Next Article in Journal
Functional Defect of Neutrophils Causing Dermatophytosis: Case Report
Previous Article in Journal
Biotransformation of Animal Fat-By Products into ARA-Enriched Fermented Bioproducts by Solid-State Fermentation of Mortierella alpina
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel 4,6-Disubstituted s-Triazin-2-yl Amino Acid Derivatives as Promising Antifungal Agents

by
Rakia Abd Alhameed
1,
Zainab Almarhoon
1,
Essam N. Sholkamy
2,
Salman Ali Khan
3,
Zaheer Ul-Haq
3,
Anamika Sharma
4,5,
Beatriz G. de la Torre
4,5,
Fernando Albericio
1,5,6,7,* and
Ayman El-Faham
1,8,*
1
Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
2
Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
3
Dr. Panjwani Center for Molecular medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
4
KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
5
Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
6
CIBER-BBN (Networking Centre on Bioengineering, Biomaterials and Nanomedicine) and Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
7
Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
8
Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Ibrahimia 12321, Alexandria, Egypt
*
Authors to whom correspondence should be addressed.
J. Fungi 2020, 6(4), 237; https://doi.org/10.3390/jof6040237
Submission received: 9 September 2020 / Revised: 14 October 2020 / Accepted: 15 October 2020 / Published: 21 October 2020

Abstract

:
A novel series of 4,6-disubstituted s-triazin-2-yl amino acid derivatives was prepared and characterized. Most of them showed antifungal activity against Candida albicans compared to clotrimazole (standard drug). Compounds bearing aniline derivatives, piperidine and glycine on the triazine core showed the highest inhibition zones at concentrations of 50, 100, 200, and 300 μg per disc. In addition, docking studies revealed that all the compounds accommodated well in the active site residues of N-myristoltransferase (NMT) and exhibited complementarity, which explains the observed antifungal activity. Interestingly, none of these compounds showed antibacterial activity.

Graphical Abstract

1. Introduction

An important component of a drug design program’s quest for new leads is the synthesis of molecules that are novel but mimic recognized biologically active molecules by virtue of the existence of certain crucial structural characteristics, especially in the development of several specific synthetic protocols for the preparation of a range of substituted s-triazine derivatives owing to their involvement in several applications in the medicinal field and the synthesis of numerous pharmaceutical agents [1,2]. The reported literature reveals that substituted s-triazine derivatives are consistent with a number of marked antimicrobial activities against Staphylococcus aureus, Bacillus cereus, Bacillus subtilis, Salmonella typhimurium, Escherichia coli Klebsiella aerogenes, and Candida albicans as human microbial pathogens [3].
C. albicans is a constituent of the normal part of the human commensal flora that lives in the human mouth and gastrointestinal tract. However, it is also is a diploid fungus that produces as both yeast and filamentous cells [4]. Systemic fungal infections (fungemias) including those by C. albicans have emerged as important causes of morbidity and mortality in immunocompromised patients (such as those with AIDS or under antitumor chemotherapy) [5,6]. In addition, hospital-acquired infections by C. albicans have become a cause of major health concerns [7,8,9,10].
In medicinal chemistry, the s-triazine ring has proved to be a privileged structure, and, therefore, its derivatives have been extensively studied against a broad number of biological targets. In this respect, s-triazine derivatives have shown antiprotozoal [11], anti-HIV [12], anticancer [13,14], antimalarial [15,16], antibacterial [17,18,19], antifungal [20,21,22], and antileishmanial activity [23], carbonic anhydrase inhibitors [24,25], and human monoamine oxidase (MAO) inhibitors [26].
Cyanuric chloride (TCT) is the starting reagent for the synthesis of s-triazine derivatives. TCT is a symmetrical tridentate compound (it has three reactive Cl) with the particularity from a synthetic point of view that once the first Cl has reacted, the two remaining Cl show different reactivity. This allows TCT to undergo sequential nucleophilic substitution by different nucleophiles (S, O, N) under a controlled temperature to furnish a full array of s-triazine derivatives [1,27,28].
Based on the previously reported data, here, we focused on the preparation of a novel series of disubstituted s-triazin-2-yl amino acid derivatives (5a–q) containing cyclic amines (morpholine, piperidine, or pyrrolidine), and 4-substituted aniline derivatives which are biologically interesting moieties [29]. To this end, we used different techniques, including conventional heating, microwave irradiation and ultrasound. The resulted disubstituted s-triazin-2-yl amino acid derivatives were screened against bacteria and fungi as a preliminary biological screening. In addition, molecular docking studies with N-Myristoltransferase (NMT) as an anti-fungal target will be discussed.

2. Materials and Methods

All reagents, chemicals and solvents were purchased from commercial suppliers. Reactions were monitored by using thin-layer chromatography (TLC, silica gel 60-F254 protected aluminum sheets). Melting points were conducted in open capillary tubes using a Gallenkamp melting point apparatus (Sigma-Aldrich Chemie GmbH, Taufkirchen, Germany) and were uncorrected. Fourier transform infrared spectroscopy (FTIR) spectra were recorded on a Nicolet 6700 spectrometer (Thermo Electron Scientific Instruments Corporation, Madison, WI, USA) using KBr discs. 1H- and 13C-NMR spectra were recorded on a Varian-Agilent-NMR 600 MHz spectrometer (Varian, Inc., Palo Altro, CA, USA). Elemental analyses were recorded on a Perkin-Elmer 2400 elemental analyzer (Perkin-Elmer Inc. Waltham, MA, USA). The ultrasonic bath was purchased from Selecta (Barcelona, Spain). Microwave experiments were performed in a multimode reactor (Monowave 300, Aton Paar GmbH, 1400 W maximum magnetron, Germany). Heating time (5 min at 70 °C and 350 watt) and reaction time were held for 12–15 min at identical watt and temperature with stirring speed (800 rpm); then, cooling was performed until 25 °C.

2.1. General Procedure for the Synthesis of Disubstituted Amino Acids-Triazine Derivatives

2.1.1. One-Pot Synthesis

A mixture of amino acid (10 mmol) and NaHCO3 (22 mmol) in 50 mL distilled water was added dropwise to a solution of cyanuric chloride (TCT; 1, 10 mmol) in 50 mL acetone at 0 °C. The reaction mixture was kept under stirring at 0 °C for 2 h, and then a solution of the amine (10 mmol) in 10 mL acetone was added dropwise (5 min) at 0 °C, followed by addition of a solution of NaHCO3 (11 mmol) in 20 mL distilled water. After complete addition, the reaction mixture was stirred overnight at room temperature. The solvent was concentrated under vacuum and then extracted with dichloromethane (10 mL). The aqueous layer was collected and acidified with 1N HCl. The white precipitate of the products (4a–f) was collected by filtration.

2.1.2. Stepwise Synthesis

The method was performed in the following two steps: Step (i) synthesis of monosubstituted s-triazine 3: a solution of the amine (1 equiv.) in 10 mL acetone was added dropwise at 0 °C in 5 min to a mixture of cyanuric chloride (1 equiv.) in 50 mL acetone, followed by the addition of a solution of NaHCO3 (1.5 Equation.) in 20 mL distilled water. The reaction mixture was kept under stirring at 0 °C for 2 h to afford mono substituted s-triazine derivatives. Step (ii) synthesis of disubstituted amino acid s-triazine: a mixture of amino acid (1 equiv.) and NaHCO3 (2.5 eq.) in 50 mL distilled water was added dropwise (10 min) to a solution of the previously prepared monosubstituted s-triazine 3 (1 equiv.) in 50 mL acetone; the reaction mixture was stirred at room temperature overnight and then acidified with 1N HCl to furnish the target compounds 4a–f.

2.2. General Procedures for the Synthesis of 4,6,-Disubstituted s-Triazin-2-yl Amino Acid Derivatives 5a–q

Method A: Conventional heating
Disubstituted s-triazine (1 mmol) was reacted with different amines (morpholine, piperidine and pyrrolidine; 1.1 mmol) in tetrahydrofuran (THF, 20 mL) and in the presence of diisopropylethylamine (DIEA; 2.2 mmol) under reflux for 24 h. The solvent was evaporated under vacuum to produce a white precipitate, which was neutralized by 1N HCl to afford trisubstituted s-triazine derivatives in 89–93% yield.
Method B: Microwave technique
Disubstituted s-triazine (1 mmol) was dissolved in 5 mL THF, and then 1.1 mmol of amine was added, followed by DIEA (2.2 mmol). This mixture was then subjected to microwave irradiation using a multimode reactor (Monowave 300, Aton Paar GmbH), where heating time (5 min, at 70 °C, 350 watt) and the reaction were held for 12–15 min at the same wattage and temperature with a stirring speed of 800 rpm. After cooling, the solvent was evaporated, and the resulting residue was subjected to neutralization with 1N HCl to obtain trisubstituted s-triazine derivatives in excellent yields and purities.
Method C: Ultrasonication method
An amine (1.1 mmol) was added to a solution of disubstituted-triazine (1 mmol) in 20 mL THF in the presence of DIEA (2.2 mmol), and the reaction mixture was subjected to ultrasound irradiation (1.5 h) at room temperature. TLC was used to monitor the completion of reaction, followed by workup similar to conventional methodology to obtain the product with highly yield (90–93%) and purity.
All compounds obtained from the three methods were purified via washing with a hot mixture of THF and a few drops of acetone.

2.2.1. (4-Morpholino-6-(Phenylamino)-1,3,5-Triazin-2-yl) Glycine (5a)

White solid in 89% (Method A), 91% (Method B) 92% (Method C) yield; mp 245–247 °C. IR (KBr, cm−1): 3341 (NH); 1669 (CO); 1H-NMR (DMSO-d6; ppm): δ 3.60 (4H, s, CH2-O-CH2), 3.66 (4H, s, CH2-N-CH2), 3.87 (2H, dd, J = 5.4 Hz, CH2COOH), 6.90 (1H, s.br, H4`), 7.19–7.23 (3H, m, H3`, H5`& NH), 7.68 (2H, dd, J = 7.2 Hz, H2` & H6`), 9.08 (1H, s, NH); 13C-NMR (DMSO-d6; ppm): δ 42.5 (CH2COOH); 43.4 (CH2-N-CH2), 66.2 (CH2-O-CH2), 119.7 (C2` & C6`); 121.6 (C4`); 128.4 (C3`& C5`); 140.4 (C1`); 164.2, 164.8 & 165.9 (triazine), 172.3 (CO). Anal. Calcd for C15H18N6O3 (330.35): C, 54.54; H, 5.49; N, 25.44. Found: C, 54.22; H, 5.56; N, 25.63; HRMS: m/z: calcd. 331.35[M+H]+; Found: 331.08.

2.2.2. (4-((4-Chlorophenyl) Amino)-6-Morpholino-1,3,5-Triazin-2-yl) Glycine (5b)

White powder in 87% (Method A), 90% (Method B), 92% (Method C) yield; mp 246–248 °C. IR (KBr, cm−1): 3350 & 3412 (NH); 1678 (CO). 1H-NMR (DMSO-d6; ppm): δ 3.60 (4H, s, CH2-O-CH2), 3.65 (4H, s, CH2-N-CH2), 3.87 (2H, dd, J = 4.8, 5.4 Hz, CH2COOH), 7.23–7.27 (3H, m, H3`, H5`& NH), 7.71 (2H, dd, J = 7.2, 8.4 Hz, H2` & H6`), 9.25 (1H, s, NH); 13C-NMR (DMSO-d6; ppm): δ 42.7 (CH2COOH); 43.7 (CH2-N-CH2), 66.4 (CH2-O-CH2), 121.5 (C2` & C6`); 125.4 (C4`); 128.5 (C3`& C5`); 139.7 (C1`); 164.2, 164.9 & 165.9 (triazine), 172.5 (CO). Anal. Calcd for C15H17ClN6O3 (364.79): C, 49.39; H, 4.70; N, 23.04. Found: C, 49.54; H, 4.81; N, 23.19; HRMS: m/z: calcd. 365.80[M+H]+, Found: 365.64.

2.2.3. (4-((4-Methoxyphenyl) Amino)-6-Morpholino-1,3,5-Triazin-2-yl) Glycine (5c)

Off-white powder in 86% (Method A), 90% (Method B), 90% (Method C) yield; mp 246–248 °C. IR (KBr, cm−1): 3349 (NH); 1674 (CO);1H-NMR (DMSO-d6; ppm): δ 3.59 (4H, s, CH2-O-CH2), 3.64 (4H, s, CH2-N-CH2), 3.69 (3H, s, OCH3), 3.83 (2H, d, J = 5.4 Hz, CH2COOH), 6.80 (2H, t, J = 7.8, 10.8 Hz, H3`& H5`), 7.54 (2H, d, J = 9 Hz, H2` & H6`), 7.57 (1H, s, NH), 8.91 (1H, s, NH); 13C-NMR (DMSO-d6; ppm): δ 42.7 (CH2COOH); 43.6 (CH2-N-CH2), 55.4 (OCH3), 66.3 (CH2-O-CH2), 113.8 (C3` & C5`); 121.6 (C2`& C6`); 133.7 (C1`); 154.5 (C4`); 164.1, 165.0 & 166.0 (triazine), 172.6 (CO). Anal. Calcd for C16H20N6O4 (360.37): C, 53.33; H, 5.59; N, 23.32. Found: C, 53.54; H, 5.71; N, 23.17; HRMS: m/z: calcd. 361.38 [M+H]+; Found: 361.13.

2.2.4. (4-(Phenylamino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Glycine (5d)

White powder in 89% (Method A), 93% (Method B), 92% (Method C) yield; mp 268–270 °C. IR (KBr, cm−1): 3339 (NH); 1674 (CO);1H-NMR (DMSO-d6; ppm): δ1.46 (4H, s, H3` & H5`), 1.59 (2H, s, H4`),3.67 (4H, s, CH2-N-CH2 ), 3.86 (2H, dd, J = 5.4, 6 Hz, NHCH2COOH), 6.88 (1H, t, J = 7.2, 7.8 Hz, H4``), 7.08 (1H, s, NH), 7.20 (2H, q, J = 7.8, 8.4 Hz, H3`` & H5``), 7.69 (2H, dd, J = 7.2, 8.4 Hz, H2`` & H6``), 8.96 (1H, d, NH); 13C-NMR (DMSO-d6; ppm): δ24.5 (C4`), 25.6(C3`& C5`), 42.5 (HOOC-CH2NH), 43.7 (C2`& C6`); 119.6 (C2`` & C6``); 121.4 (C4``); 128.4 (C3``& C5``); 140.6 (C1``); 164.1, 165. 8, 166.2 (triazine), 172.4 (CO). Anal. Calcd for C16H20N6O2 (328.38): C, 58.52; H, 6.14; N, 25.59. Found: C, 58.41; H, 6.10; N, 25.71. HRMS: m/z: calcd. 329.38 [M+H]+, Found: 329.13.

2.2.5. (4-((4-Chlorophenyl) Amino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Glycine (5e)

White powder in 88% (Method A), 91% (Method B), 92% (Method C) yield; mp 260–262 °C. IR (KBr, cm−1): 3396 (NH); 1654 (CO); 1H-NMR (DMSO-d6; ppm): δ 1.46 (4H, s, H3`& H5`), 1.59 (2H, s, H4`), 3. 67 (4H, s, CH2-N-CH2), 3.85 (2H, dd, J = 4.8 Hz, CH2COOH), 7.15 (1H, s, NH), 7.24 (2H, t, J = 9,12.6 Hz, H3`` & H5``), 7.73 (2H, dd, J = 7.2, 7.8Hz, H2`` & H6``), 9.13 (1H, s, NH); 13C-NMR (DMSO-d6; ppm): δ24.8 (C4`), 25.8 (C3`& C5`), 42.8 (HOOC-CH2NH), 44.0 (C2`& C6`); 121.3 (C2`` & C6``); 125.1 (C4``); 128.5 (C3``& C5``); 139.9 (C1``); 164. 3, 166.1, 166.5 (triazine), 172.6 (CO).Anal. Calcd for C16H19ClN6O2 (362.82): C, 52.97; H, 5.28; N, 23.16. Found: C, 53.09; H, 5.32; N, 23.40. HRMS: m/z: calcd. 363.83 [M+H]+; Found: 363.80.

2.2.6. (4-((4-Methoxyphenyl) Amino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Glycine (5f)

Off-white powder in 87% (Method A), 90% (Method B), 90% (Method C) yield; mp 261–263 °C. IR (KBr, cm−1): 3260 (NH); 1667 (CO); 1H-NMR (DMSO-d6; ppm): δ 1.45 (4H, s, H3`& H5`), 1.59 (2H, s, H4`), 3. 66 (4H, s, CH2-N-CH2), 3.68 (3H, s, OCH3), 3.84 (2H, dd, CH2COOH), 6.80 (2H, dd, J = 9, 10.8 Hz, H3`` & H5``), 7.01 (1H, s, NH), 7.57 (2H, dd, J = 6, 8.4Hz, H2`` & H6``), 8.81 (1H, d, NH);13C-NMR (DMSO-d6; ppm): δ 24.8 (C4`), 25.8 (C3`& C5`), 42.8 (CH2COOH), 44.0 (C2`& C6`); 55.6 (OCH3), 113.9 (C3``& C5``), 121.39 (C2`` & C6``); 134.0 (C1``); 154.4 (C4``), 164. 3, 164.5, 166.4 (triazine), 172.7 (CO). Anal. Calcd for C17H22N6O3 (358.40): C, 56.97; H, 6.19; N, 23.45. Found C, 56.72; H, 6.30; N, 23.67. HRMS: m/z: calcd. 359.41 [M+H]+; Found: 359.10.

2.2.7. (4-((4-Chlorophenyl) Amino)-6-(Pyrrolidin-1-yl)-1,3,5-Triazin-2-yl) Glycine (5g)

White powder in 86% (Method A), 92% (Method B), 91% (Method C) yield; mp 262–264 °C. IR (KBr, cm−1): 3371, 3246 (NH); 1661 (CO). 1H-NMR (DMSO-d6; ppm): δ 1.86 (4H, s, H3`` & H4``), 3.39–3.46 (4H, m, H2`` & H5``), 3.86 (2H, d, J = 5.4 Hz, NH CH2COOH), 7.10 (1H, d, NH). 7.23 (2H, t, J = 9, 9.6 Hz, H3` & H5`), 7.80 (2H, dd, J = 7.2, 8.4 Hz, H2` & H6`), 9.13 (1H, d, NH);13C-NMR (DMSO-d6; ppm): δ 25.3 (C3`` & C4``), 42.7 (NHCH2COOH), 46.5 (C2`` & C5``), 121.6 (C2` & C6`), 125.8 (C4`), 128.6 (C3`& C5`), 139.8 (C1`); 161.3, 163.1, 164.7 (triazine), 172.5 (CO).Anal. Calcd for C15H17ClN6O2 (348.79): C, 51.65; H, 4.91; N, 24.10. Found C, 51.81; H, 5.02; N, 24.01. HRMS: m/z: calcd. 349.80 [M+H]+; Found: 349.63.

2.2.8. (4-((4-Methoxyphenyl) Amino)-6-(Pyrrolidin-1-yl)-1,3,5-Triazin-2-yl) Glycine (5h)

White powder in 86% (Method A), 90% (Method B), 93% (Method C) yield; mp 270–272 °C. IR (KBr, cm−1): 3262 (NH); 1668 (CO);1H-NMR (DMSO-d6; ppm): δ 1.85 (4H, s, H3`` & H4``), 3.44 (4H, s, H2`` & H5``), 3.68 (3H, s, OMe), 3.86 (2H, s, NH-CH2COOH), 6.79 (2H, d, J = 7.8 Hz, H3` & H5`), 6.90 (1H, s, NH), 7.66 (2H, d, J = 7.8 Hz, H2` & H6`), 8.78 (1H, d, NH); 13C-NMR (DMSO-d6; ppm): δ 25.3 (C3``& C4``), 42.7 (CH2COOH), 46.1 (C2`` & C5``), 55.6 (OMe), 113.9 (C3` & C5`), 121.2 (C2`& C6`), 134.4 (C1`); 154.3 (C4`), 163.5, 165.8, 166.1 (triazine), 172.7 (CO). Anal. Calcd for C16H20N6O3 (344.38): C, 55.80; H, 5.85; N, 24.40. Found: C, 55.99; H, 5.97; N, 24.64. HRMS: m/z: calcd. 345.38 [M+H]+; Found: 345.56.

2.2.9. 3-((4-Morpholino-6-(Phenylamino)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5i)

White powder in 84% (Method A), 91% (Method B), 93% (Method C) yield; mp 267–269 °C. IR (KBr, cm−1): 3285 (NH); 1666 (CO). 1H-NMR (DMSO-d6; ppm): δ 2.50 (2H, t, J = 7.2 Hz, CH2COOH), 3.45 (2H, q, J = 10.8 HZ, NH-CH2CH2), 3.60 (4H, dd, J = 7.2 Hz, CH2-O-CH2), 3.66 (4H, dd, CH2-N-CH2), 6.89 (1H, s, H4`), 6.93 (1H, s, NH). 7.21 (2H, d, J = 7.8 Hz, H3` & H5`), 7.71(2H, d, J = 7.8 Hz, H2` & H6`), 9.00 (1H, d, NH), 12.19 (1H, s, COOH);13C-NMR (DMSO-d6; ppm): δ 34.5 (CH2CH2COOH); 36.8 (NH CH2CH2COOH), 43.7 (CH2-N-CH2), 66.5 (CH2-O-CH2), 119.9 (C2` & C6`); 121.7 (C4`); 128.7 (C3`& C5`); 140.9 (C1`); 164.4, 165.1 & 165.9 (triazine), 173.6 (CO). Anal. Calcd for C16H20N6O3 (344.38): C, 55.80; H, 5.85; N, 24.40. Found: C, 55.80; H, 5.85; N, 24.40. HRMS: m/z: calcd. 345.38 [M+H]+; Found: 345.44.

2.2.10. 3-((4-((4-Chlorophenyl) Amino)-6-Morpholino-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5j)

White powder in 86% (Method A), 93% (Method B), 90% (Method C) yield; mp 285–287 °C. IR (KBr, cm−1): 3277 (NH); 1678 (CO). 1H-NMR (DMSO-d6; ppm): δ2.48 (2H, s, CH2COOH), 3.44 (2H, q, J = 7.2 Hz, NH-CH2CH2), 3.60 (4H, d, J = 7.2 Hz, CH2-O-CH2), 3.64 (4H, dd, CH2-N-CH2), 6.99 (1H, s, NH), 7.24 (2H, d, J = 7.8 Hz, H3` & H5`), 7.75 (2H, d, J = 7.2 HZ, H2` & H6`), 9.17 (1H, d, NH); 13C-NMR (DMSO-d6; ppm): δ 34.4 (CH2CH2COOH), 36.8 (NHCH2CH2); 43.7 (CH2-N-CH2), 66.4 (CH2-O-CH2), 121.3 (C2`& C6`); 125.3 (C4`); 128.6 (C3`& C5`); 140.0 (C1`); 164.3, 165.0 & 165.8 (triazine), 173.5 (CO). Anal. Calcd for C16H19ClN6O3 (378.82): C, 50.73; H, 5.06; N, 22.19. Found: C, 50.95; H, 5.23; N, 22.45. HRMS: m/z: calcd. 379.82 [M+H]+; Found: 379.76.

2.2.11. 3-((4-((4-Methoxyphenyl) Amino)-6-Morpholino-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5k)

Off-white powder in 88% (Method A), 90% (Method B), 92% (Method C) yield; mp 269–271 °C. IR (KBr, cm−1): 3273 (NH); 1667 (CO).1H-NMR (DMSO-d6; ppm): δ 2.49 (2H, s, CH2COOH), 3.43 (2H, d, J = 4.2 Hz, NHCH2CH2), 3.59–3.66 (8H, m, CH2-O-CH2 & CH2-N-CH2), 3.69 (3H, s, OCH3), 6.79 (1H, s, NH), 6.80 (2H, d, J = 8.4 Hz, H3`& H5`), 7.57 (2H, s, H2` & H6`), 8.83 (1H, d, NH); 13C-NMR (DMSO-d6; ppm): δ 34.7 (CH2COOH); 37.0 (NHCH2CH2COOH), 44.0 (CH2-N-CH2), 55.7 (OCH3), 66.7 (CH2-O-CH2), 114.1 (C3` & C5`); 121.9 (C2`& C6`); 134.1 (C1`); 154. 8 (C4`); 164.5, 165.3 & 165.9 (triazine), 173.89 (CO). Anal. Calcd for C17H22N6O4 (374.40): C, 54.54; H, 5.92; N, 22.45. Found: C, 54.75; H, 6.08; N, 22.69. HRMS: m/z: calcd. 375.40 [M+H]+; Found: 375.61.

2.2.12. 3-((4-(Phenylamino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5l)

White powder in 89% (Method A); 92% (Method B), 94% (Method C) yield; mp 276–278 °C. IR (KBr, cm−1): 3275 (NH); 1663 (CO).1H-NMR (DMSO-d6; ppm): δ 1.46 (4H, s, H3` & H5`), 1.59 (2H, s, H4`), 2.49 2H, s, CH2 COOH), 3.45 (2H, t, J = 6, 10.8 Hz, CH2NH), 3.66 (4H, d, CH2-N-CH2), 6.83 (1H, s, NH), 6.87 (1H, s, H4``), 7.20 (2H, t, J = 7.2,7.8 Hz, H3`` & H5``), 7.72(2H, d, J = 7.8 Hz, H2`` & H6``), 8.92 (1H, d, NH);13C-NMR (DMSO-d6; ppm): δ 24.6 (C4`), 25.3 (C3`& C5`), 34.3 (CH2-COOH), 36.5 (NHCH2CH2COOH), 43.7 (C2`& C6`); 119.5 (C2`` & C6``); 121.3 (C4``); 128.4 (C3``& C5``); 140.8 (C1``); 164. 4, 165.7, 165.9 (triazine), 173.4 (CO). Anal. Calcd for C17H22N6O2 (342.40): C, 59.63; H, 6.48; N, 24.54. Found: C, 59.89; H, 6.66; N, 24.87. HRMS: m/z: calcd. 343.40 [M+H]+; Found: 343.61.

2.2.13. 3-((4-((4-Chlorophenyl) Amino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5m)

White powder in 86% (Method A) 92% (Method B) 92% (Method C) yield; mp 266–268 °C. IR (KBr, cm−1): 3262 (NH); 1665 (CO); 1H-NMR (DMSO-d6; ppm): δ 1.46 (4H, s, H3` & H5`), 1.59 (2H, s, H4`), 2.50 (2H, s, CH2COOH), 3.43 (2H, q, J = 6.6 Hz, CH2CH2NH), 3.67 (4H, d, CH2-N-CH2), 6.89 (1H, d, NH), 7.24 (2H, d, J = 7.8 Hz, H3``& H5``), 7.76 (2H, d, J = 8.4 Hz, H2`` & H6``), 9.09 (1H, d, NH);13C-NMR (DMSO-d6; ppm): δ24.8 (C4`), 25.8 (C3`& C5`), 34.4 (CH2CH2COOH), 36.8 (CH2CH2NH), 44.0 (C2`& C6`); 121.2 (C2`` & C6``); 125.0 (C4``); 128.5 (C3``& C5``); 140.1 (C1``); 164. 5, 165.8, 166.1 (triazine), 173.5 (CO). Anal. Calcd for C17H21ClN6O2 (376.85): C, 54.18; H, 5.62; N, 22.30. Found: C, 54.33; H, 5.76; N, 22.54. HRMS: m/z: calcd. 377.85 [M+H]+; Found: 377.63.

2.2.14. 3-((4-((4-Methoxyphenyl) Amino)-6-(Piperidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5n)

Off-white powder in 89% (Method A) 94% (Method B) 93% (Method C) yield; mp 275–277 °C. IR (KBr, cm−1): 3259 (NH); 1667 (CO); 1H-NMR (DMSO-d6; ppm): δ 1.45 (4H, s, H3` & H5`), 1.58 (2H, s, H4`), 2.50 (2H, s, CH2COOH), 3. 43 (2H, s, HOOC-CH2CH2NH), 3.65 (4H, s, CH2-N-CH2), 3.68 (3H, s, OCH3), 6.79 (2H, d, J = 9 Hz, H3`` & H5``), 7.59 (2H, s, H2`` & H6``), 8.84 (1H, d, NH);13C-NMR (DMSO-d6; ppm): δ24.9 (C4`), 25.8 (C3`& C5`), 34.6 (CH2COOH), 36.8 (CH2CH2NH) 43.9 (C2`& C6`); 55.5 (OCH3), 113.9 (C3``& C5``), 121.2 (C2`` & C6``); 134.2 (C1``); 154.4 (C4``), 164. 4, 164.7, 166.0 (triazine), 173.6 (CO). Anal. Calcd for C18H24N6O3 (372.43): C, 58.05; H, 6.50; N, 22.57. Found: C, 58.23; H, 6.62; N, 22.81. HRMS: m/z: calcd. 373.43 [M+H]+; Found: 373.55.

2.2.15. 3-((4-(Phenylamino)-6-(Pyrrolidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5o)

White powder in 87% (Method A) 91% (Method B) 92% (Method C) yield, mp 291–293°C. IR (KBr, cm−1): 3268 (NH); 1670 (CO);1H-NMR (DMSO-d6): δ 1.86 (4H, s, H3`` & H4``), 2.51 (2H, s, CH2COOH), 3.46 (6H, s, H2``, H5`` & NHCH2CH2COOH), 6.87 (1H, d, J = 6 Hz, H4`),7.19 (2H, d, J = 7.8 Hz, H3` & H5`), 7.79 (2H, s, H2` & H6`), 8.91 (1H, s, NH), 12.18 (1H, s, COOH); 13C-NMR (DMSO-d6; ppm): δ 25.3 (C3``& C4``), 34.5 (CH2COOH), 36.8 (NHCH2CH2COOH), 46.1 (C2`` & C5``), 119.6 (C2` & C6`), 121.4 (C4`), 128.7 (C3`& C5`), 141.3 (C1`), 163.7, 164. 2 (triazine), 173.6 (CO). Anal. Calcd for C16H20N6O2 (328.38): C, 58.52; H, 6.14; N, 25.59. Found: C, 58.52; H, 6.14; N, 25.59. HRMS: m/z: calcd. 329.38 [M+H]+; Found: 329.35.

2.2.16. 3-((4-((4-Chlorophenyl) Amino)-6-(Pyrrolidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5p)

White powder in 89% (Method A), 92% (Method B) 90% (Method C) yield; mp 289–291 °C. IR (KBr, cm−1): 3270 (NH); 1676 (CO);1H-NMR (DMSO-d6; ppm): δ 1.85 (4H, s, H3`` & H4``), 2.48 (2H, s, CH2COOH), 3.38 (6H, s, CH2CH2NH, H2`` & H5``), 3.45 (4H, d, J = 5.4 Hz, H2`` & H5``), 6.85 (1H, d, NH), 7.23 (2H, d, J = 7.2 Hz, H3` & H5`), 7.82 (2H, d, J = 8.4 Hz, H2` & H6`), 9.09 (1H, d, NH); 13C-NMR (DMSO-d6; ppm): δ 25.3 (C3``& C4``),34.5 (CH2COOH), 36.8 (CH2CH2NH), 46.1 (C2`` & C5``), 121.1 (C2` & C6`), 124.9 (C4`), 128.5 (C3` & C5`), 140.3 (C1`); 163.6, 164.1, 165.8 (triazine), 173.5 (CO). Anal. Calcd for C16H19ClN6O2 (362.82): C, 52.97; H, 5.28; N, 23.16. C, 52.76; H, 5.34; N, 23.43. HRMS: m/z: calcd. 363.82 [M+H]+; Found: 363.71.

2.2.17. 3-((4-((4-Methoxyphenyl) Amino)-6-(Pyrrolidin-1-yl)-1,3,5-Triazin-2-yl) Amino) Propanoic Acid (5q)

White powder in 86% (Method A), 90% (Method B), 92% (Method C) yield; mp 273–275 °C. IR (KBr, cm−1): 3261 (NH); 1660 (CO); 1H-NMR (DMSO-d6; ppm): δ 1.84 (4H, s, H3`` & H4``), 2.49 (CH2COOH), 3.39–344 (6H, m, H2``, H5``& NHCH2CH2COOH), 3.68 (3H, s, OMe), 6.79 (2H, d, J = 9 Hz, H3` & H5`), 7.66 (2H, d, J = 9.6 Hz, H2` & H6`), 8.73 (1H, d, NH); 13C-NMR (DMSO-d6): δ 25.2 (C3``& C4``), 34.6 (CH2COOH), 36.8 (NHCH2CH2COOH), 46.1 (C2`` & C5``), 55.5 (OMe), 113.9 (C3` & C5`), 121.1 (C2`& C6`), 134.4 (C1`), 154.3 (C4`); 163.7, 164. 0, 165.7 (triazine), 173.6 (CO).Anal. Calcd for C17H22N6O3 (358.40): C, 56.97; H, 6.19; N, 23.45. Found: C, 57.12; H, 6.31; N, 23.29. HRMS: m/z: calcd. 359.40 [M+H]+; Found: 359.53.

2.3. Antimicrobial Activity

2.3.1. Preparation of Microbial Inoculums

The antibacterial activity of 5a–q was evaluated against Gram-positive bacteria “S. aureus ATCC 29213 and S. epidermidis ATCC 12228”, Gram-negative bacteria “E. coli ATCC 25922 and S. typhimurium ATCC 14028” and C. albicans ATCC 60193.

2.3.2. Antimicrobial Assay Using the Disc Diffusion Method

The microbial inoculums were prepared as described in our previous work [30]. The antibacterial activity of 5a–q was studied by employing a micro-dilution method, using Mueller–Hinton broth and following the guidelines of the Clinical and Laboratory Standards Institute (CLSI) (M07, 11th Edition, 2018) [31], and serial dilution was performed using dimethyl sulfoxide (DMSO) to prepare 200 μg/mL of each compound. Paper discs (6 mm diameter) previously dipped into each compound were placed gently onto the microorganism-seeded plates. The antibacterial assay plates were then incubated at 24 h at 37 ± 1 °C. The diameters of the inhibition zones were expressed and measured in mm. Each assay was repeated three times in this experiment, and the results were expressed as a mean value ± standard deviation.

2.3.3. Minimum Inhibitory (MIC) and Minimum Fungicidal Concentration (MFC)

The MICs and MFCs were carried out as described by Rasadah and Muharnad [32]. In this regard, the nutrient agar and Sabouraud agar medium were inoculated with freshly prepared cells of C. albicans. The discs were dipped into the DMSO containing each compound and placed on the plates at concentration ranging between 50 and 300 μg per disc. Next, the discs were placed gently onto the surface of the Petri dishes after the bacterial growth and evaporation of DMSO (where it was used as a negative control). After the incubation of the Petri dishes at 37 °C for 24 h, antifungal activity was measured. Each assay was repeated three times in this experiment, and the results were expressed as a mean value.

2.3.4. Methodology of Docking Studies

In the present study, the crystal structure of N-myristoltransferase (NMT) was retrieved under the accession code 1IYL [33]. The protein structure was prepared using the protein preparation wizard in Molecular Operating Environment (MOE) Version 2018.0101 [34]. During preparation, the structures were subjected to addition of missing atoms and residues, bond order, formal charge correction, and tautomer adjustment. Hydrogens atoms were added using the protonate 3D algorithm. For protonation, GB/VI was used as the electrostatics function, with a dielectric value of 80 (for solvent). The van der Waals and electrostatics cutoffs were set at 15 and 10 A, respectively. Partial charges were applied using the AMBER10: EHT force field. The water molecules involved in the bridging ligand and protein were retained and kept rigid during energy minimization. The ligands were charged and minimized using the MMFF94x force field in MOE. The graphics were obtained using the MOE software suite.

3. Results and Discussion

3.1. Chemistry

Disubstituted s-triazinyl amino acid derivatives (5a–q) were prepared as demonstrated in Scheme 1 via either a one-pot synthesis (Route A) or stepwise synthetic pathway (Route B). In the one-pot synthesis, the amino acid was added to cyanuric chloride at 0 °C using aq. NaHCO3 as base. After 2 h, substituted aniline derivatives were added in the presence of aq. NaHCO3 as base, and the reaction was stirred overnight at rt to afford 4a–f. In contrast, the stepwise strategy (Route B, Scheme 1) involved the addition of substituted aniline derivative to cyanuric chloride at 0 °C for 2 h using NaHCO3 as base to form 3a–c. After isolation of 3a–c, underwent a nucleophilic substitution reaction with different amino acids to afford the products 4a–f.
No marked difference in the yield of 4a–f between the two routes (A and B) was observed. However, the stepwise reaction decreased the probability of side product formation from the reagents remaining in the one-pot reaction. It is important to highlight that sufficient amount of NaHCO3 was used during the synthesis to overcome/neutralize traces of cyanuric acid, which might be present in the crude cyanuric chloride.
The synthesis of 5a–q required elevated temperatures, which were achieved in three ways, namely by conventional heating, microwave irradiation or ultrasonication. Disubstituted s-triazine derivatives 4a–f (1 equiv.) and secondary amines (1.1 equiv.) were reacted in the presence of diisopropylethylamine (DIEA) as a base and THF as solvent in the three conditions mentioned above. Using conventional heating (reflux for 20–24h), 5a–q were obtained in good yields and purities. In contrast, the use of microwave irradiation afforded these compounds in better yields in only 12–15 min of reaction time, while the application of ultrasonic irradiation allowed their synthesis at room temperature in 1.5h with a greater yield and purity than conventional heating (Table 1). Of note, the prolonged reaction under conventional heating can be markedly reduced by means of microwave or ultrasound irradiation. The structures of all compounds 5a–q were confirmed by NMR spectra (1H and 13C) as shown in Supporting information (Figures S1–S17).

3.2. In Vitro Antimicrobial Assays

The antibacterial and antifungal activity of 5a–q was tested. None of the tested compounds showed antibacterial activity against the two Gram-positive bacteria (S. aureus and S. epidermidis) or two Gram-negative bacteria (E. coli and S. typhimurium). However, they showed promising antifungal activity against the human pathogenic C. albicans compared with the reference standard drug clotrimazole (Table 2). The effective bioactivity of 4,6-disubstituted s-triazin-2-yl amino acid derivatives with fungal strain, and no activity on bacterial strains may be dependent on several features of both species like cell wall, cell membrane or metabolic activity and the different amino acid moiety used for the preparation of these derivatives [35]. In addition, the proteinaceous transfer system for these bioactive compounds, primarily in the fungal strain, is responsible for the process [36].
The growth-inhibiting effects of 5a–q against C. albicans were quantitatively determined by means of the disc diffusion method at concentrations of 50, 100, 200, and 300 μg per disc. The results are shown in Table 2 and Figure 1. Compounds 5d, 5e, and 5f showed the best inhibitory capacity at 50 μg per disc of 15, 13, and 14 mm, respectively. These results revealed that the presence of the piperidine is crucial for the activity. Thus, the combination of piperidine with aniline derivatives on the triazine core bearing the glycine amino acid 5d gave the best results. In addition, among the aniline derivatives, chloro appeared to be more active than methoxy. These results are consistent with the notion that compounds bearing electron-withdrawing groups, such as chlorine atoms, have a potent inhibitory effect on the examined microbe [19,37]. This observation was also noticed in all tested compounds at different concentrations, as shown in Figure 1.
The individual minimum inhibitory concentration (MIC, μM) and minimum fungicidal concentration (MFC, μM) values of 5a–q against C. albicans are listed in Table 2. In general, all the compounds showed good antifungal activities, with a MIC value falling between 33.47 and 37.95 μM and MFC value between 66.58 and 75.90 μM. Again, this assay supports the previous findings illustrated inTable 2.

3.3. Docking Studies

The observed antifungal activity of the compounds can be attributed to the inhibition of N-myristoltransferase (NMT) enzyme, which has been validated as an anti-fungal target [32]. NMT activity is essential for the vegetative growth of C. albicans. NMT is a promising target enzyme for the development of novel fungicidal drugs with a broad anti-fungal spectrum. To date, various types of NMT inhibitors have been reported, including peptidomimetics, benzothiazoles, tetrahydro- carbazoles, sulphonamides and benzofurans. Recently, we have carried out an extensive virtual screening to identify NMT inhibitors effective against kinetoplastids. Therefore, we examined the protein–ligand interaction profile of 5b, 5d, 5j, 5n and 5o with NMT. The docking profile of compound 5d suggests that the compound is well accommodated in the binding site of C. albicans NMT. The s-triazine moiety mediates hydrogen bonds with the side chain hydroxyl of Y225. The Y225 also mediates another hydrogen bond with ligand 5o (Figure 2). The benzene ring of Y225 exhibits pi stacking interaction with the benzene ring of the ligands. Furthermore, the hydrogen bonding between the ligand and backbone amide of L355 and L394 provides anchorage. Moreover, the ligand also demonstrates hydrophobic contact with surrounding residues, which explains the inhibitory potential of 5o observed.
A slightly different binding mode was observed in the triazine moiety of 5d mediate hydrogen bond interaction with L451 (Figure 2). Y225 exhibits pi stacking interaction with the benzene ring. The compound also demonstrates polar contact with the side chain of T211. An extensive network of hydrogen and hydrophobic contacts provides further anchorage.
The protein–ligand interaction profile of 5j, 5b and 5n demonstrated that the triazine and benzene moieties of the ligands are stacked against Y359 (Figure 2). The methoxy substitution shows polar contact with L394. However, the oxygen atom of the oxathiane ring mediates the hydrogen bond with N392.
Our results demonstrate that all the compounds are well accommodated and exhibit complementarity with the active site residues of C. albicans NMT, which explains the observed antifungal activity.
Furthermore, to explore the antifungal activity against other pathogenic fungi, the active site of Cryptococcus neoformans, Aspergillus fumigatus and Histoplasma capsulatum NMTs were aligned to C. albicans NMT, as known crystal structures of NMTs from different fungi indicate that all NMTs have similar substrate binding sites despite their low sequence identity. The sequence similarity is obtained from blastp between C. albicans NMT with C. neoformans NMt (46%), A. fumigatus NMT (49%) and H. capsulatum NMT (46%), and, interestingly, they share a similar substrate binding site. As depicted in Figure 3, the alignment of the active sites of NMTs from different fungi reveals that all the active site residues in C. albicans were well aligned with C. neoformans, A. fumigatus and H. capsulatum. However, Tyr256 in C. albicans replaced Asn249 and Asn331 in A. fumigatus and H. capsulatum, respectively, while Phe339 in C. albicans replaced Ser378, Ser351 and Ser415 in A. fumigatus, C. neoformans and H. capsulatum, respectively. This high degree of similarity between binding sites may be attributed to the broad spectrum antifungal activities, and these compounds could be active against other pathogenic fungi species.

4. Conclusions

Novel 4,6-disubstituted s-triazin-2-yl amino acid derivatives were prepared via conventional heating, microwave irradiation and ultrasonication. The two latter techniques gave 5a–q in good purity and better yield within shorter time intervals compared to the conventional heating method. The synthesized derivatives were not found to be active against two Gram-positive bacteria (S. aureus ATCC 29213, S. epidermidis ATCC 12228) and two Gram-negative bacteria (E. coli ATCC 25922 and S. typhimurium ATCC 14028). However, they showed promising antifungal activities against the human pathogen C. albicans (ATCC 60193).
The key to the antifungal activity is the presence of the piperidine ring. Thus, the compounds containing piperidine were the most active, which agreed with the previously reported data [29]. Interestingly, morpholine (six-membered ring containing one O instead of C as in the case of piperidine) and pyrrolidine (five-membered ring) showed less activity. This supports the need for a degree of hydrophobicity in this position.
On the other hand, aniline without any substituent proved to be more promising than chloro (electron-withdrawing) and methoxy (electron-donating) analogs. In addition, the synthesized compounds were found to be well accommodated in the active site residues of C. albicans NMT, which explains the observed antifungal activities.
The known crystal structures of NMTs from different pathogenic fungi, namely, C. neoformans, A. fumigatus and H. capsulatum, indicate that all NMTs share similar substrate binding sites. This high degree of similarity between binding sites may be attributed to the broad-spectrum antifungal activities, and these compounds could be active against other pathogenic fungi species.
Finally, we can summarize the structure–activity relationship of our target compounds as indicated in Figure 4.
To gain a complete picture of the effect of structural modification or functionalization on antifungal activities, further development for a new series based on s-triazine amino acid derivatives with different substituents will be considered in our laboratories.

Supplementary Materials

The following are available online at https://www.mdpi.com/2309-608X/6/4/237/s1, Figures S1–S17: 1H-NMR and 13C-NMR for compound 5a–q.

Author Contributions

The work was designed and supervised by A.E.-F., Z.A., B.G.d.l.T., and F.A.; the synthesis and characterization of the reported compounds were carried out by R.A.A. and Z.A.; E.N.S. and A.S. performed the biological activity assay; S.A.K. and Z.U.-H. carried out the docking study; R.A.A., Z.A., E.N.S., and S.A.K. prepared the first drafts of the manuscript. The final version includes contributions from all authors. All authors have read and agreed to the published version of the manuscript.

Funding

Deanship of Scientific Research at King Saud University, Saudi Arabia, research group no. (RGP-1441-234). National Research Foundation (NRF) (# 105892 and Blue Sky’s Research Programme # 120386).

Acknowledgments

The authors extend their thanks to the Deanship of Scientific Research at King Saud University for funding this work through research group no. (RGP -1441-234, Saudi Arabia). The authors express their deep gratitude to one of the referees for his/her more-than-valuable comments.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Blotny, G. Recent applications of 2,4,6-trichloro-1,3,5-triazine and its derivatives in organic synthesis. Tetrahedron 2006, 62, 9507–9522. [Google Scholar] [CrossRef]
  2. Desai, N.C.; Makwana, A.H.; Rajpara, K.M. Synthesis and study of 1,3,5-triazine based thiazole derivatives as antimicrobial agents. J. Saudi Chem. Soc. 2016, 20, S334–S341. [Google Scholar] [CrossRef] [Green Version]
  3. Srinivas, K.; Srinivas, U.; Bhanuprakash, K.; Harakishore, K.; Murthy, U.S.; Rao, V.J. Synthesis and antibacterial activity of various substituted s-triazines. Eur. J. Med. Chem. 2006, 41, 1240–1246. [Google Scholar] [CrossRef] [PubMed]
  4. Ryan, K.J.; Ray, C.G. Sherris Medical Microbiology; McGraw Hill: New York, NY, USA, 2004; Volume 4, ISBN 0-8385-8529-9. [Google Scholar]
  5. Kullberg, B.J.; Oude Lashof, A.M. Epidemiology of opportunistic invasive mycoses. Eur. J. Med. Res. 2002, 7, 183–191. [Google Scholar] [PubMed]
  6. Weig, M.; Groß, U.; Mühlschlegel, F. Clinical aspects and pathogenesis of Candida infection. Trends Microbiol. 1998, 6, 468–470. [Google Scholar] [CrossRef]
  7. Zadik, Y.; Burnstein, S.; Derazne, E.; Sandler, V.; Ianculovici, C.; Halperin, T. Colonization of Candida: Prevalence among tongue-pierced and non-pierced immunocompetent adults. Oral Dis. 2010, 16, 172–175. [Google Scholar] [CrossRef] [PubMed]
  8. Achkar, J.M.; Fries, B.C. Candida infections of the genitourinary tract. Clin. Microbiol. Rev. 2010, 23, 253–273. [Google Scholar] [CrossRef] [Green Version]
  9. Kelly, M.T.; MacCallum, D.M.; Clancy, S.D.; Odds, F.C.; Brown, A.J.P.; Butler, G. The Candida albicans CaACE2 gene affects morphogenesis, adherence and virulence. Mol. Microbiol. 2004, 53, 969–983. [Google Scholar] [CrossRef]
  10. Kumamoto, C.A. Inflammation and gastrointestinal Candida colonization. Curr. Opin. Microbiol. 2011, 14, 386–391. [Google Scholar] [CrossRef] [Green Version]
  11. Melato, S.; Prosperi, D.; Coghi, P.; Basilico, N.; Monti, D. A Combinatorial Approach to 2,4,6-trisubstituted triazines with potent antimalarial activity: Combining conventional synthesis and microwave-assistance. Chem. Med. Chem. 2008, 3, 873–876. [Google Scholar] [CrossRef]
  12. Xiong, Y.-Z.; Chen, F.-E.; Balzarini, J.; De Clercq, E.; Pannecouque, C. Non-nucleoside HIV-1 reverse transcriptase inhibitors. Part 11: Structural modulations of diaryltriazines with potent anti-HIV activity. Eur. J. Med. Chem. 2008, 43, 1230–1236. [Google Scholar] [CrossRef] [PubMed]
  13. El-Faham, A.; Farooq, M.; Almarhoon, Z.; Alhameed, R.A.; Wadaan, M.A.M.; de la Torre, B.G.; Albericio, F. Di- and tri-substituted s-triazine derivatives: Synthesis, characterization, anticancer activity in human breast-cancer cell lines, and developmental toxicity in zebrafish embryos. Bioorg. Chem. 2020, 94, 103397. [Google Scholar] [CrossRef] [PubMed]
  14. Al-Rasheed, H.H.A.; Malebari, A.M.M.; Dahlous, K.A.A.; El-Faham, A. Synthesis and Characterization of New Series of 1,3-5-Triazine Hydrazone Derivatives with Promising Antiproliferative Activity. Molecules 2020, 25, 2708. [Google Scholar] [CrossRef]
  15. Kumar, A.; Srivastava, K.; Kumar, S.R.; Puri, S.K.; Chauhan, P.M.S. Synthesis of 9-anilinoacridine triazines as new class of hybrid antimalarial agents. Bioorg. Med. Chem. Lett. 2009, 19, 6996–6999. [Google Scholar] [CrossRef] [PubMed]
  16. Bhat, H.R.; Singh, U.P.; Gahtori, P.; Ghosh, S.K.; Gogoi, K.; Prakash, A.; Singh, R.K. 4-Aminoquinoline-1,3,5-triazine: Design, synthesis, in vitro antimalarial activity and docking studies. New J. Chem. 2013, 37, 2654–2662. [Google Scholar] [CrossRef]
  17. Pathak, P.; Thakur, A.; Bhat, H.R.; Singh, U.P. Hybrid 4-Aminoquinoline-1,3,5-triazine Derivatives: Design, Synthesis, Characterization, and Antibacterial Evaluation. J. Heterocycl. Chem. 2015, 52, 1108–1113. [Google Scholar] [CrossRef]
  18. Haibaa, N.S.; Khalil, H.H.; Abdel Moniem, M.; El-Wakil, M.H.; Bekhit, A.A.; Khattab, S.N. Design, synthesis and molecular modeling studies of new series of s-triazine derivatives as antimicrobial agents against multi-drug resistant clinicalisolates. Bioorg. Chem. 2019, 89, 103013. [Google Scholar] [CrossRef]
  19. Ramadan, D.R.; Elbardan, A.A.; Bekhit, A.A.; El-Faham, A.; Khattab, S.N. Synthesis and characterization of novel dimeric s-triazine derivatives as potential anti-bacterial agents against MDR clinical isolates. New J. Chem. 2018, 42, 10676–10688. [Google Scholar] [CrossRef]
  20. Sarmah, K.; Sarmah, N.; Kurmi, K.; Patel, T. Synthesis and studies of antifungal activity of 2,4,6-trisubstituted 1,3,5-triazines. Adv. Appl. Sci. Res. 2012, 3, 1459–1462. [Google Scholar]
  21. Saeed, S.; Rashid, N.; Jones, P.G.; Yunas, U. 2-substituted 4H-[1,3]thiazolo[3,2-a][1,3,5]triazine-4-thiones: Synthesis, crystal structure, and antifungal activity. J. Heterocycl. Chem. 2010, 47, 908–912. [Google Scholar] [CrossRef]
  22. Romani, L. Immunity to fungal infections. Nat. Rev. Immunol. 2011, 11, 275–288. [Google Scholar] [CrossRef] [PubMed]
  23. Khattab, S.N.; Khalil, H.H.; Bekhit, A.A.; Abd El-Rahman, M.M.; de la Torre, B.G.; El-Faham, A.; Albericio, F. 1,3,5-triazino peptide derivatives: Synthesis, characterization, and preliminary antileishmanial activity. ChemMedChem 2018, 13, 725–735. [Google Scholar] [CrossRef] [Green Version]
  24. Mikulová, M.B.; Kružlicová, D.; Pecher, D.; Supuran, C.T.; Mikuš, P. Synthetic Strategies and Computational Inhibition Activity Study for Triazinyl-Substituted Benzenesulfonamide Conjugates with Polar and Hydrophobic Amino Acids as Inhibitors of Carbonic Anhydrases. Int. J. Mol. Sci. 2020, 21, 3661. [Google Scholar] [CrossRef] [PubMed]
  25. Mikuš, P.; Krajčiová, D.; Mikulová, M.; Horváth, B.; Pecher, D.; Garaj, V.; Bua, S.; Angeli, A.; Supuran, C.T. Novel sulfonamides incorporating 1,3,5-triazine and amino acid structural motifs as inhibitors of the physiological carbonic anhydrase isozymes I, II and IV and tumor-associated isozyme IX. Bioorg. Chem. 2018, 81, 241–252. [Google Scholar] [CrossRef] [PubMed]
  26. Khattab, S.N.; Khalil, H.H.; Bekhit, A.A.; El-Rahman, M.M.A.; El-Faham, A.; Albericio, F. Synthesis and preliminary biological evaluation of 1,3,5-triazine amino acid derivatives to study their MAO inhibitors. Molecules 2015, 20, 15976–15988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sharma, A.; El-Faham, A.; de la Torre, B.G.; Albericio, F. Exploring the Orthogonal Chemoselectivity of 2,4,6-Trichloro-1,3,5-Triazine (TCT) as a Trifunctional Linker With Different Nucleophiles: Rules of the Game. Front. Chem. 2018, 6, 516. [Google Scholar] [CrossRef] [Green Version]
  28. Calvete, M.J.F.; Pinto, S.M.A.; Burrows, H.D.; Castro, M.M.C.A.; Geraldes, C.F.G.C.; Pereira, M.M. Multifunctionalization of cyanuric chloride for the stepwise synthesis of potential multimodal imaging chemical entities. Arab. J. Chem. 2020, 13, 2517–2525. [Google Scholar] [CrossRef]
  29. Desai, N.C.; Makwana, A.H.; Senta, R.D. Synthesis, characterization and antimicrobial activity of some novel 4-(4-(arylamino)-6-(piperidin-1-yl)-1,3,5-triazine-2-ylamino)-N-(pyrimidin-2-yl)benzenesulfon amides. J. Saudi Chem. Soc. 2016, 20, 686–694. [Google Scholar] [CrossRef] [Green Version]
  30. Al-Marhoon, Z.; Abdel-Megeed, A.; Sholkamy, E.N.; Siddiqui, M.R.H.; El-Faham, A. Synthesis of Phenylcarbamic Acid and 2-[2-Oxo-3-(4-substituted phenylimino)-indolin-1-yl] acetohydrazide Derivatives as Promising Antifungal Agents. Asian J. Chem. 2014, 26, 7665–7672. [Google Scholar] [CrossRef]
  31. Wayne, P.A. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically, 11th ed.; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2018. [Google Scholar]
  32. Rasadah, M.A.; Muharnad, Z. Prosid. In PerubatanTraditional Malaysia Ke-5; Universiti Malaya: Kuala Lumpur, Malaysia, 1988; p. 173. [Google Scholar]
  33. Toraskar, M.; Prasad, K.; Kadam, V. N-myristoyltransferase: A novel target. Mini. Rev. Med. Chem. 2008, 8, 142–149. [Google Scholar] [CrossRef]
  34. Molecular Operating Environment (MOE), Version 2018.0101; Chemical Computing Group Inc.: Montreal, QC, Canada, 2016.
  35. Scheffers, D.-J.; Pinho, M.G. Bacterial Cell Wall Synthesis: New Insights from Localization Studies. Microbiol. Mol. Biol. Rev. 2005, 69, 585–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Dare, K.; Ibba, M. Roles of tRNA in cell wall biosynthesis. Wiley Interdiscip. Rev. RNA 2012, 3, 247–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mewada, N.S.; Shah, D.R.; Lakum, H.P.; Chikhalia, K.H. Synthesis and biological evaluation of novel s-triazine based aryl/heteroaryl entities: Design, rationale and comparative study. J. Assoc. Arab Univ. Basic Appl. Sci. 2016, 20, 8–18. [Google Scholar] [CrossRef] [Green Version]
Scheme 1. Synthesis of di- and tri-substituted s-triazine amino acid derivatives.
Scheme 1. Synthesis of di- and tri-substituted s-triazine amino acid derivatives.
Jof 06 00237 sch001
Figure 1. Inhibition zones of 5a–q at concentrations of 50,100, 200 and 300 μg per disc against C. albicans. Error bars mean the STDV of the average values of the triplicate experiments.
Figure 1. Inhibition zones of 5a–q at concentrations of 50,100, 200 and 300 μg per disc against C. albicans. Error bars mean the STDV of the average values of the triplicate experiments.
Jof 06 00237 g001
Figure 2. The protein–ligand interaction profile of the ligands (5b, 5d, 5j, 5n, and 5o).
Figure 2. The protein–ligand interaction profile of the ligands (5b, 5d, 5j, 5n, and 5o).
Jof 06 00237 g002
Figure 3. Alignment of active site of C. albicans N-myristoltransferase (NMT) (firebrick color) with C. neoformans NMT (grey), Aspergillus fumigatus (spring green color) NMT and Histoplasma capsulatum NMT (salmon color).
Figure 3. Alignment of active site of C. albicans N-myristoltransferase (NMT) (firebrick color) with C. neoformans NMT (grey), Aspergillus fumigatus (spring green color) NMT and Histoplasma capsulatum NMT (salmon color).
Jof 06 00237 g003
Figure 4. The structure–activity relationship.
Figure 4. The structure–activity relationship.
Jof 06 00237 g004
Table 1. Yield (%) of the synthesis of 5a–q derivatives via conventional heating, microwave irradiation and ultrasonic methods.
Table 1. Yield (%) of the synthesis of 5a–q derivatives via conventional heating, microwave irradiation and ultrasonic methods.
CompoundConventional
(20–24 h)
Microwave
(12–15 min)
Ultrasound
(1.5 h)
5a899192
5b879092
5c869090
5d899392
5e889192
5f879090
5g869291
5h869093
5i849193
5j869390
5k889092
5l899294
5m869292
5n899493
5o879192
5p899290
5q869092
Table 2. Zone of inhibition (mm) *, Minimum Inhibitory Concentration (MIC) and Minimum Fungicidal Concentration (MFC) of the target compounds against Candida albicans growth.
Table 2. Zone of inhibition (mm) *, Minimum Inhibitory Concentration (MIC) and Minimum Fungicidal Concentration (MFC) of the target compounds against Candida albicans growth.
CompoundC. albicans (mm)MIC
(μM)
MFC
(μM)
50 µg100 µg200 µg300 µg
5a7 ± 0.19 ± 0.49 ± 0.210 ± 0.137.76 ± 0.775.52 ± 0.7
5b7 ± 0.38 ± 0.210 ± 0.112 ± 0.334.14 ± 0.168.28 ± 0.1
5c7 ± 0.38 ± 0.39 ± 0.310 ± 0.134.58 ± 0.869.16 ± 0.7
5d15 ± 0.218 ± 0.421 ± 0.323 ± 0.437.95 ± 0.275.90 ± 0.9
5e13 ± 0.116 ± 0.118 ± 0.320 ± 0.534.36 ± 0.668.72 ± 0.8
5f14 ± 0.416 ± 0.717 ± 0.419 ± 0.134.74 ± 0.369.48 ± 0.3
5g7 ± 0.18 ± 0.310 ± 0.311 ± 0.235.73 ± 0.771.46 ± 0.5
5h7 ± 0.29 ± 0.19 ± 0.310 ± 0.136.19 ± 0.272.38 ± 1.0
5i8 ± 0.310 ± 0.212 ± 0.412 ± 0.136.19 ± 0.472.38 ± 0.9
5j10 ± 0.413 ± 0.514 ± 0.315 ± 0.633.29 ± 0.366.58 ± 0.6
5k7 ± 0.28 ± 0.19 ± 0.310 ± 0.136.19 ± 0.272.38 ± 1.2
5l8 ± 0.210 ± 0.211 ± 0.312 ± 0.236.40 ± 0.372.8 ± 0.4
5m7 ± 0.314 ± 0.315 ± 0.215 ± 0.333.09 ± 0.166.18 ± 0.9
5n7 ± 0.29 ± 0.29 ± 0.110 ± 0.133.47 ± 0.566.94 ± 1.2
5o7 ± 0.18 ± 0.310 ± 0.210 ± 0.337.95 ± 0.775.90 ± 0.9
5p7 ± 0.18 ± 0.111 ± 0.311 ± 0.134.35 ± 0.468.70 ± 0.6
5q7 ± 0.28 ± 0.19 ± 0.410 ± 0.134.78 ± 0.669.56 ± 0.4
Clotrimazole **---23 ± 0.2--
* Inhibition zone (mm) at 300 µg per disc; ** 100 µg per disc, ± means Standard deviation (STDV) of triplicate experiments.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abd Alhameed, R.; Almarhoon, Z.; N. Sholkamy, E.; Ali Khan, S.; Ul-Haq, Z.; Sharma, A.; G. de la Torre, B.; Albericio, F.; El-Faham, A. Novel 4,6-Disubstituted s-Triazin-2-yl Amino Acid Derivatives as Promising Antifungal Agents. J. Fungi 2020, 6, 237. https://doi.org/10.3390/jof6040237

AMA Style

Abd Alhameed R, Almarhoon Z, N. Sholkamy E, Ali Khan S, Ul-Haq Z, Sharma A, G. de la Torre B, Albericio F, El-Faham A. Novel 4,6-Disubstituted s-Triazin-2-yl Amino Acid Derivatives as Promising Antifungal Agents. Journal of Fungi. 2020; 6(4):237. https://doi.org/10.3390/jof6040237

Chicago/Turabian Style

Abd Alhameed, Rakia, Zainab Almarhoon, Essam N. Sholkamy, Salman Ali Khan, Zaheer Ul-Haq, Anamika Sharma, Beatriz G. de la Torre, Fernando Albericio, and Ayman El-Faham. 2020. "Novel 4,6-Disubstituted s-Triazin-2-yl Amino Acid Derivatives as Promising Antifungal Agents" Journal of Fungi 6, no. 4: 237. https://doi.org/10.3390/jof6040237

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop