Next Article in Journal
A Survey of Marine Natural Compounds and Their Derivatives with Anti-Cancer Activity Reported in 2011
Previous Article in Journal
Efficient Synthesis and Anti-Fungal Activity of Oleanolic Acid Oxime Esters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Antiviral Activity of N-Phenylbenzamide Derivatives, a Novel Class of Enterovirus 71 Inhibitors

Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2013, 18(3), 3630-3640; https://doi.org/10.3390/molecules18033630
Submission received: 6 January 2013 / Revised: 14 March 2013 / Accepted: 18 March 2013 / Published: 21 March 2013
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
A series of novel N-phenylbenzamide derivatives were synthesized and their anti-EV 71 activities were assayed in vitro. Among the compounds tested, 3-amino-N-(4-bromophenyl)-4-methoxybenzamide (1e) was active against the EV 71 strains tested at low micromolar concentrations, with IC50 values ranging from 5.7 ± 0.8–12 ± 1.2 μM, and its cytotoxicity to Vero cells (TC50 = 620 ± 0.0 μM) was far lower than that of pirodavir (TC50 = 31 ± 2.2 μM). Based on these results, compound 1e is a promising lead compound for the development of anti-EV 71 drugs.

Graphical Abstract

1. Introduction

Enterovirus 71 (EV 71) is a single-strained and positive sense RNA virus that belongs to the Picornaviridae family. It was first isolated and identified from patients with central nervous system (CNS) diseases in California between 1969 and 1974 [1]. Since then, outbreaks of EV 71 have been reported in several countries, especially in the Asia Pacific region [2,3,4]. Diseases caused by an EV 71 infection include hand-foot-mouth disease (HFMD), herpangina (HA) and CNS involvement with fatal pulmonary edema [5]. EV 71 infections are a global public health problem causing severe clinical illness and even death in young children. However, there are currently no antiviral drugs or vaccines available in the clinic against EV 71, and the majority of treatments are simply supportive symptom management treatments. Consequently, there is an increasing need for the discovery of effective drugs against EV 71. Additionally, due to the high mutation rate of EV 71, several genotypes have been isolated in the clinic [6], and, at times, more than one genotype has been associated with both complicated and uncomplicated diseases [7]. Therefore, pinpointing the active strain has been unsuccessful. Consequently, the development of anti-EV 71 drugs should be mainly focused on the discovery of compounds with activity against several genotypes of EV 71.
Antiviral screening of our privileged structure library was carried out, and compound 2a (Figure 1) exhibited moderate activity against EV 71 virus (strain H, C2 genotype) in vitro with an IC50 value of 18 ± 1.2 μM. In order to obtain compounds with more potent activity and superior physicochemical profiles, we synthesized a series of N-phenylbenzamide derivatives and evaluated their antiviral activity against four strains (SZ-98, JS-52-3, H, BrCr) of EV 71 in Vero cells. The structure modification was mainly made to the substituents at the C-3 position of the benzene ring A to investigate the importance of amide group for the anti-EV 71 activity. Meanwhile, because there was no substituent on the ring B of compound 2a, which possibly made it vulnerable to metabolism, we also synthesized derivatives with substituents on the ring B to make them metabolically stable, and also to explore the structure-activity relationships on the ring B. This is the first report of the anti-EV 71 activity of the N-phenylbenzamide scaffold, to the best of our knowledge.
Figure 1. The structure of compound 2a.
Figure 1. The structure of compound 2a.
Molecules 18 03630 g001

2. Results and Discussion

2.1. Chemistry

As shown in Scheme 1 and Scheme 2, a series of N-phenylbenzamide derivatives were designed and synthesized according to our previous reported method [8]. In Scheme 1, 3-amino-4-methoxybenzoic acid was chosen as the starting material and was condensed with a variety of amines, using N,N′-diisopropylcarbodiimide (DIC) as the coupling reagent and N-hydroxybenzotriazole (HOBt) as the activating reagent [9], to yield the intermediate compounds 1ae. Under the same conditions, compounds 1a, b were condensed with 2-fluoropropionic acid, which was obtained by the hydrolysis of methyl 2-fluoropropanoate, to afford the target compounds 2a, b. Alkylation of the amine groups of compounds 1a, c and d was accomplished by a nucleophilic substitution reaction. To avoid cleavage of the amide bond during the reaction, a mild base (NaHCO3) was employed as the acid-binding agent. When iodomethane (MeI) was used as the alkylating agent in excess, dialkylation products 3b, d were also obtained in addition to monoalkylated products. Additionally, by a similar method used to synthesize compounds 2a, b, we also synthesized compound 2c using 3-methoxy-4-aminobenzoic acid as the starting material.
Scheme 1. The synthetic route to compounds 2ac and 3ag.
Scheme 1. The synthetic route to compounds 2ac and 3ag.
Molecules 18 03630 g003
Reagents and conditions: (i) DIC, HOBt, R1NH2, CH2Cl2, rt, 5 h, 60–70%, (ii) 2-fluoropropionic acid, DIC, HOBt, CH2Cl2, rt, 5 h, 60–70%, (iii) MeI/PrI, NaHCO3, CH2Cl2, rt, 12 h, 30–50%.
Scheme 2. The synthetic route to compounds 5af.
Scheme 2. The synthetic route to compounds 5af.
Molecules 18 03630 g004
Reagents and conditions: (i) TEA, CH2Cl2, R4COCl, 0 °C-rt, 6 h, 70–80%, (ii) DIC, HOBt, amines, CH2Cl2, rt, 5 h, 60–70%, or SOCl2, reflux 1 h, then CH2Cl2, TEA, amines , 0 °C-rt, 6 h, 60–70%.
As depicted in Scheme 2, we also synthesized several derivatives with propionyloxy (acetoxy) and propionylamino substituents on the benzene ring A. 3-Hydroxyl/amino-4-methoxybenzoic acid was first reacted in dichloromethane with propionyl chloride or acetyl chloride using triethylamine (TEA) as the base to afford compounds 4ac. By using a similar method to that used for compounds 1ae, compounds 5af were thus obtained. Alternatively, compounds 4ac were first reacted with thionyl chloride under refluxing conditions to give intermediate chloride products, which were condensed with a substituted arylamine to yield the target compounds 5af. Ultimately, compounds 5af were obtained in overall yields of 40–60%.

2.2. Anti-EV 71 Activity

The EV 71 was divided into three genotypes A, B and C based on nucleotide sequence comparisons, and genotypes B and C were subdivided into B1-B5 and C1-C5 [6,10]. Genotype C is the most common genotype found in mainland China, especially the C4 subgenotype [4,11]. Consequently, the intermediate compounds 1a, 1c and 1e, and the final products 2ac, 3ag, and 5af were evaluated for their anti-EV 71 activity in Vero cells against genotypes C4 (strains SZ-98 and JS-52-3), C2 (strain H) and A (strain BrCr). Pirodavir (Figure 2) is a rather potent and broad-spectrum picornavirus inhibitor. It was reported to be effective against 80% of 16 enteroviruses tested at the concentration of 1.3 μg/mL [12]. Therefore, we employed pirodavir as positive control to validate our model. The IC50 values were calculated by the Reed and Muench method, and the cytotoxicity was monitored by the CPE method.
Figure 2. The chemical structure of pirodavir.
Figure 2. The chemical structure of pirodavir.
Molecules 18 03630 g002
As indicated in Table 1, the cytotoxicity of all the synthesized compounds was far lower than that of pirodavir, and most of the synthesized N-phenylamide derivatives exhibited some activity against several strains of EV 71 simultaneously, especially compound 1e, which was active against all the strains tested at low micromolar concentrations (5.7 ± 0.80–12 ± 1.2 μM). In addition to compound 1e, other compounds also showed moderate levels of inhibitory activity with IC50 values of approximately 15 μM including, compound 1c against strain SZ-98, compound 3g against strains H and BrCr, 3e against strain H, 5c against strains SZ-98, JS-52-3 and H, and compound 5e against strain SZ-98. Moreover, the selectivity index (SI) values of compounds 1c (10–36), 1e (51–110), 3g (16–35), 5b (>16 – >27), 5c (16–63) and 5e (15–37) were also comparable or superior to those of pirodavir (25–52).
Table 1. The activity of the synthesized N-phenylbenzamide derivatives against several strains of EV 71.
Table 1. The activity of the synthesized N-phenylbenzamide derivatives against several strains of EV 71.
CpdsTC50
(μM)
SZ-98JS-52-3HBrCr
IC50
(μM)
SIIC50
(μM)
SIIC50
(μM)
SIIC50
(μM)
SI
1a>820350 ± 242.3160 ± 7.95.2190 ± 144.3160 ± 125.2
1c520 ± 2915 ± 0.63646 ± 5.71234 ± 3.61656 ± 4.210
1e620 ± 0.012 ± 1.1519.8 ± 0.4645.7 ± 0.81109.1 ± 1.468
2a630 ± 0.0>630-160 ± 5.7>3.918 ± 1.2>35>630-
2b>620>620-210 ± 0.0>3.0430 ± 36>1.4360 ± 26>1.7
2c>630110 ± 12>5.834 ± 2.8 41 ± 2.9>1690 ± 9.5>7.0
3a780 ± 0.0>260-260 ± 0.03.0260 ± 0.03.0180 ± 144.3
3b510 ± 3873 ± 8.67.042 ± 3.11231 ± 4.31650 ± 3.810
3c280 ± 5.964 ± 0.04.337 ± 1.97.537 ± 1.27.544 ± 1.06.2
3d>560320 ± 32>1.7>560-190 ± 0.0>3.0190 ± 0.0>3.0
3e410 ± 22>78-34 ± 4.61220 ± 1.22045 ± 0.09.0
3f120>70->70->70->70-
3g530>59-34 ± 2.51615 ± 0.03520 ± 3.127
5a>670>75-58 ± 4.9>1296 ± 10>7.045 ± 109.0
5b>60038 ± 2.9> 1639 ± 1.9>1622 ± 2.9>27>67-
5c530 ± 0.011 ± 0.84720 ± 3.1278.4 ± 1.26334 ± 0.016
5d280 ± 21190 ± 121.444 ± 3.76.265 ± 0.04.353 ± 2.45.2
5e470 ± 2213 ± 2.43732 ± 3.61422 ± 4.72132 ± 3.915
5f360 ± 3990 ± 114.098 ± 4.73.747 ± 6.17.695 ± 9.03.8
Pirodavir31 ± 2.21.2 ± 0.2251.0 ± 0.2300.6 ± 0.1521.0 ± 0.230
TC50 was defined as the concentration that inhibits 50% cellular growth in comparison with untreated controls and calculated by Reed and Muench method. IC50 (50% inhibitory concentration) was calculated by Reed & Muench method. The selectivity index (SI) was calculated as the ratio of TC50/IC50. “-” stands for no antiviral selectivity.
In terms of structure-activity relationships, benzene ring B was essential for anti-EV 71 activity. For example, when benzene ring B was replaced by a cyclohexyl group (2b), no anti-EV 71 activity was observed. On the basis of the activity of compounds 1c, 1e, 5b, 5c and 5e against EV 71, it was concluded that the introduction of electron-withdrawing groups (Cl and Br, especially Br) at the para position of benzene ring B increased anti-EV 71 activity. Substitution of acetoxy groups (5e, f) for the propionylamino group (5a, b) on benzene ring A had little influence on anti-EV 71 activity. Because the activity of compounds with propionyl substituent at the amino group (5b, c) was slightly inferior to their unsubstituted counterparts (1c, e), it can be concluded that the propionyl substituent at the amino group disfavors the anti-EV 71 activity. Compound 5d presented less potent activity against EV 71 in comparison to 5e. Therefore, it is possible that H-bond donor group at the position of R6 substituent favors the anti-EV 71 activity.

3. Experimental

3.1. General

All reagents and solvents were purchased from J&K and Alfa Aesar Chemicals, and were used without purification. 1H-NMR and 13C-NMR spectra were recorded in CDCl3 or DMSO-d6 on a Varian Inova 400/500 MHz spectrometer (Varian, San Francisco, CA, USA). Chemical shift was reported in parts per million relative to tetramethylsilane as the internal standard. Melting points were determined with a X6 microscope melting point apparatus and were uncorrected. Electrospray ionisation (ESI) high-resolution mass spectra (HRMS) were obtained on an MDS SCIEX Q-Trap mass spectrometer. The area normalization purities of the tested compounds were >95% as determined using analytical high-performance liquid chromatography (HPLC method for all compounds: detector wavelength: 260 nm, column temperature: 25 °C, column: ODS (5 μm, 4.6 × 250 mm), mobile phase: methanol/water = 75/25, flow rate = 1 mL/min). For the synthesis and spectral data of compounds 1a, 2a, 4a, 4b, 5a and 5c, please refer to our previous paper [8].

3.1.1. General Procedure for the Synthesis of Compound 1bf, 5b, 5e, 5f

3-Amino-4-methoxybenzoic acid/4a, c (1.20 mmol) was dissolved in CH2Cl2 (20 mL), and DIC (1.82 mmol) and HOBt (1.82 mmol) was added to the solution. The resulting mixture was stirred for 0.5 h at room temperature, after which amines (1.68 mmol) was added. After approximately 12 h, the reaction was quenched by the addition of 0.5 N NaOH solution (20 mL), and the organic layer was separated and washed with dilute hydrochloric acid (10%, 30 mL) and brine (30 mL), successively. The solution was then dried over anhydrous MgSO4, filtered and concentrated, and the crude residue was purified over silica gel (petroleum/ethyl acetate = 3/2) to give the corresponding compounds.
3-Amino-N-cyclohexyl-4-methoxybenzamide (1b). White solid, yield: 65%. mp: 171–172 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 1.13 (1H, m), 1.24 (4H, m), 1.62 (1H, m), 1.75 (4H, m), 3.63 (1H, m), 3.79 (3H, s), 4.79 (2H, br), 6.80 (1H, d, J = 8.4 Hz), 7.07 (1H, d, J = 8.4 Hz), 7.12 (1H, s), 8.25 (1H, s). ESI-HRMS m/z: 249.16018 (M+H+) (Calcd for C14H21N2O2: 249.16030).
3-Amino-N-(4-chlorophenyl)-4-methoxybenzamide (1c). White solid, yield: 66%. mp 152–154 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 3.84 (s, 3H), 5.46 (2H, br), 6.88 (d, 1H, J = 6.8 Hz), 7.19 (m, 2H), 7.37 (d, 2H, J = 8.0Hz), 7.78 (d, 2H, J = 8.0Hz), 10.07 (s, 1H). 13C-NMR (100 MHz, CDCl3) δ: 55.6, 109.6, 113.4, 117.4, 121.2, 124.7, 128.9, 129.2, 136.5, 150.1, 156.9. ESI-HRMS m/z: 277.07487 (M+H+) (Calcd for C14H14ClN2O2: 277.07438).
3-Amino-4-methoxy-N-(3,4,5-trimethoxyphenyl)benzamide (1d). White solid, yield: 65%. mp: 140–142 °C. 1H-NMR (500 MHz, DMSO-d6) δ: 3.62 (3H, s), 3.76 (6H, s), 3.83 (3H, s), 4.92 (2H, s), 6.88 (1H, d, J = 8.0 Hz), 7.22 (4H, m), 9.85 (1H, s). ESI-HRMS m/z: 333.14778 (M+H+) (Calcd for C17H21N2O5: 333.14505).
3-Amino-N-(4-bromophenyl)-4-methoxybenzamide (1e). White solid, yield: 68%. mp 190–192°C. 1H-NMR (500 MHz, DMSO-d6) δ: 3.84 (s, 3H), 4.97 (s, 2H), 6.90 (d, 1H, J = 8.0 Hz), 7.21 (m, 2H), 7.51 (d, 2H, J = 8.0 Hz), 7.74 (d, 2H, J = 8.0 Hz), 10.10 (s, 1H). 13C-NMR (100 MHz, CDCl3) δ: 55.6, 109.6, 113.6, 116.6, 117.3, 121.5, 127.3, 131.9, 136.5, 137.3, 150.1, 165.5. ESI-HRMS m/z: 321.02365 (M+H+) (Calcd for C14H14BrN2O2: 321.02387).
4-Amino-3-methoxy-N-phenylbenzamide (1f). White solid, yield: 70%. mp: 174–176°C. 1H-NMR (400 MHz, DMSO-d6) δ: 3.82 (s, 3H), 5.37 (2H, s), 6.66 (1H, d, J = 8.0 Hz), 7.03 (1H, d, J = 8.0 Hz), 7.32 (2H, m), 7.45 (2H, m), 7.73 (2H, d, J = 7.6 Hz), 9.77 (1H, s). ESI-HRMS m/z: 243.11329 (M+H+) (Calcd for C14H15N2O2: 243.11335).
N-(4-Chlorophenyl)-4-methoxy-3-propionamidobenzamide (5b). White solid, yield: 70%. mp: 130–132 °C. 1H-NMR (400 MHz, CDCl3) δ: 1.07 (3H, t, J = 7.6 Hz), 2.42 (2H, q, J = 7.6 Hz), 3.96 (3H, s), 6.98 (1H, d, J = 8.0 Hz), 7.32 (1H, d, J = 8.0 Hz), 7.62 (2H, d, J = 8.0 Hz), 7.79 (1H, dd, J = 8.0 and 2.0 Hz), 7.82 (1H, s), 8.09 (1H, s), 8.87 (1H, s). ESI-HRMS m/z: 333.10200 (M+H+) (Calcd for C17H18ClN2O3: 333.10059).
N-(4-Chlorophenyl)-4-methoxy-3-acetoxybenzamide (5e). White solid, yield: 58%. mp: 177–179 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.34 (3H, s), 3.89 (3H, s), 6.73 (1H, d, J = 8.0 Hz), 7.01 (2H, d, J = 8.4 Hz), 7.13 (1H, d, J = 8.0 Hz), 7.22 (2H, d, J = 8.4 Hz), 8.30 (1H, s), 9.01 (1H, s). ESI-HRMS m/z: 320.06891 (M+H+) (Calcd for C16H15ClNO4: 320.06896).
4-Methoxy-N-phenyl-3-acetoxybenzamide (5f). White solid, yield: 61%. mp: 153–155 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.29 (3H, s), 3.85 (3H, s), 7.08 (1H, t, J = 8.0 Hz), 7.26 (1H, d, J = 8.4 Hz), 7.33 (2H, t, J = 8.0 Hz), 7.73 (3H, m), 7.92 (d, 1H, J = 8.4 Hz), 10.11 (1H, s). ESI-HRMS m/z: 286.10799 (M+H+) (Calcd for C16H16NO4: 286.10793).

3.1.2. General Procedure for the Synthesis of Compounds 2b, c

2-Fluoropropanoic acid (2.40 mmol) was dissolved in CH2Cl2 (20 mL), and DIC (3.60 mmol) and HOBt (3.60 mmol) was added to the solution. The resulting mixture was stirred for 0.5 h at room temperature, after which compound 1b/1e (1.71 mmol) was added. After approximately 12 h, the reaction was quenched by the addition of 0.5 N NaOH solution (20 mL), and the organic layer was separated and washed with dilute hydrochloric acid (10%, 30 mL) and brine (30 mL), successively. The solution was then dried over anhydrous MgSO4, filtered and concentrated, and the crude residue was purified over silica gel (petroleum/ethyl acetate = 4/1) to give compounds 2b, c.
N-Cyclohexyl-3-(2-fluoropropanamido)-4-methoxybenzamide (2b). White solid, yield: 73%. mp: 192–194 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 1.15 (1H, m), 1.26 (4H, m), 1.55 (3H, dd, J = 6.4 and 16.8 Hz), 1.61 (1H, m), 1.72 (4H, m), 3.73 (1H, m), 3.93 (3H, s), 5.34 (1H, qq, J = 6.4 and 48.8 Hz), 7.15 (1H, d, J = 8.0 Hz), 8.06 (1H, d, J = 8.0 Hz),8.33 (1H, s), 9.16 (1H, s), 13.63 (1H, s). ESI-HRMS m/z: 323.17695 (M+H+) (Calcd for C17H24FN2O3: 323.17710).
4-(2-Fluoropropanamido)-3-methoxy-N-phenylbenzamide (2c). White solid, yield: 59%. mp: 142–143 °C. 1H-NMR (500 MHz, DMSO-d6) δ: 1.55 (3H, dd, J = 6.5 and 20.0 Hz), 3.96 (3H, s), 5.34 (1H, qq, J = 7.0 and 48.5 Hz), 7.09 (1H, t, J = 8.0 Hz), 7.35 (2H, t, J = 8.0 Hz), 7.61 (2H, m), 7.74 (2H, d, J = 8.0 Hz), 8.17 (1H, d, J = 8.0 Hz), 9.22 (1H, s), 10.12 (1H, s). ESI-HRMS m/z: 317.13087 (M+H+) (Calcd for C17H18FN2O3: 317.13015).

3.1.3. General Procedure for the Synthesis of Compound 3a–g

Compound 1a/1c/1d (2.2 mmol) was dissolved in CH2Cl2 (15 mL), and NaHCO3 (2.5 mmol) and MeI/n-PrI (22 mmol) were added. The resulting mixture was stirred for 12 h at room temperature. The mixture was filtered, and the filtrate was concentrated in vacuo to give a residue which was purified over silica gel (petroleum/ethyl acetate = 2/1) to give the target compounds 3ag.
4-Methoxy-3-methylamino-N-phenylbenzamide (3a). White solid, yield: 33%. mp: 170–172 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.77 (d, 3H, J = 5.2 Hz), 3.84 (s, 3H), 5.19 (q, 1H, J = 5.2 Hz), 6.88 (d, 1H, J = 8.0 Hz), 7.05 (m, 2H), 7.23 (d, 1H, J = 8.0 Hz), 7.32 (t, 2H, J = 8.0 Hz), 7.74 (d, 2H, J = 8.0 Hz), 9.93 (s, 1H). 13C-NMR (100 MHz, CDCl3) δ: 30.2, 55.6, 107.8, 108.2, 115.0, 119.9, 124.1, 128.1, 129.0, 138.3, 139.5, 149.6, 166.1. ESI-HRMS m/z: 257.12934 (M+H+) (Calcd for C15H17N2O2: 257.12900).
N-(4-Chlorophenyl)-3-dimethylamino-4-methoxybenzamide (3b). White solid, yield: 40%. mp: 160–162 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.77 (6H, s), 3.86 (3H, s), 7.03 (1H, d, J = 8.0 Hz), 7.37 (2H, d, J = 8.0 Hz), 7.43 (1H, s), 7.60 (1H, d, J = 8.0 Hz), 7.79 (2H, d, J = 8.0 Hz), 10.12 (1H, s). 13C-NMR (100 MHz, DMSO-d6) δ: 23.8, 56.2, 111.4, 117.7, 122.2, 127.2, 127.4, 128.8, 138.9, 142.3, 155.1, 157.3, 165.9. ESI-HRMS m/z: 305.10524 (M+H+) (Calcd for C16H18ClN2O2: 305.10568).
4-Methoxy-3-methylamino-N-(3,4,5-trimethoxyphenyl)benzamide (3c). White solid, yield: 30%. mp: 147–148 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.77 (3H, s), 3.62 (3H, s), 3.75 (6H, s), 3.84 (3H, s), 5.21 (1H, br), 6.89 (1H, d, J = 8.0 Hz), 7.01 (1H, s), 7.24 (3H, m), 9.84 (1H, s). 13C-NMR (100 MHz, CDCl3) δ: 30.2, 55.6, 56.1, 60.9, 65.8, 97.6, 107.8, 108.6, 114.9, 127.9, 134.4, 134.5, 139.5, 149.6, 153.3, 166.1. ESI-HRMS m/z: 347.16079 (M+H+) (Calcd for C18H23N2O5: 347.16070).
3-Dimethylamino-4-methoxy-N-(3,4,5-trimethoxyphenyl)benzamide (3d). White solid, yield: 21%. mp: 120–122 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.76 (6H, s), 3.62 (3H, s), 3.75 (6H, s), 3.87 (3H, s), 7.08 (1H, d, J = 8.0 Hz), 7.20 (2H, s), 7.48 (1H, s), 7.82 (1H, d, J = 8.0 Hz), 9.98 (1H, s). 13C-NMR (100 MHz, CDCl3) δ: 30.2, 55.6, 56.1, 56.1, 60.9, 97.9, 107.8, 108.4, 110.2, 115.2, 116.4, 124.8, 127.2, 127.8, 134.2, 153.2, 166.3. ESI-HRMS m/z: 361.17621 (M+H+) (Calcd for C19H25N2O5: 361.17635).
4-Methoxy-N-phenyl-3-(propylamino)benzamide (3e). White solid, yield: 46%. mp: 121–123 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 0.94 (3H, t, J = 7.2 Hz), 1.62 (2H, m), 3.09 (2H, m), 3.84 (3H, s), 4.93 (1H, t, J = 5.6 Hz), 6.88 (1H, d, J = 8.4 Hz), 7.05 (2H, m), 7.23 (1H, d, J = 8.4 Hz), 7.31 (2H, t, J = 8.0 Hz), 7.73 (2H, d, J = 8.0 Hz), 9.93 (1H, s). 13C-NMR (100 MHz, DMSO-d6) δ: 11.6, 21.7, 40.1, 55.6, 107.8, 108.6, 115.6, 120.3, 123.2, 127.4, 128.5, 137.8, 139.4, 148.9, 165.7. ESI-HRMS m/z: 285.15975 (M+H+) (Calcd for C17H21N2O2: 285.16006).
N-(4-Chlorophenyl)-4-methoxy-3-propylaminobenzamide (3f). White solid, yield: 37%. mp: 168–171 °C. 1H-NMR (500 MHz, DMSO-d6) δ: 0.93 (3H, t, J = 7.0 Hz), 1.59 (2H, m), 3.09 (2H, m), 3.84 (3H, s), 4.93 (1H, t, J = 5.0 Hz), 6.89 (1H, d, J = 8.0 Hz), 7.05 (1H, s), 7.24 (1H, dd, J = 1.5 and 8.0 Hz), 7.36 (2H, d, J = 8.0 Hz), 7.79 (2H, d, J = 8.0 Hz), 10.05 (1H, s). 13C-NMR (100 MHz, DMSO-d6) δ: 11.6, 21.7, 39.9, 55.6, 107.8, 108.7, 115.7, 121.8, 126.8, 127.2, 128.4, 137.9, 138.5, 149.0, 165.9. ESI-HRMS m/z: 319.12139 (M+H+) (Calcd for C17H20ClN2O2: 319.12133).
4-Methoxy-3-(propylamino)-N-(3,4,5-trimethoxyphenyl)benzamide (3g). White solid, yield: 41%. mp: 164–167 °C. 1H-NMR (500 MHz, DMSO-d6) δ: 0.92 (3H, t, J = 7.0 Hz), 1.60 (2H, m), 3.09 (2H, m), 3.62 (3H, s), 3.75 (6H, s), 3.84 (3H, s), 4.93 (1H, br), 6.89 (1H, d, J = 8.0 Hz), 7.06 (1H, s), 7.23 (3H, m), 9.84 (1H, s). 13C-NMR (100 MHz, CDCl3) δ: 11.5, 22.5, 45.2, 55.5, 56.0, 60.9, 97.6, 108.2, 108.3, 114.6, 127.8, 134.5, 134.5, 138.7, 149.4, 153.3, 166.1. ESI-HRMS m/z: 375.19227 (M+H+) (Calcd for C20H27N2O5: 375.19200).
3-Acetoxy-4-methoxybenzoic acid (4c). To a solution of 3-hydroxy-4-methoxybenzoic acid (5.0 g, 30 mmol) and Et3N (4.5 g, 45 mmol) in CH2Cl2 (100 mL) was added acetyl chloride (3.5 g, 45 mmol) at 0 °C, and the resulting mixture was warmed to room temperature and then stirred for another 2 h. The reaction mixture was then washed with water (100 mL), and brine (100 mL) successively. The organic layer was dried over anhydrous MgSO4, filtered and concentrated, and the crude residue was purified over silica gel (petroleum/ethyl acetate = 4/1) to give compounds 4c (3.8 g, yield: 60%), white solid, mp: 230–231 °C. 1H-NMR (400 MHz, DMSO-d6) δ: 2.23 (3H, s), 3.85 (3H, s), 7.35 (1H, d, J = 8.0 Hz), 7.83 (1H, s), 7.90 (1H, d, J = 8.0 Hz), 12.21 (1H, s). ESI-HRMS m/z: 209.04513 (M−H) (Calcd for C10H9O5: 209.04500).
N-(4-Chlorophenyl)-N-methyl-4-methoxy-3-propionyloxybenzamide (5d). A mixture of compound 4b (0.5 g, 2.23 mmol) and SOCl2 (15 mL, 206 mmol) was heated under reflux for 1 h. The excess SOCl2 was evaporated and the corresponding chloride product was obtained as yellow oil, which was dissolved in CH2Cl2 (5 mL) and used without further purification. N-methyl-4-chlorobenzenamine (0.38 g, 2.68 mmol) was dissolved in CH2Cl2 (20 mL), and Et3N (0.34 g, 3.34 mmol) was added, and the mixture was cooled to 0 °C (ice bath). A solution of the acid chloride product in CH2Cl2 (5 mL) was added to the solution dropwise. The resulting mixture was warmed to room temperature and then stirred for another 4 h, after which water (20 mL) was added, and the organic layer was separated, washed with brine (30 mL), and dried over anhydrous MgSO4. It was then filtered and concentrated to give a residue, which was purified over silica gel (petroleum/ethyl acetate = 1/1) to yield compound 5d as a white solid (0.5 g, yield: 64%). mp: 139–141 °C. 1H-NMR (400 MHz, CDCl3) δ: 1.23 (3H, t, J = 7.6 Hz), 2.55 (2H, q, J = 7.6 Hz), 3.44 (3H, s), 3.77 (3H, s), 6.72 (1H, d, J = 8.4 Hz), 6.98 (1H, d, J = 8.4 Hz), 7.06 (1H, s), 7.11 (1H, d, J = 8.8 Hz), 7.22 (2H, d, J = 8.8 Hz). ESI-HRMS m/z: 348.10021 (M+H+) (Calcd for C18H19ClNO4: 348.10026).

3.2. Biological Activity Test Procedures

Vero cells were purchased from the American Type Culture Collection and cultured in Minimum Essential Medium (MEM) supplemented with 10% Fetal Bovine Serum (FBS) and antibiotics (100 U/mL penicillin G, 100 μg/mL streptomycin). Enterovirus 71 (EV 71) strain SZ-98 was kindly provided by Dr. Qi Jin, Institute of Pathogen Biology, Chinese Academy of Medical Science and Peking Union Medical School. Enterovirus 71 (EV 71) strain BrCr (VR-1775) was purchased from the American Type Culture Collection. Enterovirus 71 (EV 71) strain JS-52-3 was kindly provided by Dr. Xiang-zhong Ye, Beijing Wantai Biological Pharmacy Enterprise Co., Ltd. All Enterovirus 71 strains were propagated in Vero cells.
All compounds were dissolved in DMSO at 20 mg/mL as a stock solution and further diluted in culture medium prior to use. The initial concentration in all cytotoxicity assay was 200 µg/mL, and inhibition assay for anti-EV71 experiments was from maximum non-toxic concentration, and proceeded serially 3-fold dilution with culture medium. All cytotoxicity and activity assay for each compound was repeated for three times, and the data was indicated as average ± SD.

3.2.1. Cytotoxicity Determination

The cytotoxicity of compounds in the presence of Vero cells was monitored by cytopathic effect (CPE). Vero cells (2.5 × 104/well) were plated into a 96-well plate. A total of 24 h later, the monolayer cells were incubated in the presence of various concentrations of test compounds. After 48 h of culture at 37 °C and 5% CO2 in a carbon-dioxide incubator, the cells were monitored by CPE. The median toxic concentration (TC50) was calculated by Reed & Muench analyses.

3.2.2. Anti EV 71 Activity Assay

Confluent Vero cells grown in 96-well microplates were infected respectively with 100 median tissue culture infective dose (TCID50) EV 71. After 1 h adsorption at 37 °C, the monolayers were washed by PBS and incubated at 37 °C in the maintenance medium with or without different concentrations of test compounds. Viral cytopathic effect (CPE) was observed when the viral control group reached 4 and the antiviral activity of tested compounds was determined by the Reed & Muench analyses.

4. Conclusions

In summary, a series of N-phenylbenzamide derivatives were synthesized and their anti-EV 71 activities were evaluated in vitro. All of the synthesized compounds were less toxic to Vero cells in comparison to pirodavir, and several compounds were comparable or superior to pirodavir in terms of SI values, especially compound 1e. This compound has a molecular weight and an ALogP of 321.169 g/mol and 2.6 (data obtained from Discovery Studio 3.0), respectively, and shows potential for optimization to improve its activity against EV 71 without sacrificing its druggability. Consequently, compound 1e is a promising lead compound for the development of a new anti-EV 71 agent, and further lead optimization based on compound 1e will be performed.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Grant 30873138 and 81202414).

References

  1. Schmidt, N.J.; Lennette, E.H.; Ho, H.H. An apparently new enterovirus isolated from patients with disease of the central nervous system. J. Infect. Dis. 1974, 129, 304–309. [Google Scholar] [CrossRef]
  2. Samuda, G.M.; Chang, W.K.; Yeung, C.Y.; Tang, P.S. Monoplegia caused by Enterovirus 71: An outbreak in Hong Kong. Pediatr. Infect. Dis. J. 1987, 6, 206–208. [Google Scholar] [CrossRef]
  3. Singh, S.; Chow, V.T.; Phoon, M.C.; Chan, K.P.; Poh, C.L. Direct detection of enterovirus 71 (EV71) in clinical specimens from a hand, foot, and mouth disease outbreak in Singapore by reverse transcription-PCR with universal enterovirus and EV71-specific primers. J. Clin. Microbiol. 2002, 40, 2823–2827. [Google Scholar] [CrossRef]
  4. Yang, F.; Ren, L.; Xiong, Z.; Li, J.; Xiao, Y.; Zhao, R.; He, Y.; Bu, G.; Zhou, S.; Wang, J.; et al. Enterovirus 71 outbreak in the People's Republic of China in 2008. J. Clin. Microbiol. 2009, 47, 2351–2352. [Google Scholar] [CrossRef]
  5. Lee, T.C.; Guo, H.R.; Jenny Su, H.J.; Yang, Y.C.; Chang, S.L.; Chen, K.T. Diseases caused by enterovirus 71 infection. Pediatr. Infect. Dis. J. 2009, 28, 904–910. [Google Scholar] [CrossRef]
  6. Brown, B.A.; Oberste, M.S.; Alexander, J.P, Jr.; Kennett, M.L.; Pallansch, M.A. Molecular epidemiology and evolution of enterovirus 71 strains isolated from 1970 to 1998. J. Virol. 1999, 73, 9969–9975. [Google Scholar]
  7. Shin, S.R.; Ho, M.S.; Lin, K.H.; Wu, S.L.; Chen, Y.T.; Wu, C.N.; Lin, T.Y.; Chang, L.Y.; Tsao, K.C.; Ning, H.C.; et al. Genetic analysis of enterovirus 71 isolated from fatal and non-fatal cases of hand, Foot and mouth disease during an epidemic in Taiwan, 1998. Virus Res. 2000, 68, 127–136. [Google Scholar] [CrossRef]
  8. Hao, L.H.; Li, Y.P.; He, W.Y.; Wang, H.Q.; Shang, G.Z.; Jiang, J.D.; Li, Y.H.; Li, Z.R. Synthesis and antiviral activity of substituted bisaryl amide compounds as novel influenza virus inhibitors. Eur. J. Med. Chem. 2012, 55, 117–124. [Google Scholar] [CrossRef]
  9. Humphrey, J.M.; Chamberlin, A.R. Chemical synthesis of natural product peptides: Coupling methods for the incorporation of noncoded amino acids into peptides. Chem. Rev. 1997, 97, 2243–2266. [Google Scholar] [CrossRef]
  10. Huang, Y.P.; Lin, T.L.; Kuo, C.Y.; Lin, M.W.; Yao, C.Y.; Liao, H.W.; Hsu, L.C.; Yang, C.F.; Yang, J.Y.; Chen, P.J.; et al. The circulation of subgenogroups B5 and C5 of enterovirus 71 in Taiwan from 2006 to 2007. Virus. Res. 2008, 137, 206–212. [Google Scholar] [CrossRef]
  11. Tan, X.J.; Huang, X.J.; Zhu, S.L.; Chen, H.; Yu, Q.L.; Wang, H.Y.; Huo, X.X.; Zhou, J.H.; Wu, Y.; Yan, D.M.; et al. The persistent circulation of enterovirus 71 in People's Republic of China: causing emerging nationwide epidemics since 2008. PLoS One 2008, 6, e25662. [Google Scholar]
  12. Andries, K.; Dewindt, B.; Snoeks, J.; Willebrords, R.; van Eemeren, K.; Stokbroekx, R.; Janssen, P.A. In vitro activity of pirodavir (R 77975), a substituted phenoxy-pyridazinamine with broad-spectrum antipicornaviral activity. Antimicrob. Agents Chemother. 1992, 36, 100–107. [Google Scholar] [CrossRef]
  • Sample Availability: Samples of the compounds 1a, 1c, 1e, 2ac, 3ag, and 5af are available from the authors.

Share and Cite

MDPI and ACS Style

Ji, X.-Y.; Wang, H.-Q.; Hao, L.-H.; He, W.-Y.; Gao, R.-M.; Li, Y.-P.; Li, Y.-H.; Jiang, J.-D.; Li, Z.-R. Synthesis and Antiviral Activity of N-Phenylbenzamide Derivatives, a Novel Class of Enterovirus 71 Inhibitors. Molecules 2013, 18, 3630-3640. https://doi.org/10.3390/molecules18033630

AMA Style

Ji X-Y, Wang H-Q, Hao L-H, He W-Y, Gao R-M, Li Y-P, Li Y-H, Jiang J-D, Li Z-R. Synthesis and Antiviral Activity of N-Phenylbenzamide Derivatives, a Novel Class of Enterovirus 71 Inhibitors. Molecules. 2013; 18(3):3630-3640. https://doi.org/10.3390/molecules18033630

Chicago/Turabian Style

Ji, Xing-Yue, Hui-Qiang Wang, Lan-Hu Hao, Wei-Ying He, Rong-Mei Gao, Yan-Ping Li, Yu-Huan Li, Jian-Dong Jiang, and Zhuo-Rong Li. 2013. "Synthesis and Antiviral Activity of N-Phenylbenzamide Derivatives, a Novel Class of Enterovirus 71 Inhibitors" Molecules 18, no. 3: 3630-3640. https://doi.org/10.3390/molecules18033630

Article Metrics

Back to TopTop