2,782
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Synthesis, antiviral activity and structure–activity relationship of 1-(1-aryl-4,5-dihydro-1H-imidazoline)-3-chlorosulfonylureas and products of their cyclization

, , , , , & show all
Pages 787-795 | Received 16 Apr 2015, Accepted 07 Jun 2015, Published online: 27 Jul 2015

Abstract

Novel 1-(1-aryl-4,5dihydro-1H-imidazoline)-3-chlorosulfonylourea derivatives 3a3f were synthesized in the reaction of 1-aryl-4,5-dihydro-1H-imidazol-2-amines with chlorosulfonyl isocyanate. The second series of compounds 4a4f was prepared from the respective 1-(1-aryl-4,5-dihydro-1H-imidazoline)-3-chlorsulfonylureas 3a3f and 1,1′-carbonyldiimidazole (CDI). The selected compounds were tested for their activity against Herpes simplex virus and coxsackievirus B3 (CVB3). It was determined that three derivatives, i.e 3d, 4a and 4d are active against Herpes simplex virus (HSV-1). Compounds 3d and 4c are active against CVB3. Their favorable activity can be primarily attributed to their low lipophilicity values. Moreover, the lack of substituent in the phenyl moiety or 4-methoxy substitution can be considered as the most beneficial for the antiviral activity.

Introduction

Viral infections are a permanent health problem of mankind. More and more often used therapeutic methods like organ or bone marrow transplantations, when patients are subject to strong immunosuppression, result in numerous viral infections caused by viruses from the Herpesviridae family. Herpes simplex virus type l (HSV-1) infection can cause several clinical conditions such as keratitis, cutaneous herpes and encephalitisCitation1–3. These pathologies may result from a primary infection or, alternatively, from a reactivation of a latent infection. Human herpesviruses cause mild or asymptomatic infections in patients with properly functioning immunological systemCitation4.

The illness is more serious in patients with deteriorated cellular immunity, e.g. human immunodeficiency virus (HIV) infected patients who must receive chronic treatment with antiviral agents favoring the selection of resistant variantsCitation5. Infectious complications can include central nervous system inflammation, pneumonia and congenital malformation. A number of nucleoside analogues, especially the guanosine analogue acyclovir have been developed as antiherpetic agents. The therapeutic limitation of these nucleoside analogues is that drug resistant strains develop readily through mutations in viral genes for thymidine kinase and polymeraseCitation6. There are many drugs active against herpesviruses now, but despite their high selectivity they have many defects, like numerous side-effectsCitation7. Therefore, the continuous search for new compounds as antiviral agents in the HSV therapy is urgently needed.

Myocarditis is defined as inflammation of the heart muscle and is commonly associated with viral infections. Coxsackieviruses, adenoviruses and parvovirus B19 have been implicated as causes of myocarditis. However, the main pathogen associated with this condition appears to be coxsackievirus B3 (CVB3)Citation8. CVB3 is a positive-stranded RNA virus belonging to Picornaviridae. CVB3 infection can lead to both acute and chronic viral myocarditis in infants, children, young adults and immunocompromised individuals. Myocarditis caused by CVB3 often contributes to the development of inflammatory dilatative cardiomyopathy (DCMi), and most patients with DCMi eventually require heart transplantation. Besides heart infections, CVB3 can be responsible for chronic inflammatory diseases of the pancreas and the central nervous systemCitation9,Citation10. CVB3 infections are increasing risk for public health and specific antiviral treatment for CVB3 is not available at the moment. Recently, N-benzenesulfonyl sophocarpinic acid/ester and sophocarpinol derivatives were synthesized and tested for anti CVB3 activity. Among the tested compounds, sophocarpinol exerted the most promising activity against not only CVB3 but also CVB1, CVB2, CVB5 and CVB6 with IC50 ranging from 0.62 to 3.63 μM (SI from 46 to 275), indicating a broad-spectrum anti-CVB activityCitation10. Extract from Selaginella moellendorffii Hieron and its main constituent amentoflavone also exhibit anti-CVB3 activity, both in vitro by preventing cytopathic effect formation in HEp-2 cells and in vivo by reducing mean viral titres in the heart and kidneys as well as mortality of CVB3 infected miceCitation11. It was recently reported that, oxysterol-binding protein (OSBP), a PI4P-binding protein that shuttles cholesterol between membrane compartments, is implicated as another host factor for enterovirus replicationCitation12. OSW-1 (3β,16β,17α-trihydroxycholest-5-en-22-one16-O-{O-(2-O-(4-methoxybenzoyl)-β-d-xylopyranosyl)-(1→3)-2-O-acetyl-α-arabinopyranoside, a natural compound extracted from the bulbs of the Ornithogalum saundersiae, and itraconazole, a well-known antifungal agent, are inhibitors of viral RNA replication by targeting oxysterol-binding protein (OSBP) and can be potentially used as broad-spectrum inhibitors of enterovirusesCitation12,Citation13.

In this study, we present the evaluation of antiviral activity of N-substituted derivatives of 1-arylimidazolidyn-2-ylideneurea against the SV-1Citation14,Citation15 and CVB3. The rationale of this work can be summarized as follows: (1) the design of compounds was based on previously reported imidazo[1,2-a][1,3,5]triazinesCitation16 with antiviral activity () and aryl(heteroaryl)sulfonyl ureasCitation17; (2) the set of substituents was selected based on our previous experience enabling to explore structure–activity relationship. In this study, we present synthesis, antiviral activity against the HSV-1 and CVB3 viruses and structure–activity relationship studies for the 1-(1-aryl-4,5dihydro-1H-imidazoline)-3-chlorosulfonyloureas 3a3f and products their cyclization 4a4f ().

Figure 1. Previously reported antiviral compoundCitation16.

Figure 1. Previously reported antiviral compoundCitation16.

Figure 2. The scheme of synthesis of the investigated compounds.

Figure 2. The scheme of synthesis of the investigated compounds.

Methods

Chemistry

All commercial reagents and solvents were purchased from Sigma-Aldrich (Sigma Aldrich Corp., St. Louis, MO) and used without purification. Reactions were routinely monitored by thin-layer chromatography (TLC) in silica gel (60 F254 plates Merck, Darmstadt, Germany) and the products were visualized with ultraviolet light of 254 nm wavelength. All NMR spectra were acquired on a Bruker AVANCE III 300 MHz spectrometer (Bruker Bioscience, Billerica, MA) equipped with BBO Z-gradient probe. Spectra were recorded at 25 °C using DMSO as a solvent with a non-spinning sample in 5  mm NMR-tubes. MS spectra were recorded on Bruker microTOF-Q II and processed using Compass Data Analysis software. The elementary analysis was performed with the application of Perkin-Elmer analyzer (Perkin Elmer Inc., Waltham, MA). Melting points were determined with a Boetius apparatus (Jena, Germany).

General procedure for the synthesis of 1-(1-aryl-4,5-dihydro-1H-imidazoline)-3-chlorosulfonylureas (3a–3f)

1-Aryl-4,5-dihydro-1H-imidazol-2-amines (1a1f) (0.01 mol) were dissolved in 25 mL of dichloromethane under the atmosphere of dry nitrogen and added to the solution chlorosulfonyl isocyanate 2 1.41 g (0.01 mol) dissolved in 25 mL of dichloromethne. The mixture was shaken for 24 h at room temperature. Solvent was removed by distillation and the rubber-like residue was treated with warm propan-2-ol. The solid product was filtrated off and purified by crystallization from propan-2-ol.

1-(1-Phenyl-4,5-dihydro-1H-imidazoline)-3-chlorosulfonylurea (3a)

From a general procedure with 1.61 g of 1a and 1.41 g of 2, obtaining 2.0 g of 3a (66% yield), white crystalline solid, mp 200–202 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.45 (s, 1H, NH); 8.62 (s, 1H, NH); 7.34–6.95 (m, 5H, Ar–H); 4.35 (dd, 2H, CH2, J = 6.7/J′ = 5.8 Hz); 4.56 (dd, 2H, CH2, J = 6.8/J′ = 5.6 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 170.6 (C = O); 159.6 (C = N); 126.6, 126.1, 124.9, 122.1, 114.9, 113.5, (Ar–C); 46.3 C3 (CH2); 39.7 C2 (CH2); EIMS m/z 303.2 [M+H]+. HREIMS (m/z): 302.1049, (calcd for. C10H11ClN4O3S 302.75); Anal. Calcd for C10H11ClN4O3S C, 39.67; H, 3.66; S, 10.59; Cl, 11.71. Found C, 39.63; H, 3.56; N, 18.39; S, 10.64; Cl, 11.80.

1-[1-(4-Methylphenyl-4,5-dihydro-1H-imidazoline]-3-chlorosulfonylurea (3b)

From a general procedure with 1.75 g of 1b and 1.41 g of 2, obtaining 2.59 g of 3b (82% yield), white crystalline solid, mp 206–208 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.19 (s, 1H, NH); 8.72 (s, 1H, NH); 7.51–7.05 (m, 5H, Ar–H); 4.24 (dd, 2H, CH2, J = 6.8/J′ = 5.7 Hz); 4.52 (dd, 2H, CH2, J = 6.8/J′ = 5.5 Hz); 1.82 (s, 3H, CH3); 13C NMR (DMSO-d6, 75 MHz): δ = 170.8 (C = O); 165.3 (C = N); 128.6, 128.2, 125.1, 124.1, 116.4, 115.94, (Ar–C); 47.3 C3 (CH2); 42.6 C2 (CH2); 18.5 CH3; EIMS m/z 317.3321 [M+H]+. HREIMS (m/z): 316.8 [M+] (calcd. for C11H13ClN4O3S 316.78); Anal. Calcd for C11H13ClN4O3S C, 41.70; H, 4.13; N, 17.68; Cl, 11.19; S, 10.12. Found C, 41.61; H, 4.21; N, 17.61, Cl 11.08; S, 10.04.

1-[1-(2-Methoxyphenyl-4,5-dihydro-1H-imidazoline]-3-chlorosulfonylurea (3c)

From a general procedure with 1.91 g of 1c and 1.41 g of 2, obtaining 2.46 g of 3a (74% yield), white crystalline solid, mp 183–184 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.11 (s, 1H, NH); 8.79 (s, 1H, NH); 7.44–6.91 (m, 4H, Ar–H); 4.31 (dd, 2H, CH2, J = 6.6/J′ = 5.5 Hz); 4.50 (dd, 2H, CH2, J = 6.9/J′ = 5.4 Hz); 2.12 (s, 3H, OCH3); 13C NMR (DMSO-d6, 75 MHz): δ = 171.1 (C = O); 163.6 (C = N); 129.3, 129.1, 128.3, 125.1, 122.8, 120.6, (Ar–C); 45.8 C3 (CH2); 39.7 C2 (CH2); 24.5 OCH3; EIMS m/z 333.1 [M+H]+. HREIMS (m/z): 332.1540 [M+] (calcd. for C11H13ClN4O4S 332.78), Anal. Calcd for C11H13ClN4O4S: C, 39.70; H 3.93; N, 16.83; Cl, 10.65; S, 9.63. Found C, 39.61; H, 3.98; N, 16.91; Cl, 10.75; S, 9.54.

1-[1-(4-Methoxyphenyl-4,5-dihydro-1H-imidazoline]-3-chlorosulfonylurea (3d)

From a general procedure with 1.91 g of 1d and 1.41 g of 2, obtaining 1.39 g of 3d (42% yield), white crystalline solid, mp 197–199 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.22 (s, 1H, NH); 8.56 (s, 1H, NH); 7.11–6.83 (m, 4H, Ar–H); 7.31 (s, 1H, NH); 4.30 (dd, 2H, CH2, J = 7.1/J′ = 5.9 Hz); 4.61 (dd, 2H, CH2, J = 6.9/J′ = 5.8 Hz); 2.68 (s, 3H, OCH3); 13C NMR (DMSO-d6, 75 MHz): δ = 171.2 (C = O); 161.5 (C = N); 128.6, 123.1, 122.6, 120.1, 115.4, 113.1, (Ar–C); 45.8 C3 (CH2); 39.3 C2 (CH2); 23.2 OCH3; EIMS m/z 333.1 [M+H]+. HREIMS (m/z): 332.2349 [M+] (calcd for C11H13ClN4O4S 332.78), Anal. Calcd for C11H13ClN4O4S: C, 39.70; H, 3.93; N, 16.83; Cl, 10.65; S, 9.63. Found C, 39.68; H, 3.85; N, 16.93; Cl, 10.55; S, 9.71.

1-[1-(3-chlorophenyl-4,5-dihydro-1H-imidazoline]-3-chlorosulfonylurea (3e)

From a general procedure with 1.96 g of 1e and 1.41 g of 2, obtaining 1.52 g of 3e (45% yield), white crystalline solid, mp 141–142 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.41 (s, 1H, NH); 8.93 (s, 1H, NH); 7.60–7.15 (m, 4H, Ar–H); 4.26 (dd, 2H, CH2, J = 6.8/J′ = 5.5 Hz); 4.54 (dd, 2H, CH2, J = 6.4/J′ = 5.6 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 171,3 (C = O); 162.4 (C = N); 129.6, 123.1, 122.5, 121.1, 119.5, 117.1, (Ar–C); 46.9 C3 (CH2); 41.7 C2 (CH2); EIMS m/z 338.2 [M+H]+. HREIMS (m/z): 337.1289 [M+] (calcd. for C10H10Cl2N4O3S 337.20) Anal. Calcd for C10H10Cl2N4O3S: C, 35.61; H, 2.98; N, 16.61; Cl, 21.03; S, 9.51. Found C, 35.69; H, 3.05; N, 16.79; Cl, 21.07; S, 9.60.

1-[1-(4-chlorophenyl-4,5-dihydro-1H-imidazoline]-3-chlorosulfonylurea (3f)

From a general procedure with 1.96 g of 1f and 1.41 g of 2, obtaining 2.16 g of 3f (64% yield), white crystalline solid, mp 160–162 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 9.17 (s, 1H, NH); 8.82 (s, 1H, NH); 7.36–7.05 (m, 4H, Ar–H); 4.39 (dd, 2H, CH2, J = 6.5/J′ = 5.6 Hz); 4.76 (dd, 2H, CH2, J = 6.9/J′ = 5.6 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 170.9 (C = O); 162.7 (C = N); 129.8, 129.1, 125.9, 122.8, 122.4, 115.3, (Ar–C); 47.8 C3 (CH2); 40.4 C2 (CH2); EIMS m/z 338.2 [M+H]+. HREIMS (m/z): 337.1633 [M+] (calcd. for C10H10Cl2N4O3S 337.20) Anal. Calcd. for C10H10Cl2N4O3S: C, 35.61; H, 2.98; N, 16.61; Cl, 21.03; S, 9.51. Found C, 35.51; H, 2.94; N, 16.68, Cl, 21.15; S, 9.59.

General procedure for synthesis 1-aryl-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]triazines (4a4f)

A mixture containing appropriate 1-(1-aryl-2-amine-4,5-dihydro-1H-imidazoline)-3-chlorsulfonylurea (3a3f) (0.01 mol) and 0.01 mol (1.6 g) of 1,1′-carbonyldiimidazole (CDI) was dissolved in 50 mL of DMF (N,N-dimethlformamide) and heated under reflux for 6 h. The solvent was removed by the low-pressure evaporation and the resulting solid was crystallized from methanol:propan-2-ol (1:1) mixture.

1-Phenyl-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]triazine (4a)

From a general procedure with 3.03 g of 3a and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 1.68 g of 4a (51% yield), white crystalline solid, mp 180–182 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.40–6.85 (m, 5H, Ar–H); 4.26 (dd, 2H, CH2, J = 6.9/J′ = 5.6 Hz); 4.65 (dd, 2H, CH2, J = 6.8/J′ = 5.5 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 171.2 (C = O); 170.5 (C = O); 162.6 (C = N); 129.7, 129.1, 127.1, 121.8, 121.4, 119.7, (Ar–C); 45.8 C3 (CH2); 39.9 C2 (CH2); EIMS m/z 329.7 [M+H]+. HREIMS (m/z): 329.1330 [M+] (calcd for C11H9ClN4O4S 328.75) Anal. Calcd for C11H9ClN4O4S: C, 40.19; H, 2.76; N, 17.04; Cl, 10.78; S, 9.75. Found C, 40.11; H, 2.62; N, 17.09; Cl, 10.85; S, 9.64.

1-(4-Methylphenyl)-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]-triazine (4b)

From a general procedure with 3.16 g of 3b and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 2.70 g of 4b (79% yield), white crystalline solid, mp 198–200 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.12–6.67 (m, 4H, Ar–H); 4.41 (dd, 2H, CH2, J = 7.1/J′ = 5.9 Hz); 4.69 (dd, 2H, CH2, J = 6.9/J′ = 5.6 Hz); 1.56 (s, 3H, CH3); 13C NMR (75 MHz, DMSO-d6): δ = 172.1 (C = O); 170.7 (C = O); 161.5 (C = N); 129.6, 128.4, 128.4, 127.2, 122.3, 114.6, (Ar–C); 44.6 C3 (CH2); 38.2 C2 (CH2); 14.6 CH3; EIMS m/z 343.7 [M+H]+. HREIMS (m/z): 342.3245 [M+] (calcd for C12H11ClN4O4S 342.78); Anal. Calcd for C12H11ClN4O4S: C, 42.04; H, 3.23; N, 16.34; Cl, 10.34; S, 9.35. Found C, 42.11; H, 3.31; N, 16.39; Cl, 10.11; S, 9.31.

1-(2-Methyoxyphenyl)-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]-triazine (4c)

From a general procedure with 3.59 g of 3c and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 2.16 g of 4c (64% yield), white crystalline solid, mp 212–214 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.54–6.95 (m, 4H, Ar–H); 4.33 (dd, 2H, CH2, J = 6.9/J′ = 5.5 Hz); 4.56 (dd, 2H, CH2, J = 6.8/J′ = 5.4 Hz); 3.22 (s, 3H, OCH3); 13C NMR (DMSO-d6, 75 MHz): δ = 171.4 (C = O); 171.4 (C = O); 159.6 (C = N); 127.6, 127.1, 124.9, 121.1, 120.6, 119.1, (Ar–C); 46.8 C3 (CH2); 41.5 C2 (CH2); 19.9 (OCH3); EIMS m/z 359.3 [M+H]+. HREIMS (m/z): 358.1255 [M+] (calcd for C12H11ClN4O5S 358.78), Anal. Calcd for C12H11ClN4O5S: Calcd C, 40.17; H, 3.09; N, 15.62; Cl, 9.88; S, 8.94. Found C, 40.11; H, 3.18; N, 15.49; Cl, 9.80; S, 8.84.

1-(2-Methyoxyphenyl)-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]-triazine (4d)

From a general procedure with 3.59 g of 3d and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 2.16 g of 4d (64% yield), white crystalline solid, mp 219–220 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.20–6.85 (m, 4H, Ar–H); 4.39 (dd, 2H, CH2, J = 6.8/J′ = 5.6 Hz); 4.61 (dd, 2H, CH2, J = 6.8/J′ = 5.7 Hz); 3.23 (s, 3H, OCH3); 13C NMR (DMSO-d6, 75 MHz): δ = 171.8 (C = O); 170.7 (C = O); 160.2 (C = N); 125.1, 123.9, 122.6, 121.8, 121.56, 118.2, (Ar–C); 45.6 C3 (CH2); 40.1 C2 (CH2); 18.6 (OCH3); EIMS m/z 359.8 [M+H]+. HREIMS (m/z): 358.1120 [M+] (calcd. for C12H11ClN4O5S 358.78); Anal. Calcd for C12H11ClN4O5S: C, 40.17; H, 3.09; N, 15.62, Cl, 9.88; S, 8.94%. Found C, 40.05; H, 3.19; N, 15.68; Cl, 9.95; S, 8.78.

1-(3-Chlorophenyl)-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]-triazine (4e)

From a general procedure with 3.27 g of 3e and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 2.21 g of 4e (61% yield), white crystalline solid, mp 201–202 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.60–7.15 (m, 4H, Ar–H.); 4.20 (dd, 2H, CH2, J = 6.8/J′ = 5.8 Hz); 4.56 (dd, 2H, CH2, J = 6.8/J′ = 5.6 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 173.4 (C = O); 171.7 (C = O); 160.5 (C = N); 126.6, 123.4, 122.3, 120.9, 119.7, 114.6, (Ar–C); 45.9 C3 (CH2); 42.1 C2 (CH2); EIMS m/z 364.1 [M+H]+. HREIMS (m/z): 363.9861 [M+] (calcd. for C11H8Cl2N4OS 363.20); Anal. Calcd for C11H8Cl2N4OS 363.20: C, 36.37; H, 2.22; N, 15.43; Cl, 19.52; S, 8.82. Found C, 36.44; H, 2.35; N, 15.51; Cl 19.40; S, 8.75.

1-(4-Chlorophenyl)-6-chlorosulfonyl-5,7(1H)dioxo-2,3-dihydroimidazo[1,2-a][1,3,5]-triazine (4f)

From a general procedure with 3.27 g of 3f and 1.6 g of 1,1′-carbonylodiimidazole, obtaining 2.16 g of 4f (53% yield), white crystalline solid, mp 231–232 °C; 1H NMR (DMSO-d6, 300 MHz) δ = 7.58–7.05 (m, 4H, Ar–H); 4.31 (dd, 2H, CH2, J = 7.1/J′ = 5.9 Hz); 4.59 (dd, 2H, CH2, J = 6.8/J′ = 5.8 Hz); 13C NMR (DMSO-d6, 75 MHz): δ = 171.8 (C = O); 170.1 (C = O); 158.4 (C = N); 125.6, 125.1, 123.9, 123.2, 120.5, 117.1, (Ar–C); 46.7 C3 (CH2); 40.4 C2 (CH2); EIMS m/z 364.2 [M+H]+. HREIMS (m/z): 363.1423 [M+] (calcd. for C11H8Cl2N4OS 363.20), Anal. Calcd for C11H8Cl2N4OS: Calcd C, 36.37; H, 2.22; N, 15.43; Cl, 19.52; S, 8.82. Found C, 36.50; H, 2.45; N, 15.39; Cl, 19.47; S, 8.89.

Molecular modeling

The investigated compounds were modeled using the LigPrep protocol from the Schrödinger SuiteCitation18. In order to sample different protonation states of ligands in physiological pH, Epik module was usedCitation19. The compounds were further optimized using Hartree–Fock approach and 6–31 g (d,p) basis set of Spartan 10Citation20. Parameters to evaluate drug-likeness were calculated using VegaZZ version 3.0.1Citation21 (number of atoms), Discovery Studio version 3.1Citation22 (molar mass, number of rings, lipophilicity, number of rotatable bonds), ACDLabs (molar refractivity, number of hydrogen bond donors and acceptors) and the Schrödinger Suite (a number of rigid bonds) as described previouslyCitation23–26. ADMET parameters were calculated with Discovery Studio 3.1 (solubility, blood–brain permeation) or Osiris Property ExplorerCitation27 (toxicity risks) as previously describedCitation23–26. The prediction of toxicity is based on a pre-computed set of structural fragment that give rise to toxicity alerts in case they are encountered in the investigated compoundCitation27. These fragment lists were obtained by rigorously shreddering all compounds of the RTECS database known to responsible for a certain toxicity classCitation27. During the shreddering, any molecule was first cut at every rotatable bonds leading to a set of core fragmentsCitation27. These were applied to reconstruct all possible bigger fragments being a substructure of the investigated compound. Next, a substructure search process determined the occurrence frequency of any fragment (core and constructed fragments) within all compounds of that toxicity class. It also determined these fragment's frequencies within the structures of more than 3000 commercial drugsCitation27. Based on the assumption that drugs present on the market are free of toxic effects, any fragment was considered a risk factor if it occurred often as substructure of harmful compounds but never or rarely in traded drugsCitation27. The predicted toxicity of 1.0 means the lack of a toxic effect and decreasing value means increasing risk of toxicity. For structure–activity relationship studies, HOMO and LUMO energies, lipophilicity and polarizability were calculated with Discovery Studio 3.1Citation23 HOMO and LUMO orbitals as well as a map of the electrostatic potential (ESP) onto a surface of the electron density were visualized with ArgusLabCitation28. Molecular surface area, polar surface area, molecular volume and ovality were calculated with VegaZZCitation21.

Antiviral activity

Antiviral activity assay

Antiviral activity assay were similar for HSV-1 and CBV3. After 24 h incubation, the cell culture was infected with viruses in the dose of 100 TCID50/mL. After 1 h incubation at 37 °C, the suspension of the viruses was removed and the media with 2% of serum together with the tested compounds in the non-toxic concentration were added to the cell cultures. The viruses diluted in the culture media without tested compounds were used as a control. Acyclovir (Polpharma SA, Starogard Gdański, Poland) and ribavirin (Meduna Pharma GmbH, Isernhagen, Germany) were used as reference compounds. After 48 h incubation at 37 °C, the cells were frozen and after thawing, the viruses were titrated in the Vero cell culture. The cytopathic effect (CPE) of the viruses was examined by a light microscope and the titre of virus was estimated according to the Reed–Muench methodCitation29. Viral titres were determined by tissue culture infection dose (TCID50) assays.

Cells and viruses

The Vero cell (ECACC No. 84113001-established from the kidney of a normal adult African Green monkey) was used in the experiment. The media in the culture (Dulbecco’s Modified Eagle Medium – DMEM, Sigma-Aldrich, St. Louis, MO), 100 U/mL of penicillin and of streptomycin (Polfa Tarchomin, Warsaw, Poland). The cell culture was incubated at 37 °C in the 5% CO2 atmosphere. For antiviral activity of examined compounds the HSV-1 (ATCC No. VR-260) and CVB3 (ATCC No. VR-30) from the American Type Culture Collection were used. The viruses were propagated in the Vero cell culture. Virus stocks were stored at −70 °C until used.

Cytotoxicity assay

Compounds were dissolved in dimethyl sulfoxide – DMSO (POCH, Gliwice, Poland) in the concentration of 50 mg/mL and further diluted with a complete test medium. About 100 µl of the Vero cell culture prepared was seeded into 96-well plastic plates (Becton Dickinson and Company, Franklin Lakes, NJ) at a cell density 1.5 × 104 cells per well. After 24 h incubation at 37 °C the media were removed and the cells were treated with a solution of the examined substance diluted in the media with 2% of serum. The cells were submitted to a series of compound concentrations, from 1000 µg/mL to 1.9 µg/mL. Two-fold serial dilutions of compounds were added to the cells in triplicates. The cell cultures were incubated for 72 h at 37 °C in the 5% CO2 atmosphere.

Cytotoxicity of tested compounds was estimated with the use of the MTT method, described by Takenouchi and MunekataCitation30. The MTT method is a quantitative colorimetric toxicity test, based on the transformation of yellow, soluble tetrazolium salts (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) to purple-blue insoluble formasane. This process occurs naturally in mitochondria of living cells. After 72 h incubation with compounds, cell cultures were supplemented with 10 µl per well of 5 mg/mL MTT (Sigma-Aldrich, St. Louis, MO) stock in PBS (BIOMED, Lublin, Poland), and the incubation was continued for 4 h at 37 °C. Then, 100 µl of aqueous solution containing 50% dimethylformamide (POCH, Gliwice, Poland) and 20% SDS (Sigma-Aldrich, St. Louis, MO) to solubilize the insoluble formasane precipitates produced by MTT was added. After the all-night incubation, the absorbance was measured by the Epoch plate reader (BioTek, Winooski, VT) at two wavelengths – 540 and 620 nm. Based on the test results, the cytotoxic concentration (CC50), which is the amount of tested substance that is required to reduce the number of viable cells by 50% compared to the control culture, was determined and was calculated using the Gen 5 2.01.14 software (BioTek, Winooski, VT). Cell viability (%) was calculated as (A540/620 of the treated/A540/620 of the control) × 100. When the CC50 value was calculated, non-toxic concentrations of examined compounds were selected to assess their antiviral activity. The investigation was carried out in triplicate.

Results and discussion

Chemistry

New derivatives 3a3f were obtained in the reaction of 1-aryl-4,5-dihydro-1H-imidazol-2-amines 1a1f with chlorosulfonyl isocyanate 2 in the dichloromethane solution and under nitrogen atmosphere (). The second series of compounds 4a4f was obtained from the respective 1-(1-aryl-4,5-dihydro-1H-imidazolin)-3-chlorosulfonylureas 3a3f and 1,1′-carbonyldiimidazole (CDI) in DMF (N,N-dimethlformamide) solution.

Estimation of drug-likeness

The descriptors applied for estimation of drug-likeness are presented in . Drug-likeness was assessed using Lipinski’s rule as well as the placement of the investigated compounds in the chemical space determined by the databases of the pharmacologically active compounds (CMC, Comprehensive Medicinal Chemistry Database, containing about 7000 compounds and MDDR, MACCS-II Drug Data Report, containing about 100 000 compounds) according to the methodology of PREADMETCitation31 service as described previouslyCitation23–26. Regarding Lipinski’s rule, all the compounds possess the molar mass below 500, the number of hydrogen bond donors below 5, the number of hydrogen bond acceptors below 10 and the lipophilicity below 5Citation32.

Table 1. Parameters for drug-likeness estimation.

Regarding subsequent criteria of drug-likeness, most compounds collected in the CMC database has lipophilicity from −0.4 to 5.6, molar refractivity in the range of 40–130 the number of atoms from 20 to 70 and molar mass from 160 to 480Citation23–26,Citation33. All the investigated compounds fulfill the first three criteria. Concerning the compounds in MDDR database, the drug-like substances have the number of rings equal or greater than 3, the number of rigid bonds equal or greater than 18, and the number of rotatable bonds equal or greater than 6Citation23–26,Citation34. Compounds 3a3f have too few rings and all the compounds have too few rotatable bonds. The criterion concerning rigid bonds is fulfilled by all the compounds.

Prediction of ADMET properties

In order to facilitate the selection of compounds for antiviral activity assessment, some ADMET parameters were calculated (). The plot presented in confirms that most of the tested compounds possess reasonably favorable ADMET properties. Comparing the plot in with lipophilicity values from and polar surface areas from , it can be concluded that compounds from 3c and 3d have less favorable blood–brain permeation properties. All compounds are well absorbed () and well soluble in water as they have of logS over −4 (solubility expressed in mol/dm3)Citation27. All the compounds were predicted to have low mutagenicity risk and possible irritating effects (0.8), however no risk of tumorigenicity and no reproductive effects were predicted (1.0; ).

Figure 3. ADMET properties of the studied compounds.

Figure 3. ADMET properties of the studied compounds.

Table 2. Solubility and toxicity of the investigated compounds.

Antiviral activity determination

The obtained compounds 3d and 4a4d have been studied as potential antiviral agents against HSV-1 and CVB3 and revealed their high efficacy as inhibitors of virus reproduction. The selection of compounds for antiviral activity determination was justified by availability from synthesis as well as our earlier experience. Acyclovir was used as a reference compound for HSV-1 and ribavirin for CVB3. The influence of 3d and 4a4d on the Vero cell culture after incubation for 72 h is presented in .

Table 3. Cytotoxicity of compounds 3d, 4a, 4b, 4c, 4d.

DMSO used as an eluent for examined compounds in the tested concentration had no toxic effect on cell cultures. All compounds were evaluated for their cytotoxicity on cell line by a standard MTT assay. CC50 values of compounds 3d, 4a, 4b and 4d were contained within the range of 650.07–840.93 µg/mL, except for compound 4c which showed cytotoxicity with CC50 of 90.08 µg/mL. Compounds were tested for in vitro antiviral activity using the cytopathic effect (CPE) inhibitory assay. The results are presented in .

Table 4. Antiviral activity of the compounds against HSV-1 and CVB3.

Table 5. Molecular descriptors for structure–activity determination.

Non-cytotoxic concentrations of 31, 62, 250, 500, 625 and 800 µg/mL were used for testing the antiviral activity of compounds. The research demonstrated that compound 3d in the concentration of 800 µg/mL influenced the HSV-1 and CVB3 replication by reducing the virus replication level by 2.76 log and 2.08 log, which resulted in reducing the titre by 43.3 and 25%, respectively. The compound 4a in all tested concentrations (500 and 625 µg/mL) demonstrated the antiviral activity. This derivative caused the decrease in the titre of HSV-1 by 2.29 log and 2.18 log, respectively (which corresponds to the following percentage levels of inhibition: 35.9 and 34.2%). In the case of compound 4c, tested in the concentration of 62 µg/mL, the titre of CVB3 was decreased by 2.99 log (35.9%). Whereas compound 4d was active only against HSV-1 it caused the decrease in the virus titre by 2.48 log (38.9%). Compound 4b did not demonstrate any significant antiviral activity. Thus, there is a need of further research to estimate the influence of 3d and 4a4c on the HSV-1 and CVB3 inactivation, inhibition of viral adsorption and penetration. Prospectively, study of the mechanisms underlying their antiviral activity is necessary for the development of antiviral therapeutics to treat patients infected with HSV-1 and CVB3.

Structure–activity relationship

Molecular descriptors for structure–activity determination are presented in . The active compound 3d (active against both viruses) compared to inactive compound 4b is characterized by greater surface area, greater polar surface area and also bigger volume. This compound has also the biggest ovality and the smallest lipophilicity value, the biggest HOMO energy and small polarizability in the whole set. In the series of compounds 4a4d, the active compound 4a (active against Herpes simplex virus) is characterized by the smallest surface area, the smallest volume, molecular weight, polarizability and one of the smallest polar surface area. Simultaneously, it has one of the smallest lipophilicity value. In contrast, another compound active against Herpes simplex virus, 4d has rather big surface area, volume, molecular weight, polarizability and polar surface area but it has still low lipophilicity value. It can be thus generalized that low lipohilicity value favors activity against Herpes simplex in both series. Furthermore, lack of the substituent in phenyl group or 4-methoxy substitution can be considered as the most beneficial for the antiviral activity against this virus. Compound 4c was active against CVB3, which can be also attributed to low lipophilicity value. depicts HOMO and LUMO orbitals for most active compounds 3d and 4a whereas presents maps of electrostatic potential for these derivatives. It can be concluded that for compound 3d HOMO orbital is mainly located on the phenyl ring and imidazole system while LUMO orbital on the chlorosulfonyl group. In the case of compound 4a, HOMO orbital is located on the phenyl group and LUMO orbital in the chlorosulfonyl group. The negative charge is gathered on heteroatoms, mainly on nitrogen atoms, oxygen atoms of the chlorosulfonyl group (for 3d and 4a) and the methoxy group of 3d.

Figure 4. HOMO (A, C) and LUMO (B, D) orbitals for 3d (A, B) and 4a (C, D).

Figure 4. HOMO (A, C) and LUMO (B, D) orbitals for 3d (A, B) and 4a (C, D).

Figure 5. The map of the electrostatic potential (ESP) onto a surface of the electron density for 3d (A) and 4a (B).

Figure 5. The map of the electrostatic potential (ESP) onto a surface of the electron density for 3d (A) and 4a (B).

Conclusions

In this study, we obtained 12 novel N-substituted derivatives of 1-arylimidazolidyn-2-ylideneurea and tested selected compounds for their activity against Herpes simplex virus and CVB3. It was determined that four derivatives, i.e. 3d, 4a, 4c and 4d are active against these viruses. Their favorable activity in comparison to other tested compounds can be primarily attributed to their low lipophilicity values. Moreover, the lack of substituent in the phenyl moiety or 4-methoxy substitution can be considered as the most beneficial for the antiviral activity. In the subsequent studies, we will work on the molecular mechanism of antiviral activity.

Acknowledgements

The paper was developed using the equipment purchased within the project “The equipment of innovative laboratories doing research on new medicines used in the therapy of civilization and neoplastic diseases” within the Operational Program Development of Eastern Poland 2007–2013, Priority Axis I modern Economy, operations I.3 Innovation promotion.

Declaration of interest

Part of the calculations of this study was performed under a computational grant by Interdisciplinary Center for Mathematical and Computational Modelling (ICM), Warsaw, Poland, grant number G30-18 and under resources and licenses by CSC, Finland.

The research was partially performed during the postdoctoral fellowship of Agnieszka A. Kaczor at University of Eastern Finland, Kuopio, Finland under Marie Curie fellowship.

References

  • Cao T, Xing Y, Yang Y, Mei H. Correlation between matrix metalloproteinase expression and activation of the focal adhesion kinase signaling pathway in herpes stromal keratitis. Exp Ther Med 2014;7:280–6
  • Bove D, Lupoli A, Caccavale S, et al. Dermatological and immunological conditions due to nerve lesions. Funct Neurol 2013;28:83–91
  • Steiner I, Benninger F. Update on Herpes Virus infections of the nervous system. Curr Neurol Neurosci Rep 2013;13:414–20
  • Kolokotronis A, Doumas S. Herpes simplex virus infection, with particular reference to the progression and complications of primary herpetic gingivostomatitis. Clin Microbiol Infect 2006;12:202–11
  • He DX, Tam SC. Trichosanthin affects HSV-1 replication in Hep-2 cells. Biochem Biophys Res Commun 2010;402:670–5
  • Ju HQ, Xiang YF, Xin BJ, et al. Synthesis and in vitro anti-HSV-1 activity of a novel Hsp90 inhibitor BJ-B11. Bioorg Med Chem Lett 2011;21:1675–7
  • Alekseev O, Donovan K, Azizkhan-Clifford J. Inhibition of ataxia telangiectasia mutated (ATM) kinase suppresses Herpes Simplex virus type 1 (HSV-1) keratitis. J Invest Ophthalmol Vis Sci 2014;55:706–15
  • Yajima T, Knowlton KU. Viral myocarditis: from the perspective of the virus. Circulation 2009;119:2615–24
  • Kemball CC, Harkins S, Whitmire JK, et al. Coxsackievirus B3 inhibits antigen presentation in vivo, exerting a profound and selective effect on the MHC class I pathway. PLoS Pathog 2009;5:e1000618
  • Tang S, Kong L, Li Y, et al. Novel N-benzenesulfonyl sophocarpinol derivatives as coxsackie B virus inhibitors. ACS Med Chem Lett 2015;6:183–6
  • Yin D, Li J, Lei X, et al. Antiviral activity of total flavonoid extracts from Selaginella moellendorffii hieron against Coxsackie Virus B3 in vitro and in vivo. Evid Based Complement Alternat Med 2014;2014:950817
  • Albulescu L, Strating JR, Thibaut HJ, et al. Broad-range inhibition of enterovirus replication by OSW-1, a natural compound targeting OSBP. Antiviral Res 2015;117:110–14
  • Strating JR, van der Linden L, Albulescu L, et al. Itraconazole inhibits enterovirus replication by targeting the oxysterol-binding protein. Cell Rep 2015;10:600–15
  • Rządkowska M, Szacoń E, Kaczor AA, et al. 407102 (Polish patent claim)
  • Szacoń E, Rządkowska M, Rajtar B, et al. 407103 (Polish patent claim)
  • Saczewski F, Debowski T, Szmigiel J. Synthesis and transformations of 2-amino-1(4,5-dihydro-1H-imidazol-2-yl)benzimidazoles. A route to 2,3-dihydrobenzo[4,5]-imidazo[1,2-a][1,3,5]triazine ring system. Heterocycles 1998;48:1879–85
  • Saczewski F, Kuchnio A, Samsel M, et al. Synthesis of novel aryl(heteroaryl)sulfonyl of possible biological interest. Molecules 2010;15:1113–26
  • LigPrep, Version 2.4: Schrödinger, LLC, New York; 2010
  • Epik, Version 2.1: Schrödinger, LLC, New York; 2010
  • Spartan 10. Wavefunction, Inc., Irvine, CA, USA. Available from: www.wavefun.com
  • Pedretti A, Villa L, Vistoli G. VEGA – an open platform to develop chemo-bio-informatic applications, using plug-in architecture and script programming. J Comput Aided Mol Des 2004;18:167–73
  • Discovery Studio 3.1, Accelrys
  • Rządkowska M, Szacoń E, Kaczor AA, et al. Synthesis, pharmacological activity and molecular modeling of 1-aryl-7-hydroxy-2,3-dihydroimidazo[1,2-a]pyrimidine-5(1H)-ones and their 6-substituted derivatives. Med Chem 2014;10:460–75
  • Rządkowska M, Szacoń E, Kaczor AA, et al. Synthesis, central nervous system activity, and structure–activity relationship of 1-aryl-6-benzyl-7-hydroxy-2,3-dihydroimidazo[1,2-a]pyrimidine-5(1H)-ones. Med Chem Res 2014;23:4221–37
  • Szacoń E, Rządkowska M, Kaczor AA, et al. Synthesis, central nervous system activity and structure-activity relationship of N-substituted derivatives of 1-arylimidazolidyn-2-ylideneurea and products of their cyclization. J Enzyme Inhib Med Chem 2015. [Epub ahead of print]. doi:10.3109/14756366.2014.965699
  • Rządkowska M, Szacoń E, Kaczor AA, et al. Synthesis and antiviral activity of 1-(1,3-disubstitutedimidazolidyn-2-ylidene)-3-ethoxycarbonylmethylurea derivatives. J Enzyme Inhib Med Chem 2015. [Epub ahead of print]. doi:10.3109/14756366.2015.1022172
  • Osiris Property Explorer. Available from: http://www.organic-chemistry.org/prog/peo/logS.html
  • http://www.arguslab.com/arguslab.com/ArgusLab.html [last accessed 20 Dec 2014]
  • Reed LJ, Muench MA. A simple method of estimating fifty percent end points. Am J Hyg 1938;27:493–7
  • Takenouchi T, Munekata E. Amyloid beta-peptide-induced inhibition of MTT reduction in PC12h and C1300 neuroblastoma cells: effect of nitroprusside. Peptides 1998;19:365–72
  • Lee SK, Chang GS, Lee IH, et al. The PreADME:pc-based program for batch prediction of adme properties, EuroQSAR 2004, 9.5-10, Istanbul, Turkey
  • Walters WP. Going further than Lipinski's rule in drug design. Expert Opin Drug Discov 2012;7:99–107
  • Oprea TI. Property distribution of drug-related chemical databases. J Comput Aid Mol Des 2000;14:251–64
  • Brown RD. Tools for designing diverse, drug like, cost-effective combinatorial libraries. In: Ghose A, Viswanadhan V, eds. Combinatorial library design and evaluation. New York: Marcel Dekker Inc.; 2001

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.