Skip to content
Publicly Available Published by De Gruyter November 10, 2016

Synthesis, in-vitro cytotoxicity of 1H-benzo[f]chromene derivatives and structure–activity relationships of the 1-aryl group and 9-position

  • Hany M. Mohamed , Ahmed M. Fouda , Essam S.A.E.H. Khattab , Ahmed M. El- Agrody and Tarek H. Afifi EMAIL logo

Abstract

A series of 1H-benzo[f]chromene-2-carbonitriles was synthesized and evaluated for their cytotoxic activities against MCF-7, HCT-116, and HepG-2 cancer cells. The SAR studies reported that the substitution in the phenyl ring at 1-position of 1H-benzo[f]chromene nucleus with the specific group, H atom, or methoxy group at 9-position increases the ability of the molecule against the different cell lines.

1 Introduction

One of the main objectives of organic and medicinal chemistry is the design and synthesis of molecules having as much value as human therapeutic agents. The benzochromene nucleus has been emerged as a promising and attractive scaffold in the development of potent antitumor agents and the treatment of human diseases. For example, Crolibulin™ (A) is currently in Phase I/II clinical trials for the treatment of advanced solid tumors [1], 2-amino-4-(3-nitrophenyl)-4H-benzo[h]chromene-3-carbonitrile (B), which is an inhibitor of diabetes-induced vascular dysfunction [2], 2-amino-5-oxo-4-phenyl-4,5-dihydropyrano[3,2-c]chromene-3-carbonitrile (C) that served as precursor for the blood anticoagulant warfarin [3], 4-substituted-2-(N-succinimido)-4H-benzo[h]chromene-3-carbonitrile (D) have shown anti-rheumatic activity [4] as shown in Figure 1. In the view of the aforementioned premises, and in a continuation of our interest in the synthesis of 2-amino-4H-chromenes and 2-amino-4H-benzochromenes with anticipated anticancer activity [5], [6], [7], [8], [9], [10], [11], [12], we aimed herein to report the microwave irradiation synthesis of two series of 3-amino-1-aryl-1H-benzo[f]chromene-2-carbonitrile (4a–h) and 3-amino-1-aryl-9-methoxy-1H-benzo[f]chromene-2-carb-onitrile (6a–h) derivatives. This article also explores the in vitro antiproliferative activity of the target compounds toward three cancer cell lines in addition to their structure–activity relationships of the substituent at 1- and 9-positions.

Figure 1: Structures of some benzochromenes with diverse biological and pharmacological activities.
Figure 1:

Structures of some benzochromenes with diverse biological and pharmacological activities.

2 Materials and methods

2.1 Analytical methods

Commercial-grade solvents and reagents were purchased from Sigma–Aldrich (St. Louis, MO, USA) and used without further purification. Melting points were measured with a Stuart Scientific (UK) apparatus and are uncorrected. IR spectra were determined as KBr pellets on a Jasco FT/IR 460 plus spectrophotometer (Jasco, Japan). 1H-NMR and 13C-NMR spectra were recorded using a Bruker AV 500 MHz spectrometer (Bruker, USA). 13C-NMR spectra were obtained using distortion-free enhancement by polarization transfer (DEPT) and the attached proton test (APT). Chemical shifts (δ) are expressed in parts per million (ppm). The MS was measured using a Shimadzu GC/MS-QP5050A spectrometer (Shimadzu, Japan). Elemental analyses were carried out at the Regional Centre for Mycology & Biotechnology (RCMP), Al-Azhar University, Cairo, Egypt, and the results were within ±0.25%. Analytical thin layer chromatography (TLC) on silica gel-precoated F254 Merck plates to check the purity of the compounds.

2.2 Chemistry

The method adopted for the synthesis of 3-amino-1-aryl-1H-benzo[f]chromene-2-carbonitriles (4a–h) is depicted in Scheme 1. Synthesis was initiated by reacting 2-naphthol (1) with a mixture of appropriate aromatic aldehydes (2a–h) and malononitrile (3) in ethanolic piperidine solution under microwave irradiation conditions for 2 min at 140°C.

Scheme 1: Synthesis of 3-amino-1-aryl-1H-benzo[f]chromene-2-carbonitriles (4a–h) Reagents: (a) MW, 400 W, 2 min., EtOH, and piperdine.
Scheme 1:

Synthesis of 3-amino-1-aryl-1H-benzo[f]chromene-2-carbonitriles (4a–h) Reagents: (a) MW, 400 W, 2 min., EtOH, and piperdine.

In a similar manner, treatment of 7-methoxy-2-naphthol (5) with a mixture of appropriate aromatic aldehydes (2a–h) and malononitrile (3) afforded the corresponding 3-amino-1-aryl-9-methoxy-1H-benzo[f]chromene-2-carbonitriles (6a–h) as illustrated in Scheme 2.

Scheme 2: Synthesis of 3-amino-1-aryl-9-methoxy-1H-benzo[f]chromene-2-carbonitriles (6a–h) Reagents: (a) MW, 400 W, 2 min., EtOH, and piperdine.
Scheme 2:

Synthesis of 3-amino-1-aryl-9-methoxy-1H-benzo[f]chromene-2-carbonitriles (6a–h) Reagents: (a) MW, 400 W, 2 min., EtOH, and piperdine.

The maximum power of microwave irradiation was optimized by repeating the reaction at different times and watt powers. The microwave irradiations at 400 W and a reaction time of 2 min gave the highest yield. The 1-position of compounds 4a–h and 6a–h are a chiral center and all the reactions were conducted using TLC technique.

The structures of the synthesized compounds were established based on spectral data, IR, 1H-NMR, 13C-NMR, 13C-NMR-DEPT, 13C- NMR-APT, and MS data (see experimental part and supplementary information).

A reaction mixture of 2-naphthol (1) or 7-methoxy-2-naphthol (5) (2 mmol), different aromatic aldehydes (2a–h) (2 mmol), malononitrile (3) (2 mmol), and piperidine (0.5 mL) in absolute ethanol (30 mL) was heated under microwave irradiation conditions for 2 min at 140°C. After the completion of the reaction, the reaction mixture was cooled at room temperature and precipitated solid was filtered off, washed with methanol, and recrystallized from ethanol or ethanol/benzene. The physical and spectral data of compounds 4a–h and 6a–h are as follows:

2.2.1 3-Amino-1-phenyl-1H-benzo[f]chromene-2-carbonitrile (4a)

Yield 87%, mp 280–281°C (Lit. mp 279°C [13]), IR (FTIR/KBr) υmax (cm−1): 3461, 3349, 3203 (NH2), 2183 (CN). 1H-NMR (DMSO-d6) δ: 5.31 (s, 1H, H-1), 6.98 (bs, 2H, NH2), 7.94–7.16 (m, 11H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.67, 146.81, 145.67, 130.80, 130.14, 129.46, 128.66, 128.43, 127.04, 126.96, 126.56, 124.89, 123.59, 120.43, 116.75, 115.66, 57.92, 38.07. Analysis for C20H14N2O, calculated C 80.52, H 4.73, N 9.39. Found: C 80.64, H 4.84, N 9.50.

2.2.2 3-Amino-1-(4-fluorophenyl)-1H-benzo[f]chromene-2-carbonitrile (4b)

Yield 84%, mp 230–231°C (Lit. mp 230°C [14]), IR (FTIR/KBr) υ (cm−1): 3461, 3348, 3208 (NH2), 2187 (CN). 1H-NMR (DMSO-d6) δ: 5.36 (s, 1H, H-1), 7.00 (bs, 2H, NH2), 7.96–7.08 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 161.74, 159.81, 146.75, 141.94, 130.82, 130.04, 129.58, 128.84, 128.47, 127.10, 124.94, 123.56, 120.34, 116.78, 115.49, 115.47, 57.76, 37.21. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.58 (↑), 128.84 (↑), 128.47 (↑), 127.10 (↑), 124.94 (↑), 123.56 (↑), 116.78 (↑), 115.49 (↑), 37.21 (↑). In the DEPT spectrum at 90°, only CH signals are (↑) δ: 129.58 (↑), 128.84 (↑), 128.47 (↑), 127.10 (↑), 124.94 (↑), 123.56 (↑), 116.78 (↑), 115.49 (↑), 37.21 (↑). In the DEPT spectrum at 45° (CH, CH2, and CH3) (↑) δ: 129.58 (↑), 128.84 (↑), 128.47 (↑), 127.10 (↑), 124.94 (↑), 123.56 (↑), 116.78 (↑), 115.49 (↑), 37.21 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 161.74 (↓), 159.81 (↓), 146.75 (↓), 141.94 (↓), 130.82 (↓), 130.04 (C- ↓), 128.84 (↑), 128.47 (↑), 127.10 (↑), 124.94 (↑), 123.56 (↑), 120.34 (↓), 116.78 (↑), 115.49 (↓), 115.47 (↑), 57.76 (↓), 37.21 (↑), 129.58 (↑); Analysis for C20H13FN2O, calculated C 75.94, H 4.14, N 6.01. Found: C 76.11, H 4.28, N 6.15.

2.2.3 3-Amino-1-(4-chlorophenyl)-1H-benzo[f]chromene-2-carbonitrile (4c)

Yield 83%; mp 220–221°C (Lit. mp 219°C [15]), IR (FTRI/KBr) υ (cm−1): 3457, 3327, 3210 (NH2), 2194 (CN). 1H-NMR (DMSO-d6) δ: 5.37 (s, 1H, H-1); 7.04 (bs, 2H, NH2), 7.94–7.21 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.70, 146.80, 144.63, 131.16, 130.82, 130.04, 129.67, 128.81, 128.48, 128.28, 127.14, 124.97, 123.51, 120.29, 116.77, 115.13, 57.46, 37.37. Analysis for C20H13ClN2O, calculated C 72.18, H 3.94, N 8.42. Found: C 72.27, H, 4.09, N 8.53.

2.2.4 3-Amino-1-(4-bromophenyl)-1H-benzo[f]chromene-2-carbonitrile (4d)

Yield 81%, mp 242–243°C (Lit. mp 241°C [16]), IR (FTRI/KBr) υ (cm−1): 3462, 3327, 3212 (NH2), 2195 (CN). 1H-NMR (DMSO-d6) δ: 5.35 (s, 1H, H-1), 7.04 (bs, 2H, NH2), 7.94–7.15 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.70, 146.80, 145.05, 131.59, 130.81, 130.03, 129.68, 128.49, 128.28, 127.16, 124.97, 123.50, 120.28, 119.66, 116.77, 115.06, 57.39, 37.45. Analysis for C20H13BrN2O, calculated C 63.68, H 3.47, N 7.43. Found: C 63.56, H 3.35, N 7.31.

2.2.5 3-Amino-1-(4-methylphenyl)-1H-benzo[f]chromene-2-carbonitrile (4e)

Yield 81%; mp 275–276°C (Lit. mp 270°C [17]), IR (KBr) υ (cm−1): 3428, 3336, 3215 (NH2), 2188 (CN). 1H-NMR (DMSO-d6) δ: 2.20 (s, 3H, CH3), 5.25 (s, 1H, H-1), 6.95 (bs, 2H, NH2), 7.94–7.07 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.57, 146.73, 142.78, 135.65, 130.78, 130.16, 129.37, 129.20, 128.40, 126.99, 126.86, 124.85, 123.63, 120.48, 116.74, 115.77, 58.03, 37.71, 20.51. 13C NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.37 (↑), 129.20 (↑), 128.40 (↑), 126.99 (↑), 126.86 (↑), 124.85 (↑), 123.63 (↑), 116.74 (↑), 37.71 (↑), 20.51 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.37 (↑), 129.20 (↑), 128.40 (↑), 126.99 (↑), 126.86 (↑), 124.85 (↑), 123.63 (↑), 116.74 (↑), 37.71 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.37 (↑), 129.20 (↑), 128.40 (↑), 126.99 (↑), 126.86 (↑), 124.85 (↑), 123.63 (↑), 116.74 (↑), 37.71 (↑), 20.51 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.57 (↓), 146.73 (↓), 142.78 (↓), 135.65 (↓), 130.78 (↓), 130.16 (↓), 129.37 (↓), 129.20 (↓), 128.40 (↑), 126.99 (↑), 126.86 (↑), 124.85 (↑), 123.63 (↑), 120.48 (↓), 116.74 (↑), 115.77 (↓), 58.03 (↓), 37.71 (↑), 20.51 (↑) Analysis for C21H16N2O, calculated C 80.75, H 5.16, N 8.97. Found: C 80.61, H 5.04, N 8.85.

2.2.6 3-Amino-1-(4-methoxyphenyl)-1H-benzo[f]chromene-2-carbonitrile (4f)

Yield 81%; mp 197–198°C (Lit. mp 192°C [18]), IR (FTIR/KBr) υ (cm−1): 3435, 3343, 3211 (NH2), 2189 (CN). 1H-NMR (DMSO-d6) δ: 3.67 (s, 3H, OCH3), 5.26 (s, 1H, H-1), 6.80 (bs, 2H, NH2), 7.93–6.81 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.55, 157.83, 146.68, 137.87, 130.81, 130.16, 129.32, 128.41, 128.01, 126.97, 124.83, 123.65, 120.55, 116.76, 115.93, 114.02, 58.25, 54.93, 37.31. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.32 (↑), 128.41 (↑), 128.01 (↑), 126.97 (↑), 124.83 (↑), 123.65 (↑), 116.76 (↑), 114.02 (↑), 54.93 (↑), 37.31 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.32 (↑), 128.41 (↑), 128.01 (↑), 126.97 (↑), 124.83 (↑), 123.65 (↑), 116.76 (↑), 37.31 (↑), 114.02 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.32 (↑), 128.41 (↑), 128.01 (↑), 126.97 (↑), 124.83 (↑), 123.65 (↑), 116.76 (↑), 54.93 (↑), 37.31 (↑), 114.02 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.55 (↓), 146.68 (↓), 130.81 (↓), 130.16 (↓), 129.32 (↑), 128.01 (↑), 126.97 (↑), 124.83 (↑), 123.65 (↑), 120.55 (↓), 116.76 (↑), 115.93 (↓), 58.25 (↓), 54.93 (↑), 37.31 (↑), 157.83 (↓), 137.87(↓), 128.41 (↑), 114.02 (↑). Analysis for C21H16N2O2, calculated C 76.81, H 4.91, N, 8.53. Found: C 76.68, H 4.77, N 8.39.

2.2.7 3-Amino-1-(2,4-dimethoxyphenyl)-1H-benzo[f]chromene-2-carbonitrile (4g)

Yield 80%; mp 205–206°C, IR (FTIR/KBr) υ (cm−1): 3466, 3326, 3186 (NH2), 2201 (CN). 1H-NMR (DMSO-d6) δ: 3.69 (s, 3H, OCH3), 3.88 (s, 3H, OCH3), 5.50 (s, 1H, H-1), 6.83 (bs, 2H, NH2), 7.91–6.36 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 160.01, 159.15, 156.60, 147.07, 130.64, 130.29, 129.17, 128.99, 128.42, 127.03, 124.74, 122.98, 120.46, 116.65, 116.04, 107.71, 105.80, 98.63, 57.31, 55.99, 55.06, 31.16. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.17 (↑), 128.99 (↑), 128.42 (↑), 127.03 (↑), 124.74 (↑), 122.98 (↑), 116.65 (↑), 105.80 (↑), 98.63 (↑), 55.99 (↑), 55.06 (↑), 31.16 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.17 (↑), 128.99 (↑), 128.42 (↑), 127.03 (↑), 124.74 (↑), 122.98 (↑), 116.65 (↑), 105.80 (↑), 98.63 (↑), 31.16 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.17 (↑), 128.99 (↑), 128.42 (↑), 127.03 (↑), 124.74 (↑), 122.98 (↑), 116.65 (↑), 105.80 (↑), 98.63 (↑), 55.99 (↑), 55.06 (↑), 31.16 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 160.01 (↓), 159.15 (↓), 156.60 (↓), 147.07 (↓), 130.64 (↓), 130.29 (↓), 129.17 (↑), 128.99 (↑), 128.42 (↑), 127.03 (↑), 124.74 (↑), 122.98 (↑), 120.46 (↓), 116.65 (↑), 116.04 (↓), 107.71 (↓), 105.80 (↑), 98.63 (↑), 57.31 (↓), 55.99 (↑), 55.06 (↑), 31.16 (↑). MS (EI, 70 eV) m/z (relative intensity): 358 (M+, 10.33), 221 (100). Analysis for C22H18N2O3, calculated C 73.73, H 5.06, N 7.82. Found: C 73.84, H 5.18, N 7.92.

2.2.8 3-Amino-1-(3,4-dimethoxyphenyl)-1H-benzo[f]chromene-2-carbonitrile (4h)

Yield 79%, mp 195–196°C, IR (FTIR/KBr) υ (cm−1): 3432, 3339, 3195 (NH2), 2186 (CN). 1H-NMR (DMSO-d6) δ: 3.67 (s, 3H, OCH3), 3.68 (s, 3H, OCH3), 5.25 (s, 1H, H-1), 6.57 (bs, 2H, NH2), 7.94–6.58 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.57, 148.63, 147.46, 146.70, 138.35, 130.78, 130.27, 129.35, 128.39, 126.98, 124.85, 123.74, 120.55, 118.97, 116.73, 115.81, 112.15, 111.11, 58.14, 55.48, 55.43, 37.61. 13C NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.35 (↑), 128.39 (↑), 126.98 (↑), 124.85 (↑), 123.74 (↑), 118.97 (↑), 116.73 (↑), 112.15 (↑), 111.11 (↑), 55.48 (↑), 55.43 (↑), 37.61 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.35 (↑), 128.39 (↑), 126.98 (↑), 124.85 (↑), 123.74 (↑), 118.97 (↑), 116.73 (↑), 112.15 (↑), 111.11 (↑), 37.61 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.35 (↑), 128.39 (↑), 126.98 (↑), 124.85 (↑), 123.74 (↑), 118.97 (↑), 116.73 (↑), 112.15 (↑), 111.11 (↑), 55.48 (↑), 55.43 (↑), 37.61 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.57 (↓), 148.63 (↓), 147.46 (↓), 146.70 (↓), 138.35 (↓), 130.78 (↓), 130.27 (↓), 129.35 (↑), 128.39 (↑), 126.98 (↑), 124.85 (↑), 123.74 (↑), 120.55 (↓), 118.97 (↑), 116.73 (↑), 115.81 ( ↓), 112.15 (↑), 111.11 (↑), 58.14 (↓), 55.48 (↑), 55.43 (↑), 37.61 (↑). MS (EI, 70 eV) m/z (relative intensity): 358 (M+, 9.14), 221 (M+, 100). Analysis for C22H18N2O3, calculated C 73.73, H 5.06, N 7.82. Found: C 73.66, H 4.94, N 7.68.

2.2.9 3-Amino-1-phenyl-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6a)

Yield 89%; mp 255–256°C, IR (FTIR/KBr) υ (cm−1): 3441, 3328, 3196 (NH2), 2182 (CN); 1H-NMR (DMSO-d6) δ: 3.74 (s, 3H, OCH3), 5.29 (s, 1H, H-1), 6.98 (bs, 2H, NH2), 7.85–7.04 (m, 10H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.61, 157.91, 147.24, 145.71, 131.66, 129.96, 129.03, 128.59, 127.20, 126.54, 125.96, 120.51, 116.78, 114.88, 114.02, 103.22, 57.89, 55.05, 38.11. MS (EI, 70 eV) m/z (relative intensity): 328 (M+, 7.11), 251 (100). Analysis for C21H16N2O2, calculated C 76.81, H 4.91, N 8.53. Found: C 76.70, H, 4.81, N 8.42%.

2.2.10 3-Amino-1-(4-flourophenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6b)

Yield 89%, mp 265–266°C, IR (FTIR/KBr) υ (cm−1): 3466, 3359, 3210 (NH2), 2183 (CN). 1H- NMR (DMSO-d6) δ: 3.75 (s, 3H, OCH3), 5.35 (s, 1H, H-1), 6.98 (bs, 2H, NH2), 7.86–7.05 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 161.72, 159.79, 158.00, 147.21, 141.94, 131.58, 130.00, 129.04, 128.97, 125.99, 120.42, 116.84, 115.43, 115.25, 114.04, 103.16, 57.77, 55.11, 37.23. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 130.00 (↑), 129.04 (↑), 128.97 (↑), 116.84 (↑), 115.43 (↑), 114.04 (↑), 103.16 (↑), 55.11 (↑), 37.23 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 130.00 (↑), 129.04 (↑), 128.97 (↑), 116.84 (↑), 115.43 (↑), 114.04 (↑), 103.16 (↑), 37.23 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) revealed δ: 130.00 (↑), 129.04 (↑), 128.97 (↑), 116.84 (↑), 115.43 (↑), 114.04 (↑), 103.16 (↑), 55.11 (↑), 37.23 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 161.72 (↓), 159.79 (↓), 158.00 (↓), 147.21 (↓), 141.94 (↓), 131.58 (↓), 130.00 (↑), 129.04 (↑), 128.97 (↑), 125.99 (↓), 120.42 (↓), 116.84 (↑), 115.25 (↓), 115.43 (↑), 114.04 (↑), 103.16 (↑), 57.77 (↓), 55.11 (↑), 37.23 (↑). MS (EI, 70 eV) m/z (relative intensity): 346 (M+, 31.63), 208 (100). Analysis for C21H15FN2O2, calculated C 72.82, H 4.37, N 8.09. Found: C 72.90; H 4.48; N 8.20.

2.2.11 3-Amino-1-(4-chlorophenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6c)

Yield 89%, mp 257–258°C, IR (FTRI/KBr) υ (cm−1): 3467, 3356, 3202 (NH2), 2180 (CN), 1H- NMR (DMSO-d6) δ: 3.75 (s, 3H, OCH3), 5.36 (s, 1H, H-1), 7.04 (bs, 2H, NH2), 7.86–7.06 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.68, 158.04, 147.24, 144.70, 131.56, 131.11, 130.03, 129.23, 128.29, 128.60, 125.97, 120.37, 116.86, 114.40, 114.03, 103.11, 57.43, 55.13, 37.32. MS (EI, 70 eV) m/z (relative intensity): 364 (M++2, 11.37), 362 (M+, 33.61), 208 (100). Analysis for C21H15ClN2O2, calculated C 69.52, H 4.17, N 7.72. Found: C 69.40, H 4.04, N 7.61.

2.2.12 3-Amino-1-(4-bromophenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6d)

Yield 87%, mp 251–252°C, IR (FTIR/KBr) υ (cm−1): 3468, 3354, 3200 (NH2), 2177 (CN). 1H NMR (DMSO-d6) δ: 3.75 (s, 3H, OCH3), 5.35 (s, 1H, H-1), 7.05 (bs, 2H, NH2), 7.86–7.06 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.68, 158.04, 147.24, 145.12, 131.55, 131.52, 130.03, 129.39, 128.28, 125.97, 120.37, 119.61, 116.85, 114.34, 114.03, 103.11, 57.36, 55.14, 37.39. MS (EI, 70 eV) m/z (relative intensity): 408 (M++2, 6.16), 406 (M+, 6.61), 252 (100). Analysis for C21H15BrN2O2, calculated C 61.93, H 3.71, N, 6.88. Found: C 61.84, H 3.49, N 6.73.

2.2.13 3-Amino-9-methoxy-1-(4-methylphenyl)-1H-benzo[f]chromene-2-carbonitrile (6e)

Yield 88%, mp 244–245°C, IR (FTIR/KBr) υ (cm−1): 3432, 3333, 3209 (NH2), 2185 (CN); 1H-NMR (DMSO-d6) δ: 2.21 (s, 3H, CH3), 3.75 (s, 3H, OCH3), 5.23 (s, 1H, H-1), 6.95 (bs, 2H, NH2), 7.84–7.04 (m, 9H, Ar-H), 13C-NMR (DMSO-d6) δ: 159.54, 157.87, 147.16, 142.80, 135.62, 131.67, 129.93, 129.12, 128.93, 127.09, 125.95, 120.54, 116.70, 115.02, 114.02, 103.26, 58.05, 55.06, 37.75, 20.53. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.93 (↑), 129.12 (↑), 128.93 (↑), 127.09 (↑), 116.70 (↑), 114.02 (↑), 103.26 (↑), 55.06 (↑), 37.75 (↑), 20.53 (↑). In the DEPT spectrum at 90°, only CH signals are (↑) δ: 129.93 (↑), 129.12 (↑), 128.93 (↑), 127.09 (↑), 116.70 (↑), 114.02 (↑), 103.26 (↑), 37.75 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.93 (↑), 129.12 (↑), 128.93 (↑), 127.09 (↑), 116.70 (↑), 114.02 (↑), 103.26 (↑), 55.06 (↑), 37.75 (↑), 20.53 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.54 (↓), 157.87 (↓), 147.16 (↓), 142.80 (↓), 135.62 (↓), 131.67 (↓), 129.93 (↑), 129.12 (↑), 128.93 (↑), 127.09 (↑), 125.95 (↓), 120.54 (↓), 116.70 (↑), 115.02 (↓), 114.02 (↑), 103.26 (↑), 58.05 (↓), 55.06 (↑), 37.75 (↑), 20.53 (↑). MS (EI, 70 eV) m/z (relative intensity): 342 (M+, 23.91), 90 (100); Analysis for C22H18N2O2, calculated C 77.17, H 5.30, N 8.18. Found: C 77.04, H 5.29, N 8.09.

2.2.14 3-Amino-1-(4-methoxyphenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6f)

Yield 85%, mp 219–220°C (Lit. mp 219°C [19]), IR (FTIR/KBr) υ (cm−1): 3441, 3328, 3212 (NH2), 2182 (CN). 1H-NMR (DMSO-d6) δ: 3.68 (s, 3H, OCH3), 3.76 (s, 3H, OCH3), 5.23 (s, 1H, H-1), 6.82 (bs, 2H, NH2), 7.83–6.84 (m, 9H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.48, 157.88, 157.80, 147.11, 137.87, 131.67, 129.93, 128.89, 128.23, 125.96, 120.60, 116.71, 115.16, 114.02, 113.94, 103.26, 58.21, 55.08, 54.94, 37.31. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.93 (↑), 128.89 (↑), 128.23 (↑), 116.71 (↑), 114.02 (↑), 113.94 (↑), 103.26 (↑), 55.08 (↑), 54.94 (↑), 37.31 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.93 (↑), 128.89 (↑), 128.23 (↑), 116.71 (↑), 114.02 (↑), 113.94 (↑), 103.26 (↑), 37.31 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.93 (↑), 128.89 (↑), 128.23 (↑), 116.71 (↑), 114.02 (↑), 113.94 (↑), 103.26 (↑), 55.08 (↑), 54.94 (↑), 37.31 (↑). 13CNMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.48 (↓), 157.88 (↓), 157.80 (↓), 147.11 (↓), 137.87 (↓), 131.67 (↓), 129.93 (↑), 128.89 (↑), 128.23 (↑), 125.96 (↓), 120.60 (↓), 116.71 (↑), 115.16 (↓), 114.02 (↑), 113.94 (↑), 103.26 (↑), 58.21 (↓), 55.08 (↑), 54.94 (↑), 37.31 (↑). Analysis for C22H18N2O3, calculated C 73.73, H 5.06, N 7.82. Found: C 73.85, H 5.17, N 7.93.

2.2.15 3-Amino-1-(2,4-dimethoxyphenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6g)

Yield 85%; mp 225–226°C, IR (FTIR/KBr) υ (cm−1): 3479, 3328, 3217 (NH2), 2199 (CN); 1H-NMR (DMSO-d6) δ: 3.69 (s, 3H, OCH3), 3.75 (s, 3H, OCH3), 3.88 (s, 3H, OCH3), 5.44 (s, 1H, H-1), 6.85 (bs, 2H, NH2), 7.80–6.39 (m, 8H, Ar-H). 13C-NMR (DMSO-d6) δ: 159.89, 159.13, 157.98, 156.48, 147.45, 131.80, 129.94, 129.55, 128.54, 125.95, 125.81, 120.50, 116.80, 115.44, 113.98, 105.87, 102.28, 98.18, 57.43, 55.94, 55.06, 54.88, 30.92. 13C-NMR-DEPT spectrum at 135о CH, CH3 (↑), CH2 (↓) δ: 129.94 (↑), 129.55 (↑), 128.54 (↑), 116.80 (↑), 113.98 (↑), 105.87 (↑), 102.28 (↑), 98.18 (↑), 55.94 (↑), 55.06 (↑), 54.88 (↑), 30.92 (↑). In the DEPT spectrum at 90° only CH signals are (↑) δ: 129.94 (↑), 129.55 (↑), 128.54 (↑), 116.80 (↑), 113.98 (↑), 105.87 (↑), 102.28 (↑), 98.18 (↑), 30.92 (↑). In the DEPT spectrum at 45° CH, CH2, and CH3 (↑) δ: 129.94 (↑), 129.55 (↑), 128.54 (↑), 116.80 (↑), 113.98 (↑), 105.87 (↑), 102.28 (↑), 98.18 (↑), 55.94 (↑), 55.06 (↑), 54.88 (↑), 30.92 (↑). 13C-NMR-APT spectrum CH, CH3 (↑), CH2, Cq (↓) δ: 159.89 (↓), 159.13 (↓), 157.98 (↓), 156.48 (↓), 147.45 (↓), 131.80 (↓), 129.94 (↑), 129.55 (↑), 128.54 (↑), 125.95 (↓), 120.50 (↓), 116.80 (↑), 115.44 (↓), 113.98 (↑), 105.87 (↑), 102.28 (↑), 98.18 (↑), 57.43 (↓), 55.94 (↑), 55.06 (↑), 54.88 (↑), 30.92 (↑), 125.81 (↓). MS (EI, 70 eV) m/z (relative intensity): 388 (M+, 21.94), 63 (100); Analysis for C23H20N2O4, calculated C 71.12, H 5.19, N 7.21. Found: C 71.01, H 5.08, N 7.11.

2.2.16 3-amino-1-(3,4-dimethoxyphenyl)-9-methoxy-1H-benzo[f]chromene-2-carbonitrile (6h)

Yield 81%, mp 258–259°C, IR (FTIR/KBr) υ (cm−1): 3392, 3325, 3210 (NH2), 2188 (CN); 1H-NMR (DMSO-d6) δ: 3.67 (s, 3H, OCH3), 3.68 (s, 3H, OCH3), 3.77 (s, 3H, OCH3), 5.24 (s, 1H, H-1), 6.94 (bs, 2H, NH2), 7.84–6.65 (m, 8H, Ar-H).13C-NMR (DMSO-d6) δ: 159.52, 157.90, 148.50, 147.38, 147.12, 138.48, 131.78, 129.91, 128.89, 125.95, 120.62, 119.30, 116.82, 115.15, 114.01, 112.17, 111.35, 103.25, 58.27, 55.49, 55.41, 55.09, 37.64. MS (EI, 70 eV) m/z (relative intensity): 388 (M+, 19.28), 51 (100); Analysis for C23H20N2O4, calculated C 71.12, H 5.19, N 7.21. Found: C 71.26, H 5.31, N 7.32.

2.3 Biological tests

2.3.1 Cell culture

The tumor cell lines breast adenocarcinoma (MCF-7), human colon carcinoma (HCT-116), and hepatocellular carcinoma (HepG-2) were derived from the American Type Culture Collection (ATCC, Rockville, MD). The cells were grown on RPMI-1640 medium supplemented with 10% inactivated fetal calf serum and 50 µg/mL gentamycin. The cells were maintained at 37°C in a humidified atmosphere with 5% CO2 and were subcultured two to three times a week.

2.3.2 Cytotoxicity evaluation using viability assay

The tumor cell lines were suspended in medium at a concentration of 5×104 cell/well in Corning® 96-well tissue culture plates and were then incubated for 24 h. The tested compounds with concentrations ranging from 0 to 50 μg/mL were then added into 96-well plates (six replicates) to achieve different concentration for each compound. Six vehicle controls with media or 0.5% DMSO were run for each 96-well plate as a control. After being incubated for 24 h, the numbers of viable cells were determined by the MTT test. Briefly, the media was removed from the 96-well plates and replaced with 100 μL of fresh culture RPMI 1640 medium without phenol red then 10 µL of the 12 mM MTT stock solution (5 mg of MTT in 1 mL of PBS) to each well including the untreated controls. The 96-well plates were then incubated at 37°C and 5% CO2 for 4 h. An 85-μL aliquot of the media was removed from the wells, and 50 µL of DMSO was added to each well and mixed thoroughly with the pipette and incubated at 37°C for 10 min. Afterward, the optical density was measured at 590 nm with the microplate reader (SunRise, TECAN, Inc, USA) to determine the number of viable cells and the percentage of viability was calculated as [1–(ODt/ODc)]×100% where ODt is the mean optical density of wells treated with the tested sample and ODc is the mean optical density of untreated cells. The relation between surviving cells and drug concentration was plotted to get the survival curve of each tumor cell line after being treated by the specified compound. The 50% inhibitory concentration (IC50); the concentration required to cause toxic effects in 50% of intact cells, was estimated from graphic plots of the dose response curve for each concentration using Graph pad Prism software (San Diego, CA. USA) [20].

2.4 Statistical analysis

All statistical calculations were conducted using Microsoft excel version 10, and SPSS (statistical package for the social science version 20.00) statistical program at 0.05, 0.01, and 0.001 level of significance [21] and are cited in Tables 27 (see supplementary information). Comparisons of inhibiting tumor growth between different treatment compounds or control were done using Student’s t-test, One-way ANOVA and post hoc-LSD tests (the least significant difference) measurement.

3 Results and discussion

Based on the reported cytotoxic activity of a great number of bioactive compounds incorporating chromene or benzochromene moieties, 3-amino-1-aryl-1H-benzo[f]chromene-2-carbonitriles (4a–h) and 3-amino-1-aryl-9-methoxy-1H-benzo[f]chromene-2-carbonitriles(6a–h) were selected to carry out a preliminary screening for its cytotoxic effect against three metastatic human cancer cell lines, including MCF-7 breast cancer, HCT-116 human colon cancer, and HepG-2 liver cancer. The selection of such cell lines was inspired by the declared anticancer activity of a number of chromenes, benzochromenes, and fused chromenes [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], [11], [12], [22], [23], [24], [25], [26], [27], [28], [29]. The cytotoxic activity was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) colorimetric assay [20], [30]. In-vitro cytotoxicity evaluation was performed at the Regional Centre for Mycology & Biotechnology (RCMP), Al-Azhar University under different concentrations (50, 25, 12.5, 6.25, 3.125, 1.56, and 0 µg/mL), Vinblastine and Colchicine are used as reference cytotoxic compounds. The results were expressed as growth inhibitory concentration (IC50) values that represent the compound concentrations required to produce a 50% inhibition of cell growth after 24 h of incubation compared to untreated controls as shown in Table 1 and Figure 2 (see supplementary information).

Table 1:

Cytotoxic activity of the compounds 4a–h and 6a–h against three cancer cell lines.

CompoundArIC50 (µg/mL)a
MCF-7HCT-116HepG-2
4aC6H546.1±0.1140.2±0.636.1±0.5
4b4-FC6H40.61±0.032.5±0.123.6±0.12
4c4-ClC6H40.7±0.20.6±0.112.1±0.12
4d4-BrC6H40.8±0.042.2±0.30.6±0.2
4e4-MeC6H42.6±0.125.2±±0.1532.1±0.14
4f4-MeOC6H40.71±0.33.4±0. 043.6±0.05
4g2,4-MeOC6H33.5±0.110.7±0.131.0±0.17
4h3,4-MeOC6H324.9±0.0111.7±0.0622.9±0.02
6aC6H532.9±0.242.1±0.1316.9±0.11
6b4-FC6H45.7±0.052.2±0.013.9±0.01
6c4-ClC6H40.18±0.240.7±0.072.6±0.08
6d4-BrC6H42.3±0.30.9±0.080.8±0.03
6e4-MeC6H42.4±0.111.0±0.013.0±0.21
6f4-MeOC6H417.1±0.13.0±0.1429.7±0.12
6g2,4-MeOC6H342.3±0.219.4±0.536.2±0.2
6h3,4-MeOC6H31.0±0.52.4±0.320.3±0.4
Vinblastine6.1±0.032.6±0.084.6±0.01
Colchicine17.7±0.0142.8±0.0210.6±0.04

aIC50 values expressed in µg/mL as the mean values of triplicate wells from at least three experiments and are reported as the mean ± standard error.

Figure 2: IC50 values expressed in (µg/mL) of 3-amino-1H-benzo[f]chromene derivatives (4a–h) and (6a–h) against MCF-7, HCT, and HepG-2 tumor cells.
Figure 2:

IC50 values expressed in (µg/mL) of 3-amino-1H-benzo[f]chromene derivatives (4a–h) and (6a–h) against MCF-7, HCT, and HepG-2 tumor cells.

Based on the results, it is evident that some of the tested compounds displayed significant growth inhibitory activity. Compounds 4b,c,f,d,e,g and 6c,h,d,e,b (IC50=0.18–5.7 µg/mL) were found to be more potent and efficacious than Vinblastine and Colchicine (IC50=6.1 and 17.7 µg/mL) against MCF-7 cancer cell, while compounds 4c,g,d,b and 6c,d,e,b,h (IC50=0.6–2.5 µg/mL) were found to be more potent and efficacious than Vinblastine and Colchicine (IC50=2.6 and 42.8 µg/mL) against HCT-116 cancer cell. Moreover, compound 4d,g,c,b,f with (IC50=0.6–3.6 µg/mL) were 7.7, 4.6, 2.2, 1.3, 1.3 and 17.7, 10.6, 5.1, 3.0, 3.0 times more active than Vinblastine and Colchicine (IC50=4.6 and 10.6 µg/mL), respectively, against HepG-2 cancer cell, while compounds 6d,c,e,b with (IC50=0.8-3.9 µg/mL) were 5.8, 1.8, 1.5, 1.2, and 13.3, 4.1, 3.5, 2.7, times more efficacious than Vinblastine and Colchicine (IC50=4.6 and 10.6 µg/mL), respectively, against HepG-2 cancer cell. In addition, compound 6f (IC50=17.1 µg/mL) exhibited a good activity against MCF-7 cancer cell as compared to Colchicine (IC50=17.7 µg/mL) and compounds 4f,e,h,a and 6f,g,a (IC50=3.4–42.1 µg/mL) were more active than Colchicine (IC50=42.8 µg/mL) against HCT-116 cancer cell, while the other compounds showed equipotent or moderate to fair cytotoxic activities against the three tumor cell types comparable to Vinblastine and Colchicine. It was also observed that MCF-7 cell line was more susceptible to the influence of most of the tested compounds than HCT-116 and HepG-2 cell lines, while HCT-116 cell line was more susceptible to the influence of most of the tested compounds than HepG-2 cell line.

The preliminary SAR study has focused on the effect of introduction of different substituents at the phenyl ring at the 1-position of the 1H-benzo[f]chromene moiety, on the antitumor activities of the target compounds. Based on the aforementioned biological data, many structure activity relationships could be deduced. With respect to the type of the pendant substituent at the phenyl ring and with H atom or methoxy group at 9-position, it was found that the unsubstituted phenyl at 1-postion with H atom or methoxy group at 9-position is not preferred for antitumor activity, compounds 4a and 6a exhibited equipotent or moderate to fair cytotoxic activities against the three tumor cell types comparable to Vinblastine and Colchicine. The introduction of halogen atoms (electron-withdrawing group) on the phenyl ring at 1-postion of compounds 4b–d (IC50=0.61, 0.7, 0.8 µg/mL, respectively) and compounds 6c,d,b (IC50=0.18, 2.3, 5.7 µg/mL, respectively) has caused a remarkable enhancement in the antitumor activity against MCF-7 with higher significance as shown in Tables 2 and 3 than the methoxy, methyl, or dimethoxy (electron-donating group) of compounds 4f,e,g,h (IC50=0.71, 2.6, 3.5, 24.9 µg/mL, respectively) or dimethoxy, methyl, methoxy, or dimethoxy of compounds 6h,e,f,g (IC50=1.0, 2.4, 17.1, 42.3 µg/mL, respectively) as compared with Vinblastine and Colchicine (IC50=6.1 and 17.7 µg/mL), respectively. This results imply that grafting a lipophilic electron-withdrawing substituent like halogens is more beneficial than an electron-donating substituent like methyl or methoxy group for the activity with H atom or methoxy group at 9-position.

Table 2:

Positive and negative controls and effectiveness of test compounds against MCF-7.

Control/Cpd.IC50 (μg/mL)Cell lineF ratiop-Value
Vinblastine6.1±0.15MCF-7233.0270.000 (HS)
4b0.61±0.03MCF-7
4c0.7±0.03MCF-7
4f0.71±0.06MCF-7
4d0.8±0.08MCF-7
4e2.6±0.15MCF-7
4g3.5±0.10MCF-7
6c0.18±0.06MCF-7
6h1.0±0.14MCF-7
6d2.3±0.15MCF-7
6e2.4±0.02MCF-7
6b5.7±0.33MCF-7

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

Table 3:

Positive and negative controls and effectiveness of test compounds against MCF-7.

Control/Cpd.IC50 (μg/mL)Cell lineF ratiop-Value
Colchicine17.7±0.11MCF-72468.4040.000 (HS)
4b0.61±0.03MCF-7
4c0.7±0.06MCF-7
4f0.71±0.05MCF-7
4d0.8±0.01MCF-7
4e2.6±0.16MCF-7
4g3.5±0.07MCF-7
6c0.18±0.07MCF-7
6h1.0±0.12MCF-7
6d2.3±0.05MCF-7
6e2.4±0.02MCF-7
6b5.7±0.15MCF-7
6f17.1±0.94MCF-7

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

Investigations of the cytotoxic activity against HCT-116 indicated that compounds 4c,g,d,b (IC50=0.6, 0.7, 2.2, 2.5 µg/mL, respectively) and 6c,d,e,b,h (IC50=0.7, 0.9, 1.0, 2.2, 2.4 µg/mL, respectively) were found to be the most potent derivative against HCT-116 as it was 4.3, 3.7, 1.2, 1.0; 71.3, 61.2, 19.5, 17.1, and 3.7, 2.9, 2.6, 1.2, 1.1; 61.2, 47.6, 42.8, 19.5, 17.8, respectively, times more potent and efficacious than Vinblastine and Colchicine (IC50=2.6 and 42.8 µg/mL), with the highest significance as shown in Tables 4 and 5 . This data suggests that the substitution in the phenyl ring at 1-postion with H atom or methoxy group at 9-position may be tolerated and also the halogens incorporation may be advantageous.

Table 4:

Positive and negative controls and effectiveness of test compounds against HCT-116.

Control/Cpd.IC50 (μmol/L)Cell lineF ratiop-Value
Vinblastine2.6±0.12HCT-11664.2100.000 (HS)
4c0.6±0.06HCT-116
4g0.7±0.06HCT-116
4d2.2±0.14HCT-116
4b2.5±0.17HCT-116
6c0.7±0.06HCT-116
6d0.9±0.08HCT-116
6e1.0±0.10HCT-116
6b2.2±0.08HCT-116
6h2.4±0.15HCT-116

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

Table 5:

Positive and negative controls and effectiveness of test compounds against HCT-116

Control/Cpd.IC50 (μmol/L)Cell lineF ratiop-Value
Colchicine42.8±0.06HCT-1166788.3430.000 (HS)
4c0.6±0.07HCT-116
4g0.7±0.08HCT-116
4d2.2±0.11HCT-116
4b2.5±0.17HCT-116
4f3.4±0.15HCT-116
4e5.2±0.011HCT-116
4h11.7±0.09HCT-116
4a40.2±0.54HCT-116
6c0.7±0.057HCT-116
6d0.9±0.08HCT-116
6e1.0±0.02HCT-116
6b2.2±0.02HCT-116
6h2.4±0.23HCT-116
6g19.4±0.24HCT-116
6a42.1±0.54HCT-116

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

On the other hand, cytotoxicity evaluation in HepG-2 cell line revealed that compounds 4d,g,c,b,f with (IC50=0.6, 1.0, 2.1, 3.6, 3.6 µg/mL, respectively) and compounds 6d,c,e,b with (IC50=0.8, 2.6, 3.0, 3.9 µg/mL, respectively) have enhanced the activity against HepG-2 cell line as compared to Vinblastine and Colchicine (IC50=4.6 and 10.6 µg/mL), respectively, as shown in Tables 6 and 7. In contrast, the other compounds 4h,f (IC50=22.9 and 32,1 µg/mL) and 6h,f,g (IC50=20.3, 29.7, and 36.2 µg/mL, respectively) exhibited moderate to fair cytotoxic activities against HepG-2 cell line and indicated that a lipophilic electron-withdrawing substituent like halogens is more beneficial than an electron-donating substituent like methyl or methoxy for the activity with H atom at 9-position.

Table 6:

Positive and negative controls and effectiveness of test compounds against HepG-2.

Control/Cpd.IC50 (μg/mL)Cell lineF ratiop-Value
Vinblastine4.6±0.24HepG-2112.5770.000 (HS)
4d0.6±0.05HepG-2
4g1.0±0.08HepG-2
4c2.1±0.15HepG-2
4b3.6±0.02HepG-2
4f3.6±0.08HepG-2
6d0.8±0.05HepG-2
6c2.6±0.02HepG-2
6e3.0±0.10HepG-2
6b3.9±0.08HepG-2

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

Table 7:

Positive and negative controls and effectiveness of test compounds against HepG-2.

Control/Cpd.IC50 (μg/mL)Cell lineF ratiop-Value
Colchicine10.6±0.18HepG-2605.7490.000 (HS)
4d0.6±0.08HepG-2
4g1.0±0.06HepG-2
4c2.1±0.15HepG-2
4b3.6±0.12HepG-2
4f3.6±0.05HepG-2
6d0.8±0.06HepG-2
6c2.6±0.01HepG-2
6e3.0±0.08HepG-2
6b3.9±0.45HepG-2

Positive control (active compounds) and negative control (standard drugs).

All statistical calculations were done as the mean values of triplicate.

HS=Higest significantly at p-Value <0.05.

Finally, it can be deduced that the substitution pattern on the phenyl group at the 1-position of 1H-benzo[f]chromene moiety is a crucial element for the antitumor activity with H atom or methoxy group at 9-position. The incorporation of halogen atoms (electron-withdrawing group) has greatly enhanced the activity than those of the electron-donating groups like methyl or methoxy groups and that the H atom is more preferred than the methoxy group at 9-position.

4 Conclusions

To summarize, 16 of 3-amino-1H-benzo[f]chromene-2-carbonitrile derivatives were synthesized and their cytotoxic activities were evaluated against three cancer cell lines (MCF-7, HCT-116, and HepG-2). Most of the synthesized compounds displayed relatively potent and selective cytotoxic activity against the three cancer cell lines. In particular, compounds 4b,c,f,d,e,g and 6c,h,d,e,b (IC50=0.18–5.7 µg/mL) displayed the highest activity against MCF-7 cancer cell as compared to Vinblastine and Colchicine (IC50=6.1 and 17.7 µg/mL), while compounds 4c,g,d,b and 6c,d,e,b,h (IC50=0.6–2.5 µg/mL) were found to be more potent and efficacious against HCT-116 cancer cell than Vinblastine and Colchicine (IC50=2.6 and 42.8 µg/mL). Furthermore, compounds 4d,g,c,b,f and 6d,c,e,b with (IC50=0.6–3.9 µg/mL) have higher activity than the standard drugs Vinblastine and Colchicine (IC50=4.6 and 10.6 µg/mL). From the structure–activity relationships (SARs), it can be deduced that the introduction of halogens substituent (electron-withdrawing group) at the 4-position of phenyl ring at the 1-position of the 1H-benzo[f]chromene moiety enhanced the antitumor activities of the target compounds more than the electron-donating substituent like methyl or methoxy groups, and the H atom is more favorable than methoxy group at 9-position.

Acknowledgments

The authors profoundly thank the Regional Center for Mycology & Biotechnology (RCMP), Al-Azhar University for carrying out the antitumor study and Elemental analyses.

References

1. Patil SA, Patil R, Pfeffer LM, Miller DD. Chromenes: potential new chemotherapeutic agents for cancer. Future Med Chem 2013;5:1647–60.10.4155/fmc.13.126Search in Google Scholar

2. Birch KA, Heath WF, Hermeling RN, Johnston CM, Stramm L, Dell C, et al. LY290181, an inhibitor of diabetes-induced vascular dysfunction, blocks protein kinase C-stimulated transcriptional activation through inhibition of transcription factor binding to a phorbol response element. Diabetes 1996;45:642–50.10.2337/diab.45.5.642Search in Google Scholar

3. Wiener C, Schroeder CH, West BD, Link KP. Studies on the 4-Hydroxycoumarins. XVIII. 3-[α-(Acetamidomethyl)benzyl]-4-Hydroxycoumarin and related products. J Org Chem 1962;27:3086–8.10.1021/jo01056a024Search in Google Scholar

4. Smith CW, Bailey JM, Billingham ME, Chandrasekhar S, Dell CP, Harvey AK, et al. The anti-rheumatic potential of a series of 2,4-di-substituted-4H-naphtho[1,2-b]pyran-3-carbonitriles. Bioorg Med Chem Lett 1995;5:2783–8.10.1016/0960-894X(95)00487-ESearch in Google Scholar

5. Kemnitzer W, Kasibhatla S, Jiang S, Zhang H, Zhao J, Jia S, et al. Discovery of 4-aryl-4h-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 2. structure–activity relationships of the 7- and 5-, 6-, 8-positions. Bioorg Med Chem Lett 2005;15:4745–51.10.1016/j.bmcl.2005.07.066Search in Google Scholar PubMed

6. Wang J-L, Liu D, Zhang Z-J, Shan S, Han X, Srinivasula SM, et al. Structure-based discovery of an organic compound that binds bcl-2 protein and induces apoptosis of tumor cells. Proc Natl Acad Sci U S A 2000;97:7124–29.10.1073/pnas.97.13.7124Search in Google Scholar PubMed PubMed Central

7. Doshi JM, Tian D, Xing C. Structure-activity relationship studies of ethyl 2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4h-chromene-3-carboxylate (ha 14-1), an antagonist for anti-apoptotic bcl-2 proteins to overcome drug resistance in cancer, J Med Chem 2006;49;7731–39.10.1021/jm060968rSearch in Google Scholar PubMed

8. Sabry NM, Mohamed HM, Khattab ES, Motlaq SS, El-Agrody AM. Synthesis of 4H-chromene, coumarin, 12H-chromeno[2,3-d]pyrimidine derivatives and some of their antimicrobial and cytotoxicity activities. Eur J Med Chem 2011;46:765–72.10.1016/j.ejmech.2010.12.015Search in Google Scholar PubMed

9. El-Agrody AM, Khattab ES, Fouda AM. Synthesis, structure–activity relationship (sar) studies on some 4-aryl-4H-chromenes and relationship between lipophilicity and antitumor activity. Lett Drug Des Discov 2014;11:1167–76.10.2174/1570180811666140623204655Search in Google Scholar

10. El-Agrody AM, Fouda AM, Khattab ES. Synthesis, antitumor activity of 2-amino-4H-benzo[h]chromene derivatives and structure-activity relationships of the 3- and 4-positions. Med Chem Res 2013;22:6105–20.10.1007/s00044-013-0602-8Search in Google Scholar

11. El-Agrody AM, Fouda AM, Al-Dies AM. Studies on the synthesis, In-Vitro antitumor activity of 4H-benzochromene, 7H-benzo[h]chromeno[2,3-d]pyrimidine derivatives and structure–activity relationships of the 2-,3- and 2,3-positions. Med Chem Res 2014;23:3187–99.10.1007/s00044-013-0904-xSearch in Google Scholar

12. Halawa AH, Fouda AM, Al-Dies AM, El-Agrody AM. Synthesis, biological evaluation and molecular docking studies of 4H-benzo[h]chromenes, 7H-benzo[h]chromeno[2,3-d]pyrimidines as antitumor agents. Lett Drug Des Discov 2016;13:77–88.10.2174/1570180812666150611185830Search in Google Scholar

13. Kidwai M, Saxena S, Khan MK, Thukralb SS. Aqua mediated synthesis of substituted 2-amino-4H-chromenes and in vitro study as antibacterial agents. Bioorg Med Chem Lett 2005;15:4295–8.10.1016/j.bmcl.2005.06.041Search in Google Scholar

14. Shi DQ, Zhang Z, Tu SJ. Reaction of substituted cinnamonitriles with β-naphthol in water. Chin J Org Chem 2003;23:809–12.Search in Google Scholar

15. Wen X-M, Wang H-Y, Li S-L. Aqueous [bmim][BF4] as green solvent in microwave-assisted organic reactions: clean synthesis of 1H-benzo[f]chromene derivatives. J Chem Res 2006;12:776–8.10.3184/030823406780199767Search in Google Scholar

16. Maggi R, Ballini R, Sartori G. Basic alumina catalysed synthesis of substituted 2-amino-2-chromenes via three-component reaction. Tetrahedron Lett 2004;45:2297–9.10.1016/j.tetlet.2004.01.115Search in Google Scholar

17. Zhuang QY, Rong LC, Shi DQ. Synthesis and crystal structure of substituted naphtho-pyran. Chin J Org Chem 2003;23:671–3.Search in Google Scholar

18. Elagamey AG, Sawllim SZ, El-Taweel FM, Elnagdi MH. Nitriles in heterocyclic synthesis: novel syntheses of benzo[b]pyrans, naphtho[1,2-b]pyrans, naphtho[2,1-b]pyrans, pyrano[3,2-h]quinolines and pyrano[3,2-c]quinolines. Collect Czech Chem Commun 1988;53:1534–8.10.1135/cccc19881534Search in Google Scholar

19. El-Agrody AM, Eid FA, Emam HA, Mohamed HM, Bedair AH. Synthesis of 9-methoxy and 9-acetoxy-3-amino-1-(4-methoxyphenyl)-1H-benzo[f]chromene-2-carbonitriles via 2-(imino-piperidin-1-yl-methyl)-3-(4-methoxyphenyl)acrylonitrile as intermediate. Z Naturforsch 2002;57b:579–85.10.1515/znb-2002-0516Search in Google Scholar

20. Mossman T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55–63.10.1016/0022-1759(83)90303-4Search in Google Scholar

21. Snedecor GM, Cochran WG. Statistical methods-7th ed. Lowa state University, Ames: loww3a USA Press, 1982:325.Search in Google Scholar

22. Kamal KA, Abd El-all KM, Elshemy HA, Kandeel MM. Design and synthesis of substituted chromenes as potential anticancer agents. Int J Pharm Res Develop 2012;4:310–22.Search in Google Scholar

23. Vosooghi M, Rajabalian S, Sorkhi M, Badinloo M, Nakhjiri M, Negahbani AS, et al. Synthesis and cytotoxic activity of some 2-amino-4-aryl-3-cyano-7-(dimethylamino)-4H-chromenes. Res Pharm Sci 2010;5:13–18.Search in Google Scholar

24. Kemnitzer W, Jiang S, Wang Y, Kasibhatla S, Crogan-Grundy C, Bubenick M, et al. Discovery of 4-Aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based hts assay. part 5: modifications of the 2- and 3-positions. Bioorg Med Chem Lett 2008;18:603–7.10.1016/j.bmcl.2007.11.078Search in Google Scholar PubMed

25. Kemnitzer W, Drewe J, Jiang S, Zhang H, Zhao J, Crogan-Grundy C, et al. Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 3. structure-activity relationships of fused rings at the 7,8-positions. J Med Chem 2007;50:2858–64.10.1016/j.bmcl.2005.07.066Search in Google Scholar PubMed

26. Kemnitzer W, Drewe J, Jiang S, Zhang H, Zhao J, Wang Y, et al. Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 4. structure-activity relationships of the 4-aryl group. J Med Chem 2004;47:6299–310.10.1021/jm049640tSearch in Google Scholar PubMed

27. Mahmoodi M, Aliabadi A, Emami S, Safavi M, Rajabalian S, Mohagheghi MA, et al. Synthesis and In-Vitro cytotoxicity of polyfunctionalized 4-(2-arylthiazol-4-yl)-4H-chromenes. Arch Pharm Chem 2010;343:411–6.10.1002/ardp.200900198Search in Google Scholar PubMed

28. Endo S, Matsunaga T, Kuwata K, Zhao H-T, El-Kabbani O, Kitade Y, et al. Chromene-3-carboxamide derivatives discovered from virtual screening as potent inhibitors of the tumor maker, AKR1B10. Bioorg Med Chem 2010;18:2485–90.10.1016/j.bmc.2010.02.050Search in Google Scholar PubMed

29. Tseng T-H, Chuang S-K, Hu C-C, Chang C-F, Huang Y-C, Lin C-W, et al. The synthesis of morusin as a potent antitumor agent. Tetrahedron 2010;66:1335–40.10.1016/j.tet.2009.12.002Search in Google Scholar

30. Rahman AU, Choudhary MI, Thomsen WJ. Bioassay technique for drug development. Reading, UK: Harwood Academic Publishers ISBN 0-203-34349-2, 2001.Search in Google Scholar


Supplemental Material:

The online version of this article (DOI: https://doi.org/10.1515/znc-2016-0139) offers supplementary material, available to authorized users.


Received: 2016-07-02
Revised: 2016-09-29
Accepted: 2016-10-13
Published Online: 2016-11-10
Published in Print: 2017-05-01

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 1.5.2024 from https://www.degruyter.com/document/doi/10.1515/znc-2016-0139/html
Scroll to top button