Next Article in Journal
Design, Synthesis and Antifibrotic Activities of Carbohydrate- Modified 1-(Substituted aryl)-5-trifluoromethyl-2(1H) Pyridones
Previous Article in Journal
Phytochemical Study and Anti-inflammatory, Antidiabetic and Free Radical Scavenger Evaluations of Krameria pauciflora Methanol Extract
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Benzothiazole, Benzimidazole and Benzoxazole Derivatives as Potential Antitumor Agents: Synthesis and Preliminary in Vitro Biological Evaluation

State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medicinal School, Sichuan University, Chengdu 610041, Sichuan, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2012, 17(1), 873-883; https://doi.org/10.3390/molecules17010873
Submission received: 12 December 2011 / Revised: 6 January 2012 / Accepted: 10 January 2012 / Published: 17 January 2012
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
In a previous hit-to-lead research program targeting anticancer agents, two promising lead compounds, 1a and 1b, were found. However, the poor solubility of 1a and 1b made difficult further in vivo studies. To solve this problem, a lead optimization was conducted through introducing N-methyl-piperazine groups at the 2-position and 6-position. To our delight, the optimized analogue 1d showed comparable antiproliferative activity in vitro with better solubility, compared with 1a. Based on this result, the replacement of the benzothiazole scaffold with benzimidazole and benzoxazole moieties afforded 1f and 1g, whose activities were fundamentally retained. In the preliminary in vitro biological evaluation, the immunofluorescence staining of HCT116 cells indicated that 1d, 1f and 1g led to cytosolic vacuolization which was not induced by 1a at low micromolecular concentrations. These results suggest that these optimized compounds might potentially constitute a novel class of anticancer agents, which merit further studies.

1. Introduction

Cancer is the leading cause of death in developed countries and the second leading cause of death in developing countries [1]. Although surgical resection is potentially curative, the risk of recurrence remains very high. Besides surgical resection, treatment strategies for high risk patients are still mainly based on adjuvant or neoadjuvant use of chemotherapeutic agents alone or in combination with radiotherapy. Unfortunately, the use of the above standard therapeutic protocols only results in a moderate decline in mortality and the risk of sustaining a recurrence of disease remains high. Thus, it is urgent to develop novel chemotherapeutic agents for the treatment of cancer. In a recent hit-to-lead research of anticancer agents [2], lead compounds 1a and 1b (Figure 1) with benzothiazole scaffolds, showed promising anticancer in vitro activities. However, the solubility of 1a and 1b was far from satisfactory for further in vivo research. As a result, a lead optimization of 1a and 1b was conducted.
Figure 1. The structure of lead compounds 1a and 1b.
Figure 1. The structure of lead compounds 1a and 1b.
Molecules 17 00873 g001
The N-methyl-piperazine group as found in imatinib is a commonly used motif in lead optimization [3]. Introduction of N-methylpiperazine moieties at the 2-position and 6-positions of the benzothiazole scaffold afforded compounds 1c, 1d and 1e. To determine the role of the benzothiazole scaffold, the benzoxazole analogue 1f and benzimidazole analogue 1g were also designed (Figure 2).
Figure 2. The structures of compounds 1c–g.
Figure 2. The structures of compounds 1c–g.
Molecules 17 00873 g002
Herein, we report the synthesis of 1c–g, preliminary structure-activity relationships (SARs) and biological evaluation of these anticancer agents.

2. Results and Discussion

2.1. Chemistry

The synthetic route of compounds 1c–g using a modified literature method [4,5] is outlined in Scheme 1. Taking 1d for example, the key intermediate 2a, 2-mercapto-6-aminobenzothiazole, was alkylated with side chain 3b under basic conditons to give 4a. Subsequent acylation of 4a with 2-methoxybenzoic acid yielded the target compound 1d. Besides the optimization of side chains, the replacement of the benzothiazole scaffold with benzimidazole and benzoxazole units (Table 1), afforded 1f and 1g which were prepared using the same synthetic strategy.
Scheme 1. General procedures for the synthesis of 1c–g.
Scheme 1. General procedures for the synthesis of 1c–g.
Molecules 17 00873 g004
Table 1. The anti-proliferative activity of 1a–g against HepG2 and HCT-116 cells.
Molecules 17 00873 i001
Table 1. The anti-proliferative activity of 1a–g against HepG2 and HCT-116 cells.
Molecules 17 00873 i001
CompoundR1R2XIC50(μM) a
HepG2HCT-116
1a4-Chlorobenzyl2-MethoxyphenylS0.641.52
1bBenzylChloromethylS1.131.16
1cN-methyl-piperazine-phenylChloromethylS>40.0>40.0
1dN-methyl-piperazine-phenyl2-MethoxyphenylS2.101.25
1e4-ChlorobenzylN-methylpiperazine-phenylS31.228.8
1fN-methyl-piperazine-phenyl2-MethoxyphenylN14.63.69
1gN-methyl-piperazine-phenyl2-MethoxyphenylO12.22.61
a The cytotoxicity effects of various compounds on cancer cells were determined by the MTT assay, and the results were expressed as the mean IC50 calculated from three independent experiments.
The key intermediate 2c was readily prepared from commercially available 2-amino-5-nitrophenol following cyclization and subsequent reduction of the nitro group according to the method represented in Scheme 2.
Scheme 2. Synthesis of 6-aminobenzofuran-2-thiol 2c.
Scheme 2. Synthesis of 6-aminobenzofuran-2-thiol 2c.
Molecules 17 00873 g005
Side chain 3b was prepared from 4-fluoronitrobenzene condensed with N-methylpiperazine, subsequent reduction and acylation as shown in Scheme 3.
Scheme 3. Synthesis of 2-chloro-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide 3b.
Scheme 3. Synthesis of 2-chloro-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide 3b.
Molecules 17 00873 g006

2.2. The MTT Assay

With 1c–g at hand, we evaluated their anti-proliferative activities (IC50) on HepG2 and HCT116 cells, using the MTT assay (Table 1). As shown in the Table, introduction of an N-methylpiperazine substituent at the 2-position (compound 1d) in the R1 group was well tolerated. To our surprise, replacement of the benzyl group in R1 with an N-methylpiperazine-phenyl group (compound 1c) led to almost total loss of activity. The introduction of N-methylpiperazine substituent at the 6-position (1e) in the R2 group had a detrimental effect on the potency, that is, about 20–50-fold decrease in potency, compared with that of 1a. After identifying that the N-methylpiperazine-phenyl substituent is well tolerated at the 2-position in the R1 group, we next turned to examining the role of the benzothiazole scaffold. Replacement of the benzothiazole ring with benzimidazole and benzoxazole yielded 1f and 1g with a slightly decrease in potency for HCT-116 cells compared with 1a, but showed no clear SAR trends. It was noteworthy that analogue 1d exhibited comparable inhibitory activity against HepG2 and HCT-116 cells.
To further study the cytotoxic profile, compound 1d was selected for evaluation of inhibitory activities against a panel of different types of human cancer cell lines: Prostate cancer cell line PC-3, human cervical carcinoma cell line Hela, ovarian cancer cell line SKOV-3, lung cancer cell line A549, melanoma cell line A375, nonsmall cell lung cancer cell line H460 and epithelial cancer cell line A431 (Table 2). As shown in Table 2, compound 1d also showed broad-spectrum inhibitory activity which was exhibited in previous research [2]. Some IC50 values were better than 1a, such as against Hela and SKOV-3 cells.
Table 2. The anti-proliferative activities of 1d against various cancer cell lines.
Table 2. The anti-proliferative activities of 1d against various cancer cell lines.
CompoundIC50(μM) a
PC-3HelaSKOV-3A549A375H460A431
1a1.34.56.21.92.22.31.3
1d1.21.51.31.12.31.82.5
a The cytotoxicity effects of compounds on cancer cells were determined by the MTT assay, and the results were expressed as the mean IC50 calculated from three independent experiments.

2.3. The Solubility Assay

The optimized analogues 1d, 1f and 1g showed comparable anti-proliferative activities in vitro compared with 1a. To conduct a preliminary assessment of the absorption in vivo, solubility was further determined (Table 3). As shown in Table 3, 1d, 1f and 1g showed better solubility both in ethanol and water after the introduction of N-methylpiperazine group.
Table 3. The solubility of compound 1a, 1d, 1f and 1g.
Table 3. The solubility of compound 1a, 1d, 1f and 1g.
Solvent (μg/mL)1a1d1f1g
ethanol 1.77317.92803572.2
water 0.0935.99981.7548.4

2.4. Immunofluorescence Staining

In our previous research, it was found that 1a could induce apoptosis of cancer cells. To further study the antitumor mechanisms of 1a, 1d, 1f and 1g in vitro, an immunofluorescence staining of HCT116 cells was conducted (Figure 3).
Figure 3. The immunofluorescence staining of 1a, 1d, 1f and 1g on HCT-116 cells for 24 h. Blue: nuclear; Green: tubulin.
Figure 3. The immunofluorescence staining of 1a, 1d, 1f and 1g on HCT-116 cells for 24 h. Blue: nuclear; Green: tubulin.
Molecules 17 00873 g003
As shown in Figure 3, the tubulin of HCT-116 cells was intact after the treatment with 1d, 1f and 1g. However, cytosolic vacuolization appeared, which was’t observed for 1a. It has been reported that this kind of cytosolic vacuolization is related with the inhibition of HSP-90 protein or the proteasome-promoted protein ubiquitination [6]. Further mechanistic studies are currently in progress.

3. Experimental

3.1. General

The human cancer cell lines, HepG2, HCT116, PC-9, Hela, SKOV-3, A549, A375, H460 and A431 were purchased from the American Type Culture Collection (ATCC, Rockville, MD, USA). Dulbecco’s modified Eagle medium (DMEM) and RPMI 1640 were purchased from Gibco (Grand Island, NY, USA). Fetal bovine serum (FBS) was obtained from Hyclone (Logan, UT, USA). The purity of compound screened in biological assays was determined to be >98% by HPLC analysis. An Symmetry C18 (75 mm × 4.6 mm, i.d. 3.5μm) (Waters, Milford, MA, USA) was used with a t elution of acetonitrile and HPLC-grade water as mobile phase. 1H-NMR were recorded at 400 MHz on a Varian spectrometer (Varian, Palo Alto, CA, USA) model Gemini 400. Mass Spectra (MS) were measured by Q-TOF Priemier mass spectrometer utilizing electrospray ionization (ESI) (Micromass, Manchester, UK).

3.2. Preparation of 6-Aminobenzo[d]oxazole-2-thiol (2c)

3.2.1. 6-Nitrobenzo[d]oxazole-2-thiol

6-Nitrobenzo[d]oxazole-2-thiol was synthesized according to a modified literature method [7]. A suspension of 2-amino-5-nitrophenol (3.08 g, 20 mmol) and potassium ethylxanthate (3.36 g, 21 mmol) in dry pyridine (40 mL) was stirred at 120 °C for 6 h, and then at room temperature for another 16 h. 2 M HCl solution was added to the solution to adjust the pH value to 6. The resulting precipitate was collected by filtration, washed with petroleum ether and then dried in vacuum to afford 3.22 g (97%) of the title compound. 1H-NMR (DMSO-d6) δ: 7.39 (d, 1H, J = 8 Hz), 7.86 (t, 1H, J = 8 Hz), 8.22 (dd, 1H, J1 = 4 Hz, J2 = 8 Hz)), 8.41 (s, 1H). ESI-MS (m/z, %): 197.1 (M+H)+.

3.2.2. 6-Aminobenzo[d]oxazole-2-thiol (2c)

A mixture of 6-nitrobenzo[d]oxazole-2-thiol (5 g, 25 mmol), iron powder (3.36 g, 60 mmol), and ammonium chloride (2.67 g, 50 mmol) was refluxed in a mixed solvent of ethanol (25 mL) and H2O (8 mL). After completion of the reaction, the mixture was filtered while hot and washed with petroleum ether. Standing at room temperature gave compound 2c as a bright yellow solid. Yield: 3.7 g, 90%. 1H-NMR (DMSO-d6) δ: 6.51 (dd, 2H, J1 = 4 Hz, J2 = 8 Hz), 6.64 (d, 2H, J = 4 Hz), 6.90 (d, 2H, J = 8 Hz). ESI-MS (m/z, %): 167.1 (M+H)+.

3.3. 2-Chloro-N-(4-chlorobenzyl)acetamide (3a)

2-Chloroacetyl chloride (4.79 g, 42 mmol) was added dropwise with stirring to a mixture of (4-chlorophenyl) methanamine (4.96 g, 35 mmol) in dichloromethane (50 mL) cooled to 0 °C. The resulting mixture was allowed to warm to ambient temperature and stirred for 1 h. After filtering, the solid was washed with petroleum ether and dried under vacuum for 12 h at 25–30 °C. The title compound 3a was obtained by recrystalization from ethanol. Yield: 7.25 g, 95%.1H-NMR (400 MHz, DMSO-d6): δ 4.13 (s, 2H), 4.29 (d, J = 6.0 Hz, 2H), 7.28 (d, J = 4.4 Hz, 2H), 7.39 (d, J = 8.0 Hz, 2H), 8.77 (s, 1H). ESI-MS (m/z, %): 218.0 (M−H)+.

3.4. 2-Chloro-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide (3b)

3.4.1. 1-Methyl-4-(4-nitrophenyl)piperazine

Briefly, 1-methylpiperazine (15.6 g, 156 mmol) was added slowly while stirring to a solution of 4-fluoronitrobenzene (20 g, 142 mmol) at room temperature. The mixture was stirred for 1 h, then filtered and washed with petroleum ether to give product as an orange solid. Yield: 30.3 g, 96.43%.1H-NMR (DMSO-d6) δ: 2.22 (s, 3H), 2.43 (t, 4H, J = 4 Hz), 3.45 (t, 4H, J = 4 Hz), 7.03 (d, 2H, J = 8 Hz), 8.05 (d, 2H, J = 8 Hz). ESI-MS (m/z, %): 222.0 (M+H)+.

3.4.2. 4-(4-Methylpiperazin-1-yl)aniline

A mixture of 1-methyl-4-(4-nitrophenyl) piperazine (14.8 g, 67 mmol), iron powder (15 g, 268 mmol), ammonium chloride (7.2 g, 134 mmol) was refluxed in a mixed solvent of ethanol (60 mL) and H2O (20 mL). After completion of the reaction, the mixture was filtered while hot and washed with petroleum ether. After standing at room temperature an off-white acicular solid was obtained. Yield: 12.2 g, 95.31%. 1H-NMR (DMSO-d6) δ: 2.22 (s, 3H), 2.5 (t, 4H, J = 4 Hz), 2.75 (t, 4H, J = 4 Hz), 3.22 (s, 2H), 6.55 (d, 2H, J = 8 Hz), 6.74 (d, 2H, J = 8 Hz). ESI-MS (m/z, %): 192.0 (M+H)+.

3.4.3. 2-Chloro-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide (3b)

2-Chloroacetyl chloride (4.79 g, 42 mmol) was added dropwise with stirring to a mixture of 1-methyl-4-(4-nitrophenyl) piperazine (6.69 g, 35 mmol) in dichloromethane (50 mL) cooled at 0 °C. The resulting mixture was allowed to warm to ambient temperature and stirred for 1 h. After filtering, the solid was washed with petroleum ether and dried under vacuum for 12 h at 25–30 °C. The title compound 3b was obtained by recrystalization from a solution of ethanol. Yield: 8.48 g, 90.51%.1H-NMR (DMSO-d6) δ: 2.21 (s, 3H), 2.43 (t, 4H, J = 4.8 Hz), 3.07 (t, 4H, J = 4.8 Hz), 4.2 (s, 2H), 6.90 (d, 2H, J = 8 Hz), 7.42 (d, 2H, J = 8 Hz), 10.10 (s, 1H). ESI-MS (m/z, %): 268.1 (M+H)+.

3.5. General Procedure for Preparing Compounds 4a–d

Compound 3a–b (13.76 mmol) was added to a mixture of 2a–c (16.51 mmol) and potassium carbonate (20.64 mmol) in acetone (25 mL) while refluxing with stirring. The resulting mixture was stirred under reflux for 4 h, then cooled to ambient temperature. After removal of acetone on a rotary evaporator, the solid was dissolved in ethyl acetate (100 mL) and washed with water (2 × 10 mL), saturated solution of sodium bicarbonate (2 × 20 mL) and sodium chloride (2 × 20 mL). Then, the organic phase was dried over Na2SO4 and concentrated in vacuo to give a solid product. The title compounds 4a–d were thus obtained, and could be used directly for the next step without further purification. The yields of 4a, 4b, 4c and 4d, were 92%, 90%, 90% and 93%, respectively.
2-(6-aminobenzo[d]thiazol-2-ylthio)-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide (4a): 1H-NMR (DMSO-d6) δ: 2.24 (s, 3H), 2.44 (t, 4H, J = 4 Hz), 3.06 (t, 4H, J = 4 Hz), 4.14 (s, 2H), 5.38 (s, 2H), 6.70 (dd, 1H, J1 = 2 Hz, J2 = 8.8 Hz), 6.88 (d, 2H, J = 9.2 Hz), 7.00 (d, 1H, J = 2.4 Hz), 7.41 (d, 2H, J = 8.8 Hz), 7.49 (d, 1H, J = 8.8 Hz), 10.16 (s, 1H). ESI-MS (m/z, %): 414.2 (M+H)+.
2-(6-amino-1H-benzo[d]imidazol-2-ylthio)-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide (4b): 1H-NMR (DMSO-d6) δ: 2.23 (s, 3H), 2.47 (s, 4H), 3.07 (s, 4H), 4.21 (s, 2H), 5.31 (s, 2H), 6.46 (d, 1H, J = 4 Hz), 6.56 (s, 1H), 6.88 (d, 2H, J = 8.8 Hz), 7.16 (s, 1H), 7.42 (d, 2H, J = 8.8 Hz), 10.40 (s, 1H), 12.13 (s, 1H). ESI-MS (m/z, %): 397.3 (M+H)+.
2-(6-aminobenzo[d]oxazol-2-ylthio)-N-(4-(4-methylpiperazin-1-yl)phenyl)acetamide (4c): 1H-NMR (DMSO-d6) δ: 2.23 (s, 3H), 2.46 (t, 4H, J = 4 Hz), 3.07 (t, 4H, J = 4 Hz), 4.23 (s, 2H), 5.31 (s, 2H), 6.56 (dd, 1H, J1 = 2 Hz, J2 = 8.4 Hz), 6.71 (d, 1H, J = 1.6 Hz), 6.89 (d, 2H, J = 8 Hz), 7.22 (t, 1H, J = 8 Hz), 7.41 (d, 2H, J = 8 Hz), 10.16 (s, 1H). ESI-MS (m/z, %): 398.2 (M+H)+.
2-(6-aminobenzo[d]thiazol-2-ylthio)-N-(4-chlorobenzyl)acetamide (4d): 1H-NMR (DMSO-d6) δ: 4.09 (s, 2H), 4.30 (d, 2H, J = 5.6 Hz), 5.40 (s, 2H), 6.73 (dd, 1H, J1 = 1.6 Hz, J2 = 8.8 Hz), 7.01 (d, 1H, J = 1.6 Hz), 7.28 (q, 4H, J = 8.4 Hz), 7.49 (d, 1H, J = 8.4 Hz), 8.81 (t, 1H, J = 5.6 Hz). ESI-MS (m/z, %): 364.0 (M+H)+.

3.6. 2-Chloro-N-(2-(2-(4-(4-methylpiperazin-1-yl)phenylamino)-2-oxoethylthio)benzothiazol-6-yl)-acetamide (1c)

2-Chloroacetyl chloride (43 mg, 0.38 mmol) was added dropwise with stirring to a solution of 4a (80 mg, 0.19 mmol) in dichloromethane (4 mL) originally at 0 °C. The resulting mixture was allowed to warm to ambient temperature and stirred for 1 h. After filtering, the solid was washed with petroleum ether and dried under vacuum for 12 h at 25–30 °C. The title compound 1c (an off-white solid) were obtained by recrystallization from ethanol. Yield: 72 mg, 77.42%. m.p. 245.8–246.1 °C. 1H-NMR (DMSO-d6) δ: 2.21 (s, 3H), 2.43 (t, J = 4.4 Hz, 4H), 3.07 (t, 4H, J = 4.8 Hz), 4.30 (s, 2H), 4.35 (s, 2H), 6.93 (d, 2H, J = 8.8Hz), 7.49 (d, 2H, J = 8.8 Hz), 7.55 (dd, 1H, J = 2 Hz, 8.8 Hz), 7.79 (d, 1H, J = 8.8 Hz), 8.39 (d, 1H, J = 2 Hz), 10.32 (s, 1H), 10.62 (s, 1H); ESI-MS (m/z, %): 490.3 (M+H)+.

3.7. General Procedure for Preparing Compounds 1d–g

A mixture of compound 4a–d (0.5 mmol), 2-methoxybenzoic acid hydrate or 4-(4-methylpiperazin-1-yl)benzoic acid (0.75 mmol), EDCI (0.75 mmol), HOBt (0.75 mmol), DIPEA (0.75 mmol) was warmed up to 80 °C in DMF (5 mL) with stirring. After completion of reaction, the mixture was diluted with ethyl acetate (100 mL). The resulting mixture was washed with washed with water (2 × 20 mL), saturated solution of sodium bicarbonate (2 × 20 mL) and sodium chloride (2 × 20 mL). Then, the organic phase was dried over Na2SO4 and concentrated in vacuo to give a solid product. The title compounds were obtained by recrystallization from ethanol or ethyl acetate.
2-Methoxy-N-(2-(2-(4-(4-methylpiperazin-1-yl)phenylamino)-2-oxoethylthio)benzo[d]thiazol-6-yl)benzamide (1d). Off-white solid. Yield: 76 mg, 75.47%. m.p. 172.1–172.4 °C. 1H-NMR (DMSO-d6) δ: 2.10 (s, 3H), 2.22 (t, 4H, J = 4.4 Hz), 3.08 (t, 4H, J = 4.8 Hz), 3.91 (s, 3H), 4.35 (s, 3H), 6.91 (d, 2H, J = 8.8 Hz), 7.09 (t, 1H, J = 7.2 Hz), 7.20 (d, 1H, J = 8.4 Hz), 7.48 (d, 2H, J = 8.4 Hz), 7.53 (t, 1H, J = 8 Hz), 7.67 (q, 2H, J = 8.8 Hz), 7.80 (d, 1H, J = 8.4 Hz), 8.55 (s, 1H), 10.22 (s,1H), 10.36 (s,1H); ESI-MS (m/z, %): 548.0 (M+H)+.
N-(2-(2-(4-Chlorobenzylamino)-2-oxoethylthio)benzo[d]thiazol-6-yl)-4-(4-methylpiperazin-1-yl)-benzamide (1e). Yellow solid. Yield: 184 mg, 65.36%. m.p. 177.8–179.2 °C. 1H-NMR (DMSO-d6) δ: 2.09 (s, 3H), 2.45 (t, 4H, J = 4.4 Hz), 3.30 (t, 4H, J = 4.8 Hz), 4.19 (s, 2H), 4.31 (d, 2H, J = 6 Hz), 7.03 (d, 2H, J = 8.8 Hz), 7.28 (d, 2H, J = 8.8 Hz), 7.32 (d, 2H, J = 8.4 Hz), 7.76 (s, 2H), 7.89 (d, 2H, J = 8.8 Hz), 8.52 (s, 1H), 8.85 (t, 1H, J = 6Hz), 10.17 (s, 1H); ESI-MS (m/z, %): 566.2 (M+H)+.
2-Methoxy-N-(2-(2-(4-(4-methylpiperazin-1-yl)phenylamino)-2-oxoethylthio)-1H-benzo[d]imidazol-6-yl)benzamide (1f). Faint yellow solid. Yield: 90 mg, 81.08%. m.p. 132.3–132.6 °C. 1H-NMR (DMSO-d6) δ: 2.21 (s, 3H), 2.43 (t, 4H, J = 4 Hz), 3.06 (t, 4H, J = 4 Hz), 3.90 (s, 3H), 4.21 (s, 2H), 6.88 (d, 2H, J = 8.8 Hz), 7.07 (t, 1H, J = 7.2 Hz), 7.18 (d, 1H, J = 8 Hz), 7.30 (q, 1H, J = 8.4 Hz), 7.43 (t, 3H, J = 8.8 Hz), 7.50 (t, 1H, J = 7.6 Hz), 7.64 (d, 1H, J = 7.2 Hz), 8.07 (d, 1H, J = 33.2 Hz), 10.08 (d, 1H, J = 32.8 Hz), 10.27 (s, 1H), 12.60 (s, 1H); ESI-MS (m/z, %): 531.3 (M+H)+.
2-Methoxy-N-(2-(2-(4-(4-methylpiperazin-1-yl)phenylamino)-2-oxoethylthio)benzo[d]oxazol-6-yl)-benzamide (1g). Yellow solid. Yield: 100 mg, 75.75%. m.p. 210.4–210.8 °C. 1H-NMR (DMSO-d6) δ: 2.24 (s, 3H), 2.40 (t, 4H, J = 4.4 Hz), 3.14 (t, 4H, J = 4.8 Hz), 3.89 (s, 3H), 4.23 (s, 2H), 6.91 (d, 2H, J = 8.8 Hz), 7.09 (t, 1H, J = 7.2 Hz), 7.20 (d, 1H, J = 8.4 Hz), 7.48 (d, 2H, J = 8.4 Hz), 7.53 (t, 1H, J = 8 Hz), 7.67 (q, 2H, J = 8.8 Hz), 7.80 (d, 1H, J = 8.4 Hz), 8.26 (s, 1H), 9.98 (s,1H), 10.29 (s,1H); ESI-MS (m/z, %): 532.2 (M+H)+.

3.8. Cell Culture

Cell lines including HepG2, Hela, Skov-3, A375 and A431 were maintained in Dulbecco’s modified Eagle medium (DMEM) containing 10% fetal bovine serum (FBS), penicillin (100 U/mL) and streptomycin (10 mg/L). Cell lines including HCT116, PC-9, A549 and H460 were maintained in RPMI 1640 containing 10% fetal bovine serum (FBS), penicillin (100 U/mL) and streptomycin (10 mg/L). Cells were grown in a 5% CO2 incubator at 37 °C.

3.9. Cell Proliferation Assay (MTT Assay)

Cells (2 × 103/well) were seeded in 96-well plates and cultured for 24 h, followed by compounds which dissolved in dimethylsulfoxide (DMSO) treatment for 48 h. Ten microliters of 10 mg/mL MTT was added and cultured for 4 h, the medium was removed and 150 μL of DMSO was added to dissolve formazan crystals. Absorbance was measured at 570 nm using an SpectraMAX M5 microplate spectrophotometer (Molecular Devices). The proliferation inhibitory effects of the compounds on cancer cells were expressed as IC50.

3.10. Immunofluorescence

Cells (1 ×105) were plated in 6-well plates. Following drug treatment for 24 h, cells were washed with PBS, punched by Triton X-100. Subsequently cells were probed by the primary antibodies. Then cells were incubated with the secondary antibody for 30 min at room temperature and examined under a fluorescence microscope [8].

4. Conclusions

In this paper, five novel benzothiazole-2-thiol, benzimidazole-2-thiol and benzoxazole-2-thiol derivatives were synthesized for lead optimization, and their in vitro antitumor activity was evaluated. To our delight, compounds 1d, 1f and 1g showed comparable antitumor activities and better solubility compared with the lead compound 1a. In a preliminary antitumor activity mechanism study, we found a novel phenomenon, that is, cytosolic vacuolization after treatment with these compounds which was not induced by 1a. Thus, it indicates that compounds 1d, 1f and 1g might possess different anticancer mechanism, which deserves further study.

Acknowledgements

We thank the Analytical and Testing Center of Sichuan University for NMR measurements.
  • Sample Availability: Samples of the compounds 1c–g are available from the authors.

References and Notes

  1. The Global Burden of Disease: 2004 Update; World Health Organization: Geneva, Switzerland, 2008.
  2. Wang, Z.; Shi, X.-H.; Wang, J.; Zhou, T.; Xu, Y.-Z.; Huang, T.-T.; Li, Y.-F.; Zhao, Y.-L.; Yang, L.; Yang, S.-Y.; et al. Synthesis, structure-activity relationships and preliminary antitumor evalutation of benzothiazole-2-thiol derivatives as novel apoptosis inducers. Bioorg. Med. Chem. Lett. 2011, 21, 1907–1101. [Google Scholar]
  3. Capdeville, R.; Buchdunger, E.; Zimmermann, J.; Matter, A. Glivec (STI571, imatinib), a rationaly developed, targeted anticancer drug. Nat. Rev. Drug Discov. 2002, 1, 493–502. [Google Scholar] [CrossRef]
  4. Baraldi, P.G.; Preti, D.; Tabrizi, M.A.; Fruttarolo, F.; Saponaro, G.; Baraldi, S.; Romagnoli, R.; Moorman, AR.; Gessi, S.; Varani, K.; Borea, P.A. N(6)-[(hetero)aryl/(cyclo)alkyl-carbamoyl-methoxy-phenyl]-(2-chloro)-5'-N-ethylcarboxamido-adenosines: The first example of adenosine-related structures with potent agonist activity at the human A(2B) adenosine receptor. Bioorg. Med. Chem. 2007, 15, 2514–2527. [Google Scholar] [CrossRef]
  5. Mussom, D.L.; Cochran, F.R.; Kelley, J.L.; Mclean, E.W.; Selph, J.L.; Rigdon, G.C.; Orr, G.F.; Davis, R.G.; Cooper, B.R.; Styles, V.L.; et al. Indanylidenes. 1. Design and synthesis of (E)-2-(4, 6-difluoro-1-indanylidene)acetamide, a potent, centrally acting muscle relaxant with antiinflammatory and analgesic activity. J. Med. Chem. 2003, 46, 399–408. [Google Scholar] [CrossRef]
  6. Mimnaugh, E.G.; Xu, W.; Vos, M. Simultaneous inhibition of hsp 90 and the proteasome promotes protein ubiquitination, causes endoplasmic reticulum-derived cytosolic vacuolization, and enhances antitumor activity. Mol. Cancer Ther. 2004, 3, 551–566. [Google Scholar]
  7. Wynne, G.M.; Wren, S.P.; Johnson, P.D.; Price, P.D.; De Moor, O.; Nugent, G.; Storer, R.; Pye, R.J.; Dorgan, C.R. Treatment of duchenne muscular dystrophy. WO 2007/091106 A2, 16 August 2007. [Google Scholar]
  8. Kanzawa, T.; Kondo, Y.; Ito, H.; Kondo, S.; Germano, I. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res. 2003, 63, 2103–2108. [Google Scholar]

Share and Cite

MDPI and ACS Style

Xiang, P.; Zhou, T.; Wang, L.; Sun, C.-Y.; Hu, J.; Zhao, Y.-L.; Yang, L. Novel Benzothiazole, Benzimidazole and Benzoxazole Derivatives as Potential Antitumor Agents: Synthesis and Preliminary in Vitro Biological Evaluation. Molecules 2012, 17, 873-883. https://doi.org/10.3390/molecules17010873

AMA Style

Xiang P, Zhou T, Wang L, Sun C-Y, Hu J, Zhao Y-L, Yang L. Novel Benzothiazole, Benzimidazole and Benzoxazole Derivatives as Potential Antitumor Agents: Synthesis and Preliminary in Vitro Biological Evaluation. Molecules. 2012; 17(1):873-883. https://doi.org/10.3390/molecules17010873

Chicago/Turabian Style

Xiang, Pu, Tian Zhou, Liang Wang, Chang-Yan Sun, Jing Hu, Ying-Lan Zhao, and Li Yang. 2012. "Novel Benzothiazole, Benzimidazole and Benzoxazole Derivatives as Potential Antitumor Agents: Synthesis and Preliminary in Vitro Biological Evaluation" Molecules 17, no. 1: 873-883. https://doi.org/10.3390/molecules17010873

Article Metrics

Back to TopTop