Next Article in Journal
3-Benzoylisoxazolines by 1,3-Dipolar Cycloaddition: Chloramine-T-Catalyzed Condensation of α-Nitroketones with Dipolarophiles
Next Article in Special Issue
Hispolon Induces Apoptosis, Suppresses Migration and Invasion of Glioblastoma Cells and Inhibits GBM Xenograft Tumor Growth In Vivo
Previous Article in Journal
The Identification of New Triterpenoids in Eucalyptus globulus Wood
Previous Article in Special Issue
New Estrone Oxime Derivatives: Synthesis, Cytotoxic Evaluation and Docking Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis and Anticancer Activity of a New Series of N-aryl-N′-[4-(pyridin-2-ylmethoxy)benzyl]urea Derivatives

1
Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
2
School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(12), 3496; https://doi.org/10.3390/molecules26123496
Submission received: 23 April 2021 / Revised: 26 May 2021 / Accepted: 29 May 2021 / Published: 8 June 2021
(This article belongs to the Special Issue Anticancer Agents: Design, Synthesis and Evaluation II)

Abstract

:
The development of cancer treatments requires continuous exploration and improvement, in which the discovery of new drugs for the treatment of cancer is still an important pathway. In this study, based on the molecular hybridization strategy, a new structural framework with an N-aryl-N’-arylmethylurea scaffold was designed, and 16 new target compounds were synthesized and evaluated for their antiproliferative activities against four different cancer cell lines A549, MCF7, HCT116, PC3, and human liver normal cell line HL7702. The results have shown seven compounds with 1-methylpiperidin-4-yl groups having excellent activities against all four cancer cell lines, and they exhibited scarcely any activities against HL7702. Among them, compound 9b and 9d showed greatly excellent activity against the four kinds of cells, and the IC50 for MCF7 and PC3 cell lines were even less than 3 μM.

Graphical Abstract

1. Introduction

Nowadays, cancer has become a major challenge in human health, and a leading cause of death [1,2]. Cancer is caused by the uncontrolled proliferation of cells, a kind of behavior unusual for cells, mostly related to some abnormal signal transduction regulation mechanisms. In the diagnosis and treatment of cancer, many great developments have been made, such as in the field of surgery, drug therapy from toxic drugs to targeted drugs, etc. [3,4]. However, traditional chemotherapy drugs often have serious side effects and adverse reactions. The emergence of small-molecule targeted drugs has eased the severe side effects of chemotherapy drugs to a certain extent, but targeted drugs are also prone to drug resistance problems with the prolonged administration time [5,6,7]. Therefore, it is a great challenge and opportunity to continuously develop new candidate drug molecules to bring new drugs to cancer treatment.
Compared with earlier targeted drugs, multi-target inhibitors can act on a variety of different targets and inhibit different signal pathways at the same time [6,8,9]. The multi-target inhibitors have a broader anti-tumor spectra and better prospects in clinical practice [7]. Urea and urea isosteres are structures that possesses both a hydrogen bond acceptor and a hydrogen bond donor, which makes it easy to form better interactions with drug target proteins [10]. These kinds of structures are an excellent pharmacodynamic structure in drug molecules [11]. In many small-molecule targeted kinase inhibitors, urea and urea isosteres, including aminopyrimidine, have been used in drug structures, some of which showed really favorable anti-cancer activity (Figure 1) [12,13].
Among the currently clinically used multi-target kinase inhibitors, sorafenib and its derivative regorafenib with a diaryl urea structure were the prime representatives, because they had excellent inhibitory effects on a variety of solid tumors [14,15]. In sorafenib, the rigidity of the diaryl urea structure causes the molecular rotation to not happen freely, thus the poor solubility of sorafenib results in low bioavailability [16,17]. This study is based on the structure of sorafenib and the retention of the urea scaffold, in which, in order to enhance the molecular flexibility, a carbon atom was inserted into the diaryl urea structure, and the basic urea scaffold was changed to an N-aryl-N’-benzylurea scaffold. Moreover, the diaryl ether fragment with a pyridyl group was also modified to a 4-(pyridylmethoxy)phenyl fragment. The nitrogen atom in the pyridine ring is believed to continue to play a key role, for example, as a hydrogen bond acceptor with some proteins. Meanwhile, in order to keep its position relative to the core fragment urea unchanged, the position of the nitrogen atom in the pyridine ring linked to the core urea fragment was replaced from the 4-position to the 2-position. Validity has been verified by a simulation using the Discovery Studio 3.0 software [18], and the distances between the nitrogen atom in the pyridine ring and the urea moiety in sorafenib and a target compound are 10.386Å and 10.604Å, respectively (Figure 2).
Furthermore, in previous reports, some proton pump inhibitors showed anticancer activity; lansoprazole was one of the drugs that performed well [19,20]. Based on the molecular hybridization and structural optimization strategies, the 3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-ylmethyl moiety in lansoprazole and the N-aryl-N’-benzylurea scaffold were retained into the target compounds (Figure 3) [21]. Considering the extra space in some intracellular protein serine/threonine kinases such as BRAF kinase binding with sorafenib, a new series of N-aryl-N’-benzylurea derivatives modified with a 1-methylpiperidin-4-yl group on the 3-position of the urea scaffold were designed, which are expected to block intracellular signal transduction and enhance their antiproliferative activity [22,23].

2. Results and discussion

2.1. Chemistry

As shown in Scheme 1, the target molecules were synthesized starting with 2-(chloromethyl)-3-methyl-4-(2,2,2-trifluoromethoxy)pyridine hydrochloride (1) and vanillin (2) via a Williamson reaction and obtained the ether compound 3, which reacted with hydroxylamine to convert the oxime 4. The oxime 4 was reduced by Ni-Al to obtain benzylamine 5. The reaction between benzylamine 5 and 1-methylpiperidin-4-one through reductive amination yielded the key intermediate 6. Finally, the different intermediate isocyanate 7, prepared by a reaction between substituting aniline or benzylamine, triphosgene, and TEA, were mixed with compound 5 or compound 6 to yield the targeted compounds 8a8i and 9a9g.

2.2. Biological Activity Evaluation

The target compounds were evaluated for their antiproliferative activity against different human cancer cell lines, including A549 (non-small cell lung cancer cell line), MCF-7 (breast cancer cell line), HCT116 (colon cancer cell line), PC-3 (prostate cancer cell line) and HL7702 (human liver normal cell line) by using the MTT assay with sorafenib as the control drug. The evaluated results as IC50 values are shown in Table 1.
As shown in Table 1, all the target compounds exhibited weak cytotoxic activities against HL7702, and most of the target compounds exhibit excellent antiproliferative activity against the A549 cell line and HCT116 cell line. The IC50 values of target compounds 8c, 9b, and 9d against the A549 cells line were less than 5 μM, and the IC50 values of compounds 9b, 9d, and 9g against the HCT116 cell line were less than 3 μM. The antiproliferative activities against the MCF7 cell line and the PC3 cell line of target compounds 9a9g with the 1-methylpiperidin-4-yl group were significantly higher than that of compounds 8a8i without the 1-methylpiperidin-4-yl group. The IC50 values of compounds 9a, 9b, 9d, and 9e against the MCF7 cell line and the PC3 cell line were less than 3 μM and 5 μM, respectively. Among them, target compounds 9b and 9d have shown a more potent antiproliferative activity against the four cancer cell lines with excellent IC50 values (under 5 μM) compared to the control drug sorafenib.
The analyses of the structure-activity relationships of the target compounds with the 1-methylpiperidin-4-yl group were summarized as follows: (1) The introduction of fluorine atoms on the R1 substituent of the benzene ring was mostly beneficial to the antiproliferative activity. For example, Compounds 9a, 9b, 9d, and 9e with the fluorine atoms in substituent on the phenyl show a better antiproliferative activity against the MCF7 cell line and the PC3 cell line than the control drug sorafenib, especially in the MCF7 cell line. The inhibitory activities against several cell lines of compound 9f with the nitro group were relatively weaker than that of several compounds with fluorine atoms on the R1 substituent of the benzene ring. (2) The introduction of electron-withdrawing group substituent R1 on the benzene ring results in an increase in antiproliferative activity. The antiproliferative activities against all four cancer cell lines of the target compounds 9b and 9d with a trifluoromethyl group were significantly higher than compound 9a with a methoxy group and 9c with a trifluoromethoxy group.
The antiproliferative activities of the target compounds without the 1-methylpiperidin-4-yl group have shown similar structure-activity relationships. For example, the antiproliferative activity against the HCT116 cell line of compounds 8a, 8b, 8c, and 8d with fluorine atoms in substituent on the phenyl were also better than that of the other compounds without fluorine atoms, and the IC50 value of compound 8c was less than 5 μM against the A549 cell line.

2.3. Cell Cycle Analysis

The effect of compound 9b on the cell cycle was assayed. After treatment of MCF-7 cells with compound 9b for 72 h at different concentrations (2.5, 5, 10, 20 μM), the percentages of cells in the G2/M phase were 16.7%, 23.5%, 27.7%, and 39.3%, respectively (Figure 4), indicating that compound 9b could cause an obvious G2/M arrest in a concentration-dependent manner with a concomitant decrease in terms of the number of cells in other phases of the cell cycle.

3. Materials and Methods

3.1. Chemistry

3.1.1. General Information

All reagents were obtained from commercial suppliers and used without further purification. The progress of the reactions was monitored by thin-layer chromatography (TLC) on silica gel plates and the spots visualized under ultraviolet (UV) light (254 nm). The column chromatography was performed using 200–300 mesh silica gel (Qingdao Haiyang, Qingdao, China). Mass spectra were measured with an electrospray (ESI-MS) on an Agilent 1100 Series LC/MSD Trap (Agilent Corporation, Santa Clara, CA, USA). 1H-NMR and 13C-NMR spectra were recorded on Bruker NMR spectrophotometers (Karlsruhe, Germany) using DMSO-d6 as the solvent. The IR spectra were measured using a Bruker Fourier number infrared spectrometer (Agilent Corporation, Santa Clara, CA, USA). 1H-NMR, 13C-NMR, ESI-MS and HRMS spectra of the target compounds are available in the Supplementary Material (Figures S1–S80).

3.1.2. Synthesis of 3-methoxy-4-[(3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy] benzaldehyde (3)

A mixture of 2-(chloromethyl)-3-methyl-4-(2,2,2-trifluoroethoxy)pyridine hydrochloride 1 (27.6 g, 100 mmol), vanillin 2 (15.22 g, 100 mmol), K2CO3 (69 g, 0.5 mol) and DMF(100 mL) was stirred at 80 °C overnight. The reaction was monitored by TLC (PE:EA = 3:1). The mixture was poured into 750 mL of water and stirred for 30 min until the K2CO3 was completely dissolved. The solids that precipitated out were filtered, washed with 2 mol/L NaOH aqueous solution and water, then dried to obtain a white solid 34.54 g. The yield was 97%, ESI-MS: 356.3([M + H]+).

3.1.3. Synthesis of (E)-3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl) methoxy)benzaldehyde oxime (4)

A mixture of hydroxylamine hydrochloride (3.06 g, 44 mmol) and NaHCO3 (3.69 g, 44 mmol) in water (50 mL) was stirred at room temperature until no gas was released. A solution of 3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzaldehyde 3 (14.21 g,40 mmol) in EtOH (50 mL) was added into the mixture and continued string for 3 h. The progress of the reaction can be confirmed by TLC. EtOH was removed under reduced pressure, and the white solid was precipitated, filtered off with suction, and washed with water. After drying, the white solid obtained was 14.6 g, with a yield of 98%, ESI-MS: 371.3([M + H]+).

3.1.4. Synthesis of (3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)phenyl)methanamine (5)

An aqueous solution of NaOH (5 mol/L, 60 mL) was added into the solution of 3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzaldehyde oxime 4 (11.10 g,30 mmol) in EtOH (50 mL) under an ice bath. Nickel-aluminum alloy (10 g) was slowly added into the mixture in several times, during which a lot of gas was generated. Then slowly returned to room temperature and stirred overnight. The progress of the reaction was monitored by TLC. After removing the solid by suction filtration, EtOH was distilled off under reduced pressure. The residual solution was extracted by EA, and the organic phase was washed by water and brine, then dried by Na2SO4. After the solvent was removed under reduced pressure, the white solid obtained was 10.14 g, with a yield of 95%, ESI-MS: 357.2([M + H]+).

3.1.5. Synthesis of N-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl) methoxy)benzyl)-1-methylpiperidin-4-amine (6)

A mixture of 1-methylpiperidin-4-one (2.26 g, 20 mmol), (3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)phenyl)methanamine(7.12 g, 20 mmol) 5, AcOH and MeOH was stirred for 1h at room temperature. NaBH3CN was added in 3 times, during which a lot of gas was generated and stirring was continued for 3 h. The progress of the reaction was monitored by TLC. After MeOH was distilled off under reduced pressure, a paste mixture was obtained. An aqueous solution of NaOH (2 mol/L) was added to the mixture and stirred until the paste dissolved. The solution was extracted by EA, and the organic phase was washed by water and brine, then dried by Na2SO4. After the solvent was removed under reduced pressure, a yellowish oil of 7.02 g was obtained, with a yield of 77%, 454.2([M + H]+).

3.1.6. General Procedure for the Synthesis of the Target Urea Derivatives

Triphosgene (0.20 g, 0.67 mmol) was dissolved in 10 mL DCM, a solution of substituted aniline or benzylamine (2 mmol) in 10 mL DCM was slowly dropped in during stirring. There were solids that gradually precipitated out. Then a solution of TEA (0.4 g, 4 mmol) in DCM (10 mL) was slowly dropped into the mixture, the solids gradually dissolved, and the solution of substituted isocyanate 7 was obtained. The solution of 5 or 6 (2 mmol) in 10 mL DCM was added. After the reaction was completed, the mixture was washed by water and brine and dried by Na2SO4. After DCM was distilled off under reduced pressure, the mixture was purified by silica gel chromatography (DCM:EA = 5:1, v/v) to afford 8a8i and silica gel chromatography (DCM:MeOH = 20:1, v/v) to afford 9a9g.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(4-(trifluoromethoxy)phenyl)urea (8a), white powder 0.99 g, yield 89%; m.p.: 158.8–159.3 °C; MS: 560.4([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.73 (s, 1H), 8.34 (d, J = 5.7 Hz, 1H), 7.54–7.47 (m, 2H), 7.23 (d, J = 8.6 Hz, 2H), 7.14 (d, J = 5.7 Hz, 1H), 7.05 (d, J = 8.2 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 6.81 (dd, J = 8.2, 2.0 Hz, 1H), 6.59 (t, J = 5.9 Hz, 1H), 5.14 (s, 2H), 4.92 (q, J = 8.8 Hz, 2H), 4.23 (d, J = 5.7 Hz, 2H), 3.74 (s, 3H), 2.22 (s, 3H).; 13C NMR (101 MHz, DMSO-d6) δ 161.82, 155.94, 155.51, 149.63, 148.02, 147.18, 142.55, 140.26, 133.67, 124.25 (q, J = 277.9 Hz), 122.08, 121.99, 120.69 (q, J = 254.9 Hz), 119.69, 119.19, 114.27, 112.07, 108.02, 71.56, 65.11 (q, J = 34.7 Hz), 55.95, 43.10, 10.30; IR: 3399, 3053, 3008, 2971, 2829, 1706, 1556, 1507, 1454, 1416, 1256, 1220, 1194, 1154, 1015, 911, 847, 796, 672, 645, 544; HRMS: 558.146914([M − H]) for C25H22F6N3O5, 560.161467([M + H]+) for C25H24F6N3O5.
 
1-(3-chloro-4-fluorophenyl)-3-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)urea (8b), white powder 0.86 g, yield 82%; m.p.: 167.8–169.1 °C; MS:528.3([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.72 (s, 1H), 8.34 (d, J = 5.7 Hz, 1H), 7.77 (dd, J = 6.8, 2.4 Hz, 1H), 7.32–7.18 (m, 2H), 7.14 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.2 Hz, 1H), 6.93 (d, J = 2.0 Hz, 1H), 6.80 (dd, J = 8.2, 1.9 Hz, 1H), 6.63 (t, J = 5.8 Hz, 1H), 5.13 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.22 (d, J = 5.7 Hz, 2H), 3.74 (s, 3H), 2.22 (s, 3H).; 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.94, 155.44, 152.33 (d, J = 240.0 Hz), 149.59, 148.04, 147.15, 138.26 (d, J = 2.8 Hz), 124.27 (q, J = 277.4 Hz), 122.05, 119.69, 119.44 (d, J = 18.9 Hz), 119.31, 118.23 (d, J = 6.5 Hz), 117.13 (d, J = 21.3 Hz), 114.26, 112.08, 108.06, 71.55, 65.08 (q, J = 34.2 Hz), 56.00, 43.09, 10.35; IR: 3313, 2944, 2883, 1641, 1564, 1500, 1477, 1420, 1390, 1308, 1258, 1209, 1164, 1131, 1008, 970, 911, 862, 800, 757, 647, 576, 445; HRMS: 526.116220([M − H]) for C24H21ClF4N3O4, 528.130773([M + H]+) for C24H23ClF4N3O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(4-(trifluoromethyl)phenyl)urea (8c), white powder 0.78 g, yield 72%; m.p: 169.9–171.0 °C; MS:544.5([M + H]+), 566.1([M + Na]+); 1H NMR (400 MHz, DMSO-d6) δ 8.95 (s, 1H), 8.34 (d, J = 5.7 Hz, 1H), 7.59 (q, J = 8.8 Hz, 4H), 7.14 (d, J = 5.7 Hz, 1H), 7.05 (d, J = 8.2 Hz, 1H), 6.95 (d, J = 2.0 Hz, 1H), 6.82 (dd, J = 8.2, 1.9 Hz, 1H), 6.69 (t, J = 5.8 Hz, 1H), 5.14 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.24 (d, J = 5.7 Hz, 2H), 3.74 (s, 3H), 2.22 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.93, 155.25, 149.61, 148.03, 147.19, 144.64, 133.47, 126.37, 125.09 (q, J = 270.6 Hz), 124.26 (q, J = 277.5 Hz), 122.06, 119.73, 117.72, 114.26, 112.10, 108.05, 71.55, 65.09 (q, J = 35.0 Hz), 55.98, 43.10, 10.31; IR: 3414, 3376, 2940, 2886, 1703, 1686, 1581, 1534, 1512, 1477, 1408, 1321, 1256, 1220, 1180, 1155, 1135, 1102, 1063, 1008, 979, 862, 842, 812, 595, 554;HRMS: 542.151999 ([M − H]) for C25H22F6N3O4, 544.166552([M + H]+)+ for C25H24F6N3O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(3-(trifluoromethyl)phenyl)urea (8d), white powder 0.83 g, yield 76%; m.p.: 153.2–154.2 °C; MS:544.2([M + H]+), 542.0([M − H]); 1H NMR (400 MHz, DMSO-d6) δ 8.90 (s, 1H), 8.34 (d, J = 5.7 Hz, 1H), 7.98 (s, 1H), 7.52 (d, J = 8.3 Hz, 1H), 7.45 (t, J = 7.9 Hz, 1H), 7.23 (d, J = 7.6 Hz, 1H), 7.14 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.2 Hz, 1H), 6.94 (d, J = 1.9 Hz, 1H), 6.81 (dd, J = 8.2, 1.9 Hz, 1H), 6.68 (t, J = 5.9 Hz, 1H), 5.13 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.23 (d, J = 5.8 Hz, 2H), 3.74 (s, 3H), 2.22 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.82, 155.94, 155.46, 149.62, 148.02, 147.18, 141.80, 133.59, 130.11, 124.73 (q, J = 272.3 Hz), 124.25 (q, J = 277.4 Hz), 122.07, 121.62, 119.69, 117.68, 114.28, 114.06, 112.08, 108.02 (d, J = 4.4 Hz), 71.56, 65.10 (q, J = 34.2 Hz), 55.97 (d, J = 3.2 Hz), 43.08, 10.33; IR: 3412, 3374, 2940, 2876, 1702, 1582, 1551, 1514, 1477, 1442, 1380, 1341, 1312, 1255, 1221, 1182, 1158, 1111, 1067, 1028, 1007, 979, 891, 864, 813, 796, 702, 664, 597, 552; HRMS: 542.151999([M − H]) for C25H22F6N3O4+, 544.161552 ([M + H]+) for C25H24F6N3O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(4-methoxyphenyl)urea (8e), white powder 0.59 g, yield 58%; m.p.: 179.5–180.3 °C; MS:566.2([M + H]+), 504.0([M − H]); 1H NMR (400 MHz, DMSO-d6) δ 8.28 (s, 1H), 7.29 (d, J = 8.9 Hz, 2H), 7.14 (d, J = 5.7 Hz, 1H), 6.93 (d, J = 1.9 Hz, 1H), 6.81 (dd, J = 9.0, 7.1 Hz, 3H), 6.40 (t, J = 5.9 Hz, 1H), 5.13 (s, 2H), 4.92 (q, J = 8.8 Hz, 2H), 4.20 (d, J = 5.8 Hz, 2H), 3.70 (s, 3H), 2.22 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.80, 155.96, 155.86, 154.40, 149.57, 148.05, 147.08, 134.08, 133.97, 124.28 (d, J = 277.9 Hz), 122.05, 119.91, 119.64, 114.33, 114.27, 112.06, 108.07, 71.58, 65.08 (q, J = 34.5 Hz), 55.99, 55.59, 43.09, 10.36; IR: 3312, 2940, 2839, 1631, 1571, 1508, 1467, 1417, 1376, 1363, 1308, 1271, 1241, 1160, 1136, 1030, 973, 862, 827, 669, 578, 524, 423; HRMS: 504.175179 ([M − H]) for C25H25F3N3O5, 506.189732 ([M + H]+) for C25H27F3N3O5.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(3-nitrophenyl)urea (8f), yellow powder 0.31 g, yield 30%; m.p.: 168.9–170.5 °C; MS: 521.2([M + H]+), 519.0([M − H]); 1H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 8.52 (t, J = 2.2 Hz, 1H), 8.33 (d, J = 5.8 Hz, 1H), 7.75 (d, J = 8.3 Hz, 1H), 7.66 (d, J = 8.4 Hz, 1H), 7.50 (dd, J = 9.1, 7.3 Hz, 1H), 7.13 (d, J = 5.9 Hz, 1H), 7.04 (d, J = 8.2 Hz, 1H), 6.96–6.92 (m, 1H), 6.85–6.71 (m, 2H), 5.13 (s, 2H), 4.91 (q, J = 8.8 Hz, 2H), 4.24 (d, J = 5.9 Hz, 2H), 3.74 (s, 3H), 2.21 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.83, 155.93, 155.35, 149.63, 148.59, 148.02, 147.20, 142.28, 133.51, 130.31, 124.26 (q, J = 277.8 Hz), 124.16, 122.07, 119.74, 115.95, 114.32, 112.14, 112.06, 108.05, 71.56, 65.11 (q, J = 34.4 Hz), 56.02, 43.11, 10.34; IR: 3410, 3010, 2943, 2882, 2832, 1701, 1584, 1527, 1503, 1480, 1383, 1347, 1318, 1264, 1207, 1161, 1138, 1122, 1033, 1000, 980, 868, 814, 794, 735, 671, 612, 585, 557, 445; HRMS: 519.149693 ([M − H]) for C24H22F3N4O6, 521.164246([M + H]+) for C24H24F3N4O6.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(4-methoxybenzyl)urea (8g), white powder 0.39 g, yield 38%; m.p.: 153.0–154.2 °C; MS: 520.2([M + H]+), 517.9([M − H]); 1H NMR (400 MHz, DMSO-d6) δ 9.08 (s, 1H), 8.52 (t, J = 2.2 Hz, 1H), 8.33 (d, J = 5.8 Hz, 1H), 7.75 (d, J = 8.3 Hz, 1H), 7.66 (d, J = 8.4 Hz, 1H), 7.50 (dd, J = 9.1, 7.3 Hz, 1H), 7.13 (d, J= 5.9 Hz, 1H), 7.04 (d, J = 8.2 Hz, 1H), 6.96–6.92 (m, 1H), 6.85–6.71 (m, 2H), 5.13 (s, 2H), 4.91 (q, J = 8.8 Hz, 2H), 4.24 (d, J = 5.9 Hz, 2H), 3.74 (s, 3H), 2.21 (s, 3H); 13C NMR (101 MHz, DMSO-d6) δ 161.82, 158.54, 158.46, 155.99, 149.57, 148.04, 146.99, 134.47, 133.30, 124.28 (q, J = 277.7 Hz), 122.06, 119.46, 114.26, 114.07, 111.86, 108.08, 71.61, 65.10 (q, J = 34.9 Hz), 55.92, 55.51, 43.25, 42.90, 10.36; IR: 3349, 3301, 2949, 2925, 2884, 2832, 1605, 1579, 1561, 1515, 1468, 1424, 1363, 1363, 1363, 1275, 1251, 1169, 1156, 1133, 1103, 1038, 975, 863, 816, 728, 637, 561; HRMS: 518.190829 ([M − H]) for C26H27F3N3O5, 520.205382 ([M + H]+) for C26H29F3N3O5.
 
1-(4-ethoxybenzyl)-3-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)urea (8h), white powder 0.57 g, yield 53%; m.p.: 148.9–149.9 °C; MS: 534.3 ([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.34 (d, J = 5.7 Hz, 1H), 7.20–7.11 (m, 3H), 7.02 (d, J = 8.2 Hz, 1H), 6.90–6.81 (m, 3H), 6.75 (dd, J = 8.3, 1.9 Hz, 1H), 6.30 (td, J = 6.1, 2.3 Hz, 2H), 5.13 (s, 2H), 4.92 (q, J = 8.8 Hz, 2H), 4.15 (d, J = 5.8 Hz, 4H), 3.99 (q, J = 7.0 Hz, 2H), 3.71 (s, 3H), 2.22 (s, 3H), 1.31 (t, J = 7.0 Hz, 3H); 13C NMR (101 MHz, DMSO-d6) δ 161.82, 158.48, 157.77, 155.96, 149.54, 148.02, 146.96, 134.46, 133.16, 128.76, 124.28 (q, J= 277.7, 277.3 Hz), 122.06, 119.44, 114.56, 114.21, 111.81, 108.08, 71.57, 65.08 (q, J = 34.5 Hz), 63.39, 55.89, 43.24, 42.89, 15.11, 10.36; IR: 3331, 2978, 2927, 2883, 1609, 1579, 1556, 1516, 1471, 1423, 1390, 1275, 1249, 1148, 1025, 974, 861, 815, 753, 728, 639, 574, 548; HRMS: 532.206479 ([M − H]) for C27H29F3N3O5, 534.221032([M + H]+) for C27H31F3N3O5.
 
1-(4-(dimethylamino)benzyl)-3-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)urea (8i), white powder 0.73 g, yield 69%; m.p.: 159.4–161.4 °C; MS:533.9([M + H]+), 555.4([M + Na]+); 1H NMR (400 MHz, DMSO-d6) δ 8.34 (d, J = 5.6 Hz, 1H), 7.14 (d, J = 5.7 Hz, 1H), 7.11–7.06 (m, 2H), 7.02 (d, J = 8.2 Hz, 1H), 6.87 (d, J = 2.0 Hz, 1H), 6.74 (dd, J = 8.2, 1.9 Hz, 1H), 6.69–6.65 (m, 2H), 6.22 (dt, J = 21.9, 5.9 Hz, 2H), 5.12 (s, 2H), 4.92 (q, J = 8.7 Hz, 2H), 4.15 (d, J = 5.9 Hz, 2H), 4.10 (d, J = 5.8 Hz, 2H), 3.71 (s, 3H), 2.85 (s, 6H), 2.22 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 158.46, 155.98, 149.97, 149.55, 148.04, 146.96, 134.50, 128.77, 128.49, 124.28 (q, J = 277.7 Hz), 122.05, 119.44, 114.22, 112.85, 111.82, 108.08, 71.59, 65.08 (q, J = 34.3 Hz), 55.92, 43.23, 43.06, 10.36; IR: 3336, 2940, 2918, 2879, 1613, 1570, 1517, 1468, 1421, 1308, 1256, 1233, 1175, 1137, 1039, 1011, 969, 922, 854, 810, 736, 650, 564; HRMS: 531.222464 ([M − H]) for C27H30F3N4O4, 522.237017 ([M + H]+) for C27H32F3N4O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)-3-(4-(trifluoromethoxy)phenyl)urea (9a), white powder 0.79 g, yield 60%; m.p.: 127.3–129.4 °C; MS:657.2([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.52 (s, 1H), 8.33 (d, J = 5.6 Hz, 1H), 7.58–7.51 (m, 2H), 7.23 (d, J = 8.6 Hz, 2H), 7.14 (d, J = 5.7 Hz, 1H), 7.03 (d, J = 8.3 Hz, 1H), 6.90 (d, J = 2.0 Hz, 1H), 6.75 (dd, J = 8.2, 1.9 Hz, 1H), 5.11 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.52 (s, 2H), 4.10 (tt, J = 12.0, 4.0 Hz, 1H), 3.70 (s, 3H), 2.83 (d, J = 11.6 Hz, 2H), 2.20 (d, J = 5.4 Hz, 6H), 2.06 (d, J = 10.0 Hz, 2H), 1.70 (tt, J = 12.4, 6.7 Hz, 2H), 1.55 (d, J = 14.4 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.73, 149.48, 148.05, 146.93, 143.15, 140.30, 133.64, 124.26(q, J =276.1Hz), 121.50, 118.77, 120.66 (q, J = 253.5 Hz), 111.28, 108.07, 71.55, 65.11 (q, J = 34.8, 34.0 Hz), 55.95, 55.08, 53.09, 45.99, 45.70, 45.16, 39.69, 29.90, 10.36; IR: 3397, 2950, 2848, 2799, 1648, 1584, 1513, 1470, 1416, 1377, 1293, 1256, 1227, 1204, 1159, 1132, 1031, 982, 921, 847, 825, 800, 754, 536; HRMS: 655.236063 ([M − H]) for C31H33F6N4O5, 657.250616 ([M + H]+) for C31H35F6N4O5.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)-3-(4-(trifluoromethyl)phenyl)urea (9b), white powder 0.73 g, yield 57%; m.p.: 138.4–140.5 °C; MS: 641.1([M + H]+), 321.5 ([M + 2H]2+), 639.5([M − H]); 1H NMR (400 MHz, DMSO-d6) δ 8.73 (s, 1H), 8.33 (d, J = 5.6 Hz, 1H), 7.67 (d, J = 8.6 Hz, 2H), 7.58 (d, J = 8.7 Hz, 2H), 7.14 (d, J = 5.7 Hz, 1H), 7.03 (d, J = 8.3 Hz, 1H), 6.89 (d, J = 2.0 Hz, 1H), 6.75 (dd, J = 8.3, 2.0 Hz, 1H), 5.10 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.54 (s, 2H), 4.16–4.07 (m, 1H), 3.70 (s, 3H), 2.84 (d, J = 10.9 Hz, 2H), 2.19 (s, 6H), 2.06 (t, J = 10.0 Hz, 2H), 1.72 (q, J = 12.6, 11.8 Hz, 2H), 1.57 (d, J = 11.4 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.92, 155.50, 149.49, 148.05, 146.97, 144.86, 133.47, 125.91 (q, J = 3.9 Hz), 125.08 (q, J = 270.9 Hz), 124.26 (q, J = 277.6, 277.1 Hz), 122.16 (q, J = 31.8 Hz), 122.08, 118.76, 114.21, 111.26, 108.06, 71.54, 65.11 (q, J = 34.4 Hz), 55.94, 55.06, 53.24, 45.70, 45.25, 29.88, 10.34; IR: 3387, 2943, 2846, 2802, 1650, 1584, 1513, 1468, 1416, 1378, 1313, 1250, 1224, 1162, 1131, 1063, 1030, 1016, 981, 862, 843, 811, 753, 577; HRMS: 639.241148 ([M − H]) for C31H33F6N4O4, 641.255701([M + H]+) for C31H35F6N4O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-3-(4-methoxyphenyl)-1-(1-methylpiperidin-4-yl)urea (9c), white powder 0.71 g, yield 59%; m.p.: 179.1–180.8; MS:603.2([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.33 (d, J = 5.7 Hz, 1H), 8.12 (s, 1H), 7.33–7.26 (m, 2H), 7.14 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 6.90 (d, J = 2.0 Hz, 1H), 6.84–6.74 (m, 3H), 5.11 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.49 (s, 2H), 4.08 (tt, J = 11.3, 3.9 Hz, 1H), 3.71 (d, J = 1.9 Hz, 6H), 2.80 (d, J = 11.0 Hz, 2H), 2.21 (s, 3H), 2.17 (s, 3H), 2.06–1.95 (m, 2H), 1.67 (qd, J = 12.1, 3.9 Hz, 2H), 1.54 (d, J = 12.0 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 156.08, 155.96, 155.07, 149.46, 148.06, 146.87, 133.99, 133.88, 124.27 (q, J = 278.0 Hz), 122.70, 122.06, 118.80, 114.19, 113.89, 111.29, 108.08, 71.57, 65.10 (q, J = 33.8 Hz), 55.96, 55.59, 55.28, 52.98, 46.03, 45.03, 30.19, 10.38; IR: 3403, 2943, 2836, 2788, 2759, 1642, 1583, 1511, 1467, 1446, 1416, 1373, 1295, 1250, 1220, 1159, 1128, 1034, 1011, 962, 860, 824, 737, 666, 576, 542, 441; HRMS: 637.241006 ([M + Cl]) for C31H37ClF3N4O5, 603.278881 ([M + H]+) for C31H38F3N4O5.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)-3-(3-(trifluoromethyl)phenyl)urea (9d), white powder 0.75 g, yield 59%; m.p.: 160.4–162.2 °C; MS: 641.2([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.69 (s, 1H), 8.33 (d, J = 5.7 Hz, 1H), 7.91 (d, J = 2.0 Hz, 1H), 7.74 (dd, J = 8.2, 2.0 Hz, 1H), 7.46 (t, J = 8.0 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 7.14 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 6.90 (d, J = 2.0 Hz, 1H), 6.76 (dd, J = 8.3, 2.0 Hz, 1H), 5.11 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.54 (s, 2H), 4.11 (tt, J = 12.1, 3.9 Hz, 1H), 3.70 (s, 3H), 2.84 (d, J = 11.2 Hz, 2H), 2.20 (s, 6H), 2.05 (d, J = 13.2 Hz, 2H), 1.72 (tt, J = 12.4, 6.8 Hz, 2H), 1.57 (d, J = 11.8 Hz, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.93, 155.63, 149.48, 148.06, 146.94, 141.89, 133.50, 129.78, 129.51 (q, J = 32.3, 31.7 Hz), 124.76 (d, J = 272.5 Hz), 124.26 (q, J = 278.9, 278.5 Hz), 123.84, 118.75, 118.39 (d, J = 3.9 Hz), 116.39 (d, J = 4.4 Hz), 114.22, 111.25, 108.08, 71.54, 65.10 (q, J = 34.6 Hz), 55.94, 55.05, 53.16, 46.04, 45.68, 45.21, 29.82, 10.35; IR: 3385, 2940, 2839, 2791, 2771, 2735, 1645, 1583, 1513, 1493, 1468, 1444, 1376, 1326, 1247, 1222, 1152, 1122, 1030, 1000, 972, 909, 835, 788, 749, 701, 667, 540, 458; HRMS: 639.241148 ([M − H]) for C31H33F6N4O4, 641.255701 ([M + H]+) for C31H35F6N4O4.
 
3-(3-chloro-4-fluorophenyl)-1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)urea (9e), white powder 0.69 g, yield 55%; m.p.: 176.2–177.7 °C; MS:625.2([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.54 (s, 1H), 8.33 (d, J = 5.6 Hz, 1H), 7.74 (dd, J = 6.9, 2.6 Hz, 1H), 7.41 (ddd, J = 9.2, 4.4, 2.6 Hz, 1H), 7.27 (t, J = 9.1 Hz, 1H), 7.14 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 6.89 (d, J = 2.0 Hz, 1H), 6.75 (dd, J = 8.2, 1.9 Hz, 1H), 5.11 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.51 (s, 2H), 4.08 (tt, J = 12.1, 3.9 Hz, 1H), 3.71 (s, 3H), 2.83 (d, J = 11.2 Hz, 2H), 2.20 (d, J = 2.9 Hz, 6H), 2.05 (d, J = 11.7 Hz, 2H), 1.71 (qd, J = 12.3, 3.7 Hz, 2H), 1.59–1.51 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 161.81, 155.93, 155.62, 152.83 (d, J = 240.9 Hz), 149.46, 148.06, 146.93, 138.30, 133.52, 124.27 (q, J = 277.7 Hz), 122.06, 121.79, 120.59 (d, J = 6.5 Hz), 119.01 (d, J = 18.2 Hz), 118.75, 116.71 (d, J = 21.4 Hz), 114.20, 111.26, 108.08, 71.54, 65.10 (q, J = 34.6 Hz), 55.97, 55.09, 53.16, 45.77, 45.19, 29.89, 10.37; IR: 3403, 3365, 2940, 2881, 2822, 1702, 1598, 1515, 1448, 1407, 1370, 1319, 1260, 1217, 1151, 1107, 1063, 965, 906, 842, 785, 713, 593, 509, 455; HRMS: 623.205307 ([M − H]) for C30H33ClF4N4O4, 625.219923 ([M + H]+) for C30H35ClF4N4O4.
 
1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)-3-(3-nitrophenyl)urea (9f), yellow powder 0.79 g, yield 64%; m.p.: 157.5–159.1 °C; MS:618.2([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.86 (s, 1H), 8.47 (t, J = 2.3 Hz, 1H), 8.33 (d, J = 5.7 Hz, 1H), 7.92 (dd, J = 8.2, 2.0 Hz, 1H), 7.79 (dd, J = 8.1, 2.2 Hz, 1H), 7.51 (t, J = 8.2 Hz, 1H), 7.13 (d, J = 5.7 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 6.91 (d, J = 2.0 Hz, 1H), 6.76 (dd, J = 8.3, 2.0 Hz, 1H), 5.10 (s, 2H), 4.91 (q, J = 8.7 Hz, 2H), 4.55 (s, 2H), 4.10 (tt, J = 11.9, 4.0 Hz, 1H), 3.71 (s, 3H), 2.80 (d, J = 11.0 Hz, 2H), 2.20 (s, 3H), 2.16 (s, 3H), 2.00 (t, J = 11.3 Hz, 2H), 1.71 (qd, J = 12.1, 3.8 Hz, 2H), 1.61–1.52 (m, 2H); 13C NMR (101 MHz, DMSO-d6) δ 161.80, 155.93, 155.52, 149.48, 148.28, 148.06, 146.95, 142.46, 133.45, 129.97, 126.24, 124.27 (q, J = 277.6 Hz), 122.06, 118.74, 116.58, 114.26, 114.21, 111.25, 108.07, 71.54, 65.10 (q, J = 34.4 Hz), 55.97, 55.21, 53.48, 46.01, 45.24, 30.08, 10.37; IR: 3366, 2940, 2842, 2781, 1657, 1585, 1512, 1467, 1426, 1376, 1343, 1248, 1222, 1161, 1131, 1032, 1011, 967, 861, 824, 737, 667, 584, 454; HRMS: 616.238842 ([M − H]) for C30H33F3N5O6, 618.253395 ([M + H]+) for C30H35F3N5O6.
 
3-(4-ethoxybenzyl)-1-(3-methoxy-4-((3-methyl-4-(2,2,2-trifluoroethoxy)pyridin-2-yl)methoxy)benzyl)-1-(1-methylpiperidin-4-yl)urea (9g), white powder 0.41 g, yield 33%; m.p.: 119.0–121.2 °C; MS 631.3([M + H]+); 1H NMR (400 MHz, DMSO-d6) δ 8.34 (d, J = 5.7 Hz, 1H), 7.13 (dd, J = 15.2, 7.1 Hz, 3H), 7.01 (d, J = 8.3 Hz, 1H), 6.84–6.75 (m, 4H), 6.72 (dd, J = 8.3, 2.0 Hz, 1H), 5.11 (s, 2H), 4.92 (q, J = 8.7 Hz, 2H), 4.36 (s, 2H), 4.18 (d, J = 5.6 Hz, 2H), 3.98 (q, J = 6.9 Hz, 3H), 3.63 (s, 3H), 2.79 (d, J = 11.0 Hz, 2H), 2.21 (s, 3H), 2.17 (s, 3H), 2.00 (t, J = 11.8 Hz, 2H), 1.61 (qd, J = 12.2, 3.8 Hz, 2H), 1.48 (d, J = 9.9 Hz, 2H), 1.32 (d, J = 7.0 Hz, 3H). 13C NMR (101 MHz, DMSO-d6) δ 161.82, 157.98, 157.62, 155.99, 149.46, 148.06, 146.82, 134.24, 133.60, 128.63, 124.28 (q, J = 277.9 Hz), 122.08, 118.80, 114.39, 114.08, 111.14, 108.09, 71.61, 65.12 (q, J = 34.4 Hz), 63.37, 55.79, 55.18, 52.47, 45.79, 44.88, 43.52, 29.99, 15.12, 10.38; IR: 3348, 2937, 2881, 2837, 2793, 1612, 1584, 1509, 1477, 1447, 1395, 1374, 1292, 1253, 1162, 1131, 1033, 1003, 971, 916, 804, 772, 577; HRMS: 665.272306 ([M + Cl]) for C33H41ClF3N4O5, 631.310182 ([M + H]+) for C33H42F3N4O5.

3.2. Biological Evaluation

3.2.1. Antiproliferative Activity Assays

The antiproliferative activities of target compounds were determined using a standard MTT assay [24,25,26,27]. Exponentially growing cells A549 (1.5 × 103 cells/well), MCF-7 (2.2 × 103 cells/well), HCT-116 (800 cells/well), PC-3 (2.0 × 103 cells/well) and HL7702 (5.0 × 103 cells/well) were seeded into 96-well plates and incubated for 24 h to allow the cells to attach. Then, a fresh medium containing various concentrations of the candidate compounds was added to each well. The cells were then incubated for 96 h, thereafter MTT assays were performed, and cell viability was assessed at 570 nm by a microplate reader (ThermoFisher Scientific (Shanghai) Instrument Co., Ltd., Shanghai, China). The optical densities (OD) at 570 nm were measured, and the IC50 of the target compounds was calculated by using GraphPad Prism 5.0 software to perform nonlinear fitting with the cell survival rate under different concentrations of the compounds.

3.2.2. Cell Cycle Analysis

As for the flow cytometric analysis of DNA content, 1 × 105 MCF-7 cells in exponential growth were treated with different concentrations of compound 9b for 24 h. After an incubation period, the cells were collected, centrifuged, and fixed with ice-cold ethanol (70%). The cells were then treated with buffer containing RNAse A and 0.1% Triton X-100 and then stained with the propidium iodide (PI). The samples were analyzed on a flow cytometer (Becton, Dickinson, Franklin Lakes, NJ, USA) [28]. Data were analyzed using Flowjo software v9.0.

4. Conclusions

In summary, based on the structure of sorafenib and lansoprazole, 16 target N-aryl-N’-arylmethylurea derivatives were designed with molecular hybridization and synthesized, and their antiproliferative activities were assayed. The target compounds 9b and 9d have shown excellent antiproliferative activities against all four kinds of tumor cell lines (non-small cell lung cancer A549, breast cancer MCF-7, colon cancer HCT116, prostate cancer PC-3). All target compounds have demonstrated weak cytotoxic activities against human liver normal cell line HL7702. The biological assay results showed these target compounds with the 1-methylpiperidin-4-yl group on the 3-position of urea in the target compounds and substituents containing fluorine atoms on the phenyl ring exhibit potently antiproliferative activities. The cell cycle evaluation has shown that compound 9b could cause an obvious G2/M arrest in a concentration-dependent manner.

Supplementary Materials

The following are available online, 1H-NMR, 13C-NMR, ESI-MS and HRMS of the target compounds (Figures S1–S80).

Author Contributions

Proposal for the subject: C.H. and X.L.; synthetic work and the characterization of all target compounds: S.H., S.L., C.Z., Y.H., J.L., H.H. and X.Z.; biological assays: S.H., X.Z. and H.Z.; preparation of the manuscript: S.H. and C.Z.; review and correction for the manuscript: C.H., X.L. and H.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Science Foundation of China (Grant No. 21342006), the Program for Innovative Research Team of the Ministry of Education of China (Grant No. IRT_14R36).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding authors.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of all target compounds are available from the authors.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Miller, K.D.; Nogueira, L.; Mariotto, A.B.; Rowland, J.H.; Yabroff, K.R.; Alfano, C.M.; Jemal, A.; Kramer, J.L.; Siegel, R.L. Cancer treatment and survivorship statistics, 2019. CA Cancer J. Clin. 2019, 69, 363–385. [Google Scholar] [CrossRef] [Green Version]
  3. Santos, R.; Ursu, O.; Gaulton, A.; Bento, A.P.; Donadi, R.S.; Bologa, C.G.; Karlsson, A.; Al-Lazikani, B.; Hersey, A.; Oprea, T.I.; et al. A comprehensive map of molecular drug targets. Nat. Rev. Drug Discov 2017, 16, 19–34. [Google Scholar] [CrossRef]
  4. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  5. Widmer, N.; Bardin, C.; Chatelut, E.; Paci, A.; Beijnen, J.; Levêque, D.; Veal, G.; Astier, A. Review of therapeutic drug monitoring of anticancer drugs. Part two—targeted therapies. Eur. J. Cancer 2014, 50, 2020–2036. [Google Scholar] [CrossRef] [PubMed]
  6. Melisi, D.; Piro, G.; Tamburrino, A.; Carbone, C.; Tortora, G. Rationale and clinical use of multitargeting anticancer agents. Curr. Opin. Pharmacol. 2013, 13, 536–542. [Google Scholar] [CrossRef] [PubMed]
  7. Guo, T.; Ma, S. Recent advances in the discovery of multitargeted tyrosine kinase inhibitors as anticancer agents. ChemMedChem 2020, 16, 600–620. [Google Scholar] [CrossRef] [PubMed]
  8. Raghavendra, N.M.; Pingili, D.; Kadasi, S.; Mettu, A.; Prasad, S.V.U.M. Dual or multi-targeting inhibitors: The next generation anticancer agents. Eur. J. Med. Chem. 2018, 143, 1277–1300. [Google Scholar] [CrossRef] [PubMed]
  9. Singh, H.; Kinarivala, N.; Sharma, S. Multi-targeting anticancer agents: Rational approaches, synthetic routes and structure activity relationship. Anti-Cancer Agent. Me. 2019, 19, 842–874. [Google Scholar] [CrossRef]
  10. Auria-Luna, F.; Marqués-López, E.; Romanos, E.; Fernández-Moreira, V.; Gimeno, M.C.; Marzo, I.; Herrera, R.P. Novel ureido-dihydropyridine scaffolds as theranostic agents. Bioorganic Chem. 2020, 105, 104364. [Google Scholar] [CrossRef] [PubMed]
  11. Türe, A.; Kahraman, D.C.; Cetin-Atalay, R.; Helvacıoğlu, S.; Charehsaz, M.; Küçükgüzel, İ. Synthesis, anticancer activity, toxicity evaluation and molecular docking studies of novel phenylaminopyrimidine—(thio)urea hybrids as potential kinase inhibitors. Comput. Biol. Chem. 2019, 78, 227–241. [Google Scholar] [CrossRef] [PubMed]
  12. Kilic-Kurt, Z.; Ozmen, N.; Bakar-Ates, F. Synthesis and anticancer activity of some pyrimidine derivatives with aryl urea moieties as apoptosis-inducing agents. Bioorganic Chem. 2020, 101, 104028. [Google Scholar] [CrossRef] [PubMed]
  13. Zi, M.; Liu, F.; Wu, D.; Li, K.; Zhang, D.; Zhu, C.; Zhang, Z.; Li, L.; Zhang, C.; Xie, M.; et al. Discovery of 6-arylurea-2-arylbenzoxazole and 6-arylurea-2-arylbenzimidazole derivatives as angiogenesis inhibitors: Design, synthesis and in vitro biological evaluation. ChemMedChem 2019, 14, 1291–1302. [Google Scholar] [CrossRef]
  14. Zhu, Y.-J.; Zheng, B.; Wang, H.-Y.; Chen, L. New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacolo. Sin. 2017, 38, 614–622. [Google Scholar] [CrossRef] [Green Version]
  15. Arai, H.; Battaglin, F.; Wang, J.; Lo, J.H.; Soni, S.; Zhang, W.; Lenz, H.J. Molecular insight of regorafenib treatment for colorectal cancer. Cancer Treat. Rev. 2019, 81, 101912. [Google Scholar] [CrossRef]
  16. Azimian, F.; Hamzeh-Mivehroud, M.; Shahbazi Mojarrad, J.; Hemmati, S.; Dastmalchi, S. Synthesis and biological evaluation of diaryl urea derivatives designed as potential anticarcinoma agents using de novo structure-based lead optimization approach. Eur. J. Med. Chem. 2020, 201, 112461. [Google Scholar] [CrossRef]
  17. Aghcheli, A.; Toolabi, M.; Ayati, A.; Moghimi, S.; Firoozpour, L.; Bakhshaiesh, T.O.; Nazeri, E.; Norouzbahari, M.; Esmaeili, R.; Foroumadi, A. Design, synthesis, and biological evaluation of 1-(5-(benzylthio)-1,3,4-thiadiazol-2-yl)-3-phenylurea derivatives as anticancer agents. Med. Chem. Res 2020, 29, 2000–2010. [Google Scholar] [CrossRef]
  18. Sanmartín, C.; Plano, D.; Domínguez, E.; Font, M.; Calvo, A.; Prior, C.; Encío, I.; Palop, J.A. Synthesis and pharmacological screening of several aroyl and heteroaroyl selenylacetic acid derivatives as cytotoxic and antiproliferative agents. Molecules 2009, 14, 3313–3338. [Google Scholar] [CrossRef]
  19. Fako, V.E.; Wu, X.; Pflug, B.; Liu, J.-Y.; Zhang, J.-T. Repositioning proton pump inhibitors as anticancer drugs by targeting the thioesterase domain of human fatty acid synthase. J. Med. Chem. 2015, 58, 778–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Zhang, B.; Ling, T.; Zhaxi, P.; Cao, Y.; Qian, L.; Zhao, D.; Kang, W.; Zhang, W.; Wang, L.; Xu, G.; et al. Proton pump inhibitor pantoprazole inhibits gastric cancer metastasis via suppression of telomerase reverse transcriptase gene expression. Cancer Lett. 2019, 452, 23–30. [Google Scholar] [CrossRef]
  21. Viegas-Junior, C.; Danuello, A.; Bolzani, V.D.; Barreir, E.J.; Fraga, C.A.M. Molecular hybridization: A useful tool in the design of new drug prototypes. Curr. Med. Chem. 2007, 14, 1829–1852. [Google Scholar] [CrossRef]
  22. Foster, S.A.; Whalen, D.M.; Ozen, A.; Wongchenko, M.J.; Yin, J.; Yen, I.; Schaefer, G.; Mayfield, J.D.; Chmielecki, J.; Stephens, P.J.; et al. Activation mechanism of oncogenic deletion mutations in BRAF, EGFR, and HER2. Cancer Cell 2016, 29, 477–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Simard, J.R.; Getlik, M.; Grutter, C.; Pawar, V.; Wulfert, S.; Rabiller, M.; Rauh, D. Development of a fluorescent-tagged kinase assay system for the detection and characterization of allosteric kinase inhibitors. J. Am. Chem. Soc. 2009, 131, 13286–13296. [Google Scholar] [CrossRef] [PubMed]
  24. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immun. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  25. Zhang, L.; Deng, X.S.; Zhang, C.; Meng, G.P.; Wu, J.F.; Li, X.S.; Zhao, Q.C.; Hu, C. Design, synthesis and cytotoxic evaluation of a novel series of benzo[d]thiazole-2-carboxamide derivatives as potential EGFR inhibitors. Med. Chem. Res. 2017, 26, 2180–2189. [Google Scholar] [CrossRef]
  26. Zhang, C.; Tan, X.; Feng, J.; Ding, N.; Li, Y.; Jin, Z.; Meng, Q.; Liu, X.; Hu, C. Design, synthesis and biological evaluation of a new series of 1-aryl-3-{4-[(pyridin-2-ylmethyl)thio]phenyl}urea derivatives as antiproliferative agents. Molecules 2019, 24, 2108. [Google Scholar] [CrossRef] [Green Version]
  27. Ahmed, N.M.; Youns, M.M.; Soltan, M.K.; Said, A.M. Design, synthesis, molecular modeling and antitumor evaluation of novel indolyl-pyrimidine derivatives with EGFR inhibitory activity. Molecules 2021, 26, 1838. [Google Scholar] [CrossRef] [PubMed]
  28. Xu, Y.M.; Jing, D.W.; Chen, R.; Rashid, U.H.; Jiang, J.; Liu, X.; Wang, L.S.; Xie, P. Design, synthesis and evaluation of novel sophoridinic imine derivatives containing conjugated planar structure as potent anticancer agents. Bioorg. Med. Chem. 2018, 26, 4136–4144. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The application of urea and its isosteres in anticancer drugs.
Figure 1. The application of urea and its isosteres in anticancer drugs.
Molecules 26 03496 g001
Figure 2. Spatial distance of the two nitrogen atoms in pyridine ring and urea moiety of sorafenib (left) and a target compound (right).
Figure 2. Spatial distance of the two nitrogen atoms in pyridine ring and urea moiety of sorafenib (left) and a target compound (right).
Molecules 26 03496 g002
Figure 3. Design of target compounds with molecular hybridization strategy.
Figure 3. Design of target compounds with molecular hybridization strategy.
Molecules 26 03496 g003
Scheme 1. Synthetic route of the target compounds. Reagents and conditions: a. K2CO3, DMF, 80 °C, yield 97%; b. NH2OH·HCl, NaHCO3, EtOH, H2O, yield 98%; c. Ni-Al, NaOH, EtOH, H2O, yield 95%; d. 1-methylpiperidin-4-one, NaBH3CN, AcOH, MeOH yield 77%; e. amine, triphosgene, TEA, DCM; f. isocyanate 7, DCM.
Scheme 1. Synthetic route of the target compounds. Reagents and conditions: a. K2CO3, DMF, 80 °C, yield 97%; b. NH2OH·HCl, NaHCO3, EtOH, H2O, yield 98%; c. Ni-Al, NaOH, EtOH, H2O, yield 95%; d. 1-methylpiperidin-4-one, NaBH3CN, AcOH, MeOH yield 77%; e. amine, triphosgene, TEA, DCM; f. isocyanate 7, DCM.
Molecules 26 03496 sch001
Figure 4. Effects of compound 9b on MCF7 cell cycle progress for 24 h. (a) Treatment of MCF7 cells with compound 9b at different concentrations (0 μM, 2.5 μM, 5 μM, 10 μM, 20 μM) for 24 h. (b) Quantitative analysis of cell cycle.
Figure 4. Effects of compound 9b on MCF7 cell cycle progress for 24 h. (a) Treatment of MCF7 cells with compound 9b at different concentrations (0 μM, 2.5 μM, 5 μM, 10 μM, 20 μM) for 24 h. (b) Quantitative analysis of cell cycle.
Molecules 26 03496 g004aMolecules 26 03496 g004b
Table 1. The chemical structures and inhibitory activities of the target compounds.
Table 1. The chemical structures and inhibitory activities of the target compounds.
No.Structure IC50(μM)
A549MCF7HCT116PC3HL7702
8a Molecules 26 03496 i0015.30 ± 1.45>507.25 ± 0.87>50>50
8b Molecules 26 03496 i00217.65 ± 5.6510.98 ± 1.689.33 ± 1.3829.13 ± 5.81>50
8c Molecules 26 03496 i0034.88 ± 1.94>5011.38 ± 3.28>50>50
8d Molecules 26 03496 i00426.47 ± 5.66>509.44 ± 1.22>50>50
8e Molecules 26 03496 i00529.80 ± 5.09>5028.81 ± 3.11>50>50
8f Molecules 26 03496 i00612.40 ± 0.6013.26 ± 2.2713.35 ± 2.7815.87 ± 0.73>50
8g Molecules 26 03496 i007>50>507.84 ± 1.40>50>50
8h Molecules 26 03496 i00810.93 ± 2.02>5023.31 ± 3.12>50>50
8i Molecules 26 03496 i0099.85 ± 4.40>5010.88 ± 2.85>50>50
9a Molecules 26 03496 i01017.53 ± 2.952.59 ± 0.294.41 ± 0.144.10 ± 0.19>50
9b Molecules 26 03496 i0114.93 ± 0.462.56 ± 0.072.90 ± 0.163.36 ± 0.17>50
9c Molecules 26 03496 i01215.76 ± 1.514.65 ± 0.7312.90 ± 1.5912.35 ± 1.75>50
9d Molecules 26 03496 i0133.17 ± 0.222.63 ± 0.082.56 ± 0.263.62 ± 0.27>50
9e Molecules 26 03496 i0145.48 ± 4.362.56 ± 0.165.82 ± 0.214.53 ± 0.33>50
9f Molecules 26 03496 i0156.09 ± 0.293.18 ± 0.305.18 ± 0.337.39 ± 0.63>50
9g Molecules 26 03496 i0166.04 ± 0.414.23 ± 0.472.93 ± 0.266.25 ± 0.43>50
sorafenib 6.16 ± 0.463.54 ± 0.193.88 ± 0.365.26 ± 0.46>50
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hou, S.; Liang, S.; Zhang, C.; Han, Y.; Liang, J.; Hu, H.; Zhang, X.; Hu, C.; Liu, X.; Zhang, H. Design, Synthesis and Anticancer Activity of a New Series of N-aryl-N′-[4-(pyridin-2-ylmethoxy)benzyl]urea Derivatives. Molecules 2021, 26, 3496. https://doi.org/10.3390/molecules26123496

AMA Style

Hou S, Liang S, Zhang C, Han Y, Liang J, Hu H, Zhang X, Hu C, Liu X, Zhang H. Design, Synthesis and Anticancer Activity of a New Series of N-aryl-N′-[4-(pyridin-2-ylmethoxy)benzyl]urea Derivatives. Molecules. 2021; 26(12):3496. https://doi.org/10.3390/molecules26123496

Chicago/Turabian Style

Hou, Shicheng, Shishao Liang, Chao Zhang, Yingmei Han, Jianhui Liang, Hongyu Hu, Xingeng Zhang, Chun Hu, Xiaoping Liu, and Hong Zhang. 2021. "Design, Synthesis and Anticancer Activity of a New Series of N-aryl-N′-[4-(pyridin-2-ylmethoxy)benzyl]urea Derivatives" Molecules 26, no. 12: 3496. https://doi.org/10.3390/molecules26123496

Article Metrics

Back to TopTop