Abstract
The molecular chaperone HSP90 plays an essential role in cancer occurrence and development. Therefore, it is an important target for the development of anticancer drugs. 1,3-Dibenzyl-2-aryl imidazolidine (8) is a previously reported inhibitor of HSP90; however, its anticancer activity is poor. In this work, chemical modification of 8 led to the discovery of 2,4-diarylimidazoles and 2,4-bis(benzyloxy)-5-arylpyrimidines as two types of novel HSP90 N-terminal inhibitors. 16l and 22k exhibited antiproliferative activity against multiple breast cancer cell lines with IC50 values at the low micromolar level. 16l and 22k induced significant degradation of the client proteins AKT and ERK and a lower level of the heat shock response in comparison with tanespimycin (17-AAG). 22k exhibited a strong affinity for the HSP90α N-terminus with an IC50 value of 0.21 μM. A molecular docking study revealed that 16l and 22k successfully bind to the geldanamycin binding site at the N-terminus of HSP90α.
Keywords: HSP90, 2 4-diarylimidazoles, 2 4-bis(benzyloxy)-5-arylpyrimidines, molecular docking, anticancer
Introduction
Because proteins play roles in nearly every cellular process, it is essential to maintain protein homeostasis to preserve normal cell functions. Molecular chaperones are a large family of proteins that guard cellular protein homeostasis by regulating the conformation and quality of client proteins1,2. Heat shock protein 90 (HSP90) is one of the most crucial molecular chaperones in eukaryotes and stabilises and activates more than 400 client proteins3,4. Because cancer cells require higher levels of proteins for survival than normal cells, HSP90 is overexpressed in cancer cells, accounting for 4–6% of the whole proteome5,6. In addition, conformations of normal HSP90 and HSP90 of the cancer phenotype are different, and the latter is more susceptible to inhibitors7. Inhibition of HSP90 in cancer cells results in the degradation of client oncoproteins via the ubiquitin-proteasome pathway and the subsequent disruption of multiple signal transduction pathways, further leading to the apoptosis of cancer cells8,9. Therefore, HSP90 is a promising therapeutic target for discovering anticancer drugs10. Beyond cancer, HSP90 has also emerged as a potential drug target in other protein-related diseases, such as neurodegenerative diseases, infectious diseases, and ageing11–14.
HSP90 consists of three domains: the N-terminus, C-terminus, and the middle domain15,16. Classical HSP90 inhibitors competitively bind to the ATP binding pocket at the N-terminus. Over twenty HSP90 N-terminal inhibitors have entered clinical trials for the treatment of a variety of cancers17,18. Allosteric binding sites are also found at the C-terminus and the middle domain. HSP90 C-terminal inhibitors have been extensively studied in recent years because they do not cause a rescue cascade known as the heat shock response, which is often observed in the modulation of HSP90 with N-terminal inhibitors19,20. Many natural products and synthetic small molecules have been identified as HSP90 C-terminal inhibitors; however, they have not yet entered clinical trials for cancer therapy21.
Although some clinical investigations of HSP90 N-terminal inhibitors have stopped or terminated because of drug resistance and/or organ toxicity, there are still a considerable number of active inhibitors in clinical trials22,23. According to the results obtained from ClinicalTrial.gov (https://www.clinicaltrials.gov/, 2022/06/17), 17 studies are ongoing or in preparation (including recruiting, enrolling by invitation, and active/not recruiting). Some representative examples (1–7) currently being evaluated in clinical trials are shown in Figure 1. It should be noted that combination therapy of HSP90 N-terminal inhibitors with other anticancer drugs represents an effective strategy to combat cancer in clinical trials at present24. Combination with HSP90 inhibitors would help to prevent the chemotherapeutic resistance of classical anticancer drugs and/or potentiate the cytotoxic effects25,26. In this context, it is still desirable to develop more HSP90 N-terminal inhibitors as novel anticancer agents.
Figure 1.
Representative HSP90 N-terminal inhibitors in clinical trials.
We previously reported that a series of 1,3-dibenzyl-2-aryl imidazolidines 8 served as HSP90 N-terminal inhibitors27 (Figure 2). These inhibitors showed a strong affinity for the HSP90 N-terminus according to the fluorescence polarisation (FP) assay; however, they exhibited weak antiproliferative activity against cancer cells such as MCF-7 and A549. Weak anticancer efficacy may be attributed to the physicochemical instability of the imidazolidine ring because 8 is converted into the corresponding benzaldehyde and N, N’-dibenzyl ethylenediamine in an aqueous medium28–30. Therefore, we hypothesised that replacing imidazolidine with stable aromatic scaffolds would lead to the discovery of novel HSP90 N-terminal inhibitors with stronger anticancer activity. Therefore, the nonaromatic imidazolidine ring was replaced by the aromatic imidazole ring and pyrimidine ring, which are frequently used in clinical drugs31,32. As shown in Figure 2, trisubstituted imidazole 9 and pyrimidine 10 were designed to develop novel HSP90 inhibitors. Compound 9 bears a benzyl group at the N1 position and two phenyl groups at the C2 and C4 positions of the imidazole ring. In the case of 10, it has two benzyloxy groups at the C2 and C4 positions and a phenyl group at the C5 position of the pyrimidine ring.
Figure 2.
Design strategy illustration.
Materials and methods
Chemistry
General method for chemistry
All chemical reagents were commercially purchased and used without further purification. Reactions were monitored by thin layer chromatography on GF254 TLC plates. Column chromatography purification was performed on silica gel (200–300 mesh). Nuclear magnetic resonance (NMR) data were collected on an AVANCE III HD 500 MHz nuclear magnetic resonance spectrometer (Bruker, Billerica, MA, USA). HRMS detection of 16 and 22 were carried out on a Q Exactive mass spectrometer (Thermo Fisher, Waltham, MA USA) with electrospray ionisation (ESI) as the ionisation source. Mass spectra of 19 and 20 were recorded by a UPLC–ESI–MS/MS system. The UPLC–ESI–MS/MS system consists of an Acquity UPLC system (Waters Corp., MA, USA) coupled with a QTRAP 6500 Plus mass spectrometer (Sciex, Toronto, Canada) equipped with a TurboIonSpray source. Analyst Software 1.6.3 was used for data acquisition and data processing.
Preparation of 1-benzyl-4-bromo-1H-imidazoles (13)
13 was synthesised using a modified reported method33. To a solution of 4-bromo-1H-imidazole (11, 8.00 g, 54.40 mmol) in acetone (70 ml), benzyl bromide (12, 7.20 ml, 60.40 mmol) and potassium carbonate (8.28 g, 60.00 mmol) were added. The reaction mixture was stirred at room temperature for 22 h. Potassium carbonate was filtered, and acetone was removed under a vacuum. Water and ethyl acetate were added to extract the product. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was dissolved in a small amount of dichloromethane and then precipitated by the addition of petroleum ether. The solid was filtered to afford 13 as a white solid (5.71 g, 44.07%).
General synthetic procedure for 1-benzyl-4-aryl-1H-imidazoles (14)
14 was synthesised using a modified procedure34. To a solution of 1-benzyl-4-bromo-1H-imidazoles (13, 2.38 g, 10 mmol) in 1,4-dioxane (18 ml), aryl boronic acid (2.5 equiv.), Pd(dppf)Cl2 (5 mol%) and aqueous Na2CO3 solution (2.12 g in 6 ml water, 20 mmol) were added. The reaction mixture was heated to reflux and stirred under a nitrogen atmosphere for 4 h. After cooling to room temperature, the reaction mixture was extracted with water and ethyl acetate. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was purified by column chromatography using petroleum ether/ethyl acetate as the eluent.
General synthetic procedure for 1-benzyl-2-bromo-4-aryl-1H-imidazoles (15)
15 was prepared using a modified reported method35. To a solution of 1-benzyl-4-aryl-1H-imidazoles (14) in CCl4 (2.0 M), N-bromosuccinimide (NBS, 2 equiv.) was added. The reaction mixture was stirred at 35 °C for 14 h. The reaction mixture was extracted with dichloromethane and water. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was purified by column chromatography using petroleum ether/ethyl acetate as the eluent.
General synthetic procedure for 1-benzyl-2,4-diaryl-1H-imidazoles (16)
16 was prepared using a procedure similar to that for 14. To a solution of 1-benzyl-2-bromo-4-aryl-1H-imidazoles (15) in 1,4-dioxane, aryl boronic acid (2.5 equiv.), Pd(dppf)Cl2 (5 mol%) and aqueous Na2CO3 solution (3.3 M, 2.5 equiv.) was added. The reaction mixture was heated to reflux and stirred under a nitrogen atmosphere for 4 h. After cooling to room temperature, the reaction mixture was extracted with water and ethyl acetate. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was purified by column chromatography using petroleum ether/ethyl acetate as the eluent.
1-Benzyl-2,4-diphenyl-1H-imidazole (16a)
White solid, yield 29.13%. 1H NMR (500 MHz, Chloroform-d) δ 7.65 (s, 1H), 7.51–7.49 (m, 2H), 7.43–7.35 (m, 3H), 7.28–7.18 (m, 7H), 7.14–7.11 (m, 1H), 6.97 (d, J = 7.0 Hz, 2H), 4.97 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 138.32, 137.12, 136.59, 134.54, 130.96, 130.56, 128.92, 128.82, 128.79, 128.71, 128.12, 127.91, 126.93, 126.52, 126.33, 48.77. HRMS (ESI, m/z) calcd for C22H19N2 [M + H]+ 311.1548, found 311.1560.
1-Benzyl-4-phenyl-2-(m-tolyl)-1H-imidazole (16b)
White solid, yield 35.28%. 1H NMR (500 MHz, Chloroform-d) δ 7.64 (d, J = 3.0 Hz, 1H), 7.54–7.49 (m, 2H), 7.28–7.25 (m, 4H), 7.23–7.17 (m, 3H), 7.23–7.10 (m, 4H), 7.03 (d, J = 8.0 Hz, 1H), 6.98 (s, 3H), 4.94 (s, 2H), 2.29 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 138.57, 137.97, 136.95, 136.62, 134.48, 131.58, 130.35, 129.51, 129.03, 128.76, 128.12, 127.90, 127.05, 126.45, 126.31, 48.85, 21.33. HRMS (ESI, m/z) calcd for C23H21N2 [M + H]+ 325.1705, found 325.1715.
1-Benzyl-2–(3-chlorophenyl)-4-phenyl-1H-imidazole (16c)
White solid, yield 32.46%. 1H NMR (500 MHz, Chloroform-d) δ 7.68 (s, 1H), 7.49–7.45 (m, 2H), 7.38–7.36 (m, 1H), 7.36–7.25 (m, 4H), 7.25–7.19 (m, 2H), 7.17–7.16 (m, 2H), 7.09–7.06 (m, 1H), 6.98–6.95 (m, 2H), 4.97 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 138.90, 137.46, 136.14, 134.68, 133.97, 132.34, 130.88, 130.15, 129.28, 128.95, 128.89, 128.24, 128.12, 127.28, 126.96, 126.70, 49.09. HRMS (ESI, m/z) calcd for C22H18ClN2 [M + H]+ 345.1159; found 345.1172.
1-Benzyl-4-phenyl-2-(p-tolyl)-1H-imidazole (16d)
White solid, yield 41.00%. 1H NMR (500 MHz, Chloroform-d) δ 7.62 (s, 1H), 7.51 (d, J = 7.5 Hz, 2H), 7.29–7.25 (m, 3H), 7.22–7.09 (m, 7H), 7.02–6.97 (m, 2H), 4.95 (s, 2H), 2.40 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 138.61, 138.10, 136.98, 136.74, 134.67, 130.79, 129.67, 128.91, 128.78, 128.10, 127.86, 127.42, 126.91, 126.46, 126.24, 48.63, 21.38. HRMS (ESI, m/z) calcd for C23H21N2 [M + H]+ 325.1705, found 325.1714.
1–(4-(1-Benzyl-4-phenyl-1H-imidazol-2-yl)phenyl)ethan-1-one (16e)
White solid, yield 64.45%. 1H NMR (500 MHz, Chloroform-d) δ 7.97–7.91 (m, 2H), 7.69 (s, 1H), 7.48–7.43 (m, 2H), 7.33–7.26 (m, 5H), 7.24–7.15 (m, 3H), 7.00–6.93 (m, 2H), 5.01 (s, 2H), 2.62 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 197.56, 139.35, 137.92, 136.88, 136.26, 135.51, 134.10, 131.06, 128.92, 128.80, 128.27, 128.10, 127.57, 126.84, 126.76, 48.99, 26.68. HRMS (ESI, m/z) calcd for C24H21N2O [M + H]+ 353.1654, found 353.1661.
1-Benzyl-2–(4-chlorophenyl)-4-phenyl-1H-imidazole (16f)
White solid, yield 34.40%. 1H NMR (500 MHz, Chloroform-d) δ 7.67 (s, 1H), 7.50–7.45 (m, 2H), 7.35–7.32 (m, 2H), 7.29–7.26 (m, 3H), 7.22–7.20 (m, 2H), 7.17–7.11 (m, 3H), 6.97–6.95 (m, 2H), 4.97 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 138.86, 137.50, 136.38, 134.85, 134.22, 132.26, 129.23, 129.02, 128.88, 128.23, 128.03, 127.42, 126.77, 126.59, 48.84. HRMS (ESI, m/z) calcd for C22H18ClN2 [M + H]+ 345.1159, found 345.1171.
4–(1-Benzyl-4-phenyl-1H-imidazol-2-yl)phenol (16g)
White solid, yield 37.00%. 1H NMR (500 MHz, DMSO-d6) δ 9.73 (s, 1H), 7.90 (s, 1H), 7.45–7.40 (m, 2H), 7.30–7.22 (m, 3H), 7.18 (t, J = 7.5 Hz, 2H), 7.09 (t, J = 7.3 Hz, 1H), 7.00 (d, J = 8.5 Hz, 2H), 6.93 (t, J = 7.5 Hz, 2H), 6.80 (t, J = 8.5 Hz, 2H), 5.02 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 158.17, 138.01, 137.88, 137.13, 135.53, 132.45, 128.96, 128.47, 127.90, 127.27, 126.29, 126.14, 120.93, 116.30, 48.10. HRMS (ESI, m/z) calcd for C22H19N2O [M + H]+ 327.1497, found 327.1493.
1-Benzyl-2-(furan-2-yl)-4-phenyl-1H-imidazole (16h)
White solid, yield 19.07%. 1H NMR (500 MHz, Chloroform-d) δ 7.64 (s, 1H), 7.58 (d, J = 7.5 Hz, 2H), 7.53 (d, J = 1.8 Hz, 1H), 7.34–7.19 (m, 6H), 7.06 (d, J = 7.0 Hz, 2H), 6.46–6.44 (m, 1H), 6.32 (d, J = 3.0 Hz, 1H), 5.08 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 143.45, 143.24, 141.88, 138.10, 136.28, 134.07, 128.82, 128.24, 127.06, 127.04, 126.75, 118.85, 112.47, 111.29, 49.36. HRMS (ESI, m/z) calcd for C20H17N2O [M + H]+ 301.1341, found 301.1349.
1-Benzyl-4-phenyl-2-(thiophen-2-yl)-1H-imidazole (16i)
White solid, yield 16.80%. 1H NMR (500 MHz, Chloroform-d) δ 7.67 (s, 1H), 7.60–7.54 (m, 2H), 7.45 (dd, J = 5.0, 1.5 Hz, 1H), 7.31–7.22 (m, 2H), 7.21–7.15 (m, 1H), 7.07 (dd, J = 5.0, 3.5 Hz, 1H), 7.05–7.00 (m, 2H), 6.93 (dd, J = 3.5, 1.1 Hz, 1H), 5.02 (s, 2H) . 13C NMR (126 MHz, DMSO-d6) δ 136.10, 133.17, 131.78, 129.38, 125.75, 125.59, 124.10, 123.88, 123.45, 123.24, 122.87, 122.19, 122.02, 121.85, 116.09, 44.07. HRMS (ESI, m/z) calcd for C20H17N2S [M + H]+ 317.1112, found 317.1123.
1-Benzyl-2–(4-chlorophenyl)-4-(p-tolyl)-1H-imidazole (16j)
White solid, yield 47.54%. 1H NMR (500 MHz, Chloroform-d) δ 7.67 (s, 1H), 7.36–7.32 (m, 4H), 7.29–7.25 (m, 3H), 7.12 (d, J = 8.5 Hz, 2H), 7.03 (d, J = 8.0 Hz, 2H), 7.00–6.94 (m, 2H), 4.96 (s, 2H), 2.29 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 138.77, 137.34, 136.34, 136.32, 134.81, 132.29, 131.15, 129.22, 129.02, 128.98, 128.89, 128.05, 127.00, 126.82, 126.55, 48.90, 21.14. HRMS (ESI, m/z) calcd for C23H20ClN2 [M + H]+ 359.1315, found 359.1327.
1-Benzyl-2–(4-chlorophenyl)-4-(m-tolyl)-1H-imidazole (16k)
White solid, yield 23.00%. 1H NMR (500 MHz, Chloroform-d) δ 7.66 (s, 1H), 7.48 (s, 1H), 7.36–7.26 (m, 5H), 7.15–7.03 (m, 4H), 7.00–6.93 (m, 3H), 4.96 (s, 2H), 2.28 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 138.95, 137.93, 137.44, 136.41, 134.82, 134.07, 132.29, 129.19, 129.08, 128.89, 128.04, 128.02, 127.47, 127.40, 126.78, 123.63, 48.86, 21.46. HRMS (ESI, m/z) calcd for C23H20ClN2 [M + H]+ 359.1315, found 359.1324.
1-Benzyl-2,4-bis(4-chlorophenyl)-1H-imidazole (16l)
White solid, yield 13.37%. 1H NMR (500 MHz, Chloroform-d) δ 7.65 (s, 1H), 7.45–7.37 (m, 2H), 7.36–7.31 (m, 2H), 7.30–7.26 (m, 3H), 7.21–7.14 (m, 2H), 7.13–7.07 (m, 2H), 6.99–6.91 (m, 2H), 4.95 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 137.87, 137.62, 136.23, 135.13, 132.81, 132.30, 132.18, 129.40, 128.94, 128.70, 128.43, 128.13, 127.84, 127.69, 126.82, 48.92. HRMS (ESI, m/z) calcd for C22H16Cl2N2 [M + H]+ 379.0769, found 379.0783.
1-Benzyl-4–(3-chlorophenyl)-2–(4-chlorophenyl)-1H-imidazole (16m)
White solid, yield 56.18%. 1H NMR (500 MHz, Chloroform-d) δ 7.67 (s, 1H), 7.58–7.57 (m, 1H), 7.37–7.34 (m, 2H), 7.30–7.26 (m, 3H), 7.22–7.20 (m, 1H), 7.13–7.10 (m, 4H), 6.96–6.93 (m, 2H), 4.96 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 137.62, 137.51, 136.09, 136.00, 135.20, 134.27, 132.13, 129.40, 129.37, 128.93, 128.43, 128.14, 128.09, 126.79, 126.69, 126.60, 124.50, 48.93. HRMS (ESI, m/z) calcd for C22H17Cl2N2 [M + H]+ 379.0769, found 379.0786.
1-Benzyl-2-phenyl-4-(p-tolyl)-1H-imidazole (16n)
White solid, yield 28.10%. 1H NMR (500 MHz, Chloroform-d) δ 7.64 (s, 1H), 7.38–7.35 (m, 5H), 7.30–7.26 (m, 3H), 7.25–7.18 (m, 2H), 7.01 (d, J = 8.0 Hz, 2H), 6.98–6.94 (m, 2H), 4.96 (s, 2H), 2.27 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 138.20, 136.93, 136.50, 136.05, 131.41, 130.99, 130.52, 128.89, 128.79, 128.68, 128.40, 127.93, 126.98, 126.50, 48.84, 21.13. HRMS (ESI, m/z) calcd for C23H21N2 [M + H]+ 325.1705, found 325.1711.
1-Benzyl-4-phenyl-2-(thiophen-2-yl)-1H-imidazole (16o)
White solid, yield 35.22%. 1H NMR (500 MHz, Chloroform-d) δ 7.60 (s, 1H), 7.45–7.40 (m, 3H), 7.27–7.25 (m, 5H), 7.08 (d, J = 5.0 Hz, 1H), 6.96–6.94 (m, 2H), 6.86–6.83 (m, 2H), 4.94 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 138.20, 137.00, 136.35, 134.15, 131.14, 129.63, 129.15, 128.95, 128.82, 128.01, 127.78, 127.17, 126.95, 123.32, 122.25, 48.92. HRMS (ESI, m/z) calcd for C20H17N2S [M + H]+, 317.1112, found 317.1125.
1-Benzyl-4–(4-chlorophenyl)-2-phenyl-1H-imidazole (16p)
White solid, yield 27.83%. 1H NMR (500 MHz, Chloroform-d) δ 7.67(s, 1H), 7.43–7.33 (m, 5H), 7.29–7.27 (m, 3H), 7.22–7.12 (m, 4H), 6.99–6.93 (m, 2H), 4.96 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 137.16, 137.10, 136.24, 133.97, 132.76, 132.15, 130.84, 130.00, 129.22, 129.04, 128.97, 128.85, 128.27, 128.04, 128.00, 127.33, 126.98, 48.93. HRMS (ESI, m/z) calcd for C22H18ClN2 [M + H]+ 345.1159, found 345.1172.
Preparation of 2,4-bis(4-chlorophenyl)-1H-imidazole (19)
19 was prepared using a modified procedure previously reported by the Li group36. A mixture of 4-chlorobenzene-1-carboximidamide hydrochloride (2.61 g, 20 mmol), THF (36 ml) and H2O (9 ml) was stirred and heated to 70 °C. α-Bromo-4-chloroacetophenone (4.20 g, 18 mmol) in THF (11 ml) was slowly added, and the reaction mixture was then stirred at 70 °C for 8 h. THF was removed under vacuum. The resulting mixture was dissolved in water and dichloromethane (30 ml/30 ml), and the slow addition of concentrated HCl led to the precipitation of the product, which was further filtered and washed with dichloromethane to afford 19 as a light yellow solid (1.12 g, 21.6%).
1H NMR (500 MHz, DMSO-d6) δ 12.81 (s, 1H), 8.07–7.99 (m, 2H), 7.91–7.85 (m, 2H), 7.82 (s, 1H), 7.58–7.52 (m, 2H), 7.47–7.42 (m, 2H). 13C NMR (126 MHz, DMSO-d6) δ 144.89, 140.00, 133.33, 132.64, 130.45, 129.18, 128.75, 128.39, 126.51, 125.94, 115.15. LC-MS (ESI, m/z) 288.8 [M + H]+.
General synthetic procedure for 1-substituted 2,4-bis(4-chlorophenyl)-1H-imidazoles (20)
Excess aliphatic halide and K2CO3 were added to a solution of 2,4-bis(4-chlorophenyl)-1H-imidazole (19) in the indicated solvent. The reaction mixture was stirred at the indicated temperature. The reaction mixture was extracted with ethyl acetate and water. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was purified by column chromatography using petroleum ether/ethyl acetate as the eluent.
1-Allyl-2,4-bis(4-chlorophenyl)-1H-imidazole (20a)
20a was prepared from 19 (0.58 g, 2 mmol) and allyl bromide (6.92 ml, 20 mmol) in acetonitrile at 80 °C. White solid (79 mg, 11.9%). 1H NMR (500 MHz, chloroform-d) δ 7.76 (d, J = 8.0 Hz, 2H), 7.59 (d, J = 8.0 Hz, 2H), 7.44 (d, J = 8.0 Hz, 2H), 7.34 (d, J = 8.0 Hz, 2H), 7.27 (s, 1H), 6.03–6.01 (m, 1H), 5.35 (d, J = 10.5 Hz, 1H), 5.19 (d, J = 17.0 Hz, 1H), 4.61 (s, 2H). 13C NMR (126 MHz, chloroform-d) δ 147.12, 140.43, 135.26, 132.97, 132.48, 132.40, 130.11, 128.91, 128.73, 126.17, 118.33, 117.06, 49.37, 29.71. LC-MS (ESI, m/z) 328.8 [M + H]+.
2,4-Bis(4-chlorophenyl)-1-pentyl-1H-imidazole (20b)
20b was prepared from 19 (0.29 g, 1 mmol) and 1-pentyl bromide (0.17 g, 1.1 mmol) in acetone at 60 °C. White solid (30 mg, 8.36%). 1H NMR (500 MHz, chloroform-d) δ 7.75 (d, J = 8.0 Hz, 2H), 7.56 (d, J = 8.0 Hz, 2H), 7.45 (d, J = 8.0 Hz, 2H), 7.33 (d, J = 8.5 Hz, 2H), 7.28 (s, 1H), 3.96 (t, J = 7.5 Hz, 2H), 1.77 (t, J = 7.5 Hz, 2H), 1.30–1.25 (m, 4H), 0.87 (t, J = 7.0 Hz, 3H). 13C NMR (126 MHz, chloroform-d) δ 147.04, 140.20, 135.12, 132.37, 130.32, 128.93, 128.71, 126.11, 116.47, 47.08, 30.80, 28.62, 22.14, 13.84. LC-MS (ESI, m/z) 359.2 [M + H]+.
Methyl 2–(2,4-bis(4-chlorophenyl)-1H-imidazol-1-yl)acetate (20c)
20c was prepared from 19 (0.58 g, 2 mmol) and methyl bromoacetate (0.46 g, 3 mmol) in acetonitrile at 80 °C. White solid (420 mg, 58.2%). 1H NMR (500 MHz, chloroform-d) δ 7.77–7.74 (m, 2H), 7.54–7.45 (m, 4H), 7.35–7.26 (m, 3H), 4.71 (s, 2H), 3.81 (s, 3H). 13C NMR (126 MHz, chloroform-d) δ 168.07, 147.72, 140.79, 135.63, 132.64, 132.16, 130.29, 129.12, 128.72, 128.17, 126.28, 117.62, 53.02, 48.22. LC-MS (ESI, m/z) 360.8 [M + H]+.
2–(2,4-Bis(4-chlorophenyl)-1H-imidazol-1-yl)acetamide (20d)
20d was prepared from 19 (0.58 g, 2 mmol) and bromoacetamide (0.414 g, 3 mmol) in DMF at 80 °C. White solid (120 mg, 17.3%). 1H NMR (500 MHz, DMSO-d6) δ 7.83–7.79 (m, 3H), 7.71 (s, 1H), 7.65 (d, J = 8.5 Hz, 2H), 7.57 (d, J = 8.5 Hz, 2H), 7.43 (d, J = 8.5 Hz, 2H), 7.39 (s, 1H), 4.71 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 168.52, 146.37, 138.18, 133.45, 133.02, 130.56, 129.92, 129.07, 128.55, 128.46, 125.79, 120.41, 49.05. LC-MS (ESI, m/z) 345.9 [M + H]+.
3–(2,4-Bis(4-chlorophenyl)-1H-imidazol-1-yl)propan-1-ol (20e)
20e was prepared from 19 (0.29 g, 1 mmol) and 3-bromo-1-propanol (132 μL, 1.1 mmol) in acetonitrile at 80 °C. White solid (143 mg, 41.2%). 1H NMR (500 MHz, chloroform-d) δ 7.76–7.73 (m, 2H), 7.60–7.57 (m, 2H), 7.46–7.43 (m, 2H), 7.35–7.30 (m, 3H), 7.27 (s, 1H), 4.16 (t, J = 7.0 Hz, 2H), 3.62 (t, J = 5.5 Hz, 2H), 1.97 (t, J = 6.0 Hz, 2H). 13C NMR (126 MHz, chloroform-d) δ 147.12, 140.29, 135.25, 132.50, 132.35, 130.28, 129.00, 128.88, 128.76, 126.15, 116.84, 58.82, 43.68, 33.30. LC-MS (ESI, m/z) 347.3 [M + H]+.
General synthetic procedure for 2,4-bis(benzyloxy)-5-arylpyrimidines (22)
2,4-Bis(benzyloxy)-5-bromopyrimidine 21 is commercially available and was used without further purification. 22 was prepared using a procedure similar to that for 14. To a solution of 21 in 1,4-dioxane (9 ml), aryl boronic acid (2.5 equiv.), Pd(dppf)Cl2 (5 mol%) and aqueous Na2CO3 solution (3.3 M, 2.0 equiv.) was added. The reaction mixture was heated to reflux and stirred under a nitrogen atmosphere for 4 h. After cooling to room temperature, the reaction mixture was extracted with water and ethyl acetate. The organic layer was collected, washed with water and brine, dried over anhydrous magnesium sulphate, and condensed under a vacuum. The resulting crude product was purified by column chromatography using petroleum ether/ethyl acetate as the eluent.
2,4-Bis(benzyloxy)-5-phenylpyrimidine (22a)
White solid, yield 38.55%. 1H NMR (500 MHz, Chloroform-d) δ 8.31 (s, 1H), 7.53–7.50 (m, 4H), 7.44–7.27 (m, 11H), 5.50 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.45, 163.83, 158.11, 136.62, 136.23, 133.22, 128.81, 128.49, 128.47, 128.43, 128.08, 128.02, 127.95, 127.64, 127.54, 116.33, 69.23, 68.34. HRMS (ESI, m/z) calcd for C24H21N2O2 [M + H]+ 369.1603, found 369.1615.
2,4-Bis(benzyloxy)-5-(o-tolyl)pyrimidine (22b)
White solid, yield 42.02%. 1H NMR (500 MHz, Chloroform-d) δ 8.13 (s, 1H), 7.51 (d, J = 7.5 Hz, 2H), 7.39 (t, J = 8.0 Hz, 2H), 7.36–7.20 (m, 9H), 7.15 (d, J = 7.5 Hz, 1H), 5.46 (s, 2H), 5.45 (s, 2H), 2.14 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 167.74, 164.14, 158.66, 137.33, 136.64, 136.26, 132.99, 130.41, 130.05, 128.50, 128.43, 128.20, 128.15, 128.06, 127.92, 127.60, 125.76, 116.56, 69.26, 68.13, 20.02. HRMS (ESI, m/z) calcd for C25H22N2O2 [M + H]+ 383.1760, found 383.1773.
2,4-Bis(benzyloxy)-5-(m-tolyl)pyrimidine (22c)
White solid, yield 39.02%. 1H NMR (500 MHz, Chloroform-d) δ 8.31 (s, 1H), 7.50 (d, J = 7.5 Hz, 2H), 7.41–7.28 (m, 11H), 7.16 (d, J = 7.0 Hz, 1H), 5.49 (s, 2H), 5.46 (s, 2H), 2.38 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 167.48, 163.79, 158.04, 138.03, 136.67, 136.32, 133.12, 129.59, 128.49, 128.35, 128.11, 128.04, 127.94, 127.52, 125.92, 116.43, 69.23, 68.32, 21.49. HRMS (ESI, m/z) calcd for C25H23N2O2 [M + H]+ 383.1760; found 383.1771.
2,4-Bis(benzyloxy)-5-(p-tolyl)pyrimidine (22d)
White solid, yield 43.70%. 1H NMR (500 MHz, Chloroform-d) δ 8.30 (s, 1H), 7.49 (d, J = 7.5 Hz, 2H), 7.44–7.28 (m, 10H), 7.27–7.19 (m, 2H), 5.49 (s, 2H), 5.45 (s, 2H), 2.38 (s, 3H). 13C NMR (126 MHz, Chloroform-d) δ 167.44, 163.66, 157.90, 137.48, 136.67, 136.30, 129.17, 128.64, 128.48, 128.46, 128.06, 127.57, 116.28, 69.18, 68.31, 21.20. HRMS (ESI, m/z) calcd for C25H23N2O2 [M + H]+ 383.1760, found 383.1771.
2,4-Bis(benzyloxy)-5–(4-fluorophenyl)pyrimidine (22e)
White solid, yield 34.33%. 1H NMR (500 MHz, Chloroform-d) δ 8.28 (s, 1H), 7.49–7.42 (m, 4H), 7.36 (m, 10H), 7.12–7.07 (m, 2H), 5.49 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.37, 163.87, 163.35, 161.39, 157.93, 136.57, 136.09, 130.49, 128.55, 128.48, 128.07, 128.06, 127.58, 115.52, 115.44, 115.35, 69.27, 68.45. HRMS (ESI, m/z) calcd for C24H20FN2O2 [M + H]+ 387.1509, found 387.1519.
2,4-Bis(benzyloxy)-5–(4-chlorophenyl)pyrimidine (22f)
White solid, yield 18.47%. 1H NMR (500 MHz, Chloroform-d) δ 8.29 (s, 1H), 7.49 (d, J = 7.5 Hz, 2H), 7.45 (d, J = 7.5 Hz, 2H), 7.41–7.25 (m, 10H), 5.48 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.34, 163.99, 157.98, 136.51, 136.00, 133.66, 131.67, 130.03, 128.65, 128.57, 128.49, 128.10, 128.07, 127.62, 115.23, 69.31, 68.52. HRMS (ESI, m/z) calcd for C24H20ClN2O2 [M + H]+ 403.1213, found 403.1230.
2,4-Bis(benzyloxy)-5–(2-chlorophenyl)pyrimidine (22g)
A colourless liquid, yield 23.55%. 1H NMR (500 MHz, Chloroform-d) δ 8.18 (s, 1H), 7.51–7.46 (m, 2H), 7.46–7.39 (m, 1H), 7.38–7.32 (m, 2H), 7.32–7.19 (m, 9H), 5.45 (s, 2H), 5.43 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.82, 164.56, 158.96, 136.64, 136.24, 134.40, 132.59, 131.94, 129.74, 129.55, 128.59, 128.49, 128.25, 128.17, 128.00, 127.68, 114.66, 69.45, 68.46. HRMS (ESI, m/z) calcd for C24H20ClN2O2 [M + H]+ 403.1213, found 403.1227.
2,4-Bis(benzyloxy)-5–(3-chlorophenyl)pyrimidine (22h)
White solid, yield 20.73%. 1H NMR (500 MHz, Chloroform-d) δ 8.31 (s, 1H), 7.54 (s, 1H), 7.52–7.47 (m, 2H), 7.41–7.30 (m, 11H), 5.50 (s, 2H), 5.47 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.36, 164.12, 158.13, 136.49, 135.99, 135.02, 134.29, 129.67, 128.90, 128.57, 128.50, 128.10, 127.71, 127.55, 126.86, 115.08, 69.36, 68.51. HRMS (ESI, m/z) calcd for C24H20ClN2O2 [M + H]+ 403.1213, found 403.1226.
2,4-Bis(benzyloxy)-5–(2,4-dichlorophenyl)pyrimidine (22i)
White solid, yield 9.56%. 1H NMR (500 MHz, Chloroform-d) δ 8.17 (s, 1H), 7.53–7.47 (m, 3H), 7.41–7.35 (m, 2H), 7.34–7.25 (m, 7H), 7.34–7.25 (m, 7H), 7.21 (d, J = 8.0 Hz, 1H), 5.46 (s, 2H), 5.44 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.64, 164.58, 158.86, 136.41, 135.93, 135.08, 134.65, 132.55, 131.08, 129.57, 128.50, 128.45, 128.16, 128.11, 128.03, 127.68, 127.11, 113.49, 69.42, 68.51. HRMS (ESI, m/z) calcd for C24H19Cl2N2O2 [M + H]+ 437.0824, found 437.0842.
(4–(2,4-bis(benzyloxy)pyrimidin-5-yl)phenyl)methanol (22j)
White solid, yield 44.04%. 1H NMR (500 MHz, Chloroform-d) δ 8.28 (s, 1H), 7.53–7.47 (m, 4H), 7.43–7.27 (m, 10H), 5.49 (s, 2H), 5.46 (s, 2H), 4.72 (d, J = 3.5 Hz, 2H), 1.84 (s, 1H). 13C NMR (126 MHz, DMSO-d6) δ 162.72, 159.08, 153.30, 135.64, 131.86, 131.42, 127.80, 124.21, 123.79, 123.75, 123.28, 122.88, 122.34, 111.27, 64.52, 63.70, 60.29. HRMS (ESI, m/z) calcd for C25H23N2O3 [M + H]+ 399.1709, found 399.1722.
4–(2,4-Bis(benzyloxy)pyrimidin-5-yl)phenol (22k)
White solid, yield 22.00%. 1H NMR (500 MHz, DMSO-d6) δ 9.56 (s, 1H), 8.34 (s, 1H), 7.47–7.30 (m, 12H), 6.83–6.78 (m, 2H), 5.46 (s, 2H), 5.41 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 166.68, 162.71, 157.36, 156.97, 136.64, 136.25, 129.77, 128.35, 128.33, 127.91, 127.86, 127.82, 127.58, 126.89, 123.09, 115.60, 115.46, 115.14, 68.33, 67.73. HRMS (ESI, m/z) calcd for C24H21N2O3 [M + H]+ 385.1552, found 385.1564.
3–(2,4-Bis(benzyloxy)pyrimidin-5-yl)phenol (22l)
White solid, yield 18.40%. 1H NMR (500 MHz, Chloroform-d) δ 8.83 (s, 1H), 8.23 (s, 1H), 7.40–7.36 (m, 2H), 7.38–7.10 (m, 9H), 7.08 (s, 1H), 7.00 (d, J = 7.5 Hz, 1H), 6.89 (dd, J = 8.0, 2.5 Hz, 1H), 5.39 (s, 2H), 5.37 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 167.58, 163.54, 157.63, 156.90, 136.39, 136.13, 134.14, 129.73, 128.62, 128.59, 128.15, 128.10, 127.86, 127.54, 120.65, 116.39, 116.15, 115.47, 69.48, 68.65. HRMS (ESI, m/z) calcd for C24H21N2O3 [M + H]+ 385.1552, found, 385.1562.
2–(2,4-Bis(benzyloxy)pyrimidin-5-yl)phenol (22m)
White solid, yield 12.53%. 1H NMR (500 MHz, Chloroform-d) δ 8.30 (s, 1H), 7.52–7.47 (m, 2H), 7.40–7.27 (m, 9H), 7.19 (dd, J = 7.5, 1.5 Hz, 1H), 7.05–6.96 (m, 2H), 5.52 (s, 2H), 5.47 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 166.97, 164.29, 160.23, 153.74, 136.38, 135.48, 131.04, 129.92, 128.66, 128.52, 128.36, 128.13, 128.07, 127.91, 121.19, 120.66, 117.38, 112.81, 69.45, 68.87. HRMS (ESI, m/z) calcd for C24H20N2O3 [M + H]+ 385.1552, found 385.1563.
4–(2,4-Bis(benzyloxy)pyrimidin-5-yl)benzoic acid (22n)
White solid, yield 20.04%. 1H NMR (500 MHz, DMSO-d6) δ 12.97 (s, 1H), 8.51 (s, 1H), 7.97 (d, J = 8.5 Hz, 2H), 7.72 (d, J = 8.5 Hz, 2H), 7.51–7.46 (m, 2H), 7.46–7.29 (m, 8H), 5.49 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 166.90, 166.82, 163.60, 158.51, 137.23, 136.44, 135.99, 129.20, 128.70, 128.39, 128.36, 127.98, 127.68, 114.59, 68.59, 68.07. HRMS (ESI, m/z) calcd for C25H20N2O4 [M + H]+ 413.1501, found 413.1518.
2,4-Bis(benzyloxy)-5-(thiophen-2-yl)pyrimidine (22o)
Brownish solid, yield 15.86%. 1H NMR (500 MHz, Chloroform-d) δ 8.56 (s, 1H), 7.48 (t, J = 7.0 Hz, 4H), 7.43–7.30 (m, 8H), 7.09–7.04 (m, 1H), 5.55 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 166.07, 163.33, 156.38, 136.54, 135.89, 134.48, 128.59, 128.51, 128.22, 128.10, 128.03, 127.30, 125.46, 125.18, 110.61, 69.37, 69.02. HRMS (ESI, m/z) calcd for C22H19N2O2S [M + H]+ 375.1167, found 375.1181.
2,4-Bis(benzyloxy)-5-(furan-2-yl)pyrimidine (22p)
White solid, yield 47.78%. 1H NMR (500 MHz, Chloroform-d) δ 8.75 (s, 1H), 7.51–7.29 (m, 11H), 6.74 (d, J = 2.5 Hz, 1H), 6.43 (d, J = 1.0 Hz, 1H), 5.54 (s, 2H), 5.46 (s, 2H). 13C NMR (126 MHz, Chloroform-d) δ 165.51, 162.97, 154.50, 146.54, 141.70, 136.55, 135.93, 128.69, 128.48, 128.34, 128.12, 128.07, 111.68, 109.53, 107.59, 69.34, 68.99. HRMS (ESI, m/z) calcd for C22H18N2O3 [M + H]+ 359.1396, found 359.1409.
Biological evaluation
Cell culture
Three breast cancer cell lines, MCF-7, MDA-MB-231, and 4T1 were used in this study. The cells were purchased from the Institute of Basic Medicine, Chinese Academy of Medical Sciences (Beijing, China). MCF-7 and MDA-MB-23 cells were cultured in DMEM, and 4T1 cells were cultured in RPMI-1640 using 10% foetal bovine serum (FBS) and 1% penicillin-streptomycin solution (PS) in humidified air containing 5% CO2 at 37 °C.
Antiproliferative activity assay
The antiproliferative activity of the target compounds against the breast cancer cell lines MCF-7, MDA-MB-231, and 4T1 was determined using Cell Counting Kit-8 (Beyotime, Shanghai, China). The target compounds were dissolved in DMSO, diluted with cell culture medium to the desired concentration, and stored at −20 °C before use. Cells were seeded into a 96-well plate at a density of 7000 cells/well (200 μL/well) and incubated overnight. Compound solutions at different concentrations were added and incubated for 48 h. CCK8 solution was added and further incubated for 1–2 h in the dark according to the manufacturer’s instructions. Absorbance was measured at a wavelength of 450 nm using Synergy H1 (BioTek Instruments, Inc., Winooski, VT, USA). IC50 values were calculated by GraphPad Prism 8 (GraphPad Software, Inc., San Diego, CA, USA). The experiments were performed in triplicate.
Western blotting assay
Primary antibodies against HSP70, HSP90, and GAPDH were purchased from Abcam (Cambridge, MA, USA). Primary antibodies against ERK and AKT were purchased from Beyotime (Shanghai, China). HRP (horseradish peroxidase)-labelled anti-rabbit immunoglobulin G (H + L) secondary antibody was purchased from Abbkine (Redlands, CA, USA). MCF-7 cells were incubated in a 6-well plate at a density of 106 cells/well (4 ml/well) at 37 °C for 12 h. The cells were then treated with the compounds and 17-AAG at different concentrations at 37 °C for 24 h. The 6-well plate was placed on ice for 15 min. Cells were collected into a centrifuge tube and lysed by radioimmunoprecipitation (RIPA) lysis solution containing 1% phenylmethanesulphonyl fluoride (PMSF). The lysate was centrifuged at a speed of 14,000 r/min in a cryogenic high-speed centrifuge for 10 min. The cell supernatant was collected to determine the protein content. Cell lysates were mixed with SDS loading buffer and boiled for 20 min. Equal amounts of protein in cell lysates were separated by 10% sodium dodecyl sulphate–polyacrylamide gel electrophoresis (SDS–PAGE), which was prepared using the PAGE Gel Fast Preparation Kit (Epizyme, PG112, China). The protein on the separating gel was transferred to a polyvinylidene fluoride (PVDF) membrane. The membrane was blocked in skim milk for 80 min and incubated with primary antibody (1:4000) at 4 °C overnight. After further incubation with HRP anti-rabbit IgG (H + L) (1:5000) at room temperature for 1 h, the protein bands were detected with a gel imager (ProteinSimple, San Jose, CA, USA). The density of proteins was determined using AlphaView SA (Alpha Innotech Corp, version 3.4.0.0, San Leandro, CA, USA). The experiments were performed in triplicate.
Fluorescence polarisation (FP) assay
The binding affinity for HSP90α was evaluated using a commercially available Hsp90α N-terminal domain assay kit (Catalogue: #50293, BPS Bioscience, CA, USA). According to the manufacturer’s instructions, 5x HSP90 assay buffer (5 μL), 40 mM dithiothreitol (5 μL), 2 mg/mL BSA (5 μL), and H2O (40 μL) were sequentially added to each well. After the addition of diluted FITC-labelled geldanamycin solution (100 nM, 5 μL), compounds at different concentrations (10 μM, 1.0 μM, 0.1 μM, 0.01 μM) were added. The reaction was initiated by the addition of 20 μL of Hsp90α (17 ng/μL). After 2 h of incubation with slow shaking at room temperature, the fluorescent polarisation was measured using Synergy H1 (BioTek Instruments, Inc.). The excitation and emission wavelengths were 485 nm and 530 nm, respectively. IC50 values were calculated based on the signal changes in the FP competition assay. The experiments were performed in triplicate.
Molecular docking
The crystal structure of the human HSP90α N-terminus was downloaded from the Protein Data Bank (PDB ID: 1YET) 37. Molecular docking was performed using BIOVIA Discovery Studio 2016 (Dassault Systèmes, San Diego, USA). The protein structure was prepared using the “Prepare Protein” procedure before docking. In this procedure, multiple tasks, including building loops, protonation at pH 7.4, and removing water molecules, were performed. Meanwhile, ligands were prepared by the “Prepare Ligand” procedure. The CHARMM force field was used in the preparation of protein and ligand structures. The binding site was defined using the module “Define site-From PDB Site Records.” The coordinates (in XYZ) of the docking region centre were as follows: x = 40.72, y = −45.98, z = 64.47, and the radius of the active site sphere was 10.3. Docking simulations were performed using CDOCKER, a semiflexible docking program. All other docking parameters were set as default. The protein-ligand docking pose with the highest -CDOCKER_INTERACTION_ENERGY score was taken from the docking results and described.
Statistical analysis
All values obtained from the experiments are expressed as the means ± SDs. The IC50 values at which the concentration of inhibitor reduces cell viability or enzyme activity by 50% were evaluated by nonlinear regression using GraphPad Prism 8.0 software (GraphPad Software, Inc., La Jolla, USA).
Results and discussions
Chemical synthesis
Synthesis of 1-benzyl-2,4-diarylimidazoles 16
1-Benzyl-2,4-diarylimidazoles 16 were synthesised via a four-step synthetic route (Scheme 1). The benzylation of 4-bromoimidazole 11 with benzyl bromide 12 in the presence of K2CO3 provided 1-benzyl-3-bromoimidazole 13, which further coupled with aryl boronic acid to afford 14 through a Pd(dppf)Cl2-catalysed Suzuki cross-coupling reaction. The bromination of the imidazole ring of 14 with NBS in CCl4 gave 15, which was then converted into the final product 16 through a second Suzuki cross-coupling reaction. The final reaction step always gave low yields, which could be attributed to the debromination of 15. Simple condition screening experiments revealed that Pd(dppf)Cl2 was more effective than other palladium catalysts, such as Pd(PPh3)4 and Pd(PPh3)2Cl2, at this step.
Scheme 1.
Synthetic route for 1-benzyl-2, 4-diarylimidazoles 16. Reagents and conditions: (a) 4-bromo-1H-imidazole, benzyl bromide, K2CO3, acetone, r.t., 22 h; (b) Pd(dppf)Cl2, Na2CO3, 1,4-dioxane/H2O, N2, reflux, 4 h; (c) NBS, CCl4, 35 °C, 14 h; (d) Pd(dppf)Cl2, Na2CO3, 1,4-dioxane/H2O, N2, reflux, 4 h.
Synthesis of bis(4-chlorophenyl)imidazoles 20
To further investigate the effect of substituents at the N1 position of the imidazole ring, bis(4-chlorophenyl)imidazoles 20 bearing different functional groups at the N1 position were synthesised (Scheme 2). 19 was prepared by a single-step cyclisation reaction from 4-chlorobenzene-1-carboximidamide hydrochloride 17 and 2-chloro-1–(4-chlorophenyl)ethan-1-one 18 in the presence of NaHCO3 in THF/H2O. Treating 19 with different alkyl halides in the presence of K2CO3 afforded the N-alkylated product 20.
Scheme 2.
Synthetic route for 2,4-bis(4-chlorophenyl) imidazoles 20. Reagents and conditions: (a) NaHCO3, THF/H2O, 70 °C, 5 h; (b) alkyl halides, K2CO3.
Synthesis of 2,4-bis(benzyloxy)-5-arylpyrimidines
2,4-Bis(benzyloxy)-5-arylpyrimidines 22 were obtained through a Pd(dppf)Cl2-catalysed Suzuki cross-coupling reaction between 2,4-bis(benzyloxy)-5-bromopyrimidine 21 and a variety of (hetero)aryl boronic acids in the presence of Na2CO3 in 1,4-dioxane/H2O (Scheme 3).
Scheme 3.
Synthetic route for 2, 4-bis(benzyloxy)-5-arylpyrimidines 22. Reagents and conditions: (a) Pd(dppf)Cl2, Na2CO3, 1,4-dioxane/H2O, N2, reflux, 4 h.
Biological evaluation
Antiproliferative activity evaluation
Female breast cancer is the most commonly diagnosed cancer, with an estimated 2.3 million new cases, and the fifth leading cause of cancer mortality, with an estimated 685,000 new deaths around the world in 202038. HSP90 inhibitors have been extensively studied in clinical trials for the treatment of breast cancer39. Although none has been approved in clinical practice, HSP90 inhibitors have shown great promise in breast cancer therapy. In this study, the antiproliferative activity of the synthesised compounds was evaluated against human breast cancer cell lines MCF-7 and MDA-MB-231 and mouse breast cancer cell line 4T1 using a CCK-8 assay kit. As shown in Table 1, it is remarkable that 1-benzyl-2,4-diphenyl imidazole (16a) bearing no substituents on any phenyl ring showed antiproliferative activity against all three cancer cell lines. The effect of substituent R2 at the C2 position of the imidazole ring was subsequently studied. The methyl group at the meta-position of the phenyl ring slightly increased the activity; however, a chlorine atom led to the loss of activity. In the case of the para-position, functional groups such as methyl, chloro, and acetyl could preserve the activity, while the hydroxyl group was ineffective. Replacing the phenyl ring with heteroaromatic rings, such as furan and thiophene rings, decreased the activity. The subsequent investigation of substituent R1 at the C4 position of the imidazole ring revealed that chloro substitution on the phenyl ring was beneficial for increasing activity. Among these 1-benzyl-2,4-diarylimidazoles, 16j, 16l, and 16m exhibited strong antiproliferative activity with IC50 values in the low micromolar range against three breast cancer cell lines. 8a, the most potent compound among the imidazolidine-based HSP90 inhibitors, was evaluated for comparison. It exhibited weak antiproliferative activity against MCF-7 and 4T1 cells with IC50 values of 31.25 ± 0.31 μM and 42.10 ± 1.10 μM, respectively, and it was not cytotoxic towards MDA-MB-231 cells. The above results supported our hypothesis that replacing the imidazolidine ring in 8 with a stable aromatic ring could improve the anticancer activity.
Table 1.
Antiproliferative activity of 1-benzyl-2, 4-diaryl imidazoles 16.
| IC50 (μM) |
|||||
|---|---|---|---|---|---|
| Comp. | R1 | R2 | MCF-7 | MDA-MB-231 | 4T1 |
| 16a |
|
|
19.89 ± 2.76 | 20.31 ± 2.16 | 23.40 ± 0.28 |
| 16b |
|
|
14.56 ± 0.25 | 16.68 ± 0.80 | 10.54 ± 0.05 |
| 16c |
|
|
>50 | >50 | >50 |
| 16d |
|
|
15.29 ± 0.65 | 20.21 ± 0.67 | 11.69 ± 1.02 |
| 16e |
|
|
14.56 ± 0.92 | 23.77 ± 0.23 | 9.87 ± 0.79 |
| 16f |
|
|
14.27 ± 0.43 | 21.41 ± 0.96 | 9.48 ± 0.35 |
| 16g |
|
|
>50 | >50 | >50 |
| 16h |
|
|
>50 | >50 | >50 |
| 16i |
|
|
>50 | 29.83 ± 2.06 | 30.93 ± 3.35 |
| 16j |
|
|
8.13 ± 0.93 | 14.12 ± 0.11 | 10.40 ± 0.84 |
| 16k |
|
|
13.95 ± 0.99 | 14.18 ± 0.86 | 11.68 ± 0.37 |
| 16l |
|
|
6.16 ± 0.16 | 7.27 ± 0.16 | 9.41 ± 0.67 |
| 16m |
|
|
6.07 ± 0.12 | 7.80 ± 0.32 | 10.21 ± 0.80 |
| 16n |
|
|
19.83 ± 0.60 | 21.20 ± 3.60 | 10.72 ± 0.99 |
| 16o |
|
|
14.75 ± 0.64 | 21.15 ± 3.10 | 22.97 ± 1.70 |
| 16p |
|
|
19.29 ± 0.76 | 17.24 ± 1.20 | 22.29 ± 3.20 |
| 8a [a] | -- | -- | 31.25 ± 0.31 | >50 | 42.10 ± 1.10 |
| 17-AAG | -- | -- | 10.60 ± 0.65 | 39.07 ± 1.30 | 0.45 ± 0.09 |
[a] 8a: 1,3-dibenzyl-2–(5-nitrothiophen-2-yl)imidazolidine.
The effect of the substituent at the N1 position of the imidazole ring was further studied. The benzyl group proved to be the best substituent at the N1 position. As shown in Table 2, the antiproliferative activity decreased after replacing the benzyl group with other functional groups, such as allyl, aliphatic chain, ester, amide, and aliphatic alcohol. This result indicated that the benzyl group may play essential roles in occupying the space and forming hydrophobic interactions with amino acid residues in the binding site of the HSP90 N-terminus.
Table 2.
Antiproliferative activity of bis(4-chlorophenyl)imidazoles 20.
| Cmpd. | R | IC50 (μM) |
||
|---|---|---|---|---|
| MCF-7 | MDA-MB-231 | 4T1 | ||
| 20a |
|
23.14 ± 3.14 | 20.79 ± 1.47 | 22.36 ± 1.99 |
| 20b |
|
>50 | >50 | >50 |
| 20c |
|
>50 | >50 | >50 |
| 20d |
|
19.46 ± 2.28 | >50 | >50 |
| 20e |
|
26.67 ± 3.39 | 39.13 ± 4.38 | 24.70 ± 3.63 |
| 17-AAG | -- | 10.60 ± 0.65 | 39.07 ± 1.30 | 0.45 ± 0.09 |
In the case of 2,4-bis(benzyloxy)-5-arylpyrimidines 22, most compounds in this series were ineffective in inhibiting the proliferation of cancer cell lines (Table 3). However, the introduction of the hydroxyl group dramatically increased the activity. Compound 22k, bearing a 4-hydroxyphenyl group at the C5 position of the pyrimidine ring, exhibited antiproliferative activity with IC50 values of 7.72 ± 0.86, 7.89 ± 0.21, and 7.86 ± 0.76 μM against three breast cancer cell lines. This result suggested that the hydroxyl group may play a crucial role in binding to the target.
Table 3.
Antiproliferative activity of 2,4-bis(benzyloxy)-5-arylpyrimidines 22.
| Cmpd. | R | IC50 (μM) |
||
|---|---|---|---|---|
| MCF-7 | MDA-MB-231 | 4T1 | ||
| 22a |
|
>50 | >50 | >50 |
| 22b |
|
>50 | >50 | >50 |
| 22c |
|
>50 | >50 | >50 |
| 22d |
|
>50 | >50 | >50 |
| 22e |
|
>50 | >50 | >50 |
| 22f |
|
>50 | >50 | >50 |
| 22g |
|
>50 | >50 | >50 |
| 22h |
|
>50 | >50 | >50 |
| 22i |
|
>50 | >50 | >50 |
| 22j |
|
>50 | >50 | >50 |
| 22k |
|
7.72 ± 0.86 | 7.89 ± 0.21 | 7.86 ± 0.76 |
| 22l |
|
12.34 ± 0.50 | 11.17 ± 0.70 | 8.53 ± 0.50 |
| 22m |
|
13.77 ± 0.28 | 12.66 ± 0.03 | 8.25 ± 0.68 |
| 22n |
|
28.78 ± 2.09 | >50 | 14.96 ± 2.03 |
| 22o |
|
>50 | >50 | >50 |
| 22p |
|
>50 | >50 | >50 |
| 17-AAG | 10.60 ± 0.65 | 39.07 ± 1.30 | 0.45 ± 0.09 | |
Western blotting assay
Treating cells with HSP90 N-terminal inhibitors causes the degradation of HSP90 client proteins and the compensatory expression of heat shock proteins. Protein kinase B (also known as AKT) is a serine/threonine kinase and plays a key role in the PI3K signalling pathway40. An extracellular regulated kinase (ERK) is a member of the mitogen-activated protein kinase (MAPK) signalling pathway41. Both AKT and ERK are client proteins of HSP90 and are essential for cancer progression. The effect of 16l and 22k on the expression of the HSP90 client proteins AKT and ERK and the heat shock proteins HSP90 and HSP70 was evaluated by western blotting. As shown in Figure 3, both 16l and 22k significantly decreased the expression levels of AKT and ERK in MCF-7 cells, which was consistent with the features of the classical HSP90 N-terminal inhibitor 17-AAG. It is interesting that, even at a concentration 3 times higher than the IC50 value, the expression level of HSP70 was not significantly increased in the 16l-treated MCF-7 cells, as was the expression level of HSP90 in the 22k treated cells. This result suggested that 16l and 22k inhibited cancer cell proliferation with a lower level of heat shock response in comparison with 17-AAG.
Figure 3.
Western blotting assay. (a) (b) Effects of 16l (L: 6 μM; H: 18 μM) and 22k (L: 8 μM; H: 24 μM) on the expression of AKT, ERK, HSP70, and HSP90 in MCF-7 cells, with GAPDH as an internal reference. 17-AAG (15 μM) was used as a positive control. (c) (d) Statistical analysis of western blotting assays of 16l and 22k, respectively. Data are presented as the means ± SDs. *p < 0.05, **p < 0.01, ***p < 0.001 versus control.
HSP90 binding affinity evaluation using FP assay
To further verify the binding of the synthesised compounds to the HSP90 N-terminus, the binding affinity of five typical compounds (16j, 16l, 16m, 22k, and 22l) was evaluated using an FP assay. The addition of compounds with HSP90 inhibitory capacity into the assay would compete with the fluorescence probe for binding into HSP90, resulting in a decrease in FP in comparison with the probe alone42. As shown in Figure 4, all five compounds exhibited similar binding affinities to HP90 in comparison with 17-AAG. Among them, 22k had the strongest affinity for the HSP90 N-terminus, with an IC50 value of 0.21 ± 0.03 μM.
Figure 4.
Binding affinity evaluation of representative compounds to HSP90 N-terminus.
Molecular docking study
Molecular docking was performed to predict the binding modes of 16l and 22k with the HSP90α N-terminus. Because the FP assay revealed that 16l and 22k could competitively bind to the geldanamycin binding site at the HSP90α N-terminus, a crystal structure of the HSP90α N-terminus and geldanamycin (PDB code: 1YET) was used to generate the receptor structure, and the geldanamycin binding site was employed as the active site for docking. To test the feasibility of our docking method, we docked geldanamycin into the prepared HSP90α N-terminus. The resulting top 10 binding poses were very similar to the experimental pose, with all RMSD values less than 1 Å, which suggested that our docking procedure could afford good pose reproduction (Figure S115 in the supporting information). The top-scoring pose has a -CDOCKER_INTERACTION_ENERGY score of 65.32 with an RMSD value of 0.70. As shown in Figure 5(a), 16l and 22k were successfully docked into the binding pocket of geldanamycin at the N-terminus of HSP90α with -CDOCKER_INTERACTION_ENERGY scores of 36.36 and 55.08, respectively.
Figure 5.
Predicted binding modes of 16l and 22k to the N-terminus of HSP90α (PDB code: 1YET). (a) Overlap of 16l (purple), 22k (yellow) and geldanamycin (green) in the hydrophobic bond surface of the binding pocket; (b) Interactions between 16l and HSP90α residues; (c) Interactions between 22k and HSP90α residues.
16l presented a “T-shaped lock” conformation and was possibly able to block the entrance of ATP into the binding pocket. The benzyloxy group at the N1 position of 16l was directed towards the bottom of the binding pocket, while two 4-chlorophenyl groups were positioned at the mouth of the binding pocket. 16l may form a halogen bond with GLY132, a Pi-cation interaction with LYS58, a Pi-sulphur interaction with MET98, and multiple hydrophobic interactions with residues ALA55, ILE96, and MET98 (Figure 5b). In the case of 22k, the benzyloxy group at the C2 position of the pyrimidine ring was oriented towards the bottom of the binding pocket. It should be noted that 22k may have formed hydrogen bonds with LYS58 and ASP102 (Figure 5c). The hydrogen bond interaction between the hydroxyl group of 22k and ASP102 indicated that the hydroxyl group may be important for stabilising the protein-ligand complex. This result is consistent with the observations in the biological evaluation that the introduction of the hydroxyl group enhanced the antiproliferative activity. In addition, hydrophobic interactions with residues ALA55, MET98, and VAL186 and a Pi-Anion interaction with ASP102 were observed in the predicted binding mode of 22k and the HSP90 N-terminus.
Conclusion
In summary, our work discovered two kinds of novel HSP90 N-terminal inhibitors bearing 2,4-diarylimidazole and 2,4-bis(benzyloxy)-5-arylpyrimidine as their scaffolds. 16l and 22k exhibited strong antiproliferative activities against three breast cancer cell lines, MCF-7, MDA-MB-231, and 4T1. Their inhibitory activity towards the HSP90 N-terminus was validated by western blotting and FP assays, and possible interaction modes were predicted by molecular docking. 16l and 22k can serve as a starting point for more in-depth research on anticancer drugs targeting the HSP90 N-terminus. Our future efforts will focus on evaluating the anticancer activity in vivo and investigating the pharmacokinetic behaviours of 16l and 22k.
Supplementary Material
Funding Statement
This work is funded by the Natural Science Foundation of Liaoning Province of China [Grant No. 2020-MS-105], and the Fundamental Research Funds for the Central Universities [Grant No. DUT20LK20; DUT22YG111].
Disclosure statement
The authors report no conflicts of interest.
References
- 1.Freilich R, Arhar T, Abrams JL, Gestwicki JE.. Protein-protein interactions in the molecular chaperone network. Acc Chem Res. 2018;51(4):940–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Kim YE, Hipp MS, Bracher A, Hayer-Hartl M, Hartl FU.. Molecular chaperone functions in protein folding and proteostasis. Annu Rev Biochem. 2013;82:323–355. [DOI] [PubMed] [Google Scholar]
- 3.Hoter A, El-Sabban ME, Naim HY.. The HSP90 family: structure, regulation, function, and implications in health and disease. IJMS. 2018;19(9):2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schopf FH, Biebl MM, Buchner J.. The HSP90 chaperone machinery. Nat Rev Mol Cell Biol. 2017;18(6):345–360. [DOI] [PubMed] [Google Scholar]
- 5.Picard D. Heat-shock protein 90, a chaperone for folding and regulation. Cell Mol Life Sci. 2002;59(10):1640–1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Whitesell L, Lindquist SL.. HSP90 and the chaperoning of cancer. Nat Rev Cancer. 2005;5(10):761–772. [DOI] [PubMed] [Google Scholar]
- 7.Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC, Burrows FJ.. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature. 2003;425(6956):407–410. [DOI] [PubMed] [Google Scholar]
- 8.Miyata Y, Nakamoto H, Neckers L.. The therapeutic target Hsp90 and cancer hallmarks. Curr Pharm Des. 2013;19(3):347–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Serwetnyk MA, Blagg BSJ.. The disruption of protein-protein interactions with co-chaperones and client substrates as a strategy towards Hsp90 inhibition. Acta Pharm Sin B. 2021;11(6):1446–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Trepel J, Mollapour M, Giaccone G, Neckers L.. Targeting the dynamic HSP90 complex in cancer. Nat Rev Cancer. 2010;10(8):537–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bohush A, Bieganowski P, Filipek A.. Hsp90 and its co-chaperones in neurodegenerative diseases. IJMS. 2019;20(20):4976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, Brooks RW, Grassi D, Gregg SQ, Stripay JL, Dorronsoro A, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8(1):422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Huang DS, LeBlanc EV, Shekhar-Guturja T, Robbins N, Krysan DJ, Pizarro J, Whitesell L, Cowen LE, Brown LE.. Design and synthesis of fungal-selective resorcylate aminopyrazole Hsp90 inhibitors. J Med Chem. 2020;63(5):2139–2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lubkowska A, Pluta W, Strońska A, Lalko A.. Role of heat shock proteins (HSP70 and HSP90) in viral infection. IJMS. 2021;22(17):9366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Noddings CM, Wang RY-R, Johnson JL, Agard DA.. Structure of Hsp90-p23-GR reveals the Hsp90 client-remodelling mechanism. Nature. 2022;601(7893):465–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Verba KA, Wang RY-R, Arakawa A, Liu Y, Shirouzu M, Yokoyama S, Agard DA.. Atomic structure of Hsp90-Cdc37-Cdk4 reveals that Hsp90 traps and stabilizes an unfolded kinase. Science. 2016;352(6293):1542–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Li L, Chen N-N, You Q-D, Xu X-L.. An updated patent review of anticancer Hsp90 inhibitors (2013-present). Expert Opin Ther Pat. 2021;31(1):67–80. [DOI] [PubMed] [Google Scholar]
- 18.Xiao Y, Liu Y.. Recent advances in the discovery of novel HSP90 inhibitors: an update from 2014. Curr Drug Targets. 2020;21(3):302–317. [DOI] [PubMed] [Google Scholar]
- 19.Bickel D, Gohlke H.. C-terminal modulators of heat shock protein of 90 kDa (HSP90): state of development and modes of action. Bioorg Med Chem. 2019;27(21):115080. [DOI] [PubMed] [Google Scholar]
- 20.Wang Y, Mcalpine SR.. N-terminal and C-terminal modulation of Hsp90 produce dissimilar phenotypes. Chem Commun. 2015;51(8):1410–1413. [DOI] [PubMed] [Google Scholar]
- 21.Li L, Wang L, You Q-D, Xu X-L.. Heat shock protein 90 inhibitors: an update on achievements, challenges, and future directions. J Med Chem. 2020;63(5):1798–1822. [DOI] [PubMed] [Google Scholar]
- 22.Azimi A, Caramuta S, Seashore-Ludlow B, Boström J, Robinson JL, Edfors F, Tuominen R, Kemper K, Krijgsman O, Peeper DS, et al. Targeting CDK2 overcomes melanoma resistance against BRAF and Hsp90 inhibitors. Mol Syst Biol. 2018;14(3):e7858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Eroglu Z, Chen YA, Gibney GT, Weber JS, Kudchadkar RR, Khushalani NI, Markowitz J, Brohl AS, Tetteh LF, Ramadan H, et al. Combined BRAF and HSP90 inhibition in patients with unresectable BRAF (V600E)-mutant melanoma. Clin Cancer Res. 2018;24(22):5516–5524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Birbo B, Madu EE, Madu CO, Jain A, Lu Y.. Role of HSP90 in cancer. IJMS. 2021;22(19):10317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Solárová Z, Mojžiš J, Solár P.. Hsp90 inhibitor as a sensitizer of cancer cells to different therapies (review). Int J Oncol. 2015; 46(3):907–926. [DOI] [PubMed] [Google Scholar]
- 26.Mortensen ACL, Mohajershojai T, Hariri M, Pettersson M, Spiegelberg D.. Overcoming limitations of cisplatin therapy by additional treatment with the HSP90 inhibitor onalespib. Front Oncol. 2020;10(532285):532285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Liu Y, Liu X, Li L, Dai R, Shi M, Xue H, Liu Y, Wang H.. Identification and structure-activity studies of 1,3-Dibenzyl-2-aryl imidazolidines as novel Hsp90 inhibitors. Molecules. 2019;24(11):2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hwang JY, Kim H-Y, Jo S, Park E, Choi J, Kong S, Park D-S, Heo JM, Lee JS, Ko Y, et al. Synthesis and evaluation of hexahydropyrimidines and diamines as novel hepatitis C virus inhibitors. Eur J Med Chem. 2013;70:315–325. [DOI] [PubMed] [Google Scholar]
- 29.Godin G, Levrand B, Trachsel A, Lehn J-M, Herrmann A.. Reversible formation of aminals: a new strategy to control the release of bioactive volatiles from dynamic mixtures. Chem Commun. 2010;46(18):3125–3127. [DOI] [PubMed] [Google Scholar]
- 30.Buchs Née Levrand B, Godin G, Trachsel A, de Saint Laumer J-Y, Lehn J-M, Herrmann A.. Reversible aminal formation: controlling the evaporation of bioactive volatiles by dynamic combinatorial/covalent chemistry. Eur J Org Chem. 2011;2011(4):681–695. [Google Scholar]
- 31.Zhang L, Peng X-M, Damu GLV, Geng R-X, Zhou C-H.. Comprehensive review in current developments of imidazole-based medicinal chemistry. Med Res Rev. 2014;34(2):340–437. [DOI] [PubMed] [Google Scholar]
- 32.Jeelan BN, Goudgaon NM.. A comprehensive review on pyrimidine analogs-versatile scaffold with medicinal and biological potential. J Mol Struct. 2021;1246:131168. [Google Scholar]
- 33.Senecal TD, Shu W, Buchwald SL.. A general, practical palladium-catalyzed cyanation of (hetero)aryl chlorides and bromides. Angew Chem Int Ed Engl. 2013;52(38):10035–10039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang D, Haseltine J.. A comparison of phenylboronic acid and phenyltrimethyltin in the palladium-catalyzed arylation of 1,5-dialkylimidazoles. J Heter Chem. 1994;31(6):1637–1639. [Google Scholar]
- 35.O'Connell JF, Parquette J, Yelle WE, Wang W, Rapoport H.. Convenient synthesis of methyl 1-methyl-2,4-dibromo-5-imidazolecarboxylate. Synthesis. 1988;1988(10):767–771. [Google Scholar]
- 36.Li B, Chiu CK-F, Hank RF, Murry J, Roth J, Tobiassen H.. An optimized process for formation of 2,4-disubstituted imidazoles from condensation of amidines and α-haloketones. Org Process Res Dev. 2002;6(5):682–683. − [Google Scholar]
- 37.Stebbins CE, Russo AA, Schneider C, Rosen N, Hartl FU, Pavletich NP.. Crystal structure of an Hsp90-geldanamycin complex: targeting of a protein chaperone by an antitumor agent. Cell. 1997;89(2):239–250. [DOI] [PubMed] [Google Scholar]
- 38.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2021;71(3):209–249. [DOI] [PubMed] [Google Scholar]
- 39.Zagouri F, Sergentanis TN, Chrysikos D, Papadimitriou CA, Dimopoulos M-A, Psaltopoulou T.. Hsp90 inhibitors in breast cancer: a systematic review. Breast. 2013;22(5):569–578. [DOI] [PubMed] [Google Scholar]
- 40.Song M, Bode AM, Dong Z, Lee M-H.. AKT as a therapeutic target for cancer. Cancer Res. 2019;79(6):1019–1031. [DOI] [PubMed] [Google Scholar]
- 41.Liu F, Yang X, Geng M, Huang M.. Targeting ERK, an Achilles’ Heel of the MAPK pathway, in cancer therapy. Acta Pharm Sin B. 2018;8(4):552–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Howes R, Barril X, Dymock BW, Grant K, Northfield CJ, Robertson AGS, Surgenor A, Wayne J, Wright L, James K, et al. A fluorescence polarization assay for inhibitors of Hsp90. Anal Biochem. 2006;350(2):202–213. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.








