Next Article in Journal
Bioactive Components of Salvia and Their Potential Antidiabetic Properties: A Review
Next Article in Special Issue
Selective Targeting of Breast Cancer by Tafuramycin A Using SMA-Nanoassemblies
Previous Article in Journal
The Phytochemical Analysis of Vinca L. Species Leaf Extracts Is Correlated with the Antioxidant, Antibacterial, and Antitumor Effects
Previous Article in Special Issue
Hybrid Drugs—A Strategy for Overcoming Anticancer Drug Resistance?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design, Synthesis, and Antitumor Activity of Olmutinib Derivatives Containing Acrylamide Moiety

School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
*
Authors to whom correspondence should be addressed.
Tel.: +86-791-8380-2393 (S.X.).
Molecules 2021, 26(10), 3041; https://doi.org/10.3390/molecules26103041
Submission received: 25 March 2021 / Revised: 11 May 2021 / Accepted: 12 May 2021 / Published: 20 May 2021

Abstract

:
Two series of olmutinib derivatives containing an acrylamide moiety were designed and synthesized, and their IC50 values against cancer cell lines (A549, H1975, NCI-H460, LO2, and MCF-7) were evaluated. Most of the compounds exhibited moderate cytotoxic activity against the five cancer cell lines. The most promising compound, H10, showed not only excellent activity against EGFR kinase but also positive biological activity against PI3K kinase. The structure–activity relationship (SAR) suggested that the introduction of dimethylamine scaffolds with smaller spatial structures was more favorable for antitumor activity. Additionally, the substitution of different acrylamide side chains had different effects on the activity of compounds. Generally, compounds H7 and H10 were confirmed as promising antitumor agents.

Graphical Abstract

1. Introduction

Recent studies have shown that the mutation rate and overexpression rate of epidermal growth factor receptor (EGFR; a transmembrane protein) of patients with non-small-cell lung cancer (NSCLC) are as high as 75% in vivo [1,2]. EGFR is associated with various growth processes of cancer, including proliferation, differentiation, migration, apoptosis, and angiogenesis [3]. Therefore, EGFR has garnered considerable attention as an antitumor drug target, giving rise to numerous investigations of EGFR kinase inhibitors to disrupt and inhibit the proliferation and growth of tumor cells [4]. The first-generation EGFR inhibitor gefitinib (1) (Figure 1) and the second-generation EGFR inhibitor afatinib (2) have good inhibitory effects on the EGFRL858R mutation and the EGFRT790M mutation, respectively [5,6,7]. However, the first- and second-generation inhibitors have poor kinase selectivity between the EGFRT790M mutant and the wild type. Moreover, their clinical efficacy is limited. The third-generation EGFR inhibitor olmutinib [8,9] (3) was developed by Hanmei Pharmaceutical Company, and is an irreversible inhibitor for the treatment of patients with locally advanced or EGFRT790M-mutant NSCLC (Figure 2) [10]. The IC50 values of 3 against HCC827 (EGFRdell19), H1975 (EGFRL858R/T790M), and A549 (EGFRWT) cells were 9.2 nM, 10 nM, and 225 nM, respectively. This indicated that 3 had strong selectivity for the EGFR mutation. However, 3 had toxic side effects such as palmoplantar keratoderma and diarrhea. Therefore, the goal of this study to obtain new olmutinib derivatives which can overcome the side effects of skeleton migration.
To guide our modification, a molecular docking simulation of 3 and EGFR protein was performed. The thienopyrimidine structure of 3 was inserted into the larger hydrophobic band of the protein and could form hydrogen bonds with MET-793 in the hinged region, as shown in Figure 2A,B. Therefore, we transformed the thiophene pyrimidine structure into thiophene and pyrimidine to explore the antiproliferative activity of the target compounds when fully occupying the protein cavity. At the same time, inspired by WZ4002 (4), we replaced the phenylpiperazine structure of 3 with an anisidine side chain and an electron-withdrawing cyano group [11]. Different acrylamide moieties were used to explore the influence of side-chain length and halogen atoms on the activities of compounds. We used this design idea to synthesize the first series of target compounds (Figure 3).
To design of the second series of target compounds (Figure 4), we retained the thiophene and pyrimidine core and introduced the triazine structure of phosphatidylinositol-3-kinase (PI3K) inhibitor ZSTK474 (5) while at the same time, the structures of thiophene and pyrimidine were transformed into triazine and pyrimidine [12,13]. The introduction of the 1,3,5-triazine ring increased the polarity of the molecule to better form key hydrogen bonds with MET-793 [14]. The oxygen of the morpholine ring of GDC-0941 (6) can form hydrogen bonds with the VAL-851 of the PI3K hinge region. Moreover, the introduction of the morpholine ring enhanced the mTOR-inhibitory activity of the compound; thus, the morpholine ring group was preserved [15,16,17]. Michael receptors were introduced in the solvent region to explore the interaction with the surrounding amino acids. Based on this design strategy, we completed the synthesis of the second series of compounds. We expected to obtain an ideal inhibitor with a better EGFR-inhibitory activity and to explore whether the target compound has an inhibitory effect on PI3K following the modifications described above.

2. Results and Discussion

2.1. Chemistry

According to the structure-based drug design (SBDD) strategy, we designed and synthesized two series of olmutinib derivatives as EGFR inhibitors containing an acrylamide moiety. The synthetic routes of target compounds H1–H16 are outlined in Scheme 1 and Scheme 2.
As shown in Scheme 1, we used commercially available 2,4,6-trichloropyrimidine (7) and thiophen-2-ylboronic acid to obtain 11a–11b through cyclization, chlorination, and nucleophilic substitution reactions. Compounds 11a–11b reacted with different amide side chains to give the target compounds H1–H8. As shown in Scheme 2, we used 1,3,5-triazine as the starting material to obtain 18a–18b with different amines via six steps of substitution, reduction, and chlorination. Finally, 18a–18b reacted with different amide side chains to give the target compounds H9–H16. The structural information of target compounds was confirmed by 1H-NMR, 13C-NMR, and TOF MS (ES+), the results of which were consistent with the structures depicted.

2.2. Biological Discussion

Four human tumor cell lines (A549, H1975, NCI-H460, and MCF-7) and human normal cell line LO2 were selected to evaluate the antiproliferative activity of all target compounds in vitro. Olmutinib was used as a positive control. Results are summarized in Table 1, where the values are the average of at least three independent experiments. Compared with the lead compound olmutinib, most target compounds were less toxic to the normal cell line LO2, which indicates that the target compounds had a selective inhibitory effect on cancer cells. After the introduction of electron-withdrawing cyanide, the antiproliferative activities of compounds H1–H6 that were substituted with cyano groups showed moderate inhibitory activity against all the cell lines. However, the antiproliferative activities of the compounds that were substituted with the anisidine side chain performed better than the cyano group chain. This indicates that the introduction of the electron-withdrawing units could not increase the antiproliferative activity of the compounds. Obviously, compound H7 showed the greatest inhibitory activities against A549 and H1975 cancer cell lines, with IC50 values of 4.37 ± 0.50 μM and 4.59 ± 0.46 μM, respectively, which were similar to the reference compound of olmutinib.
Table 1 shows that the introduction of the 1,3,5-triazine ring and morpholine ring significantly enhanced the antiproliferation activity of the compounds. At the same time, we found that the antiproliferative activities of the compounds that were substituted with dimethylamine groups, were better than those that were substituted with diethylamine groups. Therefore, we speculate that the inner cavity area of the hydrophobic region is limited and only can accommodate molecules with smaller structures. From the docking results (Figure 5B), it was found that the dimethylamine group of compound H10 penetrated into the interior of the protein (4zau) and completely occupied the space, which suggests that the introduction of a larger group than dimethylamine will not increase the activity of these compounds. The optimal compound H10 showed excellent antiproliferative activity against A549 and MCF-7 cancer cell lines, with IC50 values of 3.36 ± 1.59 μM and 13.05 ± 1.36 μM, respectively, which were superior to the drug of reference. The selectivity of compound H10 to A549 cells was 29.76 times than to LO2 cells, and about 5 times that of the lead compound olmutinib.
Compounds H7 and H10 had excellent antiproliferative activity and were further screened out for kinase inhibition testing. We evaluated compounds H7 and H10 with EGFRT790M/L858R kinases and PI3Kα kinases. As shown in Table 2, compounds H7 and H10 exhibited potent inhibition against EGFRT790M/L858R kinase. In particular, compound H10 showed better inhibitory activity against PI3Kα kinase than the control drug olmutinib. These data demonstrate that compound H10 is expected to be a dual inhibitor of EGFR and PI3K.

2.3. Molecular Docking Study

To explore the binding mode of the target compound (H10) with the active site of EGFR, molecular docking simulation was carried out using AutoDock 4.2 software. The docking tutorials and detailed explanations of the AutoDock basic methods we used can be found at the following address: http://autodock.scripps.edu/faqs help/tutorial (accessed on 25 March 2021). According to the analysis results of the cells and kinases, we chose compound H10 as the example ligand; the structures of EGFRWT (PDB CODE: 4zau) and EGFRT790M (PDB CODE: 3ika) were selected as docking models.
The combination of compound H10 with the EGFRT790M and PI3Kγ molecular active sites is shown in Figure 5. When compound H10 docked with the 3ika (Figure 5A), we observed that the dimethylamine group extended into the ATP hydrophobic pocket and formed hydrogen bonds with residues LYS-745, and the amino group formed hydrogen bonds with MET-793 residues. Figure 5C shows that the morpholine rings of compound H10 formed hydrogen bonds with the VAL-882 residues in 3L08. Among these, the acrylamide side chain of compound H10 formed hydrogen bonds with the LYS-833 residue in 3L08. This was in line with the combination model we predicted before. The abovementioned SAR (structure–activity relationship) analysis and molecular docking results indicate that compound H10 could be a potentially interesting anticancer agent.

3. Experimental Section

3.1. General Information

Unless otherwise stated, all reagents used in the experiment were purchased at commercial analytical grade and used directly without further purification. Common solvents (ethanol, methanol, petroleum ether, ethyl acetate, dichloromethane 1,2-dimethoxyethane, etc.) were absolutely anhydrous. All reactions were monitored on a GF254 thin-layer chromatography plate (Laishan Penghan Plastic Industry Store, Yantai, Shandong, China), and spots were visualized at 254 nanometers or 365 nanometers with iodine or light. The target compound (20 mg) and 3.5 mL DMSO formed the corresponding compound solution, and the structure of the target compound was confirmed by 1H-NMR and 13C-NMR on a Bruker 400 MHz spectrometer (Bruker Bioscience, Billerica, MA, USA) using tetramethylsilane (TMS) as an internal standard at room temperature (see Supplementary Materials). The target compounds (0.5 mg) and LC-MS methanol were prepared in a 0.5 µg/mL mixed solution, and mass spectrometry (MS) of target compounds was carried out on a Waters High Resolution Quadrupole Time of Flight Tandem Mass Spectrometer (Waters, Milford, MA, USA, Xevo G2-XS Tof). The purity of all compounds was determined using an Agilent 1260 liquid chromatograph equipped with an Inertex-C18 column. The purity of all target compounds was ≥95%.

3.2. Chemistry

3.2.1. Representative Procedure for the Synthesis of 2,4-dichloro-6-(thiophen-2-yl)pyrimidine (8)

2,4,6-Trichloropyrimidine 7 (70.0 g, 381.6 mmol) and thiophen-2-ylboronic acid (25.2 g, 196.8 mmol) were coupled by coupling reaction in 1,2-dimethoxyethane and water 5:1 solvent. The above solution was stirred at 90 °C for about 1.5 h. The reaction was monitored by TLC. After the reaction, the reactant was purified via silica gel column chromatography to obtain compound 8. Yield: 95%; color: yellow; m.p.: 145.1–147.5 °C. 1H NMR (400 MHz, DMSO-d6) δ 7.49 (s, 1H), 7.23 (s, 1H), 7.20 (s, 1H), 7.19 (s, 1H). TOF MS ES+ (m/z): [M + H]+, calcd for C8H4Cl2N2S: 232.1100, found, 232.1103.

3.2.2. Representative Procedure for the Synthesis of 2-chloro-4-(3-nitrophenoxy)-6-(thiophen-2-yl)pyrimidine (9)

Intermediate 8 (19.0 g, 82.3 mmol), metanitrophenol (12.0 g, 86.3 mmol), and cesium carbonate (32.0 g, 99.5 mmol) were dissolved in 1,4-dioxane (120 mL), and then stirred for 4 h at room temperature. The completion of the reaction was monitored by TLC. After the reaction, the reaction solvent was removed under reduced pressure to obtain a yellow solid 9. Yield: 91.4%; color: yellow; m.p.: 152–155 °C. 1H NMR (400 MHz, DMSO-d6) δ 8.05 (s, 1H), 8.02 (s, 1H), 7.64 (s, 1H), 7.54 (m, 1H), 7.24 (s, 1H), 7.21 (s, 1H), 7.19 (s, 1H), 7.16 (s, 1H). TOF MS ES+ (m/z): [M + H]+, calcd for C14H8ClN3O3S: 333.7869, found, 333.7872.

3.2.3. Representative Procedure for the Synthesis of 10a–10b

Intermediate 9 (5.3 g, 16.0 mmol), toluene-p-sulfonic acid (5.6 g, 32.5 mmol) and different amino side chains (16.0 mmol) were dissolved in acetonitrile (60 mL) and stirred for 4–5 h at 100 °C. After the reaction, the reaction solvent was removed under reduced pressure and a large amount of solid precipitated after adding twice the amount of water. The solid was filtered at atmospheric pressure and dried to obtain 10a–10b.
N-(3-methoxyphenyl)-4-(3-nitrophenoxy)-6-(thiophen-2-yl)pyrimidin-2-amine (10a)
 
Yield: 90.1%; color: yellow; m.p.: 169.5–172.7 °C. 1H NMR (400 MHz, DMSO-d6) δ 8.36 (s, 1H), 8.05 (s, 1H), 7.91 (s, 1H), 7.89 (s, 1H), 7.73 (s, 1H), 7.67 (s, 1H), 7.57–7.54 (m, 1H), 7.30 (dd, J = 7.5, 1.5 Hz, 1H), 7.24 (d, J = 7.4 Hz, 1H), 7.23–7.16 (m, 1H), 7.14 (dt, J = 7.5, 1.5 Hz, 1H), 6.89 (s, 1H), 6.57 (dt, J = 7.5, 1.6 Hz, 1H), 3.80 (s, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C21H15N3O4S: 405.0925, found, 405.0927.
3-((4-(3-nitrophenoxy)-6-(thiophen-2-yl)pyrimidin-2-yl)amino)benzonitrile (10b)
 
Yield: 80.7%; color: yellow; m.p.: 176.1–179.3 °C. 1H NMR (400 MHz, DMSO-d6) δ 8.23 (s, 1H), 8.05 (dt, J = 7.5, 1.6 Hz, 1H), 7.94 (s, 1H), 7.84 (s, 1H), 7.64 (dd, J = 7.3, 1.6 Hz, 1H), 7.55 (t, J = 7.5 Hz, 1H), 7.45–7.40 (m, 1H), 7.39 (d, J = 7.5 Hz, 1H), 7.33 (dt, J = 7.1, 1.7 Hz, 1H), 7.24 (dd, J = 7.5, 1.6 Hz, 1H), 7.19 (t, J = 7.4 Hz, 1H), 7.14 (dt, J = 7.5, 1.5 Hz, 1H), 6.89 (s, 1H). TOF MS ES+ (m/z): [M + H]+, calcd for C21H12N4O3S: 401.0746, found, 401.0749.

3.2.4. Representative Procedure for the Synthesis of 11a–11b

Intermediate 10a–10b (13mmol), ferric chloride (4.2 g, 15.6 mmol), and activated carbon (1.1 g, 91.0 mmol) were dissolved in ethanol. The solution was warmed to 80 °C, and then water and hydrazine were added (6.5 g, 130 mmol) and the solution stirred for 4 h. After the reaction, the reaction solvent was removed under reduced pressure, saturated sodium bicarbonate aqueous solution was added, and the solution was filtered to generate 11a–11b.
4-(3-aminophenoxy)-N-(3-methoxyphenyl)-6-(thiophen-2-yl)pyrimidin-2-amine (11a)
 
Yield: 78.3%; color: yellow; m.p.: 192.5–195.4 °C. 1H NMR (400 MHz, DMSO-d6) δ 8.46 (s, 1H), 7.83 (s, 1H), 7.74 (s, 1H), 7.66 (s, 1H), 7.27 (dt, J = 6.3, 4.5 Hz, 1H), 7.23 (dt, J = 6.8, 3.6 Hz, 1H), 7.22–7.20 (m, 1H), 7.17 (s, 1H), 7.02 (s, 1H), 6.91 (s, 1H), 6.83 (s, 1H), 6.57 (s, 1H), 6.17 (s, 1H), 5.06 (s, 2H), 3.80 (s, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C21H17N3O2S: 376.1405, found, 376.1408.
3-((4-(3-aminophenoxy)-6-(thiophen-2-yl)pyrimidin-2-yl)amino)benzonitrile (11b)
 
Yield: 71.1%; color: yellow; m.p.: 189.7–193.5 °C. 1H NMR (400 MHz, DMSO-d6) δ 8.23 (s, 1H), 7.83 (s, 1H), 7.66 (s, 1H), 7.42 (dt, J = 10.2, 9.1Hz, 1H), 7.40 (dt, J = 8.7, 5.6 Hz, 1H), 7.33 (s, 1H), 7.25 (s, 1H), 7.21 (s, 1H), 7.19 (s, 1H), 7.02 (s, 1H), 6.92 (s, 1H), 6.83 (s, 1H), 6.17 (s, 1H), 5.06 (s, 2H). TOF MS ES+ (m/z): [M + H]+, calcd for C21H14N4OS: 371.0983, found, 371.0980.

3.2.5. Representative Procedure for the Synthesis of Target Compounds H1–H8

Intermediate 11a–11b (1.5 mmol) and bicarbonate (0.25 g, 3 mmol) were dissolved in dichloromethane and reacted with different amide side chains under ice-bath conditions. The completion of the reaction was monitored by TLC. The reactant was purified via silica gel column chromatography to obtain the target compounds H1–H8 with high purity.

3.2.6. Representative Procedure for the Synthesis of 12–16

The specific operation was carried out according to our previous research and the physical data were in agreement with reported values [18].

3.2.7. Representative Procedure for the Synthesis of 17a–17b

Intermediate 16 (0.5 g, 1.5 mmol), different amino side chains (3 mmol), and two drops of N,N-diisopropylethylamine (DIPEA) were dissolved in isopropanol (50 mL) and stirred for 1 h at 75 °C. After the reaction, the reaction solvent was removed under reduced pressure to obtain a yellow liquid 17a–17b.
1-(5-(4-chloro-6-morpholino-1,3,5-triazin-2-yl)thiophen-2-yl)-N,N-dimethylmethanamine (17a)
 
Yield: 85.1%; color: yellow; m.p.: 174.2–175.4 °C. TOF MS ES+ (m/z): [M + H]+, calcd for C14H18ClN5OS: 339.9370, found, 339.9373.
N-((5-(4-chloro-6-morpholino-1,3,5-triazin-2-yl)thiophen-2-yl)methyl)-N-ethylethanamine (17b)
 
Yield: 82.2%; color: yellow; m.p.: 163.8–165.1 °C. TOF MS ES+ (m/z): [M + H]+, calcd for C16H22ClN5OS: 368.2436, found, 368.2433.

3.2.8. Representative Procedure for the Synthesis of 18a–18b

Intermediate 17a–17b (1 mmol), 3-aminophenol (0.1 g, 1.1 mmol), and tert-butoxide (0.22 g, 2 mmol) were dissolved in THF and then stirred for 1 h under ice-bath conditions. The reaction was monitored by TLC. After the reaction, the reaction solvent was removed under reduced pressure to obtain a yellow liquid 18a–18b.
3-((4-(5-((dimethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)aniline (18a)
 
Yield: 78.2%; color: yellow; m.p.: 179.3–182.9 °C. TOF MS ES+ (m/z): [M + H]+, calcd for C20H24N6O2S: 413.5214, found, 413.5211.
3-((4-(5-((diethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)aniline (18b)
 
Yield: 85.1%; color: yellow; m.p.: 178.4–181.5 °C. TOF MS ES+ (m/z): [M + H]+, calcd for C22H28N6O2S: 441.6275, found, 441.6277.

3.2.9. Representative Procedure for the Synthesis of Target Compounds H9–H16

The synthesis of target compounds H9–H16 was similar to that of target compounds H1–H8.
 
N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)acrylamide (H1). Yield: 40.3%; color: yellow; m.p.: 197.2–199.4 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 9.95 (s, 1H), 8.07 (s, 1H), 7.85 (s, 2H), 7.70 (d, J = 6.3 Hz, 1H), 7.51 (d, J = 8.3 Hz, 1H), 7.46 (t, J = 8.1 Hz, 2H), 7.33 (s, 2H), 7.28–7.24 (m, 1H), 7.16 (s, 1H), 7.03–6.99 (m, 1H), 6.42 (dd, J = 16.9, 10.1 Hz, 1H), 6.27 (dd, J = 16.9, 2.0 Hz, 1H), 5.79–5.74 (m, 1H). TOF MS ES+ (m/z): [M + H]+, calcd for C25H19N5O2S: 456.3863, found, 456.3864.
 
(E)-N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)but-2-enamide (H2). Yield: 58.3%; color: yellow; m.p.: 201.2–205.9 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.13 (s, 1H), 9.95 (s, 1H), 8.08 (d, J = 5.8 Hz, 2H), 7.85 (d, J = 5.1 Hz, 2H), 7.68 (d, J = 10.9 Hz, 1H), 7.47 (d, J = 8.2 Hz, 1H), 7.42 (d, J = 7.9 Hz, 1H), 7.34 (s, 2H), 7.27–7.24 (m, 1H), 7.16 (s, 1H), 6.99 (d, J = 8.1 Hz, 1H), 6.80 (dd, J = 15.3, 7.6 Hz, 1H), 6.14 (d, J = 15.3 Hz, 1H), 1.86 (d, J = 7.0 Hz, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C25H19N5O2S: 454.5743, found, 454.5746.
 
N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)-3-methylbut-2-enamide (H3). Yield: 66.3%; color: yellow; m.p.: 215.5–218.7 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.01 (s, 1H), 9.95 (s, 1H), 8.07 (d, J = 3.8 Hz, 1H), 7.85 (d, J = 4.9 Hz, 2H), 7.67 (s, 1H), 7.42-7.40 (m, 3H), 7.34 (s, 2H), 7.27–7.24 (m, 1H), 7.15 (s, 1H), 6.96 (d, J = 8.1 Hz, 1H), 5.86 (s, 1H), 2.12 (s, 3H), 1.85 (s, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C25H19N5O2S: 468.5745, found, 468.5747.
 
(E)-N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)hex-2-enamide (H4). Yield: 48.6%; color: yellow; m.p.: 209.6–211.4 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.15 (s, 1H), 9.95 (s, 1H), 8.07 (d, J = 3.7 Hz, 1H), 7.85 (d, J = 5.0 Hz, 2H), 7.68 (s, 1H), 7.48 (d, J = 8.4 Hz, 1H), 7.42 (t, J = 8.0 Hz, 2H), 7.34 (s, 2H), 7.26 (dd, J = 5.0, 3.8 Hz, 1H), 7.16 (s, 1H), 6.98 (d, J = 7.9 Hz, 1H), 6.82–6.76 (m, 1H), 6.10 (d, J = 15.4 Hz, 1H), 2.18 (d, J = 7.0 Hz, 2H), 1.46 (d, J = 7.3 Hz, 2H), 0.90 (d, J = 3.5 Hz, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C25H19N5O2S: 482.3266, found, 482.3265.
 
N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)-2-fluoroacrylamide (H5). Yield: 33.1%; color: yellow; m.p.: 222.9–226.8 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 9.97 (s, 1H), 8.08 (d, J = 4.0 Hz, 1H), 7.86 (d, J = 5.3 Hz, 1H), 7.70 (d, J = 10.7 Hz, 2H), 7.49 (t, J = 8.0 Hz, 2H), 7.34 (s, 2H), 7.27 (d, J = 4.7 Hz, 1H), 7.17 (s, 1H), 7.09 (d, J = 8.6 Hz, 1H), 5.78 (d, J = 3.7 Hz, 1H), 5.65 (s, 1H), 5.48–5.41 (m, 1H). TOF MS ES+ (m/z): [M + H]+, calcd for C24H16FN5O2S: 458.4987, found, 458.4989.
 
(E)-N-(3-((2-((3-cyanophenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)-4-methylpent-2-enamide (H6). Yield: 51.8%; color: yellow; m.p.: 223.1–225.4 °C 1H NMR (400 MHz, DMSO-d6) δ 10.19 (s, 1H), 9.94 (s, 1H), 8.06 (d, J = 3.8 Hz, 1H), 7.84 (d, J = 5.2 Hz, 2H), 7.68 (d, J = 4.5 Hz, 1H), 7.48 (d, J = 8.3 Hz, 1H), 7.42 (t, J = 8.1 Hz, 1H), 7.33 (s, 2H), 7.25 (t, J = 4.4 Hz, 1H), 7.15 (s, 1H), 6.98 (d, J = 8.0 Hz, 1H), 6.81 (d, J = 6.3 Hz, 1H), 6.79–6.74 (m, 1H), 6.06 (d, J = 15.4 Hz, 1H), 2.44 (dd, J = 13.1, 6.6 Hz, 1H), 1.03 (d, J = 6.5 Hz, 6H). TOF MS ES+ (m/z): [M + H]+, calcd for C27H23N5O2S: 482.5866, found, 482.5868.
 
(E)-N-(3-((2-((3-methoxyphenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)but-2-enamide (H7). Yield: 43.7%; color: yellow; m.p.: 231.7–234.3 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.15 (s, 1H), 9.43 (s, 1H), 8.00 (s, 1H), 7.23 (s, 2H), 6.76 (d, J = 6.9 Hz, 5H), 6.08 (d, J = 15.2 Hz, 6H), 3.67 (s, 3H), 1.93 (d, J = 7.8 Hz, 3H). 13C NMR (101 MHz, DMSO-d6) δ 170.28, 163.61, 163.02, 159.31, 155.14, 154.13, 140.54, 140.28, 139.03, 133.31, 132.43, 130.17, 129.72, 128.50, 127.84, 126.10, 120.67, 116.37, 115.97, 113.36, 112.62, 55.11, 17.41. TOF MS ES+ (m/z): [M + H]+, calcd for C25H22N4O3S: 459.5356, found, 459.5359.
 
(E)-N-(3-((2-((3-methoxyphenyl)amino)-6-(thiophen-2-yl)pyrimidin-4-yl)oxy)phenyl)-4-methylpent-2-enamide (H8). Yield: 22.6%; color: yellow; m.p.: 217.9–219.1 °C; 1H NMR 400 MHz, DMSO-d6) δ 9.83 (s, 2H), 7.55 (d, J = 8.8 Hz, 4H), 6.87 (d, J = 8.9 Hz, 5H), 6.75 (dd, J = 15.4, 6.4 Hz, 3H), 6.05 (s, 1H), 6.01 (s, 1H), 3.72 (s, 3H), 1.06 (s, 7H). TOF MS ES+ (m/z): [M + H]+, calcd for C27H26N4O3S: 487.6124, found, 487.6126.
 
N-(3-((4-(5-((dimethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)acrylamide (H9). Yield: 76.8%; color: yellow; m.p.: 224.9–226.1 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.39 (s, 1H), 7.25 (s, 2H), 7.08 (s, 1H), 7.06 (s, 1H), 7.03 (s, 1H), 7.01 (s, 1H), 6.48–6.47 (m, 1H), 6.25–6.21 (m, 1H), 5.74 (s, 1H), 3.64 (s, 4H), 3.58 (s, 4H), 3.02 (s, 2H), 1.24 (s, 6H). TOF MS ES+ (m/z): [M + H]+, calcd for C23H26N6O3S: 466.5668, found, 466.5670.
 
(E)-N-(3-((4-(5-((dimethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)but-2-enamide (H10). Yield: 63.5%; color: yellow; m.p.: 202.4–203.7 °C; 1H NMR ((400 MHz, DMSO-d6) δ 9.65 (s, 1H), 9.38 (s, 2H), 7.25 (s, 1H), 7.05 (s, 2H), 6.46 (s, 1H), 5.75 (s, 1H), 5.48 (s, 1H), 3.64 (s, 4H), 3.58 (s, 4H), 3.02 (s, 2H), 1.93 (s, 6H), 1.23 (s, 3H). 13C NMR (101 MHz, DMSO-d6) δ 170.01, 169.89, 168.95, 163.76, 163.12, 157.33, 157.13, 146.17, 142.55, 142.09, 136.74, 136.45, 134.96, 128.48, 124.20, 121.58, 117.67, 114.48, 113.04, 58.14, 36.37, 35.08, 32.23, 24.75. TOF MS ES+ (m/z): [M + H]+, calcd for C24H28N6O3S: 480.5873, found, 480.5875.
 
N-(3-((4-(5-((dimethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)-3-methylbut-2-enamide (H11). Yield: 84.5%; color: yellow; m.p.: 198.7–199.8 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.30 (s, 1H), 7.76 (s, 1H), 7.23 (s, 1H), 6.98 (s, 2H), 6.77 (s, 1H), 6.42 (s, 1H), 5.99 (s, 1H), 3.08-3.15 (m, 10H), 2.21 (s, 9H), 1.84 (s, 3H). TOF MS ES+ (m/z): [M + H]+, calcd for C24H28N6O3S: 482.6257, found, 482.6256.
 
(E)-N-(3-((4-(5-((dimethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)-4-methylpent-2-enamide (H12). Yield: 78.5%; color: yellow; m.p.: 199.7–201.3 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.85 (s, 1H), 9.39 (s, 1H), 7.76 (s, 1H), 7.28 (s, 1H), 7.07 (m, 2H), 6.83 (m, 1H), 6.46 (s, 1H), 6.10 (m, 1H), 3.64 (s, 4H), 3.58 (s, 4H), 3.02 (s, 2H), 1.93(s, 7H), 1.06 (s, 6H). TOF MS ES+ (m/z): [M + H]+, calcd for C26H32N6O3S: 508.6263, found, 508.6266.
 
N-(3-((4-(5-((diethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)methacrylamide (H13). Yield: 64.7%; color: yellow; m.p.: 240.7–243.5 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.71 (s, 1H), 7.78 (s, 1H), 7.35 (s, 1H), 6.97 (s, 1H), 6.54 (s, 1H), 6.02 (s, 1H), 5.82 (s, 1H), 5.60 (s, 1H), 5.51 (s, 1H), 3.75 (s, 6H), 1.98 (s, 4H), 1.89 (s, 4H), 1.16 (d, 3H), 1.00 (m, 6H). TOF MS ES+ (m/z): [M + H]+, calcd for C26H32N6O3S: 509.6956, found, 509.6953.
 
(E)-N-(3-((4-(5-((diethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)but-2-enamide (H14). Yield: 35.8%; color: yellow; m.p.: 189.7–192.1 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.84 (s, 1H), 7.74 (s, 1H), 7.29 (s, 1H), 6.96 (s, 1H), 6.78 (d, J = 6.9 Hz, 1H), 6.15 (s, 1H), 6.11 (s, 1H), 5.82 (s, 1H), 5.78 (s, 1H), 3.74 (s, 6H), 1.83 (m, 4H), 1.14 (d, 4H), 0.98 (m, 9H). TOF MS ES+ (m/z): [M + H]+, calcd for C26H32N6O3S: 509.6933, found, 509.6931.
 
N-(3-((4-(5-((diethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)-3-methylbut-2-enamide (H15). Yield: 41.5%; color: yellow; m.p.: 215.2–217.1 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.71 (s, 1H), 7.27 (s, 1H), 7.06 (s, 1H), 7.00 (s, 1H), 6.45 (s, 1H), 6.02 (s, 1H), 5.87 (s, 1H), 5.63 (s, 1H), 3.74 (s, 6H), 2.15 (s, 4H), 1.86 (s, 4H), 1.14 (s, 6H), 0.99 (s, 6H). TOF MS ES+ (m/z): [M + H]+, calcd for C26H32N6O3S: 523.8974, found, 523.8971.
 
(E)-N-(3-((4-(5-((diethylamino)methyl)thiophen-2-yl)-6-morpholino-1,3,5-triazin-2-yl)oxy)phenyl)-4-methylpent-2-enamide (H16). Yield: 26.4%; color: yellow; m.p.: 222.9.2–226.4 °C; 1H NMR (400 MHz, DMSO-d6) δ 9.88 (s, 1H), 7.75 (s, 1H), 7.30 (s, 1H), 7.05 (s, 1H), 6.79 (s, 1H), 6.48 (s, 1H), 6.10 (s, 1H), 6.07 (s, 1H), 3.72 (s, 6H), 1.14 (s, 10H), 1.05 (d, J = 6.7 Hz, 12H). TOF MS ES+ (m/z): [M + H]+, calcd for C26H32N6O3S: 536.6475, found, 536.6473.

3.3. EGFR and PI3Kα Kinase Assay

The potent compounds H7 and H10 were tested for their activities against EGFRT790M/L858R and PI3Kα enzyme using the Kinase-Glo Luminescent Kinase Assay, with olmutinib as a positive control. The specific operation was carried out according to our previous research [19,20].

3.4. Cytotoxicity Assay In Vitro

The in vitro cytotoxic activities of all compounds H1–H16 were evaluated with A549, H1975, LO2, and MCF-7 cell lines using the standard MTT assay, with olmutinib as a positive control [21].

3.5. Docking Studies

The three-dimensional structure of EGFR (PDB code: 4azu, 3L08) was obtained from the RCSB Protein Data Bank. We used AutoDock 4.2 software (The Scripps Research Institute, USA) for molecular docking. The docking process mainly included fixing the exact residues, adding hydrogen atoms, removing irrelevant water molecules, adding charges, etc. All the docking results were processed and modified in Open-Source PyMOL 1.8. x software (https://pymol.org (accessed on 25 March 2021).).

4. Conclusions

In summary, two series of olmutinib derivatives containing an acrylamide moiety (H1–H16) were synthesized and the pharmacological results indicate that most of the compounds exhibited moderate cytotoxic activity against five cell lines (A549, H1975, NCI-H460, LO2, and MCF-7). Among the most effective compounds, the IC50 values of H10 against A549 and H1975 cells were 3.36 ± 1.59 µM and 1.16 ± 1.53 µM, respectively. The structure–activity relationship (SAR) indicated that the introduction of small-molecule swelling of dimethylamine was more favorable for the activity of the compounds. The amino group and morpholine rings formed hydrogen bonds on the 3ika and 3l08 MET-793 residues, respectively. The kinase activity of compound H10 on PI3K was higher than that of the lead compound olmutinib at 1 µM, which indicates that compound H10 may be a new dual inhibitor of EGFR and PI3K. Further research will be conducted in the near future.

Supplementary Materials

The Supplementary materials are available online, Figures S1–S22: 1H-NMR spectrum of compound H1–H16, 8, 9, 10a–10b, 11a–11b. Figures S23 and S24: 13C-NMR spectrum of compound H7 and H10. Figures S25–S50: TOF MS of compound H1–H16, 8, 9, 10a–10b, 11a–11b, 17a–17b, 18a–18b.

Author Contributions

W.Z., Q.P. and X.H. conceived and designed the experiments; X.H. and S.T. performed the experiments; F.Y. ran the bioassay evaluation and statistics analysis; and X.H., P.Z., S.X., W.Z. and Q.P. started the project and wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

We gratefully acknowledge the generous support provided by The National Natural Science Funds (No. 21967009); Natural Science Foundation of Jiangxi, China (20202BABL216076); Science and Technology Project Founded by the Education Department of Jiangxi Province, China (GJJ190583) and Nanchang Key Laboratory of Molecular Targeted Anticancer Drug Design and Evaluation (2019-NCZDSY-007).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds H1–H16 are available from the authors.

References

  1. Shepherd, F.A.; Rodrigues, P.J.; Ciuleanu, T.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 2005, 353, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.S.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef] [PubMed]
  3. Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, P.S.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFR Mutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
  4. Hiramatsu, M.; Ninomiya, H.; Inamura, K.; Okui, M.; Satoh, Y.; Fujiwara, M.; Yamori, T.; Kitagawa, T.; Ishikawa, Y. EGFR signal pathways selectively activated in lung adenocarcinoma. Cancer Res. 2007, 67, 5038. [Google Scholar]
  5. Cohen, M.H.; Williams, G.A.; Sridhara, R.; Chen, G.; McGuinn, W.D., Jr.; Morse, D.; Abraham, S.; Rahmans, A.; Liang, C.; Lostritto, R.; et al. United States Food and Drug Administration Drug Approval Summary: Gefitinib (ZD1839; Iressa) Tablets. Clin. Cancer Res. 2004, 10, 1212–1218. [Google Scholar] [CrossRef] [Green Version]
  6. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.-M.; Zhao, X.; Christensen, J.; et al. MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef]
  7. Janjigian, Y.Y.; Smit, E.F.; Groen, H.J.; Horn, L.; Gettinger, S.; Camidge, D.R.; Riely, G.J.; Wang, B.; Fu, Y.; Chand, V.K.; et al. Dual Inhibition of EGFR with Afatinib and Cetuximab in Kinase Inhibitor–Resistant EGFR-Mutant Lung Cancer with and without T790M Mutations. Cancer Discov. 2014, 4, 1036–1045. [Google Scholar] [CrossRef] [Green Version]
  8. Kim, E.S. Olmutinib: First Global Approval. Drugs 2016, 76, 1153–1157. [Google Scholar] [CrossRef]
  9. Chen, K.L.; Cho, Y.T.; Yang, C.W.; Sheen, Y.S.; Liang, C.W.; Lacouture, M.E.; Chu, C.Y. Olmutinib-induced palmoplantar keratoderma. Br. J. Dermatol. 2018, 178, 129–131. [Google Scholar] [CrossRef] [PubMed]
  10. Kim, D.-W.; Lee, D.H.; Han, J.-Y.; Lee, J.; Cho, B.C.; Kang, J.H.; Lee, K.H.; Cho, E.K.; Kim, J.-S.; Min, Y.J.; et al. Safety, tolerability, and anti-tumor activity of olmutinib in non-small cell lung cancer with T790M mutation: A single arm, open label, phase 1/2 trial. Lung Cancer 2019, 135, 66–72. [Google Scholar] [CrossRef] [PubMed]
  11. Sakuma, Y.; Yamazaki, Y.; Nakamura, Y.; Yoshihara, M.; Matsukuma, S.; Nakayama, H.; Yokose, T.; Kameda, Y.; Koizume, S.; Miyagi, Y. WZ4002, a third-generation EGFR inhibitor, can overcome anoikis resistance in EGFR-mutant lung adenocarcinomas more efficiently than Src inhibitors. Lab. Investig. 2012, 92, 371–383. [Google Scholar] [CrossRef] [Green Version]
  12. Dexin, K.T. ZSTK474 is an ATP-competitive inhibitor of class I phosphatidylinositol 3 kinase isoforms. Cancer Sci. 2007, 98, 1638–1642. [Google Scholar]
  13. Namatame, N.; Tamaki, N.; Yoshizawa, Y.; Okamura, M.; Nishimura, Y.; Yamazaki, K.; Tanaka, M.; Nakamura, T.; Semba, K.; Yamori, T.; et al. Antitumor profile of the PI3K inhibitor ZSTK474 in human sarcoma cell lines. Oncotarget 2018, 9, 35141–35161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Folkes, A.J.; Ahmadi, K.; Alderton, W.K.; Alix, S.; Baker, S.J.; Box, G.; Chickowree, I.S.; Clarke, P.A.; Depledge, P.; Eccles, S.A.; et al. The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J. Med. Chem. 2008, 51, 5522–5532. [Google Scholar] [CrossRef] [PubMed]
  15. Yao, E.; Zhou, W.; Lee-Hoeflich, S.T.; Truong, T.; Haverty, P.M.; Eastham-Anderson, J.; Lewin-Koh, N.; Gunter, B.; Belvin, M.; Murray, L.J.; et al. Suppression of HER2/HER3-Mediated Growth of Breast Cancer Cells with Combinations of GDC-0941 PI3K Inhibitor, Trastuzumab, and Pertuzumab. Clin. Cancer Rese. 2009, 15, 4147–4156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Shaw, R.; Cantley, L. Ras, PI3K and mTOR signalling controls tumour cell growth. Nature 2006, 441, 424–430. [Google Scholar] [CrossRef] [PubMed]
  17. Verbitski, S.M.; Mullally, J.E.; Fitzpatrick, F.A.; Ireland, C.M. Punaglandins, chlorinated prostaglandins, function as potent Michael receptors to inhibit ubiquitin isopeptidase activity. J. Med. Chem. 2004, 47, 2062–2070. [Google Scholar] [CrossRef]
  18. Zhang, B.; Zhang, Q.; Xiao, Z.; Sun, X.; Yang, Z.; Gu, Q.; Liu, Z.; Xie, T.; Jin, Q.; Zheng, P.; et al. Design, synthesis and biological evaluation of substituted 2-(thiophen-2-yl)-1,3,5-triazine derivatives as potential dual PI3Kα/mTOR inhibitors. Bioorg. Chem. 2020, 95, 103525. [Google Scholar] [CrossRef]
  19. Lei, F.; Sun, C.; Xu, S.; Wang, Q.; Ouyang, Y.; Chen, C. Design, synthesis, biological evaluation and docking studies of novel 2-substituted-4-morpholino-7,8-dihydro-5H-thiopyrano[4,3-d]- pyrimidine derivatives as dual PI3Kα/mTOR inhibitors. Eur. J. Med. Chem. 2016, 116, 27–35. [Google Scholar] [CrossRef]
  20. Zhao, B.; Lei, F.; Wang, C.; Zhang, B.; Yang, Z.; Li, W.; Zhu, W.; Xu, S. Design, Synthesis and Biological Evaluation of Novel Phenylsulfonylurea Derivatives as PI3K/mTOR Dual Inhibitors. Molecules 2018, 23, 1553. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, L.; Xu, S.; Liu, X.; Chen, X.; Xiong, H.; Hou, S. Discovery of thinopyrimidine-triazole conjugates as c-Met targeting and apoptosis inducing agents. Bioorg. Chem. 2018, 77, 370–380. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of the representative EGFR and PI3K inhibitors.
Figure 1. Structures of the representative EGFR and PI3K inhibitors.
Molecules 26 03041 g001
Figure 2. Olmutinib docking model: (A) olmutinib-EGFRT790M (PDB code: 3ika); (B) olmutinib-EGFRWT (PDB code: 4zau).
Figure 2. Olmutinib docking model: (A) olmutinib-EGFRT790M (PDB code: 3ika); (B) olmutinib-EGFRWT (PDB code: 4zau).
Molecules 26 03041 g002
Figure 3. Design strategies for the first series of compounds.
Figure 3. Design strategies for the first series of compounds.
Molecules 26 03041 g003
Figure 4. Design strategies for the second series of compounds.
Figure 4. Design strategies for the second series of compounds.
Molecules 26 03041 g004
Scheme 1. The synthetic route for target compounds H1–H8. Reagents and conditions: (a) 1,2-dimethoxyethane, thiophen-2-yl-boronic acid, 90 °C, 1.5 h; (b) 1,4-dioxane, metanitrophenol, r.t., 4 h; (c) acetonitrile, toluene-p-sulfonic acid, amines, 100 °C, 4–5 h; (d) ethanol, ferric chloride, water and hydrazine, active carbon, 80 °C, 4 h; (e) dichloromethane, acid amides, bicarbonate, 0 °C, 1–2 h.
Scheme 1. The synthetic route for target compounds H1–H8. Reagents and conditions: (a) 1,2-dimethoxyethane, thiophen-2-yl-boronic acid, 90 °C, 1.5 h; (b) 1,4-dioxane, metanitrophenol, r.t., 4 h; (c) acetonitrile, toluene-p-sulfonic acid, amines, 100 °C, 4–5 h; (d) ethanol, ferric chloride, water and hydrazine, active carbon, 80 °C, 4 h; (e) dichloromethane, acid amides, bicarbonate, 0 °C, 1–2 h.
Molecules 26 03041 sch001
Scheme 2. The synthetic route for target compounds H9–H16. Reagents and conditions: (a) acetone, morpholine, 0 °C, 1 h; (b) 1,2-dimethoxyethane, 5-aldyl-2-thiophene boric acid, K2CO3, 75 °C, 24 h; (c) methanol, NaBH4, 0 °C, 0.5 h; (d) DCM, SOCl2, N,N-dimethylformamide, r.t., 2 h; (e) isopropanol, N,N-diisopropylethylamine, amines, 75 °C, 1 h; (f) THF, tert-butoxide, 3-aminophenol, 0 °C, 1 h; (g) dichloromethane, acid amides, bicarbonate, 0 °C, 1–2 h.
Scheme 2. The synthetic route for target compounds H9–H16. Reagents and conditions: (a) acetone, morpholine, 0 °C, 1 h; (b) 1,2-dimethoxyethane, 5-aldyl-2-thiophene boric acid, K2CO3, 75 °C, 24 h; (c) methanol, NaBH4, 0 °C, 0.5 h; (d) DCM, SOCl2, N,N-dimethylformamide, r.t., 2 h; (e) isopropanol, N,N-diisopropylethylamine, amines, 75 °C, 1 h; (f) THF, tert-butoxide, 3-aminophenol, 0 °C, 1 h; (g) dichloromethane, acid amides, bicarbonate, 0 °C, 1–2 h.
Molecules 26 03041 sch002
Figure 5. (A) The binding mode of compound H10 with EGFRT790M (3ika) kinase; (B) H10 and protein cavity binding pattern; (C) The binding mode of compound H10 with PI3Kγ (3L08) kinase.
Figure 5. (A) The binding mode of compound H10 with EGFRT790M (3ika) kinase; (B) H10 and protein cavity binding pattern; (C) The binding mode of compound H10 with PI3Kγ (3L08) kinase.
Molecules 26 03041 g005aMolecules 26 03041 g005b
Table 1. Structures and cytotoxicity of compounds H1–H16.
Table 1. Structures and cytotoxicity of compounds H1–H16.
Molecules 26 03041 i001
CompoundR1R2 IC50(µM) aSelectivity c
A549H1975NCI-H460MCF-7LO2
H1 Molecules 26 03041 i002 Molecules 26 03041 i0034.43 ± 0.509.64 ± 1.0113.09 ± 1.1718.99 ± 1.71>100>22.57
H2 Molecules 26 03041 i004 Molecules 26 03041 i00510.48 ± 1.1712.45 ± 1.0916.39 ± 0.9814.56 ± 1.1987.85 ± 1.678.38
H3 Molecules 26 03041 i006 Molecules 26 03041 i00727.69 ± 1.2421.85 ± 1.3325.79 ± 1.3835.83 ± 1.64>100>3.61
H4 Molecules 26 03041 i008 Molecules 26 03041 i00937.10 ± 1.1925.11 ± 1.3023.38 ± 1.4927.04 ± 1.63>100>2.70
H5 Molecules 26 03041 i010 Molecules 26 03041 i011>100>100>100>100>100
H6 Molecules 26 03041 i012 Molecules 26 03041 i01315.56 ± 0.8111.94 ± 1.1118.09 ± 0.7117.73 ± 1.01>100>6.43
H7 Molecules 26 03041 i014 Molecules 26 03041 i0154.37 ± 0.504.59 ± 0.4613.48 ± 0.5219.21 ± 1.12>100>22.88
H8 Molecules 26 03041 i016 Molecules 26 03041 i01719.01 ± 1.3421.52 ± 1.2124.12 ± 1.2849.13 ± 2.13 >100>5.26
H9 Molecules 26 03041 i018 Molecules 26 03041 i01923.46 ± 1.43 12.82 ± 1.2528.24 ± 1.7853.86 ± 1.73 >100>4.26
H10 Molecules 26 03041 i020 Molecules 26 03041 i0213.36 ± 1.591.16 ± 1.5310.65 ± 2.0213.05 ± 1.36>100>29.76
H11 Molecules 26 03041 i022 Molecules 26 03041 i02313.54 ± 1.135.57 ± 1.5319.72 ± 1.1625.83 ± 1.31>100>7.39
H12 Molecules 26 03041 i024 Molecules 26 03041 i02521.86 ± 1.5413.55 ± 1.9423.81 ± 2.0564.01 ± 1.80>100>4.57
H13 Molecules 26 03041 i026 Molecules 26 03041 i02723.45 ± 1.8921.52 ± 1.2154.53 ± 2.8632.45 ± 1.73>100>4.26
H14 Molecules 26 03041 i028 Molecules 26 03041 i02920.56 ± 2.3017.78 ± 1.3446.22 ± 1.4341.61 ± 1.61>100>4.86
H15 Molecules 26 03041 i030 Molecules 26 03041 i03121.35 ± 1.5819.46 ± 1.5562.43 ± 2.9438.54 ± 1.78>100>4.68
H16 Molecules 26 03041 i032 Molecules 26 03041 i03336.14 ± 1.6115.57 ± 2.5530.28 ± 1.6851.82 ± 1.27>100>2.77
Olmutinib b--4.29 ± 0.210.52 ± 0.105.29 ± 0.5926.90 ± 0.9325.76 ± 1.316.00
a The values are an average of three separate determinations; b used as a positive control; c IC50(LO2)/IC50(A549).
Table 2. Enzymatic activities of selected compounds H7 and H10 against EGFRT790M/L858R and PI3Kα (IC50, µM).
Table 2. Enzymatic activities of selected compounds H7 and H10 against EGFRT790M/L858R and PI3Kα (IC50, µM).
CompoundIC50 (µM) a
EGFRT790M/L858R PI3Kα
H70.63>10
H100.258.56
Olmutinib b0.01>10
a The values are an average of three separate determinations; b used as a positive control.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hu, X.; Tang, S.; Yang, F.; Zheng, P.; Xu, S.; Pan, Q.; Zhu, W. Design, Synthesis, and Antitumor Activity of Olmutinib Derivatives Containing Acrylamide Moiety. Molecules 2021, 26, 3041. https://doi.org/10.3390/molecules26103041

AMA Style

Hu X, Tang S, Yang F, Zheng P, Xu S, Pan Q, Zhu W. Design, Synthesis, and Antitumor Activity of Olmutinib Derivatives Containing Acrylamide Moiety. Molecules. 2021; 26(10):3041. https://doi.org/10.3390/molecules26103041

Chicago/Turabian Style

Hu, Xiaohan, Sheng Tang, Feiyi Yang, Pengwu Zheng, Shan Xu, Qingshan Pan, and Wufu Zhu. 2021. "Design, Synthesis, and Antitumor Activity of Olmutinib Derivatives Containing Acrylamide Moiety" Molecules 26, no. 10: 3041. https://doi.org/10.3390/molecules26103041

Article Metrics

Back to TopTop