Cancer continues to be a leading health hazard for human being due to its high morbidity and mortality rates worldwide [1]. Traditional cancer treatment methods include surgery, radiotherapy and chemotherapy, but in view of their shortcomings in treatment, researchers have been working on developing new cancer treatment methods. During this period, targeted therapy stood out in cancer treatment due to its strong specificity and low side effects [2]. Small molecule kinase inhibitors are a major category of cancer targeted drugs. With the deepening of people’s understanding of genes and carcinogenic molecular mechanisms, scientists have discovered that protein kinases play a key role in the growth, proliferation, differentiation, migration, and apoptosis of tumor cells [2, 3]. It has stimulated people’s enthusiasm for the research and development of small molecule kinase inhibitor drugs. However, from the perspective of current clinical treatment effect, kinase inhibitors have shown good efficacy in the initial treatment of cancer patients. With the passage of treatment time, the vast majority of cancer patients have shown obvious resistance to kinase inhibitors, such as the increase in the dosage of the drug and the decrease in the treatment effect, which seriously affects the treatment index of cancer patients. Therefore, it is necessary to develop new small molecule kinase inhibitors to overcome shortcomings.

Designing drugs for multiple protein kinase targets is a major method to overcome the resistance of kinase inhibitors [4]. This kind of drugs is called multi-target kinase inhibitors. In recent years, molecular hybridization methods have become an important strategy for drug molecular design. Molecular hybridization refers to the combination of two or more pharmacophores with biological activity to form a single molecular structure with higher biological activity [5]. Since these pharmacophores have different mechanisms of action and different binding targets, the drug molecules obtained by combining them will play a role in multiple aspects, thereby reducing the risk of drug resistance [6].

The mechanism of action of most kinase inhibitors is to competitively bind to the ATP-binding site of the N- and C-terminal clefts in the catalytic domain of protein kinases, thereby preventing the phosphorylation of proteins [7, 8]. 7-azaindole is a common pharmacophore with a variety of pharmacological activities, such as anti-microbial, anti-cancer, anti-HIV and anti-influenza [9]. In the 7-azaindole ring, the N on the pyridine structure is the hydrogen bond acceptor and the NH on the pyrrole structure is the hydrogen bond acceptor and donor which will form bidentate hydrogen bonds with the hinge region of the kinase [7, 8]. This enhances the binding ability between kinase and drug molecule. Meanwhile, it’s reported that more than 90 kinds of protein kinases have shown sensitivity to drugs containing 7-azaindole structure [7]. Therefore, 7-azaindole fragments are often introduced into the drug molecule as inhibitors of multiple protein kinases. Especially in the treatment of cancer, 7-azaindole has shown great advantages. Whether it is vemurafenib (BRAF kinase inhibitor) for the treatment of melanoma, or a variety of anti-cancer drugs targeting different kinase targets (such as JAK3 [10], CSF1R [11], STK [12] and ROCK1 [13]) that are currently undergoing clinical research, they are all designed based on the 7-azaindole structure.

1,2,3-Triazole is another important type of nitrogen-containing heterocyclic compound. In terms of structure, 1,2,3-triazole has the ability to form various non-covalent bindings such as hydrogen bonds, Π–Π stacking effect, hydrophobic interaction, van der Waals force and dipole-dipole interaction and it is less toxic and has stability under physiological conditions, which induces it being abundant in pharmacological activity [6, 1416]. The antibacterial [17], antimalarial [18], antifungal [19], antiviral [20], antitubercular [21], anti-HIV [22], and anticancer [23] activities of 1,2,3-triazole make it play an important role in a variety of drugs. On the anticancer activity, 1,2,3-triazole works by inhibiting different enzymes such as carbonic anhydrases (CAs) [24], thymidylate synthase (TS) [25], aromatase [26], tryptophan, 2, 3-dioxygenase (TDO) [27], vascular endothelial growth factor receptor (VEGFR) [28], and epidermal growth factor receptor (EGFR) [29]. Moreover, some of the azole derivatives such as cefatrizine, carboxyamidotriazole, anastrozole, and letrozole have already been used in clinics or are under clinical evaluation for the treatment of cancers, revealing their potential as putative anticancer drugs [6, 15].

Based on the above, a series of compounds containing 7-azaindole and 1,2,3-triazole fragments were designed and synthesized. Subsequently, these compounds were tested for their anticancer activity in order to find a better anticancer structure.

RESULTS AND DISCUSSION

Chemistry. In this study, the target compounds 3a3r were synthesized by a three-step reaction (Scheme 1). The first step is to synthesize compound 1 (4-chloro-1-(prop-2-yn-1-yl)-1H-pyrrolo[2,3-b]pyridine) by substitution reaction. In the second step, compounds 2a2r were synthesized by amide condensation reaction. In the last step, the target compounds 3a3r were synthesized by utilizing the azide-alkyne cycloaddition reaction of appropriate precursors 1 and 2a2r in the presence of CuSO4·5H2O/sodium ascorbate in THF : H2O (1 : 1) mixture. The azide-alkyne cycloaddition reaction is known as the click reaction and it need to be catalyzed by Cu(I) ions. This reaction has many advantages such as good selectivity, strong stereospecificity [30], mild reaction conditions and high yield. Therefore, it is widely used in the regio- and stereo- specific synthesis of 1,4-substituted-1,2,3-triazoles which is a structure with various pharmacological activity.

Scheme 1.
scheme 1

Synthesis of the target compounds 3a3r.

Through the above reaction, we obtained a series of hybrid compounds 3a3r containing 7-azaindole structure and 1,2,3-triazole structure (see Table 1 for specific structure). Then the 1H NMR spectra, 13C NMR spectra and Mass spectra analysis test methods had been used to characterize the structures of the compounds and characterization results had confirmed the chemical structures of the target compounds 3a3r.

Table 1. The structure of compound 3a3r

In vitro anticancer activity. The prepared target compounds 3a3r were further screened for their potential that inhibits the growth of cancer cells against lung cancer (A549) and hepatic cancer (HepG2) cell lines by MTT colorimetric assay [31]. In this study, Gefitinib and Sorafenib were used as positive control drugs for lung cancer and hepatic cancer cells, respectively. And the IC50 values which are some different concentrations of the compounds that induces 50% death of the cancer cells of the test compounds were determined from dose-response curves and the results are summarized in Table 2. And the inversely proportional relationship between the ability of the sample to inhibit cancer growth and the IC50 values.

Table 2. In vitro anticancer activity of the target compounds 3a3r and the positive control drugs. Values are mean ± SD expressed in µM

The results presented in Table 2 reveal that the synthetic series of compounds 3a3r have shown an obvious inhibitory effect on the growth of lung cancer cell (A549) and hepatic cancer cell (HepG2). Among these compounds, compound 3l exhibits the strongest inhibitory effect on cancer cells A549 and HepG2, and its IC50 values are 7.58 ± 0.87 μM and 6.69 ± 0.69 μM, respectively, while compound 3g exhibits the weakest inhibitory effect, and its IC50 values for the two cancer cells are 26.58 ± 1.54 μM and 24.37 ± 2.39 μM, respectively. At the same time, compared with the positive control, although the IC50 values of compounds 3d (7.83 ± 0.36 μM; 6.98 ± 0.19 μM), 3l, 3q (9.47 ± 0.96 μM; 10.18 ± 0.57 μM) and 3r (8.65 ± 0.82 μM; 7.97 ± 0.43 μM) are larger than the positive control (IC50 of Gefitinib for A549 = 6.77 ± 0.65 μM; IC50 of Sorafenib for HepG2 = 5.65 ± 0.71 μM), their IC50 values are all less than 10 μM. Therefore, the inhibitory effects of these four compounds on cancer cells are at the same level of inhibition as the positive control, which indicates that compounds 3d, 3l, 3q and 3r have greater anti-cancer activity potential. However, the IC50 values of compounds 3g is more than 20 μM, which suggests that there is little anticancer activity potential of compound 3g. In addition, the IC50 values of other compounds are between 10 and 20 μM, which reveals that they also have an inhibitory effect on cancer cells, but the inhibitory effect is worse than that of the positive control and compounds 3d, 3l, 3q, and 3r. The possible reason for the better activity of compounds 3d, 3l, 3q, and 3r is that they bind to multiple protein kinase targets in lung cancer cells A549 and hepatic cancer cells HepG2, thereby inhibiting the growth and reproduction of cancer cells.

Molecular docking studies. Tyrosine kinase is one of the main members of the protein kinase family. Because it can participate in the regulation of various physiological processes of cancer cells, including growth, proliferation, differentiation, metastasis and apoptosis, it is often used as a drug target for cancer treatment. In order to further investigate the possible binding modes and corresponding interaction energies between the synthesized compounds 3a3r and various tyrosine kinase targets in cancer cells, we docked the synthesized compounds 3a3r with CSF1R (PDB: 4R7H), JAK1 (PDB: 3EYG), JAK2 (PDB: 3TJD), JAK3 (PDB: 3LXK), EGFR1 [32] (PDB: 4WKQ) and VEGFR2 [33] (PDB: 3WZD, 3WZE, 4AGD) targets, respectively. The lowest binding energies between compounds and different targets were obtained, as shown in Table 3.

Table 3. The lowest binding energies of compounds 3a3r with different protein targets

As we can see from the docking results, on the one hand, the synthesized series of compounds have weak binding ability against 3TJD (JAK2 target) and the lowest binding energy ranges from –3.20 to –5.03 kcal/mol, while the synthesized compounds have strong binding ability against 3LXK (JAK3 target), 3WZE (VEGFR2 target) and 4AGD (VEGFR2 target) and the lowest binding energy is approximately around –6 and –7 kcal/mol. That is to say, the synthesized series of compounds have the best selectivity for protein targets 3LXK, 3WZE, and 4AGD, and the worst selectivity for protein target 3TJD. On the other hand, among all the compounds, the lowest binding energy values of compound 3l against protein targets 3EYG, 3LXK, 3WZE, and 4AGD are the smallest. In addition, the lowest binding energy values of compound 3l against other targets are also in the range of –6 to –7 kcal/mol. Therefore, the binding ability between compound 3l and the selected protein targets is stronger than that of other compounds on the whole. On the contrary, the lowest binding energy values of compound 3g against every selected protein targets are larger than that of other compounds. So there is weak binding ability between compound 3g and the selected protein targets.

Moreover, comparing the lowest binding energy data with the anticancer activity data of compounds, it can be found that the smaller the lowest binding energy data between the compound and each target, the better the anticancer activity it shown. Therefore, the lowest binding energy data can be used to explain the anticancer activity of compounds.

In order to more clearly observe the interactions between compounds 3g, 3l and protein targets 3LXK, 3WZE, 4AGD, respectively, the 2D and 3D binding model diagrams of compounds 3g, 3l with the protein targets (3LXK, 4AGD, 3WZE) were made by drawing software (Discovery Studio) and the results were shown in Fig. 1.

Fig. 1.
figure 1

The 2D and 3D binding model diagrams of compounds 3g, 3l against the protein targets (3LXK, 4AGD, 3WZE). (a) Compound 3l with the protein target 3LXK. (b) Compound 3l with the protein target 4AGD. (c) Compound 3l with the protein target 3WZE. (d) Compound 3g with the protein target 3LXK. (e) Compound 3g with the protein target 4AGD. (f) Compound 3g with the protein target 3WZE.

In the interaction between drug molecules and protein targets, hydrogen bonding is the most important mode of action, and its number determines the binding ability of drug molecules against protein targets. Furthermore, hydrophobic bonds are another important role in the interaction. When the number of hydrogen bonds is the same, the binding ability of different drug molecules with protein targets is mainly affected by hydrophobic bonds. According to the above content, analyzing Fig. 1 can find that in the binding of compound 3l with protein targets 3LXK, 4AGD and 3WZE, we can clearly see the formation of two hydrogen bonds between compound and the protein target, while compound 3g forms only one hydrogen bond with them respectively. Therefore, from the perspective of the number of hydrogen bonds, the binding ability of compound 3l with the three targets is stronger than that of compound 3g. Meanwhile, the hydrogen bond distance between compound 3l and the three targets is relatively short, with an average length of 2.54 Å, which is much smaller than 3.5 Å of the traditional hydrogen bond. This further indicates that compound 3l has strong binding ability with the three targets. Besides, we can also see from the figure that compound 3l interacts with many amino acid residues on the three protein targets, thus forming a strong hydrophobic interaction force. And the presence of these strong interactions can effectively improve the stability of the compound 3l with the active pocket on the protein target, so compound 3l is a potentially active small molecule for the three protein targets.

In contrast, in the binding of compound 3g with the three protein targets, both the number of hydrogen bonds and the number of hydrophobic bonds are less than those of compound 3l, which implies that compound 3g has a weak inhibitory effect on the three selected protein targets.

Why is there such a difference in the binding of compounds 3l and 3g with protein targets? Based on the structure of the compounds, we infer that compound 3g can not be better embedded into the protein targets due to the presence of a long alkyl structure in the compound 3g. As a result, the strong interaction between the protein and the target can’t be formed, resulting in the decreased inhibitory activity of compound 3g on the targets.

EXPERIMENTAL

Starting materials and solvents were purchased from Aladdin, Macklin, Adamas, Bidepharm and were used without purification. Compound 1 and 2a2r were purified by column chromatography before being used for the next reaction. Reaction progress were monitored by thin layer chromatography on silica gel 60 GF254 (Qingdao Haiyang Chemical Co., Ltd., China). Melting points were determined on a χ-4 precision micro melting point apparatus and were uncorrected. 1H NMR and 13C NMR were recorded on Avance AV-400 spectrometer (Bruker, Germany, 400 and 101 MHz). And tetramethylsilane (TMS) was used as an internal standard for spectra obtained in DMSO-d6 and CDCl3 solvent. Chemical shifts are given in parts per million (ppm). Mass spectra were recorded on Agilent 6224 instrument.

General procedure for the synthesis of 4-chloro-1-(prop-2-yn-1-yl)-1H-pyrrolo[2,3-b]pyridine (1). To 4-chloro-1H-indole (13.11 mmol) dissolved in DMF (30 mL) anhydrous potassium carbonate (39.33 mmol) was added. Then, 3-bromopropyne (15.73 mmol) was slowly added dropwise to the mixed system while stirring. The mixture was heated at 35°C for 12 h. Reaction was monitored by TLC until TLC showed that the reactants were completely consumed, eluent (PE–EA, 2 : 1). The cooled mixture was partitioned with Ethyl acetate (3×20 mL). The combined organic extracts were washed with brine, dried over Na2SO4 and evaporated in vacuo. The residue was purified on a short SiO2 column using the eluent PE–EA (4 : 1) to give the desired compound 1.

General procedure for the synthesis of compound 2a2r. To 4-azidobenzoic acid (1.20 mmol) dissolved in DMF (30 mL) HATU (1.44 mmol), DIPEA (2.40 mmol) and substituted aromatic amine (1.20 mmol) were added. The mixture was warmed and held at 35–50°C under N2 with stirring for 6–7 h. When TLC showed the reaction was complete, ethyl acetate (3×20 mL) and water (20 mL) was used to extract the solution containing product. Finally, the resulting organic phases was washed with brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure. The crude product was purified by silica gel column chromatography using the eluent PE–EA (8 : 1) to give the desired compound 2a2r.

General procedure for the synthesis of target compound 3a3r. To compound 1 (0.74 mmol) dissolved in THF (20 mL) anhydrous copper sulfate (0.22 mmol) and sodium ascorbate (0.37 mmol) dissolved in water (5 mL) as catalyst were added. Then, compound 2a2r (0.74 mmol) was dropped into the mixture with stirring. The mixture was heated and refluxed at 50°C under N2 for 9–10 h. The reaction was monitored by TLC. After the completion of reaction, the solution was filtered and the filtrate was portioned between ethyl acetate (3×20 mL) and water (20 mL) and the combined extracts were dried (Na2SO4), filtered and evaporated to dryness. The residue was purified on a short column of silica gel, using CH2Cl2–MeOH (160 : 1) to give the pure desired target compound 3a3r. And these obtained compounds were characterized by 1H NMR spectra, 13C NMR spectra and MS.

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-fluorobenzyl)benzamide (3a). Compound 3a was obtained from the reaction of compound 1 with compound 2a. Yield: 52%, white solid; mp 255–256°C. 1H NMR (400MHz, DMSO-d6): δ, ppm: 9.27 t (J = 5.8 Hz, 1H, NH), 8.91 s (1H, C9H), 8.32 d (J = 5.2 Hz, 1H,Ar-H), 8.13 d (J = 8.7 Hz, 2H, Ar-H), 8.06 d (J = 8.6 Hz, 2H, Ar-H), 7.86 d (J = 3.5 Hz, 1H, Ar-H), 7.43 d. d (J = 8.1, 5.9 Hz, 2H, Ar-H), 7.33 d (J = 5.1 Hz, 1H, C7H), 7.26–7.17 m (2H, Ar-H), 6.66 d (J = 3.5 Hz, 1H, C6H), 5.74 s (2H, C8H2), 4.53 d (J = 5.8 Hz, 2H, C24H2) . 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.02 (C-17), 162.36 (C-22), 159.95 (C-5), 153.02 (C-3), 147.51 (C-16), 144.66 (C-1), 143.31 (C-15), 138.30 (C-10), 135.65 (C-23), 134.39 (C-18,19), 129.29 (C-12,14), 122.95 (C-11,13), 121.73 (C-7), 119.60 (C-4), 118.88 (C-9), 118.81 (C-2), 116.98 (C-20,21), 115.90 (C-6), 57.88 (C-8), 42.01 (C-24). MS (ESI, m/z): 461.1 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-chlorobenzyl)benzamide (3b). Compound 3b was obtained from the reaction of compound 1 with compound 2b. Yield: 51.30%, white solid; mp 265–267°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 9.26 t (1H, NH), 8.88 s (1H, C9H), 8.28 d (J = 3.9 Hz, 1H, Ar-H), 8.09 d (J = 7.4 Hz, 2H, Ar-H), 8.03 d (J = 7.7 Hz, 2H, Ar-H), 7.82 s (1H, Ar-H), 7.39 d (J = 8.5 Hz, 4H, Ar-H), 7.30 d (J = 4.0 Hz, 1H,C7H), 6.59 d (J = 22.9 Hz, 1H, C6H), 5.70 s (2H, C8H2), 4.50 d (J = 4.2 Hz, 2H, C24H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.09 (C-17), 147.52 (C-5), 144.67 (C-3), 143.31 (C-16), 138.51 (C-23), 138.33 (C-18,19), 134.40 (C-1), 133.92 (C-15), 131.34 (C-10), 130.15 (C-22), 129.14 (C-20,21), 128.96 (C-12,14), 122.25 (C-11,13), 121.74 (C-7), 119.62 (C-4), 118.89 (C-9), 115.82

N-(4-Bromobenzyl)-4-(4-((4-chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)benzamide (3c). Compound 3c was obtained from the reaction of compound 1 with compound 2c. Yield: 48.82%, white solid; mp 247–248°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 8.87 t (1H, NH), 8.28 s (1H, C9H), 8.16-7.98 m (4H, Ar-H), 7.82 d (J = 3.5 Hz, 1H, Ar-H), 7.72 d (J = 8.4 Hz, 1H, Ar-H), 7.57– 7.51 m (2H,Ar-H), 7.37–7.20 m (3H, Ar-H, C7H), 6.59 d (J = 23.8, 3.5 Hz, 1H, C6H), 5.73 s (2H, C8H2), 4.48 d (J = 5.2 Hz, 2H, C24H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.09 (C-17), 147.52 (C-5), 144.67 (C-3), 143.31 (C-16), 138.93 (C-23), 138.33 (C-1), 134.39 (C-15), 133.91 (C-10), 131.16 (C-20,21), 130.14 (C-18,19), 129.51 (C-12,14), 128.95 (C-22), 124.90 (C-4), 122.52 (C-11,13), 121.74 (C-7), 119.80 (C-9), 119.62 (C-2), 115.81 (C-6), 57.89 (C-8), 42.14 (C-24). MS (ESI, m/z): 521.0 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-cyanobenzyl)benzamide (3d). Compound 3d was obtained from the reaction of compound 1 with compound 2d. Yield: 55.32%, white solid; mp 289–291°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 9.34 t (J = 5.9 Hz, 1H, NH), 8.88 s (1H, C9H), 8.31–8.25 m (1H, Ar-H), 8.11 t (J = 7.5 Hz, 2H, Ar-H), 8.0 d (J = 8.7 Hz, 2H, Ar-H), 7.83 d. d (J = 10.9, 6.4 Hz, 3H, Ar-H), 7.55 t (J = 7.8 Hz, 2H, Ar-H), 7.27 t (J = 9.3 Hz, 1H,C7H), 6.60 d (J = 16.8 Hz, 1H, C6H), 5.71 s (2H, C8H2), 4.61 d (J = 10.3 Hz, 2H, C24H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.27 (C-17), 147.50 (C-5), 145.33 (C-3), 144.67 (C-23), 143.27 (C-16), 138.40 (C-1), 134.41 (C-15), 133.72 (C-10), 132.26 (C-20,21), 130.07 (C-18,19), 128.99 (C-12,14), 122.04 (C-11,13), 121.72 (C-7), 119.61 (C-4), 118.89 (C-9), 118.45 (C-25), 115.79 (C-2), 115.21 (C-22), 114.87 (C-6), 59.73 (C-8), 42.54 (C-24). MS (ESI, m/z): 469.1 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(pyridin-4-ylmethyl)benzamide (3e). Compound 3e was obtained from the reaction of compound 1 with compound 2e. Yield: 53.02%, yellow solid; mp 298–299°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 9.26 t (J = 5.9 Hz, 1H, NH), 8.87 s (1H, C9H), 8.58 s (1H, Ar-H), 8.48 d (J = 3.8 Hz, 1H, Ar-H), 8.27 d (J = 5.2 Hz, 1H, Ar-H), 8.09 d (J = 8.8 Hz, 2H, Ar-H), 8.02 d (J = 8.8 Hz, 2H, Ar-H), 7.78 d. d (J = 22.6, 5.7 Hz, 2H, Ar-H), 7.38 d. d (J = 7.7, 4.8 Hz, 1H, Ar-H), 7.28 d (J = 5.2 Hz, 1H, C7H), 6.61 d (J = 3.6 Hz, 1H, C6H), 5.70 s (2H, C8H2), 4.53 d (J = 5.8 Hz, 2H, C23H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.70 (C-17), 149.35 (C-20, 21), 148.62 (C-5), 148.02 (C-22), 145.16 (C-3), 143.80 (C-16), 138.85 (C-1), 135.67 (C-15), 135.38 (C-10), 134.89 (C-12,14), 134.36 (C-11,13), 130.63 (C-18,19), 129.47 (C-4), 123.98 (C-9), 121.75(C-7) , 119.39 (C-2), 116.30 (C-6), 58.39 (C-8), 42.09 (C-23). MS (ESI, m/z): 444.0 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(thiophen-3-ylmethyl)benzamide (3f). Compound 3f was obtained from the reaction of compound 1 with compound 2f. Yield: 65.34%, yellow solid; mp 243–245°C. 1H NMR spectrum (400 MHz, CDCl3) δ, ppm: 8.23 t (J = 5.4 Hz, 1H, NH), 7.95 s (1H, C9H), 7.94–7.84 m (2H, Ar-H), 7.79–7.70 m (2H, Ar-H), 7.46 d (J = 3.5 Hz, 1H, Ar-H), 7.32 d. d (J = 12.4, 9.1 Hz, 2H, Ar-H), 7.25 d (J = 13.7 Hz, 1H, Ar-H), 7.12 t (J = 6.1 Hz, 1H, Ar-H), 7.03 t (J = 8.5 Hz, 1H, C7H), 6.61 d (J = 5.9 Hz, 1H, C6H), 5.62 s (2H, C8H2), 4.63 d (J = 16.1, 5.5 Hz, 2H, C22H2). 13C NMR spectrum (101 MHz, CDCl3) δC, ppm: 165.87 (C-17), 163.56 (C-5), 147.81 (C-3), 145.20 (C-16), 143.34 (C-21), 138.78 (C-1), 136.32 (C-15), 134.41 (C-10), 129.75 (C-18), 128.71 (C-12,14), 128.53 (C-19), 122.43 (C-11,13), 122.88 (C-7), 120.59 (C-4), 120.23 (C-20), 120.05 (C-9), 116.32 (C-2), 115.83 (C-6), 59.13 (C-8), 43.58 (C-22).- MS (ESI, m/z):449.0 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(3-morpholinopropyl)benzamide (3g). Compound 3g was obtained from the reaction of compound 1 with compound 2g. Yield: 67.54%, white solid; mp 260–261°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 8.84 t (J = 5.7 Hz, 1H, NH), 8.60 s (1H, C9H), 8.28 d (J = 5.1 Hz, 1H, Ar-H), 8.04 d (J = 8.8 Hz, 2H, Ar-H), 8.00 d (J = 8.7 Hz, 2H, Ar-H), 7.80 d (J = 3.5 Hz, 1H, Ar-H), 7.29 d (J = 5.1 Hz, 1H, C7H), 6.60 d (J = 11.9 Hz, 1H, C6H), 5.67 s (2H, C8H2), 3.61–3.55 m (4H,C18H2, C20H2), 2.44–2.23 m (8H, C21H2-C24H2), 1.76–1.69 m (2H, C19H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.43 (C-17), 148.04 (C-5), 145.12 (C-3), 143.80 (C-16), 138.63 (C-1), 134.90 (C-15), 130.61 (C-10), 129.28 (C-12,14), 122.22 (C-11,13), 121.06 (C-7), 120.39 (C-4), 119.40 (C-9), 116.30 (C-2), 115,43 (C-6), 98.39 (C-23,24), 66.67 (C-21,22), 56.52 (C-20), 53.81 (C-8), 38.34 (C-18), 26.37 (C-19). MS (ESI, m/z): 480.2 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-fluorophenyl)benzamide (3h). Compound 3h was obtained from the reaction of compound 1 with compound 2h. Yield: 69.63%, white solid; mp 228–230°C. 1H NMR spectrum (600 MHz, DMSO-d6), δ, ppm: 10.45 s (J = 24.2 Hz, 1H, NH), 8.89 s (1H, C9H), 8.26 t (J = 5.3 Hz, 1H, Ar-H), 8.14 d (J = 8.7 Hz, 2H, Ar-H), 8.07 d (J = 8.7 Hz, 2H, Ar-H), 7.83–7.75 m (3H, Ar-H), 7.28 d (J = 5.1 Hz, 1H, C7H), 7.21 t (J = 8.9 Hz, 2H, Ar-H), 6.61 d (J = 5.3 Hz, 1H, C6H), 5.68 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.22 (C-17), 159.58 (C-22), 157.19 (C-5), 147.52 (C-3), 144.68 (C-16), 143.29 (C-1), 138.63 (C-15), 138.50 (C-23), 135.30 (C-10), 134.42 (C-12,14), 130.10 (C-18,19), 122.39 (C-11,13), 121.77 (C-7), 120.34 (C-4), 119.59 (C-9), 115.80 (C-2), 115.31 (C-20,21), 115.09 (C-6), 57.90 (C-8). MS (ESI, m/z): 444.0 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-chlorophenyl)benzamide (3i). Compound 3i was obtained from the reaction of compound 1 with compound 2i. Yield: 58.0%, white solid; mp 222-223°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.24 s (J = 11.7 Hz, 1H, NH), 8.89 s (1H, C9H), 8.27 d (J = 5.1 Hz, 1H,Ar-H), 8.18 d (J = 8.7 Hz, 2H, Ar-H), 8.07 d (J = 8.7 Hz, 2H, Ar-H), 7.81 d (J = 3.6 Hz, 1H, Ar-H), 7.62–7.52 m (2H, Ar-H), 7.41 t (J = 7.1 Hz, 1H, Ar-H), 7.36 s (1H, Ar-H), 7.25 s (1H, C7H), 6.61 d (J = 3.6 Hz, 1H, C6H), 5.70 s 2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.28 (C-17), 147.53 (C-5), 144.73 (C-3), 143.33 (C-16), 138.68 (C-23), 134.85 (C-1), 134.41 (C-15), 133.67 (C-10), 130.17 (C-22), 129.70 (C-21), 129.59 (C-18), 129.45 (C-12,14), 128.66 (C-19), 127.72 (C-20), 122.52 (C-11,13), 121.79 (C-7), 119.73 (C-4), 118.90 (C-9), 116.83 (C-2), 115.43 (C-6), 57.91 (C-8). MS (ESI, m/z): 443.0 ([M + H]+) .

N-(3-Bromophenyl)-4-(4-((4-chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)benzamide (3j). Compound 3j was obtained from the reaction of compound 1 with compound 2j. Yield: 52.0%, yellow solid; mp 235–237°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.53 s (1H, NH), 8.92 s (1H, C9H), 8.28 s (1H, Ar-H), 8.16 t (J = 9.4 Hz, 3H, Ar-H), 8.09 d (J = 8.6 Hz, 2H, Ar-H), 7.81 d. d (J = 11.4, 5.6 Hz, 2H, Ar-H), 7.37–7.23 m (3H, Ar-H, C7H), 6.62 d (J = 3.5 Hz, 1H, C6H), 5.72 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.51 (C-17), 147.52 (C-5), 143.27 (C-3), 140.58 (C-16), 138.64 (C-23), 134.82 (C-1), 134.42 (C-15), 134.18 (C-10), 130.61 (C-20), 130.09 (C-12,14), 129.48 (C-22), 126.40 (C-21), 122.65 (C-11,13), 121.82 (C-4), 121.41 (C-7), 119.61 (C-18,19), 119.07 (C-9), 118.94 (C-2), 115.83 (C-6), 57.91 (C-8). MS (ESI, m/z): 508.9([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(3,4-dichlorophenyl)benzamide (3k). Compound 3k was obtained from the reaction of compound 1 with compound 2k. Yield: 46.03%, white solid; mp 280–281°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.56 s (1H, NH), 8.90 s (1H, C9H), 8.27 d (J = 5.1 Hz, 1H, Ar-H), 8.14 d (J = 8.8 Hz, 2H, Ar-H), 8.10–8.07 m (3H, Ar-H), 7.81 d (J = 3.6 Hz, 1H, Ar-H), 7.76–7.71 m (1H, Ar-H), 7.44 t (J = 9.1 Hz, 1H, Ar-H), 7.27 d. d (J = 19.7, 5.1 Hz, 1H, C7H), 6.61 d (J = 6.7 Hz, 1H, C6H), 5.69 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.22 (C-17), 159.58 (C-5), 157.19 (C-3), 147.52 (C-16), 144.68 (C-23), 143.29 (C-1), 138.50 (C-15), 135.30 (C-10), 135.28 (C-21), 134.45 (C-19), 134.42 (C-22), 130.10 (C-12,14), 129.39 (C-18), 122.34 (C-11,13), 121.77 (C-7), 119.59 (C-4), 118.90 (C-20), 118.80 (C-9), 118.31 (C-2), 115.09 (C-6), 57.90 (C-8). MS (ESI, m/z): 498.9 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-chloro-3-fluorophenyl)benzamide (3l). Compound 3l was obtained from the reaction of compound 1 with compound 2l. Yield: 48.33%, white solid; mp 275–276°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.56 s (1H, NH), 8.90 s (1H, C9H), 8.27 d (J = 5.1 Hz, 1H, Ar-H), 8.14 d (J = 8.8 Hz, 2H, Ar-H), 8.10–8.07 m (3H, Ar-H), 7.81 d (J = 3.6 Hz, 1H, Ar-H), 7.73 d (J = 6.4 Hz, 1H, Ar-H), 7.44 t (J = 9.1 Hz, 1H, Ar-H), 7.27 d. d (J = 19.7, 5.1 Hz, 1H, C7H), 6.61 d (J = 6.7 Hz, 1H, C6H), 5.70 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.40 (C-17), 154.65 (C-21), 152.23 (C-5), 147.46 (C-3), 143.32 (C-16), 138.63 (C-23), 136.16 (C-1), 134.50 (C-15), 134.09 (C-10), 130.14 (C-20), 129.43 (C-12,14), 122.21 (11,13), 121.88 (C-7), 121.81 (C-4), 120.69 (C-18), 119.66 (C-9), 118.90 (C-22), 118.87 (C-2), 116.88 (C-19), 116.66 (C-6), 57.92 (C-8). MS (ESI, m/z): 498.9 ([M + H]+).

N-(4-Bromo-2-fluorophenyl)-4-(4-((4-chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)benzamide (3m). Compound 3m was obtained from the reaction of compound 1 with compound 2m. Yield: 65.0%, white solid; mp 283–284°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.33 s (1H, NH), 8.88 s (1H, C9H), 8.27 d (J = 5.2 Hz, 1H, Ar-H), 8.16 d (J = 8.7 Hz, 2H, Ar-H), 8.07 d (J = 8.7 Hz, 2H, Ar-H), 7.80 d (J = 3.6 Hz, 1H, Ar-H), 7.68–7.57 m (2H, Ar-H), 7.46 d (J = 8.5 Hz, 1H, Ar-H), 7.26 t (J = 9.9 Hz, 1H, C7H), 6.61 d (J = 3.6 Hz, 1H, C6H), 5.70 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.30 (C-19), 156.83 (C-17), 154.33 (C-5), 147.53 (C-3), 144.74 (C-16), 143.32 (C-1), 138.75 (C-15), 134.41 (C-10), 133.37 (C-12,14), 130.14 (C-20), 129.60 (C-18), 122.51 (C-11,13), 121.78 (C-7), 120.66 (C-4), 120.38 (C-21), 119.90 (C-22), 119.80 (C-9), 118.64 (C-23), 118.52 (C-2), 116.93 (C-6) , 53.58 (C-8). MS (ESI, m/z): 482.1 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(m-tolyl)benzamide (3n). Compound 3n was obtained from the reaction of compound 1 with compound 2n. Yield: 62.68%, yellow solid; mp 287–288°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.31 s (1H, NH), 8.90 s (1H, C9H), 8.28 d (J = 5.2 Hz, 1H, Ar-H), 8.16 d (J = 8.7 Hz, 2H, Ar-H), 8.07 d (J = 8.7 Hz, 2H, Ar-H), 7.81 t (J = 5.5 Hz, 1H, Ar-H), 7.60 d. d (J = 19.5, 11.3 Hz, 2H, Ar-H), 7.35 s (1H, Ar-H), 7.19 s (1H, Ar-H), 6.95 d (J = 7.5 Hz, 1H, C7H), 6.62 d (J = 3.5 Hz, 1H, C6H), 5.70 s (2H, C8H2), 2.31 d (J = 11.2 Hz, 3H, C24H3). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.71 (C-17), 148.03 (C-5), 145.18 (C-3), 143.81 (C-6), 139.36 (C-21), 138.94 (C-23), 138.28 (C-1), 135.19 (C-15), 134.91 (C-10), 130.64 (C-20), 129.88 (C-12,14), 128.96 (C-19), 125.04 (C-22), 122.28 (C-11,13), 121.47 (C-7), 120.09 (C-4), 119.40 (C-9), 119.12 (C-18), 118.32 (C-2), 116.89 (C-6), 58.40 (C-8), 21.68 (C-24). MS (ESI, m/z): 526.9 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(3,5-dimethylphenyl)benzamide (3o). Compound 3o was obtained from the reaction of compound 1 with compound 2o. Yield: 55.0%, white solid; mp 249–251°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.20 s (1H,NH), 8.88 s (1H, C9H), 8.28 d (J = 5.2 Hz, 1H, Ar-H), 8.15 d (J = 8.8 Hz, 2H, Ar-H), 8.06 d (J = 8.8 Hz, 2H, Ar-H), 7.81 d (J = 3.6 Hz, 1H, Ar-H), 7.43 s (2H, Ar-H), 7.28 d (J = 5.2 Hz, 1H, Ar-H), 6.77 s (1H, C7H), 6.62 d (J = 3.6 Hz, 1H, C6H), 5.71 s (2H, C8H2), 2.28 s (6H, C24H3, C25H3). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm:164.14 (C-17), 147.53 (C-5), 144.68 (C-3), 143.31 (C-16), 138.77 (C-20,21), 138.41 (C-23), 137.56 (C-1), 134.73 (C-15), 134.42 (C-10), 130.13 (C-12,14), 129.35 (C-22), 122.37 (C-11,13), 122.13 (C-18,19) 121.78 (C-7), 119.59 (C-4), 118.91 (C-9), 118.22 (C-2), 115.82 (C-6), 57.91 (C-8), 21.10 (C-24,25). MS (ESI, m/z): 457.1 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(2-ethylphenyl)benzamide (3p). Compound 3p was obtained from the reaction of compound 1 with compound 2p. Yield: 62.0%, white solid; mp 241–242°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.08 s (1H, NH), 8.91 s (1H, C9H), 8.29 d (J = 4.8 Hz, 1H, Ar-H), 8.19 d (J = 8.5 Hz, 2H, Ar-H), 8.08 d (J = 8.6 Hz, 2H, Ar-H), 7.83 d (J = 3.5 Hz, 1H, Ar-H), 7.29 ddd (J = 14.6, 9.0, 5.2 Hz, 5H, Ar-H, C7H), 6.63 d (J = 3.5 Hz, 1H, C6H), 5.72 s (2H, C8H2), 2.67 d. d (J = 15.7, 8.2 Hz, 2H, C24H2), 1.16 t (J = 7.5 Hz, 3H, C25H3). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 164.54 (C-17), 147.54 (C-5), 144.72 (C-3), 143.31 (C-16), 139.89 (C-1), 138.48 (C-15), 138.22 (C-10), 135.53 (C-19), 134.42 (C-23), 134.25 (C-22), 130.14 (C-12,14), 129.32 (C-20), 128.52 (C-21) 122.52 (C-11,13), 121.80 (C-7), 119.68 (C-4), 118.92 (C-9), 118.82 (C-2), 118.34 (C-18), 116.80 (C-6), 57.91 (C-8), 23.95 (C-24), 14.13 (C-25). MS (ESI, m/z):457.1 ([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(4-methoxyphenyl)benzamide (3q). Compound 3q was obtained from the reaction of compound 1 with compound 2q. Yield: 45.22%, white solid; mp 270–272°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm: 10.23 s (1H, NH), 8.90 s (1H, C9H), 8.28 d (J = 5.2 Hz, 1H, Ar-H), 8.16 d (J = 8.6 Hz, 2H, Ar-H), 8.06 d (J = 8.5 Hz, 2H, Ar-H), 7.79 t (J = 5.3 Hz, 1H, Ar-H), 7.59 d. d (J = 19.8, 11.3 Hz, 2H, Ar-H), 7.36–7.20 m (2H, Ar-H), 6.99 d (J = 7.4 Hz, 1H, C7H), 6.65 d (J = 3.4 Hz, 1H, C6H), 5.70 s (2H, C8H2), 3.81 s (3H, C24H3). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm:167.45 (C-17), 164.30 (C-22), 156.22 (C-5), 148.03 (C-3), 144.98 (C-16), 135.67 (C-1), 134.89 (C-15), 132.48 (C-10), 132.19 (C-23), 130.27 (C-12,14), 122.48 (C-11,13), 122.27 (C-18,19), 121.06 (C-7), 120.63 (C-4), 119.25 (C-9), 118.28 (C-2), 116.90 (C-20,21), 116.83 (C-6), 58.91 (C-8), 55.09 (C-24). MS (ESI, m/z): 459.0([M + H]+).

4-(4-((4-Chloro-1H-pyrrolo[2,3-b]pyridin-1-yl)methyl)-1H-1,2,3-triazol-1-yl)-N-(pyridin-4-yl)benzamide (3r). Compound 3r was obtained from the reaction of compound 1 with compound 2r. Yield: 49.0%, yellow solid; mp 234–236°C. 1H NMR spectrum (400 MHz, DMSO-d6), δ, ppm:10.70 s (1H, NH), 8.89 s (1H, C9H), 8.27 d (J = 5.1 Hz, 1H, Ar-H), 8.17 d (J = 8.7 Hz, 2H, Ar-H), 8.04 d (J = 8.6 Hz, 3H, Ar-H), 7.80 d (J = 3.5 Hz, 2H, Ar-H), 7.28 t (J = 6.1 Hz, 3H, Ar-H, C7H), 6.61 d (J = 3.5 Hz, 1H, C6H), 5.70 s (2H, C8H2). 13C NMR spectrum (101 MHz, DMSO-d6), δC, ppm: 165.38 (C-17), 165.26 (C-22), 147.57 (C-20,21), 144.71 (C-5), 143.31 (C-3), 138.86 (C-16), 134.42 (C-1), 133.92 (C-15), 130.55 (C-10), 130.12 (C-12,14), 122.85 (C-11,13), 121.82 (C-7), 119.70 (C-4), 119.08 (C-9), 118.91 (C-2), 116.35 (C-18,19), 115.82 (C-6), 57.91 (C-8). MS (ESI, m/z): 430.0 ([M + H]+).

Biological activity. Cell culture maintenance. A549 (lung cancer) and HepG2 (hepatic cancer) cells were cultured in the DMEM growth media with 10% heat inactivated fetal bovine serum (FBS), 1% (v/v) of penicillin and streptomycin solution. All cell lines were cultured at 37°C in a 5% CO2, fully humidified atmosphere for 3-4 days.

Cytotoxicity assay. 100 μL of culture media containing cells were seeded into 96-well plates. After 24 h of incubation, the culture medium was removed. Then the fresh medium alone or the tested compounds dissolved in DMSO at increasing concentrations from 0.625 to 160 µM for cancerous cell lines were added into 96-well plates in which cancerous cells existed. The positive control drugs included Gefitinib and Sorafenib (0.625–160 μM, final concentration less than 0.4%) and DMSO was as blank control.

After 72 h, 20 µL/well of media having MTT (5 mg/mL) was added for 4h at incubated at 37°C. At last, the 100 µL of media was discarded and 100 µL of Dimethyl sulfoxide (DMSO) was added to solubilize the purple-colored formazon. All experimental conditions were tested intriplicate and the experiment was performed three times.

Finally, the absorbance of plate was recorded at 490 nm using MicroplateReader. Then using SPSS18 software, IC50 values were calculated. IC50 values were reported as mean ± SD.

Dock and virtual screening. Preparation of proteins. Three-dimensional structure of the target proteins (PDB file format) was obtained from RCSB database. Then using PymolWin software, the original water and ligands on the protein were removed and saved as PDB file format. Next, the obtained protein was added hydrogens (Polar Only), added the atomic type (Assign AD4 type) by AutoDock Tools–1.5.6 software and saved as PDBQT file format.

Preparation of ligands. ChemDraw 14.0 software was utilized to generate the 2D structures of the synthesized compounds. Then the 2D structures were converted into 3D structures with the best dominant conformation by Chem3D software and saved as PDB file format. Next, using AutoDock Tools–1.5.6 software, the PDB file format of ligands were converted to the PDBQT file format. This process required calculating the charge (Compute Gasteiger), adding the atomic type (Assign AD4 type), choosing root and torsions on the ligand.

Grid map calculations. AutoDock grid maps were calculated for each compound using AutoDock Tools–1.5.6 software, based on the active site coordinates of each protein crystal structure. The size of all grid boxes 126×126×126 xyz points with a grid spacing of 0.375 Å. Then the GPF file format was output and the file was calculated by running AutoGrid program.

Molecular docking simulations. Using Docking program of AutoDock Tools–1.5.6 software, the Genetic Algorithm (GA) was used for pose sampling and DPF file format was output. Then the file was calculated by running AutoDock program. Finally, the configuration with the lowest binding energy was obtained from the Analyze program and saved as PDB file format. The results of molecular docking were visualized in Discovery Studio 4.5 client software and then analyzed the docking results.

CONCLUSIONS

In summary, utilizing substitution reaction, amide condensation reaction and click reaction, we have synthesized and characterized a combinatorial library of 7-azaindole with 1,2,3-triazole. All the newly synthesized compounds 3a3r were subjected to in vitro anti-cancer activity against lung cancer cell lines and hepatic cancer cell lines. The studies reveal that compound 3d, 3l, 3q and 3r have obvious inhibitory activity on lung cancer and hepatic cancer cell lines and exhibit IC50 values comparable to the positive control. Molecular docking studies indicate that compound 3d, 3l, 3q and 3r can effectively bind to multiple protein targets especially to the target 3LXK, 4AGD and 3WZE. Of all the compounds, compound 3l shows the strongest inhibitory activity and the lowest values of binding energy. In view of this, we investigate the binding of compound 3l to the protein target 3LXK 4AGD and 3WZE. And the results show that compound 3l can form two strong hydrogen bonds with the protein target, and can form hydrophobic interactions with multiple amino acid residues on the target, so as to effectively embed into the active pocket of the target. Overall, the synthesized compounds demonstrate inhibition against multiple targets, indicating great potential as multi-kinase target inhibitors in cancer chemotherapy. Our work illustrates lead molecules for anticancer chemotherapy.