Skip to main content
Journal of Enzyme Inhibition and Medicinal Chemistry logoLink to Journal of Enzyme Inhibition and Medicinal Chemistry
. 2024 Nov 19;39(1):2423875. doi: 10.1080/14756366.2024.2423875

Discovery of novel 2,4-diarylaminopyrimidine hydrazone derivatives as potent anti-thyroid cancer agents capable of inhibiting FAK

Hongting Li a,#,, Mei-Qi Jia b,#, Zhao-Long Qin c, Changliang Lu d, Weili Chu e, Ze Zhang e, Jinbo Niu f, Jian Song b, Sai-Yang Zhang b, Lijun Fu a,
PMCID: PMC11578424  PMID: 39560175

Abstract

In this work, thirty 2,4-diarylaminopyrimidine-based hydrazones were designed, synthesised, and their anti-thyroid cancer activity were explored. The majority of compounds exhibit moderate to excellent cytotoxic activity against FAK overexpressing TPC-1 cells, with IC50 values ranging from 0.113 to 1.460 μM. Among them, compound 14f displayed exceptional anti-proliferative effect against TPC-1 cells (IC50 = 0.113 μM) and potent FAK inhibitory potency (IC50 = 35 nM). In silico studies indicated that compound 14f could well bind to FAK (Focal Adhesion Kinase) and have favourable pharmacokinetic profiles. In addition, compound 14f could inhibit the phosphorylation of FAK at Tyr397, Tyr576/577 and Tyr925, and did not affect the expression level of FAK in TPC-1 cells. Compound 14f was also effective in inhibiting the proliferation and migration of thyroid cancer cells TPC-1. Thus, these novel 4-arylaminopyrimidine hydrazone derivatives exhibited potent anti-thyroid cancer activities through the inhibition of FAK.

Keywords: Thyroid cancer; 2,4-Diarylaminopyrimidine; Hydrazone; FAK; Antiproliferative activities

Introduction

The incidence of thyroid cancer, the most prevalent endocrine malignancy, is progressively increasing over the past decades1. For the majority of subtypes, the prognosis remains favourable following traditional surgery, TSH suppression therapy and 131I treatment. However, there are still a few cases of differentiated thyroid cancer (DTC) that eventually develop resistance to radioactive 131I treatment and encounter recurrence or metastasis2. In addition, anaplastic thyroid cancer (ATC) is the type with the lowest degree of differentiation3. Although its incidence is low, the ATC has a high malignancy and could metastasise to distant sites early on, making it incurable by traditional treatments. Despite significant advancements in thyroid cancer treatment over the past decades, including the approval of various kinase inhibitors, such as VEGFR and RET inhibitors, the efficacy of these drugs remains unsatisfactory for patients with poorly DTC or ATC due to primary and acquired resistance4–7. Therefore, there is still an urgent need for new therapeutic strategies and novel chemical small molecules to address the treatment of thyroid cancer.

Recently, the 2,4-diaminopyrimidine moiety is usually used as a core fragment to design novel anticancer agents8–14, especially for the discovery of kinase inhibitors, such as ALK inhibitor ceritinib (1)15, JAK/Syk inhibitor cerdulatinib (2)16 and EGFR inhibitor 317 (Figure 1A). Among them, ceritinib has been approved by FDA for the treatment of NSCLC (non-small cell lung cancer) with excellent market performance, and cerdulatinib was also approved to clinical trials for the treatment of Relapsed/Refractory B-cell Malignancies. Further researches have shown that the pyrimidine ring of 2,4-diaminopyrimidine moiety could often form critical hydrogen bonds with kinases, leading to strong inhibitory activity11,18,19. Importantly, the 2,4-aminopyrimidine moiety also used as the core scaffold for the design of anti-thyroid cancer agents, such as CDK4/6 inhibitor Abemaciclib20 and BRAFV600E inhibitor Encorafenib21. Therefore, we here designed novel 2,4-diaminopyrimidine derivatives as anti-thyroid cancer agents. In our previous work, we also reported novel 2,4-diaminopyrimidine derivatives 422 and 523 as potent anticancer agents. Although compounds 4 and 5 had some advantageous effects on anticancer activities, further structural optimisation was still necessary for the development of new effective anticancer agents. The hydrazone moiety, due to its ability as a hydrogen bond donor and acceptor, has been widely used in drug design, especially in the design of anti-tumor drugs, and this functional group could endow flexible chemical structures to a certain extent24–31. For example, Bisantrene (6)32 has been approved for the treatment of human cancers, and SP-2577 (7)33 as a LSD1 inhibitor exhibited potent inhibitory effects on acute myelogenous leukaemia. In addition, compound 834 displayed significant antiproliferative activity against MCF7 cells (IC50 = 3.3 nM). 5-Deazaflavin hydrazone derivative 935 could also effectively inhibit the proliferative activity of MCF7 cells with an IC50 value of 0.1 nM (Figure 1B).

Figure 1.

Figure 1.

(A) Representative and reported 2,4-diaminopyrimidine derivative as anticancer agents; (B) Representative and reported anticancer agents bearing a hydrazone moiety.

TAE-226, as a Focal Adhesion Kinase (FAK) inhibitor, demonstrated high cytotoxicity against multiple types of cancer cells36. Although it has never progressed to the clinical trials, the distinctive 2,4-diaminopyrimidine scaffold of TAE-226 has become the structural foundation for many subsequent anticancer agents, especially FAK inhibitors37–42. According to the co-crystallization of TAE-226 and FAK (PDB:2JKK), the 2-amino junction of the pyrimidine core is closely connected to the DFG motif region of the FAK. And, the methyl carbamoyl moiety of TAE-226 forms a hydrogen bond. The N atoms at the 1-position of the pyrimidine ring and the amino group connected to 2-position could form hydrogen bonds with Cys502. In addition, the 2-methoxyaniline morpholine moiety delves deep into the solvent region. In this work, we used the molecular hybridisation strategy to insert the hydrazone moiety into TAE-226 at the C-2 position, and then explored structural modification on the solvent region, thereby obtaining novel 2,4-diarylaminopyrimidine hydrazone derivatives. We further explored the antiproliferative activity against thyroid cancer cells TPC-1 of the obtained compounds as well as the FAK inhibitory activity of the optimal compound (Figure 2). Among them, compound 14f displayed exceptional antiproliferative effect against TPC-1 cells (IC50 = 0.113 μM) and potent FAK inhibitory potency (IC50 = 35 nM).

Figure 2.

Figure 2.

Design strategies for 2,4-diarylaminopyrimidine hydrazone derivatives.

Chemistry

The synthesis of 2,4-diarylaminopyrimidine-based hydrazones is shown in Scheme 1. Through the nucleophilic substitution reaction, commercially available 2-amino-N-methylbenzamide (11) reacted with 2,4,5-trichloropyrimidine to give intermediate 12 in the presence of DIPEA in isopropyl alcohol, which was converted to the key intermediate 13 through anther substitution reaction with hydrazine hydrate. Finally, target compounds 14a-14ad were prepared by the condensation reactions of intermediate 3 and a commercially available aldehyde or ketone.

Scheme 1.

Scheme 1.

Synthesis of target compounds 14a-14ad. Reagents and conditions: (a) 2,4,5-trichloropyrimidine, isopropyl alcohol, DIPEA, reflux, 12 h; (b) hydrazine hydrate, ethanol, 25 °C, 4 h. (c) aldehydes or ketones, ethanol, 80 °C, 5 h.

Results and discussion

In vitro antiproliferative activity and structure-activity relationships

Considering that compounds 14a-14ad are designed as anti-thyroid cancer agents, their anti-thyroid cancer cells TPC-1 activity was evaluated with the TAE-226 as a positive control.

As a general trend, the majority of compounds exhibit moderate to excellent cytotoxic activity against FAK overexpressing TPC-1 cells, with IC50 values ranging from 0.113 to 1.460 μM (Table 1). The most promising compound 14f, demonstrates significant activity in TPC-1 cells with an IC50 value of 0.113 μM. In comparison, it is approximately 8.57 times more potent than TAE-226 (IC50 = 1.082 μM). To conduct a preliminary analysis of the structure-activity relationships, we synthesised compounds 14a-14r with various aromatic substituents (R1 group) to investigate the impact of R1 substitutions on antiproliferative activity. As shown in Table 1, the results demonstrated these compounds (14a-14r) exhibit moderate to potent inhibitory activity against TPC-1 cells, with IC50 values ranging from 0.113 to 0.693 μM. When the R1 group is a phenyl moiety, the IC50 value of 14a against TPC-1 cells is 0.306 μM. Based on the IC50 values of compounds 14a-14r, analysis of structure-activity relationships reveals that variations in R1 substitutions significantly affect the antiproliferative activity against TPC-1 cells. Compared to 14a, the incorporation of electron-withdrawing groups such as F (14f), CF3 (14e), CN (14j), and NO2 (14i) into the phenyl moiety at the para position leads to an augmentation in the antiproliferative activity of these compounds against TPC-1 cells, with IC50 values range from 0.113 to 0.193 μM. However, when electron-withdrawing groups chlorine (14 g) and bromine (14n) were introduced, a significant reduction in activity was observed with IC50 values of 0.409 and 0.590 μM, respectively. Furthermore, the introduction of electron-donating groups at the para position of the phenyl moiety (R1 group), such as methyl (14b), methoxy (14c), or hydroxy (14d), could sustain the antiproliferative activity. In addition, the results of the antiproliferative activity of compound 4k-4m further demonstrated that substituent groups positioned ortho of the phenyl moiety (R1 group) could significantly mitigate the cytotoxicity towards TPC-1 cells. The same trend was also observed in compounds 14n-14r. Compared with compound 14c, the introduction of the additional methoxy (14o) or F (14q) group at the 3-position of the phenyl moiety (R1 group) leads to a decrease in activity, while introducing the methoxy (14n) or F (14p) group at the 2-position maintains activity. Similarly, based on compound 14f, maintaining activity could be achieved by introducing a F group at the 2-position of its R1 group (14r). Further substitution of the phenyl moiety (R1 group) with five- or six-membered heterocyclic groups, such as pyridine, thiophene, indole, and quinoline, resulted in the synthesis of compounds 14s-14ab. Compared to compound 14a, compounds 14v and 14 u containing thiophene and pyridine groups exhibited enhanced inhibitory potency, while replacement with a 2-pyridine (14s) or 3-pyridine (14t) group maintained activity. Introduction of larger indole moiety (14x, 14aa and 14ab), substituted indole moiety (14 y and 14z) or quinoline group (14w) led to varying degrees of decreased antiproliferative activity against TPC-1 cells. These findings also suggested that an appropriate R1 group contributes to the binding affinity between the compound and active pocket of the kinase. As a continuation of SAR research, we synthesised compounds 14ac and 14ad by introducing a methyl group into the hydrazone moiety. Regrettably, the incorporation of the methyl group significantly attenuated the antiproliferative activity against TPC-1 cells compared to compounds 14c and 14f. Therefore, compound 14f was consequently selected for further investigation and mechanistic studies.

Table 1.

Antiproliferative activity 14a∼14ad and TAE-226 against thyroid cancer cells TPC-1.

Inline graphic
Compd. R1 R2 IC50 (μM)a Compd. R1 R2 IC50 (μM)a
14a H Ph 0.246 ± 0.038 14q H 3-F-4-diOCH3-Ph 0.386 ± 0.047
14b H 4-CH3-Ph 0.296 ± 0.021 14r H 2, 4-diF-Ph 0.122 ± 0.032
14c H 4-OCH3-Ph 0.257 ± 0.004 14s H graphic file with name IENZ_A_2423875_ILG0002_B.jpg 0.272 ± 0.016
14d H 4-OH-Ph 0.255 ± 0.025 14t H graphic file with name IENZ_A_2423875_ILG0003_B.jpg 0.243 ± 0.001
14e H 4-CF3-Ph 0.193 ± 0.029 14 u H graphic file with name IENZ_A_2423875_ILG0004_B.jpg 0.189 ± 0.005
14f H 4-F-Ph 0.113 ± 0.003 14v H graphic file with name IENZ_A_2423875_ILG0005_B.jpg 0.153 ± 0.005
14 g H 4-Cl-Ph 0.409 ± 0.093 14w H graphic file with name IENZ_A_2423875_ILG0006_B.jpg 0.413 ± 0.015
14h H 4-Br-Ph 0.590 ± 0.024 14x H graphic file with name IENZ_A_2423875_ILG0007_B.jpg 0.357 ± 0.010
14i H 4-NO2-Ph 0.157 ± 0.008 14 y H graphic file with name IENZ_A_2423875_ILG0008_B.jpg 0.425 ± 0.023
14j H 4-CN-Ph 0.126 ± 0.011 14z H graphic file with name IENZ_A_2423875_ILG0009_B.jpg 0.624 ± 0.044
14k H 3-OCH3-Ph 0.517 ± 0.031 14aa H graphic file with name IENZ_A_2423875_ILG0010_B.jpg 0.331 ± 0.012
14 l H 3-F-Ph 0.428 ± 0.009 14ab H graphic file with name IENZ_A_2423875_ILG0011_B.jpg 0.364 ± 0.015
14 m H 3-CF3-Ph 0.693 ± 0.017 14ac CH3 4-OCH3-Ph 0.612 ± 0.016
14n H 2, 4-diOCH3-Ph 0.263 ± 0.076 14ad CH3 4-F-Ph 1.460 ± 0.066
14o H 3, 4-diOCH3-Ph 0.434 ± 0.012 TAE-226 1.082 ± 0.023
14p H 2-F-4-diOCH3-Ph 0.228 ± 0.065        
a

MTT assay. Cells were treated with compounds or TAE-226 for 48 h to obtain average IC50 values with SD (n = 3, duplicate).

In vitro enzymatic assays and molecular docking study

Given its origin from the FAK inhibitor TAE-226 and remarkable antiproliferative activity against thyroid cancer cells TPC-1, the inhibitory potential of compound 14f against FAK was further, evaluated. According to the reported work, the 4-arylaminopyrimidine derivatives possessing a hydrazone moiety might also exhibit certain inhibitory activities against another kinases31. Therefore, the activity of compound 14f against ALK, EGFR, Pyk2, and TYK2 kinases was also tested. The results demonstrated that, although the FAK inhibitory activity exhibited a decrease in comparison to TAE-226, compound 14f still displayed significant inhibition against FAK with an IC50 of 35 nM (Figure 3A). Additionally, it exhibited moderate inhibitory activity against four other kinases. Consequently, compound 14f could be considered as a promising FAK inhibitor for potential applications in antithyroid cancer therapy.

Figure 3.

Figure 3.

(A) Enzymatic inhibition of compound 14f and TAE-226; (B) The binding mode comparison between compound 14f with TAE-226 (PDB: 2JKK); (C) The proposed binding mode of compound 14f with FAK.

To further elucidate the binding modes between 14f with FAK (PDB ID: 2JKK), we employed molecular docking analysis using MOE 2019. As shown in Figure 3(B), compound 14f (pink) shared similar binding modes with TAE-226 (Green). It was observed the N atom at the 1-position of the pyrimidine ring and the N atom connected to the 2-position could form two hydrogen bonds with the Cys502 and Asp564. In addition, the methyl carbamoyl moiety also formed a hydrogen-bonding interaction with and Asp564 of FAK. The NH group on the methyl carbamoyl moiety could form hydrogen bonds with Glu506 and Arg550 through water mediation.

In silico physicochemical properties, ADMET profiles, and drug-likeness data

The effectiveness of newly synthesised compounds is intricately linked to their biological activity, physicochemical, pharmacokinetics, and drug likeness properties, which are crucial for identifying potential candidate drugs.

Here, we used the Swiss ADME Online software (www.SwissADME.ch) to measure in the silico ADME profile of the compound 14f, 14r and 14v, which exhibited potent antiproliferative activity against thyroid cancer cells TPC-1. The results indicated that compounds 14f, 14r and 14v exhibited a predicted clogP in a range of 3.44–4.61, moderate water solubility, and high GIT absorption. As shown in Figure 4, the Boiled-Egg chart suggests that these three compounds have GIT absorption, due to their location in the human intestinal absorption (HIA) area. However, due to the high polarity of the hydrazine moiety, compounds 14f, 14r and 14v did not exhibit potential BBB permeability, indicating that these compounds face certain difficulties in reaching the CNS. The oral bioavailability of the compounds 14f, 14r and 14v is predicted in the radar chart (Figure 4). The results indicated the measured physicochemical properties of these three compounds located in the ideal pink area, except for the INSATU parameter.

Figure 4.

Figure 4.

(A) The boiled-egg chart of the compounds 14f, 14r and 14v; (B) The bioavailability radar chart of the compounds 14f, 14r and 14v.

Furthermore, the metabolism of compounds 14f, 14r and 14v is postulated to inhibit four or five main cytochrome P-450 (CYP) isoforms in the liver (Table 2), suggesting that these three compounds are easy to have potential drug-drug interactions. Moreover, compounds 14f, 14r and 14v are not the substrates of Pg-p (Table 2), which might also exhibit potent activities against Pg-p overexpressing cancer cells. However, these results are based only on chemoinformatic simulations, and additional experiments are needed to verify this prediction.

Table 2.

The in silico predicted ADME profiles for compounds 14f, 14r and 14v.

Compd. GI absorption BBB permeability Pg-p substrate CYP1A2 inhibitor CYP2C19 inhibitor CYP2C9 inhibitor CYP2D6 inhibitor CYP3A4 inhibitor
14f High No No Yes No Yes No Yes
14r High No No Yes Yes Yes No Yes
14v High No No Yes Yes Yes No Yes

The physicochemical properties of compounds 14f, 14r and 14v in Table 3 indicated that they have acceptable properties including molecular weight, Log Po/w, H-bond acceptors, rotatable bonds, TPSA and Log S, except for the rotatable bonds, which might help to demonstrate good physicochemical properties.

Table 3.

In silico physicochemical properties for compounds 14f, 14r and 14v.

Compd. MW < 500 Log Po/w < 5 (WLOGP) HBA < 10 HBD < 5 NRB < 5 TPSA Å2 < 160 Log S (ESOL)
14f 398.82 4.05 5 3 7 91.30 −5.21
14r 386.86 4.61 6 3 7 91.30 −5.38
14v 386.86 3.55 4 3 7 119.54 −5.10

MW: molecular weight; Log Po/w (WLOGP): partition coefficient octanol/water; HBA: number of H-bond acceptors; HBD: number of H-bond donors; NRB: number of rotatable bonds; TPSA: topological polar surface area; Log S (ESOL): aqueous solubility.

In addition, the results measured by the Swiss ADME Web-tool indicate that 14f, 14r and 14v have acceptable drug-likeness and conform to the rules (Table 4). Importantly, there are lack of PAINS alert in the view of medicinal chemistry, dedicating that the in vitro bioassays of these three compounds might be robustly obtained.

Table 4.

The drug-likeness of compounds 14f, 14r and 14v.

Compd. Lipinski #violations Ghose Veber Egan Muegge Bioavail. Score PAINS #alert
14f 0 Yes Yes Yes Yes 0.55 0
14r 0 Yes Yes Yes Yes 0.55 0
14v 0 Yes Yes Yes Yes 0.55 0

Effects of compound 14f on the FAK autophosphorylation in thyroid cancer cells TPC-1

The phosphorylation of FAK (as depicted in Figure 5A) positively regulates the proliferation and migration of cancer cells. Therefore, the effects of compound 14f on the expression levels of FAK and its phosphorylation sites in thyroid cancer cells TPC-1 were examined. As shown in Figure 5(B), the results indicated that treatment with compound 14f significantly increased the phosphorylation levels of FAK at Tyr397, Tyr576/577, and Tyr925 in a dose-dependent manner, with particularly pronounced inhibition observed at Tyr397 and Tyr576/577 sites. Importantly, no significant changes were observed in the expression level of FAK upon treatment with compound 14f (Figure 5B). These results suggested that Figure 5(B) could act on FAK to inhibit its activation through phosphorylation without affecting its expression or degradation processes.

Figure 5.

Figure 5.

The structural diagram of the FAK protein (A) and the inhibitory effect of compound 14f on FAK (B). In Figure 5(B), thyroid cancer cell TPC-1 was treated with DMSO or indicated concentrations of compound 14f (50, 100 and 200 nmol/L) for 48 h.

Effects of compound 14f on the proliferation and migration of thyroid cancer cells TPC-1

The phosphorylation and subsequent activation of FAK could facilitate the proliferation and migration of cancer cells. Suppression of FAK activation through specific compounds has been shown to effectively inhibit the proliferative and migratory abilities of cancer cells. Therefore, the effects of compound 14f on the proliferative and migratory capacities of thyroid cancer cells TPC-1 were evaluated.

Cells exhibiting positive EdU staining are indicative of robust proliferative activity. The experimental findings revealed there was a significant reduction in the number of thyroid cancer cells positive for EdU following treatment with compound 14f (Figure 6A). The colony formation assay serves as an indirect measure to assess cellular proliferation. As shown in Figure 6(B), compound 14f exhibited discernible inhibitory effects on the colony formation of TPC-1 cells starting from a concentration of 100 nmol/L over a span of 5-day treatment. In addition, we further explored the effects of compound 14f on the cell cycle distribution of TPC-1 cells, which could reflect the state of cellular proliferation. The results depicted in Figure 6(C&D) indicated that treatment with compound 14f induced a pronounced G1 phase arrest in TPC-1 cells. These above results demonstrate that compound 14f effectively inhibit the proliferation of thyroid cancer cells TPC-1.

Figure 6.

Figure 6.

Effects of compound 14f on the proliferation of thyroid cancer cells TPC-1. (A) In the EdU staining assay, TPC-1 cells treated with compound 14f for 48 h were stained with EdU and Hoechst; (B) The colony formation of TPC-1 cells after treatment with various concentrations of compound 14f for 5 days; (C&D) The changes in the cell cycle distribution of TPC-1 cells after a 48-h treatment.

In the wound healing assay, the healing capacity of the cellular wound was impeded in the presence of compound 14f, indicating that compound 14f has a significant inhibitory effect on cell migration of thyroid cancer cells (Figure 7A). The transwell migration assay serves as an additional method to evaluate cellular migratory activity. Notably, a significant reduction in the number of cells that traversed through the membrane was observed upon exposure to compound 14f (Figure 7A). These results demonstrated that compound 14f effectively inhibited the migration of thyroid cancer cells TPC-1.

Figure 7.

Figure 7.

Effects of compound 14f on the migration of thyroid cancer cells TPC-1. (A&B) In the wound healing assay, the healing of wounds in TPC-1 cells treated with compound 14f for various durations is shown; (C) In the transwell migration assay, TPC-1 cells that migrated through the membrane after 48 h of treatment with compound 14f are presented. **p < 0.01, ****p < 0.0001.

Conclusions

In this study, we reported the discovery of novel 2,4-diarylaminopyrimidine-based hydrazones as anti-thyroid cancer agents. Based on the FAK inhibitor TAE-226, we designed and synthesised thirty novel compounds by introducing a hydrazone group, and evaluated their proliferation inhibitory activities against FAK-overexpressing TPC-1 cells. The majority of compounds exhibit moderate to excellent cytotoxic activity against FAK overexpressing TPC-1 cells, with IC50 values ranging from 0.113 to 1.460 μM. Among them, compound 14f displayed exceptional anti-proliferative effect against TPC-1 cells (IC50 = 0.113 μM) and potent FAK inhibitory potency (IC50 = 35 nM). The results of molecular docking indicated compound 14f could well bind to FAK. Furthermore, the calculated physicochemical properties showed that compound 14f has favourable pharmacokinetic profiles. In addition, compound 14f could inhibit the phosphorylation of FAK at Tyr397, Tyr576/577, and Tyr925 in a dose-dependent manner, and did not affect the expression level of FAK in TPC-1 cells. Treatment with compound 14f was also effective in inhibiting the proliferation and migration of thyroid cancer cells TPC-1. Taken together, these results provided potential 2,4-diarylaminopyrimidine-based hydrazones for the treatment of thyroid cancers through the inhibition of FAK.

Experimental section

General chemical methods

All commercially available starting materials and solvents are used directly without further purification. The TLC was used to monitor the reactions. Column chromatography was performed using silica gel (200–300 mesh). Melting points were recorded by an X-5 micro melting apparatus. NMR spectra was acquired using a Bruker av400 spectrometer. HRMS was obtained on the Agilent 6545 LC/Q-TOF mass spectrometer.

Procedure for the synthesis of 12

To a stirred solution of commercially available 11 (2.0 mmol) and 2,4,5-trichloropyrimidine (2.0 mmol) in isopropanol (20 ml) was added DIPEA (0.2 mmol). Then, the mixture was stirred at 85 °C for 12 h. Subsequently, the mixture was cooled to room temperature, and then the intermediate 12 was obtained after the filtration of the precipitation and the dryness of the filter cake without further purification.

Procedure for the synthesis of 13

To a stirred solution of intermediate 12 (2.0 mmol) in ethanol (20 ml) was added hydrazine monohydrate (2.0 mmol). Then, the mixture was stirred at 25 °C for 4 h. Subsequently, the intermediate 13 was obtained after the filtration of the precipitation and the dryness of the filter cake without further purification.

Procedure for the synthesis of 14a-14ad

To a stirred solution of intermediate 13 (1.0 mmol) in ethanol (10 ml) was added substituted aldehyde or ketone (1.0 mmol). Then, the mixture was stirred at 80 °C for 5 h. Subsequently, the filtrate was evaporated in vacuo to get crude products, which was then purified by the chromatography on silica gel to obtain compounds 14a-14ad.

(E)-2-((2–(2-benzylidenehydrazineyl)-5-chloropyrimidin-4-yl)amino)-N-methylbenzamide (14a)

White solid; yield: 58%. M.p.199.1–202.1 °C.1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 11.26 (s, 1H), 9.37 (s, 1H), 8.80 (d, J = 4.5 Hz, 1H), 8.23 (s, 1H), 8.14 (s, 1H), 7.83 − 7.78 (m, 1H), 7.74 (d, J = 7.4 Hz, 2H), 7.60 (t, J = 7.4 Hz, 1H), 7.48 (t, J = 7.5 Hz, 2H), 7.39 (t, J = 7.3 Hz, 1H), 7.18 (t, J = 7.2 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H).13C NMR (100 MHz, DMSO-d6) δ 169.57, 158.13, 155.77, 155.31, 141.60, 140.49, 135.82, 132.09, 129.45, 129.29, 128.49, 126.67, 122.25, 121.38, 120.40, 105.92, 26.86. HRMS: calcd. C19H18ClN6O1 [M + H] +, 381.1225, found: 381.1222.

(E)-2-((5-chloro-2–(2-(4-methylbenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14b)

White solid; yield: 62%. M.p.207.1–208.1 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 11.19 (s, 1H), 9.37 (s, 1H), 8.80 (d, J = 4.4 Hz, 1H), 8.21 (s, 1H), 8.10 (s, 1H), 7.81 (d, J = 7.0 Hz, 1H), 7.60 (dd, J = 19.5, 7.7 Hz, 3H), 7.28 (d, J = 7.9 Hz, 2H), 7.17 (t, J = 7.3 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H), 2.35 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.61, 158.18, 155.77, 141.67, 140.56, 139.14, 133.15, 132.15, 130.31, 129.93, 129.58, 128.54, 126.68, 121.79, 121.41, 120.38, 105.75, 26.54, 21.49. HRMS: calcd. C20H20ClN6O1 [M + H] +, 395.1382, found: 395.1379.

(E)-2-((5-chloro-2–(2-(4-methoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14c)

White solid; yield: 67%. M.p.187.7–188.9 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.91 (s, 1H), 11.10 (s, 1H), 9.36 (s, 1H), 8.78 (d, J = 4.4 Hz, 1H), 8.20 (s, 1H), 8.09 (s, 1H), 7.80 (d, J = 7.7 Hz, 1H), 7.67 (d, J = 8.6 Hz, 2H), 7.60 (t, J = 7.8 Hz, 1H), 7.17 (t, J = 7.5 Hz, 1H), 7.05 (d, J = 8.6 Hz, 2H), 3.82 (s, 3H), 2.83 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.58, 160.55, 158.18, 155.73, 155.28, 141.68, 140.51, 132.17, 128.46, 128.40, 128.14, 122.20, 121.35, 120.35, 114.82, 105.51, 55.76, 26.85. HRMS: calcd. C20H20ClN6O2 [M + H] +, 411.1331, found: 411.1325.

(E)-2-((5-chloro-2–(2-(4-hydroxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14d)

White solid; yield: 49%. M.p.187.8–189.2 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 11.03 (s, 1H), 9.79 (s, 1H), 9.35 (s, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.19 (s, 1H), 8.05 (s, 1H), 7.80 (d, J = 7.4 Hz, 1H), 7.57 (t, J = 8.1 Hz, 3H), 7.17 (t, J = 7.4 Hz, 1H), 6.86 (d, J = 8.4 Hz, 2H), 2.83 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.58, 159.00, 158.21, 155.71, 155.29, 142.16, 140.54, 132.09, 128.46, 128.31, 126.85, 122.16, 121.37, 120.35, 116.17, 105.34, 26.85. HRMS: calcd. C19H18ClN6O2 [M + H] +, 397.1174, found: 397.1174.

(E)-2-((5-chloro-2–(2-(4-(trifluoromethyl)benzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14e)

White solid; yield: 56%. M.p.206.6–208.8 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.99 (s, 1H), 11.49 (s, 1H), 9.33 (s, 1H), 8.81 (d, J = 4.4 Hz, 1H), 8.26 (s, 1H), 8.19 (s, 1H), 7.93 (d, J = 8.1 Hz, 2H), 7.83 (dd, J = 11.9, 8.3 Hz, 3H), 7.67 (t, J = 7.7 Hz, 1H), 7.18 (t, J = 7.3 Hz, 1H), 2.84 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ169.59, 157.99, 155.84, 140.42, 139.85, 132.43, 132.37, 129.27, 128.96, 128.58, 127.08, 126.29, 126.16, 123.45, 121.35, 120.41, 106.52, 29.30. HRMS: calcd. C20H17ClF3N6O [M + H] +, 449.1099, found: 449.1094.

(E)-2-((5-chloro-2–(2-(4-fluorobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14f)

White solid; yield: 55%. M.p.176.1–177.8 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.94 (s, 1H), 11.25 (s, 1H), 9.34 (s, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.22 (s, 1H), 8.13 (s, 1H), 7.83 − 7.74 (m, 3H), 7.61 (t, J = 7.7 Hz, 1H), 7.32 (t, J = 8.8 Hz, 2H), 7.17 (t, J = 7.5 Hz, 1H), 2.84 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.56, 164.19, 161.74, 158.11, 155.76, 155.28, 140.49, 140.44, 132.42, 132.39, 132.19, 128.64, 128.56, 128.47, 122.27, 121.34, 120.40, 116.43, 116.21, 105.94, 26.84. HRMS: calcd. C19H17ClFN6O [M + H] +, 399.1131, found: 399.1129.

(E)-2-((5-chloro-2–(2-(4-chlorobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14 g)

White solid; yield: 58%. M.p.194.1 − 195.8 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 11.33 (s, 1H), 9.32 (s, 1H), 8.79 (d, J = 3.8 Hz, 1H), 8.23 (s, 1H), 8.12 (s, 1H), 7.77 (dd, J = 24.4, 7.9 Hz, 3H), 7.62 (t, J = 7.7 Hz, 1H), 7.54 (d, J = 8.1 Hz, 2H), 7.17 (t, J = 7.4 Hz, 1H), 2.84 (d, J = 4.0 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.59, 158.53, 158.08, 155.93, 155.80, 140.47, 140.24, 134.82, 133.79, 129.63, 129.34, 128.30, 121.36, 120.39, 112.65, 106.14, 26.75. HRMS: calcd. C19H17Cl2N6O [M + H] +, 415.10835, found: 415.10833.

(E)-2-((2–(2-(4-bromobenzylidene)hydrazineyl)-5-chloropyrimidin-4-yl)amino)-N-methylbenzamide (14h)

White solid; yield: 58%. M.p.192.1–193.2 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 11.33 (s, 1H), 9.32 (s, 1H), 8.79 (d, J = 4.2 Hz, 1H), 8.23 (s, 1H), 8.11 (s, 1H), 7.81 (d, J = 7.7 Hz, 1H), 7.70 − 7.60 (m, 5H), 7.17 (t, J = 7.5 Hz, 1H), 2.84 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.57, 160.08, 158.01, 155.77, 155.29, 140.39, 140.30, 135.11, 132.27, 128.46, 122.60, 122.45, 122.34, 121.34, 120.40, 106.15, 26.85. HRMS: calcd. C19H17ClBrN6O [M + H] +, 459.0330, found: 459.0325.

(E)-2-((5-chloro-2–(2-(4-nitrobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14i)

White solid; yield: 58%. M.p.201.7–202.8 °C. 1H NMR (400 MHz, DMSO-d6) δ 12.00 (s, 1H), 11.63 (s, 1H), 9.29 (s, 1H), 8.80 (d, J = 4.5 Hz, 1H), 8.33 (d, J = 8.7 Hz, 2H), 8.27 (s, 1H), 8.21 (s, 1H), 7.95 (d, J = 8.8 Hz, 2H), 7.81 (d, J = 7.8 Hz, 1H), 7.68 (t, J = 7.8 Hz, 1H), 7.19 (t, J = 7.4 Hz, 1H), 2.84 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.54, 157.81, 155.83, 155.32, 147.48, 142.32, 140.31, 139.00, 132.37, 128.50, 127.26, 126.45, 124.69, 122.45, 121.33, 106.86, 26.85. HRMS: calcd. C19H17ClN7O3 [M + H] +, 426.1076, found: 426.1072.

(E)-2-((5-chloro-2–(2-(4-cyanobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14j)

White solid; yield: 57%. M.p.200.7–202.1 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.99 (s, 1H), 11.54 (s, 1H), 9.31 (s, 1H), 8.80 (d, J = 4.3 Hz, 1H), 8.25 (s, 1H), 8.16 (s, 1H), 7.91 (q, J = 8.3 Hz, 4H), 7.81 (d, J = 7.7 Hz, 1H), 7.64 (t, J = 7.8 Hz, 1H), 7.18 (t, J = 7.5 Hz, 1H), 2.84 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.55, 157.87, 155.81, 155.28, 140.39, 140.36, 139.38, 133.21, 132.25, 128.48, 127.06, 122.38, 121.35, 120.44, 119.37, 111.08, 106.68, 26.85. HRMS: calcd. C20H17ClN7O [M + H] +, 406.1178, found: 406.1177.

(E)-2-((5-chloro-2–(2-(3-methoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14k)

White solid; yield: 53%. M.p.199.3–200.4 °C. 1H NMR (400 MHz, DMSO-d6) 1H NMR (400 MHz, DMSO) δ 11.93 (s, 1H), 11.27 (s, 1H), 9.32 (s, 1H), 8.80 (d, J = 4.1 Hz, 1H), 8.22 (s, 1H), 8.11 (s, 1H), 7.80 (d, J = 7.6 Hz, 1H), 7.58 (t, J = 7.6 Hz, 1H), 7.38 (t, J = 7.9 Hz, 1H), 7.29 (d, J = 6.3 Hz, 2H), 7.17 (t, J = 7.3 Hz, 1H), 6.96 (d, J = 7.3 Hz, 1H), 3.82 (s, 3H), 2.84 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.56, 160.08, 158.10, 155.78, 155.28, 141.53, 140.44, 137.23, 132.01, 130.35, 128.48, 122.23, 121.33, 120.48, 119.53, 115.25, 111.47, 105.97, 55.67, 26.84. HRMS: calcd. C20H20ClN6O2 [M + H] +, 411.1331, found: 411.1330.

(E)-2-((5-chloro-2–(2-(3-fluorobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14 l)

White solid; yield: 48%. M.p.203.3–205.1 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.98 (s, 1H), 11.41 (s, 1H), 9.37 (s, 1H), 8.81 (s, 1H), 8.19 (d, J = 44.4 Hz, 2H), 7.81 (d, J = 7.2 Hz, 1H), 7.55 (d, J = 19.0 Hz, 4H), 7.20 (d, J = 7.2 Hz, 2H), 2.85 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.53, 164.23, 161.81, 158.02, 155.78, 155.31, 140.46, 139.93, 138.56, 138.48, 131.87, 131.38, 131.30, 128.51, 123.23, 122.29, 121.33, 120.46, 116.20, 115.99, 112.29, 112.08, 106.23, 26.85. HRMS: calcd. C19H17ClFN6O [M + H] +, 399.1131, found: 399.1132.

(E)-2-((5-chloro-2–(2-(3-(trifluoromethyl)benzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14 m)

White solid; yield: 60%. M.p.207.1–209.8 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.97 (s, 1H), 11.46 (s, 1H), 9.30 (s, 1H), 8.81 (d, J = 4.4 Hz, 1H), 8.24 (d, J = 14.1 Hz, 2H), 8.10 (s, 1H), 7.96 (d, J = 6.8 Hz, 1H), 7.81 (d, J = 7.1 Hz, 1H), 7.77 − 7.66 (m, 2H), 7.55 (t, J = 7.5 Hz, 1H), 7.18 (t, J = 7.4 Hz, 1H), 2.84 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.52, 157.98, 155.82, 155.30, 140.38, 139.83, 136.96, 131.78, 130.97, 130.43, 130.25, 129.94, 128.52, 126.01, 125.52, 123.31, 122.23, 122.13, 121.28, 120.51, 106.35, 26.84. HRMS: calcd. C20H17ClF3N6O [M + H] +, 449.1099, found: 449.1096.

(E)-2-((5-chloro-2–(2-(2,4-dimethoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14n)

White solid; yield: 56%. M.p. 213.1–214.7 °C. 1H NMR (400 MHz, DMSO-d6) δ 11.90 (s, 1H), 11.08 (s, 1H), 9.36 (s, 1H), 8.78 (d, J = 4.4 Hz, 1H), 8.38 (s, 1H), 8.17 (s, 1H), 7.89 (d, J = 8.6 Hz, 1H), 7.79 (d, J = 6.9 Hz, 1H), 7.57 (t, J = 7.5 Hz, 1H), 7.16 (t, J = 7.5 Hz, 1H), 6.70 (d, J = 8.5 Hz, 1H), 6.63 (d, J = 2.2 Hz, 1H), 3.84 (d, J = 7.5 Hz, 6H), 2.83 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.58, 162.06, 158.85, 158.14, 155.66, 155.26, 138.51, 137.41, 132.13, 128.45, 122.15, 121.35, 121.26, 120.28, 116.66, 108.79, 106.83, 98.82, 56.19, 55.92, 26.85. HRMS: calcd. C21H22ClN6O3 [M + H] +, 441.1436, found: 441.1435.

(E)-2-((5-chloro-2–(2-(3,4-dimethoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14o)

White solid; yield: 56%. M.p. 214.1–215.2 °C. 1H NMR (400 MHz, DMSO-d6), 11.03 (s, 1H), 9.34 (s, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.35 (s, 1H), 8.20 (s, 1H), 7.79 (d, J = 8.5 Hz, 2H), 7.56 (t, J = 7.7 Hz, 1H), 7.16 (t, J = 7.5 Hz, 1H), 6.87 (dd, J = 31.1, 5.2 Hz, 2H), 3.80 (s, 3H), 2.84 (d, J = 4.4 Hz, 3H), 2.48 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.54, 158.13, 155.75, 155.29, 150.43, 149.50, 142.19, 140.39, 131.99, 128.50, 122.17, 121.27, 120.94, 120.84, 120.54, 112.19, 108.85, 105.55, 56.06, 55.97, 26.84. HRMS: calcd. C21H22ClN6O3 [M + H] +, 441.1436, found: 441.1430.

(E)-2-((5-chloro-2–(2-(2-fluoro-4-methoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14p)

White solid; yield: 57%. M.p. 218.1–219.4 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.94 (s, 1H), 11.25 (s, 1H), 9.33 (s, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.24 (d, J = 22.2 Hz, 2H), 7.91 (t, J = 8.7 Hz, 1H), 7.80 (d, J = 7.1 Hz, 1H), 7.59 (t, J = 7.6 Hz, 1H), 7.16 (t, J = 7.5 Hz, 1H), 7.02 − 6.86 (m, 2H), 3.84 (s, 3H), 2.83 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ169.56, 158.18, 158.04, 155.76, 155.36, 155.30, 154.25, 140.60, 140.46, 134.58, 132.18, 128.47, 126.91, 126.84, 125.46, 122.25, 121.32, 120.36, 112.19, 105.84, 56.35, 26.85. HRMS: calcd. C20H19ClFN6O2 [M + H] +, 429.1237, found: 429.1234.

(E)-2-((5-chloro-2–(2-(3-fluoro-4-methoxybenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14q)

White solid; yield: 57%. M.p. 234.1–235.7 °C. 1H NMR (400 MHz, DMSO-d6), 1H NMR (400 MHz, DMSO) δ 11.95 (s, 1H), 11.24 (s, 1H), 9.37 (s, 1H), 8.80 (d, J = 4.4 Hz, 1H), 8.21 (s, 1H), 8.06 (s, 1H), 7.81 (d, J = 7.4 Hz, 1H), 7.59 (dd, J = 16.7, 9.5 Hz, 2H), 7.45 (d, J = 8.4 Hz, 1H), 7.22 (dt, J = 14.9, 8.0 Hz, 2H), 3.90 (s, 3H), 2.84 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.55, 158.09, 155.74, 155.28, 153.45, 151.03, 148.39, 148.28, 140.52, 140.22, 131.89, 129.20, 129.14, 128.51, 123.99, 122.23, 121.32, 120.39, 114.40, 112.85, 112.66, 105.79, 56.61, 26.86. HRMS: calcd. C20H19ClFN6O2 [M + H] +, 429.1237, found: 429.1232.

(E)-2-((5-chloro-2–(2-(2,4-difluorobenzylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14r)

White solid; yield: 57%. M.p. 243.2–245.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.96 (s, 1H), 11.39 (s, 1H), 9.30 (s, 1H), 8.79 (d, J = 4.5 Hz, 1H), 8.28 (s, 1H), 8.23 (s, 1H), 8.02 (dd, J = 15.4, 8.4 Hz, 1H), 7.80 (d, J = 7.2 Hz, 1H), 7.60 (t, J = 7.6 Hz, 1H), 7.36 − 7.25 (m, 2H), 7.16 (t, J = 7.4 Hz, 1H), 2.83 (d, J = 4.4 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.55, 157.94, 155.78, 155.31, 140.38, 133.49, 133.46, 133.44, 132.23, 128.48, 127.43, 127.39, 122.33, 121.32, 120.39, 113.01, 112.97, 106.29, 104.92, 104.65, 26.86. HRMS: calcd. C19H16ClF2N6O [M + H] +, 417.1037, found: 417.1033.

(E)-2-((5-chloro-2–(2-(pyridin-2-ylmethylene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14s)

White solid; yield: 56%. M.p. 217.1–218.2 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.97 (s, 1H), 11.50 (s, 1H), 9.30 (s, 1H), 8.80 (d, J = 4.4 Hz, 1H), 8.58 (d, J = 4.6 Hz, 1H), 8.26 (s, 1H), 8.18 (s, 1H), 8.02 (d, J = 7.9 Hz, 1H), 7.93 (t, J = 7.3 Hz, 1H), 7.81 (d, J = 7.1 Hz, 1H), 7.62 (t, J = 7.5 Hz, 1H), 7.36 (dd, J = 6.3, 5.2 Hz, 1H), 7.18 (t, J = 7.4 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.54, 157.89, 155.81, 155.33, 154.71, 149.82, 142.10, 140.35, 137.15, 132.18, 128.50, 123.86, 122.35, 121.34, 120.49, 119.26, 106.57, 26.85. HRMS: calcd. C18H17ClN7O [M + H] +, 382.1178, found: 382.1173.

(E)-2-((5-chloro-2–(2-(pyridin-3-ylmethylene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14t)

White solid; yield: 52%. M.p. 217.1–218.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 12.01 (s, 1H), 11.59 (s, 1H), 9.31 (s, 1H), 8.81 (d, J = 4.2 Hz, 1H), 8.65 (d, J = 5.3 Hz, 2H), 8.27 (s, 1H), 8.10 (s, 1H), 7.82 (d, J = 7.7 Hz, 1H), 7.64 (dd, J = 11.5, 6.6 Hz, 3H), 7.19 (t, J = 7.5 Hz, 1H), 2.84 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.53, 157.99, 155.78, 155.30, 150.04, 148.28, 140.41, 138.57, 133.08, 132.03, 131.67, 128.49, 124.44, 122.29, 121.36, 120.45, 106.31. HRMS: calcd. C18H17ClN7O [M + H] +, 382.1178, found: 382.1174.

(E)-2-((5-chloro-2–(2-(pyridin-4-ylmethylene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14 u)

White solid; yield: 57%. M.p. 215.1–216.8 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.97 (s, 1H), 11.45 (s, 1H), 9.34 (s, 1H), 8.85 (d, J = 34.2 Hz, 2H), 8.57 (s, 1H), 8.16 (dd, J = 37.2, 19.4 Hz, 3H), 7.81 (d, J = 7.5 Hz, 1H), 7.57 (dd, J = 23.7, 16.2 Hz, 2H), 7.18 (t, J = 7.1 Hz, 1H), 2.84 (d, J = 3.1 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.54, 157.83, 155.83, 155.30, 150.63, 142.97, 140.35, 138.79, 132.17, 128.50, 122.40, 121.33, 120.66, 120.46, 106.81, 26.86. HRMS: calcd. C18H17ClN7O [M + H] +, 382.1178, found: 382.1176.

(E)-2-((5-chloro-2–(2-(thiophen-2-ylmethylene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14v)

White solid; yield: 57%. M.p. 214.6–216.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.98 (s, 1H), 11.25 (s, 1H), 9.39 (s, 1H), 8.80 (d, J = 4.5 Hz, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 7.80 (dd, J = 7.9, 1.3 Hz, 1H), 7.62 (dd, J = 17.4, 6.3 Hz, 2H), 7.33 (d, J = 2.8 Hz, 1H), 7.19 − 7.08 (m, 2H), 2.83 (d, J = 4.5 Hz, 3H).13C NMR (100 MHz, DMSO-d6) δ 169.61, 157.97, 155.75, 155.26, 141.02, 140.54, 137.03, 132.34, 128.99, 128.43, 128.18, 127.85, 122.13, 121.40, 120.24, 105.78, 26.85. HRMS: calcd. C17H16ClN6OS [M + H] +, 387.0789, found: 387.0784.

(E)-2-((5-chloro-2–(2-(quinolin-7-ylmethylene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14w)

White solid; yield: 46%. M.p. 241.7–242.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.96 (s, 1H), 11.53 (s, 1H), 9.43 (s, 1H), 8.99 (dd, J = 4.1, 1.7 Hz, 1H), 8.81 (d, J = 4.5 Hz, 1H), 8.48 − 8.43 (m, 2H), 8.25 (s, 1H), 8.04 (d, J = 7.1 Hz, 1H), 7.83 − 7.77 (m, 2H), 7.66 − 7.60 (m, 2H), 7.19 (t, J = 7.2 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.58, 158.13, 155.76, 155.30, 151.12, 150.59, 145.51, 140.47, 138.56, 137.05, 132.75, 132.16, 129.26, 128.63, 128.46, 127.08, 124.86, 122.27, 121.44, 120.40, 106.02, 26.85. HRMS: calcd. C22H19ClN7O [M + H] +, 432.1334, found: 432.1338.

(E)-2-((2–(2-((1H-indol-3-yl)methylene)hydrazineyl)-5-chloropyrimidin-4-yl)amino)-N-methylbenzamide (14x)

White solid; yield: 56%. M.p. 203.3–204.8 °C. 1H NMR (400 MHz, DMSO-d6),δ 11.84 (s, 1H), 11.46 (s, 1H), 10.86 (s, 1H), 9.43 (s, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.44 (d, J = 7.8 Hz, 1H), 8.36 (s, 1H), 8.19 (s, 1H), 7.81 (d, J = 7.1 Hz, 1H), 7.72 (d, J = 2.6 Hz, 1H), 7.60 (t, J = 7.4 Hz, 1H), 7.45 (d, J = 8.1 Hz, 1H), 7.23 (dd, J = 17.4, 7.6 Hz, 2H), 7.14 (t, J = 7.3 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.61, 158.37, 155.90, 155.32, 140.60, 140.25, 137.60, 132.13, 131.71, 129.64, 128.53, 124.78, 123.00, 122.46, 122.00, 121.28, 120.65, 120.46, 112.86, 112.25, 26.86. HRMS: calcd. C21H19ClN7O [M + H] +, 420.1334, found: 4202.1332.

((E)-2-((5-chloro-2–(2-((1-methyl-1H-indol-3-yl)methylene) hydrazineyl) pyrimidin-4-yl)amino)-N-methylbenzamide (14 y)

White solid; yield: 58%. M.p. 217.2–219.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.84 (s, 1H), 10.86 (s, 1H), 9.43 (s, 1H), 8.80 (d, J = 4.5 Hz, 1H), 8.45 (d, J = 7.7 Hz, 1H), 8.33 (s, 1H), 8.19 (s, 1H), 7.81 (d, J = 7.0 Hz, 1H), 7.71 (s, 1H), 7.60 (t, J = 7.5 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.30 (t, J = 7.4 Hz, 1H), 7.24 − 7.16 (m, 2H), 3.82 (s, 3H), 2.84 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.60, 158.32, 155.89, 155.32, 140.57, 139.73, 138.09, 133.39, 132.10, 128.53, 125.14, 123.08, 122.59, 122.02, 121.27, 120.87, 120.47, 111.91, 110.58, 104.57, 33.19, 26.85. HRMS: calcd. C22H21ClN7O [M + H] +, 434.1491, found: 434.1487.

(E)-2-((5-chloro-2–(2-((5-methoxy-1H-indol-3-yl)methylene) hydrazineyl) pyrimidin-4-yl)amino)-N-methylbenzamide (14z)

White solid; yield: 57%. M.p. 217.1–218.2 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.86 (s, 1H), 11.33 (s, 1H), 10.79 (s, 1H), 9.37 (s, 1H), 8.77 (d, J = 4.5 Hz, 1H), 8.35 (s, 1H), 8.19 (s, 1H), 7.90 (s, 1H), 7.78 (d, J = 6.9 Hz, 1H), 7.68 (d, J = 2.6 Hz, 1H), 7.55 (t, J = 7.4 Hz, 1H), 7.36 (d, J = 8.8 Hz, 1H), 7.12 (t, J = 7.5 Hz, 1H), 6.91 (dd, J = 8.7, 2.2 Hz, 1H), 3.68 (s, 3H), 2.84 (d, J = 4.5 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.63, 158.36, 155.80, 155.31, 154.77, 140.58, 132.75, 132.59, 130.16, 130.05, 128.45, 125.48, 121.83, 120.99, 120.09, 112.67, 112.46, 111.78, 106.00, 104.64, 56.18, 26.84. HRMS: calcd. C22H21ClN7O2 [M + H] +, 450.1440, found: 434.1487.

(E)-2-((2–(2-((1H-indol-5-yl)methylene)hydrazineyl)-5-chloropyrimidin-4-yl)amino)-N-methylbenzamide (14aa)

White solid; yield: 65%. M.p. 218.1–219.9 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.91 (s, 1H), 11.28 (s, 1H), 11.05 (s, 1H), 9.44 (s, 1H), 8.79 (d, J = 4.2 Hz, 1H), 8.21 (d, J = 5.5 Hz, 2H), 7.81 (s, 2H), 7.63 (t, J = 8.3 Hz, 2H), 7.49 (d, J = 8.5 Hz, 1H), 7.40 (s, 1H), 7.19 (t, J = 7.4 Hz, 1H), 6.51 (s, 1H), 2.84 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.60, 158.30, 155.72, 155.32, 143.83, 140.61, 136.97, 132.09, 128.47, 128.19, 127.04, 126.70, 122.16, 121.46, 120.37, 120.24, 119.39, 112.43, 105.20, 102.24, 26.85. HRMS: calcd. C21H19ClN7O [M + H] +, 420.1334, found: 4202.1334.

(E)-2-((2–(2-((1H-indol-6-yl)methylene)hydrazineyl)-5-chloropyrimidin-4-yl)amino)-N-methylbenzamide (14ab)

White solid; yield: 54%. M.p. 216.1–217.7 °C. 1H NMR (400 MHz, DMSO-d6),δ 11.94 (s, 1H), 11.29 (s, 1H), 11.09 (s, 1H), 9.44 (s, 1H), 8.80 (d, J = 4.3 Hz, 1H), 8.22 (d, J = 5.8 Hz, 2H), 7.82 (d, J = 7.7 Hz, 1H), 7.72 − 7.59 (m, 3H), 7.51 (d, J = 8.0 Hz, 1H), 7.45 (s, 1H), 7.19 (t, J = 7.4 Hz, 1H), 6.48 (s, 1H), 2.85 (d, J = 4.3 Hz, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.61, 158.23, 155.75, 155.31, 143.73, 140.58, 136.42, 132.30, 129.17, 128.93, 128.47, 127.43, 122.17, 121.40, 120.81, 120.32, 117.77, 110.70, 105.36, 102.01, 26.86. HRMS: calcd. C21H19ClN7O [M + H] +, 420.1334, found: 4202.1332.

(E)-2-((5-chloro-2–(2-(1–(4-methoxyphenyl)ethylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14ac)

White solid; yield: 56%. M.p. 216.7–217.1 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.95 (s, 1H), 10.08 (s, 1H), 9.48 (d, J = 8.2 Hz, 1H), 8.79 (d, J = 4.4 Hz, 1H), 8.23 (s, 1H), 7.88 (d, J = 8.8 Hz, 2H), 7.80 (d, J = 7.0 Hz, 1H), 7.57 (t, J = 7.4 Hz, 1H), 7.17 (t, J = 7.3 Hz, 1H), 7.03 (d, J = 8.9 Hz, 2H), 3.83 (s, 3H), 2.84 (d, J = 4.4 Hz, 3H), 2.30 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.59, 160.19, 158.96, 155.76, 155.13, 146.66, 140.58, 132.10, 131.82, 128.42, 127.51, 122.20, 121.48, 120.24, 114.17, 105.66, 55.73, 26.84, 13.75. HRMS: calcd. C21H22ClN6O2 [M + H] +, 425.1487, found: 425.1485.

(E)-2-((5-chloro-2–(2-(1–(4-fluorophenyl)ethylidene)hydrazineyl)pyrimidin-4-yl)amino)-N-methylbenzamide (14ad)

White solid; yield: 54%. M.p. 222.6–223.8 °C. 1H NMR (400 MHz, DMSO-d6), δ 11.96 (s, 1H), 10.20 (s, 1H), 9.43 (d, J = 8.4 Hz, 1H), 8.78 (d, J = 4.5 Hz, 1H), 8.25 (s, 1H), 7.97 (dd, J = 8.8, 5.6 Hz, 2H), 7.87 − 7.73 (m, 1H), 7.58 (dd, J = 11.5, 4.2 Hz, 1H), 7.31 (t, J = 8.8 Hz, 2H), 7.17 (t, J = 7.4 Hz, 1H), 2.84 (d, J = 4.5 Hz, 3H), 2.33 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 169.60, 164.10, 161.66, 158.87, 155.80, 155.12, 145.59, 140.50, 135.81, 132.12, 128.42, 128.17, 128.09, 122.28, 121.49, 120.32, 115.74, 115.52, 106.04, 26.85, 13.82. HRMS: calcd. C20H19ClFN6O [M + H] +, 413.1287, found: 413.1282.

Cell lines and cell culture

Thyroid cancer cells TCP-1 were provided by Wuhan Servicebio Co., Ltd. TCP-1 cells are cultured in RPMI-1640 medium supplemented with 10% foetal bovine serum (FBS), 100 U/mL penicillin, and 0.1 mg/mL streptomycin. All cells are maintained in a humidified incubator at 37 °C with 5% CO2.

MTT assay

The TCP-1 cell line is seeded into a 96-well plate and incubated under conditions of 37 °C and 5% CO2. After the cells adhere, different concentrations of compounds are added and the incubation continues for 48 h under the same conditions. Subsequently, MTT reagent is added to the medium and the incubation is continued for an additional 4 h at 37 °C. After the incubation, the medium is aspirated, and 150 μL of DMSO solution is added to dissolve the purple-blue crystals by shaking. The absorbance of the solution is measured at a wavelength of 490 nm. The IC50 value of the compounds is calculated using SPSS software version 20.0.

Kinase inhibition assay

The 50 µl reaction mixture contains 40 mM Tris, pH 7.4, 10 mM MgCl2, 0.1 mg/ml BSA, 1 mM DTT, 10 µM ATP, Kinase and the enzyme substrate. The compound was diluted in 10% DMSO and 5 µl of the dilution was added to a 50 µl reaction so that the final concentration of DMSO is 1% in all of the reactions. The assay was performed using a Kinase-Glo Plus luminescence kinase assay kit. It measures kinase activity by quantitating the amount of ATP remaining in solution following a kinase reaction. The luminescent signal from the assay is correlated with the amount of ATP present and is inversely correlated with the amount of kinase activity. The IC50 values were calculated using nonlinear regression with normalised dose-response fit using Prism GraphPad software.

Molecular docking

The X-ray crystal structure of 2JKK (PDB: 2JKK) was retrieved from the Protein Data Bank. The protonation state of compound 14f was set at pH = 7.4, and the compound 14f were expanded to 3D structures using Open Babel. MOE was applied to prepare and parametrise the receptor protein and ligands. The docking grid documents were generated by Auto Grid of sitemap, and MOE was used for docking simulation. The optimal pose was selected to analyse interaction. Finally, the protein-ligand interaction figure was generated by PyMOL.

ADMET prediction and drug-likeness properties

Here, we used the Swiss ADME Online software (www.SwissADME.ch) to measure in the silico ADME profile and drug-likeness properties.

Western blot

Different concentrations of compound 14f are applied to TCP-1 cells for 48 h, after which the cells are collected and lysed in RIPA lysis buffer (Solarbio, R0010). Protein concentration is determined using the BCA Protein Concentration Assay Kit (Solarbio, PC0020). Proteins were denatured using 4X SDS-PAGE loading buffer (Solarbio, P1016) at 100 °C for 10 min. The denatured protein samples are separated by SDS-PAGE electrophoresis. Proteins are transferred from the gel to nitrocellulose (NC) membrane (Pall Corporation, P/N 66485) and blocked with 5% non-fat milk for 1 h at room temperature. After washing with TBST, the primary antibody is added and incubated overnight at 4 °C. The membrane is washed with TBST and then incubated with the secondary antibody at room temperature for 1–2 h. The membrane is washed again with TBST, and the protein bands are visualised using an ECL (Solarbio, PE0010) ultra-sensitive luminescent reagent, followed by imaging with a chemiluminescence imaging analyser (Cytiva, AMERSHAM ImageQuant 800, JPN). The ladder used for Western Blot experiments was purchased from Thermo Scientific (Thermo Scientific, catalog number:26616). Information on the antibodies used in this study is provided below: Anti-FAK-antibody (Cell Signalling Technology, #3285S), Phospho-FAK (Tyr925) Antibody (Cell Signalling Technology, #3284S), Phospho-FAK (Tyr576/577) Antibody (Cell Signalling Technology, #3281S), Phospho-FAK (Tyr397) Antibody (Bioss, bs-3159R), β-Actin (Abways, AB0035), Goat Anti-Rabbit IgG (H + L) HRP (Cell Signalling Technology, #35401).

Colony formatting assay

TCP-1 cells are plated at a density of 10,000 to 20,000 cells per well in a 6-well plate. After cell adhesion, the cells are treated with different concentrations of compound 14f and cultured for 5 days in an incubator. At the end of the culture period, the medium is aspirated, and the cells are washed three times with PBS. The cells are then fixed with 4% paraformaldehyde for 20 min at room temperature. After fixation, the fixative is removed, and the cells are stained with 0.1% crystal violet at room temperature for 20 min. Following staining, the cells are rinsed with PBS until the purple colour is completely washed away, and then air-dried for image acquisition.

EdU staining

TCP-1 cells are treated with compound 14f, followed by the addition of an equal volume of 1X EdU working solution to the drug-containing medium, and incubated under conditions of 37 °C and 5% CO2 for 4 h. After incubation, the cells are fixed with 4% paraformaldehyde for 15 min. The fixative is then removed, and the cells are permeabilized with PBS containing 0.3% Triton X-100 at room temperature. After washing, the Click reaction working solution is added and incubated at 37 °C for 30 min. The cells are then washed and stained with a prepared Hoechst staining solution for 10 min to stain the cell nuclei. After staining, the staining solution is removed, the cells are washed, and images are captured and saved using a fluorescence microscope.

Wound healing assay

TCP-1 cells are seeded into a 6-well cell culture plate and cultured in an incubator at 37 °C with 5% CO2. Once the cells uniformly cover the bottom of the well, a vertical line is drawn in the centre of each well to mark the initial scratch. Fresh medium is replaced and different concentrations of 14f are added. Photographs are taken to document the healing at 0 h, 24 h, 36 h and 48 h post-scratch. Images are processed using ImageJ software. Data analysis is performed using GraphPad Prism software version 8.0.

Transwell assay

A 24-well plate is filled with 500 μL of RPMI-1640 medium containing 20% serum per well, and Transwell chambers are placed in the wells. TCP-1 cells are resuspended in serum-free medium and mixed with compound 14f, then added to the upper chamber of the Transwell chambers. The 24-well plates are incubated at 37 °C with 5% CO2 for 48 h. After incubation, the plates are removed, and the chambers are immersed in 4% paraformaldehyde and fixed at room temperature for 10 min. Following fixation, 0.1% crystal violet staining solution is added to the 24-well plates, and the chambers are stained at room temperature for 30 min. After staining, the plates are washed, the upper layer of cells is gently wiped off with a cotton swab, and the chambers are air-dried. Images are captured using a microscope.

Supplementary Material

Supplemental Material
Original Image for Figure 7A 36h Control_2.tif
Original Image for Figure 7A 0h Control_3.tif
Original Image for Figure 7A 12h 300 nM_3.tif
Original Image for Figure 7A 0h 600 nM_1.tif
Original Image for Figure 7A 0h 600 nM_2.tif
Original Image for Figure 7A 24h 600 nM_3.tif
Supporting information.docx
Original Image for Figure 7A 12h 600 nM_3.tif
Original Image for Figure 7A 0h 900 nM_2.tif
Original Image for Figure 6B 300 nM.TIF
Original Image for Figure 6A 300 nM_Hoechst.tif
Original Image for Figure 7A 48h 600 nM_2.tif
Original Image for Figure 7A 36h 900 nM_2.tif
Original Image for Figure 6A 900 nM_Hoechst.tif
Original Image for Figure 7A 12h 600 nM_2.tif
Original Image for Figure 7A 36h 900 nM_3.tif
Original Image for Figure 7C 600 nM.TIF
Original Image for Figure 6A 300 nM_EdU.tif
Original Image for Figure 6B Control.TIF
Original Image for Figure 6A control_EdU.tif
Original Image for Figure 7A 12h 900 nM_1.tif
Original Image for Figure 7A 36h 300 nM_1.tif
Original Image for Figure 7A 0h 600 nM_3.tif
Original Image for Figure 7A 24h 600 nM_1.tif
Original Image for Figure 7A 24h Control_1.tif
Original Image for Figure 7A 24h 300 nM_3.tif
Original Image for Fig 5B_p_FAK Y397.tif
Original Image for Figure 6B 500 nM.TIF
Original Image for Figure 7A 48h 600 nM_1.tif
Original Image for Fig 5B_p_FAK Y925.tif
Original Image for Figure 7A 12h 300 nM_1.tif
Original Image for Figure 7A 24h 900 nM_1.tif
Original Image for Figure 7A 24h 600 nM_2.tif
Original Image for Fig 5B_β_actin.tif
Original Image for Figure 7A 12h 600 nM_1.tif
Original Image for Figure 7A 36h 600 nM_1.tif
Original Image for Figure 7A 36h 600 nM_2.tif
Original Image for Figure 7A 36h 600 nM_3.tif
Original Image for Figure 6B 700 nM.TIF
Original Image for Figure 7C Control.TIF
Original Image for Figure 7A 36h Control_3.tif
Original Image for Figure 7A 12h Control_3.tif
Original Image for Figure 7A 48h Control_3.tif
Original Image for Figure 7A 0h Control_1.tif
Original Image for Figure 7A 36h Control_1.tif
Original Image for Figure 6A 600 nM_Hoechst.tif
Original Image for Figure 6B 900 nM.TIF
Original Image for Figure 7A 48h 300 nM_1.tif
Original Image for Figure 7A 24h Control_2.tif
Original Image for Figure 7A 48h 900 nM_2.tif
Original Image for Figure 7A 24h 300 nM_2.tif
Original Image for Figure 7A 48h 300 nM_3.tif
Original Image for Figure 7A 48h 600 nM_3.tif
Original Image for Figure 7A 24h 300 nM_1.tif
Original Image for Figure 7C 300 nM.TIF
Original Image for Figure 7A 48h 300 nM_2.tif
Original Image for Figure 7A 48h 900 nM_3.tif
Original Image for Figure 7A 48h 900 nM_1.tif
Original Image for Figure 6A control_Hoechst.tif
Original Image for Figure 7A 0h 900 nM_1.tif
Original Image for Figure 7A 36h 300 nM_2.tif
Original Image for Figure 7A 0h 300 nM_3.tif
Original Image for Figure 7A 12h Control_2.tif
Original Image for Figure 7A 0h 300 nM_2.tif
Original Image for Figure 7A 36h 900 nM_1.tif
Original Image for Figure 7A 24h Control_3.tif
Original Image for Figure 7A 0h 900 nM_3.tif
Original Image for Figure 7A 48h Control_2.tif
Original Image for Figure 7A 48h Control_1.tif
Original Image for Figure 7A 12h Control_1.tif
Original Image for Fig 5B_FAK.tif
Original Image for Figure 7A 12h 300 nM_2.tif
Original Image for Figure 7A 24h 900 nM_3.tif
Original Image for Figure 7A 0h 300 nM_1.tif
Original Image for Figure 7A 24h 900 nM_2.tif
Original Image for Figure 6A 600 nM_EdU.tif
Original Image for Figure 7A 36h 300 nM_3.tif
Original Image for Figure 7A 12h 900 nM_3.tif
Original Image for Figure 7C 900 nM.TIF
Original Image for Fig 5B_p_FAK Y576 577.tif
Original Image for Figure 7A 12h 900 nM_2.tif
Original Image for Figure 7A 0h Control_2.tif

Acknowledgements

The authors are thankful to the National Natural Science Foundation of China, the National Natural Science Foundation of Henan, and the Training Program for Young Key Teachers of Colleges and Universities in Henan Province for funding this work.

Funding Statement

This work was supported by the National Natural Science Foundation of China [No. U2004123 and 82273782], the National Natural Science Foundation of Henan [No. 242300421472] and the Training Program for Young Key Teachers of Colleges and Universities in Henan Province [2023GGJS008].

Author contributions

Hongting Li and Lijun Fu is responsible for conception and design of this study. Mei-Qi Jia, Zhao-Long Qin, Changliang Lu, Weili Chu, Ze Zhang and Jinbo Niu performed the experiments and analysed the data. Jian Song and Sai-Yang Zhang performed molecular simulation. Hongting Li and Mei-Qi Jia wrote the initial draft of the manuscript. Lijun Fu, Jian Song and Sai-Yang Zhang reviewed the submitted manuscript. All authors approved the final version of the manuscript.

Disclosure statement

Sai-Yang Zhang is an associated editor of the Journal of Enzyme Inhibition and Medicinal Chemistry. The authors declared that there was no conflict of interest about this work.

Data availability statement

Additional data may be requested from the authors.

References

  • 1.Chen DW, Lang BHH, McLeod DSA, Newbold K, Haymart MR.. Thyroid cancer. Lancet. 2023;401(10387):1531–1544. [DOI] [PubMed] [Google Scholar]
  • 2.Volpe F, Nappi C, Zampella E, Di Donna E, Maurea S, Cuocolo A, Klain M.. Current advances in radioactive iodine-refractory differentiated thyroid cancer. Curr Oncol. 2024;31(7):3870–3884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Boucai L, Zafereo M, Cabanillas ME.. Thyroid cancer: a review. JAMA. 2024;331(5):425–435. [DOI] [PubMed] [Google Scholar]
  • 4.Bhattacharya S, Mahato RK, Singh S, Bhatti GK, Mastana SS, Bhatti JS.. Advances and challenges in thyroid cancer: the interplay of genetic modulators, targeted therapies, and AI-driven approaches. Life Sci. 2023;332:122110. [DOI] [PubMed] [Google Scholar]
  • 5.Zhang Y, Xing Z, Liu T, Tang M, Mi L, Zhu J, Wu W, Wei T.. Targeted therapy and drug resistance in thyroid cancer. Eur J Med Chem. 2022;238:114500. [DOI] [PubMed] [Google Scholar]
  • 6.Fallahi P, Ferrari SM, Galdiero MR, Varricchi G, Elia G, Ragusa F, Paparo SR, Benvenga S, Antonelli A.. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin Cancer Biol. 2022;79:180–196. [DOI] [PubMed] [Google Scholar]
  • 7.La Pietra V, Sartini S, Botta L, Antonelli A, Ferrari SM, Fallahi P, Moriconi A, Coviello V, Quattrini L, Ke Y-Y, et al. Challenging clinically unresponsive medullary thyroid cancer: discovery and pharmacological activity of novel RET inhibitors. Eur J Med Chem. 2018;150:491–505. [DOI] [PubMed] [Google Scholar]
  • 8.Kong X, Pan P, Sun H, Xia H, Wang X, Li Y, Hou T.. Drug discovery targeting Anaplastic Lymphoma Kinase (ALK). J Med Chem. 2019;62(24):10927–10954. [DOI] [PubMed] [Google Scholar]
  • 9.Gerninghaus J, Zhubi R, Krämer A, Karim M, Tran DHN, Joerger AC, Schreiber C, Berger LM, Berger B-T, Ehret TAL, et al. Back-pocket optimization of 2-aminopyrimidine-based macrocycles leads to potent EPHA2/GAK kinase inhibitors. J Med Chem. 2024;67(15):12534–12552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chen Y, Singh R, Lin N, Taylor V, Masuda ES, Payan DG.. Discovery of 5-Aryl-2,4-diaminopyrimidine compounds as potent and selective IRAK4 inhibitors. ACS Med Chem Lett. 2022;13(4):714–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wu T, Qin Q, Liu N, Zhang C, Lv R, Yin W, Sun Y, Sun Y, Wang R, Zhao D, et al. Rational drug design to explore the structure-activity relationship (SAR) of TRK inhibitors with 2,4-diaminopyrimidine scaffold. Eur J Med Chem. 2022;230:114096. [DOI] [PubMed] [Google Scholar]
  • 12.Zhang J, Xu K, Yang F, Qiu Y, Li J, Li J, Wang W, Tan G, Zou Z, Kang F.. Design, synthesis and evaluation of nitric oxide releasing derivatives of 2,4-diaminopyrimidine as novel FAK inhibitors for intervention of metastatic triple-negative breast cancer. Eur J Med Chem. 2023;250:115192. [DOI] [PubMed] [Google Scholar]
  • 13.Zhang Y, Tong L, Yan F, Huang P, Zhu C-L, Pan C.. Design, synthesis, and antitumor activity evaluation of potent fourth-generation EGFR inhibitors for treatment of Osimertinib resistant non-small cell lung cancer (NSCLC). Bioorg Chem. 2024;147:107394. [DOI] [PubMed] [Google Scholar]
  • 14.Wang X, Qin ZL, Li N, Jia MQ, Liu QG, Bai YR, Song J, Yuan S, Zhang SY.. Annual review of PROTAC degraders as anticancer agents in 2022. Eur J Med Chem. 2024;267:116166. [DOI] [PubMed] [Google Scholar]
  • 15.Marsilje TH, Pei W, Chen B, Lu W, Uno T, Jin Y, Jiang T, Kim S, Li N, Warmuth M, et al. Synthesis, structure–activity relationships, and in vivo efficacy of the novel potent and selective Anaplastic Lymphoma Kinase (ALK) Inhibitor 5-Chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (LDK378) Currently in Phase 1 and Phase 2 Clinical Trials. J Med Chem. 2013;56(14):5675–5690. [DOI] [PubMed] [Google Scholar]
  • 16.Blunt MD, Koehrer S, Dobson RC, Larrayoz M, Wilmore S, Hayman A, Parnell J, Smith LD, Davies A, Johnson PWM, et al. The Dual Syk/JAK inhibitor cerdulatinib antagonizes B-cell receptor and microenvironmental signaling in chronic lymphocytic Leukemia. Clin Cancer Res. 2017;23(9):2313–2324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yan Q, Chen Y, Tang B, Xiao Q, Qu R, Tong L, Liu J, Ding J, Chen Y, Ding N, et al. Discovery of novel 2,4-diarylaminopyrimidine derivatives as potent and selective epidermal growth factor receptor (EGFR) inhibitors against L858R/T790M resistance mutation. Eur J Med Chem. 2018;152:298–306. [DOI] [PubMed] [Google Scholar]
  • 18.Lu Y, Sun H.. Progress in the development of small molecular inhibitors of Focal Adhesion Kinase (FAK). J Med Chem. 2020;63(23):14382–14403. [DOI] [PubMed] [Google Scholar]
  • 19.Qin Q, Wang R, Fu Q, Zhang G, Wu T, Liu N, Lv R, Yin W, Sun Y, Sun Y, et al. Design, synthesis, and biological evaluation of potent FAK-degrading PROTACs. J Enzyme Inhib Med Chem. 2022;37(1):2241–2255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Shapiro G, Rosen LS, Tolcher AW, Goldman JW, Gandhi L, Papadopoulos KP, Tolaney SM, Beeram M, Rasco DW, Kulanthaivel P, et al. A first-in-human phase I study of the CDK4/6 inhibitor, LY2835219, for patients with advanced cancer. JCO. 2013;31(15_suppl):2500–2500. [Google Scholar]
  • 21.Li Z, Jiang K, Zhu X, Lin G, Song F, Zhao Y, Piao Y, Liu J, Cheng W, Bi X, et al. Encorafenib (LGX818), a potent BRAF inhibitor, induces senescence accompanied by autophagy in BRAFV600E melanoma cells. Cancer Lett. 2016;370(2):332–344. [DOI] [PubMed] [Google Scholar]
  • 22.Liu Y, Kong L-J, Li N, Liu Y-H, Jia M-Q, Liu Q-G, Zhang S-Y, Song J.. Design, synthesis and biological evaluation of novel 2,4-diaminopyrimidine cinnamyl derivatives as inhibitors of FAK with potent anti-gastric cancer activities. Bioorg Chem. 2023;141:106895. [DOI] [PubMed] [Google Scholar]
  • 23.Song J, Liu X, Zhang Y-F, Tian X-Y, Deng M-Y, Huang C-Z, Zhang S-Y.. The dual FAK-HDAC inhibitor MY-1259 displays potent activities in gastric cancers in vitro and in vivo. Bioorg Chem. 2023;131:106328. [DOI] [PubMed] [Google Scholar]
  • 24.Tasneem S, Sheikh KA, Naematullah M, Mumtaz Alam M, Khan F, Garg M, Amir M, Akhter M, Amin S, Haque A, et al. Synthesis, biological evaluation and docking studies of methylene bearing cyanopyrimidine derivatives possessing a hydrazone moiety as potent Lysine specific demethylase-1 (LSD1) inhibitors: a promising anticancer agents. Bioorg Chem. 2022;126:105885. [DOI] [PubMed] [Google Scholar]
  • 25.Salum LB, Mascarello A, Canevarolo RR, Altei WF, Laranjeira AB, Neuenfeldt PD, Stumpf TR, Chiaradia-Delatorre LD, Vollmer LL, Daghestani HN, et al. N-(1’-naphthyl)-3,4,5-trimethoxybenzohydrazide as microtubule destabilizer: Synthesis, cytotoxicity, inhibition of cell migration and in vivo activity against acute lymphoblastic leukemia. Eur J Med Chem. 2015;96:504–518. [DOI] [PubMed] [Google Scholar]
  • 26.Li ZH, Yang DX, Geng PF, Zhang J, Wei HM, Hu B, Guo Q, Zhang XH, Guo WG, Zhao B, et al. Design, synthesis and biological evaluation of [1,2,3]triazolo[4,5-d]pyrimidine derivatives possessing a hydrazone moiety as antiproliferative agents. Eur J Med Chem. 2016;124:967–980. [DOI] [PubMed] [Google Scholar]
  • 27.Abdel-Aziz AA, El-Azab AS, AlSaif NA, Obaidullah AJ, Al-Obaid AM, Al-Suwaidan IA.. Synthesis, potential antitumor activity, cell cycle analysis, and multitarget mechanisms of novel hydrazones incorporating a 4-methylsulfonylbenzene scaffold: a molecular docking study. J Enzyme Inhib Med Chem. 2021;36(1):1521–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ashmawy FO, Gomha SM, Abdallah MA, Zaki MEA, Al-Hussain SA, El-Desouky MA.. Synthesis, in vitro evaluation and molecular docking studies of novel thiophenyl thiazolyl-pyridine hybrids as potential anticancer agents. Molecules. 2023;28(11):4270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Altıntop MD, Ertorun İ, Akalın Çiftçi G, Özdemir A.. Design, synthesis and biological evaluation of a new series of imidazothiazole-hydrazone hybrids as dual EGFR and Akt inhibitors for NSCLC therapy. Eur J Med Chem. 2024;276:116698. DOI: [DOI] [PubMed] [Google Scholar]
  • 30.Ahmed MF, El-Haggar R, Almalki AH, Abdullah O, El Hassab MA, Masurier N, Hammad SF.. Novel hydrazone-isatin derivatives as potential EGFR inhibitors: Synthesis and in vitro pharmacological profiling. Arch Pharm. 2023;356(9):e2300244. [DOI] [PubMed] [Google Scholar]
  • 31.Wang Y, Zhang G, Hu G, Bu Y, Lei H, Zuo D, Han M, Zhai X, Gong P.. Design, synthesis and biological evaluation of novel 4-arylaminopyrimidine derivatives possessing a hydrazone moiety as dual inhibitors of L1196M ALK and ROS1. Eur J Med Chem. 2016;123:80–89. DOI: [DOI] [PubMed] [Google Scholar]
  • 32.Yap HY, Yap BS, Blumenschein GR, Barnes BC, Schell FC, Bodey GP.. Bisantrene, an active new drug in the treatment of metastatic breast cancer. Cancer Res. 1983;43(3):1402–1404. [PubMed] [Google Scholar]
  • 33.Dai XJ, Liu Y, Xue LP, Xiong XP, Zhou Y, Zheng YC, Liu HM.. Reversible Lysine Specific Demethylase 1 (LSD1) inhibitors: a promising wrench to impair LSD1. J Med Chem. 2021;64(5):2466–2488. [DOI] [PubMed] [Google Scholar]
  • 34.Lu D, Chen J, Qin L, Bijou I, Yi P, Li F, Song X, MacKenzie KR, Yu X, Yang B, et al. Lead compound development of SRC-3 inhibitors with improved pharmacokinetic properties and anticancer efficacy. J Med Chem. 2024;67(7):5333–5350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bedewy WA, Mohamed MS, Abdelhameed AM, Elsawy MA, Al-Muhur M, Ashida N, Abdalla AN, Elwaie TA, Nagamatsu T, Ali HI.. Design, synthesis, and antitumor efficacy of novel 5-deazaflavin derivatives backed by kinase screening, docking, and ADME studies. J Enzyme Inhib Med Chem. 2023;38(1):2220570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Liu TJ, LaFortune T, Honda T, Ohmori O, Hatakeyama S, Meyer T, Jackson D, de Groot J, Yung WK.. Inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor kinase suppresses glioma proliferation in vitro and in vivo. Mol Cancer Ther. 2007;6(4):1357–1367. [DOI] [PubMed] [Google Scholar]
  • 37.Wang X, Li N, Liu Y-H, Wu J, Liu Q-G, Niu J-B, Xu Y, Huang C-Z, Zhang S-Y, Song J.. Targeting focal adhesion kinase (FAK) in cancer therapy: A recent update on inhibitors and PROTAC degraders. Eur J Med Chem. 2024;276:116678. [DOI] [PubMed] [Google Scholar]
  • 38.Wang L, Ai M, Yu J, Jin L, Wang C, Liu Z, Shu X, Tang Z, Liu K, Luo H, et al. Structure-based modification of carbonyl-diphenylpyrimidines (Car-DPPYs) as a novel focal adhesion kinase (FAK) inhibitor against various stubborn cancer cells. Eur J Med Chem. 2019;172:154–162. [DOI] [PubMed] [Google Scholar]
  • 39.Ai M, Wang C, Tang Z, Liu K, Sun X, Ma T, Li Y, Ma X, Li L, Chen L.. Design and synthesis of diphenylpyrimidine derivatives (DPPYs) as potential dual EGFR T790M and FAK inhibitors against a diverse range of cancer cell lines. Bioorg Chem. 2020;94:103408. [DOI] [PubMed] [Google Scholar]
  • 40.Pecoraro C, Carbone D, Scianò F, Terrana F, Xu G, Bergonzini C, Roeten MSF, Cascioferro S, Cirrincione G, Diana P, et al. Exploring the therapeutic potential of a novel series of imidazothiadiazoles targeting focal adhesion kinase (FAK) for pancreatic cancer treatment: synthesis, mechanistic insights and promising antitumor and safety profile. J Drug Target. 2024;2024:1–17. [DOI] [PubMed] [Google Scholar]
  • 41.Ye YX, Cao YY, Xu LS, Wang HC, Liu XH, Zhu HL.. FAK inhibitors in cancer, a patent review – an update on progress. Expert Opin Ther Pat. 2024;34(8):593–610. [DOI] [PubMed] [Google Scholar]
  • 42.Scianò F, Terrana F, Pecoraro C, Parrino B, Cascioferro S, Diana P, Giovannetti E, Carbone D.. Exploring the therapeutic potential of focal adhesion kinase inhibition in overcoming chemoresistance in pancreatic ductal adenocarcinoma. Future Med Chem. 2024;16(3):271–289. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Material
Original Image for Figure 7A 36h Control_2.tif
Original Image for Figure 7A 0h Control_3.tif
Original Image for Figure 7A 12h 300 nM_3.tif
Original Image for Figure 7A 0h 600 nM_1.tif
Original Image for Figure 7A 0h 600 nM_2.tif
Original Image for Figure 7A 24h 600 nM_3.tif
Supporting information.docx
Original Image for Figure 7A 12h 600 nM_3.tif
Original Image for Figure 7A 0h 900 nM_2.tif
Original Image for Figure 6B 300 nM.TIF
Original Image for Figure 6A 300 nM_Hoechst.tif
Original Image for Figure 7A 48h 600 nM_2.tif
Original Image for Figure 7A 36h 900 nM_2.tif
Original Image for Figure 6A 900 nM_Hoechst.tif
Original Image for Figure 7A 12h 600 nM_2.tif
Original Image for Figure 7A 36h 900 nM_3.tif
Original Image for Figure 7C 600 nM.TIF
Original Image for Figure 6A 300 nM_EdU.tif
Original Image for Figure 6B Control.TIF
Original Image for Figure 6A control_EdU.tif
Original Image for Figure 7A 12h 900 nM_1.tif
Original Image for Figure 7A 36h 300 nM_1.tif
Original Image for Figure 7A 0h 600 nM_3.tif
Original Image for Figure 7A 24h 600 nM_1.tif
Original Image for Figure 7A 24h Control_1.tif
Original Image for Figure 7A 24h 300 nM_3.tif
Original Image for Fig 5B_p_FAK Y397.tif
Original Image for Figure 6B 500 nM.TIF
Original Image for Figure 7A 48h 600 nM_1.tif
Original Image for Fig 5B_p_FAK Y925.tif
Original Image for Figure 7A 12h 300 nM_1.tif
Original Image for Figure 7A 24h 900 nM_1.tif
Original Image for Figure 7A 24h 600 nM_2.tif
Original Image for Fig 5B_β_actin.tif
Original Image for Figure 7A 12h 600 nM_1.tif
Original Image for Figure 7A 36h 600 nM_1.tif
Original Image for Figure 7A 36h 600 nM_2.tif
Original Image for Figure 7A 36h 600 nM_3.tif
Original Image for Figure 6B 700 nM.TIF
Original Image for Figure 7C Control.TIF
Original Image for Figure 7A 36h Control_3.tif
Original Image for Figure 7A 12h Control_3.tif
Original Image for Figure 7A 48h Control_3.tif
Original Image for Figure 7A 0h Control_1.tif
Original Image for Figure 7A 36h Control_1.tif
Original Image for Figure 6A 600 nM_Hoechst.tif
Original Image for Figure 6B 900 nM.TIF
Original Image for Figure 7A 48h 300 nM_1.tif
Original Image for Figure 7A 24h Control_2.tif
Original Image for Figure 7A 48h 900 nM_2.tif
Original Image for Figure 7A 24h 300 nM_2.tif
Original Image for Figure 7A 48h 300 nM_3.tif
Original Image for Figure 7A 48h 600 nM_3.tif
Original Image for Figure 7A 24h 300 nM_1.tif
Original Image for Figure 7C 300 nM.TIF
Original Image for Figure 7A 48h 300 nM_2.tif
Original Image for Figure 7A 48h 900 nM_3.tif
Original Image for Figure 7A 48h 900 nM_1.tif
Original Image for Figure 6A control_Hoechst.tif
Original Image for Figure 7A 0h 900 nM_1.tif
Original Image for Figure 7A 36h 300 nM_2.tif
Original Image for Figure 7A 0h 300 nM_3.tif
Original Image for Figure 7A 12h Control_2.tif
Original Image for Figure 7A 0h 300 nM_2.tif
Original Image for Figure 7A 36h 900 nM_1.tif
Original Image for Figure 7A 24h Control_3.tif
Original Image for Figure 7A 0h 900 nM_3.tif
Original Image for Figure 7A 48h Control_2.tif
Original Image for Figure 7A 48h Control_1.tif
Original Image for Figure 7A 12h Control_1.tif
Original Image for Fig 5B_FAK.tif
Original Image for Figure 7A 12h 300 nM_2.tif
Original Image for Figure 7A 24h 900 nM_3.tif
Original Image for Figure 7A 0h 300 nM_1.tif
Original Image for Figure 7A 24h 900 nM_2.tif
Original Image for Figure 6A 600 nM_EdU.tif
Original Image for Figure 7A 36h 300 nM_3.tif
Original Image for Figure 7A 12h 900 nM_3.tif
Original Image for Figure 7C 900 nM.TIF
Original Image for Fig 5B_p_FAK Y576 577.tif
Original Image for Figure 7A 12h 900 nM_2.tif
Original Image for Figure 7A 0h Control_2.tif

Data Availability Statement

Additional data may be requested from the authors.


Articles from Journal of Enzyme Inhibition and Medicinal Chemistry are provided here courtesy of Taylor & Francis

RESOURCES