Next Article in Journal
Reconstructing the Antibiotic Pipeline: Natural Alternatives to Antibacterial Agents
Previous Article in Journal
Complement Cascades and Brain Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of Novel 2-Substituted Aniline Pyrimidine Based Derivatives as Potent Mer/c-Met Dual Inhibitors with Improvement Bioavailability

1
School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
2
School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(8), 1180; https://doi.org/10.3390/biom15081180
Submission received: 5 May 2025 / Revised: 1 August 2025 / Accepted: 8 August 2025 / Published: 18 August 2025
(This article belongs to the Section Molecular Medicine)

Abstract

This study reports the rational design and systematic evaluation of a novel series of 2-substituted aniline pyrimidine derivatives as dual Mer/c-Met inhibitors. Among the synthesized compounds, 17c demonstrated potent dual kinase inhibition, with IC50 values of 6.4 ± 1.8 nM (Mer) and 26.1 ± 7.7 nM (c-Met). The compound exhibited significant antiproliferative activity across multiple cancer cell lines (HepG2, MDA-MB-231, and HCT116), while showing minimal hERG channel inhibition (IC50 > 40 μM), indicating favorable cardiac safety. Pharmacokinetic profiling revealed high metabolic stability in human liver microsomes (t1/2 = 53.1 min) and moderate oral bioavailability (F: 45.3%), with strong plasma protein-binding affinity (>95%). Mechanistic studies further demonstrated that 17c dose-dependently suppressed HCT116 cell migration and induced apoptosis. These integrated pharmacological properties position 17c as a promising therapeutic candidate for dual Mer/c-Met drive malignancies.

1. Introduction

Mer kinase (monocytes epithelial tissue reproductive tissue), a key member of the TAM (Tyro3-Axl-Mer) receptor tyrosine kinase family [1,2,3], orchestrates multiple cellular processes critical to immune regulation, apoptosis resistance, and inflammatory signaling. Structurally, Mer is characterized by a conserved intracellular tyrosine kinase domain and extracellular ligand-binding region. Its expression is predominantly localized to immune cells (e.g., macrophages and dendritic cells), where it mediates efferocytosis (apoptotic cell clearance) and suppresses pro-inflammatory cytokine production, thereby maintaining immune tolerance [4]. Mechanistically, Mer activation occurs through Gas6 ligand binding, initiating downstream signaling cascades, e.g., PI3K/AKT (phosphatidylinositol 3-kinase/protein kinase B) and MAPK (mitogen-activated protein kinase), that promote cell survival, chemotaxis, and tissue remodeling [4]. Importantly, Mer dysregulation has been pathologically linked to oncogenesis: overexpression in tumor cells enhances survival under chemotherapy (via BCL-2 upregulation), facilitates metastatic dissemination (through EMT activation), and fosters immunosuppressive microenvironments (by polarizing tumor-associated macrophages) [5,6,7,8]. These multifaceted roles have established Mer as a high-value therapeutic target for precision oncology strategies targeting both tumor-intrinsic and immune evasion mechanisms.
Current Mer kinase inhibitors are structurally categorized into two major chemotypes: aminopyrimidine-pyrazole (pyrrole) and aminopyrimidine derivatives (Figure 1). The first-generation inhibitor UNC569 [9,10], a pyrazole-based compound, selectively inhibits Mer kinase (IC50 = 1.2 nM) and suppresses downstream ERK (extracellular signal-regulated kinase/AKT signaling. However, its clinical translation is hindered by its rapid metabolism (human microsomal t1/2< 15 min) and low oral bioavailability (F < 10%) [11]. The structural optimization of UNC569 yielded second-generation analogs. UNC2025 enhanced metabolic stability (t1/2 = 68 min in human hepatocytes) and maintained Mer potency (IC50 = 1.8 nM) [12,13]. MRX2843, a dual Mer/Flt3 (fms-like tyrosine kinase 3) inhibitor (Mer IC50 = 1.2 nM), showed improved CNS penetration (brain/plasma ration = 0.4) [11] and is currently in phase II trails for relapsed/refractory acute lymphoblastic leukemia (NCT04872478) [14]. Notably, the aminopyrimidine-class inhibitor UNC2250 demonstrates exceptional Mer targeting (IC50 = 1.7 nM) and unique activity against Mer-EGFR (epidermal growth factor receptor) fusion protein [15,16]. In xenograft models, UNC2250 induced tumor regression (~60% volume reduction vs. control) by the dual blockade of Mer-mediated survival signaling and EGFR-driven proliferation [16].
The c-Met kinase (cellular mesenchymal–epithelial transition factor), also known as the hepatocyte growth factor receptor (HGFR) [17,18], is a transmembrane receptor tyrosine kinase that plays a pivotal role in regulating critical cellular processes, including proliferation, survival, migration, and angiogenesis [19,20]. The activation of c-Met occurs through binding to its specific ligand HGF, which triggers receptor dimerization and autophosphorylation. This activation initiates downstream signaling cascades, such as the MAPK, PI3K/AKT, and STAT (signal transducer and activator of transcription) pathways. Aberrant c-Met signaling, caused by overexpression, mutations, or gene amplification, is strongly associated with tumorigenesis, promoting cancer progression, metastasis, and therapy resistance. Given its oncogenic significance, c-Met has become a promising therapeutic target. Current clinical investigations are actively exploring both small-molecule inhibitors and monoclonal antibodies targeting c-Met for the treatment of diverse malignancies.
Multiple c-Met inhibitors have entered clinical practice or are under clinical investigation (Figure 2); these could be classified into Type I and Type II inhibitors based on their binding modes to the target kinase. Notably, crizotinib (Type I), approved by the FDA in 2011, is primarily used for ALK (anaplastic lymphoma kinase)-positive metastatic non-small cell lung cancer (NSCLC) but also exhibits c-Met inhibitory activity as part of its multi-target mechanism [21,22]. Cabozantinib (Type II), another tyrosine inhibitor targeting c-Met, VEGFR (vascular endothelial growth factor receptor), Mer, and Kit, has been approved for multiple indications: metastatic medullary thyroid cancer, as a second-line treatment of advanced renal cell carcinoma post-antiangiogenic therapy, and as a first-line therapy for advanced renal cell carcinoma [23,24]. Additionally, savolitinib (Type I), a highly selective c-Met inhibitor, is currently in Phase III clinical trials for papillary renal cell carcinoma and other c-Met-driven malignancies [25]. Capmatinib (Type I), a selective c-Met inhibitor, received FDA approval specifically for metastatic NSCLC with MET exon 14 skipping mutations [26,27].
Dual-target inhibition strategies have gained significant traction in anti-tumor drug development, as exemplified by the FDA-approved agent cabozantinib-a potent c-Met/VEGFR-2 dual kinase inhibitor. Structurally, cabozantinib’s quinoline core competitively binds to the hinge region of c-Met kinase, while its cyclopropylamide side chain enables optimal hydrophobic interaction within the VEGFR-2 active site. The bifunction design confers robust inhibitory activity against both targets, underpinning its clinical approval for advanced thyroid carcinoma, hepatocellular carcinoma (HCC), and renal cell carcinoma (RCC). Additionally, the dual-target strategy has also been effectively applied in other disease areas, such as in the treatment of neglected diseases and neurological disorders [28,29]. Consequently, the development of dual inhibitors represents a paradigm shift in oncology drug discovery, balancing enhanced therapeutic outcomes with improved safety profiles through rationally engineered polypharmacology.
Both Mer and c-Met, members of the receptor tyrosine kinase family, possess structurally homologous extracellular domains and share overlapping downstream signaling pathways. Notably, their functional convergence in regulating tumor proliferation, metastasis, and immune evasion establishes a compelling rationale for the dual targeting of Mer and c-Met kinases. To date, cabozantinib and its derivatives remain the only clinical available agent with demonstrated inhibitory activity against both targets [30]. However, cabozantinib’s clinical utility is predominantly attributed to its potent inhibition of c-Met and VEGFR-2, while the structural determinants governing its Mer kinase inhibitory activity remains poorly characterized. Furthermore, the emergence of cabozantinib resistance underscores the critical need to develop novel Mer/c-Met dual inhibitors with distinct chemical scaffolds. Importantly, the structure–activity relationship (SAR) of Mer/c-Met dual inhibitors remains underexplored, and the systematic exploration of these aspects through rational drug design is therefore essential to advance the development of next-generation dual-targeting agents.
The 2-substituted aniline scaffold has been widely applied in anti-tumor drug development. For instance, the Mer inhibitor UNC2250 (Figure 1) exhibits potent Mer inhibitory activity [15,16]. Additionally, our previous study identified compound 18c (Figure 5) as a dual Mer/c-Met dual inhibitor with potent dual-target inhibitory activity [31]. The IC50 values were 18.5 ± 2.3 nM and 33.6 ± 4.3 nM, respectively. Furthermore, compound 18c also demonstrated potent antiproliferation activities against HepG2, MDA-MB-231, and HCT116 cancer cell lines and showed a low risk in hERG assays. However, pharmacokinetic analysis revealed low oral bioavailability (F: 2.84%), promoting structural optimization. To elucidate the key interactions in dual-target inhibition, we analyzed the binding modes of cabozantinib with Mer (PDB: 4M3Q) [32,33] and c-Met (PDB: 3LQ8) [34,35]. Molecular docking showed that in Mer kinase, cabozantinib’s amide group forms hydrogen bonds with the hinge region (Pro672 and Met674), while the quinoline moiety occupies a hydrophobic cavity without direct hydrogen bonding (Figure 3). In c-Met kinase, both the quinoline core and the amide group participate in hydrogen bonding with residues in the ATP-binding pocket (Figure 4). Based on these observations and the binding mode of 18c [31], we propose modifying the 2-position of the pyrimidine ring to introduce hydrophilic groups. This strategy aims to enhance solubility and bioavailability while preserving dual-target activity.
Building upon our previous work (Figure 5), we designed and synthesized a series of 2-substituted aniline pyrimidine derivatives, with a subsequent evaluation of their biological activities. Among these, compound 17c emerged as a lead candidate due to its superior metabolic stability in human liver microsomes and potent antiproliferative activities against the HepG2 (liver), MDA-MB-231 (breast), and HCT116 (colon) cancer cell lines. 17c also revealed favorable pharmacokinetic properties, plasma protein binding, and a favorable safety profile in hERG assays. Mechanistic studies further showed that 17c induced dose-dependent apoptosis in HCT116 cells and effectively inhibited HCT116 cell migration. These results position 17c as a promising dual inhibitor of Mer and c-Met with significant development potential.

2. Materials and Methods

2.1. Chemical Part

All reagents and solvents were sourced from Titan Chemical Co., Ltd. (Shanghai, China), and Shijiazhuang Kehai Huabo Instruments Co., Ltd. (Shijiazhuang, China), respectively, and used without further purification unless specified. The reaction process was monitored by TLC analysis on silica GF254 Plates. High-resolution mass spectra (HRMS) data were acquired in ESI mode using a Water Q-Tof mass spectrometer (Waters Xevo G2-XS QTof). 1H- and 13C-NMR spectra were recorded on Bruker AM-400/500 spectrometers (Bruker Bioscience, USA) with DMSO-d6 used as solvent and tetramethylsilane (TMS) used as the internal reference, and chemical shift values were reported in ppm. The multiplicity of the signals is denoted as follows: s (singlet); d (doublet), t (triplet), q (quadruplet), qui (quintuplet), m (multiplet), dd (doublet of doublets), and dt (doublet of triplets). Coupling constants (J) are given in hertz (Hz). Flash chromatography was performed using silica gel (200–300 mesh) as the stationary phase, with a mobile phase consisting of a mixture of methanol (MeOH), ethyl acetate (EA), and petroleum ether (PE). The purity of all synthesized compounds was confirmed to be >95% by high-performance liquid chromatography (HPLC) using a Waters HPLC system equipped with a UV/visible detector (Waters Corporation, MA, USA), monitored at 254 nm. HPLC analysis was carried out on a 5 μm, 4.6 × 250 mm Hypersil ODS2 column, with a mobile phase consisting of a 3:7 (v/v) mixture of potassium dihydrogen phosphate solution and methanol, at a flow rate of 1.0 mL/min and an injection volume of 10 μL. All chemicals were of analytical grade and used without further purification.

2.1.1. Preparation of N-(4-Fluorophenyl)-N-(4-hydroxyphenyl)cyclopropane-1,1-dicarboxamide (Intermediate 11)

Intermediate 11 was prepared as a white solid in an 80.6% yield according to our previous work [31]. Consistent with prior methodology, the crude product required no purification and was employed directly in subsequent steps.

2.1.2. Preparation of N-(4-((2-Chloropyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (Intermediate 12)

Intermediate 12 was prepared as a white solid in a 75.7% yield according to our previous work [31].

2.1.3. General Procedure for Preparation of Target Compounds 13a13n

Target compounds 13a13n were prepared according to our previous work [31].
N-(4-fluorophenyl)-N-(4-((2-((4-((1-methylpiperidin-4-yl)oxy)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13a): White solid, yield: 23.27%. 1H NMR (500 MHz, DMSO-d6) δ 10.18 (s, 1H), 10.11 (s, 1H), 9.41 (s, 1H), 8.30 (d, J = 5.6 Hz, 1H), 7.70 (d, J = 8.9 Hz, 2H), 7.66 (dd, J = 9.1, 5.1 Hz, 2H), 7.43 (d, J = 7.5 Hz, 2H), 7.19–7.14 (m, 4H), 6.80 (d, J = 8.7 Hz, 2H), 6.35 (d, J = 5.6 Hz, 1H), 4.44 (s, 1H), 3.16–2.87 (m, 4H), 2.63 (s, 3H), 2.10–2.01 (m, 2H), 1.85 (s, 2H), 1.51 (d, J = 4.6 Hz, 4H). 13C NMR (126 MHz, DMSO-d6) δ 170.06, 168.73 (d, J = 4.3 Hz, 1C), 160.19, 159.70, 157.79, 151.65, 148.59, 136.63, 135.66, 134.42, 122.86 (d, J = 7.7 Hz, 1C), 122.43 (d, J = 10.0 Hz, 1C), 120.91,116.61, 115.62, 115.44, 98.07, 78.48, 50.54, 43.37, 32.01, 28.45, 15.91. HRMS: m/z C33H33FN6O4 [M + H]+ 597.2547, Found 597.2628. Purity: >95% (HPLC).
N-(4-fluorophenyl)-N-(4-((2-((3-((2-methoxyethyl)amino)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13b): White solid, yield: 20.08%. 1H NMR (500 MHz, DMSO-d6) δ 10.15 (s, 1H), 10.08 (s, 1H), 9.27 (s, 1H), 8.31 (d, J = 5.6 Hz, 1H), 7.71 (d, J = 9.0 Hz, 2H), 7.66 (dd, J = 9.0, 5.1 Hz, 2H), 7.21–7.14 (m, 4H), 6.86–6.79 (m, 3H), 6.34 (d, J = 5.6 Hz, 1H), 6.16 (d, J = 7.3 Hz, 1H), 5.21 (s, 1H), 3.44 (t, J = 5.9 Hz, 2H), 3.26 (s, 3H), 3.06 (t, J = 5.8 Hz, 2H), 1.49 (s, 4H). 13C NMR (126 MHz, DMSO-d6) δ 169.90, 168.67, 160.37, 160.22, 159.70, 157.79, 148.56, 141.27, 136.65, 135.67 (d, J = 2.0 Hz, 1C), 129.15, 122.82 (d, J = 7.7 Hz, 1C), 122.27, 115.61, 115.43, 107.93, 106.71, 103.23, 98.27, 70.93, 58.45, 42.93, 31.95, 15.86. HRMS: m/z C30H29FN6O4 [M + H]+ 557.2234, Found 557.2312. Purity: >95% (HPLC).
N-(4-((2-((1H-pyrazol-4-yl)amino)pyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (compound 13c): White solid, yield: 31.52%. 1H NMR (500 MHz, DMSO-d6) δ 12.31 (s, 1H), 10.17 (s, 1H), 10.08 (s, 1H), 9.65–9.18 (m, 1H), 8.29 (s, 1H), 7.72 (s, 2H), 7.65 (dd, J = 8.7, 5.0 Hz, 3H), 7.30–7.11 (m, 5H), 6.30 (d, J = 5.5 Hz, 1H), 1.50 (s, 4H). 13C NMR (126 MHz, DMSO-d6) δ 170.46, 168.68 (d, J = 11.7 Hz, 1C), 160.42, 159.84, 159.71, 157.80, 148.56, 136.67, 135.63, 130.12, 122.87 (d, J = 6.0 Hz, 1C), 122.40, 122.13, 115.62, 115.44, 98.04, 31.89, 15.97. HRMS: m/z C24H20FN7O3 [M + H]+ 474.1612, Found 474.1686. Purity: >95% (HPLC).
N-(4-((2-((3,5-difluorophenyl)amino)pyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (compound 13d): White solid, yield: 19.40%. 1H NMR (400 MHz, DMSO-d6) δ 10.23 (s, 1H), 10.07 (s, 1H), 10.01 (s, 1H), 8.47 (d, J = 5.6 Hz, 1H), 7.78 (d, J = 8.8 Hz, 2H), 7.69 (dd, J = 8.7, 5.1 Hz, 2H), 7.34 (d, J = 9.4 Hz, 2H), 7.28–7.17 (m, 4H), 6.67 (t, J = 9.1 Hz, 1H), 6.59 (d, J = 5.6 Hz, 1H), 1.53 (d, J = 4.1 Hz, 4H). 13C NMR (101 MHz, DMSO-d6) δ 170.09, 168.56, 160.40, 159.61, 157.59, 148.25, 143.31, 137.04, 135.59 (d, J = 2.6 Hz, 1C), 122.93 (d, J = 7.8 Hz, 1C), 122.29, 122.05, 115.62, 115.40, 101.74, 101.44, 99.97, 31.88, 15.93. HRMS: m/z C27H20F3N5O3 [M + H]+ 520.1518, Found 520.1594. Purity: >95% (HPLC).
N-(4-((2-((3-(4-acetylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (compound 13e): White solid, yield: 23.07%. 1H NMR (400 MHz, DMSO-d6) δ 10.15 (s, 1H), 10.07 (s, 1H), 9.38 (s, 1H), 8.34 (d, J = 5.6 Hz, 1H), 7.70 (d, J = 9.0 Hz, 2H), 7.64 (dd, J = 9.1, 5.1 Hz, 2H), 7.20–7.13 (m, 5H), 7.08 (d, J = 8.1 Hz, 1H), 6.97 (t, J = 8.1 Hz, 1H), 6.47 (dd, J = 8.1, 1.7 Hz, 1H), 6.39 (d, J = 5.6 Hz, 1H), 3.56–3.47 (dd, J = 10.5, 7.0 Hz, 4H), 2.98 (t, J = 4.7 Hz, 2H), 2.93 (t, J = 4.9 Hz, 2H), 2.03 (s, 3H), 1.47 (s, 4H). 13C NMR (101 MHz, DMSO-d6) δ 169.82, 168.71 (d, J = 4.3 Hz, 1C), 168.60, 160.30 (d, J = 7.60 Hz, 1C), 159.95, 157.56, 151.52, 148.56, 141.30, 136.59, 135.66 (d, J = 2.5 Hz, 1C), 129.26, 122.85 (d, J = 7.9 Hz, 1C), 122.10, 115.62, 115.40, 111.05, 110.13, 107.05, 98.72, 49.23, 45.87, 31.97, 21.64, 15.85. HRMS: m/z C33H32FN7O4 [M + H]+ 610.2500, Found 610.2576. Purity: >95% (HPLC).
N-(4-fluorophenyl)-N-(4-((2-((3-(piperidin-3-ylcarbamoyl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13f): White solid, yield: 31.00%. 1H NMR (500 MHz, DMSO-d6) δ 10.11 (s, 1H), 10.07 (s, 1H), 8.23 (d, J = 5.6 Hz, 1H), 8.10 (d, J = 8.0 Hz, 1H), 7.67 (d, J = 8.9 Hz, 2H), 7.64 (dd, J = 8.9, 5.1 Hz, 2H), 7.19–7.12 (m, 4H), 7.07 (t, J = 7.8 Hz, 1H), 7.02 (s, 1H), 6.94 (d, J = 7.7 Hz, 1H), 6.69 (dd, J = 7.9, 1.5 Hz, 1H), 6.07 (d, J = 5.4 Hz, 1H), 5.27 (s, 1H), 4.59–4.20 (m, 2H), 3.83–3.74 (m, 1H), 2.88–2.76 (m, 2H), 1.93–1.85 (m, 1H), 1.75–1.68 (m, 1H), 1.64–1.54 (m, 1H), 1.46 (s, 4H), 1.44–1.34 (m, 2H). 13C NMR (126 MHz, DMSO-d6) δ 169.81, 168.62 (d, J = 11.21 Hz, 1C), 167.20, 161.64, 160.43, 159.70, 157.79, 148.95, 148.31, 136.40, 136.11, 135.63 (d, J = 1.61 Hz, 1C), 129.02, 122.88 (d, J = 7.70 Hz, 1C), 121.96, 116.86, 115.58, 115.41, 115.05, 113.40, 95.84, 48.53, 46.15, 43.81, 31.91, 30.76, 23.97, 15.91. HRMS: m/z C24H20FN7O3 [M + H]+ 610.2500, Found 610.2576. Purity: >95% (HPLC).
N-(4-fluorophenyl)-N-(4-((2-((4-(piperidin-3-ylcarbamoyl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13g): White solid, yield: 28.15%. 1H NMR (500 MHz, DMSO-d6) δ 10.10 (s, 1H), 10.08 (s, 1H), 8.22 (d, J = 5.5 Hz, 1H), 7.81 (d, J = 7.9 Hz, 1H), 7.69–7.61 (m, 4H), 7.58 (d, J = 8.4 Hz, 2H), 7.15 (t, J = 8.2 Hz, 4H), 6.54 (d, J = 8.4 Hz, 2H), 6.06 (d, J = 4.9 Hz, 1H), 5.64 (s, 1H), 4.62–4.15 (m, 2H), 3.83–3.72 (m, 1H), 2.82 (t, J = 11.5 Hz, 2H), 1.88 (d, J = 9.6 Hz, 1H), 1.71 (d, J = 11.6 Hz, 1H), 1.61–1.51 (m, 1H), 1.46 (s, 4H), 1.44–1.33 (m, 2H). 13C NMR (126 MHz, DMSO-d6) δ 169.81, 168.62 (d, J = 9.89 Hz, 1C), 166.27, 161.62, 160.39, 159.70, 157.79, 152.00, 148.32, 136.37, 135.62 (d, J = 1.39 Hz, 1C), 129.33, 122.88 (d, J = 7.70 Hz, 1C), 122.04, 121.94, 115.59, 115.41, 112.95, 95.78, 48.76, 45.99, 43.83, 31.88, 30.93, 24.04, 15.92. HRMS: m/z C24H20FN7O3 [M + H]+ 610.2500, Found 610.2567. Purity: >95% (HPLC).
N-(4-fluorophenyl)-N-(4-((2-((4-(4-methylpiperazine-1-carbonyl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13h): White solid, yield: 18.28%. 1H NMR (400 MHz, DMSO-d6) δ 10.18 (s, 1H), 10.09 (s, 1H), 9.80 (s, 1H), 8.38 (d, J = 5.6 Hz, 1H), 7.71 (d, J = 9.0 Hz, 2H), 7.65 (dd, J = 9.1, 5.1 Hz, 2H), 7.58 (d, J = 8.5 Hz, 2H), 7.21 (d, J = 9.0 Hz, 2H), 7.19–7.13 (m, 4H), 6.48 (d, J = 5.6 Hz, 1H), 3.46 (s, 4H), 2.32 (s, 4H), 2.21 (s, 3H), 1.50 (s, 4H). 13C NMR (101 MHz, DMSO-d6) δ 170.11, 169.51, 168.71, 160.41, 159.90, 157.56, 148.52, 141.96, 136.78, 135.62 (d, J = 2.5 Hz, 1C), 128.19, 122.89 (d, J = 7.9 Hz, 1C), 122.51, 122.33, 118.39, 115.62, 115.40, 99.15, 54.92, 45.97, 31.81, 31.62, 16.07. HRMS: m/z C33H32FN7O4 [M + H]+ 610.2500, Found 610.2568. Purity: >95% (HPLC).
N-(4-((2-((4-(2-((2-(diethylamino)ethyl)amino)acetamido)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (compound 13i): White solid, yield: 16.50%. 1H NMR (400 MHz, DMSO-d6) δ 10.15 (s, 1H), 10.09 (s, 1H), 9.86 (s, 1H), 9.48 (s, 1H), 8.32 (d, J = 5.6 Hz, 1H), 7.71 (d, J = 8.9 Hz, 2H), 7.65 (dd, J = 9.0, 5.1 Hz, 2H), 7.51 (d, J = 8.7 Hz, 2H), 7.40 (d, J = 8.8 Hz, 2H), 7.20–7.13 (m, 4H), 6.36 (d, J = 5.6 Hz, 1H), 3.36 (s, 3H), 2.90 (s, 2H), 2.75 (s, 6H), 1.51 (d, J = 5.2 Hz, 4H), 1.06 (t, J = 7.1 Hz, 6H). 13C NMR (101 MHz, DMSO-d6) δ 170.02, 169.51, 168.74 (d, J = 2.1 Hz, 1C), 160.17, 159.94, 157.55, 148.51, 136.72, 136.35, 135.64 (d, J = 2.6 Hz, 1C), 133.07, 122.86 (d, J = 7.8 Hz, 1C), 122.25 (d, J = 12.7 Hz, 1C), 120.10, 119.64, 115.62, 115.40, 98.40, 52.32, 51.46, 46.90, 45.95, 31.92, 15.99, 10.73. HRMS: m/z C35H39FN8O4 [M + H]+ 655.3078, Found 655.3160. Purity: >95% (HPLC).
N-(4-((2-((4-(2-((2-(dimethylamino)ethyl)amino)acetamido)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (compound 13j): White solid, yield: 30.11%. 1H NMR (400 MHz, DMSO-d6) δ 10.16 (s, 1H), 10.12 (s, 1H), 9.91 (s, 1H), 9.48 (s, 1H), 8.32 (s, 1H), 7.71 (s, 2H), 7.65 (s, 2H), 7.49 (s, 2H), 7.42 (s, 2H), 7.18 (s, 4H), 6.36 (s, 1H), 3.32 (s, 3H), 2.71 (s, 2H), 2.55 (s, 2H), 2.32 (s, 6H), 1.51 (s, 4H). 13C NMR (101 MHz, DMSO-d6) δ 170.03, 169.76, 168.76 (d, J = 3.0 Hz, 1C), 160.18, 157.57, 148.54, 136.70, 136.30, 135.61 (d, J = 2.6 Hz, 1C) 133.12, 122.89 (d, J = 7.9 Hz, 1C), 122.32, 122.19, 120.01, 119.65, 115.62, 115.40, 98.39, 58.06, 52.53, 46.03, 44.87, 31.88, 16.02. HRMS: m/z C33H35FN8O4 [M + H]+ 627.2765, Found 627.2841. Purity: >95% (HPLC).
N-(4-fluorophenyl)-N-(4-((2-((4-(2-(4-(2-methoxyethyl)piperazin-1-yl)acetamido)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)cyclopropane-1,1-dicarboxamide (compound 13k): White solid, yield: 33.25%. 1H NMR (400 MHz, DMSO-d6) δ 10.14 (s, 1H), 10.09 (s, 1H), 9.48 (s, 1H), 9.46 (s, 1H), 8.32 (d, J = 5.6 Hz, 1H), 7.71 (d, J = 8.8 Hz, 2H), 7.64 (dd, J = 8.9, 5.1 Hz, 2H), 7.47 (d, J = 8.6 Hz, 2H), 7.37 (d, J = 8.8 Hz, 2H), 7.16 (q, J = 8.6 Hz, 4H), 6.36 (d, J = 5.6 Hz, 1H), 3.42 (t, J = 5.8 Hz, 2H), 3.22 (s, 3H), 3.04 (s, 2H), 2.50 (s, 2H), 2.47 (s, 8H), 1.51 (d, J = 8.2 Hz, 4H). 13C NMR (101 MHz, DMSO-d6) δ 170.05, 168.72, 168.18, 160.15, 157.56, 148.54, 136.71, 136.39, 135.63 (d, J = 2.5 Hz, 1C), 132.92, 122.87 (d, J = 7.9 Hz, 1C), 122.35, 122.17, 120.33, 119.54, 115.62, 115.40, 98.36, 70.34, 62.22, 58.46, 57.46, 53.42, 53.27, 31.89, 16.01. HRMS: m/z C36H39FN8O5 [M + H]+ 683.3027, Found 683.3107. Purity: >95% (HPLC).

2.1.4. Preparation of N-(4-Hydroxyphenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (Intermediate 15)

A mixture of 4-antipyrine carboxylic acid (14, 2.0 g, 8.61 mmol) and 4-aminophenol (10, 1.13 g, 10.33 mmol) in DMF (20 mL) was treated with HBTU (3.92 g, 10.33 mmol) and TEA (2.61 g, 25.84 mmol). After stirring at ambient temperature for 8 h (reaction monitored by TLC), the mixture was quenched by pouring into ice water (200 mL). The resulting precipitate was collected by filtration, washed thoroughly, and vacuum-dried to afford intermediate 15 as a white solid (1.75 g, 63.0%). This product was used directly in subsequent without purification.

2.1.5. Preparation of N-(4-((2-Chloropyrimidin-4-yl)oxy)phenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (Intermediate 16)

A mixture of intermediate 15 (1.75 g, 5.41 mmol) and 2,4-dichloropyrimidine (0.81 g, 5.41 mmol) in DMF (15 mL) was treated with K2CO3 (0.82 g, 5.95 mmol). The mixture was stirred at 80 °C for 4.5 h. The reaction solution was poured into ice water (100 mL), and the precipitate was filtered off, washed, and dried in a vacuum to yield intermediate 16 as a white solid (1.85 g, 78.0%), which can be used directly without any further purification.

2.1.6. General Procedure for Preparation of Target Compounds 17a17h

Intermediate 16 (1.2 mmol) and substituted aniline (1.0 mmol) were dissolved in DMF (8 mL), followed by the addition of PTSA (4.00 mmol). The reaction mixture was stirred at 90 °C for 4 h under N2 atmosphere. After cooling to ambient temperature, the solution was quenched with ice water (100 mL). The precipitate solid was isolated by filtration, washed, and vacuum-dried. Crude product purification via silica gel chromatography (gradient elution: DCM/MeOH 100:1–30:1) afforded compounds 17a17h.
1,5-dimethyl-N-(4-((2-((4-((1-methylpiperidin-4-yl)oxy)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17a): White solid, yield: 15.05%. 1H NMR (500 MHz, DMSO-d6) δ 10.82 (s, 1H), 9.41 (s, 1H), 8.29 (d, J = 5.6 Hz, 1H), 7.68 (d, J = 8.8 Hz, 2H), 7.60 (t, J = 7.7 Hz, 2H), 7.53 (d, J = 7.3 Hz, 1H), 7.45 (d, J = 7.5 Hz, 2H), 7.40–7.34 (m, 2H), 7.18 (d, J = 8.8 Hz, 2H), 6.71 (d, J = 7.7 Hz, 2H), 6.38 (d, J = 5.6 Hz, 1H), 4.25 (s, 1H), 3.38 (s, 3H), 2.77 (s, 2H), 2.74 (s, 3H), 2.45 (s, 2H), 2.31 (d, J = 13.7 Hz, 3H), 1.90 (s, 2H), 1.68 (s, 2H).13C NMR (126 MHz, DMSO-d6) δ 170.12, 163.52, 161.67, 160.36, 160.15, 154.26, 151.89, 148.17, 136.67, 134.09, 133.49, 129.97, 129.33, 127.59, 123.09, 120.95, 120.76, 116.33, 97.94, 97.60, 44.97, 33.83, 21.25, 11.98. HRMS: m/z C34H36N7O4 [M + H]+ 606.2751, Found 606.2832. Purity: >95% (HPLC).
N-(4-((2-((3-((2-methoxyethyl)amino)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17b): White solid, yield: 5.80%. 1H NMR (500 MHz, DMSO-d6) δ 10.82 (s, 1H), 9.28 (s, 1H), 8.31 (d, J = 5.4 Hz, 1H), 7.69 (d, J = 8.5 Hz, 2H), 7.60 (t, J = 7.4 Hz, 3H), 7.52 (t, J = 7.2 Hz, 1H), 7.45 (d, J = 7.4 Hz, 2H), 7.19 (d, J = 8.6 Hz, 2H), 6.87–6.77 (m, 3H), 6.36 (d, J = 5.4 Hz, 1H), 6.16 (d, J = 6.8 Hz, 1H), 3.42 (t, J = 5.5 Hz, 2H), 3.37 (s, 3H), 3.24 (s, 3H), 3.05 (s, 2H), 2.72 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 169.92, 163.53, 161.65, 160.35, 160.23, 154.29, 149.19, 148.09, 141.27, 136.65, 133.52, 129.95, 129.32, 129.04, 127.58, 122.77, 120.61, 107.94, 106.65, 103.26, 98.29, 97.57, 70.93, 58.42, 42.94, 33.79, 11.93. HRMS: m/z C31H21N7O4Na [M + Na]+ 588.2438, Found 588.2337. Purity: >95% (HPLC).
1,5-dimethyl-N-(4-((2-((3-((1-methylpiperidin-4-yl)oxy)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17c): White solid, yield: 7.01%. 1H NMR (500 MHz, DMSO-d6) δ 10.81 (s, 1H), 9.51 (s, 1H), 8.35 (d, J = 5.5 Hz, 1H), 7.68 (d, J = 8.7 Hz, 2H), 7.60 (t, J = 7.5 Hz, 2H), 7.52 (t, J = 7.4 Hz, 1H), 7.45 (d, J = 7.5 Hz, 2H), 7.27 (s, 1H), 7.20 (d, J = 8.7 Hz, 2H), 7.16 (d, J = 7.8 Hz, 1H), 7.00 (t, J = 8.0 Hz, 1H), 6.51 (d, J = 7.7 Hz, 1H), 6.42 (d, J = 5.5 Hz, 1H), 4.30 (s, 1H), 3.34 (s, 2H), 2.93 (s, 2H), 2.72 (s, 3H), 2.45 (s, 3H), 2.00 (s, 2H), 1.76 (s, 2H).13C NMR (126 MHz, DMSO-d6) δ 169.95, 163.51, 161.64, 160.33, 160.13, 157.35, 154.28, 147.98, 141.90, 136.69, 133.50, 129.96, 129.49, 129.33, 127.59, 122.70, 120.68, 112.14, 108.92, 107.25, 98.85, 97.57, 51.75, 44.37, 33.80, 31.62, 30.30, 11.94. HRMS: m/z C34H36N7O4 [M + H]+ 606.2751, Found 606.2823. Purity: >95% (HPLC).
1,5-dimethyl-N-(4-((2-((4-(4-methylpiperazine-1-carbonyl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17d): White solid, yield: 63.0%. 1H NMR (500 MHz, DMSO-d6) δ 10.85 (s, 1H), 9.82 (s, 1H), 8.38 (d, J = 5.6 Hz, 1H), 7.71 (d, J = 8.8 Hz, 2H), 7.60 (t, J = 7.6 Hz, 2H), 7.49–7.56 (m, 3H), 7.45 (d, J = 7.3 Hz, 2H), 7.21 (d, J = 8.8 Hz, 2H), 7.13 (d, J = 8.3 Hz, 2H), 6.51 (d, J = 5.6 Hz, 1H), 3.37 (s, 4H), 3.33 (s, 3H), 2.73 (s, 3H), 2.27 (s, 4H), 2.15 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 170.17, 169.40, 163.51, 161.67, 160.46, 159.84, 154.35, 148.05, 141.99, 136.85, 133.49, 129.95, 129.30, 128.45, 128.17, 127.57, 123.12, 120.68, 118.37, 99.11, 97.65, 54.90, 33.80, 11.98. HRMS: m/z C34H34N8O4Na [M + Na]+ 641.2703, Found 641.2603. Purity: >95% (HPLC).
N-(4-((2-((4-(4-acetylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17e): White solid, yield: 15.79%. 1H NMR (500 MHz, DMSO-d6) δ 10.85 (s, 1H), 9.36 (s, 1H), 8.28 (d, J = 5.5 Hz, 1H), 7.69 (d, J = 8.7 Hz, 2H), 7.60 (t, J = 7.6 Hz, 2H), 7.52 (t, J = 7.4 Hz, 1H), 7.47 (d, J = 7.4 Hz, 2H), 7.26 (s, 2H), 7.16 (d, J = 8.8 Hz, 2H), 6.68 (d, J = 6.9 Hz, 2H), 6.38 (d, J = 5.5 Hz, 1H), 3.48 (s, 4H), 3.40 (s, 3H), 2.99 (s, 2H), 2.87 (s, 2H), 2.74 (s, 3H), 2.00 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 170.26, 168.66, 163.48, 161.69, 160.29, 160.03, 154.04, 148.29, 145.98, 136.72, 133.47, 133.25, 129.97, 129.38, 127.69, 123.28, 120.82, 120.48, 116.55, 97.44, 50.18, 49.24, 45.98, 33.75, 21.60, 11.88. HRMS: m/z C34H34N8O4Na [M + Na]+ 641.2703, Found 641.2607. Purity: >95% (HPLC).
N-(4-((2-((3-(4-acetylpiperazin-1-yl)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17f): White solid, yield: 20.0%. 1H NMR (500 MHz, DMSO-d6) δ 10.82 (s, 1H), 9.40 (s, 1H), 8.34 (d, J = 5.5 Hz, 1H), 7.68 (d, J = 8.8 Hz, 2H), 7.60 (t, J = 7.6 Hz, 2H), 7.52 (t, J = 7.4 Hz, 1H), 7.45 (d, J = 7.4 Hz, 2H), 7.19 (d, J = 8.5 Hz, 3H), 7.09 (d, J = 7.3 Hz, 1H), 6.96 (t, J = 8.0 Hz, 1H), 6.51 (d, J = 7.5 Hz, 1H), 6.41 (d, J = 5.5 Hz, 1H), 3.51 (t, J = 5.7 Hz, 4H), 3.37 (s, 4H), 2.96 (s, 4H), 2.72 (s, 3H), 2.01 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 169.84, 168.70, 163.53, 161.64, 160.33, 160.26, 154.27, 151.57, 148.05, 141.31, 136.61, 133.51, 129.95, 129.32, 129.14, 127.60, 122.59, 120.57, 111.11, 110.15, 107.18, 98.73, 97.56, 49.29, 48.88, 45.86, 33.78, 21.63, 11.92. HRMS: m/z C34H34N8O4Na [M + Na]+ 641.2703, Found 641.2599. Purity: >95% (HPLC).
1,5-dimethyl-N-(4-((2-((6-(2-(4-methylpiperazin-1-yl)acetamido)pyridin-3-yl)amino)pyrimidin-4-yl)oxy)phenyl)-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17g): White solid, yield: 18.6%. 1H NMR (600 MHz, DMSO-d6) δ 10.81 (s, 1H), 9.72 (s, 1H), 9.67 (s, 1H), 8.49 (s, 1H), 8.35 (d, J = 5.6 Hz, 1H), 7.98 (s, 1H), 7.88 (d, J = 8.6 Hz, 1H), 7.69 (d, J = 8.9 Hz, 2H), 7.60 (t, J = 7.7 Hz, 2H), 7.52 (t, J = 7.5 Hz, 1H), 7.48–7.43 (m, 2H), 7.20 (d, J = 8.9 Hz, 2H), 6.43 (d, J = 5.6 Hz, 1H), 3.37 (s, 5H), 3.11 (s, 2H), 2.72 (s, 3H), 2.52–2.50 (m, 2H), 2.38 (s, 4H), 2.18 (s, 3H).13C NMR (126 MHz, DMSO-d6) δ 170.10, 168.61, 163.52, 161.66, 161.66, 160.40, 160.08, 154.31, 147.90, 145.69, 139.23, 136.78, 133.75, 133.50, 129.95, 129.32, 128.77, 127.57, 122.68, 120.68, 113.01, 99.15, 97.62, 61.50, 55.13, 52.97, 46.02, 33.78, 11.94. HRMS: m/z C34H36N10O4Na [M + Na]+ 671.2921, Found 671.2823. Purity: >95% (HPLC).
1,5-dimethyl-N-(4-((2-((4-(2-(4-methylpiperazin-1-yl)propanamido)phenyl)amino)pyrimidin-4-yl)oxy)phenyl)-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (compound 17h): White solid, yield: 33.8%. 1H NMR (600 MHz, DMSO-d6) δ 10.81 (s, 1H), 9.55 (s, 1H), 9.49 (s, 1H), 8.32 (d, J = 5.6 Hz, 1H), 7.71–7.67 (m, 2H), 7.60 (t, J = 7.7 Hz, 2H), 7.52 (dt, J = 8.3, 1.5 Hz, 1H), 7.51–7.47 (m, 2H), 7.45–7.42 (m, 2H), 7.37 (d, J = 8.6 Hz, 2H), 7.22–7.18 (m, 2H), 6.37 (d, J = 5.6 Hz, 1H), 3.37 (s, 3H), 3.15 (q, J = 6.8 Hz, 1H), 2.72 (s, 3H), 2.52–2.50 (m, 2H), 2.40 (d, J = 71.7 Hz, 6H), 2.13 (s, 3H), 1.13 (d, J = 6.9 Hz, 3H).13C NMR (126 MHz, DMSO-d6) δ 171.20, 170.01, 163.52, 161.65, 160.31, 160.17, 154.32, 148.03, 136.71, 133.49, 133.13, 129.95, 129.31, 127.53, 122.77, 120.64, 120.16, 119.56, 98.41, 97.65, 63.77, 55.38, 49.39, 46.16, 33.78, 13.29, 11.94. HRMS: m/z C36H40N9O4 [M + H]+ 662.3125, Found 662.3207. Purity: >95% (HPLC).

2.2. Inhibitory Activity Assessment Against Mer and c-Met

In vitro Mer/c-Met kinase inhibition profiling was outsourced to Shanghai Bioduro Biological Technology Co., Ltd. (Shanghai, China) using the following optimized protocol: Assay buffer (1X, MgCl2 5 mM, DTT 1 mM) served as the base matrix. Test compounds were diluted in DMSO to 100× final concentration and dispensed (100 nL/well) into a 384-well plate via an automated liquid handler, maintaining 1% DMSO% throughout. Enzymes were diluted to a 2X assay working concentration (1.2 nM) in buffer. Then, 5 μL was added to the assay plate manually (final 1 nM) with a multichannel pipette, span down at 1000 rpm and centrifuged for 30 s, and incubated for 15 min at 25 °C temperatures. The substrate solutions were diluted with 1X assay buffer. Then, 5 μL of mix or buffer was added to the assay plate manually (ATP final 3.5 μM and TK-Sub-Biotin final 0.5 μM) with a multichannel pipette, span down at 1000 rpm and centrifuged for 30 s. After 30 °C 60 min, 10 μL of detection solution (TK-antidody-cryptate final 0.25X and streptavidine-XL 665 final 0.3125 μM) was added to each well of the assay. This was mixed briefly with centrifuge and equilibrated for 60 min. The luminescence was recorded on Envision.

2.3. Antiproliferation Assay

The HepG2, MDA-MB-231, and HCT116 cell lines were derived from our own laboratory (the laboratory of our collaborator, Dr. Xiaolei Zhou, at College of Food Science and Biology in Hebei University of Science and Technology) [31]. The antiproliferative activities were assessed using the CCK-8 assay. Cell viability was measured according to the manufacturer’s instructions for the Cell Counting Kit-8 (Meilunbio, Dalian, China). In this assay, 2.5 × 103 cells per well were seeded into 96-well plates.
After an initial 12 h incubation, cancer cells were exposed to varying concentrations of compound 17c for 72 h. Following CCK-8 solution addition and 2 h incubation, the absorbance at 450 nm was measured using a BioTek Synergy HTX microplate reader. Experiments included triplicate wells with three independent replicates. The IC50 values for compound 17c were derived from an analysis of the dose–response curves in GraphPad PRISM 9.5.

2.4. hERG Potassium Channel Activity Assessment

hERG channel activity was evaluated in CHO cells (Cell Bank of Chinese Academy of Sciences, SCSP-507) stably expressing hERG transcripts using Sophion’s QPatch automated patch clamp system (Shanghai Institute of Materia Medica, Shanghai, China). Cells cultured in serum-supplemented F12 medium (37 °C/5% CO2) to 70–80% confluency were PBS-washed, detached with Detachin (37 °C/2 min), and resuspended in growth medium (2–5 × 106 cells/mL) before QPatch loading. Following centrifugation and washing, recording was performed using extracellular solution (140 NaCl, 5 KCl, 1 CaCl2, 1.25 MgCl2, 10 HEPES and 10 Glucose, pH 7.4 with NaOH) and intracellular solution (140 KCl, 1 MgCl2, 1 CaCl2, 10 EGTA and 10 HEPES, pH 7.2 KOH). A voltage protocol measured tail current peaks at −50 mV. After a 5 min baseline recording, compounds were applied for 2.5 min/concentration across six escalating doses (n ≥ 3 cells/dose), with terminal cisapride reference. The IC50 values were derived from concentration–inhibition curves (four-parameter logistic fit, GraphPad Prism 8) using Assay Software v5.6.4 for acquisition and Excel for supplementary analysis.

2.5. Liver Microsome Stability Assay

Each test compound was initially dissolved in DMSO to prepare a 10 mM stock solution. Test compound and positive controls were pre-diluted in acetonitrile to 200 μM. Incubation mixtures (200 μL total volume) comprised 0.1 M PBS (pH 7.4), 2 mM of NADPH, 0.2 mg/mL liver microsomes, and the diluted compound/control. Following a 5 min pre-incubation of all components (excluding NADPH) at 37 °C, reactions were initiated withNADPH addition. The mixture was pipette-mixed thoroughly, and 20 μL was immediately transferred to a “Quenching” plate as a 0 min sample, followed by pipette-mixing. At subsequent time points (5, 15, 30, and 60 min), 20 μL aliquots were transferred from the incubate to the “Quenching” plate after pipette-mixing. To each well of the plate, 200 μL of acetonitrile containing the internal standard was added. After centrifugation (4000 rpm, 10 min), 50 μL of supernatant was diluted 1:1 with deionized water and subjected to LC-MS/MS analysis.

2.6. Docking Study

Molecular docking simulations were performed in our lab. The binding interactions of the compounds were investigated with the aid of Discovery Studio 2019. For this analysis, the X-ray crystallographic structures of MerTK (PDB: 4M3Q) and c-Met (PDB: 3LQ8), both resolved at 1.84 Å resolution, were utilized. The protein–ligand interactions were visualized and analyzed through the interaction mode diagram generated by Discovery Studio 2019.

2.7. Western Blot Assay

Western blot assay was performed according to established protocols. HCT colon cancer cells were exposed to the specified concentration of compound 17c for 24 h, after which whole cell lysates were prepared using RAPI cell lysis buffer (Beyotime Institute of Biotechnology, Shanghai, China). Primary antibodies, purchased from Beyotime Institute of Biotechnology, were diluted at a ratio of 1:2000. The protein bands on polyvinylidene fluoride membranes were visualized and analyzed using Glyco Band-Scan Software Version 4.50. The antibody details are as follows:
Name: Phospho-MER/TYRO3 (Tyr753/Tyr685) antibody, Species: Rabit, Sequence: synthesized peptide derived from human MER/TYRO3 around the Phosphorylation site of Tyr753/Tyr685, Dilution: 1:2000, RRID number: AB_2840500, Catalogue number: AF8443, Supplier: Affinity Biosciences (Beijing, China).
Name: Phospho-c-Met (Tyr1234) antibody, Species: Rabit, Sequence: synthesized peptide derived from human c-Met around the Phosphorylation site of Tyr1234, Dilution: 1:2000, RRID number: AB_2834564, Catalogue number: AF3129, Supplier: Affinity Biosciences (Beijing, China).

2.8. Apotosis Study

Apoptosis was analyzed in a dose-dependent manner using the One Step TUNEL Apoptosis Assay Kit (Meilunbio, Dalian, China). HCT116 cells (1.5 × 104 cells/well) were seeded onto TC-treated glass coverslips in 24-well plates and incubated for 24 h. Following this, the cells were exposed to varying concentrations of compound 17c for 48 h. After treatment, the cells were fixed with 4% paraformaldehyde and permeabilized with 0.3% Triton-X-100. TUNEL staining was performed as per the manufacturer’s guidelines, with DAPI used for nuclear counterstaining. Fluorescent images were obtained using an Olympus microscope. The ED50 value for compound 17c was calculated using GraphPad PRISM 9.5. Each assay was performed in triplicate and repeated three times for consistency.

2.9. Transwell Assay

Migration assays were conducted using Transwell® inserts (Costar, Cambridge, MA, USA) with an 8 μm pore size polycarbonate membrane. HCT116 cells (1.5 × 105) were seeded into the upper chamber with serum-free medium containing 0.2% BSA, while the lower chamber contained medium supplemented with 15% FBS. Compound 17c was applied at the specified concentrations to both sides of the membrane. After 24 h of incubation at 37 °C, the cells were fixed in methanol and stained with 0.1% crystal violet. Non-migrated cells on the upper surface of the membrane were removed using a cotton swab. Five random fields per well were imaged under brightfield microscopy, and the number of migrated cells was quantified using ImageJ software (1.53c). Each experiment was performed in triplicate.

2.10. Plasma Protein Binding in Plasma

A stock solution (20 mM) of compound 17c was prepared in DMSO. The stock solution was then diluted into 100 μM with acetonitrile. The HT Dialysis apparatus was assembled according to the manufacturer’s instructions. Then, 150 μL of the plasma spiked with 1 μM final compound concentrations was added to one side of the well and 150 μL of PBS was added to the opposite side of the well (n = 3). The plate was equilibrated for 6 h at 37 °C in a 5% CO2 incubator. For T0 samples, 200 μL of spiked plasma was stored at –20 °C. For T6 samples, 200 μL of spiked plasma was incubated at 37 °C in a 5% CO2 incubator for 6 h. After incubation, 50 μL of the samples (plasma or PBS) was collected into a vessel containing 50 μL of the opposite matrix. Then, 300 μL of acetonitrile with IS was added into the vessels. The 96-well plate was centrifuged at 4000 rpm for 10 min. Then, 50 μL of supernatant was mixed with 50 μL of ddH2O and then injected onto the LC-MS/MS system for analysis.

2.11. Pharmacokinetic Properties

Male and female SD rats (SLRC Laboratory Animal Inc., Shanghai, China) were used for the study. In the oral (PO) administration, 3 rats per group were given a single 10 mg/kg dose. Blood samples (100 μL per time point) were collected via jugular vein cannula and taken at 5, 15, and 30 min, and 1, 2, 4, 8, and 24 h post-dose. For the intravenous (IV) administration, 3 rats per group received 2.0 mg/kg, and blood samples were collected at 5, 15, and 30 min, and 1, 2, and 4 h after dosing. Rats were fasted overnight prior to dosing and remained fasted for 6 h post-dose. Plasma was separated by centrifugation and stored at –40 °C until analysis. Compound concentrations were determined by LC/MS/MS (Shimadzu LC-30AD), and pharmacokinetic parameters, including Cmax, Tmax, T1/2, and AUC0-inf, were calculated.

3. Results and Discussion

3.1. Chemistry

The synthesis of target compounds and the key intermediates are described in Scheme 1 and Scheme 2, respectively. The structures of compounds were confirmed by 1H-NMR, 13C-NMR and HRMS spectroscopy.
As depicted In Scheme 1, a condensation reaction where 1-((4-fluorophenyl)carbamoyl)cyclopropane-1-carboxylic acid (9) was combined with readily accessible 4-aminophenol (10) at ambient temperature was employed, yielding the intermediate N-(4-fluorophenyl)-N-(4-hydroxyphenyl)cyclopropane-1,1-dicarboxamide (11) [36]. Subsequently, the intermediate N-(4-((2-chloropyrimidin-4-yl)oxy)phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (12) was produced via an SN2 reaction between intermediate 11 with 2,4-dichloropyrimidine [37]. Finally, the desired compounds 13a13k were obtained by reacting intermediate 12 with various substituted anilines, which were either commercially available or prepared in our prior research [38].
In Scheme 2, the preparation of the crucial intermediate N-(4-hydroxyphenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (15) commenced with a condensation process involving 1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxylic acid (14) and 4-aminophenol (10) [32]. Intermediate 15 was then reacted with 2,4-dichloropyrimidine, yielding the pivotal intermediate N-(4-((2-chloropyrimidin-4-yl)oxy)phenyl)-1,5-dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-pyrazole-4-carboxamide (16). The target compounds 17a17h were subsequently obtained by carrying out a nucleophilic substituted reaction between intermediate 16 and various substituted anilines.

3.2. Kinase Inhibitory Activities

All the target compounds (Supplementary Material, S1) were initially screened for their inhibitory activities on Mer and c-Met kinases compared to the lead compound (18c), and the results are depicted in Table 1 and Table 2. The corresponding curves are shown in Supplementary S3 and S4.
As summarized in Table 1, the substituents (R1) at the terminal pyrimidine amine group significantly modulate inhibitory potency against Mer and c-Met kinases. Compounds 13b, 13j, and 13k exhibited moderate to potent dual inhibitory, with Mer IC50 values ranging from 13.1 ± 2.1~37.3 ± 10.6 nM and c-Met IC50 values of 25.9 ± 7.7~48.6 ± 11.2 nM, compared to the lead compound 18c (Mer IC50: 18.5 ± 2.3 nM, c-Met IC50: 33.6 ± 4.3 nM). Strikingly, compound 13e demonstrated the highest dual potency, achieving IC50 values of 7.5 ± 1.6 nM (Mer) and 7.5 ± 1.1 nM (c-Met), representing a 2.5-fold and 4.5-fold improvement over 18c, respectively. In contrast, compounds 13c and 13h showed reduced Mer inhibitory activity while maintaining c-Met inhibitory activity comparable to 18c. Conversely, compounds 13d and 13i exhibited significantly reduced inhibitory activities against both Mer and c-Met kinase.
A novel series of pyrimidine derivatives were designed to optimize dual Mer/c-Met inhibition. As shown in Table 2, the Mer kinase inhibitory activities varied significantly across the series. Compounds 17d, 17e, and 17g exhibited reduced inhibitory potency, with Mer IC50 values of 84.1 ± 13.9, 69.8 ± 12.5, and >1000 nM, respectively. In contrast, compounds 17a, 17b, and 17h retained activity comparable to the lead compound, featuring IC50 values of 26.2 ± 7.1, 12.1 ± 1.3, and 11.9 ± 1.7 nM. Notably, compounds 17c and 17f showcased superior inhibition, achieving Mer IC50 values of 6.4 ± 1.8 nM and 5.5 ± 0.8 nM.
For c-Met kinase inhibition, compounds 17c, 17f, and 17h, exhibited comparable IC50 values ranging from 25.0 ± 3.5 to 45.0 ± 6.8 nM. However, the potency of 17a, 17b, 17d, 17e, and 17g decreased (IC50 values from 100.7 ± 18.5 nM to 235.6 ± 25.6 nM), indicating that specific structural optimization at the pyrimidine scaffold detrimentally impacts c-Met binding.
The findings converge to demonstrate that the designed compounds exhibit robust inhibitory effects on both Mer and c-Met targets. The steric bulk, electronic properties, and hydrophobicity of the R1 substituents have a decisive impact on dual activity. The R1 structure of 13e likely optimizes interactions with the kinase ATP pocket (e.g., hydrogen bonding, hydrophobic packing), whereas the R1 groups in 13d and 13i may introduce steric hindrance or disrupt critical binding interactions. Modification of the pyridine ammine scaffold can selectively enhance Mer inhibition (e.g., 17c/f), while most modifications (e.g., 17a/b/d/e/g) weaken c-Met binding, suggesting that c-Met is more sensitive to structural alterations. There may be local differences in the ATP-binding pockets of Mer and c-Met (e.g., flexibility or key amino acids), which could result in differing structural optimization on the two targets. A general structure–activity relationship can be visualized in Figure 6 and Figure 7. While the designed compounds demonstrate potent Mer kinase inhibition, their inhibitory activity still falls short of the positive control, indicating the need for further optimization to improve potency.

3.3. In Vitro Stability in Liver Microsomes

Metabolic stability is a critical determinant of drug-like properties. To prioritize compounds for further development, we evaluate the human liver microsomal stability of selected candidates demonstrating potent dual Mer/c-Met dual inhibition in vitro. As shown in Table 3, compounds 13b, 13e, 13j, 13k, 17c, and 17f were subjected to microsome assays to determine their half-life and intrinsic clearance. Compound 17c demonstrated superior metabolic stability, with an extended t1/2 of 147.0 min and low CLint of 0.026 mL/min/mg, whereas 17f exhibited a shorter t1/2 of 51.0 min and higher CLint of 0.068 mL/min/mg. Notably, all other compounds showed rapid hepatic clearance (t1/2 < 40 min, CLint > 0.068 mL/min/mg), suggesting limited metabolic resistance. The results highlight 17c as the most metabolically stable candidate within this series.

3.4. Antiproliferation Assay In Vitro

Compound 17c was further evaluated for its antiproliferative activity against three cancer cell lines with high Mer and c-Met expression (HCT116 colon cancer, CAKI-1 renal cancer, and PC-3 prostate cancer) using the CCK-8 assay [5,39,40,41,42]. As summarized in Table 4, 17c demonstrated superior antiproliferative activity compared to cabozantinib across all tested cell lines. Notably, 17c exhibited the greatest potency with HCT116 cells, with an IC50 value of 0.46 ± 0.06 μmol/L—a 17.1-fold improvement over cabozantinib. In CAKI-1 cells, 17c showed an IC50 of 2.31 ± 0.67 μmol/L, representing a 2.0-fold enhancement compared to cabozantinib. Similarly, against PC-3 cells, 17c achieved an IC50 of 3.79 ± 1.09 μmol/L, which is 4.1 times more effective than cabozantinib. These results collectively establish compound 17c as a potent dual Mer/c-Met inhibitor with significant therapeutic potential. The dose–response curves are provided in Supplementary S5.

3.5. Molecular Docking Study of Compound 17c

Molecular docking was conducted to validate the binding mode of compound 17c with Mer kinase (PDB: 4M3Q) and c-Met (PDB: 3LQ8) kinases. As illustrated in Figure 8, the docking pose of 17c in Mer kinase revealed three critical interactions: (1) the aminopyrimidine moiety acts as hydrogen bond acceptor with Leu593; (2) the amide side chain forms a hydrogen bond with Lys619; and (3) the benzene ring of the side chain engages in a π-π stacking interaction. The docking interaction energy was −68.7952 KJ/mol. For c-Met kinase (Figure 9), 17c exhibited a similar binding mode: (1) the aminopyrimidine core forms a hydrogen bond with Met1160; (2) the amide group forms hydrogen bonds with Lys1110 and Phe1223; and (3) the pyrimidine group participates in π-π interaction. The docking interaction energy was −91.7674 KJ/mol. These results confirm that the 2-substitutedaniline pyrimidine group effectively occupies the ATP-binding pockets of both kinases through conserved hydrogen bonding and hydrophobic interactions. The structural coherence between the predicted binding modes and enzymes inhibitory data strongly supports the rational design strategy, validating the scaffold’s potential for dual Mer/c-Met inhibition.
We also systematically compared our compound’s docking results with that of cabozantinib and foretinib (XL880). For c-Met kinase, the docking results of cabozantinib with c-Met kinase showed that the quinoline core could form a hydrogen bond with Met1160, and that the amide group participates in hydrogen bonding with Lys1110 and Asp1222. The docking results of foretinib with c-Met showed that the quinoline core could form a hydrogen bond with Met1160, and that the amide group participates in hydrogen bonding with Lys1110 and Asp1222. The docking results confirm that compound 17c shares a conserved binding mode with both the reference drug cabozantinib and the co-crystallized c-Met inhibitor foretinib (PDB: 3LQ8).
For Mer kinase, the binding mode of cabozantinib with Mer kinase remains unreported, and co-crystallized Mer ligands possess distinct chemotypes. We conducted the docking of cabozantinib and 17c with Mer kinase, and the results showed that cabozantinib’s amide group forms hydrogen bonds with the hinge region (Pro672 and Met674), while the quinoline moiety occupies a hydrophobic cavity without direct hydrogen bonding. Compound 17c’s aminopyrimidine moiety acts as a hydrogen bond acceptor with Leu593 and the amide side chain forms a hydrogen bond with Lys619. The docking results showed that compound 17c possesses better affinity than that of the reference drug cabozantinib.

3.6. Western Blot Assay

The inhibitory effects of compound 17c on Mer and c-Met kinase phosphorylation were assessed using Western blot analysis. As shown in Figure 10, increasing concentrations of 17c resulted in a reduction in the phosphorylation levels of both Mer and c-Met kinases. At a concentration of 2.5 μM, 17c significantly inhibited the phosphorylation of these kinases compared to the positive control, demonstrating its potential to effectively block Mer and c-Met kinase activation.

3.7. The hERG Test

To evaluate the potential cardiotoxicity risk associated with compound 17c, we assessed its inhibitory activity against the hERG potassium channel, a key determinant of cardiac safety [33]. As summarized in Table 5, 17c demonstrated negligible hERG inhibition (IC50 > 40 μM), indicating that a low risk of hERG activity was observed despite its potent dual inhibition of Mer and c-Met kinases.

3.8. Apoptosis Assay

To evaluate the apoptosis-inducing capability of compound 17c, the HCT116 colon cancer cell line was treated with varying concentrations of the compound. Apoptosis was evaluated using the TUNEL assay. DAPI staining (blue) served as the positive control, while apoptotic cells were identified by TUNEL staining (green) after 48 h of incubation with compound 17c, as shown in Figure 11. The quantity of apoptotic cells was measured at different concentrations of 17c, resulting in an ED50 value of 2.036 μM. These results demonstrate that compound 17c efficiently triggers apoptosis in HCT116 cells, thereby emphasizing its potential as a highly promising candidate for additional research and development.

3.9. Transwell Assay

The Transwell migration assay shown in Figure 12 was performed to evaluate the effect of compound 17c on the migration of HCT116 cells. A decrease in the number of cells migrating through the membrane was observed with an increasing concentration of 17c. These results suggest that compound 17c effectively inhibits the migration capacity of HCT116 colon cancer cells in a concentration-dependent manner.

3.10. Plasma Protein-Binding Affinity

The plasma protein-binding (PPB) affinity significantly influences the compound’s free fraction, bioavailability, tissue distribution, and elimination kinetics, thereby impacting the pharmacokinetic properties. The PPB profile of compound 17c was assessed via equilibrium dialysis, with warfarin serving as a positive control. As shown in Table 6, 17c exhibited high species-independent protein binding, with unbound fractions of 1.58% in human plasma and 1.78% in mouse plasma, corresponding to PPB values of 98.4% and 98.2%, respectively. Combined with its extended half-life and low clearance, the high PPB of 17c may contribute to sustained target engagement.

3.11. Pharmacokinetic Properties

The Pharmacokinetic (PK) profiles of compound 17c were evaluated in Sprague Dawley (SD) rats following single-dose administration via the oral (PO) and intravenous (iv) route, and the Animal Care and Use Committee (ACUC) approval documentation was IACUC-ALM-02. As summarized in Table 7, the oral administration of 17c achieved a peak plasma concentration (Cmax) of 740 ng/mL, at Tmax = 4.0 h, with a half-time (T1/2) of 3.46 h and a mean residence time (MRT_inf) of 6.52 h. Compared to lead compound 18c, compound 17c demonstrated significantly improved PK properties, including a prolonged oral half-time, reduced clearance, an extended mean residence time, and enhanced oral bioavailability (45.3%). These data indicate that 17c exhibits favorable PK properties, with sustained drug exposure and optimized absorption characteristics.

4. Conclusions

We report a novel series of dual Mer/c-Met inhibitors, with 17c emerging as the lead candidate. Molecular docking revealed that 17c forms hydrogen bonds with Mer (Leu593/Lys619) and c-Met (Met1160/Lys1110/Phe1223), complemented by π-π interaction. 17c exhibited potent enzymatic inhibition and superior antiproliferative activity over cabozantinib against three cancer cell lines. Pharmacokinetically, 17c showed 45.3% oral bioavailability (16-fold higher than 18c), an extended half-life (3.46 h), and low hepatic clearance (0.026 mL/min/mg). Safety profiling indicated minimal hERG inhibition. Mechanistically, 17c induced apoptosis and suppressed HCT116 migration. These data position 17c as a promising dual Mer/c-Met inhibitor warranting further development.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biom15081180/s1, S1: Supporting Tables; S2: Raw materials of biology; S3: Mer inhibitory results; S4: c-Met inhibitory results; S5: Antiproliferative activities against cancer cell lines; S6–S81: 1H-NMR, 13C-NMR, HRMS, and HPLC of target compounds; S82: A quantification and a fold change analysis of western blot analysis; S83: Re-docking files; S84: Alignment of 17c and foretinib; S85: Original Western blot images.

Author Contributions

Conceptualization, Investigation, Project administration, Writing—original draft, and Writing—review and editing, J.Y.; Investigation, Methodology, Project administration, Software, Writing—original draft, and Writing—review and editing, D.H.; Data curation, Investigation, and Methodology, R.W.; Formal analysis, Investigation, and Software, P.F.; Investigation, Methodology, and Validation, R.L.; Data curation, Investigation, Methodology, Project administration, and Writing—review and editing, D.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially funded by Science Research Project of Hebei Education Department (BJK2024144), Special Project for Basic Research Business Expenses of Provincial Universities (JCYJ2023004), Doctoral Research Fund Project (BSZ2022004), College Students’ Innovation and Entrepreneurship Training Program (202314432007), and the National Natural Science Foundation of China (No.82003601).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Animal Care and Use Committee (ACUC) (protocol code IACUC-ALM-02, approval date 2023-06-08).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding authors.

Acknowledgments

Thank the Instrumental Analysis Center of Hebei University of Science and Technology for providing HRMS and NMR tests.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wang, X.; Frye, S. Chapter Nineteen-Mer Receptor Tyrosine Kinase: Therapeutic Opportunities in Oncology, Virology, and Cardiovascular Indications. Annu. Rep. Med. Chem. 2014, 49, 301–314. [Google Scholar] [CrossRef]
  2. Graham, D.K.; DeRyckere, D.; Davies, K.D.; Earp, H.S. The TAM family: Phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat. Rev. Cancer 2014, 14, 769–785. [Google Scholar] [CrossRef] [PubMed]
  3. Yu, S.; Yi, M.; Qin, S.; Wu, K.M. Next generation chimeric antigen receptor T cells: Safety strategies to overcome toxicity. Mol. Cancer. 2019, 18, 125. [Google Scholar] [CrossRef] [PubMed]
  4. Smart, S.; Vasileiadi, E.; Wang, X. The Emerging Role of TYRO3 as a Therapeutic Target in Cancer. Cancers 2018, 10, 474. [Google Scholar] [CrossRef] [PubMed]
  5. Baladi, T.; Abet, V.; Piguel, S. State-of-the-art of small molecule inhibitors of the TAM family: The point of view of the chemist. Eur. J. Med. Chem. 2015, 105, 220–237. [Google Scholar] [CrossRef]
  6. Davra, V.; Kumar, S.; Geng, K.; Calianese, D.; Mehta, D.; Gadiyar, V.; Kasikara, C.; Lahey, K.C.; Chang, Y.J.; Wichroski, M.; et al. Axl and Mertk Receptors Cooperate to Promote Breast Cancer Progression by Combined Oncogenic Signaling and Evasion of Host Antitumor Immunity. Cancer Res. 2021, 81, 698–712. [Google Scholar] [CrossRef]
  7. Park, H.J.; Baen, J.Y.; Lee, Y.J.; Choi, Y.H.; Kang, J.L. The TAM-family receptor Mer mediates production of HGF through the RhoA-dependent pathway in response to apoptotic cells. Mol. Biol. Cell 2012, 23, 3254–3265. [Google Scholar] [CrossRef]
  8. Nguyen, K.Q.; Tsou, W.I.; Kotenko, S.; Birge, R.B. TAM receptors in apoptotic cell clearance, autoimmunity, and cancer. Autoimmunity 2013, 46, 294–297. [Google Scholar] [CrossRef]
  9. Sayama, A.; Okado, K.; Yamaguchi, M.; Samata, N.; Imaoka, M.; Kai, K.; Mori, K. The impact of the timing of dosing on the severity of UNC569-induced ultrastructural changes in the mouse retina. Toxicol. Pathol. 2020, 48, 669–676. [Google Scholar] [CrossRef]
  10. Koda, Y.; Itoh, M.; Tohda, S. Effects of MERTK inhibitors UNC569 and UNC1062 on the growth of acute myeloid Leukaemia cells. Anticancer Res. 2018, 38, 199–204. [Google Scholar] [CrossRef]
  11. DeRyckere, D.; Lee-Sherick, A.B.; Huey, M.G.; Hill, A.A.; Tyner, J.W.; Jacobsen, K.M.; Page, L.S.; Kirkpatrick, G.G.; Eryildiz, F.; Montgomery, S.A.; et al. UNC2025, a MERTK small-molecule inhibitor, is therapeutically effective alone and in combination with methotrexate in leukemia models. Clin. Cancer Res. 2017, 23, 1481–1492. [Google Scholar] [CrossRef] [PubMed]
  12. Yan, D.; Huelse, J.M.; Kireev, D.; Tan, Z.; Chen, L.; Goyal, S.; Wang, X.; Frye, S.V.; Behera, M.; Schneider, F.; et al. MERTK activation drives osimertinib resistance in EGFR-mutant non–small cell lung cancer. J. Clin. Investig. 2022, 132, e150517. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, W.; DeRyckere, D.; Hunter, D.; Liu, J.; Stashko, M.A.; Minson, K.A.; Cummings, C.T.; Lee, M.; Glaros, T.G.; Newton, D.L.; et al. UNC2025, a potent and orally bioavailable MER/FLT3 dual inhibitor. J. Med. Chem. 2014, 57, 7031–7041. [Google Scholar] [CrossRef] [PubMed]
  14. Lee-Sherick, A.B.; Jacobsen, K.M.; Henry, C.J.; Huey, M.G.; Parker, R.E.; Page, L.S.; Hill, A.A.; Wang, X.; Frye, S.V.; Earp, H.S.; et al. MERTK inhibition alters the PD-1 axis and promotes anti-leukemia immunity. JCI Insight 2018, 3, e97941. [Google Scholar] [CrossRef]
  15. Zhang, W.; Zhang, D.; Stashko, M.A.; DeRyckere, D.; Hunter, D.; Kireev, D.; Miley, M.J.; Cummings, C.; Lee, M.; Norris-Drouin, J.; et al. Pseudo-cyclization through intramolecular hydrogen bond enables discovery of pyridine substituted pyrimidines as new Mer kinase inhibitors. J. Med. Chem. 2013, 56, 9683–9692. [Google Scholar] [CrossRef]
  16. Shi, C.; Li, X.; Wang, X.; Ding, N.; Ping, Y.; Shi, Y.; Mi, L.; Lai, Y.; Song, Y.; Zhu, J. The proto-oncogene Mer tyrosine kinase is a novel therapeutic target in mantle cell lymphoma. J. Hematol. Oncol. 2018, 11, 43. [Google Scholar] [CrossRef]
  17. Hu, L.; Wang, X.; Song, Z.; Chen, F.; Wu, B. Leveraging CAR macrophages targeting c-Met for precision immunotherapy in pancreatic cancer: Insights from single-cell multi-omics. Mol. Med. 2024, 30, 231. [Google Scholar] [CrossRef]
  18. Barbosa-Matos, C.; Borges-Pereira, C.; Libório-Ramos, S.; Fernandes, R.; Oliveira, M.; Mendes-Frias, A.; Silvestre, R.; Osório, N.S.; Bastos, H.N.; Santos, R.F.; et al. Deregulated immune cell recruitment orchestrated by c-MET impairs pulmonary inflammation and fibrosis. Resp. Res. 2024, 25, 257. [Google Scholar] [CrossRef]
  19. Faiella, A.; Riccardi, F.; Cartenì, G.; Chiurazzi, M.; Onofrio, L. The emerging role of c-Met in carcinogenesis and clinical implications as a possible therapeutic target. J. Oncol. 2022, 2022, 5179182. [Google Scholar] [CrossRef]
  20. Lee, M.; Jain, P.; Wang, F.; Ma, P.C.; Borczuk, A.; Halmos, B. MET alterations and their impact on the future of non-small cell lung cancer (NSCLC) targeted therapies. Expert Opin. Ther. Tar. 2021, 25, 249–268. [Google Scholar] [CrossRef]
  21. Gou, Q.; Gou, Q.; Gan, X.; Xie, Y. Novel therapeutic strategies for rare mutations in non-small cell lung cancer. Sci. Rep. 2024, 14, 10317. [Google Scholar] [CrossRef]
  22. Cen, S.Y.; Lin, F.; Li, X.; Hu, Y.; Liu, J.P.; Xue, Z.; Gao, Y.; Sun, Y.P.; Zhu, S.; Dang, Y.; et al. Crizotinib and its enantiomer suppress ferroptosis by decreasing PE-O-PUFA content. Cell Death Discov. 2024, 10, 360. [Google Scholar] [CrossRef]
  23. Elghawy, O.; Barsouk, A.; Xu, J.; Chen, S.; Cohen, R.B.; Sun, L. Real word outcomes of cabozantinib therapy in poorly differentiated thyroid carcinoma. Eur. Thyroid J. 2024, 13, e240225. [Google Scholar] [CrossRef]
  24. Patil, J.; Bhattacharya, S.; Saoji, S.D.; Dande, P. Cabozantinib-phospholipid complex for enhanced solubility, bioavailability, and reduced toxicity in liver cancer. Ther. Deliv. 2025, 16, 25–41. [Google Scholar] [CrossRef] [PubMed]
  25. Miao, K.; Zhang, X.; Wang, H.; Si, X.; Zhang, L. Savolitinib versus crizotinib for treating MET positive non-small cell lung cancer. Thorac. Cancer 2023, 14, 1162–1170. [Google Scholar] [CrossRef] [PubMed]
  26. Qi, Y.; Li, J.; Lin, S.; Wu, S.; Chai, K.; Jiang, X.; Qian, J.; Jiang, C. A real-world pharmacovigilance study of FDA adverse event reporting system events for Capmatinib. Sci. Rep. 2024, 14, 11388. [Google Scholar] [CrossRef] [PubMed]
  27. Dhillon, S. Capmatinib: First approval. Drugs 2020, 80, 1125–1131. [Google Scholar] [CrossRef]
  28. Giusy, T.; Giacomo, L.; Pasquale, L.; Valeria, F.; Michele, T.; Lorenzo, T.; Maaria, P.C.; Stefano, M.; Cecilia, P. Evidence of Pyrimethamine and Cycloguanil Analogues as Dual Inhibitors of Trypanosoma brucei Pteridine Reductase and Dihydrofolate Reductase. Pharmaceuticals 2021, 14, 636. [Google Scholar] [CrossRef]
  29. Ballard, C.G. Advances in the treatment of Alzheimer’s disease: Benefits of dual cholinesterase inhibition. Eur. Neurol. 2002, 47, 64–70. [Google Scholar] [CrossRef]
  30. Naresh, G.K.; Guruprasad, L. Enhanced metastable state models of TAM kinase binding to cabozantinib explains the dynamic nature of receptor tyrosine kinases. J. Biomol. Struct. Dyn. 2021, 39, 1213–1235. [Google Scholar] [CrossRef]
  31. Huang, D.W.; Chen, Y.; Yang, J.X.; Zhao, B.Y.; Wang, S.Y.; Chai, T.T.; Cui, J.; Zhou, X.L.; Shang, Z.H. Design, Synthesis, and Biological Evaluation of 2-Substituted Aniline Pyrimidine Derivatives as Potent Dual Mer/c-Met Inhibitors. Molecules 2024, 29, 475. [Google Scholar] [CrossRef]
  32. Liu, J.; Zhang, W.; Stashko, M.A.; DeRyckere, D.; Cummings, C.T.; Hunter, D.; Yang, C.; Jayakody, C.N.; Cheng, N.; Simpson, C.; et al. UNC1062, a new and potent Mer inhibitor. Eur. J. Med. Chem. 2013, 65, 83–93. [Google Scholar] [CrossRef]
  33. Zhao, J.C.; Zhang, D.; Zhang, W.H.; Stashko, M.A.; DeRyckere, D.; Vasileiadi, E.; Parker, R.E.; Hunter, D.; Liu, Q.; Zhang, Y.; et al. Highly selective MERTK inhibitors achieved by a single methyl group. J. Med. Chem. 2018, 61, 10242–10254. [Google Scholar] [CrossRef] [PubMed]
  34. Lien, V.T.; Pettersen, S.; Haugen, M.H.; Olberg, D.E.; Mælandsmo, G.M.; Klaveness, J. Design, synthesis and biological evaluation of 6-substituted quinolines derived from cabozantinib as c-Met inhibitors. Arch. Pharm. 2019, 352, 1900101. [Google Scholar] [CrossRef] [PubMed]
  35. Chan, S.P.; Zhang, Y.N.; Wang, J.; Yu, Q.C.; Peng, X.; Zou, J.; Zhou, L.C.; Tan, L.; Duan, Y.X.; Zhou, Y.; et al. Discovery of 3-aminopyrazole derivatives as new potent and orally bioavailable AXL inhibitors. J. Med. Chem. 2022, 65, 15374–15390. [Google Scholar] [CrossRef] [PubMed]
  36. Wei, D.S.; Fan, H.; Zheng, K.; Qin, X.; Yang, L.; Yang, Y.; Duan, Y.; Zhang, Q.; Zeng, C.; Hu, L. Synthesis and anti-tumor activity of [1, 4] dioxino [2, 3-f] quinazoline derivatives as dual inhibitors of c-Met and VEGFR-2. Bioorg. Chem. 2019, 88, 102916. [Google Scholar] [CrossRef]
  37. Gao, G.R.; Li, M.Y.; Lv, Y.C.; Cao, S.F.; Tong, L.J.; Wei, L.X.; Ding, J.; Xie, H.; Duan, W.H. Design, synthesis and biological evaluation of biphenylurea derivatives as VEGFR-2 kinase inhibitors (II). Chin. Chem. Lett. 2016, 27, 200–204. [Google Scholar] [CrossRef]
  38. Park, H.; Jung, H.Y.; Mah, S.; Hong, S. Systematic computational design and identification of low Picomolar inhibitors of Aurora kinase A. J. Chem. Inf. Model. 2018, 58, 700–709. [Google Scholar] [CrossRef]
  39. Huang, D.W.; Yang, J.X.; Zhang, Q.W.; Wang, G.; Zhang, Z.X.; Zhang, Y.; Li, J.Q. Structure-guided design and development of novel N-phenylpyrimidin-2-amine derivatives as potential c-Met inhibitors. Eur. J. Med. Chem. 2021, 223, 113648. [Google Scholar] [CrossRef]
  40. Hwang, S.T.; Um, J.Y.; Chinnathambi, A.; Alharbi, S.A.; Narula, A.S.; Namjoshi, O.A.; Blough, B.E.; Ahn, K.S. Evodiamine mitigates cellular growth and promotes apoptosis by targeting the c-Met pathway in prostate cancer cells. Molecules 2020, 25, 1320. [Google Scholar] [CrossRef]
  41. Li, J.; Tan, G.S.; Cai, Y.B.; Liu, R.H.; Xiong, X.L.; Gu, B.H.; He, W.; Liu, B.; Ren, Q.Y.; Wu, J.P.; et al. A novel Apigenin derivative suppresses renal cell carcinoma via directly inhibiting wild-type and mutant MET. Biochem. Pharmacol. 2021, 190, 114620. [Google Scholar] [CrossRef]
  42. Gu, Y.; Chen, Y.; Wei, L.; Wu, S.; Shen, K.; Liu, C.; Dong, Y.; Zhao, Y.; Zhang, Y.; Zhang, C.; et al. ABHD5 inhibits YAP-induced c-Met overexpression and colon cancer cell stemness via suppressing YAP methylation. Nat. Commun. 2021, 12, 6711. [Google Scholar] [CrossRef]
Figure 1. The typical Mer inhibitors.
Figure 1. The typical Mer inhibitors.
Biomolecules 15 01180 g001
Figure 2. The representative c-Met inhibitors.
Figure 2. The representative c-Met inhibitors.
Biomolecules 15 01180 g002
Figure 3. Cabozantinib binding mode in Mer kinase (PDB: 4M3Q), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Figure 3. Cabozantinib binding mode in Mer kinase (PDB: 4M3Q), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Biomolecules 15 01180 g003
Figure 4. Cabozantinib binding mode in Mer kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Figure 4. Cabozantinib binding mode in Mer kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Biomolecules 15 01180 g004
Figure 5. Design strategy of target compounds.
Figure 5. Design strategy of target compounds.
Biomolecules 15 01180 g005
Figure 6. SAR study of the designed compounds with side chain A.
Figure 6. SAR study of the designed compounds with side chain A.
Biomolecules 15 01180 g006
Figure 7. SAR study of the designed compounds with side chain B.
Figure 7. SAR study of the designed compounds with side chain B.
Biomolecules 15 01180 g007
Figure 8. 17c binding mode in Mer kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Figure 8. 17c binding mode in Mer kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Biomolecules 15 01180 g008
Figure 9. 17c binding mode in c-Met kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Figure 9. 17c binding mode in c-Met kinase (PDB: 3LQ8), (A) 3D ligand–protein interactions; (B) 2D ligand interaction diagram.
Biomolecules 15 01180 g009
Figure 10. Western blot analysis was applied to analyze the expression and phosphorylation of c-Met and MERTK in HCT116 cells with staurosporine or increasing doses of compound 17c. All experiments were performed at least three times. Original Western blot images can be found in Figure S85.
Figure 10. Western blot analysis was applied to analyze the expression and phosphorylation of c-Met and MERTK in HCT116 cells with staurosporine or increasing doses of compound 17c. All experiments were performed at least three times. Original Western blot images can be found in Figure S85.
Biomolecules 15 01180 g010
Figure 11. Compound 17c induced cell apoptosis in HCT116 cancer cells. (A) Quantitative analysis of TUNEL staining. (B) Representative TUNEL and DAPI-stained images. Magnification, x100. HCT116 cells grown on cover slip, treated with various indicated doses of 17c for 48 h, and stained for TUNEL (green). The number of TUNEL-positive cells was counted from five non-overlap random fields per group. DAPI (blue). Data are representative of three independent experiments.
Figure 11. Compound 17c induced cell apoptosis in HCT116 cancer cells. (A) Quantitative analysis of TUNEL staining. (B) Representative TUNEL and DAPI-stained images. Magnification, x100. HCT116 cells grown on cover slip, treated with various indicated doses of 17c for 48 h, and stained for TUNEL (green). The number of TUNEL-positive cells was counted from five non-overlap random fields per group. DAPI (blue). Data are representative of three independent experiments.
Biomolecules 15 01180 g011
Figure 12. Compound 17c inhibited cell migration in HCT116 cancer cells. (A) Quantification of transwell assays. (B) Representative images of transwell assay of HCT116 cells treated with compound 17c for 24 h.
Figure 12. Compound 17c inhibited cell migration in HCT116 cancer cells. (A) Quantification of transwell assays. (B) Representative images of transwell assay of HCT116 cells treated with compound 17c for 24 h.
Biomolecules 15 01180 g012
Scheme 1. Reagents and conditions: (a) EDC·HCl/DMF, r.t., 6 h; (b) 2,4-dichloropyrimidine, K2CO3/DMF, 80 °C, 6 h; (c) PTSA/DMF, 90 °C, 4 h.
Scheme 1. Reagents and conditions: (a) EDC·HCl/DMF, r.t., 6 h; (b) 2,4-dichloropyrimidine, K2CO3/DMF, 80 °C, 6 h; (c) PTSA/DMF, 90 °C, 4 h.
Biomolecules 15 01180 sch001
Scheme 2. Reagents and conditions: (a) HBTU/TEA/DMF, r.t., 8 h; (b) 2,4-dichloropyrimidine, K2CO3/DMF, 80 °C, 4.5 h; (c) PTSA/DMF, 90 °C, 4.0 h.
Scheme 2. Reagents and conditions: (a) HBTU/TEA/DMF, r.t., 8 h; (b) 2,4-dichloropyrimidine, K2CO3/DMF, 80 °C, 4.5 h; (c) PTSA/DMF, 90 °C, 4.0 h.
Biomolecules 15 01180 sch002
Table 1. Inhibitory activities of compound 13a13k against Mer and c-Met kinase.
Table 1. Inhibitory activities of compound 13a13k against Mer and c-Met kinase.
Compd.R1IC50 (nM) 1
Merc-Met
13aBiomolecules 15 01180 i00121.7 ± 5.863.3 ± 9.1
13bBiomolecules 15 01180 i00224.3 ± 5.725.9 ± 7.7
13cBiomolecules 15 01180 i003110.1 ± 15.748.9 ± 6.4
13dBiomolecules 15 01180 i004640.2 ± 82.2728.1 ± 131.4
13eBiomolecules 15 01180 i0057.5 ± 1.67.5 ± 1.1
13fBiomolecules 15 01180 i006>1000>1000
13gBiomolecules 15 01180 i007>1000>1000
13hBiomolecules 15 01180 i00872.0 ± 8.941.6 ± 8.5
13iBiomolecules 15 01180 i00995.3 ± 20.188.6 ± 13.9
13jBiomolecules 15 01180 i01013.1 ± 2.148.6 ± 11.2
13kBiomolecules 15 01180 i01137.3 ± 10.635.4 ± 10.0
18cBiomolecules 15 01180 i01218.5 ± 2.333.6 ± 4.3
cabozantinib 0.6 ± 0.11.4 ± 0.2
1 Data are means from three independent experiments.
Table 2. Inhibitory activities of compound 17a17h against Mer and c-Met kinase.
Table 2. Inhibitory activities of compound 17a17h against Mer and c-Met kinase.
Compd.R2IC50 (nM) 1
Merc-Met
17aBiomolecules 15 01180 i01326.2 ± 7.1233.4 ± 25.4
17bBiomolecules 15 01180 i01412.1 ± 1.3100.7 ± 18.5
17cBiomolecules 15 01180 i0156.4 ± 1.826.1 ± 7.7
17dBiomolecules 15 01180 i01684.1 ± 13.9137.1 ± 34.3
17eBiomolecules 15 01180 i01769.8 ± 12.5235.6 ± 25.6
17fBiomolecules 15 01180 i0185.5 ± 0.825 ± 3.5
17gBiomolecules 15 01180 i019>1000139.7 ± 18.9
17hBiomolecules 15 01180 i02011.9 ± 1.745.0 ± 6.8
18cBiomolecules 15 01180 i02118.5 ± 2.333.6 ± 4.3
cabozantinib 0.6 ± 0.11.4 ± 0.2
1 Data are means from three independent experiments.
Table 3. Microsome stability study of compound 13b, 13e, 13j, 13k, 13m, 13n, 17c, and 17f.
Table 3. Microsome stability study of compound 13b, 13e, 13j, 13k, 13m, 13n, 17c, and 17f.
Cpd.HUMAN
T1/2 (min)CL (mL/min/mg)
13b38.30.0905
13e30.40.1142
13j27.90.1521
13k38.40.1243
17c147.00.0236
17f51.00.0680
Testosterone32.10.11
Table 4. Cytotoxic activities of compound 17c on cancer cell lines in vitro.
Table 4. Cytotoxic activities of compound 17c on cancer cell lines in vitro.
CpdsIC50 (μM) 1 of 3 Cell Lines
HepG2MDA-MB-231HCT116
17c0.46 ± 0.062.31 ± 0.673.79 ± 1.09
Cabozantinib7.87 ± 2.114.62 ± 0.5415.57 ± 4.39
1 Data are means from three independent experiments.
Table 5. Activity on hERG potassium currents of compound 17c.
Table 5. Activity on hERG potassium currents of compound 17c.
Cpd.IC50 (μM)
17c>40
Cisapride 0.04
Table 6. Stability recovery of compound 17c and positive control in plasma.
Table 6. Stability recovery of compound 17c and positive control in plasma.
Cpd.Con.
(μM)
SpeciesMean FuMean FbStability
(%)
17c1Human0.015898.4%99.3%
17c1Rat0.017898.2%102.2%
Warfarin1Human0.0087799.1%99.8%
Warfarin1Rat0.010299.0%98.2%
Fu (Free drug fraction): refers to the portion of the drug in the plasma that is not bound to proteins. Fu = Area ratio of compound in buffer/Area ratio of compound in plasma. Fb (Bound drug fraction): Refers to the portion of the drug that is bound to plasma proteins. Fb = 100 × (1 − Fu).
Table 7. Pharmacokinetic parameters of compound 17c.
Table 7. Pharmacokinetic parameters of compound 17c.
Adm.Dose
(mg/kg)
AUClast
(h*ng/mL)
T1/2
(h)
Tmax
(h)
Cmax
(ng/mL)
CL_obs (mL/min/kg)MRTINF_obs
(h)
F
(%)
p.o.1046263.464.007406.5245.3
i.v.221543.3316.43.23 
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, J.; Huang, D.; Wang, R.; Fan, P.; Li, R.; Ma, D. Discovery of Novel 2-Substituted Aniline Pyrimidine Based Derivatives as Potent Mer/c-Met Dual Inhibitors with Improvement Bioavailability. Biomolecules 2025, 15, 1180. https://doi.org/10.3390/biom15081180

AMA Style

Yang J, Huang D, Wang R, Fan P, Li R, Ma D. Discovery of Novel 2-Substituted Aniline Pyrimidine Based Derivatives as Potent Mer/c-Met Dual Inhibitors with Improvement Bioavailability. Biomolecules. 2025; 15(8):1180. https://doi.org/10.3390/biom15081180

Chicago/Turabian Style

Yang, Jixia, Daowei Huang, Ruojin Wang, Pengxin Fan, Rourou Li, and Donglai Ma. 2025. "Discovery of Novel 2-Substituted Aniline Pyrimidine Based Derivatives as Potent Mer/c-Met Dual Inhibitors with Improvement Bioavailability" Biomolecules 15, no. 8: 1180. https://doi.org/10.3390/biom15081180

APA Style

Yang, J., Huang, D., Wang, R., Fan, P., Li, R., & Ma, D. (2025). Discovery of Novel 2-Substituted Aniline Pyrimidine Based Derivatives as Potent Mer/c-Met Dual Inhibitors with Improvement Bioavailability. Biomolecules, 15(8), 1180. https://doi.org/10.3390/biom15081180

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop