Next Article in Journal
Difference Analysis of Gas Molecules Diffusion Behavior in Natural Ester and Mineral Oil Based on Molecular Dynamic Simulation
Previous Article in Journal
Fluorescence Polarization Immunoassay for Determination of Enrofloxacin in Pork Liver and Chicken
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Discovery of 8-Amino-Substituted 2-Phenyl-2,7-Naphthyridinone Derivatives as New c-Kit/VEGFR-2 Kinase Inhibitors

1
Nanjing Polytechnic Institute, Nanjing 210048, China
2
Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this paper.
Molecules 2019, 24(24), 4461; https://doi.org/10.3390/molecules24244461
Submission received: 12 November 2019 / Revised: 30 November 2019 / Accepted: 3 December 2019 / Published: 5 December 2019

Abstract

:
The 2,7-naphthyridone scaffold has been proposed as a novel lead structure of MET inhibitors by our group. To broaden the application of this new scaffold, a series of 8-amino-substituted 2-phenyl-2,7-naphthyridin-1(2H)-one derivatives were designed and synthesized. Preliminary biological screening resulted in the discovery of a new lead of c-Kit and VEGFR-2 kinase inhibitors. Compound 9k exhibited excellent c-Kit inhibitory activity, with an IC50 value of 8.5 nM, i.e., it is 38.8-fold more potent than compound 3 (IC50 of 329.6 nM). Moreover, the compounds 10l and 10r exhibited good VEGFR-2 inhibitory activity, with IC50 values of 56.5 and 31.7 nM, respectively, i.e., they are 5.0–8.8-fold more potent than compound 3 (IC50 of 279.9 nM). Molecular docking experiments provided further insight into the binding interactions of the new lead compounds with c-Kit and VEGFR-2 kinase. In this study, an 8-amino-substituted 2-phenyl-2,7-naphthyridin-1(2H)-one scaffold was identified as the new lead structure of c-Kit and VEGFR-2 kinase inhibitors.

Graphical Abstract

1. Introduction

N-Heterocyclic scaffolds are ubiquitous building blocks that are used for pharmaceuticals and agrochemicals and in materials science. Novel ring structures may exhibit unique biological or physical properties [1]. Naphthyridines, which act as bioisosteres of quinolone, universally exist in nature and form an important class of nitrogen-containing heterocyclic compounds [2]. They possess a conjugated π-system and coplanar stiffness, and are therefore a highly valued building block. Since 1893, a number of naphthyridine derivatives have been synthesized and explored for their biological activities in the search for novel drugs. A broad spectrum of biological activities has been associated with the functional derivatives of naphthyridines [3]. Gemifloxacin (1, Figure 1), an antibacterial agent, is the most successful example of a naphthyridine-based drug [4,5]. In recent years, many naphthyridine derivatives have been reported to inhibit protein kinase activity (e.g., PI3Kδ [6], CK2 [7], Akt1/2 [8], Tpl2 [9,10], and MET [11,12,13]) for the treatment of many types of human diseases, including inflammation and cancer [14,15,16,17,18].
Among the six isomeric sub-classes of naphthyridines, 2,7-naphthyridine derivatives have received increasing attention. A typical pharmaceutically and biologically active derivative is lophocladine A (2, Figure 1) [19,20,21,22]. In addition, 2,7-naphthyridine derivatives are widely used as inhibitors of Pdk-1 [23], lumazine synthase [24], PDE-5 [25], TNFα [26], SYK [27], MET [12,13], and other targets. However, the functionalization of 2,7-naphthyridine was found to be especially difficult and only a few methods are available [28], so its application in drug discovery is greatly limited.
In our previous work, a novel 2,7-naphthyridone scaffold was designed to conformationally restrain the key pharmacophoric groups of class II MET inhibitors, resulting in the discovery of the potent preclinical candidate compound 3, which targets MET kinase with a favorable drug-likeness [11]. To further expand the application of the 2,7-naphthyridone scaffold, a series of 8-amino-substituted 2-phenyl-2,7-naphthyridin-1(2H)-one derivatives were designed (Figure 2). A small library of 2,7-naphthyridones with structural diversity in its 8-amino groups, substituted 2-phenyl groups, and 3-substituents was constructed to discover new kinase inhibitors.

2. Results and Discussion

2.1. Chemistry

The synthetic pathway of compounds 9ak and 10as is schematically shown in Scheme 1. According to our reported procedures [11], the key step to constructing the 2-phenyl-2,7-naphthyridone scaffold is the introduction of a 2-phenyl group into the framework. The Ullmann-type coupling of pyridone 4 with 1-fluoro-4-iodobenzene produced 1-phenyl-pyridine-2-one 6a, while the condensation of 2-cyano-N-phenylacetamide 5 with 2,4-pentanedione produced pyridine-2-ones 6bf. The key building block 8-chloro-2-phenyl-2,7-naphthyridone 7 was successfully produced by a condensation–cyclization–chlorination reaction using substance 6 as the substrate. With intermediate 7 in hand, compounds 9ak and 10as were smoothly synthesized through the direct palladium-catalyzed coupling reactions of substance 7 and the corresponding aromatic amine 8.

2.2. Biological Evaluation

The ability of the synthesized compounds to inhibit MET, c-Kit, and VEGFR-2 activities was evaluated using an enzyme assay with a recombinant kinase domain [29]. Based on the structure activity relationship (SAR) of MET inhibitors [30], we proposed that the removal of the key diaryl ether fragments in compound 3 would result in a loss of MET activity. As shown in Table 1, compounds 9ak exhibited no obvious MET inhibitory activity at 5000 nM, while our previously reported lead compound 3 exhibited excellent MET inhibitory activity (IC50 of 9.9 nM). Interestingly, compound 9g (n = 1, block A-6/4-pyridyl group) exhibited a moderate inhibitory activity against c-Kit (IC50 of 832.0 nM) that was only 2.5-fold less potent than that of compound 3 (IC50 of 329.6 nM). More importantly, 9k (n = 1, block A-9/4-quinolyl group) exhibited excellent c-Kit inhibitory activity (IC50 of 8.5 nM); 9k is 38.8-fold more potent than compound 3. Moreover, compounds 9c (n = 0, block A-3/2, 6-dichloro-phenyl group), 9g (block A-6), and 9k (block A-9) exhibited moderate VEGFR-2 inhibitory activity (IC50 values of 238.5–691.2 nM), which was comparable to compound 3 (IC50 of 279.9 nM).
To further study the structure activity relationship (SAR) of the 2-phenyl group and the R2 group, compounds 10as were screened for their inhibitory activities against MET, c-Kit, and VEGFR-2 (Table 2). Similar to compounds 9ak, compounds 10as showed no obvious MET inhibitory activity. Compound 10d (2-(4-fluoro)-phenyl group and n = 1, block A-9/4-quinolyl group) exhibited weak c-Kit inhibitory activity, while compounds 10l (2-(4-chloro)-phenyl group) and 10r (2-(4-trifluoromethyoxy)phenyl group) bearing the same block A-9 (4-quinolyl group) exhibited slightly stronger c-Kit inhibitory activity than compound 3 (IC50 of 329.6 nM). Interestingly, most compounds 10 bearing block A-6 (4-pyridyl group) or A-9 (4-quinolyl group) showed different degrees of inhibiting VEGFR-2. For examples, compounds 10d, 10k, and 10o exhibited comparable VEGFR-2 inhibitory activity (IC50 values of 208–538 nM) to compound 3 (IC50 of 279.9 nM). More importantly, compounds 10l and 10r exhibited excellent VEGFR-2 inhibitory activity (IC50 values of 31.7–56.5 nM)—i.e., they are 5.0–8.8-fold more potent than compound 3.

2.3. Molecular Modeling

Molecular docking experiments were further performed to determine the SAR [31,32,33,34,35,36,37]. As shown in Figure 3A–C and Table 3, the entire molecules of 9g, 9k, and 10r were favorably located in the c-Kit binding pocket. The main weak interactions between 9g and c-Kit included: (1) the H-bond interactions with residues Asp810 and Cys673; (2) the ion–π interaction with Lys623; and (3) the hydrophobic interaction with Leu799. The stronger H-bond interactions with Cys673 and additional hydrophobic interactions with residues Leu595 and Tyr672 of compound 10r rendered 10r five-times more potent than compound 9g. The further enhancement of key H-bond interactions with residues Asp810 and Cys673 resulted in the significantly improved c-Kit inhibitory activity of 9k.
As shown in Figure 3D–F and Table 3, compounds 9g, 9k, and 10r were entirely located in the VEGFR-2 binding pocket. The main weak interactions between compound 9g and VEGFR-2 included: (1) the H-bond interactions with residues Asp1046 and Cys919; (2) ion–π interaction with Lys868; (3) the hydrophobic interaction with Leu1035. The stronger H-bond interactions with residues Cys919 and additional hydrophobic interactions with residues Leu840 and Tyr918 rendered compound 9k more potent than 9g. The further enhancement of key H-bond interactions with residues Asp1046 and additional hydrophobic interaction with residues Ile892 and Leu1019 resulted in the significantly improved VEGFR-2 inhibitory activity of 10r.
Taking the molecular docking results into account, we hypothesize that the hydrogen bond acceptor (N containing heterocycle) and hydrophobic effects (fused aromatic ring) of 8-amino substituents are crucial to improve the inhibitory activity for c-Kit and VEGFR-2 kinase.

3. Conclusions

In summary, we described the design and synthesis of a series of 8-amino-substituted 2-phenyl-2,7-naphthyridin-1(2H)-one derivatives to broaden the application of this new 2,7-naphthyridone scaffold. Preliminary biological screening resulted in the discovery of new lead compounds 9k, 10l, and 10r, which exhibit more potent c-Kit and VEGFR-2 kinase inhibitory activity than the previously reported lead compound, 3. Molecular docking results provided further insight into the binding interactions of the new lead compounds with c-Kit and VEGFR-2 kinase. We identified 8-amino-substituted 2-phenyl-2,7-naphthyridin-1(2H)-one as a new lead scaffold of c-Kit and VEGFR-2 kinase inhibitors.

4. Materials and Methods

4.1. Biochemical Kinase Assays

The ability of compounds to inhibit the activity of three kinases (MET, c-Kit, and VEGFR-2) was tested in vitro [30]. Enzyme assays were run in homogeneous time-resolved fluorescence (HTRF) format in 384-well microtiter plates using purified kinases purchased from Invitrogen (Carlsbad, CA, US). The HTRF KinEASE TK kit (contains substrate-biotin, antibody-cryptate, streptavidin-XL665, 5×enzymatic buffer, and detection buffer) was purchased from Cisbio (Codolet, France), and the kinase assays were performed according to the manufacturer’s instructions. After the kinases and the compounds incubated at 25~30 °C for 5 min, the reactions were initiated by the addition of 2 μL of mixed substrate solution (mixed solution of ATP (Sigma, Shanghai, China) and substrate-biotin). The final concentrations of kinases were at EC80 and the total reaction volume was 8 μL. Plates were incubated at 30 °C for 30~60 min, then the reactions were quenched by the addition 8 μL mixed detection solution (mixed solution of antibody-cryptate and streptavidin-XL665 in detection buffer). The fluorescence excitation wavelength was 320 nM. The fluorescence at 665 nm (acceptor emission wavelength) and 620 nm (donor emission wavelength) was measured with a PHERAstar FS plate reader (BMG, LABTECH, Ortenberg, Germany) using a time delay of 50 μs. All kinase assays were conducted using ATP concentrations below the enzyme Kmapp and kinase-specific biotinylated substrate peptides.
The data for dose responses were plotted as percentage of inhibition calculated with the data reduction formula 100 × [1 − (U1C2)/(C1C2)] versus concentration of compound, where U is the emission ratio of 665 nm and 620 nm of test sample, C1 is the average value obtained for solvent control (2% DMSO), and C2 is the average value obtained for no reaction control (no kinase sample). Inhibition curves were generated by plotting percentage of control activity versus log10 of the concentration of each kinase. The IC50 values were calculated by nonlinear regression with Graphpad Prism 5 (GraphPad Software, San Diego, CA).

4.2. Molecular Modeling

The three-dimensional structures of the small molecules were constructed and primarily optimized by Sybyl 2.0 software. Steepest descent and conjugate gradient methods were used in the optimization process. Autodock Tools was used to assign Gasteiger charges for both the receptors and inhibitors. The optimized probes were docked into the crystal structures of c-Kit (PDB ID: 4U0I) and VEGFR-2 (PDB ID: 3EFL) with Autodock 4.2.5, 1, respectively. The gird size was set to be 70 × 70 × 70 and the grid point spacing was set at default value 0.375 Å. A total of 256 runs were performed by using Lamarkian genetic algorithm (LGA) for conformational search. The best poses were selected for the binding model analysis for all the inhibitors. The figures were prepared with PyMOL 2.2.3 [31,32,33,34,35,36,37].

4.3. Chemistry

4.3.1. General Information

Unless otherwise noted, all chemical reagents were commercially available and treated with standard methods. Silica gel column chromatography (CC). Silica gel (200–400 Mesh; Qingdao Makall Group Co., Ltd.; Qingdao; China). Solvents were dried in a routine way and redistilled. All reactions involving air- or moisture-sensitive reagents were performed under a nitrogen or argon atmosphere. 1HNMR spectra (400 MHz) and 13CNMR (100 MHz) spectra were recorded on a Bruker BioSpin AG (Ultrashield Plus AV 400, Fällanden, Switzerland) spectrometer as deuterochloroform (CDCl3) or dimethyl sulfoxide-d6 (DMSO-d6) solutions using tetramethylsilane (TMS) as an internal standard (δ = 0) unless noted otherwise. MS spectra were obtained on an Agilent technologies 6120 quadrupole LC/MS (ESI). All reactions were monitored using thin-layer chromatography (TLC) on silica gel plates. Yields were of purified compounds and were not optimized.

4.3.2. General Procedure for the Preparation of Intermediates 7af

The intermediates 7af were prepared according to our previous report [11].

4.3.3. General Procedure for the Preparation of Targets 9ak and 10as

An oven-dried Schlenk tube was charged with 7 (0.4 mmol), Pd2(dba)3 (0.02 mmol), xantphos (0.04 mmol), t-BuONa (0.8 mmol), and amine (0.48 mmol), and then purged with argon three times. Ultra-dry dioxane (15 mL) was added to the Schlenk tube with a syringe at argon atmosphere. The mixture was stirred at 110 °C overnight. After cooling to room temperature, the mixture was concentrated in vacuo and the residue purified by flash chromatography on silica gel using DCM/MeOH (100:1) as eluent to obtain 9ak and 10as.
2-(4-fluorophenyl)-8-(phenylamino)-2,7-naphthyridin-1(2H)-one (9a): Yellow solid (72% yield). HPLC purity: 98.3%. 1H NMR (400 MHz, DMSO-d6) δ: 11.79 (s, 1H), 8.30 (d, J = 5.3 Hz, 1H), 7.81 (m, 2H), 7.69 (d, J = 7.3 Hz, 1H), 7.61–7.31 (m, 6H), 7.02 (m, 1H), 6.95 (d, J = 5.3 Hz, 1H), 6.68 (d, J = 7.3 Hz, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 162.59, 160.32, 155.89, 150.22, 145.72, 139.81, 137.53, 136.66, 129.47, 128.74, 120.33, 119.81, 109.91, 116.00, 109.91, 105.36; ESI-MS m/z: 332.3 ([M + H]+).
8-((2,6-dimethylphenyl)amino)-2-(4-fluorophenyl)-2,7-naphthyridin-1(2H)-one (9b): Yellow solid (82% yield). 1H NMR (400 MHz, CDCl3) δ: 10.57 (s, 1H), 8.14 (d, J = 5.6 Hz, 1H), 7.44 (m, 2H), 7.22 (m, 2H); 7.24(d, J = 7.2 Hz, 1H), 7.10 (m, 3H), 6.56 (d, J = 5.6 Hz, 1H), 6.42 (d, J = 7.2 Hz, 1H), 2.23 (s, 6H); 13C NMR (100 MHz, DMSO-d6) δ: 162.76, 162.66, 160.22, 157.44, 150.50, 145.73, 137.35, 136.81, 135.23, 129.52, 127.72, 125.91, 116.05, 108.49, 104.96, 104.71, 18.40; ESI-MS m/z: 360.4 ([M + H]+).
8-((2,6-dichlorophenyl)amino)-2-(4-fluorophenyl)-2,7-naphthyridin-1(2H)-one (9c): Yellow solid (72% yield). HPLC purity: 95.7%. 1H NMR (400 MHz, CDCl3) δ: 10.84 (s, 1H), 8.19 (d, J = 5.6 Hz, 1H), 7.43–7.13 (m, 8H), 6.70 (d, J = 5.6 Hz, 1H), 6.46 (d, J = 7.2 Hz, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 162.72, 162.54, 160.28, 156.45, 150.44, 145.62, 137.66, 136.58, 134.56, 133.64, 129.55, 128.50, 116.09, 115.86, 110.07, 105.17, 104.93; ESI-MS m/z: 401.2 ([M + H]+).
2-(4-fluorophenyl)-8-(pyridin-3-ylamino)-2,7-naphthyridin-1(2H)-one (9d): Yellow solid (85% yield). HPLC purity: 92.1%. 1H NMR (400 MHz, DMSO-d6) δ: 11.81 (s, 1H), 8.89 (s, 1H), 8.37 (d, J = 8 Hz, 1H), 8.33 (d, J = 5.2 Hz, 1H), 8.23 (d, J = 3.6 Hz, 1H), 7.71 (d, J = 7.2 Hz, 1H), 7.61–7.58 (m, 2H), 7.44–7.35 (m, 3H), 7.03 (d, J = 5.2 Hz, 1H), 6.71 (d, J = 7.2 Hz, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 160.35, 155.77, 150.06, 145.76, 143.02, 141.50, 137.74, 136.58, 129.53, 129.44, 126.57, 123.52, 116.15, 115.92, 110.67, 105.85, 105.17; ESI-MS m/z: 333.3 ([M + H]+).
2-(4-fluorophenyl)-8-(isoquinolin-7-ylamino)-2,7-naphthyridin-1(2H)-one (9e): Yellow solid (85% yield). HPLC purity: 96.0%. 1H NMR (400 MHz, DMSO-d6) δ: 11.99 (s, 1H), 9.22 (s, 1H), 8.84 (s, 1H), 8.41 (m, 2H), 7.82 (dd, J1 = 8.8 Hz, J2 = 8.8 Hz, 2H), 7.56 (d, J = 5.2 Hz, 1H), 7.43–7.40 (m, 2H), 7.30 (d, J = 7.2 Hz, 1H), 7.28 (d, J = 8.8 Hz, 1H), 7.24 (d, J = 8.8 Hz, 1H), 6.80(d, J = 5.2 Hz, 1H), 6.50 (d, J = 7.2 Hz, 1H); 13C NMR (100 MHz, DMSO-d6) δ: 162.62, 155.68, 151.44, 150.13, 145.71, 141.72, 138.55, 137.74, 136.60, 131.27, 129.54, 129.19, 127.15, 125.54, 119.98, 116.14, 115.92, 113.83, 110.74, 105.95, 105.24; ESI-MS m/z: 383.3 ([M + H]+).
8-(benzylamino)-2-(4-fluorophenyl)-2,7-naphthyridin-1(2H)-one (9f): Yellow solid (87% yield). HPLC purity: 96.6%. 1H NMR(400 MHz, DMSO-d6) δ: 9.59 (t, J = 5.2 Hz, 1H), 8.16 (d, J = 5.2 Hz, 1H), 7.59 (d, J = 7.2 Hz, 1H), 7.54–7.51 (m, 2H), 7.38–7.25 (m, 7H), 6.70 (d, J = 5.2 Hz, 1H), 6.56 (d, J = 7.2 Hz, 1H), 4.70 (d, J = 5.2 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 162.39, 160.15, 158.40, 151.01, 145.66, 139.62, 137.23, 136.75, 129.33, 128.37, 127.33, 126.78, 115.79, 107.47, 104.85, 104.64, 43.91; ESI-MS m/z: 346.3 ([M + H]+).
2-(4-fluorophenyl)-8-((pyridin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (9g): Yellow solid (87% yield). HPLC purity: 99.8%. 1H NMR (400 MHz, CDCl3) δ: 9.71 (t, J = 5.6 Hz, 1H), 8.51 (d, J = 5.2 Hz, 1H), 8.16 (d, J = 5.2 Hz, 1H), 7.39 (d, J = 7.2 Hz, 1H), 7.37 (m, 4H), 7.28 (d, J = 5.2 Hz, 1H), 7.26–7.18 (m, 2H), 6.56 (d, J = 5.2 Hz, 1H), 6.40 (d, J = 7.2 Hz, 1H), 4.79 (d, J = 5.6 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 162.33, 160.14, 158.33, 150.83, 149.45, 149.14, 145.65, 137.33, 136.72, 129.43, 122.07, 115.98, 107.85, 104.80, 42.77; ESI-MS m/z: 347.3 ([M + H]+).
2-(4-fluorophenyl)-8-((pyridin-3-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (9h): Yellow solid (77% yield). HPLC purity: 99.8%. 1H NMR (400 MHz, DMSO-d6) δ: 9.69 (t, J = 5.6 Hz, 1H), 8.62 (s, 1H), 8.49 (m, 1H), 8.19 (d, J = 5.2 Hz, 1H), 7.79 (m, 1H), 7.63 (d, J = 7.2 Hz, 1H), 7.58–7.37 (m, 5H), 6.75 (d, J = 5.2 Hz, 1H), 6.60 (d, J = 7.2 Hz, 1H), 4.77 (d, J = 5.6 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 160.14, 158.29, 150.88, 148.89, 147.79, 145.66, 137.25, 136.71, 135.34, 135.12, 129.40, 128.79, 128.52, 123.46, 115.79, 107.75, 104.83, 41.42; ESI-MS m/z: 347.3 ([M + H]+).
2-(4-fluorophenyl)-8-((furan-2-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (9i): Yellow solid (79% yield). HPLC purity: 95.6%. 1H NMR (400 MHz, CDCl3) δ: 9.48 (t, J = 5.2 Hz, 1H), 8.22 (d, J = 5.2 Hz, 1H), 7.36 (d, J = 7.2 Hz, 1H), 7.34–7.32 (m, 2H), 7.21–7.17 (m, 3H), 6.54 (d, J = 5.2 Hz, 1H), 6.36 (d, J = 7.2 Hz, 1H), 6.27 (m, 2H), 4.74 (d, J = 5.2 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 162.59, 162.32, 160.15, 158.05, 152.40, 150.85, 145.62, 142.19, 137.27, 136.69, 129.40, 115.98, 110.43, 107.72, 106.84, 104.83, 104.74, 37.18; ESI-MS m/z: 336.3 ([M + H]+).
2-(4-fluorophenyl)-8-((naphthalen-1-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (9j): Yellow solid (81% yield). HPLC purity: 99.5%. 1H NMR(400 MHz, DMSO-d6) δ: 9.60 (t, J = 5.6 Hz, 1H), 8.19 (d, J = 5.2 Hz, 1H), 8.10 (m, 1H), 7.95 (m, 1H), 7.85 (m, 1H), 7.56 (d, J = 7.2 Hz, 1H), 7.55–7.43 (m, 6H), 7.31 (m, 2H), 6.71 (d, J = 5.2 Hz, 1H), 6.55 (d, J = 7.2 Hz, 1H), 5.16 (d, J = 5.6 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 145.74, 137.26, 136.67, 136.64, 134.76, 133.38, 131.03, 129.61, 129.37, 129.29, 129.00, 128.55, 127.59, 126.32, 125.81, 125.50, 125.44, 123.39, 115.95, 107.53, 104.88, 104.70, 41.88; ESI-MS m/z: 396.4 ([M + H]+).
2-(4-fluorophenyl)-8-((quinolin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (9k): Yellow solid (87% yield). HPLC purity: 99.8%. 1H NMR (400 MHz, CDCl3) δ: 9.72 (t, J = 5.6 Hz, 1H), 8.81 (d, J = 4.4 Hz, 1H), 8.19 (d, J = 5.2 Hz, 1H), 8.13–8.10 (m, 2H), 7.71 (t, J = 15.2 Hz, 1H), 7.57 (t, J = 15.2 Hz, 1H), 7.44 (d, J = 4.4 Hz, 1H), 7.38 (d, J = 7.2 Hz, 1H), 7.37–7.35(m, 1H), 7.24–7.17 (m, 3H), 6.58 (d, J = 5.2 Hz, 1H), 6.41 (d, J = 7.2 Hz, 1H), 5.29 (d, J = 5.6 Hz, 2H); 13C NMR (100 MHz, DMSO-d6) δ: 162.59, 162.40, 160.15, 158.37, 150.87, 150.31, 147.59, 145.72, 145.12, 137.34, 136.71, 129.61, 129.42, 126.60, 126.16, 123.51, 118.82, 115.98, 115.75, 107.89, 104.91, 40.71; ESI-MS m/z: 397.4 ([M + H]+).
2-(4-fluorophenyl)-3-methyl-8-(pyridin-3-ylamino)-2,7-naphthyridin-1(2H)-one (10a): Yellow solid (77% yield). HPLC purity: 94.4%. 1H NMR (400 MHz, DMSO-d6) δ: 11.72 (s, 1H), 8.87 (s, 1H), 8.36 (m, 1H), 8.25 (d, J = 5.6 Hz, 1H), 8.20 (m, 1H), 7.50–7.32 (m, 5H), 6.90 (d, J = 5.6 Hz, 1H), 6.64 (s, 1H), 1.98 (s, 3H); ESI-MS m/z: 347.3 ([M + H]+).
2-(4-fluorophenyl)-3-methyl-8-((pyridin-3-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10b): Yellow solid (74% yield). HPLC purity: 96.3%. 1H NMR (400 MHz, DMSO-d6) δ: 9.53 (t, J = 5.6 Hz, 1H), 8.55 (s, 1H), 8.43 (m, 1H), 8.08 (d, J = 5.2 Hz, 1H), 7.72 (m, 1H), 7.40–7.30 (m, 5H), 6.58 (d, J = 5.2 Hz, 1H), 6.49 (s, 1H), 4.68 (d, J = 5.6 Hz, 2H), 1.92 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 162.89, 160.45, 158.09, 150.67, 148.91, 147.96, 145.19, 144.84, 135.39, 135.11, 134.43, 130.85, 130.76, 123.44, 116.33, 107.12, 104.31, 103.39, 21.18; ESI-MS m/z: 361.4 ([M + H]+).
2-(4-fluorophenyl)-3-methyl-8-((pyridin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10c): Yellow solid (77% yield). HPLC purity: 99.8%. 1H NMR (400 MHz, DMSO-d6) δ: 9.59 (t, J = 5.6 Hz, 1H), 8.46 (d, J = 5.0 Hz, 2H), 8.04 (d, J = 5.6 Hz, 1H), 7.45–7.35 (m, 4H), 7.28 (d, J = 5.0 Hz, 2H), 6.59 (d, J = 5.6 Hz, 1H), 6.49 (s, 1H), 4.70 (d, J = 5.6 Hz, 2H), 1.94 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 160.47, 158.17, 150.63, 149.45, 149.16, 145.20, 144.89, 134.45, 130.88, 130.79, 122.11, 116.34, 107.23, 104.31, 103.44, 42.73, 21.20; ESI-MS m/z: 361.4 ([M + H]+).
2-(4-fluorophenyl)-3-methyl-8-((quinolin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10d): Yellow solid (78% yield). HPLC purity: 95.9%. 1H NMR (400 MHz, DMSO-d6) δ: 9.64 (t, J = 5.6 Hz, 1H), 8.79 (d, J = 4.0 Hz, 1H), 8.21 (m, 1H), 8.06 (d, J = 4.0 Hz, 1H), 8.03 (m, 1H), 7.77 (t, J = 15.2 Hz, 1H), 7.64 (t, J = 15.2 Hz, 1H), 7.44 (d, J = 5.2 Hz, 1H), 7.42–7.34 (m, 4H), 6.60 (d, J = 5.2 Hz, 1H), 6.50 (s, 1H), 5.21 (d, J = 5.6 Hz, 2H), 1.94 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ: 163.60, 162.91, 160.47, 158.14, 150.61, 147.58, 145.27, 144.98, 134.42, 130.86, 129.23, 126.61, 126.17, 124.20, 123.53, 118.90, 118.38, 116.35, 107.27, 104.35, 103.54, 40.64, 21.21; ESI-MS m/z: 411.4 ([M + H]+).
3-methyl-2-phenyl-8-(pyridin-3-ylamino)-2,7-naphthyridin-1(2H)-one (10e): Yellow solid (80% yield). HPLC purity: 90.1%. 1H NMR (400 MHz, DMSO-d6) δ: 11.76 (s, 1H), 8.86 (s, 1H), 8.36 (m, 1H), 8.25 (d, J = 5.2 Hz, 1H), 8.19 (m, 1H), 7.64–7.50 (m, 4H), 7.39 (m, 1H), 7.34 (m, 1H), 6.90 (d, J = 5.2 Hz, 1H), 6.63 (s, 1H), 1.98 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 164.14, 158.29, 150.68, 145.37, 142.82, 141.34, 138.04, 136.64, 132.19, 131.50, 129.56, 128.67, 126.38, 123.48, 110.01, 104.71, 104.47, 21.23; ESI-MS m/z: 329.3 ([M + H]+).
3-methyl-2-phenyl-8-((pyridin-3-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10f): Yellow solid (78% yield). HPLC purity: 97.9%. 1H NMR (400 MHz, DMSO-d6) δ: 9.55 (t, J = 4.8 Hz, 1H), 8.55 (m, 1H), 8.43 (m, 1H), 8.08 (d, J = 4.4 Hz, 1H), 7.72–7.31 (m, 7H), 6.59 (d, J = 4.4 Hz, 1H), 6.48 (s, 1H), 4.68 (d, J = 4.8 Hz, 2H), 1.92 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.44, 158.12, 150.60, 148.91, 147.97, 145.17, 144.80, 138.23, 135.38, 135.10, 129.39, 128.55, 123.45, 107.11, 104.24, 103.45, 41.35, 21.18; ESI-MS m/z: 343.4 ([M + H]+).
3-methyl-2-phenyl-8-((pyridin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10g): Yellow solid (67% yield). HPLC purity: 97.7%. 1H NMR (400 MHz, DMSO-d6) δ: 9.60 (t, J = 5.6 Hz, 1H), 8.46 (d, J = 5.0 Hz, 2H), 8.04 (d, J = 5.6 Hz, 1H), 7.56–7.46 (m, 3H), 7.34 (m, 2H), 7.28 (d, J = 5.0 Hz, 2H), 6.59 (d, J = 5.6 Hz, 1H), 6.49 (s, 1H), 4.70 (d, J = 5.6 Hz, 2H), 1.93 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.47, 158.19, 150.55, 149.44, 149.17, 145.18, 144.85, 138.24, 129.40, 128.58, 126.56, 122.15, 107.23, 104.23, 103.51, 42.74, 21.19; ESI-MS m/z: 343.4 ([M + H]+).
3-methyl-2-phenyl-8-((quinolin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10h): Yellow solid (88% yield). 1H NMR (400 MHz, DMSO-d6) δ: 9.66 (t, J = 5.6 Hz, 1H), 8.79 (d, J = 4.0 Hz, 1H), 8.21 (m, 1H), 8.06 (d, J = 4.0 Hz, 1H), 8.03 (d, J = 5.2 Hz, 1H), 7.77 (t, J = 15.2 Hz, 1H), 7.64 (t, J = 15.2 Hz, 1H), 7.56–7.33 (m, 6H), 6.60 (d, J = 5.2 Hz, 1H), 6.51 (s, 1H), 5.20 (d, J = 5.6 Hz, 2H), 1.94 (s, 3H); ESI-MS m/z: 393.4 ([M + H]+).
2-(4-chlorophenyl)-3-methyl-8-(pyridin-3-ylamino)-2,7-naphthyridin-1(2H)-one (10i): Yellow solid (86% yield). HPLC purity: 91.1%. 1H NMR (400 MHz, DMSO-d6) δ: 11.70 (s, 1H), 8.86 (s, 1H), 8.36 (m, 1H), 8.25 (d, J = 5.2 Hz, 1H), 8.19 (m, 1H), 7.82–7.46 (m, 4H), 7.39 (m, 1H), 6.90 (d, J = 5.2 Hz, 1H), 6.65 (s, 1H), 1.98 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ:163.14, 155.29, 149.89, 145.37, 142.82, 141.34, 138.04, 136.64, 132.19, 131.50, 129.56, 128.67, 126.38, 123.48, 110.01, 104.71, 104.47, 21.23; ESI-MS m/z: 363.8 ([M + H]+).
2-(4-chlorophenyl)-3-methyl-8-((pyridin-3-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10j): Yellow solid (74% yield). HPLC purity: 92.1%. 1H NMR (400 MHz, DMSO-d6) δ: 9.50 (t, J = 5.6 Hz, 1H), 8.55 (s, 1H), 8.43 (d, J = 4.4 Hz, 1H), 8.08 (d, J = 5.2 Hz, 1H), 7.71 (d, J = 7.6 Hz, 1H), 7.61 (d, J = 8.4 Hz, 2H), 7.39 (d, J = 8.4 Hz, 2H), 7.32 (dd, J1 = 4.4 Hz, J2 = 7.6 Hz, 1H), 6.58 (d, J = 5.2 Hz, 1H), 6.49 (s, 1H), 4.68 (d, J = 5.6 Hz, 2H), 1.92 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.40, 158.08, 150.73, 148.88, 147.97, 145.21, 144.59, 137.09, 135.37, 135.14, 133.20, 130.63, 129.43, 123.47, 107.17, 104.42, 103.34, 41.35, 21.13; ESI-MS m/z: 377.8 ([M + H]+).
2-(4-chlorophenyl)-3-methyl-8-((pyridin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10k): Yellow solid (77% yield). HPLC purity: 99.8%. 1H NMR (400 MHz, DMSO-d6) δ: 9.59 (t, J = 5.6 Hz, 1H), 8.46 (d, J = 4.0 Hz, 2H), 8.04 (d, J = 5.6 Hz, 1H), 7.61 (d, J = 8.4 Hz, 2H), 7.41 (d, J = 8.4 Hz, 2H), 7.28 (d, J = 5.0 Hz, 2H), 6.59 (d, J = 5.6 Hz, 1H), 6.49 (s, 1H), 4.70 (d, J = 5.6 Hz, 2H), 1.94 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.42, 158.15, 150.68, 149.44, 149.15, 145.21, 144.63, 137.11, 133.21, 130.66, 129.43, 122.13, 107.27, 104.41, 103.40, 42.74, 21.15; ESI-MS m/z: 377.8 ([M + H]+).
2-(4-chlorophenyl)-3-methyl-8-((quinolin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10l): Yellow solid (84% yield). HPLC purity: 98.0%. 1H NMR (400 MHz, DMSO-d6) δ: 9.62 (t, J = 5.6 Hz, 1H), 8.79 (d, J = 4.0 Hz, 1H), 8.21 (m, 1H), 8.06 (d, J = 5.2 Hz, 1H), 8.03 (m, 1H), 7.77 (t, J = 15.2 Hz, 1H), 7.64 (t, J = 15.2 Hz, 1H), 7.61 (d, J = 8.4 Hz, 2H), 7.40 (d, J = 8.4 Hz, 2H), 7.37 (d, J = 4.0 Hz, 1H), 6.60 (d, J = 5.2 Hz, 1H), 6.51 (s, 1H), 5.21 (d, J = 5.6 Hz, 2H), 1.94 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 162.91, 158.14, 150.73, 150.31, 147.60, 145.28, 145.15, 144.69, 137.10, 133.22, 130.65, 129.61, 129.44, 129.23, 126.62, 126.17, 123.52, 118.91, 107.30, 104.44, 103.49, 40.63, 21.16; ESI-MS m/z: 427.9 ([M + H]+).
3-methyl-8-(pyridin-3-ylamino)-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10m): Yellow solid (79% yield). HPLC purity: 99.1%. 1H NMR (400 MHz, DMSO-d6) δ: 11.67 (s, 1H), 8.87 (s, 1H), 8.36 (d, J = 8.0 Hz, 1H), 8.25 (d, J = 5.6 Hz, 1H), 8.20 (d, J = 4.0 Hz, 1H), 7.59 (s, 4H), 7.34 (dd, J1 = 8.0 Hz, J2 = 4.0 Hz, 1H), 6.90 (d, J = 5.6 Hz, 1H), 6.65 (s, 1H), 1.98 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.62, 155.54, 149.94, 148.24, 145.29, 142.88, 141.38, 136.99, 136.58, 130.83, 126.45, 123.49, 122.14, 121.31, 118.76, 110.05, 104.91, 104.38, 21.20; ESI-MS m/z: 413.3 ([M + H]+).
3-methyl-8-((pyridin-3-ylmethyl)amino)-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10n): Yellow solid (74% yield). 1H NMR (400 MHz, DMSO-d6) δ: 9.50 (t, J = 5.6 Hz, 1H), 8.55 (s, 1H), 8.43 (d, J = 4.0 Hz, 1H), 8.08 (d, J = 5.2 Hz, 1H), 7.72 (d, J = 7.6 Hz, 1H), 7.59 (s, 4H), 7.32 (dd, J1 = 4.0 Hz, J2 = 7.6 Hz, 1H), 6.58 (d, J = 5.2 Hz, 1H), 6.50 (s, 1H), 4.68 (d, J = 5.6 Hz, 2H), 1.93 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.43, 158.08, 150.78, 148.92, 148.06, 147.98, 145.22, 144.52, 137.21, 135.36, 130.89, 123.44, 121.97, 121, 30, 107.14, 104.43, 103.33, 41.34, 21.14; ESI-MS m/z: 427.4 ([M + H]+).
3-methyl-8-((pyridin-4-ylmethyl)amino)-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10o): Yellow solid (79% yield). HPLC purity: 99.4%. 1H NMR (400 MHz, DMSO-d6) δ: 9.56 (t, J = 5.6 Hz, 1H), 8.46 (d, J = 5.2 Hz, 2H), 8.04 (d, J = 5.2 Hz, 1H), 7.54 (s, 4H), 7.28 (d, J = 5.2 Hz, 2H), 6.59 (d, J = 5.2 Hz, 1H), 6.50 (s, 1H), 4.70 (d, J = 5.6 Hz, 2H), 1.95 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.18, 158.36, 150.71, 149.39, 148.08, 145.50, 144.58, 137.21, 130.97, 123.85, 122.13, 121.30, 118.75, 107.27, 104.44, 103.38, 42.74, 21.16; ESI-MS m/z: 427.4 ([M + H]+).
8-((furan-2-ylmethyl)amino)-3-methyl-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10p): Yellow solid (82% yield). HPLC purity: 96.3%. 1H NMR (400 MHz, DMSO-d6) δ: 9.32 (t, J = 5.6 Hz, 1H), 8.12 (d, J = 5.6 Hz, 1H), 7.55–7.49 (m, 5H), 6.60 (d, J = 5.6 Hz, 1H), 6.50 (s, 1H), 6.37 (m, 1H), 6.28 (m, 1H), 4.65 (d, J = 5.6 Hz, 2H), 1.93 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.43, 157.84, 152.41, 150.76, 148.07, 145.20, 144.53, 142.19, 137.18, 130.87, 121.98, 110.42, 107.13, 106.85, 104.48, 103.31, 66.33, 37.10, 21.15; ESI-MS m/z: 416.3 ([M + H]+).
3-methyl-8-((naphthalen-1-ylmethyl)amino)-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10q): Yellow solid (79% yield). HPLC purity: 97.0%. 1H NMR (400 MHz, DMSO-d6) δ: 9.43 (t, J = 5.6 Hz, 1H), 8.15 (d, J = 5.2 Hz, 1H), 8.10 (d, J = 8.4 Hz, 1H), 7.93 (dd, J1 = 8.4 Hz, J2 = 8.0 Hz, 1H), 7.84 (d, J = 8.0 Hz, 1H), 7.54–7.42 (m, 8H), 6.60 (d, J = 5.2 Hz, 1H), 6.50 (s, 1H), 5.13 (d, J = 5.6 Hz, 2H), 1.91 (s, 3H);13C NMR(100 MHz, DMSO-d6)δ: 158.06, 150.94, 148.01, 145.29, 144.50, 137.14, 134.82, 133.37, 131.03, 130.83, 128.55, 127.59, 126.32, 125.80, 125.50, 123.39, 121.93, 121.26, 118.71, 106.93, 104.52, 103.24, 41.76, 21.14; ESI-MS m/z: 476.4 ([M + H]+).
3-methyl-8-((quinolin-4-ylmethyl)amino)-2-(4-(trifluoromethoxy)phenyl)-2,7-naphthyridin-1(2H)-one (10r): Yellow solid (80% yield). HPLC purity: 94.0%. 1H NMR (400 MHz, DMSO-d6) δ: 9.62 (t, J = 5.6 Hz, 1H), 8.81 (d, J = 4.0 Hz, 1H), 8.21 (m, 1H), 8.06 (d, J = 5.2 Hz, 1H), 8.05 (m, 1H), 7.77 (t, J = 15.2 Hz, 1H), 7.64 (t, J = 15.2 Hz, 1H), 7.55 (s, 4H), 7.37 (d, J = 4.0 Hz, 1H), 6.63 (d, J = 5.2 Hz, 1H), 6.54 (s, 1H), 5.22 (d, J = 5.6 Hz, 2H), 1.96 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.12, 158.66, 151.28, 150.80, 148.12, 145.80, 145.64, 145.12, 137.69, 131.40, 130.12, 129.71, 127.10, 126.68, 124.02, 122.46, 119.43, 107.80, 104.98, 103.99, 41.12, 21.66; ESI-MS m/z: 477.4 ([M + H]+).
2-(2,4-difluorophenyl)-3-methyl-8-((quinolin-4-ylmethyl)amino)-2,7-naphthyridin-1(2H)-one (10s): Yellow solid (88% yield). HPLC purity: 99.1%. 1H NMR (400 MHz, DMSO-d6) δ: 9.54 (t, J = 5.6 Hz, 1H), 8.80 (d, J = 4.4 Hz, 1H), 8.21 (d, J = 8.0 Hz, 1H), 8.09 (d, J = 5.6 Hz, 1H), 8.05 (d, J = 8.0 Hz, 1H), 7.77 (t, J = 15.2 Hz, 1H), 7.64 (t, J = 15.2 Hz, 1H), 7.61 (s, 1H), 7.55 (t, J = 14.8 Hz, 1H), 7.36 (d, J = 4.4 Hz, 1H), 7.29 (t, J = 14.8 Hz, 1H), 6.63 (d, J = 5.6 Hz, 1H), 6.56 (s, 1H), 5.21 (d, J = 5.6 Hz, 2H), 1.99 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ: 163.02, 158.10, 151.19, 150.32, 147.59, 145.41, 145.09, 144.56, 132.19, 132.09, 129.60, 129.23, 126.61, 126.16, 123.51, 122.04, 118.88, 122.57, 107.41, 105.31, 105.04, 104.80, 103.05, 40.68, 20.51; ESI-MS m/z: 429.4 ([M + H]+).

Author Contributions

H.S., L.Z., H.D., W.H. and N.S. conceived and designed the experiments; L.Z. and H.D., performed synthesis; H.S. performed biological work and molecular modelling; W.H. and N.S. analyzed the data; W.H. and N.S. wrote the paper. W.H. were responsible for the correspondence of the manuscript. All authors discussed, edited and approved the final version.

Funding

This work was supported by the National Natural Science Foundation of China (No. 21272086), and the Fundamental Research Funds for the Central Universities (No. CCNU19QN073 & CCNU18TS009).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

MET: mesenchymal−epithelial transition factor; c-Kit, receptor tyrosine kinase for the stem cell factor; VEGFR-2, vascular endothelial growth factor receptor 2; PI3Kδ, phosphatidylinositol 3-kinase delta; CK2, protein kinase CK2; Akt1/2, protein kinase B1/2; Tpl-2, Tumor Progression Loci-2; Pdk-1, phosphoinositide-dependent protein kinase-1; PDE-5, phosphodiesterase type 5; TNFα, tumor necrosis factor alpha; SYK, spleen tyrosine kinase; DMEDA, N,N’-Dimethyl-1,2-ethanediamine; DMF-DMA, N,N-dimethylformamide dimethyl acetal; DMF, N,N-dimethylformamide; ATP, adenosine triphosphate; DMSO, dimethyl sulfoxide; HNMR, hydrogen nuclear magnetic resonance; CNMR, carbon nuclear magnetic resonance; MS, mass spectrum; LC/MS (ESI), liquid chromatography-mass spectrometry (Electrospray ionization).

References

  1. Greiner, R.; Ziegler, D.S.; Cibu, D.; Jakowetz, A.C.; Auras, F.; Bein, T.; Knochel, P. Preparation of polyfunctional naphthyridines by cobalt-catalyzed cross-couplings of halogenated naphthyridines with magnesium and zinc organometallics. Org. Lett. 2017, 19, 6384–6387. [Google Scholar] [CrossRef] [PubMed]
  2. Brown, D.J.; Ellman, J.A.; Taylor, E.C. The Naphthyridines; John Wiley & Sons: Hoboken, NJ, USA, 2007. [Google Scholar]
  3. Litvinov, V.P.; Roman, S.V.; Dyachenko, V.D. Naphthyridines. Structure, physicochemical properties and general methods of synthesis. Russ. Chem. Rev. 2000, 69, 201–220. [Google Scholar]
  4. Paim, C.S.; Araujo, B.V.; Volpato, N.M.; Steppe, M.; Schapoval, E.E.S. Gemifloxacin mesylate (GFM): Dissolution test based on in vivo data. Drug Dev. Ind. Pharm. 2015, 41, 567–572. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, T.C.; Hsu, Y.L.; Tsai, Y.C.; Chang, Y.W.; Kuo, P.L.; Chen, Y.H. Gemifloxacin inhibits migration and invasion and induces mesenchymal–epithelial transition in human breast adenocarcinoma cells. J. Mol. Med. 2014, 92, 53–64. [Google Scholar] [CrossRef] [PubMed]
  6. Norman, P. Novel 1, 5-naphthyridine PI3Kδ inhibitors, an evaluation of WO2011075628. Expert Opin. Ther. Pat. 2011, 21, 1805–1810. [Google Scholar] [CrossRef] [PubMed]
  7. Pierre, F.; Chua, P.C.; O’Brien, S.E.; Siddiqui, J.A.; Bourbon, P.; Haddach, M.; Michaux, J.; Nagasawa, J.; Schwaebe, M.K.; Stefan, E.; et al. Discovery and sar of 5-(3-chlorophenylamino)benzo[c][2,6]naphthyridine-8-carboxylic acid (CX-4945), the first clinical stage inhibitor of protein kinase ck2 for the treatment of cancer. J. Med. Chem. 2011, 54, 635–654. [Google Scholar] [CrossRef]
  8. Bilodeau, M.T.; Balitza, A.E.; Hoffman, J.M.; Manley, P.J.; Barnett, S.F.; Defeo, J.D.; Haskell, K.; Jones, R.E.; Leander, K.; Robinson, R.G.; et al. Allosteric inhibitors of Akt1 and Akt2: A naphthyridinone with efficacy in an A2780 tumor xenograft model. Bioorg. Med. Chem. Lett. 2008, 18, 3178–3182. [Google Scholar] [CrossRef]
  9. Kaila, N.; Green, N.; Li, H.Q.; Hu, Y.; Janz, K.; Gavrin, L.K.; Thomason, J.; Tam, S.; Powell, D.; Cuozzo, J.; et al. Identification of a novel class of selective Tpl2 kinase inhibitors: 4-Alkylamino-[1, 7] naphthyridine-3-carbonitriles. Bioorg. Med. Chem. 2007, 15, 6425–6442. [Google Scholar] [CrossRef]
  10. Gavrin, L.K.; Green, N.; Hu, Y.; Janz, K.; Kaila, N.; Li, H.Q.; Tam, S.Y.; Thomason, J.R.; Gopalsamy, A.; Ciszewski, G.; et al. Inhibition of Tpl2 kinase and TNF-α production with 1, 7-naphthyridine-3-carbonitriles: Synthesis and structure–activity relationships. Bioorg. Med. Chem. Lett. 2005, 15, 5288–5292. [Google Scholar] [CrossRef]
  11. Zhuo, L.S.; Xu, H.C.; Wang, M.S.; Zhao, X.E.; Ming, Z.H.; Zhu, X.L.; Huang, W.; Yang, G.F. 2, 7-naphthyridinone-based MET kinase inhibitors: A promising novel scaffold for antitumor drug development. Eur. J. Med. Chem. 2019, 178, 705–714. [Google Scholar]
  12. Wang, Y.; Xu, Z.L.; Ai, J.; Peng, X.; Lin, J.P.; Ji, Y.C.; Geng, M.Y.; Long, Y.Q. Investigation on the 1, 6-naphthyridine motif: Discovery and SAR study of 1 H-imidazo [4, 5-h][1, 6] naphthyridin-2 (3 H)-one-based c-Met kinase inhibitors. Organic Biomol. Chem. 2013, 11, 1545–1562. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, M.S.; Zhuo, L.S.; Yang, F.P.; Wang, W.J.; Huang, W.; Yang, G.F. Synthesis and biological evaluation of new MET inhibitors with 1, 6-naphthyridinone scaffold. Eur. J. Med. Chem. 2019, 185, 111803. [Google Scholar] [CrossRef] [PubMed]
  14. Gibson, S.A.; Benveniste, E.N. Protein kinase CK2: An emerging regulator of immunity. Trends Immunol. 2018, 39, 82–85. [Google Scholar] [CrossRef] [PubMed]
  15. Madak, J.T.; Cuthbertson, C.R.; Miyata, Y.; Tamura, S.; Petrunak, E.M.; Stuckey, J.A.; Han, Y.Y.; He, M.; Sun, D.X.; Showalter, H.D.; et al. Design, synthesis, and biological evaluation of 4-quinoline carboxylic acids as inhibitors of dihydroorotate dehydrogenase. J. Med. Chem. 2018, 61, 5162–5186. [Google Scholar] [CrossRef]
  16. Kumar, V.; Jaggi, M.; Singh, A.T.; Madaan, A.; Sanna, V.; Singh, P.; Sharma, P.K.; Irchhaiya, R.; Burman, A.C. 1, 8-Naphthyridine-3-carboxamide derivatives with anticancer and anti-inflammatory activity. Eur. J. Med. Chem. 2009, 44, 3356–3362. [Google Scholar] [CrossRef]
  17. Fu, L.; Feng, X.; Wang, J.J.; Xun, Z.; Hu, J.D.; Zhang, J.J.; Zhao, Y.W.; Huang, Z.B.; Shi, D.Q. Efficient synthesis and evaluation of antitumor activities of novel functionalized 1,8-naphthyridine derivatives. ACS Comb. Sci. 2015, 17, 24–31. [Google Scholar] [CrossRef]
  18. Manera, C.; Malfitano, A.M.; Parkkari, T.; Lucchesi, V.; Carpi, S.; Fogli, S.; Bertini, S.; Laezza, C.; Ligresti, A.; Saccomanni, G.; et al. New quinolone-and 1, 8-naphthyridine-3-carboxamides as selective CB2 receptor agonists with anticancer and immuno–modulatory activity. Eur. J. Med. Chem. 2015, 97, 10–18. [Google Scholar] [CrossRef]
  19. Gross, H.; Goeger, D.E.; Hills, P.; Mooberry, S.L.; Ballantine, D.L.; Murray, T.F.; Valeriote, F.A.; Gerwick, W.H. Lophocladines, Bioactive Alkaloids from the Red Alga Lophocladia sp. J. Nat. Prod. 2006, 69, 640–644. [Google Scholar] [CrossRef] [Green Version]
  20. Kumpan, K.; Nathubhai, A.; Zhang, C.; Wood, P.J.; Lloyd, M.D.; Thompson, H.T.; Lehtio, L.; Threadgill, M.D. Structure-based design, synthesis and evaluation in vitro of arylnaphthyridinones, arylpyridopyrimidinones and their tetrahydro derivatives as inhibitors of the tankyrases. Bioorg. Med. Chem. 2015, 23, 3013–3032. [Google Scholar] [CrossRef] [Green Version]
  21. Theeramunkong, S.; Vajragupta, O.; Mudjupa, C. Synthesis and biological evaluation of simplified analogs of lophocladine B as potential antitumor agents. Med. Chem. Res. 2016, 25, 2959–2964. [Google Scholar] [CrossRef]
  22. Zhang, A.; Ding, C.; Cheng, C.; Yao, Q. Convenient synthesis of 2, 7-naphthyridine lophocladines A and B and their analogues. J. Comb. Chem. 2007, 9, 916–919. [Google Scholar] [CrossRef]
  23. Gopalsamy, A.; Shi, M.; Boschelli, D.H.; Williamson, R.; Olland, A.; Hu, Y.; Krishnamurthy, G.; Han, X.; Arndt, K.; Guo, B. Discovery of dibenzo[c,f][2,7]naphthyridines as potent and selective 3-phosphoinositide-dependent kinase-1 inhibitors. J. Med. Chem. 2007, 50, 5547–5549. [Google Scholar] [CrossRef]
  24. Zhang, Y.; Illarionov, B.; Bacher, A.; Fischer, M.; Georg, G.I.; Ye, Q.Z.; Vander, V.D.; Fanwick, P.E.; Song, Y.; Cushman, M. A novel lumazine synthase inhibitor derived from oxidation of 1, 3, 6, 8-tetrahydroxy-2, 7-naphthyridine to a tetraazaperylenehexaone derivative. J. Org. Chem. 2007, 72, 2769–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ukita, T.; Nakamura, Y.; Kubo, A.; Yamamoto, Y.; Moritani, Y.; Saruta, K.; Higashijima, T.; Kotera, J.; Fujishige, K.; Takagi, M.; et al. 1, 7-and 2, 7-naphthyridine derivatives as potent and highly specific PDE5 inhibitors. Bioorg. Med. Chem. Lett. 2003, 13, 2341–2345. [Google Scholar] [CrossRef]
  26. Kim, H.O.; Ryu, J.H.; Jung, J.Y.; Lee, N.K.; Kim, J.H.; Kim, E.J.; Joen, S.D.; Kim, J.H.; Rhee, H.I.; Cho, Y.B.; et al. Pyridine Derivatives, Methods of their Preparations, and Pharmaceutical Compositions Containing the Same. PTC Int. Appl. WO 2006112666A1, 26 October 2006. [Google Scholar]
  27. Okram, B.; Uno, T.; Ding, Q.; Liu, Y.H.; Jin, Y.H.; Jin, Q.H.; Wu, X.; Chen, J.W.; Yan, S.F. Compounds and Compositions as Kinase Inhibitors. PTC Int. Appl. WO 2009097287A1, 6 August 2009. [Google Scholar]
  28. Ziegler, D.S.; Greiner, R.; Lumpe, H.; Kqiku, L.; Karaghiosoff, K.; Knochel, P. Directed zincation or magnesiation of the 2-pyridone and 2, 7-naphthyridone scaffold using TMP bases. Org. Lett. 2017, 19, 5760–5763. [Google Scholar] [CrossRef] [PubMed]
  29. Kumar, R.; Knick, V.B.; Rudolph, S.K. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol. Cancer Ther. 2007, 6, 2012–2021. [Google Scholar] [CrossRef] [Green Version]
  30. Cui, J.J. Targeting receptor tyrosine kinase MET in cancer: Small molecule inhibitors and clinical progress. J. Med. Chem. 2014, 57, 4427–4453. [Google Scholar] [CrossRef]
  31. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [Green Version]
  32. Gasteiger, J.; Marsili, M. Iterative partial equalization of orbital electronegativity—A rapid access to atomic charges. Tetrahedron 1980, 36, 3219–3228. [Google Scholar] [CrossRef]
  33. Morris, G.M.; Goodsell, D.S.; Halliday, R.S.; Huey, R.; Hart, W.E.; Belew, R.K.; Olson, A.J. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J. Comput. Chem. 1998, 19, 1639–1662. [Google Scholar] [CrossRef] [Green Version]
  34. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The protein data bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Garner, A.P.; Gozgit, J.M.; Anjum, R.; Vodala, S.; Schrock, A.; Zhou, T.; Serrano, C.; Eilers, G.; Zhu, M.; Ketzer, J.; et al. Ponatinib inhibits polyclonal drug-resistant KIT oncoproteins and shows therapeutic potential in heavily pretreated gastrointestinal stromal tumor (GIST) patients. Clin. Cancer Res. 2014, 20, 5745–5755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Tasker, A.S.; Patel, V.F. PDB bank [3EFL]. to be published.
  37. The PyMOL Molecular Graphics System, Version 1.2r3pre, Schrödinger, LLC. Available online: https://pymol.org/2/ (accessed on 5 December 2019).
Sample Availability: Not available.
Figure 1. Examples of biologically active compounds containing a naphthyridine scaffold.
Figure 1. Examples of biologically active compounds containing a naphthyridine scaffold.
Molecules 24 04461 g001
Figure 2. The design of 8-amino-2-phenyl-2,7-naphthyridones.
Figure 2. The design of 8-amino-2-phenyl-2,7-naphthyridones.
Molecules 24 04461 g002
Scheme 1. Synthesis of target compounds 9ak and 10as. Reaction conditions and reagents: (i) 1-fluoro-4-iodobenzene, CuI, DMEDA, K3PO4, dioxane, 100 °C; (ii) 2,4-pentanedione, piperidine, EtOH, 90 °C; (iii) DMF-DMA, DMF, 90 °C; (iv) H2SO4, 110 °C; (v) POCl3, 110 °C; (vi) Pd2(dba)3, dppp, t-BuONa, dioxane, 100 °C.
Scheme 1. Synthesis of target compounds 9ak and 10as. Reaction conditions and reagents: (i) 1-fluoro-4-iodobenzene, CuI, DMEDA, K3PO4, dioxane, 100 °C; (ii) 2,4-pentanedione, piperidine, EtOH, 90 °C; (iii) DMF-DMA, DMF, 90 °C; (iv) H2SO4, 110 °C; (v) POCl3, 110 °C; (vi) Pd2(dba)3, dppp, t-BuONa, dioxane, 100 °C.
Molecules 24 04461 sch001
Figure 3. The proposed binding mode of: (A) 9g with c-Kit (green); (B) 9k with c-Kit (cyan); (C) 10r with c-Kit (magenta); (D) 9g with VEGFR-2 (green); (E) 9k with VEGFR-2 (cyan); (F) 10r with VEGFR-2 (magenta). The red dashed line represents a hydrogen bond. For clarity, only key residues are shown.
Figure 3. The proposed binding mode of: (A) 9g with c-Kit (green); (B) 9k with c-Kit (cyan); (C) 10r with c-Kit (magenta); (D) 9g with VEGFR-2 (green); (E) 9k with VEGFR-2 (cyan); (F) 10r with VEGFR-2 (magenta). The red dashed line represents a hydrogen bond. For clarity, only key residues are shown.
Molecules 24 04461 g003
Table 1. Inhibitory activity of 9a–k against MET, c-Kit, and VEGFR-2.
Table 1. Inhibitory activity of 9a–k against MET, c-Kit, and VEGFR-2.
Molecules 24 04461 i001
No.Block AnInhibitory Activity, IC50, nM a
METc-KitVEGFR-2
9aA-10>5000>5000>5000
9bA-20>5000>5000>5000
9cA-30>5000>5000691.2
9dA-40>5000>5000>5000
9eA-50>5000>5000>5000
9fA-11>5000>5000>5000
9gA-61>5000832.0601.3
9hA-41>5000>5000>5000
9iA-71>5000>5000>5000
9jA-81>5000>5000>5000
9kA-91>50008.5238.5
39.9329.6279.9
a In vitro kinase assays were performed with the indicated purified recombinant MET, c-Kit, or VEGFR-2 kinase domains (nM).
Table 2. Inhibitory activity of 10a–s against MET, c-Kit, and VEGFR-2.
Table 2. Inhibitory activity of 10a–s against MET, c-Kit, and VEGFR-2.
Molecules 24 04461 i002
No.Block AnR1Inhibitory Activity, IC50, nM a
c-Metc-KitVEGFR-2
10aA-404-F>5000>5000>5000
10bA-414-F>5000>5000>5000
10cA-614-F>5000>5000>5000
10dA-914-F>50001609208
10eA-40H>5000>5000>5000
10fA-41H>5000>5000>5000
10gA-61H>5000>5000>5000
10hA-91H>5000>50001031
10iA-404-Cl>5000>5000>5000
10jA-414-Cl>5000>5000>5000
10kA-614-Cl>5000>5000263
10lA-914-Cl>500010756.5
10mA-404-OCF3>5000>5000>5000
10nA-414-OCF3>5000>5000>5000
10oA-614-OCF3>5000>5000538
10pA-714-OCF3>5000>5000>5000
10qA-814-OCF3>5000>5000>5000
10rA-914-OCF3>500016931.7
10sA-912,4-F2>5000>5000887
39.9329.6279.9
a In vitro kinase assays were performed with the indicated purified recombinant MET, c-Kit, or VEGFR-2 kinase domains (nM).
Table 3. Individual energy components of the free energies of ligand with c-Kit and VEGFR-2.
Table 3. Individual energy components of the free energies of ligand with c-Kit and VEGFR-2.
N.O.CompoundΔEeleΔEvdwΔGnpΔGpolΔH-TΔSΔGcalIC50 (nM)
c-Kit (4U0I)9g−18.60−52.83−5.4942.12−34.8114.67−20.13832
9k−21.78−60.09−5.9144.37−43.4213.19−30.238.5
10r−18.78−59.91−6.5245.23−39.9815.90−24.08169
VEGFR-2 (3EFL)9g−26.32−53.91−5.3640.44−45.1515.18−29.97601
9k−26.56−60.46−5.8642.55−50.3415.55−34.79238.5
10r−27.82−64.08−6.3743.36−54.9216.38−38.5431.7

Share and Cite

MDPI and ACS Style

Sun, H.; Zhuo, L.; Dong, H.; Huang, W.; She, N. Discovery of 8-Amino-Substituted 2-Phenyl-2,7-Naphthyridinone Derivatives as New c-Kit/VEGFR-2 Kinase Inhibitors. Molecules 2019, 24, 4461. https://doi.org/10.3390/molecules24244461

AMA Style

Sun H, Zhuo L, Dong H, Huang W, She N. Discovery of 8-Amino-Substituted 2-Phenyl-2,7-Naphthyridinone Derivatives as New c-Kit/VEGFR-2 Kinase Inhibitors. Molecules. 2019; 24(24):4461. https://doi.org/10.3390/molecules24244461

Chicago/Turabian Style

Sun, Haiyan, Linsheng Zhuo, Huan Dong, Wei Huang, and Nengfang She. 2019. "Discovery of 8-Amino-Substituted 2-Phenyl-2,7-Naphthyridinone Derivatives as New c-Kit/VEGFR-2 Kinase Inhibitors" Molecules 24, no. 24: 4461. https://doi.org/10.3390/molecules24244461

Article Metrics

Back to TopTop