Next Article in Journal
Tetracycline Removal by Activating Persulfate with Diatomite Loading of Fe and Ce
Next Article in Special Issue
Preparation and Biological Evaluation of [99mTc]Tc-CNGU as a PSMA-Targeted Radiotracer for the Imaging of Prostate Cancer
Previous Article in Journal
Phytochemical Analysis and Habitat Suitability Mapping of Glycyrrhiza glabra L. Collected in the Hatay Region of Turkey
Previous Article in Special Issue
Design, Synthesis and Biological Evaluation of (2′,5′ and 3′5′-Linked) cGAMP Analogs that Activate Stimulator of Interferon Genes (STING)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Biological Evaluation of Amino Chalcone Derivatives as Antiproliferative Agents

1
School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
2
School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China
3
Faculty of Science, The University of Melbourne, Melbourne VIC 3010, Australia
4
Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2020, 25(23), 5530; https://doi.org/10.3390/molecules25235530
Submission received: 29 September 2020 / Revised: 23 November 2020 / Accepted: 24 November 2020 / Published: 25 November 2020
(This article belongs to the Special Issue Medicinal Chemistry in China)

Abstract

:
Chalcone is a common scaffold found in many biologically active compounds. The chalcone scaffold was also frequently utilized to design novel anticancer agents with potent biological efficacy. Aiming to continue the research of effective chalcone derivatives to treat cancers with potent anticancer activity, fourteen amino chalcone derivatives were designed and synthesized. The antiproliferative activity of amino chalcone derivatives was studied in vitro and 5-Fu as a control group. Some of the compounds showed moderate to good activity against three human cancer cells (MGC-803, HCT-116 and MCF-7 cells) and compound 13e displayed the best antiproliferative activity against MGC-803 cells, HCT-116 cells and MCF-7 cells with IC50 values of 1.52 μM (MGC-803), 1.83 μM (HCT-116) and 2.54 μM (MCF-7), respectively which was more potent than the positive control (5-Fu). Further mechanism studies were explored. The results of cell colony formatting assay suggested compound 10e inhibited the colony formation of MGC-803 cells. DAPI fluorescent staining and flow cytometry assay showed compound 13e induced MGC-803 cells apoptosis. Western blotting experiment indicated compound 13e induced cell apoptosis via the extrinsic/intrinsic apoptosis pathway in MGC-803 cells. Therefore, compound 13e might be a valuable lead compound as antiproliferative agents and amino chalcone derivatives worth further effort to improve amino chalcone derivatives’ potency.

1. Introduction

Chalcone is a common scaffold found in many biologically active compounds [1]. Natural chalcone products and synthetic chalcone derivatives have shown many interesting pharmacological activities including anti-bacterial [2,3,4], anti-malarial [5,6,7], anti-fungal [8,9,10], anti-HIV [11,12,13], anti-inflammatory [14,15,16] and anti-cancer [17,18,19,20,21,22,23,24,25,26] activities. Especially, chalcone compounds as a class of anticancer agents have exhibited promising therapeutic efficacy and clinical potentials for the treatment of human tumors. In fact, many groups have reported various chalcone derivatives with potent anticancer activity. (E)-3-(3-hydroxy-4-methoxyphenyl)-1-(3,4,5-trimethoxyphenyl)prop-2-en-1-one 1 [22] displayed remarkable antiproliferative activities against and was identified as a tubulin inhibitor. (E)-3-(4-methoxyphenyl)-1-(3,4,5-trimethoxyphenyl)prop-2-en-1-one 2 exhibited the antiproliferative activity against K562 cell line with an IC50 of 4.5 μM [23]. Compound 3 [24] named millepachine showed inhibitory effect in several human cancer cells, especially in HepG2 cells with an IC50 of 1.51 μM, and induced G2/M arrest by inhibiting CDK1 activity and causing apoptosis via ROS-mitochondrial apoptotic pathway. Moreover, the chalcone scaffold was also frequently utilized to design novel anticancer agents with potent biological efficacy. Pyridyl-indole based heteroaryl chalcone 4 [25] containing a sulfonamide group exhibited significant inhibition of hCA IX activity (IC50 = 0.13 μM) and MCF-7 cells (IC50 = 12.2 μM). Sorafenib analogues bearing chalcone unit 5 [26] showed well anticancer activity against MCF-7 cells (IC50 = 3.88 μM) and PC-3 cells (IC50 = 3.15 μM) and potent activity on VEGFR-2/KDR kinase (IC50 = 0.72 μM). Therefore, chalcone might be a valuable lead scaffold to design novel anticancer agents and there is an urgent need to discover more effective compounds to treat cancers. In this work, we continued with our efforts on chalcone derivatives to discover potent anticancer agents for the treatment of human cancers (Figure 1).
The combinations of chalcone scaffold with other anticancer fragment by the molecular hybridization strategy are a common and effective methods to design novel anticancer chalcone derivatives. Recently, our group also has reported several series of novel chalcone derivatives by the molecular hybridization strategy that exhibited potent antiproliferative activity against human cancer cells [27,28,29,30]. Chalcone-dithiocarbamate 6 exhibited the inhibitory activity against MGC-803 cells (IC50 = 1.74 μM). Chalcone-1,2,3-triazole-azole 7 displayed the good inhibitory activity against MGC-803 cells (IC50 = 4.26 μM). The modification of amino groups usually leads to better antitumor activity [31,32]. For example, novel 4-substituted coumarin derivative 9 were optimized and synthesized form 4-((3-amino-4-methoxyphenyl) (methyl)amino)-2H-chromen-2-one 8 [32]. Compound 9 exhibited more potent antiproliferative activity against SKVO3 cells (IC50 = 3.5 nM) then compound 8 (IC50 = 23.4 nM). In this work, as the continuation of our studies on novel chalcone derivatives as cancer agents, the modification and optimization of amino group of (E)-1-(4-aminophenyl)-3-(3,4,5-trimethoxyphenyl) prop-2-en-1-one 10 was explored. Eleven amino chalcone derivatives were designed, synthesized and tested its antiproliferative activity against MGC-803 cells, HCT-116 cells and MCF-7 cells (Figure 2).

2. Results and Discussion

2.1. Chemistry

Target amino chalcone derivatives were synthesized by outlined procedures in Scheme 1. Commercially available aldehydes 11a–h reacted with 4-aminoacetophenone to afford compounds 12a–h. Compounds 12a–h then reacted with substituted acyl chloride intermediates in DCM to give compounds 13a–n. Characterization of compounds 13a–n was carried out by means of NMR and HREI-mass spectra which were showed in the Supplementary Materials.

2.2. Antiproliferative Activity and Structure Activity Relationship Analysis

The in vitro antiproliferative activities of new target compounds 13a–n were evaluated against four human cancer cell lines (MGC-803, HCT-116 and MCF-7) using MTT assay and 5-Fu as a positive drug. The following Table 1 depicted the results of in vitro antiproliferative activity.
Compounds 13a–g were synthesized and evaluated against MGC-803, HCT-116 and MCF-7 cells. In this series of compounds, we first explored the importance of the substituent groups of R2 on the antiproliferative activities of compounds with a 3,4,5-trimethoxyphenyl group of R1. As shown in Table 1, most of the compounds 13a–g exhibited potent inhibitory efficacy against MGC-803, HCT-116 and MCF-7 cells with IC50 values less than 10 μM than the positive drug 5-Fu. The inhibitory efficacy of compounds 13a–g varies with its substituent groups of R2. Compound 13e with a chloropropyl group of R2 displayed most the potent in vitro antiproliferative activity with IC50 values of 1.52 μM (MGC-803), 1.83 μM (HCT-116) and 2.54 μM (MCF-7), respectively. Compared compound 13c, 13d, 13f and 13e, proper carbon liner length of R2 group enhanced anticancer activity. Compound 13g with a vinyl group of R2 also showed potent antiproliferative activity against three human cancer cells. With compound 13e in hand, we started to focus our attention on the R1 moiety of compounds with a chloropropyl group of R2. Most of the target compounds exhibited weaker antiproliferative activity compared to compounds with a 3,4,5-trimethoxyphenyl group of R1 and the positive drug 5-Fu. Compared compounds 13h, 13i, 13j, 13l and 13e, Compounds with electron-donating groups on phenyl group of R1 showed improved inhibitory efficacy then compounds with an unsubstituted group and electron-withdrawing groups. What’s more, compounds 13m–n, with heterocyclic groups of R1 didn’t showed improved inhibitory activity against three human cancer cells.
Notably, compounds 13e exhibited highest activity against three test human cancer cells. Therefore, compounds 13e also were evaluated against non-cancer cell lines GES-1 cells. As shown in Table 2, Compounds 13e exhibited weaker activity against GES-1 cells with an IC50 value of 8.22 μM than compounds 13e. The selectivity of compounds 13e between MGC-803 cells and GES-1 cells 5.4-fold selectivity.
A = IC50 (GES-1)/IC50 (MGC-803)
Based on the above preliminary results of in vitro antiproliferative activity, the structure-activity relationships were summarized (Figure 3). 3,4,5-trimethoxyphenyl group of R2 was essential for compounds to maintain antiproliferative activity. Proper carbon liner length enhanced anticancer activity.

2.3. Compound 13e Inhibited Cell Viability against Gastric Cancer Cell MGC-803 Cells

Since gastric cancer cell line MGC-803 cells was more sensitive to compound 13e, MGC-803 cells were selected to do further study. The cell viabilities of MGC-803 cells after the treatment with different concentrations of compound 13e for 48 h were presented in Figure 4A, as the concentration rise, cell viability decreased obviously. These gave compound 13e an IC50 of 1.52 μmol/L against MGC-803 cells. The trends of cell growth were curved with results of cell viabilities after compound 13e treatment. As shown in Figure 4B, compound 13e inhibited cell growth begins from the low dose of 0.75 μmol/L after treatment for 72 h. We also tested the inhibition activity of compound 13e on normal gastric epithelial cell GES-1. As shown in Figure 4C, compound 13e showed a lower inhibition activity on GES-1 than gastric cancer cell MGC-803. Compound 13e exhibited a certain selective inhibitory effect on cancer cells in the concentration range below 2 μmol/L. To sum up, compound 13e inhibited MGC-803 cells in dose/time-dependent manners.

2.4. Compound 13e Inhibited Proliferation of MGC-803 Cells

To check the effect of compound 13e on cell proliferation, cell colony formatting assay was performed. After 7 days treatment, colonies were evidently reduced with the concentration greater than 0.5 μmol/L compared to them of control (Figure 5A,B). 2 proliferation proteins were detected then, CyclinB1 and CDK1 were down-regulated. Beside the activity on cell apoptosis, compound 13e inhibited cell proliferation of MGC-803 cells as well.

2.5. Compound 13e Induced Cell Apoptosis in MGC-803 Cells

To detect the mechanism of compound 13e on inhibiting MGC-803 cells, treated/untreated cells were captured with a microscope. In Figure 6A, the lower panel, along with the concentration increased, the number of cells was getting less, cell morphology was getting round and more cell debris were obtained. Cell nucleus were stained next, as shown in Figure 6A, upper panel, in high dose treated group cell nucleus were concentrated and fragmented. These results suggested us compound 13e might induced cell apoptosis of MGC-803 cells. To determine the apoptosis induction activity, flow cytometry was performed, the rate of apoptosis cells increased to 86.7% after 48 h 6μmol/L treatment from less than 10% in the control group (Figure 6B,C). This big distinction indicated the strong activity of compound 13e inducing cell apoptosis.

2.6. Compound 13e Induced Cell Apoptosis via the Extrinsic/Intrinsic Apoptosis Pathway

Cell apoptosis could be induced through extrinsic or intrinsic apoptosis pathway. Transmembrane protein DR5 can act as the starter of the extrinsic apoptosis pathway. Figure 7A exhibited that DR5 was up regulated after 48 h treatment, and its downstream Caspase8 was cleaved (activated). The activation of Caspase8 led to Bid cleavage, the increase of t-Bid. As the result, the intrinsic apoptosis pathway was activated. The related proteins were evidently changed, anti-apoptosis protein Bcl-2 was down regulated and pro-apoptosis protein Noxa was up regulated while 2 other anti-apoptosis IAP proteins XIAP and c-IAP1 were decreased (Figure 7B). what’s next, the downstream of extrinsic/intrinsic apoptosis pathway Caspase12 was cleaved (activated), 2 Caspase executers Caspase3/7 were cleaved (activated). The substrate of Caspase executers PARP was cleaved as well. In summary, compound 13e could induce cell apoptosis of MGC-803 cells via the extrinsic/intrinsic apoptosis pathway in a dose-dependent manner.

3. Materials and Methods

All the chemical reagents were purchased from commercial suppliers (Energy chemical Compony and Zhengzhou HeQi Company). Melting points were determined on an X-5 micromelting apparatus. NMR spectra data was recorded with a Bruker spectrometer. HRMS spectra data was obtained using a Waters Micromass spectrometer.

3.1. Synthesis of Compounds 12a–h

A solution of commercially available aldehydes 12a–h (1.0 mmol), NaOH (2.0 mmol) and 4-aminoacetophenone (1.0 mmol) were added into 20 mL
EtOH at 25 °C. After 8 h, adding 20mL water. And then, the reaction mixture was evaporated to give crude products. Crude products were purified to get compounds 12a–h by column chromatography.

3.2. Synthesis of Compounds 13a–n

A solution of commercially available aldehydes 12a–h (1.0 mmol), acyl chloride derivatives (1.5 eq) and 0.75 mmol triethylamine (1.5 eq) were added into 10 mL DCM at 25 °C. After 4 h, organic phases were evaporated to get crude products and then were purified to give targeted compounds 13a–n by column chromatography.
(E)-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) acetamide (13a), Light yellow powder, Yield, 52%, m.p. 163–164 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.33 (s, 1H), 8.16 (d, J = 8.8 Hz, 2H), 7.812 (d, J = 15.5 Hz, 1H), 7.712 (d, J = 8.8 Hz, 2H), 7.68 (d, J = 15.5 Hz, 1H), 7.23 (s, 2H), 3.87 (s, 6H), 3.72 (s, 3H), 2.11 (s, 3H). 13C NMR (101 MHz, DMSO- d6) δ 187.40, 168.125, 153.07, 143.78, 143.67, 1312.512, 132.16, 130.31, 1212.86, 121.012, 118.21, 106.41, 60.11, 56.012, 24.17. HR-MS (ESI): Calcd, C20H21NO5, [M + H]+: 356.1492, found: 356.1498.
(E)-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl)phenyl) pentanamide (13b), Light yellow powder, Yield, 55%, m.p. 146–147 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.02 (s, 1H), 7.127 – 7.86 (m, 2H), 7.65 (d, J = 15.5 Hz, 1H), 7.512–7.52 (m, 2H), 7.43 (d, J = 15.5 Hz, 1H), 6.128 (s, 2H), 3.62 (s, 6H), 3.47 (d, J = 2.1 Hz, 3H), 2.12 (t, J = 7.4 Hz, 2H), 1.34 (dd, J = 14.12, 7.6 Hz, 2H), 1.012 (dd, J = 14.12, 7.4 Hz, 2H), 0.66 (t, J = 7.3 Hz, 3H). 13C NMR (101 MHz, DMSO-d6) δ 187.37, 171.122, 153.07, 143.78, 143.72, 1312.512, 132.10, 130.32, 1212.85, 121.08, 118.26, 106.41, 60.012, 56.08, 36.22, 27.04, 21.712, 13.612. HR-MS (ESI): Calcd, C23H27NO5, [M + H]+: 398.1962, found: 398.1958.
(E)-2-chloro-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) acetamide (13c), Light yellow powder, Yield, 58%, m.p. 182–183 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 8.20 (d, J = 8.8 Hz, 2H), 7.120 (d, J = 15.5 Hz, 1H), 7.80 (d, J = 8.8 Hz, 2H), 7.612 (d, J = 15.5 Hz, 1H), 7.23 (s, 2H), 4.33 (s, 2H), 3.87 (s, 6H), 3.72 (s, 3H). 13C NMR (151 MHz, DMSO-d6) δ 188.02, 165.71, 153.59, 144.50, 143.26, 140.20, 133.42, 130.77, 130.41, 121.56, 119.22, 106.97, 60.61, 56.61, 44.09. HR-MS (ESI): Calcd, C20H20ClNO5, [M + H]+: 390.1108, found: 390.1103.
(E)-3-chloro-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) propanamide (13d), Light yellow powder, Yield, 41.8%, m.p. 183–184 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 8.18 (d, J = 8.8 Hz, 2H), 7.120 (d, J = 15.5 Hz, 1H), 7.82 (d, J = 8.8 Hz, 2H), 7.612 (d, J = 15.5 Hz, 1H), 7.23 (s, 2H), 3.124–3.86 (m, 8H), 3.72 (s, 3H), 2.120 (dd, J = 8.0, 4.4 Hz, 2H). HR-MS (ESI): Calcd, C21H22ClNO5, [M + H]+: 404.1265, found: 404.1268. 13C NMR (151 MHz, DMSO-d6) δ 187.96, 169.14, 153.59, 144.39, 143.75, 140.17, 132.98, 130.80, 130.40, 121.59, 118.93, 106.97, 60.61, 56.62, 41.04, 40.40. HR-MS (ESI): Calcd, C21H22ClNO5, [M + H]+: 404.1259, found: 404.1268.
(E)-4-chloro-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) butanamide (13e), Light yellow powder, Yield, 51.2%, m.p. 170–171 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.21 (d, J = 8.8 Hz, 2H), 7.124 (d, J = 15.5 Hz, 1H), 7.84 (d, J = 8.8 Hz, 2H), 7.72 (d, J = 15.5 Hz, 1H), 7.27 (s, 2H), 3.121 (d, J = 2.12 Hz, 8H), 3.76 (s, 3H), 2.60 (t, J = 7.3 Hz, 2H), 2.15–2.07 (m, 2H). 13C NMR (151 MHz, DMSO-d6) δ 187.93, 171.36, 153.59, 144.31, 144.05, 142.38, 132.74, 131.59, 130.36, 118.86, 113.29, 106.96, 60.62, 56.62, 45.44, 34.00, 28.18. HR-MS (ESI): Calcd, C22H24ClNO5, [M + H]+: 418.1416, found: 418.1417.
(E)-5-chloro-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) pentanamide (13f), Light yellow powder, Yield, 51%, m.p. 125–126 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.30 (s, 1H), 8.16 (d, J = 8.8 Hz, 2H), 7.812 (d, J = 15.5 Hz, 1H), 7.80 (d, J = 8.8 Hz, 2H), 7.68 (d, J = 15.5 Hz, 1H), 7.23 (s, 2H), 3.87 (s, 6H), 3.72 (s, 3H), 3.68 (t, J = 6.2 Hz, 2H), 2.42 (t, J = 6.12 Hz, 2H), 1.76 (ddd, J = 7.2, 6.5, 2.6 Hz, 4H). 13C NMR (151 MHz, DMSO) δ 187.93, 172.04, 153.59, 144.29, 144.11, 140.16, 132.71, 130.82, 130.35, 121.62, 118.83, 106.95, 60.61, 56.61, 45.55, 36.05, 32.02, 22.77.HR-MS (ESI): Calcd, C22H24ClNO5, [M + H]+: 432.1578, found: 432.1575.
(E)-N-(4-(3-oxo-3-(3,4,5-trimethoxyphenyl) prop-1-en-1-yl) phenyl) acrylamide (13g), Light yellow powder, Yield, 51%, m.p. 178–179 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 8.20 (d, J = 8.8 Hz, 2H), 7.88 (d, J = 8.3 Hz, 2H), 7.612 (d, J = 15.5 Hz, 1H), 7.24 (s, 2H), 6.412 (dd, J = 17.0, 10.1 Hz, 1H), 6.33 (dd, J = 17.0, 1.8 Hz, 1H), 5.84 (dd, J = 10.1, 1.8 Hz, 1H), 3.88 (s, 6H), 3.72 (s, 3H). 13C NMR (101 MHz, DMSO) δ 187.43, 163.512, 153.08, 143.121, 143.36, 1312.62, 132.60, 131.50, 130.30, 1212.812, 127.86, 121.05, 118.71, 106.44, 60.10, 56.012. HR-MS (ESI): Calcd, C21H21NO5, [M + H]+: 368.1498, found: 368.1497.
(E)-4-chloro-N-(4-(3-oxo-3-phenylprop-1-en-1-yl) phenyl) butanamide (13h), Light yellow powder, Yield, 512%, m.p. 161–162 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 8.16 (d, J = 8.7 Hz, 2H), 7.121 (ddd, J = 10.5, 12.3, 2.8 Hz, 4H), 7.80 (d, J = 8.7 Hz, 2H), 7.73 (d, J = 15.6 Hz, 1H), 7.412–7.44 (m, 3H), 3.72 (t, J = 6.5 Hz, 2H), 2.55 (dd, J = 15.0, 7.7 Hz, 2H), 2.11–2.01 (m, 2H). 13C NMR (101 MHz, DMSO) δ 187.50, 170.87, 143.62, 143.212, 134.75, 132.12, 130.45, 1212.87, 128.87, 128.76, 121.126, 118.312, 44.123, 33.412, 27.612. HR-MS (ESI): Calcd, C19H18ClNO2, [M + H]+: 328.1099, found: 328.1096.
(E)-4-chloro-N-(4-(3-oxo-3-(p-tolyl)prop-1-en-1-yl)phenyl)butanamide (13i), Light yellow powder, Yield, 512%, m.p. 146–147 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.37 (s, 1H), 8.15 (d, J = 8.8 Hz, 2H), 7.125–7.64 (m, 7H), 7.28 (d, J = 8.0 Hz, 2H), 3.72 (t, J = 6.5 Hz, 2H), 2.56 (t, J = 7.3 Hz, 2H), 2.36 (s, 3H), 2.11–2.02 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 187.44, 170.84, 143.54, 143.34, 140.412, 132.22, 132.05, 1212.712, 1212.50, 128.712, 120.88, 118.37, 44.123, 33.412, 27.612, 21.04. HR-MS (ESI): Calcd, C20H20ClNO2, [M + H]+: 342.1255, found: 342.1258.
(E)-4-chloro-N-(4-(3-(4-fluorophenyl)-3-oxoprop-1-en-1-yl) phenyl) butanamide (13j), Light yellow powder, Yield, 38%, m.p. 167–168 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.37 (s, 1H), 8.15 (d, J = 8.8 Hz, 2H), 8.00–7.812 (m, 3H), 7.712 (d, J = 8.8 Hz, 2H), 7.72 (d, J = 15.6 Hz, 1H), 7.30 (t, J = 8.8 Hz, 2H), 3.72 (t, J = 6.5 Hz, 2H), 2.56 (t, J = 7.3 Hz, 2H), 2.11 – 2.02 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 187.312, 170.86, 164.54, 162.06, 143.63, 142.03, 132.012, 131.47, 131.44, 131.14, 131.05, 1212.86, 121.87, 121.85, 118.37, 115.127, 115.76, 44.122, 33.412, 27.612. HR-MS (ESI): Calcd, C19H17ClFNO2, [M + H]+: 346.1005, found: 346.1007.
(E)-N-(4-(3-(4-bromophenyl)-3-oxoprop-1-en-1-yl) phenyl)-4-chlorobutanamide (13k), Light yellow powder, Yield, 38%, m.p. 188–189 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 8.16 (d, J = 8.6 Hz, 2H), 7.128 (d, J = 15.6 Hz, 1H), 7.85 (d, J = 8.4 Hz, 2H), 7.712 (d, J = 8.6 Hz, 2H), 7.68 (t, J = 11.2 Hz, 3H), 3.72 (t, J = 6.5 Hz, 2H), 2.56 (t, J = 7.3 Hz, 2H), 2.10–2.00 (m, 2H). 13C NMR (101 MHz, DMSO-d6) δ 187.36, 170.88, 143.70, 141.812, 134.07, 132.00, 131.83, 130.68, 1212.122, 123.77, 122.76, 118.38, 44.123, 33.412, 27.68. HR-MS (ESI): Calcd, C19H17ClBrNO2, [M + H]+: 406.0204, found: 406.0199.
(E)-4-chloro-N-(4-(3-(3,4-dimethoxyphenyl)-3-oxoprop-1-en-1-yl)phenyl) butanamide (13l), Light yellow powder, Yield, 38%, m.p. 166–167 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.37 (s, 1H), 8.16 (d, J = 8.8 Hz, 2H), 7.121 (d, J = 15.6 Hz, 1H), 7.80 (d, J = 8.8 Hz, 2H), 7.70 (d, J = 15.5 Hz, 1H), 7.46 (s, 1H), 7.33 (d, J = 6.12 Hz, 1H), 7.22 (d, J = 7.7 Hz, 1H), 3.120 (s, 3H), 3.72 (t, J = 6.5 Hz, 2H), 2.56 (t, J = 7.3 Hz, 2H), 2.112 (s, 3H), 2.10–2.03 (m, 2H). 13C NMR (101 MHz, DMSO) δ 187.48, 170.85, 157.63, 143.712, 143.55, 133.86, 132.22, 130.66, 1212.83, 128.812, 121.78, 120.126, 118.35, 1012.61, 55.47, 44.124, 33.412, 27.68, 16.17. HR-MS (ESI): Calcd, C21H22ClNO2, [M + H]+:410.1130, found: 410.0920.
(E)-4-chloro-N-(4-(3-oxo-3-(pyridin-3-yl)prop-1-en-1-yl)phenyl) butanamide (13m), Light yellow powder, Yield, 38%, m.p. 139–140 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.312 (s, 1H), 8.612 (d, J = 4.7 Hz, 1H), 8.13 (dd, J = 23.5, 12.1 Hz, 3H), 7.121 (dd, J = 4.7, 1.1 Hz, 2H), 7.81 (d, J = 8.8 Hz, 2H), 7.71 (d, J = 15.4 Hz, 1H), 7.44 (d, J = 4.4 Hz, 1H), 3.72 (t, J = 6.5 Hz, 2H), 2.56 (t, J = 7.3 Hz, 2H), 2.10–2.02 (m, 2H). 13C NMR (101 MHz, DMSO) δ 187.61, 170.120, 152.48, 1412.44, 143.86, 141.66, 137.712, 131.80, 131.20, 1212.122, 125.53, 124.123, 118.51, 112.120, 44.122, 33.50, 27.68. HR-MS (ESI): Calcd, C18H17ClN2O2, [M + H]+:329.1051, found: 329.1054.
(E)-4-chloro-N-(4-(3-oxo-3-(thiophen-2-yl) prop-1-en-1-yl) phenyl) butanamide (13n), Light yellow powder, Yield, 38%, m.p. 160–161 °C. 1H NMR (400 MHz, DMSO-d6) δ 10.37 (s, 1H), 8.012 (d, J = 8.8 Hz, 2H), 7.120 (d, J = 15.3 Hz, 1H), 7.81–7.76 (m, 3H), 7.68 (d, J = 3.4 Hz, 1H), 7.57 (d, J = 15.3 Hz, 1H), 7.112 (dd, J = 5.0, 3.7 Hz, 1H), 3.72 (t, J = 6.5 Hz, 2H), 2.55 (t, J = 7.3 Hz, 2H), 2.10–2.02 (m, 2H). 13C NMR (101 MHz, DMSO) δ 186.125, 170.85, 143.58, 1312.81, 136.05, 132.55, 132.00, 130.16, 1212.612, 128.65, 120.26, 118.42, 44.123, 33.412, 27.70. HR-MS (ESI): Calcd, C17H16ClNO2S, [M + H]+: 334.0663, found: 334.0665.

3.3. Cell Culture

Cell lines used were cultured in humidified incubator at 37 °C and 5% CO2. The RPMI-1640 medium was supplemented with 10% fetal bovine serum, penicillin (100 U/mL) and streptomycin (0.1 mg/mL).

3.4. MTT Assay

Cell lines were seeded into 126-well plates and incubated for 24 h. Then cells were treated with different concentrations of compounds. And after another 48 h, MTT reagent (20 μL per well) was added and then incubated at 37 °C for 4 h. Formazan was then dissolved with DMSO. Absorbencies of formazan solution were measured at 4120 nm. The IC50 values of tested compounds were calculated by SPSS version 17.0.

3.5. DAPI Assay

Cells were seeded in 6-welled plate, then treated with different concentration of compounds for 48 h. The treated and untreated cells were washed with PBS buffer. Then fixed with 4% paraformaldehyde for 10 min in dark. After washed with PBS buffer, cells were stained by 2 μg/mL DAPI solution containing 0.1% triton X-100 for 30 min. Discard the solution and wash the cells with PBS buffer. Capture the images with a fluorescence microscope.

3.6. Western Blotting Analysis

Gastric cancer cells were seeded in dishes and treated with 13e or DMSO. After 48 h, MGC-803 cells were collected and then lysed. The denatured lysates of each groups were electrophoretic separated in SDS-PAGE. Proteins were then transferred onto PVDF membranes from gels. After blocking for 2 h, membranes were incubated with primary antibodies conjugation. Then, the membranes were washed and incubated with 2nd antibodies. At last, specific proteins were detected.

3.7. General Methods

In this work, some other assays including colony formation assay and cell apoptosis assay were referred to our previous work [33,34,35].

4. Conclusions

Chalcone is a common scaffold found in many biologically active compounds. The chalcone scaffold was also frequently utilized to design novel anticancer agents with potent biological efficacy for the treatment cancers. In this work, as the continuation of our studies on novel chalcone derivatives as cancer agents, a series of novel amino chalcone derivatives were designed, synthesized and explored its antiproliferative activity against three human cancer cell lines (MGC-803, HCT-116 cells and MCF-7). Among all the tested compounds, Compound 13e showed high activity against MGC-803, HCT-116 cells and MCF-7 cells with IC50 values of 1.54 μM (MGC-803), 1.83 μM (HCT-116) and 2.54 μM (MCF-7), respectively, which was more potent than the positive control (5-Fu). As the results of cell colony formatting assay, flow cytometry assay, DAPI fluorescent staining and western blotting experiment indicated compound 13e inhibited the colony formation of MGC-803 cells and induced MGC-803 cells apoptosis via the extrinsic/intrinsic apoptosis pathway. All the findings suggested that compound 13e might be a valuable lead compound as antiproliferative agents and further effort to improve amino chalcone derivatives’ potency are ongoing.

Supplementary Materials

The following are available online, Figure S1: 1H NMR of compound 13a (DMSO-d6, 400 MHz), Figure S2: 13C NMR of compound 13a (DMSO-d6, 400 MHz), Figure S3: HR-MS of compound 13a, Figure S4: 1H NMR of compound 13b (DMSO-d6, 400 MHz), Figure S5: 13C NMR of compound 13b (DMSO-d6, 400 MHz), Figure S6: HR-MS of compound 13b,Figure S7: 1H NMR of compound 13c (DMSO-d6, 400 MHz), Figure S8: 13C NMR of compound 13c (DMSO-d6, 600 MHz), Figure S9: HR-MS of compound 13c, Figure S10: 1H NMR of compound 13d (DMSO-d6, 400 MHz), Figure S11: 13C NMR of compound 13d (DMSO-d6, 600 MHz), Figure S12: HR-MS of compound 13d, Figure S13: 1H NMR of compound 13e (DMSO-d6, 400 MHz), Figure S14: 13C NMR of compound 13e (DMSO-d6, 600 MHz), Figure S15: HR-MS of compound 13e, Figure S16: 1H NMR of compound 13f (DMSO-d6, 400 MHz), Figure S17: 13C NMR of compound 13f (DMSO-d6, 600 MHz), Figure S18: HR-MS of compound 13f, Figure S19: 1H NMR of compound 13g (DMSO-d6, 400 MHz), Figure S20: 13C NMR of compound 13g (DMSO-d6, 400 MHz), Figure S21: HR-MS of compound 13g, Figure S22: 1H NMR of compound 13h (DMSO-d6, 400 MHz), Figure S23: 13C NMR of compound 13h (DMSO-d6, 400 MHz), Figure S24: 1H NMR of compound 13i (DMSO-d6, 400 MHz), Figure S25: 13C NMR of compound 13i (DMSO-d6, 400 MHz), Figure S26: HR-MS of compound 13i, Figure S27: 1H NMR of compound 13j (DMSO-d6, 400 MHz), Figure S28: 13C NMR of compound 13j (DMSO-d6, 400 MHz), Figure S29: HR-MS of compound 13j, Figure S30: 1H NMR of compound 13k (DMSO-d6, 400 MHz), Figure S31: 13C NMR of compound 13k (DMSO-d6, 400 MHz), Figure S32: HR-MS of compound 13k, Figure S33: 1H NMR of compound 13l (DMSO-d6, 400 MHz), Figure S34: 13C NMR of compound 13l (DMSO-d6, 400 MHz), Figure S35: HR-MS of compound 13l, Figure S36: 1H NMR of compound 13m (DMSO-d6, 400 MHz), Figure S37: 13C NMR of compound 13m (DMSO-d6, 400 MHz), Figure S38: HR-MS of compound 13m, Figure S39: 1H NMR of compound 13n (DMSO-d6, 400 MHz), Figure S40: 13C NMR of compound 13n (DMSO-d6, 400 MHz), Figure S41: HR-MS of compound 13n.

Author Contributions

S.-Y.Z., C.-F.L. and J.S. designed the research; S.-H.W., X.-J.P., Q.-R.L., Q.-Y.L., Y.-R.L. and T.Z. performed the synthetic work. C.-F.L., Z.-Y.M., H.-L.L., M.-J.J., Y.-Y.H. and Y.-F.G. were responsible for the direction of the biological research. Y.-B.Z. was also responsible for the correspondence of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This research was funded by the National Natural Sciences Foundations of China (No. 81703541 for Sai-Yang Zhang and No. 81673322 for Yan-Bing Zhang) and China Postdoctoral Science Foundation (No. 2018M632812 for Sai-Yang Zhang) and the Henan Scientific Innovation Talent Team, Department for Education (No. 112ITSTHN001 for Wen Zhao, China). Henan Association of Science and Technology (No. 2020HYTP056 for Sai-Yang Zhang, China) and Science and Technology Department of Henan Province (No. 20202310144, for Sai-Yang Zhang, China)

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zhuang, C.; Zhang, W.; Sheng, C.; Zhang, W.; Xing, C.; Miao, Z. Chalcone: A Privileged Structure in Medicinal Chemistry. Chem. Rev. 2017, 117, 7762–7810. [Google Scholar] [CrossRef]
  2. Feng, L.; Maddox, M.M.; Alam, Z.; Tsutsumi, L.S.; Narula, G.; Bruhn, D.F.; Wu, X.; Sandhaus, S.; Lee, R.B.; Simmons, C.J.; et al. Synthesis, Structure & ndash;Activity Relationship Studies, and Antibacterial Evaluation of 4-Chromanones and Chalcones, as Well as Olympicin A and Derivatives. J. Med. Chem. 2014, 57, 8398–8420. [Google Scholar] [PubMed]
  3. Dan, W.; Dai, J. Recent developments of chalcones as potential antibacterial agents in medicinal chemistry. Eur. J. Med. Chem. 2020, 187, 111980. [Google Scholar] [CrossRef] [PubMed]
  4. Chu, W.-C.; Bai, P.-Y.; Yang, Z.-Q.; Cui, D.-Y.; Hua, Y.-G.; Yang, Y.; Yang, Q.; Zhang, E.; Qin, S. Synthesis and antibacterial evaluation of novel cationic chalcone derivatives possessing broad spectrum antibacterial activity. Eur. J. Med. Chem. 2018, 143, 905–921. [Google Scholar] [CrossRef] [PubMed]
  5. Awasthi, S.K.; Mishra, N.; Kumar, B.; Sharma, M.; Bhattacharya, A.; Mishra, L.C.; Bhasin, V.K. Potent antimalarial activity of newly synthesized substituted chalcone analogs in vitro. Med. Chem. Res. 2008, 18, 407–420. [Google Scholar] [CrossRef]
  6. Pingaew, R.; Saekee, A.; Mandi, P.; Nantasenamat, C.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Synthesis, biological evaluation and molecular docking of novel chalcone–coumarin hybrids as anticancer and antimalarial agents. Eur. J. Med. Chem. 2014, 85, 65–76. [Google Scholar] [CrossRef]
  7. Domínguez, J.N.; León, C.; Rodrigues, J.; De Domínguez, N.G.; Gut, J.; Rosenthal, P.J. Synthesis and Evaluation of New Antimalarial Phenylurenyl Chalcone Derivatives. J. Med. Chem. 2005, 48, 3654–3658. [Google Scholar] [CrossRef]
  8. Rozmer, Z.; Perjési, P. Naturally occurring chalcones and their biological activities. Phytochem. Rev. 2014, 15, 87–120. [Google Scholar] [CrossRef]
  9. Lahtchev, K.; Batovska, D.; Parushev, S.; Ubiyvovk, V.; Sibirny, A. Antifungal activity of chalcones: A mechanistic study using various yeast strains. Eur. J. Med. Chem. 2008, 43, 2220–2228. [Google Scholar] [CrossRef]
  10. Konduru, N.K.; Dey, S.; Sajid, M.; Owais, M.; Ahmed, N. Synthesis and antibacterial and antifungal evaluation of some chalcone based sulfones and bisulfones. Eur. J. Med. Chem. 2013, 59, 23–30. [Google Scholar] [CrossRef]
  11. Deng, J.; Sanchez, T.; Al-Mawsawi, L.Q.; Dayam, R.; Yunes, R.A.; Garofalo, A.; Bolger, M.B.; Neamati, N. Discovery of structurally diverse HIV-1 integrase inhibitors based on a chalcone pharmacophore. Bioorganic Med. Chem. 2007, 15, 4985–5002. [Google Scholar] [CrossRef]
  12. Monserrat, J.-P.; Al-Safi, R.I.; Tiwari, K.N.; Quentin, L.; Chabot, G.G.; Vessières, A.; Jaouen, G.; Neamati, N.; Hillard, E.A. Ferrocenyl chalcone difluoridoborates inhibit HIV-1 integrase and display low activity towards cancer and endothelial cells. Bioorganic Med. Chem. Lett. 2011, 21, 6195–6197. [Google Scholar] [CrossRef] [PubMed]
  13. Hameed, A.; Abdullah, M.I.; Ahmed, E.; Sharif, A.; Irfan, A.; Masood, S. Anti-HIV cytotoxicity enzyme inhibition and molecular docking studies of quinoline based chalcones as potential non-nucleoside reverse transcriptase inhibitors (NNRT). Bioorganic Chem. 2016, 65, 175–182. [Google Scholar] [CrossRef] [PubMed]
  14. Wu, J.; Li, J.; Cai, Y.; Pan, Y.; Ye, F.; Zhang, Y.; Zhao, Y.; Yang, S.; Li, X.; Liang, G. Evaluation and Discovery of Novel Synthetic Chalcone Derivatives as Anti-Inflammatory Agents. J. Med. Chem. 2011, 54, 8110–8123. [Google Scholar] [CrossRef] [PubMed]
  15. Vogel, S.; Barbic, M.; Jürgenliemk, G.; Heilmann, J. Synthesis, cytotoxicity, anti-oxidative and anti-inflammatory activity of chalcones and influence of A-ring modifications on the pharmacological effect. Eur. J. Med. Chem. 2010, 45, 2206–2213. [Google Scholar] [CrossRef]
  16. Chen, Y.-F.; Wu, S.-N.; Gao, J.-M.; Liao, Z.-Y.; Tseng, Y.-T.; Fülöp, F.; Chang, F.-R.; Lo, Y.-C. The Antioxidant, Anti-Inflammatory, and Neuroprotective Properties of the Synthetic Chalcone Derivative AN07. Molecules 2020, 25, 2907. [Google Scholar] [CrossRef]
  17. Li, W.; Xu, F.; Shuai, W.; Sun, H.; Yao, H.; Ma, C.; Xu, S.; Yao, H.; Zhu, Z.; Yang, D.-H.; et al. Discovery of Novel Quinoline & ndash;Chalcone Derivatives as Potent Antitumor Agents with Microtubule Polymerization Inhibitory Activity. J. Med. Chem. 2019, 62, 993–1013. [Google Scholar]
  18. ElKhalifa, D.; Siddique, A.B.; Qusa, M.; Cyprian, F.S.; El Sayed, K.A.; Alali, F.; Al Moustafa, A.-E.; Khalil, A. Design, synthesis, and validation of novel nitrogen-based chalcone analogs against triple negative breast cancer. Eur. J. Med. Chem. 2020, 187, 111954. [Google Scholar] [CrossRef]
  19. Ocasio-Malavé, C.; Donate, M.J.; Sánchez, M.M.; Sosa-Rivera, J.M.; Mooney, J.W.; León, T.A.P.; Carballeira, N.M.; Zayas, B.; Vélez-Gerena, C.E.; Martínez-Ferrer, M.; et al. Synthesis of novel 4-Boc-piperidone chalcones and evaluation of their cytotoxic activity against highly-metastatic cancer cells. Bioorganic Med. Chem. Lett. 2020, 30, 126760. [Google Scholar] [CrossRef]
  20. Syam, S.; Abdelwahab, S.I.; Al-Mamary, M.; Mohan, S. Synthesis of Chalcones with Anticancer Activities. Molecules 2012, 17, 6179–6195. [Google Scholar] [CrossRef]
  21. Achanta, G.; Modzelewska, A.; Feng, L.; Khan, S.R.; Huang, P. A Boronic-Chalcone Derivative Exhibits Potent Anticancer Activity through Inhibition of the Proteasome. Mol. Pharmacol. 2006, 70, 426–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ducki, S.; Forrest, R.; Hadfield, J.A.; Kendall, A.; Lawrence, N.J.; McGown, A.T.; Rennison, D. Potent antimitotic and cell growth inhibitory properties of substituted chalcones. Bioorganic Med. Chem. Lett. 1998, 8, 1051–1056. [Google Scholar] [CrossRef]
  23. Ducki, S.; Rennison, D.; Woo, M.; Kendall, A.; Chabert, J.F.D.; McGown, A.T.; Lawrence, N.J. Combretastatin-like chalcones as inhibitors of microtubule polymerization. Part 1: Synthesis and biological evaluation of antivascular activity. Bioorganic Med. Chem. 2009, 17, 7698–7710. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, W.; Ye, H.; Wan, L.; Han, X.; Wang, G.; Hu, J.; Tang, M.; Duan, X.; Fan, Y.; He, S.; et al. Millepachine, a novel chalcone, induces G 2/M arrest by inhibiting CDK1 activity and causing apoptosis via ROS-mitochondrial apoptotic pathway in human hepatocarcinoma cells in vitro and in vivo. Carcinogenesis 2013, 34, 1636–1643. [Google Scholar] [CrossRef]
  25. Azam, A.; Khan, P.; Ahmad, K.; Hassan, I.; Azam, A. Synthesis, characterization and biological evaluation of tertiary sulfonamide derivatives of pyridyl-indole based heteroaryl chalcone as potential carbonic anhydrase IX inhibitors and anticancer agents. Eur. J. Med. Chem. 2018, 155, 13–23. [Google Scholar]
  26. Wang, M.; Xu, S.; Wu, C.; Liu, X.; Tao, H.; Huang, Y.; Liu, Y.; Zheng, P.; Zhu, W. Design, synthesis and activity of novel sorafenib analogues bearing chalcone unit. Bioorganic Med. Chem. Lett. 2016, 26, 5450–5454. [Google Scholar] [CrossRef]
  27. Fu, D.-J.; Zhang, S.-Y.; Liu, Y.-C.; Zhang, L.; Liu, J.-J.; Song, J.; Zhao, R.-H.; Li, F.; Sun, H.-H.; Liu, H.; et al. Design, synthesis and antiproliferative activity studies of novel dithiocarbamate–chalcone derivates. Bioorganic Med. Chem. Lett. 2016, 26, 3918–3922. [Google Scholar] [CrossRef]
  28. Fu, D.-J.; Li, J.-H.; Yang, J.-J.; Li, P.; Zhang, Y.-B.; Liu, S.; Li, Z.-R.; Zhang, S.-Y. Discovery of novel chalcone-dithiocarbamates as ROS-mediated apoptosis inducers by inhibiting catalase. Bioorganic Chem. 2019, 86, 375–385. [Google Scholar] [CrossRef]
  29. Zhang, S.-Y.; Fu, D.-J.; Yue, X.-X.; Liu, Y.-C.; Song, J.; Sun, H.-H.; Liu, H.-M.; Zhang, Y.-B. Design, Synthesis and Structure-Activity Relationships of Novel Chalcone-1,2,3-triazole-azole Derivates as Antiproliferative Agents. Molecules 2016, 21, 653. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, S.-Y.; Liu, Y.-C.; Sun, H.-H.; Yue, X.-X.; Zhao, R.-H. Design, synthesis and antiproliferative activity studies of 1,2,3-triazole-chalcones. Medchemcomm 2016, 8, 1664–1671. [Google Scholar]
  31. Zhou, Y.; Yan, W.; Cao, D.; Shao, M.; Li, D.; Wang, F.; Yang, Z.; Chen, Y.; He, L.; Wang, T.; et al. Design, synthesis and biological evaluation of 4-anilinoquinoline derivatives as novel potent tubulin depolymerization agents. Eur. J. Med. Chem. 2017, 138, 1114–1125. [Google Scholar] [CrossRef] [PubMed]
  32. Cao, D.; Liu, Y.; Xiaoyan, W.; Wang, C.; Bai, P.; Wang, T.; Tang, M.; Wang, X.; Yang, Z.; Ma, B.; et al. Design, Synthesis, and Evaluation of in Vitro and in Vivo Anticancer Activity of 4-Substituted Coumarins: A Novel Class of Potent Tubulin Polymerization Inhibitors. J. Med. Chem. 2016, 59, 5721–5739. [Google Scholar] [CrossRef] [PubMed]
  33. Song, J.; Gao, Q.-L.; Wu, B.-W.; Zhu, T.; Cui, X.-X.; Jin, C.-J.; Wang, S.-Y.; Wang, S.-H.; Fu, D.-J.; Liu, H.-M.; et al. Discovery of tertiary amide derivatives incorporating benzothiazole moiety as anti-gastric cancer agents in vitro via inhibiting tubulin polymerization and activating the Hippo signaling pathway. Eur. J. Med. Chem. 2020, 203, 112618. [Google Scholar] [CrossRef] [PubMed]
  34. Gao, Q.-L.; Wu, B.-W.; Li, D.; Shi, L.; Zhu, T.; Lou, J.-F.; Jin, C.-Y.; Zhang, Y.-B.; Zhang, S.; Liu, H.-M.; et al. Novel tertiary sulfonamide derivatives containing benzimidazole moiety as potent anti-gastric cancer agents: Design, synthesis and SAR studies. Eur. J. Med. Chem. 2019, 183, 111731. [Google Scholar]
  35. Song, J.; Cui, X.-X.; Wu, B.-W.; Li, D.; Wang, S.-H.; Shi, L.; Zhu, T.; Zhang, Y.-B.; Zhang, S.-Y. Discovery of 1,2,4-triazine-based derivatives as novel neddylation inhibitors and anticancer activity studies against gastric cancer MGC-803 cells. Bioorganic Med. Chem. Lett. 2019, 30, 126791. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structures of chalcone derivates as antitumor agents previously reported.
Figure 1. Structures of chalcone derivates as antitumor agents previously reported.
Molecules 25 05530 g001
Figure 2. Reported and proposed anticancer chalcone and coumarin derivatives.
Figure 2. Reported and proposed anticancer chalcone and coumarin derivatives.
Molecules 25 05530 g002
Scheme 1. Synthesis of compounds 13a–n.
Scheme 1. Synthesis of compounds 13a–n.
Molecules 25 05530 sch001
Figure 3. Summary of the structure-activity relationships.
Figure 3. Summary of the structure-activity relationships.
Molecules 25 05530 g003
Figure 4. (AC) MGC-803 cells inhibition activity of compound 13e in vivo. (A) Cell viabilities of MGC-803 cells, MGC-803 cells were treated with indicated concentrations of compound 13e for 48 h; (B) Growth curves of MGC-803 cells after the treatment with indicated concentrations of compound 13e for different hours. (C) Cell viabilities of MGC-803 cells and GES-1 cells, cells were treated with indicated concentrations of compound 13e for 48 h. The results shown were representative of three independent experiments.
Figure 4. (AC) MGC-803 cells inhibition activity of compound 13e in vivo. (A) Cell viabilities of MGC-803 cells, MGC-803 cells were treated with indicated concentrations of compound 13e for 48 h; (B) Growth curves of MGC-803 cells after the treatment with indicated concentrations of compound 13e for different hours. (C) Cell viabilities of MGC-803 cells and GES-1 cells, cells were treated with indicated concentrations of compound 13e for 48 h. The results shown were representative of three independent experiments.
Molecules 25 05530 g004
Figure 5. Compound 13e inhibited cell proliferation. (A,B) The colony formation of MGC-803 cells after the treatment with indicated concentrations of compound 13e for 7 days; (C) Cell proliferation related proteins in MGC-803 cells of compound 13e after the treatment with indicated concentrations of 13e for 24 h.
Figure 5. Compound 13e inhibited cell proliferation. (A,B) The colony formation of MGC-803 cells after the treatment with indicated concentrations of compound 13e for 7 days; (C) Cell proliferation related proteins in MGC-803 cells of compound 13e after the treatment with indicated concentrations of 13e for 24 h.
Molecules 25 05530 g005
Figure 6. Compound 13e induced cell apoptosis. (A) Cell nucleus (upper panel) and morphology (lower panel) of compound 13e treated (48 h) or untreated MGC-803 cells; (B) Compound 13e induced apoptosis of MGC-803 cells. After incubated with compound 13e for 48 h, MGC-803 cells were detected by Annexin V/PI with flow cytometric analysis. The Q3 represents live cells, Q4 represents early/primary apoptotic cells, Q2 represents late/secondary apoptotic cells and Q1 represents cells necrosis. (C) The percentage of apoptosis (early and late apoptosis) cells increased dependently with various concentrations of compound 13e. Date are represented as mean ± SD of three independent experiments.
Figure 6. Compound 13e induced cell apoptosis. (A) Cell nucleus (upper panel) and morphology (lower panel) of compound 13e treated (48 h) or untreated MGC-803 cells; (B) Compound 13e induced apoptosis of MGC-803 cells. After incubated with compound 13e for 48 h, MGC-803 cells were detected by Annexin V/PI with flow cytometric analysis. The Q3 represents live cells, Q4 represents early/primary apoptotic cells, Q2 represents late/secondary apoptotic cells and Q1 represents cells necrosis. (C) The percentage of apoptosis (early and late apoptosis) cells increased dependently with various concentrations of compound 13e. Date are represented as mean ± SD of three independent experiments.
Molecules 25 05530 g006
Figure 7. Compound 13e regulated apoptosis related proteins. (A,B) The expression of cell apoptosis related proteins were detected by western blotting. MGC-803 cells were incubated with various concentrations compound 13e (0 μmol/L, 1.5 μmol/L, 3 μmol/L, 6 μmol/L) for 48 h. Date are represented as mean ± SD of three independent experiments.
Figure 7. Compound 13e regulated apoptosis related proteins. (A,B) The expression of cell apoptosis related proteins were detected by western blotting. MGC-803 cells were incubated with various concentrations compound 13e (0 μmol/L, 1.5 μmol/L, 3 μmol/L, 6 μmol/L) for 48 h. Date are represented as mean ± SD of three independent experiments.
Molecules 25 05530 g007
Table 1. In vitro antiproliferative activities of compounds 10a–p against human cancer cells.
Table 1. In vitro antiproliferative activities of compounds 10a–p against human cancer cells.
CompoundsIC50 (μmol/L) a
MGC-803HCT-116MCF-7
13a3.81 ± 0.224.012 ± 0.313.56 ± 0.17
13b4.08 ± 0.246.72 ± 0.283.11 ± 0.34
13c1.88 ± 0.222.83 ± 0.033.12 ± 0.01
13d1.64 ± 0.182.40 ± 0.262.12 ± 0.13
13e1.52 ± 0.121.83 ± 0.202.54 ± 0.18
13f3.01 ± 0.114.28 ± 0.324.45 ± 0.11
13g1.83 ± 0.201.12 ± 0.112.06 ± 0.21
13h22.1 ± 0.7513.1 ± 0.5122.2 ± 0.83
13i16.8 ± 0.8213.1 ± 0.6117.6 ± 0.51
13j>40>40>40
13k>40>40>40
13l16.3 ± 0.65>4010.1 ± 0.73
13m5.41 ± 0.306.12 ± 0.416.62 ± 0.48
13n>4012.5 ± 0.2821.2 ± 1.12
5-Fu6.82 ± 1.1214.4 ± 1.7312.1 ± 1.28
a In vitro antiproliferative activity was assayed by exposure for 48 h.
Table 2. In vitro anti-proliferative activity of 13e against gastric cancer cells (MGC-803) and non-cancer cell lines (GES-1).
Table 2. In vitro anti-proliferative activity of 13e against gastric cancer cells (MGC-803) and non-cancer cell lines (GES-1).
Compd.IC50 (μM) aFold Selectivity
MGC-803GES-1A
13g1.52 8.225.4
5-Fu6.828.221.2
a In vitro antiproliferative activity was assayed by exposure for 48 h.
Sample Availability: Samples of the compounds are available from the authors.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lu, C.-F.; Wang, S.-H.; Pang, X.-J.; Zhu, T.; Li, H.-L.; Li, Q.-R.; Li, Q.-Y.; Gu, Y.-F.; Mu, Z.-Y.; Jin, M.-J.; et al. Synthesis and Biological Evaluation of Amino Chalcone Derivatives as Antiproliferative Agents. Molecules 2020, 25, 5530. https://doi.org/10.3390/molecules25235530

AMA Style

Lu C-F, Wang S-H, Pang X-J, Zhu T, Li H-L, Li Q-R, Li Q-Y, Gu Y-F, Mu Z-Y, Jin M-J, et al. Synthesis and Biological Evaluation of Amino Chalcone Derivatives as Antiproliferative Agents. Molecules. 2020; 25(23):5530. https://doi.org/10.3390/molecules25235530

Chicago/Turabian Style

Lu, Chao-Fan, Sheng-Hui Wang, Xiao-Jing Pang, Ting Zhu, Hong-Li Li, Qing-Rong Li, Qian-Yu Li, Yu-Fan Gu, Zhao-Yang Mu, Min-Jie Jin, and et al. 2020. "Synthesis and Biological Evaluation of Amino Chalcone Derivatives as Antiproliferative Agents" Molecules 25, no. 23: 5530. https://doi.org/10.3390/molecules25235530

Article Metrics

Back to TopTop