Next Article in Journal
Correction: Ding, S., et al. NNB-Type Tridentate Boryl Ligands Enabling a Highly Active Iridium Catalyst for C–H Borylation. Molecules 2019, 24, 1434
Next Article in Special Issue
Cytotoxic Effects of Newly Synthesized Heterocyclic Candidates Containing Nicotinonitrile and Pyrazole Moieties on Hepatocellular and Cervical Carcinomas
Previous Article in Journal
Silymarin’s Inhibition and Treatment Effects for Alzheimer’s Disease
Previous Article in Special Issue
Design, Synthesis, and Mechanism of Dihydroartemisinin–Coumarin Hybrids as Potential Anti-Neuroinflammatory Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design and Synthesis of Flavonoidal Ethers and Their Anti-Cancer Activity In Vitro

1
State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
2
The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
3
Bijie Medical College, Bijie 551700, China
*
Authors to whom correspondence should be addressed.
Molecules 2019, 24(9), 1749; https://doi.org/10.3390/molecules24091749
Submission received: 30 March 2019 / Revised: 2 May 2019 / Accepted: 3 May 2019 / Published: 6 May 2019
(This article belongs to the Special Issue Anticancer Agents: Design, Synthesis and Evaluation)

Abstract

:
Flavonoids are well-characterized polyphenolic compounds with pharmacological and therapeutic activities. However, most flavonoids have not been developed into clinical drugs, due to poor bioavailability. Herein, we report a strategy to increase the drugability of flavonoids by constructing C(sp2)-O bonds and stereo- as well as regioselective alkenylation of hydroxyl groups of flavonoids with ethyl-2,3-butadienoate allenes. Twenty-three modified flavonoid derivatives were designed, synthesized, and evaluated for their anti-cancer activities. The results showed that compounds 4b, 4c, 4e, 5e, and 6b exhibited better in vitro inhibitory activity against several cancer cell lines than their precursors. Preliminary structure–activity relationship studies indicated that, in most of the cancer cell lines evaluated, the substitution on position 7 was essential for increasing cytotoxicity. The results of this study might facilitate the preparation or late-stage modification of complex flavonoids as anti-cancer drug candidates.

1. Introduction

Consisting of more than 9000 compounds, flavonoids represent the most widely distributed polyphenols in nature [1,2]. In addition, flavonoids are ubiquitous in some medicinal plants and herbal remedies used in traditional medicine around the world, especially in China [2,3,4,5,6]. The characteristic skeleton of flavonoids is a phenylbenzopyrone moiety (C6-C3-C6), which could be further categorized according to the saturation level and the presence or absence of the central pyran ring. Many studies have confirmed that flavonoids are naturally occurring, pharmacologically active molecules. However, most flavonoids have not been developed as clinical drugs because of poor bioavailability (less than 5%), toxicity, and induction or inhibition of some metabolic enzymes [7]. To overcome these problems, structural modifications of the flavonoid skeletons have attracted great interest [8].
Allenes are highly reactive and have been used extensively in organic chemistry [9,10,11,12,13,14,15], due to the electronic or steric effects and their 1,2-diene functionality. The transition-metal catalyzed chemistry of allenes has become a recent focus of research [16,17]. We proposed that carbopalladation of allenes would provide a highly convenient method for the formation of a conjugated aryl ether, following β-H elimination by the introduction of a π-allyl palladium species.
In this research, we have developed a friendly and straightforward protocol to access various novel flavonoidal ethers by employing allenes as important multifunctional modules. Notably, both electrophilic groups and aliphatic chains can be embedded in the flavonoid scaffold smoothly with the above mentioned protocol. Therefore, more than 20 flavonoidal ethers were obtained successfully and evaluated for anti-cancer activities.

2. Materials and Methods

2.1. Instruments and Materials

1H- and 13C-NMR spectra were obtained on a 400 MHz (Varian, Inc) or 500 MHz (WIPM, China) spectrometer in CDCl3 or DMSO-d6 (tetramethylsilane (TMS) as internal standard). Chemical shifts (δ) were expressed in parts per million (ppm), relative to TMS (0 ppm). High resolution mass spectra (HRMS) were recorded on a ThermoFisher QE Focus apparatus. All the solvents were dried using standard methods and distilled before being used. Reagents and solvents were purchased from commercial sources. Solvents were purified according to the guidelines in the Purification of Laboratory Chemicals.

2.2. General Procedure for the Synthesis of Flavonoidal Ether Derivatives

2.2.1. Synthesis of Allenes

Allenes were synthesized according to previously described protocols [16,17,18].

2.2.2. Optimization of Reaction Conditions

We selected chrysin 2a and ethyl ester allene 1a as the model substrates in this study and carried out extensive screening of the reaction conditions. In an initial attempt, a variety of factors including types of Pd catalysts, bases, solvents, and ligands were examined. After extensive optimization, the product (3a) was obtained with an isolated yield of 41.7% in the presence of 10 mol% of PPh3/Pd(dba)2 as a catalyst, K2CO3 (2.0 equiv.) as a base, in a solution of MeCN under argon at 80 °C (Table 1, entry 1). Further investigations of the solvent indicated that the yield could be slightly improved to 43.0% by replacing MeCN with DMF (Table 1, entry 6). Furthermore, absence of Pd catalysts and ligands resulted in lower yield (Table 1, entry 14). None of the desired product was obtained when the base was abstracted away (Table 1, entries 15 and 16). Interestingly, only 11.2% yield of the expected product was obtained at lower temperatures (Table 1, entry 17). It was worth noting that 3a was the only isomer isolated, indicating a highly regio- and stereoselective process, in which the steric effect of the carbonyl or the hydrogen bond effect between the carbonyl and oxygen atom at position 7 may be critical for the highly regio- and stereoselective process.

2.2.3. General Procedure for the Synthesis of Target Derivatives 3a3i, 4a4g, 5a5e

Chrysin 2a (127.12 mg, 0.5 mmol), Pd(dba)2 (5.75 mg, 0.01 mmol), K2CO3 (138.21 mg, 1.0 mmol), PPh3 (13.11 mg, 0.05 mmol), allene (112.13 mg, 1.0 mmol), and anhydrous DMF (5 mL) were added to a 30 mL tube under argon, which was then sealed. The resulting mixture was stirred at 80 °C for 12 h. After completion, the reaction solution was cooled down to room temperature before 80 mL water was added, followed by the addition of 1 N HCl aqueous solution at 0 °C, until the pH value reached 7. Ethyl acetate (100 mL) was added to the reaction solution and the organic layer was washed sequentially with a large amount of water (4 × 200 mL), and then brine, before being dried over anhydrous Na2SO4. The solvent was evaporated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether–ethylacetate, 5:1) to give 3a (78.8 mg, 43% yield). Compounds 3b3i, 4a4g, and 5a5e were prepared according to similar procedures in which the double-substitution is a byproduct. The products 4a4g were generated in a shorter time compared with the other products (within 10 h).

2.2.4. General Synthetic Procedure for 6a6b Target Derivatives

The reaction with 1.0 mmol scale was carried out to assess the scalability of flavonoidal ether formation (Scheme 1, 4a). Under standard conditions, the yield was lower than that of the small-scale reaction. The reaction solution contained the following components: 4a (62.8 mg, 0.02 mmol), 1 mL methanol, and 1 N sodium hydroxide solution (2.0 equiv). The solution was then stirred at 60 °C for 5 min and 1 mL water was added before heating at 60 °C for 12 h. After completion, the reaction solution was cooled down to room temperature and 40 mL water as well as 30 mL ethyl acetate was added sequentially. Aqueous HCl (1 N) was added to the water layer at 0 °C until the pH value reached 4. Other post-processing procedures were similar to those performed to synthesize 4a. The desired product 6a was finally obtained (41.2 mg, 61% yield). The product 6b was prepared according to the same general procedure.
Ethyl (E)-3-((5-hydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) but-2-enoate (3a); yellow powder; yield 43.0%; HRMS(ESI): calcd. C21H17O6, m/z 365.1031 [M − H], found 365.1025; 1H-NMR (CDCl3, 500 MHz) δ(ppm): 7.88 (d, J = 7.0 Hz, 2H), 7.59–7.51 (m, 3H), 6.73 (s, 1H), 6.68 (d, J = 2.0 Hz, 1H), 6.49 (d, J = 2.0 Hz, 1H), 5.16 (s, 1H), 4.12 (q, J = 7.0 Hz, 2H), 2.49 (s, 3H), 1.23 (t, J = 7.0 Hz, 3H); 13C-NMR (CDCl3, 125 MHz) δ(ppm): 182.9, 170.5, 167.1, 164.7, 162.6, 159.4, 157.4, 132.3, 131.0, 129.3, 126.5, 108.6, 106.3, 104.8, 100.0, 99.8, 60.0, 18.2, 14.4.
Ethyl (E)-3-((5,6-dihydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) but-2-enoate (3b); yellow powder; yield 43.9 %; HRMS(ESI): calcd. C21H17O7, m/z 381.0980 [M − H], found 381.0970; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 13.02 (s, 1H), 8.04 (d, J = 7.2 Hz, 2H), 7.59–7.52 (m, 3H), 6.97 (s, 1H), 6.69 (s, 1H), 4.80 (s, 1H), 3.97 (q, J = 7.2 Hz, 2H), 2.40 (s, 3H), 1.09 (t, J = 7.2 Hz, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.3, 170.7, 166.6, 163.7, 156.7, 154.2, 152.4, 132.2, 130.7, 129.2, 126.5, 123.8, 105.0, 104.4, 94.8, 93.3, 59.2, 17.6, 14.2.
Diethyl 3,3′-((5-hydroxy-4-oxo-2-phenyl-4H-chromene-6,7-diyl) bis (oxy)) (2E,2′E)-bis (but-2-enoate) (3c); yellow powder; yield 29.3%; HRMS(ESI): calcd. C27H27O9, m/z 495.1650 [M + H]+, found 495.1647; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 11.76 (s, 1H), 8.04 (d, J = 6.0 Hz, 2H), 7.60–7.57 (m, 3H), 7.16 (s, 1H), 6.78 (s, 1 H), 4.77 (s, 1H), 4.63 (s, 1H), 4.01–3.94 (m, 4H), 2.44 (s, 3H), 2.39 (s, 3H), 1.16–1.03 (m, 6H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 174.7, 171.6, 170.4, 166.3, 166.2, 161.0, 155.4, 155.3, 143.0, 131.7, 131.3, 130.8, 129.1, 126.3, 109.8, 107.5, 102.7, 94.5, 93.9, 59.3, 59.1, 17.6, 17.3, 14.1.
Ethyl (E)-3-(5-(5,7-dihydroxy-4-oxo-4H-chromen-2-yl)-2-methoxyphenoxy)-2-methylacrylate (3d); yellow powder; yield 25.9%; HRMS(ESI): calcd. C22H19O8, m/z 411.1085 [M − H], found 411.1078; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 12.87 (s, 1H), 10.88 (s, 1H), 8.03 (dd, J = 9.0, 2.5 Hz, 1H), 7.87 (d, J = 2.5 Hz, 1H), 7.33 (d, J = 9 Hz, 1H), 6.95 (s, 1H), 6.52 (d, J = 2.0 Hz, 1H), 6.20 (d, J = 2.0 Hz, 1H), 4.66 (s, 1H), 3.99 (q, J = 7.0 Hz, 2H), 3.88 (s, 3H), 2.43 (s, 3H), 1.10 (t, J = 7.0 Hz, 3H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 181.8, 171.6, 166.3, 164.3, 162.1, 161.4, 157.4, 153.9, 141.1, 126.0, 123.6, 121.1, 113.8, 104.3, 103.8, 99.0, 94.6, 94.2, 59.2, 56.3, 17.7, 14.1.
Ethyl (E)-3-((2-(3-(((E)-4-ethoxy-4-oxobut-2-en-2-yl) oxy)-4-methoxyphenyl)-5-hydroxy-4-oxo-4H-chromen-7-yl) oxy) but-2-enoate (3e); yellow powder; yield 21.2%; HRMS(ESI): calcd. C28H29O10, m/z 525.1755 [M + H]+, found 525.1759; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 12.94 (s, 1H), 8.05 (dd, J = 8.5, 2.0 Hz, 1H), 7.86 (d, J = 2.0 Hz, 1H), 7.32 (d, J = 8.5 Hz, 1H), 7.05 (s, 1H), 6.95 (d, J = 2.0 Hz, 1H), 6.51 (d, J = 2.0 Hz, 1H), 5.05 (s, 1H), 4.66 (s, 1H), 4.05–3.95 (m, 4H), 3.90 (s, 3H), 2.42 (d, J = 3.5 Hz, 6H), 1.21–1.10 (m, 6H). 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 181.8, 171.6, 166.3, 164.3, 162.1, 161.4, 157.4, 153.9, 141.1, 126.0, 123.6, 121.1, 113.8, 104.3, 103.8, 99.0, 94.6, 94.2, 59.2, 56.3, 39.7, 39.5, 39.4, 17.7, 14.1.
Ethyl (E)-3-((5-hydroxy-2-(4-hydroxyphenyl)-4-oxo-4H-chromen-7-yl) oxy) but-2-enoate (3f); yellow powder; yield 30.3%; HRMS(ESI): calcd. C21H17O7, m/z 381.0980 [M − H], found 381.0986; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 13.10 (s, 1H), 10.45 (s, 1H), 7.97 (d, J = 8.0 Hz, 2H), 6.98–6.91 (m, 4H), 6.56 (s, 1H), 5.05 (s, 1H), 4.03 (q, J = 7.2 Hz, 2H), 2.42 (s, 3H), 1.14 (t, J = 7.2 Hz, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.3, 170.5, 166.1, 164.7, 161.6, 161.5, 158.4, 156.8, 128.8, 120.8, 116.0, 107.9, 104.0, 103.4, 100.2, 98.7, 59.4, 17.7, 14.1.
Ethyl (E)-3-(4-(5,7-dihydroxy-4-oxo-4H-chromen-2-yl) phenoxy) but-2-enoate (3g); yellow powder; yield 28.3%; HRMS(ESI): calcd. C21H17O7, m/z 381.0980 [M − H], found 381.0976; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 12.82 (s, 1H), 10.96 (s, 1H), 8.17 (d, J = 8.8 Hz, 2H), 7.34 (d, J = 8.4 Hz, 2H), 6.99 (s, 1H), 6.52 (d, J = 2.0 Hz, 1H), 6.22 (d, J = 2.0 Hz, 1H), 4.79 (s, 1H), 4.01 (q, J = 7.2 Hz, 2H), 2.45 (s, 3H), 1.12 (t, J = 7.2 Hz, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 181.9, 171.7, 166.2, 164.5, 162.4, 161.5, 157.5, 155.6, 128.8, 128.3, 122.2, 105.3, 103.9, 99.1, 96.9, 94.2, 59.3, 17.8, 14.2.
Ethyl 2-(5-(5,7-dihydroxy-4-oxo-4H-chromen-2-yl)-2-methylbenzo [d] [1,3] dioxol-2-yl) acetate (3h); yellow powder; yield 30.1%; HRMS(ESI): calcd. C21H17O8, m/z 397.0929 [M − H], found 397.0923; 1H-NMR (CDCl3, 400 MHz) δ(ppm): 12.62 (s, 1H), 7.43 (dd, J = 8.4, 1.6 Hz, 1H), 7.26 (d, J = 1.2 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.52 (s, 1H), 6.49 (d, J = 1.2 Hz, 1H), 6.38 (d, J = 1.6 Hz, 1H), 4.20 (q, J = 7.2 Hz, 2H), 3.06 (s, 2H), δ 1.88 (s, 3H), 1.25 (t, J = 7.2 Hz, 3H); 13C-NMR (CDCl3, 100 MHz) δ(ppm): 182.5, 168.3, 164.1, 164.0, 162.1, 157.9, 150.5, 148.1, 124.8, 121.6, 117.6, 108.9, 106.4, 104.9, 104.0, 100.0, 94.6, 61.4, 44.0, 25.0, 14.1.
Ethyl 2-(2-methyl-5-(3,5,7-trihydroxy-4-oxo-4H-chromen-2-yl) benzo [d] [1,3] dioxol-2-yl) acetate (3i); yellow powder; yield 30.1%; HRMS(ESI): calcd. C21H19O9, m/z 415.1024 [M + H]+, found 415.1018; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 12.40 (s, 1H), 10.83 (s, 1H), 9.57 (s, 1H), 7.74 (dd, J = 8.0, 1.5 Hz, 1H), 7.62 (d, J = 1.5 Hz, 1H), 7.03 (d, J = 8.0 Hz, 1H), 6.47 (d, J = 2.0 Hz, 1H), 6.19 (d, J = 2.0 Hz, 1H), 4.02 (q, J = 7.5 Hz, 2H), 3.13 (s, 2H), 1.78 (s, 3H), 1.10 (t, J = 7.5 Hz, 3H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 176.0, 167.6, 164.1, 160.7, 156.2, 148.2, 147.1, 145.9, 136.3, 124.5, 122.6, 117.3, 108.3, 107.3, 103.1, 98.3, 93.6, 60.3, 43.1, 24.7, 13.9.
Benzyl (E)-3-((5-hydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) but-2-enoate (4a); yellow powder; yield 33.0%; HRMS(ESI): calcd. C26H21O6, m/z 429.1333 [M + H]+, found 429.1324; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 12.93 (s, 1 H), 8.12 (d, J = 6.8 Hz, 2H), 7.74–7.56 (m, 3H), 7.32 (m, 5H), 7.07 (d, J = 2.0 Hz, 1H), 6.63 (d, J = 2.0 Hz, 1H), 5.10 (s, 1H), 5.07 (s, 2H), 2.46 (s, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.7, 171.3, 166.1, 164.2, 161.6, 158.7, 157.1, 136.3, 132.5, 130.4, 129.3, 128.5, 128.3, 128.1, 126.8, 108.3, 105.8, 104.4, 100.7, 98.3, 65.3, 17.9.
Dibenzyl 3,3′-((4-oxo-2-phenyl-4H-chromene-5,7-diyl) bis (oxy)) (2E,2′E)-bis (but-2-enoate) (4b); white powder; yield 29.8%; HRMS(ESI): calcd. C37H31O8, m/z 603.2013 [M + H]+, found 603.2010; 1H-NMR (CDCl3, 400 MHz) δ(ppm): 7.85 (d, J = 6.8 Hz, 2H), 7.52 (d, J = 7.6 Hz, 3H), 7.33 (m, J = 10H), 7.12 (d, J = 2.0 Hz, 1H), 6.71 (d, J = 2.0 Hz, 1H), 6.65 (s, 1H), 5.24 (s, 1H), 5.12 (s, 2H), 5.03 (s, 2H), 4.73 (s, 1H), 2.66 (s, 3H), 2.52 (s, 3H); 13C-NMR (CDCl3, 100 MHz) δ(ppm): 175.6, 174.0, 170.6, 167.0, 166.6, 162.3, 158.9, 157.5, 153.0, 136.3, 136.1 133.1, 131.9, 129.2, 128.7, 128.6, 128.5, 128.4, 128.3, 128.2, 126.3, 114.7, 113.3, 108.9, 108.0, 100.2, 95.2, 66.1, 65.8, 18.6, 18.3.
Dibenzyl 3,3′-((5-hydroxy-4-oxo-2-phenyl-4H-chromene-6,7-diyl) bis (oxy)) (2E,2′E)-bis (but-2-enoate) (4c); yellow powder; yield 24.2%; HRMS(ESI): calcd. C37H30O9Na, m/z 641.1782 [M + Na]+, found 641.1779; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 13.17 (s, 1H), 8.16-8.11 (m, 2H), 7.76–7.53 (m, 3H), 7.39-7.23 (m, 10H), 7.18 (s, 1H), 5.14 (s, 1H), 5.04 (s, 1H), 5.01 (s, 2H), 4.96 (s, 1H), 2.43 (s, 3H), 2.42 (s, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.9, 170.4, 170.3, 166.1, 165.8, 164.5, 153.7, 152.9, 150.5, 136.1, 130.3, 129.2, 128.4, 128.2, 128.1, 128.0, 126.7, 109.4, 105.4, 101.8, 98.0, 94.3, 65.2, 65.1, 17.5, 17.4.
Benzyl (E)-3-((5,6-dihydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) but-2-enoate (4d); yellow powder; yield 16.0%; HRMS(ESI): calcd. C26H20O7Na, m/z 467.1101 [M + Na]+, found 467.1092; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 13.06 (s, 1H), 11.37 (s, 1H), 8.08 (d, J = 6.0 Hz, 2H), 7.62–7.56 (m, 3H), 7.34-7.28 (m, 5H), 7.02 (s, 1H), 6.72 (s, 1H), 5.03 (s, 2H), 4.87 (s, 1H), 2.44 (s, 3H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 182.3, 171.2, 166.4, 163.7, 156.6, 154.2, 152.4, 136.3, 132.2, 130.7, 129.2, 128.4, 128.3, 128.0, 126.6, 123.7, 105.0, 104.4, 94.8, 92.9, 65.1, 17.7.
Benzyl (E)-3-((5-hydroxy-2-(3-hydroxy-4methoxyphenyl)-4-oxo-4H-chromen-7yl) oxy) but-2-enoate (4e); yellow powder; yield 32.5%; HRMS(ESI): calcd. C27H23O8, m/z 475.1387 [M + H]+, found 475.1375; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 13.07 (s, 1H), 9.48 (s, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.46 (d, J = 1.6 Hz, 1H), 7.32 (s, 5H), 7.08 (d, J = 8.4 Hz, 1H), 6.98 (s, 1H), 6.90 (s, 1H), 6.58 (d, J = 1.6 Hz, 1H), 5.10 (s, 1H), 5.06 (s, 2H), 3.87 (s, 3H), 2.45 (s, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.3, 171.3, 166.0, 164.5, 161.6, 158.4, 156.9, 151.5, 146.8, 132.2, 128.4, 128.2, 128.0, 122.6, 119.1, 113.3, 112.1, 108.0, 104.2, 104.1, 100.4, 98.1, 65.2, 55.8, 17.9.
Benzyl (E)-3-(4-(3,5,7-trihydroxy-4-oxo-4H-chromen-2-yl) phenoxy) but-2-enoate (4f); yellow powder; yield 23.6%; HRMS(ESI): calcd. C26H20O8Na, m/z 483.1050 [M + Na]+, found 483.1047; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 12.24 (s, 1H), 10.96 (s, 1H), 10.40 (s, 1H), 7.78–7.76 (m, 2H), 7.32–7.30 (m, 5H), 6.95–6.93 (m, 2H), 6.48 (d, J = 2.0 Hz, 1H), 6.23 (d, J = 2.0 Hz, 1H), 5.28 (s, 1H), 5.01 (s, 2H), 2.44 (s, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 175.0, 170.0, 166.3, 164.5, 161.1, 160.8, 156.8, 156.5, 136.2, 131.0, 130.1, 128.4, 128.3, 128.0, 119.5, 115.9, 104.1, 98.9, 94.8, 94.2, 65.1, 17.6.
Benzyl (E)-3-(4-(7-(((E)-3-(benzyloxy)-2-methyl-3-oxoprop-1-en-1-yl) oxy)-5-hydroxy-4-oxo-4H-chromen-2-yl) phenoxy)-2-methylacrylate (4g); yellow powder; yield: 64.9%; HRMS(ESI) calcd. C37H31O9, m/z 619.1963 [M + H]+, found 619.1954; 1H-NMR (CDCl3, 500 MHz) δ (ppm): 12.75 (s, 1H), 7.91 (d, J = 7.5 Hz, 2H), 7.33 (s, 10H), 7.18 (d, J = 8.0 Hz, 2H), 6.68 (d, J = 14.5 Hz, 2H), 6.50 (s, 1H), 5.20 (s, 1H), 5.11 (d, J = 7.5 Hz, 4H), 5.01 (s, 1H), 2.53 (d, J = 11.5 Hz, 6H); 13C-NMR (CDCl3, 125MHz) δ (ppm):182.7, 172.3, 171.2, 167.0, 166.9, 163.7, 162.6, 159.3, 157.3, 156.5, 136.19, 136.1, 128.6, 128.5, 128.4, 128.3, 122.4, 108.6, 106.2, 105.0, 100.1, 99.1, 97.5, 66.0, 65.9, 18.5, 18.3.
Ethyl (E)-3-((5-hydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) hex-2-enoate (5a); yellow powder; yield 32.0%; HRMS(ESI): calcd. C23H22O6Na, m/z 417.1309 [M + Na]+, found 417.1305; 1H-NMR (CDCl3, 400 MHz) δ(ppm): 12.79 (s, 1H), 7.89 (dd, J = 8.0, 2.0 Hz, 2H), 7.57–7.50 (m, 3H), 6.74 (s, 1H), 6.67 (d, J = 2.0 Hz, 1H), 6.49 (d, J = 2.0 Hz, 1H), 5.08 (s, 1H), 4.11 (dd, J = 2H), 2.94–2.90 (m, 2H), 1.74 (m, J = 15.2, 7.6 Hz, 2H), 1.23 (t, J = 7.2 Hz, 3H), 1.05 (t, J = 7.2 Hz, 3H); 13C-NMR (CDCl3, 100 MHz) δ(ppm): 182.9, 174.4, 166.9, 164.7, 162.6, 159.7, 157.5, 132.3, 131.0, 129.3, 126.5, 108.6, 106.3, 105.0, 100.1, 99.2, 60.0, 32.9, 20.9, 14.4, 13.9.
Ethyl (E)-3-((7-hydroxy-4-oxo-2-phenyl-4H-chromen-5-yl) oxy) hex-2-enoate (5b); yellow powder; yield 40.0%; HRMS(ESI): calcd. C23H23O6, m/z 395.1489 [M + H]+, found 395.1482; 1H-NMR (DMSO-d6, 400 MHz) δ(ppm): 8.26–8.01 (m, 2H), 7.56 (d, J = 7.2 Hz, 3H), 6.98 (d, J = 2.2 Hz, 1H), 6.74 (s, 1H), 6.49 (d, J = 2.2 Hz, 1H), 4.49 (s, 1H), 3.94 (dd, J = 14.4, 7.2 Hz, 2H), 2.94–2.90 (m, 2H), 1.76 (m, J = 2H), 1.09–0.99 (m, 6H), 1.00 (t, J = 7.2 Hz, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 176.1, 174.6, 166.2, 162.6, 160.7, 158.8, 151.8, 131.6, 130.9, 129.1, 126.2, 109.5, 108.6, 107.7, 101.5, 93.7, 32.6, 59.1, 19.9, 14.1, 13.8.
Ethyl (E)-3-((7-(((Z)-1-ethoxy-1-oxohex-2-en-3-yl) oxy)-6-hydroxy-4-oxo-2-phenyl-4H-chromen-5-yl) oxy) hex-2-enoate (5c); yellow powder; yield 7.0%; HRMS(ESI): calcd. C31H35O9, m/z 551.2276 [M + H]+, found 551.2260; 1H-NMR (CDCl3, 400 MHz) δ(ppm): 8.40 (s, 1H), 7.83 (d, J = 6.4 Hz, 2H), 7.55–7.48 (m, 3H), 7.11 (s, 1H), 6.60 (s, 1H), 4.90 (s, 1H), 4.70 (s, 1H), 4.10–4.03 (m, 4H), 3.05–3.02 (m, 2H), 2.93–2.90 (m, 2H), 1.87–1.72 (m, 4H), 1.21–1.16 (m, J = 6H), 1.07–1.02 (m, 6H); 13C-NMR (CDCl3, 125 MHz) δ(ppm): 176.5, 175.5, 173.6, 167.3, 167.1, 162.5, 156.0, 154.6, 144.1, 132.0, 131.6, 131.0, 129.2, 126.3, 111.4, 107.8, 102.7, 95.6, 94.5, 60.1, 59.9, 33.6, 33.1, 21.2, 20.7, 14.4, 14.2.
Ethyl (E)-3-((5,6-dihydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) hex-2-enoate (5d); yellow powder; yield 11.2%; HRMS(ESI): calcd. C23H21O7, m/z 409.1293 [M − H], found 409.1289; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 13.03 (s, 1H), 11.36 (s, 1H), 8.07 (d, J = 7.5 Hz, 2H), 7.61–7.56 (m, 3H), 7.01 (s, 1H), 6.70 (s, 1H), 4.74 (s, 1H), 3.98 (dd, J = 14.5, 6.0 Hz, 2H), 2.86 (t, J = 7.0 Hz, 2H), 1.71 (dd, J = 15.0, 7.5 Hz, 2H), 1.10 (t, J = 7.0 Hz, 3H), 1.00 (t, J = 7.5 Hz, 3H); 13C-NMR (DMSO-d6, 100 MHz) δ(ppm): 182.3, 173.8, 166.3, 163.6, 156.7, 154.1, 152.4, 132.1, 130.7, 129.2, 126.5, 123.7, 104.9, 104.4, 94.7, 92.8, 59.1, 32.3, 20.4, 14.1, 13.5.
Diethyl 3,3′-((5-hydroxy-4-oxo-2-phenyl-4H-chromene-6,7-diyl) bis (oxy)) (2E,2′E)-bis (hex-2-enoate) (5e); yellow powder; yield 14.8%; HRMS(ESI): calcd. C31H34O9Na, m/z 573.2095 [M + Na]+, found 573.2106; 1H-NMR (CDCl3, 500 MHz) δ(ppm): 12.96 (s, 1H), 7.89 (d, J = 7.5 Hz, 2H), 7.60–7.53 (m, 3H), 6.76 (d, J = 9.0 Hz, 1H), 5.03 (s, 1H), 4.85 (s, 1H), 4.13–4.06 (m, 4H), 2.93–2.87 (m, 4H), 1.77–1.69 (m, 4H), 1.21 (dd, J = 15.2, 7.6 Hz, 6H), 1.06–1.00 (m, 6H); 13C-NMR(CDCl3, 125 MHz) δ(ppm): 182.9, 174.0, 173.8, 167.1, 166.7, 165.2, 154.1, 153.8, 151.8, 132.5, 130.9, 129.4, 128.4, 126.6, 109.5, 105.9, 100.7, 98.7, 94.5, 60.0, 59.7, 33.1, 32.8, 21.0, 20.9, 14.4, 14.0, 13.9.
(E)-3-((5-hydroxy-4-oxo-2-phenyl-4H-chromen-7-yl) oxy) but-2-enoic acid (6a); yellow powder; yield 69.4%; HRMS(ESI): calcd. C19H13O6, m/z 337.0718 [M + H]+, found 337.0711; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 12.93 (s, 1H), 8.14 (d, J = 6.4 Hz, 2H), 7.76–7.58 (m, 3H), 7.15 (s, 1H), 7.05 (d, J = 2.0 Hz, 1H), 6.61 (d, J = 2.0 Hz, 1H), 5.09 (s, 1H), 2.41 (s, 3H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 182.8, 169.7, 167.6, 164.3, 161.7, 159.3, 157.2, 132.6, 130.6, 129.4, 126.9, 108.1, 105.9, 104.2, 100.7, 100.3, 17.7.
2-(2-methyl-5-(3,5,7-trihydroxy-4-oxo-4H-chromen-2-yl) benzo[d] [1,3] dioxol-2-yl) acetic acid (6b); yellow powder; yield 43.7%; HRMS(ESI): calcd. C19H15O9, m/z 387.0711 [M + H]+, found 387.0707; 1H-NMR (DMSO-d6, 500 MHz) δ(ppm): 12.40 (s, 1 H), 7.73 (dd, J = 10.5, 1.5 Hz, 1H), 7.61 (d, J = 1.5 Hz, 1H), 7.03 (d, J = 10.5 Hz, 1H), 6.46 (d, J = 2.5 Hz, 1H), 6.19 (d, J = 2.5 Hz, 1H), 3.02 (s, 2H), 1.78 (s, 3H); 13C-NMR (DMSO-d6, 125 MHz) δ(ppm): 217.5, 176.0, 172.1, 169.2, 164.1, 160.7, 156.2, 148.2, 147.1, 146.0, 136.2, 124.8, 122.9, 117.6, 108.4, 107.3, 103.1, 98.3, 93.7, 24.5, 21.1.

2.3. Anti-Cancer Activity Assay

2.3.1. Cell Lines and Cell Culture

The human leukemia cell lines K562 and HEL, the non-small cell lung cancer A549, and the prostate cancer cell line PC3 were obtained from the Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science (Guiyang, China). Cells were cultured as a monolayer in DMEM (Hyclone, Germany), supplemented with 10% heat-inactivated research-grade fetal bovine serum (Hyclone, Germany) and penicillin/streptomycin (Sigma, St. Louis, MO, USA) at 37 °C in a humidified atmosphere, containing 5% CO2.

2.3.2. Cytotoxic Activity Assay

The cytotoxic activity of compounds was measured by MTT assay, using adriamycin as the positive control. Cells were seeded in 96-well microculture plates at a density of 1 × 104 cells/well and left to adhere for 24 h. The cells were then exposed to different concentrations of the compounds for 48 h. MTT was added to each well at a final concentration of 0.5 mg/mL and cells were incubated at 37 °C for an additional 4 h. The medium was then discarded and 200 μL Tris-DMSO solution was added to each well. The dark blue formazan crystals formed were dissolved by slight shaking, and the absorbance was measured at 490 nm, using an ELISA plate reader.

2.3.3. Statistical Analysis

The data were analyzed using SPSS 18.0 and reported as mean ± standard deviation (SD) of the number of experiments indicated. For all the measurements, one-way ANOVA followed by a Student’s t-test was used to assess the statistical significance of the differences between each group. The statistical significance of the difference between two groups was assessed using the LSD method. p < 0.05 was considered to indicate statistical significance. The data are presented as the mean ± standard error of the mean (SEM) of three assays.

3. Results and Discussion

3.1. Synthesis

The optimal reaction conditions were established (Table 1, entry 6). The substrate scope was further extended to other types of allenes (Figure 1). As expected, various substituent groups led to a variety of products. When using either the monoallene (ethyl ester/benzyl ester) or diallene, the Pd catalyst efficiently promoted this C-O functionalization. Intriguingly, the application of triethylamine as a base instead of potassium carbonate was demonstrated to be more efficient for target product purification (4e, 4f). However, completion of the reaction is hard to achieve. Starting flavonoids with OH groups at positions 3′ and 4′ easily generated the 1,3-dioxolane byproducts, instead of the desired ones. The key intermediates 1a1c were prepared according to reported methods [18,19,20].
Under optimal conditions, as illustrated in Figure 2, a variety of flavonoid substrates was investigated. For instance, flavones and flavonols were well tolerated, providing ample opportunities for further anti-cancer activity evaluation of the derivatives. We found that the property of flavonoid and the substituted phenolic hydroxyl groups had a great impact on the formation of the desired products. Notably, it was more challenging to separate the analog products when more than two hydroxyl groups were presented. Finally, the target products (3a3i, 4a4g, 5a5e, 6a, 6b) were successfully purified by silica gel column chromatography. The configuration of the C=C bond in 3a and 4a was determined by the NOE (Nuclear Overhauser Effect) study. The structures of final compounds were characterized by 1H-, 13C-NMR, and MS spectrometry.

3.2. Anti-Cancer Activity

The anti-tumor activity of all derivatives in vitro was summarized in Table 2. Results showed that the inhibitory effect of compound 4e on the growth of K562 was six times stronger (p < 0.01) than that of the substrate, diosmetin, which indicated that the anti-tumor activity of the diosmetin derivative was associated with the more lipophilic benzyl ester than -OCH3 moiety at R6 position of the substrate. Compounds 5e, 3h, and 6b showed better activity (p < 0.01) on the proliferation of HEL than that of the substrates, baicalein, kaempferol, and quercetin, respectively. These results suggested that the anti-leukemia activity may be improved by increasing steric hindrance, such as by introducing propyl and 1,3-dioxolane groups onto baicalein. Compound 6b exhibited an enhanced inhibitory activity on the growth of leukemia cells (K562 and HEL) and non-small cell lung cancer A549 compared with those of 3i, which can be speculated as the enhanced water solubility associated with the carboxyl group. We then synthesized compounds 3e, 4b, and 4c to improve the anti-cancer activity (p < 0.01) on the proliferation of PC3 compared with the substrates, diosmetin, chrysin, and baicalein, respectively. The results suggested that introducing an ethyl and benzyl esters could increase their anti-cancer activity.

4. Conclusions

Twenty-three flavonoidal ether derivatives were synthesized without the steps of introducing protective groups followed by deprotection. The in vitro tumor growth inhibitory activities of all the derivatives were assayed using four human cancer cell lines, K562, A549, HEL, and PC3. In general, compounds containing 1,3-dioxolane, such as 6b, possessed broad-spectrum inhibitory activity against the above four cancer cells. Preliminary structure–activity relationship studies indicated that the position-7 substituents were essential for the cytotoxicities of the derivatives. These results provided new insight into developing flavonoid-derived anti-cancer agents.

Author Contributions

Conceptualization, L.J. and M.-L.W.; Methodology, L.J. and X.-Y.Z.; Software, L.J., X.-Y.Z.; Validation, X.-Y.Z. and C.C.; Formal analysis, L.J.; Resources, W.-D.P. and H.L.; Data curation, L.J., Y.L., and M.-L.W.; Writing—original draft preparation, L.J.; Writing—review and editing, L.J., H.R., W.-D.P., and H.L.; Project administration, W.-D.P. and H.L.; Funding acquisition, W.-D.P. and H.L.

Funding

Supported by the National Natural Science Foundation of China (No. 81660580, 81702914) and the Science and Technology Department of Guizhou Province, China (QKHRC[2016]4037, QKHJC[2017]1412).

Acknowledgments

We would like to acknowledge the help of my colleagues Ni Zhang, Xiao-Pan Ma, Chang-Fen Chen, and Sai Jiang.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Huang, X.; Liang, J.P.; Hao, B.C. Study on molecular modification and structure-activity relationship of flavonoids. J. Anhui Arg. Sci. 2015, 43, 57–61. [Google Scholar]
  2. Dhruti, B.; Ronak, S.; Gaurav, K.; Dashora, A. Synthesis and pharmacological activities of flavones: A review. Indo Am. J. Pharm. Res. 2016, 6, 4345–4363. [Google Scholar]
  3. Craig, W.J. Health-promoting properties of common herbs. Am. J. Clin. Nutr. 1999, 70, 491S–499S. [Google Scholar] [CrossRef] [PubMed]
  4. Kadarian, C.; Broussalis, A.M.; Mino, J.; Lopez, P.; Gorzalczany, S.; Ferraro, G.; Acevedo, C. Hepatoprotective activity of achyrocline satureioides (Lam) D. C. Pharmacol. Res. 2002, 45, 57–61. [Google Scholar] [CrossRef] [PubMed]
  5. Pascual, M.E.; Slowing, K.; Carretero, E.; Sánchez Mata, D.; Villar, A. Lippia: Traditional uses, chemistry and pharmacology: A review. J. Ethnopharmacol. 2001, 76, 201–214. [Google Scholar] [CrossRef]
  6. Samuelsen, A.B. The traditional uses, chemical constituents and biological activities of Plantago major L. A review. J. Ethnopharmacol. 2000, 71, 1–21. [Google Scholar] [CrossRef]
  7. Tang, L.; Feng, Q.; Zhao, J.; Dong, L.; Liu, W.; Yang, C.; Liu, Z. Involvenment of UDP-glucuronosyltranferases and sulfotransferases in the liver and intestinal first- pass metabolism of seven flavones in C57 mice and humans in vitro. Food Chem. Toxicol. 2012, 50, 1460–1467. [Google Scholar] [CrossRef] [PubMed]
  8. Singh, M.; Kaur, M.; Silakari, O. Flavones: An important scaffold for medicinal chemistry. Eur. J. Med. Chem. 2014, 84, 206–239. [Google Scholar] [CrossRef] [PubMed]
  9. Ma, S.M. Some typical advances in the synthetic applications of allenes. Chem. Rev. 2005, 105, 2829–2872. [Google Scholar] [CrossRef] [PubMed]
  10. Brasholz, M.; Reissig, H.U.; Zimmer, R. Sugars, alkaloids, and heteroaromatics: Exploring heterocyclic chemistry with alkoxyallenes. Acc. Chem. Res. 2009, 42, 45–56. [Google Scholar] [CrossRef] [PubMed]
  11. Pfrengle, F.; Reissig, H.U. Amino sugars and their mimetics Via 1,2-oxazines. Chem. Soc. Rev. 2010, 39, 549–557. [Google Scholar] [CrossRef] [PubMed]
  12. Yu, S.C.; Ma, S.M. Allenes in catalytic asymmetric synthesis and natural product syntheses. Angew. Chem. Int. Ed. 2012, 51, 3074–3112. [Google Scholar] [CrossRef] [PubMed]
  13. Ye, J.T.; Ma, S.M. Palladium-catalyzed cyclization reactions of allenes in the presence of unsaturated carbon-carbon bonds. Acc. Chem. Res. 2014, 47, 989–1000. [Google Scholar] [CrossRef] [PubMed]
  14. Alcaide, B.; Almendros, P.; Aragoncillo, C. Cyclization reactions of bis(allenes) for the synthesis of polycarbo(hetero)cycles. Chem. Soc. Rev. 2014, 43, 3106–3135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Neff, R.K.; Frantz, D.E. Recent applications of chiral allenes in axial-to-central chirality transfer reactions. Tetrahedron 2015, 71, 7–18. [Google Scholar] [CrossRef]
  16. Jose, L.M.; Lvan, V.; Fernando, L. Allenes and derivatives in gold(I)- and platinum(II)-catalyzed formal cycloadditions. Acc. Chem. Res. 2019, 52, 465–479. [Google Scholar]
  17. Egle, M.B.; Gianluigi, B.; Michael, S.C.; Giofrè, S. Chapter one—Transition metal- catalyzed intramolecular amination and hydroammination reactions of allenes. Adv. Organomet. Chem. 2018, 69, 1–71. [Google Scholar]
  18. Campbell, K.A.; House, H.O.; Surber, B.W.; Trahanovsky, W.S. Enones with strained double bonds. 10. use of flash vacuum pyrolysis to obtain bicyclo[3.3.1]non-1-en-3-one. J. Org. Chem. 1987, 52, 2474–2481. [Google Scholar] [CrossRef]
  19. Rout, L.; Harned, A.M. Allene carboxylates as dipolarophiles in Rh-catalyzed carbonyl ylide cycloadditions. Chem. Eur. J. 2009, 15, 12926–12928. [Google Scholar] [CrossRef] [PubMed]
  20. Li, R.D.; Leng, P.L.; Liu, B.; Wang, X.; Ge, Z.; Li, R. Efficient and regioselective one-pot synthesis of S-vinyl dithiocarbamates from electron-deficient allenes, amines and CS2. Tetrahedron 2016, 72, 5707–5712. [Google Scholar] [CrossRef]
Sample Availability: Samples of the compounds are available from the authors.
Scheme 1. Synthesis of flavonoidal ether and derivatization.
Scheme 1. Synthesis of flavonoidal ether and derivatization.
Molecules 24 01749 sch001
Figure 1. Types of allenes.
Figure 1. Types of allenes.
Molecules 24 01749 g001
Figure 2. Substrate scope of flavonoids and allenes.
Figure 2. Substrate scope of flavonoids and allenes.
Molecules 24 01749 g002
Table 1. Optimization of reaction conditions.
Table 1. Optimization of reaction conditions.
Molecules 24 01749 i001
EntryCatalystBaseLigandSolventYield (%)EntryCatalystBaseLigandSolventYield (%)
1Pd(dba)2K2CO3PPh3DMA [a]<510Pd(dba)2Cs2CO3PPh3DMF18.4
2Pd(dba)2K2CO3PPh3EtOH13.311PdCl2K2CO3-DMF15.8
3Pd(dba)2K2CO3PPh3MeCN41.712Pd(OAc)2K2CO3-DMF24.1
4Pd(dba)2K2CO3PPh3DCM<513PdCl2(PPh3)2K2CO3-DMF13.3
5Pd(dba)2K2CO3PPh3DMA [b]32.014-K2CO3-DMF14.9
6Pd(dba)2K2CO3PPh3DMF43.015Pd(OAc)2--DMFNO.
7Pd(dba)2NaHPPh3DMF14.216Pd(dba)2--DMFNO.
8Pd(dba)2NH(iPr)2PPh3DMF<517Pd(dba)2K2CO3PPh3DMF11.2[c]
9Pd(dba)2Et3NPPh3DMF17.2------
Conditions: 1a (0.4 mmol), 2a (0.2 mmol), isolated yield. [a] N,N-Dimethylaniline; [b] N,N-Dimethylacetamide; [c] Reaction occurred at rt.
Table 2. The anti-cancer activities of the synthetic derivatives.
Table 2. The anti-cancer activities of the synthetic derivatives.
CompdInhibition Rate % (5 µM)
K562A549HELPC3
3a34.1 ± 4.3−23.3 ± 2.1−2.5 ± 3.27.1 ± 3.2
3b28.4 ± 4.9−14.9 ± 2.827.5 ± 4.11.5 ± 3.4
3c23.1 ± 3.23.8 ± 1.721.2 ± 2.316.1 ± 1.2
3d14.0 ± 1.7−39.6 ± 2.92.7 ± 1.936.6 ± 3.6
3e−45.0 ± 1.9−20.5 ± 2.510.5 ± 2.951.7 ± 3.9 **
3f−13.7 ± 2.219.7 ± 2.6−24.3 ± 2.440.4 ± 2.7
3g26.2 ± 3.116.4 ± 2.424.9 ± 2.338.2 ± 2.5
3h16.3 ± 3.4−16.7 ± 2.358.6 ± 3.9 **−15.9 ± 2.6
3i26.2 ± 3.5−60.4 ± 2.724.9 ± 3.842.2 ± 2.1
4a−33.6 ± 2.3−36.9±2.024.6 ± 2.98.9 ± 2.6
4b−5.0 ± 1.7−2.7 ± 1.2−27.1 ± 2.756.2 ± 4.0 **
4c−22.8 ± 2.9−6.6 ± 1.416.2 ± 2.651.0 ± 4.7 **
4d17.3 ± 1.3−1.9 ± 4.318.1 ± 2.528.4 ± 2.4
4e61.9 ± 4.2 **−3.7 ± 4.349.5 ± 2.421.9 ± 2.1
4f−22.6 ± 2.9−17.5 ± 4.310.1 ± 2.128.8 ± 2.0
4g−23.3 ± 2.5−14.5 ± 1.72.7 ± 2.336.6 ± 1.9
5a−8.4 ± 1.219.5 ± 1.924.6 ± 2.821.9± 1.2
5b18.7 ± 3.514.0 ± 2.511.4 ± 1.932.9± 0.9
5c19.3 ± 3.115.9 ± 2.721.3 ± 3.031.8 ± 1.2
5d−10.6 ± 3.017.5 ± 2.218.7 ± 3.230.6± 1.8
5e−13.4 ± 4.221.8 ± 2.149.5 ± 2.0 **39.5 ± 1.7
6a4.3 ± 1.611.7 ± 0.928.3 ± 2.241.1 ± 1.9
6b35.7 ± 1.738.2 ± 3.544.7 ± 1.7 **35.2 ± 2.6
Chrysin51.0 ± 4.137.5 ± 1.821.9 ± 1.339.2 ± 2.8
Baicalein17.7 ± 2.229.3 ± 2.119.2 ± 3.528.3±1.8
Quercetin−5.0 ± 3.68.2 ± 3.010.2 ± 3.411.3 ± 1.9
Diosmetin10.9 ± 1.87.3 ± 1.29.2 ± 3.09.5 ± 2.1
Apigenin−1.5 ± 1.95.6 ± 1.16.4 ± 2.18.3 ± 2.5
Luteolin19.1 ± 1.714.7 ± 1.420.4 ± 1.716.5 ± 1.7
Kaempferol−25.3 ± 2.18.6 ± 1.3−24.0 ± 2.414.3 ± 2.2
Note: ** represents p < 0.01, vs. the inhibition of the substrate to the cancer cell lines. The data represented the average of three independent experiments.

Share and Cite

MDPI and ACS Style

Jin, L.; Wang, M.-L.; Lv, Y.; Zeng, X.-Y.; Chen, C.; Ren, H.; Luo, H.; Pan, W.-D. Design and Synthesis of Flavonoidal Ethers and Their Anti-Cancer Activity In Vitro. Molecules 2019, 24, 1749. https://doi.org/10.3390/molecules24091749

AMA Style

Jin L, Wang M-L, Lv Y, Zeng X-Y, Chen C, Ren H, Luo H, Pan W-D. Design and Synthesis of Flavonoidal Ethers and Their Anti-Cancer Activity In Vitro. Molecules. 2019; 24(9):1749. https://doi.org/10.3390/molecules24091749

Chicago/Turabian Style

Jin, Lu, Meng-Ling Wang, Yao Lv, Xue-Yi Zeng, Chao Chen, Hai Ren, Heng Luo, and Wei-Dong Pan. 2019. "Design and Synthesis of Flavonoidal Ethers and Their Anti-Cancer Activity In Vitro" Molecules 24, no. 9: 1749. https://doi.org/10.3390/molecules24091749

Article Metrics

Back to TopTop