Next Article in Journal
Valorization of Phosphorus Secondary Raw Materials by Acidithiobacillus ferrooxidans
Previous Article in Journal
New Roads Leading to Old Destinations: Efflux Pumps as Targets to Reverse Multidrug Resistance in Bacteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Evaluation of New Pyrazoline Derivatives as Potential Anticancer Agents in HepG-2 Cell Line

1
Department of Chemistry, Shantou University Medical College, Shantou 515041, Guangdong, China
2
Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
3
Department of Hepatobiliary Surgery II, Zhujiang Hospital of Southern Medical University, Guangzhou 510280, Guangdong, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 28 February 2017 / Revised: 9 March 2017 / Accepted: 9 March 2017 / Published: 16 March 2017

Abstract

:
Cancer is a major public health concern worldwide. Adverse effects of cancer treatments still compromise patients’ quality of life. To identify new potential anticancer agents, a series of novel pyrazoline derivatives were synthesized and evaluated for cytotoxic effects on HepG-2 (human liver hepatocellular carcinoma cell line) and primary hepatocytes. Compound structures were confirmed by 1H-NMR, mass spectrometry, and infrared imaging. An in vitro assay demonstrated that several compounds exerted cytotoxicity in the micromolar range. Benzo[b]thiophen-2-yl-[5-(4-hydroxy-3,5-dimethoxy-phenyl)-3-(2-hydroxy-phenyl)-4,5-dihydo-pyrazol-1-yl]-methanone (b17) was the most effective anticancer agent against HepG-2 cells owing to its notable inhibitory effect on HepG-2 with an IC50 value of 3.57 µM when compared with cisplatin (IC50 = 8.45 µM) and low cytotoxicity against primary hepatocytes. Cell cycle analysis and apoptosis/necrosis evaluation using this compound revealed that b17 notably arrested HepG-2 cells in the G2/M phase and induced HepG-2 cells apoptosis. Our findings indicate that compound b17 may be a promising anticancer drug candidate.

1. Introduction

Worldwide, liver cancer is the third most common cause of cancer deaths. It is the fifth and seventh most common cancer in men and women, respectively [1]. Cancer is a complex disease caused by various factors, such as high stress, bad dietary habits, aging, and smoking; uncontrolled, rapid, and pathological proliferation of abnormally transformed cells is a direct cause of a large group of diseases [2,3,4]. Great progress has been made in medical treatments, but cancer is still a major cause of mortality. Resistance to chemotherapeutic agents, lack of selectivity, and serious adverse effects are the primary challenges in the fight against cancer [2,3,4,5,6]. Therefore, new anticancer agents are continually developed and tested to selectively destroy tumor cells or at least limit their proliferation.
Pyrazolines are a class of compounds exhibiting a wide spectrum of activities. Several pyrazolines act as anticancer agents [7,8], tubulin assembly inhibitors [9]. Pyrazoloacridine (PZA) (I) (Figure 1) is a new anticancer drug currently undergoing Phase II clinical trials [10,11,12]. Doramapimod (BIRB-796) (II) is a selective p38α mitogen-activated protein kinase (MAPK) inhibitor undergoing Phase III clinical trials [13,14]. Axitinib (AG013736) (III), a vascular endothelial growth factor receptor (VEGFR) inhibitor, used in clinical treatment, is exploited by Pfizer [15,16]. Pazopanib (GW786034) (IV), a VEGFR inhibitor, is exploited by GlaxoSmithKline [17,18]. Tozasertib (VX-680, MK-0457) (V) is an Aurora kinase inhibitor [19,20] and 3-(5′-hydroxymethyl-2’-furyl)-1-benzyl indazole (YC-1) (VI) is a hypoxia-inducible factor (HIF)-1 inhibitor as well as a VEGF inhibitor [21,22]. They are promising anticancer drug candidates. Recent studies report that a large number of sulfonamide derivatives and heterocyclic compounds show antitumor activity and display a common chemical motif of aromatic/heterocyclic sulfonamide. The compounds’ antitumor action is attributed to varying mechanisms, such as cell cycle perturbation, disruption of microtubule assembly, and functional suppression of the transcriptional activator nuclear factor (NF)-Y (a protein that binds to DNA and regulates gene expression by promoting or suppressing transcription) [23,24,25,26]. In this report, we describe the synthesis of new pyrazoline derivatives (b119) with substituted sulfonyl moieties, heterocyclic rings, or other closely related variants in their molecular structures. We show the synthesis and evaluation of a new class of pyrazolines acting as potential antitumor agents against a human liver hepatocellular carcinoma cell line (HepG-2) and primary hepatocytes. We also carried out an analysis of the cell cycle, apoptosis, and necrosis processes using the most effective compound, b17.

2. Results and Discussion

2.1. Synthesis of New Pyrazoline Derivatives

The synthesis of new pyrazoline derivatives (b119) was carried out according to the steps shown in Scheme 1. First, chalcone derivatives (a15) were obtained via the base-catalyzed Claisen-Schmidt condensation with corresponding ketones and aldehydes. Chalcone ring-closure reactions were carried out with hydrazine or phenylhydrazine with tetrabutylammonium bromide (TBAB) as a catalyst to obtain compounds b14, b18, and b19. Next, compound b1 was treated with corresponding acyl chlorides or sulfonyl chlorides at 80 °C with ethanol as a solvent and pyridine as a catalyst to yield compounds b517. Compounds b119 were elucidated by infrared (IR), 1H-NMR, and mass spectrometry imaging. Thus, the synthetic procedure was shown to be versatile and applicable to the preparation of many derivatives. Compounds a15 are shown in Table 1; compounds b119 are shown in Table 2.

2.2. MTT Assay

To identify the most promising antitumor agent among the synthesized pyrazoline derivatives (b119), their cytotoxic effects were tested on HepG-2 cells; cisplatin was used as a positive control. We found that compounds b5, b9, and b1418 displayed IC50 values lower than 50 µM against HepG-2 cells at 48 h. The most effective cytotoxic agent was compound b17, with an IC50 value of 3.57 µM at 48 h; cisplatin, the positive control, had an IC50 value of 8.45 µM at 48 h (Table 3). The pyrazoline derivatives also displayed dose-dependent and time-dependent trends. We selected the three most effective compounds, b1517, along with cisplatin to generate growth-inhibitory curves (Figure 2).

2.3. MTS Assay

According to the results of MTT (thiazolyl blue tetrazolium bromide) assay on HepG-2 cell line, the cytotoxicity of compounds b5, b9, and b1418 were evaluated against primary hepatocytes using the MTS assay. (Isolated primary hepatocytes were unstable. Using the MTS assay, the solubilization steps were eliminated because the MTS formazan product is soluble in tissue culture medium. It can avoid further damage to cells.) Compounds b5, b9, and b1418 showed lower cytotoxicological activity against primary hepatocytes. The IC50 value of compound b17 against primary hepatocytes was 33.47 µM at 48 h. In terms of their anticancer potential, compound b17 can be considered as the most promising anticancer agent against HepG-2 due to its low cytotoxicity against primary hepatocytes (Table 3).

2.4. Cell Cycle Analysis

To determine whether compound b17 would affect the cell cycle in HepG-2 cells, we examined cell cycle progression using flow cytometry (Figure 3). HepG-2 cells were treated with compound b17 at concentrations of 0.9 µM, 2.7 µM, and 4.5 µM for 24 h. A notable decrease in cells in the G1 and S phases was observed. At 4.5 µM, up to 83.01% of the cells were arrested in the G2/M phase (the data shown in Figure 3 is about living cells after exclusion of dead cells). These findings indicate that compound b17 may be a potent anticancer agent.

2.5. Annexin-V Assay

A biparametric cytofluorimetric analysis was performed to determine the mode of cell death induced by compound b17 using propidium iodide (PI) and fluorescent immunolabeling of the protein annexin V. HepG-2 cells were treated with compound b17 at concentrations of 0.9 µM, 2.7 µM, and 4.5 µM for 12 h. The percentage of cell apoptosis was 0.3% at 0 µM for compound b17, and 7.6%, 8.7%, and 10.2% at 0.9 µM, 2.7 µM, and 4.5 µM, respectively (Figure 4). Thus, we conclude that compound b17 can induce apoptosis.

3. Experimental Section

3.1. Chemical Reagents and Equipment

All reagents were purchased from commercial suppliers. Melting points were determined using an Electrothermal 9100 melting point apparatus (Weiss-Gallenkamp, Loughborough, UK). IR spectra were recorded on a Bruker Tensor 27 Fourier IR spectrometer (Bruker, Karlsruhe, Germany). 1H-NMR spectra were recorded on a Bruker Avance 300 spectrometer (Bruker) using tetramethylsilane (TMS) as the internal standard. Mass spectra were recorded on a SCIEX Triple Quad 6500+ LC/MS/MS system (SCIEX, Los Angeles, CA, USA). HRMS (high-resolution mass spectrometry) was performed on a Thermo Scientific Q Exactive (Thermo, Waltham, MA, USA).

3.2. General Procedures for the Synthesis of Compounds a15

Compounds a14 were obtained by a mixture of substituted phenylethanone (0.01 mol), substituted benzaldehyde (0.012 mol), and piperidine (1 mL) as a catalyst, and stirred at 160 °C for 20 min. Next, 30% aqueous sodium hydroxide solution (30 mL) was added and stirred for 15 min [27]. Compound a5 was prepared by appropriate substituted phenylethanone (0.01 mol), substituted benzaldehyde (0.012 mol), and 30% aqueous sodium hydroxide solution (10 mL) in ethanol (30 mL), and stirred at 30 °C for 24 h. The progress of the reaction was checked by thin-layer chromatography (TLC). Upon completion, the reaction mixture was poured onto crushed ice, followed by neutralization with HCl [28]. The precipitated solid was filtered, washed with water, and dried by vacuum pump. The product was finally crystallized from ethanol.

3.3. General Procedures for the Synthesis of Compounds b119

Hydrazine hydrate (0.04 mol, 1.96 g) was added to a solution of compound a1 (0.01 mol, 3.00 g) in ethanol (10 mL). The mixture was refluxed under stirring for 4 h, and the reaction mixture was subsequently poured onto crushed ice; the precipitate was filtered out, and product b1 was later crystallized from ethanol. Compounds b24 were synthesized following this procedure. Compounds b517 were prepared by treating compound b1 (0.01 mol, 3.14 g) with corresponding substituted benzoyl chloride or substituted benzenesulfonyl chloride (0.015 mol) at 80 °C in ethanol as solvent with pyridine as catalyst for 1 h to yield the final N-substituted targeted compounds. The N-phenyl-substituted pyrazolines b18 and b19 were prepared by direct cyclization of a1 and a5, respectively, with phenylhydrazine in the presence of TBAB as a catalyst [29].
4-[5-(2-Hydroxyphenyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b1). Yield: 70.62%; M.p. 140.3–140.7 °C. IR (KBr) νmax (cm−1): 3277 (aromatic –OH), 2994, 2946 (aliphatic C–H asymmetric), 1618, 1498, 1461, 1424 (C=N and C=C), 1352, 1258, 1210, 1130 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.96–3.03 (1H, m, pyrazoline C4–HA), 3.54–3.58 (1H, m, pyrazoline C4–HB), 3.76 (6H, s, –OCH3), 4.74–4.81 (1H, m, pyrazoline C5–Hx), 6.70 (2H, s), 6.88–6.93 (2H, m), 7.24 (1H, t, J = 8.00 Hz), 7.31 (1H, d, J = 8.00 Hz), 7.78 (1H, s, –NH–), 8.31 (1H, s, –OH), 11.21 (1H, s, –OH). MS (ESI) (m/z): 315 [M + H]+.
2,6-Dimethoxy-4-[5-(4-nitrophenyl)-3,4-dihydro-2H-pyrazol-3-yl]-phenol (b2). Yield: 71.22%; M.p. 140.7–141.1 °C. IR (KBr) νmax (cm−1): 3319 (aromatic –OH), 2971, 2844 (aliphatic C–H asymmetric), 1614, 1464, 1430, 1407 (C=N and C=C), 1340, 1223, 1166, 1117 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.89–2.96 (1H, m, pyrazoline C4–HA), 3.43–3.50 (1H, m, pyrazoline C4–HB), 3.74 (6H, s, –OCH3), 4.85–4.91 (1H, m, pyrazoline C5–Hx), 6.65 (2H, s), 7.82 (2H, d, J = 8.00 Hz ), 8.19 (1H, s, –NH–), 8.22 (2H, d, J = 8.00 Hz), 8.32 (1H, s, –OH). MS (ESI) (m/z): 344 [M + H]+.
2-Bromo-4-[5-(2-hydroxyphenyl)-3,4-dihydro-2H-pyrazol-3-yl]-6-methoxy-phenol (b3). Yield: 70.01%; M.p. 181.8–183.9 °C. IR (KBr) νmax (cm−1): 3314 (aromatic –OH), 2966, 2938 (aliphatic C–H asymmetric), 1620, 1499, 1424, 1348 (C=N and C=C), 1262, 1187, 1153, 1049 (C-N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.98–3.05 (1H, m, pyrazoline C4–HA), 3.55–3.61 (1H, m, pyrazoline C4-HB ), 3.83 (3H, s, –OCH3), 4.76–4.81 (1H, m, pyrazoline C5–Hx), 6.88–6.93 (2H, m), 7.04 (1H, s), 7.10 (1H, s), 7.22–7.26 (1H, t, J = 8.00 Hz), 7.30 (1H, d, J = 8.00 Hz), 7.81 (1H, s, –NH–), 9.42 (1H, s, –OH), 11.15 (1H, s, –OH). MS (ESI) (m/z): 363 [M + H]+.
2-Bromo-6-methoxy-4-[5-(4-nitrophenyl)-3,4-dihydro-2H-pyrazol-3-yl]-phenol (b4). Yield: 76.11%; M.p. 208.6–208.9 °C. IR (KBr) νmax (cm−1): 3401, 3325 (aromatic –OH), 2965, 2934 (aliphatic C–H asymmetric), 1614, 1466, 1451, 1416 (C=N and C=C), 1333, 1274, 1187, 1049 (C-N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.91–2.98 (1H, m, pyrazoline C4–HA), 3.44–3.51 (1H, m, pyrazoline C4–HB), 3.82 (3H, s, –OCH3), 4.87–4.93 (1H, m, pyrazoline C5–Hx), 6.99 (1H, s), 7.06 (1H, s), 7.82 (2H, d, J = 8.00 Hz), 8.22 (2H, d, J = 8.00 Hz), 8.24 (1H, s, –NH–), 9.39 (1H, s, –OH). MS (ESI) (m/z): 392 [M + H]+.
4-[2-Benzenesulfonyl-5-(2-hydroxyphenyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b5). Yield: 72.44%; M.p. 163.1–164.4 °C. IR (KBr) νmax (cm−1): 3447, 3194 (aromatic –OH), 2969, 2937 (aliphatic C–H asymmetric), 1619, 1461, 1355, 1219 (C=N and C=C), 1171, 1116, 1056, 1002 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.27–3.32 (1H, m, pyrazoline C4–HA), 3.67–3.72 (1H, m, pyrazoline C4–HB), 3.77 (6H, s, –OCH3), 4.84–4.89 (1H, m, pyrazoline C5–Hx), 6.70 (2H, s), 6.90 (1H, t, J = 8.00 Hz), 6.96 (1H, d, J = 8.00 Hz), 7.35 (1H, t, J = 8.00 Hz), 7.44 (1H, d, J = 8.00 Hz), 7.66 (1H, t, J = 8.00 Hz), 7.74 (2H, t, J = 8.00 Hz), 7.85 (2H, d, J = 8.00 Hz), 8.42 (1H, s, –OH), 10.31 (1H, s, –OH). MS (ESI) (m/z): 455 [M + H]+.
4-[5-(2-Hydroxyphenyl)-2-(2-nitro-benzenesulfonyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b6). Yield: 40.03%; M.p. 151.1–152.8 °C. IR (KBr) νmax (cm−1): 3441, 3211 (aromatic –OH), 2930, 2837 (aliphatic C–H asymmetric), 1619, 1462, 1431, 1376 (C=N and C=C), 1304, 1180, 1113, 1008 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.42–3.44 (1H, m, pyrazoline C4–HA), 3.76 (6H, s, –OCH3), 3.91–3.98 (1H, m, pyrazoline C4–HB), 5.19–5.24 (1H, m, pyrazoline C5–Hx), 6.67 (2H, s), 6.91–6.98 (2H, m), 7.36 (1H, t, J = 8.00 Hz), 7.55 (1H, d, J = 8.00 Hz), 7.84 (1H, t, J = 8.00 Hz), 7.93 (1H, t, J = 8.00 Hz), 7.99–8.03 (2H, m), 8.45 (1H, s, –OH), 10.16 (1H, s, –OH). MS (ESI) (m/z): 500 [M + H]+.
4-[5-(2-Hydroxyphenyl)-2-(3-nitro-benzenesulfonyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b7). Yield: 51.37%; M.p. 131.9–133.5 °C. IR (KBr) νmax (cm−1): 3466, 3198 (aromatic –OH), 2971, 2938 (aliphatic C–H asymmetric), 1619, 1493, 1463, 1428 (C=N and C=C), 1357, 1216, 1177, 1116 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.43–3.46 (1H, m, pyrazoline C4–HA), 3.72 (6H, s, –OCH3), 3.75–3.78 (1H, m, pyrazoline C4–HB), 4.95–4.99 (1H, m, pyrazoline C5–Hx), 6.61 (2H, s), 6.91 (1H, t, J = 8.00 Hz), 6.96 (1H, d, J = 8.00 Hz), 7.35 (1H, d, J = 8.00 Hz), 7.52 (1H, d, J = 8.00 Hz), 7.92 (1H, t, J = 8.00 Hz), 8.21 (1H, d, J = 8.00 Hz), 8.34 (1H, s), 8.45 (1H, s, –OH), 8.51 (1H, d, J = 8.00 Hz), 10.24 (1H, s, –OH). MS (ESI) (m/z): 500 [M + H]+.
4-[5-(2-Hydroxyphenyl)-2-(4-nitro-benzenesulfonyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b8). Yield: 63.51%; M.p. 195.5–197.0 °C. IR (KBr) νmax (cm−1): 3385 (aromatic –OH), 2969, 2846 (aliphatic C–H asymmetric), 1620, 1464, 1430, 1368 (C=N and C=C), 1314, 1207, 1065, 1012 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.41–3.47 (1H, m, pyrazoline C4–HA), 3.73 (6H, s, –OCH3), 3.74–3.79 (1H, m, pyrazoline C4–HB), 4.90–4.95 (1H, m, pyrazoline C5–Hx), 6.63 (2H, s), 6.90 (1H,t, J = 8.00 Hz), 6.95 (1H, d, J = 8.00 Hz), 7.34 (1H, t, J = 8.00 Hz), 7.51 (1H, d, J = 8.00 Hz), 8.04 (2H, d, J = 8.00 Hz), 8.41 (1H, d, J = 8.00 Hz), 8.45 (1H, s, –OH), 10.22 (1H, s, –OH). MS (ESI) (m/z): 500 [M + H]+.
4-[5-(2-Hydroxyphenyl)-2-(toluene-4-sulfonyl)-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b9). Yield: 71.43%; M.p. 194.6–194.7 °C. IR (KBr) νmax (cm−1): 3453, 3192 (aromatic –OH), 2969, 2938 (aliphatic C–H asymmetric), 1615, 1493, 1462, 1429 (C=N and C=C), 1358, 1218, 1115, 1026 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.38 (3H, s, –CH3), 3.41–3.44 (1H, m, pyrazoline C4–HA), 3.67–3.75 (1H, m, pyrazoline C4–HB), 3.76 (6H, s, –OCH3), 4.77–4.83 (1H, m, pyrazoline C5–Hx), 6.69 (2H, s), 6.90 (1H, t, J = 8.00 Hz), 6.96 (1H, d, J = 8.00 Hz), 7.34 (1H, t, J = 8.00 Hz), 7.42–7.46 (3H, m), 7.72 (2H, d, J = 8.00 Hz), 8.41 (1H, s, –OH), 10.33 (1H, s, –OH). MS (ESI) (m/z): 469 [M + H]+.
[5-(4-Hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-(2-nitrophenyl)-methanone (b10). Yield: 71.43%; M.p. 194.6–194.7 °C. IR (KBr) νmax (cm−1): 3532, 3190 (aromatic –OH), 2936, 2836 (aliphatic C–H asymmetric), 1666 (C=O), 1618, 1524, 1441, 1342 (C=N and C=C), 1253, 1224, 1146, 1106 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.32–3.34 (1H, m, pyrazoline C4–HA), 3.78 (6H, s, –OCH3), 3.96–4.06 (1H, m, pyrazoline C4–HB), 5.56–5.60 (1H, m, pyrazoline C5–Hx), 6.62 (2H, s), 6.84–6.88 (2H, m), 7.29 (1H, t, J = 8.00 Hz), 7.38 (1H, d, J = 8.00 Hz), 7.70 (1H, d, J = 8.00 Hz), 7.77 (1H, t, J = 8.00 Hz), 7.90 (1H, t, J = 8.00 Hz), 8.20 (1H, s), 8.37 (1H, s, –OH), 9.79 (1H, s, –OH). MS (ESI) (m/z): 464 [M + H]+.
[5-(4-Hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-(3-nitrophenyl)-methanone (b11). Yield: 70.03%; M.p. 207.7–209.3 °C. IR (KBr) νmax (cm−1): 3212, 3078 (aromatic –OH), 2930, 2840 (aliphatic C–H asymmetric), 1667 (C=O), 1610, 1525, 1425, 1341 (C=N and C=C), 1310, 1214, 1159, 1113 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.35–3.37 (1H, m, pyrazoline C4–HA), 3.75 (6H, s, –OCH3), 3.97–4.05 (1H, m, pyrazoline C4–HB), 5.63–5.67 (1H, m, pyrazoline C5–Hx), 6.63 (2H, s), 6.90–6.95 (2H, m), 7.33 (1H, t, J = 8.00 Hz), 7.56 (1H, d, J = 8.00 Hz), 7.82 (1H, t, J = 8.00 Hz), 8.28–8.30 (1H, m), 8.40–8.42 (2H, m), 8.62 (1H, s), 10.03 (1H, s, –OH). MS (ESI) (m/z): 464 [M + H]+.
[5-(4-Hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-(4-nitrophenyl)-methanone (b12). Yield: 68.02%; M.p. 250.1–251.2 °C. IR (KBr) νmax (cm−1): 3378, 3266 (aromatic –OH), 2936, 2842 (aliphatic C–H asymmetric), 1650 (C=O), 1614, 1518, 1444, 1336 (C=N and C=C), 1258, 1243, 1210, 1110 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.40–3.46 (1H, m, pyrazoline C4–HA), 3.76 (6H, s, –OCH3), 3.98–4.05 (1H, m, pyrazoline C4–HB), 5.65–5.66 (1H, m, pyrazoline C5–Hx), 6.62 (2H, s), 6.90–6.94 (2H, m), 7.34 (1H, t, J = 8.00 Hz), 7.54 (1H, d, J = 8.00 Hz), 8.05 (2H, d, J = 8.00 Hz), 8.36 (2H, d, J = 8.00 Hz), 8.40 (1H, s, –OH), 10.03 (1H, s, –OH). MS (ESI) (m/z): 464 [M + H]+.
(3,5-Dinitrophenyl)-[5-(4-hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-methanone (b13). Yield: 51.38%; M.p. 240.4–240.9 °C. IR (KBr) νmax (cm−1): 3336, 3109 (aromatic –OH), 2939, 2843 (aliphatic C–H asymmetric), 1647 (C=O), 1614, 1543, 1465, 1432 (C=N and C=C), 1341, 1219, 1112, 1031 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.41–3.43 (1H, m, pyrazoline C4–HA), 3.75 (6H, s, –OCH3), 3.98–4.06 (1H, m, pyrazoline C4–HB), 5.63–5.67 (1H, m, pyrazoline C5–Hx), 6.64 (2H, s), 6.90 (1H, t, J = 8.00 Hz), 6.96 (1H, d, J = 8.00 Hz), 7.33 (1H, t, J = 8.00 Hz), 7.65 (1H, d, J = 8.00 Hz), 8.39 (1H, s, –OH), 8.97 (1H, s), 9.07 (2H, s), 10.06 (1H, s, –OH). MS (ESI) (m/z): 509 [M + H]+.
[5-(4-Hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-naphthalen-2-yl-methanone (b14). Yield: 63.51%; M.p. 185.5–186.5 °C. IR (KBr) νmax (cm−1): 3421 (aromatic –OH), 2931, 2835 (aliphatic C–H asymmetric), 1635 (C=O), 1616, 1522, 1465, 1426 (C=N and C=C), 1328, 1218, 1118, 1031 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.39–3.44 (1H, m, pyrazoline C4–HA), 3.75 (6H, s, –OCH3), 3.98–4.05 (1H, m, pyrazoline C4–HB), 5.67–5.71 (1H, m, pyrazoline C5–Hx), 6.65 (2H, s), 6.89–6.94 (2H, m), 7.30–7.34 (1H, m), 7.49–7.52 (1H, m), 7.60–7.67 (2H, m), 7.86–7.88 (1H, m), 8.01–8.04 (2H, m), 8.07 (1H, d, J = 8.00 Hz), 8.38 (1H, s), 8.41 (1H, s, –OH), 10.13 (1H, s, –OH). MS (ESI) (m/z): 469 [M + H]+.
[5-(4-Hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-thiophen-2-yl-methanone (b15). Yield: 63.41%; M.p. 196.1–197.2 °C. IR (KBr) νmax (cm−1): 3375, 3199 (aromatic –OH), 2942, 2836 (aliphatic C–H asymmetric), 1632 (C=O), 1612, 1516, 1435, 1336 (C=N and C=C), 1245, 1209, 1116, 1043 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.32–3.33 (1H, m, pyrazoline C4–HA), 3.69 (6H, s, –OCH3), 3.93–4.00 (1H, m, pyrazoline C4–HB), 5.57–5.61 (1H, m, pyrazoline C5–Hx), 6.50 (2H, s), 6.94–6.99 (2H, m), 7.20–7.23 (1H, m), 7.33–7.37 (1H, m), 7.78–7.80 (1H, m), 7.92–7.93 (1H, m), 7.99–8.00 (1H, m), 8.37 (1H, s, –OH), 10.19 (1H, s, –OH). MS (ESI) (m/z): 425 [M + H]+.
Furan-2-yl-[5-(4-hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-methanone (b16). Yield: 63.41%; M.p. 201.1–203.7 °C. IR (KBr) νmax (cm−1): 3355, 3128 (aromatic –OH), 2940, 2837 (aliphatic C–H asymmetric), 1620 (C=O), 1600, 1519, 1458, 1426 (C=N and C=C), 1340, 1248, 1209, 1117 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.29–3.34 (1H, m, pyrazoline C4–HA), 3.70 (6H, s, –OCH3), 3.88–3.97 (1H, m, pyrazoline C4–HB), 5.57–5.61 (1H, m, pyrazoline C5–Hx), 6.51 (2H, s), 671–6.72 (1H, m), 6.93–7.00 (2H, m), 7.35 (1H, t, J = 8.00 Hz), 7.51–7.52 (1H, m), 7.64 (1H, d, J = 8.00 Hz), 7.96 (1H, s), 8.36 (1H, s, –OH), 10.47 (1H, s, –OH). MS (ESI) (m/z): 409 [M + H]+.
Benzo[b]thiophen-2-yl-[5-(4-hydroxy-3,5-dimethoxyphenyl)-3-(2-hydroxyphenyl)-4,5-dihydro-pyrazol-1-yl]-methanone (b17). Yield: 66.67%; M.p. 169.2–170.1 °C. IR (KBr) νmax (cm−1): 3392, 3262 (aromatic –OH), 2943, 2842 (aliphatic C–H asymmetric), 1690 (C=O), 1616, 1514, 1440, 1376 (C=N and C=C), 1338, 1237, 1113, 1049 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.39–3.40 (1H, m, pyrazoline C4–HA), 3.70 (6H, s, –OCH3), 3.96–4.03 (1H, m, pyrazoline C4–HB), 5.63–5.67 (1H, m, pyrazoline C5–Hx), 6.53 (2H, s), 6.98–7.01 (2H, m), 7.35–7.39 (1H, m), 7.44–7.52 (2H, m), 7.85–7.87 (1H, m), 8.03–8.08 (2H, m), 8.37 (1H, s), 8.40 (1H, s, –OH), 10.23 (1H, s, –OH). 13C-NMR (600 MHz, δ ppm, DMSO-d6): 40.41, 44.01, 56.02, 103.17, 116.72, 117.00, 119.64, 122.54, 124.85, 125.49, 126.63, 129.30, 130.80, 132.04, 132.07, 135.01, 135.23, 137.92, 141.85, 148.11, 156.77, 156.86, 158.08. MS (ESI) (m/z): 475 [M + H]+. HRMS: m/z [M − H]¯ calcd for C26H22N2O5S: 473.1177; found: 474.1249.
4-[5-(2-Hydroxyphenyl)-2-phenyl-3,4-dihydro-2H-pyrazol-3-yl]-2,6-dimethoxy-phenol (b18). Yield: 48.46%; M.p. 169.7–170.8 °C. IR (KBr) νmax (cm−1): 3400 (aromatic –OH), 2939, 2842 (aliphatic C–H asymmetric), 1493, 1464, 1431 (C=N and C=C), 1373, 1245, 1113, 1026 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 3.25–3.30 (1H, m, pyrazoline C4–HA), 3.70 (6H, s, –OCH3), 4.00–4.07 (1H, m, pyrazoline C4-HB), 5.25–5.29 (1H, m, pyrazoline C5–Hx), 6.63 (2H, s), 6.81 (1H, t, J = 8.00 Hz), 6.92–7.01 (4H, m), 7.23 (2H, t, J = 8.00 Hz), 7.30 (1H, t, J = 8.00 Hz), 7.44 (1H, d, J = 8.00 Hz), 8.39 (1H, s, –OH), 10.62 (1H, s, –OH). MS (ESI) (m/z): 391 [M + H]+.
1-Phenyl-3-p-tolyl-5-(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazole (b19). Yield: 50.39%; M.p. 137.9–140.3 °C. IR (KBr) νmax (cm−1): 2934, 2834 (aliphatic C–H asymmetric), 1495, 1462, 1418 (C=N and C=C), 1348, 1319, 1235, 1125 (C–N). 1H-NMR (400 MHz, δ ppm, DMSO-d6): 2.34 (3H, s, –CH3), 3.09–3.15 (1H, m, pyrazoline C4–HA), 3.63 (3H, s, –OCH3), 3.70 (6H, s, –OCH3), 3.83–3.91 (1H, m, pyrazoline C4–HB), 5.29–5.33 (1H, m, pyrazoline C5-Hx), 6.62 (2H, s), 6.74 (1H, t, J = 8.00 Hz), 7.04 (2H, d, J = 8.00 Hz), 7.18 (2H, t, J = 8.00 Hz), 7.25 (2H, d, J = 8.00 Hz), 7.65 (2H, d, J = 8.00 Hz). MS (ESI) (m/z): 403 [M + H]+.
b1–19 1H-NMR, b17 13H-NMR and b17 HRMS are as provided as Supplementary Materials.

3.4. Pharmacology

3.4.1. Cell Culture and Treatment

Primary hepatocytes were isolated from male SD rats weighing 250–300 g using a modified in situ collagenase perfusion method and then the cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) with 2 g/L bovine serum albumin (BSA) and 50 mg/L gentamicin. HepG-2 cells were incubated in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin–streptomycin liquid. Cells were incubated at 37 °C in a humidified atmosphere of 95% air and 5% CO2. Stock solutions of compounds and cisplatin were prepared in dimethyl sulfoxide (DMSO), then added to fresh culture medium to obtain final concentrations.

3.4.2. MTT Assay

To evaluate the compounds’ cytotoxicity, 50 µM and 100 µM concentrations of each compound were used to target the more effective compounds for further study. Next, compounds b14 and b18 were prepared in five different concentrations (20 µM, 40 µM, 60 µM, 80 µM, 100 µM), and compounds b15, b16, and b17, along with cisplatin, were prepared in seven different concentrations (2.5 µM, 5 µM, 7.5 µM, 10 µM, 20 µM, 40 µM, 60 µM). A suspension of growing cells (50,000/mL) was prepared, and 100 µL/well dispensed into 96-well plates, yielding a density of 5000 cells/well. The cells were incubated for 24 h before adding the pyrazoline derivatives. The optimal cell number for cytotoxicity assays was determined in preliminary experiments. At the end of the incubation period, the medium was removed and 100 µL of different concentrations of pyrazoline derivatives were added to wells for 24 h and 48 h. Following the exposure period, 20 µL of MTT (5 mg/mL) were added to cells and incubated for a further 4 h at 37 °C. The medium was removed and the formazan crystals were solubilized by adding 150 µL DMSO to each well. After 10 min of shaking, the absorbance was measured at 550 nm using a FilterMax F5 Multi-Mode Microplate Reader (Energy Chemical, Shanghai, China). Cells were treated with each compound with four replicates per concentration, and each experiment was conducted at least three times. Dose–inhibition rate curves and IC50 values, defined as the drug concentrations which reduced absorbance to 50% of control values, were then generated.

3.4.3. MTS Assay

The cytotoxicological activity of compounds b5, b9, and b1418 were evaluated against primary hepatocytes using the MTS assay (Cell Titer 96® Aqueous Cell Proliferation Assay, Promega, Cat. No. G5421) as a fast and sensitive quantification of cell proliferation and viability. The concentrations of compounds were prepared in seven different concentrations (2.5 µM, 5 µM, 10 µM, 20 µM, 40 µM, 80 µM, 160 µM). Cells were seeded into 96-well plates at a density of 26,000 cells/well. The plates were incubated for 24 h prior to any treatment. At the end of this period, the medium was removed and 100 µL of different concentrations of pyrazoline derivatives were added to wells for 48 h. Following the exposure period, 20 μL of the combined MTS/PMS solution was added to cells and incubated for a further 2 h at 37 °C, then the absorbance was recorded at 490 nm using an ELISA Plate Reader (ELISA, Emeryville, CA, USA). The values of IC50, the effective concentration at which 50% of the primary hepatocytes were inhibited, were calculated to evaluate the cytotoxic activities.

3.4.4. Cell Cycle Analysis

Based on the results of the cytotoxicity assay, compound b17 was selected for further mechanistic study with HepG-2 cells. For flow cytometry analysis of DNA content, approximately 1.5 × 105 HepG-2 cells/well in exponential growth mode were plated in 6-well plates and allowed to adhere, then treated with different concentrations of compound b17 for 24 h. After incubation, the cells were collected, centrifuged, and fixed with ice-cold ethanol (70%). The cells were treated with RNase A and stained with PI. Samples were analyzed on a BD Accuri C6 flow cytometer (BD, New York, NY, USA). DNA histograms were analyzed using ModFit for Windows.

3.4.5. Annexin-V Assay

Approximately 1.5 × 105 HepG-2 cells/well were plated in 6-well plates and allowed to adhere. After 24 h, the medium was replaced with fresh culture medium containing compound b17 at final concentrations of 0 µM, 0.9 µM, 2.7 µM, and 4.5 µM. Cells were harvested after 12 h. The cells were trypsinized, washed in phosphate-buffered saline (PBS), and centrifuged at 1000 rpm for 5 min. The cells were then resuspended in 200 µL of staining solution (containing 10 µL PI and 5 µL Annexin V–PE in binding buffer), mixed gently, and incubated for 15 min at 25 °C in the dark. The samples were later analyzed with a BD Accuri™ C6 flow cytometer.

4. Conclusions

In the present study, we synthesized 19 new pyrazoline derivatives and investigated their antiproliferative effects on HepG-2 cells. We found that compound b17 was the most effective anticancer agent following a 48 h exposure with an IC50 value of 3.57 µM compared with the cisplatin value of 8.45 µM. Compound b17 was therefore selected for cell cycle analysis and apoptosis/necrosis evaluation. We observed that HepG-2 cells treated with compound b17 could be arrested in the G2/M phase. In addition, compound b17 can be regarded as a potent inducer of apoptosis in the cells. These results provide an important foundation for further development of compound b17 as a potent antitumor agent. We also found that compounds with a heterocyclic ring, such as b15 and b16, exhibited better pharmacological activity than most other pyrazoline derivatives synthesized in this study. Compounds b15 and b16 will be tested further to improve characterization of their antitumor activity.

Supplementary Materials

Supplementary materials are available online.

Acknowledgments

This study was funded by the Guangdong Natural Science Foundation of China (No. 9351503102000001).

Author Contributions

Weijie Xu, Ying Pan, and Hong Wang designed the research. Weijie Xu performed the synthesis work. Jinhong Zheng and Ying Pan were responsible for the direction of the biological research. Weijie Xu, Duncan Wei, Haiyan Li, Qing Peng and Cheng Chen performed the anticancer activity assays. Weijie Xu wrote the manuscript. All authors discussed, edited, and approved the final version.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Poustchi, H.; Sepanlou, S.; Esmaili, S.; Mehrabi, N.; Ansarymoghadam, A. Hepatocellular carcinoma in the world and the Middle East. Middle East J. Dig. Dis. 2010, 2, 31–41. [Google Scholar] [PubMed]
  2. Nepali, K.; Sharma, S.; Sharma, M.; Bedi, P.M.; Dhar, K.L. Rational approaches, design strategies, structure activity relationship and mechanistic insights for anticancer hybrids. Eur. J. Med. Chem. 2014, 77, 422–487. [Google Scholar] [CrossRef] [PubMed]
  3. Rebucci, M.; Michiels, C. Molecular aspects of cancer cell resistance to chemotherapy. Biochem. Pharmacol. 2013, 85, 1219–1226. [Google Scholar] [CrossRef] [PubMed]
  4. Nussbaumer, S.; Bonnabry, P.; Veuthey, J.L.; Fleury-Souverain, S. Analysis of anticancer drugs: A review. Talanta 2011, 85, 2265–2289. [Google Scholar] [CrossRef] [PubMed]
  5. Kerbel, R.S. Molecular and physiologic mechanisms of drug resistance in cancer: An overview. Cancer Metastasis Rev. 2001, 20, 1–2. [Google Scholar] [CrossRef] [PubMed]
  6. Medina, P.P.; Slack, F.J. microRNAs and cancer: An overview. Cell Cycle 2008, 7, 2485–2492. [Google Scholar] [CrossRef] [PubMed]
  7. Karabacak, M.; Altıntop, M.D.; İbrahim Çiftçi, H.; Koga, R.; Otsuka, M.; Fujita, M.; Özdemir, A. Synthesis and Evaluation of New Pyrazoline Derivatives as Potential Anticancer Agents. Molecules 2015, 20, 19066–19084. [Google Scholar] [CrossRef] [PubMed]
  8. Montoya, A.; Quiroga, J.; Abonia, R.; Nogueras, M.; Cobo, J.; Insuasty, B. Synthesis and in vitro antitumor activity of a novel series of 2-pyrazoline derivatives bearing the 4-aryloxy-7-chloroquinoline fragment. Molecules 2014, 19, 18656–18675. [Google Scholar] [CrossRef] [PubMed]
  9. Qin, Y.J.; Li, Y.J.; Jiang, A.Q.; Yang, M.R.; Zhu, Q.Z.; Dong, H.; Zhu, H.L. Design, synthesis and biological evaluation of novel pyrazoline-containing derivatives as potential tubulin assembling inhibitors. Eur. J. Med. Chem. 2015, 94, 447–457. [Google Scholar] [CrossRef] [PubMed]
  10. Dees, E.C.; Rowinsky, E.K.; Noe, D.A.; O’Reilly, S.; Adjei, A.A.; Elza-Brown, K.; Donehower, R.C. A phase I and pharmacologic study of pyrazoloacridine and cisplatin in patients with advanced cancer. Investig. New Drugs 2003, 21, 75–84. [Google Scholar] [CrossRef]
  11. Berg, S.L.; Blaney, S.M.; Sullivan, J.; Bernstein, M.; Dubowy, R.; Harris, M.B. Phase II trial of pyrazoloacridine in children with solid tumors: A Pediatric Oncology Group phase II study. J. Pediatr. Hematol. Oncol. 2000, 22, 506–509. [Google Scholar] [CrossRef] [PubMed]
  12. Ramaswamy, B.; Mrozek, E.; Kuebler, J.P.; Bekaii-Saab, T.; Kraut, E.H. Phase II trial of pyrazoloacridine (NSC#366140) in patients with metastatic breast cancer. Investig. New Drugs 2011, 29, 347–351. [Google Scholar]
  13. Jin, X.; Mo, Q.; Zhang, Y.; Gao, Y.; Wu, Y.; Li, J.; Hao, X.; Ma, D.; Gao, Q.; Chen, P. The p38 MAPK inhibitor BIRB796 enhances the antitumor effects of VX680 in cervical cancer. Cancer Biol. Ther. 2016, 17, 566–576. [Google Scholar] [CrossRef] [PubMed]
  14. Kuma, Y.; Sabio, G.; Bain, J.; Shpiro, N.; Márquez, R.; Cuenda, A. BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J. Biol. Chem. 2005, 280, 19472–19479. [Google Scholar] [CrossRef] [PubMed]
  15. George, D.J. Phase 2 studies of sunitinib and AG013736 in patients with cytokine-refractory renal cell carcinoma. Clin. Cancer Res. 2007, 13, 753–757. [Google Scholar] [CrossRef] [PubMed]
  16. Rini, B.; Wilding, G.; Hudes, G.; Stadler, W.M.; Kim, S.; Tarazi, J.; Bycott, P.; Liau, K.; Dutcher, J. Axitinib (AG-013736; AG) in patients (pts) with metastatic clear cell renal cell cancer (RCC) refractory to sorafenib. EJC Suppl. 2007, 5, 300–304. [Google Scholar] [CrossRef]
  17. Kumar, R.; Knick, V.B.; Rudolph, S.K.; Johnson, J.H.; Crosby, R.M.; Crouthamel, M.C.; Hopper, T.M.; Miller, C.G.; Harrington, L.E.; Onori, J.A.; et al. 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] [PubMed]
  18. Ranieri, G.; Mammi, M.; di Paola, E.D.; Russo, E.; Gallelli, L.; Citraro, R.; Gadaleta, C.D.; Marech, I.; Ammendola, M.; de Sarro, G. Pazopanib a tyrosine kinase inhibitor with strong anti-angiogenetic activity: A new treatment for metastatic soft tissue sarcoma. Crit. Rev. Oncol. Hematol. 2014, 89, 322–329. [Google Scholar] [CrossRef] [PubMed]
  19. Pezzani, R.; Rubin, B.; Bertazza, L.; Redaelli, M.; Barollo, S.; Monticelli, H.; Baldini, E.; Mian, C.; Mucignat, C.; Scaroni, C.; et al. The aurora kinase inhibitor VX-680 shows anti-cancer effects in primary metastatic cells and the SW13 cell line. Investig. New Drugs 2016, 34, 531–540. [Google Scholar] [CrossRef] [PubMed]
  20. Giles, F.J.; Swords, R.T.; Nagler, A.; Hochhaus, A.; Ottmann, O.G.; Rizzieri, D.A.; Talpaz, M.; Clark, J.; Watson, P.; Xiao, A.; et al. MK-0457, an Aurora kinase and BCR-ABL inhibitor, is active in patients with BCR-ABL T315I leukemia. Leukemia 2013, 27, 113–117. [Google Scholar] [CrossRef] [PubMed]
  21. Na, J.I.; Na, J.Y.; Choi, W.Y.; Lee, M.C.; Park, M.S.; Choi, K.H.; Lee, J.K.; Kim, K.T.; Park, J.T.; Kim, H.S. The HIF-1 inhibitor YC-1 decreases reactive astrocyte formation in a rodent ischemia model. Am. J. Transl. Res. 2015, 7, 751–760. [Google Scholar] [PubMed]
  22. Chang, L.C.; Lin, H.Y.; Tsai, M.T.; Chou, R.H.; Lee, F.Y.; Teng, C.M.; Hsieh, M.T.; Hung, H.Y.; Huang, L.J.; Yu, Y.L.; et al. YC-1 inhibits proliferation of breast cancer cells by down-regulating EZH2 expression via activation of c-Cbl and ERK. Br. J. Pharmacol. 2014, 171, 4010–4025. [Google Scholar] [CrossRef] [PubMed]
  23. Scozzafava, A.; Owa, T.; Mastrolorenzo, A.; Supuran, C.T. Anticancer and antiviral sulfonamides. Curr. Med. Chem. 2003, 10, 925–953. [Google Scholar] [CrossRef] [PubMed]
  24. Bashir, R.; Ovais, S.; Yaseen, S.; Hamid, H.; Alam, M.S.; Samim, M.; Singh, S.; Javed, K. Synthesis of some new 1,3,5-trisubstituted pyrazolines bearing benzene sulfonamide as anticancer and anti-inflammatory agents. Bioorg. Med. Chem. Lett. 2011, 21, 4301–43055. [Google Scholar] [CrossRef] [PubMed]
  25. Casini, A.; Scozzafava, A.; Mastrolorenzo, A.; Supuran, L.T. Sulfonamides and sulfonylated derivatives as anticancer agents. Curr. Cancer Drug Targets 2002, 2, 55–75. [Google Scholar] [CrossRef] [PubMed]
  26. Pingaew, R.; Prachayasittikul, V.; Worachartcheewan, A.; Nantasenamat, C.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Novel 1,4-naphthoquinone-based sulfonamides: Synthesis, QSAR, anticancer and antimalarial studies. Eur. J. Med. Chem. 2015, 20, 446–459. [Google Scholar] [CrossRef] [PubMed]
  27. Ying, P.; Chen, Y.; Yu, X.; Wang, J.; Zhang, L.; He, Y.; Zheng, J. The synthesis of a novel chalcone and evaluation for anti-free radical activity and antagonizing the learning impairments in Alzheimer’s model. Cell Physiol. Biochem. 2012, 29, 949–958. [Google Scholar]
  28. De Vasconcelos, A.; Campos, V.F.; Nedel, F.; Seixas, F.K.; Dellagostin, O.A.; Smith, K.R.; de Pereira, C.M.; Stefanello, F.M.; Collares, T.; Barschak, A.G. Cytotoxic and apoptotic effects of chalcone derivatives of 2-acetyl thiophene on human colon adenocarcinoma cells. Cell Biochem. Funct. 2013, 31, 289–297. [Google Scholar] [CrossRef] [PubMed]
  29. Havrylyuk, D.; Roman, O.; Lesyk, R. Synthetic approaches, structure activity relationship and biological applications for pharmacologically attractive pyrazole/pyrazoline-thiazolidine-based hybrids. Eur. J. Med. Chem. 2016, 113, 145–166. [Google Scholar] [CrossRef] [PubMed]
  • Sample Availability: Samples of compounds b119 are available from the authors.
Figure 1. Structures of pyrazoloacridine (PZA) (I); doramapimod (BIRB-796) (II); axitinib (AG013736) (III); pazopanib (GW786034) (IV); tozasertib (VX-680) (V); and 3-(5’-hydroxymethyl-2’-furyl)-1-benzyl indazole (YC-1) (VI).
Figure 1. Structures of pyrazoloacridine (PZA) (I); doramapimod (BIRB-796) (II); axitinib (AG013736) (III); pazopanib (GW786034) (IV); tozasertib (VX-680) (V); and 3-(5’-hydroxymethyl-2’-furyl)-1-benzyl indazole (YC-1) (VI).
Molecules 22 00467 g001
Scheme 1. General synthesis of compounds b119. Reagents and conditions: (i) substituted phenylethanone (0.01 mol), substituted benzaldehyde (0.012 mol), piperidine (1 mL), 160 °C, 20 min or 30% NaOH, ethanol, r.t., 24 h; (ii) ethanol, hydrazine hydrate, reflux, 4 h; (iii) substituted benzoyl chloride or substituted benzenesulfonyl chloride, 80 °C, ethanol, pyridine, 1 h; (iv) ethanol, phenylhydrazine, tetrabutylammonium bromide (TBAB), reflux, 1 h.
Scheme 1. General synthesis of compounds b119. Reagents and conditions: (i) substituted phenylethanone (0.01 mol), substituted benzaldehyde (0.012 mol), piperidine (1 mL), 160 °C, 20 min or 30% NaOH, ethanol, r.t., 24 h; (ii) ethanol, hydrazine hydrate, reflux, 4 h; (iii) substituted benzoyl chloride or substituted benzenesulfonyl chloride, 80 °C, ethanol, pyridine, 1 h; (iv) ethanol, phenylhydrazine, tetrabutylammonium bromide (TBAB), reflux, 1 h.
Molecules 22 00467 sch001
Figure 2. Inhibitory effects of compounds b1517 and cisplatin on HepG-2 cells after 24 h and 48 h.
Figure 2. Inhibitory effects of compounds b1517 and cisplatin on HepG-2 cells after 24 h and 48 h.
Molecules 22 00467 g002
Figure 3. Effects of compound b17 on cell cycle progression in HepG-2 cells. Cells were treated with b17 at concentrations of 0 µM, 0.9 µM, 2.7 µM, and 4.5 µM for 24 h.
Figure 3. Effects of compound b17 on cell cycle progression in HepG-2 cells. Cells were treated with b17 at concentrations of 0 µM, 0.9 µM, 2.7 µM, and 4.5 µM for 24 h.
Molecules 22 00467 g003
Figure 4. Compound b17 induced apoptosis in HepG-2 cells at concentrations of control, 0.9 µM, 2.7 µM, and 4.5 µM in a 12 h exposure. PI: propidium iodide.
Figure 4. Compound b17 induced apoptosis in HepG-2 cells at concentrations of control, 0.9 µM, 2.7 µM, and 4.5 µM in a 12 h exposure. PI: propidium iodide.
Molecules 22 00467 g004
Table 1. Structure of the compounds a15.
Table 1. Structure of the compounds a15.
CompoundsR1R2R3R4R5
a1–OCH3–OH–OCH3–OHH
a2–OCH3–OH–OCH3H–NO2
a3–Br–OH–OCH3–OHH
a4–Br–OH–OCH3H–NO2
a5–OCH3–OCH3–OCH3H–CH3
Table 2. Structure of the compounds b119.
Table 2. Structure of the compounds b119.
CompoundsR1R2R3R4R5R6
b1–OCH3–OH–OCH3–OHHH
b2–OCH3–OH–OCH3H–NO2H
b3–Br–OH–OCH3–OHHH
b4–Br–OH–OCH3H–NO2H
b5–OCH3–OH–OCH3–OHH Molecules 22 00467 i001
b6–OCH3–OH–OCH3–OHH Molecules 22 00467 i002
b7–OCH3–OH–OCH3–OHH Molecules 22 00467 i003
b8–OCH3–OH–OCH3–OHH Molecules 22 00467 i004
b9–OCH3–OH–OCH3–OHH Molecules 22 00467 i005
b10–OCH3–OH–OCH3–OHH Molecules 22 00467 i006
b11–OCH3–OH–OCH3–OHH Molecules 22 00467 i007
b12–OCH3–OH–OCH3–OHH Molecules 22 00467 i008
b13–OCH3–OH–OCH3–OHH Molecules 22 00467 i009
b14–OCH3–OH–OCH3–OHH Molecules 22 00467 i010
b15–OCH3–OH–OCH3–OHH Molecules 22 00467 i011
b16–OCH3–OH–OCH3–OHH Molecules 22 00467 i012
b17–OCH3–OH–OCH3–OHH Molecules 22 00467 i013
b18–OCH3–OH–OCH3–OHH Molecules 22 00467 i014
b19–OCH3–OCH3–OCH3–H–CH3 Molecules 22 00467 i015
Table 3. The cytotoxic effects of the compounds b119 on HepG-2 cell line and primary hepatocytes.
Table 3. The cytotoxic effects of the compounds b119 on HepG-2 cell line and primary hepatocytes.
CompoundsIC50/μmol·L−1CompoundsIC50/μmol·L−1
HepG-2Primary HepatocytesHepG-2Primary Hepatocytes
b1>50b12>50
b2>50b13>50
b3>50b1417.99 ± 1.3722.65 ± 1.21
b4>50b154.51 ± 1.4919.24 ± 0.08
b528.76 ± 1.3235.13 ± 2.21b164.61 ± 1.2720.73 ± 1.72
b6>50b173.57 ± 1.3933.47 ± 2.33
b7>50b1828.47 ± 1.3450.71 ± 3.21
b8>50b19>50
b912.01 ± 1.8329.66 ± 2.43Cisplatin8.45 ± 1.05
b10>50
b11>50
“—”: Not determined.

Share and Cite

MDPI and ACS Style

Xu, W.; Pan, Y.; Wang, H.; Li, H.; Peng, Q.; Wei, D.; Chen, C.; Zheng, J. Synthesis and Evaluation of New Pyrazoline Derivatives as Potential Anticancer Agents in HepG-2 Cell Line. Molecules 2017, 22, 467. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22030467

AMA Style

Xu W, Pan Y, Wang H, Li H, Peng Q, Wei D, Chen C, Zheng J. Synthesis and Evaluation of New Pyrazoline Derivatives as Potential Anticancer Agents in HepG-2 Cell Line. Molecules. 2017; 22(3):467. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22030467

Chicago/Turabian Style

Xu, Weijie, Ying Pan, Hong Wang, Haiyan Li, Qing Peng, Duncan Wei, Cheng Chen, and Jinhong Zheng. 2017. "Synthesis and Evaluation of New Pyrazoline Derivatives as Potential Anticancer Agents in HepG-2 Cell Line" Molecules 22, no. 3: 467. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules22030467

Article Metrics

Back to TopTop