Sie sind auf Seite 1von 12

European Journal of Medicinal Chemistry 157 (2018) 139e150

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Design, synthesis and pharmacological evaluation of new 3-(1H-


benzimidazol-2-yl)quinolin-2(1H)-one derivatives as potential
antitumor agents
Wen-Bin Kuang a, b, 1, Ri-Zhen Huang c, 1, Jiao-Lan Qin b, Xing Lu b, Qi-Pin Qin b,
Bi-Qun Zou b, d, Zhen-Feng Chen b, **, Hong Liang b, ***, Ye Zhang a, b, d, *
a
School of Pharmacy, Guilin Medical University, Guilin 541004, China
b
State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University,
Guilin 541004, China
c
Department of Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
d
Department of Chemistry & Pharmaceutical Science, Guilin Normal College, Xinyi Road 15, Guangxi 541001, China

a r t i c l e i n f o a b s t r a c t

Article history: A series of new 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one derivatives (5a15d6) were designed and
Received 17 January 2018 synthesized as antitumor agents. In vitro antitumor assay results showed that some compounds
Received in revised form exhibited moderate to high inhibitory activity against HepG2, SK-OV-3, NCI-H460 and BEL-7404 tumor
24 July 2018
cell lines, and most compounds exhibited much lower cytotoxicity against the HL-7702 normal cell line
Accepted 29 July 2018
Available online 30 July 2018
compared to 5-FU and cisplatin. In vivo antitumor assay results demonstrated that 5a3 exhibited effective
inhibition on tumor growth in the NCI-H460 xenograft mouse model and that 5d3 displayed excellent
antiproliferative activity in the BEL-7402 xenograft model. These results suggested that both 5a3 and 5d3
Keywords:
3-(1H-benzimidazol-2-yl)quinolin-2(1H)-
could be used as anticancer drug candidates. Mechanistic studies suggested that compounds 5a3 and 5d3
one derivatives exerted their antitumor activity by up-regulation of Bax, intracellular Ca2þ release, ROS generation,
Synthesis downregulation of Bcl-2, activation of caspase-9 and caspase-3 and subsequent cleavage of PARP, inhi-
Antitumor activity bition of CDK activity and activation of the p53 protein.
Action mechanism © 2018 Elsevier Masson SAS. All rights reserved.
p53

1. Introduction on the design and synthesis of new antitumor drugs with high ef-
ficiency and low toxicity to healthy cells and tissues. To achieve
As a leading cause of death worldwide, cancer is a global health these goals, combination principle strategy has been widely applied
problem. To combat with cancer, a lot of attention has been focused to design new antitumor molecules that act synchronously on
multiple targets, by combining two/more active pharmacophores
covalently in a single-hybrid molecule with dual/multiple anti-
tumor funtion [1]. The combination principle inspires us to design,
Abbreviation: FDA, Food and Drug Administration; Bax, Bcl-2-associated X
synthesize and evaluate a new kind of heterocyclic antitumor
protein; PI, propidium iodide; EB, ethidium bromide; MTT, 3-(4,5-dimethylthiazol-
2-yl)-2,5-diphenyltetrazolium bromide; CDC, cell division cyclin; CDK, cyclin-
molecules, by the joining of two important pharmacophores: 2-
dependent kinase; CDC25C, cell division cyclin 25 homolog C. oxo-quinoline and benzimidazole (Fig. 1).
* Corresponding author. School of Pharmacy, Guilin Medical University, Guilin As a kind of alkaloid which widely exist as same as quinolone, 2-
541004, China. oxo-quinoline derivatives exhibit numerous promising biological
** Corresponding author. State Key Laboratory for Chemistry and Molecular En-
activities [2e4], including antioxidation [5,6], antiproliferation [7]
gineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi
Normal University, Guilin 541004, China. and antitumor effect [8e10]. 2-Oxo-quinoline derivatives could
*** Corresponding author. State Key Laboratory for Chemistry and Molecular En- exert the antitumor effect through inducing cell apoptosis and
gineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi arresting the cell cycle, by the activation of caspases, p53 and Bax,
Normal University, Guilin 541004, China. inhibition of Bcl-2, and modulation of cytochrome c and some
E-mail addresses: chenzf@gxnu.edu.cn (Z.-F. Chen), hliang@gxnu.edu.cn
related genes/proteins [9,10]. The potential antitumor activity, easy
(H. Liang), zhangye81@126.com (Y. Zhang).
1
These authors contributed equally to this work. modification of structure and no evident toxicity [6] of 2-oxo-

https://doi.org/10.1016/j.ejmech.2018.07.066
0223-5234/© 2018 Elsevier Masson SAS. All rights reserved.
140 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

through the Vilsmeier-Haack-Arnold reaction which involved the


combination of acetanilide derivatives 2 (2a2d) with N,N-
dimethylformamide (DMF) in the presence of phosphorus oxy-
chloride [11,20]. 2-Oxo-quinoline 3-carbaldehyde derivatives
4(4a4d) were synthesized in decent yields by the hydrolytic re-
actions of 3 (3a3d) in an acetic acid (70%) aqueous solution [11]. 2-
Oxo-quinoline 3-benzimidazole derivatives 5 (5a15d6) were
finally obtained in moderate yields through coupling reactions of 4
(4a4d) with o-phenylenediamine derivatives in the presence of
methanol. The structures of compounds 5 (5a15d6) were then
Fig. 1. Design of 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one derivatives using the
combination principle. confirmed using various spectroscopic methods, including 1H NMR,
13
C NMR and high-resolution mass spectrometry (HR-MS)(Part 1.1
of Supplementary Data). In 1H NMR spectra, the chemical shift (d)
quinoline renders itself an ideal template for designing new che- around 8.91e8.97 and 11.91e12.82 were attributed to the hydro-
mopreventive agents. In our previous work, 2-oxo-quinoline has gens of NH groups fused in quinolone/benzimidazole, and d in the
been chose as pharmacophore to successfully design and synthe- range of 8.90e9.12 were mainly attributed to the hydrogens of
size some Schiff's-bases [5,6] and aminophosphate derivatives [8,9] HC¼C contained in quinolone ring, while d in the range of 6.16e7.93
with potential antioxidant and antitumor activities, by the joining were attributed to the aromatic hydrogens (H-Ar) in quinolone and
2-oxo-quinoline-3-carbaldehyde with amines or/and diethyl benzimidazole. In addition, d at 6.06, 2.38 and 2.32 were attributed
phosphite. In addition, benzimidazole, which consists of the fusion to the hydrogens of methyl group, respectively. For compounds
of benzene and imidazole, is also a bicyclic pharmacophore. 5d15d6, d in the range of 6.06e6.14 were attributed to the hy-
Benzimidazole and its derivatives exhibits a wide range of biolog- drogens of O-CH2-O group. In 13C NMR spectra, d in the range of
ical activities [11], containing antioxidant [12] and antitumor aci- 161.04e161.99 were attributed to the carbons in carbonyl groups,
tivity [13e16], which make it to be a valuable synthetic scaffold for and d around 95.40 was attributed to the carbons of O-CH2-O
designing new antitumor agents. Benzimidazole derivatives could groups, while d in the range of 20.59e20.94 and 55.94e56.09 were
exert the antitumor effect through modulation of cell cycle arrest attributed to the carbons of methyl group, respectively. d in the
and apoptotic responses [17]. Since that the antioxidants have been range of 99.47e148.19 were attributed to the carbons in C¼C and
forged as the antitumor drug candidates and the commercial C¼N groups and the aromatic carbons in in quinolone and benz-
anticancer drug bleomycin exerting the antitumor activity is imidazole. In addition, HR-MS offered the precise molecular weight
related with free radical scavenging [18], it is thus to consider that and composition of 5 (5a15d6), which were well consistent with
screening for new antitumor compounds based on the combining structures in Scheme 1. Moreover, to better understand the struc-
of 2-oxo-quinoline and benzimidazole may be a feasible way. ture of 5a15d6, the crystal structures of 5b1 and 5d6 were deter-
Owing to the synthetic and antitumor values of 2-oxo-quinoline mined by X-ray diffraction. As shown in Fig. 1-S97 and 1-S98, the
and benzimidazole derivatives, it is highly possible that the com- crystal structures of 5b1 and 5d6 were well consistent with the
bination of the 2-oxo-quinoline skeleton with benzimidazole structures determined by 1H NMR, 13C NMR and HR-MS. So the
pharmacophore may lead to potent antitumor activity. Further- structures of compounds 5 (5a15d6) were completely confirmed.
more, previous work has also indicated that some 3-(1H-benzi-
midazol-2-yl)quinolin-2(1H)-one derivatives exhibit potential 2.2. Biology
antitumor activity [19]. However, to the best of our knowledge, the
synthetic quantity and scale of 3-(1H-benzimidazol-2-yl)quinolin- 2.2.1. In vitro cytotoxicity
2(1H)-one derivatives, as well as their antitumor activities and The in vitro cytotoxicity of the 3-(1H-benzimidazol-2-yl)quino-
mechanisms have not been thoroughly explored. So a set of new lin-2(1H)-one derivatives 5(5a15d6) were determined using the
imidazol-2-yl-1H-quinolin-2-ones derivatives (5a15d6) with methylthiazoltetrazolium (MTT) assay [11,21] against the HepG2
different substituent group in quinolone/benzimidazo moiety were (human liver cancer cells), SK-OV-3 (human ovarian cancer cells),
designed and synthesized under the combination principle NCI-H460 (Human large cell lung cancer cells), BEL-7404 (Human
(Scheme 1), and their antitumor activity was then evaluated. The liver cancer cells) and HL-7702 (Human liver normal cells) cell
in vivo cytotoxicity of representative compounds (i.e., 5a3 and 5d3) lines. For comparison, two FDA-approved anticancer drugs 5-
was also evaluated. Mechanistic studies focused on apoptosis, cycle fluorouracil (5-FU) and cisplatin were used as positive controls.
arrest and activation of p53 induced by the representative com- The cytotoxicity test results were shown in Table 1. As shown in
pounds 5a3 and 5d3 was further performed. We aimed to provide Table 1, many 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one de-
information for understanding the chemopreventive mechanism of rivatives exhibited cytotoxicity against the HepG2, SKOV3, NCI-
3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one derivatives and a H460 and BEL-7404 cell lines. Compared to the 5-FU and
combination principle strategy for designing antioxidant-based cisplatin, some of these compounds displayed higher cytotoxicity
cancer chemoprevention agents. against these tumor cell lines but lower cytotoxicity against the HL-
7702 normal cell line. In the cytotoxicity assay with the HepG2 cell
line, compounds 5a2, 5a3, 5a4, 5a6, 5b3, 5d1, 5d2, 5d3 and 5d6
2. Results and discussion exhibited higher cytotoxicity than 5-FU (IC50 ¼ 31.98 mM), with
IC50values in the range of 7.54e29.63 mM. Notably, compound 5a3
2.1. Chemistry even exhibited higher cytotoxicity than cisplatin (IC50 ¼ 10.12 mM).
In the cytotoxicity assay with the SKOV-3 cell line, compounds 5a2,
3-(1H-Benzimidazol-2-yl)quinolin-2(1H)-one compounds 5 5a3, 5a4, 5a6, 5d1 and 5d3 showed higher cytotoxicity than 5-FU
(5a15d6) were synthesized as shown in Scheme 1. First, phenyl (IC50 ¼ 26.34 mM), with IC50 in the range of 8.83e24.69 mM. Com-
acetamide derivatives 2 (2a2d) were obtained by the acylation pounds 5a3, 5a4, 5a6 and 5d1 even exhibited higher cytotoxicity
reaction of compounds 1 (1a1d) [11]. 2-Chloro-quinoline-3- than cisplatin (IC50 ¼ 15.60 mM). In the cytotoxicity assay with the
carbaldehyde derivatives 3 (3a3d) were then synthesized NCI-H460 cell line, compounds 5a2, 5a3, 5a4, 5b3, 5d1 and 5d3
W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150 141

Scheme 1. Synthetic routes of compounds 5(5a15d6). Reagents and conditions: (a) acetic anhydride; (b) POCl3, DMF; (c) acetic acid (70%) aqueous solution; (d) o-phenyl-
enediamine derivatives, methanol.

Table 1
a
IC50 values of 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one derivatives 5(5a15d6) towards four selected tumor cell lines and a normal cell line.

Compounds IC50 (mM)

HepG2 SKOV-3 NCI-H460 BEL-7404 HL-7702

5a1 >100 >100 >100 >100 >100


5a2 15.21 ± 1.17 18.34 ± 1.46 24.18 ± 1.38 27.45 ± 1.47 >100
5a3 8.45 ± 1.63 10.52 ± 1.83 14.72 ± 1.64 16.54 ± 1.45 >100
5a4 10.81 ± 1.04 11.40 ± 0.78 16.79 ± 1.98 14.46 ± 0.77 >100
5a5 >100 >100 >100 >100 >100
5a6 29.63 13.80 >100 >100 >100
5b1 >100 >100 >100 >100 >100
5b2 >100 >100 >100 >100 >100
5b3 26.59 ± 1.79 33.02 ± 1.52 36.53 ± 1.12 38.95 ± 1.27 >100
5b4 >100 >100 >100 >100 >100
5b5 >100 >100 >100 >100 >100
5b6 >100 >100 >100 >100 >100
5c1 >100 >100 >100 >100 >100
5c2 >100 >100 >100 >100 >100
5c3 >100 >100 >100 >100 >100
5c4 >100 >100 >100 >100 >100
5c5 >100 >100 >100 >100 >100
5c6 >100 >100 >100 >100 >100
5d1 18.66 ± 1.56 10.62 ± 1.37 23.75 ± 1.72 15.71 ± 0.89 >100
5d2 12.43 ± 0.91 >100 >100 10.35 ± 1.35 >100
5d3 10.56 ± 1.38 24.69 ± 1.22 30.36 ± 1.07 9.06 ± 1.36 >100
5d4 >100 >100 >100 >100 >100
5d5 >100 >100 >100 >100 >100
5d6 25.56 32.17 >100 >100 >100
5-FU 31.98 ± 0.56 26.34 ± 0.57 45.44 ± 0.94 40.21 ± 1.98 58.74 ± 2.31
Cisplatin 10.12 ± 0.71 15.60 ± 1.7 20.36 ± 0.50 14.72 ± 0.66 15.67 ± 0.32
a
IC50 values are presented as mean ± SD (standard error of the mean) from three repeating experiments.

exhibited higher cytotoxicity than 5-FU (IC50 ¼ 45.44 mM), with IC50 antiproliferative activity than other compounds, indicating that the
in the range of 14.03e38.25 mM. Compounds 5a3 and 5a4 even benzodioxole substitution in quinolone moiety may have positive
displayed higher cytotoxicity than cisplatin (IC50 ¼ 20.36 mM). In influence on the antiproliferative activity. The respective compar-
the cytotoxicity assay with the BEL-7404 cell line, compounds 5a2, ison of 5a2, 5b2 and 5c2 showed that 5a2 exhibited the best anti-
5a3, 5a4, 5b3, 5d1, 5d2 and 5d3 exhibited higher cytotoxicity than 5- proliferative effect among them, implying that no(H) substitution
FU (IC50 ¼ 40.21 mM), with IC50 values in the range of in 6 position of quinolone moiety may also have active influence on
9.06e39.94 mM. Compounds 5a4, 5d2 and 5d3 even displayed the antiproliferative activity. In addition, the respective comparison
higher cytotoxicity than cisplatin (IC50 ¼ 14.72 mM). of 5a2, 5a3, 5a4 with other compounds in 5a(5a15a6) kind, as well
By the respective comparison of compounds 5a1, 5b1, 5c1 with as that of 5d2, 5d3 with other compounds in 5d(5d15d6) kind,
5d1, it should be found that compound 5d1 exhibited better demonstrated that halide-substitution in benzimidazole moiety
142 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

exhibited positive effect on their antiproliferative activity. The or- into the vehicle control groups, the experimental groups, and the
der listed as follow: chlorine > bromine > fluorine. In brief, based positive control groups.
on the above observation, it could be concluded that, for com- As shown in Fig. 3A, both of the low dose (25 mg/kg) and high
pounds 5(5a15d6), no(H) substitution in 6 position and the ben- dose (50 mg/kg) of 5a3 exhibited important inhibition on NCI-460
zodioxole substitution in 6,7 position in quinolone moiety, as well tumor growth, and the relative tumor increment rates (T/C) were
as methyl, fluor, chlor and bromo substitutions in benzimidazole calculated as 40.6% and 30.7%, respectively, based on the tumor
moiety may have positive influence on the antiproliferative activity. volume. In comparison, cisplatin at the 2 mg/kg dose exhibited a T/
Among all the 3-(2-benzimidazolyl)quinolone derivatives, C value of 29.9%. The tumors were harvested and weighted on day
compounds 5a3 and 5d3 showed the highest antiproliferation ac- 14, and the inhibitory rates on the growth of tumor weight were
tivity against the cancer cell lines. They were thus selected as calculated. Fig. 3C showed that 5a3 (at the 50 mg/kg dose) displayed
representative compounds for the mechanistic studies. significant antitumor activity in the NCI-460 model with an
inhibitory rate of 66.9%, greater than that of cisplatin (63.2%,
2.2.2. Preliminary safety evaluation of the selected compounds P < 0.01). These in vivo results indicated that 5a3 (at the 50 mg/kg
The in vivo toxicity of compounds 5a3 and 5d3 was then evalu- dose) exhibited higher antitumor effect than cisplatin (at the 2 mg/
ated in Kun-ming (KM) mice. Through intraperitoneal injection, the kg dose) in the NCI-460 mouse model, consistent with the results of
mice received 5a3 or 5d3 (dissolved in a 5% v/v DMSO/saline sol- the in vitro cytotoxicity assay. It should be noted that no significant
vent) at a dose of 50 mg/kg per day for 14 consecutive days. The change in body weight and no other adverse effects were observed
body weight of the animals was monitored daily and used as a among the mice treated with 5a3, suggesting that 5a3 exhibited no
parameter of systemic toxicity. significant toxicity to the mice within the 13-day period of treat-
Notably, no adverse effects were observed in the KM mice ment (Fig. 3B) and thus could be good candidate for antitumor drug
injected with the selected compounds during the 14-day moni- design.
toring period. As shown in Fig. 2 (5a3) and Fig. 3-S1 (5d3) (Part 3 of The in vivo antitumor activity of 5d3 was also investigated in the
Supplementary Data), the average body weight of the mice treated NCI-460 mouse model (Fig. 3-S3) (Part 3 of Supplementary Data).
with the two compounds showed similar growth rates to that of the The T/C values were determined to be 48.8% (at the 25 mg/kg dose)
control group. and 38.1% (at the 50 mg/kg dose). The inhibitory rates were deter-
The pathological changes of different organs of the KM mice mined to be 46.2% (at the 25 mg/kg dose) and 55.1% (at the 50 mg/
treated with 5a3 and 5d3 were also characterized. The organs, kg dose). Notably, no significant change in body weight and no
including kidney, heart, and liver, from the mice treated 5a3 and other adverse effects were observed among the mice treated with
5d3 were harvested and used to produce pathological slices for 5d3, suggesting that 5d3 exhibited no significant toxicity to the
microscopy characterization. As shown in Fig. 3-S2 (Part 3 of Sup- mice within the 13-day period of treatment (Fig. 3-S3B).
plementary Data), compared to the control groups, there were no 5-FU is the most common chemotherapeutic drug used for the
signs of peritonitis or damage to these organs in the mice treated treatment of hepatocellular carcinoma. Thus it was used as the
with 5a3 and 5d3. These observations indicated that 5a3 and 5d3 positive control in the in vivo experiments with the BEL-7402
had no significant toxicity to the KM mice at the dose of 50 mg/kg, mouse model. As shown in Fig. 3-S4A (Part 3 of Supplementary
suggesting that they may be good candidates as antitumor drugs. Data), both of the low (25 mg/kg) and high dose (50 mg/kg) of
5d3 displayed evident antiproliferative activity on the tumor
2.2.3. In vivo antitumor activity of 5a3 and 5d3 growth compared with the negative control group. The T/C values
In vivo antiproliferation experiments were performed to further of these two compounds were determined to be 51.5% and 44.8%
evaluate the antitumor activity of 5a3 and 5d3. The NCI-460 and (p < 0.01), respectively. It was worth noting that 5-FU at the 25 mg/
BEL-7402 xenograft models were selected in this study. In the NCI- kg dose showed a T/C value of 56.1%, higher than that of 5d3. In
460 tumor model, cisplatin was used as the positive control, addition, Fig. 3-S4C showed that the inhibitory rates on the growth
whereas 5-FU was used as the positive control in the BEL-7402 of tumor results weight were determined to be 41.2 and 44.9%
tumor model. The SPF BALB/c nude mice were randomly divided (p < 0.01) for the low and high dose of 5d3, respectively. In com-
parison, the inhibitory rate of 5-FU (at the 25 mg/kg dose) was
34.5%. These results indicated that 5d3 (both at 25 mg/kg and
50 mg/kg dose) showed more potent in vivo antitumor activity than
5-FU (at 25 mg/kg dose) in the BEL-7402 mouse model, consistent
with the result of the in vitro cytotoxicity assay. Moreover, no sig-
nificant changes in body weight and no other adverse effects were
found in the mice treated with 5d3 (Fig. 3-S4B), suggesting that 5d3
may be good candidate for antitumor drug design.
Pathological slices of the tumors harvested in the mice treated
with 5a3 and 5d3 were generated and characterized using micro-
scopy. As shown in Fig. 3-S5 (Part 3 of Supplementary Data), it was
evident that the treatment of 5a3 and 5d3 led to necrosis of the
tumor cells (NCI-460 and BEL-7402) compared with the vehicle
samples, indicating potent antiproliferative activity of 5a3 and 5d3.

2.3. Action mechanism

2.3.1. Investigation of cell cycle distribution


It is well known that cell cycle checkpoints are significant con-
trol mechanisms that ensure the performance of cell cycle events
Fig. 2. Changes in the body weight of the KM mice injected with compound 5a3. There [22]. To investigate the possible role of cell cycle arrest in the tumor
are six mice in each experimental group. cell death induced by 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-
W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150 143

Fig. 3. In vivo anticancer activity of 5a3 in the NCI-460 xenograft model. (A) Effect of 5a3 (at the doses of 50 and 25 mg/kg), cisplatin (at the dose of 2 mg/kg), or vehicle (5% DMSO in
saline, v/v) on tumor growth. Tumor growth was monitored by the mean tumor volume (mm3) ± SD (n ¼ 6) and calculated as the relative tumor increment rate (T/C, %). (B) Body
weight change of the mice treated with 5a3. (C) Tumor weight of the mice. The tumors were collected in the mice at day 14. The “**” signs represent p < 0.01 (versus the vehicle
control group). (D) Photographs of the harvested tumors from the mice.

one derivatives, HepG2 cells were treated with the 5a3 at different population of 30.58%, higher than that of the control group
concentrations (0, 6, 8 and 10 mM) for 48 h. For comparison, BEL- (18.84%). Thus, compound 5d3 also arrested the cell cycle of the
7404 cells were treated with compound 5d3 (9 mM). As shown in BEL-7404 cells in the G2/M stage, consistent with the scenarios of
Fig. 4, the treatment of HepG2 cells with compound 5a3 also 5a3.
increased cell cycle arrest at the G2 phase, resulting in an obvious G2/M transition is mainly regulated by timed activation of cyclin
increase in the G2-phase population (39.00% at 6 mM, 39.71% at B1/CDK1(CDC2) complexes whose activity is mainly mediated by
8 mM and 49.10% at 10 mM) compared with the control group the positive mediator cell division cyclin 25 homolog C (CDC25C)
(19.04%). As a result, the G1-phase and S-phase population of the phosphatase [23,24]. CDC25C modulates the phosphorylation of
HepG2 cells were reduced. These results demonstrated that com- CDK1 leading to the activation of cyclin B1/CDK1 complexes and
pound 5a3 arrested the cell cycle of the HepG2 cells at the G2/M execution of G2/M transition [23,24]. Responding to DNA damage
stage in a concentration-dependent manner. Fig. 3-S6 (Part 3 of and cellular activities resulting in G2/M arrest, phosphorylation of
Supplementary Data) showed that treating the BEL-7404 cells with CDC25C leads to its degradation and sequestration to the cyto-
5d3 (9 mM) also caused cell cycle arrest at the G2 phase with the plasm, and finally results in the destabilization of the cyclin-CDK

Fig. 4. Cell cycle analysis of the HepG2 cells treated with 5a3. The HepG2 cells were treated with compound 5a3 at different concentrations i.e., (a) 0 mM, (b) 6 mM, (c) 8 mM and (d)
10 mM, for 48 h.
144 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

complexes [25]. Furthermore, the proteins encoded by some anti- concentration dependent manner. These results demonstrated that
oncogenes, including p53, p27 and p21, also play an important role compound 5a3 induced apoptosis of the HepG2 cells in a dose-
in G2/M arrest through promoting the accumulation of the dormant dependent manner.
cyclin B-CDK complex [26]. In this work, the expression levels of
cyclin B1, CDK1, CDC25C, p53, p27 and p21 in the HepG2 cells were 2.3.3. ROS generation
determined using western blot. As shown in Fig. 5, compared to the It is known that production of intracellular reactive oxygen
negative control, the lysates of the HepG2 cells treated with 5a3 species (ROS) can induce apoptosis [29]. In order to study the role of
exhibited decreased levels of cyclin B1, CDK1 and CDC25C as well ROS in 5a3-induced apoptosis in the cancer cells, the intracellular
increased levels of p53, p21 and p27. Therefore, it could be ROS levels were evaluated using fluorescence microscopy using
concluded that 5a3 induced G2/M phase cell cycle arrest through 40 ,6-diamidino-2-phenylindole dihydrochloride (DAPI) and 2,7-
inhibition of CDK activity by increased expression levels of p53, p27 dichlorofluorescein diacetate (DCFH-DA) as fluorescent probes. As
and p21. shown in Fig. 9, in the DAPI staining assay, the HepG2 cells treated
The in vivo expression levels of cyclin B1, CDK1 and CDC25C, as with compound 5a3 as well as the control cells exhibited blue
well as p53, p27 and p21 in the tumor tissues of the NCI-460 and fluorescence in the cytoplasm. For the DCFH-DA staining assay, the
BEL-7402 xenograft mouse models were also investigated by HepG2 cells treated with compound 5a3 displayed stronger green
western blot assays. As shown in Fig. 3-S7 (Part 3 of Supplementary fluorescence signals compared to the control cells, suggesting that
Data), it was evident that the in vivo expression levels of p53 in the 5a3 upregulated the levels intracellular of ROS. Thus, ROS genera-
tumor tissues increased upon treating the mice with 5a3. Taken tion induced by the 3-(1H-benzimidazol-2-yl)quinolin-2(1H)-one
together, it can be concluded that compounds 5a3 may be p53 derivatives might be the underlying cause for apoptosis [28].
activator.
2.3.4. Intracellular Ca2þ release
2.3.2. Apoptosis assays Previous studies demonstrated that intracellular calcium levels
As a key pathway leading to cell death, apoptosis is deemed to participate in cell apoptosis and calcium overload could induce
be an effective mechanism in cancer treatment [27,28]. We studied apoptosis [30]. We investigated the role of calcium signalling in the
the apoptosis-inducing effect of compound 5a3 using a fluorescent cellular apoptosis induced by 5a3. The HepG2 cells were treated
probe Hoechst 33258 as well as acridine orange/ethidium bromide with 5a3 for 24 h, and the intracellular Ca2þ concentration was
(AO/EB) staining. In the Hoechst 33258 staining test, the normal determined by fluorescence microscopy using DCFH-DA as a cal-
HeGp 2 cells could emit blue fluorescence, whereas the cells treated cium probe. As shown in Fig. 10, treating HepG2 cells with 5a3
with 5a3 for 12 h displayed stronger blue fluorescence and resulted in an increase of intracellular Ca2þ concentration. The re-
exhibited characteristic apoptotic morphologies (Fig. 6). These ob- sults indicated that 5a3-induced apoptosis was associated with up-
servations revealed that compound 5a3 induced apoptosis of the regulation of the intracellular Ca2þ level.
HeGp 2 cells [25]. In the AO/EB staining experiment, the
morphology of 5a3-treated HeGp 2 cells exhibited significant 2.3.5. Caspase-dependent apoptosis in the HepG2 cells
change (Fig. 7). The cell nuclei were stained as yellow/green or In vitro assay. Previous work has shown that cancer cells can
orange, and the morphology changes indicated pycnosis, mem- restrict or escape from apoptosis in many ways, which often involve
brane blebbing and cell budding, all of which are characteristic of the perturbation of the Bcl-2 intrinsic apoptotic pathway [31-35].
apoptosis [25]. Taken together, it could be concluded that com- The Bcl-2 family proteins play significant regulating roles both as
pound 5a3 induced apoptosis in the HeGp 2 cells. pro- and anti-apoptotic proteins and a slight change in the dynamic
The apoptosis ratios in the HepG2 cells treated with 5a3 were balance of these proteins may result in either inhibition or pro-
quantitatively determined by flow cytometry and the results were motion of cell death [31,34]. Accordingly, the ratio of Bcl-2/Bax
shown in Fig. 8. Fig. 8 showed that the treatment of the HepG2 cells expression is more important than the individual levels of each
with compound 5a3 at different concentrations (6, 8 and 10 mM) Bcl-2 family protein in the regulation of apoptosis [35]. To study the
caused an increase of the apoptotic cell population in a mechanism of 5a3-induced apoptosis in cancer cells, the expression

Fig. 5. Effect of compound 5a3 on the expression levels of relevant proteins in HepG2 cells. Western blot analysis was performed with antibodies against cyclin B1, CDK1, CDC25C,
p21, p23, and p53. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as the loading standard.
W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150 145

Fig. 6. Effects of compound 5a3 on the morphology of the HepG2 cells. The cells were stained with the Hoechst 33258 dye.

Fig. 7. Compound 5a3 induced apoptosis in the HepG2 cells. The cells were visualized
via the AO/EB staining coupled with fluorescence microscopy.

levels of Bax, Bcl-2 and cytochrome c in the HepG2 cells treated


with 5a3 were determined by western blotting. Fig. 11 shows that Fig. 8. Apoptosis ratio of the HepG2 cells. (a) Negative control; (b) treated with 6 mM
treating the HepG2 cells with 5a3 resulted in an increase in the 5a3, (c) treated with 8 mM 5a3, (d) treated with 10 mM 5a3. The cells were analyzed
expression level of Bax and a decrease in the expression level of Bcl- using the Annexin V/PI assay in a flow cytometer.
2 accompanied by the release of mitochondrial cytochrome c into
the cytosol.
Bcl-2 family proteins play a key role in apoptosis by interfering the expression level of Bax increased in the 5a3-treated NCI-
with caspases, which are pivotal effectors of programmed cell 460xenograft model, but remained relatively unchanged in the
death [36,37]. As a family of cysteinyl aspartate specific proteases 5d3-treated group in the BEL-7402xenograft model. The expression
participate in apoptosis, caspases are divided into groups of initi- level of Bcl-2 decreased upon the treatment of 5a3 and 5d3 in the
ators (caspases 8, 9 and 10) and executioners (caspases 3, 6 and 7) NCI-460 xenograft model. The expression level of cytochrome c
[36,37]. The caspase cascade is initiated by the proteolysis of increased upon the treatment of 5a3 in the NCI-460xenograft
inactive procaspases and propagated by the cleavage of down- model. There was no significant change in the expression level of
stream caspases and substrates such as poly(ADP-ribose) poly- caspase-3 in both mouse models. The expression level of caspase-9
merase (PARP) [24]. To evaluate whether caspases were activated in increased upon the treatment with both 5a3 and 5d3 in the NCI-460
5a3-induced apoptosis, the expression levels of caspase-9, caspase- xenograft model but decreased upon the 5d3 treatment in BEL-
3 and PARP was determined by western blot. Fig. 11 showed that 7402xenograft model. The expression level of PARP increased
the HepG2 cells treated with 5a3 exhibited elevated expression upon the 5a3 treatment in the NCI-460 xenograft model, but
levels of caspase-9, caspase-3 and PARP, compared to the control decreased upon the 5d3 treatment in BEL-7402xenograft model.
samples. These results demonstrated that compounds 5a3 exerted The 5a3 treatment led to an increase in the expression level of Apaf-
pro-apoptotic effects through a mitochondrial mediated pathway 1 in the NCI-460 model, whereas in BEL-7402 model the 5d3
and caspase cascade. treatment resulted in a decrease in the expression level of Apaf-1.
In vivo assay. The expression levels of Bax, Bcl-2, cytochrome c, Through comparison of the in vitro and in vivo studies of the
caspase-9, caspase-3 and PARP, as well as apoptotic protease acti- expression levels of these proteins, it can be concluded that com-
vating facter-1 (Apaf-1), in the tumor tissues of the NCI-460 and pound 5a3 may inhibit the expression of Bcl-2, lead to the release of
BEL-7402 xenograft mouse models were also investigated using cytochrome c and Apaf-1, and finally activate caspase-9 and induce
western blot. As shown in Fig. 3-S8 (Part 3 of Supplementary Data), apoptosis.
146 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

2 cells treated with 5a3 compared with the negative control sample
(5a6, Fig. 12). These results demonstrated that upregulated p53
expression may be involved in the mechanism of 5a3-induced G2/M
cell cycle arrest, confirming that 5a3 was a p53 activator.
To further confirm whether p53 was involved in the antitumor
property of the HepG2 cells, disruption of p53 expression using
siRNRs was performed. The results in Fig. 13 showed that the S117,
S118 and S119 fragments can significantly reduce the p53 mRNA
level. It was evident that the S119 fragment exhibited the best
interference effect and was selected for further studies. As shown in
Fig. 3-S9 (Part 3 of Supplementary Data), treatment of the HepG2
cells with both S119 and compound 5a3 caused cell cycle arrest in
the G2/M stage, consistent with the result obtained by treating the
cells with 5a3 alone. A combinational treatment with S119 and 5d3
caused cell cycle arrest at the G1 stage, different from the result
obtained by treating the cells with 5d3 alone. Fig. 3-S10 (Part 3 of
Supplementary Data) showed that treatment of the cells with both
S119 and compound 5a3 led to an evident increase of the apoptotic
cell population, i.e., from 14% to 19.12%, and a combinational
treatment with S119 and compound 5d3 also caused an increase of
the apoptotic cell population, i.e., from 14.59% to 19.16%. Taken
together, it can be concluded that compounds 5a3 and 5d3 may
induce apoptosis through the activation of the tumor suppressor
Fig. 9. Compound 5a3 affected the levels of intracellular ROS in the HepG2 cells. p53.

3. Conclusion

In summary, we designed and synthesized a series of 3-(1H-


benzimidazol-2-yl)quinolin-2(1H)-one derivatives and evaluated
the cytotoxicity of these compounds against four cancer cell lines
(HepG2, SKOV-3, NCI-H460 and BEL-7404). We identified several
compounds exhibiting high in vitro anticancer activity and low
toxicity against normal HL-7702 cells. In vivo antitumor activity
study and animal toxicology test showed that 5a3and 5d3 exhibited
potent antitumor activity in different mice models. Mechanistic
studies showed that 5a3 and 5d3 caused apoptosis of tumor cells
possibly through regulation of the expression levels of apoptotic
proteins (such as Bax, Bcl-2, cytochrome c, caspase-9, caspase-3,
and PARP), intracellular Ca2þ release and ROS regulation. Cell cycle
studies indicated that compounds 5a3 and 5d3 induced cell cycle
arrest at the G2/M phase. The observed cell cycle arrest resulted
from the inhibition of CDK activity and activation of the p53 pro-
tein. This work may provide information for understanding the
chemopreventive mechanism of 3-(1H-benzimidazol-2-yl)quino-
lin-2(1H)-one derivatives and a combination principle strategy for
designing antioxidant-based cancer chemoprevention agents.

4. Experimental

Fig. 10. Compound 5a3 caused an increase in the level of intracellular Ca level in the
4.1. Chemistry
HepG2 cells.

All chemicals of reagent grade were commercially available and


used without further purification. NMR spectra were recorded on a
2.3.6. P53 activation BRUKER AVANCE AV 400/500 spectrometer using tetramethylsilane
p53 is a pivotal tumor suppressor protein with a key role in (TMS) as the internal standard. The mass spectra were collected on
many cellular processes and one of the most relevant cellular a BRUKER ESQUIRE HCT spectrometer. The GelRed nucleic acid stain
components related to cancer growth [38,39]. According to the was purchased from Biotium.
intracellular context, accumulation of p53 in response to various
stresses including DNA damage leads to cell cycle arrest, senes- 4.1.1. General synthesis procedure for compounds 5(5a15d6)
cence and apoptosis [40]. Since it can be functionally activated to 2-Oxo-quinoline 3-carbaldehyde derivatives 4(4a¡4d)
eradicate tumors, p53 has been considered as an attractive target of (1 mmol), o-phenylenediamine derivatives (1.0 mmol) and 3 mL
anticancer therapy [38,39]. To better understand whether the methanol were mixed in a pressure tube and heated at 90  C for 4 h.
expression of p53 was regulated by 5a3, the HepG 2 cells expressing After the reaction, the mixture was cooled to room temperature
a mutant form of the p53 protein were selected. Western blot and filtered to yield the product as yellow powder.
analysis showed an obvious increase of p53 expression in the HepG 5a1: Yield, 71.4%; 1H NMR (500 MHz, DMSO‑d6) d 12.42 (s, 2H,
W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150 147

Fig. 11. Effect of compound 5a3 on the expression levels of Bax, Bcl-2, cytochrome c, caspase-3, caspase-9 and PARP in the HepG2 cells. GAPDH was used as a loading standard.

J ¼ 7.8 Hz, 1H, H-Ar), 7.30 (t, J ¼ 7.5 Hz, 1H, H-Ar), 7.11e7.04 (m, 1H,
H-Ar); 13C NMR (126 MHz, DMSO‑d6) d 161.23 (C¼O), 161.15, 160.06,
158.25, 158.19, 149.87 (C¼N), 149.18, 139.89, 139.19 (C¼C), 132.14
(C¼C), 129.55, 119.56, 115.75, 110.56, 103.89, 99.47, 99.26; ESI-
HRMS, calculated m/z for C16H10FN3O [MH]-: 278.0730, found:
278.0740.
5a3: Yield, 72.2%; 1H NMR (500 MHz, DMSO‑d6) d 12.52 (s, 1H,
NH), 9.12 (s, 1H, HC¼C), 7.96 (s, 1H, H-Ar), 7.74 (s, 1H, H-Ar), 7.70 (d,
J ¼ 8.6 Hz, 1H, H-Ar), 7.63 (s, 1H, H-Ar), 7.45 (d, J ¼ 8.2 Hz, 1H, H-Ar),
7.30 (s, 1H, H-Ar), 7.23 (s, 1H, H-Ar); 13C NMR (126 MHz, DMSO‑d6)
Fig. 12. Effect of compounds 5a3 and 5a6 on the expression of p53. GAPDH was used as
d 161.14 (C¼O), 149.52 (C¼N), 140.16, 139.31 (C¼C), 132.37 (C¼C),
the loading standard.
129.65, 126.98, 123.20, 122.90, 119.90, 119.52, 115.78; ESI-HRMS,
calculated m/z for C16H10ClN3O [MH]-: 294.0434, found:
294.0446.
5a4: Yield, 73.2%; 1H NMR (500 MHz, DMSO‑d6) d 12.52 (s, 1H,
NH), 11.91 (s, 1H, NH), 9.12 (s, 1H, HC¼C), 7.98 (s, 1H, H-Ar), 7.88 (s,
1H, H-Ar), 7.66 (s, 1H, H-Ar), 7.64 (d, J ¼ 6.0 Hz, 1H, H-Ar), 7.45 (d,
J ¼ 8.3 Hz, 1H, H-Ar), 7.35 (s, 1H, H-Ar), 7.30 (s, 1H, H-Ar); 13C NMR
(126 MHz, DMSO‑d6) d 161.14(C¼O), 149.35 (C¼N), 140.21, 139.32
(C¼C), 132.38 (C¼C), 129.66, 125.47, 123.20, 119.88, 119.53, 115.78,
114.89; ESI-HRMS, calculated m/z for C16H10BrN3O [MH]-:
337.9929, found: 337.9947.
5a5: Yield, 69.1%; 1H NMR (500 MHz, DMSO‑d6) d 12.18 (s, 1H,
NH), 8.97 (s, 1H, NH), 8.93 (s, 1H, HC¼C), 7.93 (s, 1H, H-Ar), 7.81 (d,
J ¼ 7.3 Hz, 1H, H-Ar), 7.60 (t, J ¼ 7.2 Hz, 1H, H-Ar), 7.37 (d, J ¼ 8.2 Hz,
1H, H-Ar), 7.27 (t, J ¼ 7.5 Hz, 1H, H-Ar), 6.89 (s, 1H, H-Ar), 6.79 (d,
J ¼ 8.8 Hz, 1H, H-Ar); 13C NMR (126 MHz, DMSO‑d6) d 161.99 (C¼O),
154.36, 151.57 (C¼N), 140.35, 139.27 (C¼C), 136.45, 134.44, 132.52
(C¼C), 129.92, 126.85, 125.01, 123.01, 119.47, 115.80, 113.37, 113.24;
Fig. 13. Inhibition of p53 expression using siRNAs into the HepG2 cells. ESI-HRMS, calculated m/zfor C16H10N4O3 [MH]-: 305.0675, found:
305.0694.
5a6: Yield, 80.2%; 1H NMR (400 MHz, DMSO‑d6) d 12.66 (s, 1H,
NH), 9.05 (s, 1H, HC¼C), 7.94 (d, J ¼ 7.2 Hz, 1H, H-Ar), 7.60 (t,
NH), 12.48 (s, 1H, NH), 9.13 (s, 1H, HC¼C), 7.97 (d, J ¼ 7.3 Hz, 1H, H-
J ¼ 8.4 Hz, 1H, H-Ar), 7.44 (br, 3H, H-Ar), 7.28 (t, J ¼ 8.0 Hz, 1H, H-Ar),
Ar), 7.73 (s, 1H, H-Ar), 7.67 (s, 1H, H-Ar), 7.65e7.59 (m, 1H, H-Ar),
2.32 (s, 6H, -CH3); 13C NMR (126 MHz, DMSO‑d6) d 161.28 (C¼O),
7.45 (d, J ¼ 8.2 Hz, 1H, H-Ar), 7.30 (t, J ¼ 7.1 Hz, 1H, H-Ar), 7.24e7.19
147.31 (C¼N), 138.99, 138.83 (C¼C), 131.80 (C¼C), 129.38, 123.07,
(m, 2H, H-Ar); 13C NMR (101 MHz, DMSO‑d6) d 161.26 (C¼O), 148.19
120.75, 119.70, 115.68, 20.59 (CH3); ESI-HRMS, calculated m/zfor
(C¼N), 143.24, 139.56 (C¼C), 139.15, 134.90, 132.08 (C¼C), 129.53,
C18H15N3O [MþH]þ: 290.1293, found: 290.1286.
123.13, 122.75, 122.39, 120.48, 119.62, 118.76, 115.72, 113.26; ESI-
5a2: Yield, 71.7%; 1H NMR (500 MHz, DMSO‑d6) d 12.75 (d,
HRMS, calculated m/zfor C16H11N3O [MþH]þ: 262.0980, found:
J ¼ 12.1 Hz, 1H, NH), 12.49 (s, 1H, NH), 9.10 (d, J ¼ 10.8 Hz, 1H,
262.0986.
HC¼C), 7.96 (d, J ¼ 7.9 Hz, 1H, H-Ar), 7.74e7.65 (m, 1H, H-Ar),
5b1: Yield, 72.1%; 1H NMR (400 MHz, DMSO‑d6) d 12.36 (s, 2H,
7.64e7.58 (m, 1H, H-Ar), 7.49 (dd, J ¼ 9.3, 2.6 Hz, 1H, H-Ar), 7.44 (d,
NH), 8.96 (s, 1H, HC¼C), 7.71 (s, 1H, H-Ar), 7.45 (s, 2H, H-Ar), 7.42 (s,
148 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

1H, H-Ar), 7.34 (d, J ¼ 8.4 Hz, 1H, H-Ar), 2.38 (s, 3H, -CH3), 2.32 (s, z for C17H12FN3O2 [MþH]þ: 310.0992, found: 310.0999.
6H, -CH3); 13C NMR (101 MHz, DMSO‑d6) d 161.17 (C¼O), 147.44 5c3: Yield, 67.7%; 1H NMR (500 MHz, DMSO‑d6) d 12.82 (s, 1H,
(C¼N), 138.52 (C¼C), 137.07, 133.21, 132.14 (C¼C), 128.62, 120.65, NH), 12.42 (s, 1H, NH), 9.08 (s, 1H, HC¼C), 7.80e7.62 (m, 2H, H-Ar),
119.66, 118.79, 115.62, 113.05, 20.94 (CH3), 20.62 (CH3); ESI-HRMS, 7.52 (d, J ¼ 2.6 Hz, 1H, H-Ar), 7.38 (d, J ¼ 9.0 Hz, 1H, H-Ar), 7.29 (dd,
calculated m/z for C19H17N3O [MþH]þ: 304.1450, found: 304.1455. J ¼ 9.0, 2.8 Hz, 1H, H-Ar), 7.22 (dd, J ¼ 8.6, 2.0 Hz, 1H, H-Ar), 3.83 (s,
5b2: Yield, 71.9%; 1H NMR (500 MHz, DMSO‑d6) d 12.74 (d, 3H, -OCH3); 13C NMR (126 MHz, DMSO‑d6) d 160.69 (C¼O), 155.19,
J ¼ 11.7 Hz, 1H, NH), 12.42 (s, 1H, NH), 9.00 (d, J ¼ 11.0 Hz, 1H, 149.70, 139.56 (C¼C), 134.00, 126.89, 122.78, 122.02, 120.18, 117.11,
HC¼C), 7.72 (q, J ¼ 5.3 Hz, 2H, H-Ar), 7.49 (dd, J ¼ 10.5, 1.3 Hz, 1H, H- 110.40, 56.03 (OCH3); ESI-HRMS, calculated m/z for C17H12ClN3O2
A), 7.47e7.41 (m, 2H, H-A), 7.35 (d, J ¼ 8.4 Hz, 1H, H-A), 2.38 (s, 3H, [MþH]þ: 326.0696, found: 326.0703.
-CH3); 13C NMR (126 MHz, DMSO‑d6) d 161.11 (C¼O), 161.03, 150.00 5c4: Yield, 73.3%; 1H NMR (500 MHz, DMSO‑d6) d 12.82 (s, 1H,
(C¼N), 139.56 (C¼C), 137.32, 133.64, 132.24 (C¼C), 131.64, 128.80, NH), 12.41 (s, 1H, NH), 9.08 (s, 1H, HC¼C), 7.87 (s, 1H, H-Ar), 7.65 (s,
120.02, 119.53, 115.68, 113.92, 110.71, 104.07, 99.46, 20.91 (CH3); ESI- 1H, H-Ar), 7.51 (s, 1H, H-Ar), 7.38 (d, J ¼ 8.9 Hz, 1H, H-Ar), 7.34 (d,
HRMS, calculated m/z for C17H12FN3O [MþH]þ: 294.1043, found: J ¼ 8.5 Hz, 1H, H-Ar), 7.28 (d, J ¼ 8.9 Hz, 1H, H-Ar), 3.82 (s, 3H,
294.1053. -OCH3); 13C NMR (126 MHz, DMSO‑d6) d 160.68 (C¼O), 155.19,
5b3: Yield, 68.7%; 1H NMR (500 MHz, DMSO‑d6) d 12.79 (d, 149.55 (C¼N), 139.61 (C¼C), 134.01 (C¼C), 125.36, 122.03, 120.17,
J ¼ 27.0 Hz, 1H, NH), 12.43 (s, 1H, NH), 9.02 (s, 1H, HC¼C), 7.73e7.72 120.13, 117.11, 114.81, 110.39, 56.03 (OCH3); ESI-HRMS, calculated m/
(m, 1H, H-Ar), 7.70 (s, 1H, H-Ar), 7.66 (d, J ¼ 8.6 Hz, 1H, H-Ar), 7.46 z for C17H12BrN3O2 [MþH]þ: 370.0191, found: 370.0189.
(d, J ¼ 8.4 Hz, 1H, H-Ar), 7.35 (d, J ¼ 8.4 Hz, 1H, H-Ar), 7.24e7.20 (m, 5c5: Yield, 67.7%; 1H NMR (500 MHz, DMSO‑d6) d 12.09 (s, 1H,
1H, H-Ar), 2.38 (s, 3H, -CH3); 13C NMR (126 MHz, DMSO‑d6) d 161.04 NH), 8.93 (s, 2H, NH, HC¼C), 7.93 (s, 1H, H-Ar), 7.31 (d, J ¼ 8.8 Hz,
(C¼O), 149.52 (C¼N), 144.15, 139.82 (C¼C), 137.37, 133.74, 132.27 2H, H-Ar), 7.26 (d, J ¼ 8.9 Hz, 1H, H-Ar), 6.87 (s, 1H, H-Ar), 6.79 (d,
(C¼C), 128.86, 126.82, 122.83, 120.00, 119.50, 118.04, 115.70, 114.57, J ¼ 8.6 Hz, 1H, H-Ar), 3.82 (s, 3H, -OCH3); 13C NMR (126 MHz,
112.92, 20.92 (CH3); ESI-HRMS, calculated m/z for C17H12ClN3O DMSO‑d6) d 161.56 (C¼O), 154.94, 154.40, 151.58 (C¼N), 138.81
[MþH]þ: 310.0747, found: 310.0752. (C¼C), 136.47, 135.11, 134.39 (C¼C), 127.08, 124.99, 122.25, 120.01,
5b4: Yield, 71.8%; 1H NMR (500 MHz, DMSO‑d6) d 12.80 (d, 117.16, 113.37, 113.19, 110.44, 55.94 (OCH3); ESI-HRMS, calculated m/
J ¼ 23.9 Hz, 1H, NH), 12.44 (s, 1H, NH), 9.03 (s, 1H, HC¼C), 7.88 (d, z for C17H12N4O4 [MH]-: 335.0781, found: 335.0807.
J ¼ 28.0 Hz, 1H, H-Ar), 7.74 (s, 1H, H-Ar), 7.65 (dd, J ¼ 28.6, 8.5 Hz, 5c6: Yield, 78.7%; 1H NMR (400 MHz, DMSO‑d6) d 12.68 (s, 1H,
1H, H-Ar), 7.47 (d, J ¼ 8.3 Hz, 1H, H-Ar), 7.37e7.32 (m, 2H, H-Ar), NH), 12.40 (s, 1H, NH), 9.10 (s, 1H, HC¼C), 7.70 (s, 2H, H-Ar), 7.52 (s,
2.39 (s, 3H, -CH3); 13C NMR (126 MHz, DMSO‑d6) d 161.05 (C¼O), 1H, H-Ar), 7.39 (d, J ¼ 9.0 Hz, 1H, H-Ar), 7.29 (d, J ¼ 9.0 Hz, 1H, H-Ar),
149.69 (C¼N), 144.71, 142.33, 139.86 (C¼C), 137.40, 136.14, 133.75, 7.22 (d, J ¼ 6.1 Hz, 2H, H-Ar), 3.83 (s, 3H, -OCH3); 13C NMR
132.29 (C¼C), 128.87, 125.40, 120.44, 119.89, 119.51, 115.71, 115.05, (101 MHz, DMSO‑d6) d 160.79 (C¼O), 155.14, 148.34 (C¼N), 139.06
20.92 (CH3); ESI-HRMS, calculated m/zfor C17H12BrN3O [MþH]þ: (C¼C), 133.84 (C¼C), 122.55, 121.77, 120.63, 120.25, 117.06, 110.27,
354.0242, found: 354.0254. 56.00 (OCH3); ESI-HRMS, calculated m/z for C17H13N3O2 [MþH]þ:
5b5: Yield, 66.2%; 1H NMR (500 MHz, DMSO‑d6) d 12.09 (s, 1H, 292.1086, found: 292.1081.
NH), 8.91 (s, 1H, NH), 8.89 (s, 1H, HC¼C), 7.92 (t, J ¼ 2.6 Hz, 1H, H- 5d1: Yield, 73.9%; 1H NMR (500 MHz, DMSO‑d6) d 12.33 (s, 2H,
Ar), 7.58 (s, 1H, H-Ar), 7.43 (d, J ¼ 8.4 Hz, 1H, H-Ar), 7.27 (d, NH), 8.92 (s, 1H, HC¼C), 7.43 (s, 1H, H-Ar), 7.41 (s, 2H, H-Ar), 6.91 (s,
J ¼ 8.4 Hz, 1H, H-Ar), 6.87 (s, 1H, H-Ar), 6.79 (d, J ¼ 8.8 Hz, 1H, H-Ar), 1H, H-Ar), 6.14 (s, 2H, O-CH2-O), 2.31 (s, 6H, -CH3); 13C NMR
2.37 (s, 3H, -CH3); 13C NMR (126 MHz, DMSO‑d6) d 161.87 (C¼O), (126 MHz, DMSO‑d6) d 161.00 (C¼O), 151.67, 147.83 (C¼N), 144.44,
154.43, 151.54 (C¼N), 138.98 (C¼C), 138.44, 136.47, 134.45, 133.87, 138.55 (C¼C), 136.61, 130.77 (C¼C), 117.50, 114.31, 106.12, 102.57,
131.95 (C¼C), 129.18, 126.76, 124.94, 119.42, 115.72, 113.34, 113.17, 95.42 (CH2), 20.56 (CH3); ESI-HRMS, calculated m/z for C19H15N3O3
20.85 (CH3); ESI-HRMS, calculated m/zfor C17H12N4O3 [MH]-: [MþH]þ: 334.1192, found: 334.1188.
319.0831, found: 319.0860. 5d2: Yield, 73.1%; 1H NMR (400 MHz, DMSO‑d6) d 12.73 (s, 1H,
5b6: Yield, 79.2%; 1H NMR (400 MHz, DMSO‑d6) d 12.67 (s, 1H, NH), 12.44 (s, 1H, NH), 8.99 (s, 1H, HC¼C), 7.69 (s, 3H, H-Ar), 7.47 (s,
NH), 12.42 (s, 1H, NH), 9.04 (s, 1H, HC¼C), 7.73 (s, 2H, H-Ar), 7.67 (s, 1H, H-Ar), 6.92 (s, 1H, H-Ar), 6.16 (s, 2H, O-CH2-O); 13C NMR
1H, H-Ar), 7.46 (d, J ¼ 8.4 Hz, 1H, H-Ar), 7.36 (d, J ¼ 8.4 Hz, 1H, H-Ar), (101 MHz, DMSO‑d6) d 154.96 (C¼O), 153.51, 151.55, 149.05 (C¼N),
7.22 (s, 1H, H-Ar), 7.20 (s, 1H, H-Ar), 2.39 (s, 3H, -CH3); 13C NMR 147.94, 147.28, 137.62 (C¼C), 136.40, 134.00 (C¼C), 125.50, 124.56,
(101 MHz, DMSO‑d6) d 161.15 (C¼O), 148.32 (C¼N), 143.24, 139.24 113.71, 113.47, 111.90, 104.69, 103.79, 103.35 (CH2); ESI-HRMS,
(C¼C), 137.23, 134.88, 133.50, 132.22 (C¼C), 128.76, 122.73, 122.37, calculated m/z for C17H10FN3O3 [MH]-: 322.0628, found: 322.0657.
120.36, 119.57, 118.75, 115.66, 113.25, 20.95 (CH3); ESI-HRMS, 5d3: Yield, 68.3%; 1H NMR (400 MHz, DMSO‑d6) d 12.73 (s, 1H,
calculated m/z for C17H13N3O [MþH]þ: 276.1137, found: 276.1144. NH), 12.44 (s, 1H, NH), 8.99 (s, 1H, HC¼C), 7.69 (s, 3H, H-Ar), 7.47 (s,
5c1: Yield, 68.3%; 1H NMR (400 MHz, DMSO‑d6) d 12.44 (s, 1H, 1H, H-Ar), 6.92 (s, 1H, H-Ar), 6.16 (s, 2H, O-CH2-O); 13C NMR
NH), 12.36 (s, 1H, NH), 9.02 (s, 1H, HC¼C), 7.51 (s, 1H, H-Ar), 7.45 (s, (101 MHz, DMSO‑d6) d 160.87 (C¼O), 152.11 (C¼N), 144.54, 137.09,
2H, H-Ar), 7.37 (d, J ¼ 9.0 Hz, 1H, H-Ar), 7.27 (d, J ¼ 2.8 Hz, H-Ar), 126.63, 122.54, 116.66, 114.21, 106.26, 102.71, 95.40 (CH2); ESI-
3.83 (s, 3H, -OCH3), 2.32 (s, 6H, -CH3); 13C NMR (101 MHz, HRMS, calculated m/z for C17H10ClN3O3 [MH]-: 338.0333, found:
DMSO‑d6) d 160.78 (C¼O), 155.11, 147.42 (C¼N), 138.36 (C¼C), 338.0359.
133.64 (C¼C), 121.49, 120.90, 120.32, 117.00, 110.17, 55.99, 55.84 5d4: Yield, 73.8%; 1H NMR (500 MHz, DMSO‑d6) d 12.72 (s, 1H,
(OCH3), 20.63 (CH3); ESI-HRMS, calculated m/z for C19H17N3O2 NH), 12.42 (s, 1H, NH), 8.99 (s, 1H, HC¼C), 7.86 (s, 1H, H-Ar), 7.63 (s,
[MþH]þ: 320.1399, found: 320.1407. 1H, H-Ar), 7.46 (s, 1H, H-Ar), 7.31 (d, J ¼ 8.5 Hz, 1H, H-Ar), 6.92 (s,
5c2: Yield, 72.6%; 1H NMR (500 MHz, DMSO‑d6) d 12.76 (d, 1H, H-Ar), 6.16 (s, 2H, O-CH2-O); 13C NMR (126 MHz, DMSO‑d6)
J ¼ 11.3 Hz, 1H, NH), 12.40 (s, 1H, NH), 9.06 (d, J ¼ 10.8 Hz, 1H, d 160.89 (C¼O), 152.12 (C¼N), 144.55, 139.72, 137.12, 125.12, 114.56,
HC¼C), 7.68 (ddd, J ¼ 36.0, 8.6, 5.0 Hz, 1H, H-Ar), 7.54e7.40 (m, 2H, 114.22, 106.25, 102.69, 95.43 (CH2); ESI-HRMS, calculated m/z for
H-Ar), 7.38 (d, J ¼ 9.0 Hz, 1H, H-Ar), 7.28 (d, J ¼ 8.8 Hz, 1H, H-Ar), C17H10BrN3O3 [MþH]þ: 383.9984, found: 383.9977.
7.07 (t, J ¼ 8.4 Hz, 1H, H-Ar), 3.83 (s, 3H, -OCH3); 13C NMR (126 MHz, 5d5: Yield, 67.9%; 1H NMR (400 MHz, DMSO‑d6) d 12.48 (s, 1H,
DMSO‑d6) d 160.75 (C¼O), 155.18, 150.03 (C¼N), 140.00, 139.38 NH), 11.82 (s, 1H, NH), 7.66 (s, 1H, HC¼C), 7.50 (d, J ¼ 8.7 Hz, 1H, H-
(C¼C), 138.95, 133.92, 131.63 (C¼C), 121.92, 120.20, 117.09, 114.03, Ar), 7.15 (s, 1H, H-Ar), 7.08 (s, 1H, H-Ar), 6.84 (s, 1H, H-Ar), 6.60 (d,
110.78, 110.36, 104.07, 99.47, 56.03 (OCH3); ESI-HRMS, calculated m/ J ¼ 8.7 Hz, 1H, H-Ar), 6.06 (s, 2H, O-CH2-O); 13C NMR (101 MHz,
W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150 149

DMSO‑d6) d 161.88 (C¼O), 150.02 (C¼N), 144.57, 143.70, 137.51 derivatives containing a-aminophosphonates as potential antitumor agents,
Med. Chem. Comm. 8 (2017) 1158e1172.
(C¼C), 135.89, 135.22, 134.37 (C¼C), 127.17, 116.56, 113.78, 111.56,
[11] R. Arulmozhi, S. Sathishkumar, H.P. Kavitha, S. Arulmurugan, Biologically
105.62, 104.70, 102.09, 95.41 (CH2); ESI-HRMS, calculated m/z for active benzimidazole derivatives, Mini-Reviews Org. Chem. 12 (2015)
C17H10N4O5 [MH]-: 349.0573, found: 349.0597. 178e195.
5d6: Yield, 81.9%; 1H NMR (400 MHz, DMSO‑d6) d 12.58 (s, 1H, [12] A.S. Alpan, G. Sarıkaya, G. Çoban, S. Parlar, G. Armagan, V. Alptüzün, Mannich-
Benzimidazole derivatives as antioxidant and anticholinesterase inhibitors:
NH), 12.41 (s, 1H, NH), 9.00 (s, 1H, HC¼C), 7.69 (s, 1H, H-Ar), 7.62 (s, synthesis, biological evaluations, and molecular docking study, Arch. Phar-
1H, H-Ar), 7.47 (s, 1H, H-Ar), 7.17 (s, 2H, H-Ar), 6.93 (s, 1H, H-Ar), mazie 7 (2017) 350.
6.16 (s, 2H, O-CH2-O); 13C NMR (101 MHz, DMSO‑d6) d 160.98 [13] R. Zhou, E.B. Skibo, Chemistry of the pyrrolo[1,2-a]benzimidazole antitumor
agents: influence of the 7-substituent on the ability to alkylate DNA and
(C¼O), 151.89, 148.69 (C¼N), 144.48, 143.28, 139.21 (C¼C), 136.85, inhibit topoisomerase II, J. Med. Chem. 39 (1996) 4321e4331.
134.81 (C¼C), 122.43, 122.21, 118.51, 117.19, 114.25, 113.11, 106.21, [14] N.S. El-Gohary, M.I. Shaaban, Synthesis, antimicrobial, antiquorum-sensing
102.65, 95.40 (CH2); ESI-HRMS, calculated m/z for C17H11N3O3 and antitumor activities of new benzimidazole analogs, Eur. J. Med. Chem.
137 (2017) 439e449.
[MþH]þ: 306.0879, found: 306.0876. [15] €
F. Gümüş, G. Eren, L. Açık, A. Çelebi, F. Oztürk, Ş. Yılmaz, R.I. Saǧkan, S. Gür,

A. Ozkul, A. Elmal, Y. Elerman, Synthesis, cytotoxicity, and DNA interactions of
new cisplatin analogues containing substituted benzimidazole ligands, J. Med.
4.2. Biological assays
Chem. 52 (2009) 1345e1357.
[16] V. Kralova, V. Hanusov 
a, E. Rudolf, K. C  ova
an , L. Ska lova
, Flubendazole induces
The procedures for biological assays were described in the mitotic catastrophe and senescence in colon cancer cells in vitro, J. Pharm.
Supplementary Data (Part 2). The materials, instrumentation, and Pharmacol. 68 (2016) 208e218.
[17] V.L. Nayak, N. Nagesh, A. Ravikumar, C. Bagul, M.V.P.S. Vishnuvardhan,
methods for the cytotoxicity assay, cell cycle analysis, cell apoptosis V. Srinivasulu, A. Kamal, 2-aryl benzimidazole conjugate induced apoptosis in
analysis, western blot and p53-transfection assays were reported human breast cancer MCF-7 cells through caspase independent pathway,
previously [41e43]. Apoptosis 22 (2017) 118e134.
[18] L.W. Oberley, G.R. Bueftner, Role of superoxide dismutase in cancer: a review,
Canc. Res. 39 (1979) 1141e1149.
Acknowledgments [19] R. Abonia, J. Castillo, P. Cuervo, B. Insuasty, J. Quiroga, A. Ortíz, M. Nogueras,
J. Cobo, A Simple One-Pot Synthesis of new Imidazol-2-yl-1H-quinolin-2-ones
from the direct reaction of 2-chloroquinolin-3-carbaldehyde with aromatic o-
This study was supported by the National Natural Science diamines, Eur. J. Org Chem. 2 (2010) 317e325.
Foundation of China (No. 21501032), the Innovative Team & [20] B.F. Abdel-Wahab, R.E. Khidre, A.A. Farahat, A.eA.S. El-Ahl, 2-Chloroquinoline-
Outstanding Talent Program of Colleges and Universities in Guangxi 3-carbaldehydes: synthesis, reactions and applications, ARKIVOC 1 (2012)
211e276.
(2017-38), Guangxi Natural Science Foundation (Nos. [21] B.-Q. Zou, X. Lu, Q.-P. Qin, Y.-X. Bai, Y. Zhang, M. Wang, Y.-C. Liu, Z.-F. Chen,
2016GXNSFAA380300 and 2014GXNSFBA118050), Guangxi New H. Liang, Three novel transition metal complexes of 6-methyl-2-oxo-quino-
Century Ten, Hundred and Thousand Talents Project ([2017]42) and line-3-carbaldehyde thiosemicarbazone: synthesis, crystal structure, cyto-
toxicity, and mechanism of action, RSC Adv. 7 (2017) 17923e17933.
the State Key Laboratory Cultivation Base for Chemistry and Mo- [22] J.A. Choi, J.Y. Kim, J.Y. Lee, C.M. Kang, H.J. Kwon, Y.D. Yoo, T.W. Kim, Y.S. Lee,
lecular Engineering of Medicinal Resources (Ministry of Science and S.J. Lee, Induction of cell cycle arrest and apoptosis in human breast cancer
Technology of China, No. CMEMR2014-B14). cells by quercetin, Int. J. Oncol. 19 (2001) 837e844.
[23] M.B. Kastan, J. Bartek, Cell-cycle checkpoints and cancer, Nature 432 (2004)
316e322.
Appendix A. Supplementary data [24] S.U. Dighe, S. Khan, I. Soni, P. Jain, S. Shukla, R. Yadav, P. Sen, S.M. Meeran,
S. Batra, Synthesis of b-carboline-Based N-heterocyclic carbenes and their
antiproliferative and antimetastatic activities against human breast cancer
Supplementary data related to this article can be found at cells, J. Med. Chem. 58 (2015) 3485e3499.
https://doi.org/10.1016/j.ejmech.2018.07.066. [25] R. Boutros, V. Lobjois, B. Ducommun, CDC25 phosphatases in cancer cells: key
players? Good targets? Nat. Rev. Canc. 7 (2007) 495e507.
[26] F. Belluti, G. Fontana, L.D. Bo, N. Carenini, C. Giommarelli, F. Zunino, Design,
References synthesis and anticancer activities of stilbene-coumarin hybrid compounds:
identification of novel proapoptotic agents, Bioorg. Med. Chem. 18 (2010)
[1] A. Anighoro, J. Bajorath, G. Rastelli, Polypharmacology: challenges and op- 3543e3550.
portunities in drug discovery, J. Med. Chem. 57 (2014) 7874e7887. [27] M.O. Hengartner, The biochemistry of apoptosis, Nature 407 (2000) 770e776.
[2] E.S. Hamanaka, 4-Aryl-3-(heteroarylureido)-1,2-dihydro-2-oxo-quinoline [28] M.Y. Ye, G.Y. Yao, Y.M. Pan, Z.X. Liao, Y. Zhang, H.-S. Wang, Synthesis and
Derivatives as Antihypercholesterolemic and Antiatherosclerotic Agents, antitumor activities of novel a-aminophosphonate derivatives containing an
1996. Patent (US), US5565472. alizarin moiety, Eur. J. Med. Chem. 83 (2014) 116e128.
[3] N. Evens, G.G. Muccioli, N. Houbrechts, D.M. Lambert, A.M. Verbruggen, [29] R. Martin, J. Carvalho, E. Ibeas, M. Herna ndez, V. Ruiz-Gutierrez, M. Luisa-
L.K. Van, G.M. Bormans, Synthesis and biological evaluation of carbon-11- and Nieto, Acidic triterpenes compromise growth and survival of astrocytoma cell
fluorine-18-labeled 2-oxoquinoline derivatives for type 2 cannabinoid re- lines by regulating reactive oxygen species accumulation, Canc. Res. 67 (2007)
ceptor positron emission tomography imaging, Nucl. Med. Biol. 36 (2009) 3741e3751.
455e465. [30] L. Gomez, P.A. Thiebaut, M. Paillard, S. Ducreux, M. Abrial, C.C.D. Silva,
[4] J. DeRuiter, A.N. Brubaker, W.L. Whitmer, J.L.J. Stein, Synthesis and aldose A. Durand, M.R. Alam, F.V. Coppenolle, S.S. Sheu, M. Ovize, The SR/ER-
reductase inhibitory activity of substituted 2-oxoquinoline-1-acetic acid de- mitochondria calcium crosstalk is regulated by GSK3b during reperfusion
rivatives, J. Med. Chem. 29 (1986) 2024e2028. injury, Cell Death Differ. 23 (2016) 313e322.
[5] Y. Zhang, Y.-L. Fang, H. Liang, H.-S. Wang, K. Hu, X.-X. Liu, X.-H. Yi, Y. Peng, [31] Y.G. Shi, A structural view of mitochondria-mediated apoptosis, Nat. Struct.
Synthesis and antioxidant activities of 2-oxo-quinoline-3-carbaldehyde Biol. 8 (2001) 394e401.
Schiff-base derivatives, Bioorg. Med. Chem. Lett 23 (2013) 107e111. [32] R.G. Lea, N. al-Sharekh, M. Tulppala, H.O. Critchley, The immunolocalization of
[6] Y. Zhang, X.-H. Yi, Y.-L. Fang, X.-X. Liu, Preparation and Application of 2-oxo- Bcl-2 at the maternal-fetal interface in healthy and failing pregnancies, Hum.
quinoline-3-carbaldehyde Schiff-base Derivatives, Patent (China), 2014. Reprod. 12 (1997) 153e158.
ZL.201210407465X. [33] Y. Tsujimoto, J. Cossman, E. Jaffe, C.M. Croce, Involvement of the Bcl-2 gene in
[7] J. Rousell, E.B. Haddad, J.C. Mak, B.L. Webb, M.A. Giembycz, P.J. Barnes, Beta- human follicular lymphoma, Science 228 (1985) 1440e1443.
Adrenoceptor-medicated down-regulation of M2 muscarinic receptors: role of [34] D.M. Hockenbery, M. Zutter, W. Hickey, M. Nahm, S.J. Korsmeyer, Bcl-2 pro-
cyclic adenosine 5'-monophosphate-dependent protein kinase and protein tein is topographically restricted in tissues characterized by apoptotic cell
kinase C, Mol. Pharmacol. 49 (1996) 629e635. death, P. Natl. Acad. Sci USA. 88 (1991) 6961e6965.
[8] F.Q. Fang, H.S. Guo, J. Zhang, L.Y. Ban, J.W. Liu, P.Y. Yu, Anti-cancer effects of 2- [35] S. Qiao, T. Nagasaka, T. Harada, N. Nakashima, p53, Bax and Bcl-2 expression,
oxoquinoline derivatives on the HCT116 and LoVo human colon cancer cell and apoptosis in gestational trophoblast of complete hydatidiform mole,
lines, Mol. Med. Rep. 12 (2015) 8062e8070. Placenta 19 (1998) 361e369.
[9] Y.eL. Fang, Z.-L. Wu, M.-W. Xiao, Y.-T. Tang, K.-M. Li, Y. Jiao, J.-N. Xiang, A.- [36] K.W. Yip, J.C. Reed, Bcl-2 family proteins and cancer, Oncogene 27 (2008)
X. Hu, One-pot three-component synthesis of novel diethyl((2-oxo-1,2- 6398e6406.
dihydroquinolin-3-yl)(arylamino)-methyl)phosphonate as potential anti- [37] W.A. Siddiqui, A. Ahad, H. Ahsan, The mystery of BCL2 family: Bcl-2 proteins
cancer agents, Int. J. Mol. Sci. 17 (2016) 653. and apoptosis: an update, Arch. Toxicol. 89 (2015) 289e317.
[10] Y.-C. Yu, W.-B. Kuang, R.-Z. Huang, Y.-L. Fang, Y. Zhang, Z.-F. Chen, X.-L. Ma, [38] S. Haupt, M. Berger, Z. Goldberg, Y. Haupt, Apoptosisthe p53 network, J. Cell
Design, synthesis and pharmacological evaluation of new 2-oxoquinoline Sci. 116 (2003) 4077e4085.
150 W.-B. Kuang et al. / European Journal of Medicinal Chemistry 157 (2018) 139e150

[39] D.R. Green, G. Kroemer, Cytoplasmic functions of the tumor suppressor p53, [42] Z.-F. Chen, Q.-P. Qin, J.-L. Qin, Y.-C. Liu, K.-B. Huang, Y.-L. Li, T. Meng, G.-
Nature 458 (2009) 1127. H. Zhang, Y. Peng, X.-J. Luo, H. Liang, Stabilization of G-quadruplex DNA, in-
[40] T. Enoch, C. Norbury, Cellular responses to DNA damage: cell-cycle check- hibition of telomerase activity and tumor cell apoptosis of organoplatinum(II)
points, apoptosis and the roles of p53 and ATM, Trends Biochem. Sci. 20 complexes with oxoisoaporphine, J. Med. Chem. 58 (2015) 2159e2179.
(1995) 426. [43] Y.-L. Chu, C.T. Ho, J.G. Chung, R. Raghu, Y.C. Lo, L.Y. Sheen, Allicin induces anti-
[41] S.-X. Hua, R.-Z. Huang, M.-Y. Ye, Y.-M. Pan, G.-Y. Yao, Y. Zhang, H.-S. Wang, human liver cancer cells through p53 gene modulating apoptosis and auto-
Design, synthesis and in vitro evaluation of novel ursolic acid derivatives as phagy, J. Agric. Food Chem. 61 (2013) 9839e9848.
potential anticancer agents, Eur. J. Med. Chem. 95 (2015) 435e452.

Das könnte Ihnen auch gefallen