1 s2.0 S0223523419302259 Main

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

European Journal of Medicinal Chemistry 170 (2019) 195e202

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


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

Research paper

In vitro and in vivo antitumor activities of three novel binuclear


platinum(II) complexes with 40 -substituted-2,20 :60 ,200 -terpyridine
ligands
Qi-Pin Qin a, *, 1, Zhen-Feng Wang a, 1, Shu-Long Wang a, Dong-Mei Luo a, Bi-Qun Zou b, d,
Peng-Fei Yao c, Ming-Xiong Tan a, Hong Liang d, **
a
Guangxi Key Laboratory of Agricultural Resources, Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303
Jiaoyudong Road, Yulin, 537000, China
b
Department of Chemistry, Guilin Normal College, 9 Feihu Road, Gulin, 541001, China
c
Guangxi Colleges and Universities Key Laboratory of Regional Ecological Environment Analysis and Pollution Control of West Guangxi, College of
Chemistry and Environmental Engineering, Baise University, Baise, Guangxi, 533000, China
d
State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University,
15 Yucai Road, Guilin, 541004, China

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

Article history: Herein, we report the design and synthesis of three novel binuclear platinum(II) complexes, [Pt(tpbtpy)
Received 4 January 2019 Cl][Pt(DMSO)Cl3] (tpbtpy-Pt), [Pt(dthbtpy)Cl][Pt(DMSO)Cl3],CH3OH (dthbtpy-Pt), and [Pt(qlbtpy)Cl]
Received in revised form [Pt(DMSO)Cl3],CH3OH (qlbtpy-Pt) with 40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine (tpbtpy),
2 March 2019
40 -(3,5-bis (1,1-dimethylethyl)-2-hydroxy-benzaldehyde)-2,20 :60 ,200 -terpyridine (dthbtpy) and 40 -(2-
Accepted 5 March 2019
Available online 9 March 2019
quinolinecarboxaldehyde)-2,20 :60 ,200 -terpyridine (qlbtpy) as ligands, respectively. All three novel binu-
clear platinum(II) complexes tpbtpy-Pt, dthbtpy-Pt, and qlbtpy-Pt were characterized by single-crystal
X-ray diffraction analysis, spectroscopic analysis (ESI-MS, IR, 1H NMR), and elemental analysis. Addi-
Keywords:
40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 ;-
tionally, the cytotoxicity of tpbtpy-Pt, dthbtpy-Pt and qlbtpy-Pt was assessed with human non-small
terpyridine cell lung cancer cell line (NCIeH460 cells), yielding IC50 values in the range of 0.35e12.09 mM with
Platinum(II) complexes tpbtpy-Pt as the most potent and qlbtpy-Pt as the least potent complexes. Mechanistic studies indicated
Cell apoptosis that tpbtpy-Pt and dthbtpy-Pt induced apoptosis through mitochondrial dysfunction and telomerase
Telomerase inhibition inhibition. In a NCIeH460 xenograft model, when administered at 10.0 mg kg1 every 2 days, tpbtpy-Pt
Dysfunction of mitochondria was shown to significantly reduce tumor growth (tumor growth inhibition rate (IR) ¼ 70.1%, p < 0.05).
Therefore, tpbtpy-Pt is a promising Pt(II) complex for further translational studies and clinical evaluation
as an antitumor agent.
© 2019 Elsevier Masson SAS. All rights reserved.

1. Introduction complexes [3e10], such as platinum(IV)-benzhydrazide bis-conju-


gate, organometallic Pt(II) complexes [Pt(C^N)Cl(DMSO)] (C^N ¼
Pt(II)-based drugs are widely used in the clinical treatment of N,N-dimethyl-1-(2-aryl)methanamine- k2C2,N), Pt(IV) prodrugs,
lung, testicular (NT2), and ovarian cancers. [1e8] However, high cationic polynuclear Pt(II) complexes, and others. [1e12].
drug toxicity and drug resistance limit their clinical utility, and thus In addition, a large number of Pd(II), Fe(II/III), Pt(II), Zn(II), Ir(III),
researchers are encouraged to design and synthesize new Pt(II/IV) Cu(II), Au(III), Ru(II/III), V(III), and Os(III) terpyridyl complexes have
been designed and studied against cancer cells, and these com-
pounds showed better antitumor activity than the corresponding
terpyridyl ligands [13e43]. For instance, Che described a series of
* Corresponding author. cyclometalated platinum(II) complexes, [(C^N^N)PtII(N,N0 -n-
** Corresponding author.
Bu2NHC)]PF6 (HC^N^N ¼ 6-phenyl-2,20 -bipyridine), that are in-
E-mail addresses: qpqin2018@126.com (Q.-P. Qin), shulonghs@163.com
(S.-L. Wang), zoubiqun@163.com (B.-Q. Zou), yaopengfei1986@163.com (P.-F. Yao), hibitors of apoptosis (IAP) in vitro and in vivo [37]. Hussain
hliang@gxnu.edu.cn (H. Liang). investigated mixed-ligand copper(II) complex, [Cu(Fc-tpy)(CQ)]
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ejmech.2019.03.014
0223-5234/© 2019 Elsevier Masson SAS. All rights reserved.
196 Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202

NO3 (CQ ¼ 5-chloro-7-iodo-8-hydroxyquinoline), which is mainly


accumulated in the mitochondria [39]. Lippard and co-authors re-
ported that the cationic complexes [OsCl2N(tpy)]Cl
(tpy ¼ 2,2':60 ,200 -terpyridine) and [OsCl3N(phen)] (phen ¼ o-phe-
nanthroline) had activity against glioblastoma (GBM) initiating
tumor cells in vitro and in vivo [38]. Shul'pin and Rahiman sug-
gested that 40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine
(tpbtpy) copper(II) complexes [CuCl2(tpbtpy)] and [Cu(tpbtpy)2]Cl2
could induce HCT116 and MCF-7 cell apoptosis much more effec-
tively than other similar metal complexes [42,43]. However, no 40 -
(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine (tpbtpy) Pt(II)
complex has been reported in the literature, and the detailed
in vitro and in vivo anticancer mechanisms of 40 -(3-
thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine (tpbtpy) plati-
num(II) complexes remain unexplored.
Herein, we report the design, synthesis, and characterization of
three novel binuclear platinum(II) complexes, [Pt(tpbtpy)Cl] Fig. 1. ORTEP view of tpbtpy-Pt.
[Pt(DMSO)Cl3] (tpbtpy-Pt), [Pt(dthbtpy)Cl][Pt(DMSO)Cl3],CH3OH
(dthbtpy-Pt) and [Pt(qlbtpy)Cl] [Pt(DMSO)Cl3],CH3OH (qlbtpy-Pt)
with 40 -(3-thiophenecarboxaldehyde)- 2,20 :60 ,200 -terpyridine
(tpbtpy), 40 -(3,5-bis(1,1-dimethylethyl)-2-hydroxy- benzalde-
hyde)-2,20 :60 ,200 -terpyridine (dthbtpy) and 40 -(2-
quinolinecarboxaldehyde) 2,20 :60 ,200 -terpyridine (qlbtpy) as li-
gands, respectively (Scheme 1). In addition, the effects of the three
novel binuclear platinum(II) complexes, tpbtpy-Pt, dthbtpy-Pt and
qlbtpy-Pt, on cell proliferation and apoptosis in vitro and in vivo
were evaluated.

2. Results and discussion

2.1. Synthesis and characterization

The synthesis of 40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -


terpyridine (tpbtpy), 40 -(3,5-bis(1,1-dimethylethyl)-2-hydroxy-
benzaldehyde)-2,20 :60 ,200 -terpyridine (dthbtpy), and 40 -(2-
quinolinecarboxaldehyde)-2,20 :60 ,200 -terpyridine (qlbtpy) were
carried out as reported by Zou et al. (Scheme 1) [40e43]. Briefly, Fig. 2. ORTEP view of dthbtpy-Pt. The solvent CH3OH is omitted for clarity.
[Pt(tpbtpy)Cl][Pt(DMSO)Cl3] (tpbtpy-Pt), [Pt(dthbtpy)Cl]
[Pt(DMSO)Cl3],CH3OH (dthbtpy-Pt) and [Pt(qlbtpy)Cl][Pt(DMSO)
Cl3],CH3OH (qlbtpy-Pt) were synthesized by combining cis-
Pt(DMSO)2Cl2 (2.0 mmol) and tpbtpy, dthbtpy or qlbtpy (1.0 mmol)
in CH3OH (25.0 mL) and DMSO (0.2 mL) at 60  C for 6.0 h. All three
novel binuclear platinum(II) complexes tpbtpy-Pt, dthbtpy-Pt and
qlbtpy-Pt were characterized by single-crystal X-ray diffraction
analysis (Figs.13 and Tables S1S9), spectroscopic analyses (ESI-
MS, IR, 1H NMR), and elemental analysis (Fig. S1S9). Furthermore,
we showed that tpbtpy-Pt, dthbtpy-Pt and qlbtpy-Pt
(2.0  105 mol/L) were stable for 48 h in Tris-HCl buffer solution by
ESI-MS (Figs. 2, 4 and 6) [44e51].

Fig. 3. ORTEP view of qlbtpy-Pt. The solvent CH3OH is omitted for clarity.

2.2. Crystal structures


Scheme 1. Synthetic route of the syntheses of tpbtpy, dthbtpy, qlbtpy and their three
novel binuclear platinum(II) complexes tpbtpy-Pt, dthbtpy-Pt and qlbtpy-Pt. Re- The crystal structures of the three novel binuclear platinum(II)
agents and solvents are as follows: (a) ethanol, 25% NH3,H2O (aq), KOH; (b) cis- complexes tpbtpy-Pt, dthbtpy-Pt and qlbtpy-Pt were determined
Pt(DMSO)2Cl2 (2.0 mmol), 25.0 mL CH3OH and 0.2 mL DMSO, 60  C, 6.0 h.
Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202 197

by X-ray crystallography, as shown in Figs.13. A list of angels ( ) lung cancer cell line (NCIeH460 cells). In NCIeH460 cells, upon
and selected bond lengths (Å) is shown in Tables S1S9, which treatment with tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) for
were within the expected range [44e51]. The tpbtpy-Pt, dthbtpy- 24 h, down-regulation of c-myc and hTERT was observed compared
Pt or qlbtpy-Pt complex consists of one cationic [Pt(tpbtpy)Cl]þ, with control, especially with tpbtpy-Pt (0.35 mM) treated cells
[Pt(dthbtpy)Cl]þ or [Pt(qlbtpy)Cl]þ, and one anionic [Pt(DMSO) (Fig. 4B and C). Furthermore, telomerase activity was decreased after
Cl3], where the two central Pt(II) atoms were arranged in a four- treatment with tpbtpy-Pt (0.35 mM, inhibitory rates of 62.50%)) and
coordinated distorted square planar geometry. dthbtpy-Pt (5.53 mM, inhibitory rates of 52.25%)) at 24 h (Fig. 4A),
which may be related to the inhibition of c-myc and hTERT (Fig. 4)
2.3. In vitro cytotoxicity [47,56e67]. Finally, G1 accumulation increased rapidly at 24 h
(Fig. 5AeC), and the G1 phase populations observed in the tpbtpy-Pt
We studied the cytotoxicity of tpbtpy, dthbtpy, qlbtpy, cis- (0.35 mM) and dthbtpy-Pt (5.53 mM) groups were 69.06% and
Pt(DMSO)2Cl2, three novel binuclear platinum(II) complexes 66.73%, respectively, and cyclin D1 and CDK2 were down-regulated
tpbtpy-Pt, dthbtpy-Pt, qlbtpy-Pt, and cisplatin at various con- at 24 h in NCIeH460 cells (Fig. 5D and E). At the same time, the
centrations (0.325, 0.75, 1.25, 2.5, 5.0, 10.0 and 20.0 mM) against quinone oxidoreductase isozyme I (NQO1) Pt(IV) complexes caused
human NCIeH460 (non-small cell lung cancer cells), T-24 (bladder arrest at S phase which was different from that of tpbtpy-Pt
cancer cells), SK-OV-3 (ovarian cancer cells), A549 (lung carcinoma (0.35 mM) and dthbtpy-Pt (5.53 mM), demonstrating that the anti-
cancer cells), and human liver Hl-7702 normal cells. The IC50 values cancer mechanism of tpbtpy-Pt (0.35 mM) and dthbtpy-Pt
was obtained by the MTT assay after exposure to tpbtpy, dthbtpy, (5.53 mM) could distinguish from NQO1 Pt(IV) complexes. In general,
qlbtpy, cis-Pt(DMSO)2Cl2, tpbtpy-Pt, cisplatin, dthbtpy-Pt, and this result suggested that inhibition of telomerase activity by
qlbtpy-Pt for 48 h (Table 1). It was evident that tpbtpy-Pt con- tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) in NCIeH460 cancer
taining the 40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine cells led to partial growth arrest [47,56e67].
(tpbtpy) ligand demonstrated the greatest anticancer activity
(IC50 ¼ 0.35 ± 0.08 mM) against human NCIeH460 cells, a non-small 2.5. Dysfunction of mitochondria
cell lung cancer cell line. The antiproliferative activity of the three
novel binuclear platinum(II) complexes tpbtpy-Pt, dthbtpy-Pt, It is well-known that mitochondrial changes, including in-
qlbtpy-Pt, and cisplatin increased in the order of tpbtpy- creases in ROS generation, loss of DJm (mitochondrial membrane
Pt > dthbtpy-Pt > qlbtpy-Pt > cisplatin, which may be due to the
different substituent group (tpbtpy > dthbtpy > qlbtpy) will affect
the planarity of the angle among the pyridine ring planes in three
novel binuclear Pt(II) complexes, especially 3-
thiophenecarboxaldehyde substitution in the tpbtpy ligand of the
tpbtpy-Pt complex, very similar to the results obtained by Sadler
et al. [52e55]. Notably, the three novel binuclear platinum(II)
complexes tpbtpy-Pt, dthbtpy-Pt, and qlbtpy-Pt displayed higher
antiproliferative activity against the human non-small cell lung
cancer cell line (NCIeH460 cells) as compared to the human liver
HL-7702 normal cells (Table 1).

2.4. Inhibition of telomerase activity and the related proteins by


tpbtpy-Pt and dthbtpy-Pt in NCIeH460 cells

A large body of literature has reported that inhibition of telo-


merase activity by chemical complexes or compounds leads to the
decrease in the level of hTERT and c-myc proteins [56e66], conse-
quently inducing tumor cell cycle arrest and down-regulating
related-proteins [47,56e67]. Thus, we next studied whether two
novel binuclear platinum(II) complexes tpbtpy-Pt (0.35 mM) and
Fig. 4. Modulation of telomerase activity by tpbtpy-Pt (0.35 mM) and dthbtpy-Pt
dthbtpy-Pt (5.53 mM) could down-regulate related-proteins, (5.53 mM) in NCIeH460 cancer cells. (A) Analysis of telomerase activity. (B and C) c-myc
including c-myc, cyclin D1, hTERT and CDK2, inhibit telomerase ac- and hTRET proteins in tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) treated cells
tivity, and induce tumor cell cycle arrest in a human non-small cell were detected by Western blot.

Table 1
The IC50 values (mM) of tpbtpy, dthbtpy, qlbtpy, cis-Pt(DMSO)2Cl2, three novel binuclear platinum(II) complexes tpbtpy-Pt, dthbtpy-Pt, qlbtpy-Pt, and cisplatin towards
human NCIeH460 (non-small cell lung cancer cells), T-24 (bladder cancer cells), SK-OV-3 (ovarian cancer cells), A549 (lung carcinoma cancer cells) and human liver Hl-7702
normal cells after incubation for 48 h.

Compound NCIeH460 SK-OV-3 T-24 A549 HL-7702

tpbtpy 25.41 ± 0.81 34.83 ± 1.08 19.61 ± 0.69 14.21 ± 0.19 38.22 ± 0.66
tpbtpy-Pt 0.35 ± 0.08 4.52 ± 0.29 1.24 ± 0.55 6.03 ± 1.22 40.22 ± 1.58
dthbtpy 30.11 ± 0.56 35.22 ± 2.01 20.88 ± 0.44 16.31 ± 0.66 35.11 ± 1.69
dthbtpy-Pt 5.53 ± 0.22 12.83 ± 0.96 7.58 ± 1.01 11.28 ± 0.77 38.54 ± 0.96
qlbtpy 40.23 ± 1.89 39.23 ± 1.75 45.26 ± 2.11 49.99 ± 1.33 30.58 ± 0.34
qlbtpy-Pt 12.09 ± 1.03 18.69 ± 0.49 15.29 ± 1.14 20.62 ± 1.72 35.03 ± 2.25
cis-Pt(DMSO)2Cl2 >150 >150 >150 >150 >150
cisplatinb 14.11 ± 1.56 12.18 ± 1.71 11.09 ± 1.88 17.36 ± 1.16 19.33 ± 1.14

The IC50 was defined as mean ± SD (standard deviation of the average value) from five independent assays.
198 Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202

Fig. 5. The change in cell cycle and related-proteins in NCIeH460 cancer cells after treatment with tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) for 24 h (AC) Cell cycle status of
NCIeH460 cancer cells after treatment with tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) were analyzed by flow cytometry. (D and E) Cell cycle related-proteins in tpbtpy-Pt
(0.35 mM) and dthbtpy-Pt (5.53 mM) treated cells were detected by Western blot.

potential), the increased level of intracellular Ca2þ, and increases/ to evaluate tumor cell apoptosis upon treatment with tpbtpy-Pt
decreases of the apoptosis proteins, play very important roles in (0.35 mM) and dthbtpy-Pt (5.53 mM) for 24 h in NCIeH460 cells. As
drug induced cancer cell apoptosis [68e86]. Therefore, the effects shown in Fig. 9, NCIeH460 cancer cells were treated with tpbtpy-
of tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) on mitochondrial Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) for 24 h. The percentage of
properties of NCIeH460 cancer cells were examined by flow- apoptotic (Q2þQ4) cells increased to 98.5% for tpbtpy-Pt, and 19.5%
cytometry, fluorescence microscopy, and Western blot. As shown for dthbtpy-Pt as compared with the control (8.7%). NCIeH460 cell
in Fig. 6, upon treatment of NCIeH460 cells with tpbtpy-Pt apoptosis was greater when treated with tpbtpy-Pt as compared
(0.35 mM) and dthbtpy-Pt (5.53 mM), the fluorescent intensity with treatment with dthbtpy-Pt.
decreased remarkably (low DJm, from right to left), as compared
with the control group (high DJm in the control group leads to JC-1
emission of red fluorescence). After treatment of NCIeH460 tumor 2.7. The tpbtpy-Pt suppressed NCIeH460 tumor xenograft growth
cells, the fluorescence of DCF (Fig. 7aec) and Fluo-3 AM (Fig. 7df) in vivo
increased. These data suggested that tpbtpy-Pt (0.35 mM) and
dthbtpy-Pt (5.53 mM) can increase intracellular free Ca2þ and ROS The in vivo anti-cancer activity of tpbtpy-Pt was examined.
levels. Furthermore, levels of bcl-2 were decreased, and levels of Nude mice bearing NCIeH460 cell xenografts were treated with the
cytochrome c and apaf-1 were increased after treatment of highly soluble tpbtpy-Pt at a dose of 10.0 mg/kg once every 2 days
NCIeH460 cells with tpbtpy-Pt (0.35 mM) and dthbtpy-Pt by intraperitoneal injection over 13 days with no adverse effects
(5.53 mM) (Fig. 8). In summary, these complexes induced observed [47,60,85,87e95]. The treatment was found to inhibit
NCIeH460 cell apoptosis through mitochondrial dysfunction tumor growth by 70.1% (p < 0.05) as compared to with the vehicle
pathways and tpbtpy-Pt showed stronger effect than dthbtpy-Pt. group (5% DMSO in saline, v/v), which exhibited better anticancer
activity than that of cisplatin (tumor growth inhibition (TGI), 31.8%;
tumor growth inhibition rate (IR), 25.5%) [88]. Importantly, no
2.6. Cell apoptosis with Annex V/PI double staining method significant body weight loss, and no other adverse effects and
mouse death were observed after treatment with tpbtpy-Pt
Based on the above results, PI/Annex V double staining was used (10.0 mg/kg/q2d) at these doses (Fig. 10AeD and Table S10S12).

Fig. 6. The changes of DJm were studied after NCIeH460 tumor cells were treated with tpbtpy-Pt (0.35 mM, a) and dthbtpy-Pt (5.53 mM, b) for 24 h. (a) The cancer cells were
imaged by a flow-cytometry.
Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202 199

Fig. 7. Intracellular ROS (ac) and Ca2þ (df) levels in NCIeH460 cells (a,d) exposed to tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) for 24 h. The fluorescent intensity of DCF and
Fluo-3 AM were determined by fluorescence microscopy (400  ).

addition, tpbtpy-Pt significantly inhibited NCIeH460 tumor


xenograft growth (TGI ¼ 70.1%, p < 0.05), suggesting that it has the
potential to be further developed as a Pt-based anticancer drug.

4. Experimental methods

4.1. Synthesis

4.1.1. Synthesis of the tpbtpy, dthbtpy and qlbtpy ligands


Fig. 8. (A and B) Western blot analysis of bcl-2, cytochrome c, and apaf-1 in Synthesis of 40 -(3-thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyr-
NCIeH460 cells treated with tpbtpy-Pt (0.35 mM) and dthbtpy-Pt (5.53 mM) for 24 h.
b-actin was used as the internal control.
idine (tpbtpy), 40 -(3,5-bis(1,1-dimethylethyl)-2-hydroxy-benzalde-
hyde)-2,20 :60 ,200 -terpyridine (dthbtpy) and 40 -(2-
quinolinecarboxaldehyde)-2,20 :60 ,200 -terpyridine (qlbtpy) were
carried out according to published methods [42e44].

4.1.2. Synthesis and characterization of tpbtpy-Pt, dthbtpy-Pt and


qlbtpy-Pt
cis-Pt(DMSO)2Cl2 (2.0 mmol) was mixed with 1.0 mmol 40 -(3-
thiophenecarboxaldehyde)-2,20 :60 ,200 -terpyridine (tpbtpy), 40 -(3,5-
bis(1,1-dimethylethyl)-2-hydroxy-benzaldehyde)-2,20 :60 ,200 -terpyr-
idine (dthbtpy) and 40 -(2-quinolinecarboxaldehyde)-2,20 :60 ,200 -ter-
pyridine (qlbtpy) in CH3OH (25.0 mL) and DMSO (0.2 mL) at 60  C
Fig. 9. Apoptosis in NCHeH460 cells upon (a) exposure to control conditions, (b)
tpbtpy-Pt (0.35 mM), and (c) dthbtpy-Pt (5.53 mM) for 24 h as determined by flow
for 6.0 h to yield red brown rod-shaped crystals of [Pt(tpbtpy)Cl]
cytometry. [Pt(DMSO)Cl3] (tpbtpy-Pt), [Pt(dthbtpy)Cl][Pt(DMSO)Cl3],CH3OH
(dthbtpy-Pt) and [Pt(qlbtpy)Cl][Pt(DMSO)Cl3],CH3OH (qlbtpy-Pt),
which were isolated and characterized.
3. Conclusion Data for tpbtpy-Pt. Yield: 90.89%. ESI-MS: m/z ¼ 545.9 for
[tpbtpy þ Pt þ Cl]þ. IR (KBr): 3427, 3078, 2999, 2912, 1610, 1559,
In summary, three novel binuclear platinum(II) complexes, 1523, 1476, 1439, 1371, 1243, 1130, 1023, 876, 783, 750, 716,
[Pt(tpbtpy)Cl][Pt(DMSO)Cl3] (tpbtpy-Pt), [Pt(dthbtpy)Cl] 570 cm1. Elemental analysis: calcd (%) for C21H19Cl4OPt2S2: C
[Pt(DMSO)Cl3],CH3OH (dthbtpy-Pt) and [Pt(qlbtpy)Cl][Pt(DMSO) 27.25, H 2.07, N 4.54; found: C 27.20, H 2.11, N 4.52.1H NMR
Cl3],CH3OH (qlbtpy-Pt) with 40 -(3-thiophenecarboxaldehyde)- (500 MHz, DMSO‑d6) d 8.93 (d, J ¼ 2.1 Hz, 2H), 8.86e8.79 (m, 2H),
2,20 :60 ,200 -terpyridine (tpbtpy), 40 -(3,5-bis(1,1- dimethylethyl)-2- 8.78 (d, J ¼ 7.9 Hz, 2H), 8.74 (dd, J ¼ 2.9, 1.3 Hz, 1H), 8.50 (td, J ¼ 7.8,
hydroxy-benzaldehyde)-2,20 :60 ,200 -terpyridine (dthbtpy) and 40 - 1.7 Hz, 2H), 8.06 (d, J ¼ 5.1 Hz, 1H), 7.90 (tt, J ¼ 5.0, 1.8 Hz, 3H), 2.54
(2-quinolinecarboxaldehyde)-2,20 :60 ,200 -terpyridine (qlbtpy) as the (s, 6H).
respective ligands were synthesized and fully characterized by Data for dthbtpy-Pt. Yield: 92.06%. ESI-MS: m/z ¼ 668.0 for
spectroscopic methods. MTT assay of tpbtpy-Pt, dthbtpy-Pt and [dthbtpy þ Pt þ Cl]þ. IR (KBr): 3787, 3434, 2955, 1608, 1478, 1413,
qlbtpy-Pt showed excellent IC50 values in the range of 1137, 1018, 785, 658, 509, 442 cm1. Elemental analysis: calcd (%)
0.35e12.09 mM, comparable to that of cisplatin (14.11 ± 1.56 mM). for C31H37Cl4O2Pt2S: C 35.54, H 3.56, N 4.01; found: C 35.52, H 3.60,
Interestingly, tpbtpy-Pt and dthbtpy-Pt exerted antitumor effects, N 3.98.1H NMR (500 MHz, DMSO‑d6) d 8.98 (dd, J ¼ 5.7, 1.5 Hz, 2H),
including induction of mitochondrial dysfunction, inhibition of 8.79e8.73 (m, 4H), 8.70 (s, 1H), 8.52 (td, J ¼ 7.9, 1.6 Hz, 2H), 7.98
telomerase activity and related-proteins, and induction of cell cycle (ddd, J ¼ 7.5, 5.6, 1.4 Hz, 2H), 7.44 (d, J ¼ 2.4 Hz, 1H), 7.28 (d,
arrest at G1 phase in the order of tpbtpy-Pt > dthbtpy-Pt. In J ¼ 2.3 Hz, 1H), 2.54 (s, 6H), 1.46 (s, 9H), 1.34 (s, 9H).
200 Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202

Fig. 10. The tpbtpy-Pt complex suppressed NCIeH460 tumor xenograft growth in vivo. (A) The tumor volumes of NCIeH460 xenograft-bearing mice after treatment with tpbtpy-Pt
(10.0 mg/kg/q2d) via intraperitoneal injection of six doses over 13 days. (B) Body weight of mice in the tpbtpy-Pt (10.0 mg/kg/q2d) and vehicle (5% DMSO in saline, v/v) groups. (C)
Tumor weight of tpbtpy-Pt (10.0 mg/kg/q2d) and vehicle (5% DMSO in saline, v/v) groups were recorded after 13 days. (**) P < 0.05, drug-treated group vs. the vehicle control. (D)
Tumors treated with tpbtpy-Pt (10.0 mg/kg/q2d) and vehicle (5% DMSO in saline, v/v) were dissected from the mice.

Data for qlbtpy-Pt. Yield: 85.62%. ESI-MS: m/z ¼ 590.9 for References
[qlbtpy þ Pt þ Cl]þ. IR (KBr): 3493, 3056, 1606, 1551, 1477, 1416,
lez-Canto , J.A. Woods,
1142, 1123, 11032, 1018, 834, 783, 752, 690, 630, 477, 441 cm1. [1] A. Gandioso, E. Shaili, A. Massaguer, G. Artigas, A. Gonza
P.J. Sadler, V. Marcha n, An integrin-targeted photoactivatable Pt(IV) complex
Elemental analysis: calcd (%) for C26H22Cl4OPt2S: C 32.18, H 2.28, N as a selective anticancer pro-drug: synthesis and photoactivation studies,
5.77; found: C 32.14, H 2.26, N 5.80.1H NMR (500 MHz, DMSO‑d6) Chem. Commun. 51 (2015) 9169e9172.
d 9.39 (d, J ¼ 13.2 Hz, 2H), 8.95 (dd, J ¼ 22.7, 6.8 Hz, 4H), 8.77 (d, [2] Y. Gothe, T. Marzo, L. Messori, N. Metzler-Nolte, Iridium(I) compounds as
prospective anticancer agents: solution chemistry, antiproliferative profiles
J ¼ 8.6 Hz, 1H), 8.64e8.54 (m, 3H), 8.27 (d, J ¼ 8.5 Hz, 2H), 8.14 (d, and protein interactions for a series of iridium(I) N-heterocyclic carbine
J ¼ 8.2 Hz, 1H), 8.07e7.92 (m, 3H), 7.79 (s, 1H), 3.32 (s, 6H). complexes, Chem. Eur J. 22 (2016) 12487e12494.
[3] Y.-R. Zheng, K. Suntharalingam, T.C. Johnstone, S.J. Lippard, Encapsulation of
Pt(IV) prodrugs within a Pt(II) cage for drug delivery, Chem. Sci. 6 (2015)
4.2. Methods 1189e1193.
[4] D.Y.Q. Wong, J.Y. Lau, W.H. Ang, Harnessing chemoselective imine ligation for
tethering bioactive molecules to platinum(IV) prodrugs, Dalton Trans. 41
The in vitro anti-tumor activity of tpbtpy-Pt (0.35 mM) and (2012) 6104e6111.
dthbtpy-Pt (5.53 mM) was assessed in NCIeH460 cells after 24 h [5] J. Noh, B. Kwon, E. Han, M. Park, W. Yang, W. Cho, W. Yoo, G. Khang, D. Lee,
treatment as described by Ulukaya, Chao, Sadler, Che, and Liang Amplification of oxidative stress by a dual stimuli-responsive hybrid drug
enhances cancer cell death, Nat. Commun. 6 (2015) 6907, https://doi.org/
[47,48,55,63,83,90]. In vivo antitumor activity of tpbtpy-Pt 10.1038/ncomms7907.
(10.0 mg/kg/q2d) against a NCIeH460 tumor xenograft was deter- [6] L.H. Hurley, DNA and its associated processes as targets for cancer therapy,
mined as reported by Che and Samuelson [47,60,85,87e94]. Nat. Rev. Canc. 2 (2002) 188e200.
[7] O. Pinato, C. Musetti, C. Sissi, Pt-based drugs: the spotlight will be on proteins,
Metall 6 (2014) 380e395.
Acknowledgement [8] H. Kostrhunova, J. Florian, O. Novakova, A.F.A. Peacock, P.J. Sadler, V. Brabec,
DNA Interactions of monofunctional organometallic osmium(II) antitumor
complexes in cell-free media, J. Med. Chem. 51 (2008) 3635e3643.
We thank the National Natural Science Foundation of China [9] Z. Liu, A. Habtemariam, A.M. Pizarro, S.A. Fletcher, A. Kisova, O. Vrana,
(Nos. 21867017, 21861014 and 21761033), the Natural Science L. Salassa, P.C.A. Bruijnincx, G.J. Clarkson, V. Brabec, P.J. Sadler, Organometallic
half-sandwich iridium anticancer complexes, J. Med. Chem. 54 (2011)
Foundation of Guangxi (No. 2018GXNSFBA138021, 3011e3026.
2018GXNSFBA281188 and 2016GXNSFBA380237), the Innovative [10] V. Milacic, D. Chen, L. Ronconi, K.R. Landis-Piwowar, D. Fregona, Q.P. Dou,
Team & Outstanding Talent Program of Colleges and Universities in A novel anticancer gold(III) dithiocarbamate compound inhibits the activity of
a purified 20S proteasome and 26S proteasome in human breast cancer cell
Guangxi (2014-49 and 2017-38), the basic skills improvement cultures and xenografts, Can. Res. 66 (2006) 10478e10486.
project for the young and middle-aged teachers in Guangxi colleges [11] B. Rosenberg, L. VanCamp, J.E. Trosko, V.H. Mansour, Platinum compounds: a
and universities (2019KY0605 and 2019KY1163) as well as the new class of potent antitumour agents, Nature 222 (1969) 385e386.
[12] A. Zamora, S.A. Perez, M. Rothemund, V. Rodríguez, R. Schobert, C. Janiak,
scientific research project of Guilin Normal College (KYA201804)
J. Ruiz, Exploring the influence of the aromaticity on the anticancer and
for the financial support. antivascular activities of organoplatinum(II) complexes, Chem. Eur J. 23
(2017) 5614e5625.
[13] I. Eryazici, C.N. Moorefield, G.R. Newkome, Square-planar Pd(II), Pt(II), and
Appendix A. Supplementary data Au(III) terpyridine complexes: their syntheses, physical properties, supra-
molecular constructs, and biomedical activities, Chem. Rev. 108 (2008)
1834e1895.
Supplementary data to this article can be found online at [14] D. Co 
ci
c, S. Jovanovic, S. Radisavljevi
c, J. Korzekwa, A. Scheurer, R. Puchta,
https://doi.org/10.1016/j.ejmech.2019.03.014.
Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202 201

D. Baskic, D. Todorovi c, S. Popovic, S. Mati


c, B. Petrovic, New monofunctional [40] B. Deka, T. Sarkar, S. Banerjee, A. Kumar, S. Mukherjee, S. Deka, K.K. Saikia,
platinum(II) and palladium(II) complexes: studies of the nucleophilic substi- A. Hussain, Novel mitochondria targeted copper(II) complexes of ferrocenyl
tution reactions, DNA/BSA interaction, and cytotoxic activity, J. Inorg. Bio- terpyridine and anticancer active 8-hydroxyquinolines showing remarkable
chem. 189 (2018) 91e102. cytotoxicity, DNA and protein binding affinity, Dalton Trans. 46 (2017)
[15] S. Gama, I. Rodrigues, F. Mendes, I.C. Santos, E. Gabano, B. Klejevskaja, 396e409.
J. Gonzalez-Garcia, M. Ravera, R. Vilar, A. Paulo, Anthracene-terpyridine metal [41] H.-H. Zou, L. Wang, Z.-X. Long, Q.-P. Qin, Z.-K. Song, T. Xie, S.-H. Zhang, Y.-
complexes as new G-quadruplex DNA binders, J. Inorg. Biochem. 160 (2016) C. Liu, B. Lin, Z.-F. Chen, Preparation of 4-([2,20 :60 ,20 0 -terpyridin]-40 -yl)-N,N-
275e286. diethylaniline NiII and PtII complexes and exploration of their in vitro cyto-
[16] P. Shi, Q. Jiang, Y. Zhao, Y. Zhang, J. Lin, L. Lin, J. Ding, Z. Guo, DNA binding toxic activities, Eur. J. Med. Chem. 108 (2016) 1e12.
properties of novel cytotoxic gold(III) complexes of terpyridine ligands: the [42] Q.-P. Qin, T. Meng, M.-X. Tan, Y.-C. Liu, S.-L. Wang, B.-Q. Zou, H. Liang, Syn-
impact of steric and electrostatic effects, J. Biol. Inorg. Chem. 11 (2006) thesis, characterization and biological evaluation of six highly cytotoxic
745e752. ruthenium(III) complexes with 40 -substituted-2,20 :60 ,200 -terpyridine, Med.
[17] G. Sava, A. Bergamo, P.J. Dyson, Metal-based antitumour drugs in the post- Chem. Commun. 9 (2018) 525e533.
genomic era: what comes next? Dalton Trans. 40 (2011) 9069e9075. [43] D. Mahendiran, R.S. Kumar, V. Viswanathan, D. Velmurugan, A.K. Rahiman,
[18] Q. Jiang, J. Zhu, Y. Zhang, N. Xiao, Z. Guo, DNA binding property, nuclease Targeting of DNA molecules, BSA/c-Met tyrosine kinase receptor and anti-
activity and cytotoxicity of Zn(II) complexes of terpyridine derivatives, Bio- proliferative activity of bis(terpyridine)copper(II) complexes, Dalton Trans.
metals 22 (2009) 297e305. 45 (2016) 7794e7814.
[19] L. Messori, F. Abbate, G. Marcon, P. Orioli, M. Fontani, E. Mini, T. Mazzei, [44] K. Czerwin  ska, B. Machura, S. Kula, S. Krompiec, K. Erfurt, C. Roma-Rodrigues,
S. Carotti, T. O0 Connell, P. Zanello, Gold(III) complexes as potential antitumor A.R. Fernandes, L.S. Shul0 pina, N.S. Ikonnikove, G.B. Shul0 pin, Copper(II) com-
agents: solution chemistry and cytotoxic properties of some selected gold(III) plexes of functionalized 2,2’:6’,2’’-terpyridines and 2,6-di(thiazol-2-yl)pyri-
compounds, J. Med. Chem. 43 (2000) 3541e3548. dine: structure, spectroscopy, cytotoxicity and catalytic activity, Dalton Trans.
[20] K.K.-W. Lo, M.-W. Louie, K.Y. Zhang, Design of luminescent iridium(III) and 46 (2017) 9591e9604.
rhenium(I) polypyridine complexes as in vitro and in vivo ion, molecular and [45] L. Fang, X. Qin, J. Zhao, S. Gou, Construction of dual stimuli-responsive plati-
biological probes, Coord. Chem. Rev. 254 (2010) 2603e2622. num(IV) hybrids with NQO1 targeting ability and overcoming cisplatin
[21] V.M. Manikandamathavan, V. Rajapandian, A.J. Freddy, T. Weyhermüller, resistance, Inorg. Chem. 58 (2019) 2191e2200.
V. Subramanian, B.U. Nair, Effect of coordinated ligands on antiproliferative [46] M. Proetto, W. Liu, A. Hagenbach, U. Abram, R. Gust, Synthesis, characteriza-
activity and DNA cleavage property of three mononuclear Cu(II)-terpyridine tion and in vitro antitumour activity of a series of novel platinum(II) com-
complexes, Eur. J. Med. Chem. 57 (2012) 449e458. plexes bearing Schiff base ligands, Eur. J. Med. Chem. 53 (2012) 168e175.
[22] D. Lazic, A. Arsenijevi 
c, R. Puchta, Z.D. Bugar ci
c, Ana Rilak, DNA binding [47] Q.-P. Qin, S.-L. Wang, M.-X. Tan, Y.-C. Liu, T. Meng, B.-Q. Zou, H. Liang, Syn-
properties, histidine interaction and cytotoxicity studies of water soluble thesis of two platinum(II) complexes with 2-methyl-8-quinolinol derivatives
ruthenium(II) terpyridine complexes, Dalton Trans. 45 (2016) 4633e4646. as ligands and study of their antitumor activities, Eur. J. Med. Chem. 161
[23] K. Abdi, H. Hadadzadeh, M. Weil, M. Salimi, Mononuclear copper(II) complex (2019) 334e342.
with terpyridine and an extended phenanthroline base, [Cu(tpy)(dppz)]2þ: [48] Q.-P. Qin, J.-L. Qin, T. Meng, W.-H. Lin, C.-H. Zhang, Z.-Z. Wei, J.-N. Chen, Y.-
synthesis, crystal structure, DNA binding and cytotoxicity activity, Polyhedron C. Liu, H. Liang, Z.-F. Chen, High in vivo antitumor activity of cobalt oxoisoa-
31 (2012) 638e648. porphine complexes by targeting G-quadruplex DNA, telomerase and dis-
[24] I. Ott, On the medicinal chemistry of gold complexes as anticancer drugs, rupting mitochondrial functions, Eur. J. Med. Chem. 124 (2016) 380e392.
Coord. Chem. Rev. 253 (2009) 1670e1681. [49] Q.-P. Qin, S.-L. Wang, M.-X. Tan, Z.-F. Wang, X.-L. Huang, Q.-M. Wei, B.-B. Shi,
[25] A. Winter, M. Gottschaldt, G.R. Newkome, U.S. Schubert, Terpyridines and B.-Q. Zou, H. Liang, Synthesis and antitumor mechanisms of two novel plati-
their complexes with first row transition metal ions: cytotoxicity, nuclease num(II) complexes with 3-(20 -benzimidazolyl)-7-methoxycoumarin, Metall
activity and self-assembly of biomacromolecules, Curr. Top. Med. Chem. 12 10 (2018) 1160e1169.
(2012) 158e175. [50] Q.-P. Qin, S.-L. Wang, M.-X. Tan, Z.-F. Wang, D.-M. Luo, B.-Q. Zou, Y.-C. Liu, P.-
[26] F. Darabi, H. Hadadzadeh, J. Simpson, A. Shahpiri, A water-soluble Pd(II) F. Yao, H. Liang, Novel tacrine platinum(II) complexes display high anticancer
complex with a terpyridine ligand: experimental and molecular modeling activity via inhibition of telomerase activity, dysfunction of mitochondria, and
studies of the interaction with DNA and BSA; and in vitro cytotoxicity in- activation of the p53 signaling pathway, Eur. J. Med. Chem. 158 (2018)
vestigations against five human cancer cell lines, New J. Chem. 40 (2016) 106e122.
9081e9097. [51] E.-J. Gao, L. Wang, M.-C. Zhu, L. Liu, W.-Z. Zhang, Synthesis, characterization,
[27] T. Lazarevi 
c, A. Rilak, Z.D. Bugar
ci
c, Platinum, palladium, gold and ruthenium interaction with DNA and cytotoxicity in vitro of the complexes
complexes as anticancer agents: current clinical uses, cytotoxicity studies and [M(dmphen)(CO3)],H2O [M¼Pt(II), Pd(II)], Eur. J. Med. Chem. 45 (2010)
future perspectives, Eur. J. Med. Chem. 142 (2017) 8e31. 311e316.
[28] J.A. Todd, P. Turner, E.J. Ziolkowski, L.M. Rendina, (2,20 :60 ,20 0 -terpyridine) [52] E.E. Aranda, T.A. Matias, K. Araki, A.P. Vieira, E.A. de Mattos, P. Colepicolo,
platinum(II) complexes containing (thioalkyl)dicarba-closo-dodecabor- C.P. Luz, F.L.N. Marques, A.M. da Costa Ferreira, Design, syntheses, charac-
ane(12) ligands, Inorg. Chem. 44 (2005) 6401e6408. terization, and cytotoxicity studies of novel heterobinuclear oxindolimine
[30] A. Rilak, I. Bratsos, E. Zangrando, J. Kljun, I. Turel, Z..D. Bugarcic, E. Alessio, New copper(II)-platinum(II) complexes, J. Inorg. Biochem. 165 (2016) 108e118.
water-soluble ruthenium(II) terpyridine complexes for anticancer activity: [53] J. Li, Z. Tian, X. Ge, Z. Xu, Y. Feng, Z. Liu, Design, synthesis, and evaluation of
synthesis, characterization, activation kinetics, and interaction with guanine fluorine and naphthyridinee based half-sandwich organoiridium/ruthenium
derivatives, Inorg. Chem. 53 (2014) 6113e6126. complexes with bioimaging and anticancer activity, Eur. J. Med. Chem. 163
[31] E. Ulukaya, F. Ari, K. Dimas, E.I. Ikitimur, E. Guney, V.T. Yilmaz, Anti-cancer (2019) 830e839.
activity of a novel palladium(II) complex on human breast cancer cells in vitro [54] D. Wang, S.J. Lippard, Cellular processing of platinum anticancer drugs, Nat.
and in vivo, Eur. J. Med. Chem. 46 (2011) 4957e4963. Rev. Drug Discov. 4 (2005) 307e320.
[32] Q. Zhang, X. Tian, Z. Hu, C. Brommesson, J. Wu, H. Zhou, S. Li, J. Yang, Z. Sun, [55] J. Li, L. Guo, Z. Tian, M. Tian, S. Zhang, K. Xu, Y. Qian, Z. Liu, Novel halfsandwich
Y. Tian, K. Uvdal, A series of Zn(II) terpyridine complexes with enhanced two- iridium(III) imino-pyridyl complexes showing remarkable in vitro anticancer
photon-excited fluorescence for in vitro and in vivo bioimaging, J. Mater. activity, Dalton Trans. 46 (2017) 15520e15534.
Chem. B 3 (2015) 7213e7221. [56] Z. Liu, P.J. Sadler, Organoiridium complexes: anticancer agents and catalysts,
[33] M.M. Milutinovi c, A. Rilak, I. Bratsos, O. Klisuric, M. Vranes, N. Gligorijevic, Acc. Chem. Res. 47 (2014) 1174e1185.
S. Radulovi 
c, Z.D. Bugar ci
c, New 40 -(4-chlorophenyl)-2,20 :60 ,200 -terpyridine [57] X. Wang, C.-L. Zhou, J.-W. Yan, J.-Q. Hou, S.-B. Chen, T.-M. Ou, L.-Q. Gu, Z.-
ruthenium(II) complexes: synthesis, characterization, interaction with DNA/ S. Huang, J.-H. Tan, Synthesis and evaluation of quinazolone derivatives as a
BSA and cytotoxicity studies, J. Inorg. Biochem. 169 (2017) 1e12. new class of c-KIT G-quadruplex binding ligands, ACS Med. Chem. Lett. 4
[34] X. Wang, Z. Guo, Towards the rational design of platinum(II) and gold(III) (2013) 909e914.
complexes as antitumour agents, Dalton Trans. (2008) 1521e1532. [58] Y.-J. Lu, T.-M. Ou, J.-H. Tan, J.-Q. Hou, W.-Y. Shao, D. Peng, N. Sun, X.-D. Wang,
[35] S.D. Cummings, Platinum complexes of terpyridine: interaction and reactivity W.-B. Wu, X.-Z. Bu, Z.-S. Huang, D.-L. Ma, K.-Y. Wong, L.-Q. Gu, 5-N-methyl-
with biomolecules, Coord. Chem. Rev. 253 (2009) 1495e1516. ated quindoline derivatives as telomeric G-quadruplex stabliziing ligands:
[36] X. Wang, Z. Guo, Targeting and delivery of platinum-based anticancer drugs, effects of 5-N-positive charge on quadruplex binding affinity and cell prolif-
Chem. Soc. Rev. 42 (2013) 202e224. eration, J. Med. Chem. 51 (2008) 6381e6397.
[37] M.J. Hannon, P.S. Green, D.M. Fisher, P.J. Derrick, J.L. Beck, S.J. Watt, S.F. Ralph, [59] Y. Xu, Chemistry in human telomere biology: structure, function and targeting
M.M. Sheil, P.R. Barker, N.W. Alcock, R.J. Price, K.J. Sanders, R. Pither, J. Davis, of telomere DNA/RNA, Chem. Soc. Rev. 40 (2011) 2719e2740.
A. Rodger, An estrogen-platinum terpyridine conjugate: DNA and protein [60] Q. Cao, Y. Li, E. Freisinger, P.Z. Qin, R.K.O. Sigel, Z.-W. Mao, G-quadruplex DNA
binding and cellular delivery, Chem. Eur J. 12 (2006) 8000e8013. targeted metal complexes acting as potential anticancer drugs, Inorg. Chem.
[38] R.W.-Y. Sun, A.L.-F. Chow, X.-H. Li, J.J. Yan, S.S.-Y. Chui, C.-M. Che, Luminescent Front. 4 (2017) 10e32.
cyclometalated platinum(II) complexes containing ̌́
̌ N-heterocyclic carbene li- [61] Z.-F. Chen, Q.-P. Qin, J.-L. Qin, J. Zhou, Y.-L. Li, N. Li, Y.-C. Liu, H. Liang, Water-
gands with potent in vitro and in vivo anti-cancer properties accumulate in soluble ruthenium(II) complexes with chiral 4-(2,3-dihydroxypropyl)-form-
cytoplasmic structures of cancer cells, Chem. Sci. 2 (2011) 728e736. amide oxoaporphine (FOA): in vitro and in vivo anticancer activity by stabi-
[39] G. Berger, K. Grauwet, H. Zhang, A.M. Hussey, M.O. Nowicki, D.I. Wang, lization of G-Quadruplex DNA, inhibition of telomerase activity, and induction
E.A. Chiocca, S.E. Lawler, S.J. Lippard, Anticancer activity of osmium(VI) nitrido of tumor cell apoptosis, J. Med. Chem. 58 (2015) 4771e4789.
complexes in patient-derived glioblastoma initiating cells and in vivo mouse [62] A. De Cian, G. Cristofari, P. Reichenbach, E. De Lemos, D. Monchaud, M.-
models, Cancer Lett. 416 (2018) 138e148. P. Teulade-Fichou, K. Shin-ya, L. Lacroix, J. Lingner, J.-L. Mergny, Reevaluation
202 Q.-P. Qin et al. / European Journal of Medicinal Chemistry 170 (2019) 195e202

of telomerase inhibition by quadruplex ligands and their mechanisms of ac- saccharinate complexes of tertiary phosphines with phenyl and cyclohexyl
tion, Proc. Natl. Acad. Sci. Unit. States Am. 104 (2007) 17347e17352. groups targeting mitochondria and DNA, Eur. J. Med. Chem. 155 (2018)
[63] J.E. Reed, A.A. Arnal, S. Neidle, R. Vilar, Stabilization of G-quadruplex DNA and 609e622.
inhibition of telomerase activity by square-planar nickel(II) complexes, J. Am. [81] S.P. Wisnovsky, J.J. Wilson, R.J. Radford, M.P. Pereira, M.R. Chan, R.R. Laposa,
Chem. Soc. 128 (2006) 5592e5993. S.J. Lippard, S.O. Kelley, Targeting mitochondrial DNA with a platinum-based
[64] L. Xu, X. Chen, J. Wu, J. Wang, L. Ji, H. Chao, Dinuclear ruthenium(II) complexes anticancer agent, Chem. Biol. 20 (2013) 1323e1328.
that induce and stabilise G-quadruplex DNA, Chem. Eur J. 21 (2015) [82] S. Fulda, L. Galluzzi, G. Kroemer, Targeting mitochondria for cancer therapy,
4008e4020. Nat. Rev. Drug Discov. 9 (2010) 447e464.
[65] M. Akiyama, O. Yamada, S. Akita, M. Urashima, J. Horiguchi-Yamada, T. Ohno, [83] D. Wan, B. Tang, Y.-J. Wang, B.-H. Guo, H. Yin, Q.-Y. Yi, Y.-J. Liu, Synthesis and
H. Mizoguchi, Y. Eto, H. Yamada, Ectopic expression of c-myc fails to overcome anticancer properties of ruthenium(II) complexes as potent apoptosis in-
downregulation of telomerase activity induced by herbimycin A, but ectopic ducers through mitochondrial disruption, Eur. J. Med. Chem. 139 (2017)
hTERT expression overcomes it, Leukemia 14 (2000) 1260e1265. 180e190.
[66] I. Listerman, J. Sun, F.S. Gazzaniga, J.L. Lukas, E.H. Blackburn, The major reverse [84] V.T. Yilmaz, C. Icsel, M. Aygun, M. Erkisa, E. Ulukaya, Pd(II) and Pt(II) sac-
transcriptaseeincompetent splice variant of the human telomerase protein charinate complexes of bis(diphenylphosphino)propane/butane: synthesis,
inhibits telomerase activity but protects from apoptosis, Cancer Res. 73 (2013) structures, antiproliferative activity and mechanism of action, Eur. J. Med.
2817e2828. Chem. 158 (2018) 534e547.
[67] T.L. Jacobs, E.S. Epel, J. Lin, E.H. Blackburn, O.M. Wolkowitz, D.A. Bridwell, [85] W.-Y. Zhang, Q.-Y. Yi, Y.-J. Wang, F. Du, M. He, B. Tang, D. Wan, Y.-J. Liu, H.-
A.P. Zanesco, S.R. Aichele, B.K. Sahdra, K.A. MacLean, B.G. King, P.R. Shaver, L. Huang, Photoinduced anticancer activity studies of iridium(III) complexes
E.L. Rosenberg, E. Ferrer, B.A. Wallace, C.D. Saron, Psychoneuroendocrinology targeting mitochondria and tubules, Eur. J. Med. Chem. 151 (2018) 568e584.
36 (2011) 664e681. [86] B. Purushothaman, P. Arumugam, H. Ju, G. Kulsi, A.A.S. Samson, J.M. Song,
[68] A.K.P. Taggart, S.-C. Teng, V.A. Zakian, Est1p as a cell cycle-regulated activator Novel ruthenium(II) triazine complex [Ru(bdpta)(tpy)]2þ co-targeting drug
of telomere-bound telomerase, Science 297 (2002) 1023e1026. resistant GRP78 and subcellular organelles in cancer stem cells, Eur. J. Med.
[69] C. Balachandran, J. Haribabu, K. Jeyalakshmi, N.S.P. Bhuvanesh, R. Karvembu, Chem. 156 (2018) 747e759.
N. Emi, S. Awale, Nickel(II) bis(isatin thiosemicarbazone) complexes induced [87] D. Yugandhar, V.L. Nayak, S. Archana, K.C. Shekar, A.K. Srivastava, Design,
apoptosis through mitochondrial signaling pathway and G0/G1 cell cycle ar- synthesis and anticancer properties of novel oxa/azaspiro[4,5]trienones as
rest in IM-9 cells, J. Inorg. Biochem. 182 (2018) 208e221. potent apoptosis inducers through mitochondrial disruption, Eur. J. Med.
[70] P.E. Czabotar, G. Lessene, A. Strasser, J.M. Adams, Control of apoptosis by the Chem. 101 (2015) 348e357.
Bcl-2 protein family: implications for physiology and therapy, Nat. Rev. Mol. [88] K.-H. Diehl, R. Hull, D. Morton, R. Pfister, Y. Rabemampianina, D. Smith, J.-
Cell Biol. 15 (2014) 49e63. M. Vidal, C.V. De Vorstenbosch, A good practice guide to the administration of
[71] D. Merino, S.W. Lok, J.E. Visvader, G.J. Lindeman, Targeting Bcl-2 to enhance substances and removal of blood, including routes and volumes, J. Appl.
vulnerability to therapy in estrogen receptor-positive breast cancer, Oncogene Toxlcol. 21 (2001) 15e23.
35 (2016) 1877e1887. [89] Z.-F. Chen, Q.-P. Qin, J.-L. Qin, Y.-C. Liu, K.-B. Huang, Y.-L. Li, T. Meng, G.-
[72] K.M. Debatin, D. Poncet, G. Kroemer, Chemotherapy: targeting the mito- H. Zhang, Y. Peng, X.-J. Luo, H. Liang, Stabilization of G-quadruplex DNA, in-
chondrial cell death pathway, Oncogene 21 (2002) 8786e8803. hibition of telomerase activity and tumor cell apoptosis of organoplatinum(II)
[73] J.C. Reed, Bcl-2-family proteins and hematologic malignancies: history and complexes with oxoisoaporphine, J. Med. Chem. 58 (2015) 2159e2179.
future prospects, Blood 111 (2008) 3322e3330. [90] D. Palanimuthu, S.V. Shinde, K. Somasundaram, A.G. Samuelson, In vitro and
[74] A.R.D. Delbridge, S. Grabow, A. Strasser, D.L. Vaux, Thirty years of Bcl-2: in vivo anticancer activity of copperbis(thiosemicarbazone) complexes, J. Med.
translating cell death discoveries into novel cancer therapies, Nat. Rev. Chem. 56 (2013) 722e734.
Canc. 16 (2016) 99e109. [91] T.T.-H. Fong, C.-N. Lok, C.Y.-S. Chung, Y.-M.E. Fung, P.-K. Chow, P.-K. Wan, C.-
[75] W.-X. Chen, X.-D. Song, S.-F. He, J. Sun, J.-X. Chen, T. Wu, Z.-W. Mao, Ru(II) M. Che, Cyclometalated palladium(II) N-heterocyclic carbene complexes:
complexes bearing guanidinium ligands as potent anticancer agents, J. Inorg. anticancer agents for potent in vitro cytotoxicity and in vivo tumor growth
Biochem. 164 (2016) 91e98. suppression, Angew. Chem. Int. Ed. 55 (2016) 1e6.
[76] Q. Yu, Y. Liu, L. Xu, C. Zheng, F. Le, X. Qin, Y. Liu, J. Liu, Ruthenium(II) poly- [92] Y. Wang, J. Xiao, H. Zhou, S. Yang, X. Wu, C. Jiang, Y. Zhao, D. Liang, X. Li,
pyridyl complexes: cellular uptake, cell image and apoptosis of HeLa cancer G. Liang, A novel monocarbonyl analogue of curcumin, (1E,4E)-1,5-bis(2,3-
cells induced by double targets, Eur. J. Med. Chem. 82 (2014) 82e95. dimethoxyphenyl)penta-1,4-dien-3-one, induced cancer cell H460 apoptosis
[77] H. Zhang, S. Gou, J. Zhao, F. Chen, G. Xu, X. Liu, Cytotoxicity profile of novel via activation of endoplasmic reticulum stress signaling pathway, J. Med.
sterically hindered platinum(II) complexes with (1R,2R)-N1,N2-dibutyl-1,2- Chem. 54 (2011) 3768e3778.
diaminocyclohexane, Eur. J. Med. Chem. 96 (2015) 187e195. [93] J.-Q. Wang, P.-Y. Zhang, L.-N. Ji, H. Chao, A ruthenium(II) complex inhibits
[78] F.-U. Rahman, M.Z. Bhatti, A. Ali, H.-Q. Duong, Y. Zhang, B. Yang, S. Koppireddi, tumor growth in vivo with fewer side-effects compared with cisplatin,
Y. Lin, H. Wang, Z.-T. Li, D.-W. Zhang, Homo- and heteroleptic Pt(II) complexes J. Inorg. Biochem. 146 (2015) 89e96.
of ONN donor hydrazone and 4-picoline: a synthetic, structural and detailed [94] S. Go€schl, E. Schreiber-Brynzak, V. Pichler, K. Cseh, P. Heffeter, U. Jungwirth,
mechanistic anticancer investigation, Eur. J. Med. Chem. 143 (2018) M.A. Jakupec, W. Berger, B.K. Keppler, Comparative studies of oxaliplatin-
1039e1052. based platinum(IV) complexes in different in vitro and in vivo tumor
[79] G. Gao, Q.P. Dou, G1 Phase-dependent expression of Bcl-2 mRNA and protein models, Metall 9 (2017) 309e322.
correlates with chemoresistance of human cancer cells, Mol. Pharmacol. 58 [95] A. Weiss, R.H. Berndsen, M. Dubois, C. Muller, R. Schibli, A.W. Griffioen,
(2000) 1001e1010. P.J. Dyson, P. Nowak-Sliwinska, In vivo anti-tumor activity of the organome-
[80] V.T. Yilmaz, C. Icsel, O.R. Turgut, M. Aygun, M. Erkisa, M.H. Turkdemir, tallic ruthenium(II)-arene complex [Ru(h6-p-cymene)Cl2(pta)] (RAPTA-C) in
E. Ulukaya, Synthesis, structures and anticancer potentials of platinum(II) human ovarian and colorectal carcinomas, Chem. Sci. 5 (2014) 4742e4748.

You might also like