Sei sulla pagina 1di 16

This is an open access article published under an ACS AuthorChoice License, which permits

copying and redistribution of the article or any adaptations for non-commercial purposes.

Article

pubs.acs.org/jmc

Synthesis, Anticancer Activity, and Genome Profiling of Thiazolo


Arene Ruthenium Complexes
Adriana Grozav,*,† Ovidiu Balacescu,*,‡ Loredana Balacescu,‡ Thomas Cheminel,§
Ioana Berindan-Neagoe,‡,∥ and Bruno Therrien*,§

Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, Victor Babes Str. 41, RO-400012 Cluj-Napoca,
Romania

Department of Functional Genomics, Proteomics and Experimental Pathology, The Oncology Institute “Prof Dr. Ion Chiricuta”,
34-36 Republicii Str, RO-400015, Cluj-Napoca, Romania
§
Institut de Chimie, Université de Neuchâtel, 51 Avenue de Bellevaux, CH-2000 Neuchâtel, Switzerland

Research Center of Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu″ University of Medicine and
Pharmacy, 23 Marinescu Str, RO-400337 Cluj-Napoca, Romania
*
S Supporting Information

ABSTRACT: Sixteen hydrazinyl-thiazolo arene ruthenium complexes of the general formula [(η6-p-cymene)Ru(N,N′-
hydrazinyl-thiazolo)Cl]Cl were synthesized. All complexes were tested in vitro for their antiproliferative activity on three tumor
cell lines (HeLa, A2780, and A2780cisR) and on a noncancerous cell line (HFL-1). A superior cytotoxic activity of the ruthenium
complexes as compared to cisplatin and oxaliplatin, on both cisplatin-sensitive and cisplatin resistant ovarian cancer cells, was
observed. In addition, the biological activity of two selected derivatives was evaluated using microarray gene expression assay and
ingenuity pathway analysis. p53 signaling was identified as an important pathway modulated by both arene ruthenium
compounds. New activated molecules such as FAS, ZMAT3, PRMT2, BBC3/PUMA, and PDCD4, whose overexpressions are
correlated with overcoming resistance to cisplatin therapy, were also identified as potential targets. Moreover, the arene
ruthenium complexes can be used in association with cisplatin to prevent cisplatin resistance development and synergistically to
induce cell death in ovarian cancer cells.

■ INTRODUCTION
Ovarian cancer is the eighth leading cause of cancer-related
treatment for more than 30 years.2,3 Its inefficiency on
platinum-resistant tumors is a major disadvantage, which led
deaths in women worldwide and the second cause of to the search for alternative agents to resolve this drawback.
gynecologic cancer-related deaths, responsible for over Since then, complexes of other transition metals have been
150000 deaths per year around the world.1 While often designed as alternatives to cisplatin. Among these candidates,
combined with other techniques such as radiotherapy or ruthenium derivatives have undoubtedly stood out.
surgery, chemotherapy is certainly the most widely used Over the last few decades, many examples of anticancer
treatment for cancer. However, this technique has several ruthenium compounds have been reported, some of which are
limitations, mainly the lack of selectivity of the drug, which already into clinical trials, such as the well-known complexes
often also kills healthy cells and thus leads to undesirable side [imiH]trans-[Ru(N-imi)(S-dmso)Cl4] (NAMI-A)4 and [Na]
effects (hair loss, fatigue, nausea, etc.). The resistance of trans-[Ru(N-ind)2Cl4] (NKP1339).5 Those complexes present
cancerous cells to drugs is also a major problem in
chemotherapy; one of those drugs is the well-known transition Received: June 4, 2015
metal complex cisplatin, which has been used in cancer Published: October 21, 2015

© 2015 American Chemical Society 8475 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Scheme 1. Synthesis of Complexes 1−16a

a
R1, R2, and R3 functional groups are given in Table 1.

Table 1. Identification of the Functional Groups R1−R3 Attached to the Hydrazinyl-thiazole Compounds L1−L16

R1 R2 R3 R1 R2 R3
L1 Ph Me H L9 4-MeO-C6H4 Me H
L2 Ph Ph H L10 4-MeO-C6H4 Ph H
L3 Ph Me COMe L11 4-MeO-C6H4 Me COOEt
L4 2,4-Cl2−C6H3 CH2COOEt H L12 4-MeO-C6H4 Me COMe
L5 2,4-Cl2−C6H3 Me COMe L13 3-Cl-C6H4 Me H
L6 2,4-Cl2−C6H3 Me COOEt L14 3-Cl-C6H4 Me COMe
L7 4-HO-C6H4 Me H L15 3−Cl-C6H4 Ph H
L8 4-HO-C6H4 Ph H L16 3−Cl-C6H4 Me COOEt

some advantages over cisplatin, such as their activity against h; and (b) a microwave-assisted synthetic method, whose
cisplatin- resistant cancerous cell lines and their higher optimal reaction conditions were established after several
selectivity for cancerous over healthy cells, leading to reduced experiments, varying the solvent (methanol, acetonitrile, and
side effects.6,7 The mechanisms of action of ruthenium dichloromethane), the temperature (40 °C, 60 °C, 82 °C, and
complexes appear to be different from platinum drugs used in 100 °C), and the reaction time (0.5 h, 1 h, 1.5 h, and 2 h). The
the clinic, and many of them are not known yet. However, best yields for the complexes were obtained after 0.5 h of
some mechanisms have been proposed for the anticancer microwave irradiation at 60 °C and using dichloromethane as
activity of ruthenium complexes, such as the inhibition of solvent (see Table S1). The two alternative synthetic methods
metastasis,8,9 interaction with DNA,10 interaction with (microwave irradiation and conventional synthesis) are
proteins,11 production of reactive oxygen species,12 inhibition comparable for the resulting yield (50−80%), but the
of topoisomerase,13 induction of apoptosis,14 and antiangio- microwave-assisted method demands a shorter reaction time
genic effects.15,16 (30 min vs 10 h). In order to obtain a greener synthesis, one
In recent years, much attention was given to compounds has tried to replace standard organic solvents by water.
with pharmacophore hydrazine-thiazolo or thiazolo moieties Unfortunately, in water, the complexes could only be recovered
due to the identification of several thiazolo lead compounds in traces.
showing antiproliferative activity,17 inhibition of Bcl-XL,18 All complexes were isolated as their chloride salts, and they
inhibition of tautomerase,19 inhibition of metastatic cancer have been fully characterized by 1H NMR spectroscopy, mass
cell migration and invasion,20 and antitumor activity.21,22 spectrometry, IR spectroscopy, elemental analysis, and for 12
Therefore, in this study, we have combined a p-cymene by a single-crystal X-ray structure analysis. No attempt to
ruthenium unit with a hydrazinyl-thiazolo ligand to generate a separate the cationic enantiomers was performed, and the
series of organometallic compounds with significant antitumor chiral-at-metal complexes were isolated and used as racemic
activity, taking advantage of the synthetic versatility of mixtures. All complexes are stable in D2O and DMSO solutions
hydrazinyl-thiazolo derivatives and the promising biological as well as in biological media (aqueous solution containing
activity of ruthenium complexes. RPMI 1640 medium with 5% fetal calf serum, glutamine, and


antibiotics), showing no decomplexation of the hydrazinyl-
thiazolo ligands. The 1H NMR spectra of all complexes show,
RESULTS AND DISCUSSION
in addition to the signals of the p-cymene ring, the
A series of monocationic arene ruthenium complexes (1−16) characteristic signals of the ligand at higher chemical shifts
containing hydrazinyl-thiazole bidentate ligands (L1−L16) were related to its free form because of the deshielding effect
prepared (Scheme 1). The synthesis of the complexes was produced by the arene ruthenium unit. Typical signals for the
realized by two methods: (a) a conventional synthetic method ligands after complexation are a singlet at around 9 ppm
involving one equivalent of the ruthenium dimer (η6-p- associated with the proton of the azomethine moiety (CHN)
cymene)2Ru2Cl4 and two equivalents of the hydrazinyl-thiazole and the proton from the hydrazine moiety (N-NH), which is
derivatives (L1−L16) in methanol at room temperature for 10 the most deshielded one, appearing as a singlet at around 15
8476 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

ppm (in some cases the signal is not observed due to the bond distances are 2.090(6) (thiazole) and 2.123(6) Å
deuterium exchange). In the case of the complexes where R3 = (hydrazine), and these values are similar to those found in
H, the corresponding proton of the thiazolo ring is observed as analogous N,N′-coordinated pyridyl-thiazolo arene ruthenium
a singlet around 6.5 ppm. complexes.23−25 In the solid state, an angle of 34.6(3)° is
Crystals suitable for a single-crystal X-ray structure analysis observed between the plane formed by the hydrazinyl-thiazolo
were obtained for [(η6-p-cymene)Ru(L12)Cl]Cl. The molecular unit and the plane of the methoxyphenyl group.
structure of the cation is presented in Figure 1, together with In Vitro Antiproliferative Activity. The hydrazinyl-
thiazolo arene ruthenium complexes 1−16 were tested in
vitro for their antiproliferative activity on four cell lines: HeLa
(human cervical cancer cells), A2780 (human ovarian cancer
cells), A2780cisR (cisplatin-resistant human ovarian cancer
cells), and HFL-1 (noncancer cells, human fibroblast). IC50
values were used to determine the antiproliferative activity of
the complexes (IC50 = drug concentration necessary for 50%
inhibition of cell viability); these values are listed in Table 2.
Moreover, the calculated partition coefficients (log P) of the
hydrazinyl-thiazole compounds L1−L16 together with their IC50
values on HeLa cells17 are presented in Table 3, thus giving an
approximation of the lipophilicity of the ligands and their
cytotoxicity. Cisplatin and oxaliplatin, two metal-based drugs
currently used in the clinic to treat cancer, were used as
controls, and their IC50 values are also reported in Table 2.
Figure 1. ORTEP drawing of complex 12 at 35% probability level, The results obtained on the three tumor cell lines (HeLa,
with hydrogen atoms being omitted for clarity. Selected bond lengths A2780, and A2780cisR) show a promising profile for the
(Å) and angles (deg): Ru(1)−Cl(1) 2.408(2), Ru(1)−N(1) 2.090(6), antiproliferative activity of all complexes, most of them having a
Ru(1)−N(3) 2.123(6), N(2)−N(3) 1.396(7), and N(3)−C(17) cytotoxic activity at concentrations significantly lower than that
1.299(8); Cl(1)−Ru(1)−N(1) 85.61(15), Cl(1)−Ru(1)−N(3) of cisplatin and oxaliplatin (Tables S2 and S3). Moreover, by
90.04(15), N(1)−Ru(1)−N(3) 76.2(2), N(1)−C(16)−N(2) comparing the antiproliferative effect produced by the
120.5(6), C(16)−N(1)−Ru(1) 112.1(4), C(14)−N(1)−Ru(1) complexes on the tumor cell lines A2780 and A2780cisR, we
133.8(5), C(17)−N(3)−Ru(1) 133.7(5), N(2)−N(3)−Ru(1)
noticed that at almost the same concentrations (see Table 2), a
111.1(4), N(2)−N(3)−C(17) 114.2(6), and C(13)−S(1)−C(16)
88.2(3). similar effect is observed on both cell lines, suggesting a mode
of action different from that of cisplatin. By analyzing the effect
of the treatment with the complexes on the normal fibroblasts
selected geometrical parameters. In complex 12, the ruthenium cell line HFL-1, a significant reduction of cytotoxicity can be
center shows a typical pseudotetrahedral geometry with the noticed for complexes 4 (IC50 = 181 μM) and 14 (IC50 = 36.6
hydrazinyl-thiazolo ligand being N,N′-coordinated. The Ru−N μM) (Figure 2). When comparing the cytotoxic effect produced

Table 2. Cytotoxic Activity of Complexes 1−16 in HeLa, A2780, A2780cisR, and HFL-1 Cells
IC50 (μM)a
compd HeLa A2780 A2780cisR HFL-1
cisplatin 20.10 ± 0.02 11.30 ± 0.02 41.13 ± 1.24 9.50 ± 0.02
oxaliplatin 12.72 ± 0.02 10.10 ± 0.03 16.57 ± 0.02 13.53 ± 0.02
1 7.42 ± 0.01 9.65 ± 0.01 12.95 ± 0.04 18.48 ± 0.02
2 2.55 ± 0.02 2.49 ± 0.02 0.93 ± 0.02 6.27 ± 0.01
3 11.42 ± 0.02 6.99 ± 0.02 2.08 ± 0.03 6.43 ± 0.02
4 3.65 ± 0.03 12.21 ± 0.03 12.62 ± 0.01 181 ± 0.89
5 7.98 ± 0.01 50.08 ± 0.03 55.58 ± 0.03 47.33 ± 0.32
6 0.84 ± 0.25 2.86 ± 0.01 6.06 ± 0.02 4.52 ± 0.01
7 19.40 ± 0.03 6.13 ± 0.25 6.19 ± 0.01 49.40 ± 0.31
8 6.87 ± 0.23 5.25 ± 0.03 4.24 ± 0.01 7.30 ± 0.03
9 7.95 ± 0.03 6.97 ± 0.33 6.80 ± 0.05 6.47 ± 0.23
10 2.26 ± 0.03 5.12 ± 0.03 1.33 ± 0.02 13.88 ± 0.03
11 5.66 ± 0.01 9.47 ± 0.03 3.14 ± 0.01 4.27 ± 0.03
12 7.88 ± 0.01 4.54 ± 0.01 2.25 ± 0.03 3.84 ± 0.01
13 11.15 ± 0.01 6.13 ± 0.02 2.14 ± 0.05 6.32 ± 0.02
14 2.69 ± 0.01 2.26 ± 0.03 2.14 ± 0.04 36.61 ± 0.48
15 6.81 ± 0.03 7.70 ± 0.12 2.77 ± 0.01 13.69 ± 0.01
16 1.72 ± 0.01 3.75 ± 0.02 3.76 ± 0.03 2.81 ± 0.03

a
Data are reported as IC50 values (concentrations of complexes required to inhibit cell viability by 50%) determined by MTT assay after 24 h of
continuous exposure to each compound. The data represent the mean values ± SEM (standard error of mean) of at least three independent
experiments.

8477 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Table 3. Cytotoxic Activity of Compounds L1−L16 in HeLa pharmacological index of these hydrazinyl-thiazolo arene
Cells and Calculated Partition Coefficients ruthenium complexes.
Gene Expression Profiling As a Response to
compd log Pa IC50 (μM)b
Ruthenium Complexes Treatment. In order to identify
L1 3.1 ± 0.6 11.4 ± 0.005 molecular mechanisms induced by treatment with the thiazolo
L2 4.4 ± 0.6 >100 arene ruthenium compounds, we profiled a global mRNA
L3 2.6 ± 0.8 64.87 ± 0.005 expression assay. Two ovarian carcinoma cell lines, A2780 and
L4 4.4 ± 0.6 >100 A2780cisR, treated with two selected compounds (4 and 14)
L5 4.0 ± 0.9 >100 were used for 24 h after treatment, for microarray analysis.
L6 4.6 ± 0.9 >100 Although both compounds presented similarities in their
L7 2.9 ± 0.6 25.59 ± 0.010 structure (Figure 2), our microarray data showed different
L8 4.2 ± 0.6 20.04 ± 0.019 transcriptional patterns of the two cell lines in response to these
L9 3.3 ± 0.6 >100 compounds. The specific genes modulated by ruthenium
L10 4.6 ± 0.6 11.1 ± 0.009 complexes in each cell line were selected using a fold change
L11 3.8 ± 0.9 >100 (Fc) threshold of 1.5 and an FDR-adjusted p-value <0.05. As
L12 2.7 ± 0.9 >100 shown in the Venn diagram (Figure 3), the numbers of genes
L13 3.9 ± 0.6 57.53 ± 0.011
L14 3.4 ± 0.9 >100
L15 5.2 ± 0.6 >100
L16 4.4 ± 0.9 >100
a
The partition coefficients were calculated using ACD/ChemSketch.26
b
Taken from ref 17.

Figure 2. Molecular structures of complexes 4 and 14.


Figure 3. Venn diagram of transcriptional changes induced by
complexes 4 and 14 in the A2780 and A2780cisR ovarian carcinoma
cell lines. The overlap areas indicate the common set of genes for both
by these two complexes on the tumor cell lines (HeLa, A2780, ruthenium compounds in both cell lines. Complexes 4 and 14
and A2780cisR), one may notice selectivity in favor of the modulate a unique set of 169 differentially expressed genes (DEGs) in
noncancerous cell line, namely, a cytotoxic effect upon normal A2780 cells and 3723 common DEGs in A2780cisR cells.
fibroblasts 15 times lower than upon cancerous cells. Moreover,
the activity of the hydrazinyl-thiazole compounds L4 and L14 on regulated by 14 were higher than those modulated by 4, for
HeLa cells (Table 3) is very low (>100 μM), confirming the both ovarian cisplatin-sensitive and cisplatin-resistant cell lines.
beneficial effect of complexation to arene ruthenium units. Compound 4 induced transcriptional changes of 444 genes
Such variations in the activity appear to be dependent on (319 up-regulated genes and 125 down-regulated genes) in
some minor modifications in the molecular structure of the A2780 cells and 4840 genes (2569 up-regulated genes and 2271
complexes (see Table 1), thus revealing some interesting down-regulated genes) in the A2780cisR cell line. Compara-
trends: the presence of electron-withdrawing groups (chloro tively, complex 14 modulated 1392 genes (867 up-regulated
and dichloro) on the phenyl ring of R1 leads to a higher genes, 525 down-regulated genes) in A2780 cells and 7577
antiproliferative effect on the cancerous cell lines (HeLa, genes (3826 up-regulated genes and 3751 down-regulated
A2780, and A2780cisR), and the presence of a more genes) in A2780cisR cells. In addition, we have identified a
hydrophobic substituent (phenyl) at the R2 position of the unique set of 169 genes modulated by both compounds in
thiazole ring also increases the antiproliferative effect, with the A2780 cells and a unique set of 3723 genes in A2780cisR cells
exception of 15 (phenyl) and 13 (methyl) which show a higher (Figure 3).
activity but only on the HeLa cell line. However, the nature of Identification of the Biological Pathway Induced by
the R3 group (H, COMe, and COOEt) appears to have a less Ruthenium Complexes 4 and 14. To investigate the
predictable effect: in complexes 13, 14, and 16 where only R3 molecular mechanisms underlying the effects induced by the
varied, the cytotoxicity of the complexes is comparable, while hydrazinyl-thiazolo arene ruthenium compounds, we searched
between complexes 5 and 6 the COMe derivative is databases for gene ontology (GO) terms and canonical
significantly less cytotoxic than the COOEt analogue. More- pathways. The GO enrichment analysis showed that the
over, when looking at the log P values of L1−L16 in conjunction common set of genes (n = 169) modulated by both ruthenium
with the IC50 values of the ligands on HeLa cells (Table 3), it compounds in the A2780 cell line was significantly enriched in
appears that the lipophilicity of the ligands does not correlate response to stimulus (GO:0050896|GO:0051869, 76 genes, p =
with the activity. Therefore, the functionalization of the ligands 8.85 × 10−07), apoptotic process (GO:0006915|GO:0006917|
should be better explored for further optimization of the GO:0008632, 21 genes, p = 1.02 × 10−06), programmed cell
8478 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

death (GO:0012501|GO:0016244, 21 genes, p = 1.39 × 10−06), of the p53 signaling pathway in inducing apoptosis, in both
response to stress (GO:0006950, 39 genes, p = 5.95 × 10−06), cisplatin-sensitive and cisplatin-resistant ovarian carcinoma cell
and cell death (GO:0008219, 21 genes, p = 1.09 × 10−05). In lines (Table 5). Our data showed that activation of autophagy
Table 4, we present the 21 common genes involved in was triggered by overexpression of DRAM1, AEN, and
apoptotic and cell death-related processes. TP53INP1 in the A2780 cell line (Figure 4). Under conditions
of genotoxic stress induced by different agents, TP53INP1
Table 4. Common Set of Differentially Expressed Genes promotes autophagy-dependent cell death by binding the
Involved in Apoptotic and Cell Death-Related Processes proteins of the ATG8-family, followed by degradation of certain
Modulated by Complexes 4 and 14 on the Ovarian A2780 antiapoptotic proteins, through partial displacement of
Cell Line sequestosome 1 (SQSTM1/p62).27,28 Moreover, the inhibition
of SQSTM1 can be orchestrated by DRAM129 which regulates
fold change autophagy through increasing the lysosomal activity and
ANOVA clearance of autophagosomes.30 In our study, we found high
gene symbol description 4 14 p-value levels of TP53INP1 induced by both complexes 4 (Fc = 3.96
DRAM1 DNA-damage regulated 2.87 2.33 0.039 up) and 14 (Fc = 3.33 up) and an increased expression of
autophagy modulator 1 DRAM1 induced by 4 (Fc = 3.19 up) and 14 (Fc = 2.53 up) in
ZMAT3 zinc finger, matrin-type 3 2.79 2.92 0.038
(ZMAT3) the A2780 ovarian cell line. The p53-dependent autophagy, as
TP53INP1 tumor protein p53 inducible 2.72 2.66 0.007 an effect of treatment, was also enhanced by overexpression of
nuclear protein 1 AEN, which is involved in the growth of autophagic vacuoles
RPS27L ribosomal protein S27-like 2.15 2.25 0.006 and LC3-II after genotoxic stress.31 The role of TP53INP1,
TP53I3 tumor protein p53 inducible 2.10 1.66 0.014 DRAM1, and AEN in ovarian cancer was little studied.
protein 3 Nevertheless, in a recent study,32 it was demonstrated that
PRMT2 protein arginine 2.07 1.97 0.040 downregulation of TP53INP1 expression by miR569 provided
methyltransferase 2
to ovarian cancer cells the ability to survive and develop
SHF Src homology 2 domain 1.94 2.19 0.033
containing F metastases, while by overexpression of TP53INP1 using anti-
ID1 inhibitor of DNA binding 1, 1.94 2.69 0.019 miR569 therapy, an activation of cell death was obtained.
dominant negative helix− Moreover, it was demonstrated that the combination of anti-
loop−helix protein miR569 and cisplatin therapy significantly reduces tumor
BAX BCL2-associated X protein 1.85 1.66 0.018
weights as compared to treatment with cisplatin alone,
FAS Fas (TNF receptor superfamily, 1.83 1.65 0.006
member 6) emphasizing the role of TP53INP1 in the management of
AEN apoptosis enhancing nuclease 1.78 1.81 0.013 ovarian tumor treatment. On the basis of these findings, the
C16orf5 chromosome 16 open reading 1.72 1.66 0.014 combination of cisplatin with ruthenium compounds could be
frame 5 considered as a valuable alternative for the treatment of ovarian
HTRA2 HtrA serine peptidase 2 1.65 1.65 0.033 cancer.
TRIAP1 TP53 regulated inhibitor of 1.64 1.63 0.008 To the best of our knowledge, there is no evidence regarding
apoptosis 1 the role of DRAM 1 in ovarian cancer, but AEN has been
SERPINB9 serpin peptidase inhibitor, 1.59 2.32 0.021 linked to the activation of UCHL1, an important regulator of
clade B (ovalbumin),
member 9 cisplatin resistance in ovarian cancer. Downregulation of tumor
RHOC Ras homologue gene family, 1.56 1.65 0.019 suppressor gene UCHL1 was associated with increased cisplatin
member C resistance, and there is the assumption that an increased level of
APH1B anterior pharynx defective 1 1.54 1.59 0.024 UCHL1 mediated pathways is considered a new promising
homologue B (C. elegans)
strategy to overcome cisplatin resistance in ovarian cancer.33
SLIT2 slit homologue 2 (Drosophila) −1.51 −1.88 0.017
Moreover, UCHL1 and HTRA2 are considered two key
BNIP3L BCL2/adenovirus E1B 19 kDa −1.62 −1.57 0.048
interacting protein 3-like components of inducing of apoptosis by proteolysis, in a
HK2 hexokinase 2 −1.91 −2.23 0.016 caspase-independent way mediated by TNf-α.34 Our microarray
CXCR4 chemokine (C-X-C motif) −4.21 −3.86 0.044 data revealed up-regulation of UCHL1 expression induced by
receptor 4 (CXCR4) both 4 (Fc = 2.00 up) and 14 (Fc = 2.76 up) in the A2780
ovarian cell line and more than 1.5-fold for complexes 4 and 14
The GO enrichment analysis conducted on the A2780cisR (microarray data).
cell line showed an over-representation of 85 GO terms in the TP53-dependent apoptosis was also modulated by the
common set of genes modulated by both 4 and 14. The overexpression of FAS, BAX, ZMAT3, TRIAP1, and TP53I3
majority of the GO terms were related to cellular processes and molecules in A2780 cells. In a recent paper,35 it was
component organization, metabolic processes, apoptosis, and demonstrated that overexpression of FAS could avoid the
cell death (Table S4). Furthermore, we focused our attention development of resistance to cisplatin, and thus, FAS could be a
on the genes involved in apoptosis and cell death. Our data promising therapeutic target to restore the sensitivity to
highlighted 180 differentially expressed genes with different cisplatin in ovarian cancer cells. In sensitive ovarian cell lines,
roles in apoptosis and cell death that were taken into we observed a 2.42-fold overexpression of FAS induced by 4
consideration for further analysis (Table S5). and a 1.92-fold overexpression of FAS induced by 14.
In order to better understand the mechanisms by which BAX was previously identified as an important target
complexes 4 and 14 modulate apoptosis in ovarian cell lines, we modulated by ruthenium compounds in the treatment of
performed a functional analysis on the sets of genes, modulated patients with acquired cisplatin resistance.36 Our data showed
by these compounds, and known to be involved in apoptosis. an increased expression of BAX, emphasizing its role as an
Using ingenuity pathway analysis (IPA), we highlighted the role activator of apoptosis by ruthenium compounds. We also
8479 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Table 5. Top Five Canonical Pathways Identified in Gene Sets Related to Apoptotic Processes by Ingenuity Pathway Analysis
(IPA)
canonical pathways p-value ratio genes
A2780 cell line
p53 signaling 9.48 × 10−08 5/98 BAX, DRAM1,FAS, TP53I3, TP53INP1
(0.051)
−07
induction of apoptosis by HIV1 6.76 × 10 4/59 BAX, CRC4, FAS, HTRA2
(0.068)
tumoricidal function of hepatic 3.00 × 10−06 3/24 BAX, FAS, SERPINB9
natural killer cells (0.125)
apoptosis signaling 1.55 × 10−04 3/88 BAX, FAS, HTRA2
(0.034)
molecular mechanisms of cancer 7.76 × 10−04 4/356 APH1b, BAX, FAS, RHOC
(0.011)
A2780cisR Cell Line
protein ubiquitination pathway 4.68 × 10−12 20/252 HSPA5, HSPB8, PSMA1, PSMB1, PSMB4, PSMC1, PSMC3, PSMC5, PSMC6, PSMD3, PSMD4,
(0.079) PSMD6, PSMD11, PSMD12, PSMD13, PSME1, PSME2, SACS, UBA1, UBE2D3
p53 signaling 7.26 × 10−10 12/98 BBC3, BIRC5, E2F1, GADD45A, HIPK2, JMY, JUN, MDM4, PERP, PMAIP1, SIRT1, TP73
(0.122)
molecular mechanisms of cancer 8.86 × 10−06 15/356 ARHGEF2, ARHGEF9, ARHGEF16, ARHGEF17, BBC3, CDKN2C, E2F1, HIPK2, JAK2, JUN,
(0.042) MAX, PMAIP1, RHOB, SMAD6, TGFB3
cell cycle: G2/M DNA damage 1.91 × 10−04 5/49 CDK1, GADD45A, HIPK2, MDM4, TOP2A
checkpoint regulation (0.102)
ATM signaling 4.59 × 10−04 5/59 CDK1, GADD45A, JUN, MDM4, TP73
(0.085)

Figure 4. Activation of apoptotic and autophagy mechanisms induced by 4 and 14 in the A2780 cell line. Apoptosis is modulated by overexpression
of genes involved in p53 pathways such as FAS, BAX, HTRA2, TRIAP1, TP53INP1, CDIP1, or TP53I3, while autophagy is activated by
overexpression of DRAM1 and TP53INP1. The red color means overexpression of the mRNAs of specific genes in A2780 cells treated with 4 and 14
compared to that of untreated A2780 cells. Red intensities are proportional to mRNAs transcripts.

8480 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Figure 5. Activation of apoptosis by 4 and 14 in the A2780cisR cell line. Both complexes induce apoptosis in A2780cisR cells by activation of
TNFRSF8 and TNFRSF10D death receptors, and by the transcription factors NURP1, FOXO3, AATF, JUN, FOSL2, and SIAH2. Molecules
involved in resistance mechanisms of platinum-based compounds such as ERCC2, HMGB1, and CLSPN are not activated by 4 and 14. The intensity
of the red color is proportional to the overexpression of the mRNAs induced by 4 and 14 in A2780cisR cells compared with those of untreated
A2780cisR cells, while the green color means a loss of the activity or a downregulation of gene expression in A2780cisR cells after treatment with 4
or 14 as compared to that of untreated cells.

noticed a 2.5-fold up-regulation of ZMAT3 induced by both complexes. RPS27L represents a target of the p53 wild-type
complexes 4 and 14. ZMAT3 is involved in activation of gene, which was previously reported to be a promoter of
apoptosis, either directly through the regulation of BAX, p21, apoptosis in genotoxic stress conditions under etoposide
and FAS expression or indirectly by regulating the cell cycle treatment.43
through cyclin D1 and 14−3−3σ.37,38 Moreover, ZMAT3 The modulation of apoptosis could also be dependent on a
through its double-stranded-RNA-binding zinc finger protein negative regulation of some important antiapoptotic genes by
can bind different types of dsRNAs, increasing the effects on blocking their transcriptional factors as NF-Kb.44 Along this
cell cycle regulation and apoptosis, mediated by silencing line, PRMT2, one of the NF-kB modulators, blocks trans-
RNA.39,40 To date, no study reported ZMAT3 activation by location of NF-kB in the nucleus and therefore its antiapoptotic
ruthenium compounds or their role in cisplatin-induced target genes;45 however, there are no data related to the role of
apoptosis in ovarian cancer. PRMT2 in the modulation of apoptosis in ovarian cancer
Genotoxic agents as these ruthenium compounds could also induced by ruthenium compounds.
activate apoptosis through oxidative stress by enzymatic All of the above taken together revealed new molecules and
activity. One of the main actors involved in the control of pathways involved in the cell death of A2780 ovarian cancer
apoptosis in the generation of stress oxidative in a p53- cells by the hydrazinyl-thiazolo arene ruthenium complexes.
dependent manner is TP53I3.41 High levels of TP53I3 were Then, we focused our attention on the mechanism and
found in stress conditions during p53-mediated growth arrest, pathways modulated by complexes 4 and 14 in the cisplatin-
followed by exposure to genotoxic agents.42 Our data confirm resistant A2780cisR ovarian cancer cells (Figure 5).
the implication of TP53I3 in TP53-mediated apoptosis under Among the genes in the network generated by A2780cisR
genotoxic stress conditions, almost a 2 up Fc regulation was cells, NURP1 and TRIB3 were indicated as upstream regulators
reported for both complexes 4 and 14 (Figure 4). Likewise, we by IPA analysis. NURP1 is a transcription regulator predicted
found a 2.8 Fc expression of RPS27L gene, induced by both to be “activated” by ruthenium compounds and significantly
8481 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Table 6. Upstream Regulators Activated by 4 and 14 in the A2780cisR Cell Line


upstream fold predicted activation p-value of
regulator change molecule type state z-score overlap target molecules in data set
TRIB3 6.253 kinase inhibited −2.236 2.94 × 10−06 DDIT4, GARS, HERPUD1, PMAIP1, TRIB3
NURP1 3.829 transcription activated 3.464 1.63 × 10−03 BUB1, BUB1B, CEBPB, CYR61, DHCR24, ESPL1,
regulator FOXO3, TRIB3

Figure 6. qRT-PCR validation of microarray data in the A2780 cell line. Fold change (Fc) values were calculated by the ΔΔCt method relative to the
CTR group (untreated cells). The p-values were assessed by ANOVA, and a Tukey’s test was used for posthoc pairwise comparisons (* p < 0.05, **
p < 0.01, *** p < 0.001).

affect the expression of their target genes such as CYR61, apoptosis induced by ruthenium compounds. Nevertheless, low
FOXO3, and TRIB3. The relationship between upstream levels of GADD45A and BBC3/PUMA were previously
regulators and their target genes is presented in Table 6. associated with cisplatin resistance in ovarian cancer.52
We noticed different transcriptional patterns for A2780cisR Considering this issue, by the growth of its expression, BBC3
compared with those of the A2780 ovarian cell line in response was already proposed as a chemosensitizer in ovarian cancer
to 4 and 14. One hundred eighty differentially expressed genes therapy based on platinum compounds.53 Our qRT-PCR data
with a different role in apoptosis and cell death were commonly confirm the role of BBC3 as a sensitizer for cisplatin resistance,
modulated by 4 and 14 in the A2780cisR ovarian cell line induced by ruthenium compounds, with 2.78 and 3.38 Fc up-
(Figure 3). Our data indicated that p53 signaling was, as for the regulation for complexes 4 and 14.
cisplatin-sensitive cell line, one of the most important pathways Going deeper with functional analysis, we focused our
activated by the hydrazinyl-thiazolo arene ruthenium complexes attention on five transcriptional factors including JUN, AATF,
in the A2780cisR cell line (Table 5). However, unlike the SIAH2, NUPR1, and FOXO3 that were positively correlated
A2780 cell line, a different set of pro-apoptotic molecules such with activation of apoptosis. FOXO3 and JUN were both
as BBC3,46 BCL2L13,47 HRK,48 AIFM3,49 PMAIP1,50 and associated with BBC3/PUMA activation, their dual expression
GADD45A,51 was activated in the A2780ciR cell line (Figure being considered necessary to overcome chemo-resistance in
5). An overexpression greater than 2.5 Fc was recorded for ovarian cancer cells.54 Moreover, our data revealed that JUN-
HRK, AIFM3, PMAIP1, and GADD45A, while for BBC3 and mediated pathways have involved supplementary molecules and
BCL2L13 a minimum of 1.78 Fc up-regulation was recorded death modulators. In brief, JUN activated by the NURP1
for both complexes. To our knowledge, there are no studies to upstream regulator and FOXO3 via CYR6 led to the activation
present data related to these molecules as modulators of of two supplementary death modulators, DEDD2 and PDCD4,
8482 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Figure 7. qRT-PCR validation of microarray data in the A2780cisR cell line. Fold change (Fc) values were calculated by the ΔΔCt method relative
to the CTR group (untreated cells). The p-values were assessed by ANOVA, and a Tukey’s test was used for posthoc pairwise comparisons (* p <
0.05, ** p < 0.01, *** p < 0.001).

respectively. Recently, it was demonstrated that low levels of range of chemotherapeutic agents. Although, initially about
PDCD4 were associated with cisplatin resistance in ovarian 85% of ovarian cancers respond very well to first-line
cancer cells, and by increasing PDCD4 expression by miRNA chemotherapy based on platinum compounds and taxanes,
modulation, sensitivity to cisplatin was restored.55 With 1.66 Fc less than 15% will remain in complete remission. Unfortunately,
expression for 4 and 1.90 Fc expression for 14, our microarray the majority of patients with ovarian cancers will develop
data confirmed the activation of PDCD4 by ruthenium progressive resistance. Depending on the time of relapse, three
compounds. In addition, under cellular stress, AATF triggers kind of platinum resistance mechanisms have been recorded:
and increases the expression of JUN-mediated apoptosis.56 We increased resistance if the relapse occurs within 6 months after
found that SIAH2 activates the proteasome protein ubiquitin the first-line treatment; partial resistance if the relapse is
pathway by PSMD4 and UBE2D3 molecules; however, there recorded between 6 and 12 months after completing the initial
are still no studies that evaluated the activation of these treatment; and theoretically no resistance if relapse occurs after
molecules in relation to apoptosis induced by platinum or one year of finishing the first line therapy. Nevertheless,
ruthenium metal-based drugs. Interestingly, we noticed that cisplatin based retreatment in the second line of patients with
molecules involved in DNA repair, associated with cisplatin no platinum resistance will be effective only for about 60% of
resistance, such as ERCC2, HMGB1, CLSPN, or E2F, were not the patients, while for partial platinum resistance only a quarter
activated following the treatment with 4 or 14, which also of the patients will respond to a second platinum-based
highlights the advantage of these classes of compounds for therapy.57 Generally, the classical treatment based on platinum
treatment of ovarian cancer cells resistant to cisplatin. compounds had a modest impact on overall survival (OS) in
Microarray Data Validation. In order to assess the the last two decades.58,59 Because treatments with platinum
reliability of the microarray data, 21 common genes modulated compounds have reached a threshold of efficacy, it is necessary
by both ruthenium complexes were considered for qRT-PCR to identify new chemical compounds with less cross-resistance.
validation. The selected genes were chosen according to their Processes of Cell Death. As previously discussed, the
known functions in apoptosis and autophagy related to p53 biological pathway induced by the hydrazinyl-thiazolo arene
pathways. For the A2780 cell line, PCR data showed a good ruthenium complexes 4 and 14 suggests apoptosis as the main
correlation with the microarray data, except for the BNIP3L cell death mechanism. Therefore, all cancerous cell lines (HeLa,
gene (Figure 6). This result could be explained because BNIP3l A2780, and A2780cisR) were treated with complexes 4 and 14
had a p-value close to the threshold (0.048) in the microarray at the corresponding IC50 concentrations and with staurospor-
experiment (Figure 6). For the A2780cisR cell line, all of the ine (2 μg/mL) as a positive control (see Experimental Section).
selected genes showed the same expression pattern as in the The percentage of apoptotic cells (cells with condensed and/or
microarray experiment and were validated by qRT-PCR (Figure fragmented nuclei) was determined by Hoechst staining (see
7). Figure S1). For both compounds and on all cancer cell lines, at
One of the major problems reported for ovarian cancers is least 60% of the nuclei showed chromatin condensation and
related to the development of a multidrug resistance to a broad fragmentation, two typical apoptotic features.60 The morphol-
8483 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Figure 8. Morphology of the nuclei of HeLa cells observed by fluorescence microscopy (Hoechst staining, 24 h incubation at IC50 concentrations)
after treatment with or without (control) complexes 4 and 14, and with staurosporine (2 μg/mL).

Figure 9. Disruption of mitochondrial membrane potential by TMRE staining upon addition of complexes 4, 14, and staurosporine to the cancerous
cells A2780 and A2780cisR.

Figure 10. Combination of Hoechst (blue fluorescence) and TMRE (red fluorescence) staining experiments, showing the effect of complexes 4, 14,
and staurosporine on HeLa cells (24 h incubations at IC50 concentrations).

ogy of the nuclei of HeLa cells after 24 h of incubation with or Finally, using an inverted fluorescence microscopy, the
without (control) complexes 4 and 14 and with staurosporine implication of autophagy was also investigated on the cancerous
is presented in Figure 8. cell lines HeLa, A2780, and A2780cisR. Autophagy was
In addition, the effect of complexes 4 and 14 on the evaluated using a protocol based on monodansylcadaverine
mitochondrial membrane potential was assessed using (MDC) staining, and tamoxifen (10 μM) was used as a positive
control. Like tamoxifen, both complexes (24 h of incubation
tetramethylrhodamine ethyl ester (TMRE) staining. As can
with 4 and 14 at IC50 concentrations) clearly increase the
be appreciated in Figure 9, undisrupted mitochondrial fluorescence intensity of the staining agent as compared to that
membranes are observed for control cells (intense fluorescence of untreated cells (control), confirming that autophagy is also
staining), while upon addition of complexes 4 and 14 to A2780 involved in the cell death of HeLa, A2780, and A2780cisR
and A2780cisR cells, mitochondrial membranes are altered cancerous cells (Figure 11).


(weak or no fluorescence staining).
Moreover, by combining the Hoechst and TMRE staining CONCLUSIONS
procedures, morphological modifications of the nucleus and
In this study, we have reported the synthesis, the in vitro
disruption of the mitochondrial membranes on HeLa cells can antiproliferative activity, and the molecular mechanisms
be visualized (Figure 10). Indeed, the combination of both underlying cell death in cancer cells from a new class of
staining agents emphasized cell death and confirmed the role of hydrazinyl-thiazolo arene ruthenium complexes. Our data bring
apoptosis in sustaining tumor cell death, which further support a better understanding of the molecular mechanisms underlying
the gene expression data. biological and pharmacological profiles induced by arene
8484 DOI: 10.1021/acs.jmedchem.5b00855
J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

(50 mL), filtered, and washed multiple times with n-hexane, and the
solid was dried under vacuum. The yields are listed in Table S1.
1: [(η6-p-Cymene)Ru(L1)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
1.07 (d, 3JH−H = 7.1 Hz, 3H), 1.15 (d, 3JH−H = 7.1 Hz, 3H), 2.26 (s,
3H), 2.46 (s, 3H), 2.57 (sept, 3JH−H = 7.1 Hz, 1H), 4.52 (d, 3JH−H =
6.0 Hz, 1H), 5.06 (d, 3JH−H = 6.0 Hz, 1H), 5.18 (d, 3JH−H = 6.0 Hz,
1H), 5.44 (d, 3JH−H = 6.0 Hz, 1H), 6.49 (s, 1H), 7,55−7.57 (m, 3H),
8.12 (m, 2H), 9.32 (s, 1H), 15.63 (s, 1H) ppm. ESI-MS m/z (+):
488.05 M+. Anal. Calcd for C21H25N3SCl2Ru: C, 48.18; H, 4.81; N,
8.03. Found: C, 48.18; H, 4.83; N, 7.96. IR (KBr): 2958 (m), 2919
Figure 11. Fluorescence intensity corresponding to autophagy upon (m), 2525 (s), 1557 (s), 1505 (s), 1387 (m), 1306 (s), 1257 (m),
addition of complexes 4, 14 (24 h incubation at IC50 concentrations), 1093 (m) cm−1.
and tamoxifen (10 μM) to cancer cells (control = cells treated with 2: [(η6-p-Cymene)Ru(L2)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
only the staining agent). 0.96 (d, 3JH−H = 6.8 Hz, 3H), 1.01 (d, 3JH−H = 6.8 Hz, 3H), 2.14 (s,
3H), 2.36 (sept, 3JH−H = 6.8 Hz, 1H), 4.03 (d, 3JH−H = 5.4 Hz, 1H),
4.25 (d, 3JH−H = 5.4 Hz, 1H), 4.65−4.66 (m, 2H), 6.74 (s, 1H), 7.54−
7.56 (m, 6H), 7.90−7.91 (m, 2H), 8.12−8.13 (m, 2H), 9.30 (s, 1H),
ruthenium compounds and highlights their role in treating and 15.88 (s, 1H) ppm. ESI-MS m/z (+): 550.06 M+. Anal. Calcd for
overcoming cisplatin resistance mechanisms in ovarian cancers. C26H27N3SCl2Ru·0.5 H2O: C, 52.52; H, 4.75; N, 7.07. Found: C,
We found that the p53 signaling pathway was one of the 52.63; H, 4.55; N, 6.90. IR (KBr): 2916 (m), 2501 (m), 1575 (s),
most important activated pathways in both cisplatin-sensitive 1500 (s), 1384 (m), 1313 (m), 1090 (s) cm−1.
and cisplatin-resistant cancer cell lines, despite the fact that 3: [(η6-p-Cymene)Ru(L3)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
1.08 (d, 3JH−H = 6.9 Hz, 3H), 1.16 (d, 3JH−H = 6.9 Hz, 3H), 2.28 (s,
signal transduction occurs through different molecules in these
3H), 2.45 (s, 3H), 2.56 (sept, 3JH−H = 6.9 Hz, 1H), 2.82 (s, 3H), 4.51
cell lines. Moreover, new activated molecules such as FAS, (d, 3JH−H = 5.8 Hz, 1H), 5.09 (d, 3JH−H = 5.8 Hz, 1H), 5.22 (d, 3JH−H =
ZMAT3, PRMT2, BBC3/PUMA, GADD45A, and PDCD4 5.8 Hz, 1H), 5.41 (d, 3JH−H = 5.8 Hz, 1H), 7.58−7.60 (m, 3H), 8.14
whose overexpression is associated with overcoming resistance (d, 3JH−H = 7.1 Hz, 2H), 9.34 (s, 1H), 12.95 (s, 1H) ppm. ESI-MS m/z
to cisplatin therapy in ovarian cancers were identified. (+): 529.9 M+. Anal. Calcd for C23H27N3OSCl2Ru: C, 48.85; H, 4.81;
Consequently, these results suggest that our hydrazinyl-thiazolo N, 7.43. Found: C, 48.96; H, 5.34; N, 6.87. IR (KBr): 2965 (w), 1637
arene ruthenium complexes could be used in association with (m), 1568 (s), 1475 (s), 1374 (m), 1309 (s) cm−1.
cisplatin to prevent the development of cisplatin resistance 4: [(η6-p-Cymene)Ru(L4)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
mechanisms, thus inducing cell death in ovarian cancers under a 1.08 (d, 3JH−H = 6.8 Hz, 3H), 1.16 (d, 3JH−H = 6.8 Hz, 3H), 1.35 (t,
3
synergetic effect. This study describes for the first time on JH−H = 7.2 Hz, 3H), 2.20 (s, 3H), 2.60 (sept, 3JH−H = 6.8 Hz, 1H),
3.80 (s, 2H), 4.29 (q, 3JH−H = 7.2 Hz, 2H), 4.37 (d, 3JH−H = 5.9 Hz,
ovarian cancer cell lines the pathways and molecular
1H), 5.19 (d, 3JH−H = 5.9 Hz, 1H), 5.24 (d, 3JH−H = 5.9 Hz, 1H), 5.62
mechanisms modulated by hydrazinyl-thiazolo arene ruthenium (d, 3JH−H = 5.9 Hz, 1H), 6.83 (s, 1H), 7.47 (dd, 3JH−H = 8.4 Hz, 4JH−H
complexes.


= 1.9 Hz, 1H), 7.58 (d, 4JH−H = 1.9 Hz, 1H), 8.42 (d, 3JH−H = 8.4 Hz,
1H), 9.24 (s, 1H), 16.08 (s, 1H) ppm. ESI-MS m/z (+): 630.0 M+.
EXPERIMENTAL SECTION Anal. Calcd for C24H27N3O2SCl4Ru·H2O: C, 42.24; H, 4.28; N, 6.16.
The starting material (η6-p-cymene)2Ru2Cl461 and the hydrazinyl- Found: C, 42.43; H, 4.08; N, 6.31. IR (KBr): 2918 (m), 1732 (m),
thiazole derivatives (L1−L16)17 were prepared according to the 1589 (s), 1551 (s), 1470 (s), 1384 (s), 1259 (m), 1146 (m), 1083
literature. All other reagents were commercially available (Sigma- (m), 1056 (m) cm−1.
Aldrich, TCI Europe) and were used without further purification. The 5: [(η6-p-Cymene)Ru(L5)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
1
H NMR spectra were recorded on a Bruker Avance II 400 1.07 (d, 3JH−H = 7.1 Hz, 3H), 1.13 (d, 3JH−H = 7.1 Hz, 3H), 2.49 (s,
spectrometer, using the residual protonated solvent as an internal 3H), 2.54 (sept, 3JH−H = 7.1 Hz, 1H), 2.63 (s, 3H), 2.79 (s, 3H), 5.08
standard. Infrared spectra were recorded as KBr pellets on a (d, 3JH−H = 5.4 Hz, 1H), 5.35 (d, 3JH−H = 5.4 Hz, 1H), 5.57 (d, 3JH−H =
PerkinElmer FTIR 1720 X spectrometer. Elemental analyses were 5.4 Hz, 1H), 5.65 (d, 3JH−H = 5.4 Hz, 1H), 7.31 (d, 3JH−H = 8.6 Hz,
performed by the Mikroelementarisches Laboratorium, ETH Zürich 1H), 7.42 (s, 1H), 7.98 (d, 3JH−H = 8.6 Hz, 1H), 8.29 (s, 1H), 12.83 (s,
(Zürich, Switzerland). Elemental analyses confirm ≥95% purity for all 1H) ppm. ESI-MS m/z (+): 599.8 M + . Anal. Calcd for
of the tested compounds. Electrospray ionization mass spectra were C23H25N3OSCl4Ru·H2O: C, 42.34; H, 4.17; N, 6.44. Found: C,
recorded in positive-ion mode with a Bruker FTMS 4.7T BioAPEX II 42.66; H, 3.96; N, 6.30. IR (KBr): 2975 (m), 1604 (m), 1564 (m),
mass spectrometer (University of Fribourg, Switzerland). Microwave 1384 (s), 1328 (s), 1055 (s) cm−1.
assisted syntheses were performed in sealed vessels using a CEM 6: [(η6-p-Cymene)Ru(L6)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
Discover LabMate instrument, ensured with online inside temperature 1.07 (d, 3JH−H = 6.2 Hz, 3H), 1.15 (d, 3JH−H = 6.2 Hz, 3H), 1.33 (t,
3
and pressure controls. JH−H = 6.6 Hz, 3H), 2.21 (s, 3H), 2.62 (sept, 3JH−H = 6.2 Hz, 1H),
Synthesis and Characterization of Compounds 1−16. (a) A 2.78 (s, 3H), 4.27 (q, 3JH−H = 6.6 Hz, 2H), 5.12 (d, 3JH−H = 5.5 Hz,
mixture of (η6-p-cymene)2Ru2Cl4 (100 mg; 0.163 mmol) and 1H), 5.18 (d, 3JH−H = 5.5 Hz, 1H), 5.41 (d, 3JH−H = 5.5 Hz, 1H), 5.59
hydrazinyl-thiazole (0.327 mmol) in methanol (50 mL) was stirred (d, 3JH−H = 5.5 Hz, 1H), 7.44 (d, 3JH−H = 8.2 Hz, 1H), 7.56 (s, 1H),
at room temperature for 10 h. The solvent was then completely 8.55 (d, 3JH−H = 8.2 Hz, 1H), 8.78 (s, 1H), 13.43 (s, 1H) ppm. ESI-
removed under vacuum, and the residue was dissolved in dichloro- MS m/z (+): 630.0 M+. Anal. Calcd for C24H27N3O2SCl4Ru: C, 43.39;
methane (5 mL). The product was precipitated by pouring this H, 4.10; N, 6.32. Found: C, 43.47; H, 4.24; N, 6.26. IR (KBr): 2965
solution into 200 mL of n-hexane, filtering, washing multiple times (w), 1709 (m), 1585 (s), 1474 (s), 1373 (s), 1317 (s), 1276 (s), 1099
with n-hexane, and finally drying under vacuum to afford the (s) cm−1.
corresponding salts in good yields (Table S1). 7: [(η6-p-Cymene)Ru(L7)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
(b) A reaction mixture containing (η6-p-cymene)2Ru2Cl4 (100 mg; 1.07 (d, 3JH−H = 6.9 Hz, 3H), 1.14 (d, 3JH−H = 6.9 Hz, 3H), 2.27 (s,
0.163 mmol) and the hydrazinyl-thiazole (0.327 mmol) in dichloro- 3H), 2.46 (s, 3H), 2.54 (sept, 3JH−H = 6.9 Hz, 1H), 4.81 (d, 3JH−H =
methane (10 mL) was introduced into a quartz reaction vessel, which 5.7 Hz, 1H), 5.09 (d, 3JH−H = 5.7 Hz, 1H), 5.18 (d, 3JH−H = 5.7 Hz,
was sealed and then subjected to microwave irradiation for 30 min at 1H), 5.44 (d, 3JH−H = 5.7 Hz, 1H), 6.48 (s, 1H), 7.12 (d, 3JH−H = 8.4
an internal temperature of 60 °C. The mixture was then concentrated Hz, 2H), 7.70 (d, 3JH−H = 8.4 Hz, 2H), 8.76 (s, 1H), 14.23 (s, 1H)
under vacuum (2 mL), and the residue was precipitated with n-hexane ppm. ESI-MS m/z (+): 504.05 M + . Anal. Calcd for

8485 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

C21H25N3OSCl2Ru·0.5 H2O: C, 45.99; H, 4.78; N, 7.66. Found: C, Anal. Calcd for C26H26N3SCl3Ru·1.5 H2O: C, 48.27; H, 4.52; N, 6.49.
45.90; H, 4.98; N, 7.12. IR (KBr): 2965 (m), 1607 (s), 1552 (m), Found: C, 48.23; H, 4.01; N, 6.32. IR (KBr): 2969 (w), 2478 (m),
1509 (s), 1383 (m), 1284 (m), 1254 (m), 1174 (m) cm−1. 1578 (s), 1499 (m), 1379 (w), 1325 (w), 1257 (w), 1088 (m) cm−1.
8: [(η6-p-Cymene)Ru(L8)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = 15: [(η6-p-Cymene)Ru(L15)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
0.96 (d, 3JH−H = 6.3 Hz, 3H), 1.01 (d, 3JH−H = 6.3 Hz, 3H), 2.16 (s, 1.11 (d, 3JH−H = 6.8 Hz, 3H), 1.28 (d, 3JH−H = 6.8 Hz, 3H), 2.16 (s,
3H), 2.32 (sept, 3JH−H = 6.3 Hz, 1H), 3.95 (d, 3JH−H = 5.7 Hz, 1H), 3H), 2.37 (s, 3H), 2.80 (s, 3H), 2.92 (sept, 3JH−H = 6.8 Hz, 1H), 4.50
4.60 (d, 3JH−H = 5.7 Hz, 1H), 4.71−4.73 (m, 2H), 6.73 (s, 1H), 7.15− (d, 3JH−H = 5.9 Hz, 1H), 5.03 (d, 3JH−H = 5.9 Hz, 1H), 5.18 (d, 3JH−H =
7.18 (m, 2H), 7.54−7.55 (m, 3H), 7.75−7.77 (m, 2H), 7.90−7.92 (m, 5.9 Hz, 1H), 5.48 (d, 3JH−H = 5.9 Hz, 1H), 7.37 (m, 2H), 7.52 (d,
3
2H), 8.77 (s, 1H), 14.25 (s, 1H) ppm. ESI-MS m/z (+): 565.9 M+. JH−H = 7.4 Hz, 1H), 7.70 (s, 1H), 8.34 (s, 1H), 12.69 (s, 1H) ppm.
Anal. Calcd for C26H27N3OSCl2Ru·H2O: C, 50.40; H, 4.72; N, 6.78. ESI-MS m/z (+): 564.0 M+. Anal. Calcd for C23H26N3OSCl3Ru: C,
Found: C, 50.17; H, 4.61; N, 6.32. IR (KBr): 2969 (s), 1605 (s), 1572 46.05; H, 4.37; N, 7.00. Found: C, 45.89; H, 4.32; N, 6.98. IR (KBr):
(s), 1508 (s), 1278 (m), 1174 (m) cm−1. 2963 (w), 1638 (m), 1560 (s), 1475 (m), 1374 (m), 1308 (s), 1079
9: [(η6-p-Cymene)Ru(L9)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = (w) cm−1.
1.13 (d, 3JH−H = 6.8 Hz, 3H), 1.20 (d, 3JH−H = 6.8 Hz, 3H), 2.33 (s, 16: [(η6-p-Cymene)Ru(L16)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
3H), 2.50 (s, 3H), 2.63 (sept, 3JH−H = 6.8 Hz, 1H), 3.96 (s, 3H), 4.83 1.08 (d, 3JH−H = 6.2 Hz, 3H), 1.17 (d, 3JH−H = 6.2 Hz, 3H), 1.34 (t,
3
(d, 3JH−H = 5.8 Hz, 1H), 5.15 (d, 3JH−H = 5.8 Hz, 1H), 5.26 (d, 3JH−H = JH−H = 7.2 Hz, 3H), 2.32 (s, 3H), 2.60 (sept, 3JH−H = 6.2 Hz, 1H)
5.8 Hz, 1H), 5.50 (d, 3JH−H = 5.8 Hz, 1H), 6.51 (s, 1H), 7.10 (d, 3JH−H 2.82 (s, 3H), 4.31 (q, 3JH−H = 7.2 Hz, 2H), 4.61 (d, 3JH−H = 5.3 Hz,
= 8.8 Hz, 2H), 8.18 (d, 3JH−H = 8.8 Hz, 2H), 9.25 (s, 1H), 15.36 (s, 1H), 5.15 (d, 3JH−H = 5.3 Hz, 1H), 5.24 (d, 3JH−H = 5.3 Hz, 1H), 5.47
1H) ppm. ESI-MS m/z (+): 517.9 M + . Anal. Calcd for (d, 3JH−H = 5.3 Hz, 1H), 7.56 (m, 2H), 7.93 (m, 1H), 8.46 (s, 1H),
C22H27N3OSCl2Ru: C, 47.74; H, 4.92; N, 7.59. Found: C, 47.52; H, 9.32 (s, 1H), 12.68 (s, 1H) ppm. ESI-MS m/z (+): 593.9 M+. Anal.
4.94; N, 7.40. IR (KBr): 2966 (w), 2481 (w), 1604 (s), 1554 (m), Calcd for C24H28N3O2SCl3Ru·1.5 H2O: C, 43.88; H, 4.76; N, 6.40.
1509 (s), 1378 (w), 1306 (m), 1266 (s), 1180 (m), 1087 (w) cm−1. Found: C, 43.76; H, 4.46; N, 5.88. IR (KBr): 2965 (w), 1711 (m),
10: [(η6-p-Cymene)Ru(L10)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = 1574 (s), 1475 (s), 1372 (m), 1316 (s), 1276 (s), 1100 (s) cm−1.
0.98 (d, 3JH−H = 6.8 Hz, 3H), 1.02 (d, 3JH−H = 6.8 Hz, 3H), 2.17 (s, X-ray Crystallography. A crystal of 12 was mounted on a Stoe
3H), 2.38 (sept, 3JH−H = 6.8 Hz, 1H), 4.00 (d, 3JH−H = 6.1 Hz, 1H), Image Plate Diffraction system equipped with a Φ circle goniometer,
4.53 (d, 3JH−H = 6.1 Hz, 1H), 4.70 (d, 3JH−H = 6.1 Hz, 1H), 4.74 (d, using Mo Kα graphite monochromated radiation (λ = 0.71073 Å) with
3
JH−H = 6.1 Hz, 1H), 6.72 (s, 1H), 7.06 (d, 3JH−H = 8.7 Hz, 2H), 7.55 Φ range 0−200°. Crystal data for 12: Monoclinic space group P21/c
(m, 3H), 7.91 (m, 2H), 8.15 (d, 3JH−H = 8.7 Hz, 2H), 9.20 (s, 1H), (No. 14), cell parameters a = 16.2513(19), b = 15.1904(12), and c =
15.67 (s, 1H) ppm. ESI-MS m/z (+): 580.1 M+. Anal. Calcd for 13.3986(13) Å, β = 107.817(8), V = 3149.0(5) Å3, T = 173(2) K, Z =
C27H29N3OSCl2Ru·H2O: C, 51.18; H, 4.93; N, 6.63. Found: C, 51.36; 4, Dc = 1.256 g cm−3, F(000) 1216, λ (Mo Kα) = 0.71073 Å, 8557
H, 4.78; N, 6.49. IR (KBr): 2966 (w), 1604 (s), 1574 (m), 1509 (s), reflections measured, and 2494 unique (Rint = 0.2499), which were
1309 (m), 1271 (s), 1178 (m), 1085 (m) cm−1. used in all calculations. The structure was solved by direct methods
11: [(η6-p-Cymene)Ru(L11)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = using the program SHELXS-97, while the refinement and all further
1.10 (d, 3JH−H = 6.9 Hz, 3H), 1.16 (d, 3JH−H = 6.9 Hz, 3H), 1.33 (t, calculations were carried out using SHELXL-97.62 The hydrogen
3
JH−H = 7.1 Hz, 3H), 2.32 (s, 3H), 2.57 (sept, 3JH−H = 6.9 Hz, 1H), atoms were included in calculated positions and treated as riding
2.82 (s, 3H), 3.93 (s, 3H), 4.30 (q, 3JH−H = 7.1 Hz, 2H), 4.78 (d, 3JH−H atoms using the SHELXL default parameters. The non-hydrogen
= 5.9 Hz, 1H), 5.13 (d, 3JH−H = 5.9 Hz, 1H), 5.26 (d, 3JH−H = 5.9 Hz, atoms were refined anisotropically, using weighted full-matrix least-
1H), 5.45 (d, 3JH−H = 5.9 Hz, 1H), 7.07 (d, 3JH−H = 8.7 Hz, 2H), 8.15 squares on F2 with 304 parameters. R1 = 0.0515 (I > 2σ(I)) and wR2 =
(d, 3JH−H = 8.7 Hz, 2H), 9.20 (s, 1H), 16.01 (s, 1H) ppm. ESI-MS m/z 0.0975, GOF = 0.629; max/min residual density 0.447/−0.729 eÅ−3.
(+): 589.9 M+. Anal. Calcd for C25H31N3O3SCl2Ru·CH2Cl2: C, 43.95; Figure 1 was drawn with ORTEP63 and the structural data deposited at
H, 4.68; Found: C, 43.67; H, 4.75. IR (KBr): 2966 (w), 1710 (m), The Cambridge Crystallographic Data Centre: CCDC 1063589.
1604 (s), 1575 (s), 1509 (s), 1477 (m), 1372 (m), 1314 (s), 1268 (s), These data can be obtained free of charge from The Cambridge
1178 (m), 1099 (s) cm−1. Crystallographic Data Centre via www.ccdc.cam.ac.uk/data_request/
cif.
12: [(η6-p-Cymene)Ru(L12)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ =
Biology. Cell Cultures. The human cervical cancer cells (HeLa),
1.13 (d, 3JH−H = 6.4 Hz, 3H), 1.20 (d, 3JH−H = 6.4 Hz, 3H), 2.35 (s,
the human ovarian cancer cells (A2780), the cisplatin-resistant human
3H), 2.47 (s, 3H), 2.62 (sept, 3JH−H = 6.4 Hz, 1H), 2.86 (s, 3H), 3.96
ovarian cancer cells (A2780cisR), and the noncancerous cells (HFL-1)
(s, 3H), 4.81 (d, 3JH−H = 4.8 Hz, 1H), 5.16 (d, 3JH−H = 4.8 Hz, 1H),
were obtained from the European Centre of Cell Cultures (ECACC).
5.29 (d, 3JH−H = 4.8 Hz, 1H), 5.46 (d, 3JH−H = 4.8 Hz, 1H), 7.10 (d,
3 These cell lines were cultivated under sterile conditions by using RPMI
JH−H = 8.0 Hz, 2H), 8.20 (d, 3JH−H = 8.0 Hz, 2H), 9.18 (s, 1H), 12.46
1640 (Sigma-Aldrich) growth media. This medium was supplemented
(s, 1H) ppm. ESI-MS m/z (+): 560.1 M+. Anal. Calcd for with fetal calf serum (FCS, Sigma-Aldrich, 5%), antibiotics (penicillin−
C24H29N3O2SCl2Ru·H2O: C, 46.98; H, 5.09; N, 6.85. Found: C, streptomycin, Actavis Sindan Pharma, 0.1%), and glutamine (Sigma-
46.94; H, 4.91; N, 6.77. IR (KBr): 2963 (w), 1638 (m), 1604 (s), 1574 Aldrich, 0.1%) at 37 °C and CO2 (5%).
(m), 1510 (s), 1475 (m), 1374 (m), 1307 (s), 1262 (s), 1179 (m) Cell Proliferation Inhibition. The cytotoxicity activity was
cm−1. determined using the MTT assay. The cells were seeded in 96-well
13: [(η6-p-Cymene)Ru(L13)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = plates with 100 μL of cell solution (cca. 10.000 cells/well) and
1.06 (d, 3JH−H = 6.8 Hz, 3H), 1.15 (d, 3JH−H = 6.8 Hz, 3H), 2.30 (s, incubated for 24 h. The 2a−p compounds were initially dissolved in
3H), 2.45 (s, 3H), 2.59 (sept, 3JH−H = 6.8 Hz, 1H), 4.63 (d, 3JH−H = DMSO, followed by a series of successive dilutions using RPMI 1640
5.6 Hz, 1H), 5.11 (d, 3JH−H = 5.6 Hz, 1H), 5.17 (d, 3JH−H = 5.6 Hz, media, so that the final concentration of DMSO was under 0.1%.
1H), 5.46 (d, 3JH−H = 5.6 Hz, 1H), 6.44 (s, 1H), 7.51 (m, 2H), 7.91 (d, The cells were treated with thiazolo-ruthenium complexes
3
JH−H = 7.1 Hz, 1H), 9.24 (s, 1H), 13.95 (s, 1H) ppm. ESI-MS m/z (concentrations between 0.1 μM−100 μM) for 24 h, with a final
(+): 524.1 M+. Anal. Calcd for C21H24N3SCl3Ru·CH2Cl2: C, 41.10; H, volume in the well of 200 μL. After the treatment, the culture media
4.08; N, 6.54; Found: C, 41.40; H, 4.08; N, 6.33. IR (KBr): 2966 (w), were removed, MTT (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-
1604 (s), 1509 (s), 1375 (m), 1309 (s), 1261 (m), 1178 (m) cm−1. tetrazolium bromide)−Hanks media solution was added to each well,
14: [(η6-p-Cymene)Ru(L14)Cl]Cl. 1H NMR (CDCl3, 400 MHz): δ = and the plates were incubated for a further 2 h. The formazan crystals
0.98 (d, 3JH−H = 6.8 Hz, 3H), 1.03 (d, 3JH−H = 6.8 Hz, 3H), 2.18 (s, formed by the mitochondrial dehydrogenase activity of vital cells were
3H), 2.40 (sept, 3JH−H = 6.8 Hz, 1H), 4.02 (d, 3JH−H = 5.9 Hz, 1H), dissolved in DMSO.
4.42 (d, 3JH−H = 5.9 Hz, 1H), 4.69 (d, 3JH−H = 5.9 Hz, 1H), 4.78 (d, The optical density, quantified by colorimetric measurements, is
3
JH−H = 5.9 Hz, 1H), 6.77 (s, 1H), 7.54 (m, 5H), 7.90 (m, 3H), 8.50 directly proportional to the amount of formazan crystals formed in the
(s, 1H), 9.27 (s, 1H), 16.05 (s, 1H) ppm. ESI-MS m/z (+): 584.0 M+. cells, and it is an indicator of the cellular viability. The plates were

8486 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

measured with a multimode microplate reader (Biotek Synergy 2 (UPL#15); ZMAT3(NM_152240.2) F-ccaggaaagaagggaatgagt, R-
Multi-Mode Microplate Reader with SQ Xenon Flash light source), gcggggattgaagtaaggac (UPL#2); RPS27L (NM_015920.3) F-cagatcgc-
and absorbance was detected at 570 nm. ttgcagcttg, R-tcttccaaggacggatgtagtaa (UPL#23); PRMT2
Note: Cisplatin and oxaliplatin drugs were used as a positive control (NM_206962.2) F-ccagtgtggagaaggcaca; R-ggggaagactttttctccaac
in our experiment, in the same concentrations of the studied (UPL#21); BAX (NM_138761.3) F-caagaccagggtggttgg, R-cactcccgc-
compounds. All of the experiments were performed in triplicate. cacaaagat (UPL#55); AEN (NM_022767.3) F-tgcagaccggaagagacac,
Statistical Analysis. Values are given as the mean ± SEM. Data are
R-ggaagcctggggagtaatct (UPL#43); TRIAP1 (NM_016399.2) F-
represented as averages of independent experiments, performed in
gcagttcaggaattaagatacttgg, R- gctggcaatagcagatctttg (UPL#69);
triplicate. The experimental data were processed with Graph Pad
Prism 5 biostatistics software. BNIP3L (NM_004331.2) F-aatgtcgtcccacctagtcg, R-agctccac-
RNA Processing. Total RNA from treated and parental ovarian ccaggaactgt (UPL#70); HTRA2 (NM_013247.4) F-attggggtga-
carcinoma cell lines (A2780 and A2780cisR) was isolated with tgatgctgac, R-agcttggttctcgaagctgt (UPL#89); FAS (NM_000043.4)
TriReagent (Sigma-Aldrich) following the classical protocol and F-tttctcaggcatcaaaagca, R-ggagaggtggcaaagctcta (UPL#15); TRIB3
purified with RNeasy Mini kit (Qiagen, Hilden, Germany). RNA (NM_021158.3) F-ccgtcttgggccctatgt, R-cttcctggacggggtaca
quality and quantity were checked with a Lab on-a-chip Bioanalyzer (UPL#1); NUPR1 (NM_012385.2) F-ctatagcctggcccattcct, R-
2100 (Agilent Technologies, Santa Clara, CA, USA) and a NanoDrop tctctcttggtgcgaccttt (UPL#10); PMAIP1 (NM_021127.2) F-
ND-1000 spectrophotometer (Thermo Scientific, Wilmington, DE, ggagatgcctgggaagaag, R-ccaaatctcctgagttgagtagc (UPL#11); BBC3
USA). The RNA Integrity Number (RIN) was calculated for each (NM_014417.3) F-gacctcaacgcacagtacga, R-gagattgtacaggaccctcca
sample and was used to assess the integrity of extracted RNA. To (UPL#68); PDCD4 (NM_145341.3) F-tggaaagcgtaaagatagtgtgtg, R-
reduce biases due to poor RNA quality, only RNAs with RIN > 8 were ttctttcagcagcatatcaatctc (UPL#10); ERCC2 (NM_000400.3) F-
considered for further analysis.
cagatggcacagcccttc, R-tcctcttcagcgtctcctct (UPL#1); GADD45A
Microarray Expression Profiling. For each sample, 100 ng of total
RNA was labeled (Cy3) and amplified using Low Input Quick Amp (NM_001924.3) F-ggagagcagaagaccgaaag, R-agtgatcgtgcgctgactc
Labeling Kit (Agilent Technologies, Santa Clara, CA, USA) according (UPL#37); HMGB1 (NM_002128.4) F-gagtaatgttacagagcggagaga,
to the manufacturer’s instructions. Labeled cRNA targets were further R-aatgtactgcaatggctgtgag (UPL#75); TNFRSF8 (NM001243.3) F-
purified using the RNeasy Mini Kit (Qiagen, Hilden, Germany) and gctgtcaggaggtgctgttac, R-gtaggcctctgtgggcact (UPL#43); and RN18S1
quantified with a NanoDrop ND-1000 spectrophotometer. All samples (NR_003286.2) F-gcaattattccccatgaacg, R-gggacttaatcaacgcacgc
had a minimum of 1.65 μg of cRNA and Cy3 specific activity >6. For (UPL#48). Complementary DNA (cDNA) was generated from 500
each cell line (A2780 and A2780cisR) and each treatment, three ng of total RNA, with First Strand cDNA Synthesis Kit (Roche
biological replicates were hybridized to a 4 × 44k Whole Human Applied Science, Germany). For each PCR reaction were used 2.5 μL
Genome Oligo Microarray (Agilent Technologies, Santa Clara, CA, of 1:10 (v/v) dilution of the first cDNA, 0.5 μM of every primers, and
USA) for 17 h at 65 °C, then washed and dried following the Agilent 0.2 μM UPL. The PCR conditions included a 10 min step at 95 °C for
protocol. Slides were scanned with an Agilent Technologies Scanner enzyme activation followed by an amplification step including 35
G2505C US45102867 at 5 μm resolution, and pixel intensities were cycles of 15 s at 95 °C, 20 s at 55 °C, and 1 s at 72 °C followed by a
quantified using Feature Extraction software v. 11.5.1.1 (Agilent
final cooling step at 40 °C for 30 s. The expression of selected genes
Technologies, Santa Clara, CA, USA).
Microarray Data Analysis. Raw microarray data were analyzed with was calculated with the ΔΔCt method64 after their normalization with
Gene Spring GX v.11.5.1 (Agilent Technologies, Santa Clara, CA, the RN18S1 housekeeping gene.
USA). The signal intensities were normalized using the Quantile Apoptosis Assessment. Apoptosis was assessed by fluorescence
method. Features flagged as outliers, nonuniform, or saturated were microscopy, using the Multi-Parameter Apoptosis Assay Kit (Cayman
removed from subsequent analysis. Differentially expressed transcripts cat no 600330, Estonia), that contains tetramethylrhodamine ethyl
induced by two compounds in each line were selected using ANOVA ester (TMRE) and Hoechst dye staining agents. The HeLa, A2780,
followed by Tukey’s posthoc test and taking a fold change (Fc) cutoff and A2780cisR cells were seeded at a density of 20000 cells/well in 96-
of 1.5. To account for multiple testing, the p-values were corrected well plates, with 200 μL of culture media and treated for 24 h with
using the Benjamini−Hochberg procedure. The differentially ex- complexes 4 and 14 at concentrations corresponding to the IC50
pressed genes were subjected to agglomerative hierarchical clustering values. Similarly, as a positive control, cells were treated with
based on Euclidian distances and the Ward method. GO enrichment staurosporine (2 μg/mL). Cells were harvested, then washed once
analysis was performed for the data set of common differentially with phosphate-buffered saline (PBS), and centrifuged at 500 rpm for
expressed genes (DEGs) induced by the two compounds in the A2780
5 min. Further, the cells were resuspended and stained with Hoechst-
cell line but also in the A2780cisR cell line. The significance of GO
TMRE costaining solution and 100 μL/mL of culture medium, and
categories was assessed by a hypergeometric distribution, and the p-
value was adjusted using the Benjamini−Yekutieli correction. Differ- incubated for 15 min at 37 °C in CO2. A ZEISS LSM 510 inverted
entially expressed genes were mapped to molecular function and fluorescence microscope at 20× magnification was used to capture the
canonical pathways in IPA. The upstream regulators considered key images, in order to assess the cell death mechanisms. The number of
molecules that can affect the expression of their target genes were apoptotic cells was measured by assessing the percentage of cells
identified with IPA upstream regulator analysis. A z-score defining the displaying condensed or fragmented nuclei. Approximately 200 nuclei
activation state of the upstream regulators (“activated” or “inhibited”) were counted per sample.
was calculated based on Fischer’s exact test (p < 0.01). A z score Autophagy Assessment. Autophagy was evaluated using an
greater than 2 defined a statistically significant “activated” upstream Autophagy/Cytotoxicity Dual Staining Kit (Cayman Chemical Co,
regulator and a value smaller than −2 defined an “inhibited” status. Ann Arbor, MI, USA), in accordance with the manufacturer’s
Validation of Microarray Data by Quantitative Real-Time PCR recommendation for the multiplate reader detection. The HeLa,
(qRT-PCR). The expression of 21 genes of interest identified by
A2780, and A2780cisR cells were seeded at a density of 20000 cells/
microarray was validated with qRT-PCR reaction using a Light Cycler
well in 96-well plates, with 200 μL of culture media, and treated for 24
480 (Roche) device. A specific set of primers and a fluorescent probe,
named Universal Probe Library (UPL), were identified for every h with complexes 4 and 14 at concentrations corresponding to the
selected gene, with Roche Applied Science software as follows: IC50 values. As a positive control, cells were treated in a similar manner
DRAM1 (NM_018370.2) F-tgtctgtgcttcactaatttcca, R-tcacagatcg- with tamoxifen (10 μM). The fluorescence intensity was evaluated
cactcactacg (UPL#78); TP53INP1 (NM_033285.3) F-catagccca- using a BioTek Microplate Reader. The fluorescence intensity of the
agtagtcccaga, R-gcaagagctgcaacataacaat (UPL#43); TP53I3 autophagy marker MDC (monodansylcadaverine) was determined at
(NM_004881.4) F-gatttgcctgatcctcttcg, R-aagtgggatggcggtaca 355 nm and the emission wavelength at 512 nm.65

8487 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry


Article

ASSOCIATED CONTENT Drugs. Organometallics 2012, 31, 5677−5685. (b) Kilpin, K. J.; Crot,
*
S Supporting Information
S.; Riedel, T.; Kitchen, J. A.; Dyson, P. J. Ruthenium (II) and osmium
(II) 1,2,3-triazolylidene organometallics: a preliminary investigation
The Supporting Information is available free of charge on the into the biological activity of ’click’ carbene complexes. Dalton Trans.
ACS Publications website at DOI: 10.1021/acs.jmed- 2014, 43, 1443−1448.
chem.5b00855. (8) Sava, G.; Zorzet, S.; Turrin, C.; Vita, F.; Soranzo, M.; Zabucchi,
Comparative yields for the synthesis of the complexes; G.; Cocchietto, M.; Bergamo, A.; Giovine, S.; Pezzoni, G.; Sartor, L.;
comparison of the antiproliferative activity of complexes Garbisa, S. Dual action of NAMI-A in inhibition of solid tumor
1−16 to cisplatin and oxaliplatin on the cancerous cell metastasis: selective targeting of metastatic cells and binding to
lines, HeLa, A2780, and A2780cisR; GO terms enriched collagen. Clin. Cancer Res. 2003, 91, 1898−1905.
(9) Bergamo, A.; Masi, A.; Dyson, P. J.; Sava, G. Modulation of the
in the common set of genes modulated in the A2780cisR metastatic progression of breast cancer with an organometallic
ovarian cell line with complexes 4 and 14; the ruthenium compound. Int. J. Oncol. 2008, 33, 1281−1289.
differentially expressed genes in the ovarian A2780cisR (10) Moreno, V.; Font-Bardia, M.; Calvet, T.; Lorenzo, J.; Aviles, F.
cell line, listed in GO categories related to apoptosis and X.; Garcia, M. H.; Morais, T. S.; Valente, A.; Robalo, M. P. DNA
cell death; percentages of condensed and fragmented interaction and cytotoxicity studies of new ruthenium(II) cyclo-
nuclei in cancer cells (PDF) pentadienyl derivative complexes containing heteroaromatic ligands. J.
SMILES data (CSV) Inorg. Biochem. 2011, 105, 241−249.


(11) Martinez, A.; Suarez, J.; Shand, T.; Magliozzo, R. S.; Sanchez-
Delgado, R. A. Interactions of arene-Ru(II)-chloroquine complexes of
AUTHOR INFORMATION known antimalarial and antitumor activity with human serum albumin
Corresponding Authors (HSA) and transferrin. J. Inorg. Biochem. 2011, 105, 39−45.
*(A.G.) Phone: +40-264-597257. Fax: +40-264-597257. E- (12) Kapitza, S.; Jakupec, M. A.; Uhl, M.; Keppler, B. K.; Marian, B.
mail: adriana.ignat@umfcluj.ro. The heterocyclic ruthenium(III) complex KP1019 (FFC14A) causes
*(O.B.) Phone: +40-264-590638. Fax: +40-264-590638. E- DNA damage and oxidative stress in colorectal tumor cells. Cancer
mail: ovidiubalacescu@iocn.ro. Lett. 2005, 226, 115−121.
*(B.T.) Phone: +41-32-7182499. Fax: +41-32-7182511. E-mail: (13) Du, K. J.; Wang, J. Q.; Kou, J. F.; Li, G. Y.; Wang, L. L.; Chao,
bruno.therrien@unine.ch. H.; Ji, L. N. Synthesis, DNA-binding and topoisomerase inhibitory
activity of ruthenium(II) polypyridyl complexes. Eur. J. Med. Chem.
Author Contributions 2011, 46, 1056−1065.
A.G. and O.B. equally contributed to the manuscript. (14) Li, L.; Wong, Y.; Chen, T.; Zheng, W. Ruthenium complexes
Notes containing bis-benzimidazole derivatives as a new class of apoptosis
The authors declare no competing financial interest. inducers. Dalton Trans. 2012, 41, 1138−1141.


(15) Nazarov, A.; Baquié, M.; Nowak-Sliwinska, P.; Zava, O.; Van
ACKNOWLEDGMENTS Beijnum, J.; Groessl, M.; Chisholm, D.; Ahmadi, Z.; McIndoe, J. S.;
Griffioen, A. W.; Van den Bergh, H.; Dyson, P. J. Synthesis and
This work was supported by the Swiss Enlargement characterization of a new class of anti-angiogenic agent based on
Contribution in the framework of the Romanian-Swiss ruthenium clusters. Sci. Rep. 2013, 3, 1485.
Research Program, project number IZERZO-142198/1.


(16) Nowak-Sliwinska, P.; Van Beijnum, J.; Casini, A.; Nazarov, A.;
Wagnières, G.; Van den Bergh, H.; Dyson, P. J.; Griffioen, A. W.
REFERENCES Organometallic ruthenium(II) arene compounds with antiangiogenic
(1) Jemal, A.; Bray, F.; Center, M. M.; Ferlay, J.; Ward, E.; Forman, activity. J. Med. Chem. 2011, 54, 3895−3902.
D. Global cancer statistics. CA-Cancer J. Clin. 2011, 61, 69−90. (17) Grozav, A.; Găină, L.; Pileczki, V.; Crisan, O.; Silaghi-
(2) Muggia, F. Platinum compounds 30 years after the introduction Dumitrescu, L.; Therrien, B.; Zaharia, V.; Berindan-Neagoe, I. The
of cisplatin: implications for the treatment of ovarian cancer. Gynecol. synthesis and antiproliferative activities of new arylidene-hydrazinyl-
Oncol. 2009, 112, 275−281. thiazole derivatives. Int. J. Mol. Sci. 2014, 15, 22059−22072.
(3) Wheate, N. J.; Walker, S.; Craig, G. E.; Oun, R. The status of (18) Sleebs, B. E.; Kersten, W. J. A.; Kulasegaram, S.;
platinum anticancer drugs in the clinic and in clinical trials. Dalton Nikolakopoulos, G.; Hatzis, E.; Moss, R. M.; Parisot, J. P.; Yang, H.;
Trans. 2010, 39, 8113−8127. Czabotar, P. E.; Fairlie, W. D.; Lee, E. F.; Adams, J. M.; Chen, L.; van
(4) Rademaker-Lakhai, J. M.; Bongard, D.; Pluim, D.; Beijnen, J. H.; Delft, M. F.; Lowes, K. N.; Wei, A.; Huang, D. C.S.; Colman, P. M.;
Schellens, J. H. A phase I and pharmacological study with imidazolium- Street, I. P.; Baell, J. B.; Watson, K.; Lessene, G. Discovery of potent
trans-DMSO-imidazole-tetrachlororuthenate, a novel ruthenium anti- and selective benzothiazole hydrazone inhibitors of Bcl-XL. J. Med.
cancer agent. Clin. Cancer Res. 2004, 10, 3717−3727. Chem. 2013, 56, 5514−5540.
(5) (a) Hartinger, C. G.; Jakupec, M. A.; Zorbas-Seifried, S.; Groessl, (19) Dahlgren, M. K.; Garcia, A. B.; Hare, A. A.; Tirado-Rives, J.;
M.; Egger, A.; Bergar, W.; Zorbas, H.; Dyson, P. J.; Keppler, B. K. Leng, L.; Bucala, R.; Jorgensen, W. L. Virtual screening and
KP1019, a new redox-active anticancer agent-preclinical development optimization yield low-nanomolar inhibitors of the tautomerase
and results of a clinical phase I study in tumor patients. Chem. activity of Plasmodium falciparum macrophage migration inhibitory
Biodiversity 2008, 5, 2140−2155. (b) Trondl, R.; Heffeter, P.; Kowol, factor. J. Med. Chem. 2012, 55, 10148−10159.
C. R.; Jakupec, M. A.; Berger, W.; Keppler, B. K. NKP-1339, the first (20) Zheng, S.; Zhong, Q.; Xi, Y.; Mottamal, M.; Zhang, Q.;
ruthenium-based anticancer drug on the edge to clinical application. Schroeder, R. L.; Sridhar, J.; He, L.; McFerrin, H.; Wang, G.
Chem. Sci. 2014, 5, 2925−2932. Modification and biological evaluation of thiazole derivatives as novel
(6) (a) Gupta, G.; Nowak-Sliwinska, P.; Herrero, N.; Dyson, P. J.; inhibitors of metastatic cancer cell migration and invasion. J. Med.
Therrien, B. Highly water soluble trithiolato-bridged dinuclear arene Chem. 2014, 57, 6653−6667.
ruthenium complexes. Inorg. Chim. Acta 2014, 423, 524−529. (21) (a) Andreani, A.; Burnelli, S.; Granaiola, M.; Leoni, A.; Locatelli,
(b) Sava, G.; Bergamo, A.; Dyson, P. J. Metal-based antitumour A.; Morigi, R.; Rambaldi, M.; Varoli, L.; Calonghi, N.; Cappadone, C.;
drugs in the post-genomic era: what comes next? Dalton Trans. 2011, Farruggia, G.; Zini, M.; Stefanelli, C.; Masotti, L.; Radin, N. S.;
40, 9069−9075. Shoemaker, R. H. New antitumor imidazo[2,1-b]thiazole guanylhy-
(7) (a) Hartinger, C. G.; Metzler-Nolte, N.; Dyson, P. J. Challenges drazones and analogues. J. Med. Chem. 2008, 51, 809−816.
and Opportunities in the Development of Organometallic Anticancer (b) Thomas, C. J.; McCormick, M. M.; Vialas, C.; Tao, Z.-F.;

8488 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

Leitheiser, C. J.; Rishel, M. J.; Wu, X.; Hecht, S. M. Alteration of the (38) Bersani, C.; Xu, L.-D.; Vilborg, A.; Lui, W.-O.; Wiman, K. G.
selectivity of DNA cleavage by a deglycobleomycin analogue Wig-1 regulates cell cycle arrest and cell death through the p53 targets
containing a trithiazole moiety. J. Am. Chem. Soc. 2002, 124, 3875− FAS and 14−3-3σ. Oncogene 2014, 33, 4407−4417.
3884. (39) Méndez Vidal, C.; Prahl, M.; Wiman, K. G. The p53-induced
(22) Ignat, A.; Lovasz, T.; Vasilescu, M.; Fischer-Fodor, E.; Tatomir, Wig-1 protein binds double-stranded RNAs with structural character-
C. B.; Cristea, C.; Silaghi-Dumitrescu, L.; Zaharia, V. Heterocycles 27. istics of siRNAs and miRNAs. FEBS Lett. 2006, 580, 4401−4408.
Microwave assisted synthesis and antitumour activity of novel (40) Vilborg, A.; Glahder, J. A.; Wilhelm, M. T.; Bersani, C.;
phenothiazinyl-thiazolyl-hydrazine derivatives. Arch. Pharm. 2012, Corcoran, M.; Mahmoudi, S.; Rosenstierne, M.; Grandér, D.; Farnebo,
345, 574−583. M.; Norrild, B.; Wiman, K. G. The p53 target Wig-1 regulates p53
(23) Prasad, K. T.; Therrien, B.; Mohan Rao, K. Cationic half- mRNA stability through an AU-rich element. Proc. Natl. Acad. Sci. U. S.
sandwich complexes (Rh, Ir, Ru) containing 2-substituted-1,8- A. 2009, 106, 15756−15761.
naphthyridine chelating ligands: Syntheses, X-ray structure analyses (41) Porté, S.; Valencia, E.; Yakovtseva, E. A.; Borràs, E.; Shafqat, N.;
and spectroscopic studies. J. Organomet. Chem. 2008, 693, 3049−3056. Debreczeny, J. E.; Pike, A. C.; Oppermann, U.; Farrés, J.; Fita, I.; Parés,
(24) Prasad, K. T.; Therrien, B.; Mohan Rao, K. Mono and dinuclear X. Three-dimensional structure and enzymatic function of proapop-
complexes of half-sandwich platinum group metals (Ru, Rh and Ir) totic human p53-inducible quinone oxidoreductase PIG3. J. Biol.
bearing a flexible pyridyl-thiazole multidentate donor ligand. J. Chem. 2009, 284, 17194−17205.
Organomet. Chem. 2010, 695, 226−234. (42) Flatt, P. M.; Polyak, K.; Tang, L. J.; Scatena, C. D.; Westfall, M.
(25) Gras, M.; Therrien, B.; Süss-Fink, G.; Casini, A.; Edafe, F.; D.; Rubinstein, L. A.; Yu, J.; Kinzler, K. W.; Vogelstein, B.; Hill, D. E.;
Dyson, P. J. Anticancer activity of new organo-ruthenium, rhodium Pietenpol, J. A. p53-dependent expression of PIG3 during prolifer-
and iridium complexes containing the 2-(pyridine-2-yl)thiazole N,N- ation, genotoxic stress, and reversible growth arrest. Cancer Lett. 2000,
chelating ligand. J. Organomet. Chem. 2010, 695, 1119−1125. 156, 63−72.
(26) Advanced Chemistry Development. ACD/ChemSketch, version (43) He, H.; Sun, Y. Ribosomal protein S27L is a direct p53 target
12.0; Advanced Chemistry Development: Toronto, Canada, 2012. that regulates apoptosis. Oncogene 2007, 26, 2707−2716.
(27) Seillier, M.; Peuget, S.; Gayet, O.; Gauthier, C.; N’Guessan, P.; (44) Karin, M.; Lin, A. NF-κB at the crossroads of life and death. Nat.
Monte, M.; Carrier, A.; Iovanna, J. L.; Dusetti, N. J. TP53INP1, a Immunol. 2002, 3, 221−227.
tumor suppressor, interacts with LC3 and ATG8-family proteins (45) Ganesh, L.; Yoshimoto, T.; Moorthy, N. C.; Akahata, W.;
through the LC3-interacting region (LIR) and promotes autophagy- Boehm, M.; Nabel, E. G.; Nabel, G. J. Protein methyltransferase 2
dependent cell death. Cell Death Differ. 2012, 19, 1525−1535. inhibits NF-kappaB function and promotes apoptosis. Mol. Cell. Biol.
(28) Nihira, K.; Miki, Y.; Ono, K.; Suzuki, T.; Sasano, H. An 2006, 26, 3864−3874.
inhibition of p62/SQSTM1 caused autophagic cell death of several (46) Follis, A. V.; Chipuk, J. E.; Fisher, J. C.; Yun, M.-K.; Grace, C.
human carcinoma cells. Cancer Sci. 2014, 105, 568−575. R.; Nourse, A.; Baran, K.; Ou, L.; Min, L.; White, S. W.; Green, D. R.;
(29) Crighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Kriwacki, R. W. PUMA binding induces partial unfolding within BCL-
Harrison, P. R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K. M. xL to disrupt p53 binding and promote apoptosis. Nat. Chem. Biol.
DRAM, a p53-induced modulator of autophagy, is critical for 2013, 9, 163−168.
apoptosis. Cell 2006, 126, 121−134. (47) Kim, J.-Y.; So, K.-J.; Lee, S.; Park, J.-H. Bcl-rambo induces
(30) Zhang, X.-D.; Qi, L.; Wu, J.-C.; Qin, Z.-H. DRAM1 regulates apoptosis via interaction with the adenine nucleotide translocator.
autophagy flux through lysosomes. PLoS One 2013, 8, e63245. FEBS Lett. 2012, 586, 3142−3149.
(31) Eby, K. G.; Rosenbluth, J. M.; Mays, D. J.; Marshall, C. B.; (48) Barrera-Vilarmau, S.; Obregón, P.; de Alba, E. Intrinsic order
Barton, C. E.; Sinha, S.; Johnson, K. N.; Tang, L.; Pietenpol, J. A. and disorder in the bcl-2 member harakiri: insights into its
ISG20L1 is a p53 family target gene that modulates genotoxic stress- proapoptotic activity. PLoS One 2011, 6, e21413.
induced autophagy. Mol. Cancer 2010, 9, 95−102. (49) Xie, Q.; Lin, T.; Zhang, Y.; Zheng, J.; Bonanno, J. A. Molecular
(32) Chaluvally-Raghavan, P.; Zhang, F.; Pradeep, S.; Hamilton, M. cloning and characterization of a human AIF-like gene with ability to
P.; Zhao, X.; Rupaimoole, R.; Moss, T.; Lu, Y.; Yu, S.; Pecot, C. V.; induce apoptosis. J. Biol. Chem. 2005, 280, 19673−19681.
Aure, M. R.; Peuget, S.; Rodriguez-Aguayo, C.; Han, H. D.; Zhang, D.; (50) Sheridan, C.; Brumatti, G.; Elgendy, M.; Brunet, M.; Martin, S.
Venkatanarayan, A.; Krohn, M.; Kristensen, V. N.; Gagea, M.; Ram, P.; J. An ERK-dependent pathway to Noxa expression regulates apoptosis
Liu, W.; Lopez-Berestein, G.; Lorenzi, P. L.; Børresen-Dale, A. L.; by platinum-based chemotherapeutic drugs. Oncogene 2010, 29, 6428−
Chin, K.; Gray, J.; Dusetti, N. J.; McGuire, S. E.; Flores, E. R.; Sood, A. 6441.
K.; Mills, G. B. Copy number gain of hsa-miR-569 at 3q26.2 leads to (51) Salvador, J. M.; Brown-Clay, J. D.; Fornace, A. J., Jr. Gadd45 in
loss of TP53INP1 and aggressiveness of epithelial cancers. Cancer Cell stress signaling, cell cycle control, and apoptosis. Adv. Exp. Med. Biol.
2014, 26, 863−879. 2013, 793, 1−19.
(33) Jin, C.; Yu, W.; Lou, X.; Zhou, F.; Han, X.; Zhao, N.; Lin, B. (52) Zhang, P.; Liu, S. S.; Ngan, H. Y. S. TAp73-mediated the
UCHL1 is a putative tumor suppressor in ovarian cancer cells and activation of c-Jun N-terminal kinase enhances cellular chemo-
contributes to cisplatin resistance. J. Cancer 2013, 4, 662−670. sensitivity to cisplatin in ovarian cancer cells. PLoS One 2012, 7,
(34) Sosna, J.; Voigt, S.; Mathieu, S.; Kabelitz, D.; Trad, A.; Janssen, e42985.
O.; Meyer-Schwesinger, C.; Schütze, S.; Adam, D. The proteases (53) Yuan, Z.; Cao, K.; Lin, C.; Li, L.; Liu, H.-y.; Zhao, X.-y.; Liu, L.;
HtrA2/Omi and UCH-L1 regulate TNF-induced necroptosis. Cell Deng, H.-x.; Li, J.; Nie, C.-l.; Wei, Y.q. The p53 upregulated modulator
Commun. Signaling 2013, 11, 76−83. of apoptosis (PUMA) chemosensitizes intrinsically resistant ovarian
(35) Yang, F.; Long, W.; Xuechuan, H.; Xueqin, L.; Hongyun, M.; cancer cells to cisplatin by lowering the threshold set by Bcl-x(L) and
Yonghui, D. Upregulation of fas in epithelial ovarian cancer reverses Mcl-1. Mol. Med. 2011, 17, 1262−1274.
the development of resistance to cisplatin. BMB Rep. 2015, 48, 30−35. (54) Zhao, Z.; Wang, J.; Tang, J.; Liu, X.; Zhong, Q.; Wang, F.; Hu,
(36) Kisova, A.; Zerzankova, L.; Habtemariam, A.; Sadler, P. J.; W.; Yuan, Z.; Nie, C.; Wei, Y. JNK- and Akt-mediated Puma
Brabec, V.; Kasparkova, J. Differences in the cellular response and expression in the apoptosis of cisplatin-resistant ovarian cancer cells.
signaling pathways between cisplatin and monodentate organometallic Biochem. J. 2012, 444, 291−301.
Ru(II) antitumor complexes containing a terphenyl ligand. Mol. (55) Li, H.; Xu, H.; Shen, H.; Li, H. MicroRNA-106a modulates
Pharmaceutics 2011, 8, 949−957. cisplatin sensitivity by targeting PDCD4 in human ovarian cancer cells.
(37) Guo, W.; Zou, Y.-B.; Jiang, Y.-G.; Wang, R.-W.; Zhao, Y.-P.; Ma, Oncol. Lett. 2014, 7, 183−188.
Z. Zinc induces cell cycle arrest and apoptosis by upregulation of (56) Ferraris, S. E.; Isoniemi, K.; Torvaldson, E.; Anckar, J.;
WIG-1 in esophageal squamous cancer cell line EC109. Tumor Biol. Westermarck, J.; Eriksson, J. E. Nucleolar AATF regulates c-Jun-
2011, 32, 801−808. mediated apoptosis. Mol. Biol. Cell 2012, 23, 4323−4332.

8489 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490
Journal of Medicinal Chemistry Article

(57) Cooke, S. L.; Brenton, J. D. Evolution of platinum resistance in


high-grade serous ovarian cancer. Lancet Oncol. 2011, 12, 1169−1174.
(58) du Bois, A.; Reuss, A.; Pujade-Lauraine, E.; Harter, P.; Ray-
Coquard, I.; Pfisterer, J. Role of surgical outcome as prognostic factor
in advanced epithelial ovarian cancer: a combined exploratory analysis
of 3 prospectively randomized phase 3 multicenter trials. Cancer 2009,
115, 1234−1244.
(59) Vergote, I.; Tropé, C. G.; Amant, F.; Kristensen, G. B.; Ehlen,
T.; Johnson, N.; Verheijen, R. H. M.; van der Burg, M. E. L.; Lacave, A.
J.; Benedetti Panici, P.; Kenter, G. G.; Casado, A.; Mendiola, C.;
Coens, C.; Verleye, L.; Stuart, G. C. E.; Pecorelli, S.; Reed, N. S.
Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV
ovarian cancer. N. Engl. J. Med. 2010, 363, 943−953.
(60) Gupta, G.; Kumar, J. M.; Garci, A.; Rangaraj, N.; Nagesh, N.;
Therrien, B. Anticancer activity of half-sandwich RhIII and IrIII metalla-
prisms containing lipophilic side chains. ChemPlusChem 2014, 79,
610−618.
(61) Bennett, M. A.; Huang, T. N.; Matheson, T. W.; Smith, A. K.
(η6)-Hexamethylbenzene ruthenium complexes. Inorg. Synth. 1982, 21,
74−78.
(62) Sheldrick, G. M. A short history of SHELX. Acta Crystallogr.,
Sect. A: Found. Crystallogr. 2008, 64, 112−122.
(63) Farrugia, L. ORTEP-3 for Windows. J. Appl. Crystallogr. 1997,
30, 565.
(64) Livak, K. J.; Schmittgen, T. D. Analysis of relative gene
expression data using real-time quantitative PCR and the 2(-Delta
Delta C(T)) Method. Methods 2001, 25, 402−408.
(65) Irimie, A. I.; Braicu, C.; Zanoaga, O.; Pileczki, V.; Gherman, C.;
Berindan-Neagoe, I.; Campian, R. S. Epigallocatechin-3-gallate
suppresses cell proliferation and promotes apoptosis and autophagy
in oral cancer SSC-4 cells. OncoTargets Ther. 2015, 8, 461−470.

8490 DOI: 10.1021/acs.jmedchem.5b00855


J. Med. Chem. 2015, 58, 8475−8490

Potrebbero piacerti anche