You are on page 1of 10

View Article Online

View Journal

INorganic
chemistry
frontiers

Accepted Manuscript

This article can be cited before page numbers have been issued, to do this please use: Y. Wu, D. Wang, J.
Hu, X. Song, C. Xie, Z. Ma and J. Xu, Inorg. Chem. Front., 2019, DOI: 10.1039/C9QI00030E.

This is an Accepted Manuscript, which has been through the


Royal Society of Chemistry peer review process and has been
Volume 3 | Number 1 | January 2016

accepted for publication.

Accepted Manuscripts are published online shortly after


acceptance, before technical editing, formatting and proof reading.
Using this free service, authors can make their results available
to the community, in citable form, before we publish the edited
article. We will replace this Accepted Manuscript with the edited
and formatted Advance Article as soon as it is available.

You can find more information about Accepted Manuscripts in the


INORGANIC author guidelines.
CHEMISTRY Please note that technical editing may introduce minor changes
FRONTIERS to the text and/or graphics, which may alter content. The journal’s
standard Terms & Conditions and the ethical guidelines, outlined
in our author and reviewer resource centre, still apply. In no
http://rsc.li/frontiers-inorganic
event shall the Royal Society of Chemistry be held responsible
for any errors or omissions in this Accepted Manuscript or any
consequences arising from the use of any information it contains.

rsc.li/frontiers-inorganic
Page 1 of 9 Inorganic
Please do Chemistry
not adjustFrontiers
margins

View Article Online


DOI: 10.1039/C9QI00030E
Journal Name

ARTICLE

Inorganic Chemistry Frontiers Accepted Manuscript


An iron(III) complex selectively mediated cancer cell death: crystal
structure, DNA targeting and in vitro antitumor activities
Received 00th January 20xx,
Accepted 00th January 20xx Yi-Gang Wu,†a Dong-Bo Wang,†ab Juan-Juan Hu,a Xue-Qing Song,a Cheng-Zhi Xie,ac Zhong-Ying Ma*a
and Jing-Yuan Xu*a
DOI: 10.1039/x0xx00000x
The design, synthesis, crystal structures and in vitro biological assessment of three iron(III) complexes namely, [Fe(L1)2]Cl
www.rsc.org/
(1), [Fe(L2)2]∙FeCl4 (2) and [Fe(L3)(HL3)2]∙(FeCl4)2∙(CH3OH) (3) (HL1 = N'-(4-oxothiazolidin-2-ylidene)picolinohydrazonamide,
HL2 = 2-(2-(1-(Pyridin-2-yl)ethylidene)hydrazono)thiazolidin-4-one, HL3 = 3-ethyl-2-(2-(1-(Pyridin-2-
yl)ethylidene)hydrazono)thiazolidin-4-one, are reported in this work. Single crystal X-ray diffraction revealed that all the
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

three complexes were mononuclear molecules, approaching a coordination polyhedron of octahedron, in which the 4-
thiazolidinone derivative ligands exhibited different coordination modes and conformations. Their interactions with DNA
were investigated by UV-visible and fluorescence spectrometry, as well as by agarose gel electrophoresis. Results indicated
that they presented efficient DNA binding propensities and cleavage activities. In vitro, iron(III) complexes caused cell cycle
arrest at G1 phase in response to DNA damage, effectively inhibited CyclinD1 expression and all decreased the migration
of HeLa cells with lower wound-healing rate than that of control. Complex 3 was most sensitive to HeLa cancer cells,
followed by 2 and 1. Attractively, the tested complexes demonstrated significantly enhanced selectivity index values
determined for HeLa vs. normal LO2 cells compared with cisplatin, especially for complex 3, which was up to 13.94-fold.
Unlike cisplatin, more complex 3 would accumulate in cancer cells HeLa compared with the normal cells LO2.

Introduction Efforts to pursue novel metal anti-cancer drugs with high


efficacy and low toxicity have been continued for decades.
The initial interest in the development of metal-based drugs Chemists are encouraged to establish alternative approaches
for cancer treatment began with platinum complex cisplatin, on the basis of using endogenous metals to improve the
which is highly successful for application in clinical oncology. pharmacological properties instead of exogenous platinum.
Nevertheless, dose limiting toxicities or intrinsic/acquired Unlike platinum, iron is an essential element for
resistance limited its application in many types of cancer. 1,2 maintaining the high growth rate of cancer cells that require
the iron-dependent enzyme ribonucleotide reductase for DNA
synthesis as well as many life processes, and has attracted
considerable research interest.3−5 Excess iron acts as a major
generator of DNA-damaging hydroxylradical (OH), a type of
reactive oxygen species (ROS), which is generally associated
with toxicity.6 It is well known that cancer cells have far
greater iron demands in comparison to normal cells. 7−9 The
microenvironment may have a positive effect on promoting
cancer growth by providing iron to cells in virtue of cancer-
associated macrophages that secrete ferritin.10 In coordination
Scheme 1 Chemical structures of the thiazolidinone derivative chemistry, iron markedly prefers octahedral geometries for
ligands NH-Tone (HL1), MH-Tone (HL2) and ME-Tone (HL3). both iron(III) and iron(II) with a six coordination number, and
the iron in center is redox active and can be redox cycled, 11,12
which are important for cytotoxicity. Undoubtedly, new iron
aTianjin Key Laboratory on Technologies Enabling Development of Clinical complexes offer a promising strategy to design antitumor drug
Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical candidates.
University, Tianjin 300070, China
bShandong Provincial Western Hospital, Shandong 250022, China Numerous iron complexes13-15 are identified to possess
cKey Laboratory of Advanced Energy Materials Chemistry (Ministry of Education),
antitumor activities via different mechanisms, usually based on
Nankai University, Tianjin 300071, China.
E-mail: xujingyuan@tmu.edu.cn; mazhongying@tmu.edu.cn
direct interactions with DNA or other biomolecules, and
† These authors contributed equally. therefore inhibit the growth of various human cancer cells. For
Electronic Supplementary Information (ESI) available: Synthesis of ligands and instance, the iron complex of glycopeptide bleomycin Fe(BLM),
supplementary figures. See DOI:10.1039/x0xx00000x

This journal is © The Royal Society of Chemistry 20xx J. Name., 2013, 00, 1-3 | 1

Please do not adjust margins


Inorganic
Please do Chemistry
not adjustFrontiers
margins Page 2 of 9

ARTICLE Journal Name

which can cause double-strand breaks in DNA, is clinically used after several days. [Fe(L1)2]Cl (1); Yield: 63.3 mg, 56.5% (based
View Article Online
in cancer treatment.16-18 This successful application has on Fe). Elemental analysis (%): calc. for DOI:
C18H10.1039/C9QI00030E
16ClFeN10O2S2: C,
motivated investigations of new iron complexes with 38.62; H, 2.88; N, 25.02. Found: C, 38.35; H, 2.56; N, 25.33. FT-
functional ligands as potential antitumor agents. Small IR (KBr disc): 3793.1, 3284.0, 3159.3, 1633.9, 1536.5, 1469.7,
molecule libraries are crucial sources for discovering new 1413.5, 1310.7, 1238.1, 1187.3, 1103.2, 786.5, 634.4, 534.7,

Inorganic Chemistry Frontiers Accepted Manuscript


drugs. Thiazolidinone derivatives, especially for 4- 478.0 cm−1 (br, broad; w, weak; m, medium; s, strong).
thiazolidinone in heterocyclic, have been investigated for a [Fe(L2)2]∙FeCl4 (2); Yield: 38 mg, 53.0% (based on Fe).
range of pharmacologic indications such as cardiovascular,19 Elemental analysis (%): calc. for C20H18Cl4Fe2N8O2S2: C, 33.36; H,
anti-inflammatory,20 antihyperlipidemic,21 antidiabetic,22 2.52; N, 15.56. Found: C, 33.62; H, 2.71; N, 15.33. FT-IR (KBr
23 24
antimicrobial, antineoplastic activity. Herein, we report disc): 3795.1, 3450.1, 3084.9, 1685.5, 1594.5, 1558.9, 1506.5,
three new FeIII coordination complexes with 4-thiazolidinone 1452.0, 1331.9, 1302.1, 1279.6, 1234.4, 1203.0, 1148.9, 1100.6,
derivative ligands (Scheme 1) (2Z,N'Z)-N'-(4-oxothiazolidin-2- 1068.7, 775.7, 537.2. [Fe(L3)(HL3)2]∙(FeCl4)2∙(CH3OH) (3); Yield:
ylidene)picolinohydrazonamide (NH-Tone, HL1), (Z)-2-((E)-(1- 58.13 mg, 68.7% (based on Fe). Elemental analysis (%): calc. for
(pyridin-2-yl)ethylidene)hydrazono)thiazolid in-4-one (MH- C37H45Cl8Fe3N12O4S3: C, 35.01; H, 3.57; N, 13.24. Found: C,
Tone, HL2) and (Z)-3-ethyl-2-((E)-(1-(pyridin-2-yl)ethylidene) 35.29; H, 3.54; N, 13.15. FT-IR (KBr disc): 3747.9, 3649.3,
hydrazono)thiazolidin-4-one (ME-Tone, HL3). With the aim of 3434.4, 2976.0, 1720.4, 1649.9, 1592.7, 1521.6, 1473.1, 1391.4,
exploring novel antitumor agents, their interactions with DNA 1373.5, 1342.6, 1246.0, 1125.8, 1038.0, 901.1, 769.6, 746.3,
and in vitro antitumor activities against tumor cells were 550.3, 449.0.
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

investigated and discussed for better understanding their X-ray crystallography


structure-activity relationships.
All diffraction data were collected at 113(2) K with a Rigaku
Saturn CCD diffractometer using MoKα radiation (λ = 0.71073
Experimental Å) with the ω-2θ scan technique. The structures were solved
by direct methods and refined with full matrix least-squares
Materials and Instrumentations technique on F2 with SHELXS-97 and SHELXL-97 programs.
All chemicals were purchased from commercial sources and Hydrogen atoms were added theoretically and riding on the
used without further purification unless otherwise noted. 2- concerned atoms, and refined with fixed thermal factors. In 1,
Acetylpyridine and 2-Cyanopyridine were obtained from the highly disordered solvent molecules could not be
Shanghai Aladdin Bio-Chem Technology Co., LTD. satisfactorily modeled. To resolve these issues, the
Thiosemicarbazide and 4-ethyl-3-thiosemicarbazide were contribution of the electron density by the remaining water
purchased from J&K Scientific Ltd. (Beijing, China). 4- molecule was removed by the SQUEEZE routine in PLATON. 25
thiazolidinone derivative ligands HL1, HL2 and HL3 were This resulted in 168 electrons per unit cell or 42 electrons per
prepared (See in the ESI) and characterized using elemental asymmetric unit and a void volume of 888 Å 3. Because a
analysis, IR, 1H-/13C-NMR (Figures S1–S12). MTT (3-(4,5- methanol molecule has 16 electrons, it means that there are
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) was two disordered methanol in one asymmetric unit.
purchased from Sigma-Aldrich. FBS (Fetal bovine serum) was Crystallographic data for these iron(III) complexes were
purchased from Hyclone. DMEM and RPMI1640 medium was deposited at the Cambridge Crystallographic Data Centre,
obtained from Gibco. CyclinD1 monoclonal antibody and beta- CCDC No. 1527069, 1527068 and 1527067.
Tubulin polyclonal antibody were obtained from Proteintech. Iron complex-DNA binding studies
Propidium iodide (PI), RNase and ROS assay kit were
The binding affinity between CT-DNA and the iron complexes
purchased from Solarbio. Cells were counted using Countstar
was determined by absorption spectral titration
cell automatic counter acquired from Advanced Lab
experiments,26-28 which were carried out by increasing
Instrument & Technology Co., Ltd. 1H and 13C spectra were
amounts of CT-DNA (1.027 × 10−4 M) to the complexes in Tris-
recorded on 400 MHz Varian Bruker NMR spectrometer.
HCl/NaCl buffer (5 mM Tris-HCl, 50 mM NaCl, pH = 7.2). Before
Infrared spectra (IR) were recorded on a Nicolet 380
scanning full wavelength, the complexes-DNA solutions were
spectrometer.
balanced at room temperature for 5 min.
Preparation of [Fe(L1)2]Cl (1), [Fe(L2)2]∙FeCl4 (2) and [Fe(L3)(HL3)2]∙ The fluorescence titration experiment was also carried out
in Tris-HCl/NaCl buffer. The solutions of the iron complexes
(FeCl4)2∙( CH3OH) (3)
were titrated into the EB-DNA solution (5 × 10−5 M EB and 5 ×
The complexes 1–3 were prepared by the similar method in 10−5 M CT-DNA). The fluorescence quenching results were
which a 15 mL methanol solution of FeCl 3∙6H2O (0.054 g, 0.2 obtained by recording the variation of the fluorescence
mmol) was added dropwise to a 10 mL acetonitrile solution of emission spectra at different complex concentrations. The
4-thiazolidinone derivative ligands HL1, HL2 or HL3 (0.2 mmol) samples were excited at 510 nm, and the emission range was
under stirring for 3 h at room temperature. Then the black recorded from 530 to 700 nm.
suspension was filtered and black crystals suitable for X-ray DNA cleavage studies
diffraction were obtained by slow evaporation of the filtrate

2 | J. Name., 2012, 00, 1-3 This journal is © The Royal Society of Chemistry 20xx

Please do not adjust margins


Page 3 of 9 Inorganic Chemistry Frontiers

ARTICLE Journal Name

The DNA cleavage experiments were performed by agarose gel supernatant was transferred into an ICP-MS auto View
sampler tube
Article Online
electrophoresis.29 Compounds were dissolved and diluted to DOI: 10.1039/C9QI00030E
to be detected. The Fe/Pt content is expressed as ng Fe/Pt per
various concentrations with Tris-HCl/NaCl buffer (50 mM Tris- million cells.
HCl, 18 mM NaCl, pH = 7.2), respectively. The mixtures of 2 μL Western blot
pUC19 DNA (0.05 μg μL−1) and 18 μL complexes in Tris-

Inorganic Chemistry Frontiers Accepted Manuscript


HCl/NaCl buffer were incubated at 37 °C for 3 h. After adding 4 HeLa cells (1 × 106 cells/well) were plated in 6-well plates. After 24
μL of loading buffer, the samples were electrophoresed on a h, the cells were exposed to compounds at 20 μM. After 24 h
1.0% agarose gel in Tris-acetate-EDTA (TAE) buffer at 90 V for treatment, the cells were harvested and washed with PBS. Protein
25 min and then stained with ethidium bromide for 30 min. was extracted from the cells, and the protein concentration was
Then bands were visualized by UV light and photographed. determined by BCA protein assay kit. 40 μg of total proteins were
separated via 10% sodium dodecyl sulfate-containing
Cell culture polyacrylamide gel and transferred onto polyvinylidene fluoride
The human epithelial cervical cancer cell line HeLa, human membranes. The bands were blocked in TBST containing 5% nonfat
breast carcinoma cell line MCF-7, human colorectal milk, and probed with primary antibodies at room temperature for
adenocarcinoma cell line Caco-2, human umbilical vein 2 h. After incubation, the bands were washed, and further
endothelial cell line HUVEC and human normal heptical cell incubated with secondary antibody conjugated with horseradish
line LO2 were obtained from the American Type Culture peroxidase. Before recorded by Tannon-5200 Multi Image Analyzer
Collection (ATCC). The cells were maintained in DMEM or (Tannon, China), the bands were fully washed. And the gray values
RPMI1640 medium containing 10% FBS in a humidified were calculated by Image J software.
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

atmosphere containing 5% CO2 at 37 °C. Wound healing assay


Cell cycle analysis HeLa cells were plated in 6-well plates for 24 h to form
For cell cycle analysis,30 HeLa cells were seeded in 6-well plates confluent monolayers, which were then wounded by
at a density of 1 × 106 cells per well and incubated for 24 h. scratching with a 200 µL tip. After washing with PBS, cells were
Then, the cells were treated with 20 μM complexes and further incubated with medium containing the three iron
further incubated for 24 h. Cells were harvested by complexes at the concentration of 40 μM respectively. The
trypsinization, washed with cold PBS and fixed in 70% ethanol widths of wound were recorded and quantified using Image J
overnight at 20 °C. Cellular pellets were washed twice with software at times of 0, 24 and 48 h.
PBS and treated with 0.25 mg mL–1 RNase A. After incubated at Annexin V-FITC/PI staining assay
37 °C for 30 min, cells were stained with 50 mg mL−1 propidium
iodide for nucleic acid staining in the dark at room HeLa cells were seeded into 6-well plates at a density of 1 ×
temperature for further 30 min. Cell cycle distribution was 106 cells per well and incubated for 24 h. Cells were treated
performed with a BD FACSCalibur flow cytometry and the data with complexes 1–3 at concentration of 40 μM. After 48 h
were analyzed by a BD CellQuestTM Pro (version 5.2) software. treatments, cells were washed, digested and collected. After
centrifugation, the cells were resuspended in 250 μL binding
MTT assay buffer, and stained with 5 μL Annexin V-FITC for 15 min at
Cells were seeded in 96-well microplates at 3000 cells per well. room temperature in the dark. Then 5 μL PI were added.
24 h later, they were treated with graded amounts of Finally, the samples were measured using a BD Accuri® C6 flow
complexes, respectively. After 48 h treatments, 10 µL of 5 mg cytometry.
mL–1 MTT solution was added to each well for 4 h. Then, the
culture medium was removed and 100 µL of DMSO was added
to solubilize the formazan crystals. Finally, the absorbance was Results and discussion
measured at a wavelength of 570 nm by an Enzyme-linked
The structure-activity relationship was investigated by altering
Immunosorbent Assay (ELISA) plate reader (Tecan Infinite® 200
the substituents in various positions of the ligands. For HL1
Pro Microplate Reader).
and HL2, the unique difference was the substituent of the
Cellular uptake position of C6, while that of N4 between HL2 and HL3 (Scheme
The intracellular Fe/Pt content was tested by inductively- 1). Specifically, HL2 was the one that substituted amino at C6
coupled plasma mass spectrometry (ICP-MS) to determine the position in HL1 with a methyl, and HL3 was formed by
selectively mediated cancer cell death. The adherent HeLa cells replacing the H-atom of N4 in HL2 with an ethyl group. The
were incubated with 50 μM complexes at 37 °C for 8 h. After steric hindrance of ethyl group in HL3 was designed to prevent
treatment, the drug-containing medium was removed and N4 participating in coordination by turning tridentate ligand to
cells were washed by ice-cold PBS for three times and bidentate one, which would lead to the change in the number
harvested in a tube. All of the samples were lyophilized by a of ligands or introduction of easy-leaving group. HL1, HL2 and
freeze dry system. Before analysis, the lyophilized samples HL3 reacting with FeCl3 gave rise to complexes 1, 2 and 3,
were digested for 4 hours at room temperature in the respectively, whose structures were confirmed by IR,
presence of 200 μL of high purity nitric acid. The samples were elemental analysis and X-ray diffraction analysis. The
centrifuged after adding 2 mL deionized water. Finally, 200 μL perspective view of the complexes was shown in Figure 1.

This journal is © The Royal Society of Chemistry 20xx J. Name., 2013, 00, 1-3 | 3

Please do not adjust margins


Inorganic
Please do Chemistry
not adjustFrontiers
margins Page 4 of 9

ARTICLE Journal Name

Crystallographic data and structure refinement parameters [Fe(1)–N(1) 1.966(2), Fe(1)–N(3) 1.891(2), Fe(1)–N(5) 1.941(2),
View Article Online
were summarized in Table 1. Selected bond angles and Fe(1)–N(6) 1.967(2), Fe(1)–N(8) 1.886(2) DOI: 10.1039/C9QI00030E
and Fe(1)–N(10)
distances were listed in Table 2. 1.946(2) Å] from the two tridentate HL1 ligands. In [Fe(L1) 2]Cl,
As shown in Figure 1a, complex 1 crystallized in the atoms N1, N5, N6, N10 constituted the equatorial plane of the
monoclinic crystal system with the P2(1)/n space group, which octahedron, and the center iron(III) atom lied 0.0010(3) Å

Inorganic Chemistry Frontiers Accepted Manuscript


comprised cationic mononuclear iron(III) units and chlorine above the equatorial plane. Complex 2, which crystallized with
ions. The imino hydrogen atom in thiazolidinone ring of ligand complex cation [Fe(L2)2]+ and counter ion FeCl4-, belonged to
is deprotonated, and therefore the coordination ability of the triclinic space group P-1. The iron(III) center has a distorted
nitrogen is enhanced. iron(III) is located in a slightly disordered octahedral geometry with six coordinated nitrogen atoms
octahedron geometry, surrounded by six nitrogen atoms
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

Figure 1 DIAMOND views of 13 with the selected atom labelling scheme (hydrogen atoms are omitted for clarity).

Table 1 Summary of the crystal data for complexes 1, 2, and 3.

Complex 1 2 3
Formula C18H16ClFeN10O2S2 C20H18Cl4Fe2N8O2S2 C37H45Cl8Fe3N12O4S3
Formula weight 559.83 720.06 1269.18
T(K) 113(2) 113(2) 113(2)
Crystal system Monoclinic Triclinic Cubic
Space group P2(1)/n P-1 P2(1)3
a/ Å 11.625(2) 9.2489(7) 17.2759(8)
b/ Å 17.263(4) 11.9004(10) 17.2759(8)
c/ Å 14.828(3) 13.3311(10) 17.2759(8)
α/° 90 87.832(4) 90
β/° 104.359(4) 75.554(4) 90
γ/° 90 80.332(4) 90
V/ Å3 2882.9(10) 1400.71(19) 5156.1(4)
Z 4 2 4
ρcalcd (g cm–3) 1.290 1.707 1.635
μ (mm−1) 0.792 1.602 1.420
F(000) 1140 724 2580
Crystal size (mm) 0.20 x 0.18 x 0.15 0.18 x 0.15 x 0.12 0.20 x 0.15 x 0.12
θ Limits (°) 3.07 to 27.56 3.10 to 27.56 2.04 to 28.70
Reflections collected 36467 18034 71169
Independent reflections 6623 6324 4461
R(int) 0.0500 0.0262 0.0453
Data/restraints/parameters 6623/0/316 6324/6/355 4461/7/212
Goodness on fit on F2 1.070 1.052 1.026
R1 (I > 2σ(I)) 0.0411 0.0299 0.0331
wR2 (all data) 0.1164 0.0824 0.0869
Δρ (max./min.), (e Å−3 ) 0.900, -0.341 0.914, -0.437 0.487, -0.346

4 | J. Name., 2012, 00, 1-3 This journal is © The Royal Society of Chemistry 20xx

Please do not adjust margins


Page 5 of 9 Inorganic Chemistry Frontiers

ARTICLE Journal Name

[Fe(1)–N(1) 1.955(1), Fe(1)–N(2) 1.908(1), Fe(1)–N(4) 1.930(1), View Article Online


Fe(1)–N(5) 1.954(1), Fe(1)–N(6) 1.904(1) and Fe(1)–N(8) DOI: 10.1039/C9QI00030E
1.936(1) Å] from the two tridentate HL2 ligands. Similar to 1,
the two Ligands in 2 were not co-planar and the dihedral angle
of the two HL2 was 85.32(5)˚. N1, N4, N5, N8 constituted the

Inorganic Chemistry Frontiers Accepted Manuscript


equatorial plane of the octahedron, and the center iron(III)
atom lied 0.0013(2) Å above the equatorial plane. Complex 3
crystallized in the cubic crystal system with the P2(1)3 space
group and consisted of a mononuclear iron(III) unit, two FeCl 4–
and one methanol molecule in the structure. In
[Fe(L3)(HL3)2]∙(FeCl4)2∙(CH3OH), the iron(III) atom is
coordinated by three bidentate ligands in a distorted
octahedral coordination geometry. The Fe–N bond length
ranged from 1.954(2) to 1.968(2) Å.
By comparison of 1 and 2, the Fe–N bond length in 3
Figure 2 Absorption spectra of complex 1 (12.5 μM) in the
became longer, partly resulting from the increase of numbers
absence (black line) and presence (other lines) of increasing
of ligands involved in the coordination. As expected, both of
amounts of CT-DNA (3.65, 7.22, 10.73, 14.17, 17.55, 20.86 and
complexes 1 and 2 form 1:2 binding stoichiometry of metal to
24.11 μM) at room temperature in Tris-HCl/NaCl buffer (pH =
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

ligand, while 1:3 for 3, which implied that each iron(III) atom
7.2). Inset: Plot of (εa − εf)/(εb − εf) vs. [DNA] for the absorption
was coordinately saturated for all the three complexes and
titration of CT-DNA with complexes.
would not further covalently bind to DNA due to the steric
effect of the ligands and no easy-leaving group occupying the

Table 2 Bond Lengths [Å] and Angles [º] for 1, 2 and 3.


1
Fe(1)-N(8) 1.8862(18) Fe(1)-N(3) 1.8914(18)
Fe(1)-N(5) 1.9413(18) Fe(1)-N(10) 1.9463(18)
Fe(1)-N(1) 1.9659(18) Fe(1)-N(6) 1.9673(18)
N(8)-Fe(1)-N(3) 177.79(8) N(8)-Fe(1)-N(5) 99.08(8)
N(3)-Fe(1)-N(5) 79.72(8) N(8)-Fe(1)-N(10) 79.85(8)
N(3)-Fe(1)-N(10) 98.39(8) N(5)-Fe(1)-N(10) 95.62(8)
N(8)-Fe(1)-N(1) 100.94(8) N(3)-Fe(1)-N(1) 80.37(7)
N(5)-Fe(1)-N(1) 159.76(8) N(10)-Fe(1)-N(1) 90.85(8)
N(8)-Fe(1)-N(6) 80.35(8) N(3)-Fe(1)-N(6) 101.47(8)
N(5)-Fe(1)-N(6) 90.16(7) N(10)-Fe(1)-N(6) 160.01(8)
N(1)-Fe(1)-N(6) 90.23(7)
2
Fe(1)-N(6) 1.9041(13) Fe(1)-N(8) 1.9356(14) Figure 3 Emission spectra of EB-CT-DNA in the absence (black
Fe(1)-N(2) 1.9076(14) Fe(1)-N(5) 1.9539(14) line) and in the presence (other lines) of 1 with increasing
Fe(1)-N(4) 1.9301(14) Fe(1)-N(1) 1.9545(14) amounts (0.997.41 × 10−5 M). The arrow shows the intensity
N(6)-Fe(1)-N(2) 177.28(6) N(6)-Fe(1)-N(4) 99.55(6) changes on increasing the complex concentration. Inset: plot
N(2)-Fe(1)-N(4) 79.99(6) N(6)-Fe(1)-N(8) 80.28(6) of I0/I vs. [complex].
N(2)-Fe(1)-N(8) 97.09(6) N(4)-Fe(1)-N(8) 96.66(6)
N(6)-Fe(1)-N(5) 80.56(6) N(2)-Fe(1)-N(5) 102.11(6) coordination sphere.31 These structural features of 1, 2 and 3
N(4)-Fe(1)-N(5) 89.83(6) N(8)-Fe(1)-N(5) 160.54(6) might be important for their interaction with DNA and various
N(6)-Fe(1)-N(1) 99.82(6) N(2)-Fe(1)-N(1) 80.85(6) bioactivities. It is generally accepted that DNA is the primary
N(4)-Fe(1)-N(1) 160.23(6) N(8)-Fe(1)-N(1) 90.47(6) target for many anticancer drugs,32,33 especially for metal
N(5)-Fe(1)-N(1) 89.46(6) complexes, though there are some other biological targets in
3 tumor cells, including RNA, enzyme or proteins. In order to
Fe(1)-N(2)#1 1.954(2) Fe(1)-N(1)#1 1.969(2) investigate the interactions between these iron complexes and
N(2)#1-Fe(1)-N(2) 94.20(8) N(2)#1-Fe(1)-N(1)#1 172.15(9) DNA, electronic absorption titration and gel electrophoresis
N(2)-Fe(1)-N(1)#1 80.32(9) N(2)#2-Fe(1)-N(1)#1 91.82(8) assays were carried out.
N(1)#1-Fe(1)-N(1)#2 94.15(9) Electronic absorption titration is an effective technique to
Symmetry transformations used to generate equivalent atoms: investigate the binding properties of metal complexes to DNA.
#1 -z+1,x+1/2,-y+3/2 The titration curves for complexes 13 in Tris-HCl/NaCl buffer
#2 y-1/2,-z+3/2,-x+1 in the absence or presence of increasing amounts of CT-DNA

This journal is © The Royal Society of Chemistry 20xx J. Name., 2013, 00, 1-3 | 5

Please do not adjust margins


Inorganic
Please do Chemistry
not adjustFrontiers
margins Page 6 of 9

ARTICLE Journal Name

Table 3 Change in spectral features of Fe(III) complexes. View Article Online


DOI: 10.1039/C9QI00030E
Blue shift
Complex Change in absorptivity  (%) (M−1 cm−1)
max (nm) max (nm) Kb 104 (M–1)
[Fe(L1)2]Cl (1) Hypochromism 10.4 241 0 3.81

Inorganic Chemistry Frontiers Accepted Manuscript


[Fe(L2)3]∙FeCl4 (2) Hypochromism 10.3 241 0 4.32
[Fe(L3)(HL3)2]∙ (FeCl4)2∙( CH3OH) (3) Hypochromism 6.1 243 1 4.21

104 M–1 for 1, 4.32 × 104 for 2, and 4.21 × 104 for 3,
respectively. The observed Kb values fell in the range for other
iron complexes.29,35,36
Fluorescence titration experiments were carried out in Tris-
HCl/NaCl buffer (5 mM Tris-HCl, 50 mM NaCl, pH = 7.2) to
further clarify the mode of DNA interaction with these three
iron(III) complexes. Due to the strong intercalation between EB
and the adjacent DNA base pairs, the EB-DNA system emits
intense fluorescent light,37 which could be quenched by the
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

addition of a second molecule.38 The emission spectra of the


EB-DNA in the absence and presence of the iron complexes 1
3 were shown in Figure 3 and Figures S15 & S16. The samples
were excited at 510 nm, and the emission range was recorded
from 530 to 700 nm. With the addition of complexes 13, the
fluorescence intensities of the EB-DNA mixture decreased,
suggesting that these iron(III) complexes could bind to CT-DNA
and displace EB in the EB-DNA system. According to the
classical Stern-Volmer equation,39 I0/I = 1 + K[Q], the
quenching plots illustrated that the quenching of EB bound to
CT-DNA by complexes was in agreement with the linear Stern-
Volmer equation, in further indicating that these complexes
bound to DNA. The Kapp of the complexes were obtained from
the equation, KEB[EB] = Kapp[complex], where the complex
Figure 4. Cleavage of pUC19 DNA (0.1 μg μL−1) in the varied
concentrations were the value at a 50% reduction of the
concentrations of complex 1, 2 and 3 after 3 h incubation at
fluorescence intensity of EB and KEB = 1.0 × 107 M−1. The
37 °C in Tris-HCl/NaCl buffer (pH 7.2).
apparent binding constants (Kapp) of complexes 1, 2 and 3 at
room temperature were 3.06 × 106 M−1, 3.55 × 106 M−1 and
were shown in Figure 2 and Figures S13 & S14. In the absence
1.14 × 106 M−1, respectively, less than the binding constant of
of CT-DNA, the maximum absorbance of the three complexes
the classical intercalators and metallointercalators (107 M−1),
all appeared at 241243 nm, which could be attributed to the
which indicated that the interaction of the three complexes
π–π* electron transition of the aromatic structure of ligands.
with DNA was a moderate intercalative mode.
Upon increasing amounts of CT-DNA to 3, the band showed
The DNA cleavage activities of the compounds were
hypochromism accompanied by a slight blue shift of 1 nm
studied with pUC19 plasmid DNA in Tris-HCl/NaCl buffer (pH
(Table 3). The other two bands of 1 and 2 exhibited
7.2) in the absence of additives under similar physiological
hypochromism without any shift on the incremental addition
conditions. NH-Tone, MH-Tone and ME-Tone hardly owned
of CT-DNA. Intercalation for these iron complexes might be
cleavage activity (Figure S17). FeCl3 even inhibited the
difficult as their ligands were certainly not planar, according to
degradation of pUC19 DNA slightly. Figure 4 showed the
the crystal structures characterization of the complexes
results of the gel electrophoretic separations of pUC19 DNA
andtheir angles between the planes. The intrinsic binding
with increasing concentrations of the iron complexes. With
constant Kb was calculated as the following equations. 34
increasing concentrations (0, 50, 100, 200, 300 and 400 μM) of
[εa–εf]/[εb–εf] = [b–(b2–2Kb2Ct[DNA]/s)1/2]/2KbCt (1)
complex 3, the amount of the supercoiled form DNA (Form I)
b = 1 + KbCt + Kb[DNA]/2s (2)
decreased, while the nicked form DNA (Form II) with single-
Where εa is the extinction coefficient observed for the
strand breaks significantly increased. Complex 3 showed the
charge-transfer absorption band at a given DNA concentration,
comparable cleavage activity to cisplatin. It was worth
εf is the extinction coefficient of the complex free in solution,
mentioning that under the same conditions the plasmid DNA
εb is the extinction coefficient of the complex when fully bound
might be cleaved into different lengths and no DNA bands
to DNA, and Ct is the total metal complex concentration. As
could be observed after treatments by 1 and 2. It clearly
shown in Table 1, the intrinsic constant (Kb) values were 3.81 ×
indicated that 1 and 2 had more active nuclease activity on

6 | J. Name., 2012, 00, 1-3 This journal is © The Royal Society of Chemistry 20xx

Please do not adjust margins


Page 7 of 9 Inorganic Chemistry Frontiers

ARTICLE Journal Name

View Article Online


DOI: 10.1039/C9QI00030E

Inorganic Chemistry Frontiers Accepted Manuscript


Figure 5 Cell cycle distributions of HeLa after treatment with 20 μM complexes for 24 h.

DNA than complex 3. When we reduced the concentrations of results above suggested that the HeLa cells growth and
1 and 2 in 10-fold, the migration rate of supercoiled DNA migration were effectively inhibited by iron(III) complexes.
gradually decreased with increasing complex concentrations in To explore the correlation among DNA damage, cell cycle
a concentration-dependent manner (Figure 4b). Compared arrest and antitumor activity, cell viability of iron(III)
with 3, the enhanced DNA cleavage activity of 1 and 2 might complexes was measured using MTT assay on human cancer
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

be attributed to the differences in the stereochemical cells, including epithelial cervical cancer cell line HeLa, human
structures of the iron complexes, where a small steric
hindrance and suitable planarity were probably in favor of DNA
cleavage. Single-crystal X-ray diffraction analysis of the
complexes revealed that the hexa-coordinate ligands of 1 and
2 tended to act as tridentate ones, while bidentate for 3 which
coordinated to iron center to generate mononuclear iron
complexes with higher numbers of ligands involved in larger
steric hindrance.
In response to DNA damage, tumor cells will undergo cell
cycle arrest to provide extra time for repair.4042 If losing the
repair, DNA damage will cause cell death. The cell cycle arrest
ability of iron(III) complexes were examined and determined
by flow cytometry. HeLa cells were treated with 20 μM
complexes for 24 h, and the cell cycle distribution was shown
in Figure 5. Treatment with varied complexes caused distinct
increases of cells in G1 phase, whereas the percentage of cells
in S and G2/M phase was decreased. The population of cells in
G1 phase changed from 50.82% in control group to 63.37%, Figure 6 Compounds inhibit cell proliferation. Western blot assay
66.54% and 64.15% for complexes 1, 2 and 3, respectively. All was carried out to determine the protein expression of Cyclin D1.
the three iron(III) complexes arrested the cell cycle at the G1 Beta-Tubulin was used as an internal control.
phase in HeLa cells, which would lead to delaying the
progression of G1 cells into the S phase. The Cyclin D1 proto-
oncogene is over-expressed in many tumor cells, and which
mainly binds to the cell cycle-dependent kinase CDK4 or CDK6
to promote the cell transition from G1 to S phase. Besides, the
Cyclin D1 overexpression is always associated with poor cancer
patient survival and increasing metastasis.43,44 Therefore, the
western blot assay was performed to observe the Cyclin D1
expression after the three iron(III) complexes. As shown in
Figure 6, compared with control group, the Cyclin D1 were
markedly down-regulated especially in complex 3 treatment
group. In addition, the wound healing assay was performed to
test the effect of HeLa cell migration inhibited by iron(III)
complexes (Figure 7). The three iron(III) complexes all
decreased the migration of HeLa cells with lower wound-
Figure 7 Wound-healing assay. The HeLa cell layer was scratched
healing rate than that of control at 24 and 48 h. All of the
and incubated with varied iron(III) complexes at 40 μM for 24 and
48 h.

This journal is © The Royal Society of Chemistry 20xx J. Name., 2013, 00, 1-3 | 7

Please do not adjust margins


Inorganic
Please do Chemistry
not adjustFrontiers
margins Page 8 of 9

ARTICLE Journal Name

Table 4. Cell viability (IC50 in μM) of iron complexes and cisplatin for 48 h. View Article Online
DOI: 10.1039/C9QI00030E
Complex HeLa MCF-7 CaCo-2 LO2 SIHeLa/LO2
1 107.24 ± 8.20 72.53 ± 3.62 70.04 ± 27.52 64.45 ± 11.11 0.60
2 98.35 ± 33.40 145.93 ± 31.50 >100 108.27 ± 0.10 1.10

Inorganic Chemistry Frontiers Accepted Manuscript


3 24.16 ± 3.16 24.04 ± 0.48 >100 60.59 ± 2.24 2.51
Cisplatin 9.82 ± 0.52 5.59 ± 0.70 10.64 ± 1.11 1.80 ± 0.3 0.18

different solubility in aqueous solution. Complex 3 had a good


solubility in the medium, however, both 1 and 2 precipitated
out in the cell culture medium during the treatment process,
resulting in a reduced dose of complex to cross the cell
membrane. It implied that the biological activities of the
complexes were largely governed by the cellular uptake. As
shown in Table S1, the ICP-MS assay displayed that both Hela
and LO2 cells could take up cisplatin and the three iron
complexes after treatments. Eye-catchingly, more complex 3
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

would accumulate in cancer cells Hela compared with the


normal cells LO2, and showed approximately 4.44-fold higher
than that of cisplatin. The results were closely correlated with
the trend of cytotoxicity of compounds and confirmed the
hypothesis between cytotoxicity and drug cellular uptake. We
speculated that 3 presented significantly enhanced selectivity
index values determined for HeLa vs. normal LO2 among the
four tested compounds probably owing to its high
accumulation in tumor cells. Cell death modes are identified as
apoptosis, necrosis and autophagy.45 Annexin V-FITC/PI
staining was employed to provide a start for understanding the
pathways of cell death (Figure 8). Results indicated that the
Figure 8 Cell apoptosis of HeLa after treatment with 40 μM death of HeLa cells induced by 13 was related to apoptosis.
complexes for 48 h. The percentage of apoptosis was 4.9%, 4.5% and 20% for 1, 2
and 3, respectively, after 48 h treatment at 40 μM. Complex 3
could lead to more cells apoptosis compared with the other
breast carcinoma cell line MCF-7, human colorectal
two iron(III) complexes.
adenocarcinoma cell line Caco-2, human umbilical vein
endothelial cell line HUVEC and normal heptical cell line LO2.
All these three tested iron(III) complexes were cytotoxic to Conclusions
various cancer cells and inhibited the growth of cells at
micromolar concentrations (Table 4). Cisplatin, the most In conclusion, we present here three new iron(III) complexes
successful exogenous metal-based anticancer drug, suffers which show efficient DNA binding propensities and DNA
from severe side effects that result in treatment failure. cleavage activity, whereas electrostatic interactions for them
Despite showing most potent in the tested cancer cells, to DNA should be considered. Note that the crystal structures
cisplatin was much more cytotoxicity in normal heptical cell of iron complexes 1 and 2 consist of two tridentate ligands,
LO2 and umbilical vein endothelial cell line HUVEC compared while complex 3 shows a structure with three bidentate
with the tested iron complexes (Table 4 and Figure S18). ligands, which causes steric hindrance with higher numbers of
Notably, the cytotoxic selectivity index (the degree of ligands involved in the coordination. The results indicate that
selectivity expressed as SI = IC50 in LO2 cells/IC50 in HeLa cells) complexes 12 bind more intensively to the DNA helix than 3,
was 0.60, 1.10 and 2.51 for 1, 2 and 3, while, 0.18 for cisplatin. partially attributed to the steric factor. In response to DNA
It suggested that the selectivities of these iron(III) complexes damage, HeLa cells undergo cycle arrest at the G1 phase and
on HeLa cells were stronger than that of cisplatin with 3.33, down-regulating of Cyclin D1 expression after treatment with
6.11 and 13.94-fold, respectively. Similar IC 50 values (around these complexes. All the complexes show growth inhibitory
100 μM) were observed in 1 and 2. Unexpectedly, complex 3 effects with IC50 values ranging from 24.16 to 107.24 μM
demonstrated much lower IC50 values than that of 1 (up to against HeLa cells. Among them, complex 3 is the most
4.44-fold) and 2 (up to 4.07-fold), although 1 and 2 exhibited sensitive to HeLa, followed by 2 and 1. Attractively, the tested
significantly stronger DNA cleavage activities. We do speculate complexes demonstrate significantly enhanced SI values
that the conflictive results were mainly attributed to their determined for HeLa vs. normal LO2 compared with cisplatin,

8 | J. Name., 2012, 00, 1-3 This journal is © The Royal Society of Chemistry 20xx

Please do not adjust margins


Page 9 of 9 Inorganic Chemistry Frontiers

ARTICLE Journal Name

especially for complex 3, which is up to 13.94-fold. And the 19 S. V. Bhandari, K. G. Bothara, A. A. Patil, T. S.View Chitre, A. P.
Article Online
ICP-MS revealed that 3 selectively mediated cancer cell death Sarkate, S. T. Gore, S. C. Dangre, C. DOI: V. Khachane, Bioorg.
10.1039/C9QI00030E
Med. Chem., 2009, 17, 390400.
partly attributed to its relatively higher accumulation in cancer 20 R. Ottana, E. Mazzon, L. Dugo, F. Monforte, R. Maccari, L.
cell compared to normal cell. Moreover, complex 3 could Sautebin, G. De Luca, M. G. Vigorita, S. Alcaro, F. Ortuso, A.
effectively induce cell apoptosis. Overall positive biological P. Caputi, S. Cuzzocrea, Eur. J. Pharmacology, 2002, 448, 71

Inorganic Chemistry Frontiers Accepted Manuscript


properties of these iron complexes indicate that they deserve 80.
to be studied more deeply in connection with their possible 21 A. C. Tripathi, S. J Gupta, G. N. Fatima, P. K. Sonar, A. Verma,
S. K. Saraf, Eur. J. Med. Chem., 2014, 72, 5277.
utilization as promising anticancer drugs. 22 R. Ottanà, R. Maccari, M. Giglio, A. Del Corso, M. Cappiello,
U. Mura, S. Cosconati, L. Marinelli, E. Novellino, S. Sartini, C.
La Motta, F. Da Settimo, Eur. J. Med. Chem., 2011, 46, 2797
Acknowledgements 2806.
23 H. B. Felise, H. V. Nguyen, R. A. Pfuetzner, K. C. Barry, S. R.
This work was supported by National Natural Science Jackson, M. P. Blanc, P. A. Bronstein, T. Kline, S. I. Miller, Cell
Foundation of China (21371135), Tianjin Municipal Natural Host & Microbe., 2008, 4, 325336.
Science Foundation (17JCZDJC33100 and 13JCZDJC28200), and 24 K. S. Sharath Kumar, A. Hanumappa, M. Vetrivel, M. Hegde, Y.
R. Girish, T. R. Byregowda, S. Rao, S. C. Raghavan, K. S.
the Open Project Program of Tianjin Key Laboratory on Rangappa, Bioorg. Med. Chem. Lett., 2015, 25, 36163620.
Technologies Enabling Development of Clinical Therapeutics 25 A. L. Spek, J. Appl. Cryst., 2003, 36, 713.
and Diagnostics (Theranostics) (Tianjin Medical University), 26 B. Deka, T. Sarkar, S. Banerjee, A. Kumar, S. Mukherjee, S.
China (No. CTD2017-03). Deka, K. K. Saikia, A. Hussain, Dalton Trans., 2017, 46, 396
Published on 04 March 2019. Downloaded on 3/11/2019 1:13:02 AM.

409.
27 P. O. Asekunowo, R. A. Haque, M. R. Razali, S. W. Avicor, M. F.
F. Wajidi, Eur. J. Med. Chem., 2018, 150, 601615.
Notes and references 28 A. Presa, G. Vázquez, L. A. Barrios, O. Roubeau, L. Korrodi-
1 M. A. Miller, Y. R. Zheng, S. Gadde, C. Pfirschke, H. Zope, C. Gregório, R. Pérez-Tomás, P. Gamez, Inorg. Chem., 2018, 57,
Engblom, R. H. Kohler, Y. Iwamoto, K. S. Yang, B. Askevold, N. 40094022.
Kolishetti, M. Pittet, S. J. Lippard, O. C. Farokhzad, R. 29 Y. Gou, J. Wang, S. Chen, Z. Zhang, Y. Zhang, W. Zhang, F.
Weissleder, Nat. Commun., 2015, 6, 86928704. Yang, Eur. J. Med. Chem., 2016, 123, 354364.
2 L. Kelland, Nat. Rev. Cancer., 2007, 7, 573584. 30 P. Štarha, Z. Trávníček, R. Herchel, P. Jewula, Z. Dvořák,
3 L. M. Bystrom, S. Rivella, Free Radical Biology and Medicine, Dalton Trans., 2018, 47, 57145724.
2015, 79, 337342. 31 Y. C. Liu, Z. F. Chen, L. M. Liu, Y. Peng, X. Hong, B. Yang, H. G.
4 C. Angelé-Martínez, C. Goodman, J. Brumaghim, Liu, H. Liang, C. Orvig, Dalton Trans., 2009, 48, 1081310823.
Metallomics, 2014, 6, 13581381. 32 C. X. Zhang, S. J. Lippard, Curr. Opin. Chem. Biol., 2003, 7,
5 H. L. Elford, M. Freese, E. Passamani, H. P. Morris, J. Biol. 481489.
Chem., 1970, 245, 52285233. 33 C. H. Wu, D. H. Wu, X. Liu, G. Guoyiqibayi, D. D. Guo, G. Lv, X.
6 E. Graf, J. R. Mahoney, R. G. Bryant, J. W. Eaton, J. Biol. M. Wang, H. Yan, H. Jiang, Z. H. Lu, Inorg. Chem., 2009, 48,
Chem., 1984, 259, 36203624. 23522354.
7 N. T. Le, D. R. Richardson, Biochim. Biophys. Acta., 2002, 34 Y. Liu, R. Hammitt, D. A. Lutterman, R. P. Thummel, C. Turro,
1603, 3146. Inorg. Chem., 2007, 46, 60116021.
8 J. L. Buss, B. T. Greene, J. Turner, F. M. Torti, S. V. Torti, Curr. 35 S. Saha, R. Majumdar, M. Roy, R. R. Dighe, A. R. Chakravarty,
Top Med. Chem., 2004, 4, 16231635. Inorg. Chem., 2009, 48, 26522663.
9 D. S. Kalinowski, D. R. Richardson, Pharmacol. Rev., 2005, 57, 36 B. L. Fei, W. S. Xu, W. L. Gao, J. Zhang, Y. Zhao, J. Y. Long, C. E.
547583. Anson, A. K. Powell, J. Photochem. Photobiol. B., 2015, 142,
10 A. A. Alkhateeb, B. Han, J. R. Connor, Breast Cancer Res. 7785.
Treat., 2013, 137, 733744. 37 K. Anđelković, M. R. Milenković, A. Pevec, I. Turel, I. Z. Matić,
11 Y. Yu, D. S. Kalinowski, Z. Kovacevic, A. R. Siafakas, P. J. M. Vujčić, D. Sladić, D. Radanović, G. Brađan, S. Belošević, B.
Jansson, C. Stefani, D. B. Lovejoy, P. C. Sharpe, P. V. Čobeljić, J. Inorg. Biochem., 2017, 174, 137149.
Bernhardt, D. R. Richardson, J. Med. Chem., 2009, 52, 5271 38 M. Lee, A. L. Rhodes, M. D. Wyatt, S. Forrow, J. A. Hartley,
5294. Biochemistry, 1993, 32, 42374245.
12 D. S. Kalinowski, D. R. Richardson, Chem. Res. Toxicol., 2007, 39 K. Tummalapalli, V. C. S., P. Munusami, M. Pathak, B. M. M.,
20, 715720. Int. J. Biol. Macromol., 2017, 95, 12541266.
13 A. Hille, I. Ott, A. Kitanovic, I. Kitanovic, H. Alborzinia, E. 40 J. R. Jackson, A. Gilmartin, C. Imburgia, J. D. Winkler, L. A.
Lederer, S. Wölfl, N. Metzler-Nolte, S. Schäfer, W. S. Marshall, A. Roshak, Cancer Res., 2000, 60, 566572.
Sheldrick, C. Bischof, U. Schatzschneider, R. Gust, J. Biol. 41 P. J. Utz, P. Anderson, Cell Death and Differ., 2000, 7, 589
Inorg. Chem., 2009, 14, 711725. 602.
14 T.S. Lange, C. McCourt, R. K. Singh, K. K. Kim, A. P. Singh, B. S. 42 G. K. Dasika, S. C. J. Lin, S. Zhao, P. Sung, A. Tomkinson, E. Y-H.
Luisi, O. Alptürk, R. M. Strongin, L. Brard, Drug Des. Devel. P. Lee, Oncogene, 1999, 18, 78837899.
Ther., 2009, 3, 1726. 43 P. Neumeister, F. J. Pixley, Y. Xiong, H. Xie, K. Wu, A. Ashton,
15 A. Hille, T. Wolf, P. Schumacher, I. Ott, R. Gust, B. Kircher, M. Cammer, A. Chan, M. Symons, E. R. Stanley, R. G. Pestell,
Arch. Pharm. (Weinheim), 2011, 344, 217223. Mol. Biol. Cell., 2003, 14, 20052015.
16 M. J. Absalon, W. Wu, J. W. Kozarich, J. Stubbe, Biochemistry, 44 M. E. Ewen, J. Lamb, Trends Mol. Med., 2004, 10, 158162.
1995, 34, 20762086. 45 T. Vanden Berghe, A. Linkermann, S. Jouan-Lanhouet, H.
17 L. M. Mir, O. Tounekti, S. Orlowski, Gen. Pharmacol., 1996, Walczak, P. Vandenabeele, Nat. Rev. Mol. Cell. Biol., 2014, 15,
27, 745748. 135147.
18 L. Xie, Z. Luo, Z. Zhao, T. Chen. J. Med. Chem., 2017, 60, 202
214.

This journal is © The Royal Society of Chemistry 20xx J. Name., 2013, 00, 1-3 | 9

Please do not adjust margins

You might also like