Sunteți pe pagina 1din 12

Available online at www.sciencedirect.

com
JOURNAL OF
Inorganic
Biochemistry
Journal of Inorganic Biochemistry 102 (2008) 564575
www.elsevier.com/locate/jinorgbio

Gold(III) compounds as anticancer agents: Relevance of


goldprotein interactions for their mechanism of action
Angela Casini a,*, Christian Hartinger b, Chiara Gabbiani a, Enrico Mini c, Paul J. Dyson b,
Bernard K. Keppler d, Luigi Messori a,*
a
Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
b
Institut des Sciences et Ingenierie Chimiques, Ecole Polytechnique Federale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
c
Department of Pharmacology, University of Florence, Florence, Italy
d
Institute of Inorganic Chemistry, University of Vienna, A-1090 Vienna, Austria

Received 2 July 2007; received in revised form 1 October 2007; accepted 18 October 2007
Available online 28 November 2007

Abstract

Gold(III) compounds constitute an emerging class of biologically active substances, of special interest as potential anticancer agents.
During the past decade a number of structurally diverse gold(III) complexes were reported to be acceptably stable under physiological-
like conditions and to manifest very promising cytotoxic eects against selected human tumour cell lines, making them good candidates
as anti-tumour drugs. Some representative examples will be described in detail. There is considerable interest in understanding the precise
biochemical mechanisms of these novel cytotoxic agents. Based on experimental evidence collected so far we hypothesize that these
metallodrugs, at variance with classical platinum(II) drugs, produce in most cases their growth inhibition eects through a variety of
DNA-independent mechanisms. Notably, strong inhibition of the selenoenzyme thioredoxin reductase and associated disregulation
of mitochondrial functions were clearly documented in some selected cases, thus providing a solid biochemical basis for the pronounced
proapoptotic eects. These observations led us to investigate in detail the reactions of gold(III) compounds with a few model proteins in
order to gain molecular-level information on the possible interaction modes with possible protein targets. Valuable insight on the for-
mation and the nature of goldprotein adducts was gained through ESI MS (electrospray ionization mass spectrometry) and spectropho-
tometric studies of appropriate model systems as it is exemplied here by the reactions of two representative gold(III) compounds with
cytochrome c and ubiquitin. The mechanistic relevance of gold(III)-induced oxidative protein damage and of direct gold coordination to
protein sidechains is specically assessed. Perspectives for the future of this topics are briey outlined.
2007 Elsevier Inc. All rights reserved.

Keywords: Gold(III) complexes; Anticancer agents; Proteins; ESI mass spectrometry; Mechanism of action

1. Novel gold(III) compounds as anticancer agents: pharmacological prole of cisplatin suggested that other
structurally diverse compounds with outstanding metal-based compounds might pairwise manifest impor-
antiproliferative properties tant anti-tumour eects while (ideally) exhibiting a dierent
spectrum of biological activities and a lower systemic tox-
The discovery of the anticancer properties of cisplatin icity [2,3]. As d8 Au(III) complexes are isoelectronic and
during the 1960s triggered a great deal of interest in the isostructural with Pt(II) complexes, square planar gold(III)
eld of anti-tumour metallodrugs [1]. The very favourable compounds soon appeared to be excellent candidates for
anticancer evaluation. However, at variance with plati-
*
Corresponding authors. Fax: +39 055 457 3385.
num(II) compounds, gold(III) analogues were found to
E-mail addresses: angela.casini@uni.it (A. Casini), luigi.messori@ manifest, on the whole, a rather poor stability prole being
uni.it (L. Messori). kinetically more labile than the corresponding platinum(II)

0162-0134/$ - see front matter 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.jinorgbio.2007.11.003
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 565

compounds, light-sensitive and easily reducible to metallic The antiproliferative properties of these gold(III)poly-
gold. As a result of these diculties and, also, of detection amine were, then, measured by the sulforhodamine B assay
of important systemic toxicity in the course of the rst ani- on the representative human ovarian tumour cell line
mal studies, gold(III) compounds were quickly abandoned. A2780, either sensitive (A2780/S) or resistant (A2780/R)
Nonetheless, during the 1990s, renewed interest for to cisplatin. In most cases, the above compounds revealed
anticancer gold(III)-based compounds emerged place, a high cytotoxicity, with IC50 values generally falling in the
especially when a few novel gold(III) complexes, exhibit- low micromolar region [9], and were also found to over-
ing improved stability, lower toxicity and favourable come resistance to cisplatin in the cisplatin-resistant line.
in vitro pharmacological properties, were made available Later on, in collaboration with the group of Minghetti
for pharmacological testing [4]. For instance, a series of and Cinellu, we characterised and assayed other gold(III)
organogold(III) DAMP compounds (DAMP = 2-[(dimeth- complexes, bearing the bipyridyl motif [10]. This family of
ylamino)methyl]phenyl) were developed by Buckley et al., compounds turned out to display appreciable stability in
and screened for anti-tumour activity with encouraging solution and to cause relevant tumour growth inhibition
results in vitro [5,6]. Further these compounds demon- in vitro. In particular, our investigations focused on two
strated moderate activity in laboratory animals [5,6]. members of this family, namely [Au(bipyc-H)(OH)][PF6] 6
Later on, in the attempt of nding new biologically (where bipyc = 6-(1,1-dimethylbenzyl)-2,20 -bipyridine) and
active substances with an even better stability prole, some [Au(bipy)(OH)2][PF6] 7 (bipy = bipyridine) (Fig. 2) [9]. A
classical square planar gold(III) complexes, based on a number of analogues were also prepared and characterised
variety of structurally dierent ligands, were prepared (see Fig. 2, compounds 8 and 9) [11].
and characterised in our laboratory [7]. To improve the sta- In [Au(bipy)(OH)2][PF6] 7, the gold(III) center is coordi-
bility of the gold(III) center, polydentate ligands such as nated by two nitrogens of the bidentate bipyridyl ligand and
polyamines, cyclam, terpyridine and phenathroline were by two hydroxide groups. At variance, 6 is an organo-
preferentially employed. A few compounds, namely gold(III) complex in which donors to the gold(III) center
[Au(en)2]Cl3 1, [Au(dien)Cl]Cl2 2, [Au(cyclam)](ClO4)2Cl are two nitrogens from the bipyridyl moiety, the C2 carbon
3, [Au(terpy)Cl]Cl2 4, and [Au(phen)Cl2]Cl 5, (Fig. 1), were of the phenyl group, and a hydroxide group. Only small
characterised both in the solid state and in solution [8]. On deviations from ideal square planar geometry were seen in
the whole, a quite satisfactory stability prole in solution the classical bipyridyl complexes whereas such deviations
emerged for all these gold(III) compounds that opened are quite large in the case of cyclometallated derivatives.
the way to their in vitro pharmacological testing. This Both mentioned compounds exhibited sucient solubil-
means that the donor set comprising three or four nitrogen ity in watery solutions. Their intense LMCT visible bands,
atoms, produces a strong stabilisation of the gold(III) cen- lying in the 300370 nm region and diagnostic of gold in
ter and a net decrease of the reduction potential, thus pre- the oxidation state +3, were exploited to monitor reactions
venting gold(III) reduction and hydrolysis. with various kinds of biomolecular targets. Notably, these

3+
2+
NH2 3+
NH2
NH
NH NH 2ClO4-
Au
3Cl- Au Au Cl-
NH2 NH2
NH2 NH2
Cl
NH NH

1 2

3
2+

N N
Cl
N Au Cl Cl
2Cl Au
N N Cl

4 5

Fig. 1. Schematic drawing of (1) [Au(en)2]Cl3, (2) [Au(dien)Cl]Cl2, (3) [Au(cyclam)](ClO4)2Cl, (4) [Au(terpy)Cl]Cl2, and (5) [Au(phen)Cl2]Cl.
566 A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575

+ +
In the same period, Fregona and coworkers prepared
and characterised some interesting gold(III) dithiocarba-
N N CH3 PF6- N N PF6- mate compounds displaying a very encouraging biological
Au Au prole [14]. The compounds containing the N,N-di-
OH CH3
OH OH methyldithiocarbamate and ethylsarcosinedithiocarbamate
ligands (compounds 10 and 11 in Fig. 2) were the most
7
6 intensely studied ones. Early in vivo data on murine models
looked very promising and led to the rapid patenting of
+
these novel compounds. Afterwards, some additional
N N H3COOC N investigations were carried out aimed at elucidating the
CH3 CH3
Au PF6- Au main features of their mechanism of action, both at the cel-
CH3 HN CH3 H3COOC CH3 lular and biochemical level [15,16]. It is worth noting that
very recent results suggest that these gold(III) dithiocarba-
CH3 mate compounds most likely act through inhibition of the
9
8
O cancer cell proteasome [17], a quite novel and unexpected
H3C S X S X
mechanism for anticancer metallodrugs.
C
N C Au Au
Concomitantly, a series of interesting gold(III) meso-tet-
RO CH2 N C
raarylporphyrins complexes were developed by Chi Ming
H3C S X CH3 S X
X = Cl, Br Che, and coworkers at the University of Hong Kong [18].
10 R = CH3CH2 11 These compounds, of general formula [Au(III)(p-Y-
TPP)]Cl, [with Y being H (a), Me (b), OMe (c), Br (d) or
Fig. 2. Schematic drawing of [Au(bipyc-H)(OH)][PF6] (6), [Au(bipy)
Cl (e)] (see Fig. 3), were characterised by classical physio-
(OH)2][PF6] (7), [Au(bipydmb-H)(2.6-xylidine-H)][PF6] (8), [Au(pydmb-H)-
(AcO)2] (9) (where bipydmb = 6-(1,1-dimethylbenzyl)-2,20 -bipyridine); chemical methods [18]. Interestingly, coordination to the
pydmb = 2-(1,1-dimethylbenzyl)-pyridine)) and of the gold(III) dithiocar- tetrapyrrole ring causes a large stabilisation of the gold(III)
bamate complexes containing N,N-dimethyldithiocarbamate (10) and oxidation state leading to its high stability in aqueous solu-
ethylsarcosinedithiocarbamate (11) ligands. tions even in the presence of glutathione (GSH), the most
important intracellular reducing agent.
two complexes revealed a clearly dierent reactivity toward All the mentioned gold(III) porphyrins displayed excel-
ascorbate as only compound 7 was shown to be reduced. lent in vitro antiproliferative eects, with IC50 values of
This nding implies that the oxidation state +3 is far more 0.11.5 lM. The lack of cross-resistance with classical
stable in the case of the organogold(III) species compared platinum(II) compounds again suggests that gold(III) por-
to compound 7, in agreement with electrochemical results phyrins and cisplatin induce cytotoxicity through quite dis-
[12]. tinct mechanisms. Remarkably, a zinc(II) analogue [Zn(II)
The in vitro cytotoxic properties of these bipyridyl (TPP)] was also investigated and found to be at least 100-
Au(III) complexes were measured toward the human ovar- fold less cytotoxic than gold(III) porphyrins (IC50 >
ian carcinoma cell line A2780, either sensitive or resistant
to cisplatin. Both gold(III) complexes showed excellent
cytotoxic eects, with IC50 values staying in the low micro-
Y
molar range [9,10]. Compound 6 turned out to be the most
active with a twofold higher activity than cisplatin in the
A2780/R cell line. The antiproliferative properties of these
complexes were also investigated on the human ovarian cell
line SKOV3 (inherently resistant to cisplatin) and on the
+
CCRF-CEM leukemic cell line, either sensitive (CCRF-
CEM/S) or resistant (CCRF-CEM/R) to cisplatin: signi-
N N
cantly lower activities were measured on all these latter
Y Au Y
lines. In any case, an appreciable activity was retained
N N
toward the cisplatin-resistant A2780/R and CCRF-CEM/
R lines suggesting that the biochemical mechanisms of
resistance to cisplatin most likely a more ecient intracel-
lular detoxication and an increased repair of DNA dam-
age are modestly eective toward these gold(III)
complexes. In addition, independent studies pointed out
quite unambiguously that the interactions of these com-
plexes with nucleic acids are weak whereas stronger Y
adducts are formed in the reactions with model proteins Fig. 3. Gold(III) meso-tetraarylporphyrins complexes. Y = H (a), Me (b),
and serum proteins [13]. OMe (c), Br (d), Cl (e).
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 567

50 lM), thus stressing the crucial role of the gold(III) in vitro antiproliferative properties measured against the
center in the biological action. Nonetheless, the porphyrin reference A2780 human ovarian carcinoma cell line [21].
ligand was found to be essential for anticancer activity While ve compounds of this series manifested only
leading these authors to conclude that the porphyrin ligand moderate cytotoxic properties (with IC50 typically falling
is crucial both in stabilising the Au(III) center and in deliv- in the 1030 lM), the sixth one (Auoxo6), turned out to
ering the metal to its cellular targets [18]. be 515 times more active against both cell lines, thus
A quite common strategy in the eld of anticancer meriting further investigations. In particular, much atten-
metallodrugs is the design and synthesis of polymetallic tion was focused on the chemical and structural reasons
compounds, derived from the fusion of two or more for the higher biological activity of Auoxo6. Remarkably,
monometallic molecular fragments. It follows that the spe- a rather evident positive correlation was identied in a sub-
cic reactivity of each metal center in the resulting com- sequent study between the oxidising power of this com-
pound is further controlled by its interactions with the pound, its reactivity with biomolecular targets and its
nearby metal center(s) and by the overall molecular scaf- antiproliferative eects.
fold. Incorporation of two (or more) metal centers within In Table 1 we have summarised the cytotoxicity data for
an extended molecular framework may greatly aect the the above mentioned gold(III) compounds developed in
overall charge of the resulting polynuclear compound, its our group. According to this data the most active com-
redox properties, the kinetics of hydrolysis, and its specic pounds, compared to cisplatin, are compound 4 and
reactivity toward biomolecules in comparison to mononu- Auoxo6. For these two complexes the extension of the bio-
clear analogues. Some interesting examples of this strategy logical screening over a wider series of cancer cell lines is
are available for platinum(II) and ruthenium(III) antican- now in progress in our group. Furthermore, these most
cer metallodrugs [19,20]. promising molecular scaolds will be used for the drug
These observations prompted us to prepare novel dinu- design of novel gold(III) complexes. For example we have
clear gold(III) species to be tested as anticancer agents, recently synthesised the gold(III)-dinuclear derivative of 4
starting from the above described mononuclear gold(III) whose cytotoxicity properties are under evaluation.
bipyridyl complexes. A series of six dinuclear oxo gold(III) Overall, in the course of the last decade, thanks to the
complexes with bipyridyl ligands (Fig. 4), of general for- eorts of a few research laboratories a signicant number
mula [Au2(N,N)2(l-O)2][PF6]2 [where N,N = 2,20 -bipyri- of structurally diverse gold(III) compounds were prepared
dine (Auoxo1), 4,40 -di-tert-butyl- (Auoxo2), 6-methyl- and characterised that revealed attracting antiproliferative
(Auoxo3), 6-neopentyl- (Auoxo4), 6-(2,6-dimethylphenyl)- activities. In view of the great structural variety of the
(Auoxo5), 6,60 -dimethyl-2,20 -bipyridine (Auoxo6)] was thus investigated gold(III) species it is reasonable to assume that
prepared and characterised in our laboratory, and their their biological eects may depend on dierent biochemical

Me 3C CMe3

N O N N O N
Au Au [PF6]2 Au Au [PF6]2
N O N N O N

Me 3C CMe3

Auoxo1 Auoxo2

R Me Me

N O N N O N
Au Au [PF6]2 Au Au [PF6]2
N O N N O N

R Me Me

Auoxo3: R = Me Auoxo6
Auoxo4: R = CH2CMe 3
Auoxo5: R = C6H3Me2-2,6

Fig. 4. Schematic drawings of the dinuclear gold(III) complexes Auoxo. Auoxo3 is a ca. 1:1 mixture of the cis and trans isomer while Auoxo4 and Auoxo5
are, as depicted, exclusively trans isomers.
568 A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575

Table 1
Cytotoxicity (IC50-lM) of the gold compounds studied in Florence during the last years towards dierent tumour cell lines
Compounds A2780/S A2780/R CCRF-CEM/S CCRF-CEM/R SK-OV-3 MCF7 HT29 A549
Cisplatin 1.2 0.43 14 2.72 0.7 0.1 20.1 7.2 5.2 5.30 0.87 6.30 0.23
1 [Au(en)2]Cl3 8.36 0.77 17.0 4.24
2 [Au(dien)Cl]Cl2 8.2 0.93 18.7 2.16 12.6 2.0 32.7 6.6
3 [Au(cyclam)]ClO4)2Cl 99.0 >120.0
4 [Au(Terpy)Cl]Cl2 0.2 0.37 0.032
5 [Au(Phen)Cl2]Cl 3.8 1.1 3.49 0.91 2.3 6
6 [Au(bipyc-H)(OH)][PF6] 3.3 1.4 8.2 1.5 11.9 2.1 51.2 5.6 13.3 1.6 35.30 8.8 24.60 >50
7 [Au(bipy)(OH)2][PF6] 8.8 3.9 24.1 8.7 52.9 11.6 58.6 0.9 34.4 4.7
8 [Au(bipydmb-H)(2.6-xylidine- 2.50 0.43 5.7 0.3 5.20 0.40 25 35
H)][PF6]
9 [Au(pydmb-H)(AcO)2] 2.90 0.34 6.40 1.0 7.70 0.44 8.60 49
Auoxo1 22.8 1.53 23.3 0.35
Auoxo2 12.1 1.5 13.5 1.8
Auoxo3 25.4 2.47 29.8 3.1
Auoxo4 12.7 1.06 19.8 1.8
Auoxo5 11.0 1.5 13.2 1.2
Auoxo6 1.79 0.17 4.81 0.5
Cisplatin is reported as reference compound. Data were collected after 72 h exposure to drug.

mechanisms. Current eorts to identify those mechanisms analysed at the molecular level by taking advantage of a
are described in the next section. In any case sucient vast array of biochemical, biophysical and physicochemical
evidence has been collected so far that the presence of a methods [13,22].
gold(III) species is usually associated with outstanding A representative example is the case of gold(III) porphy-
cytotoxic properties so that the metal itself seems to be rins whose reactions with nucleic acids were investigated.
one of the major determinants of the biological actions. In particular, Sun and coworkers examined the interactions
However careful selection and chemical modication of of representative gold(III) porphyrins with duplex DNA by
the ligands might allow the respective complexes proper- UVvis absorption titrations [18]. Isosbestic changes and
ties to be eciently tuned with regard to inertness, rate signicant hypochromicity of the Soret band were noticed
of binding to biomolecules and cytotoxic activity. in the electronic spectra of gold(III) porphyrins after addi-
tion of calf-thymus DNA supporting a direct and strong
2. Current views concerning the biochemical actions of interaction with the DNA double helix. Accordingly, in a
cytotoxic gold(III) compounds: a variety of molecular subsequent study the same authors reported that treatment
mechanisms with gold(III) porphyrins induces signicant and character-
istic changes in protein expression proles [23].
As outlined above, the renaissance of interest for In general, these cellular and biochemical studies have
gold(III) compounds as potential anticancer metallodrugs provided further valuable insight into the reactivity and
has resulted, in the course of the last decade, in the synthe- the mode of action of novel gold(III)-based metallodrugs.
sis of a number of structurally diverse gold(III) species, Although the ultimate molecular mechanisms of these
endowed with sucient chemical stability and with relevant new anticancer agents remain largely unknown and elusive,
antiproliferative activities. Detection of in vitro cytotoxicity some considerations and suggestions may be anticipated
has represented, for these metal-based agents, the primary based on the available information.
screening criterion in order to assess their potential anti- Most of the mechanistic studies carried out on cytotoxic
cancer properties. IC50 values of 105 M or lower were gold(III) compounds have been generally referred and
used to indicate a promising, or at least acceptable, anti- compared to the behaviour of cisplatin, for which DNA
tumour ecacy. is one of the major putative targets [24]. However, it
Notably, for a few of these compounds, the pharmaco- emerges from the experimental results collected so far that
logical studies could be extended well beyond assessment the respective molecular mechanisms are rather distinct.
of mere in vitro cytotoxicity by analysing some of the eects Also for cisplatin the major resistance mechanisms fall
they produce at the cellular level such as direct DNA dam- into the following four categories: limitation of drug levels
age, modication of the cell cycle, alterations of mitochon- by reduced uptake and/or increased eux; increased cellu-
drial functions, induction of apoptosis, and so on. lar thiol levels; enhanced DNA repair and/or increased
Moreover, the reactions of some cytotoxic gold(III) damage tolerance; and failure of cell death pathways [25].
complexes with some specic biomolecular targets, e.g. If we consider the cytotoxic eects reported for some of
calf-thymus DNA and a few representative proteins, were the above mentioned gold(III) complexes (Table 1) it is
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 569

worth noting that most of them have similar activity Table 2


toward the cisplatin sensitive and resistant cell lines, and Inhibition (IC50-lM) of thioredoxin reductase by gold compounds
therefore may overcome the resistance problems seen with Compound IC50 (lM)
cisplatin itself. However further biological screening is Rat Rat Human
needed to demonstrate these peculiar pharmacological mitochondrial cytosolic cytosolic
properties. TrxR2 TrxR1 TrxR1
Overall, the few gold(III) compounds on which Cisplatin 36.0
advanced pharmacological testing was performed oer a Auranon 0.020
Au triethylphosphine chloride 0.065
rather variegate mechanistic scenario that drastically con- Aurothiomalate 0.280 0.005
trasts with the rather homogeneous biological pattern pro- 2 [Au(2,20 - 0.420 0.20
vided by classical platinum(II) compounds. Therefore, even diethylendiamine)Cl]Cl2
if in a few cases e.g. gold(III) porphyrins some evidence 9 [(Au(2-(1,1-dimethylbenzyl)- 1.420
for a direct DNA damage has been obtained, for most pyridine) (CH3COO)2]
6 [Au(6-(1,1-dimethylbenzyl)- 0.280
other cases, the eects on nucleic acids and on the cell cycle 2,20 - bipyridine)(OH)](PF6)
appear to be very modest so that it is very unlikely that 8 [Au(6-(1,1-dimethylbenzyl)- 0.210
DNA may be the primary target. 2,20 -bipyridine)-H)(2,6-
For instance, we have shown that the large cytotoxic xylidine)](PF6)
and proapoptotic eects induced by the gold(III) com- Tetrachloroaurate 0.0058
12 0.012
plexes 2 and 5 involve very marginal DNA damage [26]; 13 0.030
accordingly only weak cell cycle eects were detected, at 14 0.036
variance with cisplatin (that is known to produce an evi- 15 0.180
dent G2/M arrest), further suggesting a modest impact on 16 0.030
DNA related functions. A similar situation was previously 17 0.0022
18 1.80
reported for gold(III) DAMP compounds pairwise show- 19 0.68
ing very weak DNA interaction properties [5]. Thus, Au triphenylphosphine 0.001
DNA is likely not to be the primary target for several of chloride
the novel gold(III) compounds. Even in the cases where Au(III) chloride trihydrate 0.00075
some evidence of a direct interaction with DNA has been Au acetate 0.040
Au thiosulfate Na+ 0.0005
achieved, the mechanisms of DNA damage and cell death
appear to be profoundly distinct from those induced by Cisplatin is reported as reference compound.
platinum drugs.
All these arguments suggest that alternative biochemical
mechanisms must be operative, most likely associated to pounds are known to target, rather strongly and selectively,
selective modication of some crucial proteins. In this thiol and imidazole groups of proteins (as well as selenol
respect it is worth noting that gold(I) and gold(III) com- groups) [27]; this type of reactivity might reasonably repre-
sent the true molecular basis for their biological actions.
In this context, a recent hypothesis is that gold com-
pounds may exert their relevant cytotoxic and proapoptot-
ic eects through a direct antimitochondrial action: this
idea originally developed for classical antiarthritic and
cytotoxic gold(I) compounds like auranon and aurothi-
omalate (nonetheless showing a relevant cytotoxicity
in vitro) [28] has later been extended to novel anticancer
gold(III) complexes. The key event for the biological mech-
anism of gold compounds would be the direct, strong inhi-
bition of thioredoxin reductase (TrxR), a selenoenzyme
critically involved in the regulation of the intracellular
redox state and of mitochondrial functions [29]. As a con-
sequence of large TrxR inhibition the opening of the mito-
chondrial pore is induced, eventually leading to cyt c
release and apoptosis.
Recent studies have shed light into the molecular and
structural aspects of thioredoxin reductases. The thiore-
doxin reductases are enzymes belonging to the avoprotein
family of pyridine nucleotidedisulphide oxidoreductases
Fig. 5. Schematic representation of the molecular structure of mammalian that also includes lipoamide dehydrogenase, glutathione
TrxR1 visualised with the Swiss-Pdb Viewer software. reductase and mercuric ion reductase [30]. Members of this
570 A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575

N N N
Cl
Au AuCl4 AuCl3 AuCl2

Cl
N

12 13 14

NMe2 NMe2 NMe2

Au Au Au
Cl2 Ph
(OAc)2
Cl
15 16
17

Me PPh3
NMe2
Au
Me Me
Au
Me

Me
18 19

Fig. 6. Schematic drawings of gold(III) complexes screened for the inhibition of human TrxR1.

family are homodimeric proteins in which each monomer Rigobello in Padova, we have observed that a few gold(III)
includes an FAD prosthetic group, an NADPH binding complexes developed in our laboratory behave as eective
site and an active site containing a redox-active selenol inhibitors of the cytosolic form of the selenoenzyme thiore-
group. Electrons are transferred from NADPH via FAD doxin reductase (TrxR) (see Table 2), accordingly, these
to the active-site selenol of TrxR, which then reduces the compounds were also found to perturb greatly the mito-
substrate [31]. TrxRs are named for their ability to reduce chondrial functions [38].
oxidised thioredoxins (Trxs), a group of small This view is now reinforced by a recent paper by Powis
(10 12 kDa) ubiquitous redox-active peptides which have et al. reporting the inhibitory properties of a series of
a conserved -Trp-Cys-Gly-Pro-Cys-Lys- catalytic site that Au(III) complexes (Fig. 6), against human thioredoxin
undergoes reversible oxidation/reduction of the two Cys reductase 1 (TrxR1), including some gold(III) DAMP com-
residues (see Fig. 5). pounds [39]. Table 2 contains a compilation of representa-
The crystal structure of thioredoxin reductase has been tive literature data for the inhibition of TrxR by various
recently solved [32]. Notably, these structural studies evi- gold compounds.
denced the presence of a selenocysteine group at its active Thus, according to the data described above, it is
site that, in the reduced form, displays a high reactivity reasonable to propose that direct antimitochondrial
toward soft metal ions. There is today sucient evidence eects are the determinant for the large proapoptotic
that this selenol group is the primary anchoring site for a and cytotoxic eects produced by anticancer gold(III)
vast array of metals and metal complexes that are known compounds.
to inhibit thioredoxin reductase [33,34]. Nonetheless, other recent studies identied the thiol-
Thus, one can propose quite reasonably that gold(III) dependent cathepsin enzymes as possible, alternative tar-
compounds exert their cytotoxic eects by causing direct gets for gold-based anticancer agents [40]. These lysosomal
mitochondrial damage through selective modication of enzymes are mainly cysteine proteases responsible for
the selenol active site in thioredoxin reductase as previously extracellular matrix degradation, bone resorption and joint
suggested for auranon and some related gold(I) com- destruction. Their marked inhibition arising from coordi-
pounds [3537]. As a matter of fact, in a recent study con- nation of gold to the active-site cysteine has been reported
ducted in collaboration with the group of Bindoli and for cathespin L and B [41,42].
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 571

1.2

1.1

1.0

0.9

0.8

0.7

A 0.6 b
0.5

0.4

0.3

0.2
a
0.1

0.0
200 250 300 350 400 450 500 550 600 650 700

nm

0.50

0.45

0.40

0.35 b

0.30

A 0.25

0.20

0.15

0.10

0.05
a
0
200 250 300 350 400 450 500 550 600 650 70 0

nm

Fig. 7. Time dependent spectral proles of Auoxo1cyt c (A) and Auoxo1ubiquitin (B) adducts. Spectra correspond to proteins 105 M before (a) and
after the addition (b) of Auoxo1 in a 1:1 ratio. The further evolution of the various systems over time is reported up to 24 h incubation at 37 C. The buer
was 10 mM phosphate, 20 mM NaCl, pH 7.4.

Very recently a paper on the ecient inhibition of feature for gold compounds. The focus of current research is
cathepsin K and S by two gold(I) derivatives, auranon now on the search of the most important protein targets for
and gold thiomalate, has been published [43]. In this paper gold(III) compounds.
the authors present an X-ray analysis of the gold thioma-
late/cathepsin K complex, showing the selective binding 3. Modelling the interactions of gold(III) compounds with
of gold to the active-site cysteine residue of the protease. protein targets: ESI MS and spectroscopic studies of the
On the ground of the above arguments and of the large reaction of two representative gold(III) complexes with
structural variety of the tested compounds, it can be pro- cytochrome c and ubiquitin
posed that gold(III) complexes produce their biological
eect through a number of distinct biochemical mechanisms In recent years, ESI MS has emerged as an extremely
ranging from classical DNA damage to oxidative membrane powerful tool to monitor, at the molecular level, the forma-
damage, from proteasome inhibition to mitochondrial dam- tion of protein adducts of classical anticancer platinum(II)
age. However, the latter mechanism, i.e. mitochondrial drugs, and to identify the precise nature of the metallic
damage induced through inhibition of thioredoxin reduc- fragments coordinated to protein side chains in these
tase, seems to be a rather peculiar and recurrent mechanistic adducts [4447]. However, only very few ESI MS studies
572 A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575

953.067
100 885.333

95

90

85

80
826.800
75

70 1032.133

65
Relative Abundance

60

55

50 774.867

45
1125.600
40

35

30
1237.533
25
729.400
1374.467
20

15 1149.733
1545.733
1265.133
10 1274.267
689.200 1409.200
1554.800
5
651.200 1604.333 1772.333
403.133 586.000
0
400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800
m/z

953.467
100
885.133
95
1032.400
90

85

80

75

70 1125.867
825.667
65
Relative Abundance

60

55
1238.133
1374.733
50

45

40 1143.867

35
773.800
30 1261.400

25
1390.867 1545.800
1275.733
20 1400.333

15 1416.467
728.400 1423.933
10
1448.333
1564.000
5 716.933 1588.667
681.200 1664.800 1766.333
408.800 519.200
0
400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800
m/z

Fig. 8. Multicharged mass spectra of cyt c (A) and Auoxo1cyt c conjugate (B) after 24 h reaction at 37 C.

have dealt so far with protein adducts formed by non-plat- niques, to analyse the reactions of two selected gold(III)
inum anticancer drugs [4850]. compounds, namely compound 7 and its dinuclear ana-
We show here that ESI MS may be advantageously logue Auoxo1, with the model proteins ubiquitin and cyto-
employed, in association with other spectroscopic tech- chrome c. Independent information on these systems was
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 573

gained from ICP OES determinations. Overall, the results the free complexes revealed for Auoxo1 peaks at m/z
reported here give us the chance to draw a quite detailed 369.8 and 883.0 which were assigned to [Au(bipy)OH]+
description of the interactions that take place. (100%) and [Au2(bipy)2O2PF6] (14%). For compound 7
First of all, both 7 and Auoxo1 were reacted with either the main peak at m/z 387.4 was assigned to [Au(bi-
ubiquitin or cyt c under identical experimental conditions py)(OH)2]+ and a very weak one at m/z 499.4 to [Au(bi-
and the resulting samples monitored spectrophotometri- py)PF6]+ (3%). It is worth noting that mass spectra of
cally over 24 h at 37 C [21]. proteinmetallodrug samples immediately after mixing
Ubiquitin does not exhibit any absorption band above did not show any adduct formation. This observation indi-
280 nm so that the bands characteristic of the two Au(III) cated that gas phase reactions do not take place under the
compounds may be easily followed. In both cases, the pro- applied experimental conditions.
gressive decrease of the CT bands typical of these gold(III) After 24 h incubation, samples of Auoxo1 and 7 with cyt
complexes is observed, accompanied by the growth of c showed, beyond the signal of the native protein, a charac-
intense bands in the 270300 nm region, characteristic of teristic peak assigned to a 1:1 Aucyt c derivative. The mul-
the free ligand. However, the process is slow, at room tem- ticharged spectra of free cyt c and of its adduct with
perature, and reaches only 30% progression within 24 h. Auoxo1 are reported in Fig. 8. The ESI MS spectra,
On the other hand, horse heart cyt c, in its typical oxi- together with spectrophotometric data, strongly suggest
dised form, manifests intense bands in the visible, namely that upon reaction with the protein the organic ligand is
the Soret band at about 410 nm and broad Q bands lost whereas gold, most likely in the form of a Au+ ion,
between 500 and 550 nm. Nonetheless, the overlap with remains tightly attached to the protein. A similar behav-
the bands characteristic of the two tested gold(III) com- iour was previously reported by Sadler and coworkers in
plexes is not dramatic so that the spectral evolution of the case of the adducts of gold(I) triethylphosphine chlo-
the two distinct components of our system may be moni- ride with cyclophilin [51].
tored rather easily. The reaction of 7 and Auoxo1 with Conversely, ESI MS analysis of the incubation mixtures
cyt c is faster than in the case of ubiquitin. At 24 h the reac- of both complexes with Ub revealed no appreciable adduct
tion has progressed to about 70%. Spectral patterns similar formation even after 12 days of reaction.
to those of ubiquitin are observed showing a continuous In conclusion, the above results point out quite clearly
decrease of the bands of the gold(III) complex and the that independently of the nature of the gold(III) complexes,
growth of an UV band typical of the free ligand. Typical a similar reactivity with cyt c takes place, comprising a
spectral proles obtained for Auoxo1 are shown in Fig. 7. redox process with gold(III) reduction followed by release
Ultraltration experiments accompanied by ICP OES of the organic ligand and eventual coordination of a
determinations showed that a signicant amount of gold gold(I) ion to a protein side chain.
remains associated to cyt c (up to 60% after 24 h). In con- Both the in situ redox process and gold(I) coordination
trast, gold binding to ubiquitin is poor, being only around to a specic protein residue may be crucial events for direct
15% even after several days of incubation. and irreversible protein damage. Further studies are in pro-
Afterward, ESI MS studies were performed on the gress to identify the nature of the binding site for gold(I) in
adducts of 7 or Auoxo1 with either Ub or cyt c. For com- cyt c, good candidates being Met 65 and two accessible his-
parison purposes, all samples were prepared and analysed tidines (His 33 and His 26).
under the identical conditions.1 The proteins, alone, Recent advances in the cellular pharmacology of Au(III)
showed a charge distribution of +7 to +13 and +8 to compounds are leading to the identication of a few spe-
+18 for Ub and cyt c, respectively. ESI MS analyses of cic proteins as probable biomolecular targets. In this
frame it is important to develop appropriate physicochem-
ical methods to monitor reactions of gold(III) complexes
1
Auoxo1 and 7 were dissolved in DMSO and the solution was diluted with protein targets. Modelling work carried out in our
1:100 with H2O to obtain a nal concentration of DMSO of 1%. The laboratory on simple proteins, primarily relying on mass
complexes were incubated at molar ratios of 1:1 with ubiquitin and
cytochrome c in aqueous solution at 37 C yielding nal protein spectrometry and some other ancillary techniques, holds
concentrations of 250 lM. The samples were analysed immediately after much promise for a rapid and detailed characterisation of
start of the incubation, 3 h, 24 h, 48 h, 6 days and 12 days. Before ESI IT goldprotein adducts. Some representative examples have
MS analysis, the incubation solutions were diluted (99:1) with been reported concerning ubiquitin and cytochrome c. Fur-
ACN:H2O:formic acid (30:70:1) and analysed immediately after mixing. ther eorts are needed to extend the investigation to the
Analyses were performed on a Thermo Finnigan LCQ Deca XP Plus
quadrupole ion-trap instrument in positive ion mode. The capillary protein targets that were mentioned in the previous section,
temperature was set at 180 C and the source voltage to 1.50 kV, with a for which, up to now, ESI MS has been scarcely used for
mass range from 300 to 2000. The acquisition was done with Tune Plus 1.3 the characterisation of their interactions with metallodrugs.
SR1 program (Thermo Finnigan) and the deconvolution of data were
performed with Bioworks Browser 3.0 (Thermo Finnigan) on a Windows
4. Concluding remarks
2000 Prof. SR4 PC system using Biomass Calculation and Deconvolution
software. The molecular weight was deconvoluted from the multiply
charged protein and proteinAu adduct peaks with charges ranging from The renewed interest toward gold(III) compounds as
+7 to +13 and +8 to +18 for ubiquitin and cytochrome c, respectively. potential anti-tumour agents, that started at the beginning
574 A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575

of 1990s, has resulted, until now, in the identication of GSH glutathione


several gold(III) and organogold(III) compounds charac- ICP OES inductively coupled plasma optical emission
terised by a signicant structural variety and by encourag- spectroscopy
ing in vitro pharmacological properties. Notably, these LMCT ligand to metal charge transfer
gold(III) compounds constitute today an interesting family phen phenathroline
of new cytotoxic agents that undoubtedly deserve more py pyridine
extensive pharmacological testing and a careful analysis terpy terpyridine
of their mechanism of action. We have reported above that TPP meso-tetraarylporphyrin
these structural diverse gold(III) complexes exert outstand- TrxR thioredoxin reductase
ing antiproliferative eects when tested in vitro against var- TrxR1 human cytosolic thioredoxin reductase
ious tumour cell lines. The observed biological eects are TrxR2 human mitochondrial thioredoxin reductase
most likely the consequence of innovative and distinct Trxs thioredoxins
molecular mechanisms in dependence of the peculiar coor- Ub ubiquitin
dination and redox chemistry of the gold(III) center and of
the nature of its ligands. DNA seems not to be a primary Acknowledgements
target for most novel cytotoxic gold(III) compounds.
In contrast, the important reactivity detected toward The consortium C.I.R.C.M.S.B and Ente Cassa di
model proteins favours the idea that the cytotoxic Risparmio di Firenze are gratefully acknowledged for
eects of gold(III) complexes are primarily mediated by nancial support. Dr. Angela Casini is grateful to AIRC
interactions with proteins targets. For instance, relevant for providing her with a two years fellowship.
antimitochondrial eects, probably arising from direct
inhibition of thioredoxin reductase were highlighted, that
activate the apoptotic cascade after triggering mitochon- References
drial cyt c release. Additionally, various Au(III) complexes
[1] B. Rosenberg, L. VanCamp, J.E. Trosko, V.H. Mansour, Nature 222
were reported to inhibit strongly the disease-specic, thiol- (1969) 385386.
containing, cysteine-protease cathepsins. [2] B. Lippert, Cisplatin: Chemistry and Biochemistry of a Leading
We have shown here that the reactivity of gold(III) com- Anticancer Drug, John Wiley & Sons, Inc., New York, 1999.
pounds with proteins may be accurately monitored, at the [3] E. Wong, C.M. Giandomenico, Chem. Rev. 99 (1999) 24512466.
[4] E.R. Tiekink, Crit. Rev. Oncol. Hematol. 42 (2002) 225248.
molecular level, through physicochemical studies of appro-
[5] R.V. Parish, B.P. Howe, J.P. Wright, J. Mack, R.G. Pritchard, R.G.
priate model systems. In this respect, ESI MS has turned Buckley, A.M. Elsome, S.P. Fricker, Inorg. Chem. 35 (1996) 1659
out to be a very powerful method to follow formation of 1666.
protein adducts and identify the nature of protein bound [6] R.G. Buckley, A.M. Elsome, S.P. Fricker, G.R. Henderson, B.R.
metal fragments. In the reported examples, concomitant Theobald, R.V. Parish, B.P. Howe, L.R. Kelland, J. Med. Chem. 39
redox and metal binding processes seem to be crucial in (1996) 52085214.
[7] S. Carotti, A. Guerri, T. Mazzei, L. Messori, E. Mini, P. Orioli,
producing specic protein damage as documented by the Inorg. Chim. Acta 281 (1998) 9094.
case of cyt c. However, in this eld, the development of [8] L. Messori, F. Abbate, G. Marcon, P. Orioli, M. Fontani, E. Mini, T.
proteomic or metalloproteomic techniques, as well as Mazzei, S. Carotti, T. OConnell, P. Zanello, J. Med. Chem. 43 (2000)
other combined biochemical and genetic methods, will be 35413548.
fundamental to provide us with powerful tools for the dis- [9] F. Abbate, P. Orioli, B. Bruni, G. Marcon, L. Messori, Inorg. Chim.
Acta 311 (2000) 15.
covery of new proteins or pathways involved in the cellular [10] G. Marcon, S. Carotti, M. Coronnello, L. Messori, E. Mini, P. Orioli,
response to gold-based drugs. Overall, in view of their T. Mazzei, M.A. Cinellu, G. Minghetti, J. Med. Chem. 45 (2002)
peculiar chemical and biological properties, it seems to us 16721677.
that these novel cytotoxic gold(III) complexes have the [11] M. Coronnello, E. Mini, B. Caciagli, M.A. Cinellu, A. Bindoli, C.
potential of being further developed and exploited as Gabbiani, L. Messori, J. Med. Chem. 48 (2005) 67616765.
[12] A. Vaccini, Ph.D. thesis 2002, University of Florence.
experimental anticancer drugs. [13] L. Messori, P. Orioli, C. Tempi, G. Marcon, Biochem. Biophys. Res.
Commun. 281 (2001) 352360.
5. Abbreviations [14] L. Ronconi, L. Giovagnini, C. Marzano, F. Bettio, R. Graziani, G.
Pilloni, D. Fregona, Inorg. Chem. 44 (2005) 18671881.
bipy bipyridyl [15] L. Ronconi, C. Marzano, P. Zanello, M. Corsini, G. Miolo, C.
Macca, A. Trevisan, D. Fregona, J. Med. Chem. 49 (2006) 16481657.
bipyc 6-(1,1-dimethylbenzyl)-2,20 -bipyridine [16] D. Aldinucci, D. Lorenzon, L. Stefani, L. Giovagnini, A. Colombatti,
cisplatin cis-diammine-dichloro platinum(II) D. Fregona, Anticanc. Drugs 18 (2007) 323332.
cyt c horse heart cytochrome c [17] V. Milacic, D. Chen, L. Ronconi, K.R. Landis-Piwowar, D. Fregona,
CT charge transfer Q.P. Dou, Cancer Res. 66 (2006) 1047810486.
[18] C.M. Che, R.W.-Y. Sun, W.-Y. Yu, C.-B. Ko, N. Zhu, H. Sun,
DAMP 2-[(dimethylamino)methylphenyl]
Chem. Commun. 14 (2003) 17181719.
dien diethylendiamine [19] N. Farrell, Met. Ions Biol. Syst. 42 (2004) 251296.
en ethylendiamine [20] A. Bergamo, G. Stocco, B. Gava, M. Cocchietto, E. Alessio, B. Serli,
ESI MS electrospray ionization mass spectrometry E. Iengo, G. Sava, J. Pharmacol. Exp. Ther. 305 (2003) 725732.
A. Casini et al. / Journal of Inorganic Biochemistry 102 (2008) 564575 575

[21] A. Casini, M.A. Cinellu, G. Minghetti, C. Gabbiani, M. Coronnello, [39] L. Engman, M. McNaughton, M. Gajewska, S. Kumar, A. Birming-
E. Mini, L. Messori, J. Med. Chem. 49 (2006) 55245531. ham, G. Powis, Anticanc. Drugs 17 (2006) 539544.
[22] L. Messori, G. Marcon, P. Orioli, M.A. Cinellu, G. Minghetti, Eur. J. [40] V. Turk, B. Turk, D. Turk, EMBO J. 20 (2001) 46294633.
Biochem. 270 (2003) 46554661. [41] A. Chircorian, A.M. Barrios, Bioorg. Med. Chem. Lett. 14 (2004)
[23] Y. Wang, Q.Y. He, C.M. Che, J.F. Chiu, Proteomics 6 (2006) 131 51135116.
142. [42] S.P. Fricker, R. Skerjl, B.R. Cameron, R. Mosi, Y. Zhu, Recent
[24] L.H. Hurley, Nat. Rev. Cancer 2 (2002) 188200. developments in gold drugs in: Contribution to the Gold 2003
[25] D. Wang, S.J. Lippard, Nat. Rev. Drug. Discov. 4 (2005) 307320. Conference: New Industrial Applications for Gold.
[26] M. Coronnello, G. Marcon, S. Carotti, B. Caciagli, E. Mini, T. [43] E. Weidauer, Y. Yasuda, B.K. Biswal, M. Cherny, M.N. James, D.
Mazzei, P. Orioli, L. Messori, Oncol. Res. 12 (2000) 361370. Bromme, J. Biol. Chem. 388 (2007) 331336.
[27] M.T. Coer, C.F. Shaw 3rd, A.L. Hormann, C.K. Mirabelli, S.T. [44] I. Khalaila, C.S. Allardyce, C. Verma, P.J. Dyson, ChemBioChem 6
Crooke, J. Inorg. Biochem. 30 (1987) 177187. (2005) 17881795.
[28] G.F. Rush, P.F. Smith, G.D. Hoke, D.W. Alberts, R.M. Snyder, [45] T. Peleg-Shulman, Y. Najajreh, D. Gibson, J. Inorg. Biochem. 91
C.K. Mirabelli, Toxicol. Appl. Pharmacol. 90 (1987) 391400. (2002) 306311.
[29] C.H. Williams Jr., Eur. J. Biochem. 267 (2000) 6101. [46] A. Casini, G. Mastrobuoni, C. Temperini, C. Gabbiani, S. Francese,
[30] E.S.J. Arner, A. Holmgren, Eur. J. Biochem. 267 (2000) 61026109. G. Moneti, C.T. Supuran, A. Scozzafava, L. Messori, Chem.
[31] H. Nakamura, Antioxid. Redox. Signal. 7 (2005) 823828. Commun. (2007) 156158.
[32] K. Fritz-Wolf, S. Urig, K. Becker, J. Mol. Biol. 370 (2007) 116127. [47] C.G. Hartinger, W.H. Ang, A. Casini, L. Messori, B.K. Keppler, P.J.
[33] A.B. Witte, K. Anestal, E. Jerremalm, H. Ehrsson, E.S. Arner, Free Dyson, JAAS 22 (2007) 960967.
Radic. Biol. Med. 39 (2005) 696703. [48] A.R. Timerbaev, C.G. Hartinger, S.S. Aleksenko, B.K. Keppler,
[34] P.J. Barnard, S.J. Berners-Price, Coord. Chem. Rev. 251 (2007) 1889 Chem. Rev. 106 (2006) 22242248.
1902. [49] A. Casini, G. Mastrobuoni, W.H. Ang, C. Gabbiani, G. Pieraccini,
[35] M.P. Rigobello, G. Scutari, A. Folda, A. Bindoli, Biochem. G. Moneti, P.J. Dyson, L. Messori, Chem. Med. Chem. 2 (2007) 631
Pharmacol. 67 (2004) 689696. 635.
[36] Y. Omata, M. Folan, M. Shaw, R.L. Messer, P.E. Lockwood, D. [50] A. Casini, G. Mastrobuoni, M. Terenghi, C. Gabbiani, E. Monzani,
Hobbs, S. Bouillaguet, H. Sano, J.B. Lewis, J.C. Wataha, Toxicol. In G. Moneti, L. Casella, L. Messori, J. Biol. Inorg. Chem. 12 (2007)
Vitro 20 (2006) 882890. 11071117.
[37] O. Rackham, S.J. Nichols, P.J. Leedman, S.J. Berners-Price, A. [51] J. Zou, P. Taylor, J. Dornan, S.P. Robinson, M.D. Walkinshaw, P.J.
Filipovska, Biochem. Pharmacol. 74 (2007) 9921002. Sadler, Angew. Chem. Int. Ed. Engl. 39 (2000) 29312934.
[38] M.P. Rigobello, L. Messori, G. Marcon, M. Bragadin, A. Folda, G.
Scutari, A. Bindoli, J. Inorg. Biochem. 98 (2004) 16341641.

S-ar putea să vă placă și