Вы находитесь на странице: 1из 11

JOURNAL OF

Inorganic
Biochemistry
Journal of Inorganic Biochemistry 98 (2004) 402–412
www.elsevier.com/locate/jinorgbio

The hydrolysis of the anti-cancer ruthenium complex NAMI-A


affects its DNA binding and antimetastatic activity:
an NMR evaluation
Marina Bacac a,b,c, Anna C.G. Hotze a, Karlijn van der Schilden a, Jaap G. Haasnoot a,
Sabrina Pacor b, Enzo Alessio d, Gianni Sava b,c, Jan Reedijk a,*
a
Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
b
Department of Biomedical Sciences, University of Trieste, via L. Giorgieri 7, 34127 Trieste, Italy
c
Callerio Foundation Onlus, via A. Fleming 22-31, 34127 Trieste, Italy
d
Department of Chemical Sciences, University of Trieste, via L. Giorgieri 7, 34127 Trieste, Italy
Received 15 October 2003; received in revised form 19 November 2003; accepted 2 December 2003

Abstract

The coordination of the antimetastatic agent NAMI-A, [H2 im][trans-RuCl4 (dmso-S)(Him)], (Him ¼ imidazole; dmso ¼ di-
methyl sulfoxide), to the DNA model base 9-methyladenine (9-MeAde) was investigated in water. NMR spectroscopy was first
applied for the study of the molecular stability and hydrolysis of NAMI-A in aqueous solution over a range of pH (3.0–7.4) and
chloride ion concentrations (0–1 M) at 37.0 °C. In physiological conditions (phosphate buffer, pH 7.4) NAMI-A disappears from the
solution in 15 min due to chloride and dmso hydrolysis, leading to uncharacterised poly-oxo Ru species. Conversely, at lower pH
(3.0–6.0) and in water (pH  5.5), only a partial dmso hydrolysis occurs, slowly forming the [trans-RuCl4 (H2 O)(Him)] complex.
This latter species coordinates to 9-MeAde (via the N7 of 9-MeAde), forming the [trans-RuCl4 (9-MeAde)(Him)] complex. NAMI-
A and [trans-RuCl4 (H2 O)(Him)] give comparable intracellular ruthenium concentrations and accumulate in KB cells (human
mouth carcinoma) and accumulate these at the G2 /M phase, while poly-oxo Ru species do not, and their cell uptake is reduced to
50%. On the contrary, G2 /M arrest and protein content in the murine metastatic cell line metGM, are not influenced by NAMI-A
hydrolysis. Hydrolysed NAMI-A species apparently are easier taken up by the metGM cells, showing intracellular ruthenium
concentrations one order of magnitude greater than those of intact NAMI-A. Therefore, it is proposed that the selective anti-
metastatic activity of NAMI-A during in vivo experiments can be attributed to its hydrolysed species.
Ó 2003 Elsevier Inc. All rights reserved.

Keywords: Ruthenium; NAMI-A; Hydrolysis; Coordination to 9-methyladenine; Cell cycle arrest at G2 /M phase

1. Introduction antimetastatic activity of ruthenium–dmso complexes


was observed in the early 1980s with cis and trans-
The success of cisplatin and related platinum com- [RuCl2 (dmso)4 ], (dmso ¼ dimethyl sulfoxide), but the
plexes as anticancer agents stimulated the search for most relevant improvement in the ruthenium anticancer
other active transition-metal complexes in cancer che- chemistry was reached with NAMI, Na[trans-
motherapy [1,2]. Among all ruthenium-based anticancer RuCl4 (dmso-S)(Him)] [4], and NAMI-A, [H2 Im][trans-
agents, ruthenium–dmso complexes are believed to have RuCl4 (dmso-S)(Him)] [5] (Fig. 1) the last one being the
great potential because of their selectivity for solid tu- only ruthenium complex that successfully finished a
mor metastases and low host toxicity [3]. The selective Phase I clinical trial. Differently from cisplatin and
platinum related compounds, biological testing of ru-
thenium–dmso compounds pointed out that DNA is not
*
Corresponding author. Tel.: +31-71-5274459; fax: +31-71-5274671. the only target responsible for their antimetastatic ac-
E-mail address: reedijk@chem.leidenuniv.nl (J. Reedijk). tivity [5–7]. NAMI-A selectively reduces the formation

0162-0134/$ - see front matter Ó 2003 Elsevier Inc. All rights reserved.
doi:10.1016/j.jinorgbio.2003.12.003
M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412 403

- H+ activity comparable to that of the parent compound, or


N3
SOMe2 whether hydrolysis of ligands and subsequently differ-
4 2 H H
Cl ences in charge of the appearing species reduce the
Cl N
Ru
NH ability to cross cell membranes and to target biological
5 1 6
Cl Cl 5 N7 molecules.
1N Therefore, the aim of the present research is to in-
N3 8
vestigate the interaction of NAMI-A with DNA (using
4 2
2 N 4 N9 9-methyladenine as model base Fig. 1(right)) and the
3 speciation of NAMI-A in aqueous solution in a wide
NH CH3
5 1 range of pH and chloride ion concentrations by means
of NMR spectroscopy. Second, the biological activity of
Fig. 1. Chemical structure and numbering scheme of NAMI-A,
[H2 im][trans-RuCl4 (dmso-S)(Him)] (left) and 9-methyladenine (9-Me- some hydrolysis species of NAMI-A was studied by
Ade) (right). evaluating parameters correlated with cell viability such
as cell cycle, protein synthesis and ruthenium uptake.

and growth of lung metastases of malignant tumors such


as MCa mammary carcinoma, Lewis lung carcinoma 2. Results
(LLC) and TS/A adenocarcinoma [8]. In vitro studies
excluded cisplatin-like cytotoxicity of NAMI-A for 2.1. Effects of pH on hydrolysis of NAMI-A: [trans-
primary tumor cells and showed an effect on their cell RuCl4 (dmso-S)(Him)] ! [mer-RuCl3 (dmso-S)(Him)-
proliferation, which might be a consequence of the (H2 O)]
transient arrest at G2 /M phase of cell cycle [5,8]. Besides
cell cycle arrest, NAMI-A treatment of metastatic cells Hydrolytic transformation of NAMI-A (5 mM) was
induced modifications of cell membrane morphology monitored by 1 H NMR spectroscopy at 37.0 °C in
towards a less aggressive phenotype [9], down-regulated phosphate buffer (0.05 M phosphate, 0.15 M NaCl, in
CD44 expression and reduced their invasion ability the range of pH from 3.0 to 7.4) and in water (D2 O,
(manuscript in preparation). pH  5.5). Fig. 2(a) shows time-dependent 1 H NMR
Despite extensive in vitro and in vivo studies, the spectra of the hydrolysis of NAMI-A in phosphate
exact mechanism of action of NAMI-A has not been buffer (pH 7.4). Immediately after dissolution of NAMI-
elucidated as yet and further investigations are needed A (Fig. 2(a), t0 ), six signals are identified: four broad
to identify its real biological target(s). Central interest is signals (due to ligands bound to paramagnetic Ru(III))
focused on two fronts: (i) the investigation of the spe- and two sharp signals (due to the H2 imþ counter ion),
ciation of NAMI-A and other ruthenium-based com- assigned according to already published data for NAMI
pounds in biological media such as body fluids, culture [4]. In fact, the NMR of NAMI and NAMI-A are al-
media (to identify the metabolites formed) and (ii) the most identical. The broad signal at ca. )15 ppm was
interaction of the ruthenium complexes with biological assigned to the dmso-S ligand of NAMI-A while signals
targets such as proteins and DNA [1,10]. In this view, at )3.5, )5.6 and )7.8 ppm were assigned to H5 and
NAMI (the corresponding sodium salt of NAMI-A) has H2;4 , respectively, of the Him ligand. The sharp reso-
already been investigated for the interaction with DNA. nances at 8.69 and 7.49 ppm (1:2 integration ratio),
These studies pointed out some interesting differences in correspond, respectively, to H2 and H4 ,5 of the imi-
comparison to cisplatin that might account for different dazolium counter ion (H2 imþ ). Acetone (1 lL/mL) was
biological activity between these two compounds [11]. In added to the buffer as a reference for the integration, as
particular, NAMI appeared to modify the double helix it gives a sharp signal at 2.06 ppm that does not overlap
of DNA in a different manner than cisplatin and to with the resonances of NAMI-A or its hydrolysis
coordinate to DNA considerably faster than cisplatin species.
[11]. The faster binding of NAMI to DNA was attrib- As a result of the hydrolysis of one chloride at pH
uted to its higher rate of hydrolysis in comparison to 7.4, the resonances of NAMI-A are replaced within few
cisplatin. In fact, NAMI is not inert in physiological minutes by other signals. In particular, the broad reso-
solution but easily undergoes hydrolytic transforma- nance at ca. )10 ppm was attributed to dmso-S of the
tions [4,12], known to be the first necessary steps for the first hydrolysis species of NAMI-A, [mer-RuCl3 (dmso-
interaction of metal-based drugs with biological targets S)(Him)(H2 O)], (NAMI-AH2 O ), while the resonance at
[13]. ca. 0.6 ppm was attributed to the H5 of the Him ligand
Although NAMI has been investigated for its inter- of the same species (Fig. 2(a), dotted arrows). The res-
action with DNA, NAMI-A has not been studied as yet. onances of H2 and H4 of Him in NAMI-AH2 O are most
Moreover, it is still unclear whether species derived from likely too broad to be detected, as previously reported
hydrolysis of NAMI-A still maintain a pharmacological [12].
404 M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412

(a)

acetone

free
dmso

Him H 5 NAMI-AH2O dmso NAMI-AH2O

t 45 min

t 15- 20 min
t 10 min

t0
ppm 5 0 -5 -10 -1 5
H 2, H 4,5
H 2im+ Him H 5 Him H 2,4 dmso NAMI-A

(b) (c)
pH 7.4
3 3 pH 3.0
NAMI-A NAMI-A
NAMI -AH2O
LN peak area (dmso)

2 dm so 2

1 1 dm so

0 0
25 50 20 40 60 80
-1 -1

-2 Time (min) -2 Time (min)

NAMI-AH2O
[mer-RuCl3(dmso-S)(Him)(H2O)] SOMe2 OH2
SOMe2 Cl Cl Cl Cl
Cl Ru Ru
OH2
Ru Cl Cl Cl Cl
SOMe2 Cl Cl OH2 Him Him
Cl Cl Cl
Him OH2 NAMI-A [trans-RuCl4(Him)(H2O)]-
Ru 45 min. Ru
15 min. OH2
Cl Cl Cl Cl dmso released after 60 min
Cl Cl pH 6.0 40%
Him Him
Ru pH 5.0 11%
NAMI-A Cl Cl pH 3.0 4%
D2O (pH 5.5) 9%
Him
POLYNUCLEAR
[trans-RuCl4(Him)(H2O)]- Species

Fig. 2. (a) 300 MHz 1 H NMR spectra recorded during the hydrolysis of NAMI-A (5 mM) in phosphate buffer (0.05 M phosphate, 0.15 M NaCl), pH
7.4, 37.0 °C. (b, c) Transformation pathways of anionic NAMI-A and hydrolysis species formed in phosphate buffer (b, left) pH 7.4 and (c, right)
pH < 6.0, at 37.0 °C, monitored through integration of the dmso resonances at )15 (NAMI-A), )10 (NAMI-AH2 O ), and 2.7 ppm (dmso released) in
the 1 H NMR spectra. Scheme below the graph (left): hydrolysis species formed at pH 7.4, graph (right) species formed at pH < 6.0 and % dmso
released in function of pH. Note. For graphical reasons only the cis isomer of the [RuCl3 (H2 O)2 (Him)] species is designated in the scheme left; we
have no experimental evidence of the geometry of this species, either cis or trans or a mixture of the two isomers.

Integration of the dmso-S resonances at )15 and )10 although it is not stable in time. In fact, it continues to
ppm was used to monitor the disappearance of NAMI- hydrolyse, with a probable dissociation of at least one
A and the formation and subsequent disappearance of further chloride, until it disappears completely from the
NAMI-AH2 O (Fig. 2(b)). The signals for NAMI-A and solution after ca. 45 min (Fig. 2(b) and the scheme below
released dmso follow smooth curves; the curves for the figure). No new resonances attributable to hydrolytic
NAMI-AH2 O are less accurate and not easy to fit. Nev- species derived from NAMI-AH2 O could be detected,
ertheless, it was possible to determine that NAMI-AH2 O very likely because of the formation of polynuclear
is the predominant species in solution after 15–20 min, species (we have no experimental evidence of the ge-
M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412 405

ometry of this species, either cis or trans or a mixture of 3

the two isomers). Gradual darkening of the solution in

LN peak area (dmso


this condition, from light orange to dark brown, indi- 2

cated formation of poly-oxo Ru species as the result of

NAMI-A)
advanced hydrolysis of NAMI-A, as previously reported 1

for NAMI [12]. The gradual increase in intensity of the


0
sharp resonance at 2.72 ppm (free dmso) indicated that 3 6 9 12 15 18 21 24 27
slow hydrolysis of dmso from NAMI-A (and from its
-1
hydrolysis products) occurs simultaneously to that of 0M
chloride ligands (Fig. 2(b)). However, integration of the (a) -2 1M
signals of the free ligands indicated that dmso hydrolysis Time (min) 0.5 M
is only partial and that Him hydrolysis is negligible.
3
The situation at pH < 6 is different. No detectable
chloride hydrolysis occurs and only dmso is partially

LN peak area (dmso


2
released, slowly forming the [trans-RuCl4 (H2 O)(Him)]

NAMI-AH2O )
complex (Fig. 2(c) and the scheme below). Integration of 1

the resonance signal of free dmso as a function of pH


0
values pointed out that the rate of dmso hydrolysis de- 10 20 30 40 50 60 1M
creases by lowering the pH (scheme below the Fig. 2(c)). -1
This decrease contributes to the better stability of 0.5 M
-2
NAMI-A in solution and elongates its half-life to several
0M
hours (in comparison to t1=2 of 20 min at pH 7.4). The (b) -3 Time (min)
behaviour of NAMI-A in water is similar to that ob-
tained at lower pH since the dissolution of the compound Fig. 3. (a, b) Influence of chloride ion concentration on (top) the sta-
bility of 5 mM NAMI-A obtained by the integration of dmso-S res-
(5 mM) results in pH 5.5 that decreases in time (to 4)
onance at )15 ppm, and on (lower graph) the formation and
due to the acidity of the coordinated water molecule. As disappearance of the [mer-RuCl3 (dmso-S)(Him)(H2 O)] species ob-
for solutions with pH < 6, the two predominant species of tained by integration of dmso-S resonance at )10 ppm. Reaction was
NAMI-A in water (besides the cation) are the original followed in phosphate buffer (0.05 M phosphate), pH 7.4, 37.0 °C.
molecule [trans-RuCl4 (dmso-S)(Him)] and [trans- Range of [Cl] : 0–1 M.
RuCl4 (H2 O)(Him)] during several hours.
spectroscopy in situ, at 37.0 °C, in water. As reported in
2.2. Effects of Cl concentration on NAMI-A hydrolysis: the hydrolytic study (see above) only the dmso ligand
[trans-RuCl4 (dmso-S)(Him)] ! [mer-RuCl3 (dmso-S) hydrolyses slowly from NAMI-A in water and forms the
(Him)(H2 O)] [trans-RuCl4 (H2 O)(Him)] complex. Therefore, the po-
sition occupied by the labile water molecule is the only
The effects of increasing chloride ion concentration (0– available coordination site for a DNA model base under
1 M) on the hydrolysis of NAMI-A were investigated in these conditions. Fig. 4 shows some 1 H NMR spectra of
phosphate buffer (pH 7.4, 37.0 °C). Increase of [Cl ] in the reaction (during 7 h) between NAMI-A and 9-Me-
solution slows down the first chloride hydrolysis from Ade (1:4). At t0 only the resonances of the uncoordi-
NAMI-A (Fig. 3(a)) and also dmso hydrolysis (data not nated 9-MeAde at 8.18, 8.04 and 3.78 ppm (assigned by
shown), influencing the formation of NAMI-AH2 O . It NOE difference spectroscopy to H2 , H8 and CH3 , re-
also interferes with the stability of NAMI-AH2 O in solu- spectively), and of unreacted NAMI-A were identified.
tion since it slows down chloride hydrolysis from this The paramagnetic properties of Ru(III) cause severe
species: when no NaCl is added to the buffer, NAMI- shifting and broadening of the resonances of ligands
AH2 O reaches maximum values already after 15 min and coordinated to the metal ion (as seen for dmso and Him
disappears rather fast (after 40 min, Fig. 3(b)). At [NaCl] ligands of NAMI-A). Therefore, it is expected that,
1 M, NAMI-AH2 O reaches its maximum values only after upon coordination, the resonances of 9-MeAde are
30 min, remains present in solution for longer time, and shifted and broadened with respect to the free ligand.
finally disappears gradually after 60 min (Fig. 3(b)). Thus, the two new and relatively broad signals of equal
intensity that appear at 13.16 and 1.05 ppm after 1 h of
2.3. Coordination to 9-methyladenine: [trans-RuCl4 (dm- reaction were attributed to coordinated 9-MeAde in
so-S)(Him)] ! [trans-RuCl4 (H2 O)(Him)] + 9-Me- [trans-RuCl4 (9-MeAde)(Him)] .
Ade ! [trans-RuCl4 (9-MeAde)(Him)] The signals for the H2 and H8 protons of coordinated
9-methyladenine have been identified by T1 measure-
The coordination of NAMI-A to the DNA model ments using inversion recovery. Namely, nuclear relax-
base 9-methyladenine (9-MeAde) was studied by NMR ation is strongly affected by interaction with unpaired
406 M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412

16
14 H2

12

10

chemical shift (ppm)


8

2 H8

-2
1 0 2 4 6 8 10
Fig. 4. 300 MHz H NMR spectra recorded during the coordination of
9-methyladenine (20 mM) to NAMI-A (5 mM) in D2 O, 37.0 °C. (t0 ) pH
NAMI-A and 9-methyladenine immediately after addition; resonances
at 8.18, 8.04 and 3.78 ppm correspond to H2 , H8 and CH3 , respec- Fig. 5. pH titration of [trans-RuCl4 (9-MeAde)(Him)] .
tively, of free 9-methyladenine. (7 h) Broad and shifted signals at 13.16
and 1.05 ppm (arrows) are relative to H2 and H8 , respectively, of co-
ordinated 9-MeAde in [trans-RuCl4 (9-MeAde)(Him)] .
which is closest to the N1 protonation site. The resonance
at 1.05 ppm was therefore assigned to H8 of coordinated
electrons. Therefore, protons close to the Ru(III) centre 9-MeAde. This assignment is in agreement with the T1
have relatively short relaxation times. Hence, they can be values of the H2 and H8 , since the proton closest to the
distinguished from the protons of the free ligand which Ru(III) centre (which is the H8 ), displays the shortest T1
display large T1 values, i.e., in the order of hundreds of value in comparison to the more distant H2 (13 vs. 27 ms).
milliseconds. The new signals appearing at 13.16 and 1.05 Therefore, the coordination mode of 9-MeAde to
ppm exhibit T1 values of 27 and 13 ms, respectively, in- NAMI-A is different from the reported N6 coordination
dicating coordination of the ligand to Ru(III). A third observed in a structurally related complex, i.e. [trans-
signal at 5.15 ppm, which probably belongs to the CH3 RuCl3 (9-MeAde)(dmpt)2 ]  H2 O, (dmpt ¼ 5,7-dimeth-
group of 9-methyladenine, has been identified with the T1 yl[1,2,4]triazolo[1,5-a]pyrimidine) [18]. The bidentate
measurement. The signal is shifted almost under the N6–N7 coordination, found in [cis-Ru(bpy)2 (9-Me-
water signal but has been recognized by its short T1 value, Ade)](PF6 )2 and [a-Ru(azpy)2 (9-MeAde)](PF6 )2 (bpy ¼
i.e., 19 ms. However, it was impossible to identify the 2,20 -bipyridine; azpy ¼ 2-phenylazopyridine) [19] was
resonances of the Him ligand of the [trans-RuCl4 (9-Me- also excluded, since the [trans-RuCl4 (H2 O)(Him)]
Ade)Him] complex, which is reasonable considering complex has only one available coordination site.
that only ca. 10% of NAMI-A coordinates to 9-MeAde. No variations of chemical shifts were seen for H8 and
This is in agreement with the fact that also the Him res- H2 by increasing the pH to 7, while for pH > 7 the
onances in the [trans-RuCl4 (H2 O)Him] and [mer- complex started to decompose with gradual loss of
RuCl3 (dmso-S)(Him)(H2 O)] complexes could not be ‘‘paramagnetic’’ signals and darkening of the solution.
identified (with the exception of H5 of the Him in the ESI-MS (electrospray mass spectrometry) experiments,
[mer-RuCl3 (dmso-S)(Him)(H2 O)] complex) and might performed on the reaction solution with the aim of
overlap with those of NAMI-A. gaining further evidence on the nature of the product,
The assignment of the H2 and H8 resonances of the were unsuccessful and this technique proved to be in-
coordinated 9-MeAde in [trans-RuCl4 (9-Me- adequate for investigating these systems. The reaction
Ade)(Him)] was performed by monitoring their pH- between NAMI-A and 9-MeAde was also run adding
dependence. Free 9-MeAde exhibits N7 protonation at the DNA model base to an aged solution of the complex
pH < 2, and N1 protonation at pH  4 [14]. Protonation (3 days in 10 mM NaClO4 in D2 O at room tempera-
of N1 occurs at lower pH when a metal is bound at N7 ture), and in the presence of 10% dmso. 9-MeAde co-
[15–17]. The absence of N7 protonation for pH < 2 ordinated already after 30 min when the aged solution of
(no shifting of the resonances of [trans-RuCl4 (9-Me- NAMI-A was used, in which ca. 40% of the dmso ligand
Ade)Him] ), and the protonation of N1 of bound 9- was hydrolysed. On the contrary, 9-MeAde did not co-
MeAde at pH  3 (Fig. 5), clearly indicate that 9-MeAde ordinate to NAMI-A when 10% of dmso was added to
coordinates to ruthenium through its N7 atom and the solution, as dmso hydrolysis from NAMI-A was
consequently exclude N6 coordination. The signal at inhibited in agreement with previous studies [20]. These
13.16 ppm shows a larger pH-dependent shifting (Fig. 5) observations unambiguously proved that the dmso site
and was thus assigned to H2 of coordinated 9-MeAde, in NAMI-A becomes the coordination site of 9-MeAde.
M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412 407

9-MeAde did not coordinate to NAMI-A in phos- hydrolysis species. Data regarding the effects of 0.1 mM
phate buffer at pH 7.4, where only its hydrolysis species NAMI-A on cell proliferation of KB cell line have al-
and free 9-MeAde were found after overnight reaction ready been reported elsewhere and indicated a statisti-
together with black colour of solution due to poly-oxo cally significant reduction of cell proliferation in
Ru species (data not shown). Besides the hydrolytic in- comparison to controls [8,22]. The same study pointed
stability of NAMI-A at pH 7.4, the absence of detect- out a transient cell cycle arrest of KB cells at G2 /M phase.
able coordination of the model base to ruthenium in Therefore, we used this cell line for further investigations
phosphate buffer is attributed to competition with OH concerning the effects of NAMI-A and its hydrolysis
(high affinity ligand for Ru(III)) [21]. species on the cell cycle distribution of tumor cells.

2.4. Effects of NAMI-A hydrolysis on cell proliferation 2.5. Effects of NAMI-A hydrolysis on cell cycle distribu-
tion of KB cells
The effects of NAMI-A and of its hydrolysis species
on cell proliferation were evaluated on human ovarian The same species mentioned above were evaluated at
carcinoma cell line A2780 (normal) and A2780cisres the total Ru concentration of 0.1 mM. NAMI-A (1)
(resistant to cisplatin). Two different hydrolysis species increased KB cells at G2 /M phase by 64% in comparison
of NAMI-A were tested and compared to the original to controls (***p < 0:001, Table 1), similarly to NAMI-
molecule of NAMI-A; these were the poly-oxo Ru A (3) (***p < 0:001, Table 1). Only a weak G2 /M arrest
species, obtained by complete hydrolysis of NAMI-A in was observed with NAMI-A (2), (*p < 0:05, Table 1).
PBS, and a mixture of NAMI-A and [trans- Statistically significant differences were observed not
RuCl4 (H2 O)(Him)] (ca. 1.5:1), derived from hydrolysis only in comparison to controls but also within the tested
of NAMI-A in water (Section 5). Regarding this last species, and consisted in differences between NAMI-A
solution, it was impossible to further increase the (1), (3) and NAMI-A (2) and between NAMI-A (1) and
amount of [trans-RuCl4 (H2 O)(Him)] species, since NAMI-A (3) ($$$ p < 0:001, Table 1). The first one in-
longer hydrolysis of NAMI-A in water led to the for- dicates the inability of poly-oxo Ru species to affect cell
mation of poly-oxo Ru species. For easier description, cycle distribution of KB cells, the second one indicates
NAMI-A fresh is indicated as (1), poly-oxo Ru species the important role of dmso hydrolysis in favouring G2 /
as (2) and the mixture of NAMI-A and [trans- M arrest of same cells.
RuCl4 (H2 O)(Him)] as (3). Cell growth was determined In agreement with the found G2 /M arrest, NAMI-A
by MTT proliferation assay 24 h after treatment with 1 (1) and (3) caused a statistically significant increase of the
mM ruthenium (originally as NAMI-A). protein content in comparison to controls; the average
NAMI-A (1) and (3) show comparable reduction of fluorescence intensity (MFI) relative to the protein con-
cell proliferation of A2780 cells in comparison to con- tent of treated samples was found 206  2.1 for (1),
trols (***p < 0:001, Table 1), and both species are more 228  3.9 for (3) vs. 185  7.1 for control, p < 0:001,
active in comparison to NAMI-A (2) ($$$ p < 0:001, whereas NAMI-A (2) did not influence this parameter
Table 1). (Data are considered to be statistically signif- (MFI, 177  11.7 for (2) vs. 1857.1 for control, p > 0:05).
icant if the probability factor ‘‘p’’ is *p < 0:05, **p <
0:01 and ***p < 0:001.) 2.6. Effects of hydrolysis on cell uptake of ruthenium in
Effects on A2780cisres were comparable to those on KB cells
the A2780 cell line (data not shown) suggesting that the
mechanisms responsible for cisplatin resistance do not Total ruthenium content inside KB cells, determined
interfere with activity of NAMI-A nor with that of its by atomic absorption spectroscopy, indicated that

Table 1
Reduction of cell proliferation (MTT test, A2780 cells), G2 /M arrest and intracellular ruthenium content (KB and metGM cells), 24 h after treatment
with NAMI-A (0.1–1 mM)
Sample 1 mM 0.1 mM
MTT OD (590 nm) % G2 /M phase lg Ru/106 cells
A2780 KB metGM KB metGM
Control 2.57  0.06 13.4  1.00 12.5  1.20 – –
(1) 1.79  0.07***;$$$ 20.8  1.20***;$$$ 15.7  0.30* 0.02  0.002 0.03  0.006
(2) 2.29  0.16* 14.8  1.0 17.6  1.00**;^^ 0.01  0.005°° 0.34  0.027°°°
(3) 1.72  0.15***;$$$ 27.0  1.0***;$$$ 14.6  3.56 0.02  0.006 0.07  0.029
(1) NAMI-A immediately dissolved, (2) poly-oxo Ru species, (3) mixture of NAMI-A and [trans-RuCl4 (H2 O)(Him)] (1.5:1) (solutions were
prepared according to Section 5). Statistical analysis, Student–Newman–Keuls test, *p < 0:05; **p < 0:01; ***p < 0:001 vs. controls; $$$ p < 0:001
(1), (3) vs. (2); ^^ p < 0:01 (2) vs. (1); °°p < 0:01; °°°p < 0:001 (2) vs. (1) and (3).
408 M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412

NAMI-A (1) and (3) reached comparable values of in- to elucidate the stability and hydrolytic transformations
tracellular ruthenium concentration (ca. 0.02 lg Ru/ of NAMI-A in aqueous solution, since a good under-
1  106 cells, Table 1). The intracellular ruthenium standing of these processes is the crucial step to identify
concentration of NAMI-A (2) was reduced by about the active species and to unravel their mode of interac-
50% (Table 1) indicating decreased uptake of poly-oxo tion with DNA and other biological targets [13].
Ru species, derived from advanced hydrolysis of Hydrolysis of NAMI-A was studied by NMR spec-
NAMI-A. troscopy in a wide range of pH (3.0–7.4) and of chloride-
ion concentrations (0–1 M) to reproduce the conditions
2.7. Effects of hydrolysis on cell cycle distribution of that NAMI-A might encounter in vivo. Blood plasma
metGM cells presents a pH of 7.4, while the hypoxic environment of
solid tumors offers pH values between 6.0 and 7.0. Some
Similarly to KB cells, metGM cells were treated with cell compartments, like endosomes that handle trans-
solutions (1), (2) and (3), at the total Ru concentration ferrin, possess even lower pH values, i.e. 5.5, and it is
of 0.1 mM. For a better evaluation, these cells were also possible to reach extreme conditions like pH 2.0 in
treated for three consecutive times with the above the stomach, or pH 8.0 in urine. Studies were performed
mentioned solutions of NAMI-A and analysed for cell at 37.0 °C, i.e., a temperature at which hydrolysis and
cycle distribution, protein content and intracellular ru- binding to target biomolecules occur in host organisms.
thenium concentration after each treatment. In physiological conditions (phosphate buffer, pH
Differently from KB cells, NAMI-A (1), (2) and (3) 7.4), NAMI-A undergoes both chloride and dmso hy-
gave comparable effects on cell cycle arrest of metGM drolysis while forming a mixture of hydrolysis species
cells, 24 h after the first, second and third treatments. responsible for the disappearance of the parent com-
The first treatment with NAMI-A (1), (2) and (3) re- pound from the solution in about 15 min. Increased
sulted in comparable S phase arrest (data not shown) chloride-ion concentration delays the first hydrolysis
that shifted to G2 /M arrest after the second treatment step of NAMI-A and improves the stability of [mer-
(Table 1), and was still present after the third treatment. RuCl3 (dmso-S)(Him)(H2 O)] (NAMI-AH2 O ) in solution.
After the second treatment, a statistically significant This behaviour, indeed, suggests that hydrolytic de-
difference in G2 /M arrest was observed between NAMI- composition of NAMI-A in body fluids might be altered
A (1) and (2), with (2) being more active (^^ p < 0:01, by chloride ion concentration, similarly to what happens
Table 1). It is interesting to point out that both NAMI- for cisplatin.
A (1) and NAMI-A (2) significantly reduce protein At pH < 6.0 the chemical metabolism of NAMI-A
content in metGM cells after the first and the second changes, and the complex becomes much more inert.
treatment (MFI, about 750 vs. 866  30 for the control Chloride hydrolysis becomes negligible and slow dmso
after first, and about 925 vs. 1070  30 after the second hydrolysis generates the reactive aquated species, [trans-
treatment, p < 0:05). RuCl4 (H2 O)(Him)] . Treatment of NAMI-A with ex-
cess 9-methyladenine in water (pH 5.5) resulted in the
2.8. Effects of hydrolysis on cell uptake of ruthenium in partial formation of the [trans-RuCl4 (9-MeAde)(Him)]
metGM cells complex, whose resonances appeared clearly in the 1 H
NMR spectrum after 2 h of reaction. NMR pH titration
Significant differences were also observed in the in- experiments indicated that 9-MeAde coordinates to ru-
tracellular ruthenium content between NAMI-A and its thenium through its N7 atom.
hydrolysis species. It appears that cellular uptake of NAMI-A was not found to coordinate significantly to
ruthenium by metGM cells is favoured by hydrolysis of 9-MeAde in phosphate buffer (pH 7.4), where it com-
NAMI-A, since (2) showed increased uptake already petes with OH , a high affinity ligand for Ru(III), and
after the first treatment (Table 1). As shown, the intra- undergoes both chloride and dmso hydrolysis forming a
cellular ruthenium concentration of (2) is about 10-fold mixture of unidentified poly-oxo Ru species. However, it
higher than that of (1) and about fivefold higher than cannot be excluded that these species might also coor-
that of (3) (lg Ru/1  106 cells: 0.03 for (1), 0.34 for (2), dinate to 9-MeAde, but in this case their signals are too
0.07 for (3)). The same pattern of intracellular ruthe- broad and too weak to be detected by 1 H NMR spec-
nium concentrations was obtained after the second and troscopy. In support of this hypothesis is the fact that
the third treatment (data not shown). NAMI, the corresponding Naþ salt of NAMI-A, forms
intrastrand bifunctional adducts on polymeric DNA
[11]. Although with different techniques, this study
3. Discussion pointed out that NAMI binds both guanine and adenine
(with guanine being its preferential binding site), and in
To be able to understand the coordination of NAMI- this view it behaves similarly to NAMI-A, which is
A to biological targets, it was first considered necessary reasonable since the cation (Naþ or H2 imþ ) does not
M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412 409

influence the behaviour of the anion (at least not in involve different hydrolysis species formed through dif-
aqueous solution). Although not presented herewith, we ferent hydrolysis pathways. While DNA binding re-
also observed that NAMI-A coordinates to 9-ethyl- quires a rather intact species, binding to albumin might
guanine with the same efficacy as to 9-methyladenine. well occur after chloride hydrolysis.
The biological activity of the two main hydrolysis The different biological activity found for NAMI-A
species (i.e., the poly-oxo Ru species and [trans- and its hydrolysis species between KB and metGM cells
RuCl4 (H2 O)(Him)] ) was investigated, and compared to might account for the selective antimetastatic activity of
that of the parent compound NAMI-A. Hydrolysis affects NAMI-A. In the solid tumor KB cell line, the poly-oxo
the biological activity of NAMI-A in different ways in species have lost activity, whereas these species do arrest
human carcinoma cell lines and murine metastatic cells. metastatic cells at G2 /M phase and reduce their protein
Stronger reduction of cell proliferation on the human synthesis. Interestingly, the poly-oxo species show a
carcinoma cell line A2780 is achieved with solutions of larger potential to enter metastatic metGM cells and
fresh NAMI-A and of [trans-RuCl4 (H2 O)(Him)] than reach a significantly higher intracellular ruthenium
with a solution of poly-oxo Ru species. Surprisingly, al- concentration than fresh NAMI-A. Interestingly, the Ru
though the uptake in KB cells of the two former species uptake for the poly-oxo species is about thirty times
was found to be almost similar, [trans-RuCl4 (H2 O) more for the metastatic cells compared to the solid tu-
(Him)] was found more active than fresh NAMI-A in mor cells, which suggests the importance of this species
arresting the cell cycle of KB cells in the G2 /M phase. In for the antimetastatic activity. Therefore, it is proposed
contrast to the species 1 and 3, the poly-oxo Ru species that the selective antimetastatic activity of NAMI-A
were found to be only slightly able to enter KB cells, to during in vivo experiments might be, indeed, attribut-
arrest them at G2 /M phase and to affect protein synthesis. able to these poly-oxo Ru species, i.e., species probably
Unlike KB cells, the cell cycle and protein synthesis in more abundant after days of treatment.
metGM cells is not influenced by hydrolysis of NAMI-A,
since poly-oxo Ru species and NAMI-A gave comparable
effects. Contrary to KB cells, hydrolysis seems to favour 5. Experimental
ruthenium uptake in metGM cells; in fact, the intracel-
lular ruthenium concentration increased up to ten times in 5.1. Materials
the case of poly-oxo Ru species in comparison to NAMI-
A and up to thirty times compared to the uptake of poly- NAMI-A, [H2 im][trans-RuCl4 (dmso-S)(Him)] was
oxo Ru species in KB cells. prepared according to earlier published experimental
It is to be noted that in the solid tumor cell lines (KB section [5]. 9-Methyladenine was synthesised by methyl-
and A2780) reduction of proliferation occurs in the ation of adenine with methyl iodide, as previously de-
presence of NAMI-A and [trans-RuCl4 (H2 O)(Him)] , scribed [24].
and that these species do have the ability to bind to 9-
MeAde (and probably also to DNA). In metGM cell, on 5.2. Hydrolysis experiments
the other hand, the poly-oxo Ru species are taken up
easily, but these species do not bind to 9-MeAde. Hydrolysis was performed in phosphate buffer (0.05
M phosphate, 0.15 M NaCl) at pH 3.0, 5.0, 6.0, 6.5, 7.0,
7.4 and in water (D2 O). Reactions were followed di-
4. Conclusions rectly in 5 mm NMR tubes at 37.0 °C, on freshly pre-
pared 5 mM solutions of NAMI-A (2.3 mg/mL
Even though DNA is not expected to be the major phosphate buffer). Spectra were recorded automatically
biological target for the antimetastatic activity of every 2 min (pH 7.4, 7.0), 5 min (pH 6.5, 6.0), 10 min
NAMI-A, the present study indicates that it is likely that (pH 5.0, 3.0) and 30 min (D2 O) on a Bruker 300 DPX
this compound interacts with DNA, once it enters tumor spectrometer at 300.13 MHz. pH was measured by
cells and reaches the nuclei. The molecular stability re- laboratory pH Meter (PHM220, MeterLabÒ, Radiom-
quested for the binding of NAMI-A to DNA can be eter Analytical S.A), no correction for the pD values was
achieved at relatively low pH, such as in a tumor cell. In performed. All experiments were performed at least
these conditions chloride hydrolysis may be slowed twice. Acetone (1 lL/mL) was added to the buffer and
down and the dmso ligand can be hydrolysed from used as the reference signal for the integration, as it gives
NAMI-A resulting in the reactive aqua-complex [trans- a sharp signal (2.06 ppm) that does not overlap with the
RuCl4 (H2 O)(Him)] , that subsequently coordinates to resonances of NAMI-A or its hydrolysis species. 1 H
DNA. These findings differ from reports on the inter- NMR spectra were calibrated on the acetone peak set at
action of NAMI-A with albumin, achieved after chlo- 2.06 ppm. Normal 1 H NMR spectra were obtained us-
ride hydrolysis [23], suggesting that interactions of ing a spectral window of 100 ppm and an acquisition
NAMI-A with biologically relevant molecules might time of 1.0 s.
410 M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412

5.3. Effect of Cl concentration trometer (Applied Biosystems SCIEX), Department of


Biochemistry, Biophysics and Macromolecular Chem-
Influence of chloride ion concentration on the hy- istry, University of Trieste, Italy.
drolysis of NAMI-A was studied in phosphate buffer
(0.05 M phosphate, pH 7.4) additioned with NaCl to the
5.8. Cell cultures
desired concentration of Cl (0–1 M). Hydrolysis was
followed directly in 5 mm NMR tubes on freshly pre-
A2780 (human ovarian carcinoma) and A2780 cis-
pared 5 mM solutions of NAMI-A, at 37.0 °C.
platin-resistant cell lines were maintained in DulbeccoÕs
modified EagleÕs Medium (DMEM) (Gibco BRLe, In-
5.4. Coordination to DNA model base vitrogen Corporation, NL) supplemented with 10% fetal
calf serum, 1% penicillinG sodium (100 units/mL), 1%
Coordination of 9-methyladenine to NAMI-A was streptomycin (100 lg/mL), and 1% Glutamax 100 (all
performed in three different solutions; (a) in water from Duchefa Biochemie BV, Netherlands, NL). KB
(D2 O), (b) in D2 O containing NaClO4 (10 mM); and (c) cell line (ECACC No. 86103004) was cultured in EagleÕs
in phosphate buffer (0.05 M phosphate, 0.15 M NaCl, medium added with 10% FCS, 1% non-essential amino
pH 7.4). In (a) and (c) NAMI-A was freshly dissolved in acids 100, 1% sodium pyruvate 100 mM, 1% L -gluta-
solution (5 mM) and a fourfold molar ratio of 9- mine, 1% penicillin/streptomycin 100 buffered with 5%
methyladenine was added directly to the 5 mm NMR NaHCO3 (20 lM) (all from EuroCloneÒ, Paington,
tube. In (b) NAMI-A was aged in the solution for at UK). Cells were harvested from confluent monolayers
least 3 days at room temperature prior to reaction with by 0.05% trypsin solution. metGM cell line (for refer-
9-methyladenine according to [11]. The influence of ex- ence, see [10]) was maintained in RPMI 1640 medium
cess dmso on the coordination of 9-MeAde to NAMI-A (Sigma Chemical) added with 10% FBS, 1% L -glutamine
was investigated in a 5 mM solution of NAMI-A (D2 O) 100, 1% penicillin/streptomycin 100, 1% non-essen-
added with 20 mM 9-MeAde and 10% dmso-d6 . In all tial amino acids 100, 0.5% gentamycin 100, 0.37%
cases, reaction was followed in situ in 5 mm NMR tubes, sodium pyruvate 100 mM (all from EuroCloneÒ).
at 37.0 °C, recording spectra every 30 min. During re-
action in D2 O, the pH spontaneously decreased from 5.5
5.9. In vitro treatment with NAMI-A
to ca. 3.5.
KB and metGM cells were seeded in 6-wells plates
5.5. Inversion recovery (0.5  106 cells/4 mL/well, Corning CostarÒ, New York,
USA) and left to adhere for 48 h at 37.0 °C, 5% CO2 . In
1
H NMR were recorded using the inversion recovery the case of metGM cells, plastic wells were pre-coated
pulse sequence with the following typical values: spectral with MatrigelÒ (30 lg/100 lL) to allow adhesion of cells
width of 100 ppm, acquisition time of 200 ms, delay time that usually grow in suspension. Two main hydrolysis
(tau) in the range between 10 and 300 ms. The delay species of NAMI-A were tested and compared to fresh
between consecutive scans was generally 100 ms. Anal- NAMI-A (1): poly-oxo Ru species, formed by advanced
yses were performed on a Bruker 300 DPX spectrometer hydrolysis of NAMI-A in PBS (2) and [trans-
at 300.13 MHz. RuCl4 (H2 O)(Him)] , formed by hydrolysis of NAMI-A
in water (3). Solution (1) was prepared by dissolving
NAMI-A immediately before treatment, while solutions
5.6. pH titration
(2) and (3) were prepared by hydrolysing NAMI-A in
PBS and in water, respectively, for 4 h at 37.0 °C. In
pH titration was performed at 25.0 °C on a 5 mM
these conditions, at t0 the cells received the solution (1)
D2 O solution of NAMI-A added with 20 mM 9-MeAde
containing 100% of NAMI-A, the solution (2) contain-
in a 5 mm NMR tube, adjusting the pH by DCl (0.1 N)
ing 100% of poly-oxo Ru species (also confirmed by the
and NaOD (0.1 N). Direct pH-meter readings were used
black colour of the solution), and the solution (3)
to estimate the pKa values (no correction for the pD
containing a mixture of NAMI-A and [trans-
values was performed).
RuCl4 (H2 O)(Him)] species (ca. 1.5:1). Regarding the
solution (3), it was impossible to further increase the
5.7. ESI-MS amount of [trans-RuCl4 (H2 O)(Him)] species in the so-
lution, since longer hydrolysis in water led to the for-
Mass spectra analyses were performed at the Gorla- mation of poly-oxo Ru species. All solutions were
eus Laboratories of Leiden University on a Finningan sterilised before treatment by filtration (filters 0.22 lm,
MAT 900 instrument, equipped with an electrospray Sartorius Minisart AG, G€ ottingen, Germany) and tested
interface (ESI) and at the API I ion-spray mass spec- at the final concentration of 0.1 mM. After incubation
M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412 411

with NAMI-A (1 h at 37.0 °C in PBS, pH 7.4), the cells the addiction of 100 lL of tetramethylammonium hy-
were washed twice with PBS and incubated for further droxide (TMAH) at 25% in water (Aldrich Chimica, It-
24 h in complete medium, before analyses. In the case of aly) and 100 lL of milliQ directly in the vials. Volumes
repeated treatments, the same procedure was performed were adjusted to 0.5 mL with milliQ water. Ruthenium
for three consecutive times. (PBS, phosphate-buffered quantitation: the concentration of ruthenium in biolog-
saline; 8 g NaCl, 0.2 g KCl, Na2 HPO4 1.15 g, 0.2 g ical samples was measured in triplicate by means of
KH2 PO4 for 1 L, pH 7.4). Graphite Furnace Atomic Absorption Spectrometry
(GFAAS), model SpectrAA-300, supplied with a specific
5.10. MTT colorimetric assay ruthenium emission lamp (Hollow cathode lamp P/N 56-
101447-00) Varian, Mulgrave, Vic., Australia. Before
Cell proliferation was evaluated by MTT colorimetric each daily analysis session a five point calibration curve
assay [25] based on the reduction of the tetrazolium salt, was traced using Ruthenium Custom-Grade Standard
MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazo- 998 mg mL1 , Inorganic Ventures (St. Louis, USA). In
lium bromide] by actively growing cells to produce a order to correct for possible deterioration of the graphite
blue formazan product. Briefly, cells were seeded into 96 furnace during a daily working session, after every twelve
well plastic plates (2000 cells/well) and treated with 100 samples the calibration curve was re-traced and standard
lL NAMI-A (1 mM) for 1 h, controls in PBS. After was measured every six samples. The lower and higher
treatment, cells were allowed to grow for further 24 h in limits of quantitation (LLQ, HLQ) were set at the con-
complete medium. Each well was then added with 50 lL centration levels which corresponds respectively to the
MTT solution (5 mg/mL in PBS, Sigma Chemical) and lowest and higher standard concentration employed. The
incubated in thermostat for 4 h. At the end of incuba- limit of detection (LOD) was estimated according to:
tion time, formazan crystals were solubilised with 100 EURACHEM guide ‘‘The fitness for purpose of ana-
lL dimethyl sulfoxide and optical density was measured lytical experimental section’’. LLQ, HLQ and LOD were
at 570 nm (Automated Microplate Reader EL311s, respectively: 20, 100 and about 10 ng Ru mL1 of sample.
BIO-TEKÒ Instruments, VT, USA). The quantitation of ruthenium was carried out in 10 lL
samples at 349.9 nm with an atomising temperature of
5.11. Cell cycle analysis 2500 °C, using argon as purge gas at a flow rate of
3.0 L min1 . For further details concerning the furnace
(a) Double staining FITC/PI: 0.5  106 cells were parameter settings, see [27].
fixed in 70% EtOH for at least 4 h, washed twice with
PBS and allowed to balance in PBS for 2 h. Pellets were
5.14. Statistical analysis
stained with 0.5 mL of a PBS solution containing 10 lg
PI, FITC 0.25 ng fluorescein isothiocyanate (FITC) and
Data were submitted to computer-assisted statistical
4 lg RNase (all from Sigma Chemicals) and left over-
analysis (Instat II, Graphpad Software, vs. 2.05) using
night before flow cytometric analysis.
the Student–Newman–Keuls for analysis of variance
(ANOVA) and t test for grouped data. Data were con-
5.12. Flow cytometry analyses
sidered to be statistically significant if the probability
factor ‘‘p’’ was <0.05, <0.01 and <0.001. Symbols used
Samples were analysed by the CoulterÒ EpicsÒ XLe
to identify the significance are *p < 0:05, **p < 0:01 and
Flow Cytometer (Coulter Corporation, MI, FL, USA)
***p < 0:001.
at the LINFA laboratories (Callerio Foundation ON-
LUS). For each sample, at least 10000 events were ac-
quired. Data were elaborated with the WinMDI vs. 2.7
(Scripps Research Institute, La Jolla, CA, USA) and 6. Abbreviations
with MulticycleÒ (for cell cycle distribution).
NAMI-A [H2 im][trans-RuCl4 (dmso-S)(Him)]
5.13. Atomic absorption spectroscopy NAMI-AH2 O [mer-RuCl3 (dmso-S)(Him)(H2 O)]
Him imidazole
Intracellular ruthenium content in treated cells was dmso dimethyl sulfoxide
determined by atomic absorption spectroscopy. Samples dmso-S sulfur-bonded dimethyl sulfoxide
were processed according to a modification of the pro- 9-MeAde 9-methyladenine
cedures described by Tamura et al. [26]. All specimens KB human ovarian carcinoma cell line
were dried in crio-vials. A first step of drying was per- metGM murine metastatic cell line
formed overnight at 80 °C, a second step at 105 °C, until PI propidium iodide
the samples reached the constant dried weight. The de- FITC fluorescein isothiocyanate
composition of the dried samples was carried out with PBS phosphate-buffered saline
412 M. Bacac et al. / Journal of Inorganic Biochemistry 98 (2004) 402–412

Acknowledgements [9] S. Pacor, M. Vadori, F. Vita, M. Bacac, M.R. Soranzo, G.


Zabucchi, G. Sava, Anticancer Res. 21 (2001) 2523–2530.
[10] S.J. Berners Price, P.J. Sadler, Coord. Chem. Rev. 151 (1996) 1–
Marie Curie Training Fellowship (Medicinor re- 40.
search program 1.4.1; Contract No. HPMT-CT-2000- [11] J. Malina, O. Novakova, B.K. Keppler, E. Alessio, V. Brabec, J.
00192; type PHD20) and Fondazione C&D Callerio Biol. Inorg. Chem. 6 (2001) 435–445.
Onlus are acknowledged for financial support of the [12] G. Mestroni, E. Alessio, G. Sava, S. Pacor, M. Coluccia, A.
Ph.D. fellowship of M. Bacac. Work contributed by Boccarelli, Metal Based Drugs 1 (1993) 41–63.
[13] A. K€ ung, T. Pieper, R. Wissiack, E. Rosenberg, B.K. Keppler, J.
MIUR ‘‘Pharmacological Mechanisms of the Antimet- Biol. Inorg. Chem. 6 (2001) 292–299.
astatic Activity of Metal Based Drugs’’, and done within [14] R.M. Izatt, J.J. Christensen, J.H. Rytting, Chem. Rev. 71 (1971)
the framework of the EU COST D20/0005/01 and D20/ 439–481.
0001/00 Working Groups. Special thanks to Fons [15] K. Inagaki, M. Kuwayama, Y. Kidani, J. Inorg. Biochem. 16
Lefeber and Kees Erkelens for their generous help and (1982) 59–65.
[16] T.G. Spiro, Nucleic Acid-Metal Ion Interactions, Wiley, New
technical assistance during NMR measurements and to York, 1980.
Moreno Cocchietto for AAS assistance. [17] S.J. Lippard, Progress in Inorganic Chemistry, Wiley, New York,
1977.
[18] A.H. Velders, B. van der Geest, H. Kooijman, A.L. Spek, J.G.
References Haasnoot, J. Reedijk, Eur. J. Inorg. Chem. (2001) 369–372.
[19] A.C.G. Hotze, M.E.T. Broekhuisen, A.H. Velders, K. van der
[1] M.J. Clarke, F. Zhu, D.R. Frasca, Chem. Rev. 99 (1999) 2511–2533. Schilden, J.G. Haasnoot, J. Reedijk, Eur. J. Inorg. Chem. (2002)
[2] J. Reedijk, Proc. Natl. Acad. Sci. USA 100 (2003) 3611–3616. 369–376.
[3] G. Sava, A. Bergamo, Int. J. Oncol. 17 (2000) 353–365. [20] G. Sava, A. Bergamo, S. Zorzet, B. Gava, C. Casarsa, M.
[4] E. Alessio, G. Balducci, A. Lutman, G. Mestroni, H. Calligaris, Cocchietto, A. Furlani, V. Scarcia, B. Serli, E. Iengo, E. Alessio,
W.M. Attia, Inorg. Chim. Acta 203 (1993) 205–217. G. Mestroni, Eur. J. Cancer 38 (2002) 427–435.
[5] G. Sava, E. Alessio, A. Bergamo, G. Mestroni, in: M.J. Clarke, [21] M.M. Taqui Khan, G. Ramachandraiah, R.S. Shukla, Inorg.
P.J. Sadler (Eds.), Sulfoxide Ruthenium Complexes: Non Toxic Chem. 27 (1988) 3274–3278.
Tools for the Selective Treatment of Solid Tumor Metastases, [22] A. Bergamo, S. Zorzet, B. Gava, A. Sorc, E. Alessio, E. Iengo, G.
1999, pp. 143–170. Sava, Anti-cancer Drugs 11 (2000) 665–672.
[6] G. Sava, S. Zorzet, L. Peressin, G. Mestroni, G. Zassinovich, A. [23] L. Messori, P. Orioli, D. Vullo, E. Alessio, E. Iengo, Eur. J.
Bontempi, Inorg. Chim. Acta 137 (1987) 69–71. Biochem. 267 (2000) 1206–1213.
[7] E. Alessio, W. Attia, M. Calligaris, D. Cauci, L. Dolzani, G. [24] E.G. Talman, W. Br€ uning, J. Reedijk, A.L. Spek, N. Veldman,
Mestroni, C. Monti-Bragadin, G. Nardin, F. Quadrifoglio, G. Inorg. Chem. 36 (1997) 854–861.
Sava, M. Tamaro, S. Zorzet, in: M. Nicolini (ed.), Boston, [25] H. Tada, O. Shiho, Kuroshima, K. Koyama, M. Tsukamoto, J.
Platinum and Other Metal Coordination Compounds in Cancer Immunol. Meth. 93 (1986) 157–165.
Chemotheraphy, N€yhoff 1988, pp. 617–633. [26] H. Tamura, T. Arai, M. Nagase, N. Ichinose, Bunseki Kagaku 41
[8] A. Bergamo, R. Gagliardi, V. Scarcia, A. Furlani, E. Alessio, G. (1992) T13–T18.
Mestroni, G. Sava, J. Pharmacol. Exp. Ther. 289 (1999) 559–564. [27] M. Cocchietto, G. Sava, Pharmacol. Toxicol. 87 (2000) 193–197.

Вам также может понравиться