Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/233504271

Carbohydrate-Metal Complexes and their Potential as Anticancer Agents

Article in Current Medicinal Chemistry · October 2008


DOI: 10.2174/092986708785908978

CITATIONS READS
176 2,027

5 authors, including:

Christian Hartinger Alexey Nazarov


University of Auckland Lomonosov Moscow State University
322 PUBLICATIONS 17,765 CITATIONS 96 PUBLICATIONS 3,571 CITATIONS

SEE PROFILE SEE PROFILE

Muhammad Ashraf Shaheen


University of Sargodha
57 PUBLICATIONS 638 CITATIONS

SEE PROFILE

All content following this page was uploaded by Christian Hartinger on 29 May 2014.

The user has requested enhancement of the downloaded file.


2574 Current Medicinal Chemistry, 2008, 15, 2574-2591

Carbohydrate-Metal Complexes and their Potential as Anticancer Agents


Christian G. Hartinger*,1,2, Alexey A. Nazarov*,1,2, Shaheen M. Ashraf1, Paul J. Dyson2 and Bern-
hard K. Keppler1
1
University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
2
Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne,
Switzerland
Abstract: Platinum complex-based chemotherapy is one of the major treatment options of many malignancies. Although
severe side effects occur, and only a limited spectrum of tumors can be cured, Pt compounds are used in every second
therapy scheme. Thus, many different drug design strategies have been employed for improving the properties of antican-
cer drugs including pH or redox activation in the tumor, variation of the metal center and therefore the redox and ligand
exchange properties, the application of multinuclear metal complexes, the development of targeted approaches, etc. Ap-
plication of carbohydrate–metal complexes is an example of a targeted approach exploiting the biochemical and metabolic
functions of diverse sugars in living organisms for transport and accumulation. Natural carbohydrates and synthetic de-
rivatives possess a manifold of donors endowing them with the ability to coordinate metal centers and providing some ad-
ditional advantages over other ligands, e.g., biocompatibility, non-toxicity, enantiomeric purity, water solubility, and well-
explored chemistry. In recent years, several examples of carbohydrate compounds have been developed for diverse me-
dicinal applications ranging from compounds with antibiotic, antiviral, or fungicidal activity and anticancer compounds.
Herein, metal complexes with carbohydrate ligands are reviewed and the role of the carbohydrate carriers on the antineo-
plastic activity of these compounds, both in vitro and in vivo, is described.
Keywords: Antitumor chemotherapy, carbohydrates, metal complexes, review.

1. INTRODUCTION The success of cisplatin in treating solid tumors was fol-


lowed by the synthesis of a plethora of cisplatin analogues in
Malignant diseases cause approximately 25% of the the hope of finding superior agents [14]. The second genera-
deaths in developed countries and chemotherapy is a central tion platinum compounds carboplatin 2, cis-diammine(1,1-
component in preventing such deaths along with surgery and cyclobutanedicarboxylato)platinum(II), and oxaliplatin 3,
radiation therapy [1]. A variety of chemotherapeutics are [(1R,2R)-cyclohexane-1,2-diamine](oxalato)platinum(II),
available including alkylating agents, antimetabolites, anti- were approved for worldwide clinical use [15]. Oxaliplatin in
tumor antibiotics, natural products, hormones, hormone combination with 5-fluorouracil has emerged as the first line
antagonists, and metal complexes (often considered as alky- therapy for metastatic colorectal cancer [16]. Other plati-
lating agents). num-based drugs have been regionally approved (nedaplatin
Medicinal applications of metal compounds can be traced in Japan, lobaplatin in China, and heptaplatin in South Ko-
back to ancient times, with precious metals like gold and rea) and several platinum complexes are currently in clinical
silver being applied by the ancient Chinese, Egyptians, trials as single agent therapies or in combination with estab-
Greeks and Indians for treatment of various diseases [2]. lished drugs [15,17-21].
Chemotherapeutic use against cancer started with the devel- In order to circumvent the side effects of the prevalent
opment of cisplatin 1, cis-diamminedichloridoplatinum (II) anticancer drugs, to enhance the cytotoxicity profile, and to
(Fig. (1)), which was approved for the treatment of cancer augment the efficacy and specificity of the antitumor agents,
worldwide in 1978. Cisplatin was first prepared by Peyrone many new compounds have been prepared and evaluated
in 1844 [3], and its structure was correctly proposed by [9,15,22-27]. Several complexes based on different metals
Werner in 1893 [4], but the antineoplastic activity of cis- including ruthenium [28-31], gallium [32-35], rhodium and
platin was only discovered (somewhat serendipitously) in osmium, [36-38], copper and iron [32,39-41], molybdenum
1969 by Rosenberg [5,6]. Cisplatin is now one of the most [42], gold [43], palladium [44,45] and titanium [46,47] have
widely used and most effective chemotherapeutic agents in been synthesized and investigated for antineoplastic proper-
the treatment of epithelial malignancies such as lung, head, ties.
neck, ovarian, bladder and testicular cancers [7-10]. How-
The role of ligands in tuning the cytotoxic characteristics
ever, the treatment of tumors with cisplatin is compounded
of the complex is of considerable importance. Lipophilicity
by severe side effects such as renal toxicity, gastrointestinal
and stability of a metal complex strongly depend on the
toxicity, peripheral neuropathy, asthenia, myelosuppression
nature of the ligands [13]. Carefully designed/selected
and ototoxicity [9,11,12]. The problem of intrinsic and ac-
quired drug resistance during treatment further limits the ligands for metal-based drugs impart desirable features that
influence the toxicity, bioavailability, and the specificity of a
application of cisplatin [13].
metallodrug candidate [31,48].
Carbohydrates represent an excellent class of tunable
*Address correspondence to these authors at the University of Vienna,
Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, ligands for use in medicinal inorganic chemistry, they are
Austria; Tel: +43 1 4277 52609; Fax: +43 1 4277 9526; often inexpensive, available in enantiomerically pure form,
E-mail: christian.hartinger@univie.ac.at; alex.nazarov@univie.ac.at naturally available and possess functional groups for modu-

0929-8673/08 $55.00+.00 © 2008 Bentham Science Publishers Ltd.


Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2575

O
H2
H3N Cl H3N O N O O
Pt Pt Pt
H3N Cl H3N O N O
H2 O
O

1 2 3

O
O NH2 O CH3
H3N O O NH2 O
Pt Pt Pt
H3N O O NH2 O NH2 O O
O
4 5 6

Fig. (1). Pt-based anticancer complexes cisplatin 1, carboplatin 2 and oxaliplatin 3 (approved for worldwide clinical use) and nedaplatin 4,
heptaplatin (SKI2053R) 5 and lobaplatin 6 (regionally approved).

lation according to the desired properties. Non-functiona- 2. PLATINUM COMPLEXES DERIVED FROM CAR-
lized carbohydrates are week donor ligands, but carbohy- BOHYDRATE LIGANDS
drates can easily be modified with a broad range of func-
tional groups, e.g., amino, carboxylato, thiol, and different 2.1. N Donors for Coordination to Pt Centers
phosphorus containing moieties, capable of serving as strong All of the clinically established anticancer agents based
ligands [49]. on platinum are equipped with nitrogen-donor ligands that
Carbohydrate chemistry links organic chemistry with are strongly bound to the metal center. In general, these
medicinal chemistry and biology, due to the role that carbo- ligands are not substituted (released) as the platinum com-
hydrates play in glycobiology, in anti-infective therapy as pound exerts its anticancer effect. Nevertheless, the nature of
components of antibiotics, antitumor, and antiviral agents, the N-donor ligands dramatically influences the anticancer
and in related biomedical areas [50]. Attaching a carbohy- properties of platinum complexes and leads to activity
drate moiety to a metal-binding ligand invariably leads to against different types of cancers [9].
reduced toxicity, and also to improved solubility and mo- 2.1.1. Mono- and Disaccharide Ligands with Amino Func-
lecular targeting. Carbohydrate ligands can be used for tionalities
chelating metal ions in order to generate a well-defined bind-
ing environment and to enhance the stability of the resulting 2.1.1.1. Simple Aminosugar Ligands
complexes. Accordingly, the carbohydrate unit remains pen- Exploiting the high affinity of platinum towards N-donor
dant and is (potentially) available to interact with carbohy- ligands readily affords aminosugar–platinum complexes in
drate transporters in the body [48]. Modified carbohydrates which the carbohydrate moiety is bound to the platinum
and oligosaccharides are known to interfere with carbohy- center via an amino group. Shortly after the approval of
drate–protein interactions and can inhibit cell–cell recogni- cisplatin in the clinic there was a considerable interest in
tion and adhesion phenomena, which are crucial in cancer carbohydrate-analogues of cisplatin. The simplest represen-
growth and progression [51]. Moreover, there are several tatives of these compounds are based on glucosamine and
organic compounds in clinical use as anticancer agents or at their general structures are shown in Fig. (2) [58-60]. Fur-
an advanced clinical stage possessing a sugar moiety, e.g., thermore, a series of platinum(II) complexes based on 2-
the DNA strand break-inducing compound bleomycin [52], amino-2-deoxy-D-glucopyranose and related derivatives with
the alkylating agent glufosfamide [53], and the DNA interca- a variety of leaving groups were reported [58,61-64]. Unfor-
lator doxorubicin (trade name adriamycin) [54]. tunately, for these compounds very little in vitro and in vivo
anticancer data was reported, the only data we are aware of
Carbohydrate-metal chemistry has been extensively stud- is in the patent literature proving the applicability of these
ied and several important reviews [49,55,56] dealing with compounds as anticancer agents [65,66]; the characterization
carbohydrate-metal complexes have been published in recent of the compounds was not described. In comparison to cis-
years. Herein, the role of metallodrugs based on carbohy- platin, used as a standard control, higher doses of the carbo-
drates and their derivatives for the treatment of cancer is hydrate complexes were required to observe the same anti-
discussed. The other medicinal applications of carbohydrate- cancer effect. Notably, the carbohydrate does not seem to
metal complexes, e.g. in diagnosis, are beyond the scope of have much influence on the anticancer activity since the
this review, and also pharmaceutical compositions contain- median survival times for complexes with different sugar
ing cyclodextrins and having no covalent bond between the ligands were very similar in single cancer models.
metal complex and the organic moiety are not covered (for a
Tsubomura et al. used chelate-forming 2,3-diamino sug-
recent review see ref. [57]).
ars to obtain [PtCl2(methyl 2,3-diamino-2,3-dideoxy--D-
2576 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

mannopyranoside)]·H2O 10 and [PtCl2(2,3-diamino-2,3- Structurally similar compounds to 11 were obtained by


dideoxy-D-glucose)]·H2O 11 (Fig. (3)). Both compounds replacing the leaving chlorido ligands by iodido 13, oxalato
were found to exhibit promising antitumor activity against 14 and malonato 15 (Fig. (3)) and the cytotoxicities were
sarcoma 180 in mice with the glucose derivative 11 being determined in four cell lines (CH1, HeLa, SW480, U2OS),
more efficient (T/C = 411 vs. 230%) [67]. Compound 11 was with IC50 values being in the low micromolar range, see
found to be active in sarcoma 180-bearing mice, prolonged Table (1) [71]. The cytotoxicity of the chlorido complex 11
their survival time by 311% at 50 mg/kg, i.p. [68]. The ap- is one to two orders of magnitude lower than that of ox-
plied dose was 50 mg/kg for the carbohydrate–complexes vs. aliplatin (l-OHP) and its (S,S)-enantiomer (d-OHP), but
8 mg/kg for cisplatin, however, the LD50 of cisplatin was comparable to carboplatin in two of the four cell lines. To
determined in the mouse model at 13 mg/kg indicating the gain insights into the mode of action of the compounds the
significantly lower toxicity of the sugar derivatives. Fur- binding affinity toward the DNA model nucleotide dGMP
thermore, an influence of the conformation of the NH 2 was studied. Compound 11 binds to dGMP in a similar way
groups was elucidated [69], which is in agreement to the to cisplatin, whereas the iodido compound 13 reacts at a
results observed for oxaliplatin (compare R,R- vs. S,S-chxn) much lower rate, probably due to slower hydrolysis. For the
[70]. The most promising compounds in the study were de- complexes with the chelating oxalato and malonato ligands
termined as [PtCl2(benzyl 3,4-diamino-3,4-dideoxy-ß-L- 14 and 15, the stability of the dicarboxylato ligands resulted
arabinopyranoside)] 12 with a lifespan increase of 390% vs. in a lower reactivity to dGMP (similar to other series of Pt
the controls at low dose and 10 with a T/C of 410% at a compounds).
higher dosage [69]. Furthermore, the complexes were not
Diaminosugar–Pt complexes with methyl 2,3-diamino-
active against BDF1 mice bearing the L1210 tumor, with the
2,3-dideoxy-L-xylopyranoside and 2,3-diamino-2,3-dideoxy-
exception of 12 showing a T/C value of 250%, following a
1,5-anhydro-L-xylitol (Fig. (4)) have been synthesized by
single treatment with 32 mg/kg of the complex [69].
Hanessian and Wang [72,73]. The 4-hydroxo complexes 16
sugar
-
sugar sugar sugar
and 18 exhibited cytotoxicity against P338 leukemia in vitro,
and in vivo they are comparable to cisplatin, while 17 was
H2N Cl H2N NH2 H2N NH2 found to be less active (Table (2)). It was proposed that the
Pt Pt Pt
hydroxy group at C-4 position in 16 and 18 effects the solu-
Cl Cl Cl Cl Cl Cl
bility, hydrophilicity, electronic stabilization, distribution of
7 8 9 the complex in the plasma, and cytotoxicity. In analogy to
Fig. (2). Generic structures of mono- and diaminosugar–platinum the cisplatin/carboplatin couple, complex 19 showed much
complexes 7–9 (sugar = glucose, galactose, mannose, arabinose, reduced activity in comparison to 16 and 18, which is proba-
etc.). bly due to the stability of the cyclobutane-1,1-dicarboxylate
ligand as a leaving group lowering the reactivity towards
H2 DNA. Compounds 16 and 18 were active against L1210,
HO N HO
O O DDP5, P388 and their resistant cell lines although being less
HO HO BnO O
H2N H2N OH HO
potent than oxaliplatin.
NH2 NH2
Pt OMe Pt NH2 In a series of patents, Sasamori et al. reported the use of
Cl X Pt complexes derivatized with L-xylopyranose and L-
Cl X Cl Pt
xylopyranoside ligands combined with different leaving
11 X = Cl 12 Cl groups, including chlorido, nitrato, isocyanato, oxalato, etc.
10
13 X=I
14 X = oxalato [74-80] (Fig. (4)). In particular, the inhibition of mice leu-
15 X = malonato kemia (L1210) was observed, leading to increased survival
rates of >275 and >375% for the chlorido complex 20a and
Fig. (3). Structures of diaminosugar–platinum complexes 10-15.

Table 1. Cytotoxicity of Diaminoglucose-Based Platinum Complexes Compared to Carboplatin, Oxaliplatin (l-OHP) and d-OHP in
Four Human Cancer Cell Lines [71]

IC50 (M) a
Compounds
CH1 HeLa SW480 U2OS

11 5.1 ± 0.3 7.3 ± 0.8 67 ± 13 82 ± 24


13 7.9 ± 2.3 14 ± 2 79 ± 4 119 ± 16
14 4.1 ± 0.1 10 ± 1 87 ± 3 175 ± 20
15 6.3 ± 0.4 23 ± 7 151 ± 8 > 250
Carboplatin 0.52 ± 0.01 5.4 ± 1.5 61 ± 10 34 ± 1
Oxaliplatin, l-OHP 0.34 ± 0.09 0.30 ± 0.08 0.30 ± 0.08 0.72 ± 0.24
d-OHP 1.0 ± 0.1 1.1 ± 0.4 1.3 ± 0.3 2.6 ± 0.7
a
50% inhibitory concentrations in CH1 (ovarian carcinoma), HeLa (cervix carcinoma), SW480 (colon carcinoma), and U2OS (osteosarcoma) cell lines determined using the MTT
assay.
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2577

O OH
O OR O MeO
OH NH2
MeO NH2 NH2 H2N
H2N H2N Pt O
Pt Cl Pt Cl O
Cl Cl O

16 R = H O
18
17 R = Me
19

H2 H2 OH
O O N O
N
RO N Pt Cl
EtO N Pt Cl HO NH2
HO H2 Cl HO H2 Cl HO H2N
OH
Pt O
O
20a R = H 21 22 O
20b R = benzyl

Fig. (4). Structures of xylose-derived platinum complexes 16-22.

Table 2. In Vivo Antitumor Evaluation of Xylose-Derived Platinum Complexes 16-19 [72,73]

Complex Leukemia tumor model Dose i.p. / mg/kg T/C /%

16 P388 25 227
17 25 191
18 12.5 236
19 25 114
Cisplatin 4 236
16 L1210 50 323
17 50 218

its benzyl pyranoside analogue 20b in mice transplanted with R and L5178Y-S being defective in the incision step of the
L1210 leukemic cells at 1.6 and 0.9 mg/kg per day i.p., re- nucleotide excision repair system and the double-strand
spectively. In addition, the ethyl xylopyranoside Pt(IV) com- break repair mechanism, respectively. The IC50 values were
pound 21 and other related derivatives were prepared and 21 found to be similar for 23 in both cell lines, whereas the Pd
was shown to prolong lifespan by >375% at 3.1 mg/kg per compound 24 was 3-times more active in L5178Y-R com-
day in the same tumor model. pared to L5178Y-S cells. Overall, the Pt complex was more
cytotoxic than the Pd analogue, and in vitro results did not
In addition to xylopyranoses with the amino functionali-
correlate with the ability of the complexes to modify DNA
ties in vicinal positions, compounds with amino substituents
by inducing crosslinks, with the Pd compound leading to
in 3 and 5 positions were prepared [75,79]. Notably, 22 with
the 2-hydroxoacetato ligand was found to increase the sur- higher retardation of DNA than the Pt species, proposing a
non-DNA-related mode of action for at least one of the com-
vival rate by 264% at 3.8 mg/kg-day i.p. in L1210 leukemia
pounds.
bearing mice. Kolar also patented the application of platinum
complexes comprising 6-membered rings with diamino sug- +
Me Me
ars as anticancer agents. In this case a large number of ex- H2N O H2N O
amples of 2,4-diaminoglucopyranose Pt complexes were OMe OMe
Cl M Cl Pt NO3-
reported, in particular with regard to different leaving co-
NH2 NH2
ligands, and their biological activity was studied and com- Cl DMSO
pared to cisplatin. The compounds were shown to exhibit in
vitro activity at lower toxicity than cisplatin [81], but it is not 23 M = Pt 25
clear if the results are statistically significant. 24 M = Pd
Samochocka et al. studied platinum(II) (23) and palla- Fig. (5). Structures of L-lyxo-hexapyranoside-derived complexes
dium(II) (24) complexes based on methyl 3,4-diamino- 23-25.
2,3,4,6-tetradeoxy--L-lyxo-hexopyranoside (Fig. (5))
[82,83]. In vitro evaluation of the neutral platinum(II) com- The cationic platinum(II) complex [PtCl(methyl 3,4-
plex with the diaminosugar revealed antitumor activity diamino-2,3,4,6-tetradeoxy--L-lyxopyranoside)(DMSO)]NO3
against mouse lymphoma cell lines (L5178Y), with L5178Y- 25 was tested for the inhibition of mouse L1210 lymphoma
cell growth with reference to cisplatin [84]. In contrast to
2578 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

O and protein syntheses did not allow the mode of action to be


CH3
clearly defined. Thymidine incorporation into DNA was
HN found to be 77% after a 16 hour-incubation period and
uridine and labeled amino acids were incorporated into RNA
HO O N and protein to an extent of 17 and 100%, respectively.
PtCl2
O
There are a large number of platinum complexes with
nucleotides reported in the literature, most of which originate
from the reaction of dGMP and other nucleosides and nu-
NH2
2 cleotides as model substrates for metal complexes with
DNA. However, virtually all these species were not isolated
26 and tested on their anticancer activity.

Fig. (6). Formula of the nucleoside-Pt complex 26.


2.1.1.3. Polynuclear Pt Complexes
The concept of applying polynuclear metal complexes as
cisplatin, at concentrations of up to 0.1 mg/ml no signs of anticancer agents has been shown to improve anticancer
toxicity were observed. The ID50 (50% inhibitory dose) val- activity and to extend the range of treatable tumors including
ues were determined to be similar for 25 and cisplatin, but those that are otherwise resistant to platinum chemotherapy
for the ID90 cisplatin was about two times more toxic. [86]. Sachinvala et al. prepared platinum complexes based
on disaccharides by linking two sugar moieties. The sucrose-
2.1.1.2. Nucleoside-Aminoribose Ligands based complexes were found to exhibit good water solubility
Nucleoside-Pt complexes were introduced by Lin et al. as and cytotoxicities comparable to that of cisplatin (Fig. (7)).
anticancer agents [85]. The replication of murine L1210 cells A comparison of the mononuclear complexes 27–30, di-
in vitro was studied for several compounds, but only dichlor- (31) and trinuclear (32) and (33) Pt(II) complexes was re-
idobis(3-amino-3-deoxythymidine)platinum(II) 26 (Fig. ported [87,88]; the mononuclear complexes were not cyto-
(6)) was found to be a potent inhibitor with an ED50 of 0.8 toxic, whereas 31 (with notably high solubility of 10
μM (320 mg/kg, 2 per day for 3 consecutive days). A dose mg/mL) was comparable to that of cisplatin in vitro in hu-
dependent increase in survival time was observed in mice man KB cells and in vivo against implanted Lewis lung car-
bearing the L1210 tumor (Table (3)). However, 26 was less cinoma and P388 leukemia in mice with T/C  310 and 
potent than cisplatin (T/C of 205% for cisplatin at a single 291%, respectively. In contrast to cisplatin, the treatment of
treatment with 8 mg/kg). Inhibition assays on RNA, DNA the mice with 31 was not accompanied by severe toxicity,

Table 3. Effect of the Dinucleoside-Platinum Complex 26 on the Survival of Mice Bearing the L1210 Ascites Neoplasm and Com-
parison to Cisplatin [85]

Compound Dose / na  mg/kg Average survival / days T/C / %

26 control 10.3 0
6  80 10.7 104
6  160 13.2 128
6  320 18.0 175
cisplatin [212] 18 - 205
a
number of injections.

X H3N H3N
H2N Pt X H2N Pt Cl H2N Pt X
H3N Cl H3N X
Cl X
Pt Pt
MeO O HO O MeO O
NH2 H2N Cl H2N X
MeO HO MeO
MeO O O O MeO O
O O O
MeO
X NH2
OMe OH Pt OMe
MeO HO X NH3 MeO

27 X = iodido 31 32 X = chlorido
28 X = oxalate 33 X = malonate
29 X = malonate
30 X = cyclopropylmalonate

Fig. (7). Mono-, di- and trinuclear platinum complexes derived from sucrose.
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2579

although weight loss was observed during the 50-day obser- sensitive tumors (Gross leukemia and Lewis lung tumor).
vation period. The structurally similar trinuclear compound However, no advantage over treatment with doxorubicin was
32 was reasonably soluble in water (though being fully found in these tumor models.
methyl-protected) and was slightly more active in vitro than O
O OH
the dinuclear compound 31 and in vivo a life time prolonga- OH
tion of T/C  355% was observed, which might also be due
OH
to the higher lipophilicity of the compound leading fre-
quently to higher drug concentrations in the cell, although no
cellular uptake data was reported to substantiate this hy-
OMe O OH O
pothesis.
Evaluation of the anticancer activity of 34 and 35 (Fig. Me O
(8)) against P388 leukemia revealed high anticancer activity NH2
with IC50 values in the high nM range [89], notably, inde- OH
NH2 Pt Cl
pendent of the number of Pt centers coordinated to the sugar
t-Bu
moiety. Cl
36
Ph O OH Ph O OH
O O O O Fig. (9). Structure of the platinum-doxorubicin complex 36.
Dinuclear Pt compounds based on doxorubicin have also
NH OMe NH OMe been reported, but without biological data [95]. Notably, the
Pt(NH3)Cl2 Pt centers were found to coordinate to the hydroxyquinone
group. The same was observed for a series of multinuclear
Pt Cl neomycin B complexes [96].
Cl2(H3N)Pt
OMe HN Cl OMe NH A series of Pt ammine quinone complexes with naphtho-
quinone derivatives as ligands were prepared and at a dose of
O O 3  30 mg/kg i.p. a long term survival in 8 of 10 mice bear-
O O
OH O Ph OH O Ph ing an Ehrlich ascites tumor cells was observed [97]. In the
34 35 same tumor model the positive control animals were treated
with cisplatin and 4 out of 10 survived for more than 60
Fig. (8). Mono- and dinuclear mannose-based Pt(II) complexes. days. Related complexes based on Pd(II) and Fe(III) coordi-
In a patent dinuclear aminosugar Pt compounds based on nated to the anthracyclines doxorubicin and daunorubicin
an anthracenedione-linker were claimed as potential antican- were reported with antitumor activity against P388 leukemia
cer agents [90], but no biological data was reported. cells [98,99]. The interaction of the complexes with plasma
was studied and no binding was observed. It was also found
2.1.1.4. Multifunctional and Organometallic Complexes that the Pd–anthracycline bond was not cleaved on binding
Another strategy for overcoming resistance problems to DNA. Furthermore, the complexes did not increase
during chemotherapy is the development of multifunctional superoxide formation in contrast to their anthracycline
drugs, i.e., attaching a known organic drug to a platinum ligands alone.
center [91,92]. By linking two cytotoxic moieties Pasini et Metallocenes covalently linked to doxorubicin [100] have
al. synthesized the platinum-doxorubicin compound 36 (Fig. also been studied. The zirconocene-doxorubicin compound
(9)) (doxorubicin and also daunorubicin are used to treat a was found to be inactive, whereas the Ti analogues exhibited
wide range of different cancer types, and cisplatin does not antitumor activity similar to that of titanocene dichloride.
show cross-resistance to anthracyclines) [93]. Compound 36 The mode of action is thought to involve the dissociation of
was tested against murine P388 and L1210 leukemia and the the complex, and the presence of metal ions induces binding
cisplatin- and doxorubicin-resistant sublines L1210/DDP and to DNA and erythrocyte ghosts, although it is possible that
P388/Dx, respectively, and against the naturally doxorubi- the doxorubicin ligand is released from the Ti center prior to
cin-resistant solid murine early colon carcinoma #26. Prom- entry into the cell.
ising activity, also in comparison to the parent compounds
Other organometallic compounds derived from the reac-
cisplatin and doxorubicin, was found against the P388 tumor
tion of [{PtMe3(H2O)2}2SO4] with (amino)-D-glucitol and
model, whereas all the tested compounds were not very ac-
glucosamine gave rise of compounds of the general formula
tive against L1210 leukemia. In the drug resistant tumor
[PtMe3L]2SO4, whereas reaction with (2-amino)-D-gluconate
models, similar activity as for cisplatin and doxorubicin was
and D-threonate resulted in the complexes [PtMe3L] and
reported, yielding only a low number of long-term survivors.
[PtMe3L(H2O)], respectively [101]. Although proposed as
Against murine early colon carcinoma #26 36 exhibited anticancer drugs no biological data was reported. The same
similar activity to doxorubicin and was less active than cis- is true for Pt organometallics coordinated via the C1 atom of
platin. Preliminary results on the mode of action suggest the sugar ligands [63] and (acetyl)glucose and ribose deriva-
intercalation of the doxorubicin moiety into DNA, rather tives via organic linkers [102].
than covalent binding via the Pt center [94]. The preclinical
evaluation of this complex was extended to the solid doxoru- 2.1.1.5. Platinum(IV) Complexes
bicin-resistant tumors BI6 melanoma (acquired resistance) The platinum(IV) unit represents an excellent motif to
and MXT mammary carcinoma (intrinsic resistance) and to which functional ligands can be attached, which are subse-
2580 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

quently lost by reduction to Pt(II) inside cells [103,104]. logical evaluation of these complexes in vivo against P388
With sugars there are few examples although Pt(IV) sugar leukemia cells implanted in mice revealed most of the palla-
complexes were claimed in a patent by Kay et al. [105]. dium complexes to be inactive with the exception of a disac-
However, the characterization of the compounds was not charide maltose-Pd complex which had a T/C value of 120%
provided and the suggested structure, including coordination at a dose of 400 mg/kg. The lower antineoplastic activity of
to the ring-oxygen, is uncertain. Within the series of tested the palladium(II) complexes in comparison to Pt(II) com-
compounds the Pt compounds containing either a methyl - pounds is thought to be due to low genotoxicity of Pd com-
D-mannopyranoside or D-glucosamine ligand were the most plexes, differing DNA damage, and lower stability of the
active and in some of the tumor models were more active complexes, especially in biological fluids.
than cisplatin.
Talebian et al. synthesized Pt compounds linked via an
2.1.2. Coordination to Pt via a Linker ethylendiamine moiety to glucose, galactose and mannose
units, in pyranose, furanose and 2-amino-2-deoxy forms
Coordination of sugars to Pt centers via donor-atom bear-
[110]. The respective N-(6-deoxy-1,2:3,4-di-O-isopropyli-
ing spacers offers several advantages over direct coordina-
dene--D-6-galactopyranosyl) cis-dichloridoplatinum(II)
tion of the Pt moiety to aminosugars: cell penetration and
complex 39 (Fig. (11)) was administered i.p. at a single dose
drug-receptor interactions are thought to be improved and
the sugar functionality helps to maintain high solubility of 150 mg/kg and resulted in a 72% life span increase, i.e., a
16.3 days survival time, in mice with leukemia. The com-
without preventing the Pt from exhibiting its anticancer ac-
parison with cisplatin reveals a slightly higher activity of the
tivity.
established anticancer drug (83% and 17.4 days at 10
Recently Chen et al. synthesized the cisplatin analogous mg/kg). The D-galactose and D-ribose compounds were re-
glucose-Pt compound (Fig. (10); 37 sugar = glucose] and di- ported without biological data [111].
and trisaccharide derivatives with a 2-hydroxy-1,3- H2
diaminopropane linker, with the in vitro antitumor activity of N
Cl
37 being comparable to that of cisplatin against human ovar- Pt
ian cancer A2780S and melanoma cancer MeWo cell lines. Cl
No activity against the cisplatin-resistant variant A2780cP NH
O
was observed. As far as we are aware no biological data
concerning the di- and trisaccharide complexes has been O
O
reported [106,107].
O
O O
NH2 Cl
sugar-C1
O Pt
Cl H2N NH2 39
sugar-C1 NH2
Pt
Cl Cl Fig. (11). Structure of cis-dichlorido[N-(6-deoxy-1,2:3,4-di-O-
isopropylidene--D-6-galactopyranosyl)-1,2-ethylenediamine-N,N']
37 38
platinum(II) 39.
Fig. (10). Carbohydrate-pendant platinum complexes, with the Pt As far as we are aware, there is only one report dealing
center being linked via an amine linker. with cyclodextrin derivatives of Pt compounds coordinated
Shigenobu and co-workers reported analogous com- via an amine functionality [112]. However, there are many
pounds with the sugar being varied from D-glucose, D- papers describing the application of such carbohydrates for
mannose, D-galactose, D-xylose, to L-glucose (Fig. (10), 37), solving formulation problems in pharmaceutical research
coordinated to the Pt center via a (2S)-2,3-diamino-1- [57]. PtCl2(NH3) or PtCl2 fragments coordinated to amino-
propanol linking ligand (Fig. (10), 38). These sugar- and ethylenediamino-- and -cyclodextrins administered to
appended platinum(II) complexes were found to be active mice bearing a standard P388 tumor model gave a maximal
against P388 cells implanted in mice [108]. Significant dif- T/C of 121% [112].
ferences between - and -D-galactose anomers were ob-
served. The former exhibited a T/C value of 194% at 400 2.2. Ligands with O Donors Capable of Releasing the Pt
mg/kg dose, whereas the latter exhibited low toxicity at the Center
same dose. Comparison of D- and L-glucose reveals a higher 2.2.1. Carboxylate Ligands
toxicity of the non-natural carbohydrate, and the (2S)-2,3-
diamino-1-propanol linked compounds show higher toxicity In view of the reported cytotoxicity of Pt(II) complexes
than the 1,3-diamino-2-propanol derivatives. For all the containing oxaliplatin-type chxn ligands against murine
studied compounds, the carbohydrate moiety was found to leukemia L1210 [70,113], Kidani et al. reported the synthe-
reduce the toxicity significantly in comparison to cisplatin sis of platinum(II) complexes with D-glucuronate, D-
(LD50 10 mg/kg) [69]. It is supposed that the uptake into gluconate, 1,2,3,4-tetra-O-acetyl-- or -D-glucuronate and
cancer cells, and the differing intracellular processing may 2,3,4,6-tetra-O-acetyl-D-gluconate as leaving groups.
be responsible for the differences in the antitumor activity of The D-glucuronato compounds of both chxn and 2-
this complex class [108]. The same group reported the (aminomethyl)cyclohexylamine 40 [amcha; Fig. (12))
analogous palladium(II) complexes with their profiles differ- ligands were found to be water soluble and were very active
ing significantly from the platinum analogues [109]. Bio- against the P388 leukemia tumor model. Notably, similar to
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2581

NH2
-OOC OH OH O
HO O
HO OH HO
O-
OH
H2N OH OH

40 41 42

Fig. (12). Structural formulae of amcha 40, D-glucuronate 41, and D-gluconate 42.

oxaliplatin, a dependence on the conformation of the chxn physiological conditions with PtCl(D-glucuronato)[(1R,2R)-
ligand was shown, and both the trans- and cis-(1R,2R) com- chxn] showing half-lives of 56 and 52 min in water and
pounds were more active than the (1S,2S)-enantiomers. In physiological saline solution, respectively [118]. However,
the case of the amcha ligands, the D-glucuronato complexes the compound was highly active against L1210 and its cis-
with the cis-d and the trans-d ligands were more active than platin-resistant variant, whereas no tumor-inhibiting poten-
the l-analogues. From this study the amcha complexes ap- tial was observed against Lewis lung carcinoma and B16
pear to be more active and of lower general toxicity than the melanoma models. In contrast, the related complex with a
chxn compounds. In combination with their high water solu- nitrato leaving group, i.e., Pt(D-glucuronato)(NO3)[(1R,2R)-
bility, these compounds were considered suitable for further chxn], only displayed marginal activity against these models,
development. Compounds with other carbohydrate-based demonstrating the importance of the appropriate selection of
leaving groups, i.e., D-glucuronate 41, D-gluconate 42 (Fig. co-ligands.
(12)) as well as acetylated D-glucuronate and D-gluconate,
Talebian et al. synthesized and examined the bone mar-
were found to exhibit antitumor activity in mice against the
row cytotoxicities of platinum(II) complexes in which the
murine L1210 tumor. The anticancer activity was found to
sugar is attached to the Pt center via a malonic acid bridge
be higher for the more lipophilic compound Pt(1,2,3,4-tetra-
(Fig. (13)) [119]. The non-acetylated compound 43a was
O-acetyl--D-glucuronate)2[(1R,2R)-chxn] in comparison to found to be much more water soluble than 43b and was
Pt(D-gluconato)(NO3)(cis-amcha) (T/C values of 388 and
tested for antitumor activity in P388 leukemia, L1210 and its
317%, respectively, at a dose of 50 mg/kg) [114,115]. The
cisplatin resistant variant L1210/DDP. Compound 43a was
comparison of these compounds is perhaps not appropriate
found to exhibit activity similar to cisplatin and carboplatin,
due to their structural dissimilarity. The galactose analogue
but with reduced nephrotoxicity and less leucopenia than the
Pt(galacturonate)2[(1R,2R)-chxn] was tested against L1210
established drug compounds [120].
leukemia in vitro and in vivo and resulted in a T/C value of
238% at a single administration of 12.5 mg/kg [116]. In By linking a glucosamine moiety via an L-aspartate
addition, the latter compound and its analogous sacchara- linker, compound 44 (Fig. (13)) was obtained [121]. How-
tolactonato and saccharato complexes were shown to be ever, at a quite high dosage of 400 mg/kg no advantage in
active in the cisplatin-resistant variant of the L1210 cell line, comparison to cisplatin was observed for the treatment of
with the saccharato complex being more active in the cis- P388 leukemia in mice, and raising the dose to 800 mg/kg
platin-resistant cells compared to normal L1210 cells [117]. resulted in death of test mice.
This suggests that the biochemical mechanisms of cisplatin Compound 45 (Fig. (14)) was selected as a lead structure
resistance, including detoxification and repair mechanisms, from a series of compounds having a malonate-derivative as
do not affect such compounds. Comparing the in vivo activ- linker connecting the sugar unit to the Pt moiety [122]. Glu-
ity of the latter complexes revealed at dosages of 25–50 cose, galactose and mannose, as well as their 6- and 1-amino
mg/kg T/C values of about 220%, as also observed for cis- forms and the acids were all attached to the platinum center
platin. However, for cisplatin a lower number of long term using this linker. The in vitro and in vivo cytostatic activity
survivors was observed (1 in 6 for cisplatin vs. 4 in 6 for the of the platinum complexes was determined against a variety
galacturonato complex), and the sugar complexes were of of human cancer cell lines, including breast, cervix, prostate
markedly lower general toxicity. and small cell lung cancer cells. None of the tested com-
Compounds of the latter type, viz. with the sugar ligand pounds were remarkably active in all tumor models, although
coordinated via a carboxylate, were not very stable under 45 was more active than the established drugs in human

AcO
AcO O
H2N H2N
O O AcO S O O
OR OR O Pt AcHN Pt
N HN N
H2 H2
RO O N O
N H
H
OR OR O O

43 44

Fig. (13). Structures of gluconate (43a, R = H; 43b, R = acetyl) and glucosamine-derived complexes with anticancer activity.
2582 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

OH

O HO O
HO OH
O NH
O H2 OH H2N O
HO O O O
N Pt
NH Pt N
HO O O O H
H2N HO N O H2 O
O OH Pt NH3
HO OH O O O O
OH NH3
46 47
45

Fig. (14). Malonate-linked sugar-Pt conjugates.

TUR myeloid leukemia cells, and 46 showed the best activ- the anticancer activity against human hepatoma cells HepG2
ity in the cervix and prostate cancer cell lines. Studies on the with the same compound being linked only to a single galac-
effect of inhibiting sugar transport revealed a high depend- tose moiety (50) and to the analogue without sugar units. The
ence for 45, whereas the activity of cisplatin was not ef- cytotoxicity was in the same dimension as observed for cis-
fected. In an in vivo study against several tumor models, 45 platin and 49 was found to be the most cytotoxic agent, pos-
proved an effective inhibitor of tumor growth. Notably, 45 sibly due to a mediation of the cellular uptake by galactose
was highly tolerated with a promising therapeutic index. For receptors expressed on the surface of the cells [126,127].
the similar glucosamine derivative, 47, uptake was studied However, no data on cellular uptake was provided.
but no other biological data was reported [123].
Malonate modified dextran was linked to cisplatin-
Reedijk and co-workers designed the -glucuronyl- [128,129] and oxaliplatin-type [130,131] fragments. The
platinum conjugate 48 (Fig. (15)) with an (1R,2R)- former compound was tested in vivo in mice bearing Colon
cyclohexane-1,2-diamine (chxn) ligand, with the intention 26 mouse cancer cells and was shown to be more effective
that the enzyme -glucuronidase can cleave the sugar-benzyl than cisplatin. The platinum content in the organs remained
bond inducing the release of the active drug moiety [124]. high for at least 24 hours after injection, with no accompany-
The platinum complex was characterized as Pt(chxn)(4- ing side-effects observed. The oxaliplatin-type complex with
hydroxybenzylmalonate) after enzymatic cleavage, demon- a chxn ligand was investigated against p388D1 lymphocytic
strating the concept, but further biological data has not been leukemia cells in vitro and was as active as the dichlorido
reported. precursor. The stability of the administered conjugate in
serum was found to be high [130], whereas half of the cis-
NaOOC platin immobilized on the dextran was released within 48
H2N
HO O
O O
hours in physiological phosphate buffer solution [132].
HO O
Pt N Functionalization of dextran and conjugation to platinum
OH H2 moieties resulted, as envisaged, in a longer circulation time
O
and higher serum levels. The extent to which the compounds
O are retained is dependent on the size of the compound. In
48 addition, the compounds were less prone to protein binding
than cisplatin and its aqua analogue [133]. Some of the com-
Fig. (15). Structure of the -glucuronyl-platinum(II) conjugate 48,
pounds were tested in vitro against murine and human tumor
activated by -glucuronidase.
models and were of similar or lower activity in comparison
2.2.2. Dextran and Macromolecular Compounds to the drug precursors. No correlation between activity and
Employing a similar strategy as for the compounds de- the molecular weight of the carrier was observed [134,135].
scribed in section 2.2.1, i.e., employing a malonato group to In contrast, in vivo the conjugates were more active than
link the sugar moiety to a Pt center, resulted in a series of cisplatin and also the size of the carrier was shown to influ-
macromolecular drugs with antibodies, polymers or oligo- ence the activity, with the higher molecular weight conjugate
saccharide carriers linked covalently to (typically) more than being the most active. Repeated injections led to 100% sur-
one platinum center. Such carrier units attached to drugs vival of test animals. The binding of the metal fragment is,
have been found to facilitate the delivery of drugs to the however, reversible upon reaction with biological target
target tissue. Moreover, such platinum conjugates were molecules such as DNA. In another study dextran and inulin
found to have superior water solubility properties and also were modified in a similar manner, but only minor antican-
introduce a certain degree of inertness to the platinum center cer activity was observed in human or murine leukemia, lung
with regard to reaction with biomolecules in the blood- and colon cancer models [136].
stream, which has been proposed to lead to the deactivation Dextran and starch were modified by malonic anhydride
of Pt compounds or even to species exhibiting unwanted and reacted in situ to form monodentate platinum complexes
side-effects [125]. of cisplatin- and oxaliplatin-type [137-139]. T/C values of
Ohya et al. reported the synthesis of cisplatin-polymer 329% were reached against L1210 leukemia in mice, with
conjugates to 4 galactose units 49 (Fig. (16)) and compared some of the mice even cured, an effect which could not be
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2583

OH
OH OH

O
OH H H
HO O
HO N O N O
OH O O O
HO 3 H H
O N O N
OH NH N O O
OH H n
OH
O O O Pt NH3
O O O
OH H NH3
HO O NH
HO N O
OH
HO 3
O

OH
OH OH

O
OH H H
HO O
HO N O N O O
OH O
HO 3
O NH
OH NH
OH OH
O
O
OH H
HO O NH
HO N O
OH
HO 3
O

49
OH
OH OH

O
OH H H NH3
HO O
HO N O N O O O
OH O Pt NH
HO 3 H 3
O N O
N N
H H
O O
50

Fig. (16). Galactose-based macromolecular Pt conjugates.

achieved by treatment with cisplatin. Similar to the previ- der bloodstream conditions and a strong influence on the
ously described carbohydrate complexes, much higher doses systemic clearance and distribution of platinum into tissues
than that required for cisplatin were applicable. [143].
Oxidized polysaccharides have been attached to platinum 2.2.3. Organometallic Compounds
centers [140]. Amylose and dextran of different molecular
Partly inspired by the finding of Lowe and coworkers,
weights, amylopectin, polygalacturonic acid and alginic acid who reported that platinum(II) complexes of 4’-substituted
were thereby linked to a Pt(chxn) moiety. In general, at low
2,2’:6’,2’’-terpyridine were cytotoxic to human ovarian
doses excellent results were obtained against L1210 leuke-
carcinoma cells [144] as well as to Trypanosoma and
mia in mice with T/C values up to 357% (for cisplatin 175%)
Leishmania parasites [145], Ma et al. synthesized water
and 5 of 6 mice cured. Screening the amylose compound,
soluble luminescent platinum terpyridine complexes with
which was the most active against L1210, in solid tumor
glycosylated acetylide and arylacetylide ligands and studied
models revealed up to complete inhibition of tumor growth. their DNA binding properties [146,147]. Spectroscopic stud-
Schechter and colleagues followed a different approach ies showed that the Pt(II)-glycosylated acetylide or arylacet-
by linking the cytotoxic platinum moiety in a similar manner ylide moieties remain intact in aqueous solution for 72 hours
as described above to a monoclonal antibody directed against at room temperature. The compounds were found to bind to
the epidermal growth factor receptor which is over-expressed DNA in an intercalative manner, as indicated by spectropho-
on KB cells [141]. The carrier-conjugated drug was still tometric studies, and the substitution pattern of the terpyri-
capable of interacting with DNA and was pharmacologically dine ligand was shown to influence the extent of intercala-
active against tumor cells. Notably, the antibody conjugation tion. The complexes with arylacetylide (51) and glycosylated
and also biotinylation proved to be important for anticancer acetylide (such as 53) ligands were tested for their cytotoxic-
activity. By using antibodies specifically reactive to B lym- ity against human carcinoma cell lines in comparison to
phoma cells preferential cytotoxicity to this cell type was cisplatin and the non-glycosylated arylacetylide derivative
observed [142], albeit at maintained level as for the parent 52. The most active compound was identified as 51 (Fig.
inorganic drug molecules. The conjugation of platinated (17)), which is approximately 10-times more active than the
dextranes with antibodies resulted in stable compounds un- non-glycosylated compound 52 and one order of magnitude
2584 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

t-Bu t-Bu

N OAc N N OAc
AcO OAc AcO
O OAc
t-Bu N Pt O N Pt t-Bu N Pt O O
OAc OAc
N N N

51 t-Bu 52 53 t-Bu

Fig. (17). Organometallic terpyridine-Pt complexes.

Table 4. IC50 Values (M) of 51–53 in Human Carcinoma Cell Lines (HeLa, HepG2, SF-268, NCl-H460, MCF-7) and Normal 293
Cells in Comparison to Cisplatin [146,147]

Complex Cell linesa

HeLa HepG2 SF-268 NCI-H460 MCF-7 293

51 0.1 ± 0.03 0.1 ± 0.01 0.06 ± 0.02 0.1 ± 0.03 0.08 ± 0.04 0.5 ± 0.1
52 2.7 ± 0.7 3.0 ± 1.1 2.1 ± 0.8 2.5 ± 0.8 3.4 ± 1.0 4.6 ± 1.2
53 17.8 ± 0.5 22.9 ± 0.8 17.1 ± 0.4 28.5 ± 1.9 17.1 ± 0.4 50.3 ± 7.2
cisplatin 11.6 ± 0.2 20.6 ± 1.9 15.6 ± 0.2 25.1 ± 3.4 19.1 ± 1.7 > 100
a
HeLa, cervix carcinoma; HepG2, hepatocellular carcinoma; SF-268, human glioma; NCI-H460, lung cancer; MCF-7, breast adenocarcinoma; 293, non-tumorigenic kidney cell line.

more potent than the acetylide complex 53, the latter being as shown for similar aminoalcohol complexes [150]. Fur-
slightly less active than cisplatin. All the compounds showed thermore, such a ring-opening/-closing mechanism might
the lowest activity in the non-tumorigenic kidney cell line prevent platinum complexes from undergoing reactions with
293, with cisplatin being non-toxic in this model. Further- biomolecules after intravenous administration, with protein
more, cancer cell death was induced in about 52% of the interactions possibly contributing to the toxic side-effects of
cells by apoptosis and a much lower fraction of necrosis was Pt-based drugs [151-153]. Compounds 55–57 were active
observed (ca. 4.8%). Notably, these organometallic com- against P388 leukemia with an inhibition ratio of 60% at
pounds represent rare examples of Pt complexes linked to high nM concentrations. However, detailed biological data is
sugar molecules exhibiting lower IC50 values than cisplatin not available.
in cell culture studies. The dinuclear complex 58 (Fig. (18)), in which bisphos-
phonate ligands bridge the two Pt centers, was reported to be
2.3. Pt complexes of P-Containing Sugars slightly active in sarcoma S180 ascites and L1210 leukemia
tumor models at a high single dose of 320 mg/kg [154].
In an attempt to overcome resistance problems occurring Treatment resulted in an increased life span of 28% in the
during the treatment of cancer patients with cisplatin and sarcoma mice vs. 58% for cisplatin.
other platinum complexes, P-containing sugar derivatives
have been developed. Shi and co-workers synthesized sugar
3. NON-PLATINUM CARBOHYDRATE-BASED
complexes bases on carbohydrates with a PPh2 group at the
2-position, such as in ligand 54 (Fig. (18)), or at C3 METAL COMPLEXES
[148,149]. The respective Pt complex 55 was proposed to 3.1. Auranofin Analogues
exert a potential activation/deactivation mechanism by form-
ing singly- (56) and doubly ring-closed (57) species [148] by In modern clinical protocols, auranofin [(2,3,4,6-tetra-
exploiting pH differences between tumor and normal tissue, O-acetyl-1-thio- ß- D-glucopyranosato-S)(triethylphosphine)]-

OH

HO O
OH OH OH
HO
PPh2 NH
Ph O
O O OH P Cl P Cl P O
OMe Pt Pt Pt O O
= O
P Cl P O P O P P
OH P
O O O O 58
OH Pt
Pt NH2
H2N
54 55 56 57 NH2 H2N

Fig. (18). Ring-closing Pt-phosphinesugar complexes based on ligand 54, and a bisphosphonate-based dinuclear Pt complex.
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2585

gold(I) 59, Fig. (19)) is used in the treatment of rheumatoid Shi et al. reported the synthesis and characterization of
arthritis [155-158]. Auranofin and other Au complexes [both several gold(I) complexes based on ligand 54 (Fig. (18)),
Au(I) and Au(III) based] have also been evaluated for the also used in platinum complexes (see section 2.3.), and the
treatment of cancer [147,159-163], AIDS [147], bronchial analogous compound with the PPh2 group in the 3-position
asthma, and malaria [158,164]. [165,168]. These complexes have been found to be effective
in inhibiting the growth of P388 leukemia, in particular, the
AcO
AcO O activity of Au(54)(2-mercaptobenzimidazole) was found in
AcO S the high nM range [168].
Au
OAc X
Baker, Berners-Price and co-workers reported the synthe-
Fig. (19). Structures of auranofin and selected derivatives; X = sis of organometallic Au(I) N-heterocyclic carbene com-
P(CH2CH3)3 auranofin 59, P(CH3)3P 60, P[(CH3)2CH]3 61, plexes 69 (Fig. (20)) as analogues of auranofin [169]. Such
P[(CH3)3N]3 62, P(C6H5)3 63, P(CH3CH2)2(C6H5) 64, ligand systems were found to be suitable for tuning lipophil-
P[(CH3CH2)(C6H5)2] 65, P[(CH3CH2)2(CH3CH2O)] 66, icity, and in preliminary biological studies the complexes
P(CH3CH2)2[(CH3)2CH] 67, P(CH3CH2)2[HO(CH2)4] 68. were shown to exhibit antimitochondrial activity which
could be correlated to their lipophilicity.
Auranofin was found to be active in several tumor mod-
els [159,160,162,165], affecting the mitochondrial function The dinuclear Au(I) complex 70 (Fig. (20)) proved to be
[166] and inducing apoptosis. The thioredoxin reduc- active in murine tumor models. It was shown to induce an S-
tase/thioredoxin system in the mitochondria might play a phase arrest in human promyelocytic leukemia HL-60 cells
central role in the mode of action of auranofin, with au- at low concentrations, whereas at higher concentrations a
ranofin being an inhibitor of thioredoxin reductase leading to secondary block in the G1/S transition was observed [170].
an elevated level of hydrogen peroxide in the mitochondria.
A large number of auranofin analogues with different sub- 3.2. Ru(II) and Ru(III)-Sugar Complexes
stituents (see examples in Table 5) and other sugar-based
Au(I) complexes have been reported [167] and some of them Ruthenium(III) complexes are among the most exten-
were found to exhibit potent antitumor activity in vitro and sively studied anticancer compounds (after those based on
in vivo. Replacing the triethylphosphine ligand in auranofin platinum) and the two most promising compounds KP1019
by other phosphines, phosphinites or amidophosphites did 71 and NAMI-A 72 (Fig. (21)) are currently undergoing
not significantly alter the antitumor activity in B16 cells and clinical trials [28,29,171]. Their proposed mode of action is
in the P388 leukemia tumor model. A similar observation different from that of Pt anticancer agents, thus Ru com-
was made on replacing the tetraacetylglucose moiety by pounds might extend the range of treatable tumors. In gen-
glucose (both  and ), galactose and xylose. eral, Ru complexes are less generally toxic and therefore less
prone to inducing unwanted side-effects in patients, a major

Table 5. Biological Activity of Auranofin and Related Analogues In Vitro in B16 Mouse Melanoma Cells and In Vivo Against P388
Leukemia in Mice [167]

Compound B16, IC50 / μmol/kg MTD / μmol/kg P388, ILSmax / %

59 (auranofin) 1.5 18 70
60 2 9 45
61 4 14 46
62 2 8 60
63 4 7 36
64 2 13 55
65 4 6 32
66 1 14 70
67 2 17 90
68 8 17 58

AcO OH
AcO O HO
R O HO
AcO S HO OH
Au N HO S S O
OAc Au Au
OH Ph2P Ph2P OH
N
R
69 70

Fig. (20). Structures of organometallic Au(I) N-heterocyclic carbene compounds, and a dinuclear Au complex.
2586 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

HN +
H H R
NH R
N N N N
Cl Cl HN Cl Cl O
Ru Ru Ru
Cl Cl Cl Cl
HN Cl Ru N
H2N Cl Ru
O
Cl P Cl P
N NH2 Cl O
S
HN O N O
N
OR2
OR1
71 72 73 74 75

Fig. (21). Ruthenium complexes of interest for anticancer research.

draw-back in Pt-based chemotherapy. Their low toxicity is Following a different approach by using Ru-sugar com-
thought to stem from the selective transport into the tumor plexes for imaging purposes, a -emitting ruthenocene moi-
via the transferrin pathway [152,172-174] and reduction of ety (103Ru) was linked to sugar residues, in particular ami-
Ru(III) species to Ru(II) in the cell, leading to their activa- nosorbit, tetraacetylglucosamine, aminomannit and ami-
tion [175,176]. Recently, organometallic Ru(II)-arene com- nodulcit derivatives were prepared [194]. The compounds
pounds were found to exhibit potential as anticancer agents, were observed to be highly stable in vivo and after applica-
and such compounds were found to be active both against tion to mice bearing Ehrlich’s ascites tumor, it was found
primary tumors (74) and metastasis (73) [36,37,177-187]. that the compounds accumulated in the liver and kidneys
A large number of ruthenium complexes with different [195]. However, excretion was markedly accelerated for the
sugar compounds in comparison to the administration of
ligands have been synthesized and tested for their anticancer 103
RuCl3.
properties, although only a few carbohydrate complexes with
anticancer activity have been reported.
3.3. Metallocene-Based Compounds
In particular, a bleomycin-Ru(III) complex was reported
to be ineffective in DNA strand scission whilst exhibiting Titanocene dichloride 76 (Fig. (22)) was introduced as a
anticancer activity against murine leukemia L1210 cells, putative anticancer drug at the end of the 1970’s and entered
with a similar magnitude to cisplatin [188]. It seems likely clinical trials in 1993 [46,196]. Nephrotoxicity was identified
that DNA strand cleavage is not responsible for the in vitro as dose-limiting, but the absence of bone marrow toxicity
toxicity and some other mechanism is in operation. was thought to be promising and clinical phase II trials were
initiated. In these studies, setup as a single agent therapy, no
Recently, organometallic Ru(II)-arene complexes of the improved effects in comparison to other treatment regimens
general formula 75 (Fig. (21)) with phosphite-carbohydrate were observed and accordingly the development was abon-
ligands have been reported [189-191], and the compounds doned.
were found to be moderately active against human colon
adenocarcinoma (SW480), ovarian carcinoma (CH1, A2780 There are only a few examples in literature of sugar-
and cisplatin-resistant A2780), lung carcinoma (A549), derived titanium compounds with biological data [197,198].
melanoma (Me300), glioblastoma (LNZ308) and HCEC One appears to be a titanocene-cyclodextrin derivative with
endothelial cells in vitro [192], as was also observed for the titanocene group bridging two cyclodextrin moieties
certain RAPTA-type complexes and NAMI-A [178,193]. [197]. The compound was observed to exhibit potent cyto-
Similar IC50 values in A2780 and the cisplatin-resistant phe- toxicity against the human MCF-7 breast cancer cells and
notype suggest a different mode-of-action as compared to was significantly more active than titanocene dichloride.
cisplatin. Importantly, the lowest activity was found against Another series of Ti compounds bearing either one (77) or
the non-tumoral endothelial cell line, indicating selectivity two (78) ribofuranose moieties was tested and compared to
for cancer cells. cisplatin, titanocene dichloride and titanocene bisbenzoate
against L929 mouse fibroblasts, A-431 epithelial carcinoma,

OMe OH
O
OMe
O HO O
MeO Cl O O
O Ti
Cl Cl O O O O
Cl OMe
Ti Ti
O O a R=H
Cl Cl
b R = CH3 R R
O O Fe
R R
R R
76 77 78 79

Fig. (22). Organometallic complexes of interest for anticancer research.


Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2587

A-498 renal cancer and KB 3.1 cervix carcinoma cell lines patent literature, which could also hamper scientific pro-
[198]. Titanocene 78a appeared to be the most active com- gress.
pound of the series, exhibiting similar activity as cisplatin in
When comparing the type of linkages used to connect
L929 and KB 3.1 cells. Furthermore, the synthesis of several
metal complexes to sugars there are two different classes.
other sugar-derived Ti and also Zr compounds was reported,
First, where the metal center is linked to the carbohydrate via
but no biological data was published [199-204].
a group capable of releasing the cytotoxic moiety and the
Ferrocene derivatives have gained significant interest in sugar being mainly responsible for the transport, and can be
medicinal application, largely motivated by the pioneering considered a carrier molecule. Second, in complexes with
work of Jaouen and co-workers on the tamoxifen derivatives aminosugars, the sugar moiety should have a significant
of ferrocene [205] and by the development of ferroquine as influence on the anticancer activity, and if DNA is consid-
an antimalarial [206]. Itoh et al. have reported on the synthe- ered the ultimate target, on the structural changes induced in
sis of a series of ferrocene-conjugated glucose derivatives DNA.
such as 79 and tested them in a mouse cancer cell line
Although carbohydrate-metal compounds have been
(FM3A) and also against the malaria parasite P. falciparum
shown to exhibit cytostatic activities in several cases, many
[207,208]. Only moderate activity was observed against this
facets remain to be explored. Future research should cer-
cancer model and against other human tumor cell lines, in-
tainly include more detailed studies on the uptake of sugar
cluding 41M, CH1 (both ovarian carcinoma), SK-BR-3
compounds mediated by sugar transporters, since this argu-
(mammarycarcinoma), and SW480 (colon carcinoma) [209].
mentation is frequently used as a justification, and the me-
tabolization of such compounds in biological environment.
4. CONCLUSIONS AND OUTLOOK Also the type of linker, the ligand sphere of the cytotoxic
moiety, the release of the metal center, etc. need fine tuning,
Carbohydrates and their derivatives are the constituents and offer scope for further investigations. Finally, it would
of many natural antibiotics, including bleomycin, anthracy- be interesting to see the performance of a carbohydrate-
clines, neocarzinostatin chromophore, and other DNA- based metal complex in clinical trials as some described in
targeting agents. Sugar compounds such as glucose are re- this review appear to be significantly superior to the existing
quired to a higher degree by tumors, indicated by the over- chemotherapeutic agents currently available.
expression of glucose receptors on the cell surface, offering a
degree of selective transport to cytotoxic compounds at-
5. ACKNOWLEDGEMENTS
tached to carbohydrates. This requirement is also related to
the up-regulation of the glycolytic activity for energy pro- The authors are indebted to the Higher Education Com-
duction (due to lower nutrition supply because of slow mission of Pakistan, the Austrian Exchange Service, the FFG
neovascularization), one of the major metabolic alterations in – Austrian Research Promotion Agency (811591), the Aus-
cancer cells [210,211]. trian Council for Research and Technology Development
Linking metal complexes to sugars usually results in im- (IS526001), the FWF – Austrian Science Fund (J2613-N19),
proved solubility in biologically compatible media (com- the Swiss National Science Foundation (individual short
pared to established anticancer drugs and drug candidates in visit, No. 120985) and COST D20 and D39 for financial
advanced stages of development). They are suitable for in- support.
travenous application, and result in compounds with a re-
duced general toxicity, which might be related to a type of ABBREVIATIONS
targeted approach. This feature allows the drug candidates to
be applied at much higher doses and endows them with a ADEPT = antibody-directed enzyme prodrug therapy
broader therapeutic window. Notably, many examples have Amcha = 2-(aminomethyl)cyclohexylamine
been reported with higher anticancer activity, both in vitro
and in vivo, than standard chemotherapeutics, but no clear- chxn = cyclohexane-1,2-diamine
cut structure-activity relationships were found. Also, the dGMP = 2-deoxyguanosine 5’-monophosphate
mechanism of action of the sugar complexes is not clear yet.
For platinum complexes a similar mode of action as for the ED = effective dose
compounds in clinical routine is assumed, whereas not much IC50 = 50% inhibitory concentration
is known about the targets for non-platinum complexes. ID50 50% inhibitory dose
=
As far as we are aware metal-carbohydrate compounds i.p. = intraperitoneal
have not yet entered clinical trials, although for example,
chxn-Pt sugar complexes were more active than oxaliplatin ILS = increased life span
itself. The lack of clinical data might be at least in part due to T/C = treated to control
higher preparative demands in comparison to the relatively
simple benchmark chemotherapeutics. However, carbohy-
6. REFERENCES
drate chemistry is extensively developed, but frequently
includes multi-step synthetic procedures, which is a draw- [1] Fight against cancer, WHO, 2007.
back for commercialization of such compounds. Further- [2] Marks, G.; Beatty, W.K. The Precious Metals of Medicine, Charles
more, a lot of the data in this field is found exclusively in the Scribner's Sons: New York, 1975, pp. 294.
[3] Peyrone, M. Annalen der Chemie und Pharmazie, 1844, 51, 1.
2588 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

[4] Lippert, B. (Ed.) Cisplatin: Chemistry and Biochemistry of a [38] Peacock, A.F.A.; Habtemariam, A.; Fernandez, R.; Walland, V.;
Leading Anticancer Drug, Helvetica Chimica Acta / Wiley-VCH: Fabbiani, F.P.A.; Parsons, S.; Aird, R.E.; Jodrell, D.I.; Sadler, P.J.
Zürich / Weinheim, 1999, pp. 576. J. Am. Chem. Soc., 2006, 128, 1739.
[5] Rosenberg, B.; VanCamp, L.; Trosko, J.E.; Mansour, V.H. Nature, [39] Easmon, J. Expert Opin. Ther. Pat., 2002, 12, 789.
1969, 222, 385. [40] Wang, T.; Guo, Z. Curr. Med. Chem., 2006, 13, 525.
[6] Rosenberg, B. Interdiscip. Sci. Rev., 1978, 3, 134. [41] Nguyen, A.; Vessieres, A.; Hillard, E.A.; Top, S.; Pigeon, P.;
[7] Oliver, T.; Mead, G. Curr. Opin. Oncol., 1993, 5, 559. Jaouen, G. Chimia, 2007, 61, 716.
[8] Stathopoulos, G.P.; Rigatos, S.; Malamos, N.A. Oncol. Rep., 1999, [42] Köpf-Maier, P.; Leitner, M.; Voigtlander, R.; Köpf, H. Z. Natur-
6, 797. forsch., C: Biosci., 1979, 34, 1174.
[9] Galanski, M.; Arion, V.B.; Jakupec, M.A.; Keppler, B.K. Curr. [43] Messori, L.; Gabbiani, C. In Metal Compounds in Cancer Chemo-
Pharm. Design, 2003, 9, 2078. therapy; Perez, J.M.; Fuertes, M.A.; Alonso, C., Eds., Research
[10] Stathopoulos, G.P.; Boulikas, T.; Vougiouka, M.; Deliconstantinos, Signpost: Trivandrum, 2005; 355.
G.; Rigatos, S.; Darli, E.; Viliotou, V.; Stathopoulos, J.G. Oncol. [44] Al-Allaf, T.A.K.; Rashan, L.J. Boll. Chim. Farm., 2001, 140, 205.
Rep., 2005, 13, 589. [45] Gao, E.; Cheng, M.; Wang, K. Zhonghua Yiyao Zazhi, 2006, 6,
[11] Rosenberg, B. Adv. Exp. Med. Biol., 1977, 91, 129. 143.
[12] Hill, J.M.; Speer, R.J. Anticancer Res., 1982, 2, 173. [46] Köpf, H.; Köpf-Maier, P. Angew. Chem., Int. Ed. Engl., 1979, 18,
[13] Galanski, M.; Jakupec, M.A.; Keppler, B.K. Curr. Med. Chem., 477.
2005, 12, 2075. [47] Keppler, B.K.; Friesen, C.; Vongerichten, H.; Vogel, E. In Metal
[14] O'Dwyer, P.J.; Stevenson, J.P.; Johnson, S.W. Drugs, 2000, 59, 19. Complexes in Cancer Chemotherapy; Keppler, B.K., Ed., VCH:
[15] Jakupec, M.A.; Galanski, M.; Keppler, B.K. Rev. Physiol. Bio- Weinheim, 1993; 297.
chem. Pharmacol., 2003, 146, 1. [48] Storr, T.; Thompson, K.H.; Orvig, C. Chem. Soc. Rev., 2006, 35,
[16] Graham, J.; Muhsin, M.; Kirkpatrick, P. Nat. Rev. Drug Discovery, 534.
2004, 3, 11. [49] Steinborn, D.; Junicke, H. Chem. Rev., 2000, 100, 4283.
[17] Giatromanolaki, A.; Stathopoulos, G.P.; Koukourakis, M.I.; Riga- [50] Hanessian, S. (Ed.) Preparative Carbohydrate Chemistry, Marcel
tos, S.; Vrettou, E.; Kittas, C.; Fountzilas, G.; Sivridis, E. Am. J. Dekker Inc.: New York, 1986, pp. 650.
Clin. Oncol., 2001, 24, 222. [51] Nangia-Makker, P.; Conklin, J.; Hogan, V.; Raz, A. Trends Mol.
[18] Stathopoulos, G.P.; Veslemes, M.; Georgatou, N.; Antoniou, D.; Med., 2002, 8, 187.
Giamboudakis, P.; Katis, K.; Tsavdaridis, D.; Rigatos, S.K.; Dimi- [52] Morel, F.; Renoux, M.; Lachaume, P.; Alziari, S. Mutat. Res.,
troulis, I.; Bastani, S.; Loukides, S.; Vergos, K.; Marossis, K.; Fundam. Mol. Mech. Mutagen., 2008, 637, 111.
Grigoratou, T.; Kalatzi, E.; Charalambatou, M.; Paspalli, A.; [53] Chiorean, E.G.; Dragovich, T.; Hamm, J.; Langmuir, V.K.; Kroll,
Michalopoulou, P.; Stoka, M.; Gerogianni, A. Ann. Oncol., 2004, S.; Jung, D.T.; Colowick, A.B.; Tidmarsh, G.F.; Loehrer, P.J.
15, 1048. Cancer Chemother. Pharmacol., 2008, 61, 1019.
[19] Veslemes, M.; Antoniou, D.; Georgatou, N.; Giamboudakis, P.; [54] Fornari, F.A.; Randolph, J.K.; Yalowich, J.C.; Ritke, M.K.;
Dimitroulis, J.; Katis, K.; Stathopoulos, G.P. Anticancer Res., Gewirtz, D.A. Mol. Pharmacol., 1994, 45, 649.
2005, 25, 2991. [55] Gyurcsik, B.; Nagy, L. Coord. Chem. Rev., 2000, 203, 81.
[20] Androulakis, N.; Syrigos, K.; Polyzos, A.; Aravantinos, G.; [56] Dieguez, M.; Pamies, O.; Claver, C. Chem. Rev., 2004, 104, 3189.
Stathopoulos, G.P.; Ziras, N.; Mallas, K.; Vamvakas, L.; Geor- [57] Uekama, K.; Hirayama, F.; Arima, H. In Cyclodextrins Their
goulis, V. Cancer Investig., 2005, 23, 9. Complexes; Dodziuk, H., Ed., Wiley-VCH: Weinheim, 2008; 381.
[21] Aravantinos, G.; Fountzilas, G.; Kosmidis, P.; Dimopoulos, M.A.; [58] Kuduk-Jaworska, J.; Jezowska-Trzebiatowska, B. Inorg. Chim.
Stathopoulos, G.P.; Pavlidis, N.; Bafaloukos, D.; Papadimitriou, C.; Acta, 1986, 123, 209.
Karpathios, S.; Georgoulias, V.; Papakostas, P.; Kalofonos, H.P.; [59] Haroutounian, S.A.; Georgiadis, M.P.; Bailar, J.C. Inorg. Chim.
Grimani, E.; Skarlos, D.V. Ann. Oncol., 2005, 16, 1116. Acta, 1986, 124, 137.
[22] Wong, E.; Giandomenico, C.M. Chem. Rev., 1999, 99, 2451. [60] Johannesen, S.A.; Petersen, B.O.; Duus, J.O.; Skrydstrup, T. Inorg.
[23] Fuertes, M.A.; Castilla, J.; Alonso, C.; Perez, J.M. Curr. Med. Chem. (Washington, DC, U. S.), 2007, 46, 4326.
Chem., 2002, 2, 539. [61] Beck, W.; Thiel, G. cis-Platinum(II) complexes with saccharide
[24] Ho, Y.P.; Au-Yeung, S.C.F.; To, K.K.W. Med. Res. Rev., 2003, 23, derivatives as ligands. DE Patent 3108842, 1982.
633. [62] Thiel, G.; Beck, W. Z. Naturforsch., B: Anorg. Chem., Org. Chem.,
[25] Reedijk, J. Proc. Natl. Acad. Sci. USA, 2003, 100, 3611. 1983, 38B, 1081.
[26] Barnes, K.R.; Lippard, S.J. In Metal Ions in Biological Systems; [63] Nagel, Y.; Beck, W. Z. Naturforsch., B: Anorg. Chem., Org.
Sigel, A.; Sigel, H., Eds., Marcel Dekker Inc.: New York, 2004; Chem., 1985, 40B, 1181.
Vol. 42, 143. [64] Pill, T.; Polborn, K.; Beck, W. Chem. Ber., 1990, 123, 11.
[27] Jakupec, M.A.; Galanski, M.; Arion, V.B.; Hartinger, C.G.; Kep- [65] Bitha, P.; Child, R.G.; Hlavka, J.J.; Lin, Y.I. Platinum complexes.
pler, B.K. Dalton Trans., 2008, 183. EP Patent 85-116010, 1986.
[28] Rademaker-Lakhai, J.M.; Van Den Bongard, D.; Pluim, D.; Bei- [66] Bitha, P.; Child, R.G.; Hlavka, J.J.; Lin, Y.I. Preparation of tetra-
jnen, J.H.; Schellens, J.H.M. Clin. Cancer Res., 2004, 10, 3717. hydro-3,4-furandiamine platinum complexes and related com-
[29] Hartinger, C.G.; Zorbas-Seifried, S.; Jakupec, M.A.; Kynast, B.; pounds as antitumor agents. EP Patent 237829, 1987.
Zorbas, H.; Keppler, B.K. J. Inorg. Biochem., 2006, 100, 891. [67] Tsubomura, T.; Yano, S.; Kobayashi, K.; Sakurai, T.; Yoshikawa,
[30] Kostova, I. Curr. Med. Chem., 2006, 13, 1085. S. J. Chem. Soc., Chem. Commun., 1986, 6, 459.
[31] Ang, W.H.; Dyson, P.J. Eur. J. Inorg. Chem., 2006, 4003. [68] Yoshikawa, S.; Yano, S.; Tsubomura, T. Antitumor hexose-
[32] Lovejoy, D.B.; Richardson, D.R. Expert Opin. Invest. Drugs, 2000, platinum complexes. JP Patent 62059294, 1987.
9, 1257. [69] Tsubomura, T.; Ogawa, M.; Yano, S.; Kobayashi, K.; Sakurai, T.;
[33] Arion, V.B.; Jakupec, M.A.; Galanski, M.; Unfried, P.; Keppler, Yoshikawa, S. Inorg. Chem., 1990, 29, 2622.
B.K. J. Inorg. Biochem., 2002, 91, 298. [70] Noji, M.; Okamoto, K.; Kidani, Y.; Tashiro, T. J. Med. Chem.,
[34] Jakupec, M.A.; Keppler, B.K. In Metal Ions in Biological Systems; 1981, 24, 508.
Sigel, A.; Sigel, H., Eds., Marcel Dekker Inc.: New York, 2004; [71] Berger, I.; Nazarov, A.A.; Hartinger, C.G.; Groessl, M.; Valiahdi,
Vol. 42, 425. S.-M.; Jakupec, M.A.; Keppler, B.K. ChemMedChem, 2007, 2,
[35] Mohammadi, K.; Thompson, K.H.; Patrick, B.O.; Storr, T.; Mar- 505.
tins, C.; Polishchuk, E.; Yuen, V.G.; McNeill, J.H.; Orvig, C. J. [72] Hanessian, S. (Diaminotetrahydropyran)dihaloplatinum compounds
Inorg. Biochem., 2005, 99, 2217. as anticancer agents and their preparation. GB Patent 2210039,
[36] Dorcier, A.; Dyson, P.J.; Gossens, C.; Rothlisberger, U.; Scopelliti, 1989.
R.; Tavernelli, I. Organometallics, 2005, 24, 2114. [73] Hanessian, S.; Wang, J. Can. J. Chem., 1993, 71, 886.
[37] Dorcier, A.; Ang, W.H.; Bolano, S.; Gonsalvi, L.; Juillerat- [74] Sasamori, H.; Hori, C.; Chiba, K. Preparation of platinum com-
Jeannerat, L.; Laurenczy, G.; Peruzzini, M.; Phillips, A.D.; Zano- plexes as antitumor agents. JP Patent 02134395, 1990.
bini, F.; Dyson, P.J. Organometallics, 2006, 25, 4090.
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2589

[75] Sasamori, H.; Hori, C.; Chiba, K. Pyranose platinum complex, [105] Kay, H.; Palmer, J.W.; Stanko, J.A. Platinum complexes and their
antitumor agent, and intermediate for its preparation. JP Patent therapeutic use. WO Patent 2005016946, 2005.
02223591, 1990. [106] Chen, Y.; Heeg, M.J.; Braunschweiger, P.G.; Xie, W.; Wang, P.G.
[76] Sasamori, H.; Hori, C.; Niitsuma, S.; Chiba, K. Preparation of Angew. Chem., Int. Ed. Engl., 1999, 38, 1768.
platinum complexes as antitumor agents. JP Patent 03127796, [107] Chen, Y.; Janczuk, A.; Chen, X.; Wang, J.; Ksebati, M.; Wang,
1991. P.G. Carbohydr. Res., 2002, 337, 1043.
[77] Sasamori, H.; Niitsuma, S. Preparation of 3,4-diamino-3,4- [108] Mikata, Y.; Shinohara, Y.; Yoneda, K.; Nakamura, Y.; Brudziñska,
dideoxy-L-xylopyranosides as intermediates for antitumor platinum I.; Tanase, T.; Kitayama, T.; Takagi, R.; Okamoto, T.; Kinoshita,
complexes. JP Patent 03161495, 1991. I.; Doe, M.; Orvig, C.; Yano, S. Bioorg. Med. Chem. Lett., 2001,
[78] Sasamori, H.; Hori, C.; Niitsuma, S.; Chiba, K. Preparation of 11, 3045.
platinum complexes as antitumor agents. JP Patent 04178396, [109] Brudzinska, I.; Mikata, Y.; Obata, M.; Ohtsuki, C.; Yano, S.
1992. Bioorg. Med. Chem. Lett., 2004, 14, 2533.
[79] Sasamori, H.; Niitsuma, S.; Hori, C.; Chiba, K. Preparation of [110] Talebian, A.; Green, D.C.; Hammer, C.F.; Schein, P.S. Platinum-
platinum complexes as antitumor agents. JP Patent 04312593, sugar complexes for use as neoplasm inhibitors. WO Patent
1992. 8900575, 1989.
[80] Sasamori, H.; Niitsuma, S.; Hori, K.; Chiba, K. Preparation of [111] De Almeida, M.V.; Cesar, E.T.; Fontes, A.P.S.; Felicio, E.d.C.A. J.
platinum complexes as antitumor agents. JP Patent 04224596, Carbohydr. Chem., 2000, 19, 323.
1992. [112] Matsumoto, K.; Noguchi, Y.; Yoshida, N. Inorg. Chim. Acta, 1998,
[81] Kolar, C.; Kraemer, H.P.; Dehmel, K. Platinum(II) complexes with 272, 162.
diamino sugars and their pharmaceutical compositions. EP Patent [113] Kidani, Y.; Noji, M.; Tashiro, T. Gann, 1980, 71, 637.
167071, 1986. [114] Kidani, Y.; Noji, M.; Tsukagoshi, S.; Tashiro, T. Gann, 1978, 69,
[82] Samochocka, K.; Fokt, I.; Anulewicz-Ostrowska, R.; Przewloka, 263.
T.; Mazurek, A.P.; Fuks, L.; Lewandowski, W.; Kozerski, L.; Bo- [115] Noji, M.; Hanai, M.; Omori, T.; Tashiro, T.; Suzuki, K.; Kidani, Y.
cian, W.; Bednarek, E.; Lewandowska, H.; Sitkowski, J.; Priebe, Chem. Pharm. Bull., 1988, 36, 3439.
W. Dalton Trans., 2003, 2177. [116] Khokhar, A.R.; Newman, R.A.; Krakoff, I.H. 1,2-Diaminocyclo-
[83] Kruszewski, M.; Bouzyk, E.; Oldak, T.; Samochocka, K.; Fuks, L.; hexane-platinum complexes with antitumor activity. US Patent
Lewandowski, W.; Fokt, I.; Priebe, W. Teratog., Carcinog., 5132323, 1992.
Mutagen., 2003, 23, 1. [117] Khokhar, A.R.; Hacker, M.P. Inorg. Chim. Acta, 1991, 179, 289.
[84] Fuks, L.; Samochocka, K.; Anulewicz-Ostrowska, R.; Kruszewski, [118] Kidani, Y.; Achiwa, K.; Ono, H.; Tomatsu, K.; Zaikokuji, K.; Noji,
M.; Priebe, W.; Lewandowski, W. Eur. J. Med. Chem., 2003, 38, M.; Tashiro, T. J. Clin. Hematol. Oncol., 1985, 15, 35.
775. [119] Talebian, A.H.; Bensely, D.; Schein, P.S.; Green, D. Anti-Cancer
[85] Lin, T.S.; Zhou, R.X.; Scanlon, K.J.; Brubaker, W.F., Jr.; Lee, Drug Design, 1990, 5, 371.
J.J.S.; Woods, K.; Humphreys, C.; Prusoff, W.H. J. Med. Chem., [120] Talebian, A.; Green, D.C.; Schein, P.S. Platinum pharmaceutical
1986, 29, 681. agents. US Patent 4946954, 1990.
[86] Farrell, N.; Qu, Y.; Bierbach, U.; Valsecchi, M.; Menta, E. In [121] Talebian, A.; Green, D.C.; Hammer, C.F.; Schein, P.S. Preparation
Cisplatin: 1999; 479. of platinum compounds for the treatment of cancer. WO Patent
[87] Sachinvala, N.D.; Chen, H.; Niemczura, W.P.; Furusawa, E.; 8900574, 1989.
Cramer, R.E.; Rupp, J.J.; Ganjian, I. J. Med. Chem., 1993, 36, [122] Gao, Q.; Gallop, M.A.; Xiang, J.-N. Platinum-containing com-
1791. pounds exhibiting cytostatic activity, synthesis and methods of use.
[88] Sachinvala, N.D.; Chen, H.; Furusawa, E. Novel sucrose ligands, WO Patent 2006091790, 2006.
methods for their preparation, platinum complexes derived there- [123] Tidmarsh, G.; Matteucci, M.; Rao, P. Compositions and methods
from, and methods for their usage. WO Patent 9400465, 1994. for treating cancer. WO Patent 2003082301, 2003.
[89] Shi, J.; Tong, Q.; Jia, L.; Lei, N. Preparation of carbohydrate [124] Tromp, R.A.; van Boom, S.S.G.E.; Timmers, C.M.; van Zutphen,
derivative diamine and Pt-complex as anticancer agent. CN Patent S.; van der Marel, G.A.; Overkleeft, H.S.; van Boom, J.H.; Reedijk,
1995055, 2007. J. Bioorg. Med. Chem. Lett., 2004, 14, 4273.
[90] Pasini, A.; Zunino, F.; Tofanetti, O.; Gandolfi, C.A.; Tognella, S.; [125] Ouchi, T.; Matsumoto, M.; Masunaga, T.; Ohya, Y.; Ichinose, K.;
Spinelli, S. Aminoanthracenedione-bis-platinum complexes, useful Nakashima, M.; Ichikawa, M.; Kanematsu, T. In Polymeric Drugs
as antitumor agents. EP Patent 320960, 1989. & Drug Delivery Systems; Ottenbrite, R.M.; Kim, S.W., Eds.,
[91] Zunino, F.; Savi, G.; Pasini, A. Cancer Chemother. Pharmacol., Technomic Publishing Lancaster, 2001; 239.
1986, 18, 180. [126] Ohya, Y.; Nagatomi, K.; Ouchi, T. Macromol. Biosci., 2001, 1,
[92] Pasini, A.; Zunino, F. Angew. Chem., 1987, 99, 632. 355.
[93] Fischer, D.S.; Knobf, M.T.; Durivage, H.J.; Beaulieu, N. The [127] Ohya, Y.; Oue, H.; Nagatomi, K.; Ouchi, T. Biomacromolecules,
Cancer Chemotherapy Handbook (Mosby's Cancer Chemotherapy 2001, 2, 927.
Handbook), Mosby: Philadelphia, 2003, pp. 640. [128] Ichinose, K.; Tomiyama, N.; Nakashima, M.; Ohya, Y.; Ichikawa,
[94] Pasini, A.; Pratesi, G.; Savi, G.; Zunino, F. Inorg. Chim. Acta, M.; Ouchi, T.; Kanematsu, T. Anti-Cancer Drugs, 2000, 11, 33.
1987, 137, 123. [129] Ohya, Y.; Shirakawa, S.; Matsumoto, M.; Ouchi, T. Polym. Adv.
[95] Bakola-Christianopoulou, M.N.; Akrivos, P.D. Int. J. Chem., 1999, Technol., 2000, 11, 635.
9, 47. [130] Ohya, Y.; Masunaga, T.; Baba, T.; Ouchi, T. J. Biomater. Sci.,
[96] Krawielitzki, S.; Beck, W. Chem. Ber./Recl., 1997, 130, 1659. Polym. Ed., 1996, 7, 1085.
[97] Sagredos, A.N.; Papageorgiou, V.P.; Mellidis, A.S. Platinum [131] Ohya, Y.; Masunaga, T.; Baba, T.; Ouchi, T. J. Macromol. Sci.,
complexes of substituted antitumor 5,8-dihydroxy-1,4-naphthoqui- Chem., 1996, A33, 1005.
nones and their use. EP Patent 144809, 1985. [132] Nakashima, M.; Ichinose, K.; Kanematsu, T.; Masunaga, T.; Ohya,
[98] Fiallo, M.M.L.; Garnier-Suillerot, A. Biochemistry, 1986, 25, 924. Y.; Ouchi, T.; Tomiyama, N.; Sasaki, H.; Ichikawa, M. Biol.
[99] Fiallo, M.M.L.; Garnier-Suillerot, A. Inorg. Chim. Acta, 1987, 137, Pharm. Bull., 1999, 22, 756.
119. [133] Schechter, B.; Rosing, M.A.; Wilchek, M.; Arnon, R. Cancer
[100] Moustatih, A.; Fiallo, M.M.L.; Garnier-Suillerot, A. J. Med. Chem., Chemother. Pharmacol., 1989, 24, 161.
1989, 32, 336. [134] Schechter, B.; Pauzner, R.; Arnon, R.; Wilchek, M. Cancer Bio-
[101] Junicke, H.; Arendt, Y.; Steinborn, D. Inorg. Chim. Acta, 2000, chem. Biophys., 1986, 8, 277.
304, 224. [135] Schechter, B.; Pauzner, R.; Wilchek, M.; Arnon, R. Cancer Bio-
[102] Stocker, B.L.; Hoberg, J.O. Organometallics, 2006, 25, 4537. chem. Biophys., 1986, 8, 289.
[103] Ang, W.H.; Khalaila, I.; Allardyce, C.S.; Juillerat-Jeanneret, L.; [136] Serino, A.J.; Henson, G.W.; Schwartz, D.A.; Picker, D.H. Macro-
Dyson, P.J. J. Am. Chem. Soc., 2005, 127, 1382. molecular platinum antitumor agents. EP Patent 280474, 1988.
[104] Reithofer, M.R.; Galanski, M.; Arion, V.B.; Keppler, B.K. Chem.
Commun., 2008, 1091.
2590 Current Medicinal Chemistry, 2008 Vol. 15, No. 25 Hartinger et al.

[137] Miki, S.; Mori, F.; Konishi, M.; Okada, M.; Nishida, T.; Tashiro, [171] Hartinger, C.G.; Jakupec, M.A.; Zorbas-Seifried, S.; Groessl, M.;
T.; Tsukagoshi, S. Platinum complexes with dextran or hy- Egger, A.; Berger, W.; Zorbas, H.; Dyson, P.J.; Keppler, B.K.
droxyethyl starch as neoplasm inhibitors. EP Patent 307827, 1989. Chem. Biodiversity, 2008, in press.
[138] Okada, M.; Mori, F.; Konishi, M.; Miki, S.; Kageyu, A.; Tashiro, [172] Hartinger, C.G.; Hann, S.; Koellensperger, G.; Sulyok, M.; Groessl,
T.; Tsukagoshi, S. Dextran complexes as tumor inhibitors and their M.; Timerbaev, A.R.; Rudnev, A.V.; Stingeder, G.; Keppler, B.K.
manufacture. JP Patent 01123803, 1989. Int. J. Clin. Pharmacol. Ther., 2005, 43, 583.
[139] Mori, F.; Okada, M.; Miki, S.; Ebashi, I.; Nishida, T.; Kawai, K.; [173] Groessl, M.; Reisner, E.; Hartinger, C.G.; Eichinger, R.; Semenova,
Tashiro, T.; Tsukagoshi, S. Polysaccharide complexes, their prepa- O.; Timerbaev, A.R.; Jakupec, M.A.; Arion, V.B.; Keppler, B.K. J.
ration and pharmaceutical uses. WO Patent 9003402, 1990. Med. Chem., 2007, 50, 2185.
[140] Gill, D.S.; Andrulis, P.J., Jr. Platinum-polymer complexes and their [174] Hartinger, C.G.; Keppler, B.K. Electrophoresis, 2007, 28, 3436.
use as antitumor agents. WO Patent 8910928, 1989. [175] Jakupec, M.A.; Reisner, E.; Eichinger, A.; Pongratz, M.; Arion,
[141] Schechter, B.; Arnon, R.; Wilchek, M. React. Polym., 1995, 25, V.B.; Galanski, M.; Hartinger, C.G.; Keppler, B.K. J. Med. Chem.,
167. 2005, 48, 2831.
[142] Schechter, B.; Pauzner, R.; Arnon, R.; Haimovich, J.; Wilchek, M. [176] Schluga, P.; Hartinger, C.G.; Egger, A.; Reisner, E.; Galanski, M.;
Cancer Immunol. Immunother., 1987, 25, 225. Jakupec, M.A.; Keppler, B.K. Dalton Trans., 2006, 1796.
[143] McIntosh, D.P.; Cooke, R.J.; McLachlan, A.J.; Daley-Yates, P.T.; [177] Allardyce, C.S.; Dyson, P.J.; Ellis, D.J.; Heath, S.L. Chem. Com-
Rowland, M. J. Pharm. Sci., 1997, 86, 1478. mun., 2001, 1396.
[144] Lowe, G.; Droz, A.S.; Vilaivan, T.; Weaver, G.W.; Park, J.J.; Pratt, [178] Scolaro, C.; Bergamo, A.; Brescacin, L.; Delfino, R.; Cocchietto,
J.M.; Tweedale, L.; Kelland, L.R. J. Med. Chem., 1999, 42, 3167. M.; Laurenczy, G.; Geldbach, T.J.; Sava, G.; Dyson, P.J. J. Med.
[145] Lowe, G.; Droz, A.S.; Vilaivan, T.; Weaver, G.W.; Tweedale, L.; Chem., 2005, 48, 4161.
Pratt, J.M.; Rock, P.; Yardley, V.; Croft, S.L. J. Med. Chem., 1999, [179] Yan, Y.K.; Melchart, M.; Habtemariam, A.; Sadler, P.J. Chem.
42, 999. Commun., 2005, 4764.
[146] Ma, D.-L.; Shum, T.Y.-T.; Zhang, F.; Che, C.-M.; Yang, M. Chem. [180] Ang, W.H.; Daldini, E.; Scolaro, C.; Scopelliti, R.; Juillerat-
Commun., 2005, 4675. Jeannerat, L.; Dyson, P.J. Inorg. Chem., 2006, 45, 9006.
[147] Wai-Yin Sun, R.; Ma, D.-L.; Wong, E.L.-M.; Che, C.-M. Dalton [181] Dyson, P.J.; Sava, G. Dalton Trans., 2006, 1929.
Trans., 2007, 4884. [182] Scolaro, C.; Geldbach, T.J.; Rochat, S.; Dorcier, A.; Gossens, C.;
[148] Shi, J.-C.; Yueng, C.-H.; Wu, D.-X.; Liu, Q.-T.; Kang, B.-S. Or- Bergamo, A.; Cocchietto, M.; Tavernelli, I.; Sava, G.;
ganometallics, 1999, 18, 3796. Rothlisberger, U.; Dyson, P.J. Organometallics, 2006, 25, 756.
[149] Shi, J.-C.; Zheng, Y.; Wu, D.-X.; Huang, X.-Y.; Liu, Q.-T.; Kang, [183] Vock, C.A.; Scolaro, C.; Phillips, A.D.; Scopelliti, R.; Sava, G.;
B.-S. Inorg. Chim. Acta, 1999, 290, 121. Dyson, P.J. J. Med. Chem., 2006, 49, 5552.
[150] Galanski, M.; Baumgartner, C.; Meelich, K.; Arion, V.B.; Fremuth, [184] Melchart, M.; Sadler, P.J. In Bioorganometallics; Jaouen, G., Ed.,
M.; Jakupec, M.A.; Schluga, P.; Hartinger, C.G.; von Keyserlingk, Wiley-VCH: Weinheim, 2006; 39.
N.G.; Keppler, B.K. Inorg. Chim. Acta, 2004, 357, 3237. [185] Scolaro, C.; Chaplin, A.B.; Hartinger, C.G.; Bergamo, A.; Coc-
[151] Hartinger, C.G.; Schluga, P.; Galanski, M.; Baumgartner, C.; chietto, M.; Keppler, B.K.; Sava, G.; Dyson, P.J. Dalton Trans.,
Timerbaev, A.R.; Keppler, B.K. Electrophoresis, 2003, 24, 2038. 2007, 5065.
[152] Timerbaev, A.R.; Hartinger, C.G.; Aleksenko, S.S.; Keppler, B.K. [186] Mendoza-Ferri, M.G.; Hartinger, C.G.; Eichinger, R.E.; Stolyarova,
Chem. Rev., 2006, 106, 2224. N.; Jakupec, M.A.; Nazarov, A.A.; Severin, K.; Keppler, B.K. Or-
[153] Aleksenko, S.S.; Hartinger, C.G.; Semenova, O.; Meelich, K.; ganometallics, 2008, 27, 2405.
Timerbaev, A.R.; Keppler, B.K. J. Chromatogr. A, 2007, 1155, [187] Mendoza-Ferri, M.G.; Hartinger, C.G.; Nazarov, A.A.; Kandioller,
218. W.; Severin, K.; Keppler, B.K. Appl. Organomet. Chem., 2008, 22,
[154] Hollis, L.S.; Stern, E.W. Cis-Diamineplatinum complexes with 326.
diphosphonates as antitumor agents. EP Patent 409527, 1991. [188] Margalit, R.; Gray, H.B.; Clarke, M.J.; Podbielski, L. Chem.-Biol.
[155] Finkelstein, A.E.; Walz, D.T.; Batista, V.; Mizraji, M.; Roisman, Interact., 1986, 59, 231.
F.; Misher, A. Ann. Rheum. Dis., 1976, 35, 251. [189] Nazarov, A.A.; Berger, I.; Biba, F.; Hartinger, C.G.; John, R.;
[156] Berglof, F.E.; Berglof, K.; Walz, D.T. J. Rheumatol., 1978, 5, 68. Mangrum, J.B.; Farrell, N.P.; Galanski, M.; Jakupec, M.A.; Kep-
[157] Finkelstein, A.E.; Roisman, F.R.; Batista, V.; de Nudelman, F.G.; pler, B.K. J. Biol. Inorg. Chem., 2007, 12, S40.
Titto, E.H.; Misraji, M.; Walz, D.T. J. Rheumatol., 1980, 7, 160. [190] Berger, I. Anticancer Carbohydrate Metal Complexes: Synthesis
[158] Shaw, C.F. III. Chem. Rev., 1999, 99, 2589. and Characterization, PhD thesis, Vienna: University of Vienna,
[159] Simon, T.M.; Kunishima, D.H.; Vibert, G.J.; Lorber, A. Cancer 2007.
Res., 1981, 41, 94. [191] Shaheen, M.A.; Berger, I.; Nazarov, A.A.; Hartinger, C.G.; Koro-
[160] Mirabelli, C.K.; Johnson, R.K.; Sung, C.M.; Faucette, L.; Muir- teev, M.P.; Nifant'ev, E.E.; Keppler, B.K. Chem. Biodiversity,
head, K.; Crooke, S.T. Cancer Res., 1985, 45, 321. 2008, in press.
[161] Gabbiani, C.; Casini, A.; Messori, L. Gold Bull., 2007, 40, 73. [192] Berger, I.; Hanif, M.; Nazarov, A.A.; Hartinger, C.G.; John, R.;
[162] Barnard, P.J.; Berners-Price, S.J. Coord. Chem. Rev., 2007, 251, Kuznetsov, M.L.; Groessl, M.; Schmitt, F.; Zava, O.; Biba, F.;
1889. Arion, V.B.; Galanski, M.; Jakupec, M.A.; Juillerat-Jeanneret, L.;
[163] Casini, A.; Hartinger, C.; Gabbiani, C.; Mini, E.; Dyson, P.J.; Dyson, P.J.; Keppler, B.K. Chem. Eur. J., 2008, DOI:
Keppler, B.K.; Messori, L. J. Inorg. Biochem., 2008, 102, 564. 10.1002/chem.200801032.
[164] Sannella, A.R.; Casini, A.; Gabbiani, C.; Messori, L.; Bilia, A.R.; [193] Alessio, E.; Mestroni, G.; Bergamo, A.; Sava, G. Curr. Top. Med.
Vincieri, F.F.; Majori, G.; Severini, C. FEBS Lett., 2008, 582, 844. Chem. (Sharjah, United Arab Emirates), 2004, 4, 1525.
[165] Shi, J.-C.; Huang, X.-Y.; Wu, D.-X.; Liu, Q.-T. Inorg. Chem., [194] Schneider, M.; Wenzel, M. J. Lab. Comp. Radiopharm., 1981, 18,
1996, 35, 2742. 293.
[166] Rigobello, M.P.; Scutari, G.; Folda, A.; Bindoli, A. Biochem. [195] Wenzel, M.; Schneider, M. Int. J. Appl. Radiat. Isot., 1981, 32, 5.
Pharmacol., 2004, 67, 689. [196] Köpf-Maier, P. Anticancer Res., 1999, 19, 493.
[167] Mirabelli, C.K.; Johnson, R.K.; Hill, D.T.; Faucette, L.F.; Girard, [197] Lu, Z.; Lu, C.; Ren, X.; Meng, Q. J. Organomet. Chem., 2006, 691,
G.R.; Kuo, G.Y.; Sung, C.M.; Crooke, S.T. J. Med. Chem., 1986, 5895.
29, 218. [198] Sasse, F.; Erker, G.; Kehr, G.; Meyer Zu Berstenhorst, B.; Redlich,
[168] Shi, J.-C.; Chen, L.-J.; Huang, X.-Y.; Wu, D.-X.; Kang, B.-S. J. H. Preparation of anionic cyclopentadienyl-functionalized carbo-
Organomet. Chem., 1997, 535, 17. hydrates as building blocks for metallocene carbohydrate antican-
[169] Baker, M.V.; Barnard, P.J.; Berners-Price, S.J.; Brayshaw, S.K.; cer drugs. DE Patent 2006-102006053690, 2008.
Hickey, J.L.; Skelton, B.W.; White, A.H. J. Organomet. Chem., [199] Jessen, L.; Haupt, E.T.K.; Heck, J. Chem. Eur. J., 2001, 7, 3791.
2005, 690, 5625. [200] Kuentzer, D.; Jessen, L.; Heck, J. Chem. Commun., 2005, 5653.
[170] Mirabelli, C.K.; Jensen, B.D.; Mattern, M.R.; Sung, C.M.; Mong, [201] Gansauer, A.; Franke, D.; Lauterbach, T.; Nieger, M. J. Am. Chem.
S.M.; Hill, D.T.; Dean, S.W.; Schein, P.S.; Johnson, R.K.; Crooke, Soc., 2005, 127, 11622.
S.T. Anti-Cancer Drug Design, 1986, 1, 223.
Anticancer Sugar-Metal Complexes Current Medicinal Chemistry, 2008 Vol. 15, No. 25 2591

[202] Meyer zu Berstenhorst, B.; Erker, G.; Kehr, G.; Froehlich, R. [208] Hartinger, C.G.; Nazarov, A.A.; Arion, V.B.; Giester, G.; Jakupec,
Dalton Trans., 2006, 3200. M.; Galanski, M.; Keppler, B.K. New J. Chem., 2002, 26, 671.
[203] Kuentzer, D.; Tschersich, S.; Heck, J. Z. Anorg. Allg. Chem., 2007, [209] van Staveren, D.R.; Metzler-Nolte, N. Chem. Rev., 2004, 104,
633, 43. 5931.
[204] Erker, G. J. Organomet. Chem., 2007, 692, 1187. [210] Warburg, O. Science, 1956, 123, 309.
[205] Jaouen, G. (Ed.) Bioorganometallics, Wiley-VCH: Weinheim, [211] Kim, J.-W.; Gardner, L.B.; Dang, C.V. Drug Discov. Today Dis.
2006, pp. 444. Mech., 2005, 2, 233.
[206] Dive, D.; Biot, C. ChemMedChem, 2008, 3, 383. [212] Macquet, J.P.; Butour, J.L. JNCI, J. Natl. Cancer Inst., 1983, 70,
[207] Itoh, T.; Shirakami, S.; Ishida, N.; Yamashita, Y.; Yoshida, T.; 899.
Kim, H.S.; Wataya, Y. Bioorg. Med. Chem. Lett., 2000, 10, 1657.

Received: June 01, 2008 Revised: August 10, 2008 Accepted: August 15, 2008

View publication stats

You might also like