You are on page 1of 13

View Article Online / Journal Homepage / Table of Contents for this issue

NJC Dynamic Article Links

Cite this: New J. Chem., 2011, 35, 1973–1985

www.rsc.org/njc PERSPECTIVE
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

Application of ferrocene and its derivatives in cancer researchw


Cátia Ornelas*
Received (in Montpellier, France) 27th February 2011, Accepted 10th May 2011
DOI: 10.1039/c1nj20172g

The use of ferrocene-based compounds for medicinal applications is an active research area.
Many reports have demonstrated that some ferrocenyl derivatives are highly active against several
diseases, including cancer. This review focuses on the most relevant examples of ferrocene-based
molecules that show anticancer activity.

1. Introduction strategies leading to ferrocene derivatives, and explored their


applications in a wide range of scientific areas.8 Due to
Ferrocene was discovered in 1951,1,2 but its correct structure the favorable electronic properties of ferrocene and its easy
was only elucidated soon afterward independently by Wilkinson functionalization, these compounds have found many applica-
et al. and Fischer and Pfab.3,4 The resemblance of its reactivity tions in materials science including sensors,9–18 catalysts,16,19–24
to that of benzene inspired Woodward et al. to coin the new electroactive materials25–30 and aerospace materials.31,32
iron compound with the name ferrocene.5 The elucidation of The stability of ferrocene in aqueous and aerobic media, the
the structure of ferrocene was a landmark discovery in the history accessibility of a large variety of derivatives, and its favorable
of chemistry, and led to the birth of modern organometallic electrochemical properties have made ferrocenyl compounds
chemistry. The terms ‘‘sandwich compound’’ and ‘‘metallocene’’ very popular molecules for biological applications.33–39
are applied today, not only to ferrocene and its derivatives, but to The use of ferrocene in medicinal applications is an active
a much wider range of compounds that include other metals.6 research area. Many reports have shown that some ferro-
Ferrocene quickly attracted the attention of the scientific and cene derivatives are highly active in vitro and in vivo, against
technical community on account of its fascinating chemistry.6,7 several diseases including fungal and bacterial infections,40,41
Chemists wasted no time and started to develop synthetic malaria,37,42–44 human immunodeficiency virus (HIV),45 and
cancer.33–39
The anticancer potential of ferrocene derivatives was first
College of Chemistry, University of California Berkeley, California, studied in the late 1970s, when Brynes and co-workers reported
94720-1460, USA. E-mail: catiaornelas@berkeley.edu;
Fax: +1 510-643-3079 the antitumor activity of ferrocenyl compounds bearing amine
w This article is dedicated to Professor Didier Astruc, a special mentor or amide groups against lymphocytic leukemia P-388.46 The
and friend, on the occasion of his 65th birthday. compounds were administered to mice intraperitoneally with
either water or water-Tween 80 (surfactant). The antitumor
activity of these compounds was low but significant enough to
Dr Cátia Ornelas received her show that the incorporation of the ferrocenyl group into an
MS degree in chemistry from appropriate carrier could provide an agent with enhanced anti-
the University of Madeira, tumor activity (Fig. 1).46
Portugal, working with Professor Since then, several types of ferrocenyl compounds have been
João Rodrigues. For her PhD
synthesized and evaluated in terms of anticancer properties.
studies, she moved to the Univer-
sity of Bordeaux (France) to The following sections of this review describe the most relevant
work with Professor Didier examples of ferrocene-based compounds that have shown
Astruc on dendrimer chemistry.
From 2008 until the summer of
2010, she was a postdoctoral
fellow in the group of Professor
Marcus Weck at New York
University. Presently, she
Cátia Ornelas works as a postdoctoral fellow
with Professor Jean M. J.
Fre´chet at the University of California at Berkeley. Her interests
are in supramolecular chemistry, catalysis, and dendrimers for Fig. 1 Chemical structures of ferrocene (1); and a ferrocenyl compound
nanomedicine. bearing amide groups (2), tested against lymphocytic leukemia P-388.46

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1973
View Article Online

anticancer activity, which include: (i) ferricenium salts; (ii) ferro-


cene conjugated to biologically active molecules; (iii) DNA
targeting moieties linked to ferrocene; (iv) ferrocenyl derivatives
of selective estrogen modulators; (v) ferrocenyl androgens and
anti-androgens; and (vi) ferrocenyl compounds coordinated to
other metals. Section 8 describes some examples where active
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

ferrocenyl compounds were conjugated to drug delivery systems, Fig. 3 Two examples of a series of ferrocenylalkylazoles, which are
and the final section discusses the prospects and future of the field. active against solid tumors.55

by ferrocene.48,54–56 A series of ferrocenylalkyl azoles (Fig. 3)


2. Ferricenium salts against Ehrlich ascites tumor was synthesized and their tumor growth inhibition and toxicity
and Rauscher leukemia virus was studied in vivo. These ferrocenyl compounds were tolerated
In 1984, Köpf-Maier, Köpf, and Neuse disclosed the anti- well by animals, showing lower toxicity when compared to
tumor activity of ferricenium salts.47 This group used mice ferrocene and clinically used drugs. Any noticeable visual
bearing Ehrlich ascites tumor (EAT) to test ferrocene and alteration of the mice appearance, neither in the condition of
some ferricenium salts bearing different counter anions. Due its internal organs were observed.48,54,55 The best results were
to the low solubility of ferrocene in water, it was injected in obtained with compound 4 (Fig. 3) when tested against solid
mice using propylene glycol, and it was observed that the tumors, which presented up to 100% of tumor growth inhibition,
neutral ferrocene complex lacked anti-tumor efficacy against and then achieved 45% regression. The success of compound 4
EAT. On the other hand, the ferricenium salts are highly soluble was attributed to: (1) the hydrophilic benzotriazolyl group
in water, and could be directly injected in mice using saline provides transport in aqueous media and the lipophilic ferrocenyl
media. Ferricenium chloroferrates such as [Cp2Fe]+[FeCl4] moiety ensures membrane permeability; (2) the ferrocenyl
(3a) caused survival of up to 83% of the animals after treatment group is capable of forming ionic bonds (after oxidation to the
with optimum doses, leading to increases in life-span of 380%, ferricenium form), and the azolyl group can form hydrogen
when compared with the untreated controls (Fig. 2).47 bonds with phosphate groups at cleavage points of DNA;
In vivo tests by Popova and co-workers showed that ferri- (3) the planar heterocyclic ring and bulky ferrocenyl group can
cenium tri-iodide (3b) was effective against Rauscher leukemia intercalate between the planes of DNA nucleic bases; (4) the
virus (RLV), while [Cp2Fe]+[FeCl4] (3a) did not show any swinging alkyl bridge allows for the formation of the ligand–
significant activity against this disease (Fig. 2).48 receptor complexes.55
These results suggest that the anticancer activity of the
ferricenium salts strongly depends on the counter anion and 3.2. Ferrocene conjugated to peptides for lung cancer
the type of cancer. Kenny’s group synthesized a series of ferrocenyl compounds
Soon thereafter, Neuse and Kanzawa proposed that ferrocenyl that possess three key components: (i) a ferrocene moiety, (ii) a
compounds could be oxidized in the cell via normal metabolic conjugated linker that lowers the oxidation potential of the
processes, so that both ferricenium and ferrocene derivatives ferrocenyl group, and (iii) a peptide derivative that can interact
could yield an anti-proliferative effect.49,50 Further studies with biomolecules via secondary interactions such as hydrogen
suggested that the mechanism of action of the ferricenium bonding.57–60 All the ferrocenyl-peptide derivatives were tested
salts was orchestrated by generation of radicals through the in vitro against the human lung carcinoma cell line H1299, and
Fenton pathway that causes DNA damage and consequent most of them showed IC50 values below 10 mM (IC50
apoptosis of cells.36,51–53 value indicates the mean compound concentration required to
achieve 50% growth inhibition). In particular, N-(6-ferrocenyl-2-
3. Ferrocene conjugated to biologically active naphthoyl)-g-aminobutyric acid ethyl ester (compound 6, Fig. 4)
molecules as promising anti-cancer agents had an IC50 value of 0.62 mM, and is more active in vitro than the
clinical employed anti-cancer drug cisplatin.57–60
3.1. Ferrocenylalkylazoles with high activity against solid
tumors 3.3. Ferrocenylalkyl nucleobases against carcinoma 755
Azoles including imidazole, pyrazole and adenine, which A few research groups have been trying to combine the
are the central ingredients in many drugs, have been chosen potential antitumor properties of some nucleosides and nucleo-
by Snegur et al. as the objects for chemical modification bases with the properties of ferrocene by attaching them
covalently.61–63 Simenel et al. prepared ferrocenyl derivatives
of thymine, adenine, cytosine and iodo-cytosine.62 The anti-
tumor activity of 1-N-ferrocenylmethyl thymine (compound 7,
Fig. 5) was studied in vivo against some animal tumor systems
such as carcinoma 755 (Ca755), melanoma B16 (B16) and
Lewis lung carcinoma (LLC). Compound 7 showed a signifi-
cant antitumor effect against Ca755 with 70% tumor growth
Fig. 2 Molecular structure of ferricenium salts 3a47 and 3b48 which inhibition in comparison with the control. Furthermore, this
are active against EAT and RLV respectively. group suggested a synergistic effect of antitumor activity against

1974 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM. View Article Online

Fig. 7 Retinoyl derivative of ferrocene.70

3.5. Ferrocenyl derivatives of retinoids


Retinoids include active metabolites of vitamin A (retinol) and
play an important role in vertebrate growth and development,
supporting cell differentiation, embryonic development, immune
Fig. 4 Structure of N-(6-ferrocenyl-2-naphthoyl)-g-aminobutyric response, and reproduction.68 Some trans-retinoic acids are
acid ethyl ester compound 6, which showed in vitro activity against the currently used for treatment of dermatological disorders and
human lung carcinoma cell line H1299.60 as chemotherapeutic agents against various endothelial cancers,
breast cancer, and endometrial cancer. Their actions are mediated
through binding and activation of the retinoic acid receptors
(RARs) or retinoid X receptors (RXRs), which function as
ligand-dependent transcription factors.69
As an attempt to improve the anti-cancer activity of retinoids,
some ferrocenyl derivatives (compound 10, Fig. 7) were prepared,
and their antiproliferative activity toward human lung cancer cell
line (A549), human liver cancer cell line (BEL7404), and human
Fig. 5 Structure of 1N-ferrocenylmethyl thymine (7) that showed tongue cancer cell line (Tca) was investigated.70 Although the
70% tumor growth inhibition on Ca755.62 results showed that the ferrocenyl analogues exhibited higher
antiproliferative activities than the parent 13-cis-retinoic acid, the
Ca755, by combining 1-N-ferrocenylmethyl thymine 7 with the IC50 values obtained can be considered modest (19–40 mM).70
well-known antitumor drug cyclophosphamide.62

3.4. Ferrocenyl derivative of illudin M 4. Ferrocenyl compounds targeting DNA


Illudin M is a fungal cytotoxin that belongs to a family of 4.1. Combination of ferrocene with a DNA intercalator
sesquiterpenes, produced by some mushrooms.64 Although the
The attachment of a ferrocenyl group to molecules that bind
illudins are highly active against various tumors, their extreme
DNA is a promising strategy to bring the ferrocene moiety to
toxicity has prevented clinical applications.64 Some semisynthetic
close proximity of DNA, which might enhance the probability
illudin derivatives with reduced toxicity are currently in phase II
of DNA damage and cell apoptosis.
clinical trials for a number of cancers including prostate, ovarian,
Ong’s group synthesized compound 11 (Fig. 8) by linking
pancreatic, renal, breast and lung.65,66
ferrocene to acridine (a DNA intercalator) and compared its
With the intention to enlarge the therapeutic index and the
anticancer activity with the analogous ferrocenyl derivative
cell line specificity of Illudin M (compound 8, Fig. 6), new
lacking the acridine moiety (compound 12, Fig. 8).71 The
esters with 1,1 0 -ferrocenedioic acid and ferrocenoic acid were
in vitro studies indicated that the compound containing acridine
prepared by Schobert and co-workers.67 Bis(illudinyl M) 1,10 -ferro-
(11) was highly toxic to the four cancer cell lines tested, while
cenedioate (compound 9, Fig. 6) was by far the less toxic to
compound 12 was inactive. The inability of the phenyl derivative
nonmalignant fibroblasts, while more cancer selective and more
12 to bind DNA was suggested as the main cause for the lack
cell line-specific than the parent Illudin M. This improvement
of cytotoxicity. These findings are in agreement with the hypo-
partially originated from the ferrocene shielding the enone
thesis that the combination of the cytotoxic effects of the
group against attack of glutathione. TUNEL assays that allow
ferrocenyl group with the DNA binding ability of acridine
the detection of late stages of apoptosis indicated that the
ferrocenyl diester 9 induces apoptosis in cancer cells in a similar
pathway than Illudin M.67

Fig. 6 Molecular structures of illudin M (8), and its ferrocenyl Fig. 8 Molecular structures of ferrocenyl compounds linked to
derivative (9).67 acridine (11) and to a benzyl group (12).71

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1975
View Article Online

Fig. 9 Chemical structures of azalactone ferrocene (13) and thio-


Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

morpholide amido methyl ferrocene (14), potential inhibitors of topo-


isomerase II.74

allows for strong interactions with the key cellular target,


increasing cytotoxicity.71 Fig. 10 Molecular structures of Tamoxifen (15), hydroxytamoxifen
(16) and hydroxyferrocifens (17).76–78
4.2. Ferrocenyl derivatives as inhibitors of topoisomerase II
Topoisomerases are enzymes that unwind and wind DNA to
facilitate DNA replication, and to allow DNA to control the the hydroxyferrocifens 17 is comparable to that of hydroxy-
synthesis of proteins. In normal cells, the activity of topo- tamoxifen 16, being slightly more potent at a concentration of
isomerase II is highly regulated, while it is present at high 0.1 mM, and definitely superior at 1 mM. While hydroxy-
levels in rapidly proliferating cancerous cells.72 This difference tamoxifen was completely inactive on the hormone-independent
in topoisomerase II levels between healthy and cancer cells cells, the hydroxyferrocifens displayed a strong antiproliferative
make these enzymes potential anticancer targets. Thus, develop- effect, presenting a remarkable low IC50 value. Therefore,
ment of isoform-specific topoisomerase II inhibitors has been of two trends could be put forward: one which is similar to
great interest for cancer-specific drug targeting. the antiestrogenic role of hydroxytamoxifen on the estrogen
Kondapi and co-workers reported a series of ferrocenyl receptor, and one which involved the in situ activation of the
derivatives and studied their inhibitory properties against topo- ferrocenyl function.39,76–79
isomerase IIa and b.73,74 Based on structure–activity analysis, Several studies on the structure–activity relationship of hydroxy-
two compounds in particular, azalactone ferrocene 13 and ferrocifens suggest some trends concerning (a) the stereochemistry
thiomorpholide amido methyl ferrocene 14, presented a signifi- of the compounds, (b) the effect of the N,N-dimethylamino
cant effect against topoisomerase II activity (Fig. 9).74 These side chain, (c) the presence and position of the phenol group,
results indicate that the two compounds interacted with topo- (d) the role and location of the ferrocene moiety, (e) the conju-
isomerase II and inhibited its activity causing numerous genetic gation of the system, and (f) the functionalization of the phenyl
implications that ultimately resulted in neoplastic cell death.74 rings.39,80
(a) Under the conditions of relative binding affinity (RBA)
competition with ERa and ERb, the Z isomer of hydroxy-
5. Targeting breast cancer with selective estrogen tamoxifen is clearly better recognized. However, in the few cases
receptor modulators (SERM) where individual isomers of ferrocifens were used in cell studies,
no difference in activity could be ascertained due to the rapid
5.1. Ferrocifens, ferrocifenols and ferrocenophanes
isomerization of the hydroxyferrocifens in protic solvents.80
Among all the ferrocene derivatives for cancer therapeutics, (b) The binding affinity of the hydroxyferrocifens decreases
the ferrocifens are probably the most widely studied, showing as the N,N-dimethylamino side chain becomes longer. Further
very promising results for breast cancer.38,39 For patients studies showed that the side chain has a strong influence on
diagnosed with ER+ breast cancer, commonly the non-steroidal anti-oestrogenicity, however, its impact on cytotoxicity is still
selective estrogen receptor modulator (SERM) tamoxifen (15) controversial.39,81
is prescribed (Fig. 10).75 (c) The impact of the hydroxyl group position in the
In 1996, Jaouen et al. first coupled ferrocene to the active cytotoxicity against breast cancer cells was studied via a series
metabolite tamoxifen 15 by replacing a phenyl group of of para and meta-substituted mono and di-phenols, bearing
4-hydroxytamoxifen 16 with ferrocene (compound 17, one or two ferrocenyl groups.82–84 The trend indicates that
Fig. 10). Since ferrocene is intrinsically aromatic, recognition ferrocifens containing only one ferrocenyl moiety and two
of 17 by the estrogen receptor should not be deteriorated. The phenol groups in the para positions present higher toxicity
hydroxyferrocifens 17 (differentiated by the length of the towards these cancer cells.39,82–84
dimethyl amino chain, n = 2–5, 8) were designed to combine (d) The hydroxyferrocifens were compared with analogues
the antiestrogenic properties of tamoxifen while simultaneously where the ferrocene moiety was replaced by titanium, rhenium
targeting the ferrocene moiety to the receptor and thus to DNA, and ruthenium complexes, and it was clearly shown that
to possibly obtain new therapeutic advantages.76–78 among the metal-based tamoxifen derivatives, the ferrocifens
The antiproliferative activity of the hydroxyferrocifens (17) present the best anticancer activity.85,86
was evaluated on MCF-7 cells which are hormone-dependent (e) To determine the role of the conjugated p-system linking
breast cancer cells that have an important concentration the ferrocene and phenol groups, compounds containing a sp3
of ERa, and on MDA-MB-231 cells, which are classified as carbon instead of an ethylene group were designed. The cell
hormone-independent breast cancer cells because they are devoid studies suggest that compounds possessing a p-system signifi-
of ERa. It was found that on MCF-7 cells, the effect of cantly outperform their non-conjugated analogues.87,88

1976 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM. View Article Online

Fig. 11 Ferrocifens with a variety of phenyl substituents.39

(f) The influence of phenyl substituents other than OH was


evaluated by studying a set of ferrocifens analogues containing
Cl, Br, CF3, CN, NH3, NHCOMe, OCOMe, OMe, SCOMe or
SMe (Fig. 11).84,88,89 While ferrocifens bearing Cl (18), Br (19),
CF3 (20), OMe (25), SCOMe (26) or SMe (27) were inactive,
compounds bearing CN (21), NH3 (22), NHCOMe (23) or
OCOMe (24) showed significant anti-proliferative effects at
10 mM (Fig. 11). Compounds containing two OCOMe showed
similar behavior than the diphenol ferrocifen suggesting
that enzymatic hydrolysis occurs inside the cells. Therefore,
compound 24 can be considered as a ‘‘pro-drug’’.39 Fig. 12 Molecular structures of ferrocenophane compounds structur-
Rigid molecules usually bind more strongly to a receptor than ally related to tamoxifen.91,92
their flexible analogues if they have the proper geometry for
entering the active site, while a flexible molecule must adopt such group of complexes active against breast cancer, with a range of
a geometry causing entropy loss and weaker binding.39 Thus, IC50 values between 0.09 and 13 mM.39 It would be interesting
Jaouen and co-workers reported a set of compounds containing to carry out in vivo studies on the ferrocifen compounds, and
the cyclic [3]ferrocenophane moiety, and compared their anti- detect if these ferrocenyl analogues of tamoxifen present less
cancer activity with their non-cyclic analogues (Fig. 12).90–93 For side effects than the tamoxifen itself, which include the increased
each p-R-phenyl substitution pattern investigated, the [3]ferroceno- risk of endometrial uterine cancer and blood clotting in
phanyl derivatives showed higher cytotoxicity than the corres- the lungs.
ponding ferrocenyl molecules, with the highest activity found
5.2. Ferrocenyl raloxifen derivatives
for compounds with protic substituents (Fig. 12).91,92
Direct comparison between compound 28 that possesses a Following the same line as for the tamoxifen derivatives,
direct linkage between the cyclopentadienyl ring and the double Marques and co-workers have explored the possibility to
bond, and compound 29 in which the ferrocenyl group is linked to replace a phenyl ring by a ferrocenyl moiety, on raloxifen
the double bond through a methylene bridge, showed that the (compound 30, Fig. 13) which is another SERM.94 They
conjugated compound 28 is one order of magnitude more cyto- studied raloxifen derivatives containing one ferrocenyl group,
toxic than the non-conjugated analogue. Compound 28 showed and all the tested compounds showed considerable cytotoxic
an exceptional antiproliferative effect on the hormone-independent activity against ovarian, cervical, lung, colon and breast
MDA-MB-231 and PC-3 cells with an IC50 value of 0.09 mM.91,92 cancer cell lines. Among all the compounds, [3-ferrocenyl-6-
The extensive study on tamoxifen derivatives containing the methoxybenzo[b]thiophen-2-yl][4-(piperazin-1-yl)methylphenyl]
ferrocenyl moiety has provided the scientific community with a methanone (compound 31, Fig. 13) is worthy of note, showing

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1977
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM. View Article Online

Fig. 13 Molecular structures of Raloxifen (30) and a ferrocenyl


compound (31) structurally related to Raloxifen.94

IC50 values in the low-micromolar range and more than one


order of magnitude lower than those of cisplatin. In addition,
chemosensitivity tests on resistant phenotypes indicated that
this compound caused no cross-resistance with cisplatin, although
compound 31 is not considered a potential multidrug-resistant
Fig. 14 Chemical structures of testosterone and dihydrotestosterone
(MDR) substrate. This group also showed that compound 31 and their ferrocenyl derivatives.97,98
activates caspase-3 in ovarian cells, suggesting that the mecha-
nism of cell death was apoptosis rather than necrosis.94
The studies on the SERMs containing ferrocenyl groups DHT, with an ethynyl substituent (Fig. 14). The androgen
have shown that this type of vectorization can be successful, activity of the resulting compounds was rather low, probably
because some of the compounds present both anti-oestrogenic because the receptor does not tolerate substitution in the C-17
and cytotoxic properties. position; however compounds 34 and 35 (Fig. 14) presented
One could then imagine that merely transporting the ferrocene strong anti-proliferative activity on hormone-independent
unit into the nucleus via the receptor–bioligand interaction PC-3 prostate cancer cells with IC50 values of 4.7 and
would be sufficient to produce a cytotoxic effect.39 To investi- 8.3 mM respectively.97
gate this hypothesis, a ferrocenyl group was attached onto A related study was carried out by Manosroi et al. where
the oestradiol skeleton, and these compounds present lower the ferrocenyl group was introduced in the C-2 position of the
anti-cancer activity than the ferrocifens and raloxifen testosterone skeleton (compound 36, Fig. 14). Although the
derivatives.95,96 These results suggest that the anticancer receptor activity of this compound was not yet studied, it showed
activity of the ferrocenyl-SERMs compounds is due to a high cytotoxic activity (comparable to that of doxorubicin)
synergetic effect between the tamoxifen or raloxifen moiety against HeLa cells.98
and the ferrocenyl group, rather than the mere presence of the
ferrocene in the interior of the cells.70,71
6.2. Ferrocenyl derivative of nilutamide
Nilutamide (37) is a non-steroidal anti-androgen therapeutic
6. Targeting prostate cancer with ferrocenyl agent used in prostate cancer patients (Fig. 15). As an attempt
androgens and anti-androgens to improve the therapeutic activity of nilutamide, ferrocenyl
derivatives were synthesized.99 It was expected to obtain
6.1. Testosterone and dihydrotestosterone ferrocenyl
anticancer activity for the nilutamide-ferrocenyl compounds
complexes
through an anti-androgen effect; however, these compounds
Testosterone (32), the endogenous hormone, and its active showed an unusual behavior. Compound 38 (Fig. 15) showed
metabolite dihydrotestosterone (33) (DHT), can promote the significant cytotoxic activity against PC-3 hormone-independent
malignant growth of the prostate gland (Fig. 14). Thus, the prostate cancer cells with an IC50 value of 5.4 mM, which
latter therapy for prostate cancer consists in using anti-androgens, suggests that its activity does not occur via the androgen
which block the androgen-induced hormonal effect by binding receptor, but probably via recognition by other receptors.
to the receptor instead of testosterone. The attachment of Furthermore, this study emphasized that in this case the
ferrocene to anti-androgens could enhance their anticancer introduction of the ferrocenyl group produces the same effect
activity. Thus, several ferrocenyl derivatives of testosterone than a purely organic aromatic moiety.39,99
and dihydrotestosterone (DHT) were synthesized, and their The idea of attaching ferrocene to androgens and anti-
anticancer activity was studied. androgens appeared to be a promising strategy to create efficient
Jaouen and co-workers introduced the ferrocenyl group at drugs for hormone-dependent prostate cancers. However, the
the C-17 position of the steroid skeleton of testosterone and results obtained for the ferrocenyl derivatives of testosterone,

1978 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM. View Article Online

Fig. 15 Chemical structures of nilutamide (37) and ferrocenyl Fig. 16 Platinum complexes bearing ferrocenyl groups, which are
compound 38 which is structurally related to nilutamide.99 active against leukemia cell line P388.110

dihydrotestosterone and nilutamide contradict this hypothesis


because the resulting compounds showed low androgen activity
and higher cytotoxic activity against hormone-independent
prostate cancer cells. This unexpected behavior is still not
clearly explained but it might be partially due to the fact that the
ferrocenyl group somehow precludes binding to the receptors.

7. Compounds containing ferrocenyl group and


Fig. 17 Rhodium complexes bearing ferrocenyl groups, which anti-
other transition metals cancer activity is similar to that of cisplatin.112
The success of cisplatin100 as an anticancer agent has stimulated
the search for cytotoxic compounds based on platinum and
other transition metals, in a continuous pursuit for fewer side
effects.101–103 Attempts to combine the anti-tumor activity of
ferrocenyl derivatives to that of other transition metals started
in the late 80’s.104–109

7.1. Ferrocene compounds with platinum, palladium, rhodium


and iridium
Since the Rosenberg’s discovery100 of the potent antitumor Fig. 18 Palladacycles that show high selectivity for tumor cells.115,116
activity of cisplatin, several amino-platinum complexes were
prepared and evaluated for their antitumor activity. Among
them, complexes containing ferrocenyl groups were also agents against solid tumors. The results revealed that the
studied.105,110,111 Al-Allaf et al. prepared a few Pt(II) complexes palladium compound 43 (Fig. 18) was able to inhibit cathepsin
with the formula cis-[Pt(Fc2PhP)(DMSO)X2] and tested their B activity in a reversible fashion by binding to free cathepsin B
antitumor activity in vitro, against leukemia cell line P388.110 as well as to the enzyme–substrate complex.
Complexes 39 and 40 (Fig. 16) showed IC50 values close to The application of this complex, in Walker tumor-bearing
those of cisplatin and 5-fluorouracil.110 rats, resulted in 90% inhibition of the tumor growth, reducing
Rajput et al. have prepared a series of ferrocenyl compounds the solid tumors mass from 4.0 g to 0.3 g. Toxicological studies
containing pyridine ligands and studied their coordination using mice treated with high doses of the complex did not
behavior with platinum, palladium, rhodium and iridium. The show any alterations in red or white blood cell morphology
new complexes were tested for cytotoxic activity and compared 14 days after the drug administration. Similar results were
to cisplatin. Several of the complexes displayed significant obtained with hepatic, kidney, and spleen tissues, making
cytotoxic activity against the cancer cell line WHCO1, being this complex a promising antitumoral drug with reduced
the rhodium complexes 41 and 42 the most active, with growth toxicity.114,116
inhibitory activities similar to that of cisplatin (Fig. 17).112 The same research group also showed that the ionic and
A recent study by Spencer et al. where a series of palladacycles mononucleated palladacycle complex 44 (Fig. 18) triggers
were evaluated in vitro as cytotoxic agents against A2780/S lysosomal membrane permeabilization with release of lyso-
cancer cells suggested that the complexes containing 1,2-bis- somal enzymes, especially cathepsin B to the cytosol of K562
(diphenylphosphino)ethane were more active than their analogues cells, which lack the oncoprotein p53.115 This finding was of
with 1,2-bis(diphenylphosphino)ferrocene.113 particular importance from the point of view that development
Caires and co-workers carried out interesting in vivo studies of anticancer agents with different modes of action is a
involving chiral cyclopalladated complexes derived from key step in overcoming clinical therapy resistance. Moreover,
N,N-dimethyl-1-phenethylamine and the coordinating ligand toxicological studies demonstrated that the palladacycle
1,10 -bis(diphenylphosphine)ferrocene (dppf).114–116 The complexes complex 44 presents low level of toxicity for normal tissue,
were investigated as cathepsin B inhibitors and antitumoral suggesting high selectivity for tumor cells.115,116

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1979
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM. View Article Online

Fig. 19 Gold(I) carbene complex bearing two ferrocenyl groups.119

7.2. Gold complexes bearing ferrocenyl groups


Fig. 20 Ruthenium compounds bearing ferrocenyl groups, which
In order to combine the anticancer properties of ferrocenyl present anticancer activity.125,128
compounds, diphenylphosphine derivatives and gold complexes,
several research groups have synthesized a variety of gold
complexes containing dppf. Despite the presence of the three Ott et al. showed that their activity against colon carcinoma
moieties, all the complexes tested presented only modest anti- and breast cancer cells is relatively close to that of cisplatin.128
cancer activity, being less active than cisplatin.106,108,109,117 Comparison of these complexes with the related monometallic
Recently, Raubenheimer and co-workers showed a successful ruthenium complex and ferrocene derivative indicates that an
combination of the anticancer properties of the gold(I) carbene enhanced electron delocalization might be of high relevance
complexes118 and the ferrocenyl moiety.119 They designed a for the bioactivity.128
gold(I) carbene complex containing two ferrocenyl groups
(compound 45, Fig. 19) and screened it against human HeLa 7.4. Ferrocenyl compounds coordinated to iron, nickel, cobalt,
cervix epithelioid carcinoma cell line, the human colon adeno- copper or zinc
carcinoma line CoLo 320 DM, and the Jurkat leukemia cell Kraatz and co-workers have tested the potential anti-cancer
line. The phosphine-free gold(I) complex 45 presented tumor properties of the redox-active ferrocenyl modified pyrazole
specificity and achieved growth inhibition at lower concen- ligand and its corresponding complexes with iron, nickel
trations than the well-known cisplatin, for two of the three and cobalt.129 Using the human mammary adenocarcinoma
tested cancer cell lines.119 MCF-7 cell line, they verified that the three metal complexes
induced higher cytotoxicity than the free ligand, however the
7.3. Ruthenium complexes containing ferrocenyl groups obtained IC50 values can be considered modest, ranging from
46 to 73 mM.129
Ruthenium complexes have been studied as an alternative to Abd-Elzaher et al. synthesized a ferrocenyl ligand that
platinum, due to its range of oxidation states (II, III and IV) contains salicylaldehyde moieties and studied the anti-cancer
accessible under physiological conditions, and general lower properties of its complexes with Co(II), Ni(II), Cu(II), and
toxicity than the platinum analogues.120–123 The lower toxicity Zn(II), against human breast cancer cell line MCF-7.130 Treat-
of ruthenium complexes towards healthy cells can be partially ments of the MCF-7 cells with gradually increasing doses of
explained by the fact that ruthenium compounds specifi- the complexes revealed that the activity of superoxide dismutase
cally accumulate in rapidly dividing cells, such as tumors. This and the level of hydrogen peroxide were significantly increased,
accumulation of ruthenium in tumors is due to the ability of while the activities of catalase and glutathione peroxidase, and
ruthenium to mimic iron in binding to transferrin (the protein the levels of reduced glutathione were significantly lowered
which delivers iron to cells) which receptors are overexpressed compared with the control MCF-7 cells. These metal complexes
in cancer cells.124 showed significant anticancer activity comparable to that of
As an attempt to combine the cytotoxic properties of nitrosyl cisplatin, being the highest activity achieved by the zinc complex
ruthenium complexes and ferrocene, Von Poelhsitz et al. (compound 48, Fig. 21). This report suggests that the antitumor
reported the ruthenium complex fac-[RuCl3(NO)(dppf)] effect of these metal complexes may be exerted partially by the
(compound 46, Fig. 20).125 This complex was tested on increasing hydrogen peroxide production and by the depletion
MDA-MB-231 breast tumor cells and it showed higher cyto- of intracellular catalase, glutathione peroxidase, and reduced
toxicity than the precursor complex RuCl3NO2H2O and the glutathione.130
free dppf ligand, being six times more active than cisplatin.
Considering the lack of activity of the RuCl3NO2H2O complex,
it is obvious that the ferrocene ligand plays a crucial role on the 8. Ferrocenyl-based molecules with anticancer
anticancer activity of this compound.125
activity conjugated to drug delivery systems
Dyson and co-workers investigated a few arene–ruthenium
complexes containing nitrogen-based ferrocenyl ligands and In the last decade, drug delivery systems have become a promising
tested them against ovarian carcinoma cells; however all the solution to increase water-solubility, selectivity and blood
compounds presented much lower anticancer activity than circulation life-time of small molecule drugs. Several drug
cisplatin.126,127 delivery systems including polymer microcapsules, liposomes,
For complexes where the ferrocene is conjugated to the nanoparticles and dendrimers are being explored to carry a
ruthenium through an ethylene linker (compound 47, Fig. 20), wide variety of drugs into diseased tissue.131–146

1980 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
View Article Online

the use of heptakis-2,3,6-tri-O-methyl-b-CD (TRIMEB) or


2-hydroxypropyl-b-CD (HPbCD) instead of b-CD as hosts
for the ferrocenyl compounds significantly increased their
cytotoxic activity (up to 2.5 times).148
The fact that b-CD significantly increases the water solubility
of the ferrocenyl guests without loss of cytotoxic activity seems
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

to be an important feature that makes these host compounds


interesting ingredients for drug formulations of ferrocenyl
compounds with relevant anticancer activity.147,148 Moreover,
Fig. 21 Zinc complex bearing a ferrocenyl ligand with anticancer the substituted CDs TRIMEB and HPbCD, not only increase
activity comparable to that of cisplatin against MCF-7 breast cancer water solubility of the ferrocenyl compounds, as they also
cells.130 enhance their cytotoxic activity.148

Despite the high anticancer activity of some ferrocenyl 8.2. Polyaspartamide–ferrocene conjugates
compounds described in this review, only a very few reports Neuse and co-workers have focused their investigation on the
focus on the use of drug delivery systems to improve their bio- cytotoxic behavior of polyaspartamide–ferrocene conjugates
logical properties, which include host–guest complexes with against HeLa cells and the Colo 320 DM, a human colon
cyclodextrin,147,148 polyaspartamide copolymers,149–152 polymeric adenocarcinoma line which is known to be intrinsically
micelles153,154 and lipid nanocapsules.155,156 drug-resistant.149–152 Ferrocene was conjugated to water-soluble
polyaspartamide copolymers using a wide variety of linkers
8.1 Host–guest complexes of cyclodextrin and anticancer and side groups. A quick glance at the in vitro data revealed a
active ferrocenyl compounds good performance pattern for most of the conjugates against
both cell lines, with IC50 values in general much lower than
Cyclodextrin (CD) inclusion complexes are interesting for
those of cisplatin.152 Copolymers 49 and 50 (Fig. 22) stand out
pharmaceutical applications due to high water solubility and
as top performers giving IC50 values in the range of 3.6–36 mM
stability.157 Other potential benefits of using CD drug delivery
of ferrocene. The critical compositional difference between the
systems in chemotherapy include improved activities and reduc-
ferrocenyl polymers tested is the presence in 49 and 50 of
tion of toxic side effects.158 CDs are known to form stable inclusion
tertiary amine side groups that enable the polymers to convert
complexes with a large number of ferrocenyl derivatives,159 there-
to cationic species through amine protonation at physiological
fore the CDs have the potential to serve as carriers for cytotoxic
pH.152 Polymers of this type have shown to undergo preferential
ferrocenyl compounds.
cell entry in the cationic state, which may render them potentially
Jaouen’s group have studied the inclusion of ferrocifens
more cytotoxic than analogous polymers lacking the amine
(see Section 5.1) into b-CD.147 It was found that the inter-
groups.152
action with b-CD significantly increases the water solubility of
ferrocifens, while their cytotoxic properties are not affected.147
8.3. Polymeric micelles cross-linked with ferrocene
A similar approach was undertaken by Romão and co-workers,
in which they used CDs to form host–guest complexes with Zhang and co-workers have used 1,1 0 -ferrocenedicarboxylic
1,2-disubstituted ferrocenes.148 The cytotoxicity of the complexes acid as a difunctional cross-linker, and developed a novel
with b-CD was found to be similar to those obtained in the ferrocene-containing shell cross-linked thermoresponsive micelle
absence of cyclodextrin. Interestingly, they also showed that system via a two-step procedure (Fig. 23): (1) self-assembly of

Fig. 22 Polyaspartamide–ferrocene copolymers.152

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1981
View Article Online

9. Conclusion and prospects


Application of ferrocene derivatives in cancer therapeutics is
an active research area, and many reports have demonstrated
that some ferrocenyl compounds are highly cytotoxic in vitro
against several cancer cell lines, including breast, prostate,
lung, colon and leukemia cancer cells. Besides the significant
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

amount of successful in vitro results for a wide variety of ferro-


cenyl compounds, only a few were tested in vivo. Significant
tumor growth inhibition was obtained in vivo for ferricenium salts
(up to 83% growth inhibition of EAT),47 ferrocenylalkylazoles
(up to 100% growth inhibition and 45% regression of solid
tumors),55 ferrocenyl-nucleobases (up to 70% growth inhibition
of Ca755),62 palladacyles with dppf (up to 90% growth inhibition
of Walker tumors),114–116 and lipid nanocapsules loaded with
hydroxyferrocifen (longer survival rate).155,156 The success of the
in vivo anticancer activity of some ferrocenyl-based molecules is
encouraging and should inspire scientists to design compounds
with even higher efficiency against several cancer types.
It would be of great interest to study the potential side effects
caused by the ferrocenyl compounds, in order to evaluate the
possibility of its use in humans. Particular attention should be
Fig. 23 Formation of the ferrocene-containing shell cross-linked given to the possibility of iron overload in the body, which can
thermoresponsive micelle system (reprinted with permission).154 cause liver damage.
I believe that the future of this area relies on the search for
a poly(N-isopropylacrylamide-co-aminoethyl methacrylate)- efficient biodegradable drug delivery systems to carry the active
b-polymethyl methacrylate (P(NIPAAm-co-AMA)-b-PMMA) ferrocenyl compounds, which should increase water solubility
copolymer into polymeric micelle in aqueous solution; (2) cross- and blood circulation life-time, and enhance selectivity towards
linking of the hydrophilic shell layer via the amidation reaction cancer tissue. Furthermore, the use of multifunctional drug
between the amine groups of AMA units and the carboxylic delivery carriers would allow the attachment or encapsulation
acid functions of 1,1 0 -ferrocenedicarboxylic acid.154 In vitro of the ferrocenyl compounds, together with imaging agents
studies demonstrated that the thermoresponsive ferrocene- and/or other drugs, opening the door for the use of these
containing shell cross-linked micelles exhibited significant compounds in theranostics.
cytotoxicity against HeLa cells, due to the presence of the
ferrocenyl groups.154
Acknowledgements
8.4. Lipid nanocapsules loaded with dihydroxyferrocifen
I am grateful to Prof. Jean M. J. Fréchet and National Science
Lipid nanocapsules (LNC) of around 50 nm were used to Foundation (NSF) for support, and to Dr Jackson D. Megiatto
encapsulate the active ferrocenyl diphenol tamoxifen derivative Jr for useful discussions about this review.
(Fc-diOH, see Section 5.1), in order to increase its solubility in
biological media.155 The Fc-diOH was encapsulated in high
loading because of the large hydrophobic core in the LNC Notes and references
structure. The loaded LNCs were up taken by 9L-glioma cells 1 T. J. Kealy and P. L. Pauson, Nature, 1951, 168, 1039–1040.
and the cytostatic activity of the Fc-diOH was conserved 2 S. A. Miller, J. A. Tebboth and J. F. Tremaine, J. Chem. Soc.,
presenting an IC50 value of about 0.6 mM. Interestingly, the 1952, 632–635.
Fc-diOH-loaded LNCs showed low toxicity levels when in 3 G. Wilkinson, M. Rosenblum, M. C. Whiting and R. B. Woodward,
J. Am. Chem. Soc., 1952, 74, 2125–2126.
contact with healthy cells, conferring a functional specificity of 4 E. O. Fischer and W. Z. Pfab, Z. Naturforsch., B: Anorg. Chem.
this compound for tumor cells. Promising in vivo experiments Org. Chem. Biochem. Biophys. Biol., 1952, 7b, 377.
in rats demonstrated that the Fc-diOH-loaded LNC dramati- 5 R. B. Woodward, M. Rosenblum and M. C. Whiting, J. Am.
Chem. Soc., 1952, 74, 3458–3459.
cally reduced the tumor mass and glioma volume, showing for 6 M. Rosenblum, Chemistry of the Iron-Group Metallocenes, Part I,
the first time the in vivo efficacy of hydroxyferrocifens.155 Interscience, New York, 1965.
This work was then taken to the next level by evaluating the 7 A. Togni and T. Hayashi, Ferrocenes, Wiley-VCH, Weinheim,
in vivo efficacy of the Fc-diOH-loaded LNCs in combination 1994.
8 R. D. Adams, J. Organomet. Chem., 2001, 637–639, 1.
with external radiotherapy in the intracerebral 9L-glioma 9 S. P. Hendry, M. F. Cardosi, A. P. F. Turner and E. W. Neuse,
model.156 This combination resulted in a significantly longer Anal. Chim. Acta, 1993, 281, 453–459.
survival rate when compared to Fc-diOH-LNC-treated animals 10 A. Labande and D. Astruc, Chem. Commun., 2000, 1007–1008.
and to the group treated with blank LNCs + radiotherapy, 11 A. Labande, J. Ruiz and D. Astruc, J. Am. Chem. Soc., 2002, 124,
1782–1789.
suggesting a synergetic antitumor activity on 9L-glioma cells, 12 M. C. Daniel, J. Ruiz and D. Astruc, J. Am. Chem. Soc., 2003,
probably caused by a closely oxidative relationship.156 125, 1150–1151.

1982 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
View Article Online

13 D. Astruc, M. C. Daniel and J. Ruiz, Chem. Commun., 2004, 48 L. V. Popova, V. N. Babin, Y. A. Belousov, Y. S. Nekrasov,
2637–2649. A. E. Snegireva, N. P. Borodina, G. M. Shaposhnikova,
14 M. P. G. Armada, J. Losada, M. Zamora, B. Alonso, O. B. Bychenko, P. M. Raevskii, N. B. Morozova, A. I.
I. Cuadrado and C. M. Casado, Bioelectrochemistry, 2006, 69, Iiyina and K. G. Shitkov, Appl. Organomet. Chem., 1993, 7,
65–73. 85–94.
15 C. Ornelas, J. R. Aranzaes, E. Cloutet, S. Alves and D. Astruc, 49 P. Köpf-Maier and H. Köpf, Bioinorganic Chemistry, Springer
Angew. Chem., Int. Ed., 2007, 46, 872–877. Berlin/Heidelberg, 1988, vol. 70, pp. 103–185.
16 D. Astruc, C. Ornelas and J. R. Aranzaes, J. Inorg. Organomet. 50 E. W. Neuse and F. Kanzawa, Appl. Organomet. Chem., 1990, 4,
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

Polym. Mater., 2008, 18, 4–17. 19–26.


17 C. Ornelas, J. Ruiz and D. Astruc, Organometallics, 2009, 28, 51 H. Tamura and M. Miwa, Chem. Lett., 1997, 1177–1178.
4431–4437. 52 A. Houlton, R. M. G. Roberts and J. Silver, J. Organomet.
18 R. Djeda, A. Rapakousiou, L. Liang, N. Guidolin, J. Ruiz and Chem., 1991, 418, 107–112.
D. Astruc, Angew. Chem., Int. Ed., 2010, 49, 8152–8156. 53 D. Osella, M. Ferrali, P. Zanello, F. Laschi, M. Fontani, C. Nervi
19 Ferrocenes Homogeneous Catalysis Organic Synthesis Material and G. Cavigiolio, Inorg. Chim. Acta, 2000, 306, 42–48.
Science, ed. A. Togni and T. Hayashi, VCH, Weinheim, 1995. 54 L. V. Snegur, A. A. Simenel, Y. S. Nekrasov, E. A. Morozova,
20 V. Wei, R. Vajtai, Y. Y. Choi, P. M. Ajayan, H. Zhu, C. Xu and Z. A. Starikova, S. M. Peregudova, Y. V. Kuzmenko, V. N.
D. Wu, Nano Lett., 2002, 2, 1105–1107. Babin, L. A. Ostrovskaya, N. V. Bluchterova and M. M. Fomina,
21 X. Zhang, A. Cao, B. Wei, Y. Li, J. Wei, C. Xu and D. Wu, Chem. J. Organomet. Chem., 2004, 689, 2473–2479.
Phys. Lett., 2002, 362, 285–290. 55 L. V. Snegur, Y. S. Nekrasov, N. S. Sergeeva, Z. V. Zhilina,
22 C. Ornelas, L. Salmon, J. R. Aranzaes and D. Astruc, Chem. V. V. Gumenyuk, Z. A. Starikova, A. A. Simenel, N. B. Morozova,
Commun., 2007, 4946–4948. I. K. Sviridova and V. N. Babin, Appl. Organomet. Chem., 2008,
23 A. K. Diallo, C. Ornelas, L. Salmon, J. R. Aranzaes and 22, 139–147.
D. Astruc, Angew. Chem., Int. Ed., 2007, 46, 8644–8648. 56 A. A. Simenel, S. V. Samarina, L. V. Snegur, Z. A. Starikova,
24 C. Ornelas, J. R. Aranzaes, L. Salmon and D. Astruc, Chem.–Eur. J., L. A. Ostrovskaya, N. V. Bluchterova and M. M. Fomina,
2008, 14, 50–64. Appl. Organomet. Chem., 2008, 22, 276–280.
25 M. C. Daniel, A. Sakamoto, J. Ruiz, D. Astruc and H. Nishihara, 57 A. J. Corry, A. Goel, S. R. Alley, P. N. Kelly, D. O’Sullivan,
Chem. Lett., 2006, 38–39. D. Savage and P. T. M. Kenny, J. Organomet. Chem., 2007, 692,
26 D. Astruc, C. Ornelas and J. Ruiz, Acc. Chem. Res., 2008, 41, 1405–1410.
841–856. 58 A. Goel, D. Savage, S. R. Alley, P. N. Kelly, D. O’Sullivan,
27 C. Ornelas, J. Ruiz, C. Belin and D. Astruc, J. Am. Chem. Soc., H. Mueller-Bunz and P. T. M. Kenny, J. Organomet. Chem.,
2008, 131, 590–601. 2007, 692, 1292–1299.
28 A. Wang, C. Ornelas, D. Astruc and P. Hapiot, J. Am. Chem. 59 A. Mooney, A. J. Corry, D. O’Sullivan, D. K. Rai and P. T. M.
Soc., 2009, 131, 6652–6653. Kenny, J. Organomet. Chem., 2009, 694, 886–894.
29 D. Astruc, C. Ornelas and J. Ruiz, Chem.–Eur. J., 2009, 15, 60 A. Mooney, A. J. Corry, C. N. Ruairc, T. Mahgoub,
8936–8944. D. O’Sullivan, N. O’Donovan, J. Crown, S. Varughese, S. M.
30 J. D. Megiatto, K. Li, D. I. Schuster, A. Palkar, M. A. n. Draper, D. K. Rai and P. T. M. Kenny, Dalton Trans., 2010,
Herranz, L. Echegoyen, S. Abwandner, G. de Miguel and 39, 8228–8239.
D. M. Guldi, J. Phys. Chem. B, 2010, 114, 14408–14419. 61 P. Meunier, I. Ouattara, B. Gautheron, J. Tirouflet, D. Camboli
31 E. W. Neuse, Adv. Macromol. Chem., 1968, 1, 1–138. and J. Besançon, Eur. J. Med. Chem., 1991, 26, 351–362.
32 E. W. Neuse, J. R. Woodhouse, G. Montaudo and C. Puglisi, 62 A. A. Simenel, E. A. Morozova, L. V. Snegur, S. I. Zykova,
Appl. Organomet. Chem., 1988, 2, 53–57. V. V. Kachala, L. A. Ostrovskaya, N. V. Bluchterova and
33 J. C. Swarts, E. W. Neuse and G. J. Lamprecht, J. Inorg. M. M. Fomina, Appl. Organomet. Chem., 2009, 23, 219–224.
Organomet. Polym., 1994, 4, 143–153. 63 A. A. Simenel, G. A. Dokuchaeva, L. V. Snegur, A. N. Rodionov,
34 D. R. van Staveren and N. Metzler-Nolte, Chem. Rev., 2004, 104, M. M. Ilyin, S. I. Zykova, L. A. Ostrovskaya, N. V. Bluchterova,
5931–5986. M. M. Fomina and V. A. Rikova, Appl. Organomet. Chem., 2011,
35 (a) C. S. Allardyce, A. Dorcier, C. Scolaro and P. J. Dyson, Appl. 25, 70–75.
Organomet. Chem., 2005, 19, 1–10; (b) L. V. Snegur, V. N. Babin, 64 M. J. Kelner, T. C. McMorris, W. T. Beck, J. M. Zamora and
A. A. Simenel, Y. S. Nekrasov, L. A. Ostrovskaya and N. S. R. Taetle, Cancer Res., 1987, 47, 3186–3189.
Sergeeva, Russ. Chem. Bull., Int. Ed., 2010, 59, 2167–2178. 65 F. R. Kinder, R.-M. Wang, W. E. Bauta and K. W. Bair, Bioorg.
36 E. W. Neuse, J. Inorg. Organomet. Polym. Mater., 2005, Med. Chem. Lett., 1996, 6, 1029–1034.
15, 3–32. 66 M. J. Kelner, T. C. McMorris, L. Estes, W. Wnag, K. M. Samson
37 M. F. R. Fouda, M. M. Abd-Elzaher, R. A. Abdelsamaia and and R. Taetle, Invest. New Drugs, 1996, 14, 161–167.
A. A. Labib, Appl. Organomet. Chem., 2007, 21, 613–625. 67 S. Knauer, B. Biersack, M. Zoldakova, K. Effenberger, W. Milius
38 A. Nguyen, A. Vessières, E. A. Hillard, S. Top, P. Pigeon and and R. Schobert, Anti-Cancer Drugs, 2009, 20, 676–681.
G. Jaouen, Chimia, 2007, 61, 716–724. 68 J. Paik, S. Vogel, R. Piantedosi, A. Sykes, W. S. Blaner and
39 E. A. Hillard, A. Vessières and G. Jaouen, Top. Organomet. K. Swisshelm, Biochemistry, 2000, 39, 8073–8084.
Chem., 2010, 32, 81–117. 69 A. Nudelman and A. Rephaeli, J. Med. Chem., 2000, 43,
40 C. Biot, N. François, L. Maciejewski, J. Brocard and D. Poulain, 2962–2966.
Bioorg. Med. Chem. Lett., 2000, 10, 839–841. 70 B. Long, S. Liang, D. Xin, Y. Yang and J. Xiang, Eur. J. Med.
41 J. Zhang, Appl. Organomet. Chem., 2008, 22, 6–11. Chem., 2009, 44, 2572–2576.
42 C. Biot, G. Glorian, L. A. Maciejewski, J. S. Brocard, O. Domarle, 71 C.-W. Ong, J.-Y. Jeng, S.-S. Juang and C.-F. Chen, Bioorg. Med.
G. Blampain, P. Millet, A. J. Georges, H. Abessolo, D. Dive and Chem. Lett., 1992, 2, 929–932.
J. Lebibi, J. Med. Chem., 1997, 40, 3715–3718. 72 J. Roca, Trends Biochem. Sci., 1995, 20, 156–160.
43 L. Delhaes, C. Biot, L. Berry, L. A. Maciejewski, D. Camus, 73 Y. N. Vashisht Gopal, D. Jayaraju and A. K. Kondapi, Arch.
J. S. Brocard and D. Dive, Bioorg. Med. Chem., 2000, 8, Biochem. Biophys., 2000, 376, 229–235.
2739–2745. 74 A. D. Sai Krishna, G. Panda and A. K. Kondapi, Arch. Biochem.
44 T. Itoh, S. Shirakami, N. Ishida, Y. Yamashita, T. Yoshida, Biophys., 2005, 438, 206–216.
H.-S. Kim and Y. Wataya, Bioorg. Med. Chem. Lett., 2000, 10, 75 V. Craig Jordan, Curr. Probl. Cancer, 1992, 16, 134–176.
1657–1659. 76 S. Top, J. Tang, A. Vessières, D. Carrez, C. Provot and
45 A. K. Kondapi, N. Satyanarayana and A. D. Saikrishna, Arch. G. Jaouen, Chem. Commun., 1996, 955–956.
Biochem. Biophys., 2006, 450, 123–132. 77 S. Top, B. Dauer, J. Vaissermann and G. Jaouen, J. Organomet.
46 V. J. Fiorina, R. J. Dubois and S. Brynes, J. Med. Chem., 1978, Chem., 1997, 541, 355–361.
21, 393–395. 78 S. Top, A. Vessières, G. Leclercq, J. Quivy, J. Tang,
47 P. Köpf-Maier, H. Köpf and E. W. Neuse, J. Cancer Res. Clin. J. Vaissermann, M. Huché and G. Jaouen, Chem.–Eur. J., 2003,
Oncol., 1984, 108, 336–340. 9, 5223–5236.

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1983
View Article Online

79 E. Hillard, A. Vessières, F. Le Bideau, D. Plażuk, D. Spera, 108 M. Viotte, B. Gautheron, M. M. Kubicki, I. E. Nifant’ev and
M. Huché and G. Jaouen, ChemMedChem, 2006, 1, 551–559. S. P. Fricker, Met.-Based Drugs, 1995, 2, 311–326.
80 A. Nguyen, S. Top, P. Pigeon, A. Vessières, E. A. Hillard, 109 M. Viotte, B. Gautheron, I. Nifant’ev and L. G. Kuz’mina, Inorg.
M.-A. Plamont, M. Huché, C. Rigamonti and G. Jaouen, Chim. Acta, 1996, 253, 71–76.
Chem.–Eur. J., 2009, 15, 684–696. 110 T. A. K. Al-Allaf and L. J. Rashan, Appl. Organomet. Chem.,
81 I. Shiina, Y. Sano, K. Nakata, T. Kikuchi, A. Sasaki, M. Ikekita, 1999, 13, 63–68.
Y. Nagahara, Y. Hasome, T. Yamori and K. Yamazaki, Biochem. 111 W. Henderson and S. R. Alley, Inorg. Chim. Acta, 2001, 322,
Pharmacol., 2008, 75, 1014–1026. 106–112.
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

82 E. A. Hillard, P. Pigeon, A. Vessières, C. Amatore and G. Jaouen, 112 J. Rajput, J. R. Moss, A. T. Hutton, D. T. Hendricks,
Dalton Trans., 2007, 5073–5081. C. E. Arendse and C. Imrie, J. Organomet. Chem., 2004, 689,
83 E. A. Hillard, A. Vessières, S. Top, P. Pigeon, K. Kowalski, 1553–1568.
M. Huché and G. Jaouen, J. Organomet. Chem., 2007, 692, 113 J. Spencer, A. Casini, O. Zava, R. P. Rathnam, S. K. Velhanda,
1315–1326. M. Pfeffer, S. K. Callear, M. B. Hursthouse and P. J. Dyson,
84 J. B. Heilmann, E. A. Hillard, M.-A. Plamont, P. Pigeon, Dalton Trans., 2009, 10731–10735.
M. Bolte, G. Jaouen and A. Vessières, J. Organomet. Chem., 114 C. Bincoletto, I. L. S. Tersariol, C. R. Oliveira, S. Dreher,
2008, 693, 1716–1722. D. M. Fausto, M. A. Soufen, F. D. Nascimento and A. C. F.
85 G. Jaouen, S. Top, A. Vessières, P. Pigeon, G. Leclercq and Caires, Bioorg. Med. Chem., 2005, 13, 3047–3055.
I. Laios, Chem. Commun., 2001, 383–384. 115 C. M. V. Barbosa, C. R. Oliveira, F. D. Nascimento, M. C. M.
86 S. Top, A. Vessières, P. Pigeon, M.-N. Rager, M. Huché, Smith, D. M. Fausto, M. A. Soufen, E. Sena, R. C. Araújo,
E. Salomon, C. Cabestaing, J. Vaissermann and G. Jaouen, I. L. S. Tersariol, C. Bincoletto and A. C. F. Caires, Eur. J.
ChemBioChem, 2004, 5, 1104–1113. Pharmacol., 2006, 542, 37–47.
87 E. Hillard, A. Vessières, L. Thouin, G. Jaouen and C. Amatore, 116 A. C. F. Caires, Anti-Cancer Agents Med. Chem., 2007, 7,
Angew. Chem., Int. Ed., 2006, 45, 285–290. 484–491.
88 O. Zekri, E. A. Hillard, S. Top, A. Vessières, P. Pigeon, 117 M. Viotte, B. Gautheron, M. M. Kubicki, Y. Mugnier and
M.-A. Plamont, M. Huché, S. Boutamine, M. J. McGlinchey, R. V. Parish, Inorg. Chem., 1995, 34, 3465–3473.
H. Muller-Bunz and G. Jaouen, Dalton Trans., 2009, 4318–4326. 118 M. V. Baker, P. J. Barnard, S. J. Berners-Price, S. K. Brayshaw,
89 P. Pigeon, S. Top, O. Zekri, E. A. Hillard, A. Vessières, J. L. Hickey, B. W. Skelton and A. H. White, Dalton Trans., 2006,
M.-A. Plamont, O. Buriez, E. Labbé, M. Huché, S. Boutamine, 3708–3715.
C. Amatore and G. Jaouen, J. Organomet. Chem., 2009, 694, 119 U. E. I. Horvath, G. Bentivoglio, M. Hummel, H. Schottenberger,
895–901. K. Wurst, M. J. Nell, C. E. J. van Rensburg, S. Cronje and
90 G. Erker, Macromol. Symp., 2006, 236, 1–13. H. G. Raubenheimer, New J. Chem., 2008, 32, 533–539.
91 D. Plazuk, A. Vessières, E. A. Hillard, O. Buriez, E. Labbé, 120 M. Galanski, V. B. Arion, M. A. Jakupec and B. K. Keppler,
P. Pigeon, M.-A. Plamont, C. Amatore, J. Zakrzewski and Curr. Pharm. Des., 2003, 9, 2078–2089.
G. r. Jaouen, J. Med. Chem., 2009, 52, 4964–4967. 121 C. G. Hartinger, S. Zorbas-Seifried, M. A. Jakupec, B. Kynast,
92 M. Görmen, P. Pigeon, S. Top, E. A. Hillard, M. Huché, H. Zorbas and B. K. Keppler, J. Inorg. Biochem., 2006, 100,
C. G. Hartinger, F. de Montigny, M.-A. Plamont, A. Vessières 891–904.
and G. Jaouen, ChemMedChem, 2010, 5, 2039–2050. 122 W. Han Ang and P. J. Dyson, Eur. J. Inorg. Chem., 2006,
93 M. Gormen, D. Plazuk, P. Pigeon, E. A. Hillard, M.-A. Plamont, 4003–4018.
S. Top, A. Vessières and G. Jaouen, Tetrahedron Lett., 2010, 51, 123 I. Kostova, Curr. Med. Chem., 2006, 13, 1085–1107.
118–120. 124 A. R. Timerbaev, C. G. Hartinger, S. S. Aleksenko and
94 A. P. Ferreira, J. L. F. da Silva, M. T. Duarte, M. F. M. da B. K. Keppler, Chem. Rev., 2006, 106, 2224–2248.
Piedade, M. P. Robalo, S. G. Harjivan, C. Marzano, 125 G. Von Poelhsitz, A. L. Bogado, M. P. de Araujo, H. S.
V. Gandin and M. M. Marques, Organometallics, 2009, 28, Selistre-de-Araújo, J. Ellena, E. E. Castellano and A. A. Batista,
5412–5423. Polyhedron, 2007, 26, 4707–4712.
95 A. Vessières, D. Spera, S. Top, B. Misterkiewicz, J.-M. Heldt, 126 M. Auzias, B. Therrien, G. Suss-Fink, P. Štěpnička, W. H. Ang
E. Hillard, M. Huché, M.-A. Plamont, E. Napolitano, R. Fiaschi and P. J. Dyson, Inorg. Chem., 2008, 47, 578–583.
and G. Jaouen, ChemMedChem, 2006, 1, 1275–1281. 127 M. Auzias, J. Gueniat, B. Therrien, G. Süss-Fink, A. K.
96 A. Vessières, S. Top, W. Beck, E. Hillard and G. Jaouen, Dalton Renfrew and P. J. Dyson, J. Organomet. Chem., 2009, 694,
Trans., 2006, 529–541. 855–861.
97 S. Top, C. Thibaudeau, A. Vessières, E. Brulé, F. Le Bideau, 128 I. Ott, K. Kowalski, R. Gust, J. Maurer, P. Mücke and
J.-M. Joerger, M.-A. Plamont, S. Samreth, A. Edgar, J. r. m. R. F. Winter, Bioorg. Med. Chem. Lett., 2010, 20, 866–869.
Marrot, P. Herson and G. Jaouen, Organometallics, 2009, 28, 129 W. C. M. Duivenvoorden, Y.-n. Liu, G. Schatte and
1414–1424. H.-B. Kraatz, Inorg. Chim. Acta, 2005, 358, 3183–3189.
98 J. Manosroi, K. Rueanto, K. Boonpisuttinant, W. Manosroi, 130 M. Abd-Elzaher, S. Moustafa, A. Labib and M. Ali, Monatsh.
C. Biot, H. Akazawa, T. Akihisa, W. Issarangporn and A. Manosroi, Chem., 2010, 141, 387–393.
J. Med. Chem., 2010, 53, 3937–3943. 131 K. E. Uhrich, S. M. Cannizzaro, R. S. Langer and
99 O. Payen, S. Top, A. Vessières, E. Brulé, M.-A. Plamont, M. J. K. M. Shakesheff, Chem. Rev., 1999, 99, 3181–3198.
McGlinchey, H. Müller-Bunz and G. Jaouen, J. Med. Chem., 132 M. C. Daniel and D. Astruc, Chem. Rev., 2004, 104, 293–346.
2008, 51, 1791–1799. 133 C. C. Lee, J. A. MacKay, J. M. J. Frechet and F. C. Szoka,
100 B. Rosenberg, L. Vancamp, J. E. Trosko and V. H. Mansour, Nat. Biotechnol., 2005, 23, 1517–1526.
Nature, 1969, 222, 385–386. 134 S. Svenson and D. A. Tomalia, Adv. Drug Delivery Rev., 2005, 57,
101 D. Lebwohl and R. Canetta, Eur. J. Cancer, 1998, 34, 1522–1534. 2106–2129.
102 E. Wong and C. M. Giandomenico, Chem. Rev., 1999, 99, 135 B. Helms and E. W. Meijer, Science, 2006, 313, 929–930.
2451–2466. 136 C. Ornelas, E. Boisselier, V. Martinez, I. Pianet, J. R. Aranzaes
103 M. J. Clarke, F. Zhu and D. R. Frasca, Chem. Rev., 1999, 99, and D. Astruc, Chem. Commun., 2007, 5093–5095.
2511–2534. 137 Biomedical Nanostructures, ed. C. R. H. Kenneth, E. Gonsalves,
104 V. Scarcia, A. Furlani, B. Longato, B. Corain and G. Pilloni, Cato T. Laurencin and Lakshmi S. Nair, John Wiley & Sons, Inc.,
Inorg. Chim. Acta, 1988, 153, 67–70. 2008.
105 E. W. Neuse, M. G. Meirim and N. F. Blom, Organometallics, 138 I. J. Majoros, C. R. Williams and J. R. Baker, Curr. Top. Med.
1988, 7, 2562–2565. Chem., 2008, 8, 1165–1179.
106 D. T. Hill, G. R. Girard, F. L. McCabe, R. K. Johnson, 139 J. B. Wolinsky and M. W. Grinstaff, Adv. Drug Delivery Rev.,
P. D. Stupik, J. H. Zhang, W. M. Reiff and D. S. Eggleston, 2008, 60, 1037–1055.
Inorg. Chem., 1989, 28, 3529–3533. 140 S. F. M. van Dongen, H.-P. M. de Hoog, R. J. R. W. Peters,
107 A. Rosenfeld, J. Blum, D. Gibson and A. Ramu, Inorg. Chim. M. Nallani, R. J. M. Nolte and J. C. M. van Hest, Chem. Rev.,
Acta, 1992, 201, 219–221. 2009, 109, 6212–6274.

1984 New J. Chem., 2011, 35, 1973–1985 This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011
View Article Online

141 R. K. Tekade, P. V. Kumar and N. K. Jain, Chem. Rev., 2009, 151 E. W. Neuse, Macromol. Symp., 2001, 172, 127–138.
109, 49–87. 152 M. T. Johnson, E. Kreft, D. D. N’Da, E. W. Neuse and C. E. J.
142 S. H. Medina and M. E. H. El-Sayed, Chem. Rev., 2009, 109, van Rensburg, J. Inorg. Organomet. Polym., 2003, 13, 255–267.
3141–3157. 153 A. Nguyen, V. Marsaud, C. Bouclier, S. Top, A. Vessières,
143 C. Ornelas and M. Weck, Chem. Commun., 2009, 5710–5712. P. Pigeon, R. Gref, P. Legrand, G. Jaouen and J.-M. Renoir,
144 M. E. Fox., F. C. Szoka and J. M. J. Fréchet, Acc. Chem. Res., Int. J. Pharm., 2008, 347, 128–135.
2009, 42, 1141–1151. 154 H. Wei, C.-Y. Quan, C. Chang, X.-Z. Zhang and R.-X. Zhuo,
145 C. Ornelas, J. Broichhagen and M. Weck, J. Am. Chem. Soc., J. Phys. Chem. B, 2010, 114, 5309–5314.
Published on 16 June 2011. Downloaded by University of California - Santa Barbara on 7/8/2018 3:43:19 PM.

2010, 132, 3923–3931. 155 E. Allard, C. Passirani, E. Garcion, P. Pigeon, A. Vessières,


146 C. Ornelas, R. Pennell, L. F. Liebes and M. Weck, Org. Lett., G. Jaouen and J.-P. Benoit, J. Controlled Release, 2008, 130,
2011, 13, 976–979. 146–153.
147 O. Buriez, J. M. Heldt, E. Labbé, A. Vessières, G. Jaouen and 156 E. Allard, D. Jarnet, A. Vessières, S. Vinchon-Petit, G. Jaouen,
C. Amatore, Chem.–Eur. J., 2008, 14, 8195–8203. J.-P. Benoit and C. Passirani, Pharm. Res., 2010, 27, 56–64.
148 Z. Petrovski, M. R. P. Norton de Matos, S. S. Braga, C. C. L. 157 T. Loftsson and M. E. Brewster, J. Pharm. Sci., 1996, 85,
Pereira, M. L. Matos, I. S. Gonçalves, M. Pillinger, P. M. Alves 1017–1025.
and C. C. Romão, J. Organomet. Chem., 2008, 693, 675–684. 158 T. Utsuki, H. Brem, J. Pitha, T. Loftsson, T. Kristmundsdottir,
149 G. Caldwell, M. G. Meirim, E. W. Neuse and C. E. J. B. M. Tyler and A. Olivi, J. Controlled Release, 1996, 40, 251–260.
van Rensburg, Appl. Organomet. Chem., 1998, 12, 793–799. 159 F. Hapiot, S. Tilloy and E. Monflier, Chem. Rev., 2006, 106,
150 E. W. Neuse, Polym. Adv. Technol., 1998, 9, 786–793. 767–781.

This journal is c The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2011 New J. Chem., 2011, 35, 1973–1985 1985

You might also like