Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

Ruthenium compounds as

anticancer agents
New ruthenium-based compounds with fewer and less severe side effects, could replace
longstanding platinum-based anticancer drugs
Simon Page

In short This observation led to cis-


diamminedichloroplatinum(II),
●● As clinical trials
commonly known as cisplatin, being
progress, ruthenium
compounds may
approved by the American Food and
provide a less toxic and Drugs Administration (FDA) for
more effective cancer therapy in 1978. It has since
alternative to become the most widely used
platinum therapies anticancer drug, with an estimated
●● Many different
70% of patients receiving the
ruthenium compounds compound as part of their treatment.
1

have been tested for


their anticancer Cisplatin
properties, without Cisplatin was heralded as a completely
enough investigation novel type of antitumour agent, and its
into their mode(s) of discovery enticed a veritable army of
action inorganic chemists to devise and test
other precious metal-based therapies.
Disappointingly however, fifty years
and countless clinical candidates later,
there have only been two more
worldwide drug approvals for
precious metal-containing anticancer
drugs: carboplatin and oxaliplatin –
both of which are direct analogues of
cisplatin (fig 1) – approved in 1993
and 2002 respectively.
The mode of action of these
platinum complexes is known: the
chloride or dicarboxylate ligands are
hydrolysed within the cell to
generate a bis-aqua species, which
binds irreversibly to DNA – usually
to two adjacent guanine bases –
shutterstock

(fig 2) and the cell, unable to


replicate, defaults to apoptosis (that
is, controlled cell death).

Platinum therapies Rosenberg used a platinum electrode From platinum to ruthenium


For half a century, the field of metal- to apply an electric field to a colony of Whilst the chemotherapeutic success
based anticancer drugs has been E. coli, which was observed to inhibit of platinum is undeniable, it is by no
dominated by the precious metal their growth. A diligent investigation means the perfect drug. It is not
platinum. The discovery of platinum’s into the cause of this effect concluded effective against many common types
anticancer properties was made by that the platinum electrode was of cancer, drug resistance is common
chance during an experiment in 1965, breaking down to generate and it has a deplorable range of side
conducted at Michigan State platinum(II) species in situ, which was effects, which can include nerve
University by Barnett Rosenberg. stopping the cells from multiplying. damage, hair loss and nausea.

26 | Education in Chemistry | January 2012 www.rsc.org/eic


1 O H concentration of molecular oxygen
H2N Cl O N (due to their higher metabolic rate and
H2N O O
Pt Pt Pt remoteness from the blood supply).
H2N Cl H2N O O O
N These two factors taken in parallel
O H mean that compounds of ruthenium
can be administered in the (relatively
To overcome these limitations, Fig 1 Furthermore, ruthenium tends to inert) III oxidation state, causing
some compounds based on ruthenium The chemical form octahedral complexes, which minimal damage to healthy cells, but
have been developed and tested structures of gives the chemist two more ligands to being reduced to the (active) II
against cancer cell lines. These cisplatin, exploit compared with platinum(II) oxidation state in cancer cells.3
compounds tend to cause fewer (and carboplatin and complexes, which adopt a square Recently, however, this theory has
less severe) side effects compared to oxaliplatin planar geometry. Ruthenium can also come under considerable criticism
platinum drugs. form strong chemical bonds with a and even the question of how
Ruthenium’s properties are well range of different elements of varying ruthenium compounds enter cells has
suited towards pharmacological chemical ‘hardness’ and been the subject of some literature
debate.
2
Can ruthenium impersonate
iron?
Ruthenium is a transition metal in
group 8 – the same chemical group as
iron. Iron, in spite of its potential
reactivity and biological toxicity, is a
key element without which most cells
cannot survive. In fact, nature has
developed considerable machinery to
sequester, transport and make use of
iron. The fact that iron and ruthenium
are in the same chemical group has
american chemical society

led to some chemists postulating that


it is capable of taking iron’s place in
some proteins, most notably in the
chaperone and uptake protein
transferrin.4 However, others remain
skeptical that the two metals are
applications. It can access a range of Fig 2 electronegativities, meaning that similar enough to be interchangeable;
oxidation states (II, III and IV) under A crystal structure ruthenium can bind to a range of specifically, ruthenium binds tighter
physiologically relevant conditions. showing cisplatin biomolecules, not just DNA. and more slowly than iron and has a
Also, the energy barriers to (red) binding to preference for ‘softer’ ligands. Nature
interconversion between these and ‘kinking’ DNA Why is ruthenium less toxic? has also developed extremely high
oxidation states is relatively low, (grey)2 One hypothesis as to why ruthenium affinity iron binding sites (eg
allowing for ready oxidation state compounds are less toxic in general transferrin holds iron with a binding
changes when inside the cell. In spite than platinum drugs is ‘Activation by constant of 1023 M-1), and it has been
of this flexibility in oxidation state, Reduction’. This theory is based on the questioned whether ruthenium can
ruthenium complexes display observation that ruthenium(III) outcompete iron in these contexts.
relatively slow ligand exchange rates in Fig 3 complexes are more inert than Another suggestion is that ruthenium
water (fig 3) – its kinetics are on the The ligand exchange ruthenium(II), which can partially be can bind to other sites on transferrin,
timescale of cellular reproduction rates of platinum attributed to its higher effective and ‘piggy-back’ into the cell when
(mitosis), meaning that if a ruthenium group metals are nuclear charge (Zeff ). Also, cancerous iron is taken into the cell (fig 4).5
ion does bind to something in the cell, considerably slower cells tend to have a more chemically
it is likely to remain bound for the than those of other reducing environment than healthy Key molecules
remainder of that cell’s lifetime. metals cells, owing to their lower Two ruthenium compounds are
currently undergoing clinical
3 evaluation as anticancer drugs (as far
as the author is aware): NAMI-A and
KP1019 (fig 5). A great deal of the
remaining literature can be
categorised as follows:

Coordination Compounds
Simon Page/J ReedJick

In ‘classical’ coordination compounds,


the metal is surrounded by Lewis
bases with lone pairs. The compounds
KP1019 and NAMI-A fall into this

www.rsc.org/eic January 2012 | Education in Chemistry | 27


4 Fig 6 6
An example of a
RAPTA complex

presence of a facially-coordinated
aromatic ring (which is relatively
hydrophobic) and a PTA
(1,3,5-triaza-7-phosphaadamantane)
ligand (which is highly water
soluble). Oddly, RAPTA complexes
display a similar spectrum of activity
to the coordination complex
NAMI-A, in spite of their apparent
differences in oxidation state, ligands,
charge and geometry.

Fig 4 Multinuclear
Simon page/richard wheeler

Ruthenium The ability of ruthenium species to


‘piggy-backing’ form multinuclear and
into cells on the supramolecular architectures has been
surface of the iron known for some time, but their
uptake protein application to medicinal chemistry has
transferrin. only been explored recently.
Some particularly interesting
category. Given that these compounds few treatment options for secondary strategies include ruthenium-
have some ligands that can feasibly be (metastatic) cancers, and the platinum mixed-metal compounds,6
hydrolysed (eg chloride, DMSO), it is prognosis for patients who develop ruthenium cluster complexes,7
thought that the actual ‘active’ this form of the disease is much worse. ruthenium DNA intercalators8 and
molecules which reach the cancer cell supramolecular ‘Trojan Horses’, which
are unlikely to be the same as the Organometallics contain a cytotoxic payload that is
molecular structures shown (fig 5). An organometallic complex is often released upon entry to the cancer cell9
KP1019 and NAMI-A appear to be defined as a molecule with a distinct (fig 7).
quite similar structurally (both are metal-carbon bond. Although they
Ru(III)+ complexes with chloride and have a reputation as being unstable Directed therapies
heterocyclic ligands and a heterocyclic compounds, better known for Recently, chemists have developed
counterion) yet they display spontaneous combustion than compounds where a precious metal
remarkably different types of therapeutic effects, some ruthenium is chemically linked to an ‘organic
anticancer activity: KP1019 is active organometallics have displayed high directing molecule’ (ODM), which
against primary cancers (ie the main water- and air-stability and an has a known biological target – eg a
tumour mass which forms first in a interesting spectrum of anticancer drug molecule. It has been
patient), whereas NAMI-A is active activity. speculated that the organic molecule
against secondary tumour cells (ie the Arguably the most successful can ‘lead’ the metal into the cell and
metastases which form after cells from ruthenium organometallic anticancer to a specific target. If the metal then
the primary tumour have moved to a complexes have been the so-called binds directly to the cellular target, it
different organ, eg via the ‘RAPTA’ complexes (fig 6). RAPTA could massively increase the potency
bloodstream). Currently there are very complexes are characterised by the of the drug (fig 8). The main
difference between this strategy and
5 the above work is that here the
biological target of the molecule is
known, making this strategy a more
NH rational form of drug design.
HN
N N
Cl Cl
NH
Cl
Fig 5 Biological target of ruthenium
Cl
Cl
Ru
Cl Ru Chemical drugs
N Cl Cl
H HN structures of (left A recent trend in medicinal chemistry
O SH N N
NH H to right) NAMI-A has been a trend away from high-
and KP1019, the throughput approaches to drug
first ruthenium discovery (ie those where vast
anticancer databases of molecules are screened
compounds to against a biological target) towards
enter clinical trials structure-based drug discovery (ie

28 | Education in Chemistry | January 2012 www.rsc.org/eic


7 high affinity for cancer targets, with
far less severe side effects. This
strategy could be highly advantageous
if a target can be identified.
The final and most recent approach
to ruthenium drug design would be
the generation of further
ruthenium-ODM complexes, in which
an organic molecule binds to the
active site of an enzyme and the
Ruthenium cluster complex Ruthenium-platinum mixed attached ruthenium ion binds to a
metal complex nearby residue of the same protein.
The high-energy interaction set up
between the metal and the target
offers medicinal chemists a high-
energy mode of bonding which isn’t
available to ‘traditional’ organic
medicines. The advantage to this
approach is that the compound has a
known (or, at least, desired) biological
target against which enzymological
studies can be performed – such as
enzyme inhibition studies and protein
crystallography.
Of course, in reality, it is likely that
Ruthenium DNA intercalator all three of these approaches will be
followed to a greater or lesser degree,
as more and more chemists enter the
Supramolecular ‘Trojan Horse’ newly emerging field of ruthenium
anticancer drugs. The continued
progress of ruthenium compounds in
those where drug design is based on Fig 7 against different targets and cell lines. clinical trials, and the frequent and
specific structural information about a Selected This approach could include exciting reports of new ruthenium-
biological target) which recent multinuclear structure-activity relationships (SARs) containing drug candidates in the
research estimates can save around ruthenium in order to determine how altering literature point towards a future where
50% of the cost associated with anticancer different functional groups on the medicinal chemists look beyond the
discovering a drug.10 However, a major complexes molecule affect its anticancer activity. classical ‘biological’ elements of
limitation in the field is the lack of an This strategy has the advantage of carbon, hydrogen, nitrogen and
agreed biological target. Currently, the simplicity, but it would be inefficient oxygen, and begin to consider the
two main theories are that ruthenium and a lack of information about the potential of the less explored regions
compounds target DNA or (as yet physiological target of the resultant of the periodic table to generate
unknown) proteins. To date, a great complexes could complicate powerful and effective drugs.
deal of research has been directed at regulatory approval.
generating novel complexes for testing, Alternatively, more research could Simon Page is a PhD student in the Cancer
with relatively little rational design or be directed at eliciting the mode of Research UK PhD Programme in Medicinal
work aimed at elucidating the modes Fig 8 action of existing ruthenium Chemistry at the University of Cambridge
of action of these complexes. An organic molecule compounds. Then, armed with
directs a metal (M) to detailed information about how References
1. A Dorcier et al, Organometallics, 2006, 25, 4090
Future ruthenium drugs a Lewis Basic Amino ruthenium behaves in biological (DOI: 10.1021/om060394o)
In conclusion, ruthenium compounds Acid (B) near the media – what it prefers to bind to, how 2. P Takahara et al, J. Am. Chem. Soc, 1996, 118, 12309
have shown highly promising active site of a it gets into cells etc – chemists can DOI: 10.1021/ja9625079)
anticancer activity in cells, animals target protein begin to design 3. P Schluga et al, Dalton Trans., 2006, 1796
(DOI: 10.1039/B511792E)
and humans. To date, two compounds ruthenium 4. J Vincent and S Love, Biochim. Biophys. Acta, 2011,
are being evaluated in phase II clinical 8 drugs which DOI: 10.1016/j.bbagen.2011.07.003
trials. However, a major limitation in have a 5. S Page et al, Future Med. Chem., 2009, 1, 541
ruthenium drug discovery is their (DOI: 10.4155/fmc.09.25)
6. A Herman et al, Inorg. Chem., 2008, 47, 274
unknown mode of action. This leads (DOI: 10.1021/ic062419h)
to three possible strategies for future 7. B Therrien et al, J. Cluster Sci., 2007, 18, 741
ruthenium drug design. (DOI: 10.1007/s10876-007-0140-y)
Chemists could continue the 8. A Hotze et al, Eur. J. Inorg. Chem., 2005, 2648
current approach, without a clear (DOI: 10.1002/ejic.200500110)
9. B Therrien et al, Angew. Chem. Int. Ed., 2008, 47, 3773
biological target for their compounds,
Simon page

(DOI: 10.1002/anie.200800186)
but instead generating large numbers 10. R Stevens, Nat. Struct. Mol. Biol., 2004, 11, 293
of different compounds to screen (DOI: 10.1038/nsmb0404-293)

www.rsc.org/eic January 2012 | Education in Chemistry | 29

You might also like