You are on page 1of 22

Cancer Chemother Pharmacol

DOI 10.1007/s00280-016-2976-z

REVIEW ARTICLE

Platinum‑based drugs: past, present and future


Shahana Dilruba1 · Ganna V. Kalayda1 

Received: 31 August 2015 / Accepted: 20 January 2016


© Springer-Verlag Berlin Heidelberg 2016

Abstract  Platinum-based drugs cisplatin, carboplatin and Cisplatin, the first platinum anticancer drug
oxaliplatin are widely used in the therapy of human neo-
plasms. Their clinical success is, however, limited due to Cisplatin, cis-diamminedichloroplatinum(II) (Fig. 1), is
severe side effects and intrinsic or acquired resistance to the known as Peyrone’s chloride since the end of nineteenth
treatment. Much effort has been put into the development century (named after Michele Peyrone, who synthesized
of new platinum anticancer complexes, but none of them it first). The cytostatic property of cisplatin was discov-
has reached worldwide clinical application so far. Neda- ered by Barnett Rosenberg in the late 1960s, while he
platin, lobaplatin and heptaplatin received only regional was doing experiments to analyze the effect of electric
approval. Some new platinum complexes and platinum field on bacterial growth. He observed that bacterial pro-
drug formulations are undergoing clinical trials. Here, we liferation was ceased and pinned down the cause of this
review the main classes of new platinum drug candidates, phenomenon to the platinum electrode that was used.
such as sterically hindered complexes, monofunctional Rosenberg and colleagues identified cisplatin as a key
platinum drugs, complexes with biologically active ligands, compound responsible for the anti-proliferative effect.
trans-configured and polynuclear platinum complexes, Clinical trials were initiated in 1971, and after circum-
platinum(IV) prodrugs and platinum-based drug delivery venting a number of obstacles, cisplatin was finally
systems. For each class of compounds, a detailed over- approved for use in testicular and ovarian cancer by the
view of the mechanism of action is given, the cytotoxicity US Food and Drug Administration and in several Euro-
is compared to that of the clinically used platinum drugs, pean countries in 1979 [1].
and the clinical perspectives are discussed. A critical analy- Cisplatin is active against a wide spectrum of solid
sis of lessons to be learned is presented. Finally, a general neoplasms, including ovarian, testicular, bladder, colo-
outlook regarding future directions in the field of new plati- rectal, lung and head and neck cancers [2, 3]. The drug
num drugs is given. often leads to an initial therapeutic response associated
with complete disease remission, partial response or dis-
Keywords  Platinum drugs · Resistance · Toxicity · ease stabilization. However, cisplatin therapy requires
Clinical perspective · Future directions additional medication and is accompanied by severe side
effects including dose-limiting nephrotoxicity, cumula-
tive peripheral sensory neuropathy, ototoxicity due to
irreversible damage of the hair cells in Corti organ, as
well as nausea and vomiting. Furthermore, resistance
to cisplatin represents a major hurdle to the success of
* Ganna V. Kalayda the treatment. Many tumors are intrinsically resistant
akalayda@uni‑bonn.de to the platinum drug. Moreover, many originally sensi-
1
Department of Clinical Pharmacy, Institute of Pharmacy,
tive tumors develop resistance gradually after initial
University of Bonn, An der Immenburg 4, 53121 Bonn, response [4].
Germany

13
Cancer Chemother Pharmacol

Fig. 1  Chemical structures of O H2
the clinically used platinum N O
H3N Cl H3N O O
drugs Pt Pt Pt
H3N Cl O O
H3N N O
O H2

cisplatin carboplatin oxaliplatin

O
O NH2 O O NH2 O
H3N O
Pt Pt
Pt
O NH2 O
H3N O NH2 O O
O

nedaplatin lobaplatin heptaplatin

Fig. 2  Schematic representa-
tion of the mechanism of action
of cisplatin and resistance to
the drug

Mechanism of action of cisplatin and resistance in drug resistance to the treatment (Fig. 2). The mecha-
mechanisms nisms of cisplatin resistance have been classified into pre-
target (i.e., those interfering with cisplatin transport prior
Multiple cellular events appear to contribute to the cyto- to DNA binding), on-target (repair of Pt-DNA lesions),
toxic effect of cisplatin, although DNA platination is seen post-target (cellular events taking place after DNA platina-
as a crucial step. Alterations in any of these events manifest tion) and off-target (alterations in signaling pathways not

13
Cancer Chemother Pharmacol

directly engaged by cisplatin but interfering with cisplatin- by increasing the efflux of drug [16]. Furthermore, over-
induced proapoptotic events) [4]. expression of this transporter seems to correlate with poor
Cisplatin had been long presumed to enter the cells by prognosis [17].
passive diffusion as its uptake is concentration-depend- The activated platinum species also react with nucleo-
ent and non-saturable. However, copper transporter 1 philic centers on purine bases of DNA, in particular, N7
(CTR1), which is a transmembrane protein involved in positions of guanosine and adenosine residues. The two
copper homeostasis, was found to play an important role reactive sites on the platinum center permit the formation
in the uptake of cisplatin as well [5, 6]. Ctr1−/− (CTR1- of a cross-link between two adjacent guanines. In addition,
deficient) mouse embryonic fibroblasts accumulated platinum can coordinate to guanine bases of different DNA
much less cisplatin than their wild-type variants and were strands to form interstrand cross-links. The intrastrand
several times more resistant to the drug [7]. Downregula- 1,2-d(GpG) cross-linking induces a significant distortion
tion of CTR1 is often observed in cisplatin-resistant cells in the DNA double helix [18]. This DNA lesion, which is
[8]. In non-small cell lung cancer patients, low levels of assumed to largely account for cisplatin cytotoxicity, is rec-
CTR1 correlated with poor therapeutic response [9]. On ognized by several cellular proteins leading to the repair,
the other hand, copper-extruding P-type ATPases, ATP7A replication bypass or initiation of apoptosis. Several protein
and ATP7B, were found to participate in cisplatin trans- families are involved in the recognition of Pt-DNA adducts
port as well. Detailed studies showed that ATP7A seques- including non-histone chromosomal high-mobility group
ters cisplatin in vesicular structures preventing further proteins 1 and 2 (HMG1 and HMG2), nucleotide exci-
distribution of the drug, while ATP7B is responsible for sion repair (NER) proteins and mismatch repair (MMR)
cisplatin efflux [6, 10]. These transporters were reported proteins.
to be upregulated in cisplatin-resistant cancer cells [11], HMG proteins are mainly involved in gene regulation
and patients with high levels of ATP7A and ATP7B had and chromatin structure. HMG1 and HMG2 recognize
significantly poorer overall survival [9, 12]. Other trans- intrastrand DNA adducts between adjacent guanines chang-
porters appear to be involved, too. For instance, inhibition ing cell cycle events and subsequently inducing apoptosis
of Na+, K+-ATPase leads to a reduction in cisplatin accu- [19]. HMG proteins also contribute to cisplatin cytotoxic-
mulation, and the transporter is downregulated in cispl- ity by shielding platinum-DNA adducts from repair [20].
atin-resistant ovarian carcinoma cells [13]. And although On the contrary, NER is the pathway employed by cancer
defects in cellular accumulation represent the most fre- cells for removal of platinum-DNA adducts and for DNA
quently and well-documented feature of cell lines selected damage repair. A cellular defect in this pathway resulted
for cisplatin resistance [6], we are still far from having in hypersensitivity to cisplatin and the restoration of this
a complete picture of transport mechanisms influencing pathway reversed the process [21]. On the other hand,
tumor cell sensitivity to the drug. Very recently, the loss enhanced NER activity is associated with cisplatin resist-
of LRRC8A and LRRC8D subunits of the heteromeric ance [4]. Mismatch repair (MMR) protein complex does
volume-regulated anion channels (VRACs) was associ- not actually repair cisplatin-DNA adducts, rather it tries to
ated with resistance to cisplatin [14]. repair DNA damage, but after failing to do so, it initiates
Once inside the cells, where the chloride concen- apoptosis. Downregulation or mutation of MMR genes is
tration is much lower (4–20 mM) than in the blood- consistently documented in context of cisplatin resistance
stream (100 mM), cisplatin is hydrolyzed yielding [22]. Another protein family, poly(ADP-ribosyl)ated pro-
monoaqua [Pt(NH3)2Cl(H2O)]+ and diaqua complexes teins (PARP), critically involved in base excision repair, is
[Pt(NH3)2(H2O)2]2+ [15]. These reactive species bind getting attention, as PARP are highly expressed and consti-
to cytoplasmic nucleophiles such as glutathione (GSH), tutively hyperactivated in cisplatin-resistant cells [23].
methionine, metallothioneins and other cysteine-rich pro- DNA damage activates the signal transduction path-
teins. This leads to the depletion of cytoplasmic antioxi- ways that eventually lead to apoptosis. Post-target resist-
dant reserves resulting in the oxidative stress in cells. On ance results from defects in the signaling pathways as well
the other hand, these nucleophilic species act as scavengers as from problems with the cell death execution machinery
limiting the available reactive cisplatin and thus contribut- itself. The inactivation of tumor suppressor p53 is one of
ing to cisplatin resistance. Elevated levels of glutathione the most predominant mechanisms of post-target resist-
or glutathione-S-transferase, an enzyme mediating cispl- ance, as it is the case in half of the human neoplasms [24].
atin coupling to GSH, were observed in cisplatin-resistant HMG1 and HMG2 facilitate the binding of p53 to DNA to
cells [4]. Platinum–glutathione conjugates were reported induce the transactivation of several target genes involved
to be readily excreted out of the cells by multidrug in cell cycle progression (p21), DNA repair and apoptosis
resistance protein MRP2, a member of the ABC family (Bax). Ovarian cancer patients with a wild-type p53 have a
ATPases. Thus, MRP2 also mediates cisplatin resistance higher probability of recovering following cisplatin-based

13
Cancer Chemother Pharmacol

therapy than the patients with mutations in this protein leaving group. The bidentate oxalate significantly reduces
[25]. Moreover, testicular neoplasms are particularly sensi- reactivity of oxaliplatin and thereby limits the toxic side
tive to cisplatin since they almost lack mutated p53 [26]. effects to peripheral sensory neuropathy [15]. The DACH
The contribution of the off-target mechanisms to cisplatin ligand is more lipophilic increasing passive uptake of oxali-
resistance is increasingly recognized. For example, over- platin compared to cisplatin and carboplatin. Higher lipo-
expression of the ERBB2 protooncogene (also known as philicity may also be a reason why oxaliplatin also employs
HER-2) encoding a member of epidermal growth factor some other routes of cellular entry than first- and second-
receptor (EGFR) family was associated with resistance to generation drugs. Organic cation transporters OCT1 and
cisplatin in non-small cell lung cancer patients [4]. OCT2 have been implicated to mediate oxaliplatin uptake,
as their overexpression significantly increases the cellular
accumulation of oxaliplatin, but not cisplatin or carbopl-
Carboplatin and other second‑generation drugs atin. Colorectal cancer cells overexpress organic cation
transporters, which may explain the efficacy of the oxali-
The second-generation platinum drug carboplatin (cis- platin in this particular type of cancer [31]. The clinical
diammine(1,1-cyclobutane dicarboxylato)platinum(II), relevance of these transporters remains, however, unclear.
Fig. 1) was developed in order to reduce the dose-limiting In some clinical studies, increased expression of OCT2 cor-
toxicity of cisplatin. Carboplatin’s mechanism of action is related with prolonged progression-free survival of patients
similar to that of cisplatin. However, the second-generation with metastatic colorectal cancer [32]. On the other hand,
drug has a lower aquation rate due to the bidentate cyclobu- OCT2 mRNA was barely expressed in nine colorectal can-
tane dicarboxylate ligand [27]. As a result of reduced reac- cer cell lines and in ovarian cancer patients [33]. Regarding
tivity, the neurotoxicity and ototoxicity after carboplatin CTR1, evidence that the transporter is involved in oxalipl-
treatment are much less pronounced. Due to its reduced atin uptake is not that strong as in the case of cisplatin; nev-
toxicity profile, carboplatin is suitable for more aggressive ertheless, acquisition of oxaliplatin resistance was reported
high-dose chemotherapy. This drug is accepted worldwide to be accompanied by CTR1 downregulation. Also reduced
and almost replaced cisplatin in combination regimens with expression of the β1-subunit of Na+, K+-ATPase was found
paclitaxel for treatment of ovarian cancer [28]. But the dose in some oxaliplatin-resistant cells. Copper efflux transport-
is limited by myelosuppression, with thrombocytopenia ers appear to play an important role in oxaliplatin sensitiv-
being more severe than neutropenia and anemia. Carbopl- ity, too. Low levels of ATP7B in colorectal cancer patients
atin has limited efficacy against testicular germ-cell can- were associated with a favorable outcome [33].
cers, squamous cell carcinoma of the head and neck and Similar to cisplatin, oxaliplatin mainly forms cross-
bladder cancer. As a consequence, cisplatin still remains links on the adjacent guanine bases or between guanine
the drug of choice for treating these diseases. As in the case and adenine, however, to a lower extent. Nevertheless,
of cisplatin, tumor resistance to carboplatin poses a major oxaliplatin-DNA adducts are more efficient in inhibition of
clinical problem. The mechanisms underlying carboplatin DNA synthesis. Due to the bulkier DACH ligand, oxalipl-
resistance are generally similar to the mechanisms of cispl- atin induces different conformational distortion on DNA.
atin resistance [29]. The bulkiness and lipophilicity of DACH are considered
Nedaplatin (cis-diammineglycolatoplatinum(II), Fig. 1) responsible for differential processing of oxaliplatin-DNA
has improved toxicity profile compared to cisplatin and adducts. The latter are not recognized by MMR proteins.
pharmacokinetic properties similar to cisplatin [30]. It has Interestingly, it does not lead to decreased cytotoxicity but
limited regional approval (Japan) for treatment of small cell makes oxaliplatin antitumor activity MMR independent.
lung cancer (SCLC), non-small cell lung cancer (NSCLC), Besides, increase in replicative bypass, i.e., DNA synthesis
head and neck and esophageal cancer [15]. bypassing platinum-DNA adducts, was reported not to cor-
relate with cytotoxicity of oxaliplatin [34]. All these factors
result in different activity spectrum of the drug compared
Oxaliplatin and other third‑generation drugs to cisplatin or carboplatin. At present, oxaliplatin is being
evaluated in clinical trials for treatment of gastric, pancre-
The third-generation platinum drug oxaliplatin (Fig. 1) was atic, breast and non-small cell lung cancers [15].
developed to overcome resistance against cisplatin and car- Among other third-generation drugs, lobaplatin and
boplatin. In combination with 5-fluorouracil and folinate, heptaplatin received only regional approval. Lobaplatin
this drug is used as an efficient treatment of adjuvant and (Fig. 1) is approved in China for the therapy of metastatic
metastatic colorectal cancer intrinsically insensitive to cis- breast cancer, chronic myelogenous leukemia and SCLC
platin [15]. Oxaliplatin is a platinum complex with (1R,2R)- [15]. Its dose-limiting toxicity is thrombocytopenia. Hepta-
1,2-diaminocyclohexane (DACH) ligand and oxalate as a platin (Fig. 1) has a bulkier amine ligand but is structurally

13
Cancer Chemother Pharmacol

similar to lobaplatin. The drug was demonstrated to be sta- to that of carboplatin, with thrombocytopenia being a dose-
ble in solution, to have no remarkable toxicity and to retain limiting factor. The new platinum drug showed antitumor
cytotoxic activity in cisplatin-resistant cell lines. Heptapl- activity against platinum-sensitive ovarian cancer in a
atin is currently applied in the Republic of Korea for the Phase II clinical trial [36]. In a Phase II study in SCLC,
treatment of gastric cancer. A Phase III study showed that the therapeutic effect of picoplatin in platinum-sensitive
heptaplatin combination with 5-fluorouracil is comparable and platinum-resistant tumors was evaluated [37]. The
to cisplatin/5-fluorouracil regimen with less severe hemato- observed overall response rates of 8.3 and 15.4 %, respec-
logical side effects [15]. tively, were modest. The median overall survival was 35.7
and 27.3 weeks, respectively. Following these studies, a
larger Phase III SPEAR (Study of Picoplatin Efficacy After
New platinum‑based drugs Relapse) trial has been launched. Unfortunately, it has not
shown any advantage of the picoplatin arm over the con-
Given the toxic side effects and tumor cell resistance trol group (best supportive care) [38]. This disappoint-
against clinically used platinum drugs, much effort was ing outcome may have been caused, at least partly, by an
devoted to the development of platinum compounds, which unbalanced proportion of patients in the control group who
would overcome these disadvantages. Numerous com- received post-study chemotherapy. In addition, the subset
plexes have been synthesized, thoroughly investigated and analysis of refractory patients, i.e., those who showed no
evaluated in cancer cell lines and xenograft models. Some response or relapsed within 45 days after first line of plat-
platinum drug candidates have reached clinical trials. Here, inum-based regimen, has shown a significant advantage
we review the main classes of platinum anticancer com- of the picoplatin treatment in terms of progression-free
plexes in the context of their mechanism of action, cytotox- survival [38]. It is unclear why best supportive care was
icity and clinical perspective. chosen as control instead of drugs like topotecan known to
show activity in the second-line treatment. Apparently, the
choice of the control group has played a decisive role in the
Sterically hindered platinum complexes outcome of the SPEAR trial seriously damaging the clini-
cal perspective of picoplatin.
Since one of the resistance mechanisms is increased cyto- Besides increasing steric hindrance of cisplatin, attempts
solic deactivation of platinum drugs by sulfur-containing to modify oxaliplatin in order to improve its therapeu-
peptides and proteins, researchers aimed at decreasing tic characteristics were made. For this purpose, a series of
reactivity through introduction of a bulky carrier ligand. oxaliplatin analogs with various alkyl substituents was devel-
Decreased reactivity was also expected to improve the oped. A comparative study showed that oxaliplatin deriva-
toxicity profile, as was the case with carboplatin. The tives with higher lipophilicity had a potential to overcome
most prominent compound of this group is picoplatin, cis- oxaliplatin resistance; however, their absolute cytotoxicity
[PtCl2(NH3)(2-methylpyridine)] (ZD0473, Fig. 3). This was decreased [39]. Further evaluation revealed the methyl-
complex was indeed shown to be less susceptible to deac- substituted analogs {(1R,2R,4R)-4-methyl-1,2-cyclohexan-
tivation by intracellular thiols as compared to cisplatin ediamine}oxalatoplatinum(II), KP1537, and {(1R,2R,4S)-
[35]. Picoplatin retains its cytotoxicity in cisplatin- and 4-methyl-1,2-cyclohexanediamine}oxalatoplatinum(II),
oxaliplatin-resistant cell lines independently of the resist- KP1691 (Fig. 3), as the most promising candidates. Their
ance mechanisms prevailing [36]. An interesting feature activity in vitro was better than that of oxaliplatin in plati-
of the drug is that it was the first platinum(II) complex, num-sensitive and platinum-resistant cell lines, with KP1691
which could be administered orally. Applied either orally being more cytotoxic than its isomer [40, 41]. Interestingly,
or intravenously it exhibits antitumor activity in vivo. The of the two platinum complexes, KP1537 accumulated in
combination of picoplatin and paclitaxel was found to act human colon carcinoma cells to a much higher extent than
synergistically [36]. Toxicity profile of picoplatin is similar KP1691. Even though KP1537 was susceptible to extensive

Fig. 3  Chemical structures of H2 H2
some sterically hindered plati- N O N O
O O
num anticancer complexes N Cl Pt Pt
Pt O O
N O N O
H3N Cl H2 H2

picoplatin KP1537 KP1691

13
Cancer Chemother Pharmacol

efflux in contrast to oxaliplatin and KP1691, it induced sig- Platinum(II) complexes with biologically active
nificantly higher amount of early apoptosis and more late ligands
apoptosis as compared to KP1691 [41]. These features of
KP1537 are likely to account for its superior antitumor activ- Introduction of bulky ligands got a new twist with biologi-
ity in vivo. In L1210 leukemia-bearing mice, KP1537 led to cally active carriers. It was shown, for instance, that estro-
an increase in life span of more than 200 % with five out of gen enhances cisplatin cytotoxicity by increasing the levels
six animals being long-term survivors, as compared to 152 of HMG1 protein, which shields platinum-DNA adducts
and 122 % increase in life span for oxaliplatin and KP1691, from repair [43]. As a number of human tumors have estro-
respectively, and two out of six long-term survivors in each gen receptors, various platinum(II) complexes with recep-
case. In another study, oxaliplatin derivatives were evalu- tors substrates were synthesized. The most interesting
ated in colon cancer xenograft models [41]. Since the neces- compound of this group is VP-128 (Fig. 4), an estradiol-
sity of active immune response for oxaliplatin activity was tethered platinum complex with excellent receptor binding
recently shown [42], immunocompetent BALB/c mice have affinity [44]. In ovarian cancer cells, the complex induced
been compared with immunodeficient SCID/BALB/c ani- caspase-independent apoptosis and autophagy due to the
mals. All three drugs have been active in immunocompetent formation of acidic vesicular organelles [45]. Without an
mice and retarded tumor growth significantly, with KP1537 increase in systemic toxicity, VP-128 showed markedly
being the most potent compound. Oxaliplatin has shown no improved in vitro and in vivo activity in ERα (+) breast
activity in immunodeficient mice; in contrast, tumor growth cancer (MCF-7), which has weak sensitivity toward cis-
retardation has been significant for both oxaliplatin analogs platin. In ERα (−) breast carcinoma (MDA-MB-468),
with minor difference between them [41]. Both isomers have VP-128, and not cisplatin, resulted in nuclear accumulation
shown lower systemic toxicity, which allowed to increase the of apoptosis-inducing factor. However, its in vivo activity
maximum tolerated dose [40, 41]. In spite of these promis- was comparable to that of cisplatin [46].
ing findings, more detailed studies are needed to find out A recent report has introduced a glucose-conjugated
whether the methyl-substituted oxaliplatin derivatives are oxaliplatin-based complex Glu-Pt (Fig. 4) designed to
worth further clinical evaluation.

Fig. 4  Chemical structures of OH
some platinum complexes with
biologically active ligands HN
H Cl Pt N
HO Cl
VP-128

H2 O
N O
O O
OH
Pt
F
N O HO OH
H2 O
OH

Glu-Pt

N NH2
N + N

N
(CH2)n
O N
Cl HN H
HN NH2
Pt Pt
Cl N Cl Cl
H2

cis-[Pt{AO(CH2)6en}Cl2]+ 9-aminoac-PtenCl2 (n=2-5)

13
Cancer Chemother Pharmacol

specifically target glucose transporters (GLUTs). The com- Monofunctional platinum complexes
pound shares the mechanistic principles with oxaliplatin
but exhibits improved cytotoxicity. In DBA/2 mice bear- Later on, researchers turned to monofunctional platinum
ing L1210 leukemia xenografts, maximum tolerated dose complexes, i.e., those featuring only one chloride as a
has been sixfold higher resulting in improved efficacy. The leaving group. Such complexes had for a long time been
complex has also shown superior synergistic interaction considered inactive as the parent compound [Pt(dien)Cl]
with 5-fluorouracil and folinate making it interesting for Cl (dien = diethylenetriamine) is not cytotoxic. However,
further evaluation [47]. when complexes with intercalators were concerned, it was
A widely studied group of biologically active ligands presumed that a platinum moiety would be covalently
are intercalators. Combination of platinum with interca- linked to a nucleobase and an intercalator would addition-
lators enables binding to DNA via two different sites— ally interact with DNA forming stable and structurally
through covalent bonds of the platinum moiety and by different adducts than cisplatin. This approach appeared
intercalation of the ligand between DNA base pairs. It to be fruitful and has produced many interesting com-
was expected that such bifunctional complexes would pounds such as PT-ACRAMTU and its amidine derivative
form DNA adducts, which would not be susceptible to (Fig.  5). These complexes rapidly intercalate into DNA
the repair mechanisms. One of the first complexes was followed by platination of a nucleobase at the intercala-
prepared by Lippard and co-workers and contained tion pocket. This mechanism efficiently drives platinum
acridine orange (cis-[Pt{AO(CH2)6en}Cl2]+, Fig. 4). It away from the classical binding sites [51]. Such mono-
showed the same sequence selectivity in cross-linking functional-intercalative adducts induced much less dis-
DNA as cisplatin, and the ligand was found to interca- tortion on DNA than cisplatin-d(GpG)-cross-link result-
late into DNA in the distance of one or two base pairs ing in a decreased or negligible affinity of HMG domain
from the Pt-DNA adduct [48]. Subsequently developed proteins to recognize the adduct [52], which implies
acridine-containing platinum complexes (9-aminoac- differences in adduct processing compared to cisplatin.
PtenCl2, Fig. 4) had different sequence specificity [49] The amidine derivative formed DNA adducts faster than
but failed to show an advantage over cisplatin in terms PT-ACRAMTU. Moreover, these adducts were repaired
of biological activity. The acridine–platinum compounds less efficiently. Rapid and irreversible DNA adduct for-
were more active than cisplatin or than the correspond- mation correlated with higher cytotoxicity. In general,
ing free ligands in CH1 ovarian carcinoma cell lines. PT-ACRAMTU and especially its amidine analog were
However, in contrast to the free acridines platinum com- more active in vitro than cisplatin and the free ligands
plexes exhibited much lower activity in the cisplatin- with the most prominent results obtained in non-small cell
resistant variant [50]. lung cancer cell line NCI-H460. Antitumor activity of the

Fig. 5  Chemical structures of 2+
some monofunctional platinum +
H3N Cl
complexes
H2 NH Pt
N S N
N H3N N
Pt H
N Cl HN
H2
PT-ACRAMTU pyriplatin

2+ +
H3N Cl
NH Pt
H2 H
N N N H3N N
N
Pt H
N Cl
H2

amidine analogue of PT-ACRAMTU phenathriplatin

13
Cancer Chemother Pharmacol

hybrid compounds was attributed to higher cellular accu- Platinum(II) complexes with trans‑geometry
mulation, faster formation and less efficient removal of
DNA adducts as compared to cisplatin [51]. The mono- For a long time, cis-geometry was believed to be a neces-
functional-intercalative adducts were capable of induc- sary feature for platinum complexes to exhibit antitumor
ing cell cycle arrest in the S phase, while cisplatin causes activity. However, increased repair of platinum-induced
cells to accumulate in G2/M phase [53]. Unfortunately, DNA damage by resistant cells turned researchers’ atten-
these advantages were not reflected in in vivo activity. PT- tion to platinum complexes able to build structurally dif-
ACRAMTU failed to slow tumor growth in mice. Moreo- ferent DNA adducts. Such unprecedented adducts were
ver, the compounds were quite toxic to animals with max- supposed to be processed by the cellular machinery in the
imum tolerated doses an order of magnitude lower than in other way compared to the cross-links formed by the con-
the case of cisplatin. Such lack of cancer cell selectivity ventional platinum drugs. Due to their geometry, trans-
makes these complexes unattractive for further clinical platinum complexes are predestined to interact differently
development. Specific delivery to cancer cells may sig- with DNA than their cis-counterparts.
nificantly improve the therapeutic profile of the amidine Transplatin (trans-[Pt(NH3)2Cl2]) is known to lack anti-
analog of PT-ACRAMTU and lead to a promising antitu- tumor activity, which has been attributed to the fast deacti-
mor drug candidate. vation by sulfur proteins [58]. Interestingly, if transplatin
Another group of monofunctional compounds has been is applied to the cells irradiated with UVA light, it forms
developed in the Lippard’s laboratory. They focused on interstrand cross-links and protein–DNA adducts and
complexes with a general formula cis-[Pt(NH3)2AmCl]+, exerts cytotoxic effects comparable to those of cisplatin
where Am was a planar aromatic base like pyridine, [59]. Introduction of the bulky ligands instead of ammonia
purine, pyrimidine or an intercalator. The complexes decreased the rate of chloride exchange positively affecting
with the latter (9-aminoacridine or chloroquine) were the kinetics of DNA adduct formation and antitumor activ-
found highly toxic in preliminary animal studies and ity [58].
were not considered further [54]. Of these complexes, Trans-configured platinum anticancer complexes, apart
[Pt(NH3)2(pyridine)Cl]+, later referred to as pyriplatin from Pt(IV) and polynuclear platinum complexes, can be
(Fig. 5), had a remarkable affinity for organic cation trans- roughly divided into three groups: complexes with planar
porters OCT1 and OCT2, which are believed to contribute aromatic bases, with iminoethers and with aliphatic amines
to oxaliplatin influx. Interestingly, pyriplatin displayed (Fig.  6). All three groups of compounds feature highly
higher cytotoxicity in the cells overexpressing OCT1 than cytotoxic representatives, many of them retaining their
in cells with low levels of the transporter [55]. The com- activity in resistant cell lines [60–62]. A special subgroup
plex forms monofunctional adducts, which induce only of the complexes with planar aromatic bases was developed
little distortion on the DNA double helix [56]. Pyriplatin- substituting chloride leaving groups by carboxylates, e.g.,
DNA adducts are mainly repaired by NER and not by trans-[Pt(OAc)2(NH3)(3-pic)] (Fig. 6) [58]. The water-
MMR or double-strand break repair. NER of the mono- soluble carboxylate analogs were found to be slower in
functional adducts is, however, less efficient than in the DNA binding than their chloride counterparts [61], which
case of cisplatin [54]. As a result, pyriplatin demonstrated did not necessarily result in decreased antitumor activity
a different in vitro activity spectrum as compared to the [63]. Slower interaction with DNA may be compensated by
clinically used platinum drugs. But its cytotoxicity was enhanced cellular accumulation of these compounds [63].
much lower than that of cisplatin in all cell lines tested As expected, trans-platinum complexes formed different
[57]. Since the studies with RNA polymerase II suggested DNA adducts than cisplatin, carboplatin and oxaliplatin.
the relevance of steric hindrance of the amine ligand for Most of them first build monofunctional adducts, which
enzyme inhibition, the structure of the monofunctional tend to persist for a longer period of time. Trans-platinum
complexes was varied to introduce a bulkier ligand. The complexes with planar aromatic bases later form inter-
platinum complex with phenanthridine, later called phen- strand cross-links to a higher degree than cisplatin. As com-
anthriplatin, turned out to be the most potent of the series pared to transplatin, the rate of monofunctional to bifunc-
(Fig. 5) [56]. Its cellular uptake is higher than that of cis- tional adduct conversion is much lower (trans-[PtCl2(NH3)
platin and pyriplatin, and the steric hindrance provides (quin)] adducts persist for 48 h as monofunctional, whereas
protection from thiol deactivation similar to that found the conversion rate for transplatin reaches 70 %) [58]. In
for picoplatin [54]. Phenathriplatin’s spectrum of antitu- contrast, platinum complexes with iminoethers barely form
mor activity on NCI panel of cell lines was different from interstrand cross-links, they build relatively stable mono-
other anticancer agents [56]. However, its activity in vivo functional adducts first [60] later converting to protein–
has not been reported. DNA cross-links [64]. The recognition and the processing

13
Cancer Chemother Pharmacol

Fig. 6  Chemical structures of trans complexes with planar amines


some platinum(II) complexes
with trans-geometry
S O
N N N O
Cl Cl
Pt Pt O Pt
H3N Cl H3N Cl O NH3

trans-[PtCl2(NH3)(quin)] trans-[PtCl2(NH3)(tz)] trans-[Pt(OAc)2(NH3)(3-pic)]

trans complexes with iminoethers

MeO H
N Cl
Me Pt Me
Cl N
H OMe

trans-EE

trans complexes with aliphatic amines

Cl NH3
Cl NH2
H2 Pt
H Pt
Cl N N Cl H
N Cl
Pt
H
N Cl N .HCl
N .HCl

trans-[PtCl2(ipa)(dma)] trans-[PtCl2(NH3)(pip-pip)] trans-[PtCl2(n-butylamine)(pip-pip)]

of the DNA adducts formed by trans-platinum complexes active against P388 leukemia. A platinum complex with
are also different from that of the conventional platinum iminoethers trans-[PtCl2(E-iminoether)2] (trans-EE)
drugs. Many of them are not recognized by HMG proteins decreased the level of lung metastases but less efficiently
and are not repaired by nuclear excision repair system [58, than cisplatin. Treatment with this complex also did not
64]. This explains antitumor activity of trans-complexes in lead to increased survival in mice compared to cisplatin.
platinum-resistant cell lines. Protein–DNA cross-links also Trans-[PtCl2(ipa)(dma)] exhibited no antitumor activity
appear to be of major importance for the anticancer activity in the human ovarian CH1 xenograft, possibly due to its
of these compounds. The biological consequences of these inactivation by plasma proteins [58]. Trans-[PtCl2(NH3)
cross-links are yet to be clarified. (pip–pip)] and trans-[PtCl2(n-butylamine)(pip–pip)] were
A distinguishing feature of the trans complexes with studied in BALB/c mice bearing A2780 ovarian carcinoma
planar aromatic bases is their ability to induce single- and mice with cisplatin-resistant counterpart A2780cisR. In
stranded protein-associated DNA strand breaks in a fashion spite of similar activity in vitro, trans-complexes extended
similar to topoisomerase inhibitors. This group of platinum life expectancy by 79 and 46 %, respectively, in contrast
complexes forms DNA-topoisomerase I complexes. Inter- to 160 % increase by cisplatin. Being the most promising
estingly, cis-counterparts barely produce DNA–protein compound with respect to the cytotoxicity in cisplatin-
cross-links and protein-associated strand breaks [58]. Fur- resistant cell line, trans-[PtCl2(NH3)(pip–pip)] showed no
thermore, complexes with planar bases decrease telomerase activity in BALB/c mice with A2780cisR tumor. Trans-
activity. Noteworthy, many telomerase inhibitors also pos- [PtCl2(n-butylamine)(pip–pip)] was less effective than cis-
sess planar aromatic moieties [58]. platin [58].
As in vivo activity is concerned, there is little data Platinum complexes with trans-geometry do indeed
available. Trans-[PtCl2(NH3)(tz)] was only moderately form structurally different DNA adducts, which are also

13
Cancer Chemother Pharmacol

processed in the other fashion than adducts of the conven- reported [66]. The DNA adducts of the dinuclear com-
tional platinum drugs. Trans-configured complexes show plexes partially unwind the double helix; however, in con-
promising activity in vitro, especially in platinum-resistant trast to cisplatin, only a small extent of DNA bending is
cells. Unfortunately, these findings do not transform into observed. These relatively little conformational changes
therapeutic efficacy in vivo. It is likely that altered bio- result in the lack of recognition by HMG proteins [67]. The
distribution of trans complexes compared to the approved cross-links of the azolato-bridged complexes on DNA are
platinum drugs, e.g., fast deactivation by plasma proteins, also expected to be poor substrates for the NER enzymes
is a key hurdle on the way to the clinical success. [65]. These dinuclear complexes exhibit much higher cyto-
toxicity than cisplatin. They have a different spectrum of
antitumor activity compared to the clinically approved
Polynuclear platinum complexes platinum drugs as found on a panel of 39 tumor cell lines
(JFCR39) [68]. The azolato-bridged complexes largely
As 1,2-d(GpG)-intrastrand cross-link induced by cisplatin overcome resistance in PC-9 and PC-14 cisplatin-resistant
causes DNA bending toward the major groove creating a non-small cell lung cancer cell lines with resistance fac-
kink susceptible to DNA damage recognition and repair tors ranging from 0.5 to 0.8 for PC-9 cells and from 0.9
proteins, the attention turned to polynuclear (also called to 2.0 for PC-14 cells [69]. A tetrazolato-bridged dinuclear
multinuclear) platinum complexes. They were expected to platinum complex 5-H-Y shows remarkably high antitumor
form DNA adducts, which would span over several base activity in pancreatic cancer [70]. However, as these com-
pairs and would induce less distortion on the DNA double plexes were not patented, they have no chance to be devel-
helix. One class of interesting anticancer polynuclear plati- oped for clinical use.
num compounds is represented by azolato-bridged dinu- A different class of polynuclear platinum complexes
clear platinum complexes (Fig. 7). was designed by Farrell and colleagues. They first focused
Similarly to cisplatin, these compounds preferentially on highly charged trans-configured complexes with ali-
form intrastrand cross-links on adjacent guanines [65]. phatic amines. The most prominent member of this class
For the triazolato-bridged member of this compound class is the trinuclear complex BBR3464 (Fig. 7). This com-
AMTA, additional electrostatic interaction with DNA was pound showed better uptake by tumor cells than cisplatin

H3N 2+ H3N 2+ H3N 2+


NH3 NH3 NH3
Pt Pt N Pt
N N N
OH OH OH
N N N
Pt N Pt N Pt
NH3 NH3 NH3
H3N H3N H3N

AMPZ AMTA 5-H-Y

4+
H2 H3N Cl
H2 H3N N
H3N N Pt
Pt
Pt N NH3
Cl N NH3 H2
NH3 H2

BBR3464

H2 8+
H2 H3N N
H2 H3N N NH3
H3N N Pt
Pt
H3N Pt N NH3
N N NH3 H2
H2 NH3 H2

Triplatin-NC

Fig. 7  Chemical structures of some polynuclear platinum complexes

13
Cancer Chemother Pharmacol

and overcame accumulation defects present in cisplatin- affinity for DNA. Such trinuclear platinum complex tri-
resistant cell lines [71]. As in the case of cisplatin, CTR1 is platin-NC (Fig. 7) showed higher accumulation in A2780
involved in cellular uptake of BBR3464. However, strong ovarian carcinoma cells than BBR3464, supposedly due to
interaction of BBR3464 with phospholipid membrane the increased charge. The complex was reported to enter
models due to the high positive charge may point out at an the cells via macropinocytosis facilitated by its interaction
alternative mechanism of the cellular entry [72]. BBR3464 with cell surface glycosaminoglycans, which was observed
was shown to platinate DNA to a higher extent than cis- to a lesser extent for BBR3464 and not at all for cispl-
platin [71]. The trinuclear complex forms long-range delo- atin and oxaliplatin [80]. Triplatin-NC appears to avoid
calized intra- and interstrand cross-links between guanines deactivation by intracellular nucleophiles [81]. It binds to
spanning up to six base pairs compared to a maximum of DNA in a non-covalent fashion: square planar tetraam(m)
three base pairs in the case of cisplatin [73]. As a result of ineplatinum moieties form bidentate N–O–N complexes
increased flexibility, DNA adducts induced by BBR3464 with phosphate oxygens through hydrogen bonding (phos-
lack severe distortion due to bending and twisting. While phate clamps) [82]. Triplatin-NC also induces DNA con-
both intra- and interstrand cross-links are not recognized by densation in nucleoli and transfer RNA aggregation lead-
HMG proteins, intrastrand lesions are effectively removed ing to DNA transcription inhibition at the concentrations
by NER in contrast to the interstrand cross-links [74, 75]. lower compared to the natural condensing agents like
The central tetraam(m)ineplatinum unit also plays an spermine [83]. The complex induces a robust G1 arrest in
important role as it interacts with DNA by pre-association a p53-independent manner [80]. These features account
[76]. The trinuclear complex provokes differential cellular for high cytotoxicity in various cancer cell lines, which is
response to p53 activation and recognition, which is likely barely affected by p53 or Bax mutations [83]. Therefore, it
to account for its activity in cells featuring p53 mutations is possible that triplatin-NC has overcome the limitations
and therefore resistant to cisplatin [77]. All these proper- of BBR3464 and may represent a promising antitumor
ties are reflected in the cytotoxicity of BBR3464. The com- drug candidate.
pound was found highly active in various cell lines (often
more than 100-fold compared to cisplatin), and it overcame
resistance in neuroblastoma, ovarian carcinoma and mela- Platinum(IV) prodrugs
noma cells with different mechanisms of resistance [78].
Testing BBR3464 on the NCI cell line panel revealed a Platinum(IV) prodrugs are a new class of molecules
markedly different spectrum of activity compared to the that might improve the pharmacological properties
clinically used drugs. The trinuclear complex was highly of the platinum(II) anticancer agents currently in use.
active in mice bearing GFX214 and MKN45 gastric car- Platinum(IV) complexes feature additional ligands in the
cinoma xenografts, which did not respond to cisplatin axial positions, which allows further chemical modifica-
therapy. Interestingly, BBR3464 prolonged tumor growth tion and adjusting properties. Platinum(IV) complexes
inhibition even after the end of the treatment. However, are substitution inert. As a result, they are more stable in
the maximum tolerated dose of BBR3464 was tenfold to blood circulation due to decreased interaction with plasma
15-fold lower than that of cisplatin [78]. In spite of these proteins. Platinum(IV) prodrugs are activated by reduction
encouraging results, in Phase II trials in non-small cell lung transforming them into square planar platinum(II) com-
cancer, small cell lung cancer, ovarian and gastric cancer plexes. Glutathione and ascorbate are believed to act as
only sporadic response was observed [76]. The maximum reducing agents, but substantial reduction by cellular pro-
tolerated dose was manifold lower than for cisplatin. The teins was also reported [84]. As the axial ligands are lost
patients suffered from severe dose-limiting gastrointesti- during activation step, they can be used to carry favorable
nal and hematological side effects [76], which are likely pharmacological properties in the prodrugs, for instance,
to have resulted from higher plasma protein binding in to maximize cellular uptake. In contrast, the nature and
human as compared to mice [79]. Apparently, progressive geometry of equatorial ligands determine cytotoxicity of
biotransformation and fast drug degradation facilitated by platinum(IV) compounds. Platinum(IV) complexes have
the trans-configuration of the chloride leaving groups are several advantages over their platinum(II) counterparts:
responsible for the disappointing results of the Phase II (1) due to their high stability, they can be administered
studies with BBR3464. orally; (2) they have diminished side effects as they are less
As the central platinum unit of BBR3464 interacts with prone to react with proteins; (3) their axial ligands can be
DNA electrostatically, it was hypothesized that complexes modified to improve the pharmacological properties, e.g.,
with several tetraam(m)ineplatinum units may repre- to target the platinum moiety to the tumor. Platinum(IV)
sent an inert alternative to BBR3464 in terms of reactiv- anticancer complexes can be classified into three catego-
ity. Moreover, a high positive charge would increase the ries: complexes with axial ligands without any bioactivity

13
Cancer Chemother Pharmacol

O
O

O
H2 Cl H2 HO H3N Cl O
N Cl N Cl Pt H3N Cl
N Cl Pt
Pt Pt
H2 O N Cl
N Cl H2 O
N Cl
H2 Cl H2 OH
O O
tetraplatin iproplatin satraplatin LA-12

Cl
O O Cl
O H O O
N O
O
O n O
H3N O H3N Cl
Cl O
Pt Pt
O O
H3N Cl H3N Cl
O O
N O O
n
H O
O O Cl
O O
Cl

Pt(IV)- estradiol conjugate Pt(IV) complex with ethacrynic acid

O
O

CHCl2 O OH OH OH
O
H3N Cl H3N N3 H3N N3 H3N N3
Cl NH3
Pt Pt Pt Pt Pt
H3N Cl H3N N3 N3 NH3 N3 N
Cl NH3
O O OH OH OH
CHCl2
O O

mitaplatin Pt(IV)-VPA photoactivatable Pt(IV) complexes

Fig. 8  Chemical structures of some platinum(IV) prodrugs

(hydroxides, chlorides, acetates, etc.), complexes with bio- platinum(II) [15]. In a Phase III trial in hormone-refracted
active or targeting axial ligands and photoactivatable com- prostate cancer, the complex led to an increase in progres-
plexes (Fig. 8). sion-free survival and reduced the risk of disease progres-
sion by 40 %. However, because of the lack of convincing
Platinum(IV) complexes without bioactive ligands benefit in terms of overall survival, approval by the US
Food and Drug Administration did not follow. It was attrib-
Tetraplatin, iproplatin, satraplatin (formerly JM-216) and uted to the insufficient power of the study and/or difficulty
LA-12 (Fig. 8) are platinum(IV) complexes without bio- to determine the actual reasons of death [15]. At present,
active axial ligands that entered clinical trials. Tetraplatin satraplatin is undergoing Phase I, II and III clinical trials
was abandoned due to severe cumulative neurotoxicity, in combination with various drugs for treatment of prostate
and iproplatin did not show any better activity than cispl- and non-small cell lung cancer as well as advanced solid
atin or carboplatin [15]. Satraplatin was the first lipophilic tumors. However, as these studies are in a much earlier
platinum(IV) drug developed for oral administration. It stage and will require further funding, it appears unlikely
is rapidly taken up by cells and overcomes accumulation that the company would keep on developing satraplatin.
defects in cisplatin-resistant cell lines [85]. In human, satra- LA-12, an analog of satraplatin, showed higher uptake
platin is readily absorbed and yields several products upon in A2780 and cisplatin-resistant A2780cisR cell lines
reduction in the bloodstream, the most abundant and active than cisplatin, although its accumulation was reduced in
species is thereby cis-amminedichlorido(cyclohexylamine) A2780cisR cells compared to the sensitive counterparts.

13
Cancer Chemother Pharmacol

The complex induced DNA lesions similar to those of cis- activation in tumor tissue by radiation. The complexes are
platin [85]. It overcame cisplatin resistance in A2780cisR inert in the dark and stable toward deactivation by glu-
cells caused by elevated GSH levels, as glutathione is tathione. Interestingly, trans isomers were more cytotoxic
responsible for activation of Pt(IV) complex through reduc- than their cis-counterparts. The use of pyridine ligands led
tion, and not for deactivation as in the case of Pt(II) com- to a marked increase in potency and activity at longer wave-
plexes [85]. LA-12 showed promising in vivo activity in lengths [90]. When irradiated with blue light, the activity of
mice bearing ADJ/PC6 plasmacytoma and A2780 ovarian trans, trans, trans-[Pt(N3)2(OH)2(NH3)(py)] (Fig. 8) toward
cancer [86] and has just finished Phase I clinical trials. esophageal cancer was enhanced in vivo [90]. The mecha-
nism of action is distinct from that of conventional plati-
Platinum(IV) complexes with bioactive ligands num drugs such as cisplatin. The photodecomposition route
involves two one-electron transfers from azido ligands
The axial sites of platinum(IV) prodrugs offer the opportu- generating N2 and Pt(II) species, which in turn form DNA
nity to increase the selectivity for cancerous tissue or intra- lesions. These lesions are able to stall RNA polymerase II
cellular targeting. As mentioned above, pre-treatment with more efficiently than cisplatin [90].
estrogen increased cisplatin cytotoxicity in hormone-positive Taken together, platinum(IV) prodrugs represent a
tumors [43]. Thus, estrogen receptor-positive [ER (+)] breast promising class of anticancer compounds; especially, the
and ovarian cancer cells were targeted by a Pt(IV) complex employment of the axial ligands targeting the drug to the
with estradiol tethered to the axial positions (Fig. 8). How- tumor has potential. In this case, it is important to ensure
ever, an extracellular mixture of cisplatin and estrogen was that the ligands stay attached to platinum all the way to
more cytotoxic than the Pt(IV) prodrug indicating its inef- the tumor but are cleaved off at the disease site. In gen-
ficient cellular uptake or different pharmacokinetics [43]. eral, many questions regarding cellular accumulation, bio-
Platinum(IV) complex with ethacrynic acid in axial transformation in the bloodstream and inside the cells and
position (Fig. 8) was developed to overcome glutathione- in vivo activity of platinum(IV) prodrugs remain unan-
S-transferase (GST) resistance. Ethacrynic acid is an inhib- swered. Better understanding of how these complexes
itor of cytosolic GST. This prodrug was rapidly taken up work may provide a key to the development of the first
and had higher efficacy than cisplatin after short incubation platinum(IV) compound in clinical use.
but only a moderate effect after 72 h [87]. A similar strat-
egy was employed by Lippard and colleagues who added
dichloroacetate ligands in axial positions of platinum(IV) Improved delivery
prodrugs (mitaplatin, Fig. 8). Dichloroacetate alters the
mitochondrial membrane potential in cancer cells. Sub- Although platinum drugs have brought major advances in
sequently, the cytochrome c is released and the apotosis- oncology, their clinical success is often hindered by the
inducing factor is translocated to nucleus. The cytotoxicity adverse side effects and development of resistance. Addi-
of mitaplatin is equivalent or exceeded that of other plati- tional obstacles include low bioavailability and low water
num compounds and comparable to that of cisplatin [88]. solubility. Carrier-based delivery of platinum drugs spe-
A Pt(IV) derivative of cisplatin-containing valproic acid cifically to the tumor is a strategy to overcome these lim-
(VPA) as axial ligands (Fig. 8) kills cancer cells more effi- itations because of its potential to improve drug efficacy,
ciently than cisplatin. VPA inhibits histone deacetylase reduce unwanted side effects and overcome defects in cel-
activity to decondense chromatin and thereby increases the lular accumulation accounting for drug resistance. Drug
accessibility of DNA within chromatin for DNA-binding delivery systems often exploit the differences between
agents. The treatment of ovarian cancer cells with Pt(IV)- healthy and tumor tissue in order to increase drug selectiv-
VPA complex resulted in enhanced histone H3 acetyla- ity for the malignant tissue. Passive drug delivery is based
tion and decondensation of heterochromatin compared to on the ability of macromolecules to accumulate better in
free VPA. Pt(IV)-valproic acid conjugate binds to DNA to tumor tissue because of its increased permeability and poor
a higher extent than the complexes with biologically inac- lymphatic clearance, a phenomenon known as increased
tive ligands. Together with increased cellular accumulation, permeability and retention (EPR) effect [91]. Drugs cou-
these factors seem to account for the enhanced cytotoxicity pled to liposomes, lipid particles, micelles and various
of Pt(IV)-VPA [89]. other polymeric carriers selectively accumulate in tumors,
while the clinically used cancer drugs have low molecular
Photoactivatable platinum(IV) prodrugs weight and easily pass through the membranes of cancer-
ous as well as normal tissue. Therefore, the nanoscale size
Sadler and co-workers prepared a series of Pt(IV) com- of the carrier is important, as it prevents extravasion in
plexes with azides (Fig. 8) to enable their selective normal tissue and removal by renal clearance. Active drug

13
Cancer Chemother Pharmacol

delivery achieves selectivity of a carrier-conjugated anti- of DNA in tumor cells [95]. The example of SPI-77 shows
cancer drug by using specific antibodies or ligands recog- that not only successful delivery but also efficient release
nizing receptors present exclusively in tumor cells. of the drug is important for the efficacy of a drug delivery
The most widely used motif in anticancer drug carri- system.
ers is the hydrophilic polymer polyethylene glycol (PEG). Aroplatin (L-NDDP, originally Aronex Pharmaceuti-
Due to its high aqueous solubility, high mobility and large cals, now Agenus, Inc.) is a liposomal formulation of cis-
exclusion volume, hydrated PEG forms a dense brush of bis-(neodecanoato)-{(1R,2R)-1,2-diaminocyclohexane}
polymer chains stretching out and covering the particle sur- platinum(II) (NDDP, Fig. 9), a structural analog of oxali-
face. Thus, PEG coating serves to decrease particle opsoni- platin with two branched aliphatic leaving groups of ten
zation and thus make the carrier less recognizable by the carbon atoms, incorporated in a matrix of dimyristoylphos-
reticuloendothelial system in the liver and spleen [92]. phatidylcholine and dimyristoyl phosphatidyl glycerol
(DMPG). Due to high lipophilicity of the oxaliplatin
Liposomal formulations of platinum drugs analog, the drug-to-lipid ratio is 1:15 [96]. The liposo-
mal carrier plays a crucial role in mediating the cytotox-
Liposomes prepared from various amphiphilic phospholip- icity and antitumor activity of the drug as the free drug
ids are lipid bilayer vesicles with an aqueous interior. The itself has a very low cytotoxicity. It has been suggested
major advantage of this technique is that it can be used for that active intermediates are formed within a lipid bilayer.
both hydrophobic and hydrophilic drugs utilizing the phos- The activation appears to rely on the presence of DMPG
pholipid bilayer and aqueous cavity, respectively. and lipophilic leaving groups of NDDP [92]. A Phase II
Lipoplatin (Regulon, Inc.) is one of the most promising study showed significant but manageable toxicity in mes-
liposomal platinum drugs currently under investigation. othelioma patients. In general, the response was promis-
This drug is prepared using soy phosphatidylcholine (SPC- ing; however, in some patients the areas of mesothelioma,
3), cholesterol, dipalmitoyl phosphatidylglycerol (DPPG) which were not in direct contact with pleural space, were
and methoxy-PEG-distearoyl phosphatidylethanolamine not exposed to the drug leading to poor efficacy [92].
(mPEG2000-DSPE). Lipoplatin comprises 9 % cisplatin Lipoxal (Regulon, Inc.) is another oxaliplatin formula-
and 91 % lipid (w/w) corresponding to drug-to-lipid ratio tion prepared in a manner similar to lipoplatin. A Phase I
of 1:10. Preclinical studies of lipoplatin in mice, rats and in study in advanced gastrointestinal cancer patients showed
severe combined immunodeficient mice revealed that it has adequate effectiveness and greatly reduced side effects
lower side effects, especially nephrotoxicity, compared to compared to oxaliplatin [97]. MBP-426 is an oxaliplatin
cisplatin [93]. A Phase I clinical trial showed elevated accu- formulation made of transferrin-conjugated N-glutaryl
mulation of cisplatin in tumor tissues compared to normal phosphatidylethanolamine. This drug binds preferentially
tissue and did not show cisplatin-specific side effects such to transferrin receptors expressed in tumor cells. Thus, the
as nephro-, neuro-, ototoxicity and hair loss. Phase II stud- drug delivery is enhanced via uptake of MBP-426 by trans-
ies of lipoplatin in combination with gemcitabine showed ferrin receptors in human cancer cells. This drug has shown
significant clinical benefit in patients who were previously encouraging anticancer activity in the preclinical setting
resistant to first- and second-line chemotherapy. Phase III and has entered clinical trials in gastric and esophageal
trials in NSCLC and pancreatic cancer are ongoing. Moreo- cancer [92].
ver, lipoplatin has obtained an orphan drug status from the Cisplatin nanocapsules contain the platinum complex
European Medicines Agency (EMA) for treatment of pan- with high encapsulation efficiency (the average drug-to-
creatic adenocarcinoma [93]. lipid ratio exceeds 10:1). They are taken up by caveolae-
SPI-77 (Alza Pharmaceuticals, formerly Sequus Phar- mediated endocytosis or by the clathrin-mediated route in
maceuticals) is another liposomal formulation of cisplatin. cells, which do not express caveolin-1. The cytotoxicity
In this case, cisplatin is encapsulated in stealth liposomes of cisplatin nanocapsules was twofold greater than that of
composed of hydrogenated soy phosphatidylcholine, cho- the free drug [98]. This formulation did not show enhanced
lesterol and PEG-modified phosphatidyl-ethanolamine. The antitumor efficacy in animal models of ovarian cancer,
drug loading capacity of SPI-77 is very low (drug-to-lipid which was attributed to insufficient accumulation of nano-
ratio ~1:70). Phase I studies of SPI-77 showed good toler- capsules in the tumor tissue and disruption of the capsule
ance in all patients with lack of toxicity typical for the cis- structure due to the interaction with plasma proteins [99].
platin regimen [94]. But the trial was discontinued due to Nevertheless, this encapsulation strategy is rather encour-
modest antitumor response. This could be due to low drug aging, and if the problems of plasma stability and poor
loading capacity and/or high stability of liposomes and delivery can be properly addressed, for example, through
inefficient release of the drug as was clear from low plasma active targeting, platinum drug nanocapsules may get a
concentration of free cisplatin and diminished platination clinical perspective.

13
Cancer Chemother Pharmacol

Fig. 9  Chemical structures of H2 O
NDDP (an active complex of N O
Aroplatin), AP5280, AP5346 Pt
(ProLindac), EFG-tethered O
N
cisplatin–nanotube conjugate H2 O
and Pt(IV) complex attached to
a SWNT nanotube NDDP

C C C C
H2 H2
O
O NH 95 HN 5
C C C C
H2 H2
O
O O NH x HN y
HO NH
O
O HO NH
HN
O
O HN
NH
O H2N
O O O N Pt
N
N H2
+
Na-O NH3 +
Na-O O
Pt
O NH3 O
O

AP5280 AP5346 (ProLindac)

H O
Cl NH3
N Pt = EFG
O NH3
O

EFG-tethered cisplatin-nanotube conjugate

OEt
OEt
H3N Cl
H3N Cl Pt
Pt
O H3N Cl
H3 N Cl
O
O
OH
O

Pt(IV) complex and its SWNT-conjugate


O
O
O O
= H O- H
O O O N
P N OCH2CH2
H 45
O O O

Platinum‑polymer conjugates clinical trials, which demonstrated promising efficacy and


less platinum-based toxicity [100]. However, the company
This concept is based on covalent binding of the drug to chose to proceed with a more promising conjugate AP5346
a hydrophilic polymer. The most frequently used polymer (Fig.  9), which represents an improved polymer carrier
for conjugation of anticancer compounds is the N-(2-hy- linked to a more potent oxaliplatin-like DACH-Pt moiety.
droxypropyl)methacrylamide (HPMA). AP5280 is one of AP5346 or ProLindac (Access Pharmaceuticals, Inc.)
the platinum-HPMA-copolymers (Fig. 9). It contains cispl- consists of a biocompatible and water-soluble polymer
atin linked to the polymer through a malonate end group. bound to DACH-Pt via a pH-sensitive chelating group.
This conjugate with platinum loading capacity of 10 % The chelate is stable at physiological pH but releases the
by weight was 20-fold less toxic than cisplatin in vivo but platinum moiety in more acidic environment of hypoxic
showed 19-fold increase in platinum accumulation in B16 tumors or endosomal/lysosomal vesicles inside the cells.
mouse tumors. Based on the improved therapeutic index ProLindac showed better tumor inhibition, reduced toxicity
in other mouse models, AP5280 advanced into Phase I/II toward normal cells, increased and more sustained plasma

13
Cancer Chemother Pharmacol

platinum levels and up to 16-fold increased platinum deliv- nanotubes are single-walled carbon nanotubes (SWCNTs)
ery to the tumor and 14-fold to tumor DNA. The conjugate or multi-walled carbon nanotubes (MWCNTs), peptide
works better than oxaliplatin in three colon xenograft mod- nanotubes and template-synthesized nanotubes. The pres-
els (Colo-26, HT-29 and HCT116), in the L1210 murine ence of open ends and a large inner volume facilitate incor-
leukemia and 0157 hybridoma models [101]. ProLindac poration of pharmaceutical species at high-loading capaci-
was well tolerated and showed no neutropenia or signifi- ties [92]. The possibility of incorporating and releasing
cant hematologic toxicity in patients with advanced solid cisplatin from SWCNTs was demonstrated. The released
tumors in Phase I trial [92]. Phase II study in recurrent cisplatin retained the ability to kill human lung cancer
ovarian cancer was initiated; however, in the last two years cells, while the SWCNTs themselves were not toxic [105].
there has been no update on the status of the trial, which The cytotoxicity of cis, cis, trans-[Pt(NH3)2Cl2(OEt)(OOC-
may indicate disappointing outcome. Interestingly, ProLin- CH2CH2COOH)], a platinum(IV) complex, was increased
dac is no longer listed as a drug under development by the by >100-fold after it was tethered to a surface of carbon
company [102]. nanotubes (Fig. 9). The conjugate was taken up by endo-
cytosis, and lower endosomal pH triggered the reduction
Platinum delivery using dendrimers and release of the active Pt(II) complex [106]. Targeted
cisplatin–nanotube conjugates modified with folate or epi-
Dendrimers are highly branched polymers with multiple dermal growth factor (EGF) (Fig. 9) demonstrated selective
end groups, which allow encapsulation or conjugation of accumulation and enhanced activity against folate recep-
various drug molecules in the core or at the surface. There tor-positive tumor cells or EGF-overexpressing head and
is a wide variety of dendrimers, which are made from poly- neck squamous carcinoma cells, respectively [107]. Ultra-
amidoamines, polyamines, polypeptides, polyesters, car- short carbon nanotubes (USCNTs) of ca. 1.4-nm-diameter
bohydrates or DNA. The most frequently studied ones are encapsulating cisplatin were more potent than the free
polyamidoamines (PAMAM). PAMAM dendrimers can platinum drug in two breast cancer cell lines (MCF7 and
carry either amino groups or carboxylate groups at the end MDA-MB-231). Wrapping of USCNTs with a surfactant
of their branches. Cisplatin conjugate with PAMAM den- hindered the release of cisplatin resulting in higher cyto-
drimers induced growth retardation of the subcutaneous toxicity. For in vivo application, the surfactant molecules
B16F10 murine melanoma, while cisplatin alone had no could be replaced with a cancer-specific protein [108].
antitumor activity [103]. Oxaliplatin conjugate with den-
drimers has also been reported. The PAMAM dendrimers Platinum complexes in polymer micelles
with carboxylic acid terminal group containing up to 40
DACH-Pt moieties at the surface retained water solubility Polymer micelles represent aggregates of block copoly-
and showed sustained release of active platinum species mers featuring core–shell architecture. They entrap drugs
over 24 h at physiological conditions. But further study did in the micelle core and increase the solubility of a drug.
not progress due to the modest efficiency of the conjugate Poly(amino acid)-based copolymers, such as poly(aspartic
[92]. A possible reason for this is irreversible binding of acid), PAsp and poly(glutamic acid), PGlu, have been used
platinum to easily accessible amino groups abundantly pre- most frequently for platinum drug delivery. Incorporation
sent in the dendrimer. Two dendrimers featuring a PEG unit of cisplatin in such polymer micelles decreased nephro-
as a core and citric acid on the periphery with molecular toxicity, increased the circulation of micelle-bound drug in
weight of ca. 1000 Da and ca. 2000 Da, respectively, were plasma and thereby increased exposure of the drug to the
synthesized. Cisplatin conjugate of the latter polymer dem- tumors. The size of cisplatin-containing micelles is about
onstrated greater cytotoxicity in both sensitive and resistant 30 nm, which allows them to penetrate into tumor tissues
HT1080 human fibrosarcoma cells, CT26 fibroblasts and including poorly permeable tumors. PGlu-based cisplatin
SKOV3 human ovarian cells compared to cisplatin, while micelles incorporating cisplatin showed prolonged blood
cisplatin conjugate of the former polymer demonstrated circulation and better accumulation in tumors. This for-
greater cytotoxicity toward HT1080 and CT26 cell lines. mulation, called NC-6004 or Nanoplatin, led to complete
Increased cytotoxicity of both conjugates over the parent tumor regression in some mice bearing C26 tumor, which
drug is encouraging for the possible use of dendrimer con- was not accompanied by a significant weight loss [92].
jugates as carriers for platinum drugs [104]. The cytotoxicity of NC-4006 was comparable to that of
free cisplatin in mice implanted with MKN-45 human gas-
Platinum drugs in nanotubes tric cancer cells [109]. A Phase I clinical trial of NC-6004
showed significantly better tolerability than free cisplatin
Nanotubes have tubular shape with one dimension in the with reduced side effects [110]. A Phase II study of the
nanometer scale, usually a diameter. Some examples of combination with gemcitabine in pancreatic cancer patients

13
Cancer Chemother Pharmacol

demonstrated that platinum hypersensitivity could be com- In addition, there is growing interest in combining plati-
pletely inhibited by prophylactic treatment and that there num drugs with PARP inhibitors targeting DNA repair sys-
was no need for pre-hydration as required for the standard tem. A combination of cisplatin with PARP inhibitors acted
cisplatin regimen. Oxaliplatin-containing micelles (NC- synergistically in several NSCLC cell lines independently
4016) were also prepared and could overcome oxaliplatin of their p53 status, whereas treatment with either drug alone
resistance in vivo because of drug release in the perinu- had no effect. Synergistic interaction was attributed to the
clear region targeting it directly to DNA. Clinical trials of induction of DNA damage foci, mitochondrial membrane
NC-4016 are currently under way [92]. permeabilization, caspase activation and plasma membrane
In summary, specific delivery of platinum drugs to rupture [23]. In Phase I studies, combination of olaparib
the tumor is an attractive way to improve their therapeu- with cisplatin or cisplatin/gemcitabine regimen was not
tic properties. A great diversity of carriers facilitates rapid very well tolerated, but promising activity in patients with
design of new formulations. Although most of constructs BRCA1/2 mutations warrants further investigation [115]. A
under clinical evaluation rely on EPR effect, active target- Phase II randomized study in NSCLC showed no benefit
ing is likely to become a major strategy in the future. In the from iniparib addition to the standard cisplatin/gemcitabine
era of targeted drugs, this may be the best opportunity to therapy. However, the results could have been influenced
bring a new platinum-based drug to the market. The avail- by imbalances in key baseline characteristics, such as per-
able nanotechnology provides a good basis for the develop- formance status, gender and disease stage [116].
ment in this direction.

Lessons to be learned and future directions


Novel combinations
When considering efforts put into understanding how plati-
Platinum drugs approved for clinical use are seldom given num drugs work and into development of new anticancer
to patients as monotherapy. They are usually combined platinum complexes, one may wonder why we have not yet
with other drugs in order to achieve a synergistic effect got another platinum drug approved for clinical use. A vast
through different mechanisms of action and thereby to number of new compounds have been prepared and tested
enable dose reduction for each drug. Good understanding in cell line models. Unfortunately, the latter only poorly
of how platinum complexes work and which mechanisms reflect the clinical situation. High cytotoxicity in tumor
are involved in platinum drug resistance paves the way for cell cultures should be in the first place viewed as a sign of
the development of new combination regimens. Here, we general toxicity, and not of anticancer activity. And indeed
summarize the most interesting recent developments in this some very promising drug candidates were abandoned due
field. to their toxic effects (Table 1). Parallel testing on healthy
As downregulation of copper transporter 1 is associated cells (e.g., human fibroblasts) could help to discriminate
with cisplatin resistance, copper-chelating agents (e.g., tri- between toxic and antitumor effects at the early stage. It is
entine), which stimulate expression of this transporter, have also important not to discard the compounds with moderate
been supposed to boost cisplatin uptake and thereby to sen- activity in vitro right in the beginning but to estimate the
sitize tumor cells to the drug. Despite encouraging results therapeutic window in the early animal studies. After all,
in cell line models, therapeutic response in patients was the widely and successfully used carboplatin is only mod-
low (19 %). It may be due to the heterogeneous cohort of erately cytotoxic. Regarding platinum drug delivery sys-
patients and tumor entities in the study resulting in differ- tems, which largely avoid the toxicity problems, not only
ent capacity of copper chelation to induce CTR1 [111]. successful delivery but also sufficient release of the drug at
Reports on synergistic interaction between platinum the tumor site is important (Table 1).
drugs and various inhibitors of EGFR signaling have drawn The differences between test animals and humans also
attention of clinicians to these combinations. However, so need to be taken into account. Besides different pharma-
far the results have been mostly disappointing. Combina- cokinetics, the perception of tolerability and efficacy in
tion of carboplatin/paclitaxel with sorafenib even increased animal experiments is distinct from that in the clinical set-
mortality in patients with squamous cell lung carcinoma in ting. The accepted level of toxicity is high (body weight
a Phase III trial [112]. Addition of gefitinib did not improve loss  ≤15 % and lethality ≤10 % [76]) and may result in
efficacy of the cisplatin/gemcitabine regimen in a Phase II severe intolerable side effects in patients. In animal stud-
study [113]. Nevertheless, compared to the standard pem- ies, inhibition of tumor growth is seen as a sufficient end-
etrexed/platinum treatment gefitinib was able to improve point, whereas therapeutic response in patients requires the
progression-free survival in NSCLC patients with EGFR reduction in tumor lesions. Clinical development requires
mutations [114]. improvement as well (Table 1). The setup of clinical trials

13
Cancer Chemother Pharmacol

Table 1  Overview of factors halting the development of platinum drug candidates


Drug The furthest development stage and tumor entity Reason(s) for failure to get approval

Picoplatin Phase III, SCLC Poor setup of clinical study (choice of the control group)
BBR3464 Phase II, metastatic SCLC, Toxicity due to fast biotransformation, low maximum tolerated dose
metastatic pancreatic cancer resulting in only sporadic response
Tetraplatin Phase I, refractory solid tumors Toxicity, likely due to inactivation
Iproplatin Phase II, ovarian cancer, metastatic breast cancer No improved activity
Satraplatin Phase III, prostate cancer Lack of benefit in overall survival, insufficient
power of the study, wrong choice of tumor entity
SPI-77 Phase II, platinum-sensitive recurrent ovarian cancer Lack of efficacy due to low drug loading capacity
and insufficient drug release

should be carefully planned to avoid influence of external have been developed. At present, it is, however, inconclu-
factors such as post-study chemotherapy within a control sive, which estimation formula is better [117].
group (as in the case of picoplatin). Pharmaceutical com- Understanding the factors leading to the lack of clinical
panies sometimes tend to aim at wider indication than the success holds a key to the rational design of new drugs. In
drug candidate actually offers. On the contrary, clinical the times of targeted drugs and personalized medicine, drug
studies would benefit from the focus on the advantages of development relies on distinguishing between healthy and
novel drugs. Satraplatin was developed as an oral plati- tumor tissue and between patients. Therefore, a new plati-
num drug, which after reduction yields active platinum(II) num drug featuring only a different activity profile would
species sharing mechanistic principles with cisplatin and not have the potential to attract the sponsors. Consequently,
carboplatin. It might have been advantageous to evalu- compounds and formulations stable in the bloodstream and
ate satraplatin in the salvage setting in platinum-sensitive in healthy tissues releasing active species in tumor environ-
tumors and not in refractory prostate cancer. Phase II tri- ment (e.g., carrier-based formulations or Pt(IV) prodrugs)
als in ovarian cancer suggested at least similar activity of appear the most promising. In the future, they will likely
satraplatin and approved platinum drugs. Even in the case utilize active targeting strategies, e.g., through binding to
of comparable efficacy an oral platinum drug could be a cancer-specific receptors. It should be noted, however,
rational choice in the salvage therapy. that the issues of platinum drug attachment and controlled
In the clinical trials, it is also of importance to choose the release still pose a challenge, since delivery vehicles are
appropriate dosing. For instance, in the case of carboplatin often peptides rich in nitrogen and sulfur donors with high
the dose is calculated based on the target AUC (area under affinity for platinum.
curve) of a plot of drug concentration versus time and based Development of platinum drug conjugates with biologi-
on the renal function represented by the glomerular filtra- cally active molecules sometimes showed that the conju-
tion rate (GFR), the so-called Calvert formula. This dosing gates were less effective than a simple drug combination.
strategy helps to compensate for the interpatient variability. The advantage of a combination is that active substances
It relies on the specific pharmacokinetics of carboplatin, as are not bound to each other and can exert their effect inde-
nearly three quarters of the drug is excreted in the urine in pendently. Combination of platinum drugs with copper-
the intact form. Such pharmacology-based dosing could be chelating agents is worth further investigation because
considered for new inert platinum complexes, since their it could become a low-cost treatment option and because
plasma protein binding is expected to be low resulting in resistant cells with low CTR1 levels could be more sus-
predominantly renal clearance. Nevertheless, the Calvert ceptible to the induction of the transporter. This strategy
formula has its drawbacks. It does not cover specific patient would get more promising if CTR1 expression levels could
groups such as older and overweight patients, as well as be determined influencing the decision to use a copper che-
children. For children, for example, a modification of the lator or not. Combing platinum complexes with inhibitors
equation by Newell has been introduced. Another prob- of EGFR signaling may be considered for the treatment
lem is measurement or estimation of the GFR or creatinine of patients with EGFR mutations. Platinum combinations
clearance, which substitutes GFR in some new algorithms. with PARP inhibitors, e.g., olaparib, show promise for the
Measurements are either inconvenient and expensive (radi- group of patients featuring BRCA1/2 mutations. In this
oisotopic methods) or inaccurate (24-h urine collection). case, sequential treatment may be beneficial as olaparib
For this reason, several new methods of GFR estimation response correlated with prior platinum sensitivity [116].

13
Cancer Chemother Pharmacol

The example of combinations clearly demonstrates and ATP7B associated with cisplatin resistance in human ovar-
interpatient variability and the necessity to identify predic- ian carcinoma cells. BMC Cancer 8:175
11. Samimi G, Safaei R, Katano K, Holzer AK, Rochdi M, Tomioka
tive factors for therapeutic response and toxicity. Predictors M, Goodman M, Howell SB (2004) Increased expression of the
of severe ototoxicity and of cisplatin efficacy in head and copper efflux transporter ATP7A mediates resistance to cispl-
neck cancer have been reported, but many more are yet to atin, carboplatin, and oxaliplatin in ovarian cancer cells. Clin
be revealed [102]. Rapid development of functional genom- Cancer Res 10:4661–4669
12. Nakayama K, Kanzaki A, Terada K, Mutoh M, Ogawa K,
ics and imaging technology opens possibilities to personal- Sugiyama T, Takenoshita S, Itoh K, Yaegashi N, Miyazaki K,
ize platinum-based treatment of cancer. The application of Neamati N, Takebayashi Y (2004) Prognostic value of the Cu-
these techniques is likely to influence platinum chemother- transporting ATPase in ovarian carcinoma patients receiving
apy to a similar extent as the development of new drugs or cisplatin-based chemotherapy. Clin Cancer Res 10:2804–2811
13. Schneider V, Krieger ML, Bendas G, Jaehde U, Kalayda GV
drug combinations. (2013) Contribution of intracellular ATP to cisplatin resistance
Platinum drugs represent a cornerstone of treatment of tumor cells. J Biol Inorg Chem 18:165–174
of various solid neoplasms. Although the interest in new 14. Planells-Cases R, Lutter D, Guyader C, Gerhards NM, Ullrich
platinum compounds has been fading in the era of targeted F, Elger DA, Kucukosmanoglu A, Xu G, Voss FK, Reincke SM,
Stauber T, Blomen VA, Vis DJ, Wessels LF, Brummelkamp
drugs, the potential to improve on platinum-based treat- TR, Borst P, Rottenberg S, Jentsch TJ (2015) Subunit com-
ment has not yet been fully exploited. New drugs and for- position of VRAC channels determines substrate specificity
mulations actively targeted to the tumors and successfully and cellular resistance to Pt-based anti-cancer drugs. EMBO J
released there, but also new platinum-based drug combi- 34:2993–3008
15. Wheate NJ, Walker S, Craig GE, Oun R (2010) The status of
nations and identification of predictive markers for patient platinum anticancer drugs in the clinic and in clinical trials.
response and side effects are likely to shape future develop- Dalton Trans 39:8113–8127
ments in the field. 16. Borst P, Evers R, Kool M, Wijnholds J (2000) A family of drug
transporters: the multidrug resistance-associated proteins. J Natl
Acknowledgments  The authors are grateful to Ulrich Jaehde (Insti- Cancer Inst 92:1295–1302
tute of Pharmacy, University of Bonn) for fruitful discussions. 17. Yamasaki M, Makino T, Masuzawa T, Kurokawa Y, Miyata H,
Takiguchi S, Nakajima K, Fujiwara Y, Matsuura N, Mori M,
Doki Y (2011) Role of multidrug resistance protein 2 (MRP2)
in chemoresistance and clinical outcome in oesophageal squa-
mous cell carcinoma. Br J Cancer 104:707–713
References 18. Takahara PM, Rosenzweig AC, Frederick CA, Lippard SJ
(1995) Crystal structure of double-stranded DNA contain-
1. Wiltshaw E (1979) Cisplatin in the treatment of cancer. Platin ing the major adduct of the anticancer drug cisplatin. Nature
Met Rev 23:90–98 377:649–652
2. Galanski M (2006) Recent developments in the field of antican- 19. Brown SJ, Kellett PJ, Lippard SJ (1993) Ixr1, a yeast protein
cer platinum complexes. Recent Pat Anticancer Drug Discov that binds to platinated DNA and confers sensitivity to cisplatin.
1:285–295 Science 261:603–605
3. Lebwohl D, Canetta R (1998) Clinical development of platinum 20. Huang JC, Zamble DB, Reardon JT, Lippard SJ, Sancar
complexes in cancer therapy: an historical perspective and an A (1994) HMG-domain proteins specifically inhibit the
update. Eur J Cancer 34:1522–1534 repair of the major DNA adduct of the anticancer drug cispl-
4. Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, atin by human excision nuclease. Proc Natl Acad Sci USA
Castedo M, Kroemer G (2012) Molecular mechanisms of cispl- 91:10394–10398
atin resistance. Oncogene 31:1869–1883 21. Furuta T, Ueda T, Aune G, Sarasin A, Kraemer KH, Pommier
5. Kalayda GV, Wagner CH, Jaehde U (2012) Relevance of cop- Y (2002) Transcription-coupled nucleotide excision repair as a
per transporter 1 for cisplatin resistance in human ovarian carci- determinant of cisplatin sensitivity of human cells. Cancer Res
noma cells. J Inorg Biochem 116:1–10 62:4899–4902
6. Safaei R (2006) Role of copper transporters in the uptake and 22. Vaisman A, Varchenko M, Umar A, Kunkel TA, Risinger
efflux of platinum containing drugs. Cancer Lett 234:34–39 JI, Barrett JC, Hamilton TC, Chaney SG (1998) The role of
7. Holzer AK, Manorek GH, Howell SB (2006) Contribution of hMLH1, hMSH3, and hMSH6 defects in cisplatin and oxalipl-
the major copper influx transporter CTR1 to the cellular accu- atin resistance: correlation with replicative bypass of platinum-
mulation of cisplatin, carboplatin, and oxaliplatin. Mol Pharma- DNA adducts. Cancer Res 58:3579–3585
col 70:1390–1394 23. Michels J, Vitale I, Senovilla L, Enot DP, Garcia P, Lissa D,
8. Zisowsky J, Koegel S, Leyers S, Devarakonda K, Kassack Olaussen KA, Brenner C, Soria J, Castedo M, Kroemer G
MU, Osmak M, Jaehde U (2007) Relevance of drug uptake and (2013) Synergistic interaction between cisplatin and PARP
efflux for cisplatin sensitivity of tumor cells. Biochem Pharma- inhibitors in non-small cell lung cancer. Cell Cycle 12:877–883
col 73:298–307 24. Kirsch DG, Kastan MB (1998) Tumor-suppressor p53: impli-
9. Yang T, Chen M, Chen T, Thakur A (2015) Expression of cations for tumor development and prognosis. J Clin Oncol
the copper transporters hCtr1, ATP7A and ATP7B is associ- 16:3158–3168
ated with the response to chemotherapy and survival time in 25. Feldman DR, Bosl GJ, Sheinfeld J, Motzer RJ (2008) Medical
patients with resected non-small cell lung cancer. Oncol Lett treatment of advanced testicular cancer. JAMA 299:672–684
10:2584–2590 26. Peng HQ, Hogg D, Malkin D, Bailey D, Gallie BL, Bulbul M,
10. Kalayda GV, Wagner CH, Buss I, Reedijk J, Jaehde U (2008) Jewett M, Buchanan J, Goss PE (1993) Mutations of the p53
Altered localisation of the copper efflux transporters ATP7A gene do not occur in testis cancer. Cancer Res 53:3574–3578

13
Cancer Chemother Pharmacol

27. Calvert AH, Harland SJ, Newell DR, Siddik ZH, Jones AC, Mendiboure J, Pignon J, Jooste V, van Endert P, Ducreux M,
McElwain TJ, Raju S, Wiltshaw E, Smith IE, Baker JM, Peck- Zitvogel L, Piard F, Kroemer G (2010) Immunogenic death
ham MJ, Harrap KR (1982) Early clinical studies with cis- of colon cancer cells treated with oxaliplatin. Oncogene
diammine-1,1-cyclobutane dicarboxylate platinum II. Cancer 29:482–491
Chemother Pharmacol 9:140–147 43. Barnes KR, Kutikov A, Lippard SJ (2004) Synthesis, char-
28. Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, acterization, and cytotoxicity of a series of estrogen-tethered
Burger RA, Mannel RS, De Geest K, Hartenbach EM, Baergen platinum(IV) complexes. Chem Biol 11:557–564
R (2003) Phase III trial of carboplatin and paclitaxel compared 44. Saha P, Descôteaux C, Brasseur K, Fortin S, Leblanc V, Parent
with cisplatin and paclitaxel in patients with optimally resected S, Asselin E, Bérubé G (2012) Synthesis, antiproliferative activ-
stage III ovarian cancer: a Gynecologic Oncology Group study. ity and estrogen receptor α affinity of novel estradiol-linked
J Clin Oncol 21:3194–3200 platinum(II) complex analogs to carboplatin and oxaliplatin.
29. Stewart DJ (2007) Mechanisms of resistance to cisplatin and Potential vector complexes to target estrogen-dependent tissues.
carboplatin. Crit Rev Oncol Hematol 63:12–31 Eur J Med Chem 48:385–390
30. Kuwahara A, Yamamori M, Nishiguchi K, Okuno T, Chaya- 45. Brasseur K, Leblanc V, Fabi F, Parent S, Descôteaux C, Bérubé
hara N, Miki I, Tamura T, Kadoyama K, Inokuma T, Takemoto G, Asselin E (2013) ERα-targeted therapy in ovarian cancer
Y, Nakamura T, Kataoka K, Sakaeda T (2010) Effect of dose- cells by a novel estradiol-platinum(II) hybrid. Endocrinology
escalation of 5-fluorouracil on circadian variability of its phar- 154:2281–2295
macokinetics in Japanese patients with Stage III/IVa esophageal 46. van Themsche C, Parent S, Leblanc V, Descôteaux C, Simard
squamous cell carcinoma. Int J Med Sci 7:48–54 A, Bérubé G, Asselin E (2009) VP-128, a novel oestradiol-
31. Zhang S, Lovejoy KS, Shima JE, Lagpacan LL, Shu Y, Lapuk platinum(II) hybrid with selective anti-tumour activity towards
A, Chen Y, Komori T, Gray JW, Chen X, Lippard SJ, Giaco- hormone-dependent breast cancer cells in vivo. Endocr Relat
mini KM (2006) Organic cation transporters are determinants of Cancer 16:1185–1195
oxaliplatin cytotoxicity. Cancer Res 66:8847–8857 47. Li H, Gao X, Liu R, Wang Y, Zhang M, Fu Z, Mi Y, Wang Y,
32. Tashiro A, Tatsumi S, Takeda R, Naka A, Matsuoka H, Hashi- Yao Z, Gao Q (2015) Glucose conjugated platinum(II) com-
moto Y, Hatta K, Maeda K, Kamoshida S (2014) High expres- plex: antitumor superiority to oxaliplatin, combination effect
sion of organic anion transporter 2 and organic cation trans- and mechanism of action. Eur J Med Chem 101:400–408
porter 2 is an independent predictor of good outcomes in 48. Bowler BE, Lippard SJ (1986) Modulation of platinum antitu-
patients with metastatic colorectal cancer treated with FOL- mor drug binding to DNA by linked and free intercalators. Bio-
FOX-based chemotherapy. Am J Cancer Res 4:528–536 chemistry 25:3031–3038
33. Martinez-Balibrea E, Martínez-Cardús A, Ginés A, Ruiz de Por- 49. Temple MD, McFadyen WD, Holmes RJ, Denny WA, Mur-
ras V, Moutinho C, Layos L, Manzano JL, Bugés C, Bystrup ray V (2000) Interaction of cisplatin and DNA-targeted 9-ami-
S, Esteller M, Abad A (2015) Tumor-related molecular mecha- noacridine platinum complexes with DNA. Biochemistry
nisms of oxaliplatin resistance. Mol Cancer Ther 14:1767–1776 39:5593–5599
34. Di Francesco AM, Ruggiero A, Riccardi R (2002) Cellular and 50. Holmes RJ, McKeage MJ, Murray V, Denny WA, McFadyen
molecular aspects of drugs of the future: oxaliplatin. Cell Mol WD (2001) cis-Dichloroplatinum(II) complexes tethered to
Life Sci 59:1914–1927 9-aminoacridine-4-carboxamides: synthesis and action in resist-
35. Holford J, Raynaud F, Murrer BA, Grimaldi K, Hartley JA, ant cell lines in vitro. J Inorg Biochem 85:209–217
Abrams M, Kelland LR (1998) Chemical, biochemical and 51. Suryadi J, Bierbach U (2012) DNA metalating-intercalating
pharmacological activity of the novel sterically hindered plati- hybrid agents for the treatment of chemoresistant cancers.
num co-ordination complex, cis-[amminedichloro(2-methylpyr- Chemistry 18:12926–12934
idine)] platinum(II) (AMD473). Anticancer Drug Des 13:1–18 52. Kostrhunova H, Malina J, Pickard AJ, Stepankova J, Vojtiskova
36. Kelland L (2007) The resurgence of platinum-based cancer M, Kasparkova J, Muchova T, Rohlfing ML, Bierbach U, Bra-
chemotherapy. Nat Rev Cancer 7:573–584 bec V (2011) Replacement of a thiourea with an amidine group
37. Treat J, Schiller J, Quoix E, Mauer A, Edelman M, Modiano M, in a monofunctional platinum-acridine antitumor agent. Effect
Bonomi P, Ramlau R, Lemarie E (2002) ZD0473 treatment in on DNA interactions, DNA adduct recognition and repair. Mol
lung cancer: an overview of the clinical trial results. Eur J Can- Pharm 8:1941–1954
cer 38(Suppl 8):S13–S18 53. Smyre CL, Saluta G, Kute TE, Kucera GL, Bierbach U (2011)
38. Chan BA, Coward Jermaine I G (2013) Chemotherapy advances Inhibition of DNA synthesis by a platinum-acridine hybrid
in small-cell lung cancer. J Thorac Dis 5(Suppl 5):S565–S578 agent leads to potent cell kill in non-small cell lung cancer. ACS
39. Buss I, Garmann D, Galanski M, Weber G, Kalayda GV, Kep- Med Chem Lett 2:870–874
pler BK, Jaehde U (2011) Enhancing lipophilicity as a strategy 54. Johnstone TC, Wilson JJ, Lippard SJ (2013) Monofunctional
to overcome resistance against platinum complexes? J Inorg and higher-valent platinum anticancer agents. Inorg Chem
Biochem 105:709–717 52:12234–12249
40. Abramkin SA, Jungwirth U, Valiahdi SM, Dworak C, Habala 55. Lovejoy KS, Todd RC, Zhang S, McCormick MS, D’Aquino
L, Meelich K, Berger W, Jakupec MA, Hartinger CG, Nazarov JA, Reardon JT, Sancar A, Giacomini KM, Lippard SJ (2008)
AA, Galanski M, Keppler BK (2010) {(1R,2R,4R)-4-methyl- cis-Diammine(pyridine)chloroplatinum(II), a monofunctional
1,2-cyclohexanediamine}oxalatoplatinum(II): a novel enanti- platinum(II) antitumor agent: uptake, structure, function, and
omerically pure oxaliplatin derivative showing improved anti- prospects. Proc Natl Acad Sci USA 105:8902–8907
cancer activity in vivo. J Med Chem 53:7356–7364 56. Johnstone TC, Park GY, Lippard SJ (2014) Understanding and
41. Jungwirth U, Xanthos DN, Gojo J, Bytzek AK, Körner W, Hef- improving platinum anticancer drugs–phenanthriplatin. Anti-
feter P, Abramkin SA, Jakupec MA, Hartinger CG, Windberger cancer Res 34:471–476
U, Galanski M, Keppler BK, Berger W (2012) Anticancer activ- 57. Lovejoy KS, Serova M, Bieche I, Emami S, D’Incalci M, Brog-
ity of methyl-substituted oxaliplatin analogs. Mol Pharmacol gini M, Erba E, Gespach C, Cvitkovic E, Faivre S, Raymond E,
81:719–728 Lippard SJ (2011) Spectrum of cellular responses to pyriplatin,
42. Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghir- a monofunctional cationic antineoplastic platinum(II) com-
inghelli F, Aymeric L, Michaud M, Apetoh L, Barault L, pound, in human cancer cells. Mol Cancer Ther 10:1709–1719

13
Cancer Chemother Pharmacol

58. Aris SM, Farrell NP (2009) Towards antitumor active trans- 73. Summa N, Maigut J, Puchta R, van Eldik R (2007) Possible
platinum compounds. Eur J Inorg Chem 2009:1293 biotransformation reactions of polynuclear Pt(II) complexes.
59. Heringova P, Woods J, Mackay FS, Kasparkova J, Sadler PJ, Inorg Chem 46:2094–2104
Brabec V (2006) Transplatin is cytotoxic when photoacti- 74. Kasparkova J, Zehnulova J, Farrell N, Brabec V (2002) DNA
vated: enhanced formation of DNA cross-links. J Med Chem interstrand cross-links of the novel antitumor trinuclear plati-
49:7792–7798 num complex BBR3464. Conformation, recognition by high
60. Coluccia M, Boccarelli A, Mariggiò MA, Cardellicchio N, mobility group domain proteins, and nucleotide excision repair.
Caputo P, Intini FP, Natile G (1995) Platinum(II) complexes J Biol Chem 277:48076–48086
containing iminoethers: a trans platinum antitumour agent. 75. Zehnulova J, Kasparkova J, Farrell N, Brabec V (2001) Con-
Chem Biol Interact 98:251–266 formation, recognition by high mobility group domain proteins,
61. Ma ES, Bates WD, Edmunds A, Kelland LR, Fojo T, Far- and nucleotide excision repair of DNA intrastrand cross-links of
rell N (2005) Enhancement of aqueous solubility and stability novel antitumor trinuclear platinum complex BBR3464. J Biol
employing a trans acetate axis in trans planar amine platinum Chem 276:22191–22199
compounds while maintaining the biological profile. J Med 76. Jakupec MA, Galanski M, Arion VB, Hartinger CG, Keppler
Chem 48:5651–5654 BK (2008) Antitumour metal compounds: more than theme and
62. Montero EI, Díaz S, González-Vadillo AM, Pérez JM, Alonso variations. Dalton Trans (2):183–194
C, Navarro-Ranninger C (1999) Preparation and characteriza- 77. Kasparkova J, Fojta M, Farrell N, Brabec V (2004) Differential
tion of novel trans-[PtCl(2)(amine)(isopropylamine)] com- recognition by the tumor suppressor protein p53 of DNA modi-
pounds: cytotoxic activity and apoptosis induction in ras-trans- fied by the novel antitumor trinuclear platinum drug BBR3464
formed cells. J Med Chem 42:4264–4268 and cisplatin. Nucleic Acids Res 32:5546–5552
63. Quiroga AG, Pérez JM, Alonso C, Navarro-Ranninger C, Far- 78. Manzotti C, Pratesi G, Menta E, Di Domenico R, Cavalletti E,
rell N (2006) Novel transplatinum(II) complexes with [N2O2] Fiebig HH, Kelland LR, Farrell N, Polizzi D, Supino R, Pezzoni
donor sets. Cellular pharmacology and apoptosis induction in G, Zunino F (2000) BBR 3464: a novel triplatinum complex,
Pam 212-ras cells. J Med Chem 49:224–231 exhibiting a preclinical profile of antitumor efficacy different
64. Novakova O, Kasparkova J, Malina J, Natile G, Brabec V (2003) from cisplatin. Clin Cancer Res 6:2626–2634
DNA-protein cross-linking by trans-[PtCl(2)(E-iminoether)(2)]. 79. Jodrell DI, Evans TR, Steward W, Cameron D, Prendiville
A concept for activation of the trans geometry in platinum anti- J, Aschele C, Noberasco C, Lind M, Carmichael J, Dobbs N,
tumor complexes. Nucleic Acids Res 31:6450–6460 Camboni G, Gatti B, de Braud F (2004) Phase II studies of
65. Mlcouskova J, Kasparkova J, Suchankova T, Komeda S, Brabec BBR3464, a novel tri-nuclear platinum complex, in patients
V (2012) DNA conformation and repair of polymeric natural with gastric or gastro-oesophageal adenocarcinoma. Eur J Can-
DNA damaged by antitumor azolato-bridged dinuclear Pt(II) cer 40:1872–1877
complex. J Inorg Biochem 114:15–23 80. Peterson EJ, Menon VR, Gatti L, Kipping R, Dewasinghe D,
66. Kida N, Katsuda Y, Yoshikawa Y, Komeda S, Sato T, Saito Y, Perego P, Povirk LF, Farrell NP (2015) Nucleolar targeting by
Chikuma M, Suzuki M, Imanaka T, Yoshikawa K (2010) Char- platinum: p53-independent apoptosis follows rRNA inhibition,
acteristic effect of an anticancer dinuclear platinum(II) com- cell-cycle arrest, and DNA compaction. Mol Pharm 12:287–297
plex on the higher-order structure of DNA. J Biol Inorg Chem 81. Benedetti BT, Peterson EJ, Kabolizadeh P, Martínez A, Kipping
15:701–707 R, Farrell NP (2011) Effects of noncovalent platinum drug–pro-
67. Mlcouskova J, Malina J, Novohradsky V, Kasparkova J, Kom- tein interactions on drug efficacy: use of fluorescent conjugates
eda S, Brabec V (2012) Energetics, conformation, and recogni- as probes for drug metabolism. Mol Pharm 8:940–948
tion of DNA duplexes containing a major adduct of an antican- 82. Prisecaru A, Molphy Z, Kipping RG, Peterson EJ, Qu Y, Kel-
cer azolato-bridged dinuclear Pt(II) complex. Biochim Biophys lett A, Farrell NP (2014) The phosphate clamp: sequence selec-
Acta 1820:1502–1511 tive nucleic acid binding profiles and conformational induction
68. Komeda S, Takayama H, Suzuki T, Odani A, Yamori T, Chi- of endonuclease inhibition by cationic Triplatin complexes.
kuma M (2013) Synthesis of antitumor azolato-bridged dinu- Nucleic Acids Res 42:13474–13487
clear platinum(II) complexes with in vivo antitumor effi- 83. Malina J, Farrell NP, Brabec V (2014) Substitution-inert tri-
cacy and unique in vitro cytotoxicity profiles. Metallomics nuclear platinum complexes efficiently condense/aggregate
5:461–468 nucleic acids and inhibit enzymatic activity. Angew Chem Int
69. Uemura M, Suzuki T, Nishio K, Chikuma M, Komeda S (2012) Ed Engl 53:12812–12816
An in vivo highly antitumor-active tetrazolato-bridged dinuclear 84. Wexselblatt E, Gibson D (2012) What do we know about the
platinum(II) complex largely circumvents in vitro cisplatin reduction of Pt(IV) pro-drugs? J Inorg Biochem 117:220–229
resistance: two linkage isomers yield the same product upon 85. Hall MD, Mellor HR, Callaghan R, Hambley TW (2007) Basis
reaction with 9-ethylguanine but exhibit different cytotoxic pro- for design and development of platinum(IV) anticancer com-
files. Metallomics 4:686–692 plexes. J Med Chem 50:3403–3411
70. Komeda S, Lin Y, Chikuma M (2011) A tetrazolato-bridged 86. Kozubík A, Horváth V, Svihálková-Sindlerová L, Soucek K,
dinuclear platinum(II) complex exhibits markedly high in vivo Hofmanová J, Sova P, Kroutil A, Zák F, Mistr A, Turánek J
antitumor activity against pancreatic cancer. ChemMedChem (2005) High effectiveness of platinum(IV) complex with ada-
6:987–990 mantylamine in overcoming resistance to cisplatin and sup-
71. Perego P, Gatti L, Caserini C, Supino R, Colangelo D, Leone R, pressing proliferation of ovarian cancer cells in vitro. Biochem
Spinelli S, Farrell N, Zunino F (1999) The cellular basis of the Pharmacol 69:373–383
efficacy of the trinuclear platinum complex BBR 3464 against 87. Ang WH, Khalaila I, Allardyce CS, Juillerat-Jeanneret L,
cisplatin-resistant cells. J Inorg Biochem 77:59–64 Dyson PJ (2005) Rational design of platinum(IV) compounds
72. Kabolizadeh P, Ryan J, Farrell N (2007) Differences in the to overcome glutathione-S-transferase mediated drug resistance.
cellular response and signaling pathways of cisplatin and J Am Chem Soc 127:1382–1383
BBR3464 ([[trans-PtCl(NH3)(2)]2mu-(trans-Pt(NH3)(2) 88. Dhar S, Lippard SJ (2009) Mitaplatin, a potent fusion of cis-
(H2N(CH2)(6)-NH2)2)]4+) influenced by copper homeostasis. platin and the orphan drug dichloroacetate. Proc Natl Acad Sci
Biochem Pharmacol 73:1270–1279 USA 106:22199–22204

13
Cancer Chemother Pharmacol

89. Novohradsky V, Zerzankova L, Stepankova J, Vrana O, Raveen- 105. Ajima K, Yudasaka M, Murakami T, Maigné A, Shiba K, Iijima
dran R, Gibson D, Kasparkova J, Brabec V (2015) New insights S (2005) Carbon nanohorns as anticancer drug carriers. Mol
into the molecular and epigenetic effects of antitumor Pt(IV)- Pharm 2:475–480
valproic acid conjugates in human ovarian cancer cells. Bio- 106. Feazell RP, Nakayama-Ratchford N, Dai H, Lippard SJ (2007)
chem Pharmacol 95:133–144 Soluble single-walled carbon nanotubes as longboat delivery
90. Butler JS, Sadler PJ (2013) Targeted delivery of platinum-based systems for platinum(IV) anticancer drug design. J Am Chem
anticancer complexes. Curr Opin Chem Biol 17:175–188 Soc 129:8438–8439
91. Maeda H, Bharate GY, Daruwalla J (2009) Polymeric drugs for 107. Bhirde AA, Patel V, Gavard J, Zhang G, Sousa AA, Mase-
efficient tumor-targeted drug delivery based on EPR-effect. Eur dunskas A, Leapman RD, Weigert R, Gutkind JS, Rusling JF
J Pharm Biopharm 71:409–419 (2009) Targeted killing of cancer cells in vivo and in vitro with
92. Oberoi HS, Nukolova NV, Kabanov AV, Bronich TK (2013) EGF-directed carbon nanotube-based drug delivery. ACS Nano
Nanocarriers for delivery of platinum anticancer drugs. Adv 3:307–316
Drug Deliv Rev 65:1667–1685 108. Guven A, Rusakova IA, Lewis MT, Wilson LJ (2012) Cispl-
93. Boulikas T (2009) Clinical overview on Lipoplatin: a successful atin@ US-tube carbon nanocapsules for enhanced chemothera-
liposomal formulation of cisplatin. Expert Opin Investig Drugs peutic delivery. Biomaterials 33:1455–1461
18:1197–1218 109. Matsumura Y, Kataoka K (2009) Preclinical and clinical studies
94. Veal GJ, Griffin MJ, Price E, Parry A, Dick GS, Little MA, of anticancer agent-incorporating polymer micelles. Cancer Sci
Yule SM, Morland B, Estlin EJ, Hale JP, Pearson AD, Wel- 100:572–579
bank H, Boddy AV (2001) A phase I study in paediatric 110. Plummer R, Wilson RH, Calvert H, Boddy AV, Griffin M, Slud-
patients to evaluate the safety and pharmacokinetics of SPI-77, den J, Tilby MJ, Eatock M, Pearson DG, Ottley CJ, Matsumura
a liposome encapsulated formulation of cisplatin. Br J Cancer Y, Kataoka K, Nishiya T (2011) A phase I clinical study of cis-
84:1029–1035 platin-incorporated polymeric micelles (NC-6004) in patients
95. Kim ES, Lu C, Khuri FR, Tonda M, Glisson BS, Liu D, Jung with solid tumours. Br J Cancer 104:593–598
M, Hong WK, Herbst RS (2001) A phase II study of STEALTH 111. Chen HH, Chen W, Liang Z, Tsai W, Long Y, Aiba I, Fu S,
cisplatin (SPI-77) in patients with advanced non-small cell lung Broaddus R, Liu J, Feun LG, Savaraj N, Kuo MT (2015) Target-
cancer. Lung Cancer 34:427–432 ing drug transport mechanisms for improving platinum-based
96. Perez-Soler R, Khokhar AR, Lopez-Berestein G (1987) Treat- cancer chemotherapy. Expert Opin Ther Targets 19:1307–1317
ment and prophylaxis of experimental liver metastases of 112. Scagliotti G, Novello S, von Pawel J, Reck M, Pereira JR,
M5076 reticulosarcoma with cis-bis-neodecanoato-trans-R, Thomas M, Abrão Miziara JE, Balint B, de Marinis F, Keller
R-1,2-diaminocyclohexaneplatinum(II) encapsulated in multila- A, Arén O, Csollak M, Albert I, Barrios CH, Grossi F, Krza-
mellar vesicles. Cancer Res 47:6462–6466 kowski M, Cupit L, Cihon F, Dimatteo S, Hanna N (2010)
97. Stathopoulos GP, Boulikas T, Kourvetaris A, Stathopoulos J Phase III study of carboplatin and paclitaxel alone or with
(2006) Liposomal oxaliplatin in the treatment of advanced can- sorafenib in advanced non-small-cell lung cancer. J Clin Oncol
cer: a phase I study. Anticancer Res 26:1489–1493 28:1835–1842
98. Hamelers IH, Staffhorst RW, Voortman J, de Kruijff B, Reedijk 113. Miller K, Morant R, Stenzl A, Zuna I, Wirth M (2016) A Phase
J, van Bergen en Henegouwen PM, de Kroon AI (2009) High II Study of the Central European Society of Anticancer-Drug
cytotoxicity of cisplatin nanocapsules in ovarian carcinoma Research (CESAR) Group: results of an Open-Label Study
cells depends on uptake by caveolae-mediated endocytosis. Clin of Gemcitabine plus Cisplatin with or without concomitant or
Cancer Res 15:1259–1268 sequential gefitinib in patients with advanced or metastatic tran-
99. Staffhorst RW, van der Born K, Erkelens CA, Hamelers IH, sitional cell carcinoma of the urothelium. Urol Int 96:5–13
Peters GJ, Boven E, de Kroon AI (2008) Antitumor activity and 114. Yu H, Zhang J, Wu X, Luo Z, Wang H, Sun S, Peng W, Qiao
biodistribution of cisplatin nanocapsules in nude mice bear- J, Feng Y, Wang J, Chang J (2014) A phase II randomized trial
ing human ovarian carcinoma xenografts. Anticancer Drugs evaluating gefitinib intercalated with pemetrexed/platinum
19:721–727 chemotherapy or pemetrexed/platinum chemotherapy alone in
100. Rademaker-Lakhai JM, Terret C, Howell SB, Baud CM, De unselected patients with advanced non-squamous non-small cell
Boer RF, Pluim D, Beijnen JH, Schellens JH, Droz J (2004) lung cancer. Cancer Biol Ther 15:832–839
A Phase I and pharmacological study of the platinum polymer 115. Balmaña J, Tung NM, Isakoff SJ, Graña B, Ryan PD, Saura
AP5280 given as an intravenous infusion once every 3 weeks in C, Lowe ES, Frewer P, Winer E, Baselga J, Garber JE (2014)
patients with solid tumors. Clin Cancer Res 10:3386–3395 Phase I trial of olaparib in combination with cisplatin for the
101. Nowotnik DP, Cvitkovic E (2009) ProLindac (AP5346): a treatment of patients with advanced breast, ovarian and other
review of the development of an HPMA DACH platinum poly- solid tumors. Ann Oncol 25:1656–1663
mer therapeutic. Adv Drug Deliv Rev 61:1214–1219 116. Novello S, Besse B, Felip E, Barlesi F, Mazieres J, Zalcman
102. Apps MG, Choi Eugene H Y, Wheate NJ (2015) The state- G, von Pawel J, Reck M, Cappuzzo F, Ferry D, Carcereny E,
of-play and future of platinum drugs. Endocr Relat Cancer Santoro A, Garcia-Ribas I, Scagliotti G, Soria J (2014) A phase
22:R219–R233 II randomized study evaluating the addition of iniparib to gem-
103. Malik N, Evagorou EG, Duncan R (1999) Dendrimer-platinate: citabine plus cisplatin as first-line therapy for metastatic non-
a novel approach to cancer chemotherapy. Anticancer Drugs small-cell lung cancer. Ann Oncol 25:2156–2162
10:767–776 117. Nagao S, Fujiwara K, Imafuku N, Kagawa R, Kozuka Y, Oda
104. Haririan I, Alavidjeh MS, Khorramizadeh MR, Ardestani MS, T, Maehata K, Ishikawa H, Koike H, Aotani E, Kohno I (2005)
Ghane ZZ, Namazi H (2010) Anionic linear-globular den- Difference of carboplatin clearance estimated by the Cockroft-
drimer-cis-platinum(II) conjugates promote cytotoxicity in vitro Gault, Jelliffe, Modified-Jelliffe, Wright or Chatelut formula.
against different cancer cell lines. Int J Nanomed 5:63–75 Gynecol Oncol 99:327–333

13

You might also like