You are on page 1of 17

Coordination Chemistry Reviews 360 (2018) 17–33

Contents lists available at ScienceDirect

Coordination Chemistry Reviews


journal homepage: www.elsevier.com/locate/ccr

Review

The mechanism of tumour cell death by metal-based anticancer drugs is


not only a matter of DNA interactions
Alberta Bergamo a, Paul J. Dyson b, Gianni Sava a,c,⇑
a
Callerio Foundation Onlus, via A. Fleming 22, 34127 Trieste, Italy
b
Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
c
Department of Life Sciences, University of Trieste, via A. Fleming 22, 34127 Trieste, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Platinum drugs are extensively used in the clinic to treat cancer, often leading to a palliative response
Received 4 July 2017 rather than a cure. While DNA is considered to be the primary target of platinum drugs, there is no clear
Accepted 7 January 2018 relationship between cellular platinum accumulation, DNA platination and Pt-DNA adduct removal, and
herein we describe new mechanistic insights of platinum drugs related to the hallmarks of cancer and
how they interfere with the tumour microenvironment. We then proceed to describe the properties of
Keywords: other metal drugs, including both non-targeted compounds that do not significantly interact with DNA
Platinum drugs
and targeted compounds that interfere more selectively with specific pathways responsible for tumour
Metal-based compounds
Cancer chemotherapy
growth and invasion. Our analysis of the cancer biology and the selected drugs allows us to propose pos-
DNA adducts sible routes for future drug development based on metal scaffolds.
Molecular tumour targets Ó 2018 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.1. Mechanism of action of platinum-based drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
1.2. Lessons from testicular cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
1.3. The post, post-genomic paradigms of cancer growth and treatment. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
1.3.1. Recognizing cancer complexity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
1.3.2. Tumour plasticity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
1.3.3. The microenvironment partnership. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
1.3.4. Where to orientate in such complexity?. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
1.3.5. Current trends in handling neoplastic diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
2. Re-visiting the concept of treating tumours with cisplatin in the era of the targeted therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
3. Treating tumours or the tumour microenvironment? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
4. Metal-complexes can target the determinants of tumour malignancy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

Abbreviations: 15-LOX-1, 15-lipoxygenase; AIF, apoptosis inducing factor; APC, adenomatous polyposis coli; AKT or PKB, protein kinase B; ATG16L2, autophagy related 16
like 2; AUC, area under the curve; Bad, Bcl-2 associated death promoter; Bcl-2, B-cell lymphoma 2; Bcl-xL, B-cell lymphoma extra large; bFGF, basic FGF; CAF, cancer
associated fibroblast; CASP, caspase; CLNDP, clearance of nedaplatin; CML, chronic myeloid leukemia; COX-2, cycloxygenase 2; CSC, cancer stem cell; CTL, cytotoxic T
lymphocyte; CTLA-4, cytotoxic T-lymphocyte antigen 4; DC, dendritic cells; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EMT, epithelial mesenchymal
transdifferentiation; ERK, extracellular signal-regulated kinase; FAK, focal adhesion kinase; FGF, fibroblast growth factor; HGF, hepatocyte growth factor; GLUT, glucose
transporter 1; HH, Hedgehog; HK2, hexokinase 2; LC3, light chain 3; LDH, lactate dehydrogenase; MAPK, mitogen activated protein kinase; Mcl-1, myeloid cell leukemia 1;
MDM4, mouse double minute 4; MDSC, myeloid-derived suppressor cells; MMP-9, matrix metalloproteinase 9; NCP, nucleosome core particle; NDP, Nedaplatin; NK, natural
killer; NSCLC, non-small cell lung cancer; PDGF-C, platelet-derived growth factor C; PDK, pyruvate dehydrogenase kinase; PDH, pyruvate dehydrogenase; PD-L, programmed
death receptor ligand; PI3K, phosphoinositide 3-kinase; PTEN, phosphatase and tensin homolog; STAT, signal transducer and activator of transcription; TGF-b, transforming
growth factor beta; TKI, tyrosine kinase inhibitor; TME, tumour microenvironment; TRAIL, tumour necrosis factor-related apoptosis inducing ligand; Treg, regulatory T cells;
TSP-1, thrombospondin-1; UCHL5, ubiquitin C-terminal hydrolase L5; ULK2, unc-51-like kinase; uPA, urokinase-typa plasminogen activator; uPAR, uPA receptor; UPS,
ubiquitinproteasome system; USP14, ubiquitin-specific protease; VEGF-A, vascular endothelial growth factor-A; VDAC, voltage-dependent anoin channels.
⇑ Corresponding author at: Dept. of Life Sciences, University of Trieste, Via A. Fleming 22, 34127 Trieste, Italy.
E-mail addresses: a.bergamo@callerio.org (A. Bergamo), paul.dyson@epfl.ch (P.J. Dyson), gsava@units.it (G. Sava).

https://doi.org/10.1016/j.ccr.2018.01.009
0010-8545/Ó 2018 Elsevier B.V. All rights reserved.
18 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

4.1. Sustaining proliferative signalling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24


4.2. Resisting cell death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.3. Inducing angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
4.4. Activating invasion and metastasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
4.5. Genome instability and mutation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
4.6. Reprogramming energy metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
4.7. Evading immune destruction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
5. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
Conflict of interest. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30

1. Introduction 1.1. Mechanism of action of platinum-based drugs

New metal-based anticancer compounds are systematically The principle mode of action of cisplatin and its analogues is
benchmarked against cisplatin, with respect to its chemical proper- believed to be due to binding to DNA in cancer cells with the cor-
ties, biological activity and therapeutic applications. Occasionally responding negative consequences for cell survival (Fig. 2) [5,6].
the other registered analogues of cisplatin (Fig. 1) are also used to Considerable progress has also been made in unravelling other
benchmark new putative metal-based anticancer drugs. Conse- mechanistic details, for example, understanding how platinated
quently, the approaches used to transfer metal-based compounds DNA regions are repaired (see below), which can lead to drug resis-
from the laboratory to the clinic, to add to the arsenal of chemother- tance. However, only a small fraction of cisplatin binds to DNA and
apeutics used to control cancer growth, has been ‘conditioned’ by the details surrounding the fate of the majority of the drug has yet
attempts to obtain compounds with similar therapeutic profiles to be fully elucidated, i.e. the full mechanism of action of cisplatin
to cisplatin, but without the drawbacks of cisplatin. Therefore, and the other platinum-based drugs remains largely unknown.
approaches to overcome the toxicity of cisplatin and/or tumour A rather simple molecule such as cisplatin is unable to distin-
resistance to cisplatin-containing chemotherapies dominate the guish between different cancer cells and the way cisplatin binds
field. This strategy had successfully produced two further drugs to cellular DNA is presumably independent of the type of cell. Stud-
approved world-wide (carboplatin and oxaliplatin) and three other ies on DNA repair mechanisms may explain resistance phenomena
analogues approved by China, Japan and South Korea (lobaplatin, of certain cancer cells to cisplatin, but it is often difficult to attri-
nedaplatin and heptaplatin, respectively) (Fig. 1 and Table 1) [1]. bute these repair mechanisms to different types of cancer cell
The introduction of new platinum-based derivatives into clini- [7,8]. Overall, the way in which cisplatin binds to DNA and the
cal studies has significantly slowed down and, in the last two dec- known resistance mechanisms do not adequately explain why,
ades, only very few analogues were selected, none of which even with a very responsive tumour such as testicular cancer, there
successfully completed the clinical trials and were proposed to are individuals who are completely cured, independent of the stage
the FDA or EMA for approval [1–4]. Since it is highly questionable of their disease, and others where treatment is only partially suc-
whether cisplatin would make it into the clinic today, we should, cessful or even completely ineffective [9]. The hypothesis that
perhaps, not be surprized that new metal-based drugs with pro- the main mechanism of action of cisplatin is based on its ability
files similar to cisplatin fail to gain clinical approval. An under- to form adducts with the cellular DNA, which cause distortions
standing of the reasons for this failure should help to identify that are not recognized by DNA repair mechanisms, has led to a
other approaches that may turn out to be more effective. large number of cisplatin-derivatives that failed to progress

Fig. 1. Structures of the main platinum drugs approved for cancer therapy. In black those approved for use world-wide; in blue those approved in specific countries
(Lobaplatin-China, Nedaplatin-Japan, Heptaplatin-Korea). Carboplatin represents the prototype of 2nd generation and oxaliplatin is the representative of the 3rd generation
of the clinically approved platinum drugs. The arrows trace the ancestry of the second and third generation derivatives of cisplatin.
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 19

Table 1
Main characteristics of the clinically approved platinum drugs (from S. Dilruba and G.V. Kalayda, 2013 [1]).

Drug Main characteristics Target tumours


Cisplatin The most clinically used platinum drug; included in many combination regimens Various cancers, among which ovarian, testicular, bladder, colorectal,
comprising biological/biotechnological drugs. lung and head and neck cancers
Carboplatin Same as cisplatin, perhaps less toxic and suitable for more aggressive high-dose Same as cisplatin but with limited efficacy against testicular germ-
chemotherapy. cell cancers, squamous cell carcinoma of the head and neck and
bladder cancer
Oxaliplatin Bulkiness and lipophilicity are believed responsible for differential processing of Mainly colorectal cancers but under clinical trials for pancreatic,
platinum-DNA adducts. gastric, breast and non-small cell lung cancers.
Nedaplatin Recommended therapeutic dose 80–100 mg/m2; PK profile similar to that of Non-small and small cell lung cancer, esophageal cancer, uterine
carboplatin. Ishibashi’s formula to predict the correct individual dose: Dose NDP cervical cancer, head and neck cancer, or urothelial cancer
= AUC  CLNDP, where CLNDP = 0.0738  creatinine clearance + 4.47. Limited
use in Japan. (Shimada et al., 2013, [181])
Lobaplatin Structurally very similar to oxaliplatin; standard dose: 35 mg/m2. Limited use in CML, small cell lung cancer, breast cancer
China.
Heptaplatin Active in cisplatin resistant tumours. Limited use in the Republic of Korea. Gastric cancer

AUC: area under the curve; CLNDP: clearance of NDP; CML = chronic myeloid leukemia; NDP: Nedaplatin.

However, palliation should not be confused with cure, and corre-


sponds to prolongation of the time to the exitus compared to the
natural history of the tumour. Nevertheless, the addition of plat-
inum compounds to chemotherapy regimens is useful for overall
survival in, for example, ovarian, lung, colorectal and breast cancers
and, in general, head-and-neck cancers, although in these cancers
cures are rarely obtained [see for example: [18–20]].
The natural question arising from this difference between tes-
ticular and other cancers is, therefore, what is the molecular basis
for this difference, which makes the latter group less responsive to
platinum drugs? And, if these differences are known, can they be
used to design new metal-based structures (of platinum or other
metals) capable of curing these cancers?
However, before answering these questions, it should be noted
that testicular cancer is not a unique type of tumour and there are
at least five types of tumours with different histologies and differ-
ent molecular pathways referred to as testicular cancer. Testicular
cancers can be roughly divided into seminomatous and non-
Fig. 2. Schematic representation of the mechanism of cytotoxicity induced by seminomatous tumours, and there are further sub-categories
platinum-based drugs. The drug enters the cell, undergoes activation (aquation)
within each of these two groups [21]. If differences between testic-
and then binds to guanine N-atoms in DNA forming inter- and intra-strand
crosslinks that lead to cell death. ular cancers can explain why some are cured whereas others are
not, then it should be possible to define the molecular pathways
that are activated by cisplatin-induced damage leading to the acti-
beyond pre-clinical studies or were subsequently abandoned dur- vation of cell death processes. An in vitro study exploring the sen-
ing clinical studies. sitivity of testicular tumours to cisplatin led to the conclusion that
The DNA targeting mechanism has conditioned the preclinical ‘‘. . .there seems to be no clear relationship between cisplatin-induced
development of drugs based on other transition metals. The over- apoptosis and (a) growth characteristics, (b) cellular Pt accumulation,
whelming majority of new metal compounds proposed for the (c) DNA platination, (d) Pt-DNA adduct removal, (e) p53 status, (f)
treatment of tumours are evaluated for their cytotoxic effects intrinsic Bcl-2 and Bax expression and (g) cisplatin-induced modula-
against a few cell lines to see if they are as active as cisplatin. This tion of the Bcl-2/Bax ratio.” (Fig. 3) [22]. Despite this analysis, many
approach ignores the biological target of the drug since it is usually attempts to design more effective platinum-based drugs have been
assumed that the active drug component interacts with DNA to based on obtaining compounds which lead to (a) higher amounts
produce lesions responsible for the cell death [10–12]. Mechanistic of DNA platination, (b) higher resistance to the removal of Pt-
studies therefore tend to be centred on how the compound DNA adducts and (c) higher levels of cellular platinum
releases or is transformed into the active species that subsequently accumulation.
binds to the DNA bases and on the speciation of the compound in Extending the analysis concerning testicular cancer to other
biological fluids [13–16]. cancer types should be taken with caution. Recent studies have
unravelled some aspects of the complexity of the sensitivity of tes-
ticular cancer to platinum drugs and, as mentioned above, the
1.2. Lessons from testicular cancer
expression of p53, Bcl-2 and Bax were not particularly relevant
for the induction of apoptosis by cisplatin [22]. In another
Of all cancers, testicular cancer is the most responsive to plat-
in vitro study of different testicular cancer cell lines it was con-
inum therapies, in which the chemotherapy is curative in a large
cluded that ‘‘. . .wild-type TP53 and the low levels of p53 in complex
proportion of the treated subjects even when started in advanced
with the p53 negative feed-back regulator MDM2 contribute to cis-
stages of tumour growth [9,17]. The activity of platinum drugs on
platin sensitivity.” and that ‘‘. . .the high levels of the pluripotency reg-
testicular cancer has not been reproduced in any other cancer types
ulator Oct4 . . . the high levels of miR-17/106b seed family and pro-
even if the chemotherapy regimens with platinum agents are used
apoptotic Noxa and the low levels of cytoplasmic p21 (WAF1/Cip1)
with success in the palliative treatment of many other tumours.
20 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

Fig. 3. Cytotoxicity of platinum drugs and characteristics of the pathways activated in treated testicular cancer cells [9,17,22].

appear to be causative for the exquisite sensitivity to cisplatin-based heterogeneity of neoplastic diseases. Heterogeneity concerns the
therapy of testicular cancer.” (Fig. 3) [9,17]. genetics, the epigenetics, the proteomics and the biochemistry of
In other words, it is not possible to expect the efficacy of cis- the tumour, and is manifest at intra-tumoural, inter-metastatic,
platin of tumours that do not show the same characteristics of intra-metastatic and inter-patient levels. From a genetic stance,
the responsive tumours, because only the cells of these tumours cells located at distant sites within a tumour mass show more dif-
have the possibility to activate the corrected pathways necessary ferences than neighbouring cells [23]. Such intra-tumour hetero-
to induce cell death. Therefore, it is not possible to overcome the geneity is relevant to tumour progression, since it provides the
resistance of tumours to cisplatin with new derivatives endowed basis for inter-metastatic heterogeneity among different meta-
with a higher capacity than cisplatin to penetrate the tumour cell, static lesions of the same patient, each originating from a founder
of producing Pt-DNA adducts, and being less susceptible to the cell, or small group of cells, with a very different mutation kit, and
mechanisms that remove these adducts. Instead, it is necessary likely originating from different and distinct primary tumour areas
to identify the characteristics of the pathways that regulate [23]. This situation has important implications regarding
tumour cell survival in order to design metal-based drugs that chemotherapeutic sensitivity and responses. In addition, the off-
induce the changes required to induce cell death. spring of the founder cell(s), as they continue to divide, can acquire
new and different mutations generating heterogeneity among the
1.3. The post, post-genomic paradigms of cancer growth and treatment cells of an individual metastasis, affecting the response to systemic
therapies and providing the seeds for drug resistance. Somatic
In recent years powerful new research tools and refined exper- mutations within tumours further underpin the heterogeneity
imental models have become available, and critical regulatory among tumours of different patients, and account for the unique
genes were identified changing the anatomical, histological, cellu- clinical course of each patient typically observed in clinical prac-
lar and molecular genetic landscape of cancer. This wealth of new tice. However, patient outcome is also determined by additional
information has shaped a new history and geography of tumours factors such as pharmacokinetics and non-genetic factors [24].
superseding the reductive view that tumours are merely masses
of abnormally proliferating cancer cells. 1.3.2. Tumour plasticity
During multistep tumour progression, certain clonal expansions
1.3.1. Recognizing cancer complexity may be elicited by non-mutational changes altering regulation of
Today we have a clearer view of cancer complexity. Tumours gene expression, e.g. through epigenetic mechanisms, i.e. DNA
are considered as real tissues, which are dependent on multiple methylation and histone modifications [25–27]. Human tumours
distinct cell types. The participation of cancer and host cells in het- harbour large numbers of epigenetic changes [28]. Interestingly,
erotypic interactions result in their constant and dynamic co- a feature of epigenetics is plasticity [29], implying that epigenetic
evolution, changing their characteristics with time. This implies changes are subject to microenvironmental influences [28].
that studying and understanding the characteristics of tumours It is noteworthy that phenotypic heterogeneity of cancer cells
should not be separated from considering the microenvironment, within a tumour mass may also develop from a genetically homo-
which is both a cause and a consequence of tumorigenesis. The geneous population due to the presence of cells in different states
main trait underlying tumour complexity is the tremendous of differentiation. This phenotypic plasticity can be traced to the
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 21

presence of a subclass of neoplastic cells within tumours, i.e. can- genesis, resisting cell death, deregulating cellular energetics, avoiding
cer stem cells (CSCs) [30,31]. In recent years, evidence has accumu- immune destruction, are expressed at varying degrees in almost
lated that CSCs are a common constituent of most tumours and all tumours. In addition, the acquisition of such features depends
they have been shown to express markers that are also present on two ‘‘enabling characteristics” i.e. genome instability and
in the normal stem cells of the tissue of origin, with which they tumour-promoting inflammation [42].
share self-renewal capability, and have the ability to generate Cancer is a genetic disease originating from and sustained by
new tumours [32,33]. The origin of CSCs remains to be clarified genomic instability. In the ‘‘breakthrough phase” a cell begins to
and may even vary from one tumour type to another. The acquisi- proliferate abnormally after the acquisition of a ‘‘driver-gene muta-
tion of CSC traits has been recently related with the epithelial mes- tion”, i.e. a mutation that confers a selective growth advantage to
enchymal transdifferentiation (EMT) program [34–36]. EMT allows the tumour cell [44]. Thus, a Mut-driver-gene is a gene that, if
cancer cells to be disseminated from the primary tumour, and can altered by intragenic mutations, can drive tumorigenesis. During
also confer the self-renewal capability needed for the clonal expan- the following ‘‘expansion phase” a second driver-gene mutation
sion at sites of dissemination. The connection between EMT and enables the cell to grow in its environment despite hostile condi-
CSCs implies that the heterotypic signals that elicit the transdiffer- tions, and subsequent mutations allow cells to invade and metas-
entiation process, such as those released by an inflammatory tasize. Sequencing analyses for more than 22,000 cancers have
stroma, may be important in creating and maintaining CSCs. By revealed more than 3 million somatic mutations, but around only
definition plasticity is a distinctive feature of CSCs that can shift 140 Mut-driver-genes [28,44]. Furthermore, considering Mut-
to a non-CSCs status and vice versa, but can also turn to produce driver-genes at a pathway level, rather than at an individual level,
stromal cell types as observed in glioblastomas [37–40]. The pres- there are only a limited number of cellular signalling pathways
ence of CSCs within the tumour mass affects cancer therapies since through which a growth advantage can be gained. All the known
resistance to treatments, and disease recurrence, may be attributed driver genes can be traced to one or more of 12 pathways that reg-
to the lower sensitivity of CSCs to commonly used drugs, and to ulate three core cellular processes, these being (i) cell fate (NOTCH,
their ability to regenerate the tumour once therapy has ceased. HH (Hedgehog), APC (Adenomatous Polyposis Coli), Chromatin
modification, Transcriptional regulation), (ii) cell survival (cell
1.3.3. The microenvironment partnership cycle/apoptosis, RAS, PI3K (Phosphoinositide 3-kinase), STAT (Sig-
Most heterogeneity is manifest in the tumour microenviron- nal Transducer and Activator of Transcription), MAPK (Mitogen
ment (TME), which is populated by a set of specialized cell types Activated Protein Kinase), TGF-b (Transforming Growth Factor
including multipotent stromal cells/mesenchymal stem cells, beta)) and (iii) genome maintenance (DNA damage control). A typ-
fibroblasts, endothelial cell precursors, and immune cells [41]. ical tumour contains 2 to 8 driver-gene mutations, moreover the
During tumour progression the abundance and the phenotypic protein products of genes regulate cell fate, cell survival and gen-
characteristics of these cell types change, as well as the composi- ome maintenance and often interact with one another, so that
tion of the extracellular matrix (ECM) due to the interplay between the pathways overlap [44]. This means that therapeutic efforts
the tumour cells (parenchyma) and the mesenchymal cells forming can be restricted to a few pathways.
the tumour-associated stroma. These heterotypic interactions cre-
ate a succession of tumour microenvironments that change with 1.3.5. Current trends in handling neoplastic diseases
time and enable primary, invasive and then metastatic growth Based on the current understanding of neoplastic diseases the
[42]. Cancer cell biology is governed by extremely sophisticated paradigms of cancer pharmacological treatments are also evolving
networks of interactions between neoplastic and stromal cells that with complete cancer eradication often considered to be a seem-
affect and, at the same time, are influenced by the evolution of the ingly unattainable goal. A better approach is to try to circumvent
surrounding ECM. This continuously changing dynamic is essential neoplastic disease by deploying an array of different strategies.
for tumour establishment, growth and dissemination. Indeed, the First, it is necessary to change the perspective and to consider
reciprocal interactions between cancer and stromal cells activate the treatment of cancer with an integrated, holistic view. Since cur-
dissemination and, in turn, enhance the neoplastic character of ing advanced cancers (those that cannot be eradicated by surgery
the cancer cells. Moreover, cancer cells can further stimulate the and have already metastasized) is unlikely, a more successful route
stroma to facilitate and expedite their reprogramming allowing is to consider cancer a chronic and manageable disease. Therefore,
them to invade adjacent tissues and disseminate. Such dynamics to reduce morbidity and mortality, efforts to potentiate prevention
characterizes multi-stage tumour development and prevents a full and make early diagnosis, two areas that benefit greatly from
understanding of cancer pathogenesis. recent developments in cancer biology, offer considerable opportu-
nities. In addition, molecular targeted therapies, targeting particu-
1.3.4. Where to orientate in such complexity? lar cancer mechanisms and hallmark capabilities, represent a
There is enormous cancer diversity comprising hundreds of significant advance in the treatment of cancer [45,46], but they
cancer types and subtypes that affect most organs and tissues of are not magic bullets. Indeed, over time, tumours engender resis-
the body and is brought about by genetic mutations and rearrange- tance to treatment, and relapses often occur after only a few
ments and by epigenetic reprogramming. This multiplicity also months [28]. Consequently, it is necessary to target as many cancer
relies on different recruitment of normal supporting cells at differ- capabilities as possible, rather than merely target a single mecha-
ent rates of disease progression. While this complexity is daunting, nism. In practice, it is usual to select an array of targets (the ‘‘Targe-
neoplastic disease is the result of an evolutionary process based on tome”) to develop a broad-spectrum therapeutic approach. This
a small set of organizing principles, which display an underlying implies employing a ‘‘network pharmacology” plan, shifting from
order. A rationalization of cancer complexity was shaped in 2000 a target-oriented approach towards a network-directed strategy,
by Hanahan and Weinberg [43], and was more recently revisited with the goal of interfering with multiple parts of a biological sys-
by the same authors [42]. They traced cancer to eight hallmarks, tem. Thus, it is possible to directly target the tumour cells or indi-
‘‘distinctive and complementary capabilities that enable tumour rectly inactivate and/or neutralize cells of the microenvironment.
growth and metastatic dissemination”, allowing the biology of neo- However, in order to select which cancer hallmark to target, it is
plastic disease to be understood. These hallmarks, i.e. sustaining necessary to consider the detailed cancer biology. The search for
proliferative signalling, evading growth suppressors, activating inva- drugs for advanced cancers should not be generic, as each tumour
sion and metastasis, enabling replicative immortality, inducing angio- type has peculiar characteristics depending on the growth stage,
22 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

Fig. 4. Schematic of the pathways involved in the modulation of the response of tumour cells to platinum drugs.

organ and composition of the microenvironment. Therefore, one or It appears that only targeted therapies can effectively treat resis-
more molecules sustaining a specific hallmark in a well-defined tant tumours. Note that data from clinical trials suggest that the
tumour, at a particular stage of growth, should be targeted. anti-tumour effects of the platinum-based drugs depend on the
Each tumour and even each tumour setting is unique and two molecular characteristics of the transcriptome/proteome of the
examples are particularly relevant to illustrate this concept. First, cells (see below).
a gene can function differently in different tumour types, as in It is known that only a small amount of cisplatin that enters a
the case of NOTCH, which genetic data indicate to be an oncogene cancer cell is found in the nucleus and therefore the different cel-
in lymphomas and leukemias and a tumour suppressor gene in lular responses to platinum-drugs could be based on the differ-
squamous cell carcinomas [47–50]. Second, pathway functions ences in pathways activated by the individual cells rather than
are different, depending on the organism, cell type, and precise only to DNA binding. In this respect a number of clinical studies
genetic alterations in a cell [51]. This difference explains why treat- show the dependence of the tumour response to cisplatin on the
ment with drugs inhibiting mutant BRAF kinase activity gives dra- presence of specific mutations in specific pathways of the tumour
matic remissions in the majority of melanoma patients harbouring cells (Fig. 4).
the BRAF V600E mutation [52], whereas the same drugs have no The significantly greater response of non small cell lung cancer
therapeutic effect in colorectal cancer patients harbouring the (NSCLC) patients with the WEE1 rs3910384 G/G homozygote geno-
same genetic alteration [53]. The expression of Epidermal Growth type compared to those with the A/A + A/G genotype to the combi-
Factor Receptor (EGFR) in colorectal cancer, but not in melanoma, nation therapy of cisplatin and gemcitabine [55], suggests the
is liable for the circumvention of the growth-inhibitory effects of greater importance of downstream pathways over the initial DNA
the BRAF inhibitors. damage, where the activity of an important checkpoint, such as
WEE1 for the regulation of the cell cycle progression, becomes
the limiting step for cell survival to the drug-induced changes
2. Re-visiting the concept of treating tumours with cisplatin in [56]. Similarly, the expression of the translational initiating factor
the era of the targeted therapy eIF3a negatively modulates the cells capacity to react to the
platinum-induced DNA lesions and, correspondingly, the tumours
The cytotoxicity of platinum drugs cannot be solely attributed in which this factor is down-regulated (e.g. in ovarian cancer,
to the principles of their reactivity with DNA of cancer cells, a con- NSCLC and nasopharyngeal carcinomas) gain a survival benefit
cept that has often been used to support the lack of effectiveness of and become resistant to these drugs [57–59].
many derivatives in experimental models [see for example [54]]. Growing evidence supports the role of non-coding microRNAs in
The anticancer activity of a platinum-based complex, and indeed the sensitivity/resistance of tumours to platinum drugs. An example
of any metal-based compound, whether expressed as cytotoxicity involves miR-193b, a tumour suppressor factor in association with
or inhibition of cancer growth, depends on the ability of the com- Mcl-1 (Myeloid cell leukemia 1) in hepatocellular carcinoma, which
pound to activate cancer cell death pathways (see the example in becomes determinant to enhance the sensitivity of this tumour to
Fig. 3). The study of cell growth and differentiation has revealed platinum therapy [60]. In squamous cell carcinoma miR 885-3p also
that all cells have several pathways capable of activating their plays a crucial role, through the modulation of the cell mRNAs of
own suicide or their survival. Tumour cells do not differ from this MDM4 (Mouse Double Minute 4), AKT1 (also known as Protein
general rule, although they may have hidden the death pathways Kinase B PKB), BCL2 (B-Cell Lymphoma 2), ATG16L2 (Autophagy
by overexpression of other signals. Hence, targeted drugs are able Related 16 Like 2), ULK2 (Unc-51-Like Kinase), CASP2 and CASP3
to act against tumours that are apparently resistant to conven- (Caspase 2 and 3), leading to an increased sensitivity to cisplatin
tional therapies as they disturb the overexpression of these signals. [61,62]. Similarly, the down-regulation of Breast Related Cancer
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 23

Antigen BRCA1-IRIS, a pathway involved in the promotion of metas- may lead to the induction of malignant traits. It is now established
tasis and in the resistance to anoikis of tumour cells, sensitizes ovar- that the gain of a malignant phenotype is not the result of a direct
ian cancer cells to low doses of platinum therapy [63]. Support to the effect of the stimuli on tumour cells but, rather, the consequence of
role of the presence of BRCA mutations was also given by a study [64] a stimulus-promoted cross-talk between tumour cells and other
in patients with metastatic triple negative breast cancer who cell types within the TME [80]. Furthermore, recent findings have
received cisplatin or carboplatin in monotherapy. Similarly, another uncovered novel roles for the TME in modulating therapeutic effi-
study showed that oxaliplatin-based therapy of colorectal cancer cacy, and stromal signatures have been shown to have powerful
patients is much more effective when the tumours are represented prognostic value in predicting treatment outcome [81–84]. There
by the KRAS mutant subtypes [65]. are many examples of how the TME can affect drug response.
Another interesting aspect involves the capacity of platinum Tumours resistant to alkylating agents become sensitive to the
drugs to prevent the development of acquired resistance of certain same agents when grown ex vivo in the absence of other cells
cancer cells to the targeted therapies. Although there are only a [85]. Colon carcinoma cells injected in orthotopic metastatic sites
few examples available, the study with Gefitinib in advanced show a different response to doxorubicin when compared with
NSCLC patients clearly shows that the addition of a platinum ther- an ectopic subcutaneous implant [86]. Melanoma may become
apy prevents the onset of resistance after the initial sensitivity resistant to inhibitors of mutant V600E BRAF due to Hepatocyte
shown to this specific tyrosine kinase inhibitor (TKI) [66]. This syn- Growth Factor (HGF), a stroma-derived paracrine growth factor
ergy with targeted therapies is an important aspect of the actual secreted in the TME [83,84]. Refractoriness to anti-angiogenic ther-
use of the platinum drugs in cancer chemotherapy and can over- apy in vivo can result from elevation of pro-angiogenic Platelet-
come cancer resistance due to specific mutations, e.g. the combina- Derived Growth Factor C (PDGF-C) expression by cancer associated
tion therapy comprising oxaliplatin and dovitinib, a TKI, in fibroblasts (CAFs) [87]. In glioblastomas, anti-angiogenic therapies
colorectal cancers irrespective of their Ras-Raf mutation state may even increase local invasion and metastasis as cancer cells
[67]. In other situations, targeted therapies may suppress the may reduce their dependence on a particular hallmark capability
capacity of cancer cells to repair the damage induced to DNA, (angiogenesis), becoming more dependent on another (invasion
therefore amplifying the activity of platinum drugs, as shown by and metastasis) [42]. This last example introduces a basic notion,
the use of a Bcl-2 inhibitor in NSCLC [68]. i.e. partially redundant signalling pathways regulate each of the
A new frontier for conventional platinum drugs, i.e. cisplatin, core hallmark capabilities. In addition, stromal paracrine factors,
carboplatin and oxaliplatin, is represented by their utility in com- acting as mediators of malignant traits, also appear highly redun-
bination with the novel anticancer immunotherapies [69]. Since it dant [80]. The main consequence for therapeutic design and plan-
was demonstrated that the damage induced by cisplatin or oxali- ning is that the inhibition of one key pathway in a tumour may not
platin on tumour cells leads to the release of factors that promote completely eradicate a hallmark capability [42].
the recruitment and activation of macrophage-dependent lympho- Tumour and non-tumour cells work to the development and
cyte reactions against the tumour cells, combinations of platinum progression of several neoplastic diseases, thereby non-tumour
drugs with MoAb-based treatments have found new perspectives cells may constitute legitimate targets for therapeutic intervention.
in the clinic [70]. Signs of the induction of immunogenic cell death, In addition to the cell type, a rationale therapeutic approach should
such as the exposure of calreticulin at the cell surface, by the com- comprise the orchestrated inhibition of multiple key molecules
bined regimen of cisplatin plus pyridoxine in NSCLC cells, require and pathways.
an intact immune system in order to lead to a significant anti- A particularly attractive example is the targeting of the immune
tumour response [71]. With this action platinum drugs negatively cells of the TME. Both the innate and adaptive immune system
regulate the signal transducer and activation transcription factor 6 infiltrate many tumours [88,89], which may be interpreted as a
(STAT6) dependent programmed death receptor ligand 2 (PD-L2) in sign of inflammatory conditions within the tumour mass, reflecting
dendritic cells, resulting in an enhanced immune response against an attempt by the immune system to inhibit tumour development.
the tumour cells [72–74]. Now it is recognized that tumour-associated inflammatory cells
The feature that connects all these events is the interaction of have a conflicting dual role. Indeed, both tumour-antagonizing
the platinum drugs with DNA together with the corresponding and tumour-promoting leukocytes can be found in most cancers.
descending events [7]. However, the interaction with DNA is only It is now widely accepted that tumour inflammation is a potent
one step in the anti-tumour activity of cisplatin and of its clinically motor that promotes a microenvironment that favours many steps
used analogues. Besides inducing DNA lesions, it is equally, if not of carcinogenesis, namely the expansion of genomic aberrations,
more important, that the target cell is unable to repair such damage cellular transformation, apoptosis evasion, invasion, amongst
or, alternatively, that the platinum drug interacts also with other others [90–94]. As tumour growth progresses, macrophages and
targets to lead the cell to activate its death mechanisms [75]. The mast cells secrete matrix degrading enzymes, chemokines and
interaction with multiple targets, besides nucleic acids, may ulti- cytokines, which elicit local stromal cells to recruit circulating
mately be responsible for the success or of the failure of the therapy leukocytes into tumour tissue causing an acute inflammatory sta-
[76–79]. Therefore, to some extent platinum drugs behave similarly tus [95]. However, the imbalance between the two processes driv-
to targeted therapies, mimicking what is often observed with small ing the repair of the damaged tissues, i.e. apoptosis and wound
molecules used to target tyrosine kinase receptors, the proteasome healing, turns an acute process into a chronic inflammation, which
or something else overexpressed by a cancer cell. led to the description of tumours as ‘‘wounds that never heal”
[89,96]. The re-establishment of the balance between the contrast-
ing inflammatory responses in tumours is behind the concept of
3. Treating tumours or the tumour microenvironment? therapies designed to redirect immune system cells towards
tumour destruction [97,98]. This can be achieved by both selective
Tumours in patients inherently grow in an interactive milieu of anti-inflammatory drugs, and by immune re-education, stimulat-
tumour cells and stroma, a term referring to all non-malignant ing again the immune arm able to combat tumour cells. It seems
cells, including endothelial cells, fibroblasts, and infiltrating leuko- likely that cancer immunotherapy will become a major part of
cytes, as well as extracellular matrix proteins. The tumour the combination treatment plan for patients with many cancer
microenvironment (TME) produces different types of stimuli cap- types [99]. Indeed, patients with cancers at advanced stages of
able of endowing tumour cells with aggressive behaviour, which growth show a durable anti-tumour response to drugs targeting
24 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

a a

Fig. 5a. The ruthenium(II) compound [(g6-p-cymene)Ru(6-(2-(2-(1H-imidazol-


1yl))ethoxy)-4-(30 -chloro-40 fluoroanilino)-7-methoxy-quinazolin)Cl2] induces
apoptosis at earlier stage than the ruthenium moiety and the anilinoquinazoline Fig. 6a. Examples of metal compounds that contrast the resistance of tumour cells
ligand alone through the inhibition of EGFR and the interaction with the DNA minor to cell death mechanisms. The ruthenium polypyridyl complex RuPOP ([Ru(phen)2-
groove [105]. (EGFR = Epithelial Growth Factor Receptor). p-MOPIP][PF6]2) induces mitochondria-mediated and caspase-dependent apoptosis
[106]. NiPT, a nickel pyrithione compound potently inhibits two deubiquitinases
associated to the 19S proteasome [109,110]. The ruthenium complex K-WH0402
(K-bipyridine 2-(2-trifluoromethphenyl)imidazole[4,5-f][1,10]phenanthroline
b ruthenium(II) chloride, K-[Ru(bpy)2-(o-tFPIP)]Cl2 induces autophagy by increasing
the expression of Beclin-1 and the conversion of the cytoplasmic form of
microtubule-associated protein 1 light chain 3 (LC3 I) to LC3 II [114]. (LC3 I =
Light Chain 3 I; LC3 II = Light Chain 3 II; UPS = Ubiquitin-Proteasome System).

Fig. 5b. Examples of Ru(II) and Ru(III) derivatives of 4-anilinoquinazoline.

pathways of the immune response, such as the T-cell receptors


Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) and programmed
death ligand 1 (PD-L1), in therapeutic approaches that can be tai-
lored also to different TMEs [99,100].
Fig. 6b. Structures of ruthenium polypyridyl (RuPOP) complexes.

4. Metal-complexes can target the determinants of tumour


malignancy
binding subsequently leads to a number of down-stream signalling
While DNA is often the assumed target of metal-based drugs, pathways. The epidermal growth factor receptor (EGFR) is at the
recent studies show that some compounds act via non-DNA dam- forefront of many of these pathways and several therapeutic
aging mechanisms and are able to target the hallmarks of cancer. strategies have been developed to target it [101,102]. A series of
Indeed, these mechanisms might be much more prevalent than dual-functional ruthenium anti-tumour complexes exploit the
currently thought as most new metal-based anticancer compounds ‘‘pharmacophore conjugation” strategy [103–105]. These complexes
are merely evaluated for cytotoxic effects in vitro and compared to contain 4-anilinoquinazoline ligands, an EGFR inhibitor. The EGFR
cisplatin, as mentioned above. inhibitory activity is retained and, in addition, a high affinity
Here we report selected examples of metal-complexes by cate- towards DNA via minor groove binding was observed. In particular,
gorizing them according to the specific cancer hallmark affected by both Ru(II) and Ru(III) derivatives of 4-anilinoquinazoline have
the compound. anti-proliferative activity towards the EGF-stimulated growth of
MCF-7 human breast cancer cells [103,104]. Related complexes
4.1. Sustaining proliferative signalling with a 6-(2-(2-(1H-imidazol-1yl))ethoxy)-4-(30 -chloro-40 fluoroani
lino)-7-methoxy-quinazoline) ligand were shown to induce apop-
The uncontrolled proliferation of cancer cells is enabled by tosis at an earlier stage than the 4-anilinoquinazoline-based com-
growth factors that interact with cell-surface receptors in which pounds (Figs. 5a and 5b) [105].
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 25

Fig. 7. Structure of K-WH0402.

4.2. Resisting cell death

Among cell death mechanisms, apoptosis is considered a hurdle


to cancer development and growth. Elucidation of molecular cir-
cuitries governing the apoptotic process has revealed various Fig. 8a. Examples of anti-angiogenic metal compounds. The 8-hydroxyquinoline
ruthenium(II) complexes BQ ([Ru(bpy)2(8-HQ)]+) and PQ ([Ru(phen)2(8-HQ)]+)
inducing stresses, both naturally occurring during tumorigenesis,
interfere with the binding of bFGF to its receptor thus disrupting the down-stream
or consequent to anti-cancer therapy. Certain ruthenium com- signalling pathway and suppressing several features of endothelial cells [124]. Ru-
pounds display anti-cancer properties through the activation of SeNPs (ruthenium(II) polypyridyl functionalized selenium nanoparticles) interact
apoptotic processes (Figs. 6a and b). For example, a series of ruthe- also with bFGF causing profound change of its conformation [125]. (bFGF = basic
Fibroblast Growth Factor; bFGFR = basic Fibroblast Growth Factor Receptor).
nium functionalised polypyridyl complexes, e.g. [Ru(phen)2-p-
MOPIP][PF6]2 (RuPOP) induce mitochondria-mediated and
caspase-dependent apoptosis in human cancer cell lines, thus
exerting anti-proliferative activity [106]. RuPOP suppresses the b
expression of the anti-apoptotic Bcl-2 and Bcl-xL (B-cell lymphoma
extra large) proteins, and up-regulates the expression of the pro-
apoptotic protein Bad (Bcl-2 associated death promoter). These
changes lead to loss of the mitochondrial membrane potential
and to the release of apoptogenic factors such as cytochrome c
and apoptosis inducing factor (AIF). More recently, RuPOP was
shown to synergistically enhance tumor necrosis factor-related
apoptosis inducing ligand (TRAIL)-induced apoptosis in the triple
negative breast cancer cell line MDA-MB-231, suggesting that the
Fig. 8b. Structure of 8-hydroxyquinoline ruthenium(II) complexes [Ru(bpy)2(8-
combined treatment of RuPOP and TRAIL represents a novel strat- HQ)]+ (BQ) and [Ru(phen)2(8-HQ)]+ (PQ) (left) and the complex that has been
egy to inhibit the growth of this aggressive subtype of breast can- attached ot selenium nanoparticles (right).
cer [107].
Cancer cell apoptosis can be induced by metal complexes
through the inhibition of the ubiquitin–proteasome system (UPS) tein 1 light chain 3 (LC3 I) to LC3 II, which is a marker of
[108]. Notably, proteasome inhibition alters the homeostatic autophagosome accumulation. It is worth noting that autophagy
mechanisms of cell protein catabolism, inducing cell death. Metal appears to have a conflicting role on cancer cells and on tumour
complexes based on copper, zinc and nickel exhibit this activity progression depending on the stimulus and the cell type
[109,110]. Notably, the nickel pyrithione compound NiPT potently [115,116]. In this case, however, the induction of autophagy inhi-
inhibits the UPS by targeting Ubiquitin C-terminal Hydrolase L5 bits tumour growth and progression in agreement with the activity
(UCHL5) and Ubiquitin-specific protease 14 (USP14), two deubiq- of a series of ruthenium(II) compounds containing a b-carboline
uitinases associated with the 19S proteasome, leading to apoptosis alkaloid as ligand [117].
in both cultured cancer cells and cells from acute myeloid leukae-
mia patients. The proteasome inhibition of NiPT has been validated 4.3. Inducing angiogenesis
in a xenograft model in nude mice where it affects tumour growth.
Autophagy is a key process induced when cells suffer stresses During cancer progression angiogenesis is always in play, which
such as nutrient deficiency [111,112]. Cells break down intracellu- is triggered mainly by vascular endothelial growth factor-A (VEGF-
lar proteins and organelles, through lysosome-mediated degrada- A) and its receptors, and by members of the fibroblast growth fac-
tion, to recycle the resulting catabolites and to use them for tor (FGF) family [118,119], and countered by factors that oppose
metabolism and biosynthesis. Growing evidence indicates that angiogenesis such as thrombospondin-1 (TSP-1) [119,120].
autophagy and apoptosis are intertwined [113]. The pro- Anti-angiogenesis agents have been introduced in various clin-
apoptotic protein Bcl-2 binds Beclin-1, which is responsible for ical cancer chemotherapies [121–123], and metal-based com-
triggering the canonical autophagy pathway. A ruthenium com- plexes also have been investigated for their anti-angiogenic
plex, K-bipyridine 2-(2-trifluoromethphenyl)imidazole[4,5-f] potential. 8-Hydroxyquinoline ruthenium(II) complexes [Ru
[1,10]phenanthroline ruthenium(II) chloride, K-WH0402 (Fig. 7), (bpy)2(8-HQ)]+ (BQ) and [Ru(phen)2(8-HQ)]+ (PQ) suppress the
induces autophagy in hepatocellular carcinoma cells HCCLM6 proliferation, migration, invasion, tube formation and microvessel
through a Beclin-1-dependent pathway [114]. K-WH0402 growth of endothelial cells in vitro [124]. The mechanism of action
increases the expression levels of Beclin-1 and decreases the inter- involves interfering with binding of bFGF to its receptors on the
action between Beclin-1 and Bcl-2. In addition, it promotes the cell surface and the consequent inactivation of bFGF-mediated sig-
conversion of the cytoplasmic form of microtubule-associated pro- nalling (Figs. 8a and 8b). The ability to interact with bFGF was
26 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

demonstrated also for ruthenium(II) polypyridyl functionalized


a selenium nanoparticles (Ru-SeNPs) [125]. In the presence of Ru-
SeNPs, bFGF undergoes a dramatic change of conformation, as
showed by circular dichroism experiments, and the direct interac-
tion of Ru-SeNPs was further confirmed by their ability to protect
bFGF from trypsin digestion. Other ruthenium(II) complexes have
been shown to provide angiostatic cancer treatments in vivo when
optimally combined with other agents [126].

4.4. Activating invasion and metastasis

Over the past decade the mechanisms underlying invasion and


metastasis have been the subject of numerous investigations and,
although many aspects need to be clarified, relevant features of
these processes have been revealed. The role of cell-to-cell and
cell-to-ECM adhesion molecules has been ascertained, as well that
of matrix-degrading enzymes, of epithelial to mesenchymal transi-
tion (EMT), and of the heterotypic contributions emanating from
Fig. 9a. Examples of metal compounds that inhibit invasion and metastasis. The the tumour stroma. Correspondingly, many metal compounds have
ruthenium complex RAWQ11 ([(g6-C6H6)Ru(H2iip)Cl]Cl) inhibits invasion and been tested as inhibitors of cancer cell invasion and metastasis,
metastasis through the targeting of miR-21 that affects downstream PTEN and
both in vitro and in vivo, and evaluated for their potential to target
AKT pathway [127]. RuPOP ([Ru(phen)2-p-MOPIP](PF6)22H2O) suppresses the
phosphorylation of FAK and then inactivates AKT and ERK signalling [107]. The these malignant characteristics (Figs. 9a and 9b). A ruthenium
phosphorylation of FAK is central also in the mechanism of action of the ruthenium complex of formula [(g6-C6H6)Ru(H2iip)Cl]Cl (RAWQ11) inhibits
compound NAMI-A ([ImH][RuCl4(DMSO)Im) [130], and the vanadium compound the invasion and metastasis of MDA-MB-231 cells, changing their
VOChrys ([VO(chrysin)2EtOH]2) [131]. (AKT/PKB = Protein Kinase B; MMP-9 = morphology, affecting the number of focal adhesions, and contrast-
Matrix Metallo Proteinase 9; p-ERK = phospho-Extracellular signal Regulated
Kinase; p-FAK = phospho-Focal Adhesion Kinase; PTEN = Phosphatase and Tensin
ing the invadopodia formation [127]. These functional effects
homologue; uPA = urokinase-type Plasminogen Activator; uPAR = urokinase-type result from the down-regulation of miR-21 and the subsequent
Plasminogen Activator Receptor). up-regulation of PTEN (Phosphatase and tensin homolog) that
leads to the negative regulation of the AKT pathway. This pathway
is considered to be central in metastasis inhibition induced by
another ruthenium complex, i.e. RuPOP [Ru(phen)2-p-MOPIP]
(PF6)2 (Fig. 6b), in MDA-MB-231 cells [107]. RuPOP inactivates
b
AKT and ERK (extracellular signal-regulated kinase) signalling by
decreasing the expression levels of their phosphorylated forms,
via the suppression of the phosphorylation of the Focal Adhesion
Kinase (FAK) at Tyr397. An additional effect of impaired FAK acti-
vation corresponds to the down-regulation of the ECM degrading
Fig. 9b. Structure of the ruthenium(II) complex RAWQ11.
enzymes including urokinase-type plasminogen activator (uPA),
urokinase-type plasminogen activator receptor (uPAR), and matrix
metalloproteinase 9 (MMP-9), that FAK usually stimulate through
the Ras/MEK/ERK and PI3K/Akt pathways [128,129].
FAK is emerging as a target of other metal complexes. The
ruthenium compound [RuCl4(DMSO)Im][ImH] (NAMI-A, Fig. 10)
reduces FAK auto-phosphorylation on Tyr397 by a mechanism
involving the a5b1 integrin, which modulation determines the
anti-migratory effect of NAMI-A in a colorectal cancer cell line,
and presumably being important for the anti-metastatic activity
displayed in a model of hepatic metastases in vivo [130]. An oxo-
vanadium(IV) complex with a flavonoid chrysin ligand [VO
(chrysin)2EtOH] (VOChrys) is also able to reduce the phosphoryla-
tion level of in situ expressed FAK, leading to alteration of the
related cell signalling pathway [131]. Ruthenium complexes devel-
oped as 15-lipoxygenase-1 (15-LOX-1) inhibitors for inflammatory
and respiratory diseases [132] could also exhibit anti-metastatic
properties. Lipoxygenases catalyse the formation of signalling
molecules such as leukotrienes, lipoxins and eoxins from polyun-
saturated fatty acids, which have a role in inflammatory lung dis-
eases [133]. Two types of complexes with the general formula
[Ru([9]aneS3)(dmso)(N,N- or N,O-donor ligand)](PF6)2 and [(g6-p-
cymene)(RuCl(O,O-ligand)]Cl (Fig. 10) inhibit 15-LOX-1 in the
micromolar range, comparable to that of a known 15-LOX inhibi-
tor, and occurring through an uncompetitive mechanism. LOXs
also have a role in cancer, both pro- and anti-metastatic, depending
Fig. 10. Structure of NAMI-A (top, left), VOChrys (top, right) and the 15-LOX-1
on the type of tumour [134,135]. In a melanoma model, the prod-
inhibitors [Ru([9]aneS3)(dmso)(N,N- or N,O-donor ligand)]2+ and [(g6-p-cymene) ucts of 15-LOX-1 enzymatic activity potentiate the lung homing of
(RuCl(O,O-ligand)]Cl inhibit (bottom). B16F10 melanoma cells in vivo and favour the in vitro adhesion on
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 27

collagen and fibronectin [136]. Notably, the 15-LOX-1 products


stem from the activation of ERK and FAK signalling pathways, b
thereby up-regulating the adhesion and the metastatic potential
of B16F10 melanoma cells. The inhibition of 15-LOX-1 by the
ruthenium compounds in this tumour model could therefore break
FAK signalling and attenuate the downstream metastatic regula-
tory proteins.

4.5. Genome instability and mutation

Alterations to the genome provide cancer cells with acquired


functional capabilities that allow them to survive, proliferate and
disseminate, e.g. by inactivating tumour suppressor genes. Among
the caretaker genes that maintain genome integrity, BRCA1 is
involved in the repair of DNA double-strand breaks, protein ubiq-
uitination, and transcriptional regulation [137], therefore its muta-
tions enhance the risk of developing cancers. Correspondingly, an
impaired BRCA1 function in chemotherapy treated patients is
related to increased sensitivity to treatment and improved survival
following platinum-containing regimens [138,139]. The ruthenium
(II) complexes containing the bidentate ligand 5-chloro-2- Fig. 11b. Structures of the ruthenium(II) complexes containing the bidentate 5-
(phenylazo)pyridine interact with the holo, Zn2+-bound BRCA1 chloro-2-(phenylazo)pyridine ligand (top) and RAPTA-C and RAED-C (bottom).
protein affecting its overall conformation, and impairing the
BRCA1-mediated ubiquitination, in particular it reduces the BRCA1
NCPs exhibit unique conformational features, determined by the
E3 ligase activity (Fig. 11a and 11b) [140]. Both these findings are
DNA sequence, which influences where the histone proteins prefer
observed also with platinum-based drugs [141,142] and other
to localize [144,145]. In addition, DNA conformational features are
ruthenium compounds [143]. The final effects of the direct interac-
also affected by nucleosome positioning, i.e. the same sequence
tion of these ruthenium compounds with the BRCA1 RING domain
positioned at different histone binding sites displays different con-
is cell death by apoptosis in breast tumour cell lines, suggest that
formational features, and is cell-type and cell-status dependent
this protein could be an attractive target for metal-complexes, in
[146]. In other words, beside epigenetic differences such as DNA
particular for the treatment of triple negative breast cancers.
methylation, type of histones and their acetylation, cancer and
Besides genome instability conferred by mutations, cancer cells
healthy cells can be distinguished on the basis of their nucleosome
earn selective advantages through epigenetic mechanisms that
positioning and compaction state depending on the particular gene
affect the regulation of gene expression. Most of the current
and its activation status. The understanding of the sequence-
knowledge on DNA-adduct formation of metal-based drugs was
dependent properties of nucleosomes has shown that small mole-
derived from studies with naked, i.e. protein-free, DNA, or short
cules can selectively recognize nucleosomal DNA, opening the way
oligonucleotide fragments. However, the actual substrate for
for highly site-selective nucleosomal double helix recognition by
DNA-targeting agents is chromatin, where DNA is packed around
metal-based compounds [146]. Studies on [(g6-p-cymene)Ru(1,3,
histone proteins in well-defined nucleosome core particles (NCPs).
5-triaza-7-phosphaadamantane)Cl2] RAPTA-C [147] show it to be
associated with the protein component of chromatin in treated
cancer cells, in agreement with the formation of adducts in the
nucleosome core at specific histone protein sites detected in vitro
a [148]. Conversely, [(g6-p-cymene)Ru(ethylenediamine)Cl]PF6
RAED-C, a ruthenium compound differing from RAPTA-C in only
an apparently modest ligand substitution, associates mainly with
the DNA.
A very recent work of the same authors further documents the
discovery that metal-based compounds can form site-specific
adducts on the histone proteins that package DNA into the nucle-
osome [149]. Auranofin and [(g6-toluene)Ru(1,3,5-triaza-7-phos
phaadamantane)Cl2] RAPTA-T (Fig. 12) both form histone protein
adducts in the nucleosome core, albeit at distantly related sites.
In addition, the presence of RAPTA-T facilitates auranofin adduct
formation. The synergism by RAPTA-T and auranofin appears to
be mediated via an allosteric mechanism within the nucleosome
and yields a synergistic activity in killing cancer cells. Further
recent findings also support the ability of other ruthenium com-
pounds to interact with histones. An a organoruthenium complex
[Ru(II)(phenanthroline)( j -C,N-(2-phenyl-pyridine)(NCMe)2]PF6
Fig. 11a. Metal compounds affecting genome caretakers and epigenetic mecha- (RDC11) binds to histones H3.1, H2A and H2B in cells and in vitro
nisms. The ruthenium(II) complexes containing the bidentate ligand 5-chloro-2- on purified histones (Fig. 11a). In addition, RDC11 impacts on H3
(phenylazo)pyridine interact with BRCA1 protein affecting its overall conformation, post-translational modification, in which these changes are differ-
and impairing the BRCA1-mediated ubiquitination [140]. RDC11 [Ru(II)(phenan-
ent from those caused by cisplatin, and they are also observed
throline)(j-C,N-(2-phenyl-pyridine)(NCMe)2]PF6 binds to histones H3.1, H2A and
H2B in cells and in vitro on purified histones [150]. (BRCA1 = Breast Cancer 1; H2A in vivo, corroborating their significance. Such epigenetic regulation
= Histone 2A; H2B = Histone 2B; H3 = Histone 3; H4 = Histone 4). suggests that RDC11 might alter several signalling pathways [150].
28 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

Fig. 12. Structure of RAPTA-T (left), auranofin (center) and RDC11 (right).

been shown that cisplatin re-directs cancer cells to oxidative phos-


a phorylation from the Warburg effect (Fig. 13a). A possible target to
achieve this effect is PDK, which mRNA levels are reduced by cis-
platin treatment, limiting the final step of glycolysis by inhibiting
pyruvate dehydrogenase (PDH) activity and subsequently the flux
of pyruvate into the Krebs cycle [158]. The interference with glu-
cose metabolism can also proceed through the cisplatin-induced
detachment of hexokinase 2 (HK2) from voltage-dependent anion
channels (VDAC) [159]. This binding provides access to mitochon-
drial ATP, therefore it has been suggested that up-regulation of
hexokinase expression is responsible for the Warburg effect
[160]. Conversely, the interruption of the HK2-VDAC binding fos-
ters the activation of pro-apoptotic proteins [159]. In support of
this mechanism is the finding that AKT, which inhibits apoptosis
and stimulates the binding of HK2 to VDAC, is highly activated in
cisplatin-resistant ovarian cancer cells [161].
The anti-metabolic activity of cisplatin was also demonstrated
in breast and cervical cancer cells [162]. In these models, cisplatin
Fig. 13a. Metal compounds interfering with glucose metabolism. Cisplatin affects reduces glycolysis by suppression of glycolysis-related proteins
several molecules of glucose metabolism, such as PDK, GLUT1 and HK-2/VDAC such as GLUT1, GLUT4 and LDH (Lactate dehydrogenase), restrain-
interaction [158,159]. The cyclometalated ruthenium(II) complex [Ru(phpy)
ing cell growth and proliferation. These effects take place through
(bpy)2]+ (bpy = 2,20 -bipyridine, phpyH = 2-phenylpyridine) is a non-competitive
inhibitor of LDH [163]. (GLUT1 = Glucose Transporter 1; HK-2 = Hexokinase 2; LDH
down-regulation of the integrin b5/FAK signalling pathway. Ruthe-
= Lactate Dehydrogenase; PDH = Pyruvate dehydrogenase; PDK = Pyruvate Dehy- nium compounds also impact on tumour metabolism, notably on
drogenase Kinase; VDAC = Voltage-Dependent Anion Channel). LDH activity [163]. The cyclometalated 2-phenylpyridinato ruthe-
nium(II) complex [Ru(phpy)(bpy)2]+ (bpy = 2,20 -bipyridine, phpy
H = 2-phenylpyridine), is cytotoxic towards gastric and colon can-
b

Fig. 13b. Structures of [Ru(bpy)3]2+ (left) and the cyclometalated 2-phenylpyrid-


inato ruthenium(II) complex [Ru(phpy)(bpy)2]+ (right).

4.6. Reprogramming energy metabolism

The Warburg effect describes the metabolic shift from oxidative


phosphorylation to aerobic glycolysis that takes place in tumours
[151–153]. To implement this switch cancer cells up-regulate the
Fig. 14. Metal compounds that act on the immune system. Platinum compounds
expression of pyruvate dehydrogenase kinase (PDK) and glucose potentiate the activation of T cells [74]. Notably, oxaliplatin induces tumour-
transporters, notably GLUT1, in order to increase glucose uptake specific immune responses [172], and cisplatin prompts granzyme-B tumour cell
and utilization [154–156]. Tumour cells benefit from this shift with killing. The ruthenium(II) organometallic compound UNICAM-1 ([Ru(p-cymene)(bis
the increased glycolysis allowing the utilization of glycolytic inter- (3,5 dimethylpyrazol-1-yl)methane)Cl]Cl) reverses tumour-elicited immune sup-
pression by significantly reducing the number of Treg cells infiltrating the primary
mediates in various synthetic pathways for molecules needed for
tumour [174]. (CTLs = Cytotoxic T Lymphocytes; MDSC = Myeloid Derived Suppres-
assembling new cells. In addition, the switch to aerobic glycolysis sor Cells; PD-L 2 = Programmed Death Ligand 2; STAT6 = Signal Transducer and
allows tumour cells to evade apoptosis [157]. Interestingly, it has Activator of Transcription 6; Treg = T Regulatory cells).
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 29

anilinoquinazoline compounds, and comparatively simple and less


rational compounds that affect cancer hallmarks eliciting some
intriguing effects. Both classes of compounds are important in can-
cer chemotherapy, the former being more targeted whereas the
latter induce a broad range of responses on both the tumour and
TME.
Fig. 15. The structure of UNICAM-1.

5. Conclusions
cer and inhibits purified LDH (Figs. 13a and 13b). The kinetic inhi-
bition mechanism reveals a non-competitive inhibition, suggesting Analysis of the most recent data on the anti-tumour activity of
that the compound does not interact with the enzyme near the lac- platinum drugs is providing new insights into their mechanism of
tate/pyruvate or NAD+/NADH binding sites. Inhibition of LDH action. Unlike targeted drugs used in cancer chemotherapy, plat-
activity was confirmed also in treated cancer cells, and it could inum drugs and more generally most metal-based anticancer
be part of the mode of action of this compound, although it is unli- drugs, do not target an overexpressed pathway. Rather, they have
kely to account for the entire cytotoxic effect. Interestingly, the in common the same target, e.g. cellular DNA, and their different
related polypyridine ruthenium(II) complex [Ru(bpy)3]2+, where reactivity distinguishes how they bind that target, e.g. how
one Ru-N bond replaces one Ru-C bond, interacts with LDH differ- strongly, for how long, etc. This paradigm has long supported the
ently, and correspondingly is not cytotoxic to tumour cells. concept that the resistance to these therapies is mainly based on
the acquired (or innate) capacity of the tumour cells to repair the
DNA lesions. However, this view is losing impact and new, consis-
4.7. Evading immune destruction tent evidence, supports alternative possibilities that resistance or
sensitivity is a matter of the characteristic hallmark pathways
In recent years, an increasing body of evidence suggests the exhibited by the cancer cells. The necessity to treat more targets
two-faced role of the immune system in tumour formation and in the same cell in order to acquire a stronger and sustained cyto-
progression. Notably, the innate and adaptive arms of the immune toxicity is well known, but becomes more relevant with the intro-
system can be instrumental in tumour eradication [164,165]. How- duction of the targeted drugs. However, cells are able to overcome
ever, cancer cells may escape the actions put in place by the targeted drugs by rearranging the pathways that sustain their sur-
immune system to hold them in check. For example, cancer cells vival. Since the binding of a molecule to the DNA may rapidly
secrete immunosuppressive factors to restrain the activity of cyto- induce the activation of death pathways, it is not surprising that
toxic T lymphocytes (CTL) and natural killer (NK) cells [166,167]. In platinum drugs have been (and are still) used in order to synergize
addition, cancer cells can hamper the actions of CTLs by mobilizing with the targeted therapies for the treatment of many human car-
inflammatory cells possessing immunosuppressive features, such cinomas. It is also apparent that most metal-based drugs have
as regulatory T cells (Tregs) and myeloid-derived suppressor cells more than one target and potentially multiple targets. This charac-
(MDSCs) [168,169]. Recent reports have shed light on the modula- teristic should be considered as an opportunity that has not yet
tion of the immune system by metal-compounds as part of their received an adequate attention. Moreover, certain non-targeted
anti-tumour mechanism of action, including several examples of drugs, e.g. NAMI-A and RAPTA-C, do not primarily bind to DNA,
platinum compounds (Fig. 14) [72,170]. Platinum compounds which opens up further options in cancer treatment. Indeed,
potentiate the activation of T cells via dendritic cells (DCs) through NAMI-A and cisplatin were shown to function synergistically when
the inhibition of the STAT6 signalling pathway that causes the applied in combination [175]. Rational design of metal-based com-
down-regulation of the T cell-inhibitory molecule programmed pounds to target the multiple targets that sustain cancer hallmarks
death ligand 2 (PD-L 2) [74,171]. Oxaliplatin causes immunogenic would limit the off-targets effects often observed with drugs that
cell death, a combination of tumour cell stress and death that can affect additional targets beyond their own target. A better under-
induce tumour-specific immune responses, through non-DNA- standing of the ability of metal-based drugs to interact with pro-
binding effects [172]. In addition, platinum drugs sensitize cancer teins and other cell components (e.g. organelles such
cells to CTL-mediated attack, e.g. cisplatin promotes granzyme-B mitochondria rather than miRNAs or long non-coding RNAs, ribo-
killing by increasing the expression of mannose-6-phosphate on somes and ribozimes rather than the cell cytoskeleton and its com-
the cell surface [173]. ponents) should drive the design of more effective compounds.
The ruthenium(II) organometallic compound [Ru(p-cymene)(bi When the ruthenium compound cis-dichlorotetrakisdimethylsul
s(3,5dimethylpyrazol-1-yl)methane)Cl]Cl (UNICAM-1, Fig. 15) phoxideruthenium(II) was demonstrated to control the metastatic
reverses tumour-elicited immune suppression by significantly ability of cancer cells, the complexity of the biological phe-
reducing the number of Treg cells infiltrating the primary tumour nomenon of the metastatic growth and the limited knowledge of
in an experimental model of triple negative breast cancer [174]. the molecular aspects involved, prevented the precise mechanism
This effect is accompanied by an increase in CD4+ and CD8+ T lym- of that action being disclosed [176]. Subsequent research with the
phocytes. The modulation of the immune system by UNICAM-1 second generation ruthenium complexes (NAMI [RuCl4(DMSO)Im]
seems related to the reduction of Cyclooxygenase 2 (COX2) expres- Na and its analogues) have provided some insights into the
sion and is considered to be the prominent mechanism responsible potency of their anti-metastasis ability, albeit with limited useful
of the in vivo anti-tumour effects. information on the mechanism of action, except a subtle sugges-
The categorization of metal compounds on the basis of the tar- tion to a crucial role of the DMSO ligand(s) [177]. More recently,
geted hallmarks as reported above should be taken with caution. data showing the lack of penetration of NAMI-A into the cell and
Alternative categories are sometimes possible since partially the capacity to interact with integrins positioned on the outer cell
redundant signalling pathways regulate each of the core hallmarks membrane [130,178–180] are amongst the most important find-
capabilities and the same targets can belong to different pathways ings that might be useful for the synthesis of novel metal-based
and be involved in different cellular processes. An alternative view drugs.
is that there are two types of metal drugs, those with a functional Combined, these findings should represent the new paradigm
organic moiety attached that induces the response, e.g. the 4- for the definition of new and innovative metal-based drugs for
30 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

the treatment of malignant tumours, i.e. drugs that change the way [15] B. Michalke, Platinum speciation used for elucidating activation or inhibition
of Pt-containing anti-cancer drugs, J. Trace Elem. Med. Biol. 24 (2010) 69–77.
the tumours interact with the healthy cells in the niche of the pri-
[16] C.F. Harrington, A. Taylor, Analytical approaches to investigating metal-
mary or secondary sites by creating an in-hospital environment in containing drugs, J. Pharm. Biomed. Anal. 15 (2015) 210–217.
which tumour cells cannot receive the signals necessary to allow [17] A. di Pietro, E.G.E. de Vries, J.A. Gietema, D.C.J. Spierings, S. de Jong, Testicular
them to divide and grow. To achieve this effect it is necessary to germ cell tumours: The paradigm of chemo-sensitive solid tumours, Int. J.
Biochem. Cell Biol. 37 (2005) 2437–2456.
have a drug capable of interacting with more than one target and [18] X. Guan, F. Ma, Y. Fan, W. Zhu, R. Hong, B. Xu, Platinum-based chemotherapy
the metal-based drugs, because of their capacity to fill the biolog- in triple-negative breast cancer: a systematic review and meta-analysis of
ical space with a number of ligands, variably oriented in a tri- randomized-controlled trials, Anticancer Drugs 26 (2015) 894–901.
[19] D.D. Rosa, L.R. Medeiros, M.I. Edelweiss, P.R. Pohlmann, A.T. Stein, Adjuvant
dimensional way, can do that more efficiently than organic mole- platinum-based chemotherapy for early stage cervical cancer, Cochrane
cules. Finally, although beyond the scope of this review, it should Database Syst. Rev. 6 (2012) CD005342.
be stressed that, beyond focusing on the cancer target/pathways, [20] W.X. Qi, L.N. Tang, A.N. He, Z. Shen, F. Lin, Y. Yao, Doublet versus single
cytotoxic agent as first-line treatment for elderly patients with advanced
there are important strategies that influence the status of the effi- non-small-cell lung cancer: a systematic review and meta-analysis, Lung 190
cacy/effectiveness of metal-based compounds for cancer therapy. (2012) 477–485.
These include formulation aspects, modulation of the uptake and [21] Y. Sheikine, E. Genega, J. Melamed, P. Lee, V.E. Reuter, H. Ye, Molecular
genetics of testicular germ cell tumors, Am. J. Cancer Res. 2 (2012) 153–
more generally on the bio-distribution of the active species, as well 167.
as on their capacity to synergise with novel therapeutic biotechno- [22] H. Burger, K. Nooter, A.W. Boersma, C.J. Kortland, G. Stoner, Expression of p53,
logical interventions [182,183]. These key PK-PD aspects need to Bcl-2 and Bax in cisplatin-induced apoptosis in testicular germ cell tumour
cell lines, Br. J. Cancer 77 (1998) 1562–1567.
be considered when developing efficient anticancer metal drug
[23] S. Yachida, S. Jones, I. Bozic, T. Antal, R. Leary, B. Fu, M. Kamiyama, R.H.
therapies. Hruban, J.R. Eshleman, M.A. Nowak, V.E. Velculescu, K.W. Kinzler, B.
Vogelstein, C.A. Iacobuzio-Donahue, Distant metastasis occurs late during
the genetic evolution of pancreatic cancer, Nature 467 (2010) 1114–1117.
Funding [24] A. Kreso, C.A. O’Brien, P. van Galen, O.I. Gan, F. Notta, A.M. Brown, K. Ng, J. Ma,
E. Wienholds, C. Dunant, A. Pollett, S. Galliger, J. McPherson, C.G. Mullighan,
D. Shibata, J.E. Dick, Variable clonal repopulation dynamics influence
This research did not receive any specific grant from funding chemotherapy response in colorectal cancer, Science 339 (2013) 543–548.
agencies in the public, commercial, or not-for-profit sectors. [25] M. Berdasco, M. Esteller, Aberrant epigenetic landscape in cancer: how
cellular identity goes awry, Dev. Cell 19 (2010) 698–711.
[26] M. Esteller, Cancer epigenomics: DNA methylomes and histone-modification
Conflict of interest maps, Nat. Rev. Genet. 8 (2007) 286–298.
[27] P.A. Jones, S.B. Baylin, The epigenomics of cancer, Cell 128 (2007) 683–692.
[28] B. Vogelstein, N. Papadopoulos, V.E. Velculescu, S. Zhou, L.A. Diaz Jr, K.W.
None. Kinzler, Cancer genome landscapes, Science 339 (2013) 1546–1558.
[29] M. Pelizzola, J.R. Ecker, The DNA methylome, FEBS Lett. 585 (2011) 1994–
2000.
Acknowledgements [30] J. Ye, D. Wu, P. Wu, Z. Chen, J. Huang, The cancer stem cell niche: cross talk
between cancer stem cells and their microenvironment, Tumour Biol. 35
(2014) 3945–3951.
The authors would like to thank the EU COST Actions from D1 to [31] Y.E. Lau, N.P. Ho, T.K. Lee, Cancer stem cells and their microenvironment:
CM1105 and the entire inorganic chemistry community that took biology and therapeutic implications, Stem Cells Int. 2017 (2017) 3714190.
part in stimulating and profitable reflections and collaborations. [32] R.W. Cho, M.F. Clarke, Recent advances in cancer stem cells, Curr. Opin. Genet.
Dev. 18 (2008) 1–6.
[33] N.A. Lobo, Y. Shimono, D. Qian, M.F. Clarke, The biology of cancer stem cells,
References Annu. Rev. Cell. Dev. Biol. 23 (2007) 675–699.
[34] A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance: an
emerging axis of evil in the war on cancer, Oncogene 29 (2010) 4741–4751.
[1] S. Dilruba, G.V. Kalyda, Platinum-based drugs: past, present and future,
[35] S.A. Mani, W. Guo, M.J. Liao, E.N. Eaton, A. Ayyanan, A.Y. Zhou, M. Brooks, F.
Cancer Chemother. Pharmacol. 77 (2016) 1103–1124.
Reinhard, C.C. Zhang, M. Shipitsin, L.L. Campbell, K. Polyak, C. Brisken, J. Yang,
[2] M.G. Apps, E.H.Y. Choi, N.J. Wheate, The state-of-play and future of platinum
R.A. Weinberg, The epithelial-mesenchymal transition generates cells with
drugs, Endocr. Relat. Cancer 22 (2015) R219–R233.
properties of stem cells, Cell 133 (2008) 704–715.
[3] N.J. Wheate, S. Walker, G.E. Craig, R. Oun, The status of platinum anticancer
[36] A.P. Morel, M. Lievre, C. Thomas, G. Hinkal, S. Ansieau, A. Puisieux, Generation
drugs in the clinic and in clinical trials, Dalton Trans. 39 (2010) 8113–8127.
of breast cancer stem cells through epithelial-mesenchymal transition, PLoS
[4] T.C. Johnstone, K. Suntharalingam, S.J. Lippard, The next generation of
One 3 (2008) e2888.
platinum drugs: targeted Pt(II) agents, nanoparticle delivery, and Pt(IV)
[37] Y. Soda, T. Marumoto, D. Friedmann-Morvinski, M. Soda, F. Liu, H. Michiue, S.
prodrugs, Chem. Rev. 116 (2016) 3416–3486.
Pastorino, M. Yang, R.M. Hoffman, S. Kesari, I.M. Verma, Transdifferentiation
[5] R.C. Todd, S.J. Lippard, Inhibition of transcription by platinum antitumor
of glioblastoma cells into vascular endothelial cells, Proc. Natl. Acad. Sci. U.S.
compounds, Metallomics 1 (2009) 280–291.
A. 108 (2011) 4274–4280.
[6] D. Wang, S.J. Lippard, Cellular processing of platinum anticancer drugs, Nat.
[38] S. El Hallani, B. Boisselier, F. Peglion, A. Rousseau, C. Colin, A. Idbaih, Y. Marie,
Rev. Drug Discov. 4 (2005) 307–320.
K. Mokhtari, J.L. Thomas, A. Eichman, J.Y. Delattre, A.J. Maniotis, M. Sanson, A
[7] W.H. Ang, M. Myint, S.J. Lippard, Transcription inhibition by platinum DNA
new alternative mechanism in glioblastoma vascularization: tubular
cross-links in live mammalian cells, J. Am. Chem. Soc. 132 (2010) 7429–7435.
vasculogenic mimicry, Brain 133 (2010) 973–982.
[8] V. Brabec, J. Kasparkova, Modifications of DNA by platinum complexes.
[39] L. Ricci-Vitiani, R. Pallini, M. Biffoni, M. Todaro, G. Invernici, T. Cenci, G. Maira,
Relation to resistance of tumors to platinum antitumor drugs, Drug Resist.
E.A. Parati, G. Stassi, L.M. Larocca, R. De Maria, Tumour vascularization via
Updat. 8 (2005) 131–146.
endothelial differentiation of glioblastoma stem-like cells, Nature 468 (2010)
[9] R. Koster, M.A.T.M. van Vugt, H. Timmer-Bosscha, J.A. Gietema, S. de Jong,
824–828.
Unravelling mechanisms of cisplatin sensitivity and resistance in testicular
[40] R. Wang, K. Chadalavada, J. Wilshire, U. Kowalik, K.E. Hovinga, A. Geber, B.
cancer, Exp. Rev. Mol. Med. 15 (2013) e12.
Fligelman, M. Leversha, C. Brennan, V. Tabar, Glioblastoma stem-like cells
[10] V. Brabec, O. Novakova, DNA binding mode of ruthenium complexes and
give rise to tumour endothelium, Nature 468 (2010) 829–833.
relationship to tumor cell toxicity, Drug. Resist. Updat. 9 (2006) 111–122.
[41] P.A. Kenny, G.Y. Lee, M.J. Bissell, Targeting the tumor microenvironment,
[11] M. Chikuma, T. Sato, S. Komeda, Current status of and future perspectives for
Front. Biosci. 12 (2007) 3468–3474.
platinum antitumor drugs, Yakugaku Zasshi 128 (2008) 307–316.
[42] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell
[12] N. Farrell, Nonclassical platinum antitumor agents: perspectives for design
144 (2011) 646–674.
and development of new drugs complementary to cisplatin, Cancer Invest. 11
[43] D. Hanahan, R.A. Weinberg, The hallmarks of cancer, Cell 100 (2000) 57–70.
(1993) 578–589.
[44] B. Vogelstein, K.W. Kinzler, The path to cancer – three strikes and you’re out,
[13] J.W. Cox, S.J. Berners-Price, M.S. Davies, Y. Qu, N. Farrell, Kinetic analysis of
N. Engl. J. Med. 373 (2015) 1895–1898.
the stepwise formation of a long-range DNA interstrand cross-link by a
[45] M.O. Palumbo, P. Kavan, W.H. Miller Jr, L. Panasci, S. Assouline, N. Johnson, V.
dinuclear platinum antitumor complex: evidence for aquated intermediates
Cohen, F. Patenaude, M. Pollak, R.T. Jagoe, G. Batist, Systemic cancer therapy:
and formation of both kinetically and thermodynamically controlled
achievements and challenges that lie ahead, Front. Pharmacol. 4 (2013) 57.
conformers, J. Am. Chem. Soc. 123 (2001) 1316–1326.
[46] V. Kruse, S. Rottey, O. De Backer, S. Van Belle, V. Cocquyt, H. Denys, PARP
[14] R.A. Ruhayel, I. Zgani, S.J. Berners-Price, N.P. Farrell, Solution studies of
inhibitors in oncology: a new synthetic lethal approach to cancer therapy,
dinuclear polyamine-linked platinum-based antitumour complexes, Dalton
Acta Clin. Belg. 66 (2011) 2–9.
Trans. 40 (2011) 4147–4154.
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 31

[47] A.P. Weng, A.A. Ferrando, W. Lee, J.P. Morris 4th, L.B. Silverman, C. Sanchez- [66] S. Kanda, H. Horinouchi, Y. Fujiwara, H. Nokihara, N. Yamamoto, I. Sekine, H.
Irizarry, S.C. Blacklow, A.T. Look, J.C. Aster, Activating mutations of Kunitoh, K. Kubota, T. Tamura, Y. Ohe, Cytotoxic chemotherapy may
NOTCH1 in human T cell acute lymphoblastic leukemia, Science 306 (2004) overcome the development of acquired resistance to epidermal growth
269–271. factor receptor tyrosine kinase inhibitors (EGFR-TKIs) therapy, Lung Cancer
[48] N. Agrawal, M.J. Frederick, C.R. Pickering, C. Bettegowda, K. Chang, R.J. Li, C. 89 (2015) 287–293.
Fakhry, T.X. Xie, J. Zhang, J. Wang, N. Zhang, A.K. El-Naggar, S.A. Jasser, J.N. [67] S. Gaur, L. Chen, V. Ann, W.C. Lin, Y. Wang, V.H. Chang, N.Y. Hsu, H.S. Shia, Y.
Weinstein, L. Trevino, J.A. Drummond, D.M. Muzny, Y. Wu, L.D. Wood, R.H. Yen, Dovitinib synergizes with oxaliplatin in suppressing cell proliferation
Hruban, W.H. Westra, W.M. Koch, J.A. Califano, R.A. Gibbs, D. Sidransky, B. and inducing apoptosis in colorectal cancer cells regardless of RAS-RAF
Vogelstein, V.E. Velculescu, N. Papadopoulos, D.A. Wheeler, K.W. Kinzler, J.N. mutation status, Mol. Cancer 13 (2014).
Myers, Exome sequencing of head and neck squamous carcinoma reveals [68] T. Ren, J. Shan, M. Li, Y. Qing, C. Qian, G. Wang, Q. Li, G. Lu, C. Li, Y. Peng, H. Luo,
inactivating mutations in NOTCH1, Science 333 (2011) 1154–1157. S. Zhang, Y. Yang, Y. Cheng, D. Wang, S.F. Zhou, Small-molecule BH3 mimetic
[49] N. Stransky, A.M. Egloff, A.D. Tward, A.D. Kostic, K. Cibulskis, A. Sivachenko, G. and pan-Bcl-2 inhibitor AT-101 enhances the antitumor efficacy of cisplatin
V. Kryukov, M.S. Lawrence, C. Sougnez, A. McKenna, E. Shefler, A.H. Ramos, P. through inhibition of APE1 repair and redox activity in non-small-cell lung
Stojanov, S.L. Carter, D. Voet, M.L. Cortes, D. Auclair, M.F. Berger, G. Saksena, C. cancer, Drug Des. Devel. Ther. 9 (2015) 2887–2910.
Guiducci, R.C. Onofrio, M. Parkin, M. Romkes, J.L. Weissfeld, R.R. Seethala, L. [69] M. van Gelder, M.H. van Oers, W.G. Alemayehu, M.C. Abrahamse-Testroote, J.
Wang, C. Rangel-Escareno, J.C. Fernandez-Lopez, A. Hidalgo-Miranda, J. J. Cornelissen, M.E. Chamuleau, P. Zachée, M. Hoogendoorn, M. Nijland, E.J.
Melendez-Zajgla, W. Winckler, K. Ardlie, S.B. Gabriel, M. Meyerson, E.S. Petersen, A. Beeker, G.J. Timmers, L. Verdonck, M. Westerman, O. de Weerdt,
Lander, G. Getz, T.R. Golub, L.A. Garraway, J.R. Grandis, The mutational A.P. Kater, Efficacy of cisplatin-based immunochemotherapy plus alloSCT in
landscape of head and neck squamous cell carcinoma, Science 333 (2011) high-risk chronic lymphocytic leukemia: final results of a prospective
1157–1160. multicenter phase 2 HOVON study, Bone Marrow Transplant. 51 (2016)
[50] N. Agrawal, Y. Jiao, C. Bettegowda, S.M. Hutfless, Y. Wang, S. David, Y. Cheng, 799–806.
W.S. Twaddel, N.L. Latt, E.J. Shin, L.D. Wang, L. Wang, W. Yang, V.E. Velculescu, [70] R.K. Mehmood, Review of cisplatin and oxaliplatin in current immunogenic
B. Vogelstein, N. Papadopoulos, K.W. Kinzler, S.J. Meltzer, Comparative and monoclonal antibody treatments, Oncology Rev. 8 (2014) 256–263.
genomic analysis of esophageal adenocarcinoma and squamous cell [71] F. Aranda, N. Bloy, J. Pesquet, B. Petit, K. Chaba, A. Sauvat, O. Kepp, N. Khadra,
carcinoma, Cancer Discov. 2 (2012) 899–905. D. Enot, C. Pfirschke, M. Pittet, L. Zitvogel, G. Kroemer, L. Senovilla, Immune-
[51] D.A. Grueneberg, S. Gegot, J. Pearlberg, W. Li, J.E. Davies, A. Baldwin, W. dependent antineoplastic effects of cisplatin plus pyridoxine in non-small-
Endege, J. Doench, J. Sawyer, Y. Hu, F. Boyce, J. Xian, K. Munger, E. Harlow, cell lung cancer, Oncogene 34 (2015) 3053–3062.
Kinase requirements in human cells: I. Comparing kinase requirements [72] S.V. Hato, A. Khong, I.J.M. de Vries, W.J. Lesterhuis, Molecular pathways: the
across various cell types, Proc. Natl. Acad. Sci. U.S.A. 105 (2008) 16472– immunogenic effects of platinum-based chemotherapeutics, Clin. Cancer Res.
16477. 20 (2014) 2831–2837.
[52] P.B. Chapman, A. Hauschild, C. Robert, J.B. Haanen, P. Ascierto, J. Larkin, R. [73] S.V. Hato, I.J.M. de Vries, W.J. Lesterhuis, STATing the importance of immune
Dummer, C. Garbe, A. Testori, M. Maio, D. Hogg, P. Lorigan, C. Lebbe, T. Jouary, modulation by platinum chemotherapeutics, OncoImmunology 1 (2012)
D. Schadendorf, A. Ribas, S.J. O’Day, J.A. Sosman, J.M. Kirkwood, A.M. 234–236.
Eggermont, B. Dreno, K. Nolop, J. Lo, B. Nelson, J. Hou, R.J. Lee, K.T. Flaherty, [74] W.J. Lesterhuis, C.J.A. Punt, S.V. Hato, D. Eleveld-Trancikova, B.J.H. Jansen, S.
G.A. McArthur, BRIM-3 Study Group, Improved survival with vemurafenib in Nierkens, G. Schreibelt, A. de Boer, C.M.L. van Herpen, J.H. Kaanders, J.H.J.M.
melanoma with BRAF V600E mutation, N. Engl. J. Med. 364 (2011) 2507– van Krieken, G.J. Adema, C.G. Figdor, I.J.M. de Vries, Platinum-based drugs
2516. disrupt STAT6-mediated suppression of immune responses against cancer in
[53] M. Mao, F. Tian, J.M. Mariadason, C.C. Tsao, R. Lemos Jr, F. Dayyani, Y.N. Gopal, humans and mice, J. Clin. Invest. 121 (2011) 3100–3108.
Z.Q. Jiang, I.I. Wistuba, X.M. Tang, W.G. Bornman, G. Bollag, G.B. Mills, G. [75] Q.H. Miow, T.Z. Tan, J. Ye, J.A. Lau, T. Yokomizo, J.P. Thiery, S. Mori, Epithelial–
Powis, J. Desai, G.E. Gallick, M.A. Davies, S. Kopetz, Resistance to BRAF mesenchymal status renders differential responses to cisplatin in ovarian
inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition cancer, Oncogene 34 (2015) 1899–1907.
or demethylating agents, Clin. Cancer Res. 19 (2013) 657–667. [76] L. Gatti, G. Cassinelli, N. Zaffaroni, C. Lanzi, P. Perego, New mechanisms for old
[54] D.S. Godsell, The molecular perspective: Cisplatin, Stem Cells 24 (2006) 514– drugs: Insights into DNA-unrelated effects of platinum compounds and drug
515. resistance determinants, Drug. Resist. Updates 20 (2015) 1–11.
[55] D. Liu, C. Wu, Y. Jiao, L. Hou, D. Lu, H. Zheng, C. Chen, J. Qian, K. Fei, B. Su, [77] A. Macciò, C. Madeddu, Cisplatin : an old drug with a newfound efficacy -
WEE1 kinase polymorphism as a predictive biomarker for efficacy of from mechanisms of action to cytotoxicity, Exp. Opin. Pharmacother. 13
platinum-gemcitabine doublet chemotherapy in advanced non-small cell (2013) 1839–1857.
lung cancer patients, Sci. Rep. 5 (2015) 11114. [78] R. Mezencev, Interactions of cisplatin with non-DNA targets and their
[56] A. Brozovic, J. Damrot, R. Tsaryk, L. Helbig, T. Nikolova, C. Hartig, M. Osmak, influence on anticancer activity and drug toxicity: the complex world of
W.P. Roos, B. Kaina, G. Fritz, Cisplatin sensitivity is related to late DNA the platinum complex, Curr. Cancer Drug Targets 14 (2014) 1–23.
damage processing and checkpoint control rather than to the early DNA [79] P.M. Bruno, Y. Liu, G.Y. Park, J. Murai, C.E. Koch, T.J. Eisen, J.R. Pritchard, Y.
damage response, Mutat. Res. 670 (2009) 32–41. Pommier, S.J. Lippard, M.T. Heman, A subset of platinum-containing
[57] R.Y. Liu, Z. Dong, J. Liu, J.Y. Yin, L. Zhou, X. Wu, Y. Yang, W. Mo, W. Huang, S.K. chemotherapeutic agents kills cells by inducing ribosome biogenesis stress,
Khoo, J. Chen, D. Petillo, B.T. Teh, C.N. Qian, J.T. Zhang, Role of eIF3a in Nat. Med. 23 (2017) 461–473.
regulating cisplatin sensitivity and nucleotide excision repair of [80] F. Marcucci, M. Bellone, C.A. Caserta, A. Corti, Pushing tumor cells towards a
nasopharyngeal carcinomas, Oncogene 30 (2011) 4814–4823. malignant phenotype: stimuli from the microenvironment, intercellular
[58] J.Y. Yin, J. Shen, Z.Z. Dong, Q. Huang, M.Z. Zhong, D.Y. Feng, H.H. Zhou, J.T. communications and alternative roads, Int. J. Cancer 135 (2013) 1265–1276.
Zhang, Z.Q. Liu, Effect of eIF3a on response of lung cancer patients to [81] S.S. Dave, G. Wright, B. Tan, A. Rosenwald, R.D. Gascoyne, W.C. Chan, R.I.
platinum-based chemotherapy by regulating DNA repair, Clin. Cancer Res. 17 Fisher, R.M. Braziel, L.M. Rimsza, T.M. Grogan, T.P. Miller, M. LeBlanc, T.C.
(2011) 4–9. Greiner, D.D. Weisenburger, J.C. Lynch, J. Vose, J.O. Armitage, E.B. Smeland, S.
[59] Y. Zhang, J.J. Yu, Y. Tian, Z.Z. Li, C.Y. Zhang, S.F. Zhang, L.Q. Cao, Y. Zhang, C.Y. Kvaloy, H. Holte, J. Delabie, J.M. Connors, P.M. Lansdorp, Q. Ouyang, T.A. Lister,
Qian, W. Zhang, H.H. Zhou, J.Y. Yin, Z.Q. Liu, EIF3a improve cisplatin A.J. Davies, A.J. Norton, H.K. Muller-Hermelink, G. Ott, E. Campo, E.
sensitivity in ovarian cancer by regulating XPC and p27Kip1 translation, Montserrat, W.H. Wilson, E.S. Jaffe, R. Simon, L. Yang, J. Powell, H. Zhao, N.
Oncotarget 6 (2015) 25441–25451. Goldschmidt, M. Chiorazzi, L.M. Staudt, Prediction of survival in follicular
[60] W. Yin, Y. Nie, Z. Zhang, L. Xie, X. He, MiR-193b acts as a cisplatin sensitizer lymphoma based on molecular features of tumor-infiltrating immune cells,
via the caspase-3-dependent pathway in HCC chemotherapy, Oncol. Rep. 34 N. Engl. J. Med. 351 (2004) 2159–2169.
(2015) 368–374. [82] G. Finak, N. Bertos, F. Pepin, S. Sadekova, M. Souleimanova, H. Zhao, H. Chen,
[61] Y. Huang, A.Y. Chuang, E.A. Ratovitski, Phospho-DNp63a/miR-885-3p axis in G. Omeroglu, S. Meterissian, A. Omeroglu, M. Hallet, M. Park, Stromal gene
tumor cell life and cell death upon cisplatin exposure, Cell Cycle 10 (2011) expression predicts clinical outcome in breast cancer, Nat. Med. 14 (2008)
3938–3947. 518–527.
[62] Y. Huang, D. Kesselman, D. Kizub, R. Guerrero-Preston, E.A. Ratovitski, [83] R. Straussman, T. Morikawa, K. Shee, M. Barzily-Rokni, Z.R. Qian, J. Du, A.
Phospho-DNp63a/microRNA feedback regulation in squamous carcinoma Davia, M.M. Mongare, J. Gould, D.T. Frederick, Z.A. Cooper, P.B. Chapman, D.B.
cells upon cisplatin exposure, Cell Cycle 12 (2013) 684–697. Solit, A. Ribas, R.S. Lo, K.T. Flaherty, S. Ogino, J.A. Wargo, T.R. Golub, Tumour
[63] B.T. Paul, Z. Blanchard, M. Ridgway, W.M. ElShamy, BRCA1-IRIS inactivation micro-environment elicits innate resistance to RAF inhibitors through HGF
sensitizes ovarian tumors to cisplatin, Oncogene 34 (2015) 3036–3052. secretion, Nature 487 (2012) 500–504.
[64] S.J. Isakoff, E.L. Mayer, L. He, T.A. Traina, L.A. Carey, K.J. Krag, H.S. Rugo, M.C. [84] T.R. Wilson, J. Fridlyand, Y. Yan, E. Penuel, L. Burton, E. Chan, J. Peng, E. Lin, Y.
Liu, V. Stearns, S.E. Come, K.M. Timms, A.R. Hartman, D.R. Borger, D.M. Wang, J. Sosman, A. Ribas, J. Li, J. Moffat, D.P. Sutherlin, H. Koeppen, M.
Finkelstein, J.E. Garber, P.D. Ryan, E.P. Winer, P.E. Goss, L.W. Ellisen, Merchant, R. Neve, J. Settleman, Widespread potential for growth-factor-
TBCRC009: a multicenter phase II clinical trial of platinum monotherapy driven resistance to anticancer kinase inhibitors, Nature 487 (2012) 505–509.
with biomarker assessment in metastatic triple-negative breast cancer, J. [85] B.A. Teicher, T.S. Herman, S.A. Holden, Y.Y. Wang, M.R. Pfeffer, J.W. Crawford,
Clin. Oncol. 33 (2015) 1902–1909. E. Frei 3rd, Tumor resistance to alkylating agents conferred by mechanisms
[65] Y.L. Lin, Y.H. Liang, J.H. Tsai, J.Y. Liau, J.T. Liang, B.R. Lin, J.S. Hung, L.I. Lin, L.H. operative only in vivo, Science 247 (1990) 1457–1461.
Tseng, Y.L. Chang, K.H. Yeh, A.L. Cheng, Oxaliplatin-based chemotherapy is [86] I.J. Fidler, C. Wilmanns, R. Staroselsky, Z. Dong, D. Fan, Modulation of tumor
more beneficial in KRAS mutant than in KRAS wild-type metastatic colorectal cell response to chemotherapy by the organ environment, Cancer Metastasis
cancer patients, PLoS One 9 (2014) e86789. Rev. 13 (1994) 209–222.
32 A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33

[87] Y. Crawford, I. Kasman, L. Yu, C. Zhong, X. Wu, Z. Modrusan, J. Kaminker, N. autophagy to antagonize mitochondrial-mediated apoptosis, J. Med. Chem.
Ferrara, PDGF-C mediates the angiogenic and tumorigenic properties of 53 (2010) 7613–7624.
fibroblasts associated with tumors refractory to anti-VEGF treatment, Cancer [118] D. Hanahan, J. Folkman, Patterns and emerging mechanisms of the
Cell 15 (2009) 21–34. angiogenic switch during tumorigenesis, Cell 86 (1996) 353–364.
[88] R. Virchow, Cellular Pathology as based upon Physiological and Pathological [119] V. Baeriswyl, G. Christofori, The angiogenic switch in carcinogenesis, Semin.
Histology, J.B. Lippincott ed., Philadelphia, 1863. Cancer Biol. 19 (2009) 329–337.
[89] H.F. Dvorak, Tumors: wounds that do not heal. Similarities between tumor [120] G. Bergers, L.E. Benjamin, Tumorigenesis and the angiogenic switch, Nat. Rev.
stroma generation and wound healing, N. Engl. J. Med. 315 (1986) 1650– Cancer 8 (2008) 592–603.
1659. [121] Z. Wang, C. Dabrosin, X. Yin, M.M. Fuster, A. Arreola, W.K. Rathmell, D.
[90] A. Mantovani, Cancer: inflaming metastasis, Nature 457 (2009) 36–37. Generali, G.P. Nagaraju, B. El-Rayes, D. Ribatti, Y.C. Chen, K. Honoki, H. Fujii, A.
[91] S. Demaria, E. Pikarsky, M. Karin, L.M. Coussens, Y.C. Chen, E.M. El-Omar, G. G. Georgakilas, S. Nowsheen, A. Amedei, E. Niccolai, A. Amin, S.S. Ashraf, B.
Trinchieri, S.M. Dubinett, J.T. Mao, E. Szabo, A. Krieg, G.J. Weiner, B.A. Fox, G. Helferich, X. Yang, G. Guha, D. Bhakta, M.R. Ciriolo, K. Aquilano, S. Chen, D.
Coukos, E. Wang, R.T. Abraham, M. Carbone, M.T. Lotze, Cancer and Halicka, S.I. Mohammed, A.S. Azmi, A. Bilsland, W.N. Keith, L.D. Jensen, Broad
inflammation: promise for biologic therapy, J. Immunother. 33 (2010) 335– targeting of angiogenesis for cancer prevention and theraphy, Semin. Cancer
351. Biol. 35 (2015) S224–S243.
[92] A. Mantovani, P. Allavena, A. Sica, F. Balkwill, Cancer-related inflammation, [122] Y. Zhao, A.A. Adjei, Targeting angiogenesis in cancer therapy: moving beyond
Nature 454 (2008) 436–444. vascular endothelial growth factor, Oncologist 20 (2015) 660–673.
[93] B.B. Aggarwal, S. Shishodia, S.K. Sandur, M.K. Pandey, G. Sethi, Inflammation [123] A.E. El-Kenawi, A.B. El-Remessy, Angiogenesis inhibitors in cancer therapy:
and cancer: how hot is the link?, Biochem Pharmacol. 72 (2006) 1605–1621. mechanistic perspective on classification and treatment rationales, Br. J.
[94] S. Ohnishi, N. Ma, R. Thanan, S. Pinlaor, O. Hammam, M. Murata, S. Kawanishi, Pharmacol. 170 (2013) 712–729.
DNA damage in inflammation-related carcinogenesis and cancer stem cells, [124] L. Yang, J. Zhang, C. Wang, X. Qin, Q. Yu, Y. Zhou, J. Liu, Interaction between 8-
Oxid. Med. Cell. Longev. 2013 (2013) 387014. hydoxyquinoline ruthenium(II) complexes and basis fibroblast growth
[95] L.M. Coussens, L. Zitvogel, A.K. Palucka, Neutralizing tumor-promoting factors (bFGF): inhibiting angiogenesis and tumor growth through ERK and
chronic inflammation: a magic bullet?, Science 339 (2013) 286–291 AKT signalling pathways, Metallomics 6 (2014) 518–531.
[96] M. Schaefer, S. Werner, Cancer as an overhealing wound: an old hypothesis [125] D. Sun, Y. Liu, Q. Yu, Y. Zhou, R. Zhang, X. Chen, A. Hong, J. Liu, The effects of
revisited, Nat. Rev. Mol. Cell. Biol. 9 (2008) 628–638. luminescent ruthenium(II) polypyridyl functionalized selenium
[97] J.C. Roeser, S.D. Leach, F. McAllister, Emerging strategies for cancer nanoparticles on bFGF-induced angiogenesis and AKT/ERK signalling,
immunoprevention, Oncogene 34 (2015) 6029–6039. Biomaterials 34 (2013) 171–180.
[98] D.S. Chen, I. Mellman, Oncology meets immunology: the cancer-immunity [126] A. Weiss, X. Ding, J.R. van Beijnum, I. Wong, T.J. Wong, R.H. Berndsen, O.
cycle, Immunity 39 (2013) 1–10. Dormond, M. Dalliga, L. Shen, R.O. Schlingemann, R. Pili, C.M. Ho, P.J. Dyson,
[99] M.J. Smyth, S.F. Ngiow, A. Ribas, M.W.L. Teng, Combination cancer H. van den Bergh, A.W. Griffioen, P. Nowak-Sliwinska, Rapid optimization of
immunotherapies tailored to the tumour microenvironment, Nat. Rev. Clin. drug combination for the optimal angiostatic treatment of cancer,
Oncol. 13 (2016) 143–158. Angiogenesis 18 (2015) 233–244.
[100] I. Mellman, G. Coukos, G. Dranoff, Cancer immunotherapy comes of age, [127] Q. Wu, J. He, W. Mei, Z. Zhang, X. Wu, F. Sun, Arene ruthenium(II) complex, a
Nature 480 (2011) 480–489. potent inhibitor against proliferation, migration and invasion of breast cancer
[101] C. Yewale, D. Baradia, I. Vhora, S. Patil, A. Misra, Epidermal growth factor cells, reduces stress fibers, focal adhesions and invadopodia, Metallomics 6
receptor targeting in cancer: a review of trends and strategies, Biomaterials (2014) 2204–2212.
34 (2013) 8690–8707. [128] H.W. Smith, C.J. Marshall, Regulation of cell signalling by uPAR, Nat. Rev. Mol.
[102] J.R. Goffin, K. Zbuk, Epidermal growth factor receptor: pathway, therapies, Cell. Biol. 11 (2010) 23–36.
and pipeline, Clin. Ther. 35 (2013) 1282–1303. [129] J. Zhang, S.N. Hochwald, The role of FAK in tumor metabolism and therapy,
[103] W. Zheng, Q. Luo, Y. Lin, Y. Zhao, X. Wang, Z. Du, X. Hao, Y. Yu, S. Lu, L. Ji, X. Li, Pharmacol. Ther. 142 (2014) 154–163.
L. Yang, F. Wang, Complexation with organometallic ruthenium [130] C. Pelillo, H. Mollica, J.A. Eble, J. Grosche, L. Herzog, B. Codan, G. Sava, A.
pharmacophores enhances the ability of 4-anilinoquinazolines inducing Bergamo, Inhibition of adhesion, migration and of a5b1 integrin in the HCT-
apoptosis, Chem. Commun. 49 (2013) 10224–10226. 116 colorectal cancer cells treated with the ruthenium drug NAMI-A, J. Inorg.
[104] L. Ji, W. Zheng, Y. Lin, X. Wang, S. Lu, X. Hao, Q. Luo, X. Li, L. Yang, F. Wang, Biochem. 160 (2016) 225–235.
Novel ruthenium complexes ligated with 4-anilinoquinazoline derivatives: [131] I.E. Leon, P. Diez, S.B. Etcheverry, M. Fuentes, Deciphering the effect of an
synthesis, characterisation and preliminary evaluation of biological activity, oxovanadium(IV) complex with the flavonoid chrysin (VOChrys) on
Eur. J. Med. Chem. 77 (2014) 110–120. intracellular cell signalling pathways in an osteosarcoma cell line,
[105] J. Du, E. Zhang, Y. Zhao, W. Zheng, Y. Zhang, Y. Lin, Z. Wang, Q. Luo, K. Wu, F. Metallomics 8 (2016) 739–749.
Wang, Discovery of a dual-targeting organometallic ruthenium complex with [132] K. Traven, N. Eleftheriadis, S. Sersen, J. Kliun, J. Bezensek, B. Stanovnik, I. Turel,
high activity inducing early stage apoptosis of cancer cells, Metallomics 7 F.J. Dekker, Ruthenium complexes as inhibitors of 15-lipoxygenase-1,
(2015) 1573–1583. Polyhedron 101 (2015) 306–313.
[106] T. Chen, Y. Liu, W.J. Zheng, J. Liu, Y.S. Wong, Ruthenium polypyridyl [133] H.E. Claesson, On the biosynthesis and biological role of eoxins and 15-
complexes that induce mitochondria-mediated apoptosis in cancer cells, lipoxygenase-1 in airway inflammation and Hodgkin lymphoma,
Inorg. Chem. 49 (2010) 6366–6368. Prostaglandins Other Lipid Mediat. 89 (2009) 120–125.
[107] W. Cao, W. Zheng, T. Chen, Ruthenium polypyridyl complex inhibits growth [134] M.Y. Li, H.L. Yuan, F.W.S. Ko, B. Wu, X. Long, J. Du, J. Wu, C.S.H. Ng, I.Y.P. Wan,
and metastasis of breast cancer cells by suppressing FAK signalling with T.S.K. Mok, D.S.C. Hui, M.J. Underwood, G.G. Chen, Antineoplastic effects of 15
enhancement of TRAIL-induced apoptosis, Sci. Rep. 5 (2015) 9157. (S)-hydoxyeicosatetraenoic acid and 13-S-hydoxyoctadecadienoic acid in
[108] C. Zhao, X. Chen, D. Zang, X. Lan, S. Liao, C. Yang, P. Zhang, J. Wu, X. Li, N. Liu, Non-Small Cell Lung Cancer, Cancer 121 (2015) 3130–3145.
Y. Liao, H. Huang, S. Shi, L. Jiang, X. Liu, Z. He, Q.P. Dou, X. Wang, J. Liu, A novel [135] J. Chang, L. Jiang, Y. Wang, B. Yao, S. Yang, B. Zhang, M.Z. Zhang, 12/15
nickel complex works as a proteasomal deubiquitinase inhibitor for cancer lipoxygenase regulation of colorectal tumorigenesis is determined by the
therapy, Oncogene 35 (2016) 5916–5927. relative tumor levels of its metabolite 12-HETE and 13-HODE in animal
[109] S.M. Schmitt, M. Frezza, Q.P. Dou, New applications of old metal-binding models, Oncotarget 6 (2015) 2879–2888.
drugs in the treatment of human cancer, Front. Biosci. 4 (2012) 375–391. [136] K.H. Kang, T.Y. Ling, H.H. Liou, Y.K. Huang, M.J. Hour, H.C. Liou, W.M. Fu,
[110] D. Buac, S. Schmitt, G. Ventro, F.R. Kona, Q.P. Dou, Dithiocarbamate-based Enhancement role of host 12/15-lipoxygenase in melanoma progression, Eur.
coordination compounds as potent proteasome inhibitors in human cancer J. Cancer 49 (2013) 2747–2759.
cells, Mini Rev. Med. Chem. 12 (2012) 1193–1201. [137] B. Friedenson, The BRCA1/2 pathway prevents hematologic cancers in
[111] B. Levine, G. Kroemer, Autophagy in the pathogenesis of disease, Cell 132 addition to breast and ovarian cancers, BMC Cancer 7 (2007) 152–162.
(2008) 27–42. [138] M. Taron, R. Rosell, E. Felip, P. Mendez, J. Souglakos, M.S. Ronco, C. Queralt, J.
[112] N. Mizushima, Autophagy: process and function, Genes Dev. 21 (2007) 2861– Majo, J.M. Sanchez, J.J. Sanchez, J. Maestre, BRCA1 mRNA expression levels as
2873. an indicator of chemoresistance in lung cancer, Hum. Mol. Genet. 13 (2004)
[113] S.W. Ryter, K. Mizumura, A.M. Choi, The impact of autophagy on cell death 2443–2449.
modalities, Int. J. Cell. Biol. 2014 (2014) 502676. [139] C. Papadaki, M. Sfakianaki, G. Ioannidis, E. Lagoudaki, M. Trypaki, K.
[114] J. Yuan, Z. Lei, X. Wang, F. Zhu, D. Chen, Ruthenium complex K-WH0402 Tryfonidis, D. Mavroudis, E. Stathopoulos, V. Georgoulias, J. Souglakos,
induces hepatocellular carcinoma LM6 (HCCLM6) cell death by triggering the ERCC1 and BRCA1 mRNA expression levels in the primary tumour could
Beclin-1-dependent autophagy pathway, Metallomics 7 (2015) 896–907. predict the effectiveness of the second-line cisplatin-based chemotherapy in
[115] A. Apel, H. Zentgraf, M.W. Buechler, I. Herr, Autophagy-A double-edged pre-treated patients with metastatic non-small cell lung cancer, J. Thorac.
sword in oncology, Int. J. Cancer 125 (2009) 991–995. Oncol. 7 (2012) 663–671.
[116] M.D. Bareford, M.A. Park, A. Yacoub, H.A. Hamed, Y. Tang, N. Cruickshanks, P. [140] T. Nhukeaw, P. Temboot, K. Hansongnem, A. Ratanaphan, Cellular responses
Eulitt, N. Hubbard, G. Tye, M.E. Burow, P.B. Fisher, R.G. Moran, K.P. Nephew, S. of BRCA1-defective and triple-negative breast cancer cells and in vitro BRCA1
Grant, P. Dent, Sorafenib enhances pemetrexed cytotoxicity through an interactions induced by metallo-intercalator ruthenium(II) complexes
autophagy-dependent mechanism in cancer cells, Cancer Res. 71 (2011) containing chloro-substituted phenylazopyridine, BMC Cancer 14 (2014) 73.
4955–4967. [141] A. Atipairin, B. Canyuk, A. Ratanaphan, Cisplatin affects the conformation of
[117] C. Tan, S. Lai, S. Wu, S. Hu, L. Zhou, Y. Chen, M. Wang, Y. Zhu, W. Lian, W. Peng, apo form, not holo form, of BRCA1 RING finger domain and confers thermal
L. Ji, A. Xu, Nuclear permeable ruthenium(II) b-carboline complexes induce stability, Chem. Biodivers. 7 (2010) 1949–1967.
A. Bergamo et al. / Coordination Chemistry Reviews 360 (2018) 17–33 33

[142] A. Atipairin, B. Canyuk, A. Ratanaphan, The RING heterodimer BRCA1-BARD1 lactate dehydrogenase activity and cytotoxicity in gastric and colon cancer
is a ubiquitin ligase inactivated by the platinum-based anticancer drugs, cells, J. Inorg. Biochem. 163 (2016) 28–38.
Breast Cancer Res. Treat. 126 (2011) 203–209. [164] M.W.L. Teng, J.B. Swann, C.M. Koebel, R.D. Schreiber, M.J. Smyth, Immune-
[143] A. Ratanaphan, T. Nhukeaw, K. Hongthong, P.J. Dyson, Differential mediated dormancy: an equilibrium with cancer, J. Leukoc. Biol. 84 (2008)
cytotoxicity, cellular uptake, apoptosis and inhibition of BRCA1 expression 988–993.
of BRCA1-defective and sporadic breast cancer cells induced by an anticancer [165] R. Kim, M. Emi, K. Tanabe, Cancer immunoediting from immune surveillance
ruthenium(II)-arene compound, RAPTA-EA1, Anticancer Agents Med. Chem. to immune escape, Immunology 121 (2007) 1–14.
17 (2017) 212–220. [166] L. Yang, Y. Pang, H.L. Moses, TGF-beta and immune cells: an important
[144] W.K. Olson, A.A. Gorin, X.J. Lu, L.M. Hock, V.B. Zhurkin, DNA sequence- regulatory axis in the tumor microenvironment and progression, Trends
dependent deformability deduced from protein-DNA crystal complexes, Proc. Immunol. 31 (2010) 220–227.
Natl. Acad. Sci. U.S.A. 95 (1998) 11163–11168. [167] J.D. Shields, I.C. Kourtis, A.A. Tomei, J.M. Roberts, M.A. Swartz, Induction of
[145] A. Krueger, E. Protozanova, M.D. Frank-Kamenetskii, Sequence-dependent lymphoid-like stroma and immune escape by tumors that express the
base pair opening in DNA double helix, Biophys. J. 90 (2006) 3091–3099. chemokine CCL21, Science 328 (2010) 749–752.
[146] C.A. Davey, Exposure to metals can be therapeutic, Chimia 69 (2015) 125– [168] D. Mougiakakos, A. Choudhury, A. Lladser, R. Kiessling, C.C. Johansson,
130. Regulatory T cells in cancer, Adv. Cancer Res. 107 (2010) 57–117.
[147] C. Scolaro, A. Bergamo, L. Brescacin, R. Delfino, M. Cocchietto, G. Laurenczy, T. [169] S. Ostrand-Rosenberg, P. Sinha, Myeloid-derived suppressor cells: linking
J. Geldbach, G. Sava, P.J. Dyson, In vitro and in vivo evaluation of ruthenium inflammation and cancer, J. Immunol. 182 (2009) 4499–4506.
(II)-arene PTA complexes, J. Med. Chem. 48 (2005) 4161–4171. [170] A. Terenzi, C. Pirker, B.K. Keppler, W. Berger, Anticancer metal drugs and
[148] Z. Adhireksan, G.E. Davey, P. Campomanes, M. Groessl, C.M. Clavel, H. Yu, A.A. immunogenic cell death, J. Inorg. Biochem. 165 (2016) 71–79.
Nazarov, C.H. Yeo, W.H. Ang, P. Droege, U. Rothlisberger, P.J. Dyson, C.A. [171] P. Loke, J.P. Allison, PD-L1 and PD-L2 are differentially regulated by Th1 and
Davey, Ligand substitutions between ruthenium-cymene compounds can Th2 cells, Proc. Natl. Acad. Sci. U.S.A. 100 (2003) 5336–5341.
control protein versus DNA targeting and anticancer activity, Nat. Commun. 5 [172] M. Obeid, A. Tesniere, F. Ghiringhelli, G.M. Fimia, L. Apetoh, J.L. Perfettini, M.
(2014) 3462. Castedo, G. Mignot, T. Panaretakis, N. Casares, D. Metivier, N. Larochette, P.
[149] Z. Adhireksan, G. Palermo, T. Riedel, Z. Ma, R. Muhammad, U. Rothlisberger, P. van Endert, F. Ciccosanti, M. Piacentini, L. Zitvogel, G. Kroemer, Calreticulin
J. Dyson, C.A. Davey, Allosteric cross-talk in chromatin can mediate drug-drug exposure dictates the immunogenicity of cancer cell death, Nat. Med. 13
synergy, Nat. Commun. 8 (2017) 14860. (2007) 54–61.
[150] C. Licona, M.E. Spaety, A. Capuozzo, M. Ali, R. Santamaria, O. Armant, F. [173] R. Ramakrishnan, D. Assudani, S. Nagaraj, T. Hunter, H.I. Cho, S. Antonia, S.
Delalande, A. Van Dorsselaer, S. Cianferani, J. Spencer, M. Pfeffer, G. Mellitzer, Altiok, E. Celis, D.I. Gabrilovich, Chemotherapy enhances tumor cell
C. Gaiddon, A ruthenium anticancer compound interacts with histones and susceptibility to CTL-mediated killing during cancer immunotherapy in
impacts differently on epigenetic and death pathways compared to cisplatin, mice, J. Clin. Invest. 120 (2010) 1111–1124.
Oncotarget 8 (2017) 2568–2584. [174] M. Montani, G.V. Badillo Pazmay, A. Hysi, G. Lupidi, R. Pettinari, V. Gambini,
[151] O.H. Warburg, The Metabolism of Tumours: Investigations from the Kaiser M. Tilio, F. Marchetti, C. Pettinari, S. Ferraro, M. Iezzi, C. Marchini, A. Amici,
Wilhelm Institute for Biology, Berlin-Dahlem, Arnold Constable ed., London, The water soluble ruthenium(II) organometallic compound [Ru(p-cymene)
1930. (bis(3,5 dimethylpyrazol-1-yl)methane)Cl]Cl suppresses triple negative
[152] O. Warburg, On the origin of cancer cells, Science 123 (1956) 309–314. breast cancer growth by inhibiting tumor infiltration of regulatory T cells,
[153] O. Warburg, On respiratory impairment in cancer cells, Science 124 (1956) Pharmacol. Res. 107 (2016) 282–290.
269–270. [175] I. Khalaila, A. Bergamo, F. Bussy, G. Sava, P.J. Dyson, The role of cisplatin and
[154] R.G. Jones, C.B. Thompson, Tumor suppressors and cell metabolism: a recipe NAMI-A plasma-protein interactions in relation to combination therapy, Int.
for cancer growth, Genes Dev. 23 (2009) 537–548. J. Oncol. 29 (2006) 261–268.
[155] R.J. DeBerardinis, J.J. Lum, G. Hatzivassiliou, C.B. Thompson, The biology of [176] E. Alessio, G. Mestroni, G. Nardin, W.M. Attia, M. Calligaris, G. Sava, S. Zorzet,
cancer: metabolic reprogramming fuels cell growth and proliferation, Cell Cis- and trans-dihalotetrakis(dimethylsulfoxide)ruthenium(II) complexes
Metab. 7 (2008) 11–20. (RuX2(DMSO)4; X = Cl, Br): synthesis, structure, and antitumor activity,
[156] P.P. Hsu, D.M. Sabatini, Cancer cell metabolism: Warburg and beyond, Cell Inorg. Chem. 27 (1988) 4099–4106.
134 (2008) 703–707. [177] G. Sava, S. Pacor, A. Bergamo, M. Cocchietto, G. Mestroni, E. Alessio, Effects of
[157] A. Tomiyama, S. Serizawa, K. Tachibana, K. Sakurada, H. Samejima, Y. ruthenium complexes on experimental tumors: irrelevance of cytotoxicity
Kuchino, C. Kitanaka, Critical role for mitochondrial oxidative for metastasis inhibition, Chem. Biol. Interact. 95 (1995) 109–125.
phosphorylation in the activation of tumor suppressors Bax and Bak, J. [178] G. Sava, F. Frausin, M. Cocchietto, F. Vita, E. Podda, P. Spessotto, A. Furlani, V.
Natl. Cancer Inst. 98 (2006) 1462–1473. Scarcia, G. Zabucchi, Actin-dependent tumour cell adhesion after short-term
[158] R.L. Safirstein, Acute renal failure: from renal physiology to the renal exposure to the antimetastasis ruthenium complex NAMI-A, Eur. J. Cancer 40
transcriptome, Kidney Int. 66 (2004) S62–S66. (2004) 1383–1396.
[159] N. Shulga, R. Wilson-Smith, J.G. Pastorino, Hexokinase II detachment from the [179] M. Groessl, O. Zava, P.J. Dyson, Cellular uptake and subcellular distribution of
mitochondria potentiates cisplatin induced cytotoxicity through a caspase-2 ruthenium-based metallodrugs under clinical investigation versus cisplatin,
dependent mechanism, Cell Cycle 8 (2009) 3355–3364. Metallomics 3 (2011) 591–599.
[160] T. Egawa-Takata, H. Endo, M. Fujita, Y. Ueda, T. Miyatake, H. Okuyama, K. [180] J.B. Aitken, S. Antony, C.M. Weekley, B. Lai, L. Spiccia, H.H. Harris, Distinct
Yoshino, S. Kamiura, T. Enomoto, T. Kimura, M. Inoue, Early reduction of cellular fates for KP1019 and NAMI-A determined by X-ray fluorescence
glucose uptake after cisplatin treatment is a marker of cisplatin sensitivity in imaging of single cells, Metallomics 4 (2012) 1051–1056.
ovarian cancer, Cancer Sci. 101 (2010) 2171–2178. [181] M. Shimada, H. Itamochi, J. Kigawa, Nedaplatin: a cisplatin derivative in
[161] R.L. Elstrom, D.E. Bauer, M. Buzzai, R. Karnauskas, M.H. Harris, D.R. Plas, H. cancer chemotherapy, Cancer Manag. Res. 5 (2013) 67–76.
Zhuang, R.M. Cinalli, A. Alavi, C.M. Rudin, C.B. Thompson, Akt stimulates [182] A. Doucette, K.N. Hassel, D.C. Crans, Selective speciation improves efficacy
aerobic glycolysis in cancer cells, Cancer Res. 64 (2004) 3892–3899. and lowers toxicity of platinum anticancer and vanadium antidiabetic drugs,
[162] S. Wang, J. Xie, J. Li, F. Liu, X. Wu, Z. Wang, Cisplatin suppresses the growth J. Inorg. Biochem. 165 (2016) 56–70.
and proliferation of breast and cervical cancer cell lines by inhibiting integrin [183] M. Selman, C. Rousso, A. Bergeron, H.H. Son, R. Krishnan, N.A. El-Sayes, O.
b5-mediated glycolysis, Am. J. Cancer Res. 6 (2016) 1108–1117. Varette, A. Chen, F. Le Boeuf, F. Tzelepis, J.C. Bell, D.C. Crans, J.S. Diallo, Multi-
[163] H. Rico Bautista, R.O. Saavedra Diaz, L.Q. Shen, C. Orvain, C. Gaiddon, R. Le modal potentiation of oncolytic virotherapy by vanadium compounds, Mol.
Lagadec, A.D. Ryabov, Impact of cyclometalated ruthenium(II) complexes on Ther. 26 (2018) 56–69.

You might also like