Vous êtes sur la page 1sur 15

European Journal of Pharmacology ()

1 Contents lists available at ScienceDirect


2
3
4 European Journal of Pharmacology
5
6
journal homepage: www.elsevier.com/locate/ejphar
7
8
9
Review
10
11
12
Cisplatin in cancer therapy: Molecular mechanisms of action
13
Q1
14 Shaloam Dasari, Paul Bernard Tchounwou n
15 Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science,
16 Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA
17
18
19 art ic l e i nf o a b s t r a c t
20
21 Article history: Cisplatin, cisplatinum, or cis-diamminedichloroplatinum (II), is a well-known chemotherapeutic drug.
22 Received 27 March 2014 It has been used for treatment of numerous human cancers including bladder, head and neck, lung,
Received in revised form ovarian, and testicular cancers. It is effective against various types of cancers, including carcinomas, germ
23
13 July 2014
24 cell tumors, lymphomas, and sarcomas. Its mode of action has been linked to its ability to crosslink with
Accepted 14 July 2014
the purine bases on the DNA; interfering with DNA repair mechanisms, causing DNA damage, and
25
subsequently inducing apoptosis in cancer cells. However, because of drug resistance and numerous
26 Keywords: undesirable side effects such as severe kidney problems, allergic reactions, decrease immunity to
27 Cisplatin infections, gastrointestinal disorders, hemorrhage, and hearing loss especially in younger patients, other
28 Platinum-based drugs
platinum-containing anti-cancer drugs such as carboplatin, oxaliplatin and others, have also been used.
29 Mechanisms of action
Furthermore, combination therapies of cisplatin with other drugs have been highly considered to
Cancer treatment
30 overcome drug-resistance and reduce toxicity. This comprehensive review highlights the physicochem-
31 ical properties of cisplatin and related platinum-based drugs, and discusses its uses (either alone or in
32 combination with other drugs) for the treatment of various human cancers. A special attention is paid to
33 its molecular mechanisms of action, and its undesirable side effects.
34 & 2014 Published by Elsevier B.V.
35
36
37
38 Contents
39
40 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
41 2. Cisplatin and other platinum-containing drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
42 3. Uses of cisplatin for cancer treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1. Cisplatin and lung cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
43
3.2. Cisplatin and ovarian cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
44
3.3. Cisplatin and carcinoma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
45
3.4. Cisplatin and breast cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
46 3.5. Cisplatin and brain cancer. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
47 4. Combination therapy of cisplatin with other cancer drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
48 4.1. Cisplatin and paclitaxel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
49 4.2. Cisplatin and tegafur-uracil (UFT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
50 4.3. Cisplatin and doxorubicin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
51 4.4. Cisplatin and gemcitabine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
52 4.5. Cisplatin and vitamin D. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
53 4.6. Other possible drug combinations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
5. Molecular mechanisms of cisplatin pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
54
5.1. Cisplatin-induced oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
55
5.2. Cisplatin modulation of calcium signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
56 5.3. Cisplatin-induced cell apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
57 5.4. Cisplatin and protein kinase C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
58 5.5. Cisplatin and mitogen-activated protein kinase (MAPK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
59 5.6. Cisplatin and jun amino-terminal kinase (JNK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
60
61
n
62 Corresponding author. Tel.: 1 601 979 0777.
63 E-mail address: paul.b.tchounwou@jsums.edu (P. Bernard Tchounwou).
64 http://dx.doi.org/10.1016/j.ejphar.2014.07.025
65 0014-2999/& 2014 Published by Elsevier B.V.
66

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
2 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 5.7. Cisplatin and p38 mitogen-activated protein kinase (MAPK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8


2 5.8. Cisplatin and AKT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3 5.9. Cisplatin and signaling for DNA damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4 5.10. p53 and DNA damage response to cisplatin treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.11. Cyclobutanedicarboxylate (c-abl) and DNA damage response to cisplatin treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5
5.12. Cisplatin modulation of gene expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6
5.13. Computational studies of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
7 6. Toxicological effects of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8 6.1. Hepatotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
9 6.2. Cardiotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
10 6.3. Nephrotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
11 6.4. Other organ toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
12 7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
13 Uncited references . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
14 Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
15
16
17
18
19
1. Introduction
20 drugs) for the treatment of various human cancers, to examine
21 its molecular mechanisms of action, and to discuss it potential
Cisplatin (CAS no. 15663-27-1, MF-Cl2H6N2Pt; NCF-119875),
22 side effects.
cisplatinum, also called cis-diamminedichloroplatinum(II), is a
23
metallic (platinum) coordination compound with a square planar
24
geometry. It is a white or deep yellow to yelloworange crystalline
25
powder at room temperature. It is slightly soluble in water
26 2. Cisplatin and other platinum-containing drugs
and soluble in dimethylprimanide and N,N-dimethylformamide.
27
Cisplatin is stable under normal temperatures and pressures, but
28 Since the early seminal work in the preclinical and clinical
may transform slowly over time to the trans-isomer (IARC, 1981;
29 development of cisplatin, several thousand analogs have been
Akron, 2009). Cisplatin has a molecular weight of 301.1 g/mol, a
30 synthesized and tested for properties that would enhance its
density of 3.74 g/cm3, a melting point of 270 1C, a log Kow of  2.19
31 therapeutic index. About 13 of these analogs have been evaluated
and a water solubility of 2.53 g/L at 25 1C (HSDB 2009).
32 in clinical trials, but only one (carboplatin) has provided denite
Cisplatin was rst synthesized by M. Peyrone in 1844 and its
33 advantage over cisplatin and achieved worldwide approval. Nine
chemical structure was rst elucidated by Alfred Werner in 1893.
34 platinum analogs are currently in clinical trials around the world
However, the compound did not gain scientic investigations until
35 ormaplatin (tetraplatin), oxaliplatin, DWA2114R, enloplatin, loba-
the 1960s when the initial observations of Rosenberg et al. (1965)
36 platin, CI-973 (NK-121), 254-S, JM-216, and liposome-entrapped
at Michigan State University pointed out that certain electrolysis
37 cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum
products of platinum mesh electrodes were capable of inhibiting
38 (II) (LNDDP)] (Weiss and Christian, 1993). Fig. 1 presents the
cell division in Escherichia coli created much interest in the
39 chemical structures of cisplatin and four of its analogs including
possible use of these products in cancer chemotherapy. Since the
40 carboplatin, oxaliplatin, ormaplatin and enloplatin.
identication of cis-dichlorodiammineplatinum (II) (cisplatin, r) as
41 From the molecular perspective, cisplatin represents a perfect
the agent responsible for this activity, considerable interest has
42 example of how a small alteration in chemical structure can signi-
been generated in the use of coordination complexes of platinum,
43 cantly affect biological activity in target cell (Goodsell, 2006). As
palladium, and other noble metals in the treatment of cancer.
44 illustrated in Fig. 2, cisplatin, carboplatin and oxaliplatin are composed
Cisplatin has been especially interesting since it has shown
45 of doubly charged platinum ion surrounded by four ligands, with the
anticancer activity in a variety of tumors including cancers of the
46 amine ligands on the left forming stronger interactions with the
ovaries, testes, and solid tumors of the head and neck. It was
47 platinum ion, and the chloride ligands or carboxylate compounds on
discovered to have cytotoxic properties in the 1960s, and by the
48 the right forming leaving groups allowing the platinum ion to form
end of the 1970s it had earned a place as the key ingredient in the
49 bonds with DNA bases (Goodsell, 2006).
systemic treatment of germ cell cancers. Among many chemother-
50 Carboplatin or Cis diammine (1,1-cyclobutanecarboxylato) pla-
apy drugs that are widely used for cancer, cisplatin is one of the
51 tinum (II) is a chemotherapeutic drug used for cancers of ovaries,
most compelling ones. It was the rst FDA-approved platinum
52 lung, head and neck. In terms of its structure, carboplatin differs
compound for cancer treatment in 1978 (Kelland, 2007). This has
53 from cisplatin in that it has a bidentate dicarboxylate (CBDCA)
led to interest in platinum (II)and other metalcontaining com-
54 ligand in place of the two chloride ligands, which are the leaving
pounds as potential anticancer drugs (Frezza et al., 2010).
55 groups in cisplatin (Figs. 1 and 2). It exhibits lower reactivity
Cisplatin is clinically proven to combat different types of cancers
56 and slower DNA binding kinetics, although it forms the same
including sarcomas, cancers of soft tissue, bones, muscles, and blood
57 reaction products in vitro at equivalent doses with cisplatin. Unlike
58 vessels. Although such cancers have recently received better prog- cisplatin, carboplatin may be susceptible to alternative mechan-
59 nosis and therefore have become less life threatening (Desoize and isms. Some studies show that cisplatin and carboplatin cause
60 Madoulet, 2002), signicant challenges remain with regard to their different morphological changes in MCF-7 cell lines while exerting
61 cure. Also, because of drug resistance and considerable side effects, their cytotoxic behavior (Natarajan et al., 1999). The dimin-
62 combination therapy of cisplatin with other cancer drugs has been ished reactivity limits proteincarboplatin complexes, which are
63 applied as novel therapeutic strategies for many human cancers. In excreted. The lower excretion rate of carboplatin means that more
64 this research, we aim to provide a comprehensive review of the is retained in the body, and hence its effects are longer lasting
65 physicochemical properties of cisplatin and related platinum-based (a retention half-life of 30 h for carboplatin, compared to 1.53.6 h
66 drugs, to discuss its uses (either alone or in combination with other in the case of cisplatin).

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 3

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28 Fig. 1. Chemical structures of selected platinum drugs: A-cisplatin; B-carboplatin; C-oxaliplatin; D-ormaplatin; E-enloplatin (Weiss and Christian, 1993).
29
30
31 Relative to cisplatin, the greatest benet of carboplatin is its
32 reduced side effects, particularly the elimination of nephrotoxic
33 effects. The main drawback of carboplatin is its myelo suppressive
34 effect which causes the blood cell and platelet output of bone
35 marrow in the body to decrease quite dramatically, sometimes as
36 low as 10% of its usual production levels (Canetta et al., 1985).
37 Carboplatin is less potent than cisplatin; depending on the type
38 of cancer, carboplatin may only be 1/8 to 1/45 as effective. The
39 clinical standard of dosage of carboplatin is usually a 4:1 ratio
40 compared to cisplatin; that is, for a dose that usually requires a
41 particular dose of cisplatin, four times more carboplatin is needed
42 to achieve the same effectiveness. The stable property of carbo-
43 platin is a mixed blessing; once uptake of the drug occurs, its
44 retention half-life is considerably longer than cisplatin, but it is
45 also this inertness that causes carboplatin to go right through the
46 human body, and up to 90% of the carboplatin given can be
47 recovered in urine (Go and Adjei, 1999).
48 In order to overcome cisplatin resistance, other platinum (Pt)
49 compounds have been developed and, in the last ten years,
50 Pt-derivatives with reporting activity have also been synthesized.
51 The rst generation of reporting Pt-compounds was based on
52 linking a uorescent molecule (e.g. cyanine) to cisplatin, but more
53 recent studies have focused on strategies to synthesize intrinsi-
54 cally uorescent derivatives. Accordingly, bile acid platinum com-
55 pounds have shown uorescence intensity that is stable at room
56 temperature for a long time; this uorescence is maintained after
57 binding to oligonucleotides or DNA. Because of this, the binding
58 mode of these compounds to DNA can be easily analyzed both by
59 ow injection and uorescence techniques, showing that although
60 these compounds target the nuclei, they form adduct with the
61 DNA that are different from those due to cisplatin. In line with this,
62 Fig. 2. Computational molecular structures of cisplatin, carboplatin and oxalipla- these bile acid derivatives have shown increased cytotoxicity and
63 tion. These platinum compounds are composed of doubly charged platinum ion ability to overcome resistance as compared to cisplatin in several
surrounded by four ligands; with the amine ligands on the left forming stronger
64 interactions with the platinum ion, and the chloride ligands or carboxylate
cell lines. Moreover, in contrast to cisplatin, the activity of these
65 compounds on the right forming leaving groups allowing the platinum ion to form compounds does not seem to be restricted to cycling cells but they
66 bonds with DNA bases (Goodsell, 2006). also seem to kill resting cells (Rodriguez-Fernandez et al., 2009)

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
4 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 3. Uses of cisplatin for cancer treatment survival rate of approximately 50% has not changed over the last
2 decades. Cisplatin alone is not an effective drug in treating the
3 3.1. Cisplatin and lung cancer disease. A randomized comparison of cisplatin alone or in combi-
4 nation with methotrexate, vinblastine, doxorubicin, and or gemci-
5 Lung cancer remains one of the most common types of fatal tabine in patients with metastatic urothelial carcinoma has been
6 malignancies (Youlden et al., 2008). Small cell lung cancers (SCLCs) reported (Scher, 1992; Matsuki et al., 2013).
7 represent 15% of all lung cancers (Chen et al., 2009). At present,
8 platinum based treatments are key drugs for SCLC (Abrams et al., 3.4. Cisplatin and breast cancer
9 2003). Cisplatin and carboplatin are two of the most common
10 types of platinum based treatments used in SCLC chemotherapy Breast cancer is one of the leading causes of women mortality
11 (Go and Adjei, 1999). In clinical trials, cisplatin is often selected worldwide. Chemotherapy is the only option for treating the
12 due to its strong antitumor activity, but its adverse effects include malignant breast cancer and condition for increases the lifespan
13 renal toxicity (Iwasaki et al., 2005), nausea and vomiting (Kosmas of the patient (Decatris et al., 2004). Chemotherapeutic agents
14 et al., 2001). Therefore, to avoid renal toxicity, urine volumes have been developed to counter the continuing breast cancer
15 should be monitored and large-dose infusion is mandatory in problem. However, most chemotherapeutic drugs effectively tar-
16 cisplatin based chemotherapy. In clinical practice, carboplatin has get rapidly dividing cells causing damage and are thus referred to
17 been considered to be a substitute for cisplatin without any as cytotoxic drugs. Cisplatin is an important chemotherapeutic
18 apparent loss of therapeutic efcacy since aggressive hydration is agent used widely or the treatment of a variety of malignancies,
19 often problematic. including breast, testicular, ovarian, cervical, prostate, head and
20 The standard of care for localized non-small-cell lung cancer neck, bladder, lung and refractory non-Hodgkin's lymphomas
21 (NSCLC) is surgery followed by, in case of stage II and III disease, (Tsimberidou et al., 2009; Dhar et al., 2011). The cytotoxic effect
22 adjuvant cisplatin-based chemotherapy. The Lung Adjuvant is likely a result of inhibition of replication by cisplatin-DNA
23 Cisplatin Evaluation program, a pooled analysis of the ve largest adducts and induction of apoptosis (Siddik, 2003).
24 trials, recently showed an absolute 5-year survival benet of 5.3%
25 with adjuvant chemotherapy as well as the NSCLC meta-analysis 3.5. Cisplatin and brain cancer
26 (Pignon et al., 2008).
27 CD133, a surface glycoprotein linked to organ-specic stem Glioblastomamultiforme (GBM) is the most common primary
28 cells, has been described as a marker of cancer-initiating cells in malignant brain tumor, and with rare exception, is invariably fatal
29 different tumor types. It has also been reported that a CD133 , (Stupp et al., 2005). The current standard of care for patients
30 epithelial-specic antigen-positive (CD133 ESA ) population is with GBMs consists of surgery and radiotherapy in combination
31 increased in primary non-small cell lung cancer (NSCLC) compared with temozolomide, followed by repetitive cycles of temozolo-
32 with normal lung tissue (Bertolini et al., 2009). mide (Stupp et al., 2009). Although the survival advantage of
33 this combined treatment regimen was still evident at 5 years, the
34 3.2. Cisplatin and ovarian cancer increase in overall median survival was only for 2.5 months.
35 Cisplatin therapy is also used for recurrent childhood brain
36 Ovarian cancer has the highest mortality among gynecologic tumors(Khan et al., 1982),as well as in other cancers such as
37 cancers. Most patients with ovarian cancer are diagnosed at late gastric cancer (Koizumi et al., 2008), anal cancer (Ajani et al.,
38 stages due to lack of effective screening strategies and specic 2008), and leukemia (Previati et al., 2006).
39 symptoms associated with early-stage disease. Conventional treat-
40 ment for late stages of ovarian cancers is surgical excision followed
41 by platinum/taxane combination chemotherapy. Although this 4. Combination therapy of cisplatin with other cancer drugs
42 treatment regime is effective as the rst-line treatment, recur-
43 rence occurs in up to 75% of ovarian cancer patients. Patients Cisplatin combination chemotherapy is the basis of treatment
44 with recurrent ovarian cancer ultimately develop resistance to of many cancers. Platinum responsiveness is high primarily
45 chemotherapy and eventually succumb to the disease (Agarwal but many cancer patients will ultimately relapse with cisplatin-
46 and Kaye, 2003). About 90% of ovarian cancers arise originally resistant disease. Hence, drug resistance has been observed in
47 from ovaries with an unknown reason, while the remainder has many patients who have relapsed from cisplatin treatment. The
48 hereditary background, or are associated with breast and colon proposed mechanisms of cisplatin resistance include changes in
49 cancers (Lynch et al., 2009). Cisplatin derivatives are used as the cellular uptake and efux of cisplatin, increased biotransformation
50 mainline treatment of ovarian cancer, despite their severe side and detoxication in the liver, and increase in DNA repair and anti-
51 effects and development of resistance. Cisplatin is used in combi- apoptotic mechanisms (Gottesman et al., 2002). To overcome
52 nation with other chemical agents or compounds to treat ovarian resistance, cisplatin is commonly used in combination with some
53 cancer in both the resistant and sensitive cell lines. For example other drugs in treating ovarian cancer, biliary tract cancer, lung
54 cisplatin is used along with honey venom (Alizadehnohi et al., cancer (diffuse malignant pleural mesothelioma), gastric cancer,
55 2012), withaferin (Kakar et al., 2012), trichostatin A or 5-aza-20 - carcinoma of salivary gland origin, breast, colon, lung, prostate,
56 deoxycytidine (Meng et al., 2013), overcoming chemotherapy melanoma and pancreatic cancer cell lines, squamous cell carci-
57 resistance of ovarian cancer cells by liposomal cisplatin (Koch noma of male genitial tract, urothelial bladder cancer, and cervical
58 et al., 2013) cancer. Table 1 presents a synopsis of cisplatin combination
59 therapy with other cancer drugs and targeted cancers.
60 3.3. Cisplatin and carcinoma
61 4.1. Cisplatin and paclitaxel
62 Head and neck squamous cell carcinoma (HNSCC) is a common
63 malignant disease with more than 600,000 new cases registered Paclitaxel is a mitotic agent that binds preferentially to micro-
64 worldwide every year (Parkin et al., 2005). Despite improved tubules (Parness et al., 1981) and the resulting stabilization
65 treatment options, including surgery, radiation and chemotherapy, of microtubules inhibits the reorganization of the microtubule
66 HNSCC is associated with a high mortality rate. The overall 5-year network. Paclitaxel has been demonstrated to be active against

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 5

1 Table 1
2 Combination therapy of cisplatin and other cancer drugs.
3
Combination drug(s) Cancer type Reference(s)
4
5 Paclitaxel Ovarian carcinoma, breast carcinoma, lung carcinoma, Sarosy, Kohn et al. (1992),McGuire et al. (1989),
6 melanoma, head and neck carcinoma Einzig et al. (1992),Holmes et al. (1991),
7 Murphy et al. (1993),
Legha et al. (1990), Einzig et al. (1991), Forastiere (1994)
8 Paclitaxel and 5-FU Gastric and esophagogastric adenocarcinoma Kim et al. (1999)
9 UFT Non small lung carcinoma Ichinose et al. (2000)
10 Doxorubicin Diffuse malignant pleural mesothelioma Ardizzoni et al. (1991)
11 Cyclophosphamide and doxorubicin Salivary gland advanced carcinoma Dreyfuss et al. (1987)
Gemcitabine Biliary cancer Valle et al. (2010)
12
Osthole Lung cancer Xu et al. (2013)
13 Honeybee venom Ovarian cancer Alizadehnohi et al. (2012)
14 Anvirzel Breast, colon, lung, prostate, melanoma and pancreatic Apostolou et al. (2013)
15 cancer
16 Bevacizumab Non small lung carcinoma Du et al. (2013)
Vinbalstine and bleomycin Metastatic granulosa cell tumors in ovary Colombo et al. (1986)
17 Methotrexate and bleomycin Advanced squamous cell carcinoma of the male Dexeus et al. (1991)
18 genital tract
19 Everolimus Urothelial bladder cancer Pinto-Leite, et al. (2013)
20 Fluorouracil, doxorubicin and Salivary gland carcinoma Dimery et al. (1990)
cyclophosphamide
21
Metformin Lung adenocarcinoma Lin et al. (2013)
22 Oxaliplatin, quercetin and thymoquinone Ovarian cancer Nessa et al. (2011)
23 Olaparib PTEN decient lung cancer Minami et al. (2013)
24 Tetra arsenic oxide Cervical cancer Byun et al. (2013)
25 Vindesine Non small lung carcinoma Le et al. (1994)
-galactosyl-pyrrolidinyldiazeniumdiolate Glial cells of brain Human cervical cancer Deng et al. (2013)
26 Gliosarcoma cell lines
27
28
29 previously treated ovarian carcinoma (Sarosy et al., 1992; McGuire 4.5. Cisplatin and vitamin D
30 et al., 1989; Einzig et al., 1992), breast carcinoma (Holmes et al.,
31 1991), lung carcinoma (Murphy et al., 1993), and melanoma (Legha Vitamin D and its analogs regulate gene expression by binding
32 et al., 1990; Einzig et al., 1991) as well as for head and neck to specic vitamin D receptors (VDR). Upon ligand activation and
33 carcinoma (Forastiere, 1994). The combination chemotherapy with dimerization with retinoid X receptor (RXR), VDRRXR hetero-
34 paclitaxel, cisplatin and uorouracil is an active and tolerable as dimers bind specic nucleotide sequences, vitamin D response
35 rst-line and second line therapies in Chinese patients with elements (VDREs), in target genes to activate or repress their
36 advanced gastric and esophagogastric junction adenocarcinoma expression (Baniahmad and Tsai, 1993). A number of vitamin D
37 which showed a better tolerance has also been reported (Kim et target genes have been identied in several tumor cell types: p21,
38 al., 1999) E-cadherin, c-Jun N-terminal kinase (JNK), c-Myc oncogene,
39 insulin-like transforming growth factor family and their receptors
40 (Saramaki et al., 2006; Nagpal et al., 2005).
41 4.2. Cisplatin and tegafur-uracil (UFT) Cisplatin is used along with vitamin D in treating squamous cell
42 carcinoma (Light et al., 1997) and colon cancer (Milczarek et al.,
43 UFT is an oral anticancer agent comprised of tegafur and uracil 2013). Published research has shown that pre-treatment for 72 h
44 in a 1:4 xed molar ratio and is absorbed very well from the small of human promyelocytic leukemia (HL-60) cells with calcitriol or
45 intestine (Fujii et al., 1979). Combination chemotherapy comprised its new analogs signicantly potentiated their sensitivity to the
46 of oral UFT (a combination of tegafur and uracil) and cisplatin was antiproliferative effect in vitro of cisplatin, doxorubicin or genis-
47 shown to be an effective regimen for the treatment of advanced tein. Moreover, for all cytotoxic agents tested a synergistic anti-
48 non-small cell lung carcinoma (Ichinose et al., 2000). proliferative effect was observed. This effect was expressed as a
49 signicant decrease of the ID50 (inhibitory dose 50%) values for
50 4.3. Cisplatin and doxorubicin each cytotoxic agent applied after pretreatment of HL-60 cells
51 with calcitriol or its analogs in comparison with the effect of
52 The combination of doxorubicin and cisplatin is effective and cytotoxic agent applied alone (Siwinska et al., 2001).
53 well tolerated. It might be considered for palliation of symptomatic
54 patients with diffuse malignant pleural mesothelioma DMPM
55 (Ardizzoni et al., 1991). Cyclophosphamide, doxorubicin, and cispla- 4.6. Other possible drug combinations
56 tin combination chemotherapy for advanced carcinomas of salivary
57 gland origin showed encouraging results (Dreyfuss et al., 1987). Cisplatin is also used in combination with natural compounds
58 like osthole in lung cancer cell lines (Xu et al., 2013), honey bee
59 venom in ovarian cancer cells (Alizadehnohi et al., 2012), anvirzel
60 4.4. Cisplatin and gemcitabine in breast, colon, lung, prostate, melanoma and pancreatic cancer
61 cell lines (Apostolou et al., 2013), bevacizumab in non-small cell
62 As compared with gemcitabine alone, cisplatin plus gemcita- lung cancer mediated malignant pleural effusion (Du et al., 2013),
63 bine was associated with a signicant survival advantage without vinblastine and bleomycin in metastatic granulosa cell tumor of
64 the addition of substantial toxicity. Cisplatin plus gemcitabine is an the ovary (Colombo et al., 1986), methotrexate, bleomycin and
65 appropriate option for the treatment of patients with advanced cisplatin for advanced squamous cell carcinoma of the male genital
66 biliary cancer (Valle et al., 2010). tract (Dexeus et al., 1991).

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
6 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 Everolimus combined with cisplatin has a potential role in cancer is due to adaptive activation of Nrf2 may contribute to the
2 treatment of urothelial bladder cancer (Pinto-Leite et al., 2013), development of acquired drug-resistance (Chian et al., 2014). It is
3 Fluorouracil, doxorubicin, cyclophosphamide, and cisplatin com- also indicated that AQP5 is associated with drug resistance of
4 bination chemotherapy has been used in advanced or recurrent colon cancer (Shi et al., 2014). In chondrosarcoma cells over-
5 salivary gland carcinoma (Dimery et al., 1990). Metformin has been expression of EGFR contributed to cisplatin resistance (Song et
6 shown to enhance cisplatin cytotoxicity by suppressing Stat3 al., 2014). Due to loss of homeodomain-interacting protein kinase-
7 activity independently of the LKB1-AMPK pathway (Lin et al., 2 (HIPK2) contributes to cell proliferation and tumorigenesis in
8 2013). bladder cancer (Lin et al., 2014).
9 Synergistic interactions have been reported for -galacto-
10 syl-pyrrolidinyldiazeniumdiolate and cisplatin combination
11 chemotherapy against glial cells of brain, human cervical and
12 gliosarcoma cell lines (Deng et al., 2013). Cisplatin and oxaliplatin 5. Molecular mechanisms of cisplatin pharmacology
13 in combination with quercetin and thymoquinone in human
14 ovarian tumour models is the best combination to treat ovarian Several membrane transporters of platinum compounds analo-
15 cancer (Nessa et al., 2011). Olaparib in combination with cisplatin gous to MDR1 including the efux ATPases (MRPs, ATP7A/B), and
16 has a synergistic effect on PTEN-Decient lung cancer Cells the solute carrier importers (CTR1, the SLCs, AQP2, and AQP9),
17 (Minami et al., 2013). Tetraarsenic oxide and cisplatin induce have been reported. MDR1 is an ATP-binding cassette transporter
18 apoptotic synergism in cervical cancer (Byun et al., 2013). Vinde- known for years as P-glycoprotein (Shen et al , 2012; Johnson et al.,
19 sine and cisplatin combination chemotherapy compared with 1996). The uptake of cisplatin is mediated by the copper trans-
20 vindesine as a single agent in the management of non-small cell porter Ctr1 in yeast and mammals (Ishida et al., 2002). It has been
21 lung cancer seemed to be more effective (Le et al., 1994). further conrmed in human cells that cisplatin triggers rapid
22 In ovarian carcinoma treatment, the combination of cisplatin degradation of the copper membrane transporter CTR1, with
23 and paclitaxel produced a more encouraging response rate (73%) diminished inux of cisplatin, resulting in resistance to the drug
24 (McGuire et al., 1996). A single institutional Phase II trial using (Lin et al., 2002; Holzer et al., 2006). Genetic knockout of CTR1
25 cisplatin and oral UFT administration in place of protracted results in cellular resistance to cisplatin in vivo. Cells with
26 intravenous injection of 5-FU yielded a 35% response rate and a increased CTR1 expression exhibit increased platinum accumula-
27 median survival time of 11 months in 31 patients with advanced tion and, in most instances, increased sensitivity to cisplatin.
28 Q3 NSCLC (Ichinose, Takanashi et al., 1995). The addition of cisplatin to TMEM205, a membrane protein has been associated with cellular
29 cyclophosphamide and doxorubicin (CAP) has been recently asso- resistance to cisplatin was identied. Analysis of TMEM205
30 ciated with overall response rates of 5700% and complete expression proles in normal human tissues indicates a differen-
31 response rates of 2675% in small series of patients (Creagan et tial expression pattern with higher expression levels in the liver,
32 al., 1983; Alberts et al., 1981; Eisenberger, 1985). Cisplatin is known pancreas, and adrenal glands. So, overexpression of TMEM205 in
33 to have an additive or synergistic effect in combination with CP-r cells may play a role in cellular resistance to platinum and
34 gemcitabine in a number of different tumor types (Crino et al., would also be valuable as a biomarker or target in cancer
35 1997; von der et al., 2005; Hitt et al., 1998). chemotherapy (Shen et al., 2012). Glucose Transporter 1 (Glut1)
36 Photodynamic therapy (PDT), which is the administration of a is not proposed to directly transport cisplatin, the mislocalization
37 photosensitiser followed by visible light activation, is a promising of the transporter exacerbates the cisplatin resistance phenotype
38 route to avoid damage to healthy cells and the surrounding tissue. (Shen et al., 2012).
39 Transition metal complexes as photochemotherapeutic agents are Cisplatin becomes activated once it enters the cell. In the
40 an attractive option for further development in the eld of cytoplasm the chloride atoms on cisplatin are displaced by water
41 photoactivated chemotherapy (PACT). These complexes exhibit molecules. This hydrolyzed product is a potent electrophile that
42 different numbers and types of excited states which are easily can react with any nucleophile, including the sulfhydryl groups on
43 accessible upon light irradiation, subsequently giving rise to the proteins and nitrogen donor atoms on nucleic acids. Cisplatin
44 formation of various photoproducts that can enable a distinct binds to the N7 reactive center on purine residues and as such can
45 mode of action. Platinumdiazido complexes are promising can- cause deoxyribonucleic acid (DNA) damage in cancer cells, block-
46 didates for PACT due to the low cytotoxicity when irradiated with ing cell division and resulting in apoptotic cell death. The 1,2-
47 visible light (Shaili, 2014). intrastrand cross-links of purine bases with cisplatin are the most
48 Cisplatin resistance is correlated with autophagy induction in notable among the changes in DNA. These include the 1,2-
49 a panel of ovarian cancer cells (Wang and Wu, 2014). Previous intrastrand d(GpG) adducts 1,2-intrastrand d(ApG) adducts repre-
50 studies demonstrated that A549/CDDP cells acquired an epithelial- senting about 90% and 10% of adducts, respectively. 1,3-intrastrand
51 mesenchymal transition (EMT) phenotype, with morphological d(GpXpG) adducts and other adducts such as inter-strand cross-
52 changes including acquisition of a spindle-like broblastic pheno- links and nonfunctional adducts have been reported to contribute
53 type, downregulation of E-cadherin, upregulation of mesenchymal to cisplatin's toxicity. Hence, published research from many
54 markers (vimentin, Snail and Slug), and increased capability of laboratories has implicated DNA as a critical target for cisplatin
55 invasion and migration in case of human lung cancer cell lines. cytotoxicity, the most revealing evidence being the hypersensitiv-
56 (Demonstrated that A549/CDDP cells acquired an epithelial- ity to cisplatin by both prokaryotic and eukaryotic cells decient in
57 mesenchymal transition (EMT) phenotype, with morphological DNA repair (Beck and Brubaker, 1973; Fraval et al., 1978). As
58 changes including acquisition of a spindle-like broblastic pheno- illustrated in Fig. 3, several molecular mechanisms leading to
59 type, downregulation of E-cadherin, upregulation of mesenchymal apoptosis have been implicated in cisplatin treatment of human
60 markers (vimentin, Snail and Slug), and increased capability of cancers.
61 invasion and migration in case of human lung cancer cell lines
62 (Wang et al., 2014). 5.1. Cisplatin-induced oxidative stress
63 It has been hypothesized that Rac1, a Rho GTPase, is implicated
64 in HNSCC insensitivity to chemo-radiotherapy resulting in tumour Under normal physiological conditions, cells control reactive oxy-
65 recurrence development in head and neck squamous cell carci- gen species levels by balancing the generation of reactive oxygen
66 noma (Skvortsov et al., 2014). Cisplatin resistance in colorectal species with their elimination by scavenging system (reduced

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 7

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42 Fig. 3. Overview of molecular mechanisms of cisplatin in cancer treatment.

43
44 glutathione-GSH, superoxide dismutase-SOD, and catalase-CAT). But Excessive reactive oxygen species can induce apoptosis through
45 under oxidative stress conditions, excessive reactive oxygen species both the extrinsic and intrinsic pathways (Ozben, 2007). In the
46 can damage cellular proteins, lipids and DNA, leading to fatal lesions in extrinsic pathway of apoptosis, reactive oxygen species are gener-
47 cells that contribute to carcinogenesis. Cancer cells exhibit greater ated by Fas ligand as an upstream event for Fas activation via
48 reactive oxygen species stress than normal cells do, partly due to phosphorylation, which is necessary for subsequent recruitment of
49 oncogenic stimulation, increased metabolic activity and mitochondrial Fas-associated protein with death domain and caspase 8 as well as
50 malfunction. Oxidative stress is the one of most important mechan- apoptosis induction (Gupta et al., 2012). In the intrinsic pathway,
51 isms involved in cisplatin toxicity. The mitochondrion is the primary reactive oxygen species function to facilitate cytochrome c release
52 target for cisplatin induced oxidative stress, resulting in loss of by activating pore-stabilizing proteins (Bcl-2 and Bcl-xL) as well as
53 mitochondrial protein sulfhydrl group, calcium uptake inhibition and inhibiting pore-destabilizing proteins (Bcl-2-associated X protein,
54 reduction of mitochondrial membrane potential (Saad et al., 2004) Bcl-2 homologous antagonist/killer) (Martindale and Holbrook,
55 Exposure to oxidative stress can upset regular biological func- 2002). An even higher reactive oxygen species level can result in
56 tions. Cisplatin also induces reactive oxygen species that trigger both apoptosis and necrosis in cancer cells (Hampton and
57 cell death besides DNA damage. Cell death occurs upon immediate Orrenius, 1997). Reactive oxygen species can also induce cell death
58 activation of numerous signaling pathways, whereas the denite through autophagy, which is a self-catabolic process involving
59 pathways depend on the (cancer) cell. The formation of reactive sequestration of cytoplasmic contents (exhausted organelles and
60 oxygen species depends on the concentration of cis-diamminedi- protein aggregates), for degradation in lyposomes (Shrivastava
61 chloro platinum(II) and the length of exposure (Brozovic et al., et al., 2011)
62 2010). The intracellular redox homeostasis is maintained by the
63 thiol group (SH) containing molecules. Under certain conditions a 5.2. Cisplatin modulation of calcium signaling
64 thiol group may lead to formation of thiyl radicals that in turn can
65 interact with molecular oxygen, therefore generating reactive Cisplatin, under low intracellular chloride ion concentrations,
66 oxygen species (Desoize, 2002). has been shown to hydrolyze into variously charged reactive

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
8 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 species including monoaqua [cis-(NH)PtCI(HO)] and diaquated regulating the release of mitochondrial cytochrome c in response
2 [cis-(NH)Pt(HO)]2 forms (Jennerwein and Andrews, 1995; to DNA damage. Cisplatin-induced genotoxic stress activates
3 Aggarwal et al., 1980). These hydrolyzed forms of cisplatin have multiple signal transduction pathways, which can contribute to
4 been shown to be 1000 times more reactive than normal cisplatin, apoptosis or chemo resistance.
5 and act through the inhibition of mitochondrial respiration by
6 uncoupling oxidative phosphorylation (Aggarwal, 1993). This 5.4. Cisplatin and protein kinase C
7 result in an efux of calcium from the mitochondria and a
8 temporary increase in the cellular calcium levels, which is thought Protein kinase C [PKC] is a family closely related phospholipid
9 to play a signicant role in the disruption of normal calcium dependent enzymes that play critical roles in signal transduction
10 homeostasis, and hence cell function. and cell regulation (Basu and Sivaprasad, 2007; Newton, 2003;
11 Mitochondrial glutathione (GSH) seems to be essential in the Basu, 1993; Nishizuka, 1992). There have been contrasting on
12 regulation of inner mitochondrial permeability and enzyme func- reports whether activation or down regulation of PKC is necessary
13 tion by keeping SH in the reduced state. When the SH-groups of for cisplatin sensitization (Isonishi et al., 1990; Hirata et al., 1993).
14 enzymes are not maintained in a reduced form, they become The effects of PKC on cellular sensitivity/resistance to cisplatin
15 inactivated. Cisplatin induced toxicities, especially nephrotoxicity, depend on the pattern of the PKC isozymes as well as on the
16 seem to be related to a decrease in the intracellular concentrations cellular context (Basu and Krishnamurthy, 2010).
17 of GSH and protein bound SH-groups. Nicotinamide adenine
18 dinucleotide (NADH), which helps to maintain SH groups, declines 5.5. Cisplatin and mitogen-activated protein kinase (MAPK)
19 with cisplatin treatment. Consequently, this depletion of GSH and
20 NADH appears to result in the inhibition of some dehydrogenases, Mitogen activated protein kinases are a family of structurally
21 resulting in the uncoupling of oxidative phosphorylation leading related serine/threonine protein kinases that coordinate various
22 to hydroxyl radical formation and oxidative stress. These free extra cellular signals to regulate cell growth and survival (Chang
23 radicals attack polyunsaturated lipids and proteins and initiate and Karin, 2001; Johnson and Lapadat, 2002; Marshall, 1995).
24 lipid per oxidation. This process becomes autolytic and causes Cisplatin has been shown to cause activation of ERK in several cell
25 severe damage to membrane integrity (Aggarwal, 1998). types although there are controversies whether activation of ERK
26 In summary, cisplatin's disruption of calcium homeostasis prevents or contributes to cisplatin induced cell death (Tang et al.,
27 initiates primary events such as lipid per oxidation and enzyme 2010; Wang et al., 2000; Yeh et al., 2002; Nowak, 2002; Hayakawa
28 inhibition. These events damage the cells through mitochondrial et al., 1999; Persons et al., 1999; Basu and Tu, 2005). Cisplatin-
29 damage, inhibition of mitochondrial function, depletion of adeno- induced extracellular-signal-regulated kinase (ERK) activation pre-
30 sine triphosphate (ATP) and other cofactors. This probably leads to cedes p53-mediated DNA damage response since ERK directly
31 apoptosis and tissue necrosis. Thus, it seems that elevated calcium phosphorylates p53 causing up regulation of p21, 45kd-growth
32 levels, via calcium supplementation, may act as another means of arrest and DNA damage (GADD45), and mouse double minute
33 cytoprotection, by competing for binding sites with cisplatin and 2 homolog (Mdm2) (DeHaan et al., 2001).Thus, activation of ERK
34 prevent various toxicities associated with it. may cause cell cycle arrest allowing time for the repair of cisplatin-
35 induced DNA damage via p53.
36 5.3. Cisplatin-induced cell apoptosis
37 5.6. Cisplatin and jun amino-terminal kinase (JNK)
38 Apoptosis is a controlled type of cell death which is energy-
39 dependent leading to cell shrinkage, chromatin condensation, c-Jun N-terminal kinase or stress activated protein kinase is
40 membrane budding, phosphatidylserine externalization, and acti- activated by various stress stimuli, including DNA damage. Both cis
41 vation of a family of cysteine proteases called caspases (Salvesen and trans forms of cisplatin activate the JNK pathway. p73, a
42 and Dixit, 1997; Cummings et al., 2000). Caspase activation is the proapoptotic member of p53 forms a complex with JNK leads to
43 key step in the beginning of apoptosis, and several stimuli activate cisplatin induced apoptosis (Jones et al., 2007).
44 caspases, including those that activate plasma membrane death
45 receptors (caspase 8) and cause mitochondrial dysfunction (cas- 5.7. Cisplatin and p38 mitogen-activated protein kinase (MAPK)
46 pase 9). Caspases are either initiators or executioners of apoptosis.
47 Initiator caspases include caspases 8 and 9, and activation of these Environmental stress is an important moderator of cisplatin-
48 caspases results in activation of downstream or executioner induced apoptosis, by activating p38 MAPK family. It has already
49 caspases such as caspases 3 and 7 (Salvesen and Abrams, 2004). been identied that EGFR as a substrate for p38 MAPK and
50 Executioner caspases are accountable for many of the biochemical cisplatin-induced receptor internalization was triggered by p38-
51 characteristics of apoptosis, including cleavage and activation of mediated phosphorylation of the receptor (Winograd-Katz et al.,
52 poly (ADP-ribose) polymerase and of the inhibitor of caspase 2006). p38 MAPK has been shown to mediate its effect via p18
53 activator domain protein, which leads to DNA fragmentation. (Hamlet), a p38 MAPK-regulated protein, which interacts with p53
54 Cisplatin primarily induces cell death by apoptosis and a defect and stimulates the transcription of proapoptotic genes PUMA and
55 in apoptotic signaling could also confer cisplatin resistance. There NOXA to induce apoptosis (Cuadrado et al., 2007). Therefore, the
56 are two major pathways of apoptotic cell death (Nunez et al., 1998; p38 MAPK pathway plays a critical role in regulating cisplatin-
57 Kischkel et al., 1995). The extrinsic pathway is initiated when induced apoptosis.
58 ligands bind to the tumor necrosis factor- (TNF) receptor super
59 family followed by oligomerization and recruitment of procaspase- 5.8. Cisplatin and AKT
60 8 via adapter molecules to form the death-inducing signaling
61 complex (DISC). The intrinsic pathway is initiated by cellular Akt belongs to a family of serine/threonine kinases which act
62 stress, such as DNA damage, resulting in release of cytochrome-c downstream of phosphoinositide 3-kinase (P13K) and plays a
63 from the mitochondria causing activation of procaspase-9 through critical role in the cell survival (Brazil and Hemmings, 2001).
64 the interaction with apoptosis promoting activating factor-1 It has been demonstrated that cisplatin-induced DNA damage
65 (APAF-1) and formation of an active apoptosome complex. Bcl-2 caused phosphorylation of BAD at ser136 via Akt (Hayakawa
66 family proteins regulate DNA damage-induced apoptosis by et al., 2000). Basically, BAD is phosphorylated in cells with

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 9

1 cisplatin- induced DNA damage and this BAD phosphorylation is 5.12. Cisplatin modulation of gene expression
2 required for cell viability after cisplatin treatment in both cisplatin
3 resistant and -sensitive cells. Previous study showed that cisplatin- In vitro studies suggest that cisplatin resistance can result from
4 induced DNA damage triggers the phosphorylation of both BAD epigenetic changes at the molecular and cellular levels, including
5 Ser-112 via an ERK cascade and BAD Ser-136 via a PI-3 K-PKB/Akt reduced accumulation of the platinum compounds by either active
6 cascade. Iinhibition of either of these cascades sensitizes ovarian efux/sequestration/secretion or impaired inux, detoxication
7 cancer cells to cisplatin (Hayakawa et al., 2000). by GSH conjugates, metallothioneins and other antioxidants,
8 It has been demonstrated that cisplatin-induced phosphoryla- increased levels of DNA damage repair (nucleotide excision repair
9 tion of BAD Ser-112 is MEK-dependent; while cisplatin-induced and mismatch repair), changes in DNA methylation status, altera-
10 phosphorylation of BAD Ser- 136 is PI-3K-Akt-dependent. In tions of membrane protein trafcking as a result of defective
11 addition, cisplatin induced phosphorylation of both BAD Ser-112 organization and distribution of the cytoskeleton, overexpression
12 and Ser-136 is involved in maintaining cell viability after cisplatin of chaperones, up- or down-regulated expression of microRNA
13 treatment. All these results suggest that the ERK and PI-3K-Akt (miRNA1), transcription factors and small GTPases, inactivation
14 signaling cascades converge at BAD to suppress the apoptotic (Shen et al., 2012).
15 effect of BAD (Hayakawa et al., 2000). As a consequence of reduced uptake or retention, the formation
16 of platinum-DNA adducts is correspondingly decreased, reducing
17 5.9. Cisplatin and signaling for DNA damage cytotoxicity, resulting in more resistance to the platinum com-
18 pound. Signicant reductions in platinum-DNA adduct formation
19 Mutations are caused by several signals associated with stress. (9-fold) and ribosomal RNA gene-specic interstrand cross-
20 DNA once is damaged is followed by activation of cell cycle check link formation (12-fold) have been reported in studies with
21 points, there by delaying the cell cycle progression either to repair human hepatoma cisplatin-resistant 7404-CP20 cells. However
22 or to permanently eliminate the cells by inducing cell death. The the removal rates of the total platinum-DNA adducts and gene-
23 response of cells to cisplatin induced DNA damage will decide the specic interstrand cross-links were similar to those in their
24 fate of a cell to live or die (Basu and Krishnamurthy, 2010). parental 7404 cells (Johnson et al., 1996).
25 Cisplatin also induces endoplasmic reticulum stress and
26 5.10. p53 and DNA damage response to cisplatin treatment nucleus-independent apoptotic signaling (Mandic et al., 2003).
27 Because cisplatin is a DNA-damaging agent and an inducer of
28 p53 is a short lived protein, which is activated (phosphoryla- apoptosis, it is reasonable to expect changes in HSPs in the
29 tion) by Ataxia telangiectasia mutated (ATM) on DNA damage development of cellular resistance to the drug. HSP60 was sig-
30 signal. The activated p53 now in turn activates Mdm2, which is E3 nicantly expressed in both human cervical and liver carcinoma
31 ubiquitin ligase. p53 can transactivate genes involved in cell cycle (Shen et al., 2012). Up-regulation of HSP27, HSP70, HSP72, GRP78,
32 progression, DNA repair and apoptosis. p53 can regulate cisplatin and HSP90 have been reported to be involved in CP-r ovarian
33 induced cell death by several mechanisms like: degradation of cancer (Arts et al., 1999; Liu et al., 2002), breast cancer (Vargas-
34 ice-like inhibitory protein (FLIP), direct binding and counter- Roig et al., 1998), colon cancer (Bel et al., 1999), cervical HeLa
35 acting the antiapoptotic function of B-cell lymphoma-extra-large cells (Huang et al., 2000) and laryngeal carcinoma cells (Brozovic
36 (Bcl-xL), over expression of phosphatase and tensin homolog et al., 2001).
37 (PTEN) and inhibition of AMPK (Basu and Krishnamurthy, 2010). The low molecular weight GTP-binding proteins of the Rho
38 Although p53 plays an important role in cisplatin induced DNA family, including RhoA, RhoB, RhoC, and Rac, all belong to the Ras
39 damage response, p53 negative cells also respond to cisplatin tumor suppressor gene family. Reduced expression of small
40 induced DNA damage. This suggests the existence of alternate GTPases such as Rab5, Rac1, and RhoA was detected in human
41 pathway for this event to occur upon stress. cisplatin-resistant cells in association with decreased accumula-
42 tion of radiolabeled compounds (Shen et al., 2004). The ribosomal
43 5.11. Cyclobutanedicarboxylate (c-abl) and DNA damage response protein L36 was found to present cisplatin resistance in human
44 to cisplatin treatment carcinoma cells (Shen et al., 2006). In human breast cancer MCF-7
45 cells, reduced expression of the ribosomal protein P0 was found by
46 c-Abl is tyrosine kinase receptor and contains nuclear localiza- proteomic analysis (Smith et al., 2007). Moreover, ectopic L37 over
47 tion motifs and nuclear export signals. Upon DNA damage, c-Abl is expression can attenuate the DNA damage response mediated by
48 recruited from cytoplasm to nucleus, later associate with and p53 (Llanos and Serrano, 2010).
49 phosphorylates MEK kinase 1. This complex activates jun amino- It has been reported that transcription factors such as Y-box
50 terminal kinase /stress-activated protein kinases (JNK/SAPK) binding protein-1, CCAAT-binding transcription factor 2, activating
51 (Kharbanda et al., 2000). It is also demonstrated that activation transcription factor 4, zinc nger factor 143, the nuclear transcrip-
52 of c-Abl and JNK is conditional upon the recognition of cisplatin tion factor- B, the microphthalmia-associated transcription factor,
53 induced DNA damage by the mismatch repair (MMR) system since the forkhead transcription factor O, and mitochondrial transcrip-
54 c-Abl response is absent in MMR-decient cells (Nehme et al., tion factor A, play a major role in cisplatin resistance (Shen et al.,
55 1999). 2012). It was rst found that the levels of nuclear factor (Erythroid-
56 The MMR/c-Abl binds to p73, a member of p53 family to start Derived 2)-like 2 (Nrf2) expression in Nrf2-decient murine
57 apoptosis (Gong et al., 1999). Phosphorylation and stabilization of embryonic cells and human ovarian cancer SK-OV cisplatin-resis-
58 p73 can increase its proapoptotic function by disassociating from tant cells correlate with the extent of resistance to cisplatin. Loss of
59 p63, another p53 family member (Basu and Krishnamurthy, 2010). Nrf2 or inhibition by siRNA resulted in increased cell death,
60 C-Jun will enhance the p73 stability by binding to transcription co cytotoxicity, and apoptosis in response to cisplatin treatment
61 activator Yap1, preventing proteasomal degradation of p73 and compared with control cells (Cho et al., 2008). Also, increased
62 results in the recruitment of p300 to trigger transcription of expression in GCF2-transfected KB-3-1 cells results in reduced
63 proapoptotic genes (Levy et al., 2008).Cisplatin can trigger clea- RhoA expression, disruption of actin-lamin dynamics, mislocali-
64 vage of c-Abl which is a substrate for caspase and proteolytic zation of the membrane protein MRP1, and reduction in cisplatin
65 cleavage of c-Abl was shown to be important for cisplatin-induced accumulation, leading to 3-fold resistance to cisplatin (Shen et al.,
66 apoptosis (Machuy et al., 2004). 2012).

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
10 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 5.13. Computational studies of cisplatin Thermodynamics of hydrolysis of cisplatin and bis(ethylene-


2 diamine) dichloroplatinum(II) using a combination of molecular
3 Cisplatin is one of the most effective anticancer drugs currently mechanics for obtaining optimized geometries of the reactants
4 in use. Following the nding of its antitumor activity over three and products, and the extended Huckel method for deriving
5 decades ago, strong research has been carried out to reveal the charge distributions and electronic energies were studied. This
6 details of its cytotoxic activity and to design analogs with reduced hydrolysis was studied by several others using various levels of
7 side effects (Mantri and Baik, 2006). Recently, computational theory, where a common approach adopted has been to use DFT to
8 studies have been conducted to complement experimental works. optimize the geometries of the key intermediates and reevaluate
9 The hydrolysis process of cisplatin which activates the drug was their energies using higher level ab initio methods, and/or adding
10 the goal of past research. cisplatinDNA interactions are the next solvation corrections based on continuum dielectric models.
11 theoretical studies, since DNA is the primary target of the drug. describes the interaction of transplatin and the fragments trans-
12 At present, to study the thermodynamics and kinetics of not only PtCl2(NH3) and Pt(NH3)23 with G:C and A:T base pairs using DFT,
13 cisplatinDNA complexes, but also of other complexes such as Pt followed by an MP2-based energy analysis (Nikolov et al., 1994;
14 (II)-based cisplatin analogs, other transition metal complexes, and Mantri and Baik, 2006; Zhang et al., 2001; Lau and Deubel, 2005).
15 DNA binding organic molecules, both quantum mechanical and Cisplatin has been known to bind to guanine with much greater
16 molecular mechanical methods are being used. Future research is preference over adenine, which is somewhat surprising because it
17 aiming at elucidating the role of repair enzymes in modulating the could be argued that the inductive effects of the electron-
18 cytotoxic activity of DNA binding agents (Mantri and Baik, 2006). withdrawing oxo group at the C6 position of guanine should
19 The combination of dramatically improved computer hardware reduce the electron density at N7 compared to adenine that has
20 and robust, sophisticated, and numerically efcient modeling software an electron-donating amino group at the C6 position, thus making
21 has allowed for employing high levels of theory to examine various the N7 of guanine less nucleophilic compared to adenine (Mantri
22 aspects of cisplatin chemistry using computational chemistry techni- and Baik, 2006). The major difference between guanine and
23 ques. The very rst computational studies on cisplatin aimed at better adenine is that N1 of guanine is protonated, while adenine
24 understanding the hydrolysis of cisplatin, because the activation by exposes an N1 lone pair. The presence of the N1 proton diminishes
25 hydrolysis had long been established as the rate-limiting step (Basch the delocalization of electron density over the nitrogen lone pairs
26 et al., 1986). An intriguing question had been the preferential of the purine skeleton resulting in greater localization of electron
27 antitumor activity of the cis isomer over the trans isomer, a fact then density on the N3 and N7 atoms of guanine, compared to adenine
28 attributed to the steric effects of binding DNA bases. It was eventually where the electron density is delocalized over the N1, N3, and N7
29 discovered that the cis orientation of the leaving groups allowed atoms (Mantri and Baik, 2006). In an attempt to identify yet
30 binding of two adjacent guanine bases on the same strand, which another subtle electronic feature that could help in distinguishing
31 resulted in a large kink in the DNA helix, causing polymerases to be between the reactivity of adenine and guanine, it was discovered
32 stalled at the kink. However, it was demonstrated that the trans that the [Pt] fragment is a poor -donor; thus -back donation
33 isomer is favored over the cis isomer in all cases due to reduction in does not appear to play a major role (Mantri and Baik, 2006).
34 ligandligand repulsion, especially in the case of anionic ligands. These Cisplatin binds to other cellular components, resulting in either
35 differences become negligibly small when favorable hydrogen bonding deactivation of the drug and/or disruption of normal biochemical
36 interactions are possible between the ammine ligands and the other pathways. Thus, signicant efforts have focused on understanding
37 labile ligands (Basch et al., 1986). the binding of cisplatin with other entities found in the cells; in
38 There is increasing awareness now that one important reason particular S- and N-containing ligands that are expected have the
39 for cisplatin's performance is associated to the high mobility group greatest afnity for platinum based on good hardness- softness
40 (HMG) proteins that bind to DNA and protect the drugDNA matching. It was reported a thorough DFT study which compared
41 adducts against excision repair (Wang and Lippard, 2005). To the PtL bond strengths of a series of tri- ammine platinum(II)
42 enable comparison of the binding of Pt(II) to various positions in complexes with oxygen-, nitrogen- and sulfur-donor ligands as
43 the four nucleobases in DNA and to keep computational costs to a models of competing ligands encountered in a biological system
44 minimum, Pt(NH3)23 was used as the fragment that interacted (Deubel, 2002). In another study, the kinetics of the substitution
45 with the bases. The ranking of the bases followed the order: G(N7) reaction of various nitrogen- and sulfur ligands with the activated
46 4C(N3)4C(O2)4G(O6)4A(N3)EA(N1)4A(N7)4G(N3)4T(O4) cisplatin complex was modeled using DFT. This study revealed that
47 4T(O2), based on differential Pt(II) binding energies. On the basis of kinetically N-donor ligands were preferred over S-donors, with the
48 the ranking above and the fact that most of the potential binding selectivity originating from electrostatic rather than orbital-based
49 sites are in reality unavailable for binding due to WatsonCrick interaction (Deubel, 2004).
50 base pairing, the N7 position of guaninein particular intrastrand Other areas of active study on cisplatin are degree of local
51 binding to two adjacent guanineswas conrmed as the preferred bending/unwinding upon cisplatin binding, the disruption of base
52 binding site, in full agreement with experimental evidence (Mantri stacking and other local distortions, the thermodynamics of the
53 and Baik, 2006) various possible adducts, the directional preference in the forma-
54 Density functional theory was applied to provide new insights tion of the bifunctional adducts, the ineffectiveness of transplatin,
55 into the structure and reactivity of cis- and transplatin and related the effect of cisplatin binding on the dynamics of DNA, and the
56 hydrolysis product (Carloni et al., 2000) and also to compare the binding and recognition of HMG domain and/or DNA repair
57 electronic structures of cisplatin and its second-generation analog proteins to these adducts (Mantri and Baik, 2006). Computational
58 carboplatin (Carloni and Andreoni, 1996). Results suggested that models have made signicant contributions to understanding the
59 the replacement of the chloride ligands of cisplatin by carboxylate nature and reactivity of cisplatin and allowed for delineating many
60 ligands in carboplatin resulted in a greater stability of the metal- features of how it binds to DNA.
61 ligand bonds in the latter, leading to potentially higher activation
62 energy for substitution reactions. Later, a more comprehensive
63 study comparing the geometric, electronic, and vibrational proper- 6. Toxicological effects of cisplatin
64 ties of cisplatin using different basis sets, including pure ECP and
65 hybrid ECP with various electron correlation methods up to the Cisplatin interacts with DNA, and forms covalent adduct with
66 MP4 level, and DFT, was performed (Pavankumar et al., 1999). purine DNA bases and this platinum compound, interaction is the

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 11

1 root cause for cytotoxic effect of cisplatin (Yousef et al., 2009). suggesting an active accumulation of drug by renal parenchymal
2 Cisplatin treatment has been associated with several toxic side cells. Studies in recent years have identied two different mem-
3 effects including nephrotoxicity (de Jongh et al., 2003), hepato- brane transporters capable of transporting cisplatin into cells: Ctr1
4 toxicity and Cardiotoxicity (Al-Majed, 2007). Many cardiac events and OCT2 (Ishida et al., 2002). Later, cisplatin is biotransformed in
5 have been reported in many case reports including electro- the kidney into cysteinyl glycine conjugates and other high thiols
6 cardiographicchanges, arrhythmias, myocarditis, cardiomyopathy by some localized enzymes.
7 and congestive heart failure (Yousef et al., 2009). Decrease
8 in antioxidant defense system is reported due to oxidative stress 6.4. Other organ toxicity
9 through the generation of reactive oxygen species, including
10 antioxidant enzymes and non enzymatic molecules, reduced Other cisplatin-induced organ toxicities such as ototoxicity,
11 glutathione, are major alterations in the cisplatin toxicity (Kart gastrotoxicity, myelosuppression, allergic reactions and some
12 et al., 2010). reproductive toxic effects have also been reported (Hartmann
13 et al., 2000; Hartmann and Lipp, 2003)
14 6.1. Hepatotoxicity
15
16 High dosage of cisplatin may lead to hepatotoxicity (dos Santos 7. Conclusions
17 et al., 2007).Oxidative stress is the main reason for cisplatin-
18 induced toxicity possibly due to depletion of reduced glutathione Cisplatin is one of the most effective anticancer agents widely
19 GSH (Yilmaz et al., 2004), also many studies reported that there used in the treatment of solid tumors. It has been extensively used
20 were a signicant elevation in the hepatic malonaldehyde (MDA) for the cure of different types of neoplasms including head and
21 and reduction in the level of antioxidant enzymes in rats treated neck, lung, ovarian, leukemia, breast, brain, kidney and testicular
22 with cisplatin (Yilmaz, et al., 2005; Mansour et al., 2006). The most cancers. In general, cisplatin and other platinum-based com-
23 sensitive biomarkers directly concerned in causing the cellular pounds are considered as cytotoxic drugs which kill cancer cells
24 damage and toxicity are transaminases, because they are cyto- by damaging DNA, inhibiting DNA synthesis and mitosis, and
25 plasmic in location and are released into the circulation after inducing apoptotic cell death. Several molecular mechanisms of
26 cellular damage. Elevation of the hepatic enzymes level in serum action including induction of oxidative stress as characterized by
27 and bilirubin are the indicators for impaired liver functions (Iseri reactive oxygen species production and lipid peroxidation, induc-
28 et al., 2007). Cisplatin hepatotoxicity was shown to be exacerbated tion of p53 signaling and cell cycle arrest, down-regulation of
29 by increased expression of cytochrome P450-2E1 enzyme (Caro proto-oncogenes and anti-apoptotic proteins, and activation of
30 and Cederbaum, 2004).Observed histopathological changes will be both intrinsic and extrinsic pathways of apoptosis. However,
31 necrosis and degeneration of hepatocytes with inammatory cells cisplatin chemotherapy is also associated with substantial side
32 inltration around portal area with sinusoidal dilatation (Kart effects that include hepatotoxic, nephrotoxic, cardiotoxic, neuro-
33 et al., 2010; Cetin et al., 2006). Recent studies have focused on toxic and/or hematotoxic damage. Also, some patients may relapse
34 methods for protection of cisplatin-induced Hepatotoxicity using from cisplatin treatment with their cancers being refractory to
35 various agents, such as selenium (Liao et al., 2008) and vitamin E cisplatin regimen. Hence, combination therapies of cisplatin with
36 (Naziroglu et al., 2004; Iraz et al., 2005). other drugs are common practice in the treatment of human
37 cancers. Findings of several studies have suggested that other
38 6.2. Cardiotoxicity compounds combined with cisplatin constitute the best therapeu-
39 tic approach to overcome drug resistance and reduce the undesir-
40 Leakage of lactate dehydrogenase (LDH) and creatine kinase able side effects. Moving forward, combinatorial strategies which
41 (CK) from cardiac myocytes is due to cardiotoxicity could be a target multiple mechanisms, such as reducing cisplatin uptake and
42 secondary event following cisplatin-induced lipid peroxidation of reducing inammation, may offer the best chance for clinically
43 cardiac membranes. Degeneration and necrosis of cardiac muscle meaningful prevention.
44 ber cells with brous tissue reaction and vacuolated cytoplasm of
45 many muscle cells and blood vessels are inated with blood are
46 the histological changes of cisplatin induced toxicology (Al-Majed Uncited references Q2
47 et al., 2006).
48 (Hayakawa et al. (2004), Hazardous (2014), Ohta et al., (2006);
49 6.3. Nephrotoxicity The chemical book data base (2014))
50
51 The kidney accumulates cisplatin to a greater degree than other
52 organs and is the major route for its excretion. The cisplatin Acknowledgments
53 concentration in proximal tubular epithelial cells is about 5 times
54 the serum concentration (Kuhlmann et al., 1997).The dispropor- The research described in this publication was made possible Q4
55 tionate accumulation of cisplatin in kidney tissue contributes to by a grant from the National Institutes of Health (Grant no.
56 cisplatin-induced nephrotoxicity (Arany and Sarstein, 2003). G12MD007581) through the RCMI Center for Environmental
57 Biosynthesis of amino acid lysine and methionine yields a qua- Health at Jackson State University.
58 ternary ammonium compound called Carnitine, which is required for
59 the transport of fatty acids from the cytosol into the mitochondria
References
60 during the breakdown of lipids to generate metabolic energy. Kidney
61 damage is caused by the inhibition of Carnitine synthesis and also by
Abrams, T.J., Lee, L.B., Murray, L.J., Pryer, N.K., Cherrington, J.M., 2003. SU11248
62 the Carnitine reabsorption by the proximal tubule of nephron, which inhibits KIT and platelet-derived growth factor receptor beta in preclinical
63 is due to declined production of Carnitine. models of human small cell lung cancer. Mol. Cancer Ther. 2, 471478.
64 Cisplatin is cleared by the kidney by both glomerular ltration Agarwal, R., Kaye, S.B., 2003. Ovarian cancer: strategies for overcoming resistance to
chemotherapy. Nat. Rev. Cancer 3, 502516.
65 and tubular secretion (Yao et al., 2007; Ishida et al., 2002). Aggarwal, S.K., 1993. A histochemical approach to the mechanism of action of
66 Cisplatin concentrations within the kidney exceed those in blood cisplatin and its analogues. J. Histochem. Cytochem. 41, 10531073.

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
12 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 Aggarwal, S.K., 1998. Calcium modulation of toxicities due to cisplatin. Met. Based Chian, S., Li, Y.Y., Wang, X.J., Tang, X.W., 2014. Luteolin sensitizes two oxaliplatin-
2 Drugs 5, 7781. resistant colorectal cancer cell lines to chemotherapeutic drugs via inhibition of
Aggarwal, S.K., Broomhead, J.A., Fairlie, D.P., Whitehouse, M.W., 1980. Platinum the nrf2 pathway. Asian Pac. J. Cancer Prev. 15, 29112916.
3 drugs: combined anti-lymphoproliferative and nephrotoxicity assay in rats. Chang, L., Karin, M., 2001. Mammalian MAP kinase signalling cascades. Nature 410,
4 Cancer Chemother. Pharmacol. 4, 249258. 3740.
5 Ajani, J.A., Winter, K.A., Gunderson, L.L., Pedersen, J., Benson III, A.B., Thomas Jr., C.R., Chen, G., Huynh, M., Fehrenbacher, L., West, H., Lara Jr., P.N., Yavorkovsky, L.L.,
Mayer, R.J., Haddock, M.G., Rich, T.A., Willett, C., 2008. Fluorouracil, mitomycin, Russin, M., Goldstein, D., Gandara, D., Lau, D., 2009. Phase II trial of irinotecan
6 and radiotherapy vs uorouracil, cisplatin, and radiotherapy for carcinoma of the and carboplatin for extensive or relapsed small-cell lung cancer. J. Clin. Oncol.
7 anal canal: a randomized controlled trial. JAMA 299, 19141921. 27, 14011404.
8 Akron. 2009. The Chemical Database. The Department of Chemistry at the Cho, J.M., Manandhar, S., Lee, H.R., Park, H.M., Kwak, M.K., 2008. Role of the Nrf2-
University of Akron.http://ull.chemistry.uakron.edu/erd and search on CAS antioxidant system in cytotoxicity mediated by anticancer cisplatin: implica-
9
number. (accessed 25.03.14.). tion to cancer cell resistance. Cancer Lett. 260, 96108.
10 Alberts, D.S., Manning, M.R., Coulthard, S.W., Koopmann Jr., C.F., Herman, T.S., 1981. Colombo, N., Sessa, C., Landoni, F., Sartori, E., Pecorelli, S., Mangioni, C., 1986.
11 Adriamycin/cis-platinum/cyclophosphamide combination chemotherapy for Cisplatin, vinblastine, and bleomycin combination chemotherapy in metastatic
12 advanced carcinoma of the parotid gland. Cancer 47, 645648. granulosa cell tumor of the ovary. Obstet. Gynecol. 67, 265268.
Al-Majed, A.A., 2007. Carnitine deciency provokes cisplatin-induced hepatotoxi- Creagan, E.T., Woods, J.E., Schutt, A.J., OFallon, J.R., 1983. Cyclophosphamide,
13 city in rats. Basic Clin. Pharmacol. Toxicol. 100, 145150. adriamycin, and cis-diamminedichloroplatinum (II) in the treatment of
14 Al-Majed, A.A., Sayed-Ahmed, M.M., Al-Yahya, A.A., Aleisa, A.M., Al-Rejaie, S.S., advanced nonsquamous cell head and neck cancer. Cancer 52, 20072010.
15 Al-Shabanah, O.A., 2006. Propionyl-L-carnitine prevents the progression of Crino, L., Scagliotti, G., Marangolo, M., Figoli, F., Clerici, M., De, M.F., Salvati, F.,
cisplatin-induced cardiomyopathy in a carnitine-depleted rat model. Pharmacol. Cruciani, G., Dogliotti, L., Pucci, F., Paccagnella, A., Adamo, V., Altavilla, G.,
16 Res. 53, 278286. Incoronato, P., Trippetti, M., Mosconi, A.M., Santucci, A., Sorbolini, S., Oliva, C.,
17 Alizadehnohi, M., Nabiuni, M., Nazari, Z., Safaeinejad, Z., Irian, S., 2012. The Tonato, M., 1997. Cisplatin-gemcitabine combination in advanced non-small-
18 synergistic cytotoxic effect of cisplatin and honey bee venom on human cell lung cancer: a phase II study. J. Clin. Oncol. 15, 297303.
ovarian cancer cell line A2780cp. J. Venom. Res. 3, 2227. Cuadrado, A., Lafarga, V., Cheung, P.C., Dolado, I., Llanos, S., Cohen, P., Nebreda, A.R.,
19 Apostolou, P., Toloudi, M., Chatziioannou, M., Ioannou, E., Knocke, D.R., Nester, J., 2007. A new p38 MAP kinase-regulated transcriptional coactivator that
20 Komiotis, D., Papasotiriou, I., 2013. AnvirzelTM in combination with cisplatin in stimulates p53-dependent apoptosis. EMBO J. 26, 21152126.
21 breast, colon, lung, prostate, melanoma and pancreatic cancer cell lines. BMC Cummings, B.S., Lasker, J.M., Lash, L.H., 2000. Expression of glutathione-dependent
Pharmacol. Toxicol. 14, 18.
22 enzymes and cytochrome P450s in freshly isolated and primary cultures of
Arany, I., Sarstein, R.L., 2003. Cisplatin nephrotoxicity. Semin. Nephrol. 23, proximal tubular cells from human kidney. J. Pharmacol. Exp. Ther. 293,
23 460464. 677685.
24 Ardizzoni, A., Rosso, R., Salvati, F., Fusco, V., Cinquegrana, A., De, P.M., Serrano, J., de Jongh, F.E., van Veen, R.N., Veltman, S.J., de, W.R., van der, Burg M.E., van den,
25 Pennucci, M.C., Soresi, E., Crippa, M., 1991. Activity of doxorubicin and cisplatin Bent M.J., Planting, A.S., Graveland, W.J., Stoter, G., Verweij, J., 2003. Weekly
combination chemotherapy in patients with diffuse malignant pleural
26 high-dose cisplatin is a feasible treatment option: analysis on prognostic factors
mesothelioma. An Italian lung cancer task force (FONICAP) phase II study.
for toxicity in 400 patients. Br. J. Cancer 88, 11991206.
27 Cancer 67, 29842987.
Decatris, M.P., Sundar, S., OByrne, K.J., 2004. Platinum-based chemotherapy in
28 Arts, H.J., Hollema, H., Lemstra, W., Willemse, P.H., De Vries, E.G., Kampinga, H.H.,
metastatic breast cancer: current status. Cancer Treat. Rev. 30, 5381.
Van der Zee, A.G., 1999. Heat-shock-protein-27 (hsp27) expression in ovarian
29 DeHaan, R.D., Yazlovitskaya, E.M., Persons, D.L., 2001. Regulation of p53 target gene
carcinoma: relation in response to chemotherapy and prognosis. Int. J. Cancer
expression by cisplatin-induced extracellular signal-regulated kinase. Cancer
30 84, 234238.
Chemother. Pharmacol. 48, 383388.
31 Baniahmad, A., Tsai, M.J., 1993. Mechanisms of transcriptional activation by steroid
Deng, L., Zhang, E., Chen, C., 2013. Synergistic interaction of beta-galactosyl-
hormone receptors. J. Cell. Biochem. 51, 151156.
32 Basch, H., Krauss, M., Stevens, W.J., Cohen, D., 1986. Binding of Pt(NH3)23 to nucleic
pyrrolidinyl diazeniumdiolate with cisplatin against three tumor cells. Arch.
33 Pharm. Res. 36, 619625.
acid bases. Inorg. Chem. 25, 684688.
Desoize, B., 2002. Cancer and metals and metal compounds: part Icarcinogenesis.
34 Basu, A., 1993. The potential of protein kinase C as a target for anticancer treatment.
Crit. Rev. Oncol. Hematol. 42, 13.
Pharmacol. Ther. 59, 257280.
35 Desoize, B., Madoulet, C., 2002. Particular aspects of platinum compounds used at
Q5 Basu, A., Krishnamurthy, S., 2010. Cellular responses to cisplatin-induced DNA
36 present in cancer treatment. Crit. Rev. Oncol. Hematol. 42, 317325.
damage. J. Nucleic Acids, 2010.
Deubel, D.V., 2002. On the competition of the purine bases, functionalities of
37 Basu, A., Sivaprasad, U., 2007. Protein kinase cepsilon makes the life and death
peptide side chains, and protecting agents for the coordination sites of
38 decision. Cell. Signal. 19, 16331642.
dicationic cisplatin derivatives. J. Am. Chem. Soc. 124, 58345842.
Basu, A., Tu, H., 2005. Activation of ERK during DNA damage-induced apoptosis
39 involves protein kinase Cdelta. Biochem. Biophys. Res. Commun. 334,
Deubel, D.V., 2004. Factors governing the kinetic competition of nitrogen and sulfur
ligands in cisplatin binding to biological targets. J. Am. Chem. Soc. 126,
40 10681073.
Beck, D.J., Brubaker, R.R., 1973. Effect of cis-platinum(II)diamminodichloride on 59996004.
41 Dexeus, F.H., Logothetis, C.J., Sella, A., Amato, R., Kilbourn, R., Fitz, K., Striegel, A.,
wild type and deoxyribonucleic acid repair decient mutants of Escherichia coli.
42 J. Bacteriol. 116, 12471252. 1991. Combination chemotherapy with methotrexate, bleomycin and cisplatin
43 Bel, C.A., Chatterjee, S., Gosky, D.M., Berger, S.J., Berger, N.A., 1999. Increased for advanced squamous cell carcinoma of the male genital tract. J. Urol. 146,
sensitivity of human colon cancer cells to DNA cross-linking agents after GRP78 12841287.
44 Dhar, S., Kolishetti, N., Lippard, S.J., Farokhzad, O.C., 2011. Targeted delivery of a
up-regulation. Biochem. Biophys. Res. Commun. 257, 361368.
45 Bertolini, G., Roz, L., Perego, P., Tortoreto, M., Fontanella, E., Gatti, L., Pratesi, G., cisplatin prodrug for safer and more effective prostate cancer therapy in vivo.
46 Fabbri, A., Andriani, F., Tinelli, S., Roz, E., Caserini, R., Lo, V.S., Camerini, T., Proc. Natl. Acad. Sci. U.S.A. 108, 18501855.
Dimery, I.W., Legha, S.S., Shirinian, M., Hong, W.K., 1990. Fluorouracil, doxorubicin,
47 Mariani, L., Delia, D., Calabro, E., Pastorino, U., Sozzi, G., 2009. Highly
tumorigenic lung cancer CD133 cells display stem-like features and are cyclophosphamide, and cisplatin combination chemotherapy in advanced or
48 recurrent salivary gland carcinoma. J. Clin. Oncol. 8, 10561062.
spared by cisplatin treatment. Proc. Natl. Acad. Sci. U.S.A. 106, 1628116286.
49 Brazil, D.P., Hemmings, B.A., 2001. Ten years of protein kinase B signalling: a hard dos Santos, N.A., Martins, N.M., Curti, C., Pires Bianchi, M.L., dos Santos, A.C., 2007.
50 Akt to follow. Trends Biochem. Sci. 26, 657664. Dimethylthiourea protects against mitochondrial oxidative damage induced by
Brozovic, A., Ambriovic-Ristov, A., Osmak, M., 2010. The relationship between cisplatin in liver of rats. Chem. Biol. Interact. 170, 177186.
51 Dreyfuss, A.I., Clark, J.R., Fallon, B.G., Posner, M.R., Norris Jr., C.M., Miller, D., 1987.
cisplatin-induced reactive oxygen species, glutathione, and BCL-2 and resis-
52 tance to cisplatin. Crit. Rev. Toxicol. 40, 347359. Cyclophosphamide, doxorubicin, and cisplatin combination chemotherapy for
53 Brozovic, A., Simaga, S., Osmak, M., 2001. Induction of heat shock protein 70 in advanced carcinomas of salivary gland origin. Cancer 60, 28692872.
drug-resistant cells by anticancer drugs and hyperthermia. Neoplasma 48, Du, N., Li, X., Li, F., Zhao, H., Fan, Z., Ma, J., Fu, Y., Kang, H., 2013. Intrapleural
54
99103. combination therapy with bevacizumab and cisplatin for non-small cell lung
55 Byun, J.M., Jeong, D.H., Lee, D.S., Kim, J.R., Park, S.G., Kang, M.S., Kim, Y.N., Lee, K.B., cancermediated malignant pleural effusion. Oncol. Rep. 29, 23322340.
56 Sung, M.S., Kim, K.T., 2013. Tetraarsenic oxide and cisplatin induce apoptotic Einzig, A.I., Hochster, H., Wiernik, P.H., Trump, D.L., Dutcher, J.P., Garowski, E.,
57 synergism in cervical cancer. Oncol. Rep. 29, 15401546. Sasloff, J., Smith, T.J., 1991. A phase II study of taxol in patients with malignant
Canetta, R., Rozencweig, M., Carter, S.K., 1985. Carboplatin: the clinical spectrum to melanoma. Investig. New Drugs 9, 5964.
58 date. Cancer Treat. Rev. 12 (Suppl. A), S125S136. Einzig, A.I., Wiernik, P.H., Sasloff, J., Runowicz, C.D., Goldberg, G.L., 1992. Phase II
59 Carloni, P., Andreoni, W., 1996. Platinum-modied nucleobase pairs in the solid study and long-term follow-up of patients treated with taxol for advanced
60 state: a theoretical study. J. Phys. Chem. 100, 1779717800. ovarian adenocarcinoma. J. Clin. Oncol. 10, 17481753.
Carloni, P., Sprik, M., Andreoni, W., 2000. Key steps of the cis-platinDNA Eisenberger, M.A., 1985. Supporting evidence for an active treatment program for
61 advanced salivary gland carcinomas. Cancer Treat. Rep. 69, 319321.
interaction: density functional theory-based molecular dynamics simulations.
62 J. Phys. Chem. B 104, 823835. Forastiere, A.A., 1994. Paclitaxel (Taxol) for the treatment of head and neck cancer.
63 Caro, A.A., Cederbaum, A.I., 2004. Oxidative stress, toxicology, and pharmacology of Semin. Oncol. 21, 4952.
CYP2E1. Annu. Rev. Pharmacol. Toxicol. 44, 2742. Fraval, H.N., Rawlings, C.J., Roberts, J.J., 1978. Increased sensitivity of UV-repair-
64
Cetin, R., Devrim, E., Kilicoglu, B., Avci, A., Candir, O., Durak, I., 2006. Cisplatin decient human cells to DNA bound platinum products which unlike thymine
65 impairs antioxidant system and causes oxidation in rat kidney tissues: possible dimers are not recognized by an endonuclease extracted from Micrococcus
66 protective roles of natural antioxidant foods. J. Appl. Toxicol. 26, 4246. luteus. Mutat. Res. 51, 121132.

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 13

1 Frezza, M., Hindo, S., Chen, D., Davenport, A., Schmitt, S., Tomco, D., Dou, Q.P., 2010. Jennerwein, M., Andrews, P.A., 1995. Effect of intracellular chloride on the cellular
2 Novel metals and metal complexes as platforms for cancer therapy. Curr. pharmacodynamics of cis-diamminedichloroplatinum(II). Drug Metab. Dispos.
Pharm. Des. 16, 18131825. 23, 178184.
3 Fujii, S., Kitano, S., Ikenaka, K., Shirasaka, T., 1979. Effect of coadministration of Johnson, G.L., Lapadat, R., 2002. Mitogen-activated protein kinase pathways
4 uracil or cytosine on the anti-tumor activity of clinical doses of 1-(2-tetrahy- mediated by ERK, JNK, and p38 protein kinases. Science 298, 19111912.
5 drofuryl)-5-uorouracil and level of 5-uorouracil in rodents. Gann 70, Johnson, S.W., Shen, D., Pastan, I., Gottesman, M.M., Hamilton, T.C., 1996. Cross-
209214. resistance, cisplatin accumulation, and platinum-DNA adduct formation and
6
Go, R.S., Adjei, A.A., 1999. Review of the comparative pharmacology and clinical removal in cisplatin-sensitive and -resistant human hepatoma cell lines. Exp.
7 activity of cisplatin and carboplatin. J. Clin. Oncol. 17, 409422. Cell Res. 226, 133139.
8 Gong, J.G., Costanzo, A., Yang, H.Q., Melino, G., Kaelin Jr., W.G., Levrero, M., Wang, J.Y., Jones, E.V., Dickman, M.J., Whitmarsh, A.J., 2007. Regulation of p73-mediated
9 1999. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin- apoptosis by c-Jun N-terminal kinase. Biochem. J. 405, 617623.
induced DNA damage. Nature 399, 806809. Kakar, S.S., Jala, V.R., Fong, M.Y., 2012. Synergistic cytotoxic action of cisplatin and
10 Gottesman, M.M., Fojo, T., Bates, S.E., 2002. Multidrug resistance in cancer: role of withaferin A on ovarian cancer cell lines. Biochem. Biophys. Res. Commun. 423,
11 ATP-dependent transporters. Nat. Rev. Cancer 2, 4858. 819825.
12 Goodsell, D.S., 2006. The molecular perspective: cisplatin. Stem Cells 24, 514515. Kart, A., Cigremis, Y., Karaman, M., Ozen, H., 2010. Caffeic acid phenethyl ester
Gupta, S.C., Hevia, D., Patchva, S., Park, B., Koh, W., Aggarwal, B.B., 2012. Upsides and (CAPE) ameliorates cisplatin-induced hepatotoxicity in rabbit. Exp. Toxicol.
13 downsides of reactive oxygen species for cancer: the roles of reactive oxygen Pathol. 62, 4552.
14 species in tumorigenesis, prevention, and therapy. Antioxid. Redox Signal. 16, Kelland, L., 2007. The resurgence of platinum-based cancer chemotherapy. Nat. Rev.
15 12951322. Cancer 7, 573584.
Hampton, M.B., Orrenius, S., 1997. Dual regulation of caspase activity by hydrogen Khan, A.B., DSouza, B.J., Wharam, M.D., Champion, L.A., Sinks, L.F., Woo, S.Y.,
16
peroxide: implications for apoptosis. FEBS Lett. 414, 552556. McCullough, D.C., Leventhal, B.G., 1982. Cisplatin therapy in recurrent child-
17 Hartmann, J.T., Fels, L.M., Knop, S., Stolt, H., Kanz, L., Bokemeyer, C., 2000. hood brain tumors. Cancer Treat. Rep. 66, 20132020.
18 A randomized trial comparing the nephrotoxicity of cisplatin/ifosfamide-based Kharbanda, S., Pandey, P., Yamauchi, T., Kumar, S., Kaneki, M., Kumar, V., Bharti, A.,
19 combination chemotherapy with or without amifostine in patients with solid Yuan, Z.M., Ghanem, L., Rana, A., Weichselbaum, R., Johnson, G., Kufe, D., 2000.
tumors. Investig. New Drugs 18, 281289. Activation of MEK kinase 1 by the c-Abl protein tyrosine kinase in response to
20 Hartmann, J.T., Lipp, H.P., 2003. Toxicity of platinum compounds. Expert Opin. DNA damage. Mol. Cell. Biol. 20, 49794989.
21 Pharmacother. 4, 889901. Kim, Y.H., Shin, S.W., Kim, B.S., Kim, J.H., Kim, J.G., Mok, Y.J., Kim, C.S., Rhyu, H.S.,
22 Hayakawa, J., Mittal, S., Wang, Y., Korkmaz, K.S., Adamson, E., English, C., Ohmichi, M., Hyun, J.H., Kim, J.S., 1999. Paclitaxel, 5-uorouracil, and cisplatin combination
McClelland, M., Mercola, D., 2004. Identication of promoters bound by c-Jun/
23 chemotherapy for the treatment of advanced gastric carcinoma. Cancer 85,
ATF2 during rapid large-scale gene activation following genotoxic stress. Mol. 295301.
24 Cell. 16, 521535. Kischkel, F.C., Hellbardt, S., Behrmann, I., Germer, M., Pawlita, M., Krammer, P.H.,
25 Hayakawa, J., Ohmichi, M., Kurachi, H., Ikegami, H., Kimura, A., Matsuoka, T., Peter, M.E., 1995. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated pro-
26 Jikihara, H., Mercola, D., Murata, Y., 1999. Inhibition of extracellular signal-
teins form a death-inducing signaling complex (DISC) with the receptor. EMBO
regulated protein kinase or c-Jun N-terminal protein kinase cascade, differen-
27 J. 14, 55795588.
tially activated by cisplatin, sensitizes human ovarian cancer cell line. J. Biol.
Koch, M., Krieger, M.L., Stolting, D., Brenner, N., Beier, M., Jaehde, U., Wiese, M.,
28 Chem. 274, 3164831654.
Royer, H.D., Bendas, G., 2013. Overcoming chemotherapy resistance of ovarian
29 Hayakawa, J., Ohmichi, M., Kurachi, H., Kanda, Y., Hisamoto, K., Nishio, Y., Adachi, K.,
cancer cells by liposomal cisplatin: molecular mechanisms unveiled by gene
Tasaka, K., Kanzaki, T., Murata, Y., 2000. Inhibition of BAD phosphorylation
30 expression proling. Biochem. Pharmacol. 85, 10771090.
either at serine 112 via extracellular signal-regulated protein kinase cascade or
31 Koizumi, W., Narahara, H., Hara, T., Takagane, A., Akiya, T., Takagi, M., Miyashita, K.,
at serine 136 via Akt cascade sensitizes human ovarian cancer cells to cisplatin.
Nishizaki, T., Kobayashi, O., Takiyama, W., Toh, Y., Nagaie, T., Takagi, S.,
32 Cancer Res. 60, 59885994.
Yamamura, Y., Yanaoka, K., Orita, H., Takeuchi, M., 2008. S-1 plus cisplatin
33 Hazardous Substances Data Bank.National Library of Medicine. http://toxnet.nlm.
versus S-1 alone for rst-line treatment of advanced gastric cancer (SPIRITS
nih.gov/ cgi-bin/sis/htmlgen?HSDB and search on CAS number. (accessed
34 19.03.14.).
trial): a phase III trial. Lancet Oncol. 9, 215221.
Kosmas, C., Tsavaris, N.B., Malamos, N.A., Vadiaka, M., Koufos, C., 2001. Phase II
35 Hirata, J., Kikuchi, Y., Kita, T., Imaizumi, E., Tode, T., Ishii, K., Kudoh, K., Nagata, I.,
study of paclitaxel, ifosfamide, and cisplatin as second-line treatment in
36 1993. Modulation of sensitivity of human ovarian cancer cells to cis-diammi-
relapsed small-cell lung cancer. J. Clin. Oncol. 19, 119126.
nedichloroplatinum(II) by 12-O-tetradecanoylphorbol-13-acetate and D,L-
37 buthionine-S,R-sulphoximine. Int. J. Cancer 55, 521527.
Kuhlmann, M.K., Burkhardt, G., Kohler, H., 1997. Insights into potential cellular
38 mechanisms of cisplatin nephrotoxicity and their clinical application. Nephrol.
Hitt, R., Castellano, D., Hidalgo, M., Garcia-Carbonero, R., Pena, M., Brandariz, A.,
Dial. Transplant. 12, 24782480.
39 Millan, J.M., Alvarez Vincent, J.J., Cortes-Funes, H., 1998. Phase II trial of cisplatin
Lau, J.K.-C., Deubel, D.V., 2005. Hydrolysis of the anticancer drug cisplatin: pitfalls
and gemcitabine in advanced squamous-cell carcinoma of the head and neck.
40 in the interpretation of quantum chemical calculations. J. Chem. Theory
Ann. Oncol. 9, 13471349.
41 Holmes, F.A., Walters, R.S., Theriault, R.L., Forman, A.D., Newton, L.K., Raber, M.N., Comput. 2, 103106.
42 Buzdar, A.U., Frye, D.K., Hortobagyi, G.N., 1991. Phase II trial of taxol, an active Le, C.T., Brisgand, D., Douillard, J.Y., Pujol, J.L., Alberola, V., Monnier, A., Riviere, A.,
drug in the treatment of metastatic breast cancer. J. Natl. Cancer Inst. 83, Lianes, P., Chomy, P., Cigolari, S., 1994. Randomized study of vinorelbine and
43 cisplatin versus vindesine and cisplatin versus vinorelbine alone in advanced
17971805.
44 Holzer, A.K., Manorek, G.H., Howell, S.B., 2006. Contribution of the major copper non-small-cell lung cancer: results of a European multicenter trial including
45 inux transporter CTR1 to the cellular accumulation of cisplatin, carboplatin, 612 patients. J. Clin. Oncol. 12, 360367.
Legha, S.S., Ring, S., Papadopoulos, N., Raber, M., Benjamin, R.S., 1990. A phase II
46 and oxaliplatin. Mol. Pharmacol. 70, 13901394.
Huang, T.G., Ip, S.M., Yeung, W.S., Ngan, H.Y., 2000. Changes in p21WAF1, pRb, trial of taxol in metastatic melanoma. Cancer 65, 24782481.
47 Levy, D., Adamovich, Y., Reuven, N., Shaul, Y., 2008. Yap1 phosphorylation by c-Abl
Mdm-2, Bax and Bcl-2 expression in cervical cancer cell lines transfected with a
48 p53 expressing adenovirus. Eur. J. Cancer 36, 249256. is a critical step in selective activation of proapoptotic genes in response to DNA
49 IARC, 1981. Cisplatin. In Some Antineoplastic and Immunosuppressive Agents, damage. Mol. Cell. 29, 350361.
IARCMonographs on the Evaluation of Carcinogenic Risk of Chemicals to Liao, Y., Lu, X., Lu, C., Li, G., Jin, Y., Tang, H., 2008. Selection of agents for prevention
50 of cisplatin-induced hepatotoxicity. Pharmacol. Res. 57, 125131.
Humans. International Agency for Research on Cancer, Lyon, France, pp.
51 151164. Light, B.W., Yu, W.D., McElwain, M.C., Russell, D.M., Trump, D.L., Johnson, C.S., 1997.
52 Ichinose, Y., Yosimori, K., Yoneda, S., Kuba, M., Kudoh, S., Niitani, H., 2000. UFT plus Potentiation of cisplatin antitumor activity using a vitamin D analogue in a
cisplatin combination chemotherapy in the treatment of patients with murine squamous cell carcinoma model system. Cancer Res. 57, 37593764.
53
advanced nonsmall cell lung carcinoma: a multiinstitutional phase II trial. For Lin, C.C., Yeh, H.H., Huang, W.L., Yan, J.J., Lai, W.W., Su, W.P., Chen, H.H., Su, W.C.,
54 2013. Metformin enhances cisplatin cytotoxicity by suppressing signal trans-
the Japan UFT Lung Cancer Study Group. Cancer 88, 318323.
55 Iraz, M., Kalcioglu, M.T., Kizilay, A., Karatas, E., 2005. Aminoguanidine prevents ducer and activator of transcription-3 activity independently of the liver kinase
56 ototoxicity induced by cisplatin in rats. Ann. Clin. Lab. Sci. 35, 329335. B1-AMP-activated protein kinase pathway. Am. J. Respir. Cell Mol. Biol. 49,
Iseri, S., Ercan, F., Gedik, N., Yuksel, M., Alican, I., 2007. Simvastatin attenuates 241250.
57 Lin, J., Zhang, Q., Lu, Y., Xue, W., Xu, Y., Zhu, Y., Hu, X., 2014. Downregulation of
cisplatin-induced kidney and liver damage in rats. Toxicology 230, 256264.
58 Ishida, S., Lee, J., Thiele, D.J., Herskowitz, I., 2002. Uptake of the anticancer drug HIPK2 increases resistance of bladder cancer cell to cisplatin by regulating
59 cisplatin mediated by the copper transporter Ctr1 in yeast and mammals. Proc. Wip1. PLoS One 9, e98418.
60 Natl. Acad. Sci. U.S.A. 99, 1429814302. Lin, X., Okuda, T., Holzer, A., Howell, S.B., 2002. The copper transporter CTR1
Isonishi, S., Andrews, P.A., Howell, S.B., 1990. Increased sensitivity to cis-diammi- regulates cisplatin uptake in Saccharomyces cerevisiae. Mol. Pharmacol. 62,
61 nedichloroplatinum(II) in human ovarian carcinoma cells in response to 11541159.
62 treatment with 12-O-tetradecanoylphorbol 13-acetate. J. Biol. Chem. 265, Liu, J.R., Opipari, A.W., Tan, L., Jiang, Y., Zhang, Y., Tang, H., Nunez, G., 2002.
63 36233627. Dysfunctional apoptosome activation in ovarian cancer: implications for
Iwasaki, Y., Nagata, K., Nakanishi, M., Natuhara, A., Kubota, Y., Ueda, M., Arimoto, T., chemoresistance. Cancer Res. 62, 924931.
64
Hara, H., 2005. Double-cycle, high-dose ifosfamide, carboplatin, and etoposide Llanos, S., Serrano, M., 2010. Depletion of ribosomal protein L37 occurs in response
65 followed by peripheral blood stem-cell transplantation for small cell lung to DNA damage and activates p53 through the L11/MDM2 pathway. Cell Cycle
66 cancer. Chest 128, 22682273. 9, 40054012.

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
14 S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology ()

1 Lynch, H.T., Casey, M.J., Snyder, C.L., Bewtra, C., Lynch, J.F., Butts, M., Godwin, A.K., 2009. Parness, J., Horwitz, S.B., 1981. Taxol binds to polymerized tubulin in vitro. J. Cell
2 Hereditary ovarian carcinoma: heterogeneity, molecular genetics, pathology, and Biol. 91, 479487.
management. Mol. Oncol. 3, 97137. Persons, D.L., Yazlovitskaya, E.M., Cui, W., Pelling, J.C., 1999. Cisplatin-induced
3 Machuy, N., Rajalingam, K., Rudel, T., 2004. Requirement of caspase-mediated activation of mitogen-activated protein kinases in ovarian carcinoma cells:
4 cleavage of c-Abl during stress-induced apoptosis. Cell Death. Differ. 11, inhibition of extracellular signal-regulated kinase activity increases sensitivity
5 290300. to cisplatin. Clin. Cancer Res. 5, 10071014.
Mandic, A., Hansson, J., Linder, S., Shoshan, M.C., 2003. Cisplatin induces endo- Pignon, J.P., Tribodet, H., Scagliotti, G.V., Douillard, J.Y., Shepherd, F.A., Stephens, R.J.,
6 Dunant, A., Torri, V., Rosell, R., Seymour, L., Spiro, S.G., Rolland, E., Fossati, R.,
plasmic reticulum stress and nucleus-independent apoptotic signaling. J. Biol.
7 Chem. 278, 91009106. Aubert, D., Ding, K., Waller, D., Le, C.T., 2008. Lung adjuvant cisplatin evaluation:
8 Mansour, H.H., Hafez, H.F., Fahmy, N.M., 2006. Silymarin modulates Cisplatin- a pooled analysis by the LACE Collaborative Group. J. Clin. Oncol. 26,
induced oxidative stress and hepatotoxicity in rats. J. Biochem. Mol. Biol. 39, 35523559.
9
656661. Pinto-Leite, R., Arantes-Rodrigues, R., Palmeira, C., Colaco, B., Lopes, C., Colaco, A.,
10 Q6 Mantri, Y., Baik, M.H., 2006. Computational Studies: Cisplatin, Encyclopedia of Costa, C., da Silva, V.M., Oliveira, P., Santos, L., 2013. Everolimus combined with
11 Inorganic Chemistry. John Wiley & Sons, Ltd.. cisplatin has a potential role in treatment of urothelial bladder cancer. Biomed.
12 Marshall, C.J., 1995. Specicity of receptor tyrosine kinase signaling: transient Pharmacother. 67, 116121.
versus sustained extracellular signal-regulated kinase activation. Cell 80, Previati, M., Lanzoni, I., Corbacella, E., Magosso, S., Guaran, V., Martini, A., Capitani, S.,
13 179185. 2006. Cisplatin-induced apoptosis in human promyelocytic leukemia cells. Int. J.
14 Martindale, J.L., Holbrook, N.J., 2002. Cellular response to oxidative stress: signaling Mol. Med. 18, 511516.
15 for suicide and survival. J. Cell. Physiol. 192, 115. Rodriguez-Fernandez, E., Manzano, J.L., Alonso, A., Almendral, M.J., Perez-Andres, M.,
Matsuki, M., Takahashi, A., Katou, S., Takayanagi, A., Takagi, Y., Kamata, K., 2013. Orfao, A., Criado, J.J., 2009. Fluorescent cisplatin analogues and cytotoxic activity.
16 Curr. Med. Chem. 16, 43144327.
Pathological complete response to gemcitabine and cisplatin chemotherapy for
17 advanced upper tract urothelial carcinoma: a case report. Nihon Hinyokika Rosenberg, b., Vancamp, L., Krigas, T., 1965. Inhibition of cell division in escherichia
18 Gakkai Zasshi 104, 3337. coli by electrolysis products from a platinum electrode. Nature 205, 698699.
McGuire, W.P., Rowinsky, E.K., Rosenshein, N.B., Grumbine, F.C., Ettinger, D.S., Saad, S.Y., Najjar, T.A., Alashari, M., 2004. Role of non-selective adenosine receptor
19 blockade and phosphodiesterase inhibition in cisplatin-induced nephrogonadal
Armstrong, D.K., Donehower, R.C., 1989. Taxol: a unique antineoplastic agent
20 with signicant activity in advanced ovarian epithelial neoplasms. Ann. Intern. toxicity in rats. Clin. Exp. Pharmacol. Physiol. 31, 862867.
21 Med. 111, 273279. Salvesen, G.S., Abrams, J.M., 2004. Caspase activationstepping on the gas or
releasing the brakes? Lessons from humans and ies. Oncogene 23, 27742784.
22 McGuire, W.P., Hoskins, W.J., Brady, M.F., Kucera, P.R., Partridge, E.E., Look, K.Y.,
Clarke-Pearson, D.L., Davidson, M., 1996. Cyclophosphamide and cisplatin Salvesen, G.S., Dixit, V.M., 1997. Caspases: intracellular signaling by proteolysis. Cell
23 91, 443446.
compared with paclitaxel and cisplatin in patients with stage III and stage IV
24 ovarian cancer. N. Engl. J. Med. 334, 16. Saramaki, A., Banwell, C.M., Campbell, M.J., Carlberg, C., 2006. Regulation of the
25 Meng, F., Sun, G., Zhong, M., Yu, Y., Brewer, M.A., 2013. Anticancer efcacy of human p21(waf1/cip1) gene promoter via multiple binding sites for p53 and
cisplatin and trichostatin A or 5-aza-20 -deoxycytidine on ovarian cancer. the vitamin D3 receptor. Nucleic Acids Res. 34, 543554.
26 Sarosy, G., Kohn, E., Stone, D.A., Rothenberg, M., Jacob, J., Adamo, D.O., Ognibene, F.P.,
Br. J. Cancer 108, 579586.
27 Milczarek, M., Rosinska, S., Psurski, M., Maciejewska, M., Kutner, A., Wietrzyk, J.,
Cunnion, R.E., Reed, E., 1992. Phase I study of taxol and granulocyte colony-
28 stimulating factor in patients with refractory ovarian cancer. J. Clin. Oncol. 10,
2013. Combined colonic cancer treatment with vitamin D analogs and irinote-
11651170.
29 can or oxaliplatin. Anticancer Res. 33, 433444.
Scher, H.I., 1992. A randomized comparison of cisplatin alone or in combination
Minami, D., Takigawa, N., Takeda, H., Takata, M., Ochi, N., Ichihara, E., Hisamoto, A.,
30 with methotrexate, vinblastine, and doxorubicin in patients with metastatic
Hotta, K., Tanimoto, M., Kiura, K., 2013. Synergistic effect of olaparib with
31 urothelial carcinoma: a cooperative group study. J. Urol. 148, 16251626.
combination of cisplatin on PTEN-decient lung cancer cells. Mol. Cancer Res.
Shaili, E., 2014. Platinum anticancer drugs and photochemotherapeutic agents:
32 11, 140148.
recent advances and future developments. Sci. Prog. 97, 2040.
33 Murphy, W.K., Fossella, F.V., Winn, R.J., Shin, D.M., Hynes, H.E., Gross, H.M., Davilla, E.,
Shen, D.W., Liang, X.J., Gawinowicz, M.A., Gottesman, M.M., 2004. Identication of
Leimert, J., Dhingra, H., Raber, M.N., 1993. Phase II study of taxol in patients with
34 cytoskeletal [14C]carboplatin-binding proteins reveals reduced expression and
untreated advanced non-small-cell lung cancer. J. Natl. Cancer Inst. 85, 384388.
disorganization of actin and lamin in cisplatin-resistant cell lines. Mol.
35 Nagpal, S., Na, S., Rathnachalam, R., 2005. Noncalcemic actions of vitamin D
Pharmacol. 66, 789793.
36 receptor ligands. Endocr. Rev. 26, 662687.
Shen, D.W., Liang, X.J., Suzuki, T., Gottesman, M.M., 2006. Identication by
Natarajan, G., Malathi, R., Holler, E., 1999. Increased DNA-binding activity of cis-1,1-
37 cyclobutanedicarboxylatodiammineplatinum(II) (carboplatin) in the presence
functional cloning from a retroviral cDNA library of cDNAs for ribosomal
38 protein L36 and the 10-kDa heat shock protein that confer cisplatin resistance.
of nucleophiles and human breast cancer MCF-7 cell cytoplasmic extracts:
Mol. Pharmacol. 69, 13831388.
39 activation theory revisited. Biochem. Pharmacol. 58, 16251629. Shen, D.W., Pouliot, L.M., Hall, M.D., Gottesman, M.M., 2012. Cisplatin resistance: a
Naziroglu, M., Karaoglu, A., Aksoy, A.O., 2004. Selenium and high dose vitamin E
40 cellular self-defense mechanism resulting from multiple epigenetic and genetic
administration protects cisplatin-induced oxidative damage to renal, liver and changes. Pharmacol. Rev. 64, 706721.
41 lens tissues in rats. Toxicology 195, 221230. Shi, X., Wu, S., Yang, Y., Tang, L., Wang, Y., Dong, J., Lu, B., Jiang, G., Zhao, W., 2014.
42 Nehme, A., Baskaran, R., Nebel, S., Fink, D., Howell, S.B., Wang, J.Y., Christen, R.D., AQP5 silencing suppresses p38 MAPK signaling and improves drug resistance Q7
43 1999. Induction of JNK and c-Abl signalling by cisplatin and oxaliplatin in in colon cancer cells. Tumour Biol. ([Epub ahead of print]).
mismatch repair-procient and -decient cells. Br. J. Cancer 79, 11041110. Shrivastava, A., Kuzontkoski, P.M., Groopman, J.E., Prasad, A., 2011. Cannabidiol
44 Nessa, M.U., Beale, P., Chan, C., Yu, J.Q., Huq, F., 2011. Synergism from combinations induces programmed cell death in breast cancer cells by coordinating the cross-
45 of cisplatin and oxaliplatin with quercetin and thymoquinone in human ovarian talk between apoptosis and autophagy. Mol. Cancer Ther. 10, 11611172.
46 tumour models. Anticancer Res. 31, 37893797. Siddik, Z.H., 2003. Cisplatin: mode of cytotoxic action and molecular basis of
Newton, A.C., 2003. Regulation of the ABC kinases by phosphorylation: protein
47 resistance. Oncogene 22, 72657279.
kinase C as a paradigm. Biochem. J. 370, 361371. Siwinska, A., Opolski, A., Chrobak, A., Wietrzyk, J., Wojdat, E., Kutner, A., Szelejewski, W.,
48 Nikolov, G.S., Trendalova, N., Schnenberger, H., Gust, R., Kritzenberger, J., Yersin, H., Radzikowski, C., 2001. Potentiation of the antiproliferative effect in vitro of
49 1994. Molecular mechanical and quantum chemical study on the species doxorubicin, cisplatin and genistein by new analogues of vitamin D. Anticancer
50 involved in the hydrolysis of cis-diamminedichloroplatinum(II) and substituted Res. 21, 19251929.
bis(ethylenediamine)dichloroplatinum(II) complexes Part I. Reactants and pro- Skvortsov, S., Dudas, J., Eichberger, P., Witsch-Baumgartner, M., Loefer-Ragg, J., Pritz,
51 ducts. Inorg. Chim. Acta 217, 159170. C., Schartinger, V.H., Maier, H., Hall, J., Debbage, P., Riechelmann, H., Lukas, P.,
52 Nishizuka, Y., 1992. Intracellular signaling by hydrolysis of phospholipids and Skvortsova, I., 2014. Rac1 as a potential therapeutic target for chemo-
53 activation of protein kinase C. Science 258, 607614. radioresistant head and neck squamous cell carcinomas (HNSCC). Br. J. Cancer
Nowak, G., 2002. Protein kinase C-alpha and ERK1/2 mediate mitochondrial 110 (11), 26772687.
54
dysfunction, decreases in active Na transport, and cisplatin-induced apopto- Smith, L., Welham, K.J., Watson, M.B., Drew, P.J., Lind, M.J., Cawkwell, L., 2007.
55 sis in renal cells. J. Biol. Chem. 277, 4337743388. The proteomic analysis of cisplatin resistance in breast cancer cells. Oncol. Res.
56 Nunez, G., Benedict, M.A., Hu, Y., Inohara, N., 1998. Caspases: the proteases of the 16, 497506.
57 apoptotic pathway. Oncogene 17, 32373245. Song, Y.D., Zhang, K.F., Liu, D., Guo, Y.Q., Wang, D.Y., Cui, M.Y., Li, G., Sun, Y.X., Shen, J.H.,
Ohta, T., Ohmichi, M., Hayasaka, T., Mabuchi, S., Saitoh, M., Kawagoe, J., Takahashi, K., Li, X.G., Zhang, L., Shi, F.J., 2014. Inhibition of EGFR-induced glucose metabolism Q8
58 Igarashi, H., Du, B., Doshida, M., Mirei, I.G., Motoyama, T., Tasaka, K., Kurachi, H., sensitizes chondrosarcoma cells to cisplatin. Tumour Biol. ([Epub ahead of print]).
59 2006. Inhibition of phosphatidylinositol 3-kinase increases efcacy of cisplatin in Stupp, R., Hegi, M.E., Mason, W.P., van den Bent, M.J., Taphoorn, M.J., Janzer, R.C., Ludwin,
60 in vivo ovarian cancer models. Endocrinology 147, 17611769. S.K., Allgeier, A., Fisher, B., Belanger, K., Hau, P., Brandes, A.A., Gijtenbeek, J., Marosi,
Ozben, T., 2007. Oxidative stress and apoptosis: impact on cancer therapy. J. Pharm. C., Vecht, C.J., Mokhtari, K., Wesseling, P., Villa, S., Eisenhauer, E., Gorlia, T., Weller, M.,
61 Sci. 96, 21812196. Lacombe, D., Cairncross, J.G., Mirimanoff, R.O., 2009. Effects of radiotherapy with
62 Pavankumar, P.N.V., Seetharamulu, P., Yao, S., Saxe, J.D., Reddy, D.G., Hausheer, F.H., concomitant and adjuvant temozolomide versus radiotherapy alone on survival in
63 1999. Comprehensive ab initio quantum mechanical and molecular orbital glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC
(MO) analysis of cisplatin: structure, bonding, charge density, and vibrational trial. Lancet Oncol. 10, 459466.
64
frequencies. J. Comput. Chem. 20, 365382. Stupp, R., Mason, W.P., van den Bent, M.J., Weller, M., Fisher, B., Taphoorn, M.J.,
65 Parkin, D.M., Bray, F., Ferlay, J., Pisani, P., 2005. Global cancer statistics, 2002. CA Belanger, K., Brandes, A.A., Marosi, C., Bogdahn, U., Curschmann, J., Janzer, R.C.,
66 Cancer J. Clin. 55, 74108. Ludwin, S.K., Gorlia, T., Allgeier, A., Lacombe, D., Cairncross, J.G., Eisenhauer, E.,

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i
S. Dasari, P. Bernard Tchounwou / European Journal of Pharmacology () 15

1 Mirimanoff, L.K., 2005. Radiotherapy plus concomitant and adjuvant temozo- Wang, X., Martindale, J.L., Holbrook, N.J., 2000. Requirement for ERK activation in
2 lomide for glioblastoma. N. Engl. J. Med. 352, 987996. cisplatin-induced apoptosis. J. Biol. Chem. 275, 3943539443.
Tang, M.K., Zhou, H.Y., Yam, J.W., Wong, A.S., 2010. c-Met overexpression con- Weiss, R.B., Christian, M.C., 1993. New cisplatin analogues in development. A
3 tributes to the acquired apoptotic resistance of nonadherent ovarian cancer review. Drugs 46, 360377.
4 cells through a cross talk mediated by phosphatidylinositol 3-kinase and Winograd-Katz, S.E., Levitzki, A., 2006. Cisplatin induces PKB/Akt activation and
5 extracellular signal-regulated kinase 1/2. Neoplasia 12, 128138. p38(MAPK) phosphorylation of the EGF receptor. Oncogene 25, 73817390.
The chemical book data base http://www.chemicalbook.com/ProductIndex_EN. Xu, X.M., Zhang, Y., Qu, D., Liu, H.B., Gu, X., Jiao, G.Y., Zhao, L., 2013. Combined
6 aspxand searchon product details. (accessed 21.03.14.). anticancer activity of osthole and cisplatin in NCI-H460 lung cancer cells
7 Tsimberidou, A.M., Braiteh, F., Stewart, D.J., Kurzrock, R., 2009. Ultimate fate of in vitro. Exp. Ther. Med. 5, 707710.
8 oncology drugs approved by the us food and drug administration without a Yao, X., Panichpisal, K., Kurtzman, N., Nugent, K., 2007. Cisplatin nephrotoxicity: a
randomized Trial. J. Clin. Oncol. 27, 62436250. review. Am. J. Med. Sci. 334, 115124.
9 Valle, J., Wasan, H., Palmer, D.H., Cunningham, D., Anthoney, A., Maraveyas, A., Yeh, P.Y., Chuang, S.E., Yeh, K.H., Song, Y.C., Ea, C.K., Cheng, A.L., 2002. Increase of the
10 Madhusudan, S., Iveson, T., Hughes, S., Pereira, S.P., Roughton, M., Bridgewater, J., resistance of human cervical carcinoma cells to cisplatin by inhibition of the
11 2010. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N. MEK to ERK signaling pathway partly via enhancement of anticancer drug-
Engl. J. Med. 362, 12731281. induced NF kappa B activation. Biochem.Pharmacol. 63, 14231430.
12
Vargas-Roig, L.M., Gago, F.E., Tello, O., Aznar, J.C., Ciocca, D.R., 1998. Heat shock Yilmaz, H.R., Iraz, M., Sogut, S., Ozyurt, H., Yildirim, Z., Akyol, O., Gergerlioglu, S.,
13 protein expression and drug resistance in breast cancer patients treated with 2004. The effects of erdosteine on the activities of some metabolic enzymes
14 induction chemotherapy. Int. J. Cancer 79, 468475. during cisplatin-induced nephrotoxicity in rats. Pharmacol. Res. 50, 287290.
15 von der, M.H., Sengelov, L., Roberts, J.T., Ricci, S., Dogliotti, L., Oliver, T., Moore, M.J., Yilmaz, H.R., Sogut, S., Ozyurt, B., Ozugurlu, F., Sahin, S., Isik, B., Uz, E., Ozyurt, H.,
Zimmermann, A., Arning, M., 2005. Long-term survival results of a randomized 2005. The activities of liver adenosine deaminase, xanthine oxidase, catalase,
16 trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, superoxide dismutase enzymes and the levels of malondialdehyde and nitric
17 doxorubicin, plus cisplatin in patients with bladder cancer. J. Clin. Oncol. 23, oxide after cisplatin toxicity in rats: protective effect of caffeic acid phenethyl
18 46024608. ester. Toxicol. Ind. Health 21, 6773.
Wang, D., Lippard, S.J., 2005. Cellular processing of platinum anticancer drugs. Nat. Youlden, D.R., Cramb, S.M., Baade, P.D., 2008. The international epidemiology of
19 Rev. Drug Discov. 4, 307320. lung cancer: geographical distribution and secular trends. J. Thorac. Oncol. 3,
20 Wang, H., Zhang, G., Zhang, H., Zhang, F., Zhou, B., Ning, F., Wang, H.S., Cai, S.H., Du, J., 819831.
21 2014. Acquisition of epithelial-mesenchymal transition phenotype and cancer Yousef, M.I., Saad, A.A., El-Shennawy, L.K., 2009. Protective effect of grape seed
stem cell-like properties in cisplatin-resistant lung cancer cells through AKT/ proanthocyanidin extract against oxidative stress induced by cisplatin in rats.
22 beta-catenin/Snail signaling pathway. Eur. J. Pharmacol. 723, 156166. Food Chem. Toxicol. 47, 11761183.
23 Q9 Wang, J., Wu, G.S., 2014. Role of autophagy in cisplatin resistance in ovarian cancer Zhang, Y., Guo, Z., You, X.Z., 2001. Hydrolysis theory for cisplatin and its analogues
24 cells. J. Biol. Chem. ([Epub ahead of print]). based on density functional studies. J. Am. Chem. Soc. 123, 93789387.
25

Please cite this article as: Dasari, S., Bernard Tchounwou, P., Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J
Pharmacol (2014), http://dx.doi.org/10.1016/j.ejphar.2014.07.025i

Vous aimerez peut-être aussi