ijms-logo

Journal Browser

Journal Browser

Platinum-Based Anti-Tumor Drugs

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Bioinorganic Chemistry".

Deadline for manuscript submissions: closed (28 September 2018) | Viewed by 43416

Special Issue Editor


E-Mail Website
Guest Editor
Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
Interests: cancer chemotherapy; platinum-based drugs; synthetic anticancer substances; cytotoxic natural products; aromatase inhibitors

Special Issue Information

Dear Colleagues,

Platinum-based anti-tumor drugs, such as cisplatin, carboplatin and oxaliplatin, are establish agents in the treatment of a diverse group of solid tumor types. Over the last few decades, thousands of platinum complexes have been synthesized and tested in preclinical screens for anti-tumor activity, but only a handful of these substances ever reached the clinic. Is the development of new platinum-base anti-cancer drugs at an end? Or is it only beginning?

The situation is improving as many non-conventional Pt anti-tumor agents begin to emerge from pre-clinical research with very promising activity. Newer Pt complexes, that violate the classical structure-activity rules, appear to act by mechanisms different to the traditional agents like cisplatin. Recently, there has been increasing interest to develop Pt(IV) anti-cancer agents due to greater possibilities for structural diversity compared to Pt(II) complexes; this can help to improve the pharmacological and pharmaceutical properties of the drug, leading to better efficacy and selectivity. Another recent trend has been to chemically couple an anti-tumor Pt(IV) complex with a second anticancer drug to generate a dual-acting drug. Once the Pt(IV) is reduced in the biological system to the Pt(II) anticancer agent,  the second anti-cancer drug is released to can act on its own. An exciting development is also the coupling of Pt anti-cancer agents to nanoparticles to improve their pharmaceutical and therapeutic properties. The mechanisms of drug uptake by cancer cells and cell death have been intensely investigated for traditional anti-cancer Pt complexes but how are the non-conventional Pt complexes taken up by cells, and which pathways do they activate to selectively kill cancer cells? Another important question is how to design new Pt complexes that overcome cancer cell resistance to the traditional Pt anti-cancer agents, and at the same time avoiding resistance altogether. Finally, what are the prospects for new Pt complexes entering clinical trials.

Prof. Dr. Patrick J. Bednarski
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Cisplatin
  • Newer anti-tumor platinum complexes
  • Non-conventional platinum complexes
  • Chiral platinum complexes
  • Nanoparticles
  • Drug-design and -targeting
  • Dual-drugs
  • Prodrugs
  • Mechanisms of cell death
  • Resistance to platinum complexes
  • Drug uptake
  • Structure-activity-relationships
  • Toxicity
  • Pharmacokinetics
  • Metabolism
  • Cancer treatment
  • Clinical trials

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

12 pages, 2273 KiB  
Article
5-Azacitidine Exerts Prolonged Pro-Apoptotic Effects and Overcomes Cisplatin-Resistance in Non-Seminomatous Germ Cell Tumor Cells
by Christoph Oing, Izudin Verem, Wael Y. Mansour, Carsten Bokemeyer, Sergey Dyshlovoy and Friedemann Honecker
Int. J. Mol. Sci. 2019, 20(1), 21; https://doi.org/10.3390/ijms20010021 - 21 Dec 2018
Cited by 27 | Viewed by 3851
Abstract
Despite high cure rates, about 20% of patients with advanced germ cell tumors (GCTs) fail cisplatin-based chemotherapy. High levels of DNA methylation have been identified in GCTs and linked to cisplatin resistance. Here, we examined the effects of DNA hypomethylating 5-azacitidine (5-aza) on [...] Read more.
Despite high cure rates, about 20% of patients with advanced germ cell tumors (GCTs) fail cisplatin-based chemotherapy. High levels of DNA methylation have been identified in GCTs and linked to cisplatin resistance. Here, we examined the effects of DNA hypomethylating 5-azacitidine (5-aza) on two embryonal carcinoma cell lines (NCCIT, 2102Ep) and their cisplatin-resistant isogenic derivatives. Effects on cell viability and cisplatin sensitivity were assessed by the trypan blue exclusion method. Western blotting was used to examine induction of apoptosis 5-aza and results were validated by flow cytometry. Single agent treatment with 5-aza strongly impacted viability and induced apoptosis at low nanomolar concentrations, both in cisplatin-sensitive and -resistant cell lines. 5-aza exerted an immediate apoptotic response, followed by a prolonged inhibitory effect on cell viability and cell-cycle progression. Sequential treatment with 5-aza and cisplatin reduced cellular survival of the cisplatin-resistant sublines already at nanomolar concentrations, suggesting a partial restoration of cisplatin sensitivity by the compound. 5-aza demonstrated anti-tumor activity as a single agent at low nanomolar concentrations in GCT cells, irrespective of cisplatin-sensitivity. 5-aza may also have the potential at least to partially restore cisplatin-sensitivity in non-seminoma cells, supporting the hypothesis that combining DNA demethylating agents with cisplatin-based chemotherapy may be a valid therapeutic approach in patients with refractory GCTs. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Figure 1

11 pages, 1378 KiB  
Article
A Pt(IV) Prodrug Combining Chlorambucil and Cisplatin: A Dual-Acting Weapon for Targeting DNA in Cancer Cells
by Diego Montagner, Dina Tolan, Emma Andriollo, Valentina Gandin and Cristina Marzano
Int. J. Mol. Sci. 2018, 19(12), 3775; https://doi.org/10.3390/ijms19123775 - 27 Nov 2018
Cited by 17 | Viewed by 4854
Abstract
In this study, two DNA-targeting agents, cisplatin and chlorambucil, were combined in a Pt(IV) prodrug, 1, which was thoroughly characterized by means of spectroscopic and spectrometric techniques. Tested towards a panel of various human tumor cell lines, this compound showed superior in [...] Read more.
In this study, two DNA-targeting agents, cisplatin and chlorambucil, were combined in a Pt(IV) prodrug, 1, which was thoroughly characterized by means of spectroscopic and spectrometric techniques. Tested towards a panel of various human tumor cell lines, this compound showed superior in vitro antitumor potential than the reference drug cisplatin. In addition, an antitumor potential of 1 was found, which is comparable to that of oxaliplatin in 3D spheroid models of colon cancer cells. Mechanistic studies performed in colon cancer cells confirmed that the conjugation of chlorambucil to Pt(IV) cisplatin-based scaffold tunes the lipophilicity of the prodrug, consequently improving the ability of the compound to accumulate into cancer cells and to target DNA, ultimately leading to apoptotic cancer cell death. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

11 pages, 2845 KiB  
Article
N-Heterocyclic Carbene-Polyethyleneimine (PEI) Platinum Complexes Inducing Human Cancer Cell Death: Polymer Carrier Impact
by May Wantz, Mathilde Bouché, Georges Dahm, Neïla Chekkat, Sylvie Fournel and Stéphane Bellemin-Laponnaz
Int. J. Mol. Sci. 2018, 19(11), 3472; https://doi.org/10.3390/ijms19113472 - 05 Nov 2018
Cited by 10 | Viewed by 3726
Abstract
The high interest in N-Heterocyclic platinum carbene complexes in cancer research stems from their high cytotoxicity to human cancer cells, their stability, as well as their ease of functionalization. However, the development of these new molecules as anticancer agents still faces multiple [...] Read more.
The high interest in N-Heterocyclic platinum carbene complexes in cancer research stems from their high cytotoxicity to human cancer cells, their stability, as well as their ease of functionalization. However, the development of these new molecules as anticancer agents still faces multiple challenges, in particular solubility in aqueous media. Here, we synthesized platinum-NHC bioconjugates that combine water-solubility and cytotoxicity by using polyethyleneimine as polymer carrier. We showed on 8 different types of cells that the activity of these conjugates is modulated by the size of the polymer and the overall density of metal ions onto polymer chains. Using HCT116 cells, the conjugates displayed an effective activity after only 45 min of exposure in vitro correlated with a quick uptake by the cells as shown by the use of various fluorescent-tagged derivatives. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

21 pages, 5334 KiB  
Article
Evaluation for Synergistic Effects by Combinations of Photodynamic Therapy (PDT) with Temoporfin (mTHPC) and Pt(II) Complexes Carboplatin, Cisplatin or Oxaliplatin in a Set of Five Human Cancer Cell Lines
by Carsten Lange and Patrick J. Bednarski
Int. J. Mol. Sci. 2018, 19(10), 3183; https://doi.org/10.3390/ijms19103183 - 16 Oct 2018
Cited by 17 | Viewed by 4273
Abstract
The platinum(II) complexes carboplatin (CBDCA), cisplatin (CDDP) and oxaliplatin (1-OHP) are used as anticancer drugs in a large number of tumour chemotherapy regimens. Many attempts have been made to combine Pt(II)-based chemotherapy with alternative treatment strategies. One such alternative anticancer approach is known [...] Read more.
The platinum(II) complexes carboplatin (CBDCA), cisplatin (CDDP) and oxaliplatin (1-OHP) are used as anticancer drugs in a large number of tumour chemotherapy regimens. Many attempts have been made to combine Pt(II)-based chemotherapy with alternative treatment strategies. One such alternative anticancer approach is known as photodynamic therapy (PDT), where a non-toxic photosensitizer (PS) produces oxidative stress via the formation of reactive oxygen species (ROS) after local illumination of the affected tissue. A very promising PS is 5,10,15,20-tetra(m-hydroxyphenyl)chlorin (mTHPC, Temoporfin), which is approved for the treatment of head and neck cancer in Europe. In the present study, a combination of mTHPC-mediated PDT and either CBDCA, CDDP, or 1-OHP was applied to five human cancer cell lines from different tumour origins. Cytotoxicity was determined by the MTT assay and synergistic effects on cytotoxicity were evaluated by calculation of Combination Indices (CI). Synergy was identified in some of the combinations, for example, with 1-OHP in three of the tested cell lines but antagonism was also observed for a number of combinations in certain cell lines. In cases of synergy, elevated ROS levels were observed after combination but apoptosis induction was not necessarily increased compared to a treatment with a single compound. Cell cycle analysis revealed a formation of apoptotic subG1 populations and S phase as well as G2/M phase arrests after combination. In conclusion, pre-treatment with mTHPC-PDT has the potential to sensitize some types of tumour cells towards Pt(II) complexes, in particular 1-OHP but synergy is highly dependent on the type of cancer. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

15 pages, 1569 KiB  
Article
Platinum Complexes Can Bind to Telomeres by Coordination
by Lina Saker, Samar Ali, Caroline Masserot, Guillaume Kellermann, Joel Poupon, Marie-Paule Teulade-Fichou, Evelyne Ségal-Bendirdjian and Sophie Bombard
Int. J. Mol. Sci. 2018, 19(7), 1951; https://doi.org/10.3390/ijms19071951 - 03 Jul 2018
Cited by 4 | Viewed by 3973
Abstract
It is suggested that several compounds, including G-quadruplex ligands, can target telomeres, inducing their uncapping and, ultimately, cell death. However, it has never been demonstrated whether such ligands can bind directly and quantitatively to telomeres. Here, we employed the property of platinum and [...] Read more.
It is suggested that several compounds, including G-quadruplex ligands, can target telomeres, inducing their uncapping and, ultimately, cell death. However, it has never been demonstrated whether such ligands can bind directly and quantitatively to telomeres. Here, we employed the property of platinum and platinum-G-quadruplex complexes to target G-rich sequences to investigate and quantify their covalent binding to telomeres. Using inductively coupled plasma mass spectrometry, surprisingly, we found that, in cellulo, in the presence of cisplatin, a di-functional platinum complex, telomeric DNA was platinated 13-times less than genomic DNA in cellulo, as compared to in vitro data. On the contrary, the amount of mono-functional platinum complexes (Pt-ttpy and Pt-tpy) bound either to telomeric or to genomic DNA was similar and occurred in a G-quadruplex independent-manner. Importantly, the quantification revealed that the low level of cisplatin bound to telomeric DNA could not be the direct physical cause of TRF2 displacement from telomeres. Altogether, our data suggest that platinum complexes can affect telomeres both directly and indirectly. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

16 pages, 2858 KiB  
Article
Synthesis and Biological Evaluation of Zeise’s Salt Derivatives with Acetylsalicylic Acid Substructure
by Alexander Weninger, Daniel Baecker, Victoria Obermoser, Dorothea Egger, Klaus Wurst and Ronald Gust
Int. J. Mol. Sci. 2018, 19(6), 1612; https://doi.org/10.3390/ijms19061612 - 30 May 2018
Cited by 15 | Viewed by 6796
Abstract
The development of novel biologically active organometallic compounds bearing an acetylsalicylic acid (ASA) substructure led to the synthesis of analogical Zeise-type salts that accordingly inhibit cyclooxygenase (COX) enzymes. In order to determine the influence of the length of the alkyl chain between the [...] Read more.
The development of novel biologically active organometallic compounds bearing an acetylsalicylic acid (ASA) substructure led to the synthesis of analogical Zeise-type salts that accordingly inhibit cyclooxygenase (COX) enzymes. In order to determine the influence of the length of the alkyl chain between the platinum(II) center and the ASA moiety, compounds with varying methylene groups (n = 1–4) were synthesized and characterized. For the propene derivative structural elucidation by X-ray crystallography was possible. Prior to evaluation of biological activity, the complexes were investigated regarding their stability in different media, such as water, physiological sodium chloride, and phosphate buffered saline. Therefore, an analytical method based on capillary electrophoresis was established. All of the compounds were tested for their COX inhibitory potential. In general, complexes with longer alkyl chains caused higher inhibition of COX enzymes and the inhibitory potential towards COX enzymes was enhanced when compared to Zeise’s salt. The growth inhibitory effects of the synthesized substances were investigated in vitro against colon carcinoma (HT-29) and breast cancer (MCF-7) cells. The IC50 values of the new derivatives ranged from 30 to 50 µM, whereas neither Zeise’s salt itself nor ASA showed any antiproliferative activity at the used concentrations. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

18 pages, 3448 KiB  
Article
Key Players of Cisplatin Resistance: Towards a Systems Pharmacology Approach
by Navin Sarin, Florian Engel, Florian Rothweiler, Jindrich Cinatl, Martin Michaelis, Roland Frötschl, Holger Fröhlich and Ganna V. Kalayda
Int. J. Mol. Sci. 2018, 19(3), 767; https://doi.org/10.3390/ijms19030767 - 07 Mar 2018
Cited by 26 | Viewed by 5001
Abstract
The major obstacle in the clinical use of the antitumor drug cisplatin is inherent and acquired resistance. Typically, cisplatin resistance is not restricted to a single mechanism demanding for a systems pharmacology approach to understand a whole cell’s reaction to the drug. In [...] Read more.
The major obstacle in the clinical use of the antitumor drug cisplatin is inherent and acquired resistance. Typically, cisplatin resistance is not restricted to a single mechanism demanding for a systems pharmacology approach to understand a whole cell’s reaction to the drug. In this study, the cellular transcriptome of untreated and cisplatin-treated A549 non-small cell lung cancer cells and their cisplatin-resistant sub-line A549rCDDP2000 was screened with a whole genome array for relevant gene candidates. By combining statistical methods with available gene annotations and without a previously defined hypothesis HRas, MAPK14 (p38), CCL2, DOK1 and PTK2B were identified as genes possibly relevant for cisplatin resistance. These and related genes were further validated on transcriptome (qRT-PCR) and proteome (Western blot) level to select candidates contributing to resistance. HRas, p38, CCL2, DOK1, PTK2B and JNK3 were integrated into a model of resistance-associated signalling alterations describing differential gene and protein expression between cisplatin-sensitive and -resistant cells in reaction to cisplatin exposure. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

17 pages, 4937 KiB  
Article
Multifaceted Mechanisms of Cisplatin Resistance in Long-Term Treated Urothelial Carcinoma Cell Lines
by Margaretha A. Skowron, Margarita Melnikova, Joep G. H. Van Roermund, Andrea Romano, Peter Albers, Jürgen Thomale, Wolfgang A. Schulz, Günter Niegisch and Michèle J. Hoffmann
Int. J. Mol. Sci. 2018, 19(2), 590; https://doi.org/10.3390/ijms19020590 - 16 Feb 2018
Cited by 26 | Viewed by 5122
Abstract
Therapeutic efficacy of cisplatin-based treatment of late stage urothelial carcinoma (UC) is limited by chemoresistance. To elucidate underlying mechanisms and to develop new approaches for overcoming resistance, we generated long-term cisplatin treated (LTT) UC cell lines, characterised their cisplatin response, and determined the [...] Read more.
Therapeutic efficacy of cisplatin-based treatment of late stage urothelial carcinoma (UC) is limited by chemoresistance. To elucidate underlying mechanisms and to develop new approaches for overcoming resistance, we generated long-term cisplatin treated (LTT) UC cell lines, characterised their cisplatin response, and determined the expression of molecules involved in cisplatin transport and detoxification, DNA repair, and apoptosis. Inhibitors of metallothioneins and Survivin were applied to investigate their ability to sensitise towards cisplatin. Cell growth, proliferation, and clonogenicity were examined after cisplatin treatment by MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, EdU (5-ethynyl-2’-deoxyuridine) incorporation assay, and Giemsa staining, respectively. Cell cycle distribution and apoptosis were quantified by flow cytometry. mRNA and protein expressions were measured by real-time quantitative (qRT)-PCR, western blot, or immunofluorescence staining. LTTs recovered rapidly from cisplatin stress compared to parental cells. In LTTs, to various extents, cisplatin exporters and metallothioneins were induced, cisplatin adduct levels and DNA damage were decreased, whereas expression of DNA repair factors and specific anti-apoptotic factors was elevated. Pharmacological inhibition of Survivin, but not of metallothioneins, sensitised LTTs to cisplatin, in an additive manner. LTTs minimise cisplatin-induced DNA damage and evade apoptosis by increased expression of anti-apoptotic factors. The observed diversity among the four LTTs highlights the complexity of cisplatin resistance mechanisms even within one tumour entity, explaining heterogeneity in patient responses to chemotherapy. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

Review

Jump to: Research

19 pages, 2076 KiB  
Review
Facilitating the Cellular Accumulation of Pt-Based Chemotherapeutic Drugs
by Ian Henry Lambert and Belinda Halling Sørensen
Int. J. Mol. Sci. 2018, 19(8), 2249; https://doi.org/10.3390/ijms19082249 - 01 Aug 2018
Cited by 12 | Viewed by 4424
Abstract
Cisplatin, carboplatin, and oxaliplatin are Pt-based drugs used in the chemotherapeutic eradication of cancer cells. Although most cancer patient cells initially respond well to the treatment, the clinical effectiveness declines over time as the cancer cells develop resistance to the drugs. The Pt-based [...] Read more.
Cisplatin, carboplatin, and oxaliplatin are Pt-based drugs used in the chemotherapeutic eradication of cancer cells. Although most cancer patient cells initially respond well to the treatment, the clinical effectiveness declines over time as the cancer cells develop resistance to the drugs. The Pt-based drugs are accumulated via membrane-bound transporters, translocated to the nucleus, where they trigger various intracellular cell death programs through DNA interaction. Here we illustrate how resistance to Pt-based drugs, acquired through limitation in the activity/subcellular localization of canonical drug transporters, might be circumvented by the facilitated uptake of Pt-based drug complexes via nanocarriers/endocytosis or lipophilic drugs by diffusion. Full article
(This article belongs to the Special Issue Platinum-Based Anti-Tumor Drugs)
Show Figures

Graphical abstract

Back to TopTop