Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Curr Pharm Des ; 29(13): 1067, 2023.
Article in English | MEDLINE | ID: mdl-37231765

ABSTRACT

An article was published in the journal "Current Pharmaceutical Design", Volume 9, No. 25, 2003, pp: 2078-2089 [1]. The first author is requesting an alteration in the name. Details of a correction are provided here. The original name published was Markus Galanski. The request is to change the name to Mathea Sophia Galanski. The original article can be found online at: https://www.eurekaselect.com/article/8545 We regret the error and apologize to readers.

2.
Chem Sci ; 7(5): 3052-3061, 2016 May 01.
Article in English | MEDLINE | ID: mdl-29997796

ABSTRACT

Nano-scale secondary ion mass spectrometry (NanoSIMS) enables trace element and isotope analyses with high spatial resolution. This unique capability has recently been exploited in several studies analyzing the subcellular distribution of Au and Pt anticancer compounds. However, these studies were restricted to cell culture systems. To explore the applicability to the in vivo setting, we developed a combined imaging approach consisting of laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS), NanoSIMS and transmission electron microscopy (TEM) suitable for multi-scale detection of the platinum distribution in tissues. Applying this approach to kidney and tumor samples upon administration of selected platinum(iv) anticancer prodrugs revealed uneven platinum distributions on both the organ and subcellular scales. Spatial platinum accumulation patterns were quantitatively assessed by LA-ICP-MS in histologically heterogeneous organs (e.g., higher platinum accumulation in kidney cortex than in medulla) and used to select regions of interest for subcellular-scale imaging with NanoSIMS. These analyses revealed cytoplasmic sulfur-rich organelles accumulating platinum in both kidney and malignant cells. Those in the tumor were subsequently identified as organelles of lysosomal origin, demonstrating the potential of the combinatorial approach for investigating therapeutically relevant drug concentrations on a submicrometer scale.

3.
Dalton Trans ; 44(2): 659-68, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25385191

ABSTRACT

The very promising results of Na-trans-[RuCl4(1H-indazole)2] (NKP-1339) in clinical studies have fuelled renewed interest in the research and development of ruthenium(III) coordination compounds for cancer therapy. By applying an improved synthetic approach to this class of coordination compounds, six new examples of the general formula (cation)-trans-[RuCl4(azole)2], where (cation) = tetrabutylammonium (Bu4N)(+) (1, 2), sodium (3, 4), azolium (5, 6), and azole = 1-methyl-indazole (1, 3, 5), 1-ethyl-indazole (2, 4, 6), have been prepared. All compounds have been characterized by elemental analysis, electrospray ionization (ESI) mass spectrometry, UV-vis-, and NMR-spectroscopy and, if possible, X-ray diffraction analysis. Furthermore, the influence of the alkyl substituent at the position N1 of the indazole backbone on the stability in aqueous media as well as on the biological activity in three human cancer cell lines (CH1, A549, and SW480) and on the cellular accumulation in SW480 cells is discussed.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Proton Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization , Spectrum Analysis/methods
4.
Met Based Drugs ; 2009: 681270, 2009.
Article in English | MEDLINE | ID: mdl-19789639

ABSTRACT

The effects of indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019, or FFC14A), the second ruthenium compound that entered clinical trials, in an in vitro model of tumour invasion and metastasis show that the antitumour effects of this compound might include also the modulation of cell behaviour although its cytotoxicity appears to be predominant over these effects. The comparison with its imidazole analogue KP418 shows however its superiority, being able to control in vitro cell growth and in some instances also in vivo tumour development. These results suggest that the activity of KP1019 is predominantly due to direct cytotoxic effects for tumour cells, evident also in vivo on primary tumour growth and that the effects on modulation of the biological behaviour of the cancer cell can be present but might have only a partial role.

5.
Curr Cancer Drug Targets ; 9(5): 595-607, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19508176

ABSTRACT

KP772 is a new lanthanum complex containing three 1,10-phenathroline molecules. Recently, we have demonstrated that the promising in vitro and in vivo anticancer properties of KP772 are based on p53-independent G(0)G(1) arrest and apoptosis induction. A National Cancer Institute (NCI) screen revealed significant correlation of KP772 activity with that of the ribonucleotide reductase (RR) inhibitor hydroxyurea (HU). Consequently, this study aimed to investigate whether KP772 targets DNA synthesis in tumor cells by RR inhibition. Indeed, KP772 treatment led to significant reduction of cytidine incorporation paralleled by a decrease of deoxynucleoside triphosphate (dNTP) pools. This strongly indicates disruption of RR activity. Moreover, KP772 protected against oxidative stress, suggesting that this drug might interfere with RR by interaction with the tyrosyl radical in subunit R2. Additionally, several observations (e.g. increase of transferrin receptor expression and protective effect of iron preloading) indicate that KP772 interferes with cellular iron homeostasis. Accordingly, co-incubation of Fe(II) with KP772 led to generation of a coloured iron complex (Fe-KP772) in cell free systems. In electron paramagnetic resonance (EPR) measurements of mouse R2 subunits, KP772 disrupted the tyrosyl radical while Fe-KP772 had no significant effects. Moreover, coincubation of KP772 with iron-loaded R2 led to formation of Fe-KP772 suggesting chelation of RR-bound Fe(II). Summarizing, our data prove that KP772 inhibits RR by targeting the iron centre of the R2 subunit. As also Fe-KP772 as well as free lanthanum exert significant -though less pronounced- cytotoxic/static activities, additional mechanisms are likely to synergise with RR inhibition in the promising anticancer activity of KP772.


Subject(s)
Antineoplastic Agents/pharmacology , Dinucleoside Phosphates/metabolism , Organometallic Compounds/pharmacology , Phenanthrolines/pharmacology , Ribonucleotide Reductases/antagonists & inhibitors , Cell Line, Tumor , DNA/biosynthesis , Drug Synergism , Female , Humans , Hydroxyurea/pharmacology , Iron/metabolism , Iron Chelating Agents/pharmacology , Nucleotides/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Receptors, Transferrin/biosynthesis
6.
Biochem Pharmacol ; 73(12): 1873-86, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17445775

ABSTRACT

Recently, we have introduced [tris(1,10-phenanthroline)lanthanum(III)] trithiocyanate (KP772, FFC24) as a new lanthanum compound which has promising anticancer properties in vivo and in vitro. Aim of this study was to investigate the impact of ABC transporter-mediated multidrug resistance (MDR) on the anticancer activity of KP772. Here, we demonstrate that all MDR cell models investigated, overexpressing ABCB1 (P-glycoprotein), ABCC1 (multidrug resistance protein 1), or ABCG2 (breast cancer resistance protein) either due to drug selection or gene transfection, were significantly hypersensitive against KP772. Using ABCB1-overexpressing KBC-1 cells as MDR model, KP772 hypersensitivity was demonstrated to be based on stronger apoptosis induction and/or cell cycle arrest at unaltered cellular drug accumulation. KP772 did neither stimulate ABCB1 ATPase activity nor alter rhodamine 123 accumulation arguing against a direct interaction with ABCB1. Accordingly, several drug resistance modulators did not sensitize but rather protect MDR cells against KP772-induced cytotoxicity. Moreover, long-term KP772 treatment of KBC-1 cells at subtoxic concentrations led within 20 passages to a complete loss of drug resistance based on blocked MDR1 gene expression. When exposing parental KB-3-1 cells to subtoxic, stepwise increasing KP772 concentrations, we observed, in contrast to several other metallo-drugs, no acquisition of KP772 resistance. Summarizing, our data demonstrate that KP772 is hyperactive in MDR cells and might have chemosensitizing properties by blocking ABCB1 expression. Together with the disability of tumor cells to acquire KP772 resistance, our data suggest that KP772 should be especially active against notoriously drug-resistant tumor types and as second line treatment after standard chemotherapy failure.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple , Lanthanum/pharmacology , Organometallic Compounds/pharmacology , Phenanthrolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Carcinoma, Small Cell/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Formazans/metabolism , HL-60 Cells , Humans , Lanthanum/chemistry , Lanthanum/therapeutic use , Lung Neoplasms/drug therapy , Molecular Structure , Neoplasm Proteins/metabolism , Organic Anion Transporters/metabolism , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Phenanthrolines/chemistry , Phenanthrolines/therapeutic use , Sensitivity and Specificity , Tetrazolium Salts/metabolism
10.
Rev Physiol Biochem Pharmacol ; 153: 101-11, 2005.
Article in English | MEDLINE | ID: mdl-15674649

ABSTRACT

Cerium is a member of the lanthanide series or rare earth elements which exert diverse biological effects mainly by their resemblance to calcium. This similarity, which is particularly characteristic for the lighter members of the lanthanide series, enables these elements to replace calcium in biomolecules without necessarily substituting for it functionally. While the inhibitory effects on calcium-dependent physiological processes (such as those involved in the blood clotting cascade as well as in neuronal and muscular functions) are well-known, their relevance for the pharmacological properties of cerium are less clear. Historically, cerium oxalate was used as an antiemetic, especially in vomiting of pregnancy and kinetoses, although its mechanism of action has never been clarified. At present, cerium nitrate is available as an adjunct to silver sulfadiazine cream for the topical treatment of extensive burns not amenable to early wound excision. Apart from direct antiseptic effects, cerium helps to prevent postburn sepsis and systemic inflammatory response by fixing burn toxins. The antineoplastic potential of cerium compounds, which had fallen into oblivion, is currently being re-explored in experimental settings, though the mechanistic basis remains to be elucidated.


Subject(s)
Burns/drug therapy , Cerium , Neoplasms/drug therapy , Vomiting/drug therapy , Animals , Biological Availability , Burns/metabolism , Cerium/pharmacokinetics , Cerium/pharmacology , Cerium/therapeutic use , Humans , Neoplasms/metabolism , Vomiting/metabolism
11.
J Cancer Res Clin Oncol ; 131(2): 101-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15503135

ABSTRACT

PURPOSE: The ruthenium complex salt indazolium trans-[tetrachlorobisindazole-ruthenate(III)] (KP1019) and the analogous sodium salt KP1339 are effective tumor-inhibiting drugs in experimental therapy of autochthonous colorectal carcinomas in rats. This paper examines the cell biological mechanisms underlying their antineoplastic effects. METHODS: Colorectal tumor cell lines were used to analyze uptake of the ruthenium(III) complexes into the cells and the mechanism as well as the efficacy of their cytotoxic effects. RESULTS: KP1019 and KP1339 are efficiently taken up into the cells: 100 microM ruthenium(III) complex in the growth medium led to the uptake of 120-160 ng ruthenium per 10(6) cells within 30 min. Uptake of KP418 was tenfold lower correlating with its lower cytotoxic efficiency. KP1019 and KP1339 induced apoptosis in SW480 and HT29 cells predominantly by the intrinsic mitochondrial pathway as indicated by loss of mitochondrial membrane potential. Correspondingly sensitivity of the cells paralleled expression of bcl(2) while it was only slightly affected by mutations in Ki-ras. CONCLUSIONS: Our data demonstrate that trans-[tetrachlorobisindazole-ruthenate(III)] complex salts are promising candidate drugs in the second-line treatment of colorectal cancers resistant to other cytostatic drugs and has been introduced into phase I clinical trials.


Subject(s)
Apoptosis , Carcinoma/pathology , Colorectal Neoplasms/pathology , Indazoles/pharmacology , Organometallic Compounds/pharmacology , Humans , Indazoles/chemistry , Indazoles/pharmacokinetics , Mitochondria/physiology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Ruthenium , Salts , Tumor Cells, Cultured
12.
J Pharmacol Exp Ther ; 312(1): 281-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15331656

ABSTRACT

KP1019 [indazolium trans-[tetrachlorobis(1H-indazole)ruthenate (III)] (FFC14A) is a metal complex with promising anticancer activity. Since chemoresistance is a major obstacle in chemotherapy, this study investigated the influence of several drug resistance mechanisms on the anticancer activity of KP1019. Here we demonstrate that the cytotoxic effects of KP1019 are neither substantially hampered by overexpression of the drug resistance proteins multidrug resistance-related protein 1, breast cancer resistance protein, and lung resistance protein nor the transferrin receptor and only marginally by the cellular p53 status. In contrast, P-glycoprotein overexpression weakly but significantly (up to 2-fold) reduced KP1019 activity. P-glycoprotein-related resistance was based on reduced intracellular KP1019 accumulation and reversible by known P-glycoprotein modulators. KP1019 dose dependently inhibited ATPase activity of P-glycoprotein with a K(i) of approximately 31 microM. Furthermore, it potently blocked P-glycoprotein-mediated rhodamine 123 efflux under serum-free conditions (EC(50), approximately 8 microM), however, with reduced activity at increased serum concentrations (EC(50) at 10% serum, approximately 35 microM). Moreover, P-glycoprotein-mediated daunomycin resistance could only be marginally restored by KP1019 in serum-containing medium, also indicating an influence of serum proteins on the interaction between KP1019 and P-glycoprotein. Acquired KP1019 resistance was investigated by selecting KB-3-1 cells against KP1019 for more than 1 year. Only an approximately 2-fold KP1019 resistance could be induced, which unexpectedly was not due to overexpression of P-glycoprotein or other efflux pumps. Accordingly, KP1019-resistant cells did not display reduced drug accumulation. Their unique cross-resistance pattern confirmed an ABC transporter-independent resistance phenotype. In summary, the likeliness of acquiring insensitivity to KP1019 during therapy is expected to be low, and resistance should not be based on overexpression of drug efflux transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Indazoles/pharmacology , Ruthenium Compounds/pharmacology , Adenosine Triphosphatases/metabolism , Genes, MDR/physiology , HL-60 Cells , Humans , KB Cells , Organometallic Compounds , Receptors, Transferrin/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
13.
Curr Pharm Des ; 9(25): 2078-89, 2003.
Article in English | MEDLINE | ID: mdl-14529417

ABSTRACT

25 years after the first approval of cisplatin in the clinic against a number of cancer diseases, cisplatin and related compounds continue to be among the most efficient anticancer drugs used so far. Efforts are focused to develop novel platinum- and non-platinum-based antitumor drugs to improve clinical effectiveness, to reduce general toxicity and to broaden the spectrum of activity. In the field of non-platinum compounds exhibiting anticancer properties, ruthenium complexes are very promising, showing activity on tumors which developed resistance to cisplatin or in which cisplatin is inactive. Furthermore, general toxicity was found to be very low. The first ruthenium compound NAMI-A entered phase I clinical trials in 1999 as an antimetastatic drug, whereas the ruthenium complex KP1019 will enter phase I clinical trials in 2003 as an anticancer drug which is among others very active against colon carcinomas and their metastases. Remarkable progress is also seen in developing tumor inhibiting gallium compounds. One of them, KP46, will also enter phase I clinical trials in 2003. This article reviews briefly the achievements in the field of anticancer metal complexes focusing the discussion onto the impact of the group of Bioinorganic Chemistry at the Department of Inorganic Chemistry at the University of Vienna. The development of pH sensitive platinum prodrugs, platinum-based drug targeting strategies with low-molecular-weight carriers, kinetically inert platinum(IV) complexes, as well as tumor inhibiting non-platinum anticancer drugs based on ruthenium and gallium is covered in the following sections.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems/trends , Metals, Heavy/chemistry , Metals, Heavy/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Humans , Ligands
14.
Article in English | MEDLINE | ID: mdl-12605304

ABSTRACT

Thirty years after the onset of the first clinical studies with cisplatin, the development of antineoplastic platinum drugs continues to be a productive field of research. This article reviews the current preclinical and clinical status, including a discussion of the molecular basis for the activity of the parent drug cisplatin and platinum drugs of the second and third generation, in particular their interaction with DNA. Further emphasis is laid on the development of third generation platinum drugs with activity in cisplatin-resistant tumours, particularly on chelates containing 1,2-diaminocyclohexane (DACH) and on the promising and more recently evolving field of non-classic ( trans- and multinuclear) platinum complexes. The development of oral platinum drugs and drug targeting strategies using liposomes, polymers or low-molecular-weight carriers in order to improve the therapeutic index of platinum chemotherapy are also covered.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Platinum Compounds/therapeutic use , Animals , Drug Design , Humans , Models, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...