Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Chemotherapy ; 53(2): 148-52, 2007.
Article in English | MEDLINE | ID: mdl-17308381

ABSTRACT

The reaction of platinum(II) [Pt(II)] or palladium(II) [Pd(II)] with 2-acetyl pyridine 4N-ethyl thiosemicarbazone, HAc4Et (1) results in the complexes [Pt(Ac4Et)(2)] (2) and [Pd(Ac4Et)(2)] (3). In a panel of human tumor cell lines of different origins (breast, colon, and ovary cancers), and containing also cisplatin-refractory/resistant cell lines, the in vitro growth inhibitory effect of 1-3 was compared to that of cisplatin by using the sulforodamine B assay. After a 96-hour continuous treatment, both the thiosemicarbazone HAc4Et and the metal compounds [Pt(Ac4Et)(2)] and [Pd(Ac4Et)(2)] exhibit very remarkable growth inhibitory activities with mean IC(50) values of 0.9 nM (0.22-2.47 nM), 0.7 nM (0.15-2 nM) and 0.5 nM (0.17-1.02 nM), respectively. In contrast, cisplatin shows a markedly lower growth inhibitory potency, the mean IC(50) in the panel being 2.8 muM (0.2-8 muM). In addition to their major cell growth inhibitory potency, complexes 1-3 are characterized by a growth inhibitory profile different from that of cisplatin, being active towards cisplatin-refractory tumor cell lines. These findings, along with the ability of completely overcoming acquired cisplatin resistance from either multifocal or reduced uptake origin, confirm the antitumor potential of HAc4Et and support the hypothesis that both [Pt(Ac4Et)(2)] and [Pd(Ac4Et)(2)] complexes can be characterized by cellular pharmacological properties distinctly different from those of cisplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Organometallic Compounds/pharmacology , Thiosemicarbazones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Palladium/chemistry , Platinum/chemistry , Thiosemicarbazones/chemistry
2.
Br J Cancer ; 86(6): 993-8, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11953835

ABSTRACT

NAMI-A is a ruthenium-based compound with selective anti-metastasis activity in experimental models of solid tumours. We studied whether this activity was dependent on anti-angiogenic ability of NAMI-A. We thus investigated its in vitro effects on endothelial cell functions necessary for angiogenesis to develop, as well as its in vivo effects in the chick embryo chorioallantoic membrane model. Endothelial cell proliferation, chemotaxis, and secretion of the matrix-degrading enzyme metalloproteinase-2 were inhibited by NAMI-A in a dose-dependent manner, and without morphologic signs of cell apoptosis or necrosis. Lastly, NAMI-A displayed a dose-dependent in vivo anti-angiogenic activity in the chorioallantoic membrane model. These data suggest that the anti-angiogenic activity of NAMI-A can contribute to its anti-metastatic efficacy in mice bearing malignant solid tumours.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Endothelium, Vascular/drug effects , Neoplasm Metastasis/prevention & control , Organometallic Compounds/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Endothelium, Vascular/cytology , Humans , Matrix Metalloproteinase Inhibitors , Ruthenium Compounds
3.
Mol Pharmacol ; 58(6): 1525-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11093793

ABSTRACT

To investigate the modifications of antitumor activity and DNA binding mode of transplatin after replacement of one nonleaving group NH(3) by an iminoether group, trans-[PtCl(2)(Z-HN=C(OMe)Me)(NH(3)] and trans-[PtCl(2)(E-HN=C(OMe)Me)(NH(3)] complexes (differing in the Z or E configuration of iminoether, and abbreviated mixed Z and mixed E, respectively), have been synthesized. In a panel of human tumor cell lines, both mixed Z and mixed E show a cytotoxic potency higher than that of transplatin, the mean IC(50) values being 103, 37, and 215 microM, respectively. In vivo mixed Z is more active and less toxic than mixed E in murine P388 leukemia and retains its efficacy against SK-OV-3 human cancer cell xenograft in nude mice. In the reaction with naked DNA, mixed Z forms monofunctional adducts that do not evolve into intrastrand cross-links but close slowly into interstrand cross-links between complementary guanine and cytosine residues. The monofunctional mixed Z adducts are removed by thiourea and glutathione. The interstrand cross-links behave as hinge joints, increasing the flexibility of DNA double helix. The mixed Z, transplatin, and cisplatin interstrand cross-links, as well as mixed Z monofunctional adducts are not specifically recognized by HMG1 protein, which was confirmed to be able to specifically recognize cisplatin d(GpG) intrastrand cross-links. These data demonstrate that the DNA interaction properties of the antitumor-active mixed Z are very similar to those of transplatin, thus suggesting that clinical inactivity of transplatin could not depend upon its peculiar DNA binding mode.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cisplatin/chemistry , Cisplatin/metabolism , Cisplatin/therapeutic use , Cross-Linking Reagents/metabolism , DNA/chemistry , DNA/drug effects , DNA/metabolism , DNA Adducts/drug effects , DNA Footprinting , DNA Restriction Enzymes/metabolism , DNA-Directed RNA Polymerases/metabolism , Disease Models, Animal , Drug Screening Assays, Antitumor , High Mobility Group Proteins/metabolism , Humans , Leukemia P388/drug therapy , Mice , Nucleic Acid Conformation , Oligonucleotides/metabolism , Rats , Thiourea/pharmacology , Tumor Cells, Cultured
4.
J Inorg Biochem ; 78(4): 347-54, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10857916

ABSTRACT

The reactions of Na2PtCl4 with pyridine-2-carbaldehyde and 2-acetyl pyridine N(4)-ethyl-thiosemicarbazones, HFo4Et and HAc4Et respectively, afforded the complexes [Pt(Fo4Et)Cl], [Pt(HFo4Et)2]Cl2, [Pt(Fo4Et)2] and [Pt(Ac4Et)Cl], [Pt(HAc4Et)2]Cl2 x 2H2O, [Pt(Ac4Et)2]. The new complexes have been characterized by elemental analyses and spectroscopic studies. The crystal structure of the complex [Pt(Ac4Et)Cl] has been solved. The anion of Ac4E coordinates in a planar conformation to the central platinum(II) through the pyridyl N, azomethine N and thiolato S atoms. Intermolecular hydrogen, non-hydrogen bonds, pi-pi and weak Pt-pi contacts lead to aggregation and a supramolecular assembly. The cytotoxic activity for the platinum(II) complexes in comparison to that of cisplatin and thiosemicarbazones was evaluated in a pair of cisplatin-sensitive and -resistant ovarian cancer cell lines A2780 and A2780/Cp8. The platinum(II) complexes showed a cytotoxic potency in a very low micromolar range and were found able to overcome the cisplatin resistance of A2780/Cp8 cells.


Subject(s)
Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Ligands , Magnetic Resonance Spectroscopy , Models, Chemical , Models, Molecular , Spectrophotometry, Infrared , Spectroscopy, Fourier Transform Infrared , Thiosemicarbazones/chemistry , Tumor Cells, Cultured
5.
J Inorg Biochem ; 79(1-4): 173-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10830863

ABSTRACT

A novel class of dianionic Ru(III) dimers of formula Na2[[trans-RuCl4(Me2SO)]2(mu-L)], with L = pyrazine (pyz, 1), pyrimidine (pym, 2), 4,4'-bipyridine (bipy, 3), and 1,2-bis(4-pyridine) ethane (etbipy, 4), was developed by us with the specific aim of assessing their antitumor properties. The dimers are in fact structurally related to the antimetastatic mononuclear compound (ImH) [trans-RuCl4(Me2SO)(Im)] (NAMI-A, Im = imidazole). Preliminary results concerning the antineoplastic activity of 1-4 against the murine MCa carcinoma model, a tumor which spontaneously metastasizes in the lungs, are reported. Similarly to what is normally observed with NAMI-A, the treatment with the dimeric complexes was scarcely effective against the growth of the primary tumor. However, dimers 1, 2, and 4 reduced very effectively the number and, in particular, the weight of lung metastases (to about 5% with respect to controls); in particular, Na2[[trans-RuCl4(Me2SO)]2(mu-etbipy)] (4) was as effective as NAMI-A in reducing the spontaneous metastases at a dosage which, in terms of moles of ruthenium, is about 3.5 times lower compared to that normally used for NAMI-A. Furthermore, in vitro tests showed that dimers 1-4 are capable of forming interstrand cross-links with linearized plasmidic DNA in a time-dependent manner. All the dimeric species are more active in inducing cross-links compared to NAMI-A, and the dimer bridged by the etbipy ligand (4) is the most effective among those tested.


Subject(s)
Antineoplastic Agents/chemistry , DNA/chemistry , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Organometallic Compounds/chemistry , Ruthenium , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Dimerization , Female , Ligands , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred CBA , Organometallic Compounds/chemical synthesis , Organometallic Compounds/therapeutic use
6.
Met Based Drugs ; 7(1): 23-32, 2000.
Article in English | MEDLINE | ID: mdl-18475920

ABSTRACT

The single-stranded oligonucleotide 5'-d(CCTCGCTCTC) (I) was reacted with the antitumor trans platinum iminoderivative trans-[PtCl(2){E-HN = C(OMe)Me}(2)] (trans-EE) and subsequently annealed with its complementary strand 5'-d(GAGAGCGAGG) (II). The platinated duplex was characterized by 1D and 2D proton NMR spectroscopy at 600 MHz. In agreement with previous studies by different techniques trans-EE was found to form a monofunctional adduct with the duplex involving the guanine residue. The modification by trans-EE has been found to induce only minor local distortion in the duplex geometry. Two key crosspeaks observed in the NOESY map corresponding to a close contact between G5-H8 and the methoxy and the methyl group, respectively, enabled us to dock the trans-EE complex with the duplex by geometry optimization. The results support the idea that the antitumor activity of trans-EE is related to lesion of DNA fundamentally different from that of cisplatin. Unexpectedly, the NOESY spectra indicated that at the high NaCl concentration used (0.2 M) the duplex was found to undergo slow deplatination. This was subsequently proved by HPLC. In a separate experiment on platination of the single strand in a salt free environment the HPLC analysis showed that the monofunctional adduct was not deplatinated, however, after 24 hours, additidnal minor isomers were detected.

7.
Int J Oncol ; 15(5): 1039-44, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10536190

ABSTRACT

The platinum complex trans-[PtCl2¿E-HN=C(OMe)Me¿2] was compared to cisplatin for cytotoxicity towards tumour cells, and for cellular pharmacological properties in A2780 and cisplatin-resistant A2780/Cp8 ovarian cancer cells. Trans-[PtCl2¿E-HN=C(OMe)Me¿2] was comparably cytotoxic to cisplatin (mean IC50 after 72 h exposure = 6. 1 microM and 7 microM, respectively) and did not show cross-resistance in A2780/Cp8 cells (resistance factor = 0.9). Cellular accumulation measurements after treatment with equimolar drug concentrations showed that trans-[PtCl2¿E-HN=C(OMe)Me¿2] entered both A2780 and A2780/Cp8 cells much more efficiently than cisplatin, whose accumulation was reduced in A2780/Cp8 cells. Unlike cisplatin, trans-[PtCl2¿E-HN=C(OMe)Me¿2] induced rapidly cell death and cell cycle modifications of treated cells, thus indicating substantially different mechanistic properties.


Subject(s)
Antineoplastic Agents/toxicity , Organometallic Compounds/toxicity , Biological Transport , Cell Cycle/drug effects , Cell Division/drug effects , Cisplatin/pharmacokinetics , Cisplatin/toxicity , Drug Resistance, Neoplasm , Female , Humans , Organometallic Compounds/pharmacokinetics , Ovarian Neoplasms , Platinum/pharmacokinetics , Platinum/toxicity , Tumor Cells, Cultured
8.
Anticancer Drug Des ; 14(3): 253-64, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10500500

ABSTRACT

The platinum-iminoether complexes trans-[PtCl2[E - HN = C(OEt)Me]2] (1) and trans-[PtCl2[Z - HN = C(OEt)Me[2] (2), differing in the configuration of the iminoether ligands, were investigated for cytotoxicity towards human tumor cell lines, the involvement of DNA as a cytotoxic target, and their DNA binding mode. The cytotoxicity of isomer 1 was comparable to that of cisplatin, whereas isomer 2 was slightly less active. Excision-repair-deficient xeroderma pigmentosum group A cells were four times more sensitive to both isomers than normal cells, thus implicating cellular DNA as the cytotoxic target. Replication mapping experiments showed that both isomers interact preferentially with guanine residues at py-G-py sites. Oligodeoxyribonucleotides containing unique N7-guanine monofunctional adducts of the more cytotoxic isomer 1 were prepared and investigated for chemical reactivity, stability and DNA conformational alterations. The results showed that the ability of thiourea to labilize the monofunctional adducts depends upon the DNA secondary structure, but not upon the sequence context. Monofunctional adducts evolve to bidentate adducts in single-stranded oligonucleotides, but they are stable in double-stranded oligonucleotides and produce conformational distortions selectively located at the 5'-adjacent base pair. This study gives new insight into the mechanism of action of trans platinum-iminoether complexes, enabling for the first time comparison between different ligand isomers.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , DNA, Neoplasm/metabolism , Organoplatinum Compounds/metabolism , Organoplatinum Compounds/toxicity , Cell Division/drug effects , DNA Adducts/metabolism , DNA Footprinting , DNA, Single-Stranded/metabolism , Drug Stability , Female , Growth Inhibitors/metabolism , Growth Inhibitors/pharmacology , Humans , Ligands , Nucleic Acid Conformation , Oligonucleotides/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Stereoisomerism , Thiourea/pharmacology , Tumor Cells, Cultured/drug effects
9.
J Inorg Biochem ; 77(1-2): 31-5, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10626350

ABSTRACT

In order to widen our knowledge on antitumour trans-[PtCl2(iminoether)2] complexes, we have synthesised two new derivatives, trans-[PtCl2¿E-HN = C(OEt)Me¿2] (1) and trans-[PtCl2¿Z-HN = C(OEt)Me¿2] (2), which differ in the configuration of the iminoether ligands. Isomer 1 showed an in vitro cytotoxicity similar to that of cisplatin in a panel of human tumour cell lines (mean IC50 = 8 and 7.7 microM, respectively), whereas isomer 2 showed a lower activity (IC50 = 14.3 microM). Both 1 and 2 isomers overcame cisplatin resistance of ovarian cancer cell line A2780/Cp8. In agreement with the n-octanol/saline partition ratios, intracellular platinum content (and DNA platination) after a 2-h exposure to equimolar drug concentrations was in the order 1 > 2 >> cisplatin, thus indicating that substitution of imminoethers for ammines determines a major lipophilicity and cellular uptake of the platinum drug. Both 1 and 2 showed a major toxic effect towards an excision repair-defective Drosophila strain, thus indicating cellular DNA as cytotoxic target. Finally, both 1 and 2 were active in vivo against the murine P388 system, but, contrary to the in vitro activity, isomer 2 was slightly more active than 1. On the whole, the results confirm the antitumour activity of trans-[PtCl2(iminoether)2] complexes, and indicate that the configuration of the iminoether ligands may affect the pharmacological properties of this class of complexes.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA, Neoplasm/drug effects , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Animals , Carcinoma/drug therapy , Carcinoma/genetics , Cell Division/drug effects , Cisplatin/pharmacology , DNA Repair/drug effects , DNA Repair/genetics , Dose-Response Relationship, Drug , Drosophila/genetics , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , Imines/chemistry , Leukemia P388/drug therapy , Mice , Mice, Inbred Strains , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Mol Pharmacol ; 52(3): 354-61, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9281596

ABSTRACT

Recent findings that an analogue of clinically ineffective transplatin, trans-[PtCl2(E-iminoether)2], exhibits antitumor activity has helped reevaluation of the empirical structure-antitumor activity relationship generally accepted for platinum(II) complexes. According to this relationship, only the cis geometry of leaving ligands in the bifunctional platinum(II) complexes, should be therapeutically active. Global modifications of natural DNAs in cell-free media by trans-[PtCl2(E-iminoether)2] were studied through various molecular biophysical methods and compared with modifications by cis-[PtCl2(E-iminoether)2], transplatin, cisplatin, and monofunctional chlorodiethylenetriamineplatinum(II) chloride. Thus, the results of this study have extended our recent finding, indicating that the prevalent lesion occurring in double-helical DNA on its modification by trans-[PtCl2(E-iminoether)2] is a monofunctional adduct at guanine residues. The modification by trans-[PtCl2(E-iminoether)2] has been found to induce local distortions in DNA, which have a character differing fundamentally from those induced by both clinically ineffective or antitumor platinum complexes tested in this study. The different character of alterations induced in DNA by the adducts of trans-[PtCl2(E-iminoether)2] and transplatin has been suggested to be relevant to the unexpected observation that the new complex with leaving chloride groups in trans position exhibits antitumor efficacy. In addition, the results support the idea that platinum drugs that bind to DNA in a manner fundamentally different from that of cisplatin can exhibit altered biological properties, including differing spectra and intensities of antitumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/drug effects , Nucleic Acid Conformation/drug effects , Organometallic Compounds/pharmacology , Animals , Cattle , Chemical Phenomena , Chemistry, Physical , Cisplatin/pharmacology , DNA/chemistry , DNA/metabolism , DNA, Superhelical/drug effects , DNA, Superhelical/metabolism , Drug Stability , Stereoisomerism , Substrate Specificity , Transcription, Genetic/drug effects
12.
Nucleic Acids Res ; 24(2): 336-41, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8628659

ABSTRACT

It has been shown recently that some analogues of clinically ineffective trans-diamminedichloroplatinum (II) (transplatin) exhibit antitumor activity. This finding has inverted the empirical structure-antitumor activity relationships delineated for platinum(II) complexes, according to which only the cis geometry of leaving ligands in the bifunctional platinum complexes is therapeutically active. As a result, interactions of trans platinum compounds with DNA, which is the main pharmacological target of platinum anticancer drugs, are of great interest. The present paper describes the DNA binding of antitumor trans-[PtCl(2)(E-imino ether)(2)] complex (trans-EE) in a cell-free medium, which has been investigated using three experimental approaches. They involve thiourea as a probe of monofunctional DNA adducts of platinum (II) complexes with two leaving ligands in the trans configuration, ethidium bromide as a probe for distinguishing between monofunctional and bifunctional DNA adducts of platinum complexes and HPLC analysis of the platinated DNA enzymatically digested to nucleosides. The results show that bifunctional trans-EE preferentially forms monofunctional adducts at guanine residues in double-helical DNA even when DNA is incubated with the platinum complex for a relatively long time (48 h at 37 degrees C in 10 mM NaCIO(4). It implies that antitumor trans-EE modifies DNA in a different way than clinically ineffective transplatin, which forms prevalent amount of bifunctional DNA adducts after 48 h. This result has been interpreted to mean that the major adduct of trans-EE, occurring in DNA even after long reaction times, is a monofunctional adduct in which the reactivity of the second leaving group is markedly reduced. It has been suggested that the different properties of the adducts formed on DNA by transplatin and trans-EE are relevant to their distinct clinical efficacy.


Subject(s)
DNA Adducts/metabolism , Organoplatinum Compounds/metabolism , Animals , Cattle , Cisplatin/analogs & derivatives , Cisplatin/metabolism , DNA/chemistry , DNA/metabolism , DNA Adducts/chemical synthesis , Deoxyguanosine/chemistry , Ethidium , Fluorescent Dyes , Molecular Probes , Nucleic Acid Denaturation , Organoplatinum Compounds/chemical synthesis , Structure-Activity Relationship , Thiourea
13.
Chem Biol Interact ; 98(3): 251-66, 1995 Dec 22.
Article in English | MEDLINE | ID: mdl-8548863

ABSTRACT

The biological activity of cis and trans complexes of formula [PtCl2(HN = C(OMe)Me)2] has been investigated. The iminoether ligands can have either E or Z configuration about the C = N double bond, therefore EE, EZ and ZZ isomers are obtainable. Substitution of iminoether with EE configuration for amine leads to unexpectedly high antitumor activity for the complex with trans geometry which turns out to be more active than the cis congener in the P388 leukaemia system. The same trans-EE complex shows an activity comparable to that of cisplatin in reducing the primary tumour mass and lung metastases in mice bearing Lewis lung carcinoma, thus representing a trans platinum complex active on both limphoproliferative and solid metastasizing murine tumours. Also the cytotoxicity, the inhibition of DNA synthesis and the mutagenic activity, which are greater for the cis- with respect to the trans-isomer in the amine complexes, are instead greater for the trans- than for the cis- isomer in the case of iminoether compounds. Binding to calf thymus DNA is slower for iminoether complexes than it is for amine complexes, however after 24 h reaction time the level of binding is similar for both types of complexes. Trans-EE, like trans-DDP, does not give the DNA conformational alterations (terbium fluorescence) typical of antitumour-active cis- platinum compounds, but, under strictly analogous experimental conditions, shows a greatly reduced DNA interstrand cross-linking ability (heat denaturation/renaturation assay) with respect to either trans-DDP or cis-EE and cis-DDP. The data in hand point to a new trans platinum antitumour complex with a mechanism of action different from that of cis-DDP and classical analogues.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/drug therapy , Leukemia P388/drug therapy , Platinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Lewis Lung/pathology , Cell Count/drug effects , Cell Division/drug effects , Cisplatin/pharmacology , Cross-Linking Reagents , DNA/drug effects , DNA/metabolism , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , Leukemia P388/pathology , Mice , Mutagenicity Tests , Mutagens , Platinum Compounds/chemical synthesis , Platinum Compounds/chemistry , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Met Based Drugs ; 2(4): 195-9, 1995.
Article in English | MEDLINE | ID: mdl-18472766

ABSTRACT

The combinational treatment between the selective antimetastatic agent, sodium-trans-rutheniumtetrachloridedimethylsulfoxideimidazole, Na[trans-RuCl(4)(DMSO)Im], and the cytotoxic drug 5-fluorouracil (5-FU) on primary tumor growth and on the survival time of experimental tumors results in an effect significantly greater than that of each single agent used alone either with the solid metastasizing MCa mammary carcinoma of the CBA mouse or with the lymphocytic leukemia P388 and its platinum resistant P388/DDP subline. Thus the inorganic compound Na[trans-RuCl(4)(DMSO)Im], known for its potent and selective antimetastatic effects, positively interacts with the antitumor action of an organic anticancer agent such as 5-FU on both a solid metastasizing tumor and a tumor of lymphoproliferative type. In particular, the effects of the combinational treatment on the survival time of tumor bearing mice seem to be related to the selective antimetastatic activity of the ruthenium complex that joins the potent cytotoxicity of 5-FU for the tumor. Moreover, these data show that Na[trans-RuCl(4)(DMSO)Im] is almost as effective on the subline of P388 made resistant to cisplatin as it was on the parental line.

15.
Met Based Drugs ; 2(5): 249-56, 1995.
Article in English | MEDLINE | ID: mdl-18472776

ABSTRACT

A platinum(II) complex with the antiviral drug acyclovir was synthesized and its antiviral and anticancer properties were investigated in comparison to those of acyclovir and cisplatin. The platinum-acyclovir complex maintained the antiviral activity of the parent drug acyclovir, though showing a minor efficacy on a molar basis (ID(50) = 7.85 and 1.02 muMu for platinum-acyclovir and cisplatin, respectively). As anticancer agent, the platinum-acyclovir complex was markedly less potent than cisplatin on a mole-equivalent basis, but it was as effective as cisplatin when equitoxic dosages were administered in vivo to P388 leukaemia-bearing mice (%T/C = 209 and 211 for platinum-acyclovir and cisplatin, respectively). The platinum-acyclovir complex was also active against a cisplatin-resistant subline of the P388 leukaemia (%T/C = 140), thus suggesting a different mechanism of action. The DNA interaction properties (sequence specificity and interstrand cross-linking ability) of platinum-acyclovir were also investigated in comparison to those of cisplatin and [Pt(dien)Cl](+), an antitumour-inactive platinum-triamine compound. The results of this study point to a potential new drug endowed, at the same time, with antiviral and anticancer activity and characterized by DNA interaction properties different from those of cisplatin.

17.
Met Based Drugs ; 1(1): 41-63, 1994.
Article in English | MEDLINE | ID: mdl-18476216

ABSTRACT

In this paper we report a review of the results obtained in the last few years by our group in the development of ruthenium(III) complexes characterized by the presence of sulfoxide ligands and endowed with antitumor properties. In particular, we will focus on ruthenates of general formula Na[trans-RuCl(4)(R(1)R(2)SO)(L)], where R(1)R(2)SO = dimethylsulfoxide (DMSO) or tetramethylenesulfoxide (TMSO) and L = nitrogen donor ligand. The chemical behavior of these complexes has been studied by means of spectroscopic techniques both in slightly acidic distilled water and in phosphate buffered solution at physiological pH. The influence of biological reductants on the chemical behavior is also described. The antitumor properties have been investigated on a number of experimental tumors. Out of the effects observed, notheworthy appears the capability of the tested ruthenates to control the metastatic dissemination of solid metastasizing tumors. The analysis of the antimetastatic action, made in particular on the MCa mammary carcinoma of CBA mouse, has demonstrated a therapeutic value for these complexes which are able to significantly prolong the survival time of the treated animals. The antimetastatic effect is not attributable to a specific cytotoxicity for metastatic tumor cells although in vitro experiments on pBR322 double stranded DNA has shown that the test ruthenates bind to the macromolecule, causing breaks corresponding to almost all bases, except than thymine, and are able to cause interstrand bonds, depending on the nature of the complex being tested, some of which results active as cisplatin itself.

19.
Eur J Cancer ; 29A(13): 1873-9, 1993.
Article in English | MEDLINE | ID: mdl-8260245

ABSTRACT

Two ruthenium(II) complexes, characterised by the presence of dimethylsulphoxide ligands, were investigated in comparison to cisplatin on mouse P388 leukaemia and on a subline made resistant to cisplatin (P388/DDP). Both cis- and trans-RuCl2(DMSO)4 significantly prolonged the survival time of leukaemic mice, independently of the tumour line used. Unlike cisplatin, the prolongation of life-span of tumour-bearing hosts caused by ruthenium complexes was not supported by a parallel inhibition of the number of tumour cells in the treated hosts, as evidenced by tumour cell count in the peritoneal cavity and by vivo-vivo bioassays of blood samples and of whole brains. Thus, cis- and trans-RuCl2(DMSO)4 appear capable of preventing leukaemic spread into the central nervous system also when the number of tumour cells in the peritoneal cavity and in the blood stream is as high as in untreated controls. When the drug-induced DNA damage was investigated by modifying double stranded DNA and identifying the lesions able to inhibit DNA synthesis in vitro, trans-RuCl2(DMSO)4 and, to a lesser extent, cis-RuCl2(DMSO)4 formed blocking lesions at the same sites of cisplatin; nevertheless, the mechanism of antitumour activity of ruthenium complexes appears to be different from that of cisplatin for the absence of any relationship between cytotoxicity and prevention of leukaemic dissemination into the central nervous system. These data indicate that the activity of cis- and trans-RuCl2(DMSO)4 on the P388 leukaemia is characterised by the lack of cross-resistance with cisplatin and by the alteration of the metastasising behaviour of leukaemic cells which lose their natural capacity to invade the central nervous system.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/prevention & control , Leukemia P388/drug therapy , Organometallic Compounds/therapeutic use , Animals , Brain Neoplasms/secondary , Cisplatin/therapeutic use , DNA, Neoplasm/drug effects , Drug Resistance , Leukemia P388/mortality , Male , Mice , Mice, Inbred Strains , Ruthenium , Tumor Cells, Cultured/drug effects
20.
Int J Oncol ; 2(4): 527-9, 1993 Apr.
Article in English | MEDLINE | ID: mdl-21573587

ABSTRACT

Some Ru(II)-DMSO complexes have antimetastatic properties in experimental tumors. Since plasminogen activators are thought to play an important role in the expression of cancer cell metastatic capacity, we evaluated the effect of two Ru(II)-DMSO complexes on the fibrinolytic activity of Lewis lung carcinoma. Tumor-bearing mice were given daily, for 14 days, an i.p. injection of antimetastatic dosages of cis-RuCl2(DMSO)4 (700 mg/kg/die) or trans-RUCl2(DMSO)4 (37 mg/kg/die), or vehicle. Tumor extracts obtained on day 15 from treatment groups had significantly lower (plasminogen-dependent) fibrinolytic activity than extracts from control animals (p<0.001). Urokinase inhibitor activity in tumor extracts did not differ among groups and did not correlate with plasminogen activator activity, Fibrin autography of control tumor extracts revealed the presence of a main fibrinolytic band co-migrating with urinary plasminogen activator (urokinase-type) and of minor bands with a higher molecular weight. In samples from animals treated with either Ru(II)-DMSO complex the most striking finding was a reduction of the band corresponding to free urokinase. These findings suggest that ruthenium complexes decrease the fibrinolytic activity of tumor cells by reducing urokinase production rather than by enhancing inhibitor production. Treatment of tumor-bearing mice with cis-RuCl2(DMSO)4 at a dosage equimolar to the trans isomer, neither reduced metastasis formation nor decreased plasminogen activator activity of tumor extracts. The depression of tumor-associated proteolytic activity could contribute to the antimetastatic properties of ruthenium complexes.

SELECTION OF CITATIONS
SEARCH DETAIL
...