Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Dalton Trans ; 44(4): 1905-13, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25489765

RESUMO

NAMI-A is a ruthenium-based drug endowed with the unique property of selectively targeting solid tumour metastases. Although two clinical studies had already been completed, limited information exists on the behavior of NAMI-A after injection into the bloodstream. PK data in humans informs us of a rather low free drug concentration, of a relatively high half-life time of elimination and of a linear relationship between the administered dose and the corresponding AUC for up to toxic doses. In the present study, we examined the chemical kinetics of albumin binding with or without the presence of reducing agents, and we evaluated how these chemical aspects might influence the in vivo PK and the in vitro ability of NAMI-A to inhibit cell migration, which is a bona fide, rapid and easy way to suggest anti-metastatic properties. The experimental data support the binding of NAMI-A to serum albumin. The reaction is facilitated when the drug is in its reduced form and, in agreement with already reported data, the adduct formed with albumin maintains the biological activity of the ruthenium drug. The formation of the adduct is favored by low ratios of NAMI-A : HSA and by the reduction of the drug with ascorbic acid. The difference in in vivo PK and the faster binding to albumin of the reduced NAMI-A seem to suggest that the drug is not rapidly reduced immediately upon injection, even at low doses. Most probably, cell and protein binding prevail over the reduction of the drug. This observation supports the thesis that the reduction of the drug before injection must be considered relevant for the pharmacological activity of NAMI-A against tumour metastases.


Assuntos
Antineoplásicos , Dimetil Sulfóxido/análogos & derivados , Compostos Organometálicos , Albumina Sérica/química , Albumina Sérica/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Ácido Ascórbico/química , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dimetil Sulfóxido/química , Dimetil Sulfóxido/farmacocinética , Dimetil Sulfóxido/farmacologia , Humanos , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos ICR , Compostos Organometálicos/química , Compostos Organometálicos/farmacocinética , Compostos Organometálicos/farmacologia , Oxirredução , Rodaminas/metabolismo , Rutênio/sangue , Rutênio/metabolismo , Compostos de Rutênio
2.
Eur J Cancer ; 38(3): 427-35, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11818210

RESUMO

The influence of chemical stability on the antimetastatic ruthenium(III) compound imidazolium trans-imidazoletetrachlorodimethylsulphoxideruthenium(III) (NAMI-A) in aqueous solution was studied both in vitro and in vivo. The loss of dimethyl-sulphoxide (DMSO) ligand from the compound was tested by using a NAMI-A solution acidified with HCl at pH 3.0 and aged for 0, 4, 8 and 24 h prior to intraperitoneal (i.p.) injection into CBA mice bearing advanced MCa mammary carcinoma. The activity of NAMI-A on lung metastases showed no change even after the loss of DMSO ligand from up to 50% of the molecules. The reduction of NAMI-A did not modify the number of KB cells blocked in the S+G2M phases, independent of whether the reduction occurred outside the cells or after loading the cells with the compound prior to treatment with the reductants (ascorbic acid, glutathione or cysteine). In vivo, the complete reduction of NAMI-A with equivalent amounts of ascorbic acid, glutathione or cysteine prior to administration to mice bearing advanced MCa mammary carcinoma was more active than NAMI-A alone. The data show that NAMI-A, although undergoing a series of chemical modifications, maintains its antimetastatic activity in a broad range of experimental conditions.


Assuntos
Antineoplásicos/química , Dimetil Sulfóxido/análogos & derivados , Dimetil Sulfóxido/química , Neoplasias Mamárias Experimentais/tratamento farmacológico , Compostos Organometálicos/química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Divisão Celular , Dimetil Sulfóxido/metabolismo , Dimetil Sulfóxido/farmacocinética , Dimetil Sulfóxido/uso terapêutico , Feminino , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Compostos Organometálicos/farmacocinética , Compostos Organometálicos/uso terapêutico , Fase S , Células Tumorais Cultivadas
3.
Anticancer Res ; 20(1A): 197-202, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10769655

RESUMO

The content of ruthenium in blood and different organs of healthy CBA mice was determined by AAS after single i.v. treatment of 200 mg kg-1 of NAMI-A, a new antimetastatic ruthenium compound. Ruthenium concentration in blood falls 5 min after i.v. treatment. In the kidney, ruthenium concentration is markedly higher than in any other analysed tissue. No ruthenium was detected in brains. Pharmacokinetic parameters for a mono- or a bi-compartment model are identifiable: t1/2 is 10.45 h vs 12.02 (t1/2 alpha 0.023 h + t1/2 beta 12 h) with Cltot of 1.60 ml*h-1 vs 1.59); Vd is 24.15 vs 27.48 ml and (model dependent) AUC is 689 vs 694 mg*L-1*h. AUC(0-->infinity) calculated by noncompartmental method (linear trapezoidal rule) is 719.77 mg*L-1*h. NAMI-A is rapidly cleared from the blood compartment immediately after i.v. administration. Apparently, there is no differential accumulation of ruthenium in the lungs which might account for a selective antimetastatic effect caused by a cytotoxic concentration in this site, nor in any other specific organ examined.


Assuntos
Antineoplásicos/farmacocinética , Dimetil Sulfóxido/análogos & derivados , Metástase Neoplásica/prevenção & controle , Compostos Organometálicos/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/sangue , Antineoplásicos/toxicidade , Área Sob a Curva , Compartimentos de Líquidos Corporais , Dimetil Sulfóxido/administração & dosagem , Dimetil Sulfóxido/sangue , Dimetil Sulfóxido/farmacocinética , Dimetil Sulfóxido/toxicidade , Injeções Intravenosas , Nefropatias/induzido quimicamente , Camundongos , Camundongos Endogâmicos CBA , Modelos Biológicos , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/sangue , Compostos Organometálicos/toxicidade , Compostos de Rutênio , Distribuição Tecidual
4.
Anticancer Res ; 19(2A): 969-72, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10368640

RESUMO

The effects of NAMI-A, a novel ruthenium compound endowed with selective antimetastatic action, were tested on solid metastasising tumours of the mouse in comparison to cisplatin, cyclophosphamide and dacarbazine. Each compound was administered i.p. as freshly prepared solutions in isotonic saline in combination with surgical removal of primary tumour, and was used at the maximum tolerated dose. NAMI-A significantly reduced the growth of lung metastases either when given prior to surgery (early growing tumours) on TS/A adenocarcinoma or after surgical ablation of primary tumours (already established lung metasases) on Lewis lung carcinoma. The postsurgical treatment of mice bearing MCa mammary carcinoma caused a significant prolongation of the life-span of the treated animals. In the comparison experiments, dacarbazine was completely ineffective, cisplatin was as active as NAMI-A on MCa mammary carcinoma, slightly less active than NAMI-A on TS/A adenocarcinoma and inactive on Lewis lung carcinoma, and cyclophosphamide was always more active than any other treatment performed. These data stress that NAMI-A, independently of the lack of direct cell cytotoxicity, when compared to the reference drugs, has a potent therapeutic effect in mice bearing solid metastasising tumours.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Ciclofosfamida/uso terapêutico , Dacarbazina/uso terapêutico , Dimetil Sulfóxido/análogos & derivados , Metástase Neoplásica/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Dimetil Sulfóxido/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Compostos de Rutênio
5.
Anticancer Drugs ; 10(1): 129-38, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10194556

RESUMO

NAMI-A (imidazolium trans-imidazoledimethylsulfoxidetetrachlororuthenate, ImH[trans-RuCl4(DMSO)Im]) is a new ruthenium compound active against lung metastasis of solid metastasizing tumors. We have tested this compound in mice with Lewis lung carcinoma or MCa mammary carcinoma in order to compare the effects on primary tumor and lung metastases with possible alterations of cell cycle distribution of tumor cells. We have also investigated whether there were unequal tissue accumulations of the compound itself at different dose levels ranging from 17.5 to 70 mg/kg/day given for six consecutive days. NAMI-A caused a reduction of metastasis weight larger than that of metastasis number; we explain this finding as the capacity of NAMI-A to selectively interfere with the growth of metastases already settled in the lungs. However, this specificity is not simply related to a larger concentration of NAMI-A in the lungs than in other tissues. Following i.p. treatment, NAMI-A rapidly disappeared from the peritoneal cavity; its low blood concentration may be caused by rapid renal clearance. These data provide further evidence for a selective anti-metastasis effect of the ruthenium complex NAMI-A. The reduction of lung metastasis is followed by a significant prolongation of the host's life-time expectancy, indicating a therapeutic benefit of NAMI-A on lung metastases from solid tumors.


Assuntos
Antineoplásicos/farmacologia , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Compostos Organometálicos/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/secundário , Ciclo Celular/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Dimetil Sulfóxido/toxicidade , Feminino , Citometria de Fluxo , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos , Compostos Organometálicos/toxicidade , Rutênio/análise , Rutênio/farmacocinética , Compostos de Rutênio , Espectrofotometria Atômica , Distribuição Tecidual
6.
J Pharmacol Exp Ther ; 289(1): 559-64, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10087050

RESUMO

The effects of NAMI-A (imidazolium trans-imidazoledimethyl sulfoxide-tetrachlororuthenate) are compared with cisplatin on tumor cells cultured in vitro at doses of 1 to 100 microM and on tumor metastases in vivo at maximum tolerated doses. Using mouse tumors that metastasize to the lungs, NAMI-A given i.p. for 6 consecutive days at 35 mg/kg/day, was effective independently of the tumor line being treated and of the stage of metastasis growth. Conversely, cisplatin (2 mg/kg/day for 6 days) was as effective as NAMI-A on MCa mammary carcinoma and TS/A adenocarcinoma and less effective than NAMI-A on Lewis lung carcinoma. Cisplatin reduced body weight gain and spleen weight during treatment and was much more toxic than NAMI-A on liver sinusoids, kidney tubules, and lung epithelium. In vitro NAMI-A caused a transient cell cycle arrest of tumor cells in the premitotic G2/M phase, whereas cisplatin caused a progressive dose-dependent disruption of cell cycle phases. Correspondingly, NAMI-A did not modify cell growth, whereas cisplatin caused a dose-dependent reduction of cell proliferation, as determined by sulforhodamine B test. Thus, NAMI-A, unlike cisplatin, is a potent agent for the treatment of solid tumor metastases as well as when these tumor lesions are in an advanced stage of growth. NAMI-A is endowed with a mechanism of action unrelated to direct tumor cell cytotoxicity, and such mechanism of action is responsible for a reduced host toxicity.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/patologia , Compostos Organometálicos/farmacologia , Animais , Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/toxicidade , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Dimetil Sulfóxido/toxicidade , Citometria de Fluxo , Humanos , Células KB , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Compostos Organometálicos/toxicidade , Propídio , Rodaminas , Compostos de Rutênio , Células Tumorais Cultivadas
7.
Clin Exp Metastasis ; 16(4): 371-9, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9626816

RESUMO

Imidazolium trans-imidazoledimethylsulphoxidetetrachlororuthenate ImH[trans-RuCl4(DMSO)Im] (NAMI-A), a ruthenium compound that replaces Na+ with ImH+ in the molecule of Na[trans-RuCl4(DMSO)Im] (NAMI), was studied for the anti-metastasis effects in models of solid metastasizing tumours of the mouse. NAMI-A, given i.p. at 35 mg/kg/day for six consecutive days, a dose equimolar to that of NAMI, to mice bearing Lewis lung carcinoma and MCa mammary carcinoma, markedly reduces lung metastasis weight by 80-90%, with an effect equal or even superior to that of NAMI, depending on the experimental system adopted. Correspondingly, NAMI-A increases the content of connective tissue in the tumour matrix, around blood vessels, and in the tumour capsule, augments the percentage of tumour cells in G2/M phase and reduces the amount of CD45+ cells infiltrating the tumour parenchyma. The effects of the same doses on spleen lymphocytes correspond to an increase of CD8+ subset without any change of the distribution of cells in G0/G1, S and G2/M phases. The study shows that NAMI-A behaves similarly to NAMI on the several parameters examined in comparison experiments and therefore we suggest to credit NAMI-A with all the biological actions already described for NAMI during the last 3 years. The replacement of Na+ with ImH+ therefore, besides the better chemical stability of the molecule, confers to [trans-RuCl4(DMSO)Im]- a closer similarity with a true drug to be used in humans, and suggests this molecule for future studies of preclinical toxicology and phase I and II clinical trials.


Assuntos
Antineoplásicos/farmacologia , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Compostos Organometálicos/farmacologia , Rutênio/farmacologia , Animais , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Carcinoma/secundário , Dimetil Sulfóxido/farmacologia , Feminino , Citometria de Fluxo , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/secundário , Camundongos , Camundongos Endogâmicos C57BL
8.
Pathol Oncol Res ; 4(1): 30-6, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9555118

RESUMO

The effects of the new molecule ImH[trans-RuCl4(DMSO)Im] (NAMI-A), administered orally or intraperitoneally to adjuvant-arthritic rats or orally to mice bearing s.c. or i.m. implants of MCa mammary carcinoma, were studied. NAMI-A was not able to modify the progression of chronic inflammation in the complete Freund-adjuvant injected animals. Histology indicated a significant worsening of the inflammatory process, characterised by an increased infiltration of inflammatory cells, as well as by a remarkable deposition of connective tissue fibres around the blood vessels and alveolar walls. NAMI-A had no effect on primary i.m. implanted MCa mammary carcinoma growth and its lung metastasis formation, but significantly interfered with the cell cycle of primary tumor cells following bolus oral administration. On the contrary, NAMI-A caused a significant inhibition of lung metastasis accompanied by a dramatic deposition of connective tissue fibres around the primary tumor mass, when given as medicated food to mice implanted s.c. with MCa tumor. These data indicated that NAMI-A is well absorbed after oral administration although there is no connection between lung concentration and the antimetastatic activity. Conversely, the marked deposition of connective tissues in NAMI-A treated animals is in agreement with the reported effects of the compound on extracellular matrix and tumor blood vessels.


Assuntos
Anti-Inflamatórios/administração & dosagem , Antineoplásicos/administração & dosagem , Artrite/tratamento farmacológico , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Compostos Organometálicos/administração & dosagem , Compostos de Rutênio/administração & dosagem , Animais , Dimetil Sulfóxido/administração & dosagem , Feminino , Camundongos , Metástase Neoplásica , Ratos
9.
Int J Cancer ; 68(1): 60-6, 1996 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-8895542

RESUMO

The anti-metastatic ruthenium complex Na[trans-RuCl4(DMSO)Im] was given i.p. at 22 and 44 mg/kg/day, on days 8-13 after tumour implantation, to mice carrying s.c. implants of MCa mammary carcinoma. The aim of the study was to compare the effects on lung metastasis formation with those on primary tumour cells. This investigation was based on flow cytometry analysis after propidium iodide and acridine orange staining, histology of tumour parenchyma and RT-PCR analysis for the type-IV collagenases MMP-9 and MMP-2 and their respective inhibitors TIMP-1 and TIMP-2 mRNAs. Na[trans-RuCl4(DMSO)Im] is not cytotoxic for tumour cells but has the capacity of interacting with nucleic acids, giving a general reduction of nucleic acid content as shown by a marked reduction of acridine orange staining and a tendency to a reduction of DNA polyploidy with marked reduction of 8n and 4n cell populations. Na[trans-RuCl4(DMSO)Im] also influences a proteolytic system which has the potential of degrading the basement membrane and has been related to metastatic aggressiveness: it markedly reduces, in a dose-dependent manner, MMP-2/TIMP-2 balance, but not that of MMP-9/TIMP-1. The different enzyme/inhibitor mRNA levels between untreated and treated tumours seem to be unaffected by tumour-infiltrating lymphocytes and are paralleled by the maintenance of connective tissue around blood vessels in the tumour mass. Correspondingly, lung metastasis formation is markedly reduced, to less than 10% of that seen in controls.


Assuntos
Antineoplásicos/uso terapêutico , Dimetil Sulfóxido/análogos & derivados , Gelatinases/metabolismo , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Compostos Organometálicos/uso terapêutico , Inibidores de Proteases/metabolismo , Laranja de Acridina , Animais , Colagenases/genética , Colagenases/metabolismo , Corantes , Dimetil Sulfóxido/uso terapêutico , Endotélio/patologia , Feminino , Citometria de Fluxo , Gelatinases/antagonistas & inibidores , Gelatinases/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/prevenção & controle , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Reação em Cadeia da Polimerase , Propídio , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/metabolismo , DNA Polimerase Dirigida por RNA , Inibidor Tecidual de Metaloproteinase-2 , Inibidores Teciduais de Metaloproteinases
10.
Anticancer Drugs ; 7(6): 697-702, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8913439

RESUMO

Treatment of MCa mammary carcinoma metastases by i.p. administration of a total dose of 450 mg/kg Na[trans-RuCl4(DMSO)lm], after successful surgical removal of primary tumor mass, causes a significant prolongation of the host's life-time expectancy. This effect, related to lung metastasis inhibition, seems not attributable to a direct inhibition of tumor cells since antimetastatic effects can be achieved also when drug treatment occurs before tumor cell injection into the host. Also, the activity of Na[trans-RuCl4(DMSO)lm] seems independent of its concentration in tumor cells. Rather it must be stressed that the fate of this compound in the blood, following i.v. administration, is fast and only a very low percent of the total dose reaches the tumor target in the lungs. These data emphasize the possibility that Na[trans-RuCl4(DMSO)lm] increases the resistance of the host against metastasis formation, possibly by the already shown mechanism of potentiation of the extracellular matrix and reduction of blood stream invasion by tumor cells.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Rutênio/uso terapêutico , Animais , Antineoplásicos/metabolismo , Dimetil Sulfóxido/uso terapêutico , Neoplasias Pulmonares/prevenção & controle , Camundongos , Neoplasia Residual/tratamento farmacológico , Rutênio/metabolismo , Análise de Sobrevida
11.
Met Based Drugs ; 3(2): 67-73, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-18472798

RESUMO

The effects of the treatment of tumor cells of MCa mammary carcinoma and TLX5 lymphoma with the ruthenium complex Na[trans-RuCl(4) (DMSO)lm] for several transplant generations were studied on tumor growth and metastases formation. On TLX5 lymphoma cells, treatment was performed in vitro prior to in vivo inoculation of tumor cells in intact or immunesuppressed mice. Either considering tumor take and growth or its capacity to invade the brain of the inoculated hosts, Na[trans-RuCl(4)(DMSO)lm] did not induce any significant modification. Conversely, in mice with MCa mammary carcinoma, the in vivo treatment of tumor cells in immunesuppressed hosts caused a progressive increase of DNA activity and, starting from the 4th transplant generation, a significantly increased susceptibility of lung metastasis formation to a further treatment in intact mice. These data seem to suggest that Na[trans-RuCl(4)(DMSO)Im] does not induce chemical xenogenization of tumor cells nor its repeated treatment induces resistance in tumor cells. Conversely, it appears that Na[trans-RuCl(4)(DMSO)lm] may select a tumor cell population which maintains its capacity to metastasise to the lung but with enhanced sensitivity to the antimetastatic properties of this compound.

12.
Chem Biol Interact ; 95(1-2): 109-26, 1995 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-7697744

RESUMO

A series of 18 ruthenium(III) complexes, structurally related to the selective antimetastatic drug Na[trans-RuCl4(DMSO)Im], and characterized by the presence of sulfoxide and nitrogen-donor ligands were tested on TLX5 lymphoma and some of them on MCa mammary carcinoma to evaluate the dependence of the degree of cytotoxicity and of antimetastatic activity on the chemical properties. In vitro cytotoxicity is present only at high concentrations (> 10(-4) M), depends upon lipophilicity and is markedly affected by the presence of 5% serum or plasma samples in the culture medium. The comparison of the effects on in vitro cytotoxicity with in vivo antitumor and antimetastatic action points out that these compounds reduce metastasis formation by a mechanism unrelated to a direct tumor cell cytotoxicity. If on one hand Na[trans-RuCl4(TMSO)Iq], the compound that shows the most potent in vitro cytotoxic effects, is the least effective against metastases, on the other Na[trans-RuCl4(DMSO)Im], the compound that better reduces metastasis formation, is rather devoid of cytotoxic effects on tumor cells kept in vitro. In particular, Na[trans-RuCl4(DMSO)Im] seems to distinguish between artificially induced metastases and spontaneous metastases and reduces only the former by a cytotoxic mechanism. Out of all the tested compounds, with the exception of Na[trans-RuCl4(DMSO)Ox], Na[trans-RuCl4(DMSO)Im] is confirmed to be the most selective antimetastatic agent of the group.


Assuntos
Antineoplásicos/toxicidade , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Linfoma/tratamento farmacológico , Linfoma/patologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Compostos de Rutênio/toxicidade , Animais , Neoplasias Encefálicas/secundário , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Camundongos , Camundongos Endogâmicos CBA , Transplante de Neoplasias
13.
Met Based Drugs ; 2(4): 195-9, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-18472766

RESUMO

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.

14.
Clin Exp Metastasis ; 12(2): 93-100, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8306532

RESUMO

The ruthenium-dimethylsulfoxide complex Na(trans-RuCl4(DMSO)Im] was given i.v. to mice bearing MCa mammary carcinoma and its effects on tumor growth and on healthy host tissues were studied by macroscopic examination of primary tumor growth, by survival time, and by histological analysis using light microscopy and SEM. Either by means of vivo-vivo bioassays or by microscopic examination it appeared that the growth of lung tumors was markedly reduced, whereas the growth of the i.m. primary tumor was much less affected. These effects account for the prolongation of survival time and for the cure rate observed. The favourable effect on survival time was also influenced by the lack of significant cytotoxicity for normal tissues such as lung and kidney epithelia, muscle and liver cells, splenocytes and bone marrow. It thus appears that the selective interaction with tumor cells in the lungs cannot simply be attributed to a selectively higher localization of the compound at this site, nor to a modification of the histological structure of primary tumor. These results highlight the pharmacologic properties of this compound for the control of solid tumor metastases, an effect that was shown to be similarly exerted on advanced tumor metastases.


Assuntos
Dimetil Sulfóxido/análogos & derivados , Dimetil Sulfóxido/uso terapêutico , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Compostos de Rutênio/uso terapêutico , Animais , Peso Corporal/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Esquema de Medicação , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos CBA
15.
Met Based Drugs ; 1(1): 41-63, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-18476216

RESUMO

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.

16.
Eur J Cancer ; 29A(13): 1873-9, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8260245

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/prevenção & controle , Leucemia P388/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Neoplasias Encefálicas/secundário , Cisplatino/uso terapêutico , DNA de Neoplasias/efeitos dos fármacos , Resistência a Medicamentos , Leucemia P388/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos , Rutênio , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Int J Oncol ; 2(4): 527-9, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21573587

RESUMO

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.

18.
Clin Exp Metastasis ; 10(4): 273-80, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1617835

RESUMO

The antitumor and antimetastatic effects of a ruthenium(III) complex, Na[trans-RuCl4(DMSO)Im], have been examined in mice bearing MCa mammary carcinoma. Na[trans-RuCl4(DMSO)Im] is capable of reducing primary tumor growth and of prolonging the survival time with different schedules of administration. However, better effects were obtained (a) with treatments started soon after tumor implantation, (b) with daily administration rather than with treatments at 4-day intervals and (c) using relatively low daily doses. The prolongation of survival time in tumor-bearing hosts, greater than that obtained with cisplatin, cannot be simply related to the effect on primary tumor growth, always less than that of cisplatin. Rather, emphasis is placed on the reduction of lung metastasis formation, obtained either by i.v. injection of tumor cells (lung colonization or spontaneously from i.m. tumor implants, which is significantly greater than the reduction of primary tumor growth. The antimetastatic effect depends on the treatment schedule and is greater with low doses given daily than with high doses administrated with drug-free intervals. Metastasis reduction involved mainly large metastatic nodules, reducing the average weight of each metastasis by 8-fold for spontaneous metastases and by 5-fold for lung colonies. The antimetastatic action of Na[trans-RuCl4(DMSO)Im] thus indicates the possibility that related analogs can represent a new generation of antitumor compounds capable of selectively interacting with metastasis formation of solid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Dimetil Sulfóxido/análogos & derivados , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Compostos Organometálicos/uso terapêutico , Animais , Cisplatino/uso terapêutico , Esquema de Medicação , Feminino , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos CBA , Transplante de Neoplasias , Fatores de Tempo
20.
Anticancer Drugs ; 3(1): 25-31, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1623212

RESUMO

The effects of two new Ru(III) complexes, [mer-RuCl3(DMSO)2Im] degrees and Na[trans-RuCl4(DMSO)Im], were investigated on primary tumor growth and on the survival time using three solid metastasizing tumors of the mouse: Lewis lung carcinoma, B16 melanoma and MCa mammary carcinoma. Na[trans-RuCl4(DMSO)Im] appears to be the most promising compound, in that: (1) it is soluble in water and therefore easy to handle in comparison with the neutral species [mer-RuCl3(DMSO)2Im]degrees or to the already described BBR2382; (2) similarly to cisplatin, though at a lower level, it reduces tumor growth in its primary site in each tumor model employed; (3) unlike cisplatin, it increases the life span of tumor-bearing hosts in all tumors used, independently of the effects on primary tumor growth; and (4) it is also effective in reducing spontaneous metastasis formation when the effects on primary tumor growth are completely absent. Dimethylsulfoxide (DMSO), used for solubilizing poorly water-soluble compounds (i.e. [mer-RuCl3(DMSO)2Im]degrees) or for stabilizing the compound in the solution before injection (i.e. Na[trans-RuCl4(DMSO)Im]), reduces the anti-tumor potency. Conversely, the antitumor effects of Na[trans-RuCl4(DMSO)Im] are more pronounced in mice hydrated with isotonic saline. We conclude that Na[trans-RuCl4(DMSO)Im] is a good candidate for further investigations aimed at ascertaining the mechanism of the anti-metastatic activity and of the positive effects on survival time of mice bearing solid metastasizing tumors.


Assuntos
Carcinoma/tratamento farmacológico , Dimetil Sulfóxido/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Compostos Organometálicos/farmacologia , Animais , Cisplatino/uso terapêutico , Dimetil Sulfóxido/farmacologia , Formas de Dosagem , Esquema de Medicação , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA