Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Invest New Drugs ; 22(4): 359-67, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15292706

RESUMEN

The pattern of inhibition of cell proliferation and cytotoxicity in vitro by 1,4-bis(1-naphthyl)-2,3-dinitro-1,3-butadiene (Naph-DNB) was evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and the trypan blue (TB) dye exclusion assays in nine murine and human cell lines of different histologic origin. In our culture conditions Naph-DNB showed a good inhibiting activity against all cell lines tested, with IC(50)s varying within a narrow micromolar range of concentrations (2.0 +/- 0.2-14.3 +/- 2.3 microM). In particular, murine P388 (leukemia), human Jurkat (leukemia), A2780, PA-1 (ovarian carcinoma) and Saos-2 (osteosarcoma) cells showed the highest sensitivity to the inhibiting potential of Naph-DNB, while human A549 (non small cell lung cancer, NSCLC), MDA-MB-231 (breast cancer), HGC-27 (gastric cancer) and HCT-8 (colon carcinoma) were the least sensitive cell lines. Moreover, the analysis of cytotoxicity of Naph-DNB evaluated by the TB test showed that this compound was able to kill cells with IC(50)s ranging from 1.7 to 39.2 microM. The study of the induction of apoptosis was carried out by 4'-6-diamidine-2'-phenylindole (DAPI) staining of segmented nuclei, western blot of p53 protein and TdT-mediated dUTP-biotin nick end labeling (TUNEL) method, while the interaction with DNA was evaluated through the analysis of interstrand cross-link (ISCL) formation. Our data show that in all cell lines tested Naph-DNB was able to form ISCLs, to upregulate p53 oncosuppressor-protein and to induce apoptosis. Moreover, TUNEL analysis also suggested that Naph-DNB, similarly to other anticancer drugs, was able to block cells in the G (0)/ G (1) phase of the cell cycle. In conclusion our data suggest that Naph-DNB may be an effective novel lead molecule for the design of new anticancer compounds.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Butadienos/farmacología , Proliferación Celular/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Microscopía Fluorescente , Proteína p53 Supresora de Tumor/biosíntesis
2.
Oncol Rep ; 12(1): 91-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15201965

RESUMEN

The inhibition of cell proliferation by 1,4-bis (1-naphthyl)-2,3-dinitro-1,3-butadiene (Naph-DNB) was evaluated in vitro against 4 cell lines (L1210/DDP, A2780/DX3, HCT-8/FU7dR, A549-T12) selected for their resistance to cisplatin, doxorubicin, 5-fluorouracil and taxol, and their wild-type counterparts. Naph-DNB is a novel anti-cancer compound obtained years ago within a research project of Organic Chemistry aimed at synthesizing 2,3-dinitrobutadiene derivatives. Because of its chemical structure, Naph-DNB was suggested to interact with nucleic acids, in particular DNA, and the other cellular macromolecules. This hypothesis made us consider Naph-DNB as a candidate for studies concerning its antitumour activity. We used the MTT assay to test the inhibition of cell proliferation after incubation of the cell lines with Naph-DNB for 72 h. For comparison, resistant and wild-type cell lines were also tested against those anticancer drugs used in vitro for their selection. In these culture conditions Naph-DNB retained its inhibiting activity against all resistant cells with IC50 values similar to those obtained in corresponding wild-type cell lines. Moreover, Naph-DNB was twice as effective as 5-fluorouracil against wild-type HCT-8 cells. Our previous findings about the interaction of Naph-DNB with DNA through the formation of interstrand cross-links suggested a mechanism of action similar to that of platinum/alkylating agents or topoisomerase inhibitors (intercalating agents). Our present data obtained by the K-SDS precipitation assay in A2780 and A549 cells showed that Naph-DNB is not able to form a stable topoisomerase-DNA complex as is the case for topoisomerase inhibitors. In conclusion, our results indicate that Naph-DNB is able to overcome some of the classical mechanisms of resistance selected by some anticancer drugs mainly used in clinical setting.


Asunto(s)
Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Butadienos , Línea Celular Tumoral , Cisplatino/farmacología , Doxorrubicina/farmacología , Fluorouracilo/farmacología , Humanos , Cinética , Paclitaxel/farmacología
3.
Invest New Drugs ; 22(1): 3-16, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14707490

RESUMEN

Cis-diaminechloro-[2-(diethylamino) ethyl 4-amino-benzoate, N(4)]-chloride platinum (II) monohydrochloride monohydrate (DPR) is a new platinum triamine complex obtained from the synthesis of cisplatin and procaine. In this paper we analyzed, adopting a disease-oriented strategy, the tumour selectivity of this compound, its ability to induce apoptosis and its mechanism of interaction with DNA. The inhibition of cell proliferation was evaluated by the MTT assay using a panel of 51 tumour cell lines. Some of them were also evaluated for the induction of apoptosis by 4'-6-diamidine-2'-phenylindole (DAPI) staining, Western blot of p53 protein and agarose gel electrophoresis of ladder DNA. Finally, interstand cross-links (ISCL) were evaluated by ethidium bromide fluorescence technique. When evaluated by the MTT assay, DPR showed a high selective activity for neuroblastoma, small cell lung cancer (SCLC), ovarian cancer and leukemia cell lines. The comparison of mean graphs of DPR and cisplatin suggested that our compound possesses a mechanism of action similar to that, at least in part, of its parent compound. Moreover, DPR showed itself to be a good trigger of programmed cell death, as demonstrated by DAPI staining, activation of p53 protein and agarose gel electrophoresis of ladder DNA. Finally, the study of the formation of ISCLs demonstrated that DPR, despite being a monofunctional platinum compound, is able to form bifunctional adducts through the release of procaine residue. Data presented here suggest that DPR is an antitumour agent able to trigger apoptosis, and that it is endowed with a peculiar mechanism(s) of action and a special selective activity against two tumours, namely neuroblastoma and SCLC, which are still characterized by a low incidence of long-term survivors.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/análogos & derivados , Cisplatino/farmacología , Compuestos Organoplatinos/farmacología , Procaína/análogos & derivados , Procaína/farmacología , Animales , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fragmentación del ADN , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Electroforesis en Gel de Agar , Colorantes Fluorescentes , Humanos , Indoles/farmacología , Concentración 50 Inhibidora , Ratones , Microscopía Fluorescente , Procaína/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba
4.
Arch Toxicol ; 77(10): 584-90, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14574446

RESUMEN

Embryo-lethal and teratogenic effects caused by the cisplatin-procaine complex cis-diaminechloro-[2-(diethylamino) ethyl 4-amino-benzoate, N(4)]-chlorideplatinum(II) monohydrochloride monohydrate (DPR) were examined in CD-1 mice after a single administration of 7, 14, 21 or 28 mg/kg, injected on day 6, 9, 13 or 16 of pregnancy. At day 18 of pregnancy fetuses were removed and carefully examined for external, visceral and skeletal malformations under a dissecting microscope. A significant reduction of maternal weight gain was observed in pregnant mice after the administration of 21 (day 13) or 28 mg/kg (days 9 and 13) DPR. The exposure to DPR during the organogenesis and early histogenesis periods of prenatal development (administration on day 9 or 13) induced a significant reduction of the mean percentage of live fetuses and a significant increase of the mean percentage of dead and resorbed fetuses. A dose-dependent reduction of fetal body weight was observed in surviving specimens exposed to DPR on embryonic day 9, 13 or 16. The analysis of surviving fetuses killed on day 18 of gestation showed that a few, but statistically significant, external malformations and visceral anomalies were observed after administration of 21 or 28 mg/kg DPR on embryonic day 13. External malformations consisted of three hepato-omphalocele and six palatoschisis (one random palatoschisis was also observed at 21 mg/kg DPR given on day 9), while visceral anomalies included only renal pelvis dilatation. Skeletal anomalies affected fetuses independently of the day of treatment and were more frequent at the highest doses of DPR. They consisted of a delay in skull ossifications, vertebral and sternal anomalies, and formation of extra ribs. A low and non-significant incidence of skeletal malformations (asymmetric sternum) was noticed in fetuses. Our data demonstrated that DPR can cause embryotoxic effects if administered during the period of organogenesis and early histogenesis. Beside embryo-lethality, DPR induced growth retardation and malformations in surviving fetuses.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Antineoplásicos/toxicidad , Cisplatino/análogos & derivados , Cisplatino/toxicidad , Muerte Fetal/inducido químicamente , Compuestos Organoplatinos/toxicidad , Procaína/análogos & derivados , Procaína/toxicidad , Animales , Antineoplásicos/administración & dosificación , Huesos/anomalías , Huesos/embriología , Cisplatino/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Feto/anomalías , Feto/efectos de los fármacos , Inyecciones Intraperitoneales , Ratones , Compuestos Organoplatinos/administración & dosificación , Embarazo , Procaína/administración & dosificación , Vísceras/anomalías , Vísceras/embriología
5.
Anticancer Res ; 23(2B): 1509-16, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12820417

RESUMEN

BACKGROUND: Our previous studies showed that procainamide hydrochloride may be an important modulator of cisplatin toxicity and antitumour activity. This study was performed in order to investigate if procainamide hydrochloride may influence the therapeutic index of cisplatin by inducing modifications of its pharmacokinetics and pharmacodymanics in vivo. MATERIALS AND METHODS: The pharmacokinetic profile of cisplatin administered either in the presence or absence of procainamide hydrochloride was investigated in BDF1 female mice bearing 6-day P388 leukemia. Procainamide hydrochloride was administered i.v. at the dose of 50 mg/kg, immediately before cisplatin which, in turn, was administered i.p. at the dose of 8 mg/kg. RESULTS: The combined administration of the antiarrhythmic drug and cisplatin caused significant differences in the pharmacokinetic profiles of Pt in plasma, ascites fluid and tissues. Filterable Pt was significantly increased both in plasma and ascites fluid in animals given the combined treatment. Similarly, a small increase was also found for total plasma Pt. These differences caused some changes of the pharmacokinetic parameters of filterable (plasma: AUC0-1 h = +16%, t1/2 alpha = +29%, t1/2b = +14%, K2p = -32%; ascites fluid: AUC0-1 h = +23%, t1/2 alpha = +78%, t1/2 beta = -49%, and total Pt (plasma: AUC0-1 h = +19%, t1/2 alpha = +27%, t1/2 beta = -22%; ascites fluid: AUC0-1 h = +6%, AUC0-infinity = +43%, t1/2 alpha = +30%). The analysis of tissue Pt content showed the general increase of Pt concentration in the main organs of animals treated with cisplatin and procainamide hydrochloride, with AUC0-24 h increased by 95%, 22%, 90% and 28% in kidney, liver, spleen and lung, respectively. The analysis of binding of Pt to DNA and percent interstrand cross-links (%ISCL) in P388 tumour cells showed that the % ISCL (10.44 +/- 3.81% vs. 3.51 +/- 0.01%) and the efficiency of ISCL formation (0.51 +/- 0.14 vs. 0.17 +/- 0.02 %ISCL.microgram DNA/pg Pt) were significantly greater when cisplatin was administered in association with procainamide hydrochloride. CONCLUSION: Our results show that procainamide hydrochloride may alter the pharmacodynamics and the pharmacokinetics and distribution of Pt in tumored mice treated with cisplatin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/farmacocinética , Leucemia P388/tratamiento farmacológico , Platino (Metal)/análisis , Procainamida/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Área Bajo la Curva , Líquido Ascítico/química , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cisplatino/administración & dosificación , Cisplatino/farmacología , Cisplatino/toxicidad , Reactivos de Enlaces Cruzados/administración & dosificación , Reactivos de Enlaces Cruzados/farmacocinética , Reactivos de Enlaces Cruzados/farmacología , Reactivos de Enlaces Cruzados/toxicidad , Aductos de ADN , ADN de Neoplasias/química , ADN de Neoplasias/efectos de los fármacos , Femenino , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Ratones , Especificidad de Órganos , Platino (Metal)/farmacocinética , Procainamida/administración & dosificación , Distribución Tisular
6.
Anticancer Drugs ; 14(2): 163-6, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12569303

RESUMEN

We evaluated in vitro the inhibitory effect of cis-diaminechloro-[2-(diethylamino) ethyl 4-amino-benzoate, N4]-chlorideplatinum(II) monohydrochloride monohydrate (DPR) on colony formation by granulocyte/macrophage (CFU-GM) peripheral blood progenitor cells, representing a method to quantitate the toxicity of drugs to the hematopoietic system, and human leukemic cell lines. The results were compared with those obtained exposing cells to cisplatin and carboplatin. Our data showed that while DPR had a significantly better cytotoxic activity than cisplatin and carboplatin against HL60 and K562, and than carboplatin against Molt 4 cells, it showed 12 and 43 times less inhibitory effect on CFU-GM than cisplatin and carboplatin, respectively. These results suggest that the myelosuppressive activity of DPR could be lower than that of cisplatin and carboplatin, and, furthermore, that leukemic cells represent a preferential target for its cytotoxic activity compared to normal committed hemopoietic progenitor cells. All our results speak in favor of a better therapeutic index for DPR than for the other platinum compounds considered here.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Cisplatino/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Procaína/análogos & derivados , Procaína/farmacología , Antineoplásicos/efectos adversos , Carboplatino/farmacología , Cisplatino/efectos adversos , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Células HL-60/efectos de los fármacos , Humanos , Técnicas In Vitro , Células K562/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Compuestos Organoplatinos/efectos adversos , Procaína/efectos adversos
7.
Anticancer Drugs ; 13(10): 1043-54, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12439338

RESUMEN

Efforts have been made to reduce the undesirable side effects of cisplatin, mainly nephro- and neurotoxicity, but their reduction is usually accompanied by a concomitant inhibition of antitumor activity. The local anesthetic procaine hydrochloride (P.HCl) improves the therapeutic index of cisplatin not only by the reduction of its nephro- and hemotoxicity, but also by an increase of its antitumor activity. We therefore investigated the effects of a combined treatment of cisplatin and P.HCl on rat kidneys and compared this to kidneys from rats treated with a toxic dose of cisplatin or P.HCl alone. Treatment with a saline solution was used as control. Dehydrogenase activities [succinate dehydrogenase (SDH) and NADPH diaphorase reaction demonstrating nitric oxide synthase (NOS/NADPHd)] and phosphatase activities [K -nitrophenyl phosphatase (K pNPPase), alkaline phosphatase (AlPase) and acid phosphatase (AcPase)] were studied on cryostatic sections of kidneys from controls and treated rats. Evidence of heavy morphological damage and altered AlPase and AcPase activities induced by cisplatin were observed in the S3 segment of the proximal tubules. In addition, SDH and K pNPPase activities showed some changes in the distal tubule cells. The NOS/NADPHd activity in macula densa was drastically reduced. Combined treatment of cisplatin and P.HCl greatly attenuated morphological alterations of the rat kidney and reduced the changes in enzyme activities, except for NOS/NADPHd activity, compared to the cisplatin-treated group of animals. The study indicates that, in cisplatin-induced nephrotoxicity, a significant role is played by enzyme activities, in particular K pNPPase and NOS/NADPHd, and that P.HCl can mitigate the nephrotoxicity of cisplatin, possibly by influencing some enzyme activities involved in important renal metabolic pathways.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Riñón/efectos de los fármacos , Procaína/farmacología , Animales , Combinación de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Inyecciones Intraperitoneales , Riñón/enzimología , Riñón/ultraestructura , Corteza Renal/efectos de los fármacos , Corteza Renal/enzimología , Corteza Renal/ultraestructura , Médula Renal/efectos de los fármacos , Médula Renal/enzimología , Médula Renal/ultraestructura , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/ultraestructura , Masculino , Microscopía Electrónica , Óxido Nítrico Sintasa/metabolismo , Oxidorreductasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Ratas , Ratas Sprague-Dawley
8.
Eur J Pharmacol ; 442(3): 265-72, 2002 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-12065080

RESUMEN

In preceding papers, we proposed that procainamide hydrochloride, a class I antiarrhythmic agent, was able to protect mice and rats from cisplatin-induced nephrotoxicity and that it could exert its action through accumulation in kidneys followed by coordination with cisplatin (or its hydrolysis metabolites) and formation of a less toxic platinum compound similar to the new platinum(II) triamine complex cis-diamminechloro-[2-(diethylamino)ethyl 4-amino-benzoate, N4]-chlorideplatinum(II) monohydrochloride monohydrate, obtained by the reaction of cisplatin with procaine hydrochloride. Hepatotoxicity is not considered as a dose-limiting toxicity for cisplatin, but liver toxicity can occur when the antineoplastic drug is administered at high doses. Here, we report that procainamide hydrochloride, at an i.p. dose of 100 mg/kg, reduces cisplatin-induced hepatotoxicity, as evidenced by the normalization of plasma activity of glutamic oxalacetic transaminase and gamma-glutamyl transpeptidase, as well as by histological examination of the liver tissue. Twenty-four hours after i.p. treatment with the combination of 7.5 mg/kg cisplatin and 100 mg/kg procainamide, a significant increase of procainamide (+56%, P<0.05), total platinum (+31%, P<0.05), platinum-DNA adducts (+31%, P<0.05) and percent DNA-DNA interstrand cross-links (+69%, P<0.02) was found in liver tissue, as compared to animals treated with cisplatin alone. Moreover, in accordance with these findings, we also observed a slightly lower concentration and cumulative excretion of platinum in the feces. Since mitochondrial injury is considered a central event in the early stages of the nephrotoxic effect of cisplatin, the distribution of platinum in these subcellular organelles obtained from hepatocytes was determined after treatment with cisplatin with or without procainamide hydrochloride, together with platinum concentration in their cytosolic fraction. Our data show that the coadministration of procainamide hydrochloride produced a rearrangement of subcellular platinum distribution in hepatocytes with a slight decrease in mitochondria (-15%, P<0.10) and a slight increase in the cytosolic fraction (+40%, P<0.10) of platinum content, compared to the treatment with cisplatin alone. In analogy with our previous results in the kidney, confirmed here by our data in vitro, we suggest that the hepatoprotective activity of procainamide hydrochloride is linked to the formation of a less toxic platinum complex, which leads to inactivation of cisplatin itself and/or its highly toxic hydrolysis metabolites and to a different subcellular distribution of platinum.


Asunto(s)
Antiarrítmicos/farmacología , Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Hígado/efectos de los fármacos , Procainamida/farmacología , Animales , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Citosol/metabolismo , Interacciones Farmacológicas , Heces/química , Hígado/metabolismo , Hígado/patología , Masculino , Mitocondrias Hepáticas/metabolismo , Platino (Metal)/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...