Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Biol Inorg Chem ; 15(5): 737-48, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20221888

RESUMEN

The ruthenium compound KP1019 has demonstrated promising anticancer activity in a pilot clinical trial. This study aims to evaluate the intracellular uptake/binding patterns of KP1019 and its sodium salt KP1339, which is currently in a phase I-IIa study. Although KP1339 tended to be moderately less cytotoxic than KP1019, IC(50) values in several cancer cell models revealed significant correlation of the cytotoxicity profiles, suggesting similar targets for the two drugs. Accordingly, both drugs activated apoptosis, indicated by caspase activation via comparable pathways. Drug uptake determined by inductively coupled plasma mass spectrometry (ICP-MS) was completed after 1 h, corresponding to full cytotoxicity as early as after 3 h of drug exposure. Surprisingly, the total cellular drug uptake did not correlate with cytotoxicity. However, distinct differences in intracellular distribution patterns suggested that the major targets for the two ruthenium drugs are cytosolic rather than nuclear. Consequently, drug-protein binding in cytosolic fractions of drug-treated cells was analyzed by native size-exclusion chromatography (SEC) coupled online with ICP-MS. Ruthenium-protein binding of KP1019- and KP1339-treated cells distinctly differed from the platinum binding pattern observed after cisplatin treatment. An adapted SEC-SEC-ICP-MS system identified large protein complexes/aggregates above 700 kDa as initial major binding partners in the cytosol, followed by ruthenium redistribution to the soluble protein weight fraction below 40 kDa. Taken together, our data indicate that KP1019 and KP1339 rapidly enter tumor cells, followed by binding to larger protein complexes/organelles. The different protein binding patterns as compared with those for cisplatin suggest specific protein targets and consequently a unique mode of action for the ruthenium drugs investigated.


Asunto(s)
Antineoplásicos/metabolismo , Indazoles/metabolismo , Compuestos Organometálicos/metabolismo , Proteínas/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Citosol/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indazoles/síntesis química , Indazoles/química , Indazoles/farmacología , Espectrometría de Masas , Peso Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Unión Proteica , Compuestos de Rutenio , Relación Estructura-Actividad , Factores de Tiempo , Células Tumorales Cultivadas
2.
Naunyn Schmiedebergs Arch Pharmacol ; 389(1): 17-32, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26319048

RESUMEN

Extrusion of chemotherapeutics by ATP-binding cassette (ABC) transporters like ABCB1 (P-glycoprotein) represents a crucial mechanism of multidrug resistance in cancer therapy. We have previously shown that the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor simvastatin directly inhibits ABCB1, alters the glycosylation of the transporter, and enhances the intracellular accumulation of doxorubicin with subsequent anti-cancer action. Here, we show that simvastatin reduces endogenous dolichol levels and ABCB1 in human neuroblastoma SH-SY5Y cells. Coapplication with dolichol prevents the downregulation of the ABCB1 transporter. Importantly, dolichol also attenuated simvastatin-induced apoptosis, unmasking involvement of unfolded protein response. Direct monitoring of the fluorescent fusion protein YFP-ABCB1 further confirms concentration-dependent reduction of ABCB1 in HEK293 cells by simvastatin. In simvastatin-treated murine xenografts, ABCB1 was also reduced in the liver and rhabdomyosarcoma but did not reach significance in neuroblastoma. Nevertheless, the in vivo anti-cancer effects of simvastatin are corroborated by increased apoptosis in tumor tissues. These findings provide experimental evidence for usage of simvastatin in novel chemotherapeutic regimens and link dolichol depletion to simvastatin-induced anti-cancer activity.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Dolicoles/farmacología , Regulación hacia Abajo , Femenino , Células HEK293 , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Simvastatina/uso terapéutico , Carga Tumoral/efectos de los fármacos
3.
Melanoma Res ; 25(4): 284-94, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26020489

RESUMEN

The interleukin (IL)-6 inhibits the growth of early-stage melanoma cells, but not metastatic cells. Metastatic melanoma cells are susceptible to statin-induced apoptosis, but this is not clear for early-stage melanoma cells. This study aimed to investigate the IL-6 susceptibility of melanoma cells from different stages in the presence of simvastatin to overcome loss of growth arrest. ELISA was used to detect secreted IL-6 in human melanoma cells. The effects of IL-6 were measured by western blots for STAT3 and Bcl-2 family proteins. Apoptosis and proliferation were measured by caspase 3 activity, Annexin V staining, cell cycle analysis, and a wound-healing assay. Human metastatic melanoma cells A375 and 518A2 secrete high amounts of IL-6, in contrast to early-stage WM35 cells. Canonical IL-6 signaling is intact in these cells, documented by transient phosphorylation of STAT3. Although WM35 cells are highly resistant to simvastatin-induced apoptosis, coadministration with IL-6 enhanced the susceptibility to undergo apoptosis. This proapoptotic effect of IL-6 might be explained by a downregulation of Bcl-XL, observed only in WM35 cells. Furthermore, the IL-6 receptor blocking antibody tocilizumab was coadministered and unmasked an IL-6-sensitive proportion in the simvastatin-induced caspase 3 activity of metastatic melanoma cells. These results confirm that simvastatin facilitates apoptosis in combination with IL-6. Although endogenous IL-6 secretion is sufficient in metastatic melanoma cells, exogenously added IL-6 is needed for WM35 cells. This effect may explain the failure of simvastatin to reduce melanoma incidence in clinical trials and meta-analyses.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Apoptosis/efectos de los fármacos , Interleucina-6/metabolismo , Melanoma/patología , Simvastatina/farmacología , Antineoplásicos/farmacología , Sitios de Unión/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Melanoma/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Cicatrización de Heridas/efectos de los fármacos , Proteína bcl-X/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 386(7): 605-17, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23564041

RESUMEN

Tumours, which are initially sensitive to cytotoxic agents, often develop resistance to a broad spectrum of structurally unrelated drugs. The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors have been shown to inhibit ATP-binding cassette (ABC) transporters but have also impact on glycosylation of such proteins. Doxorubicin is a substrate for ABC transporters like P-glycoprotein (ABCB1) which is present in human RD rhabdomyosarcoma cells. It was therefore the aim of this study to identify the compartmentalisation and action of doxorubicin in simvastatin-treated RD cells. Due to autofluorescence of doxorubicin, intracellular distribution was monitored by confocal microscopy. The biological effects were traced on the level of colony formation, caspase activation and DNA injury. Here we show that simvastatin treatment leads to ABCB1 inhibition and down-regulation of the transporter. Consequently, these cells accumulate significant amounts of doxorubicin, predominantly in the nucleus and lysosomes. While clearance of the anthracycline into lysosomes is not altered by simvastatin treatment, it significantly enhanced nuclear accumulation in a HMG-CoA reductase-independent manner. Thus, in such treated cells, topoisomerase II activity is significantly inhibited, which is further corroborated by augmented double-strand DNA breaks. Moreover, colony formation was synergistically inhibited by the combination of simvastatin and doxorubicin. Given the fact that ABCB1 expression correlates with an adverse prognosis in many tumours, adjuvant chemotherapy including statins might represent a novel therapeutic concept to overcome ABCB1-mediated multidrug resistance by direct inhibition and down-regulation.


Asunto(s)
Adyuvantes Farmacéuticos/administración & dosificación , Doxorrubicina/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Simvastatina/administración & dosificación , Inhibidores de Topoisomerasa II/administración & dosificación , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo II/metabolismo , Histonas/metabolismo , Humanos , Ácido Mevalónico/administración & dosificación , Rabdomiosarcoma/metabolismo
5.
Eur J Cancer ; 49(15): 3366-75, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23790465

RESUMEN

KP1339 is a promising ruthenium-based anticancer compound in early clinical development. This study aimed to test the effects of KP1339 on the in vitro and in vivo activity of the multi-kinase inhibitor sorafenib, the current standard first-line therapy for advanced hepatoma. Anticancer activity of the parental compounds as compared to the drug combination was tested against a panel of cancer cell lines with a focus on hepatoma. Combination of KP1339 with sorafenib induced in the majority of all cases distinctly synergistic effects, comprising both sorafenib-resistant as well as sorafenib-responsive cell models. Several mechanisms were found to underlie these multifaceted synergistic activities. Firstly, co-exposure induced significantly enhanced accumulation levels of both drugs resulting in enhanced apoptosis induction. Secondly, sorafenib blocked KP1339-mediated activation of P38 signalling representing a protective response against the ruthenium drug. In addition, sorafenib treatment also abrogated KP1339-induced G2/M arrest but resulted in check point-independent DNA-synthesis block and a complete loss of the mitotic cell populations. The activity of the KP1339/sorafenib combination was evaluated in the Hep3B hepatoma xenograft. KP1339 monotherapy led to a 2.4-fold increase in life span and, thus, was superior to sorafenib, which induced a 1.9-fold prolonged survival. The combined therapy further enhanced the mean survival by 3.9-fold. Synergistic activity was also observed in the VM-1 melanoma xenograft harbouring an activating braf mutation. Together, our data indicate that the combination of KP1339 with sorafenib displays promising activity in vitro and in vivo especially against human hepatoma models.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Niacinamida/análogos & derivados , Compuestos Organometálicos/farmacología , Compuestos de Fenilurea/farmacología , Rutenio/farmacología , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Xenoinjertos , Humanos , Ratones , Ratones SCID , Niacinamida/administración & dosificación , Niacinamida/farmacología , Compuestos Organometálicos/administración & dosificación , Compuestos de Fenilurea/administración & dosificación , Sorafenib , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA