Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Invest New Drugs ; 32(6): 1071-82, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25064374

RESUMEN

Irinotecan is a water-soluble camptothecin derivative with clinical activity against colorectal and small cell lung cancers and is currently a standard of care therapeutic in the treatment of colorectal cancer in combination with 5-fluorouracil. One of the major clinical issues limiting the use of irinotecan is gastrointestinal toxicity manifested as life-threatening diarrhea which is reported in up to 45% of treated patients. The studies summarized here tested, in a rat model of irinotecan-associated gastro-intestinal toxicity, whether a lipid nanoparticle formulation of irinotecan, Irinophore C™, mitigated early-onset or late-onset diarrhea when given at doses equivalent to unformulated irinotecan that engenders both early- and late-onset diarrhea. Specifically, rats administered intravenously on two consecutive days with unformulated irinotecan at 170 mg/kg then 160 mg/kg experienced transient early-onset diarrhea after each administration and then experienced significant late-onset diarrhea peaking 4 days after treatment. Irinophore C™ given at the identical dose and schedule did not elicit either early- or late-onset diarrhea in any animals. When Irinophore C™ was combined with 5-fluorouracil there was also no early- or late-onset diarrhea observed. Histopathological analysis of the gastro-intestinal tract confirmed that the effects associated with irinotecan treatment were absent in rats given Irinophore C™ at the identical dose. Pharmacokinetic analysis demonstrated significantly higher systemic concentrations of irinotecan in rats given the nanoparticle formulation compared to those given unformulated irinotecan. These results demonstrate that the Irinophore C™ formulation is significantly less toxic than irinotecan, used either as a single agent or in combination with 5-fluorouracil, in a rat model of irinotecan-induced gastrointestinal toxicity.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Camptotecina/análogos & derivados , Diarrea/prevención & control , Nanopartículas/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/sangre , Antineoplásicos Fitogénicos/farmacocinética , Camptotecina/administración & dosificación , Camptotecina/efectos adversos , Camptotecina/sangre , Camptotecina/farmacocinética , Colesterol/química , Colon/patología , Diarrea/inducido químicamente , Diarrea/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Fluorouracilo/administración & dosificación , Intestino Delgado/patología , Irinotecán , Liposomas , Fosfatidilcolinas/química , Ratas Sprague-Dawley
2.
PLoS One ; 8(4): e62349, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626804

RESUMEN

PURPOSE: To investigate the use of liposomal irinotecan (Irinophore C™) plus or minus 5-fluorouracil (5-FU) for the treatment of colorectal cancer. EXPERIMENTAL DESIGN: The effect of irinotecan (IRI) and/or 5-FU exposure times on cytotoxicity was assessed in vitro against HT-29 or LS174T human colon carcinoma cells. The pharmacokinetics and biodistribution of Irinophore C™ (IrC™) and 5-FU, administered alone or in combination, were compared in vivo. A subcutaneous model of HT-29 human colorectal cancer in Rag2-M mice was utilized to assess the efficacy of IrC™ alone, and in combination with 5-FU. RESULTS: The cytotoxicity of IRI and 5-FU were strongly dependent on exposure time. Synergistic interactions were observed following prolonged exposure to IRI/5-FU combinations. Pharmacokinetics/biodistribution studies demonstrated that the 5-FU elimination rate was decreased significantly when 5-FU was co-administered intravenously with IrC™, versus alone. Significant decreases in 5-FU elimination were also observed in plasma, with an associated increase of 5-FU in some tissues when 5-FU was given by intraperitoneal injection and IrC™ was given intravenously. The elimination of IrC™ was not significantly different when administered alone or in combination with 5-FU. Therapeutic studies demonstrated that single agent IrC™ was significantly more effective than the combination of IRI/5-FU; surprisingly, IrC™/5-FU combinations were no more effective than IrC™ alone. The administration of combinations of 5-FU (16 mg/kg) and IrC™ (60 mg IRI/kg) showed increased toxicity when compared to IrC™ alone. Treatment with IrC™ alone (60 mg IRI/kg) delayed the time required for a 5-fold increase in initial tumor volume to day 49, compared to day 23 for controls. When IrC™ (40 mg IRI/kg) was used in combination with 5-FU (16 mg/kg), the time to increase tumor volume 5-fold was 43 days, which was comparable to that achieved when using IrC™ alone (40 mg IRI/kg). CONCLUSIONS: Single agent IrC™ was well tolerated and has significant therapeutic potential. IrC™ may be a suitable replacement for IRI treatment, but its use with free 5-FU is complicated by IrC™-engendered changes in 5-FU pharmacokinetics/biodistribution which are associated with increased toxicity when using the combination.


Asunto(s)
Camptotecina/análogos & derivados , Neoplasias Colorrectales/patología , Fluorouracilo/farmacología , Animales , Camptotecina/administración & dosificación , Camptotecina/farmacología , Camptotecina/toxicidad , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/toxicidad , Células HT29 , Humanos , Irinotecán , Liposomas , Masculino , Ratones , Carga Tumoral/efectos de los fármacos
4.
Nanomedicine (Lond) ; 6(9): 1645-54, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22077466

RESUMEN

A number of studies have outlined the antiangiogenic effects of cytotoxic agents when administered frequently at low doses. These studies suggest that the effect of the cytotoxic agent is on the vasculature within the tumor and it is assumed that there is little or negligible cytotoxicity. Liposomal drug delivery systems have the ability to provide a dual mechanism of activity where tumor accumulation can deliver high local concentrations of the drug at the site of action with concomitant slow release of the drug from carriers in the blood compartment that results in antivascular effects, similar to that achieved when dosing frequently at low levels. Although this dual mechanism of activity may be linked to other lipid nanoparticle formulations of anticancer drugs, this article summarizes the evidence supporting direct (cytotoxic) and indirect (antivascular) actions of a liposomal formulation of irinotecan.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Lípidos/química , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Camptotecina/análogos & derivados , Química Farmacéutica/métodos , Humanos , Irinotecán , Liposomas/química , Modelos Biológicos , Modelos Químicos , Neoplasias/tratamiento farmacológico
7.
Mol Cancer Ther ; 7(1): 59-70, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18202010

RESUMEN

Integrin-linked kinase (ILK) was assesed as a therapeutic target in glioblastoma xenograft models through multiple endpoints including treatment related changes in the tumor microenvironment. Glioblastoma cell lines were tested in vitro for sensitivity toward the small-molecule inhibitors QLT0254 and QLT0267. Cell viability, cell cycle, and apoptosis were evaluated using MTT assay, flow cytometry, caspase activation, and DAPI staining. Western blotting and ELISA were used for protein analysis (ILK, PKB/Akt, VEGF, and HIF-1alpha). In vivo assessment of growth rate, cell proliferation, BrdUrd, blood vessel mass (CD31 labeling), vessel perfusion (Hoechst 33342), and hypoxia (EF-5) was done using U87MG glioblastoma xenografts in RAG2-M mice treated orally with QLT0267 (200 mg/kg q.d.). ILK inhibition in vitro with QLT0254 and QLT0267 resulted in decreased levels of phospho-PKB/Akt (Ser473), secreted VEGF, G2-M block, and apoptosis induction. Mice treated with QLT0267 exhibited significant delays in tumor growth (treated 213 mm3 versus control 549 mm3). In situ analysis of U87MG tumor cell proliferation from QLT0267-treated mice was significantly lower relative to untreated mice. Importantly, VEGF and HIF-1alpha expression decreased in QLT0267-treated tumors as did the percentage of blood vessel mass and numbers of Hoechst 33342 perfused tumor vessels compared with control tumors (35% versus 83%). ILK inhibition with novel small-molecule inhibitors leads to treatment-associated delays in tumor growth, decreased tumor angiogenesis, and functionality of tumor vasculature. The therapeutic effects of a selected ILK inhibitor (QLT0267) should be determined in the clinic in cancers that exhibit dysregulated ILK, such as PTEN-null glioblastomas.


Asunto(s)
Glioblastoma/enzimología , Glioblastoma/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/irrigación sanguínea , Glioblastoma/tratamiento farmacológico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Estructura Molecular , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cancer Ther ; 6(3): 844-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17339368

RESUMEN

Various methods have been explored to enhance antibody-based cancer therapy. The use of multivalent antibodies or fragments against tumor antigens has generated a great deal of interest, as various cellular signals, including induction of apoptosis, inhibition of cell growth/survival, or internalization of the surface molecules, can be triggered or enhanced on extensive cross-linking of the target/antibody complex by the multivalent form of the antibody. The goal of the studies reported here was to develop multivalent antibody constructs via grafting of antibody molecules onto liposome membranes to enhance antibody activity. Using trastuzumab and rituximab as examples, up to a 25-fold increase in the antibody potency in cell viability assay was observed when the antibodies were presented in the multivalent liposome formulation. Key cell survival signaling molecules, such as phosphorylated Akt and phosphorylated p65 nuclear factor-kappaB, were down-regulated on treatment with multivalent liposomal trastuzumab and liposomal rituximab, respectively. Potent in vivo antitumor activity was shown for liposomal trastuzumab. The data presented here showed the potential of liposome technology to enhance the therapeutic effect of antibodies via a mechanism that modulates cell survival through clustering of the target/antibody complex.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/terapia , Animales , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Anticuerpos Antineoplásicos , Antígenos CD20/inmunología , Antígenos de Neoplasias/inmunología , Western Blotting , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo , Femenino , Citometría de Flujo , Genes erbB-2/genética , Genes erbB-2/inmunología , Humanos , Liposomas , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/inmunología , Rituximab , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Trastuzumab
10.
Pharm Res ; 22(6): 915-22, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15948035

RESUMEN

PURPOSE: We assessed the combination of doxorubicin or liposomal doxorubicin with trastuzumab for alterations in peak serum drug levels, as these agents are increasingly being paired in the treatment of aggressive breast cancer. We hypothesized that trastuzumab would exhibit a slower rate of elimination from the serum when in combination with liposomal doxorubicin based on the known effects of liposomal doxorubicin on phagocytic cells of the mononuclear phagocyte system (MPS), which are responsible in part for the uptake and degradation of antibodies. METHODS: Doxorubicin and trastuzumab serum levels were assessed following injection of free doxorubicin, liposomal doxorubicin, or trastuzumab into female RAG2-M mice bearing subcutaneous MCF-7(HER-2) tumors. The effects of combination drug treatment on tumor growth were compared to single-agent treatment. RESULTS: Peak serum trastuzumab levels were not altered as a result of addition of doxorubicin therapy, nor were doxorubicin levels altered over 24 h as a result of coadministration of trastuzumab. Liposomal doxorubicin administration did result in serum doxorubicin levels 200- to 1000-fold higher than with injection of free doxorubicin. CONCLUSIONS: For the specific combination of trastuzumab with doxorubicin, either in free or liposomal form, coadministered in mice, there was no impact of one drug on the other in terms of peak serum drug levels or efficacy.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/sangre , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Doxorrubicina/administración & dosificación , Doxorrubicina/sangre , Combinación de Medicamentos , Interacciones Farmacológicas , Ensayo de Inmunoadsorción Enzimática , Femenino , Indicadores y Reactivos , Inyecciones Intravenosas , Liposomas , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/patología , Trasplante Heterólogo , Trastuzumab
11.
Methods Enzymol ; 391: 40-57, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15721373

RESUMEN

Vincristine is a dimeric Catharanthus alkaloid derived from the Madagascan periwinkle that acts by binding to tubulin and blocking metaphase in actively dividing cells. While vincristine is widely used in the treatment of a number of human carcinomas, its use is associated with dose-limiting neurotoxicity, manifested mainly as peripheral neuropathy. It is known that the therapeutic activity of vincristine can be significantly enhanced after its encapsulation in appropriately designed liposomal systems. Enhanced efficacy is also associated with a slight decrease in drug toxicity. Thus, the therapeutic index of vincristine can be enhanced significantly through the use of a liposomal delivery system. Vincristine may be encapsulated into liposomes of varying lipid composition by several techniques, including passive loading, pH gradient loading, and ionophore-assisted loading. However, most research has focused on the encapsulation of vincristine in response to a transbilayer pH gradient, which actively concentrates the drug within the aqueous interior of the liposome. This chapter details the preparation and evaluation of liposomal vincristine. Specifically, we elaborate on the components (choice of lipids, molar proportions, etc.), methods (preparation of liposomes, drug loading methods, etc.), critical design features (size, surface charge, etc.), and key biological endpoints (circulation lifetime, bioavailability, efficacy measurements) important to the development of a formulation of vincristine with enhanced therapeutic properties.


Asunto(s)
Antineoplásicos Fitogénicos , Portadores de Fármacos , Liposomas , Vincristina , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Humanos , Concentración de Iones de Hidrógeno , Ionóforos/metabolismo , Lípidos/química , Liposomas/síntesis química , Liposomas/química , Estructura Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Vincristina/administración & dosificación , Vincristina/química , Vincristina/farmacocinética , Vincristina/uso terapéutico
12.
Methods Enzymol ; 391: 71-97, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15721375

RESUMEN

Doxorubicin is the best known and most widely used member of the anthracycline antibiotic group of anticancer agents. It was first introduced in the 1970s, and since that time has become one of the most commonly used drugs for the treatment of both hematological and solid tumors. The therapy-limiting toxicity for this drug is cardiomyopathy, which may lead to congestive heart failure and death. Approximately 2% of patients who have received a cumulative (lifetime) doxorubicin dose of 450-500 mg?m(2) will experience this condition. An approach to ameliorating doxorubicin-related toxicity is to use drug carriers, which engender a change in the pharmacological distribution of the drug, resulting in reduced drug levels in the heart. Examples of these carrier systems include lipid-based (liposome) formulations that effect a beneficial change in doxorubicin biodistribution, with two formulations approved for clinical use. Drug approval was based, in part, on data suggesting that beneficial changes in doxorubicin occurred in the absence of decreased therapeutic activity. Preclinical (animal) and clinical (human) studies showing that liposomes can preferentially accumulate in tumors have provided a rationale for improved activity. Liposomes represent ideal drug delivery systems, as the microvasculature in tumors is typically discontinuous, having pore sizes (100-780 nm) large enough for liposomes to move from the blood compartment into the extravascular space surrounding the tumor cells. Liposomes, in the size range of 100-200 nm readily extravasate within the site of tumor growth to provide locally concentrated drug delivery, a primary role of liposomal formulation. Although other liposomal drugs have been prepared and characterized due to the potential for liposomes to improve antitumor potency of the encapsulated drug, the studies on liposomal doxorubicin have been developed primarily to address issues of acute and chronic toxicity that occur as a consequence of using this drug. It is important to recognize that research programs directed toward the development of liposomal doxorubicin occurred concurrently with synthetic chemistry programs attempting to introduce safer and more effective anthracycline analogues. Although many of these drugs are approved for use, and preliminary liposomal formulations of these analogues have been prepared, doxorubicin continues to be a mainstay of drug cocktails used in the management of most solid tumors. It will be of great interest to observe how the approved formulations of liposomal doxorubicin are integrated into combination regimes for treatment of cancer. In the meantime, we have learned a great deal about liposomes as drug carriers from over 20 years of research on different liposomal doxorubicin formulations, the very first of which were identified in the late 1970s. This chapter will discuss the various methods for encapsulation of doxorubicin into liposomes, as well as some of the important interactions between the formulation components of the drug and how this may impact the biological activity of the associated drug. This review of methodology, in turn, will highlight research activities that are being pursued to achieve better performance parameters for liposomal formulations of doxorubicin, as well as other anticancer agents being considered for use with lipid-based carriers.


Asunto(s)
Antibióticos Antineoplásicos/metabolismo , Doxorrubicina/metabolismo , Portadores de Fármacos , Liposomas , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/química , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/química , Humanos , Iones , Liposomas/química , Estructura Molecular , Fosfolípidos/química
13.
Mol Cancer Res ; 2(11): 606-19, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15561777

RESUMEN

The effects of HER-2/neu overexpression on the tumor microenvironment in an aggressive breast cancer xenograft model were investigated. These studies focused on tumors derived following the subcutaneous injection of MDA-MB-435/LCC6 cells transfected with human c-erbB2 (LCC6(HER-2)) into SCID-Rag2M mice. LCC6(HER-2) tumors were more viable (H&E-stained tumor sections) than isogenic vector control tumors (LCC6(Vector)). Correspondingly, a 2.7-fold increase in trypan blue-excluding cells (P = 0.00056) and a 4.8-fold increase in clonogenic cells (P = 0.00146) were noted in cell suspensions derived from disaggregated LCC6(HER-2) versus LCC6(Vector) tumors. Tumor sections stained with the antibody detecting 2-(2-nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl)-acetamide (EF5), a marker of hypoxia, showed a greater fraction of hypoxic tissue in LCC6(HER-2) tumors compared with control tumors. Flow cytometric analyses based on viable tumor cells (DNA content >/= 2N) in cell suspensions from disaggregated tumors confirmed that there were significantly more EF5-positive cells (i.e., hypoxic) in LCC6(HER-2) than in LCC6(Vector) tumors (16.41 +/- 8.1% and 5.96 +/- 4.1%, respectively; P = 0.0015). Protein levels of phosphorylated (Ser(536)) nuclear factor-kappaB p65 were significantly elevated in LCC6(HER-2) tumors (P = 0.00048), and a trend in increased hypoxia-inducible factor-1alpha protein levels was observed in LCC6(HER-2) compared with LCC6(Vector) tumors. Despite the substantial viable hypoxic cell fraction and a 1.7-fold increase of vascular endothelial growth factor protein (P = 0.05) in LCC6(HER-2) tumors, no significant differences were found (P > 0.05) between LCC6(HER-2) and LCC6(Vector) vasculature (CD31 staining and Hoechst 33342 perfusion). These results suggest that HER-2/neu overexpression may be linked with overall increased tumor viability and a significant increase in the population of viable hypoxic cells, which is not due to differences in tumor vascularization.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Etanidazol/análogos & derivados , Neovascularización Patológica/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Adaptación Fisiológica/genética , Animales , Biomarcadores , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/genética , Carcinoma/irrigación sanguínea , Carcinoma/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral/trasplante , Proliferación Celular , Respiración de la Célula/genética , Supervivencia Celular/genética , Células Clonales/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Supervivencia de Injerto/fisiología , Humanos , Hidrocarburos Fluorados , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones SCID , FN-kappa B/metabolismo , Factor de Transcripción ReIA , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Cancer Biol Ther ; 3(2): 197-204, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14726684

RESUMEN

The nature of anti-cancer therapeutics is currently undergoing a paradigm change, with biologic agents slowly being introduced into the therapeutic armory, displacing or complimenting the traditionally used cytotoxic agents. These new agents include monoclonal antibodies, recombinant DNA, antisense oligonucleotides (ASO) and others. To assess the new therapeutics, new predictive models are required. Utilizing the MDA435/LCC6 human breast cancer xenograft model, the pharmacokinetic behavior of antisense oligonucleotides targeted against vascular endothelial growth factor and HER-2/neu was assessed. For pharmacodynamic analysis, ASO in buffer or encapsulated in a liposomal formulation were injected intravenously or intraperitoneally into MDA435/LCC6 ascites tumor-bearing mice. Plasma antisense elimination, tissue distribution, total peritoneal antisense and peritoneal cell associated antisense levels were determined. Liposomal encapsulation led to significant decreases in the plasma elimination rate, as evidenced by an approximate 10-fold increase in mean AUC over 24 hours, as well as enhanced peritoneal cell delivery in mice bearing ascites tumors. Tissue distribution studies of both free and liposome encapsulated ASO indicated that ASO distribution was dictated primarily by the liposomal carrier when administered in liposomal form.


Asunto(s)
Ascitis/tratamiento farmacológico , Liposomas , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Oligonucleótidos Antisentido/uso terapéutico , Receptor ErbB-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacocinética , Animales , Ascitis/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Humanos , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones SCID , Ratones Transgénicos , Proteínas Nucleares , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética
15.
J Exp Ther Oncol ; 3(5): 261-71, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14696623

RESUMEN

This study assessed the in vivo therapeutic activity of an antisense molecule targeted against HER-2/neu expressing mRNA. Antisense activity was evaluated in female SCID/Rag2m mice bearing subcutaneous tumors derived from HER-2/neu-transfected MDA-MB-435 (MDA-MB-435(HER2)) cells, a transfected line derived from the human breast cancer MDA-MB-435 cell line. Animals were treated with free or liposome-encapsulated antisense. The area under the curve (AUC(0-24h)) of the liposomal formulated antisense was demonstrated to be more than 30-fold greater than that of free antisense following intravenous administration. Efficacy was determined by assessing changes in tumor growth rate as well as by an immunohistological end-point evaluating HER-2/neu expression. HER-2/neu protein expression was reduced in mice bearing HER-2/neu-transfected MDA-MB-435 tumors when treated with liposomal antisense. However, tumors in these mice grew at a faster rate than the control, a result that was interpreted to be a consequence of selection of a more rapidly proliferating HER-2/neu-negative subpopulation of cells. Effective control of the MDA-MB-435(HER2) tumors was achieved when antisense treatment was combined with doxorubicin. Tumors derived from animals treated with the combination of doxorubicin and the liposomal antisense against HER-2/neu exhibited no detectable levels of HER-2/neu expression. Antisense targeted against HER-2/neu mRNA was effective in reducing or eliminating HER-2/neu protein expression, and when combined wtih doxorubicin treatment was efficacious in the treatment of mice bearing HER-2/neu-overexpressing human xenograft tumors.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Oligonucleótidos Antisentido/uso terapéutico , Receptor ErbB-2/genética , Animales , Área Bajo la Curva , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Quimioterapia Combinada , Femenino , Citometría de Flujo , Humanos , Liposomas , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones SCID , Proteínas Nucleares , Receptor ErbB-2/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...