Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 7: 40142, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28071663

RESUMEN

Prosthetic grafts and patches are commonly used in cardiovascular surgery, however neointimal hyperplasia remains a significant concern, especially under low flow conditions. We hypothesized that delivery of rapamycin from nanoparticles (NP) covalently attached to patches allows sustained site-specific delivery of therapeutic agents targeted to inhibit localized neointimal hyperplasia. NP were covalently linked to pericardial patches using EDC/NHS chemistry and could deliver at least 360 ng rapamycin per patch without detectable rapamycin in serum; nanoparticles were detectable in the liver, kidney and spleen but no other sites within 24 hours. In a rat venous patch angioplasty model, control patches developed robust neointimal hyperplasia on the patch luminal surface characterized by Eph-B4-positive endothelium and underlying SMC and infiltrating cells such as macrophages and leukocytes. Patches delivering rapamycin developed less neointimal hyperplasia, less smooth muscle cell proliferation, and had fewer infiltrating cells but retained endothelialization. NP covalently linked to pericardial patches are a novel composite delivery system that allows sustained site-specific delivery of therapeutics; NP delivering rapamycin inhibit patch neointimal hyperplasia. NP linked to patches may represent a next generation of tissue engineered cardiovascular implants.


Asunto(s)
Angioplastia/métodos , Inhibidores de Crecimiento/administración & dosificación , Hiperplasia/prevención & control , Nanopartículas/administración & dosificación , Neointima/patología , Sirolimus/administración & dosificación , Trasplantes/cirugía , Animales , Portadores de Fármacos/administración & dosificación , Inhibidores de Crecimiento/farmacocinética , Histocitoquímica , Hiperplasia/patología , Ratas , Sirolimus/farmacocinética , Resultado del Tratamiento
2.
Eur J Pharm Sci ; 97: 70-78, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27816627

RESUMEN

The purpose of this study was to establish a population pharmacokinetic/pharmacodynamic (PK/PD) model linking etoposide free tumor and total plasma concentrations to the inhibition of solid tumor growth in rats. Walker-256 tumor cells were inoculated subcutaneously in the right flank of Wistar rats, which were randomly divided in control and two treated groups that received etoposide 5 or 10mg/kg i.v. bolus every day for 8 and 4days, respectively, and tumor volume was monitored daily for 30days. The plasma and intratumoral concentrations-time profiles were obtained from a previous study and were modeled by a four-compartment population pharmacokinetic (popPK) model. PK/PD analysis was conducted using MONOLIX v.4.3.3 on average data and by mean of a nonlinear mixed-effect model. PK/PD data were analyzed using a modification of Simeoni Tumor Growth Inhibition (TGI) model by introduction of an Emax function to take into account the concentration dependency of k2variable parameter (variable potency). The Simeoni TGI-Emax model was capable to fit schedule-dependent antitumor effects using the tumor growth curves from the control and two different administered schedules. The PK/PD model was capable of describing the tumor growth inhibition using total plasma or free tumor concentrations, resulting in higher k2max (maximal potency) for free concentrations (25.8mL·µg-1·day-1 - intratumoral vs. 12.6mL·µg-1·day-1 total plasma). These findings indicate that the plasma concentration may not be a good surrogate for pharmacologically active free tumor concentrations, emphasizing the importance of knowing drug tumor penetration to choose the best antitumor therapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacocinética , Carcinoma 256 de Walker/metabolismo , Modelos Animales de Enfermedad , Etopósido/farmacocinética , Inhibidores de Crecimiento/farmacocinética , Carga Tumoral/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma 256 de Walker/tratamiento farmacológico , Carcinoma 256 de Walker/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Etopósido/uso terapéutico , Inhibidores de Crecimiento/uso terapéutico , Masculino , Ratas , Ratas Wistar , Carga Tumoral/fisiología
3.
J Interferon Cytokine Res ; 34(10): 759-68, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24841172

RESUMEN

Interferon gamma (IFN-γ) is a 28 kDa homodimeric cytokine that exhibits potent immunomodulatory, anti-proliferative, and antiviral properties. The protein is used to treat chronic granulomatous disease and malignant osteopetrosis, and it is under investigation as a treatment for a variety of cancer, fungal and viral diseases. IFN-γ has a short circulating half life in vivo, which necessitates frequent administration to patients. An unusual feature of IFN-γ is that the protein contains no native cysteines. To create a longer-acting and potentially more effective form of the protein, we introduced a cysteine residue into the IFN-γ coding sequence at amino acid position 103, which is located in a surface-exposed, non-helical region of the protein. The added cysteine residue served as the site for targeted modification of the protein with a cysteine-reactive polyethylene glycol (PEG) reagent. The recombinant protein was expressed in bacteria, purified and modified with 10, 20, and 40 kDa maleimide PEGs. The purified, PEGylated proteins had in vitro bioactivities comparable to IFN-γ, as measured using an in vitro cell growth inhibition assay. The PEGylated proteins displayed 20- to 32-fold longer half lives than IFN-γ in rats, and they were significantly more effective than IFN-γ at inhibiting growth of a human tumor xenograft in athymic mice.


Asunto(s)
Cisteína/química , Inhibidores de Crecimiento/farmacocinética , Interferón gamma/farmacocinética , Polietilenglicoles/química , Animales , Línea Celular Tumoral , Cisteína/genética , Femenino , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/farmacología , Humanos , Inmunomodulación , Interferón gamma/química , Interferón gamma/farmacología , Masculino , Ratones , Ratones Desnudos , Mutación/genética , Unión Proteica , Conformación Proteica , Ingeniería de Proteínas , Estabilidad Proteica , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biochem Pharmacol ; 83(6): 715-22, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22222427

RESUMEN

Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/farmacocinética , Aspirina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Inhibidores de Crecimiento/farmacocinética , Sulfuro de Hidrógeno/metabolismo , Ibuprofeno/análogos & derivados , Naproxeno/análogos & derivados , Sulindac/análogos & derivados , Tionas/farmacocinética , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aspirina/química , Aspirina/farmacocinética , Línea Celular Tumoral , Inhibidores de Crecimiento/química , Humanos , Sulfuro de Hidrógeno/química , Ibuprofeno/química , Ibuprofeno/farmacocinética , Naproxeno/química , Naproxeno/farmacocinética , Sulindac/química , Sulindac/farmacocinética , Tionas/química
5.
Nutr Cancer ; 61(4): 538-43, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19838926

RESUMEN

The effect of macelignan, a phytoestrogen, on P-gp function was investigated using multidrug resistant cancer cells overexpressing P-gp (NCI/ADR-RES) and the fluorescent P-gp substrates, daunorubicin and rhodamine 123. Macelignan (40 microM) increased the cellular accumulation of daunorubicin by approximately threefold in NCI/ADR-RES cells, whereas it did not alter the cellular accumulation of daunorubicin in MCF-7/sensitive cells. Similarly, the presence of macelignan also enhanced significantly (P < 0.05) the cellular accumulation of rhodamine 123 in a concentration-dependent manner in NCI/ADR-RES cells. Furthermore, cancer cells were more susceptible to the cytotoxicity of vinblastine, a P-gp substrate, in the presence of macelignan. Those results suggest that macelignan has inhibitory effects on P-gp mediated cellular efflux. However, P-gp activity did not affect the cellular accumulation of macelignan itself. Taken all together, macelignan was identified as a novel inhibitor of P-gp activity and may be a promising lead compound for the rational design of more efficacious drugs to reverse multidrug resistance in cancer.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos , Lignanos/farmacología , Adenocarcinoma , Análisis de Varianza , Antineoplásicos Fitogénicos/farmacocinética , Transporte Biológico Activo/efectos de los fármacos , Neoplasias de la Mama , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daunorrubicina/farmacocinética , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Resistencia a Múltiples Medicamentos , Sinergismo Farmacológico , Femenino , Colorantes Fluorescentes/farmacocinética , Inhibidores de Crecimiento/farmacocinética , Inhibidores de Crecimiento/farmacología , Humanos , Lignanos/farmacocinética , Fitoestrógenos/farmacocinética , Fitoestrógenos/farmacología , Fitoterapia , Rodamina 123/farmacocinética , Vinblastina/farmacocinética
6.
Cancer Chemother Pharmacol ; 64(2): 425-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19280191

RESUMEN

PURPOSE: The toxicities, pharmacokinetics and recommended dose of oral once daily ZK 304709, a novel multi-targeted growth inhibitor (MTGI) with activity against cell-cycle progression and angiogenesis, was investigated in patients by administration for 14 consecutive days followed by 14 days recovery. METHODS: Patients with solid tumours resistant to standard treatments were enrolled in an accelerated titration design. RESULTS: Thirty-seven patients received ZK 304709 from 15 to 285 mg daily. The most common drug-related adverse events were vomiting, diarrhoea and fatigue. Systemic exposure to ZK 304709 increased with dose up to 90 mg daily but plateaued thereafter, with high inter-individual variability at all doses. Thirteen patients had stable disease as best response as per RECIST criteria. CONCLUSIONS: There was no increase in exposure to ZK 304709 with dose escalation above 90 mg, and the MTD was not determined. This study illustrates the importance of phase I pharmacokinetic data to guide dose escalation and drug development.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Antineoplásicos/farmacocinética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inhibidores de Crecimiento/farmacocinética , Neoplasias/metabolismo , Administración Oral , Adulto , Anciano , Inhibidores de la Angiogénesis/efectos adversos , Antineoplásicos/efectos adversos , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Crecimiento/efectos adversos , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/patología , Pronóstico , Seguridad , Tasa de Supervivencia , Distribución Tisular , Resultado del Tratamiento
7.
Molecules ; 13(7): 1441-54, 2008 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-18719516

RESUMEN

A series of amino acid monoester prodrugs of floxuridine was synthesized and evaluated for the improvement of oral bioavailability and the feasibility of target drug delivery via oligopeptide transporters. All floxuridine 5'-amino acid monoester prodrugs exhibited PEPT1 affinity, with inhibition coefficients of Gly-Sar uptake (IC50) ranging from 0.7 - 2.3 mM in Caco-2 and 2.0 - 4.8 mM in AsPC-1 cells, while that of floxuridine was 7.3 mM and 6.3 mM, respectively. Caco-2 membrane permeabilities of floxuridine prodrugs (1.01 - 5.31 x 10(-6 )cm/sec) and floxuridine (0.48 x 10(-6 )cm/sec) were much higher than that of 5-FU (0.038 x 10(-6) cm/sec). MDCK cells stably transfected with the human oligopeptide transporter PEPT1 (MDCK/hPEPT1) exhibited enhanced cell growth inhibition in the presence of the prodrugs. This prodrug strategy offers great potential, not only for increased drug absorption but also for improved tumor selectivity and drug efficacy.


Asunto(s)
Aminoácidos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Floxuridina/farmacología , Floxuridina/farmacocinética , Profármacos/farmacología , Profármacos/farmacocinética , Simportadores/metabolismo , Absorción/efectos de los fármacos , Aminoácidos/síntesis química , Aminoácidos/farmacocinética , Animales , Antimetabolitos Antineoplásicos/síntesis química , Células CACO-2 , Perros , Sistemas de Liberación de Medicamentos , Ésteres , Inhibidores de Crecimiento/farmacocinética , Inhibidores de Crecimiento/farmacología , Humanos , Transportador de Péptidos 1 , Profármacos/síntesis química , Simportadores/biosíntesis , Simportadores/genética
8.
Curr Pharm Des ; 12(26): 3411-21, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17017935

RESUMEN

Dehydroepiandrosterone (DHEA) is the most abundant adrenal androgenic steroid in young adult humans. The physiological functions of DHEA in preventing human carcinogenesis are still controversial, but a lot of reports have shown that pharmacological doses of DHEA show chemopreventive and anti-proliferative effects on tumors in rodents. Although a therapeutic dose of DHEA has been reported to promote hepatocarcinogenesis in rats due to peroxisomal proliferation, it remains unclear whether DHEA is a peroxisome proliferator in human liver. The chemopreventive and anti-proliferative effects of DHEA are not explained by a single mechanism, and at least four mechanisms seem to contribute to these effects: 1) depletion of NADPH and ribose-5-phosphate due to the inhibition of glucose-6-phosphate dehydrogenase activity, 2) suppression of cholesterol biosynthetic pathway by inhibition of HMG-CoA reductase, 3) interference with cell proliferation signaling pathways, and 4) suppression of nitric oxide generation through down-regulation of nitric oxide synthase II. In addition to studies of the mechanisms underlying the anti-neoplastic effects, searches for more potent and less androgenic DHEA derivatives are ongoing. A small amount of DHEA is endogenously metabolized to 7-oxygenated DHEA, and this may represent a metabolic pathway to more potent steroid hormones. Androsterone, epiandrosterone and etiocholanolone have been considered to be merely inactive end products of DHEA, but may in fact be physiological effectors in their own right. In addition, DHEA analogs such as 3beta-methyl-5-androsten-17-one, 16alpha-fluoro-5-androsten-17-one and 16alpha-fluoro-5alpha-androstan-17-one have been synthesized and shown to be more effective inhibitors of tumor growth, compared with DHEA itself. However, to design potent and safe DHEA derivatives, identification of the DHEA receptor and clarification of the mechanisms of DHEA action are required.


Asunto(s)
Antineoplásicos/química , Deshidroepiandrosterona/química , Inhibidores de Crecimiento/química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Deshidroepiandrosterona/farmacocinética , Deshidroepiandrosterona/uso terapéutico , Inhibidores de Crecimiento/farmacocinética , Inhibidores de Crecimiento/uso terapéutico , Humanos
9.
Ther Drug Monit ; 27(6): 770-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306853

RESUMEN

We report here a specific, automated LC/LC-MS/MS assay for the quantification of ABT-578 in human and rabbit blood and rabbit tissues for drug-eluting stent development. After protein precipitation, samples were injected into the HPLC system and extracted online using a high flow of 5 mL/min. The extracts were then backflushed onto the analytic column. The [M+Na] of ABT-578 (m/z 988.6-->369.4) and its internal standard sirolimus (m/z 936.5-->409.3) were monitored. Extraction and analysis took 4 minutes. The assay was validated following the US Food & Drug Administration guidelines. Linearity was 0.025-25 ng/mL for most matrices. In human blood, interday accuracies were 81.8% (at 0.025 ng/mL), 91.0% (1 ng/mL), and 99.5% (50 ng/mL), and interday precisions were 10.7% (0.025 ng/mL), 3.0% (1 ng/mL), and 1.8% (50 ng/mL).


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Sirolimus/análogos & derivados , Espectrometría de Masa por Ionización de Electrospray/métodos , Animales , Inhibidores de Crecimiento/sangre , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/farmacocinética , Humanos , Estructura Molecular , Conejos , Reproducibilidad de los Resultados , Sirolimus/sangre , Sirolimus/farmacocinética , Tecnología Farmacéutica/métodos , Distribución Tisular
10.
J Control Release ; 94(1): 63-74, 2004 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-14684272

RESUMEN

Systemic application of anticancer drugs often causes severe toxic side effects. To reduce the undesired effects, advanced drug delivery systems are needed which are based on specific cell targeting vehicles. In this study, bacterial ghosts from Mannheimia haemolytica were used for site-specific delivery of doxorubicin (DOX) to human colorectal adenocarcinoma cells (Caco-2). Bacterial ghosts are non-denatured envelopes of Gram-negative bacteria with fully intact surface structures for specific attachment to mammalian cells. The in vitro release profile of DOX-ghosts demonstrated that the loaded drug was non-covalently associated with the bacterial ghosts and that the drug delivery vehicles themselves represent a slow release system. Adherence studies showed that the M. haemolytica ghosts more efficiently than E. coli ghosts targeted the Caco-2 cells and released the loaded DOX within the cells. Cytotoxicity assays revealed that the DOX-ghosts exhibited potent antiproliferative activities on Caco-2 cells as the DOX associated with ghosts was two magnitude of orders more cytotoxic than free DOX provided in the medium at the same concentrations. Notably, a significant reduction in the cell viability was measured with DOX-ghosts at low DOX concentrations, which had no inhibitory effect when applied as free DOX after incubation for 16 h or when applied at higher concentrations for only 10 min to the cells. As the higher antiproliferative effects of DOX on Caco-2 cells were mediated by the specific drug targeting properties of the bacterial ghosts, the bacterial ghost system represents a novel platform for advanced drug delivery.


Asunto(s)
Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Inhibidores de Crecimiento/administración & dosificación , Mannheimia haemolytica , Células CACO-2 , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacocinética , Inhibidores de Crecimiento/farmacocinética , Humanos , Mannheimia haemolytica/metabolismo
12.
Drug Deliv ; 10(2): 95-100, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12746055

RESUMEN

Some selected lipophilic conjugates of the antifolate drug methotrexate (MTX) with lipoamino acids (LAA), previously described, were incorporated in liposomes with a different composition and charge (neutral, positive, or negative). The properties of the liposomal systems were determined. The inhibitory activity of the conjugates after incorporation in the vesicles was determined in a preliminary assessment against a human erythroleukemic cell line (K562 cells) and compared with the activity of the parent drug and of free conjugates. The influence of liposome surface charge and of the type of conjugate (i.e., in the carboxylic or ester form) on the biological effect is discussed.


Asunto(s)
Aminoácidos Neutros/administración & dosificación , Aminoácidos Neutros/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Preparaciones de Acción Retardada/uso terapéutico , Portadores de Fármacos/farmacocinética , Metotrexato/administración & dosificación , Metotrexato/farmacocinética , Preparaciones de Acción Retardada/administración & dosificación , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Estabilidad de Medicamentos , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/farmacocinética , Humanos , Células K562 , Lípidos/química , Liposomas , Fosfolípidos/química , Tecnología Farmacéutica
13.
Pharm Res ; 19(6): 744-54, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12134943

RESUMEN

PURPOSE: The attainment of effective intracellular delivery remains an important issue for pharmacologic applications of antisense oligonucleotides. Here, we describe the synthesis, binding properties, and biologic properties of peptide-oligonucleotide conjugates comprised of the Tat and Ant cell-penetrating peptides with 2'-O-methyl phosphorothioate oligonucleotides. METHODS: The biologic assay used in this study measures the ability of the antisense molecule to correct splicing of an aberrant intron inserted into the Luciferase gene; thus, this assay clearly demonstrates the delivery of functional antisense molecules to the splicing machinery within the nucleus. The binding affinities of the conjugates to their target sequences were measured by surface plasmon resonance (BIAcor) techniques. RESULTS: The peptide-oligonucleotide conjugates progressively entered cells over a period of hours and were detected in cytoplasmic vesicles and in the nucleus. Peptide-oligonucleotide conjugates targeted to the aberrant splice site, but not mismatched controls, caused an increase in Luciferase activity in a dose-responsive manner. The kinetics of Luciferase appearance were consistent with the course of the uptake process for the conjugates. The effects of peptide conjugation on the hybridization characteristics of the oligonucleotides were also examined using surface plasmon resonance. The peptide-oligonucleotide conjugates displayed binding affinities and selectivities similar to those of unconjugated oligonucleotides. CONCLUSIONS: Conjugation with cell-penetrating peptides enhances oligonucleotide delivery to the nucleus without interfering with the base-pairing function of antisense oligonucleotides.


Asunto(s)
Productos del Gen tat/farmacocinética , Proteínas de Homeodominio/farmacocinética , Proteínas Nucleares , Oligonucleótidos Antisentido/farmacocinética , Factores de Transcripción , Secuencia de Aminoácidos , Proteína con Homeodominio Antennapedia , División Celular/efectos de los fármacos , División Celular/genética , Sistemas de Liberación de Medicamentos/métodos , Productos del Gen tat/administración & dosificación , Productos del Gen tat/genética , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/farmacocinética , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Proteínas de Homeodominio/administración & dosificación , Proteínas de Homeodominio/genética , Humanos , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/genética , Unión Proteica/genética
14.
Cancer Res ; 61(9): 3669-74, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11325837

RESUMEN

The therapeutic efficacy and tumor accumulation of a liposome formulation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU), an effective agent used in the treatment of malignant brain tumors, was examined in an animal tumor model. Pharmacokinetic studies in normal and tumor-bearing rats indicated that a 2-fold greater plasma exposure was achieved with liposome-formulated CCNU compared with the free drug. In Fisher rats bearing s.c. tumors 36B-10, tumor growth was delayed substantially when liposomal CCNU was delivered compared with free-drug treatment. In single-dose treatments of 20, 35, and 50 mg/kg, tumor progression after each dose was reduced approximately 2-fold with liposomal compared with free CCNU (four animals in each treatment group). Multiple-dose treatments (given as three weekly doses with eight animals in each treatment group) with cumulative doses of 80 and 100 mg/kg of free and liposomal CCNU also resulted in a 2-fold reduction in tumor progression when compared with free-drug treatment. When drug levels in tumors relative to plasma were examined, it was observed that tumor drug concentrations did not exceed those found in plasma after administration of free CCNU; after administration of liposomal CCNU, however, tumor concentrations exceeded those in plasma by nearly 10-fold. These results suggest that the increased efficacy of liposome-formulated CCNU may be attributable to enhanced drug accumulation in tumor tissues.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Astrocitoma/tratamiento farmacológico , Neoplasias Encefálicas/tratamiento farmacológico , Lomustina/administración & dosificación , Animales , Antineoplásicos Alquilantes/sangre , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacología , Astrocitoma/sangre , Astrocitoma/metabolismo , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/metabolismo , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/sangre , Inhibidores de Crecimiento/farmacocinética , Inhibidores de Crecimiento/farmacología , Liposomas , Lomustina/sangre , Lomustina/farmacocinética , Lomustina/farmacología , Ratas , Ratas Endogámicas F344
15.
Cancer Res ; 61(5): 1983-90, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280756

RESUMEN

4-demethoxy-3'-deamino-3'-aziridinyl-4'-methylsulphonyl-daunorubicin (PNU-159548) belongs to a novel class of antitumor compounds (termed alkycyclines) and is currently undergoing Phase II clinical trial. In the present study, we investigated the in vitro and in vivo antitumor activity, the pharmacokinetics, and the toxicological profile of this compound. PNU-159548 showed good cytotoxic activity in murine and human cancer cells growing in vitro, with an average concentration for 50% growth inhibition of 15.8 ng/ml. The drug showed strong antitumor efficacy in vivo after i.v. and p.o. administration against rapidly proliferating murine leukemias and slowly growing transplantable human xenografts. At non-toxic doses, PNU-159548 produced complete regression and cures in ovarian, breast, and human small cell lung carcinomas. Fourteen of 16 models studied, including colon, pancreatic, gastric, and renal carcinomas, astrocytoma and melanoma, were found to be sensitive to PNU-159548. In addition, PNU-159548 was effective against intracranially implanted tumors. Toxicological studies revealed myelosuppression as the main toxicity in both mice and dogs. The maximum tolerated doses, after a single administration, were 2.5 mg/kg of body weight in mice, 1.6 mg/kg in rats, and 0.3 mg/kg in dogs. In the cyclic studies, the maximum tolerated doses were 0.18 mg/kg/day (cumulative dose/cycle: 0.54 mg/kg) in rats and 0.05 mg/kg/day (cumulative dose/cycle: 0.15 mg/kg) in dogs. PNU-159548 showed minimal cardiotoxicity, when compared with doxorubicin in the chronic rat model at a dose level inducing similar myelotoxicity. Animal pharmacokinetics, carried out in mice, rats, and dogs, was characterized by high volumes of distribution, plasma clearance of the same order of the hepatic blood flow, and short terminal half-life. These findings support the conclusion that PNU-159548 is an excellent candidate for clinical trials in the treatment of cancer.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Daunorrubicina/farmacología , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Daunorrubicina/análogos & derivados , Daunorrubicina/farmacocinética , Daunorrubicina/toxicidad , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/toxicidad , Femenino , Inhibidores de Crecimiento/farmacocinética , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/toxicidad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Inhibidores de Topoisomerasa II , Células Tumorales Cultivadas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Anticancer Res ; 20(5B): 3591-601, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11131667

RESUMEN

The relationship between growth and the antioxidant enzyme defence system in human MCF-7 (breast) cancer cells treated with bovine milk fat enriched with conjugated linoleic acid (CLA) was studied. Milk enriched in CLA was obtained from cows on pasture supplemented with full fat rapeseeds and full fat soyabeans (1). Cell number decreased up to 90% (p < 0.05) and lipid peroxidation increased 15-fold (p < 0.05) following incubation of MCF-7 cells for 8 days with increasing levels of milk fat yielding CLA concentrations between 16.9 and 22.6 ppm. Growth suppression and prooxidant effects of milk fat CLA were independent of the variable composition of the milk fat samples, suggesting that CLA was the active ingredient in milk fat responsible for the cytotoxic effect. Mixtures containing isomers of CLA (c9, t11-, t10, c12-, c11, t13- and minor amounts of other isomers) and linoleic acid (LA) at similar concentrations to the milk fat samples were as effective at inhibiting growth and stimulating peroxidation of MCF-7 cells as the milk fatty acids. Incubation of the cells with the c9, t11 CLA isomer (20 ppm) or the mixture of CLA isomers (20 ppm) for 8 days resulted in a 60% decrease (p < 0.05) in viability compared with untreated controls and was significantly (p < 0.05) more effective than incubation with the t10, c12 CLA isomer (20 ppm), which caused only a 15% decrease in cell numbers under similar conditions. A 25% increase (p < 0.05) in cell proliferation occurred when LA (20 ppm) alone was incubated with MCF-7 cells for 8 days. 14C-CLA was preferentially incorporated into the phospholipid fraction of the MCF-7 cell lipids in a dose-dependent manner and CLA accumulated in cell membranes more efficiently when the cells were incubated in the presence of milk fat than the c9, t11 synthetic CLA isomer. Superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx) activities were induced in MCF-7 cells exposed to milk fat (containing 16.9-22.6 ppm CLA) over 8 days. The data indicate that milk fat triglyceride-bound CLA, consisting primarily of the c9, t11 isomer, was cytotoxic towards MCF-7 cells.


Asunto(s)
Neoplasias de la Mama/patología , Ácidos Grasos/farmacología , Inhibidores de Crecimiento/farmacología , Ácidos Linoleicos/farmacología , Leche/química , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Radioisótopos de Carbono , Catalasa/metabolismo , Bovinos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dieta , Relación Dosis-Respuesta a Droga , Ácidos Grasos/farmacocinética , Femenino , Glutatión Peroxidasa/metabolismo , Inhibidores de Crecimiento/farmacocinética , Humanos , Ácidos Linoleicos/farmacocinética , Peroxidación de Lípido/efectos de los fármacos , Glycine max , Superóxido Dismutasa/metabolismo , Células Tumorales Cultivadas
17.
Exp Eye Res ; 71(5): 483-7, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11040083

RESUMEN

The purpose of this study was to investigate the use of dietary isoflavonoids and flavonoids for the treatment of ocular neovascularization. Corneal blood vessels were induced by intrastromal implantation of pellets containing bFGF. Isoflavonoids and flavonoids (Genistein, Fisetin and Luteolin) were dissolved in a microemulsion to increase bioavailability and applied topically in concentrations between 0.5 and 1 ng ml(-1). Corneal neovascularization was quantified under the microscope. In comparison to control eyes, all three substances significantly inhibited corneal neovascularization (P < or = 0.05). Fisetin had the strongest effect followed by Genistein and Luteolin. No significant topical side effects were observed. We concluded that the isoflavonoid Genistein and two structurally related flavonoids are potent inhibitors of corneal angiogenesis in vivo. The wide distribution of the flavonoids in the plant kingdom together with the presented results suggests that flavonoids may contribute to the preventive effect of a plant-based diet on neovascular disease of the eye.


Asunto(s)
Neovascularización de la Córnea/terapia , Flavonoides/uso terapéutico , Genisteína/uso terapéutico , Inhibidores de Crecimiento/uso terapéutico , Administración Tópica , Animales , Disponibilidad Biológica , Emulsiones , Flavonoides/farmacocinética , Flavonoles , Genisteína/farmacocinética , Inhibidores de Crecimiento/farmacocinética , Luteolina , Conejos , Resultado del Tratamiento
18.
J Nutr ; 130(7): 1766-71, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10867048

RESUMEN

An experiment clarifying the influence of food deprivation on the isoflavone conjugation pattern in rats was conducted. Food-deprived and fed rats were administered daidzein and genistein at 7.9 mcmol/kg body, and changes in their plasma metabolites (i.e., free compounds, sulfates, glucuronides, sulfates/glucuronides) were measured quantitatively as a function of time. In the food-deprived group, total plasma daidzein and genistein reached maximum concentrations of 20.9 +/- 4.4 and 11.4 +/- 3.1 mcmol/L, respectively, 10 min after administration, whereas in the fed group, the maxima were 2.4 +/- 0.8 mcmol/L for daidzein after 2 h and 1. 8 +/- 0.2 mcmol/L for genistein after 4 h. In both groups, there were significantly more daidzein sulfates than genistein sulfates. Moreover, depriving rats of food before daidzein and genistein administration significantly increased plasma isoflavone sulfates with simultaneous significant decreases in plasma isoflavone glucuronides compared with fed rats. Additionally, nonconjugated daidzein and genistein appeared in plasma of food-deprived rats for 1 h after administration. Plasma concentrations of conjugates having both sulfate and glucuronide moieties were not significantly different between the groups.


Asunto(s)
Privación de Alimentos , Genisteína/farmacocinética , Glycine max , Inhibidores de Crecimiento/farmacocinética , Isoflavonas/farmacocinética , Animales , Cromatografía Líquida de Alta Presión , Glucurónidos/metabolismo , Masculino , Ratas , Ratas Wistar , Sulfatos/metabolismo
19.
J Neuroimmunol ; 106(1-2): 172-80, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10814795

RESUMEN

Leukemia inhibitory factor (LIF) is a neurotrophic cytokine now under clinical investigation for its effects on the CNS. We studied its passage across the blood-brain barrier (BBB) from blood to brain and spinal cord. Although a large amount of LIF was reversibly associated with the cerebral vasculature, intact LIF did reach brain parenchyma. Multiple-time regression analysis showed ready access of LIF to the CNS at a rate much faster than that of the vascular marker albumin. Excess LIF inhibited the entry of 125I-LIF after administration i.v. or by in-situ perfusion in blood-free buffer. Efflux of LIF from brain to blood was slower than reabsorption by CSF bulk flow, indicating that LIF tended to be retained in the brain. Although ciliary neurotrophic factor (CNTF) and LIF bind to the same receptor complex, CNTF did not cross-inhibit the entry of LIF into the CNS. A monoclonal antibody to LIF, however, abolished the entry of LIF. Our results show that peripherally administered LIF readily enters the brain and spinal cord by a saturable transport system across the BBB that may have biological implications.


Asunto(s)
Sistema Nervioso Central/metabolismo , Inhibidores de Crecimiento/farmacocinética , Interleucina-6 , Linfocinas/farmacocinética , Animales , Anticuerpos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Factor Neurotrófico Ciliar/farmacología , Relación Dosis-Respuesta a Droga , Estabilidad de Medicamentos , Inhibidores de Crecimiento/antagonistas & inhibidores , Inhibidores de Crecimiento/sangre , Inhibidores de Crecimiento/inmunología , Humanos , Factor Inhibidor de Leucemia , Linfocinas/antagonistas & inhibidores , Linfocinas/sangre , Linfocinas/inmunología , Ratones , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacocinética , Distribución Tisular
20.
Biol Trace Elem Res ; 78(1-3): 241-54, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11314982

RESUMEN

The metabolism of arsenic, its affinity to metallothionein (MT), its influence on selenium levels, and its biotransformation to different metabolites in the liver tissue of laying hens exposed to arsenic trioxide (As2O3) was investigated. The experiment was performed with two groups of hens fed for 19 d with either a standard diet or with the same diet enriched in arsenic (30 microg/g). The major findings were as follows: 1. After 19 d exposure, about 65% of the total liver As was found in the water-soluble phase (100,000g centrifuged supernatant). In liver supernatant, As binding was found mostly in the range of very low-molecular-weight proteins (Mr < 10,000). Although after exposure the amount of MT-like proteins increased, the As bound to it was only in trace amounts. The protein was identified by convential procedures as Zn,Cu-thionein with traces of selenium and arsenic. 2. Arsenic exposure resulted in almost unchanged Se levels regarding its tissue concentrations and distribution between supernatant and pellet, where about 10% of total Se was found in the supernatant. On the contrary, As exposure did affect Cd levels. Tissue Cd concentration was slightly diminished, but the percentage of tissue Cd found in the water-soluble phase was increased from 20% to 40%. 3. In methanol extracts of tissue and supernatant of the As-exposed group, only two arsenic compounds were detected, As(III) and dimethylarsinic acid (DMA), the latter prevailing.


Asunto(s)
Arsenicales/farmacología , Pollos , Inhibidores de Crecimiento/farmacología , Hígado/metabolismo , Metalotioneína/metabolismo , Óxidos/farmacología , Animales , Arsénico/metabolismo , Trióxido de Arsénico , Arsenicales/farmacocinética , Bioquímica/métodos , Cadmio/metabolismo , Cobre/metabolismo , Femenino , Inhibidores de Crecimiento/farmacocinética , Inactivación Metabólica , Hígado/efectos de los fármacos , Metalotioneína/efectos de los fármacos , Metanol/química , Óxidos/farmacocinética , Selenio/metabolismo , Extractos de Tejidos/química , Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...