Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nutrients ; 12(6)2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32516922

RESUMEN

We investigated the effect of evodiamine-containing microalga Dunaliella tertiolecta (DT) on the prevention of diet-induced obesity in a thermoneutral C57BL/6J male (30 °C). It attenuates the activity of brown adipose tissue (BAT), which accelerates diet-induced obesity. Nine-week-old mice were fed a high-fat diet supplemented with 10 g (Low group) or 25 g (High group) DT powder per kg food for 12 weeks. Compared to control mice without DT supplementation, body weight gain was significantly reduced in the High group with no difference in food intake. Tissue analyses indicated maintenance of multilocular morphology in BAT and reduced fat deposition in liver in DT-supplemented mice. Molecular analysis showed a significant decrease in mammalian target of rapamycin-ribosomal S6 protein kinase signaling pathway in white adipose tissue and upregulation in mRNA expression of brown fat-associated genes including fibroblast growth factor-21 (Fgf21) and uncoupling protein 1 (Ucp1) in BAT in the High group compared to the control. In the experiments using C3H10T1/2 adipocytes, DT extract upregulated mRNA expression of brown fat-associated genes in dose-dependent and time-dependent manners, accompanied by a significant increase in secreted FGF21 levels. Our data show the ability of DT as a nutraceutical to prevent brown fat attenuation and diet-induced obesity in vivo.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Microalgas/química , Obesidad/metabolismo , Obesidad/prevención & control , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Termogénesis/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Obesidad/etiología , Quinazolinas/aislamiento & purificación , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína Desacopladora 1/metabolismo
2.
Mol Cancer Ther ; 19(4): 976-987, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32241871

RESUMEN

HER2 is a transmembrane tyrosine kinase receptor that mediates cell growth, differentiation, and survival. HER2 is overexpressed in approximately 20% of breast cancers and in subsets of gastric, colorectal, and esophageal cancers. Both antibody and small-molecule drugs that target HER2 and block its tyrosine kinase activity are effective in treating HER2-driven cancers. In this article, we describe the preclinical properties of tucatinib, an orally available, reversible HER2-targeted small-molecule tyrosine kinase inhibitor. In both biochemical and cell signaling experiments, tucatinib inhibits HER2 kinase activity with single-digit nanomolar potency and provides exceptional selectivity for HER2 compared with the related receptor tyrosine kinase EGFR, with a >1,000-fold enhancement in potency for HER2 in cell signaling assays. Tucatinib potently inhibits signal transduction downstream of HER2 and HER3 through the MAPK and PI3K/AKT pathways and is selectively cytotoxic in HER2-amplified breast cancer cell lines in vitro. In vivo, tucatinib is active in multiple HER2+ tumor models as a single agent and shows enhanced antitumor activity in combination with trastuzumab or docetaxel, resulting in improved rates of partial and complete tumor regression. These preclinical data, taken together with the phase-I tucatinib clinical trial results demonstrating preliminary safety and activity, establish the unique pharmacologic properties of tucatinib and underscore the rationale for investigating its utility in HER2+ cancers. GRAPHICAL ABSTRACT: http://mct.aacrjournals.org/content/molcanther/19/4/976/F1.large.jpg.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/metabolismo , Animales , Apoptosis , Proliferación Celular , Docetaxel/administración & dosificación , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Oxazoles/administración & dosificación , Piridinas/administración & dosificación , Quinazolinas/administración & dosificación , Trastuzumab/administración & dosificación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Pharm ; 581: 119278, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32229284

RESUMEN

Berberine chloride (BBR) and evodiamine (EVO) are two main active ingredients of "ZuoJinWan", a classical Chinese herbal medicine, and these compounds are known to have a synergistic inhibitory effect on various cancer cell lines. Several recent studies have reported anti-melanoma effects for both BBR and EVO. However, topical delivery of the two compounds has been challenging, due to their poor aqueous solubility and their low skin penetration. In the current study, we have combined BBR and EVO into an ethosomes delivery system with the future aim to design a novel topical anti-melanoma formulation. The ethosomes formulations were characterized using particle size, entrapment efficiency and an in vitro skin drug deposition study. The ethosome formulation displaying maximum drug deposition in the epidermis was selected for further study. This formulation contained ethosomes with mean size of 171 nm and 90% or above entrapment efficiency for both BBR and EVO. Cell viability tests proved the optimized ethosomes increased the inhibitory effect on B16 melanoma cells. These results corroborate that ethosomes containing a combination of BBR and EVO are a promising delivery system for potential use in melanoma therapy.


Asunto(s)
Berberina/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Desarrollo de Medicamentos/métodos , Melanoma Experimental/metabolismo , Quinazolinas/metabolismo , Absorción Cutánea/efectos de los fármacos , Animales , Berberina/administración & dosificación , Berberina/metabolismo , Femenino , Humanos , Liposomas , Melanoma Experimental/tratamiento farmacológico , Ratones , Persona de Mediana Edad , Extractos Vegetales/administración & dosificación , Extractos Vegetales/metabolismo , Quinazolinas/administración & dosificación
4.
Clin Drug Investig ; 40(3): 237-247, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31965548

RESUMEN

BACKGROUND AND OBJECTIVES: Carotegrast methyl, a novel prodrug, oral antagonist of α4-integrin, is in development for the treatment of active ulcerative colitis. This randomised, placebo-controlled, double-blind, crossover study evaluated the effect of food on the pharmacokinetics and pharmacodynamics as well as the safety profile after a single dose of carotegrast methyl in healthy male subjects. METHODS: Subjects were randomised to receive a single dose of carotegrast methyl (240, 480 or 960 mg) or placebo in a 6:2 ratio and received the study drug under both fed and fasted conditions separated by an 8-day washout. The pharmacokinetic profiles of carotegrast methyl and its active metabolite, carotegrast, were assessed. The pharmacodynamic profile was evaluated according to a change in the peripheral lymphocyte count. Safety was monitored throughout. RESULTS: Based on the area under the time curve from zero to the time of the last quantifiable concentration (AUClast), food reduced systemic exposure to both carotegrast methyl and carotegrast by 21-57% and 5-29%, respectively. The fed-to-fasted ratio of least square means for the increase in the lymphocyte count was almost at unity in each dose, indicating no food effect on pharmacodynamics. The time ≥ 90% of maximum effect was prolonged dose dependently, suggesting that a 960 mg-dose can provide a long-lasting effect. Reported adverse events were all mild. CONCLUSIONS: Despite the reduced systemic exposure to both carotegrast methyl and carotegrast, food had no effect on the increase in lymphocyte count. A single administration of carotegrast methyl up to 960 mg was found to be safe.


Asunto(s)
Interacciones Alimento-Droga , Integrina alfa4/antagonistas & inhibidores , Fenilalanina/análogos & derivados , Quinazolinas/administración & dosificación , Administración Oral , Adulto , Área Bajo la Curva , Estudios Cruzados , Método Doble Ciego , Ayuno , Humanos , Masculino , Fenilalanina/administración & dosificación , Quinazolinonas , Adulto Joven
5.
Oncol Rep ; 43(2): 415-426, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31894286

RESUMEN

Colon cancer is one of the most common malignancies. Although there has been great development in treatment regimens over the last few decades, its prognosis remains poor. There is still a clinical need to find new drugs for colon cancer. Evodiamine (Evo) is a quinolone alkaloid extracted from the traditional herbal medicine plant Evodia rutaecarpa. In the present study, CCK­8, flow cytometry, reverse transcription quantitative polymerase chain reaction, western blot analysis and a xenograft tumor model were used to evaluate the anti­cancer activity of Evo in human colon cancer cells and determine the possible mechanism underlying this process. It was revealed that Evo exhibited prominent anti­proliferation and apoptosis­inducing effects in HCT116 cells. Bone morphogenetic protein 9 (BMP9) was notably upregulated by Evo in HCT116 cells. Exogenous BMP9 potentiated the anti­cancer activity of Evo, and BMP9 silencing reduced this effect. In addition, HIF­1α was also upregulated by Evo. The anticancer activity of Evo was enhanced by HIF­1α, but was reduced by HIF­1α silencing. BMP9 potentiated the effect of Evo on the upregulation of HIF­1α, and enhanced the antitumor effect of Evo in colon cancer, which was clearly reduced by HIF­1α silencing. In HCT116 cells, Evo increased the phosphorylation of p53, which was enhanced by BMP9 but reduced by BMP9 silencing. Furthermore, the effect of Evo on p53 was potentiated by HIF­1α and reduced by HIF­1α silencing. The present findings therefore strongly indicated that the anticancer activity of Evo may be partly mediated by BMP9 upregulation, which can activate p53 through upregulation of HIF­1α, at least in human colon cancer.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Factor 2 de Diferenciación de Crecimiento/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quinazolinas/administración & dosificación , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Ratones , Fosforilación , Quinazolinas/farmacología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Pharmacol Res Perspect ; 7(6): e00523, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31624633

RESUMEN

Many compounds that appear promising in preclinical species, fail in human clinical trials due to safety concerns. The FDA has strongly encouraged the application of modeling in drug development to improve product safety. This study illustrates how DILIsym, a computational representation of liver injury, was able to reproduce species differences in liver toxicity due to PF-04895162 (ICA-105665). PF-04895162, a drug in development for the treatment of epilepsy, was terminated after transaminase elevations were observed in healthy volunteers (NCT01691274). Liver safety concerns had not been raised in preclinical safety studies. DILIsym, which integrates in vitro data on mechanisms of hepatotoxicity with predicted in vivo liver exposure, reproduced clinical hepatotoxicity and the absence of hepatotoxicity observed in the rat. Simulated differences were multifactorial. Simulated liver exposure was greater in humans than rats. The simulated human hepatotoxicity was demonstrated to be due to the interaction between mitochondrial toxicity and bile acid transporter inhibition; elimination of either mechanism from the simulations abrogated injury. The bile acid contribution occurred despite the fact that the IC50 for bile salt export pump (BSEP) inhibition by PF-04895162 was higher (311 µmol/L) than that has been generally thought to contribute to hepatotoxicity. Modeling even higher PF-04895162 liver exposures than were measured in the rat safety studies aggravated mitochondrial toxicity but did not result in rat hepatotoxicity due to insufficient accumulation of cytotoxic bile acid species. This investigative study highlights the potential for combined in vitro and computational screening methods to identify latent hepatotoxic risks and paves the way for similar and prospective studies.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/antagonistas & inhibidores , Anticonvulsivantes/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Biológicos , Quinazolinas/toxicidad , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Administración Oral , Adolescente , Adulto , Animales , Anticonvulsivantes/administración & dosificación , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Simulación por Computador , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/normas , Epilepsia/tratamiento farmacológico , Células HEK293 , Voluntarios Sanos , Hepatocitos , Humanos , Concentración 50 Inhibidora , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Quinazolinas/administración & dosificación , Ratas , Especificidad de la Especie , Ácido Taurocólico/metabolismo , Adulto Joven
7.
J Ethnopharmacol ; 236: 288-301, 2019 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-30872168

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Aerial parts of Peganum harmala Linn are a Uighur traditional medicinal herb in China used to treat amnesia, bronchial asthma, and cough. Deoxyvasicine (DVAS), a potent cholinesterase inhibitor exhibiting anti-senile dementia activity, is one of the chief active ingredients in aerial parts of P. harmala and plays a key role in mediating the pharmacological effects of P. harmala. However, the metabolic profiling and in vivo pharmacokinetic characteristics of DVAS still remain unknown. AIM OF THE STUDY: The aim of this present study was to investigate the metabolism and pharmacokinetic properties of DVAS in rats by using ultra-performance liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry (UPLC-ESI-QTOF-MS) and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-ESI-MS/MS) method. MATERIALS AND METHODS: The metabolic profiling of DVAS was evaluated in vitro and in vivo by rat liver microsomes (RLMs) incubation and by rat bio-specimens, such as urine, feces, plasma, and bile, after the oral administration of 45 mg/kg DVAS. An efficient and sensitive UPLC-ESI-MS/MS method was developed and validated to simultaneously determine DVAS and its major four metabolites, namely, vasicine, deoxyvasicinone, vasicinone, and 1,2,3,9-tetrahydropyrrolo[2,1-b]quinazolin-3-ß-D-glucuronide in rat plasma. For pharmacokinetic studies, 32 Sprague-Dawley rats were randomly divided into four groups, namely, intravenous dosage group (2 mg/kg DVAS) and three oral dosage groups (5, 15, and 45 mg/kg DVAS). In addition, the activity of the components in plasma after intravenous administration of DVAS was evaluated by in vitro anti-butyrylcholinesterase (BChE) assays. RESULTS: A total of 23 metabolites were found in RLMs, plasma, urine, feces, and bile by UPLC-ESI-QTOF-MS. The metabolic pathway of DVAS in vivo and in vitro mainly involved hydroxylation, dehydrogenation, acetylation, methylation, glucuronidation, and O-sulphate conjugation, and the C-3 and C-9 sites were the main metabolic soft spots. All 23 metabolites were detected in the urine sample, and 13, 8, 22, and 6 metabolites were identified from rat feces, plasma, bile, and RLMs, respectively. The standard curves of DVAS and four metabolites in rat plasma showed good linearity in the concentration range of 0.82-524.00 ng/mL with acceptable selectivity, precision, accuracy, recovery, and stability. DVAS exhibited linear dose-proportional pharmacokinetics at doses of 5, 15, and 45 mg/kg after oral administration, and the average oral absolute bioavailability of DVAS was 47.46%. The in vitro anti-BChE assays implied that the inhibitive activities were mainly due to the different concentrations of prototype DVAS. CONCLUSIONS: DVAS can be rapidly absorbed and excreted by blood, and it is also extensively metabolized in vivo, and the anti-BChE activity in blood is mainly attributed to DVAS. These findings can lay a foundation for new drug development for DVAS.


Asunto(s)
Alcaloides/farmacocinética , Inhibidores de la Colinesterasa/farmacocinética , Peganum/química , Quinazolinas/farmacocinética , Administración Intravenosa , Administración Oral , Alcaloides/administración & dosificación , Alcaloides/sangre , Alcaloides/metabolismo , Animales , Butirilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/administración & dosificación , Inhibidores de la Colinesterasa/sangre , Inhibidores de la Colinesterasa/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Pruebas de Enzimas , Femenino , Masculino , Medicina Tradicional , Microsomas Hepáticos , Componentes Aéreos de las Plantas/química , Quinazolinas/administración & dosificación , Quinazolinas/sangre , Quinazolinas/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos
8.
Br J Haematol ; 185(4): 691-700, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30919941

RESUMEN

Anagrelide is an established treatment option for essential thrombocythaemia (ET). A prolonged release formulation was developed with the aim of reducing dosing frequency and improving tolerability, without diminishing efficacy. This multicentre, randomized, double blind, active-controlled, non-inferiority trial investigated the efficacy, safety and tolerability of anagrelide prolonged release (A-PR) over a reference product in high-risk ET patients, either anagrelide-naïve or -experienced. In a 6 to 12-week titration period the individual dose for the consecutive 4-week maintenance period was identified. The primary endpoint was the mean platelet count during the maintenance period (3 consecutive measurements, day 0, 14, 28). Of 112 included patients 106 were randomized. The mean screening platelet counts were 822 × 109 /l (95% confidence interval (CI) 707-936 × 109 /l) and 797 × 109 /l (95% CI 708-883 × 109 /l) for A-PR and the reference product, respectively. Both treatments effectively reduced platelet counts, to mean 281 × 109 /l for A-PR (95% CI 254-311) and 305 × 109 /l (95% CI 276-337) for the reference product (P < 0·0001, for non-inferiority). Safety and tolerability were comparable between both drugs. The novel prolonged-release formulation was equally effective and well tolerated compared to the reference product. A-PR provides a more convenient dosing schedule and will offer an alternative to licensed immediate-release anagrelide formulations.


Asunto(s)
Inhibidores de Agregación Plaquetaria/administración & dosificación , Quinazolinas/administración & dosificación , Trombocitemia Esencial/tratamiento farmacológico , Preparaciones de Acción Retardada , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Composición de Medicamentos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/farmacocinética , Recuento de Plaquetas , Calidad de Vida , Quinazolinas/efectos adversos , Quinazolinas/farmacocinética , Resultado del Tratamiento
9.
Mol Pharm ; 16(1): 422-436, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30525641

RESUMEN

Evodiamine (EVO) and Berberine (BBR), from Euodiae Fructus and Coptidis rhizoma, have been used as an herbal medicine pair in traditional Chinese medicine to exert synergistic antitumor effects against various types of tumor cells. However, their clinical use is limited by their poor solubility and adverse toxic side effects. Mesoporous silica nanoparticles (MSNs) possess excellent properties such as a readily functionalized surface, prominent biocompatibility, and huge specific surface area for loading with hydrophobic and hydrophilic drug. On this basis, a novel temperature- and pH-responsive dual drug delivery platform has been developed, in which lipid-coated MSN@p(NIPAM- co-MA) codelivers EVO and BBR. The results indicate that the nanocarrier improves the efficacy and biocompatibility of the drug pair and maintain desirable drug profiles at the low pH and higher temperature of the tumor microenvironment. The dual drug-loaded MSNs showed excellent synergistic therapy effects in vitro (cytotoxicity, cell migration and invasion, angiogenesis) and in vivo (growth of tumor grafts in mice). Meanwhile, the dual drug-loaded nanoparticles showed lower systemic toxicity than either drug alone, the free drug combination, or Taxol. These results suggest that the temperature- and pH-sensitive lipid-coated MSNs are a promising novel carrier for both hydrophobic and hydrophilic drugs.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Dióxido de Silicio/química , Animales , Berberina/administración & dosificación , Berberina/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Portadores de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Quinazolinas/administración & dosificación , Quinazolinas/uso terapéutico , Temperatura , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Anticancer Drugs ; 29(10): 1021-1025, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30134285

RESUMEN

The liver is the most common site of colorectal cancer metastases. The present study aimed to evaluate the efficacy and safety of transarterial chemoembolization (TACE) with raltitrexed and oxaliplatin for colorectal liver metastases in a prospective, multicenter, single-arm trial conducted in 12 hospitals from different areas in China. A total of 90 patients with colorectal liver metastases were enrolled and treated by TACE with raltitrexed 4 mg and oxaliplatin 100 mg, followed by embolotherapy with 50 mg oxaliplatin and 5-20 ml lipiodol, administered every 28 days for four cycles. Patients were followed up every 3 months after the treatment and up to 12 months. The primary endpoint was time to progression. For the full analysis set (FAS), the median time to progression and overall survival were 9.1 and 17.8 months, respectively. The disease control rate in FAS was 71 (78.9%). Grade 3 or 4 adverse events were reported for 24 (26.7%) out of all 90 patients. Grade 3 thrombocytopenia, transglutaminase abnormality, and decreased neutrophil were observed in eight (8.9%), six (6.7%), and five (5.6%) patients, respectively. No unexpected adverse events or toxic deaths were observed. TACE with raltitrexed plus oxaliplatin is feasible, clinically beneficial, and well tolerated with low-grade toxicity for colorectal cancer patients with liver metastases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Quimioembolización Terapéutica/métodos , Neoplasias Colorrectales/terapia , Neoplasias Hepáticas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Quimioembolización Terapéutica/efectos adversos , China , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Aceite Etiodizado/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Oxaliplatino/administración & dosificación , Estudios Prospectivos , Quinazolinas/administración & dosificación , Sobrevida , Tiofenos/administración & dosificación
11.
Drug Deliv ; 25(1): 1426-1437, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29890855

RESUMEN

Most antitumor ingredients found in nature have poor solubility. These ingredients are expected to have much better absorption and higher bioavailability than synthetic antitumor agents. Woody oil emulsive nanosystems carrying poorly soluble natural alkaloids were fabricated (evodiamine (EA) carried by fructus bruceae oil-based emulsive nanosystems, or EFEN). Fructus bruceae oil has two excipient-like properties (oil phase and stabilizer) that contribute to the formulation and one drug-like property (antitumor effects) that synergizes with the antitumor effect of EA. The properties of EFEN were compared with free EA, a blank nanoemulsion, an EA-loaded emulsive nanosystem, and a fructus bruceae oil-loaded emulsive nanosystem. For the first time, this suggests that increases in the sensitivity of lung cancer cells to poorly soluble natural alkaloids can be achieved by delivering drugs using woody oil-based emulsive nanosystems. In this study, woody oil-based emulsive nanosystems efficiently deliver poorly soluble natural alkaloids.


Asunto(s)
Alcaloides/química , Emulsiones/administración & dosificación , Emulsiones/química , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/química , Aceites de Plantas/química , Lana/química , Células A549 , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Disponibilidad Biológica , Brucea javanica , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Medicamentos Herbarios Chinos/química , Excipientes/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Quinazolinas/administración & dosificación , Quinazolinas/química , Ratas , Ratas Sprague-Dawley , Solubilidad/efectos de los fármacos
12.
Chin J Integr Med ; 24(6): 460-466, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29335861

RESUMEN

OBJECTIVE: To evaluate the potential pharmacokinetic interactions of the anticancer agent gefitinib (Iressa®) and the oriental medications Guipi Decoction (, GPD, Guibi-tang in Korean) and Bawu Decoction (, BWD, Palmul-tang in Korean). METHODS: Methylcellulose (MC, control), GPD (1,200 mg/kg), or BWD (6,000 mg/kg) was orally administered to rats either as a single dose or multiple doses prior to gefitinib administration. To examine the effects of a single dose of the herbal medicines, gefitinib (10 mg/kg) was orally administered after 5 min or 1 h of MC or the herbal medicine pretreatments. To examine the effects of the multiple doses of the herbal medicines, gefitinib (10 mg/kg) was orally administered following 7 consecutive days of the administration of MC or each herbal medicine. The plasma concentrations of gefitinib were determined with liquid chromatography-tandem mass spectrometry assay. The plasma concentration-time profiles of gefitinib were analyzed with a noncompartmental analysis. RESULTS: Gefitinib was rapidly absorbed and showed a monoexponential decline with an elimination half-life of 3.7-4.1 h. The pharmacokinetics of gefitinib was not affected by GPD pretreatment. However, a significantly lower maximum plasma concentration (Cmax, P<0.05) and area under the curve (P<0.05), and a delayed time to reach Cmax (Tmax, P<0.01) were observed in both single- and multipledose BWD-pretreated rats compared with the control rats. CONCLUSIONS: BWD and not GPD might delay and interfere with gefitinib absorption. Further evaluations of the clinical significance of these findings are needed.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética , Animales , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Gefitinib , Masculino , Quinazolinas/sangre , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Factores de Tiempo
13.
J Nat Med ; 72(1): 145-154, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28936800

RESUMEN

Liver fibrosis is considered to be a result of chronic liver pathological changes, and hepatic stellate cells (HSCs) play an important role during this process. Evodiamine, an indole alkaloid derived from Evodia rutaecarpa, exhibits pharmacological activities. This study focused on the effects of evodiamine on carbon tetrachloride (CCl4)-induced liver fibrosis in rats and HSCs in vitro via the TGF-ß1/Smad signaling pathway. A liver fibrosis rat model was established by the intraperitoneal injection of CCl4 (3 ml/kg, 30% in olive oil). Evodiamine (15 and 25 mg/kg) was administered orally for 8 weeks. HSCs were treated with different evodiamine concentrations. The results indicated that evodiamine could improve the histopathological abnormalities in liver tissues and decrease the level of aspartate aminotransferase (AST), alanine aminotransferase (ALT), hydroxyproline, and total bilirubin (TBIL). Concentrations of IL-6, tumor necrosis factor-α (TNF-α), collagen-I (COL-I), and collagen-III (COL-III) were reduced by evodiamine. Western blotting and real-time PCR showed that protein expression of transforming growth factor-ß (TGF-ß1), p-Smad 2/3 (phosphorylation of Smad 2/3), and smooth muscle alpha-actin (α-SMA) as well as mRNA expression of TGF-ß1 and α-SMA in liver tissues were downregulated by evodiamine. The cell proliferation, production of hydroxyproline, and the protein expression of TGF-ß1, p-Smad 2/3, and α-SMA in HSCs were dose-dependently reduced by evodiamine. Collectively, evodiamine had an antifibrosis effect in CCl4-induced liver fibrosis, and reduced HSCs proliferation and collagen metabolism in vitro. The major mechanism was downregulation of relative expression of TGF-ß1, p-Smad 2/3, and α-SMA.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , Quinazolinas/uso terapéutico , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Modelos Animales de Enfermedad , Cirrosis Hepática/patología , Masculino , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Ratas , Ratas Wistar
14.
Antioxid Redox Signal ; 28(5): 339-357, 2018 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28665143

RESUMEN

AIMS: Systemic diseases often have common characteristics. The aim of this study was to investigate the feasibility of targeting common pathological metabolism to inhibit the progression of malignant and proliferative diseases. RESULTS: Gefitinib-resistant (G-R) nonsmall-cell lung cancer (NSCLC) and rheumatoid arthritis (RA) were studied as conditions representative of malignant and proliferative diseases, respectively. Strong lipogenic activity and high expression of sterol regulatory element-binding protein 1 (SREBP1) were found in both G-R NSCLC cells and synovial fibroblasts from RA patients (RASFs). Berberine (BBR), an effective suppressor of SREBP1 and lipogenesis regulated through reactive oxygen species (ROS)/AMPK pathway, selectively inhibited the growth of G-R NSCLC cells and RASFs but not that of normal cells. It effectively caused mitochondrial dysfunction, activated ROS/AMPK pathway, and finally suppressed cellular lipogenesis and cell proliferation. Addition of ROS blocker, AMPK inhibitor, and palmitic acid significantly reduced the effect of BBR. In an in vivo study, treatment of BBR led to significant inhibition of mouse tumor xenograft growth and remarkably slowed down the development of adjuvant-induced arthritis in rats. Innovation and Conclusion: Targeting ROS/AMPK/lipogenesis signaling pathway selectively inhibited the growth of G-R NSCLC cells and the progress of RASFs in vitro and in vivo, which provides a new avenue for treating malignancies and proliferative diseases. Antioxid. Redox Signal. 28, 339-357.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Artritis Reumatoide/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Lipogénesis/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Berberina/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Gefitinib , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Oxidación-Reducción , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Líquido Sinovial/efectos de los fármacos , Líquido Sinovial/metabolismo
15.
Expert Opin Pharmacother ; 18(15): 1637-1648, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28933580

RESUMEN

INTRODUCTION: Treatment options for relapsed ovarian cancer have increased over the decade with the addition of targeted agents, such as PARP inhibitors and antiangiogenic agents. Bevacizumab, a monoclonal antibody binding vascular endothelial growth factor (VEGF), was the first anti-angiogenic agent to be incorporated in the ovarian cancer treatment landscape. Other molecules utilising different mechanisms of action to target angiogenesis have been developed, including cediranib, an oral potent inhibitor of VEGF Tyrosine Kinase Inhibitor that has demonstrated activity in both phase II and phase III studies. Areas covered: Herein we will review cediranib as well as the evidence for its use in ovarian cancer, both as monotherapy and in combination with chemotherapy, PARP inhibitors and immunotherapy. A literature search was made in PubMed and on ClinicalTrials.gov for clinical trials with cediranib. Expert opinion: The addition of cediranib for the treatment of ovarian cancer is promising, and has demonstrated a significant improvement in progression free survival in a phase III trial in combination with chemotherapy and maintenance treatment. Cediranib is currently being explored in ovarian cancer and other gynaecological malignancies aiming to improve patient care; further research will help define its role in standard clinical practice for patients with ovarian cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Quinazolinas/uso terapéutico , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ensayos Clínicos Fase III como Asunto , Terapia Combinada , Supervivencia sin Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Neoplasias Ováricas/irrigación sanguínea , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos
16.
Molecules ; 22(7)2017 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-28708106

RESUMEN

Evodiamine is one of the main components isolated from Evodia rutaecarpa, and it has been reported to exert inhibitory effects on cancers by anti-proliferative and apoptosis-inducing activities. Although the anti-cancer activity of evodiamine has been identified, the precise mechanisms of this action remain obscure. While previous studies indicated that evodiamine exerts anti-tumor effects through inhibiting ß-catenin activity, and WW domain-containing oxidoreductase (WWOX) regulates ß-catenin accumulation in cytoplasm, the effects of evodiamine on the expression of WWOX are still unknown. In this study, we provide evidence that evodiamine dose- and time-dependently inhibits both Mus musculus and Homo sapiens hepatocellular carcinoma (HCC) cells, as well as Hepa1-6 and HepG2 cell proliferation. We further tested the therapeutic effects of evodiamine in Hepa1-6 hepatoma-bearing mice, and we found that treatment of evodiamine by oral gavage significantly decreased the tumor size of the mice. Moreover, the expressions of WWOX were dose-dependently increased in HCC cell lines as well as in Hepa1-6 hepatoma-bearing mice after the treatment with evodiamine. Knockdown of WWOX in HepG2 and Hepa1-6 cells diminished the effects of evodiamine on the inhibitory effect of cancer cell growth, indicating that evodiamine induced anti-cancer activity through a WWOX-dependent pathway. As such, evodiamine activated WWOX to exert an anti-HCC activity, and might be a potential therapeutic or preventive candidate for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Evodia/química , Quinazolinas/química , Quinazolinas/farmacología , Oxidorreductasa que Contiene Dominios WW/efectos de los fármacos , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen/métodos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Extractos Vegetales/química , Quinazolinas/administración & dosificación , beta Catenina
17.
Cancer Chemother Pharmacol ; 80(3): 535-544, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28714036

RESUMEN

PURPOSE: To determine the pharmacokinetics of radiolabeled copanlisib (BAY 80-6946) in healthy male volunteers and to investigate the disposition and biotransformation of copanlisib. METHODS: A single dose of 12 mg copanlisib containing 2.76 MBq [14C]copanlisib was administered as a 1-h intravenous infusion to 6 volunteers with subsequent sampling up to 34 days. Blood, plasma, urine and feces were collected to monitor total radioactivity, parent compound and metabolites. RESULTS: Copanlisib treatment was well tolerated. Copanlisib was rapidly distributed throughout the body with a volume distribution of 1870 L and an elimination half-life of 52.1-h (range 40.4-67.5-h). Copanlisib was the predominant component in human plasma (84% of total radioactivity AUC) and the morpholinone metabolite M1 was the only circulating metabolite (about 5%). Excretion of drug-derived radioactivity based on all 6 subjects was 86% of the dose within a collection interval of 20-34 days with 64% excreted into feces as major route of elimination and 22% into urine. Unchanged copanlisib was the main component excreted into urine (15% of dose) and feces (30% of dose). Excreted metabolites (41% of dose) of copanlisib resulted from oxidative biotransformation. CONCLUSIONS: Copanlisib was eliminated predominantly in the feces compared to urine as well as by hepatic biotransformation, suggesting that the clearance of copanlisib would more likely be affected by hepatic impairment than by renal dysfunction. The dual mode of elimination via unchanged excretion of copanlisib and oxidative metabolism decreases the risk of clinically relevant PK-related drug-drug interactions.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Quinazolinas/uso terapéutico , Administración Intravenosa , Fosfatidilinositol 3-Quinasa Clase I/farmacocinética , Fosfatidilinositol 3-Quinasa Clase I/uso terapéutico , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética
18.
Biomed Pharmacother ; 92: 149-156, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28535418

RESUMEN

Peritoneal metastasis of colorectal cancer is one of the most incident and fateful diseases among relapse cases. It shows a certain resistance to systemic chemotherapy. The perfusion system in clinic is complex and hard to be used in fundamental researches. This study aims at evaluating the effect of an improved hyperthermic intraperitoneal chemotherapy with Raltitrexed used in tumor-bearing mice with peritoneal metastatic colorectal carcinoma. The results showed that no severe adverse effect was observed. All control animals developed extensive peritoneal and mesenteric metastatic nodes. Tumor sites in the treatment groups were reduced significantly. The administration dose of Raltitrexed influenced concentration in systemic blood and peritoneal tissues. Temperature promoted the intracellular absorption of Raltitrexed significantly. Our findings reveal that hyperthermic intraperitoneal chemotherapy is an efficient therapy in treating peritoneal metastatic carcinoma in nude mice. It can effectively reduce the extension of carcinoma cells from macro and micro examination. The combination of hyperthermia and Raltitrexed resulted in an improved therapeutic effect on animal models.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Carcinoma/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Hipertermia Inducida , Neoplasias Peritoneales/tratamiento farmacológico , Quinazolinas/administración & dosificación , Tiofenos/administración & dosificación , Absorción Fisiológica , Animales , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/farmacocinética , Antimetabolitos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma/patología , Carcinoma/secundario , Carcinoma/terapia , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Terapia Combinada/efectos adversos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Femenino , Semivida , Calor , Humanos , Hipertermia Inducida/efectos adversos , Infusiones Parenterales , Masculino , Ratones Desnudos , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/terapia , Quinazolinas/efectos adversos , Quinazolinas/farmacocinética , Quinazolinas/uso terapéutico , Distribución Aleatoria , Tiofenos/efectos adversos , Tiofenos/farmacocinética , Tiofenos/uso terapéutico , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Tumour Biol ; 39(4): 1010428317697555, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28443459

RESUMEN

Astragaloside IV, the active component of Astragalus membranaceus, exhibits diverse biological roles including the anti-tumor activity. In this study, we evaluated the chemosensitive role of astragaloside IV in non-small cell lung cancer cells. Cell Counting Kit-8 analysis was performed to determine cell viability. Real-time polymerase chain reaction and western blot were used to measure the messenger RNA and protein expression. Results showed that astragaloside IV treatment could suppress the proliferation of non-small cell lung cancer cells. In addition, combined treatment with astragaloside IV remarkably enhanced the chemosensitivity to gefitinib in three non-small cell lung cancer cell lines including NCI-H1299, HCC827, and A549. Furthermore, compared with gefitinib-treated cells, the messenger RNA expression of SIRT6 was obviously increased in non-small cell lung cancer cells treated with gefitinib combined with astragaloside IV. In addition, downregulation of SIRT6 was accomplished using small interference RNA technology. As a result, SIRT6 inhibition abolished the sensitization role of astragaloside IV in non-small cell lung cancer cells. Taken together, these data demonstrated that astragaloside IV sensitized tumor cells to gefitinib via regulation of SIRT6, suggesting that astragaloside IV may serve as potential therapeutic approach for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinazolinas/administración & dosificación , Saponinas/administración & dosificación , Sirtuinas/biosíntesis , Triterpenos/administración & dosificación , Células A549 , Apoptosis/efectos de los fármacos , Astragalus propinquus/química , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sirtuinas/genética
20.
Pharm Biol ; 55(1): 294-298, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27927077

RESUMEN

CONTEXT: Rutaecarpine is an active indoloquinazoline alkaloid ingredient originating from Evodia rutaecarpa (Wu-zhu-yu in Chinese), which possesses a variety of effects. However, its metabolism has not been investigated thoroughly yet. OBJECTIVE: This study develops a highly sensitive and effective method for detection and characterization of the metabolites of rutaecarpine in Sprague-Dawley (SD) rats. MATERIALS AND METHODS: In this study, an efficient method was developed using ultra-high-performance liquid chromatography coupled with linear ion trap-Orbitrap mass spectrometer (UHPLC-LTQ-Orbitrap MS) to detect the metabolism profile of rutaecarpine in rat plasma. First, a blood sample (1 mL) was withdrawn 2 h after oral administration of rutaecarpine in SD rats (50 mg/kg). Second, the blood was centrifuged at 4000 rpm for 10 min and pretreated by solid-phase extraction method. Third, 2 µL of the plasma was injected into UHPLC-LTQ-Orbitrap MS for analysis. Finally, the metabolites of rutaecarpine were tentatively identified based on accurate mass measurements, fragmentation patterns and chromatographic retention times. RESULTS: A total of 16 metabolites (four new metabolites, viz., dihydroxylation and sulphate conjugation products of rutaecarpine (M8-M11)) as well as parent drug itself, including three phase I and 12 phase II metabolites were detected and identified in rat plasma. Hydroxylation, sulphate conjugation and glucuronidation were confirmed as the primary metabolic pathways for rutaecarpine in rat plasma. DISCUSSION AND CONCLUSION: These results provide an insight into the metabolism of rutaecarpine and also can give strong indications on the effective forms of rutaecarpine in vivo.


Asunto(s)
Cromatografía Líquida de Alta Presión , Medicamentos Herbarios Chinos/farmacocinética , Alcaloides Indólicos/farmacocinética , Espectrometría de Masas , Quinazolinas/farmacocinética , Administración Oral , Animales , Medicamentos Herbarios Chinos/administración & dosificación , Glucurónidos/farmacocinética , Hidroxilación , Alcaloides Indólicos/administración & dosificación , Alcaloides Indólicos/sangre , Masculino , Fase I de la Desintoxicación Metabólica , Fase II de la Desintoxicación Metabólica , Estructura Molecular , Quinazolinas/administración & dosificación , Quinazolinas/sangre , Ratas Sprague-Dawley , Extracción en Fase Sólida , Sulfatos/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA