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.
Eur J Med Chem ; 270: 116375, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38604095

RESUMEN

Spleen tyrosine kinase (Syk) is a key signal transduction mediator of the B cell receptor (BCR) signaling pathway. Abnormal BCR signaling plays a key role in initiation and development of B-cell-derived hematological malignancies, therefore, Syk represents a potential target for inhibiting the BCR signaling resulting in a therapeutic effect in these cancers. Herein, we describe a novel series of SYK inhibitors with 4-(3'-pyrazolyl)-2-amino-pyrimidine scaffold. Extensive study of structure-activity relationships led to the identification of 1 (NMS-0963), a highly potent Syk inhibitor (IC50 = 3 nM) endowed with high selectivity within a panel of tested kinases and high antiproliferative activity in SYK-dependent BaF3-TEL/SYK cells and in other BCR-dependent hematological tumor cell lines. Additionally, 1 effectively inhibited Syk phosphorylation and downstream signaling mediators of the BCR in treated cells. In in vivo pharmacokinetics studies, 1, displayed good pharmacokinetics properties, with linear exposure with dose and excellent oral bioavailability. These findings suggest that 1 is a promising new Syk inhibitor for treating BCR-dependent hematological cancers.


Asunto(s)
Neoplasias Hematológicas , Proteínas Tirosina Quinasas , Pirimidinas , Humanos , Quinasa Syk/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Fosforilación , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico
2.
Nat Commun ; 10(1): 3151, 2019 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320631

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
J Med Chem ; 62(12): 5863-5884, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31070916

RESUMEN

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that cross-links collagens and elastin in the extracellular matrix and is a critical mediator of tumor growth and metastatic spread. LOX is a target for cancer therapy, and thus the search for therapeutic agents against LOX has been widely sought. We report herein the medicinal chemistry discovery of a series of LOX inhibitors bearing an aminomethylenethiophene (AMT) scaffold. High-throughput screening provided the initial hits. Structure-activity relationship (SAR) studies led to the discovery of AMT inhibitors with sub-micromolar half-maximal inhibitory concentrations (IC50) in a LOX enzyme activity assay. Further SAR optimization yielded the orally bioavailable LOX inhibitor CCT365623 with good anti-LOX potency, selectivity, pharmacokinetic properties, as well as anti-metastatic efficacy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Administración Oral , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Metástasis de la Neoplasia/tratamiento farmacológico , Relación Estructura-Actividad , Tiofenos/química , Tiofenos/farmacocinética , Tiofenos/farmacología , Tiofenos/uso terapéutico
4.
Nat Commun ; 8: 14909, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28416796

RESUMEN

Lysyl oxidase (LOX) remodels the tumour microenvironment by cross-linking the extracellular matrix. LOX overexpression is associated with poor cancer outcomes. Here, we find that LOX regulates the epidermal growth factor receptor (EGFR) to drive tumour progression. We show that LOX regulates EGFR by suppressing TGFß1 signalling through the secreted protease HTRA1. This increases the expression of Matrilin2 (MATN2), an EGF-like domain-containing protein that traps EGFR at the cell surface to facilitate its activation by EGF. We describe a pharmacological inhibitor of LOX, CCT365623, which disrupts EGFR cell surface retention and delays the growth of primary and metastatic tumour cells in vivo. Thus, we show that LOX regulates EGFR cell surface retention to drive tumour progression, and we validate the therapeutic potential of inhibiting this pathway with the small molecule inhibitor CCT365623.


Asunto(s)
Membrana Celular/metabolismo , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Aminopropionitrilo/química , Aminopropionitrilo/farmacología , Animales , Técnicas Biosensibles , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Perros , Activación Enzimática , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Humanos , Proteínas Matrilinas/metabolismo , Ratones , Modelos Biológicos , Metástasis de la Neoplasia , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
6.
Cancer Discov ; 6(3): 286-99, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26715644

RESUMEN

UNLABELLED: Targeted therapies and immunotherapies have transformed melanoma care, extending median survival from ∼9 to over 25 months, but nevertheless most patients still die of their disease. The aim of precision medicine is to tailor care for individual patients and improve outcomes. To this end, we developed protocols to facilitate individualized treatment decisions for patients with advanced melanoma, analyzing 364 samples from 214 patients. Whole exome sequencing (WES) and targeted sequencing of circulating tumor DNA (ctDNA) allowed us to monitor responses to therapy and to identify and then follow mechanisms of resistance. WES of tumors revealed potential hypothesis-driven therapeutic strategies for BRAF wild-type and inhibitor-resistant BRAF-mutant tumors, which were then validated in patient-derived xenografts (PDX). We also developed circulating tumor cell-derived xenografts (CDX) as an alternative to PDXs when tumors were inaccessible or difficult to biopsy. Thus, we describe a powerful technology platform for precision medicine in patients with melanoma. SIGNIFICANCE: Although recent developments have revolutionized melanoma care, most patients still die of their disease. To improve melanoma outcomes further, we developed a powerful precision medicine platform to monitor patient responses and to identify and validate hypothesis-driven therapies for patients who do not respond, or who develop resistance to current treatments.


Asunto(s)
Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Melanoma/diagnóstico , Melanoma/tratamiento farmacológico , Medicina de Precisión , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biopsia , Análisis por Conglomerados , Manejo de la Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Terapia Molecular Dirigida , Mutación , Estadificación de Neoplasias , Reproducibilidad de los Resultados , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
7.
EMBO Mol Med ; 7(8): 1063-76, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26077591

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Animales , Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Humanos , Hipoxia , Ratones , Metástasis de la Neoplasia/prevención & control , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Resultado del Tratamiento , Gemcitabina
8.
Cancer Cell ; 27(1): 85-96, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25500121

RESUMEN

BRAF and MEK inhibitors are effective in BRAF mutant melanoma, but most patients eventually relapse with acquired resistance, and others present intrinsic resistance to these drugs. Resistance is often mediated by pathway reactivation through receptor tyrosine kinase (RTK)/SRC-family kinase (SFK) signaling or mutant NRAS, which drive paradoxical reactivation of the pathway. We describe pan-RAF inhibitors (CCT196969, CCT241161) that also inhibit SFKs. These compounds do not drive paradoxical pathway activation and inhibit MEK/ERK in BRAF and NRAS mutant melanoma. They inhibit melanoma cells and patient-derived xenografts that are resistant to BRAF and BRAF/MEK inhibitors. Thus, paradox-breaking pan-RAF inhibitors that also inhibit SFKs could provide first-line treatment for BRAF and NRAS mutant melanomas and second-line treatment for patients who develop resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Pirazinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/patología , Melanoma Experimental , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Oncol ; 8(6): 1140-58, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25178978

RESUMEN

The discovery that BRAF is a driver oncogene in cancer, and complementary improvements in our understanding of the immune system have resulted in new targeted and immune-therapies for metastatic melanoma. Targeted therapies achieve impressive clinical results in carefully selected patients but the development of resistance seems inevitable in most cases. Conversely, immune-checkpoints inhibitors can achieve long-term remission and cures, but in a smaller proportion of patients, and biomarkers to predict which patients will respond are not available. Nevertheless, melanoma has led the evolution of cancer treatment from relatively nonspecific cytotoxic agents to highly selective therapies and here we review the lessons from this paradigm shift in treatment and the opportunities for further improvements in outcomes for melanoma patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Melanoma/terapia , Terapia Molecular Dirigida/métodos , Neoplasias Cutáneas/terapia , Animales , Antineoplásicos/farmacología , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Piel/inmunología , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
10.
Sci Signal ; 7(318): ra30, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24667377

RESUMEN

Melanoma is a highly metastatic and lethal form of skin cancer. The protein kinase BRAF is mutated in about 40% of melanomas, and BRAF inhibitors improve progression-free and overall survival in these patients. However, after a relatively short period of disease control, most patients develop resistance because of reactivation of the RAF-ERK (extracellular signal-regulated kinase) pathway, mediated in many cases by mutations in RAS. We found that BRAF inhibition induces invasion and metastasis in RAS mutant melanoma cells through a mechanism mediated by the reactivation of the MEK (mitogen-activated protein kinase kinase)-ERK pathway, increased expression and secretion of interleukin 8, and induction of protease-dependent invasion. These events were accompanied by a cell morphology switch from predominantly rounded to predominantly elongated cells. We also observed similar responses in BRAF inhibitor-resistant melanoma cells. These data show that BRAF inhibitors can induce melanoma cell invasion and metastasis in tumors that develop resistance to these drugs.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Melanoma/fisiopatología , Invasividad Neoplásica/fisiopatología , Metástasis de la Neoplasia/fisiopatología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/fisiopatología , Animales , Western Blotting , Forma de la Célula/fisiología , Dimetilsulfóxido , Resistencia a Antineoplásicos , Ensayo de Inmunoadsorción Enzimática , Humanos , Indoles/farmacología , Interleucina-8/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Estadísticas no Paramétricas , Sulfonamidas/farmacología
11.
Oncotarget ; 4(8): 1185-98, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23852390

RESUMEN

TRAIL has been shown to induce apoptosis in cancer cells, but in some cases they fail to respond to this ligand. We explored the ability of representative phosphatidylinositol-3-kinase (PI3 Kinase)/mTOR and HSP90 inhibitors to overcome TRAIL resistance by increasing apoptosis in colorectal cancer models. We determined the sensitivity of 27 human colorectal cancer and 2 non-transformed colon epithelial cell lines to TRAIL treatment. A subset of the cancer cell lines with a range of responses to TRAIL was selected from the panel for treatment with TRAIL combined with the PI3 Kinase/mTOR inhibitor PI-103 or the HSP90 inhibitor 17-AAG (tanespimycin). Two TRAIL-resistant cell lines were selected for in vivo combination studies with TRAIL and 17-AAG. We found that 13 colorectal cancer cell lines and the 2 non-transformed colon epithelial cell lines were resistant to TRAIL. We demonstrated that co-treatment of TRAIL and PI-103 or 17-AAG was synergistic or additive and significantly enhanced apoptosis in colorectal cancer cells. This was associated with decreased expression or activity of survival protein biomarkers such as ERBB2, AKT, IKKα and XIAP. In contrast, the effect of the combination treatments in non-transformed colon cells was minimal. We show here for the first time that co-treatment in vivo with TRAIL and 17-AAG in two TRAIL-resistant human colorectal cancer xenograft models resulted in significantly greater tumor growth inhibition compared to single treatments. We propose that combining TRAIL with PI3 Kinase/mTOR or HSP90 inhibitors has therapeutic potential in the treatment of TRAIL-resistant colorectal cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benzoquinonas/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Apoptosis/efectos de los fármacos , Benzoquinonas/administración & dosificación , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ensayos de Selección de Medicamentos Antitumorales , Furanos/administración & dosificación , Furanos/farmacología , Células HCT116 , Proteínas HSP90 de Choque Térmico/metabolismo , Células HT29 , Humanos , Lactamas Macrocíclicas/administración & dosificación , Ratones , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Piridinas/administración & dosificación , Piridinas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Toxicol Pathol ; 35(7): 972-83, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18098043

RESUMEN

Protein-kinase inhibitors are among the most advanced compounds in development using the new drug discovery paradigm of developing small-molecule drugs against specific molecular targets in cancer. After treatment with a cyclin dependent kinase CDK2 inhibitor in monkey, histopathological analysis of the eye showed specific cellular damage in the photoreceptor layer. Since this CDK2 inhibitor showed activity also on other CDKs, in order to investigate the mechanism of toxicity of this compound, we isolated cones and rods from the retina of normal monkey and humans by Laser Capture Microdissection. Using Real-Time PCR we first measured the expression of cyclin dependent protein-kinases (CDK)1, 2, 4, 5, Glycogen synthase kinase 3beta (GSK3beta) and microtubule associated protein TAU. We additionally verified the presence of these proteins in monkey eye sections by immuno-histochemistry and immunofluorescence analysis and afterwards quantified GSK3beta, phospho-GSK3beta and TAU by Reverse Phase Protein Microarrays. With this work we demonstrate how complementary gene expression and protein-based technologies constitute a powerful tool for the understanding of the molecular mechanism of a CDK2 inhibitor induced toxicity. Moreover, this investigative approach is helpful to better understand and characterize the mechanism of species-specific toxicities and further support a rational, molecular mechanism-based safety assessment in humans.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Serina-Treonina Quinasas/análisis , Retina/efectos de los fármacos , Retina/enzimología , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3/análisis , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inmunohistoquímica , Macaca fascicularis , Masculino , Microdisección , Fosforilación , Reacción en Cadena de la Polimerasa , Retina/patología , Proteínas tau/análisis
13.
Curr Opin Drug Discov Devel ; 10(1): 53-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17265742

RESUMEN

Currently, several protein kinase-modulating compounds have received market approval across a range of diverse therapeutic indications. Furthermore, a large number of chemical and biological protein kinase-modulating compounds are undergoing testing at the preclinical and clinical level. Protein kinases are both major pharmacological targets and diagnostically useful. Progression of kinase modulators toward clinically viable therapies is aided by a reversible mechanism of action, short treatment durations and patient-compliant administration routes. However, the physiological role and essential functional activity of protein kinases in many organs and tissues complicates, to different extents, the development of useful, highly potent protein kinase modulators. In this review, we will highlight common problems in the development of these compounds and lessons learned from the extensive preclinical and clinical characterization of some key protein kinase modulators, some of which have either entered and successfully completed clinical trials or have been abandoned as a consequence of unacceptable toxicity issues. We will ultimately explore how molecular profiling tools combined with histopathological endpoints can be adopted to address and further understand these toxicities in humans and understand their relevance and characterization when identified during early animal experiments.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Inhibidores de Proteínas Quinasas/efectos adversos , Animales , Ensayos Clínicos como Asunto , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico
14.
Toxicol In Vitro ; 20(1): 125-31, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16098711

RESUMEN

The scope of this study was to compare in vitro and in vivo cytochrome P450 (CYP) gene induction in mice, using liver slices as an in vitro model. We have chosen to study mice to be able to better interpret CYP induction during long-term safety studies in this species. Mouse liver slices were incubated with beta-naphthoflavone (betaNF), phenobarbital (PB) or dexamethasone (DEX) for 24 h. In addition, in an in vivo study, mice were treated with the same compounds for three days. The mRNA expression of cyp1a1, cyp1a2, cyp2b10 and cyp3a11, which are important for drug metabolism and inducible by xenobiotics, were investigated in vivo and in vitro by real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). Both in mouse liver slices and in vivo, betaNF was found to be a potent inducer of cyp1a1 and to a lesser extent of cyp1a2. All three compounds induced cyp2b10 mRNA levels, while the cyp3a11 mRNA level was induced only by DEX. Overall, these data demonstrated a good predictive in vitro-in vivo correlation of CYP induction.


Asunto(s)
Bioensayo , Sistema Enzimático del Citocromo P-450/metabolismo , Hígado/enzimología , Animales , Sistema Enzimático del Citocromo P-450/genética , Dexametasona/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos , Fenobarbital/farmacología , ARN Mensajero/metabolismo , Activación Transcripcional , beta-naftoflavona/farmacología
15.
Chem Biol Interact ; 151(1): 1-11, 2004 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-15607757

RESUMEN

Xenobiotics, including drugs, can influence cytochrome P450 (CYP) activity by upregulating the transcription of CYP genes. To minimize potential drug interactions, it is important to ascertain whether a compound will be an inducer of CYP enzymes early in the development of new therapeutic agents. In vivo and in vitro studies are reported that demonstrate the use of liver and intestinal slices as an in vitro model to predict potential CYP induction in vivo. Rat liver slices and intestinal slices were incubated, for 24 h and 6 h, respectively, with beta-naphthoflavone (betaNF), phenobarbital (PB) or dexamethasone (DEX). In an in vivo study, rats were treated with the same compounds for 3 days. In vivo and in vitro CYP mRNA levels were measured by using real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). In addition, CYP enzyme activities were determined in rat liver slices after 48 h incubation. In both rat liver and intestinal slices, betaNF significantly induced CYP1A1, CYP1A2 and CYP2B1 mRNA levels. PB significantly induced CYP2B1. In liver slices a minor induction of CYP1A1 and CYP3A1 by PB was observed, whereas DEX significantly induced CYP3A1, CYP2B1 and CYP1A2 mRNA levels. The induction profiles (qualitative and quantitative) observed in vivo and in vitro are quite similar. All together, these data demonstrate that liver and intestinal slices are a useful and predictive tool to study CYP induction.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Intestinos/efectos de los fármacos , Hígado/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Animales , Secuencia de Bases , Sistema Enzimático del Citocromo P-450/genética , Cartilla de ADN , Dexametasona/farmacología , Inducción Enzimática , Técnicas In Vitro , Intestinos/enzimología , Hígado/enzimología , Masculino , Fenobarbital/farmacología , ARN Mensajero/genética , Ratas , Ratas Wistar , beta-naftoflavona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA