Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Drug Metab Dispos ; 49(7): 572-580, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33941543

RESUMEN

Givosiran is an N-acetylgalactosamine-conjugated RNA interference therapeutic that targets 5'-aminolevulinate synthase 1 mRNA in the liver and is currently marketed for the treatment of acute hepatic porphyria. Herein, nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion properties of givosiran were characterized. Givosiran was completely absorbed after subcutaneous administration with relatively short plasma elimination half-life (t1/2; less than 4 hours). Plasma exposure increased approximately dose proportionally with no accumulation after repeat doses. Plasma protein binding was concentration dependent across all species tested and was around 90% at clinically relevant concentration in human. Givosiran predominantly distributed to the liver by asialoglycoprotein receptor-mediated uptake, and the t1/2 in the liver was significantly longer (∼1 week). Givosiran was metabolized by nucleases, not cytochrome P450 (P450) isozymes, across species with no human unique metabolites. Givosiran metabolized to form one primary active metabolite with the loss of one nucleotide from the 3' end of antisense strand, AS(N-1)3' givosiran, which was equipotent to givosiran. Renal and fecal excretion were minor routes of elimination of givosiran as approximately 10% and 16% of the dose was recovered intact in excreta of rats and monkeys, respectively. Givosiran is not a substrate, inhibitor, or inducer of P450 isozymes, and it is not a substrate or inhibitor of uptake and most efflux transporters. Thus, givosiran has a low potential of mediating drug-drug interactions involving P450 isozymes and drug transporters. SIGNIFICANCE STATEMENT: Nonclinical pharmacokinetics and absorption, distribution, metabolism, and excretion (ADME) properties of givosiran were characterized. Givosiran shows similar pharmacokinetics and ADME properties across rats and monkeys in vivo and across human and animal matrices in vitro. Subcutaneous administration results in adequate exposure of givosiran to the target organ (liver). These studies support the interpretation of toxicology studies, help characterize the disposition of givosiran in humans, and support the clinical use of givosiran for the treatment of acute hepatic porphyria.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Pirrolidinas/farmacocinética , 5-Aminolevulinato Sintetasa/antagonistas & inhibidores , Acetilgalactosamina/administración & dosificación , Acetilgalactosamina/farmacocinética , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Femenino , Semivida , Inyecciones Subcutáneas , Eliminación Intestinal , Macaca fascicularis , Masculino , Modelos Animales , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/tratamiento farmacológico , Pirrolidinas/administración & dosificación , Ratas , Eliminación Renal , Distribución Tisular
3.
Bioanalysis ; 11(21): 1955-1965, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31829055

RESUMEN

Aim: A novel single-stranded deaminated oligonucleotide metabolite resulting from a REVERSIR™ oligonucleotide was discovered and identified in monkey liver after subcutaneous administration. Results & methodology: REVERSIR-A and its metabolites were extracted from biological matrices by solid phase extraction and analyzed using LC coupled with high-resolution MS under negative ionization mode. A novel 9-mer metabolite of REVERSIR-A, resulting from deamination of the 3' terminal 2'-O-methyl-adenosine nucleotide to 2'-O-methyl-inosine, was discovered at significant levels in monkey liver. The metabolite's identity was confirmed by LC-MS/MS. Conclusion: This report describes the first observation of a long-chain deaminated metabolite of a single-stranded REVERSIR oligonucleotide in vivo in monkey liver.


Asunto(s)
Espectrometría de Masas/métodos , Oligonucleótidos/metabolismo , Animales , Desaminación , Inosina/metabolismo , Hígado/metabolismo , Macaca fascicularis
4.
Bioanalysis ; 11(21): 1967-1980, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31829056

RESUMEN

Aim: Advancements in RNA interference therapeutics have triggered development of improved bioanalytical methods for oligonucleotide metabolite profiling and high-throughput quantification in biological matrices. Results & methodology: HPLC coupled with high-resolution mass spectrometry (LC-HRMS) methods were developed to investigate the metabolism of a REVERSIR™ molecule in vivo. Plasma and tissue samples were extracted using solid-phase extraction followed by LC-HRMS analysis for metabolite profiling and quantification. The method was qualified from 10 to 5000 ng/ml (plasma) and 100 to 50000 ng/g (liver and kidney). In rat liver, intra and interday accuracy ranged from 80.9 to 118.5% and 88.4 to 111.9%, respectively, with acceptable precision (<20% CV). Conclusion: The LC-HRMS method can be applied for metabolite profiling and quantification of oligonucleotides in biological matrices.


Asunto(s)
Espectrometría de Masas/métodos , Metabolómica/métodos , Oligonucleótidos/metabolismo , Animales , Secuencia de Bases , Cromatografía Liquida , Hígado/metabolismo , Macaca fascicularis , Oligonucleótidos/sangre , Oligonucleótidos/genética , Oligonucleótidos/orina
5.
Nucleic Acids Res ; 47(7): 3306-3320, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30820542

RESUMEN

For oligonucleotide therapeutics, chemical modifications of the sugar-phosphate backbone are frequently used to confer drug-like properties. Because 2'-deoxy-2'-fluoro (2'-F) nucleotides are not known to occur naturally, their safety profile was assessed when used in revusiran and ALN-TTRSC02, two short interfering RNAs (siRNAs), of the same sequence but different chemical modification pattern and metabolic stability, conjugated to an N-acetylgalactosamine (GalNAc) ligand for targeted delivery to hepatocytes. Exposure to 2'-F-monomer metabolites was low and transient in rats and humans. In vitro, 2'-F-nucleoside 5'-triphosphates were neither inhibitors nor preferred substrates for human polymerases, and no obligate or non-obligate chain termination was observed. Modest effects on cell viability and mitochondrial DNA were observed in vitro in a subset of cell types at high concentrations of 2'-F-nucleosides, typically not attained in vivo. No apparent functional impact on mitochondria and no significant accumulation of 2'-F-monomers were observed after weekly administration of two GalNAc-siRNA conjugates in rats for ∼2 years. Taken together, the results support the conclusion that 2'-F nucleotides can be safely applied for the design of metabolically stabilized therapeutic GalNAc-siRNAs with favorable potency and prolonged duration of activity allowing for low dose and infrequent dosing.


Asunto(s)
Acetilgalactosamina/efectos adversos , Acetilgalactosamina/química , Desoxirribonucleótidos/efectos adversos , Desoxirribonucleótidos/química , Flúor/química , ARN Interferente Pequeño/efectos adversos , ARN Interferente Pequeño/química , Animales , Femenino , Flúor/efectos adversos , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
6.
Bioorg Med Chem Lett ; 27(2): 232-236, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27923619

RESUMEN

The design, synthesis and assessment of ß-carboline core-based compounds as potential multifunctional agents against several processes that are believed to play a significant role in Alzheimer's disease (AD) pathology, are described. The activity of the compounds was determined in Aß self-assembly (fibril and oligomer formation) and cholinesterase (AChE, BuChE) activity inhibition, and their antioxidant properties were also assessed. To obtain insight into the mode of action of the compounds, HR-MS studies were carried out on the inhibitor-Aß complex formation and molecular docking was performed on inhibitor-BuChE interactions. While several compounds exhibited strong activities in individual assays, compound 14 emerged as a promising multi-target lead for the further structure-activity relationship studies.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/análisis , Antioxidantes/farmacología , Carbolinas/farmacología , Inhibidores de la Colinesterasa/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/biosíntesis , Antioxidantes/síntesis química , Antioxidantes/química , Carbolinas/síntesis química , Carbolinas/química , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Colinesterasas/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad
7.
Cancer Res ; 72(6): 1494-503, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22266222

RESUMEN

Continued reliance on the androgen receptor (AR) is now understood as a core mechanism in castration-resistant prostate cancer (CRPC), the most advanced form of this disease. While established and novel AR pathway-targeting agents display clinical efficacy in metastatic CRPC, dose-limiting side effects remain problematic for all current agents. In this study, we report the discovery and development of ARN-509, a competitive AR inhibitor that is fully antagonistic to AR overexpression, a common and important feature of CRPC. ARN-509 was optimized for inhibition of AR transcriptional activity and prostate cancer cell proliferation, pharmacokinetics, and in vivo efficacy. In contrast to bicalutamide, ARN-509 lacked significant agonist activity in preclinical models of CRPC. Moreover, ARN-509 lacked inducing activity for AR nuclear localization or DNA binding. In a clinically valid murine xenograft model of human CRPC, ARN-509 showed greater efficacy than MDV3100. Maximal therapeutic response in this model was achieved at 30 mg/kg/d of ARN-509, whereas the same response required 100 mg/kg/d of MDV3100 and higher steady-state plasma concentrations. Thus, ARN-509 exhibits characteristics predicting a higher therapeutic index with a greater potential to reach maximally efficacious doses in man than current AR antagonists. Our findings offer preclinical proof of principle for ARN-509 as a promising therapeutic in both castration-sensitive and castration-resistant forms of prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Tiohidantoínas/uso terapéutico , Antagonistas de Andrógenos/farmacocinética , Anilidas/farmacocinética , Anilidas/uso terapéutico , Animales , Antineoplásicos Hormonales/sangre , Antineoplásicos Hormonales/farmacocinética , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Nitrilos/farmacocinética , Nitrilos/uso terapéutico , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/sangre , Feniltiohidantoína/farmacocinética , Feniltiohidantoína/uso terapéutico , Ratas , Receptores Androgénicos/efectos de los fármacos , Tiohidantoínas/sangre , Tiohidantoínas/síntesis química , Tiohidantoínas/farmacocinética , Compuestos de Tosilo/farmacocinética , Compuestos de Tosilo/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Res ; 71(15): 5164-74, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21646475

RESUMEN

In contrast to normal cells, cancer cells avidly take up glucose and metabolize it to lactate even when oxygen is abundant, a phenomenon referred to as the Warburg effect. This fundamental alteration in glucose metabolism in cancer cells enables their specific detection by positron emission tomography (PET) following i.v. injection of the glucose analogue (18)F-fluorodeoxy-glucose ((18)FDG). However, this useful imaging technique is limited by the fact that not all cancers avidly take up FDG. To identify molecular determinants of (18)FDG retention, we interrogated the transcriptomes of human-cancer cell lines and primary tumors for metabolic pathways associated with (18)FDG radiotracer uptake. From ninety-five metabolic pathways that were interrogated, the glycolysis, and several glycolysis-related pathways (pentose phosphate, carbon fixation, aminoacyl-tRNA biosynthesis, one-carbon-pool by folate) showed the greatest transcriptional enrichment. This "FDG signature" predicted FDG uptake in breast cancer cell lines and overlapped with established gene expression signatures for the "basal-like" breast cancer subtype and MYC-induced tumorigenesis in mice. Human breast cancers with nuclear MYC staining and high RNA expression of MYC target genes showed high (18)FDG-PET uptake (P < 0.005). Presence of the FDG signature was similarly associated with MYC gene copy gain, increased MYC transcript levels, and elevated expression of metabolic MYC target genes in a human breast cancer genomic dataset. Together, our findings link clinical observations of glucose uptake with a pathologic and molecular subtype of human breast cancer. Furthermore, they suggest related approaches to derive molecular determinants of radiotracer retention for other PET-imaging probes.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Radioisótopos de Flúor , Fluorodesoxiglucosa F18 , Perfilación de la Expresión Génica , Genes myc , Glucólisis , Proteínas de Neoplasias/biosíntesis , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Radiofármacos , Adenocarcinoma/clasificación , Adenocarcinoma/patología , Astrocitoma/metabolismo , Astrocitoma/patología , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Línea Celular Tumoral/metabolismo , Femenino , Radioisótopos de Flúor/farmacocinética , Fluorodesoxiglucosa F18/farmacocinética , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucólisis/genética , Humanos , Masculino , Melanoma/patología , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Radiofármacos/farmacocinética
10.
Science ; 324(5928): 787-90, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19359544

RESUMEN

Metastatic prostate cancer is treated with drugs that antagonize androgen action, but most patients progress to a more aggressive form of the disease called castration-resistant prostate cancer, driven by elevated expression of the androgen receptor. Here we characterize the diarylthiohydantoins RD162 and MDV3100, two compounds optimized from a screen for nonsteroidal antiandrogens that retain activity in the setting of increased androgen receptor expression. Both compounds bind to the androgen receptor with greater relative affinity than the clinically used antiandrogen bicalutamide, reduce the efficiency of its nuclear translocation, and impair both DNA binding to androgen response elements and recruitment of coactivators. RD162 and MDV3100 are orally available and induce tumor regression in mouse models of castration-resistant human prostate cancer. Of the first 30 patients treated with MDV3100 in a Phase I/II clinical trial, 13 of 30 (43%) showed sustained declines (by >50%) in serum concentrations of prostate-specific antigen, a biomarker of prostate cancer. These compounds thus appear to be promising candidates for treatment of advanced prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos/uso terapéutico , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata/tratamiento farmacológico , Antagonistas de Andrógenos/metabolismo , Antagonistas de Andrógenos/farmacocinética , Antagonistas de Andrógenos/farmacología , Anilidas/metabolismo , Anilidas/farmacología , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Benzamidas , Disponibilidad Biológica , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Nitrilos/metabolismo , Nitrilos/farmacología , Feniltiohidantoína/metabolismo , Feniltiohidantoína/farmacocinética , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata/patología , Receptores Androgénicos/química , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Compuestos de Tosilo/metabolismo , Compuestos de Tosilo/farmacología , Transcripción Genética/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Cell ; 11(6): 555-69, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17560336

RESUMEN

Although most oncogenic phenotypes of PTEN loss are attributed to AKT activation, AKT alone is not sufficient to induce all of the biological activities associated with PTEN inactivation. We searched for additional PTEN-regulated pathways through gene set enrichment analysis (GSEA) and identified genes associated with JNK activation. PTEN null cells exhibit higher JNK activity, and genetic studies demonstrate that JNK functions parallel to and independently of AKT. Furthermore, PTEN deficiency sensitizes cells to JNK inhibition and negative feedback regulation of PI3K was impaired in PTEN null cells. Akt and JNK activation are highly correlated in human prostate cancer. These findings implicate JNK in PI3K-driven cancers and demonstrate the utility of GSEA to identify functional pathways using genetically defined systems.


Asunto(s)
Transformación Celular Neoplásica , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Activación Enzimática , Retroalimentación Fisiológica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
12.
J Nucl Med ; 47(6): 999-1006, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16741310

RESUMEN

UNLABELLED: Small-animal PET scanning with (18)F-FDG is increasingly used in murine models of human diseases. However, the impact of dietary conditions, mode of anesthesia, and ambient temperature on the biodistribution of (18)F-FDG in mice has not been systematically studied so far. The aim of this study was to determine how these factors affect assessment of tumor glucose use by (18)F-FDG PET and to develop an imaging protocol that optimizes visualization of tumor xenografts. METHODS: Groups of severe combined immunodeficient (SCID) mice were first imaged by microPET with free access to food, at room temperature (20 degrees C), and no anesthesia during the uptake period (reference condition). Subsequently, the impact of (a) fasting for 8-12 h, (b) warming the animals with a heating pad (30 degrees C), and (c) general anesthesia using isoflurane or ketamine/xylazine on the (18)F-FDG biodistribution was evaluated. Subcutaneously implanted human A431 epidermoid carcinoma and U251 glioblastoma cells served as tumor models. RESULTS: Depending on the study conditions, (18)F-FDG uptake by normal tissues varied 3-fold for skeletal muscle, 13-fold for brown adipose tissue, and 15-fold for myocardium. Warming and fasting significantly reduced the intense (18)F-FDG uptake by brown adipose tissue observed under the reference condition and markedly improved visualization of tumor xenografts. Although tumor (18)F-FDG uptake was not above background activity under the reference condition, tumors demonstrated marked focal (18)F-FDG uptake in warmed and fasted animals. Quantitatively, tumor (18)F-FDG uptake increased 4-fold and tumor-to-organ ratios were increased up to 17-fold. Ketamine/xylazine anesthesia caused marked hyperglycemia and was not further evaluated. Isoflurane anesthesia only mildly increased blood glucose levels and had no significant effect on tumor (18)F-FDG uptake. Isoflurane markedly reduced (18)F-FDG uptake by brown adipose tissue and skeletal muscle but increased the activity concentration in liver, myocardium, and kidney. CONCLUSION: Animal handling has a dramatic effect on (18)F-FDG biodistribution and significantly influences the results of microPET studies in tumor-bearing mice. To improve tumor visualization mice should be fasted and warmed before (18)F-FDG injection and during the uptake period. Isoflurane appears well suited for anesthesia of tumor-bearing mice, whereas ketamine/xylazine should be used with caution, as it may induce marked hyperglycemia.


Asunto(s)
Crianza de Animales Domésticos/métodos , Artefactos , Conducta Animal/fisiología , Fluorodesoxiglucosa F18/farmacocinética , Actividad Motora/fisiología , Tomografía de Emisión de Positrones/veterinaria , Animales , Ambiente , Masculino , Tasa de Depuración Metabólica , Ratones , Especificidad de Órganos , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular , Imagen de Cuerpo Entero/métodos , Imagen de Cuerpo Entero/veterinaria
13.
Nat Med ; 12(1): 122-7, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16341243

RESUMEN

Inhibitors of the kinase mammalian target of rapamycin (mTOR) have shown sporadic activity in cancer trials, leading to confusion about the appropriate clinical setting for their use. Here we show that loss of the Von Hippel-Lindau tumor suppressor gene (VHL) sensitizes kidney cancer cells to the mTOR inhibitor CCI-779 in vitro and in mouse models. Growth arrest caused by CCI-779 correlates with a block in translation of mRNA encoding hypoxia-inducible factor (HIF1A), and is rescued by expression of a VHL-resistant HIF1A cDNA lacking the 5' untranslated region. VHL-deficient tumors show increased uptake of the positron emission tomography (PET) tracer fluorodeoxyglucose (FDG) in an mTOR-dependent manner. Our findings provide preclinical rationale for prospective, biomarker-driven clinical studies of mTOR inhibitors in kidney cancer and suggest that FDG-PET scans may have use as a pharmacodynamic marker in this setting.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Renales/metabolismo , Proteínas Quinasas/metabolismo , Regiones no Traducidas 5' , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , ADN/química , Cartilla de ADN/química , ADN Complementario/metabolismo , Densitometría , Fluorodesoxiglucosa F18/farmacología , Glucosa/metabolismo , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Immunoblotting , Luciferasas/metabolismo , Ratones , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa , Tomografía de Emisión de Positrones , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Radiofármacos/farmacología , Serina-Treonina Quinasas TOR , Factores de Tiempo , Transfección
14.
Nature ; 434(7035): 921-6, 2005 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-15829968

RESUMEN

Defining the molecular strategies that integrate diverse signalling pathways in the expression of specific gene programmes that are critical in homeostasis and disease remains a central issue in biology. This is particularly pertinent in cancer biology because downregulation of tumour metastasis suppressor genes is a common occurrence, and the underlying molecular mechanisms are not well established. Here we report that the downregulation of a metastasis suppressor gene, KAI1, in prostate cancer cells involves the inhibitory actions of beta-catenin, along with a reptin chromatin remodelling complex. This inhibitory function of beta-catenin-reptin requires both increased beta-catenin expression and recruitment of histone deacetylase activity. The coordinated actions of beta-catenin-reptin components that mediate the repressive state serve to antagonize a Tip60 coactivator complex that is required for activation; the balance of these opposing complexes controls the expression of KAI1 and metastatic potential. The molecular mechanisms underlying the antagonistic regulation of beta-catenin-reptin and the Tip60 coactivator complexes for the metastasis suppressor gene, KAI1, are likely to be prototypic of a selective downregulation strategy for many genes, including a subset of NF-kappaB target genes.


Asunto(s)
Acetiltransferasas/metabolismo , Antígenos CD/genética , Proteínas del Citoesqueleto/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Glicoproteínas de Membrana/genética , Metástasis de la Neoplasia/genética , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/metabolismo , Transcripción Genética/genética , Acetiltransferasas/genética , Animales , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Colágeno , Regulación hacia Abajo/genética , Combinación de Medicamentos , Histona Acetiltransferasas , Humanos , Proteína Kangai-1 , Laminina , Lisina Acetiltransferasa 5 , Masculino , Ratones , FN-kappa B/metabolismo , Trasplante de Neoplasias , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/metabolismo , Proteoglicanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , beta Catenina
15.
J Nucl Med ; 46(1): 114-20, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632041

RESUMEN

UNLABELLED: The aim of this study was to evaluate, whether PET with (18)F-FDG and 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) may be used to monitor noninvasively the antiproliferative effects of tyrosine kinase inhibitors. METHODS: Using a high-resolution small animal scanner, we measured the effect of the ErbB-selective kinase inhibitor PKI-166 on the (18)F-FDG and (18)F-FLT uptake of ErbB1-overexpressing A431 xenograft tumors. RESULTS: Treatment with PKI-166 markedly lowered tumor (18)F-FLT uptake within 48 h of drug exposure; within 1 wk (18)F-FLT uptake decreased by 79%. (18)F-FLT uptake by the xenografts significantly correlated with the tumor proliferation index as determined by proliferating cell nuclear antigen staining (r = 0.71). Changes in (18)F-FLT uptake did not reflect inhibition of ErbB kinase activity itself but, rather, the effects of kinase inhibition on tumor cell proliferation. Tumor (18)F-FDG uptake generally paralleled the changes seen for (18)F-FLT. However, the baseline signal was significantly lower than that for (18)F-FLT. CONCLUSION: These results indicate that (18)F-FLT PET provides noninvasive, quantitative, and repeatable measurements of tumor cell proliferation during treatment with ErbB kinase inhibitors and provide a rationale for the use this technology in clinical trials of kinase inhibitors.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Carcinoma de Células Escamosas/metabolismo , Didesoxinucleósidos/farmacocinética , Fluorodesoxiglucosa F18/farmacocinética , Invasividad Neoplásica/diagnóstico por imagen , Invasividad Neoplásica/prevención & control , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Radiofármacos/farmacocinética , Resultado del Tratamiento
16.
Cancer Cell ; 6(5): 517-27, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15542435

RESUMEN

Given the role of the EGFR/HER2 family of tyrosine kinases in breast cancer, we dissected the molecular basis of EGFR/HER2 kinase signaling in prostate cancer. Using the small molecule dual EGFR/HER2 inhibitor PKI-166, we show that the biologic effects of EGFR/HER-2 pathway inhibition are caused by reduced AR transcriptional activity. Additional genetic and pharmacologic experiments show that this modulation of AR function is mediated by the HER2/ERBB3 pathway, not by EGFR. This HER2/ERBB3 signal stabilizes AR protein levels and optimizes binding of AR to promoter/enhancer regions of androgen-regulated genes. Surprisingly, the downstream signaling pathway responsible for these effects appears to involve kinases other than Akt. These data suggest that the HER2/ERBB3 pathway is a critical target in hormone-refractory prostate cancer.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , Receptor ErbB-2/fisiología , Receptores Androgénicos/metabolismo , Animales , Células COS , Chlorocebus aethiops , ADN de Neoplasias/metabolismo , Receptores ErbB/fisiología , Humanos , Masculino , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , ARN Interferente Pequeño/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal , Transfección , Células Tumorales Cultivadas
17.
Science ; 305(5682): 399-401, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15256671

RESUMEN

Resistance to the ABL kinase inhibitor imatinib (STI571 or Gleevec) in chronic myeloid leukemia (CML) occurs through selection for tumor cells harboring BCR-ABL kinase domain point mutations that interfere with drug binding. Crystallographic studies predict that most imatinib-resistant mutants should remain sensitive to inhibitors that bind ABL with less stringent conformational requirements. BMS-354825 is an orally bioavailable ABL kinase inhibitor with two-log increased potency relative to imatinib that retains activity against 14 of 15 imatinib-resistant BCR-ABL mutants. BMS-354825 prolongs survival of mice with BCR-ABL-driven disease and inhibits proliferation of BCR-ABL-positive bone marrow progenitor cells from patients with imatinib-sensitive and imatinib-resistant CML. These data illustrate how molecular insight into kinase inhibitor resistance can guide the design of second-generation targeted therapies.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Sustitución de Aminoácidos , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Benzamidas , Sitios de Unión , División Celular/efectos de los fármacos , Línea Celular , Ensayos Clínicos Fase I como Asunto , Dasatinib , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Mesilato de Imatinib , Ratones , Ratones SCID , Mutación , Piperazinas/uso terapéutico , Conformación Proteica , Pirimidinas/metabolismo , Pirimidinas/uso terapéutico , Tiazoles/metabolismo , Tiazoles/uso terapéutico , Transfección
18.
Cancer Res ; 64(4): 1252-4, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14973081

RESUMEN

An isocaloric low-fat diet has been shown to slow androgen-sensitive Los Angeles Prostate Cancer-4 (LAPC-4) tumor growth in a mouse xenograft model. LAPC-4 cells were injected into male severe combined immunodeficient mice. After palpable tumors developed, the mice were divided into three groups, high-fat intact, high-fat castration, and low-fat castration. Tumor latency (18 versus 9 weeks; P < 0.001) and mouse survival (20.8 +/- 1.3 versus 13 +/- 0.7 weeks; P < 0.01) were significantly longer in the low-fat castration versus high-fat castration group. Reduced dietary fat intake delayed conversion from androgen-sensitive to -insensitive prostate cancer and significantly prolonged survival of severe combined immunodeficient mice bearing LAPC-4 xenografts.


Asunto(s)
Andrógenos/fisiología , Grasas de la Dieta/administración & dosificación , Neoplasias de la Próstata/dietoterapia , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/patología , Trasplante Heterólogo
19.
Nat Med ; 10(1): 33-9, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702632

RESUMEN

Using microarray-based profiling of isogenic prostate cancer xenograft models, we found that a modest increase in androgen receptor mRNA was the only change consistently associated with the development of resistance to antiandrogen therapy. This increase in androgen receptor mRNA and protein was both necessary and sufficient to convert prostate cancer growth from a hormone-sensitive to a hormone-refractory stage, and was dependent on a functional ligand-binding domain. Androgen receptor antagonists showed agonistic activity in cells with increased androgen receptor levels; this antagonist-agonist conversion was associated with alterations in the recruitment of coactivators and corepressors to the promoters of androgen receptor target genes. Increased levels of androgen receptor confer resistance to antiandrogens by amplifying signal output from low levels of residual ligand, and by altering the normal response to antagonists. These findings provide insight toward the design of new antiandrogens.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias de la Próstata/tratamiento farmacológico , Antagonistas de Andrógenos/farmacología , Antineoplásicos Hormonales/farmacología , Secuencia de Bases , Cartilla de ADN , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Receptores Androgénicos/fisiología
20.
J Biol Chem ; 279(4): 2737-46, 2004 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-14576155

RESUMEN

Prior work demonstrates that AKT activity regulates sensitivity of cells to G(1) arrest induced by mammalian target of rapamycin (mTOR) inhibitors such as rapamycin and CCI-779. To investigate this, a novel high-throughput microarray polysome analysis was performed to identify genes whose mRNA translational efficiency was differentially affected following mTOR inhibition. The analysis also allowed the assessment of steady-state transcript levels. We identified two transcripts, cyclin D1 and c-myc, which exhibited differential expression in an AKT-dependent manner: High levels of activated AKT resulted in rapamycin-induced down-regulation of expression, whereas low levels resulted in up-regulation of expression. To ectopically express these proteins we exploited the finding that the p27(kip1) mRNA was efficiently translated in the face of mTOR inhibition irrespective of AKT activity. Thus, the p27(kip1) 5'-untranslated region was fused to the cyclin D1 and c-myc coding regions and these constructs were expressed in cells. In transfected cells, expression of cyclin D1 or c-myc was not decreased by rapamycin. Most importantly, this completely converted sensitive cells to a phenotype resistant to G(1) arrest. Furthermore, the AKT-dependent differential expression patterns of these two genes was also observed in a mouse xenograft model following in vivo treatment with CCI-779. These results identify two critical downstream molecular targets whose expression is regulated by AKT activity and whose down-regulation is required for rapamycin/CCI-779 sensitivity.


Asunto(s)
Ciclina D1/biosíntesis , Inhibidores de Proteínas Quinasas , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Sirolimus/análogos & derivados , Línea Celular , Ciclina D1/genética , Fase G1/efectos de los fármacos , Fase G1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Genes myc , Humanos , Proteínas Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...