Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Ther ; 116(1): 186-193, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38654484

RESUMEN

HepQuant tests quantify liver function from clearance of deuterium- and 13C-labeled cholates administered either intravenously and orally (SHUNT) or orally (DuO). Hepatic impairment studies have relied on clinical or laboratory criteria like Child-Pugh classification to categorize the degree of hepatic dysfunction. We compared HepQuant tests with Child-Pugh classification in predicting the pharmacokinetics of ampreloxetine. Twenty-one subjects with hepatic impairment (8 Child-Pugh A, 7 Child-Pugh B, and 6 Child-Pugh C), and 10 age- and sex-matched controls were studied. The pharmacokinetics of ampreloxetine were measured after oral administration of a single dose of 10 mg. Disease severity index (DSI), portal-systemic shunting (SHUNT%), hepatic reserve, and hepatic filtration rates (HFRs) were measured from serum samples obtained after intravenous administration of [24-13C]-cholate and oral administration of [2,2,4,4-2H]cholate. Ampreloxetine plasma exposure (AUC0-inf) was similar to controls in Child-Pugh A, increased 1.7-fold in subjects with Child-Pugh B, and 2.5-fold in subjects with Child-Pugh C and correlated with both Child-Pugh score and HepQuant parameters. The variability observed in ampreloxetine exposure (AUC0-inf) in subjects with moderate (Child-Pugh B) and severe hepatic impairment (Child-Pugh C) was explained by HepQuant parameters. Multivariable regression models demonstrated that DSI, SHUNT%, and Hepatic Reserve from SHUNT and DuO were superior predictors of ampreloxetine exposure (AUC0-inf) compared to Child-Pugh score. HepQuant DSI, SHUNT%, and hepatic reserve were more useful predictors of drug exposure than Child-Pugh class for ampreloxetine and thus may better optimize dose recommendations in patients with liver disease. The simple-to-administer, oral-only DuO version of the HepQuant test could enhance clinical utility.


Asunto(s)
Hepatopatías , Morfolinas , Humanos , Masculino , Femenino , Persona de Mediana Edad , Hepatopatías/metabolismo , Anciano , Administración Oral , Morfolinas/farmacocinética , Morfolinas/administración & dosificación , Adulto , Pruebas de Función Hepática/métodos , Índice de Severidad de la Enfermedad , Isótopos de Carbono , Deuterio , Hígado/metabolismo , Alcohol Feniletílico/análogos & derivados
2.
Science ; 367(6477): 586-590, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32001657

RESUMEN

The initiating mutations that contribute to cancer development are sometimes present in premalignant cells. Whether therapies targeting these mutations can eradicate premalignant cells is unclear. Acute myeloid leukemia (AML) is an attractive system for investigating the effect of preventative treatment because this disease is often preceded by a premalignant state (clonal hematopoiesis or myelodysplastic syndrome). In Npm1c/Dnmt3a mutant knock-in mice, a model of AML development, leukemia is preceded by a period of extended myeloid progenitor cell proliferation and self-renewal. We found that this self-renewal can be reversed by oral administration of a small molecule (VTP-50469) that targets the MLL1-Menin chromatin complex. These preclinical results support the hypothesis that individuals at high risk of developing AML might benefit from targeted epigenetic therapy in a preventative setting.


Asunto(s)
Terapia Genética/métodos , Leucemia Experimental/prevención & control , Leucemia Mieloide Aguda/prevención & control , Proteínas Nucleares/genética , Preleucemia/terapia , Animales , Cromatina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Técnicas de Sustitución del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Experimental/genética , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Células Progenitoras Mieloides/patología , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Nucleofosmina , Preleucemia/genética , Preleucemia/patología , Proteínas Proto-Oncogénicas/metabolismo
3.
Oncotarget ; 10(32): 3040-3050, 2019 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-31105884

RESUMEN

DNA methylation can mediate epigenetic silencing of tumor suppressor and cancer protective genes. The protein ubiquitin-like containing PHD and ring finger domains 1 (UHRF1) is an essential component in cells for DNA methylation maintenance. The SET- and RING-associated (SRA) domain of UHRF1 can bind hemimethylated DNA, and mediate recruitment of DNA methyltransferases to copy the methylation pattern to the newly synthesized daughter strand. Loss of UHRF1 function can lead to demethylation and re-expression of epigenetically silenced tumor suppressor genes and can reduce cancer cell growth and survival. We created a high-throughput time-resolved fluorescence resonance energy transfer (TR-FRET) assay to screen for inhibitors capable of disrupting the interaction between the UHRF1-SRA domain and hemimethylated DNA. Using this assay (Z' factor of 0.74 in 384-well format) we screened the Library of Pharmacologically Active Compounds (LOPAC) for UHRF1-SRA inhibitors, and validated 7 hit compounds. These compounds included the anthracycline derivatives idarubicin and mitoxantrone, which are commonly used chemotherapeutic drugs known to mediate cytotoxicity by acting as class II topoisomerase (TOP2) poisons. In a panel of additional known topoisomerase poisons, only the anthracycline derivatives showed dose responsive inhibition of UHRF1-SRA. Additionally, mitoxantrone and doxorubicin showed dose-responsive global DNA demethylation and demonstrated a synergistic growth inhibition of multiple cancer cell lines when combined with the DNA methyltransferase (DNMT) inhibitor decitabine. These data validate a novel TR-FRET assay for identification of UHRF1 inhibitors, and revealed unexpected epigenetic properties of commonly used chemotherapeutic drugs that showed synergistic cytotoxicity of cancer cells when combined with a demethylating agent.

4.
Cancer Chemother Pharmacol ; 83(1): 61-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30327876

RESUMEN

PURPOSE: Obesity may alter mononuclear phagocyte system (MPS) function and the pharmacology and efficacy of nanoparticles therapies, such as PEGylated liposomal doxorubicin (PLD). We aimed to evaluate the relationships between hormone and chemokine mediators of MPS function and the pharmacokinetic (PK) exposure of PLD in obese and normal weight patients with ovarian and endometrial cancer. METHODS: Hormone and chemokine mediators in obese and normal weight ovarian and endometrial cancer patients were measured. A separate pharmacology study was performed that evaluated the relationship between serum hormone concentrations, MPS function, and PK disposition of PLD in refractory ovarian cancer patients. RESULTS: Univariate analysis revealed a significant relationship between serum estradiol and body mass index (OR 8.64, 95% CI 2.67-28.0, p < 0.001). Estrone and testosterone concentrations were positively correlated with MPS function (ρ = 0.57 and 0.53, p = 0.14 and 0.18, respectively) and inversely correlated with PLD PK exposure (ρ = - 0.75 and - 0.76, respectively, p = 0.02 for both). CONCLUSIONS: Higher MPS function resulting in reduced PLD exposure is a potential mechanism for reduced efficacy of PLD and other nanoparticles observed in obese patients with cancer. PK simulations suggest higher doses of PLD are required in obese patients to achieve similar exposures as standard dosing in normal weight patients.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/análogos & derivados , Neoplasias Endometriales/patología , Sistema Mononuclear Fagocítico/fisiopatología , Nanopartículas/administración & dosificación , Obesidad/fisiopatología , Neoplasias Ováricas/patología , Antibióticos Antineoplásicos/química , Estudios de Casos y Controles , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Neoplasias Endometriales/tratamiento farmacológico , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Nanopartículas/química , Neoplasias Ováricas/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Polietilenglicoles/química , Pronóstico
5.
Artículo en Inglés | MEDLINE | ID: mdl-27082761

RESUMEN

The epigenetic and anti-cancer activities of the nucleoside analog DNA methyltransferase (DNMT) inhibitors decitabine (5-aza-2'-deoxycytidine, DAC), azacitidine, and guadecitabine are thought to require cellular uptake, metabolism to 5-aza-2'-deoxycytidine triphosphate, and incorporation into DNA. This genomic incorporation can then lead to trapping and degradation of DNMT enzymes, and ultimately, passive loss of DNA methylation. To facilitate measurement of critical exposure-response relationships of nucleoside analog DNMT inhibitors, a sensitive and reliable method was developed to simultaneously quantitate 5-aza-2'-deoxycytidine genomic incorporation and genomic 5-methylcytosine content using LC-MS/MS. Genomic DNA was extracted and digested into single nucleosides. Chromatographic separation was achieved with a Thermo Hyperpcarb porous graphite column (100mm×2.1mm, 5µm) and isocratic elution with a 10mM ammonium acetate:acetonitrile with 0.1% formic acid (70:30, v/v) mobile phase over a 5min total analytical run time. An AB Sciex 5500 triple quadrupole mass spectrometer operated in positive electrospray ionization mode was used for the detection of 5-aza-2'-deoxycytidine, 2'-deoxycytidine, and 5-methyl-2'-deoxycytidine. The assay range was 2-400ng/mL for 5-aza-2'-deoxycytidine, 50-10,000ng/mL for 2'-deoxycytidine, and was 5-1000ng/mL for 5-methyl-2'-deoxycytidine. The assay proved to be accurate (93.0-102.2%) and precise (CV≤6.3%) across all analytes. All analytes exhibited long-term frozen digest matrix stability at -70°C for at least 117 days. The method was applied for the measurement of genomic 5-aza-2'-deoxycytidine and 5-methyl-2'-deoxycytidine content following exposure of in vitro cell culture and in vivo animal models to decitabine.


Asunto(s)
Azacitidina/análogos & derivados , Cromatografía Liquida/métodos , Metilación de ADN/fisiología , ADN/química , Metiltransferasas/antagonistas & inhibidores , Espectrometría de Masas en Tándem/métodos , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Animales , Azacitidina/análisis , Azacitidina/química , Azacitidina/metabolismo , Línea Celular Tumoral , ADN/análisis , ADN/metabolismo , Decitabina , Humanos , Límite de Detección , Modelos Lineales , Ratones , Ratones Desnudos , Reproducibilidad de los Resultados
6.
Cancer Chemother Pharmacol ; 77(3): 565-73, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26822231

RESUMEN

PURPOSE: Significant variability in the pharmacokinetics and pharmacodynamics of PEGylated liposomal doxorubicin (PLD) exists. PLD undergoes clearance via the mononuclear phagocyte system (MPS). Technetium Tc 99m sulfur colloid (TSC) is approved for imaging MPS cells. We investigated TSC as a phenotypic probe of PLD pharmacokinetics and pharmacodynamics in women with epithelial ovarian cancer. METHODS: TSC 10 mCi IVP was administered and followed by dynamic planar and SPECT/CT imaging and blood pharmacokinetics sampling. PLD 30-40 mg/m(2) IV was administered with or without carboplatin, followed by plasma pharmacokinetics sampling. RESULTS: There was a linear relationship between TSC clearance and encapsulated doxorubicin clearance (R(2) = 0.61, p = 0.02), particularly in patients receiving PLD alone (R(2) = 0.81, p = 0.04). There was a positive relationship (ρ = 0.81, p = 0.01) between maximum grade palmar-plantar erythrodysesthesia toxicity developed and estimated encapsulated doxorubicin concentration in hands. CONCLUSIONS: TSC is a phenotypic probe for PLD pharmacokinetics and pharmacodynamics and may be used to individualize PLD therapy in ovarian cancer and for other nanoparticles in development.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/análogos & derivados , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Radiofármacos/administración & dosificación , Azufre Coloidal Tecnecio Tc 99m/administración & dosificación , Adulto , Anciano , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacocinética , Carboplatino/administración & dosificación , Carcinoma Epitelial de Ovario , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/farmacocinética , Femenino , Síndrome Mano-Pie/etiología , Humanos , Persona de Mediana Edad , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Fenotipo , Polietilenglicoles/administración & dosificación , Polietilenglicoles/efectos adversos , Polietilenglicoles/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada por Rayos X/métodos
7.
J Biomol Screen ; 19(7): 1060-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24608100

RESUMEN

Methylated DNA binding proteins such as Methyl-CpG Binding Domain Protein 2 (MBD2) can transduce DNA methylation alterations into a repressive signal by recruiting transcriptional co-repressor complexes. Interfering with MBD2 could lead to reactivation of tumor suppressor genes and therefore represents an attractive strategy for epigenetic therapy. We developed and compared fluorescence polarization (FP) and time-resolved fluorescence resonance energy transfer (TR-FRET)-based high-throughput screening (HTS) assays to identify small-molecule inhibitors of the interaction between the methyl binding domain of MBD2 (MBD2-MBD) and methylated DNA. Although both assays performed well in 96-well format, the TR-FRET assay (Z' factor = 0.58) emerged as a superior screening strategy compared with FP (Z' factor = 0.08) when evaluated in an HTS 384-well plate format. Using TR-FRET, we screened the Sigma LOPAC library for MBD2-MBD inhibitors and identified four compounds that also validated in a dose-response series. This included two known DNA intercalators (mitoxantrone and idarubicin) among two other inhibitory compounds (NF449 and aurintricarboxylic acid). All four compounds also inhibited the binding of SP-1, a transcription factor with a GC-rich binding sequence, to a methylated oligonucleotide, demonstrating that the activity was nonspecific. Our results provide proof of principle for using TR-FRET-based HTS to identify small-molecule inhibitors of MBD2 and other DNA-protein interactions.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Transferencia Resonante de Energía de Fluorescencia/métodos , Ácido Aurintricarboxílico/química , Bencenosulfonatos/química , ADN/química , Metilación de ADN , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Colorantes Fluorescentes/química , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Idarrubicina/química , Mitoxantrona/química , Oligonucleótidos/química , Unión Proteica , Factor de Transcripción Sp1/química
8.
Proc Natl Acad Sci U S A ; 110(27): 11199-204, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23776246

RESUMEN

Oxaliplatin is an integral component of colorectal cancer therapy, but its clinical use is associated with a dose-limiting peripheral neurotoxicity. We found that the organic cation transporter 2 (OCT2) is expressed on dorsal root ganglia cells within the nervous system where oxaliplatin is known to accumulate. Cellular uptake of oxaliplatin was increased by 16- to 35-fold in cells overexpressing mouse Oct2 or human OCT2, and this process was associated with increased DNA platination and oxaliplatin-induced cytotoxicity. Furthermore, genetic or pharmacologic knockout of Oct2 protected mice from hypersensitivity to cold or mechanical-induced allodynia, which are established tests to assess acute oxaliplatin-induced neurotoxicity. These findings provide a rationale for the development of targeted approaches to mitigate this debilitating toxicity.


Asunto(s)
Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Proteínas de Transporte de Catión Orgánico/fisiología , Compuestos Organoplatinos/toxicidad , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Síndromes de Neurotoxicidad/genética , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Proteínas de Transporte de Catión Orgánico/deficiencia , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico , Compuestos Organoplatinos/farmacocinética , Oxaliplatino
9.
Drug Resist Updat ; 15(1-2): 5-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22459901

RESUMEN

Members of the solute carrier family of transporters are responsible for the cellular uptake of a broad range of endogenous compounds and xenobiotics in multiple tissues. Several of these solute carriers are known to be expressed in cancer cells or cancer cell lines, and decreased cellular uptake of drugs potentially contributes to the development of resistance. As result, the expression levels of these proteins in humans have important consequences for an individual's susceptibility to certain drug-induced side effects, interactions, and treatment efficacy. In this review article, we provide an update of this rapidly emerging field, with specific emphasis on the direct contribution of solute carriers to anticancer drug uptake in tumors, the role of these carriers in regulation of anticancer drug disposition, and recent advances in attempts to evaluate these proteins as therapeutic targets.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Humanos , Proteínas de Transporte de Membrana/genética , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Ratas
10.
Inorg Chem ; 48(4): 1577-87, 2009 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-19146434

RESUMEN

New stable cationic organogold(III) complexes containing the "pincer" iminophosphorane ligand (2-C(6)H(4)-PPh(2)=NPh) have been prepared by reaction of the previously described [Au{kappa(2)-C,N-C(6)H(4)(PPh(2)=N(C(6)H(5))-2}Cl(2)] 1 and a combination of sodium or silver salts and appropriate ligands. The presence of the P atom in the PR(3) fragment has been used as a "spectroscopic marker" to study the in vitro stability (and oxidation state) of the new organogold complexes in solvents like dimethyl sulfoxide and water. Compounds with dithiocarbamato ligands and water-soluble phosphines of the general type [Au{kappa(2)-C,N-C(6)H(4)(PPh(2)=N(C(6)H(5))-2}(S(2)CN-R(2))]PF(6) (R = Me 2; Bz 3) and [Au{kappa(2)-C,N-C(6)H(4)(PPh(2)=N(C(6)H(5))-2}(PR(3))(n)Cl]PF(6) (PR(3) = P{Cp(m-C(6)H(4)-SO(3)Na)(2)} n = 1 4, n = 2 TPA {1,3,5-triaza-7-phosphaadamantane} 5) have been synthesized and characterized in solution and in the solid state (the crystal structure of 2 has been determined by X-ray diffraction studies). Complexes 1-5 have been tested as potential anticancer agents, and their cytotoxicity properties were evaluated in vitro against HeLa human cervical carcinoma and Jurkat-T acute lymphoblastic leukemia cells. Compounds 2 and 3 are quite cytotoxic for these two cell lines. There is a preferential induction of apoptosis in HeLa cells after treatment with 1-5. However in the case of the more cytotoxic complex (2), cell death is activated because of both apoptosis and necrosis. The interactions of 1-5 with Calf Thymus DNA have been evaluated by Thermal Denaturation methods. All these complexes show no or little (electrostatic) interaction with DNA. The interaction of 2 with two model proteins (cytochrome c and thioredoxin reductase) has been analyzed by spectroscopic methods (vis-UV and fluorescence). Compound 2 manifests a high reactivity toward both proteins. The mechanistic implications of these results are discussed here.


Asunto(s)
Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Compuestos Orgánicos de Oro/síntesis química , Compuestos Orgánicos de Oro/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Citocromos c/efectos de los fármacos , Estabilidad de Medicamentos , Humanos , Ligandos , Estructura Molecular , Fosforanos/química , Solubilidad , Reductasa de Tiorredoxina-Disulfuro/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...