Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Bioorg Med Chem Lett ; 76: 129009, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36174836

RESUMEN

Strategically replacing hydrogen with deuterium at sites of metabolism in small molecule drugs can significantly alter clearance and potentially enhance clinical safety. Bupropion is an antidepressant and smoking cessation medication with the potential to cause seizures. We hypothesized that incorporating deuterium at specific sites in bupropion may greatly reduce epimerization, potentially slow metabolism, and reduce the formation of toxic metabolites, namely hydroxybupropion which has been associated with bupropion's toxicity. Four deuterated analogues were synthesized incorporating deuterium at sites of metabolism and epimerization with the aim of altering the metabolic profile of bupropion. Spectroscopic binding and metabolism studies with bupropion and R-or S-d4 and R-or S-d10 analogs were performed with recombinant CYP2B6, human liver microsomes, and human hepatocytes. Results demonstrate that deuterated bupropion analogues exhibited 20-25% decrease in racemization and displayed a significant decrease in the formation of CYP2B6-mediated R,R - or S,S-hydroxybupropion with recombinant protein and human liver microsomes. In primary human hepatocytes, metabolism of deuterated analogs to R,R - and S,S-hydroxybupropion and threo- and erythro-hydrobupropion was significantly less than R/S-d0 bupropion. Selective deuterium substitution at metabolic soft spots in bupropion has the potential to provide a drug with a simplified pharmacokinetic profile, reduced toxicity and improved tolerability in patients.


Asunto(s)
Bupropión , Humanos , Bupropión/farmacología , Bupropión/metabolismo , Citocromo P-450 CYP2B6 , Deuterio , Proteínas Recombinantes
2.
J Pharmacol Exp Ther ; 362(2): 359-367, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28611092

RESUMEN

Ivacaftor is currently used for the treatment of cystic fibrosis as both monotherapy (Kalydeco; Vertex Pharmaceuticals, Boston, MA) and combination therapy with lumacaftor (Orkambi; Vertex Pharmaceuticals). Each therapy targets specific patient populations: Kalydeco treats patients carrying one of nine gating mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) protein, whereas Orkambi treats patients homozygous for the F508del CFTR mutation. In this study, we explored the pharmacological and metabolic effects of precision deuteration chemistry on ivacaftor by synthesizing two novel deuterated ivacaftor analogs, CTP-656 (d9-ivacaftor) and d18-ivacaftor. Ivacaftor is administered twice daily and is extensively converted in humans to major metabolites M1 and M6; therefore, the corresponding deuterated metabolites were also prepared. Both CTP-656 and d18-ivacaftor showed in vitro pharmacologic potency similar to that in ivacaftor, and the deuterated M1 and M6 metabolites showed pharmacology equivalent to that in the corresponding metabolites of ivacaftor, which is consistent with the findings of previous studies of deuterated compounds. However, CTP-656 exhibited markedly enhanced stability when tested in vitro. The deuterium isotope effects for CTP-656 metabolism (DV = 3.8, DV/K = 2.2) were notably large for a cytochrome P450-mediated oxidation. The pharmacokinetic (PK) profile of CTP-656 and d18-ivacaftor were assessed in six healthy volunteers in a single-dose crossover study, which provided the basis for advancing CTP-656 in development. The overall PK profile, including the 15.9-hour half-life for CTP-656, suggests that CTP-656 may be dosed once daily, thereby enhancing patient adherence. Together, these data continue to validate deuterium substitution as a viable approach for creating novel therapeutic agents with properties potentially differentiated from existing drugs.


Asunto(s)
Aminofenoles/administración & dosificación , Aminofenoles/farmacocinética , Deuterio/administración & dosificación , Deuterio/farmacocinética , Metaboloma/efectos de los fármacos , Quinolonas/administración & dosificación , Quinolonas/farmacocinética , Administración Oral , Aminofenoles/química , Animales , Estudios Cruzados , Deuterio/química , Perros , Descubrimiento de Drogas , Femenino , Humanos , Masculino , Metaboloma/fisiología , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Quinolonas/química , Ratas , Ratas Sprague-Dawley
3.
Drug Metab Dispos ; 44(10): 1584-97, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27450182

RESUMEN

Metabolic activation of the dual-tyrosine kinase inhibitor lapatinib by cytochromes CYP3A4 and CYP3A5 has been implicated in lapatinib-induced idiosyncratic hepatotoxicity; however, the relative enzyme contributions have not been established. The objective of this study was to examine the roles of CYP3A4 and CYP3A5 in lapatinib bioactivation leading to a reactive, potentially toxic quinoneimine. Reaction phenotyping experiments were performed using individual human recombinant P450 enzymes and P450-selective chemical inhibitors. Lapatinib metabolites and quinoneimine-glutathione (GSH) adducts were analyzed using liquid chromatography-tandem mass spectrometry. A screen of cDNA-expressed P450s confirmed that CYP3A4 and CYP3A5 are the primary enzymes responsible for quinoneimine-GSH adduct formation using lapatinib or O-dealkylated lapatinib as the substrate. The mean kinetic parameters (Km and kcat) of lapatinib O-dealkylation revealed that CYP3A4 was 5.2-fold more efficient than CYP3A5 at lapatinib O-dealkylation (CYP3A4 kcat/Km = 6.8 µM(-1) min(-1) versus CYP3A5 kcat/Km = 1.3 µM(-1) min(-1)). Kinetic analysis of GSH adduct formation indicated that CYP3A4 was also 4-fold more efficient at quinoneimine-GSH adduct formation as measured by kcat (maximum relative GSH adduct levels)/Km (CYP3A4 = 0.0082 vs. CYP3A5 = 0.0021). In human liver microsomal (HLM) incubations, CYP3A4-selective inhibitors SR-9186 and CYP3cide reduced formation of GSH adducts by 78% and 72%, respectively, compared with >90% inhibition by the pan-CYP3A inhibitor ketoconazole. The 16%-22% difference between CYP3A- and CYP3A4-selective inhibition indicates the involvement of remaining CYP3A5 activity in generating reactive metabolites from lapatinib in pooled HLMs. Collectively, these findings support the conclusion that both CYP3A4 and CYP3A5 are quantitatively important contributors to lapatinib bioactivation.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Quinazolinas/farmacocinética , Alquilación , Biocatálisis , Cromatografía Liquida , Citocromo P-450 CYP3A/efectos de los fármacos , Inhibidores del Citocromo P-450 CYP3A/farmacología , Glutatión/metabolismo , Humanos , Lapatinib , Espectrometría de Masas en Tándem
4.
J Pharmacol Exp Ther ; 354(1): 43-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25943764

RESUMEN

Selective deuterium substitution as a means of ameliorating clinically relevant pharmacokinetic drug interactions is demonstrated in this study. Carbon-deuterium bonds are more stable than corresponding carbon-hydrogen bonds. Using a precision deuteration platform, the two hydrogen atoms at the methylenedioxy carbon of paroxetine were substituted with deuterium. The new chemical entity, CTP-347 [(3S,4R)-3-((2,2-dideuterobenzo[d][1,3]dioxol-5-yloxy)methyl)-4-(4-fluorophenyl)piperidine], demonstrated similar selectivity for the serotonin receptor, as well as similar neurotransmitter uptake inhibition in an in vitro rat synaptosome model, as unmodified paroxetine. However, human liver microsomes cleared CTP-347 faster than paroxetine as a result of decreased inactivation of CYP2D6. In phase 1 studies, CTP-347 was metabolized more rapidly in humans and exhibited a lower pharmacokinetic accumulation index than paroxetine. These alterations in the metabolism profile resulted in significantly reduced drug-drug interactions between CTP-347 and two other CYP2D6-metabolized drugs: tamoxifen (in vitro) and dextromethorphan (in humans). Our results show that precision deuteration can improve the metabolism profiles of existing pharmacotherapies without affecting their intrinsic pharmacologies.


Asunto(s)
Inhibidores del Citocromo P-450 CYP2D6/farmacología , Citocromo P-450 CYP2D6/metabolismo , Paroxetina/farmacología , Animales , Encéfalo/metabolismo , Inhibidores del Citocromo P-450 CYP2D6/farmacocinética , Deuterio , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Humanos , Marcaje Isotópico , Microsomas Hepáticos/metabolismo , Paroxetina/farmacocinética , Paroxetina/uso terapéutico , Ensayo de Unión Radioligante , Ratas , Receptores de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sinaptosomas/metabolismo , Tamoxifeno/metabolismo
5.
Drug Metab Dispos ; 39(4): 693-702, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21177487

RESUMEN

Oxymetazoline (6-tert-butyl-3-(2-imidazolin-2-ylmethyl)-2,4-dimethylphenol) has been widely used as a nonprescription nasal vasoconstrictor for >40 years; however, its metabolic pathway has not been investigated. This study describes the in vitro metabolism of oxymetazoline in human, rat, and rabbit liver postmitochondrial supernatant fraction from homogenized tissue (S9) fractions and their microsomes supplemented with NADPH. The metabolites of oxymetazoline identified by liquid chromatography (LC)/UV/tandem mass spectrometry (MS/MS), included M1 (monohydroxylation of the t-butyl group), M2 (oxidative dehydrogenation of the imidazoline to an imidazole moiety), M3 (monohydroxylation of M2), M4 (dihydroxylation of oxymetazoline), and M5 (dihydroxylation of M2). Screening with nine human expressed cytochromes P450 (P450s) identified CYP2C19 as the single P450 isoform catalyzing the formation of M1, M2, and M3. Glutathione conjugates of oxymetazoline (M6) and M2 (M7) were identified in the liver S9 fractions, indicating the capability of oxymetazoline to undergo bioactivation to reactive intermediate species. M6 and M7 were not detected in those liver S9 incubations without NADPH. Cysteine conjugates (M8 and M9) derived from glutathione conjugates and hydroxylated glutathione conjugates (M10 and M11) were also identified. The reactive intermediate of oxymetazoline was trapped with glutathione and N-acetyl cysteine and identified by LC/MS/MS. M6 was isolated and identified by one-dimensional or two-dimensional NMR as the glutathione conjugate of a p-quinone methide. We have shown the tendency of oxymetazoline to form p-quinone methide species via a bioactivation mechanism involving a CYP2C19-catalyzed two-electron oxidation. Nevertheless, we conclude that the formation of this reactive species might not be a safety concern for oxymetazoline nasal products because of the typical low-dose and brief dosage regimen limited to nasal delivery.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Glutatión/metabolismo , Oximetazolina/metabolismo , Simpatomiméticos/metabolismo , Acetilcisteína/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Citocromo P-450 CYP2C19 , Humanos , Hidroxilación , Técnicas In Vitro , Indolquinonas/metabolismo , Hígado/metabolismo , Masculino , Microsomas Hepáticos/metabolismo , NADP/metabolismo , Oxidación-Reducción , Oximetazolina/química , Conejos , Ratas , Simpatomiméticos/química
6.
J Med Chem ; 49(9): 2669-72, 2006 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-16640325

RESUMEN

Activation of CCR8 by its ligand CCL1 may play an important role in diseases such as asthma, multiple sclerosis, and cancer. The study of small molecule CCR8 antagonists will help establish the validation of these hypotheses. We report the design, synthesis, and progress toward optimization of potent small molecule CCR8 antagonists identified from a high-throughput screen. These analogues exhibit good potency in binding and chemotaxis assays, show good selectivity versus the hERG channel, and have good eADME (early absorption, distribution, metabolism, and excretion) profiles.


Asunto(s)
Diseño de Fármacos , Receptores de Quimiocina/antagonistas & inhibidores , Aminación , Línea Celular , Quimiotaxis/efectos de los fármacos , Éter/química , Humanos , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Piperidinas/farmacología , Pirrolidinas/síntesis química , Pirrolidinas/química , Pirrolidinas/farmacología , Receptores CCR8 , Relación Estructura-Actividad
7.
J Neurotrauma ; 32(20): 1621-32, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25794265

RESUMEN

Post-traumatic seizures can exacerbate injurious outcomes of severe brain trauma, yet effective treatments are limited owing to the complexity of the pathology underlying the concomitant occurrence of both events. In this study, we tested C-10068, a novel deuterium-containing analog of (+)-N-methyl-3-ethoxymorphinan, in a rat model of penetrating ballistic-like brain injury (PBBI) and evaluated the effects of C-10068 on PBBI-induced nonconvulsive seizures (NCS), acute neuroinflammation, and neurofunctional outcomes. NCS were detected by electroencephalographic monitoring. Neuroinflammation was evaluated by immunohistochemical markers, for example, glial fibrillary acidic protein and major histocompatibility complex class I, for activation of astrocytes and microglia, respectively. Neurofunction was tested using rotarod and Morris water maze tasks. Three infusion doses of C-10068 (1.0, 2.5, and 5.0 mg/kg/h × 72 h) were tested in the antiseizure study. Neuroinflammation and neurofunction were evaluated in animals treated with 5.0 mg/kg/h × 72 h C-10068. Compared to vehicle treatment, C-10068 dose dependently reduced PBBI-induced NCS incidence (40-50%), frequency (20-70%), and duration (30-82%). The most effective antiseizure dose of C-10068 (5.0 mg/kg/h × 72 h) also significantly attenuated hippocampal astrocyte activation and perilesional microglial reactivity post-PBBI. Within C-10068-treated animals, a positive correlation was observed in reduction in NCS frequency and reduction in hippocampal astrocyte activation. Further, C-10068 treatment significantly attenuated astrocyte activation in seizure-free animals. However, C-10068 failed to improve PBBI-induced motor and cognitive functions with the dosing regimen used in this study. Overall, the results indicating that C-10068 exerts both potent antiseizure and antiinflammatory effects are promising and warrant further investigation.


Asunto(s)
Antiinflamatorios , Anticonvulsivantes , Dextrometorfano , Antagonistas de Aminoácidos Excitadores , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Dextrometorfano/administración & dosificación , Dextrometorfano/análogos & derivados , Dextrometorfano/farmacología , Modelos Animales de Enfermedad , Electroencefalografía , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/farmacología , Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/inmunología , Hipocampo/efectos de los fármacos , Inflamación/etiología , Masculino , Microglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Convulsiones/etiología
8.
J Pharm Sci ; 100(2): 784-93, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20669329

RESUMEN

The incubation of oxymetazoline, a nonprescription nasal decongestant, with human liver microsomes (HLMs) supplemented with uridine-5-diphosphoglucuronic acid (UDPGA) generated glucuronide metabolite as observed by LC/MS/MS. The uridine glucuronosyltransferases (UGTs) responsible for the O-glucuronidation of oxymetazoline remain thus far unidentified. The glucuronide formed in HLMs was identified by LC/MS/MS and characterized by one- and two-dimensional NMR to be the ß-O-glucuronide of oxymetazoline. UGT screening with expressed UGTs identified UGT1A9 as the single UGT isoform catalyzing O-glucuronidation of oxymetazoline. Oxymetazoline O-glucuronidation by using HLMs was best fitted to the allosteric sigmoidal model. The derived S(50) and V(max) values were 2.42 ± 0.40 mM and 8.69 ± 0.58 pmole/(min mg of protein), respectively, and maximum clearance (CL(max)) was 3.61 L/min/mg. Oxymetazoline O-glucuronidation by using expressed UGT1A9 was best fitted to the substrate inhibition model. The derived K(m) and V(max) values were 2.53 ± 1.03 mM and 54.18 ± 16.92 pmole/(min mg of protein), respectively, and intrinsic clearance (CL(int)) was 21.41 L/(min mg). Our studies indicate that oxymetazoline is not glucuronidated at its nanomolar intranasal dose and thus is eliminated unchanged, because UGT1A9 would only contribute to its elimination at the toxic plasma concentrations.


Asunto(s)
Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Microsomas Hepáticos/metabolismo , Descongestionantes Nasales/metabolismo , Oximetazolina/metabolismo , Uridina Difosfato Ácido Glucurónico/metabolismo , Cromatografía Liquida , Humanos , Cinética , Microsomas Hepáticos/enzimología , Isoformas de Proteínas/metabolismo , Espectrometría de Masas en Tándem , UDP Glucuronosiltransferasa 1A9
9.
J Med Chem ; 54(13): 4752-72, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21650221

RESUMEN

Histone deacetylase (HDAC) inhibitors have shown promise in treating various forms of cancer. However, many HDAC inhibitors from diverse structural classes have been associated with QT prolongation in humans. Inhibition of the human ether a-go-go related gene (hERG) channel has been associated with QT prolongation and fatal arrhythmias. To determine if the observed cardiac effects of HDAC inhibitors in humans is due to hERG blockade, a highly potent HDAC inhibitor devoid of hERG activity was required. Starting with dacinostat (LAQ824), a highly potent HDAC inhibitor, we explored the SAR to determine the pharmacophores required for HDAC and hERG inhibition. We disclose here the results of these efforts where a high degree of pharmacophore homology between these two targets was discovered. This similarity prevented traditional strategies for mitigating hERG binding/modulation from being successful and novel approaches for reducing hERG inhibition were required. Using a hERG homology model, two compounds, 11r and 25i, were discovered to be highly efficacious with weak affinity for the hERG and other ion channels.


Asunto(s)
Acrilamidas/toxicidad , Antineoplásicos/toxicidad , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/toxicidad , Ácidos Hidroxámicos/toxicidad , Acrilamidas/síntesis química , Acrilamidas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Canal de Potasio ERG1 , Células HCT116 , Semivida , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Técnicas In Vitro , Ratones , Ratones Desnudos , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Trasplante de Neoplasias , Técnicas de Placa-Clamp , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular , Trasplante Heterólogo
10.
Drug Metab Dispos ; 34(9): 1600-5, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16790553

RESUMEN

Apparent intrinsic clearance (CL(int,app)) of 7-ethoxycoumarin, phenacetin, propranolol, and midazolam was measured using rat and human liver microsomes and freshly isolated and cryopreserved hepatocytes to determine factors responsible for differences in rates of metabolism in these systems. The cryopreserved and freshly isolated hepatocytes generally provided similar results, although there was greater variability using the latter system. The CL(int,app) values in hepatocytes are observed to be lower than that in microsomes, and this difference becomes greater for compounds with high CL(int,app). This could partly be attributed to the differences in the free fraction (fu). The fu in hepatocyte incubations (fu,hep-inc) was influenced not only by the free fraction of compounds in the incubation buffer (fu,buffer) but also by the rate constants of uptake (k(up)) and metabolism (k(met)). This report provides a new derivation for fu,hep-inc, which can be expressed as fu,hep-inc = [k(up)/(k(met) + k(up))]/[1 + (C(hep)/C(buffer)) x (V(hep)/V(buffer))], where the C(hep), C(buffer), V(hep), and V(buffer) represent the concentrations of a compound in hepatocytes and buffer and volumes of hepatocytes and buffer, respectively. For midazolam, the fu,hep-inc was calculated, and the maximum metabolism rate in hepatocytes was shown to be limited by the uptake rate.


Asunto(s)
Evaluación Preclínica de Medicamentos , Hepatocitos/metabolismo , Microsomas Hepáticos/metabolismo , Animales , Transporte Biológico , Cumarinas/metabolismo , Criopreservación , Difusión , Evaluación Preclínica de Medicamentos/métodos , Humanos , Técnicas In Vitro , Cinética , Tasa de Depuración Metabólica , Midazolam/metabolismo , Fenacetina/metabolismo , Propranolol/metabolismo , Ratas , Reproducibilidad de los Resultados
11.
Drug Metab Dispos ; 34(4): 702-8, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16443666

RESUMEN

Bortezomib (Velcade, PS-341), a dipeptidyl boronic acid, is a first-in-class proteasome inhibitor approved in 2003 for the treatment of multiple myeloma. In a preclinical toxicology study, bortezomib-treated rats resulted in liver enlargement (35%). Ex vivo analyses of the liver samples showed an 18% decrease in cytochrome P450 (P450) content, a 60% increase in palmitoyl coenzyme A beta-oxidation activity, and a 41 and 23% decrease in CYP3A protein expression and activity, respectively. Furthermore, liver samples of bortezomib-treated rats had little change in CYP2B and CYP4A protein levels and activities. To address the likelihood of clinical drug-drug interactions, the P450 inhibition potential of bortezomib and its major deboronated metabolites M1 and M2 and their dealkylated metabolites M3 and M4 was evaluated in human liver microsomes for the major P450 isoforms 1A2, 2C9, 2C19, 2D6, and 3A4/5. Bortezomib, M1, and M2 were found to be mild inhibitors of CYP2C19 (IC(50) approximately 18.0, 10.0, and 13.2 microM, respectively), and M1 was also a mild inhibitor of CYP2C9 (IC(50) approximately 11.5 microM). However, bortezomib, M1, M2, M3, and M4 did not inhibit other P450s (IC(50) values > 30 microM). There also was no time-dependent inhibition of CYP3A4/5 by bortezomib or its major metabolites. Based on these results, no major P450-mediated clinical drug-drug interactions are anticipated for bortezomib or its major metabolites. To our knowledge, this is the first report on P450-mediated drug-drug interaction potential of proteasome inhibitors or boronic acid containing therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Microsomas Hepáticos/enzimología , Pirazinas/farmacología , Animales , Antineoplásicos/metabolismo , Bortezomib , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Cinética , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley
12.
Drug Metab Dispos ; 33(11): 1723-8, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16103134

RESUMEN

VELCADE (bortezomib, PS-341), reversibly inhibits the 20S proteasome and exhibits cytotoxic and antitumor activities. Pretreatment of cancer cells with bortezomib increases the chemosensitivity of these cells, suggesting that bortezomib may be used in combination chemotherapy. The relative contributions of the five major human cytochromes P450 (P450s), 1A2, 2C9, 2C19, 2D6, and 3A4 (the focus of the present study), to the metabolism of bortezomib are an important aspect of potential drug interactions. Relative activity factor (RAF), chemical inhibition, and immunoinhibition using monoclonal antibodies were three approaches employed to determine the relative contributions of the major human P450s to the net hepatic metabolism of bortezomib. RAFs for the P450 isoform-selective substrates were determined; the ratio of the rate of metabolism of bortezomib with cDNA-expressed P450s versus rate of metabolism with human liver microsomes was normalized with respect to the RAF for each P450 isoform to determine the percentage contributions of the P450s to the net hepatic metabolism of bortezomib. CYP3A4 followed by CYP2C19 were determined to be the major contributors to the metabolism of bortezomib. Chemical inhibition and immunoinhibition confirmed that CYP3A4 and CYP2C19 were the major P450s responsible for the hepatic metabolism of bortezomib. The studies were conducted with 2 muM bortezomib, and the disappearance of bortezomib, rather than appearance of a specific metabolite, was quantified to determine the contributions of the P450s to the overall hepatic metabolism of bortezomib in humans.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Ácidos Borónicos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/metabolismo , Oxigenasas de Función Mixta/metabolismo , Inhibidores de Proteasas/metabolismo , Pirazinas/metabolismo , Anticuerpos Monoclonales/farmacología , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/inmunología , Bortezomib , Citocromo P-450 CYP1A2/inmunología , Citocromo P-450 CYP1A2/metabolismo , Inhibidores del Citocromo P-450 CYP1A2 , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/inmunología , Citocromo P-450 CYP2D6/metabolismo , Inhibidores del Citocromo P-450 CYP2D6 , Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/inmunología , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/inmunología , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA