Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Sep Sci ; 44(21): 3914-3923, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34463059

RESUMEN

An intelligent chemometric second-order calibration method called alternating trilinear decomposition- assisted multivariate curve resolution combined with high-performance liquid chromatography-diode array detection was used for the simultaneous quantification of nine tyrosine kinase inhibitors in three complex biological systems. The method allows simultaneous quantification of the components in different biological matrices without the need for cumbersome pre-treatment steps, complex elution conditions, and complete peak separation. Even with the varying time shift, severe peak overlap, and various unknown interferences, the proposed method can extract pure chromatographic and spectroscopic information for each analyte, while providing accurate qualitative and quantitative results of nine common tyrosine kinase inhibitors in three different biological matrices. All the drugs were eluted in 7 min. The results showed that the nine drugs in each matrix showed good linearity (r > 0.984) in the calibration range with a root mean square error of calibration less than 0.9 µg/mL. The average spiked recoveries of the target analytes were all in the range of 83.4-110.0%, with standard deviations less than 9.0%. Finally, the classical method was used to validate the proposed method. In comparison to the traditional method, the proposed strategy is accuracy, simultaneous, and interference-free.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Inhibidores de Proteínas Quinasas , Calibración , Quimiometría , Humanos , Límite de Detección , Modelos Lineales , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/orina , Reproducibilidad de los Resultados
2.
Invest New Drugs ; 39(3): 803-811, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33462752

RESUMEN

BACKGROUND: Entrectinib is an oral, CNS-active, potent inhibitor of tyrosine receptor kinases A/B/C, tyrosine kinase ROS proto-oncogene 1, and anaplastic lymphoma kinase approved for use in patients with solid tumors. We describe 3 clinical studies, including one investigating the single/multiple dose pharmacokinetics of entrectinib in patients and two studies in healthy volunteers investigating the absorption/distribution/metabolism/excretion (ADME) of entrectinib, its relative bioavailability, and effect of food on pharmacokinetics. METHODS: The patient study is open-label with dose-escalation and expansion phases. Volunteers received entrectinib (100-400 mg/m2, and 600-800 mg) once daily with food in continuous 28-day cycles. In the ADME study, volunteers received a single oral dose of [14C]entrectinib 600 mg. In the third study, volunteers received single doses of entrectinib 600 mg as the research and marketed formulations in the fasted state (Part 1), and the marketed formulation in the fed and fasted states (Part 2). Entrectinib and its major active metabolite M5 were assessed in all studies. RESULTS: Entrectinib was absorbed in a dose-dependent manner with maximum concentrations at ~4 h postdose and an elimination half-life of ~20 h. Entrectinib was cleared mainly through metabolism and both entrectinib and metabolites were eliminated mainly in feces (minimal renal excretion). At steady-state, the M5-to-entrectinib AUC ratio was 0.5 (with 600 mg entrectinib research formulation in patients). The research and marketed formulations were bioequivalent and food had no relevant effect on pharmacokinetics. CONCLUSIONS: Entrectinib is well absorbed, with linear PK that is suitable for once-daily dosing, and can be taken with or without food.


Asunto(s)
Antineoplásicos/farmacocinética , Benzamidas/farmacocinética , Indazoles/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Antineoplásicos/orina , Benzamidas/administración & dosificación , Benzamidas/sangre , Benzamidas/orina , Cápsulas , Estudios Cruzados , Ayuno/metabolismo , Heces/química , Femenino , Interacciones Alimento-Droga , Voluntarios Sanos , Humanos , Indazoles/administración & dosificación , Indazoles/sangre , Indazoles/orina , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Equivalencia Terapéutica , Adulto Joven
3.
Curr Drug Metab ; 21(5): 357-367, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407271

RESUMEN

BACKGROUND: Surufatinib is a potent small-molecule tyrosine kinase inhibitor and exhibited significant efficacy in the treatment of neuroendocrine tumors in clinical trials. OBJECTIVE: The absorption, metabolism and excretion of surufatinib were investigated in rats and human volunteers following a single oral dose of [14C] surufatinib. METHODS: The radioactivity was measured in plasma, urine, feces and bile by liquid scintillation counting, and the metabolites were characterized by liquid chromatography coupled to mass spectrometry. RESULTS: Surufatinib was orally absorbed similarly in rats and human volunteers, with the median Tmax of 4 hours post-dose. The estimated t1/2 appeared longer in humans than in rats (mean t1/2: 3.12 hour for male rats, 6.48 hours for female rats and 23.3 hours for male human volunteers). The excretion of surufatinib was almost complete in rats and human volunteers in the studies, with the total radioactivity recovery of >90% of the dose. Similarly, in rats and humans, fecal excretion predominated (approximately 87% of the dose recovered in feces and only 5% in urine). The parent drug was the major radioactive component detected in the plasma extracts of rats and humans, and no single circulating metabolite accounted for >10% of the total radioactivity. Unchanged drug was a minor radioactive component in the excreta of rats and humans. CONCLUSION: Fecal excretion was the predominant way for the elimination of surufatinib and its metabolites in rats and humans. No disproportionate circulating metabolite was observed in humans.


Asunto(s)
Antineoplásicos/farmacocinética , Indoles/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Administración Oral , Adulto , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/orina , Bilis/metabolismo , Heces/química , Femenino , Humanos , Indoles/efectos adversos , Indoles/orina , Absorción Intestinal , Masculino , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/orina , Pirimidinas/efectos adversos , Pirimidinas/orina , Ratas Sprague-Dawley , Sulfonamidas/efectos adversos , Sulfonamidas/orina
4.
Spectrochim Acta A Mol Biomol Spectrosc ; 238: 118438, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32388416

RESUMEN

A highly selective and simple micellar synchronous spectrofluorimetric method was described for simultaneous analysis of two tyrosine kinase inhibitors (TKIs); namely lapatinib (LPB) and foretinib (FTB) in human urine. The method depended on measuring synchronous fluorescence of the two drugs in micellar media composed of cremophor RH 40 (Cr RH 40) surfactant using feed-forward and cascade-forward neural networks preceded by genetic algorithm for data manipulation. Different experimental conditions that affect fluorescence of the cited drugs are optimized including pH, diluting solvent, surfactant's type and concentration. A training set of nine mixtures containing different concentrations of both drugs was prepared for models' construction. Extra validation set composed of other nine mixtures was prepared to validate prediction performance for the constructed models. Root mean square error of prediction (RMSEP) was used as a tool to compare prediction power of each model. The method was extended for quantification of LPB and FTB in spiked human urine.


Asunto(s)
Anilidas/orina , Lapatinib/orina , Inhibidores de Proteínas Quinasas/orina , Quinolinas/orina , Humanos , Límite de Detección , Micelas , Redes Neurales de la Computación , Espectrometría de Fluorescencia/métodos
5.
Biopharm Drug Dispos ; 41(4-5): 206-220, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32383777

RESUMEN

Fasudil hydrochloride as an intracellular calcium ion antagonist that dilates blood vessels has exhibited a very potent pharmacological effect in the treatment of angina pectoris. The purpose of this study was to determine the absorption, distribution, and excretion profiles of fasudil in rats and beagle dogs, respectively, to clarify its pharmacokinetic pattern. A sensitive and reliable LC-MS/MS method has been developed and established and successfully applied to pharmacokinetic study, including absorption, tissue distribution, and excretion. The results revealed that in the range of 2-6 mg/kg, the pharmacokinetic behavior for instance, AUC and Cmax , in rats was observed in a dose dependent manner. However, the plasma concentrations were indicative of a significant gender difference in the pharmacokinetics of fasudil in rats, in terms of absolute bioavailability and excretion. Interestingly, the resulting data obtained from beagle dogs showed that there was no gender difference in the absolute bioavailability of fasudil hydrochloride after single or repeated administrations. In conclusion, this study characterized the pharmacokinetic pattern fasudil both in rats and beagle dogs through absorption, tissue distribution and excretion study. The findings may be valuable and provide a rationale for further study and its safe use in clinical practice.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Inhibidores de Proteínas Quinasas/farmacocinética , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/sangre , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacocinética , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/orina , Animales , Área Bajo la Curva , Bilis/química , Cromatografía Liquida , Perros , Heces/química , Femenino , Masculino , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Ratas Wistar , Espectrometría de Masas en Tándem , Distribución Tisular
6.
Invest New Drugs ; 38(5): 1507-1519, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32221754

RESUMEN

Tepotinib (MSC2156119J) is an oral, potent, highly selective MET inhibitor. This open-label, phase I study in healthy volunteers (EudraCT 2013-003226-86) investigated its mass balance (part A) and absolute bioavailability (part B). In part A, six participants received tepotinib orally (498 mg spiked with 2.67 MBq [14C]-tepotinib). Blood, plasma, urine, and feces were collected up to day 25 or until excretion of radioactivity was <1% of the administered dose. In part B, six participants received 500 mg tepotinib orally as a film-coated tablet, followed by an intravenous [14C]-tepotinib tracer dose (53-54 kBq) 4 h later. Blood samples were collected until day 14. In part A, a median of 92.5% (range, 87.1-96.9%) of the [14C]-tepotinib dose was recovered in excreta. Radioactivity was mainly excreted via feces (median, 78.7%; range, 69.4-82.5%). Urinary excretion was a minor route of elimination (median, 14.4% [8.8-17.7%]). Parent compound was the main constituent in excreta (45% [feces] and 7% [urine] of the radioactive dose). M506 was the only major metabolite. In part B, absolute bioavailability was 72% (range, 62-81%) after oral administration of 500 mg tablets (the dose and formulation used in phase II trials). In conclusion, tepotinib and its metabolites are mainly excreted via feces; parent drug is the major eliminated constituent. Oral bioavailability of tepotinib is high, supporting the use of the current tablet formulation in clinical trials. Tepotinib was well tolerated in this study with healthy volunteers.


Asunto(s)
Antineoplásicos/farmacocinética , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridazinas/farmacocinética , Pirimidinas/farmacocinética , Administración Oral , Adulto , Antineoplásicos/sangre , Antineoplásicos/orina , Disponibilidad Biológica , Heces/química , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/sangre , Piperidinas/orina , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Piridazinas/sangre , Piridazinas/orina , Pirimidinas/sangre , Pirimidinas/orina , Adulto Joven
7.
Eur J Drug Metab Pharmacokinet ; 45(1): 101-111, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31673875

RESUMEN

BACKGROUND AND OBJECTIVES: Erdafitinib, an oral selective pan-fibroblast growth factor receptor (FGFR) kinase inhibitor, is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. The aim of this phase 1 study was to assess the pharmacokinetics and safety of erdafitinib in healthy participants when coadministered with fluconazole (moderate CYP2C9 and CYP3A inhibitor), and itraconazole (a strong CYP3A4 and P-glycoprotein inhibitor). The effect of CYP2C9 genotype variants (*1/*1, *1/*2, *1/*3) on the pharmacokinetics of erdafitinib was also investigated. METHODS: In this open-label, parallel-group, single-center study, eligible healthy adults were randomized by CYP2C9 genotype to receive Treatment A (single oral dose of erdafitinib 4 mg) on day 1, Treatment B (fluconazole 400 mg/day orally) on days 1-11, or Treatment C (itraconazole 200 mg/day orally) on days 1-11. Healthy adults randomized to Treatment B and C received a single oral 4-mg dose of erdafitinib on day 5. The pharmacokinetic parameters, including mean maximum plasma concentration (Cmax), area under the curve (AUC) from time 0 to 168 h (AUC168h), AUC from time 0 to the last quantifiable concentration (AUClast), and AUC from time 0 to infinity (AUC∞) were calculated from individual plasma concentration-time data using standard non-compartmental methods. RESULTS: Coadministration of erdafitinib with fluconazole increased Cmax of erdafitinib by approximately 21%, AUC168h by 38%, AUClast by 49%, and AUC∞ by 48% while coadministration with itraconazole resulted in no change in erdafitinib Cmax and increased AUC168h by 20%, AUClast by 33% and AUC∞ by 34%. Erdafitinib exposure was comparable between participants with CYP2C9 *1/*2 or *1/*3 and with wild-type CYP2C9 genotype. The ratio of total amount of erdafitinib excreted in the urine (inhibited to non-inhibited) was 1.09, the ratio of total amount of excreted metabolite M6 was 1.21, and the ratio of the metabolite to parent ratio in the urine was 1.11, when coadministration of erdafitinib with itraconazole was compared with single-dose erdafitinib. Treatment-emergent adverse events (TEAEs) were generally Grade 1 or 2 in severity; the most commonly reported TEAE was headache. No safety concerns were identified with single-dose erdafitinib when administered alone and in combination with fluconazole or itraconazole in healthy adults. CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. TRIAL REGISTRATION: ClinicalTrials.gov identifier number: NCT03135106.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Interacciones Farmacológicas , Fluconazol/farmacología , Itraconazol/farmacología , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Quinoxalinas/farmacocinética , Adulto , Área Bajo la Curva , Citocromo P-450 CYP2C9/genética , Combinación de Medicamentos , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Pirazoles/efectos adversos , Pirazoles/sangre , Pirazoles/orina , Quinoxalinas/efectos adversos , Quinoxalinas/sangre , Quinoxalinas/orina , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores
8.
Drug Metab Dispos ; 47(12): 1457-1468, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31649125

RESUMEN

An innovative open-label, crossover clinical study was used to investigate the excretion balance, pharmacokinetics, and metabolism of nemiralisib-an inhaled phosphoinositide 3-kinase delta inhibitor being developed for respiratory diseases. Six healthy men received a single intravenous microtracer of 10 µg [14C]nemiralisib with a concomitant inhaled nonradiolabeled 1000 µg dose followed by an oral 800 µg dose of [14C]nemiralisib 14 days later. Complementary methods including accelerator mass spectrometry allowed characterization of a range of parameters including oral absorption (Fabs), proportion of nemiralisib escaping gut wall metabolism (Fg), hepatic extraction (Eh), fraction of dose absorbed from inhaled dose (Flung), and renal clearance. Intravenous pharmacokinetics of nemiralisib were characterized by low blood clearance (10.0 l/h), long terminal half-life (55 hours), and high volume of distribution at steady state (728 l). Nemiralisib exhibited moderate inhaled and oral bioavailability (38% and 35%) while Flung was 29%. Absorption and first-pass parameters were corrected for blood renal clearance and compared with values without correction. Any swallowed nemiralisib was relatively well absorbed (Fabs, 0.48) with a high fraction escaping gut wall metabolism and low extraction by the liver (Fg and Eh being 0.83 and 0.10, respectively). There were no major human plasma metabolites requiring further qualification in animal studies. Both unchanged nemiralisib and its oxidative/conjugative metabolites were secreted in bile, with nemiralisib likely subject to further metabolism through enterohepatic recirculation. Direct renal clearance and metabolism followed by renal clearance were lesser routes of elimination. SIGNIFICANCE STATEMENT: A number of innovative features have been combined into one small clinical study enabling a comprehensive description of the human pharmacokinetics and metabolism of an inhaled molecule. Design elements included an intravenous 14C tracer administration concomitant with an inhalation dose that enabled derivation of parameters such as fraction absorbed (Fabs), the proportion of drug escaping first-pass extraction through the gut wall and liver (Fg and Fh) and hepatic extraction (Eh). Entero-test bile sampling enabled characterization of biliary elimination pathways.


Asunto(s)
Monitoreo de Drogas/métodos , Indazoles/farmacocinética , Indoles/farmacocinética , Oxazoles/farmacocinética , Piperazinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Administración por Inhalación , Administración Intravenosa , Administración Oral , Adulto , Disponibilidad Biológica , Radioisótopos de Carbono , Estudios Cruzados , Heces/química , Voluntarios Sanos , Humanos , Indazoles/administración & dosificación , Indazoles/sangre , Indazoles/orina , Indoles/administración & dosificación , Indoles/sangre , Indoles/orina , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Oxazoles/administración & dosificación , Oxazoles/sangre , Oxazoles/orina , Piperazinas/administración & dosificación , Piperazinas/sangre , Piperazinas/orina , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Distribución Tisular
9.
Br J Pharmacol ; 176(13): 2162-2178, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30875096

RESUMEN

BACKGROUND AND PURPOSE: Sunitinib is a small-molecule TK inhibitor associated with hepatotoxicity. The mechanisms of its toxicity are still unclear. EXPERIMENTAL APPROACH: In the present study, mice were treated with 60, 150, and 450 mg·kg-1 sunitinib to evaluate sunitinib hepatotoxicity. Sunitinib metabolites and endogenous metabolites in liver, serum, faeces, and urine were analysed using ultra-performance LC electrospray ionization quadrupole time-of-flight MS-based metabolomics. KEY RESULTS: Four reactive metabolites and impaired clearance of sunitinib in liver played a dominant role in sunitinib-induced hepatotoxicity. Using a non-targeted metabolomics approach, various metabolic pathways, including mitochondrial fatty acid ß-oxidation (ß-FAO), bile acids, lipids, amino acids, nucleotides, and tricarboxylic acid cycle intermediates, were disrupted after sunitinib treatment. CONCLUSIONS AND IMPLICATIONS: These studies identified significant alterations in mitochondrial ß-FAO and bile acid homeostasis. Activation of PPARα and inhibition of xenobiotic metabolism may be of value in attenuating sunitinib hepatotoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedades Metabólicas/inducido químicamente , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Sunitinib/efectos adversos , Sunitinib/farmacocinética , Animales , Antineoplásicos/sangre , Antineoplásicos/orina , Ácidos y Sales Biliares/metabolismo , Ácidos Grasos/metabolismo , Heces/química , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Sunitinib/sangre , Sunitinib/orina
10.
Clin Pharmacol Drug Dev ; 8(1): 22-31, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30256541

RESUMEN

In this open-label study (NCT02142920), we investigated the distribution, pharmacokinetics, and metabolism of the pan-class-I isoform phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor gedatolisib (PF-05212384), following a single intravenous administration in healthy male subjects. A single, 89-mg, intravenous dose of gedatolisib was associated with a favorable safety profile in the 6 healthy subjects evaluated. Peak plasma concentrations for unchanged gedatolisib and total radioactivity were observed at the end of the 30-minute infusion. The only observed drug-related material in plasma was the parent drug, gedatolisib. Terminal half-life for plasma gedatolisib was ∼37 hours. Following the dose, 66%-73% of drug-related material was recovered in the feces. Metabolism of gedatolisib was trace; only 1 oxidative metabolite, M5, was identified in feces (<1% of total dose). Identification of gedatolisib in feces suggests that biliary and/or intestinal secretion of unchanged parent drug significantly contributes to gedatolisib clearance.


Asunto(s)
Morfolinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Triazinas/farmacocinética , Adulto , Células Cultivadas , Ayuno/metabolismo , Heces/química , Voluntarios Sanos , Hepatocitos , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Morfolinas/administración & dosificación , Morfolinas/sangre , Morfolinas/orina , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Triazinas/administración & dosificación , Triazinas/sangre , Triazinas/orina
11.
Invest New Drugs ; 37(4): 666-673, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30397836

RESUMEN

Aims This two-part, phase I study evaluated the mass balance, excretion, pharmacokinetics and safety of the investigational aurora A kinase inhibitor, alisertib, in three patients with advanced malignancies. Methods Part A; patients received a single 35-mg dose of [14C]-alisertib oral solution (~80 µCi total radioactivity [TRA]). Serial blood, urine, and fecal samples were collected up to 336 h post-dose for alisertib mass balance and pharmacokinetics in plasma and urine by liquid chromatography-tandem mass spectrometry, and mass balance/recovery of [14C]-radioactivity in urine and feces by liquid scintillation counting. Part B; patients received non-radiolabeled alisertib 50 mg as enteric-coated tablets twice-daily for 7 days in 21-day cycles. Results In part A, absorption was fast (median plasma Tmax, 1 h) for alisertib and TRA. Mean plasma t1/2 for alisertib and TRA were 23.4 and 42.0 h, respectively. Mean plasma alisertib/TRA AUC0-inf ratio was 0.45, indicating presence of alisertib metabolites in circulation. Mean TRA blood/plasma AUC0-last ratio was 0.60, indicating preferential distribution of drug-related material in plasma. On average, 87.8% and 2.7% of administered radioactivity was recovered in feces and urine, respectively (total recovery, 90.5% by 14 days post-dose). In part B, patients received a median 3 cycles of alisertib. The most common any-grade adverse events were fatigue and alopecia. Conclusions Findings suggest that alisertib is eliminated mainly via feces, consistent with hepatic metabolism and biliary excretion of drug-related material. Further investigation of alisertib pharmacokinetics in patients with moderate-severe hepatic impairment is warranted to inform dosing recommendations in these patient populations.


Asunto(s)
Antineoplásicos/farmacocinética , Aurora Quinasa A/antagonistas & inhibidores , Azepinas/farmacocinética , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/farmacocinética , Administración Oral , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Antineoplásicos/orina , Azepinas/efectos adversos , Azepinas/sangre , Azepinas/orina , Heces/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Pirimidinas/efectos adversos , Pirimidinas/sangre , Pirimidinas/orina
12.
Xenobiotica ; 49(8): 953-960, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30215545

RESUMEN

1. Leniolisib is a novel oral phosphatidylinositol-3-kinase (PI3K) delta inhibitor, currently in clinical development for the treatment of inflammatory and autoimmune diseases. 2. We investigated the absorption, metabolism, and excretion of leniolisib in healthy subjects after a single oral 400 mg dose as part of a first-in-human clinical study. The parent drug and metabolites were quantified by 19F-NMR in plasma, urine and faeces after liquid chromatography separation, and structures were determined by liquid chromatography coupled to tandem mass spectrometry. 3. Drug-related material was mainly excreted as oxidative metabolites in urine and faeces, providing evidence that elimination occurs mainly by metabolism. No metabolites were abundant in plasma relative to the parent drug. An average mass balance of 66% was obtained, demonstrating that relatively extensive elimination/excretion data can be obtained by 19F-NMR in a first in human clinical study without the use of a radiolabeled drug.


Asunto(s)
Absorción Fisiológica , Flúor/química , Voluntarios Sanos , Espectroscopía de Resonancia Magnética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/metabolismo , Piridinas/metabolismo , Pirimidinas/metabolismo , Administración Oral , Adolescente , Adulto , Heces , Femenino , Humanos , Masculino , Metaboloma , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/orina , Piridinas/sangre , Piridinas/farmacocinética , Piridinas/orina , Pirimidinas/sangre , Pirimidinas/farmacocinética , Pirimidinas/orina , Adulto Joven
14.
Eur J Pharm Sci ; 104: 335-343, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28433749

RESUMEN

Untargeted metabolite profiling using high-resolution mass spectrometry coupled with liquid chromatography (LC-HRMS), followed by data analysis with the Compound Discoverer 2.0™ software, was used to study the metabolism of imatinib in humans with chronic myeloid leukemia. Plasma samples from control (drug-free) and patient (treated with imatinib) groups were analyzed in full-scan mode and the unknown ions occurring only in the patient group were then, as potential imatinib metabolites, subjected to multi-stage fragmentation in order to elucidate their structure. The application of an untargeted approach, as described in this study, enabled the detection of 24 novel structurally unexpected metabolites. Several sulphur-containing compounds, probably originating after the reaction of reactive intermediates of imatinib with endogenous glutathione, were found and annotated as cysteine and cystine adducts. In the proposed mechanism, the cysteine adducts were formed after the rearrangement of piperazine moiety to imidazoline. On the contrary, in vivo S-N exchange occurred in the case of the cystine adducts. In addition, N-O exchange was observed in the collision cell in the course of the fragmentation of the cystine adducts. The presence of sulphur in the cysteine and cystine conjugates was proved by means of ultra-high resolution measurements using Orbitrap Elite. The detection of metabolites derived from glutathione might improve knowledge about the disposition of imatinib towards bioactivation and help to improve understanding of the mechanism of its hepatotoxicity or nephrotoxicity in humans.


Asunto(s)
Antineoplásicos/metabolismo , Mesilato de Imatinib/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Azufre/metabolismo , Antineoplásicos/sangre , Antineoplásicos/orina , Cromatografía Liquida , Cisteína/metabolismo , Cistina/metabolismo , Humanos , Mesilato de Imatinib/sangre , Mesilato de Imatinib/orina , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Azufre/sangre , Azufre/orina , Espectrometría de Masas en Tándem/métodos
15.
Bioanalysis ; 9(5): 459-468, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28176528

RESUMEN

AIM: Alectinib (Alecensa®) is an anaplastic lymphoma kinase inhibitor for the treatment of anaplastic lymphoma kinase positive non-small-cell lung cancer, and M4 is its major pharmacologically active metabolite. To characterize the pharmacokinetics and excretion of alectinib and M4 in human urine, a bioanalytical method was required. RESULTS: An LC-MS/MS method using supported liquid extraction was developed for the determination of alectinib and M4 in human urine over the concentration range 0.5-500 ng/ml. Accuracy ranged from 92.0 to 112.2% and precision (CV) was below 9.6%. CONCLUSION: The method was successfully employed to determine alectinib and M4 concentrations in urine samples from a clinical mass balance study. Addition of the surfactant Tween-20 to urine prevented nonspecific binding of the analytes.


Asunto(s)
Carbazoles/orina , Cromatografía Líquida de Alta Presión/métodos , Piperidinas/orina , Inhibidores de Proteínas Quinasas/orina , Espectrometría de Masas en Tándem/métodos , Carbazoles/metabolismo , Carbazoles/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cromatografía Líquida de Alta Presión/normas , Humanos , Piperidinas/metabolismo , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Control de Calidad , Espectrometría de Masas en Tándem/normas
16.
Bioanalysis ; 8(18): 1919-36, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27523799

RESUMEN

AIM: Selumetinib is an inhibitor of MEK1/2 in Phase III development that has activity in multiple tumor types. Validated bioanalytical methods were required to quantitate selumetinib and its N-desmethyl and amide metabolites in a variety of human biological matrices. Methodology & results: LC-MS/MS assays were developed and validated that demonstrated acceptable precision, accuracy and selectivity for selumetinib and the two metabolites in human plasma, urine, blood dialysate and plasma ultrafiltrate. Incurred sample re-analysis was acceptable and issues observed in plasma with the amide metabolite, due to potential instability, were addressed. CONCLUSION: Robust and sensitive LC-MS/MS assays for the quantification of selumetinib and two of its metabolites were validated in human biological matrices and are being used to support the clinical development program.


Asunto(s)
Bencimidazoles/sangre , Bencimidazoles/orina , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Espectrometría de Masas en Tándem/métodos , Amidas/sangre , Amidas/metabolismo , Amidas/orina , Bencimidazoles/metabolismo , Cromatografía Liquida/métodos , Monitoreo de Drogas/métodos , Humanos , Límite de Detección , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/metabolismo
17.
Clin Pharmacokinet ; 55(1): 47-77, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26201307

RESUMEN

Scientists have identified the impact of angiogenesis on tumor growth and survival. Among other efficient drugs, several small-molecule tyrosine kinase inhibitors (TKIs) targeting the vascular endothelial growth factor receptor (VEGFR) have been developed and have already been integrated into the treatment of various advanced malignancies. This review provides a compilation of current knowledge on the pharmacokinetic aspects of all VEGFR-TKIs already approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) and of those still under investigation. Additional information on substance metabolism, potential for drug-drug interactions (DDIs), and the need for dose adaptation in patients with predominant renal and/or hepatic impairment has been included. All TKIs introduced in this review were administered orally, allowing for easy drug handling for healthcare professionals and patients. For almost all substances, the maximum plasma concentrations were reached within a short period of time. The majority of the substances showed a high plasma protein binding and their excretion occurred via the feces and, to a lesser extent, via the urine. In most cases, dose adaptation in patients with mild to moderate renal or hepatic impairment is not recommended. Cytochrome P450 (CYP) 3A4 was found to play a crucial role in the drug metabolic processes of many compounds. In order to prevent unwanted DDIs, co-administration of VEGFR TKIs together with CYP3A4 inhibitors or inducers should be avoided. Throughout all TKIs, the data indicate high inter-individual variability. The causes of this are still unclear and require further research to allow for individualization of treatment regimens.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Oral , Interacciones Farmacológicas , Heces/química , Humanos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/orina
18.
Drug Metab Dispos ; 44(1): 28-39, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26451002

RESUMEN

The pharmacokinetics, metabolism, and excretion of cobimetinib, a MEK inhibitor, were characterized in healthy male subjects (n = 6) following a single 20 mg (200 µCi) oral dose. Unchanged cobimetinib and M16 (glycine conjugate of hydrolyzed cobimetinib) were the major circulating species, accounting for 20.5% and 18.3% of the drug-related material in plasma up to 48 hours postdose, respectively. Other circulating metabolites were minor, accounting for less than 10% of drug-related material in plasma. The total recovery of the administered radioactivity was 94.3% (±1.6%, S.D.) with 76.5% (±2.3%) in feces and 17.8% (±2.5%) in urine. Metabolite profiling indicated that cobimetinib had been extensively metabolized with only 1.6% and 6.6% of the dose remaining as unchanged drug in urine and feces, respectively. In vitro phenotyping experiments indicated that CYP3A4 was predominantly responsible for metabolizing cobimetinib. From this study, we concluded that cobimetinib had been well absorbed (fraction absorbed, Fa = 0.88). Given this good absorption and the previously determined low hepatic clearance, the systemic exposures were lower than expected (bioavailability, F = 0.28). We hypothesized that intestinal metabolism had strongly attenuated the oral bioavailability of cobimetinib. Supporting this hypothesis, the fraction escaping gut wall elimination (Fg) was estimated to be 0.37 based on F and Fa from this study and the fraction escaping hepatic elimination (Fh) from the absolute bioavailability study (F = Fa × Fh × Fg). Physiologically based pharmacokinetics modeling also showed that intestinal clearance had to be included to adequately describe the oral profile. These collective data suggested that cobimetinib was well absorbed following oral administration and extensively metabolized with intestinal first-pass metabolism contributing to its disposition.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Azetidinas/administración & dosificación , Azetidinas/farmacocinética , Absorción Intestinal , Mucosa Intestinal/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Eliminación Renal , Administración Oral , Adulto , Antineoplásicos/sangre , Antineoplásicos/orina , Azetidinas/sangre , Azetidinas/orina , Disponibilidad Biológica , Biotransformación , Radioisótopos de Carbono , Citocromo P-450 CYP3A/metabolismo , Heces/química , Glicina/metabolismo , Voluntarios Sanos , Humanos , Hidrólisis , Intestinos/enzimología , Masculino , Microsomas Hepáticos/metabolismo , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Piperidinas/sangre , Piperidinas/orina , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Especificidad por Sustrato , Adulto Joven
19.
Biomed Chromatogr ; 30(4): 596-600, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26284315

RESUMEN

A novel and selective liquid chromatographic-mass spectrometric method (LC-MS/MS) has been established and validated for simultaneous determination of subutinib and active metabolite in human urine. Urine samples were extracted by liquid-liquid extraction with ethyl acetate and separated on a Wondasil C18 (150 × 2.1 mm, 3.5 µm), with methanol-0.2% formic acid solution (73:27, v/v) as mobile phase at flow rate of 0.2 mL/min. The linear range was 0.5000-200.0 ng/mL for subutinib and active metabolite, with a lower limit of quantitation of 0.5000 ng/mL. Intra- and inter-run precisions were all <11.8 and 14.3%, and the accuracies were all <4.5 and 5.4%, with the extraction recoveries 88.8-97.5 and 93.8-99.4% for the two analytes, respectively. The carryover values were all <15% for the two anayltes. The method was successfully applied to study urinary excretion of subutinib and active metabolite in human after oral administration of subutinib maleate capsules in fed and fasting states.


Asunto(s)
Antineoplásicos/orina , Indoles/orina , Inhibidores de Proteínas Quinasas/orina , Pirroles/orina , Espectrometría de Masas en Tándem/métodos , Adulto , Antineoplásicos/metabolismo , Cromatografía Liquida/métodos , Femenino , Humanos , Indoles/metabolismo , Límite de Detección , Masculino , Inhibidores de Proteínas Quinasas/metabolismo , Pirroles/metabolismo , Adulto Joven
20.
Xenobiotica ; 45(10): 887-902, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25986538

RESUMEN

1. The human mass balance of (14)C-labelled ASP015K ([(14)C]ASP015K), an orally bioavailable Janus kinase (JAK) inhibitor, was characterized in six healthy male subjects after a single oral dose of [(14)C]ASP015K (100 mg, 3.7 MBq) in solution. [(14)C]ASP015K was rapidly absorbed with tmax of 1.6 and 1.8 h for ASP015K and total radioactivity in plasma, respectively. Mean recovery in urine and feces amounted to 36.8% and 56.6% of the administered dose, respectively. The main components of radioactivity in plasma and urine were ASP015K and M2 (5'-O-sulfo ASP015K). In feces, ASP015K and M4 (7-N-methyl ASP015K) were the main components. 2. In vitro study of ASP015K metabolism showed that the major isozyme contributing to the formation of M2 was human sulfotransferase (SULT) 2A1 and of M4 was nicotinamide N-methyltransferase (NNMT). 3. The in vitro intrinsic clearance (CLint_in vitro) of M4 formation from ASP015K in human liver cytosol (HLC) was 11-fold higher than that of M2. The competitive inhibitory effect of nicotinamide on M4 formation in the human liver was considered the reason for high CLint_in vitro of M4 formation, while each metabolic pathway made a near equal contribution to the in vivo elimination of ASP015K. ASP015K was cleared by multiple mechanisms.


Asunto(s)
Adamantano/análogos & derivados , Quinasas Janus/antagonistas & inhibidores , Niacinamida/análogos & derivados , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Adamantano/administración & dosificación , Adamantano/farmacocinética , Administración Oral , Adolescente , Adulto , Radioisótopos de Carbono/administración & dosificación , Radioisótopos de Carbono/farmacocinética , Heces , Humanos , Quinasas Janus/metabolismo , Masculino , Tasa de Depuración Metabólica , Metilación , Persona de Mediana Edad , Niacinamida/administración & dosificación , Niacinamida/farmacocinética , Nicotinamida N-Metiltransferasa/genética , Nicotinamida N-Metiltransferasa/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/orina , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...