Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Ther Drug Monit ; 46(5): 634-641, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38531816

RESUMEN

BACKGROUND: Ibrutinib and zanubrutinib are Bruton tyrosine kinase inhibitors used to treat mantle cell lymphoma, chronic lymphocytic leukemia, and small lymphocytic lymphoma. Dihydroxydiol ibrutinib (DHI) is an active metabolite of the drug. A liquid chromatography-tandem mass spectrometry method was developed to detect ibrutinib, DHI, and zanubrutinib in human plasma. METHODS: The method involved a protein precipitation step, followed by chromatographic separation using a gradient of 10 mM ammonium acetate (containing 0.1% formic acid)-acetonitrile. Ibrutinib-d5 was used as an internal standard. Analytes were separated within 6.5 minutes. The optimized multiple reaction monitoring transitions of m/z 441.1 → 304.2, 475.2 → 304.2, 472.2 → 455.2, and 446.2 → 309.2 were selected to inspect ibrutinib, DHI, zanubrutinib, and the internal standards in positive ion mode. RESULTS: The validated curve ranges included 0.200-800, 0.500-500, and 1.00-1000 ng/mL for ibrutinib, DHI, and zanubrutinib, respectively. The precisions of the lower limit of quantification of samples were below 15.5%, the precisions of the other level samples were below 11.4%, and the accuracies were between -8.6% and 8.4%. The matrix effect and extraction recovery of all compounds ranged between 97.6%-109.0% and 93.9%-105.2%, respectively. The selectivity, accuracy, precision, matrix effect, and extraction recovery results were acceptable according to international method validation guidelines. CONCLUSIONS: A simple and rapid method was developed and validated in this study. This method was used to analyze plasma concentrations of ibrutinib and zanubrutinib in patients with mantle cell lymphoma, chronic lymphocytic leukemia/small lymphocytic lymphoma, or diffuse large B-cell lymphoma. The selected patients were aged between 44 and 74 years.


Asunto(s)
Adenina , Piperidinas , Pirazoles , Pirimidinas , Espectrometría de Masas en Tándem , Humanos , Piperidinas/sangre , Piperidinas/uso terapéutico , Adenina/análogos & derivados , Adenina/uso terapéutico , Adenina/sangre , Pirimidinas/sangre , Pirimidinas/uso terapéutico , Espectrometría de Masas en Tándem/métodos , Pirazoles/sangre , Pirazoles/uso terapéutico , Cromatografía Liquida/métodos , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/uso terapéutico , Reproducibilidad de los Resultados , Pirazinas/sangre , Pirazinas/uso terapéutico , Cromatografía Líquida con Espectrometría de Masas
2.
Luminescence ; 39(8): e4837, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39113185

RESUMEN

During the pandemic, Favipiravir (FVP) and Molnupiravir (MPV) have been widely used for COVID-19 treatment, leading to their presence in the environment. A green synchronous spectrofluorimetric method was developed to simultaneously detect them in environmental water, human plasma, and binary mixtures. Maximum fluorescence intensity was achieved at pH 8, with MPV exhibiting two peaks at 300 and 430 nm, and FVP showing one peak at 430 nm. A fluorescence subtraction method effectively removed interference, enabling direct determination of MPV at 300 nm and FVP at 430 nm. The method showed linearity within 2-13 ng/mL for FVP and 50-600 ng/mL for MPV, with recoveries of 100.35% and 100.12%, respectively. Limits of detection and quantification were 0.19 and 0.57 ng/mL for FVP and 10.52 and 31.88 ng/mL for MPV. Validation according to ICH and FDA guidelines yielded acceptable results. The method demonstrated good recoveries of FVP and MPV in pharmaceuticals, tap water and Nile water (99.62% ± 0.96% and 99.69% ± 0.64%) as per ICH guidelines and spiked human plasma (94.87% ± 2.111% and 94.79% ± 1.605%) following FDA guidelines, respectively. Its environmental friendliness was assessed using Green Analytical Procedure Index (GAPI) and the Analytical Greenness Metric (AGREE) tools.


Asunto(s)
Amidas , Antivirales , Pirazinas , Espectrometría de Fluorescencia , Pirazinas/análisis , Pirazinas/sangre , Pirazinas/química , Amidas/análisis , Amidas/química , Amidas/sangre , Espectrometría de Fluorescencia/métodos , Humanos , Antivirales/análisis , Antivirales/sangre , Uridina/análisis , Uridina/sangre , Límite de Detección , Citidina/análisis , Citidina/sangre , Citidina/análogos & derivados , Tratamiento Farmacológico de COVID-19 , Mercaptopurina/sangre , Mercaptopurina/análisis , SARS-CoV-2 , Hidroxilaminas
3.
Mikrochim Acta ; 189(3): 125, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35229221

RESUMEN

A novel electrochemical sensor is reported for the detection of the antiviral drug favipiravir based on the core-shell nanocomposite of flower-like molybdenum disulfide (MoS2) nanospheres and molecularly imprinted polymers (MIPs). The MoS2@MIP core-shell nanocomposite was prepared via the electrodeposition of a MIP layer on the MoS2 modified electrode, using o-phenylenediamine as the monomer and favipiravir as the template. The selective binding of target favipiravir at the MoS2@MIP core-shell nanocomposite produced a redox signal in a concentration dependent manner, which was used for the quantitative analysis. The preparation process of the MoS2@MIP core-shell nanocomposite was optimized. Under the optimal conditions, the sensor exhibited a wide linear response range of 0.01 ~ 100 nM (1.57*10-6 ~ 1.57*10-2 µg mL-1) and a low detection limit of 0.002 nM (3.14*10-7 µg mL-1). Application of the sensor was demonstrated by detecting favipiravir in a minimum amount of 10 µL biological samples (urine and plasma). Satisfied results in the recovery tests indicated a high potential of favipiravir monitoring in infectious COVID-19 samples.


Asunto(s)
Amidas/análisis , Antivirales/análisis , Disulfuros/química , Polímeros Impresos Molecularmente/química , Molibdeno/química , Nanocompuestos/química , Nanosferas/química , Pirazinas/análisis , Amidas/sangre , Amidas/uso terapéutico , Amidas/orina , Antivirales/sangre , Antivirales/uso terapéutico , Antivirales/orina , COVID-19/virología , Técnicas Electroquímicas/métodos , Humanos , Límite de Detección , Oxidación-Reducción , Pirazinas/sangre , Pirazinas/uso terapéutico , Pirazinas/orina , Reproducibilidad de los Resultados , SARS-CoV-2/aislamiento & purificación , Tratamiento Farmacológico de COVID-19
4.
Biomed Chromatogr ; 35(7): e5098, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33606892

RESUMEN

A novel, simple and rapid UPLC-MS/MS method was developed and validated for determination of favipiravir (FAV) in human plasma. Lamivudine was used as an internal standard (IS). The Xevo TQD LC-MS/MS was operated under the multiple-reaction monitoring mode using electrospray ionization. Precipitation with acetonitrile was used in sample preparation as it gives relatively cleaner plasma samples. The prepared samples were chromatographed using an Acquity UPLC® HSS C18 (100 × 2.1 mm, 1.8 µm) column. The mobile phase was composed of ammonium formate and methanol in a gradient mode that was pumped at a flow rate of 0.35 ml/min. The developed method was validated as per the FDA guidelines and linearity was in the range of 0.25-16 µg/ml for FAV. The intra- and inter-day precision and accuracy results were within the acceptable limits. A run time of 4.5 min and a low quantification limit of FAV allowed the application of the developed method for the determination of FAV in a bioequivalence study in healthy human volunteers.


Asunto(s)
Amidas/sangre , Cromatografía Líquida de Alta Presión/métodos , Pirazinas/sangre , Espectrometría de Masas en Tándem/métodos , Adulto , Amidas/química , Amidas/farmacocinética , Femenino , Humanos , Límite de Detección , Masculino , Persona de Mediana Edad , Pirazinas/química , Pirazinas/farmacocinética , Reproducibilidad de los Resultados , Equivalencia Terapéutica , Adulto Joven
5.
Biomed Chromatogr ; 35(4): e5028, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33179270

RESUMEN

Gilteritinib, an oral inhibitor of FMS-like tyrosine kinase 3 (FLT3), is a standard treatment for FLT3-mutated acute myeloid leukemia. We developed a simple HPLC-UV-based method for determining the concentration of gilteritinib in human plasma. The analysis requires the extraction of a 200-µL plasma sample and the precipitation of proteins by solid-phase extraction. Gilteritinib was isocratically separated within 10 min using a mobile phase of acetonitrile:0.5% monopotassium phosphate (KH2 PO4 , pH 3.5, 28:72, v/v) on a Capcell Pack C18 MG II (250 × 4.6 mm) column at a flow rate of 1.0 mL/min and monitored at 250 nm. The calibration curve was found to be linear within a plasma concentration range of 25-2500 ng/mL, with the coefficient of determination (r2 ) being 0.9997. The coefficients of intra-day and inter-day validation were 2.3-3.7 and 1.3-5.2%, respectively. The accuracy and recovery of the assay were -9.6 to 0.1 and >81.8%, respectively. This HPLC-UV method for determining the plasma concentration of gilteritinib is simple and can be effectively applied to routine drug monitoring.


Asunto(s)
Compuestos de Anilina/sangre , Cromatografía Líquida de Alta Presión/métodos , Pirazinas/sangre , Anciano , Compuestos de Anilina/uso terapéutico , Antineoplásicos/uso terapéutico , Femenino , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Modelos Lineales , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazinas/uso terapéutico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrofotometría Ultravioleta
6.
Molecules ; 26(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206357

RESUMEN

In the current work, a simple, economical, accurate, and precise HPLC method with UV detection was developed to quantify Favipiravir (FVIR) in spiked human plasma using acyclovir (ACVR) as an internal standard in the COVID-19 pandemic time. Both FVIR and ACVR were well separated and resolved on the C18 column using the mobile phase blend of methanol:acetonitrile:20 mM phosphate buffer (pH 3.1) in an isocratic mode flow rate of 1 mL/min with a proportion of 30:10:60 %, v/v/v. The detector wavelength was set at 242 nm. Maximum recovery of FVIR and ACVR from plasma was obtained with dichloromethane (DCM) as extracting solvent. The calibration curve was found to be linear in the range of 3.1-60.0 µg/mL with regression coefficient (r2) = 0.9976. However, with acceptable r2, the calibration data's heteroscedasticity was observed, which was further reduced using weighted linear regression with weighting factor 1/x. Finally, the method was validated concerning sensitivity, accuracy (Inter and Intraday's % RE and RSD were 0.28, 0.65 and 1.00, 0.12 respectively), precision, recovery (89.99%, 89.09%, and 90.81% for LQC, MQC, and HQC, respectively), stability (% RSD for 30-day were 3.04 and 1.71 for LQC and HQC, respectively at -20 °C), and carry-over US-FDA guidance for Bioanalytical Method Validation for researchers in the COVID-19 pandemic crisis. Furthermore, there was no significant difference for selectivity when evaluated at LLOQ concentration of 3 µg/mL of FVIR and relative to the blank.


Asunto(s)
Amidas/análisis , Amidas/sangre , Antivirales/análisis , Antivirales/sangre , Bioensayo/métodos , Tratamiento Farmacológico de COVID-19 , Cromatografía Líquida de Alta Presión/métodos , Extracción Líquido-Líquido/métodos , Pirazinas/análisis , Pirazinas/sangre , Aciclovir/análisis , Aciclovir/sangre , COVID-19/sangre , Calibración , Estabilidad de Medicamentos , Congelación , Humanos , Estándares de Referencia , Reproducibilidad de los Resultados , Solventes/química
7.
Biomed Chromatogr ; 34(9): e4869, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32346872

RESUMEN

To investigate the effect of ligustrazine on the pharmacokinetic profile of tanshinol after intravenous administration in rats, a sensitive liquid chromatography tandem mass spectrometry method was developed and validated for quantitative determination of tanshinol and ligustrazine in rat plasma. After prepared by protein precipitation, the analytes were separated on a Waters Acquity HSS T3 column (100 × 2.1 mm, 1.8µm) and eluted by 0.1% formic acid in water and acetonitrile at a flow rate of 0.4 ml/min. The precursor-product ion transitions were m/z 197.0 → 135.0 for tanshinol, m/z 417.1 → 255.1 for liquiritin (internal standard) in negative ion mode and m/z 137.1 → 55.0 for ligustrazine in positive ion mode. To avoid the interference of tanshinol metabolite transformation, the stability of analytes in samples collected after administration was assessed. The validated method was successfully applied to a pharmacokinetic study after intravenous administration of single tanshinol and Danshen Chuanxiongqin Injection. After Danshen Chuanxiongqin injection administration, the values of elimination half-time, area under the concentration-time curve and Co were 0.36 ± 0.13 h, 1.29 ± 0.37 µg/ml h and 10.51 ± 2.58 µg/ml for male rats, respectively. In the single tanshinol group, the corresponding values were 0.56 ± 0.24 h, 1.85 ± 0.44 µg/ml h and 14.11 ± 2.26 µg/ml for male rats-30-40% higher than those for the Danshen Chuanxiongqin Injection group. There was a significant different between male and female rats. This study provided information on the influence of ligustrazine on the pharmacokinetic characteristics of tanshinol after intravenous administration of Danshen Chuanxiongqin Injection in rats, which will be helpful for its clinical application.


Asunto(s)
Ácidos Cafeicos , Pirazinas , Administración Intravenosa , Animales , Ácidos Cafeicos/administración & dosificación , Ácidos Cafeicos/sangre , Ácidos Cafeicos/química , Ácidos Cafeicos/farmacocinética , Cromatografía Líquida de Alta Presión/métodos , Femenino , Modelos Lineales , Masculino , Pirazinas/administración & dosificación , Pirazinas/sangre , Pirazinas/química , Pirazinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Salvia miltiorrhiza , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem/métodos
8.
Pharm Dev Technol ; 24(10): 1236-1242, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31407940

RESUMEN

Objective: Tanshinone IIA (TSN) and Tetramethylpyrazine (TMP) were combined in a composite, oil-in-water nanoemulsions (TSN/TMP O/W NEs) was prepared to prolong in vitro and vivo circulation time, and enhance the bioavailability of TSN. Material and methods: Physicochemical characterization of TSN/TMP O/W NEs was characterized systematically. The in vitro dissolution and in vivo pharmacokinetic experiments of TSN/TMP O/W NEs were also evaluated. Result: A formulation was optimized, yielding a 32.5 nm average particle size, an encapsulation efficiency of over 95 %, and were spherical in shape as shown by TEM. TSN/TMP O/W NEs were shown to extend the release and availability in vitro compared to raw compounds. In pharmacokinetic study, the AUC0→∞ and t1/2 of the TSN/TMP O/W NEs were 481.50 mg/L*min and 346.39 min higher than TSN solution, respectively. Brain tissue concentration of TSN was enhanced with TSN/TMP O/W NEs over raw TSN and even TSN O/W NEs. Conclusions: Therefore, nanoemulsions are an effective carrier to increase encapsulation efficiency of drugs, improve bioavailability and brain penetration for TSN - which is further enhanced by pairing with the co-delivery of TMP, providing a promising drug delivery.


Asunto(s)
Abietanos/química , Abietanos/farmacocinética , Encéfalo/metabolismo , Composición de Medicamentos/métodos , Nanocompuestos/química , Pirazinas/química , Pirazinas/farmacocinética , Abietanos/sangre , Animales , Disponibilidad Biológica , Combinación de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Emulsiones , Tamaño de la Partícula , Pirazinas/sangre , Ratas Sprague-Dawley , Solubilidad , Propiedades de Superficie , Distribución Tisular
9.
Artículo en Inglés | MEDLINE | ID: mdl-30249687

RESUMEN

The in vivo antimalarial efficacies of two phosphatidylinositol 4-kinase (PI4K) inhibitors, a 3,5-diaryl-2-aminopyrazine sulfoxide and its corresponding sulfone metabolite, were evaluated in the NOD-scid IL2Rγnull (NSG) murine malaria disease model of Plasmodium falciparum infection. We hypothesized that the sulfoxide would serve as a more soluble prodrug for the sulfone, which would lead to improved drug exposure with oral dosing. Both compounds had similar efficacy (90% effective dose [ED90], 0.1 mg kg-1 of body weight) across a quadruple-dose regimen. Pharmacokinetic profiling revealed rapid sulfoxide clearance via conversion to sulfone, with sulfone identified as the major active metabolite. When the sulfoxide was dosed, the exposure of the sulfone achieved was as much as 2.9-fold higher than when the sulfone was directly dosed, thereby demonstrating that the sulfoxide served as an effective prodrug for the treatment of malaria.


Asunto(s)
Antimaláricos/farmacología , Malaria Falciparum/tratamiento farmacológico , Parasitemia/tratamiento farmacológico , Profármacos/farmacología , Pirazinas/farmacología , Sulfonas/farmacología , Sulfóxidos/farmacología , 1-Fosfatidilinositol 4-Quinasa/antagonistas & inhibidores , 1-Fosfatidilinositol 4-Quinasa/genética , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Animales , Antimaláricos/sangre , Antimaláricos/síntesis química , Antimaláricos/farmacocinética , Biotransformación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Eritrocitos/parasitología , Expresión Génica , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Parasitemia/patología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Profármacos/síntesis química , Profármacos/farmacocinética , Pirazinas/sangre , Pirazinas/síntesis química , Pirazinas/farmacocinética , Sulfonas/sangre , Sulfonas/síntesis química , Sulfonas/farmacocinética , Sulfóxidos/sangre , Sulfóxidos/síntesis química , Sulfóxidos/farmacocinética , Resultado del Tratamiento
10.
Xenobiotica ; 48(7): 704-719, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28737453

RESUMEN

1. The metabolism of selexipag has been studied in vivo in man and the main excreted metabolites were identified. Also, metabolites circulating in human plasma have been structurally identified and quantified. 2. The main metabolic pathway of selexipag in man is the formation of the active metabolite ACT-333679. Other metabolic pathways include oxidation and dealkylation reactions. All primary metabolites undergo subsequent hydrolysis of the sulphonamide moiety to their corresponding acids. ACT-333679 undergoes conjugation with glucuronic acid and aromatic hydroxylation to P10, the main metabolite detected in human faeces. 3. The formation of the active metabolite ACT-333679 is catalysed by carboxylesterases, while the oxidation and dealkylation reactions are metabolized by CYP2C8 and CYP3A4. CYP2C8 is the only P450 isoform catalysing the aromatic hydroxylation to P10. CYP2C8 together with CYP3A4 are also involved in the formation of several minor ACT-333679 metabolites. UGT1A3 and UGT2B7 catalyse the glucuronidation of ACT-333679. 4. The potential of selexipag to inhibit or induce cytochrome P450 enzymes or drug transport proteins was studied in vitro. Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. Also, selexipag inhibits the transporters OATP1B1, OATP1B3, OAT1, OAT3, and BCRP. However, due to its low dose and relatively low unbound exposure, selexipag has a low potential for causing drug-drug interactions.


Asunto(s)
Acetamidas/metabolismo , Acetamidas/farmacología , Pirazinas/metabolismo , Pirazinas/farmacología , Receptores de Epoprostenol/agonistas , Acetamidas/sangre , Acetamidas/química , Acetatos/farmacología , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Esterasas/antagonistas & inhibidores , Esterasas/metabolismo , Hepatocitos/metabolismo , Humanos , Proteínas de Transporte de Membrana/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Metaboloma , Metabolómica , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , NADP/metabolismo , Pirazinas/sangre , Pirazinas/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Epoprostenol/metabolismo , Proteínas Recombinantes/metabolismo
11.
Biomed Chromatogr ; 32(6): e4210, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29431191

RESUMEN

Tetramethylpyrazine (TMP) has been widely used in the treatment of ischemic cerebrovascular disease. However, the mechanism of TMP and how to increase its bioavailability need to be further explored. In our study, an in vivo microdialysis sampling technique coupled with ultra-performance liquid chromatography-mass spectrometry method was developed to investigate the pharmacokinetic properties of TMP and its interaction with different doses of borneol (BO) in rats. Linearity of TMP in brain and blood dialysates exhibited good linear relationships over the concentration range of 0.991-555.14 ng/mL. The specificity, linearity, accuracy, precision, matrix effect and stability were within acceptable ranges. The results demonstrated that BO had a marked impact on the pharmacokinetic properties of TMP. After co-administration, the areas under the concentration-time curve (AUC) of TMP in brain and blood were significantly increased. Meanwhile, the peak concentration of TMP in brain was also enhanced. The AUCBrain /AUCBlood of TMP, increased from 44% to 56 and 60.8% after co-administration with BO (15 and 30 mg/kg). The pharmacodynamic results showed that TMP co-administration with BO enhanced the cerebral blood flow during the period of ischemia and reduced the infarct volume. Overall, it might be an effective way to treat stroke to use TMP co-administered with BO.


Asunto(s)
Encéfalo/metabolismo , Canfanos/farmacocinética , Cromatografía Líquida de Alta Presión/métodos , Microdiálisis/métodos , Pirazinas , Animales , Química Encefálica , Canfanos/química , Estabilidad de Medicamentos , Modelos Lineales , Masculino , Pirazinas/análisis , Pirazinas/sangre , Pirazinas/química , Pirazinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem/métodos
12.
Lancet Oncol ; 18(8): 1061-1075, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28645776

RESUMEN

BACKGROUND: Internal tandem duplication mutations in FLT3 are common in acute myeloid leukaemia and are associated with rapid relapse and short overall survival. The clinical benefit of FLT3 inhibitors in patients with acute myeloid leukaemia has been limited by rapid generation of resistance mutations, particularly in codon Asp835 (D835). We aimed to assess the highly selective oral FLT3 inhibitor gilteritinib in patients with relapsed or refractory acute myeloid leukaemia. METHODS: In this phase 1-2 trial, we enrolled patients aged 18 years or older with acute myeloid leukaemia who either were refractory to induction therapy or had relapsed after achieving remission with previous treatment. Patients were enrolled into one of seven dose-escalation or dose-expansion cohorts assigned to receive once-daily doses of oral gilteritinib (20 mg, 40 mg, 80 mg, 120 mg, 200 mg, 300 mg, or 450 mg). Cohort expansion was based on safety and tolerability, FLT3 inhibition in correlative assays, and antileukaemic activity. Although the presence of an FLT3 mutation was not an inclusion criterion, we required ten or more patients with locally confirmed FLT3 mutations (FLT3mut+) to be enrolled in expansion cohorts at each dose level. On the basis of emerging findings, we further expanded the 120 mg and 200 mg dose cohorts to include FLT3mut+ patients only. The primary endpoints were the safety, tolerability, and pharmacokinetics of gilteritinib. Safety and tolerability were assessed in the safety analysis set (all patients who received at least one dose of gilteritinib). Responses were assessed in the full analysis set (all patients who received at least one dose of study drug and who had at least one datapoint post-treatment). Pharmacokinetics were assessed in a subset of the safety analysis set for which sufficient data for concentrations of gilteritinib in plasma were available to enable derivation of one or more pharmacokinetic variables. This study is registered with ClinicalTrials.gov, number NCT02014558, and is ongoing. FINDINGS: Between Oct 15, 2013, and Aug 27, 2015, 252 adults with relapsed or refractory acute myeloid leukaemia received oral gilteritinib once daily in one of seven dose-escalation (n=23) or dose-expansion (n=229) cohorts. Gilteritinib was well tolerated; the maximum tolerated dose was established as 300 mg/day when two of three patients enrolled in the 450 mg dose-escalation cohort had two dose-limiting toxicities (grade 3 diarrhoea and grade 3 elevated aspartate aminotransferase). The most common grade 3-4 adverse events irrespective of relation to treatment were febrile neutropenia (97 [39%] of 252), anaemia (61 [24%]), thrombocytopenia (33 [13%]), sepsis (28 [11%]), and pneumonia (27 [11%]). Commonly reported treatment-related adverse events were diarrhoea (92 [37%] of 252]), anaemia (86 [34%]), fatigue (83 [33%]), elevated aspartate aminotransferase (65 [26%]), and increased alanine aminotransferase (47 [19%]). Serious adverse events occurring in 5% or more of patients were febrile neutropenia (98 [39%] of 252; five related to treatment), progressive disease (43 [17%]), sepsis (36 [14%]; two related to treatment), pneumonia (27 [11%]), acute renal failure (25 [10%]; five related to treatment), pyrexia (21 [8%]; three related to treatment), bacteraemia (14 [6%]; one related to treatment), and respiratory failure (14 [6%]). 95 people died in the safety analysis set, of which seven deaths were judged possibly or probably related to treatment (pulmonary embolism [200 mg/day], respiratory failure [120 mg/day], haemoptysis [80 mg/day], intracranial haemorrhage [20 mg/day], ventricular fibrillation [120 mg/day], septic shock [80 mg/day], and neutropenia [120 mg/day]). An exposure-related increase in inhibition of FLT3 phosphorylation was noted with increasing concentrations in plasma of gilteritinib. In-vivo inhibition of FLT3 phosphorylation occurred at all dose levels. At least 90% of FLT3 phosphorylation inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher. 100 (40%) of 249 patients in the full analysis set achieved a response, with 19 (8%) achieving complete remission, ten (4%) complete remission with incomplete platelet recovery, 46 (18%) complete remission with incomplete haematological recovery, and 25 (10%) partial remission INTERPRETATION: Gilteritinib had a favourable safety profile and showed consistent FLT3 inhibition in patients with relapsed or refractory acute myeloid leukaemia. These findings confirm that FLT3 is a high-value target for treatment of relapsed or refractory acute myeloid leukaemia; based on activity data, gilteritinib at 120 mg/day is being tested in phase 3 trials. FUNDING: Astellas Pharma, National Cancer Institute (Leukemia Specialized Program of Research Excellence grant), Associazione Italiana Ricerca sul Cancro.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Pirazinas/farmacología , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética , Anciano , Compuestos de Anilina/sangre , Compuestos de Anilina/uso terapéutico , Antineoplásicos/sangre , Antineoplásicos/uso terapéutico , Plaquetas , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Pirazinas/sangre , Pirazinas/uso terapéutico , Recurrencia , Retratamiento , Tirosina Quinasa 3 Similar a fms/metabolismo
13.
Blood ; 125(15): 2336-43, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25696919

RESUMEN

Small-molecule inhibitors of kinases involved in B-cell receptor signaling are an important advance in managing lymphoid malignancies. Entospletinib (GS-9973) is an oral, selective inhibitor of spleen tyrosine kinase. This multicenter, phase 2 study enrolled subjects with relapsed or refractory chronic lymphocytic leukemia (CLL; n = 41) or non-Hodgkin lymphoma (n = 145). Participants received 800 mg entospletinib twice daily. We report efficacy outcomes in the CLL cohort (n = 41) and safety outcomes in all cohorts (N = 186). The primary end point was a progression-free survival (PFS) rate at 24 weeks in subjects with CLL. The PFS rate at 24 weeks was 70.1% (95% confidence interval [CI], 51.3%-82.7%); median PFS was 13.8 months (95% CI, 7.7 months to not reached). The objective response rate was 61.0% (95% CI, 44.5%-75.8%), including 3 subjects (7.3%) who achieved nodal response with persistent lymphocytosis. Fifty-four subjects (29.0%) had serious adverse events (SAEs). The most common treatment-emergent SAEs included dyspnea, pneumonia, febrile neutropenia, dehydration, and pyrexia. Common grade 3/4 laboratory abnormalities included neutropenia (14.5%) and reversible alanine aminotransferase/aspartate aminotransferase elevations (13.4%). Entospletinib demonstrates clinical activity in subjects with relapsed or refractory CLL with acceptable toxicity. This trial was registered at www.clinicaltrials.gov as #NCT01799889.


Asunto(s)
Indazoles/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazinas/uso terapéutico , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Humanos , Indazoles/efectos adversos , Indazoles/sangre , Leucemia Linfocítica Crónica de Células B/enzimología , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/sangre , Pirazinas/efectos adversos , Pirazinas/sangre , Quinasa Syk
14.
Br J Clin Pharmacol ; 83(12): 2778-2788, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28715853

RESUMEN

AIMS: Based on in vitro data, there is evidence to suggest that cytochrome P450 (CYP) 2C8 is involved in the metabolism of selexipag and its active metabolite, ACT-333679. The present study evaluated the possible pharmacokinetic interactions of selexipag with gemfibrozil, a strong CYP2C8 inhibitor, and rifampicin, an inducer of CYP2C8. METHODS: The study consisted of two independent parts, each conducted according to an open-label, randomized, crossover design. The pharmacokinetics and safety of selexipag and ACT-333679 were studied following single-dose administration either alone or in the presence of multiple-dose gemfibrozil (part I) or rifampicin (part II) in healthy male subjects. RESULTS: Gemfibrozil had comparatively small effects on selexipag (less than 2-fold difference in any pharmacokinetic variable) but, with respect to ACT-333679, increased the maximum plasma concentration (Cmax ) 3.6-fold [90% confidence interval (CI) 3.1, 4.3] and the area under the plasma concentration-time curve from zero to infinity (AUC0-∞ ) 11.1-fold (90% CI 9.2, 13.4). The marked increased exposure to ACT-333679, which mediates the majority of the pharmacological activity of selexipag, was accompanied by significantly more adverse events such as headache, nausea and vomiting. Coadministration of rifampicin increased the Cmax of selexipag 1.8-fold (90% CI 1.4, 2.2) and its AUC0-∞ 1.3-fold (90% CI 1.1, 1.4); its effects on ACT-333679 were to increase its Cmax 1.3-fold (90% CI 1.1, 1.6), shorten its half-life by 63% and reduce its AUC0-∞ by half (90% CI 0.45, 0.59). CONCLUSION: Concomitant administration of selexipag and strong inhibitors of CYP2C8 must be avoided, whereas when coadministered with inducers of CYP2C8, dose adjustments of selexipag should be envisaged.


Asunto(s)
Acetamidas/farmacocinética , Acetatos/farmacocinética , Antihipertensivos/farmacocinética , Inductores del Citocromo P-450 CYP2C8/administración & dosificación , Inhibidores del Citocromo P-450 CYP2C8/administración & dosificación , Gemfibrozilo/administración & dosificación , Profármacos/farmacocinética , Pirazinas/farmacocinética , Rifampin/administración & dosificación , Acetamidas/administración & dosificación , Acetamidas/efectos adversos , Acetamidas/sangre , Acetatos/administración & dosificación , Acetatos/efectos adversos , Acetatos/sangre , Activación Metabólica , Adolescente , Adulto , Antihipertensivos/administración & dosificación , Antihipertensivos/efectos adversos , Antihipertensivos/sangre , Área Bajo la Curva , Estudios Cruzados , Citocromo P-450 CYP2C8/metabolismo , Inductores del Citocromo P-450 CYP2C8/efectos adversos , Inhibidores del Citocromo P-450 CYP2C8/efectos adversos , Interacciones Farmacológicas , Gemfibrozilo/efectos adversos , Alemania , Semivida , Voluntarios Sanos , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Profármacos/administración & dosificación , Profármacos/efectos adversos , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Pirazinas/sangre , Rifampin/efectos adversos , Medición de Riesgo , Adulto Joven
15.
Eur J Clin Pharmacol ; 73(2): 151-156, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27885399

RESUMEN

PURPOSE: The aim of this single-center, open-label study was to assess the absolute bioavailability of an oral (tablet) versus intravenous (i.v.) formulation of selexipag in healthy subjects. METHODS: A pilot phase in three healthy male subjects, which preceded the main study, consisted of a single 20-minute i.v. infusion of 50 µg selexipag. Its objectives were to ensure the safety of the i.v. formulation and to select the i.v. dose for the main study. The main study had a randomized, two-way crossover design in 16 healthy male subjects. Subjects received a single oral dose of 400 µg selexipag and a single 80-minute i.v. infusion of 200 µg selexipag. RESULTS: Three subjects in the pilot and 15 in the main phase completed the study as planned, whereas one subject of the main study withdrew the consent. A geometric mean total body clearance (95% confidence interval (CI)) of 17.9 L/h (15.0-21.5) and a volume of distribution of 11.7 L (10.6-13.0) were determined. The absolute oral bioavailability of selexipag (90% CI) was 49.4% (42.6-57.2). Selexipag was well-tolerated; no adverse event (AE) occurred during the pilot phase, and the main observed AEs were headache, nausea, and vomiting. CONCLUSION: A single i.v. administration of selexipag in healthy subjects was safe and well-tolerated. The bioavailability of selexipag after oral administration is approximately 50%.


Asunto(s)
Acetamidas/farmacocinética , Antihipertensivos/farmacocinética , Pirazinas/farmacocinética , Receptores de Prostaglandina/agonistas , Acetamidas/efectos adversos , Acetamidas/sangre , Acetatos/sangre , Administración Oral , Adulto , Antihipertensivos/efectos adversos , Antihipertensivos/sangre , Disponibilidad Biológica , Estudios Cruzados , Humanos , Infusiones Intravenosas , Masculino , Pirazinas/efectos adversos , Pirazinas/sangre , Receptores de Epoprostenol , Adulto Joven
16.
Eur J Clin Pharmacol ; 73(9): 1121-1128, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28639119

RESUMEN

PURPOSE: In vitro data showed that selexipag and its active metabolite (ACT-333679) have an inductive effect on CYP3A4, CYP2B6, and CYP2C9 at concentrations approximately 100-fold higher than the maximum plasma concentration (C max) measured under steady-state conditions. In order to confirm in vivo the lack of induction at the enterocyte level, we assessed the effect of selexipag on midazolam, a substrate of hepatic and intestinal CYP3A4. METHODS: This study was conducted according to an open-label, randomized, two-way crossover design. A total of 20 subjects received a single oral dose of 7.5 mg midazolam alone (treatment A) or on top of steady-state selexipag (treatment B). Selexipag was administered twice daily using an up-titration scheme consisting of three steps: 400, 600, 1000, and 1600 µg with increments every fourth day. A 24-h pharmacokinetic profile was performed following midazolam administration, and bioequivalence criteria were investigated on an exploratory basis. RESULTS: The C max of midazolam and 1-hydroxymidazolam was decreased by approximately 20 and 14%, respectively, following treatment B compared to A. The time to reach C max for midazolam and 1-hydroxymidazolam was similar between treatments. The terminal half-life was reduced in treatment B compared to A for both midazolam (16%) and 1-hydroxymidazolam (20%). Exposure (area under the curve) to midazolam and 1-hydroxymidazolam was similar between treatments, and the 90% confidence intervals of geometric mean ratios were within the bioequivalence interval. Treatment with midazolam, selexipag, and the combination was safe and well tolerated. CONCLUSION: Exposure to midazolam and 1-hydroxymidazolam was not affected by treatment with selexipag.


Asunto(s)
Acetamidas/farmacocinética , Midazolam/farmacocinética , Pirazinas/farmacocinética , Acetamidas/efectos adversos , Acetamidas/sangre , Acetamidas/farmacología , Adulto , Antihipertensivos/efectos adversos , Antihipertensivos/sangre , Antihipertensivos/farmacocinética , Antihipertensivos/farmacología , Área Bajo la Curva , Estudios Cruzados , Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Semivida , Voluntarios Sanos , Humanos , Masculino , Midazolam/efectos adversos , Midazolam/sangre , Midazolam/farmacología , Pirazinas/efectos adversos , Pirazinas/sangre , Pirazinas/farmacología , Adulto Joven
17.
J Sep Sci ; 40(19): 3782-3791, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28749011

RESUMEN

A rapid, simple, and accurate procedure was developed and validated for the simultaneous quantification of two anticancer agents, volitinib and gefitinib in rat plasma by high-performance liquid chromatography with tandem mass spectrometry. The samples were separated by gradient elution from a cyano column within five minutes, using 0.1% formic acid in acetonitrile and 10 mM ammonium formate solution (pH 3.0) as mobile phase. When plasma samples were deproteinated by adding methanol, the analytes in the extract were detected in the positive ionization mode with the tracer ion mass of 346.1 → 145.1 for volitinib and 446.8 → 128.1 for gefitinib. The assay was determined to be valid in the concentration ranges of 2 to 1000 ng/mL for volitinib, and of 1 to 500 ng/mL for gefitinib. Intra- and interday accuracies ranged from 88.0 to 104.7% for volitinib and from 90.3 to 101%, for gefitinib. The precision of the assay ranged from 2.1 to 9.71% for volitinib and 2.31 to 12.1% for gefitinib. This method was successfully applied to a pharmacokinetic study of volitinib and gefitinib after the administration of an intravenous or oral dose, indicating that the developed assay can be used to simultaneously determine the concentrations of volitinib and gefitinib in rat plasma.


Asunto(s)
Pirazinas/sangre , Quinazolinas/sangre , Triazinas/sangre , Animales , Cromatografía Líquida de Alta Presión , Gefitinib , Pirazinas/farmacocinética , Quinazolinas/farmacocinética , Ratas , Espectrometría de Masas en Tándem , Triazinas/farmacocinética
20.
Drug Metab Dispos ; 43(10): 1441-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26203069

RESUMEN

This study focused on the mechanistic interpretation of ex vivo oxidation of a candidate drug in blood plasma samples. An unexpected lipid peroxide-mediated epoxidation followed by a dramatic rearrangement led to production of a five-membered oxazole derivative from the original six-membered pyrazinone-carboxamide core of a human neutrophil elastase inhibitor, 6-(1-(4-cyanophenyl)-1H-pyrazol-5-yl)-N-ethyl-5-methyl-3-oxo-4-(3-(trifluoromethyl)phenyl)-3,4-dihydropyrazine-2-carboxamide (AZD9819). The rearranged oxidation product 2-(1-(4-cyanophenyl)-1H-pyrazol-5-yl)-5-(N-ethylacetamido)-N-(3-(trifluoromethyl)phenyl)oxazole-4-carboxamide was characterized by accurate-mass tandem mass spectrometry fragmentations, by two-dimensional NMR and X-ray crystallography of an authentic standard, and by incorporation of an (18)O atom from molecular (18)O2 to the location predicted by our proposed mechanism. The lipid peroxide-mediated oxidation was demonstrated by using human low-density lipoprotein (LDL) in pH 7.4 phosphate buffer and by inhibiting the oxidation with ascorbic acid or l-glutathione, two antioxidants effective in both plasma and the LDL incubation. A nucleophilic mechanism for the epoxidation of AZD9819 by lipid hydroperoxides explains the prevention of its ex vivo oxidation by acidification of the plasma samples. The discovery of the lipid peroxide-dependent oxidation of an analyte and the means of prevention could provide valuable information for biotransformation and bioanalysis.


Asunto(s)
Peróxidos Lipídicos/sangre , Proteínas Inhibidoras de Proteinasas Secretoras/sangre , Proteínas Inhibidoras de Proteinasas Secretoras/química , Amidas/sangre , Amidas/química , Animales , Femenino , Humanos , Peróxidos Lipídicos/química , Masculino , Oxidación-Reducción , Pirazinas/sangre , Pirazinas/química , Pirazoles/sangre , Pirazoles/química , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA