Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 446
Filtrar
1.
Clin Pharmacol Ther ; 115(6): 1336-1345, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38404228

RESUMEN

Daprodustat is the first oral hypoxia-inducible factor prolyl hydroxylase inhibitor approved recently for the treatment of anemia caused by chronic kidney disease (CKD) in adults receiving dialysis. We evaluated the role of organic anion transporting polypeptide (OATP)1B-mediated hepatic uptake transport in the pharmacokinetics (PKs) of daprodustat using in vitro and in vivo studies, and physiologically-based PK (PBPK) modeling of its drug-drug interactions (DDIs) with inhibitor drugs. In vitro, daprodustat showed specific transport by OATP1B1/1B3 in the transfected cell systems and primary human and monkey hepatocytes. A single-dose oral rifampin (OATP1B inhibitor) reduced daprodustat intravenous clearance by a notable 9.9 ± 1.2-fold (P < 0.05) in cynomolgus monkeys. Correspondingly, volume of distribution at steady-state was also reduced by 5.0 ± 1.1-fold, whereas the half-life change was minimal (1.5-fold), corroborating daprodustat hepatic uptake inhibition by rifampin. A PBPK model accounting for OATP1B-CYP2C8 interplay was developed, which well described daprodustat PK and DDIs with gemfibrozil (CYP2C8 and OATP1B inhibitor) and trimethoprim (weak CYP2C8 inhibitor) within 25% error of the observed data in healthy subjects. About 18-fold increase in daprodustat area under the curve (AUC) following gemfibrozil treatment was found to be associated with strong CYP2C8 inhibition and moderate OATP1B inhibition. Moreover, PK modulation in hepatic dysfunction and subjects with CKD, in comparison to healthy control, was well-captured by the model. CYP2C8 and/or OATP1B inhibitor drugs (e.g., gemfibrozil, clopidogrel, rifampin, and cyclosporine) were predicted to perpetrate moderate-to-strong DDIs in healthy subjects, as well as, in target CKD population. Daprodustat can be used as a sensitive CYP2C8 index substrate in the absence of OATP1B modulation.


Asunto(s)
Citocromo P-450 CYP2C8 , Interacciones Farmacológicas , Hepatocitos , Transportador 1 de Anión Orgánico Específico del Hígado , Insuficiencia Renal Crónica , Rifampin , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Adulto , Animales , Femenino , Humanos , Masculino , Citocromo P-450 CYP2C8/metabolismo , Inhibidores del Citocromo P-450 CYP2C8/farmacocinética , Glicina/análogos & derivados , Glicina/farmacocinética , Células HEK293 , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Hepatopatías/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/antagonistas & inhibidores , Macaca fascicularis , Insuficiencia Renal Crónica/metabolismo , Rifampin/farmacología , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/antagonistas & inhibidores
2.
Cancer Chemother Pharmacol ; 93(5): 471-479, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38278871

RESUMEN

PURPOSE: Report pharmacokinetic (PK)/pharmacodynamic (PD) findings from the phase III ClarIDHy study and any association between PK/PD parameters and treatment outcomes in this population. METHODS: Patients with mutant isocitrate dehydrogenase 1 (mIDH1) advanced cholangiocarcinoma were randomized at a 2:1 ratio to receive ivosidenib or matched placebo. Crossover from placebo to ivosidenib was permitted at radiographic disease progression. Blood samples for PK/PD analyses, a secondary endpoint, were collected pre-dose and up to 4 h post-dose on day (D) 1 of cycles (C) 1 - 2, pre-dose and 2 h post-dose on D15 of C1 - 2, and pre-dose on D1 from C3 onwards. Plasma ivosidenib and D-2-hydroxyglutarate (2-HG) were measured using liquid chromatography-tandem mass spectrometry. All clinical responses were centrally reviewed previously. RESULTS: PK/PD analysis was available for samples from 156 ivosidenib-treated patients. Ivosidenib was absorbed rapidly following single and multiple oral doses (time of maximum observed plasma concentration [Tmax] of 2.63 and 2.07 h, respectively). Ivosidenib exposure was higher at C2D1 than after a single dose, with low accumulation. In ivosidenib-treated patients, mean plasma 2-HG concentration was reduced from 1108 ng/mL at baseline to 97.7 ng/mL at C2D1, close to levels previously observed in healthy individuals. An average 2-HG inhibition of 75.0% was observed at steady state. No plasma 2-HG decreases were seen with placebo. Plasma 2-HG reductions were observed in ivosidenib-treated patients irrespective of best overall response (progressive disease, or partial response and stable disease). CONCLUSION: Once-daily ivosidenib 500 mg has a favorable PK/PD profile, attesting the 2-HG reduction mechanism of action and, thus, positive outcomes in treated patients with advanced mIDH1 cholangiocarcinoma. CLINICAL TRIAL REGISTRATION: NCT02989857 Registered February 20, 2017.


Asunto(s)
Antineoplásicos , Neoplasias de los Conductos Biliares , Colangiocarcinoma , Glicina , Glicina/análogos & derivados , Isocitrato Deshidrogenasa , Mutación , Piridinas , Humanos , Colangiocarcinoma/tratamiento farmacológico , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Glicina/farmacocinética , Glicina/administración & dosificación , Glicina/uso terapéutico , Glicina/farmacología , Piridinas/farmacocinética , Piridinas/administración & dosificación , Piridinas/farmacología , Piridinas/uso terapéutico , Masculino , Persona de Mediana Edad , Femenino , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/genética , Anciano , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Adulto , Método Doble Ciego , Anciano de 80 o más Años , Estudios Cruzados , Resultado del Tratamiento
3.
Clin Pharmacol Drug Dev ; 11(5): 562-575, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35355447

RESUMEN

Daprodustat is a hypoxia-inducible factor-prolyl hydroxylase inhibitor in development for treatment of anemia of chronic kidney disease. We evaluated the role of hepatic impairment on daprodustat pharmacokinetics, pharmacodynamics, and tolerability. Participants with mild (Child-Pugh Class A, score 5-6) and moderate (Child-Pugh Class B, score 7-9) hepatic impairment and matched healthy controls were administered single 6-mg doses of daprodustat. Exposure parameters were determined for daprodustat and its six metabolites. Comparisons resulted in 1.5- and 2.0-fold higher daprodustat Cmax and area under the curve (AUC) exposures in participants with mild and moderate hepatic impairment, respectively, versus controls; Cmax in mild hepatic impairment was comparable to controls. Similarly, aligned with parent drug, unbound daprodustat Cmax and AUC exposures increased 1.6- to 2.3-fold in hepatic-impaired participants versus controls, and metabolite exposures were 1.2- to 2.0-fold higher in participants with hepatic impairment. Erythropoeitin (EPO) baseline-corrected AUC exposures were between 0.3-fold lower and 2.2-fold higher in matched controls versus hepatic-impaired participants. No serious or study drug-related adverse events were reported. Daprodustat exposure was increased in participants with moderate and mild hepatic impairment compared with matched controls; however, no meaningful differences in EPO were observed and no new safety concerns were identified (ClinicalTrials.gov: NCT03223337).


Asunto(s)
Hepatopatías , Inhibidores de Prolil-Hidroxilasa , Barbitúricos , Femenino , Glicina/efectos adversos , Glicina/análogos & derivados , Glicina/farmacocinética , Humanos , Hepatopatías/metabolismo , Masculino , Inhibidores de Prolil-Hidroxilasa/efectos adversos , Inhibidores de Prolil-Hidroxilasa/farmacocinética
4.
Drug Metab Dispos ; 50(1): 58-64, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34670777

RESUMEN

Hepatobiliary imaging is increasingly used by pharmacologists to quantify liver concentrations of transporter-dependent drugs. However, liver imaging does not quantify concentrations in extracellular space, hepatocytes, and bile canaliculi. Our study compared the compartmental distribution of two hepatobiliary substrates gadobenate dimeglumine [BOPTA; 0.08 liver extraction ratio (ER)] and mebrofenin (MEB; 0.93 ER) in a model of perfused rat liver. A gamma counter placed over livers measured liver concentrations. Livers were preperfused with gadopentetate dimeglumine to measure extracellular concentrations. Concentrations coming from bile canaliculi and hepatocytes were calculated. Transporter activities were assessed by concentration ratios between compartments and pharmacokinetic parameters that describe the accumulation and decay profiles of hepatocyte concentrations. The high liver concentrations of MEB relied mainly on hepatocyte and bile canaliculi concentrations. In contrast, the three compartments contributed to the low liver concentrations obtained during BOPTA perfusion. Nonlinear regression analysis of substrate accumulation in hepatocytes revealed that cellular efflux is measurable ∼4 minutes after the start of perfusion. The hepatocyte-to-extracellular concentration ratio measured at this time point was much higher during MEB perfusion. BOPTA transport by multidrug resistance associated protein 2 induced an aquaporin-mediated water transport, whereas MEB transport did not. BOPTA clearance from hepatocytes to bile canaliculi was higher than MEB clearance. MEB did not efflux back to sinusoids, whereas BOPTA basolateral efflux contributed to the decrease in hepatocyte concentrations. In conclusion, our ex vivo model quantifies substrate compartmental distribution and transport across hepatocyte membranes and provides an additional understanding of substrate distribution in the liver. SIGNIFICANCE STATEMENT: When transporter-dependent drugs target hepatocytes, cellular concentrations are important to investigate. Low concentrations on cellular targets impair drug therapeutic effects, whereas excessive hepatocyte concentrations may induce cellular toxicity. With a gamma counter placed over rat perfused livers, we measured substrate concentrations in the extracellular space, hepatocytes, and bile canaliculi. Transport across hepatocyte membranes was calculated. The study provides an additional understanding of substrate distribution in the liver.


Asunto(s)
Medios de Contraste/farmacocinética , Hígado/diagnóstico por imagen , Hígado/metabolismo , Compuestos de Anilina/farmacocinética , Animales , Canalículos Biliares/metabolismo , Sistema Biliar/diagnóstico por imagen , Diagnóstico por Imagen , Espacio Extracelular/metabolismo , Genes erbB-2/genética , Glicina/farmacocinética , Hepatocitos/metabolismo , Técnicas In Vitro , Masculino , Modelos Biológicos , Dinámicas no Lineales , Ratas , Ratas Sprague-Dawley
5.
Clin Pharmacokinet ; 61(3): 347-362, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34905154

RESUMEN

The pharmacokinetics of roxadustat are well characterized, with an apparent volume of distribution after oral administration of 22-57 L, apparent clearance of 1.2-2.65 L/h, and renal clearance of 0.030-0.026 L/h in healthy volunteers; the elimination half-life is 9.6-16 h. Plasma binding is 99% and the fraction eliminated by hemodialysis is 2.34%. As an interpretation of the pharmacodynamics of roxadustat, we proposed a concept with a hypothetical cascade of two subsequent effects, first on erythropoetin (EPO) and second on hemoglobin (delta Hb). The primary effect on EPO is observed within a few hours after roxadustat administration and can be modeled using the sigmoidal Hill equation. The concentration at half-maximum effect can be inferred at 10-36 µg/mL, the Hill coefficient at 3.3, and the effect bisection time at 10-17 h, corresponding to EPO half-life. The subsequent effect on hemoglobin (delta Hb) is observed after several weeks and can be interpreted as an irreversible, dose proportional, unsaturable effect, continuing in agreement with the lifespan of red blood cells of 63-112 days.


Asunto(s)
Glicina , Isoquinolinas , Glicina/análogos & derivados , Glicina/farmacocinética , Hemoglobinas , Humanos , Isoquinolinas/farmacocinética
6.
Artículo en Inglés | MEDLINE | ID: mdl-34663555

RESUMEN

99mTc-mebrofenin hepatobiliary scintigraphy with SPECT/CT (HBS-M) has become an important quantitative method to evaluate global liver function and future liver remnant (FLR) function in patients who are candidates for resective liver surgery. The purpose of this work was to describe the method in the prediction of post-surgical liver failure. The overall liver function and that of the FLR are obtained by analysis of the initial dynamic phase of the scan. Liver volume to be preserved is expressed as a percentage of the total liver volume measured in both CT sections. HBS-M is able to accurately gauge regional liver function abnormalities that could be represented as normal liver tissue parenchyma in the CT study. This technique can provide very valuable prognostic information for the estimation of the postoperative risk of liver failure in all patients who are candidates for resective liver surgery.


Asunto(s)
Compuestos de Anilina/farmacocinética , Glicina/farmacocinética , Hepatectomía/efectos adversos , Fallo Hepático/diagnóstico por imagen , Hígado/metabolismo , Compuestos de Organotecnecio/farmacocinética , Complicaciones Posoperatorias/diagnóstico por imagen , Radiofármacos/farmacocinética , Compuestos de Anilina/sangre , Glicina/sangre , Humanos , Hígado/anatomía & histología , Hígado/diagnóstico por imagen , Tasa de Depuración Metabólica , Tamaño de los Órganos , Factores de Tiempo , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada por Rayos X
7.
Drug Metab Dispos ; 49(10): 870-881, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34321251

RESUMEN

Point mutations in isocitrate dehydrogenase 1 (IDH1) result in conversion of α-ketoglutarate to the oncometabolite, d-2-hydroxyglutarate (2-HG). Ivosidenib is a once daily (QD), orally available, potent, mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor approved for the treatment of patients with relapsed or refractory acute myeloid leukemia (AML) and intensive chemotherapy-ineligible newly diagnosed AML, with a susceptible IDH1 mutation. We characterized the protein binding, metabolism, metabolites, cell permeability, and drug-drug interaction potential of ivosidenib in humans, monkeys, dogs, rats, and/or mice in in vitro experiments. In vivo pharmacokinetic (PK) profiling and assessment of drug distribution and excretion was undertaken in rats, dogs, and monkeys administered single-dose ivosidenib. The PK/pharmacodynamic (PD) relationship between ivosidenib and 2-HG was analyzed in an mIDH1 xenograft mouse model. Ivosidenib was well absorbed, showed low clearance, and moderate to long terminal half-life (5.3-18.5 hours) in rats, dogs, and monkeys. Brain to plasma exposure ratio was low (2.3%), plasma protein binding was high, and oxidative metabolism was the major elimination pathway. Ivosidenib had high cell permeability and was identified as a substrate for P-glycoprotein. There was moderate induction of cytochrome P450 (P450) enzymes CYP3A4 and CYP2B6 but minimal P450 inhibition or autoinduction. Tumor 2-HG reduction appeared to be dose- and drug-exposure-dependent. Ivosidenib showed a favorable PK profile in several animal species, along with a clear PK/PD relationship demonstrating 2-HG inhibition that translated well to patients with AML. SIGNIFICANCE STATEMENT: Ivosidenib is a mutant IDH1 (mIDH1) inhibitor approved for the treatment of certain patients with mIDH1 acute myeloid leukemia. In Sprague-Dawley rats, beagle dogs, and cynomolgus monkeys, ivosidenib demonstrated a favorable pharmacokinetic profile, and in female BALB/c mice showed clear dose- and exposure-dependent inhibition of the oncometabolite, d-2-hydroxyglutarate, which is present at abnormal levels in mIDH1 tumors. These findings led to the further development of ivosidenib and are consistent with data from patients with mIDH1 cancers and healthy participants.


Asunto(s)
Glicina/análogos & derivados , Isocitrato Deshidrogenasa/metabolismo , Leucemia Mieloide Aguda , Piridinas/farmacocinética , Animales , Antineoplásicos/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Vías de Eliminación de Fármacos , Interacciones Farmacológicas , Glutaratos/metabolismo , Glicina/farmacocinética , Haplorrinos , Humanos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Ácidos Cetoglutáricos/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Tasa de Depuración Metabólica , Ratones , Mutación Puntual , Unión Proteica , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
CPT Pharmacometrics Syst Pharmacol ; 10(6): 577-588, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33822485

RESUMEN

Ivosidenib is a potent, targeted, orally active, small-molecule inhibitor of mutant isocitrate dehydrogenase 1 (IDH1) that has been approved in the United States for the treatment of adults with newly diagnosed acute myeloid leukemia (AML) who are greater than or equal to 75 years of age or ineligible for intensive chemotherapy, and those with relapsed or refractory AML, with a susceptible IDH1 mutation. Ivosidenib is an inducer of the CYP2B6, CYP2C8, CYP2C9, and CYP3A4 and an inhibitor of P-glycoprotein (P-gp), organic anion transporting polypeptide-1B1/1B3 (OATP1B1/1B3), and organic anion transporter-3 (OAT3) in vitro. A physiologically-based pharmacokinetic (PK) model was developed to predict drug-drug interactions (DDIs) of ivosidenib in patients with AML. The in vivo CYP3A4 induction effect of ivosidenib was quantified using 4ß-hydroxycholesterol and was subsequently verified with the PK data from an ivosidenib and venetoclax combination study. The verified model was prospectively applied to assess the effect of multiple doses of ivosidenib on a sensitive CYP3A4 substrate, midazolam. The simulated midazolam geometric mean area under the curve (AUC) and maximum plasma concentration (Cmax ) ratios were 0.18 and 0.27, respectively, suggesting ivosidenib is a strong inducer. The model was also used to predict the DDIs of ivosidenib with CYP2B6, CYP2C8, CYP2C9, P-gp, OATP1B1/1B3, and OAT3 substrates. The AUC ratios following multiple doses of ivosidenib and a single dose of CYP2B6 (bupropion), CYP2C8 (repaglinide), CYP2C9 (warfarin), P-gp (digoxin), OATP1B1/1B3 (rosuvastatin), and OAT3 (methotrexate) substrates were 0.90, 0.52, 0.84, 1.01, 1.02, and 1.27, respectively. Finally, in accordance with regulatory guidelines, the Simcyp modeling platform was qualified to predict CYP3A4 induction using known inducers and sensitive substrates.


Asunto(s)
Antineoplásicos/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Inductores de las Enzimas del Citocromo P-450/farmacocinética , Glicina/análogos & derivados , Leucemia Mieloide Aguda/metabolismo , Modelos Biológicos , Piridinas/farmacocinética , Antineoplásicos/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Células CACO-2 , Inductores de las Enzimas del Citocromo P-450/administración & dosificación , Interacciones Farmacológicas , Femenino , Glicina/administración & dosificación , Glicina/farmacocinética , Células HEK293 , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Midazolam/farmacocinética , Piridinas/administración & dosificación , Sulfonamidas/farmacocinética
9.
Pharmacol Res ; 167: 105537, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33684510

RESUMEN

Preclinical and clinical investigation on proteasome as a druggable target in cancer has led to the development of proteasome inhibitors (PIs) with different pharmacodynamic and pharmacokinetic properties. For example, carfilzomib has a better safety profile and a lower risk of clinically relevant drug-drug interactions than bortezomib, whereas ixazomib can be orally administered on a weekly basis due to a very long elimination half-life and high systemic exposure. The purpose of this review article is to elucidate the quantitative and qualitative differences in potency, selectivity, pharmacokinetics, safety and drug-drug interactions of clinically validated PIs to provide useful information for their clinical use in real life setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Compuestos de Boro/efectos adversos , Compuestos de Boro/farmacocinética , Compuestos de Boro/farmacología , Compuestos de Boro/uso terapéutico , Bortezomib/efectos adversos , Bortezomib/farmacocinética , Bortezomib/farmacología , Bortezomib/uso terapéutico , Interacciones Farmacológicas , Glicina/efectos adversos , Glicina/análogos & derivados , Glicina/farmacocinética , Glicina/farmacología , Glicina/uso terapéutico , Humanos , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/efectos adversos , Oligopéptidos/farmacocinética , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/efectos adversos , Inhibidores de Proteasoma/farmacocinética , Inhibidores de Proteasoma/farmacología
10.
Clin Pharmacol Drug Dev ; 10(8): 950-958, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33661566

RESUMEN

Vadadustat is a hypoxia-inducible factor prolyl hydroxylase inhibitor in development for the treatment of anemia of chronic kidney disease. This phase 1, open-label, parallel-group, single-dose study evaluated the pharmacokinetics of 450-mg vadadustat in adults with moderate hepatic impairment (Child-Pugh class B) vs those with normal hepatic function. Primary end points were area under the plasma concentration-time curve (AUC) from dosing to last concentration and to infinity, as well as maximum concentration (Cmax ); additional pharmacokinetic parameters included time to Cmax (Tmax ) and half-life. Safety and tolerability were also assessed. All enrolled participants (n = 16) completed the study. Demographics were similar in both groups (overall, 100% White; 62.5% female; mean age, 59.2 years). Vadadustat plasma exposure was higher in the moderate hepatic impairment group, whereas maximum concentration was similar between groups. Point estimates of the hepatic impairment : normal geometric mean ratios (90% confidence interval) for AUC from dosing to last concentration, AUC from dosing to infinity, and Cmax were 1.05 (0.82-1.35), 1.06 (0.82-1.36), and 1.02 (0.79-1.32), respectively. Mean elimination half-life was 5.8 and 7.8 hours in the normal and hepatic impairment groups, respectively. Treatment-emergent adverse events were mostly mild in severity, and vadadustat was generally well tolerated. In conclusion, moderate hepatic impairment did not significantly impact vadadustat systemic exposure, and mild hepatic impairment is unlikely to alter vadadustat exposure.


Asunto(s)
Ayuno/sangre , Glicina/análogos & derivados , Hepatopatías/fisiopatología , Ácidos Picolínicos/administración & dosificación , Inhibidores de Prolil-Hidroxilasa/administración & dosificación , Administración Oral , Anciano , Área Bajo la Curva , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/farmacocinética , Humanos , Masculino , Persona de Mediana Edad , Ácidos Picolínicos/efectos adversos , Ácidos Picolínicos/farmacocinética , Inhibidores de Prolil-Hidroxilasa/efectos adversos , Inhibidores de Prolil-Hidroxilasa/farmacocinética
11.
Clin Transl Sci ; 14(3): 942-953, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33493392

RESUMEN

Ivosidenib is a once daily (q.d.), orally available, potent mutant isocitrate dehydrogenase 1 (mIDH1) inhibitor approved for treatment of patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) and intensive chemotherapy ineligible AML with a susceptible IDH1 mutation. Population pharmacokinetics (PKs; N = 253), exposure-response (efficacy [n = 201] and safety [n = 253]), and concentration-corrected electrocardiogram QT interval (QTc; n = 171) analyses were performed using phase I data (100 mg twice daily and 300-1200 mg q.d.). Ivosidenib disposition was well-described by a two-compartment PK model with first-order absorption and elimination. Between-subject variability was moderate for PK parameters. Intrinsic factors did not affect ivosidenib PKs. Moderate/strong CYP3A4 inhibitors increased the area under the plasma ivosidenib concentration-time curve at steady state (AUCss ) by 60%. Efficacy responders and nonresponders had similar ivosidenib exposures. Based on AUCss , there was no apparent relationship between ivosidenib exposure and efficacy or adverse events. The plasma ivosidenib concentration-QT analysis showed a mean change in QTc using Fridericia's method (ΔQTcF) of 17.2 msec at the approved 500 mg q.d. dose. Because of the direct association between ivosidenib exposure and QTcF, patients should have their electrocardiograms and electrolytes monitored, and comedications that increase ivosidenib exposure or prolong the QT interval should be avoided. These model-based analyses quantitatively provide a framework to describe the relationship among ivosidenib dose, exposure, and clinical end points. With precautions for QTc prolongation, the exposure-response analyses support the 500 mg q.d. dose in patients with AML with a susceptible IDH1 mutation.


Asunto(s)
Antineoplásicos/farmacocinética , Glicina/análogos & derivados , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndrome de QT Prolongado/diagnóstico , Piridinas/farmacocinética , Administración Oral , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Área Bajo la Curva , Variación Biológica Poblacional , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/administración & dosificación , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Interacciones Farmacológicas , Electrocardiografía , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/farmacocinética , Humanos , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/genética , Síndrome de QT Prolongado/inducido químicamente , Masculino , Persona de Mediana Edad , Modelos Biológicos , Mutación , Piridinas/administración & dosificación , Piridinas/efectos adversos , Distribución Tisular , Resultado del Tratamiento , Adulto Joven
12.
J Environ Sci Health B ; 56(3): 235-240, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33449826

RESUMEN

Fertiactyl® is a foliar fertilizer with the potential to minimize the phytotoxicity effects caused by glyphosate drift in eucalyptus plants. As the interactions of the glyphosate and Fertiactyl® in tank mix on the plant behavior are not yet known, the objective was to evaluate the absorption and translocation of 14C-glyphosate, applied isolated and mixed in tank with Fertiactyl®, in young eucalyptus plants (clone I-144, Eucalyptus urophylla x E. grandis). The addition of Fertiactyl® to the mixture of 14C-glyphosate reduced the absorption by 94.3% in relation to the total absorbed at the end of the evaluation compared to plants treated only with 14C-glyphosate, i.e., Fertiactyl® protected the eucalyptus plants of the glyphosate intoxication by drift. The translocation rates from the treated leaves to the rest of the shoots and roots were low (<2% of the total recovered) in both treatments, suggest that restricted translocation is a mechanism of natural tolerance to glyphosate in plants of clone I-144. It is concluded that Fertiactyl®, mixed in the solution with glyphosate, protects young eucalyptus plants against glyphosate drift by reducing the amount of herbicide absorbed.


Asunto(s)
Eucalyptus/efectos de los fármacos , Fertilizantes , Glicina/análogos & derivados , Herbicidas/farmacocinética , Radioisótopos de Carbono/farmacocinética , Eucalyptus/metabolismo , Glicina/farmacocinética , Glicina/toxicidad , Herbicidas/toxicidad , Hojas de la Planta/efectos de los fármacos , Brotes de la Planta/efectos de los fármacos , Distribución Tisular , Glifosato
13.
J Environ Sci Health B ; 56(1): 10-15, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33043792

RESUMEN

The application of glyphosate to coffee crops can cause injuries to plants. Fertiactyl® foliar fertilizer reduces injuries when mixed with glyphosate; however, it is important to establish which mechanisms are responsible for this protective action. This study aimed to evaluate the absorption and translocation of glyphosate applied separately and in mixture with Fertiactyl® in coffee seedlings. Absorption and translocation were performed with 14C-glyphosate applied separately and in mixture with Fertiactyl® at 0, 6, 12, 24, 48, 96, and 144 hours after application (HAA). Most of the 14C-glyphosate applied to coffee seedlings was not absorbed. The 14C-glyphosate applied separately had a higher absorption by coffee seedlings (6.5%) than in a mixture with Fertiactyl® (2.7%) at 144 HAA. The maximum translocation of the 14C-glyphosate applied separately was 0.69% at 81.2 HAA and in mixture with Fertiactyl® was 0.41% at 41.2 HAA. The treated leaves retained a higher percentage of 14C-glyphosate when applied separately (5.6% at 144 HAA) than in a mixture with Fertiactyl® (2.2% at 144 HAA). Low translocation (<1%) for the rest of the plant shoots was observed both for the 14C-glyphosate applied separately and in combination with Fertiactyl®. Therefore, Fertiactyl® decreased the absorption and translocation of 14C-glyphosate in coffee seedlings.


Asunto(s)
Coffea/metabolismo , Fertilizantes , Glicina/análogos & derivados , Herbicidas/farmacocinética , Absorción Fisiológica , Transporte Biológico , Glicina/farmacocinética , Hojas de la Planta/metabolismo , Plantones/metabolismo , Glifosato
14.
Eur J Drug Metab Pharmacokinet ; 46(1): 141-153, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33165773

RESUMEN

BACKGROUND AND OBJECTIVES: Roxadustat is an orally active hypoxia-inducible factor prolyl hydroxylase inhibitor for anemia in chronic kidney disease. The pharmacokinetics, metabolic profile, and pharmacodynamics of roxadustat were investigated in subjects with different degrees of kidney function. METHODS: This phase 1 open-label study enrolled subjects with normal and severely impaired kidney function, and end-stage renal disease (ESRD) on continuous ambulatory peritoneal dialysis (CAPD) or automated peritoneal dialysis (APD) or hemodialysis/hemodiafiltration (HD/HDF). All subjects received a single 100-mg dose of oral roxadustat. Within a single-sequence, two-treatment period design (P1/P2), subjects with ESRD on HD/HDF received roxadustat 2 h after (P1) and 2 h before (P2) a dialysis session. Area under the plasma concentration-time curve (AUC) from administration to infinity (AUCinf), maximum concentration (Cmax), and terminal elimination half-life (t1/2) were assessed for roxadustat; AUC and Cmax were assessed for erythropoietin. RESULTS: Thirty-four subjects were enrolled and received roxadustat (normal kidney function, n = 12; severely impaired kidney function, n = 9; ESRD on CAPD/APD, n = 1; ESRD on HD/HDF, n = 12). The geometric least-square mean ratio of AUCinf was 223% and 195% in subjects with severely impaired kidney function and ESRD on HD/HDF, respectively, relative to subjects with normal kidney function; Cmax and t1/2 were comparable. The pharmacokinetic profile of roxadustat was not affected by HD/HDF. AUCinf and t1/2 for the metabolites of roxadustat increased in subjects with kidney impairment. The AUC and Cmax of erythropoietin increased in subjects with severely impaired kidney function or ESRD on HD/HDF. Roxadustat was well tolerated. CONCLUSIONS: Kidney function impairment increased the AUC of roxadustat and its metabolites. The Cmax and t1/2 of roxadustat were comparable among groups. Roxadustat and its metabolites were not cleared by HD/HDF. Clinical Trials Registration Number: NCT02965040.


Asunto(s)
Glicina/análogos & derivados , Isoquinolinas/farmacocinética , Fallo Renal Crónico/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacocinética , Diálisis Renal/tendencias , Administración Oral , Adulto , Anciano , Femenino , Glicina/administración & dosificación , Glicina/farmacocinética , Humanos , Isoquinolinas/administración & dosificación , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Inhibidores de Prolil-Hidroxilasa/administración & dosificación
15.
Clin Pharmacol Drug Dev ; 10(1): 99-109, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32648303

RESUMEN

Ivosidenib, a small-molecule inhibitor of mutant isocitrate dehydrogenase 1, is primarily cleared by hepatic metabolism. This open-label study investigated the impact of hepatic impairment on ivosidenib pharmacokinetics (ClinicalTrials.gov: NCT03282513). Otherwise healthy participants with mild (n = 9) or moderate (n = 8) hepatic impairment (Child-Pugh score) and matched participants with normal hepatic function (n = 16) received 1 oral dose of 500-mg ivosidenib. Mild hepatic impairment had a negligible effect on total ivosidenib plasma exposure, with geometric mean ratios (90% confidence interval [CI]) of 0.933 (0.715-1.22) for maximum concentration (Cmax ) and 0.847 (0.624-1.15) for area under the plasma concentration-time curve (AUC) in participants with mild hepatic impairment versus matched controls. Moderate hepatic impairment reduced total ivosidenib exposure by 28% to 44%, with geometric mean ratios (90%CI) of 0.565 (0.419-0.763) for Cmax and 0.716 (0.479-1.07) for AUC, although the 90%CI for AUC included 1.00. The ivosidenib unbound fraction was concentration dependent and higher in participants with mild/moderate hepatic impairment compared with matched controls. There was no apparent trend to increasing unbound Cmax with increased hepatic impairment severity. A single 500-mg ivosidenib dose was well tolerated, with no serious or severe adverse events and no adverse events leading to discontinuation. We conclude that mild/moderate hepatic impairment did not lead to clinically relevant changes in ivosidenib exposure following a single 500-mg dose.


Asunto(s)
Antineoplásicos/farmacocinética , Glicina/análogos & derivados , Hepatopatías/metabolismo , Piridinas/farmacocinética , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/sangre , Glicina/farmacocinética , Voluntarios Sanos , Humanos , Hígado/metabolismo , Masculino , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/sangre
16.
J Med Chem ; 63(23): 14989-15012, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33205975

RESUMEN

Increasing evidence implicates the orphan G protein-coupled receptor 88 (GPR88) in a number of striatal-associated disorders. In this study, we report the design and synthesis of a series of novel (4-alkoxyphenyl)glycinamides (e.g., 31) and the corresponding 1,3,4-oxadiazole bioisosteres derived from the 2-AMPP scaffold (1) as GPR88 agonists. The 5-amino-1,3,4-oxadiazole derivatives (84, 88-90) had significantly improved potency and lower lipophilicity compared to 2-AMPP. Compound 84 had an EC50 of 59 nM in the GPR88 overexpressing cell-based cAMP assay. In addition, 84 had an EC50 of 942 nM in the [35S]GTPγS binding assay using mouse striatal membranes but was inactive in membranes from GPR88 knockout mice, even at a concentration of 100 µM. In vivo pharmacokinetic testing of 90 in rats revealed that the 5-amino-1,3,4-oxadiazole analogues may have limited brain permeability. Taken together, these results provide the basis for further optimization to develop a suitable agonist to probe GPR88 functions in the brain.


Asunto(s)
Glicina/análogos & derivados , Glicina/farmacología , Oxadiazoles/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Diseño de Fármacos , Glicina/farmacocinética , Masculino , Ratones Noqueados , Estructura Molecular , Oxadiazoles/síntesis química , Oxadiazoles/farmacocinética , Ratas Long-Evans , Solubilidad , Relación Estructura-Actividad
17.
Cancer Chemother Pharmacol ; 86(5): 619-632, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32978634

RESUMEN

PURPOSE: Develop a physiologically based pharmacokinetic (PBPK) model of ivosidenib using in vitro and clinical PK data from healthy participants (HPs), refine it with clinical data on ivosidenib co-administered with itraconazole, and develop a model for patients with acute myeloid leukemia (AML) and apply it to predict ivosidenib drug-drug interactions (DDI). METHODS: An HP PBPK model was developed in Simcyp Population-Based Simulator (version 15.1), with the CYP3A4 component refined based on a clinical DDI study. A separate model accounting for the reduced apparent oral clearance in patients with AML was used to assess the DDI potential of ivosidenib as the victim of CYP3A perpetrators. RESULTS: For a single 250 mg ivosidenib dose, the HP model predicted geometric mean ratios of 2.14 (plasma area under concentration-time curve, to infinity [AUC0-∞]) and 1.04 (maximum plasma concentration [Cmax]) with the strong CYP3A4 inhibitor, itraconazole, within 1.26-fold of the observed values (2.69 and 1.0, respectively). The AML model reasonably predicted the observed ivosidenib concentration-time profiles across all dose levels in patients. Predicted ivosidenib geometric mean steady-state AUC0-∞ and Cmax ratios were 3.23 and 2.26 with ketoconazole, and 1.90 and 1.52 with fluconazole, respectively. Co-administration of the strong CYP3A4 inducer, rifampin, predicted a greater DDI effect on a single dose of ivosidenib than on multiple doses (AUC ratios 0.35 and 0.67, Cmax ratios 0.91 and 0.81, respectively). CONCLUSION: Potentially clinically relevant DDI effects with CYP3A4 inducers and moderate and strong inhibitors co-administered with ivosidenib were predicted. Considering the challenges of conducting clinical DDI studies in patients, this PBPK approach is valuable in ivosidenib DDI risk assessment and management.


Asunto(s)
Antineoplásicos/farmacocinética , Inductores del Citocromo P-450 CYP3A/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Itraconazol/farmacocinética , Leucemia Mieloide Aguda/tratamiento farmacológico , Administración Oral , Antineoplásicos/administración & dosificación , Área Bajo la Curva , Simulación por Computador , Citocromo P-450 CYP3A/metabolismo , Inductores del Citocromo P-450 CYP3A/administración & dosificación , Inhibidores del Citocromo P-450 CYP3A/administración & dosificación , Interacciones Farmacológicas , Femenino , Fluconazol/administración & dosificación , Fluconazol/farmacocinética , Glicina/administración & dosificación , Glicina/análogos & derivados , Glicina/farmacocinética , Voluntarios Sanos , Humanos , Itraconazol/administración & dosificación , Cetoconazol/administración & dosificación , Cetoconazol/farmacocinética , Masculino , Microsomas Hepáticos , Modelos Biológicos , Piridinas/administración & dosificación , Piridinas/farmacocinética , Rifampin/administración & dosificación , Rifampin/farmacocinética
18.
J Med Chem ; 63(17): 10045-10060, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787144

RESUMEN

The design and discovery of a new series of (5-alkynyl-3-hydroxypicolinoyl)glycine inhibitors of prolyl hydroxylase (PHD) are described. These compounds showed potent in vitro inhibitory activity toward PHD2 in a fluorescence polarization-based assay. Remarkably, oral administration of 17, with an IC50 of 64.2 nM toward PHD2, was found to stabilize HIF-α, elevate erythropoietin (EPO), and alleviate anemia in a cisplatin-induced anemia mouse model with an oral dose of 25 mg/kg. Rat and dog studies showed that 17 has good pharmacokinetic properties, with oral bioavailabilities of 55.7 and 54.0%, respectively, and shows excellent safety profiles even at a high dose of 200 mg/kg in these animals. Based on these results, 17 is currently being evaluated in a phase I clinical trial for anemia.


Asunto(s)
Anemia/tratamiento farmacológico , Glicina/análogos & derivados , Glicina/uso terapéutico , Ácidos Picolínicos/uso terapéutico , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Anemia/inducido químicamente , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cisplatino , Perros , Diseño de Fármacos , Eritropoyetina/metabolismo , Femenino , Glicina/farmacocinética , Glicina/toxicidad , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Ácidos Picolínicos/síntesis química , Ácidos Picolínicos/farmacocinética , Ácidos Picolínicos/toxicidad , Inhibidores de Prolil-Hidroxilasa/síntesis química , Inhibidores de Prolil-Hidroxilasa/farmacocinética , Inhibidores de Prolil-Hidroxilasa/toxicidad , Ratas Sprague-Dawley , Relación Estructura-Actividad
19.
J Clin Pharmacol ; 60(11): 1432-1440, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32603526

RESUMEN

Roxadustat (FG-4592), an oral hypoxia-inducible factor prolyl hydroxylase inhibitor that stimulates erythropoiesis, was evaluated in a phase 1b study in patients with end-stage renal disease with anemia on hemodialysis. Seventeen patients, on epoetin-alfa maintenance therapy with stable hemoglobin levels ≥10 g/dL, had epoetin-alfa discontinued on day 3 and were enrolled in this double-blind placebo-controlled study. Two cohorts were randomized 3:1 (roxadustat: placebo). Patients received single doses of roxadustat (1 or 2 mg/kg) or placebo 1 hour after hemodialysis on day 1 and 2 hours before dialysis on day 8. Maximum plasma concentration and area under the plasma concentration-time curve for patients receiving roxadustat were slightly more than dose proportional and elimination half-life ranged from 14.7 to 19.4 hours. Roxadustat was highly protein bound (99%) in plasma, and dialysis contributed a small fraction of the total clearance: only 4.56% and 3.04% of roxadustat recovered from the 1 and 2 mg/kg dose groups, respectively. Roxadustat induced transient elevations of endogenous erythropoietin that peaked between 7 and 14 hours after dosing and returned to baseline by 48 hours after dosing. Peak median endogenous erythropoietin levels were 96 mIU/mL and 268 mIU/mL for the 1- and 2-mg/kg doses, respectively, within physiologic range of endogenous erythropoietin responses to hypoxia at high altitude or after blood loss. No serious adverse events were reported, and there were no treatment- or dose-related trends in adverse event incidence.


Asunto(s)
Anemia/tratamiento farmacológico , Glicina/análogos & derivados , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Fallo Renal Crónico/complicaciones , Inhibidores de Prolil-Hidroxilasa/administración & dosificación , Inhibidores de Prolil-Hidroxilasa/farmacocinética , Administración Oral , Adulto , Anciano , Anemia/etiología , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Eritropoyesis/efectos de los fármacos , Eritropoyetina/sangre , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/sangre , Glicina/farmacocinética , Humanos , Hipoxia , Isoquinolinas/efectos adversos , Isoquinolinas/sangre , Masculino , Persona de Mediana Edad , Inhibidores de Prolil-Hidroxilasa/efectos adversos , Inhibidores de Prolil-Hidroxilasa/sangre , Diálisis Renal , Resultado del Tratamiento
20.
J Pharmacol Exp Ther ; 374(2): 342-353, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32487538

RESUMEN

Anemia of chronic kidney disease (CKD) is a multifactorial disorder caused by impaired erythropoietin (EPO) production and altered iron homeostasis associated with inflammation. Hypoxia-inducible factor (HIF) is a transcription factor that stimulates erythropoiesis via a coordinated response involving increased EPO production and enhanced iron availability for Hb synthesis. HIF degradation is regulated by HIF-prolyl hydroxylase (HIF-PH) enzymes. We hypothesized that roxadustat, an orally available small-molecule inhibitor of HIF-PH, would increase EPO production and promote erythropoiesis in animal models of anemia. In cells, roxadustat increased both HIF-1α and HIF-2α proteins, leading to an increase in EPO production, even in the presence of EPO-suppressing inflammatory cytokines. Roxadustat administered intermittently to healthy rats and cynomolgus monkeys increased circulating EPO levels, reticulocytes, blood Hb, and hematocrit in a dose-dependent manner. Roxadustat corrected anemia in a rat model of CKD after five-sixth nephrectomy and in a rat model of anemia of inflammation with impaired iron metabolism induced by peptidoglycan-polysaccharide (PG-PS). In the PG-PS model, roxadustat significantly decreased hepatic expression of hepcidin, a hormone responsible for iron sequestration and functional iron deficiency, and increased expression of two genes involved in duodenal iron absorption: divalent metal transporter 1 and duodenal cytochrome b. In conclusion, by activating the HIF pathway, roxadustat increased EPO production, elevated Hb, corrected anemia, and improved iron homeostasis. The coordinated erythropoietic response stimulated by roxadustat, involving both EPO production and mobilization of iron stores, makes this compound a promising treatment of anemia of CKD and anemia associated with functional iron deficiency. SIGNIFICANCE STATEMENT: Roxadustat is a novel orally available small-molecule inhibitor of HIF prolyl hydroxylase enzymes that reversibly stabilizes HIF-α, thus activating transcription of HIF-dependent genes, including EPO and regulators of iron homeostasis. Activation of the HIF pathway by roxadustat induces erythropoiesis in healthy rats and monkeys and corrects experimentally induced anemia in rats. The coordinated erythropoietic response that increases EPO production and mobilizes iron stores makes roxadustat a promising treatment for anemia of chronic kidney disease and anemia associated with functional iron deficiency.


Asunto(s)
Anemia/complicaciones , Anemia/tratamiento farmacológico , Glicina/análogos & derivados , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Isoquinolinas/farmacología , Insuficiencia Renal Crónica/complicaciones , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Eritropoyesis/efectos de los fármacos , Eritropoyetina/metabolismo , Glicina/farmacocinética , Glicina/farmacología , Glicina/uso terapéutico , Haplorrinos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/farmacocinética , Isoquinolinas/uso terapéutico , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA