Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 88(1): 248-259, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34182590

RESUMEN

Alemtuzumab is a lymphodepleting monoclonal antibody utilized in conditioning regimens for allogeneic haematopoietic cell transplantation (HCT). A recently proposed therapeutic range of 0.15-0.6 µg/mL on the day of transplantation is associated with better HCT outcomes. The purpose of this study was to characterize alemtuzumab population pharmacokinetic/pharmacodynamic (PK/PD) and to propose individualized subcutaneous dosing schemes to achieve this optimal level for paediatric patients. METHODS: Alemtuzumab concentration and absolute lymphocyte count (ALC) profiles were obtained from 29 paediatric and young adult patients (median age 6.4 y; range 0.28-21.4 y) with nonmalignant disorders undergoing HCT. PK/PD analyses were performed using nonlinear mixed effects modelling. Monte Carlo simulation was conducted to evaluate different improved dosing approaches. RESULTS: A one-compartment model with sequential zero- and first-order absorption adequately described subcutaneously administered alemtuzumab PK. Model fit was significantly improved by including allometrically scaled body weight on clearance (0.080 L/h/70 kg) and volume of distribution (17.4 L/70 kg). ALC reduction following subcutaneous alemtuzumab was swift. An inhibitory Emax model best characterized the relationship between alemtuzumab concentration and ALC. Emax and EC50 were estimated as 1.18 × 103 /µL and 0.045 µg/mL, respectively. The currently used per kg dosing was found to cause uneven alemtuzumab exposure across different age and weight cohorts. Simulations indicated optimal target achieving dose as allometry-based dose of 18 mg × (weight/70)0.75 or body surface area-based dose of 10 mg/m2 , divided over 3 days, with a potential individualized top-up dose; both of which yielded similar results. CONCLUSION: An allometry- or body surface area-based starting dosing regimen in combination with individualized Bayesian PK estimation using concentration feedback is proposed for alemtuzumab precision dosing in children undergoing allogeneic HCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Alemtuzumab , Teorema de Bayes , Niño , Simulación por Computador , Humanos , Acondicionamiento Pretrasplante , Adulto Joven
2.
Ther Drug Monit ; 44(3): 404-413, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34629445

RESUMEN

BACKGROUND: Teicoplanin is a glycopeptide antibiotic used for the treatment of methicillin-resistant Staphylococcus aureus infections. To ensure successful target attainment, therapeutic drug monitoring-informed dosage adjustment is recommended. However, it relies on the experience of the clinician and the frequency of drug measurements. This study aimed to design a new optimal dosing regimen of teicoplanin with a maintenance dosing strategy for neonates and children based on their physiological characteristics. METHODS: Data from teicoplanin-treated patients (n = 214) were collected from electronic medical records. Covariate analyses were performed using population pharmacokinetic (PK) modeling with 399 serum teicoplanin concentrations from 48 neonates and 166 children. Multiple PK simulations were conducted to explore optimal dosing regimens that would allow control of the trough concentration to the target of 15-30 mg/L quicker than the current standard regimen. RESULTS: Allometrically scaled body weight, postmenstrual age (PMA), renal function, and serum albumin were implemented as substantial covariates for teicoplanin clearance in a two-compartment PK model. Covariate analyses and comprehensive simulation assessments recommended the following modifications to the current regimen: (1) decreased dose for premature babies (PMA ≤28 weeks), (2) decreased dose for children with renal dysfunction, and (3) increased dose for children (0.5-11 years) with an estimated glomerular filtration rate of ≥90 mL/min/1.73 m2. CONCLUSIONS: This study leverages real-world clinical information and proposes new optimal dosing regimens for teicoplanin in neonates and children through PK modeling and simulation analyses, taking into account the age, including PMA, and renal function of patients.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Teicoplanina , Antibacterianos/farmacocinética , Niño , Monitoreo de Drogas , Humanos , Recién Nacido , Método de Montecarlo , Teicoplanina/farmacocinética
3.
Eur J Clin Pharmacol ; 77(8): 1157-1168, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33527208

RESUMEN

PURPOSE: Variability in teicoplanin pharmacokinetics has been explained by multiple factors such as body weight, renal function, and serum albumin level. To improve mechanistic understanding of the causes of variability, a physiologically based pharmacokinetic (PBPK) model can be used as a systematic platform. In this study, a PBPK model of teicoplanin was developed to quantitatively assess the effects of physiological changes due to disease status using virtual populations. METHODS: Predictive performance of the models was evaluated by comparing simulated and observed concentration-time profiles of teicoplanin. Subsequently, sensitivity analyses were conducted to identify potential factors contributing to individual differences in teicoplanin PK. RESULTS: The developed PBPK model generated concentration-time profiles that were comparable to clinical observations in healthy adults, including Caucasians and Japanese, and after single-dose and multiple-dose administration. The predicted PK parameters (i.e., Cmax, AUC, clearance) were within a two-fold range of the observed data in patients with renal impairments as well as healthy adults. Changes in total and unbound teicoplanin concentrations at 72 h, after various dosing regimens (tested 4-14 mg/kg q12h for three doses as a loading dose and then 4-14 mg/kg daily as a maintenance dose), were sensitive to renal function and serum albumin concentrations. CONCLUSION: The PBPK model of teicoplanin provides mechanistic insight into the factors altering its disposition and allows assessments of the theoretical and quantitative impact of individual changes in physiological parameters on its PK even when an actual assessment with adequate sample sizes of patients is challenging.


Asunto(s)
Antibacterianos/farmacocinética , Modelos Biológicos , Insuficiencia Renal/metabolismo , Albúmina Sérica/metabolismo , Teicoplanina/farmacocinética , Anciano , Área Bajo la Curva , Pueblo Asiatico , Simulación por Computador , Femenino , Tasa de Filtración Glomerular , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Gravedad del Paciente , Población Blanca
4.
Br J Clin Pharmacol ; 86(5): 933-943, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31851762

RESUMEN

AIMS: Fanconi anaemia (FA) is a rare disorder characterized by progressive bone marrow failure that requires haematopoietic cell transplantation (HCT). Busulfan is used in conditioning regimens prior to HCT. Doses used in non-FA patients cause life-threatening toxicities in FA patients and data on busulfan pharmacokinetics (PK) in this population are limited. This study characterized busulfan PK in paediatric FA patients using population PK modelling and evaluated the effect of body composition on steady-state concentrations (Css ). METHODS: A total of 200 busulfan plasma concentrations in 29 FA patients from a recent study (Clinicaltrials.gov; NCT01082133) were available for population PK modelling. The effect of different body size-scaled doses and body compositions on Css was investigated using population PK modelling. RESULTS: Fat free mass (FFM) was identified as the best size descriptor in a two-compartment busulfan PK model in FA patients. Conventional dosing, based on an amount of busulfan per kilogram of total body mass, resulted in higher Css in FA patients with higher body mass index (BMI). A newly proposed FFM-based dosing strategy would eliminate the observed trend of higher concentrations in high BMI patients, and achieve consistent Css across a wide BMI spectrum. CONCLUSIONS: This is the first study to describe the population PK of busulfan in paediatric FA patients. The proposed model will facilitate PK model-informed precision dosing. FFM-based dosing is expected to improve the probability of achieving target Css , particularly in obese patients, while minimizing the risk of overdosing.


Asunto(s)
Alquilantes , Busulfano , Anemia de Fanconi , Trasplante de Células Madre Hematopoyéticas , Alquilantes/farmacocinética , Composición Corporal , Busulfano/farmacocinética , Niño , Anemia de Fanconi/terapia , Femenino , Humanos , Masculino , Acondicionamiento Pretrasplante
5.
Biol Blood Marrow Transplant ; 25(12): 2416-2421, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31326610

RESUMEN

It is well known that pharmacokinetics (PK)-guided busulfan (BU) dosing increases engraftment rates and lowers hepatotoxicity in patients undergoing hematopoietic cell transplantation (HCT). However, there are no published PK data in patients with Fanconi anemia (FA), who are known to have baseline DNA repair defect and related inherent sensitivity to chemotherapy. In our prospective, multi-institutional study of alternative donor HCT for FA using chemotherapy-only conditioning, we replaced the single dose of total-body irradiation with BU at initial doses of 0.8 to 1.0 mg/kg and 0.6 to 0.8 mg/kg given i.v. every 12 hours for 4 doses. Patients received the first dose of i.v. busulfan on day -8, and blood levels for PK were obtained. PK samples were drawn following completion of infusion. BU PK levels were collected at 2 hours, 2 hours and 15 minutes, and 4, 5, 6, and 8 hours from the start of infusion. The remaining 3 doses of BU were given on days -7 and -6. Thirty-seven patients with available BU PK data with a median age of 9.2 years (range, 4.3 to 44 years) are included in the final analyses. The overall BU PK profile in patients with FA is similar to non-FA patients after considering their body weight. In our cohort, a strong correlation between BU clearance and weight supports current practice of per kilogram dosing. However, not surprisingly, we show that the disease (ie, host) sensitivity related to FA is the main determinant of total dose of BU that can be safely administered to patients in this high-risk population. On the basis of our results, we propose an optimal BU concentration at steady-state level of ≤350 ng/mL (equivalent to total cumulative exposure of 16.4 mg*h/L for 4 doses over 2 days) for patients with FA undergoing HCT. To our knowledge, this is the first and largest report of prospective BU PK in patients with FA undergoing HCT, providing an optimal BU target cutoff to achieve stable donor engraftment while avoiding excessive toxicity.


Asunto(s)
Busulfano/administración & dosificación , Busulfano/farmacocinética , Anemia de Fanconi/terapia , Trasplante de Células Madre Hematopoyéticas , Acondicionamiento Pretrasplante , Adolescente , Adulto , Aloinjertos , Busulfano/efectos adversos , Niño , Preescolar , Femenino , Humanos , Masculino
6.
Am J Transplant ; 19(10): 2944-2948, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31012522

RESUMEN

Cannabidiol (CBD), a major purified nonpsychoactive component of cannabis with anticonvulsant properties, was approved by the U.S. Food and Drug Administration (FDA) in June 2018 as an adjuvant treatment for refractory epilepsy (Epidiolex; GW Pharmaceuticals). CBD is metabolized by cytochrome P450 (CYP)3A4 and CYP2C19 with a growing body of evidence suggesting it is also a potent inhibitor of these pathways. We report for the first time a significant drug-drug interaction between the purified CBD product and tacrolimus. A participant in a CBD clinical trial for epilepsy who was also receiving tacrolimus showed an approximately 3-fold increase in dose-normalized tacrolimus concentrations while receiving 2000-2900 mg/day of CBD. Our report delineates an important concern for the transplant community with the increasing legalization of cannabis and advent of an FDA-approved CBD product. Larger studies are needed to better understand the impact of this drug-drug interaction in solid organ transplant recipients.


Asunto(s)
Cannabidiol/metabolismo , Epilepsia/tratamiento farmacológico , Inmunosupresores/metabolismo , Nefritis Intersticial/tratamiento farmacológico , Tacrolimus/metabolismo , Adulto , Cannabidiol/uso terapéutico , Interacciones Farmacológicas , Epilepsia/complicaciones , Epilepsia/metabolismo , Epilepsia/patología , Femenino , Humanos , Inmunosupresores/uso terapéutico , Nefritis Intersticial/complicaciones , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Pronóstico , Tacrolimus/uso terapéutico
7.
Biopharm Drug Dispos ; 40(9): 350-357, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31691978

RESUMEN

We previously analysed the serum concentrations of dihydrocodeine in a 1-month-old infant with respiratory depression after being prescribed dihydrocodeine phosphate 2.0 mg/day divided t.i.d. for 2 days. The purpose was to develop a full physiologically based pharmacokinetic (PBPK) model that could account for these and other drug monitoring results. Based on experiments in Caco-2 cell monolayers, the effective permeability of dihydrocodeine in human jejunum was established as 1.28 × 10-4 cm/s. The in vitro Vmax /Km values for dihydrocodeine demethylation mediated by recombinant cytochrome P450 2D6 and 3A4 were 0.19 and 0.066 µl/min/pmol, respectively, and for dihydrocodeine 6-O-glucuronidation mediated by recombinant UGT2B4 and 2B7, the Vmax /Km values were 0.14 and 0.22 µl/min/mg protein, respectively. Renal clearance was calculated as 5.37 L/h on the total clearance value multiplied by the fraction recovered in urine. The reported plasma concentration-time profiles of dihydrocodeine after intravenous administration in healthy volunteers were used to adjust the tissue partitioning ratios. The developed model simulated the pharmacokinetic profiles of dihydrocodeine after single and multiple oral administrations reasonably well in the same population. Subsequently, the validated model was used to simulate pharmacokinetic profiles for five pediatric cases, including the 1-month-old Japanese boy and a 14-year-old Japanese girl who took an overdose of dihydrocodeine phosphate (37 mg). The simulated pharmacokinetic profiles for five virtual pediatric subjects matching the age, gender, and P450 2D6 phenotype of each case approximately reflected the observed values. These results suggested that our dihydrocodeine PBPK model reproduced the results of clinical cases reasonably well for subjects.


Asunto(s)
Codeína/análogos & derivados , Modelos Biológicos , Administración Intravenosa , Administración Oral , Adolescente , Adulto , Analgésicos Opioides/farmacocinética , Codeína/administración & dosificación , Codeína/farmacocinética , Femenino , Humanos , Recién Nacido , Masculino
8.
Biol Blood Marrow Transplant ; 23(4): 635-641, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28089878

RESUMEN

Alemtuzumab is frequently used as part of reduced-intensity conditioning (RIC) regimens for allogeneic hematopoietic cell transplantation (HCT) in pediatric patients with nonmalignant diseases. We previously suggested an optimal day 0 targeted range of alemtuzumab, but there are no pediatric data regarding the pharmacokinetics (PK) of subcutaneous alemtuzumab to guide precision dosing trials. The goal of this study was to prospectively characterize alemtuzumab PK and to explore absolute lymphocyte count (ALC) as a predictor of interindividual variability. We prospectively enrolled 23 patients who received an alemtuzumab, fludarabine, and melphalan RIC regimen. Seventeen patients completed study and received 1 mg/kg alemtuzumab divided over 5 days subcutaneously, starting on day -14. The median age was 7 years (range, .5 to 18). Blood sampling for PK measurements and descriptive PK analyses were performed. The median maximum alemtuzumab concentration was 2.39 µg/mL (interquartile range, 1.98 to 2.92). The median terminal half-life was 5.2 days (interquartile range, 2.7 to 7.8). The median concentration at day 0 was 1.27 µg/mL (interquartile range, .35 to 1.51). Importantly, day 0 alemtuzumab levels and area under the curve negatively correlated with predose ALC and ALC area-time, respectively. In conclusion, we reported the PK of subcutaneous alemtuzumab given to pediatric allogeneic HCT patients and observed that almost all patients have persistence of lytic levels of alemtuzumab beyond day 0, at levels in excess of that needed to reduce the risk of acute graft-versus-host disease. Additionally, levels correlate with pretransplant ALC. These results will allow the development of population PK models for precision dosing trials.


Asunto(s)
Alemtuzumab/farmacocinética , Recuento de Linfocitos , Acondicionamiento Pretrasplante/métodos , Adolescente , Alemtuzumab/administración & dosificación , Antineoplásicos Inmunológicos , Niño , Preescolar , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/prevención & control , Semivida , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Melfalán/uso terapéutico , Estudios Prospectivos , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico
9.
Drug Metab Dispos ; 45(1): 23-26, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27780835

RESUMEN

Organic cation transporter 1 (OCT1) plays an important role in the disposition of clinically important drugs, and the capacity of OCT1 activity is presumed to be proportional to the protein expression level in organ tissues. Knowledge of OCT1 protein expression in children, especially neonates and small infants, is currently very limited. Here, we report on the characterization of OCT1 protein expression in neonatal, infant, and pediatric liver samples performed using immunoblot analysis. OCT1 protein expression was detected in liver samples from neonates as early as postnatal days 1 and 2. This youngest group showed significantly lower OCT1 expression normalized by glyceraldehyde-6-phosphate dehydrogenase (values given as means ± S.D. in arbitrary units; 0.03 ± 0.02, n = 7) compared with samples from patients aged 3 to 4 weeks (0.08 ± 0.03, n = 5, P < 0.01), 3 to 6 months (0.23 ± 0.15, n = 7, P < 0.01), 11 months to 1 year (0.42 ± 0.32, n = 6, P < 0.01), and 8 to 12 years (1.00 ± 0.44, n = 7, P < 0.01). These data demonstrate an age-dependent increase in OCT1 expression from birth up to 8 to 12 years of age, and the findings of this study contribute to the understanding of OCT1 functional capacity and its effect upon the disposition of OCT1 substrates in neonates and small infants.


Asunto(s)
Envejecimiento/metabolismo , Hígado/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/biosíntesis , Envejecimiento/genética , Niño , Preescolar , Femenino , Genotipo , Hepatocitos/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Factor 1 de Transcripción de Unión a Octámeros/genética
10.
Pediatr Blood Cancer ; 64(8)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28205374

RESUMEN

BACKGROUND: Sirolimus has recently been shown to be efficacious and tolerable in pediatric patients with complicated vascular anomalies. Nevertheless, dosing information remains very limited especially for neonates and infants. The purpose of this study was to develop an age-appropriate sirolimus starting dosing regimen based on the developmental changes in drug elimination capacity using data collected in neonates and infants. PROCEDURE: A recently developed sirolimus maturation model [Emoto et al. CPT Pharmacometrics Syst Pharmacol, 2016] was used to simulate clearance estimates using realistic age and weight covariates for age cohorts aged 0-24 months. Next, predose concentrations at steady state were generated for each age cohort of neonates and infants. Dose requirements to attain predefined target trough concentration ranges (10-15 and 5-10 ng/ml) were simulated across the different age groups. Starting doses were chosen to maximize the likelihood of achieving sirolimus-targeted concentrations. RESULTS: The trajectory of simulated sirolimus clearances increased with age and was in agreement with the previous findings in the Phase 2 study. The proposed dosing regimens covered eight age cohorts and resulted in target attainment of more than 75-95% across selected regimens. CONCLUSIONS: This study identified age-appropriate sirolimus dosing regimens for neonates and infants. The algorithm in combination with therapeutic drug management will facilitate sirolimus precision dosing in young children with vascular anomalies. A prospective evaluation is being planned.


Asunto(s)
Algoritmos , Vasos Sanguíneos/anomalías , Sirolimus/farmacocinética , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Modelos Teóricos
11.
Biol Blood Marrow Transplant ; 22(12): 2220-2225, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27664325

RESUMEN

We describe a single-center prospective study of alemtuzumab as a second-line agent for steroid-refractory (SR) acute graft-versus-host disease (aGVHD) in pediatric and young adult allogeneic hematopoietic stem cell transplant recipients. Alemtuzumab was administered for grades II to IV aGVHD if patients did not improve within 5 days or worsened within 48 hours after corticosteroids. Interim analyses of alemtuzumab levels and response were performed after every 5 patients enrolled, resulting in 3 dosing cohorts, as follows: (1) .2 mg/kg alemtuzumab subcutaneously on days 1 to 5 (maximum of 31 mg over 5 days) and .2 mg/kg/dose (not exceeding 10 mg/dose) on days 15, 22, and 29; (2) .2 mg/kg alemtuzumab subcutaneously on days 1 to 5 (maximum of 43 mg over 5 days) and .2 mg/kg/dose on day 7, 10, 15, 22, and 29; and (3) .2 mg/kg subcutaneously on days 1 to 5 and .2 mg/kg/dose on day 7, 10, 15, and 22. Alemtuzumab levels were assessed before starting alemtuzumab and at days 1, 3, 6, 10, and 14 and weekly until day 99, where day 1 was the day of first alemtuzumab dose. Fifteen patients (median age, 10 years; range, 1.4 to 27) received alemtuzumab for grades II (6%), III (74%), and IV (20%) SR-aGVHD. The overall response rate was 67%, with complete response (CR) in 40%, partial response (PR) in 27%, and no response in 33%. The median day 6 alemtuzumab level was 2.79 µg/mL (interquartile range, 1.34 to 4.89) in patients with CR compared with .62 µg/mL (interquartile range, .25 to 1.45) in patients with PR + no response (P < .05). Ninety percent (n = 9) of patients with a CR or PR reduced corticosteroid doses within 8 weeks from first alemtuzumab dose. Side effects included fever (26%) and transient thrombocytopenia (53%). Asymptomatic viremias occurred in all patients but invasive viral disease occurred in 2 patients. One patient developed Epstein-Barr virus-post-transplantation lymphoproliferative disorder. Eighty percent (n = 12) of patients were alive at 6 months, of whom 53% (n = 8) were free of GVHD whereas 13% (n = 2) developed chronic GVHD. Alemtuzumab is an effective second-line agent for children and young adults with SR-aGVHD. Higher alemtuzumab levels are associated with CR. A real-time dose adjusted alemtuzumab study is needed to further optimize the dose of alemtuzumab in aGVHD.


Asunto(s)
Alemtuzumab/administración & dosificación , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/métodos , Terapia Recuperativa/métodos , Enfermedad Aguda , Adolescente , Adulto , Niño , Preescolar , Esquema de Medicación , Femenino , Fiebre/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Lactante , Masculino , Estudios Prospectivos , Inducción de Remisión , Esteroides/farmacología , Esteroides/uso terapéutico , Trombocitopenia/etiología , Trasplante Homólogo , Resultado del Tratamiento , Virosis/etiología , Adulto Joven
12.
Ther Drug Monit ; 38(5): 607-13, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27310200

RESUMEN

BACKGROUND: Sirolimus is a mammalian target of rapamycin inhibitor that is being used to prevent organ rejection in kidney transplant patients often in combination with calcineurin inhibitors (CNIs; cyclosporine and tacrolimus). All 3 drugs are metabolized primarily by CYP3As. Clinical drug-drug interaction (DDI) studies of cyclosporine on sirolimus pharmacokinetics have been reported; however, there are a few clinical DDI data related to tacrolimus. METHODS: In vitro inhibition assay with sirolimus were conducted using recombinant CYP3As and human microsomes in the presence and absence of CNIs. Sirolimus concentrations were determined by validated high-performance liquid chromatography-tandem mass spectrometry (LC/MS-MS) assay. The DDI risk in terms of increase in sirolimus area under the curve (AUC) was evaluated by a mechanistic model using in vitro inhibition data and published pharmacokinetic parameters of CNIs. RESULTS: Both CNIs showed similar inhibitory effects on sirolimus metabolism in human liver and intestinal microsomes. Cyclosporine predominantly inhibited CYP3A4 (half maximal inhibitory concentration = 0.71 µM) rather than CYP3A5 (>5 µM), whereas tacrolimus showed similar inhibition for CYP3A4 (0.29 µM) and CYP3A5 (0.41 µM). The predicted increase in AUC of sirolimus during the coadministration of cyclosporine was 3.9-fold, which was comparable to the observed clinical data (3.3-fold) in healthy volunteers. Sirolimus AUC was estimated to a 2.8- to 3.2-fold increase during the coadministration of tacrolimus, based on the reported Cmax values and doses of tacrolimus in kidney transplant patients. In addition, exploratory sensitivity analysis indicated that the predicted increase in sirolimus AUC was sensitive to the free fraction of cyclosporine but not to the free fraction of tacrolimus. CONCLUSIONS: This study suggests that tacrolimus has a lower clinical DDI risk potential affecting sirolimus pharmacokinetics compared with cyclosporine in kidney transplant patients.


Asunto(s)
Interacciones Farmacológicas , Trasplante de Riñón , Sirolimus/farmacocinética , Tacrolimus/farmacología , Ciclosporina/farmacología , Citocromo P-450 CYP3A/efectos de los fármacos , Inhibidores del Citocromo P-450 CYP3A/farmacología , Humanos , Inmunosupresores/farmacocinética , Técnicas In Vitro , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Medición de Riesgo
13.
Br J Clin Pharmacol ; 80(6): 1438-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26256674

RESUMEN

AIMS: Sirolimus is an mTOR inhibitor metabolized by CYP3A4 and CYP3A5. Reported effects of CYP3A5 polymorphisms on sirolimus pharmacokinetics (PK) have shown unexplained discrepancies across studies. We quantitatively assessed the effect of CYP3A5*3 status on sirolimus PK by in vitro assessment and simulation using a physiologically-based PK (PBPK) model. In addition, we explored designs for an adequately powered pharmacogenetic association study. METHOD: In vitro metabolism studies were conducted to confirm individual CYP contribution to sirolimus metabolism. PK profiles were simulated in CYP3A5 expressers and non-expressers with a PBPK model. The pre-dose concentration predictions were used as the outcome parameter to estimate the required sample size for a pharmacogenetic association study. RESULTS: Sirolimus metabolism was inhibited by over 90% by ketoconazole, a CYP3A specific inhibitor. The PBPK model developed based on CL(int) of recombinant CYP3A4, CYP3A5 and CYP2C8 predicted a small CYP3A5*3 effect on simulated sirolimus PK profiles. A subsequent power analysis based on these findings indicated that at least 80 subjects in an enrichment design, 40 CYP3A5 expressers and 40 non-expressers, would be required to detect a significant difference in the predicted trough concentrations at 1 month of therapy (P < 0.05, 80% power). CONCLUSIONS: This study suggests that CYP3A5 contribution to sirolimus metabolism is much smaller than that of CYP3A4. Observed discrepancies across studies could be explained as the result of inadequate sample size. PBPK model simulations allowed mechanism-based evaluation of the effects of CYP3A5 genotype on sirolimus PK and provided preliminary data for the design of a future prospective study.


Asunto(s)
Citocromo P-450 CYP3A/genética , Inmunosupresores/farmacocinética , Polimorfismo Genético , Sirolimus/farmacocinética , Humanos , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Modelos Biológicos , Modelos Genéticos , Estudios Retrospectivos
15.
Ther Drug Monit ; 37(3): 395-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25162215

RESUMEN

BACKGROUND: Sirolimus is an inhibitor of mammalian target of rapamycin, which exhibits large interindividual pharmacokinetic variability. We report sirolimus pharmacokinetic data collected as part of a concentration-controlled multicenter phase II clinical trial in pediatric patients with neurofibromatosis type 1. The purpose of this study was to explore the effect of growth on age-dependent changes in sirolimus clearance with a focus on cytochrome P450 3A (CYP3A) subfamily mediated metabolism. METHODS: Predose blood samples were obtained at steady state from 18 patients with neurofibromatosis type 1. Sirolimus and its 5 CYP3A-dependent primary metabolites were quantified by HPLC-UV/MS. Concentration ratios of metabolites to sirolimus (metabolic ratio) were calculated as an index of metabolite formation. RESULTS: Metabolic ratios of the main metabolites, 16-O-demethylsirolimus (16-O-DM) and 24-hydroxysirolimus (24OH), were significantly correlated with sirolimus clearance, whereas this was not the case for the other 3 metabolites (25-hydroxysirolimus, 46-hydroxysirolimus, and 39-O-demethylsirolimus). The ratios for the 16-O-DM and 24OH metabolites were lower in children than adults. No significant difference in allometrically scaled metabolic ratios of 16-O-DM and 24OH was observed between children and adults. CONCLUSIONS: This study suggests that the age-dependent changes in sirolimus clearance can be explained by size-related increases in CYP3A metabolic capacity, most likely due to liver and intestinal growth. These findings will help facilitate the development of age-appropriate dosing algorithms for sirolimus in infants and children.


Asunto(s)
Envejecimiento/metabolismo , Inmunosupresores/metabolismo , Inmunosupresores/farmacocinética , Neurofibromatosis 1/metabolismo , Sirolimus/metabolismo , Sirolimus/farmacocinética , Adolescente , Adulto , Envejecimiento/sangre , Niño , Preescolar , Citocromo P-450 CYP3A/metabolismo , Femenino , Humanos , Inmunosupresores/sangre , Masculino , Persona de Mediana Edad , Neurofibromatosis 1/sangre , Sirolimus/análogos & derivados , Sirolimus/sangre , Adulto Joven
16.
Biopharm Drug Dispos ; 36(3): 148-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25450725

RESUMEN

Teneligliptin is a type 2 diabetes drug that has an inhibitory effect on dipeptidyl peptidase-4. The aim of this study was to establish a physiologically based pharmacokinetic (PBPK) model to elucidate in detail the pharmacokinetics of teneligliptin. A PBPK model of teneligliptin was developed using the population-based Simcyp simulator incorporating the results of in vitro and in vivo studies. Model validation was conducted by comparison of simulated teneligliptin plasma concentrations with those from clinical trials. Using the PBPK model, predicted drug-drug interactions with concomitant medication were examined. The robustness of the PBPK model was demonstrated by the accurate simulation of clinically measured plasma concentrations of teneligliptin after oral administration in different ethnic groups, in subjects belonging to different age groups and in patients with kidney or liver impairment; none of these factors were incorporated during model development. The fraction absorbed and intestinal availability of teneligliptin predicted by the model were 0.62 and 0.99, respectively. The predicted ratios of areas under the time-concentration curves (AUCs) in patients with moderate and severe renal impairment who were concomitantly administered ketoconazole, a potent inhibitor of P450 3A4, were, respectively, 2.1- and 2.2-fold those in healthy adults who were given teneligliptin alone. A robust PBPK model reflecting the pharmacokinetic properties of teneligliptin was constructed. The final optimized PBPK model enabled us to elucidate in detail the factors affecting the pharmacokinetics of teneligliptin and to predict changes in exposure in drug-drug interactions or in specific populations.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Modelos Biológicos , Pirazoles/farmacocinética , Tiazolidinas/farmacocinética , Adulto , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A/farmacología , Inhibidores de la Dipeptidil-Peptidasa IV/sangre , Interacciones Farmacológicas , Femenino , Humanos , Mucosa Intestinal/metabolismo , Cetoconazol/farmacología , Hígado/metabolismo , Hepatopatías/metabolismo , Masculino , Persona de Mediana Edad , Pirazoles/sangre , Insuficiencia Renal/metabolismo , Reproducibilidad de los Resultados , Tiazolidinas/sangre , Adulto Joven
17.
Br J Pharmacol ; 181(7): 1091-1106, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37872109

RESUMEN

BACKGROUND AND PURPOSE: Fraction metabolized (fm ) and fraction transported (ft ) are important for understanding drug-drug interactions (DDIs) in drug discovery and development. However, current in vitro systems cannot accurately estimate in vivo fm due to inability to reflect the ft by efflux transporters (ft,efflux ). This study demonstrates how CYP3A-mediated DDI for CYP3A/P-gp substrates can be predicted using Hu-PXB mice as human liver chimeric mice. EXPERIMENTAL APPROACH: For estimating human in vitro fm by CYP3A enzyme (fm,CYP3A,in vitro ), six drugs, including CYP3A/P-gp substrates (alprazolam, cyclosporine, docetaxel, midazolam, prednisolone, and theophylline) and human hepatocytes were incubated with or without ketoconazole as a CYP3A inhibitor. We calculated fm,CYP3A,in vitro based on hepatic intrinsic clearance. To estimate human in vivo fm,CYP3A (fm,CYP3A,in vivo ), we collected information on clinical DDI caused by ketoconazole for these six drugs. We calculated fm,CYP3A,in vivo using the change of total clearance (CLtotal ). For evaluating the human DDI predictability, the six drugs were administered intravenously to Hu-PXB and SCID mice with or without ketoconazole. We calculated the change of CLtotal caused by ketoconazole. We compared the CLtotal change in humans with that in Hu-PXB and SCID mice. KEY RESULTS: The fm,CYP3A,in vitro was overestimated compared to the fm,CYP3A,in vivo . Hu-PXB mice showed much better correlation in the change of CLtotal with humans (R2 = 0.95) compared to SCID mice (R2 = 0.0058). CONCLUSIONS AND IMPLICATIONS: CYP3A-mediated DDI can be predicted by correctly estimating human fm,CYP3A,in vivo using Hu-PXB mice. These mice could be useful predicting hepatic fm and ft,efflux .


Asunto(s)
Citocromo P-450 CYP3A , Cetoconazol , Humanos , Ratones , Animales , Citocromo P-450 CYP3A/metabolismo , Cetoconazol/metabolismo , Ratones SCID , Hígado/metabolismo , Interacciones Farmacológicas
18.
J Clin Pharmacol ; 63(10): 1156-1168, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37496106

RESUMEN

Physiologically based pharmacokinetic (PBPK) models are useful in bridging drug exposure in different ethnic groups, and there is increasing regulatory application of this approach in adults. Reported pediatric PBPK models tend to focus on the North European population, with few examples in other ethnic groups. This study describes the development and verification of a Japanese pediatric PBPK population. The development of the model was based on the existing North European pediatric population. Japanese systems and clinical data were collated from public databases and the literature, and the underlying demographics and equations were optimized so that physiological outputs represented the Japanese pediatric population. The model was tested using 14 different small molecule drugs, eliminated by a variety of pathways, including cytochrome P450 3A4 (CYP3A4) metabolism and renal excretion. Given the limitations of the clinical data, the overall performance of the model was good, with 44/62 predictions for PK parameters (area under the plasma drug concentration-time curve, AUC; maximum serum concentration, Cmax ; clearance, CL) being within 0.8- to 1.25-fold, 56/62 within 0.67- to 1.5-fold, and 61/62 within 0.5- to 2.0-fold of the observed values. Specific results for the 5 CYP3A4 substrates showed 20/31 cases were predicted within 0.8- to 1.25-fold, 27/31 within 0.67- to 1.5-fold, and all were within 0.5- to 2.0-fold of the observed values. Given the increased regulatory use of pediatric PBPK in drug development, expanding these models to other ethnic groups are important. Considering qualifying these models based on the context of use, there is a need to expand on the current research to include a larger range of drugs with different elimination pathways. Collaboration among academic, industry, model providers, and regulators will facilitate further development.


Asunto(s)
Citocromo P-450 CYP3A , Eliminación Renal , Niño , Humanos , Simulación por Computador , Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Pueblos del Este de Asia , Modelos Biológicos
19.
Xenobiotica ; 42(8): 719-30, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22324396

RESUMEN

Cynomolgus monkeys are widely used to predict human pharmacokinetic and/or toxic profiles in the drug developmental stage. Characterization of cynomolgus monkey P450s such as the mRNA expression level, substrate specificity, and inhibitor selectivity were conducted to provide helpful information in designing monkey in vivo studies and monkey-to-human extrapolation. The expression levels of 12 monkey P450 mRNAs, which are considered to be important P450 subfamilies in drug metabolism, were investigated in the liver, small intestine (duodenum, jejunum, and ileum), and colon of individual monkeys. 3. In vitro activities and intrinsic clearance values were determined in monkey intestinal and liver microsomes (MIM and MLM, respectively) using nine typical oxidative reactions for human P450s. Paclitaxel 6α-hydroxylation, diclofenac 4'-hydroxylation, and S-mephenytoin 4'-hydroxylation showed low activities in MIM and MLM. IC50 values of eight selective inhibitors of human P450s were determined in MIM and MLM. Inhibitory effects of furafylline and sulfaphenazole were weak in monkeys on phenacetin O-deethylation and diclofenac 4'-hydroxylation, respectively. These results show profiles of monkey P450s in both the intestine and liver in detail and contribute to a better understanding of the species difference in substrate specificity and inhibitor selectivity between cynomolgus monkeys and humans.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores Enzimáticos/farmacología , Intestinos/enzimología , Hígado/enzimología , Macaca fascicularis/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Inactivación Metabólica , Concentración 50 Inhibidora , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Masculino , Espectrometría de Masas , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Oxidación-Reducción/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estándares de Referencia , Especificidad por Sustrato/efectos de los fármacos , Teofilina/análogos & derivados , Teofilina/farmacología , Factores de Tiempo
20.
Adv Pharmacol ; 95: 365-391, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35953161

RESUMEN

Cytochrome P450 enzymes play an important role in the pharmacokinetics, efficacy, and toxicity of drugs. Age-dependent changes in P450 enzyme expression have been studied based on several detection systems, as well as by deconvolution of in vivo pharmacokinetic data observed in pediatric populations. The age-dependent changes in P450 enzyme expression can be important determinants of drug disposition in childhood, in addition to the changes in body size and the other physiological parameters, and effects of pharmacogenetics and disease on organ functions. As a tool incorporating drug-specific and body-specific factors, physiologically-based pharmacokinetic (PBPK) models have become increasingly used to characterize and explore mechanistic insights into drug disposition. Thus, PBPK models can be a bridge between findings from basic science and utilization in predictive science. Pediatric PBPK models incorporate additional system specific information on developmental physiology and ontogeny and have been used to predict pharmacokinetic parameters from preterm neonates onwards. These models have been advocated by regulatory authorities in order to support pediatric clinical trials. The purpose of this chapter is to highlight accumulated knowledge and findings from basic research focusing on P450 enzymes, as well as the current status and future challenges of expanding the utilization of pediatric PBPK modeling.


Asunto(s)
Sistema Enzimático del Citocromo P-450 , Modelos Biológicos , Niño , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Recién Nacido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA