Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Clin Pharmacol Ther ; 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38752712

RESUMEN

The landscape of oncology drug development has witnessed remarkable advancements over the last few decades, significantly improving clinical outcomes and quality of life for patients with cancer. Project Optimus, introduced by the U.S. Food and Drug Administration, stands as a groundbreaking endeavor to reform dose selection of oncology drugs, presenting both opportunities and challenges for the field. To address complex dose optimization challenges, an Oncology Dose Optimization IQ Working Group was created to characterize current practices, provide recommendations for improvement, develop a clinical toolkit, and engage Health Authorities. Historically, dose selection for cytotoxic chemotherapeutics has focused on the maximum tolerated dose, a paradigm that is less relevant for targeted therapies and new treatment modalities. A survey conducted by this group gathered insights from member companies regarding industry practices in oncology dose optimization. Given oncology drug development is a complex effort with multidimensional optimization and high failure rates due to lack of clinically relevant efficacy, this Working Group advocates for a case-by-case approach to inform the timing, specific quantitative targets, and strategies for dose optimization, depending on factors such as disease characteristics, patient population, mechanism of action, including associated resistance mechanisms, and therapeutic index. This white paper highlights the evolving nature of oncology dose optimization, the impact of Project Optimus, and the need for a tailored and evidence-based approach to optimize oncology drug dosing regimens effectively.

2.
Br J Clin Pharmacol ; 89(7): 2272-2282, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36823349

RESUMEN

AIMS: Patients with solid tumours were treated with the anti-PD-1 antibody dostarlimab in the Phase I GARNET trial. This study aimed to examine dostarlimab's effect on corrected QT (QTc) interval and the systemic concentration-QTc interval relationship. METHODS: In GARNET Part 2B, patients received 500 mg dostarlimab every 3 weeks (Q3W) for four cycles, then 1000 mg Q6W. Triplicate 12-lead ECGs were recorded and time-matched pharmacokinetic (PK) samples collected at screening, on Day 1 of Cycles 1, 4, 5, 8, 12 (pre-dose and 0.5 h after infusion end), and at treatment end. Concentration-change from baseline QTcF (ΔQTcF) analysis using a linear mixed effects model, summary statistics, incidence of clinically noteworthy ECG values and rhythm abnormalities were evaluated. RESULTS: A total of 377 patients were considered for evaluation (n = 15 excluded from concentration-ΔQTcF). There was a non-significant concentration-ΔQTcF relationship (0.001589 ms/µg/mL; P = .5906). Mean ΔQTcF increase was <6 ms (upper-bound two-sided 90% confidence interval [CI], <10 ms at all post-dose timepoints). Highest geometric mean concentration was 414.1 µg/mL (Cycle 5 Day 1, 0.5 h) with predicted mean ∆QTcF of 3.064 ms (upper-bound two-sided 90% CI: 5.071). Mean QTcF prolongation (all concentrations) was 2.4 ms. QTcF prolongation ≥500 ms occurred in five patients (1.3%); 51 (13.6%) and nine patients (2.4%) had ΔQTcF ≥30 ms and ≥60 ms, respectively. Ten patients (2.7%) reported rhythm abnormalities. No U-wave abnormalities, torsades de pointes, ventricular tachycardia or ventricular fibrillation/flutter were observed. CONCLUSIONS: Dostarlimab does not cause clinically significant QTcF prolongation exceeding the regulatory concern threshold.


Asunto(s)
Síndrome de QT Prolongado , Neoplasias , Humanos , Anticuerpos Monoclonales/efectos adversos , Neoplasias/tratamiento farmacológico , Neoplasias/inducido químicamente , Anticuerpos Monoclonales Humanizados/efectos adversos , Electrocardiografía , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/epidemiología , Frecuencia Cardíaca
3.
CPT Pharmacometrics Syst Pharmacol ; 12(1): 87-94, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36317409

RESUMEN

Dostarlimab (JEMPERLI) is an anti-programmed cell death protein-1 (PD-1) monoclonal antibody (mAb) which is approved by the US Food and Drug Administration for patients with recurrent/advanced mismatch repair-deficient solid tumors, including endometrial cancer, following progression on prior treatment, with approval based on data from the phase I GARNET trial. To support dostarlimab dose regimen recommendations, we estimated and compared the potency of dostarlimab relative to anti-PD-1 mAb pembrolizumab using both data published from the KEYNOTE-001 trial of pembrolizumab and data from the GARNET trial. PD-1 target engagement was assessed ex vivo in blood samples via a super antigen staphylococcal enterotoxin B stimulation assay and interleukin-2 (IL-2) stimulation ratios calculated for dostarlimab. A non-linear mixed-effect sigmoid maximum effect inhibitory model was fitted to dostarlimab IL-2 stimulation ratios using extracted pembrolizumab data as informative priors. The estimated half-maximal effective concentration was 1.95 µg ml-1 (95% credibility interval: 0.21-5.87) for dostarlimab and 1.59 µg ml-1 (95% confidence interval: 0.42-6.12) for pembrolizumab. These findings suggest dostarlimab and pembrolizumab to be equipotent for peripheral PD-1 suppression based on analysis of ex vivo IL-2 stimulation ratios. Accounting for a three-fold dilution between serum and tumor, a target dostarlimab trough concentration of ~54 µg ml-1 would be needed for 90% suppression in the tumor. These data support the use of dostarlimab as a potent PD-1 suppressor and the recommended dostarlimab monotherapy dose regimen of 500 mg Q3W ×4 cycles followed by 1000 mg Q6W thereafter in recurrent/advanced solid tumors.


Asunto(s)
Antineoplásicos Inmunológicos , Neoplasias , Humanos , Interleucina-2/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología
4.
Br J Clin Pharmacol ; 88(9): 4142-4154, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35357027

RESUMEN

AIM: Develop a population pharmacokinetic (PopPK) model to characterise the pharmacokinetics (PK) of anti-programmed cell death protein-1 (PD-1) antibody dostarlimab, identify covariates of clinical relevance, and investigate efficacy/safety exposure-response (ER) relationships. METHODS: A PopPK model was developed using Phase 1 GARNET (NCT02715284) trial data for dostarlimab (1, 3 or 10 mg kg-1 every 2 wk; 500 mg every 3 wk or 1000 mg every 6 wk; 500 mg every 3 wk × 4 then 1000 mg every 6 wk [recommended regimen]) serum concentrations over time. Concentration-time data were analysed using nonlinear mixed effects modelling with standard stepwise covariate modelling. ER was explored for treatment-related adverse events and overall response rate (ORR) using logistic regression. RESULTS: PopPK model/adverse event ER analyses included 546 patients (ORR ER analysis n = 362). Dostarlimab PK was well described by a 2-compartment model with time-dependent linear elimination. Time-dependent clearance decreased over time to a maximum of 14.9%. At steady state, estimated dostarlimab geometric mean coefficient of variation % clearance was 0.179 (30.2%) L d-1 ; volume of distribution was 5.3 (14.2%) L; terminal elimination half-life was 23.5 (22.4%) days. Statistically significant covariates were age, body weight, sex, time-varying albumin and alanine aminotransferase for clearance; body weight, albumin and sex for volume of distribution of the central compartment. Hepatic or renal impairment did not affect PK. There were no clinically significant ER relationships. CONCLUSION: Dostarlimab PK parameters are similar to other anti-programmed cell death protein-1 antibodies. The clinical impact of covariates on exposure was limited-to-moderate, supporting recommended dostarlimab monotherapy therapeutic dosing.


Asunto(s)
Neoplasias , Receptor de Muerte Celular Programada 1 , Albúminas , Anticuerpos Monoclonales , Anticuerpos Monoclonales Humanizados , Peso Corporal , Muerte Celular , Ensayos Clínicos Fase I como Asunto , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptor de Muerte Celular Programada 1/uso terapéutico
5.
Pharm Res ; 37(6): 102, 2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32440783

RESUMEN

PURPOSE: To model absolute neutrophil count (ANC) suppression in response to acute radiation (AR) exposure and evaluate ANC time course as a predictor of overall survival (OS) in response to AR exposure with or without treatment with granulocyte colony-stimulating factor in nonhuman primates. METHODS: Source data were obtained from two pivotal studies conducted in rhesus macaques exposed to 750 cGy of whole body irradiation on day 0 that received either placebo, daily filgrastim, or pegfilgrastim (days 1 and 8 after irradiation). Animals were observed for 60 days with ANC measured every 1 to 2 days. The population model of ANC response to AR and the link between observed ANC time course and OS consisted of three submodels characterizing injury due to radiation, granulopoiesis, and a time-to-event model of OS. RESULTS: The ANC response model accurately described the effects of AR exposure on the duration of neutropenia. ANC was a valid surrogate for survival because it explained 76% (95% CI, 41%-97%) and 73.2% (95% CI, 38.7%-99.9%) of the treatment effect for filgrastim and pegfilgrastim, respectively. CONCLUSION: The current model linking radiation injury to neutropenia and ANC time course to OS can be used as a basis for translating these effects to humans.


Asunto(s)
Filgrastim/administración & dosificación , Modelos Biológicos , Neutropenia/prevención & control , Neutrófilos , Polietilenglicoles/administración & dosificación , Traumatismos Experimentales por Radiación/prevención & control , Animales , Estudios de Factibilidad , Femenino , Recuento de Leucocitos , Leucopoyesis/efectos de los fármacos , Leucopoyesis/efectos de la radiación , Macaca mulatta , Masculino , Neutropenia/sangre , Neutropenia/etiología , Neutropenia/mortalidad , Traumatismos Experimentales por Radiación/sangre , Traumatismos Experimentales por Radiación/mortalidad , Factores de Tiempo
6.
Clin Transl Sci ; 13(4): 807-817, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32112517

RESUMEN

Acute exposure to high doses of radiation leads to severe myelosuppression, but few treatments are currently available to treat hematopoietic syndrome of acute radiation syndrome. Granulocyte colony stimulating factors (e.g., filgrastim) stimulate proliferation of neutrophil precursors and enhance mature neutrophil function. Owing to ethical constraints on conducting clinical research in lethally irradiated humans, we developed a model-based strategy to integrate preclinical experience in irradiated nonhuman primates (NHPs) and other clinical myelosuppressive conditions to inform filgrastim dosing to treat hematopoietic syndrome of acute radiation syndrome. Models predicting neutrophil counts and overall survival based on drug exposures were calibrated and scaled from NHPs to adult and pediatric human subjects. Several scenarios were examined investigating variations in filgrastim doses, dose frequency, treatment initiation, and duration, as well as the effect of age and radiation dose rate. Model-based simulations and established safety profiles supported that a subcutaneous filgrastim dose of 10 µg/kg once daily provides a significant survival benefit (50%) over placebo in both adults and children, provided that the treatment is initiated within 1-14 days after radiation exposure and lasts 2-3 weeks. For treatment durations of longer than 3 weeks, filgrastim treatment is not expected to provide significantly greater benefit. This survival benefit is expected to hold for the wide range of radiation doses and dose rates (0.01-1,000 Gy/hours) examined.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Filgrastim/administración & dosificación , Fármacos Hematológicos/administración & dosificación , Síndrome de Radiación Aguda/mortalidad , Adulto , Factores de Edad , Animales , Niño , Simulación por Computador , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Evaluación Preclínica de Medicamentos , Femenino , Células Precursoras de Granulocitos/efectos de los fármacos , Humanos , Inyecciones Subcutáneas , Macaca mulatta , Masculino , Mielopoyesis/efectos de los fármacos , Medición de Riesgo/métodos , Resultado del Tratamiento
7.
Br J Clin Pharmacol ; 85(6): 1312-1325, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30756425

RESUMEN

AIMS: The aims of this study were to develop a pharmacokinetic (PK) and PK-pharmacodynamic (PK/PD) model of cinacalcet in adults and paediatrics with secondary hyperparathyroidism (SHPT) on dialysis, to test covariates of interest, and to perform simulations to inform dosing in paediatrics with SHPT. METHODS: Cinacalcet PK, intact parathyroid hormone (iPTH) and corrected calcium (cCa) time courses following multiple daily oral doses (1-300 mg) were modelled using a nonlinear mixed effects modelling approach using data from eight clinical studies. Model-based trial simulations, using adult or paediatric titration schemas, predicted efficacy (iPTH change from baseline and proportion achieving iPTH decrease ≥30%) and safety (cCa change from baseline and proportion achieving cCa ≤8.4 mg/dL) endpoints at 24 weeks. RESULTS: Cinacalcet PK parameters were described by a two-compartment linear model with delayed first-order absorption-elimination (apparent clearance = 287.74 L h-1 ). Simulations suggested that paediatric starting doses (1, 2.5, 5, 10 and 15 mg) would provide PK exposures less than or similar to a 30 mg adult dose. The titrated dose simulations suggested that the mean (prediction interval) proportion of paediatric and adult subjects achieving ≥30% reduction in iPTH from baseline at Week 24 was 49% (36%, 62%), and 70.1% (62.5%, 77%), respectively. Additionally, the mean (confidence interval) proportion of paediatric and adult subjects achieving cCa ≤8.4 mg dL-1 at Week 24 was 8% (2%, 18%) and 23.6% (17.5%, 30.5%), respectively. CONCLUSIONS: Model-based simulations showed that the paediatric cinacalcet starting dose (0.2 mg kg-1 ), titrated to effect, would provide the desired PD efficacy (PTH suppression <30%) while minimizing safety concerns (hypocalcaemia).


Asunto(s)
Calcimiméticos/farmacocinética , Cinacalcet/farmacocinética , Hiperparatiroidismo Secundario/tratamiento farmacológico , Modelos Biológicos , Diálisis Renal , Insuficiencia Renal Crónica/terapia , Adolescente , Adulto , Factores de Edad , Biomarcadores/sangre , Calcimiméticos/administración & dosificación , Calcimiméticos/efectos adversos , Niño , Preescolar , Cinacalcet/administración & dosificación , Cinacalcet/efectos adversos , Simulación por Computador , Cálculo de Dosificación de Drogas , Femenino , Humanos , Hiperparatiroidismo Secundario/sangre , Hiperparatiroidismo Secundario/diagnóstico , Hiperparatiroidismo Secundario/etiología , Hipocalcemia/inducido químicamente , Masculino , Hormona Paratiroidea/sangre , Diálisis Renal/efectos adversos , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/diagnóstico , Resultado del Tratamiento
8.
CPT Pharmacometrics Syst Pharmacol ; 7(11): 771-779, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30246497

RESUMEN

Optimal dose selection in clinical trials is problematic when efficacious and toxic concentrations are close. A novel quantitative approach follows for optimizing dose titration in clinical trials. A system of pharmacokinetics (PK), pharmacodynamics, efficacy, and toxicity was simulated for scenarios characterized by varying degrees of different types of variability. Receiver operating characteristic (ROC) and clinical trial simulation (CTS) were used to optimize drug titration by maximizing efficacy/safety. The scenarios included were a low-variability base scenario, and high residual (20%), interoccasion (20%), interindividual (40%), and residual plus interindividual variability scenarios, and finally a shallow toxicity slope scenario. The percentage of subjects having toxicity was reduced by 87.4% to 93.5%, and those having efficacy was increased by 52.7% to 243%. Interindividual PK variability may have less impact on optimal cutoff values than other sources of variability. ROC/CTS methods for optimizing dose titration offer an individualized approach that leverages exposure-response relationships.


Asunto(s)
Ensayos Clínicos como Asunto , Curva ROC , Algoritmos , Relación Dosis-Respuesta a Droga , Humanos , Modelos Biológicos , Farmacocinética , Farmacología
10.
J Clin Pharmacol ; 58(6): 717-726, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29534286

RESUMEN

Etelcalcetide, a d-amino acid peptide, is an intravenous calcimimetic approved for the treatment of secondary hyperparathyroidism. Etelcalcetide binds the calcium-sensing receptor and increases its sensitivity to extracellular calcium, thereby decreasing secretion of parathyroid hormone (PTH) by chief cells. Etelcalcetide and its low-molecular-weight transformation products are rapidly cleared by renal excretion in healthy subjects, but clearance is substantially reduced and dependent on hemodialysis in end-stage renal disease. The effective half-life is 3-5 days in patients undergoing hemodialysis 3 times a week. A clinical study using a single microtracer intravenous dose of [14 C]etelcalcetide indicated that 60% of the administered dose was eliminated in dialysate. Etelcalcetide undergoes reversible disulfide exchange with serum albumin to form a serum albumin peptide conjugate that is too large (67 kDa) to be dialyzed, until a subsequent exchange forms etelcalcetide or a low-molecular-weight transformation product. This exchange from albumin is apparent after hemodialysis, when it partially restores etelcalcetide concentrations in plasma. Etelcalcetide has no known risks for drug-drug interactions. In phase 3 studies, 74%-75% of hemodialysis patients with secondary hyperparathyroidism who received etelcalcetide achieved a >30% PTH reduction from baseline versus 8%-10% of patients who received placebo. The pharmacokinetics and pharmacodynamics of etelcalcetide in hemodialysis patients supports a 5-mg starting dose administered after hemodialysis and uptitration in 2.5- or 5-mg increments every 4 weeks to a maximum dose of 15 mg 3 times a week.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Hiperparatiroidismo Secundario/metabolismo , Péptidos/farmacología , Péptidos/farmacocinética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/terapia , Administración Intravenosa , Calcimiméticos/farmacocinética , Calcimiméticos/farmacología , Interacciones Farmacológicas , Humanos , Diálisis Renal , Eliminación Renal/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico
11.
Br J Clin Pharmacol ; 84(5): 911-925, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29318653

RESUMEN

AIM: The objective of the present study was to use pharmacokinetic-pharmacodynamic modelling to characterize the effects of chemotherapy on the granulopoietic system and to predict the absolute neutrophil counts (ANCs) for patients with chemotherapy-induced neutropenia treated with filgrastim and pegfilgrastim. METHODS: Data were extracted from 10 phase I-III studies conducted in 110 healthy adults, and 618 adult and 52 paediatric patients on chemotherapy following administration of filgrastim or pegfilgrastim. The structural model accounted for ANC dynamics and the effects of filgrastim and pegfilgrastim, chemotherapy and corticosteroids. The impact of neutrophils on drug disposition was based on a drug receptor-binding model that assumed quasi-equilibrium and stimulation of the production and maturation of neutrophils upon treatment. The chemotherapy and corticosteroid effects were represented by kinetic-pharmacodynamic-type models, where chemotherapy stimulated elimination of neutrophil precursors at the mitotic stage, and corticosteroids stimulated neutrophil production. RESULTS: The systemic half-lives of filgrastim (2.6 h) and pegfilgrastim (10.1 h) were as expected. The effective half-life of chemotherapy was 9.6 h, with a 2-day killing effect. The rate of receptor elimination from mitotic compartments exhibited extreme interindividual variability (% coefficient of variation >200), suggesting marked differences in sensitivity to chemotherapy effects on ANCs. The stimulatory effects of pegfilgrastim were significantly greater than those of filgrastim. Model qualification confirmed the predictive capability of this model. CONCLUSION: This qualified model simulates the time course of ANC in the absence or presence of chemotherapy and predicts nadir, time to nadir and time of recovery from different grades of neutropenia upon treatment with filgrastim and pegfilgrastim.


Asunto(s)
Filgrastim/efectos adversos , Filgrastim/farmacocinética , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Polietilenglicoles/efectos adversos , Polietilenglicoles/farmacocinética , Ensayos Clínicos Controlados Aleatorios como Asunto/estadística & datos numéricos , Corticoesteroides/efectos adversos , Adulto , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Femenino , Filgrastim/farmacología , Voluntarios Sanos , Fármacos Hematológicos/efectos adversos , Fármacos Hematológicos/sangre , Fármacos Hematológicos/farmacocinética , Fármacos Hematológicos/farmacología , Humanos , Recuento de Leucocitos , Masculino , Neutropenia/inducido químicamente , Polietilenglicoles/farmacología
12.
Clin Pharmacokinet ; 57(1): 71-85, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28508378

RESUMEN

INTRODUCTION: Etelcalcetide is a novel calcimimetic that binds and activates calcium-sensing receptors (CaSRs) for the treatment of secondary hyperparathyroidism (SHPT). METHODS: To assess titrated dosing regimens, population pharmacokinetic (PK) and PK/pharmacodynamic (PKPD) modeling of etelcalcetide was performed using NONMEM 7.2. In this analysis, plasma etelcalcetide, serum parathyroid hormone (PTH) and calcium (Ca) concentration-time data were collected from five phase I, II, and III clinical trials following single or multiple intravenous doses of etelcalcetide ranging from 2.5 to 60 mg. A semi-mechanistic model was used to describe the relationship between etelcalcetide, PTH, and Ca. This model included the role of PTH in Ca regulation, the feedback of Ca onto PTH production via the CaSR, and the activity of etelcalcetide plasma levels in increasing the sensitivity of the CaSR to Ca via the cooperative binding model. The impact of relevant covariates was evaluated by stepwise forward/backward selection. Model evaluation was based on standard goodness-of-fit plots and prediction-corrected visual predictive checks (pcVPCs). Simulation was conducted to evaluate titrated dosing regimens. RESULTS AND DISCUSSION: The time courses of etelcalcetide, PTH, and Ca were well-described by the model. The clearance and central volume of distribution (Vc) of etelcalcetide were 0.472 L/h and 49.9 L, respectively, while estimates of the turnover half-lives of PTH and Ca were 0.36 and 23 h, respectively. The extent of interindividual variability in model parameters was low to moderate (6-67%), and no covariates were identified as significant predictors of PK and PD variability. pcVPCs confirmed the predictive ability of the model. CONCLUSIONS: The current analysis confirms the putative mechanism of action of etelcalcetide as an allosteric activator of CaSR. Simulations showed that dose titration of etelcalcetide, rather than fixed dose, is needed to effectively decrease the PTH level in patient populations.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Modelos Biológicos , Péptidos/administración & dosificación , Insuficiencia Renal Crónica/terapia , Administración Intravenosa , Adulto , Anciano , Anciano de 80 o más Años , Calcio/sangre , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Femenino , Semivida , Humanos , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Péptidos/farmacocinética , Péptidos/farmacología , Diálisis Renal/métodos , Distribución Tisular , Adulto Joven
13.
J Pharmacokinet Pharmacodyn ; 44(1): 43-53, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28063122

RESUMEN

Etelcalcetide (AMG 416) is an allosteric activator of the calcium-sensing receptor for treatment of secondary hyperparathyroidism in patients with chronic kidney disease (CKD) on hemodialysis. To characterize the time course of etelcalcetide in different matrices (plasma, dialysate, urine, and feces), a drug disposition model was developed. Nonlinear mixed-effect modeling was used to describe data from six adults with CKD on hemodialysis who received a single intravenous dose of [14C]etelcalcetide (10 mg; 710 nCi) after hemodialysis (study NCT02054572). A three-compartment model with the following attributes adequately described the observed concentration-time profiles of etelcalcetide in the different matrices: biotransformation in the central compartment; elimination in dialysate, urine, and feces; and a nonspecific elimination process. The terminal half-life of total C-14 in plasma was approximately 56 days. The ratio of conjugation-deconjugation rate constants between etelcalcetide and biotransformed products was 11.3. Simulations showed that three hemodialysis sessions per week for 52 weeks would contribute to 60.1% of the total clearance of etelcalcetide following single-dose intravenous etelcalcetide administration. Minimal amounts were eliminated in urine (2.5%) and feces (5.7%), whereas nonspecific elimination accounted for 31.2% of total elimination. In addition to removal of etelcalcetide, ~10% of small-molecular weight biotransformed products was estimated to have been removed through hemodialysis and in urine. This model provided a quantitative approach to describe biotransformation, distribution, and elimination of etelcalcetide, a unique synthetic D-amino acid peptide, in the relevant patient population.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Modelos Biológicos , Péptidos/farmacocinética , Diálisis Renal , Insuficiencia Renal Crónica/terapia , Radioisótopos de Carbono , Ensayos Clínicos Fase I como Asunto , Simulación por Computador , Heces/química , Humanos , Hiperparatiroidismo Secundario/metabolismo , Péptidos/administración & dosificación , Péptidos/sangre , Péptidos/orina , Diálisis Renal/efectos adversos , Insuficiencia Renal Crónica/complicaciones , Distribución Tisular
14.
Sci Transl Med ; 8(324): 324ra15, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26843190

RESUMEN

Pharmacokinetic and pharmacodynamic models estimate the potency of antiviral agents but do not capture viral and immunologic factors that drive the natural dynamics of infection. We designed a mathematical model that synthesizes pharmacokinetics, pharmacodynamics, and viral pathogenesis concepts to simulate the activity of pritelivir, a DNA helicase-primase inhibitor that targets herpes simplex virus. Our simulations recapitulate detailed viral kinetic shedding features in five dosage arms of a phase 2 clinical trial. We identify that in vitro estimates of median effective concentration (EC50) are lower than in vivo values for the drug. Nevertheless, pritelivir potently decreases shedding at appropriate doses based on its mode of action and long half-life. Although pritelivir directly inhibits replication in epithelial cells, our model indicates that pritelivir also indirectly limits downstream viral spread from neurons to genital keratinocytes, within genital ulcers, and from ulcer to new mucosal sites of infection. We validate our model based on its ability to predict outcomes in a subsequent trial with a higher dose. The model can therefore be used to optimize dose selection in clinical practice.


Asunto(s)
Antivirales/farmacología , Herpesvirus Humano 2/efectos de los fármacos , Herpesvirus Humano 2/fisiología , Modelos Biológicos , Piridinas/farmacología , Tiazoles/farmacología , Ensayos Clínicos como Asunto , Simulación por Computador , Relación Dosis-Respuesta a Droga , Humanos , Cinética , Sulfonamidas , Resultado del Tratamiento , Esparcimiento de Virus/efectos de los fármacos
15.
J Clin Pharmacol ; 55(6): 620-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25581635

RESUMEN

This study characterizes the population pharmacokinetics of AMG 416, an allosteric activator of the calcium-sensing receptor, in subjects with secondary hyperparathyroidism receiving hemodialysis. AMG 416 doses ranging from 2.5 to 60 mg were administered intravenously as single or multiple thrice weekly (TIW) doses at the end of hemodialysis during rinseback. The influence of demographics, concomitant medications, and other disease-related biomarkers on pharmacokinetic parameters was explored. The predictability of the final model was evaluated using bootstrapping and visual predictive checks. A 3-compartment linear pharmacokinetic model that accounts for the hemodialysis clearance best described the data. Plasma clearance (interindividual variability) was 0.564 L/h (14.0%CV). The hemodialysis clearance was 22.2 L/h. The volume of distribution at steady-state was approximately 624 L (82%CV). The mean time to achieve 90% steady-state predialysis concentrations with 3- and 6-hour hemodialysis TIW was 46 and 32 days, respectively. No statistically significant (P < .01) covariates effect was found on pharmacokinetic parameters. Bootstrapping and predictive checks supported model predictive ability. AMG 416 exhibits linear and stationary pharmacokinetics within the range of doses evaluated. Within the range of covariate values investigated, pharmacokinetically driven adjustments of AMG 416 dosing on the basis of these covariates were not warranted.


Asunto(s)
Hiperparatiroidismo Secundario/tratamiento farmacológico , Tasa de Depuración Metabólica/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/farmacocinética , Receptores Sensibles al Calcio/agonistas , Diálisis Renal/métodos , Administración Intravenosa , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Hiperparatiroidismo Secundario/etiología , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
16.
Clin Pharmacol Drug Dev ; 4(5): 387-94, 2015 09.
Artículo en Inglés | MEDLINE | ID: mdl-27137148

RESUMEN

The aim of this study was to evaluate whether genetic polymorphisms in CYP3A5 and ABCB1 are responsible for the interindividual variability observed in quetiapine pharmacokinetics. Pharmacokinetic data from a randomized crossover study evaluating 2 quetiapine 25 mg immediate-release tablets after single oral dose were used to develop a population pharmacokinetic model. The single nucleotide polymorphisms (SNPs) evaluated for the genotype effects of quetiapine pharmacokinetics were CYP3A5 A6986G and ABCB1 C3435T, along with other demographic variables and formulations. A one-compartment distribution model with linear elimination plus four transit compartments for the delayed absorption adequately described quetiapine disposition. CYP3A5 *1/*1 individuals (n = 3) had 29% increased clearance compared to *1/*3 and *3/*3 individuals. The impact of an increased clearance was evaluated by simulations. By computing the probability of target attainment (PTA) of steady-state therapeutic goal at 1-hour and 12-hour time points after 50-400 mg twice-daily regimens, the results indicated that CYP3A5 genotype has minimal impact on the PTA of the 1-hour concentrations but a significant impact on the 12-hour concentrations. The interpretation based on the simulations does not call for a genotype-based dosing scheme and is consistent with consensus guidelines for quetiapine that therapeutic drug monitoring is considered useful. Clinical Pharmacology in Drug Development.


Asunto(s)
Antipsicóticos/farmacocinética , Citocromo P-450 CYP3A/genética , Modelos Biológicos , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Fumarato de Quetiapina/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adulto , Antipsicóticos/administración & dosificación , Antipsicóticos/efectos adversos , Antipsicóticos/sangre , Simulación por Computador , Estudios Cruzados , Citocromo P-450 CYP3A/metabolismo , Genotipo , Voluntarios Sanos , Humanos , Jordania , Masculino , Persona de Mediana Edad , Método de Montecarlo , Dinámicas no Lineales , Farmacogenética , Fenotipo , Fumarato de Quetiapina/administración & dosificación , Fumarato de Quetiapina/efectos adversos , Fumarato de Quetiapina/sangre , Adulto Joven
17.
J Pharmacol Exp Ther ; 347(1): 2-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23857976

RESUMEN

Translation of central nervous system receptor occupancy from animal models to humans has been elusive for many therapeutic targets. However, this may represent a valuable link to clinical efficacy for drugs acting within the brain and spinal cord. The introduction of positron emission tomography marked a significant noninvasive advance in determination of target engagement in the central nervous system. Pharmacokinetic/pharmacodynamic (PK/PD) modeling represents a valuable tool to translate ex vivo receptor occupancy from relevant animal models to humans. Whereas PK properties usually are reasonably scaled across species using standard allometric principles, PD properties related to receptor occupancy are usually species-independent. The translational value and applicability of PK/PD approaches are more directly evident when comparable modeling assumptions and mathematical model structures are employed across experiments and analyses. The purpose of this article is to review the basic principles of PK/PD analysis of receptor occupancy determined using noninvasive positron emission tomography imaging and first principles of allometric PK scaling and PD prediction based on animal data. By use of the data from the area of pain management, we also provide a case study of PK/PD analysis showcasing the importance of PK/PD model assumptions in predicting receptor occupancy in humans based on data from animal models.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Modelos Animales , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Receptores de Neurotransmisores/metabolismo , Animales , Humanos , Modelos Teóricos , Tomografía de Emisión de Positrones , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Receptores de Neurotransmisores/fisiología
18.
Clin Pharmacokinet ; 52(10): 907-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23719682

RESUMEN

BACKGROUND AND OBJECTIVE: Hydrocodone is a semi-synthetic narcotic analgesic and antitussive. Although hydrocodone products have been on the market for over 50 years, relatively little is known about its pharmacokinetics. Additionally, there are no published reports of population pharmacokinetic analyses for hydrocodone. Furthermore, current labeling of hydrocodone-containing products provides little guidance in terms of the impact of patient descriptors on the pharmacokinetics of hydrocodone. The objectives of this analysis were to develop a population pharmacokinetic model that characterizes the pharmacokinetics of hydrocodone following single and multiple oral doses of hydrocodone extended-release capsules (hydrocodone bitartrate ER capsules) in healthy subjects and patients, to examine the impact of patient descriptors on pharmacokinetic parameters and to assess the dose-proportionality of hydrocodone pharmacokinetic. METHODS: A total of 4,714 plasma hydrocodone concentrations from 220 subjects were available for this analysis. The data were extracted from seven studies (five phase 1 and two phase 2 studies). A two-compartment open mamillary model with linear elimination and a complex absorption model was used to fit the data, using NONMEM(®) version 7.1.2 software. The absorption model involved two sequential first-order absorption processes with the delay in the first process accomplished by means of multiple transit compartments. Covariate analysis was performed using standard forward selection followed by backward elimination processes. Model evaluation was performed using a prediction-corrected visual predictive check (pcVPC). RESULTS: The population estimates of apparent oral central volume of distribution and apparent oral linear clearance were 714 L and 64.4 L/h, respectively. The first absorption process was rapid. Creatinine clearance and body surface area (BSA) were statistically significant predictors of the apparent oral clearance and apparent oral volume of distribution. The pcVPC indicated that the model provided a robust and unbiased fit to the data. CONCLUSIONS: A linear model for hydrocodone elimination provided an adequate fit to the observed data over the entire dose range, which supports that hydrocodone bitartrate ER capsules exhibit dose-proportional pharmacokinetics. The formulation of hydrocodone bitartrate ER capsules results in absorption profiles that are variable across and within subjects. Despite the variability in absorption profiles, a relatively simple model provided an adequate fit to the data. Creatinine clearance and BSA were statistically significant predictors of the apparent oral clearance and apparent oral volume of distribution. Absorption characteristics of hydrocodone bitartrate ER capsules should still allow effective plasma concentrations of hydrocodone to be reached quickly and for effective concentrations to be maintained for a long period.


Asunto(s)
Analgésicos Opioides/farmacocinética , Antitusígenos/farmacocinética , Hidrocodona/farmacocinética , Modelos Biológicos , Adulto , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/sangre , Antitusígenos/administración & dosificación , Antitusígenos/sangre , Estudios Cruzados , Preparaciones de Acción Retardada , Humanos , Hidrocodona/administración & dosificación , Hidrocodona/sangre , Persona de Mediana Edad , Adulto Joven
19.
Cancer Chemother Pharmacol ; 66(6): 1039-49, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20140617

RESUMEN

PURPOSE: The pharmacokinetic profiles of bendamustine and active metabolites were defined in patients with rituximab-refractory, relapsed indolent B-cell non-Hodgkin's lymphoma, and supported understanding of exposure-response relationships for efficacy and safety. METHODS: Bendamustine was administered as a 60-min 120 mg/m(2) intravenous infusion on days 1 and 2 of six 21-day cycles. Pharmacokinetic models were developed, with covariate assessment. Correlations between bendamustine exposure and responder status or occurrence of neutropenia, thrombocytopenia, fatigue, nausea, and vomiting were examined. RESULTS: Following a single dose of bendamustine HCl, concentrations declined in a triphasic manner, with rapid distribution, intermediate, and slow terminal phases. The intermediate t (1/2) (40 min) was considered the pharmacologically relevant (beta elimination) t (1/2) since the initial phases accounted for 99% of the AUC. Age, sex, mild/moderate renal, or mild liver impairment did not alter pharmacokinetics. Metabolite concentrations were low relative to parent. No correlation was observed between exposure and safety or efficacy measures because of the limited range of exposures after 120 mg/m(2) administration, except bendamustine C (max) was a significant (P value = 0.013) predictor of the probability of nausea in patients, most of whom were pretreated with antiemetics. CONCLUSIONS: The BSA-based dosing regimen for bendamustine achieved the targeted exposure and was associated with a high incidence of therapeutic response. Given the short t (1/2) and low concentrations of bendamustine observed by 12 h after dosing, the single-dose profile for bendamustine described by these analyses is expected to be representative of the multiple-dose profile. The occurrence of nausea was significantly related to bendamustine exposure, with the probability of nausea increasing as bendamustine C (max) increases.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/metabolismo , Compuestos de Mostaza Nitrogenada/administración & dosificación , Compuestos de Mostaza Nitrogenada/farmacocinética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Área Bajo la Curva , Clorhidrato de Bendamustina , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Humanos , Estimación de Kaplan-Meier , Linfoma no Hodgkin/etnología , Masculino , Persona de Mediana Edad , Neutropenia/inducido químicamente , Compuestos de Mostaza Nitrogenada/efectos adversos , Factores de Tiempo , Resultado del Tratamiento
20.
Nephrol Dial Transplant ; 21(9): 2425-31, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16822794

RESUMEN

BACKGROUND: Haemodialysis vascular access dysfunction is currently a huge clinical problem. In an attempt to reduce the morbidity associated with haemodialysis vascular access dysfunction, we have previously developed and validated a local perivascular paclitaxel release system that has been shown to release paclitaxel for at least 3 weeks. The aim of the current study was to evaluate the in vivo use of these perivascular wraps (for both safety and efficacy) at different time points in our pig model of arteriovenous graft stenosis. METHODS: Paclitaxel-loaded ethylene vinyl acetate wraps were placed around the graft-vein anastomosis on one side, with control polymers being placed on the contralateral side in our pig model of arteriovenous graft stenosis. Animals were sacrificed at early (10-11 days), middle (23-24 days) and late (32-38 days) time points. The entire graft-vein anastomosis was removed at the time of sacrifice and assessed for the extent of luminal stenosis using histomorphometric techniques. RESULT: Graft-vein anastomoses treated with the paclitaxel-loaded polymers had an almost complete absence of luminal stenosis at the middle (23-24 days) and late (32-38 days) time points (when one would expect the development of neointimal hyperplasia) as compared with the contralateral control graft-vein anastomoses (37.90% luminal stenosis in the controls vs 0.10% in the paclitaxel group). There were minimal local side effects from this procedure. CONCLUSIONS: Our results demonstrate the safety and efficacy of paclitaxel-loaded perivascular wraps in the setting of a pig model of arteriovenous graft stenosis. We believe that such a local approach which could be easily applied at the time of surgery is ideally suited for use in the clinical setting of haemodialysis vascular access dysfunction. It is likely that this novel approach could result in a significant reduction in the huge economic and health morbidity costs currently associated with this recalcitrant clinical problem.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Derivación Arteriovenosa Quirúrgica/efectos adversos , Oclusión de Injerto Vascular/tratamiento farmacológico , Paclitaxel/administración & dosificación , Animales , Modelos Animales de Enfermedad , Implantes de Medicamentos , Oclusión de Injerto Vascular/etiología , Oclusión de Injerto Vascular/patología , Porcinos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...