Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Pharmacol ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38752556

ABSTRACT

Belzutifan (Welireg, Merck & Co., Inc., Rahway, NJ, USA) is an oral, potent hypoxia-inducible factor-2α inhibitor, recently approved in the United States for the treatment of von Hippel-Lindau (VHL) disease-associated renal cell carcinoma (RCC) and other VHL disease-associated neoplasms. Safety and efficacy were investigated in two clinical studies: a Phase 1 dose escalation/expansion study in solid tumors and RCC and a Phase 2 study in VHL-RCC. A population pharmacokinetic model was used to estimate belzutifan exposures to facilitate exposure-response (E-R) analyses for efficacy and safety endpoints. Relationships between exposure and efficacy (overall response rate, disease control rate, progression-free survival, best overall tumor size response, and other endpoints), safety outcomes (Grade ≥3 anemia, Grade ≥3 hypoxia, and time to first dose reduction/dose interruption), and pharmacodynamic biomarkers (erythropoietin [EPO] and hemoglobin [Hgb]) were evaluated using various regression techniques and time-to-event analyses. Efficacy E-R was generally flat with non-significant positive trends with exposure. The safety E-R analyses demonstrated a lack of relationship for Grade ≥3 hypoxia and a positive relationship for Grade ≥3 anemia, with incidences also significantly dependent on baseline Hgb. Exposure-dependent reductions in EPO and Hgb were observed. Based on the cumulative benefit-risk assessment in VHL disease-associated neoplasms using E-R, no a priori dose adjustment is recommended for any subpopulation. These analyses supported the benefit-risk profile of belzutifan 120 mg once daily dosing in patients with VHL-RCC for labeling and the overall development program.

2.
CPT Pharmacometrics Syst Pharmacol ; 12(10): 1499-1510, 2023 10.
Article in English | MEDLINE | ID: mdl-37596839

ABSTRACT

Belzutifan (Welireg, Merck & Co., Inc., Rahway, NJ, USA) is an oral, potent inhibitor of hypoxia-inducible factor 2α, approved for the treatment of certain patients with von Hippel-Lindau (VHL) disease-associated renal cell carcinoma (RCC), central nervous system hemangioblastomas, and pancreatic neuroendocrine tumors. It is primarily metabolized by the polymorphic uridine 5'-diphospho-glucuronosyltransferase (UGT) 2B17 and cytochrome (CYP) 2C19. A population pharmacokinetic (PK) model was built, using NONMEM version 7.3, based on demographics/PK data from three clinical pharmacology (food effect, formulation bridging, and genotype/race effect) and two clinical studies (phase I dose escalation/expansion in patients with RCC and other solid tumors; phase II in patients with VHL). Median (range) age for the combined studies was 55 years (19-84) and body weight was 73.6 kg (42.1-165.8). Belzutifan plasma PK was well-characterized by a linear two-compartment model with first-order absorption and elimination. For patients with VHL, the predicted geometric mean (% coefficient of variation) apparent clearance was 7.3 L/h (51%), apparent total volume of distribution was 130 L (35%), and half-life was 12.39 h (42%). There were no clinically relevant differences in belzutifan PK based on the individual covariates of age, sex, ethnicity, race, body weight, mild/moderate renal impairment, or mild hepatic impairment. In this model, dual UGT2B17 and CYP2C19 poor metabolizers (PMs) were estimated to have a 3.2-fold higher area under the plasma concentration-time curve compared to UGT2B17 extensive metabolizer and CYP2C19 non-PM patients. This population PK analysis enabled an integrated assessment of PK characteristics with covariate effects in the overall population and subpopulations for belzutifan labeling.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cytochrome P-450 CYP2C19/metabolism , Kidney Neoplasms/drug therapy , Body Weight
3.
Br J Clin Pharmacol ; 86(9): 1795-1806, 2020 09.
Article in English | MEDLINE | ID: mdl-32162721

ABSTRACT

AIMS: In this exposure-response analysis, the dosing regimen for tildrakizumab, an antibody for treating moderate-to-severe chronic plaque psoriasis, was determined using data from 3 randomised controlled trials (P05495/NCT01225731: phase 2b, n = 355; reSURFACE 1/NCT01722331: phase 3, n = 772; reSURFACE 2/NCT01729754: phase 3, n = 1090). METHODS: A maximum drug effect (Emax ) logistic-regression exposure-efficacy model was used to describe the week 12 Psoriasis Area and Severity Index (PASI) responses with average concentration of tildrakizumab during weeks 1-12 (Cavg12 ) as exposure metric. The impact of covariates (e.g., body weight, region) was tested. Exposure-safety, longitudinal pharmacokinetic-pharmacodynamic and risk-benefit analyses were also conducted. RESULTS: At week 12, Emax was estimated at 62.2, 37.9 and 14.6% of responders for PASI75/90/100, respectively. Exposure-response curves plateaued at exposures >5 µg mL-1 . Heavier subjects had a lower response rate to placebo as measured by PASI75/90/100 than lighter subjects. PASI100 placebo response was less in subjects with higher baseline PASI score and older age. Simulated week 12 PASI75 increased by ≤4% on increasing the dose from 100 to 200 mg every 12 weeks (Q12W). The pharmacokinetic-pharmacodynamic model adequately described the time course of PASI change after treatment in the entire population and in each subject. Risk-benefit profiles were favourable for the 100- and 200-mg doses in different weight subgroups. CONCLUSIONS: Patients with moderate-to-severe psoriasis should receive 100-mg subcutaneous tildrakizumab Q12W. Patients with high body weight (>90 kg) may benefit from a higher dose (200-mg Q12W).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Psoriasis , Aged , Double-Blind Method , Humans , Pharmaceutical Preparations , Psoriasis/drug therapy , Severity of Illness Index , Treatment Outcome
4.
J Clin Pharmacol ; 52(12): 1861-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22174430

ABSTRACT

A mechanistic drug-disease model was developed on the basis of a previously published integrated glucose-insulin model by Jauslin et al. A glucokinase activator was used as a test compound to evaluate the model's ability to identify a drug's mechanism of action and estimate its effects on glucose and insulin profiles following oral glucose tolerance tests. A kinetic-pharmacodynamic approach was chosen to describe the drug's pharmacodynamic effects in a dose-response-time model. Four possible mechanisms of action of antidiabetic drugs were evaluated, and the corresponding affected model parameters were identified: insulin secretion, glucose production, insulin effect on glucose elimination, and insulin-independent glucose elimination. Inclusion of drug effects in the model at these sites of action was first tested one-by-one and then in combination. The results demonstrate the ability of this model to identify the dual mechanism of action of a glucokinase activator and describe and predict its effects: Estimating a stimulating drug effect on insulin secretion and an inhibiting effect on glucose output resulted in a significantly better model fit than any other combination of effect sites. The model may be used for dose finding in early clinical drug development and for gaining more insight into a drug candidate's mechanism of action.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucokinase/metabolism , Glucose/metabolism , Insulin/metabolism , Models, Biological , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Glucose Tolerance Test , Humans , Male , Middle Aged
5.
J Clin Pharmacol ; 51(2): 153-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20220046

ABSTRACT

A model able to simultaneously characterize and simulate 24-hour glucose and insulin profiles following multiple meal tests was developed, extending an integrated glucose-insulin model for oral glucose tolerance tests that was previously published. The analysis was based on glucose and insulin measurements from 59 placebo-treated patients with type 2 diabetes. Circadian variations in glucose homeostasis were assessed on relevant parameters based on literature review. They were best described by a nighttime dip in insulin secretion between approximately 9 p.m. and 5 a.m. using a modulator function. The integrated glucose-insulin model has thus been shown to be applicable to real-life situations determined by multiple meals over the course of a day. This provides the basis for the analysis and simulation of long-term glucose and insulin data. The model may also prove useful for understanding antidiabetic drug actions and requirements in the context of circadian changes in glucose-insulin regulation.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin/blood , Models, Biological , Circadian Rhythm , Female , Glucose Tolerance Test , Homeostasis , Humans , Male , Middle Aged , Nonlinear Dynamics
6.
Basic Clin Pharmacol Toxicol ; 106(3): 189-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20050839

ABSTRACT

The integrated glucose-insulin model was originally developed on a variety of intravenous glucose provocation experiments in healthy volunteers and type 2 diabetic patients. The model, which simultaneously describes time-courses of glucose and insulin based on mechanism-based components for production, elimination and homeostatic feedback, has been further extended to oral glucose provocations, meal tests and insulin administration. The model has been used to describe experiments ranging from 4 to 24 hr. Applications of the integrated glucose-insulin model include the clinical assessment of the mechanism of action of anti-diabetic drugs and the magnitude of their effects. Finally, the model was used for optimizing the design of provocation experiments.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Models, Biological , Blood Glucose/metabolism , Computer Simulation , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose Tolerance Test , Glycated Hemoglobin/metabolism , Homeostasis , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin Secretion
7.
J Clin Pharmacol ; 47(10): 1244-55, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17906159

ABSTRACT

An integrated model for the glucose-insulin system describing oral glucose tolerance test data was developed, extending on a previously introduced model for intravenous glucose provocations. Model extensions comprised the description of glucose absorption by a chain of transit compartments with a mean transit time of 35 minutes, a bioavailability of 80%, and a representation of the incretin effect, expressed as a direct effect of the glucose absorption rate on insulin secretion. The ability of the model to predict the incretin effect was assessed by simulating the observed difference in insulin response following an oral glucose tolerance test compared with an isoglycemic glucose infusion mimicking an oral glucose tolerance test profile. The extension of the integrated glucose-insulin model to gain information from oral glucose tolerance test data considerably expands its range of applications because the oral glucose tolerance test is one of the most common glucose challenge experiments for assessing the efficacy of hypoglycemic agents in clinical drug development.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Insulin/metabolism , Models, Biological , Computer Simulation , Drug Design , Female , Glucose/biosynthesis , Glucose Tolerance Test , Humans , Insulin Secretion , Kinetics , Male , Middle Aged
8.
J Clin Pharmacol ; 47(9): 1159-71, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17766701

ABSTRACT

An integrated model for the regulation of glucose and insulin concentrations following intravenous glucose provocations in healthy volunteers and type 2 diabetic patients was developed. Data from 72 individuals were included. Total glucose, labeled glucose, and insulin concentrations were determined. Simultaneous analysis of all data by nonlinear mixed effect modeling was performed in NONMEM. Integrated models for glucose, labeled glucose, and insulin were developed. Control mechanisms for regulation of glucose production, insulin secretion, and glucose uptake were incorporated. Physiologically relevant differences between healthy volunteers and patients were identified in the regulation of glucose production, elimination rate of glucose, and secretion of insulin. The model was able to describe the insulin and glucose profiles well and also showed a good ability to simulate data. The features of the present model are likely to be of interest for analysis of data collected in antidiabetic drug development and for optimization of study design.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Insulin/metabolism , Models, Biological , Clinical Trials as Topic , Computer Simulation , Diabetes Mellitus, Type 2/drug therapy , Drug Design , Glucose/biosynthesis , Glucose Tolerance Test , Homeostasis , Humans , Hypoglycemic Agents/pharmacology , Insulin Secretion , Kinetics , Nonlinear Dynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...