Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
CPT Pharmacometrics Syst Pharmacol ; 12(10): 1398-1410, 2023 10.
Article in English | MEDLINE | ID: mdl-37675853

ABSTRACT

N,N-dimethyltryptamine (DMT) is a psychedelic compound that is being studied as a therapeutic option in various psychiatric disorders. Due to its short half-life, continuous infusion of DMT has been proposed to extend the psychedelic experience and potential therapeutic effects. The primary aim of this work was to design an infusion protocol for DMT based on a desired level of psychedelic intensity using population pharmacokinetic/pharmacodynamic modeling. As a secondary aim, the impact of choosing a continuous variable or a bounded integer pharmacokinetic/pharmacodynamic model to inform such an infusion protocol was investigated. A previously published continuous variable model and two newly developed bounded integer models were used to assess optimal doses for achieving a target response. Simulations were performed to identify an optimal combination of a bolus dose and an infusion rate. Based on the simulations, optimal doses to achieve intensity ratings between 7 and 9 (possible range = 0-10) were a bolus dose of 16 mg DMT fumarate followed by an infusion rate of 1.4 mg/min based on the continuous variable model and 14 mg with 1.2 mg/min for the two bounded integer models. However, the proportion within target was low (<53%) for all models, indicating that individual dose adjustments would be necessary. Furthermore, some differences between the models were observed. The bounded integer models generally predicted lower proportions within a target of 7-9 with higher proportions exceeding target compared with the continuous variable model. However, results varied depending on target response with the major differences observed at the boundaries of the scale.


Subject(s)
Hallucinogens , Humans , Hallucinogens/pharmacology , Hallucinogens/therapeutic use , N,N-Dimethyltryptamine/pharmacology , N,N-Dimethyltryptamine/therapeutic use , Infusions, Intravenous , Computer Simulation
2.
CPT Pharmacometrics Syst Pharmacol ; 12(4): 474-486, 2023 04.
Article in English | MEDLINE | ID: mdl-36762714

ABSTRACT

N,N-dimethyltryptamine (DMT) is a psychedelic substance and is being used as a research tool in investigations of the neurobiology behind the human consciousness using different brain imaging techniques. The effects of psychedelics have commonly been studied using electroencephalography (EEG) and have been shown to produce suppression of alpha power and increase in signal diversity. However, the relationship between DMT exposure and its EEG effects has never been quantified. In this work, a population pharmacokinetic/pharmacodynamic analysis was performed investigating the relationship between DMT plasma concentrations and its EEG effects. Data were obtained from a clinical study where DMT was administered by intravenous bolus dose to 13 healthy subjects. The effects on alpha power, beta power, and Lempel-Ziv complexity were evaluated. DMT was shown to fully suppress alpha power. Beta power was only partially suppressed, whereas an increase in Lempel-Ziv complexity was observed. The relationship between plasma concentrations and effects were described using effect compartment models with sigmoidal maximum inhibitory response or maximum stimulatory response models. Values of the concentration needed to reach half of the maximum response (EC50,e ) were estimated at 71, 137, and 54 nM for alpha, beta, and Lempel-Ziv complexity, respectively. A large amount of between-subject variability was associated with both beta power and Lempel-Ziv complexity with coefficients of variability of 75% and 77% for the corresponding EC50,e values, respectively. Alpha power appeared to be the most robust response, with a between-subject variability in EC50,e of 29%. Having a deeper understanding of these processes might prove beneficial in choosing appropriate doses and response biomarkers in the future clinical development of DMT.


Subject(s)
Electroencephalography , N,N-Dimethyltryptamine , Humans , N,N-Dimethyltryptamine/pharmacology , Electroencephalography/methods
3.
Clin Transl Sci ; 15(12): 2928-2937, 2022 12.
Article in English | MEDLINE | ID: mdl-36088656

ABSTRACT

N,N-dimethyltryptamine (DMT) is a psychedelic compound that is believed to have potential as a therapeutic option in several psychiatric disorders. The number of clinical investigations with DMT is increasing. However, very little is known about the pharmacokinetic properties of DMT as well as any relationship between its exposure and effects. This study aimed to characterize population pharmacokinetics of DMT as well as the relationship between DMT plasma concentrations and its psychedelic effects as measured through subjective intensity ratings. Data were obtained from 13 healthy subjects after intravenous administration of DMT. The data were analyzed using nonlinear mixed-effects modeling in NONMEM. DMT plasma concentrations were described by a two-compartment model with first-order elimination leading to formation of the major metabolite indole 3-acetic acid. The relationship between plasma concentrations and psychedelic intensity was described by an effect site compartment model with a sigmoid maximum effect (Emax ) response. DMT clearance was estimated at 26 L/min, a high value indicating elimination of DMT to be independent of blood flow. Higher concentrations of DMT were associated with a more intense experience with the concentration of DMT at the effect site required to produce half of the maximum response estimated at 95 nM. The maximum achievable intensity rating was 10 and the simulated median maximum rating was zero, 2, 4, 8, and 9 after doses of 1, 4, 7, 14, and 20 mg, respectively. The model can be useful in predicting suitable doses for clinical investigations of DMT based on the desired intensity of the subjective experience.


Subject(s)
Hallucinogens , N,N-Dimethyltryptamine , Humans , N,N-Dimethyltryptamine/pharmacology , Hallucinogens/pharmacokinetics , Hallucinogens/therapeutic use , Infusions, Intravenous
4.
Thromb Haemost ; 122(9): 1573-1583, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35909257

ABSTRACT

BACKGROUND: Dabigatran etexilate, a direct oral thrombin inhibitor, is approved to treat venous thromboembolism (VTE) in both adults and children. OBJECTIVES: This population analysis characterized relationships between dabigatran total plasma concentrations and coagulation laboratory parameters (activated partial thromboplastin time [aPTT]; diluted thrombin time [dTT]; ecarin clotting time [ECT]). METHODS: Data from three phase 2a and one single-arm and one randomized, comparative phase 2b/3 pediatric studies (measurements: aPTT 2,925 [N = 358]; dTT 2,348 [N = 324]; ECT 2,929 [N = 357]) were compared with adult data (5,740 aPTT, 3,472 dTT, 3,817 ECT measurements; N = 1,978). Population models were fitted using nonlinear mixed-effects modeling. Covariates (e.g., sex, age) were assessed on baseline and drug-effect parameters, using a stepwise covariate model-building procedure. RESULTS: Overall, relationships between dabigatran, aPTT, dTT, and ECT were similar in children and adults. For children aged <6 months, a higher proportion of baseline samples were outside or close to the upper aPTT and ECT adult ranges. No age-related differences were detected for dTT. With increasing dabigatran concentration, aPTT rose nonlinearly (half the maximum effect at 368 ng/mL dabigatran) while dTT and ECT increased linearly (0.37 and 0.73% change per ng/mL dabigatran, respectively). Mean baseline aPTT (45 vs. 36 seconds) and ECT (40 vs. 36 seconds) were slightly increased for those aged <6 months versus older children. CONCLUSION: The similar relationships of laboratory parameters observed across pediatric age groups suggests that developmental changes in the hemostatic system may have little effect on response to dabigatran.


Subject(s)
Anticoagulants , Dabigatran , Adolescent , Antithrombins , Blood Coagulation , Blood Coagulation Tests , Child , Child, Preschool , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Female , Humans , Infant , Male , Partial Thromboplastin Time , Randomized Controlled Trials as Topic
5.
J Thromb Haemost ; 19(5): 1259-1270, 2021 05.
Article in English | MEDLINE | ID: mdl-33636042

ABSTRACT

BACKGROUND: Dabigatran etexilate (DE), a direct oral thrombin inhibitor, has been evaluated in children with venous thromboembolism (VTE) using oral solution, pellets, or capsules. OBJECTIVES: This study evaluated DE pharmacokinetics (PK) in children with VTE and the appropriateness of a DE pediatric age- and weight-based dosing algorithm. PATIENTS/METHODS: A population PK model was fitted to data from four single-arm and one randomized, comparative pediatric VTE studies (358 children aged birth to <18 years; 2748 PK observations) and one healthy-adult study (32 males aged <40 years; 1523 PK observations) using nonlinear mixed-effects modeling. A stepwise, covariate, model-building procedure evaluated the influence of covariates (e.g., age, body weight, body surface area [BSA]-normalized renal function, and sex). The final model was used to evaluate the pediatric dosing algorithm, with simulations comparing pediatric trough exposure with reference exposure defined for the pediatric studies. RESULTS: The population PK of dabigatran was adequately described by a two-compartment model with first-order elimination and absorption. Age, weight, BSA-normalized renal function, and sex were statistically significant covariates (all P < .05). Apparent clearance increased with age (independently of body weight), diminished with decreasing BSA-normalized renal function, and was lower in females than males. All disposition parameters increased with body weight escalation (allometric scaling). Simulations confirmed that for all DE formulations, the final pediatric dosing algorithms achieved reference exposure without dose adjustment. CONCLUSIONS: Using a population PK model of DE for children with VTE, simulations showed that the final dosing algorithms were appropriate for all DE formulations; no dose titration was needed.


Subject(s)
Dabigatran , Venous Thromboembolism , Adolescent , Adult , Antithrombins , Body Weight , Child , Computer Simulation , Female , Humans , Male , Venous Thromboembolism/diagnosis , Venous Thromboembolism/drug therapy
6.
Br J Clin Pharmacol ; 85(2): 413-421, 2019 02.
Article in English | MEDLINE | ID: mdl-30414387

ABSTRACT

AIMS: To characterize ticagrelor exposure-response relationship for platelet inhibition in patients with stable coronary artery disease (CAD) and a history of myocardial infarction (MI), using nonlinear mixed effects modelling and simulation. METHODS: Platelet function data were integrated with plasma concentration data of ticagrelor and its active metabolite AR-C1249010XX in a population pharmacokinetic (PK) and pharmacodynamic (PD) model, based on two clinical studies. In the ONSET/OFFSET study, PK and platelet function were assessed in 123 CAD patients receiving placebo, ticagrelor (180 mg followed by 90 mg twice daily) or clopidogrel (600 mg followed by 75 mg once daily). In the PEGASUS-TIMI 54 platelet function substudy, PK and platelet function were assessed during maintenance dosing in 180 prior MI patients receiving placebo, ticagrelor 60 mg or ticagrelor 90 mg twice daily. RESULTS: Platelet inhibition by ticagrelor was described by a sigmoidal Emax model. On average, half maximal inhibition was reached at ticagrelor concentrations of 116 (RSE: 5.3%) nmol l-1 . Simulations showed that near maximal platelet inhibition is achieved with both ticagrelor 60 and 90 mg twice daily. At simulated lower doses, platelet inhibition is overall reduced, more variable between patients, and show greater peak-to-trough variability. Ticagrelor antiplatelet response was similar between the studied patient populations. CONCLUSIONS: In patients with stable CAD or a history of MI, near maximal platelet inhibition is achieved with both ticagrelor 60 and 90 mg twice daily. At modelled doses <60 mg, the response is reduced overall, more variable between patients, and patients will display greater peak-to-trough variability.


Subject(s)
Coronary Artery Disease/drug therapy , Models, Biological , Myocardial Infarction/prevention & control , Purinergic P2Y Receptor Antagonists/pharmacology , Ticagrelor/pharmacology , Adult , Aged , Aged, 80 and over , Blood Platelets/drug effects , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Coronary Artery Disease/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Multicenter Studies as Topic , Myocardial Infarction/blood , Platelet Aggregation/drug effects , Platelet Function Tests , Purinergic P2Y Receptor Antagonists/therapeutic use , Randomized Controlled Trials as Topic , Ticagrelor/therapeutic use , Treatment Outcome , Young Adult
7.
J Pharmacokinet Pharmacodyn ; 44(6): 581-597, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29103208

ABSTRACT

Population model-based (pharmacometric) approaches are widely used for the analyses of phase IIb clinical trial data to increase the accuracy of the dose selection for phase III clinical trials. On the other hand, if the analysis is based on one selected model, model selection bias can potentially spoil the accuracy of the dose selection process. In this paper, four methods that assume a number of pre-defined model structure candidates, for example a set of dose-response shape functions, and then combine or select those candidate models are introduced. The key hypothesis is that by combining both model structure uncertainty and model parameter uncertainty using these methodologies, we can make a more robust model based dose selection decision at the end of a phase IIb clinical trial. These methods are investigated using realistic simulation studies based on the study protocol of an actual phase IIb trial for an oral asthma drug candidate (AZD1981). Based on the simulation study, it is demonstrated that a bootstrap model selection method properly avoids model selection bias and in most cases increases the accuracy of the end of phase IIb decision. Thus, we recommend using this bootstrap model selection method when conducting population model-based decision-making at the end of phase IIb clinical trials.


Subject(s)
Acetates/administration & dosage , Anti-Asthmatic Agents/administration & dosage , Indoles/administration & dosage , Nonlinear Dynamics , Acetates/pharmacokinetics , Anti-Asthmatic Agents/pharmacokinetics , Clinical Trials, Phase II as Topic/statistics & numerical data , Dose-Response Relationship, Drug , Female , Humans , Indoles/pharmacokinetics , Male
8.
Int J Clin Pharmacol Ther ; 55(5): 416-424, 2017 May.
Article in English | MEDLINE | ID: mdl-28139972

ABSTRACT

OBJECTIVE: The population pharmacokinetics of ticagrelor and its active metabolite AR-C124910XX were characterized following ticagrelor 60 mg or 90 mg twice daily oral long-term treatment in 4,426 patients with a history of myocardial infarction. METHODS: The ticagrelor and AR-C124910XX plasma concentration-time data were described by one-compartment models with first-order absorption or metabolite formation and elimination. RESULTS: Systemic exposure to ticagrelor and AR-C124910XX were stable over time. Ticagrelor apparent clearance (CL/F) was 17 L/h for the 60-mg and 15.4 L/h for the 90-mg dose. The CL/F of AR-C124910XX was 11.1 L/h for the 60-mg and 9.95 L/h for the 90-mg dose. Both ticagrelor and AR-C124910XX CL/F were independently influenced by body weight, sex, age, smoking, and Japanese ethnicity. Female sex and age > 75 years were the only categorical covariates, having more than 20% effect on AR-C124910XX CL/F. Ticagrelor CL/F was 6% higher and 11% lower, whereas AR-C124910XX CL/F was 26% higher and 34% lower for patients weighing 110 and 50 kg, respectively, compared with an 83 kg patient. CONCLUSIONS: The small differences in exposure to both ticagrelor and AR-C124910XX between demographic subgroups were in accordance with the consistent efficacy and safety outcomes observed across the population. The results were similar to those observed previously in patients with acute coronary syndromes.
.


Subject(s)
Adenosine/analogs & derivatives , Models, Biological , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Activation, Metabolic , Adenosine/administration & dosage , Adenosine/blood , Adenosine/pharmacokinetics , Administration, Oral , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Gastrointestinal Absorption , Humans , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/blood , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/blood , Ticagrelor
9.
J Clin Pharmacol ; 57(5): 573-583, 2017 05.
Article in English | MEDLINE | ID: mdl-27859337

ABSTRACT

The relationships between drug exposure and the composite risk of cardiovascular (CV) death, myocardial infarction (MI), and stroke as well as the risk of TIMI major bleeding were estimated following long-term treatment with ticagrelor 60 or 90 mg twice daily in 20,942 patients with prior MI. These analyses support the primary reported efficacy and safety evaluations by showing that there were clear separations from placebo early in treatment with both doses, regardless of ticagrelor exposure, for both endpoints. In addition, the exposure-response analyses provided new insight into the contribution of individual exposure levels, rather than dose, as a predictor of events and accounted for differences in the baseline risk between patients. The predicted risks of CV death/MI/stroke were similar despite an increase in the median predicted ticagrelor average steady-state concentration from 606 nmol/L with ticagrelor 60 mg to 998 nmol/L with ticagrelor 90 mg (hazard ratios vs placebo of 0.83 and 0.81, respectively). The corresponding predicted risk of TIMI major bleeding slightly increased (hazard ratios vs placebo of 2.4 and 2.6, respectively). Apart from Japanese patients, showing a lower risk of CV death/MI/stroke, the response to ticagrelor was consistent across the study population, as supported by the combination of relatively flat exposure-response relationships in the studied exposure range, similar sensitivity to ticagrelor exposure, and small exposure differences. Consequently, the present analyses support the selection of the 60-mg dose for all demographic subgroups of patients studied.


Subject(s)
Adenosine/analogs & derivatives , Myocardial Infarction/chemically induced , Myocardial Infarction/mortality , Adenosine/adverse effects , Adenosine/blood , Adenosine/pharmacokinetics , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Double-Blind Method , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Models, Biological , Myocardial Infarction/blood , Myocardial Infarction/drug therapy , Platelet Aggregation Inhibitors/adverse effects , Risk Factors , Ticagrelor
10.
Drugs R D ; 16(2): 165-72, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27139012

ABSTRACT

OBJECTIVE: The aim of this study was to characterise the population pharmacokinetics of FE 999049, a novel recombinant human follicle-stimulating hormone (FSH), after multiple dosing in healthy women, taking into account endogenous FSH levels and the reproductive hormone dynamics. METHODS: Longitudinal measurements of FSH, luteinising hormone, progesterone, estradiol, and inhibin B levels were collected after repeated subcutaneous dosing with 225 IU of FE 999049 in 24 gonadotropin downregulated healthy women. The FSH data were described using nonlinear mixed-effects modelling. RESULTS: The measured FSH levels were modelled as a sum of endogenous FSH and FE 999049. The FE 999049 population pharmacokinetics were best described using a one-compartment model with first-order absorption and elimination, and a transit model for delayed absorption. The apparent clearance and volume of distribution increased with body weight in accordance with an allometrically scaled power exponent of 0.75 and 1, respectively. Endogenous FSH levels were lower in individuals with higher progesterone levels at baseline and were further suppressed over time with increasing inhibin B levels. CONCLUSIONS: This characterisation of FE 999049 population pharmacokinetics after repeated dosing is in line with previous findings after single-dose administration. The results provide a basis for study design and data evaluation in the future development of recombinant FSH products, and show it can be of importance to account for endogenous FSH levels and its variation over time for accurate estimation of exogenously administered FSH pharmacokinetic parameters. Thus, correcting FSH concentrations by the observed endogenous FSH baseline value at all time points may be incorrect.


Subject(s)
Follicle Stimulating Hormone, Human/blood , Recombinant Proteins/pharmacokinetics , Adult , Dose-Response Relationship, Drug , Double-Blind Method , Estradiol/blood , Female , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/pharmacokinetics , Half-Life , Humans , Immunoassay , Inhibins/blood , Progesterone/blood , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Treatment Outcome , Young Adult
11.
Drugs R D ; 16(2): 173-80, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27003895

ABSTRACT

OBJECTIVE: The purpose of this analysis was to develop a population pharmacokinetic model for a novel recombinant human follicle-stimulating hormone (FSH) (FE 999049) expressed from a human cell line of foetal retinal origin (PER.C6(®)) developed for controlled ovarian stimulation prior to assisted reproductive technologies. METHODS: Serum FSH levels were measured following a single subcutaneous FE 999049 injection of 37.5, 75, 150, 225 or 450 IU in 27 pituitary-suppressed healthy female subjects participating in this first-in-human single ascending dose trial. Data was analysed by nonlinear mixed effects population pharmacokinetic modelling in NONMEM 7.2.0. RESULTS: A one-compartment model with first-order absorption and elimination rates was found to best describe the data. A transit model was introduced to describe a delay in the absorption process. The apparent clearance (CL/F) and apparent volume of distribution (V/F) estimates were found to increase with body weight. Body weight was included as an allometrically scaled covariate with a power exponent of 0.75 for CL/F and 1 for V/F. CONCLUSIONS: The single-dose pharmacokinetics of FE 999049 were adequately described by a population pharmacokinetic model. The average drug concentration at steady state is expected to be reduced with increasing body weight.


Subject(s)
Follicle Stimulating Hormone, Human/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Adult , Area Under Curve , Contraceptives, Oral, Combined , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follicle Stimulating Hormone, Human/administration & dosage , Follicle Stimulating Hormone, Human/blood , Humans , Immunoassay , Ovulation Induction , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Treatment Outcome , Young Adult
12.
J Pharmacokinet Pharmacodyn ; 40(4): 487-96, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23843051

ABSTRACT

This work explores the advantages of a model based drug development (MBDD) approach for the design and analysis of antiretroviral phase II trials. Two different study settings were investigated: (1) a 5-arm placebo-controlled parallel group dose-finding/proof of concept (POC) study and (2) a comparison of investigational drug and competitor. Studies were simulated using a HIV-1 dynamics model in NONMEM. The Monte-Carlo Mapped Power method determined the sample size required for detecting a dose-response relationship and a significant difference in effect compared to the competitor using a MBDD approach. Stochastic simulation and re-estimation were used for evaluation of model parameter precision and bias given different sample sizes. Results were compared to those from an unpaired, two-sided t test and ANOVA (p ≤ 0.05). In all scenarios, the MBDD approach resulted in smaller study sizes and more precisely estimated treatment effect than conventional statistical analysis. Using a MBDD approach, a sample size of 15 patients could be used to show POC and estimate ED50 with a good precision (relative standard error, 25.7 %). A sample size of 10 patients per arm was needed using the MBDD approach for detecting a difference in treatment effect of ≥20 % at 80 % power, a 3.4-fold reduction in sample size compared to a t test. The MBDD approach can be used to achieve more precise dose-response characterization facilitating decision making and dose selection. If necessitated, the sample size needed to reach a desired power can potentially be reduced compared to traditional statistical analyses. This may allow for comparison against competitors already in early clinical studies.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Clinical Trials, Phase II as Topic/methods , Drug Discovery/methods , HIV Infections/drug therapy , HIV-1 , Models, Statistical , Computer Simulation , Dose-Response Relationship, Drug , Humans , Monte Carlo Method , Research Design , Sample Size
13.
AAPS J ; 15(2): 308-15, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23224752

ABSTRACT

Atazanavir increases plasma bilirubin levels in a concentration-dependent manner. Due to less costly and readily available assays, bilirubin has been proposed as a marker of atazanavir exposure. In this work, a previously developed nomogram for detection of suboptimal atazanavir exposure is validated against external patient populations. The bilirubin nomogram was validated against 311 matching bilirubin and atazanavir samples from 166 HIV-1-infected Norwegian, French, and Italian patients on a ritonavir-boosted regimen. In addition, the nomogram was evaluated in 56 Italian patients on an unboosted regimen. The predictive properties of the nomogram were validated against observed atazanavir plasma concentrations. The use of the nomogram to detect non-adherence was also investigated by simulation. The bilirubin nomogram predicted suboptimal exposure in the patient populations on a ritonavir-boosted regimen with a negative predictive value of 97% (95% CI 95-100). The bilirubin nomogram and monitoring of atazanavir concentrations had similar predictive properties for detecting non-adherence based on simulations. Although both methods performed adequately during a period of non-adherence, they had lower predictive power to detect past non-adherence episodes. Using the bilirubin nomogram for detection of suboptimal atazanavir exposure in patients on a ritonavir-boosted regimen is a rapid and cost-effective alternative to routine measurements of the actual atazanavir exposure in plasma. Its application may be useful in clinical settings if atazanavir concentrations are not available.


Subject(s)
Bilirubin/blood , Drug Monitoring/methods , HIV Infections/drug therapy , HIV Protease Inhibitors/blood , HIV-1/pathogenicity , Nomograms , Oligopeptides/blood , Pyridines/blood , Adult , Atazanavir Sulfate , Biomarkers/blood , Computer Simulation , Drug Therapy, Combination , Europe , Female , HIV Infections/blood , HIV Infections/virology , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/pharmacokinetics , Humans , Male , Medication Adherence , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Reproducibility of Results , Ritonavir/administration & dosage
14.
Int J Clin Pharmacol Ther ; 50(11): 765-77, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22943931

ABSTRACT

OBJECTIVES: AZD1656 is a novel glucokinase activator with a postulated dual mechanism of action by activating glucokinase in both the pancreas and the liver, and with the potential to deliver effective glucose-lowering in Type 2 diabetes mellitus. Here, we present the tolerability, pharmacokinetics and pharmacodynamics of AZD1656 in two single-blind, randomized, placebo-controlled studies, one with Western and the other with Japanese healthy adult male subjects. METHODS: Both studies evaluated oral single ascending doses of AZD1656 of up to 180 mg, administered during euglycemic clamp conditions to explore a wide dose range without risking hypoglycemia. Safety, pharmacokinetics and effects on serum insulin and glucose infusion rate were assessed. A population pharmacokinetics analysis was also conducted. RESULTS: AZD1656 was well tolerated in single doses up to 180 mg in both populations. AZD1656 was rapidly absorbed, and a dose-proportional increase in total exposure was observed for AZD1656 and the equipotent metabolite, AZD5658. Taking differences in body weight into account, there were no differences in pharmacokinetic parameters between Western and Japanese subjects. A dose-dependent blood glucose lowering effect was indirectly demonstrated by the increased glucose infusion rate required to maintain euglycemia, which was of similar magnitude in both populations. Dose-dependent increases in insulin secretion were also observed. CONCLUSIONS: No safety concerns were raised. AZD1656 displayed uncomplicated pharmacokinetics and dose-dependent pharmacodynamics effects were observed. The results suggest no ethnic differences in AZD1656 tolerability, pharmacokinetics or pharmacodynamics.


Subject(s)
Azetidines/administration & dosage , Azetidines/pharmacokinetics , Blood Glucose/drug effects , Enzyme Activators/administration & dosage , Enzyme Activators/pharmacokinetics , Glucokinase/metabolism , Glucose Clamp Technique , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Pyrazines/administration & dosage , Pyrazines/pharmacokinetics , Administration, Oral , Adult , Asian People , Azetidines/adverse effects , Biomarkers/blood , Blood Glucose/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Activators/adverse effects , Humans , Hypoglycemic Agents/adverse effects , Insulin/blood , Male , Middle Aged , Models, Biological , Pyrazines/adverse effects , Single-Blind Method , United States , Young Adult
15.
J Pharmacokinet Pharmacodyn ; 38(6): 727-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21964996

ABSTRACT

The objective of this analysis was to compare three methods of handling HIV-RNA data below the limit of quantification (LOQ) when describing the time-course of antiretroviral drug response using a drug-disease model. Treatment naïve Scandinavian HIV-positive patients (n = 242) were randomized to one of three study arms. Two nucleoside reverse transcriptase inhibitors were administrated in combination with 400/100 mg lopinavir/ritonavir twice daily, 300/100 mg atazanavir/ritonavir once a day or 600 mg efavirenz once a day. The viral response was monitored at screening, baseline and at 1, 2, 3, 4, 12, 24, 48, 96, 120, and 144 weeks after study initiation. Data up to 400 days was fitted using a viral dynamics non-linear mixed effects drug-disease model in NONMEM. HIV-RNA data below LOQ of 50 copies/ml plasma (39%) was omitted, replaced by LOQ/2 or included in the analysis using a likelihood-based method (M3 method). Including data below LOQ using the M3 method substantially improved the model fit. The drug response parameter expressing the fractional inhibition of viral replication was on average (95% CI) estimated to 0.787 (0.721-0.864) for lopinavir and atazanavir treatment arms and 0.868 (0.796-0.923) for the efavirenz containing regimen. At 400 days after treatment initiation 90% (76-100) of the lopinavir and atazanavir treated patients were predicted to have undetectable viral levels and 96% (89-100%) for the efavirenz containing treatment. Including viral data below the LOQ rather than omitting or replacing data provides advantages such as better model predictions and less biased parameter estimates which are of importance when quantifying antiretroviral drug response.


Subject(s)
Anti-HIV Agents/therapeutic use , Benzoxazines/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Lopinavir/therapeutic use , Nonlinear Dynamics , Oligopeptides/therapeutic use , Pyridines/therapeutic use , Ritonavir/therapeutic use , Viral Load/statistics & numerical data , Adult , Aged , Alkynes , Anti-HIV Agents/administration & dosage , Atazanavir Sulfate , Benzoxazines/administration & dosage , Cyclopropanes , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination/statistics & numerical data , Female , HIV Infections/virology , Humans , Limit of Detection , Lopinavir/administration & dosage , Male , Middle Aged , Oligopeptides/administration & dosage , Pyridines/administration & dosage , RNA, Viral/drug effects , Ritonavir/administration & dosage
16.
AAPS J ; 13(4): 598-605, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21913053

ABSTRACT

The objective of this work was to examine the atazanavir-bilirubin relationship using a population-based approach and to assess the possible application of bilirubin as a readily available marker of atazanavir exposure. A model of atazanavir exposure and its concentration-dependent effect on bilirubin levels was developed based on 200 atazanavir and 361 bilirubin samples from 82 patients receiving atazanavir in the NORTHIV trial. The pharmacokinetics was adequately described by a one-compartment model with first-order absorption and lag-time. The maximum inhibition of bilirubin elimination rate constant (I(max)) was estimated at 91% (95% CI, 87-94) and the atazanavir concentration resulting in half of I(max) (IC50) was 0.30 µmol/L (95% CI, 0.24-0.37). At an atazanavir/ritonavir dose of 300/100 mg given once daily, the bilirubin half-life was on average increased from 1.6 to 8.1 h. A nomogram, which can be used to indicate suboptimal atazanavir exposure and non-adherence, was constructed based on model simulations.


Subject(s)
Bilirubin/blood , Biomarkers/blood , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Atazanavir Sulfate , HIV Infections/blood , HIV Protease Inhibitors/therapeutic use , Humans , Oligopeptides/therapeutic use , Pyridines/therapeutic use
17.
Clin Pharmacokinet ; 50(8): 531-40, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21740076

ABSTRACT

BACKGROUND: Pharmacokinetic studies of antiretroviral drugs are often conducted in adult healthy subjects, and the results are extrapolated to HIV/AIDS patients. HIV/AIDS, however, is known to cause morphological and physiological changes that may alter the pharmacokinetics of antiretroviral drugs. We examined the effect of HIV/AIDS on the pharmacokinetics of efavirenz in Ugandans. METHODS: After a first oral dose of efavirenz 600 mg in treatment-naïve HIV-infected patients, blood samples were collected at nine time points up to 24 hours. The plasma-concentration time data from these patients were merged with previously reported data from adult healthy subjects. Population pharmacokinetic models were fitted to the data, using NONMEM VI software. Covariate analyses were performed to estimate the effects of HIV/AIDS disease, demographic characteristics (sex, bodyweight, age), biochemical variables (serum creatinine, urea, alanine aminotransferase) and pharmacogenetic variation in cytochrome P450 (CYP) 2B6, CYP3A5 and adenosine triphosphate-binding cassette, sub-family B, member 1 (ABCB1) on the population pharmacokinetic parameters. RESULTS: Efavirenz plasma concentration-time data obtained from 29 HIV-1-infected, treatment-naïve patients were merged with previously reported data from 32 adult healthy subjects. The model identified sex and HIV/AIDS disease as statistically significant categorical predictors of efavirenz pharmacokinetics. Females were predicted to have a 2-fold higher volume of distribution of the peripheral compartment after oral administration (V(2)/F) than males (95% CI 1.53, 2.63), while HIV/AIDS patients were found to have 30% lower relative bioavailability (95% CI 18.7, 40.7) than healthy subjects. The increased V(2)/F in females resulted in a 2-fold longer elimination half-life than in males. CONCLUSION: On the basis of the findings of this analysis, we conclude that, apart from bodyweight-based differences, both HIV/AIDS disease and sex affect efavirenz pharmacokinetics in Ugandans. HIV/AIDS disease is associated with reduced relative bioavailability of efavirenz. We recommend that findings from healthy subject studies be confirmed in HIV/AIDS patients and that caution be applied in direct extrapolation of exposure data to the target patient population.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacokinetics , Benzoxazines/pharmacokinetics , HIV Infections/drug therapy , Administration, Oral , Adolescent , Adult , Alkynes , Biological Availability , Body Weight , Cyclopropanes , Female , Half-Life , Humans , Male , Middle Aged , Models, Biological , Nonlinear Dynamics , Pharmacogenetics , Sex Factors , Tissue Distribution , Uganda , Young Adult
18.
Br J Clin Pharmacol ; 71(4): 536-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21395646

ABSTRACT

AIMS: This study aimed to test whether a pharmacokinetic simulation model could extrapolate nonclinical drug data to predict human efavirenz exposure after single and continuous dosing as well as the effects of concomitant rifampicin and further to evaluate the weight-based dosage recommendations used to counteract the rifampicin-efavirenz interaction. METHODS: Efavirenz pharmacokinetics were simulated using a physiologically based pharmacokinetic model implemented in the Simcyp™ population-based simulator. Physicochemical and metabolism data obtained from the literature were used as input for prediction of pharmacokinetic parameters. The model was used to simulate the effects of rifampicin on efavirenz pharmacokinetics in 400 virtual patients, taking into account bodyweight and CYP2B6 phenotype. RESULTS: Apart from the absorption phase, the simulation model predicted efavirenz concentration-time profiles reasonably well, with close agreement with clinical data. The simulated effects of rifampicin co-administration on efavirenz treatment showed only a minor decrease of 16% (95% confidence interval 13-19) in efavirenz area under the concentration-time curve, of the same magnitude as what has been clinically observed (22%). Efavirenz exposure depended on CYP2B6 phenotype and bodyweight. Increasing the efavirenz dose during concomitant rifampicin was predicted to be most successful in patients over 50 kg regardless of CYP2B6 status. CONCLUSIONS: Our findings, although based on a simulation approach using limited in vitro data, support the current recommendations for using a 50 kg bodyweight cut-off for efavirenz dose increment when co-treating with rifampicin.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Antibiotics, Antitubercular/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/genetics , Benzoxazines/pharmacokinetics , HIV Infections/drug therapy , Oxidoreductases, N-Demethylating/genetics , Rifampin/pharmacokinetics , Tuberculosis/drug therapy , Alkynes , Anti-HIV Agents/therapeutic use , Antibiotics, Antitubercular/therapeutic use , Benzoxazines/therapeutic use , Body Weight/drug effects , Computer Simulation , Cyclopropanes , Cytochrome P-450 CYP2B6 , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Forecasting/methods , HIV Infections/genetics , Humans , Models, Theoretical , Phenotype , Rifampin/therapeutic use , Tuberculosis/genetics
19.
Br J Clin Pharmacol ; 68(5): 690-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19916993

ABSTRACT

AIMS: Efavirenz exhibits pharmacokinetic variability causing varied clinical response. The aim was to develop an integrated population pharmacokinetic/pharmacogenetic model and investigate the impact of genetic variations, sex, demographic and biochemical variables on single-dose efavirenz pharmacokinetics among Ugandan subjects, using NONMEM. METHODS: Efavirenz plasma concentrations (n = 402) from 121 healthy subjects were quantified by high-performance liquid chromatography. Subjects were genotyped for 30 single nucleotide polymorphisms (SNPs), of which six were novel SNPs in CYP2B6, CYP3A5 and ABCB1. The efavirenz pharmacokinetics was described by a two-compartment model with zero- followed by first-order absorption. RESULTS: Apparent oral clearance (95% confidence interval) was 4 l h l(-1) (3.5, 4.5) in extensive metabolizers. In the final model, incorporating multiple covariates, statistical significance was found only for CYP2B6*6 and CYP2B6*11 on apparent oral clearance as well as ABCB1 (rs3842) on the relative bioavailability. Subjects homozygous for CYP2B6*6 (G516T, A785G) and *11 displayed 21 and 20% lower apparent oral clearance, respectively. Efavirenz relative bioavailability was 26% higher in subjects homozygous for ABCB1 (rs3842). The apparent peripheral volume of distribution was twofold higher in women compared with men. CONCLUSIONS: The model identified the four factors CYP2B6*6, CYP2B6*11, a novel variant allele in ABCB1 (rs3842) and sex as major predictors of efavirenz plasma exposure in a healthy Ugandan population after single-dose administration. Use of mixed-effects modelling allowed the analysis and integration of multiple pharmacogenetic and demographic covariates in a pharmacokinetic population model.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Benzoxazines/pharmacokinetics , Black People/genetics , Polymorphism, Genetic/genetics , Adult , Alkynes , Cyclopropanes , Dose-Response Relationship, Drug , Female , Humans , Male , Models, Biological , Models, Genetic , Sex Factors , Uganda , Young Adult
20.
Eur J Clin Pharmacol ; 64(4): 357-65, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18057928

ABSTRACT

OBJECTIVE: The study sought to investigate the relationship between efavirenz exposure and the CYP2B6 516G-->T(*6) genotype in HIV/AIDS outpatients, using pharmacokinetic modelling and simulation. METHODS: Blood samples where obtained from 74 outpatients treated with a combination regimen including 600 mg efavirenz daily for a duration of at least 3 weeks at clinics in Harare, Zimbabwe. The subjects were genotyped for the major CYP2B6 variant, CYP2B6*6, associated with reduced enzyme activity, using a PCR-RFLP method. Efavirenz plasma concentrations were determined by HPLC-UV. Population pharmacokinetic modelling and simulation of the data were performed in NONMEM VI. RESULTS: A high allele frequency of the CYP2B6*6 allele of 49% was observed. Efavirenz plasma concentrations were above 4 mg/L in 50% of the patients. Genotype and sex were identified as predictive covariates of efavirenz disposition. Pharmacokinetic parameter estimates indicate that a dose reduction to 400 mg efavirenz per day is possible in patients homozygous for the CYP2B6*6 genotype without compromising therapeutic efficacy. CONCLUSION: The CYP2B6*6 allele occurs at a high frequency in people of African origin and is associated with high efavirenz concentrations. Simulations indicate that an a priori 35% dose reduction in homozygous CYP2B6*6 patients would maintain drug exposure within the therapeutic range in this group of patients. Our preliminary results suggest the conduct of a prospective clinical dose optimization study to evaluate the utility of genotype-driven dose adjustment in this population.


Subject(s)
Acquired Immunodeficiency Syndrome/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Benzoxazines/pharmacokinetics , HIV Infections/genetics , Oxidoreductases, N-Demethylating/genetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/virology , Adult , Algorithms , Alkynes , Benzoxazines/administration & dosage , Benzoxazines/therapeutic use , Cyclopropanes , Cytochrome P-450 CYP2B6 , Female , Gene Frequency , Genetic Variation , Genotype , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/virology , Humans , Male , Models, Statistical , Population , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/therapeutic use , Sex Factors , Zimbabwe/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...