Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Infect Dis Ther ; 12(7): 1861-1873, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37329415

ABSTRACT

INTRODUCTION: Bamlanivimab and etesevimab (BAM + ETE) are monoclonal antibodies (mAbs) effective in reducing COVID-19-related hospitalizations and all-cause mortality in adult participants at increased risk for severe disease. We present pharmacokinetic (PK), efficacy, and safety results from pediatric participants (< 18 years of age) with COVID-19 who were treated with BAM + ETE. METHODS: In an addendum to the phase 2/3 BLAZE-1 clinical trial (NCT04427501), pediatric participants received open-label weight-based dosing (WBD, n = 94) based on exposure-matching to the authorized dose of BAM + ETE in adult participants. For efficacy and safety assessments, placebo (n = 14) and BAM + ETE (n = 20)-treated adolescent participants (> 12 to < 18 years of age) from the BLAZE-1 trial were included in the overall pediatric population (N = 128). All participants had mild to moderate COVID-19 upon enrollment and ≥ 1 risk factor for severe COVID-19. The primary objective was to characterize the PK of BAM and ETE in the WBD population. RESULTS: The median age of the participants was 11.2 years, 46.1% were female, 57.9% were Black/African American, and 19.7% were Hispanic/Latino. The area under the curve for BAM and ETE in the WBD population was similar to that previously observed in adults. There were no COVID-19-related hospitalizations or deaths. All adverse events (AE) except one were mild or moderate, with one participant reporting a serious AE. CONCLUSION: WBD in pediatric participants achieved similar drug exposures compared to adult participants that received the authorized BAM + ETE dose. The pediatric efficacy and safety data were consistent with adults receiving mAbs for COVID-19. TRIAL REGISTRATION NUMBER: NCT04427501.

2.
CPT Pharmacometrics Syst Pharmacol ; 11(11): 1443-1457, 2022 11.
Article in English | MEDLINE | ID: mdl-35899461

ABSTRACT

Glycated hemoglobin (HbA1c) is the main biomarker of diabetes drug development. However, because of its delayed turnover, trial duration is rarely shorter than 12 weeks, and being able to predict long-term HbA1c with precision using data from shorter studies would be beneficial. The feasibility of reducing study duration was therefore investigated in this study, assuming a model-based analysis. The aim was to investigate the predictive performance of 24- and 52-week extrapolations using data from up to 4, 6, 8 or 12 weeks, with six previously published pharmacometric models of HbA1c. Predictive performance was assessed through simulation-based dose-response predictions and model averaging (MA) with two hypothetical drugs. Results were consistent across the methods of assessment, with MA supporting the results derived from the model-based framework. The models using mean plasma glucose (MPG) or nonlinear fasting plasma glucose (FPG) effect, driving the HbA1c formation, showed good predictive performance despite a reduced study duration. The models, using the linear effect of FPG to drive the HbA1c formation, were sensitive to the limited amount of data in the shorter studies. The MA with bootstrap demonstrated strongly that a 4-week study duration is insufficient for precise predictions of all models. Our findings suggest that if data are analyzed with a pharmacometric model with MPG or FPG with a nonlinear effect to drive HbA1c formation, a study duration of 8 weeks is sufficient with maintained accuracy and precision of dose-response predictions.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 2 , Humans , Glycated Hemoglobin , Diabetes Mellitus, Type 2/drug therapy , Fasting , Biomarkers
3.
CPT Pharmacometrics Syst Pharmacol ; 11(6): 721-730, 2022 06.
Article in English | MEDLINE | ID: mdl-35289125

ABSTRACT

The relationship between severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) viral load reduction and disease symptom resolution remains largely undefined for coronavirus disease 2019 (COVID-19). While the vaccine-derived immunity takes time to develop, neutralizing monoclonal antibodies offer immediate, passive immunity to patients with COVID-19. Bamlanivimab and etesevimab are two potent neutralizing monoclonal antibodies directed to the receptor binding domain of the spike protein of SARS-CoV-2. This study aims to describe the relationship between viral load and resolution of eight common COVID-19-related symptoms in patients following treatment with neutralizing monoclonal antibodies (bamlanivimab alone or bamlanivimab and etesevimab together), in a phase II clinical trial. Corresponding pharmacokinetics (PKs), viral load, and COVID-19-related symptom data were modeled using Nonlinear Mixed Effects Modeling to describe the time-course of eight COVID-19-related symptoms in an ordered categorical manner (none, mild, moderate, and severe), following administration of bamlanivimab or bamlanivimab and etesevimab together to participants with COVID-19. The PK/pharmacodynamic (PD) models characterized the exposure-viral load-symptom time course of the eight preselected COVID-19-related symptoms. Baseline viral load (BVL), change in viral load from baseline, and time since the onset of symptoms, demonstrated statistically significant effects on symptom score probabilities. Higher BVL generally indicated an increased probability of symptom severity. The severity of symptoms decreased over time, partially driven by the decrease in viral load. The effect of increasing time resulting in decreased severity of symptoms was over and above the effect of decreasing viral load. Administration of bamlanivimab alone or together with etesevimab results in a faster time to resolution of COVID-19-related symptoms compared to placebo.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing , Antibodies, Viral , Humans , Viral Load
4.
Clin Pharmacol Ther ; 111(3): 595-604, 2022 03.
Article in English | MEDLINE | ID: mdl-34687040

ABSTRACT

Neutralizing monoclonal antibodies (mAb), novel therapeutics for the treatment of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), have been urgently researched from the start of the pandemic. The selection of the optimal mAb candidate and therapeutic dose were expedited using open-access in silico models. The maximally effective therapeutic mAb dose was determined through two approaches; both expanded on innovative, open-science initiatives. A physiologically-based pharmacokinetic (PBPK) model, incorporating physicochemical properties predictive of mAb clearance and tissue distribution, was used to estimate mAb exposure that maintained concentrations above 90% inhibitory concentration of in vitro neutralization in lung tissue for up to 4 weeks in 90% of patients. To achieve fastest viral clearance following onset of symptoms, a longitudinal SARS-CoV-2 viral dynamic model was applied to estimate viral clearance as a function of drug concentration and dose. The PBPK model-based approach suggested that a clinical dose between 175 and 500 mg of bamlanivimab would maintain target mAb concentrations in the lung tissue over 28 days in 90% of patients. The viral dynamic model suggested a 700 mg dose would achieve maximum viral elimination. Taken together, the first-in-human trial (NCT04411628) conservatively proceeded with a starting therapeutic dose of 700 mg and escalated to higher doses to evaluate the upper limit of safety and tolerability. Availability of open-access codes and application of novel in silico model-based approaches supported the selection of bamlanivimab and identified the lowest dose evaluated in this study that was expected to result in the maximum therapeutic effect before the first-in-human clinical trial.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antiviral Agents/administration & dosage , Models, Biological , SARS-CoV-2/drug effects , Antibodies, Monoclonal/pharmacokinetics , Antiviral Agents/pharmacokinetics , Clinical Trials as Topic , Computer Simulation , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Humans , SARS-CoV-2/immunology
5.
J Clin Pharmacol ; 61(2): 234-243, 2021 02.
Article in English | MEDLINE | ID: mdl-32895980

ABSTRACT

Weight loss has been associated with improvement in insulin sensitivity. It is consequently a cornerstone in the management of type 2 diabetes mellitus (T2DM). However, the strictly quantitative relationship between weight loss, insulin sensitivity, and clinically relevant glucose homeostasis biomarkers as well as changes therein as T2DM progresses is not yet fully understood. Therefore, the objective of our research was to establish a body weight-directed disease trial model for glucose homeostasis. To that end, we conducted a model-based meta-analysis using time course data of body weight loss (following lifestyle change or surgical procedure) and corresponding improvement of insulin sensitivity expressed as the Matsuda index. Changes in body weight were best described by a sigmoidal Emax model, whereas changes in the Matsuda index were best described by a linear model with a slope of 3.49. Once developed and verified, the model-based meta-analysis was linked to a disease-drug trial model for T2DM previously developed by our group to characterize and predict the impact of weight loss on clinically relevant glucose homeostasis biomarkers. The joint model was then used to conduct clinical trial simulations, which showed that weight loss can greatly improve clinically relevant glucose homeostasis biomarkers in T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance/physiology , Models, Biological , Weight Loss/physiology , Biomarkers , Blood Glucose , Glycated Hemoglobin , Humans , Insulin/blood
6.
Clin Pharmacol Ther ; 104(4): 699-708, 2018 10.
Article in English | MEDLINE | ID: mdl-29271001

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a chronic, progressive disease characterized by persistently elevated blood glucose concentration (hyperglycemia). We developed a mechanistic drug-disease modeling platform based on data from more than 4,000 T2DM subjects in seven phase II/III clinical trials. The model integrates longitudinal changes in clinically relevant biomarkers of glycemic control with information on baseline disease state, demographics, disease progression, and different therapeutic interventions, either when given alone or as add-on combination therapy. The model was able to simultaneously characterize changes in fasting plasma glucose, fasting serum insulin, and glycated hemoglobin A1c following administration of sulfonylurea, metformin, and thiazolidinedione as well as disease progression in clinical trials ranging from 16-104 weeks of treatment. The mechanistic components of this generalized mechanism-based platform, based on knowledge of pharmacology, insulin-glucose homeostatic feedback, and diabetes pathophysiology, allows its application to be further expanded to other antidiabetic drug classes and combination therapies.


Subject(s)
Blood Glucose/drug effects , Clinical Trials as Topic/methods , Diabetes Mellitus, Type 2/drug therapy , Endpoint Determination , Hypoglycemic Agents/therapeutic use , Models, Biological , Research Design , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Insulin/blood , Male , Metformin/therapeutic use , Middle Aged , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/therapeutic use , Time Factors , Treatment Outcome
7.
Diabetes Technol Ther ; 19(8): 483-490, 2017 08.
Article in English | MEDLINE | ID: mdl-28700249

ABSTRACT

BACKGROUND: The purpose of this prospective, model-based simulation approach was to evaluate the impact of various rapid-acting mealtime insulin dose-titration algorithms on glycemic control (hemoglobin A1c [HbA1c]). METHODS: Seven stepwise, glucose-driven insulin dose-titration algorithms were evaluated with a model-based simulation approach by using insulin lispro. Pre-meal blood glucose readings were used to adjust insulin lispro doses. Two control dosing algorithms were included for comparison: no insulin lispro (basal insulin+metformin only) or insulin lispro with fixed doses without titration. RESULTS: Of the seven dosing algorithms assessed, daily adjustment of insulin lispro dose, when glucose targets were met at pre-breakfast, pre-lunch, and pre-dinner, sequentially, demonstrated greater HbA1c reduction at 24 weeks, compared with the other dosing algorithms. Hypoglycemic rates were comparable among the dosing algorithms except for higher rates with the insulin lispro fixed-dose scenario (no titration), as expected. The inferior HbA1c response for the "basal plus metformin only" arm supports the additional glycemic benefit with prandial insulin lispro. CONCLUSIONS: Our model-based simulations support a simplified dosing algorithm that does not include carbohydrate counting, but that includes glucose targets for daily dose adjustment to maintain glycemic control with a low risk of hypoglycemia.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin Glargine/administration & dosage , Insulin Lispro/administration & dosage , Metformin/administration & dosage , Models, Theoretical , Algorithms , Computer Simulation , Diabetes Mellitus, Type 2/blood , Dose-Response Relationship, Drug , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/therapeutic use , Insulin Glargine/therapeutic use , Insulin Lispro/therapeutic use , Metformin/therapeutic use
8.
Clin Pharmacokinet ; 55(7): 769-788, 2016 07.
Article in English | MEDLINE | ID: mdl-26798033

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease, which affects millions of people worldwide. The disease is characterized by chronically elevated blood glucose concentrations (hyperglycaemia), which result in comorbidities and multi-organ dysfunction. This is due to a gradual loss of glycaemic control as a result of increasing insulin resistance, as well as decreasing ß-cell function. The objective of T2DM drug interventions is, therefore, to reduce fasting and postprandial blood glucose concentrations to normal, healthy levels without hypoglycaemia. Several classes of novel antihyperglycaemic drugs with various mechanisms of action have been developed over the past decades or are currently under clinical development. The development of these drugs is routinely supported by the application of pharmacokinetic/pharmacodynamic modelling and simulation approaches. They integrate information on the drug's pharmacokinetics, clinically relevant biomarker information and disease progression into a single, unifying approach, which can be used to inform clinical study design, dose selection and drug labelling. The objective of this review is to provide a comprehensive overview of the quantitative approaches that have been reported since the 2008 review by Landersdorfer and Jusko in an increasing order of complexity, starting with glucose homeostasis models. Each of the presented approaches is discussed with respect to its strengths and limitations, and respective knowledge gaps are highlighted as potential opportunities for future drug-disease model development in the area of T2DM.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Models, Biological , Biomarkers , Blood Glucose/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/agonists , Glucokinase/biosynthesis , Glycated Hemoglobin , Humans , Hyperglycemia/prevention & control , Hypoglycemic Agents/pharmacokinetics , Incretins/biosynthesis , Insulin/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, Glucagon/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors
9.
Clin Pharmacokinet ; 55(5): 625-34, 2016 May.
Article in English | MEDLINE | ID: mdl-26507721

ABSTRACT

BACKGROUND AND OBJECTIVE: Dulaglutide is a long-acting glucagon-like peptide-1 receptor agonist administered as once-weekly subcutaneous injections for the treatment of type 2 diabetes (T2D). The clinical pharmacokinetics of dulaglutide were characterized in patients with T2D and healthy subjects. METHODS: The pharmacokinetics of dulaglutide were assessed throughout clinical development, including conventional pharmacokinetic analysis in clinical pharmacology studies and population pharmacokinetic analyses of data from combined phase 2 and phase 3 studies in patients with T2D. The effects of potential covariates on dulaglutide population pharmacokinetics were evaluated using nonlinear mixed-effects models. RESULTS: Dulaglutide gradually reached the maximum concentration in 48 h and had a terminal elimination half-life of 5 days. Steady state was achieved between the second and fourth doses. The accumulation ratio was 1.56 for the 1.5 mg dose. Intra-individual variability estimates for the area under the plasma concentration-time curve and the maximum concentration were both <17% [coefficient of variation (CV)]. There was no difference in pharmacokinetics between injection sites (arm, thigh or abdomen). Dulaglutide pharmacokinetics were well described by a two-compartment model with first-order absorption and elimination. The population clearance was estimated at 0.126 L/h [inter-individual variability (CV) 33.8%]. Age, body weight, sex, race and ethnicity did not influence dulaglutide pharmacokinetics to any clinically relevant degree. CONCLUSION: The pharmacokinetics of dulaglutide support once-weekly administration in patients with T2D. The pharmacokinetic findings suggest that dose adjustment is not necessary on the basis of body weight, sex, age, race or ethnicity or site of injection.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptides/analogs & derivatives , Hypoglycemic Agents/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Adult , Aged , Aged, 80 and over , Drug Administration Routes , Female , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/blood , Glucagon-Like Peptides/pharmacokinetics , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/blood , Male , Middle Aged , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Young Adult
10.
Cell Metab ; 18(3): 333-40, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24011069

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a recently discovered metabolic regulator. Exogenous FGF21 produces beneficial metabolic effects in animal models; however, the translation of these observations to humans has not been tested. Here, we studied the effects of LY2405319 (LY), a variant of FGF21, in a randomized, placebo-controlled, double-blind proof-of-concept trial in patients with obesity and type 2 diabetes. Patients received placebo or 3, 10, or 20 mg of LY daily for 28 days. LY treatment produced significant improvements in dyslipidemia, including decreases in low-density lipoprotein cholesterol and triglycerides and increases in high-density lipoprotein cholesterol and a shift to a potentially less atherogenic apolipoprotein concentration profile. Favorable effects on body weight, fasting insulin, and adiponectin were also detected. However, only a trend toward glucose lowering was observed. These results indicate that FGF21 is bioactive in humans and suggest that FGF21-based therapies may be effective for the treatment of selected metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factors/therapeutic use , Obesity/drug therapy , Adiponectin/blood , Adolescent , Adult , Aged , Blood Glucose/analysis , Body Mass Index , Body Weight , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/complications , Double-Blind Method , Dyslipidemias/complications , Dyslipidemias/drug therapy , Female , Humans , Insulin/blood , Male , Middle Aged , Obesity/complications , Placebo Effect , Triglycerides/blood , Young Adult
11.
Drug Metab Dispos ; 41(7): 1329-38, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23584886

ABSTRACT

Ketoconazole is a potent CYP3A inhibitor used to assess the contribution of CYP3A to drug clearance and quantify the increase in drug exposure due to a strong inhibitor. Physiologically based pharmacokinetic (PBPK) models have been used to evaluate treatment regimens resulting in maximal CYP3A inhibition by ketoconazole but have reached different conclusions. We compare two PBPK models of the ketoconazole-midazolam interaction, model 1 (Chien et al., 2006) and model 2 implemented in Simcyp (version 11), to predict 16 published treatment regimens. With use of model 2, 41% of the study point estimates of area under the curve (AUC) ratio and 71% of the 90% confidence intervals were predicted within 1.5-fold of the observed, but these increased to 82 and 100%, respectively, with model 1. For midazolam, model 2 predicted a maximal midazolam AUC ratio of 8 and a hepatic fraction metabolized by CYP3A (f(m)) of 0.97, whereas model 1 predicted 17 and 0.90, respectively, which are more consistent with observed data. On the basis of model 1, ketoconazole (400 mg QD) for at least 3 days and substrate administration within 2 hours is required for maximal CYP3A inhibition. Ketoconazole treatment regimens that use 200 mg BID underestimate the systemic fraction metabolized by CYP3A (0.86 versus 0.90) for midazolam. The systematic underprediction also applies to CYP3A substrates with high bioavailability and long half-lives. The superior predictive performance of model 1 reflects the need for accumulation of ketoconazole at enzyme site and protracted inhibition. Model 2 is not recommended for inferring optimal study design and estimation of fraction metabolized by CYP3A.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Midazolam/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Drug Interactions , Humans , Models, Biological , Research Design
12.
J Pharm Sci ; 101(8): 2668-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22573521

ABSTRACT

There are 94,709 clinical trials across 179 countries. Approximately half (47,467) are related to the three categories within the scope of the free online resource "Drug Delivery Trends in Clinical Trials and Translational Medicine," which are (1) drug delivery technology and systems, (2) biological molecule platforms, and (3) pharmacokinetic and pharmacodynamic interactions. In this commentary, trends in biological molecule platforms and their impacts are discussed. The sales of top 15 biologic drugs have reached over $63 billion in 2010. In the past 10 years, major pharmaceutical companies have acquired biological molecule platforms and have become integrated biopharmaceutical companies, highlighting the role of biotechnology in driving new therapeutic product development. The top three products--Remicade, Enbrel, and Humira--indicated for arthritis and colitis and targeted to tumor necrosis factor-alpha (TNF-α), each generated over $6 billion in annual sales. In addition to TNF-α, biologic candidates targeted to other inflammatory molecules are in clinical development, partly driven by commercial interests and medical need. Although clinical experience indicates that all the anti-TNF-α molecular platforms are effective for rheumatoid arthritis, Crohn's disease, and colitis, whether the new agents can provide additional relief or cures remains to be seen.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Colitis/drug therapy , Crohn Disease/drug therapy , Drug Delivery Systems/methods , Drug Discovery/methods , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Clinical Trials as Topic , Drug Delivery Systems/economics , Drug Discovery/economics , Humans , Pharmacokinetics , Translational Research, Biomedical
13.
J Pharm Sci ; 100(10): 4127-57, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21541937

ABSTRACT

The objective of this study is to assess the effectiveness of physiologically based pharmacokinetic (PBPK) models for simulating human plasma concentration-time profiles for the unique drug dataset of blinded data that has been assembled as part of a Pharmaceutical Research and Manufacturers of America initiative. Combinations of absorption, distribution, and clearance models were tested with a PBPK approach that has been developed from published equations. An assessment of the quality of the model predictions was made on the basis of the shape of the plasma time courses and related parameters. Up to 69% of the simulations of plasma time courses made in human demonstrated a medium to high degree of accuracy for intravenous pharmacokinetics, whereas this number decreased to 23% after oral administration based on the selected criteria. The simulations resulted in a general underestimation of drug exposure (Cmax and AUC0- t ). The explanations for this underestimation are diverse. Therefore, in general it may be due to underprediction of absorption parameters and/or overprediction of distribution or oral first-pass. The implications of compound properties are demonstrated. The PBPK approach based on in vitro-input data was as accurate as the approach based on in vivo data. Overall, the scientific benefit of this modeling study was to obtain more extensive characterization of predictions of human PK from PBPK methods.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Computer Simulation , Cooperative Behavior , Drug Evaluation, Preclinical , Gastrointestinal Absorption , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Reproducibility of Results , Species Specificity
14.
J Pharm Sci ; 100(10): 4090-110, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21541938

ABSTRACT

The objective of this study was to evaluate the performance of various allometric and in vitro-in vivo extrapolation (IVIVE) methodologies with and without plasma protein binding corrections for the prediction of human intravenous (i.v.) clearance (CL). The objective was also to evaluate the IVIVE prediction methods with animal data. Methodologies were selected from the literature. Pharmaceutical Research and Manufacturers of America member companies contributed blinded datasets from preclinical and clinical studies for 108 compounds, among which 19 drugs had i.v. clinical pharmacokinetics data and were used in the analysis. In vivo and in vitro preclinical data were used to predict CL by 29 different methods. For many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. In addition, 66 methods of predicting oral (p.o.) area under the curve (AUCp.o. ) were evaluated for 107 compounds using rational combinations of i.v. CL and bioavailability (F), and direct scaling of observed p.o. CL from preclinical species. Various statistical and outlier techniques were employed to assess the predictability of each method. Across methods, the maximum success rate in predicting human CL for the 19 drugs was 100%, 94%, and 78% of the compounds with predictions falling within 10-fold, threefold, and twofold error, respectively, of the observed CL. In general, in vivo methods performed slightly better than IVIVE methods (at least in terms of measures of correlation and global concordance), with the fu intercept method and two-species-based allometry (rat-dog) being the best performing methods. IVIVE methods using microsomes (incorporating both plasma and microsomal binding) and hepatocytes (not incorporating binding) resulted in 75% and 78%, respectively, of the predictions falling within twofold error. IVIVE methods using other combinations of binding assumptions were much less accurate. The results for prediction of AUCp.o. were consistent with i.v. CL. However, the greatest challenge to successful prediction of human p.o. CL is the estimate of F in human. Overall, the results of this initiative confirmed predictive performance of common methodologies used to predict human CL.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Animals , Area Under Curve , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Protein Binding , Rats , Reproducibility of Results , Species Specificity
15.
J Pharm Sci ; 100(10): 4050-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21523782

ABSTRACT

This study is part of the Pharmaceutical Research and Manufacturers of America (PhRMA) initiative on predictive models of efficacy, safety, and compound properties. The overall goal of this part was to assess the predictability of human pharmacokinetics (PK) from preclinical data and to provide comparisons of available prediction methods from the literature, as appropriate, using a representative blinded dataset of drug candidates. The key objectives were to (i) appropriately assemble and blind a diverse dataset of in vitro, preclinical in vivo, and clinical data for multiple drug candidates, (ii) evaluate the dataset with empirical and physiological methodologies from the literature used to predict human PK properties and plasma concentration-time profiles, (iii) compare the predicted properties with the observed clinical data to assess the prediction accuracy using routine statistical techniques and to evaluate prediction method(s) based on the degree of accuracy of each prediction method, and (iv) compile and summarize results for publication. Another objective was to provide a mechanistic understanding as to why one methodology provided better predictions than another, after analyzing the poor predictions. A total of 108 clinical lead compounds were collected from 12 PhRMA member companies. This dataset contains intravenous (n = 19) and oral pharmacokinetic data (n = 107) in humans as well as the corresponding preclinical in vitro, in vivo, and physicochemical data. All data were blinded to protect the anonymity of both the data and the company submitting the data. This manuscript, which is the first of a series of manuscripts, summarizes the PhRMA initiative and the 108 compound dataset. More details on the predictability of each method are reported in companion manuscripts.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Computer Simulation , Cooperative Behavior , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Program Development , Program Evaluation , Reproducibility of Results , Risk Assessment , Risk Factors , Species Specificity
16.
J Pharm Sci ; 100(10): 4074-89, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21452299

ABSTRACT

The objective of this study was to evaluate the performance of various empirical, semimechanistic and mechanistic methodologies with and without protein binding corrections for the prediction of human volume of distribution at steady state (Vss ). PhRMA member companies contributed a set of blinded data from preclinical and clinical studies, and 18 drugs with intravenous clinical pharmacokinetics (PK) data were available for the analysis. In vivo and in vitro preclinical data were used to predict Vss by 24 different methods. Various statistical and outlier techniques were employed to assess the predictability of each method. There was not simply one method that predicts Vss accurately for all compounds. Across methods, the maximum success rate in predicting human Vss was 100%, 94%, and 78% of the compounds with predictions falling within tenfold, threefold, and twofold error, respectively, of the observed Vss . Generally, the methods that made use of in vivo preclinical data were more predictive than those methods that relied solely on in vitro data. However, for many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. It is recommended to initially use the in vitro tissue composition-based equations to predict Vss in preclinical species and humans, putting the assumptions and compound properties into context. As in vivo data become available, these predictions should be reassessed and rationalized to indicate the level of confidence (uncertainty) in the human Vss prediction. The top three methods that perform strongly at integrating in vivo data in this way were the Øie-Tozer, the rat -dog-human proportionality equation, and the lumped-PBPK approach. Overall, the scientific benefit of this study was to obtain greater characterization of predictions of human Vss from several methods available in the literature.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Animals , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Humans , Interdisciplinary Communication , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Protein Binding , Rats , Reproducibility of Results , Species Specificity
17.
J Pharm Sci ; 100(10): 4111-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21480234

ABSTRACT

The objective of this study was to evaluate the performance of the Wajima allometry (Css -MRT) approach published in the literature, which is used to predict the human plasma concentration-time profiles from a scaling of preclinical species data. A diverse and blinded dataset of 108 compounds from PhRMA member companies was used in this evaluation. The human intravenous (i.v.) and oral (p.o.) pharmacokinetics (PK) data were available for 18 and 107 drugs, respectively. Three different scenarios were adopted for prediction of human PK profiles. In the first scenario, human clearance (CL) and steady-state volume of distribution (Vss ) were predicted by unbound fraction corrected intercept method (FCIM) and Øie-Tozer (OT) approaches, respectively. Quantitative structure activity relationship (QSAR)-based approaches (TSrat-dog ) based on compound descriptors together with rat and dog data were utilized in the second scenario. Finally, in the third scenario, CL and Vss were predicted using the FCIM and Jansson approaches, respectively. For the prediction of oral pharmacokinetics, the human bioavailability and absorption rate constant were assumed as the average of preclinical species. Various statistical techniques were used for assessing the accuracy of the simulation scenarios. The human CL and Vss were predicted within a threefold error range for about 75% of the i.v. drugs. However, the accuracy in predicting key p.o. PK parameters appeared to be lower with only 58% of simulations falling within threefold of observed parameters. The overall ability of the Css -MRT approach to predict the curve shape of the profile was in general poor and ranged between low to medium level of confidence for most of the predictions based on the selected criteria.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Access to Information , Administration, Intravenous , Administration, Oral , Animals , Biological Availability , Computer Simulation , Cooperative Behavior , Dogs , Drug Evaluation, Preclinical , Gastrointestinal Absorption , Humans , Interdisciplinary Communication , Metabolic Clearance Rate , Models, Statistical , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Program Development , Program Evaluation , Rats , Reproducibility of Results , Species Specificity
18.
J Pharm Sci ; 100(1): 53-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20589750

ABSTRACT

In spite of the recent advances in technology to optimize the absorption, distribution, metabolism and elimination (ADME) properties of new and promising medicinal products to reduce clinical failures, the investigation of drug disposition in the pediatric and elderly populations continues to be under evaluated. With the increasing prevalence of aging populations world-wide, there is a growing concern from health care providers, regulators and the general public that drug delivery is still less than optimal for the vulnerable patient populations likely to be more sensitive to adverse effects of the new investigational drugs. This review of the ClinicalTrials.gov database revealed a rapidly increasing number of clinical trials and a trend towards wider inclusion criteria of the elderly population in clinical trials over the past 10 years. However, when we summarized trials by drug delivery, biological platforms, and disease categories, less than 10% of these trials included pharmacokinetic evaluation in elderly subjects greater than 65 years of age, and less than 4% included pharmacokinetic evaluation in children less than 17 years of age. Across the various disease areas, the percentage of trials that included pharmacokinetic evaluation in the children and elderly has remained low and is consistently less than the studies that included the younger 18 to 65 age group. Therefore, it is not known whether the right information is being generated from the growing number of clinical trials to guide optimal dosing recommendations in special patient populations.


Subject(s)
Drug Delivery Systems , Drugs, Investigational/pharmacokinetics , Adolescent , Aged , Aged, 80 and over , Aging , Animals , Child , Child, Preschool , Clinical Trials as Topic/statistics & numerical data , Clinical Trials as Topic/trends , Databases, Factual , Drug Delivery Systems/adverse effects , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Humans , Infant , National Institutes of Health (U.S.) , United States
19.
Br J Clin Pharmacol ; 70(4): 523-36, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20840444

ABSTRACT

AIMS: To develop a population pharmacokinetic model to describe the pharmacokinetics of desipramine in healthy subjects, after oral administration of a 50mg dose. Additional objectives were to develop a semi-mechanistic population pharmacokinetic model for desipramine, which allowed simulation of CYP2D6-mediated inhibition, when using desipramine as a probe substrate, and to evaluate certain study design elements, such as duration of desipramine pharmacokinetic sampling, required sample size and optimal pharmacokinetic sampling schedule for intermediate, extensive and ultrarapid metabolizers of CYP2D6 substrates. RESULTS: The mean population estimates of the first order absorption rate constant (k(a) ), apparent clearance (CL/F) and apparent volume of distribution at steady state (V(ss) /F) were 0.15h(-1) , 111 lh(-1) and 2950 l, respectively. Further, using the proposed semi-mechanistic hepatic intrinsic clearance model with Bayesian inference, mean population desipramine hepatic intrinsic clearance was estimated to be 262 lh(-1) with between-subject variability of 84%. d-optimal PK sampling times for intermediate metabolizers were calculated to be approximately 0.25, 24, 75 and 200h. Similar sampling times were found for ultrarapid and extensive metabolizers except that the second d-optimal sample was earlier at 14 and 19h, respectively, compared with 24h for intermediate metabolizers. This difference in sampling times between the three genotypes can be attributed to the different intrinsic clearances and elimination rates. CONCLUSIONS: A two compartment population pharmacokinetic model best described desipramine disposition. The semi-mechanistic population model developed is suitable to describe the pharmacokinetic behaviour of desipramine for the dose routinely used in drug-drug interaction (DDI) studies. Based on this meta-analysis of seven trials, a sample size of 21 subjects in cross-over design is appropriate for assessing CYP2D6 interaction with novel compounds.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors , Desipramine/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Models, Biological , Administration, Oral , Adult , Bayes Theorem , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Female , Genotype , Humans , Male , Middle Aged , Phenotype
20.
J Pharm Sci ; 98(6): 1928-34, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19117050

ABSTRACT

While the number of clinical trials has continued to grow by about 20% in the past six months, no corresponding growth in product approval by the food and drug administration is seen or anticipated in the near future. Late-stage clinical failures due to lack of efficacy or toxicity continues to be a challenge. The optimization of absorption, distribution, metabolism and elimination (ADME) has improved drug candidate selection and reduced early clinical failure. The current challenge is how to avoid late stage clinical failures. Expanded knowledge of drug target distribution, pharmacokinetics and validated biomarkers will allow implementation of appropriate drug delivery and clinical trial designs to reduce drug exposure to off-target organs such as the liver and kidney and could reduce potential untoward effects. In essence, integration of drug delivery and targeting to reduce exposure in off-target tissues in the preclinical and clinical program may hold the key to increasing the odds of success in drug development. In this update, we briefly review data on clinical trials pertinent to drug delivery in the current regulatory environment. It also provides our analysis on the emerging trends in second generation antibody therapeutics in drug delivery and targeting.


Subject(s)
Drug Delivery Systems/methods , Drug Delivery Systems/trends , Clinical Trials as Topic , Humans
SELECTION OF CITATIONS
SEARCH DETAIL