Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Article in English | MEDLINE | ID: mdl-38782726

ABSTRACT

BACKGROUND AND HYPOTHESIS: Volenrelaxin, is a half-life-extended recombinant human relaxin protein developed for improving kidney perfusion and cardiorenal function. This study assessed the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of volenrelaxin following single- and multiple-ascending doses (SAD and MAD) administration. METHODS: In this Phase 1, 4-part, randomized, double-blinded, placebo-controlled SAD and MAD study in healthy participants, SAD participants (n = 56) received an intravenous (IV) or subcutaneous (SC) dose of volenrelaxin or placebo in a dose-ascending manner. MAD participants (n = 77) received volenrelaxin or placebo SC once weekly for 5 weeks. Effective renal plasma flow (ERPF) and measured glomerular filtration rate (mGFR) were determined by para-aminohippurate and iohexol clearance, respectively. RESULTS: Volenrelaxin demonstrated an extended half-life and increased acute and chronic placebo-adjusted ERPF change from baseline by 50% and 44%, respectively (p < 0.0001). Measured GFR was unchanged, while filtration fraction and afferent/efferent renal arteriolar resistances were reduced. Systolic and diastolic blood pressures decreased, and pulse rate increased with increasing volenrelaxin exposures, demonstrating maximal model-derived placebo-adjusted changes (90% confidence interval) of -6.16 (-8.04, -4.28) mmHg, -6.10 (-7.61, -4.58) mmHg, and + 4.39 (3.38, 5.39) bpm, respectively. Adverse events were mild, with no difference in orthostatic hypotension between volenrelaxin and placebo. CONCLUSION: Volenrelaxin was well-tolerated, safe and suitable for weekly SC dosing. Volenrelaxin showed a sustained improvement in kidney perfusion upon repeated dosing, supporting further clinical development in chronic kidney disease and chronic heart failure. Clinical trial registration: NCT04768855.

2.
J Clin Pharmacol ; 64(2): 215-226, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37853524

ABSTRACT

Dulaglutide 3.0 and 4.5 mg weekly doses were approved for additional glycemic control in adult patients with type 2 diabetes inadequately controlled with metformin and 0.75 or 1.5 mg weekly doses of dulaglutide. Effects such as nausea and vomiting are commonly reported with dulaglutide and other glucagon-like peptide-1 receptor agonist therapies. Based on a pharmacokinetic/pharmacodynamic model-informed approach, a stepwise dose-escalation scheme with 4-week intervals between dose increments was suggested to mitigate gastrointestinal events for dulaglutide. These gastrointestinal events are dose dependent and attenuate over time with repeated dosing. A Markov chain Monte Carlo pharmacokinetic/pharmacodynamic joint model was developed using AWARD-11 data (N = 1842) to optimize dulaglutide dose escalation to 3.0 and 4.5 mg to mitigate gastrointestinal events. Model simulations evaluated probabilities of nausea and vomiting events for various dosing scenarios in patients needing higher doses for additional glycemic control. The model indicated that patients may dose escalate from 1.5 to 3.0 mg, then 4.5 mg weekly after at least 4 weeks on each dose. No clinically meaningful differences in nausea or vomiting events were expected when patients escalated to 3.0 or 4.5 mg following initiation at 0.75 or 1.5 mg dulaglutide. Based on the findings of this model, a minimum 4-week duration at each dose before escalation was appropriate to reduce gastrointestinal events of dulaglutide, consistent with observed gastrointestinal events data from the AWARD-11 study and supporting the currently recommended dose-escalation regimen of dulaglutide doses of 3.0 and 4.5 mg for additional glycemic control.


Subject(s)
Diabetes Mellitus, Type 2 , Adult , Humans , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/adverse effects , Blood Glucose , Glycated Hemoglobin , Glucagon-Like Peptides/adverse effects , Immunoglobulin Fc Fragments/adverse effects , Recombinant Fusion Proteins/adverse effects , Vomiting/chemically induced , Vomiting/drug therapy , Nausea/chemically induced , Nausea/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists
4.
Diabetes Obes Metab ; 24(2): 302-311, 2022 02.
Article in English | MEDLINE | ID: mdl-34697882

ABSTRACT

AIM: To evaluate HbA1c and body weight changes when semaglutide 0.5- or 1.0-mg once-weekly (QW) is switched to dulaglutide 3.0- or 4.5-mg QW via exposure-response modelling. METHODS: HbA1c and body weight time-course models were developed and validated with data from the SUSTAIN 1 to 10 trials for semaglutide and the AWARD-11 trial for dulaglutide. Simulations were conducted for HbA1c and body weight over 52 weeks. In the initial 26 weeks, semaglutide was initiated at 0.25-mg and titrated to 0.5- or 1.0-mg QW via 4-weekly stepwise titration, followed by 26 weeks of dulaglutide initiated at 0.75- or 1.5-mg QW and escalated to 3.0- or 4.5-mg QW via 4-weekly stepwise titration. RESULTS: At 26 weeks, model-predicted mean changes from baseline in HbA1c and weight for semaglutide 0.5 mg were up to -1.55% and -3.44 kg, respectively. After switching to dulaglutide 3.0 mg, further reductions were 0.19% and 1.40 kg, respectively, at 52 weeks. Predicted mean HbA1c and weight changes for semaglutide 1.0 mg at 26 weeks were -1.84% and -4.96 kg, respectively; after switching to dulaglutide 4.5 mg, HbA1c was maintained with additional weight reduction of up to 0.57 kg at 52 weeks. Glycaemic control was preserved when switching from semaglutide 1.0 mg to dulaglutide 3.0 mg. CONCLUSION: Switching from semaglutide 0.5 mg to dulaglutide 3.0 or 4.5 mg with dose escalation potentially yields additional HbA1c and weight reductions; switching from semaglutide 1.0 mg to dulaglutide 4.5 mg may enhance weight loss.


Subject(s)
Diabetes Mellitus, Type 2 , Glycemic Control , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptides/adverse effects , Glucagon-Like Peptides/analogs & derivatives , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/adverse effects , Immunoglobulin Fc Fragments/adverse effects , Recombinant Fusion Proteins/adverse effects , Weight Loss
5.
Adv Ther ; 39(1): 488-503, 2022 01.
Article in English | MEDLINE | ID: mdl-34787823

ABSTRACT

INTRODUCTION: This study evaluated the pharmacokinetics, pharmacodynamics, and safety of a single dulaglutide dose in Chinese healthy subjects and of multiple dulaglutide doses in Chinese patients with type 2 diabetes mellitus (T2DM). METHODS: This two-part, double-blind, placebo-controlled study included 16 healthy subjects randomized to receive a single dose of placebo and two of three dulaglutide doses (0.5 mg, 0.75 mg, or 1.5 mg) in three treatment periods, and 42 patients with T2DM randomized to receive placebo or one of the three dulaglutide doses once weekly for 4 weeks. Pharmacokinetics and safety parameters were assessed in all participants, and pharmacodynamics effects were investigated in patients with T2DM. RESULTS: Following a single-dose administration of 0.5 mg, 0.75 mg, or 1.5 mg dulaglutide in healthy subjects, geometric mean maximum concentrations (Cmax) were 29.4, 44.2, and 81.5 ng/mL, respectively. Following weekly administration in patients with T2DM for 4 weeks, Cmax were 26.3, 41.4, and 70.2 ng/mL, respectively, with accumulation ratios of 1.33-1.39. Geometric mean for half-life of 4-5 days and median time to Cmax (tmax) of approximately 48 h were observed in both study populations. Dose-proportional increases in drug exposure were observed after both single and multiple dosing. Significant reductions in baseline-corrected fasting glucose and hemoglobin A1c (HbA1c) were observed in patients with T2DM who received dulaglutide 0.75 mg and 1.5 mg. Dulaglutide was well tolerated, with the majority of adverse events being gastrointestinal disorders of mild severity. CONCLUSIONS: Pharmacokinetics, pharmacodynamics, and safety profiles of dulaglutide demonstrated in the present study support a once-weekly dosing regimen in Chinese patients with T2DM. TRIAL REGISTRATION: NCT01667900 (ClinicalTrials.gov).


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptides , Immunoglobulin Fc Fragments , Recombinant Fusion Proteins , Blood Glucose , China , Diabetes Mellitus, Type 2/drug therapy , Dose-Response Relationship, Drug , Double-Blind Method , Glucagon-Like Peptides/adverse effects , Glucagon-Like Peptides/analogs & derivatives , Healthy Volunteers , Humans , Hypoglycemic Agents/adverse effects , Immunoglobulin Fc Fragments/adverse effects , Recombinant Fusion Proteins/adverse effects
6.
Clin Pharmacol Drug Dev ; 10(12): 1452-1459, 2021 12.
Article in English | MEDLINE | ID: mdl-34410042

ABSTRACT

Insulin glargine (IGlar) and LY2963016 (LY IGlar) are long-acting insulin analogs with identical primary amino acid sequences. We conducted a randomized, open-label, 2-treatment, 2-period, crossover study in healthy Chinese subjects to evaluate the relative bioavailability of LY IGlar to IGlar and pharmacokinetic (PK) and pharmacodynamic (PD) characteristics of LY IGlar. Subjects (n = 58) were randomized to receive single subcutaneous doses (0.5 U/kg) of LY IGlar and IGlar with a ≥7-day washout period between study treatments. Serum was collected before and up to 24 hours after dosing to assess PK characteristics. PD characteristics were assessed by euglycemic clamp up to 24 hours after dosing. Linear mixed-effects models were used to fit the log-transformed primary PK (maximum observed concentration and area under the concentration-time curve from time 0 to 24 hours) and PD parameters (maximum glucose infusion rate and total amount of glucose infused during clamp period). The geometric least squares means ratios (90% confidence interval) of LY IGlar to IGlar for maximum observed concentration and area under the concentration-time curve from time 0 to 24 hours were 0.961 (0.887-1.04) and 0.941 (0.872-1.01), respectively. The geometric least squares means ratios (90% confidence interval) of LY IGlar to IGlar were 0.91 (0.85-0.98) for maximum glucose infusion rate and 0.89 (0.82-0.97) for total amount of glucose infused during clamp period. LY IGlar demonstrated similarity to IGlar in PK and PD characteristics following single-dose (0.5 U/kg) administration in healthy Chinese subjects.


Subject(s)
Glucose , Hypoglycemic Agents , Biological Availability , Blood Glucose/metabolism , China , Cross-Over Studies , Glucose Clamp Technique , Humans , Insulin Glargine
7.
CPT Pharmacometrics Syst Pharmacol ; 8(9): 664-675, 2019 09.
Article in English | MEDLINE | ID: mdl-31250974

ABSTRACT

The drug-drug interaction profile of atorvastatin confirms that disposition is determined by cytochrome P450 (CYP) 3A4 and organic anion transporting polypeptides (OATPs). Drugs that affect gastric emptying, including dulaglutide, also affect atorvastatin pharmacokinetics (PK). Atorvastatin is a carboxylic acid that exists in equilibrium with a lactone form in vivo. The purpose of this work was to assess gastric acid-mediated lactone equilibration of atorvastatin and incorporate this into a physiologically-based PK (PBPK) model to describe atorvastatin acid, lactone, and their major metabolites. In vitro acid-to-lactone conversion was assessed in simulated gastric fluid and included in the model. The PBPK model was verified with in vivo data including CYP3A4 and OATP inhibition studies. Altering the gastric acid-lactone equilibrium reproduced the change in atorvastatin PK observed with dulaglutide. The model emphasizes the need to include gastric acid-lactone conversion and all major atorvastatin-related species for the prediction of atorvastatin PK.


Subject(s)
Atorvastatin/pharmacokinetics , Gastroparesis/complications , Glucagon-Like Peptides/analogs & derivatives , Lactones/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Atorvastatin/administration & dosage , Cells, Cultured , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Drug Interactions , Gastric Acid/metabolism , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacokinetics , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunoglobulin Fc Fragments/administration & dosage , Models, Biological , Organic Anion Transporters , Recombinant Fusion Proteins/administration & dosage
8.
J Clin Pharmacol ; 59(2): 258-270, 2019 02.
Article in English | MEDLINE | ID: mdl-30339268

ABSTRACT

Insulin replacement therapy is a fundamental treatment for glycemic control for managing diabetes. The engineering of insulin analogues has focused on providing formulations with action profiles that mimic as closely as possible the pattern of physiological insulin secretion that normally occurs in healthy individuals without diabetes. Hence, it may be helpful to practitioners to visualize insulin concentration profiles and associated glucose action profiles. Expanding on a previous analysis that established a pharmacokinetic (PK) model to describe typical profiles of insulin concentration over time following subcutaneous administration of various insulin formulations, the goal of the current analysis was to link the PK model to an integrated glucose-insulin (IGI) systems pharmacology model. After the pharmacokinetic-pharmacodynamic (PK-PD) model was qualified by comparing model predictions with clinical observations, it was used to project insulin (PK) and glucose (PD) profiles of common insulin regimens and dosing scenarios. The application of the PK-PD model to clinical scenarios was further explored by incorporating the impact of several hypothetical factors together, such as changing the timing or frequency of administration in a multiple-dosing regimen over the course of a day, administration of more than 1 insulin formulation, or insulin dosing adjusted for carbohydrates in meals. Visualizations of insulin and glucose profiles for commonly prescribed regimens could be rapidly generated by implementing the linked subcutaneous insulin PK-IGI model using the R statistical program (version 3.4.4) and a contemporary web-based interface, which could enhance clinical education on glycemic control with insulin therapy.


Subject(s)
Blood Glucose/drug effects , Insulin/pharmacology , Insulin/pharmacokinetics , Adult , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Drug Administration Schedule , Humans , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Middle Aged , Models, Biological
9.
CPT Pharmacometrics Syst Pharmacol ; 6(12): 804-813, 2017 12.
Article in English | MEDLINE | ID: mdl-28891251

ABSTRACT

Baricitinib is an oral inhibitor of Janus kinases (JAKs), selective for JAK1 and 2. It demonstrated dose-dependent efficacy in patients with moderate-to-severe rheumatoid arthritis (RA) in a phase IIb study up to 24 weeks. Population pharmacokinetic/pharmacodynamic (PopPK/PD) models were developed to characterize concentration-time profiles and dose/exposure-response (D/E-R) relationships for the key efficacy (proportion of patients achieving American College of Rheumatology 20%, 50%, or 70% response rate) and safety endpoints (incidence of anemia) for the phase IIb study. The modeling suggested that 4 mg q.d. was likely to offer the optimum risk/benefit balance, whereas 2 mg q.d. had the potential for adequate efficacy. In addition, at the same total daily dose, a twice-daily regimen is not expected to provide an advantage over q.d. dosing for the efficacy or safety endpoints. The model-based simulations formed the rationale for key aspects of dosing, such as dose levels and dosing frequency for phase III development.


Subject(s)
Antirheumatic Agents/administration & dosage , Antirheumatic Agents/pharmacokinetics , Arthritis, Rheumatoid/drug therapy , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Models, Biological , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Administration, Oral , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/enzymology , Azetidines/adverse effects , Clinical Trials, Phase III as Topic , Computer Simulation , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Predictive Value of Tests , Purines , Pyrazoles , Sulfonamides/adverse effects , Treatment Outcome
10.
J Clin Pharmacol ; 57(9): 1126-1137, 2017 09.
Article in English | MEDLINE | ID: mdl-28394405

ABSTRACT

Insulin pharmacokinetics following subcutaneous administration were modeled, simulated, and displayed through an interactive and user-friendly interface to illustrate the time course of administered insulins frequently prescribed, providing a simple tool for clinicians through a straightforward visualization of insulin regimens. Pharmacokinetic data of insulin formulations with different onset and duration of action from several clinical studies, including insulin glargine, regular insulin, neutral protamine Hagedorn (NPH), insulin lispro, and premixed preparations of NPH with regular insulin (Mix 70/30), and insulin lispro protamine suspension with insulin lispro (Mix 50/50, Mix 75/25), were used to develop a predictive population pharmacokinetic model of insulins with consideration of factors such as insulin formulation, weight-based dosing, body-weight effect on volume of distribution, and administration time relative to meals, on the insulin time-action profile. The model-predicted insulin profile of each insulin was validated and confirmed to be comparable to observed data via an external validation method. Model-based simulations of clinically relevant insulin-dosing scenarios to cater to specific initial patient and prescribing conditions were then implemented with differential equations using the R statistical program (version 3.2.2). The R package Shiny was subsequently applied to build a web browser interface to execute and visualize the model simulation outputs. The application of insulin pharmacokinetic modeling enabled informative visualization of insulin time-action profiles and provided an efficient and intuitive educational tool to quickly convey and interactively explore many insulin time-action profiles to ease the understanding of insulin formulations in clinical practice.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Insulins/pharmacokinetics , Models, Biological , Adolescent , Adult , Dose-Response Relationship, Drug , Female , Humans , Hypoglycemic Agents/administration & dosage , Injections, Subcutaneous , Insulins/administration & dosage , Male , Middle Aged , Young Adult
12.
Int J Clin Pharmacol Ther ; 53(3): 199-205, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25546161

ABSTRACT

OBJECTIVE: Changes in the cortisol diurnal rhythm have been found in neuropsychiatric diseases, including anxiety and depression. The aim of this study was to investigate the reliability and reproducibility of cortisol diurnal rhythm in healthy males, through determination of the inter- and intrasubject variability, to facilitate evaluation of this biomarker for drug target engagement. MATERIALS AND METHODS: This open-label, 2-period study design evaluated serum cortisol release over a 24-hour period in 18 healthy males. A cosinor model was used to model the cortisol diurnal rhythm, and the inter- and intrasubject coefficients of variation (CVs) were calculated across the 2 periods. RESULTS: Three significant cortisol concentration peaks were observed at ~7 AM, 1 PM, and 7 PM. The intersubject CVs (%) for the amplitude, acrophase, and midline estimating statistic of rhythm (MESOR) were 18.2, 19.3, and 16.8, respectively. The intrasubject CVs (%) were 11.2, 7.6, and 6.9, respectively. The inter- and intrasubject CVs (%) for the lunch-induced 1 PM peak and the >dinner-induced 7 PM peak were 22.1, 17.3, 44.7, and 22.1 respectively. CONCLUSIONS: Assessment of serum cortisol diurnal rhythm suggests that the amplitude, acrophase, and MESOR, but not the meal-induced peaks, have the potential to be a reliable biomarker in drug development targeting the hypothalamic-pituitary-adrenocortical (HPA) axis.


Subject(s)
Circadian Rhythm , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Adult , Biomarkers/blood , Fasting/blood , Healthy Volunteers , Humans , Hydrocortisone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Male , Middle Aged , Models, Biological , Pituitary-Adrenal System/metabolism , Postprandial Period , Reproducibility of Results , Sex Factors , Time Factors , Young Adult
13.
Int J Clin Pharmacol Ther ; 52(9): 717-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24887333

ABSTRACT

OBJECTIVES: A modified insulin tolerance test (ITT) can be used to simulate a physiological stress state through the induction of controlled hypoglycemia in healthy volunteers. This allows for evaluation of hypothalamic-pituitary-adrenocortical axis response to stress via a surge in cortical release. However, a consequence of severe, prolonged hypoglycemia is QT interval prolongation. The aim of this analysis was to confirm that blood glucose lowering to 60 mg/dL (previously identified as adequate for inducing stress) has low risk of inducing clinically significant QT prolongation. MATERIALS AND METHODS: Continuous ECG monitoring was conducted as a planned sub study of an open-label, 2-period study involving 18 healthy male subjects. The QTcF response to hypoglycemia was measured over 2 identical periods, ~ 7 days apart. RESULTS: An indirect- response model adequately described the pharmacological relationship between blood glucose and QTcF intervals over the time-course of the ITT. The model correctly identified the steep glucose-QT relationship as an on-off response with a large Hill coefficient of 59 and the threshold glucose, EC50, as ~ 57 mg/dL with narrow between-subject variability of 10%. Simulated QTcF profiles over the course of an ITT did not demonstrate any QTcF interval changes of clinical concern, defined as QTcF observation > 500 ms, if hypoglycemia did not reach below 60 mg/dL. The statistical prediction that the chance of a mean QTcF observation > 500 ms was < 0.0001. CONCLUSIONS: Results support that an ITT maintained at or above 60 mg/dL is unlikely to cause QT prolongation in healthy volunteers and does not warrant continuous ECG monitoring in this group of subjects.


Subject(s)
Arrhythmias, Cardiac/etiology , Blood Glucose/metabolism , Computer Simulation , Diagnostic Techniques, Endocrine , Hypoglycemia/complications , Hypoglycemic Agents , Insulin , Models, Biological , Adult , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/physiopathology , Biomarkers/blood , Electrocardiography , Healthy Volunteers , Humans , Hypoglycemia/blood , Hypoglycemia/diagnosis , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Infusions, Intravenous , Insulin/administration & dosage , Insulin/adverse effects , Male , Predictive Value of Tests , Risk Assessment , Risk Factors , Time Factors
14.
J Clin Pharmacol ; 54(10): 1117-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24752880

ABSTRACT

Ixekizumab (LY2439821), a humanized immunoglobulin G subclass 4 (IgG4) monoclonal antibody that selectively binds and neutralizes interleukin (IL) 17A has demonstrated efficacy in the treatment of psoriasis. A population pharmacokinetics-pharmacodynamics model was developed using NONMEM 7.2 to describe the temporal relationship between ixekizumab concentrations and absolute Psoriasis Area and Severity Index (PASI) scores from a phase 2 dose-finding study in chronic plaque psoriasis. The objective was to inform dose-selection for further development. The primary endpoint, PASI 75 (75% or greater improvement from baseline PASI score) was then derived from each individual's absolute PASI score. The population pharmacokinetics of ixekizumab was characterized by a two-compartment model, while the exposure-response relationship was characterized using an indirect response model that described the pharmacological effects of ixekizumab and placebo in the form of inhibition of the formation of psoriatic skin lesions. PASI 75 responder status at the Week 12 primary endpoint was found to be a significant covariate on the concentration producing half maximal effect (EC50 ). While the results suggested patient may have different levels of sensitivity to ixekizumab, it is possible that nonresponder patients assigned to lower doses of ixekizumab may potentially become responders to ixekizumab if given doses that yield adequate exposures.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Models, Biological , Psoriasis/drug therapy , Adult , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Chronic Disease , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Nonlinear Dynamics , Psoriasis/pathology , Severity of Illness Index , Treatment Outcome
15.
J Diabetes ; 5(2): 127-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23332026

ABSTRACT

BACKGROUND: This open-label, single-period study assessed the pharmacokinetics, safety, tolerability, and pharmacodynamics of exenatide once weekly (q.w.), following single and multiple weekly subcutaneous (s.c.) injections in native Chinese patients with type 2 diabetes (T2D). METHODS: Patients (n = 25; mean [±SD] age 51.3 ± 8.2 years; body mass index 25.6 ± 2.4 kg/m(2) ; HbA1c 7.4 ± 1.2%; duration of diabetes 3.1 ± 3.1 years) previously treated with diet modification and exercise alone or incombination with stable metformin doses were enrolled in the study. Twenty-five patients received weekly doses of 2 mg, s.c., exenatide q.w. for 10 weeks, followed by 10 weeks observation. Pharmacokinetic parameters of exenatide, fasting plasma glucose (FPG), HbA1c, and body weight were summarized using descriptive statistics. RESULTS: Steady state plasma exenatide concentrations (299 pg/mL) were attained within 8 weeks. Exenatide q.w. was generally well tolerated, and the majority of adverse events reported were mild in severity. The most frequent study drug-related adverse events were diarrhea and vomiting. Decreases were observed from baseline to 10 weeks in FPG (~3.0 mmol/L), HbA1c (~1.0%), and body weight (~3.8 kg). CONCLUSIONS: This is the first clinical trial of exenatide q.w. in native Chinese patients with T2D. The results suggest that exenatide q.w. has a pharmacokinetic profile in this patient population similar to that observed in other ethnic and racial populations, and appears to be safe and generally well tolerated, with the potential to improve glycemic control and decrease body weight without increasing the risk of hypoglycemia.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Peptides/pharmacokinetics , Venoms/pharmacokinetics , Adult , Asian People , Blood Glucose/metabolism , Body Mass Index , Body Weight , Diabetes Mellitus, Type 2/drug therapy , Drug Tolerance , Exenatide , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Injections, Subcutaneous , Male , Middle Aged , Peptides/therapeutic use , Venoms/therapeutic use
16.
Regul Pept ; 179(1-3): 77-83, 2012 Nov 10.
Article in English | MEDLINE | ID: mdl-22960288

ABSTRACT

Exenatide is a glucagon-like peptide-1 receptor agonist for the treatment of type 2 diabetes and has been shown to lower blood glucose through multiple mechanisms, including glucose-dependent insulin secretion, suppression of postprandial glucagon release and slowing of gastric emptying. The effects of exenatide on biliary motility are unknown. This study evaluated the effect of a single dose of exenatide on cholecystokinin (CCK)-induced gallbladder emptying. Healthy subjects participated in this randomized, 2-period, double-blind crossover study. Fasting subjects received a single subcutaneous injection of exenatide (10 µg) or placebo 60 min before CCK infusion. Gallbladder volume and ejection fraction (EF) were assessed by ultrasonography before, during, and after CCK infusion (0.003 µg/kg infused over 50 min at 2 mL/min). The diameters of the main pancreatic duct and common bile duct were measured sonographically at the same time points before, during, and following CCK infusion. Administration of exenatide did not affect pre-CCK infusion gallbladder volume or EF compared to placebo. During the CCK-infusion, the mean minimum gallbladder volume was similar for exenatide (13.68 mL) and placebo (11.05 mL) (least squares mean [LSM] difference of 2.62 mL; 95% confidence interval [CI], -0.53, 5.78), but the mean maximum EF was lower for exenatide (28.79%) versus placebo (46.13%) (LSM difference of -17.34%; 95% CI, -30.54, -4.13). Exenatide had no clinically significant effects on pancreatic or bile duct diameters. In conclusion, exenatide reduced CCK-induced gallbladder emptying compared with placebo in fasting healthy subjects.


Subject(s)
Cholecystokinin/pharmacology , Fasting , Gallbladder Emptying/drug effects , Peptides/pharmacology , Venoms/pharmacology , Adolescent , Adult , Aged , Bile Ducts/anatomy & histology , Bile Ducts/diagnostic imaging , Blood Glucose/analysis , Cholecystokinin/administration & dosage , Confidence Intervals , Cross-Over Studies , Double-Blind Method , Exenatide , Female , Gallbladder/diagnostic imaging , Gallbladder/drug effects , Gallbladder/metabolism , Glucagon-Like Peptide 1/agonists , Humans , Least-Squares Analysis , Male , Middle Aged , Organ Size/drug effects , Pancreatic Ducts/anatomy & histology , Pancreatic Ducts/diagnostic imaging , Peptides/administration & dosage , Ultrasonography , Venoms/administration & dosage , Young Adult
17.
Lung Cancer ; 63(1): 121-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18538445

ABSTRACT

OBJECTIVE: Pharmacogenetics suggests variants of genes involved in gemcitabine pharmacology could be useful markers for predicting inter-ethnic and inter-patient outcomes from treatment with the agent. Here, we have characterized the distribution of variants of genes involved in gemcitabine pharmacology in ethnic Asian populations and their association with non-small cell lung cancer (NSCLC) patient outcome. METHODS: All genes involved in gemcitabine transport, metabolism and activity were screened for suitable variants for analysis using publications and public databases. By pyrosequencing, the frequency of qualifying variants was characterized from germline DNA of 94 healthy Asian donors and 53 NSCLC patients receiving gemcitabine-based chemotherapy. RESULTS: Significant differences in genotype distribution between Caucasians and Asians were seen at 10/25 (45%) variant loci. In NSCLC patients, CDA+435 C>T variants were associated with response (p=0.026) and time to progression (p=0.016) and SLC28A1+1561 G>A variants were associated with neutropenia (p=0.030) and thrombocytopenia nadir (p=0.037). CONCLUSIONS: Many genotypes in gemcitabine pharmacology vary in their frequency between Caucasians and Asians. CDA+435, and SLC28A1+1561 are worthy of further investigation as potential indicators of patient outcome after gemcitabine treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Deoxycytidine/analogs & derivatives , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Pharmacogenetics/methods , Adolescent , Adult , Aged , Antimetabolites, Antineoplastic/pharmacology , Asian People , Carcinoma, Non-Small-Cell Lung/ethnology , Cytidine Deaminase/genetics , Deoxycytidine/pharmacology , Female , Genotype , Humans , Lung Neoplasms/ethnology , Male , Middle Aged , Retrospective Studies , Singapore , Treatment Outcome , Gemcitabine
18.
Clin Cancer Res ; 14(13): 4213-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18594002

ABSTRACT

PURPOSE: This tumor response pharmacodynamic model aims to describe primary lesion shrinkage in non-small cell lung cancer over time and determine if concentration-based exposure metrics for gemcitabine or that of its metabolites, 2',2'-difluorodeoxyuridine or gemcitabine triphosphate, are better than gemcitabine dose for prediction of individual response. EXPERIMENTAL DESIGN: Gemcitabine was given thrice weekly on days 1 and 8 in combination with carboplatin, which was given only on day 1 of every cycle. Gemcitabine amount in the body and area under the concentration-time curves of plasma gemcitabine, 2',2'-difluorodeoxyuridine, and intracellular gemcitabine triphosphate in white cells were compared to determine which best describes tumor shrinkage over time. Tumor growth kinetics were described using a Gompertz-like model. RESULTS: The apparent half-life for the effect of gemcitabine was 7.67 weeks. The tumor turnover time constant was 21.8 week.cm. Baseline tumor size and gemcitabine amount in the body to attain 50% of tumor shrinkage were estimated to be 6.66 cm and 10,600 mg. There was no evidence of relapse during treatment. CONCLUSIONS: Concentration-based exposure metrics for gemcitabine and its metabolites were no better than gemcitabine amount in predicting tumor shrinkage in primary lung cancer lesions. Gemcitabine dose-based models did marginally better than treatment-based models that ignored doses of drug administered to patients. Modeling tumor shrinkage in primary lesions can be used to quantify individual sensitivity and response to antitumor effects of anticancer drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxycytidine/analogs & derivatives , Lung Neoplasms/drug therapy , Aged , Area Under Curve , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Kinetics , Male , Middle Aged , Neoplasms/metabolism , Time Factors , Gemcitabine
19.
Cancer Chemother Pharmacol ; 63(1): 55-64, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18305939

ABSTRACT

AIM: This study aims to determine if intracellular formation of gemcitabine triphosphate (dFdCTP), an active metabolite of gemcitabine, is saturable at doses used for treatment of Asian patients with lung cancer. METHODS: From a phase II trial, plasma concentrations of gemcitabine, its inactive metabolite 2'-2'-difluorodeoxyuridine (dFdU), and mononuclear cell concentrations of gemcitabine-triphosphate were measured in 56 and 33 patients, respectively. The pharmacokinetics of gemcitabine and metabolites were modeled using nonlinear mixed effects modeling (NONMEM). A reduced dataset of ten randomly selected patients was employed to compare first-order and saturable formation of dFdCTP from gemcitabine. RESULTS: The median population clearance estimate for dFdCTP formation with the full dataset was 70.2 L/h/70 kg/1.7 m. Modeling Michaelis-Menten formation of dFdCTP on a reduced dataset estimated K(m) to be 3.6 times higher than the maximum gemcitabine concentration (72.2 microM) measured in this study. CONCLUSIONS: The results showed that first-order and nonsaturable clearance described intracellular dFdCTP formation at clinically applied doses of gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cytidine Triphosphate/analogs & derivatives , Deoxycytidine/analogs & derivatives , Lung Neoplasms/metabolism , Prodrugs/pharmacokinetics , Adipose Tissue , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biotransformation , Body Weight , Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Clinical Trials, Phase II as Topic , Creatinine/blood , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/pharmacology , Deoxycytidine/pharmacokinetics , Female , Humans , Lung Neoplasms/drug therapy , Male , Middle Aged , Models, Biological , Prospective Studies , Randomized Controlled Trials as Topic , Gemcitabine
20.
Cancer Chemother Pharmacol ; 62(2): 243-51, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17909805

ABSTRACT

PURPOSE: The aims were to determine the maximum tolerable dose (MTD) of docetaxel with CYP3A inhibition by ketoconazole, and to correlate the pharmacokinetics of docetaxel with midazolam phenotyping of CYP3A activity. METHODS: Forty-one patients with refractory metastatic cancers were treated with an escalating dose of intravenous docetaxel once in every 3 week of 10 mg/m(2), concurrently with oral ketoconazole 200 mg twice daily for 3 days starting 2 days before the administration of docetaxel. Midazolam phenotyping test with ketoconazole modulation was performed before the first cycle of docetaxel. Docetaxel and midazolam pharmacokinetics were compared to our previous study of docetaxel treatment without ketoconazole modulation. RESULTS: Neutropenia was the dose-limiting toxicity. The maximum tolerated dose was 70 mg with mean AUC at 70 mg similar to 75 mg/m(2) of docetaxel without ketoconazole. The plasma clearances of docetaxel and midazolam were reduced by 1.7- and 6-fold, respectively. The variability of midazolam AUC was reduced from 157 to 67%, but variability of docetaxel clearance was not reduced by CYP3A inhibition. Docetaxel clearance correlated with renal function and maximum concentration of ketoconazole, but not midazolam clearance or other variables of hepatic function. CONCLUSION: Fixed dosing was found to be feasible, without increased variability of clearance or neutrophil toxicity compared to BSA-based dosing. With ketoconazole modulation, docetaxel clearance correlated with renal function but not CYP3A phenotype.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/therapeutic use , Ketoconazole/therapeutic use , Neoplasms/drug therapy , Taxoids/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A , Docetaxel , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Ketoconazole/administration & dosage , Ketoconazole/adverse effects , Ketoconazole/pharmacokinetics , Male , Maximum Tolerated Dose , Midazolam/administration & dosage , Midazolam/pharmacokinetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/pathology , Neutropenia/chemically induced , Taxoids/administration & dosage , Taxoids/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...