Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Article in English | MEDLINE | ID: mdl-39153154

ABSTRACT

Pharmacokinetics and pharmacodynamics of many biologics are influenced by their complex binding to biological receptors. Biologics consist of diverse groups of molecules with different binding kinetics to its receptors including IgG with simple one-to-one drug receptor bindings, bispecific antibody (BsAb) that binds to two different receptors, and antibodies that can bind to six or more identical receptors. As the binding process is typically much faster than elimination (or internalization) and distribution processes, quasi-equilibrium (QE) binding models are commonly used to describe drug-receptor binding kinetics of biologics. However, no general QE modeling framework is available to describe complex binding kinetics for diverse classes of biologics. In this paper, we describe novel approaches of using differential algebraic equations (DAE) to solve three QE multivalent drug-receptor binding (QEMB) models. The first example describes the binding kinetics of three-body equilibria of BsAb that binds to 2 different receptors for trimer formation. The second example models an engineered IgG variant (Multabody) that can bind to 24 identical target receptors. The third example describes an IgG with modified neonatal Fc receptor (FcRn) binding affinity that competes for the same FcRn receptor as endogenous IgG. The model parameter estimates were obtained by fitting the model to all data simultaneously. The models allowed us to study potential roles of cooperative binding on bell-shaped drug exposure-response relationships of BsAb, and concentration-depended distribution of different drug-receptor complexes for Multabody. This DAE-based QEMB model platform can serve as an important tool to better understand complex binding kinetics of diverse classes of biologics.

2.
Article in English | MEDLINE | ID: mdl-38639818

ABSTRACT

The paper extended the TMDD model to drugs with more than two (N > 2) identical binding sites (N-to-one TMDD). The quasi-steady-state (N-to-one QSS), quasi-equilibrium (N-to-one QE), irreversible binding (N-to-one IB), and Michaelis-Menten (N-to-one MM) approximations of the model were derived. To illustrate properties of new equations and approximations, N = 4 case was investigated numerically. Using simulations, the N-to-one QSS approximation was compared with the full N-to-one TMDD model. As expected, and similarly to the standard TMDD for monoclonal antibodies (mAb), N-to-one QSS predictions were nearly identical to N-to-one TMDD predictions, except for times of fast changes following initiation of dosing, when equilibrium has not yet been reached. Predictions for mAbs with soluble targets (slow elimination of the complex) were simulated from the full 4-to-one TMDD model and were fitted to the 4-to-one TMDD model and to its QSS approximation. It was demonstrated that the 4-to-one QSS model provided nearly identical description of not only the observed (simulated) total drug and total target concentrations, but also unobserved concentrations of the free drug, free target, and drug-target complexes. For mAb with a membrane-bound target, the 4-to-one MM approximation adequately described the data. The 4-to-one QSS approximation converged 8 times faster than the full 4-to-one TMDD.

3.
Invest New Drugs ; 37(6): 1218-1230, 2019 12.
Article in English | MEDLINE | ID: mdl-30820810

ABSTRACT

Background AR-67 is a novel camptothecin analogue at early stages of drug development. The phase 1 clinical trial in cancer patients with solid tumors was completed and a population pharmacokinetic model (POP PK) was developed to facilitate further development of this investigational agent. Methods Pharmacokinetic data collected in the phase 1 clinical trial were utilized for the development of a population POP PK by implementing the non-linear mixed effects approach. Patient characteristics at study entry were evaluated as covariates in the model. Subjects (N = 26) were treated at nine dosage levels (1.2-12.4Ā mg/m2/day) on a daily Ɨ 5 schedule. Hematological toxicity data were modeled against exposure metrics. Results A two-compartment POP PK model best described the disposition of AR-67 by fitting a total of 328 PK observations from 25 subjects. Following covariate model selection, age remained as a significant covariate on central volume. The final model provided a good fit for the concentration versus time data and PK parameters were estimated with good precision. Clearance, inter-compartmental clearance, central volume and peripheral volume were estimated to be 32.2Ā L/h, 28.6Ā L/h, 6.83Ā L and 25.0Ā L, respectively. Finally, exposure-pharmacodynamic analysis using Emax models showed that plasma drug concentration versus time profiles are better predictors of AR-67-related hematologic toxicity were better predictors of leukopenia and thrombocytopenia, as compared to total dose. Conclusions A POP PK model was developed to characterize AR-67 pharmacokinetics and identified age as a significant covariate. Exposure PK metrics Cmax and AUC were shown to predict hematological toxicity. Further efforts to identify clinically relevant determinants of AR-67 disposition and effects in a larger patient population are warranted.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Models, Biological , Neoplasms/metabolism , Organosilicon Compounds/pharmacokinetics , Adult , Aged , Antineoplastic Agents, Phytogenic/blood , Camptothecin/blood , Camptothecin/pharmacokinetics , Female , Humans , Male , Middle Aged , Neoplasms/blood , Organosilicon Compounds/blood
4.
Biomed Chromatogr ; 33(7): e4527, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30830964

ABSTRACT

A high-performance liquid chromatography-tandem mass spectrometry method was developed and validated for the simultaneous quantification of morphine, morphine's major metabolites morphine-3-glucuronide and morphine-6-glucuronide, and clonidine, to support the pharmacokinetic analysis of an ongoing double-blinded randomized clinical trial that compares the use of morphine and clonidine in infants diagnosed with neonatal abstinence syndrome. Plasma samples were processed by solid-phase extraction and separated on an Inertsil ODS-3 (4 Āµm) column using an 0.1% formic acid in water-0.1% formic acid in methanol gradient. Detection of the analytes was conducted in the positive multiple reaction monitoring mode. The range of quantitation was 1-1000 ng/mL for morphine, morphine-3-glucuronide and morphine-6-glucuronide, and 0.25-100 ng/mL for clonidine. Intra-day and inter-day accuracy and precision were ≤15% for all analytes across the quantitation range. Extraction recovery rates were ≥94% for morphine, ≥90% for M3G, ≥87% for M6G and ≥ 79% for clonidine. Matrix effect ranged from 85-94% for clonidine to 101-106% for M3G. The method fulfilled all predetermined acceptance criteria and required only 100 ĀµL of starting plasma volume. Furthermore, it was successfully applied to 30 clinical trial plasma samples.


Subject(s)
Chromatography, High Pressure Liquid/methods , Clonidine/blood , Morphine Derivatives/blood , Tandem Mass Spectrometry/methods , Clonidine/chemistry , Drug Stability , Humans , Infant, Newborn , Limit of Detection , Linear Models , Morphine Derivatives/chemistry , Neonatal Abstinence Syndrome/blood , Neonatal Abstinence Syndrome/diagnosis , Randomized Controlled Trials as Topic , Reproducibility of Results
5.
Br J Clin Pharmacol ; 84(3): 520-532, 2018 03.
Article in English | MEDLINE | ID: mdl-29077992

ABSTRACT

AIMS: Congenital hyperinsulinism (HI) is the most common cause of persistent hypoglycaemia in infants and children. Exendin-(9-39), an inverse glucagon-like peptide 1 (GLP-1) agonist, is a novel therapeutic agent for HI that has demonstrated glucose-raising effect. We report the first population pharmacokinetic (PopPK) model of the exendin-(9-39) in patients with HI and propose the optimal dosing regimen for future clinical trials in neonates with HI. METHODS: A total of 182 pharmacokinetic (PK) observations from 26 subjects in three clinical studies were included for constructing the PopPK model using first order conditional estimation (FOCE) with interaction method in nonlinear mixed-effects modelling (NONMEM). Exposure metrics (area under the curve [AUC] and maximum plasma concentration [Cmax ]) at no observed adverse effect levels (NOAELs) in rats and dogs were determined in toxicology studies. RESULTS: Observed concentration-time profiles of exendin-(9-39) were described by a linear two-compartmental PK model. Following allometric scaling of PK parameters, age and creatinine clearance did not significantly affect clearance. The calculated clearance and elimination half-life for adult subjects with median weight of 69Ā kg were 11.8Ā lĀ h-1 and 1.81Ā h, respectively. The maximum recommended starting dose determined from modelling and simulation based on the AUC0-last at the NOAEL and predicted AUC0-inf using the PopPK model was 27Ā mgĀ kg-1 Ā day-1 intravenously. CONCLUSIONS: This is the first study to investigate the PopPK of exendin-(9-39) in humans. The final PopPK model was successfully used with preclinical toxicology findings to propose the optimal dosing regimen of exendin-(9-39) for clinical studies in neonates with HI, allowing for a more targeted dosing approach to achieve desired glycaemic response.


Subject(s)
Congenital Hyperinsulinism/drug therapy , Models, Biological , Peptide Fragments/administration & dosage , Adolescent , Adult , Age Factors , Animals , Area Under Curve , Child , Child, Preschool , Cross-Over Studies , Dogs , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infant , Infant, Newborn , Male , Middle Aged , No-Observed-Adverse-Effect Level , Nonlinear Dynamics , Peptide Fragments/adverse effects , Peptide Fragments/pharmacokinetics , Pilot Projects , Rats , Rats, Sprague-Dawley , Young Adult
6.
Biopharm Drug Dispos ; 37(2): 107-19, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26581439

ABSTRACT

PURPOSE: The two-compartment linear model used to describe the population pharmacokinetics (PK) of many therapeutic monoclonal antibodies (TMAbs) offered little biological insight to antibody disposition in humans. The purpose of this study is to develop a semi-mechanistic FcRn-mediated IgG disposition model to describe the population PK of TMAbs in clinical patients. METHODS: A standard two-compartment linear PK model from a previously published population PK model of pertuzumab was used to simulate intensive PK data of 100 subjects for model development. Two different semi-mechanistic FcRn-mediated IgG disposition models were developed and First Order Conditional Estimation (FOCE) with the interaction method in NONMEM was used to obtain the final model estimates. The performances of these models were then compared with the two-compartment linear PK model used to simulate the data for model development. RESULTS: A semi-mechanistic FcRn-mediated IgG disposition model consisting of a peripheral tissue compartment and FcRn-containing endosomes in the central compartment best describes the simulated pertuzumab population PK data. This developed semi-mechanistic population PK model had the same number of model parameters, produced very similar concentration-time profiles but provided additional biological insight to the FcRn-mediated IgG disposition in human subjects compared with the standard linear two-compartment linear PK model. CONCLUSION: This first reported semi-mechanistic model may serve as an important model framework for developing future population PK models of TMAbs in clinical patients.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Models, Biological , Receptors, Fc/immunology , Humans
7.
Pharm Res ; 32(6): 1907-19, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25467958

ABSTRACT

PURPOSE: A semi-mechanistic multiple-analyte population pharmacokinetics (PK) model was developed to describe the complex relationship between the different analytes of monomethyl auristatin E (MMAE) containing antibody-drug conjugates (ADCs) and to provide insight regarding the major pathways of conjugate elimination and unconjugated MMAE release in vivo. METHODS: For an anti-CD79b-MMAE ADC the PK of total antibody (Tab), conjugate (evaluated as antibody conjugated MMAE or acMMAE), and unconjugated MMAE were quantified in cynomolgus monkeys for single (0.3, 1, or 3Ā mg/kg), and multiple doses (3 or 5Ā mg/kg, every-three-weeks for 4 doses). The PK data of MMAE in cynomolgus monkeys, after intravenous administration of MMAE at single doses (0.03 or 0.063Ā mg/kg), was included in the analysis. A semi-mechanistic model was developed and parameter estimates were obtained by simultaneously fitting the model to all PK data using a hybrid ITS-MCPEM method. RESULTS: The final model well described the observed Tab, acMMAE and unconjugated MMAE concentration-time profiles. Analysis suggested that conjugate is lost via both proteolytic degradation and deconjugation, while unconjugated MMAE in systemic circulation appears to be mainly released via proteolytic degradation of the conjugate. CONCLUSIONS: Our model improves the understanding of ADC catabolism, which may provide useful insights when designing future ADCs.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Models, Biological , Oligopeptides/pharmacokinetics , Administration, Intravenous , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/blood , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Biotransformation , Macaca fascicularis , Oligopeptides/administration & dosage , Oligopeptides/blood , Proteolysis
8.
Pediatr Crit Care Med ; 15(2): 162-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24141658

ABSTRACT

OBJECTIVE: To evaluate the effectiveness of methadone for the treatment of neonatal abstinence syndrome when used according to a preexisting clinical pathway. DESIGN: This is a 3-year retrospective study conducted at a single institution. In this study, neonates who received methadone for the treatment of neonatal abstinence syndrome according to a predefined clinical treatment pathway were evaluated for treatment success: defined as adherence to the methadone regimen with no residual signs of withdrawal. Data were collected for methadone dosages, Lipsitz scores, length of methadone treatment, total length of hospital stay, and relevant clinical data. SETTING: Level III neonatal ICU. PATIENTS: Newborn infants with in utero exposure to substances of abuse. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Sixty patients were included. The mean gestational age and birth weight were 37.07 Ā± 3.05 weeks and 2.77 Ā± 0.6 kg. All 60 patients exhibited neonatal abstinence syndrome within first 72 hours of life. Fifty-seven of 60 patients (95%) initiated methadone treatment according to protocol. There was deviation from the protocol at 48 and 72 hours of treatment with approximately 59% and 13% of the patients still on methadone at more than the prescribed amount to control neonatal abstinence syndrome. The mean Ā± SD total methadone exposure was 1.99 Ā± 1.63 mg/kg, length of treatment 11.66 Ā± 9.02 days, and total hospital length of stay 22.43 Ā± 29.3 days, suggesting significant variability in response. No significant correlation was found between birth weight or gestational age and length of treatment. CONCLUSION: Clinical pathway for treating neonatal abstinence syndrome was closely followed at the initial diagnosis. The doses of methadone used in the first 24-48 hours of this study were insufficient for adequate symptom control. Despite a formal treatment protocol, there was substantial variability in total methadone exposure, length of treatment, and length of stay, suggesting other contributory factors for the observed variability.


Subject(s)
Analgesics, Opioid/administration & dosage , Methadone/administration & dosage , Neonatal Abstinence Syndrome/drug therapy , Pregnancy Complications/drug therapy , Prenatal Exposure Delayed Effects/drug therapy , Analgesics, Opioid/therapeutic use , Clinical Protocols , Female , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Length of Stay , Male , Methadone/therapeutic use , Pregnancy , Retrospective Studies , Treatment Outcome
9.
Pediatr Crit Care Med ; 15(6): 546-53, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24797719

ABSTRACT

OBJECTIVES: To determine the population pharmacokinetics of unbound hydrocortisone in critically ill neonates and infants receiving IV hydrocortisone for treatment of vasopressor-resistant hypotension and to identify patient-specific sources of pharmacokinetic variability. DESIGN: Prospective observational cohort study. SETTING: Level 3 neonatal ICU. PATIENTS: Sixty-two critically ill neonates and infants receiving IV hydrocortisone as part of standard of care for the treatment of vasopressor-resistant hypotension: median gestational age 28 weeks (range, 23-41), median weight 1.2 kg (range, 0.5-4.4), and 29 females. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Unbound baseline cortisol and postdose hydrocortisone concentrations measured from blood samples being drawn for routine laboratory tests. A one-compartment model best described the data. Allometric weight and postmenstrual age were significant covariates on unbound hydrocortisone clearance and volume of distribution. Final population estimates for clearance, volume of distribution, and baseline cortisol concentration were 20.2 L/hr, 244 L, and 1.37 ng/mL, respectively. Using the median weight and postmenstrual age of our subjects (i.e., 1.2 kg and 28 wk) in the final model, the typical unbound hydrocortisone clearance and volume of distribution were 1.0 L/hr and 4.2 L, respectively. The typical half-life for unbound hydrocortisone was 2.9 hours. A sharp and continuous increase in unbound hydrocortisone clearance was observed at 35 weeks postmenstrual age. CONCLUSIONS: We report the first pharmacokinetic data for unbound hydrocortisone, the pharmacologically active moiety, in critically ill neonates and infants with vasopressor-resistant hypotension. Unbound hydrocortisone clearance increased with body weight and was faster in children with an older postmenstrual age. Unbound hydrocortisone clearance increased sharply at 35 weeks postmenstrual age and continued to mature thereafter. This study lays the groundwork for evaluating unbound hydrocortisone exposure-response relationships and drawing definitive conclusions about the dosing of IV hydrocortisone in critically ill neonates and infants with vasopressor-resistant hypotension.


Subject(s)
Drug Resistance , Hormones/pharmacokinetics , Hormones/therapeutic use , Hydrocortisone/pharmacokinetics , Hydrocortisone/therapeutic use , Hypotension/drug therapy , Blood Pressure/drug effects , Body Weight , Critical Illness , Female , Gestational Age , Half-Life , Hormones/blood , Humans , Hydrocortisone/blood , Hypotension/blood , Infant , Infant, Newborn , Male , Prospective Studies , Vasopressins/pharmacology
10.
Mol Pharmacol ; 84(2): 182-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23680637

ABSTRACT

Organic cation transporters (OCTs) are members of the solute carrier 22 family of transporter proteins that are involved in absorption, distribution, and excretion of organic cations. OCT3 is localized in the apical (AP) membrane of enterocytes, but the literature is ambiguous about OCT1 (mOct1) localization, with some evidence suggesting a basolateral (BL) localization in human and mouse enterocytes. This is contrary to our preliminary findings showing AP localization of OCT1 in Caco-2 cell monolayers, an established model of human intestinal epithelium. Therefore, this study aims at determining the localization of OCT1 (mOct1) in Caco-2 cells, and human and mouse enterocytes. Functional studies using OCT1-specific substrate pentamidine showed transporter-mediated AP but not BL uptake in Caco-2 cells and human and mouse intestinal tissues. OCT1 inhibition decreased AP uptake of pentamidine by Ć¢ĀˆĀ¼50% in all three systems with no effect on BL uptake. A short hairpin RNA-mediated OCT1 knockdown in Caco-2 cells decreased AP uptake of pentamidine by Ć¢ĀˆĀ¼50% but did not alter BL uptake. Immunostaining and confocal microscopy in all three systems confirmed AP localization of OCT1 (mOct1). Our studies unequivocally show AP membrane localization of OCT1 (mOct1) in Caco-2 cells and human and mouse intestine. These results are highly significant as they will require reinterpretation of previous drug disposition and drug-drug interaction studies where conclusions were drawn assuming BL localization of OCT1 in enterocytes. Most importantly, these results will require revision of the regulatory guidance for industry in the United States and elsewhere because it has stated that OCT1 is basolaterally localized in enterocytes.


Subject(s)
Enterocytes/metabolism , Intestinal Mucosa/metabolism , Organic Cation Transporter 1/metabolism , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Protein Transport
11.
Pharmacogenet Genomics ; 23(5): 251-61, 2013 May.
Article in English | MEDLINE | ID: mdl-23459029

ABSTRACT

OBJECTIVE: Tacrolimus is used clinically for the long-term treatment of antirejection of transplanted organs in liver and kidney transplant recipients, although dose optimization is poorly managed. The aim of this study was to examine the association between tacrolimus pharmacokinetic variability and CYP3A4 and CYP3A5 genotypes by a population pharmacokinetic analysis based on routine drug monitoring data in adult renal transplant recipients. MATERIALS AND METHODS: Trough tacrolimus concentrations were obtained from 161 adult kidney transplant recipients after transplantation. The population pharmacokinetic analysis was carried out using the nonlinear mixed-effect modeling software NONMEM version 7.2. The CYP3A4*1G and CYP3A5*3 genetic polymorphisms from the patients studied were determined by direct sequencing using a validated automated genetic analyzer. RESULTS: A one-compartment model with first-order absorption and elimination adequately described the pharmacokinetics of tacrolimus. Covariates including CYP3A5*3 and CYP3A4*1G alleles and hematocrit were retained in the final model. The apparent clearance of tacrolimus was about two-fold higher in kidney transplant patients with higher enzymatic activity of CYP3A5*1 and CYP3A4*1G (with the CYP3A5*1/*1 or *1/*3 and CYP3A4*1/*1G or CYP3A4*1G/*1G) compared with those with lower enzymatic activity (CYP3A5*3/*3 and CYP3A4*1/*1). CONCLUSION: This is the first study to extensively determine the effect of CYP3A4*1G and CYP3A5*3 genetic polymorphisms and hematocrit value on tacrolimus pharmacokinetics in Chinese renal transplant recipients. The findings suggest that CYP3A5*3 and CYP3A4*1G polymorphisms and hematocrit are determinant factors in the apparent clearance of tacrolimus. The initial dose design is mainly based on CYP3A5 and CYP3A4 genotypes as well as hematocrit. This result may also be useful for maintenance tacrolimus dose optimization and may help to avoid fluctuating tacrolimus levels and improve the efficacy and tolerability of tacrolimus in kidney transplant recipients.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Graft Rejection/genetics , Kidney Transplantation , Tacrolimus/administration & dosage , Adolescent , Adult , Aged , Alleles , China , Female , Genotype , Graft Rejection/drug therapy , Hematocrit , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/genetics , Middle Aged , Polymorphism, Genetic , Tacrolimus/pharmacokinetics
12.
Pediatr Crit Care Med ; 14(2): 194-202, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23254984

ABSTRACT

OBJECTIVE: Phenobarbital is the first-line treatment for neonatal seizures. Many neonates with hypoxic ischemic encephalopathy are treated with therapeutic hypothermia, and about 40% have clinical seizures. Little is known about the pharmacokinetics of phenobarbital in infants with hypoxic ischemic encephalopathy who undergo therapeutic hypothermia. The objective of this study was to determine the effect of therapeutic hypothermia on phenobarbital pharmacokinetics, taking into account maturational changes. SETTING: Level 3 neonatal ICU. PATIENTS: Infants with hypoxic ischemic encephalopathy and suspected seizures, all treated with phenobarbital. Some of these infants also received treatment with therapeutic hypothermia. INTERVENTIONS: None. DESIGN: A retrospective cohort study of 39 infants with hypoxic ischemic encephalopathy treated with phenobarbital (20 were treated with therapeutic hypothermia and 19 were not). MEASUREMENTS AND MAIN RESULTS: Data on phenobarbital plasma concentrations were collected in 39 subjects with hypoxic ischemic encephalopathy with or without therapeutic hypothermia. Using nonlinear mixed-effects modeling, population pharmacokinetics of phenobarbital were developed with a total of 164 plasma concentrations. A one-compartment model best described the pharmacokinetics. The clearance of phenobarbital was linearly related to body weight and matured with increasing age with a maturation half-life of 22.1 days. Therapeutic hypothermia did not influence the pharmacokinetic parameters of phenobarbital. CONCLUSIONS: Therapeutic hypothermia does not influence the clearance of phenobarbital after accounting for weight and age. Standard phenobarbital dosing is appropriate for the initial treatment of seizures in neonates with hypoxic ischemic encephalopathy treated with therapeutic hypothermia.


Subject(s)
Anticonvulsants/pharmacokinetics , Hypothermia, Induced , Hypoxia-Ischemia, Brain/therapy , Phenobarbital/pharmacokinetics , Age Factors , Anticonvulsants/therapeutic use , Body Temperature , Body Weight , Female , Humans , Hypoxia-Ischemia, Brain/complications , Infant, Newborn , Male , Phenobarbital/therapeutic use , Retrospective Studies , Seizures/complications , Seizures/drug therapy
13.
Yao Xue Xue Bao ; 47(7): 941-6, 2012 Jul.
Article in Zh | MEDLINE | ID: mdl-22993862

ABSTRACT

Reasonable sampling scheme is the important basis for establishing reliable population pharmacokinetic model. It is an effective method for estimation of population pharmacokinetic parameters with sparse data to perform population pharmacokinetic analysis using the nonlinear mixed-effects models. We designed the sampling scheme for amlodipine based on D-optimal sampling strategy and Bayesian estimation method. First, optimized sample scenarios were designed using WinPOPT software according to the aim, dosage regimen and visit schedule of the clinical study protocol, and the amlodipine population model reported by Rohatagi et al. Second, we created a NONMEM-formatted dataset (n = 400) for each sample scenario via Monte Carlo simulation. Third, the estimation of amlodipine pharmacokinetic parameters (clearance (CL/F), volume (V/F) and Ka) was based on the simulation results. All modeling and simulation exercises were conducted with NONMEM version 7.2. Finally, the accuracy and precision of the estimated parameters were evaluated using the mean prediction error (MPE) and the mean absolute error (MAPE), respectively. Among the 6 schemes, schemes 6 and 3 have good accuracy and precision. MPE is 0.1% for scheme 6 and -0.6% for scheme 3, respectively. MAPE is 0.7% for both schemes. There is no significant difference in MPE and MAPE of volume among them. Therefore, we select scheme 3 as the final sample scenario because it has good accuracy and precision and less sample points. This research aims to provide scientific and effective sampling scheme for population pharmacokinetic (PK) study of amlodipine in patients with renal impairment and hypertension, provide a scientific method for an optimum design in clinical population PK/PD (pharmacodynamics) research.


Subject(s)
Amlodipine/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Calcium Channel Blockers/pharmacokinetics , Hypertension/metabolism , Adult , Age Factors , Alanine Transaminase/blood , Amlodipine/pharmacology , Antihypertensive Agents/pharmacology , Bayes Theorem , Body Weight , Calcium Channel Blockers/pharmacology , Humans , Metabolic Clearance Rate , Middle Aged , Models, Biological , Monte Carlo Method , Nonlinear Dynamics , Renal Insufficiency/metabolism , Software
14.
Clin Pharmacol Ther ; 110(3): 803-807, 2021 09.
Article in English | MEDLINE | ID: mdl-33884615

ABSTRACT

One of the challenges in translational medicine is to select first-in-human doses of investigational drugs based on findings in preclinical studies. Despite substantial progress in the optimization of recombinant adeno-associated virus (AAV) vectors of in vivo gene therapy for treating various diseases, there remain significant limitations to the use of preclinical data to guide dose selection in clinical trials. Here we introduce a novel concept of gene efficiency factor (GEF) to describe the efficiency of the gene transfer system and describe and apply the concept of GEF in AAV-mediated in vivo gene transfer systems. We explore the utility of allometric scaling to translate GEF across species using AAV-mediated in vivo factor IX (FIX) gene therapy for hemophilia B and to demonstrate the use of GEF in predicting efficacious AAV vector doses in humans. We show for the first time that an allometric relationship exists for GEF of AAV-mediated in vivo gene therapy. Furthermore, we demonstrate the feasibility of using the allometric relationship of GEF to select efficacious first-in-human doses of virus-mediated invivo gene therapy. Based on our findings, allometry of GEF can be used to translate biological efficiency from animal studies to clinical studies and provide a rational basis of setting first-in-human doses for new virus-mediated invivo gene therapy products.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Animals , Factor IX/genetics , Gene Transfer Techniques , Hemophilia B/genetics , Humans
15.
AAPS J ; 23(2): 37, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33660056

ABSTRACT

One important objective of population pharmacokinetic (PPK) analyses is to identify and quantify relationships between covariates and model parameters such as clearance and volume. To improve upon existing covariate model development methods including stepwise procedures and Wald's approximation method (WAM), this paper introduces an innovative method named the hybrid first-order conditional estimation (FOCE)/Monte-Carlo parametric expectation maximization (MCPEM)-based Wald's approximation method with backward elimination (BE), or H-WAM-BE. Compared with WAM, this new method uses MCPEM to obtain full covariance matrix after running FOCE to obtain full model parameter estimates, followed by BE to select the final covariate model. Two groups of datasets (simulation datasets and rituximab datasets) were used to compare the performance of H-WAM-BE with two other methods, likelihood ratio test (LRT)-based stepwise covariate method (SCM) and H-WAM with full subset approach (H-WAM-F) in NONMEM. Different scenarios with different sample sizes and sampling schemes were used for simulating datasets. The nominal model was used as the reference to evaluate the three methods for their ability to accurately identify parameter-covariate relationships. The methods were compared using the number of true and false positive covariates identified, number of times that they identified the reference model, computation times, and predictive performance. Best-performing H-WAM-BE methods (M2 and M4) showed comparable results with LRT-based SCM. H-WAM-BE required shorter or comparable computation times than LRT-based SCM and H-WAM-F regardless of the model structure, sample size, or sampling design used in this study.


Subject(s)
Biological Variation, Population , Models, Biological , Rituximab/pharmacokinetics , Clinical Trials, Phase II as Topic , Computer Simulation , Datasets as Topic , Humans , Likelihood Functions , Monte Carlo Method , Rituximab/administration & dosage
16.
Clin Transl Sci ; 14(4): 1231-1249, 2021 07.
Article in English | MEDLINE | ID: mdl-33650314

ABSTRACT

In this paper, we review the management of neonatal opioid withdrawal syndrome (NOWS) and clinical pharmacology of primary treatment agents in NOWS, including morphine, methadone, buprenorphine, clonidine, and phenobarbital. Pharmacologic treatment strategies in NOWS have been mostly empirical, and heterogeneity in dosing regimens adds to the difficulty of extrapolating study results to broader patient populations. As population pharmacokinetics (PKs) of pharmacologic agents in NOWS become more well-defined and knowledge of patient-specific factors affecting treatment outcomes continue to accumulate, PK/pharmacodynamic modeling and simulation will be powerful tools to aid the design of optimal dosing regimens at the patient level. Although there is an increasing number of clinical trials on the comparative efficacy of treatment agents in NOWS, here, we also draw attention to the importance of optimizing the dosing regimen, which can be arguably equally important at identifying the optimal treatment agent.


Subject(s)
Analgesics, Opioid/adverse effects , Narcotic Antagonists/administration & dosage , Neonatal Abstinence Syndrome/drug therapy , Opiate Substitution Treatment/methods , Biological Variation, Population , Dose-Response Relationship, Drug , Humans , Infant, Newborn , Narcotic Antagonists/pharmacokinetics , Neonatal Abstinence Syndrome/etiology , Treatment Outcome
17.
PLoS One ; 15(3): e0230571, 2020.
Article in English | MEDLINE | ID: mdl-32208461

ABSTRACT

Androgen deprivation therapy (ADT) is a widely used treatment for patients with hormone-sensitive prostate cancer (PCa). However, duration of treatment response varies, and most patients eventually experience disease progression despite treatment. Leuprorelin is a luteinizing hormone-releasing hormone (LHRH) agonist, a commonly used form of ADT. Prostate-specific antigen (PSA) is a biomarker for monitoring disease progression and predicting treatment response and survival in PCa. However, time-dependent profile of tumor regression and growth in patients with hormone-sensitive PCa on ADT has never been fully characterized. In this analysis, nationwide medical claims database provided by Humana from 2007 to 2011 was used to construct a population-based disease progression model for patients with hormone-sensitive PCa on leuprorelin. Data were analyzed by nonlinear mixed effects modeling utilizing Monte Carlo Parametric Expectation Maximization (MCPEM) method in NONMEM. Covariate selection was performed using a modified Wald's approximation method with backward elimination (WAM-BE) proposed by our group. 1113 PSA observations from 264 subjects with malignant PCa were used for model development. PSA kinetics were well described by the final covariate model. Model parameters were well estimated, but large between-patient variability was observed. Hemoglobin significantly affected proportion of drug-resistant cells in the original tumor, while baseline PSA and antiandrogen use significantly affected treatment effect on drug-sensitive PCa cells (Ds). Population estimate of Ds was 3.78 x 10-2 day-1. Population estimates of growth rates for drug-sensitive (Gs) and drug-resistant PCa cells (GR) were 1.96 x 10-3 and 6.54 x 10-4 day-1, corresponding to a PSA doubling time of 354 and 1060 days, respectively. Proportion of the original PCa cells inherently resistant to treatment was estimated to be 1.94%. Application of population-based disease progression model to clinical data allowed characterization of tumor resistant patterns and growth/regression rates that enhances our understanding of how PCa responds to ADT.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Leuprolide/therapeutic use , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Biomarkers/blood , Databases, Factual , Disease Progression , Drug Resistance, Neoplasm , Humans , Male , Middle Aged , Monte Carlo Method , Prostate-Specific Antigen/blood , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Survival Rate
18.
Circ Genom Precis Med ; 13(4): e002602, 2020 08.
Article in English | MEDLINE | ID: mdl-32510984

ABSTRACT

BACKGROUND: Warfarin is an effective treatment for thromboembolic disease but has a narrow therapeutic index; optimal anticoagulation dosage can differ tremendously among individuals. We aimed to evaluate whether genotype-guided warfarin dosing is superior to routine clinical dosing for the outcomes of interest in Chinese patients. METHODS: We conducted a multicenter, randomized, single-blind, parallel-controlled trial from September 2014 to April 2017 in 15 hospitals in China. Eligible patients were ≥18 years of age, with atrial fibrillation or deep vein thrombosis without previous treatment of warfarin or a bleeding disorder. Nine follow-up visits were performed during the 12-week study period. The primary outcome measure was the percentage of time in the therapeutic range of the international normalized ratio during the first 12 weeks after starting warfarin therapy. RESULTS: A total of 660 participants were enrolled and randomly assigned to a genotype-guided dosing group or a control group under standard dosing. The genotype-guided dosing group had a significantly higher percentage of time in the therapeutic range than the control group (58.8% versus 53.2% [95% CI of group difference, 1.1-10.2]; P=0.01). The genotype-guided dosing group also achieved the target international normalized ratio sooner than the control group. In subgroup analyses, warfarin normal sensitivity group had an even higher percentage of time in the therapeutic range during the first 12 weeks compared with the control group (60.8% versus 48.9% [95% CI, 1.1-24.4]). The incidence of adverse events was low in both groups. CONCLUSIONS: The outcomes of genotype-guided warfarin dosing were superior to those of clinical standard dosing. These findings raise the prospect of precision warfarin treatment in China. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02211326.


Subject(s)
Anticoagulants/therapeutic use , Asian People/genetics , Atrial Fibrillation/drug therapy , Venous Thrombosis/drug therapy , Warfarin/therapeutic use , Aged , Anticoagulants/adverse effects , Atrial Fibrillation/genetics , China , Cytochrome P-450 CYP2C9/genetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Genotype , Hemorrhage/etiology , Humans , International Normalized Ratio , Male , Middle Aged , Single-Blind Method , Treatment Outcome , Venous Thrombosis/genetics , Vitamin K Epoxide Reductases/genetics , Warfarin/adverse effects
20.
Clin Cancer Res ; 13(16): 4832-9, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699862

ABSTRACT

PURPOSE: To characterize the pharmacokinetics and pharmacodynamics of oxaliplatin in cancer patients with impaired renal function. EXPERIMENTAL DESIGN: Thirty-four patients were stratified by 24-h urinary creatinine clearance (CrCL) into four renal dysfunction groups: group A (control, CrCL, >or=60 mL/min), B (mild, CrCL, 40-59 mL/min), C (moderate, CrCL, 20-39 mL/min), and D (severe, CrCL, <20 mL/min). Patients were treated with 60 to 130 mg/m2 oxaliplatin infused over 2 h every 3 weeks. Pharmacokinetic monitoring of platinum in plasma, plasma ultrafiltrates, and urine was done during cycles 1 and 2. RESULTS: Plasma ultrafiltrate platinum clearance strongly correlated with CrCL (r2 = 0.712). Platinum elimination from plasma was triphasic, and maximal platinum concentrations (Cmax) were consistent across all renal impairment groups. However, only the beta-half-life was significantly prolonged by renal impairment, with values of 14.0 +/- 4.3, 20.3 +/- 17.7, 29.2 +/- 29.6, and 68.1 h in groups A, B, C, and D, respectively (P = 0.002). At a dose level of 130 mg/m2, the area under the concentration time curve increased in with the degree of renal impairment, with values of 16.4 +/- 5.03, 39.7 +/- 11.5, and 44.6 +/- 14.6 mug.h/mL, in groups A, B, and C, respectively. However, there was no increase in pharmacodynamic drug-related toxicities. Estimated CrCL using the Cockcroft-Gault method approximated the measured 24-h urinary CrCL (mean prediction error, -5.0 mL/min). CONCLUSIONS: Oxaliplatin pharmacokinetics are altered in patients with renal impairment, but a corresponding increase in oxaliplatin-related toxicities is not observed.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Kidney Diseases/metabolism , Neoplasms/drug therapy , Organoplatinum Compounds/pharmacokinetics , Adult , Aged , Aged, 80 and over , Creatinine/metabolism , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , Platinum/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL