Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Chee M. Ng is active.

Publication


Featured researches published by Chee M. Ng.


Pharmacogenetics and Genomics | 2013

Effects of CYP3A4 and CYP3A5 polymorphisms on tacrolimus pharmacokinetics in Chinese adult renal transplant recipients: a population pharmacokinetic analysis.

Xiao-cong Zuo; Chee M. Ng; Jeffrey S. Barrett; Ai-jing Luo; Bi-kui Zhang; Chen-hui Deng; Lanyan Xi; Ke Cheng; Yingzi Ming; Guoping Yang; Qi Pei; Li-jun Zhu; Hong Yuan; Hai-qiang Liao; Jun-jie Ding; Di Wu; Ya-nan Zhou; Ningning Jing; Zhijun Huang

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.


Pediatric Critical Care Medicine | 2013

Population pharmacokinetics of phenobarbital in infants with neonatal encephalopathy treated with therapeutic hypothermia

Renée A. Shellhaas; Chee M. Ng; Christina H. Dillon; John Barks; Varsha Bhatt-Mehta

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.


European Journal of Pharmaceutical Sciences | 2014

Modeling approach to investigate the effect of neonatal Fc receptor binding affinity and anti-therapeutic antibody on the pharmacokinetic of humanized monoclonal anti-tumor necrosis factor-α IgG antibody in cynomolgus monkey.

Chee M. Ng; Kelly M. Loyet; Suhasini Iyer; Paul J. Fielder; Rong Deng

PURPOSE Several neonatal Fc receptor (FcRn) variants of an anti-tumor necrosis factor (TNF)-α humanized monoclonal IgG antibodies (mAbs) were developed but the effect of their differential FcRn binding affinities on pharmacokinetic (PK) behavior were difficult to be definitively measured in vivo due to formation of anti-therapeutic antibody (ATA). A semi-mechanistic model was developed to investigate the quantitative relationship between the FcRn binding affinity and PK of mAbs in cynomolgus monkey with the presence of ATA. METHODS PK and ATA data from cynomolgus monkeys which received a single intravenous dose of adalimumab, wild-type or two FcRn variant (N434H and N434A) anti-TNF-α mAbs were included in the analysis. Likelihood-based censored data handling method was used to include many PK observations with BQL values for model development. A fully integrated PK-ATA model was developed and used to fit simultaneously to the PK/ATA data. RESULTS AND CONCLUSIONS The PK and ATA time-profiles and effect of FcRn-binding affinity on PK of mAbs were well described by the model and the parameters were estimated with good precision. The model was used successfully to construct quantitative relationships between FcRn binding affinity and PK of anti-TNF-α mAbs in the presence of the ATA-mediated elimination and interferences.


Pharmacotherapy | 2015

Population Pharmacokinetic Model of Sublingual Buprenorphine in Neonatal Abstinence Syndrome

Chee M. Ng; Erin Dombrowsky; Hopi Lin; Michelle E. Erlich; David E. Moody; Jeffrey S. Barrett; Walter K. Kraft

Neonatal abstinence syndrome (NAS)—a clinical entity of infants from in utero exposure to psychoactive xenobiotic and buprenorphine—has been successfully used to treat NAS. However, nothing is known about the pharmacokinetics (PK) of buprenorphine in neonates with NAS. To our knowledge, this is the first study to investigate the population pharmacokinetic of sublingual buprenorphine in neonates with NAS.


Cancer Chemotherapy and Pharmacology | 2011

Mechanism-based pharmacokinetic/pharmacodynamic model for troxacitabine-induced neutropenia in cancer patients

Chee M. Ng; A. Patnaik; M. Beeram; Chia Chi Lin; C. H. Takimoto

PurposesThe objective of this study was to develop a mechanism-based population pharmacokinetic/pharmacodynamic (PK/PD) model in describing troxacitabine-induced neutropenia in patients with cancer.MethodsA total of 727 PK/PD samples from 31 patients with cancer were included in the analysis. A mechanism-based population PD model was developed to describe neutropenia and the final model consisted of (1) a drug-sensitive uncommitted progenitor cell compartment (2) three transit compartments, and (3) a circulating neutrophil compartment with feedback mechanism. The troxacitabine affected the proliferation of sensitive progenitor cells through an inhibitory Emax model. The model parameters were estimated using the MCPEM algorithm that was implemented in a parallel computing platform consisting of a single computer equipped with a quad-core INTEL central processor unit.Results and conclusionsThe mechanism-based PK/PD model developed using parallelized MCPEM method adequately describes the complex relationship between the exposure and absolute neutrophil counts in troxacitabine-treated patients with cancer. The simulation results suggested that the less frequent dosing schedule of troxacitabine used currently in clinical studies was associated with less incidence of neutropenia compared to more frequent dosing schedule.


Aaps Journal | 2016

Mechanism-Based Competitive Binding Model to Investigate the Effect of Neonatal Fc Receptor Binding Affinity on the Pharmacokinetic of Humanized Anti-VEGF Monoclonal IgG1 Antibody in Cynomolgus Monkey

Chee M. Ng; Paul J. Fielder; Jin Jin; Rong Deng

The quantitative relationship between neonatal Fc receptor (FcRn) binding affinity at both acidic and physiological pH and the pharmacokinetics of protein engineered FcRn IgG1 variants has not yet been reported. Our objective was to develop a quantitatively mechanism-based competitive binding model to describe the effects of FcRn binding affinity at acidic and physiological pH on the pharmacokinetics of anti-VEGF IgG1 antibodies when both endogenous and exogenous antibodies are competing for the same FcRn. Pharmacokinetic (PK) and FcRn binding data from five Fc variants of humanized anti-VEGF IgG1 monoclonal antibodies with wide range of FcRn binding affinity were used for the analysis. Sixty-seven anti-VEGF IgG1 antibody-treated animals and 25 control animals with simulated endogenous IgG levels were used to develop the final model. A hybrid iterative two stages and Monte Carlo parametric expectation-maximization method was used to obtain the final model parameters estimates. The final model well described the observed PK data. Quantitative FcRn binding affinity-pharmacokinetics relationships was constructed to provide important biological insights in better understanding of the FcRn binding effect on pharmacokinetics of anti-VEGF IgG1 antibodies in cynomolgus monkeys and served as an important model-based drug discovery platform to guide the design and development of the future generation of anti-VEGF or other therapeutic IgG1 antibodies.


Magnetic Resonance Imaging | 2013

GPU-accelerated nonparametric kinetic analysis of DCE-MRI data from glioblastoma patients treated with bevacizumab

Yu-Han H. Hsu; Gregory Z. Ferl; Chee M. Ng

Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) is often used to examine vascular function in malignant tumors and noninvasively monitor drug efficacy of antivascular therapies in clinical studies. However, complex numerical methods used to derive tumor physiological properties from DCE-MRI images can be time-consuming and computationally challenging. Recent advancement of computing technology in graphics processing unit (GPU) makes it possible to build an energy-efficient and high-power parallel computing platform for solving complex numerical problems. This study develops the first reported fast GPU-based method for nonparametric kinetic analysis of DCE-MRI data using clinical scans of glioblastoma patients treated with bevacizumab (Avastin®). In the method, contrast agent concentration-time profiles in arterial blood and tumor tissue are smoothed using a robust kernel-based regression algorithm in order to remove artifacts due to patient motion and then deconvolved to produce the impulse response function (IRF). The area under the curve (AUC) and mean residence time (MRT) of the IRF are calculated using statistical moment analysis, and two tumor physiological properties that relate to vascular permeability, volume transfer constant between blood plasma and extravascular extracellular space (K(trans)) and fractional interstitial volume (ve) are estimated using the approximations AUC/MRT and AUC. The most significant feature in this method is the use of GPU-computing to analyze data from more than 60,000 voxels in each DCE-MRI image in parallel fashion. All analysis steps have been automated in a single program script that requires only blood and tumor data as the sole input. The GPU-accelerated method produces K(trans) and ve estimates that are comparable to results from previous studies but reduces computational time by more than 80-fold compared to a previously reported central processing unit-based nonparametric method. Furthermore, it is at least several orders of magnitudes faster than standard parametric methods that perform compartmental modeling. This finding indicates that the GPU-based method can significantly shorten the computational times required to assess tumor physiology from DCE-MRI data in preclinical and clinical development of antivascular therapies.


The Journal of Clinical Pharmacology | 2010

Synergy Between Scientific Advancement and Technological Innovation, Illustrated by a Mechanism-Based Model Characterizing Sodium-Glucose Cotransporter-2 Inhibition

Liping Zhang; Chee M. Ng; James F. List; Marc Pfister

Advances in experimental medicine and technological innovation during the past century have brought tremendous progress in modern medicine and generated an ever‐increasing amount of data from bench and bedside. The desire to extend scientific knowledge motivates effective data integration. Technological innovation makes this possible, which in turn accelerates the advancement in science. This mutually beneficial interaction is illustrated by the development of an expanded mechanism‐based model for understanding a novel mechanism, sodium‐glucose cotransporter‐2 SGLT2 inhibition for potential treatment of type 2 diabetes mellitus.


Biopharmaceutics & Drug Disposition | 2016

Incorporation of FcRn-mediated disposition model to describe the population pharmacokinetics of therapeutic monoclonal IgG antibody in clinical patients.

Chee M. Ng

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. Copyright


Pediatric Critical Care Medicine | 2014

Population pharmacokinetics of unbound hydrocortisone in critically ill neonates and infants with vasopressor-resistant hypotension.

Heather Vezina; Chee M. Ng; Delia M. Vazquez; John Barks; Varsha Bhatt-Mehta

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.

Collaboration


Dive into the Chee M. Ng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey S. Barrett

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Barks

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Walter K. Kraft

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Yu-Han H. Hsu

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge