Network


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

Hotspot


Dive into the research topics where Chie Emoto is active.

Publication


Featured researches published by Chie Emoto.


CPT: Pharmacometrics & Systems Pharmacology | 2013

Development of a Physiologically‐Based Pharmacokinetic Model for Sirolimus: Predicting Bioavailability Based on Intestinal CYP3A Content

Chie Emoto; Tsuyoshi Fukuda; Samuel H. Cox; Uwe Christians; Alexander A. Vinks

Sirolimus is an inhibitor of mammalian target of rapamycin (mTOR) and is increasingly being used in transplantation and cancer therapies. Sirolimus has low oral bioavailability and exhibits large pharmacokinetic variability. The underlying mechanisms for this variability have not been explored to a large extent. Sirolimus metabolism was characterized by in vitro intrinsic clearance estimation. Pathway contribution ranked from CYP3A4 > CYP3A5 > CYP2C8. With the well stirred and Qgut models sirolimus bioavailability was predicted at 15%. Interindividual differences in bioavailability could be attributed to variable intestinal CYP3A expression. The physiologically‐based pharmacokinetics (PBPK) model developed in Simcyp predicted a high distribution of sirolimus into adipose tissue and another elimination pathway in addition to CYP‐mediated metabolism. PBPK model predictive performance was acceptable with Cmax and area under the curve (AUC) estimates within 20% of observed data in a dose escalation study. The model also showed potential to assess the impact of hepatic impairment and drug–drug interaction (DDI) on sirolimus pharmacokinetics.


British Journal of Clinical Pharmacology | 2015

The impact of CYP3A5*3 polymorphism on sirolimus pharmacokinetics: insights from predictions with a physiologically-based pharmacokinetic model

Chie Emoto; Tsuyoshi Fukuda; Raja Venkatasubramanian; Alexander A. Vinks

AIMS Sirolimus is an mTOR inhibitor metabolized by CYP3A4 and CYP3A5. Reported effects of CYP3A5 polymorphisms on sirolimus pharmacokinetics (PK) have shown unexplained discrepancies across studies. We quantitatively assessed the effect of CYP3A5*3 status on sirolimus PK by in vitro assessment and simulation using a physiologically-based PK (PBPK) model. In addition, we explored designs for an adequately powered pharmacogenetic association study. METHOD In vitro metabolism studies were conducted to confirm individual CYP contribution to sirolimus metabolism. PK profiles were simulated in CYP3A5 expressers and non-expressers with a PBPK model. The pre-dose concentration predictions were used as the outcome parameter to estimate the required sample size for a pharmacogenetic association study. RESULTS Sirolimus metabolism was inhibited by over 90% by ketoconazole, a CYP3A specific inhibitor. The PBPK model developed based on CL(int) of recombinant CYP3A4, CYP3A5 and CYP2C8 predicted a small CYP3A5*3 effect on simulated sirolimus PK profiles. A subsequent power analysis based on these findings indicated that at least 80 subjects in an enrichment design, 40 CYP3A5 expressers and 40 non-expressers, would be required to detect a significant difference in the predicted trough concentrations at 1 month of therapy (P < 0.05, 80% power). CONCLUSIONS This study suggests that CYP3A5 contribution to sirolimus metabolism is much smaller than that of CYP3A4. Observed discrepancies across studies could be explained as the result of inadequate sample size. PBPK model simulations allowed mechanism-based evaluation of the effects of CYP3A5 genotype on sirolimus PK and provided preliminary data for the design of a future prospective study.


Biology of Blood and Marrow Transplantation | 2017

Pretransplant Absolute Lymphocyte Counts Impact the Pharmacokinetics of Alemtuzumab

Rebecca Marsh; Tsuyoshi Fukuda; Chie Emoto; Lisa Neumeier; Pooja Khandelwal; Sharat Chandra; Ashley Teusink-Cross; Alexander A. Vinks; Parinda A. Mehta

Alemtuzumab is frequently used as part of reduced-intensity conditioning (RIC) regimens for allogeneic hematopoietic cell transplantation (HCT) in pediatric patients with nonmalignant diseases. We previously suggested an optimal day 0 targeted range of alemtuzumab, but there are no pediatric data regarding the pharmacokinetics (PK) of subcutaneous alemtuzumab to guide precision dosing trials. The goal of this study was to prospectively characterize alemtuzumab PK and to explore absolute lymphocyte count (ALC) as a predictor of interindividual variability. We prospectively enrolled 23 patients who received an alemtuzumab, fludarabine, and melphalan RIC regimen. Seventeen patients completed study and received 1 mg/kg alemtuzumab divided over 5 days subcutaneously, starting on day -14. The median age was 7 years (range, .5 to 18). Blood sampling for PK measurements and descriptive PK analyses were performed. The median maximum alemtuzumab concentration was 2.39 µg/mL (interquartile range, 1.98 to 2.92). The median terminal half-life was 5.2 days (interquartile range, 2.7 to 7.8). The median concentration at day 0 was 1.27 µg/mL (interquartile range, .35 to 1.51). Importantly, day 0 alemtuzumab levels and area under the curve negatively correlated with predose ALC and ALC area-time, respectively. In conclusion, we reported the PK of subcutaneous alemtuzumab given to pediatric allogeneic HCT patients and observed that almost all patients have persistence of lytic levels of alemtuzumab beyond day 0, at levels in excess of that needed to reduce the risk of acute graft-versus-host disease. Additionally, levels correlate with pretransplant ALC. These results will allow the development of population PK models for precision dosing trials.


Drug Metabolism and Disposition | 2016

Developmental Changes in Hepatic Organic Cation Transporter OCT1 Protein Expression from Neonates to Children.

David Hahn; Chie Emoto; Alexander A. Vinks; Tsuyoshi Fukuda

Organic cation transporter 1 (OCT1) plays an important role in the disposition of clinically important drugs, and the capacity of OCT1 activity is presumed to be proportional to the protein expression level in organ tissues. Knowledge of OCT1 protein expression in children, especially neonates and small infants, is currently very limited. Here, we report on the characterization of OCT1 protein expression in neonatal, infant, and pediatric liver samples performed using immunoblot analysis. OCT1 protein expression was detected in liver samples from neonates as early as postnatal days 1 and 2. This youngest group showed significantly lower OCT1 expression normalized by glyceraldehyde-6-phosphate dehydrogenase (values given as means ± S.D. in arbitrary units; 0.03 ± 0.02, n = 7) compared with samples from patients aged 3 to 4 weeks (0.08 ± 0.03, n = 5, P < 0.01), 3 to 6 months (0.23 ± 0.15, n = 7, P < 0.01), 11 months to 1 year (0.42 ± 0.32, n = 6, P < 0.01), and 8 to 12 years (1.00 ± 0.44, n = 7, P < 0.01). These data demonstrate an age-dependent increase in OCT1 expression from birth up to 8 to 12 years of age, and the findings of this study contribute to the understanding of OCT1 functional capacity and its effect upon the disposition of OCT1 substrates in neonates and small infants.


Biology of Blood and Marrow Transplantation | 2016

A Prospective Study of Alemtuzumab as a Second-Line Agent for Steroid-Refractory Acute Graft-versus-Host Disease in Pediatric and Young Adult Allogeneic Hematopoietic Stem Cell Transplantation

Pooja Khandelwal; Chie Emoto; Tsuyoshi Fukuda; Alexander A. Vinks; Lisa Neumeier; Christopher E. Dandoy; Javier El-Bietar; Sharat Chandra; Stella M. Davies; J.J. Bleesing; Michael B. Jordan; Parinda A. Mehta; Sonata Jodele; Michael Grimley; Ashish Kumar; Kasiani C. Myers; Rebecca A. Marsh

We describe a single-center prospective study of alemtuzumab as a second-line agent for steroid-refractory (SR) acute graft-versus-host disease (aGVHD) in pediatric and young adult allogeneic hematopoietic stem cell transplant recipients. Alemtuzumab was administered for grades II to IV aGVHD if patients did not improve within 5 days or worsened within 48 hours after corticosteroids. Interim analyses of alemtuzumab levels and response were performed after every 5 patients enrolled, resulting in 3 dosing cohorts, as follows: (1) .2 mg/kg alemtuzumab subcutaneously on days 1 to 5 (maximum of 31 mg over 5 days) and .2 mg/kg/dose (not exceeding 10 mg/dose) on days 15, 22, and 29; (2) .2 mg/kg alemtuzumab subcutaneously on days 1 to 5 (maximum of 43 mg over 5 days) and .2 mg/kg/dose on day 7, 10, 15, 22, and 29; and (3) .2 mg/kg subcutaneously on days 1 to 5 and .2 mg/kg/dose on day 7, 10, 15, and 22. Alemtuzumab levels were assessed before starting alemtuzumab and at days 1, 3, 6, 10, and 14 and weekly until day 99, where day 1 was the day of first alemtuzumab dose. Fifteen patients (median age, 10 years; range, 1.4 to 27) received alemtuzumab for grades II (6%), III (74%), and IV (20%) SR-aGVHD. The overall response rate was 67%, with complete response (CR) in 40%, partial response (PR) in 27%, and no response in 33%. The median day 6 alemtuzumab level was 2.79 µg/mL (interquartile range, 1.34 to 4.89) in patients with CR compared with .62 µg/mL (interquartile range, .25 to 1.45) in patients with PR + no response (P < .05). Ninety percent (n = 9) of patients with a CR or PR reduced corticosteroid doses within 8 weeks from first alemtuzumab dose. Side effects included fever (26%) and transient thrombocytopenia (53%). Asymptomatic viremias occurred in all patients but invasive viral disease occurred in 2 patients. One patient developed Epstein-Barr virus-post-transplantation lymphoproliferative disorder. Eighty percent (n = 12) of patients were alive at 6 months, of whom 53% (n = 8) were free of GVHD whereas 13% (n = 2) developed chronic GVHD. Alemtuzumab is an effective second-line agent for children and young adults with SR-aGVHD. Higher alemtuzumab levels are associated with CR. A real-time dose adjusted alemtuzumab study is needed to further optimize the dose of alemtuzumab in aGVHD.


Therapeutic Drug Monitoring | 2015

Age-dependent changes in sirolimus metabolite formation in patients with neurofibromatosis type 1.

Chie Emoto; Tsuyoshi Fukuda; Tomoyuki Mizuno; Shareen Cox; Björn Schniedewind; Uwe Christians; Brigitte C. Widemann; Michael J. Fisher; Brian Weiss; John P. Perentesis; Alexander A. Vinks

Background: Sirolimus is an inhibitor of mammalian target of rapamycin, which exhibits large interindividual pharmacokinetic variability. We report sirolimus pharmacokinetic data collected as part of a concentration-controlled multicenter phase II clinical trial in pediatric patients with neurofibromatosis type 1. The purpose of this study was to explore the effect of growth on age-dependent changes in sirolimus clearance with a focus on cytochrome P450 3A (CYP3A) subfamily mediated metabolism. Methods: Predose blood samples were obtained at steady state from 18 patients with neurofibromatosis type 1. Sirolimus and its 5 CYP3A-dependent primary metabolites were quantified by HPLC–UV/MS. Concentration ratios of metabolites to sirolimus (metabolic ratio) were calculated as an index of metabolite formation. Results: Metabolic ratios of the main metabolites, 16-O-demethylsirolimus (16-O-DM) and 24-hydroxysirolimus (24OH), were significantly correlated with sirolimus clearance, whereas this was not the case for the other 3 metabolites (25-hydroxysirolimus, 46-hydroxysirolimus, and 39-O-demethylsirolimus). The ratios for the 16-O-DM and 24OH metabolites were lower in children than adults. No significant difference in allometrically scaled metabolic ratios of 16-O-DM and 24OH was observed between children and adults. Conclusions: This study suggests that the age-dependent changes in sirolimus clearance can be explained by size-related increases in CYP3A metabolic capacity, most likely due to liver and intestinal growth. These findings will help facilitate the development of age-appropriate dosing algorithms for sirolimus in infants and children.


Pediatric Blood & Cancer | 2017

Developmental pharmacokinetics of sirolimus: Implications for precision dosing in neonates and infants with complicated vascular anomalies

Tomoyuki Mizuno; Tsuyoshi Fukuda; Chie Emoto; Paula S. Mobberley-Schuman; Adrienne M. Hammill; Denise M. Adams; Alexander A. Vinks

Sirolimus has recently been shown to be efficacious and tolerable in pediatric patients with complicated vascular anomalies. Nevertheless, dosing information remains very limited especially for neonates and infants. The purpose of this study was to develop an age‐appropriate sirolimus starting dosing regimen based on the developmental changes in drug elimination capacity using data collected in neonates and infants.


European Journal of Pharmaceutical Sciences | 2017

Model-based precision dosing of sirolimus in pediatric patients with vascular anomalies

Tomoyuki Mizuno; Chie Emoto; Tsuyoshi Fukuda; Adrienne M. Hammill; Denise M. Adams; Alexander A. Vinks

Abstract Sirolimus is the first drug to show efficacy in the treatment of patients with complicated vascular anomalies. The current study expands on the evolution of a PK model‐based strategy for the precision dosing of sirolimus as part of prospective concentration controlled clinical trials in pediatric patients with vascular anomalies. Twelve month follow up data collected from 52 pediatric patients participating in the Phase 2 clinical trial were analyzed. Target attainment across the age range of 3 weeks to 18 years after 2–3 months of therapy was 94% (49 out of 52 patients). The mean sirolimus dose to achieve the target of ˜10 ng/mL for patients older than 2 years was 1.8 mg/m2 twice daily (range 0.8–2.9), while it was 0.7 to 1.6 mg/m2 twice daily for patients 3 weeks of age to 2 years. A total of 676 blood concentration data were used for the population PK analysis by nonlinear mixed effect modeling using NONMEM. The final model included a maturation function for sirolimus clearance and allometrically scaled body weight to account for size differences. The mean allometrically scaled sirolimus clearance estimates increased from 3.9 to 17.0 L/h per 70 kg with age from shortly after the birth to 2 years of age while the mean estimate for patients older than 2 years was 18.5 L/h per 70 kg. The developed model based dosing strategy provides a foundation for ongoing efforts to define the sirolimus exposure‐response and clinical outcome relationships across the pediatric age spectrum from birth to adolescence. Graphical abstract No caption available.


Therapeutic Drug Monitoring | 2016

Risk Assessment of Drug-Drug Interactions of Calcineurin Inhibitors Affecting Sirolimus Pharmacokinetics in Renal Transplant Patients.

Chie Emoto; Alexander A. Vinks; Tsuyoshi Fukuda

Background: Sirolimus is a mammalian target of rapamycin inhibitor that is being used to prevent organ rejection in kidney transplant patients often in combination with calcineurin inhibitors (CNIs; cyclosporine and tacrolimus). All 3 drugs are metabolized primarily by CYP3As. Clinical drug–drug interaction (DDI) studies of cyclosporine on sirolimus pharmacokinetics have been reported; however, there are a few clinical DDI data related to tacrolimus. Methods: In vitro inhibition assay with sirolimus were conducted using recombinant CYP3As and human microsomes in the presence and absence of CNIs. Sirolimus concentrations were determined by validated high-performance liquid chromatography-tandem mass spectrometry (LC/MS-MS) assay. The DDI risk in terms of increase in sirolimus area under the curve (AUC) was evaluated by a mechanistic model using in vitro inhibition data and published pharmacokinetic parameters of CNIs. Results: Both CNIs showed similar inhibitory effects on sirolimus metabolism in human liver and intestinal microsomes. Cyclosporine predominantly inhibited CYP3A4 (half maximal inhibitory concentration = 0.71 µM) rather than CYP3A5 (>5 µM), whereas tacrolimus showed similar inhibition for CYP3A4 (0.29 µM) and CYP3A5 (0.41 µM). The predicted increase in AUC of sirolimus during the coadministration of cyclosporine was 3.9-fold, which was comparable to the observed clinical data (3.3-fold) in healthy volunteers. Sirolimus AUC was estimated to a 2.8- to 3.2-fold increase during the coadministration of tacrolimus, based on the reported Cmax values and doses of tacrolimus in kidney transplant patients. In addition, exploratory sensitivity analysis indicated that the predicted increase in sirolimus AUC was sensitive to the free fraction of cyclosporine but not to the free fraction of tacrolimus. Conclusions: This study suggests that tacrolimus has a lower clinical DDI risk potential affecting sirolimus pharmacokinetics compared with cyclosporine in kidney transplant patients.


Clinical Pharmacology & Therapeutics | 2018

Influence of OCT1 ontogeny and genetic variation on morphine disposition in critically ill neonates: lessons from PBPK modeling and clinical study

David Hahn; Chie Emoto; Joshua C. Euteneuer; Tomoyuki Mizuno; Alexander A. Vinks; Tsuyoshi Fukuda

Morphine is commonly used for analgesia in the neonatal intensive care unit (NICU) despite having highly variable pharmacokinetics (PKs) between individual patients. The pharmacogenetic (PG) effect of variants at the loci of organic cation transporter 1 (OCT1) and UDP‐glucuronosyltransferase 2B7 (UGT2B7) on age‐dependent morphine clearance were evaluated in a cohort of critically ill neonatal patients using an opportunistic sampling design. Our primary results demonstrate the significant influence of OCT1 genotype (P < 0.05) and gestational age (P ≤ 0.005) on morphine PKs. A physiologically based pharmacokinetic (PBPK) model for morphine that accounted for OCT1 ontogeny and PG effect in post‐term neonates adequately described the clinically observed variability in morphine PKs. This study serves as a proof of concept for genotype‐dependent drug transporter ontogeny in neonates.

Collaboration


Dive into the Chie Emoto's collaboration.

Top Co-Authors

Avatar

Alexander A. Vinks

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tsuyoshi Fukuda

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tomoyuki Mizuno

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lisa Neumeier

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Parinda A. Mehta

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Adrienne M. Hammill

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Hahn

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Denise M. Adams

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Pooja Khandelwal

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rebecca A. Marsh

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge