Network


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

Hotspot


Dive into the research topics where Alexander A. Vinks is active.

Publication


Featured researches published by Alexander A. Vinks.


Pediatrics | 2016

Efficacy and Safety of Sirolimus in the Treatment of Complicated Vascular Anomalies.

Denise M. Adams; Cameron C. Trenor; Adrienne M. Hammill; Alexander A. Vinks; Manish N. Patel; Gulraiz Chaudry; Mary Sue Wentzel; Paula S. Mobberley-Schuman; Lisa M. Campbell; Christine Brookbank; Anita Gupta; Carol Chute; Jennifer Eile; Jesse McKenna; Arnold C. Merrow; Lin Fei; Lindsey Hornung; Michael Seid; A. Roshni Dasgupta; Belinda Dickie; Ravindhra G. Elluru; Anne W. Lucky; Brian Weiss; Richard G. Azizkhan

BACKGROUND AND OBJECTIVES: Complicated vascular anomalies have limited therapeutic options and cause significant morbidity and mortality. This Phase II trial enrolled patients with complicated vascular anomalies to determine the efficacy and safety of treatment with sirolimus for 12 courses; each course was defined as 28 days. METHODS: Treatment consisted of a continuous dosing schedule of oral sirolimus starting at 0.8 mg/m2 per dose twice daily, with pharmacokinetic-guided target serum trough levels of 10 to 15 ng/mL. The primary outcomes were responsiveness to sirolimus by the end of course 6 (evaluated according to functional impairment score, quality of life, and radiologic assessment) and the incidence of toxicities and/or infection-related deaths. RESULTS: Sixty-one patients were enrolled; 57 patients were evaluable for efficacy at the end of course 6, and 53 were evaluable at the end of course 12. No patient had a complete response at the end of course 6 or 12 as anticipated. At the end of course 6, a total of 47 patients had a partial response, 3 patients had stable disease, and 7 patients had progressive disease. Two patients were taken off of study medicine secondary to persistent adverse effects. Grade 3 and higher toxicities attributable to sirolimus included blood/bone marrow toxicity in 27% of patients, gastrointestinal toxicity in 3%, and metabolic/laboratory toxicity in 3%. No toxicity-related deaths occurred. CONCLUSIONS: Sirolimus was efficacious and well tolerated in these study patients with complicated vascular anomalies. Clinical activity was reported in the majority of the disorders.


Clinical Pharmacology & Therapeutics | 2015

Clinical Pharmacogenetics Implementation Consortium (CPIC) Guidelines for CYP3A5 Genotype and Tacrolimus Dosing

Kelly A. Birdwell; B. Decker; Julia M. Barbarino; Josh F. Peterson; C.M. Stein; Wolfgang Sadee; Danxin Wang; Alexander A. Vinks; Y. He; Jesse J. Swen; J.S. Leeder; Ron H.N. van Schaik; Kenneth E. Thummel; Teri E. Klein; Kelly E. Caudle; I.A.M. MacPhee

Tacrolimus is the mainstay immunosuppressant drug used after solid organ and hematopoietic stem cell transplantation. Individuals who express CYP3A5 (extensive and intermediate metabolizers) generally have decreased dose‐adjusted trough concentrations of tacrolimus as compared with those who are CYP3A5 nonexpressers (poor metabolizers), possibly delaying achievement of target blood concentrations. We summarize evidence from the published literature supporting this association and provide dosing recommendations for tacrolimus based on CYP3A5 genotype when known (updates at www.pharmgkb.org).


Clinical Pharmacology & Therapeutics | 2015

Modeling and simulation in pediatric drug therapy: Application of pharmacometrics to define the right dose for children

Alexander A. Vinks; Chie Emoto; Tsuyoshi Fukuda

During the past decades significant progress has been made in our understanding of the importance of age‐appropriate development of new drug therapies in children. Importantly, several regulatory initiatives in Europe and the US have provided a framework for a rationale. In the US, most notably the enactment of the Best Pharmaceuticals for Children Act (BPCA) and Product Research and Equity Act (PREA) has facilitated the studying of on‐patent and off‐patent drugs in children. The biggest challenge in pediatric studies is defining a safe and effective dose or dose range in a patient population that can span from premature neonates to adolescents. From a mechanism‐based perspective, advances in the science of quantitative pharmacology and pharmacometrics have resulted in the development of model‐based approaches to better describe and understand important age‐related factors influencing drug disposition and response in pediatric patients. The application of modeling and simulation has been shown to result in better estimates of pediatric doses as evidenced by several studies, although the optimal approach is still being debated. The extrapolation of efficacy findings from adults to the pediatric population has streamlined the development process especially for studies in older children. However, a focus on developmental changes in neonates and infants as well as further developing a paradigm for conducting pharmacodynamic studies in neonates, infants, and children remain important unmet needs. In this overview we will review current approaches for age‐appropriate dose selection and highlight ongoing efforts to define exposure–response and clinical outcome relationships across the pediatric age spectrum.


CPT: Pharmacometrics & Systems Pharmacology | 2015

Development of a Pediatric Physiologically Based Pharmacokinetic Model for Sirolimus: Applying Principles of Growth and Maturation in Neonates and Infants

Chie Emoto; Tsuyoshi Fukuda; Trevor N. Johnson; Denise M. Adams; Alexander A. Vinks

This study describes the maturation of sirolimus clearance in a cohort of very young pediatric patients with vascular anomalies. The relationship between allometrically scaled in vivo clearance and age was described by the Emax model in patients aged 1 month to 2 years. Consistent with the observed increase, in vitro intrinsic clearance of sirolimus using pediatric liver microsomes showed a similar age‐dependent increase. In children older than 2 years, allometrically scaled sirolimus clearance did not show further maturation. Simulated clearance estimates with a sirolimus physiologically based pharmacokinetic model that included CYP3A4/5/7 and CYP2C8 maturation profiles were in close agreement with observed in vivo clearance values. In addition, physiologically based pharmacokinetic model‐simulated sirolimus pharmacokinetic profiles predicted the actual observations well. These results demonstrate the utility of a physiologically based pharmacokinetic modeling approach for the prediction of the developmental trajectory of sirolimus metabolic activity and its effects on total body clearance in neonates and infants.


CPT: Pharmacometrics & Systems Pharmacology | 2017

Characterization of Contributing Factors to Variability in Morphine Clearance Through PBPK Modeling Implemented With OCT1 Transporter

Chie Emoto; Tsuyoshi Fukuda; Trevor N. Johnson; Sibylle Neuhoff; S Sadhasivam; Alexander A. Vinks

Morphine shows large interindividual variability in its pharmacokinetics; however, the cause of this has not been fully addressed. The variability in morphine disposition is considered to be due to a combination of pharmacogenetic and physiological determinants related to morphine disposition. We previously reported the effect of organic cation transporter (OCT1) genotype on morphine disposition in pediatric patients. To further explore the underlying mechanisms for variability arising from relevant determinants, including OCT1, a physiologically based pharmacokinetic (PBPK) model of morphine was developed. The PBPK model predicted morphine concentration‐time profiles well, in both adults and children. Almost all of the observed morphine clearances in pediatric patients fell within a twofold range of median predicted values for each OCT1 genotype in each age group. This PBPK modeling approach quantitatively demonstrates that OCT1 genotype, age‐related growth, and changes in blood flow as important contributors to morphine pharmacokinetic (PK) variability.


The Journal of Clinical Pharmacology | 2014

Meropenem in children receiving continuous renal replacement therapy: Clinical trial simulations using realistic covariates

Edward J. Nehus; Samer Mouksassi; Alexander A. Vinks; Stuart L. Goldstein

Meropenem is frequently prescribed in children receiving continuous renal replacement therapy (CRRT). Fluid overload is often present in critically ill children and affects drug disposition. The purpose of this study was to develop a pharmacokinetic model to (1) evaluate target attainment of meropenem dosing regimens against P. aeruginosa in children receiving CRRT and (2) estimate the effect of fluid overload on target attainment. Clinical trial simulations were employed to evaluate target attainment of meropenem in various age groups and degrees of fluid overload in children receiving CRRT. Pharmacokinetic parameters were extracted from published literature, and 287 patients from the prospective pediatric CRRT registry database provided realistic clinical covariates including patient weight, fluid overload, and CRRT prescription characteristics. Target attainment at 40% and 75% time above the minimum inhibitory concentration was evaluated. Clinical trial simulations demonstrated that children greater than 5 years of age achieved acceptable target attainment with a dosing regimen of 20 mg/kg every 12 hours. In children less than 5, however, increased dosing of 20 mg/kg every 8 hours was needed to optimize target attainment. Fluid overload did not affect target attainment. These in silico model predictions will need to be verified in vivo in children receiving meropenem and CRRT.


The Journal of Clinical Pharmacology | 2016

Pharmacokinetics of meropenem in children receiving continuous renal replacement therapy: Validation of clinical trial simulations

Edward J. Nehus; Tomoyuki Mizuno; Shareen Cox; Stuart L. Goldstein; Alexander A. Vinks

Meropenem is frequently prescribed in critically ill children receiving continuous renal replacement therapy (CRRT). We previously used clinical trial simulations to evaluate dosing regimens of meropenem in this population and reported that a dose of 20 mg/kg every 12 hours optimizes target attainment. Meropenem pharmacokinetics were investigated in this prospective, open‐label study to validate our previous in silico predictions. Seven patients received meropenem (13.8–22 mg/kg) administered intravenously every 12 hours as part of standard care. A mean dose of 18.6 mg/kg of meropenem was administered, resulting in a mean peak concentration of 80.1 μg/mL. Meropenem volume of distribution was 0.35 ± 0.085 L/kg. CRRT clearance was 40.2 ± 6.6 mL/(min · 1.73 m2) and accounted for 63.4% of the total clearance of 74.8 ± 36.9 mL/(min · 1.73 m2). Simulations demonstrated that a dose of 20 mg/kg every 12 hours resulted in a time above the minimum inhibitory concentration (%fT > MIC) of 100% in 5 out of 7 subjects, with a %fT > MIC of 93% and 43% in the remaining 2 subjects. We conclude that CRRT contributed significantly to the total clearance of meropenem. A dosing regimen of 20 mg/kg achieved good target attainment in critically ill children receiving CRRT, which is consistent with our previously published in silico predictions.


Clinical Pharmacology & Therapeutics | 2017

Precision Medicine—Nobody Is Average

Alexander A. Vinks

Medicine gets personal and tailor‐made treatments are underway. Hospitals have started to advertise their advanced genomic testing capabilities and even their disruptive technologies to help foster a culture of innovation. The prediction in the lay press is that in decades from now we may look back and see 2017 as the year precision medicine blossomed. It is all part of the Precision Medicine Initiative that takes into account individual differences in peoples genes, environments, and lifestyles.


Clinical Pharmacology & Therapeutics | 2017

Learning Health Systems as Facilitators of Precision Medicine

Lb Ramsey; T Mizuno; Alexander A. Vinks; Pa Margolis

The concept of the learning health system offers promise to facilitate personalized medicine. In children, serious illness is uncommon so generating and applying new knowledge therefore requires networks of institutions. To illustrate the concept of the Learning Health System, we describe the example of the ImproveCareNow Network. We then use a network case study to illustrate how the concept of precision medicine can be achieved through a Learning Health System in a real-world clinical environment.


CPT: Pharmacometrics & Systems Pharmacology | 2018

PBPK Model of Morphine Incorporating Developmental Changes in Hepatic OCT1 and UGT2B7 Proteins to Explain the Variability in Clearances in Neonates and Small Infants

Chie Emoto; Trevor N. Johnson; Sibylle Neuhoff; David Hahn; Alexander A. Vinks; Tsuyoshi Fukuda

Morphine has large pharmacokinetic variability, which is further complicated by developmental changes in neonates and small infants. The impacts of organic cation transporter 1 (OCT1) genotype and changes in blood‐flow on morphine clearance (CL) were previously demonstrated in children, whereas changes in UDP‐glucuronosyltransferase 2B7 (UGT2B7) activity showed a small effect. This study, targeting neonates and small infants, was designed to assess the influence of developmental changes in OCT1 and UGT2B7 protein expression and modified blood‐flow on morphine CL using physiologically based pharmacokinetic (PBPK) modeling. The implementation of these three age‐dependent factors into the pediatric system platform resulted in reasonable prediction for an age‐dependent increase in morphine CL in these populations. Sensitivity of morphine CL to changes in cardiac output increased with age up to 3 years, whereas sensitivity to changes in UGT2B7 activity decreased. This study suggests that morphine exhibits age‐dependent extraction, likely due to the developmental increase in OCT1 and UGT2B7 protein expression/activity and hepatic blood‐flow.

Collaboration


Dive into the Alexander A. Vinks's collaboration.

Top Co-Authors

Avatar

Richard Wenstrup

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

John Pestian

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tracy A. Glauser

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

Chie Emoto

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Denise M. Adams

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Stuart L. Goldstein

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

Brian Weiss

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge