Edgar Schuck
Eisai
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Edgar Schuck.
Bioorganic & Medicinal Chemistry Letters | 2011
Sridhar Narayan; Eric M. Carlson; Hongsheng Cheng; Krista Condon; Hong Du; Sean Eckley; Yongbo Hu; Yimin Jiang; Vipul Kumar; Bryan M. Lewis; Philip Saxton; Edgar Schuck; Boris M. Seletsky; Karen TenDyke; Huiming Zhang; Wanjun Zheng; Bruce A. Littlefield; Murray J. Towle; Melvin J. Yu
Eribulin mesylate is a newly approved treatment for locally advanced and metastatic breast cancer. We targeted oral bioavailability and efficacy against multidrug resistant (MDR) tumors for further work. The design, synthesis and evaluation of novel amine-containing analogs of eribulin mesylate are described in this part. Attenuation of basicity of the amino group(s) in the C32 side-chain region led to compounds with low susceptibility to PgP-mediated drug efflux. These compounds were active against MDR tumor cell lines in vitro and in xenograft models in vivo, in addition to being orally bioavailable.
The Journal of Clinical Pharmacology | 2014
Yanke Yu; Veronika Logovinsky; Edgar Schuck; June Kaplow; Min-Kun Chang; Takehiko Miyagawa; Nancy Wong; Jim Ferry
E2212, a novel γ‐secretase modulator, is under development for the treatment of Alzheimers disease. The safety, tolerability, pharmacokinetics, and pharmacodynamics of single ascending oral doses (10–250 mg, double‐blind, placebo‐controlled, randomized) of E2212 were evaluated. In this phase I clinical trial, E2212 was found to be well tolerated in single doses. Maximum tolerated dose was not achieved up to 250 mg. Most AEs were mild to moderate in severity with no identifiable dose related pattern. There were no clinically significant findings on physical and ophthalmologic examinations as well as vital signs, laboratory, ECG and C‐SSRS assessments. E2212 was rapidly absorbed, with median tmax values ranging from 0.5 to 1.0 h. E2212 exhibited biphasic disposition with the terminal t1/2 of 12.5–19.0 h. Renal excretion was the minor pathway for E2212 elimination. Increased PD response (reduction in plasma concentrations of Aβ(x–42)) was observed with increasing doses. The maximum PD response was observed in the highest dose 250 mg cohort, with ΔAUAC(0–24 h) of 44.1% and Amax of 53.6%. These results support further clinical development of E2212.
Xenobiotica | 2011
Jan-Shiang Taur; Christopher DesJardins; Edgar Schuck; Y. Nancy Wong
Eribulin is a new anticancer agent currently in Phase III clinical trials for the treatment of metastatic breast cancer. In the current studies, we have investigated the effects of P-glycoprotein (P-gp) on the in vivo disposition of eribulin using CF-1 abcb1a-deficient mice, and the influence of eribulin on P-gp-mediated efflux of digoxin in Caco-2 cells. Eribulin was administered intravenously and orally in both CF-1 wild-type and CF-1 abcb1a-deficient mice. P-gp-mediated efflux of digoxin in Caco-2 cell monolayers was measured in the presence of eribulin. The plasma exposure to eribulin was higher in CF-1 abcb1a-deficient mice than that in CF-1 wild-type mice after intravenous (IV) and oral (PO) administrations. The oral bioavailability of eribulin was 62.3% in CF-1 abcb1a-deficient mice compared with 7.6% in wild-type mice. The brain penetration of eribulin in CF-1 abcb1a-deficient mice was 30-fold greater than that in wild-type mice. Eribulin decreased the efflux ratio of digoxin in a concentration-dependent manner, with the result of IC50 greater than 10 µM. The [I]/IC50 of eribulin was estimated to be <0.05. P-gp is likely to limit the oral absorption and brain penetration of eribulin in CF-1 wild-type mice. Eribulin inhibited the efflux of digoxin with IC50 greater than 10 µM in Caco-2 cells. These results suggest that eribulin, given intravenously at the clinically relevant concentrations, may not alter P-gp-mediated disposition of concurrently administered drugs.
Bioorganic & Medicinal Chemistry Letters | 2011
Sridhar Narayan; Eric M. Carlson; Hongsheng Cheng; Krista Condon; Hong Du; Sean Eckley; Yongbo Hu; Yimin Jiang; Vipul Kumar; Bryan M. Lewis; Philip Saxton; Edgar Schuck; Boris M. Seletsky; Karen TenDyke; Huiming Zhang; Wanjun Zheng; Bruce A. Littlefield; Murray J. Towle; Melvin J. Yu
Novel second generation analogs of eribulin mesylate, a tubulin agent recently approved for the treatment of breast cancer, are reported. Our recent efforts have focused on expanding the target indications for this class of compounds to other tumor types. Herein, we describe the design, synthesis and evaluation of eribulin analogs active against brain tumor cell lines in vitro and corresponding brain tumor models in mice. Attenuation of basicity of the amino group(s) in the C32 side-chain region led to compounds with lower susceptibility to P-gp mediated drug efflux, allowing these compounds to permeate through the blood-brain barrier. In preclinical in vivo studies, these compounds showed significantly higher levels in the brain and cerebrospinal fluid as compared to eribulin. In addition, analogs within this series showed antitumor activity in an orthotopic murine model of human glioblastoma.
Alzheimers & Dementia | 2010
Christa Nagy; Edgar Schuck; Akira Ishibashi; Yosuke Nakatani; Bhaskar Rege; Veronika Logovinsky
Results: E2012 reduced plasma Aβ40 and Aβ42 in healthy subjects in a dose-related manner. Aβ42 decreased to a greater extent than Aβ40 (maximum reductions at 400 mg ~ 50% and ~30%, respectively) at 4-6 hrs post-dose, without a rebound effect. AUC0-inf increased proportionally up to 400 mg. Cmax increased proportionally up to 200 mg and less than proportionally thereafter. Results from rat studies with E2012 were predictive of the results obtained in humans for plasma Aβ effect over 24 hr. Effects on rat CSF and brain Aβ followed similar trends, supporting the use of CSF as a surrogate matrix for effects on brain Aβ in humans.
International Journal of Pharmaceutics | 2013
Yanke Yu; Christopher DesJardins; Phil Saxton; George Lai; Edgar Schuck; Y. Nancy Wong
Eribulin mesylate (E7389), a tubulin and microtubule inhibitor, has been approved to treat metastatic breast cancer in certain patient populations. A liposomal formulation of E7389, E7389-LF, aims to increase the therapeutic profile of E7389. As determining the free drug concentration is crucial for the assessment of efficacy and toxicity of liposomal drug, in this study, an ultracentrifugation method coupled with LC-MS/MS was developed to separate the free E7389 from liposomal and protein bound E7389. The pharmacokinetics of the free E7389 after dosing either E7389 or E7389-LF was characterized. The concentration ratio of E7389 in ultracentrifuged mice plasma (UCM) vs E7389 in plasma after a 2mg/kg i.v. of E7389 ranged from 54.19% to 65.41%, which was similar to the free fraction in the mouse plasma. The respective concentration ratio of E7389 in UCM vs E7389 in plasma after a 2mg/kg i.v. of E7389-LF ranged from 0.07% to 0.59%, and the exposure, expressed as AUC, of UCM/plasma ratio was determined to be 0.2%. Pharmacokinetic modeling was performed to estimate the release kinetics of E7389 from E7389-LF, and the release was best described by a first order rate constant k(rel) 0.078 h(-1). Sensitivity analysis demonstrated that further decrease the release rate constant by adjusting liposome formulation would lead to decreased C(max) and much longer half-life of UCM E7389, which might result in better efficacy and lower toxicity.
Clinical Pharmacology & Therapeutics | 2018
Mohamad Shebley; Punam Sandhu; Arian Emami Riedmaier; Masoud Jamei; Rangaraj Narayanan; Aarti Patel; Sheila Annie Peters; Venkatesh Pilla Reddy; Ming Zheng; Loeckie de Zwart; Maud Bénéton; Francois Bouzom; Jun Chen; Yuan Chen; Yumi Cleary; Christiane Collins; Gemma L. Dickinson; Nassim Djebli; Heidi J. Einolf; Iain Gardner; Felix Huth; Faraz Kazmi; Feras Khalil; Jing Lin; Aleksandrs Odinecs; Chirag Patel; Haojing Rong; Edgar Schuck; Pradeep Sharma; Shu‐Pei Wu
This work provides a perspective on the qualification and verification of physiologically based pharmacokinetic (PBPK) platforms/models intended for regulatory submission based on the collective experience of the Simcyp Consortium members. Examples of regulatory submission of PBPK analyses across various intended applications are presented and discussed. European Medicines Agency (EMA) and US Food and Drug Administration (FDA) recent draft guidelines regarding PBPK analyses and reporting are encouraging, and to advance the use and acceptability of PBPK analyses, more clarity and flexibility are warranted.
European Journal of Paediatric Neurology | 2016
Alexis Arzimanoglou; Jose Ferreira; Andrew Satlin; Shannon Mendes; Betsy Williams; David Critchley; Edgar Schuck; Ziad Hussein; Dinesh Kumar; Shobha Dhadda; Francesco Bibbiani
OBJECTIVE A good knowledge of safety and age group-specific pharmacokinetics (PK) of antiepileptic drugs (AEDs) in young pediatric patients is of great importance in clinical practice. This paper presents 6-month interim safety and PK from an ongoing 2-year open-label study (Study 303) of adjunctive rufinamide treatment in pediatric subjects ≥ 1 to < 4 years with inadequately controlled epilepsies of the Lennox-Gastaut syndrome (LGS) spectrum. METHODS Subjects (N = 37) were randomized to either rufinamide or any other approved AED chosen by the investigator as adjunctive therapy to the subjects existing regimen of 1-3 AEDs. RESULTS Interim safety results showed that treatment-emergent adverse events (TEAEs) were similar between the rufinamide (22 [88.0%]) and any-other-AED group (9 [81.8%]), with most events considered mild or moderate. A population PK analysis was conducted including plasma rufinamide concentrations from Study 303 and two other study populations of LGS subjects ≥ 4 years. The rufinamide PK profile was dose independent. The apparent clearance (CL/F) estimated from the PK model was 2.19 L/h; it was found to increase significantly as a function of body weight. Coadministration of valproic acid significantly decreased rufinamide CL/F. CL/F was not significantly affected by other concomitant AEDs, age, gender, race, hepatic function, or renal function. No adjustments to body weight-based rufinamide dosing in subjects ≥ 1 to < 4 years are necessary. SIGNIFICANCE Rufinamide was safe and well tolerated in these pediatric subjects. Results from the interim analysis demonstrate that rufinamides safety and PK profile is comparable in subjects ≥ 1 to < 4 and ≥ 4 years with LGS. CLINICAL TRIAL REGISTRATION Study 303 (clinicaltrials.gov: NCT01405053).
Epilepsy Research | 2017
Barry E. Gidal; Rama Maganti; Antonio Laurenza; Haichen Yang; David Verbel; Edgar Schuck; Jim Ferry
OBJECTIVES Perampanel, a selective, noncompetitive AMPA receptor antagonist, is indicated as adjunctive therapy for the treatment of partial seizures with or without secondarily generalized seizures and primary generalized tonic-clonic seizures in patients with epilepsy aged 12years and older. In vitro studies and Phase I trials indicate that perampanel is metabolized almost exclusively by CYP3A, with an elimination half-life (t1/2) averaging approximately 105h. Understanding of pharmacokinetic (PK) interactions-enzyme inhibition or induction-and anticipating their occurrence are important for management of patients with epilepsy. Here we report PK results from a Phase I drug-drug interaction (DDI) study (Study 005) combining perampanel with the CYP3A inhibitor ketoconazole, as well as supplementary in silico predictions further exploring this interaction. METHODS A Phase I, randomized, open-label, two-period, two-treatment, two-way crossover study was conducted in 26 healthy adult male volunteers. Subjects were randomized to 1 of 2 treatment sequences. In one period, subjects received a single 1-mg fasting dose of perampanel (Day1); in the other period, subjects received ketoconazole 400mg once daily for 10days with a single 1-mg perampanel dose while fasting (Day3). Blood samples were drawn at multiple time points up to 288h after the perampanel dose. Pharmacokinetic parameters of perampanel were calculated by noncompartmental analysis, and safety was recorded. An integrated, physiologically based PK model built in Simcyp® provided additional insight into this interaction. Drug-drug interaction intensity was measured by the ratio of systemic exposure (area under plasma concentration-time curve [AUC]) of perampanel in the presence or absence of concomitant ketoconazole. RESULTS Single oral doses of 1mg perampanel and once-daily oral doses of ketoconazole 400mg were safe and well tolerated. Maximum perampanel plasma concentration (Cmax) and time to Cmax showed no apparent differences when perampanel was administered alone versus with ketoconazole. Ketoconazole co-administration resulted in an approximate 20% increase in perampanel AUC (P<0.001). This increase, although statistically significant, was a<2.0-fold AUC change and alone would suggest a modest effect of ketoconazole. To further explore these results, DDI simulations were performed to query the findings and test additional study conditions. Using the actual trial conditions of Study 005, the simulations also predicted an AUC ratio increase <2-fold, providing verification of the simulation assumptions and the modest effect of ketoconazole for 10days. Simulations further suggested that an interaction effect of ketoconazole on perampanel exposure (>2-fold) of potential clinical significance could be predicted when using larger doses of ketoconazole (e.g., 200mg every 6h) coadministered for a greater time period (e.g., 30days), with AUC ratio as high as 3.36. Additionally, simulations suggested that a significant interaction with co-administration of perampanel and an inhibitor more potent than ketoconazole (such as itraconazole) could not be ruled out. CONCLUSIONS Selecting an appropriate study design is critical to fully characterize the PK interaction for drugs such as perampanel that have a long t1/2. Although a negligible effect on perampanel PK was observed following co-administration of ketoconazole 400mg/day for 10days, this is likely due in part to the relatively brief co-administration period of ketoconazole and perampanel (<3 times the t1/2 of perampanel). While short-term administration of a CYP3A inhibitor may not significantly increase perampanel exposure, such increases may be expected following chronic and larger dosing or with a more potent inhibitor.
Drug Metabolism Letters | 2013
Jan-Shiang Taur; Edgar Schuck; Nancy Wong
To provide a fast assessment in predicting P-gp-mediated DDI risk during early stage of drug development, a transcellular P-gp inhibition assay using two concentrations is presented in the present study. The efflux ratios of loperamide in the presence of forty-five commercial compounds at two concentrations were measured and compared to that of six concentrations in human P-gp cDNA-expressing LLC-PK1 cells (LLC-MDR1). The inhibition potency calculated from the change on the efflux ratio (ER) and on the net secretory flux (NSF) of loperamide was investigated. The P-gp inhibition potency was defined as potent (IC50 < 1 µM), moderate (1 µM < IC50 < 10 µM), or weak (IC50 > 10 µM). The results using 1 µM and 10 µM of inhibitor concentrations provided the best correlation and are most consistent with those generated from a 6-point approach.