Network


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

Hotspot


Dive into the research topics where Venkat Sethuraman is active.

Publication


Featured researches published by Venkat Sethuraman.


Clinical Pharmacology & Therapeutics | 2010

Clinical Pharmacokinetics of the BCR–ABL Tyrosine Kinase Inhibitor Nilotinib

Chiaki Tanaka; Ophelia Q. P. Yin; Venkat Sethuraman; Tom Smith; X Wang; Karen Grouss; H. Kantarjian; Francis J. Giles; Oliver G. Ottmann; Lawrence Galitz; Horst Schran

This article describes studies that investigated the pharmacokinetics of nilotinib, a highly specific, oral, second‐generation BCR–ABL tyrosine kinase inhibitor. After a once‐ or twice‐daily regimen at doses ranging from 50 to 1,200 mg/day in 119 patients with chronic myeloid leukemia (CML), the area under the serum concentration–time curve (AUC) and peak serum concentration (Cmax) of nilotinib were found to be nearly dose proportional up to a dose of 400 mg once daily. Solubility‐limited absorption at higher doses was observed, but this was partially overcome by dividing the daily dose into two. For instance, the administration of 400 mg nilotinib twice daily resulted in a 35% increase in AUC as compared to a once‐daily dose of 800 mg. Exploratory pharmacodynamic assessment showed a general trend of greater reduction in white blood cell (WBC) levels with increase in nilotinib concentrations. This finding was consistent with the observation of an 82% reduction in WBC levels in patients after a regimen of 400 mg nilotinib twice daily for 15 days. The type and quantity of food intake variably affected nilotinib absorption. When administered after a high‐fat meal, the AUC of nilotinib increased by 50% in CML patients (n = 10) and by 82% in healthy volunteers (n = 44).


British Journal of Clinical Pharmacology | 2008

Effects of imatinib (Glivec) on the pharmacokinetics of metoprolol, a CYP2D6 substrate, in Chinese patients with chronic myelogenous leukaemia.

Yanfeng Wang; Li Zhou; Catherine Dutreix; Elisabeth Leroy; Qi Yin; Venkat Sethuraman; Gilles-Jacques Riviere; Ophelia Q. P. Yin; Horst Schran; Zhi-Xiang Shen

WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT Imatinib, a tyrosine kinase inhibitor, exhibits a competitive inhibition on the CYP450 2D6 isozyme with a K(i) value of 7.5 microm. However, the clinical significance of the inhibition and its relevance to 2D6 polymorphisms have not been evaluated. The pharmacokinetics of imatinib have been well studied in Caucasians, but not in a Chinese population. Metoprolol, a CYP2D6 substrate, has different clearances among patients with different CYP2D6 genotypes. It is often used as a CYP2D6 probe substrate for clinical drug-drug interaction studies. WHAT THIS STUDY ADDS Co-administration of imatinib at 400 mg twice daily increased the plasma AUC of metoprolol by approximately 23% in 20 Chinese patients with chronic myeloid leukaemia (CML), about 17% increase in CYP2D6 intermediate metabolizers (IMs) (n = 6), 24% in extensive metabolizers (EMs) (n = 13), and 28% for the subject with unknown 2D6 status (n = 1) suggesting that imatinib has a weak to moderate inhibition on CYP2D6 in vivo. * The clearance of imatinib in Chinese patients with CML showed no difference between CYP2D6 IMs and EMs, and no major difference from Caucasian patients with CML based on data reported in the literature. AIMS To investigate the effect of imatinib on the pharmacokinetics of a CYP2D6 substrate, metoprolol, in patients with chronic myeloid leukaemia (CML). The pharmacokinetics of imatinib were also studied in these patients. METHODS Patients (n = 20) received a single oral dose of metoprolol 100 mg on day 1 after an overnight fast. On days 2-10, imatinib 400 mg was administered twice daily. On day 8, another 100 mg dose of metoprolol was administered 1 h after the morning dose of imatinib 400 mg. Blood samples for metoprolol and alpha-hydroxymetoprolol measurement were taken on study days 1 and 8, and on day 8 for imatinib. RESULTS Of the 20 patients enrolled, six patients (30%) were CYP2D6 intermediate metabolizers (IMs), 13 (65%) extensive metabolizers (EMs), and the CYP2D6 status in one patient was unknown. In the presence of 400 mg twice daily imatinib, the mean metoprolol AUC was increased by 17% in IMs (from 1190 to 1390 ng ml(-1) h), and 24% in EMs (from 660 to 818 ng ml(-1) h). Patients classified as CYP2D6 IMs had an approximately 1.8-fold higher plasma metoprolol exposure than those classified as EMs. The oral clearance of imatinib was 11.0 +/- 2.0 l h(-1) and 11.8 +/- 4.1 l h(-1) for CYP2D6 IMs and EMs, respectively. CONCLUSIONS Co-administration of a high dose of imatinib resulted in a small or moderate increase in metoprolol plasma exposure in all patients regardless of CYP2D6 status. The clearance of imatinib showed no difference between CYP2D6 IMs and EMs.


The Journal of Clinical Pharmacology | 2011

Effects of rifampin and ketoconazole on the pharmacokinetics of nilotinib in healthy participants.

Chiaki Tanaka; Ophelia Q. P. Yin; Tom Smith; Venkat Sethuraman; Karen Grouss; Lawrence Galitz; Robert Harrell; Horst Schran

Nilotinib (Tasigna), an orally bioavailable second‐generation BCR‐ABL tyrosine kinase inhibitor, is approved for use in patients with chronic myeloid leukemia in chronic phase and accelerated phase who are resistant or intolerant to prior therapy, including imatinib. Previous in vitro studies indicated that nilotinib metabolism is primarily mediated by CYP3A4. To investigate the effect of CYP3A4 induction and inhibition on nilotinib pharmacokinetics, 2 studies were conducted in healthy volunteers prior to and following treatment with a strong inducer (rifampin) or inhibitor (ketoconazole). In the induction study, administration of rifampin 600 mg once daily for 8 days significantly increased urinary 6β‐hydroxycortisol/cortisol ratio, from a preinduction baseline of 5.8 ± 2.7 to 18.0 ± 10.2 after 8 days of rifampin treatment, confirming an inductive effect on CYP3A4. Nilotinib oral clearance was increased by 4.8‐fold, and the maximum serum concentration (Cmax) and area under the serum concentration‐time curve (AUC) were decreased by 64% and 80%, respectively, in the induced state compared with baseline. In the inhibition study, ketoconazole 400 mg once daily for 6 days increased the Cmax and AUC of nilotinib by 1.8‐ and 3‐fold, respectively, compared with nilotinib alone. These results indicate that concurrent use of strong CYP3A4 inducers or inhibitors may necessitate dosage adjustments of nilotinib and should be avoided when possible.


Clinical Therapeutics | 2009

Effects of hepatic impairment on the pharmacokinetics of nilotinib: An open-label, single-dose, parallel-group study

Ophelia Q. P. Yin; Neil Gallagher; Chiaki Tanaka; Deirdre Fisher; Venkat Sethuraman; Wei Zhou; Tsu-Han Lin; Douglas Heuman; Horst Schran

BACKGROUND Nilotinib is a second-generation BCR-ABL tyrosine kinase inhibitor approved for the treatment of patients who have imatinib-resistant Philadelphia chromosome-positive chronic myeloid leukemia in the chronic or accelerated phase or who are unable to tolerate imatinib. Nilotinib is metabolized in the liver via oxidation and hydroxylation pathways, mediated primarily by the cytochrome P450 3A4 isozyme. Interpatient variability in systemic exposure to nilotinib has been reported to range from 32% to 64%. OBJECTIVE This study compared the pharmacokinetics of nilotinib in subjects with hepatic impairment and subjects with normal hepatic function. METHODS Hepatic impairment was classified as mild (Child-Pugh grade A), moderate (Child-Pugh grade B), or severe (Child-Pugh grade C). Healthy control subjects were matched with hepatically impaired subjects by age (+/-10 years) and body weight (+/-20%). All subjects received a single oral dose of nilotinib 200 mg under fasted conditions, and serial blood samples were collected at specific times up to 120 hours after dosing. Serum nilotinib concentrations were measured using a validated LC-MS/MS assay with a lower limit of quantification of 2.5 ng/mL. The pharmacokinetic parameters analyzed were C(max), T(max), AUC(0-last), AUC(0-infinity), t(1/2), CL/F, and Vz/F. Tolerability assessments included adverse events (AEs), regular monitoring of clinical laboratory measures (eg, hematology, blood chemistry, urinalysis), physical examinations, vital signs, and ECGs. Each AE was evaluated in terms of its clinical significance, severity, duration, relation to study drug, and action taken. RESULTS The study enrolled 18 subjects with hepatic impairment (all male; age range, 47-67 years; weight range, 73.9-103.9 kg) and 9 healthy controls (all male; age range, 36-62 years; weight range, 73.3-109.5 kg). Among subjects with hepatic impairment, 6 had mild impairment, 6 moderate impairment, and 6 severe impairment. The nilotinib AUC(0-infinity) was a mean of 35%, 35%, and 19% higher in subjects with mild, moderate, and severe impairment, respectively, compared with healthy controls. The nilotinib CL/F was lower in all hepatic-impairment groups compared with healthy controls. The mean (SD) t(1/2) was 15.1 (4.97) and 16.0 (9.13) hours in the mild-impairment and control groups, respectively, but was 21.6 (7.77) and 32.4 (10.7) hours in the moderate- and severe-impairment groups, respectively, reflecting the decrease in CL/F and/or increase in Vz/F in the latter 2 groups. All AEs were mild or moderate, and the frequency of AEs was not associated with the degree of hepatic impairment. AEs included abdominal pain (1 subject with mild impairment), dyspepsia (2 with mild impairment), flatulence (1 with severe impairment), nausea (1 with mild impairment), urinary tract infection (1 with mild impairment), back pain (1 each with mild impairment and severe impairment, 1 control subject), and headache (1 each with mild impairment and severe impairment). CONCLUSIONS After a single 200-mg dose, nilotinib pharmacokinetics were modestly affected by hepatic impairment. The extent of change in nilotinib exposure in subjects with hepatic impairment was generally within the range of variability that has been observed clinically. The results of this study suggest that dose adjustment may not be necessary in patients with hepatic impairment. Nilotinib should be used with caution, and careful clinical monitoring is recommended in this population. ClinicalTrials.gov identifier: NCT00418626.


Journal of Biopharmaceutical Statistics | 2009

Balanced 2(n) factorial designs when observations are spatially correlated.

Venkat Sethuraman; Damaraju Raghavarao

In this article we focus on the optimal factorial and fractional-factorial designs when observations within blocks are correlated. The topic was motivated by a problem when the pharmaceutical experimenter needed to develop a controlled release, once-daily tablet formulation. Typically, in order to compare different formulations, trials are conducted in healthy human volunteers where each formulation is administered and bioavailability is estimated. Since each subject is administered more than one formulation, the observations within subjects are correlated. Balanced designs for 2 n factorial experiments when observations within blocks are spatially correlated, AR(1) with positive correlation (ρ > 0), are characterized. An explicit construction and analytical proof of balanced designs for both 2 n full and 2 n −1 fractional factorial experiments is provided. In order to illustrate the construction, two examples using a complete 2 3 factorial and a half replicate of 2 4 factorial experiment are provided. We consider the optimal or near-optimal designs provided by Cheng and Steinberg (1991), Martin et al. (1998c), and Elliott et al. (1999) as the starting point to obtain balanced designs. We compare the relative efficiencies of our balanced designs with these designs.


Journal of Biopharmaceutical Statistics | 2010

On Power and Sample Size Calculation in Ethnic Sensitivity Studies

Wei Zhang; Venkat Sethuraman

In ethnic sensitivity studies, it is of interest to know whether the same dose has the same effect over populations in different regions. Glasbrenner and Rosenkranz (2006) proposed a criterion for ethnic sensitivity studies in the context of different dose-exposure models. Their method is liberal in the sense that their sample size will not achieve the target power. We will show that the power function can be easily calculated by numeric integration, and the sample size can be determined by bisection.


Pharmaceutical Statistics | 2009

Impact of baseline ECG collection on the planning, analysis and interpretation of 'thorough' QT trials.

Venkat Sethuraman; Qiankun Sun


Pharmaceutical Statistics | 2007

Sample size calculation for the Power Model for dose proportionality studies

Venkat Sethuraman; Sergei L. Leonov; Lisa Squassante; Toni R. Mitchell; Michael Hale


Clinical Pharmacology & Therapeutics | 2010

Clinical Pharmacokinetics of the BCRABL Tyrosine Kinase Inhibitor Nilotinib

Chiaki Tanaka; Ophelia Q. P. Yin; Venkat Sethuraman; Tom Smith; Xin Wang; Karen Grouss; Hagop M. Kantarjian; Francis J. Giles; Oliver G. Ottmann; Lawrence Galitz; Horst Schran; Oqp Yin


Journal of Biopharmaceutical Statistics | 2010

Statistical consideration in testing for assay sensitivity in a "thorough" QT study.

Venkat Sethuraman; Shuang Wu; Jixian Wang

Collaboration


Dive into the Venkat Sethuraman's collaboration.

Researchain Logo
Decentralizing Knowledge