Network


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

Hotspot


Dive into the research topics where Umesh A. Shukla is active.

Publication


Featured researches published by Umesh A. Shukla.


Clinical Pharmacology & Therapeutics | 2002

The pharmacokinetics and pharmacodynamics of enoxaparin in obese volunteers

Ger‐Jan Sanderink; Aimé Le Liboux; Navin Jariwala; Neasa Harding; Marie‐Laure Ozoux; Umesh A. Shukla; Guy Montay; Bruno X Boutouyrie; Adelaida Miro

The objective of this study was to compare the pharmacokinetics of the low‐molecular‐weight heparin enoxaparin in obese and nonobese volunteers, by means of two administration regimens.


The Journal of Clinical Pharmacology | 1995

Nonlinear Pharmacokinetics of Nefazodone After Escalating Single and Multiple Oral Doses

Sanjeev Kaul; Umesh A. Shukla; Rashmi H. Barbhaiya

The single‐ and multiple‐dose pharmacokinetics of nefazodone and its metabolites, hydroxynefazodone, p‐hydroxynefazodone, and m‐chlorophenylpiperazine were investigated in two groups of 18 healthy male volunteers, employing three‐period complete crossover designs. In one group, single 50‐mg, 100‐mg, and 200‐mg oral doses of nefazodone hydrochloride were administered with a 1‐week washout between treatments. In the other group, doses of 50 mg, 100 mg, and 200 mg were administered twice a day (every 12 hours) for 7.5 days (15 doses) with a 1‐week washout between treatments. Serial plasma samples were obtained in both groups and assayed for nefazodone, hydroxynefazodone, m‐chlorophenylpiperazine, and p‐hydroxynefazodone. Cmax plasma levels of nefazodone and hydroxynefazodone were attained within 2 hours of administration of nefazodone; tmax for m‐chlorophenylpiperazine was more delayed, and p‐hydroxynefazodone levels were generally below the assay limit. On repeated twice‐daily dosing of nefazodone, steady‐state levels of the drug and its metabolites were reached within 3 days. Mean single‐dose plasma half‐life (t1/2) values for nefazodone increased from ∼1 hour at a 50‐mg dose to ∼2 hours at a 200‐mg dose; at steady state, t1/2 values increased from ∼2 hours at 50 mg twice daily to ∼3.7 hours at 200 mg twice daily. Whereas dose increased in the proportion of 1:2:4, mean single‐dose AUC0‐∞ for nefazodone increased in the proportion of 1:3.3:8.9 and mean steady‐state AUC0‐r for nefazodone increased in the proportion of 1:4.2:16.8. Plasma levels of hydroxynefazodone paralleled those of nefazodone and were approximately 33% of nefazodone levels at each dose level. Plasma levels of m‐chlorophenylpiperazine were only ∼10% those of nefazodone. Within the dosage range of 50–200 mg of nefazodone hydrochloride, nefazodone and hydroxynefazodone exhibited nonlinear pharmacokinetics; m‐chlorophenylpiperazine, a minor metabolite, appeared to exhibit linear pharmacokinetics.


Antimicrobial Agents and Chemotherapy | 1990

Comparison of cefprozil and cefaclor pharmacokinetics and tissue penetration.

Rashmi H. Barbhaiya; Umesh A. Shukla; C R Gleason; Wen Chyi Shyu; Richard Wilber; Kenneth A. Pittman

The pharmacokinetics and tissue penetration, as judged by skin blister fluid, of cefprozil and cefaclor were examined in 12 healthy male volunteers. Doses of 250 and 500 mg of each drug were given to fasting subjects in a crossover fashion. Serially obtained plasma, skin blister fluid, and urine samples were analyzed for cefprozil or cefaclor by validated high-pressure liquid chromatographic methods. After oral administration of 250 and 500 mg of cefprozil, mean concentrations in plasma rose to peak levels (Cmax) of 6.1 and 11.2 micrograms/ml, respectively, and those of cefaclor were 10.6 and 17.3 micrograms/ml, respectively. The elimination half-life of cefprozil (1.3 h) was significantly longer than that of cefaclor (0.6 h), and as a result, the area under the curve for cefprozil was about two times greater than that for cefaclor. Both cephalosporins were primarily excreted unchanged in urine. The mean skin blister Cmax values were 3.0 and 5.8 micrograms/ml for cefprozil and 3.6 and 6.5 micrograms/ml for cefaclor after the 250- and 500-mg oral doses, respectively. The mean Cmax values in skin blister fluid for both cephalosporins were comparable and were significantly lower than the corresponding Cmax values in plasma. However, the levels of cefprozil and cefaclor in skin blister fluid declined more slowly than they did in plasma. The skin blister fluid half-life estimates for cefprozil were significantly longer than they were for cefaclor. Parallel to the observation in plasma, the mean skin blister fluid areas under the curve for cefprozil were significantly higher than they were for cefaclor. The plasma and skin blister fluid pharmacokinetic analyses suggest that the exposure of humans to cefprozil is significantly greater than that to cefaclor at the same dose.


European Journal of Clinical Pharmacology | 1995

Single-dose pharmacokinetics of nefazodone in healthy young and elderly subjects and in subjects with renal or hepatic impairment.

Rashmi H. Barbhaiya; Douglas S. Greene; Umesh A. Shukla

The single-dose pharmacokinetics of nefazodone (NEF) and its metabolites hydroxynefazodone (HO-NEF) and m-chlorophenylpiperazine (mCPP) were examined in 12 healthy younger subjects ≤55 years of age (YNG), 12 elderly subjects ≥65 years of age (ELD), 12 patients with biopsy proven hepatic cirrhosis (HEP) and 12 patients with moderate renal impairment (REN), ClCR 20–60 ml·min−1. The study was of parallel group design, with each of the four subject groups receiving escalating single oral doses of 50, 100 and 200 mg of nefazodone at 1 week intervals. Serial blood samples for pharmacokinetic analysis were collected for 48 h following each dose and plasma samples were assayed for NEF, HO-NEF and mCPP by a validated HPLC method.Single oral doses up to 200 mg of nefazodone were well tolerated by all subjects. Maximum plasma levels of NEF and HO-NEF were generally attained within 1 h after administration of nefazodone. HO-NEF and mCPP plasma levels were about 1/3 and <1/10 those of NEF, respectively. There were no apparent gender-related pharmacokinetic differences in any group of subjects. NEF and HO-NEF pharmacokinetics were dose dependent in all four subject groups; a superproportional increase in AUC and an increase in t1/2 with increasing dose was obtained, indicative of nonlinear pharmacokinetics. Relative to normal subjects, elderly and cirrhotic subjects exhibited increased systemic exposure to NEF and HO-NEF, as reflected by AUC, at all doses of nefazodone; subjects with moderate renal impairment did not.Elderly and cirrhotic patients may require lower doses of NEF to achieve and maintain therapeutic effectiveness.


The Journal of Clinical Pharmacology | 2006

Pharmacokinetics of otamixaban, a direct factor Xa inhibitor, in healthy male subjects: pharmacokinetic model development for phase 2/3 simulation of exposure.

Anne Paccaly; Annke Frick; Shashank Rohatagi; Jingli Liu; Umesh A. Shukla; Ronald Rosenburg; Markus Hinder; Bradford K. Jensen

The pharmacokinetics of otamixaban was investigated in healthy male subjects over a wide range of intravenous doses, with duration of administration varying between 1‐minute infusions (bolus dose) and 24‐hour infusions, using noncompartmental and multicompartmental methods. A global compartmental analysis (2 and 3 compartments) generated a single set of pharmacokinetic parameters, regardless of infusion rate and duration, and took into account the 30% decrease in clearance and volume of distribution observed over the dose range. The 2‐compartment model was retained to predict bolus plus 3‐hour‐infusion doses of otamixaban for future phase 2/3 studies. Otamixaban exhibited in healthy subjects several interesting pharmacokinetic features in view of its potential therapeutic use in coronary thrombosis: a rapid plasma distribution and elimination, a well‐described dose‐exposure relationship, a low intersubject variability in plasma exposure, and a mixed renal and biliary excretion with constant renal clearance.


Journal of Clinical Psychopharmacology | 1995

Coadministration of nefazodone and benzodiazepines: I. Pharmacodynamic assessment.

Patricia D. Kroboth; M. Margaret Folan; Richard M. Lush; Phillip C. Chaikin; Umesh A. Shukla; Rashmi H. Barbhaiya; Daniel E. Salazar

One hundred two healthy men were evaluated in one of three studies conducted to evaluate the coadministration of nefazodone, 200 mg twice daily, and three benzodiazepines: triazolam, 0.25 mg; alprazolam, 1 mg twice daily; or lorazepam, 2 mg twice daily. In the first study, psychomotor performance, memory, and sedation were assessed at 0, 0.5, 1.5, 2.5, and 9 hours after single doses of triazolam alone and again after 7 days of nefazodone. Data from 6 of 12 subjects in this study were evaluable because of a dosing error in the other 6 subjects. In the subsequent two parallel design studies, groups of 12 volunteers received 7 days of either placebo; nefazodone, 200 mg; alprazolam, 1 mg twice daily; or alprazolam plus nefazodone or, in the second study, either placebo; nefazodone; lorazepam, 2 mg twice daily; or lorazepam plus nefazodone; the studies were identical, double-dummy, double-blind designs. Psychomotor performance, memory, and sedation were assessed at 0, 1, 3, and 8 hours after the 8 a.m. dose on days 1, 3, 5, and 7 of the studies. In all studies, blood samples were also obtained at testing times so that effect/concentration comparisons could be made and so full pharmacokinetic analyses could be done for separate studies. Nefazodone had no effect on psychomotor performance, memory, or sedation relative to placebo in any study. The mean maximum observed effect (MaxOE) on psychomotor performance and sedation were increased when triazolam was given after 7 days of nefazodone (p < 0.05); also, triazolam concentration was 60% higher at this time. Alprazolam and lorazepam impaired performance on day 1 (mean MaxOE, 34 and 30%, respectively) relative to placebo and nefazodone. By day 7 of alprazolam or lorazepam, psychomotor impairment decreased, indicating the development of tolerance. Alprazolam plus nefazodone increased psychomotor impairment (MaxOE, approximately 50%) and sedation relative to alprazolam alone on days 3, 5, and 7 (p < 0.05). Higher alprazolam concentrations explained the increased impairment in the alprazolam plus nefazodone treatment group; however, it is also possible that there was a delay in the development of tolerance. There were no differences in psychomotor impairment, memory, sedation, or lorazepam concentration detected between the lorazepam alone and lorazepam plus nefazodone treatments. This is consistent with the absence of a pharmacokinetic interaction between nefazodone and lorazepam. These results indicate that if the coadministration of a benzodiazepine is required in patients receiving nefazodone therapy, clinically significant interactions would be less likely with those eliminated by conjugative metabolism such as lorazepam. In cases where a benzodiazepine eliminated by oxidative metabolism is required, a reduction in initial dosage and careful clinical evaluation for signs of psychomotor impairment may be appropriate.


Thrombosis and Haemostasis | 2005

Pharmacodynamic markers in the early clinical assessment of otamixaban, a direct factor Xa inhibitor

Anne Paccaly; Marie‐Laure Ozoux; Valeria Chu; Kelly Simcox; Vanessa Marks; Geneviève Freyburger; Michel Sibille; Umesh A. Shukla

This manuscript reports the assessment of pharmacodynamic (PD) markers of anti-coagulation in the first-in-man study with the novel direct Factor Xa (FXa) inhibitor, otamixaban, with a brief description of safety and pharmacokinetic (PK) findings. The study comprised ten consecutive parallel groups of healthy male subjects (6 active, 2 placebo per group). Eight groups received escalating intravenous doses of otamixaban as 6-hour infusions (1.7 to 183 microg/kg/h) and two groups received a bolus dose (30 or 120 microg/kg) with a 6-hour infusion (60 or 140 microg/ kg/h, respectively). PD markers included anti-FXa activity and clotting time measurements, i.e. activated Thromboplastin Time (aPTT), Prothrombin Time (PT), Heptest Clotting Time (HCT), and Russells Viper Venom-induced clotting Time (RVVT). In addition, Endogenous Thrombin Potential (ETP) was assessed in the bolus-plus-infusion dose groups. Otamixaban was well tolerated. Otamixaban plasma concentrations increased with escalating dose, were maximal at the end-of-infusion (C(eoi)), and decreased rapidly as the infusion was stopped. Anti-FXa activity coincided with otamixaban plasma concentrations and clotting time measurements followed the same pattern. Maximal changes from baseline at C(eoi) were 1.9 +/- 0.2 for aPTT, 2.0 +/- 0.2 for PT, 5.1 +/- 0.6 for HCT, and 4.5 +/- 1.2 for RVVT. Otamixaban inhibited thrombin generation (24% decrease in ETP) and a delay in thrombin generation was noticed in vitro at high concentrations.


The Journal of Clinical Pharmacology | 1992

Population Pharmacokinetic Analysis of Didanosine (2′,3′‐Dideoxyinosine) Plasma Concentrations Obtained in Phase I Clinical Trials in Patients with AIDS or AIDS‐Related Complex

Sudhakar M. Pai; Umesh A. Shukla; Thaddeus H. Grasela; Catherine A. Knupp; Raphael Dolin; Fred T. Valentine; Colin McLaren; Howard A. Liebman; R. Russell Martin; Kenneth A. Pittman; Rashmi H. Barbhaiya

Plasma didanosine concentration data from 36 patients receiving once‐a‐day therapy and from 33 patients receiving twice‐a‐day therapy were subject to population pharmacokinetic analysis with the computer program NONMEM. Once‐ or twice‐a‐day regimens of didanosine were administered intravenously (IV) (dose: 0.8–33 mg/kg) during the first 2 weeks of therapy, and orally (dose: 1.6–66 mg/kg) for the remaining 4 weeks of therapy. Plasma pharmacokinetics were determined after the first and last (steady‐state) IV and oral doses. Population pharmacokinetic parameters for the combined IV and oral steady‐state data were (mean [%CV]): systemic clearance, CL, 0.70 (5.2) L/h/kg; central compartment volume, Vc, 0.18 (32) L/kg; steady‐state distribution volume, Vdss, 0.84 (6.8) L/kg; first‐order absorption rate constant, Ka, 1.3 (9.5) hr−1; and bioavailable fraction, F, 0.34 (8.5). Interindividual variability (omega) was (%CV) 22.3 and 71.0 for CL and Vc, respectively. Intraindividual (residual) variability (sigma) in plasma concentrations (%CV) was 50.2. Body weight, sex, and age did not account for the variability in either CL or Vc, and the use of alternate pharmacokinetic models did not reduce the value of intraindividual variability. Population parameters for the combined IV and oral first‐dose data were generally similar to those for the steady‐state data. The parameters can be used to design dosing regimens in patients using the Bayesian feedback approach.


European Journal of Clinical Pharmacology | 1994

Disposition kinetics of buspirone in patients with renal or hepatic impairment after administration of single and multiple doses

Rashmi H. Barbhaiya; Umesh A. Shukla; Marc A. Pfeffer; Kenneth A. Pittman; R. Shrotriya; C. Laroudie; Richard E. Gammans

AbstractThe single dose and steady-state pharmacokinetics of buspirone and its metabolite 1-pyrimidinyl piperazine (1-PP) have been evaluated in normal volunteers and patients with renal or hepatic impairment, using a parallel group design, with assignment of patients to study group on the basis of the degree of renal (mild, moderate, severe) or hepatic (compensated or decompensated) impairment. Each healthy volunteer or patient received a single dose of 10 mg buspirone on Day 1 of the study, and starting 36 h after the first dose, healthy volunteers and patients received 10 mg doses of buspirone every 12 hours for 9 days. On the morning of Day 10 they received the last dose. Serial blood samples were collected on Days 1, 5 and 10 and plasma was analysed for buspirone and 1-PP. The plasma concentrations of buspirone and 1-PP were highly variable regardless of the renal or hepatic function. The peak concentrations (Cmax) and area under the curves (AUC) of buspirone and 1-PP on Days D 5 and 10 were higher than on Day D 1. The trough levels (Cmin) and AUCs (D 5 and 10) of buspirone and 1-PP indicated, that, regardless of renal or hepatic function, steady state was reached after 3 to 5 days of dosing. At steady-state, patients with renal or hepatic impairment had significantly higher Cmax and AUC values of buspirone than in normal volunteers. However, the intensity and frequency of adverse experiences in patients with renal or hepatic impairment were not significantly different from those observed in normal volunteers. There was no correlation between the average plasma concentrations of buspirone (


Antimicrobial Agents and Chemotherapy | 1990

Comparison of the effects of food on the pharmacokinetics of cefprozil and cefaclor.

Rashmi H. Barbhaiya; Umesh A. Shukla; Carol Gleason; Wen Chyi Shyu; Kenneth A. Pittman

Collaboration


Dive into the Umesh A. Shukla's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. Russell Martin

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge