Network


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

Hotspot


Dive into the research topics where Avraham Yacobi is active.

Publication


Featured researches published by Avraham Yacobi.


Pharmaceutical Research | 2000

Bioanalytical Method Validation—A Revisit with a Decade of Progress

Vinod P. Shah; Kamal K. Midha; John W. A. Findlay; Howard M. Hill; James D. Hulse; Iain J. McGilveray; G. McKay; Krys J. Miller; Rabindra Patnaik; Mark L. Powell; Alfred P. Tonelli; C. T. Viswanathan; Avraham Yacobi

This report is a synthesis of (1) the earlier conference on Analytical Methods Validation−Bioavailability, Bioequivalence and Pharmacokinetic Studies (Conference held in Arlington, VA, December 3–5, 1990 and the report published in Pharmaceutical Research, 9: 588-592, 1992) and (2) the workshop on “Bioanalytical Methods Validation—A Revisit with a Decade of Progress,” (Workshop held in Arlington, VA, January 12–14, 2000), sponsored by the American Association of Pharmaceutical Scientists and the U. S. Food and Drug Administration. The bioanalytical method validation workshop of January 12–14, 2000 was directed towards small molecules. A separate workshop was held in March 1–3, 2000 to discuss validation principles for macromolecules. The purpose of this report is to represent the progress in analytical methodologies over the last decade and assessment of the major agreements and issues discussed with regard to small molecules at both the conference and the workshop. The report is also intended to provide guiding principles for validation of bioanalytical methods employed in support of bioavailability, bioequivalence, and pharmacokinetic studies in man and in animals.


Clinical Pharmacology & Therapeutics | 1983

Kinetics of esmolol, an ultra-short-acting beta blocker, and of its major metabolite

Check Y. Sum; Avraham Yacobi; Ronald Kartzinel; Herman F. Stampfli; Charles S Davis; Chii-Ming Lai

Esmolol is an ultra‐short‐acting beta blocker. Its kinetics was studied in eight healthy subjects after continuous intravenous infusion of 400 μg/kg/min over 2 hr. The concentrations of esmolol and its major metabolite, 3‐[4‐(2‐hydroxy‐3‐{isopropylamino}propoxy)phenyl]propionic acid, in blood and urine were determined by gas chromatographic‐mass spectrometric assay and HPLC. The distribution and elimination t½s of esmolol averaged 2.03 and 9.19 min. The apparent volume of distribution of esmolol averaged 3.43 l/kg and was four times the volume of the central compartment. The total clearance of esmolol averaged 285 ml/min/kg, indicating that nonhepatic routes play a predominant role in its clearance. The t½s of formation and elimination of the metabolite averaged 2.82 min and 3.72 hr. The ratio of the metabolite formation and elimination rate constants of the parent drug (kf/k10) averaged 0.829, suggesting that 82.9% of esmolol was converted to the metabolite (which is consistent with the urinary recovery of 71% of the dose as unconjugated metabolite). The volume of distribution and total clearance of the metabolite averaged 0.411 l/kg and 1.28 ml/min/kg. Esmolol was followed by a significant reduction of isoproterenol‐induced increase in heart rate and systolic blood pressure at doses of 50, 150, and 400 μg/kg/min. There was a strong correlation between the magnitude of these effects and the logarithm of the steady‐state blood concentrations of esmolol.


International Journal of Pharmaceutics | 1992

Analytical methods validation: Bioavailability, bioequivalence and pharmacokinetic studies: Sponsored by the American Association of Pharmaceutical Chemists, U.S. Food and Drug Administration, Fédération Internationale Pharmaceutique, Health Protection Branch (Canada) and Association of Official Analytical Chemists

Vinod P. Shah; Kamal K. Midha; Shrikant Dighe; Iain J. McGilveray; Jerome P. Skelly; Avraham Yacobi; Thomas Layloff; C. T. Viswanathan; C. Edgar Cook; R. D. McDowall; Kenneth A. Pittman; Sidney Spector; Kenneth S. Albert; Sanford Bolton; Michael R. Dobrinska; William H. Doub; Michael Eichelbaum; John W. A. Findlay; Keith Gallicano; William Garland; Dwight J. Hardy; James D. Hulse; H. Thomas Karnes; Ron Lange; William D. Mason; G. McKay; Eric Ormsby; James Overpeck; H.D. Plattenberg; Gerald K. Shiu

Abstract This is a summary report of the conference on ‘Analytical Methods Validation: Bioavailability, Bioequivalence and Pharmacokinetic Studies.’ The conference was held from December 3 to 5, 1990, in the Washington, DC area and was sponsored by the American Association of Pharmaceutical Scientists, U.S. Food and Drug Administration, Federation Internationale Pharmaceutique, Health Protection Branch (Canada) and Association of Official Analytical Chemists. The purpose of the report is to represent our assessment of the major agreements and issues discussed at the conference. This report is also intended to provide guiding principles for validation of analytical methods employed in bioavailability, bioequivalence and pharmacokinetic studies in man and animals. The objectives of the conference were: (1) to reach a consensus on what should be required in analytical methods validation and the procedures to establish validation; (2) to determine processes of application of the validation procedures in the bioavailability, bioequivalence and pharmacokinetic studies; and (3) to develop a report on analytical methods validation (which may be referred to in developing future formal guidelines). Acceptable standards for documenting and validating analytical methods with regard to processes, parameters or data treatments were discussed because of their importance in assessment of pharmacokinetic. bioavailability, and bioequivalence studies. Other topics which were considered essential in the conduct of pharmacokinetic studies or in establishing bioequivalency criteria, including measurement of drug metabolites and stereoselectivc determinations, were also deliberated.


The Journal of Clinical Pharmacology | 1982

Pharmacokinetics of Hydroxyethyl Starch in Normal Subjects

Avraham Yacobi; Roger G. Stoll; Check Y. Sum; Chii-Ming Lai; Surinder D. Gupta; James D. Hulse

Abstract: To determine the elimination of high‐molecular‐weight hydroxyethyl starch (HES, Mw 450,000) in normal subjects, ten volunteers were given 500 ml 6% HES solution by intravenous infusion, and serial blood and urine samples were collected for nonglucose total carbohydrate determination. On the average, 46 and 64 per cent of the dose was excreted in the urine within two and eight days, respectively. The plasma concentration declined rapidly during the first week after infusion. The average terminal half‐life was 17 days during the first 42 days, which accounted for elimination of about 90 per cent of the dose. The remainder was eliminated with a terminal half‐life of 48 days determined between days 42 and 83 of the study. As expected, the infusion of HES resulted in plasma volume expansion over a 48‐hour period during which time levels of nonglucose carbohydrates were above 3.5 mg/ml. HES is metabolized by α‐amylase in the body. During the first 48 hours after infusion of HES, plasma α‐amylase activity was significantly increased over control. Concomitantly, α‐amylase activity in urine was also elevated but not significantly so.


The Journal of Clinical Pharmacology | 1994

Opportunities for Integration of Pharmacokinetics, Pharmacodynamics, and Toxicokinetics in Rational Drug Development

Carl C. Peck; William H. Barr; Leslie Z. Benet; Jerry M. Collins; Robert E. Desjardins; Daniel E. Furst; John G. Harter; Gerhard Levy; Thomas M. Ludden; John H. Rodman; Lilly Sanathanan; Jerome J. Schentag; Vinod P. Shah; Lewis B. Sheiner; Jerome P. Skelly; Donald R. Stanski; Robert Temple; C. T. Viswanathan; Judi Weissinger; Avraham Yacobi

Carl C. Peck, MD, William H. Barr, PharmD, PhD, Leslie Z. Benet, PhD, Jerry Collins, PhD, Robert E. Desjardins, MD, Daniel E. Furst, MD, John G. Harter, MD, Gerhard Levy, PharmD, Thomas Ludden, PhD, John H. Rodman, PharmD, Lilly Sanathanan, PhD, Jerome J. Schentag, Pharmfl, Vinod P. Shah, PhD, Lewis B. Sheiner, MD, Jerome P. Skelly, PhD, Donald R. Stanski, MD, Robert J. Temple, MD, C. T. Viswanathan, PhD, Judi Weissinger, PhD, and Avraham Yacobi, PhD


Annals of Pharmacotherapy | 1983

Hetastarch: An Overview of the Colloid and its Metabolism:

James D. Hulse; Avraham Yacobi

Hetastarch, ethoxylated amylopectin, has found clinical utility as a plasma volume expansion agent, a sedimenting agent during pheresis, and a pump priming fluid. Hetastarch is a complex mixture of derivatized amylopectin molecules of various molecular sizes. The derivatization causes resistance to enzymatic hydrolysis, therefore, allowing hetastarch sufficient vascular residence time to be an effective vascular osmotic agent. This has led to its use as a volume expander and to its consequent use as a pump priming fluid. The metabolism of hetastarch proceeds through α-amylase hydrolysis of glycosidic bonds, yielding molecules small enough for renal clearance, but does not result in complete hydrolysis. Hence, glucose is not a significant product of hetastarch metabolism. Metabolism proceeds at such a rate that volume expansion is seen for 24–36 hours with a maximum effect (100–172 percent of the infused volume) occurring shortly after infusion. Ninety percent of the dose is eliminated with a half-life of about 17 days.


Drug Metabolism Reviews | 1986

Pharmacokinetics of Mitoxantrone in Man and Laboratory Animals

Vijay K. Batra; John A. Morrison; David L. Woodward; Nancy S. Siverd; Avraham Yacobi

Mitoxantrone (NOVANTRONE), an anthracenedione, is a novel anticancer agent with a wide spectrum of antitumor activity. Its anticancer activity is comparable to that of doxorubicin but with apparently significantly reduced cardiotoxicity. The recommended dosage regimen for the treatment of breast carcinoma is 12-14 mg/m2 given intravenously once every 21 days. Intravenously administered mitoxantrone disappears from the plasma of man and laboratory animals with multiexponential kinetics and with the terminal half-life ranging from 38 h to several days. It is rapidly cleared from the plasma by extensive sequestration into the tissues of the rat, dog, monkey, and man. However, redistribution back into the plasma and elimination from the body are slow processes. In both animals and man it is metabolized to the mono- and dicarboxylic acid derivatives, as well as glucuronide conjugates of these acids. Following intravenous administration, it is unchanged mitoxantrone that binds to most tissues. Rats, dogs, monkeys, and man, all eliminate mitoxantrone and its metabolites slowly by both renal and biliary excretion, with the biliary route predominating.


Pediatric Infectious Disease Journal | 1987

Pharmacokinetic profile of cefixime in man

Robert D. Faulkner; Avraham Yacobi; Joseph S. Barone; Stanley A. Kaplan; B. Michael Silber

Cefixime is an orally absorbed third generation cephalosporin with a broad spectrum of activity against Gram-positive and Gram-negative bacteria and is highly resistant to beta-lactamase degradation. In general serum and urinary concentrations of cefixime achieved after recommended daily doses are well above the minimal inhibitory concentrations at 90% for indicated microorganisms. The pharmacokinetics of cefixime were determined in adult and pediatric subjects. In general the half-life of the drug is about 3 to 4 hours and is not dependent on dose. The drug is not extensively bound to serum proteins; the free fraction is about 31% and is concentration-independent. The absolute bioavailability, based on comparisons of area under the serum concentration-time curve values after 200-mg intravenous, 200-mg oral solution, and 200- and 400-mg capsule doses, ranged from 40 to 52%, showing a comparable bioavailability for cefixime at single 200- and 400-mg oral doses. In a dose proportionality study, over an oral dose range of 200 to 2000 mg, peak serum concentration (Cmax) and area under the concentration-time values increased linearly but were not directly proportional with dose. Upon multiple dosing for 2 weeks on a 400-mg daily or 200-mg twice a day regimen, serum concentrations and urinary recovery of unchanged drug were similar for each group, and there was no drug accumulation. Peak serum concentration and area under the concentration-time curve values after a 400-mg dose were almost double those after a 200-mg dose. All formulations of cefixime were bioequivalent to an oral reference solution at doses of 200 and 400 mg.(ABSTRACT TRUNCATED AT 250 WORDS)


Archive | 1993

Integration of pharmacokinetics, pharmacodynamics, and toxicokinetics in rational drug development

Avraham Yacobi; Jerome P. Skelly; Vinod P. Shah; Leslie Z. Benet

Twenty-five papers from a conference on [title] held April 1991, in Arlington, Virginia, were edited and updated for presentation in this proceedings volume. Contributions on the topics announced in the title are from academic, governmental, and industrial scientists aiming to identify the roles and


Xenobiotica | 1987

Deposition of viprostol (a synthetic PGE2 vasodilator) in the skin following topical administration to laboratory animals

Gabriela Nicolau; R. A. Baughman; A. Tonelli; W. McWilliams; J. Schiltz; Avraham Yacobi

1. Topical application of 14C-viprostol, a synthetic prostaglandin E2 analogue, to laboratory animals resulted in a significant depot of radioactivity in the skin at the application site in all species studied: mouse, rat, guinea pig, rabbit and monkey, with longer residence times in the larger species. 2. The location of the 14C-label in the skin in mice and monkeys was determined by microscopic autoradiography. Evaluation of the autoradiograms show rapid penetration of the drug into the skin via the hair follicles. 3. In mouse distribution of radioactivity was evident in the stratum corneum and down the hair shafts by 30 min. after dosing. By 2 h radioactivity was also observed throughout the viable epidermis; in the dermis only the hair shafts contained significant radioactivity. At 72 h after dose removal, radioactivity was evident only in the hair follicles and hair shaft. 4. In monkey the residence time of radioactivity in the skin was significantly longer than in mouse, but the general distribution pattern was similar in both species. 5. The presence of viprostol in the hair follicles and epidermal layer after topical administration is consistent with its extensive skin metabolism previously reported.

Collaboration


Dive into the Avraham Yacobi's collaboration.

Top Co-Authors

Avatar

Vinod P. Shah

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel A. Moros

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Barrie Levitt

New York Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William H. Barr

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge