Network


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

Hotspot


Dive into the research topics where Punit Marathe is active.

Publication


Featured researches published by Punit Marathe.


Journal of Pharmaceutical Sciences | 2011

PHRMA CPCDC initiative on predictive models of human pharmacokinetics, part 5: Prediction of plasma concentration–time profiles in human by using the physiologically‐based pharmacokinetic modeling approach

Patrick Poulin; Rhys D.O. Jones; Hannah M. Jones; Christopher R. Gibson; Malcolm Rowland; Jenny Y. Chien; Barbara J. Ring; Kimberly K. Adkison; M. Sherry Ku; Handan He; Ragini Vuppugalla; Punit Marathe; Volker Fischer; Sandeep Dutta; Vikash Sinha; Thorir Björnsson; Thierry Lavé; James W.T. Yates

The objective of this study is to assess the effectiveness of physiologically based pharmacokinetic (PBPK) models for simulating human plasma concentration-time profiles for the unique drug dataset of blinded data that has been assembled as part of a Pharmaceutical Research and Manufacturers of America initiative. Combinations of absorption, distribution, and clearance models were tested with a PBPK approach that has been developed from published equations. An assessment of the quality of the model predictions was made on the basis of the shape of the plasma time courses and related parameters. Up to 69% of the simulations of plasma time courses made in human demonstrated a medium to high degree of accuracy for intravenous pharmacokinetics, whereas this number decreased to 23% after oral administration based on the selected criteria. The simulations resulted in a general underestimation of drug exposure (Cmax and AUC0- t ). The explanations for this underestimation are diverse. Therefore, in general it may be due to underprediction of absorption parameters and/or overprediction of distribution or oral first-pass. The implications of compound properties are demonstrated. The PBPK approach based on in vitro-input data was as accurate as the approach based on in vivo data. Overall, the scientific benefit of this modeling study was to obtain more extensive characterization of predictions of human PK from PBPK methods.


Journal of Pharmaceutical Sciences | 2011

PhRMA CPCDC initiative on predictive models of human pharmacokinetics, part 3: Comparative assessement of prediction methods of human clearance

Barbara J. Ring; Jenny Y. Chien; Kimberly K. Adkison; Hannah M. Jones; Malcolm Rowland; Rhys D.O. Jones; James W.T. Yates; M. Sherry Ku; Christopher R. Gibson; Handan He; Ragini Vuppugalla; Punit Marathe; Volker Fischer; Sandeep Dutta; Vikash Sinha; Thorir Björnsson; Thierry Lavé; Patrick Poulin

The objective of this study was to evaluate the performance of various allometric and in vitro-in vivo extrapolation (IVIVE) methodologies with and without plasma protein binding corrections for the prediction of human intravenous (i.v.) clearance (CL). The objective was also to evaluate the IVIVE prediction methods with animal data. Methodologies were selected from the literature. Pharmaceutical Research and Manufacturers of America member companies contributed blinded datasets from preclinical and clinical studies for 108 compounds, among which 19 drugs had i.v. clinical pharmacokinetics data and were used in the analysis. In vivo and in vitro preclinical data were used to predict CL by 29 different methods. For many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. In addition, 66 methods of predicting oral (p.o.) area under the curve (AUCp.o. ) were evaluated for 107 compounds using rational combinations of i.v. CL and bioavailability (F), and direct scaling of observed p.o. CL from preclinical species. Various statistical and outlier techniques were employed to assess the predictability of each method. Across methods, the maximum success rate in predicting human CL for the 19 drugs was 100%, 94%, and 78% of the compounds with predictions falling within 10-fold, threefold, and twofold error, respectively, of the observed CL. In general, in vivo methods performed slightly better than IVIVE methods (at least in terms of measures of correlation and global concordance), with the fu intercept method and two-species-based allometry (rat-dog) being the best performing methods. IVIVE methods using microsomes (incorporating both plasma and microsomal binding) and hepatocytes (not incorporating binding) resulted in 75% and 78%, respectively, of the predictions falling within twofold error. IVIVE methods using other combinations of binding assumptions were much less accurate. The results for prediction of AUCp.o. were consistent with i.v. CL. However, the greatest challenge to successful prediction of human p.o. CL is the estimate of F in human. Overall, the results of this initiative confirmed predictive performance of common methodologies used to predict human CL.


Journal of Medicinal Chemistry | 2008

Discovery of Brivanib Alaninate ((S)-((R)-1-(4-(4-Fluoro-2-methyl-1H-indol-5-yloxy)-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate), A Novel Prodrug of Dual Vascular Endothelial Growth Factor Receptor-2 and Fibroblast Growth Factor Receptor-1 Kinase Inhibitor (BMS-540215)

Zhen-Wei Cai; Yongzheng Zhang; Robert M. Borzilleri; Ligang Qian; Stephanie Barbosa; Donna D. Wei; Xiaoping Zheng; Lawrence Wu; Junying Fan; Zhongping Shi; Barri Wautlet; Steve Mortillo; Robert Jeyaseelan; Daniel W. Kukral; Amrita Kamath; Punit Marathe; Celia D’Arienzo; George Derbin; Joel C. Barrish; Jeffrey A. Robl; John T. Hunt; Louis J. Lombardo; Joseph Fargnoli; Rajeev S. Bhide

A series of amino acid ester prodrugs of the dual VEGFR-2/FGFR-1 kinase inhibitor 1 (BMS-540215) was prepared in an effort to improve the aqueous solubility and oral bioavailability of the parent compound. These prodrugs were evaluated for their ability to liberate parent drug 1 in in vitro and in vivo systems. The l-alanine prodrug 8 (also known as brivanib alaninate/BMS-582664) was selected as a development candidate and is presently in phase II clinical trials.


Journal of Medicinal Chemistry | 2008

Discovery of Pyrrolopyridine-Pyridone Based Inhibitors of Met Kinase : Synthesis, X-ray Crystallographic Analysis, and Biological Activities

Kyoung S. Kim; Liping Zhang; Robert J. Schmidt; Zhen-Wei Cai; Donna D. Wei; David K. Williams; Louis J. Lombardo; George L. Trainor; Dianlin Xie; Yaquan Zhang; Yongmi An; John S. Sack; John S. Tokarski; Celia D'Arienzo; Amrita Kamath; Punit Marathe; Yueping Zhang; Jonathan Lippy; Robert Jeyaseelan; Barri Wautlet; Benjamin Henley; Johnni Gullo-Brown; Veeraswamy Manne; John T. Hunt; Joseph Fargnoli; Robert M. Borzilleri

Conformationally constrained 2-pyridone analogue 2 is a potent Met kinase inhibitor with an IC50 value of 1.8 nM. Further SAR of the 2-pyridone based inhibitors of Met kinase led to potent 4-pyridone and pyridine N-oxide inhibitors such as 3 and 4. The X-ray crystallographic data of the inhibitor 2 bound to the ATP binding site of Met kinase protein provided insight into the binding modes of these inhibitors, and the SAR of this series of analogues was rationalized. Many of these analogues showed potent antiproliferative activities against the Met dependent GTL-16 gastric carcinoma cell line. Compound 2 also inhibited Flt-3 and VEGFR-2 kinases with IC50 values of 4 and 27 nM, respectively. It possesses a favorable pharmacokinetic profile in mice and demonstrates significant in vivo antitumor activity in the GTL-16 human gastric carcinoma xenograft model.


Clinical Pharmacokinectics | 2005

Steady-State Pharmacokinetics of a Novel Extended-Release Metformin Formulation

Peter Timmins; Steve Donahue; Jeff Meeker; Punit Marathe

AbstractBackground and objective: Metformin is an effective treatment for type 2 diabetes mellitus. The pharmacokinetic characteristics of the conventional immediate-release (IR) formulation of metformin (Glucophage®), however, necessitate two- or three-times-daily dosing. Development of a novel extended-release (XR) formulation of metformin (Glucophage® XR) using GelShield Diffusion System technology provides a once-daily dosing option. The objective of this study was to assess the steady-state pharmacokinetics of metformin XR tablets. Study design: This was an open-label, multiple-dose, five-regimen, twosequence clinical study lasting 5 weeks. Methods: Subjects were 16 healthy volunteers aged 18–40 years. Three 1-week regimens of metformin XR (500, 1000 and 1500mg once daily) were administered sequentially. Subjects were alternately given either metformin XR 2000mg once daily or metformin IR 1000mg twice daily during weeks 4 and 5. The pharmacokinetic properties of metformin XR were assessed on two separate days at steady state and compared with those of metformin IR. Results: Absorption of metformin XR was slower than that of metformin IR (time to maximum plasma concentration = 7 versus 3 hours). Maximum plasma concentrations (Cmax) following the administration of metformin XR 2000mg once daily was 36% higher than that following the evening dose of metformin IR 1000mg twice daily. The extent of absorption, determined by area under the plasma concentration-time curve (AUC), was equivalent for both formulations. The mean accumulation ratio of metformin XR was 1.0, indicating no accumulation with multiple-dose administration. Intrasubject variabilities in Cmax and AUC of metformin were comparable between metformin XR and metformin IR. This novel formulation of metformin XR was well tolerated at single doses up to 2000mg once daily for 7 days, and adverse events were similar to those reported with metformin IR. Conclusion: The pharmacokinetic parameters of metformin XR tablet using GelShield Diffusion System technology were similar to those of metformin IR. Metformin XR was well tolerated at single doses up to 2000mg once daily.


Journal of Pharmaceutical Sciences | 2011

PhRMA CPCDC initiative on predictive models of human pharmacokinetics, part 2: Comparative assessment of prediction methods of human volume of distribution

Rhys D.O. Jones; Hannah M. Jones; Malcolm Rowland; Christopher R. Gibson; James W.T. Yates; Jenny Y. Chien; Barbara J. Ring; Kimberly K. Adkison; M. Sherry Ku; Handan He; Ragini Vuppugalla; Punit Marathe; Volker Fischer; Sandeep Dutta; Vikash Sinha; Thorir Björnsson; Thierry Lavé; Patrick Poulin

The objective of this study was to evaluate the performance of various empirical, semimechanistic and mechanistic methodologies with and without protein binding corrections for the prediction of human volume of distribution at steady state (Vss ). PhRMA member companies contributed a set of blinded data from preclinical and clinical studies, and 18 drugs with intravenous clinical pharmacokinetics (PK) data were available for the analysis. In vivo and in vitro preclinical data were used to predict Vss by 24 different methods. Various statistical and outlier techniques were employed to assess the predictability of each method. There was not simply one method that predicts Vss accurately for all compounds. Across methods, the maximum success rate in predicting human Vss was 100%, 94%, and 78% of the compounds with predictions falling within tenfold, threefold, and twofold error, respectively, of the observed Vss . Generally, the methods that made use of in vivo preclinical data were more predictive than those methods that relied solely on in vitro data. However, for many compounds, in vivo data from only two species (generally rat and dog) were available and/or the required in vitro data were missing, which meant some methods could not be properly evaluated. It is recommended to initially use the in vitro tissue composition-based equations to predict Vss in preclinical species and humans, putting the assumptions and compound properties into context. As in vivo data become available, these predictions should be reassessed and rationalized to indicate the level of confidence (uncertainty) in the human Vss prediction. The top three methods that perform strongly at integrating in vivo data in this way were the Øie-Tozer, the rat -dog-human proportionality equation, and the lumped-PBPK approach. Overall, the scientific benefit of this study was to obtain greater characterization of predictions of human Vss from several methods available in the literature.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of orally active pyrrolopyridine- and aminopyridine-based Met kinase inhibitors

Zhen-Wei Cai; Donna D. Wei; Gretchen M. Schroeder; Lyndon A. M. Cornelius; Kyoung S. Kim; Xiao-Tao Chen; Robert J. Schmidt; David K. Williams; John S. Tokarski; Yongmi An; John S. Sack; Veeraswamy Manne; Amrita Kamath; Yueping Zhang; Punit Marathe; John T. Hunt; Louis J. Lombardo; Joseph Fargnoli; Robert M. Borzilleri

A series of acylurea analogs derived from pyrrolopyridine and aminopyridine scaffolds were identified as potent inhibitors of Met kinase activity. The SAR at various positions of the two kinase scaffolds was investigated. These studies led to the discovery of compounds 3b and 20b, which demonstrated favorable pharmacokinetic properties in mice and significant antitumor activity in a human gastric carcinoma xenograft model.


The Journal of Clinical Pharmacology | 2000

Pharmacokinetics and Bioavailability of a Metformin/Glyburide Tablet Administered Alone and with Food

Punit Marathe; Mark E. Arnold; Jeff Meeker; Douglas S. Greene; Rashmi H. Barbhaiya

Two randomized crossover studies were conducted to evaluate the pharmacokinetics (including food effect) of fixed‐combination metformin/glyburide tablets. Pharmacokinetics and bioavailability of two strengths (500 mg/2.5 mg and 500 mg/5 mg) of metformin/glyburide tablets were assessed relative to coadministered metformin and glyburide tablets in study 1. The effect of a high‐fat meal on the bioavailability of a metformin/glyburide (500 mg/5 mg) tablet was assessed relative to the fasted condition in study 2. The fixed combination metformin/glyburide tablets showed bioequivalence for the metformin component with the reference metformin tablet and comparable bioavailability for the glyburide component with the reference glyburide tablet. Food does not appear to affect the bioavailability of either component to an appreciable extent.


Bioorganic & Medicinal Chemistry Letters | 2008

Benzothiazole based inhibitors of p38α MAP kinase

Chunjian Liu; James Lin; Sidney Pitt; Rosemary Zhang; John S. Sack; Susan E. Kiefer; Kevin Kish; Arthur M. Doweyko; Hongjian Zhang; Punit Marathe; James M. Trzaskos; Murray McKinnon; John H. Dodd; Joel C. Barrish; Gary L. Schieven; Katerina Leftheris

Rational design, synthesis, and SAR studies of a novel class of benzothiazole based inhibitors of p38alpha MAP kinase are described. The issue of metabolic instability associated with vicinal phenyl, benzo[d]thiazol-6-yl oxazoles/imidazoles was addressed by the replacement of the central oxazole or imidazole ring with an aminopyrazole system. The proposed binding mode of this new class of p38alpha inhibitors was confirmed by X-ray crystallographic studies of a representative inhibitor (6a) bound to the p38alpha enzyme.


Journal of Pharmacology and Experimental Therapeutics | 2013

Cynomolgus Monkey as a Potential Model to Assess Drug Interactions Involving Hepatic Organic Anion Transporting Polypeptides: In Vitro, In Vivo, and In Vitro-to-In Vivo Extrapolation

Hong Shen; Zheng Yang; Mintier G; Han Yh; Cliff Chen; Balimane P; Jemal M; Zhao W; Zhang R; Kallipatti S; Selvam S; Sukrutharaj S; Krishnamurthy P; Punit Marathe; Rodrigues Ad

Organic anion–transporting polypeptides (OATP) 1B1, 1B3, and 2B1 can serve as the loci of drug–drug interactions (DDIs). In the present work, the cynomolgus monkey was evaluated as a potential model for studying OATP-mediated DDIs. Three cynomolgus monkey OATPs (cOATPs), with a high degree of amino acid sequence identity (91.9, 93.5, and 96.6% for OATP1B1, 1B3, and 2B1, respectively) to their human counterparts, were cloned, expressed, and characterized. The cOATPs were stably transfected in human embryonic kidney cells and were functionally similar to the corresponding human OATPs (hOATPs), as evident from the similar uptake rate of typical substrates (estradiol-17β-d-glucuronide, cholecystokinin octapeptide, and estrone-3-sulfate). Moreover, six known hOATP inhibitors exhibited similar IC50 values against cOATPs. To further evaluate the appropriateness of the cynomolgus monkey as a model, a known hOATP substrate [rosuvastatin (RSV)]-inhibitor [rifampicin (RIF)] pair was examined in vitro; the monkey-derived parameters (RSV Km and RIF IC50) were similar (within 3.5-fold) to those obtained with hOATPs and human primary hepatocytes. In vivo, the area under the plasma concentration-time curve of RSV (3 mg/kg, oral) given 1 hour after a single RIF dose (15 mg/kg, oral) was increased 2.9-fold in cynomolgus monkeys, consistent with the value (3.0-fold) reported in humans. A number of in vitro–in vivo extrapolation approaches, considering the fraction of the pathways affected and free versus total inhibitor concentrations, were also explored. It is concluded that the cynomolgus monkey has the potential to serve as a useful model for the assessment of OATP-mediated DDIs in a nonclinical setting.

Collaboration


Dive into the Punit Marathe'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

Sandhya Mandlekar

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge