Network


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

Hotspot


Dive into the research topics where Anima Ghosal is active.

Publication


Featured researches published by Anima Ghosal.


Rapid Communications in Mass Spectrometry | 2009

A retention‐time‐shift‐tolerant background subtraction and noise reduction algorithm (BgS‐NoRA) for extraction of drug metabolites in liquid chromatography/mass spectrometry data from biological matrices

Peijuan Zhu; Wei Ding; Wei Tong; Anima Ghosal; Kevin B. Alton; Swapan K. Chowdhury

A retention-time-shift-tolerant background subtraction and noise reduction algorithm (BgS-NoRA) is implemented using the statistical programming language R to remove non-drug-related ion signals from accurate mass liquid chromatography/mass spectrometry (LC/MS) data. The background-subtraction part of the algorithm is similar to a previously published procedure (Zhang H and Yang Y. J. Mass Spectrom. 2008, 43: 1181-1190). The noise reduction algorithm (NoRA) is an add-on feature to help further clean up the residual matrix ion noises after background subtraction. It functions by removing ion signals that are not consistent across many adjacent scans. The effectiveness of BgS-NoRA was examined in biological matrices by spiking blank plasma extract, bile and urine with diclofenac and ibuprofen that have been pre-metabolized by microsomal incubation. Efficient removal of background ions permitted the detection of drug-related ions in in vivo samples (plasma, bile, urine and feces) obtained from rats orally dosed with (14)C-loratadine with minimal interference. Results from these experiments demonstrate that BgS-NoRA is more effective in removing analyte-unrelated ions than background subtraction alone. NoRA is shown to be particularly effective in the early retention region for urine samples and middle retention region for bile samples, where the matrix ion signals still dominate the total ion chromatograms (TICs) after background subtraction. In most cases, the TICs after BgS-NoRA are in excellent qualitative correlation to the radiochromatograms. BgS-NoRA will be a very useful tool in metabolite detection and identification work, especially in first-in-human (FIH) studies and multiple dose toxicology studies where non-radio-labeled drugs are administered. Data from these types of studies are critical to meet the latest FDA guidance on Metabolite in Safety Testing (MIST).


Drug Metabolism and Disposition | 2011

Identification of human liver cytochrome P450 enzymes involved in the metabolism of SCH 530348 (Vorapaxar), a potent oral thrombin protease-activated receptor 1 antagonist.

Anima Ghosal; Xiaowen Lu; Natalia A. Penner; Lan Gao; Ragu Ramanathan; Swapan K. Chowdhury; Narendra S. Kishnani; Kevin B. Alton

Vorapaxar (SCH 530348), a potent oral thrombin protease-activated receptor 1 antagonist, is being developed as an antiplatelet agent for patients with established vascular disease. The objective of this study was to identify the human liver cytochrome P450 (P450) enzyme(s) responsible for the metabolism of SCH 530348. Human liver microsomes metabolized SCH 530348 to M19, an amine metabolite formed via carbamate cleavage, and M20 (monohydroxy-SCH 530348). Recombinant human CYP3A4 exhibited the most activity (11.5% profiled radioactivity) for the formation of M19, followed by markedly less substrate conversion with CYP1A1 and CYP2C19. Trace levels of M19, a major excreted human metabolite, were detected with CYP1A2, CYP3A5, and CYP4F3A. Formation of M19 by human liver microsomes was inhibited 89% by ketoconazole (IC50, 0.73 μM), 34% by tranylcypromine, and 89% by anti-CYP3A4 monoclonal antibody. There was a significant correlation between the rate of M19 formation and midazolam 1′-hydroxylation (r = 0.75) or M19 formation and testosterone 6β-hydroxylation (r = 0.92). The results of screening, inhibition, and correlation studies confirmed that CYP3A4 is the major P450 enzyme responsible for M19 formation from SCH 530348. In contrast, formation of M20, a major circulating human metabolite at steady state, was primarily catalyzed by CYP3A4 and CYP2J2. M20 is pharmacologically equipotent to SCH 530348, whereas M19 is an inactive metabolite. Formation of M20 by human liver microsomes was inhibited 89% by ketoconazole, 75% by astemizole (a CYP2J2 inhibitor), and 43% by CYP3A4 monoclonal antibody. These results suggest that CYP3A4 and CYP2J2 are both involved in the formation of M20 metabolite.


Drug Metabolism and Disposition | 2007

IDENTIFICATION OF HUMAN LIVER CYTOCHROME P450 ENZYMES INVOLVED IN BIOTRANSFORMATION OF VICRIVIROC, A CCR5 RECEPTOR ANTAGONIST

Anima Ghosal; Ragu Ramanathan; Yuan Yuan; Neil Hapangama; Swapan K. Chowdhury; Narendra S. Kishnani; Kevin B. Alton

Vicriviroc (SCH 417690), a CCR5 receptor antagonist, is currently under investigation for the treatment of human immunodeficiency virus infection. The objective of this study was to identify human liver cytochrome P450 enzyme(s) responsible for the metabolism of vicriviroc. Human liver microsomes metabolized vicriviroc via N-oxidation (M2/M3), O-demethylation (M15), N,N-dealkylation (M16), N-dealkylation (M41), and oxidation to a carboxylic acid metabolite (M35b/M37a). Recombinant human CYP3A4 catalyzed the formation of all these metabolites, whereas CYP3A5 catalyzed the formation of M2/M3 and M41. CYP2C9 only catalyzed the formation of M15. There was a high correlation between the rates of formation of M2/M3, M15, and M41, which was determined using 10 human liver microsomal samples and testosterone 6β-hydroxylation catalyzed by CYP3A4/5 (r ≥ 0.91). Ketoconazole and azamulin (inhibitors of CYP3A4) were potent inhibitors of the formation of M2/M3, M15, M41, and M35b/M37a from human liver microsomes. A CYP3A4/5-specific monoclonal antibody (1 μg/μg of protein) inhibited the formation of all metabolites from human liver microsomes by 86 to 100%. The results of this study suggest that formation of the major vicriviroc metabolites in human liver microsomes is primarily mediated via CYP3A4. CYP2C9 and CYP3A5 most likely play a minor role in the biotransformation of this compound. These enzymology data will provide guidance to design clinical studies to address any potential drug-drug interactions.


Drug Metabolism Letters | 2009

Metabolism of Loratadine and Further Characterization of Its In Vitro Metabolites

Anima Ghosal; Samir Gupta; Ragu Ramanathan; Yuan Yuan; Xiaowen Lu; A.-D. Su; N. Alvarez; Shmuel Zbaida; Swapan K. Chowdhury; Kevin B. Alton

The present study demonstrated that in addition to CYP3A4 and CYP2D6, the metabolism of loratadine is also catalyzed by CYP1A1, CYP2C19, and to a lesser extent by CYP1A2, CYP2B6, CYP2C8, CYP2C9 and CYP3A5. The biotransformation of loratadine was associated with the formation of desloratadine (DL) and further hydroxylation of both DL and the parent drug (loratadine). Based on the inhibition and correlation studies contribution of CYP2C19 in the formation of the major circulating metabolite DL seems to be minor. Reported clinical results suggest that the steady state mean (%CV) plasma Cmax and AUC(24hr) of loratadine were 4.73 ng/ml (119%) and 24.1 ng.hr/ml (157%), respectively, after dosing with 10 mg loratadine tablets for 10 days. High inter-subject variability in loratadine steady-state data is probably due to the phenotypical characteristics of CYP2D6, CYP2C19, and CYP3A4. The relative abundance of CYP3A4 in the human liver exceeds that of CYP2C19 and CYP2D6 and therefore the contribution of CYP3A4 in the metabolism of loratadine should be major (approximately 70%).


Drug Metabolism and Disposition | 2006

IDENTIFICATION OF HUMAN LIVER CYTOCHROME P450 ENZYMES RESPONSIBLE FOR THE METABOLISM OF LONAFARNIB (SARASAR)

Anima Ghosal; Swapan Chowdhury; Wei Tong; Neil Hapangama; Yuan Yuan; Ai-Duen Su; Shmuel Zbaida

Lonafarnib (Sarasar), a farnesyl transferase inhibitor, is currently under development for the treatment of solid tumors. Incubation of lonafarnib with human liver microsomes resulted in the formation of four oxidative metabolites (M1, M2, M3, and M4). Minor to trace levels of these metabolites were detected in humans after multiple-dose administration of lonafarnib. Liquid chromatography-mass spectrometry analyses exhibited a mass to charge ratio (m/z) for the (M+H)+ ion of M1, M2, M3, and M4 at 653, 635, 669, and 653 Th, respectively. These metabolites, respectively, resulted from changes of +O, –2H, +2O, and +O relative to lonafarnib. Recombinant human CYP3A4 and CYP3A5 exhibited catalytic activity with respect to the formation of M1, M2, and M3, whereas CYP2C8 exhibited catalytic activity with respect to the formation of M4. There was a high correlation between the formation of M1, determined in 10 human liver microsomal samples, and 6β-hydroxylation of testosterone catalyzed by CYP3A4/5 (r = 0.93). The IC50 values of ketoconazole for inhibition of M1 and M2 were 0.61 and 0.92 μM, respectively. The formation of M4 by human liver microsomes was inhibited 72% by 50 μM quercetin, suggesting that the formation of M4 was mediated via CYP2C8. A CYP3A4/5-specific inhibitory monoclonal antibody inhibited the formation of M1, M2, and M3 by 85, 75, and 100%, respectively. In conclusion, the formation of metabolites M1, M2, and M3 from lonafarnib was mediated via CYP3A4 and CYP3A5.


Xenobiotica | 2005

Identification of human liver cytochrome P450 enzymes involved in the metabolism of SCH 351125, a CCR5 antagonist

Anima Ghosal; Swapan K. Chowdhury; Samir Gupta; Yuan Yuan; Robert M. Iannucci; H. Zhang; Shmuel Zbaida; James E. Patrick; Kevin B. Alton

The identification and relative contribution of human cytochrome P450 enzyme(s) involved in the metabolism of SCH 351125 were investigated. In human liver microsomes, O-deethylation was the major metabolic pathway, whereas aromatization of a piperidine ring to pyridine and the reduction of the N-oxide moiety were minor routes. Recombinant human CYP3A4 and CYP2C9 both exhibited catalytic activity with respect to the formation of rotameric O-deethylated metabolites (M12, M13), the metabolites resulting from aromatization (M22/M24) and N-oxide reduction (M31). Using the relative activity factor (RAF) approach, the relative contributions of CYP3A4 and CYP2C9 to M13 formation were estimated to be 76 and 24%, respectively. There was a high correlation (r > 0.96) between the rate of formation of M12 and M13 and 6β-hydroxylation of testosterone catalysed by CYP3A4/5. Ketoconazole (2 µM) and CYP3A4/5-specific inhibitory monoclonal antibody inhibited the formation of M12 and M13 from human liver microsomes by approximately 60 and 71%, respectively. The results demonstrate that the in vitro metabolism of SCH 351125 is mediated primarily via CYP3A4 and that CYP2C9 plays a minor role. Clinical study designs should encompass these enzymology data to address any potential drug interactions.


Retrovirology | 2005

Characterization of the In Vitro Human Liver Cytochrome P450 (CYP) Mediated Metabolism and Inhibition Potential of Vicriviroc

Anima Ghosal; Mary Barecki-Roach; Ragulan Ramanathan; Yuan Yuan; Chris Casciano

Vicriviroc (formerly SCH 417690), a CCR5 receptor antagonist, is currently under investigation for the treatment of HIV infection. Human liver microsomes (HLM) metabolized vicriviroc via N-oxidation (M2/M3), Odemethylation (M15), N, N-dealkylation (M16), Ndealkylation (M41) and carboxylic acid formation (M35b/M37a). The metabolites generated under in vitro conditions were also detected in clinical studies after oral doses of vicriviroc. Incubation with recombinant human CYP3A4 formed all metabolites listed above, while CYP2C9 formed M15 and CYP3A5 formed M2/M3 and M41.


Progress in Pharmaceutical and Biomedical Analysis | 2005

Chapter 12 Cytochrome p450 (cyp) and udp-glucuronosyltransferase (ugt) enzymes: role in drug metabolism, polymorphism, and identification of their involvement in drug metabolism

Anima Ghosal; Ragulan Ramanathan; Narendra S. Kishnani; Swapan K. Chowdhury; Kevin B. Alton

Publisher Summary The biotransformation of drugs is divided into two categories—Phase I and Phase II metabolism. Phase I reactions include oxidation, reduction, hydrolysis, and hydration. Metabolic oxidations usually occur through the action of cytochrome P450 (CYP) oxidative enzymes. Although there are at least 50 different P450 isoforms, drug metabolism in humans most likely involve CYP1A2, CYP3A4, CYP2C9, CYP2C19, and CYP2D6. P450 enzymes catalyze aromatic hydroxylation, aliphatic hydroxylation, N-, O-, and S-dealkylation; N-hydroxylation, N-oxidation, sulfoxidation, deamination,and dehalogenation. This chapter focuses on CYP enzymes for Phase I metabolism and UDP-glucuronosyltransferase (UGT) enzymes for Phase II metabolism. Understanding the involvement of P450 enzymes in drug metabolism is critical to assessing the potential for drug interaction with concomitant drugs, food, and endogenous substances. Phase I enzymes primarily modify lipophilic molecules by creating polar functionalities to increase hydrophilicity, which thereby facilitates clearance from the body. Such additional functionalities may be readily amenable to Phase II conjugation reactions. Glucuronidation is the major conjugation pathway probably due to the relatively high natural abundance of the reaction co-factor, UDP-glucuronic acid. This process occurs with alcohols, phenols, hydroxylamines, carboxylic acids, amines, sulfonamides, and thiols. The role of these enzymes in drug metabolism is reviewed within the context of their polymorphism and analytical technologies used today in determining their involvement in drug metabolism are presented.


Drug Metabolism and Disposition | 2004

Identification of human UDP-glucuronosyltransferase enzyme(s) responsible for the glucuronidation of posaconazole (noxafil)

Anima Ghosal; Neil Hapangama; Yuan Yuan; Joana Achanfuo-Yeboah; Robert M. Iannucci; Swapan Chowdhury; Kevin B. Alton; James E. Patrick; Shmuel Zbaida


Drug Metabolism and Disposition | 2004

IDENTIFICATION OF HUMAN UDP-GLUCURONOSYLTRANSFERASE ENZYME(S) RESPONSIBLE FOR THE GLUCURONIDATION OF EZETIMIBE (ZETIA)

Anima Ghosal; Neil Hapangama; Yuan Yuan; Joana Achanfuo-Yeboah; Robert M. Iannucci; Swapan Chowdhury; Kevin B. Alton; James E. Patrick; Shmuel Zbaida

Collaboration


Dive into the Anima Ghosal'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
Researchain Logo
Decentralizing Knowledge