Network


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

Hotspot


Dive into the research topics where Narendra S. Kishnani is active.

Publication


Featured researches published by Narendra S. Kishnani.


Drug Metabolism and Disposition | 2011

Characterization of Human Liver Enzymes Involved in the Biotransformation of Boceprevir, a Hepatitis C Virus Protease Inhibitor

Anima Ghosal; Yuan Yuan; Wei Tong; Ai-Duen Su; Chunyan Gu; Swapan K. Chowdhury; Narendra S. Kishnani; Kevin B. Alton

Boceprevir (SCH 503034), a protease inhibitor, is under clinical development for the treatment of human hepatitis C virus infections. In human liver microsomes, formation of oxidative metabolites after incubations with [14C]boceprevir was catalyzed by CYP3A4 and CYP3A5. In addition, the highest turnover was observed in recombinant CYP3A4 and CYP3A5. After a single radiolabeled dose to human, boceprevir was subjected to two distinct pathways, namely cytochrome P450-mediated oxidation and ketone reduction. Therefore, attempts were made to identify the enzymes responsible for the formation of carbonyl-reduced metabolites. Human liver S9 and cytosol converted ∼28 and ∼68% of boceprevir to M28, respectively, in the presence of an NADPH-generating system. Screening of boceprevir with recombinant human aldo-keto reductases (AKRs) revealed that AKR1C2 and AKR1C3 exhibited catalytic activity with respect to the formation of M+2 metabolites (M28 and M31). The formation of M28 was inhibited by 100 μM flufenamic acid (80.3%), 200 μM mefenamic acid (83.7%), and 100 μM phenolphthalein (86.1%), known inhibitors of AKRs, suggesting its formation through carbonyl reduction pathway. Formation of M28 was also inhibited by 100 μM diazepam (75.1%), 1 mM ibuprofen (70%), and 200 μM diflunisal (89.4%). These data demonstrated that CYP3A4 and CYP3A5 are primarily responsible for the formation of oxidative metabolites and the formation of M28 and M31, the keto-reduced metabolites, are most likely mediated by AKR1C2 and AKR1C3. Because the biotransformation and clearance of boceprevir involves two different enzymatic pathways, boceprevir is less likely to be a victim of significant drug-drug interaction with concomitant medication affecting either of these pathways.


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.


Pharmaceutical Research | 2003

Assessment of Temporal Biochemical and Gene Transcription Changes in Rat Liver Cytochrome P450: Utility of Real-Time Quantitative RT-PCR

Federico M. Goodsaid; Jairam Palamanda; Diana Montgomery; George Mandakas; Chunyan Gu; Zhiling Li; Xiaoli You; Laura Norton; Roger Smith; Inhou Chu; Tony Soares; Kevin B. Alton; Narendra S. Kishnani; I.Y. Rosenblum

AbstractPurpose. A conventional approach to assess cytochrome P450 (CYP) induction in preclinical animal models involves daily dosing for a least a week followed by Western blot and/or enzyme activity analysis. To evaluate the potential benefit of a third more specific and sensitive assay, real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), with the objective of reducing the duration of the conventional 1-week study, we simultaneously assessed gene expression by qRT-PCR along with Western blots and enzyme activity assays as a time course in an in vivo model. Methods. Rats were dosed daily for 8 days with model inducers of CYP1A, CYP2B, CYP3A, or CYP4A. Liver P450 levels were measured after 0.5, 1, 2, 4, and 8 days of dosing by qRT-PCR, Western blot, and enzyme activity. Results: CYP1A, CYP3A, and CYP4A genes were maximally induced very rapidly (0.5-1 day), whereas the CYP2B gene was maximally induced after a lag time of 4 days. In all cases, fold changes in induction detected by qRT-PCR were greater than fold changes in protein levels and enzyme activities. Conclusions. Maximal persistent and larger fold changes observed by qRT-PCR either preceded or occurred simultaneously with maximal sustained fold changes in protein levels as measured by Western blots and enzyme activity assays. Our data show that qRT-PCR provides increased sensitivity and specificity over conventional assays and may be key information for reliable assessment of drug-related changes in CYP induction during the transition from discovery to toxicology studies.


Cutaneous and Ocular Toxicology | 2007

SCH 412499: Biodistribution and Safety of an Adenovirus Containing P21WAF-1/CIP-1 Following Subconjunctival Injection in Cynomolgus Monkeys

Robert W. Veneziale; Christopher M. Bral; Dinesh P. Sinha; Robert W. Watkins; Mark E. Cartwright; Irwin Y. Rosenblum; Kimberley A. Treinen; Narendra S. Kishnani; Joyce Nelson; Zoe Chen; Barbara Faha; Daniel C. Maneval; Robert J. Munger; Xiao Yan Cai; Constance Cullen; Joseph C. Arezzo

Monkey studies were conducted for the preclinical safety assessment of SCH 412499, an adenovirus encoding p21, administered by subconjunctival injection prior to trabeculectomy for postoperative maintenance of the surgical opening. Biodistribution of SCH 412499 was minimal and there was no systemic toxicity. Findings included swollen, partially closed or shut eye(s) and transient congestion in the conjunctiva. A mononuclear cell infiltrate was present in the conjunctiva, choroid and other ocular tissues, but completely or partially resolved over time. Electroretinograms and visual evoked potentials revealed no adverse findings. Thus, the findings are not expected to preclude the clinical investigation of SCH 412499.


Journal of Chromatography B | 2002

High-performance liquid chromatographic method for the quantification of unbound evernimicin in human plasma ultrafiltrate.

Ruyun Zhong; Abraham Hernandez; Kevin B. Alton; Narendra S. Kishnani; James E. Patrick

A rapid HPLC method was developed for quantification of unbound evernimicin in human plasma. Protein-free samples prepared by ultrafiltration were injected directly onto a polymeric reversed-phase column and the eluent monitored at 302 nm. Evernimicin that eluted within 3.5 min was well resolved from endogenous components. Linearity was established between peak height and evernimicin concentration from 25 to 2500 ng/ml. Assay precision (C.V.) was within 5% while bias was no greater than 3%. This method has been used for the ex vivo assessment of evernimicin protein binding in human plasma from safety and tolerance as well as liver dysfunction and renal insufficiency studies.


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.


Archive | 2006

Medicaments and methods combining a HCV protease inhibitor and an AKR competitor

Anima Ghosal; Narendra S. Kishnani; Kevin B. Alton; Ronald E. White


Archive | 2006

Interferon-igg fusion

Leonard G. Presta; Narendra S. Kishnani


Archive | 2011

Inhibition of cyp3a drug metabolism

Anima Ghosal; Samir Gupta; Narendra S. Kishnani; Claudia Kasserra; Edward O'Mara

Collaboration


Dive into the Narendra S. Kishnani'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
Researchain Logo
Decentralizing Knowledge