Network


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

Hotspot


Dive into the research topics where Ragu Ramanathan is active.

Publication


Featured researches published by Ragu Ramanathan.


Journal of the American Society for Mass Spectrometry | 2002

In Electrospray Ionization Source Hydrogen/ Deuterium Exchange LC-MS and LC-MS/MS for Characterization of Metabolites

Wing Lam; Ragu Ramanathan

A new method is described for performing hydrogen/deuterium (H/D) exchange in an electrospray ionization (ESI) source. The use of liquid chromatography (LC)-mass spectrometer equipped with an ESI source and deuterium oxide (D2O) as the sheath liquid allows H/D exchange experiments to be performed on-line. This directly provides information for determining the number and position of exchangeable hydrogens, aiding in the elucidation of the structures of drug metabolites. To demonstrate the utility of this method, LC-mass spectrometry (MS) and LC-MS/MS experiments were performed using either H2O or D2O as sheath liquid on a matrix metalloprotease (MMP) inhibitor (PD 0200126) and its metabolites. Examination of the mass shift of the deuteriated molecule from that of the protonated molecule allowed the number of exchangeable protons to be determined. Interpretation of the production-spectra helped to determine the location of the exchanged protons and assisted in the assignment of the site(s) of modification for each metabolite.


Bioanalysis | 2010

Novel MS solutions inspired by MIST

Ragu Ramanathan; Jonathan L. Josephs; Mohammed Jemal; Mark E. Arnold; W. Griffith Humphreys

To improve patient safety and to help avoid costly late-stage failures, the pharmaceutical industry, along with the US FDA and International Committee on Harmonization (ICH), recommends the identification of differences in drug metabolism between animals used in nonclinical safety assessments and humans as early as possible during the drug-development process. LC-MS is the technique of choice for detection and characterization of metabolites, however, the widely different LC-MS response observed for a new chemical entity (NCE) and its structurally related metabolites limits the direct use of LC-MS responses for quantitative determination of NCEs and metabolites. While no method provides completely accurate universal response, UV, corona charged aerosol detection (CAD), radioactivity, NMR and low-flow (< 20 µl/min) nanospray approaches provide opportunities to quantify metabolites in the absence of reference standards or radiolabeled material with enough precision to meet the needs of early clinical development.


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%).


Journal of Pharmaceutical and Biomedical Analysis | 2002

Comparison of heterogeneous and homogeneous radioactivity flow detectors for simultaneous profiling and LC-MS/MS characterization of metabolites

Laura A Egnash; Ragu Ramanathan

Methods for simultaneous liquid chromatography-radioactivity monitor (LC-RAM) metabolite profiling and LC-tandem mass spectrometry (MS/MS) characterization of metabolites are described. Profiling and characterization of metabolites from three drug candidates from different therapeutic areas were compared using in-line heterogeneous LC-RAM-MS/MS and homogeneous LC-RAM-MS/MS methods. Although comparison shows that simultaneous metabolite profiling and characterization can be achieved using either heterogeneous or homogeneous-LC-RAM-MS/MS systems, a homogeneous system has the advantage in the following aspects, (1) sensitivity; (2) ease of method transfer; (3) less peak broadening problems due to the drug or metabolites adhering to the RAM cell; (4) accuracy in quantitation of the metabolites; and (5) the ability to load larger volumes of unprocessed biological fluids. Furthermore, the study shows that some of the possible metabolites that do not ionize well with electrospray ionization (ESI) and eluded detection by heterogeneous-LC-RAM detection could be very easily detected and characterized using a homogeneous-LC-RAM-MS/MS system.


Drug Metabolism and Disposition | 2011

Plasma stability-dependent circulation of acyl glucuronide metabolites in humans: how circulating metabolite profiles of muraglitazar and peliglitazar can lead to misleading risk assessment.

Donglu Zhang; Nirmala Raghavan; Lifei Wang; Yongjun Xue; Mary T. Obermeier; Stephanie Y. Chen; Shiwei Tao; Hao Zhang; Peter T. W. Cheng; Wenying Li; Ragu Ramanathan; Zheng Yang; W. Griffith Humphreys

Muraglitazar and peliglitazar, two structural analogs differing by a methyl group, are dual peroxisome proliferator-activated receptor-α/γ activators. Both compounds were extensively metabolized in humans through acyl glucuronidation to form 1-O-β-acyl glucuronide (AG) metabolites as the major drug-related components in bile, representing at least 15 to 16% of the dose after oral administration. Peliglitazar AG was the major circulating metabolite, whereas muraglitazar AG was a very minor circulating metabolite in humans. Peliglitazar AG circulated at lower concentrations in animal species than in humans. Both compounds had a similar glucuronidation rate in UDP-glucuronic acid-fortified human liver microsomal incubations and a similar metabolism rate in human hepatocytes. Muraglitazar AG and peliglitazar AG were chemically synthesized and found to be similarly oxidized through hydroxylation and O-demethylation in NADPH-fortified human liver microsomal incubations. Peliglitazar AG had a greater stability than muraglitazar AG in incubations in buffer, rat, or human plasma (pH 7.4). Incubations of muraglitazar AG or peliglitazar AG in plasma produced more aglycon than acyl migration products compared with incubations in the buffer. These data suggested that the difference in plasma stability, not differences in intrinsic formation, direct excretion, or further oxidation of muraglitazar AG or peliglitazar AG, contributed to the observed difference in the circulation of these AG metabolites in humans. The study demonstrated the difficulty in doing risk assessment based on metabolite exposure in plasma because the more reactive muraglitazar AG would not have triggered a threshold of concern based on the recent U.S. Food and Drug Administration guidance on Metabolites in Safety Testing, whereas the more stable peliglitazar AG would have.


Bioanalysis | 2016

2016 White Paper on recent issues in bioanalysis: focus on biomarker assay validation (BAV) (Part 1 – small molecules, peptides and small molecule biomarkers by LCMS)

Eric Yang; Jan Welink; Stephanie Cape; E.J. Woolf; Jens Sydor; C. A. James; Dina Goykhman; Mark E. Arnold; Neil Addock; Ronald Bauer; Michael Buonarati; Eugene Ciccimaro; Raj Dodda; Christopher Evans; Fabio Garofolo; Nicola Hughes; Rafiq Islam; Corey Nehls; Amanda Wilson; Chad Briscoe; Mark Bustard; Laura Coppola; Stephanie Croft; Dieter M. Drexler; Luca Ferrari; Daniela Fraier; Rand Jenkins; John Kadavil; Lloyd King; Wenkui Li

The 2016 10th Workshop on Recent Issues in Bioanalysis (10th WRIB) took place in Orlando, Florida with participation of close to 700 professionals from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations, and regulatory agencies worldwide. WRIB was once again a 5-day, weeklong event - A Full Immersion Week of Bioanalysis including Biomarkers and Immunogenicity. As usual, it was specifically designed to facilitate sharing, reviewing, discussing and agreeing on approaches to address the most current issues of interest including both small and large molecule analysis involving LCMS, hybrid LBA/LCMS, and LBA approaches, with the focus on biomarkers and immunogenicity. This 2016 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. This white paper is published in 3 parts due to length. This part (Part 1) discusses the recommendations for small molecules, peptides and small molecule biomarkers by LCMS. Part 2 (Hybrid LBA/LCMS and regulatory inputs from major global health authorities) and Part 3 (large molecule bioanalysis using LBA, biomarkers and immunogenicity) will be published in the Bioanalysis journal, issue 23.


Bioanalysis | 2016

Unconjugated payload quantification and DAR characterization of antibody–drug conjugates using high-resolution MS

Leanne Grafmuller; Ragu Ramanathan; Frank Barletta; Rick Steenwyk; Joseph Tweed

AIM The application of high-resolution MS to antibody-drug conjugate (ADC) drug development may provide insight into their safety and efficacy. Quantification of unconjugated cytotoxic drug (payload) and characterization of drug-to-antibody ratio distribution were determined in plasma using orthogonal acceleration quadrupole-time-of-flight MS. RESULTS Unconjugated payload quantification determined by quadrupole-time-of-flight-based MRM(highresolution) and triple quadrupole-based multiple reaction monitoring were comparable and achieved detection limits of 0.030 and 0.015 ng/ml, respectively. As determined by immunocapture and TOF-MS, drug-to-antibody ratio remained unchanged up to 3-weeks postdose for an ADC containing engineered glutamine linkers, but declined from four to three over 2 weeks in an ADC containing engineered cysteine linkers. CONCLUSION The use of high-resolution MS in ADC drug discovery confirms its utility within the bioanalytical discipline.


Journal of Pharmaceutical and Biomedical Analysis | 2002

Application of semi-automated metabolite identification software in the drug discovery process for rapid identification of metabolites and the cytochrome P450 enzymes responsible for their formation

Ragu Ramanathan; Donald L. McKenzie; Meera Tugnait; Kristen Siebenaler

Rapid identification of metabolites of compound X using data dependent scan function of a quadrupole ion trap mass spectrometer and semi-automated metabolite identification software is described. Compound X is metabolized via monooxygenation and desmethylation. LC-ESI-MS spectra obtained, following incubations of Compound X with microsomes in the presence and absence of chemical inhibitors specific for CYP1A2, CYP3A4, CYP2D6, CYP2C9 and CYP2E1, were processed using semi-automated metabolite identification software to extract information and to identify the cytochrome P450 enzymes responsible for metabolite formation. Chemical inhibition data suggests that the primary cytochrome P450 (CYP450) isozyme responsible for the metabolism of compound X is CYP3A4 with a minor contribution from both CYP2D6 and CYP2E1. Additionally, neither CYP2C9 nor CYP1A2 appears to contribute to the metabolism of compound X.

Collaboration


Dive into the Ragu Ramanathan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Walter A. Korfmacher

National Center for Toxicological Research

View shared research outputs
Top Co-Authors

Avatar

Chad Briscoe

University of Nebraska–Lincoln

View shared research outputs
Researchain Logo
Decentralizing Knowledge