Network


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

Hotspot


Dive into the research topics where David L. Bourdet is active.

Publication


Featured researches published by David L. Bourdet.


Journal of Pharmacology and Experimental Therapeutics | 2006

Ritonavir, Saquinavir, and Efavirenz, but Not Nevirapine, Inhibit Bile Acid Transport in Human and Rat Hepatocytes

MaryPeace McRae; Carolina M. Lowe; Xianbin Tian; David L. Bourdet; Richard Ho; Brenda F. Leake; Richard B. Kim; Kim L. R. Brouwer; Angela D. M. Kashuba

Human immunodeficiency virus-infected patients on antiretroviral drug therapy frequently experience hepatotoxicity, the underlying mechanism of which is poorly understood. Hepatotoxicity from other compounds such as bosentan and troglitazone has been attributed, in part, to inhibition of hepatocyte bile acid excretion. This work tested the hypothesis that antiretroviral drugs modulate hepatic bile acid transport. Ritonavir (28 μM), saquinavir (15 μM), and efavirenz (32 μM) inhibited [3H]taurocholate transport in bile salt export pump expressing Sf9-derived membrane vesicles by 90, 71, and 33%, respectively. In sandwich-cultured human hepatocytes, the biliary excretion index (BEI) of [3H]taurocholate was maximally decreased 59% by ritonavir, 39% by saquinavir, and 20% by efavirenz. Likewise, in sandwich-cultured rat hepatocytes, the BEI of [3H]taurocholate was decreased 100% by ritonavir and 94% by saquinavir. Sodium-dependent and -independent initial uptake rates of [3H]taurocholate in suspended rat hepatocytes were significantly decreased by ritonavir, saquinavir, and efavirenz. [3H]Taurocholate transport by recombinant NTCP and Ntcp was inhibited by ritonavir (IC50 = 2.1 and 6.4 μM in human and rat, respectively), saquinavir (IC50 = 6.7 and 20 μM, respectively), and efavirenz (IC50 = 43 and 97 μM, respectively). Nevirapine (75 μM) had no effect on bile acid transport in any model system. In conclusion, ritonavir, saquinavir, and efavirenz, but not nevirapine, inhibited both the hepatic uptake and biliary excretion of taurocholate.


Journal of Medicinal Chemistry | 2013

Discovery of a Novel Series of Potent Non-Nucleoside Inhibitors of Hepatitis C Virus NS5B

Ryan Craig Schoenfeld; David L. Bourdet; Ken A. Brameld; Elbert Chin; Javier de Vicente; Amy Fung; Seth F. Harris; Eun Kyung Lee; Sophie Le Pogam; Vincent Leveque; Jim Li; Alfred Sui-Ting Lui; Isabel Najera; Sonal Rajyaguru; Michael Sangi; Sandra Steiner; Francisco Xavier Talamas; Joshua Paul Gergely Taygerly; Junping Zhao

Hepatitis C virus (HCV) is a major global public health problem. While the current standard of care, a direct-acting antiviral (DAA) protease inhibitor taken in combination with pegylated interferon and ribavirin, represents a major advancement in recent years, an unmet medical need still exists for treatment modalities that improve upon both efficacy and tolerability. Toward those ends, much effort has continued to focus on the discovery of new DAAs, with the ultimate goal to provide interferon-free combinations. The RNA-dependent RNA polymerase enzyme NS5B represents one such DAA therapeutic target for inhibition that has attracted much interest over the past decade. Herein, we report the discovery and optimization of a novel series of inhibitors of HCV NS5B, through the use of structure-based design applied to a fragment-derived starting point. Issues of potency, pharmacokinetics, and early safety were addressed in order to provide a clinical candidate in fluoropyridone 19.


Journal of Pharmacology and Experimental Therapeutics | 2012

Prediction of Human Serotonin and Norepinephrine Transporter Occupancy of Duloxetine by Pharmacokinetic/Pharmacodynamic Modeling in the Rat

David L. Bourdet; Pamela R. Tsuruda; Glenmar P. Obedencio; Jacqueline A.M. Smith

Translation of central nervous system occupancy and clinical effect from animal models to humans has remained elusive for many pharmacological targets. The current studies evaluate the ability of a rodent pharmacokinetic/pharmacodynamic (PK/PD) modeling approach to translate ex vivo occupancy determined in rats to that observed after positron emission tomography (PET) imaging in humans for the dual serotonin transporter (SERT) and norepinephrine transporter (NET) inhibitor duloxetine. Ex vivo transporter occupancy in rat spinal cord was evaluated after single oral doses of 0.3 to 60 mg/kg. A novel methodology, based on the initial rates of association of transporter selective radioligands to tissue homogenates, was developed and validated for the assessment of ex vivo transporter occupancy. Duloxetine exhibited selectivity for occupancy of SERT over NET in rat spinal cord with ED50 values of 1 and 9 mg/kg, respectively. Corresponding EC50 values for the inhibition of SERT and NET based on unbound duloxetine spinal cord concentrations were 0.5 and 8 nM. An effect compartment PK/PD modeling approach was used to analyze the relationship between the time course of duloxetine plasma concentration and SERT and NET occupancy. Duloxetine inhibited SERT and NET in rat spinal cord with a plasma EC50 of 2.95 and 59.0 ng/ml, respectively. Similar plasma EC50 values for the inhibition of SERT (2.29–3.7 ng/ml) have been reported from human PET studies. This study illustrates the value of translational PK/PD modeling approaches and suggests that the preclinical modeling approach used in the current study is capable of predicting plasma concentrations associated with 50% occupancy of SERT in the human central nervous system.


Journal of Medicinal Chemistry | 2015

Novel Series of Dihydropyridinone P2X7 Receptor Antagonists

Francisco Javier Lopez-Tapia; Keith Adrian Murray Walker; Christine Brotherton-Pleiss; Joanie Caroon; Dov Nitzan; Lee Edwin Lowrie; Shelley K. Gleason; Shu-Hai Zhao; Jacob Berger; Debra Cockayne; Deborah Phippard; Rebecca T. Suttmann; William L. Fitch; David L. Bourdet; Pankaj D. Rege; Xiaojun Huang; Scott Broadbent; Charles Alois Dvorak; Jiang Zhu; Paul J. Wagner; Fernando Padilla; Brad Loe; Alam Jahangir; André Alker

Identification of singleton P2X7 inhibitor 1 from HTS gave a pharmacophore that eventually turned into potential clinical candidates 17 and 19. During development, a number of issues were successfully addressed, such as metabolic stability, plasma stability, GSH adduct formation, and aniline mutagenicity. Thus, careful modification of the molecule, such as conversion of the 1,4-dihydropyridinone to the 1,2-dihydropyridinone system, proper substitution at C-5″, and in some cases addition of fluorine atoms to the aniline ring allowed for the identification of a novel class of potent P2X7 inhibitors suitable for evaluating the role of P2X7 in inflammatory, immune, neurologic, or musculoskeletal disorders.


Journal of Pharmaceutical Sciences | 2016

Why Does the Intestine Lack Basolateral Efflux Transporters for Cationic Compounds? A Provocative Hypothesis

William R. Proctor; Xin Ming; David L. Bourdet; Tianxiang Kevin Han; Ruth S. Everett; Dhiren R. Thakker

Transport proteins in intestinal epithelial cells facilitate absorption of nutrients/compounds that are organic anions, cations, and zwitterions. For two decades, we have studied intestinal absorption and transport of hydrophilic ionic compounds, with specific focus on transport properties of organic cations and their interactions with intestinal transporters and tight junction proteins. Our data reveal how complex interactions between a compound and transporters in intestinal apical/basolateral (BL) membranes and tight junction proteins define oral absorption, and that the BL membrane lacks an efflux transporter that can transport positively charged compounds. Based on our investigations of transport mechanisms of zwitterionic, anionic, and cationic compounds, we postulate that physicochemical properties of these ionic species, in relation to the intestinal micro pH environment, have exerted evolutionary pressure for development of transporters that can handle apical uptake/efflux of all 3 ionic species and BL efflux of anions and zwitterions, but such evolutionary pressure is lacking for development of a BL efflux transporter for cationic compounds. This review provides an overview of intestinal uptake/efflux transporters and describes our studies on intestinal transport of cationic, anionic, and zwitterionic drugs that led to hypothesize that there are no cation-selective BL efflux transporters in the intestine.


Clinical Pharmacology & Therapeutics | 2018

Can BSEP Inhibition Testing In Drug Discovery And Development Reduce Liver Injury Risk? ‐ An International Transporter Consortium Perspective

J Gerry Kenna; Kunal S. Taskar; Christina Battista; David L. Bourdet; Kim L. R. Brouwer; Kenneth R. Brouwer; David Dai; Christoph Funk; Michael J. Hafey; Yurong Lai; Jonathan Maher; Y. Anne Pak; Jenny M. Pedersen; Joseph W. Polli; A. David Rodrigues; Paul B. Watkins; Kyunghee Yang; Robert Yucha

Bile salt export pump (BSEP) inhibition has emerged as an important mechanism that may contribute to the initiation of human drug‐induced liver injury (DILI). Proactive evaluation and understanding of BSEP inhibition is recommended in drug discovery and development to aid internal decision making on DILI risk. BSEP inhibition can be quantified using in vitro assays. When interpreting assay data, it is important to consider in vivo drug exposure. Currently, this can be undertaken most effectively by consideration of total plasma steady state drug concentrations (Css,plasma). However, because total drug concentrations are not predictive of pharmacological effect, the relationship between total exposure and BSEP inhibition is not causal. Various follow‐up studies can aid interpretation of in vitro BSEP inhibition data and may be undertaken on a case‐by‐case basis. BSEP inhibition is one of several mechanisms by which drugs may cause DILI, therefore, it should be considered alongside other mechanisms when evaluating possible DILI risk.


Journal of Pharmacology and Experimental Therapeutics | 2005

Differential Substrate and Inhibitory Activities of Ranitidine and Famotidine toward Human Organic Cation Transporter 1 (hOCT1; SLC22A1), hOCT2 (SLC22A2), and hOCT3 (SLC22A3)

David L. Bourdet; John B. Pritchard; Dhiren R. Thakker


Pharmaceutical Research | 2006

Intestinal Absorptive Transport of the Hydrophilic Cation Ranitidine: A Kinetic Modeling Approach to Elucidate the Role of Uptake and Efflux Transporters and Paracellular vs. Transcellular Transport in Caco-2 Cells

David L. Bourdet; Gary M. Pollack; Dhiren R. Thakker


Pharmaceutical Research | 2006

Saturable Absorptive Transport of the Hydrophilic Organic Cation Ranitidine in Caco-2 Cells: Role of pH-Dependent Organic Cation Uptake System and P-Glycoprotein

David L. Bourdet; Dhiren R. Thakker


Journal of Medicinal Chemistry | 2004

Photoaffinity labeling of the anionic sites in Caco-2 cells mediating saturable transport of hydrophilic cations ranitidine and famotidine

David L. Bourdet; Kiho Lee; Dhiren R. Thakker

Collaboration


Dive into the David L. Bourdet's collaboration.

Top Co-Authors

Avatar

Dhiren R. Thakker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kim L. R. Brouwer

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angela D. M. Kashuba

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge