TY - JOUR T1 - The Role of Aldehyde Oxidase and Xanthine Oxidase in the Biotransformation of a Novel Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 5 JF - Drug Metabolism and Disposition JO - Drug Metab Dispos SP - 1834 LP - 1845 DO - 10.1124/dmd.112.046136 VL - 40 IS - 9 AU - Ryan D. Morrison AU - Anna L. Blobaum AU - Frank W. Byers AU - Tammy S. Santomango AU - Thomas M. Bridges AU - Donald Stec AU - Katrina A. Brewer AU - Raymundo Sanchez-Ponce AU - Melany M. Corlew AU - Roger Rush AU - Andrew S. Felts AU - Jason Manka AU - Brittney S. Bates AU - Daryl F. Venable AU - Alice L. Rodriguez AU - Carrie K. Jones AU - Colleen M. Niswender AU - P. Jeffrey Conn AU - Craig W. Lindsley AU - Kyle A. Emmitte AU - J. Scott Daniels Y1 - 2012/09/01 UR - http://dmd.aspetjournals.org/content/40/9/1834.abstract N2 - Negative allosteric modulation (NAM) of metabotropic glutamate receptor subtype 5 (mGlu5) represents a therapeutic strategy for the treatment of childhood developmental disorders, such as fragile X syndrome and autism. VU0409106 emerged as a lead compound within a biaryl ether series, displaying potent and selective inhibition of mGlu5. Despite its high clearance and short half-life, VU0409106 demonstrated efficacy in rodent models of anxiety after extravascular administration. However, lack of a consistent correlation in rat between in vitro hepatic clearance and in vivo plasma clearance for the biaryl ether series prompted an investigation into the biotransformation of VU0409106 using hepatic subcellular fractions. An in vitro appraisal in rat, monkey, and human liver S9 fractions indicated that the principal pathway was NADPH-independent oxidation to metabolite M1 (+16 Da). Both raloxifene (aldehyde oxidase inhibitor) and allopurinol (xanthine oxidase inhibitor) attenuated the formation of M1, thus implicating the contribution of both molybdenum hydroxylases in the biotransformation of VU0409106. The use of 18O-labeled water in the S9 experiments confirmed the hydroxylase mechanism proposed, because 18O was incorporated into M1 (+18 Da) as well as in a secondary metabolite (M2; +36 Da), the formation of which was exclusively xanthine oxidase-mediated. This unusual dual and sequential hydroxylase metabolism was confirmed in liver S9 and hepatocytes of multiple species and correlated with in vivo data because M1 and M2 were the principal metabolites detected in rats administered VU0409106. An in vitro-in vivo correlation of predicted hepatic and plasma clearance was subsequently established for VU0409106 in rats and nonhuman primates. ER -