Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 50, 2020 - Issue 1
752
Views
22
CrossRef citations to date
0
Altmetric
Review Articles

Aldehyde oxidase; new approaches to old problems

Pages 34-50 | Received 29 Apr 2019, Accepted 28 May 2019, Published online: 19 Jun 2019

References

  • Adkins DE, Clark SL, Åberg K, et al. (2012). Genome-wide pharmacogenomic study of citalopram-induced side effects in STAR*D. Transl Psychiat 2:e129.
  • Akabane T. (2012) Impacts of species differences in drug metabolizing enzymes on human bioavailability prediction. [PhD Thesis] Chiba, Japan: Chiba University.
  • Akabane T, Gerst N, Naritomi Y, et al. (2012). A practical and direct comparison of intrinsic metabolic clearance of several non-CYP enzyme substrates in freshly isolated and cryopreserved hepatocytes. Drug Metab Pharmacok 27:181–91.
  • Akabane T, Tanaka K, Irie M, et al. (2011). Case report of extensive metabolism by aldehyde oxidase in humans: pharmacokinetics and metabolite profile of FK3453 in rats, dogs, and humans. Xenobiotica 41:372–84.
  • Al Salhen KS. (2014). In vitro oxidation of aldehyde oxidase from rabbit liver: specificity toward endogenous substrates. J King Saud Univ 26:67–74.
  • Alfaro JF, Jones JP. (2008). Studies on the mechanism of aldehyde oxidase and xanthine oxidase. J Org Chem 73:9469–72.
  • Al-Omar MA, Beedham C, Belal F, et al. (2005). Fluorimetric measurement of hydrogen peroxide produced during aldehyde oxidase catalysed oxidation using scopoletin. J Med Sci 5:10–20.
  • Alousi AM, Boinpally R, Wiegand R, et al. (2006). A phase 1 trial of XK469: toxicity profile of a selective topoisomerase IIβ inhibitor. Invest New Drugs 25:147–54.
  • Amano T, Fukami T, Ogiso T, et al. (2018). Identification of enzymes responsible for dantrolene metabolism in the human liver: a clue to uncover the cause of liver injury. Biochem Pharmacol 151:69–78.
  • Anderson LW, Collins JM, Klecker RW, et al. (2005). Metabolic profile of XK469, 2(R)-[4-(7-chloro-2-quinoxalinyl)oxyphenoxy]-propionic acid; NSC698215) in patients and in vitro: low potential for active or toxic metabolites or for drug–drug interactions. Cancer Chemoth Pharm 56:351–7.
  • Aoyama K, Matsubara K, Okada K, et al. (2000). N-Methylation ability for azaheterocyclic amines is higher in Parkinsons disease: nicotinamide loading test. J Neural Transm 107:985–95.
  • Apenova N, Peng H, Hecker M, et al. (2018). A rapid and sensitive fluorometric method for determination of aldehyde oxidase activity. Toxicol Appl Pharm 341:30–7.
  • Argikar UA, Potter PM, Hutzler JM, et al. (2016). Challenges and opportunities with non-CYP enzymes aldehyde oxidase, carboxylesterase, and UDP-glucuronosyltransferase: focus on reaction phenotyping and prediction of human clearance. AAPS J 18:1391–405.
  • Austin NE, Baldwin SJ, Cutler L, et al. (2001). Pharmacokinetics of the novel, high-affinity and selective dopamine D3 receptor antagonist SB-277011 in rat, dog and monkey: in vitro/in vivo correlation and the role of aldehyde oxidase. Xenobiotica 31:677–86.
  • Bannon P, Yu P, Cook JM, et al. (1998). Identification of quinine metabolites in urine after oral dosing in humans. J Chromatogr B Biomed Sci Appl 715:387–93.
  • Barr JT, Choughule KV, Nepal S, et al. (2014). Why do most human liver cytosol preparations lack xanthine oxidase activity? Drug Metab Dispos 42:695–9.
  • Barr JT, Jones JP, Joswig-Jones CA, Rock DA. (2013). Absolute quantification of aldehyde oxidase protein in human liver using liquid chromatography–tandem mass spectrometry. Mol Pharm 10:3842–9.
  • Bathelt CM, Ridder L, Mulholland AJ, et al. (2004). Mechanism and structure-reactivity relationships for aromatic hydroxylation by cytochrome P450. Org Biomol Chem 2:2998–3005.
  • Beedham C, Al-Tayib Y, Smith JA. (1992). Role of guinea pig and rabbit hepatic aldehyde oxidase in oxidative in vitro metabolism of cinchona antimalarials. Drug Metab Dispos 20:889–95.
  • Beedham C, Critchley DJP, Rance DJ. (1995a). Substrate specificity of human liver aldehyde oxidase toward substituted quinazolines and phthalazines: a comparison with hepatic enzyme from guinea pig, rabbit, and baboon. Arch Biochem Biophys 319:481–90.
  • Beedham C, Peet CF, Panoutsopoulos GI, et al. (1995b). Role of aldehyde oxidase in biogenic amine metabolism. Prog Brain Res 106:345–53.
  • Bender DA. (2012). Amino acid metabolism. Chichester, England: Wiley-Blackwell.
  • Berg SL, Murry DJ, McCully CL, et al. (1998). Pharmacokinetics of O6-benzylguanine and its active metabolite 8-oxo-O6-benzylguanine in plasma and cerebrospinal fluid after intrathecal administration of O6-benzylguanine in the nonhuman primate. Clin Cancer Res 4:2891–4.
  • Cantú Reinhard FG, Sainna MA, Upadhyay P, et al. (2016). A systematic account on aromatic hydroxylation by a cytochrome P450 model compound I: a low‐pressure mass spectrometry and computational study. Chem Eur J 22:18608–19.
  • Carroll MB, Smith DM, Shaak TL. (2017). Genomic sequencing of uric acid metabolizing and clearing genes in relationship to xanthine oxidase inhibitor dose. Rheumatol Int 37:445–53.
  • Castellino S, O’Mara M, Koch K, et al. (2012). Human metabolism of lapatinib, a dual kinase inhibitor: implications for hepatotoxicity. Drug Metab Dispos 40:139–50.
  • Cerqueira NMFSA, Coelho C, Brás NF, et al. (2015). Insights into the structural determinants of substrate specificity and activity in mouse aldehyde oxidases. J Biol Inorg Chem 20:209–17.
  • Chen H, Evarts J, Webb H, et al. (2012). Biotransformation of GS-1101, CAL-101), a potent and selective inhibitor of PI3K delta for the treatment of patients with hematologic malignancies. FASEB J 26:850.
  • Chen H, Lin Y, Han M, et al. (2010). Simultaneous quantitative analysis of fasudil and its active metabolite in human plasma by liquid chromatography electro-spray tandem mass spectrometry. J Pharm Biomed Anal 52:242–8.
  • Choughule KV, Barr JT, Jones JP. (2013). Evaluation of rhesus monkey and guinea pig hepatic cytosol fractions as models for human aldehyde oxidase. Drug Metab Dispos 41:1852–8.
  • Coelho C, Foti A, Hartmann T, et al. (2015). Structural insights into xenobiotic and inhibitor binding to human aldehyde oxidase. Nat Chem Biol 11:779–83.
  • Coelho C, Mahro M, Trincão J, et al. (2012). The first mammalian aldehyde oxidase crystal structure: insights into substrate specificity. J Biol Chem 287:40690–702.
  • Coene KLM, Kluijtmans LAJ, van der Heeft E, et al. (2018). Next-generation metabolic screening: targeted and untargeted metabolomics for the diagnosis of inborn errors of metabolism in individual patients. J Inherit Metab Dis 41:337–53.
  • Critchley DJP, Rance DJ, Beedham C. (1994). Biotransformation of carbazeran in guinea pig: effect of hydralazine pretreatment. Xenobiotica 24:37–47.
  • Crnogorac–Jurcevic T, Gangeswaran R, Bhakta V, et al. (2005). Proteomic analysis of chronic pancreatitis and pancreatic adenocarcinoma. Gastroenterology 129:1454–63.
  • Crouch RD, Blobaum AL, Felts AS, et al. (2017). Species-specific involvement of aldehyde oxidase and xanthine oxidase in the metabolism of the pyrimidine-containing mGlu5-negative allosteric modulator VU0424238, auglurant. Drug Metab Dispos 45:1245–59.
  • Crouch RD, Hutzler JM, Daniels JS. (2018). A novel in vitro allometric scaling methodology for aldehyde oxidase substrates to enable selection of appropriate species for traditional allometry. Xenobiotica 48:219–31.
  • Crouch RD, Morrison RD, Byers FW, et al. (2016). Evaluating the disposition of a mixed aldehyde oxidase/cytochrome P450 substrate in rats with attenuated P450 activity. Drug Metab Dispos 44:1296–303.
  • Cruciani G, Milani N, Benedetti P, et al. (2018). From experiments to a fast easy-to-use computational methodology to predict human aldehyde oxidase selectivity and metabolic reactions. J Med Chem 61:360–71.
  • Cuneo A, Barosi G, Danesi R, et al. (2019). Management of adverse events associated with idelalisib treatment in chronic lymphocytic leukemia and follicular lymphoma: a multidisciplinary position paper. Hematol Oncol 37:3–14.
  • Dalgaard L. (2015). Comparison of minipig, dog, monkey and human drug metabolism and disposition. J Pharmacol Toxicol 74:80–92.
  • Dalvie D, Sun H, Xiang C, et al. (2012). Effect of structural variation on aldehyde oxidase-catalyzed oxidation of zoniporide. Drug Metab Dispos 40:1575–87.
  • Dalvie D, Xiang C, Kang P, Zhou S. (2013). Interspecies variation in the metabolism of zoniporide by aldehyde oxidase. Xenobiotica 43:399–408.
  • Dalvie D, Zhang C, Chen W, et al. (2010). Cross-species comparison of the metabolism and excretion of zoniporide: contribution of aldehyde oxidase to interspecies differences. Drug Metab Dispos 38:641–54.
  • Day RO, Graham GG, Hicks M, et al. (2007). Clinical pharmacokinetics and pharmacodynamics of allopurinol and oxypurinol. Clin Pharmacokinet 46:623–44.
  • Diamond S, Boer J, Maduskuie T, et al. (2010). Species-specific metabolism of SGX523 by aldehyde oxidase and the toxicological implications. Drug Metab Dispos 38:1277–85.
  • Dick RA. (2018). Refinement of in vitro methods for identification of aldehyde oxidase substrates reveals metabolites of kinase inhibitors. Drug Metab Dispos 46:846–59.
  • Dick RA, Kanne DB, Casida JE. (2005). Identification of aldehyde oxidase as the neonicotinoid nitroreductase. Chem Res Toxicol 18:317–23.
  • Dick RA, Kanne DB, Casida JE. (2006). Substrate specificity of rabbit aldehyde oxidase for nitroguanidine and nitromethylene neonicotinoid insecticides. Chem Res Toxicol 19:38–43.
  • Dick RA, Kanne DB, Casida JE. (2007). Nitroso-imidacloprid irreversibly inhibits rabbit aldehyde oxidase. Chem Res Toxicol 20:1942–6.
  • Dittrich C, Greim G, Borner M, et al. (2002). Phase I and pharmacokinetic study of BIBX 1382 BS, an epidermal growth factor receptor, EGFR) inhibitor, given in a continuous daily oral administration. Eur J Cancer 38:1072–80.
  • Dubbelman AC, Nijenhuis CM, Jansen RS, et al. (2016). Metabolite profiling of the multiple tyrosine kinase inhibitor lenvatinib: a cross-species comparison. Invest New Drugs 34:300–18.
  • Fagerberg L, Hallström BM, Oksvold P, et al. (2014). Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomic. Mol Cell Proteomics 13:397–406.
  • Foti RS, Dalvie DK. (2016). Cytochrome P450 and non-cytochrome P450 oxidative metabolism: contributions to the pharmacokinetics, safety, and efficacy of xenobiotics. Drug Metab Dispos 44:1229–45.
  • Foti A, Dorendorf F, Leimkühler S. (2017). A single nucleotide polymorphism causes enhanced radical oxygen species production by human aldehyde oxidase. PLoS One 12:e0182061.
  • Foti A, Hartmann T, Coelho C, et al. (2016). Optimization of the expression of human aldehyde oxidase for investigations of single-nucleotide polymorphisms. Drug Metab Dispos 44:1277–85.
  • Fu C, Di L, Han X, et al. (2013). Aldehyde oxidase 1 in human liver cytosols: quantitative characterization of AOX1 expression level and activity relationship. Drug Metab Dispos 41:1797–804.
  • Gadepalli RSVS, Rimoldi JM, Fronczek FR, et al. (2007). Synthesis of fenthion sulfoxide and fenoxon sulfoxide enantiomers: effect of sulfur chirality on acetylcholinesterase activity. Chem Res Toxicol 20:257–62.
  • Gamberi T, Magherini F, Modesti A, et al. (2018). Adiponectin signaling pathways in liver diseases. Biomedicines 6:52.
  • Garattini E, Fratelli M, Terao M. (2008). Mammalian aldehyde oxidases: genetics, evolution and biochemistry. Cell Mol Life Sci 65:1019–48.
  • Garattini E, Fratelli M, Terao M. (2009). The mammalian aldehyde oxidase gene family. Hum Genomics 4:119–30.
  • Garattini E, Terao M. (2013). Aldehyde oxidase and its importance in novel drug discovery: present and future challenges. Expert Opin Drug Dis 8:641–54.
  • Haldrup C, Mundbjerg K, Vestergaard EM, et al. (2013). DNA methylation signatures for prediction of biochemical recurrence after radical prostatectomy of clinically localized prostate cancer. J Clin Oncol 31:3250–8.
  • Hall WW, Krenitsky TA. (1986). Aldehyde oxidase from rabbit liver: specificity toward purines and their analogs. Arch Biochem Biophys 251:36–46.
  • Hartmann T, Terao M, Garattini E, et al. (2012). The impact of single nucleotide polymorphisms on human aldehyde oxidase. Drug Metab Dispos 40:856–64.
  • Hayes A, Mok NY, Liu M, et al. (2017). Pyrido [3, 4-d] pyrimidin-4, 3 H)-one metabolism mediated by aldehyde oxidase is blocked by C2-substitution. Xenobiotica 47:771–7.
  • Hille R, Hall J, Basu P. (2014). The mononuclear molybdenum enzymes. Chem Rev 114:3963–4038.
  • Hori H. (2014). Methylated nucleosides in tRNA and tRNA methyltransferases. Front Genet 5:144.
  • Hu T, Khambatta ZS, Hayden PJ, et al. (2010). Xenobiotic metabolism gene expression in the EpiDermin vitro 3D human epidermis model compared to human skin. Toxicol In Vitro 24:1450–63.
  • Huang S, Adams E, Van Schepdael A. (2019). Study of aldehyde oxidase by micellar electrokinetic chromatography separation of O6-benzylguanine and 8-oxo-O6-benzylguanine. Electrophoresis 40:330–5.
  • Huang DY, Furukawa A, Ichikawa Y. (1999). Molecular cloning of retinal oxidase/aldehyde oxidase cDNAs from rabbit and mouse livers and functional expression of recombinant mouse retinal oxidase cDNA in Escherichia coli. Arch Biochem Biophys 364:264–72.
  • Huang F, Koenen-Bergmann M, MacGregor TR, et al. (2008). Pharmacokinetic and safety evaluation of BILR 355, a second-generation nonnucleoside reverse transcriptase inhibitor, in healthy volunteers. Antimicrob Agents Ch 52:4300–7.
  • Hussein Z, Mizuo H, Hayato S, et al. (2017). Clinical pharmacokinetic and pharmacodynamic profile of lenvatinib, an orally active, small-molecule, multitargeted tyrosine kinase inhibitor. Eur J Drug Metab Ph 42:903–14.
  • Hutzler JM, Cerny MA, Yang Y-S, et al. (2014a). Cynomolgus monkey as a surrogate for human aldehyde oxidase metabolism of the EGFR inhibitor BIBX1382. Drug Metab Dispos 42:1751–60.
  • Hutzler JM, Yang YS, Albaugh D, et al. (2012). Characterization of aldehyde oxidase enzyme activity in cryopreserved human hepatocytes. Drug Metab Dispos 40:267–75.
  • Hutzler JM, Yang Y-S, Brown C, et al. (2014b). Aldehyde oxidase activity in donor-matched fresh and cryopreserved human hepatocytes and assessment of variability in 75 donors. Drug Metab Dispos 42:1090–7.
  • Ichida K, Amaya Y, Okamoto K, et al. (2012). Mutations associated with functional disorder of xanthine oxidoreductase and hereditary xanthinuria in humans. Int J Mol Sci 13:15475–95.
  • Ichida K, Matsumura T, Sakuma R, et al. (2001). Mutation of human molybdenum cofactor sulfurase gene is responsible for classical xanthinuria type II. Biochem Biophys Res Commun 282:1194–200.
  • Ichida K, Yoshida M, Sakuma R, et al. (1998). Two siblings with classical xanthinuria type 1: significance of allopurinol loading test. Intern Med 37:77–82.
  • Infante JR, Rugg T, Gordon M, et al. (2013). Unexpected renal toxicity associated with SGX523, a small molecule inhibitor of MET. Invest New Drug 31:363–9.
  • Inoue K, Mizuo H, Kawaguchi S, et al. (2014). Oxidative metabolic pathway of lenvatinib mediated by aldehyde oxidase. Drug Metab Dispos 42:1326–33.
  • Isobe T, Ohta M, Kaneko Y, et al. (2016). Species differences in metabolism of ripasudil (K-115) are attributed to aldehyde oxidase. Xenobiotica 46:579–90.
  • Itoh K. (2009). [Individual and strain differences of aldehyde oxidase in the rat]. Yakugaku Zasshi 129:1487–93.
  • Itoh K, Masubuchi A, Sasaki T, et al. (2007). Genetic polymorphism of aldehyde oxidase in donryu rats. Drug Metab Dispos 35:734–9.
  • Itoh K, Wakabayashi N, Katoh Y, et al. (1999). Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev 13:76–86.
  • Itoh K, Yamamura M, Takasaki W, et al. (2006). Species differences in enantioselective 2‐oxidations of RS‐8359, a selective and reversible MAO‐A inhibitor, and cinchona alkaloids by aldehyde oxidase. Biopharm Drug Dispos 27:133–9.
  • Jackman JE, Montange RK, Malik HS, et al. (2003). Identification of the yeast gene encoding the tRNA m1G methyltransferase responsible for modification at position 9. RNA 9:574–85.
  • Janßen H, Janßen PHE, Broelsch CE. (2004). UW is superior to celsior and HTK in the protection of human liver endothelial cells against preservation injury. Liver Transplant 10:1514–23.
  • Jensen KG, Jacobsen A-M, Bundgaard C, et al. (2016). Lack of exposure in a first-in-man study due to aldehyde oxidase metabolism: investigated by use of 14C-microdose, humanized mice, monkey pharmacokinetics, and in vitro methods. Drug Metab Dispos 45:68–75.
  • Jin F, Robeson M, Zhou H, et al. (2014). Drug interaction profile of idelalisib and its major metabolite, GS-563117. J Clin Oncol 32:2593.
  • Jin F, Robeson M, Zhou H, et al. (2015). Clinical drug interaction profile of idelalisib in healthy subjects. J Clin Pharmacol 55:909–19.
  • Jones JP, Korzekwa KR. (2013). Predicting intrinsic clearance for drugs and drug candidates metabolized by aldehyde oxidase. Mol Pharm 10:1262–68.
  • Kamli MR, Kim J, Pokharel S, et al. (2014). Expressional studies of the aldehyde oxidase, AOX1) gene during myogenic differentiation in C2C12 cells. Biochem Biophys Res Commun 450:1291–96.
  • Kaye B, Offerman JL, Reid JL, et al. (1984). A species difference in the presystemic metabolism of carbazeran in dog and man. Xenobiotica 14:935–45.
  • Kaye B, Rance DJ, Waring L. (1985). Oxidative metabolism of carbazeran in vitro by liver cytosol of baboon and man. Xenobiotica 15:237–42.
  • Kimura K, Wakamatsu A, Suzuki Y, et al. (2005). Diversification of transcriptional modulation: large-scale identification and characterization of putative alternative promoters of human genes. Genome Res 16:55–65.
  • Kitamura S, Nakatani K, Ohashi K, et al. (2001). Extremely high drug-reductase activity based on aldehyde oxidase in monkey liver. Biol Pharm Bull 24:856–859.
  • Kitamura S, Nitta K, Tayama Y, et al. (2008). Aldehyde oxidase-catalyzed metabolism of N-1-methylnicotinamide in vivo and in vitro in chimeric mice with humanized liver. Drug Metab Dispos 36:1202–1205.
  • Kitamura S, Sugihara K, Ohta S. (2006). Drug-metabolizing ability of molybdenum hydroxylases. Drug Metab Pharmacok 21:83–98.
  • Kitamura S, Suzuki T, Kadota T, et al. (2003). In vitro metabolism of fenthion and fenthion sulfoxide by liver preparations of sea bream, goldfish, and rats. Drug Metab Dispos 31:179–186.
  • Knox WE. (1946). The quinine-oxidizing enzyme and liver aldehyde oxidase. J Biol Chem 163:699–711.
  • Konishi K, Fukami T, Gotoh S, et al. (2017). Identification of enzymes responsible for nitrazepam metabolism and toxicity in human. Biochem Pharmacol 140:150–160.
  • Kosel M, Amey M, Aubert A-C, Baumann P. (2001). In vitro metabolism of citalopram by monoamine oxidase B in human blood. Eur Neuropsychopharm 11:75–8.
  • Kosel M, Gnerre C, Voirol P, et al. (2002). In vitro biotransformation of the selective serotonin reuptake inhibitor citalopram, its enantiomers and demethylated metabolites by monoamine oxidase in rat and human brain preparations. Mol Psychiatry 7:181–8.
  • Kozminski K, Heyward S, Zientek M. (2019). Aldehyde oxidase activity in human vascular tissue and its potential contribution to extra-hepatic metabolism. Drug Metab Pharmacok 34:S62–S3.
  • Kratochwil NA, Meille C, Fowler S, et al. (2017). Metabolic profiling of human long-term liver models and hepatic clearance predictions from in vitro data using nonlinear mixed-effects modeling. AAPS J 19:534–50.
  • Kremer JI, Gömpel K, Bakuradze T, et al. (2018). Urinary excretion of niacin metabolites in humans after coffee consumption. Mol Nutr Food Res 62:e1700735.
  • Krenitsky TA, Neil SM, Elion GB, et al. (1972). A comparison of the specificities of xanthine oxidase and aldehyde oxidase. Arch Biochem Biophys 150:585–99.
  • Kücükgöze G, Terao M, Garattini E, Leimkühler S. (2017). Direct comparison of the enzymatic characteristics and superoxide production of the four aldehyde oxidase enzymes present in mouse. Drug Metab Dispos 45:947–55.
  • Kundu TK, Hille R, Velayutham M, et al. (2007). Characterization of superoxide production from aldehyde oxidase: an important source of oxidants in biological tissues. Arch Biochem Biophys 460:113–21.
  • Kurosaki M, Bolis M, Fratelli M, et al. (2013). Structure and evolution of vertebrate aldehyde oxidases: from gene duplication to gene suppression. Cell Mol Life Sci 70:1807–30.
  • Kurzawski M, Dziewanowski K, Safranow K, Drozdzik M. (2012). Polymorphism of genes involved in purine metabolism (XDH, AOX1, MOCOS) in kidney transplant recipients receiving azathioprine. Ther Drug Monit 34:266–274.
  • Lee KWK, Pausova Z. (2013). Cigarette smoking and DNA methylation. Front Genet 4:132.
  • Lenglet A, Liabeuf S, Bodeau S, et al. (2016). N-methyl-2-pyridone-5-carboxamide (2PY)-major metabolite of nicotinamide: an update on an old uremic toxin. Toxins 8:339.
  • Leoni C, Buratti FM, Testai E. (2008). The participation of human hepatic P450 isoforms, flavin-containing monooxygenases and aldehyde oxidase in the biotransformation of the insecticide fenthion. Toxicol Appl Pharm 233:343–52.
  • Lepri S, Ceccarelli M, Milani N, et al. (2017). Structure-metabolism relationships in human-AOX: chemical insights from a large database of aza-aromatic and amide compounds. Proc Natl Acad Sci USA 114:E3178.
  • Li AC, Cui D, Yu E, et al. (2019). Identification and human exposure prediction of two aldehyde oxidase-mediated metabolites of a methylquinoline-containing drug candidate. Xenobiotica 49:302–12.
  • Li Y, Lai WG, Whitcher-Johnstone A, et al. (2012a). Metabolic switching of BILR 355 in the presence of ritonavir. I. Identifying an unexpected disproportionate human metabolite. Drug Metab Dispos 40:1122–9.
  • Liu L, Halladay JS, Shin Y, et al. (2011). Significant species difference in amide hydrolysis of GDC-0834, a novel potent and selective Brutons tyrosine kinase inhibitor. Drug Metab Dispos 39:1840–49.
  • Li Y, Xu J, Lai WG, et al. (2012b). Metabolic switching of BILR 355 in the presence of ritonavir II: uncovering novel contributions by gut bacteria and aldehyde oxidase. Drug Metab Dispos 40:1130–37.
  • Lolkema M, Bohets HH, Arkenau H, et al. (2015). The c-met tyrosine kinase inhibitor JNJ-38877605 causes renal toxicity through species-specific insoluble metabolite formation. Clin Cancer Res 21:2297–304.
  • Long L, Dolan ME. (2001). Role of cytochrome P450 isoenzymes in metabolism of O6-benzylguanine: implications for dacarbazine activation. Clin Cancer Res 7:4239–44.
  • Lu K, Alcivar AL, Ma J, et al. (2017). NRF2 induction supporting breast cancer cell survival is enabled by oxidative stress-induced DPP3-KEAP1 interaction. Cancer Res 77:2881–92.
  • Maeda K, Ohno T, Igarashi S, et al. (2012). Aldehyde oxidase 1 gene is regulated by Nrf2 pathway. Gene 505:374–8.
  • Maia LB, Moura JJG. (2018). Putting xanthine oxidoreductase and aldehyde oxidase on the NO metabolism map: nitrite reduction by molybdoenzymes. Redox Biol 19:274–89.
  • Maia LB, Pereira V, Mira L, Moura JJG. (2015). Nitrite reductase activity of rat and human xanthine oxidase, xanthine dehydrogenase, and aldehyde oxidase: evaluation of their contribution to NO formation in vivo. Biochemistry 54:685–710.
  • Manevski N, Balavenkatraman KK, Bertschi B, et al. (2014). Aldehyde oxidase activity in fresh human skin. Drug Metab Dispos 42:2049–57.
  • Mao Z, Wu Y, Li Q, et al. (2018). Aldehyde oxidase-dependent species difference in hepatic metabolism of fasudil to hydroxyfasudil. Xenobiotica 48:170–7.
  • Mirzaei S, Taherpour AA, Mohamadi S. (2016). Mechanistic study of allopurinol oxidation using aldehyde oxidase, xanthine oxidase and cytochrome P450 enzymes. RSC Adv 6:109672–80.
  • Monick MM, Beach SRH, Plume J, et al. (2012). Coordinated changes in AHRR methylation in lymphoblasts and pulmonary macrophages from smokers. Am J Med Genet B 159B:141–51.
  • Montefiori M, Jørgensen FS, Olsen L. (2017). Aldehyde oxidase: reaction mechanism and prediction of site of metabolism. ACS Omega 2:4237–44.
  • Moriwaki Y, Yamamoto T, Takahashi S, et al. (2001). Widespread cellular distribution of aldehyde oxidase in human tissues found by immunohistochemistry staining. Histol Histopathol 16:745–53.
  • Mota C, Coelho C, Leimkühler S, et al. (2018). Critical overview on the structure and metabolism of human aldehyde oxidase and its role in pharmacokinetics. Coord Chem Rev 368:35–59.
  • Mota C, Esmaeeli M, Coelho C, et al. (2019). Human aldehyde oxidase (hAOX1): structure determination of the moco-free form of the natural variant G1269R and biophysical studies of single nucleotide polymorphisms. FEBS Open Bio 9:925–34.
  • Mraz M, Hurba O, Bartl J, et al. (2015). Modern diagnostic approach to hereditary xanthinuria. Urolithiasis 43:61–7.
  • Naritomi Y, Sanoh S, Ohta S. (2019). Utility of chimeric mice with humanized liver for predicting human pharmacokinetics in drug discovery: comparison with in vitro–in vivo extrapolation and allometric scaling. Biol Pharm Bull 42:327–36.
  • Neumeier M, Weigert J, Schäffler A, et al. (2006). Aldehyde oxidase 1 is highly abundant in hepatic steatosis and is downregulated by adiponectin and fenofibric acid in hepatocytes in vitro. Biochem Bioph Res Co 350:731–5.
  • Nirogi R, Kandikere V, Palacharla RC, et al. (2014). Identification of a suitable and selective inhibitor towards aldehyde oxidase catalyzed reactions. Xenobiotica 44:197–204.
  • Nishimura M, Naito S. (2006). Tissue-specific mRNA expression profiles of human phase I metabolizing enzymes except for cytochrome P450 and phase II metabolizing enzymes. Drug Metab Pharmacok 21:357–74.
  • O’Hara F, Burns AC, Collins MR, et al. (2014). A simple litmus test for aldehyde oxidase metabolism of heteroarenes. J Med Chem 57:1616–20.
  • Oesch F, Fabian E, Landsiedel R. (2018). Xenobiotica-metabolizing enzymes in the skin of rat, mouse, pig, guinea pig, man, and in human skin models. Arch Toxicol 92:2411–56.
  • Ogiso T, Fukami T, Mishiro K, et al. (2018). Substrate selectivity of human aldehyde oxidase 1 in reduction of nitroaromatic drugs. Arch Biochem Biophys 659:85–92.
  • Øster B, Thorsen K, Lamy P, et al. (2011). Identification and validation of highly frequent CpG island hypermethylation in colorectal adenomas and carcinomas. Int J Cancer 129:2855–66.
  • Otwell CJ, Woodworth Z, Buckley D. (2013). International Society for the Study of Xenobiotics, Washington, DC. 10th International ISSX Meeting; 2013, Toronto, Ontario, Canada.
  • Paragas EM, Humphreys SC, Min J, et al. (2017). The two faces of aldehyde oxidase: oxidative and reductive transformations of 5-nitroquinoline. Biochem Pharmacol 145:210–7.
  • Payes B, Greenberg DM. (1968). Studies on the enzymic decomposition of urocanic acid: VII. Identification of the enzyme catalyzing the oxidation of 4(5)-imidazolone-5(4)-propionic acid as an aldehyde oxidase. Arch Biochem Biophys 125:911–7.
  • Peretz H, Naamati MS, Levartovsky D, et al. (2007). Identification and characterization of the first mutation (Arg776Cys) in the C-terminal domain of the human molybdenum cofactor sulfurase, HMCS) associated with type II classical xanthinuria. Mol Genet Metab 91:23–9.
  • Peretz H, Watson DG, Blackburn G, et al. (2012). Urine metabolomics reveals novel physiologic functions of human aldehyde oxidase and provides biomarkers for typing xanthinuria. Metabolomics 8:951–9.
  • Propper D, Jones K, Anthoney DA, et al. (2016). Phase II study of TP300 in patients with advanced gastric or gastro-oesophageal junction adenocarcinoma. BMC Cancer 16:779.
  • Pryde DC, Dalvie D, Hu Q, et al. (2010). Aldehyde oxidase: an enzyme of emerging importance in drug discovery. J Med Chem 53:8441–60.
  • Ramanathan S, Jin F, Sharma S, et al. (2016). Clinical pharmacokinetic and pharmacodynamic profile of idelalisib. Clin Pharmacokinet 55:33–45.
  • Ramírez J, Kim TW, Liu W, et al. (2014). A pharmacogenetic study of aldehyde oxidase I in patients treated with XK469. Pharmacogenet Genom 24:129–32.
  • Rivera SP, Choi HH, Chapman B, et al. (2005). Identification of aldehyde oxidase 1 and aldehyde oxidase homologue 1 as dioxin-inducible genes. Toxicology 207:401–9.
  • Roberts R, Zhang M, Marinaki A, et al. (2010). Does genetic variability in aldehyde oxidase and molybdenum cofactor sulfurase predict nonresponse to allopurinol? Aliment Pharm Ther 32:310–1.
  • Rochat B, Kosel M, Boss G, et al. (1998). Stereoselective biotransformation of the selective serotonin reuptake inhibitor citalopram and its demethylated metabolites by monoamine oxidases in human liver. Biochem Pharmacol 56:15–23.
  • Romão MJ, Coelho C, Santos-Silva T, et al. (2017). Structural basis for the role of mammalian aldehyde oxidases in the metabolism of drugs and xenobiotics. Curr Opin Chem Biol 37:39–47.
  • Roy SK, Korzekwa KR, Gonzalez FJ, et al. (1995). Human liver oxidative metabolism of O6−benzylguanine. Biochem Pharmacol 50:1385–9.
  • Ruenitz PC, Bai X. (1995). Acidic metabolites of tamoxifen. Aspects of formation and fate in the female rat. Drug Metab Dispos 23:993–8.
  • Sadler NC, Nandhikonda P, Webb-Robertson BJ, et al. (2016). Hepatic cytochrome P450 activity, abundance, and expression throughout human development. Drug Metab Dispos 44:984–91.
  • Sangkuhl K, Klein TE, Altman RB. (2011). PharmGKB summary: citalopram pharmacokinetics pathway. Pharmacogenet Genom 21:769–72.
  • Sanoh S, Horiguchi A, Sugihara K, et al. (2012a). Prediction of in vivo hepatic clearance and half-life of drug candidates in human using chimeric mice with humanized liver. Drug Metab Dispos 40:322–8.
  • Sanoh S, Nozaki K, Murai H, et al. (2012b). Prediction of human metabolism of FK3453 by aldehyde oxidase using chimeric mice transplanted with human or rat hepatocytes. Drug Metab Dispos 40:76–82.
  • Sanoh S, Ohta S. (2014). Chimeric mice transplanted with human hepatocytes as a model for prediction of human drug metabolism and pharmacokinetics. Biopharm Drug Dispos 35:71–86.
  • Sanoh S, Tayama Y, Sugihara K, et al. (2015). Significance of aldehyde oxidase during drug development: effects on drug metabolism, pharmacokinetics, toxicity, and efficacy. Drug Metab Pharmacok 30:52–63.
  • Sebesta I, Stiburkova B, Krijt J. (2018). Hereditary xanthinuria is not so rare disorder of purine metabolism. Nucleos Nucleot Nucl 37:324–8.
  • Shui IM, Wong CJ, Zhao S, et al. (2016). Prostate tumor DNA methylation is associated with cigarette smoking and adverse prostate cancer outcomes. Cancer 122:2168–77.
  • Sigruener A, Buechler C, Orso E, et al. (2007). Human aldehyde oxidase 1 interacts with ATP-binding cassette transporter-1 and modulates its activity in hepatocytes. Horm Metab Res 39:781–9.
  • Simmonds HA, Reiter S, Nishino T. (1995). Hereditary xanthinuria, In: Scriver, C.R, ed. The metabolic and molecular basis of inherited disease. New York: McGraw-Hill, 137–41.
  • Slominska EM, Rutkowski P, Smolenski RT, et al. (2004). The age-related increase in N-methyl-2-pyridone-5-carboxamide (NAD catabolite) in human plasma. Mol Cell Biochem 267:25–30.
  • Smith M. A, Marinaki T, Ansari A, et al. (2008). Common polymorphism in the aldehyde oxidase gene is a marker of non-response to azathioprine therapy in inflammatory bowel disease. Annual general meeting of the British-society-of-gastroenterology, 10–13 March, Birmingham, England. Gut 57:094.
  • Sodhi JK, Wong S, Kirkpatrick DS, et al. (2015). A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos 43:908–15.
  • Stanulović M, Chaykin S. (1971). Aldehyde oxidase: catalysis of the oxidation of N 1 -methylnicotinamide and pyridoxal. Arch Biochem Biophys 145:27–34.
  • Stavrinou P, Mavrogiorgou MC, Polyzoidis K, et al. (2015). Expression profile of genes related to drug metabolism in human brain tumors. PLoS One 10:e0143285.
  • Stiburkova B, Pavelcova K, Petru L, et al. (2018). Thiopurine-induced toxicity is associated with dysfunction variant of the human molybdenum cofactor sulfurase gene (xanthinuria type II). Toxicol Appl Pharm 353:102–8.
  • Stock W, Undevia SD, Bivins C, et al. (2008). A phase I and pharmacokinetic study of XK469R, NSC 698215), a quinoxaline phenoxypropionic acid derivative, in patients with refractory acute leukemia. Invest New Drugs 26:331–8.
  • Strelevitz TJ, Orozco CC, Obach RS. (2012). Hydralazine as a selective probe inactivator of aldehyde oxidase in human hepatocytes: estimation of the contribution of aldehyde oxidase to metabolic clearance. Drug Metab Dispos 40:1441–8.
  • Suzuki Y, Shibuya M, Satoh Si, et al. (2008). Safety and efficacy of fasudil monotherapy and fasudil-ozagrel combination therapy in patients with subarachnoid hemorrhage: sub-analysis of the post-marketing surveillance study. Neurol Med Chir 48:241–8.
  • Swenson TL, Casida JE. (2013). Aldehyde oxidase importance in vivo in xenobiotic metabolism: imidacloprid nitroreduction in mice. Toxicol Sci 133:22–8.
  • Takaoka N, Sanoh S, Okuda K, et al. (2018). Inhibitory effects of drugs on the metabolic activity of mouse and human aldehyde oxidases and influence on drug–drug interactions. Biochem Pharmacol 154:28–38.
  • Takasaki W, Yamamura M, Nozaki A, et al. (2005). Stereoselective pharmacokinetics of RS‐8359, a selective and reversible MAO‐A inhibitor, by species‐dependent drug‐metabolizing enzymes. Chirality 17:135–41.
  • Tanoue C, Sugihara K, Uramaru N, et al. (2013). Prediction of human metabolism of the sedative-hypnotic zaleplon using chimeric mice transplanted with human hepatocytes. Xenobiotica 43:956–62.
  • Tayama Y, Miyake K, Sugihara K, et al. (2007). Developmental changes of aldehyde oxidase activity in young Japanese children. Clin Pharmaco Ther 81:567–72.
  • Tayama Y, Sugihara K, Sanoh S, et al. (2012). Developmental changes of aldehyde oxidase activity and protein expression in human liver cytosol. Drug Metab Pharmacokinet 27:543–7.
  • Taylor SM, Stubley-Beedham C, Stell JG. (1984). Simultaneous formation of 2- and 4-quinolones from quinolinium cations catalysed by aldehyde oxidase. Biochem J 220:67–74.
  • Terao M, Kurosaki M, Barzago MM, et al. (2006). Avian and canine aldehyde oxidases. Novel insights into the biology and evolution of molybdo-flavoenzyme. J Biol Chem 281:19748–61.
  • Terao M, Kurosaki M, Barzago MM, et al. (2009). Role of the molybdoflavoenzyme aldehyde oxidase homolog 2 in the biosynthesis of retinoic acid: generation and characterization of a knockout mouse. Mol Cell Biol 29:357–77.
  • Terao M, Romão MJ, Leimkühler S, et al. (2016). Structure and function of mammalian aldehyde oxidases. Arch Toxicol 90:753–80.
  • Tomizawa M, Casida JE. (2000). Imidacloprid, thiacloprid, and their imine derivatives up-regulate the alpha 4 beta 2 nicotinic acetylcholine receptor in M10 cells. Toxicol Appl Pharm 169:114–29.
  • Torres RA, Korzekwa KR, McMasters DR, et al. (2007). Use of density functional calculations to predict the regioselectivity of drugs and molecules metabolized by aldehyde oxidase. J Med Chem 50:4642–7.
  • Undevia SD, Innocenti F, Ramirez J, et al. (2008). A phase I and pharmacokinetic study of the quinoxaline antitumour agent R(+)XK469 in patients with advanced solid tumours. Eur J Cancer 44:1684–92.
  • van Eijl S, Zhu Z, Cupitt J, et al. (2012). Elucidation of xenobiotic metabolism pathways in human skin and human skin models by proteomic profiling. PLoS One 7:e41721.
  • Vanholder R, De Smet R, Glorieux G, et al. (2003). Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 63:1934–43.
  • Vardavas AI, Ozcagli E, Fragkiadaki P, et al. (2018). The metabolism of imidacloprid by aldehyde oxidase contributes to its clastogenic effect in New Zealand rabbits. Mutat Res 829–830:26–32.
  • Vardavas A, Tzatzarakis M, Stivaktakis P, et al. (2017). Aldehyde oxidase metabolism route inhibition via sodium tungstate in imidacloprid exposed rabbits. Toxicol Lett 280:S253.
  • Varisli L. (2013). Identification of new genes downregulated in prostate cancer and investigation of their effects on prognosis. Genet Test Mol Bioma 17:562–6.
  • Waldron M, Winter A, Hill BT. (2017). Pharmacokinetic and pharmacodynamic considerations in the treatment of chronic lymphocytic leukemia: ibrutinib, idelalisib, and venetoclax. Clin Pharmacokinet 56:1255–66.
  • Wang X, Anadón A, Wu Q, et al. (2018). Mechanism of neonicotinoid toxicity: impact on oxidative stress and metabolism. Annu Rev Pharmacol Toxicol 58:471–507.
  • Wanwimolruk S, Wong SM, Zhang HU, et al. (1995). Metabolism of quinine in man: identification of a major metabolite, and effects of smoking and rifampicin pretreatment. J Pharm Pharmacol 47:957–63.
  • Weigert J, Neumeier M, Bauer S, et al. (2008). Small-interference RNA-mediated knock-down of aldehyde oxidase 1 in 3T3-L1 cells impairs adipogenesis and adiponectin release. FEBS Lett 582:2965–72.
  • Wilkinson DJ, Southall RL, Li M, et al. (2017). Minipig and human metabolism of aldehyde oxidase substrates: in vitro-in vivo comparisons. AAPS J 19:1163–74.
  • Wright RM, Vaitaitis GM, Weigel LK, et al. (1995). Identification of the candidate ALS2 gene at chromosome 2q33 as a human aldehyde oxidase gene. Redox Rept 1:313–21.
  • Xie J, Saburulla NF, Chen S, et al. (2019). Evaluation of carbazeran 4-oxidation and O6-benzylguanine 8-oxidation as catalytic markers of human aldehyde oxidase: impact of cytosolic contamination of liver microsomes. Drug Metab Dispos 47:26–37.
  • Xu Y, Li L, Wang Y, et al. (2017). Aldehyde oxidase mediated metabolism in drug-like molecules: a combined computational and experimental study. J Med Chem 60:2973–82.
  • Yagi K, Akagi K, Hayashi H, et al. (2010). Three DNA methylation epigenotypes in human colorectal cancer. Clin Cancer Res 16:21–33.
  • Yang X, Johnson N, Di L. (2019). Evaluation of cytochrome P450 selectivity for hydralazine as an aldehyde oxidase inhibitor for reaction phenotyping. J Pharm Sci 108:1627–30.
  • Zhang X, Liu H, Weller P, et al. (2011). In silico and in vitro pharmacogenetics: aldehyde oxidase rapidly metabolizes a p38 kinase inhibitor. Pharmacogenomics J 11:15–24.
  • Zhang JW, Xiao W, Gao ZT, et al. (2018). Metabolism of c-met kinase inhibitors containing quinoline by aldehyde oxidase, electron donating and steric hindrance effect. Drug Metab Dispos 46:1847–55.
  • Zhou L, Satonin D, Chappell J, et al. (2019). Elimination of [14C] LY3023414 by aldehyde oxidase and CYP enzymes in humans following oral administration. Drug Metab Pharmacok 34:S63.
  • Zhou Y, Zhang X, Ding R, et al. (2015). Using next-generation sequencing to identify a mutation in human MCSU that is responsible for type II xanthinuria. Cell Physiol Biochem 35:2412–21.
  • Zientek MA, Youdim K. (2014). Reaction phenotyping: advances in the experimental strategies used to characterize the contribution of drug-metabolizing enzymes. Drug Metab Dispos 43:163–81.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.