Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 50, 2020 - Issue 1
497
Views
21
CrossRef citations to date
0
Altmetric
Review Articles

Uridine diphosphate glucuronosyltransferase 1A1

ORCID Icon
Pages 64-76 | Received 26 Mar 2019, Accepted 08 May 2019, Published online: 03 Jun 2019

References

  • Abang AM. (1998). The clinical pharmacology of topoisomerase I inhibitors. Semin. Hematol 35:13–21.
  • AIDS Info Guidelines. (2019) Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents. Department of Health and Human Services. http://www.aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf>. Accessed on March 4th, 2019.
  • Atasilp C, Chansriwong P, Sirachainan E, et al. (2016). Correlation of UGT1A1(*)28 and (*)6 polymorphisms with irinotecan‑induced neutropenia in Thai colorectal cancer patients. Drug Metab Pharmacokinet 31:90–4.
  • Babu SR, Lakshmi VM, Huang G-W, et al. (1996). Glucuronide conjugates of 4-aminobiphenyl and its N-hydroxy metabolites. pH stability and synthesis by human and dog liver. Biochem Pharmacol 51:1679–85.
  • Bajro MH, Josifovski T, Panovski M, et al. (2012). Promoter length polymorphism in UGT1A1 and the risk of sporadic colorectal cancer. Cancer Genet 205:163–7.
  • Banerjee SS, Aher N, Patil R, Khandare J. (2012). Poly(ethylene glycol)-Prodrug conjugates: concept, design, and applications. J Drug Deliv 2012:103973
  • Barbarino JM, Haidar CE, Klein TE, Altman RB. (2014). PharmGKB summary: Very important pharmacogene information for UGT1A1. Pharmacogenet Genomics 24:177–83.
  • Barua AB, Olson JA. (1987). Chemical synthesis and growth-promoting activity of all-trans-retinyl beta-D-glucuronide. Biochem J 244:231–4.
  • Bauer TM, Ritz R, Haberthur C, et al. (1995). Prolonged sedation due to accumulation of conjugated metabolites of midazolam. Lancet 346:145–7.
  • Bernabeu I, Cameselle‑Teijeiro J, Casanueva FF, Marazuela M. (2009). Pegvisomant‑induced cholestatic hepatitis with jaundice in a patient with Gilbert's syndrome. Eur J Endocrinol 160:869–72.
  • Bernabeu I, Marazuela M, Lucas T, et al. (2010). Pegvisomant‑induced liver injury is related to the UGT1A1*28 polymorphism of Gilbert's syndrome. J Clin Endocrinol Metab 95:2147–54.
  • Beutler E, Gelbart T, Demina A. (1998). Racial variability in the UDP‑glucuronosyltransferase 1 (UGT1A1) promoter: A balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci 95:8170–4.
  • Bock KW. (1991). Roles of UDP-glucuronosyltransferases in chemical carcinogenesis. Crit Rev Biochem Mol Biol 26:129–50.
  • Boelsterli UA, Zimmerman HJ, Kretz-Rommel A. (1995). Idiosyncratic liver toxicity of nonsteroidal antiinflammatory drugs: molecular mechanisms and pathology. Crit Rev Toxicol 25:207–35.
  • Bosma PJ. (2003). Inherited disorders of bilirubin metabolism. J Hepatol 38:107–17.
  • Bosma PJ, Chowdhury JR, Bakker C, et al. (1995). The genetic basis of the reduced expression of bilirubin UDP‑glucuronosyltransferase 1 in Gilbert's syndrome. N Engl J Med 333:1171–5.
  • Bosma PJ, Chowdhury NR, Goldhoorn BG, et al. (1992). Sequence of exons and the flanking regions of human bilirubin‑UDP‑glucuronosyltransferase gene complex and identification of a genetic mutation in a patient with Crigler‑Najjar syndrome, type I. Hepatology 15:941–7.
  • Bosma PJ, Goldhoorn B, Oude Elferink RP, et al. (1993). A mutation in bilirubin uridine 5'-diphosphate-glucuronosyltransferase isoform 1 causing Crigler-Najjar syndrome type II. Gastroenterology 105:216–20.
  • Bosma PJ, Seppen J, Goldhoorn B, et al. (1994). Bilirubin UDP-glucuronosyltransferase 1 is the only relevant bilirubin glucuronidating isoform in man. J Biol Chem 269:17960–4.
  • Braun MS, Richman SD, Thompson L, et al. (2009). Association of molecular markers with toxicity outcomes in a randomized trial of chemotherapy for advanced colorectal cancer: the FOCUS trial. J Clin Oncol 27:5519–28.
  • Buchfelder M, Schlaffer S, Droste M, et al. (2009). German Pegvisomant Observational Study: The German ACROSTUDY: Past and present. Eur J Endocrinol 161: S3–S10.
  • Canu G, Minucci A, Zuppi C, Capoluongo E. (2013). Gilbert and Crigler Najjar syndromes: An update of the UDP‑glucuronosyltransferase 1A1 (UGT1A1) gene mutation database. Blood Cells Mol Dis 50:273–80.
  • Chen K, Jin M, Zhu Y, et al. (2006). Genetic polymorphisms of the uridine diphosphate glucuronosyltransferase 1A7 and colorectal cancer risk in relation to cigarette smoking and alcohol drinking in a Chinese population. J Gastroenterol Hepatol 21:1036–41.
  • Choe PG, Park WB, Song JS, et al. (2010). Incidence of atazanavir‑associated hyperbilirubinemia in Korean HIV patients: 30 months follow‑up results in a population with low UDP‑glucuronosyltransferase1A1*28 allele frequency. J Korean Med Sci 25:1427–30.
  • Ciotti M, Werlin SL, Owens IS. (1999). Delayed response to phenobarbital treatment of a Crigler‑Najjar type II patient with partially inactivating missense mutations in the bilirubin UDP‑glucuronosyltransferase gene. J Pediatr Gastroenterol Nutr 28:210–3.
  • Clarke DJ, Burchell B. (1994) Conjugation-deconjugation reactions in drug metabolism and toxicity, Vol 112, (Kauffman FC, Ed.) Berlin, Germany, Springer-Verlag, 3–43.
  • Cote JF, Kirzin S, Kramar A, et al. (2007). UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res 13:3269–75.
  • Daar ES, Tierney C, Fischl MA, et al. (2011). Atazanavir plus ritonavir or efavirenz as part of a 3-drug regimen for initial treatment of HIV-1. Ann Intern Med 154:445–56.
  • Dean L. (2018) Irinotecan Therapy and UGT1A1 Genotype. In: Medical Genetics Summaries (Eds Pratt V, McLeod H, Rubinstein W, Dean L, Kattman B, Adriana Malheiro A.), Bethesda, MD: National Center for Biotechnology Information. Available at: https://www.ncbi.nlm.nih.gov/books/NBK294473/. Accessed 4th March, 2019.
  • Dickinson RG, Hooper WD, Eadie MJ. (1984). pH-dependent rearrangement of the biosynthetic ester glucuronide of valproic acid to beta-glucuronidase-resistant forms. Drug Metab Disp 12:247–52.
  • Ding A, Ojingwa JC, McDonagh AF, et al. (1993). Evidence for covalent binding of acyl glucuronides to serum albumin via an imine mechanism as revealed by tandem mass spectrometry. Proc Natl Acad Sci USA 90:3797–801.
  • Ding A, Zia-Amirhosseini P, McDonagh AF, et al. (1995). Reactivity of tolmetin glucuronide with human serum albumin. Identification of binding sites and mechanisms of reaction by tandem mass spectrometry. Drug Metab Disp 23:369–76.
  • Ebner T, Burchell B. (1993). Substrate specificities of two stably expressed human liver UDP-glucuronosyltransferases of the UGT1 gene family. Drug Metab Disp 21:50–5.
  • Eggers NJ, Doust K. (1981). Isolation and identification of probenecid acyl glucuronide. J Pharm Pharmacol 33:123–4.
  • Eskandari-Nasab E, Hashemi M, Rezaei H, et al. (2012). Evaluation of UDP‑glucuronosyltransferase 2B17 (UGT2B17) and dihydrofolate reductase (DHFR) genes deletion and the expression level of NGX6 mRNA in breast cancer. Mol Biol Rep 39:10531–9.
  • Fang JL, Lazarus P. (2004). Correlation between the UDP‑glucuronosyltransferase (UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype: significantly decreased glucuronidating activity against benzo(a)pyrene‑7,8‑dihydrodiol(‑) in liver microsomes from subjects with the UGT1A1*28 variant. Cancer Epidemiol Biomarkers Prev 13:102–9.
  • FDA. (2013). Clinical Pharmacogenomics: Premarketing Evaluation in Early-Phase Clinical Studies and Recommendations for Labeling, Available at: https://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM337169.pdf. Accessed 5th, March 2019.
  • FDA. (2019). Table of Pharmacogenomic Biomarkers in Drug Labeling, Available at: https://www.fda.gov/Drugs/ScienceResearch/ucm572698.htm. Accessed March 5th, 2019.
  • Fenselau C. (1994) Conjugation-deconjugation reactions in drug metabolism and toxicity, Vol. 112, (Kauffman FC, Ed.) Berlin, Germany: Springer-Verlag, 367–389.
  • Ferraldeschi R, Minchell LJ, Roberts SA, et al. (2009). UGT1A1*28 genotype predicts gastrointestinal toxicity in patients treated with intermediate-dose irinotecan. Pharmacogenomics 10:733–9.
  • Filopanti M, Barbieri AM, Mantovani G, et al. (2014). Role of UGT1A1 and ADH gene polymorphisms in pegvisomant‑induced liver toxicity in acromegalic patients. Eur J Endocrinol 170:247–54.
  • Fournel-Gigleux S, Hamar-Hansen C, Motassim N, et al. (1988). Substrate specificity and enantioselectivity of arylcarboxylic acid glucuronidation. Drug Metab Dispos 16:627–34.
  • Frances B, Gout R, Monsarrat B, et al. (1992). Further evidence that morphine-6 beta-glucuronide is a more potent opioid agonist than morphine. J Pharmacol Exp Ther 262:25–30.
  • Gallant JE, Koenig E, Andrade-Villanueva J, et al. (2013). Cobicistat versus ritonavir as a pharmacoenhancer of atazanavir plus emtricitabine/tenofovir disoproxil fumarate in treatment-naive HIV type 1-infected patients: week 48 results. J Infect Dis 208:32–9.
  • Gammal RS, Court MH, Haidar CE, Iwuchukwu OF, et al. (2016). Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and Atazanavir Prescribing. Clin Pharmacol Therap 99:363–9.
  • Girard H, Lévesque E, Bellemare J, et al. (2007). Genetic diversity at the UGT1 locus is amplified by a novel 3' alternative splicing mechanism leading to nine additional UGT1A proteins that act as regulators of glucuronidation activity. Pharmacogenet Genomics 17:1077–89.
  • Giustina A, Ambrosio MR, Beck Peccoz P, et al. (2014). Use of Pegvisomant in acromegaly. An Italian society of endocrinology guideline. J Endocrinol Invest 37:1017–30.
  • Glimelius B, Garmo H, Berglund A, et al. (2011). Prediction of irinotecan and 5‑fluorouracil toxicity and response in patients with advanced colorectal cancer. Pharmacogenomics J 11:61–71.
  • Goey AK, Sissung TM, Peer CJ, et al. (2016). Effects of UGT1A1 genotype on the pharmacokinetics, pharmacodynamics and toxicities of belinostat administered by 48 h continuous infusion in patients with cancer. J Clin Pharmacol 56:461–73.
  • Goldman JM. (2007). How I treat chronic myeloid leukemia in the imatinib era. Blood 110:2828–37.
  • Green MD, King CD, Mojarrabi B, et al. (1998). Glucuronidation of amines and other xenobiotics catalyzed by expressed human UDP-glucuronosyltransferase 1A3. Drug Metab Disp 26:507–12.
  • Guillemette C. (2003). Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J 3:136–58.
  • Guillemette C, Bélanger A, Lépine J. (2004). Metabolic inactivation of estrogens in breast tissue by UDP‑glucuronosyltransferase enzymes: An overview. Breast Cancer Res 6:246–54.
  • Hoskins JM, Goldberg RM, Qu P, et al. (2007). UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst 99:1290–5.
  • Hu ZY, Yu Q, Pei Q, Guo C. (2010). Dose-dependent association betweenUGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Clin Cancer Res 2010:3832–42.
  • Huangfu H, Pan H, Wang B, et al. (2016). Association between UGT1A1 polymorphism and risk of laryngeal squamous cell carcinoma. Int J Environ Res Public Health 13:112.
  • Jancova P, Anzenbacher P, Anzenbacherova E. (2010). Phase II drug metabolizing enzymes. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 154:103–16.
  • Jin C, Miners JO, Lillywhite KJ, et al. (1993). Complementary deoxyribonucleic acid cloning and expression of a human liver uridine diphosphate-glucuronosyltransferase glucuronidating carboxylic acid-containing drugs. J Pharmacol Exp Ther 264:475–9.
  • Jinno H, Tanaka‑Kagawa T, Hanioka N, et al. (2003). Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Drug Metab Disp 31:108–13.
  • Kantarjian H, Shah NP, Hochhaus A, et al. (2010). Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 362:2260–70.
  • Kaplan M, Hammerman C, Maisels MJ. (2003). Bilirubin genetics for the nongeneticist: Hereditary defects of neonatal bilirubin conjugation. Pediatrics 111:886–93.
  • Köhle C, Möhrle B, Münzel PA, et al. (2003). Frequent co‑occurrence of the TATA box mutation associated with Gilbert's syndrome (UGT1A1*28) with other polymorphisms of the UDP‑glucuronosyltransferase‑1 locus (UGT1A6*2 and UGT1A7*3) in Caucasians and Egyptians. Biochem Pharmacol 65:1521–7.
  • Kweekel DM, Gelderblom H, Van der Straaten T, et al. (2008). UGT1A1*28 genotype and irinotecan dosage in patients with metastatic colorectal cancer: a Dutch Colorectal Cancer Group study. Br J Cancer 99:275–82.
  • Lamas MJ, Duran G, Balboa E, et al. (2012). The value of genetic polymorphisms to predict toxicity in metastatic colorectal patients with irinotecan-based regimens. Cancer Chemother Pharmacol 69:1591–9.
  • Lankisch TO, Moebius U, Wehmeier M, et al. (2006). Gilbert's disease and atazanavir: from phenotype to UDP‑glucuronosyltransferase haplotype. Hepatology 44:1324–32.
  • Lavelle F, Bissery MC, Andre S, et al. (1996). Preclinical evaluation of CPT-11 and its active metabolite SN-38. Semin Oncol 23:11–20.
  • Lennox JL, Landovitz RJ, Ribaudo HJ, et al. (2014). Efficacy and tolerability of 3 nonnucleoside reverse transcriptase inhibitor-sparing antiretroviral regimens for treatment-naive volunteers infected with HIV-1: a randomized, controlled equivalence trial. Ann Intern Med 161:461–71.
  • Liu X, Cheng D, Kuang Q, et al. (2014). Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Pharmacogenomics J 14:120–9.
  • Lodoso Torrecilla B, Palomo Atance E, Camarena Grande C, et al. (2006). Crigler‑Najjar syndrome: diagnosis and treatment. An Pediatr (Barc) 65:73–8.
  • Lowenberg D, Thorn CF, Whirl-Carrillo M, et al (2001) Irinotecan Pathway, Pharmacokinetics: Pharmacogenomics Knowledge Base (PharmGKB) and Stanford University. Available at: https://www.pharmgkb.org/pathway/PA2001. Accessed 21/02/2019.
  • Mackenzie PI, Bock KW, Burchell B, et al. (2005). Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 15:677–85.
  • Magdalou J, Chajes V, Lafaurie C, et al. (1990). Glucuronidation of 2-arylpropionic acids pirprofen, flurbiprofen, and ibuprofen by liver microsomes. Drug Metab Disp 18:692–7.
  • Maisels MJ. (2015). Managing the jaundiced newborn: A persistent challenge. CMAJ 187:335–43.
  • Malan DR, David KE, Wirtz N, et al. (2008). Efficacy and safety of atazanavir, with or without ritonavir, as part of once-daily highly active antiretroviral therapy regimens in antiretroviral-naive patients. J Acquir Immune Defic Syndr 47:161–7.
  • Mallea-Gil MS, Bernabeu I, et al. (2016). Pegvisomant‑induced cholestatic hepatitis in an acromegalic patient with UGT1A1*28 mutation. Case Rep Endocrinol 216:Article ID 2087102, Available at: http://dx.doi.org/10.1155/2016/2087102.
  • Marcuello E, Altés A, Menoyo A, et al. (2004). UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br J Cancer 91:678–82.
  • Marks PA, Richon VM, Rifkind RA. (2000). Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst 92:1210–6.
  • Marques SC, Ikediobi ON. (2010). The clinical application of UGT1A1pharmacogenetic testing: gene–environment interactions. Hum Genom 4:238–49.
  • Marsh S, Hoskins JM. (2010). Irinotecan pharmacogenomics. Pharmacogenomics 11:1003–10.
  • Martinez-Balibrea E, Abad A, Martínez-Cardús A, et al. (2010). UGT1A and TYMS genetic variants predict toxicity and response of colorectal cancer patients treated with first-line irinotecan and fluorouracil combination therapy. Br J Cancer 103:581–9.
  • Maruo Y, Nakahara S, Yanagi T, et al. (2016). Genotype of UGT1A1 and phenotype correlation between Crigler‑Najjar syndrome type II and Gilbert syndrome. J Gastroenterol Hepatol 31:403–8.
  • McLeod HL, Sargent DJ, Marsh S, et al. (2010). Pharmacogenetic predictors of adverse events and response to chemotherapy in metastatic colorectal cancer: results from North American Gastrointestinal Intergroup Trial N9741. J Clin Oncol 28:3227–33.
  • Meech R, Mackenzie PI. (1997). Structure and function of uridine diphosphate glucuronosyltransferases. Clin Exp Pharmacol Physiol 24:907–15.
  • Memon N, Weinberger BI, Hegyi T, et al. (2016). Inherited disorders of bilirubin clearance. Pediatr Res 79:378–86.
  • Miura M. (2015). Therapeutic drug monitoring of imatinib, nilotinib, and dasatinib for patients with chronic myeloid leukemia. Biol Pharm Bull 38:645–54.
  • Molina JM, Andrade-Villanueva J, Echevarria J, et al. (2008). Once-daily atazanavir/ritonavir versus twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 48 week efficacy and safety results of the CASTLE study. Lancet 372:646–55.
  • Mulder GJ, Coughtrie MWH, Burchell B. (1990) Conjugation reactions in drug metabolism. (Mulder GJ, Ed.) London, YK: Taylor and Francis, 51–105.
  • Nicholls AW, Akira K, Lindon JC, et al. (1996). NMR spectroscopic and theoretical chemistry studies on the internal acyl migration reactions of the 1-O-acyl-beta-D-glucopyranuronate conjugates of 2-, 3-, and 4-(trifluoromethyl) benzoic acids. Chem Res Toxicol 9:1414–24.
  • O’Brien SG, Guilhot F, Larson RA, et al. (2003). IRIS Investigators. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 348:994–1004.
  • Olson JA, Moon RC, Anders MW, et al. (1992). Enhancement of biological activity by conjugation reactions. J Nutr 122:615–24.
  • Osawa K. (2009). Gene polymorphisms and chemotherapy in Non-small Cell Lung Cancer. Zhongguo Fei Ai Za Zhi 12:837–40.
  • Osborne R, Thompson P, Joel S, et al. (1992). The analgesic activity of morphine-6-glucuronide. Br J Clin Pharmacol 34:130–8.
  • Park WB, Choe PG, Song KH, et al. (2010). Genetic factors influencing severe atazanavir‑associated hyperbilirubinemia in a population with low UDP‑glucuronosyltransferase 1A1*28 allele frequency. Clin Infect Dis 51:101–6.
  • Paulik A, Grim J, Filip S. (2012). Predictors of irinotecan toxicity and efficacy in treatment of metastatic colorectal cancer. Acta Medica (Hredac Kralove) 55:153–159.
  • Peer CJ, Goey AKL, Sissung TM, et al. (2016). UGT1A1 genotype-dependent dose adjustment of belinostat using population pharmacokinetic modelling and simulation in patients with small cell lung cancer and other advanced cancers. Clin Pharmacol Ther 56:450–60.
  • Petit F, Gajdos V, Capel L, et al. (2006). Crigler-Najjar type II syndrome may result from several types and combinations of mutations in the UGT1A1 gene. Clin Genet 69:525–7.
  • Phelip JM, Mineur L, De la Fouchardière C, et al. (2016). High resectability rate of initially unresectable colorectal liver metastases after UGT1A1‑adapted high‑dose irinotecan combined with LV5FU2 and cetuximab: A multicenter phase II study (ERBIFORT). Ann Surg Oncol 23:2161–6.
  • Pichereau S, Le Louarn A, Lecomte T, et al. (2010). Cost‑Effectiveness of UGT1A1*28 genotyping in preventing severe neutropenia following FOLFIRI therapy in colorectal cancer. J Pharm Sci 13:615–25.
  • Qui Y, Burlingame AL, Benet LZ. (1998). Mechanisms for covalent binding of benoxaprofen glucuronide to human serum albumin. Studies By tandem mass spectrometry. Drug Metab Disp 26:246–56.
  • Ramos-Leví AM, Bernabeu I, Sampedro-Núñez M, et al. (2016). Genetic predictors of response to different medical therapies in acromegaly. Prog Mol Biol Transl Sci 138:85–114.
  • Rodrigues C, Vieira E, Santos R, et al. (2012). Impact of UGT1A1 gene variants on total bilirubin levels in Gilbert syndrome patients and in healthy subjects. Blood Cells Mol Dis 48:166–72.
  • Rothenberg ML. (1996). The current status of irinotecan (CPT-11) in the United States. Ann N Y Acad Sci 803:272–81.
  • Rouits E, Boisdron-Celle M, Dumont A, et al. (2004). Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res 10:5151–9.
  • Saglio G, Kim DW, Issaragrisil S, et al. (2010). Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med 362:2251–9.
  • Sai K, Saito Y. (2011). Ethnic differences in the metabolism, toxicology and efficacy of three anticancer drugs. Expert Opin Drug Metab Toxicol 7:967–88.
  • Sallustio BC, Sabordo L, Evans AM, et al. (2000). Hepatic disposition of electrophilic acyl glucuronide conjugates. Cur Drug Metab 1:163–80.
  • Sallustio BC, Fairchild BA, Pannall PR. (1997). Interaction of human serum albumin with the electrophilic metabolite 1-O-gemfibrozil-β-D-glucuronide. Drug Metab Disp 25:55–60.
  • Sanchez‑Dominguez CN, Gallardo-Blanco HL, Salinas‑Santander MA, et al. (2018). Uridine 5'‑diphospho‑glucronosyltrasferase: Its role in pharmacogenomics and human disease (Review). Exp Therap Med 16:3–11.
  • Satoh T, Ura T, Yamada Y, et al. (2011). Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Cancer Sci 102:1868–73.
  • Seco ML, del Río E, Barcelo MJ, et al. (2002). [Interest in the study of genetic variants of the promoter region of the UGT1A1 gene in neonatal jaundice]. An Esp Pediatr 56:139–43.
  • Servedio V, d'Apolito M, Maiorano N, et al. (2005). Spectrum of UGT1A1 mutations in Crigler-Najjar (CN) syndrome patients: identification of twelve novel alleles and genotype-phenotype correlation . Hum Mutat 25:325
  • Shimomura K, Kamata O, Ueki S, et al. (1971). Analgesic effect of morphine glucuronides. Tohoku J Exp Med 105:45–52.
  • Shulman K, Cohen I, Barnett-Griness O, et al. (2011). Clinical implications of UGT1A1*28 genotype testing in colorectal cancer patients. Cancer 117:3156–62.
  • Singer JB, Shou Y, Giles F, et al. (2007). UGT1A1 promoter polymorphism increases risk of nilotinib-induced hyperbilirubinemia. Leukemia 21:2311–5.
  • Spahn-Langguth H, Benet LZ. (1992). Acyl glucuronides revisited: is the glucuronidation process a toxification as well as a detoxification mechanism? Drug Metab Rev 24:5–48.
  • Spectrum Pharmaceuticals Inc. (2014) Full prescribing information Beleodaq. Available from: Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/206256lbl.pdf.
  • Strassburg CP, Vogel A, Kneip S, et al. (2002). Polymorphisms of the human UDP-glucuronosyltransferase (UGT) 1A7 gene in colorectal cancer. Gut 50:851–6.
  • Swami U, Goel S, Mani S. (2013). Therapeutic targeting of CPT-11 induced diarrhea: a case for prophylaxis. Curr Drug Targets 14:777–97.
  • Thibaudeau J, Lépine J, Tojcic J, et al. (2006). Characterization of common UGT1A8, UGT1A9, and UGT2B7 variants with different capacities to inactivate mutagenic 4‑hydroxylated metabolites of estradiol and estrone. Cancer Res 66:125–33.
  • Thomas A, Rajan A, Szabo E, et al. (2014). A Phase I/II Trial of Belinostat in Combination with Cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study. Clin Cancer Res 20:5392–402.
  • Toffoli G, Cecchin E, Corona G, et al. (2006). The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. J Clin Oncol 24:3061–8.
  • Tourancheau A, Margaillan G, Rouleau M, et al. (2016). Unravelling the transcriptomic landscape of the major phase II UDP‑glucuronosyltransferase drug metabolizing pathway using targeted RNA sequencing. Pharmacogenomics J 16:60–70.
  • UGT allele web site. (2019). Available at: https://www.pharmacogenomics.pha.ulaval.ca/ugt-alleles-nomenclature. Accessed 4th March 2019.
  • Ushijima K, Kamura T, Tamura K, et al. (2013). Docetaxel/irinotecan combination chemotherapy in platinum/taxane-refractory and -resistant ovarian cancer: JGOG/WJGOG Intergroup Study. Int J Clin Oncol 18:126–31.
  • Vogel A, Kneip S, Barut A, et al. (2001). Genetic link of hepatocellular carcinoma with polymorphisms of the UDP-glucuronosyltransferase UGT1A7 gene. Gastroenterology 121:1136–‑44.
  • Vogel A, Ockenga J, Ehmer U, et al. (2002). Polymorphisms of the carcinogen detoxifying UDP‑glucuronosyltransferase UGT1A7 in proximal digestive tract cancer. Z Gastroenterol 40:497–502.
  • Vore M, Liu Y, Huang L. (1997). Cholestatic properties and hepatic transport of steroid glucuronides. Drug Metab Rev 29:183–203.
  • Wall ME. (1998). Camptothecin and taxol: discovery to clinic. Med Res Rev 8:299–314.
  • Wang LZ, Ramirez J, Yeo W, et al. (2013). Glucuronidation by UGT1A1 is the dominant pathway of the metabolic disposition of belinostat in liver cancer patients. PLoS One 8:e54522.
  • Wang Y, Zukowski K, Yamada H, et al. (1990). Production of urothelial tumors in the heterotopic bladder of rat by benzidine derivatives. Cancer Res 50:2868–71.
  • Wells DS, Janssen FW, Ruelius HW. (1987). Interactions between oxaprozin glucuronide and human serum albumin. Xenobiotica 17:1437–49.
  • Wells PG, Mackenzie PI, Chowdhury JR, et al. (2004). Glucuronidation and the UDP-glucuronosyltransferases in health and disease. Drug Metab Dispos 32:281–90.
  • Wen F, Li Q. (2016). Treatment dilemmas of cetuximab combined with chemotherapy for metastatic colorectal cancer. World J Gastroenterol 22:5332–41.
  • Wen Z, Tallman MN, Ali SY, et al. (2007). UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: Structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Drug Metab Dispos 35:371–80.
  • Whirl-Carrillo M, McDonagh EM, Hebert JM, et al. (2012). Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther 92:414–7.
  • Williams AM, Dickinson RG. (1994). Studies on the reactivity of acyl glucuronides-VI. Modulation of reversible and covalent interaction of diflunisal acyl glucuronide and its isomers with human plasma protein in vitro. Biochem Pharmacol 47:457–67.
  • Yang C, Liu Y, Xi WQ, et al. (2015). Relationship between UGT1A1*6/*28 polymorphism and severe toxicities in Chinese with pancreatic or biliary tract cancer treated with irinotecan-containg regimens. Drug Des Devel Ther 9:3677–3683.
  • Zheng Z, Park JY, Guillemette C, et al. (2001). Tobacco carcinogen‑detoxifying enzyme UGT1A7 and its association with orolaryngeal cancer risk. J Natl Cancer Inst 93:1411–8.
  • Zhou Y, Wang SN, Li H, et al. (2014). Quantitative trait analysis of polymorphisms in two bilirubin metabolism enzymes to physiologic bilirubin levels in Chinese newborns. J Pediatr 165:1154–60.
  • Zia-Amirhosseini P, Spahn-Langguth H, Benet LZ. (1994). Bioactivation by glucuronide-conjugate formation. Adv Pharmacol 27:385–97.
  • Zile MH, Cullum ME, Simpson RU, et al. (1987). Induction of differentiation of human promyelocytic leukemia cell line HL-60 by retinoyl glucuronide, a biologically active metabolite of vitamin A. Proc Natl Acad Sci USA 84:2208–12.

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.