Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 50, 2020 - Issue 1
771
Views
19
CrossRef citations to date
0
Altmetric
Review Articles

Reducing risk in thiopurine therapy

&
Pages 101-109 | Received 21 Oct 2019, Accepted 30 Oct 2019, Published online: 12 Nov 2019

References

  • Alves S, Amorim A, Ferreira F, Prata MJ. (2001). Influence of the variable number of tandem repeats located in the promoter region of the thiopurine methyltransferase gene on enzymatic activity. Clin Pharmacol Ther 70:165–74.
  • Alves S, Rocha J, Amorim A, Prata MJ. (2004). Tracing the origin of the most common thiopurine methyltransferase (TPMT) variants: preliminary data from the patterns of haplotypic association with two CA repeats. Ann Hum Genet 68:313–23.
  • Ansari A, Arenas M, Greenfield SM, et al. (2008). Prospective evaluation of the pharmacogenetics of azathioprine in the treatment of inflammatory bowel disease. Aliment Pharmacol Ther 28:973–83.
  • Ansari A, Elliott T, Baburajan B, et al. (2008). Long-term outcome of using allopurinol co-therapy as a strategy for overcoming thiopurine hepatotoxicity in treating inflammatory bowel disease. Aliment Pharmacol Ther 28:734–41.
  • Appell ML, Berg J, Duley J, et al. (2013). Nomenclature for alleles of the thiopurine methyltransferase gene. Pharmacogenet Genomics 23:242–8.
  • Arenas M, Duley J, Sumi S, et al. (2007). The ITPA c.94C > A and g.IVS2 + 21A > C sequence variants contribute to missplicing of the ITPA gene. Biochim Biophys Acta 1772:96–102.
  • Askanase AD, Wallace DJ, Weisman MH, et al. (2009). Use of pharmacogenetics, enzymatic phenotyping, and metabolite monitoring to guide treatment with azathioprine in patients with systemic lupus erythematosus. J Rheumatol 36:89–95.
  • Berth-Jones J, Takwale A, Tan E, et al. (2002). Azathioprine in severe adult atopic dermatitis: a double-blind, placebo-controlled, crossover trial. Br J Dermatol 147:324–30.
  • Blaker PA, Arenas-Hernandez M, Smith MA, et al. (2013). Mechanism of allopurinol induced TPMT inhibition. Biochem Pharmacol 86:539–47.
  • Boonsrirat U, Angsuthum S, Vannaprasaht S, et al. (2008). Azathioprine-induced fatal myelosuppression in systemic lupus erythematosus patient carrying TPMT*3C polymorphism. Lupus 17:132–4.
  • Brem R, Karran P. (2012). Oxidation-mediated DNA cross-linking contributes to the toxicity of 6-thioguanine in human cells. Cancer Res 72:4787–95.
  • Brouwer C, Marinaki AM, Lambooy LH, et al. (2001). Pitfalls in the determination of mutant alleles of the thiopurine methyltransferase gene. Leukemia 15:1792–3.
  • Budhiraja P, Popovtzer M. (2011). Azathioprine-related myelosuppression in a patient homozygous for TPMT*3A. Nat Rev Nephrol 7:478–84.
  • Burchenal JH, Murphy ML, Ellison RR, et al. (1953). Clinical evaluation of a new antimetabolite, 6-mercaptopurine, in the treatment of leukemia and allied diseases. Blood 8:965–99.
  • Calne RY. (1960). The rejection of renal homografts. Inhibition in dogs by 6-mercaptopurine. Lancet 275:417–8.
  • Cargnin S, Genazzani AA, Canonico PL, Terrazzino S. (2018). Diagnostic accuracy of NUDT15 gene variants for thiopurine-induced leukopenia: a systematic review and meta-analysis. Pharmacol Res 135:102–11.
  • Carter M, Jemth AS, Hagenkort A, et al. (2015). Crystal structure, biochemical and cellular activities demonstrate separate functions of MTH1 and MTH2. Nat Commun 6:7871.
  • Chang JY, Park SJ, Jung ES, et al. (2019). Genotype-based treatment with thiopurine reduces incidence of myelosuppression in patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol. doi.10.1016/j.cgh.2019.08.034.
  • Chao K, Wang X, Cao Q, et al. (2017). Combined detection of NUDT15 variants could highly predict thiopurine-induced leukopenia in chinese patients with inflammatory bowel disease: a multicenter analysis. Inflamm Bowel Dis 23:1592–9.
  • Cheung ST, Allan RN. (2003). Mistaken identity: misclassification of TPMT phenotype following blood transfusion. Eur J Gastroenterol Hepatol 15:1245–7.
  • Choi R, Sohn I, Kim MJ, et al. (2019). Pathway genes and metabolites in thiopurine therapy in korean children with acute lymphoblastic leukemia. Br J Clin Pharmacol 85:1585.
  • Coenen MJH, de Jong DJ, van Marrewijk CJ, et al.; TOPIC Recruitment Team. (2015). Identification of patients with variants in TPMT and dose reduction reduces hematologic events during thiopurine treatment of inflammatory bowel disease. Gastroenterology 149:907–17.
  • Colleoni L, Kapetis D, Maggi L, et al. (2013). A new thiopurine s-methyltransferase haplotype associated with intolerance to azathioprine. J Clin Pharmacol 53:67–74.
  • Collie-Duguid ES, Pritchard SC, Powrie RH, et al. (1999). The frequency and distribution of thiopurine methyltransferase alleles in Caucasian and Asian populations. Pharmacogenetics 9:37–42.
  • Colombel JF, Ferrari N, Debuysere H, et al. (2000). Genotypic analysis of thiopurine S-methyltransferase in patients with Crohn’s disease and severe myelosuppression during azathioprine therapy. Gastroenterology 118:1025–30.
  • Coulthard SA, McGarrity S, Sahota K, et al. (2018). Three faces of mercaptopurine cytotoxicity in vitro: methylation, nucleotide homeostasis, and deoxythioguanosine in DNA. Drug Metab Dispos 46:1191–9.
  • Daehn I, Brem R, Barkauskaite E, Karran P. (2011). 6-Thioguanine damages mitochondrial DNA and causes mitochondrial dysfunction in human cells. FEBS Lett 585:3941–6.
  • Dorababu P, Naushad SM, Linga VG, et al. (2012). Genetic variants of thiopurine and folate metabolic pathways determine 6-MP-mediated hematological toxicity in childhood ALL. Pharmacogenomics 13:1001–8.
  • Dubinsky MC, Lamothe S, Yang HY, et al. (2000). Pharmacogenomics and metabolite measurement for 6-mercaptopurine therapy in inflammatory bowel disease. Gastroenterology 118:705–13.
  • Dubinsky MC, Yang H, Hassard PV, et al. (2002). 6-MP metabolite profiles provide a biochemical explanation for 6-MP resistance in patients with inflammatory bowel disease. Gastroenterology 122:904–15.
  • Elion GB. (1989). The purine path to chemotherapy. Science. 244:41–7.
  • Elion GB, Callahan S, Rundles RW, Hitchings GH. (1963). Relationship between metabolic fates and antitumor activities of thiopurines. Cancer Res 23:1207–17.
  • Evans WE, Hon YY, Bomgaars L, et al. (2001). Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathioprine. J Clin Oncol 19:2293–301.
  • Fazel-Najafabadi E, Vahdat Ahar E, Fattahpour S, Sedghi M. (2015). Structural and functional impact of missense mutations in TPMT: an integrated computational approach. Comput Biol Chem 59:48–55.
  • Ford L, Prout C, Gaffney D, Berg J. (2004). Whose TPMT activity is it anyway? Ann Clin Biochem 41:498–500.
  • Gerbek T, Ebbesen M, Nersting J, et al. (2018). Role of TPMT and ITPA variants in mercaptopurine disposition. Cancer Chemother Pharmacol 81:579–86.
  • Higgs JE, Payne K, Roberts C, Newman WG. (2010). Are patients with intermediate TPMT activity at increased risk of myelosuppression when taking thiopurine medications? Pharmacogenomics 11:177–88.
  • Hindorf U, Appell ML. (2012). Genotyping should be considered the primary choice for pre-treatment evaluation of thiopurine methyltransferase function. J Crohn’s Colitis 6:655–9.
  • Hung SI, Chung WH, Liou LB, et al. (2005). HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc Natl Acad Sci U S A 102:4134–9.
  • Khanna R, Bressler B, Levesque BG, et al.; REACT Study Investigators. (2015). Early combined immunosuppression for the management of Crohn’s disease (REACT): a cluster randomised controlled trial. Lancet 386:1825–34.
  • Khera S, Trehan A, Bhatia P, et al. (2019). Prevalence of TPMT, ITPA and NUDT 15 genetic polymorphisms and their relation to 6MP toxicity in north Indian children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol 83:341–8.
  • Krynetski EY, Krynetskaia NF, Yanishevski Y, Evans WE. (1995a). Methylation of mercaptopurine, thioguanine, and their nucleotide metabolites by heterologously expressed human thiopurine S-methyltransferase. Mol Pharmacol 47:1141–7.
  • Krynetski EY, Schuetz JD, Galpin AJ, et al. (1995b). A single point mutation leading to loss of catalytic activity in human thiopurine S-methyltransferase. Proc Natl Acad Sci U S A 92:949–53.
  • Kurzawski M, Dziewanowski K, Ciechanowski K, Drozdzik M. (2005). Severe azathioprine-induced myelotoxicity in a kidney transplant patient with thiopurine S-methyltransferase-deficient genotype (TPMT*3A/*3C). Transplant Int 18:623–5.
  • Lee JH, Kim TJ, Kim ER, et al. (2017). Measurements of 6-thioguanine nucleotide levels with TPMT and NUDT15 genotyping in patients with Crohn’s disease. PLoS One 12:e0188925.
  • Leipold G, Schütz E, Haas JP, Oellerich M. (1997). Azathioprine-induced severe pancytopenia due to a homozygous two-point mutation of the thiopurine methyltransferase gene in a patient with juvenile HLA-B27-associated spondylarthritis. Arthritis Rheum 40:1896–8.
  • Lennard L, Cartwright CS, Wade R, et al. (2013). Thiopurine methyltransferase genotype-phenotype discordance and thiopurine active metabolite formation in childhood acute lymphoblastic leukaemia. Br J Clin Pharmacol 76:125–36.
  • Lennard L, Gibson BE, Nicole T, Lilleyman JS. (1993). Congenital thiopurine methyltransferase deficiency and 6-mercaptopurine toxicity during treatment for acute lymphoblastic leukaemia. Arch Dis Child 69:577–9.
  • Lennard L, Lilleyman JS, Van Loon J, Weinshilboum RM. (1990). Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemia. Lancet 336:225–9.
  • Leung M, Piatkov I, Rochester C, et al. (2009). Normal thiopurine methyltransferase phenotype testing in a Crohn disease patient with azathioprine induced myelosuppression. Int Med J 39:121–6.
  • Levin RH, Landy M, Frei E. (1964). The effect of 6-mercaptopurine on immune response in man. N Engl J Med 271:16–22.
  • Liu Y, Meng Y, Wang L, et al. (2018). Associations between the NUDT15 R139C polymorphism and susceptibility to thiopurine-induced leukopenia in Asians: a meta-analysis. OncoTarget Ther 11:8309–17.
  • Liu YP, Wu HY, Yang X, et al. (2015). Association between thiopurine S-methyltransferase polymorphisms and thiopurine-induced adverse drug reactions in patients with inflammatory bowel disease: a meta-analysis. PLoS One 10:e0121745.
  • Liu YP, Xu HQ, Li M, et al. (2015). Association between thiopurine S-methyltransferase polymorphisms and azathioprine-induced adverse drug reactions in patients with autoimmune diseases: a meta-analysis. PLoS One 10:e0144234.
  • Loennechen T, Yates CR, Fessing MY, et al. (1998). Isolation of a human thiopurine S-methyltransferase (TPMT) complementary DNA with a single nucleotide transition A719G (TPMT*3C) and its association with loss of TPMT protein and catalytic activity in humans. Clin Pharmacol Ther 64:46–51.
  • Maeda T, Sumi S, Ueta A, et al. (2005). Genetic basis of inosine triphosphate pyrophosphohydrolase deficiency in the Japanese population. Mol Genet Metab 85:271–9.
  • Marinaki AM, Ansari A, Duley JA, et al. (2004). Adverse drug reactions to azathioprine therapy are associated with polymorphism in the gene encoding inosine triphosphate pyrophosphatase (ITPase). Pharmacogenetics 14:181–7.
  • Marinaki AM, Arenas M, Khan ZH, et al. (2003). Genetic determinants of the thiopurine methyltransferase intermediate activity phenotype in British Asians and Caucasians. Pharmacogenetics 13:97–105.
  • Mazel P, Henderson JF, Axelrod J. (1964). S-Demethylation by microsomal enzymes. J Pharmacol Exp Ther 143:1–6.
  • Meggitt SJ, Gray JC, Reynolds NJ. (2006). Azathioprine dosed by thiopurine methyltransferase activity for moderate-to-severe atopic eczema: a double-blind, randomised controlled trial. Lancet 367:839–46.
  • Milosevic G, Kotur N, Krstovski N, et al. (2018). Variants in TPMT, ITPA, ABCC4 and ABCB1 genes as predictors of 6-mercaptopurine induced toxicity in children with acute lymphoblastic leukemia. J Med Biochem 37:320–7.
  • Moradveisi B, Muwakkit S, Zamani F, et al. (2019). ITPA, TPMT, and NUDT15 genetic polymorphisms predict 6-mercaptopurine toxicity in middle eastern children with acute lymphoblastic leukemia. Frontiers Pharmacol 10:916.
  • Moriyama T, Nishii R, Lin TN, et al. (2017a). The effects of inherited NUDT15 polymorphisms on thiopurine active metabolites in Japanese children with acute lymphoblastic leukemia. Pharmacogenet Genom 27:236–9.
  • Moriyama T, Nishii R, Perez-Andreu V, et al. (2016). NUDT15 polymorphisms alter thiopurine metabolism and hematopoietic toxicity. Nature Genet 48:367–73.
  • Moriyama T, Yang YL, Nishii R, et al. (2017b). Novel variants in NUDT15 and thiopurine intolerance in children with acute lymphoblastic leukemia from diverse ancestry. Blood 130:1209–12.
  • Naushad SM, Dorababu P, Rupasree Y, et al. (2019). Classification and regression tree-based prediction of 6-mercaptopurine-induced leucopenia grades in children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol 83:875–80.
  • Nelson JA, Carpenter JW, Rose LM, Adamson DJ. (1975). Mechanisms of action of 6-thioguanine, 6-mercaptopurine, and 8-azaguanine. Cancer Res 35:2872–8.
  • Osterman MT, Kundu R, Lichtenstein GR, Lewis JD. (2006). Association of 6-thioguanine nucleotide levels and inflammatory bowel disease activity: a meta-analysis. Gastroenterology 130:1047–53.
  • Otterness D, Szumlanski C, Lennard L, et al. (1997). Human thiopurine methyltransferase pharmacogenetics: gene sequence polymorphisms. Clin Pharmacol Ther 62:60–73.
  • Pazmiño PA, Sladek SL, Weinshilboum RM. (1980). Thiol S-methylation in uremia: erythrocyte enzyme activities and plasma inhibitors. Clin Pharmacol Ther 28:356–67.
  • Planet G, Fox IH. (1976). Inhibition of phosphoribosylpyrophosphate synthesis by purine nucleosides in human erythrocytes. J Biol Chem 251:5839–44.
  • Poppe D, Tiede I, Fritz G, et al. (2006). Azathioprine suppresses ezrin-radixin-moesin-dependent T cell-APC conjugation through inhibition of Vav guanosine exchange activity on Rac proteins. J Immunol 176:640–51.
  • Rappaport HP. (1993). Replication of the base pair 6-thioguanine/5-methyl-2-pyrimidine with the large Klenow fragment of Escherichia coli DNA polymerase I. Biochemistry 32:3047–57.
  • Relling MV, Schwab M, Whirl-Carrillo M, et al. (2019). Clinical Pharmacogenetics Implementation Consortium Guideline for thiopurine dosing based on TPMT and NUDT15 genotypes: 2018 update. Clin Pharmacol Ther 105:1095–105.
  • Roberts RL, Gearry RB, Bland MV, et al. (2008). Trinucleotide repeat variants in the promoter of the thiopurine S-methyltransferase gene of patients exhibiting ultra-high enzyme activity. Pharmacogenet Genom 18:434–8.
  • Salavaggione OE, Wang L, Wiepert M, et al. (2005). Thiopurine S-methyltransferase pharmacogenetics: variant allele functional and comparative genomics. Pharmacogenet Genom 15:801–15.
  • Sandborn W, Sutherland L, Pearson D, et al. (2000). Azathioprine or 6-mercaptopurine for inducing remission of Crohn’s disease. Cochrane Database Syst Rev (2):CD000545.
  • Sarcione EJ, Stutzman L. (1960). A comparison of the metabolism of 6-mercaptopurine and its 6-methyl analog in the rat. Cancer Res 20:387–92.
  • Schaeffeler E, Fischer C, Brockmeier D, et al. (2004). Comprehensive analysis of thiopurine S-methyltransferase phenotype-genotype correlation in a large population of German-Caucasians and identification of novel TPMT variants. Pharmacogenetics 14:407–17.
  • Schaeffeler E, Jaeger SU, Klumpp V, et al. (2019). Impact of NUDT15 genetics on severe thiopurine-related hematotoxicity in patients with European ancestry. Genet Med 21:2145.
  • Shah SAV, Paradkar M, Desai D, Ashavaid TF. (2017). Nucleoside diphosphate-linked moiety X-type motif 15 C415T variant as a predictor for thiopurine-induced toxicity in Indian patients. J Gastroenterol Hepatol 32:620–4.
  • Shih DQ, Nguyen M, Zheng L, et al. (2012). Split-dose administration of thiopurine drugs: a novel and effective strategy for managing preferential 6-MMP metabolism. Aliment Pharmacol Ther 36:449–58.
  • Sluiter RL, van Marrewijk C, de Jong D, et al. (2019). Genotype-guided thiopurine dosing does not lead to additional costs in patients with inflammatory bowel disease. J Crohns Colitis 13:838–45.
  • Smith M, Blaker P, Patel C, et al. (2013). The impact of introducing thioguanine nucleotide monitoring into an inflammatory bowel disease clinic. Int J Clin Pract 67:161–9.
  • Sparrow MP, Hande SA, Friedman S, et al. (2005). Allopurinol safely and effectively optimizes tioguanine metabolites in inflammatory bowel disease patients not responding to azathioprine and mercaptopurine. Aliment Pharmacol Ther 22:441–6.
  • Spire-Vayron de la Moureyre C, Debuysère H, Fazio F, et al. (1999). Characterization of a variable number tandem repeat region in the thiopurine S-methyltransferase gene promoter. Pharmacogenetics 9:189–98.
  • Stocco G, Cheok MH, Crews KR, et al. (2009). Genetic polymorphism of inosine triphosphate pyrophosphatase is a determinant of mercaptopurine metabolism and toxicity during treatment for acute lymphoblastic leukemia. Clin Pharmacol Ther 85:164–72.
  • Sumi S, Marinaki AM, Arenas M, et al. (2002). Genetic basis of inosine triphosphate pyrophosphohydrolase deficiency. Hum Genet 111:360–7.
  • Szumlanski C, Otterness D, Her C, et al. (1996). Thiopurine methyltransferase pharmacogenetics: human gene cloning and characterization of a common polymorphism. DNA Cell Biol 15:17–30.
  • Tai HL, Fessing MY, Bonten EJ, et al. (1999). Enhanced proteasomal degradation of mutant human thiopurine S-methyltransferase (TPMT) in mammalian cells: mechanism for TPMT protein deficiency inherited by TPMT*2, TPMT*3A, TPMT*3B or TPMT*3C. Pharmacogenetics 9:641–50.
  • Tai HL, Krynetski EY, Yates CR, et al. (1996). Thiopurine S-methyltransferase deficiency: two nucleotide transitions define the most prevalent mutant allele associated with loss of catalytic activity in Caucasians. Am J Hum Genet 58:694–702.
  • Takagi Y, Setoyama D, Ito R, et al. (2012). Human MTH3 (NUDT18) protein hydrolyzes oxidized forms of guanosine and deoxyguanosine diphosphates: comparison with MTH1 and MTH2. J Biol Chem 287:21541–9.
  • Takatsu N, Matsui T, Murakami Y, et al. (2009). Adverse reactions to azathioprine cannot be predicted by thiopurine S-methyltransferase genotype in Japanese patients with inflammatory bowel disease. J Gastroenterol Hepatol 24:1258–64.
  • Tassaneeyakul W, Srimarthpirom S, Reungjui S, et al. (2003). Azathioprine-induced fatal myelosuppression in a renal-transplant recipient who carried heterozygous TPMT*1/*3C. Transplantation 76:265–6.
  • Tay BS, Lilley RM, Murray AW, Atkinson MR. (1969). Inhibition of phosphoribosyl pyrophosphate amidotransferase from Ehrlich ascites-tumour cells by thiopurine nucleotides. Biochem Pharmacol 18:936–8.
  • Tiede I, Fritz G, Strand S, et al. (2003). CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. J Clin Invest 111:1133–45.
  • Ugajin T, Miyatani H, Demitsu T, et al. (2009). Severe myelosuppression following alopecia shortly after the initiation of 6-mercaptopurine in a patient with Crohn’s disease. Int Med 48:693–5.
  • Urowitz MB, Gordon DA, Smythe HA, et al. (1973). Azathioprine in rheumatoid arthritis. A double-blind, cross over study. Arthritis Rheum 16:411–8.
  • Valerie NCK, Hagenkort A, Page BDG, et al. (2016). NUDT15 hydrolyzes 6-thio-deoxyGTP to mediate the anticancer efficacy of 6-thioguanine. Cancer Res 76:5501–11.
  • Van Dieren JM, Hansen BE, Kuipers EJ, et al. (2007). Meta-analysis: inosine triphosphate pyrophosphatase polymorphisms and thiopurine toxicity in the treatment of inflammatory bowel disease. Aliment Pharmacol Ther 26:643–52.
  • van Egmond R, Chin P, Zhang M, et al. (2012). High TPMT enzyme activity does not explain drug resistance due to preferential 6-methylmercaptopurine production in patients on thiopurine treatment. Aliment Pharmacol Ther 35:1181–9.
  • van Gennep S, Konté K, Meijer B, et al. (2019). Systematic review with meta-analysis: risk factors for thiopurine-induced leukopenia in IBD. Aliment Pharmacol Ther 50:484–506.
  • Vasudevan A, Gibson PR, Van Langenberg DR. (2019). Systematic review: cost-effective strategies of optimizing anti-tumor necrosis and immunomodulators in inflammatory bowel disease. Inflammat Bowel Dis 25:1462.
  • Walker GJ, Harrison JW, Heap GA, et al.; IBD Pharmacogenetics Study Group. (2019). Association of genetic variants in NUDT15 with thiopurine-induced myelosuppression in patients with inflammatory bowel disease. JAMA 321:773–85.
  • Warren DJ, Andersen A, Slørdal L. (1995). Quantitation of 6-thioguanine residues in peripheral blood leukocyte DNA obtained from patients receiving 6-mercaptopurine-based maintenance therapy. Cancer Res 55:1670–4.
  • Wee JS, Marinaki A, Smith CH. (2011). Life threatening myelotoxicity secondary to azathioprine in a patient with atopic eczema and normal thiopurine methyltransferase activity. BMJ 342:d1417.
  • Weinshilboum RM, Sladek SL. (1980). Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet 32:651–62.
  • Yamanaka H, Kamatani N, Nishida Y, et al. (1984). Relationship between phosphorylation and cytotoxicity of 2-chloroadenosine and 6-methylmercaptopurine riboside in human cells. Biochimica Et Biophysica Acta 798:291–4.
  • Yang SK, Hong M, Baek J, et al. (2014). A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 46:1017–20.
  • Yang JJ, Landier W, Yang W, et al. (2015). Inherited NUDT15 variant is a genetic determinant of mercaptopurine intolerance in children with acute lymphoblastic leukemia. J Clin Oncol 33:1235–42.
  • Yan L, Zhang S, Eiff B, et al. (2000). Thiopurine methyltransferase polymorphic tandem repeat: genotype-phenotype correlation analysis. Clin Pharmacol Therap 68:210–9.
  • Yi ES, Choi YB, Choi R, et al. (2018). NUDT15 variants cause hematopoietic toxicity with low 6-TGN levels in children with acute lymphoblastic leukemia. Cancer Res Treat 50:872–82.
  • You C, Dai X, Yuan B, Wang Y. (2012). Effects of 6-thioguanine and S6-methylthioguanine on transcription in vitro and in human cells. J Biol Chem 287:40915–23.
  • Yuan B, Zhang J, Wang H, et al. (2011). 6-Thioguanine reactivates epigenetically silenced genes in acute lymphoblastic leukemia cells by facilitating proteasome-mediated degradation of DNMT1. Cancer Res 71:1904–11.
  • Zgheib NK, Akika R, Mahfouz R, et al. (2017). NUDT15 and TPMT genetic polymorphisms are related to 6-mercaptopurine intolerance in children treated for acute lymphoblastic leukemia at the Children’s Cancer Center of Lebanon. Pediatric Blood Cancer 64:146–50.
  • Zhou H, Li L, Yang P, et al. (2018). Optimal predictor for 6-mercaptopurine intolerance in Chinese children with acute lymphoblastic leukemia: NUDT15, TPMT, or ITPA genetic variants? BMC Cancer 18:516.
  • Zhu X, Wang XD, Chao K, et al. (2016). NUDT15 polymorphisms are better than thiopurine S-methyltransferase as predictor of risk for thiopurine-induced leukopenia in Chinese patients with Crohn’s disease. Aliment Pharmacol Ther 44:967–75.
  • Zur RM, Roy LM, Ito S, et al. (2016). Thiopurine S-methyltransferase testing for averting drug toxicity: a meta-analysis of diagnostic test accuracy. Pharmacogenom J 16:305–11.

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.