Page 101 - Read Online
P. 101

Page 268                                              Franca et al. Cancer Drug Resist 2019;2:256-70 I http://dx.doi.org/10.20517/cdr.2019.004

                   2011;3:RRN1236.
               8.   Dubinsky MC, Lamothe S, Yang HY, Targan SR, Sinnett D, et al. Pharmacogenomics and metabolite measurement for 6-mercaptopurine
                   therapy in inflammatory bowel disease. Gastroenterology 2000;118:705-13.
               9.   Ben-Horin S, Van Assche G, Chowers Y, Fudim E, Ungar B, et al. Pharmacokinetics and immune reconstitution following
                   discontinuation of thiopurine analogues: implications for drug withdrawal strategies. J Crohns Colitis 2018;12:1410-7.
               10.  Derijks LJ, Gilissen LP, Engels LG, Bos LP, Bus PJ, et al. Pharmacokinetics of 6-mercaptopurine in patients with inflammatory bowel
                   disease: implications for therapy. Ther Drug Monit 2004;26:311-8.
               11.  Lancaster DL, Patel N, Lennard L, Lilleyman JS. Leucocyte versus erythrocyte thioguanine nucleotide concentrations in children taking
                   thiopurines for acute lymphoblastic leukaemia. Cancer Chemother Pharmacol 2002;50:33-6.
               12.  Cuffari C, Seidman EG, Latour S, Théorêt Y. Quantitation of 6-thioguanine in peripheral blood leukocyte DNA in Crohn’s disease
                   patients on maintenance 6-mercaptopurine therapy. Can J Physiol Pharmacol 1996;74:580-5.
               13.  Stocco G, Franca R, Verzegnassi F, Londero M, Rabusin M, et al. Pharmacogenomic approaches for tailored anti-leukemic therapy in
                   children. Curr Med Chem 2013;20:2237-53.
               14.  Franca R, Kuzelicki NK, Sorio C, Toffoletti E, Montecchini O, et al. Targeting kinase-activating genetic lesions to improve therapy of
                   pediatric acute lymphoblastic leukemia. Curr Med Chem 2018;25:2811-25.
               15.  Ling Y, Xie Q, Zhang Z, Zhang H. Protein kinase inhibitors for acute leukemia. Biomark Res 2018;6:8.
               16.  Avallone EV, Pica R, Cassieri C, Zippi M, Paoluzi P, et al. Azathioprine treatment in inflammatory bowel disease patients: type and time
                   of onset of side effects. Eur Rev Med Pharmacol Sci 2014;18:165-70.
               17.  Ledder O, Lemberg DA, Day AS. Thiopurine-induced pancreatitis in inflammatory bowel diseases. Expert Rev Gastroenterol Hepatol
                   2015;9:399-403.
               18.  Citterio-Quentin A, El Mahmoudi A, Perret T, Conway A, Ryan A, et al. Red Blood cell IMPDH activity in adults and children with or
                   without azathioprine: relationship between thiopurine metabolites, ITPA and TPMT activities. Basic Clin Pharmacol Toxicol 2018.
               19.  Urbančič D, Kotar A, Šmid A, Jukič M, Gobec S, et al. Methylation of selenocysteine catalysed by thiopurine S-methyltransferase.
                   Biochim Biophys Acta Gen Subj 2019;1863:182-90.
               20.  Schwarz G, Csaszar J, Schaeffeler E, Schwab, M. Diagnostic in vitro method, 2013 [Application number: US20160160264A1].
                   Available from: https://patents.google.com/patent/US20160160264A1/en. [Last accessed on 15 Apr 2019].
               21.  Collie-Duguid ES, Pritchard SC, Powrie RH, Sludden J, Collier DA, et al. The frequency and distribution of thiopurine
                   methyltransferase alleles in Caucasian and Asian populations. Pharmacogenetics 1999;9:37-42.
               22.  Schaeffeler E, Fischer C, Brockmeier D, Wernet D, Moerike K, et al. Comprehensive analysis of thiopurine S-methyltransferase
                   phenotype-genotype correlation in a large population of German-Caucasians and identification of novel TPMT variants.
                   Pharmacogenetics 2004;14:407-17.
               23.  Liu C, Yang W, Pei D, Cheng C, Smith C, et al. Genomewide approach validates thiopurine methyltransferase activity is a monogenic
                   pharmacogenomic trait. Clin Pharmacol Ther 2017;101:373-81.
               24.  Tiede I, Fritz G, Strand S, Poppe D, Dvorsky R, et al. CD28-dependent Rac-1 activation is the molecular target of azathioprine in
                   primary human CD4+ T lymphocytes. J Clin Invest 2003;111:1133-45.
               25.  Yates CR, Krynetski EY, Loennechen T, Fessing MY, Tai HL, et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency:
                   genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997;126:608-14.
               26.  Hindorf U, Appell ML. Genotyping should be considered the primary choice for pre-treatment evaluation of thiopurine
                   methyltransferase function. J Crohns Colitis 2012;6:655-9.
               27.  Chadli Z, Kerkeni E, Hannachi I, Chouchene S, Ben Fredj N, et al. Distribution of genetic polymorphisms of genes implicated in
                   thiopurine drugs metabolism. Ther Drug Monit 2018;40:655-9.
               28.  Saiz-Rodríguez M, Ochoa D, Belmonte C, Román M, Martínez-Ingelmo C, et al. Influence of thiopurine S-methyltransferase
                   polymorphisms in mercaptopurine pharmacokinetics in healthy volunteers. Basic Clin Pharmacol Toxicol 2019;124:449-55.
               29.  Oliveira E, Quental S, Alves S, Amorim A, Prata MJ. Do the distribution patterns of polymorphisms at the thiopurine S-methyltransferase
                   locus in sub-Saharan populations need revision? Hints from Cabinda and Mozambique. Eur J Clin Pharmacol 2007;63:703-6.
               30.  Tai HL, Krynetski EY, Schuetz EG, Yanishevski Y, Evans WE. Enhanced proteolysis of thiopurine S-methyltransferase (TPMT)
                   encoded by mutant alleles in humans (TPMT*3A, TPMT*2): mechanisms for the genetic polymorphism of TPMT activity. Proc Natl
                   Acad Sci U S A. 1997;94:6444-9.
               31.  Wang L, Sullivan W, Toft D, Weinshilboum R. Thiopurine S-methyltransferase pharmacogenetics: chaperone protein association and
                   allozyme degradation. Pharmacogenetics 2003;13:555-64.
               32.  Wang L, Nguyen TV, McLaughlin RW, Sikkink LA, Ramirez-Alvarado M, et al. Human thiopurine S-methyltransferase
                   pharmacogenetics: variant allozyme misfolding and aggresome formation. Proc Natl Acad Sci U S A. 2005;102:9394-9.
               33.  Coenen MJ, de Jong DJ, van Marrewijk CJ, Derijks LJ, Vermeulen SH, et al. Identification of patients with variants in tpmt and dose
                   reduction reduces hematologic events during thiopurine treatment of inflammatory bowel disease. Gastroenterology 2015;149:907-17.e7.
               34.  Hartford C, Vasquez E, Schwab M, Edick MJ, Rehg JE, et al. Differential effects of targeted disruption of thiopurine methyltransferase
                   on mercaptopurine and thioguanine pharmacodynamics. Cancer Res 2007;67:4965-72.
               35.  Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, et al. Clinical pharmacogenetics implementation consortium
                   guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther 2011;89:387-91.
               36.  Relling MV, Schwab M, Whirl-Carrillo M, Suarez-Kurtz G, Pui CH, et al. Clinical pharmacogenetics implementation consortium (CPIC)
                   guideline for thiopurine dosing based on TPMT and NUDT15 genotypes: 2018 update. Clin Pharmacol Ther 2018.
   96   97   98   99   100   101   102   103   104   105   106