Thiopurine methyltransferase genotype and thiopurine S-methyltransferase activity in Greek children
with inflammatory bowel disease

Ìaria Gazoulia, Ioanna Pachoulab, Ioanna Panayotoub, Georgios Chouliarasb,
Nicholas P. Anagnoua,c, George Chroussosb, Eleftheria Romab

Laboratory of Biology, School of Medicine University of Athens, Aghia Sophia Children’s Hospital, Foundation
of Biomedical Research of the Academy of Athens, Athens, Greece

aDepartment of Basic Medical Science, Laboratory of Biology,
School of Medicine, University of Athens, (Maria Gazouli,
Nicholas P. Anagnou); bFirst Department of Pediatrics,
“Aghia Sophia” Children’s Hospital, School of Medicine,
University of Athens (Ioanna Pachoula, Ioanna Panayotou,
Georgios Chouliaras, George Chroussos, Eleftheria Roma); cLaboratory of Cell and Gene Therapy, Centre of Basic Research II, Foundation for Biomedical Research of the Academy of Athens (IIBEAA) (Nicholas P. Anagnou), Athens, Greece

Conflict of Interest: None

Correspondence to: Maria Gazouli, PhD, Department of Basic Medical Science, Laboratory of Biology, School of Medicine, University of Athens, Michalakopoulou 176, 11527 Athens, Greece, e-mail: mgazouli@med.uoa.gr

Received 30 April 2012; accepted 7 May 2012

Abstract

Background Azathioprine (AZA) and 6-mercaptopurine (6MP) are used in the treatment of pediatric inflammatory bowel disease (IBD). Genetic variations in thiopurine S-methyltranfarase (TPMT) gene have been correlated with enzyme activity and with the occurrence of adverse events to AZA and 6MP. The aim of the present study was to examine the sensitivity and specificity of TPMT genotyping for TPMT enzymatic activity, reducing harm from thiopurine by pretesting, and the association of thiopurine toxicity with TPMT status in children with IBD.

Methods TPMT red blood cell (RBC) activity was measured by using a radiochemical method and genotype was determined for the TPMT alleles *2, *3A, *3B and *3C in 108 thiopurine-treated pediatric IBD patients with a mean age of 11.3 years (range 3-16).

Results Significant TPMT activity differences between wild-type and heterozygous and homozygous mutated subjects were observed. We divided TPMT activity into three categories according to frequency distribution: low (16.67%), intermediate (25.92%) and high (57.41%). The whole population included a total of 77.78% of homozygous wild-type subjects, 15.74% heterozygous variants, 1.85% homozygous variants and five (4.63%) compound heterozygous variant TPMT*3B/*3C. The overall concordance rate between TPMT genotypes and phenotypes was 88.2%. Seven carriers of at least one variant allele and low or intermediate TPMT activity developed adverse effects.

Conclusions Our findings suggest that carriers of at least one variant allele and both intermediate and absent TPMT activity have an increased risk of developing thiopurine-induced myelotoxicity compared with individuals with normal genotype and TPMT activity.

Keywords azathioprine, 6MP, pharmacogenetics, TPMT, inflammatory bowel disease

Introduction

Azathioprine (AZA) and 6-mercaptopurine (6MP) are widely used as immunosuppressive drugs in the treatment of children with inflammatory bowel disease (IBD) [Crohn’s disease (CD) and ulcerative colitis (UC)]. A number of questions remain regarding the need to optimize AZA metabolism in IBD patients treated with thiopurines. Thiopurines are metabolized by a complex multistep enzymatic pathway. After it is absorbed, AZA is metabolized to 6MP in the liver then during metabolism, hypoxanthine-guanine phosphoribosyltransferase (HGPRT) converts 6MP to cytotoxic 6-thioguanine nucleotide analogues, while thiopurine methyltransferase (TPMT) inactivates 6MP through methylation to form 6-methylmercaptopurine [1,2]. The metabolism and the production of active metabolites are mainly regulated by TPMT.

The TPMT gene has an autosomal codominant inheritance and the TPMT activity is largely influenced by polymorphisms, which results in a trimodal distribution; those patients heterozygous or homozygous for the “low activity” mutation gene might have an increased susceptibility for myelotoxicity with thiopurine therapy [3-6]. It has been reported that patients with inherited TPMT deficiency treated with standard doses of thiopurines present with higher levels of the thioguanine active metabolites and have an increased risk for adverse events. Unless patients with two defective alleles are treated with 10- to 15-fold lower doses of this medication, potentially fatal, hematopoietic toxicity, which requires immediate discontinuation of treatment, may follow [7-9].

TPMT genetic polymorphism was first described by Weinshilboum and Sladek [10]. In Caucasians, approximately 11% of the population harbour heterozygous and 0.3% homozygous TPMT mutations, leading to an intermediate or low TPMT activity, respectively. In these patients, thiopurine metabolism was shunted towards an increased production of active and toxic compounds. A high degree of concordance was demonstrated between TPMT genotype and phenotype in Caucasians [11,12]. Whether determining TPMT status prior to the start of thiopurine therapy, and adapting the dose accordingly, should be systematically performed in order to minimize the risk of myelotoxicity remains controversial [13-15]. In contrast to European [16] guidelines, American guidelines suggest the use of TPMT determination before thiopurine administration [17-19].

Therefore, in our study we aimed to examine the sensitivity and specificity of TPMT genotyping for TPMT enzymatic activity, reducing harm from thiopurine by pretesting, and the association of thiopurine toxicity with TPMT status in children with IBD.

Material and Methods

Patients with IBD who presented between February 2007 and August 2011 at the First Department of Pediatrics of the ‘Aghia Sophia’ Children’s Hospital were consecutively enrolled. IBD was diagnosed based on clinical, endoscopic, radiological and histological criteria [20]. The study included patients who had been taking AZA or 6MP for at least 3 months or who had experienced adverse effects during treatment with these drugs. Thiopurine dosage had to be 0.3-2.5 mg⁄kg. Bone marrow suppression was defined as leukopenia (WBC <3000 ⁄mm3) and/or thrombocytopenia (platelets <100 000 ⁄mm3), hepatotoxicity by serum alanine transaminase levels greater than twice the upper normal limit resolving after withdrawal of thiopurine drug, and pancreatitis by severe abdominal pain and hyperamylasemia resolving after withdrawal of thiopurine drug. The study was approved by the Ethics Committees of the participant centers.

After lysis of red blood cell (RBC), RBC TPMT activity was measured by a radiochemical method, as previously described [21].

For genotype analysis, venous blood samples (2 mL from each pediatric patient) were collected. Genomic screening was accomplished by a polymerase chain reaction (PCR) and restriction fragment length polymorphisms assay as previously described [15]. DNA of the patients was screened for TPMT*3A (both G460A and A719G mutation), TPMT*3B (only G460A mutation), TPMT*3C (only A719G mutation) and TPMT*2 (G238C mutation). Differences in allele frequencies were compared with the chi-square test (GRAPHPAD V. 3.00; GraphPad Software, San Diego, CA, USA). The statistical associations were tested using two-sided Fisher’s exact test, and compared using the odds ratios and 95% confidence intervals. Strong association (significance) was assumed at p <0.05.

 

Results

The clinical data of the 108 patients enrolled in the study are presented in Table 1. Seventy-seven were diagnosed with CD (71.29%), 20 with UC (18.52%) and 11 with indeterminate colitis (IC) (10.18%). All patients received standard recommended thiopurine doses for 6MP 1.0-1.5 mg/kg/day and for AZA 2.0-2.5 mg/kg/day. The patients were closely monitored through blood tests and regular medical visits in order to evaluate the clinical response and development of adverse events. According to toxicity, 13 children presented side effects to AZA and 6MP (Table 2).

 

 

 

 

 

The TPMT activity ranged from 0.36 up to 78 U/mL RBC. Among all patients included, 62 (57.41%) had normal to high TPMT activity (>15.6 U/mL RBC), 28 (28.92%) had intermediate (5.6 – 15.5 U/mL RBC) and 18 (16.67%) had low (<5.5 U/mL RBC) TPMT activity. Distribution of TPMT activity is represented in Fig. 1.

 

 

 

Among the 108 IBD children enrolled in the study a total of 77.78% were homozygous wild-type subjects, 15.74% heterozygous variants, 1.85% homozygous variants and 5 (4.63%) compound heterozygous variant TPMT*3B/*3C. The median TPMT activity in patients with a wild-type genotype was 27.27 U/mL RBC (range 15.8 - 78 U/mL RBC), in subjects with heterozygous phenotype 7.64 U/mL RBC (range 0.79 – 15.46 U/mL RBC) and in carriers homozygous mutated genotype 1.25 U/mL RBC (range 0.36 – 5.7 U/mL RBC). The difference between the values of patients with wild-type genotype and those of carriers of at least one TPMT variant is statistically significant (p <0.0001). The overall concordance rate between TPMT genotypes and phenotypes was 88.2 % (Fig. 2). Ten of the 46 carriers of at least one variant allele and/or with low or intermediate TPMT activity developed adverse effects, compared with 3 out of 62 with a wild-type TPMT gene and normal or high TPMT activity (O.R. 5.46, 95% CI 1.41 – 21.18, p = 0.014). More specifically, 2 patients with high TPMT activity and one with intermediate TPMT activity, all with wild-type genotype, developed hepatotoxicity. One patient with high TPMT activity, and 2 with intermediate TPMT activity, all with wild-type genotype; developed pancreatitis. The majority of the patients with low or intermediate TPMT activity developed bone marrow toxicity. All these patients’ thiopurines (AZA or 6MP) were withdrawn or reduced due to adverse effects.

 

 

 

Discussion

Thiopurines (AZA and 6MP) are widely used drugs in IBD treatment and are especially effective for the maintenance of remission in IBD [22], even though their use is limited by the occurrence of adverse effects.

Several genetics variants in TPMT have been systematically related to the occurrence of toxicity in thiopurine treatment. More than 25 mutations are now indexed but the clinical relevance of some of them remains unclear [23]. TPMT*3A, TPMT*3C, and TPMT*2 represent the most prevalent mutant alleles in Caucasians and African-Americans and account for 80-95% of intermediate or deficient methylator phenotypes [24,25]. It is accepted that patients carrying TPMT alleles associated with low TPMT activity, especially homozygosis, show a significant increase in the risk of adverse effects as a result of excessive accumulation of 6-TGN (Q3. Please explain abbreviation) [26,27]. TPMT activity might be determined in erythrocytes prior to therapy; nevertheless TPMT activity might be decreased by the use of commonly used drugs like sulfalazine, salicylic acid and their derivatives [28]. Therefore, Yates et al [29] reported that the predictability of TPMT genotype on the patients TPMT phenotype resulted in a greater than 95% correlation.

In agreement with previous studies, among the 108 subjects examined in the present study, the average level of enzymatic activity measured in patients with a mutated TPMT genotype was significantly lower than that of the remaining group carrying a wild-type TPMT genotype [28-30]. There are numerous studies on the relationship between TPMT low enzymatic activity and the adverse effects of thiopurines. Some of these studies show a notable increase in adverse effects such as bone marrow toxicity in patients with a mutated TPMT gene [16,31]. On the other hand, other studies suggest that thiopurine-induced adverse effects can be caused by factors other than TPMT mutations [32]. Our results are not in agreement with studies suggested that myelosuppression during thiopurine therapy of IBD patients are caused by factors other than mutations leading to reduced TPMT activity [29].

Our concordance rate between genotype and phenotype assays is 88.2% and is comparable to that reported in previous studies [13,28]. It is possible that additional genetic and epigenetic factors can contribute to an explanation of the differences between genotype and phenotype [33].

Even though it seems clear that patients with severe deficiency should not receive thiopurines because of the high risk of adverse effects [34], it is also well known that TPMT deficiency only explains a portion of all thiopurine-related adverse effects [3]. Additional single nucleotide polymorphisms in genes encoding enzymes involved in thiopurine metabolism (i.e. MDR1, ITPA, MTHFR etc) may also be responsible for this inter-individual response variability [25]. Consequently, opinion is still divided as to whether TMPT activity and TPMT genotyping determination is needed prior to starting AZA or 6MP therapy [35]. On the other hand, some authors have reported that patients with high TPMT activity are at high risk of suffering from hepatotoxicity because they accumulate methylated metabolites [36]. In this study, two patients with high TPMT activity and wild-type genotype developed hepatotoxicity. Even though this study has important limitations such as the management of the patients including concomitant drugs, it is suggested that the practice of examining TPMT genotype and TPMT activity prior to starting therapy with thiopurines has improved our ability to manage these patients more effectively and safely than previously. The practice of monitoring thiopurine metabolism has been well established in several countries such as Sweden, UK, New Zealand [19,37,38], where the national guidelines for treatment of IBD state that TPMT genotype and/or phenotype determination should be considered in all patients prior to initiation of thiopurine treatment. Recently, Chouchana et al [18] proposed a therapeutic algorithm allowing a dosage individualization to optimize and secure the management of patients under thiopurines. Incorporating pharmacogenetic factors into therapeutic management is promising for ensuring maximal therapeutic response of thiopurines with a minimization of the risk for adverse effects in IBD patients. Further studies evaluating such factors as well as the potential effects on patients’ safety are needed.

 

Summary Box

What is already known:

The patients with inherited TPMT deficiency treated with standard doses of thiopurines present with higher levels of the thioguanine active metabolites and have an increased risk for adverse events

In Caucasians, approximately 11% of the population harbour heterozygous and 0.3% homozygous TPMT mutations, leading to an intermediate or low TPMT activity, respectively

More than 25 mutations are now indexed but the clinical relevance of some of them remains unclear

TPMT*3A, TPMT*3C, and TPMT*2 represent the most prevalent mutant alleles in Caucasians and African-Americans and account for 80–95% of intermediate or deficient methylator phenotypes

What the new findings are:

Significant TPMT activity differences between wild-type and heterozygous and homozygous mutated subjects were observed

The carriers of at least one variant allele and with both intermediate and absent TPMT activity have an increased risk of developing thiopurine-induced myelotoxicity compared with individuals with normal genotype and TPMT activity

A high degree of concordance was demonstrated between TPMT genotype and phenotype

 

References

1.     Sahasranaman S, Howard D, Roy S. Clinical pharmacology and pharmacogenetics of thiopurines. Eur J Clin Pharmacol 2008;64:753-767.

2.     Lennard L. TPMT in the treatment of Crohn’s disease with azathioprine. Gut 2002;51:143-146.

3.     Gisbert JP, Gomollón F, Cara C, et al. Thiopurine methyltransferase activity in Spain: a study of 14,545 patients. Dig Dis Sci 2007;52:1262-1269.

4.     Schaeffeler E, Fischer C, Brockmeier D, 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;14:407-417.

5.     Gisbert JP, Niño P, Rodrigo L, Cara C, Guijarro LG. Thiopurine methyltransferase (TPMT) activity and adverse effects of azathioprine in inflammatory bowel disease: long-term follow-up study of 394 patients. Am J Gastroenterol 2006;101:2769-2776.

6.     Gisbert JP, Gomollón F. Thiopurine-induced myelotoxicity in patients with inflammatory bowel disease: a review. Am J Gastroenterol 2008;103:1783-1800.

7.     Evans WE, Horner M, Chu YQ, Kalwinsky D, Roberts WM. Altered mercaptopurine metabolism, toxic effects, and dosage requirement in a thiopurine methyltransferase-deficient child with acute lymphocytic leukaemia. J Pediatr 1991;119:985-989.

8.     Evans WE, Hon YY, Bomgaars L, et al. Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathioprine. J Clin Oncol 2001;19:2293-2301.

9.     Evans WE. Pharmacogenetics of thiopurine S-methyltransferase and thiopurine therapy. Ther Drug Monitor 2004;26:186-191.

10.   Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet 1980;32:651-662.

11.   Yates CR, Krynetski EY, Loennechen T, et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997;126:608-614.

12.   Rossi AM, Bianchi M, Guarnieri C, Barale R, Pacifici GM. Genotype-phenotype correlation for thiopurine S-methyltransferase in healthy Italian subjects. Eur J Clin Pharmacol 2001;57:51-54.

13.   Candy S, Wright J, Gerber M, Adams G, Gerig M, Goodman R. A controlled double blind study of azathioprine in the management of Crohn’s disease. Gut 1995;37:674-678.

14.   Roblin X, Oussalah A, Chevaux JB, Sparrow M, Peyrin-Biroulet L. Use of thiopurine testing in the management of inflammatory bowel diseases in clinical practice: a worldwide survey of experts. Inflamm Bowel Dis 2011;17:2480-2487.

15.   Gazouli M, Pachoula I, Panayotou I, et al. Thiopurine S-methyltransferase genotype and the use of thiopurines in paediatric inflammatory bowel disease Greek patients. J Clin Pharm Ther 2010;35:93-97.

16.   Travis SP, Stange EF, Lémann M, et al. European evidence based consensus on the diagnosis and management of Crohn’s disease: current management. Gut 2006;55(Suppl 1):i16-i35.

17.   Colombel JF, Ferrari N, Debuysere H, et al. Genotypic analysis of thiopurine S-methyltransferase in patients with Crohn’s disease and severe myelosuppression during azathioprine therapy. Gastroenterology 2000;118:1025-1030.

18.   Chouchana L, Narjoz C, Beaune P, Loriot MA, Roblin X. Review article: the benefits of pharmacogenetics for improving thiopurine therapy in inflammatory bowel disease. Aliment Pharmacol Ther 2012;35:15-36.

19.   Ford LT, Berg JD. Thiopurine S-methyltransferase (TPMT) assessment prior to starting thiopurine drug treatment; a pharmacogenomic test whose time has come. J Clin Pathol 2010;63:288-295.

20.   Lennard-Jones JE. Classification of inflammatory bowel disease. Scand J Gastroenterol 1989; (Suppl)170:2-6.

21.   Menor C, Fueyo JA, Escribano O, et al. Determination of thiopurine methyltransferase activity in human erythrocytes by high-performance liquid chromatography: comparison with the radiochemical method. Ther Drug Monit 2001;23:536-541.

22.   Peyrin-Biroulet L, Deltenre P, Ardizzone S, et al. Azathioprine and 6-mercaptopurine for the prevention of postoperative recurrence in Crohn’s disease: a meta-analysis. Am J Gastroenterol 2009;104:2089-2096.

23.   Derijks LJ, Wong DR. Pharmacogenetics of thiopurines in inflammatory bowel disease. Curr Pharm Des 2010;16:145-154.

24.   Hamdan-Khalil R, Allorge D, Lo-Guidice JM, et al. In vitro characterization of four novel non-functional variants of the thiopurine S-methyltransferase. Biochem Biophys Res Commun 2003;309:1005-1010.

25.   Katsanos K, Tsianos EV. Non-TPMT determinants of azathioprine toxicity in inflammatory bowel disease. Ann Gastroenterol 2010;23:95-101.

26.   Gearry RB, Barclay ML. Azathioprine and 6-mercaptopurine pharmacogenetics and metabolite monitoring in inflammatory bowel disease. J Gastroenterol Hepatol 2005;20:1149-1157.

27.   De Vroey B, Colombel JF. IBD in 2010: optimizing treatment and minimizing adverse events. Nat Rev Gastroenterol Hepatol 2011;8:74-76.

28.   Lennard L. TPMT in the treatment of Crohn’s disease with azathioprine. Gut 2002;51:143-146.

29.   Yates CR, Krynetski EY, Loennechen T, et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann Intern Med 1997;126:608-614.

30.   Stocco G, Martelossi S, Barabino A, et al. TPMT genotype and the use of thiopurines in paediatric inflammatory bowel disease. Dig Liver Dis 2005;37:940-945.

31.   Rossi AM, Bianchi M, Guarnieri C, Barale R, Pacifici GM. Genotype-phenotype correlation for thiopurine S-methyltransferase in healthy Italian subjects. Eur J Clin Pharmacol 2001;57:51-54.

32.   Teml A, Schaeffeler E, Herrlinger KR, Klotz U, Schwab M. Thiopurine treatment in inflammatory bowel disease: clinical pharmacology and implication of pharmacogenetically guided dosing. Clin Pharmacokinet 2007;46:187-208.

33.   Dubinsky MC, Lamothe S, Yang HY, et al. Pharmacogenomics and metabolite measurement for 6-mercaptopurine therapy in inflammatory bowel disease. Gastroenterology 2000;118:705-713.

34.   Krynetski EY, Evans WE. Genetic polymorphism of thiopurine S-methyltransferase: molecular mechanisms and clinical importance. Pharmacology 2000;61:136-146.

35.   Lichtenstein GR, Abreu MT, Cohen R, Tremaine W; American Gastroenterological Association. American Gastroenterological Association Institute technical review on corticosteroids, immunomodulators, and infliximab in inflammatory bowel disease. Gastroenterology 2006;130:940-987.

36.   Dignass A, Van Assche G, Lindsay JO, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Current management. J Crohns Colitis 2010;4:28-62.

37.   Dubinsky MC, Yang H, Hassard PV, et al. 6-MP metabolite profiles provide a biochemical explanation for 6-MP resistance in patients with inflammatory bowel disease. Gastroenterology 2002;122:904-915.

38.   Hindorf U, Andersson P. How are thiopurines used and monitored by Swedish gastroenterologists when treating patients with inflammatory bowel disease? Scand J Gastroenterol 2011;46:1215-1221.