You are on page 1of 7

CLINICAL GASTROENTEROLOGY AND HEPATOLOGY 2008;6:654 – 660

Thiopurine Dose in Intermediate and Normal Metabolizers of Thiopurine


Methyltransferase May Differ Three-Fold

SHARON J. GARDINER,*,‡ RICHARD B. GEARRY,*,§ EVAN J. BEGG,*,‡ MEI ZHANG,*,‡ and MURRAY L. BARCLAY*,‡,§
*Department of Medicine, ‡Department of Clinical Pharmacology, and §Department of Gastroenterology, Christchurch Hospital and Christchurch School of Medicine,
Christchurch, New Zealand

phically and integral to the metabolism of both azathioprine


See Van Assche G et al on page 1861 for and 6-MP (Figure 1).
companion article in the June 2008 issue of Testing for TPMT by genotype or phenotype (enzyme activ-
Gastroenterology. ity within erythrocytes) is aimed primarily at detecting the 0.3%
to 0.6% of individuals with negligible enzyme activity5,6 who
achieve very increased concentrations of the active 6-thiogua-
See CME exam on page 604.
nine nucleotides (6-TGNs) with standard thiopurine doses and
have a high likelihood of profound myelosuppression.7,8 Al-
Background & Aims: Patients with inflammatory bowel though these individuals are best managed with an alternate
disease (IBD) may have different thiopurine dose require- drug, case reports suggest that a thiopurine dose reduction to
ments in relation to thiopurine methyltransferase (TPMT) approximately 10% of normal may be safe and effective pro-
genotype and/or phenotype. The purpose of this study was vided that close monitoring may occur.7,9,10 Because myelosup-
to determine thiopurine dose requirements in intermediate pression may occur between routine blood counts it seems
versus normal TPMT metabolism status. Methods: Con- prudent to restrict the use of thiopurine drugs in cases of
secutive patients starting azathioprine or 6-mercaptopurine TPMT deficiency to those centers where 6-TGN monitoring can
for IBD were followed up for 9 months. The thiopurine be undertaken in a timely manner.
dose was individualized using 6-thioguanine nucleotide (6- There is comparatively less information on how to individ-
TGN) concentrations (range, 235– 450 pmol/8 ⴛ 108 red ualize thiopurine dose in intermediate versus normal metabo-
blood cells [RBCs]) and clinical status. Additional assess- lizers of TPMT who comprise, when combined, more than 99%
ments undertaken every three months included measures of of the population (⬃10% and ⬃90%, respectively). TPMT activ-
disease activity. Results: Eight (10%) of 77 participants ity varies 7-fold across these 2 groups, with 6-TGN concentra-
were withdrawn because of protocol violation. Fifty-two tions inversely related to TPMT activity.11,12 When comparing
(75%) of the remaining 69 subjects (⬃90% and 10% with the the 2 TPMT groups, intermediate metabolizers have an approx-
TPMT*1/*1 and *1/*3 genotypes, respectively) completed imately 5-fold greater risk of mild to moderate leukopenia and
follow-up on azathioprine (n ⴝ 46) or 6-mercaptopurine (n ⴝ an increased likelihood of response compared with normal
6). The mean initial dose (as azathioprine equivalents) was metabolizers.11–13 These findings have lead some investigators
similar (⬃1 mg/kg/d) for the 2 TPMT genotypes, but after 9 to suggest that intermediate metabolizers could benefit from a
months the dose was 50% lower in the TPMT*1/*3 group (0.9 reduction in thiopurine dose to approximately 50% of normal.13
vs 1.8 mg/kg/d, P < .0001). Despite dose adjustment, median However, others believe that there is insufficient published
6-TGN concentrations still were 2-fold higher in the evidence to warrant routine implementation of a reduced dose
TPMT*1/*3 group at the end of the follow-up period (505 vs in intermediate metabolizers.14 One small study showed similar
273 pmol/8 ⴛ 108 RBCs, P ⴝ .02). This difference was 3-fold therapeutic outcomes for atopic eczema when intermediate
when the concentration was adjusted for dose (578 vs 183 metabolizers received 40% of the azathioprine dose of patients
pmol/8 ⴛ 108 per mg/kg/d, P ⴝ .0007). Results were similar with the normal phenotype (1 and 2.5 mg/kg/d, respectively).15
if TPMT phenotype was used instead of genotype. Clinical Thus, although the clinical use of tests for TPMT (mainly
outcomes did not differ between groups. Conclusions: phenotype) seems to be escalating,4,16,17 more research is re-
Initial target doses to attain therapeutic 6-TGN concentra- quired to determine if TPMT-based dose adjustment can im-
tions (>235 pmol/8 ⴛ 108 RBCs) in patients with IBD prove outcomes of intermediate and normal metabolizers. We
might be 1 and 3 mg/kg/d in intermediate and normal aimed to determine the difference in thiopurine dose to attain
metabolizers, respectively. a similar outcome between individuals with the intermediate

U p to 50% of patients with inflammatory bowel disease


(IBD) treated with azathioprine and its metabolite,
6-mercaptopurine (6-MP), experience significant toxicity1,2 or
Abbreviations used in this paper: CI, confidence interval; CRP,
C-reactive protein; ESR, erythrocyte sedimentation rate; IBD, inflamma-
tory bowel disease; IQ, interquartile; 6-MMPN, 6-methylmercaptopurine
nucleotides; 6-MP, 6-mercaptopurine; RBC, red blood cell; 6-TGN, 6-thio-
inadequate response.3 As a result, much research has been guanine nucleotides; TPMT, thiopurine methyltransferase.
directed toward improving outcomes with these drugs. The © 2008 by the AGA Institute
most successful of these involves testing for thiopurine meth- 1542-3565/08/$34.00
yltransferase (TPMT),4 an enzyme that is expressed polymor- doi:10.1016/j.cgh.2008.02.032
June 2008 THIOPURINE DOSE AND TPMT STATUS 655

treatment and at the same times as the sampling for inflam-


matory markers (months 3, 6, and 9). Quality of life was
assessed every 3 months using the validated short IBD ques-
tionnaire.19 Compliance with these study assessments (blood
tests, feces samples, and questionnaire) was encouraged via
telephone contact in the week before each scheduled assess-
ment point. At these points of contact, details such as the
current thiopurine dose and concomitant drug therapy also
were recorded. All decisions regarding patient care including
initial thiopurine dose and dose adjustments were made by each
patient’s treating clinician independently of the research study.
Figure 1. Metabolism of azathioprine and 6-MP. GMPS, guanosine The clinicians could access the hematologic results from the
monophosphate synthetase; HGPRT, hypoxanthine guanine phospho- study (including TPMT activity) via the hospital intranet but
ribosyltransferase; IMPDH, inosine monophosphate dehydrogenase; not the calprotectin concentrations or short IBD questionnaire
TG, thioguanine; TGMP, thioguanine monophosphate; TGN, thiogua-
scores.
nine nucleotides; TIMP, thionosine monophosphate; TXMP, thioxan-
thosine monophosphate.
Participants were withdrawn from the study if they repeat-
edly failed to comply with drug therapy or assessments, or if
they had to stop thiopurine treatment for any reason. Subjects
genotype or phenotype of TPMT and those with the normal with side effects to their initial drug (usually azathioprine in
metabolizer status. this institution) who then commenced treatment with another
thiopurine (6-MP) were re-enrolled in the study where possible.
For these subjects, the protocol continued as if it had been
Materials and Methods uninterrupted.
Participants and Study Design
A prospective population-based study was undertaken Outcome Definitions
in Canterbury, New Zealand. The characteristics of the individ- The final tolerated thiopurine dose was defined as the
uals with IBD in Canterbury have been described previously.18 dose recorded for the patient at the last assessment point
Consecutive patients starting treatment with azathioprine or (month 9). The dose of 6-MP was adjusted to azathioprine
6-MP for IBD were identified by their gastroenterologist either equivalents by multiplying the 6-MP dose by 2.08.20 6-TGN and
via attendance at outpatient clinics or hospitalization. Patients 6-MMPN concentrations were both reported as the measured
were eligible to participate if they were at least 16 years of age (observed) values (pmol/8 ⫻ 108 RBCs) and as the concentra-
and had normal (ⱖ9.3 IU/mL) or intermediate (5–9.2 IU/mL) tion adjusted for each individual’s thiopurine dose and weight
TPMT activity. Eligibility criteria were broad to reflect the (pmol/8 ⫻ 108 RBCs per mg/kg/d). An adverse reaction to
real-life use of these drugs, with no one excluded on the basis of thiopurine treatment was defined as one that necessitated ces-
concurrent disease state or drug therapy unless these consti- sation of the drug and could be categorized as hepatotoxicity
tuted a reason to avoid thiopurine treatment (eg, pre-existing (transaminase concentration greater than 2 times the upper
neutropenia). Approval was obtained from the Canterbury Eth- limit of normal), pancreatitis (severe abdominal pain with a
ics Committee (Christchurch, New Zealand). Written informed serum amylase concentration greater than 3 times the upper
consent was obtained from all participants. limit of normal), myelosuppression (white cell count ⬍3.0 ⫻
Each subject commenced thiopurine treatment and under- 109/L and/or neutrophil count ⬍2.0 ⫻ 109/L), flu-like/hyper-
went dose adjustment and monitoring according to the usual sensitivity illness (combination of arthralgia, myalgia, fever,
practice of their gastroenterologist. This comprised assess-
and/or rash), nausea/vomiting, or rash. Decisions regarding
ment of TPMT activity (phenotype), and dose-individualiza-
drug discontinuation because of an adverse reaction were made
tion based on concentrations of the active 6-TGNs (local
by the physician responsible on a case-by-case basis.
target range, 235– 450 pmol/8 ⫻ 108 red blood cells [RBCs])
and assessment of clinical response including hematologic
monitoring. Concentrations of the major product of TPMT
Analytic Procedures
metabolism, 6-methylmercaptopurine nucleotides (6-MMPN), The concentrations of 6-TGN and 6-MMPN in RBCs
also were monitored in some patients for clinical purposes, to were determined by using a previously described high-perfor-
assess compliance, predict hepatotoxicity, and identify patients mance liquid chromatography method.21,22 Standard curves
who preferentially produce high 6-MMPN and subtherapeutic were linear over the concentration range of 30 to 2400 pmol/8 ⫻
6-TGN concentrations. Complete blood counts and liver func- 108 RBCs for 6-TGN (r2 ⬎ 0.99) and 30 to 12,000 for 6-MMPN
tion tests were monitored as per the usual practice of each (r2 ⬎ 0.99). Intraday and interday coefficients of variation were
gastroenterologist. For the purposes of this noninterventional less than 10% and the limit of quantification was approximately
study, additional assessments were undertaken at monthly in- 30 pmol/8 ⫻ 108 RBCs. TPMT activity was determined using a
tervals during the 9-month follow-up period. Complete blood radiochemical method23 based on that published by Wein-
counts and liver function tests were measured once per month, shilboum et al,24 whereas TPMT genotype was determined using
whereas inflammatory markers (C-reactive protein [CRP] and a multiplexed amplification refractory mutation system assay to
erythrocyte sedimentation rate [ESR] in blood, and calprotectin screen for TPMT*2, *3A, and *3C.25 Fecal calprotectin concen-
in feces) were determined every 3 months. 6-TGN and 6-MMPN trations were determined using a commercial single-step en-
concentrations were determined after 1 month of thiopurine zyme-linked immunosorbent assay (PhiCal Test; Calpro, Oslo,
656 GARDINER ET AL CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 6, No. 6

Norway). Liver function tests, complete blood counts, CRP, and attempted therapy with 6-MP. Five had recurrence of the ad-
ESR were determined via usual laboratory methods. verse effect (3 flu-like reaction, 1 nausea/vomiting, 1 pancreati-
tis) whereas 6 subjects (1 flu-like reaction, 4 hepatotoxicity, 1
Statistics nausea/vomiting) tolerated the 6-MP and were re-enrolled in
Statistical analyses were conducted using GraphPad the study after a median break from thiopurine treatment of 23
Prism version 4.0 for Windows (GraphPad Software, San Diego, days (interquartile [IQ] range, 14 –33 d). One of these stopped
CA). Comparisons within groups including post hoc analyses 6-MP after 3 months’ treatment because of the need for surgery.
were undertaken using the paired t test or the Wilcoxon signed This left 52 subjects who completed the 9-month evaluation on
rank test whereas comparisons between groups were made azathioprine (n ⫽ 46) or 6-MP (n ⫽ 6). The majority of these
using the unpaired t test, repeated-measures 1-way analysis of (78%) took mesalazine, which has been suggested to inhibit
variance, the Mann–Whitney U test, or the Friedman test as TPMT, but there was no difference in the median TPMT activity
appropriate. Categoric variables were compared using the chi- among those who did (12.2; IQ range, 10.3–14.3) or did not
square test. Relationships were assessed using the Pearson and (13.1; IQ range, 10.4 –14.5) take mesalazine (P ⫽ .565).
Spearman correlations for parametric and nonparametric vari-
ables, respectively. The percentage variation (r2) in dose owing
Thiopurine Dose
to TPMT activity was determined from the correlation coeffi- The 52 subjects had a mean initial thiopurine dose of
cient (r) identified in the Pearson correlation. For all statistical 72 mg/d (95% CI, 63–97 mg/d) or 1.0 mg/kg/d (95% CI, 0.9 –1.2
analyses, 2-sided P values of less than .05 were considered mg/kg/d) (as azathioprine equivalents). The mean dose in-
significant. creased during the course of the study (P ⬍ .0001), with post
hoc analyses revealing significant differences between month 0
(starting dose) and each of the subsequent monthly assessment
Results points (⬃1 vs 1.6 mg/kg/d, P ⬍ .0001) (Table 1). Most of the
Seventy-seven patients consented to participate in this dose escalation occurred within the first month of treatment,
study between September 2003 and May 2005. Seven subjects reflecting the approach of many clinicians to start treatment
subsequently were withdrawn as a result of failure to commence with a small dose and increase slowly within the first few weeks
thiopurine treatment (n ⫽ 2), poor compliance (n ⫽ 2), inabil- in an attempt to reduce early side effects. The TPMT*1/*1 and
ity to be contacted (n ⫽ 2), and early drug cessation (⬍1 week *1/*3 genotypes had comparable mean doses at baseline
of treatment) owing to surgery (n ⫽ 1). Another subject was (month 0) of 1.0 and 1.1 mg/kg/d, respectively (P ⫽ .780), and
found to be TPMT deficient and is described separately.10 The after 1 month of treatment (month 1) of 1.5 and 1.2 mg/kg/d,
remaining 69 subjects (34 men) had a mean age of 39.2 years respectively (P ⫽ .283). However, significant differences were
(95% confidence interval [CI], 35.4 – 42.9 y) and weight of 73.5 seen from months 2 (1.6 and 1.0 mg/kg/d, P ⫽ .033) to 9 (1.8
kilograms (95% CI, 69.7–77.3 kg), respectively. The majority of and 0.9 mg/kg/d, P ⫽ .0006), that is, individuals with the
subjects (76.8%) had Crohn’s disease, with the remainder having TPMT*1/*1 genotype were titrated to a dose that was 2-fold
ulcerative colitis (18.8%) or IBD unspecified (4.3%). Sixty-eight of larger than that of the TPMT*1/*3 group (Figure 2). Similar
the 69 subjects were tested for both TPMT genotype and pheno- differences in thiopurine dose were seen between those with the
type, with 61 (89.7%) and 7 (10.3%) individuals having the intermediate (5–9.2 IU/mL) versus normal (ⱖ9.3 IU/mL) pheno-
TPMT*1/*1 and *1/*3 genotype, respectively. As expected, the type (Figure 2). Approximately 30% of the variance in dose was
mean TPMT activity was higher in the TPMT*1/*1 genotype explained by TPMT activity (r2 ⫽ 0.295, P ⬍ .0001) (Figure 3).
group at 13.1 (95% CI, 12.5–13.7) IU/mL versus 8.3 (95% CI,
6.8 –9.8) IU/mL for the TPMT*1/*3 group (P ⬍ .0001). Geno- 6-Thioguanine Nucleotide and
type did not predict phenotype in 6 of 68 subjects (8.8%). Four 6-Methylmercaptopurine Nucleotide
of 61 subjects (6.6%) with the TPMT*1/*1 genotype had activity Concentrations
(7.9 –9.2 IU/mL) in the intermediate range of 5 to 9.2 IU/mL, The mean 6-TGN concentrations for the 52 subjects
whereas 2 of 7 (28.6%) subjects with the TPMT*1/*3 genotype (including the 5 subjects who had switched to 6-MP within the
had activity (9.8 and 9.9 IU/mL) in the normal range of 9.3 to first 1–2 months of treatment) were stable during evaluation,
17.6 IU/mL. It should be noted that the radiochemical assay and were approximately 270 to 280 pmol/8 ⫻ 108 RBCs at
used in our study and institution results in lower ranges for months 1, 3, 6, and 9 (Table 1). Mean 6-TGN concentrations at
TPMT activity than identified via the high-performance liquid the final assessment point were 2-fold higher in the TPMT*1/*3
chromatography methods used in some laboratories. For exam- genotype than the TPMT*1/*1 group at 505 pmol/8 ⫻ 108 RBCs
ple, Prometheus (San Diego, CA) reports a range of 6.7 to 23.6 (95% CI, 188 – 823 pmol/8 ⫻ 108 RBCs) versus 273 pmol/8 ⫻ 108
enzyme units for the intermediate group whereas those with RBCs (95% CI, 234 –312 pmol/8 ⫻ 108 RBCs) despite receiving
activity greater than 23.6 enzyme units are classified as normal a 50% lower thiopurine dose (P ⫽ .016). This difference was
metabolizers. 3-fold when the 6-TGN concentration was dose- and weight-
Forty-seven of the 69 subjects (68%) completed the 9-month adjusted, at 578 pmol/8 ⫻ 108 RBCs per mg/kg/d (95% CI,
follow-up on their original thiopurine drug, which was azathio- 407–749 pmol/8 ⫻ 108 RBCs per mg/kg/d) versus 183 pmol/8
prine in all but 1 case. The remaining 22 (32%) subjects devel- ⫻ 108 RBCs per mg/kg/d (95% CI, 142–224 pmol/8 ⫻ 108 RBCs
oped toxicity (8 flu-like reactions, 6 hepatotoxicity, 4 nausea/ per mg/kg/d), respectively (P ⫽ .0007) (Figure 4). Twenty-four
vomiting, 2 pancreatitis, 1 headache, and 1 abdominal pain) of 49 subjects (49%) with evaluable data had 6-TGN concentra-
that necessitated discontinuation of azathioprine, which oc- tions outside the local therapeutic range of 235 to 450 pmol/8
curred after a median of 30 days (range, 7–99 d) of treatment. ⫻ 108 RBCs after 9 months of treatment. Those with the
After resolution of the adverse reaction, 11 of these 22 subjects TPMT*1/*1 genotype were more likely than those with the
Table 1. Thiopurine Dose, Metabolite Concentrations, and Markers of Disease Activity

Month P
June 2008

valuea
(between
0 1 2 3 4 5 6 7 8 9 months)

Dose, mg 72b (63–97) 111 (97–125) 110 (97–123) 111 (98–124) 116 (103–129) 118 (105–130) 117 (104–130) 119 (106–131) 121 (108–134) 122 (109–135) <.0001
Dose, mg/kg 1.04b (0.90–1.18) 1.56 (1.37–1.74) 1.56 (1.37–1.75) 1.57 (1.39–1.74) 1.64 (1.46–1.81) 1.65 (1.48–1.82) 1.64 (1.46–1.81) 1.66 (1.49–1.82) 1.64 (1.47–1.82) 1.66 (1.48–1.84) <.0001
6-TGN, pmol/8 ⫻ 274 (183–372) 266 (182–374) 266 (214–407) 282 (195–401) .797
108 RBCs
6-MMPN, pmol/8 ⫻ 571 (364–1407) 438 (240–1278) 511 (216–1716) 408 (275–1117) .829
108 RBCs
Calprotectin, ␮g/g 370c (64–881) 82 (26–361) 182 (42–403) 158 (50–444) .001
CRP, mg/L 9d (4–29) 5 (4–13) 5 (3–12) 4 (3–8) .009
ESR, mm/h 13 (5–27) 10 (5–24) 10 (6–21) 9 (5–19) .539
Short IBD 40e (30–52) 49f (42–63) 55 (47–62) 56 (47–66) <.0001
questionnaire

NOTE. Mean (95% CI) shown for thiopurine dose, and median (IQ range) shown for metabolite concentrations markers of disease activity.
aRepeated-measures 1-way analysis of variance (dose) or Friedman test (metabolite concentrations and markers of disease activity).
bMonth 0 versus months 1 to 9 (P ⬍ .0001 each).
cMonth 0 versus month 3 (P ⬍ .001), vs month 6 (P ⬍ .01), and vs month 9 (P ⫽ .02).
dMonth 0 versus month 3 (P ⬍ .01), vs month 6 (P ⬍ .01), and vs month 9 (P ⬍ .001).
eMonth 0 versus month 3 (P ⬍ .0001), vs month 6 (P ⬍ .0001), and vs month 9 (P ⬍ .0001).
fMonth 3 versus month 6 (P ⬍ .01), and vs month 9 (P ⫽ .02).

TPMT activity (r ⫽ ⫺0.509, P ⫽ .0002) (Figure 5).


pmol/8 ⫻ 108 RBCs (1.7 and 1.8 mg/kg/d, respectively).
THIOPURINE DOSE AND TPMT STATUS

Figure 3. TPMT activity versus weight-adjusted thiopurine dose.


and 660 pmol/8 ⫻ 108 RBCs (IQ range, 334 –1470 pmol/8 ⫻
pmol/8 ⫻ 108 RBCs (IQ range, 86 –185 pmol/8 ⫻ 108 RBCs)
among those who had concentrations above or below 235
concentrations above the range (4 of 44 vs 2 of 5, P ⫽ .046).

with the TPMT*1/*3 versus TPMT*1/*1 genotypes at 154


Median 6-MMPN concentrations were lower in individuals
.09) or TPMT activity (r ⫽ ⫺0.269, P ⫽ .059). However, when
cantly with either thiopurine dose (mg/kg/d) (r ⫽ ⫺0.250, P ⫽
TPMT*1/*1 (⌬) and *1/*3 (Œ) genotypes and in (B) normal (⌬, ⱖ9.3

(weight-adjusted) there was a significant relationship with


6-TGN concentrations were adjusted for thiopurine dose
Measured 6-TGN concentrations did not correlate signifi-
Within the TPMT*1/*1 group, there was no difference in dose
44 vs 0 of 5, respectively; P ⫽ .072) but were less likely to have
*1/*3 genotype to have concentrations below the range (18 of
657

IU/mL) phenotypes and intermediate (Œ, 5–9.2 IU/mL) and presented


Figure 2. (A) Thiopurine dose (as azathioprine equivalents) in

as mean and 95% CI. †Groups became significantly different (P ⬍ .05).


658 GARDINER ET AL CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 6, No. 6

month 3 for calprotectin, CRP, and short IBD scores, respec-


tively) and were maintained during the 9-month follow-up
(Table 1). There was no significant change in ESR during the
9-month follow-up period (P ⫽ .539). The percentage change in
calprotectin (P ⫽ .352), CRP (P ⫽ .921), ESR (P ⫽ .597), or
short IBD questionnaire (P ⫽ .714) did not correlate with
thiopurine dose (mg/kg/d), and only the percentage change in
short IBD score correlated significantly with 6-TGN concentra-
tions at 9 months (r ⫽ 0.371, P ⫽ .01) (data not shown).
The patients with TPMT*1/*1 versus *1/*3 genotypes did
not experience any difference in the percentage change in cal-
protectin, CRP, ESR, and short IBD questionnaire from base-
line, despite a 2-fold difference in 6-TGN concentrations (data
not shown). Consistent with this, no significant differences
were observed between the TPMT*1/*3 versus *1/*1 genotypes
in the proportion of patients on steroids at completion of the
study (2 of 5 and 7 of 46, respectively; P ⫽ .167) or the median
lymphocyte count (1.5 ⫻ 109/L each).
TPMT genotype did not relate to azathioprine toxicity, with
the TPMT*1/*3 genotype comprising approximately 10% of
each of the tolerator and nontolerator groups. Further, mean
TPMT activity was comparable in the 2 groups at 12.6 (95% CI,
11.8 –13.4) and 12.8 (95% CI, 11.4 –14.1), respectively (P ⫽ .779).
There were insufficient data to compare thiopurine dose be-
tween the tolerators and nontolerators of azathioprine. How-
ever, 14 of the 22 nontolerators underwent blood sampling for
metabolite concentrations within 2 days of stopping azathio-
prine (⬃30 days into treatment), enabling crude comparison
with the tolerator group at the month 1 (30 day) assessment
point. There was a significant difference (P ⫽ .024) in median
6-TGN concentration between tolerators and nontolerators at
265 pmol/8 ⫻ 108 RBCs (IQ range, 180 –370 pmol/8 ⫻ 108 RBCs)
and 165 pmol/8 ⫻ 108 RBCs (IQ range, 130 –259 pmol/8 ⫻ 108
RBCs), respectively. However, this was in the opposite direction to
what might be expected if toxicity was concentration related. This
may reflect the blood sampling approximately 2 days after drug
cessation and the use of lower does in individuals with adverse
effects. The median 6-MMPN concentration in the tolerators at
month 1 was 583 pmol/8 ⫻ 108 RBCs (IQ range, 291–1422
Figure 4. (A) Actual and (B) dose-adjusted 6-TGN concentrations ver-
pmol/8 ⫻ 108 RBCs), which was comparable with the median of
sus TPMT genotype at month 9. Data are presented as mean and 95%
CI. Mean actual and dose-adjusted 6-TGN concentrations were 2-fold
339 pmol/8 ⫻ 108 RBCs (IQ range, 245–1220 pmol/8 ⫻ 108
(P ⫽ .016) and 3-fold higher (P ⫽ .0007) in the TPMT*1/*3 versus the RBCs) in the nontolerators (P ⫽ .288).
*1/*1 genotype, respectively.
Discussion
The principal aim of this prospective study was to
108 RBCs), respectively (P ⫽ .001). This difference was less determine the difference in thiopurine dose requirements in
impressive when 6-MMPN concentrations were adjusted for
thiopurine dose at 218 pmol/8 ⫻ 108 RBCs per mg/kg/d (IQ
range, 126 –260 pmol/8 ⫻ 108 RBCs per mg/kg/d) versus 411
pmol/8 ⫻ 108 RBCs per mg/kg/d (IQ range, 231– 804 pmol/8 ⫻
108 RBCs per mg/kg/d), respectively (P ⫽ .058). 6-MMPN con-
centrations correlated with both thiopurine dose (r ⫽ 0.610, P ⬍
.0001) and TPMT activity (r ⫽ 0.348, P ⫽ .014) (data not shown).

Clinical Outcomes
Within subject calprotectin concentrations (P ⫽ .001),
CRP (P ⫽ .009) and short IBD questionnaire scores (P ⬍ .0001)
improved significantly during the course of the 9-month study
(Table 1). The improvements were seen at the first assessment
point (3 months) after initiation of thiopurine treatment (P ⬍ Figure 5. Dose-adjusted 6-TGN concentrations versus TPMT activity
.001, P ⬍ .01, and P ⬍ .0001 for comparisons of month 0 vs (includes line of best fit plus 95% CI of this line).
June 2008 THIOPURINE DOSE AND TPMT STATUS 659

individuals with intermediate versus normal TPMT metabolizer 6-TGN concentrations, which should be maintained at a rea-
status. The mean initial thiopurine dose (as azathioprine equiv- sonable level (perhaps no higher than 400 –500 pmol/8 ⫻ 108
alents) was similar (⬃1 mg/kg/d) in the 2 groups but with RBCs) because increased concentrations also have been linked
continued treatment individuals with the TPMT*1/*1 genotype with nodular regenerative hyperplasia.28,29 The reduced dose
were titrated to a dose that was 2-fold higher than that used in approach (1 mg/kg/d) in intermediate metabolizers already has
the TPMT*1/*3 genotype group (1.8 and 0.9 mg/kg/d, respec- been implemented in the practice of some clinicians30,31 despite
tively). Despite this difference, individuals with the TPMT*1/*3 the limited supportive evidence. Because there is a 3-fold dif-
genotype attained a 2-fold higher mean concentration of the ference in the dose required to achieve comparable 6-TGN
active 6-TGN metabolites (505 pmol/8 ⫻ 108 RBCs) than the concentrations in the normal metabolizers, it seems reasonable
wild-type (273 pmol/8 ⫻ 108 RBCs; P ⫽ .02). Further, the mean to aim for 3 mg/kg/d in the normal metabolizer group, which
difference in 6-TGN concentrations between groups was 3-fold is consistent with the upper target dose recommended for
when each individual’s 6-TGN concentrations were adjusted for patients with IBD, irrespective of TPMT status, in some guide-
their thiopurine dose (578 vs 183 pmol/8 ⫻ 108 RBCs per lines.32
mg/kg/d; P ⫽ .0007). This suggests that individuals with the Although either genotyping or phenotyping for TPMT can
TPMT*1/*3 genotype require, on average, one third of the dose be used to facilitate individualization of thiopurine dose, phe-
of those with the normal genotype to achieve comparable notyping may have an advantage over genotyping because
6-TGN concentrations. TPMT activity varies approximately 4-fold across normal and
These principal findings may not seem surprising in light of intermediate metabolizers, and varies inversely with 6-TGN
the mechanisms for dose adjustments in this study. The clini- concentrations as shown in the current study. However, our
cians adjusted dose via their usual methods, which included study suggests that TPMT activity explains only 30% of the
consideration of TPMT activity and 6-TGN concentrations and variation in thiopurine dose in the absence of consideration of
overall an improvement in IBD disease activity (calprotectin, the 6-TGN concentrations achieved. Examination of further
CRP, and IBD questionnaire score) was seen (Table 1). The final factors influencing thiopurine dose were beyond the scope of this
doses achieved, with an approximately 2-fold difference be- study. Further study is needed to determine whether dosing in
tween TPMT*1/*1 and *1/*3 genotypes, are consistent with the direct relationship to measured TPMT activity enables better in-
suggestion from previous researchers that individuals with the dividualization of thiopurine dose than classification of an indi-
intermediate metabolizer status may require half of the dose of vidual as 1 of 3 genotypes or phenotypes.
normal metabolizers.13 However, if 6-TGN concentrations are Overall, the findings of this study suggest that intermediate
used to indicate likely efficacy, the true difference in dose could metabolizers should receive approximately one third of the dose
be 3-fold. The use of 6-TGN concentrations as a clinical end of normal metabolizers to achieve similar 6-TGN concentra-
point is reasonable because concentrations above 235 to 260 tions. Our results support a target dose of 3 mg/kg/d in normal
pmol/8 ⫻ 108 RBCs are associated with a 3-fold greater likeli- metabolizers and around 1 mg/kg/d for intermediate metabo-
hood of remission (odds ratio, 3.27; 95% CI, 1.71– 6.27).26 The lizers. Lower initial starting doses, for example, 2 and 0.5 mg/
lack of observation of any other clinical differences (eg, change kg/d, initially may help to minimize the occurrence of some of
in inflammatory markers from baseline) between the 2 groups, the nonmyelosuppression-related toxicities of this class such as
despite a 2-fold difference in measured 6-TGN concentrations, nausea and vomiting or hepatotoxicity, which seem unrelated
may reflect some of the difficulties in using clinical end points to either TPMT activity or 6-TGN concentrations. Although
to assess efficacy (eg, white cell count, steroid use), the small these adverse effects usually are regarded as idiosyncratic, there
number of intermediate metabolizers studied, and the complex- is some evidence of a dose-response relationship.33 Because the
ity of thiopurine pharmacokinetics. upper limit for 6-TGN concentrations is poorly established and
More detailed examination of the 6-TGN concentrations other active metabolites exist, dose adjustments should always
achieved in the study reinforces the need for differential dose occur in conjunction with conventional monitoring including
requirements in the 2 TPMT genotype groups. Half of all par- complete blood counts and liver function tests.
ticipants (24 of 49 subjects with evaluable data) achieved
6-TGN concentrations outside the target range of 235 to 450 References
pmol/8 ⫻ 108 RBCs. Importantly, almost half of those with
1. Sandborn W, Sutherland L, Pearson D, et al. Azathioprine or
normal metabolizer status (18 of 44) had 6-TGN concentra-
6-mercaptopurine for induction of remission in Crohn’s disease.
tions of less than 235 pmol/8 ⫻ 108 RBCs on a mean dose of 1.8 Cochrane Database Syst Rev 2000;2:CD000545.
mg/kg/d. This may suggest inadequate drug exposure for many 2. Gearry RB, Barclay ML, Burt MJ, et al. Thiopurine drug adverse
patients, although it also is possible that some patients effects in a population of New Zealand patients with inflammatory
achieved adequate disease control with values less than 235 bowel disease. Pharmacoepidemiol Drug Saf 2004;13:563–567.
pmol/8 ⫻ 108 RBCs, or experienced toxicity with higher con- 3. Pearson DC, May GR, Fick GH, et al. Azathioprine and 6-mercap-
centrations. All 5 individuals with the TPMT*1/*3 genotype and topurine in Crohn disease: a meta-analysis. Ann Intern Med
evaluable data had 6-TGN concentrations above 235 pmol/8 ⫻ 1995;123:132–142.
108 RBCs (276, 417, 424, 465, and 945 pmol/8 ⫻ 108 RBCs) on 4. Gardiner SJ, Begg EJ. Pharmacogenetics testing for drug metab-
olizing enzymes—is it happening in practice? Pharmacogenet
a mean dose of approximately 0.9 mg/kg/d. Because the upper
Genomics 2005;15:365–369.
limit of the target range (450 pmol/8 ⫻ 108 RBCs) is poorly 5. Schaeffeler E, Fischer C, Brockmeier D, et al. Comprehensive anal-
established27 and there was no evidence of leukopenia in any of ysis of thiopurine S-methyltransferase phenotype-genotype correla-
the study participants, it seems reasonable to aim for a mean tion in a large population of German-Caucasians and identification of
dose of 1 mg/kg/d in the intermediate metabolizers. However, novel TPMT variants. Pharmacogenetics 2004;14:407– 417.
the dose then should be tailored against clinical outcomes and 6. Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenet-
660 GARDINER ET AL CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 6, No. 6

ics: monogenic inheritance of erythrocyte thiopurine ferese and 6-thioguanine nucleotide measurement: early
methyltransferase activity. Am J Hum Genet 1980;32:651– 662. experience of use in clinical practice. Intern Med J 2005;35:
7. Evans WE, Horner M, Chu YQ, et al. Altered mercaptopurine 580 –585.
metabolism, toxic effects and dosage requirement in a thiopurine 22. Gardiner SJ, Gearry RB, Roberts RL, et al. Exposure to thiopurine
methyltransferase-deficient child with acute lymphocytic leuke- drugs through breast milk is low based on metabolite concentra-
mia. J Pediatr 1991;119:985–989. tions in mother-infant pairs. Br J Clin Pharmacol 2006;62:
8. Lennard L, Gibson BE, Nicole T, et al. Congenital thiopurine 453– 456.
methyltransferase deficiency and 6-mercaptopurine toxicity dur- 23. Sies C, Florkowski CM, George PM, et al. Measurement of thio-
ing treatment for acute lymphoblastic leukaemia. Arch Dis Child purine methyl transferase activity guides does-initiation and pre-
1993;69:577–579. vents toxicity from azathioprine. N Z Med J 2005;118:1210.
9. Kaskas BA, Louis E, Hindorf U, et al. Safe treatment of thiopurine 24. Weinshilboum RM, Raymond FA, Pazmino PA. Human erythrocyte
S-methyltransferase deficient Crohn’s disease patients with aza- thiopurine methyltransferase: radiochemical microassay and bio-
thioprine. Gut 2003;52:140 –142. chemical properties. Clin Chim Acta 1978;85:323–333.
10. Gardiner SJ, Gearry RB, Barclay ML, et al. Two cases of TPMT 25. Roberts RL, Barclay ML, Gearry RB, et al. A multiplexed allele-
deficiency—a lucky save and a near miss with azathioprine. Br J specific polymerase chain reaction assay for the detection of
Clin Pharmacol 2006;62:473– 476. common thiopurine S-methyltransferase (TPMT) mutations. Clin
11. Lennard L, van Loon J, Lilleyman JS, et al. Thiopurine pharmaco- Chim Acta 2004;341:49 –53.
genetics in leukemia: correlation of erythrocyte thiopurine meth- 26. Osterman MT, Kundu R, Lichtenstein GR, et al. Association of
yltransferase activity and 6-thioguanine nucleotide concentra- 6-thioguanine nucleotide levels and inflammatory bowel disease
tions. Clin Pharmacol Ther 1987;41:18 –25. activity: a meta-analysis. Gastroenterology 2006;130:1047–
1053.
12. Lennard L, Lilleyman JS, Van Loon J, et al. Genetic variation in
27. Schutz E, Gummert J, Mohr FW, et al. Should 6-thioguanine
response to 6-mercaptopurine for childhood acute lymphoblastic
nucleotides be monitored in heart transplant recipients given
leukaemia. Lancet 1990;336:225–229.
azathioprine? Ther Drug Monit 1996;18:228 –233.
13. Sanderson J, Ansari A, Marinaki T, et al. Thiopurine methyltrans-
28. de Boer NKH, Mulder DJJ, Van Bodegraven AA. Nodular regener-
ferase: should it be measured before commencing thiopurine
ative hyperplasia and thiopurines: the case for level-dependent
drug therapy? Ann Clin Biochem 2004;41:294 –302.
toxicity. Liver Transplantation 2005;11:1300 –1301.
14. Lichtenstein GR. Monitoring 6-mercaptopurine/azathioprine me-
29. Dubinsky MC, Vasiliauskas EA, Singh H, et al. 6-Thioguanine can
tabolite levels. Am J Gastroenterol 2007;102:S14 –S17.
cause serious liver injury in inflammatory bowel disease patients.
15. Meggitt SJ, Gray JC, Reynolds NJ. Azathioprine dosed by thiopurine
Gastroenterology 2003;125:298 –303.
methyltransferase activity for moderate-to-severe atopic eczema: a
30. Dassopoulos T. Pharmacogenomics of IBD therapies. Gastroen-
double-blind, randomised controlled trial. Lancet 2006;367:839 – terol Hepatol 2007;3:338 –339.
846. 31. Cuffari C. A physician’s guide to azathioprine metabolite testing.
16. Tan BB, Lear JT, Gawkrodger DJ, et al. Azathioprine in dermatol- Gastroenterol Hepatol 2006;2:58 – 63.
ogy: a survey of current practice in the U.K. Br J Dermatol 32. Lichtenstein GR, Abreu MT, Cohen R, et al. American Gastroen-
1997;136:351–355. terological Association Institute technical review on corticoste-
17. Fargher EA, Tricker K, Newman W, et al. Current use of pharma- roids, immunomodulators, and infliximab in inflammatory bowel
cogenetic testing: a national survey of thiopurine methyltrans- disease. Gastroenterology 2006;130:940 –987.
ferase testing prior to azathioprine prescription. J Clin Pharm 33. Gisbert JP, Gonzalez-Lama Y, Mate J. Thiopurine-induced liver
Ther 2007;32:187–195. injury in patients with inflammatory bowel disease: a systematic
18. Gearry RB, Richardson A, Frampton CMA, et al. High incidence of review. Am J Gastroenterol 2007;102:1518 –1527.
Crohn’s disease in Canterbury, New Zealand: results of an epi-
demiological study. Inflamm Bowel Dis 2006;12:936 –943.
19. Irvine EJ, Zhou Q, Thompson AK. The short inflammatory bowel Address requests for reprints to: Sharon Gardiner, PhD, Department
disease questionnaire: a quality of life instrument for community of Medicine, Christchurch School of Medicine, P.O. Box 4345,
physicians managing inflammatory bowel disease. Am J Gastro- Christchurch, New Zealand. e-mail: sharon.gardiner@cdhb.govt.nz; fax:
enterol 1996;91:1571–1578. (64) 3-364-1003.
20. Sandborn WJ. A review of immune modifier therapy for inflamma- The authors wish to acknowledge the University of Otago, Tertiary
tory bowel disease: azathioprine, 6-mercaptopurine, cyclosporine Education Commission, Canterbury Medical Research Foundation, and
and methotrexate. Am J Gastroenterol 1997;91:423– 433. the Jim and Mary Carney Charitable Trust for financial support of this
21. Gearry RB, Barclay ML, Roberts RL, et al. Thiopurine methyltrans- study.

You might also like