Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2017

Open Access 01.12.2017 | Research article

The effect of kidney function on the urate lowering effect and safety of increasing allopurinol above doses based on creatinine clearance: a post hoc analysis of a randomized controlled trial

verfasst von: Lisa K. Stamp, Peter T. Chapman, Murray Barclay, Anne Horne, Christopher Frampton, Paul Tan, Jill Drake, Nicola Dalbeth

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2017

Abstract

Background

The use of allopurinol in people with chronic kidney disease (CKD) remains one of the most controversial areas in gout management. The aim of this study was to determine the effect of baseline kidney function on safety and efficacy of allopurinol dose escalation to achieve serum urate (SU) <6 mg/dl.

Methods

We undertook a post hoc analysis of a 24-month allopurinol dose escalation treat-to-target SU randomized controlled trial, in which 183 people with gout were randomized to continue current dose allopurinol for 12 months and then enter the dose escalation phase or to begin allopurinol dose escalation immediately. Allopurinol was increased monthly until SU was <6 mg/dl. The effect of baseline kidney function on urate lowering and adverse effects was investigated.

Results

Irrespective of randomization, there was no difference in the percentage of those with creatinine clearance (CrCL) <30 ml/min who achieved SU <6 mg/dl at the final visit compared to those with CrCL ≥30 to <60 ml/min and those with CrCL ≥60 ml/min, with percentages of 64.3% vs. 76.4% vs. 75.0%, respectively (p = 0.65). The mean allopurinol dose at month 24 was significantly lower in those with CrCL <30 ml/min as compared to those with CrCL ≥30 to <60 ml/min or CrCL ≥60 ml/min (mean (SD) 250 (43), 365 (22), and 460 (19) mg/day, respectively (p < 0.001)). Adverse events were similar among groups.

Conclusions

Allopurinol is effective at lowering urate even though and accepting that there were small numbers of participants with CrCL <30 ml/min, these data indicate that allopurinol dose escalation to target SU is safe in people with severe CKD. The dose required to achieve target urate is higher in those with better kidney function.

Trial registration

Australian and New Zealand Clinical trials Registry, ACTRN12611000845​932. Registered on 10 August 2011.
Abkürzungen
ACR
American College of Rheumatology
AE
Adverse event
AHS
Allopurinol hypersensitivity syndrome
ALP
Alkaline phosphatase
ALT
Alanine transaminase
ANOVA
Analysis of variance
AST
Aspartate aminotransferase
CKD
Chronic kidney disease
control/DE
Dose escalation phase group
CrCL
Creatinine clearance
CTCAE
Common Terminology Criteria for Adverse Events
DE/DE
Immediate dose escalation group
EULAR
European League Against Rheumatism
GGT
Gamma glutamyl transferase
HAQ
Health assessment questionnaire
N
Number
NSAID
Non steroidal anti-inflammatory drug
SAE
Serious adverse event
SD
Standard deviation
SE
Standard error
SJC
Swollen joint count
SU
Serum urate
TJC
Tender joint count
ULT
Urate lowering therapy
VAS
Visual analogue scale

Background

The use of allopurinol in people with chronic kidney disease (CKD) remains one of the most controversial areas in gout management. While recommendations from the American College of Rheumatology (ACR) [1] and European League Against Rheumatism (EULAR) [2] both advocate allopurinol as a first-line urate lowering therapy (ULT), for dosing guidance the ACR advocates a gradual dose escalation even in those with CKD [1], while EULAR recommends dose restriction based on creatinine clearance (CrCL) [2]. This discrepancy is due to concerns over increased risk of adverse effects, particularly allopurinol hypersensitivity syndrome (AHS) and limited data on the use of allopurinol in CKD.
Since the initial allopurinol dosing guidelines by Hande et al., based on an association between allopurinol dose, oxypurinol and AHS, were published [3], a number of other risk factors for AHS have been identified [4]. The Hande dosing strategy does not differentiate between starting dose, which has been associated with AHS [5], and maintenance dose (i.e. dose required to achieve target serum urate (SU)). Once AHS occurs, people with CKD have higher mortality [6].
Clinical trials of ULT have excluded those with significant CKD and restricted the allopurinol dose to ≤ 300 mg daily even in those with normal kidney function [79]. We have previously reported results from a randomized controlled trial of allopurinol dose escalation to achieve target urate in which 52% of the 183 participants had CrCL <60 ml/min, suggesting that such an approach is effective and safe [10]. Herein we undertook a post hoc analysis to determine the effect of baseline kidney function on the safety and efficacy of the allopurinol dose escalation strategy.

Methods

Study design

A 24-month open, randomized, controlled, parallel-group, comparative clinical trial was undertaken (ACTRN12611000845932). Ethical approval was obtained from the Multi-Regional Ethics Committee, New Zealand. Written informed consent was obtained from each participant. Full methods have been reported previously [10, 11]. In brief, 183 people with gout as defined by the American Rheumatism Association 1977 preliminary classification criteria for gout [12], receiving at least the CrCl based dose of allopurinol for  ≥ 1 month and with SU ≥6 mg/dl were recruited. People with a history of intolerance to allopurinol and those receiving azathioprine were excluded. Chronic kidney disease was not an exclusion criterion. Participants were randomized to continue current dose allopurinol for 12 months and then enter the dose escalation phase (control/DE) or to begin allopurinol dose escalation immediately (DE/DE). Allopurinol was increased monthly until SU was <6 mg/dl; in those with CrCL ≤60 ml/min allopurinol was increased by 50 mg increments and in those with CrCL >60 ml/min it was increased by 100 mg. Participants were not stratified by renal function at randomization. For the purposes of this post hoc analysis, participants were grouped according to kidney function at baseline as having (1) none/mild impairment, CrCL ≥60 ml/min (CKD stage 1 and 2), (2) moderate impairment, CrCL ≥30 to <60 ml/min (CKD stage 3) and (3) severe impairment, CrCL <30 ml/min (CKD stage 4 and 5).

Adverse and serious advent event reporting

Treatment-emergent adverse events (AE) were defined as any AE occurring after entry into the study until the end of month 24. Worsening laboratory-defined AEs were those where there was an increase in AE grade from baseline between month 12 and month 24, using the Common Terminology Criteria for Adverse Events (CTCAE v4.0).

Study outcomes

The primary efficacy outcome was reduction in SU at the final visit (month 24 or the final visit for those deceased or lost to follow up). Secondary efficacy outcomes included (1) the proportion of participants reaching target SU levels from baseline to months 12 and 24 and from months 12 to 24, (2) the percentage reduction in SU from baseline to months 12 and 24 and from months 12 to 24 and (3) the dose of allopurinol required to achieve SU <6 mg/dl. The primary safety outcome was serious adverse events (SAEs) and treatment-emergent or worsening AEs related to liver or kidney function.

Statistical analysis

Baseline demographics and clinical features were summarized using standard descriptive statistics including mean, standard deviation (SD), range, frequency and percent as appropriate.
Changes from baseline to months 12 and 24 and from month 12 to month 24, and levels at 12 and 24 months were compared between kidney function groups using analysis of variance (ANOVA), which included the randomized group and the interaction between the randomized group and kidney function groups as factors. Comparisons of baseline levels were compared using one-way ANOVA, which only included kidney function group as the factor. Dichotomous outcome measures were compared using logistic regression, which included CrCL and randomized group and the interaction between the randomized group and kidney function groups as factors. A two-tailed p value <0.05 was taken to indicate statistical significance.

Results

Demographics

There were 183 participants who entered the study; 93 in the control/DE group (n = 14 with CrCL <30 ml/min; n = 31 with CrCL ≥30 to <60 ml/min; n = 48 with CrCL ≥60 ml/min 48) and 90 in the DE/DE group (n = 10 with CrCL <30 ml/min; n = 40 with CrCL ≥30 to <60 ml/min; n = 40 with CrCL ≥60 ml/min). There were 143 participants who completed the month-12 visit; 73 in the control/DE group (n = 8 with CrCL <30 ml/min; n = 24 with CrCL ≥30 to <60 ml/min; n = 41 with CrCL ≥60 ml/min) and 70 in the DE/DE group (n = 7 with CrCL <30 ml/min; n = 35 with CrCL ≥30 to <60 ml/min; n = 28 with CrCL ≥60 ml/min). There were 137 participants who completed the month-24 visit; 68 in the control/DE group (n = 7 with CrCL <30 ml/min; n = 22 with CrCL ≥30 to <60 ml/min; n = 39 with CrCL ≥60 ml/min) (73.1%) and 69 in the DE/DE group (n = 7 with CrCL <30 ml/min; n = 33 with CrCL ≥30 to <60 ml/min; n = 29 with CrCL ≥60 ml/min). Demographics for each of the randomized groups according to baseline kidney function group are outlined in Table 1.
Table 1
Participant baseline demographics and clinical features
Variable
Control/dose escalation (n = 93)
Dose escalation/dose escalation (n = 90)
CrCL <30 ml/min (n = 14)
CrCL ≥30 to <60 ml/min (n = 31)
CrCL ≥60 ml/min (n = 48)
CrCL <30 ml/min (n = 10)
CrCL ≥30 to <60 ml/min (n = 40)
CrCL ≥60 ml/min (n = 40)
Age yearsa
68.2 (14.2)
66.6 (9.3)
54.8 (11.9)
66.8 (12.5)
64.6 (9.6)
52.5 (12.1)
Male, n (%)
7 (50%)
25 (80.6%)
46 (95.8)
9 (90%)
34 (85%)
39 (97.5)
Ethnicity, n (%)
 NZ European
6 (42.9%)
13 (41.9%)
20 (41.7%)
2 (20%)
21 (52.5%)
14 (35%)
 Maori
3 (21.4%)
10 (32.3%)
9 (18.8%)
3 (30.0%)
13 (32.5%)
13 (32.5%)
 Pacific Island
4 (28.6%)
6 (19.4%)
17 (35.4%)
5 (50%)
5 (12.5%)
9 (22.5%)
 Asian
1 (7.1)
2 (6.5%)
1 (2.1%)
0 (0%)
1 (2.5%)
4 (10.0%)
 Other
0 (0%)
0 (0%)
1 (2.1%)
0 (0%)
0 (0%)
0 (0%)
Duration of gout (years)
16.8 (14.8)
18.2 (14.7)
18.1 (11.9)
13.1 (11.2)
16.9 (11.2)
16.9 (11.2)
Baseline serum urate mg/dla
8.3 (1.5)
7.1 (1.6)
6.8 (1.5)
8.0 (1.6)
7.6 (1.6)
6.5 (1.3)
CrCL (ml/min)
19.8 (5.9)
44.3 (7.9)
82.4 (16.6
21.1 (6.7)
44.5 (8.1)
85.5 (17.7)
Body mass index (kg/m2)a
34.6 (7.2)
35.8 (8.3)
35.1 (7.5)
36.9 (8.4)
35.9 (8.4)
33.7 (6.8)
Baseline allopurinol dose mg/dayb
135.7 (100-250)
258.1 (150-400)
328.1 (200-600)
160.0 (100-300)
231.9 (100-600)
317.5 (150-600)
Allopurinol dose, n (%)
  ≤ 200 mg/day
13 (92.9%)
13 (41.9%)
5 (10.4%)
9 (90%)
25 (62.5%)
3 (7.5%)
  > 200–300 mg/day
1 (7.1%)
16 (51.6%)
33 (68.8%)
1 (10%)
13 (32.5%)
32 (80%)
  > 300 mg/day
0 (0%)
2 (6.5%)
10 (20.8%)
0 (0%)
2 (5%)
7 (7.8%)
Presence of palpable tophi, n (%)
10 (71.4%)
14 (45.2%)
22 (45.8%)
4 (40%)
13 (32.5%)
18 (45%)
Co-existing conditions, n (%)
 Obesityc
11 (78.6%)
23 (74.2%)
36 (75%)
8 (80%)
29 (72.5%)
27 (67.5%)
 Kidney stones
0 (0%)
1 (3.2%)
2 (4.2%)
1 (10%)
3 (7.5%)
1 (2.5%)
 Cardiovascular diseased
13 (92.9%)
14 (45.2%)
11 (22.9%)
5 (50%)
26 (65%)
10 (25%)
 Diabetes mellitus
8 (57.1%)
12 (38.7%)
13 (27.1%)
7 (70%)
18 (45%)
4 (10%)
 Hypertension
11 (78.6%)
29 (93.5%)
25 (52.1%)
9 (90%)
36 (90%)
22 (55%)
 Hyperlipidemia
12 (85.7%)
19 (61.3%)
27 (56.3%)
7 (70%)
22 (55%)
18 (45%)
Concurrent medications, n (%)
 Diuretic
13 (92.9)
19 (61.3%)
11 (22.9%)
7 (70%)
23 (57.5%)
8 (20.0%)
 Aspirin
11 (78.6%)
17 (54.8%)
13 (27.1%)
7 (70%)
23 (57.5%)
10 (25%)
Any anti-inflammatory prophylaxis
5 (35.7%)
15 (48.4%)
25 (52.1%)
4 (40%)
24 (60%)
23 (57.5%)
 Colchicine
2 (14.3%)
11 (25.5%)
22 (45.8%)
3 (30%)
13 (32.5%)
18 (45%)
 NSAID
0 (0%)
3 (9.7%)
6 (12.5%)
2 (20%)
4 (10%)
9 (22.5%)
 Prednisone
3 (21.4%)
8 (19.4%)
3 (6.3%)
1 (10%)
9 (22.5%)
2 (5%)
CrCL creatinine clearance, NSAID non steroidal anti-inflammatory drug
aMean (SD)
bMean (range)
cObesity defined as body mass index ≥30 kg/m2
dCardiovascular disease defined as ischemic heart disease, heart failure or peripheral vascular disease

Serum urate

In both the control/DE and DE/DE groups the mean baseline SU was significantly higher in those with CrCL <30 ml/min compared to those with CrCL ≥30 to <60 ml/min and ≥60 ml/min (Fig. 1a). Mean SU was below 6 mg/dl in all kidney function groups by month 24 (Fig. 1a). The percentage with SU <6 mg/dl at the final visit by randomization and by kidney function groups is shown in Fig. 1b. Irrespective of randomization, there was no significant difference in the percentage of those with CrCL <30 ml/min who achieved SU <6 mg/dl at the final visit compared to those with CrCL ≥30 to <60 ml/min and ≥60 ml/min, with percentages of 64.3% vs. 76.4% vs. 75.0%, respectively (p = 0.65). The mean (standard error (SE)) change in SU from baseline to month 24 irrespective of randomization, was significantly higher in those with CrCL <30mls/min; mean change -2.23 (0.88) mg/dl in those with CrCL <30mls/min, -1.98 (0.23) mg/dl in those with CrCL ≥30 to <60 ml/min and -1.00 (0.79) mg/dl in those with CrCL ≥60 ml/min (p = 0.002). The mean change in SU by kidney function and randomization group is shown in Table 2.
Table 2
Primary and secondary efficacy endpoints
Variable
C/DE (n = 93)
DE/DE (n = 90)
P valuesa
CrCL <30 ml/min (n = 14)
CrCL ≥30 to <60 ml/min (n = 31)
CrCL ≥60 ml/min (n = 48)
CrCL <30 ml/min (n = 10)
CrCL ≥30 to<60 ml/min (n = 40)
CrCL ≥60 ml/min (n = 40)
 
Change in serum urate (mg/dl), mean (SE)
 Baseline to month 12
-0.67 (0.67)
-0.05 (0.33)
-0.25 (0.26)
-1.49 (0.47)
-2.27 (0.29)
-1.06 (0.24)
0.04
 Baseline to month 24
-2.23 (0.88)
-1.41 (0.36)
-1.10 (0.25)
-2.47 (0.68)
-2.35 (0.28)
-0.86 (0.27)
0.15
 Month 12 to month 24
-1.42 (0.45)
-1.51 (0.41)
-0.87 (0.25)
-1.20 (0.29)
0.01 (0.17)
0.34 (0.23)
0.82
Serum urate <6 mg/dl, %
 Month 12
13%
25%
49%
57%
86%
86%
0.50
 Month 24
57%
68%
72%
71%
82%
79%
0.93
Mean (SE) serum urate
 Baseline
8.25 (0.41)
7.10 (0.29)
6.82 (0.21)
8.02 (0.46)
7.63 (0.26)
6.52 (0.20)
<0.001b
 Month 12
7.38 (0.57)
7.13 (0.32)
6.37 (0.24)
5.91 (0.59)
5.25 (0.12)
5.34 (0.19)
0.20
 Month 24
5.93 (0.71)
5.72 (0.25)
5.62 (0.19)
5.21 (0.30)
5.27 (0.18)
5.61 (0.25)
0.46
Percentage change in serum urate from baseline, mean (SE)
 Baseline to month 12
-6.2 (7.2)
1.6 (4.9)
-1.9 (3.4)
-20.2 (6.0)
-27.5 (2.9)
-15.0 (3.5)
0.08
 Baseline to month 24
-25.5 (8.5)
-16.6 (4.8)
-13.9 (3.2)
-30.0 (5.8)
-29.1 (2.8)
-11.8 (4.1)
0.14
 Month 12 to month 24
-19.4 (5.4)
-17.9 (4.6)
-10.1 (3.5)
-2.4 (5.4)
0.87 (3.2)
7.75 (4.6)
0.99
Percentage with at least one flare in preceding month
 Baseline
50%
38.7%
56.3%
40.0%
32.5%
42.5%
0.17b
 Month 12
37.5%
12.5%
39%
14.3%
34.3%
32.1%
0.10
 Month 24
14.3%
13.6%
20.5%
0.0%
15.2%
3.4%
0.30
Allopurinol dose (mg/day) to achieve target SU at month 24, mean (range)
262.5 (150–500)
389.3 (250–650)
439.3 (300–800)
350.0 (250–600)
396.3 (200–700)
491.3 (300–900)
0.002b
Number of participants requiring >300 mg/day to achieve target SU at month 24
1/4 (25%)
9/14 (64.3%)
20/28 (71.4%)
1/5 (20%)
16/27 (59.3%)
19/23 (82.6%)
0.013
Percentage of individuals receiving anti-inflammatory prophylaxis
 Baseline
35.7%
48.4%
52.1%
40.0%
60.0%
57.5%
0.30b
 Month 12
37.5%
29.2%
29.3%
71.4%
34.3%
14.3%
0.15
 Month 24
14.3%
18.2%
15.4%
57.1%
9.1%
10.3%
0.16
HAQ mean (SE) change
 Baseline to month 12
-0.10 (0.19)
0.10 (0.15)
-0.14 (0.09)
0.47 (0.28)
0.06 (0.12)
-0.13 (0.11)
0.25
 Baseline to month 24
-0.09 (0.21)
0.14 (0.11)
-0.32 (0.07)
-0.19 (0.30)
-0.05 (0.11)
-0.20 (0.12)
0.67
 Month 12 to month 24
0.10 (0.04)
-0.09 (0.18)
-0.22 (0.08)
-0.72 (0.35)
-0.05 (0.13)
-0.05 (0.11)
0.04
Pain VAS mean (SE) change
 Baseline to month 12
-0.25 (0.90)
1.04 (0.51)
-0.02 (0.40)
0.43 (0.61)
0.17 (0.40)
-0.43 (0.54)
0.57
 Baseline to month 24
-1.15 (1.3)
0.27 (0.62)
-1.10 (0.35)
-0.50 (1.36)
-0.64 (0.37)
-1.28 (0.53)
0.56
 Month 12 to month 24
-0.71 (0.71)
-0.52 (0.84)
-1.03 (0.37)
-0.60 (0.87)
-0.67 (0.34)
-0.78 (0.57)
0.93
SJC mean (SE) change
 Baseline to month 12
-2.88 (1.84)
-0.04 (1.05)
-0.07 (0.72)
0.86 (1.6)
-1.17 (0.89)
0.17 (0.53)
0.19
 Baseline to month 24
-2.14 (1.39)
-2.86 (1.65)
-1.26 (0.73)
-0.67 (0.67)
-1.85 (0.93)
-0.24 (0.27)
0.99
 Month 12 to month 24
1.23 (1.16)
-2.91 (2.24)
-1.34 (0.98)
0 (0)
-0.52 (0.42)
-0.37 (0.59)
0.57
TJC mean (SE) change
 Baseline to month 12
2.13 (3.56)
0.04 (1.33)
-2.83 (1.07)
-0.57 (0.62)
-1.57 (0.90)
1.18 (1.42)
0.04
 Baseline to month 24
-2.57 (1.97)
-1.73 (1.49)
-2.08 (0.88)
-0.50 (0.34)
-0.42 (1.54)
-0.97 (0.50)
0.97
 Month 12 to month 24
-5.0 (3.0)
-2.57 (1.83)
0.65 (0.55)
-0.20 (0.20)
1.18 (1.32)
-1.96 (1.37)
0.02
C/DE control/dose escalation phasegroup, DD immediate dose escalation group, CrCL creatinine clearance, SU serum urate, HAQ health assessment questionnaire, VAS visual analogue scale, SJC swollen joint count, TJC tender joint count
aFor levels and changes after baseline the p value tests the significance of randomization depending on baseline CrCL
bFor baseline assessments the p value represents the significance of differences between kidney function groups
The mean percentage change in SU by randomization and by kidney function groups is shown in Fig. 1c. Irrespective of randomization, the mean (SE) percentage change in SU from baseline to final visit in those with CrCL <30 ml/min was similar to those with CrCL ≥30 to <60 ml/min and significantly higher than those with CrCL ≥60 ml/min (-27.7% (5.0%) vs. -24.1% (2.7%) vs. -13.0% (2.5%) (p = 0.003)).

Allopurinol dose

Mean allopurinol dose during the study period by randomization and by kidney function groups is shown in Fig. 1d. Irrespective of randomization, mean (SE) allopurinol dose at baseline was lower in those with lower CrCL; 146 (18) mg/day, 243 (10) mg/day and 323 (9) mg/day (p < 0.001) in those with CrCL <30mls/min, ≥30 to <60 ml/min and ≥60 ml/min, respectively. Irrespective of randomization, the mean (SE) allopurinol dose at month 24 was significantly lower in those with CrCL <30 ml/min as compared to those with CrCL ≥30 to <60 ml/min or CrCL ≥60 ml/min (250 (43) mg/day, 365 (22) mg/day and 460 (19) mg/day, respectively (p < 0.001)). Allopurinol dose in those with SU <6 mg/dl at month 24 is shown in Fig. 2.
The allopurinol dose required to achieve target SU was associated with baseline kidney function (Table 2). Of those with CrCL <30 ml/min (n = 14), only one participant in the control/DE and one in the DE/DE group required >300 mg/day to achieve target SU. In those with higher CrCL, the number of participants requiring >300 mg/day allopurinol to achieve target SU was higher (Table 2). Irrespective of randomization, the mean (range) allopurinol dose required to achieve target SU was higher in those with better kidney function (311.1 mg/day (150–600) vs. 393.9 mg/day (200–700) vs. 462.8 mg/day (300–900)).

Adverse events

There were 17 deaths during the study period, details of these have been published previously [10, 11]. During the RCT phase of the study there were five deaths in the control group of which four occurred in those with CrCL <30mls/min. In comparison none of the five deaths in the dose escalation groups had a CrCL <30 ml/min (Fig. 3). During the open extension phase of the study there were four deaths in the control/DE group, of which one occurred in those with CrCL <30mls/min. In comparison, there were three deaths in the DE/DE group, of which one occurred in those with CrCL <30mls/min. Of note there were high rates of co-morbidities and in particular cardiovascular disease at baseline in those with CrCL <30 ml/min (Table 1). The number of SAEs according to kidney function group and randomization group are shown in Table 3. The type and number of SAEs was as expected and was similar between groups. The percentage of participants with treatment emergent or worsening gamma glutamyl transferase (GGT), alanine transaminase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), creatinine and CrCL by kidney function groups are shown in Fig. 3.
Table 3
Serious adverse events during months 0–12 and months13–24 by kidney function group
CTCAE category
Time period
Control (n = 93)
Dose escalation (n = 90)
CrCL <30 ml/min (n = 14)
CrCL ≥30 to <60 ml/min (n = 31)
CrCL ≥60 ml/min (n = 48)
CrCL <30 ml/min (n = 10)
CrCL ≥30 to <60 ml/min (n = 40)
CrCL ≥60 ml/min (n = 40)
Cardiac disorders
Month 0–12
6 (3)
7 (4)
1 (1)
1 (1)
9 (7)
4 (3)
Month 13–24
3 (2)
8 (5)
0
0
6 (5)
2 (2)
Gastrointestinal disorders
Month 0–12
1 (1)
3 (3)
2 (2)
0
3 (3)
0
Month 13–24
0
5 (4)
1 (1)
0
3 (3)
0
General disorders
Month 0–12
1 (1)
0
0
0
0
1 (1)
Month 13–24
1 (1)
0
1 (1)
0
0
1 (1)
Hepatobiliary disorders
Month 0–12
0
0
0
0
1 (1)
0
Month 13–24
0
0
0
0
0
0
Infections and infestations
Month 0–12
2 (2)
3 (3)
3 (3)
1 (1)
2 (1)
1 (1)
Month 13–24
0
6 (4)
2 (2)
3 (3)
5 (3)
1 (1)
Injury, poisoning and procedural complications
Month 0–12
1 (1)
0
1 (1)
0
1 (1)
0
Month 13–24
1 (1)
3 (2)
1 (1)
0
1 (1)
0
Investigations
Month 0–12
0
0
0
0
1 (1)
0
Month 13–24
0
0
0
0
0
0
Metabolism and nutrition
Month 0–12
0
0
0
1 (1)
1 (1)
0
Month 13–24
0
0
0
0
0
0
Musculoskeletal
Month 0–12
0
0
1 (1)
1 (1)
0
0
Month 13–24
0
0
2 (1)
0
1 (1)
0
Nervous system disorders
Month 0–12
1 (1)
1 (1)
1 (1)
0
0
1 (1)
Month 13–24
3 (2)
0
0
1 (1)
3 (3)
0
Renal and urinary disorders
Month 0–12
3 (3)
2 (2)
0
1 (1)
1 (1)
0
Month 13–24
0
0
0
2 (2)
1 (1)
0
Respiratory, thoracic and mediastinal disorders
Month 0–12
0
1 (1)
1 (1)
1 (1)
1 (1)
0
Month 13–24
0
0
0
0
0
0
Skin and subcutaneous tissue disorders
Month 0–12
1 (1)
0
0
0
0
2 (1)
Month 13–24
0
0
0
0
0
0
Psychiatric disorders
Month 0–12
0
0
0
0
0
0
Month 13–24
1 (1)
0
0
0
0
1 (1)
Vascular disorders
Month 0–12
0
0
0
0
0
0
Month 13–24
0
0
0
0
1 (1)
0
Data reported are number of events (number of individuals)

Discussion

Long-term urate lowering in the setting of CKD is challenging and controversial. Although allopurinol is considered first-line ULT, there are limited data in those with CKD. Herein, we showed that use of allopurinol is safe and effective even in those with CKD.
There are few treatment options for urate lowering in people with stage 4 and 5 CKD and gout. There are limited data on the use of febuxostat in those with CrCL <30 ml/min [13]; uricosuric agents are either contraindicated or not effective in those with CrCL <30mls/min, and while pegloticase needs no dose adjustment in CKD, it is not widely available [14]. Thus it is important that clinicians can safely and effectively use allopurinol, given its widespread availability and low cost.
In the current study, similar proportions of participants in each kidney function group achieved target urate, suggesting the strategy is effective even in those with more severe CKD. The dose required to achieve target SU in those with CrCL <30 ml/min was ≤ 300 mg/day in the majority of participants. Doses >300 mg/day are only infrequently used in clinical practice even in those with normal kidney function, frequently due to physician inertia or concern about dose escalation [2, 15]. The data presented herein suggest that in people with CrCL <30 ml/min, allopurinol doses >300 mg/day are rarely needed to achieve target SU.
The numbers and types of SAEs were similar in both groups but as expected the number of cardiac events was higher in those with CrCL <30 ml/min even during the first 12 months when dose escalation was not undertaken [16]. While a number of abnormalities in kidney function were noted, they were similar between randomized groups over the study period.
There are several limitations to this study. There were only small numbers of participants with CrCL <30 ml/min and the study was not powered to detect the rare AHS. Further adequately powered studies of people with stage 4/5 CKD may be required to clarify the impact on clinical outcomes such as flares, activity limitation and health-related quality of life, and to confirm the safety of this approach. However, studies of sufficient size to detect the rare AHS are unlikely to be undertaken given the large numbers of participants that would be required.

Conclusion

Allopurinol is effective at lowering urate even in those with severe CKD. The dose required to achieve target SU is higher in those with better kidney function. Accepting that there were small numbers of participants with CrCL <30 ml/min, these data indicate that allopurinol dose escalation to target SU is safe in people with CKD.

Acknowledgements

We are grateful to the Health Research Council of New Zealand Independent Data Safety Monitoring Committee for monitoring the study.

Funding

This study was funded by the Health Research Council of New Zealand.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Ethical approval was obtained from the Multi-Regional Ethics Committee, New Zealand. Written informed consent was obtained from each participant.
Not applicable.

Competing interests

Prof. Stamp reports grants from Health Research Council of New Zealand, outside the conduct of the study; grants from Ardea Biosciences, grants from Health Research Council of the submitted work; Dr. Chapman has nothing to disclose; Prof. Barclay has nothing to disclose; Dr Horne reports grants from Health Research Council of New Zealand, during the conduct of the study; Prof. Frampton has nothing to disclose; Mrs. Drake has nothing to disclose; Mr Tan has nothing to disclose; Prof. Dalbeth reports grants from Health Research Council of New Zealand, during the conduct of the study, grants from Health Research Council of New Zealand, grants and personal fees from AstraZeneca, grants and personal fees from Ardea Biosciences, personal fees from Takeda, personal fees from Teijin, personal fees from Menarini, grants from Fonterra, personal fees from Pfizer, personal fees from Crealta and personal fees from Cymabay, outside the submitted work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Khanna D, Fitzgerald J, Khanna P, Sangmee B, Singh M, Neogi T, Pillinger M, Merill J, Lee S, Prakash S, et al. 2012 American College of Rheumatology guidelines for the management of gout. Part 1: Systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricaemia. Arthritis Care Res. 2012;64(10):1431–46.CrossRef Khanna D, Fitzgerald J, Khanna P, Sangmee B, Singh M, Neogi T, Pillinger M, Merill J, Lee S, Prakash S, et al. 2012 American College of Rheumatology guidelines for the management of gout. Part 1: Systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricaemia. Arthritis Care Res. 2012;64(10):1431–46.CrossRef
2.
Zurück zum Zitat Richette P, Doherty M, Pascual E, Barskova V, Becce F, Castaneda-Sanabria J, Coyfish M, Guillo S, Janesen T, Janssens H, et al. 2016 updated EULAR evidence-based recommendations for the management of gout. Ann Rheum Dis. 2017;76(1):29–42.CrossRefPubMed Richette P, Doherty M, Pascual E, Barskova V, Becce F, Castaneda-Sanabria J, Coyfish M, Guillo S, Janesen T, Janssens H, et al. 2016 updated EULAR evidence-based recommendations for the management of gout. Ann Rheum Dis. 2017;76(1):29–42.CrossRefPubMed
3.
Zurück zum Zitat Hande K, Noone R, Stone W. Severe allopurinol toxicity. Description and guidelines for prevention in patients with renal insufficiency. Am J Med. 1984;76:47–56.CrossRefPubMed Hande K, Noone R, Stone W. Severe allopurinol toxicity. Description and guidelines for prevention in patients with renal insufficiency. Am J Med. 1984;76:47–56.CrossRefPubMed
4.
Zurück zum Zitat Stamp L, Day R, Yun J. Allopurinol hypersensitivity: investigating the cause and minimizing the risk. Nat Rev Rheum. 2016;12(4):235–42.CrossRef Stamp L, Day R, Yun J. Allopurinol hypersensitivity: investigating the cause and minimizing the risk. Nat Rev Rheum. 2016;12(4):235–42.CrossRef
5.
Zurück zum Zitat Stamp L, Taylor W, Jones P, Dockerty J, Drake J, Frampton C, Dalbeth N. Starting dose, but not maximum maintenance dose, is a risk factor for allopurinol hypersensitivity syndrome: a proposed safe starting dose of allopurinol. Arthritis Rheum. 2012;64(8):2529–36.CrossRefPubMed Stamp L, Taylor W, Jones P, Dockerty J, Drake J, Frampton C, Dalbeth N. Starting dose, but not maximum maintenance dose, is a risk factor for allopurinol hypersensitivity syndrome: a proposed safe starting dose of allopurinol. Arthritis Rheum. 2012;64(8):2529–36.CrossRefPubMed
6.
Zurück zum Zitat Chung W-H, Chang W-C, Stocker S, Juo C-G, Graham G, Lee M-H, Williams KM, Tian YC, Juan KC, Jan Wu YJ, Yang C-H, et al. Insights into the poor prognosis of allopurinol-induced severe cutaneous adverse reactions: the impact of renal insufficiency, high plasma levels of oxypurinol and granulysin. Ann Rheum Dis. 2015;74(12):2157–64.CrossRefPubMed Chung W-H, Chang W-C, Stocker S, Juo C-G, Graham G, Lee M-H, Williams KM, Tian YC, Juan KC, Jan Wu YJ, Yang C-H, et al. Insights into the poor prognosis of allopurinol-induced severe cutaneous adverse reactions: the impact of renal insufficiency, high plasma levels of oxypurinol and granulysin. Ann Rheum Dis. 2015;74(12):2157–64.CrossRefPubMed
7.
Zurück zum Zitat Becker M, Schumacher HR, Wortmann R, MacDonald P, Eustace D, Palo W, Streit J, Joseph-Ridge N. Febuxostat compared with allopurinol in patients with hyperuricaemia and gout. N Engl J Med. 2005;353:2450–61.CrossRefPubMed Becker M, Schumacher HR, Wortmann R, MacDonald P, Eustace D, Palo W, Streit J, Joseph-Ridge N. Febuxostat compared with allopurinol in patients with hyperuricaemia and gout. N Engl J Med. 2005;353:2450–61.CrossRefPubMed
8.
Zurück zum Zitat Sundy J, Baraf H, Yood R, Edwards N, Gutierrez-Urena S, Treadwell E, Vázquez-Mellado J, White W, Lipsky P, Horowitz Z, et al. Efficacy and tolerability of pegloticase for the treatment of chronic gout in patients refractory to conventional treatment: two randomized controlled trials. JAMA. 2011;306(7):711–20.CrossRefPubMed Sundy J, Baraf H, Yood R, Edwards N, Gutierrez-Urena S, Treadwell E, Vázquez-Mellado J, White W, Lipsky P, Horowitz Z, et al. Efficacy and tolerability of pegloticase for the treatment of chronic gout in patients refractory to conventional treatment: two randomized controlled trials. JAMA. 2011;306(7):711–20.CrossRefPubMed
9.
Zurück zum Zitat Saag K, Fitz-Patrick D, Kopicko J, Fung M, Bhakta N, Adler S, Storgard C, Baumgartner S, Becker M. Lesinurad combined with allopurinol: randomized, double-blind, placebo-controlled study in gout subjects with inadequate response to standard of care allopurinol (a US-based study). Arthritis Rheum. 2017;69(1):203–12.CrossRef Saag K, Fitz-Patrick D, Kopicko J, Fung M, Bhakta N, Adler S, Storgard C, Baumgartner S, Becker M. Lesinurad combined with allopurinol: randomized, double-blind, placebo-controlled study in gout subjects with inadequate response to standard of care allopurinol (a US-based study). Arthritis Rheum. 2017;69(1):203–12.CrossRef
10.
Zurück zum Zitat Stamp L, Chapman P, Barclay M, Horne A, Frampton C, Tan P, Drake J, Dalbeth N. A randomised controlled trial of the efficacy and safety of allopurinol dose escalation to achieve target serum urate in people with gout. Ann Rheum Dis. 2017;76:1522–8.CrossRefPubMed Stamp L, Chapman P, Barclay M, Horne A, Frampton C, Tan P, Drake J, Dalbeth N. A randomised controlled trial of the efficacy and safety of allopurinol dose escalation to achieve target serum urate in people with gout. Ann Rheum Dis. 2017;76:1522–8.CrossRefPubMed
11.
Zurück zum Zitat Stamp L, Chapman P, Barclay M, Horne A, Frampton C, Tan P, Drake J, Dalbeth N. Allopurinol dose escalation to achieve serum urate below 6 mg/dl: an open label extension study. Ann Rheum Dis. 2017;76(12):2065–70.CrossRefPubMed Stamp L, Chapman P, Barclay M, Horne A, Frampton C, Tan P, Drake J, Dalbeth N. Allopurinol dose escalation to achieve serum urate below 6 mg/dl: an open label extension study. Ann Rheum Dis. 2017;76(12):2065–70.CrossRefPubMed
12.
Zurück zum Zitat Wallace S, Robinson H, Masi A, Decker J, McCarty D, Yu T. Preliminary criteria for the classification of the acute arthritis of primary gout. Arthritis Rheum. 1977;20(3):895–900.CrossRefPubMed Wallace S, Robinson H, Masi A, Decker J, McCarty D, Yu T. Preliminary criteria for the classification of the acute arthritis of primary gout. Arthritis Rheum. 1977;20(3):895–900.CrossRefPubMed
13.
Zurück zum Zitat Saag K, Whelton A, Becker M, MacDonald P, Hunt B, Gunawardhana L. Impact of febuxostat on renal function in gout subjects with moderate-to-severe renal impairment. Arthritis Rheum. 2016;68(8):2035–43.CrossRef Saag K, Whelton A, Becker M, MacDonald P, Hunt B, Gunawardhana L. Impact of febuxostat on renal function in gout subjects with moderate-to-severe renal impairment. Arthritis Rheum. 2016;68(8):2035–43.CrossRef
14.
Zurück zum Zitat Vargas-Santos AB, Neogi T. Management of gout and hyperuricaemia in CKD. Am J Kidney Dis. 2017. https://doi.org/10.1053/j.ajkd.2017.01.055. Vargas-Santos AB, Neogi T. Management of gout and hyperuricaemia in CKD. Am J Kidney Dis. 2017. https://​doi.​org/​10.​1053/​j.​ajkd.​2017.​01.​055.​
15.
Zurück zum Zitat Becker M, Fitz-Patrick D, Choi H, Dalbeth N, Storgarde C, Cravetsf M, Baumgartner S. An open-label, 6-month study of allopurinol safety in gout: the LASSO study. Semin Arthritis Rheum. 2015;2015(45):174–83.CrossRef Becker M, Fitz-Patrick D, Choi H, Dalbeth N, Storgarde C, Cravetsf M, Baumgartner S. An open-label, 6-month study of allopurinol safety in gout: the LASSO study. Semin Arthritis Rheum. 2015;2015(45):174–83.CrossRef
16.
Zurück zum Zitat Gupta T, Harikrishnan P, Kolte D, Khera S, Subramanian K, Mujib M, Masud A, Palaniswamy C, Sule S, Jain D, et al. Trends in management and outcomes of ST-elevation myocardial infarction in patients with end-stage renal disease in the United States. Am J Cardiol. 2015;115(8):1033–41.CrossRefPubMed Gupta T, Harikrishnan P, Kolte D, Khera S, Subramanian K, Mujib M, Masud A, Palaniswamy C, Sule S, Jain D, et al. Trends in management and outcomes of ST-elevation myocardial infarction in patients with end-stage renal disease in the United States. Am J Cardiol. 2015;115(8):1033–41.CrossRefPubMed
Metadaten
Titel
The effect of kidney function on the urate lowering effect and safety of increasing allopurinol above doses based on creatinine clearance: a post hoc analysis of a randomized controlled trial
verfasst von
Lisa K. Stamp
Peter T. Chapman
Murray Barclay
Anne Horne
Christopher Frampton
Paul Tan
Jill Drake
Nicola Dalbeth
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2017
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-017-1491-x

Weitere Artikel der Ausgabe 1/2017

Arthritis Research & Therapy 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.