Sie sind auf Seite 1von 14

New Drug Class

Gout therapeutics: new drugs for an old disease


Christopher M Burns, Robert L Wortmann

The approval of febuxostat, a non-purine-analogue inhibitor of xanthine oxidase, by the European Medicines Agency and
the US Food and Drug Administration heralds a new era in the treatment of gout. The use of modied uricases to rapidly
reduce serum urate concentrations in patients with otherwise untreatable gout is progressing. Additionally, advances in
our understanding of the transport of uric acid in the renal proximal tubule and the inammatory response to
monosodium urate crystals are translating into potential new treatments. In this Review, we focus on the clinical trials of
febuxostat. We also review results from studies of pegloticase, a pegylated uricase in development, and we summarise
data for several other pipeline drugs for gout, such as the selective uricosuric drug RDEA594 and various interleukin-1
inhibitors. Finally, we issue a word of caution about the proper use of the new drugs and the already available drugs for
gout. At a time of important advances, we need to recommit ourselves to a rational approach to the treatment of gout.

Introduction
50 years after McCarty and Hollander rediscovered
monosodium urate crystals in gouty joints,1 up to 5 million
people in the European Union and a similar number in
the USA have gout. This disease is the most common form
of inammatory arthritis in men older than 40 years, and
aects 12% of adults in developed countries,25 although
the best way to estimate the incidence and prevalence of
gout is debated.6 The last drug approved by the US Food
and Drug Administration for the treatment of gout was
allopurinol, in 1965. With approval of febuxostat by the
European Medicines Agency in 2008 and by the US Food
and Drug Administration in 2009, and with the appearance
of several novel drugs in the therapeutic pipeline, a new
era in gout treatment is beginning.
In this Review, we discuss febuxostat and several other
drugs still in development that target various steps in the
pathogenesis of gout (gure 1). New urate-lowering drugs
use two traditional strategies: inhibition of xanthine
oxidase to reduce production of uric acid and promotion
of uricosuria to increase its renal excretion. A new
approach uses pegloticase, a polymer-coupled form of
uricase, to rapidly reduce serum urate concentrations.
We focus on the approved drug febuxostat and on
pipeline drugs such as pegloticase, RDEA594, and various
interleukin-1 inhibitors in the treatment of gout (table 1).
For an update on clinical features of gout, please see the
Seminar in The Lancet.7

Approved drug: febuxostat


Febuxostat, 2-[3-cyano-4-(2-methylpropoxy) phenyl]-4methyl-thiazole-5-carboxylic acid (gure 2), is a potent
oral inhibitor of xanthine oxidase, and lowers serum
urate concentration in a predictable way in man.810
Unlike allopurinol and its active metabolite oxypurinol,
febuxostat is not a purine analogue and inhibits only
xanthine oxidase, not other enzymes in the purine and
pyrimidine metabolic pathways.10 With 85% absorption
1 h after oral administration, and a half-life of 418 h,
once-daily dosing is eective.912 Metabolism occurs
mainly in the liver via glucuronide formation and
oxidation, with about 50% of the drug excreted in stool
and 50% appearing in urine, either unchanged or as
www.thelancet.com Vol 377 January 8, 2011

metabolic products.9 Febuxostat dose adjustments


are not needed in patients with mild to moderate
renal failure.13

Lancet 2011; 377: 16577


Published Online
August 17, 2010
DOI:10.1016/S01406736(10)60665-4
See Editorial page 97
Dartmouth Medical School,
Lebanon, NH, USA
(C M Burns MD,
Prof R L Wortmann MD)
Correspondence to:
Dr Christopher M Burns,
Dartmouth Medical School, 1
Medical Center Drive, Lebanon,
NH03756, USA
chris.burns@hitchcock.org

Randomised controlled trials


Febuxostat has been studied in one phase 2 and three
phase 3 randomised controlled trials.1417 Table 2 summarises
these phase 3 trials. In FACT, 762 patients with serum
urate concentrations of 476 mol/L or higher were
randomly assigned to receive febuxostat 80 mg or 120 mg
daily, or allopurinol 300 mg daily, for 52 weeks.15 Febuxostat
was better at achieving the primary endpoint of a serum
urate concentration lower than 357 mol/L at each of the
last three monthly measurements. In APEX, 1072 gout
patients were given febuxostat 80 mg, 120 mg, or 240 mg
daily; allopurinol 300 mg or 100 mg daily according to renal
function; or placebo for 26 weeks.16 All doses of febuxostat
were better than allopurinol and placebo, including in the
subset of patients with renal insuciency (serum
creatinine concentration of 132176 mol/L). In
CONFIRMS, 2269 patients were randomly assigned to
Search strategy and selection criteria
We searched the PubMed (Medline), EmBase, and Cochrane
library databases for articles published in English since 1990
with the search terms gout, and hyperuricemia. We also
searched for gout or hyperuricemia combined with
treatment. We searched the reference lists of publications
identied through the initial search for further citations.
Additionally, we searched the American College of
Rheumatology (ACR) and the European League Against
Rheumatism (EULAR) websites to access abstracts presented
at recent meetings so as to identify the latest information
available, with the understanding that these reports have not
all undergone peer review. Finally, we searched the websites
of the US Food and Drug Administration and European
Medicines Agency for more detailed information about the
application processes for the relevant drugs. We generally
focused on studies published in the past 5 years, but also
included often cited, older publications. Finally, select review
articles and book chapters are also cited for more thorough
coverage of topics beyond the scope of this New Drug review.

165

New Drug Class

Inhibitory eect
Stimulatory eect

Purine nucleotide
metabolism

Xanthine

Xanthine oxidase inhibitors:


allopurinol, oxypurinol,
febuxostat

Uric acid

Allantoin
Uricases:
rasburicase,
pegloticase

Uricosurics:
probenecid,
benzbromarone,
RDEA594

Renal
excretion

Gastrointestinal tract
excretion

Circulating
in excess

Urate
deposition

Uricosuria

Inammation inhibitors:
Non-steroidal anti-inammatory drugs,
colchicine, steroids, interleukin-1 inhibitors

Tophi

Inammasome activation
with acute gout attack

Figure 1: Targets for intervention in the treatment and prophylaxis of gout

Description

Dosing

Status

Non-purine xanthine
oxidase inhibitor

Oral, once daily

FDA, EMA approved for gout

Pegloticase

Pegylated recombinant
porcine-like uricase

Intravenously, every
2 or 4 weeks

Phase 3 for gout completed;


resubmission to FDA in early 2010

RDEA594

Selective URAT1 inhibitor,


uricosuric

Oral, once daily

Phase 2b underway

Anakinra

Recombinant, nonglycosylated form of


human IL-1R

Subcutaneously, daily
for 3 days for acute
gout

FDA approved for rheumatoid


arthritis, not gout

Rilonacept

Soluble receptorfragment-crystallisable
fusion protein; inhibits
interleukins 1 and 1

Subcutaneously, every
week for acute gout or
prophylaxis for
urate-lowering

FDA approved for


cryopyrin-associated periodic
syndromes, not gout; phase 2

Approved drug
Febuxostat
Pipeline drugs

IL-1 Inhibitors

Canakinumab

One subcutaneous
Fully human antiinterleukin-1 monoclonal dose for acute gout
antibody

FDA approved for cryopyrinassociated periodic syndromes,


not gout; phase 2

EMA=European Medicines Agency. FDA=US Food and Drug Administration.

Table 1: Description of new drugs for the treatment of gout

febuxostat 40 mg or 80 mg daily; or allopurinol 300 mg


daily, or 200 mg daily in patients with creatinine clearance
of 3059 mL/min.17 Febuxostat 80 mg was better than
40 mg and than allopurinol in achieving a concentration of
serum urate lower than 357 mol/L at nal visit, including
in patients with renal insuciency. Febuxostat 40 mg daily
was better than allopurinol 200 mg daily only in patients
with renal insuciency. The only signicant reduction in
tophi was recorded in patients given febuxostat 120 mg
daily compared with placebo in APEX.
In FACT, the febuxostat groups had more gout ares
than the allopurinol group in the rst 8 weeks during
166

which patients received prophylaxis (naproxen or


colchicine). Thereafter, ares decreased and did not dier
signicantly. Similar results were reported in APEX. In
CONFIRMS, prophylaxis continued for the entire
26-week study. Flares were reduced in general, but were
still more frequent with febuxostat than with allopurinol.
Other than ares, frequencies of serious and treatmentrelated adverse events were similar among groups, with
signicant dierences only noted in febuxostat 120 mg
daily versus placebo in APEX. The most frequent adverse
events related to treatment were liver function test
abnormalities, diarrhoea, headache, dizziness, and
musculoskeletal symptoms. Discontinuation rates were
highest in the febuxostat groups in all three trials. Nine
deaths occurred during the trials, none deemed related
to treatment.
In APEX, cardiovascular events, dened as chest pain,
coronary artery disease, myocardial infarction, and atrial
brillation, occurred more frequently in the febuxostat
80 mg and 120 mg groups than in the other groups (ve
in each of febuxostat groups versus one in each of the
other three groups, including febuxostat 240 mg).
Although these dierences were not signicant, the US
Food and Drug Administration required CONFIRMS to
be completed before approval. In CONFIRMS, all
cardiovascular adverse events were studied by a masked
cardiovascular safety committee using the Anti-Platelet
Trialists Collaboration (APTC) endpoints.18 One death
occurred in each of the febuxostat groups (40 mg and
80 mg), and three deaths occurred in the allopurinol
groups. Three cardiovascular events dened by APTC
occurred in patients given febuxostat 80 mg and three in
the allopurinol groups. There were no statistically
signicant dierences among groups. Findings were
similar for the 26 non-APTC cardiovascular events.17

Open label extension trials


In FOCUS, an open label extension trial of a 28-day
phase 2 randomised controlled trial, consenting patients
completing the randomised trial were given febuxostat
80 mg daily, which could subsequently be adjusted to
40 mg or 120 mg daily.19 Patients with mild renal
insuciency were included. Patients with appropriate
control of serum urate were maintained in study. At
5 years, 93% (54/58) of completing patients had
concentrations of serum urate lower than 357 mol/L.
Half of the enrolled patients discontinued prematurely,
38 in the rst year, but only 13 of them discontinued
because of an adverse event. No deaths were reported.
Gout ares were essentially eliminated and tophus
resolution occurred in 69% of 26 aected patients.
In EXCEL, an open label extension of two phase 3
randomised controlled trials, 1086 patients were
assigned to febuxostat 80 mg or 120 mg daily, or
allopurinol 300 mg daily.20 The goal was to maintain
patients at serum urate concentrations lower than
357 mol/L for up to 3 years. Adjustments were allowed
www.thelancet.com Vol 377 January 8, 2011

New Drug Class

during the rst 6 months to achieve serum urate


concentrations lower than 357 mol/L. At the rst
month, 80% of patients given febuxostat, but only 46%
of those given allopurinol, achieved target concentrations
of serum urate. Failures were reassigned from
allopurinol to febuxostat 80 mg daily, and from
febuxostat 80 mg to 120 mg daily. More than 80% of
patients maintained serum urate concentrations lower
than 357 mol/L thereafter, with disappearance of gout
ares. Tophi resolved in 46% of patients on febuxostat
80 mg, 36% of patients on febuxostat 120 mg, and 29%
of patients on allopurinol. Adverse events did not dier
among groups. Importantly, allopurinol dosing was
xed, inherently favouring the febuxostat groups, for
which dose adjustments were allowed.

Recommendations
The US Food and Drug Administration approved
febuxostat for gout at 40 mg and 80 mg daily in February,
2009. The European Medicines Agency approved
febuxostat at 80 mg and 120 mg daily in May, 2008, before
the CONFIRMS results.17 Cardiovascular events in FACT
and APEX were numerically higher with febuxostat than
with other treatments. In the open label extension
studies, cardiovascular events were more common in
febuxostat groups (27%) than in allopurinol groups
(112%), but when total drug exposures were taken into
account, the incidence was the same.21 Risk factors for
APTC events were a history of atherosclerotic disease or
myocardial infarction, baseline congestive heart failure,
and age older than 60 years at baseline (p=0001 for all
four factors). No association was noted for hypertension,
stroke, diabetes, or hyperlipidaemia. In Europe, the
European Medicines Agency concluded that treatment
with febuxostat of patients with ischaemic heart disease
or congestive heart failure was not recommended. The
US Food and Drug Administration accepted the results
of CONFIRMS as mitigating cardiovascular risk, but
only approved dosing of 40 mg and 80 mg daily. Takeda
has committed to a post-marketing study, designed along
with the US Food and Drug Administration.
In the USA, the recommended starting dose of
febuxostat is 40 mg daily. No dose adjustment is needed
in patients with creatinine clearance higher than
30 mL/min. Although small numbers suggest that the
use of febuxostat in patients with severe renal
insuciency is safe, no strong evidence exists to
recommend it.13 Febuxostat is safe in patients with mild
to moderate hepatic impairment, but cannot be assumed
to be risk-free in those with severe liver disease.22 Liver
function should be monitored since abnormalities are
common. A possible signal for thyroid abnormalities
induced by febuxostat has been recorded21 although the
mechanism is not obvious. In Europe, an increased
thyroid-stimulating hormone concentration is regarded
as a potential adverse event, whereas in the USA, it is
mentioned only as a possible laboratory abnormality.
www.thelancet.com Vol 377 January 8, 2011

Just as with allopurinol, febuxostat should not be used


with drugs metabolised by xanthine oxidase such as
theophylline, 6-mercaptopurine, and azathioprine. Toxic
concentrations with potentially fatal results such as bone
marrow failure are possible. In healthy people, no
signicant interactions with colchicine, naproxen,
indomethacin, hydrochlorothiazide, warfarin, or desipramine have been identied.2327
Concentrations of serum urate should be measured
24 weeks after starting febuxostat treatment. If values of
serum urate are 357 mol/L or higher, the dose should
be increased. The principle of dosing to target should be
followed whatever the urate-lowering drug used, with the
exception of pegloticase. The more rapid and substantial
the fall in serum urate concentration is, the greater is the
likelihood of gout ares. In patients treated with
febuxostat, ares happen earlier during treatment, but
also stop occurring sooner than with allopurinol. This
phenomenon re-emphasises the importance of
prophylaxis and education of patients. Prophylaxis should
be continued for at least 6 months irrespective of the
urate-lowering agent.

Appropriate candidates for febuxostat


Febuxostat and allopurinol have similar safety proles,
although no cases of hypersensitivity syndrome have
been reported with febuxostat. Patients with
Xanthine

Febuxostat
O

O
H

H
O

N
N

H
Allopurinol
O

N
N

O
N

H
Oxypurinol
O

H
N
N
O

N
H

Figure 2: Chemical structures of xanthine, allopurinol and its metabolite


oxypurinol, and febuxostat

167

New Drug Class

FACT (n=762), 52 weeks15

APEX (n=1072), 28 weeks16

CONFIRMS (n=2269), 26 weeks17

Treatment (n)

Febuxostat 80 mg (256)
Febuxostat 120 mg (251)
Allopurinol 300 mg (253)

Febuxostat 80 mg (267)
Febuxostat 120 mg (269)
Febuxostat 240 mg (134)
Allopurinol 100300 mg (268)
Placebo (134)

Febuxostat 40 mg (757)
Febuxostat 80 mg (756)
Allopurinol 200/300 mg (755)
300 mg (611); 200 mg (145 with creatinine clearance
of 3059 mL/min)

Primary Endpoint

Serum urate <357 mol/L last three monthly visits

Serum urate <357 mol/L last three monthly visits

Serum urate <357 mol/L at last visit

Primary endpoint
achieved

Febuxostat 80 mg: 53% (p<0001 vs allopurinol)


Febuxostat 120 mg: 62% (p<0001 vs allopurinol)
Allopurinol 300 mg: 21%

Febuxostat 80 mg: 48% (p<0001 vs allopurinol, placebo)


Febuxostat 120 mg: 65% (p<0001 vs allopurinol, placebo)
Febuxostat 240 mg: 69% (p<0001 vs allopurinol, placebo)
Allopurinol 100/300 mg: 22% (p<0001 vs placebo)
Placebo: 0%

Febuxostat 40 mg: 45%


Febuxostat 80 mg: 67% (p<0001 vs febuxostat
40 mg, allopurinol)
Allopurinol 200/300 mg: 42%

Primary endpoint
achieved in renal
insuciency

NA

Subset: creatinine clearance >15, 20 (n=40)


Febuxostat 80 mg: 4/9=44% (p<005 vs allopurinol)
Febuxostat 120 mg: 5/11=45% (p<005 vs allopurinol)
Febuxostat 240 mg: 3/5=60% (p<005 vs allopurinol)
Allopurinol 100 mg: 0/10=0%
Placebo: 0/5=0%

Subset: creatinine clearance 3089 ml/min (n=1483)


Febuxostat 40 mg: 50% (p=0021 vs allopurinol)
Febuxostat 80 mg: 72% (p<0001 vs febuxostat
40 mg, allopurinol)
Allopurinol 200/300 mg: 42%

First 8 weeks

Febuxostat 80 mg: 22%,


Febuxostat 120 mg: 36% (p<0001 vs others)
Allopurinol: 300 mg: 21%

Febuxostat 80 mg: 28%


Febuxostat 120 mg: 36% (p005 vs febuxostat 80 mg,
allopurinol, placebo)
Febuxostat 240 mg: 46% (p005 vs febuxostat 80 mg,
allopurinol, placebo)
Allopurinol 100/300 mg: 23%
Placebo: 20%

Weeks 952

Febuxostat 80 mg: 64%,


Febuxostat 120 mg: 70%,
Allopurinol 300 mg: 64%

Entire study

No signicant dierences among groups

Febuxostat 40 mg: 31%


Febuxostat 80 mg: 31%
Allopurinol 200/300 mg: 25%
Febuxostat 40 & 80 mg signicantly more than
allopurinol, but p value not reported

Secondary endpoint: All groups with decrease in median size and


tophi reduction
number, but no signicant dierences

No signicant dierences, apart from mean % decrease in


number of tophi in febuxostat 120 mg vs placebo (p005)

NA

Discontinued
treatment (%)

Febuxostat 80 mg: 34% (p=004 vs allopurinol)


Febuxostat 120 mg: 39% (p=0002 vs allopurinol)
Allopurinol: 26%

Febuxostat 80 mg: 35% (p005 vs febuxostat 120 mg,


allopurinol, placebo)
Febuxostat 120 mg: 26%
Febuxostat 240 mg: 36% (p005 vs febuxostat 120 mg,
allopurinol)
Allopurinol 21%
Placebo 25%

Febuxostat 40 mg: 17% (p=0.021 vs allopurinol)


Febuxostat 80 mg: 21% (p<0.001 vs febuxostat
40 mg, allopurinol)
Allopurinol 200/300 mg: 18%

Deaths (n)

Febuxostat 80 mg: 2; febuxostat 120 mg: 2;


allopurinol: 0

None

Febuxostat 40 mg: 1; febuxostat 80 mg: 1;


allopurinol: 3

Adverse events or
serious adverse
events, other than
gout ares

Liver function test abnormalities, diarrhoea,


headaches, and musculoskeletal symptoms most
common in all groups with no signicant
dierences

Febuxostat 80 mg and 120 mg more cardiovascular events


(5 in each, vs 1 in each of other 3 groups), but no signicant
dierences.
Diarrhoea and dizziness more common in febuxostat
240 mg vs other groups except placebo

No signicant dierences
APTC Events: febuxostat 40 mg: 0; febuxostat 80 mg:
3 (1 non-fatal MI, 2 non-fatal strokes); allopurinol:
3 (2 deaths, 1 non-fatal MI); no signicant dierences

Secondary endpoint:
gout ares (%)

APTC= Anti-Platelet Trialists Collaboration. MI=myocardial infarction. NA=not applicable/not done.

Table 2: Summary of three phase 3 randomised controlled trials of febuxostat

hypersensitivity reactions to allopurinol who were given


febuxostat were able to tolerate the new drug.28
Allopurinol doses used in these trials were xed and
too low; this fact cannot be emphasised enough. One
could reasonably argue that if allopurinol doses were
titrated to serum urate concentrations, the febuxostat
advantage might vanish, although the side-eect prole
of allopurinol might also change. Accordingly,
febuxostat should be considered mainly for patients
168

intolerant to allopurinol, for those whose gout is not


controlled with other urate-lowering treatments, and
for those with renal insuciency (but whose creatinine
clearance is higher than 30 mL/min). Febuxostat should
be tried before an attempt at allopurinol desensitisation,
a cumbersome and often unsuccessful strategy for
overcoming milder allopurinol reactions. Finally,
febuxostat should be used before uricosuric drugs in
patients with nephrolithiasis.
www.thelancet.com Vol 377 January 8, 2011

New Drug Class

Pipeline drug: pegloticase, the mammalian


uricasereversing evolution?
Uricase was lost to man and some non-human primates
via a missense mutation in the gene encoding the
enzyme.29 In other species, uricase converts urate to
allantoin, which is ve to ten times more water-soluble
and more readily eliminated than urate. Non-recombinant
uricase from Aspergillus avus was developed and used
for human tumour lysis syndrome in the late 1960s.30,31
This syndrome occurs after chemotherapy of rapidly
proliferating neoplasms, with profound hyperuricaemia
and possible renal failure.32 The non-recombinant
enzyme was eective but its production was dicult, the
half-life was short, and severe allergic reactions were
common (5%).31
Recombinant enzyme produced by genetically
modied Saccharomyces cerevisiae expressing uricase
cloned from A avus was developed in the 1990s.33 In
2002, rasburicase, a recombinant A avus uricase, was
approved by the US Food and Drug Administration for
acute tumour lysis syndrome in children with cancer.34
This agent dramatically lowers serum urate
concentrations, but enthusiasm for its use in gout was
dampened by persistent immunogenicity and short
half-life (less than 24 h).3538 In a promising study,
patients with gout that could not be treated with
allopurinol were given rasburicase; ve patients
received six monthly infusions and ve patients received
ve daily infusions of 02 mg/kg rasburicase intravenously, both after pretreatment with 60 mg of
methylprednisolone intravenously.37 Patients given the
monthly infusions had signicantly lower serum urate
concentrations after 6 months, and two of ve had
reduction in size of tophi. Patients given daily infusions
did not maintain lower serum urate concentrations and
had no change in tophi size. Unfortunately, eight of ten
patients had at least one adverse event, including two
who had hypersensitivity reactions, and eight of ten had
gout attacks despite prophylaxis. No long-term followup was reported. Non-oncological use of rasburicase is
not common.
Strategies devised to overcome these problems were
the use of mammalian uricase and the pegylation of the
uricase by covalent attachment to poly(ethylene glycol).39
Pegloticase (gure 3), a pegylated mammalian (porcinelike) recombinant uricase, has been studied in gout.39
Phase 1 studies established that intravenous administration of pegloticase was better than subcutaneous
administration.40,41 With intravenous administration of
05 mg to 12 mg, the plasma uricase activity increases
linearly with doses up to 8 mg, referring specically to
the mass of the uricase protein within the molecule. This
activitys half-life is 64138 days. With intravenous
doses equal or higher than 4 mg, the concentration of
serum urate falls dramatically in 2472 h, from a mean of
660 mol/L (SD 36) to 59 mol/L (SD 30), staying low for
21 days after infusion.40
www.thelancet.com Vol 377 January 8, 2011

Clinical trials
Pegloticase clinical trials are summarised in table 3. A
phase 2 open label trial randomly assigned 41 patients
whose treatment for gout had failed to 1214 weeks of
intravenous pegloticase at 412 mg every 2 or 4 weeks.42
The mean concentration of plasma urate fell to less than
357 mol/L within 6 h for all doses. The primary endpoint
of plasma urate concentrations lower than 357 mol/L
for 80% of the study duration was achieved in 5088% of
patients. The optimum dose was 8 mg given every
2 weeks. Several patients had striking reductions in tophi
after just 12 weeks.43 Most patients had gout ares (88%).
Development of antibodies to pegloticase in 31 of
41 patients led to a reduced drug half-life. Because of
reactions, infusion concentration and rate were decreased
later in the study. Infusion reaction prophylaxis was
allowed only in patients with previous reactions.
Phase 3 data from replicate 6-month randomised
controlled trials (212 patients total) were presented at the
American College of Rheumatology annual meeting in
2008 (ACR 2008) and at the European League Against
Rheumatism annual meeting in 2009 (EULAR 2009).4446
Patients were randomly assigned to either pegloticase
8 mg intravenously every 2 weeks or 4 weeks or placebo.44
The primary endpoint was plasma urate concentration
lower than 357 mol/L for 80% of the time in months 3
and 6. Colchicine or non-steroidal anti-inammatory
drugs were used for gout prophylaxis. Infusion reaction
prophylaxis consisted of oral fexofenadine and
paracetamol, and hydrocortisone 200 mg intravenously
before infusion. Pegloticase was signicantly more
eective than placebo.44 Secondary endpoints of tophus
reduction, quality of life, and disability measures favoured
pegloticase over placebo in some, but not all,
comparisons.45,46 Complete tophus response, dened as
resolution of one or more tophi without increase in size
in other tophi or development of new ones, was better
than placebo only in the group of 8 mg every 2 weeks,
probably because of measurement technique. It seemed
clear that, when tolerated, pegloticase was ecacious at
reducing tophi.45
Gout ares, infusion reactions, and serious adverse
events were signicantly more frequent in patients given
pegloticase (table 3) than in other patients. The most
common reason for withdrawal was infusion reaction.
Important relations were noted between immunogenicity,
infusion reactions, and ecacy.47 High-titre antibodies to
pegloticase (higher than 1:7290) were associated with
loss of response and infusion reactions. Antibodies to
poly(ethylene glycol) were even more predictive. All
patients with detectable antibodies to poly(ethylene
glycol) also had antibodies to pegloticase, but not vice
versa, probably indicating dierent sensitivities for each
ELISA. Importantly, 96% of patients with antibodies to
poly(ethylene glycol) from the groups assigned every-2week or every-4-week pegloticase were non-responders,
and 50% and 76%, respectively, had infusion reactions.
169

New Drug Class

Figure 3: Molecular models of uricase tetramer (AC) and of pegloticase containing 36 strands of 10-kDa poly(ethylene glycol) (PEG) per uricase tetramer (D)
(A) Cartoon model, (B) space-lling model showing tunnel, and (C) space-lling model rotated around the vertical axis so that the tunnel is not visible, of uricase
tetramer with subunits in dierent colours, based on crystal structure of A avus uricase tetramer. (D) Space-lling model of uricase tetramer, in the same orientation
as in (B), with nine strands of 10 kDa poly(ethylene glycol) attached to each uricase subunit. The scale of (D) is about half that of (AC). Reprinted from reference 39
with permission of authors and publisher (Elsevier).

These antibodies did not neutralise uricase activity in


vitro. Low-titre antibodies to pegloticase were not
predictive. At ACR 2009, further analysis showed that
development of antipegloticase antibodies was associated
with a loss of response in patients, with their plasma
urate concentrations rising above 357 mol/L.48
Furthermore, 71% of infusion reactions occurred after
this loss of response. In the group assigned pegloticase
every 2 weeks, cessation of treatment when concentrations
of plasma urate were higher than 357 mol/L would
have avoided 91% of infusion reactions. This nding
suggests that careful monitoring of concentrations of
serum urate could allow clinicians to avoid most
infusion reactions.
All patients in the pegloticase randomised trials had
some cardiovascular risk factor.44 The cardiovascular
170

adjudication committee of Savient Pharmaceuticals, Inc


identied ten of 169 patients (6%) with major
cardiovascular events in the pegloticase groups and none
of 43 patients in the placebo group. Results from the US
Food and Drug Administrations independent analysis
of these data showed eight events in the pegloticase
groups (5%) and one in the placebo groups (2%).49 The
agency noted that all events occurred in patients with
pre-existing risk factors and concluded that the number
of events was too small to lend support to a denite
cardiovascular signal.49
157 of 212 patients completed the randomised trials;
151 of them enrolled in an open label extension study.
6-month data were presented at ACR 2009.50 82 patients
received 8 mg of pegloticase every 2 weeks and 67 every 4
weeks. Two patients were merely observed. Gout attacks
www.thelancet.com Vol 377 January 8, 2011

New Drug Class

Phase 2 (n=41), 1214 weeks42

Phase 3 GOUT1/GOUT2 (n=212 total), 6 months each4447,50

Treatment (n)

4 mg every 2 weeks (7)


8 mg every 2 weeks (8)
8 mg every 4 weeks (13)
12 mg every 4 weeks (13)

8 mg every 2 weeks (85)


8 mg every 4 weeks (84)
Placebo (43)

Withdrawals

15, all for adverse events, 12 for infusion day events

8 mg every 2 weeks: 26 (31%), due to adverse events 18 (21%)


8 mg every 4 weeks: 25 (30%), due to adverse events 18 (21%)
Placebo: 4 (9%), due to adverse events 1 (2%)

Primary endpoint:
(Percentage of
patients with plasma
urate 357 mol/L
80% of study)

Intention-to-treat:
4 mg every 2 weeks: 4/7 (57%)
8 mg every 2 weeks: 7/8 (88%)
8 mg every 4 weeks: 7/13 (54%)
12 mg every 4 weeks: 8/13 (62%)
(no signicant dierences among groups)

GOUT1 & GOUT2 (n=212):


8 mg every 2 weeks: 36/85 (42%) (p<0001 vs placebo)
8 mg every 4 weeks: 29/84 (35%) (p<0001 vs placebo)
Placebo: 0% 0/43 (0%)

Resolution of
1 tophus

NA
Anecdotal reports of remarkable improvement over
12 weeks43

8 mg every 2 weeks: 21/52 (p=0002 vs placebo)


8 mg every 4 weeks: 11/52
Placebo: 2/29

Gout ares

4 mg every 2 weeks: 86%


8 mg every 2 weeks: 63%
8 mg every 4 weeks: 92%
12 mg every 4 weeks: 100%

Months 13:
8 mg every 2 weeks: 64/85 (77%) (p=0016)
8 mg every 4 weeks: 68/84 (81%) (p=0002)
Placebo: 23/43 (54%)
(p values vs placebo)
Months 46:
8 mg every 2 weeks: 28/69 (41%) (p=0007)
8 mg every 4 weeks: 39/69 (57%)
Placebo: 29/43 (67%)
(p value vs placebo)

Adverse events

166 adverse events, excluding gout ares


93% of patients had an adverse event
40% related to pegloticase treatment
93% mild or moderate severity
Most common: nephrolithiasis 15%, arthralgia 12%, and
infusion day events 127%

Including infusion reactions and gout ares


8 mg every 2 weeks: 80 (94%), led to discontinuation in 16 (19%)
8 mg every 4 weeks: 84 (100%), led to discontinuation in 17 (20%)
Placebo: 41 (95%), led to discontinuation in 1 (2%)
Nausea, headache, and nasopharyngitis most common after infusion
reaction and gout ares

Infusion day events

18/41 patients (44%)


9/18 withdrawn without rechallenge
3/9 rechallenged withdrew because of another infusion day
event
Commonest infusion day events: muscle spasm, dyspnoea,
hypersensitivity

Infusion reactions
8 mg every 2 weeks: 26%, 4 severe (5%), led to discontinuation in
9 (11%)
8 mg every 4 weeks: 40%, 8 severe (10%), led to discontinuation in
11 (13%)
Placebo: 5%, none severe (0%), led to discontinuation in none

Serious adverse
events

13 in 9 patients, 22% of patients


5 deemed pegloticase-related: anaemia, hypersensitivity,
infected tophus, gout ares (2 patients)
2 serious adverse events led to discontinuation:
hypersensitivity, infected tophus

8 mg every 2 weeks: 20 (24%), led to discontinuation in 9 (11%)


8 mg every 4 weeks: 19 (23%), led to discontinuation in 7 (8%)
Placebo: 5 (12%), led to discontinuation in none
Most common: infusion day events, musculoskeletal including gout
ares, and infection

Deaths

No deaths

8 mg every 2 weeks: 2 (2%); 8 mg every 4 weeks: 1 (1%); placebo: 0 (0%)

Antibody analysis

To pegloticase:
4 mg intravenously every 2 weeks: 86%
8 mg intravenously every 2 weeks: 63%
8 mg intravenously every 4 weeks: 69%
12 mg intravenously every 4 weeks: 85%
(Led to shortened half-life, higher plasma urate levels in
some patients, but no signicant dierences)

If high titre (>1:7290) antipegloticase:


1/68 (1%) responders
51/85 (60%) non-responders (p<0001)
13/25 (52%) every 2 weeks group had infusion reactions
18/27 (67%) every 4 weeks had infusion reactions
If any antibody to poly(ethylene glycol):
27/28 (96%) every 2 weeks non-responders
24/25 (96%) every 4 weeks non-responders
14/28 (50%) every 2 weeks had infusion reactions
19/25 (76%) every 4 weeks had infusion reactions

NA=not applicable/not done.

Table 3: Summary of phase 2 and phase 3 clinical trials of pegloticase

continued to decline in patients given pegloticase. Of


48 patients with serum urate concentrations
consistently lower than 357 mol/L on pegloticase in the
randomised trials, 43 maintained that response in the
extension. An additional 20 patients, beyond 32 patients
in the randomised trials, had complete resolution of at
least one tophus. Infusion reactions were common and
www.thelancet.com Vol 377 January 8, 2011

sometimes serious, frequently leading to discontinuation,


especially in patients who had received placebo in the
randomised trials (9% in placebo to every 2-week
pegloticase open label conversion and 19% in placebo to
every 4-week pegloticase open label conversion). Three
patients died during the open label extension, all deemed
unrelated to treatment.49,50
171

New Drug Class

Basolateral membrane
to circulation

Probenecid
Benzbromarone
RDEA-594

Tubule cell

Uric acid

Uric acid
OAT1
OAT3

URAT1
Organic anions
Monocarboxylates
Na+

Probenecid
Benzbromarone

SLC5A8
SLC5A12
Monocarboxylates

Uric acid
Glucose
Fructose

SLC2A9v1
(GLUT9)

SLC2A9v2
(GLUT9N)

SLC13A3

OAT4

Uric acid
Glucose
Fructose

Dicarboxylates

Uric acid

Na+

Dicarboxylates

Apical membrane
tubule lumen

Figure 4: Present understanding of uric acid reabsorption and eects of uricosuric drugs in the proximal
renal tubule
Filtered uric acid is exchanged for monocarboxylates through URAT1 and dicarboxylates through OAT4 on the
apical side of the tubule cell. SLC2A9v2 (GLUT9N) also transports uric acid into the cell, then SLC2A9v1 (GLUT9)
transports it out of the cell through the basolateral membrane and back into the circulation, along with glucose
and fructose. OAT1 and OAT3 are involved in the movement of uric acid through the basolateral membrane,
although details are unclear. RDEA594 seems to be distinct from traditional uricosuric drugs in that it inhibits only
URAT1 and not the basolateral transporters. The sodium-dependent monocarboxylate transporters SLC5A8 and
SLC5A12 and dicarboxlyate transporter SLC13A3 are also shown. Transporters involved in uric acid secretion into
the tubule are not shown. See text for references. Modied from reference 60 with permission of authors and
publisher ( Oxford University Press).

Licensing status and recommendations


A US Food and Drug Administration arthritis advisory
committee voted pegloticase to be safe and eective on
June 16, 2009. However, the agencys complete response
letter, dated July 31, 2009, denied approval and cited
production concerns due to changes in the
manufacturing process after completion of the
randomised trials. The agency also wanted more
communication with prescribing doctors about safety
and monitoring. In September, Savient announced that
a return to the original manufacturing was in process,
and that the US Food and Drug Administration would
not ask for further trials before resubmission in early
2010 for US approval.
If approved, pegloticase will have a much more
restricted target population than febuxostat does.
Careful selection will be crucial. Terkeltaub38 has
proposed guidelines for use of uricase in gout. Uricases
are appropriate for patients with tophaceous gout with
a large excess of total body urate and persisting gout
attacks, or with damaging arthropathy, who are
intolerant to conventional treatments or for whom they
were unsuccessful. Presumably, many such patients
will respond to febuxostat, but others wont. The notion
of debulking the urate load is a good way to
172

conceptualise the pegloticase strategy, and emphasises


it as adjunctive and best followed by another uratelowering treatment. Indeed, pegloticase can reduce
tophi dramatically and more quickly than
conventional treatment.43,46,50
Can patients continue pegloticase indenitely if no
alternative exists? Great strides have been made in
reduction of immunogenicity, but the issue remains,
and continuous use of pegloticase might be restricted
by antibody development. At least 25% of patients
develop antibodies, with subsequent infusion reactions,
restricted ecacy, or drug withdrawal. The estimated
frequency of anaphylaxis was 51% (14 of 273 patients),
73% in 2-week regimen and 39% in 4-week regimen.49
Gout ares were very frequent and sometimes severe.
Infusion reactions and ares occurred despite
concomitant corticosteroids with infusions, which
might further restrict use in patients with
contraindications to corticosteroids such as diabetes or
glaucoma. Pegloticase should not be given to patients
with glucose-6-phosphate-dehydrogenase deciency
since it can induce haemolysis. Yet the absence of other
options for these dicult-to-manage patients warrants
some degree of acceptance of a less than optimum
performance with novel interventions. The open label
extension data are encouraging.50 Identication of rising
concentrations of serum urate as a sign of antibody
development and impending infusion reaction should
allow for safer administration.48

Pipeline drug:
RDEA594a more selective uricosuric?
In man, most ltered urate is reabsorbed, followed by its
secretion and post-secretory reabsorption in the renal
proximal tubule, with about 10% excretion in urine.51
Most patients with gout have inecient renal excretion
of uric acid as the mechanism of hyperuricaemia.51,52
Understanding of these processes has advanced greatly.
Urateanion exchanger transporter 1 (URAT1) has been
identied as a primary transporter of uric acid from the
tubule lumen into epithelial cells of the proximal tubule
in exchange for monocarboxylates (gure 4).53,54 Organic
anion transporter 4 seems to have a similar role in
exchanging uric acid for dicarboxylates.55 Glucose
transporter 9 (GLUT9, SLC2A9), an electrogenic hexose
transporter, and its splicing variants mediate reabsorption
of uric acid, along with glucose and fructose, at the apical
membrane, through the basolateral membrane, and into
the circulation.5659 Probenecid, sulnpyrazone, and
benzbromarone are traditional uricosuric drugs used in
gout. These drugs are now known to inhibit uric acid
reabsorption by URAT1 and GLUT9.58,60

Pharmocokinetics and pharmacodynamics


Serendipitously, during clinical trials in patients with
HIV infection, the non-nucleoside reverse transcription
inhibitor RDEA806 (Ardea Biosciences, San Diego, CA,
www.thelancet.com Vol 377 January 8, 2011

New Drug Class

USA) was noted to have substantial uricosuric eects


traced to its active metabolite RDEA594, which inhibits
URAT1.61,62 Subsequently, RDEA594 itself was given to
healthy individuals.63 Ascending single doses (38 patients,
28 given drug) of 5 mg to 600 mg, and then ascending
daily doses (64 patients, 48 given drug) of 100 mg to
400 mg for 10 days were given to male volunteers, aged
1845 years. Mean maximum plasma concentration
occurred about 1 h after oral dosing. About 2050% of
RDEA594 was excreted unchanged in the urine within
24 h. Individuals given 100 mg and 200 mg daily doses
had 15% and 30% reductions in serum urate
concentrations, respectively, correlating with increased
uricosuria. Adverse events were mild to moderate, most
often diarrhoea, with no withdrawals or deaths.
Investigators concluded that an immediate-release dose
at breakfast would be used in gout trials.
In vitro, RDEA594 at clinically relevant doses did not
inhibit OAT1 or OAT3, whereas probenecid did.64 In vivo,
administration of parental RDEA806 and RDEA594 to
24 individuals did not alter plasma concentrations or
urinary excretion of tenofovir and emtricitabine, reverse
transcription inhibitors dependent on OAT1 and OAT3
for clearance. Thus, RDEA594 might have more
specicity for URAT1 than available uricosurics, and less
potential for drug interactions. No concerning interactions
exist in terms of pharmacokinetics and urinary excretion
between RDEA594 and allopurinol or oxypurinol in
Cynomolgus monkeys, or between RDEA594 and
febuxostat in Sprague-Dawley rats.65 These data suggest
that co-administration of a xanthine oxidase inhibitor
and RDEA594 could safely accelerate urate reduction in
gout patients.

Clinical trials and recommendations


A phase 2a open study of RDEA594 randomly assigned
21 male patients with gout who had concentrations of
serum urate equal or higher than 476 mol/L and
creatinine clearance equal or higher than 50 mL/min to
be given RDEA594 200 mg daily for 1 week, followed by
400 mg daily for 1 week; allopurinol 300 mg daily for
2 weeks; or placebo.66 Colchicine prophylaxis was used.
With such small numbers, the randomisation yielded
signicant baseline dierences among groups.
Nevertheless, at 2 weeks, six of 11 (55%) patients given
RDEA594 had achieved a serum urate concentration
lower than 357 mol/L versus ve of ve (100%) patients
given allopurinol, and none of patients given placebo.
The lowest serum urate concentration was 462%
(SEM 24) lower than baseline with RDEA, 487 % (57)
lower than baseline with allopurinol, and 24% (24)
lower with placebo. No statistical comparisons were
possible. RDEA594 enhanced urinary uric acid excretion
only to values recorded in healthy people. Adverse
eects were mild, apart from acute coronary syndrome
in one patient given allopurinol. A phase 2b study of
RDEA594 is underway.
www.thelancet.com Vol 377 January 8, 2011

Urate crystals

Cell surface recognition by toll-like receptors 2 and 4,


FC receptors, integrins

Crystal uptake

NALP3 inammasome recognition

Caspase cleavage and activation

Prointerleukin 1

Interleukin 1

Monocyte or
macrophage
Interleukin 1

Acute gout

Figure 5. Central role of the innate immune system and the NALP3
inammasome in acute gout
Monosodium urate crystals are recognised on the surface of monocytes by
innate immune system receptors such as toll-like receptors 2 and 4, fragmentcrystallisable receptors and integrins. The crystals are taken up by the cell and
recognised by the NALP3 (cryopyrin) inammasome. Activation of caspase
follows, with cleavage of the precursor prointerleukin 1 to active interleukin 1.
The proinammatory cytokine interleukin 1 is then secreted from the cell,
along with interleukin 18 and tumour necrosis factor . This signal is amplied
through the recruitment of other cells and the acute gout attack ensues. 6569
FC=fragment crystallisable.

The development of RDEA594 is at too early a stage for


recommendations. However, in addition to RDEA594s
selectivity, investigators point to the safety of its
concomitant use with other urate-lowering treatments.
Combination therapy can more eectively lower
concentrations of serum urate and reduce tophi, and is
worth further investigation.67,68 More selective uricosuric
drugs could improve that treatment. The tradeo is a
likelihood of earlier, more frequent, and more severe
gout ares. Aggressive prophylaxis would be essential.
Finally, patients with nephrolithiasis generally should
not be given uricosurics, including RDEA594.

Pipeline drug: interleukin-1 inhibitors


rounding up the number 1 suspect?
In animals, the NALP3 inammasome and interleukin
1 are crucial players in the gout inammatory pathway.
After cell surface recognition by the innate immune
system, including toll-like receptors 2 and 4,69 the
monosodium urate crystals are phagocytised by
monocytes or macrophages (gure 5). Resultant poorly
characterised intracellular changes lead to NALP3
assembly, caspase-1 activation, cleavage of prointerleukin 1, and secretion of active interleukin 1, a
major driver of gout inammation.70 Indeed, we have
known for more than 20 years that murine macrophages
produce interleukin 1 in response to monosodium urate
crystals.71 This eect is blocked in vitro by colchicine.70
Downstream, interleukin 1 activates other macrophages
173

New Drug Class

Urate crystal
recognition

Inammasome activation phase


Proinammatory mediators:
interleukin 1, interleukin 6, interleukin 18,
tumour necrosis factor , interleukin 10

Amplication phase
Intercritical gout
Quiescent phase

The cycle
of gout

Tissue damage mediators:


matrix metalloproteinases,
arginase, prostaglandins,
toll-like receptor signalling

Resolution phase
Anti-inammatory mediators:
CD68, PPAR-, TGF-,
-prostaglandin D synthase

Figure 6: Elucidation of the molecular details behind the characterisation of the


acute attack of gout, from twinge, to full-blown are, to subsequent resolution
On the basis of more than 20 years of research, along with new studies using the
murine air pouch model of gout, three distinct phases are likely at play. These
phases are an early proinammatory cytokine-driven phase after activation of
the inammasome, an amplication phase marked by tissue damage, and a late
resolution phase, in which anti-inammatory mediators predominate. 6569
Immune recognition of crystals is also dependent on proteins coating the
crystals such as immunoglobulin and apolipoprotein E which are
pro-inammatory and anti-inammatory, respectively. Specic mediators in
each phase have been or potentially could be exploited therapeutically.

through the myeloid dierentiation primary response


gene 88 (MyD88)-dependent interleukin-1 receptor to
produce tumour necrosis factor , interleukin 6, and
neutrophil chemotactants.69,70,72 On the basis of murine
air pouch model studies, three distinct phases might be
at play in acute gout: an early pro-inammatory cytokinedriven phase, an amplication phase marked by tissue
damage, and a late resolution phase in which antiinammatory mediators predominate (gure 6).73

Clinical trials and recommendations


Acute gout treatment traditionally consists of nonsteroidal
anti-inammatory
drugs,
colchicine,
corticotropin, or systemic or intra-articular corticosteroids.
Identication of interleukin 1 as crucial in animal
models prompted a pilot study in man with anakinra, an
interleukin-1 receptor antagonist approved for treatment
of rheumatoid arthritis.74 After demonstration that
anakinra was eective in a murine model, ten patients
with acute gout for whom standard treatment had failed
were given 100 mg of anakinra daily subcutaneously for
3 days. Complete resolution of arthritis was recorded in
nine of ten patients at day 3.
Rilonacept is a soluble interleukin-1 receptor fused to
the fragment-crystallisable (Fc) portion of human
immunoglobulin that inhibits interleukin 1 and
interleukin 1. It was approved by the US Food and Drug
Administration in 2008 for use in children with the
autoinammatory
cryopyrin-associated
periodic
174

syndromes in which interleukin 1 has a pathogenetic


role. In an acute gout trial, ten patients with one or more
inamed joints for 4 weeks or longer were given
rilonacept 320 mg subcutaneously, followed by 160 mg
subcutaneously per week for 5 subsequent weeks, in a
14-week, multicentre, non-randomised cross-over study.75
One patient withdrew because of an injection site
reaction, which is the most common adverse event. No
serious adverse events were reported. Three patients
developed non-neutralising antibodies to rilonacept
without consequence. Although there was no signicant
eect on the number of aected joints, patients selfreported median pain scores on a visual analogue scale
decreased from 50 to 28 after 2 weeks of rilonacept
treatment (p<0049), with sustained improvement at
8 weeks (13; p<0.049). C-reactive protein concentration
fell signicantly. At last treatment, 60% of patients
reported more than 50% improvement in pain
(p=000015) and 50% of patients had more than 75%
improvement (p001).
In a phase 2 double-blind randomised trial, 83 patients
with concentrations of serum urate equal or higher than
446 mol/L, with two or more gout attacks per year, and
starting allopurinol treatment, were randomly assigned
to are prophylaxis with either rilonacept 160 mg subcutaneously per week or placebo.76 Patients with acute
attacks were given non-steroidal anti-inammatory drugs
or prednisone. During the 16-week trial, 39 gout ares
occurred in 42 patients given placebo versus nine in
41 patients given rilonacept (p=00036). Of patients on
placebo, 48% had gout ares, and of those, 26% had more
than one are (p=00209). 22% of patients on rilonacept
had ares and none of them had more than one are
(p=00005). Other adverse events were similar and not
serious; infection was the most common of these events
(26% in placebo and 15% in rilonacept).
Canakinumab is a fully human monoclonal antiinterleukin 1 antibody with a 28-day half-life that
received approval from US Food and Drug Administration
in 2009 for cryopyrin-associated periodic syndromes. In
an 8-week, multicentre, masked, double-dummy study,
147 patients with acute gout, refractory or with
contraindications to non-steroidal anti-inammatory
drugs and colchicine, received one subcutaneous dose of
canakinumab (10 mg, 25 mg, 50 mg, 90 mg, or 150 mg).
57 patients with similar background received one
intramuscular injection of 40 mg triamcinolone acetonide
(half-life of 14 days).77 Canakinumab 150 mg was better
than triamcinolone acetonide at reducing pain (p<005),
and did so more rapidly (p=00006). Recurrent ares
occurred in 37% of patients given canakinumab 150 mg
versus 454% of patients given triamcinolone acetonide
during the 8-week follow up (p=0006). Single cases of
appendicitis and carotid artery stenosis in canakinumab
groups and cerebrovascular disorder in the triamcinolone
acetonide group were deemed unrelated to treatment. No
withdrawals due to adverse events occurred. One could
www.thelancet.com Vol 377 January 8, 2011

New Drug Class

argue that this comparison was unfair because these


drugs have very dierent half-lives, and corticosteroids
can produce rebound ares, but eective one visit-one
treatment is an advantage, and canakinumab outperformed triamcinolone acetonide in general.
Interleukin-1 inhibition is a promising treatment for
patients with gout who cannot tolerate traditional
agents, including corticosteroids. Larger clinical trials
are underway with rilonacept and canakinumab, both
for acute gout and for prophylaxis. Interleukin-1
inhibitor studies without eective treatment in the
comparator groups should be interpreted cautiously.
Furthermore, after the febuxostat and pegloticase trials,
we believe that true placebo groups in are prophylaxis
trials are now probably unacceptable, apart from
patients who are unable to take available prophylactic
drugs. Finally, these biological agents are very expensive,
but their short-term use in specic clinical situations
might be humane and also ultimately cost eective.70,78,79

Final observations
Time will tell how febuxostat or the pipeline drugs t
into gout treatment algorithms. At the risk of sounding
critical, we remind readers that confusion about how to
use long-available drugs is a persisting issue with our
management of gout. Addition of new drugs alone will
not correct these pre-existing misconceptions. The
diagnosis should be substantiated by identication of
intra-articular monosodium urate crystals, guidelines
about the start of urate-lowering treatment should be
followed, prophylaxis against ares early in such
treatment should be given, low enough concentrations
of serum urate should be targeted, and long-term
adherence by patients should be sought.8090 Most
patients with gout are handled by primary care providers
and
fewer
than
3%
by
rheumatologists.80
Rheumatologists seem to be somewhat, but not
impressively, better at achieving target concentrations
of serum urate than primary care physicians (mean
serum urate concentrations 353 versus 413 mol/L,
respectively, p=00004).81
A major concern is the continued use of allopurinol at
300 mg daily, or less in renal insuciency, since these
doses are clearly inadequate in most patients.1517, 19,20,9193
Clinicians often use suboptimum doses of allopurinol in
patients with renal insuciency for fear of precipitating
worsening renal failure or the sometimes fatal
allopurinol hypersensitivity syndrome.94 Nevertheless,
the approach should be to start low and go slow, with
careful monitoring while titrating upward to achieve
target serum urate values.95 Evidence that treatment of
hyperuricaemia in these patients might improve renal
function is encouraging.96,97 Higher doses of allopurinol
in patients with gout and renal insuciency are not
associated with increased risk of hypersensitivity, but are
associated with better achievement of target serum urate
values (86%).98,99 Undertreatment for fear of side-eects
www.thelancet.com Vol 377 January 8, 2011

does disservice to patients since they are on a dose of


medicine that will not achieve the therapeutic goal,
which is to stop attacks and resolve tophi, but merely
slow the rate of progression.
These same principles should apply to dosing of
febuxostat, or other urate-lowering treatments, apart
from drugs like pegloticase, for which the reason for
failure is generally the development of antibodies against
the drugs, not inadequate dosing. The excitement
generated by the appearance of new therapies oers an
ideal opportunity to re-educate our patients and ourselves
about gout so as to improve outcome in this highly
treatable disease.8890
Contributors
Both authors were involved in the conception, literature review, writing,
editing, gure creation, and reference selection for this Review.
Conicts of interest
CMB declares he has no conicts of interest. RLW is a paid consultant
for TAP/Takeda and Savient Pharmaceuticals, Inc.
References
1
McCarty DJ Jr, Hollander JL. Identication of urate crystals in gouty
synovial uid. Ann Intern Med 1961; 54: 45260.
2
Wallace KL, Riedel AA, Joseph-Ridge N, Wortmann R. Increasing
prevalence of gout and hyperuricemia over 10 years among older
adults in a managed care population. J Rheumatol 2004;
31: 158287.
3
Saag KG, Choi H. Epidemiology, risk factors, and lifestyle
modications for gout. Arthritis Res Ther 2006; 8: S2.
4
Roddy E, Zhang W, Doherty M. The changing epidemiology of gout.
Nat Clin Pract Rheumatol 2007; 3: 44349.
5
Annemans L, Spaepen E, Gaskin M, et al. Gout in the UK and
Germany: prevalence, comorbidities and management in general
practice 20002005. Ann Rheum Dis 2008; 67: 96066.
6
Harrold LR, Saag K, Yood RA, et al. Validity of gout diagnoses in
administrative data. Arthritis Rheum 2007; 57: 10308.
7
Richette P, Bardin T. Gout. Lancet 2010; 375: 31828.
8
Okamoto K, Eger BT, Nishino T, Kondo S, Pai EF, Nishino T.
An extremely potent inhibitor of xanthine oxidoreductase. Crystal
structure of the enzyme-inhibitor complex and mechanism of
inhibition. J Biol Chem 2003; 278: 184855.
9
Becker MA, Kisicki J, Khosravan R, et al. Febuxostat (TMX-67),
a novel, nonpurine, selective inhibitor of xanthine oxidase, is safe
and decreases serum urate in healthy volunteers.
Nucleosides Nucleotides Nucleic Acids 2004; 23: 111116.
10 Takano Y, Hase-Aoki K, Horiuchi H, et al. Selectivity of febuxostat,
a novel non-purine inhibitor of xanthine oxidase/xanthine
dehydrogenase. Life Sci 2005; 76: 183547.
11 Komoriya K, Hoshide S, Takeda K, et al. Pharmacokinetics and
pharmacodynamics of febuxostat (TMX-67), a non-purine selective
inhibitor of xanthine oxidase/xanthine dehydrogenase (NPSIXO) in
patients with gout and/or hyperuricemia.
Nucleosides Nucleotides Nucleic Acids 2004; 23: 111922.
12 Khosravan R, Grabowski BA, Wu JT, Joseph-Ridge N, Vernillet L.
Pharmacokinetics, pharmacodynamics and safety of febuxostat,
a non-purine selective inhibitor of xanthine oxidase, in a dose
escalation study in healthy subjects. Clin Pharmacokinet 2006;
45: 82141.
13 Mayer MD, Khosravan R, Vernillet L, Wu JT, Joseph-Ridge N,
Mulford DJ. Pharmacokinetics and pharmacodynamics of
febuxostat, a new selective nonpurine inhibitor of xanthine oxidase,
in subjects with renal impairment. Am J Ther 2005; 12: 2234.
14 Becker MA, Schumacher HR Jr, Wortmann RL, et al. Febuxostat,
a novel nonpurine selective inhibitor of xanthine oxidase: a
twenty-eight-day, multicenter, phase II, randomized, double-blind,
placebo-controlled, dose-response clinical trial examining safety and
ecacy in patients with gout. Arthritis Rheum 2005; 52: 91623.
15 Becker MA, Schumacher HR Jr, Wortmann RL, et al. Febuxostat
compared with allopurinol in patients with hyperuricemia and gout.
N Engl J Med 2005; 353: 245061.

175

New Drug Class

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

176

Schumacher HR, Becker MA, Wortmann RL, et al. Eects of


febuxostat versus allopurinol and placebo in reducing serum urate
in subjects with hyperuricemia and gout: a 28-week, phase III,
randomized, double-blind, parallel group trial. Arthritis Care Res
2008; 59: 154048.
Becker M, Schumacher HR Jr, Espinoza L, et al. A phase 3
randomized, controlled, multicenter, double-blind trial (RCT)
comparing ecacy and safety of daily febuxostat (FEB) and
allopurinol (ALLO) in subjects with gout. Arthritis Rheum 2008;
58: S402930.
Collaborative overview of randomised trials of antiplatelet
therapyI: Prevention of death, myocardial infarction, and stroke
by prolonged antiplatelet therapy in various categories of patients.
BMJ 1994; 308: 81106.
Schumacher HR Jr, Becker MA, Lloyd E, MacDonald PA,
Lademacher C. Febuxostat in the treatment of gout: 5-yr ndings
of the FOCUS ecacy and safety study. Rheumatology 2009;
48: 18894.
Becker MA, Schumacher HR, MacDonald PA, Lloyd L,
Lademacher C. Clinical ecacy and safety of successful longterm
urate lowering with febuxostat or allopurinol in subjects with gout.
J Rheumatol 2009; 36: 127382.
European Medicines Agency. CHMP assessment report for
Adenuric. 2008. http://www.emea.europa.eu/humandocs/PDFs/
EPAR/adenuric/H-777-en6.pdf (accessed Feb 1, 2010).
Khosravan R, Grabowski BA, Mayer MD, Wu JT, Joseph-Ridge N,
Vernillet L. The eect of mild and moderate hepatic impairment on
pharmacokinetics, pharmacodynamics, and safety of febuxostat,
a novel nonpurine selective inhibitor of xanthine oxidase.
J Clin Pharmacol 2006; 46: 88102.
Khosravan R, Mayer MD, Wu JT, et al. Eect of concomitant
administration of febuxostat and colchicine on pharmacokinetics of
febuxostat and colchicine at steady state. Arthritis Rheum 2005;
52: S10203.
Khosravan RP, Erdman KB, Vernillet LPP, et al. Eect of febuxostat
on pharmacokinetics of desipramine, a CYP2D6 substrate, in
healthy subjects. Clin Pharmacol Ther 2005; 77: P43P43.
Grabowski BA, Khosravan R, Wu JT, et al. Eect of
hydrochlorothiazide on pharmacokinetics and pharmacodynamics
of febuxostat. Arthritis Rheum 2005; 52: S10304.
Khosravan R, Wu JT, Joseph-Ridge N, et al. Pharmacokinetic
interactions of concomitant administration of febuxostat and
NSAIDs. J Clin Pharmacol 2006; 46: 85566.
Mukoyoshi M, Nishimura S, Hoshide S, et al. In vitro drugdrug
interaction studies with febuxostat, a novel non-purine selective
inhibitor of xanthine oxidase: plasma protein binding, identication
of metabolic enzymes and cytochrome P450 inhibition. Xenobiotica
2008; 38: 496510.
Becker MA, Schumacher HR, Wortmann RL, et al. Allopurinol
intolerant patients treated with febuxostat for 4 years.
Arthritis Rheum 2006; 54: S646.
Wu XW, Lee CC, Muzny DM, Caskey CT. Urate oxidase: primary
structure and evolutionary implications. Proc Natl Acad Sci USA
1989; 86: 941216.
Laboureur P, Langlois C. Urate-oxydase dAspergillus avus: I.
Obtention, purication, proprits. Bull Soc Chim Biol 1968;
50: 81125.
Pui C-H, Relling MV, Lascombes F, et al. Urate oxidase in
prevention and treatment of hyperuricemia associated with
lymphoid malignancies. Leukemia 1997; 11: 181316.
Davidson MB, Thakkar S, Hix JK, Bhandarkar ND, Wong A,
Schreiber MJ. Pathophysiology, clinical consequences and
treatment of tumor lysis syndrome. Am J Med 2004; 116: 54654.
Leplatois P, Le Douarin B, Loison G. High-level production of a
peroxisomal enzyme: Aspergillus avus uricase accumulates
intracellularly and is active in Saccharomyces cerevisiae. Gene 1992;
122: 13945.
Navolanic PM, Pui C-H, Larson RA, et al. Elitekrasburicase: an
eective means to prevent and treat hyperuricemia associated with
tumor lysis syndrome, a Meeting Report, Dallas, Texas, January
2002. Leukemia 2003 17: 499514.
de Bont JM, Pieters R. Management of hyperuricemia with
rasburicase review. Nucleosides Nucleotides Nucleic Acids 2004;
23: 143140.

36

37

38
39

40

41

42

43

44

45

46

47

48

49

50
51

52
53

54

55

56

57

Richette P, Bardin T. Successful treatment with rasburicase of


a tophaceous gout in a patient allergic to allopurinol.
Nat Clin Pract Rheumatol 2006; 2: 33842.
Richette P, Briere C, Honene-Claviert V, Loeuille D, Bardin T.
Rasburicase for tophaceous gout not treatable with allopurinol:
An exploratory study. J Rheumatol 2007; 34: 1093-98.
Terkeltaub R. Learning how and when to employ uricase as bridge
therapy in refractory gout. J Rheumatol 2007; 34: 195558.
Sherman MR, Saifer MGP, Perez-Ruiz F. PEG-uricase in the
management of treatment-resistant gout and hyperuricemia.
Adv Drug Deliv Rev 2008; 60: 5968.
Sundy JS, Ganson NJ, Kelly SJ, et al. Pharmacokinetics and
pharmacodynamics of intravenous PEGylated recombinant
mammalian urate oxidase in patients with refractory gout.
Arthritis Rheum 2007; 56: 102128.
Ganson NJ, Kelly SJ, Scarlett E, Sundy JS, Hersheld MS. Control of
hyperuricemia in subjects with refractory gout, and induction of
antibody against poly(ethylene glycol) (PEG), in a phase I trial of
subcutaneous PEGylated urate oxidase. Arthritis Res Ther 2006; 8: R12.
Sundy JS, Becker MA, Baraf HSB, et al. Reduction of plasma urate
levels following treatment with multiple doses of pegloticase
(polyethylene glycol-conjugated uricase) in patients with treatmentfailure gout. Arthritis Rheum 2008; 58: 288291.
Baraf HS, Matsumoto AK, Maroli AN, Waltrip RW. Resolution of
gouty tophi after twelve weeks of pegloticase treatment.
Arthritis Rheum 2008; 58: 363234.
Sundy JS, Baraf HS, Becker MA, et al. Ecacy and safety of
intravenous pegloticase (PGL) in treatment failure gout (TFG):
results from GOUT1 and GOUT2. Ann Rheum Dis 2009; 68: 318.
Baraf HS, Becker MA, Edwards NL, et al. Tophus response to
pegloticase (PGL) therapy: pooled results from GOUT1 and
GOUT2, PGL phase 3 randomized, double blind, placebo-controlled
trials. Arthritis Rheum 2008; 58: S176.
Edwards NL, Baraf HS, Becker MA, et al. Improvement in healthrelated quality of life (HRQL) and disability index in treatment
failure gout (TFG) after pegloticase (PGL) therapy: pooled results
from GOUT1 and GOUT2, phase 3, randomized, double blind,
placebo (PBO)-controlled trials. Arthritis Rheum 2008; 58: S178.
Becker MA, Treadwell EL, Baraf HS, et al. Immunoreactivity and
clinical response to pegloticase (PGL): pooled data from GOUT1
and GOUT2, PGL phase 3 randomized, double blind,
placebo-controlled trials. Arthritis Rheum 2008; 58: S880.
Wright D, Sundy JS, Rosario-Jansen T. Routine serum uric acid
(SUA) monitoring predicts antibody-mediated loss of response and
infusion reaction risk during pegloticase therapy. Arthritis Rheum
2009; 60: S413.
US Food and Drug Administration. Brieng Document for the
Arthritis Advisory Committee Meeting http://www.fda.gov/
downloads/AdvisoryCommittees/CommitteesMeetings Materials/
Drugs/ArthritisDrugsAdvisoryCommittee/UCM165714.pdf
(accessed Feb 15, 2010).
Sundy JS, Baraf HS, Gutierrez-Urena SR, et al. Chronic use of
pegloticase: study and ecacy update. Arthritis Rheum 2009; 60: S417.
Wyngaarden JB, Kelley WN. Disposition of uric acid in primary
gout. In: Wyngaarden JB, Kelley WN, eds. Gout and hyperuricemia.
New York: Grune & Stratton, 1976: 149157.
Simkin PA. New standards for uric acid excretion: evidence for an
inducible transporter. Arthritis Care Res 2003; 49: 73536.
Enomoto A, Kimura H, Chairoungdu A, et al. Molecular
identication of a renal urate anion exchanger that regulates blood
urate levels. Nature 2002; 417: 44752.
Endou H, Anzai N. Urate transport across the apical membrane of
renal proximal tubules. Nucleosides Nucleotides Nucleic Acids 2008;
27: 57884.
Hagos Y, Stein D, Ugele B, Burckhardt G, Bahn A. Human renal
organic anion transporter 4 operates as an asymmetric urate
transporter. J Am Soc Nephrol 2007; 18: 43039.
Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, Moley JF,
Moley KH. Identication and characterization of human glucose
transporter-like protein-9 (GLUT9): alternative splicing alters
tracking. J Biol Chem 2004; 279: 1622936.
Vitart V, Rudan I, Hayward C, et al. SLC2A9 is a newly identied
urate transporter inuencing serum urate concentration, urate
excretion and gout. Nat Genet 2008; 40: 43742.

www.thelancet.com Vol 377 January 8, 2011

New Drug Class

58

59
60

61

62

63

64

65

66

67

68

69

70

71

72

73

74
75

76

77

78

Anzai N, Ichida K, Jutabha P, et al. Plasma urate level is directly


regulated by a voltage-driven urate eux transporter URATv1
(SLC2A9) in humans. J Biol Chem 2008; 283: 2683438.
Cauleld MJ, Munroe PB, ONeill D, et al. SLC2A9 is a
high-capacity urate transporter in humans. PLoS Med 2008; 5: e197.
Dalbeth N, Merriman T. Crystal ball gazing: new therapeutic
targets for hyperuricaemia and gout. Rheumatology 2009;
48: 22226.
Yeh L, Hingorani V, Manhard K, et al. Safety and uric acid lowering
eect in humans following multiple doses of RDEA806, a novel
prodrug for the potential treatment of hyperuricemia.
Ann Rheum Dis 2008; 67: 248.
Yeh L, Tamai I, Hamatake R, et al. Mode of action of RDEA594 as a
uric acid lowering agent in humans following multiple doses of its
prodrug, RDEA806. Ann Rheum Dis 2008; 67: 249.
Yeh L-T, Shen Z, Kerr B, et al. Safety, pharmacokinetics, and serum
uric acid lowering eect of RDEA594, a novel uricosuric agent, in
healthy volunteers. Ann Rheum Dis 2009; 68: 320.
Yeh L-T, Shen Z, Kerr B, et al. RDEA594, a potent URAT1 inhibitor
without aecting other important renal transporters OAT1 and
OAT3. Ann Rheum Dis 2009; 68: 320.
Yang X, Dick R, Borges V, et al. Evaluation of drug-drug interaction
potential between RDEA594, allopurinol and febuxostat in
preclinical species. Arthritis Rheum 2009; 60: S41213.
Lasko B, Sheedy B, Hingorani V, et al. RDEA594, a novel uricosuric
agent, signicantly reduced serum urate levels and was well
tolerated in a phase 2a pilot study in hyperuricemic gout patients.
Arthritis Rheum 2009; 60: S41314.
Goldfarb E, Smythe CJ. Eects of allopurinol, a xanthine oxidase
inhibitor, and sulnpyrazone upon the urinary and serum urate
concentrations in eight patients with tophaceous gout.
Arthritis Rheum 2007; 9: 41423.
Perez-Ruiz F, Calabozo M, Pijoan JI, Herrero-Beites AM, Ruibal A.
Eect of urate-lowering therapy on the velocity of size of reduction
of tophi in chronic gout. Arthritis Rheum 2002; 47: 35660.
Liu-Bryan R, Scott P, Sydlaske A, Rose DM, Terkeltaub R. Innate
immunity conferred by Toll-like receptors 2 and 4 and myeloid
dierentiation factor 88 expression is pivotal to monosodium urate
monohydrate crystal-induced inammation. Arthritis Rheum 2005;
52: 293646.
Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J.
Gout-associated uric acid crystal activate the NALP3 inammasome.
Nature 2006; 440: 23741.
Di Giovine FS, Malawista SE, Nuki G, Du GW. Interleukin 1 (IL 1)
as a mediator of crystal arthritis. Stimulation of T cell and synovial
broblast mitogenesis by urate crystal-induced IL-1. J Immunol
1987; 138: 321318.
Chen CJ, Shi Y, Hearn A, et al. MyD88-dependent IL-1 receptor
signaling is essential for gouty inammation stimulated by
monosodium urate crystals. J Clin Invest 2006; 116: 226271.
Mayer CT, Tobias JW, Menetski JP, Schumacher HR Jr, Pessler F.
Dynamics of transcription regulation of urate crystal inammation
in a synovium-like tissue. Arthritis Rheum 2009; 60: S729.
So A, De Smedt T, Revaz S Tschopp J. A pilot study of IL-1
inhibition by anakinra in acute gout. Arthritis Res Ther 2007; 9: R28.
Terkeltaub R, Sundy JS, Schumacher HR, et al. The IL-1 inhibitor
rilonacept in treatment of chronic gouty arthritis: Results of a
placebo-controlled, monosequence crossover, nonrandomized,
single-blind pilot study. Ann Rheum Dis 2009; 68: 161317.
Schumacher HR Jr, Sundy JS, Terkeltaub R, et al. Placebo-controlled
study of rilonacept for gout are prophylaxis during initiation of
urate-lowering therapy. Arthritis Rheum 2009; 60: S410.
So A, De Meulemeester M, Shamim T, et al. Canakinumab
(ACZ885) vs. triamcinolone acetonide for treatment of acute ares
and prevention of recurrent ares in gouty arthritis patients
refractory to or contraindicated to NSAIDs and/or colchicine.
Arthritis Rheum 2009; 60: LB4.
Cronstein BN, Terkeltaub R. The inammatory process of gout and
its treatment. Arthritis Res Ther 2006; 8: S3.

www.thelancet.com Vol 377 January 8, 2011

79
80
81

82

83

84

85
86

87

88

89
90
91

92

93

94

95

96

97

98

99

Terkeltaub R. Novel therapies for treatment of gout and


hyperuricemia. Arthritis Res Ther 2009; 11: 236.
Krishnan E, Lienesch D, Kwoh CK. Gout in Ambulatory Care
Settings in the United States. J Rheumatol 2008; 35: 498501.
Keenan RT, Lehman RA, OBrien WR, Crittenden DB, Lee KH,
Pillinger MH. Gout management in primary care vs. rheumatology:
evidence for suboptimal treatment. Arthritis Rheum 2009; 60: S415.
Zhang W, Doherty M, Pascual E, et al. EULAR evidence based
recommendations for gout. Part I: diagnosis: report of a task force
of the Standing committee for International Clinical Studies
Including Therapeutics (ESCISIT). Ann Rheum Dis 2006;
65: 130111.
Zhang W, Doherty M, Bardin T, et al. EULAR evidence bases
recommendations for gout. Part II: management: report of a task
force of the EULAR Standing Committee for International Clinical
Studies Including Therapeutics (ESCISIT). Ann Rheum Dis 2006;
65: 131224.
Stamp L, Sharples K, Gow P, Raill B. The optimal use of
allopurinol: an audit of allopurinol use in South Auckland.
Aust N Z J Med 2000; 30: 56772.
Schlesinger N, Schumacher HR Jr. Gout: can management be
improved? Curr Opin Rheumatol 2001; 13: 240244.
Sarawate CA, Brewer KK, Yang W, et al. Gout medication treatment
patterns and adherence to standards of care from a managed care
perspective. Mayo Clin Proc 2006; 81: 92534.
Sarawate CA, Patel PA, Schumacher HR Jr, Yang W, Brewer KK,
Bakst AW. Serum urate levels and gout ares: analysis from
managed care data. J Clin Rheum 2006; 12: 6165.
Wortmann RL, Ryan LM. Management of crystal-induced
arthropathies. In: Wortmann RL, Schumacher HR Jr, Becker MA,
Ryan LM, eds. Crystal induced arthropathies: gout, pseudogout and
apatite-associated syndromes. New York: Taylor & Francis, 2006.
Becker MA, Chohan S. We can make gout management more
successful now. Curr Opin Rheumatol 2008; 20: 167172.
Sundy JS. Gout management: lets get it right this time.
Arthritis Care Res 2008; 59: 153537.
Perez-Ruiz F, Alonso-Ruiz A, Calabozo M, Herrero-Beites A,
Garcia-Erauskin G, Ruiz-Lucea E. Ecacy of allopurinol and
benzbromarone for the control of hyperuricemia: a pathogenic
approach to the therapy of chronic primary gout. Ann Rheum Dis
1998; 57: 54549.
Dalbeth N, Kumar S, Stamp L, Gow P. Dose adjustment of
allopurinol according to creatinine clearance does not provide
adequate control of hyperuricemia in patients with gout.
J Rheumatol 2006; 33: 164650.
Perez-Ruiz F, Liot F. Lowering serum uric acid levels: what is the
optimal target for improving clinical outcomes in gout?
Arthritis Care & Research 2007; 57: 132428.
Hande KR, Noone RM, Stone WJ. Severe allopurinol toxicity:
description and guidelines for prevention in patients with renal
insuciency. Am J Med 1984; 76: 4756.
Dalbeth N, Stamp L. Allopurinol dosing in renal impairment:
walking the tightrope between adequate urate lowering and adverse
events. Semin Dial 2007; 20: 39195.
Perez-Ruiz F, Calabozo M, Herrero-Beites AM, Garca-Erauskin G,
Pijoan JI. Improvement in renal function in patients with chronic
gout after proper control of hyperuricemia and gouty bouts.
Nephron 2000; 86: 28791.
Siu YP, Leung KT, Tong MK, Kwan TH. Use of allopurinol in
slowing the progression of renal disease through its ability to lower
serum uric acid. Am J Kidney Dis 2006; 47: 5159.
Silverberg MS, Mallela R, Lesse AJ, Bonner MR, Baer AN, Li C.
Allopurinol hypersensitivity reaction: a case-control study of the
role of renal dosing. Arthritis Rheum 2009; 60: S414.
Stamp L, ODonnell JL, Zhang M, Frampton C, Barclay M,
Chapman PT. Increasing allopurinol dose above the recommended
range is eective and safe in chronic gout, including in those with
renal impairment a pilot study. Arthritis Rheum 2009; 60: S729.

177

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

Das könnte Ihnen auch gefallen