SlideShare une entreprise Scribd logo
1  sur  13
Télécharger pour lire hors ligne
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  271
Division of Academic
Rheumatology,
University of
Nottingham, Clinical
Sciences Building, City
Hospital Nottingham,
Hucknall Road,
Nottingham NG5 1PB,
UK (F.R., M.H., M.D.).
Correspondence to:
M.D.
michael.doherty@
nottingham.ac.uk
Optimizing current treatment of gout
Frances Rees, Michelle Hui and Michael Doherty
Abstract | Gout is the most common inflammatory arthritis worldwide. Although effective treatments exist
to eliminate sodium urate crystals and to ‘cure’ the disease, the management of gout is often suboptimal.
This article reviews available treatments, recommended best practice and barriers to effective care, and
how these barriers might be overcome. To optimize the management of gout, health professionals need to
know not only how to treat acute attacks but also how to up-titrate urate-lowering therapy against a specific
target level of serum uric acid that is below the saturation point for crystal formation. Current perspectives
are changing towards much earlier use of urate-lowering therapy, even at the time of first diagnosis of gout.
Holistic assessment and patient education are essential to address patient-specific risk factors and ensuring
adherence to individualized therapy. Shared decision-making between a fully informed patient and practitioner
greatly increases the likelihood of curing gout.
Rees, F. et al. Nat. Rev. Rheumatol. 10, 271–283 (2014); published online 11 March 2014; doi:10.1038/nrrheum.2014.32
Introduction
Gout is the most common form of inflammatory arthri-
tis, with crude estimates of adult prevalence of 2.5% in
the UK,1
3.9% in the USA,2
and 3.8% in Taiwan.3
The
prevalence and incidence of gout increase with age,
and it is more common in men than women (approxi-
mately 3:1).2
Fortunately, the pathogenesis of gout
is relatively well understood. We know the causative
agent—­monosodium urate crystals—and the multi-
ple risk factors that lead to their formation, primar-
ily through chronic elevation of uric acid levels above
the saturation point at which urate crystals can form
(Figure 1). Urate crystals are deposited preferentially
in and around peripheral joints in the feet, knees, hands
and elbows, especially those affected by osteoarthritis.4
The urate crystals predominantly form in the super­ficial
layers of articular cartilage, in subchondral bone and in
fibrous peri-articular tissues, but might also be evident
clinically as subcutaneous tophi. During a prolonged
asymptomatic phase (the duration of which is unknown
but presumed to be measured in years), the concentra-
tion of crystals gradually builds up and eventu­ally, once
deposition is extensive, it is hypothesized that urate
crystals ‘shed’ from the articular cartilage into the joint
space and trigger an acute ‘attack’ of extremely painful
joint inflammation. This acute attack is usually the
reason patients first seek medical attention. However, if
left untreated, continuing urate crystal deposition not
only causes further acute attacks and involvement of
additional joint sites, but also, and importantly, could
cause eventual irreversible joint damage with chronic
symptoms and disability. Apart from causing acute gout
and chronic joint damage, there is increasing evidence,
though still controversial,5
that a high serum uric acid
(SUA) level is an independent risk factor for cardio­
vascular disease, stroke and chronic kidney disease
(CKD).6–8
Therefore, there are several reasons to con-
sider definitive treatment of elevated SUA levels once a
patient is known to have gout.
Fortunately, several treatment strategies exist to
effectively reduce and maintain SUA levels below the
saturation point, thus preventing further crystal for-
mation and encouraging the dissolution of exist­ing
crystals. Once the pathogenic urate crystals are elimi-
nated, further acute attacks are impossible and the risk
of further crystal-induced joint damage is removed.
How­ever, although gout is the only form of chro­nic
arthritis for which such a ‘cure’ exists, audits pub-
lished in the past 4 years confirm that the treatment of
gout both in primary care and hospital practice is far
from optimal.1,9,10
This article reviews recommended available treat-
ments, and highlights new perspectives relating to
current standards of care and barriers that need to be
addressed if gout is to be managed successfully. The
Review largely reflects the latest EULAR11
and British
Society for Rheumatology (BSR)12
guidelines, published
in 2006 and 2007 respectively (both are currently being
updated), as well as the more recent 2012 ACR guide-
lines.13,14
In addition, we systematically searched the
literature for individual treatments. Some drug options
(including biologic agents and uricases) are not dis-
cussed in detail because they are largely unlicenced for
gout, are very expensive and have limited application.
We first review the available options for managing acute
attacks and for reducing SUA levels, and then focus
on important barriers to care and how to successfully
implement ‘curative’ management strategies.
Competing interests
M.D. has received honoraria for being a member of ad hoc
advisory boards for Ardea Biosciences, Menarini, Novartis and
Savient. F.R. and M.H. declare no competing interests.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
272  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
Management of acute attacks
Acute gout is recognized as one of the most painful
experi­ences known, on the same level as childbirth or
visceral colic. Therefore, treatment of an attack should
be initiated as soon as possible following initial diagno-
sis or, in a patient with known gout, as soon as the first
warning symptoms are perceived. Only a few recom­
mended options are available, and formal research evi-
dence for most of these options is limited. This reflects
not only the logistical challenges of undertaking ran-
domized controlled trials (RCTs) in an unpredict-
able and episodic condition that reaches its maximum
intensity within just 24 h of onset, but also the fact that
current treatments have been available for decades and
are accepted as normal clinical practice without a per-
ceived need for further research evidence to confirm
their efficacy.
NSAIDs
An oral NSAID is by far the most commonly used treat-
ment for acute gout in general practice.15
However, just
one placebo-controlled RCT has confirmed that an
Key points
■■ Many established treatments are already available for managing gout effectively
■■ Lowering serum uric acid to a target level below the saturation point for urate
can effectively ‘cure’ gout
■■ Health professionals need to be aware of current best practice for the
management of gout
■■ Patient education and discussion of illness perceptions is key to addressing
patient-specific risk factors and ensuring adherence to an individualized
management plan
■■ Shared decision-making between a fully informed patient and a health
practitioner, whether a doctor, nurse or pharmacist, can greatly increase the
likelihood of a ‘cure’ and lead to improvements in patient outcomes
NSAID (tenoxicam 40 mg daily) is better than placebo
at reducing pain at 24 h.16
Because of the severity of
pain experienced in acute gout, the NSAID should be
commenced at its maximum dose, rather than titrat-
ing upwards from the lower end of the dose range.11,12,14
Although some doctors still hold the traditional belief
that indomethacin is more effective than other NSAIDs,
studies comparing different quick-acting NSAIDs dem­
on­strate their equal efficacy in acute gout.17–21
The
main issue with oral NSAIDs is, of course, their safety.
To reduce the risk of upper gastrointestinal ulcera­
tion, bleeding and perforation, co-administration of
a ­proton-pump inhibitor (PPI) is recommended,22
although the addition of a PPI does not alter the risk of
small and large bowel involvement. In addition, NSAIDs
can cause acute renal failure and are contra­indicated in
patients with CKD.23
Selective cyclo­oxygenase (COX)‑2
inhibitors compare favourably with non­selective
NSAIDs24,25
for the treatment of acute gout in terms of
similar efficacy but lower incidence of serious gastro­
intestinal events; however, concerns remain over the
cardio­vascular and renal toxicity of COX‑2 inhibitors
with frequent or chronic use.26
In the UK, co-­prescription
of a PPI is recommended with these drugs.22
Colchicine
Along with oral NSAIDs, oral colchicine is recom­mended
as a first-line treatment for acute gout attacks.11,12,14
Interest in IL‑1 inhibition with bio­logic agents for
acute gout has arisen due to the action of these drugs
on the NLRP3 inflammasome; however, colchi­cine is
also known to exert one of its many actions here27
and
is a much less expensive option. Figure 2 demonstrates
the metabolism and various mechanisms of action
of colchicine.
Oral colchicine has been used in clinical practice for
centuries, but only two placebo-controlled RCTs have
been published, both of which confirm its efficacy in
acute attacks.28,29
These studies both demonstrated that
colchicine significantly reduced pain compared with
placebo, but high-dose regimens (4.8 mg total over 6 h)
were associated with substantial gastrointestinal dis-
turbance (nausea, vomiting or diarrhoea). Low-dose
regimens (1.8 mg total over 1 h) were equally effective
at reducing pain but adverse effects were minimized.
Hence, current EULAR guidelines recommend a maxi­
mum dose of 0.5 mg colchicine three times a day,11
and
the ACR guidelines14
recommend a regimen involving
a loading dose of 1.2 mg then 0.6 mg 1 h later, followed
after a further 12 h by 0.6 mg once or twice daily. Such
low doses seem to be effective and well-tolerated in
clinical practice. Even lower doses (0.5 mg once or twice
daily) can be tried in patients with persistent adverse
effects and in those with moderate to severe CKD.30
Following the publication of the ACR guidelines, an
equivalent dosing is likely to become more widely
adopted in Europe—with a modified dosage of a 1.0 mg
loading dose followed 1 h later by 0.5 mg on the first day,
then 0.5 mg two to four times a day on subsequent days.
Colchicine is considered to work best when commenced
Dietary purines
Uric acid pool
(serum, tissues)
30% 70%
70% renal
excretion
30% gut
elimination
MSU crystals
Purine nucleotides
and bases
Tissue nucleotide
synthesis and metabolism
Osteoarthritis
Ageing
ObesityHigh-purine
diet
■ Genetic factors
■ Metabolic syndrome
(hypertension,
insulin resistance,
hyperlipidaemia)
■ Renal impairment
■ Drugs that reduce
renal function
Figure 1 | Metabolism of uric acid and risk factors for gout. Crystallization of MSU
occurs when uric acid levels exceed the saturation point, through inefficient
elimination or excessive production of uric acid. The multiple risk factors for the
formation of urate crystals are shown in yellow boxes at their sites of action.
Abbreviation: MSU, monosodium urate.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  273
within 24 h of first symptoms of an acute attack, and the
ACR guidelines recommend that colchicine is not initi-
ated more than 36 h after symptom onset. Treatment is
usually continued until the acute attack has subsided.
Colchicine should be used at low doses and with cau­
tion in patients taking a cytochrome P450 3A4 inhibitor
(ciclosporin, ketoconazole, ritonavir, clari­thromycin,
erythromycin, extended-release verapamil, extended-
release diltiazem)27,30,31
owing to drug–drug inter­
actions,31,32
particularly in people with renal impairment,
and those taking a statin should stop this medication
temporarily while on colchicine.27
Intravenous colchicine
is highly toxic and not recommended.11,12,14
Corticosteroids
Intra-articular corticosteroids
Aspiration of an affected joint followed by injection of
corticosteroids (Figure 3) is ideal for treatment of acute
gout attacks,11
particularly in a hospital setting where
first-line use of this intervention can provide prompt
relief with minimal adverse effects. This treatment is
a particularly useful option where colchicine, NSAIDs
or oral corticosteroids are contraindicated. Aspiration
itself very quickly reduces the extreme pain of gout,
pre­sumably by lowering the marked intra-articular
hyper­tension caused by acute crystal-induced synovitis.
Prac­tical difficulties with intra-articular aspiration and
injection arise if the gout attack is polyarticular or affects
the midfoot, or if no physician with sufficient expertise
is available (the usual situation in general practice). One
uncontrolled trial has been published, which supports
the use of intra-articular corticosteroid injection.33
Although no evidence exists to guide optimal dosage, as
for other arthropathies most physicians use a long-acting
corticosteroid and adjust the dose according to the size
of joint affected.34
Oral corticosteroids
Oral prednisolone can be used where colchicine or
NSAIDs are contraindicated or have failed to provide
sufficient relief,11
and this method of delivery of cortico­
steroids is the one most commonly used in general
practice. As with intra-articular corticosteroids, no
evidence is available to guide optimal dosage. One key
trial demonstrated that, in patients with acute gout, oral
prednisolone 35 mg daily was equivalent in efficacy
to oral naproxen 500 mg twice a day.35
Another trial
demon­strated that 30 mg prednisolone daily for 6 days
was equivalent in efficacy to intramuscular diclofenac
75 mg plus 50 mg indomethacin orally on day 1 followed
by oral indomethacin 50 mg three times a day for 2 days
then 25 mg three times a day for 3 days.36
Such doses
of oral corticosteroids administered in short courses are
commonly used in general practice for other conditions
(for example, asthma) and considered overall to be a safe
option, especially in patients with comorbidities.
Intramuscular corticosteroids
Intramuscular delivery of corticosteroids is another
alternative treatment that is mainly limited to hospi-
tal settings. Intramuscular corticosteroids are usually
admini­stered as a single injection but, again, there is
no con­sensus on dose. In one trial, 60 mg intra­mus­
cular triamcinolone acetonide was equivalent in effi-
cacy to oral indomethacin 50 mg three times daily for
the treatment of acute gout.37
However, higher intra­
muscular doses—120 mg of triamcinolone acetonide or
m­ethylprednisolone—are often used to control an acute
flare of rheumatoid arthritis or other painful inflamma-
tory arthropathies38
and many physicians use such doses
for acute gout.
Biologic agents
Considerable interest exists among rheumatologists in
the use of biologic agents for the rare situation where
all of the above treatments are considered to be contra-­
indicated or impractical. However, available biologic
agents that inhibit IL‑1 and which have been assessed
in treatment of acute attacks of gout—specifically ana­
kinra,39,40
canakinumab41–43
and rilonacept44–46
—have
either had no comparator or have been compared to a
possibly suboptimal dose of triamcinolone (40 mg intra-
muscularly) rather than a more commonly used short
course of oral prednisolone (for example, 30–35 mg
prednisolone daily). Although some modest advan-
tages of biologic agents have been reported, these treat-
ments remain comparatively very expensive, and as yet
are largely unlicenced for acute gout in most countries
(canakinumab is now licenced for use in Europe, but not
in the UK or USA).
Main effects of colchicine
■ Binds to tubulin in cells
(affecting cell division)
■ Inhibits function of leukocytes
■ Inhibits chemotactic factors
■ Inhibits inflammasome and
IL-1 production
■ Inhibits release of histamine
from mast cells
Oral administration
80–90% faecal excretion
Liver
Kidney
10–20% renal excretion
Enterohepatic
recycling
Figure 2 | Metabolism and mechanisms of action of colchicine. After oral
administration and gastrointestinal absorption, colchicine is primarily metabolized
by the liver but undergoes substantial enterohepatic circulation. Its pharmacological
effects are multiple and varied. Colchicine is excreted primarily in faeces but also
in urine.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
274  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
Physical treatment
In support of widespread clinical experience, one small
trial (n = 19) examining the effect of adding localized ice
therapy to a standardized treatment for acute attacks47
found greater improvements at 1 week in those who
received ice. Locally applied ice packs are therefore
recom­mended as a simple and safe adjunct to pharmaco­
logical therapy. No RCTs are available to support resting
of the affected joint,12
but such common-sense advice
obviously accords with the patient’s natural inclination.
Issues related to treatment of acute attacks
NSAIDs are readily available but unfortunately are
contra­indicated in many patients with gout owing to
age, comorbidity, concomitant drug therapy or renal
impair­ment.48,49
Pharmacoeconomic analysis has
also shown oral NSAIDs to be less cost-effective than
oral corticosteroids or colchicine, largely because of
their adverse effects.50,51
NSAIDs and colchicine have
not been directly compared with respect to their effi-
cacy, but colchicine might be more applicable to most
patients, with the caveats of severe renal impairment and
certain drug interactions listed above. Where expertise
is available, joint aspiration and injection with cortico­
steroids is often the safest option and this treatment
should be regarded as recommended best practice in a
hospital setting. The key to successful management of
acute attacks of gout is to assess and treat each patient
individually, discussing with them which option is
most acceptable. Any treatment should be started as
early as possible and drugs can be used in combination
if needed (for example, intra-articular corticosteroids
with oral colchicine).14
Long-term management
The aim of long-term therapy is to ‘cure’ gout by lowering
SUA levels to below the saturation point for urate—that
is, to at least below 360 μmol/l (6 mg/dl)11
—thus promot-
ing crystal dissolution and preventing formation of new
crystals. The most effective method of achieving this
SUA level is the use of urate-lowering therapy (ULT).
Before commencement of ULT, each patient should be
given a clear verbal explanation of the causes of gout,
backed up by written information (for example, Arthritis
Research UK’s patient information booklet52
), along with
an individualized explanation of the relevant risk factors
that could elevate their SUA levels. The content listed in
Box 1 was used in a study to explain to patients the causes
and risk factors for the development of gout, and the
patients subsequently demon­strated excellent ad­herence
to individualized management plans.53
One of the many barriers to care is a stereotyped view
of gout and the feeling that gout is self-induced through
poor lifestyle, gluttony and excessive alcohol intake.54
Many patients have seen old cartoons that empha­
size such lifestyle factors (Figure 4). Patients can be
embarrassed to have gout—particularly women, who
often think it is a man’s disease—which can lead to a
dis­inclination to seek help. Negative views about gout
are associated with poorly controlled disease and lower
adher­ence to ULT.55–57
Therefore, it is important that the
explana­tion should be non-accusatory, emphasizing that
having gout is not necessarily the patient’s fault, and
that the condition is curable with treatment. Exploration
and full discussion of the patient’s perception of their
illness is crucial if they are to be sufficiently motivated to
adhere to an appropriately developed management plan.
Individualized, constructive advice on lifestyle modi-
fications that might reduce predisposition to hyper­
uricaemia should be given if appropriate. This advice
might include, for example: reduce weight by use of
recom­mended strategies if overweight or obese;58
reduce
excessive intake of beer and spirits;59
reduce exces-
sive intake of purine-rich foods (such as red meat and
seafood) and consume proportionately more dairy pro­
ducts;60
reduce intake of fructose-rich drinks and fruit
(apples, oranges);61
and cease any retinol supplementa-
tion (retinol increases SUA compared with β-carotene,
which reduces SUA).62
Although these recommendations
are based on observational studies rather than RCTs,
clearly many health reasons exist for tackling being over-
weight or drinking more than the recommended levels
of alcohol, and receiving indivi­dualized lifestyle advice
might enable the person with gout to take control of their
condition and improve their self-efficacy. However, the
benefit to gout of lifestyle modification should not be
overemphasized since, firstly, this emphasis reinforces
the negative perceptions of gout as a self-inflicted condi-
tion and, secondly, ULT will still be required to achieve
SUA levels low enough to treat the condition within a
reasonable timescale.
With respect to predisposition to hyperuricaemia and
development of gout, being overweight or obese increases
endogenous production of uric acid63
and is probably a
a b
Figure 3 | Management of acute gout attack. a | Example of an acute attack at the
first metatarsophalangeal joint, known as ‘podagra’. b | Aspiration of an acutely
inflamed joint; note the turbidity of the synovial fluid. Aspiration followed by
injection of corticosteroids is safe, rapidly reduces the extreme pain of acute gout
and enables confirmation of the diagnosis. In a hospital setting such treatment,
especially in an ill patient with multiple comorbidities, should be considered
best practice.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  275
more important factor than individual dietary compo-
nents. However, interest is growing in diet­ary altera­tions
that might reduce the frequency of acute attacks in people
with established gout, rather than influence hyper­
uricaemia and crystal deposition. A long-­recognized
paradox is that many patients link the trig­gering of an
acute attack to excessive consumption of dietary purine
food and alcohol, which might be expected to increase
SUA levels, whereas the accepted triggers for an acute
attack (for example, intercurrent illness, surgery, initia-
tion of ULT) are associated with a rapid decrease in uric
acid levels that is hypothesized to encourage partial dis-
solution and subsequent ‘shedding’ of preformed crys-
tals into the joint space. A possible explanation for this
paradox is that an increase in free fatty acids following
a large meal might act as a ‘second signal’ and syner-
gize with urate crystals to trigger the inflam­masome
via ASC/caspase 1‑driven (but not NALP3-driven)
release of IL‑1β.64
Conversely, the dairy fractions glyco­
macropeptide and G600 milk fat extract might inhibit
triggering of the inflammasome by urate crystals.65
An
RCT published in 2012 showed that daily intake of a
skimmed milk powder enriched with glycomacropeptide
and G600 reduces the frequency of acute gout attacks
over a 3‑month period.66
Similarly, evidence from a
1-year case–crossover study showed that regular intake
of cherry reduces the frequency of acute attacks, possibly
owing to high levels of anthocyanins, which possess anti-
inflammatory and antioxidant properties.67
There­fore,
if patients are interested in dietary alterations, it might
be very reasonable to advise avoidance of large, rich
meals and to consider appropriate dairy supplements
and cherry or cherry extract as safe self-­management
st­rategies for reducing attack frequency.
Other modifiable risk factors for hyperuricaemia
can also sometimes be addressed. For example, chronic
diuretic therapy for hypertension (rather than for heart
failure) could be switched, where feasible, to an alterna-
tive antihypertensive regimen, bearing in mind that in
an observational cohort β-adrenergic blocking agents,
angiotensin-converting enzyme inhibitors and non-
losartan angiotensin II receptor blockers increased
SUA whereas losartan and calcium-channel blockers
lowered SUA68
(via reduced or enhanced renal excretion
of uric acid, respectively). In some patients with end-
stage renal disease, renal transplantation might nor-
malize their SUA. Importantly, comorbidities that are
associated with hyperuricaemia, such as hyper­tension,
hyperlipidaemia and hyperglycaemia,7,69–71
need to be
considered and optimally managed. These conditions are
important in their own right, but tight control of each
can indepen­dently reduce SUA levels through improved
renal ex­cretion of uric acid.72,73
Urate-lowering therapy
ULT is a key component of curative gout therapy and
needs to be fully explained to each patient and upwardly
titrated against a specific target SUA level. Patient edu-
cation is crucial and, as shown in a proof-of-principle
study,53
ensures excellent adherence to ULT. Upwards
titration of ULT to achieve a level of SUA well below
the saturation threshold of 360 μmol/l is important to
enable the dissolution of existing crystals and prevent
joint damage—in effect ‘curing’ gout. The lower the level
of SUA obtained, the faster the dissolution of crystals
and reduction in size of any tophi.74,75
It is important to
explain to patients that they will remain at risk of acute
Box 1 | Key messages when explaining the nature of gout
Explanation of the causes of gout, including the following key points:
■■ We know its cause: deposition of urate crystals in and around peripheral joints
■■ Crystals form when SUA levels rise above the critical ‘saturation point’ for
crystal formation
■■ In people with persistently raised SUA levels, crystals slowly but continuously
accumulate without causing symptoms
■■ When sufficient crystals have formed in cartilage, some occasionally ‘spill
out’ into the joint cavity, triggering severe inflammation of the joint lining and
presenting as an acute attack
■■ Over many years, acute attacks can increase in frequency and spread to involve
other joints
■■ In addition to acute attacks, continuing crystal deposition might eventually
result in hard, slowly expanding lumps of crystals (tophi) that can cause
pressure damage to joint cartilage and bone and can even appear as palpable
lumps under the skin
■■ In some people, tophi could result in irreversible joint damage and cause
regular chronic pain on using the joints
■■ Reduction and maintenance of SUA levels below the saturation point stops
production of new crystals and encourages existing crystals to dissolve, so
eventually there are no crystals and therefore no gout
Explanation of relevant risk factors that elevate SUA levels above the saturation
point, including:
■■ Hereditary factors that result in some people having relatively inefficient renal
excretion of uric acid
■■ High body mass—the majority (around two-thirds) of uric acid is made by the
body’s cells and this production increases with obesity
■■ A purine-rich diet—around one-third of uric acid comes from the diet
■■ Drugs (e.g. diuretics) that reduce the kidney’s ability to excrete uric acid
■■ Chronic renal impairment associated with ageing or kidney disease
Abbreviation: SUA, serum uric acid.
Figure 4 | “Punch cures the gout, the colic, and the ‘tisick” (anonymous). Historical
sterotypes of gout persist and can act as barriers to adequate care.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
276  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
attacks until all existing crystals are dissolved, that if
they do develop an attack it is not a sign that their ULT is
ineffec­tive or making things worse, and that they should
continue the ULT while they manage an acute attack.
Gradual upward titration is probably important for reduc-
ing the risk of acute flare, and in one study this strategy
for initiating ULT seemed not to increase the frequency
of attacks more than would have been expected (based on
the frequency of self-reported attacks the previous year).53
Limited evidence is available to guide the commence-
ment of ULT. Currently, indications for ULT include
recurrent attacks of gout, clinically detectable tophi,
joint damage or nephrolithiasis11
—in other words, well-
established and relatively severe gout. However, urate
crystal deposition is increasingly realized to be far more
extensive than would be thought by clinical assessment
alone. For example, ultrasonography studies published in
2011 and 2012 demonstrate obvious crystal deposition
in a substantial proportion of people with asympto­matic
hyperuricaemia,76,77
and dual-energy CT has shown
extensive urate deposition in people with established
gout, including at unsuspected sites such as the distal
patellar tendon.78
In patients with gout, synovial fluid
aspirated from knees that have not been the site of an
acute attack will often show urate crystals.79
Therefore,
it seems that crystal deposition is likely to be exten-
sive in many peripheral joints even at the time of first
presentation and that every person with gout is techni-
cally ‘tophaceous’ in terms of having microtophi in and
around their joints. Such a perspective is causing a trend
towards earlier commencement of ULT, closer to the
time of first diagnosis, particularly if a patient is young
(for example, in their twenties, thirties or forties) or has
a low estimated glomerular filtration rate.12
Certainly the
patient should at least be informed at this time about
the availability and expected benefits of ULT and be
involved in the decision as to whether to start ULT.
Most clinicians advocate delaying initiation of ULT
until an acute attack has settled, owing to concern that
commencing ULT will prolong the attack or precipitate
a polyarticular flare. However, one placebo-controlled
RCT found no difference in pain or flare rate when
ULT with allopurinol (plus colchicine prophylaxis) was
started during an acute attack compared with a delay of
10 days.80
Many general practitioners perceive logistical
advantages to initiating ULT at the time a patient presents
with an acute attack and argue that many patients will not
re­attend 4 weeks later when everything is back to normal.
Nevertheless, it seems sensible to provide enough infor-
mation at the time of the acute attack to encourage sub-
sequent reattendance and to wait until the second visit,
when the patient is not in severe pain, to fully explain the
disease and discuss its long-term management.
Xanthine oxidase inhibitors
Xanthine oxidase inhibitors (XOIs) are the usual first-
choice ULT, with allopurinol favoured over febuxostat
due to cost considerations and long-term safety data.
XOIs reduce endogenous production of uric acid by
inhibiting the conversion of hypoxanthine to xanthine
and of xanthine to uric acid. Table 1 compares the key
features of the two currently available XOIs.
Allopurinol
Allopurinol is a purine analogue and a nonspecific com-
petitive inhibitor of xanthine oxidase. Its active metabo-
lite, oxypurinol, is predominantly excreted via the kidney.
Although no placebo-controlled RCTs have confirmed
the efficacy of allopurinol, decades of clinical experience
and several head-to-head studies81–83
have shown it to be
an effective, well-established, cheap and relatively safe
ULT, and it is therefore recommended as first-line ULT.11
The dose range of allopurinol is large, meaning it can
be titrated in small increments for maximum effective-
ness. In a 2013 observational study in the UK,53
the
median dose of allopurinol required to reach the thera-
peutic target of SUA ≤360 μmol/l (achieved in 90% of
participants) was 400 mg once daily. This dose is higher
than the commonly prescribed daily dose of 300 mg,
demonstrating the importance of adjusting therapy for
maximum effectiveness in individual patients. It might
be that 300 mg was adequate for most people when the
first dose-ranging studies for allopurinol were published
around the 1960s, but that population-level increases in
height and BMI mean 300 mg is now suboptimal; when
used in RCTs in the USA in 2005, this dose achieved the
therapeutic target in only around 20% of patients.84,85
An
initial dose of 100 mg daily is recom­mended, with sub-
sequent incremental increases of 100 mg approximately
every month (up to a maximum of 900 mg), stopping
these increases at the dose at which SUA is lowered
to 360 μmol/l (6 mg/dl)13
or preferably 300 μmol/l
(5 mg/dl). The evidence for dose-­adjustment of allo­
purinol in patients with renal impairment, using 50 mg
increments, is inconclusive.86–89
Stamp et al.89
suggest that
dose-adjustment algorithms based on creatinine clear-
ance of allopurinol were overly cautious and that with
gradual upward titration the target SUA level can often
be reached safely, even in the setting of renal impairment,
and at doses that are above those recommended.
Table 1 | Comparison of XOIs
Characteristic Allopurinol Febuxostat
Action Nonspecific XOI Specific XOI
Elimination Renal Hepatic
Daily dose* (50 mg), 100–900 mg (40 mg), 80 mg, 120 mg
DRESS Rare Rare
Cautions Severe renal impairment Heart failure (NYHA class III
or IV), hepatic failure,
eGFR 30 ml/min/1.73 m2
Interactions Cytotoxic drugs metabolized
by XO (for example,
azathioprine, mercaptopurine)
Warfarin
Cytotoxic drugs metabolized by
XO (for example, azathioprine,
mercaptopurine)
Cost of 28 tablets‡
£1.06 (300 mg tablets) £24.36 (80 mg or 120 mg
tablets)
*The doses in parentheses can be used in patients with renal impairment but are not routinely used.
‡
According to information from the British National Formulary.135
Abbreviations: DRESS, drug reaction with
eosinophilia and systemic symptoms; eGFR, estimated glomerular filtration rate; NYHA, New York Heart
Association; XO, xanthine oxidase; XOI, XO inhibitor.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  277
Allopurinol is usually well-tolerated, by approximately
8–9 out of 10 patients,53,82
the usual cause of intolerance
being nausea, gastrointestinal disturbance, headache or
rash. However, allopurinol can rarely cause a serious syn-
drome of drug reaction or rash with eosinophilia and sys-
temic symptoms (DRESS; previously termed allopurinol
hypersensitivity syndrome), which can cause fever, severe
cutaneous adverse reactions (SCAR; including toxic
epidermal necrolysis and Stevens–Johnson syndrome),
hepatitis, vasculitis, renal impairment and sometimes
death.90
Allopurinol-induced DRESS usually occurs
within the first few months of treatment and reported
risk factors include renal impairment, concomitant use
of diuretics, initiation of treatment with a fixed dose of
300mg and, particularly in Korean, Chinese and Thai
populations, presence of the HLA‑B*5801 allele.12,91
It might be reasonable to consider screening for this
polymorphism in populations with a high prevalence
of HLA‑B*5801.91
The precise incidence of DRESS from
allopurinol use is unclear but allopurinol is reportedly the
most frequent cause of SCAR.90
Although rare, the occur-
rence of severe DRESS and SCAR is another reason to
start low and gradually increase the dose of allopurinol.92
Allopurinol has also been shown to be beneficial in
cardiovascular disease93,94
and renal disease,95,96
both of
which commonly coexist with or occur as a complication
of gout. Although still controversial and not currently
licenced for these indications in most countries, this is
another potential benefit of ULT which, if explained to
patients at risk of cardiovascular or renal disease, might
further encourage adherence to treatment. Whether the
cardio-renal benefits result from lowering of SUA or
from xanthine oxidase inhibition remains uncertain.97
Febuxostat
Licenced in 2008 in Europe, febuxostat is a non-purine,
highly specific XOI that undergoes predominantly
hepatic metabolism. Two placebo-controlled double-
blind RCTs have demonstrated its efficacy in reducing
SUA.84,98
Two further double-blind RCTs compared
doses of 40–240 mg febuxostat against a fixed daily dose
of 300 mg allopurinol85,99
and showed that febuxo­stat was
superior at doses of 80 mg and 120 mg daily. How­ever, the
trials that included allopurinol did not allow for upward
titration of the allopurinol dose against SUA level, which
is the established best clinical practice. Furthermore,
in these RCTs84,85
approximately 30% of participants
on febuxostat do not reach the therapeutic target (SUA
360 umol/l) at the highest recommended dose (120 mg
daily), whereas in clinical practice upward titra­tion
of allopurinol in those who tolerate it would usually
achieve this target at doses well under the maximum dose
(900 mg daily).53
The most common adverse event deemed attributable
to febuxostat in the early clinical trials was liver-­function
test abnormalities, but the possibility of an adverse
cardio­vascular signal (more myocardial infarction, stroke
and cardiovascular death in febuxostat than allopurinol
participants, though nonsignificant) led to febuxo­stat
not being recommended in people with heart failure.100
Long-term safety studies to address these con­cerns
are in progress. Also, although febuxostat was initially
thought to be free of the serious SCAR and DRESS reac-
tions associated with allopurinol, some cases of severe
hypersensitivity cutaneous reactions have subsequently
been reported.101,102
Undoubtedly, the main advantage of
febuxostat over allopurinol is that it can be used without
dose-adjustment or concern over toxicity in people with
renal impairment (although it is not recommended for
use in those with an estimated glomerular filtration rate
30 ml/min/1.73 m2
). Disadvantages include the inability
to slowly increase the dose (only 80 mg and 120 mg tablets
are available in most countries, and provocation of acute
gout attacks at the starting dose of 80 mg is likely),84,85,98,99
cardiovascular safety concerns, and high cost. Although
the ACR guidelines (which did not consider cost) do
not favour one XOI over the other,13
the UK National
Institute for Health and Clinical Excellence (which does
consider cost utility) recommends allo­purinol as the
first-line XOI, with febuxostat recommended as a second
option in those who cannot tolerate allopurinol or in
whom it is contraindicated or ineffective.103
Uricosuric drugs
Available uricosuric drugs act predominantly on
urate anion exchanger 1 (URAT1)—an organic anion
t­ransporter—to prevent reuptake of uric acid at the proxi­
mal renal tubule and thus increase renal excretion of uric
acid (Figure 5). The resulting higher con­centration of
uric acid in the collecting tubules can predispose to uric
acid stone formation, so the patient should be advised to
drink plenty of fluids and remain well-hydrated. Three
uricosuric drugs are licenced and well-established as ULT:
benzbromarone (50–200 mg daily), probenecid (250–
500 mg twice daily) and sulfin­pyrazone (200–800 mg
daily). Although inexpensive, their use is limited by lack
of availability in most countries. These medications are
all effective at reducing SUA82,104–107
and have dose ranges
that readily enable stepwise dosage increases. Probenecid
and sulfin­pyrazone are contraindicated in patients with
severe renal impairment or nephrolithiasis, which further
Basolateral
membrane
Renal proximal
tubule
epithelial cell
Apical (brush border)
membrane
Probenecid
Benzbromarone
Sulfinpyrazone
Sulfinpyrazone
OAT1
OAT3
GLUT9
URAT1
OAT4
Uric acid
Uric acid
Probenecid
Benzbromarone
Proximal tubule lumenCirculation
Figure 5 | Mechanism of action of uricosuric drugs at the proximal renal tubule.
Uricosuric drugs licenced for gout (shown in blue boxes) act on URAT1 to prevent
re-uptake of uric acid and thus increase its renal excretion. As this is a simplified
schema, not all transporters are shown. Abbreviations: OAT, organic anion
transporter; GLUT9, glucose transporter type 9; URAT1, urate anion exchanger 1.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
278  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
limits their use. Benzbromarone is not contraindicated
in those with mild or moderate renal impairment and
can still be very effective in this situation. The avail-
ability of benz­bromarone became limited mainly due
to concerns over reports of hepatotoxicity, particularly
in Asia, although the reported risk in European popula-
tions is estimated to be at most 1 in 17,000 patients.108
Nevertheless, liver function should be checked whenever
SUA level is meas­ured, especially in the first few months
of initiation and dose-adjustment of uricosuric therapy.
Other available ULTs
Losartan,109
oral vitamin C supplements,110,111
and feno­
fibrate112
all have direct uricosuric effects and have
been shown to reduce SUA levels in people with hyper­
uricaemia. For losartan and fenofibrate, this uricos­
uric action is in addition to their beneficial effects on
hypertension and hyperlipidaemia, respectively. How­
ever, the urate-lowering effects of all three approaches
are fairly modest and, on their own, are unlikely to
achieve the therapeutic target level of SUA.113
These
agents remain unlicenced for use in gout, but losartan
and feno­fibrate can be considered as useful adjunctive
ther­apy for patients who require treatment for coexistent
hy­pertension or hyperlipidaemia.
Unlike some other animals and bacteria, humans lack
uricase—an enzyme that converts uric acid to highly
soluble allantoin. Pegloticase is a pegylated uricase
that was recently licenced (2010 in the USA, 2013 in
Europe) for use in ‘treatment-refractory’ gout. This drug
is administered by repeated intravenous infusion (8 mg
every 2 weeks seems optimal), a regimen that limits
its use to the hospital setting. Pegloticase is extremely
effective at rapidly reducing SUA to very low levels for
several weeks.114
However, it is also antigenic,115
with a
risk of anaphylaxis and infusion reaction, so premedi-
cation with antihistamine and corticosteroids in an
appropriately staffed facility is recommended.116
Con­
cerns also remain over cardiovascular risk, and this
drug should be used with caution in people with heart
failure. A large proportion of patients undergoing repeat
infusions develop blocking antibodies against the PEG
component,115
which not only reduces ULT efficacy but
also increases the risk of adverse reactions. Therefore,
following successful initiation of treatment, a subse-
quent preinfusion SUA 360 μmol/l is an indication to
withdraw treatment (recom­mended by the manu­fac­
turers). Although pegloticase is seemingly very efficient
at induc­ing rapid regression of tophi, the number of
patients who are truly refractory to other standard ULTs
is likely to be extremely small, and because of safety and
tolerability concerns, and the cost and logistics of deliv-
ery of this drug, its use is likely to be very restricted.
Rasburicase, a uricase that has been licenced for many
years for tumour lysis prophylaxis, has also been used to
treat ‘refractory’ gout where intolerance or renal impair-
ment precludes the use of other ULTs.117,118
However,
although some patients seem to tolerate repeated doses,
rasburicase is unlicenced for gout and is more antigenic
than pegloticase.119,120
ULTs in development
Other drugs that are undergoing clinical investigation
as potential ULTs include RDEA594 (lesinurad), which
is a uricosuric,121
and BCX4208 (ulodesine), which is a
purine nucleoside phosphorylase inhibitor.122
How­ever,
these agents are not yet available or licenced for use in
gout, either as monotherapy or in combination with
an XOI.
What is the optimum SUA level?
Current guidelines agree that SUA should be reduced and
maintained to a level below 360 μmol/l (6 mg/dl)11,13
to
prevent further crystal formation and to encourage dis-
solution of exisiting crystals.123
However, the lower the
SUA, the faster the dissolution of crystals and tophi75
and
the sooner the patient reaches the state of being ‘cured’.
Therefore, particularly in patients with a long history
of gout79
or the presence of tophi, an initial target of
300 μmol/l (5 mg/dl) might be more successful; this
is the target recommended by the BSR.12
Concern is
increasing, however, that lifelong maintenance (that
is, after acute attacks have stopped and any tophi have
resolved) of SUA at very low levels might increase the
risk of neuro­degenerative diseases such as Parkinson,
dementia and multiple sclerosis, which appear to be
negatively associated with gout.124
This is because uric
acid is a strong antioxidant97
and although this benefit
is countered by its other detrimental effects on vascular
endothelium, its antioxidant benefits may predominate
within the central nervous system. Although there is no
absolute agreement on the optimal SUA level that asso-
ciates with no disease risk in humans, the concept of an
initial crystal-clearing ‘induction phase’ followed by a
later ‘maintenance phase’ of ULT is one that is increas-
ingly discussed.125
We suggest that the initial SUA target
to effect a ‘cure’ should be well under 300 μmol/l for
the first 3–5 years of treatment but that subsequently,
when no further attacks have occurred and any tophi
have regressed, the dose of ULT should be reduced to
allow the SUA to rise somewhat but remain below the
saturation point to prevent crystal formation (that is,
in the range 300–360 μmol/l) (Figure 6). The length of
time the patient should continue ULT is guided by SUA
level. For most patients, use of this target could mean
lifelong therapy, but at a low dose. However, in patients
with an attributable risk factor that can be success­fully
modified (for example, by stopping long-term thia­
zide therapy, successful renal transplantation, marked
reduction in BMI) cessation of ULT might be consid-
ered after the induction phase or some time into the
maintenance phase.
Flare prophylaxis
Initiation of ULT can provoke acute attacks, some-
times involving several joints and even joints pre­viously
un­affected by gout. This flare of disease is particularly
likely if SUA is rapidly and substantially lowered by a
high or very efficient dose of ULT (for example, allo­
purinol 300 mg daily, febuxostat 80 mg daily, uricase).
It is thought to result from the rapid partial dissolution
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  279
of and thus easier ‘shedding’ of crystals. If the patient
is not fully informed about the possibility of this flare
in advance, such an event might cause a patient, quite
reason­ably, to give up on their ULT and even to be
unwilling to consider further advice for their gout.
To reduce the risk of ULT-induced flares, the EULAR
guidelines recom­mend co-prescription of either low-
dose colchicine (usually 0.5 mg once or twice daily) or
an oral NSAID (plus PPI) as prophylaxis for the first few
months of ULT.11
Certainly one RCT has confirmed that
colchicine is more effective than placebo in reducing
the number of acute attacks during the first 6 months of
allopurinol treatment.126
Additionally, a post hoc analysis
of three RCTs of febuxostat, in which patients received
either 8 weeks or 6 months of prophylaxis with naproxen
250 mg twice daily or colchicine 0.6 mg daily, found an
increase in flare rate (up to 40%) at the end of 8 weeks of
prophylaxis, whereas flare rates remained low (3–5%) at
the end of 6 months of prophylaxis.127
However, low-dose
colchicine, and possibly NSAIDs, might reduce the flare
rate in people who are not on ULT, and a key question is
whether initiation of ULT using the recommended slow
up-titration regimen (as is possible with allo­purinol
in particular) actually causes more attacks than may
have been expected otherwise. Furthermore, in a study
(n = 96) where patients were given a full explanation of
the possible risk of flare from ULT and of the avail­ability
of prophylaxis, the vast majority (96%) preferred to take
just the one drug (ULT) without prophylaxis and to just
manage acute flares as they arose.53
Most patients in this
study (65%) experienced fewer attacks in the year fol-
lowing ULT initiation than in the preceding year and
only one in five reported more attacks (mean 2.7 more).
Further studies of whether or not slow up-titration
increases the risk of flare are warranted to better inform
shared decision-making. Rilonacept and canakinumab
have shown efficacy in gout prophylaxis, but as already
noted these agents are very expensive and currently not
licenced for this use.45,46
Improving the standard of care
Despite the availability of effective treatments for gout
and of published recommendations to guide best prac-
tice, it is apparent that the current standard of care for
patients with gout is more than just “suboptimal”.128–132
This criticism applies to both primary and secondary care
physicians, including rheumatologists. Few patients are
given an adequate explanation of the cause of gout and its
possible outcomes, or educated as to the avail­ability and
possible benefits of treatment. Most doctors concentrate
solely on the management of acute attacks rather than
long-term therapy, and only one-third to one-half of gout
patients ever receive ULT.1,10,15
Very few general practi-
tioners who offer ULT aim for a speci­fic SUA level or
serially check SUA133
and most use allopurinol at a fixed
dose of 300 mg (which is insufficient for most patients).53
This ‘standard of care’ leads to many people with gout
continuing to experience acute attacks and being at
unnecessary risk of developing joint damage. Because
appropriate information is not provided, patients with
gout show poor adherence to any ULT offered and, if
they stop ULT because ‘the tr­eatment’s not working’, they
are often misclassified as ‘treatment failures’.130
Barriers
to effective care can develop both from the patient and
the physician (Box 2).54
Full clinical assessment followed by clear informa-
tion from the practitioner and subsequent involvement
Time
SUAlevel
Dissolution of
MSU crystals
360μmol/l
Introduction
of ULT
Figure 6 | Graph representing clearance induction and
maintenance phases of therapy for gout. In the induction
phase, ULT reduces the SUA level to well below the
saturation threshold (indicated by the dashed line), to enable
dissolution of existing crystals and ’cure’ of gout. In the
maintenance phase, ULT is reduced following this ‘cure’, and
the SUA level is allowed to rise to just below the threshold for
maintenance. Abbreviations: MSU, monosodium urate; SUA,
serum uric acid; ULT, urate-lowering therapy.
Box 2 | Barriers to effective management of gout54,130
Patient barriers
■■ Patients lack knowledge and understanding about the
causes and consequences of gout
■■ Gout is often not considered as ‘arthritis’ or as
something serious, and is often associated with humour
■■ Gout is considered a man’s disease; women might be
unwilling or embarrassed to receive the diagnosis, men
might be reluctant to seek medical attention
■■ Gout is considered as a normal part of ageing, self-
inflicted by overindulgence and poor lifestyle, stigma
and negative stereotypical view
■■ Patients do not adhere to pharmacological treatment
Physician barriers
■■ Many primary care staff lack knowledge and
understanding about the causes and consequences
of gout
■■ Gout is not always managed as a long-term chronic
disease but as an acute condition
■■ Patients are not educated or given information to
encourage lifestyle modification and adherence
to long-term urate-lowering therapy
■■ Clinical guidelines for gout are not known or utilized,
particularly in primary care
■■ Training and education in best practice for the
treatment and management of gout is lacking at the
undergraduate and postgraduate level
■■ Care providers lack incentives to improve treatment
and long-term management of gout in general practice
Adapted by permission from BMJ Publishing Group Limited.
Spencer, K. et al. Ann. Rheum. Dis. 71, 1490–1495 (2012).54
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
280  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
of the informed patient in decision-making is impor-
tant in any medical condition. This process is central
to the principles of health care and is considered a key
professional responsibility by the UK General Medical
Council.134
Fortunately, gout is a very easy disease to
explain—certainly easier than many other less-well-
understood musculoskeletal conditions—and in the
vast majority of cases the practitioner can be very posi-
tive when discussing management options and progno-
sis (Figure 7). Poor adherence is predominantly due to
lack of patient education by the doctor. As demonstrated
in a proof-of-­principle study,53
a package of care that
includes full patient education on gout and its treatment,
discussion of illness perceptions, and development of an
individual­ized management plan (that is, recommended
best practice) can result in excellent patient outcomes.
When given appropriate information (Box 1), 100% of
patients in this study opted for ULT and during 1 year
of follow-up adherence was excellent, with more than
90% achieving and maintaining the therapeutic target of
SUA level 360 μmol/l and beginning to show improve-
ments in quality of life.53
Although participants in this
study were first seen by a rheumatologist in a hospi­tal
set­ting (for approximately 1 h), subsequent follow-up
was carried out by a specially trained nurse, often in
the patient’s home. As is the case with other prevalent
chronic conditions (for example, diabetes, asthma), it is
likely that nurse led-care of gout could be successful and
cost-effective in a community setting; an RCT is currently
comparing nurse-led care to ongoing general practitioner
care over a 2‑year period in UK general practice.
Given the high prevalence of gout and its potentially
serious consequences, it is clearly important that health
professionals have appropriate training in the ‘best prac-
tice’ management of gout, to enable them to educate and
treat their patients appropriately. General practitioners in
the UK admit in general that incentives, especially finan-
cial incentives, are required for them to alter their inter-
est and practice,54
and this change could be facilitated if
gout became a designated condition within the Quality
and Outcomes Framework for general practice in the
UK—a scheme in the UK that offers financial incentives
to general practices for achieving specified medical targets
for common conditions. Should the growing evidence that
high SUA level is an independent risk factor for cardio-
vascular disease, stroke and CKD become accepted, and
should ULT become licenced for the treatment of hyper­
uricaemia per se, these developments would have a major
impact on the prevalence and incidence of gout. How­ever,
given the currently high prevalence of gout, the impact it
has on patient health and quality of life, andthe availability
of effective ‘curative’ treatment, it seems justified to argue
for appropriate education of practitioners to improve
standards of care on the basis of gout itself.
Conclusions
A number of established and effective treatments exist
for managing gout, with new options on the horizon.
Despite this, gout care is often suboptimal and it is
apparent that just having effective drugs is not sufficient
to effect a ‘cure’.131
Education of patients can empower
them to make lifestyle changes and ensure they under-
stand the importance of adherence to long-term ULT. In
conjunction with this, health-care professionals need to
be aware of current best practice and how to up-titrate
ULT against a specific target level of SUA, but more
importantly they need to know how to address illness
perceptions and educate their patients appropriately, so
that individualized management plans can be developed
on the basis of shared decision-making. Current trends
are towards much earlier consideration of ULT, increas-
ingly towards the time of first diagnosis, and although
still controversial the arguments to treat elevated SUA
for cardiovascular and renal disease rather than just gout
are becoming stronger and have been accepted in some
countries including Japan. Although gout is relatively
well-understood, further research is required into various
aspects including the optimum level of SUA for long-term
manage­ment and, most importantly, how current stand-
ards of care can be improved using existing knowledge
and effective treatments.
Figure 7 | A full and holistic assessment followed by
individualized patient education is core to the management
of people with gout. In this instance, a specialist nurse
practitioner is assessing a patient and her explanation of
his chronic tophaceous gout, its treatment options, and
the likely benefits he can expect from ULT will be backed
up by focused patient literature. Written consent for
publication was obtained from the patient and nurse.
Image provided courtesy of M. Doherty.
Review criteria
Articles for this Review were found by systematic review
of the following databases: MEDLINE (1946–present),
Embase (1974–present), PubMed (from inception to
present), Cochrane Controlled Trials Register (from
inception to present), ISI Web of Science and AMED
(1985–present). The search strategy used terms for
gout, including “gouty arthritis”, “podagra”, “tophi”,
“monosodium urate crystals” and “hyperuricaemia”, in
combination with terms for studies or trials, including
“RCTs”, “cohort studies”, “case–control studies”
and “systematic reviews”. Preliminary searches were
undertaken in March 2012, and were updated July 2012.
Subsequent relevant articles were added as published.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  281
1.	 Kuo, C. F., Grainge, M. J., Mallen, C., Zhang, W.
 Doherty, M. Rising burden of gout in the UK
but continuing suboptimal management:
a nationwide population study. Ann. Rheum. Dis.
http://dx.doi.org/10.1136/annrheumdis‑
2013‑204463.
2.	 Zhu, Y., Pandya, B. J.  Choi, H. K. Prevalence of
gout and hyperuricemia in the US general
population: the National Health and Nutrition
Examination Survey 2007–2008. Arthritis
Rheum. 63, 3136–3141 (2011).
3.	 Kuo, C. F. et al. Risk of myocardial infarction
among patients with gout: a nationwide
population-based study. Rheumatology (Oxford)
52, 111–117 (2013).
4.	 Roddy, E., Zhang, W.  Doherty, M. Are joints
affected by gout also affected by osteoarthritis?
Ann. Rheum. Dis. 66, 1374–1377 (2007).
5.	 Palmer, T. M. et al. Association of plasma uric
acid with ischaemic heart disease and blood
pressure: Mendelian randomisation analysis of
two large cohorts. BMJ 347, f4262 (2013).
6.	 Stack, A. G., Independent and conjoint
associations of gout and hyperuricaemia with
total and cardiovascular mortality. QJM 106,
647–658 (2013).
7.	 Holme, I., Aastveit, A. H., Hammar, N., Jungner, I.
 Walldius, G. Uric acid and risk of myocardial
infarction, stroke and congestive heart failure in
417,734 men and women in the Apolipoprotein
MOrtality RISk study (AMORIS). J. Intern. Med.
266, 558–570 (2009).
8.	 Jolly, S. E. et al. Uric acid, hypertension, and
chronic kidney disease among Alaska Eskimos:
the Genetics of Coronary Artery Disease in
Alaska Natives (GOCADAN) study. J. Clin.
Hypertens. 14, 71–77 (2012).
9.	 Gnanenthiran, S. R., Hassett, G. M.,
Gibson, K. A.  McNeil, H. P. Acute gout
management during hospitalization: a need for a
protocol. Intern. Med. J. 41, 610–617 (2011).
10.	 Roddy, E., Mallen, C. D., Hider, S. L. 
Jordan, K. P. Prescription and comorbidity
screening following consultation for acute gout
in primary care. Rheumatology (Oxford) 49,
105–111 (2010).
11.	 Zhang, W. et al. EULAR evidence based
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. 65, 1312–1324 (2006).
12.	 Jordan, K. et al. British Society for Rheumatology
and British Health Professionals in
Rheumatology guideline for the management of
gout. Rheumatology (Oxford) 46, 1372–1374
(2007).
13.	 Khanna, D. et al. 2012 American College of
Rheumatology guidelines for management of
gout. Part 1: systematic nonpharmacologic and
pharmacologic therapeutic approaches to
hyperuricemia. Arthritis Care Res. (Hoboken) 64,
1431–1446 (2012).
14.	 Khanna, D. et al. 2012 American College of
Rheumatology guidelines for management of
gout. Part 2: therapy and antiinflammatory
prophylaxis of acute gouty arthritis. Arthritis Care
Res. (Hoboken) 64, 1447–1461 (2012).
15.	 Mikuls, T. R. et al. Gout epidemiology: results
from the UK General Practice Research
Database, 1990–1999. Ann Rheum Dis. 64,
267–272 (2005).
16.	 Garcia de la Torre, I. Double-blind parallel study
comparing tenoxicam and placebo in acute gouty
arthritis [Spanish]. Invet. Med. Int. 14, 92–97
(1987).
17.	 Altman, R. D., Honig, S., Levin, J. M. 
Lightfoot, R. W. Ketoprofen versus indomethacin
in patients with acute gouty arthritis:
a multicenter, double blind comparative study.
J. Rheumatol. 15, 1422–1426 (1988).
18.	 Butler, R. C. et al. Double-blind trial of
flurbiprofen and phenylbutazone in acute gouty
arthritis. Br. J. Clin. Pharmacol. 20, 511–513
(1985).
19.	 Shrestha, M. et al. Randomized double-blind
comparison of the analgesic efficacy of
intramuscular ketorolac and oral indomethacin
in the treatment of acute gouty arthritis. Ann.
Emerg. Med. 26, 682–686 (1995).
20.	 Lederman, R. A double-blind comparison of
etodolac and high doses of naproxen in the
treatment of acute gout. Adv.Ther. 7, 344–354
(1990).
21.	 Maccagno, A., Di Giorgio, E.  Romanowicz, A.
Effectiveness of etodolac (‘Lodine’) compared
with naproxen in patients with acute gout. Curr.
Med. Res. Opin. 12, 423–429 (1991).
22.	 National Collaborating Centre for Chronic
Conditions. Osteoarthritis: national clinical
guideline for care and management in adults
(Royal College of Physicians, London, 2008).
23.	 Brater, D. C. Anti-inflammatory agents and renal
function. Semin.Arthritis Rheum. 32 (Suppl. 1),
33–42 (2002).
24.	 Rubin, B. R. et al. Efficacy and safety profile of
treatment with etoricoxib 120 mg once daily
compared with indomethacin 50 mg three times
daily in acute gout: a randomized controlled trial.
Arthritis Rheum. 50, 598–606 (2004).
25.	 Schumacher, H. R. Jr et al. Randomised double
blind trial of etoricoxib and indometacin in
treatment of acute gouty arthritis. BMJ 324,
1488–1492 (2002).
26.	 Chen, L.  Ashcroft, D. Risk of myocardial
infarction associated with selective COX‑2
inhibitors: meta-analysis of randomised
controlled trials. Pharmacoepidemiol. Drug Saf.
16, 762–772 (2007).
27.	 Terkeltaub, R. A. Colchicine update: 2008.
Semin.Arthritis Rheum. 38, 411–419 (2009).
28.	 Ahern, M. J. et al. Does colchicine work? The
results of the first controlled study in acute gout.
Aust. NZ J. Med. 17, 301–304 (1987).
29.	 Terkeltaub, R. A. et al. High versus low dosing of
oral colchicine for early acute gout flare:
twenty‑four‑hour outcome of the first multicenter,
randomized, double-blind, placebo-controlled,
parallel-group, dose-comparison colchicine
study. Arthritis Rheum. 62, 1060–1068 (2010).
30.	 Terkeltaub, R. A. et al. Novel evidence-based
colchicine dose-reduction algorithm to predict
and prevent colchicine toxicity in the presence of
cytochrome P450 3A4/P-glycoprotein inhibitors.
Arthritis Rheum. 63, 2226–2237 (2011).
31.	 Clive, D. M. Renal transplant-associated
hyperuricemia and gout. J.Am. Soc. Nephrol. 11,
974–979 (2000).
32.	 US Food and Drug Administration. Information
for healthcare professionals: new safety
information for colchicine (marketed as Colcrys)
[online], http://www.fda.gov/Drugs/DrugSafety/
PostmarketDrugSafetyInformationforPatientsand
Providers/DrugSafetyInformationforHeathcare
Professionals/ucm174315.htm (2009).
33.	 Fernández, C., Noguera, R., González, J. A. 
Pascual, E. Treatment of acute attacks of gout
with a small dose of intraarticular triamcinolone
acetonide. J. Rheumatol. 26, 2285–2286
(1999).
34.	 Courtney, P.  Doherty, M. Joint aspiration and
injection and synovial fluid analysis. Best Pract.
Res. Clin. Rheumatol. 23, 161–192 (2009).
35.	 Janssens, H. J., Janssen, M., van de
Lisdonk, E. H., van Riel, P. L.  van Weel, C.
Use of oral prednisolone or naproxen for the
treatment of gout arthritis: a double-blind,
randomised equivalence trial. Lancet 371,
1854–1860 (2008).
36.	 Man, C. Y., Cheung, I. T., Cameron, P. A. 
Rainer, T. H. Comparison of oral prednisolone/
paracetamol and oral indomethacin/
paracetamol combination therapy in the
treatment of acute goutlike arthritis: a double-
blind, randomized, controlled trial. Ann. Emerg.
Med. 49, 670–677 (2007).
37.	 Alloway, J. A., Moriarty, M. J., Hoogland, Y. T.
 Nashel, D. J. Comparison of triamcinolone
acetonide with indomethacin in the treatment of
acute gouty arthritis. J. Rheumatol. 20, 111–113
(1993).
38.	 Choy, E. H., Kingsley, G. H., Khoshaba, B.,
Pipitone, N.  Scott, D. L. Intramuscular
Methylprednisolone Study Group. A two year
randomised controlled trial of intramuscular
depot steroids in patients with established
rheumatoid arthritis who have shown an
incomplete response to disease modifying
antirheumatic drugs. Ann. Rheum. Dis. 64,
1288–1293 (2005).
39.	 So, A., De Smedt, T., Revaz, S.  Tschopp, J.
A pilot study of IL‑1 inhibition by anakinra in
acute gout. Arthritis Res.Ther. 9, R28 (2007).
40.	 Chen, K., Fields, T., Mancuso, C. A., Bass, A. R.
 Vasanth, L. Anakinra’s efficacy is variable in
refractory gout: report of ten cases. Semin.
Arthritis Rheum. 40, 210–214 (2010).
41.	 Schlesinger, N. et al. Canakinumab relieves
symptoms of acute flares and improves
health-related quality of life in patients with
difficult‑to‑treat gouty arthritis by suppressing
inflammation: results of a randomized, dose-
ranging study.Arthritis Res.Ther. 13, R53
(2011).
42.	 So, A. et al. Canakinumab for the treatment of
acute flares in difficult‑to‑treat gouty arthritis:
results of a multicenter, phase II, dose-ranging
study. Arthritis Rheum. 62, 3064–3076 (2010).
43.	 Schlesinger, N. et al. Canakinumab for acute
gouty arthritis in patients with limited treatment
options: results from two randomised,
multicentre, active-controlled, double-blind trials
and their initial extensions. Ann. Rheum. Dis. 71,
1839–1848 (2012).
44.	 Terkeltaub, R. et al. The interleukin 1 inhibitor
rilonacept in treatment of chronic gouty arthritis:
results of a placebo-controlled, monosequence
crossover, non-randomised, single-blind pilot
study. Ann. Rheum. Dis. 68, 1613–1617 (2009).
45.	 Schumacher, H. R. Jr et al. Rilonacept
(interleukin‑1 Trap) in the prevention of acute
gout flares during initiation of urate-lowering
therapy: results of a phase II randomized,
double-blind, placebo-controlled trial. Arthritis
Rheum. 64, 876–884 (2012).
46.	 Schumacher, H. R. Jr et al. Rilonacept
(interleukin‑1 Trap) for prevention of gout flares
during initiation of uric acid-lowering therapy:
results from a phase III randomized, double-
blind, placebo-controlled, confirmatory efficacy
study. Arthritis Care Res. 64, 1462–1470
(2012).
47.	 Schlesinger, N. et al. Local ice therapy during
bouts of acute gouty arthritis. J. Rheumatol. 29,
331–334 (2002).
48.	 Gambaro, G.  Perazella, M. A. Adverse renal
effects of anti-inflammatory agents: evaluation
of selective and nonselective cyclooxygenase
inhibitors. J. Intern. Med. 253, 643–652 (2003).
49.	 Stamp, L. K.  Jordan, S. The challenges of gout
management in the elderly. Drugs Aging 28,
591–603 (2011).
50.	 Cattermole, G. N., Man, C. Y., Cheng, C. H.,
Graham, C. A.  Rainer, T. H. Oral prednisolone
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
282  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum
is more cost-effective than oral indomethacin for
treating patients with acute gout-like arthritis.
Eur. J. Emerg. Med. 16, 261–266 (2009).
51.	 Wertheimer, A. I., Davis, M. W.  Lauterio, T. J.
A new perspective on the pharmacoeconomics
of colchicine. Curr. Med. Res. Opin. 27, 931–937
(2011).
52.	 Arthritis Research UK. Gout [online], http://
www.arthritisresearchuk.org/arthritis-
information/conditions/gout.aspx (2013).
53.	 Rees, F., Jenkins, W.  Doherty, M. Patients with
gout adhere to curative treatment if informed
appropriately: proof‑of‑concept observational
study. Ann. Rheum. Dis.72, 826–830 (2013).
54.	 Spencer, K., Carr, A.  Doherty, M. Patient and
provider barriers to effective management of
gout in general practice: a qualitative study.
Ann. Rheum. Dis. 71, 1490–1495 (2012).
55.	 Dalbeth, N. et al. Illness perceptions in patients
with gout and the relationship with progression
of musculoskeletal disability. Arthritis Care Res.
(Hoboken) 63, 1605–1612 (2011).
56.	 Lindsay, K., Gow, P., Vanderpyl, J., Logo, P. 
Dalbeth, N. The experience and impact of living
with gout: a study of men with chronic gout using
a qualitative grounded theory approach. J. Clin.
Rheumatol. 17, 1–6 (2011).
57.	 Harrold, L. R. et al. Patients’ knowledge and
beliefs concerning gout and its treatment:
a population based study. BMC Musculoskelet.
Disord. 13, 180 (2012).
58.	 Choi, H. K., Atkinson, K., Karlson, E. W. 
Curhan, G. Obesity, weight change, hypertension,
diuretic use, and risk of gout in men: the Health
Professionals Follow-up Study. Arch. Intern. Med.
165, 742–748 (2005).
59.	 Choi, H. K., Atkinson, K., Karlson, E. W.,
Willett, W.  Curhan, G. Alcohol intake and risk
of incident gout in men: a prospective study.
Lancet. 363, 1277–1281 (2004).
60.	 Choi, H. K., Atkinson, K., Karlson, E. W.,
Willett, W.  Curhan, G. Purine-rich foods, dairy
and protein intake, and the risk of gout in men.
N. Engl. J. Med. 350, 1093–1103 (2004).
61.	 Choi, H. K.  Curhan, G. Soft drinks, fructose
consumption, and the risk of gout in men:
prospective cohort study. BMJ 336, 309–312
(2008).
62.	 Choi, W. J., Ford, E. S., Curhan, G., Rankin, J. I. 
Choi, H. K. Independent association of serum
retinol and β‑carotene levels with hyperuricemia:
a national population study. Arthritis Care Res.
(Hoboken) 64, 389–396 (2012).
63.	 Lyngdoh, T. et al. Serum uric acid and adiposity:
deciphering causality using a bidirectional
Mendelian randomization approach. PLoS ONE
7, e39321 (2012).
64.	 Joosten, L. A. B. et al. Engagement of fatty acids
with Toll-like receptor 2 drives interleukin‑1
production via the ASC/caspase 1 pathway in
monosodium urate monohydrate crystal-induced
gouty arthritis. Arthritis Rheum. 62, 3237–3248
(2010).
65.	 Dalbeth, N. et al. Identification of dairy fractions
with antiinflammatory properties in models of
acute gout. Ann. Rheum. Dis. 69, 766–769
(2010).
66.	 Dalbeth, N. et al. Effects of skim milk powder
enriched with glycomacropeptide and G600 milk
fat extract on frequency of gout flares:
a proof‑of‑concept randomised controlled trial.
Ann. Rheum. Dis. 71, 929–934 (2012).
67.	 Zhang, Y. et al. Cherry consumption and
decreased risk of recurrent gout attacks.
Arthritis Rheum. 64, 4004–4011 (2012).
68.	 Choi, H. K., Soriano, L. C., Zhang, Y. 
Rodríguez, L. A. G. Antihypertensive drugs
and risk of incident gout among patients with
hypertension: population based case-control
study. BMJ 344, d8190 (2012).
69.	 Kaya, E. B. et al. Serum uric acid levels predict
the severity and morphology of coronary
atherosclerosis detected by multidetector
computed tomography. Atherosclerosis 213,
178–183 (2010).
70.	 Grayson, P. C., Kim, S. Y., Lavalley, M. 
Choi, H. K. Hyperuricemia and incident
hypertension: a systematic review and meta-
analysis. Arthritis Care Res. (Hoboken) 63,
102–110 (2011).
71.	 Numata, T., Miyatake, N., Wada, J.  Makino, H.
Comparison of serum uric acid levels between
Japanese with and without metabolic
syndrome. Diabetes Res. Clin. Pract. 80,
e1–e5 (2008).
72.	 Li, C., Hsieh, M. C.  Chang, S. J. Metabolic
syndrome, diabetes, and hyperuricaemia. Curr.
Opin. Rheumatol. 25, 210–216 (2013).
73.	 Stamp, L. K.  Chapman, P. T. Gout and its
comorbidities: implications for therapy.
Rheumatology (Oxford) 52, 34–44 (2013).
74.	 Perez-Ruiz, F. Treating to target: a strategy to
cure gout. Rheumatology (Oxford) 48 (Suppl. 2),
ii9-ii14 (2009).
75.	 Perez-Ruiz, F., Calabozo, M., Pijoan, J. I., Herrero-
Beites, A. M.  Ruibal, A. Effect of urate-lowering
therapy on the velocity of size reduction of tophi
in chronic gout. Arthritis Rheum. 47, 356–360
(2002).
76.	 De Miguel, E. et al. Diagnosis of gout in patients
with asymptomatic hyperuricaemia: a pilot
ultrasound study. Ann. Rheum. Dis. 71, 157–158
(2012).
77.	 Pineda, C. et al. Joint and tendon subclinical
involvement suggestive of gouty arthritis in
asymptomatic hyperuricemia: an ultrasound
controlled study. Arthritis Res.Ther. 13, R4
(2011).
78.	 Choi, H. K. et al. Dual energy CT in gout:
a prospective validation study.Ann. Rheum. Dis.
71, 1466–1471 (2012).
79.	 Pascual, E. Persistence of monosodium urate
crystals and low grade inflammation in the
synovial fluid of untreated gout. Arthritis Rheum.
34, 141–145 (1991).
80.	 Taylor, T., Mecchella, J., Larson, R., Kerin, K. 
MacKenzie, T. Initiation of allopurinol at first
medical contact for acute attacks of gout:
a randomized clinical trial. Am. J. Med. 125,
1126–1134 (2012).
81.	 Reinders, M. K. et al. A randomised controlled
trial on the efficacy and tolerability with dose
escalation of allopurinol 300–600 mg/day
versus benzbromarone 100–200 mg/day in
patients with gout. Ann. Rheum. Dis. 68,
892–897 (2009).
82.	 Kuzell, W. C., Seebach, L. M., Glover, R. P. 
Jackman, A. E. Treatment of gout with allopurinol
and sulphinpyrazone in combination and with
allopurinol alone. Ann. Rheum. Dis. 25, 634–642
(1966).
83.	 Bull, P. W.  Scott, J. T. Intermittent control of
hyperuricemia in the treatment of gout.
J. Rheumatol. 16, 1246–1248 (1989).
84.	 Schumacher, H. R. Jr et al. Effects 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 Rheum. 59,
1540–1548 (2008).
85.	 Becker, M. A. et al. Febuxostat compared with
allopurinol in patients with hyperuricemia and
gout. N. Engl. J. Med. 353, 2450–2461 (2005).
86.	 Perez-Ruiz, F., Hernando, I., Villar, I.  Nolla, J. M.
Correction of allopurinol dosing should be based
on clearance of creatinine, but not plasma
creatinine levels: another insight to allopurinol-
related toxicity. J. Clin. Rheumatol. 11, 129–133
(2005).
87.	 Vazquez-Mellado, J., Morales, E. M., Pacheco-
Tena, C.  Burgos-Vargas, R. Relation between
adverse events associated with allopurinol and
renal function in patients with gout. Ann. Rheum.
Dis. 60, 981–983 (2001).
88.	 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. 33, 1646–1650 (2006).
89.	 Stamp, L. K. et al. Using allopurinol above the
dose based on creatinine clearance is effective
and safe in patients with chronic gout, including
those with renal impairment. Arthritis Rheum.
63, 412–421 (2011).
90.	 Kim, S. C., Newcomb, C., Margolis, D., Roy, J.
 Hennessy, S. Severe cutaneous reactions
requiring hospitalization in allopurinol initiators:
a population-based cohort study. Arthritis Care
Res. (Hoboken) 65, 578–584 (2013).
91.	 Yeo, S. I. HLA‑B*5801: utility and cost-
effectiveness in the Asia-Pacific Region. Int. J.
Rheum. Dis. 16, 254–257 (2013).
92.	 Stamp, L. K. et al. Starting dose is a risk factor
for allopurinol hypersensitivity syndrome:
a proposed safe starting dose of allopurinol.
Arthritis Rheum. 64, 2529–2536 (2012).
93.	 Doehner, W. et al. Effects of xanthine oxidase
inhibition with allopurinol on endothelial function
and peripheral blood flow in hyperuricemic
patients with chronic heart failure: results from
2 placebo-controlled studies. Circulation 105,
2619–2624 (2002).
94.	 Noman, A., Ang, D., Ogston, S., Lang, C. 
Struthers, A. Effect of high-dose allopurinol on
exercise in patients with chronic stable angina:
a randomised, placebo controlled crossover trial.
Lancet 375, 2161–2167 (2010).
95.	 Goicoechea, M. et al. Effect of allopurinol in
chronic kidney disease progression and
cardiovascular risk. Clin. J.Am. Soc. Nephrol. 5,
1388–1393 (2010).
96.	 Momeni, A., Shahidi, S., Seirafian, S., Taheri, S.
 Kheiri, S. Effect of allopurinol in decreasing
proteinuria in type 2 diabetic patients. Iran. J.
Kidney Dis. 4, 128–132 (2010).
97.	 Neogi, T. et al. Are either or both hyperuricemia
and xanthine oxidase directly toxic to the
vasculature? A critical appraisal. Arthritis
Rheum. 64, 327–338 (2012).
98.	 Becker, M. A. 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
efficacy in patients with gout. Arthritis Rheum.
52, 916–923 (2005).
99.	 Becker, M. A. et al. The urate-lowering efficacy
and safety of febuxostat in the treatment of the
hyperuricemia of gout: the CONFIRMS trial.
Arthritis Res.Ther. 12, R63 (2010).
100.	European Medicines Agency. Summary of
product characteristics: adenuric film-coated
tablets [online], http://www.medicines.org.uk/
emc/medicine/22830/SPC (2012).
101.	Abeles, A. M. Febuxostat hypersensitivity.
J. Rheumatol. 39, 659 (2012).
102.	Chohan, S. Safety and efficacy of febuxostat
treatment in subjects with gout and severe
allopurinol adverse reactions. J. Rheumatol. 38,
1957–1959 (2011).
103.	National Institute for Health and Care
Excellence. Febuxostat for the management
of hyperuricaemia in people with gout. NICE
technology appraisal guidance 164 [online], 
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS | RHEUMATOLOGY 	 VOLUME 10  |  MAY 2014  |  283
http://www.nice.org.uk/guidance/TA164
(2008).
104.	Ogino, K. et al. Uric acid-lowering treatment with
benzbromarone in patients with heart failure:
a double-blind placebo-controlled crossover
preliminary study. Circ. Heart Fail. 3, 73–81
(2010).
105.	Reinders, M. K. et al. Efficacy and tolerability of
urate-lowering drugs in gout: a randomised
controlled trial of benzbromarone versus
probenecid after failure of allopurinol. Ann.
Rheum. Dis. 68, 51–56 (2009).
106.	Scott, J. T. Comparison of allopurinol and
probenecid. Ann. Rheum. Dis. 25, 623–626
(1966).
107.	Reinders, M. K., van Roon, E. N., Houtman, P. M.,
Brouwers, J. R.  Jansen, T. L. Biochemical
effectiveness of allopurinol and allopurinol-
probenecid in previously benzbromarone-treated
gout patients. Clin. Rheumatol. 26, 1459–1465
(2007).
108.	Lee, M. H., Graham, G. G., Williams, K. M.
 Day, R. O. A benefit-risk assessment of
benzbromarone in the treatment of gout. Was its
withdrawal from the market in the best interest
of patients? Drug Saf. 31, 643–665 (2008).
109.	Zhu, X. et al. Efficacy and safety of losartan
in treatment of hyperuricemia and
posttransplantation erythrocytosis: results of a
prospective, open, randomized, case–control
study. Transplant. Proc. 41, 3736–3742 (2009).
110.	Huang, H. Y. et al. The effects of vitamin C
supplementation on serum concentrations of
uric acid: results of a randomized controlled
trial. Arthritis Rheum. 52, 1843–1847 (2005).
111.	Juraschek, S. P., Miller, E. R. 3rd
 Gelber, A. C.
Effect of oral vitamin C supplementation on
serum uric acid: a meta-analysis of randomized
controlled trials. Arthritis Care Res. (Hoboken)
63, 1295–1306 (2011).
112.	Bastow, M. D., Durrington, P. N.  Ishola, M.
Hypertriglyceridemia and hyperuricemia: effects
of two fibric acid derivatives (bezafibrate and
fenofibrate) in a double-blind, placebo-controlled
trial. Metabolism 37, 217–220 (1988).
113.	Stamp, L. K. et al. Clinically insignificant effect of
supplemental vitamin C on serum urate in
patients with gout. A pilot randomized controlled
trial. Arthritis Rheum. 65, 1636–1642 (2013).
114.	Sundy, J. S. et al. Efficacy and tolerability of
pegloticase for the treatment of chronic gout in
patients refractory to conventional treatment:
two randomized controlled trials. JAMA 306,
711–720 (2011).
115.	Ganson, N. J., Kelly, S. J., Scarlett, E.,
Sundy, J. S.  Hershfield, M. S. 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. 8,
R12 (2006).
116.	Krystexxa®( pegloticase) prescribing
information. Savient Pharmaceuticals Inc.
[online], http://krystexxa.com/hcp/pdf/
KRYSTEXXA_Prescribing_Information.pdf (2014).
117.	Malaguarnera, M. et al. A single dose of
rasburicase in elderly patients with
hyperuricaemia reduces serum uric acid levels
and improves renal function. Expert Opin.
Pharmacother. 10, 737–742 (2009).
118.	De Angelis, S. et al. Is rasburicase an effective
alternative to allopurinol for management of
hyperuricemia in renal failure patients? A double
blind-randomized study. Eur. Rev. Med.
Pharmacol. Sci. 11, 179–184 (2007).
119.	Pui, C. H. et al. Recombinant urate oxidase for
the prophylaxis or treatment of hyperuricemia in
patients with leukemia or lymphoma. Clin. Oncol.
19, 697–704 (2001).
120.	Richette, P., Brière, C., Hoenen-Clavert, V.,
Loeuille, D.  Bardin, T. Rasburicase for
tophaceous gout not treatable with allopurinol:
an exploratory study. J. Rheumatol. 34,
2093–2098 (2007).
121.	Lasko, B. et al. RDEA594, a novel uricosuric
agent, significantly reduced serum urate levels
and was well tolerated in a phase 2a pilot study
in hyperuricemic gout patients [abstract].
Arthritis Rheum. 60 (Suppl. 10), 1105 (2009).
122.	Becker, M. et al. BCX4208 combined with
allopurinol increases response rates in patients
with gout who fail to reach goal range serum
urate on allopurinol alone: a randomized, double-
blind, placebo-controlled trial [L10]. Presented at
the 2011 ACR/ARHP Annual Scientific Meeting.
123.	Li-Yu, J. et al. Treatment of chronic gout. Can we
determine when urate stores are depleted
enough to prevent attacks of gout? J. Rheumatol.
28, 577–580 (2001).
124.	de Oliveira, E. P.  Burini, R. C. High plasma uric
acid concentration: causes and consequences.
Diabetol. Metab. Syndr. 4, 12 (2012).
125.	Perez-Ruiz, F., Herrero-Beites, A. M. 
Carmona, L. A two-stage approach to the
treatment of hyperuricemia in gout: the “dirty
dish” hypothesis. Arthritis Rheum. 63,
4002–4006 (2011).
126.	Borstad, G. C. et al. Colchicine for prophylaxis
of acute flares when initiating allopurinol for
chronic gouty arthritis. J. Rheumatol. 31,
2429–2432 (2004).
127.	Wortmann, R., MacDonald, P., Hunt, B. 
Jackson, R. Effect of prophylaxis on gout flares
after the initiation of urate-lowering therapy:
analysis of data from three phase III trials. Clin.
Ther. 32, 2386–2397 (2010).
128.	Mikuls, T. R., Farrar, J. T., Bilker, W. B.,
Fernandes, S.  Saag, K. G. Suboptimal
physician adherence to quality indicators for the
management of gout and asymptomatic
hyperuricaemia: results from the UK General
Practice Research Database (GPRD).
Rheumatology (Oxford) 44, 1038–1042 (2005).
129.	Roddy, E., Zhang, W.  Doherty, M. Concordance
of the management of chronic gout in a UK
primary-care population with the EULAR gout
recommendations. Ann. Rheum. Dis. 66,
1311–1315 (2007).
130.	Doherty, M. et al. Gout: why is this curable
disease so seldom cured? Ann. Rheum. Dis. 71,
1765–1770 (2012).
131.	Lioté, F.  Choi, H. Managing gout needs more
than drugs: ‘Il faut le savoir-faire, l’Art et la
manière’. Ann. Rheum. Dis. 72, 791–793
(2013).
132.	Harrold, L. R. et al. Primary care providers’
knowledge, beliefs and treatment practices for
gout: results of a physician questionnaire.
Rheumatology (Oxford) 52, 1623–1629 (2013).
133.	Owens, D., Whelan, B.  McCarthy, G. A survey
of the management of gout in primary care.
Ir. Med. J. 101, 147–149 (2008).
134.	General Medical Council. Tomorrow’s doctors:
outcomes and standards for undergraduate
medical education (General Medical Council,
London, 2009).
135. British National Formulary. BNF [online],
http://www.bnf.org/bnf/index.htm (2014).
Author contributions
All authors made substantial contributions to
discussions of content, writing, and reviewing/editing
the manuscript before submission.
REVIEWS
© 2014 Macmillan Publishers Limited. All rights reserved

Contenu connexe

Tendances

Slide deck 2018_updated_eular_recommendations_management_hand_oa
Slide deck 2018_updated_eular_recommendations_management_hand_oaSlide deck 2018_updated_eular_recommendations_management_hand_oa
Slide deck 2018_updated_eular_recommendations_management_hand_oaAnwar Sholeh Manik
 
Efficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia
Efficacy of Helicobacter pylori Eradication Therapy on Functional DyspepsiaEfficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia
Efficacy of Helicobacter pylori Eradication Therapy on Functional DyspepsiaDLambertus
 
Lec levels of prevention intervention 2021-22
Lec levels of prevention intervention 2021-22Lec levels of prevention intervention 2021-22
Lec levels of prevention intervention 2021-22Tauseef Jawaid
 
Low Back Pain Clinical Practice Guideline
Low Back Pain Clinical Practice GuidelineLow Back Pain Clinical Practice Guideline
Low Back Pain Clinical Practice GuidelineMr. Saeed Al-Amri
 
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricas
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricasBronquiolitis: estudio variabilidad manejo en urgencias pediatricas
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricasJavier González de Dios
 
Experimental study in epidemiology methods ppt
Experimental  study in epidemiology methods pptExperimental  study in epidemiology methods ppt
Experimental study in epidemiology methods pptanjalatchi
 
552_rachel_bowe_Project Proposal Draft Final
552_rachel_bowe_Project Proposal Draft Final552_rachel_bowe_Project Proposal Draft Final
552_rachel_bowe_Project Proposal Draft FinalRachel Bowe
 
Concepts of Screening for disease
Concepts of Screening for diseaseConcepts of Screening for disease
Concepts of Screening for diseaseMohan Jangwal
 
Disease prevention strategies: Primary
Disease prevention strategies: PrimaryDisease prevention strategies: Primary
Disease prevention strategies: PrimaryTanveerRehman4
 
Time Out Project, Woodhull Hospital
Time Out Project, Woodhull HospitalTime Out Project, Woodhull Hospital
Time Out Project, Woodhull Hospitalminan05
 
Arthur Vinnitsky Poster 4 [Autosaved]
Arthur Vinnitsky Poster 4 [Autosaved]Arthur Vinnitsky Poster 4 [Autosaved]
Arthur Vinnitsky Poster 4 [Autosaved]Arthur Vinnitsky
 
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...Nutricia
 
Prevention and control of diseases for third year mbbs students
Prevention and control of diseases for third year mbbs studentsPrevention and control of diseases for third year mbbs students
Prevention and control of diseases for third year mbbs studentswrigveda
 
disease prevention and control
disease prevention and controldisease prevention and control
disease prevention and controlPreetika Maurya
 

Tendances (19)

perforation
perforationperforation
perforation
 
Dr. Born CV
Dr. Born CVDr. Born CV
Dr. Born CV
 
Slide deck 2018_updated_eular_recommendations_management_hand_oa
Slide deck 2018_updated_eular_recommendations_management_hand_oaSlide deck 2018_updated_eular_recommendations_management_hand_oa
Slide deck 2018_updated_eular_recommendations_management_hand_oa
 
Efficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia
Efficacy of Helicobacter pylori Eradication Therapy on Functional DyspepsiaEfficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia
Efficacy of Helicobacter pylori Eradication Therapy on Functional Dyspepsia
 
Lec levels of prevention intervention 2021-22
Lec levels of prevention intervention 2021-22Lec levels of prevention intervention 2021-22
Lec levels of prevention intervention 2021-22
 
Low Back Pain Clinical Practice Guideline
Low Back Pain Clinical Practice GuidelineLow Back Pain Clinical Practice Guideline
Low Back Pain Clinical Practice Guideline
 
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricas
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricasBronquiolitis: estudio variabilidad manejo en urgencias pediatricas
Bronquiolitis: estudio variabilidad manejo en urgencias pediatricas
 
Experimental study in epidemiology methods ppt
Experimental  study in epidemiology methods pptExperimental  study in epidemiology methods ppt
Experimental study in epidemiology methods ppt
 
CONCEPT OF PREVENTION
CONCEPT OF PREVENTIONCONCEPT OF PREVENTION
CONCEPT OF PREVENTION
 
552_rachel_bowe_Project Proposal Draft Final
552_rachel_bowe_Project Proposal Draft Final552_rachel_bowe_Project Proposal Draft Final
552_rachel_bowe_Project Proposal Draft Final
 
Concepts of Screening for disease
Concepts of Screening for diseaseConcepts of Screening for disease
Concepts of Screening for disease
 
Concept of control
Concept of controlConcept of control
Concept of control
 
20150300.0 00030
20150300.0 0003020150300.0 00030
20150300.0 00030
 
Disease prevention strategies: Primary
Disease prevention strategies: PrimaryDisease prevention strategies: Primary
Disease prevention strategies: Primary
 
Time Out Project, Woodhull Hospital
Time Out Project, Woodhull HospitalTime Out Project, Woodhull Hospital
Time Out Project, Woodhull Hospital
 
Arthur Vinnitsky Poster 4 [Autosaved]
Arthur Vinnitsky Poster 4 [Autosaved]Arthur Vinnitsky Poster 4 [Autosaved]
Arthur Vinnitsky Poster 4 [Autosaved]
 
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...
Tolerability and Safety of Souvenaid in Patients with Mild Alzheimer’s Diseas...
 
Prevention and control of diseases for third year mbbs students
Prevention and control of diseases for third year mbbs studentsPrevention and control of diseases for third year mbbs students
Prevention and control of diseases for third year mbbs students
 
disease prevention and control
disease prevention and controldisease prevention and control
disease prevention and control
 

Similaire à Optimizing current treatment of gout nrrheum.2014.32

Surgical vs Conservative Management of Colonic Diverticulitis
Surgical vs Conservative Management of Colonic DiverticulitisSurgical vs Conservative Management of Colonic Diverticulitis
Surgical vs Conservative Management of Colonic DiverticulitisVedica Sethi
 
Stress ulcer prophylaxis.pdf
Stress ulcer prophylaxis.pdfStress ulcer prophylaxis.pdf
Stress ulcer prophylaxis.pdfDr-Marwa
 
2017 cpg secondary and primary prevention guideline (1)
2017 cpg secondary and primary prevention guideline (1)2017 cpg secondary and primary prevention guideline (1)
2017 cpg secondary and primary prevention guideline (1)Ibn Abdullah
 
8.1 Fistula Crohn000341961.pdf
8.1 Fistula Crohn000341961.pdf8.1 Fistula Crohn000341961.pdf
8.1 Fistula Crohn000341961.pdfssuser7764be
 
Gout ii acr 2012
Gout ii   acr 2012Gout ii   acr 2012
Gout ii acr 2012Hummd Mdhum
 
Abbreviated Laparotomy
Abbreviated LaparotomyAbbreviated Laparotomy
Abbreviated LaparotomyNHS
 
How to achieve deep remission in treatment of inflammatory bowel disease.
How to achieve deep remission in treatment of inflammatory bowel disease.How to achieve deep remission in treatment of inflammatory bowel disease.
How to achieve deep remission in treatment of inflammatory bowel disease.Younis I Munshi
 
Guias peritonitis
Guias peritonitisGuias peritonitis
Guias peritonitisEze Dzul
 
Healthcare Problem PresentationBy Vannelyn Oriel1
Healthcare Problem PresentationBy Vannelyn Oriel1Healthcare Problem PresentationBy Vannelyn Oriel1
Healthcare Problem PresentationBy Vannelyn Oriel1SusanaFurman449
 
Trauma y sangrado guias europeas
Trauma y sangrado guias europeasTrauma y sangrado guias europeas
Trauma y sangrado guias europeasLucia Tacanga
 
pancreatitis aguda .pdf
pancreatitis aguda .pdfpancreatitis aguda .pdf
pancreatitis aguda .pdfRoblesKR
 
abdominal sepsis
abdominal sepsisabdominal sepsis
abdominal sepsisSa Ju
 
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...ISPD peritonitis guideline recommendations 2022 update on prevention and trea...
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...Maria Martinez
 
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASE
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASERECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASE
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASEPARUL UNIVERSITY
 
cp119 juvenile-arthritis.pdf
cp119 juvenile-arthritis.pdfcp119 juvenile-arthritis.pdf
cp119 juvenile-arthritis.pdfshelladepari
 

Similaire à Optimizing current treatment of gout nrrheum.2014.32 (20)

Surgical vs Conservative Management of Colonic Diverticulitis
Surgical vs Conservative Management of Colonic DiverticulitisSurgical vs Conservative Management of Colonic Diverticulitis
Surgical vs Conservative Management of Colonic Diverticulitis
 
05 n141 16396
05 n141 1639605 n141 16396
05 n141 16396
 
Stress ulcer prophylaxis.pdf
Stress ulcer prophylaxis.pdfStress ulcer prophylaxis.pdf
Stress ulcer prophylaxis.pdf
 
Art end clasificacion
Art end clasificacionArt end clasificacion
Art end clasificacion
 
2017 cpg secondary and primary prevention guideline (1)
2017 cpg secondary and primary prevention guideline (1)2017 cpg secondary and primary prevention guideline (1)
2017 cpg secondary and primary prevention guideline (1)
 
Pulsatile dry cupping in patients with
Pulsatile dry cupping in patients withPulsatile dry cupping in patients with
Pulsatile dry cupping in patients with
 
8.1 Fistula Crohn000341961.pdf
8.1 Fistula Crohn000341961.pdf8.1 Fistula Crohn000341961.pdf
8.1 Fistula Crohn000341961.pdf
 
Gout ii acr 2012
Gout ii   acr 2012Gout ii   acr 2012
Gout ii acr 2012
 
Abbreviated Laparotomy
Abbreviated LaparotomyAbbreviated Laparotomy
Abbreviated Laparotomy
 
Damage control surgery for abdominal emergencies
Damage control surgery for abdominal emergenciesDamage control surgery for abdominal emergencies
Damage control surgery for abdominal emergencies
 
How to achieve deep remission in treatment of inflammatory bowel disease.
How to achieve deep remission in treatment of inflammatory bowel disease.How to achieve deep remission in treatment of inflammatory bowel disease.
How to achieve deep remission in treatment of inflammatory bowel disease.
 
Guias peritonitis
Guias peritonitisGuias peritonitis
Guias peritonitis
 
Healthcare Problem PresentationBy Vannelyn Oriel1
Healthcare Problem PresentationBy Vannelyn Oriel1Healthcare Problem PresentationBy Vannelyn Oriel1
Healthcare Problem PresentationBy Vannelyn Oriel1
 
19, 20 dm_dec_clin_donley
19, 20 dm_dec_clin_donley19, 20 dm_dec_clin_donley
19, 20 dm_dec_clin_donley
 
Trauma y sangrado guias europeas
Trauma y sangrado guias europeasTrauma y sangrado guias europeas
Trauma y sangrado guias europeas
 
pancreatitis aguda .pdf
pancreatitis aguda .pdfpancreatitis aguda .pdf
pancreatitis aguda .pdf
 
abdominal sepsis
abdominal sepsisabdominal sepsis
abdominal sepsis
 
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...ISPD peritonitis guideline recommendations 2022 update on prevention and trea...
ISPD peritonitis guideline recommendations 2022 update on prevention and trea...
 
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASE
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASERECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASE
RECENT ADVANCES IN THE MANAGEMENT OF INFLAMMATORY BOWEL DISEASE
 
cp119 juvenile-arthritis.pdf
cp119 juvenile-arthritis.pdfcp119 juvenile-arthritis.pdf
cp119 juvenile-arthritis.pdf
 

Plus de Elsa von Licy

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Elsa von Licy
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiElsa von Licy
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
NeuropsychophysiologieElsa von Licy
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)Elsa von Licy
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuroElsa von Licy
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01Elsa von Licy
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercicesElsa von Licy
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pElsa von Licy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsElsa von Licy
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesElsa von Licy
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiantsElsa von Licy
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficaceElsa von Licy
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introElsa von Licy
 

Plus de Elsa von Licy (20)

Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
Styles of Scientific Reasoning, Scientific Practices and Argument in Science ...
 
Strategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfiStrategie Decisions Incertitude Actes conference fnege xerfi
Strategie Decisions Incertitude Actes conference fnege xerfi
 
Rainville pierre
Rainville pierreRainville pierre
Rainville pierre
 
Neuropsychophysiologie
NeuropsychophysiologieNeuropsychophysiologie
Neuropsychophysiologie
 
L agressivite en psychanalyse (21 pages 184 ko)
L agressivite en psychanalyse (21 pages   184 ko)L agressivite en psychanalyse (21 pages   184 ko)
L agressivite en psychanalyse (21 pages 184 ko)
 
C1 clef pour_la_neuro
C1 clef pour_la_neuroC1 clef pour_la_neuro
C1 clef pour_la_neuro
 
Hemostase polycop
Hemostase polycopHemostase polycop
Hemostase polycop
 
Antiphilos
AntiphilosAntiphilos
Antiphilos
 
Vuillez jean philippe_p01
Vuillez jean philippe_p01Vuillez jean philippe_p01
Vuillez jean philippe_p01
 
Spr ue3.1 poly cours et exercices
Spr ue3.1   poly cours et exercicesSpr ue3.1   poly cours et exercices
Spr ue3.1 poly cours et exercices
 
Plan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 pPlan de cours all l1 l2l3m1m2 p
Plan de cours all l1 l2l3m1m2 p
 
M2 bmc2007 cours01
M2 bmc2007 cours01M2 bmc2007 cours01
M2 bmc2007 cours01
 
Feuilletage
FeuilletageFeuilletage
Feuilletage
 
Chapitre 1
Chapitre 1Chapitre 1
Chapitre 1
 
Biophy
BiophyBiophy
Biophy
 
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutionsBioph pharm 1an-viscosit-des_liquides_et_des_solutions
Bioph pharm 1an-viscosit-des_liquides_et_des_solutions
 
Poly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicalesPoly histologie-et-embryologie-medicales
Poly histologie-et-embryologie-medicales
 
Methodes travail etudiants
Methodes travail etudiantsMethodes travail etudiants
Methodes travail etudiants
 
Atelier.etude.efficace
Atelier.etude.efficaceAtelier.etude.efficace
Atelier.etude.efficace
 
There is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_introThere is no_such_thing_as_a_social_science_intro
There is no_such_thing_as_a_social_science_intro
 

Optimizing current treatment of gout nrrheum.2014.32

  • 1. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  271 Division of Academic Rheumatology, University of Nottingham, Clinical Sciences Building, City Hospital Nottingham, Hucknall Road, Nottingham NG5 1PB, UK (F.R., M.H., M.D.). Correspondence to: M.D. michael.doherty@ nottingham.ac.uk Optimizing current treatment of gout Frances Rees, Michelle Hui and Michael Doherty Abstract | Gout is the most common inflammatory arthritis worldwide. Although effective treatments exist to eliminate sodium urate crystals and to ‘cure’ the disease, the management of gout is often suboptimal. This article reviews available treatments, recommended best practice and barriers to effective care, and how these barriers might be overcome. To optimize the management of gout, health professionals need to know not only how to treat acute attacks but also how to up-titrate urate-lowering therapy against a specific target level of serum uric acid that is below the saturation point for crystal formation. Current perspectives are changing towards much earlier use of urate-lowering therapy, even at the time of first diagnosis of gout. Holistic assessment and patient education are essential to address patient-specific risk factors and ensuring adherence to individualized therapy. Shared decision-making between a fully informed patient and practitioner greatly increases the likelihood of curing gout. Rees, F. et al. Nat. Rev. Rheumatol. 10, 271–283 (2014); published online 11 March 2014; doi:10.1038/nrrheum.2014.32 Introduction Gout is the most common form of inflammatory arthri- tis, with crude estimates of adult prevalence of 2.5% in the UK,1 3.9% in the USA,2 and 3.8% in Taiwan.3 The prevalence and incidence of gout increase with age, and it is more common in men than women (approxi- mately 3:1).2 Fortunately, the pathogenesis of gout is relatively well understood. We know the causative agent—­monosodium urate crystals—and the multi- ple risk factors that lead to their formation, primar- ily through chronic elevation of uric acid levels above the saturation point at which urate crystals can form (Figure 1). Urate crystals are deposited preferentially in and around peripheral joints in the feet, knees, hands and elbows, especially those affected by osteoarthritis.4 The urate crystals predominantly form in the super­ficial layers of articular cartilage, in subchondral bone and in fibrous peri-articular tissues, but might also be evident clinically as subcutaneous tophi. During a prolonged asymptomatic phase (the duration of which is unknown but presumed to be measured in years), the concentra- tion of crystals gradually builds up and eventu­ally, once deposition is extensive, it is hypothesized that urate crystals ‘shed’ from the articular cartilage into the joint space and trigger an acute ‘attack’ of extremely painful joint inflammation. This acute attack is usually the reason patients first seek medical attention. However, if left untreated, continuing urate crystal deposition not only causes further acute attacks and involvement of additional joint sites, but also, and importantly, could cause eventual irreversible joint damage with chronic symptoms and disability. Apart from causing acute gout and chronic joint damage, there is increasing evidence, though still controversial,5 that a high serum uric acid (SUA) level is an independent risk factor for cardio­ vascular disease, stroke and chronic kidney disease (CKD).6–8 Therefore, there are several reasons to con- sider definitive treatment of elevated SUA levels once a patient is known to have gout. Fortunately, several treatment strategies exist to effectively reduce and maintain SUA levels below the saturation point, thus preventing further crystal for- mation and encouraging the dissolution of exist­ing crystals. Once the pathogenic urate crystals are elimi- nated, further acute attacks are impossible and the risk of further crystal-induced joint damage is removed. How­ever, although gout is the only form of chro­nic arthritis for which such a ‘cure’ exists, audits pub- lished in the past 4 years confirm that the treatment of gout both in primary care and hospital practice is far from optimal.1,9,10 This article reviews recommended available treat- ments, and highlights new perspectives relating to current standards of care and barriers that need to be addressed if gout is to be managed successfully. The Review largely reflects the latest EULAR11 and British Society for Rheumatology (BSR)12 guidelines, published in 2006 and 2007 respectively (both are currently being updated), as well as the more recent 2012 ACR guide- lines.13,14 In addition, we systematically searched the literature for individual treatments. Some drug options (including biologic agents and uricases) are not dis- cussed in detail because they are largely unlicenced for gout, are very expensive and have limited application. We first review the available options for managing acute attacks and for reducing SUA levels, and then focus on important barriers to care and how to successfully implement ‘curative’ management strategies. Competing interests M.D. has received honoraria for being a member of ad hoc advisory boards for Ardea Biosciences, Menarini, Novartis and Savient. F.R. and M.H. declare no competing interests. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 2. 272  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum Management of acute attacks Acute gout is recognized as one of the most painful experi­ences known, on the same level as childbirth or visceral colic. Therefore, treatment of an attack should be initiated as soon as possible following initial diagno- sis or, in a patient with known gout, as soon as the first warning symptoms are perceived. Only a few recom­ mended options are available, and formal research evi- dence for most of these options is limited. This reflects not only the logistical challenges of undertaking ran- domized controlled trials (RCTs) in an unpredict- able and episodic condition that reaches its maximum intensity within just 24 h of onset, but also the fact that current treatments have been available for decades and are accepted as normal clinical practice without a per- ceived need for further research evidence to confirm their efficacy. NSAIDs An oral NSAID is by far the most commonly used treat- ment for acute gout in general practice.15 However, just one placebo-controlled RCT has confirmed that an Key points ■■ Many established treatments are already available for managing gout effectively ■■ Lowering serum uric acid to a target level below the saturation point for urate can effectively ‘cure’ gout ■■ Health professionals need to be aware of current best practice for the management of gout ■■ Patient education and discussion of illness perceptions is key to addressing patient-specific risk factors and ensuring adherence to an individualized management plan ■■ Shared decision-making between a fully informed patient and a health practitioner, whether a doctor, nurse or pharmacist, can greatly increase the likelihood of a ‘cure’ and lead to improvements in patient outcomes NSAID (tenoxicam 40 mg daily) is better than placebo at reducing pain at 24 h.16 Because of the severity of pain experienced in acute gout, the NSAID should be commenced at its maximum dose, rather than titrat- ing upwards from the lower end of the dose range.11,12,14 Although some doctors still hold the traditional belief that indomethacin is more effective than other NSAIDs, studies comparing different quick-acting NSAIDs dem­ on­strate their equal efficacy in acute gout.17–21 The main issue with oral NSAIDs is, of course, their safety. To reduce the risk of upper gastrointestinal ulcera­ tion, bleeding and perforation, co-administration of a ­proton-pump inhibitor (PPI) is recommended,22 although the addition of a PPI does not alter the risk of small and large bowel involvement. In addition, NSAIDs can cause acute renal failure and are contra­indicated in patients with CKD.23 Selective cyclo­oxygenase (COX)‑2 inhibitors compare favourably with non­selective NSAIDs24,25 for the treatment of acute gout in terms of similar efficacy but lower incidence of serious gastro­ intestinal events; however, concerns remain over the cardio­vascular and renal toxicity of COX‑2 inhibitors with frequent or chronic use.26 In the UK, co-­prescription of a PPI is recommended with these drugs.22 Colchicine Along with oral NSAIDs, oral colchicine is recom­mended as a first-line treatment for acute gout attacks.11,12,14 Interest in IL‑1 inhibition with bio­logic agents for acute gout has arisen due to the action of these drugs on the NLRP3 inflammasome; however, colchi­cine is also known to exert one of its many actions here27 and is a much less expensive option. Figure 2 demonstrates the metabolism and various mechanisms of action of colchicine. Oral colchicine has been used in clinical practice for centuries, but only two placebo-controlled RCTs have been published, both of which confirm its efficacy in acute attacks.28,29 These studies both demonstrated that colchicine significantly reduced pain compared with placebo, but high-dose regimens (4.8 mg total over 6 h) were associated with substantial gastrointestinal dis- turbance (nausea, vomiting or diarrhoea). Low-dose regimens (1.8 mg total over 1 h) were equally effective at reducing pain but adverse effects were minimized. Hence, current EULAR guidelines recommend a maxi­ mum dose of 0.5 mg colchicine three times a day,11 and the ACR guidelines14 recommend a regimen involving a loading dose of 1.2 mg then 0.6 mg 1 h later, followed after a further 12 h by 0.6 mg once or twice daily. Such low doses seem to be effective and well-tolerated in clinical practice. Even lower doses (0.5 mg once or twice daily) can be tried in patients with persistent adverse effects and in those with moderate to severe CKD.30 Following the publication of the ACR guidelines, an equivalent dosing is likely to become more widely adopted in Europe—with a modified dosage of a 1.0 mg loading dose followed 1 h later by 0.5 mg on the first day, then 0.5 mg two to four times a day on subsequent days. Colchicine is considered to work best when commenced Dietary purines Uric acid pool (serum, tissues) 30% 70% 70% renal excretion 30% gut elimination MSU crystals Purine nucleotides and bases Tissue nucleotide synthesis and metabolism Osteoarthritis Ageing ObesityHigh-purine diet ■ Genetic factors ■ Metabolic syndrome (hypertension, insulin resistance, hyperlipidaemia) ■ Renal impairment ■ Drugs that reduce renal function Figure 1 | Metabolism of uric acid and risk factors for gout. Crystallization of MSU occurs when uric acid levels exceed the saturation point, through inefficient elimination or excessive production of uric acid. The multiple risk factors for the formation of urate crystals are shown in yellow boxes at their sites of action. Abbreviation: MSU, monosodium urate. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 3. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  273 within 24 h of first symptoms of an acute attack, and the ACR guidelines recommend that colchicine is not initi- ated more than 36 h after symptom onset. Treatment is usually continued until the acute attack has subsided. Colchicine should be used at low doses and with cau­ tion in patients taking a cytochrome P450 3A4 inhibitor (ciclosporin, ketoconazole, ritonavir, clari­thromycin, erythromycin, extended-release verapamil, extended- release diltiazem)27,30,31 owing to drug–drug inter­ actions,31,32 particularly in people with renal impairment, and those taking a statin should stop this medication temporarily while on colchicine.27 Intravenous colchicine is highly toxic and not recommended.11,12,14 Corticosteroids Intra-articular corticosteroids Aspiration of an affected joint followed by injection of corticosteroids (Figure 3) is ideal for treatment of acute gout attacks,11 particularly in a hospital setting where first-line use of this intervention can provide prompt relief with minimal adverse effects. This treatment is a particularly useful option where colchicine, NSAIDs or oral corticosteroids are contraindicated. Aspiration itself very quickly reduces the extreme pain of gout, pre­sumably by lowering the marked intra-articular hyper­tension caused by acute crystal-induced synovitis. Prac­tical difficulties with intra-articular aspiration and injection arise if the gout attack is polyarticular or affects the midfoot, or if no physician with sufficient expertise is available (the usual situation in general practice). One uncontrolled trial has been published, which supports the use of intra-articular corticosteroid injection.33 Although no evidence exists to guide optimal dosage, as for other arthropathies most physicians use a long-acting corticosteroid and adjust the dose according to the size of joint affected.34 Oral corticosteroids Oral prednisolone can be used where colchicine or NSAIDs are contraindicated or have failed to provide sufficient relief,11 and this method of delivery of cortico­ steroids is the one most commonly used in general practice. As with intra-articular corticosteroids, no evidence is available to guide optimal dosage. One key trial demonstrated that, in patients with acute gout, oral prednisolone 35 mg daily was equivalent in efficacy to oral naproxen 500 mg twice a day.35 Another trial demon­strated that 30 mg prednisolone daily for 6 days was equivalent in efficacy to intramuscular diclofenac 75 mg plus 50 mg indomethacin orally on day 1 followed by oral indomethacin 50 mg three times a day for 2 days then 25 mg three times a day for 3 days.36 Such doses of oral corticosteroids administered in short courses are commonly used in general practice for other conditions (for example, asthma) and considered overall to be a safe option, especially in patients with comorbidities. Intramuscular corticosteroids Intramuscular delivery of corticosteroids is another alternative treatment that is mainly limited to hospi- tal settings. Intramuscular corticosteroids are usually admini­stered as a single injection but, again, there is no con­sensus on dose. In one trial, 60 mg intra­mus­ cular triamcinolone acetonide was equivalent in effi- cacy to oral indomethacin 50 mg three times daily for the treatment of acute gout.37 However, higher intra­ muscular doses—120 mg of triamcinolone acetonide or m­ethylprednisolone—are often used to control an acute flare of rheumatoid arthritis or other painful inflamma- tory arthropathies38 and many physicians use such doses for acute gout. Biologic agents Considerable interest exists among rheumatologists in the use of biologic agents for the rare situation where all of the above treatments are considered to be contra-­ indicated or impractical. However, available biologic agents that inhibit IL‑1 and which have been assessed in treatment of acute attacks of gout—specifically ana­ kinra,39,40 canakinumab41–43 and rilonacept44–46 —have either had no comparator or have been compared to a possibly suboptimal dose of triamcinolone (40 mg intra- muscularly) rather than a more commonly used short course of oral prednisolone (for example, 30–35 mg prednisolone daily). Although some modest advan- tages of biologic agents have been reported, these treat- ments remain comparatively very expensive, and as yet are largely unlicenced for acute gout in most countries (canakinumab is now licenced for use in Europe, but not in the UK or USA). Main effects of colchicine ■ Binds to tubulin in cells (affecting cell division) ■ Inhibits function of leukocytes ■ Inhibits chemotactic factors ■ Inhibits inflammasome and IL-1 production ■ Inhibits release of histamine from mast cells Oral administration 80–90% faecal excretion Liver Kidney 10–20% renal excretion Enterohepatic recycling Figure 2 | Metabolism and mechanisms of action of colchicine. After oral administration and gastrointestinal absorption, colchicine is primarily metabolized by the liver but undergoes substantial enterohepatic circulation. Its pharmacological effects are multiple and varied. Colchicine is excreted primarily in faeces but also in urine. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 4. 274  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum Physical treatment In support of widespread clinical experience, one small trial (n = 19) examining the effect of adding localized ice therapy to a standardized treatment for acute attacks47 found greater improvements at 1 week in those who received ice. Locally applied ice packs are therefore recom­mended as a simple and safe adjunct to pharmaco­ logical therapy. No RCTs are available to support resting of the affected joint,12 but such common-sense advice obviously accords with the patient’s natural inclination. Issues related to treatment of acute attacks NSAIDs are readily available but unfortunately are contra­indicated in many patients with gout owing to age, comorbidity, concomitant drug therapy or renal impair­ment.48,49 Pharmacoeconomic analysis has also shown oral NSAIDs to be less cost-effective than oral corticosteroids or colchicine, largely because of their adverse effects.50,51 NSAIDs and colchicine have not been directly compared with respect to their effi- cacy, but colchicine might be more applicable to most patients, with the caveats of severe renal impairment and certain drug interactions listed above. Where expertise is available, joint aspiration and injection with cortico­ steroids is often the safest option and this treatment should be regarded as recommended best practice in a hospital setting. The key to successful management of acute attacks of gout is to assess and treat each patient individually, discussing with them which option is most acceptable. Any treatment should be started as early as possible and drugs can be used in combination if needed (for example, intra-articular corticosteroids with oral colchicine).14 Long-term management The aim of long-term therapy is to ‘cure’ gout by lowering SUA levels to below the saturation point for urate—that is, to at least below 360 μmol/l (6 mg/dl)11 —thus promot- ing crystal dissolution and preventing formation of new crystals. The most effective method of achieving this SUA level is the use of urate-lowering therapy (ULT). Before commencement of ULT, each patient should be given a clear verbal explanation of the causes of gout, backed up by written information (for example, Arthritis Research UK’s patient information booklet52 ), along with an individualized explanation of the relevant risk factors that could elevate their SUA levels. The content listed in Box 1 was used in a study to explain to patients the causes and risk factors for the development of gout, and the patients subsequently demon­strated excellent ad­herence to individualized management plans.53 One of the many barriers to care is a stereotyped view of gout and the feeling that gout is self-induced through poor lifestyle, gluttony and excessive alcohol intake.54 Many patients have seen old cartoons that empha­ size such lifestyle factors (Figure 4). Patients can be embarrassed to have gout—particularly women, who often think it is a man’s disease—which can lead to a dis­inclination to seek help. Negative views about gout are associated with poorly controlled disease and lower adher­ence to ULT.55–57 Therefore, it is important that the explana­tion should be non-accusatory, emphasizing that having gout is not necessarily the patient’s fault, and that the condition is curable with treatment. Exploration and full discussion of the patient’s perception of their illness is crucial if they are to be sufficiently motivated to adhere to an appropriately developed management plan. Individualized, constructive advice on lifestyle modi- fications that might reduce predisposition to hyper­ uricaemia should be given if appropriate. This advice might include, for example: reduce weight by use of recom­mended strategies if overweight or obese;58 reduce excessive intake of beer and spirits;59 reduce exces- sive intake of purine-rich foods (such as red meat and seafood) and consume proportionately more dairy pro­ ducts;60 reduce intake of fructose-rich drinks and fruit (apples, oranges);61 and cease any retinol supplementa- tion (retinol increases SUA compared with β-carotene, which reduces SUA).62 Although these recommendations are based on observational studies rather than RCTs, clearly many health reasons exist for tackling being over- weight or drinking more than the recommended levels of alcohol, and receiving indivi­dualized lifestyle advice might enable the person with gout to take control of their condition and improve their self-efficacy. However, the benefit to gout of lifestyle modification should not be overemphasized since, firstly, this emphasis reinforces the negative perceptions of gout as a self-inflicted condi- tion and, secondly, ULT will still be required to achieve SUA levels low enough to treat the condition within a reasonable timescale. With respect to predisposition to hyperuricaemia and development of gout, being overweight or obese increases endogenous production of uric acid63 and is probably a a b Figure 3 | Management of acute gout attack. a | Example of an acute attack at the first metatarsophalangeal joint, known as ‘podagra’. b | Aspiration of an acutely inflamed joint; note the turbidity of the synovial fluid. Aspiration followed by injection of corticosteroids is safe, rapidly reduces the extreme pain of acute gout and enables confirmation of the diagnosis. In a hospital setting such treatment, especially in an ill patient with multiple comorbidities, should be considered best practice. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 5. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  275 more important factor than individual dietary compo- nents. However, interest is growing in diet­ary altera­tions that might reduce the frequency of acute attacks in people with established gout, rather than influence hyper­ uricaemia and crystal deposition. A long-­recognized paradox is that many patients link the trig­gering of an acute attack to excessive consumption of dietary purine food and alcohol, which might be expected to increase SUA levels, whereas the accepted triggers for an acute attack (for example, intercurrent illness, surgery, initia- tion of ULT) are associated with a rapid decrease in uric acid levels that is hypothesized to encourage partial dis- solution and subsequent ‘shedding’ of preformed crys- tals into the joint space. A possible explanation for this paradox is that an increase in free fatty acids following a large meal might act as a ‘second signal’ and syner- gize with urate crystals to trigger the inflam­masome via ASC/caspase 1‑driven (but not NALP3-driven) release of IL‑1β.64 Conversely, the dairy fractions glyco­ macropeptide and G600 milk fat extract might inhibit triggering of the inflammasome by urate crystals.65 An RCT published in 2012 showed that daily intake of a skimmed milk powder enriched with glycomacropeptide and G600 reduces the frequency of acute gout attacks over a 3‑month period.66 Similarly, evidence from a 1-year case–crossover study showed that regular intake of cherry reduces the frequency of acute attacks, possibly owing to high levels of anthocyanins, which possess anti- inflammatory and antioxidant properties.67 There­fore, if patients are interested in dietary alterations, it might be very reasonable to advise avoidance of large, rich meals and to consider appropriate dairy supplements and cherry or cherry extract as safe self-­management st­rategies for reducing attack frequency. Other modifiable risk factors for hyperuricaemia can also sometimes be addressed. For example, chronic diuretic therapy for hypertension (rather than for heart failure) could be switched, where feasible, to an alterna- tive antihypertensive regimen, bearing in mind that in an observational cohort β-adrenergic blocking agents, angiotensin-converting enzyme inhibitors and non- losartan angiotensin II receptor blockers increased SUA whereas losartan and calcium-channel blockers lowered SUA68 (via reduced or enhanced renal excretion of uric acid, respectively). In some patients with end- stage renal disease, renal transplantation might nor- malize their SUA. Importantly, comorbidities that are associated with hyperuricaemia, such as hyper­tension, hyperlipidaemia and hyperglycaemia,7,69–71 need to be considered and optimally managed. These conditions are important in their own right, but tight control of each can indepen­dently reduce SUA levels through improved renal ex­cretion of uric acid.72,73 Urate-lowering therapy ULT is a key component of curative gout therapy and needs to be fully explained to each patient and upwardly titrated against a specific target SUA level. Patient edu- cation is crucial and, as shown in a proof-of-principle study,53 ensures excellent adherence to ULT. Upwards titration of ULT to achieve a level of SUA well below the saturation threshold of 360 μmol/l is important to enable the dissolution of existing crystals and prevent joint damage—in effect ‘curing’ gout. The lower the level of SUA obtained, the faster the dissolution of crystals and reduction in size of any tophi.74,75 It is important to explain to patients that they will remain at risk of acute Box 1 | Key messages when explaining the nature of gout Explanation of the causes of gout, including the following key points: ■■ We know its cause: deposition of urate crystals in and around peripheral joints ■■ Crystals form when SUA levels rise above the critical ‘saturation point’ for crystal formation ■■ In people with persistently raised SUA levels, crystals slowly but continuously accumulate without causing symptoms ■■ When sufficient crystals have formed in cartilage, some occasionally ‘spill out’ into the joint cavity, triggering severe inflammation of the joint lining and presenting as an acute attack ■■ Over many years, acute attacks can increase in frequency and spread to involve other joints ■■ In addition to acute attacks, continuing crystal deposition might eventually result in hard, slowly expanding lumps of crystals (tophi) that can cause pressure damage to joint cartilage and bone and can even appear as palpable lumps under the skin ■■ In some people, tophi could result in irreversible joint damage and cause regular chronic pain on using the joints ■■ Reduction and maintenance of SUA levels below the saturation point stops production of new crystals and encourages existing crystals to dissolve, so eventually there are no crystals and therefore no gout Explanation of relevant risk factors that elevate SUA levels above the saturation point, including: ■■ Hereditary factors that result in some people having relatively inefficient renal excretion of uric acid ■■ High body mass—the majority (around two-thirds) of uric acid is made by the body’s cells and this production increases with obesity ■■ A purine-rich diet—around one-third of uric acid comes from the diet ■■ Drugs (e.g. diuretics) that reduce the kidney’s ability to excrete uric acid ■■ Chronic renal impairment associated with ageing or kidney disease Abbreviation: SUA, serum uric acid. Figure 4 | “Punch cures the gout, the colic, and the ‘tisick” (anonymous). Historical sterotypes of gout persist and can act as barriers to adequate care. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 6. 276  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum attacks until all existing crystals are dissolved, that if they do develop an attack it is not a sign that their ULT is ineffec­tive or making things worse, and that they should continue the ULT while they manage an acute attack. Gradual upward titration is probably important for reduc- ing the risk of acute flare, and in one study this strategy for initiating ULT seemed not to increase the frequency of attacks more than would have been expected (based on the frequency of self-reported attacks the previous year).53 Limited evidence is available to guide the commence- ment of ULT. Currently, indications for ULT include recurrent attacks of gout, clinically detectable tophi, joint damage or nephrolithiasis11 —in other words, well- established and relatively severe gout. However, urate crystal deposition is increasingly realized to be far more extensive than would be thought by clinical assessment alone. For example, ultrasonography studies published in 2011 and 2012 demonstrate obvious crystal deposition in a substantial proportion of people with asympto­matic hyperuricaemia,76,77 and dual-energy CT has shown extensive urate deposition in people with established gout, including at unsuspected sites such as the distal patellar tendon.78 In patients with gout, synovial fluid aspirated from knees that have not been the site of an acute attack will often show urate crystals.79 Therefore, it seems that crystal deposition is likely to be exten- sive in many peripheral joints even at the time of first presentation and that every person with gout is techni- cally ‘tophaceous’ in terms of having microtophi in and around their joints. Such a perspective is causing a trend towards earlier commencement of ULT, closer to the time of first diagnosis, particularly if a patient is young (for example, in their twenties, thirties or forties) or has a low estimated glomerular filtration rate.12 Certainly the patient should at least be informed at this time about the availability and expected benefits of ULT and be involved in the decision as to whether to start ULT. Most clinicians advocate delaying initiation of ULT until an acute attack has settled, owing to concern that commencing ULT will prolong the attack or precipitate a polyarticular flare. However, one placebo-controlled RCT found no difference in pain or flare rate when ULT with allopurinol (plus colchicine prophylaxis) was started during an acute attack compared with a delay of 10 days.80 Many general practitioners perceive logistical advantages to initiating ULT at the time a patient presents with an acute attack and argue that many patients will not re­attend 4 weeks later when everything is back to normal. Nevertheless, it seems sensible to provide enough infor- mation at the time of the acute attack to encourage sub- sequent reattendance and to wait until the second visit, when the patient is not in severe pain, to fully explain the disease and discuss its long-term management. Xanthine oxidase inhibitors Xanthine oxidase inhibitors (XOIs) are the usual first- choice ULT, with allopurinol favoured over febuxostat due to cost considerations and long-term safety data. XOIs reduce endogenous production of uric acid by inhibiting the conversion of hypoxanthine to xanthine and of xanthine to uric acid. Table 1 compares the key features of the two currently available XOIs. Allopurinol Allopurinol is a purine analogue and a nonspecific com- petitive inhibitor of xanthine oxidase. Its active metabo- lite, oxypurinol, is predominantly excreted via the kidney. Although no placebo-controlled RCTs have confirmed the efficacy of allopurinol, decades of clinical experience and several head-to-head studies81–83 have shown it to be an effective, well-established, cheap and relatively safe ULT, and it is therefore recommended as first-line ULT.11 The dose range of allopurinol is large, meaning it can be titrated in small increments for maximum effective- ness. In a 2013 observational study in the UK,53 the median dose of allopurinol required to reach the thera- peutic target of SUA ≤360 μmol/l (achieved in 90% of participants) was 400 mg once daily. This dose is higher than the commonly prescribed daily dose of 300 mg, demonstrating the importance of adjusting therapy for maximum effectiveness in individual patients. It might be that 300 mg was adequate for most people when the first dose-ranging studies for allopurinol were published around the 1960s, but that population-level increases in height and BMI mean 300 mg is now suboptimal; when used in RCTs in the USA in 2005, this dose achieved the therapeutic target in only around 20% of patients.84,85 An initial dose of 100 mg daily is recom­mended, with sub- sequent incremental increases of 100 mg approximately every month (up to a maximum of 900 mg), stopping these increases at the dose at which SUA is lowered to 360 μmol/l (6 mg/dl)13 or preferably 300 μmol/l (5 mg/dl). The evidence for dose-­adjustment of allo­ purinol in patients with renal impairment, using 50 mg increments, is inconclusive.86–89 Stamp et al.89 suggest that dose-adjustment algorithms based on creatinine clear- ance of allopurinol were overly cautious and that with gradual upward titration the target SUA level can often be reached safely, even in the setting of renal impairment, and at doses that are above those recommended. Table 1 | Comparison of XOIs Characteristic Allopurinol Febuxostat Action Nonspecific XOI Specific XOI Elimination Renal Hepatic Daily dose* (50 mg), 100–900 mg (40 mg), 80 mg, 120 mg DRESS Rare Rare Cautions Severe renal impairment Heart failure (NYHA class III or IV), hepatic failure, eGFR 30 ml/min/1.73 m2 Interactions Cytotoxic drugs metabolized by XO (for example, azathioprine, mercaptopurine) Warfarin Cytotoxic drugs metabolized by XO (for example, azathioprine, mercaptopurine) Cost of 28 tablets‡ £1.06 (300 mg tablets) £24.36 (80 mg or 120 mg tablets) *The doses in parentheses can be used in patients with renal impairment but are not routinely used. ‡ According to information from the British National Formulary.135 Abbreviations: DRESS, drug reaction with eosinophilia and systemic symptoms; eGFR, estimated glomerular filtration rate; NYHA, New York Heart Association; XO, xanthine oxidase; XOI, XO inhibitor. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 7. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  277 Allopurinol is usually well-tolerated, by approximately 8–9 out of 10 patients,53,82 the usual cause of intolerance being nausea, gastrointestinal disturbance, headache or rash. However, allopurinol can rarely cause a serious syn- drome of drug reaction or rash with eosinophilia and sys- temic symptoms (DRESS; previously termed allopurinol hypersensitivity syndrome), which can cause fever, severe cutaneous adverse reactions (SCAR; including toxic epidermal necrolysis and Stevens–Johnson syndrome), hepatitis, vasculitis, renal impairment and sometimes death.90 Allopurinol-induced DRESS usually occurs within the first few months of treatment and reported risk factors include renal impairment, concomitant use of diuretics, initiation of treatment with a fixed dose of 300mg and, particularly in Korean, Chinese and Thai populations, presence of the HLA‑B*5801 allele.12,91 It might be reasonable to consider screening for this polymorphism in populations with a high prevalence of HLA‑B*5801.91 The precise incidence of DRESS from allopurinol use is unclear but allopurinol is reportedly the most frequent cause of SCAR.90 Although rare, the occur- rence of severe DRESS and SCAR is another reason to start low and gradually increase the dose of allopurinol.92 Allopurinol has also been shown to be beneficial in cardiovascular disease93,94 and renal disease,95,96 both of which commonly coexist with or occur as a complication of gout. Although still controversial and not currently licenced for these indications in most countries, this is another potential benefit of ULT which, if explained to patients at risk of cardiovascular or renal disease, might further encourage adherence to treatment. Whether the cardio-renal benefits result from lowering of SUA or from xanthine oxidase inhibition remains uncertain.97 Febuxostat Licenced in 2008 in Europe, febuxostat is a non-purine, highly specific XOI that undergoes predominantly hepatic metabolism. Two placebo-controlled double- blind RCTs have demonstrated its efficacy in reducing SUA.84,98 Two further double-blind RCTs compared doses of 40–240 mg febuxostat against a fixed daily dose of 300 mg allopurinol85,99 and showed that febuxo­stat was superior at doses of 80 mg and 120 mg daily. How­ever, the trials that included allopurinol did not allow for upward titration of the allopurinol dose against SUA level, which is the established best clinical practice. Furthermore, in these RCTs84,85 approximately 30% of participants on febuxostat do not reach the therapeutic target (SUA 360 umol/l) at the highest recommended dose (120 mg daily), whereas in clinical practice upward titra­tion of allopurinol in those who tolerate it would usually achieve this target at doses well under the maximum dose (900 mg daily).53 The most common adverse event deemed attributable to febuxostat in the early clinical trials was liver-­function test abnormalities, but the possibility of an adverse cardio­vascular signal (more myocardial infarction, stroke and cardiovascular death in febuxostat than allopurinol participants, though nonsignificant) led to febuxo­stat not being recommended in people with heart failure.100 Long-term safety studies to address these con­cerns are in progress. Also, although febuxostat was initially thought to be free of the serious SCAR and DRESS reac- tions associated with allopurinol, some cases of severe hypersensitivity cutaneous reactions have subsequently been reported.101,102 Undoubtedly, the main advantage of febuxostat over allopurinol is that it can be used without dose-adjustment or concern over toxicity in people with renal impairment (although it is not recommended for use in those with an estimated glomerular filtration rate 30 ml/min/1.73 m2 ). Disadvantages include the inability to slowly increase the dose (only 80 mg and 120 mg tablets are available in most countries, and provocation of acute gout attacks at the starting dose of 80 mg is likely),84,85,98,99 cardiovascular safety concerns, and high cost. Although the ACR guidelines (which did not consider cost) do not favour one XOI over the other,13 the UK National Institute for Health and Clinical Excellence (which does consider cost utility) recommends allo­purinol as the first-line XOI, with febuxostat recommended as a second option in those who cannot tolerate allopurinol or in whom it is contraindicated or ineffective.103 Uricosuric drugs Available uricosuric drugs act predominantly on urate anion exchanger 1 (URAT1)—an organic anion t­ransporter—to prevent reuptake of uric acid at the proxi­ mal renal tubule and thus increase renal excretion of uric acid (Figure 5). The resulting higher con­centration of uric acid in the collecting tubules can predispose to uric acid stone formation, so the patient should be advised to drink plenty of fluids and remain well-hydrated. Three uricosuric drugs are licenced and well-established as ULT: benzbromarone (50–200 mg daily), probenecid (250– 500 mg twice daily) and sulfin­pyrazone (200–800 mg daily). Although inexpensive, their use is limited by lack of availability in most countries. These medications are all effective at reducing SUA82,104–107 and have dose ranges that readily enable stepwise dosage increases. Probenecid and sulfin­pyrazone are contraindicated in patients with severe renal impairment or nephrolithiasis, which further Basolateral membrane Renal proximal tubule epithelial cell Apical (brush border) membrane Probenecid Benzbromarone Sulfinpyrazone Sulfinpyrazone OAT1 OAT3 GLUT9 URAT1 OAT4 Uric acid Uric acid Probenecid Benzbromarone Proximal tubule lumenCirculation Figure 5 | Mechanism of action of uricosuric drugs at the proximal renal tubule. Uricosuric drugs licenced for gout (shown in blue boxes) act on URAT1 to prevent re-uptake of uric acid and thus increase its renal excretion. As this is a simplified schema, not all transporters are shown. Abbreviations: OAT, organic anion transporter; GLUT9, glucose transporter type 9; URAT1, urate anion exchanger 1. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 8. 278  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum limits their use. Benzbromarone is not contraindicated in those with mild or moderate renal impairment and can still be very effective in this situation. The avail- ability of benz­bromarone became limited mainly due to concerns over reports of hepatotoxicity, particularly in Asia, although the reported risk in European popula- tions is estimated to be at most 1 in 17,000 patients.108 Nevertheless, liver function should be checked whenever SUA level is meas­ured, especially in the first few months of initiation and dose-adjustment of uricosuric therapy. Other available ULTs Losartan,109 oral vitamin C supplements,110,111 and feno­ fibrate112 all have direct uricosuric effects and have been shown to reduce SUA levels in people with hyper­ uricaemia. For losartan and fenofibrate, this uricos­ uric action is in addition to their beneficial effects on hypertension and hyperlipidaemia, respectively. How­ ever, the urate-lowering effects of all three approaches are fairly modest and, on their own, are unlikely to achieve the therapeutic target level of SUA.113 These agents remain unlicenced for use in gout, but losartan and feno­fibrate can be considered as useful adjunctive ther­apy for patients who require treatment for coexistent hy­pertension or hyperlipidaemia. Unlike some other animals and bacteria, humans lack uricase—an enzyme that converts uric acid to highly soluble allantoin. Pegloticase is a pegylated uricase that was recently licenced (2010 in the USA, 2013 in Europe) for use in ‘treatment-refractory’ gout. This drug is administered by repeated intravenous infusion (8 mg every 2 weeks seems optimal), a regimen that limits its use to the hospital setting. Pegloticase is extremely effective at rapidly reducing SUA to very low levels for several weeks.114 However, it is also antigenic,115 with a risk of anaphylaxis and infusion reaction, so premedi- cation with antihistamine and corticosteroids in an appropriately staffed facility is recommended.116 Con­ cerns also remain over cardiovascular risk, and this drug should be used with caution in people with heart failure. A large proportion of patients undergoing repeat infusions develop blocking antibodies against the PEG component,115 which not only reduces ULT efficacy but also increases the risk of adverse reactions. Therefore, following successful initiation of treatment, a subse- quent preinfusion SUA 360 μmol/l is an indication to withdraw treatment (recom­mended by the manu­fac­ turers). Although pegloticase is seemingly very efficient at induc­ing rapid regression of tophi, the number of patients who are truly refractory to other standard ULTs is likely to be extremely small, and because of safety and tolerability concerns, and the cost and logistics of deliv- ery of this drug, its use is likely to be very restricted. Rasburicase, a uricase that has been licenced for many years for tumour lysis prophylaxis, has also been used to treat ‘refractory’ gout where intolerance or renal impair- ment precludes the use of other ULTs.117,118 However, although some patients seem to tolerate repeated doses, rasburicase is unlicenced for gout and is more antigenic than pegloticase.119,120 ULTs in development Other drugs that are undergoing clinical investigation as potential ULTs include RDEA594 (lesinurad), which is a uricosuric,121 and BCX4208 (ulodesine), which is a purine nucleoside phosphorylase inhibitor.122 How­ever, these agents are not yet available or licenced for use in gout, either as monotherapy or in combination with an XOI. What is the optimum SUA level? Current guidelines agree that SUA should be reduced and maintained to a level below 360 μmol/l (6 mg/dl)11,13 to prevent further crystal formation and to encourage dis- solution of exisiting crystals.123 However, the lower the SUA, the faster the dissolution of crystals and tophi75 and the sooner the patient reaches the state of being ‘cured’. Therefore, particularly in patients with a long history of gout79 or the presence of tophi, an initial target of 300 μmol/l (5 mg/dl) might be more successful; this is the target recommended by the BSR.12 Concern is increasing, however, that lifelong maintenance (that is, after acute attacks have stopped and any tophi have resolved) of SUA at very low levels might increase the risk of neuro­degenerative diseases such as Parkinson, dementia and multiple sclerosis, which appear to be negatively associated with gout.124 This is because uric acid is a strong antioxidant97 and although this benefit is countered by its other detrimental effects on vascular endothelium, its antioxidant benefits may predominate within the central nervous system. Although there is no absolute agreement on the optimal SUA level that asso- ciates with no disease risk in humans, the concept of an initial crystal-clearing ‘induction phase’ followed by a later ‘maintenance phase’ of ULT is one that is increas- ingly discussed.125 We suggest that the initial SUA target to effect a ‘cure’ should be well under 300 μmol/l for the first 3–5 years of treatment but that subsequently, when no further attacks have occurred and any tophi have regressed, the dose of ULT should be reduced to allow the SUA to rise somewhat but remain below the saturation point to prevent crystal formation (that is, in the range 300–360 μmol/l) (Figure 6). The length of time the patient should continue ULT is guided by SUA level. For most patients, use of this target could mean lifelong therapy, but at a low dose. However, in patients with an attributable risk factor that can be success­fully modified (for example, by stopping long-term thia­ zide therapy, successful renal transplantation, marked reduction in BMI) cessation of ULT might be consid- ered after the induction phase or some time into the maintenance phase. Flare prophylaxis Initiation of ULT can provoke acute attacks, some- times involving several joints and even joints pre­viously un­affected by gout. This flare of disease is particularly likely if SUA is rapidly and substantially lowered by a high or very efficient dose of ULT (for example, allo­ purinol 300 mg daily, febuxostat 80 mg daily, uricase). It is thought to result from the rapid partial dissolution REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 9. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  279 of and thus easier ‘shedding’ of crystals. If the patient is not fully informed about the possibility of this flare in advance, such an event might cause a patient, quite reason­ably, to give up on their ULT and even to be unwilling to consider further advice for their gout. To reduce the risk of ULT-induced flares, the EULAR guidelines recom­mend co-prescription of either low- dose colchicine (usually 0.5 mg once or twice daily) or an oral NSAID (plus PPI) as prophylaxis for the first few months of ULT.11 Certainly one RCT has confirmed that colchicine is more effective than placebo in reducing the number of acute attacks during the first 6 months of allopurinol treatment.126 Additionally, a post hoc analysis of three RCTs of febuxostat, in which patients received either 8 weeks or 6 months of prophylaxis with naproxen 250 mg twice daily or colchicine 0.6 mg daily, found an increase in flare rate (up to 40%) at the end of 8 weeks of prophylaxis, whereas flare rates remained low (3–5%) at the end of 6 months of prophylaxis.127 However, low-dose colchicine, and possibly NSAIDs, might reduce the flare rate in people who are not on ULT, and a key question is whether initiation of ULT using the recommended slow up-titration regimen (as is possible with allo­purinol in particular) actually causes more attacks than may have been expected otherwise. Furthermore, in a study (n = 96) where patients were given a full explanation of the possible risk of flare from ULT and of the avail­ability of prophylaxis, the vast majority (96%) preferred to take just the one drug (ULT) without prophylaxis and to just manage acute flares as they arose.53 Most patients in this study (65%) experienced fewer attacks in the year fol- lowing ULT initiation than in the preceding year and only one in five reported more attacks (mean 2.7 more). Further studies of whether or not slow up-titration increases the risk of flare are warranted to better inform shared decision-making. Rilonacept and canakinumab have shown efficacy in gout prophylaxis, but as already noted these agents are very expensive and currently not licenced for this use.45,46 Improving the standard of care Despite the availability of effective treatments for gout and of published recommendations to guide best prac- tice, it is apparent that the current standard of care for patients with gout is more than just “suboptimal”.128–132 This criticism applies to both primary and secondary care physicians, including rheumatologists. Few patients are given an adequate explanation of the cause of gout and its possible outcomes, or educated as to the avail­ability and possible benefits of treatment. Most doctors concentrate solely on the management of acute attacks rather than long-term therapy, and only one-third to one-half of gout patients ever receive ULT.1,10,15 Very few general practi- tioners who offer ULT aim for a speci­fic SUA level or serially check SUA133 and most use allopurinol at a fixed dose of 300 mg (which is insufficient for most patients).53 This ‘standard of care’ leads to many people with gout continuing to experience acute attacks and being at unnecessary risk of developing joint damage. Because appropriate information is not provided, patients with gout show poor adherence to any ULT offered and, if they stop ULT because ‘the tr­eatment’s not working’, they are often misclassified as ‘treatment failures’.130 Barriers to effective care can develop both from the patient and the physician (Box 2).54 Full clinical assessment followed by clear informa- tion from the practitioner and subsequent involvement Time SUAlevel Dissolution of MSU crystals 360μmol/l Introduction of ULT Figure 6 | Graph representing clearance induction and maintenance phases of therapy for gout. In the induction phase, ULT reduces the SUA level to well below the saturation threshold (indicated by the dashed line), to enable dissolution of existing crystals and ’cure’ of gout. In the maintenance phase, ULT is reduced following this ‘cure’, and the SUA level is allowed to rise to just below the threshold for maintenance. Abbreviations: MSU, monosodium urate; SUA, serum uric acid; ULT, urate-lowering therapy. Box 2 | Barriers to effective management of gout54,130 Patient barriers ■■ Patients lack knowledge and understanding about the causes and consequences of gout ■■ Gout is often not considered as ‘arthritis’ or as something serious, and is often associated with humour ■■ Gout is considered a man’s disease; women might be unwilling or embarrassed to receive the diagnosis, men might be reluctant to seek medical attention ■■ Gout is considered as a normal part of ageing, self- inflicted by overindulgence and poor lifestyle, stigma and negative stereotypical view ■■ Patients do not adhere to pharmacological treatment Physician barriers ■■ Many primary care staff lack knowledge and understanding about the causes and consequences of gout ■■ Gout is not always managed as a long-term chronic disease but as an acute condition ■■ Patients are not educated or given information to encourage lifestyle modification and adherence to long-term urate-lowering therapy ■■ Clinical guidelines for gout are not known or utilized, particularly in primary care ■■ Training and education in best practice for the treatment and management of gout is lacking at the undergraduate and postgraduate level ■■ Care providers lack incentives to improve treatment and long-term management of gout in general practice Adapted by permission from BMJ Publishing Group Limited. Spencer, K. et al. Ann. Rheum. Dis. 71, 1490–1495 (2012).54 REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 10. 280  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum of the informed patient in decision-making is impor- tant in any medical condition. This process is central to the principles of health care and is considered a key professional responsibility by the UK General Medical Council.134 Fortunately, gout is a very easy disease to explain—certainly easier than many other less-well- understood musculoskeletal conditions—and in the vast majority of cases the practitioner can be very posi- tive when discussing management options and progno- sis (Figure 7). Poor adherence is predominantly due to lack of patient education by the doctor. As demonstrated in a proof-of-­principle study,53 a package of care that includes full patient education on gout and its treatment, discussion of illness perceptions, and development of an individual­ized management plan (that is, recommended best practice) can result in excellent patient outcomes. When given appropriate information (Box 1), 100% of patients in this study opted for ULT and during 1 year of follow-up adherence was excellent, with more than 90% achieving and maintaining the therapeutic target of SUA level 360 μmol/l and beginning to show improve- ments in quality of life.53 Although participants in this study were first seen by a rheumatologist in a hospi­tal set­ting (for approximately 1 h), subsequent follow-up was carried out by a specially trained nurse, often in the patient’s home. As is the case with other prevalent chronic conditions (for example, diabetes, asthma), it is likely that nurse led-care of gout could be successful and cost-effective in a community setting; an RCT is currently comparing nurse-led care to ongoing general practitioner care over a 2‑year period in UK general practice. Given the high prevalence of gout and its potentially serious consequences, it is clearly important that health professionals have appropriate training in the ‘best prac- tice’ management of gout, to enable them to educate and treat their patients appropriately. General practitioners in the UK admit in general that incentives, especially finan- cial incentives, are required for them to alter their inter- est and practice,54 and this change could be facilitated if gout became a designated condition within the Quality and Outcomes Framework for general practice in the UK—a scheme in the UK that offers financial incentives to general practices for achieving specified medical targets for common conditions. Should the growing evidence that high SUA level is an independent risk factor for cardio- vascular disease, stroke and CKD become accepted, and should ULT become licenced for the treatment of hyper­ uricaemia per se, these developments would have a major impact on the prevalence and incidence of gout. How­ever, given the currently high prevalence of gout, the impact it has on patient health and quality of life, andthe availability of effective ‘curative’ treatment, it seems justified to argue for appropriate education of practitioners to improve standards of care on the basis of gout itself. Conclusions A number of established and effective treatments exist for managing gout, with new options on the horizon. Despite this, gout care is often suboptimal and it is apparent that just having effective drugs is not sufficient to effect a ‘cure’.131 Education of patients can empower them to make lifestyle changes and ensure they under- stand the importance of adherence to long-term ULT. In conjunction with this, health-care professionals need to be aware of current best practice and how to up-titrate ULT against a specific target level of SUA, but more importantly they need to know how to address illness perceptions and educate their patients appropriately, so that individualized management plans can be developed on the basis of shared decision-making. Current trends are towards much earlier consideration of ULT, increas- ingly towards the time of first diagnosis, and although still controversial the arguments to treat elevated SUA for cardiovascular and renal disease rather than just gout are becoming stronger and have been accepted in some countries including Japan. Although gout is relatively well-understood, further research is required into various aspects including the optimum level of SUA for long-term manage­ment and, most importantly, how current stand- ards of care can be improved using existing knowledge and effective treatments. Figure 7 | A full and holistic assessment followed by individualized patient education is core to the management of people with gout. In this instance, a specialist nurse practitioner is assessing a patient and her explanation of his chronic tophaceous gout, its treatment options, and the likely benefits he can expect from ULT will be backed up by focused patient literature. Written consent for publication was obtained from the patient and nurse. Image provided courtesy of M. Doherty. Review criteria Articles for this Review were found by systematic review of the following databases: MEDLINE (1946–present), Embase (1974–present), PubMed (from inception to present), Cochrane Controlled Trials Register (from inception to present), ISI Web of Science and AMED (1985–present). The search strategy used terms for gout, including “gouty arthritis”, “podagra”, “tophi”, “monosodium urate crystals” and “hyperuricaemia”, in combination with terms for studies or trials, including “RCTs”, “cohort studies”, “case–control studies” and “systematic reviews”. Preliminary searches were undertaken in March 2012, and were updated July 2012. Subsequent relevant articles were added as published. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 11. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  281 1. Kuo, C. F., Grainge, M. J., Mallen, C., Zhang, W. Doherty, M. Rising burden of gout in the UK but continuing suboptimal management: a nationwide population study. Ann. Rheum. Dis. http://dx.doi.org/10.1136/annrheumdis‑ 2013‑204463. 2. Zhu, Y., Pandya, B. J. Choi, H. K. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007–2008. Arthritis Rheum. 63, 3136–3141 (2011). 3. Kuo, C. F. et al. Risk of myocardial infarction among patients with gout: a nationwide population-based study. Rheumatology (Oxford) 52, 111–117 (2013). 4. Roddy, E., Zhang, W. Doherty, M. Are joints affected by gout also affected by osteoarthritis? Ann. Rheum. Dis. 66, 1374–1377 (2007). 5. Palmer, T. M. et al. Association of plasma uric acid with ischaemic heart disease and blood pressure: Mendelian randomisation analysis of two large cohorts. BMJ 347, f4262 (2013). 6. Stack, A. G., Independent and conjoint associations of gout and hyperuricaemia with total and cardiovascular mortality. QJM 106, 647–658 (2013). 7. Holme, I., Aastveit, A. H., Hammar, N., Jungner, I. Walldius, G. Uric acid and risk of myocardial infarction, stroke and congestive heart failure in 417,734 men and women in the Apolipoprotein MOrtality RISk study (AMORIS). J. Intern. Med. 266, 558–570 (2009). 8. Jolly, S. E. et al. Uric acid, hypertension, and chronic kidney disease among Alaska Eskimos: the Genetics of Coronary Artery Disease in Alaska Natives (GOCADAN) study. J. Clin. Hypertens. 14, 71–77 (2012). 9. Gnanenthiran, S. R., Hassett, G. M., Gibson, K. A. McNeil, H. P. Acute gout management during hospitalization: a need for a protocol. Intern. Med. J. 41, 610–617 (2011). 10. Roddy, E., Mallen, C. D., Hider, S. L. Jordan, K. P. Prescription and comorbidity screening following consultation for acute gout in primary care. Rheumatology (Oxford) 49, 105–111 (2010). 11. Zhang, W. et al. EULAR evidence based 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. 65, 1312–1324 (2006). 12. Jordan, K. et al. British Society for Rheumatology and British Health Professionals in Rheumatology guideline for the management of gout. Rheumatology (Oxford) 46, 1372–1374 (2007). 13. Khanna, D. et al. 2012 American College of Rheumatology guidelines for management of gout. Part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. (Hoboken) 64, 1431–1446 (2012). 14. Khanna, D. et al. 2012 American College of Rheumatology guidelines for management of gout. Part 2: therapy and antiinflammatory prophylaxis of acute gouty arthritis. Arthritis Care Res. (Hoboken) 64, 1447–1461 (2012). 15. Mikuls, T. R. et al. Gout epidemiology: results from the UK General Practice Research Database, 1990–1999. Ann Rheum Dis. 64, 267–272 (2005). 16. Garcia de la Torre, I. Double-blind parallel study comparing tenoxicam and placebo in acute gouty arthritis [Spanish]. Invet. Med. Int. 14, 92–97 (1987). 17. Altman, R. D., Honig, S., Levin, J. M. Lightfoot, R. W. Ketoprofen versus indomethacin in patients with acute gouty arthritis: a multicenter, double blind comparative study. J. Rheumatol. 15, 1422–1426 (1988). 18. Butler, R. C. et al. Double-blind trial of flurbiprofen and phenylbutazone in acute gouty arthritis. Br. J. Clin. Pharmacol. 20, 511–513 (1985). 19. Shrestha, M. et al. Randomized double-blind comparison of the analgesic efficacy of intramuscular ketorolac and oral indomethacin in the treatment of acute gouty arthritis. Ann. Emerg. Med. 26, 682–686 (1995). 20. Lederman, R. A double-blind comparison of etodolac and high doses of naproxen in the treatment of acute gout. Adv.Ther. 7, 344–354 (1990). 21. Maccagno, A., Di Giorgio, E. Romanowicz, A. Effectiveness of etodolac (‘Lodine’) compared with naproxen in patients with acute gout. Curr. Med. Res. Opin. 12, 423–429 (1991). 22. National Collaborating Centre for Chronic Conditions. Osteoarthritis: national clinical guideline for care and management in adults (Royal College of Physicians, London, 2008). 23. Brater, D. C. Anti-inflammatory agents and renal function. Semin.Arthritis Rheum. 32 (Suppl. 1), 33–42 (2002). 24. Rubin, B. R. et al. Efficacy and safety profile of treatment with etoricoxib 120 mg once daily compared with indomethacin 50 mg three times daily in acute gout: a randomized controlled trial. Arthritis Rheum. 50, 598–606 (2004). 25. Schumacher, H. R. Jr et al. Randomised double blind trial of etoricoxib and indometacin in treatment of acute gouty arthritis. BMJ 324, 1488–1492 (2002). 26. Chen, L. Ashcroft, D. Risk of myocardial infarction associated with selective COX‑2 inhibitors: meta-analysis of randomised controlled trials. Pharmacoepidemiol. Drug Saf. 16, 762–772 (2007). 27. Terkeltaub, R. A. Colchicine update: 2008. Semin.Arthritis Rheum. 38, 411–419 (2009). 28. Ahern, M. J. et al. Does colchicine work? The results of the first controlled study in acute gout. Aust. NZ J. Med. 17, 301–304 (1987). 29. Terkeltaub, R. A. et al. High versus low dosing of oral colchicine for early acute gout flare: twenty‑four‑hour outcome of the first multicenter, randomized, double-blind, placebo-controlled, parallel-group, dose-comparison colchicine study. Arthritis Rheum. 62, 1060–1068 (2010). 30. Terkeltaub, R. A. et al. Novel evidence-based colchicine dose-reduction algorithm to predict and prevent colchicine toxicity in the presence of cytochrome P450 3A4/P-glycoprotein inhibitors. Arthritis Rheum. 63, 2226–2237 (2011). 31. Clive, D. M. Renal transplant-associated hyperuricemia and gout. J.Am. Soc. Nephrol. 11, 974–979 (2000). 32. US Food and Drug Administration. Information for healthcare professionals: new safety information for colchicine (marketed as Colcrys) [online], http://www.fda.gov/Drugs/DrugSafety/ PostmarketDrugSafetyInformationforPatientsand Providers/DrugSafetyInformationforHeathcare Professionals/ucm174315.htm (2009). 33. Fernández, C., Noguera, R., González, J. A. Pascual, E. Treatment of acute attacks of gout with a small dose of intraarticular triamcinolone acetonide. J. Rheumatol. 26, 2285–2286 (1999). 34. Courtney, P. Doherty, M. Joint aspiration and injection and synovial fluid analysis. Best Pract. Res. Clin. Rheumatol. 23, 161–192 (2009). 35. Janssens, H. J., Janssen, M., van de Lisdonk, E. H., van Riel, P. L. van Weel, C. Use of oral prednisolone or naproxen for the treatment of gout arthritis: a double-blind, randomised equivalence trial. Lancet 371, 1854–1860 (2008). 36. Man, C. Y., Cheung, I. T., Cameron, P. A. Rainer, T. H. Comparison of oral prednisolone/ paracetamol and oral indomethacin/ paracetamol combination therapy in the treatment of acute goutlike arthritis: a double- blind, randomized, controlled trial. Ann. Emerg. Med. 49, 670–677 (2007). 37. Alloway, J. A., Moriarty, M. J., Hoogland, Y. T. Nashel, D. J. Comparison of triamcinolone acetonide with indomethacin in the treatment of acute gouty arthritis. J. Rheumatol. 20, 111–113 (1993). 38. Choy, E. H., Kingsley, G. H., Khoshaba, B., Pipitone, N. Scott, D. L. Intramuscular Methylprednisolone Study Group. A two year randomised controlled trial of intramuscular depot steroids in patients with established rheumatoid arthritis who have shown an incomplete response to disease modifying antirheumatic drugs. Ann. Rheum. Dis. 64, 1288–1293 (2005). 39. So, A., De Smedt, T., Revaz, S. Tschopp, J. A pilot study of IL‑1 inhibition by anakinra in acute gout. Arthritis Res.Ther. 9, R28 (2007). 40. Chen, K., Fields, T., Mancuso, C. A., Bass, A. R. Vasanth, L. Anakinra’s efficacy is variable in refractory gout: report of ten cases. Semin. Arthritis Rheum. 40, 210–214 (2010). 41. Schlesinger, N. et al. Canakinumab relieves symptoms of acute flares and improves health-related quality of life in patients with difficult‑to‑treat gouty arthritis by suppressing inflammation: results of a randomized, dose- ranging study.Arthritis Res.Ther. 13, R53 (2011). 42. So, A. et al. Canakinumab for the treatment of acute flares in difficult‑to‑treat gouty arthritis: results of a multicenter, phase II, dose-ranging study. Arthritis Rheum. 62, 3064–3076 (2010). 43. Schlesinger, N. et al. Canakinumab for acute gouty arthritis in patients with limited treatment options: results from two randomised, multicentre, active-controlled, double-blind trials and their initial extensions. Ann. Rheum. Dis. 71, 1839–1848 (2012). 44. Terkeltaub, R. et al. The interleukin 1 inhibitor rilonacept in treatment of chronic gouty arthritis: results of a placebo-controlled, monosequence crossover, non-randomised, single-blind pilot study. Ann. Rheum. Dis. 68, 1613–1617 (2009). 45. Schumacher, H. R. Jr et al. Rilonacept (interleukin‑1 Trap) in the prevention of acute gout flares during initiation of urate-lowering therapy: results of a phase II randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 64, 876–884 (2012). 46. Schumacher, H. R. Jr et al. Rilonacept (interleukin‑1 Trap) for prevention of gout flares during initiation of uric acid-lowering therapy: results from a phase III randomized, double- blind, placebo-controlled, confirmatory efficacy study. Arthritis Care Res. 64, 1462–1470 (2012). 47. Schlesinger, N. et al. Local ice therapy during bouts of acute gouty arthritis. J. Rheumatol. 29, 331–334 (2002). 48. Gambaro, G. Perazella, M. A. Adverse renal effects of anti-inflammatory agents: evaluation of selective and nonselective cyclooxygenase inhibitors. J. Intern. Med. 253, 643–652 (2003). 49. Stamp, L. K. Jordan, S. The challenges of gout management in the elderly. Drugs Aging 28, 591–603 (2011). 50. Cattermole, G. N., Man, C. Y., Cheng, C. H., Graham, C. A. Rainer, T. H. Oral prednisolone REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 12. 282  |  MAY 2014  |  VOLUME 10 www.nature.com/nrrheum is more cost-effective than oral indomethacin for treating patients with acute gout-like arthritis. Eur. J. Emerg. Med. 16, 261–266 (2009). 51. Wertheimer, A. I., Davis, M. W. Lauterio, T. J. A new perspective on the pharmacoeconomics of colchicine. Curr. Med. Res. Opin. 27, 931–937 (2011). 52. Arthritis Research UK. Gout [online], http:// www.arthritisresearchuk.org/arthritis- information/conditions/gout.aspx (2013). 53. Rees, F., Jenkins, W. Doherty, M. Patients with gout adhere to curative treatment if informed appropriately: proof‑of‑concept observational study. Ann. Rheum. Dis.72, 826–830 (2013). 54. Spencer, K., Carr, A. Doherty, M. Patient and provider barriers to effective management of gout in general practice: a qualitative study. Ann. Rheum. Dis. 71, 1490–1495 (2012). 55. Dalbeth, N. et al. Illness perceptions in patients with gout and the relationship with progression of musculoskeletal disability. Arthritis Care Res. (Hoboken) 63, 1605–1612 (2011). 56. Lindsay, K., Gow, P., Vanderpyl, J., Logo, P. Dalbeth, N. The experience and impact of living with gout: a study of men with chronic gout using a qualitative grounded theory approach. J. Clin. Rheumatol. 17, 1–6 (2011). 57. Harrold, L. R. et al. Patients’ knowledge and beliefs concerning gout and its treatment: a population based study. BMC Musculoskelet. Disord. 13, 180 (2012). 58. Choi, H. K., Atkinson, K., Karlson, E. W. Curhan, G. Obesity, weight change, hypertension, diuretic use, and risk of gout in men: the Health Professionals Follow-up Study. Arch. Intern. Med. 165, 742–748 (2005). 59. Choi, H. K., Atkinson, K., Karlson, E. W., Willett, W. Curhan, G. Alcohol intake and risk of incident gout in men: a prospective study. Lancet. 363, 1277–1281 (2004). 60. Choi, H. K., Atkinson, K., Karlson, E. W., Willett, W. Curhan, G. Purine-rich foods, dairy and protein intake, and the risk of gout in men. N. Engl. J. Med. 350, 1093–1103 (2004). 61. Choi, H. K. Curhan, G. Soft drinks, fructose consumption, and the risk of gout in men: prospective cohort study. BMJ 336, 309–312 (2008). 62. Choi, W. J., Ford, E. S., Curhan, G., Rankin, J. I. Choi, H. K. Independent association of serum retinol and β‑carotene levels with hyperuricemia: a national population study. Arthritis Care Res. (Hoboken) 64, 389–396 (2012). 63. Lyngdoh, T. et al. Serum uric acid and adiposity: deciphering causality using a bidirectional Mendelian randomization approach. PLoS ONE 7, e39321 (2012). 64. Joosten, L. A. B. et al. Engagement of fatty acids with Toll-like receptor 2 drives interleukin‑1 production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 62, 3237–3248 (2010). 65. Dalbeth, N. et al. Identification of dairy fractions with antiinflammatory properties in models of acute gout. Ann. Rheum. Dis. 69, 766–769 (2010). 66. Dalbeth, N. et al. Effects of skim milk powder enriched with glycomacropeptide and G600 milk fat extract on frequency of gout flares: a proof‑of‑concept randomised controlled trial. Ann. Rheum. Dis. 71, 929–934 (2012). 67. Zhang, Y. et al. Cherry consumption and decreased risk of recurrent gout attacks. Arthritis Rheum. 64, 4004–4011 (2012). 68. Choi, H. K., Soriano, L. C., Zhang, Y. Rodríguez, L. A. G. Antihypertensive drugs and risk of incident gout among patients with hypertension: population based case-control study. BMJ 344, d8190 (2012). 69. Kaya, E. B. et al. Serum uric acid levels predict the severity and morphology of coronary atherosclerosis detected by multidetector computed tomography. Atherosclerosis 213, 178–183 (2010). 70. Grayson, P. C., Kim, S. Y., Lavalley, M. Choi, H. K. Hyperuricemia and incident hypertension: a systematic review and meta- analysis. Arthritis Care Res. (Hoboken) 63, 102–110 (2011). 71. Numata, T., Miyatake, N., Wada, J. Makino, H. Comparison of serum uric acid levels between Japanese with and without metabolic syndrome. Diabetes Res. Clin. Pract. 80, e1–e5 (2008). 72. Li, C., Hsieh, M. C. Chang, S. J. Metabolic syndrome, diabetes, and hyperuricaemia. Curr. Opin. Rheumatol. 25, 210–216 (2013). 73. Stamp, L. K. Chapman, P. T. Gout and its comorbidities: implications for therapy. Rheumatology (Oxford) 52, 34–44 (2013). 74. Perez-Ruiz, F. Treating to target: a strategy to cure gout. Rheumatology (Oxford) 48 (Suppl. 2), ii9-ii14 (2009). 75. Perez-Ruiz, F., Calabozo, M., Pijoan, J. I., Herrero- Beites, A. M. Ruibal, A. Effect of urate-lowering therapy on the velocity of size reduction of tophi in chronic gout. Arthritis Rheum. 47, 356–360 (2002). 76. De Miguel, E. et al. Diagnosis of gout in patients with asymptomatic hyperuricaemia: a pilot ultrasound study. Ann. Rheum. Dis. 71, 157–158 (2012). 77. Pineda, C. et al. Joint and tendon subclinical involvement suggestive of gouty arthritis in asymptomatic hyperuricemia: an ultrasound controlled study. Arthritis Res.Ther. 13, R4 (2011). 78. Choi, H. K. et al. Dual energy CT in gout: a prospective validation study.Ann. Rheum. Dis. 71, 1466–1471 (2012). 79. Pascual, E. Persistence of monosodium urate crystals and low grade inflammation in the synovial fluid of untreated gout. Arthritis Rheum. 34, 141–145 (1991). 80. Taylor, T., Mecchella, J., Larson, R., Kerin, K. MacKenzie, T. Initiation of allopurinol at first medical contact for acute attacks of gout: a randomized clinical trial. Am. J. Med. 125, 1126–1134 (2012). 81. Reinders, M. K. et al. A randomised controlled trial on the efficacy and tolerability with dose escalation of allopurinol 300–600 mg/day versus benzbromarone 100–200 mg/day in patients with gout. Ann. Rheum. Dis. 68, 892–897 (2009). 82. Kuzell, W. C., Seebach, L. M., Glover, R. P. Jackman, A. E. Treatment of gout with allopurinol and sulphinpyrazone in combination and with allopurinol alone. Ann. Rheum. Dis. 25, 634–642 (1966). 83. Bull, P. W. Scott, J. T. Intermittent control of hyperuricemia in the treatment of gout. J. Rheumatol. 16, 1246–1248 (1989). 84. Schumacher, H. R. Jr et al. Effects 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 Rheum. 59, 1540–1548 (2008). 85. Becker, M. A. et al. Febuxostat compared with allopurinol in patients with hyperuricemia and gout. N. Engl. J. Med. 353, 2450–2461 (2005). 86. Perez-Ruiz, F., Hernando, I., Villar, I. Nolla, J. M. Correction of allopurinol dosing should be based on clearance of creatinine, but not plasma creatinine levels: another insight to allopurinol- related toxicity. J. Clin. Rheumatol. 11, 129–133 (2005). 87. Vazquez-Mellado, J., Morales, E. M., Pacheco- Tena, C. Burgos-Vargas, R. Relation between adverse events associated with allopurinol and renal function in patients with gout. Ann. Rheum. Dis. 60, 981–983 (2001). 88. 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. 33, 1646–1650 (2006). 89. Stamp, L. K. et al. Using allopurinol above the dose based on creatinine clearance is effective and safe in patients with chronic gout, including those with renal impairment. Arthritis Rheum. 63, 412–421 (2011). 90. Kim, S. C., Newcomb, C., Margolis, D., Roy, J. Hennessy, S. Severe cutaneous reactions requiring hospitalization in allopurinol initiators: a population-based cohort study. Arthritis Care Res. (Hoboken) 65, 578–584 (2013). 91. Yeo, S. I. HLA‑B*5801: utility and cost- effectiveness in the Asia-Pacific Region. Int. J. Rheum. Dis. 16, 254–257 (2013). 92. Stamp, L. K. et al. Starting dose is a risk factor for allopurinol hypersensitivity syndrome: a proposed safe starting dose of allopurinol. Arthritis Rheum. 64, 2529–2536 (2012). 93. Doehner, W. et al. Effects of xanthine oxidase inhibition with allopurinol on endothelial function and peripheral blood flow in hyperuricemic patients with chronic heart failure: results from 2 placebo-controlled studies. Circulation 105, 2619–2624 (2002). 94. Noman, A., Ang, D., Ogston, S., Lang, C. Struthers, A. Effect of high-dose allopurinol on exercise in patients with chronic stable angina: a randomised, placebo controlled crossover trial. Lancet 375, 2161–2167 (2010). 95. Goicoechea, M. et al. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin. J.Am. Soc. Nephrol. 5, 1388–1393 (2010). 96. Momeni, A., Shahidi, S., Seirafian, S., Taheri, S. Kheiri, S. Effect of allopurinol in decreasing proteinuria in type 2 diabetic patients. Iran. J. Kidney Dis. 4, 128–132 (2010). 97. Neogi, T. et al. Are either or both hyperuricemia and xanthine oxidase directly toxic to the vasculature? A critical appraisal. Arthritis Rheum. 64, 327–338 (2012). 98. Becker, M. A. 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 efficacy in patients with gout. Arthritis Rheum. 52, 916–923 (2005). 99. Becker, M. A. et al. The urate-lowering efficacy and safety of febuxostat in the treatment of the hyperuricemia of gout: the CONFIRMS trial. Arthritis Res.Ther. 12, R63 (2010). 100. European Medicines Agency. Summary of product characteristics: adenuric film-coated tablets [online], http://www.medicines.org.uk/ emc/medicine/22830/SPC (2012). 101. Abeles, A. M. Febuxostat hypersensitivity. J. Rheumatol. 39, 659 (2012). 102. Chohan, S. Safety and efficacy of febuxostat treatment in subjects with gout and severe allopurinol adverse reactions. J. Rheumatol. 38, 1957–1959 (2011). 103. National Institute for Health and Care Excellence. Febuxostat for the management of hyperuricaemia in people with gout. NICE technology appraisal guidance 164 [online],  REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved
  • 13. NATURE REVIEWS | RHEUMATOLOGY VOLUME 10  |  MAY 2014  |  283 http://www.nice.org.uk/guidance/TA164 (2008). 104. Ogino, K. et al. Uric acid-lowering treatment with benzbromarone in patients with heart failure: a double-blind placebo-controlled crossover preliminary study. Circ. Heart Fail. 3, 73–81 (2010). 105. Reinders, M. K. et al. Efficacy and tolerability of urate-lowering drugs in gout: a randomised controlled trial of benzbromarone versus probenecid after failure of allopurinol. Ann. Rheum. Dis. 68, 51–56 (2009). 106. Scott, J. T. Comparison of allopurinol and probenecid. Ann. Rheum. Dis. 25, 623–626 (1966). 107. Reinders, M. K., van Roon, E. N., Houtman, P. M., Brouwers, J. R. Jansen, T. L. Biochemical effectiveness of allopurinol and allopurinol- probenecid in previously benzbromarone-treated gout patients. Clin. Rheumatol. 26, 1459–1465 (2007). 108. Lee, M. H., Graham, G. G., Williams, K. M. Day, R. O. A benefit-risk assessment of benzbromarone in the treatment of gout. Was its withdrawal from the market in the best interest of patients? Drug Saf. 31, 643–665 (2008). 109. Zhu, X. et al. Efficacy and safety of losartan in treatment of hyperuricemia and posttransplantation erythrocytosis: results of a prospective, open, randomized, case–control study. Transplant. Proc. 41, 3736–3742 (2009). 110. Huang, H. Y. et al. The effects of vitamin C supplementation on serum concentrations of uric acid: results of a randomized controlled trial. Arthritis Rheum. 52, 1843–1847 (2005). 111. Juraschek, S. P., Miller, E. R. 3rd Gelber, A. C. Effect of oral vitamin C supplementation on serum uric acid: a meta-analysis of randomized controlled trials. Arthritis Care Res. (Hoboken) 63, 1295–1306 (2011). 112. Bastow, M. D., Durrington, P. N. Ishola, M. Hypertriglyceridemia and hyperuricemia: effects of two fibric acid derivatives (bezafibrate and fenofibrate) in a double-blind, placebo-controlled trial. Metabolism 37, 217–220 (1988). 113. Stamp, L. K. et al. Clinically insignificant effect of supplemental vitamin C on serum urate in patients with gout. A pilot randomized controlled trial. Arthritis Rheum. 65, 1636–1642 (2013). 114. Sundy, J. S. et al. Efficacy and tolerability of pegloticase for the treatment of chronic gout in patients refractory to conventional treatment: two randomized controlled trials. JAMA 306, 711–720 (2011). 115. Ganson, N. J., Kelly, S. J., Scarlett, E., Sundy, J. S. Hershfield, M. S. 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. 8, R12 (2006). 116. Krystexxa®( pegloticase) prescribing information. Savient Pharmaceuticals Inc. [online], http://krystexxa.com/hcp/pdf/ KRYSTEXXA_Prescribing_Information.pdf (2014). 117. Malaguarnera, M. et al. A single dose of rasburicase in elderly patients with hyperuricaemia reduces serum uric acid levels and improves renal function. Expert Opin. Pharmacother. 10, 737–742 (2009). 118. De Angelis, S. et al. Is rasburicase an effective alternative to allopurinol for management of hyperuricemia in renal failure patients? A double blind-randomized study. Eur. Rev. Med. Pharmacol. Sci. 11, 179–184 (2007). 119. Pui, C. H. et al. Recombinant urate oxidase for the prophylaxis or treatment of hyperuricemia in patients with leukemia or lymphoma. Clin. Oncol. 19, 697–704 (2001). 120. Richette, P., Brière, C., Hoenen-Clavert, V., Loeuille, D. Bardin, T. Rasburicase for tophaceous gout not treatable with allopurinol: an exploratory study. J. Rheumatol. 34, 2093–2098 (2007). 121. Lasko, B. et al. RDEA594, a novel uricosuric agent, significantly reduced serum urate levels and was well tolerated in a phase 2a pilot study in hyperuricemic gout patients [abstract]. Arthritis Rheum. 60 (Suppl. 10), 1105 (2009). 122. Becker, M. et al. BCX4208 combined with allopurinol increases response rates in patients with gout who fail to reach goal range serum urate on allopurinol alone: a randomized, double- blind, placebo-controlled trial [L10]. Presented at the 2011 ACR/ARHP Annual Scientific Meeting. 123. Li-Yu, J. et al. Treatment of chronic gout. Can we determine when urate stores are depleted enough to prevent attacks of gout? J. Rheumatol. 28, 577–580 (2001). 124. de Oliveira, E. P. Burini, R. C. High plasma uric acid concentration: causes and consequences. Diabetol. Metab. Syndr. 4, 12 (2012). 125. Perez-Ruiz, F., Herrero-Beites, A. M. Carmona, L. A two-stage approach to the treatment of hyperuricemia in gout: the “dirty dish” hypothesis. Arthritis Rheum. 63, 4002–4006 (2011). 126. Borstad, G. C. et al. Colchicine for prophylaxis of acute flares when initiating allopurinol for chronic gouty arthritis. J. Rheumatol. 31, 2429–2432 (2004). 127. Wortmann, R., MacDonald, P., Hunt, B. Jackson, R. Effect of prophylaxis on gout flares after the initiation of urate-lowering therapy: analysis of data from three phase III trials. Clin. Ther. 32, 2386–2397 (2010). 128. Mikuls, T. R., Farrar, J. T., Bilker, W. B., Fernandes, S. Saag, K. G. Suboptimal physician adherence to quality indicators for the management of gout and asymptomatic hyperuricaemia: results from the UK General Practice Research Database (GPRD). Rheumatology (Oxford) 44, 1038–1042 (2005). 129. Roddy, E., Zhang, W. Doherty, M. Concordance of the management of chronic gout in a UK primary-care population with the EULAR gout recommendations. Ann. Rheum. Dis. 66, 1311–1315 (2007). 130. Doherty, M. et al. Gout: why is this curable disease so seldom cured? Ann. Rheum. Dis. 71, 1765–1770 (2012). 131. Lioté, F. Choi, H. Managing gout needs more than drugs: ‘Il faut le savoir-faire, l’Art et la manière’. Ann. Rheum. Dis. 72, 791–793 (2013). 132. Harrold, L. R. et al. Primary care providers’ knowledge, beliefs and treatment practices for gout: results of a physician questionnaire. Rheumatology (Oxford) 52, 1623–1629 (2013). 133. Owens, D., Whelan, B. McCarthy, G. A survey of the management of gout in primary care. Ir. Med. J. 101, 147–149 (2008). 134. General Medical Council. Tomorrow’s doctors: outcomes and standards for undergraduate medical education (General Medical Council, London, 2009). 135. British National Formulary. BNF [online], http://www.bnf.org/bnf/index.htm (2014). Author contributions All authors made substantial contributions to discussions of content, writing, and reviewing/editing the manuscript before submission. REVIEWS © 2014 Macmillan Publishers Limited. All rights reserved