You are on page 1of 9

Rheumatology 2015;54:20–28

RHEUMATOLOGY doi:10.1093/rheumatology/keu237
Advance Access publication 14 August 2014

Review
Psoriatic arthritis: current therapy and future
approaches
DoQuyen Huynh1 and Arthur Kavanaugh2

Abstract

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
PsA is a systemic inflammatory condition that affects 20–30% of patients with psoriasis. It is characterized
by potential involvement of diverse tissues, including peripheral and axial joints, enthesitis, dactylitis
and skin and nail disease. The degree of involvement in each domain can vary over time in individual
patients and can differ substantially between PsA patients. The clinical heterogeneity along with the
varying extent of severity and activity can pose significant challenges to treatment. Although some studies
had suggested immunopathophysiological similarities between PsA and RA, more recently important
R EV I E W

distinctions have been defined. Similarly, although some immunomodulatory therapies have proved ef-
fective for both PsA and RA, recent data suggest distinct responses to certain targeted therapies. Herein,
current DMARDs and biologic agents as well as the potential role of emerging therapeutics will be
reviewed.
Key words: psoriatic arthritis, tumour necrosis factor inhibitors, disease-modifying anti-rheumatic drug, treat-
ment, review, ustekinumab, apremilast, IL-17 inhibitor, rituximab, methotrexate.

Clinical features and diagnosis reactants, such as ESR or CRP, however, many patients
with active disease will have normal levels. The majority
PsA is a chronic inflammatory arthritis that occurs in pa- of PsA patients are seronegative, in that serum tests for
tients with psoriasis. Recent studies have suggested that RF and ACPA are negative; however, there is a greater
20–30% of patients with psoriasis may have PsA [1]. Skin prevalence of RF/ACPA positivity among PsA patients
manifestations precede the arthritis in >80% of PsA compared with the general population [3]. The older
patients, at times by a decade or more. In addition to Moll and Wright criteria to classify PsA have been largely
synovitis, other manifestations common in PsA include supplanted by the Classification of Psoriatic Arthritis
enthesitis, dactylitis and anterior uveitis or iritis. (CASPAR) classification criteria, which have been shown
Gastrointestinal involvement with a resemblance to IBD to have high sensitivity and specificity in diverse
is common, although not always clinically apparent. The settings [4].
peak age at onset of PsA is between 30 and 50 years of
age. There is equal distribution between men and women Treatment guidelines and paradigms
with the exception of axial disease, which favours men
3:1. There are significant genetic associations common The clinical heterogeneity of PsA poses a challenge to
to psoriasis and PsA, some of which overlap with other clinicians in selecting the most appropriate treatments.
autoimmune conditions [2]. To help address this, international groups such as the
The diagnosis of PsA is clinical as there are no pathog- Group for Research and Assessment of Psoriasis and
nomonic, serological, imaging or other diagnostic tests. Psoriatic arthritis (GRAPPA) and the European League
Some patients may have elevations in acute phase Against Rheumatism (EULAR) have published treatment
recommendations [5, 6], as have some individual coun-
tries. The GRAPPA recommendations consider five do-
1
Division of Rheumatology and 2Division of Rheumatology, Allergy and mains of involvement (peripheral arthritis, skin and nail
Immunology, University of California, San Diego, La Jolla, CA, USA. involvement, enthesitis, dactylitis and axial arthritis) and
Submitted 3 December 2013; revised version accepted 9 April 2014. use a grid approach to account for various levels of dis-
Correspondence to: DoQuyen Huynh, Division of Rheumatology, ease activity and severity. The EULAR recommendations
University of California, San Diego, Fellow 9444 Medical Center Drive,
Suite 0943, La Jolla, CA 92093-0943, USA. E-mail: d4huynh@ucsd.edu employ an algorithmic approach that focuses mainly on
or doquyen9@gmail.com musculoskeletal manifestations, specifically peripheral

! The Author 2014. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For Permissions, please email: journals.permissions@oup.com
Psoriatic arthritis: treatment

arthritis; manifestations such as dactylitis, enthesitis and suggested that overall NSAIDs may be used safely with
skin and nail involvement were to be considered separ- MTX without an increased risk of toxic adverse events or
ately. With PsA research proceeding apace, inclusion of liver, renal or pulmonary dysfunction [12]. Also, a placebo-
newer therapies into clinical guidelines requires ongoing controlled study of nimesulide, a selective cyclooxygen-
modification and updating. ase 2 (COX-2) inhibitor, did result in improved signs and
Other therapeutic concepts are also being explored in symptoms of PsA and, contrary to prior suggestions,
PsA. Central to any assessment of the efficacy of thera- NSAID use did not worsen skin disease [13].
pies are the outcome measures used to quantify disease Systemic and IA steroids are commonly used for PsA,
activity across various domains of disease, both in clinical although again there are limited data confirming their util-
studies and in the clinic. As many of the outcome meas- ity specifically in PsA [14, 15]. Nevertheless, it should be
ures used in PsA are borrowed from other conditions, noted that between 40% and 50% of patients in clinical
such as assessment of peripheral synovitis as has long trials are or have previously been on oral steroids. One
been done for RA, there has been substantial effort in small study compared the use of early IA steroid injection

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
devising and validating PsA-specific outcome measures vs conservative management with NSAIDs in early PsA
[7, 8]. A recent review of the various outcomes measures patients presenting with oligoarticular disease. In this
has been performed [9];. where there is no single univer- study, patients also received SSZ if synovitis persisted
sally accepted measure, in practice, minimal disease ac- or if polyarticular disease occurred. By week 52, 81% of
tivity has become to be considered an acceptable target patients on the IA steroid achieved complete response,
for treatment. defined by the absence of synovitis, compared with only
Newer treatment paradigms, some of which have also 51% in the conservative group. It should be noted that
been developed in some rheumatic and other diseases, SSZ was started in a greater percentage of the steroid
are also being assessed in PsA. Further, there has been group (45%) as compared with the conservative group
growing interest in the concept of treat to target in PsA. In (14%) and the study contained a mixture of patients with
RA, data have shown that regular assessments of disease oligoarticular arthritis, of whom few had an established
activity and alteration of therapies aimed at achieving the diagnosis of PsA [16].
lowest levels of disease activity possible are associated
with optimal benefits in clinical response, joint damage SSZ
and functional ability. The Tight Control of Psoriatic Among DMARDs, SSZ has one of the larger bodies of
Arthritis (TICOPA) study addressed this, with PsA patients evidence specifically in PsA. In a placebo-controlled trial
randomized 1:1 to receive either (i) standard of care with of 221 PsA patients there was improvement in peripheral
changes in medication and dose escalation to be deter- arthritis in 59% of SSZ-treated patients compared with
mined by a physician every 12 weeks or (ii) a 4-week 47% of controls; however, the overall effect size was
follow-up with dose escalation and medication changes small [17]. Similarly a smaller double-blind randomized
as outlined by a protocol aimed at minimal disease activity controlled trial (RCT) suggested that the use of SSZ pro-
[10]. PsA patients under tight control achieved signifi- vided improvement in functional outcomes and to a lesser
cantly better clinical outcomes in ACR 20%, 50% and degree skin disease [18]. Unfortunately there is no docu-
70% responses (ACR20/50/70) of 62%, 51% and 38% mented benefit of SSZ with regard to inhibition of joint
at 48 weeks compared with 45%, 25% and 17% in the damage as assessed by radiographic progression [19].
standard care group [11]. Overall skin improvement as
measured by the Psoriasis Area and Severity Index 75 LEF
(PASI75) was reached in 59% of the tight control group
LEF, an inhibitor of pyrimidine synthesis, was studied in a
compared with only 33% in the standard care group. multinational double-blind RCT called the Treatment of
However, there were more adverse effects noted in the PsA Study (TOPAS) [20]. Overall, 58.9% of the LEF-trea-
tight control group. Further analysis may elucidate the ted patients were clinical responders, compared with
risk/benefit considerations of treat to target in PsA. 29.7% of placebo controls. LEF was superior to placebo
Lastly, another concept that has received considerable in functional status scores, CRP reduction and 75% im-
interest in RA and will be also assessed in PsA is whether provement in the PASI75. In a small retrospective study of
therapies such as biologic agents might be withdrawn patients who were considered non-responders to MTX,
for PsA patients achieving treatment goals, with clinical patients who received a combination of LEF and MTX
response being maintained. had improvements in their 28-joint DAS (DAS28) and
EULAR response criteria compared with those receiving
Common conventional treatment agents MTX monotherapy [21, 22]. The combination of MTX and
LEF can be associated with elevated liver function tests
NSAIDs and corticosteroids and requires careful monitoring [23].
NSAIDs and corticosteroids are often prescribed as part
of the management of inflammatory joint diseases, includ- CSA and tacrolimus
ing PsA. While concerns had been raised about their CSA and tacrolimus inhibit calcineurin, resulting in the in-
safety, a recent Cochrane review evaluating the safety of hibition of T lymphocyte activation. CSA has been shown
NSAIDs and paracetamol in inflammatory arthritides to be effective in improving both skin and joint

www.rheumatology.oxfordjournals.org 21
DoQuyen Huynh and Arthur Kavanaugh

involvement in a number of clinical studies in psoriasis Administration (FDA) for use in PsA: infliximab, etanercept,
and PsA [24]. CSA has also been shown to be effective ADA, golimumab and, most recently, certolizumab pegol
in combination with MTX and TNF inhibitor (TNFi). A (Table 1) [38–48]. Overall, efficacy among the agents ap-
double-blind RCT that compared combination pears comparable, although there have not been head-to-
CSA + MTX with MTX or placebo in PsA showed signifi- head trials. An indirect comparison reviewing data from
cant improvement in CRP, PASI, swollen joint count and RCTs of the first four approved TNFis, and focusing on
US-defined synovitis with combination CSA + MTX [25]. changes in skin, peripheral joint and axial involvement,
Combinations of CSA with TNFi have also shown positive suggested that there were no significant differences be-
results in an open label prospective study of 160 patients tween the TNFis in PsA [49, 50]. Studies of certolizumab
receiving monotherapy CSA, monotherapy adalimumab pegol, the most recently approved agent, included pa-
(ADA) or combination CSA + ADA. PsA patients on com- tients who had previously been treated with other TNFis,
bination CSA + ADA had statistically significant improve- and showed efficacy comparable to TNFi-naive patients
ments in Psoriatic Arthritis Response Criteria (PsARC),

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
[46]. This suggests that TNFi switching may be viable in
ACR50 and HAQ assessments at 12 months compared PsA, as has been established in RA. Data supporting this
with those on monotherapy [26], although there was little concept also come from registries. In the Norwegian
difference in PASI outcomes between the groups. The DMARD (NOR-DMARD) registry, the durability of
popularity of CSA use has been tempered by the need
for close monitoring due to concerns about renal toxicity
and hypertension, especially at doses >3 mg/kg/day.
TABLE 1 Summary of results of clinical trials of TNFis in
PsA
MTX
MTX is widely accepted as a cornerstone therapy in PsA,
Patients meeting
as reflected in both the GRAPPA and EULAR treatment reponse criteria, %
recommendations [27–29]. Despite its popularity there is a Study
paucity of studies assessing MTX use in PsA, and results Agent/trial size ACR20 PASI75
have been disparate. The MTX in PsA trial randomized
221 PsA patients to placebo or 15 mg of MTX weekly for Etanercept [38, 39]
Week 12 205 59 38
6 months [30]. The overall results suggested significant
Week 24 55 40
efficacy for MTX only in the physician global assessment
Week 48 169 64 40
and PASI, and the primary outcomes were not achieved. Infliximab/IMPACT [40, 47, 48]
However, a number of factors in study design may have Week 14 200 58 64
contributed to these results, including a high dropout rate, Week 24 54 60
a relatively low dose of MTX and inclusion of PsA patients Week 54 173 59 50
with less severe disease. Indeed, when analysis was re- Week 98 104 62 64
stricted to polyarticular patients, MTX was effective. ADA/ADEPT [41, 42]
A recent systematic analysis reviewed the five RCTs in Week 12 315 58 49
the medical literature and about a dozen other observa- Week 24 57 59
Week 48 245 59 59
tional studies [31]. Although disparate results were re-
Week 104 57 58
ported in the individual studies, in general it appeared Golimumab/GO-REVEAL [43–45]
that trials using weekly doses of MTX of 515 mg achieved 50 mg
clinical benefit, whereas those using smaller doses did Week 14 405 51 40
not. Further support for the efficacy of MTX comes from Week 24 52 56
the open-label RESPOND [32] study, in which 115 treat- Week 52 360 67 62
ment-naive PsA patients were randomized to receive Week 104 335 67 86
combination MTX + infliximab or MTX monotherapy at a 100 mg
dose of 15 mg. At 16 weeks, 86% of patients received Week 14 45 58
Week 24 61 66
an ACR20 response rate with combination therapy, as
Week 52 360 71 68
did 66% of patients on MTX monotherapy. Overall, des-
Week 104 335 70 86
pite the paucity of evidence, MTX remains a mainstay of Certolizumab/ 409
drug therapy in PsA. rapid-PsA [46]
200 mg
TNF inhibitors Week 12 58 47
Week 24 138 64 62
The introduction of biologic therapies with anti-TNFis has 400 mg
greatly improved the clinician’s ability to treat all of the Week 12 51 47
various manifestations of PsA. The clinical efficacy of Week 24 135 56 61
TNFis exceeds that of traditional DMARDs and TNFis
have been shown to significantly inhibit joint damage as TNFis: tumour necrosis factor inhibitors; ADA: adalimumab;
assessed by radiographic progression [33–37]. Currently ACR20: 20% response to ACR criteria; PASI75: Psoriasis
five TNFis are approved by the US Food and Drug Area and Severity Index 75.

22 www.rheumatology.oxfordjournals.org
Psoriatic arthritis: treatment

remaining on a second TNFi was less than that for the FIG. 1 Proposed immunopathogenesis in the skin
first TNFi course, but suggested that some patients will
respond to TNFi switching [52]. Similarly in DANBIO
(a nationwide registry of biologic therapies in Denmark),
retention rates on a second or even a third TNFi sug-
gest that switching TNFis can result in clinical
improvement [52].
Factors such as cost and patients’ concerns regarding
side effects over the long term have raised the question of
whether treatments might be tapered or even withdrawn
in patients achieving low levels of disease activity. In RA,
studies have suggested that such a strategy might be
viable for a subset of patients, particularly those with ear-

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
lier disease who achieve very low levels of disease activity
on treatment. In PsA, to date there has been only one
prospective case–control study addressing this. Dose re-
duction of ADA to 40 mg every 4 weeks was done in a
population that included 76 PsA patients who had ob-
tained clinical remission [53]. At the 28-week follow-up, Dendritic and T helper (Th17, Th1) cells secrete varying
88.6% of PsA patients remained in clinical remission; implicated cytokines that further increase and recruit
interestingly, only 17.6% of the RA patients in the study leucocyte activation and ultimately systemic inflammation.
remained in remission. Other studies addressing dose re- DC: dendritic cell.
duction and withdrawal are currently under way and may
help define the patient characteristics that predict suc-
cess on lower doses of specific agents. patients who had previously received treatment with
TNFis were included. Of note, the magnitude of clinical
Newer therapies and agents response was less than in TNF-naive patients [57].
Analysis of radiographic data from the combined
Despite advances in therapy, there remain patients who P-SUMMIT I and II databases showed inhibition of radio-
fail to respond to classic DMARDs and TNFis or who have graphic progression [58]. In the studies, ustekinumab was
loss of efficacy over time. Driven in part by this unmet generally safe and well tolerated.
clinical need, newer classes and targets of therapy have
emerged. This has also been facilitated by studies further
IL-17 inhibitors
advancing our understanding of the immunopathophysiol-
ogy of disease (Figs. 1 and 2). For example, one study IL-17, an inflammatory cytokine secreted by Th17 T cells
suggested a potentially key role for IL-23 in arthritides and other cells, has been identified in psoriatic plaques
characterized by enthesitis, including PsA [54]. Thus, in and inflamed entheses. Currently there are three IL-17 in-
an animal model of enthesitis, a specific subset T cell hibitors in advanced phase clinical trials, including secu-
residing in the enthesis, distinct from Th17 cells, pos- kinumab, ixekizumab and brodalumab. Secukinumab and
sessed the ability to respond to IL-23, leading to up- ixekizumab are mAbs against IL-17A, while brodalumab is
regulation of other inflammatory mediators, including a mAb directed against the IL-17 receptor A (IL-17RA).
IL-6, IL-17 and IL-22. Of note, this model had a resemb- Data for these agents have shown very robust improve-
lance to human disease, with enthesitis and entheseal ments in skin psoriasis. For example, in a phase IIb RCT of
new bone formation as well as inflammation in the aortic secukinumab at 150 and 75 mg, there was PASI75 im-
root and valve. provement of 81% and 57% of the patients, respectively,
compared with 9% for placebo at 12 weeks [59]. A dose-
Ustekinumab: IL-12/IL-23 inhibitor finding randomized study of ixekizumab also showed sig-
Ustekinumab is a human mAb directed against the p40 nificant PASI improvement in >77% of patients compared
subunit common to IL-12 and IL-23 that has received with 8% for placebo [60]. These authors also mention a
regulatory approval for psoriasis and PsA. In P-SUMMIT significant reduction in joint pain assessed by a visual
I, 615 PsA patients with no prior TNFi exposure were ran- analogue scale (VAS) in 12 patients who also carried the
domized 1:1:1 to receive placebo vs 45 mg or 90 mg of diagnosis of PsA. Similar to secukinumab and ixekizumab,
ustekinumab. ACR20/50/70 and PASI 75 responses were a phase II dose-finding trial of brodalumab showed early
examined at 24 weeks. In the 90 mg ustekinumab group improvement in PASI75 at 12 weeks compared with pla-
the ACR20/50/70 and PASI75 were 49.5%/27.9%/14.2% cebo (70 mg, 45%; 140 mg, 85.9%; 210 mg, 86.3%;
and 62.4%, whereas control responses were 22.8%/ 280 mg, 76%; placebo, 16%) [61].
8.7%/2.4% and 11%, respectively. There were also sig- In PsA, a small (n = 24) double-blind RCT of secukinu-
nificant improvements in the enthesitis, dactylitis and mab has demonstrated significant improvement in the
function as measured by the HAQ. Responses were main- HAQ disability index (HAQ-DI) and CRP; however, the pri-
tained up to 108 weeks [55, 56]. In P-SUMMIT II, PsA mary endpoint of ACR20 improvement was not met [62].

www.rheumatology.oxfordjournals.org 23
DoQuyen Huynh and Arthur Kavanaugh

FIG. 2 Proposed immunopathogenesis in PsA FIG. 3 Mechanism of action of apremilast and tofacitinib

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
Apremilast: In immune cells, Toll-like receptor 4 (TLR4)
activation leads to downstream signalling that activates
nuclear factor kappa B (NFkB, which increases tran-
scription of pro-inflammatory mediators such as IL-23 and
TNF-a. Simultaneously G protein coupled receptors
(GPCRs) activate adenylyl cyclase (AC), leading to ele-
vated levels of cyclic adenosine monophosphate (cAMP).
In cells such as monocytes, cAMP is degraded by
phosphodiesterase enzyme 4 (PDE4). Apremilast inhibits
PDE4, which leads to elevated levels of cAMP. High levels
of cAMP activate protein kinase A (pKa), leading to
phosphorylation of cAMP-responsive element binding
Local cytokines IL-17, IL-6 and IL-23 further increase in- transcriptions factors such as cAMP-responsive element
flammation in joint and entheseal complex. Localized in- binding protein (CREB), which can bind to promoter se-
flammation leads to activation of osteoclasts and erosion quences of anti-inflammatory genes such as IL-10. Both
formation. The presence of novel resident entheseal T TLR4 and GPCR downstream signalling require shared
cells expressing RORgt receptors respond to IL-23 acti- co-factors such as CREB binding protein (CBP) and p300.
vation and further secrete IL-22 and enhance IL-6. IL-22 Greater activation of the anti-inflammatory pathway by
may activate local osteoblast cells leading to bony prolif- apremilast may further decrease pro-inflammatory sig-
eration characteristically seen in PsA. OC: osteoclast; OB: nalling by recruiting important co-factors needed in acti-
osteoblast; RORgt: RAR-related orphan receptor; RANK: vation. Tofacitinib: Janus kinase (JAK) activation-specific
receptor activator of nuclear factor kappa B. cytokines such as IL-6 lead to phosphorylation of the
signal transducer and activator of transcription (STAT)
Interestingly, in RA, IL-17 inhibitors have also been stu- pathway, which mediates transcription of pro-inflamma-
died, and clinical responses appeared to be modest com- tory cytokines. Tofacitinib blocks JAK activity and therein
pared with therapies with other mechanisms of action. suppresses STAT signalling. IRAK: interleukin receptor-
Brodalumab has been studied in PsA as well [63]. Doses associated kinase; TRAF6: tumour necrosis factor recep-
of 140 mg and 280 mg given subcutaneously resulted in tor-associated factor 6; STAT3: signal transducer and
ACR20 responses at 12 weeks of 36.8% and 39.3%, re- activator of transcription 3.
spectively, compared with 18.2% for placebo. Additional
longer-term studies will be necessary to define the effect demonstrated significant ACR20 responses of 43.5%
of IL-17 inhibitors on the various manifestations of PsA. and 35.8%, respectively, compared with 11.8% for pla-
cebo controls at week 12 [64]. Results from phase III trials,
Apremilast: phosphodiesterase inhibitor dubbed PALACE 1, 2, 3 and 4, have also recently been
Elevated levels of cyclic adenosine monophosphate reported. In PALACE 1, 504 patients were randomized
(cAMP) down-regulate pro-inflammatory cytokines such 1:1:1 to receive placebo, apremilast 20 mg twice a day
as IL-12, IL-23, TNF-a and IFN-g (Fig. 3). Phosphodiester- or apremilast 30 mg twice a day [65]. At 16 weeks, 31%
ase enzymes that degrade cAMP, e.g. phosphodiesterase and 40% of apremilast 20 and apremilast 30 mg patients,
enzyme 4 (PDE4), have become a potential therapeutic respectively, achieved ACR20, compared with only 19%
target. The PDE4 inhibitor apremilast has been assessed on placebo. Long-term follow-up to week 52 in PALACE 4
in several studies in PsA patients. In a phase II study, revealed persistence of benefit, with 58% of patients
apremilast at a dose of 20 mg or 40 mg twice daily meeting ACR20, although fewer patients met ACR50 or

24 www.rheumatology.oxfordjournals.org
Psoriatic arthritis: treatment

70 and benefit to the skin was modest [66]. Overall, apre- disease while the remaining 4 had peripheral disease.
milast appeared to be well tolerated with minimal effects Other open-label studies also suggest quite modest im-
on any laboratory parameters. provement in PsA patients treated with RTX, although
some improvement may be due to concomitant steroid
Abatacept: CTLA-4 inhibitor use [75, 76].
T cells appear to play an important role in the pathogen-
esis in PsA. T cell activation is dependent on both specific Conclusion
antigen presentation and co-stimulation. CTLA-4 inhibits
CD28 interactions with CD80 and CD86 and thereby PsA is a chronic inflammatory condition with significant
down-regulates T cell activation. Abatacept, a fusion pro- heterogeneity in clinical manifestations, including skin
tein composed of the extracellular domain of CTLA-4 and nail psoriasis, enthesitis, dactylitis, axial arthritis and
linked to an IgG1 Fc portion, has proven efficacy in nu- peripheral arthritis. Among DMARDs, MTX is most com-
monly used, despite a paucity of high-quality studies sup-

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
merous studies in patients with RA [67, 68]. In PsA, the
results of a randomized double-blind placebo-controlled porting its use. LEF, ciclosporin and SSZ are all
trial evaluating varying doses over a 6-month period have efficacious for arthritis and psoriasis to varying degrees
been reported [69]. The results suggest abatacept-treated in some PsA patients. Unarguably, the class of anti-
patients, particularly those at a dose of 10 mg/kg, had a TNFis has established itself as most effective for all the
significantly higher percentage of ACR20 responses (48%) various manifestations of PsA, and in fact could be con-
compared with placebo (19%). Trends toward improve- sidered the standard of care, particularly in patients with
ment in erosion, osteitis and synovitis as seen on MRI severe, active disease. There remain unanswered ques-
and general improvement in physical function and quality tions concerning established therapies in PsA. Prominent
of life were observed. Unfortunately, the skin response among these is whether TNFi and DMARD combinations,
was inconsistent and patients previously exposed to especially with MTX, might be synergistic in PsA as they
TNFis had a less robust response than TNFi-naive pa- are in RA. To date, all trials of TNFis in PsA have allowed
tients. Further studies evaluating the potential efficacy of MTX, but have required active disease as an eligibility cri-
abatacept in PsA are currently ongoing. terion. Such a study design obviates the ability to assess
the additive or synergistic benefit of TNFi and MTX. As
Tocilizumab: IL-6 inhibitor noted, there is great interest in newer treatment
approaches, such as treat to target, and possibly tapering
Tocilizumab, a humanized mAb to the IL-6 receptor, has
or discontinuing therapy in patients achieving treatment
been approved for RA and it is being studied in PsA. Of
goals. Finally, pharmacoeconomic assessments specific
note, a randomized trial of tocilizumab in AS showed no
to PsA will help establish the value of currently available
clinical efficacy despite a decrease in CRP [70]. In a case
agents and optimize their use.
report of two patients with PsA, there was a reduction in
Success with established therapies has helped drive
CRP levels but no benefit in skin or joint symptoms, which
interest in newer therapies. Among the newer agents,
were alleviated subsequently with the use of anti-TNFis
the greatest amount of data to date are for the IL-12/
[71]. Currently there is only one documented case report
IL-23 inhibitor ustekinumab. This agent may have its
of tocilizumab improving tender joint count, swollen joint
main role in PsA patients in whom skin manifestations
count and patient global VAS in an atypical PsA patient
are particularly active or severe. On the horizon it appears
that had loss of efficacy on TNFis [72].
that apremilast may offer an effective option for PsA, par-
ticularly for patients with more moderate disease activity
Janus kinase (JAK) inhibitors
and in whom safety concerns might impact the choice of
Another target of drug therapy are the JAK molecules: other treatments. There has been interest in the effects in
JAK1, JAK2, JAK3 and TYK2. These intracellular mol- PsA of therapies already approved for use in RA.
ecules are important in signal transduction of cytokines Interestingly, some therapies effective in RA, such as
such as IL-2, IL-12 and IL-6 (Fig. 3). Tofacitinib was FDA RTX, seem not to be very effective in PsA. Abatacept
approved in late 2012 for the treatment of RA. Of note, it has some efficacy in PsA, but that too appears to be
has been shown to be effective in phase II trials of mod- less than its efficacy in RA. This suggests that whereas
erate to severe psoriasis at doses of 5 and 15 mg twice these disease states both respond to TNFis, differences in
daily at 12 weeks of therapy [73]. Further studies in PsA their immunopathogenesis may be revealed by distinct
are under way. responsiveness to other targeted therapies. In that
regard, although there are limited data currently available,
Rituximab: CD20 inhibitor it might be argued that the efficacy of IL-17 inhibition in
Rituximab (RTX), a chimeric mAb directed against the arthritis of PsA seems to be notably less than the re-
CD20, has been shown to be effective and is approved sponse of skin psoriasis. Currently the treatment outlook
for use in RA. Recently an open-label trial of RTX in 26 in PsA appears optimistic, with numerous studies under
patients who had either AS or PsA showed modest im- way. Undoubtedly these studies will allow the introduction
provement in 11 of 26 patients (47.8%) as measured by of additional agents and novel treatment strategies, and
the BASDAI [74]. Of note, 23 of the 26 patients had pre- ultimately help optimize the care of patients with this im-
vious exposure to TNFis. Of the 11 patients, 7 had axial portant condition.

www.rheumatology.oxfordjournals.org 25
DoQuyen Huynh and Arthur Kavanaugh

Rheumatology key messages 13 Sarzi-Puttini P, Santandrea S, Boccassini L, Panni B,


. PsA is a chronic inflammatory disease treated by Caruso I. The role of NSAIDs in psoriatic arthritis: evidence
the use of DMARDs and TNF inhibitors. from a controlled study with nimesulide. Clin Exp
. Newer agents to treat PsA show promise and clin- Rheumatol 2001;19(Suppl 22):S17–20.
ical trials of agents with different mechanisms of 14 Brenner M, Molin S, Ruebsam K et al. Generalized pus-
activity are under way. tular psoriasis induced by systemic glucocorticosteroids:
four cases and recommendations for treatment. Br J
Dermatol 2009;161:964–6.
Disclosure statement: A.K. has conducted clinical re- 15 Eder L, Chandran V, Ueng J et al. Predictors of response
search activities for AbbVie, Amgen, Janssen, UCB, to intra-articular steroid injection in psoriatic arthritis.
Pfizer, Roche and Sanofi. The other author has declared Rheumatology 2010;49:1367–73.
no conflicts of interest. 16 Marzo-Ortega H, Green MJ, Keenan AM et al. A rando-

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
mized controlled trial of early intervention with intra ar-
ticular corticosteroids followed by sulfasalazine versus
References conservative treatment in early oligoarthritis. Arthritis Care
Res 2007;57:154–60.
1 Ceponis A, Kavanaugh A. Use of methotrexate in patients
with psoriatic arthritis. Clin Exp Rheumatol 2010;28(Suppl 17 Clegg DO, Reda DJ, Abdellatif M. Comparison of
61):s132–7. sulfasalazine and placebo for the treatment of axial
2 Chandran V, Gladmann DD. Update on biomarkers in and peripheral articular manifestations of the seronega-
psoriatic arthritis. Curr Rheumatol Rep 2010;12:288–94. tive spondyloarthopathies. Arthritis Rheum 1999;25:
1957–61.
3 Vander CB, Hoffman IE, Zmierczak H et al. Anti-citrulli-
nated peptide antibodies may occur in patients with 18 Gupta AK, Grober JS, Hamilton TA. Sulfasalazine therapy
psoriatic arthritis. Ann Rheum Dis 2005;64:1145–9. for psoriatic arthritis: a double blind, placebo controlled
trial. J Rheumatol 1995;22:894–8.
4 Taylor W, Gladman D, Helliwell P et al. Classification cri-
teria for psoriatic arthritis; development of new criteria 19 Rahman P, Gladman DD, Cook RJ, Zhou Y, Young G. The
from a large international study. Arthritis Rheum 2006;54: use of sulfasalazine in psoriatic arthritis: a clinic experi-
2665–73. ence. J Rheumatol 1998;25:1957–61.

5 Gossec L, Smolen JS, Gaujoux-Viala C et al. European 20 Jaimes-Hernandez J, Melendez-Mercado CI, Mendoza-
League Against Rheumatism recommendations for the Fuentes A et al. Efficacy of leflunomide 100 mg weekly
management of psoriatic arthritis with pharmacological compared to low dose methotrexate in patients with active
therapies. Ann Rheum Dis 2012;71:4–12. rheumatoid arthritis: double blind, randomized clinical trial.
Rheumatol Clin 2012;8:243–9.
6 Ritchlin CT, Kavanaugh A, Gladman DD et al. Treatment
recommendations for psoriatic arthritis. Ann Rheum Dis 21 Behrens F, Finkenwirth C, Pavelka K et al. Leflunomide in
2009;68:1387–94. psoriatic arthritis: results from a large European pro-
spective observational study. Arthritis Care Res 2013;65:
7 Healy PJ, Helliwell PS. Measuring clinical enthesitis in
464–70.
psoriatic arthritis: assessment of existing measures and
development of an instrument specific to psoriatic arth- 22 Sakellariou GT, Sayegh FE, Anastasilakis AD,
ritis. Arthritis Rheum 2008;59:686–91. Kapetanos GA. Leflunomide addition in patients with ar-
ticular manifestations of psoriatic arthritis resistant to
8 Palominos PE, Gaujoux-Viala C et al. Clinical outcomes in
methotrexate. Rheumatol Int 2013;33:2917–20.
psoriatic arthritis: a systematic literature review. Arthritis
Care Res 2012;64:397–406. 23 Curtis JR, Beukelman T, Onofrei A et al. Elevated liver
enzyme tests among patients with rheumatoid arthritis or
9 Wajed J, Helliwell P, Korendowych E et al. Disease as-
psoriatic arthritis treated with methotrexate and or leflu-
sessment in psoriatic spectrum: a modular approach for
nomide. Ann Rheum Dis 2012;69:43–7.
use in routine clinical practice. Ann Rheum Dis 2014;73:
318–9. 24 Salvarani C, Macchioni P, Olivieri I et al. A comparison of
cyclosporine, sulfasalazine, and symptomatic therapy in
10 Coates LC, Navarro-Coy N, Brown SR et al. The TICOPA
the treatment of psoriatic arthritis. J Rheumatol 2001;28:
protocol (TIght COntrol of Psoriatic Arthritis): a rando-
2274–82.
mized controlled trial to compare intensive management
versus standard care in early psoriatic arthritis. BMC 25 Fraser AD, van Kuijk AWR, Westhovens R et al. A rando-
Musculoskelet Disord 2012;21:101. mized double blind, placebo controlled, multicenter trial of
combination therapy with methotrexate plus cyclosporine
11 Coates LC, Moverly A, McParland L et al. Results of a
in patients with active psoriatic arthritis. Ann Rheum Dis
randomized control trial comparing tight control of psori-
2005;64:859–64.
atic arthritis (TICOPA) with standard of care: tight control
improves outcome. ACR meeting, San Diego, 2013. 26 Karanikolas GN, Koukli EM, Katsalira A et al. Adalimumab
Abstract 814. or cyclosporine as monotherapy and in combination in
12 Colebatch AN, Marks JL, van der Heijde DM, Edwards CJ. severe psoriatic arthritis: results from a prospective 12-
Safety of nonsteroidal anti-inflammatory drugs and/or month nonrandomized unblinded clinical trial. J
paracetamol in people receiving methotrexate for inflam- Rheumatol 2011;38:2466–74.
matory arthritis: a Cochrane systematic review. J 27 Chandran V, Schentag CT, Gladman DD. Reappraisal
Rheumatol Suppl 2012;90:62–73. of the effectiveness of methotrexate in psoriatic

26 www.rheumatology.oxfordjournals.org
Psoriatic arthritis: treatment

arthritis: results from a longitudinal observational cohort. J 44 Kavanaugh A, van der Heijde D, McInnes I et al.
Rheumatol 2008;35:469–71. Golimumab in psoriatic arthritis: one-year clinical efficacy,
28 Scarpa R, Peluso R, Atteno M et al. The effectiveness of a radiographic, and safety results from a randomized, pla-
traditional therapeutic approach in early psoriatic arthritis: cebo-controlled trial. Arthritis Rheum 2012;64:2504–17.
results of a pilot randomized 6month trial with metho- 45 Kavanaugh A, McInnes I, Mease P et al. Clinical efficacy,
trexate. Clin Rheumatol 2008;27:823–6. radiographic and safety findings through 2 years of goli-
29 Ritchlin CT, Kavanaugh A, Gladman DD et al. Treatment mumab treatment in patients with active psoriatic arthritis:
recommendations for psoriatic arthritis. Ann Rheum Dis results from a long-term extension of the randomized
2009;68:1387–94. placebo-controlled GO-REVEAL study. Ann Rheum Dis
2013;72:1777–85.
30 Kingsley GH, Kowalczyk A, Taylor H et al. A randomized
placebo-controlled trial of methotrexate in psoriatic arth- 46 Mease PJ, Fleischmann R, Deadhar AA et al. Effect of
ritis. Rheumatology 2012;51:1368–77. certolizumab pegol on signs and symptoms in patients
with psoriatic arthritis: 24 week results of a phase 3

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
31 Ceponis A, Kavanaugh A. Use of methotrexate in patients
double-blind randomized placebo controlled study
with psoriatic arthritis. Clin Exp Rheumatol 2010;28(Suppl
(RAPID-PsA). Ann Rheum Dis 2014;73:233–7.
61):S132–7.
47 Antoni C, Krueger GG, de Vlam K et al. Infliximab improves
32 Baranauskaite A, Raffayova H, Kungurov NV et al.
signs and symptoms of psoriatic arthritis: results of the
Infliximab plus methotrexate is superior to methotrexate
IMPACT 2 trial. Ann Rheum Dis 2005;64:1150–7.
alone in the treatment of psoriatic arthritis in methotrex-
ate-naive patients: the RESPOND study. Ann Rheum Dis 48 Kavanaugh A, Krueger GG, Beutler A et al. Infliximab
2012;71:541–8. maintains a high degree of clinical response in patients
with active psoriatic arthritis through 1 year of treatment:
33 Ceponis A, Kavanaugh A. Treatment of psoriatic arthritis
results from the IMPACT2 trial. Ann Rheum Dis 2007;66:
with biological agents. Semin Cutan Med Surg 2010;29:
498–505.
56–62.
49 Atteno M, Peluso R, Costa L et al. Comparison of effect-
34 Boehncke WH, Prinz J, Gottlieb A. Biologic therapies for
iveness and safety of infliximab, etanercept and adalimu-
psoriasis. A systematic review. J Rheumatol 2006;33:
mab in psoriatic arthritis patients who experienced an
1147–51.
inadequate response to previous disease-modifying anti-
35 Salvarani C, Pipitone N, Catanoso M et al. Adalimumab in rheumatic drugs. Clin Rheumatol 2010;29:399–403.
psoriatic arthritis. J Rheumatol 2012;39(Suppl 89):77–81.
50 Thorlund K, Druyts E, Avina-Zubieta JA, Mills EJ. Anti-
36 Spadaro A, Lubrano E, Ferrara NM, Scarpa R. Etanercept tumor necrosis factor (TNF) drugs for the treatment of
in psoriatic arthritis. J Rheumatol 2012;39(Suppl 89):74–6. psoriatic arthritis: an indirect comparison meta-analysis.
37 Cantini F, Niccoli L, Nannini C, Kaloudi O, Cassara E. Biologics 2012;6:417–27.
Infliximab in psoriatic arthritis. J Rheumatol 2012;39(Suppl 51 Fagerli K, Lie E, van der Heijde D et al. Switching
89):71–3. between TNF inhibitors in psoriatic arthritis: data
38 Mease PJ, Goffe BS, Metz J et al. Etanercept treatment of from the NOR-DMARD study. Ann Rheum Dis 2013;72:
psoriatic arthritis and psoriasis: a randomized trial. Lancet 1840–4.
2000;356:385–90. 52 Glintborg B, Ostergaard M, Krogh NS et al. Clinical re-
39 Mease PJ, Kivitz AJ, Burch FX et al. Continued inhibition of sponse, drug survival, and predictors thereof among 548
radiographic progression in patients with psoriatic arthritis patients with psoriatic arthritis who switched tumor ne-
following 2 years of treatment with etanercept. J crosis factor a inhibitor therapy: results from the Danish
Rheumatol 2006;33:712–21. Nationwide DANBIO registry. Arthritis Rheum 2013;65:
40 Antoni C, Kavanaugh A, van der Heijde D et al. Two-year 1213–23.
efficacy and safety of infliximab treatment in patients with 53 Cantini F, Niccoli L, Cassara E, Kaloudi O, Nannini C.
active psoriatic arthritis: findings of the Infliximab multi- Sustained maintenance of clinical remission after adali-
national psoriatic arthritis controlled trial (IMPACT). J mumab dose reduction in patients with early psoriatic
Rheumatol 2008;35:869–76. arthritis: a long term follow up study. Biologics 2012;6:
41 Mease PJ, Ory P, Sharp JT et al. Adalimumab for long- 201–6.
term treatment of psoriatic arthritis: 2 year data from the 54 Sherlock P, Joyce-Shaikh B, Turner S et al. IL-23 induces
adalimumab effectiveness in Psoriatic Arthritis Trial spondylarthropathy by acting on ROR-gt+
(ADEPT). Ann Rheum Dis 2009;68:702–9. CD3+CD4 CD8 entheseal resident T cells. Nat Med
42 Mease PJ, Gladman DD, Ritchlin CT et al. Adalimumab for 2012;18:1069–77.
the treatment of patients with moderately to severely 55 McInnes I, Kavanaugh A, Gottlieb A et al. Ustekinumab in
active psoriatic arthritis: results of a double-blind, rando- patients with active psoriatic arthritis: results of the phase
mized, placebo-controlled trial. Arthritis Rheum 2005;52: 3, multicenter, double-blind, placebo-controlled
3279–89. PSUMMIT I study. Ann Rheum Dis 2012;71(Suppl):
43 Kavanaugh A, McInnes I, Mease P et al. Golimumab, a S107–48.
new human tumor necrosis factor alpha antibody, ad- 56 McInnes IB, Kavanaugh A, Gottlieb AB et al. Efficacy and
ministered every four weeks as a subcutaneous injection safety of ustekinumab in patients with active psoriatic
in psoriatic arthritis: twenty-four-week efficacy and safety arthritis: 1 year results of the phase 3 multicentre, double
results of a randomized, placebo-controlled study. blind, placebo-controlled PSUMMIT 1 trial. Lancet 2013;
Arthritis Rheum 2009;60:976–86. 382:780–9.

www.rheumatology.oxfordjournals.org 27
DoQuyen Huynh and Arthur Kavanaugh

57 Kavanaugh A, Puig L, Gottlieb A et al. Efficacy and safety abatacept in patients with rheumatoid arthritis receiving
of ustekinumab in patients with active psoriatic arthritis: 2 background methotrexate: a 5 year extended phase IIB
year results from a phase 3, multicenter, double-blind, study. J Rheumatol 2009;36:736–42.
placebo-controlled study. ACR meeting, San Diego, 2013. 68 Schiff M, Weinblatt ME, Valente R et al. Head-to-head
Abstract L10. comparison of subcutaneous abatacept versus adalimu-
58 McInnes I, Ritchlin C, Rahman P. Ustekinumab is effective mab for rheumatoid arthritis: two-year efficacy and
in inhibiting radiographic progression in patients with safety findings from AMPLE trial. Ann Rheum Dis 2014;73:
active psoriatic arthritis: integrated data analysis of two 86–94.
phase 3, randomized, placebo-controlled studies. ACR 69 Mease P, Genovese MC, Gladstein G et al. Abatacept in
meeting, San Diego, 2013. Abstract 1695. the treatment of patients with psoriatic arthritis: results of
59 Papp KA, Langley RG, Sigurgeirsson MA et al. Efficacy a six-month, multicenter, randomized double-blind pla-
and safety of secukinumab in the treatment of moderate- cebo controlled, phase II trial. Arthritis Rheum 2011;63:
to-severe plaque psoriasis: a randomized, double-blind, 939–48.

Downloaded from http://rheumatology.oxfordjournals.org/ at Universidad Nacional Autonoma de Mexico on August 19, 2015
placebo-controlled phase II does-ranging study. Br J 70 Sieper J, Porter-Brown B, Thompson L, Harari O,
Dermatol 2013;168:412–21. Dougados MJ. Tocilizumab (TCZ) is not effective for
60 Leonardi C, Matheson R, Zachariae C et al. Anti-interleu- the treatment of ankylosing spondylitis (AS): results of
kin-17 monoclonal antibody ixekizumab in chronic plaque phase II international, multicenter, randomized, double-
45. psoriasis. N Engl J Med 2012;366:1190–9. blind, placebo-controlled trial. Ann Rheum Dis 2012;
61 Papp K, Leonardi C, Menter A et al. Brodalumab, an anti- 1(Suppl 3):110.
interleukin-17-receptor antibody for psoriasis. N Engl J 71 Ogata A, Umegaki N, Katavama I, Kumanogo A, Tanaka T.
Med 2012;366:1181–9. Psoriatic arthritis in two patients with an inadequate re-
62 McInnes IB, Sieper J, Braun J et al. Efficacy and safety of sponse to treatment with tocilizumab. Joint Bone Spine
secukinumab, a fully human anti-interleukin-17A mono- 2012;79:85–7.
clonal antibody, in patients with moderate-to-severe 72 Hughes M, Chinoy H. Successful use of tocilizumab in a
psoriatic arthritis: a 24-week, randomized, double blind, patient with psoriatic arthritis. Rheumatology 2013;52:
placebo controlled, phase II proof of concept trial. Ann 1728–9.
Rheum Dis 2014;73:349–56.
73 Mamolo C, Harness J, Tan H, Menter A. Tofacitinib (CP-
63 Genovese M, Mease P, Greenwald M et al. Clinical re- 690,550), an oral Janus kinase inhibitor, improves patient-
sponse to brodalumab, an anti-interleukin-17 receptor reported outcomes in phase 2b, randomized, double-
antibody, in subjects with psoriatic arthritis. ACR meeting, blind, placebo-controlled study in patients with moderate-
San Diego, 2013. Abstract 817. to-severe psoriasis. J Eur Acad Dermatol Venereol 2013;
64 Schett G, Wollenhautpt J, Papp K et al. Oral apremilast in 28:192–203.
the treatment of active psoriatic arthritis: results of a 74 Wendling D, Dougados M, Berenbaum F et al. A nation-
multicenter, randomized, double-blind, placebo-con- wide series: data from the AIR registry of the French
trolled study. Arthritis Rheum 2012;64:3156–67. Society of Rheumatology. J Rheumatol 2012;39:2327–31.
65 Kavanaugh A, Mease PJ, Adebajo AO et al. Long term (52 75 Mease P, Kavanaugh A, Genovese M et al. Rituximab in
week) results of a phase 3, randomized controlled trial of psoriatic arthritis provides modest clinical improvement
apremilast, an oral phosphodiesterase 4 inhibitor, in pa- and reduces expression of inflammatory biomarkers in
tients with psoriatic arthritis. Ann Rheum Dis 2013; skin lesions [abstract]. Arthritis Rheum 2010;62(Suppl):
72(Suppl 3):163. S818.
66 Wells A, Edwards CJ, Adebajo A et al. Apremilast in the 76 Song IH, Heldmann F, Rudwaleit M et al. Different re-
treatment of DMARD naı̈ve psoriatic arthritis patients: re- sponse to rituximab in tumor necrosis factor blocker-naive
sults of a phase 3 randomized controlled trial (PALACE 4). patients with active ankylosing spondylitis and in patients
ACR meeting, San Diego, 2013. Abstract L4. in whom tumor necrosis factor blockers have failed: a
67 Westhovens R, Kremer J, Moreland L et al. Safety and twenty-four week clinical trial. Arthritis Rheum 2012;62:
efficacy of the selective costimulation modulator 1290–7.

28 www.rheumatology.oxfordjournals.org

You might also like