You are on page 1of 19

Journal of Dermatological Treatment

ISSN: 0954-6634 (Print) 1471-1753 (Online) Journal homepage: https://www.tandfonline.com/loi/ijdt20

Psoriatic arthritis for dermatologists

Alice Gottlieb & Joseph F. Merola

To cite this article: Alice Gottlieb & Joseph F. Merola (2020) Psoriatic arthritis
for dermatologists, Journal of Dermatological Treatment, 31:7, 662-679, DOI:
10.1080/09546634.2019.1605142

To link to this article: https://doi.org/10.1080/09546634.2019.1605142

© 2019 The Author(s). Published with


license by Taylor & Francis Group, LLC

Published online: 07 May 2019.

Submit your article to this journal

Article views: 13159

View related articles

View Crossmark data

Citing articles: 21 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ijdt20
JOURNAL OF DERMATOLOGICAL TREATMENT
2020, VOL. 31, NO. 7, 662–679
https://doi.org/10.1080/09546634.2019.1605142

REVIEW ARTICLE

Psoriatic arthritis for dermatologists


Alice Gottlieba and Joseph F. Merolab
a
Department of Dermatology, Icahn School of Medicine at Mt Sinai, New York, NY, USA; bDepartment of Medicine, Division of Rheumatology
and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA

ABSTRACT ARTICLE HISTORY


Psoriatic arthritis (PsA) affects up to one-third of patients with psoriasis. It is the major comorbidity of Received 27 March 2019
psoriasis because of the likelihood that loss of function and permanent disability will develop if initiation Accepted 28 March 2019
of treatment is delayed. Dermatologists are uniquely positioned to recognize early signs of PsA and be
KEYWORDS
the first-line healthcare practitioners to detect PsA in patients with psoriasis. PsA can affect six clinical
Psoriatic arthritis; psoriasis;
domains: peripheral arthritis, dactylitis, enthesitis, psoriasis, psoriatic nail disease, and axial disease. peripheral arthritis;
However, not every patient will have involvement of all domains and the domains affected can change dactylitis; enthesitis;
over time. Complicating the diagnosis is the condition’s similarity with other arthritic diseases and poten- psoriatic nail disease; axial
tial heterogeneity. In this article, we provide practical guidance for dermatologists for detecting PsA in disease; disability; loss
patients with psoriasis. We also review the available treatment options by each clinical domain of PsA of function
and give advice on how to interpret the results of PsA clinical trials. Through early recognition of PsA in
patients with psoriasis and initiation of proper treatment, dermatologists can help to prevent PsA disease
progression, irreversible joint damage, and resultant permanent disability, and improve quality of life.

Introduction with first- and second-degree relatives of affected individuals hav-


ing increased risk of PsA (16,17).
Psoriatic arthritis (PsA) is a potentially progressive, erosive,
The pathophysiology of PsA is similar to that of psoriasis and
chronic, heterogeneous, and systemic inflammatory disease that
involves important cytokines in the interleukin (IL)-23-Th17-IL-17
develops in up to 30% of patients with psoriasis and can manifest
and tumor necrosis factor (TNF) pathway (e.g. IL-12, IL-17, IL-23,
in up to six different clinical domains, including peripheral arth-
and TNF-a), as well as cytokines such as IL-22 (13,18,19). Thus, it
ritis, dactylitis, enthesitis, psoriasis, psoriatic nail disease, and axial
disease (1–3). Peripheral arthritis can cause pain in a variety of is not surprising that many cytokine-targeting biologics and
joints in PsA and commonly involves the knee (41%), finger small-molecule inhibitors that provide effective skin clearance in
(26%), hip (19%), ankle (19%), and wrist (16%) (3). Dactylitis is col- psoriasis also improve joint symptoms and slow radiographic pro-
loquially referred to as ‘sausage digit,’ and is a distinguishing fea- gression in PsA.
ture of PsA, characterized by uniform swelling of an entire digit Although PsA is a widely recognized comorbidity in psoriasis,
that occurs in up to 48% of patients (4). Another distinguishing nearly 52% of patients with psoriasis have joint pain without a
feature of PsA, enthesitis is present in 35% of patients and is diagnosis of PsA and the average diagnostic delay for PsA is
defined as inflammation where the tendon, ligament, or joint cap- 5 years (3,20). Dermatologists have an essential role in reducing
sule inserts into bone (5–7). Psoriatic lesions can develop on the this diagnostic delay by screening patients with psoriasis for PsA,
skin and may affect the nails (e.g. pitting). Axial symptoms, which as 85% of patients develop psoriasis before PsA, and in these
may occur in up to 50% of patients with PsA, result in back stiff- cases, PsA frequently develops within 10 years following the
ness and pain that improves with movement (8,9). appearance of psoriasis (21–23). There are a number of comorbid-
Etiologically, the familial aggregation of psoriasis and PsA is ities that are more common in patients with PsA than in patients
indicative of a genetic basis for psoriatic disease. The major with psoriasis without signs of PsA, including inflammatory bowel
histocompatibility complex is a known susceptibility locus for PsA disease, cardiovascular diseases (including obesity, hypertension,
and psoriasis (10–12), as shown by the observation that nearly and type 2 diabetes mellitus), uveitis, depression, anxiety, and
25% of patients with PsA are positive for human leukocyte fatty liver disease (24,25). Thus, this indicates a need for screening
antigen (HLA)-B27 (13). Specific HLA alleles are associated with all patients with psoriasis.
different PsA manifestations, including symmetric sacroiliitis Early detection and treatment of PsA are critically important
(HLA-B27:05), asymmetric sacroiliitis (HLA-B08:01 and HLA- for improving long-term patient outcomes, as the disease can
C07:01), enthesitis (HLA-B27:05 and HLA-C01:02), dactylitis progress rapidly and cause irreversible joint damage (21,26).
(HLA-B27:05 and HLA-B08:01), and synovitis (HLA-B08:01) (14). Specifically, diagnostic and treatment delays of more than
HLA-Cw0602 is more common in PsA than the general 6 months contribute to peripheral joint erosions and poor func-
population and is associated with an earlier age of psoriasis onset tional outcomes (27). When PsA is identified early, initiation of
(15). Additionally, there is a strong familial aggregation of PsA treatment with targeted therapies can significantly improve the

CONTACT Alice Gottlieb alicegottliebderm@gmail.com Department of Dermatology, Icahn School of Medicine at Mt Sinai, 10 Union Square East, New York,
NY, USA
ß 2019 The Author(s). Published with license by Taylor & Francis Group, LLC
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in
any way.
JOURNAL OF DERMATOLOGICAL TREATMENT 663

signs and symptoms of PsA and prevent radiographic progres- at the more commonly affected entheses (lateral epicondyle, med-
sion (28–30). ial femoral condyle, and Achilles tendon insertion) and then pal-
The remainder of this review will provide dermatologists with pate and look for signs of inflammation (Figure 1). Somatic
practical insights into how to recognize the varied clinical mani- symptoms and tender point counts can help differentiate PsA
festations of PsA, properly diagnose PsA, and interpret results from fibromyalgia, in which patients have more tender points and
from clinical trials of PsA. greater enthesitis scores (37). Detecting diffuse tenderness in soft
tissue areas away from joints and entheses is also helpful in differ-
entiating fibromyalgia from PsA. Patients with reactive arthritis
Diagnosis of PsA
can also exhibit enthesitis that commonly involves the Achilles
Inflammatory versus non-inflammatory arthritis tendon and plantar fascia (38). Other factors that can help to dif-
ferentiate reactive arthritis from PsA include younger age at onset,
Recognizing subtle symptomatic differences between inflamma-
sacroiliitis, and articular symptoms primarily affecting the lower
tory and non-inflammatory arthritis is critical for proper diagnosis,
limbs and the presence of acute anterior uveitis, dysuria, or diar-
as these diseases share similar characteristics such as pain and
rhea/inflammatory bowel disease (38). Being cognizant of these
joint stiffness. In general, clinical characteristics of inflammatory
differences, a diagnosis of reactive arthritis or PsA requires rheu-
arthritis include prolonged stiffness upon immobility, joint pain,
matologic evaluation and the dermatologist will have benefited
joint tenderness, joint swelling, pain at entheses, swollen digits,
the patient greatly even if the diagnosis is reactive arthritis rather
and worsened back pain upon immobility (9,31). Patients with
than PSA.
non-inflammatory arthritis typically present with normal acute-
Although PsA is a type of inflammatory arthritis, it may be
phase reactants, bony crepitus, asymmetric joint involvement, dis-
challenging to identify, because its symptoms may mimic non-
tal interphalangeal (DIP) joint involvement, a lack of prolonged
inflammatory arthritis. More specifically, in patients with PsA,
morning stiffness, and bony outgrowths around the joints (9,31).
the erythrocyte sedimentation rate (ESR) and C-reactive protein
Of note, non-inflammatory arthritis may be suspected in patients
(CRP) level are within the normal range nearly 50% of the time,
with PsA because inflammatory markers are normal in 50% of
joints are affected asymmetrically, DIP joint involvement may
patients with PsA and DIP involvement in PsA may be confused
be present, and radiographic manifestations include new bone
with osteoarthritis on plain radiography (31,32). Questions to help formation and concurrent bone loss as opposed to only bony
practitioners identify inflammatory arthritis are presented in erosion (13,31,32,39,40). Characteristics that can help dermatol-
Table 1. ogists differentiate PsA from non-inflammatory arthritis (osteo-
arthritis), inflammatory arthritis (gout and rheumatoid arthritis
Clinical diagnosis of PsA [RA]), and pain (fibromyalgia) are described in Table 2. These
characteristics, however, can be challenging to differentiate
A simple mnemonic, ‘PSA,’ representing pain (in the joints), stiff-
when two or more diseases are present simultaneously, and
ness (>30 min after inactivity/sausage digit [dactylitis]), and axial
osteoarthritis and/or fibromyalgia can present with PsA (41,42).
(axial spine involvement/back pain associated with stiffness and
Practitioners should also be aware that the risk of gout is sig-
pain that improves with activity), has been developed as a con- nificantly increased in patients with concurrent PsA and
venient method to help practitioners quickly recognize specific psoriasis (43).
characteristics of PsA (9). Enthesitis may present before symptoms To address the need for early screening tools to identify PsA
of arthritis in individuals with PsA, and early enthesitis is not in patients with psoriasis, three validated screening tools exist:
detectable by radiography. Thus, because enthesitis may be the the psoriasis and epidemiology screening tool (PEST), the psori-
only presenting musculoskeletal symptom of PsA, it is important atic arthritis screening and evaluation (PASE), and the Toronto
for dermatologists to query patients regarding enthesitis symp- psoriatic arthritis screening (ToPAS) (44–48). PEST is a patient
toms. During clinical examinations, dermatologists can screen for self-assessment tool consisting of five questions and a drawing
visible signs of enthesitis such as redness and swelling at insertion of a mannequin diagram on which patients indicate locations of
sites (33). Although enthesitis is typically evaluated by manual stiff, swollen, or painful joints (49,50). PASE is a questionnaire of
palpation, it is particularly challenging to assess because objective 15 items designed specifically for use in dermatology clinics to
signs of inflammation such as redness and swelling are often identify patients with psoriasis and symptoms of inflammatory
absent and symptoms of enthesitis are known to mimic those of musculoskeletal disease (44,51,52). ToPAS is a 12-item question-
fibromyalgia, which is also associated with pain on digital palpa- naire in which patients use pictures of psoriatic skin, nail disease,
tion at tender points (34–37). Evaluation of enthesitis is a chal- inflamed joints, and dactylitis to identify symptoms (49). In a
lenge amongst dermatologists and rheumatologists due to these comparison of these tools, PEST, the only nonproprietary screen-
issues and additional education on techniques for evaluating ing tool and available on the free GRAPPA mobile phone app,
enthesitis would be beneficial. We suggest to first ask about pain and ToPAS had slightly better sensitivity than PASE in detecting
PsA in patients with psoriasis; however, all questionnaires had
Table 1. Questions to ask patients to help identify the presence of inflamma- high false-positive rates, resulting in low specificity (53).
tory arthritis. Although screening tools can be useful in some circumstances,
‘Yes’ supports the presence of inflammatory arthritis they are often inconvenient and challenging to include during
 Do you have a family history of psoriasis or psoriatic arthritis? routine office visits with patients (9). Additionally, it is important
 Have you ever had tender/swollen fingers or toes? for dermatologists to first recognize the presence of inflammatory
 Do you experience morning stiffness lasting longer than a half hour
to 1 h? arthritis before attempting differential diagnosis for PsA so that
 Does your pain improve with exercise? other causes of joint pain are not overlooked.
 Have your symptoms of joint pain persisted for > 3 months? The ClASsification criteria for Psoriatic ARthritis (CASPAR) crite-
 Does your pain improve with nonsteroidal anti-inflammatory drugs? ria, which were developed to standardize enrollment in PsA clin-
 How would you describe your pain (gnawing, throbbing, deep)?
ical trials, can help dermatologists identify patients with PsA but
664 A. GOTTLIEB AND J. F. MEROLA

Figure 1. Enthesitis sites evaluated by clinical assessment instruments. LEI: Leeds Enthesitis Index; MASES: Maastricht Ankylosing Spondylitis Enthesitis Score; SPARCC:
Spondyloarthritis Research Consortium of Canada. For scoring purposes, the inferior patella and tibial tuberosity are considered one site because of their anatomical
proximity. Image from Mease 2017 (178).

are not required for diagnosis (54). The diagnosis of PsA is clinical treatments that are effective for other PsA domains may not
and based on patient history and physical examination and sup- improve axial disease.
ported by imaging and laboratory evaluation. Patients can fulfill The numerically valued criteria of CASPAR include psoriasis or
the CASPAR criteria to be diagnosed with PsA and dermatologists a personal/family history of psoriasis, psoriatic nail dystrophy, a
may find these criteria useful when evaluating patients for pos- negative test result for the presence of rheumatoid factor (RF),
sible signs of PsA (55). These classification criteria consist of two current dactylitis or history of dactylitis (recorded by a rheuma-
groupings: the stem (or required criteria) and criteria associated tologist), and juxta-articular new-bone formation (54).
with a numerical value (54,55). To fulfill the CASPAR criteria, a Among the CASPAR criteria, ongoing psoriasis (which is
patient must present with at least one of the stem components weighted more than all other criteria) or a personal or family his-
and 3 points from the criteria listed in Table 3. tory of psoriasis is encompassed within one category, whereas
The stem criteria of CASPAR are defined under the broad term nail dystrophy is counted as a separate category. Of the remain-
‘inflammatory articular disease,’ which consists of inflammatory ing criteria, dactylitis requires the most careful clinical attention,
joint (peripheral) disease, enthesitis, and inflammatory axial dis- because it may occur in any digit, but most frequently manifests
ease. Inflammatory joint disease is generally recognized by joint in the toes (59). A dactylitic digit, which is very specific to PsA,
swelling, erythema, and pain (even at rest) (55). may be red, hot, and tender (acute dactylitis) or swollen without
As part of the clinical examination for peripheral arthritis (part acute inflammatory changes (chronic dactylitis) and may occur in
of the stem criteria of CASPAR), dermatologists should evaluate isolation or as one of several digits affected (6,59,60). During clin-
patients for signs of swelling, which is indicative of active syno- ical examinations, dermatologists can identify dactylitis by screen-
vitis, and tenderness, which is another sign of inflammation. ing for ‘sausage-shaped’ digits, which present with uniform
Practical assessments can be conducted by applying light pres- inflammation such that soft tissue between the metacarpophalan-
sure (e.g. enough to blanch the tip of a fingernail) at the joint geal and proximal interphalangeal, proximal and DIP, and/or DIP
line. Key joints to evaluate include DIP joints, proximal interpha- and digital tuft are diffusely swollen (61). The entire digit is
langeal joints, metacarpophalangeal and metatarsophalangeal affected and swelling at each joint cannot be independently rec-
joints, and wrist, elbow, shoulder, sternoclavicular, temporoman- ognized, and flexion is difficult or impossible (62). The presence
dibular, hip, knee, ankle, and midtarsal joints (56). Novel educa- of RF, a type of autoantibody first found to be associated with RA
tional tools are being developed to help train dermatologists on and not typically present in PsA, is determined through a blood
how to accurately perform a musculoskeletal examination to test (63). Although negative RF is one of the numerically valued
screen for PsA. CASPAR criteria, RF is present in approximately 13% of patients
Axial inflammatory articular disease (part of the stem criteria with PsA, and the incidence of positive RF increases with age
of CASPAR) is typically characterized by slow-developing back (21,64). Anti-cyclic citrullinated peptide antibodies are an add-
pain persisting for >3 months before age 40 years, alternating itional negative marker of PsA (not included in the CASPAR crite-
buttock pain (caused by sacroiliitis), pain causing waking from ria); however, roughly 13% of patients with PsA present with
sleep during the second half of the night and prolonged morning these antibodies (65). When positive in patients with PsA, both RF
stiffness or stiffness upon immobility (57,58). Additionally, and anti-cyclic citrullinated peptide antibodies typically have a
JOURNAL OF DERMATOLOGICAL TREATMENT 665

Table 3. CASPAR criteria.

1:5 to 1:10
Female-to-
male ratio
3:1 to 4:1

1:1 to 2:1
Presence of inflammatory articular disease (joint, spine, or entheseal) with  3
points from the list below (54)

9:1

1:1
Characteristics (points)
Psoriasis (2) or Personal history of psoriasis (1) or Family history of

secondary to tophi eruption, eye


involvement, and manifestations
psoriasis (1)

Rarely: dermatologic manifestation

mimicking a tumor or infection


Extra-articular manifestations

Myalgias, tender points, irritable


Dactylitis or a history of dactylitis recorded by a rheumatologist (1)

Psoriasis, dactylitis, tendonitis,


Subcutaneous nodules, carpal

Juxta-articular new bone formation (1)


Negative for the rheumatoid factor (1)
Psoriatic nail dystrophy (1)

onychodystrophy
tunnel syndrome

bowel syndrome

Extensor tendon
None

Superficial &
deep laminae
Nail
90% ACPAþ

13% ACPAþ
75–80% RFþ
Laboratory

and ACPA

and ACPA

and ACPA
findings

Normal RF

Normal RF

Normal RF
13% RFþ
bone growth, enthesitis
Key radiographic findings

sclerosis, osteophytosis
Bone erosion, soft tissue
swelling, joint space
narrowing, marginal

Bone erosion and new


Joint space narrowing,

Deep lamina
Tophi, bone erosion

Flexor tendon

Figure 2. The fascia of the nail root is an extension of the entheses. Image from
McGonagle 2009 (179).
erosions

low titer, especially in comparison to patients with RA (66,67).


None

Finally, the last numerically valued CASPAR criteria, juxta-articular


new bone formation, is not typically a feature of early PsA.
involvement

Cervical and

Yes and no
lumbar

ACPA, anti-citrullinated peptide antibodies; PsA, psoriatic arthritis; RA, rheumatoid arthritis; RF, rheumatoid factor.
Axial

Atypical
Cervical

Psoriasis subtypes and PsA


Yes

Although psoriasis can occur in a variety of locations on the


Symmetrical

body, specific manifestations are clinical predictors of PsA (68).


Several studies have found an increased risk of PsA has correla-
Variably

Variably

tions with psoriatic scalp lesions, intertriginous/inverse psoriasis,


Yes

Yes

No

and nail dystrophy (68–71). The hazard ratios for PsA in patients
with scalp, intergluteal/perianal, and nail psoriasis are 3.89, 2.35,
Classically metatarsophalangeal/podagra

and 2.93, respectively (68), and in mild psoriasis, 83% of patients


Lower extremity, proximal and distal
Pattern of joint involvement

with scalp and nail psoriasis, 40% of patients with intergluteal/


Increased risk in patients with
may involve joints of any size

concurrent psoriasis and PSA

perianal lesions, and 37% of patients with isolated scalp psoriasis


interphalangeal joints, first

met CASPAR criteria for PsA (69). Similarly, the incidences of scalp,
carpometacarpal joint

intergluteal, and nail psoriasis were higher in patients with PsA


Small and large joints

(100, 83, and 64%, respectively) than in patients with psoriasis


alone (67, 25, and 40%, respectively) (70). Recognition of inverse
Large and small

psoriasis is an important clue for informing a diagnosis of PsA


that is commonly missed during physical examination. Inverse
Reproduced with modification from Mies Richie (150).
Diffuse

psoriasis has typically been considered uncommon but contem-


porary findings report a prevalence of 21–30% for inverse psoria-
sis in patients with psoriasis (72).
Inflammation

Although nail psoriasis is common in moderate-to-severe psor-


Table 2. Differential diagnosis of PsA.

iasis without PsA, it is even more prevalent in PsA (over 80% of


patients affected) and is hypothesized to indicate involvement of
Yes

Yes

Yes
No

No

the distal phalanx in PsA (55,68,73). This unique link between nail
In premenopausal women.

dystrophy and PsA can be explained by the physiological relation-


or acute
Chronic

Chronic

Chronic

Chronic

Chronic

ship between nails and entheses. Histological studies have dem-


Both

onstrated that the extensor tendon, which is attached to the


terminal phalanx, extends distally and connects with the nail root,
(43,148,149)
Osteoarthritis

Fibromyalgia

revealing that the fascia of the nail root is an extension of the


PsA (64,65)

enthesis and providing evidence of how inflammation of these


RA (145)

(146)

(147)
Disease

entheses often gives rise to nail pitting (Figure 2) (74). The link
Gout

between nail disease and adjacent enthesitis has further been


666 A. GOTTLIEB AND J. F. MEROLA

demonstrated through magnetic resonance imaging (MRI) and instruments are used to evaluate patient health status (patient-
ultrasound studies (75,76). Therefore, although nails are develop- reported outcomes [PROs]), and several individual clinical and
mentally related to the skin, they are functionally linked with PRO measures have been integrated into composite indices to
entheses, underscoring the correlation between nail disease/oste- create more comprehensive measures.
olysis and PsA – particularly in DIP joints (77,78).
Overall, these observations indicate that dermatologists should
Peripheral arthritis measures
pay particular attention for PsA manifestations in patients with
psoriasis affecting the scalp, intergluteal/perianal areas, and Peripheral arthritis is predominantly assessed by indices, including
nails (79). the 68 tender and 66 swollen joint count, the 28-joint Disease
Activity Score (DAS28), a composite measure that assesses 28
peripheral joints along with a measure of ESR or CRP (in the
Treatment of PsA
DAS28-CRP measure), and the American College of Rheumatology
The European League Against Rheumatism (EULAR) and the (ACR) criteria, another composite measure that specifies a
Group for Research and Assessment of Psoriasis and Psoriatic percentage improvement (i.e. 20, 50, or 70%) in tender or swollen
Arthritis (GRAPPA) have developed recommendations for the joint counts, as well as 3 of 5 additional measures (Table 4)
treatment of PsA, with guidelines for appropriate use of (56,83). These measures include the patient’s assessment of pain,
nonsteroidal anti-inflammatory drugs (NSAIDs), traditional disease- the patient’s global assessment of disease activity, the physician’s
modifying anti-rheumatic drugs (DMARDs; e.g. methotrexate, assessment of physical function, the patient’s assessment of
sulfasalazine, leflunomide, cyclosporine), biologics, and small-mol- physical function, and acute-phase reactant value. Furthermore,
ecule inhibitors (80,81). These recommendations provide specific when trials last longer than 1 year and agents are being tested as
guidance for treatment by PsA clinical domain. Choice of therapy DMARDs, ACR includes radiography or another imaging technique
should be optimized to target the PsA symptoms that are the (83,84). Across PsA clinical trials, 20% improvement in ACR criteria
most burdensome to each patient, with the overarching goals of (ACR20) has been used as a primary endpoint to assess the
maximizing long-term health-related quality of life and preventing efficacy of biologics and small-molecule inhibitors for the
irreversible joint structural damage (80,81). Briefly, NSAIDs are treatment of articular symptoms. Results from these trials are
recommended to control pain and inflammation in patients with summarized in Figure 3. However, because DAS28 was originally
peripheral arthritis, axial disease, and enthesitis, and traditional developed for RA, it does not evaluate some of the most
DMARDs are recommended as treatments for peripheral arthritis, frequently involved joints in PsA (particularly in the lower
dactylitis, and skin/nail disease. Guidelines clarify that the term extremities) and all PsA domains. Although ACR response is used
‘DMARD’ to describe these agents was chosen for historical as the primary endpoint in PsA clinical trials, it is not employed in
purposes and that these agents do not provide any disease-modi- clinical practice and does not evaluate all PsA domains.
fying effects on radiographic damage. Biologics that inhibit TNF-a,
IL-17A, or IL-12/23 are recommended across all PsA domains, and
Dactylitis measures
the small-molecule phosphodiesterase-4 inhibitor, apremilast, is
recommended for all domains except axial disease (80,81). These Dactylitis is also measured using multiple approaches (Table 4).
guidelines were developed in 2015, when data were available for The simplest assessment is counting dactylitic digits (tender and
the TNF-a inhibitors, etanercept, adalimumab, infliximab, certolizu- non-tender or just tender) (85). Alternative clinical indices include
mab pegol, and golimumab, and for the IL-12/23 inhibitor, usteki- a 0–3 scale of physician-rated severity to assess all 20 digits (86),
numab (80,81). Since then, additional data have been published the Leeds Dactylitis Index (LDI), and LDI-Basic (87,88). The LDI and
for apremilast, the IL-17A inhibitors secukinumab and ixekizumab, LDI-Basic multiply the tenderness score of each affected digit
the Janus kinase inhibitor tofacitinib, and the T-cell inhibitor (based on the Ritchie index – graded 0–3 for LDI or binary score
abatacept, all of which are now approved for adults with active for LDI-Basic) by the ratio of the circumference of the affected
PsA. The IL-23 inhibitor guselkumab is approved for moderate-to- digit to the circumference of the digit on the opposite hand or
severe plaque psoriasis and is in late-stage development for PsA. foot. Results from clinical trials using these indices to evaluate the
Additionally, NSAIDs, adalimumab, certolizumab pegol, etanercept, effects of treatment on dactylitis are summarized in Table 5.
golimumab, infliximab, and secukinumab are approved for the Drugs that have shown statistically significant improvements in
treatment of ankylosing spondylitis (AS), a condition related dactylitis indices compared with placebo include infliximab,
to PsA. certolizumab pegol, intravenous golimumab, subcutaneous
golimumab 100 mg, ustekinumab, guselkumab, secukinumab, and
ixekizumab. Statistical significance was not reached or was not
Domain-specific evaluation of PsA in clinical trials
tested for in trials of adalimumab, etanercept, tofacitinib,
Dermatologists should be particularly aware of measures used to apremilast, and abatacept. It is not possible to indirectly compare
assess PsA, as the International Dermatology Outcome Measures results across trials because different clinical indices and different
(IDEOM) group has recently encouraged screening and subse- reporting methods were used to assess dactylitis across studies.
quent measurement of PsA symptoms in all psoriasis clinical trials At present, there is no consensus on the best method for
(82). The IDEOM group evaluated the patient global (PG)-arthritis, assessing dactylitis in PsA clinical trials, and minimal clinically
routine assessment patient index data (RAPID)-3, and Psoriatic important difference (MCID) thresholds have not been established
Arthritis Impact of Disease (PsAID)-9 tools for PsA symptom evalu- for available dactylitis indices.
ation and recommended that PsAID9 and RAPID3 are potentially
more appropriate measures of PsA symptoms than PG-arthritis
Enthesitis measures
(82). In PsA clinical trials, various measures (Table 4) are used to
assess peripheral arthritis, dactylitis, enthesitis, skin/nail disease, Enthesitis can be measured with imaging techniques, such as
and radiographic outcomes. Additionally, several different ultrasound and MRI; however, in dermatology clinics, use of
JOURNAL OF DERMATOLOGICAL TREATMENT 667

Table 4. Indices used to measure PsA activity by clinical domain (56,89,96–99,121,151–158).


PsA domain Measure Description/guidance for practical use Advantages/disadvantages
Peripheral arthritis 66 SJC, 68 TJC Specific joints are palpated for signs of tenderness and  Easy to perform; simple scoring
swelling, which are signs of inflammation and active  Can be completed in 2 min
synovitis. To perform, apply 4 kg/cm2 of pressure at  Good responsiveness in PsA clinical trials
the joint line  Severity of tenderness and swelling is
not graded
DAS28 A composite joint count measure that includes joints of  Good performance characteristics in PsA
both shoulders, elbows, knees, and wrists, as well as  Not recommended for use in PsA inclusion
all MCP joints, and all PIP joints of the hands criteria or as a primary endpoint in clinical
trials because it may underestimate disease
in lower extremities and DIP joints
 Severity of tenderness and swelling is
not graded
 Does not evaluate foot involvement, which
is common in PsA
ACR The ACR joint count documents the number of joints  Reliable measure of peripheral disease activ-
with tenderness and swelling ity in PsA clinical trials
 Can be used for patient follow-up in clin-
ical practice
Dactylitis 0–3 score assigned to Overall scores range from 0 (none) to 60 (severe)  Simple counting and scoring method
each digit of the  Demonstrated responsiveness in PsA trials
hands and feet of biologics
 Not as quantitative as LDI
LDI Uses an instrument called a dactylometer to measure  Minimal burden for patients
digit circumference and evaluate pain and tenderness  Objective quantitative measure
on a 0–3 scale; size comparisons are made vs. contra-  Requires a specialized tool (dactylometer)
lateral digits  More complicated scoring system that
involves comparison of the ratio of circum-
ferences between affected and contralat-
eral digits
Enthesitis 4-point enthesitis Enthesitis at Achilles tendon and plantar fascia graded  Demonstrated responsiveness in PsA clin-
measure as present or absent or scored on a 0–3 scale ical trials
 Does not perform as well as indices that
evaluate more sites
LEI Bilateral evaluation of six entheseal sites (lateral epicon-  Developed specifically for PsA
dyles, medial femoral condyles, and Achilles tendon  Can be completed in 30 s
insertions) for the presence (1) or absence (0)  Correlates well with other PsA disease activ-
of tenderness ity measures
 Least floor effect of available enthesi-
tis indices
MASES Evaluation of 13 entheseal sites (bilateral first costo-  Recommended by ASAS for use in SpA
chondral joints, seventh costochondral joints, poster- randomized controlled trials
ior superior iliac spines, anterior superior iliac spines,  Can be completed in 2–5 min
iliac crests, proximal insertion of Achilles tendons,  Demonstrated responsiveness in PsA clin-
and fifth lumbar spinous process) for the presence ical trials
(1) or absence (0) of tenderness  Not validated in PsA
 Lower ICC in patients with PsA vs. AS
SPARCC Bilateral evaluation of 16 entheseal sites (Achilles ten-  Can be completed in 2–5 min
dons, plantar fascia insertion at the calcaneus, patel-  Correlates well with AS disease activ-
lar tendon insertion at base of the patella, quadriceps ity measures
insertion into the superior border of the patella,  Not validated in PsA
supraspinatus insertion into the greater tuberosity of
the humerus, and medial and lateral epicondyles) for
the presence (1) or absence (0) of tenderness.
Modified versions are available evaluating 6 and
8 sites
Berlin Evaluation of 12 entheseal sites scored as absent (0) or  Has not been used in PsA clinical trials
present (1)  Lower ICC in patients with PsA vs. AS
San Francisco Evaluation of 17 tendon-insertion sites scored based on  More sensitive than Berlin index and MASES
pain and tenderness on a scale of 0–3 in AS clinical trial
 Has not been used in PsA clinical trials
 Lower ICC in patients with PsA vs. AS
Skin PASI Quantitative assessment of skin lesions based on BSA  Widely used in psoriasis and PsA clin-
affected and severity of erythema, induration, and ical trials
scale, weighted by body area  Can be administered in 5 min
 Not used in clinical practice, in part because
of complexity
PGA Assessment of skin lesions scored on a scale from 0  Simpler to use than PASI
(clear) to 6 (very severe)  Widely recognized and accepted by
dermatologists
 Less quantitative than PASI
BSA Assessed by considering the surface area of skin  Basic, easy-to-perform assessment that can
affected by psoriasis lesions; patient’s handprint be used in daily practice
(palm and fingers) is estimated to be 1% BSA  Does not include assessment of lesion sever-
ity or body location affected
(continued)
668 A. GOTTLIEB AND J. F. MEROLA

Table 4. Continued.
PsA domain Measure Description/guidance for practical use Advantages/disadvantages
Nails NAPSI Nails are scored based on the presence or absence of  Has been used extensively in psoriasis clin-
nail bed and nail matrix psoriasis, with each nail div- ical trials
ided into four quadrants (scores for each nail range  Relatively straightforward physical
from 0 to 8) examination
 Can be completed in 5–10 min
 Not validated in psoriasis or PsA
 Not used in clinical practice
Modified NAPSI Each nail is evaluated for seven features: pitting, ony-  Shorter and easier to use than NAPSI
cholysis and oil-dropping dyschromia, nail plate  Good inter-rater reliability
crumbling, leukonychia, splinter hemorrhages, hyper-  Used in PsA clinical trials
keratosis, and red spots in the lunula  Well correlated with other clinical measures
of PsA
 Relatively straightforward physical
examination
 Can be completed in <5 min
Radiographic Modified Steinbrocker 40 joints in the hands and feet are classified on a 0–4  Simple scoring method used since 1949
progression global scoring radiographic scale in which 0 ¼ normal, 1 ¼ juxta-  Can be performed quickly in clinical prac-
method articular osteoporosis or soft-tissue swelling, 2 ¼ ero- tice (6 min)
sions, 3 ¼ erosion and joint-space narrowing or sub-  Does not include use of any radio-
luxation, and 4 ¼ total joint destruction (lysis graphic standards
or ankylosis)  Less sensitive than other methods
Sharp method Evaluates joints in the hands (40–44 joints) and feet  Various Sharp score modifications have been
for PsA (8–12 joints) for erosions and joint-space narrowing used in different PsA clinical trials
on a scale of 0–5; separate grading is used for pen-  More sensitive to detecting change in RA
cil-in-cup phenomenon, gross osteolysis, periostitis, than PsA
and tuft resorption  Time-consuming and not practical for use in
a clinical setting
Sharp/van der Heijde Erosions are scored from 0 to 5 and joint-space narrow-  Has been widely used in PsA clinical trials
method for PsA ing from 0 to 4, with pencil-in-cup and osteolysis  Can be performed in 15 min
scored separately  Changes > MCID have not been observed in
PsA clinical trials because disease progres-
sion is slow relative to study durations
PARS Assessment of 40 joints in the hands and feet scored  Validated in PsA
separately for destruction (0–5 scale) and bony prolif-  Only instrument to evaluate proliferation,
eration (0–4 scale) for a total score range of 0–360 which is a PsA-specific feature
 Can be performed in 10 min
 Destruction and proliferation scales are only
weakly correlated
 Has not been used in any clinical trials
Spine BASDAI Set of 6 questionnaires scored on a 0–10 cm VAS about  Most commonly used measure in AS clin-
assessment symptoms of fatigue, pain, and stiffness ical trials
 Can be completed in 2 min
 Does not discriminate well between periph-
eral and axial PsA disease activity
 Fails to discriminate between high and low
PsA disease activity
BASFI Set of 10 items related to functional anatomy (bending,  Commonly used in AS disease
reaching, changing position, standing, turning, and impact studies
climbing steps) and ability to cope with everyday life  Can be completed in <3 min
rated on a 0–10 cm VAS  Does not discriminate well between periph-
eral and axial disease activity
ACR: American College of Rheumatology; AS: ankylosing spondylitis; ASAS: Assessment of SpondyloArthritis international Society; BASDAI: Bath Ankylosing
Spondylitis Disease Activity Index; BASFI: Bath Ankylosing Spondylitis Function Index; BSA: body surface area; DAS28: Disease Activity Score in 28 joints; DIP: distal
interphalangeal; ICC: intraclass correlation coefficient; LDI: Leeds Dactylitis Index; LEI: Leeds Enthesitis Index; MASES: Maastricht Ankylosing Spondylitis Enthesitis
Score; MCID: minimal clinically important difference; MCP: metacarpophalangeal; NA: not available; NAPSI: nail psoriasis severity index; PARS: Psoriatic Arthritis
Ratingen score; PASI: psoriasis area and severity index; PGA: physician global assessment; PIP: proximal interphalangeal; PsA: psoriatic arthritis; RA: rheumatoid arth-
ritis; SDD: smallest detectable difference; SJC: swollen joint count; SpA: spondyloarthritis; SPARCC: spondyloarthritis research consortium of Canada; TJC: tender joint
count; VAS: visual analog scale.

domain-specific clinical indices is more practical. Instruments avail- different studies, differences in reporting methods and study
able for the assessment of enthesitis include the Leeds Enthesitis designs restrict the ability to indirectly compare results across
Index (LEI), the Maastricht Ankylosing Spondylitis Enthesitis Score studies. Furthermore, the clinical significance of improvements on
(MASES), the 4-point enthesitis measure, the Spondyloarthritis different indices is not well established, and MCID is not available
Research Consortium of Canada (SPARCC) enthesitis measure, the for any enthesitis measure (89,90). However, it is noteworthy that
Berlin index, and the San Francisco index (Table 4) (56). statistically significant improvements in enthesitis have been
As with dactylitis, there is currently no consensus on which reported compared with placebo in phase 3 studies of infliximab,
enthesitis index is best; therefore, PsA trials of biologics and certolizumab pegol, golimumab (subcutaneous and intravenous
small-molecule inhibitors have used different instruments. As formulations), secukinumab, ustekinumab, and apremilast, and in
shown in Table 6, even when the same instrument was used in a phase 2 study of guselkumab (Table 6).
JOURNAL OF DERMATOLOGICAL TREATMENT 669

100

76.8

% of patients achieving
80 71.7

ACR20 at Week 24
69
63.8 65
61 60
57 56.3 58
60 52 53 51
54
48 49 49.5
42.4
39.4
36.6
40
26.4

20

0

Et rce l 4 mg ID †

Q W‡

um 10 W ‡
G b 5 mg §
um g §

el V §

cu izu ab b ║
to ma em st 2 um t
um pe last m ab*

pt mg Q4 *
ol m W/Q *

iz lix b

Se kin ma 2W

1 W

ci 30 g
U fac 10 g
U kinu ib g*
in b g*

90 g
g
C lizu Ap ila alim cep

Et b p ol 2 0 m BID

er 50 m 2W

50 BI W

ek Inf ma

m
To tinib 0 m

ab m
m
im 0 m

ki ab 4

e itin m
e k ma m
Se Ixek um ima
us b I
ol ab /Q
B

Q
cu um b Q

To ma 50

um 45
re A ata

an pt 00 Q

ku
a 0

5
a
g
g

G um W
g
Ab

fa b
0

im g
3

e go 0
d

G
ol
0

nu
im

Ix
i
m

st
st
an e

ce

G
r
Ap

liz b
a
to
er
er
C

Figure 3. ACR20 response rates reported at week 24 in PsA trials of biologics and small-molecule inhibitors (86,102,103,132,134–136,138,139,159–161,180). For agents
without a dose specified, results are presented for the approved dosage. Figure is for visualization purposes only and should not be used to make direct comparisons
of efficacy between therapies. Data values have not been statistically corrected. Unless otherwise indicated, the proportion of patients with ACR20 at week 24 was
the primary endpoint of the study. ACR: American College of Rheumatology; BID: two times daily; BIW: twice weekly; PsA: psoriatic arthritis; Q2W: every 2 weeks;
Q4W; every 4 weeks; QW: once weekly. Primary endpoint: ACR20 at week 12; †Primary endpoint: ACR20 at week 16; ‡ACR20 at week 24 is a secondary endpoint;
§
Primary endpoint: ACR20 at week 14; ||Results reported at week 16 (primary endpoint).

Table 5. Results of dactylitis assessments by clinical index from trials of biologics and small-molecule inhibitors in PsA.
Dactylitis index Trial name/description Key dactylitis outcomes
Scale of 0–3 for each ADEPT (Adalimumab Effectiveness in Psoriatic Mean improvement in dactylitis was greater with adalimumab vs. placebo
digit of the hands Arthritis Trial) (159) but statistical significance was not reached (values not reported)
and feet PRESTA (Psoriasis Randomized Etanercept STudy in Mean percent change from baseline to week 24 was 85% with etanercept
Subjects with Psoriatic Arthritis) (160) 50 mg BIW/QW and 85% with etanercept 50 mg QW/QW
IMPACT (Infliximab Multinational Psoriatic Arthritis At week 16, mean percent improvement was 85% in the infliximab group
Controlled Trial) (86) vs. 29% in the placebo group (p < .001)
GO-REVEAL (A Randomized Evaluation of Safety Median percent change from baseline to week 24 was 100% with
and Efficacy in Subjects with Psoriatic Arthritis golimumab 50 mg, 100% with golimumab 100 mg, and 42% with
Using a Human Anti-TNF Monoclonal placebo (both p  .09 vs. placebo)
Antibody) (161)
GO-VIBRANT (A study of golimumab in participants Mean change from baseline to week 24 in dactylitis score was –8.2 with IV
with active PsA) (102) golimumab 2 mg/kg and –5.0 with placebo (p < .001)
PSUMMIT 1 (A study of the safety and effectiveness Among patients with dactylitis at baseline, dactylitis was present at week
of ustekinumab in patients with PsA) (138) 24 in 56% of patients treated with ustekinumab 45 or 90 mg vs. 76% of
the placebo group (p¼.0013)
OPAL Beyond (Oral Psoriatic Arthritis Trial; Mean dactylitis score change from baseline to month 3 was –5.2 with
Tofacitinib in PsA subjects with inadequate tofacitinib 5 mg, –5.4 with tofacitinib 10 mg, and –1.9 with placebo
response to TNF inhibitors) (134) (statistical significance not tested)
Efficacy and safety of guselkumab in patients with Median percent improvement from baseline to week 24 was 100% with
active PsA: a phase 2a, randomized, double- guselkumab 100 mg and 33% with placebo (p < .001)
blind, placebo-controlled study (132)
Presence (1) or absence (0) FUTURE 1 (Efficacy at 24 weeks and long-term At week 24, complete resolution of dactylitis was achieved by 52% of
for each digit of the safety, tolerability and efficacy up to 2 years of patients receiving secukinumab 75 or 150 mg compared with 16% of
hands and feet secukinumab (AIN457) in patients with active the placebo group (p < .05)
PsA (133)
PALACE 1 (Psoriatic Arthritis Long-term Assessment The percentage of patients with complete resolution of dactylitis at week
of Clinical Efficacy 1) (135) 24 was 51% with apremilast 20 mg BID, 48% with apremilast 30 mg
BID, and 41% with placebo (statistical significance not reached)
LDI or LDI-Basic RAPID-PsA (Certolizumab pegol in subjects with Mean LDI change at week 24 was –40.7 with certolizumab pegol 200 mg
adult onset active and progressive PsA) (139) Q2W, –53.5 with certolizumab pegol 400 mg Q4W, and –22.0 with pla-
cebo (both p  .002)
SPIRIT-P1 (A study of ixekizumab in participants Mean change from baseline in LDI-Basic was –75.4 with ixekizumab Q4W,
with active PsA) (136) –66.1 with ixekizumab Q2W, and –33.7 with placebo (both p < .05 vs.
placebo). Complete resolution of dactylitis (LDI-Basic score ¼ 0) was
reported by 80% of the ixekizumab Q4W group, 77% in the ixekizumab
Q2W group, and 25% of the placebo group (both p  .001 vs. placebo)
ASTRAEA (Active PSoriaTic Arthritis RandomizEd Using LDI-Basic, complete resolution of dactylitis at week 24 was achieved
TriAl) (103) by 44% of patients treated with abatacept vs. 34% with placebo; statis-
tical significance was not reached
BID: two times daily; IV: intravenous; LDI: Leeds Dactylitis Index; PsA: psoriatic arthritis; Q2W: every 2 weeks; Q4W: every 4 weeks; TNF: tumor necrosis factor.
670 A. GOTTLIEB AND J. F. MEROLA

Table 6. Results of enthesitis assessments by clinical index from trials of biologics and small-molecule inhibitors in PsA.
Enthesitis index Trial name/description Key enthesitis outcomes
4-point scale ADEPT (Adalimumab Effectiveness in Psoriatic Arthritis Mean improvement in enthesitis was greater with adalimumab vs placebo,
Trial) (159) but statistical significance was not reached (values not reported)
PRESTA (Psoriasis Randomized Etanercept STudy in 81% of patients treated with etanercept 50 mg BIW/QW, and 81% of patients
Subjects with Psoriatic Arthritis) (160) treated with etanercept 50 mg QW/QW had a reduction in enthesitis sites
at week 24
IMPACT (Infliximab Multinational Psoriatic Arthritis At week 16, 14% of patients treated with infliximab compared with 31% of
Controlled Trial) (86) the placebo group had enthesitis (p < .001)
FUTURE 1 (Efficacy at 24 weeks and long term safety, At week 24, complete resolution of enthesitis was achieved by 48% of
tolerability and efficacy up to 2 years of patients treated with secukinumab 75 or 150 mg compared with 13% of
secukinumab (AIN457) in patients with active the placebo group (p < .05)
PsA (133)
LEI RAPID-PsA (Certolizumab pegol in subjects with adult In patients with LEI >1 at baseline, mean change at week 24 was –2.0 with
onset active and progressive PsA) (139) certolizumab pegol 200 mg Q2W, –1.8 with certolizumab pegol 400 mg
Q4W, and –1.1 with placebo (both p  .003)
GO-VIBRANT (A study of golimumab in participants Mean change from baseline to week 14 in LEI was –2.1 with IV golimumab
with active PsA) (102) 2 mg/kg and –1.1 with placebo (p < .001)
SPIRIT-P1 (A study of ixekizumab in participants with Mean LEI score change from baseline to week 24 was 1.3 for IXEQ4W, 1.4
active PsA) (136) for IXEQ2W, and 0.8 for placebo (statistical significance not reached)
OPAL Beyond (Oral Psoriatic Arthritis Trial; Tofacitinib Mean LEI score change from baseline to month 3 was –1.3 with tofacitinib
in PsA subjects with inadequate response to TNF 5 mg, –1.3 with tofacitinib 10 mg, –0.5 with placebo (statistical significance
inhibitors) (134) not tested)
ASTRAEA (Active PSoriaTic Arthritis RandomizEd Complete resolution of enthesitis at week 24 was achieved by 33% of
TriAl) (103) patients treated with abatacept vs. 21% with placebo (statistical signifi-
cance not reached)
Efficacy and safety of guselkumab in patients with Median percent improvement from baseline to week 24 was 100% with
active PsA: a phase 2a, randomized, double-blind, guselkumab 100 mg and 33% with placebo (p ¼ .009)
placebo-controlled study (132)
MASES or modified GO-REVEAL (A Randomized Evaluation of Safety and Median percent change on modified MASES from baseline to week 24 was
MASES Efficacy in Subjects with Psoriatic Arthritis Using a 60% with golimumab 50 mg, 67% with golimumab 100 mg, and 12% with
Human Anti-TNF Monoclonal Antibody) (161) placebo (both p < .001 vs. placebo)
PSUMMIT 1 (A study of the safety and effectiveness of Among patients with modified MASES 1 at baseline, enthesitis was present
ustekinumab in patients with PsA) (138) at week 24 in 65% of patients treated with ustekinumab 45 or 90 mg vs.
81% of the placebo group (p ¼ .0006)
PALACE 1 (Psoriatic Arthritis Long-term Assessment of The percentage of patients with complete resolution of enthesitis at week 24
Clinical Efficacy 1) (135) was 32% with apremilast 20 mg BID, 34% with apremilast 30 mg BID, and
14% with placebo; both p  .0037 vs. placebo
BID: two times daily; BIW: twice weekly; IV: intravenous; IXEQ2W: ixekizumab 80 mg once every 2 weeks; IXEQ4W: ixekizumab 80 mg once every 4 weeks; LEI: Leeds
Enthesitis Index; MASES: Maastricht Ankylosing Spondylitis Enthesitis Score; PsA: psoriatic arthritis; Q2W: every 2 weeks; Q4W: every 4 weeks; QW: once weekly; TNF:
tumor necrosis factor.

Skin and nail psoriasis measures While PsA clinical trials have included skin assessments,
results from psoriasis trials can provide the most specific
Psoriasis is measured by tools frequently used in dermatology
and focused information on the effects of different therapies
trials, including the psoriasis area and severity index (PASI),
on clearance of skin and nail lesions. Table 7 summarizes
body surface area (BSA), physicians global assessment (PGA),
PASI and NAPSI response rates for biologic and small-mol-
Investigator’s Global Assessment (IGA), or IGA modified 2011
ecule inhibitor therapies approved for the treatment of
(Table 4) (56,91). The primary endpoint in psoriasis trials and the moderate-to-severe plaque psoriasis, including etanercept,
key skin endpoint in PsA trials are PASI responses (usually infliximab, adalimumab, ustekinumab, guselkumab,
percentage of patients achieving at least 75% improvement in secukinumab, ixekizumab, and apremilast.
PASI scores [PASI75]). Nail psoriasis is assessed using the nail
psoriasis severity index (NAPSI) or modified NAPSI (Table 4) (56).
The National Psoriasis Foundation recently defined Radiographic measures
acceptable response as either BSA 3% or 75% BSA As shown in Table 4, the most common radiographic scoring
improvement from baseline and target response as BSA methods for PsA clinical trials are modified from RA scoring meth-
1% for patients with psoriasis (92). Additionally, the ods and include the modified Steinbrocker, the modified Sharp
product of PGA and BSA (PGAxBSA) is a simple tool for method for PsA, the modified Sharp/van der Heijde method for
assessing psoriasis severity and response to treatment with PsA (also reported as the van der Heijde-modified total Sharp
concordance to PASI response that can evaluate treatment score [mTSS]), and the Psoriatic Arthritis Ratingen Score (PARS)
targets for psoriasis (93–95). As use of newer biologics and (96). Although these tools all analyze joints in the hands and feet,
small-molecule inhibitors will be needed to meet these scoring criteria vary between methods. For example, both
goals, it is critical that dermatologists understand the modified Sharp and mTSS separately assess erosions and joint
impact that these drugs have on PsA and other domains of space narrowing (a product of cartilage destruction), whereas the
psoriatic disease so that patients are appropriately treated. PARS method separately assesses erosions and bony prolifera-
Treatment of skin disease by dermatologists is an import- tions. Further, the modified Steinbrocker uses only one scale that
ant component of comprehensive PsA disease management. considers erosions, joint space narrowing, and ankylosis. GRAPPA
JOURNAL OF DERMATOLOGICAL TREATMENT 671

Table 7. Results of psoriasis assessments from trials of biologics and small-molecule inhibitors in chronic plaque psoriasis.
Skin outcomes Nail outcomes
Drug Trial PASI result Trial NAPSI result
Etanercept Etanercept as monotherapy Week 12 PASI 75 Open-label trial of the effects Mean target fingernail NAPSI
in patients with psoria-  49% with 50 mg BIW (p < .001 of etanercept 50 mg BIW/ score change from baseline
sis (162) vs. placebo) QW or QW/QW on nail to week 24:
 34% with 25 mg BIW (p < .001 psoriasis in patients with  –4.3 with etanercept
vs. placebo) moderate-to-severe plaque 50 mg BIW/QW
 14% with 25 mg QW (p < .001 psoriasis (163)  –4.4 with etanercept
vs. placebo) 100 mg QW/QW
 4% with placebo
Week 12 PASI 90
 22% with 50 mg BIW (p < .001
vs. placebo)
 12% with 25 mg BIW (p < .001
vs. placebo)
 3% with 25 mg QW
 1% with placebo
Infliximab EXPRESS (Infliximab induction Week 10 EXPRESS (164) NAPSI % improvement from
and maintenance therapy  PASI 75 achieved by 80% with baseline to week 24:
for moderate-to-severe infliximab vs. 3% with pla-  56% with infliximab vs.
psoriasis) (164) cebo (p < .0001) –3% with pla-
 PASI 90 achieved by 57% with cebo (p < .0001)
infliximab vs. 1% with pla-
cebo (p < .0001)
Adalimumab REVEAL (Adalimumab therapy Week 12 Post hoc nail psoriasis suba- Median NAPSI response at
for moderate to severe  PASI 75 achieved by 68% with nalysis of data from the week 16, 40%
psoriasis) (165) adalimumab vs. 5% with pla- BELIEVE study in patients
cebo (p < .001 vs. placebo) with moderate-to-severe
 PASI 90 achieved by 37% with psoriasis (166)
adalimumab vs. 2% with pla-
cebo (p < .001 vs. placebo)
Ustekinumab PHOENIX 1 (Efficacy and Week 12 PASI 75 PHOENIX 1 (168) NAPSI % improvement from
safety of ustekinumab in  66% with ustekinumab 90 mg baseline to week 24:
patients with psoria- (p < .0001 vs. placebo)  47% with ustekinu-
sis) (167)  67% with ustekinumab 45 mg mab 45 mg
(p < .0001 vs. placebo)  49% with ustekinu-
 Placebo 3% mab 90 mg
Week 12 PASI 90
 37% with ustekinumab 90 mg
(p < .0001 vs. placebo)
 42% with ustekinumab 45 mg
(p < .0001 vs. placebo)
 Placebo 2%
Secukinumab ERASURE (Efficacy or Week 12 PASI 75 TRANSFIGURE (Phase 3b Mean NAPSI % change from
response and safety of  82% with secukinumab study of secukinumab vs. baseline to week 16:
two fixed secukinumab 300 mg (p < .001 vs. placebo) placebo in patients with  –45% with secukinumab
regimens in psoria-  72% with secukinumab moderate-to-severe psoria- 300 mg (p < .0001
sis) (169) 150 mg (p < .001 vs. placebo) sis with nail involve- vs. placebo)
 5% with placebo ment (170)  –38% with secukinumab
Week 12 PASI 90 150 mg (p < .0001
 59% with secukinumab vs. placebo)
300 mg (p < .001 vs. placebo)  –11% with placebo
 39% with secukinumab
150 mg (p < .001 vs. placebo)
 1% with placebo
Ixekizumab UNCOVER-3 (Comparison of Week 12 PASI 75 UNCOVER-3 (172) Mean NAPSI % change from
ixekizumab with  84% with ixekizumab Q4W baseline to week 12:
etanercept or placebo in (p < .0001 vs. placebo)  –39% with
moderate-to-severe  87% with ixekizumab Q2W ixekizumab Q2W
psoriasis) (171) (p < .0001 vs. placebo)  –40% with
 7% with placebo ixekizumab Q4W
Week 12 PASI 90  þ5% with placebo
 65% with ixekizumab Q4W
(p < .0001 vs. placebo)
 68% with ixekizumab Q2W
(p < .0001 vs. placebo)
 3% with placebo
Apremilast ESTEEM 1 (Efficacy and safety Week 16 PASI 75 Subanalysis of ESTEEM 1 and Mean NAPSI % change vs pla-
trial evaluating the effects  33% with apremilast 30 mg ESTEEM 2 data for apremi- cebo at week 16:
of apremilast in psoria- BID (p < .0001 vs. placebo) last 30 mg BID vs. placebo  –23% vs. þ6.5% in
sis) (173)  5% with placebo in patients with moderate- ESTEEM 1; p < .0001
to-severe psoriasis (174)  –29% vs. –7.1% in
ESTEEM 2; p < .0001
(continued)
672 A. GOTTLIEB AND J. F. MEROLA

Table 7. Continued.
Skin outcomes Nail outcomes
Drug Trial PASI result Trial NAPSI result
Guselkumab VOYAGE 1 (Efficacy and Week 16 PASI 75 VOYAGE 1 (175) Mean NAPSI % change from
safety of guselkumab com-  91% with guselkumab baseline to week 16:
pared with adalimumab (p < .001 vs. placebo)  34% with guselkumab
for the continuous treat-  6% with placebo  –1% with placebo
ment of patients with Week 16 PASI 90
moderate-to-severe psoria-  73% with guselkumab
sis (175) (p < .001 vs. placebo)
 3% with placebo
BIW: twice weekly; NAPSI: Nail Psoriasis Severity Index; PASI: Psoriasis Area and Severity Index; Q2W: every 2 weeks; Q4W: every 4 weeks; QW: once weekly.

Table 8. Results of radiographic progression assessments from trials of biologics and small-molecule inhibitors in PsA.
Trial name/description Key radiographic outcomes
Etanercept treatment of PsA: safety, efficacy, and effect on Annualized rate of change in modified TSS at 12 months was –0.03 units with etanercept vs.
disease progression (137) þ1.00 unit for placebo (p ¼ .0001)
ADEPT (Adalimumab Effectiveness in Psoriatic Arthritis At week 24, mean change from baseline in modified TSS was –0.1 with adalimumab and þ0.9
Trial (131) with placebo; changes at week 48 were þ0.1 and þ1.0, respectively (SDC ¼ 1.88)
FUTURE 5 (Study to Demonstrate the Efficacy [Including At week 24, mean change from baseline in modified TSS was þ0.08 with secukinumab 300 mg
Inhibition of Structural Damage], Safety and Tolerability up with loading dose (p < .01 vs. placebo), þ0.17 with secukinumab 150 mg with loading dose
to 2 Years of Secukinumab in Active Psoriatic (p < .05 vs. placebo), –0.09 with secukinumab 150 mg without loading dose (p < .05 vs. pla-
Arthritis) (176) cebo), and þ0.50 with placebo
SPIRIT-P1 (A study of ixekizumab in participants with active At week 24, mean change from baseline in modified TSS was þ0.17 with ixekizumab Q4W,
PsA) (136) þ0.08 with ixekizumab Q2W, and þ0.49 with placebo (both p  .01)
IMPACT 2 (Infliximab Multinational Psoriatic Arthritis On a 0–528 scale (SDC ¼ 2.7), mean PsA-modified SHS change from baseline to week 24 was
Controlled Trial 2) (100) –0.70 with infliximab 5 mg and þ0.82 with placebo (p ¼ .001); 97% of the infliximab group
and 88% of the placebo group had scores  SDD
GO-REVEAL (A Randomized Evaluation of Safety and Efficacy On a 0–528 scale (SDC ¼ 1.56), mean changes from baseline to week 24 on the PsA-modified
in Subjects with Psoriatic Arthritis Using a Human Anti-TNF SHS were –0.16 with golimumab 50 mg (p ¼ .011 vs. placebo), –0.02 with golimumab 100 mg
Monoclonal Antibody) (101) (p ¼ .086 vs. placebo), and þ0.27 with placebo. The percent of patients with a PsA-modified
SHS change from baseline  SDD was 96% with golimumab 50 mg, 94% with golimumab
100 mg, and 89% with placebo
GO-VIBRANT (A study of golimumab in participants with active On a 0–528 scale (SDC ¼ 2.49), mean change in total PsA-modified SHS was –0.4 with intraven-
PsA) (102) ous golimumab 2 mg/kg vs. 2.0 in the placebo group (p < .001)
RAPID-PsA (Certolizumab pegol in subjects with adult onset Imputation method significantly affected assessment and reporting of radiographic progression
active and progressive PsA) (177) With no imputation, mTSS change from baseline to week 24 was þ0.06 with certolizumab
pegol (pooled doses) and þ 0.29 with placebo (p ¼ .008)
PSUMMIT-1 and PSUMMIT-2 (Study of the safety and effective- On a 0–528 scale (SDC ¼ 2.01), mean changes from baseline to week 24 was 0.4 with
ness of ustekinumab in patients with PsA) (29) ustekinumab 45 or 90 mg and 1.0 with placebo (p < .001). In both groups, change in SHS
was  SDD for most patients (92% with ustekinumab and 84% with placebo; p < .001)
ASTRAEA (Active PSoriaTic Arthritis RandomizEd TriAl) (103) Mean change from baseline in PsA-modified total SHS score was 0.30 with abatacept vs. 0.35
with placebo at week 24; statistical significance and SDC not reported
mTSS: modified total Sharp score; PsA: psoriatic arthritis; Q2W: every 2 weeks; Q4W: every 4 weeks; QW: once weekly; SDC: smallest detectable change; SHS: Sharp/
van der Heijde score; TSS: total Sharp score.

consensus is that the Sharp/van der Heijde method for PsA is the radiographic progression (118–120). Apremilast does not have
optimal tool for use in randomized controlled trials (97). approval for inhibition of radiographic progression from
Radiographic progression results from PsA trials are summar- either agency.
ized in Table 8. All drugs, except abatacept, were associated with
statistically significant differences compared with placebo in
Spine measures
mTSS/modified Sharp/van der Heijde score (not reported for
guselkumab, apremilast, or tofacitinib). However, differences The most commonly used indices for assessment of spondylitis
between active treatment and placebo were below the MCID include the Bath Ankylosing Spondylitis Disease Activity Index
based on the smallest detectable difference observed in RA (BASDAI) and the Bath Ankylosing Spondylitis Functional Index
studies (98,99). In many cases, most (>80%) patients in both (BASFI) (Table 4). Components of the BASDAI and BASFI, along
placebo and active-treatment groups had a change in Sharp/van with patient assessments of spinal pain and global impression of
der Heijde score that was below the smallest detectable change, disease activity rated on a 0–10 cm visual analog scale, are
indicating that no measurable progression was observed in the included in the composite Assessment of SpondyloArthritis inter-
majority of patients during the study period (29,100–102). This national Society (ASAS) improvement criteria (121), which is the
lack of progression has made it difficult to detect meaningful most common primary endpoint in recent AS clinical trials.
treatment differences in radiographic progression (103). Of the While axial disease is an important clinical domain of PsA,
drugs available for PsA, approval for inhibition of radiographic fewer than 5% of patients have isolated spinal joint involvement
progression is given for adalimumab, etanercept, certolizumab (26). Thus, because not all patients with PsA present with spinal
pegol, infliximab, ixekizumab, secukinumab, and abatacept by involvement and use of MRI in clinical trials is expensive, formal
both the FDA and EMA (104–117). For golimumab, ustekinumab, assessment of spondylitis has not been performed in randomized
and tofacitinib, only the EMA gives approval for inhibition of controlled PsA trials. However, the effects of biologic and small-
JOURNAL OF DERMATOLOGICAL TREATMENT 673

molecule inhibitor therapies on axial disease in PsA can be control goal had significantly improved joint outcomes compared
inferred from results of controlled clinical trials of these agents in with patients receiving standard care.
patients with AS. All TNF-a inhibitors approved for the treatment
of active PsA (etanercept, adalimumab, infliximab, golimumab,
Summary
and certolizumab pegol) are also approved for the treatment of
AS. In AS clinical trials, ASAS20 response rates were similar to all PsA is a heterogeneous, rapidly developing disease with poten-
TNF-a inhibitors, ranging from 58 to 61% (122). In the MEASURE 1 tially deleterious effects and frequently occurs within 10 years fol-
and MEASURE 2 studies of the IL-17A inhibitor secukinumab in lowing the appearance of psoriasis (21). We consider PsA to be
patients with AS, ASAS20 response rates at week 16 were 61 and the major comorbidity of psoriasis due to the likelihood of PsA to
61%, respectively, with the 150-mg dose and 60 and 41%, result in permanent disability if left untreated. Early PsA diagnosis
respectively, with the 75-mg dose (123). ASAS20 response rates and initiation of treatment can prevent possibly adverse conse-
were significantly higher with tofacitinib 5 mg (80.8%) compared quences, whereas a diagnostic delay of more than 6 months con-
with placebo (41.2%; p < .001) in a phase 2 dose-ranging study in tributes to poor radiographic and functional outcomes (27).
AS (124). In a small open-label pilot study of abatacept in AS, Therefore, while providing important care to manage the skin
27% of TNF inhibitor-naïve and 20% of TNF inhibitor-experienced symptoms of psoriasis, dermatologists have the unique ability to
patients achieved ASAS20 responses, suggesting limited efficacy recognize early signs of PsA in patients with psoriasis and be the
of abatacept for this indication (125). Methotrexate has not been first healthcare practitioners to detect PsA. Dermatologists can
demonstrated to show benefit in patients with AS (126). A clinical then initiate treatment with agents that are effective for both
trial of ixekizumab (NCT02696798) is ongoing in AS. psoriasis and PsA. There are currently available PsA drugs that
block TNF-a and IL-17A. These medications can inhibit radio-
PRO measures graphic progression, control signs and symptoms, improve phys-
ical function, prevent disability, and improve quality of life. Thus,
PRO measures were developed to assess patients’ perspectives of dermatologists, by detecting PsA early, are key to preventing dis-
various quality-of-life domains. These measures include the pain ability in these patients. Treatments should be selected that tar-
visual analog scale (VAS), the health assessment questionnaire get the most burdensome or progressive PsA clinical domain(s)
disability index (HAQ-DI), the EuroQoL 5 dimension questionnaire for each patient based on the efficacy profiles of available treat-
(EQ-5D), and the 36-item short form survey (SF-36), which is bro- ments for these domains.
ken into the physical component summary (PCS) and mental
component summary (MCS) (56). Changes from baseline of
10 mm for pain VAS and 0.30–0.35 for HAQ-DI have been Acknowledgments
used as MCID in PsA trials, and an MCID for change from baseline
Technical assistance with editing and styling of the manuscript for
in SF-36 has been defined as 5–10 (2.5–5 for PCS and MCS
submission was provided by Oxford PharmaGenesis Inc. and was
individually) in PsA (101,127–130). Clinically important improve-
funded by Novartis Pharmaceuticals Corporation. The authors
ments in these PROs have been reported in PsA clinical trials of
were fully responsible for all content and editorial decisions and
adalimumab, etanercept, infliximab, golimumab (subcutaneous
received no financial support or other form of compensation
and intravenous), certolizumab pegol, ustekinumab, guselkumab,
related to the development of this manuscript.
secukinumab, ixekizumab, apremilast, tofacitinib, and abatacept
(100–103,131–139). Additionally, the PsAID12, which is available
on the GRAPPA mobile-phone app, and the RAPID3 question- Disclosure statement
naires are useful for dermatologists to use in clinical practice to
Alice Gottlieb: Consultant/advisory board agreements for Janssen,
evaluate the impact of PsA symptoms on patients (140,141).
Celgene, Bristol-Myers Squibb, AbbVie, UCB, Novartis, Incyte, Lilly,
Reddy Labs, Valeant, Dermira, Allergan, Sun, XBiotech, and Leo.
Additional composite measures Pharmaceutical industry research/educational grants from Janssen,
Incyte, UCB, Novartis, and Lilly, and XBiotech.
Composite indices, such as the Psoriatic Arthritis Disease Activity
Joseph F. Merola: Consultant and/or investigator for Merck
Score (PASDAS), have been developed as a means of integrating
Research Laboratories, Abbvie, Eli Lilly and Company, Novartis,
information from a variety of individual outcome indices, includ-
Janssen, UCB, Samumed, Celgene, Sanofi Regeneron, GSK, Almirall,
ing physician’s global assessment scores, disease activity indices,
and PROs into one meaningful measure of PsA (142). Minimal dis- Sun Pharma, Biogen, Pfizer, Incyte, Aclaris, and Leo Pharma.
ease activity (MDA), a measure that encompasses all aspects of
PsA, is different from PASDAS and similar composite indices Funding
because it was created to be an objective target for treatment.
This work was supported by Novartis Pharmaceuticals Corporation.
When MDA criteria are met, PsA is considered to be in a satisfac-
tory state of disease activity for both patients and physicians.
Patients are classified as achieving MDA when they meet 5 of 7 References
criteria, including tender joint count 1; swollen joint count 1;
PASI 1 or body surface area 3%; patient pain VAS 15; patient 1. Acosta Felquer ML, FitzGerald O. Peripheral joint involve-
global disease activity VAS 20; health assessment questionnaire ment in psoriatic arthritis patients. Clin Exp Rheumatol.
0.5; and tender entheseal points 1 (143). The value of using 2015;33:S26–S30.
MDA as a treatment target was demonstrated in the TIght 2. Mease PJ, Gladman DD, Papp KA, et al. Prevalence of
COntrol of Psoriatic Arthritis (TICOPA) trial (144). In this study, rheumatologist-diagnosed psoriatic arthritis in patients
MDA was successfully used as an objective goal in a treat-to-tar- with psoriasis in European/North American dermatology
get approach as patients with early PsA treated with this tight clinics. J Am Acad Dermatol. 2013;69:729–735.
674 A. GOTTLIEB AND J. F. MEROLA

3. Kavanaugh A, Helliwell P, Ritchlin CT. Psoriatic arthritis and 22. Catanoso M, Pipitone N, Salvarani C. Epidemiology of
burden of disease: patient perspectives from the popula- psoriatic arthritis. Reumatismo. 2012;64:66–70.
tion-based multinational assessment of psoriasis and psori- 23. Gottlieb AB, Kircik L, Eisen D, et al. Use of etanercept for
atic arthritis (MAPP) survey. Rheumatol Ther. 2016;3: psoriatic arthritis in the dermatology clinic: the experience
91–102. diagnosing, understanding care, and treatment with eta-
4. Coates LC, Hodgson R, Conaghan PG, et al. MRI and ultra- nercept (EDUCATE) study. J Dermatolog Treat. 2006;17:
sonography for diagnosis and monitoring of psoriatic arth- 343–352.
ritis. Best Pract Res Clin Rheumatol. 2012;26:805–822. 24. Husted JA, Thavaneswaran A, Chandran V, et al.
5. Terslev L, Naredo E, Iagnocco A, et al. Defining enthesitis Cardiovascular and other comorbidities in patients with
in spondyloarthritis by ultrasound: results of a Delphi pro- psoriatic arthritis: a comparison with patients with psoria-
cess and of a reliability reading exercise. Arthritis Care Res. sis. Arthritis Care Res (Hoboken). 2011;63:1729–1735.
2014;66:741–748. 25. de Oliveira M, de Oliveira Rocha B, Vieira Duarte G.
6. Gladman DD. Clinical features and diagnostic considera- Psoriasis: classical and emerging comorbidities. An Bras
tions in psoriatic arthritis. Rheum Dis Clin North Am. 2015; Dermatol. 2015;90:9–20.
41:569–579. 26. Gottlieb A, Korman NJ, Gordon KB, et al. Guidelines of care
7. Polachek A, Li S, Chandran V, et al. Clinical enthesitis in a for the management of psoriasis and psoriatic arthritis:
prospective longitudinal psoriatic arthritis cohort: inci- section 2. Psoriatic arthritis: overview and guidelines of
dence, prevalence, characteristics, and outcome. Arthritis care for treatment with an emphasis on the biologics. J
Care Res. 2017;69:1685–1691. Am Acad Dermatol. 2008;58:851–864.
8. Liu JT, Yeh HM, Liu SY, et al. Psoriatic arthritis: epidemi- 27. Haroon M, Gallagher P, FitzGerald O. Diagnostic delay of
ology, diagnosis, and treatment. WJO. 2014;5:537–543. more than 6 months contributes to poor radiographic and
9. Cohen JM, Husni ME, Qureshi AA, et al. Psoriatic arthritis: functional outcome in psoriatic arthritis. Ann Rheum Dis.
it’s as easy as “PSA”. J Am Acad Dermatol. 2015;72: 2015;74:1045–1050.
905–906. 28. Gladman DD. Recent advances in understanding and man-
10. Eder L, Chandran V, Gladman DD. What have we learned aging psoriatic arthritis. F1000Res. 2016;5:2670.
about genetic susceptibility in psoriasis and psoriatic arth- 29. Kavanaugh A, Ritchlin C, Rahman P, et al. Ustekinumab, an
ritis? Curr Opin Rheumatol. 2015;27:91–98. anti-IL-12/23 p40 monoclonal antibody, inhibits radio-
11. Eder L, Chandran V, Pellet F, et al. Human leucocyte anti- graphic progression in patients with active psoriatic arth-
gen risk alleles for psoriatic arthritis among patients with ritis: results of an integrated analysis of radiographic data
psoriasis. Ann Rheum Dis. 2012;71:50–55. from the phase 3, multicentre, randomised, double-blind,
12. Rahman P, Roslin NM, Pellett FJ, et al. High resolution placebo-controlled PSUMMIT-1 and PSUMMIT-2 trials. Ann
mapping in the major histocompatibility complex region Rheum Dis. 2014;73:1000–1006.
identifies multiple independent novel loci for psoriatic 30. Merola JF, Lockshin B, Mody EA. Switching biologics in the
arthritis. Ann Rheum Dis. 2011;70:690–694. treatment of psoriatic arthritis. Semin Arthritis Rheum.
13. Ritchlin CT, Colbert RA, Gladman DD. Psoriatic arthritis. N 2017;47:29–37.
Engl J Med. 2017;376:957–970. 31. Mease PJ. Distinguishing inflammatory from noninflamma-
14. FitzGerald O, Haroon M, Giles JT, et al. Concepts of patho- tory arthritis, enthesitis, and dactylitis in psoriatic arthritis:
genesis in psoriatic arthritis: genotype determines clinical a report from the GRAPPA 2010 annual meeting. J
phenotype. Arthritis Res Ther. 2015;17:115. Rheumatol. 2012;39:415–417.
15. Gladman DD, Cheung C, Ng CM, et al. HLA-C locus alleles 32. Punzi L, Podswiadek M, Oliviero F, et al. Laboratory find-
in patients with psoriatic arthritis (PsA). Hum Immunol. ings in psoriatic arthritis. Reumatismo. 2007;59(1):52–55.
1999;60:259–261. 33. Sakkas LI, Alexiou I, Simopoulou T, et al. Enthesitis in psori-
16. Chandran V, Schentag CT, Brockbank JE, et al. Familial atic arthritis. Semin Arthritis Rheum. 2013;43:325–334.
aggregation of psoriatic arthritis. Ann Rheum Dis. 2009;68: 34. Karreman MC, Weel AE, van der Ven M, et al. Prevalence of
664–667. psoriatic arthritis in primary care patients with psoriasis.
17. Karason A, Love TJ, Gudbjornsson B. A strong heritability Arthritis Rheumatol. 2016;68:924–931.
of psoriatic arthritis over four generations—the Reykjavik 35. Almodo var R, Carmona L, Zarco P, et al. Fibromyalgia in
Psoriatic Arthritis Study. Rheumatology. 2009;48: patients with ankylosing spondylitis: prevalence and utility
1424–1428. of the measures of activity, function and radiological dam-
18. Fitzgerald O, Winchester R. Psoriatic arthritis: from patho- age. Clin Exp Rheumatol. 2010;28:S33–S39.
genesis to therapy. Arthritis Res Ther. 2009;11:214. 36. Wolfe F, Clauw DJ, Fitzcharles MA, et al. The American
19. Suzuki E, Mellins ED, Gershwin ME, et al. The IL-23/IL-17 College of Rheumatology preliminary diagnostic criteria for
axis in psoriatic arthritis. Autoimmun Rev. 2014;13:496–502. fibromyalgia and measurement of symptom severity.
20. Lebwohl MG, Kavanaugh A, Armstrong AW, et al. US Arthritis Care Res. 2010;62:600–610.
Perspectives in the management of psoriasis and psoriatic 37. Marchesoni A, Atzeni F, Spadaro A, et al. Identification of
arthritis: patient and physician results from the popula- the clinical features distinguishing psoriatic arthritis and
tion-based Multinational Assessment of Psoriasis and fibromyalgia. J Rheumatol. 2012;39:849–855.
Psoriatic Arthritis (MAPP) Survey. Am J Clin Dermatol. 38. Stavropoulos PG, Soura E, Kanelleas A, et al. Reactive arth-
2016;17:87–97. ritis. J Eur Acad Dermatol Venereol. 2015;29:415–424.
21. Gladman DD, Antoni C, Mease P, et al. Psoriatic arthritis: 39. Cervini C, Leardini G, Mathieu A, et al. Psoriatic arthritis:
epidemiology, clinical features, course, and outcome. Ann epidemiological and clinical aspects in a cohort of 1.306
Rheum Dis. 2005;64:ii14–ii17. Italian patients. Reumatismo. 2005;57:283–290.
JOURNAL OF DERMATOLOGICAL TREATMENT 675

40. Mensah KA, Schwarz EM, Ritchlin CT. Altered bone remod- (SPARCC), Maastricht Ankylosing Spondylitis Enthesis Score
eling in psoriatic arthritis. Curr Rheumatol Rep. 2008;10: (MASES), Leeds Dactylitis Index (LDI), Patient Global for
311–317. Psoriatic Arthritis, Dermatology Life Quality Index (DLQI),
41. McGonagle D, Hermann KG, Tan AL. Differentiation Psoriatic Arthritis Quality of Life (PsAQOL), Functional
between osteoarthritis and psoriatic arthritis: implications Assessment of Chronic Illness Therapy-Fatigue (FACIT-F),
for pathogenesis and treatment in the biologic therapy Psoriatic Arthritis Response Criteria (PsARC), Psoriatic
era. Rheumatology. 2015;54:29–38. Arthritis Joint Activity Index (PsAJAI), Disease Activity in
42. Brikman S, Furer V, Wollman J, et al. The effect of the pres- Psoriatic Arthritis (DAPSA), and Composite Psoriatic Disease
ence of fibromyalgia on common clinical disease activity Activity Index (CPDAI). Arthr Care Res. 2011;63:S64–S85.
indices in patients with psoriatic arthritis: a cross-sectional 57. Strand V, Singh JA. Evaluation and management of the
study. J Rheumatol. 2016;43:1749–1754. patient with suspected inflammatory spine disease. Mayo
43. Merola JF, Wu S, Han J, et al. Psoriasis, psoriatic arthritis Clin Proc. 2017;92:555–564.
and risk of gout in US men and women. Ann Rheum Dis. 58. Golder V, Schachna L. Ankylosing spondylitis: an update.
2015;74:1495–1500. Aust Fam Physician. 2013;42:780–784.
44. Husni ME, Meyer KH, Cohen DS, et al. The PASE question- 59. Brockbank JE, Stein M, Schentag CT, et al. Dactylitis in
naire: pilot-testing a psoriatic arthritis screening and evalu- psoriatic arthritis: a marker for disease severity? Ann
ation tool. J Am Acad Dermatol. 2007;57:581–587. Rheum Dis. 2005;64:188–190.
45. Ibrahim GH, Buch MH, Lawson C, et al. Evaluation of an 60. Kaeley GS, Eder L, Aydin SZ, et al. Dactylitis: a hallmark of
existing screening tool for psoriatic arthritis in people with psoriatic arthritis. Semin Arthritis Rheum. 2018;48:263–273.
psoriasis and the development of a new instrument: the 61. Rothschild BM, Pingitore C, Eaton M. Dactylitis: implications
Psoriasis Epidemiology Screening Tool (PEST) question- for clinical practice. Semin Arthritis Rheum. 1998;28:41–47.
naire. Clin Exp Rheumatol. 2009;27:469–474. 62. Olivieri I, Padula A, Scarano E, et al. Dactylitis or “sausage-
46. Chandran V, Gladman DD. Toronto Psoriatic Arthritis shaped” digit. J Rheumatol. 2007;34:1217–1222.
Screening (ToPAS) questionnaire: a report from the 63. Falkenburg WJ, van Schaardenburg D, Ooijevaar-de Heer P,
GRAPPA 2009 annual meeting. J Rheumatol. 2011;38: et al. IgG subclass specificity discriminates restricted IgM
546–547. rheumatoid factor responses from more mature anti-citrul-
47. Karreman MC, Weel A, van der Ven M, et al. Performance linated protein antibody-associated or isotype-switched
of screening tools for psoriatic arthritis: a cross-sectional IgA responses. Arthritis Rheumatol. 2015;67:3124–3134.
study in primary care. Rheumatology. 2017;56:597–602. 64. Waller M, Toone EC, Vaughan E. Study of rheumatoid fac-
48. Boehncke WH, Qureshi A, Merola JF, et al. Diagnosing and tor in a normal population. Arthritis Rheum. 1964;7:
treating psoriatic arthritis: an update. Br J Dermatol. 2014; 513–520.
170:772–786. 65. Inanc N, Dalkilic E, Kamali S, et al. Anti-CCP antibodies in
49. Dominguez P, Gladman DD, Helliwell P, et al. Development rheumatoid arthritis and psoriatic arthritis. Clin Rheumatol.
of screening tools to identify psoriatic arthritis. Curr 2007;26:17–23.
Rheumatol Rep. 2010;12:295–299. 66. Candia L, Marquez J, Gonzalez C, et al. Low frequency of
50. Walsh JA, Callis Duffin K, Krueger GG, et al. Limitations in anticyclic citrullinated peptide antibodies in psoriatic arth-
screening instruments for psoriatic arthritis: a comparison ritis but not in cutaneous psoriasis. J Clin Rheumatol. 2006;
of instruments in patients with psoriasis. J Rheumatol. 12:226–229.
2013;40:287–293. 67. Popescu C, Zofota S, Bojinca V, et al. Anti-cyclic citrulli-
51. Merola JF, Husni ME, Qureshi AA. Screening instruments nated peptide antibodies in psoriatic arthritis—cross-sec-
for psoriatic arthritis. J Rheumatol. 2013;40:1623. tional study and literature review. J Med Life. 2013;6:
52. Husni ME, Qureshi AA, Koenig AS, et al. Utility of the PASE 376–382.
questionnaire, psoriatic arthritis (PsA) prevalence and PsA 68. Wilson FC, Icen M, Crowson CS, et al. Incidence and clinical
improvement with anti-TNF therapy: results from the predictors of psoriatic arthritis in patients with psoriasis: a
PRISTINE trial. J Dermatolog Treat. 2014;25:90–95. population-based study. Arthritis Rheum. 2009;61:233–239.
53. Coates LC, Aslam T, Al Balushi F, et al. Comparison of three 69. Patrizi A, Venturi M, Scorzoni R, et al. Nail dystrophies,
screening tools to detect psoriatic arthritis in patients with scalp and intergluteal/perianal psoriatic lesions: risk factors
psoriasis (CONTEST study). Br J Dermatol. 2013;168: for psoriatic arthritis in mild skin psoriasis? G Ital Dermatol
802–807. Venereol. 2014;149:177–184.
54. Taylor W, Gladman D, Helliwell P, et al. Classification crite- 70. Takata T, Takahashi A, Taniguchi Y, et al. Detection of
ria for psoriatic arthritis: development of new criteria from asymptomatic enthesitis in psoriasis patients: an onset of
a large international study. Arthritis Rheum. 2006;54: psoriatic arthritis? J Dermatol. 2016;43:650–654.
2665–2673. 71. Papadavid E, Katsimbri P, Kapniari I, et al. Prevalence of
55. Mease PJ, Armstrong AW. Managing patients with psoriatic psoriatic arthritis and its correlates among patients with
disease: the diagnosis and pharmacologic treatment of psoriasis in Greece: results from a large retrospective study.
psoriatic arthritis in patients with psoriasis. Drugs. 2014;74: J Eur Acad Dermatol Venereol. 2016;30:1749–1752.
423–441. 72. Merola JF, Li T, Li WQ, et al. Prevalence of psoriasis pheno-
56. Mease PJ. Measures of psoriatic arthritis: tender and types among men and women in the USA. Clin Exp
Swollen Joint Assessment, Psoriasis Area and Severity Dermatol. 2016;41:486–489.
Index (PASI), Nail Psoriasis Severity Index (NAPSI), Modified 73. Williamson L, Dalbeth N, Dockerty JL, et al. Extended
Nail Psoriasis Severity Index (mNAPSI), Mander/Newcastle report: nail disease in psoriatic arthritis—clinically import-
Enthesitis Index (MEI), Leeds Enthesitis Index (LEI), ant, potentially treatable and often overlooked.
Spondyloarthritis Research Consortium of Canada Rheumatology (Oxford). 2004;43:790–794.
676 A. GOTTLIEB AND J. F. MEROLA

74. Tan AL, Benjamin M, Toumi H, et al. The relationship development of an instrument specific to psoriatic arthritis.
between the extensor tendon enthesis and the nail in dis- Arthritis Rheum. 2008;59:686–691.
tal interphalangeal joint disease in psoriatic arthritis—a 90. Maksymowych WP, Mallon C, Morrow S, et al.
high-resolution MRI and histological study. Rheumatology Development and validation of the Spondyloarthritis
(Oxford). 2006;46:253–256. Research Consortium of Canada (SPARCC) Enthesitis index.
75. Aydin SZ, Castillo-Gallego C, Ash ZR, et al. Ann Rheum Dis. 2009;68:948–953.
Ultrasonographic assessment of nail in psoriatic disease 91. Langley RG, Feldman SR, Nyirady J, et al. The 5-point
shows a link between onychopathy and distal interphalan- Investigator’s Global Assessment (IGA) scale: a modified
geal joint extensor tendon enthesopathy. Dermatology. tool for evaluating plaque psoriasis severity in clinical trials.
2012;225:231–235. J Dermatolog Treat. 2015;26:23–31.
76. Tan AL, Grainger AJ, Tanner SF, et al. A high-resolution 92. Armstrong AW, Siegel MP, Bagel J, et al. From the medical
magnetic resonance imaging study of distal interphalan- board of the national psoriasis foundation: treatment tar-
geal joint arthropathy in psoriatic arthritis and osteoarth- gets for plaque psoriasis. J Am Acad Dermatol. 2017;76:
ritis: are they the same? Arthritis Rheum. 2006;54: 290–298.
1328–1333. 93. Gottlieb AB, Merola JF, Chen R, et al. Assessing clinical
77. McGonagle D. Enthesitis: an autoinflammatory lesion link- response and defining minimal disease activity in plaque
ing nail and joint involvement in psoriatic disease. J Eur psoriasis with the Physician Global Assessment and body
Acad Dermatol Venereol. 2009;23: 9–13. surface area (PGA x BSA) composite tool: an analysis of
78. Sobolewski P, Walecka I, Dopytalska K. Nail involvement in apremilast phase 3 ESTEEM data. J Am Acad Dermatol.
psoriatic arthritis. Reumatologia. 2017;55:131–135. 2017;77:1178–1180.
79. Rouzaud M, Sevrain M, Villani AP, et al. Is there a psoriasis 94. Merola JF, Amato DA, See K, et al. Evaluation of
skin phenotype associated with psoriatic arthritis? sPGA  BSA as an outcome measure and treatment target
Systematic literature review. J Eur Acad Dermatol Venereol. for clinical practice. J Invest Dermatol. 2018;138:1955–1961.
95. Walsh JA, McFadden M, Woodcock J, et al. Product of the
2014;28: 17–26.
80. Coates LC, Kavanaugh A, Mease PJ, et al. Group for physician global assessment and body surface area: a sim-
ple static measure of psoriasis severity in a longitudinal
research and assessment of psoriasis and psoriatic arthritis
cohort. J Am Acad Dermatol. 2013;69:931–937.
2015 treatment recommendations for psoriatic arthritis.
96. Wassenberg S. Radiographic scoring methods in psoriatic
Arthritis Rheumatol. 2016;68:1060–1071.
arthritis. Clin Exp Rheumatol. 2015;33:S55–S59.
81. Gossec L, Smolen JS, Ramiro S, et al. European League
97. Tillett W, Jadon D, Shaddick G, et al. Feasibility, reliability,
Against Rheumatism (EULAR) recommendations for the
and sensitivity to change of four radiographic scoring
management of psoriatic arthritis with pharmacological
methods in patients with psoriatic arthritis. Arthritis Care
therapies: 2015 update. Ann Rheum Dis. 2016;75:499–510.
Res. 2014;66:311–317.
82. Perez-Chada LM, Cohen JM, Gottlieb AB, et al. Achieving
98. van der Heijde D, Lassere M, Edmonds J, et al. Minimal
international consensus on the assessment of psoriatic
clinically important difference in plain films in RA: group
arthritis in psoriasis clinical trials: an International
discussions, conclusions, and recommendations. OMERACT
Dermatology Outcome Measures (IDEOM) initiative. Arch Imaging Task Force. J Rheumatol. 2001;28:914–917.
Dermatol Res. 2018;310:701–710. 99. Bruynesteyn K, van der Heijde D, Boers M, et al.
83. Felson DT, Anderson JJ, Boers M, et al. American College Determination of the minimal clinically important differ-
of Rheumatology. Preliminary definition of improvement in ence in rheumatoid arthritis joint damage of the Sharp/
rheumatoid arthritis. Arthritis Rheum. 1995;38:727–735. van der Heijde and Larsen/Scott scoring methods by clin-
84. Felson DT, Anderson JJ, Boers M, et al. The American ical experts and comparison with the smallest detectable
College of Rheumatology preliminary core set of disease difference. Arthritis Rheum. 2002;46:913–920.
activity measures for rheumatoid arthritis clinical trials. The 100. van der Heijde D, Kavanaugh A, Gladman DD, et al.
Committee on Outcome Measures in Rheumatoid Arthritis Infliximab inhibits progression of radiographic damage in
Clinical Trials. Arthritis Rheum. 1993;36:729–740. patients with active psoriatic arthritis through one year of
85. Clegg DO, Reda DJ, Mejias E, et al. Comparison of sulfa- treatment: results from the induction and maintenance
salazine and placebo in the treatment of psoriatic arthritis. psoriatic arthritis clinical trial 2. Arthritis Rheum. 2007;56:
A department of veterans affairs cooperative study. 2698–2707.
Arthritis Rheum. 1996;39:2013–2020. 101. Kavanaugh A, van der Heijde D, McInnes IB, et al.
86. Antoni CE, Kavanaugh A, Kirkham B, et al. Sustained bene- Golimumab in psoriatic arthritis: one-year clinical efficacy,
fits of infliximab therapy for dermatologic and articular radiographic, and safety results from a phase III, random-
manifestations of psoriatic arthritis: results from the inflixi- ized, placebo-controlled trial. Arthritis Rheum. 2012;64:
mab multinational psoriatic arthritis controlled trial 2504–2517.
(IMPACT). Arthritis Rheum. 2005;52:1227–1236. 102. Kavanaugh A, Husni ME, Harrison DD, et al. Safety and
87. Helliwell PS, Firth J, Ibrahim GH, et al. Development of an efficacy of intravenous golimumab in patients with active
assessment tool for dactylitis in patients with psoriatic psoriatic arthritis: results through week twenty-four of
arthritis. J Rheumatol. 2005;32:1745–1750. the GO-VIBRANT study. Arthritis Rheumatol. 2017;69:
88. Ferguson EG, Coates LC. Optimisation of rheumatology 2151–2161.
indices: dactylitis and enthesitis in psoriatic arthritis. Clin 103. Mease PJ, Gottlieb AB, van der Heijde D, et al. Efficacy and
Exp Rheumatol. 2014;32:S-113–S-117. safety of abatacept, a T-cell modulator, in a randomised,
89. Healy PJ, Helliwell PS. Measuring clinical enthesitis in psori- double-blind, placebo-controlled, phase III study in psori-
atic arthritis: assessment of existing measures and atic arthritis. Ann Rheum Dis. 2017;76:1550–1558.
JOURNAL OF DERMATOLOGICAL TREATMENT 677

104. Humira (adalimumab) [prescribing information]. North 129. Gladman D, Fleischmann R, Coteur G, et al. Effect of certo-
Chicago (IL): AbbVie Inc.; 2018. lizumab pegol on multiple facets of psoriatic arthritis as
105. Humira [summary of product characteristcs]. Ludwigshafen, reported by patients: 24-week patient-reported outcome
Germany: AbbVie Deutschland GmbH & Co. KG.; 2017. results of a phase III, multicenter study. Arthritis Care Res.
106. Remicade (infliximab) [prescribing information]. Horsham 2014;66:1085–1092.
(PA): Janssen Biotech, Inc.; 2018. 130. Busse JW, Bartlett SJ, Dougados M, et al. Optimal strategies
107. Remicade [summary of product characteristics]. Leiden, for reporting pain in clinical trials and systematic reviews:
The Netherlands: Janssen Biologics B.V.; 2018. recommendations from an OMERACT 12 workshop. J
108. Enbrel (etanercept) [prescribing information]. Thousand Rheumatol. 2015;42:1962–1970.
Oaks (CA): Immunex Corporation; 2017. 131. Gladman DD, Mease PJ, Ritchlin CT, et al. Adalimumab for
109. Enbrel [summary of product characteristics]. Kent (UK): long-term treatment of psoriatic arthritis: forty-eight week
Pfizer Limited; 2014. data from the adalimumab effectiveness in psoriatic arth-
110. Cimzia (certolizumab pegol) [prescribing information]. ritis trial. Arthritis Rheum. 2007;56:476–488.
Smyrna (GA): UCB, Inc.; 2017. 132. Deodhar A, Gottlieb A, Boehncke WH, et al. OP0218
111. Cimzia [summary of product characteristics]. Bruxelles, Efficacy and safety results of guselkumab, and anti-IL23
Belgium: UCB Pharma S.A.; 2019. monoclonal antibody, in patients with active psoriatic arth-
112. Taltz (ixekizumab) [prescribing information]. Indianapolis ritis over 24 weeks: a phase 2a, randomized, double-blind,
(IN): Eli Lilly and Company; 2018. placebo-controlled study [abstract]. Ann Rheum Dis. 2017;
113. Taltz [summary of product characteristics]. Utrecht, The 76:142–143.
Netherlands: Eli Lilly Nederland B.V.; 2016. 133. Mease PJ, McInnes IB, Kirkham B, et al. Secukinumab inhib-
114. Cosentyx (secukinumab) [prescribing information]. East ition of interleukin-17A in patients with psoriatic arthritis.
Hanover (NJ): Novartis Pharmaceuticals Corporation; 2018. N Engl J Med. 2015;373:1329–1339.
115. Cosentyx [summary of product characteristics]. Camberley 134. Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for
(UK): Novartis Europharm Limited; 2016. psoriatic arthritis in patients with an inadequate response
116. Orencia (abatacept) [prescribing information]. Princeton to TNF inhibitors. N Engl J Med. 2017;377:1525–1536.
(NJ): Bristol-Myers Squibb Company; 2017. 135. Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment
117. Orencia [summary of product characteristics]. Uxbridge of psoriatic arthritis in a phase 3 randomised, placebo-con-
(UK): Bristol-Myers Squibb Pharma EEIG; 2019. trolled trial with apremilast, an oral phosphodiesterase 4
118. Simponi [summary of product characteristics]. Leiden, The inhibitor. Ann Rheum Dis. 2014;73:1020–1026.
Netherlands: Janssen Biologics B.V.; 2019. 136. Mease PJ, van der Heijde D, Ritchlin CT, et al. Ixekizumab,
119. Stelara [summary of product characteristics]. Beerse, an interleukin-17A specific monoclonal antibody, for the
Belgium: Janssen-Cilag International NV.; 2016. treatment of biologic-naive patients with active psoriatic
120. Xeljanz [summary of product characteristics]. Kent (UK): arthritis: results from the 24-week randomised, double-
Pfizer Limited; 2018. blind, placebo-controlled and active (adalimumab)-con-
121. Sieper J, Rudwaleit M, Baraliakos X, et al. The Assessment trolled period of the phase III trial SPIRIT-P1. Ann Rheum
Dis. 2017;76:79–87.
of SpondyloArthritis international Society (ASAS) hand-
137. Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment
book: a guide to assess spondyloarthritis. Ann Rheum Dis.
of psoriatic arthritis: safety, efficacy, and effect on disease
2009;68: ii1–i44.
progression. Arthritis Rheum. 2004;50:2264–2272.
122. Toussirot E. Biologics in spondyloarthritis: TNFa inhibitors
138. McInnes IB, Kavanaugh A, Gottlieb AB, et al. Efficacy and
and other agents. Immunotherapy. 2015;7:669–681.
safety of ustekinumab in patients with active psoriatic
123. Baeten D, Sieper J, Braun J, et al. Secukinumab, an inter-
arthritis: 1 year results of the phase 3, multicentre, double-
leukin-17A inhibitor, in ankylosing spondylitis. N Engl J
blind, placebo-controlled PSUMMIT 1 trial. Lancet. 2013;
Med. 2015;373:2534–2548.
382:780–789.
124. van der Heijde D, Deodhar A, Wei JC, et al. Tofacitinib in
139. Mease PJ, Fleischmann R, Deodhar AA, et al. Effect of cer-
patients with ankylosing spondylitis: a phase II, 16-week, tolizumab pegol on signs and symptoms in patients with
randomised, placebo-controlled, dose-ranging study. Ann psoriatic arthritis: 24-week results of a Phase 3 double-
Rheum Dis. 2017;76:1340–1347. blind randomised placebo-controlled study (RAPID-PsA).
125. Song IH, Heldmann F, Rudwaleit M, et al. Treatment of Ann Rheum Dis. 2014;73:48–55.
active ankylosing spondylitis with abatacept: an open- 140. Holland R, Tillett W, Korendowych E, et al. Validation of
label, 24-week pilot study. Ann Rheum Dis. 2011;70: the Psoriatic Arthritis Impact of Disease (PsAID)
1108–1110. Questionnaire and its potential as a single-item outcome
126. Chen J, Veras MM, Liu C, et al. Methotrexate for ankylosing measure in clinical practice. Ann Rheum Dis. 2018;77:
spondylitis. Cochrane Database Syst Rev. 2013;CD004524. 343–347.
127. Mease PJ, Woolley JM, Bitman B, et al. Minimally important 141. Coates LC, Tillett W, Shaddick G, et al. Value of the Routine
difference of Health Assessment Questionnaire in psoriatic Assessment of Patient Index Data 3 in patients with psori-
arthritis: relating thresholds of improvement in functional atic arthritis: results from a tight-control clinical trial and
ability to patient-rated importance and satisfaction. J an observational cohort. Arthritis Care Res. 2018;70:
Rheumatol. 2011;38:2461–2465. 1198–1205.
128. Strand V, Sharp V, Koenig AS, et al. Comparison of health- 142. Helliwell PS, FitzGerald O, Fransen J, et al. The develop-
related quality of life in rheumatoid arthritis, psoriatic arth- ment of candidate composite disease activity and
ritis and psoriasis and effects of etanercept treatment. Ann responder indices for psoriatic arthritis (GRACE project).
Rheum Dis. 2012;71:1143–1150. Ann Rheum Dis. 2013;72:986–991.
678 A. GOTTLIEB AND J. F. MEROLA

143. Coates LC, Fransen J, Helliwell PS. Defining minimal dis- active psoriatic arthritis: results of a double-blind, random-
ease activity in psoriatic arthritis: a proposed objective tar- ized, placebo-controlled trial. Arthritis Rheum. 2005;52:
get for treatment. Ann Rheum Dis. 2010;69:48–53. 3279–3289.
144. Coates LC, Moverley AR, McParland L, et al. Effect of tight 160. Sterry W, Ortonne JP, Kirkham B, et al. Comparison of two
control of inflammation in early psoriatic arthritis (TICOPA): etanercept regimens for treatment of psoriasis and psori-
a UK multicentre, open-label, randomised controlled trial. atic arthritis: PRESTA randomised double blind multicentre
Lancet. 2015;386:2489–2498. trial. BMJ. 2010;340:c147.
145. Taylor PC, Maini RN. Biologic markers in the diagnosis and 161. Kavanaugh A, McInnes I, Mease P, et al. Golimumab, a new
assessment of rheumatoid arthritis. In: O’Dell JR, editor. human tumor necrosis factor a antibody, administered
UpToDate. Waltham, MA: UpToDate. [cited 28 Nov 2018]. every four weeks as a subcutaneous injection in psoriatic
146. Doherty M, Abhishek A. Clinical manifestations and diagno- arthritis: twenty-four-week efficacy and safety results of a
sis of osteoarthritis. In: Hunter D, editor. UpToDate. randomized, placebo-controlled study. Arthritis Rheum.
Waltham (MA): UpToDate. [cited 28 Nov 2018]. 2009;60:976–986.
147. Goldenberg DLG. Clinical manifestations and diagnosis of 162. Leonardi CL, Powers JL, Matheson RT, et al. Etanercept as
fibromyalgia in adults. In: Schur PH, editor. UpToDate. monotherapy in patients with psoriasis. N Engl J Med.
Waltham (MA): UpToDate. [cited 28 Nov 2018]. 2003;349:2014–2022.
148. Singh JA. Racial and gender disparities among patients 163. Ortonne JP, Paul C, Berardesca E, et al. A 24-week random-
with gout. Curr Rheumatol Rep. 2013;15:307. ized clinical trial investigating the efficacy and safety of
149. Rymal E, Rizzolo D. Gout: a comprehensive review. JAAPA. two doses of etanercept in nail psoriasis. Br J Dermatol.
2014;27:26–31. 2013;168:1080–1087.
150. Mies Richie A, Francis ML. Diagnostic approach to polyar- 164. Reich K, Nestle FO, Papp K, et al. Infliximab induction and
ticular joint pain. Am Fam Physician. 2003;68:1151–1160. maintenance therapy for moderate-to-severe psoriasis: a
151. van der Heijde D, Braun J, Deodhar A, et al. Comparison of phase III, multicentre, double-blind trial. Lancet. 2005;366:
three enthesitis indices in a multicentre, randomized, pla- 1367–1374.
cebo-controlled trial of golimumab in ankylosing spondyl- 165. Menter A, Tyring SK, Gordon K, et al. Adalimumab therapy
itis (GO-RAISE). Rheumatology. 2013;52:321–325. for moderate to severe psoriasis: a randomized, controlled
152. Rahman P, Gladman DD, Cook RJ, et al. Radiological assess- phase III trial. J Am Acad Dermatol. 2008;58:106–115.
ment in psoriatic arthritis. Br J Rheumatol. 1998;37: 166. Thaci D, Unnebrink K, Sundaram M, et al. Adalimumab for
760–765. the treatment of moderate to severe psoriasis: subanalysis
153. Wassenberg S, Fischer-Kahle V, Herborn G, et al. A method of effects on scalp and nails in the BELIEVE study. J Eur
to score radiographic change in psoriatic arthritis. Z Acad Dermatol Venereol. 2015;29:353–360.
Rheumatol. 2001;60:156–166. 167. Leonardi CL, Kimball AB, Papp KA, et al. Efficacy and safety
154. Mattei PL, Corey KC, Kimball AB. Psoriasis Area Severity of ustekinumab, a human interleukin-12/23 monoclonal
Index (PASI) and the Dermatology Life Quality Index antibody, in patients with psoriasis: 76-week results from a
(DLQI): the correlation between disease severity and psy- randomised, double-blind, placebo-controlled trial
chological burden in patients treated with biological thera- (PHOENIX 1). Lancet. 2008;371:1665–1674.
pies. J Eur Acad Dermatol Venereol. 2014;28:333–337. 168. Rich P, Bourcier M, Sofen H, et al. Ustekinumab improves
155. Carlin CS, Feldman SR, Krueger JG, et al. A 50% reduction nail disease in patients with moderate-to-severe psoriasis:
in the Psoriasis Area and Severity Index (PASI 50) is a clin- results from PHOENIX 1. Br J Dermatol. 2014;170:398–407.
ically significant endpoint in the assessment of psoriasis. J 169. Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in
Am Acad Dermatol. 2004;50:859–866. plaque psoriasis-results of two phase 3 trials. N Engl J
156. Ward MM, Guthrie LC, Alba MI. Brief report: rheumatoid Med. 2014;371:326–338.
arthritis response criteria and patient-reported improve- 170. Reich K, Arenberger P, Mrowietz U, et al. Secukinumab
ment in arthritis activity: is an American College of shows high and sustained efficacy in nail psoriasis: 1.5 year
Rheumatology twenty percent response meaningful to results from the TRANSFIGURE study. Presented at: The
patients? Arthritis Rheumatol. 2014;66:2339–2343. 2017 American Academy of Dermatology Annual Meeting;
157. Zochling J. Measures of symptoms and disease status in 2017 March 3–7; Orlando, FL.
ankylosing spondylitis: Ankylosing Spondylitis Disease 171. Griffiths CE, Reich K, Lebwohl M, et al. Comparison of ixeki-
Activity Score (ASDAS), Ankylosing Spondylitis Quality of zumab with etanercept or placebo in moderate-to-severe
Life Scale (ASQoL), Bath Ankylosing Spondylitis Disease psoriasis (UNCOVER-2 and UNCOVER-3): results from two
Activity Index (BASDAI), Bath Ankylosing Spondylitis phase 3 randomised trials. Lancet. 2015;386:541–551.
Functional Index (BASFI), Bath Ankylosing Spondylitis 172. Dennehy EB, Zhang L, Amato D, et al. Ixekizumab is effect-
Global Score (BAS-G), Bath Ankylosing Spondylitis ive in subjects with moderate to severe plaque psoriasis
Metrology Index (BASMI), Dougados Functional Index with significant nail involvement: results from UNCOVER 3.
(DFI), and Health Assessment Questionnaire for the J Drugs Dermatol. 2016;15:958–961.
Spondylarthropathies (HAQ-S). Arthritis Care Res. 2011; 173. Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral
63:S47–S58. phosphodiesterase 4 (PDE4) inhibitor, in patients with
158. Englbrecht M, Wang Y, Ronneberger M, et al. Measuring moderate to severe plaque psoriasis: results of a phase III,
joint involvement in polyarticular psoriatic arthritis: an randomized, controlled trial (Efficacy and Safety Trial
introduction of alternatives. Arthritis Care Res. 2010;62: Evaluating the Effects of Apremilast in Psoriasis [ESTEEM]
977–983. 1). J Am Acad Dermatol. 2015;73:37–49.
159. Mease PJ, Gladman DD, Ritchlin CT, et al. Adalimumab for 174. Rich P, Gooderham M, Bachelez H, et al. Apremilast, an
the treatment of patients with moderately to severely oral phosphodiesterase 4 inhibitor, in patients with
JOURNAL OF DERMATOLOGICAL TREATMENT 679

difficult-to-treat nail and scalp psoriasis: results of 2 phase evaluation of radiographic progression in patients with
III randomized, controlled trials (ESTEEM 1 and ESTEEM 2). psoriatic arthritis: results of the RAPID-PsA 24-week phase
J Am Acad Dermatol. 2016;74:134–142. III double-blind randomised placebo-controlled study of
175. Blauvelt A, Papp KA, Griffiths CE, et al. Efficacy and safety certolizumab pegol. Ann Rheum Dis. 2014;73:233–237.
of guselkumab, an anti-interleukin-23 monoclonal anti- 178. Mease PJ, Van den Bosch F, Sieper J, et al. Performance
body, compared with adalimumab for the continuous of 3 ethesitis indices in patients with peripheral
treatment of patients with moderate to severe psoriasis: spondyloarthritis during treatment with adalimumab.
results from the phase III, double-blinded, placebo- and J Rheumatol. 2017;44:599–608.
active comparator-controlled VOYAGE 1 trial. J Am Acad 179. McGonagle D, Tan AL, Benjamin M. The nail as a
Dermatol. 2017;76:405–417. musculoskeletal appendage-implications for an
176. Mease P, van der Heijde D, Landewe R, et al. Secukinumab improved understanding of the link between psoriasis
improves active psoriatic arthritis symptoms and inhibits and arthritis. Dermatology. 2009;218:97–102.
radiographic progression: primary results from the rando- 180. McInnes IB, Mease PJ, Kirkham B, et al. Secukinumab, a
mised, double-blind, phase III FUTURE 5 study. Ann Rheum human anti-interleukin-17A monoclonal antibody, in
Dis. 2018;77:890–897. patients with psoriatic arthritis (FUTURE 2): a
177. van der Heijde D, Fleischmann R, Wollenhaupt J, et al. randomised, double-blind, placebo-controlled, phase
Effect of different imputation approaches on the 3 trial. Lancet. 2015;386:1137–1146.

You might also like