BJD

British Journal of Dermatology

R E V I E W A RT I C L E

Diagnosing and treating psoriatic arthritis: an update W.H. Boehncke,1 A. Qureshi,2 J.F. Merola,2,3 D. Thacßi,4 G.G. Krueger,5 J. Walsh,6 N. Kim7 and A.B. Gottlieb8 1

Geneva University Hospital, Rue Gabrielle Perret-Gentil 4, 1211 Geneva 14, Switzerland Department of Dermatology, Brigham and Women’s Hospital, 221 Longwood Avenue, Boston, MA 02115, U.S.A. 3 Department of Dermatology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, U.S.A. 4 Comprehensive Center for Inflammation Medicine, University of L€ubeck, Ratzeburger Allee 160, 23435 L€ubeck, Germany 5 Dermatology, University Hospital, University of Utah, Clinic 28, 50 North Medical Drive, Salt Lake City, UT 84132, U.S.A. 6 University of Utah, 50 North Medical Drive, Salt Lake City, UT 84132, U.S.A. 7 Tufts Medical Center, 800 Washington Street, #231, Boston, MA 02111, U.S.A. 8 Tufts Medical Center, 800 Washington Street, #114, Boston, MA 02111, U.S.A. 2

Summary Correspondence Alice B. Gottlieb. E-mail: [email protected]

Accepted for publication 18 November 2013

Funding sources AbbVie Inc. funded medical writing assistance but had no role in the content of the manuscript.

Conflicts of interest See the Appendix DOI 10.1111/bjd.12748

Psoriatic arthritis (PsA) is an inflammatory arthritis of uncertain pathogenesis, affecting approximately one in four patients with psoriasis. Onset of psoriasis typically precedes the development of PsA. Therefore, the dermatologist is ideally positioned to recognize the early signs and symptoms of PsA for diagnosis and subsequent treatment. The role of the dermatologist in early diagnosis and treatment is essential for preventing pain and functional disabilities, as well as the joint deterioration that accompanies progressive forms of PsA. Diagnosis of PsA is a key aspect of the clinical decision process for the dermatologist, as psoriasis plus PsA requires a different therapeutic approach from that required for psoriasis alone. Furthermore, PsA is associated with an increased risk of cardiovascular comorbidities that present significant health concerns. In this review, the pathogenesis and comorbidities of PsA are discussed. In addition, screening and imaging tools that aid in the diagnosis of PsA, as well as tools used for efficacy assessment, are reviewed. Available therapies are presented, with a focus on targeted biologics and emerging treatments.

What’s already known about this topic?

• • • •

Psoriatic arthritis (PsA) is a significant comorbidity of psoriasis. Physical disability, cardiovascular comorbidities, psychological issues, and a generally poor quality of life negatively impact many to most patients with PsA. Several tools have been developed to aid in PsA diagnosis and assessment. A large body of work has been devoted to researching factors contributing to PsA and developing targeted biologic therapies.

What does this study add?

• • •

772

Dermatologists are in a position to recognize the early signs of PsA and therefore can be instrumental in achieving the goal of early initiation of adequate therapy. Early intervention may also help slow disease progression and improve patient outcomes. This review may be used by dermatologists as an educational guide on the pathogenesis, screening, diagnosis, comorbidities, existing treatments and emerging therapies for PsA.

British Journal of Dermatology (2014) 170, pp772–786

© 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 773

Psoriatic arthritis (PsA) is a chronic inflammatory arthritis that is uncommon in the general population (025% prevalence in the U.S.A.)1 but is quite common in patients with psoriasis (~25% prevalence worldwide).1–3 Psoriasis typically precedes the development of PsA by ~12 years;4,5 therefore, dermatologists are ideally situated to recognize and diagnose PsA in its early stages. Varied skin lesions accompany PsA, including localized, diffuse, guttate or pustular lesions. Among patients with psoriasis, nail dystrophy, scalp lesions, intergluteal/perianal lesions, and percentage of body surface area affected with psoriasis all appear to be associated with an increased risk for PsA.1,6 Erosive joint deformities have been reported in 40– 57% of patients with PsA;5,7 joint damage occurs in as many as 27% of patients within 10 months and in 47% of patients within 2 years of symptom onset.8 PsA has been placed in the family of spondyloarthropathies negative for rheumatoid factor, which includes ankylosing spondylitis, reactive arthritis and inflammatory bowel diseaseassociated arthropathy.9,10 PsA may afflict any synovial joint and the most common forms of PsA involve the joints of the hands and feet. PsA is characterized by swollen and tender joints, dactylitis [inflammation of an entire digit (‘sausage digit’)] and enthesitis (inflammation at sites of tendon insertion to bone), and axial involvement (inflammation in the sacroiliac joints or spine); inflammation may lead to progressive joint deterioration.5 PsA differs from other forms of inflammatory arthritis in that excessive bone is sometimes produced at joint margins; a diagnostic feature is the simultaneous presence of erosions and excessive bone within the same joint unit. In a combined effort, an international group of rheumatologists developed CASPAR (ClASsification criteria for Psoriatic ARthritis), a specific set of criteria for research (Table S1; see Supporting Information).11 Diagnosis of PsA using CASPAR requires peripheral or axial articular inflammatory disease or enthesitis and ≥ 3 points earned by assessing the following: evidence of skin lesions of psoriasis, past psoriasis, family history (the latter two weighted at half the points of a current psoriatic lesion), psoriatic nail dystrophy, rheumatoid factor negativity, dactylitis and specific juxta-articular radiological abnormalities.11 CASPAR was validated in patients with inflammatory arthritis and arthropathies.

Aetiology and pathogenesis The pathogenesis of PsA is complex, influenced by genetic, immunological and environmental factors. Familial aggregation of PsA was first noted in an English population.12 PsA certainly appears to be heritable – in Icelandic, Canadian and English populations, the risk in first-degree relatives of patients with PsA was 30–55-fold higher than for the general population.13– 15 Genetic and molecular techniques have established the association of PsA with the major histocompatibility complex, and several class I human leucocyte antigen (HLA) genes have been associated with specific PsA phenotypes. For example, HLAB27 has been associated with axial disease and HIV,16–19 while HLA-B38 and HLA-B39 have been associated with peripheral © 2013 British Association of Dermatologists

arthritis.20,21 Furthermore, PsA progression has been associated with HLA-B39 alone, HLA-B27 in the presence of HLA-DR7, and HLA-DQw3 in the absence of HLA-DR7.16–19 In contrast, patients with PsA with both HLA-Cw6 and HLA-DRB1*07 have been reported to have less severe PsA.22 The pathogenesis of psoriasis and PsA is mediated by inflammatory elements and the cytokine pathways they activate (Fig. 1).23,24 It has been proposed that the process leading to PsA starts at the enthesis (the point of insertion of tendons and ligaments in bone), where T cells, dendritic cells, mast cells, neutrophils and macrophages infiltrate the enthesium and synovium of joints in a manner similar to the processes that drive psoriatic skin lesions.25,26 Additionally, tumour necrosis factor (TNF)-a levels are elevated in the skin and synovium and are detectable in the plasma of patients with PsA and psoriasis.27,28 Recently, findings of increased Thelper (Th)17 cell counts in the circulation, synovial tissue and synovial fluid of patients with PsA have suggested that Th17 cells may be involved in the inflammatory response.24,29 Although the exact mechanisms of PsA are not yet elucidated, in models of autoimmune arthritis, overproduction of TNF-a, interleukin (IL)-17 and other inflammatory cytokines leads to osteoclastogenesis, bone resorption and cartilage destruction – changes that also occur in PsA.24,30,31 Additionally, studies conducted in a mouse model of spondyloarthropathy demonstrated that expression of IL-22 induces genes that upregulate entheseal and periosteal bone formation.32 Environmental factors also influence PsA development. Lifting heavy loads, bacterial infections, cigarette smoking and injuries may be associated with PsA;33,34 stressful life events have been identified as additional risk factors.35 Whether this mimics psoriasis onset following skin injury (Koebner response) remains to be elucidated.

Diagnostic and screening tools Psoriatic skin lesions usually precede the onset of joint symptoms; therefore, dermatologists are ideally situated to screen and diagnose PsA early in disease progression, thus mitigating many of its crippling elements. Dermatologists should ask their patients with psoriasis about signs and symptoms suggestive of an inflammatory arthritis like PsA, such as morning stiffness lasting > 30 min; typically, the arthritis symptoms of PsA improve with activity, but can present as hot, red, swollen joints.36 Dermatologists will want to query their patients about a family history of psoriasis or arthritis. During the physical exam, dermatologists should look for signs of PsA, such as joint swelling, dactylitis and enthesitis, as well as axial manifestations of PsA, such as pain, stiffness and range of motion limitations in the neck or back. Dermatologists may consider laboratory tests for some patients when screening for PsA. The acute-phase reactant C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) are elevated in approximately 50% of patients with PsA, and a lower level of ESR has been shown to reduce the risk of clinical damage progression.37,38 However, British Journal of Dermatology (2014) 170, pp772–786

774 Management of PsA, W.H. Boehncke et al.

Fig 1. Schematic presentation of immunological factors contributing to the pathogenesis of psoriatic arthritis. IL-12 induces differentiation of Th1 cells, while IL23 primarily stimulates proliferation and activation of Th17 cells, which are characterized by the expression of IL-17 A and F, IL-21 and IL-22.24 Cytokines produced by Th1 and Th17 cells induce the production of TNF-a in macrophages and other cell types, adding to inflammation. APC, antigenpresenting cell; ICAM, intercellular adhesion molecule; IFN, interferon; IL, interleukin; TGF, transforming growth factor; Th, T helper; TNF, tumour necrosis factor. Adapted with permission from Raychaudhuri24

normal ESR and CRP findings do not preclude a diagnosis of PsA. In addition, dermatologists should ask their patients with psoriasis about enthesopathies (e.g. ‘tennis elbow’ and Achilles tendonitis). Enthesopathies are frequently observed in patients with PsA and sometimes are the only presenting rheumatic manifestation.39,40 Furthermore, questionnaires such as the Psoriatic Arthritis Screening and Evaluation (PASE; Fig. S1),41 the Toronto Psoriatic Arthritis Screening (ToPAS; Fig. S2)42 and the Psoriatic Epidemiology Screening Tool (PEST; Fig. S3; all provided as Supporting Information)43 were developed and validated by the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) to facilitate diagnosis of PsA by dermatologists and rheumatologists (Table 1).41–49 Although these questionnaires allow early screening of PsA in

patients with psoriasis by dermatologists, recent validation studies demonstrated that these screening instruments have performed inconsistently with regard to specificity and sensitivity.47–49 Imaging is helpful for differential diagnosis, prognosis, monitoring disease progression and guiding therapy decisions. Imaging can be useful for diagnosis of axial disease (Fig. 2)50 and for establishing the severity of peripheral arthritis.51 Radiographs are useful for identifying typical proliferative and destructive bone lesions in PsA;52,53 baseline joint damage, if present, predicts further destruction.54 Ultrasonography and magnetic resonance imaging (MRI) are valuable for assessing synovial tissue, joint effusion, erosions, dactylitis, tenosynovitis and enthesitis.55–57 Imaging procedures are best

Table 1 Comparison of three screening tools for psoriatic arthritis

Based on Sensitivity Specificity Skin/nail assessment Axial disease ‘Active’ disease ‘Remission’ Marker of treatment Unique features

ToPAS42

PEST43

PASE41,45

Rheumatology, dermatology and methodology input 41–87%42,47–49 297–93%42,47–49 Yes Yes Yes Yes No Pictures of skin/nail involvement

Modification of PAQ by Alenius et al.46

De novo dermatology–rheumatology input and Delphi exercise 24–82%41,45,47–49 385–94%41,45,47–49 No Yes Yes No Yes Symptom and function subscales

275–92%43,47–49 372–98%43,47–49 Yes Yes Yes Yes No Figure of mannequin to indicate areas of soreness

PAQ, Psoriasis Assessment Questionnaire; PASE, Psoriatic Arthritis Screening and Evaluation; PEST, Psoriasis Epidemiology Screening Tool; ToPAS, Toronto Psoriatic Arthritis Screening. Adapted with permission from Qureshi et al.44

British Journal of Dermatology (2014) 170, pp772–786

© 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 775

quately reflecting PsA disease activity in patients with oligoarticular and distal interphalangeal disease. Several questionnaires were developed to evaluate QoL in patients with PsA. The modified Health Assessment Questionnaire (HAQ)-Skin consists of questions from the original Stanford HAQ, used for QoL assessments in rheumatic diseases,66,67 as well as questions about psoriasis severity and functional impact.68 However, the HAQ-Skin does not adequately address the dermatological, social or psychological impact of PsA; the Psoriatic Arthritis Quality of Life (PsAQoL) is a reliable, easily administered tool developed to address this shortcoming.69 The negative psychosocial effects of psoriasis on mental health and QoL are well documented, especially regarding feelings of stigmatization, stress, anxiety and depression.70 Patients with PsA reported more bodily pain and role limitations due to emotional distress than patients with RA.71 Similarly, patients reported that PsA negatively affected mental health and social functioning.72

Comorbidities

Fig 2. A patient with psoriatic arthritis exhibiting asymmetric marginal and nonmarginal syndesmophytes (white circle) randomly distributed along the lumbar tract of the spine. Used with permission from Scarpa et al.50

performed in early disease stages, in consultation with a rheumatologist; physicians should appreciate that a negative finding for PsA does not preclude later onset.

Assessment tools The Outcome MEasures in Rheumatoid Arthritis Clinical Trials (OMERACT) and GRAPPA consortium achieved consensus on six core domains that should be assessed in clinical trials for PsA: peripheral joint activity, skin activity, pain, patient’s global assessment, physical function and health-related quality of life (QoL).58 Treatment efficacy tools for PsA include the American College of Rheumatology (ACR) score, the modified Disease Activity Score (DAS), PsA response criteria (PsARC), Disease Activity index for Psoriatic Arthritis (DAPSA), PsA Joint Activity Index (PsAJAI) and Composite Psoriatic Disease Activity Index (CPDAI); see Table 2.41,59–64 Additionally, GRAPPA recently developed two new composite measures that incorporate the assessment of joints, entheses, skin, dactylitis and axial disease. The Psoriatic ArthritiS Disease Activity Score (PASDAS) and Arithmetic Mean of Desirability Functions (AMDF) composite measures were better able to discriminate between high and low disease activity than the CPDAI, DAPSA and DAS28; further validation testing is underway.65 ACR and DAS are the most widely used assessment tools in clinical trials of PsA; both were originally developed for the evaluation of rheumatoid arthritis (RA). The DAS28 is limited by not ade© 2013 British Association of Dermatologists

PsA was initially considered benign, but is now known to increase the risk of cardiovascular disease, cerebrovascular disease, type II diabetes, hyperlipidaemia, hypertension and mortality.73–75 Although the risk of cardiovascular disease is increased with psoriasis alone, the risk is greater in association with PsA.76,77 Patients with PsA exhibit signs of subclinical atherosclerosis, demonstrated by increased carotid intima– media thickness.78–80 There may be a link between increased prevalence of cardiovascular disease in patients with PsA and metabolic syndrome, which encompasses overlapping symptoms, including obesity, impaired glucose tolerance and hypertension. Metabolic syndrome increases the risk of developing type II diabetes and atherosclerotic cardiovascular disease; in one study, metabolic syndrome was present in 58% of patients with PsA.81 Obesity has been associated with an increased risk of developing PsA82,83 and may be associated with an unfavourable response to therapy.84,85 Additionally, elevated levels of triglycerides, total cholesterol, high-density cholesterol and glucose have been reported in patients with PsA with subclinical atherosclerosis.80 Increased mortality of patients with PsA may be compounded by chronic obstructive pulmonary disease (COPD), which is more prevalent in patients with established PsA than in those with early-stage PsA.86 Although an aetiological relationship between PsA and COPD has not been fully identified, evidence shows an interplay between smoking status, genetic makeup and the prevalence of PsA in patients with psoriasis.87 Thus, interventions aimed at lifestyle modifications for preventing obesity or smoking are areas in which dermatologists can make an important impact early in the course of PsA. An important question, too early for an answer, is whether markers of and interventional treatment for these comorbid states will alter the natural history of psoriasis and PsA. It is interesting to note that treatment of RA with an anti-TNF agent has been associated with decreases in N-terminal probrain natriuretic British Journal of Dermatology (2014) 170, pp772–786

776 Management of PsA, W.H. Boehncke et al. Table 2 Comparison of psoriatic arthritis treatment efficacy assessment tools41,59–64 Tool

Disease domain

Instrument

Score range

ACR

Peripheral arthritis

66 SJC; 68 TJC PtPain PtGDA MDGDA Acute-phase reactants PtPain PtGDA MDGDA PtPain PtGDA MDGDA PtFunc (e.g. HAQ) Mathematical equation involving Ritchie Articular Index, total number of swollen joints, erythrocyte sedimentation rate and general health assessment General health assessment Total score Symptom score

66/68 VAS 10 cm VAS 10 cm VAS 10 cm

Function score 66 SJC; 68 TJC HAQ Acute-phase reactants Pt assessment Erythema Scaling MDGDA Enthesopathy index Tender/swollen digits Spondylitis Articular Index PtPain assessment 66 SJC; 68 TJC PtGA PtPain CRP Simplified instrument using weighted measures of a 30% improvement in each of: JNT, CRP, MDGDA, PtGDA, PtPain and HAQ MDGDA PtGDA PtPain MDGDA PtGDA PtPain HAQ 66 SJC; 68 TJC HAQ PASI DLQI Leeds enthesitis score HAQ Digit score HAQ BASDAI ASQoL

8–40 0–4 0–3

Dactylitis

Spinal disease

DAS

Physical function/disability Peripheral arthritis

Physical function/disability PASE

PsARC

Peripheral arthritis Enthesitis Dactylitis Spinal disease Physical function/disability Peripheral arthritis

Skin disease

Enthesitis Dactylitis Spinal disease DAPSAa

Peripheral arthritis

PsAJAI

Peripheral arthritis

Dactylitis

Spinal disease

CPDAI

Physical function/disability Peripheral arthritis Skin disease Enthesitis Dactylitis

Spinal disease

VAS VAS VAS VAS VAS VAS 0–3

10 10 10 10 10 10

cm cm cm cm cm cm

15–75 7–35

0–4 0–4 0–4 0–6 0–4 0–4 0–4 VAS 10 cm 66/68

0–9

VAS 10 VAS 10 VAS 10 VAS 10 VAS 10 VAS 10 0–3 66/68 0–3 0–72 0–30 0–6 0–3 0–20 0–3 0–10 0–18

cm cm cm cm cm cm

ACR, American College of Rheumatology; ASQoL, Ankylosing Spondylitis Quality of Life; BASDAI, Bath Ankylosing Spondylitis Disease Activity Index; CPDAI, Composite Psoriatic Disease Activity Index; CRP, C-reactive protein; DAPSA, Disease Activity Index for Psoriatic Arthritis; DAS, Disease Activity Score; DLQI, Dermatology Life Quality Index; HAQ, Health Assessment Questionnaire; JNT, combined tender and swollen joint count scores; MDGDA, Physician Global Disease Activity Assessment; PASI, Psoriasis Area and Severity Index; PsAJAI, Psoriatic Arthritis Joint Activity Index; PsARC, psoriatic arthritis response criteria; Pt, Patient; PtFunc, Patient Function Assessment; PtGA, Patient Global Assessment; PtGDA, Patient Global Disease Activity Assessment; PtPain, Patient Pain Assessment; SJC, swollen joint count; TJC, tender joint count; VAS, visual analogue scale. aDomain

specifically indicated as peripheral arthritis only.

British Journal of Dermatology (2014) 170, pp772–786

© 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 777

peptide, a biomarker for cardiovascular risk.88 A large retrospective study of patients from the Consortium of Rheumatology Researchers of North America (CORRONA) registry points to improvement with disease-modifying therapies, especially the anti-TNF agents, which were shown to modulate positively the risk and incidence of cardiovascular events.89

Treatment Disease severity influences the selection of an appropriate PsA treatment. Nonsteroidal anti-inflammatory drugs (NSAIDs) and physical therapy are commonly used to treat mild symptoms. The presence of PsA significantly impacts treatment decisions for patients with psoriasis, as several drugs indicated for RA may be ineffective for psoriatic skin lesions or even worsen symptoms. For example, systemic corticosteroids may be prescribed for short-term management of PsA, but are not recommended by dermatologists due to the risk of severe flares of psoriasis on treatment withdrawal. Likewise, some drugs, such as ciclosporin (see below), are very effective treatments for

psoriasis but are generally not effective in the treatment of most forms of PsA. Furthermore, the chronic nature of PsA and the increased risk of cardiovascular comorbidities necessitate comprehensive long-term management.90

Traditional nonbiologic disease-modifying antirheumatic drugs The American Academy of Dermatology, European League Against Rheumatism and GRAPPA have established treatment guidelines for PsA, including some therapies not currently approved for PsA (Table 3).51,91,92 Nonbiologic disease-modifying antirheumatic drugs (DMARDs) are recommended for patients with moderate-to-severe PsA. The synthetic DMARD methotrexate reduces inflammation by increasing the release of adenosine, a potent anti-inflammatory autocoid, at sites of inflammation.93 Methotrexate is commonly prescribed as an initial treatment for moderate-to-severe PsA; however, conflicting evidence exists regarding its efficacy in PsA disease modification. Methotrexate reduces joint tenderness and

Table 3 Available and emerging treatments for psoriatic arthritis

Drug

Class

Treatments approved for PsA Nonbiologic DMARD Methotrexate105,106 Leflunomide159 Nonbiologic DMARD Adalimumab101,106 TNF inhibitor Etanercept106,122 TNF inhibitor

Region(s) where approved for PsA

Golimumab106,123 Infliximab106,124

TNF inhibitor TNF inhibitor

Canada, Europe Europe U.S.A., Europe, Japan U.S.A., Canada Europe U.S.A., Europe, Canada U.S.A., Europe, Canada

Certolizumab pegol159,167

TNF inhibitor

U.S.A.

Ustekinumab155,159

IL-12/23 inhibitor

U.S.A., Europe

Treatments not approved for PsA Sulfasalazine116,159 Nonbiologic DMARD Ciclosporin A121,159 Nonbiologic DMARD Abatacept159,168 T-cell costimulatory inhibitor

Not applicable Not applicable Not applicable

Tocilizumab159,169 Secukinumab159–161 Brodalumab159 Apremilast164 Tofacitinib159,165,170

IL-6R inhibitor IL-17 inhibitor IL-17R inhibitor PDE-4 inhibitor Selective JAK inhibitor

Not Not Not Not Not

applicable applicable applicable applicable applicable

Dose Up to 25 mg per week (oral) 20 mg per day (oral) 40 mg SC every other week 50 mg SC every week 25 mg SC twice per week 50 mg SC every month 5 mg kg1 body weight IV at weeks 0, 2 and 6, and every 8 weeks thereafter 400 mg SC at weeks 0, 2 and 4, followed by 200 mg every other week; for maintenance dosing, can consider 400 mg every 4 weeks 45 mg SC initially and 4 weeks later, followed by 45 mg every 12 weeks. For patients > 100 kg with coexistent moderate-to-severe plaque psoriasis: 90 mg SC initially and 4 weeks later, followed by 90 mg every 12 weeks 2–3 g per day (oral) in two divided dosesa 25 mg kg1 per day in two divided dosesa,b 10 mg kg1 2 and 4 weeks after first infusion, then once per month IV or a single infusion loading dose (10 mg kg1), followed by 125 mg SC within 1 day, and 125 mg every week thereafterc 4–8 mg kg1 IV every 4 weeksc Up to 300 mg per month (SC)d 70–210 mg (SC) every 2 weeks or 280 mg (SC) monthlye 20 mg twice daily or 40 mg once daily (oral) 5 mg twice daily (oral)f

DMARD, disease-modifying antirheumatic drug; IL, interleukin; IV, intravenous; JAK, Janus kinase; PDE, phosphodiesterase; PsA, psoriatic arthritis; RA, rheumatoid arthritis; SC, subcutaneous; TNF, tumour necrosis factor. aDose approved by the Food and Drug Administration (FDA) for RA; bdose approved by the FDA for psoriasis; cdose approved by the European Medicines Agency for RA; dinvestigational dose for RA; einvestigational dose for psoriasis; fdose approved by the FDA for RA.

© 2013 British Association of Dermatologists

British Journal of Dermatology (2014) 170, pp772–786

778 Management of PsA, W.H. Boehncke et al.

swelling and suppresses skin manifestations, but the impact of methotrexate on radiographic progression remains unclear.94–96 More recently, the placebo-controlled Methotrexate in PsA (MIPA) study failed to demonstrate a significant advantage of methotrexate in modifying disease activity at 6 months, although global symptom measures improved.97 However, the MIPA study may have been underpowered, and the minimum entry criteria allowed for the inclusion of patients with active disease in fewer joints than in some other PsA trials. Another limitation of the MIPA study was the rate of patient withdrawal, which may help to explain the negative results. In contrast, methotrexate administered early in disease progression reduced joint inflammation compared with baseline in 68% of patients in a longitudinal cohort study.98 A recent meta-analysis reported that the addition of methotrexate to anti-TNF therapy in patients with immune-mediated inflammatory diseases reduced the development of antibodies against anti-TNF agents.99 Furthermore, methotrexate increases the serum concentration of infliximab and adalimumab.100,101 Methotrexate use has also been associated with a significantly decreased risk of cardiovascular disease in patients with psoriasis or RA.102,103 Therefore, adding methotrexate to TNF-inhibitor therapy may provide additional clinical benefit. Methotrexate is associated with the potential for a range of serious adverse effects.104–107 Methotrexate is a teratogen and abortifacient; it is contraindicated in women who are pregnant, attempting to conceive, or breastfeeding an infant. Methotrexate is also contraindicated in patients with alcoholism, alcoholic liver disease, other chronic liver disease, immunodeficiency or blood dyscrasias. Methotrexate should not be given to patients with renal impairment, hepatitis, cirrhosis, leuco-penia or thrombocytopenia. Acute toxicity due to drug– drug interactions may occur when methotrexate is administered to patients who are taking sulfamethoxazole plus trimethoprim, as well as some types of NSAIDs. A number of other substances may also interact with methotrexate; these include penicillins and many other antibiotics, hepatotoxic agents, theophylline and preparations of folate or its derivatives [e.g. folinic acid (leucovorin)]. Methotrexate-induced hepatotoxicity is a risk for patients with PsA, especially during long-term treatment.108 The consensus among dermatologists is to stratify patients on methotrexate as low risk or high risk. The latter category includes patients with pre-existing liver disease or other risk factors such as nonalcoholic steatohepatitis (NASH), excessive alcohol consumption, obesity and diabetes.91,104,109 NASH and nonalcoholic fatty liver disease associated with psoriasis (but not with RA) may confound identification of methotrexate hepatotoxicity.104,109 Liver function tests should be performed every 1–3 months in low-risk patients, and more often when starting therapy or changing the dose; monitoring procollagen peptide III is common outside the U.S.A.91,104 Opinions differ on whether the value of information from liver biopsy outweighs the risks of the procedure. Guidelines for PsA and psoriasis issued by the American Academy of Dermatology recommend considering liver biopsies for patients at risk for British Journal of Dermatology (2014) 170, pp772–786

methotrexate-induced hepatotoxicity.91 Biopsy may be warranted in high-risk patients after a cumulative oral methotrexate dose of 10–15 g and in low-risk patients after 35– 40 g.91,104 In contrast, the ACR guidelines for the use of methotrexate in RA do not recommend liver biopsy.110 Methotrexate may lead to myelosuppression and (more rarely) bone marrow failure and pancytopenia, although the chance of these complications is greater in patients with existing risk factors. The immunosuppressive nature of methotrexate therapy has been associated with developing malignancies. Lung disease such as pneumonitis may also occur during methotrexate therapy. Less serious but more common are gastrointestinal issues (ulcerative colitis, nausea, anorexia and stomatitis), leucopenia, fatigue, chills and fever, dizziness, susceptibility to infection, and malaise, which can sometimes be alleviated by folate supplementation or altering the route, number or timing of methotrexate doses. Although not approved for treating PsA, sulfasalazine has been examined in several clinical studies. Patients with PsA treated for 6 months with sulfasalazine showed improvement in clinical measures, including morning stiffness, number of painful joints, articular score and grip strength compared with placebo; however, skin manifestations did not improve.111–114 Most adverse events (AEs) associated with sulfasalazine were mild. In contrast, a subsequent small study suggested that sulfasalazine may not alter radiographic progression in PsA; additionally, due to gastrointestinal AEs, treatment was not well tolerated.115 Adverse gastrointestinal effects may make sulfasalazine treatment intolerable for some patients, although the incidence may be reduced at doses < 4 g per day. The use of sulfasalazine is contraindicated in patients with intestinal or urinary obstruction; porphyria (due to the risk of an acute attack); and hypersensitivity to sulfasalazine, its metabolites, other sulfonamide drugs, or salicylates.116 Caution must be used in treating patients who are breastfeeding and those with hepatic or renal damage, severe allergies or bronchial asthma, and blood dyscrasias. The anti-inflammatory drug leflunomide exhibited improvements in PsARC response and QoL compared with placebo in patients with PsA117 and resulted in modest but significant improvement in Psoriasis Area and Severity Index (PASI) scores and target lesion response.118 Due to the strong potential for fetal death and birth defects, leflunomide is contraindicated in pregnant women and women who could conceive, unless reliable contraception is used.119 Severe and occasionally fatal liver injury has occurred in some patients; therefore, leflunomide should not be given to patients who have existing liver disease or alanine aminotransferase > 2 9 the upper limit of normal. Caution is advised when leflunomide is coadministered with drugs, such as methotrexate, that also may be hepatotoxic, and in patients with chronic renal insufficiency. Leflunomide is a potential immunosuppressant and is not recommended in the settings of severe immunodeficiency, bone marrow dysplasia or severe, uncontrolled infections. Diarrhoea, nausea and hypertension were the most common AEs associated with leflunomide in clinical trials. © 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 779

Ciclosporin A (CsA) has demonstrated clinical benefit for psoriatic skin lesions, peripheral arthritis and axial symptoms in patients with PsA.120 However, direct evidence that CsA reduces radiographic damage is lacking. CsA is contraindicated in patients undergoing treatment with phototherapy, immunosuppressants (e.g. methotrexate), coal tar or radiation; it is also contraindicated in patients with abnormal renal function, uncontrolled hypertension and malignancies.121 CsA should not be given to women who are breastfeeding and should be used to treat a pregnant woman only if the benefits to her outweigh the risks to the fetus, which include premature birth and low weight. CsA has been placed in pregnancy category C. CsA can cause nephrotoxicity and renal dysfunction; thus, close monitoring of renal function is needed, particularly in elderly patients, in order to guide dose adjustments and avoid structural kidney damage. CsA can also cause hepatotoxicity. Blood pressure and laboratory parameters should be evaluated before starting treatment, monitored every other week during months 1–3, and checked every month thereafter. Similar to other immunosuppressive agents, there is an increased risk of lymphoproliferative malignancies and skin cancers (e.g. squamous and basal cell carcinomas) with CsA treatment. CsA is metabolized by cytochrome P450 3A enzymes and interacts with many substances, including calcium channel blockers, antibiotics, antifungals and grapefruit juice.121 The primary AEs associated with CsA therapy include hypertension, headaches, tremors, immunosuppression and nausea; other AEs include gum hyperplasia, hyperlipidaemia and hypertrichosis.

Biologic disease-modifying antirheumatic drugs Five TNF inhibitors-adalimumab, etanercept, golimumab, infliximab and certolizumab pegol-and one IL-12/IL-23 inhibitor, ustekinumab, are currently approved for the treatment of PsA (Table 3). TNF inhibitors improve cutaneous psoriasis, peripheral arthritis, QoL and function. Importantly, they slow or prevent structural damage in peripheral joints. In contrast to traditional DMARDs, TNF inhibitors exhibit efficacy for enthesitis, dactylitis and axial disease. Adalimumab, etanercept, golimumab, infliximab and certolizumab pegol share a number of safety considerations because of their common mechanism of action. Anti-TNF agents elevate the risk of opportunistic infections in general and have been associated with serious infections that can result in hospitalization or death.101,122–124 Treatment should not be started in a patient with an active infection; live vaccines should not be given concomitantly. The risk of infection may be greater in patients who are > 65 years of age or have weakened immunity due to comorbid disease or concomitant medications (e.g. methotrexate or corticosteroids). Besides new infections, reactivation of latent tuberculosis and hepatitis B virus infections may occur; all patients should be screened for tuberculosis and hepatitis B virus exposure. All five anti-TNF agents have been placed in Pregnancy Category B by the U.S. Food and Drug Administration and should be © 2013 British Association of Dermatologists

used in pregnant women only if clearly needed; it is unknown whether breastfeeding while on therapy is safe for the infant. Cautious consideration of risk and benefit is advised when considering adalimumab or other anti-TNF agents for patients with a history of malignancy due to associations with increased risk of developing malignancies, particularly lymphoma and skin cancer (mainly basal cell carcinoma and squamous cell carcinoma). In adolescents, cases of aggressive hepatosplenic T-cell lymphoma have been observed, although nearly all were in patients with Crohn disease or ulcerative colitis who were taking other immunosuppressants concurrently. Caution is advised in treating patients with demyelinating syndromes (e.g. multiple sclerosis) or congestive heart failure, due to the chance of new onset or exacerbation of symptoms. Psoriasis worsening or new onset may occur on beginning treatment with an anti-TNF agent. On rare occasions, a lupus-like disorder may result when autoantibodies form. Adalimumab is a fully human, anti-TNF monoclonal antibody. In the Adalimumab Effectiveness in Psoriatic Arthritis Trial (ADEPT), treatment with adalimumab resulted in significantly higher ACR20 response rates and significant inhibition of radiographic structural damage compared with placebo after 24 weeks.125 Radiographic damage was inhibited and clinical improvements were maintained in most patients at 48 and 144 weeks.126,127 Additionally, adalimumab significantly reduced the extent of psoriatic skin lesions and improved function and QoL.125,128 Adalimumab is generally well tolerated, with most AEs being mild to moderate in severity. Injection-site reactions were common in clinical trials, but most were mild, and they usually did not require the discontinuation of treatment.101 During 2 years of adalimumab exposure, the most common AEs were upper respiratory tract infection (URI), nasopharyngitis and sinusitis; serious infections and malignancies were observed at lower rates.127 Etanercept (fusion protein of soluble TNF receptor linked to human immunoglobulin Fc) is a receptor decoy and was the first TNF inhibitor reported to benefit patients with PsA. In a 12-week study, etanercept demonstrated efficacy for clinical measures of PsA and psoriasis, with significantly greater numbers of patients treated with etanercept achieving PsARC, ACR20 and PASI responses compared with placebo.129 In another study, etanercept significantly reduced disease activity, improved joint symptoms, reduced psoriatic lesions, and inhibited radiographic progression in patients with PsA.130 Two-year follow-up studies demonstrated sustained inhibition of radiographic progression and improved functional outcomes.131,132 Patients randomized to two different etanercept treatment regimens in the Psoriasis Randomized Etanercept Study in Subjects with Psoriatic Arthritis (PRESTA) trial exhibited significant improvements in skin, joints and QoL.133,134 A small observational study showed that etanercept improved axial manifestations of PsA.135 Etanercept was well tolerated in clinical studies; most AEs were mild to moderate. The most common AEs were injection-site reactions and URI.130,134 Golimumab is a fully human monoclonal anti-TNF antibody. In patients with active PsA (GO-REVEAL study), 48% of British Journal of Dermatology (2014) 170, pp772–786

780 Management of PsA, W.H. Boehncke et al.

patients receiving golimumab achieved an ACR20 response after 14 weeks, compared with a significantly smaller proportion (9%) receiving placebo.136 Golimumab improved skin and nail manifestations, physical function and QoL. Improvements in disease activity were maintained with longer treatment (52 weeks); additionally, golimumab inhibited radiological progression.137 AEs were similar to those associated with other TNF inhibitors and included nasopharyngitis and URI.136 The dosing and efficacy of golimumab for psoriasis have not yet been adequately studied. The Infliximab Multinational Psoriatic Arthritis Controlled Trial (IMPACT) demonstrated the significant benefit of infliximab (chimeric anti-TNF monoclonal antibody) over placebo for improving measures of arthritis, dactylitis, enthesitis and psoriasis after 14–50 weeks.138–140 Longer treatment resulted in sustained improvement in joint and skin symptoms and inhibition of radiographic progression.141,142 Most AEs associated with infliximab were mild to moderate. After 2 years of treatment, the most common AE was URI.141 Infliximab is the only anti-TNF agent that is given via intravenous infusion and administered on a dose-per-weight basis. Infliximab doses of >5 mg kg1 are contraindicated in patients with moderate-tosevere heart failure.124 Infliximab is also contraindicated in patients with hypersensitivity to murine proteins. Infusion reactions occurred in 18% of patients treated with infliximab in phase III studies; however, < 1% of patients experienced a serious infusion reaction. Certolizumab pegol (pegylated Fab fragment of a humanized anti-TNF monoclonal antibody) demonstrated efficacy compared with placebo in patients with RA or psoriasis,143,144 and was recently approved for the treatment of PsA. In the RAPIDPsA study, ACR20 response rates were superior in patients treated with certolizumab pegol compared with placebo at weeks 12 and 24.145 Physical function was significantly improved after 24 weeks of treatment with certolizumab pegol, and improvements in psoriatic skin involvement, nail disease, enthesitis and dactylitis were also observed. Among patients treated with certolizumab pegol, the most common infectious AEs were nasopharyngitis and URI, while diarrhoea and headache were the most common noninfectious AEs. No head-to-head studies have compared anti-TNF agents for the treatment of PsA. Consequently, there have been attempts to determine the relative efficacy of these therapies via observational studies and analyses of separate well-controlled trials.120,146–151 Most research has suggested that improvements in joint manifestations of PsA were similar among the antiTNF agents. In several analyses, infliximab was more effective than etanercept in achieving PASI responses.120,147,149 Some analyses have shown that adalimumab may also be more effective than etanercept in achieving PASI responses.120,149,152 The certainty of these results is limited by the open-label nature of observational studies and by the heterogeneity of meta-analyses due to varying study end points, treatment duration and patient characteristics. Ustekinumab is a human monoclonal antibody that was recently approved for the treatment of PsA. It targets the p40 British Journal of Dermatology (2014) 170, pp772–786

subunit shared by IL-12 and IL-23, thus blocking its interaction with the receptor and inhibiting downstream signalling pathways, differentiation and cytokine production. Ustekinumab improved joint tenderness, enthesitis, HAQ and PASI measures compared with placebo in patients with PsA.153 Most recently, the 1-year PSUMMIT 1 study demonstrated significantly more ACR20 responses at week 24 in patients with PsA who received ustekinumab 45 or 90 mg compared with placebo.154 Ustekinumab was generally well tolerated, with the severity of the most common AEs reported as mild to moderate; the most frequent AEs were URI, nasopharyngitis and headache.153 Potentially serious reactivation of previous infections is possible with ustekinumab; it should not be given to patients with clinically important active infections, and caution is needed in patients with a history of recurrent infections.155 Patients should be checked for active tuberculosis infection before starting ustekinumab. They should not be given live vaccines and should avoid other people who have been immunized recently with live vaccines. Genetic deficiency in IL-12 and IL-23 is associated with disseminated infections caused by mycobacteria, salmonella and bacillus Calmette–Guerin, but it is unknown whether IL blockade with ustekinumab carries the same risks. As with the TNF inhibitors, ustekinumab is classified as a Pregnancy Category B agent and should be used in pregnant women only if the benefits outweigh risks to the fetus; caution is advised when nursing, due to a lack of data on the presence of ustekinumab in human breast milk and its effects on infants. Ustekinumab may also increase the risk of malignancies; rapidly developing squamous cell carcinomas were observed in patients with risk factors during postmarketing surveillance. A single case of reversible posterior leucoencephalopathy syndrome occurred during ustekinumab treatment; the patient recovered with appropriate treatment.

Emerging treatments Other biologics and small-molecule compounds are under development to treat PsA (Table 3). Some of these compounds have mechanisms of action that do not target TNF and several have shown promising results in phase II and III trials. Abatacept blocks interactions between antigen-producing cells and T cells, thus inhibiting T-cell activation. A placebocontrolled phase II trial of abatacept in patients with PsA demonstrated significant improvements in ACR20 response rate, MRI-monitored joint erosion and synovitis, PASI score and QoL.156 The IL-6 inhibitors tocilizumab and ALD518 have demonstrated efficacy and tolerability in the treatment of RA in clinical trials.157,158 The IL-17 inhibitors secukinumab and ixekizumab, as well as brodalumab (which, in general, blocks the IL-17 family of cytokines from binding to the IL-17 receptor) have shown efficacy in the treatment of psoriasis and RA.159–161 Unexpectedly, secukinumab had limited efficacy for PsA in a phase II study;162 a more definitive phase II or III trial will need to be completed to determine whether this is indeed the case. © 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 781

Apremilast (phosphodiesterase-4 inhibitor) and tofacitinib (selective Janus kinase inhibitor) function by inhibiting cytokine signalling pathways. Apremilast has demonstrated clinical benefit in the treatment of psoriasis;163 results of a phase II study in patients with PsA have been reported164 and larger trials are underway. Tofacitinib has recently been approved for the treatment of RA.165

Conclusions PsA is among the most significant comorbidities associated with psoriasis, resulting in pain and deformities that limit function and reduce QoL. Additionally, patients with PsA are at greater risk for developing metabolic syndrome and cardiovascular disease and should be routinely monitored for cardiovascular risk factors.90 PsA may lead to progressive joint deterioration, which treatment may slow or prevent. Because psoriasis often presents years before the onset of PsA, the dermatologist is uniquely positioned to recognize the early signs of PsA. In many cases, dermatologists may choose to work closely with rheumatologists to minimize symptoms, slow disease progression and maximize QoL. Many treatment options are currently approved for PsA, with biologic DMARDs producing significant improvements in disease activity and progression. Interestingly, antiTNF therapy was recently shown to improve cardiovascular comorbidities associated with PsA,166 suggesting clinical benefit beyond treating joint symptoms and supporting a comprehensive approach to long-term care.90 Several additional biologics targeting inflammatory pathways are currently under investigation for the treatment of PsA.

Acknowledgments AbbVie funded development of the publication by Complete Publication Solutions (CPS). Amanda L Sheldon PhD, of CPS, provided medical writing and editorial support to the authors in the development of this publication. AbbVie had no role in content development of the publication. All decisions regarding content were made by the authors.

References 1 Gelfand JM, Gladman DD, Mease PJ et al. Epidemiology of psoriatic arthritis in the population of the United States. J Am Acad Dermatol 2005; 53:573–7. 2 Ibrahim G, Waxman R, Helliwell PS. The prevalence of psoriatic arthritis in people with psoriasis. Arthritis Rheum 2009; 61: 1373–8. 3 Prey S, Paul C, Bronsard V et al. Assessment of risk of psoriatic arthritis in patients with plaque psoriasis: a systematic review of the literature. J Eur Acad Dermatol Venereol 2010; 24 (Suppl. 2): 31–5. 4 Gottlieb AB, Kircik L, Eisen D et al. Use of etanercept for psoriatic arthritis in the dermatology clinic: the Experience Diagnosing, Understanding Care, and Treatment with Etanercept (EDUCATE) study. J Dermatolog Treat 2006; 17:343–52. 5 Gladman DD, Shuckett R, Russell ML et al. Psoriatic arthritis (PSA) – an analysis of 220 patients. Q J Med 1987; 62:127–41. © 2013 British Association of Dermatologists

6 Wilson FC, Icen M, Crowson CS et al. Incidence and clinical predictors of psoriatic arthritis in patients with psoriasis: a population-based study. Arthritis Rheum 2009; 61:233–9. 7 Torre Alonso JC, Rodriguez Perez A, Arribas Castrillo JM et al. Psoriatic arthritis (PA): a clinical, immunological and radiological study of 180 patients. Br J Rheumatol 1991; 30:245–50. 8 Kane D, Stafford L, Bresnihan B et al. A prospective, clinical and radiological study of early psoriatic arthritis: an early synovitis clinic experience. Rheumatology (Oxford) 2003; 42:1460–8. 9 Colombo E, Latiano A, Palmieri O et al. Enteropathic spondyloarthropathy: a common genetic background with inflammatory bowel disease? World J Gastroenterol 2009; 15:2456–62. 10 Arvikar SL, Fisher MC. Inflammatory bowel disease associated arthropathy. Curr Rev Musculoskelet Med 2011; 4:123–31. 11 Taylor W, Gladman D, Helliwell P et al. Classification criteria for psoriatic arthritis: development of new criteria from a large international study. Arthritis Rheum 2006; 54:2665–73. 12 Moll JM, Wright V. Familial occurrence of psoriatic arthritis. Ann Rheum Dis 1973; 32:181–201. 13 Karason A, Love TJ, Gudbjornsson B. A strong heritability of psoriatic arthritis over four generations – the Reykjavik Psoriatic Arthritis Study. Rheumatology (Oxford) 2009; 48:1424–8. 14 Chandran V, Schentag CT, Brockbank JE et al. Familial aggregation of psoriatic arthritis. Ann Rheum Dis 2009; 68:664–7. 15 Rahman P, Elder JT. Genetic epidemiology of psoriasis and psoriatic arthritis. Ann Rheum Dis 2005; 64 (Suppl. 2):ii37–9; discussion ii40–1. 16 Fife DJ, Waller JM, Jeffes EW et al. Unraveling the paradoxes of HIV-associated psoriasis: a review of T-cell subsets and cytokine profiles. Dermatol Online J 2007; 13:4. 17 Winchester R, Minevich G, Steshenko V et al. HLA associations reveal genetic heterogeneity in psoriatic arthritis and in the psoriasis phenotype. Arthritis Rheum 2012; 64:1134–44. 18 Zachariae H, Overgaard Petersen H, Kissmeyer Nielsen F, Lamm L. HL-A antigens in pustular psoriasis. Dermatologica 1977; 154:73–7. 19 Duvic M. Immunology of AIDS related to psoriasis. J Invest Dermatol 1990; 95:38S–40S. 20 Gladman DD, Anhorn KA, Schachter RK et al. HLA antigens in psoriatic arthritis. J Rheumatol 1986; 13:586–92. 21 Gladman DD, Farewell VT. The role of HLA antigens as indicators of disease progression in psoriatic arthritis. Multivariate relative risk model. Arthritis Rheum 1995; 38:845–50. 22 Ho PY, Barton A, Worthington J et al. HLA-Cw6 and HLADRB1*07 together are associated with less severe joint disease in psoriatic arthritis. Ann Rheum Dis 2007; 66:807–11. 23 Torti DC, Feldman SR. Interleukin-12, interleukin-23, and psoriasis: current prospects. J Am Acad Dermatol 2007; 57:1059–68. 24 Raychaudhuri SP. Role of IL-17 in psoriasis and psoriatic arthritis. Clin Rev Allergy Immunol 2012; 44:183–93. 25 Rottman JB, Smith TL, Ganley KG et al. Potential role of the chemokine receptors CXCR3, CCR4, and the integrin alphaEbeta7 in the pathogenesis of psoriasis vulgaris. Lab Invest 2001; 81:335– 47. 26 Kruithof E, Baeten D, De Rycke L et al. Synovial histopathology of psoriatic arthritis, both oligo- and polyarticular, resembles spondyloarthropathy more than it does rheumatoid arthritis. Arthritis Res Ther 2005; 7:R569–80. 27 Ritchlin C, Haas-Smith SA, Hicks D et al. Patterns of cytokine production in psoriatic synovium. J Rheumatol 1998; 25:1544– 52. 28 Ettehadi P, Greaves MW, Wallach D et al. Elevated tumour necrosis factor-alpha (TNF-alpha) biological activity in psoriatic skin lesions. Clin Exp Immunol 1994; 96:146–51.

British Journal of Dermatology (2014) 170, pp772–786

782 Management of PsA, W.H. Boehncke et al. 29 Jandus C, Bioley G, Rivals JP et al. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum 2008; 58:2307–17. 30 Koshy PJ, Henderson N, Logan C et al. Interleukin 17 induces cartilage collagen breakdown: novel synergistic effects in combination with proinflammatory cytokines. Ann Rheum Dis 2002; 61:704–13. 31 Moran EM, Mullan R, McCormick J et al. Human rheumatoid arthritis tissue production of IL-17A drives matrix and cartilage degradation: synergy with tumour necrosis factor-alpha, oncostatin M and response to biologic therapies. Arthritis Res Ther 2009; 11:R113. 32 Sherlock JP, Joyce-Shaikh B, Turner SP et al. IL-23 induces spondyloarthropathy by acting on ROR-gammat+ CD3+CD4–CD8– entheseal resident T cells. Nat Med 2012; 18:1069–76. 33 Eder L, Law T, Chandran V et al. Association between environmental factors and onset of psoriatic arthritis in patients with psoriasis. Arthritis Care Res (Hoboken) 2011; 63:1091–7. 34 Li W, Han J, Qureshi AA. Smoking and risk of incident psoriatic arthritis in US women. Ann Rheum Dis 2012; 71:804–8. 35 Pattison E, Harrison BJ, Griffiths CE et al. Environmental risk factors for the development of psoriatic arthritis: results from a case–control study. Ann Rheum Dis 2008; 67:672–6. 36 Garg A, Gladman D. Recognizing psoriatic arthritis in the dermatology clinic. J Am Acad Dermatol 2010; 63:733–48; quiz 749–50. 37 Gladman DD, Farewell VT, Nadeau C. Clinical indicators of progression in psoriatic arthritis: multivariate relative risk model. J Rheumatol 1995; 22:675–9. 38 Punzi L, Podswiadek M, Sfriso P et al. Pathogenetic and clinical rationale for TNF-blocking therapy in psoriatic arthritis. Autoimmun Rev 2007; 6:524–8. 39 Frediani B, Falsetti P, Storri L et al. Ultrasound and clinical evaluation of quadricipital tendon enthesitis in patients with psoriatic arthritis and rheumatoid arthritis. Clin Rheumatol 2002; 21:203–6. 40 Sadek HA, Abdel-Nasser AM, El-Amawy TA et al. Rheumatic manifestations of psoriasis. Clin Rheumatol 2007; 26:488–98. 41 Husni ME, Meyer KH, Cohen DS et al. The PASE questionnaire: pilot-testing a psoriatic arthritis screening and evaluation tool. J Am Acad Dermatol 2007; 57:581–7. 42 Gladman DD, Schentag CT, Tom BD et al. Development and initial validation of a screening questionnaire for psoriatic arthritis: the Toronto Psoriatic Arthritis Screen (ToPAS). Ann Rheum Dis 2009; 68:497–501. 43 Ibrahim GH, Buch MH, Lawson C et al. Evaluation of an existing screening tool for psoriatic arthritis in people with psoriasis and the development of a new instrument: the Psoriasis Epidemiology Screening Tool (PEST) questionnaire. Clin Exp Rheumatol 2009; 27:469–74. 44 Qureshi AA, Dominguez P, Duffin KC et al. Psoriatic arthritis screening tools. J Rheumatol 2008; 35:1423–5. 45 Dominguez PL, Husni ME, Holt EW et al. Validity, reliability, and sensitivity-to-change properties of the psoriatic arthritis screening and evaluation questionnaire. Arch Dermatol Res 2009; 301:573–9. 46 Alenius GM, Stenberg B, Stenlund H et al. Inflammatory joint manifestations are prevalent in psoriasis: prevalence study of joint and axial involvement in psoriatic patients, and evaluation of a psoriatic and arthritic questionnaire. J Rheumatol 2002; 29:2577– 82. 47 Coates LC, Aslam T, Al Balushi F et al. Comparison of three screening tools to detect psoriatic arthritis in patients with psoriasis (CONTEST study). Br J Dermatol 2013; 168:802–7. 48 Haroon M, Kirby B, Fitzgerald O. High prevalence of psoriatic arthritis in patients with severe psoriasis with suboptimal perfor-

British Journal of Dermatology (2014) 170, pp772–786

49

50 51 52 53

54

55 56

57 58 59

60

61

62

63

64

65

66

67 68

69

mance of screening questionnaires. Ann Rheum Dis 2013; 72:736– 40. Walsh JA, Callis Duffin K, Krueger GG et al. Limitations in screening instruments for psoriatic arthritis: a comparison of instruments in patients with psoriasis. J Rheumatol 2013; 40:287–93. Scarpa R, Peluso R, Atteno M. Clinical presentation of psoriatic arthritis. Reumatismo 2007; 59 (Suppl. 1):49–51. Ritchlin CT, Kavanaugh A, Gladman DD et al. Treatment recommendations for psoriatic arthritis. Ann Rheum Dis 2009; 68:1387–94. Siannis F, Farewell VT, Cook RJ et al. Clinical and radiological damage in psoriatic arthritis. Ann Rheum Dis 2006; 65:478–81. De Simone C, Caldarola G, D’Agostino M et al. Usefulness of ultrasound imaging in detecting psoriatic arthritis of fingers and toes in patients with psoriasis. Clin Dev Immunol 2011; 2011:390726. Bond SJ, Farewell VT, Schentag CT et al. Predictors for radiological damage in psoriatic arthritis: results from a single centre. Ann Rheum Dis 2007; 66:370–6. Ory PA, Gladman DD, Mease PJ. Psoriatic arthritis and imaging. Ann Rheum Dis 2005; 64 (Suppl. 2):ii55–7. Gutierrez M, Filippucci E, De Angelis R et al. A sonographic spectrum of psoriatic arthritis: ‘the five targets’. Clin Rheumatol 2010; 29:133–42. Spira D, Kotter I, Henes J et al. MRI findings in psoriatic arthritis of the hands. AJR Am J Roentgenol 2010; 195:1187–93. Gladman DD, Mease PJ, Strand V et al. Consensus on a core set of domains for psoriatic arthritis. J Rheumatol 2007; 34:1167–70. Clegg DO, Reda DJ, Mejias E et al. Comparison of sulfasalazine and placebo in the treatment of psoriatic arthritis. A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 1996; 39:2013–20. Felson DT, Anderson JJ, Boers M et al. The American College of Rheumatology preliminary core set of disease activity measures for rheumatoid arthritis clinical trials. The Committee on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Arthritis Rheum 1993; 36:729–40. Prevoo ML, van MA, Kuper HH et al. Modified disease activity scores that include twenty-eight-joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum 1995; 38:44–8. Gladman DD, Tom BD, Mease PJ et al. Informing response criteria for psoriatic arthritis (PsA) II: further considerations and a proposal – the PsA joint activity index. J Rheumatol 2010; 37:2559–65. Mumtaz A, Gallagher P, Kirby B et al. Development of a preliminary composite disease activity index in psoriatic arthritis. Ann Rheum Dis 2011; 70:272–7. Schoels M, Aletaha D, Funovits J et al. Application of the DAREA/ DAPSA score for assessment of disease activity in psoriatic arthritis. Ann Rheum Dis 2010; 69:1441–7. Helliwell PS, Fitzgerald O, Fransen J et al. The development of candidate composite disease activity and responder indices for psoriatic arthritis (GRACE project). Ann Rheum Dis 2013; 72:986–91. Bruce B, Fries JF. The Stanford Health Assessment Questionnaire: a review of its history, issues, progress, and documentation. J Rheumatol 2003; 30:167–78. Fries JF, Spitz P, Kraines RG et al. Measurement of patient outcome in arthritis. Arthritis Rheum 1980; 23:137–45. Husted JA, Gladman DD, Long JA et al. A modified version of the Health Assessment Questionnaire (HAQ) for psoriatic arthritis. Clin Exp Rheumatol 1995; 13:439–43. McKenna SP, Doward LC, Whalley D et al. Development of the PsAQoL: a quality of life instrument specific to psoriatic arthritis. Ann Rheum Dis 2004; 63:162–9.

© 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 783 70 Schmid-Ott G, Schallmayer S, Calliess IT. Quality of life in patients with psoriasis and psoriasis arthritis with a special focus on stigmatization experience. Clin Dermatol 2007; 25:547–54. 71 Husted JA, Gladman DD, Farewell VT et al. Health-related quality of life of patients with psoriatic arthritis: a comparison with patients with rheumatoid arthritis. Arthritis Rheum 2001; 45:151–8. 72 Salaffi F, Carotti M, Gasparini S et al. The health-related quality of life in rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis: a comparison with a selected sample of healthy people. Health Qual Life Outcomes 2009; 7:25. 73 Wong K, Gladman DD, Husted J et al. Mortality studies in psoriatic arthritis: results from a single outpatient clinic. I. Causes and risk of death. Arthritis Rheum 1997; 40:1868–72. 74 Gladman DD, Farewell VT, Wong K et al. Mortality studies in psoriatic arthritis: results from a single outpatient center. II. Prognostic indicators for death. Arthritis Rheum 1998; 41:1103–10. 75 Han C, Robinson DW Jr, Hackett MV et al. Cardiovascular disease and risk factors in patients with rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis. J Rheumatol 2006; 33:2167– 72. 76 Husted JA, Thavaneswaran A, Chandran V et al. Cardiovascular and other comorbidities in patients with psoriatic arthritis: a comparison with patients with psoriasis. Arthritis Care Res (Hoboken) 2011; 63:1729–35. 77 Li WQ, Han JL, Manson JE et al. Psoriasis and risk of nonfatal cardiovascular disease in U.S. women: a cohort study. Br J Dermatol 2012; 166:811–18. 78 Kimhi O, Caspi D, Bornstein NM et al. Prevalence and risk factors of atherosclerosis in patients with psoriatic arthritis. Semin Arthritis Rheum 2007; 36:203–9. 79 Gonzalez-Juanatey C, Llorca J, Amigo-Diaz E et al. High prevalence of subclinical atherosclerosis in psoriatic arthritis patients without clinically evident cardiovascular disease or classic atherosclerosis risk factors. Arthritis Rheum 2007; 57:1074–80. 80 Tam LS, Shang Q, Li EK et al. Subclinical carotid atherosclerosis in patients with psoriatic arthritis. Arthritis Rheum 2008; 59:1322–31. 81 Raychaudhuri SK, Chatterjee S, Nguyen C et al. Increased prevalence of the metabolic syndrome in patients with psoriatic arthritis. Metab Syndr Relat Disord 2010; 8:331–4. 82 Li W, Han J, Qureshi AA. Obesity and risk of incident psoriatic arthritis in US women. Ann Rheum Dis 2012; 71:1267–72. 83 Love TJ, Zhu Y, Zhang Y et al. Obesity and the risk of psoriatic arthritis: a population-based study. Ann Rheum Dis 2012; 71:1273–7. 84 Di Minno MN, Peluso R, Iervolino S et al. Obesity and the prediction of minimal disease activity: a prospective study in psoriatic arthritis. Arthritis Care Res (Hoboken) 2013; 65:141–7. 85 Di Minno MN, Iervolino S, Peluso R et al. Weight loss and induction of minimal disease activity in psoriatic arthritis patients starting TNF-a blockers treatment. Ann Rheum Dis 2012; 71:109. 86 Khraishi M, MacDonald D, Rampakakis E et al. Prevalence of patient-reported comorbidities in early and established psoriatic arthritis cohorts. Clin Rheumatol 2011; 30:877–85. 87 Eder L, Shanmugarajah S, Thavaneswaran A et al. The association between smoking and the development of psoriatic arthritis among psoriasis patients. Ann Rheum Dis 2012; 71:219–24. 88 Peters MJ, Welsh P, McInnes IB et al. Tumour necrosis factor a blockade reduces circulating N-terminal pro-brain natriuretic peptide levels in patients with active rheumatoid arthritis: results from a prospective cohort study. Ann Rheum Dis 2010; 69:1281–5. 89 Greenberg JD, Kremer JM, Curtis JR et al. Tumour necrosis factor antagonist use and associated risk reduction of cardiovascular events among patients with rheumatoid arthritis. Ann Rheum Dis 2011; 70:576–82.

© 2013 British Association of Dermatologists

90 Boehncke WH, Boehncke S. Cardiovascular mortality in psoriasis and psoriatic arthritis: epidemiology, pathomechanisms, therapeutic implications, and perspectives. Curr Rheumatol Rep 2012; 14:343–8. 91 American Academy of Dermatology Work Group, Menter A, Korman NJ et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 6 guidelines of care for the treatment of psoriasis and psoriatic arthritis: case-based presentations and evidence-based conclusions. J Am Acad Dermatol 2011; 65: 137–74. 92 Gossec L, Smolen JS, Gaujoux-Viala C et al. European League Against Rheumatism recommendations for the management of psoriatic arthritis with pharmacological therapies. Ann Rheum Dis 2012; 71:4–12. 93 Cronstein BN, Naime D, Ostad E. The antiinflammatory mechanism of methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte accumulation in an in vivo model of inflammation. J Clin Invest 1993; 92:2675–82. 94 Black RL, O’Brien WM, Vanscott EJ et al. Methotrexate therapy in psoriatic arthritis; double-blind study on 21 patients. JAMA 1964; 189:743–7. 95 Abu-Shakra M, Gladman DD, Thorne JC et al. Longterm methotrexate therapy in psoriatic arthritis: clinical and radiological outcome. J Rheumatol 1995; 22:241–5. 96 Spadaro A, Riccieri V, Sili-Scavalli A et al. Comparison of cyclosporin A and methotrexate in the treatment of psoriatic arthritis: a one-year prospective study. Clin Exp Rheumatol 1995; 13:589– 93. 97 Kingsley GH, Kowalczyk A, Taylor H et al. A randomized placebo-controlled trial of methotrexate in psoriatic arthritis. Rheumatology (Oxford) 2012; 51:1368–77. 98 Chandran V, Schentag CT, Gladman DD. Reappraisal of the effectiveness of methotrexate in psoriatic arthritis: results from a longitudinal observational cohort. J Rheumatol 2008; 35:469–71. 99 Maneiro JR, Salgado E, Gomez-Reino JJ. Immunogenicity of monoclonal antibodies against tumor necrosis factor used in chronic immune-mediated inflammatory conditions: systematic review and meta-analysis. JAMA Intern Med 2013; 173:1416–28. 100 Maini RN, Breedveld FC, Kalden JR et al. Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor alpha monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41:1552–63. 101 Humiraâ (adalimumab) [prescribing information]. North Chicago, IL: AbbVie Inc.; 2013. 102 Prodanovich S, Ma F, Taylor JR et al. Methotrexate reduces incidence of vascular diseases in veterans with psoriasis or rheumatoid arthritis. J Am Acad Dermatol 2005; 52:262–7. 103 Micha R, Imamura F, Wyler von Ballmoos M et al. Systematic review and meta-analysis of methotrexate use and risk of cardiovascular disease. Am J Cardiol 2011; 108:1362–70. 104 Kalb RE, Strober B, Weinstein G et al. Methotrexate and psoriasis: 2009 National Psoriasis Foundation Consensus Conference. J Am Acad Dermatol 2009; 60:824–37. 105 Methotrexate [prescribing information]. Lake Forest, IL: Hospira; 2008. 106 Chang CA, Gottlieb AB, Lizzul PF. Management of psoriatic arthritis from the view of the dermatologist. Nat Rev Rheumatol 2011; 7:588–98. 107 Menter A, Gottlieb A, Feldman SR et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol 2008; 58:826–50.

British Journal of Dermatology (2014) 170, pp772–786

784 Management of PsA, W.H. Boehncke et al. 108 Whiting-O’Keefe QE, Fye KH, Sack KD. Methotrexate and histologic hepatic abnormalities: a meta-analysis. Am J Med 1991; 90:711–16. 109 Lindsay K, Gough A. Psoriatic arthritis, methotrexate and the liver – are rheumatologists putting their patients at risk? Rheumatology (Oxford) 2008; 47:939–41. 110 Saag KG, Teng GG, Patkar NM et al. American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis. Arthritis Rheum 2008; 59:762–84. 111 Farr M, Kitas GD, Waterhouse L et al. Sulphasalazine in psoriatic arthritis: a double-blind placebo-controlled study. Br J Rheumatol 1990; 29:46–9. 112 Fraser SM, Hopkins R, Hunter JA et al. Sulphasalazine in the management of psoriatic arthritis. Br J Rheumatol 1993; 32:923–5. 113 Dougados M, vam der Linden S, Leirisalo-Repo M et al. Sulfasalazine in the treatment of spondylarthropathy. A randomized, multicenter, double-blind, placebo-controlled study. Arthritis Rheum 1995; 38:618–27. 114 Clegg DO, Reda DJ, Weisman MH et al. Comparison of sulfasalazine and placebo in the treatment of reactive arthritis (Reiter’s syndrome). A Department of Veterans Affairs Cooperative Study. Arthritis Rheum 1996; 39:2021–7. 115 Rahman P, Gladman DD, Cook RJ et al. The use of sulfasalazine in psoriatic arthritis: a clinic experience. J Rheumatol 1998; 25:1957–61. 116 Azulfidineâ (sulfasalazine) [prescribing information]. New York, NY: Pharmacia & Upjohn Co, Division of Pfizer Inc; 2011. 117 Kaltwasser JP, Nash P, Gladman D et al. Efficacy and safety of leflunomide in the treatment of psoriatic arthritis and psoriasis: a multinational, double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum 2004; 50:1939–50. 118 Nash P, Thaci D, Behrens F et al. Leflunomide improves psoriasis in patients with psoriatic arthritis: an in-depth analysis of data from the TOPAS study. Dermatology 2006; 212:238–49. 119 Aravaâ (leflunomide) [prescribing information]. Bridgewater, NJ: Sanofi-Aventis U.S. LLC; 2012. 120 Ash Z, Gaujoux-Viala C, Gossec L et al. A systematic literature review of drug therapies for the treatment of psoriatic arthritis: current evidence and meta-analysis informing the EULAR recommendations for the management of psoriatic arthritis. Ann Rheum Dis 2012; 71:319–26. 121 Neoralâ (cyclosporine) [prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals Corporation; 2009. 122 Enbrelâ (etanercept) [prescribing information]. Thousand Oaks, CA: Immunex Corporation; 2013. 123 Simponiâ (golimumab) [prescribing information]. Horsham, PA: Janssen Biotech, Inc.; 2013. 124 Remicadeâ (infliximab) [prescribing information]. Horsham, PA: Janssen Biotech, Inc.; 2013. 125 Mease PJ, Gladman DD, Ritchlin CT et al. Adalimumab for the treatment of patients with moderately to severely active psoriatic arthritis: results of a double-blind, randomized, placebo-controlled trial. Arthritis Rheum 2005; 52:3279–89. 126 Gladman DD, Mease PJ, Ritchlin CT et al. Adalimumab for longterm treatment of psoriatic arthritis: forty-eight week data from the Adalimumab Effectiveness in Psoriatic Arthritis Trial. Arthritis Rheum 2007; 56:476–88. 127 Mease PJ, Ory P, Sharp JT et al. Adalimumab for long-term treatment of psoriatic arthritis: 2-year data from the Adalimumab Effectiveness in Psoriatic Arthritis Trial (ADEPT). Ann Rheum Dis 2009; 68:702–9. 128 Gladman DD, Mease PJ, Cifaldi MA et al. Adalimumab improves joint-related and skin-related functional impairment in patients

British Journal of Dermatology (2014) 170, pp772–786

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

with psoriatic arthritis: patient-reported outcomes of the Adalimumab Effectiveness in Psoriatic Arthritis Trial. Ann Rheum Dis 2007; 66:163–8. Mease PJ, Goffe BS, Metz J et al. Etanercept in the treatment of psoriatic arthritis and psoriasis: a randomised trial. Lancet 2000; 356:385–90. Mease PJ, Kivitz AJ, Burch FX et al. Etanercept treatment of psoriatic arthritis: safety, efficacy, and effect on disease progression. Arthritis Rheum 2004; 50:2264–72. Mease PJ, Kivitz AJ, Burch FX et al. Continued inhibition of radiographic progression in patients with psoriatic arthritis following 2 years of treatment with etanercept. J Rheumatol 2006; 33:712–21. Mease PJ, Woolley JM, Singh A et al. Patient-reported outcomes in a randomized trial of etanercept in psoriatic arthritis. J Rheumatol 2010; 37:1221–7. Prinz JC, Fitzgerald O, Boggs RI et al. Combination of skin, joint and quality of life outcomes with etanercept in psoriasis and psoriatic arthritis in the PRESTA trial. J Eur Acad Dermatol Venereol 2011; 25:559–64. Sterry W, Ortonne JP, Kirkham B et al. Comparison of two etanercept regimens for treatment of psoriasis and psoriatic arthritis: PRESTA randomised double blind multicentre trial. BMJ 2010; 340:c147. Lubrano E, Spadaro A, Marchesoni A et al. The effectiveness of a biologic agent on axial manifestations of psoriatic arthritis. A twelve months observational study in a group of patients treated with etanercept. Clin Exp Rheumatol 2011; 29:80–4. Kavanaugh A, McInnes I, Mease P et al. Golimumab, a new human tumor necrosis factor a antibody, administered every four weeks as a subcutaneous injection in psoriatic arthritis: twentyfour-week efficacy and safety results of a randomized, placebocontrolled study. Arthritis Rheum 2009; 60:976–86. Kavanaugh A, van der Heijde D, McInnes IB et al. Golimumab in psoriatic arthritis: one-year clinical efficacy, radiographic, and safety results from a phase III, randomized, placebo-controlled trial. Arthritis Rheum 2012; 64:2504–17. Antoni C, Krueger GG, de Vlam K et al. Infliximab improves signs and symptoms of psoriatic arthritis: results of the IMPACT 2 trial. Ann Rheum Dis 2005; 64:1150–7. Antoni CE, Kavanaugh A, Kirkham B et al. Sustained benefits of infliximab therapy for dermatologic and articular manifestations of psoriatic arthritis: results from the infliximab multinational psoriatic arthritis controlled trial (IMPACT). Arthritis Rheum 2005; 52:1227–36. Kavanaugh A, Antoni CE, Gladman D et al. The Infliximab Multinational Psoriatic Arthritis Controlled Trial (IMPACT): results of radiographic analyses after 1 year. Ann Rheum Dis 2006; 65:1038– 43. Antoni CE, Kavanaugh A, van der Heijde D et al. Two-year efficacy and safety of infliximab treatment in patients with active psoriatic arthritis: findings of the Infliximab Multinational Psoriatic Arthritis Controlled Trial (IMPACT). J Rheumatol 2008; 35:869–76. van der Heijde D, Kavanaugh A, Gladman DD et al. Infliximab inhibits progression of radiographic damage in patients with active psoriatic arthritis through one year of treatment: results from the induction and maintenance psoriatic arthritis clinical trial 2. Arthritis Rheum 2007; 56:2698–707. Fleischmann R, Vencovsky J, van Vollenhoven RF et al. Efficacy and safety of certolizumab pegol monotherapy every 4 weeks in patients with rheumatoid arthritis failing previous disease-modifying antirheumatic therapy: the FAST4WARD study. Ann Rheum Dis 2009; 68:805–11.

© 2013 British Association of Dermatologists

Management of PsA, W.H. Boehncke et al. 785 144 Reich K, Ortonne JP, Gottlieb AB et al. Successful treatment of moderate to severe plaque psoriasis with the PEGylated Fab0 certolizumab pegol: results of a phase II randomized, placebocontrolled trial with a re-treatment extension. Br J Dermatol 2012; 167:180–90. 145 Mease PJ, Fleischmann R, Deodhar AA et al. Effect of certolizumab pegol on signs and symptoms in patients with psoriatic arthritis: 24-week results of a phase 3 double-blind randomised placebocontrolled study (RAPID-PsA). Ann Rheum Dis 2014; 73:48–55. 146 Atteno M, Peluso R, Costa L et al. Comparison of effectiveness and safety of infliximab, etanercept, and adalimumab in psoriatic arthritis patients who experienced an inadequate response to previous disease-modifying antirheumatic drugs. Clin Rheumatol 2010; 29:399–403. 147 Fenix-Caballero S, Alegre-Del RE, Castano-Lara R et al. Direct and indirect comparison of the efficacy and safety of adalimumab, etanercept, infliximab and golimumab in psoriatic arthritis. J Clin Pharm Ther 2013; 38:286–93. 148 Migliore A, Bizzi E, Broccoli S et al. Indirect comparison of etanercept, infliximab, and adalimumab for psoriatic arthritis: mixed treatment comparison using placebo as common comparator. Clin Rheumatol 2012; 31:133–7. 149 Rodgers M, Epstein D, Bojke L et al. Etanercept, infliximab and adalimumab for the treatment of psoriatic arthritis: a systematic review and economic evaluation. Health Technol Assess 2011; 15: i–xxi, 1–329. 150 Saougou I, Markatseli TE, Papagoras C et al. Sustained clinical response in psoriatic arthritis patients treated with anti-TNF agents: a 5-year open-label observational cohort study. Semin Arthritis Rheum 2011; 40:398–406. 151 Thorlund K, Druyts E, Avina-Zubieta JA et al. Anti-tumor necrosis factor (TNF) drugs for the treatment of psoriatic arthritis: an indirect comparison meta-analysis. Biologics 2012; 6:417–27. 152 Kirson NY, Rao S, Birnbaum HG et al. Matching-adjusted indirect comparison of adalimumab vs etanercept and infliximab for the treatment of psoriatic arthritis. J Med Econ 2013; 16:479–89. 153 Gottlieb A, Menter A, Mendelsohn A et al. Ustekinumab, a human interleukin 12/23 monoclonal antibody, for psoriatic arthritis: randomised, double-blind, placebo-controlled, crossover trial. Lancet 2009; 373:633–40. 154 McInnes IB, Kavanaugh A, Gottlieb AB et al. Efficacy and safety of ustekinumab in patients with active psoriatic arthritis: 1 year results of the phase 3, multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet 2013; 382:780–9. 155 Stelaraâ (ustekinumab) [prescribing information]. Horsham, PA: Janssen Biotech, Inc.; 2013. 156 Mease P, Genovese MC, Gladstein G et al. Abatacept in the treatment of patients with psoriatic arthritis: results of a six-month, multicenter, randomized, double-blind, placebo-controlled, phase II trial. Arthritis Rheum 2011; 63:939–48. 157 Ogata A, Tanaka T. Tocilizumab for the treatment of rheumatoid arthritis and other systemic autoimmune diseases: current perspectives and future directions. Int J Rheumatol 2012; 2012:946048. 158 Mease P, Strand V, Shalamberidze L et al. A phase II, doubleblind, randomised, placebo-controlled study of BMS945429 (ALD518) in patients with rheumatoid arthritis with an inadequate response to methotrexate. Ann Rheum Dis 2012; 71:1183–9. 159 Mease PJ. Psoriatic arthritis – treatment update. Bull NYU Hosp Jt Dis 2011; 69:243–9. 160 Hueber W, Patel DD, Dryja T et al. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med 2010; 2:52ra72.

© 2013 British Association of Dermatologists

161 Genovese MC, Durez P, Richards HB et al. Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study. Ann Rheum Dis 2013; 72:863–9. 162 McInnes IB, Sieper J, Braun J et al. Efficacy and safety of secukinumab, a fully human anti-interleukin-17A monoclonal antibody, in patients with moderate-to-severe psoriatic arthritis: a 24-week, randomised, double-blind, placebo-controlled, phase II proof-of-concept trial. Ann Rheum Dis 2014; 73:349–56. 163 Papp K, Cather JC, Rosoph L et al. Efficacy of apremilast in the treatment of moderate to severe psoriasis: a randomised controlled trial. Lancet 2012; 380:738–46. 164 Schett G, Wollenhaupt J, Papp K et al. Oral apremilast in the treatment of active psoriatic arthritis: results of a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum 2012; 64:3156–67. 165 Xeljanzâ (tofacitinib) [prescribing information]. New York, NY: Pfizer Labs; 2012. 166 Angel K, Provan SA, Fagerhol MK et al. Effect of 1-year anti-TNFa therapy on aortic stiffness, carotid atherosclerosis, and calprotectin in inflammatory arthropathies: a controlled study. Am J Hypertens 2012; 25:644–50. 167 Cimziaâ (certolizumab pegol) [prescribing information]. Smyrna, GA: UCB, Inc.; 2013. 168 Orenciaâ (abatacept) [prescribing information]. Princeton, NJ: Bristol-Myers Squibb; 2011. 169 RoActemraâ (tocilizumab) [prescribing information]. GrenzachWyhlen, Germany: Roche; 2011. 170 Kremer JM, Bloom BJ, Breedveld FC et al. The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: results of a double-blind, placebo-controlled phase IIa trial of three dosage levels of CP-690,550 versus placebo. Arthritis Rheum 2009; 60:1895–905. 171 Cantini F, Niccoli L, Nannini C et al. Psoriatic arthritis: a systematic review. Int J Rheum Dis 2010; 13:300–17.

Appendix Conflicts of interest W.H.B. has current consulting/advisory board agreements with Biogen Idec, Janssen, Pfizer and MSD. A.Q. has current consulting agreements with AbbVie, Janssen, Novartis and the U.S. Centers for Disease Control; has licensed his questionnaire to Pfizer and Merck for clinical trials; and has received a research/educational grant from Amgen. J.F.M. is an investigator for Amgen and has a consulting agreement with Biogen Idec. D.T. has current consulting/advisory board agreements with Astellas, Biogen, Janssen, Celgene Corp., AbbVie, Pfizer, MSD and Lilly. D.T. receives research/educational grants (paid to the Department of Dermatology, Goethe University) from Pfizer. G.G.K., currently and in the past 2 years, has served on the steering committees for Centocor/Janssen for psoriasis and psoriatic arthritis and has given educational talks on these subjects. He also serves on the steering committees for PSOLAR/ Janssen and is compensated for these activities. G.G.K. chairs the data monitoring safety board for Pfizer for a new psoriasis indication and is compensated for these activities. He has given educational talks on psoriasis and psoriatic arthritis and has received fees and/or honoraria for these as well as for

British Journal of Dermatology (2014) 170, pp772–786

786 Management of PsA, W.H. Boehncke et al.

services as a consultant or as an advisory board member for AbbVie, Amgen, Boehringer Ingelheim, Lilly, L’Oreal, Medicis, Novo Nordisk and Vascular Biogenics Limited. J.W. has current consulting agreements with Celgene Corp. and AbbVie. N.K. has no relevant conflict of interests. A.B.G. has current consulting/advisory board agreements with Amgen Inc., Astellas, Janssen-Ortho Inc., Celgene Corp., Bristol-Myers Squibb Co., Beiersdorf Inc., Abbott Labs (AbbVie), TEVA, Actelion, UCB, Novo Nordisk, Novartis, DermiPsor Ltd, Incyte, Pfizer, Canfite, Lilly, Coronado, Vertex, Karyopharm, CSL Behring Biotherapies for Life, Merck & Co. Inc. and GlaxoSmithKline. She has received research/educational grants (paid to Tufts Medical Center) from Janssen-Ortho Inc., Amgen, Abbott Labs (AbbVie), Novartis, Celgene, Pfizer, Lilly, Coronado, Immune Control, Lerner Medical Devices Inc., Novo Nordisk and UCB.

British Journal of Dermatology (2014) 170, pp772–786

Supporting Information Additional Supporting Information may be found in the online version of this article at the publisher’s website: Table S1. CASPAR (ClASsification criteria for Psoriatic Arthritis) – specificity 987%, sensitivity 914%.11 Reproduced with permission from Cantini et al.171 Fig S1. The PASE (Psoriatic Arthritis Screening and Evaluation) questionnaire. Used with permission from Husni et al.41 Fig S2. The ToPAS (Toronto Psoriatic Arthritis Screening) questionnaire. Used with permission from Gladman et al.42 Fig S3. The PEST (Psoriatic Epidemiology Screening Tool) questionnaire. One point is scored for each question answered in the affirmative. A total score ≥ 3 is indicative of psoriatic arthritis. Used with permission from Ibrahim et al.43

© 2013 British Association of Dermatologists

Diagnosing and treating psoriatic arthritis: an update.

Psoriatic arthritis (PsA) is an inflammatory arthritis of uncertain pathogenesis, affecting approximately one in four patients with psoriasis. Onset o...
247KB Sizes 0 Downloads 0 Views