Abstract
Objective
Psoriatic arthritis (PsA), a chronic inflammatory disease characterized by heterogeneous clinical manifestations, substantially impacts the quality of life of affected individuals. This article aims at developing consensus recommendations for the management of PsA and associated comorbidities and screening and monitoring requirements of PsA therapies in the United Arab Emirates (UAE) population.
Methods
An extensive review of present international and regional guidelines and publications on the pharmacological management, monitoring of therapies in the context of PsA was performed. Key findings from guidelines and literature were reviewed by a panel of experts from the UAE at several meetings to align with current clinical practices. Consensus statements were formulated based on collective agreement of the experts and members of Emirates Society for Rheumatology.
Results
The consensus recommendations were developed to aid practitioners in clinical decision‐making with respect to dosage recommendations for pharmacological therapies for PsA, including conventional drugs, non‐biologic, and biologic therapies. Consensus recommendations for therapeutic options for the treatment of PsA domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, psoriasis, and nail disease, were developed. The panel emphasized the importance of monitoring PsA therapies and arrived at a consensus on monitoring requirements for PsA therapies. The expert panel proposed recommendations for the management of common comorbidities associated with PsA.
Conclusion
These consensus recommendations can guide physicians and healthcare professionals in the UAE in making proper treatment decisions, as well as efficiently managing comorbidities and monitoring therapies in patients with PsA.
Keywords: biologics, comorbidities, disease‐modifying drugs, domains, dosage, monitoring, pharmacological management, psoriatic arthritis, recommendations
1. INTRODUCTION
Psoriatic arthritis (PsA) is a heterogeneous chronic inflammatory disease occurring in about 30% of patients with psoriasis. 1 The global prevalence of PsA varies by geographic region and ranges from 0.001% to 0.42%, while the prevalence of PsA is 0.01% to 0.3% in Middle East countries. 2 , 3 , 4 , 5 , 6 Approximately 20% of patients diagnosed with PsA may develop a more aggressive form of arthritis resulting in joint damage. 4 In patients with PsA, quality of life (QoL) is substantially impacted consequent to stress, depression, mood changes, pain, and compromised physical functioning. 7 , 8
Nearly all the current treatment recommendations for PsA are reflective of the treatment and disease landscape in developed countries, particularly Europe and the United States. 9 , 10 , 11 Currently, not much is known about the epidemiology and treatment practices specific to PsA in the Middle East. There are several local challenges that may not be adequately accounted for, in currently available treatment recommendations for PsA. 12
Multiple factors necessitate national recommendations for the management of PsA specific to the United Arab Emirates (UAE), including wide variability in healthcare systems, patient access to advanced care, affordability of treatment, practicing rheumatologists trained in different countries and implementing different approaches to treatment, and ethnic diversities among patients from almost 200 countries in the UAE. Many of the newer approved therapies such as biological disease‐modifying antirheumatic drugs (DMARDs) may not be accessible to patients who do not have insurance coverage, given their prohibitive cost. Other factors that preclude the implementation of global treatment recommendations in local clinical practice are lack of disease awareness among both patients and healthcare providers, shortage of healthcare resources, and lack of multidisciplinary healthcare clinics. 1
The objectives of this article are to develop consensus statements for the pharmacological management of PsA and associated comorbidities and screening and monitoring requirements of PsA therapies, to assist practicing physicians in the UAE.
2. METHODS
For the development of the consensus guidelines, 6 experts with international board certifications, and more than 15 years experience in rheumatology and an interest in psoriatic arthritis representing different healthcare sectors (government and private) of the UAE were chosen from the Emirates Society for Rheumatology and convened in several meetings.
A targeted literature review was conducted. Current international and local treatment guidelines for PsA were identified through an extensive literature search and reviewed by members of the panel to identify unmet needs in local treatment practices in the UAE. Regional guidelines were compared with the latest international guidelines from the American College of Rheumatology/National Psoriasis Foundation (ACR/NPF) Guideline for the Treatment of Psoriatic Arthritis 2018, European League Against Rheumatism (EULAR) 2019, The Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA) 2020 Update, GRAPPA 2015 (detailed), and 2014 Saudi Practical Guidelines on Biologic Treatment of Psoriasis. 9 , 10 , 11 , 13 , 14
As of August 2021, there is a dearth for guidelines on management of PsA specific to the Arab region.
Based on a review of international and regional guidelines, consensus statements were developed focusing on pharmacological treatment options, screening and monitoring requirements for PsA therapies, and management of comorbidities associated with PsA. The results were discussed with all the members of Emirates Society of Rheumatology meeting to arrive at the final consensus statements. Key findings from the review were presented as statements.
The key objectives of the meetings were:
to critically review the available regional and international recommendations on the management of patients with PsA
to develop regional recommendations for effective management of PsA
to develop regional recommendations for screening and monitoring requirements for PsA therapies.
Several meetings were held to generate consensus statements regarding pharmacological management of psoriatic arthritis. The first expert panel meeting was conducted on September 23, 2020 and the meeting lasted for 2 hours. The second expert panel meeting was conducted on October 7, 2020 and the meeting lasted for 3 hours. The third expert panel meeting was held on December 16, 2020 and lasted for 2 hours. The fourth and fifth meetings were held in the presence of Emirates Society for Rheumatology members on December 18, 2020 and May 22, 2021 respectively; each meeting lasted for almost 2 hours. The final meeting was held on August 10, 2021 and lasted for 2 hours when the consensus statements were approved.
The consensus statements have been written in 2 separate papers. The first paper focuses on overarching principles, evaluation of PsA and non‐pharmacological treatment options of PsA. 15 The present article, which is the second part covers consensus statements related to pharmacological management of PsA (dosing and administration recommendations, treatment recommendations for PsA domains and consensus statements on efficacy and safety profile of non‐biologic and biologic therapies), screening and monitoring requirements for therapies and management of comorbidities.
3. RESULTS AND DISCUSSION
3.1. Pharmacological treatment options
GRAPPA recommendations (2015) are centered around major subdomains within PsA, including peripheral arthritis, axial disease, enthesitis, dactylitis, psoriasis, and nail disease. In accordance with the overarching principles outlined in the GRAPPA recommendations, the expert panel agreed that treatment selection should be based on shared decision‐making between the physician and patient. The current treatment recommendations were developed to align with the core recommendations from GRAPPA. 9
Pharmacological therapies for the management of PsA include:
Symptomatic treatments: non‐steroidal anti‐inflammatory drugs (NSAIDs) and glucocorticoids (GCCs) 9
- Biologic DMARDs:
-
otumor necrosis factor inhibitors (TNFi): adalimumab, etanercept, infliximab, certolizumab pegol, golimumab 9
-
ointerleukin‐12/23 inhibitors (IL‐12/23i): ustekinumab 9
-
oIL‐23 inhibitors (IL‐23i): guselkumab 17
-
oIL‐17 inhibitors (IL‐17i): secukinumab, ixekizumab 10
-
ocytotoxic T‐lymphocyte‐associated protein 4‐immunoglobulin (CTLA4‐Ig): abatacept 10
- o
-
o
3.2. Dosage and administration
The dosage recommendations for the pharmacological agents are shown in Table 1.
TABLE 1.
Therapeutic class | Dosage | Route of administration |
---|---|---|
NSAIDs a | Oral and IM | |
GCCs b | Lowest effective dose | IM and IA |
csDMARDs | ||
Methotrexate 23 | 7.5–30 mg/wk | Oral, SC |
Sulfasalazine 26 , 27 | 2 to 3 g/d | Oral |
Leflunomide 28 | Daily loading dose of 100 mg for 3 d followed by 20 mg/d | Oral |
tsDMARDs | ||
Apremilast 30 , 31 , 32 , 33 | Apremilast 30mg twice daily (maintenance dose) | Oral |
Tofacitinib 34 | 5 mg twice daily (immediate release) or 11 mg once daily (extended release) in combination with non‐biologic DMARDs | Oral |
Upadacitinib 16 | 15 mg once daily | Oral |
TNFi b | ||
Adalimumab 107 | 40 mg every 2 wks in patients with PsA with inadequate response to DMARDs | SC |
Etanercept 108 , 109 , 110 | 50 mg once weekly | SC |
Infliximab 111 , 112 | 5 mg/kg at 0, 2, 6 wks, and every 8 wks thereafter. | IV |
Certolizumab pegol 113 , 114 |
400 mg at wks 0, 2, 4 Maintenance dose: 200 mg every 2 wks, or 400 mg every 4 wks once clinical response is confirmed |
SC |
Golimumab 115 |
SC: 50 mg monthly IV: 2 mg/kg over 30 min at Wks 0 and 4 (loading dose), thereafter every 8 wks (maintenance) |
SC, IV |
IL‐12/23i c | ||
Ustekinumab 37 | 45 mg or 90 mg (based on weight) at 0 and 4 wks, and every 12 wks thereafter | SC |
IL‐17i | ||
Secukinumab 116 |
300 mg in patients with concomitant moderate‐to‐ severe plaque psoriasis or who are TNFi inadequate responders at wks 0, 1, 2, 3 and 4 and every 4 wks thereafter. 150 mg in other patients at wks 0, 1, 3, 4 and every 4 wks thereafter and based on clinical response the dose can be increased to 300 mg. |
SC |
Ixekizumab 38 , 117 |
160 mg followed by 80 mg every 2 or 4 wks in patients who previously had inadequate response to TNFi. For patients with arthritis and moderate‐to‐severe plaque psoriasis, using the dosing regimen for plaque psoriasis;160 mg SC at wk 0, then 80 mg SC at wks 2, 4, 6, 8, 10 and 12, then 80 mg SC every 4 wks, starting wk 16. |
SC |
IL‐23i | ||
Guselkumab 40 | 100 mg at wks 0 and 4, and every 8 wks thereafter (maintenance) | SC |
(CTLA4‐Ig) T‐cell co‐stimulation inhibitor | ||
Abatacept 43 , 118 | 500 mg, 750 mg, or 1000 mg (based on weight range); following initial IV infusion, subsequent infusions should be administered at 2 and 4 wks and every 4 wks thereafter) 125 mg of abatacept injection should be administered SC once weekly | IV, SC |
Abbreviations: csDMARD, conventional synthetic disease‐modifying antirheumatic drug; CTLA4‐Ig, cytotoxic T‐lymphocyte‐associated antigen4‐immunoglobulin; GCCs, glucocorticoids; IA, intra‐articular; IL‐17i, interleukin‐17 inhibitor; IL‐12/23i, interleukin‐12/23 inhibitor; IL‐23i, interleukin‐23 inhibitor; IM, intramuscular; IV, intravenous; JAK, Janus kinase; NSAIDs, non‐steroidal anti‐inflammatory drugs; PsA, psoriatic arthritis; PDE‐4i, phosphodiesterase‐4 inhibitor (apremilast); SC, subcutaneous; TNFi, tumor necrosis factor inhibitor; tsDMARD, targeted systemic disease‐modifying antirheumatic drug.
Mentioning the doses of all available NSAIDs is beyond the scope of this paper.
European League Against Rheumatism 2019 and Group for Research and Assessment of Psoriasis and Psoriatic Arthritis 2015 guidelines recommend administering the lowest effective doses of GCC.
Loading dosage might vary according to the severity of psoriasis and other domains of the disease.
3.2.1. NSAIDs
The expert panel urged that a thorough safety evaluation should precede the use of NSAIDs in patients with comorbid medical conditions (eg peptic ulcer disease, chronic kidney disease [CKD], cardiovascular disease [CVD]).
3.2.2. GCCs
In the treatment of peripheral arthritis, GCC is recommended conditionally, and the lowest effective doses should be administered to reduce the risk of side effects. 9
3.2.3. csDMARDs
Conventional DMARDs are indicated for the treatment of moderate‐to‐severe PsA and in patients who have failed to respond to short‐term NSAID therapy. Methotrexate has been shown to improve disease activity and health‐related QoL in patients with PsA. Methotrexate has a broad therapeutic dose range (7.5–30 mg/wk) and different administration forms (oral, or subcutaneous). 23 Evidence suggests that monotherapy with methotrexate offers moderate improvement in joint and skin disease in patients with PsA, and doses >15 mg/wk are associated with greater clinical efficacy compared to lower doses. 24 , 25
In patients with mild‐to‐moderate peripheral arthritis, use of sulfasalazine at a dose of 2–3 g/d may improve functional outcomes. 26 , 27
Leflunomide monotherapy with a daily loading dose of 100 mg/d for 3 days, followed by 20 mg/d is effective in the management of patients with mild‐to‐moderate PsA. 28 In an ongoing randomized, placebo‐controlled, double‐blind trial, the effectiveness of combination therapy of methotrexate and leflunomide in the treatment of patients with PsA is being evaluated, and the outcomes of the study are expected to provide key information for treatment strategies in PsA. 29
3.2.4. tsDMARDs
Phosphodiesterase‐4 inhibitors
Apremilast at a dose of 30 mg twice daily improves signs and symptoms and physical function in patients with active PsA. 30 , 31 , 32 , 33
JAK inhibitors
The recommended dosage of upadacitinib, a selective JAK inhibitor, is 15 mg once daily orally in patients with active PsA, who have not adequately responded or are intolerant to 1 or more DMARDs. 16
According to EULAR 2019 recommendations, tofacitinib should be administered after inadequate response or intolerance to at least 1 bDMARD, or in case bDMARDs are not considered appropriate (due to patient preference for oral therapy or adherence issues to injectable formulations). 10 The recommended dose of tofacitinib is 5 mg twice daily (immediate release) or 11 mg once daily (extended release) in combination with non‐biologic DMARDs. 34
3.2.5. Biologic DMARDs
TNF inhibitors
TNFi agents approved by the US Food and Drug Administration (FDA) and other health authorities worldwide for PsA treatment include etanercept, infliximab, adalimumab, golimumab, and certolizumab pegol. They are recommended for use in PsA after inadequate response to at least 1 synthetic DMARD, although they may also be used as initial therapy. 35 TNFi agents are recommended in peripheral arthritis, and are also the first choice of therapy in enthesitis, dactylitis, and nail psoriasis. 36
The dosage recommendations for TNFi (adalimumab, etanercept, infliximab, certolizumab pegol, and golimumab) are shown in Table 1.
IL‐12/23 inhibitors
The IL‐12/23 inhibitor ustekinumab has been recommended alongside other biologics such as TNFi and IL‐17 inhibitors after DMARD therapy in patients with active PsA. 9 , 10 The recommended dose of ustekinumab is 45 mg or 90 mg (subcutaneous) at 0 and 4 weeks and every 12 weeks thereafter. 37
IL‐17 inhibitors
The recommended dose of IL‐17i ixekizumab in patients with active PsA and inadequate response to TNFi agents is starting dose of 160 mg followed by 80 mg every 2 or 4 weeks for the safe and effective management of PsA. 38 IL‐17 inhibitor secukinumab is administered subcutaneously at doses of 300 mg in patients with concomitant moderate‐to‐severe plaque psoriasis or who are TNFi inadequate responders at weeks 0, 1, 2, 3 and 4 and every 4 weeks thereafter. The loading dose can be 150 mg in other patients at weeks 0, 1, 3, 4 and every 4 weeks thereafter and based on clinical response the dose can be increased to 300 mg. 39
IL‐23 inhibitors
Guselkumab, a specific IL‐23 inhibitor, has been recently approved for the treatment of PsA by the US FDA and European Medicines Agency (EMA). 17 The recommended dose of guselkumab is 100 mg at weeks 0 and 4, and every 8 weeks thereafter. 40 Risankizumab was not approved for PsA by the FDA and EMA when our consensus statements were drafted. However, the EMA and FDA approved it on November 22, 2021 and January 21, 2022 respectively for treatment of active psoriatic arthritis. 41 , 42
CTLA4‐Ig
Abatacept is a biologic agent that targets T‐cell costimulatory signals selectively and is approved for the treatment of PsA patients with inadequate response to csDMARDs, excluding those with uncontrolled skin lesions and axial disease. 43
3.2.6. Biosimilars
In recent years, biosimilars of infliximab, etanercept, and adalimumab have been approved by regulatory bodies in Europe and the USA for the treatment of PsA. These agents have been approved for the treatment of PsA based on similar efficacy to the reference product in psoriasis and/or rheumatoid arthritis (RA), by the extrapolation principle. 44 , 45 , 46 Biosimilars are more cost‐effective compared to biologics, and thereby represent a solution for better patient accessibility to therapy and reduction in associated healthcare costs. 47
Consensus statements on dosage recommendations for non‐biologic and biologic therapies are provided in Table 1.
3.3. Treatment recommendations based on PsA domains
3.3.1. Peripheral arthritis
In DMARD‐naïve patients with peripheral arthritis, csDMARDs (methotrexate, sulfasalazine, and leflunomide), PDE‐4i, IL‐12/23i, IL‐17i, IL‐23i, JAKi, and TNFi are recommended therapeutic options. 9 , 11 , 33 In patients with monoarthritis or oligoarthritis accompanied by factors such as dactylitis or joint damage, the use of csDMARDs and intra‐articular (IA) GCC should be considered. 9 In patients with polyarticular disease, csDMARDs should be considered either as first‐line treatment or after a short course of NSAIDs. In patients with inadequate response to csDMARDs, TNFi, IL‐12/23i, IL23i, IL17i, PDE‐4i and JAKi are recommended therapeutic options. 9 , 10 , 34 , 48 , 49 In patients with inadequate response to 1 biologic treatment, switching to another biologic within the same drug class, or to a drug with a different mode of action, should be considered. 9 , 10 In all patients with peripheral arthritis, IA and systemic GCC are conditionally recommended at the lowest dosages and for a short duration.
3.3.2. Axial disease
At the time of drafting consensus statements, there were no studies on management of psoriatic spondylitis. Therefore, management of this condition depends on current treatment modalities in ankylosing spondylitis. In patients with psoriatic spondylitis/axial disease who are biologic‐naïve, NSAIDs, physiotherapy, simple analgesia, TNFi or IL‐17i, JAKi are recommended therapeutic options. Tofacitinib was not approved for PsA by the FDA and EMA when the consensus statements were drafted. However, FDA and EMA approved it on November 18, 2021 and December 14, 2021 respectively for treatment of ankylosing spondylitis. Other therapeutic options such as sacroiliac joint GCC injections and bisphosphonates can be used, but with caution. 9 , 14 , 50 , 51 , 52 , 53 , 54 , 55 , 56
3.3.3. Enthesitis
For the management of PsA patients with enthesitis (inflammation at the sites of attachment of ligaments, tendons, and joint capsules to bone), 57 treatment with TNFi, IL‐17i, IL‐23i, IL‐12/23i and JAKi are recommended therapeutic options. Other therapeutic options include NSAIDs, physiotherapy, methotrexate, CTLA4‐Ig, and PDE‐4i. 9 , 10 , 49 , 58 , 59 , 60 , 61 , 62
3.3.4. Dactylitis
The recommended therapies for the management of PsA patients with dactylitis include TNFi therapies (infliximab, adalimumab, golimumab, and certolizumab pegol), IL‐17i, IL‐12/23i, IL‐23i, JAKi, and PDE‐4i. 9 , 58 , 62 , 63 Other therapeutic options include NSAIDs, GCC injections, and methotrexate. 9
3.3.5. Nail disease
For the management of PsA patients with moderate‐to‐severe nail disease, TNFi, IL‐17i, PDE‐4i, IL‐23i, IL‐12/23i and acitretin are recommended therapeutic options. 9 , 10 , 64 , 65 , 66 , 67 Other options include topical therapies, procedural therapies, csDMARDs (cyclosporine, leflunomide, and methotrexate). 9 , 68 , 69
3.3.6. Skin disease
The expert panel recommends the use of topical therapies, phototherapy, acitretin and csDMARDs (methotrexate, leflunomide, cyclosporine) as first‐line therapeutic options, especially for PsA patients with milder skin disease. 9 TNFi, IL‐17i, IL12/23i, IL‐23i, JAKi and PDE‐4i are recommended therapeutic options for treatment of PsA patients with significant skin involvement. 9 Furthermore, biologic agents such as IL‐17i are preferred to TNFi, with or without topical treatments and DMARDs, in PsA patients with active psoriasis (≥3% of body surface area of skin involvement). 70 In accordance with GRAPPA 2015 recommendations, the expert panel recommends switching from one DMARD to another, or to biologic treatment, or from one biologic treatment to another. 9
Consensus recommendations for the treatment of PsA domains were based on GRAPPA 2015, GRAPPA 2020, EULAR 2019 recommendations and literature review, and are presented in Table 2.
TABLE 2.
Domain | Recommended therapeutic options (strongly recommended) | Other therapeutic options (conditionally recommended) | To be avoided (strongly not recommended) |
---|---|---|---|
Peripheral arthritis DMARD‐naïve 9 , 11 , 33 |
|
|
|
|
|
||
|
|
||
|
|
|
|
|
|
||
|
|
||
|
|||
|
Abbreviations: CSA, cyclosporine; CTLA‐4 Ig, cytotoxic T lymphocyte‐associated antigen‐4 immunoglobulin; DMARD, disease‐modifying antirheumatic drug; GCC, glucocorticoids; IA, intra‐articular; IL‐12/23i, interleukin‐12/23 inhibitor; IL‐17i, interleukin‐17 inhibitor; IL‐23i, interleukin‐23 inhibitor; IV, intravenous; JAKi, Janus kinase inhibitor; LEF, leflunomide; MTX, methotrexate; NSAIDs, non‐steroidal anti‐inflammatory drugs; PsA, psoriatic arthritis; PDE‐4i phosphodiesterase‐4 inhibitor (apremilast); SC, subcutaneous; TNFi, tumor necrosis factor inhibitor.
3.4. Treatment response
Evaluating response to therapy in patients with PsA can be difficult due to its complex nature, which encompasses a multitude of clinical manifestations. To date, there is no standardized outcome measure for PsA.
In terms of efficacy and safety profile, experts agreed to adhere to the recommendations from guidelines, latest literature evidence and prescription label. Accordingly, consensus statements were developed for efficacy and safety of non‐biologic and biologic therapies.
3.5. Efficacy and safety profile of non‐biologic pharmacological therapies
Consensus statements on efficacy/safety profile of non‐biologic pharmacological therapies are detailed in Table 3
TABLE 3.
Symptomatic treatments |
NSAIDs
|
GCCs
|
csDMARDs |
Methotrexate
|
Leflunomide
|
Sulfasalazine
|
tsDMARDs |
Apremilast
|
Tofacitinib
|
Upadacitinib
|
Abbreviations: bDMARD, biologic disease‐modifying antirheumatic drug; csDMARD, conventional synthetic disease‐modifying antirheumatic drug; COVID‐19, coronavirus disease‐2019; GCC, glucocorticoids; HIV, human immunodeficiency virus; IR, inadequate response; JAKi, Janus kinase inhibitor; MTX, methotrexate; NSAIDs, non‐steroidal anti‐inflammatory drugs; PsA, psoriatic arthritis; TNF, tumor necrosis factor; tsDMARD, targeted synthetic disease‐modifying antirheumatic drug.
3.6. Efficacy and safety profile of biologic pharmacological therapies
Consensus statements on the efficacy and safety of biologic pharmacological therapy are presented in Table 4.
TABLE 4.
TNFi |
|
|
IL‐12/23i |
|
IL‐17i |
|
IL‐23i |
|
CTLA4‐Ig |
|
Abbreviations: ACR, American College of Rheumatology; CTLA4‐Ig, cytotoxic T‐lymphocyte‐associated protein 4‐immunoglobulin; DMARD, disease‐modifying antirheumatic drug; HBV, hepatitis B virus; HCV, hepatitis C virus; HIV, human immunodeficiency virus; IBD, inflammatory bowel disease; IL‐12/23i, interleukin‐12/23 inhibitor; IL‐17i, interleukin‐17 inhibitor; IL‐23i, interleukin‐23 inhibitor; PsA, psoriatic arthritis; SC, subcutaneous; TNFi, tumor necrosis factor inhibitor.
3.7. Treatment failure and switching therapy
The expert panel agreed that transitioning to alternative TNFi should be considered in PsA patients with primary or secondary failure of TNFi therapy. The response to alternative treatment should be assessed with the same criteria as those used for the first TNFi agent. Furthermore, possible consequences for control of skin disease should be considered and referral to a dermatologist also considered, if required. 11
Similarly, switching within a class or between a class (bDMARD to tsDMARD) can also be considered in cases of primary or secondary failure of a bDMARD. However, it is more advisable to change class after a second failure within a given class. 11
3.8. Screening and monitoring requirements for PsA therapies
The expert panel suggested that before initiation of any systemic therapy, a practical approach should be adopted to monitor PsA patients based on medical history, physical examination, and tests (laboratory and imaging). The expert panel opined that monitoring of systemic therapies is crucial to maximize the benefits and minimize the risks associated with these drugs.
Owing to an increased risk of hepatotoxicity and renal toxicity associated with most systemic DMARDs, experts recommended monitoring tests including complete blood count, comprehensive liver function tests, renal function test, erythrocyte sedimentation rate (ESR), C‐reactive protein (CRP), and serum creatinine levels.
As most bDMARDs are immunomodulators, there is a high risk of serious infections, including tuberculosis, hepatitis, and human immunodeficiency virus (HIV). Therefore, it is important that patients are routinely screened for tuberculosis (QuantiFERON), hepatitis B virus (HBV), hepatitis C virus (HCV), and HIV prior to initiating any biologic therapy.
Furthermore, patients should be assessed for their vaccination status before initiating any systemic therapy. Routine vaccination for pertussis and inactivated influenza, pneumococcal, and HBV is recommended in high‐risk patients and in highly prevalent regions at baseline. Varicella‐zoster antibody (IgG) test, especially in patients taking JAKi, should be conducted at baseline. Live vaccines should be avoided during treatment with biologics.
In accordance with the recommendations provided by the Saudi guideline and GRAPPA 2015 and evidence from the literature, the expert panel agreed upon consensus statements on screening and monitoring requirements for PsA therapies presented in Table 5.
TABLE 5.
Recommendations |
---|
|
|
|
|
|
|
|
|
|
Abbreviations: ALP, alkaline phosphatase; ALT, alanine transaminase; ASCVD, atherosclerotic cardiovascular disease; CRP, C‐reactive protein; CVD, cardiovascular disease; ESR, erythrocyte sedimentation rate; GGT, G‐glutamyl transferase; HBV, hepatitis B virus; HCV, hepatitis C virus; HIV, human immunodeficiency virus; IBD, inflammatory bowel disease; IgG, immunoglobulin G; JAKi, Janus kinase inhibitor; PsA, psoriatic arthritis.
3.9. Management of comorbidities
Identifying comorbidities is critical to the optimal management and treatment of PsA. Common comorbidities in patients with PsA include CVD, obesity, metabolic syndrome, hypercholesterolemia, hypertension, diabetes mellitus, chronic kidney disease, malignancy, osteoporosis, non‐alcoholic fatty liver disease (NAFLD), and depression. Moreover, some comorbidities such as inflammatory bowel disease (IBD) and ophthalmic disease (eg uveitis) might present as extra‐articular manifestations of disease.
3.9.1. CVD
The risk of major adverse cardiovascular events has been found to be higher in patients with PsA not prescribed DMARDs compared to the general population or those with psoriasis only. 71 , 72 Based on clinical evidence and GRAPPA 2020 recommendations, JAKi, ustekinumab, IL‐17i, IL‐23i, abatacept could be considered as treatment options in management of PsA patients with comorbid CVD. 14 , 54 , 73 , 74 , 75 , 76 GRAPPA recommends caution with the use of TNFi, glucocorticoids and NSAIDs in patients with congestive heart failure (CHF). 9
3.9.2. Obesity and metabolic syndrome
GRAPPA 2020 recommends that physicians should be cautious about prescribing glucocorticoids to patients with metabolic syndrome, and methotrexate to patients with obesity and metabolic syndrome. 14 , 54
3.9.3. Hypercholesterolemia
He expert panel emphasized the importance of lipid‐lowering drugs and nutritionist referral in PsA patients with comorbid hypercholesterolemia. Evidence suggests that tofacitinib should be used with caution in PsA patients with comorbid hypercholesterolemia. 77
3.9.4. Hypertension
Statins, angiotensin‐converting enzyme inhibitors, and/or angiotensin II blockers are preferred treatment options in patients with PsA and hypertension. 9 Caution should be taken when prescribing NSAIDs, cyclo‐oxygenase‐2 (COX2) inhibitors, or prednisone, as they are associated with an increased risk of CVD. 9 , 78
3.9.5. Diabetes mellitus
The prevalence of type 2 diabetes mellitus in patients with PsA has been reported to be between 6.1% and 20.2% with a higher risk noted in women with more severe forms of PsA. 79 When selecting the treatment for PsA in such patients, most guidelines recommend taking caution with glucocorticoids and methotrexate, as they could worsen glycemic homeostasis and/or influence cardiovascular risk factors such as arterial hypertension. 9 , 11
3.9.6. IBD
There is increased prevalence of IBD and subclinical bowel inflammation among patients with PsA. 80 , 81 Sulfasalazine, TNFi, and tofacitinib (only ulcerative colitis) are approved treatments for IBD. According to 2018 ACR/NPF guidelines, for patients with active PsA and concomitant active IBD who are DMARD‐naïve, monoclonal TNFi are the preferred choice. 11 In patients who are contraindicated for TNFi, IL‐12/23i can be prescribed. The use of NSAIDs and IL‐17i should be avoided, as they may exacerbate IBD symptoms. 82 , 83 Interim analysis reports from a phase II study suggest that guselkumab could be effectively used in patients with Crohn's disease. 84 A long‐term study on the efficacy of adalimumab in the treatment of patients with Crohn's disease with intolerance or inadequate response to infliximab reported sustained clinical remission and response with adalimumab maintenance therapy. 85
3.9.7. Uveitis
The management of concomitant uveitis in PsA patients varies depending on the severity of the disease and its impact on daily activities. 86 Both infliximab and adalimumab are effective treatment options87, 88, 89 certolizumab pegol/golimumab has shown moderate success, while etanercept has demonstrated only limited success. 90 Secukinumab showed promising results in phase II clinical trials; however, primary efficacy endpoint was not met in the phase III study91, 92 The efficacy of IL‐17i, IL‐12/23i, and JAK/STAT (signal transducer and activator of transcription) inhibitors is currently under evaluation. 91 , 92
3.9.8. Depression
The prevalence of depression and anxiety among patients with PsA is 9% to 36% and 15% to 30%, respectively. 93 , 94 As both depression and anxiety may affect pain perception, QoL, and treatment outcomes; it is important to take appropriate screening measures and treatment decisions in such patients. Apremilast should be used with caution in patients with PsA and comorbid depression. 95
3.9.9. Hyperuricemia and gout
Hyperuricemia is common in patients with PsA, especially in those with longer CVD, metabolic syndrome, and disease duration. 96 , 97 , 98 It is therefore important to regularly monitor serum uric acid levels in patients with PsA. 97 Gout is an important differential diagnosis of PsA and, therefore, awareness about its increased incidence in this population is critical. 99
3.9.10. Hypothyroidism
Due to increased incident cases of hypothyroidism, thyroid dysfunction, positive antithyroid peroxidase antibodies (AbTPO), and appearance of a hypoechoic thyroid pattern in patients with PsA, especially women, it is important to evaluate AbTPO levels, thyroid function, and thyroid ultrasound, with regular follow‐up visits. 100 , 101
3.9.11. Osteoporosis
Screening for osteoporosis in psoriatic patients is performed by measuring bone mineral density through dual‐energy X‐ray absorptiometry (DEXA) or assessing the Fracture Risk Assessment (FRAX) score. For management, specific guidelines should be followed for patients treated with chronic systemic glucocorticoids, as it may modify bone mineral density due to bone loss. 102
3.9.12. Malignancy
Given the potential risk of de novo or recurrent malignancy being associated with the use of TNFi, regular screening is recommended, especially in patients with a history of cancer. 9 In contrast, IL‐17i, abatacept have a better safety profile for malignancy and are preferred treatment options in these patients.
3.9.13. Fatty liver disease
Liver disease, particularly NAFLD, has an increased prevalence in patients with psoriasis and PsA. 103 Given the potential risk of liver damage with specific PsA treatments, regular monitoring of liver function abnormalities is deemed necessary. Liver biopsy should also be considered, based on the presence or absence of risk factors for hepatotoxicity and cumulative methotrexate dose. 9 , 11 Furthermore, caution should be taken when prescribing methotrexate, leflunomide, sulfasalazine, and NSAIDs in patients with established liver disease due to the increased risk of hepatotoxicity.
3.9.14. Chronic kidney disease
Methotrexate should be avoided in patients with significant renal insufficiency or end‐stage renal disease on hemodialysis, given that renal impairment is a major risk factor for developing methotrexate toxicity. 104 NSAIDs should also be avoided, given that they may increase the risk for acute kidney injury. 105
3.9.15. Serious infections
There is a high risk of serious infections, including tuberculosis, hepatitis, and HIV, associated with the use of certain PsA treatments—including TNFi. 9 Due to the low incidence of serious infections observed with IL‐12/23i, IL‐17i, and abatacept in comparison to TNFi, the former are preferred treatment options for PsA. 9
Consensus statements on the management of comorbidities in patients with PsA are presented in Table 6.
TABLE 6.
Comorbid condition/s | Treatment options/referral/monitoring | Treatment requiring caution |
---|---|---|
Cardiovascular disease and congestive heart failure | JAKi, ustekinumab, IL‐17i, IL‐23i, abatacept 73 , 74 , 75 , 76 |
NSAIDs, GCCs, TNFi (TNFi should be avoided in patients with severe CHF [NYHA class III and IV] and should be used with caution in patients with mild CHF [NYHA class I and II]) 9 , 54 , 143 , 144 |
Obesity and metabolic syndrome | Weight reduction, nutritionist referral, obesity/endocrine clinic referral | MTX, GCCs 9 |
Hypercholesterolemia | Lipid‐lowering agents, nutritionist referral | Tofacitinib 77 |
Hypertension | Statins, angiotensin‐converting enzyme inhibitors, and/or angiotensin II blockers 9 |
NSAIDs, GCCs 9 |
Diabetes mellitus | Hypoglycemic medications | MTX, GCCs 9 |
IBD |
TNFi (excluding etanercept) for UC (tofacitinib, IL‐12/23i) IL‐23i 84 |
IL‐17i |
Uveitis | TNFi (especially adalimumab and infliximab), MTX | NSAIDs, IL‐17i 145 |
Depression | Psychiatry referral | Apremilast |
Hyperuricemia and gout |
Monitoring serum uric acid levels Urate‐lowering therapy if indicated |
|
Thyroid disease | Routine thyroid tests/endocrine referral | |
Osteoporosis | Monitor with DEXA as indicated in non‐PsA patients | GCCs |
Malignancy | Oncology referral, IL‐17i, abatacept | All biological agents |
Fatty liver disease |
GI referral Weight loss Dietician referral |
NSAIDs, SSZ, MTX, LEF, tofacitinib 54 |
Chronic kidney disease | Nephrologist referral | NSAIDs, MTX 54 |
HBV |
Ustekinumab GI referral, monitor HBV PCR (once a mo for first 3 mo and every 3 mo thereafter) 146 |
NSAIDs, MTX, LEF, biologics (for carriers) 54 |
HCV | GI referral, monitor HCV PCR (once in every 3‐6 mo) 146 | NSAIDs, MTX, LEF, biologics (for carriers) 54 |
Tuberculosis | IL‐17i, abatacept, referral to respiratory or infectious disease specialist | TNFi especially infliximab |
Abbreviations: CHF, congestive heart failure; DEXA, dual‐energy X‐ray absorptiometry; GCCs, glucocorticoids; GI, gastrointestinal; HBV, hepatitis B virus; HCV, hepatitis C virus; IBD, inflammatory bowel disease; IL‐12/23i, interleukin‐12/23 inhibitor; IL‐17i, interleukin‐17 inhibitor; IL‐23i, interleukin‐23 inhibitor; JAKi, Janus kinase inhibitor; LEF, leflunomide; MTX, methotrexate; NHYA, New York Heart Association; NSAIDs, non‐steroidal anti‐inflammatory drugs; PCR, polymerase chain reaction; SSZ, sulfasalazine; TNFi, tumor necrosis factor inhibitor; UC, ulcerative colitis.
3.9.16. Immunization
Immunization status of the patient should be assessed. Routine vaccination for pertussis and inactivated influenza, pneumococcal, and HBV (in high‐risk patients and in highly prevalent regions) should be performed at baseline. 9
4. CONCLUSION
The present consensus statements for the pharmacological management of PsA are in corroboration with established global guidelines on the different aspects of PsA, especially highlighting the management of PsA and associated comorbid conditions and monitoring of therapies in patients with PsA. There is a scarcity of such consensus‐based statements in the Arab world. Furthermore, our consensus statements are aligned with the most recently published Saudi consensus recommendations. 106 Our detailed consensus recommendations can help physicians and healthcare professionals in the UAE to make informed treatment decisions, improvise treatment strategies, monitor therapies, as well as effectively manage comorbidities in patients with PsA.
AUTHOR CONTRIBUTIONS
KAA had a substantive role in drafting the final manuscript. The authors are fully responsible for all the content and editorial decisions; the authors involved themselves at all stages of manuscript development and approved the final version.
CONFLICT OF INTEREST
The authors have no conflict of interest related to this work. The consensus guideline was funded by the Emirates Society for Rheumatology, a non‐profit organization.
ACKNOWLEDGEMENTS
The authors would like to thank Dr Arun Jayarame Gowda and Dr Kavitha Ganesha from IQVIA for providing medical writing support. The authors are fully responsible for all the content and editorial decisions; the authors have involved themselves at all stages of manuscript development and approved the final version.
Alnaqbi KA, Hannawi S, Namas R, Alshehhi W, Badsha H, Al‐Saleh J. Consensus statements for pharmacological management, monitoring of therapies, and comorbidity management of psoriatic arthritis in the United Arab Emirates. Int J Rheum Dis. 2022;25:1107‐1122. doi: 10.1111/1756-185X.14406
REFERENCES
- 1. Bedaiwi M, Al‐Homood IA, El‐Garf A, et al. Disease burden and treatment challenges of psoriatic arthritis in Africa and the Middle East. Rheumatol Int. 2019;39(8):1321‐1329. [DOI] [PubMed] [Google Scholar]
- 2. Stolwijk C, van Onna M, Boonen A, van Tubergen A. Global prevalence of Spondyloarthritis: a systematic review and meta‐regression analysis. Arthritis Care Res (Hoboken). 2016;68(9):1320‐1331. [DOI] [PubMed] [Google Scholar]
- 3. Alamanos Y, Voulgari PV, Drosos AA. Incidence and prevalence of psoriatic arthritis: a systematic review. J Rheumatol. 2008;35(7):1354‐1358. [PubMed] [Google Scholar]
- 4. 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(4):573‐573.e13. [DOI] [PubMed] [Google Scholar]
- 5. Al‐Awadhi AM, Olusi SO, Moussa M, et al. Musculoskeletal pain, disability and health‐seeking behavior in adult Kuwaitis using a validated Arabic version of the WHO‐ILAR COPCORD Core questionnaire. Clin Exp Rheumatol. 2004;22(2):177‐183. [PubMed] [Google Scholar]
- 6. Al Saleh J, Sayed ME, Monsef N, Darwish E. The prevalence and the determinants of musculoskeletal diseases in Emiratis attending primary health care clinics in Dubai. Oman Med J. 2016;31(2):117‐123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kimball AB, Gladman D, Gelfand JM, et al. National Psoriasis Foundation clinical consensus on psoriasis comorbidities and recommendations for screening. J Am Acad Dermatol. 2008;58(6):1031‐1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Husni ME, Merola JF, Davin S. The psychosocial burden of psoriatic arthritis. Semin Arthritis Rheum. 2017;47(3):351‐360. [DOI] [PubMed] [Google Scholar]
- 9. Coates LC, Kavanaugh A, Mease PJ, et al. Group for Research and Assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68(5):1060‐1071. [DOI] [PubMed] [Google Scholar]
- 10. Gossec L, Baraliakos X, Kerschbaumer A, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis. 2020;79(6):700‐712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Singh JA, Guyatt G, Ogdie A, et al. 2018 American College of Rheumatology/National Psoriasis Foundation guideline for the treatment of psoriatic arthritis. Arthritis Rheumatol. 2019;71(1):5‐32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Abdulghani M, Al Sheik A, Alkhawajah M, et al. Management of psoriasis in Africa and the Middle East: a review of current opinion, practice and opportunities for improvement. J Int Med Res. 2011;39(5):1573‐1588. [DOI] [PubMed] [Google Scholar]
- 13. Hamadah IR, Al Raddadi AA, Bahamdan KA, et al. Saudi practical guidelines on biologic treatment of psoriasis. J Dermatolog Treat. 2015;26(3):223‐229. [DOI] [PubMed] [Google Scholar]
- 14. Coates LC, Corp N, van der Windt DA, O'Sullivan D, Soriano ER, Kavanaugh A. GRAPPA treatment recommendations: 2021 update. J Rheumatol. 2022;49(6 Suppl 1):52‐54. [DOI] [PubMed] [Google Scholar]
- 15. Alnaqbi KA, Hannawi S, Namas R, Alshehhi W, Badsha H, Al‐Saleh J. Consensus statements for evaluation and nonpharmacological management of psoriatic arthritis in UAE. Int J Rheum Dis. 2022;25(7):725‐732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kaufman MB. European commission approves upadacitinib for psoriatic arthritis & ankylosing spondylitis; 2021. [11/05/2021]. Available from:. https://www.the‐rheumatologist.org/article/european‐commission‐approves‐upadacitinib‐for‐psoriatic‐arthritis‐ankylosing‐spondylitis/
- 17. Boehncke WH, Brembilla NC, Nissen MJ. Guselkumab: the first selective IL‐23 inhibitor for active psoriatic arthritis in adults. Expert Rev Clin Immunol. 2021;17(1):5‐13. [DOI] [PubMed] [Google Scholar]
- 18. van der Heijde D, Baraliakos X, Gensler LS, et al. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active ankylosing spondylitis (TORTUGA): results from a randomised, placebo‐controlled, phase 2 trial. Lancet. 2018;392(10162):2378‐2387. [DOI] [PubMed] [Google Scholar]
- 19. AbbVie . AbbVie announces first regulatory approval of SKYRIZI™ (risankizumab) for the treatment of plaque psoriasis, generalized pustular psoriasis and erythrodermic psoriasis and psoriatic arthritis in Japan. 2019. [cited 26 March, 2019]. Available from: https://news.abbvie.com/news/abbvie‐announces‐first‐regulatory‐approval‐skyrizi‐risankizumab‐for‐treatment‐plaque‐psoriasis‐generalized‐pustular‐psoriasis‐and‐erythrodermic‐psoriasis‐and‐psoriatic‐arthritis‐in‐japan.htm
- 20. Mease PJ, Helliwell PS, Hjuler KF, Raymond K, McInnes I. Brodalumab in psoriatic arthritis: results from the randomised phase III AMVISION‐1 and AMVISION‐2 trials. Ann Rheum Dis. 2021;80(2):185‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ritchlin CT, Kavanaugh A, Merola JF, et al. Bimekizumab in patients with active psoriatic arthritis: results from a 48‐week, randomised, double‐blind, placebo‐controlled, dose‐ranging phase 2b trial. Lancet. 2020;395(10222):427‐440. [DOI] [PubMed] [Google Scholar]
- 22. PDA Mease, van der Heijde D, Behrens F, Kivitz A, Kim J. Efficacy and safety of deucravacitinib (BMS‐986165), an oral, selective tyrosine kinase 2 inhibitor, in patients with active psoriatic arthritis: results from a phase 2, randomized, double‐blind, placebo‐controlled trial. Abstact number LO3. ACR Convergence; 2020.
- 23. Goodman SM, Cronstein BN, Bykerk VP. Outcomes related to methotrexate dose and route of administration in patients with rheumatoid arthritis: a systematic literature review. Clin Exp Rheumatol. 2015;33(2):272‐278. [PMC free article] [PubMed] [Google Scholar]
- 24. Ceponis A, Kavanaugh A. Use of methotrexate in patients with psoriatic arthritis. Clin Exp Rheumatol. 2010;28(5 Suppl 61):S132‐S137. [PubMed] [Google Scholar]
- 25. Elmamoun M, Chandran V. Role of methotrexate in the management of psoriatic arthritis. Drugs. 2018;78(6):611‐619. [DOI] [PubMed] [Google Scholar]
- 26. Clegg DO, Reda DJ, Abdellatif M. Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondylarthropathies: a Department of Veterans Affairs cooperative study. Arthritis Rheum. 1999;42(11):2325‐2329. [DOI] [PubMed] [Google Scholar]
- 27. Gupta AK, Grober JS, Hamilton TA, et al. Sulfasalazine therapy for psoriatic arthritis: a double blind, placebo controlled trial. J Rheumatol. 1995;22(5):894‐898. [PubMed] [Google Scholar]
- 28. 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(6):1939‐1950. [DOI] [PubMed] [Google Scholar]
- 29. Mulder MLM, Vriezekolk JE, den Broeder N, et al. Comparing methotrexate monotherapy with methotrexate plus leflunomide combination therapy in psoriatic arthritis: protocol of a randomized, placebo‐controlled, double‐blind clinical trial (COMPLETE‐PsA). Trials. 2020;21(1):155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Cutolo M, Myerson GE, Fleischmann RM, et al. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J Rheumatol. 2016;43(9):1724‐1734. [DOI] [PubMed] [Google Scholar]
- 31. Edwards CBF, Crowley J, Hu C, Stevens R, Birbara C. Long‐term (52‐week) results of a phase 3, randomized, controlled trial of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement (PALACE 3). 2013. ACR/ARHP Annual Meeting 2013.
- 32. Kavanaugh A, Mease PJ, Gomez‐Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomised, placebo‐controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020‐1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wells FAEC, Adebajo A, Kivitz A, Bird P, Shah K, et al. A premilast in the treatment of DMARD‐naïve psoriatic arthritis patients: results of a phase 3 randomized, controlled trial (PALACE 4).: L4. Arthritis Rheumatism. 2013;65:3320‐3321. [Google Scholar]
- 34. Ly K, Beck KM, Smith MP, Orbai AM, Liao W. Tofacitinib in the management of active psoriatic arthritis: patient selection and perspectives. Psoriasis (Auckl). 2019;9:97‐107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kang EJ, Kavanaugh A. Psoriatic arthritis: latest treatments and their place in therapy. Ther Adv Chronic Dis. 2015;6(4):194‐203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. D'Angelo S, Tramontano G, Gilio M, Leccese P, Olivieri I. Review of the treatment of psoriatic arthritis with biological agents: choice of drug for initial therapy and switch therapy for non‐responders. Open Access Rheumatol. 2017;9:21‐28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Savage LJ, Wittmann M, McGonagle D, Helliwell PS. Ustekinumab in the treatment of psoriasis and psoriatic arthritis. Rheumatol Ther. 2015;2(1):1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Nash P, Kirkham B, Okada M, et al. Ixekizumab for the treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from the 24‐week randomised, double‐blind, placebo‐controlled period of the SPIRIT‐P2 phase 3 trial. Lancet. 2017;389(10086):2317‐2327. [DOI] [PubMed] [Google Scholar]
- 39. Nash P, Mease PJ, McInnes IB, et al. Efficacy and safety of secukinumab administration by autoinjector in patients with psoriatic arthritis: results from a randomized, placebo‐controlled trial (FUTURE 3). Arthritis Res Ther. 2018;20(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. USFDA . TREMFYA (guselkumab) injection, for subcutaneous use. Administration USFaD. US FDA; 2017. [Google Scholar]
- 41. Agency EM . Skyrizi. Available from:. https://www.ema.europa.eu/en/medicines/human/EPAR/skyrizi
- 42. U.S. FDA approves second indication for SKYRIZI® (risankizumab‐rzaa) to treat adults with active psoriatic arthritis [press release]. 2022.
- 43. Zizzo G, Gremese E, Ferraccioli G. Abatacept in the treatment of psoriatic arthritis: biological and clinical profiles of the responders. Immunotherapy. 2018;10(9):807‐821. [DOI] [PubMed] [Google Scholar]
- 44. Chingcuanco F, Segal JB, Kim SC, Alexander GC. Bioequivalence of biosimilar tumor necrosis factor‐alpha inhibitors compared with their reference biologics: a systematic review. Ann Intern Med. 2016;165(8):565‐574. [DOI] [PubMed] [Google Scholar]
- 45. Cohen S, Kay J. Biosimilars: implications for rheumatoid arthritis therapy. Curr Opin Rheumatol. 2017;29(3):260‐268. [DOI] [PubMed] [Google Scholar]
- 46. Agency EM . Biosimilars in EU; 2017. Available from: https://www.ema.europa.eu/en/documents/leaflet/biosimilars‐eu‐information‐guide‐healthcare‐professionals_en.pdf
- 47. Kabir ER, Moreino SS, Sharif Siam MK. The breakthrough of Biosimilars: a twist in the narrative of biological therapy. Biomolecules. 2019;9(9):410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Deodhar A, Gottlieb AB, Boehncke WH, et al. Efficacy and safety of guselkumab in patients with active psoriatic arthritis: a randomised, double‐blind, placebo‐controlled, phase 2 study. Lancet. 2018;391(10136):2213‐2224. [DOI] [PubMed] [Google Scholar]
- 49. Mease PJ, Gottlieb AB, van der Heijde D, et al. Efficacy and safety of abatacept, a T‐cell modulator, in a randomised, double‐blind, placebo‐controlled, phase III study in psoriatic arthritis. Ann Rheum Dis. 2017;76(9):1550‐1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Deodhar A RR, Ganz F, Gao T, Anderson JK, Oster A, et al. Efficacy and safety of upadacitinib in patients with psoriatic arthritis and axial involvement. ACR Convergence 2020. 2020.
- 51. van der Heijde D, Song IH, Pangan AL, et al. Efficacy and safety of upadacitinib in patients with active ankylosing spondylitis (SELECT‐AXIS 1): a multicentre, randomised, double‐blind, placebo‐controlled, phase 2/3 trial. Lancet. 2019;394(10214):2108‐2117. [DOI] [PubMed] [Google Scholar]
- 52. Baeten D, Sieper J, Braun J, et al. Secukinumab, an interleukin‐17a inhibitor in ankylosing spondylitis. N Engl J Med. 2015;373(26):2534‐2548. [DOI] [PubMed] [Google Scholar]
- 53. Mease PJ, Lertratanakul A, Anderson JK, et al. Upadacitinib for psoriatic arthritis refractory to biologics: SELECT‐PsA 2. Ann Rheum Dis. 2020;80(3):312‐320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Coates LC, Corp N, van der Windt DA, Soriano ER, Kavanaugh A. GRAPPA treatment recommendations: an update from the 2020 GRAPPA annual meeting. J Rheumatol. 2021;97:jrheum.201681‐jrheum.201666. [DOI] [PubMed] [Google Scholar]
- 55. Agency EM . Xeljanz: Pfizer Europe MA EEIG; 2022. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/xeljanz
- 56. U.S. FDA Approves Pfizer's XELJANZ® (tofacitinib) for the Treatment of Active Ankylosing Spondylitis . Available from: https://www.pfizer.com/news/press‐release/press‐release‐detail/us‐fda‐approves‐pfizers‐xeljanzr‐tofacitinib‐treatment‐0
- 57. Orbai AM, Weitz J, Siegel EL, et al. Systematic review of treatment effectiveness and outcome measures for enthesitis in psoriatic arthritis. J Rheumatol. 2014;41(11):2290‐2294. [DOI] [PubMed] [Google Scholar]
- 58. Mease PJ, Gladman DD, Deodhar A, et al. Impact of guselkumab, an interleukin‐23 p19 subunit inhibitor, on enthesitis and dactylitis in patients with moderate to severe psoriatic arthritis: results from a randomised, placebo‐controlled, phase II study. RMD Open. 2020;6(2):e001217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Koppikar S, Eder L. The management of enthesitis in clinical practice. Curr Opin Rheumatol. 2020;32(4):380‐386. [DOI] [PubMed] [Google Scholar]
- 60. Gladman DD, Kavanaugh A, Gomez‐Reino JJ, et al. Therapeutic benefit of apremilast on enthesitis and dactylitis in patients with psoriatic arthritis: a pooled analysis of the PALACE 1‐3 studies. RMD Open. 2018;4(1):e000669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Coates LC, Wallman JK, McGonagle D, et al. Secukinumab efficacy on resolution of enthesitis in psoriatic arthritis: pooled analysis of two phase 3 studies. Arthritis Res Ther. 2019;21(1):266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nash P, Coates LC, Fleischmann R, et al. Efficacy of Tofacitinib for the treatment of psoriatic arthritis: pooled analysis of two phase 3 studies. Rheumatol Ther. 2018;5(2):567‐582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Kavanaugh A, Mease PJ, Reimold AM, et al. Secukinumab for Long‐term treatment of psoriatic arthritis: a two‐year Followup from a phase III, randomized, double‐blind placebo‐controlled study. Arthritis Care Res (Hoboken). 2017;69(3):347‐355. [DOI] [PubMed] [Google Scholar]
- 64. Reich K, Warren RB, Coates LC, Di Comite G. Long‐term efficacy and safety of secukinumab in the treatment of the multiple manifestations of psoriatic disease. J Eur Acad Dermatol Venereol. 2020;34(6):1161‐1173. [DOI] [PubMed] [Google Scholar]
- 65. Ricceri F, Pescitelli L, Tripo L, Bassi A, Prignano F. Treatment of severe nail psoriasis with acitretin: an impressive therapeutic result. Dermatol Ther. 2013;26(1):77‐78. [DOI] [PubMed] [Google Scholar]
- 66. Lanna C, Cesaroni GM, Mazzilli S, et al. Apremilast as a target therapy for nail psoriasis: a real‐life observational study proving its efficacy in restoring the nail unit. J Dermatolog Treat. 2020;33(2):1097‐1101. [DOI] [PubMed] [Google Scholar]
- 67. Foley P, Gordon K, Griffiths CEM, et al. Efficacy of Guselkumab compared with Adalimumab and placebo for psoriasis in specific body regions: a secondary analysis of 2 randomized clinical trials. JAMA Dermatol. 2018;154(6):676‐683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Gumusel M, Ozdemir M, Mevlitoglu I, Bodur S. Evaluation of the efficacy of methotrexate and cyclosporine therapies on psoriatic nails: a one‐blind, randomized study. J Eur Acad Dermatol Venereol. 2011;25(9):1080‐1084. [DOI] [PubMed] [Google Scholar]
- 69. Behrens F, Finkenwirth C, Pavelka K, et al. Leflunomide in psoriatic arthritis: results from a large European prospective observational study. Arthritis Care Res (Hoboken). 2013;65(3):464‐470. [DOI] [PubMed] [Google Scholar]
- 70. Mease PJ, Smolen JS, Behrens F, et al. A head‐to‐head comparison of the efficacy and safety of ixekizumab and adalimumab in biological‐naïve patients with active psoriatic arthritis: 24‐week results of a randomised, open‐label, blinded‐assessor trial. Rheumatol Ther. 2020;79(1):123‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Ogdie A, Yu Y, Haynes K, et al. Risk of major cardiovascular events in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a population‐based cohort study. Ann Rheum Dis. 2015;74(2):326‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. 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(12):1729‐1735. [DOI] [PubMed] [Google Scholar]
- 73. Xie W, Xiao S, Huang Y, Sun X, Zhang Z. Effect of tofacitinib on cardiovascular events and all‐cause mortality in patients with immune‐mediated inflammatory diseases: a systematic review and meta‐analysis of randomized controlled trials. Ther Adv Musculoskelet Dis 2019;11:1759720X19895492, 1759720X1989549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lee MP, Desai RJ, Jin Y, Brill G, Ogdie A, Kim SC. Association of Ustekinumab vs TNF inhibitor therapy with risk of atrial fibrillation and cardiovascular events in patients with psoriasis or psoriatic arthritis. JAMA Dermatol. 2019;155(6):700‐707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ramonda R, Lorenzin M, Carriero A, Chimenti MS, Scarpa R, Marchesoni A, Lubrano di Scorpaniello E, Salvarani C, Cauli A, Semeraro A, Santo L, Ortolan A, Doria A, Fracassi E, Virelli G, Masia M, Fanizzi R, Visalli E, Amato G, Carletto A, Foti R, on behalf Spondyloartritis and Psoriatic Arthritis SIR Study Group “Antonio Spadaro” . Effectiveness and safety of secukinumab in 608 patients with psoriatic arthritis in real life: a 24‐month prospective, multicentre study. RMD Open 2021;7(1):e001519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Kang EH, Jin Y, Brill G, et al. Comparative cardiovascular risk of Abatacept and tumor necrosis factor inhibitors in patients with rheumatoid arthritis with and without diabetes mellitus: a multidatabase cohort study. J Am Heart Assoc. 2018;7(3):e007393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Gladman DD, Charles‐Schoeman C, McInnes IB, et al. Changes in lipid levels and incidence of cardiovascular events following Tofacitinib treatment in patients with psoriatic arthritis: a pooled analysis across phase III and Long‐term extension studies. Arthritis Care Res (Hoboken). 2019;71(10):1387‐1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Zhu TY, Li EK, Tam LS. Cardiovascular risk in patients with psoriatic arthritis. Int J Rheumatol 2012;2012:714321, 1, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Dal Bello G, Gisondi P, Idolazzi L, Girolomoni G. Psoriatic arthritis and diabetes mellitus: a narrative review. Rheumatol Ther. 2020;7(2):271‐285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Li WQ, Han JL, Chan AT, Qureshi AA. Psoriasis, psoriatic arthritis and increased risk of incident Crohn's disease in US women. Ann Rheum Dis. 2013;72(7):1200‐1205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Scarpa R, Manguso F, D'Arienzo A, et al. Microscopic inflammatory changes in colon of patients with both active psoriasis and psoriatic arthritis without bowel symptoms. J Rheumatol. 2000;27(5):1241‐1246. [PubMed] [Google Scholar]
- 82. Hohenberger M, Cardwell LA, Oussedik E, Feldman SR. Interleukin‐17 inhibition: role in psoriasis and inflammatory bowel disease. J Dermatolog Treat. 2018;29(1):13‐18. [DOI] [PubMed] [Google Scholar]
- 83. Long MD, Kappelman MD, Martin CF, Chen W, Anton K, Sandler RS. Role of nonsteroidal anti‐inflammatory drugs in exacerbations of inflammatory bowel disease. J Clin Gastroenterol. 2016;50(2):152‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sandborn WJCD, Johanns J, Lang G, Adedokun OJ, Afzali A, et al. The efficacy and safety OF guselkumab induction therapy IN patients with moderately to severely active CROHN'S disease: week 12 interim analyses from the phase 2 GALAXI 1 study. United Eur Gastroenterol J. 2020;160:S‐350‐S‐351. [Google Scholar]
- 85. Panaccione R, Sandborn WJ, D'Haens G, et al. Clinical benefit of Long‐term Adalimumab treatment in patients with Crohn's disease following loss of response or intolerance to infliximab: 96‐week efficacy data from GAIN/ADHERE trials. J Crohns Colitis. 2018;12(8):930‐938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Rosenbaum JT. Uveitis in spondyloarthritis including psoriatic arthritis, ankylosing spondylitis, and inflammatory bowel disease. Clin Rheumatol. 2015;34(6):999‐1002. [DOI] [PubMed] [Google Scholar]
- 87. Rudwaleit M, Rodevand E, Holck P, et al. Adalimumab effectively reduces the rate of anterior uveitis flares in patients with active ankylosing spondylitis: results of a prospective open‐label study. Ann Rheum Dis. 2009;68(5):696‐701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Fotiadou C, Lazaridou E, Kemanetzi C, Kyrmanidou E, Ioannides D. Recalcitrant psoriatic uveitis and anti‐tumor necrosis factor‐alpha monoclonal antibodies: experience from a psoriasis referral center. Int J Dermatol. 2015;54(9):1105‐1108. [DOI] [PubMed] [Google Scholar]
- 89. Guignard S, Gossec L, Salliot C, et al. Efficacy of tumour necrosis factor blockers in reducing uveitis flares in patients with spondylarthropathy: a retrospective study. Ann Rheum Dis. 2006;65(12):1631‐1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Arepalli S, Rosenbaum JT. The use of biologics for uveitis associated with spondyloarthritis. Curr Opin Rheumatol. 2019;31(4):349‐354. [DOI] [PubMed] [Google Scholar]
- 91. Rosenbaum JT, Bodaghi B, Couto C, et al. New observations and emerging ideas in diagnosis and management of non‐infectious uveitis: a review. Semin Arthritis Rheum. 2019;49(3):438‐445. [DOI] [PubMed] [Google Scholar]
- 92. Pepple KL, Lin P. Targeting Interleukin‐23 in the treatment of noninfectious uveitis. Ophthalmology. 2018;125(12):1977‐1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. McDonough E, Ayearst R, Eder L, et al. Depression and anxiety in psoriatic disease: prevalence and associated factors. J Rheumatol. 2014;41(5):887‐896. [DOI] [PubMed] [Google Scholar]
- 94. Kamalaraj N, El‐Haddad C, Hay P, Pile K. Systematic review of depression and anxiety in psoriatic arthritis. Int J Rheum Dis. 2019;22(6):967‐973. [DOI] [PubMed] [Google Scholar]
- 95. Zerilli T, Ocheretyaner E. Apremilast (Otezla): a new Oral treatment for adults with psoriasis and psoriatic arthritis. P T. 2015;40(8):495‐500. [PMC free article] [PubMed] [Google Scholar]
- 96. Li X, Miao X, Wang H, et al. Association of serum uric acid levels in psoriasis: a systematic review and meta‐analysis. Medicine (Baltimore). 2016;95(19):e3676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. AlJohani R, Polachek A, Ye JY, Chandran V, Gladman DD. Characteristic and outcome of psoriatic arthritis patients with hyperuricemia. J Rheumatol. 2018;45(2):213‐217. [DOI] [PubMed] [Google Scholar]
- 98. Lai TL, Yim CW, Wong PY, Leung MC, Ng WL. Hyperuricemia in Asian psoriatic arthritis patients. Int J Rheum Dis. 2018;21(4):843‐849. [DOI] [PubMed] [Google Scholar]
- 99. Merola JF, Wu S, Han J, Choi HK, Qureshi AA. Psoriasis, psoriatic arthritis and risk of gout in US men and women. Ann Rheum Dis. 2015;74(8):1495‐1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Ruffilli I, Ragusa F, Benvenga S, et al. Psoriasis, psoriatic arthritis, and thyroid autoimmunity. Front Endocrinol (Lausanne). 2017;8:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Fallahi P, Ferrari SM, Ruffilli I, et al. Increased incidence of autoimmune thyroid disorders in patients with psoriatic arthritis: a longitudinal follow‐up study. Immunol Res. 2017;65(3):681‐686. [DOI] [PubMed] [Google Scholar]
- 102. Grossman JM, Gordon R, Ranganath VK, et al. American College of Rheumatology 2010 recommendations for the prevention and treatment of glucocorticoid‐induced osteoporosis. Arthritis Care Res (Hoboken). 2010;62(11):1515‐1526. [DOI] [PubMed] [Google Scholar]
- 103. Madanagobalane S, Anandan S. The increased prevalence of non‐alcoholic fatty liver disease in psoriatic patients: a study from South India. Australas J Dermatol. 2012;53(3):190‐197. [DOI] [PubMed] [Google Scholar]
- 104. Boey O, Van Hooland S, Woestenburg A, Van der Niepen P, Verbeelen D. Methotrexate should not be used for patients with end‐stage kidney disease. Acta Clin Belg. 2006;61(4):166‐169. [DOI] [PubMed] [Google Scholar]
- 105. Zhang X, Donnan PT, Bell S, Guthrie B. Non‐steroidal anti‐inflammatory drug induced acute kidney injury in the community dwelling general population and people with chronic kidney disease: systematic review and meta‐analysis. BMC Nephrol. 2017;18(1):256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Al Rayes H, Alazmi M, Attar S, et al. Consensus‐based recommendations on the diagnosis, referral and clinical management of patients with psoriatic arthritis. Rheumatol Int. 2022;42(3):391‐401. [DOI] [PubMed] [Google Scholar]
- 107. Genovese MC, Mease PJ, Thomson GT, et al. Safety and efficacy of adalimumab in treatment of patients with psoriatic arthritis who had failed disease modifying antirheumatic drug therapy. J Rheumatol. 2007;34(5):1040‐1050. [PubMed] [Google Scholar]
- 108. 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(4):712‐721. [PubMed] [Google Scholar]
- 109. Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment of psoriatic arthritis: safety, efficacy, and effect on disease progression. Arthritis Rheum. 2004;50(7):2264‐2272. [DOI] [PubMed] [Google Scholar]
- 110. Mease PJ, Goffe BS, Metz J, VanderStoep A, Finck B, Burge DJ. Etanercept in the treatment of psoriatic arthritis and psoriasis: a randomised trial. Lancet. 2000;356(9227):385‐390. [DOI] [PubMed] [Google Scholar]
- 111. 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(5):869‐876. [PubMed] [Google Scholar]
- 112. Kavanaugh A, Krueger GG, Beutler A, et al. Infliximab maintains a high degree of clinical response in patients with active psoriatic arthritis through 1 year of treatment: results from the IMPACT 2 trial. Ann Rheum Dis. 2007;66(4):498‐505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Acosta‐Felquer ML, Rosa J, Soriano ER. An evidence‐based review of certolizumab pegol in the treatment of active psoriatic arthritis: place in therapy. Open Access Rheumatol. 2016;8:37‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Esposito M, Carubbi F, Giunta A, Alunno A, Giacomelli R, Fargnoli MC. Certolizumab pegol for the treatment of psoriatic arthritis and plaque psoriasis. Expert Rev Clin Immunol. 2020;16(2):119‐128. [DOI] [PubMed] [Google Scholar]
- 115. Weinblatt ME, Bingham CO 3rd, Mendelsohn AM, et al. Intravenous golimumab is effective in patients with active rheumatoid arthritis despite methotrexate therapy with responses as early as week 2: results of the phase 3, randomised, multicentre, double‐blind, placebo‐controlled GO‐FURTHER trial. Ann Rheum Dis. 2013;72(3):381‐389. [DOI] [PubMed] [Google Scholar]
- 116. Blair HA. Secukinumab: a review in psoriatic arthritis. Drugs. 2021;81(4):483‐494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. USFDA . Taltz(ixekizumab) injection, for subcutaneous use. 2017. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125521s004lbl.pdf
- 118. Ursini F, Russo E, De Giorgio R, De Sarro G, D'Angelo S. Current treatment options for psoriatic arthritis: spotlight on abatacept. Ther Clin Risk Manag. 2018;14:1053‐1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Edwards CJ, Blanco FJ, Crowley J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann Rheum Dis. 2016;75(6):1065‐1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Reich K, Puig L, Szepietowski JC, et al. Secukinumab dosing optimization in patients with moderate‐to‐severe plaque psoriasis: results from the randomized, open‐label OPTIMISE study. Br J Dermatol. 2020;182(2):304‐315. [DOI] [PubMed] [Google Scholar]
- 121. Chiricozzi A, Panduri S, Dini V, Tonini A, Gualtieri B, Romanelli M. Optimizing acitretin use in patients with plaque psoriasis. Dermatol Ther. 2017;30(2):e12453. [DOI] [PubMed] [Google Scholar]
- 122. Valenzuela F, Korman NJ, Bissonnette R, et al. Tofacitinib in patients with moderate‐to‐severe chronic plaque psoriasis: long‐term safety and efficacy in an open‐label extension study. Br J Dermatol. 2018;179(4):853‐862. [DOI] [PubMed] [Google Scholar]
- 123. Drvar DL, Vlahinic T, Males Z, Turcic P, Ceovic R. A modern approach to the treatment of plaque psoriasis. Acta Pharm. 2019;69(4):511‐523. [DOI] [PubMed] [Google Scholar]
- 124. Rogojan C, Hetland ML. Depigmentation‐‐a rare side effect to intra‐articular glucocorticoid treatment. Clin Rheumatol. 2004;23(4):373‐375. [DOI] [PubMed] [Google Scholar]
- 125. Sakellariou GT, Sayegh FE, Anastasilakis AD, Kapetanos GA. Leflunomide addition in patients with articular manifestations of psoriatic arthritis resistant to methotrexate. Rheumatol Int. 2013;33(11):2917‐2920. [DOI] [PubMed] [Google Scholar]
- 126. Farr M, Kitas GD, Waterhouse L, Jubb R, Felix‐Davies D, Bacon PA. Sulphasalazine in psoriatic arthritis: a double‐blind placebo‐controlled study. Br J Rheumatol. 1990;29(1):46‐49. [DOI] [PubMed] [Google Scholar]
- 127. Keskin Y, Koz G, Nas K. What has changed in the treatment of psoriatic arthritis after COVID‐19? Eurasian J Med. 2021;53(2):132‐136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Seo MR, Kim JW, Park EJ, et al. Recommendations for the management of patients with systemic rheumatic diseases during the coronavirus disease pandemic. Korean J Intern Med. 2020;35(6):1317‐1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. 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(10):3279‐3289. [DOI] [PubMed] [Google Scholar]
- 130. 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(5):702‐709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. 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(8):2698‐2707. [DOI] [PubMed] [Google Scholar]
- 132. 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(4):1227‐1236. [DOI] [PubMed] [Google Scholar]
- 133. 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(8):1150‐1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. 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 placebo‐controlled study (RAPID‐PsA). Ann Rheum Dis. 2014;73(1):48‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Mease P, Deodhar A, Fleischmann R, et al. Effect of certolizumab pegol over 96 weeks in patients with psoriatic arthritis with and without prior antitumour necrosis factor exposure. RMD Open. 2015;1(1):e000119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Kavanaugh A, McInnes IB, Mease P, et al. Clinical efficacy, radiographic and safety findings through 5 years of subcutaneous golimumab treatment in patients with active psoriatic arthritis: results from a long‐term extension of a randomised, placebo‐controlled trial (the GO‐REVEAL study). Ann Rheum Dis. 2014;73(9):1689‐1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. 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(9894):780‐789. [DOI] [PubMed] [Google Scholar]
- 138. Deodhar A, Mease PJ, McInnes IB, et al. Long‐term safety of secukinumab in patients with moderate‐to‐severe plaque psoriasis, psoriatic arthritis, and ankylosing spondylitis: integrated pooled clinical trial and post‐marketing surveillance data. Arthritis Res Ther. 2019;21(1):111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Mease PJ, van der Heijde D, Ritchlin CT, et al. Ixekizumab, an interleukin‐17A specific monoclonal antibody, for the treatment of biologic‐naive patients with active psoriatic arthritis: results from the 24‐week randomised, double‐blind, placebo‐controlled and active (adalimumab)‐controlled period of the phase III trial SPIRIT‐P1. Ann Rheum Dis. 2017;76(1):79‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Deodhar A, Helliwell PS, Boehncke WH, et al. Guselkumab in patients with active psoriatic arthritis who were biologic‐naive or had previously received TNFalpha inhibitor treatment (DISCOVER‐1): a double‐blind, randomised, placebo‐controlled phase 3 trial. Lancet. 2020;395(10230):1115‐1125. [DOI] [PubMed] [Google Scholar]
- 141. Mease PJ, Rahman P, Gottlieb AB, et al. Guselkumab in biologic‐naive patients with active psoriatic arthritis (DISCOVER‐2): a double‐blind, randomised, placebo‐controlled phase 3 trial. Lancet. 2020;395(10230):1126‐1136. [DOI] [PubMed] [Google Scholar]
- 142. Sunzini F, McInnes I, Siebert S. JAK inhibitors and infections risk: focus on herpes zoster. Ther Adv Musculoskelet Dis. 2020;12:1759720X20936059, 1759720X2093605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Gerriets V BP, Goyal A, Khaddour K. Tumor necrosis factor inhibitors. StatPearls Publishing LLC; 2021. Available from: https://www.ncbi.nlm.nih.gov/books/NBK482425/ [PubMed] [Google Scholar]
- 144. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT, Anti TNFTACHFI . Randomized, double‐blind, placebo‐controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor‐alpha, in patients with moderate‐to‐severe heart failure: results of the anti‐TNF therapy against congestive heart failure (ATTACH) trial. Circulation. 2003;107(25):3133‐3140. [DOI] [PubMed] [Google Scholar]
- 145. McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL‐17A in axial spondyloarthritis and psoriatic arthritis: recent advances and controversies. Ann Rheum Dis. 2019;78(9):1167‐1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Bonifati C, Lora V, Graceffa D, Nosotti L. Management of psoriasis patients with hepatitis B or hepatitis C virus infection. World J Gastroenterol. 2016;22(28):6444‐6455. [DOI] [PMC free article] [PubMed] [Google Scholar]