Abstract
Tumor necrosis factor a (TNF-α)-targeted therapies have expanded the therapeutic options for patients with inflammatory bowel disease (IBD), rheumatoid arthritis (RA), psoriasis, and psoriatic arthritis (PsA) and have significantly improved patients’ quality of life. Paradoxically, anti-TNF-α agents may induce psoriatic eruptions or worsen preexisting psoriatic skin disease. Currently, there is no standard approach for the management of TNF inhibitor-induced psoriasis. Here, we conduct a literature review on TNF inhibitor-induced psoriasis and introduce a novel treatment algorithm for maintaining otherwise effective anti-TNF therapy versus switching to a different class as appropriate in the management of patients with IBD, RA, psoriasis, or PsA.
Keywords: TNF inhibitor-induced psoriasis, biologic agents, TNF-α
Introduction
There is substantial evidence to support the role of tumor necrosis factor α (TNF-α) in the pathogenesis of several inflammatory conditions which has led to the increased use of TNF-α-targeted therapies to treat inflammatory bowel disease (IBD), rheumatoid arthritis (RA), psoriasis, and psoriatic arthritis (PsA).1,2 Currently, there are 5 existing TNF-α inhibitors approved by the Food and Drug Administration (FDA): etanercept, infliximab, adalimumab, certolizumab pegol, and golimumab. The development of these biologic drugs expanded the therapeutic options for patients with these conditions and has significantly improved their quality of life.3–5
Paradoxically, anti-TNF-α agents may also induce and/or worsen psoriatic skin lesions in about 0.6% to 5.3% of patients, with Crohn disease and RA being the most common underlying diseases.6 This cutaneous reaction can be present clinically as plaque psoriasis (15.8%–50%), palmoplantar pustular psoriasis (33.3%–45%), psoriasiform (19.9%), guttate (7%–15%), generalized pustular psoriasis (5.3%–12%), and inverse psoriasis(1.7%–4%).7–11 Eruptions can also appear on the nails and scalp.6 Interestingly, patients can exhibit lesions with different morphological features.12
The histopathologic features involve a spectrum of cutaneous reactions that include psoriasis-like patterns, eosinophilic hypersensitivity reactions, lichen planus–like dermatitis, or a sterile pustular folliculitis.12,13 In our experience, aside from the palmoplantar variant, the presentation of a psoriasiform dermatitis with clinical overlapping features of eczematous hypersensitivity and psoriasis (supported by histopathology of a spongiotic psoriasiform dermatitis with eosinophils) is one of the more common presentations suggestive of this entity.12
In the current literature, infliximab (52.6%–62.5%) is the most reported TNF-α inhibitor to elicit psoriasiform eruptions; etanercept (12–29%), adalimumab (14.4–34%), certolizumab pegol (1%), and golimumab (0.5%) have also been implicated.6,7,9,10 Because these lesions have been reported with all TNF-α inhibitor agents, TNF inhibitor-induced psoriasis is suggested to be a class effect.14
Currently, there is no standard approach for the management of TNF inhibitor-induced psoriasis. Here, we present a literature review on TNF inhibitor-induced psoriasis and introduce a treatment algorithm for maintaining otherwise effective anti-TNF therapy versus switching to another class as appropriate in the management of patients with IBD, RA, psoriasis, or PsA in the setting of TNF inhibitor-induced psoriasis.
Tumor Necrosis Factor Inhibitor-Induced Psoriasis in Inflammatory Conditions
Rheumatoid Arthritis
The prevalence of psoriasis during anti-TNF treatment in RA is estimated at 2.3% to 5%.15,16 Data from a large prospective cohort in the United Kingdom evidenced that the incidence rate of psoriasis in patients with RA treated with anti-TNF agents is significantly higher (1.04; 95% confidence interval [CI]: 0.67–1.54 per 1000 person years) compared to patients treated with conventional diseasemodifying antirheumatic drugs (rate of 0; upper 97.5% CI: 0.71 per 1000 person-years) in a 5207 person-years of follow-up. Furthermore, they found that the incidence rate of psoriasis among those treated with adalimumab was significantly higher compared to those treated with etanercept and infliximab17; these findings were later supported by Joyau et al.11 Interestingly, Hernandez et al found a global incident rate of 2.3/1000 patient-years, but subanalysis among different biologic agents does not show significant differences.18 Overall, patients with RA developing anti TNF-induced psoriasis tended to be females, with no family history of psoriasis, and the median time from initiation of therapy to the onset of psoriasis was about 6 months.17 Pustular psoriasis was the phenotype most described followed by plaque psoriasis.9 Of note, treatment with certolizumab pegol and golimumab has also been associated with anti-TNF-induced psoriasis in RA.19,20
Inflammatory Bowel Disease
Although the prevalence of TNF inhibitor-induced psoriasis in IBD is estimated at 1.6% to 2.7%,21,22 the psoriasis incidence rate is 0.53% per patient year with a cumulative incidence estimated at 1% at year 1, 2.5% at 5 years, and 4.5% at 10 years.23 Of note, being a smoker or having history of smoking was associated with a higher risk of developing TNF inhibitor-induced psoriasis in several studies.23 Furthermore, Guerra et al also found that female sex increased the risk of anti-TNF-induced psoriasis in patients with IBD.23 Although a higher absolute frequency number of cases have been reported among patients treated with infliximab, the incident rate of TNF inhibitor-induced psoriasis was similar for all anti-TNF agents.23 A possible explanation for the high absolute frequency is that adalimumab was introduced as an IBD therapy later compared to infliximab.22 In general, patients with IBD presenting with TNF inhibitor-induced psoriasis did not have personal or family history of psoriasis, the median time from initiation of therapy to the development of skin lesions was 12 months (maintenance period), and the most common phenotype of psoriasis was palmoplantar pustulosis.21–23 Interestingly, in 1 case series of 14 pediatric patients with IBD treated with anti-TNF-α therapy, psoriasis lesions most commonly occurred during IBD remission.24
Psoriasis and PsA
Tumor necrosis factor inhibitor-induced psoriasis has also been reported in patients with psoriasis and PsA, though the prevalence of new-onset TNF-induced psoriasis is unclear.25,26 These paradoxical psoriatic eruptions sometimes manifest in originally unaffected skin, suggesting de novo psoriasis rather than exacerbation of previous lesions.9,11 Importantly, patients may exhibit a new or different morphology from their original presentation of psoriasis.6,9 Indeed, patients with a history of plaque or guttate psoriasis may develop pustular lesions after TNF-α inhibitor therapy.8 Moreover, patients with psoriasis with no history of guttate psoriasis may develop guttate lesions during TNF-α inhibitor therapy.11 In fact, the prevalence of guttate psoriasis in these patients is estimated at 53% to 78%.8,9 Paradoxical onset of PsA was also observed by Napolitano et al in a cohort of 327 patients receiving biologic therapy.26 This study identified 22 cases, of which 18 patients were receiving anti-TNF therapy and 4 were receiving ustekinumab. The majority of the cases presented with peripheral arthritis, with oligoarthritis being the most common presentation.26 Unlike the cases in RA and IBD, women and men were equally affected in these studies.
Proposed Pathogenesis of TNF-α Inhibitor Induced Psoriasiform Dermatitis
The mechanism of TNF inhibitor-induced psoriasis remains unclear. Several theories have been proposed to explain the etiology of this cutaneous reaction, which are briefly discussed below.
Increased Type 1 Interferon
Classical psoriasis is thought to be initiated by skin plasmacytoid dendritic cells (pDCs), which are normally downregulated by TNF-α.27 These pDCs produce type 1 interferons (IFNs) which induce the autoimmune response by promoting activation and maturation of conventional dendritic cells that stimulate CD8+ T cells to migrate into the epidermis and promote the growth of psoriatic lesions.28Accordingly, one theory regarding the mechanism of TNF-α inhibitor-induced psoriasis hypothesizes that blocking TNF-α allows an increased and uncontrolled production of type 1 IFNs by pDCs, which may induce and/or worsen psoriasiform lesions.8 Indeed, skin lesions from patients with TNF-α inhibitor-induced psoriasis, when compared to those with classical psoriasis, are characterized by increased levels of type I IFNs and pDC.29 Furthermore, treatment with IFN-α has been implicated to induce and exacerbate psoriasis.30
Interleukin-23/T-helper 17 Axis Theory
The role of interleukin (IL)-23/T-helper (TH)-17 axis has also been implicated in the pathogenesis of TNF inhibitor-induced psoriasis.9,31–34 Interleukin-23 is a proinflammatory cytokine that promotes TH-17 activation that has been implicated in the pathogenesis of chronic inflammatory diseases such as RA, IBD, and psoriatic disease.35,36 Interestingly, patients with Crohn disease with TNF inhibitor-induced psoriasis were more likely to be homozygous for polymorphisms in the IL-23 receptor gene compared to disease-matched controls.32,37 Histological analysis of anti-TNF-induced psoriasis lesions showed an increased number of IFN-y-secreting Th-1 and IL-17-/IL-22- secreting Th17 cells, similar to genuine psoriasis. Accordingly, Tillack et al demonstrated for the first time the efficacy of ustekinumab, an IL-12/23 antagonist, for paradoxical anti-TNF psoriasis. Furthermore, they showed that patients with severe forms of anti-TNF-induced psoriasis requiring ustekinumab presented a higher number of IL-17A-expressing T cells compared to patients not requiring ustekinumab.32 In short, the emerging evidence for the role of IL-23/TH-17 axis has suggested that this is a new potential target for therapy.
Permissive Infections
Infections are a known trigger for psoriatic lesions.38 Patients receiving anti-TNF-α therapy have increased risk for infection.39,40 Accordingly, infection may play a role in the manifestation of TNF inhibitor-induced psoriasis. It has been suggested that exposure to infectious organisms may induce keratoderma blennorhagicum, histologically indistinguishable from pustular psoriasis, which may be the lesions described as TNF inhibitor-induced psoriasis.41 These palmoplantar psoriasiform lesions have been linked to chlamydial infection, for which TNF-α has been implicated in controlling and limiting.42
Already at Risk for Psoriasis
It is well established that patients with chronic rheumatologic and gastrointestinal inflammatory disease such as IBD have higher incidence of psoriasis.43,44 The prevalence of IBD in patients with psoriasis is estimated to be around 1.4%.45 In contrast, the prevalence of psoriasis in patients with IBD is approximately 9.6% of patients with Crohn disease and 5.7% of patients with ulcerative colitis.46,47 Indeed, in a retrospective chart review of patients with IBD, those with TNF-α inhibitor-induced psoriasiform eruptions had a greater genetic predisposition for psoriasis compared to patients who did not develop psoriasis while receiving TNF-α inhibitor therapy.48 Thus, patients who have a history of these diseases may be predisposed to TNF inhibitor-induced psoriasis.
Treatment of Anti-TNF-Induced Psoriasis
The treatment of TNF-inhibitor-induced psoriasis is challenging. The decision to continue, suspend, or replace the anti-TNF therapy has to be carefully discussed with the patient considering the severity of the skin eruption, the needs of the underlying disease, and the availability of other treatment options. Of note, the severity of skin eruption should be defined considering the extent of the psoriatic eruption (eg, body surface area) as well as its impact on the quality of life of the patient (eg, involvement of palms, soles, face, genitals, and psychological impact) as recommended by the National Psoriasis Foundation.49 Thus, in this review, we will define mild psoriatic eruption as those cases in which both the extension of the disease and the impact on quality of life are considered tolerable by the patient, and moderate to severe otherwise. Here, we propose an algorithm illustrating different options for the treatment of TNF inhibitor-induced psoriasis in Figure 1.
Figure 1.
Upon the onset of anti-TNF-induced psoriasiform skin disease, the severity of the skin eruption and the efficacy of the anti-TNF drug on the underlying primary disease (RA, IBD, psoriasis, and PsA). For those patients with mild psoriasiform disease and stable underlying disease, we propose a “treat through” strategy in which the anti-TNF therapy will be continued and conventional psoriasis-specific therapy will be added (eg, topical steroids, UV therapy, methotrexate, cyclosporine, and acitretin). If the skin eruption is recalcitrant or worsens, or if the primary condition is poorly controlled, we propose switching to another anti-TNF or to another class. For patients with moderate to severe skin eruption and stable underlying disease, we propose switching to a different anti-TNF and treat the psoriasis symptoms concomitantly. If this fails, a different class should be considered. Instead, for those cases in which the underlying disease is not well controlled with an anti-TNF, we suggest switching to another class. *Controlled underlying disease: rheumatoid arthritis, inflammatory bowel disease, psoriasis, or psoriatic arthritis. **Consider also cases in which anti-TNF therapy is required or favored as in uveitis. IBD indicates inflammatory bowel disease; PsA, psoriatic arthritis; RA, rheumatoid arthritis; TNF, tumor necrosis factor.
Mild Skin Eruption
Controlled underlying disease.
In the setting of mild TNF inhibitor-induced psoriasis and when a patient’s underlying disease state is otherwise well managed on current anti-TNF therapy, a “treat-through” approach with typical therapies for psoriasis may be the most prudent. The psoriasis therapies to be considered include topical steroids, UV therapy, methotrexate, cyclosporine (although not favored long-term therapy option), or acitretin, as appropriate. Notably, we consider cyclosporine with some caution in combination with other biologics, as this therapy is typically used only for short-term bridge therapy when necessary and not as a long-term agent. Indeed, adverse events with administration of concomitant anti-TNF agents and cyclosporine in patients with severe IBD, most notably infections and at least 2 deaths,50,51 have been reported in the literature.
This “treat through” strategy has demonstrated complete resolution in 26% to 41% of the cases and a partial response in 25% to 57.4% (Table 1).7–10 Historically, “treating through” TNF inhibitor-induced psoriasis has been important for patients with IBD given the evidence that discontinuation of biological therapy can aggravate gastrointestinal symptoms and the limited therapeutic class options.52
Table 1.
Summary of Clinical Outcomes per Treatment Modality for Anti-TNF-Induced Psoriasis Across Multiple Underlying Diseases Reported in the Literature.
| Ko et al, 20096 (N = 127) | Collamer et al,8 2008 (N= 114) | Collamer et al,9 2010 (n = 207) | Brown et al,7 2017 (N =216) | Schmidt etal,10 2012 (N = 56) | |
|---|---|---|---|---|---|
| Continued on TNF therapy | N =82 | N =22 | |||
| Resolved, n (%) | 31 (30) | 53 (26) | 27 (32.9) | 9(41) | |
| Improved, n (%) | 32(31) | 52 (25) | 47 (57.4) | 8(36) | |
| No improvement, n (%) | 2(2) | 2(1) | 8 (9.8) | 5(23) | |
| Partial or no resolution combined | |||||
| Discontinued off TNF therapy | N =65 | N =21 | |||
| Resolved, n (%) | 14 (13%) | 50 (24) | 31 (47.7) | 12(57) | |
| Improved, n (%) | 30 (46.2) | 4(19) | |||
| No improvement, n (%) | 4 (6.2) | 1 (5) | |||
| Partial or no resolution combined | 4(4) | 11 (5) | |||
| Switched to different TNF agent | N = 13 | N =49 | |||
| Resolved/no recurrence at follow-up, n (%) | 2(15.4) | 5(5) | 13(6) | 18 (36.7) | |
| Improved, n (%) | 9 (18.4) | ||||
| No improvement, n (%) | 22 (44.9) | ||||
| Resolved off anti-TNF and recurred with a different anti-TNF | 8(8) | 12(6) |
Abbreviation: TNF, tumor necrosis factor.
Pustular psoriasis, which is one of the most frequently observed clinical presentation of anti-TNF induced psoriasis, can be treated with dapsone or acitretin. Dapsone is an “anti-neutrophil” agent that has been shown to improve pustular psoriasis in patients who had previously failed several topical and systemic therapies.53 Of note, dapsone has fewer side effects than other systemic therapies.54 Furthermore, topical dapsone 5% gel has a safe side effect profile given its low systemic absorption.55 In addition to dapsone, clinicians can also consider acitretin, an oral retinoid used as the first-line therapy to treat generalized pustular psoriasis.56 Indeed, there has been one case report of ustekinumab-induced pustular psoriasis that resolved after acitretin treatment, with no recurrence of these lesions at the patient’s 6-month follow-up.57
Uncontrolled underlying disease.
When patients present with mild TNF inhibitor-induced psoriasis and the underlying disease state is not well controlled, we propose switching to a different anti-TNF agent while maintaining psoriasis-specific therapy. However, the rate of success of this approach is limited since complete resolution was only observed in 5% to 36.7% of cases, with partial response in 18.4% (Table 1).6–9
Moderate to Severe Skin Eruption
Controlled underlying disease.
When patients have moderate to severe skin eruption and their underlying disease is well controlled, switching to a different anti-TNF agent can be considered. As cited before, complete regression of psoriasis lesion was seen in only 5% to 36.7% of cases (Table 1).6–9
Uncontrolled underlying disease and refractory cases.
For patients with moderate to severe anti-TNF-induced psoriasis whose underlying disease is active despite anti-TNF-therapy and refractory cases, agents outside the anti-TNF class should be considered, combined with topical or systemic therapy for psoriasis as appropriate. Indeed, it has been reported that in up to approximately 64.3% of cases, patients who discontinue TNF inhibitors and switch to therapies outside the TNF class experience resolution of their psoriasiform lesions.6 Below, we discuss different alternative therapies for RA, IBD, psoriasis, and PsA. An overview of these agents is presented in Table 2.
Table 2.
Summary of Therapy Options for Patients With RA, IBD, Psoriasis, and PsA With TNF Inhibitor-Induced Psoriasis.
| Drug/Brand Name | Mechanism of Action | FDA-Approved Indication(s) of Relevance | Special Considerations |
|---|---|---|---|
| RA | |||
| Tocilizumab/Actemra | Anti-IL-6 monoclonal antibody |
|
Tocilizumab has demonstrated to be a viable treatment option for patients with RA who have failed TNF-a inhibitor therapy.58 Of note, tocilizumab may be more effective when combined with methotrexate than when used as monotherapy.59‘60 |
| Rituximab/Rituxan | Anti-CD20 monoclonal antibody |
|
Patients with RA who previously failed TNF inhibitor therapy who switched to rituximab saw significant improvement in their RA compared to those who switched therapy to a second TNF inhibitor.61 This effect was particularly striking in seropositive patients. Other studies have demonstrated similar results.62‘63 |
| Abatacept/Orencia | Selective T-cell costimulation blocker |
|
Abatacept is a viable treatment option for patients with moderate to severe RA who failed one or more TNF-a inhibitor therapies or disease-modifying antirheumatic drugs (DMARDs). Patients with active RA who failed prior TNF-a inhibitor therapy showed significantly more improvement in physical function when treated with abatacept for 6 months.64 Notably, clinicians should discontinue etanercept treatment before treating with abatacept, as an increase in serious adverse events was observed when these 2 therapies were used in combination.65 |
| Tofacitinib/Xeljanz | Janus-associated kinase QAK) inhibitor |
|
Tofacitinib is a treatment option for patients with RA and demonstrated to have similar efficiency to adalimumab in treating RA.66 Additionally, tofacitinib used in combination with methotrexate improved RA in patients who failed prior TNF-a inhibitor therapy.67‘68 |
| IBD | |||
| Ustekinumab/Stelara | Anti-IL-12 and anti-IL-23 monoclonal antibody |
|
Patients with Crohn disease who have failed anti-TNF-a agents have reported resolution of psoriatic lesions after treatment with ustekinumab.69‘70 Moreover, in a cohort of anti-TNF resistant patients with Crohn disease, 73.7% of patients responded to subcutaneous ustekinumab.71 Ustekinumab therapy has also significantly improved TNF inhibitor-induced palmoplantar pustulosis in a patient with a history of Crohn disease.72 |
| Vedolizumab/Entyvio | a4|37 Integrin antagonist monoclonal antibody |
|
Vedolizumab is a treatment option for patients who failed prior conventional treatment of TNF-a inhibitor therapy.73‘74 Of note, patients with IBD who primarily received vedolizumab had higher rates of response and remission than those who were treated with anti-TNF-a agents in the past, though previous treatment to anti-TNF-a agents does not significantly affect vedolizumab efficacy.75‘76 |
| Psoriasis/PsA | |||
| Secukinumab/Cosentyx | Anti-IL-I7A monoclonal antibody |
|
Secukinumab is a treatment option for moderate to severe plaque psoriasis and PsA that can effectively treat patients with PsA who failed prior TNF-a therapy.77,78 In a recent systematic review comparing the effectiveness of several treatments for PsA, secukinumab was found to be the most effective in treating PsA in both the anti-TNF-a-naive and anti-TNF-a-experienced cohorts.79 Furthermore, patients with PsA who have previously been treated with ustekinumab seem to have a greater response to secukinumab.80 |
| Ixekizumab/Taltz | Anti-IL-17 A monoclonal antibody |
|
Ixekizumab is a treatment option for adult moderate to severe psoriasis. There are limited data regarding the efficiency of ixekizumab in treating patients with psoriatic disease who failed TNF-a inhibitors, 1 study demonstrated improved signs and symptoms after 24 weeks of treatment with ixekizumab in patients with PsA who failed prior anti-TNF-a therapy.81 Of note, in the UNCOVER trials, patients with moderate to severe plaque psoriasis exhibited greater responses to ixekizumab therapy compared to etanercept and placebo.82 |
| Apremilast/Otezla | Phosphodiesterase-4-inhibitor |
|
Apremilast is a treatment option for moderate to severe plaque psoriasis and PsA. Although there are limited data on the efficiency of apremilast in treating psoriasis or PsA in patients who failed TNF-a inhibitors, apremilast has been demonstrated to be effective and safe in treating patients with active PsA who were treated with DMARDs or biologies in the past.83 The safety and efficacy of switching from etanercept to apremilast in treating psoriasis is supported in the LIBERATE study.84 |
| Ustekinumab/Stelara | Anti-IL-12 and anti-IL-23 monoclonal antibody |
|
Ustekinumab is a treatment option for moderate to severe psoriasis and PsA, which can efficiently treat TNF inhibitor-induced psoriasis.85 Two case reports saw resolution of psoriasiform eruptions secondary to adalimumab in females with PsA treated with ustekinumab every 3 months.86 Furthermore, ustekinumab has also been implicated in successfully treating 4 cases of recalcitrant palmoplantar pustulosis, one of which was anti-TNF inhibitor induced.87 Interestingly, there has been 1 case report of a 30-year-old male who previously failed infliximab therapy whose psoriasis was exacerbated after treatment with ustekinumab.88 |
| Abatacept/Orencia | Selective T-cell costimulation blocker |
|
Abatacept is a treatment option for PsA used where other drugs have failed. Several studies support the efficacy of abatacept in treating patients with PsA who have failed prior anti-TNF therapy, particularly in improving musculoskeletal manifestations of PsA.89‘90 |
| Tofacitinib/Xeljanz | Janus-associated kinase QAK) inhibitor |
|
Tofacitinib is a treatment option for patients with PsA that has been shown to reduce disease activity in patients with PsA who failed prior TNF inhibitor therapy.91 Additionally, tofacitinib has also been shown to effectively treat patients with PsA with previous inadequate response to conventional synthetic DMARDs.92 Notably, tofacitinib is not FDA approved to treat psoriasis. |
| Guselkumab/Tremfya | Anti-IL-23 monoclonal antibody |
|
Guselkumab is a treatment option for moderate to severe psoriasis that has demonstrated superior efficacy compared to adalimumab for treating psoriasis.93‘94 Importantly, guselkumab has been shown to be effective in treating patients with psoriasis who had failed prior anti-TNF therapy.95 |
Abbreviations: FDA, Food and Drug Administration; IBD, inflammatory bowel disease; IL, interleukin; PsA, psoriatic arthritis; RA, rheumatoid arthritis; TNF, tumor necrosis factor.
Rheumatoid Arthritis
Non-anti-TNF biologic agents have been demonstrated to be, in some cases, as or more effective treatment compared to a second anti-TNF drug for patients with RA with insufficient primary response to anti-TNF-α inhibitor, suggesting the need for a different mechanism of action.96 Indeed, tocilizumab, rituximab, abatacept, and tofacitinib have been significantly more effective in treating RA when compared to placebo in these patients.97 Importantly, abatacept and tofacitinib are an FDA-approved drugs for PsA.
Inflammatory Bowel Disease
Although TNF-α inhibitors have improved clinical outcomes for patients with IBD, many patients fail to attain remission with anti-TNF-α therapy.98 For this patient population that have failed TNF-inhibitor therapy, we recommend ustekinumab specifically in the setting of Crohn disease or vedolizumab as viable treatment options for patients with IBD. Notably, both ustekinumab and vedolizumab seem to have the same response and remission rate in patients with Crohn disease who failed prior TNF-α inhibitor therapy.99 Of note, substantial evidence supports the long-term efficacy and safety of ustekinumab in the treatment of psoriasis in both clinical trials and clinical settings.100 Accordingly, Guerra et al reported 8 cases of TNF-induced psoriasis cases treated with ustekinumab, of which 75% experienced complete resolution.23 Furthermore, Pugliese et al achieved complete clearance of skin lesions in all 3 cases treated with ustekinumab.101
Additionally, we also recommend discussions with IBD-treating physician or gastroenterology regarding traditional IBD treatments such as immunosuppressive agents including 6-mercaptopurine and azathioprine versus newer biologic therapies.
Psoriasis and PsA
Several treatment options are available for patients with psoriasis and PsA who failed TNF-α inhibitor therapy. Guselkumab is a viable treatment option for patients with psoriasis, while ustekinumab, ixekizumab, secukinumab, and apremilast have demonstrated efficiency in treating patients with both psoriasis and PsA. Finally, tofacitinib and abatacept are viable treatment options for patients with PsA only.
Discussion
Tumor necrosis factor inhibitor-induced psoriasis is not uncommon. This cutaneous reaction can manifest in several forms including plaque psoriasis, palmoplantar pustular psoriasis, psoriasiform, guttate, and inverse psoriasis.11 Growing evidence supports the notion that paradoxical psoriasis is a class effect.6,7 The increasing incidence of TNF-α induced psoriasis in a crowded landscape of therapeutic options supports the need for algorithmic guidance to treat TNF inhibitor-induced psoriasis. Here, we propose an algorithm from a dermatologic standpoint in which the severity of the skin eruption and the characteristics of the underlying disease guide the medical decision. In contrast, previously published algorithms in the rheumatologic literature founded their treatment choices exclusively on the severity of skin disease.8 Taken together, our algorithm outlines specific therapies for each underlying disease, including the most recently approved agents by the FDA.
Our algorithm proposes that for those cases with mild induced psoriasis in patients whose underlying disease is well controlled, a “treat through” approach with traditional therapies for psoriasis is the preferred option. Conversely, for those patients with mild skin eruption and uncontrolled underlying disease or patients with moderate-to-severe induced psoriasis with good control of underlying disease, temporal withdrawal of anti-TNF therapy or switching to other anti-TNF agents could be considered. However, it is important to acknowledge that more than half of the patients may experience recurrence of skin lesions after the reintroduction of either the same or a different anti-TNF.23,102,103 Thus, for refractory cases of induced psoriasis as well as for patients with moderate to severe skin eruption whose underlying disease is not well controlled, our algorithm emphasizes switching the drug class to increase the likelihood of resolution of the paradoxical psoriasis.
Understanding the inflammatory pathways involved in the onset of this adverse event and that of the underlying disease is of paramount importance to select the most appropriate treatment for patients who have failed or are otherwise intolerant to anti-TNF-α agents. Indeed, the identification of the role of the IL-23/Th-17 axis in paradoxical psoriasis has become critical in the management of these cases. For example, ustekinumab is an anti-IL12/23 agent that had led to resolution of skin lesions in more than 75% of the cases in patients with IBD.32,104,105 Likewise, the efficacy of anti-IL17 agents in genuine psoriasis and PsA suggests anti-IL17 agents as another promising class of agents for paradoxical psoriasis. Of note, anti-IL17 agents should be used with caution in patients with both psoriasis and IBD, since there is evidence that they may exacerbate or induce IBD in patients with psoriatic disease.106
Despite the advances in the pathogenesis of paradoxical psoriasis, significant gaps remain as this cutaneous reaction has also been described for some of the alternative therapies described in this review. Indeed, paradoxical psoriasis and PsA have been described in patients treated with ustekinumab.105 Then rituximab has been associated with de novo and flare of existing psoriasis in patients with seronegative RA and systemic lupus erythematosus.107,108 Similarly, new onset of psoriasis and exacerbation of previous psoriasis were observed in a few patients with RA and PsA treated with abatacept.109,110 Finally, 2 cases of de novo psoriasis were described in patients with RA treated with tocilizumab.111,112
Other indications of anti-TNF agents include uveitis, ankylosing spondylitis, and juvenile RA. As paradoxical psoriasis in these conditions has been less frequently reported compared to RA, psoriatic disease, and IBD, we did not include them in our discussion.
Conclusion
Tumor necrosis factor inhibitor-induced psoriasis is not uncommon and has been observed to be a class effect. Here, we propose a novel algorithm that incorporates new treatment options for patients with RA, IBD, psoriasis, and PsA with TNF inhibitor-induced psoriasis and highlights the strategy to switch therapy outside the TNF class when clinically appropriate. Because paradoxical psoriasis has also been described for some of these classes of drugs, further studies are required to better characterize the safety and efficacy of this approach. Thus, we recommend discussing these treatment options in a case-by-case basis and incorporating multidisciplinary care physicians for treating the underlying disease.
Funding
The author(s) received no financial support for the research, authorship, and/or publication of this article.
Footnotes
Declaration of Conflicting Interests
The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: Joseph F. Merola is a consultant and/or investigator for Biogen IDEC, AbbVie, Amgen, Eli Lilly, Novartis, Pfizer, Janssen, UCB, Samumed, Science 37, Celgene, Sanofi Regeneron, Merck, and GSK.
References
- 1.Campa M, Ryan C, Menter A. An overview of developing TNF-alpha targeted therapy for the treatment of psoriasis. Expert Opin Investig Drugs. 2015;24(10):1343–1354. [DOI] [PubMed] [Google Scholar]
- 2.Feldman M, Taylor P, Paleolog E, Brennan FM, Maini RN. Anti-TNF alpha therapy is useful in rheumatoid arthritis and Crohn’s disease: analysis of the mechanism of action predicts utility in other diseases. Transplant Proc. 1998;30(8):4126–4127. [DOI] [PubMed] [Google Scholar]
- 3.Feldman SR, Gordon KB, Bala M, et al. Infliximab treatment results in significant improvement in the quality of life of patients with severe psoriasis: a double-blind placebo-controlled trial. Br J Dermatol. 2005;152(5):954–960. [DOI] [PubMed] [Google Scholar]
- 4.Strand V, Sharp V, Koenig AS, et al. Comparison of healthrelated quality of life in rheumatoid arthritis, psoriatic arthritis and psoriasis and effects of etanercept treatment. Ann Rheum Dis. 2012;71(7):1143–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Holdam AS, Bager P, Dahlerup JF. Biological therapy increases the health-related quality of life in patients with inflammatory bowel disease in a clinical setting. Scand J Gastroenterol. 2016; 51(6):706–711. [DOI] [PubMed] [Google Scholar]
- 6.Ko JM, Gottlieb AB, Kerbleski JF. Induction and exacerbation of psoriasis with TNF-blockade therapy: a review and analysis of 127 cases. J Dermatolog Treat. 2009;20(2):100–108. [DOI] [PubMed] [Google Scholar]
- 7.Brown G, Wang E, Leon A, et al. Tumor necrosis factor-alpha inhibitor-induced psoriasis: systematic review of clinical features, histopathological findings, and management experience. J Am Acad Dermatol. 2017;76(2):334–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Collamer AN, Guerrero KT, Henning JS, Battafarano DF. Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy: a literature review and potential mechanisms of action. Arthritis Rheum. 2008;59(7):996–1001. [DOI] [PubMed] [Google Scholar]
- 9.Collamer AN, Battafarano DF. Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy: clinical features and possible immunopathogenesis. Semin Arthritis Rheum. 2010; 40(3):233–240. [DOI] [PubMed] [Google Scholar]
- 10.Shmidt E, Wetter DA, Ferguson SB, Pittelkow MR. Psoriasis and palmoplantar pustulosis associated with tumor necrosis factoralpha inhibitors: the Mayo Clinic experience, 1998 to 2010. J Am Acad Dermatol. 2012;67(5):e179–e185. [DOI] [PubMed] [Google Scholar]
- 11.Joyau C, Veyrac G, Dixneuf V, Jolliet P. Anti-tumour necrosis factor alpha therapy and increased risk of de novo psoriasis: is it really a paradoxical side effect? Clinical and experimental rheumatology. 2012;30(5):700–706. [PubMed] [Google Scholar]
- 12.Laga AC, Vleugels RA, Qureshi AA, Velazquez EF. Histopathologic spectrum of psoriasiform skin reactions associated with tumor necrosis factor-alpha inhibitor therapy. A study of 16 biopsies. Am J Dermatopathol. 2010;32(6):568–573. [DOI] [PubMed] [Google Scholar]
- 13.Seneschal J, Milpied B, Vergier B, Lepreux S, Schaeverbeke T, Taieb A. Cytokine imbalance with increased production of interferon-alpha in psoriasiform eruptions associated with antitumour necrosis factor-alpha treatments. Br J Dermatol. 2009; 161(5):1081–1088. [DOI] [PubMed] [Google Scholar]
- 14.Nguyen K, Vleugels RA, Velez NF, Merola JF, Qureshi AA. Psoriasiform reactions to anti-tumor necrosis factor alpha therapy. J Clin Rheumatol. 2013;19(7):377–381. [DOI] [PubMed] [Google Scholar]
- 15.Flendrie M, Vissers WH, Creemers MC, de Jong EM, van de Kerkhof PC, van Riel PL. Dermatological conditions during TNF-alpha-blocking therapy in patients with rheumatoid arthritis: a prospective study. Arthritis Res Ther. 2005;7(3):R666–R676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sfikakis PP, Iliopoulos A, Elezoglou A, Kittas C, Stratigos A. Psoriasis induced by anti-tumor necrosis factor therapy: a paradoxical adverse reaction. Arthritis Rheum. 2005;52(8): 2513–2518. [DOI] [PubMed] [Google Scholar]
- 17.Harrison MJ, Dixon WG, Watson KD, et al. Rates of new-onset psoriasis in patients with rheumatoid arthritis receiving antitumour necrosis factor alpha therapy: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis. 2009;68(2):209–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hernandez MV, Sanmarti R, Canete JD, et al. Cutaneous adverse events during treatment of chronic inflammatory rheumatic conditions with tumor necrosis factor antagonists: study using the Spanish registry of adverse events of biological therapies in rheumatic diseases. Arthritis Care Res. 2013;65(12):2024–2031. [DOI] [PubMed] [Google Scholar]
- 19.Koizumi H, Tokuriki A, Oyama N, et al. Certolizumab pegol, a pegylated anti-TNF-αlpha antagonist, caused de novo-onset palmoplantar pustulosis followed by generalized pustular psoriasis in a patient with rheumatoid arthritis. J Dermatol. 2017;44(6): 723–724. [DOI] [PubMed] [Google Scholar]
- 20.Mateo S, Garcia-Martinez FJ, Sanchez-Aguilar D, Amarelo J, Toribio J. Psoriasiform exfoliative erythroderma induced by golimumab. Clin Exp Dermatol. 2014;39(7):813–815. [DOI] [PubMed] [Google Scholar]
- 21.Afzali A, Wheat CL, Hu JK, Olerud JE, Lee SD. The association of psoriasiform rash with anti-tumor necrosis factor (anti-TNF) therapy in inflammatory bowel disease: a single academic center case series. J Crohns Colitis. 2014;8(6):480–488. [DOI] [PubMed] [Google Scholar]
- 22.Guerra I, Algaba A, Perez-Calle JL, et al. Induction of psoriasis with anti-TNF agents in patients with inflammatory bowel disease: a report of 21 cases. J Crohns Colitis. 2012;6(5):518–523. [DOI] [PubMed] [Google Scholar]
- 23.Guerra I, Pérez-Jeldres T, Iborra M, et al. Incidence, clinical characteristics, and management of psoriasis induced by Anti-TNF therapy in patients with inflammatory bowel disease: a nationwide cohort study. Inflamm Bowel Dis. 2016;22(4): 894–901. [DOI] [PubMed] [Google Scholar]
- 24.Eickstaedt JB, Killpack L, Tung J, Davis D, Hand JL, Tollefson MM. Psoriasis and Psoriasiform eruptions in pediatric patients with inflammatory bowel disease treated with anti-tumor necrosis factor alpha agents. Pediatr Dermatol. 2017;34(3):253–260. [DOI] [PubMed] [Google Scholar]
- 25.Gottlieb AB, Evans R, Li S, et al. Infliximab induction therapy for patients with severe plaque-type psoriasis: a randomized, double-blind, placebo-controlled trial. J Am Acad Dermatol. 2004;51(4): 534–542. [DOI] [PubMed] [Google Scholar]
- 26.Napolitano M, Balato N, Caso F, et al. Paradoxical onset of psoriatic arthritis during treatment with biologic agents for plaque psoriasis: a combined dermatology and rheumatology clinical study. Clin Exp Rheumatol. 2017;35(1):137–140. [PubMed] [Google Scholar]
- 27.Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med. 2005;202(1):135–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Conrad C, Boyman O, Tonel G, et al. Alpha1beta1 integrin is crucial for accumulation of epidermal T cells and the development of psoriasis. Nat Med. 2007;13(7):836–842. [DOI] [PubMed] [Google Scholar]
- 29.Conrad C, Di Domizio J, Mylonas A, et al. TNF blockade induces a dysregulated type I interferon response without autoimmunity in paradoxical psoriasis. Nat Commun. 2018;9(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ladoyanni E, Nambi R. Psoriasis exacerbated by interferon-alpha in a patient with chronic myeloid leukemia. J Drugs Dermatol. 2005;4(2):221–222. [PubMed] [Google Scholar]
- 31.Ma HL, Napierata L, Stedman N, et al. Tumor necrosis factor alpha blockade exacerbates murine psoriasis-like disease by enhancing Th17 function and decreasing expansion of Treg cells. Arthritis Rheum. 2010;62(2):430–440. [DOI] [PubMed] [Google Scholar]
- 32.Tillack C, Ehmann LM, Friedrich M, et al. Anti-TNF antibodyinduced psoriasiform skin lesions in patients with inflammatory bowel disease are characterised by interferon-gamma-expressing Th1 cells and IL-17A/IL-22-expressing Th17 cells and respond to anti-IL-12/IL-23 antibody treatment. Gut. 2014;63(4):567–577. [DOI] [PubMed] [Google Scholar]
- 33.Chan JR, Blumenschein W, Murphy E, et al. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med. 2006;203(12):2577–2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Lee E, Trepicchio WL, Oestreicher JL, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med. 2004;199(1):125–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tabarkiewicz J, Pogoda K, Karczmarczyk A, Pozarowski P, Giannopoulos K. The role of IL-17 and Th17 lymphocytes in autoimmune diseases. Arch Immunol Ther Exp. 2015;63(6):435–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Duffin KC, Krueger GG. Genetic variations in cytokines and cytokine receptors associated with psoriasis found by genomewide association. J Invest Dermatol. 2009;129(4):827–833. [DOI] [PubMed] [Google Scholar]
- 37.Sherlock ME, Walters T, Tabbers MM, et al. Infliximab-induced psoriasis and psoriasiform skin lesions in pediatric Crohn disease and a potential association with IL-23 receptor polymorphisms. J Pediatr Gastroenterol Nutr. 2013;56(5):512–518. [DOI] [PubMed] [Google Scholar]
- 38.Fry L, Baker BS. Triggering psoriasis: the role of infections and medications. Clin Dermatol. 2007;25(6):606–615. [DOI] [PubMed] [Google Scholar]
- 39.Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 2006;295(19):2275–2285. [DOI] [PubMed] [Google Scholar]
- 40.Dommasch ED, Abuabara K, Shin DB, Nguyen J, Troxel AB, Gelfand JM. The risk of infection and malignancy with tumor necrosis factor antagonists in adults with psoriatic disease: a systematic review and meta-analysis of randomized controlled trials. J Am Acad Dermatol. 2011;64(6):1035–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Carter JD. Tumour necrosis factor inhibition causing psoriasis? A more plausible explanation exists. Ann Rheum Dis. 2006;65(12): 1680–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Inman RD, Chiu B. Early cytokine profiles in the joint define pathogen clearance and severity of arthritis in Chlamydia-induced arthritis in rats. Arthritis Rheum. 2006;54(2):499–507. [DOI] [PubMed] [Google Scholar]
- 43.Najarian DJ, Gottlieb AB. Connections between psoriasis and Crohn’s disease. J Am Acad Dermatol. 2003;48(6):805–821; quiz 822–804. [DOI] [PubMed] [Google Scholar]
- 44.Kimball AB, Gladman D, Gelfand JM, et al. National Psoriasis Foundation clinical consensus on psoriasis co-morbidities and recommendations for screening. J Am Acad Dermatol. 2008; 58(6):1031–1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Makredes M, Robinson D Jr., Bala M, Kimball AB. The burden of autoimmune disease: a comparison of prevalence ratios in patients with psoriatic arthritis and psoriasis. J Am Acad Dermatol. 2009; 61(3):405–410. [DOI] [PubMed] [Google Scholar]
- 46.Lee FI, Bellary SV, Francis C. Increased occurrence of psoriasis in patients with Crohn’s disease and their relatives. Am J Gastro enterol. 1990;85(8):962–963. [PubMed] [Google Scholar]
- 47.Yates VM, Watkinson G, Kelman A. Further evidence for an association between psoriasis, Crohn’s disease and ulcerative colitis. Br J Dermatol. 1982;106(3):323–330. [DOI] [PubMed] [Google Scholar]
- 48.Vedak P, Kroshinsky D, St. John J, Xavier RJ, Yajnik V, Ananthakrishnan AN. Genetic basis of TNF-α antagonist associated psoriasis in inflammatory bowel diseases: a genotype-phenotype analysis. Aliment Pharmacol Ther. 2016;43(6):697–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pariser DM, Bagel J, Gelfand JM, et al. National Psoriasis Foundation clinical consensus on disease severity. Arch Dermatol. 2007;143(2):239–242. [DOI] [PubMed] [Google Scholar]
- 50.Maser EA, Deconda D, Lichtiger S, Ullman T, Present DH, Kornbluth A. Cyclosporine and infliximab as rescue therapy for each other in patients with steroid-refractory ulcerative colitis. Clin Gastroenterol Hepatol. 2008;6(10):1112–1116. [DOI] [PubMed] [Google Scholar]
- 51.Leblanc S, Allez M, Seksik P, et al. Successive treatment with cyclosporine and infliximab in steroid-refractory ulcerative colitis. Am J Gastroenterol. 2011;106(4):771–777. [DOI] [PubMed] [Google Scholar]
- 52.Denadai R, Teixeira FV, Steinwurz F, Romiti R, Saad-Hossne R. Induction or exacerbation of psoriatic lesions during anti-TNF-alpha therapy for inflammatory bowel disease: a systematic literature review based on 222 cases. J Crohns Colitis. 2013;7(7): 517–524. [DOI] [PubMed] [Google Scholar]
- 53.Sheu JS, Divito SJ, Enamandram M, Merola JF. Dapsone therapy for pustular psoriasis: case series and review of the literature. Dermatology (Basel, Switzerland). 2016;232(1):97–101. [DOI] [PubMed] [Google Scholar]
- 54.Lorenz M, Wozel G, Schmitt J. Hypersensitivity reactions to dapsone: a systematic review. Acta Derm Venereol. 2012;92(2): 194–199. [DOI] [PubMed] [Google Scholar]
- 55.Stotland M, Shalita AR, Kissling RF. Dapsone 5% gel: a review of its efficacy and safety in the treatment of acne vulgaris. Am J Clin Dermatol. 2009;10(4):221–227. [DOI] [PubMed] [Google Scholar]
- 56.Robinson A, Van Voorhees AS, Hsu S, et al. Treatment of pustular psoriasis: from the Medical Board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67(2):279–288. [DOI] [PubMed] [Google Scholar]
- 57.Dai Y-X, Chen C-C. Flare-up of pustular psoriasis after ustekinumab therapy: case report and literature review [published online ahead of print June 1, 2018] Dermatologica Sinica. 2018. [Google Scholar]
- 58.Baganz L, Richter A, Kekow J, et al. Long-term effectiveness of tocilizumab in patients with rheumatoid arthritis, stratified by number of previous treatment failures with biologic agents: results from the German RABBIT cohort. Rheumatol Int. 2018. 38(4):579–587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Emery P, Keystone E, Tony HP, et al. IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebocontrolled trial. Ann Rheum Dis. 2008;67(11):1516–1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wakabayashi H, Hasegawa M, Nishioka Y, Minami Y, Nishioka K, Sudo A. Clinical outcome in patients with rheumatoid arthritis switched to tocilizumab after etanercept or infliximab failure. Clin Rheumatol. 2013;32(2):253–259. [DOI] [PubMed] [Google Scholar]
- 61.Emery P, Gottenberg JE, Rubbert-Roth A, et al. Rituximab versus an alternative TNF inhibitor in patients with rheumatoid arthritis who failed to respond to a single previous TNF inhibitor: SWITCH-RA, a global, observational, comparative effectiveness study. Ann Rheum Dis. 2015;74(6):979–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Soliman MM, Hyrich KL, Lunt M, et al. Rituximab or a second anti-tumor necrosis factor therapy for rheumatoid arthritis patients who have failed their first anti-tumor necrosis factor therapy? Comparative analysis from the British Society for Rheumatology Biologics Register. Arthritis Care Res (Hoboken). 2012;64(8):1108–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gomez-Reino JJ, Maneiro JR, Ruiz J, et al. Comparative effectiveness of switching to alternative tumour necrosis factor (TNF) antagonists versus switching to rituximab in patients with rheumatoid arthritis who failed previous TNF antagonists: the MIRAR Study. Ann Rheum Dis. 2012;71(11):1861–1864. [DOI] [PubMed] [Google Scholar]
- 64.Genovese MC, Becker JC, Schiff M, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med. 2005;353(11): 1114–1123. [DOI] [PubMed] [Google Scholar]
- 65.Weinblatt M, Schiff M, Goldman A, et al. Selective costimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomised clinical trial. Ann Rheum Dis. 2007;66(2):228–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367(6):508–519. [DOI] [PubMed] [Google Scholar]
- 67.Burmester GR, Blanco R, Charles-Schoeman C, et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet. 2013;381(9865):451–460. [DOI] [PubMed] [Google Scholar]
- 68.Singh JA, Hossain A, Tanjong Ghogomu E, et al. Biologics or tofacitinib for people with rheumatoid arthritis unsuccessfully treated with biologics: a systematic review and network metaanalysis. Cochrane Database Syst Rev. 2017;3:CD012591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sanso Sureda A, Rocamora Durán V, Sapiña Camaró A, Royo Escosa V, Bosque López MJ. Ustekinumab in a patient with Crohn’s disease and anti-TNF-alpha-induced psoriasis [in Spanish]. Gastroenterol Hepatol. 2011;34(8):546–550. [DOI] [PubMed] [Google Scholar]
- 70.Puig L, Morales-Múnera CE, López-Ferrer A, Geli C. Ustekinumab treatment of TNF antagonist-induced paradoxical psoriasis flare in a patient with psoriatic arthritis: case report and review. Dermatology. 2012;225(1):14–17. [DOI] [PubMed] [Google Scholar]
- 71.Kopylov U, Afif W, Cohen A, et al. Subcutaneous ustekinumab for the treatment of anti-TNF resistant Crohn’s disease—the McGill experience. J Crohns Colitis. 2014;8(11):1516–1522. [DOI] [PubMed] [Google Scholar]
- 72.Chu DH, Van Voorhees AS, Rosenbach M. Treatment of refractory tumor necrosis factor inhibitor-induced palmoplantar pustulosis: a report of 2 cases. Arch Dermatol. 2011;147(10):1228–1230. [DOI] [PubMed] [Google Scholar]
- 73.Feagan BG, Rubin DT, Danese S, et al. Efficacy of vedolizumab induction and maintenance therapy in patients with ulcerative colitis, regardless of prior exposure to tumor necrosis factor antagonists. Clin Gastroenterol Hepatol. 2017;15(2):229–239 e5. [DOI] [PubMed] [Google Scholar]
- 74.Sands BE, Feagan BG, Rutgeerts P. Effects of vedolizumab induction therapy for patients with Crohn’s disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology. 2014;147(3):618–627 e3. [DOI] [PubMed] [Google Scholar]
- 75.Sands BE, Sandborn WJ, Van Assche G, et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease in patients naive to or who have failed tumor necrosis factor antagonist therapy. Inflamm Bowel Dis. 2017;23(1):97–106. [DOI] [PubMed] [Google Scholar]
- 76.Feagan BG, Rutgeerts P, Sands BE. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369(8):699–710. [DOI] [PubMed] [Google Scholar]
- 77.Kavanaugh A, McInnes IB, Mease PJ, et al. Efficacy of subcutaneous secukinumab in patients with active psoriatic arthritis stratified by prior tumor necrosis factor inhibitor use: results from the randomized placebo-controlled FUTURE 2 study. J Rheumatol. 2016;43(9):1713–1717. [DOI] [PubMed] [Google Scholar]
- 78.Pelechas E, Memi T, Voulgari PV, Drosos AA. A case of recalcitrant psoriatic arthritis to TNF inhibitors improved after administration of secukinumab, an IL-17A inhibitor. Rheumatol Ther. 2017;4(2):509–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kawalec P, Holko P, Moćko P, Pilc A. Comparative effectiveness of abatacept, apremilast, secukinumab and ustekinumab treatment of psoriatic arthritis: a systematic review and network meta-analysis. Rheumatol Int. 2018;38(2):189–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Blauvelt A, Reich K, Tsai TF, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-to-severe plaque psoriasis up to 1 year: results from the CLEAR study. J Am Acad Dermatol. 2017;76(1):60–69 e9. [DOI] [PubMed] [Google Scholar]
- 81.Nash P, Kirkham B, Okada M. 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]
- 82.Griffiths CE, Reich K, Lebwohl M, et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet. 2015;386(9993):541–551. [DOI] [PubMed] [Google Scholar]
- 83.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]
- 84.Reich K, Gooderham M, Bewley A, et al. Safety and efficacy of apremilast through 104 weeks in patients with moderate to severe psoriasis who continued on apremilast or switched from etanercept treatment: findings from the LIBERATE study. J Eur Acad Dermatol Venereol. 2018;32(3):397–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ritchlin C, Rahman P, Kavanaugh A, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal antibody, ustekinumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour necrosis factor therapy: 6-month and 1-year results of the phase 3, multicentre, double-blind, placebo-controlled, randomised PSUMMIT 2 trial. Ann Rheum Dis. 2014;73(6):990–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bogaards NA, de Rie MA. Psoriasiform eruption and worsening of pustulosis palmoplantaris after treatment with two Anti-TNF-alpha inhibitors, followed by successful treatment with ustekinumab. Dermatol Ther (Heidelb). 2016;6(4):683–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gerdes S, Franke J, Domm S, Mrowietz U. Ustekinumab in the treatment of palmoplantar pustulosis. Br J Dermatol. 2010; 163(5):1116–1118. [DOI] [PubMed] [Google Scholar]
- 88.Suh HY, Ahn JY, Park MY, Youn JI. Exacerbation of infliximab-induced paradoxical psoriasis after ustekinumab therapy. J Dermatol. 2018;45(3):332–333. [DOI] [PubMed] [Google Scholar]
- 89.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]
- 90.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(4): 939–948. [DOI] [PubMed] [Google Scholar]
- 91.Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–1536. [DOI] [PubMed] [Google Scholar]
- 92.Mease P, Hall S, FitzGerald O, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377(16): 1537–1550. [DOI] [PubMed] [Google Scholar]
- 93.Blauvelt A, Papp KA, Griffiths CE, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial. J Am Acad Dermatol. 2017;76(3):405–417. [DOI] [PubMed] [Google Scholar]
- 94.Reich K, Armstrong AW, Foley P, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: results from the phase III, double-blind, placebo-and active comparator-controlled VOYAGE 2 trial. J Am Acad Dermatol. 2017;76(3):418–431. [DOI] [PubMed] [Google Scholar]
- 95.Megna M, Balato A, Raimondo A, Balato N. Guselkumab for the treatment of psoriasis. Expert Opin Biol Ther. 2018;18(4):459–468. [DOI] [PubMed] [Google Scholar]
- 96.Gottenberg JE, Brocq O, Perdriger A, et al. Non-TNF-targeted biologic vs a second anti-TNF drug to treat rheumatoid arthritis in patients with insufficient response to a first anti-TNF drug: a randomized clinical trial. JAMA. 2016;316(11):1172–1180. [DOI] [PubMed] [Google Scholar]
- 97.Wei W, Knapp K, Wang L, et al. Treatment persistence and clinical outcomes of tumor necrosis factor inhibitor cycling or switching to a new mechanism of action therapy: real-world observational study of rheumatoid arthritis patients in the united states with prior tumor necrosis factor inhibitor therapy. Adv Ther. 2017;34(8):1936–1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Rutgeerts P, Sandborn WJ, Feagan BG, et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005;353(23):2462–2476. [DOI] [PubMed] [Google Scholar]
- 99.Kawalec P, Mocko P. An indirect comparison of ustekinumab and vedolizumab in the therapy of TNF-failure Crohn’s disease patients. J Comp Eff Res. 2018;7(2):101–111. [DOI] [PubMed] [Google Scholar]
- 100.Kimball AB, Papp KA, Wasfi Y, et al. Long-term efficacy of ustekinumab in patients with moderate-to-severe psoriasis treated for up to 5 years in the PHOENIX 1 study. J Eur Acad Dermatol Venereol. 2013;27(12):1535–1545. [DOI] [PubMed] [Google Scholar]
- 101.Pugliese D, Guidi L, Ferraro PM, et al. Paradoxical psoriasis in a large cohort of patients with inflammatory bowel disease receiving treatment with anti-TNF alpha: 5-year follow-up study. Aliment Pharmacol Ther. 2015;42(7):880–888. [DOI] [PubMed] [Google Scholar]
- 102.Feuerstein JD, Cheifetz AS. Miscellaneous adverse events with biologic agents (excludes infection and malignancy). Gastroenterol Clin North Am. 2014;43(3):543–563. [DOI] [PubMed] [Google Scholar]
- 103.Iborra M, Beltran B, Bastida G, Aguas M, Nos P. Infliximab and adalimumab-induced psoriasis in Crohn’s disease: a paradoxical side effect. J Crohns Colitis. 2011;5(2):157–161. [DOI] [PubMed] [Google Scholar]
- 104.Andrisani G, Marzo M, Celleno L, et al. Development of psoriasis scalp with alopecia during treatment of Crohn’s disease with infliximab and rapid response to both diseases to ustekinumab. Eur Rev Med Pharmacol Sci. 2013;17(20): 2831–2836. [PubMed] [Google Scholar]
- 105.Lee HY, Woo CH, Haw S. Paradoxical flare of psoriasis after ustekinumab therapy. Ann Dermatol. 2017;29(6): 794–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Whitlock SM, Enos CW, Armstrong AW, et al. Management of psoriasis in patients with inflammatory bowel disease: from the Medical Board of the National Psoriasis Foundation. J Am Acad Dermatol. 2018;78(2):383–394. [DOI] [PubMed] [Google Scholar]
- 107.Thomas L, Canoui-Poitrine F, Gottenberg JE, et al. Incidence of new-onset and flare of preexisting psoriasis during rituximab therapy for rheumatoid arthritis: data from the French AIR registry. J Rheumatol. 2012;39(5):893–898. [DOI] [PubMed] [Google Scholar]
- 108.Dass S, Vital EM, Emery P. Development of psoriasis after B cell depletion with rituximab. Arthritis Rheum. 2007;56(8): 2715–2718. [DOI] [PubMed] [Google Scholar]
- 109.Jost C, Hermann J, Caelen Lel S, Graninger W. New onset psoriasis in a patient receiving abatacept for rheumatoid arthritis. BMJ Case Rep. 2009;2009:pii:bcr09.2008.0845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ursini F, Naty S, Russo E, Grembiale RD. Abatacept in psoriatic arthritis: case report and short review. J Pharmacol Pharmac-other. 2013;4(suppl 1):S29–S32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Palmou-Fontana N, Sanchez Gavino JA, McGonagle D, Garcia-Martinez E, Iniguez de Onzono Martin L. Tocilizumab-induced psoriasiform rash in rheumatoid arthritis. Dermatology (Basel, Switzerland). 2014;228(4):311–313. [DOI] [PubMed] [Google Scholar]
- 112.Laurent S, Le Parc JM, Clerici T, Breban M, Mahe E. Onset of psoriasis following treatment with tocilizumab. Br J Dermatol. 2010;163(6):1364–1365. [DOI] [PubMed] [Google Scholar]

