23,856
Views
106
CrossRef citations to date
0
Altmetric
Review

An update on generalized pustular psoriasis

ORCID Icon, &
Pages 907-919 | Received 17 Jun 2019, Accepted 23 Jul 2019, Published online: 05 Sep 2019

ABSTRACT

Introduction: Generalized pustular psoriasis (GPP) is a rare, severe relapsing/remitting, multisystem disease that can be difficult to treat. Recent clinical, histological, and genetic evidence suggests that GPP is a distinct clinical entity from plaque psoriasis and requires a separate diagnosis. The interleukin-36 pathway appears to be central to GPP pathogenesis. As no therapeutic agents have been approved for GPP to date in the United States or Europe, the introduction of anti-IL-36 therapies may change disease management.

Areas covered: Using PubMed and Google Scholar, we reviewed the literature for articles related to GPP, psoriasis, and the genetics, pathogenesis, and treatment thereof.

Expert opinion: New therapeutic options and updated guidelines for GPP treatment are needed. Ideal agents would have rapid onset of action and rapid time to achieve disease clearance, have the ability to prevent acute flares and avert recurrence, and possess a favorable safety profile. Such therapies should be readily accessible via approval or listing on formularies. Scoring systems to establish GPP disease burden and objective outcome measures could also help with further evaluation of therapies and treatment access issues. IL-36 remains a promising target, as supported by early phase data suggesting efficacy and safety for a novel anti-IL-36 therapy.

Plain language summary

Generalized pustular psoriasis (GPP) is a rare dermatological condition; it is characterized by the sudden appearance of multiple small blisters filled with pus (called pustules) on large areas of the skin of the body, arms, and legs, that also become red and painful. The pustules are not infectious. Other symptoms may include fever, chills, and loss of appetite. These episodes are called flares, and can cause – in the most severe cases – life-threatening complications (such as heart failure, and/or serious infection) that may require emergency medical treatment. GPP can last from days to weeks, and often comes back. The cause of GPP is unknown, but specific factors are known to trigger a flare (such as taking certain medicines, or suddenly stopping steroid medicines). Problems with the immune system are likely to be important in causing GPP. The interleukin-36 pathway involves a group of proteins that act on immune cells in the skin (and other tissues). These proteins are linked to the development of GPP in some people. This article aims to provide a summary of the main features of GPP, including possible immune and genetic factors (particularly the interleukin-36 pathway), and the limitations of treatment options available to doctors.

Video Abstract

Read the transcript

Watch the video on Vimeo. Copyright is retained by Boehringer Ingelheim Pharmaceuticals, Inc., under a CC-BY license.

1. Introduction

Generalized pustular psoriasis (GPP) is a severe multisystem disease characterized by the sudden and widespread eruption of superficial sterile pustules [Citation1,Citation2]. It may or may not be preceded by a history of plaque psoriasis, and the systemic complications of GPP – when present – have the potential to be life-threatening, particularly in older patients (e.g., bacterial infection [Citation3], cardiorespiratory failure [Citation4]). Although children and infants can be affected by GPP, cases usually occur in adults. GPP is a rare condition, and the prevalence is greater in Asian than Caucasian populations (7.46/million in Japan versus 1.76/million in France) [Citation5,Citation6]. There is no standard treatment for GPP, and guidance often follows that for plaque psoriasis treatment. No therapeutic agents have been approved for GPP treatment in the United States or Europe to date, although both oral and conventional biologic agents have been used to successfully treat GPP. Several biologic agents have been approved in Japan for use in the treatment of GPP, including monoclonal antibodies against interleukin (IL)-17 (secukinumab, ixekizumab, and brodalumab [Citation7]) and IL-23 (guselkumab [Citation8]). Targeted biologic therapies have had a profound effect on the clinical management of individuals with plaque psoriasis (moderate and severe disease) [Citation9], although their use in GPP has been more limited. This report provides an overview of GPP, including the features of GPP and related pustular skin conditions, a review of proposed key mediators in the immunopathogenesis of GPP, and a discussion of the current options for GPP treatment and their limitations. Literature was retrieved using Boolean searches for English language articles in PubMed and Google Scholar, and including terms related to GPP, psoriasis, and the genetics, pathogenesis, and treatment thereof. The reference lists from retrieved articles were also considered. Additional data were obtained from websites of the US Food and Drug Administration (FDA) and from websites pertaining to individual therapeutic agents of interest.

2. Clinical presentation

Japanese guidelines (2018) diagnose GPP on the basis of the following primary parameters (definitive diagnosis if all four parameters are present; suspected diagnosis with two or three parameters): a) systemic symptoms such as fever and fatigue; b) systemic or extensive flushing accompanied by multiple sterile pustules that sometimes merge to form lakes of pus; c) neutrophilic subcorneal pustules histopathologically characterized by Kogoj’s spongiform pustules; and d) repeated recurrence of the above-stated clinical and histological findings [Citation7]. European guidelines (2017) define GPP as primary, sterile, macroscopically visible pustules on nonacral skin (excluding cases where pustulation is restricted to psoriatic plaques) that can occur with or without systemic inflammation, and with or without plaque psoriasis, and can be either relapsing (more than one episode) or persistent (more than 3 months) [Citation2]. Laboratory test findings in patients with GPP commonly include elevated C-reactive protein, leukocytosis and neutrophilia, and liver function abnormalities [Citation4,Citation10]. GPP may be classified into the following types: acute GPP (von Zumbusch variant; representing more than 50% to more than 90% of adult cases [Citation3,Citation11,Citation12]), pustular psoriasis of pregnancy (previously referred to incorrectly as ‘impetigo herpetiformis’ [Citation13]), and infantile/juvenile pustular psoriasis [Citation14]. Pustular psoriasis may occur in localized forms, including palmoplantar pustular psoriasis and acrodermatitis continua of Hallopeau (ACH) [Citation14]. Some clinicians also consider subcorneal pustular dermatosis (Sneddon–Wilkinson syndrome) to be a localized variant of pustular psoriasis [Citation15]. In addition, acute generalized exanthematous pustulosis (AGEP) shares many features in common with GPP [Citation16], and has been described as a drug-induced form of pustular psoriasis, although the clinical features, treatments, course, and triggers differ substantially from GPP [Citation4]. The main clinical and histological features of GPP are summarized in [Citation4,Citation13,Citation14,Citation17Citation20] and [Citation21]. Skin lesions typically observed in patients with GPP are shown in . Recently, GPP cases with autoinflammatory patho-mechanisms have been classified as an ‘autoinflammatory keratinization disease’ (AiKD) [Citation22].

Figure 1. Microscopic and macroscopic features of GPP. Histopathology of psoriasis. (a) Psoriasis vulgaris characteristically shows acanthosis, parakeratosis, and dermal inflammatory infiltrates. (b) In pustular psoriasis, acanthotic changes are accompanied by epidermal predominantly neutrophilic infiltrates, which cause pustule formation. (Reproduced from “Psoriasis Pathogenesis and Treatment” by Adriana Rendon and Knut Schäkel [Int. J. Mol. Sci. 2019; 20: 1475; http://dx.doi.org/10.3390/ijms20061475] licensed under CC-BY 4.0). (c) Macroscopic features of GPP. (Reproduced with permission from DermNet New Zealand Trust, Hamilton, New Zealand). GPP: generalized pustular psoriasis

Figure 1. Microscopic and macroscopic features of GPP. Histopathology of psoriasis. (a) Psoriasis vulgaris characteristically shows acanthosis, parakeratosis, and dermal inflammatory infiltrates. (b) In pustular psoriasis, acanthotic changes are accompanied by epidermal predominantly neutrophilic infiltrates, which cause pustule formation. (Reproduced from “Psoriasis Pathogenesis and Treatment” by Adriana Rendon and Knut Schäkel [Int. J. Mol. Sci. 2019; 20: 1475; http://dx.doi.org/10.3390/ijms20061475] licensed under CC-BY 4.0). (c) Macroscopic features of GPP. (Reproduced with permission from DermNet New Zealand Trust, Hamilton, New Zealand). GPP: generalized pustular psoriasis

Table 1. Main features of GPP

GPP is a relapsing and remitting disease with a highly variable clinical course, and symptom severity may vary with each flare episode in an individual patient [Citation17]. Recurrence of GPP flares may occur years after the initial diagnosis; conversely, patients may experience multiple flares per year. Although idiopathic in many cases, GPP flares may be caused by internal and external triggers, including infection [Citation1,Citation3,Citation11], pregnancy [Citation11], withdrawal of corticosteroids [Citation11], and initiation of other drugs (e.g., ustekinumab [Citation23], some tumor necrosis factor [TNF] antagonists [Citation24]). In some cases of drug-related disease, it may be difficult to distinguish between GPP and AGEP, as the latter also presents as a disseminated pustular eruption. However, certain medications are strongly associated with AGEP, including pristinamycin, ampicillin/amoxicillin, quinolones, anti-infective sulfonamides, terbinafine, and diltiazem [Citation25]. In contrast, the withdrawal of systemic corticosteroids in a patient with plaque psoriasis is a leading cause of GPP. Complications and morbidity from GPP may arise for a variety of reasons, including secondary bacterial infection, hypoalbuminemia (secondary to loss of plasma proteins into the tissues), hypocalcemia, renal tubular necrosis (secondary to oligemia), liver damage (secondary to oligemia and neutrophilic cholangitis), and malnutrition; furthermore, untreated patients have an increased risk of cardiorespiratory failure [Citation4]. Providing treatment is initiated promptly, the prognosis for most patients with GPP is generally good; however, elderly patients may have poorer outcomes [Citation4]. Mortality data from studies of patients with GPP are limited, but rates of 3% [Citation3] and 7% [Citation11] have been reported. As GPP often presents in individuals with existing/history of plaque psoriasis (reported in more than 65% of GPP cases [Citation11,Citation26]), GPP has been categorized as a variant of plaque psoriasis. However, recent clinical, histological, and genetic evidence suggests that GPP is a distinct clinical entity and requires a separate diagnosis [Citation2,Citation7,Citation27Citation30].

3. Pathoimmunology

The pathoimmunology of GPP is not yet fully understood. Currently suggested pathoimmunogenetic mechanisms for GPP (and plaque psoriasis) are complex, and a detailed description is beyond the scope of this report; however, these pathways are reviewed comprehensively by Johnston and colleagues [Citation29], Furue and colleagues [Citation31], and Ogawa and colleagues [Citation32]. A summary of the main points is provided below.

The IL-36 cytokines IL-36α, IL-36β, IL-36γ, and IL-36 receptor antagonist (IL-36Ra), which are part of the IL-1 family, are expressed in a variety of cell types, including keratinocytes. Binding of IL-36α, IL-36β, or IL-36γ to the IL-36 receptor (IL-36R) leads to the stimulation of inflammatory responses, inducing the release of chemokines that promote activation of neutrophils, macrophages, dendritic cells, and T cells. Gene IL36RN encodes protein IL-36Ra, which suppresses the pro-inflammatory responses triggered by IL-36α, IL-36β, and IL-36γ. Mutation of IL36RN leads to functional impairment of IL-36Ra, caused by decreased messenger RNA/protein expression or structural changes to the amino acid sequence, and subsequent enhancement of the downstream inflammatory cascade, i.e., loss-of-function mutation of IL36RN leads to exaggerated IL-36 signaling [Citation18,Citation33]. In GPP, loss-of-function mutations of IL36RN were identified in both familial [Citation18] and sporadic [Citation33] cases. Subsequent studies found that IL36RN mutations were often present in patients with GPP, particularly in those individuals without associated plaque psoriasis [Citation34,Citation35]; the reported frequency of IL36RN mutation in GPP cohorts ranges from 23% to 37% [Citation34Citation37]. The disease observed in patients with acute GPP and IL36RN mutations resulting in reduced IL-36Ra activity was named DITRA (deficiency of IL-36Ra); high-grade fever and general malaise were noted to occur during a DITRA flare [Citation18]. The influence of IL36RN mutation status on disease severity was reported in a meta-analysis of 233 patients with GPP, in which the presence of recessive IL36RN alleles (found in 49/233 [21%] cases) was associated with early disease onset (17 ± 2.4 years vs 33 ± 1.5 years in those without pathogenic IL36RN alleles; P = 5.9 x 10-7), systemic inflammation (83% vs 56%; P = 1.5 x 10-3), and absence of concurrent plaque psoriasis (36 vs 69%; P = 5.0 x 10-4) [Citation35]. Other gene mutations have been identified in different groups of patients with GPP (and related conditions) that lead to an enhanced inflammatory cascade and the recruitment of neutrophils and macrophages. These include mutations in CARD14 encoding caspase-activating recruitment domain member 14 and AP1S3 encoding adaptor protein complex 1 subunit sigma 3 (Supplementary Table 1) [Citation18,Citation20,Citation33,Citation34,Citation38Citation45].

Recent gene expression analyses have demonstrated that the transcriptome of GPP shares features in common with that of plaque psoriasis, but is inclined more toward innate immune inflammation (i.e., immediate, nonspecific responses to antigen) [Citation29,Citation46], whereas adaptive (i.e., antigen-specific) immune responses appear to have a more central role in plaque psoriasis [Citation29]. Functional IL-36R activation and loss-of-function mutation of its antagonist IL-36Ra are believed to be central to the pathogenesis of pustular psoriasis ( and online video animation), while the TNF-α/IL-23/IL-17/IL-22 axis drives plaque psoriasis (reviewed in [Citation29,Citation31,Citation32]). For comparison, acute forms of psoriasis are driven by type 1 interferons [Citation47]. However, these pathways are closely linked, and active ‘cross-talk’ between the two may result in a progressive cycle of inflammation (reviewed in [Citation31]). Microarray analyses profiling gene expression in skin biopsy samples (GPP, N = 28; plaque psoriasis, N = 12) found that both GPP and plaque psoriasis lesions overexpressed IL-17A, TNF-α, IL-1, IL-36, and interferons (IFNs), but GPP lesions had higher expression levels of IL-1β, IL-36α, and IL-36γ, with lower levels of IL-17A and IFNγ than plaque psoriasis lesions [Citation29]. GPP lesions also had greater expression of neutrophil chemokines (CXCL1, CXCL2, and CXCL8 [IL-8]) and greater enrichment of neutrophil and monocyte transcripts when compared with plaque psoriasis lesions [Citation29]. Elevated expression of IL-36 in GPP was localized to keratinocytes surrounding neutrophilic pustules [Citation29].

Figure 2. Pathoimmunology of GPP. (a) Role of IL-36 cytokines. (b) Role of IL-36 receptor antagonist. (c) Role of IL36RN gene mutation. (d) Proposed central mediators in GPP [Citation29,Citation31]. (See also online video animation)

GPP: generalized pustular psoriasis; IL: interleukin; IL36RN: IL-36 receptor antagonist gene; TNF: tumor necrosis factor.
Figure 2. Pathoimmunology of GPP. (a) Role of IL-36 cytokines. (b) Role of IL-36 receptor antagonist. (c) Role of IL36RN gene mutation. (d) Proposed central mediators in GPP [Citation29,Citation31]. (See also online video animation)

A further gene expression study using biopsies from pustular skin diseases (GPP, N = 30; palmoplantar pustulosis, N = 17; and AGEP, N = 14) reported common alterations in expression of genes affecting neutrophil chemo-attraction (including IL36RN and IL8), as well as the novel shared feature of upregulation of the six-transmembrane epithelial antigens of prostate (STEAP) proteins STEAP1 and STEAP4, which correlated with overexpression of pro-inflammatory cytokines, including IL-1, IL-36, CXCL1, and CXCL8 [Citation46]. Interestingly, STEAP1 and STEAP4 were not overexpressed in samples from patients with plaque psoriasis. Further analyses suggested that STEAP1 and STEAP4 may promote neutrophil chemotaxis in pustular psoriasis, possibly by stimulating the production of neutrophil-activating cytokines (e.g., IL-36 group, CXCL1, and CXCL8) [Citation46].

IL-17 is one of the main cytokines produced by T-helper (Th) 17/Th1 cells, which have a key role in the pathogenesis of plaque psoriasis. IL-17A is a potent inducer of tissue inflammation and neutrophil recruitment [Citation48], and is assumed to have a role in GPP pathogenesis [Citation30]. Although activation of Th17/Th1 signaling is common to both GPP and plaque psoriasis, expression levels of Th17-/Th1-related cytokines (such as IL-17A, IL-22, IL-23p19, IFNγ, and IL-18) were observed to be significantly increased in plaque psoriasis lesions compared with GPP lesions [Citation29]. Studies of palmoplantar pustular psoriasis and palmoplantar pustulosis have also observed increased gene expression of IL-17 without an increase in IL-23 [Citation49,Citation50]. These various data provide the rationale for the development of newer therapies (biologics) to target pro-inflammatory pathways in GPP [Citation29].

4. Current treatments and their limitations

Treatment options for GPP are summarized in [Citation7,Citation13,Citation14,Citation17,Citation51Citation58], and benefits and limitations of the various treatment classes are summarized in [Citation4,Citation17,Citation29,Citation30,Citation51,Citation53,Citation54,Citation56,Citation57,Citation59Citation79]. It should be reiterated that no GPP-specific therapeutic agents have been approved in the United States or Europe to date. There is also no standard guidance for GPP therapy; treatment is informed by the extent of involvement, disease severity, and the presence of any underlying risk factors [Citation70]. Treatment for GPP often follows the existing guidance for plaque psoriasis. US guidance on GPP treatment from the National Psoriasis Foundation Medical Board was published in 2012 [Citation70], prior to the availability of many of the targeted biologic therapies now used in psoriasis (plaque or pustular types). The rarity of GPP cases and the relapsing-remitting nature of this disease make it difficult to initiate randomized controlled clinical trials to investigate treatment efficacy and safety; thus, available data originate mainly from case reports and small single-arm studies.

Table 2. Treatment of GPP

Table 3. Benefits and limitations of treatment classes used in GPP

Systemic therapy in adults with GPP typically includes oral retinoids (acitretin), cyclosporine, methotrexate, or an appropriate biologic agent with a rapid speed of onset. In Japan, other types of biologics are approved for the treatment of GPP, including antagonists of IL-17 (secukinumab, ixekizumab, and brodalumab [Citation7]) and IL-23 (guselkumab [Citation8]). These and other biologics used in GPP are discussed below. It should be noted, however, that paradoxical induction of GPP has been reported with some biologics [Citation23,Citation24,Citation80]. Retinoids provide effective therapy in many GPP cases, but they also have dose-dependent adverse effects. Oral retinoids are contraindicated in women who are pregnant or who may become pregnant [Citation76]. A recent systematic review that identified 101 published adult cases of patients with GPP (predominantly acute GPP/von Zumbusch type) who had received targeted immunotherapy reported that the most common treatment agents applied before or concomitantly with targeted immunotherapy were cyclosporine and retinoids (including acitretin, etretinate, isotretinoin, and bexarotene; however, it should be noted that only acitretin is currently approved by the FDA for systemic use in psoriasis) [Citation30], followed by methotrexate and systemic corticosteroids. Case reports of GPP treatment using other agents, such as the antineutrophil agent dapsone (five cases treated) [Citation54] and the small molecule (phosphodiesterase-4 inhibitor) apremilast (one case treated) [Citation81], have also been published. Published data on the use of small molecules in GPP are generally lacking; although agents such as Janus kinase (JAK) inhibitors are in clinical development as a possible treatment for plaque psoriasis (reviewed in [Citation82]).

Second-line therapy in adults with GPP may include combination therapy of a biologic plus nonbiologic agent. Topical treatments (e.g., corticosteroids, calcipotriene, and tacrolimus) may be useful as an adjunct to systemic therapy or as first-line therapy for mild (or localized) disease. Oral corticosteroids may be considered in GPP if other treatment choices are not possible, but these agents should be used with caution and with gradual tapering of medication, as rapid withdrawal may induce a pustular flare. Psoralen ultraviolet (UV)-A phototherapy may be considered as second-line treatment in adults (although availability may be limited) but it is not recommended for acute forms of pustular psoriasis, and is not considered to be safe during pregnancy. Granulocyte and monocyte apheresis have also been utilized successfully in some refractory cases of GPP [Citation83,Citation84].

Recommendations for systemic therapy for GPP in childhood are similar to those for adults, and include acitretin (monotherapy, or in combination with oral prednisone in severe disease), methotrexate, and cyclosporine. None of these agents are FDA approved for psoriasis treatment in children due to the lack of randomized controlled trials data in this age group [Citation55]. Oral retinoids are contraindicated in young females of reproductive potential. Second-line therapy for GPP in children may include anti-TNF-α agents [Citation85] or UV-B phototherapy.

Data for the use of systemic therapy for GPP in pregnant and lactating females are also limited. Treatment options include cyclosporine (at a reduced dose), oral corticosteroids, topical agents, and phototherapy with narrow-band UV-B [Citation13]. Controlled clinical data are lacking on the safety of biologics during pregnancy in patients with psoriasis [Citation58]; however, these agents are generally classified as FDA pregnancy risk category B (under the pre-2015 lettering scheme [Citation86]) [Citation13]. Currently, anti-TNF-α agents can be used during the first half of pregnancy (due to active transplacental transport after gestational week 22), and longer-term use may be considered on an individual patient basis depending upon disease severity [Citation58]. Treatment must balance the possible adverse effects of drug therapy with the serious and potentially life-threatening consequences of GPP to the mother and fetus.

Case reports/series and some small open-label trials have been published on newer biologic therapies that target cytokines associated with GPP pathoimmunology (reviewed recently by Boehner et al. [Citation30] and Zhou et al. [Citation51]). GPP case reports of agents targeting the IL-1–/IL-36–chemokine-neutrophil axis include the recombinant IL-1 receptor antagonist anakinra [Citation87,Citation88] and the anti-IL-1β monoclonal antibodies canakinumab [Citation89] and gevokizumab (in clinical development; not yet FDA approved) [Citation90]. Further anti-IL-36 agents are in early clinical development, including ANB019 (anti-IL-36R) [Citation91], and BI 655130 (anti-IL-36R) [Citation92]. GPP case reports describe the use of the monoclonal antibody ustekinumab [Citation93,Citation94] targeting the shared p40 subunit of IL-12 and IL-23, and an open-label phase 3 study reported treatment success in 7/9 patients with GPP receiving guselkumab [Citation52], which also targets IL-23 by binding to the p19 subunit. Open-label phase 3 studies reported treatment success in 10/12 patients with GPP receiving the monoclonal antibody secukinumab (anti-IL-17A) [Citation95], 11/12 patients with GPP receiving brodalumab (anti-IL-17RA) [Citation96], and 3/5 patients with GPP receiving ixekizumab (anti-IL-17A) [Citation97,Citation98]. Secukinumab has also been used to treat a case of refractory juvenile GPP [Citation99]. Although these reports describe positive clinical outcomes (i.e., skin clearance, disease remission for varying periods, and few significant adverse events) for patients with GPP using the various biologic agents, the data are limited by the small number of patients, lack of control groups, potential bias in publishing only positive therapeutic results, and many of the studies having been carried out in Japanese populations [Citation30]. Furthermore, none of the GPP clinical trials cited above studied treatment efficacy in acute flares, but rather assessed chronic GPP using an open-label design and with the primary endpoint at 16 months or earlier. Additional research into these new biologics is needed for GPP involving larger-scale, randomized controlled clinical trials with participants from different ethnicities to provide robust efficacy and safety data. A number of clinical trials investigating the use of biologic therapies in GPP have been completed recently, and several more are ongoing (Supplementary Table 2).

5. Expert opinion

GPP is a serious and potentially life-threatening condition that must be diagnosed accurately and treated quickly. GPP adversely affects every aspect of the patient’s life during disease flare-up. The condition is painful and distressing; affected skin is covered in sterile pustules, and any contact with clothing or bedding often causes staining with blood and pus. Additionally, the skin loses all protective function, and the risk of infection (especially bacterial infection) is increased, which may be a particular issue when the patient is in the hospital. Optimal treatment for GPP would offer both a rapid onset of action for the management of acute disease flares, and a rapid time to achieve complete disease clearance. Speed of action is vital to minimize disease severity and duration of complications, and decrease the overall disease burden for the patient. Effective long-term maintenance therapy is also needed to prevent repeated GPP flares.

There are several key weaknesses in the current clinical management of GPP. The first issue is the lack of recent specific guidelines for GPP management in the United States and Europe; however, updated guidelines on GPP pathogenesis and treatment were published recently (September 2018) by the Japanese Dermatological Society [Citation7]. Secondly, the current choice of treatments for GPP is limited, and the options available generally follow what is used for plaque psoriasis. There is a profound need for GPP-specific therapeutic agents to be licensed in the United States and Europe, as are now available in Japan. Future research should include potential differential treatment responses between ethnic groups to current and novel therapies as they become approved and available in different regions. Robust efficacy data to guide treatment choices in GPP are also lacking – this applies to both conventional agents and newer therapies, such as biologics. Stronger evidence to support the use of biologics might then facilitate more rapid patient access to these medical therapies. Time to efficacy data are of particular importance for any GPP therapeutic agent being investigated, as rapid onset of action is critical in GPP patient management. There are also problems in finding effective treatments for patients with refractory disease. However, generating meaningful GPP clinical trial data is difficult, as the rarity of this condition makes it problematic to recruit sufficient numbers of patients. Furthermore, the relapsing/remitting nature of GPP and the unpredictability of acute flares present a challenge for clinical study design. Current treatment options in GPP may also be limited by other factors, such as agent-specific safety concerns. For example, retinoids are contraindicated in pregnancy and risk of pregnancy, while the risk of malignancy or infection may be increased in agents with immunosuppressive effects. With the latter, it may also be necessary to perform screening tests for certain infections (e.g., hepatitis or tuberculosis) prior to gaining access to the desired agents, which may delay GPP treatment.

Additionally, initiating treatment with newer therapies, such as biologics, may be hindered by issues in obtaining payer approval and/or prior authorization. There is a tendency for clinicians to initiate therapy with retinoids (unless contraindicated) or cyclosporine, as these agents are more accessible; however, biologics may offer clinical benefits for GPP, such as faster reduction of symptoms and a more favorable safety profile. Therapies for GPP that are readily available and approved or listed on formularies are required. To expedite future drug development, the question of whether GPP could be approached as an orphan disease may also be considered. Improved understanding of the pathogenesis of GPP could help to identify potential new therapeutic targets. The ongoing Pustular Psoriasis Elucidating Underlying Mechanisms (PLUM) study is investigating the genetic causes of GPP and biomarkers associated with the disease. The PLUM study may also help in the quest for personalized medical treatment, as targeted therapies could be appropriately prescribed for an individual instead of the ‘try-it-and-see’ approach currently in use. The localized forms of pustular psoriasis (palmoplantar pustular psoriasis, ACH), which present more frequently in the clinic than GPP, may also help in the search for an effective GPP treatment. Currently, IL-36 seems to be a promising target for GPP treatment and, possibly, also for localized pustular psoriasis. In addition, there is a requirement for objective outcome measures that are tailored to assess the clinical features of GPP (e.g., changes in overall erythema, pustules, number of lesions, body surface area affected, disease severity, etc.) that could facilitate comparisons between different therapies. A scoring system to establish disease burden in GPP could also help with treatment access issues, rather than using the Psoriasis Area and Severity Index (PASI). A recent GPP study report evaluated patients using adaptations of PASI [Citation100] and the Investigator’s Global Assessment (IGA) [Citation101]. The Generalized Pustular PASI (GPPASI) replaces the induration component with a pustule component, while the Generalized Pustular Psoriasis Physician Global Assessment (GPPGA) is a clinical assessment of the severity of pustules, erythema, and scaling of GPP lesions [Citation92]. These tools may prove useful in future GPP studies.

It is anticipated that the results from clinical trials investigating novel therapies – such as IL-36 inhibitors and other targeted biologics – will bring new treatment options and provide much-needed data on long-term maintenance of efficacy, safety, and the impact of withdrawal/re-treatment with these newer agents. The importance of targeting IL-36R signaling in GPP is supported by a recent phase 1 study (N = 7) that reported good efficacy and safety was shown by anti-IL-36R agent BI 655130 [Citation92]. Once new data from current clinical trials are published, we may also have a better understanding of the impact of the new treatments on associated comorbidities: for example, current biologics may also treat some cases of psoriatic arthritis (IL-17 inhibition) or inflammatory bowel disease (TNF/IL-23 inhibition). It is also expected that new guidelines for GPP management in North America and Europe will be developed and published within the next several years. Increased understanding of GPP pathogenesis will continue to improve our comprehension of this condition and the availability of newly developed targeted therapies will, hopefully, improve the treatment and quality of life of affected individuals.

Article highlights

  • GPP is a rare and severe multisystem disease, characterized by the sudden and widespread eruption of superficial sterile pustules – that may or may not be preceded by a history of plaque psoriasis – and potentially severe systemic complications may occur.

  • There is no standard treatment for GPP – guidance often follows that for plaque psoriasis treatment – and no therapeutic agents have been approved for GPP in the United States or Europe to date.

  • Ideal therapeutic agents in GPP would have a rapid onset of action, a rapid time to achieve disease clearance, the ability to prevent acute flares, and a favorable safety profile; such therapies should be readily accessible via approval or listing on formularies.

  • A scoring system to establish GPP disease burden could help with treatment access issues; objective outcome measures that are tailored to assess the clinical features of GPP are also required, and could facilitate comparisons between therapies.

  • Lack of inhibition of IL-36R (i.e., exaggerated IL-36 signaling), associated with a loss-of-function mutation in IL36RN that leads to functional impairment of the antagonist IL-36Ra, appears to be a key mechanism in the pathogenesis of GPP.

  • The IL-36 axis is associated with preferential expression of IL-36 cytokines, keratinocyte expression of neutrophil chemokines, and induction of inflammatory keratinocyte responses.

  • New GPP therapies using biologic agents that target pro-inflammatory pathways, including the IL-36 axis, are in various stages of clinical development.

Declaration of interest

M.J. Gooderham has been a speaker, advisory board member, and/or clinical investigator for AbbVie, Amgen, Akros, Arcutis, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Dermira, Galderma, GSK, Janssen, Kyowa Kirin, Leo Pharma, Medimmune, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi Genzyme, Sun Pharma, UCB, and Valeant/Bausch Health.

A.S. Van Voorhees has been a consultant for AbbVie, Amgen, Aqua, AstraZeneca, Celgene, Corrona, Dermira, Janssen, Leo, Novartis, and Pfizer; received a research grant from AbbVie; and has other relationship with Merck.

M.G. Lebwohl is an employee of Mount Sinai, which receives research funds from AbbVie, Amgen, Boehringer Ingelheim, Celgene, Eli Lilly, Incyte, Janssen/Johnson & Johnson, Kadmon, Leo Pharmaceuticals, Medimmune/AstraZeneca, Novartis, Pfizer, SCIderm, UCB, Ortho Dermatologics, and Vidac; M.G. Lebwohl is also a consultant for Allergan, Aqua, Arcutis, Boehringer Ingelheim, Bristol-Myers Squibb, LEO Pharma, Menlo, Mitsubishi Pharma/NeuroDerm Ltd., Promius/Dr. Reddy, Regeneron, Theravance Biopharma, and Verrica.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

A reviewer on this manuscript has disclosed that they have served as a speaker, advisory board member, and/or clinical investigator for AbbVie, Lilly, Almirall, Amgen, Celgene, Dermira, Galderma, Janssen, Leo Pharma, Novartis, Pfizer, Sanofi, UCB. Peer reviewers on this manuscript have no other relevant financial relationships or otherwise to disclose.

Supplemental material

Plain Language Summary

Download PDF (2.9 MB)

Supplemental Material

Download MS Word (41.1 KB)

Acknowledgments

Medical writing assistance, supported financially by BIPI, was provided by Debra Brocksmith, MB ChB, PhD, of Elevate Scientific Solutions during the preparation of this manuscript. BIPI was given the opportunity to check the data used in this manuscript for factual accuracy only.

Supplementary material

Supplemental data for this article can be accessed here.

Additional information

Funding

This paper was funded by Boehringer Ingelheim Pharmaceuticals, Inc. (BIPI). The authors received no direct compensation related to the development of the manuscript.

References

  • Baker H , Ryan TJ. Generalized pustular psoriasis. A clinical and epidemiological study of 104 cases. Br J Dermatol. 1968 Dec;80(12):771–793.
  • Navarini AA , Burden AD , Capon F , et al. European consensus statement on phenotypes of pustular psoriasis. J Eur Acad Dermatol Venereol. 2017 Nov;31(11):1792–1799.
  • Zelickson BD , Muller SA. Generalized pustular psoriasis. A review of 63 cases. Arch Dermatol. 1991 Sep;127(9):1339–1345.
  • Morrison AO. Pustular psoriasis. 2017 [cited 2018 Aug 08 ]. Medscape. Available from: https://emedicine.medscape.com/article/1108220-overview
  • Ohkawara A , Yasuda H , Kobayashi H , et al. Generalized pustular psoriasis in Japan: two distinct groups formed by differences in symptoms and genetic background. Acta Derm Venereol. 1996 Jan;76(1):68–71.
  • Augey F , Renaudier P , Nicolas JF . Generalized pustular psoriasis (Zumbusch): a French epidemiological survey. Eur J Dermatol. 2006 Nov-Dec;16(6):669–673.
  • Fujita H , Terui T , Hayama K , et al. Japanese guidelines for the management and treatment of generalized pustular psoriasis: the new pathogenesis and treatment of GPP. J Dermatol. 2018 Nov;45(11):1235–1270.
  • MorphoSys . MorphoSys announces approval of Tremfya® (guselkumab) for the treatment of moderate to severe forms of psoriasis and psoriatic arthritis in Japan. 2018 Apr 6 [cited 2018 Nov 12]. Available from: https://www.morphosys.com/media-investors/media-center/morphosys-announces-approval-of-tremfyar-guselkumab-for-the-treatment
  • Rønholt K , Iversen L . Old and New Biological Therapies for Psoriasis. Int J Mol Sci. 2017 Nov 1;18(11):2297.
  • Viguier M , Allez M , Zagdanski AM , et al. High frequency of cholestasis in generalized pustular psoriasis: evidence for neutrophilic involvement of the biliary tract. Hepatology. 2004 Aug;40(2):452–458.
  • Choon SE , Lai NM , Mohammad NA , et al. Clinical profile, morbidity, and outcome of adult-onset generalized pustular psoriasis: analysis of 102 cases seen in a tertiary hospital in Johor, Malaysia. Int J Dermatol. 2014 Jun;53(6):676–684.
  • Jin H , Cho HH , Kim WJ , et al. Clinical features and course of generalized pustular psoriasis in Korea. J Dermatol. 2015 Jul;42(7):674–678.
  • Trivedi MK , Vaughn AR , Murase JE . Pustular psoriasis of pregnancy: current perspectives. Int J Womens Health. 2018;10:109–115.
  • Benjegerdes KE , Hyde K , Kivelevitch D , et al. Pustular psoriasis: pathophysiology and current treatment perspectives. Psoriasis (Auckl). 2016;6:131–144.
  • Kandolf Sekulovic L . Overview of subcornal pustular dermatosis. Medscape. 2016 [cited 2018 Aug 08 ]. Available from: https://emedicine.medscape.com/article/1124252-overview#a1
  • Feldmeyer L , Heidemeyer K , Yawalkar N . Acute generalized exanthematous pustulosis: pathogenesis, genetic background, clinical variants and therapy. Int J Mol Sci. 2016 Jul 27;17:8.
  • Umezawa Y , Ozawa A , Kawasima T , et al. Therapeutic guidelines for the treatment of generalized pustular psoriasis (GPP) based on a proposed classification of disease severity. Arch Dermatol Res. 2003 Apr;295(Suppl 1):S43–S54.
  • Marrakchi S , Guigue P , Renshaw BR , et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N Engl J Med. 2011 Aug 18;365(7):620–628.
  • Berki DM , Liu L , Choon SE , et al. Activating CARD14 mutations are associated with generalized pustular psoriasis but rarely account for familial recurrence in psoriasis vulgaris. J Invest Dermatol. 2015 Dec;135(12):2964–2970.
  • Setta-Kaffetzi N , Simpson MA , Navarini AA , et al. AP1S3 mutations are associated with pustular psoriasis and impaired Toll-like receptor 3 trafficking. Am J Hum Genet. 2014 May 1;94(5):790–797.
  • Rendon A , Schakel K . Psoriasis Pathogenesis and Treatment. Int J Mol Sci. 2019 Mar 23;20:6.
  • Akiyama M , Takeichi T , McGrath JA , et al. Autoinflammatory keratinization diseases: an emerging concept encompassing various inflammatory keratinization disorders of the skin. J Dermatol Sci. 2018 May;90(2):105–111.
  • Dai Y-X , Chen -C-C Flare-up of pustular psoriasis after ustekinumab therapy: case report and literature review. Dermatol Sin. 2018;36:222–225. epub.
  • Collamer AN , Battafarano DF . Psoriatic skin lesions induced by tumor necrosis factor antagonist therapy: clinical features and possible immunopathogenesis. Semin Arthritis Rheum. 2010 Dec;40(3):233–240.
  • Sidoroff A , Dunant A , Viboud C , et al. Risk factors for acute generalized exanthematous pustulosis (AGEP)-results of a multinational case-control study (EuroSCAR). Br J Dermatol. 2007 Nov;157(5):989–996.
  • Borges-Costa J , Silva R , Goncalves L , et al. Clinical and laboratory features in acute generalized pustular psoriasis: a retrospective study of 34 patients. Am J Clin Dermatol. 2011 Aug 1;12(4):271–276.
  • Capon F . IL36RN mutations in generalized pustular psoriasis: just the tip of the iceberg? J Invest Dermatol. 2013 Nov;133(11):2503–2504.
  • Sugiura K . The genetic background of generalized pustular psoriasis: IL36RN mutations and CARD14 gain-of-function variants. J Dermatol Sci. 2014 Jun;74(3):187–192.
  • Johnston A , Xing X , Wolterink L , et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol. 2017 Jul;140(1):109–120.
  • Boehner A , Navarini AA , Eyerich K . Generalized pustular psoriasis - a model disease for specific targeted immunotherapy, systematic review. Exp Dermatol. 2018 May 31;27:1067–1077. epub (31 May 2018).
  • Furue K , Yamamura K , Tsuji G , et al. Highlighting interleukin-36 signalling in plaque psoriasis and pustular psoriasis. Acta Derm Venereol. 2018 Jan 12;98(1):5–13.
  • Ogawa E , Sato Y , Minagawa A , et al. Pathogenesis of psoriasis and development of treatment. J Dermatol. 2018 Mar;45(3):264–272.
  • Onoufriadis A , Simpson MA , Pink AE , et al. Mutations in IL36RN/IL1F5 are associated with the severe episodic inflammatory skin disease known as generalized pustular psoriasis. Am J Hum Genet. 2011 Sep 9;89(3):432–437.
  • Sugiura K , Takemoto A , Yamaguchi M , et al. The majority of generalized pustular psoriasis without psoriasis vulgaris is caused by deficiency of interleukin-36 receptor antagonist. J Invest Dermatol. 2013 Nov;133(11):2514–2521.
  • Hussain S , Berki DM , Choon SE , et al. IL36RN mutations define a severe autoinflammatory phenotype of generalized pustular psoriasis. J Allergy Clin Immunol. 2015 Apr;135(4):1067–1070.e9.
  • Körber A , Mossner R , Renner R , et al. Mutations in IL36RN in patients with generalized pustular psoriasis. J Invest Dermatol. 2013 Nov;133(11):2634–2637.
  • Twelves S , Mostafa A , Dand N , et al. Clinical and genetic differences between pustular psoriasis subtypes. J Allergy Clin Immunol. 2019 Mar;143(3):1021–1026.
  • National Institutes of Health . IL36RN gene, interleukin 36 receptor antagonist U.S. National Library of Medicine; [cited 2018 Aug 18 ]. Available from: https://ghr.nlm.nih.gov/gene/IL36RN
  • National Institutes of Health . CARD14 gene, caspase recruitment domain family member 14: U.S. National Library of Medicine; [cited 2018 Aug 18 ]. Available from: https://ghr.nlm.nih.gov/gene/CARD14
  • National Institutes of Health . AP1S3 gene, adaptor related protein complex 1 subunit sigma 3: U.S. National Library of Medicine; [ cited 2018 Aug 18]. Available from: https://ghr.nlm.nih.gov/gene/AP1S3
  • Setta-Kaffetzi N , Navarini AA , Patel VM , et al. Rare pathogenic variants in IL36RN underlie a spectrum of psoriasis-associated pustular phenotypes. J Invest Dermatol. 2013 May;133(5):1339–1366.
  • Jordan CT , Cao L , Roberson ED , et al. PSORS2 is due to mutations in CARD14. Am J Hum Genet. 2012 May 4;90(5):784–795.
  • Fuchs-Telem D , Sarig O , van Steensel MA , et al. Familial pityriasis rubra pilaris is caused by mutations in CARD14. Am J Hum Genet. 2012 Jul 13;91(1):163–170.
  • Israel L , Mellett M . Clinical and genetic heterogeneity of CARD14 mutations in psoriatic skin disease. Front Immunol. 2018;9:2239.
  • Mahil SK , Twelves S , Farkas K , et al. AP1S3 mutations cause skin autoinflammation by disrupting keratinocyte autophagy and up-regulating IL-36 production. J Invest Dermatol. 2016 Nov;136(11):2251–2259.
  • Liang Y , Xing X , Beamer MA , et al. Six-transmembrane epithelial antigens of the prostate comprise a novel inflammatory nexus in patients with pustular skin disorders. J Allergy Clin Immunol. 2017 Apr;139(4):1217–1227.
  • Nestle FO , Conrad C , Tun-Kyi A , et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med. 2005 Jul 4;202(1):135–143.
  • Nograles KE , Zaba LC , Guttman-Yassky E , et al. Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways. Br J Dermatol. 2008 Nov;159(5):1092–1102.
  • Bissonnette R , Nigen S , Langley RG , et al. Increased expression of IL-17A and limited involvement of IL-23 in patients with palmo-plantar (PP) pustular psoriasis or PP pustulosis; results from a randomised controlled trial. J Eur Acad Dermatol Venereol. 2014 Oct;28(10):1298–1305.
  • Bissonnette R , Suarez-Farinas M , Li X , et al. Based on molecular profiling of gene expression, palmoplantar pustulosis and palmoplantar pustular psoriasis are highly related diseases that appear to be distinct from psoriasis vulgaris. PLoS One. 2016;11(5):e0155215.
  • Zhou LL , Georgakopoulos JR , Ighani A , et al. Systemic monotherapy treatments for generalized pustular psoriasis: a systematic review. J Cutan Med Surg. 2018;1:1203475418773358.
  • Sano S , Kubo H , Morishima H , et al. Guselkumab, a human interleukin-23 monoclonal antibody in Japanese patients with generalized pustular psoriasis and erythrodermic psoriasis: efficacy and safety analyses of a 52-week, phase 3, multicenter, open-label study. J Dermatol. 2018 May;45(5):529–539.
  • Janssen Biotech I . Prescribing information: TREMFYA (guselkumab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761061s000lbl.pdf
  • Sheu JS , Divito SJ , Enamandram M , et al. Dapsone therapy for pustular psoriasis: case series and review of the literature. Dermatology. 2016;232(1):97–101.
  • Napolitano M , Megna M , Balato A , et al. Systemic treatment of pediatric psoriasis: a review. Dermatol Ther (Heidelb). 2016 6;Jun(2):125–142.
  • Janssen Biotech I . Prescribing information: REMICADE (infliximab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/103772s5385lbl.pdf
  • Amgen Inc . Prescribing information: ENBREL® (etanercept). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/103795s5563s5564lbl.pdf
  • Porter ML , Lockwood SJ , Kimball AB . Update on biologic safety for patients with psoriasis during pregnancy. Int J Womens Dermatol. 2017. 3. Mar(1):21–25.
  • AbbVie Inc . Prescribing information: HUMIRA (adalimumab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125057s406lbl.pdf
  • Swedish Orphan Biovitrum AB (publ) . Prescribing information: KINERET® (anakinra). [cited 20 Aug 2018]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/103950s5182lbl.pdf
  • Novartis Pharmaceuticals Corporation . Prescribing information: ILARIS® (canakinumab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/125319s088lbl.pdf
  • Novartis Pharmaceuticals Corporation . Prescribing information: COSENTYX® (secukinumab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125504s013lbl.pdf
  • Valeant Pharmaceuticals North America LLC . Prescribing information: SILIQ™ (brodalumab). [cited 2018 Aug 20]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761032lbl.pdf
  • Eli Lilly and Company . Prescribing information: TALTZ (ixekizumab). [cited 2018 Aug 20 ]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125521s009lbl.pdf
  • Janssen Biotech I . Prescribing information: STELARA® (ustekinumab). [cited 2018 Aug 20]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/761044lbl.pdf
  • Prens EP , van Joost T , Hegmans JP , et al. Effects of cyclosporine on cytokines and cytokine receptors in psoriasis. J Am Acad Dermatol. 1995 Dec;33(6):947–953.
  • Wozel G , Blasum C . Dapsone in dermatology and beyond. Arch Dermatol Res. 2014 Mar;306(2):103–124.
  • Uva L , Miguel D , Pinheiro C , et al. Mechanisms of action of topical corticosteroids in psoriasis. Int J Endocrinol. 2012;2012:561018.
  • Wong T , Hsu L , Liao W . Phototherapy in psoriasis: a review of mechanisms of action. J Cutan Med Surg. 2013 Jan-Feb;17(1):6–12.
  • 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 Aug;67(2):279–288.
  • Langley RG , Elewski BE , Lebwohl M , et al. Secukinumab in plaque psoriasis–results of two phase 3 trials. N Engl J Med. 2014 Jul 24;371(4):326–338.
  • 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 Jan;76(1):60–69 e9.
  • Hunt MJ , Lee SH , Salisbury EL , et al. Generalized pustular psoriasis responsive to PUVA and oral cyclosporin therapy. Australas J Dermatol. 1997 Nov;38(4):199–201.
  • Kopp T , Karlhofer F , Szepfalusi Z , et al. Successful use of acitretin in conjunction with narrowband ultraviolet B phototherapy in a child with severe pustular psoriasis, von Zumbusch type. Br J Dermatol. 2004 Oct;151(4):912–916.
  • Honigsmann H , Gschnait F , Konrad K , et al. Photochemotherapy for pustular psoriasis (von Zumbusch). Br J Dermatol. 1977 Aug;97(2):119–126.
  • Khalil S , Bardawil T , Stephan C , et al. Retinoids: a journey from the molecular structures and mechanisms of action to clinical uses in dermatology and adverse effects. J Dermatolog Treat. 2017 Dec;28(8):684–696.
  • American Academy of Dermatology . Psoriasis: recommendations for cyclosporine. [cited 2019 Jan 22 ]. Available from: https://www.aad.org/practicecenter/quality/clinical-guidelines/psoriasis/systemic-agents/recommendations-for-cyclosporine
  • Medscape . Dapsone (Rx). [cited 2018 Nov 15 ]. Available from: https://reference.medscape.com/drug/aczone-dapsone-342559#10
  • Bandoli G , Palmsten K , Forbess Smith CJ , et al. A review of systemic corticosteroid use in pregnancy and the risk of select pregnancy and birth outcomes. Rheum Dis Clin North Am. 2017 Aug;43(3):489–502.
  • Almutairi D , Sheasgreen C , Weizman A , et al. Generalized pustular psoriasis induced by infliximab in a patient with inflammatory bowel disease. J Cutan Med Surg. 2018;1:507–510.
  • Jeon C , Nakamura M , Sekhon S , et al. Generalized pustular psoriasis treated with apremilast in a patient with multiple medical comorbidities. JAAD Case Rep. 2017 Nov;3(6):495–497.
  • Fragoulis GE , McInnes IB , Siebert S . JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford). 2019 Feb 1;58(Supplement_1):i43–i54.
  • Ikeda S , Takahashi H , Suga Y , et al. Therapeutic depletion of myeloid lineage leukocytes in patients with generalized pustular psoriasis indicates a major role for neutrophils in the immunopathogenesis of psoriasis. J Am Acad Dermatol. 2013 Apr;68(4):609–617.
  • Fujisawa T , Suzuki S , Mizutani Y , et al. Granulocyte and monocyte adsorption apheresis for generalized pustular psoriasis: therapeutic outcomes in three refractory patients. Ther Apher Dial. 2015 Aug;19(4):336–341.
  • Saikaly SK , Mattes M . Biologics and pediatric generalized pustular psoriasis: an emerging therapeutic trend. Cureus. 2016 Jun 22;8(6):e652.
  • US Food and Drug Administration . Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (Federal Register/Vol. 73, No. 104/Thursday, May 29, 2008). [cited 2019 Jan 23 ]. Available from: https://www.govinfo.gov/content/pkg/FR-2008-05-29/pdf/E8-11806.pdf
  • Viguier M , Guigue P , Pages C , et al. Successful treatment of generalized pustular psoriasis with the interleukin-1-receptor antagonist Anakinra: lack of correlation with IL1RN mutations. Ann Intern Med. 2010 Jul 6;153(1):66–67.
  • Huffmeier U , Watzold M , Mohr J , et al. Successful therapy with anakinra in a patient with generalized pustular psoriasis carrying IL36RN mutations. Br J Dermatol. 2014 Jan;170(1):202–204.
  • Skendros P , Papagoras C , Lefaki I , et al. Successful response in a case of severe pustular psoriasis after interleukin-1beta inhibition. Br J Dermatol. 2017 Jan;176(1):212–215.
  • Mansouri B , Richards L , Menter A . Treatment of two patients with generalized pustular psoriasis with the interleukin-1beta inhibitor gevokizumab. Br J Dermatol. 2015 Jul;173(1):239–241.
  • Khanskaya I , Pinkstaff J , Marino MH , et al. A phase 1 study of ANB019, an anti-IL-36 receptor monoclonal antibody, in healthy volunteers. Poster presented at the 2018 European Academy of Allergy and Clinical Immunology (EAACI) Congress; 2018 May 26–30; Munich, Germany. [cited 2018 Aug 14 ]. Available from: https://www2.anaptysbio.com/wp-content/uploads/ANB019-Phase-1-Study-Poster-EAACI-2018.pdf
  • Bachelez H , Choon S-E , Marrakchi S , et al. Inhibition of the interleukin-36 pathway for the treatment of generalized pustular psoriasis. N Engl J Med. 2019 Mar 7;380(10):981–983.
  • Arakawa A , Ruzicka T , Prinz JC . Therapeutic efficacy of interleukin 12/interleukin 23 blockade in generalized pustular psoriasis regardless of IL36RN mutation status. JAMA Dermatol. 2016 Jul 1;152(7):825–828.
  • Storan ER , O’Gorman SM , Markham T . Generalized pustular psoriasis treated with ustekinumab. Clin Exp Dermatol. 2016 Aug;41(6):689–690.
  • Imafuku S , Honma M , Okubo Y , et al. Efficacy and safety of secukinumab in patients with generalized pustular psoriasis: A 52-week analysis from phase III open-label multicenter Japanese study. J Dermatol. 2016 Sep;43(9):1011–1017.
  • Yamasaki K , Nakagawa H , Kubo Y , et al. Efficacy and safety of brodalumab in patients with generalized pustular psoriasis and psoriatic erythroderma: results from a 52-week, open-label study. Br J Dermatol. 2017 Mar;176(3):741–751.
  • Saeki H , Nakagawa H , Nakajo K , et al. Efficacy and safety of ixekizumab treatment for Japanese patients with moderate to severe plaque psoriasis, erythrodermic psoriasis and generalized pustular psoriasis: results from a 52-week, open-label, phase 3 study (UNCOVER-J). J Dermatol. 2017 Apr;44(4):355–362.
  • Saeki H , Nakagawa H , Ishii T , et al. Efficacy and safety of open-label ixekizumab treatment in Japanese patients with moderate-to-severe plaque psoriasis, erythrodermic psoriasis and generalized pustular psoriasis. J Eur Acad Dermatol Venereol. 2015 Jun;29(6):1148–1155.
  • Ho P-H , Tsai T-F . Successful treatment of refractory juvenile generalized pustular psoriasis with secukinumab monotherapy: A case report and review of published work. J Dermatol. 2018 Sep 19;45:1353–1356.
  • Fredriksson T , Pettersson U . Severe psoriasis–oral therapy with a new retinoid. Dermatologica. 1978;157(4):238–244.
  • Langley RG , Feldman SR , Nyirady J , et al. The 5-point Investigator’s Global Assessment (IGA) Scale: A modified tool for evaluating plaque psoriasis severity in clinical trials. J Dermatolog Treat. 2015 Feb;26(1):23–31.