Back to Journals » Psoriasis: Targets and Therapy » Volume 9

Diagnosis and screening of patients with generalized pustular psoriasis

Authors Ly K , Beck KM, Smith MP, Thibodeaux Q , Bhutani T

Received 31 March 2019

Accepted for publication 1 June 2019

Published 20 June 2019 Volume 2019:9 Pages 37—42

DOI https://doi.org/10.2147/PTT.S181808

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Prof. Dr. Uwe Wollina



Karen Ly,* Kristen M Beck*, Mary P Smith, Quinn Thibodeaux, Tina Bhutani

Department of Dermatology, University of California, San Francisco, CA, USA

*These authors contributed equally to this work

Abstract: Generalized pustular psoriasis (GPP) is a rare and potentially life-threatening variant of psoriasis that is characterized by recurrent, acute onset, widely distributed pustular eruptions on inflamed, erythematous skin. It is important to recognize acute GPP as a subtype of psoriasis associated with high morbidity and mortality so therapy can be initiated without delay. Since GPP was first described in 1910 by Leopold von Zumbusch, it has been inconsistently defined, stratified, and diagnosed in the literature. Multiple definitions and diagnostic criteria have been proposed over the years. Recently, formal consensus guidelines on GPP have been published by international groups. This article reviews the current evidence and understanding in the diagnosis and screening of GPP.

Keywords: generalized pustular psoriasis, acute generalized pustular psoriasis of von Zumbusch, pustular psoriasis, diagnostic criteria

Introduction

Generalized pustular psoriasis (GPP) is a rare, severe, and potentially life-threatening variant of psoriasis that is characterized by recurrent, acute onset, widely distributed pustular eruptions on inflamed, erythematous skin. Prompt diagnosis of GPP may prevent morbidity from complications including sepsis, acute renal failure, high-output congestive heart failure, and acute respiratory distress syndrome.3 This article reviews the current evidence and understanding in the diagnosis and screening of GPP.

Background

In contrast to chronic plaque psoriasis, which accounts for the majority of psoriasis vulgaris (PV) cases worldwide, GPP is considered to be a rare disease. A 2018 Japanese epidemiological analysis reported that GPP represented just 1.8% of all clinical types of psoriasis.4 However, the exact prevalence of GPP is unknown. One reason for this is that since its first description in 1910 by Leopold von Zumbusch,1 it has been inconsistently defined, stratified, and diagnosed in the literature.2 Rigorous studies characterizing GPP have been limited by both the rarity of the disease and by multiple definitions and diagnostic criteria that have been proposed over the years.

Historically, pustular psoriasis has been classified as either generalized or localized. The generalized forms are then further stratified by the acuity of presentation, acute GPP (also known as GPP, generalized pustular psoriasis of von Zumbusch, or von Zumbusch type GPP) and subacute GPP (also known as annular pustular psoriasis (APP) or annular pattern GPP).57

Acute onset GPP is the most common type, which accounts for more than two-thirds of GPP cases.3,8 When acute GPP occurs in pregnancy, it is often termed impetigo herpetiformis (IH). Juvenile pustular psoriasis (JPP) is a subtype of GPP that occurs in the pediatric population.9,10

Although some studies have included APP as a variant of GPP, this is controversial due to its subacute course and mild systemic symptoms.11 Subacute GPP is characterized by annular lesions with pustules, erythema, and scaling at the advancing edge.3,11,12 Patients typically lack systemic symptoms and laboratory abnormalities.

GPP is also distinguished from the transient, primary pustule formation that may occur at the periphery of psoriasis plaques in PV. This may occur in patients with PV during periods of disease exacerbation or following the application of irritants, such as tars. Although this phenomenon has been termed “localized GPP” and “psoriasis cum pustulatione”2 (psoriasis with pustules), it is generally recognized that it is not a type of pustular psoriasis.

Clinical diagnosis

The diagnosis of GPP should be suspected in patients with acute onset erythema and pustulosis, and subsequently evaluated using clinicopathologic correlation between physical examination findings, patient history, review of symptoms, and histopathology.

GPP presents as the rapid onset of widespread, erythematous, inflamed skin studded with 2- to 3-mm sterile pustules.12 Pustules may expand and coalesce into irregular “lakes of pus.” During episodes of pustulation, patients may develop abnormal clinical findings associated with systemic inflammation. Patients appear systemically ill, with high-grade fever, chills, malaise, and anorexia.8,11,12 Erythroderma may occur. After 1–2 days, the pustules typically resolve with residual erythema and desquamation.3

A physical examination of the skin and oral mucosa is important to evaluate the extent of skin involvement. Associated cutaneous symptoms may include pain, burning, and pruritus.8 Associated mucosal findings include a geographic or fissured tongue, cheilitis, and ocular involvement (eg, conjunctivitis, uveitis, iritis).3,13 Additional extracutaneous findings may include nail abnormalities, arthralgias, jaundice, and lower extremity edema.15,16

A history of concurrent or previous PV may be helpful in confirming the diagnosis; however, not all patients have a history of psoriasis. GPP may occur with or without a history of PV.3,17,18 GPP may affect patients at any age, but most commonly affects patients in the fourth decade.3,13,16 GPP is also reported to occur at a higher incidence in women.16,18

Although many cases appear to be idiopathic, medications, infections, and pregnancy have been reported as triggers of GPP.3,8,12 A detailed drug history is important as it can associate the initiation or withdrawal of a drug with the onset of disease.3,7 Rapid tapering of systemic corticosteroids is a well-known and frequently reported trigger for GPP flares.19,20 Many other medications have also been implicated as a trigger, including antibiotics such as amoxicillin,21 terbinafine,22 calcipotriol ointment,23 betamethasone ointment,23 tumor necrosis factor-alpha (TNF-α) inhibitors,24,25 ustekinumab,24,26 and withdrawal of cyclosporine.27

Infections reported as potential etiological factors in GPP include streptococcal,28 Trichophyton rubrum,29 cytomegalovirus,30,31 Epstein-Barr virus,32 and varicella-zoster virus.33 GPP has also been associated with various medical conditions including Turner syndrome,4 hypoparathyroidism,34 hypocalcemia,34 allogeneic stem cell transplantation,35 rheumatoid arthritis,36 and cardiomyopathy.4

On initial evaluation, it is important to determine which patients require immediate hospitalization due to risk of complication. Systemic symptoms including fever, chills, shortness of breath, and altered mental status may indicate the need for inpatient admission. Obtaining a history of comorbid medical conditions may also help triage patients who are at higher risk for complications. These include advanced age, congestive heart failure, renal insufficiency, diabetes mellitus, or immunodeficiency.

Patients with GPP may develop life-threatening complications, including sepsis, acute renal failure, neutrophilic cholangitis, high-output congestive heart failure, acute respiratory distress syndrome, and death.3,8 Mortality in GPP is most commonly due to complications from sepsis, acute respiratory distress syndrome, and cardiac failure.3,17 It is therefore critical to identify which patients require hospitalization for stabilization, rapid treatment, and to reduce morbidity and mortality.

There is no cure for GPP. GPP may be relapsing, with recurrence either idiopathic or only when exposed to a trigger, or persistent, with symptoms lasting for months. Symptoms may self-resolve or require aggressive treatment.8

Laboratory abnormalities

Laboratory evaluations are strongly recommended and deemed necessary to assess for severity and potential complications associated with GPP. Laboratory abnormalities would be consistent with the systemic involvement seen in GPP, the most common of which are leukocytosis with neutrophilia and elevated erythrocyte sedimentation rate (ESR).3 Complete blood count with differential (to evaluate for leukocytosis and lymphopenia) and a comprehensive metabolic panel (to evaluate for hypocalcemia, other electrolyte abnormalities, hypoalbuminemia, and to evaluate liver and renal function) are common initial assessments. Skin desquamation and epidermal barrier disruption may result in electrolyte abnormalities. Hypocalcemia can occur as a result of hypoalbuminemia, but ionized calcium is typically normal and patients are asymptomatic.8,12 Patients may have increased alkaline phosphatase, transaminase, and bilirubin levels.6,8 Patient who report an antecedent upper respiratory infection may have positive anti-streptolysin titers.

Cultures of pustules and blood samples, gram stains, and potassium hydroxide preparation can also be performed to rule out other causes of pustulosis.17

Histopathology

Skin biopsy is important to confirm the diagnosis of GPP.38 The histopathology of GPP is characterized by Kogoj’s spongiform pustules, which are the accumulations of neutrophils under the stratum corneum, as well as the classic findings of psoriasis, which include parakeratosis, acanthosis, hyperkeratosis, elongation of rete ridges, diminished stratum granulosum, capillary dilation of the papillary dermis, Munro’s microabscesses, and superficial perivascular mononuclear cell infiltrations. The edema and inflammatory cell infiltrate is notably greater than what is observed in PV.11,3739

Other diseases with similar histopathology include acute generalized exanthematous pustulosis (AGEP). However, the additional presence of eosinophils and necrotic keratinocytes, which are suggestive of an underlying drug trigger, would likely be seen.40

Differential diagnosis

The list of differential diagnoses for GPP is vast and includes many cutaneous pustular diseases, including AGEP, subacute annular pustular psoriasis (APP), localized forms of pustular psoriasis (eg, palmoplantar pustular psoriasis, Acrodermatitis continua of Hallopeau), pemphigus foliaceus, IgA pemphigus, and subcorneal pustular dermatosis.11,38

The most important diagnosis to exclude is AGEP, a rare and severe pustular skin reaction, most commonly due to medications.41 It presents as sterile pustules on an erythematous base and can be clinically and histologically difficult to differentiate from GPP. Clinically, AGEP has a more abrupt onset, shorter duration, does not recur, has no personal or family history of PV, and is associated with a recently initiated medication.38,41 AGEP may also be accompanied by blood eosinophilia.38

Genetic screening

Although the etiology of GPP remains to be fully elucidated, a genetic basis that may cause or contribute to pustular variants of psoriasis, distinct from that of chronic plaque psoriasis, has been identified.

In recent years, genetic studies have identified three gene mutations in one or more forms of pustular psoriasis-mutations in IL36RN (encodes IL-36 receptor antagonist), CARD14 (encodes a keratinocyte adaptor protein), and AP1S3 (encodes a subunit of the adaptor protein 1 complex). These three mutations account for fewer than 30% of GPP cases.42 IL36RN mutations are the most frequent genetic abnormality observed in pustular psoriasis and occur in approximately 20% of GPP patients.43 AP1S3 and CARD14 variants are found in fewer than 10% of these patients.

Currently, genetic screenings for IL36RN, CARD14, and AP1S3 mutations are not routinely indicated. However, IL36RN mutations are being increasingly used to aid in the diagnosis of GPP. Genotype–phenotype studies have shown that IL36RN mutations are associated with an earlier age of onset in GPP, widespread inflammation, and are not associated with concurrent PV.18,43,44 When this clinical triad is present in familial IL36RN mutations, it is described by the acronym DITRA (deficiency of the interleukin 36 receptor antagonist).45 Twelves et al demonstrated that IL36RN mutations were associated with an earlier age of onset across all pustular psoriasis subtypes, with the youngest age of onset found in GPP patients (mean age of onset 31 years ±19.7 years). Based on this finding, this study recommended that patients who present with GPP before age 30 be screened for IL36RN mutations.18

Proposed diagnostic criteria

Recent consensus guidelines on GPP have been published by European and Japanese groups in order to distinguish GPP from other pustular diseases (Table 1). One goal of these guidelines is to develop an extended cohort using standard classification in order to reveal clinical and genetic insights into patient demographics, clinical features, and genetic mutations that may improve diagnosis and early treatment of this potentially life-threatening disease.

Table 1 Summary of diagnostic criteria

In 2017, the European Rare and Severe Psoriasis Expert Network (ERASPEN) published the first European consensus statement2 on the phenotypes of pustular psoriasis. GPP was defined as primary, sterile, macroscopically visible pustules on non-acral skin. In addition to this, the criteria included subclassifiers that indicate if GPP occurs with or without PV and with or without systemic inflammation. The criteria also specify that a diagnosis of GPP can only be made when the condition has relapsed or persisted for more than three months. No severity assessment was published with this report. However, ERASPEN did acknowledge the need for updated severity criteria. It was thought that using the number of pustules present to measure severity would be limited by the diverse morphology of GPP, which may include either discrete or confluent pustules. If a patient presents with mixed types of pustular psoriasis (eg, both GPP and a localized form of pustular psoriasis, such as Arodermatitis continua of Hallopeau), then the patient should be classified according to the most predominant feature.

The Japanese guideline was published in 2018 by the Japanese Dermatological Association and the Study Group for Rare Intractable Skin Diseases as an update to Umezawa et al’s37 2003 GPP guideline. This guideline by Fujita et al defines GPP as a rare disease in which acute fever, generalized skin rashes, and many sterile pustules develop. Histopathologically, GPP forms subcorneal pustules characterized by Kogoj’s spongiform pustules. GPP may or may not be preceded by PV and is characterized by repeated disease recurrence. During the course of the disease, patients have abnormal clinical findings associated with systemic inflammation and often present with mucosal symptoms and arthritis as complications. Although rare, GPP may be accompanied by certain eye symptoms and secondary amyloidosis.

Fujita et al made a few important additions to the definition of GPP. The definition was updated to include that GPP may have potential complications of arthritis, mucosal and ocular symptoms, and secondary amyloidosis. This addition to the definition was made to emphasize that GPP involves more than just skin lesions, it causes systemic inflammation and has the potential for serious complications. Another significant difference between the 2003 and 2018 Japanese criteria was the removal of laboratory abnormalities from the criteria. This was due to insufficient sensitivity and specificity of laboratory abnormalities in the diagnosis of GPP; instead, laboratory abnormalities were strongly recommended and deemed necessary to assess for severity and potential complications (Table 2).

Table 2 Summary of assessments included in criteria

Conclusion

The diagnosis of GPP can be challenging due to its rarity, heterogeneous presentation, and lack of consistent classification, but it is important to recognize acute GPP as a potentially fatal subtype of psoriasis so therapy can be initiated without delay. GPP is diagnosed based on the presence of visible pustules on erythematous skin with confirmation from histopathology, and supportive evidence from laboratory and genetic evaluations. The most important diagnosis to exclude is AGEP.

These findings are reflected in recent consensus guidelines by the ERASPEN and the Japanese Dermatological Association.2,14 Both guidelines share two features: the clinical presence of pustules and disease recurrence. Other requirements for diagnosis such as systemic inflammation, Kogoj’s spongiform pustules, and laboratory abnormalities vary depending on the criteria used.14,37,46 These diagnostic criteria can be utilized for the assessment and diagnosis of GPP, but there remains a need for a unified diagnostic criteria that can be universally adopted and allow for further characterization of GPP in larger cohorts. Further genetic studies are needed to determine the clinical significance of known genetic mutations in the classification, diagnosis, and screening of GPP.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Zumbusch V. Leopold. Psoriasis and pustuloses Exanthem. Arch Dermatol Syphilol. 1910;99:335–346. doi:10.1007/BF01910970

2. Navarini AA, Burden AD, Capon F, et al. European consensus statement on phenotypes of pustular psoriasis. J Eur Acad Dermatol Venereol. 2017;31(11):1792–1799. doi:10.1111/jdv.14386

3. Choon SE, Lai NM, Mohammad NA, Nanu NM, Tey KE, Chew SF. 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;53(6):676–684. doi:10.1111/ijd.12070

4. Ito T, Takahashi H, Kawada A, Iizuka H, Nakagawa H. Epidemiological survey from 2009 to 2012 of psoriatic patients in Japanese Society for Psoriasis Research. J Dermatol. 2018;45(3):293–301. doi:10.1111/1346-8138.14105

5. Griffiths CE, Christophers E, Barker JN, et al. A classification of psoriasis vulgaris according to phenotype. Br J Dermatol. 2007;156(2):258–262. doi:10.1111/j.1365-2133.2006.07675.x

6. Benjegerdes KE, Hyde K, Kivelevitch D, Mansouri B. Pustular psoriasis: pathophysiology and current treatment perspectives. Psoriasis (Auckl). 2016;6:131–144. doi:10.2147/PTT.S98954

7. Raychaudhuri SK, Maverakis E, Raychaudhuri SP. Diagnosis and classification of psoriasis. Autoimmun Rev. 2014;13(4–5):490–495. doi:10.1016/j.autrev.2014.01.008

8. Borges-Costa J, Silva R, Goncalves L, Filipe P, Soares de Almeida L, Marques Gomes M. Clinical and laboratory features in acute generalized pustular psoriasis: a retrospective study of 34 patients. Am J Clin Dermatol. 2011;12(4):271–276. doi:10.2165/11586900-000000000-00000

9. de Oliveira ST, Maragno L, Arnone M, Fonseca Takahashi MD, Romiti R. Generalized pustular psoriasis in childhood. Pediatr Dermatol. 2010;27(4):349–354. doi:10.1111/j.1525-1470.2010.01084.x

10. Lau BW, Lim DZ, Capon F, Barker JN, Choon SE. Juvenile generalized pustular psoriasis is a chronic recalcitrant disease: an analysis of 27 patients seen in a tertiary hospital in Johor, Malaysia. Int J Dermatol. 2017;56(4):392–399. doi:10.1111/ijd.13489

11. Naik HB, Cowen EW. Autoinflammatory pustular neutrophilic diseases. Dermatol Clin. 2013;31(3):405–425. doi:10.1016/j.det.2013.04.001

12. Baker H, Ryan TJ. Generalized pustular psoriasis. A clinical and epidemiological study of 104 cases. Br J Dermatol. 1968;80(12):771–793.

13. Jin H, Cho HH, Kim WJ, et al. Clinical features and course of generalized pustular psoriasis in Korea. J Dermatol. 2015;42(7):674–678. doi:10.1111/1346-8138.12863

14. 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;45(11):1235–1270. doi:10.1111/1346-8138.14523

15. 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;40(2):452–458. doi:10.1002/hep.20305

16. Tay YK, Tham SN. The profile and outcome of pustular psoriasis in Singapore: a report of 28 cases. Int J Dermatol. 1997;36(4):266–271.

17. Hoegler KM, John AM, Handler MZ, Schwartz RA. Generalized pustular psoriasis: a review and update on treatment. J Eur Acad Dermatol Venereol. 2018;32(10):1645–1651. doi:10.1111/jdv.14949

18. Twelves S, Mostafa A, Dand N, et al. Clinical and genetic differences between pustular psoriasis subtypes. J Allergy Clin Immunol. 2019;143(3):1021–1026. doi:10.1016/j.jaci.2018.06.038

19. Brenner M, Molin S, Ruebsam K, Weisenseel P, Ruzicka T, Prinz JC. Generalized pustular psoriasis induced by systemic glucocorticosteroids: four cases and recommendations for treatment. Br J Dermatol. 2009;161:964–966. doi:10.1111/j.1365-2133.2009.09348.x

20. Westphal DC, Schettini AP, Souza PP, Castiel J, Chirano CA, Santos M. Generalized pustular psoriasis induced by systemic steroid dose reduction. An Bras Dermatol. 2016;91(5):664–666. doi:10.1590/abd1806-4841.20163804

21. Sugiura K, Shoda Y, Akiyama M. Generalized pustular psoriasis triggered by amoxicillin in monozygotic twins with compound heterozygous IL36RN mutations: comment on the article by Navarini et al. J Invest Dermatol. 2014;134(2):578–579. doi:10.1038/jid.2013.354

22. Ozturk G, Turk BG, Karaca N, et al. Generalized pustular eruptions due to terbinafine. Cutan Ocul Toxicol. 2012;31(1):81–84. doi:10.3109/15569527.2011.607202

23. Tobin AM, Langan SM, Collins P, Kirby B. Generalized pustular psoriasis (von Zumbusch) following the use of calcipotriol and betamethasone dipropionate ointment: a report of two cases. Clin Exp Dermatol. 2009;34:629–630. doi:10.1111/j.1365-2230.2008.03034.x

24. Hay RA, Pan JY. Paradoxical flare of pustular psoriasis triggered by ustekinumab, which responded to adalimumab therapy. Clin Exp Dermatol. 2014;39(6):751–752. doi:10.1111/ced.12392

25. Goiriz R, Dauden E, Perez-Gala S, Guhl G, Garcia-Diez A. Flare and change of psoriasis morphology during the course of treatment with tumour necrosis factor blockers. Clin Exp Dermatol. 2007;32(2):176–179. doi:10.1111/j.1365-2230.2006.02315.x

26. Gregoriou S, Kazakos C, Christofidou E, Kontochristopoulos G, Vakis G, Rigopoulos D. Pustular psoriasis development after initial ustekinumab administration in chronic plaque psoriasis. Eur J Dermatol. 2011;21(1):104–105. doi:10.1684/ejd.2011.1164

27. Georgala S, Koumantaki E, Rallis E, Papadavid E. Generalized pustular psoriasis developing during withdrawal of short-term cyclosporin therapy. Br J Dermatol. 2000;142:1057–1058.

28. Maiolo C, Kwok SM, Ross C, Ibbetson J. Perianal streptococcal infection precipitating pustular psoriasis in an adult. JAAD Case Reports. 2016;2(4):281. doi:10.1016/j.jdcr.2016.05.010

29. Feily A, Namazi MR, Seifmanesh H. Generalized pustular psoriasis-like dermatophytosis due to Trichophyton rubrum. Acta Dermatovenerol Croat. 2011;19(3):209–211.

30. Yoneda K, Matsuoka-Shirahige Y, Demitsu T, Kubota Y. Pustular psoriasis precipitated by cytomegalovirus infection. Br J Dermatol. 2012;167(5):1186–1189. doi:10.1111/j.1365-2133.2012.11044.x

31. Ali FR, Green R, McMullen E, Motta L, Judge MR. Cutaneous cytomegalovirus complicating pustular psoriasis. Br J Dermatol. 2014;171(3):670–671. doi:10.1111/bjd.13026

32. Jiyad Z, Moriarty B, Creamer D, Higgins E. Generalized pustular psoriasis associated with Epstein-Barr virus. Clin Exp Dermatol. 2015;40(2):146–148. doi:10.1111/ced.12493

33. Sugiura K, Uchiyama R, Okuyama R, Akiyama M. Varicella zoster virus-associated generalized pustular psoriasis in a baby with heterozygous IL36RN mutation. J Am Acad Dermatol. 2014;71(5):e216–e218. doi:10.1016/j.jaad.2014.07.015

34. Kawamura A, Kinoshita MT, Suzuki H. Generalized pustular psoriasis with hypoparathyroidism. Eur J Dermatol. 1999;9(7):574–576.

35. George L, Mathews V, George B, Thomas M, Pulimood SA. Generalized pustular psoriasis following allogeneic stem cell transplantation. Clin Exp Dermatol. 2015;40(2):160–162. doi:10.1111/ced.12508

36. Deng W, Miao C, Zhang X. Abrupt generalized pustules in patients with rheumatoid arthritis and interstitial lung disease. J Dermatol. 2018;45(2):198–201. doi:10.1111/1346-8138.14126

37. 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;295(Suppl 1):S43–S54. doi:10.1007/s00403-002-0371-6

38. Kardaun SH, Kuiper H, Fidler V, Jonkman MF. The histopathological spectrum of acute generalized exanthematous pustulosis (AGEP) and its differentiation from generalized pustular psoriasis. J Cutan Pathol. 2010;37(12):1220–1229. doi:10.1111/j.1600-0560.2010.01612.x

39. Sugiura K. The genetic background of generalized pustular psoriasis: IL36RN mutations and CARD14 gain-of-function variants. J Dermatol Sci. 2014;74(3):187–192. doi:10.1016/j.jdermsci.2014.02.006

40. Diaz A. Deficiency of the Interleukin-36 Receptor Antagonist (DITRA) and generalized pustular psoriasis. In: Efthimiou P, editor. Auto-Inflammatory Syndromes: Pathophysiology, Diagnosis, and Management. Cham: Springer International Publishing; 2019:85–94.

41. Szatkowski J, Schwartz RA. Acute generalized exanthematous pustulosis (AGEP): a review and update. J Am Acad Dermatol. 2015;73(5):843–848. doi:10.1016/j.jaad.2015.07.017

42. Mahil SK, Barker JN, Capon F. Pustular forms of psoriasis related to autoinflammation. In: Hashkes PJ, Laxer RM, Simon A, editors. Textbook of Autoinflammation. Cham: Springer International Publishing; 2019:471–484.

43. Hussain S, Berki DM, Choon SE, et al. IL36RN mutations define a severe autoinflammatory phenotype of generalized pustular psoriasis. J Allergy Clin Immunol. 2015;135(4):1067–1070.e1069. doi:10.1016/j.jaci.2014.09.043

44. Mossner R, Wilsmann-Theis D, Oji V, et al. The genetic basis for most patients with pustular skin disease remains elusive. Br J Dermatol. 2018;178(3):740–748. doi:10.1111/bjd.15867

45. Marrakchi S, Guigue P, Renshaw BR, et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. The New England Journal of Medicine. 2011;365(7):620–628. doi:10.1056/NEJMoa1013068

46. Navarini AA, Simpson MA, Borradori L, Yawalkar N, Schlapbach C. Homozygous missense mutation in IL36RN in generalized pustular dermatosis with intraoral involvement compatible with both AGEP and generalized pustular psoriasis. JAMA Dermatol. 2015;151(4):452–453. doi:10.1001/jamadermatol.2014.3848

Creative Commons License © 2019 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.