The Skin as a Window into Primary Immune Deficiency Diseases: Atopic Dermatitis and Chronic Mucocutaneous Candidiasis

The Skin as a Window into Primary Immune Deficiency Diseases: Atopic Dermatitis and Chronic Mucocutaneous Candidiasis

Clinical Management Review The Skin as a Window into Primary Immune Deficiency Diseases: Atopic Dermatitis and Chronic Mucocutaneous Candidiasis Heat...

568KB Sizes 0 Downloads 35 Views

Clinical Management Review

The Skin as a Window into Primary Immune Deficiency Diseases: Atopic Dermatitis and Chronic Mucocutaneous Candidiasis Heather Lehman, MD, and Christopher Gordon, DO Buffalo, NY

INFORMATION FOR CATEGORY 1 CME CREDIT Credit can now be obtained, free for a limited time, by reading the review articles in this issue. Please note the following instructions. Method of Physician Participation in Learning Process: The core material for these activities can be read in this issue of the Journal or online at the JACI: In Practice Web site: www.jaci-inpractice.org/. The accompanying tests may only be submitted online at www.jaciinpractice.org/. Fax or other copies will not be accepted. Date of Original Release: March 1, 2019. Credit may be obtained for these courses until February 28, 2020 Copyright Statement: Copyright Ó 2019-2021. All rights reserved. Overall Purpose/Goal: To provide excellent reviews on key aspects of allergic disease to those who research, treat, or manage allergic disease. Target Audience: Physicians and researchers within the field of allergic disease. Accreditation/Provider Statements and Credit Designation: The American Academy of Allergy, Asthma & Immunology (AAAAI) is accredited by the Accreditation Council for Continuing Medical Education (ACCME) to provide continuing medical education for physicians. The AAAAI designates this journal-based CME activity for 1.00

Primary immune deficiency diseases characteristically present with recurrent, severe, or unusual infections. These infections may often involve the skin, with mucocutaneous candidal infections seen in a variety of different primary immune deficiencies. Primary immune deficiencies may also present with noninfectious cutaneous complications, of which eczema is the most common. In a patient with suspected primary immune deficiency, the presence of eczema or candidal skin infections offers critical information about the underlying immune defect, either the presence of atopy or defect in the TH17 pathway, Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY No funding was received for this work. Conflicts of interest: The authors declare that they have no relevant conflicts of interest. Received for publication September 6, 2018; revised November 19, 2018; accepted for publication November 25, 2018. Corresponding author: Heather Lehman, MD, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1001 Main Street, 5th floor, Buffalo, NY 14203. E-mail: [email protected]. 2213-2198 Ó 2018 American Academy of Allergy, Asthma & Immunology https://doi.org/10.1016/j.jaip.2018.11.026

788

AMA PRA Category 1 CreditÔ. Physicians should claim only the credit commensurate with the extent of their participation in the activity. List of Design Committee Members: Heather Lehman, MD, and Christopher Gordon, DO (authors); Robert S. Zeiger, MD, PhD (editor) Learning objectives: 1. To recognize the manifestations of primary immune deficiencies that commonly present with eczematous dermatitis. 2. To understand the inheritance patterns of genetic causes of chronic mucocutaneous candidiasis. 3. To differentiate the associated infections and noninfectious findings among the known genetic defects underlying chronic mucocutaneous candidiasis. 4. To describe an approach to diagnosis and treatment of chronic mucocutaneous candidiasis. Recognition of Commercial Support: This CME has not received external commercial support. Disclosure of Relevant Financial Relationships with Commercial Interests: The authors declare that they have no relevant conflicts of interest. R. S. Zeiger declares no relevant conflicts of interest.

respectively. These skin manifestations also are often early or heralding findings of the underlying immunologic disease. Therefore, awareness of associations between these skin findings and specific immune deficiencies may aide in the early detection and treatment of serious or life-threatening immunologic defects. This review specifically will focus on the primary immune deficiencies commonly associated with eczema or mucocutaneous candidiasis. Ó 2018 American Academy of Allergy, Asthma & Immunology (J Allergy Clin Immunol Pract 2019;7:788-98) Key words: Skin; PIDD; Atopic dermatitis; Chronic mucocutaneous candidiasis

Primary immune deficiency diseases (PIDDs) are a group of more than 350 genetic defects that lead to an aberrantly functioning immune system.1 Cutaneous manifestations are common in PIDD, affecting between 40% and 70% of patients with primary immunodeficiency.2,3 These skin findings in PIDD are often a presenting feature at the time of initial immune deficiency diagnosis in childhood.2-4 Skin infections are the most common cutaneous manifestation found in PIDD, and eczematous dermatitis is the

J ALLERGY CLIN IMMUNOL PRACT VOLUME 7, NUMBER 3

Abbreviations used AD-HIES- Autosomal dominant hyper-IgE syndrome AIRE- Autoimmune regulator APECED- Autoimmune polyendocrinopathy, candidiasis, and ectodermal dysplasia AR-HIES- Autosomal recessive hyper-IgE syndrome CARD9- Caspase recruitment domain 9 CID- Combined immune deficiency CMCC- Chronic mucocutaneous candidiasis CMV- Cytomegalovirus DGS- DiGeorge syndrome DOCK8- Dedicator of cytokinesis 8 FOXP3- Forkhead box protein 3 GlcNAc-6-P- N-Acetylglucosamine-6-phosphate GM-CSF- Granulocyte-macrophage-colony stimulating factor GOF- Gain-of-function GVHD- Graft-versus-host disease HDAC- Histone deacetylase HPV- Human papillomavirus HSCT- Hematopoietic stem cell transplant HSV- Herpes simplex virus IPEX- Immune dysregulation, polyendocrinopathy, and enteropathy, X-linked JAK- Janus kinase NF-kB- Nuclear factor-kB NK- Natural killer PGM3- Phosphoglucomutase 3 PIDD- Primary immune deficiency disease PRR- Pattern recognition receptor RORC- Retinoic aciderelated orphan receptor C SCID- Severe combined immunodeficiency SPINK5- Serine protease inhibitor of Kazal type 5 STAT- Signal transducer and activator factor of transcription TLR- Toll-like receptor WAS- Wiskott-Aldrich syndrome WASp- Wiskott-Aldrich syndrome protein

second most common skin finding reported in PIDD. This review will discuss 2 very important dermatologic manifestations of underlying immune deficiency: atopic dermatitis and mucocutaneous candidiasis.

ATOPIC DERMATITIS/ECZEMA IN PIDD Atopic or eczematous dermatitis is a common finding in the general population. It affects an estimated 10% to 20% of children in developed countries5 and 11% of children in the United States.6 It appears to be increasing in overall incidence and prevalence.7 In addition, higher rates of atopic dermatitis are observed in those affected by PIDD. In cohorts of pediatric PIDD populations, 13% to 22% have been affected by eczema.2-4 In 1 cohort, 75 patients with well-established severe atopic dermatitis underwent immune evaluation with 7.9% diagnosed with underlying PIDD.8 Eczema may be a presenting clinical manifestation of those immunodeficiencies presenting in the allergy clinic as well as the clinical immunology clinic. Several primary immunodeficiencies are also associated with the presence of infantile erythroderma, a severe exfoliative dermatitis affecting more than 90% of the body, which can be confused with a severe inflammatory eczema. Early diagnosis of immunodeficiency is crucial and

LEHMAN AND GORDON

789

therefore it is key to recognize the immunodeficiencies and additional clinical features that may present with atopic dermatitis or erythroderma.

Autosomal dominant hyper-IgE syndrome Early onset, severe eczema is a shared feature of both autosomal dominant and autosomal recessive forms of hyper-IgE syndrome (AD-HIES and AR-HIES). AD-HIES was originally described in 1966 as “Job syndrome.” It is a multisystem primary immunodeficiency syndrome of recurrent abscesses with Staphylococcus spp., sinopulmonary infections, and severe eczematous dermatitis.9,10 Mutations in the signal transducer and activator factor of transcription-3 (STAT3) were determined to be the cause of AD-HIES, first reported in 2007.11,12 The mutation is a dominant negative loss-of-function mutation, in which the abnormal gene product suppresses the function of the normal gene copy. STAT3 is a member of the Janus kinase (JAK)-STAT family of proteins. It is integral for the differentiation of naïve CD4þ T cells into TH17 cells. Therefore, patients with AD-HIES have a lack of TH17 cells leading to impaired IL-17/IL-22 production and signaling.13-15 Defective response to multiple cytokines, including IL-6 and IL-21, have also been reported.16,17 STAT3 is also key for maintenance of CD8þ T-cell memory. Likely related to this defect is an increased reactivation rate of varicella zoster virus and Epstein-Barr virus observed in AD-HIES.18,19 B-cell memory differentiation is also impacted and can result in derangements of specific antibody production, leading to the requirement of immunoglobulin replacement or prophylactic antibiotic use.20 Most patients with AD-HIES develop an early-onset (often in the first week of life) papulopustular eruption that begins on the face and scalp, but can become generalized.21 The rash morphology progresses to an eczematous dermatitis within the first year that can be lichenified.22,23 Colonization and infection with Staphylococcus aureus is strongly associated with this chronic dermatitis. Keratinocytes and bronchial epithelial cells normally produce antistaphylococcal factors, but require stimulation of TH17 cytokines to do so,24-26 which are impaired in patients with AD-HIES. In addition, approximately 80% of patients show some degree of impaired chemotactic effects of neutrophils.25,27,28 This may help explain the characteristic “cold” abscesses of some patients with STAT3 dominant negative mutation. Pneumonias can occur frequently and early in AD-HIES. In a French survey, the mean age at first infection was 10.5 months.29 Frequently, these infections are complicated by bronchiectasis, pneumatocele, and bronchopleural fistulae.10 Predisposition to other infections, including Pseudomonas, Aspergillus, and nontuberculous mycobacteria, has also been observed.29-32 Patients may actually report feeling well and are afebrile, despite serious infection. Patients with AD-HIES have elevated serum IgE levels with peak levels greater than 2000 IU/mL in the majority of cases, though normal IgE levels have been reported. Eosinophilia is observed in 93% of cases. Physical patient characteristics include facial asymmetry, large bulbous nose, prominent chin, higharched palate, and retained primary teeth. Other notable musculoskeletal features include scoliosis in 75% of patients.33 Vascular abnormalities such as Chiari I malformations are often detected. Recent studies have shown increased incidence of lacunar infarcts, coronary aneurysms, and coronary dilation.

790

LEHMAN AND GORDON

These coronary abnormalities have been associated with an increased risk of myocardial infarction.10,34 In contrast to AR-HIES, anaphylaxis is rare and food allergies are not a major concern in AD-HIES.35 Compared with patients with atopic dermatitis with similarly elevated IgE, both food allergy and food-induced anaphylaxis in AD-HIES were significantly lower (38% vs 58% and 8% vs 33%, respectively).36 Life expectancy is into the fifth or sixth decade with infection as the most common cause of death.10

DOCK8 deficiency The majority of cases of AR-HIES are attributed to dedicator of cytokinesis 8 (DOCK8) mutations.37 AR-HIES is similar in presentation to AD-HIES with elevated serum IgE levels, eosinophilia, eczematous dermatitis, and recurrent sinopulmonary and skin infections. Patients present with early onset dermatitis with classic atopic distribution and appearance.35 They often lack the pustular eruption observed in AD-HIES during the neonatal period.23 Another somewhat unique characteristic of DOCK8 deficiency is the increased frequency of multiple severe food allergies and asthma.35 DOCK8 deficiency can also be clinically distinguished from AD-HIES by severe cutaneous viral infections with herpes simplex virus (HSV), varicella zoster virus, molluscum contagiosum virus, and human papillomavirus (HPV).38 This susceptibility to these viral infections is likely due to associated impairment of natural killer (NK) cell development.38 CD4þ, CD8þ T cells, and plasmacytoid dendritic cells are also reduced in number. This results in profound decrease in production of IFN-a in response to viral infections through Toll-like receptor 9 (TLR9). A reduction in memory B cells has been found and is likely an explanation for the elevated risk of recurrent sinopulmonary infections observed due to specific antibody deficiency.38,39 Compared with AD-HIES, mucocutaneous candidiasis is observed less frequently in DOCK8 deficiency. However, this population is at higher risk of malignancy, particularly squamous cell carcinoma related to HPV infection sites and lymphoma. Later in life, malignancy becomes a frequent cause of death.10 Treatment of the dermatitis in AD-HIES and DOCK8 deficiency is quite difficult due to concurrent cutaneous viral infections and superimposed bacterial infections with S. aureus. Use of sodium hypochlorite baths and occasionally systemic antibiotics is recommended based on the literature published regarding the treatment of general atopic dermatitis population.40 Given reduced life expectancy, allogeneic hematopoietic stem cell transplant (HSCT) is often a pursued treatment.41 Cutaneous viral infections and dermatitis improve after transplant.42 Phosphoglucomutase 3 deficiency First described in 2014, phosphoglucomutase 3 (PGM3) deficiency results in a syndrome associated with atopic dermatitis, recurrent infections, and elevated IgE levels. It has been considered another cause of AR-HIES in addition to DOCK8 deficiency. PGM3 protein catalyzes the conversion of N-acetylglucosamine-6-phosphate (GlcNAc-6-P) into GlcNAc-1-P in the synthesis of uridine diphosphate-GlcNAc, a critical component of the glycosylation pathway. All patients described have had atopic dermatitis as a feature. These patients were also described as having other significant atopic diseases including asthma and

J ALLERGY CLIN IMMUNOL PRACT MARCH 2019

allergies. Similar to DOCK8 deficiency, a susceptibility to cutaneous bacterial and viral infections (HSV, cytomegalovirus [CMV]) was observed. Frequent sinopulmonary infections are reported with complications including pneumatocele development and bronchiectasis. One series of 3 patients with PGM3 deficiency had a more severe immunophenotype of T-B-NKþ severe combined immunodeficiency (SCID) along with skeletal anomalies in 2 of these 3 patients.43 Setting PGM3 deficiency apart from AD-HIES and DOCK8 deficiency is the observed neurologic impairment including developmental delay, ataxia, myoclonus, dysarthria, and sensorineural hearing loss. Another potentially unique cutaneous finding of PGM3 deficiency is leukocytoclastic vasculitis. Chronic mucocutaneous candidiasis can also develop, but with more variability than in AD-HIES.44,45

IPEX Immune dysregulation, polyendocrinopathy, and enteropathy, X-linked (IPEX) syndrome is an often fatal, X-linked recessive condition caused by loss-of-function mutations in forkhead box protein 3 (FOXP3). Foxp3 is fundamental to the function of CD4þ T-regulatory lymphocytes. Lack of functional regulatory T cells leads to immune dysregulation including autoimmune disease. Affecting males, IPEX typically presents during the first few months of life with a triad of severe dermatitis, insulin-dependent diabetes mellitus and other autoimmune phenomena, and intractable diarrhea.46 The primary skin manifestation is severe eczematous dermatitis. Other less common cutaneous manifestations include erythroderma, urticaria, psoriasiform dermatitis, pemphigoid nodularis, cheilitis, onychodystrophy, and alopecia universalis.46-48 IgE and eosinophil levels are often elevated along with increased incidence of food allergies.49,50 Antibodies to organ-specific antigens have been documented as likely explanation for autoimmune manifestations observed in IPEX syndrome.51 Impaired skin barrier and gut epithelium leads to infections. In addition, immunosuppressive drugs used to manage autoimmune disease can increase susceptibility to infection. In a cohort of 50 patients afflicted with IPEX, more than 50% experienced significant infections, including sepsis, meningitis, pneumonia, and osteomyelitis.52 The most commonly reported pathogens are Enterococcus spp., Staphylococcus spp., CMV, and Candida spp.53 Currently, the only potential curative treatment is hematopoietic stem cell transplant. A retrospective, multicenter review published in 2018 identified 96 patients with IPEX and demonstrated similar overall survival rates for individuals managed with chronic immune suppression compared with those who underwent HSCT, although there was greater mortality in the HSCT group in the peritransplant period.54 The disease progressed in individuals managed with chronic immune suppression, resulting in lower long-term, disease-free survival. Wiskott-Aldrich syndrome Wiskott-Aldrich syndrome (WAS) is an X-linked disorder due to mutations in the WAS gene, encoding the Wiskott-Aldrich syndrome protein (WASp). WASp, only expressed by hematopoietic cells, plays a key role for actin cytoskeleton remodeling that is vital for immunologic synapse formation between T cells and antigen-presenting cells.55 Mutations lead to

J ALLERGY CLIN IMMUNOL PRACT VOLUME 7, NUMBER 3

dysregulation of lymphocyte and NK cell migration and homing functions.56 The classic phenotype of Wiskott-Aldrich includes thrombocytopenia with small platelet size, eczema, recurrent infections, and increased risk of autoimmune disorders and malignancies. However, there is a broad spectrum of phenotypic presentations of WAS mutations, ranging from severe eczema with associated bacterial and viral infection with the risk of malignancy (B-cell lymphoma or leukemia), to a mild form of isolated thrombocytopenia (X-linked thrombocytopenia).57,58 The high variability in presentation is due to many possible mutations in the WAS gene resulting in variable effects on protein expression.58 Isolated Xlinked congenital neutropenia has been described due to gain-offunction (GOF) mutations in the GTPase-binding domain of WASp.59 Eczema affected 81% of patients with WAS in 1 large multiinstitutional case series, and typically presents during their first year of life.60 Immunologic findings include defective antibody responses to vaccines with normal IgG and normal-to-high IgE and IgA, low IgM levels, reduced NK cell cytotoxicity, decreased function of regulatory Treg cells, and impaired chemotaxis of phagocytic cells.49,61-63 Streptococcus pneumoniae, Neisseria meningitidis, and Haemophilus influenzae are frequent microbial agents isolated in WAS, manifesting as sinopulmonary infections, meningitis, and colitis. Recommended antimicrobial therapy is prophylactic trimethoprim-sulfamethoxazole and acyclovir. Some patients require immune globulin replacement at higher rates typically owing to an increased catabolic rate observed in WAS.64 HSCT can be completed in hopes of curative treatment. For autoimmune cytopenias, rituximab has been used with effectiveness. Those with increased bleeding risk can be treated with splenectomy; however, this can increase the risk of severe infection.65

DiGeorge syndrome DiGeorge syndrome (DGS) is a disorder characterized by a constellation of signs and symptoms associated with defective development of the pharyngeal pouch system. It is caused by a heterozygous chromosomal deletion at 22q11.2. Most patients with DGS have mildly-impaired T-cell production and normal humoral immunity. Other abnormalities include cleft palate, heart defects, dysmorphism, and hypoparathyroidism.66 Less than 1% of patients with 22q11.2 deletion have “complete DGS” with profoundly decreased T-cell counts consistent with T-BþNKþ SCID. Only a small subset of patients with complete DSG develop atypical complete DGS, with oligoclonal T-cell expansion resulting in inflammatory manifestations resembling those of Omenn syndrome. Skin findings include a papular or eczematous dermatitis that can be diffuse and erythrodermic.67-69 Omenn syndrome Hypomorphic mutations in various SCID genes may lead to Omenn syndrome due to the expansion of an oligoclonal, activated T-cell population. The classic presentation of Omenn syndrome is the development of exfoliative erythroderma associated with diffuse alopecia, lymphadenopathy, hepatosplenomegaly, recurrent severe opportunistic infections, eosinophilia, and failure to thrive in the early neonatal period.70 The findings are reminiscent of graft-versus-host disease (GVHD), although histology is not consistent with GVHD and there is no evidence of maternal engraftment of lymphocytes.71

LEHMAN AND GORDON

791

Although the initial cutaneous presentation of Omenn syndrome is most commonly described as erythroderma, it may present with a neonatal eczematous rash. Infants with classic SCID mutations may develop GVHD secondary to engraftment of transplacentally derived maternal T cells, or of T cells derived from transfusion of nonirradiated blood products. The GVHD that develops in these patients with SCID appears identical to the clinical picture of Omenn syndrome, including the cutaneous findings of erythroderma and alopecia.72,73

Comel-Netherton syndrome Comel-Netherton syndrome is a rare autosomal recessive disorder caused by mutations in the serine protease inhibitor of Kazal type 5 gene (SPINK5).74 The protein product, a serine protease inhibitor, is expressed on epithelial and mucosal surfaces. The initial skin abnormality in infants is described as generalized exfoliative erythroderma.75,76 Some affected infants are born with a tight, clear sheath covering their skin called a collodion membrane. Eventually, infants will develop a triad of ichthyosiform erythroderma, trichorrhexis invaginata (“bamboo hair”), and atopic disease, potentially including a superimposed atopic dermatitis rash. Infants are at high risk of life-threatening complications due to severe erythroderma. Because of the degradation of the epidermal barrier, many proinflammatory cytokines are increased including thymic stromal lymphopoietin, inducing a TH2 differentiation, which can predispose to the development of atopic manifestations.77 Therefore, older children with Comel-Netherton syndrome manifest a wide range of atopic diseases including asthma, atopic dermatitis, and allergic rhinitis with markedly elevated serum IgE levels and eosinophils. Angioedema and anaphylaxis to foods have also been reported.78 Children with Comel-Netherton syndrome may also show short stature, growth hormone deficiency, mild development delay, recurrent infections, and pancreatic insufficiency.79,80 There is no targeted therapy at present for Comel-Netherton syndrome, although kallikrein inhibitors may alleviate symptoms in animal models.81 Gene therapy using a lentiviral vector encoding SPINK5 is also being studied.82 EVALUATION FOR PRIMARY IMMUNODEFICIENCY IN CASES OF ATOPIC DERMATITIS Most often, severe atopic dermatitis is seen in patients without any underlying primary immune deficiency. Awareness of associations between skin findings and immunodeficiency diseases may lead to early detection and treatment of serious or lifethreatening immunologic defects in patients with PIDD presenting as eczema. Thorough medical history, family history, and physical examination may lead the clinician to suspect and investigate specific primary immune deficiencies. Genetic testing is required for most of the PIDDs with eczema, though protein detection by flow cytometry is available for WAS and DOCK8 deficiency. A clinical challenge remains to differentiate patients early in the course of HIES versus children with eczema and atopy. A scoring system was proposed in 1999 to identify patients with hyper-IgE syndromes.83 This was developed before the genetic causes of AD-HIES and AR-HIES were known. The initial scoring cutoff of 15 in this system has subsequently been shown to not be specific for HIES, with many patients with simple eczema and atopy reaching this score. However, it has since been

792

LEHMAN AND GORDON

shown that a score 40 along with low TH17 cell enumeration is sensitive for STAT3 dominant negative mutation.84 However, this scoring system may also pick up patients with DOCK8 deficiency in the group with score 40. A new “DOCK8 score” has been described to differentiate patients with DOCK8 deficiency versus STAT3 dominant negative mutation.85

CHRONIC MUCOCUTANEOUS CANDIDIASIS IN PIDDs Host defense against Candida and dermatophytes involves both the innate and cellular immune pathways. These fungi encounter the human host’s innate immune response as a first-line combatant to infection. Fungal pathogen-associated molecular patterns are recognized by pattern recognition receptors (PRRs) on phagocytes including TLRs and C-type lectin receptors. TLR2 and TLR4 recognize O-linked mannan on the fungal cell wall, leading to the activation of adaptor protein MyD88 and downstream activation of nuclear factor-kB (NF-kB). An example of C-type lectin receptor is Dectin-1, which recognizes b-glucans in the fungal cell wall and also leads to the activation of NF-kB through adaptor protein caspase recruitment domain 9 (CARD9). The downstream effect of NF-kB activation in phagocytes is transcription of proinflammatory cytokines that bind to the receptors of TH17 cells. Defects along these described pathways lead to primary immune deficiencies that are associated with chronic mucocutaneous candidiasis (Figure 1).86,87 It has long been recognized that PIDDs with major defects in cellular immunity, including SCID, complete DGS, and acquired immunodeficiency syndrome, may present with mucocutaneous candidal infections as one of many infectious susceptibilities. This is due to the lack of TH17 cells, whereas the presence of other opportunistic infections is due to the absence of other T-cell subsets. In contrast, patients with a phenotype of primary chronic mucocutaneous candidiasis (CMCC), with few other major infectious susceptibilities, have been identified in having one of several genetic defects leading to isolated TH17 impairment, including STAT1 GOF mutation, IL-17F or IL-17RA/RC mutations, ACT1 mutation, and retinoic aciderelated orphan receptor C (RORC) mutation. Complex syndromes with mucocutaneous candidiasis as a main infectious feature include autoimmune polyendocrinopathy, candidiasis, and ectodermal dysplasia (APECED) and autosomal dominant hyper-IgE syndrome. Table I summarized genetic defects with CMCC as a major feature. STAT1 GOF mutations The most common genetic mutation identified in patients with a CMCC phenotype is an autosomal dominant, GOF STAT1 mutation. STAT1 is a transcription factor important to TH1 cell development. It plays a pivotal role in cellular response to interferons and is central to many pathways involving hormones, interleukins, and growth factors.88 STAT1 GOF mutations have been found in either the coiled-coil domain or the DNA-binding domain of STAT1. These mutations result in prolonged phosphorylation of the STAT1 protein, leading to prolonged transcription factor activity in response to IFN-a/b, IFN-g, and IL-27, which suppress TH17 cell development, as well as in response to IL-6 and IL-21, which normally activate the STAT3 transcription factor rather than STAT1.89-92

J ALLERGY CLIN IMMUNOL PRACT MARCH 2019

GOF mutations in STAT1 result in a phenotype of mucocutaneous candidiasis and invasive fungal infections including coccidioidomycosis and histoplasmosis. In addition, patients are at increased susceptibility to bacterial sinopulmonary, mycobacterial, and herpesvirus family infections.93-95 Autoimmunity is a common finding in STAT1 GOF, with autoimmune thyroid disease being reported in 44% of 1 large patient cohort.96 Other autoimmune conditions reported include autoimmune cytopenias, autoimmune hepatitis, vitiligo, and alopecia.89,96 Malignancy as well as aortic and cerebral aneurysms has also been reported.89,97,98

Dectin-1 deficiency Dectin-1 is a PRR expressed on phagocytes that is able to bind to b-glucans on fungal cell walls. Dectin-1-mediated signaling promotes secretion of IL-2 and IL-23, which induces a tightly regulated TH17 differentiation and response.99,100 Homozygous mutations in CLEC7A (DECTIN1) resulting in Dectin-1 deficiency were reported in a single family in 2009.101 Three affected siblings homozygous for a Y238X polymorphism developed recurrent vulvovaginal candidiasis and onychomycosis, but were not predisposed to invasive candida infections. However, because the Y238X polymorphism is actually quite common in several populations and healthy homozygotes exist, the significance of Dectin-1 defects as a cause of CMCC is unclear.102,103 It has been suggested that this polymorphism of CLEC7A may confer the risk of CMCC in the presence of other modifying factors, rather than represent a monogenic cause of CMCC on its own.103 CARD9 deficiency CARD9 is an adaptor protein in the signaling pathway downstream of the Dectin-1 and Dectin-2 PRRs. Autosomal recessive mutations that result in CARD9 deficiency lead to defects in TH17 differentiation and impaired neutrophil killing of fungi.104 Production of IL-6 and IL-1b in response to Candida albicans requires CARD9. In contrast to Dectin-1 polymorphisms, patients with CARD9 deficiency appear susceptible to invasive fungal infection, particularly candidal meningitis,105,106 in addition to mucocutaneous candidiasis. These patients also develop deep dermatophytosis, in which dermatophytes may initially infect the epidermis and nails, but then invade the dermis and disseminate into underlying tissues including bone, lymph nodes, and brain.106 Although CARD9-deficient mice have been shown to be susceptible to Mycobacterium tuberculosis and Listeria monocytogenes,107,108 these infections have not been seen in patients with CARD9 deficiency to date. Gene mutations in IL-17RA, IL-17RC, IL-17F, and ACT1 associated with CMCC Autosomal recessive mutations in the IL-17RA, IL-17RC, and ACT1 genes and autosomal dominant mutations in the IL-17F gene have also been found to cause the TH17 dysfunction underlying CMCC in small numbers of patients.109,110 These mutations have been described to disrupt IL-17 signaling, including downstream activation of pathways such as NF-kB. IL17RA and IL-17F mutations were actually the first 2 genetic causes of CMCC disease identified, but all of the above defects have remained rare causes of CMCC overall.109 The phenotype of these defects is much more limited than that of STAT1 GOF. IL-17F and IL-17RC mutations have

J ALLERGY CLIN IMMUNOL PRACT VOLUME 7, NUMBER 3

LEHMAN AND GORDON

793

been shown to present with only isolated chronic mucocutaneous candidiasis, without other infectious risks. Patients with IL-17RA and ACT1 mutations show an additional predisposition to superficial staphylococcal infections, including dermatitis and blepharitis.109,111 The differences have been suggested to lie in the fact that IL-17RC and IL-17F mutations leave IL17E function intact, whereas IL-17RA and ACT1 mutations impact IL-17E function as well as IL-17A and IL-17F.110

Autoimmune polyendocrinopathy, candidiasis, and ectodermal dysplasia APECED is a monogenic disease caused by mutations in the autoimmune regulator (AIRE) gene. Its inheritance is typically autosomal recessive, but dominant negative mutations have been described as well. This condition was first described in 1929, when an association between hypoparathyroidism and chronic candidiasis was observed.113 The triad of CMCC, hypoparathyroidism, and adrenal insufficiency was reported first in 1946, with clinical diagnosis of classic APECED requiring 2 of these 3 findings.99,114 However, additional autoimmune manifestations in the skin and other organs are common and some of these may present before 2 manifestations of the classic triad for APECED have developed. Therefore, expanded diagnostic criteria have been proposed, adding an “adjunct triad” of urticarial eruption, intestinal dysfunction, and enamel hypoplasia. The presence of any 2 of the 6 features of the classic and adjunct triad should prompt diagnostic testing for APECED, including genetic testing and evaluation for IFN-u autoantibodies.115 In 1997, AIRE mutation was identified as the cause of APECED.116,117 AIRE is found in medullary thymic epithelial cells that fulfill an important role in self-antigen presentation to developing thymocytes and is required for deletion of autoreactive T cells. Dysfunctional or absent AIRE allows survival of these autoreactive T cells.118,119 The presence of various autoantibodies is hallmark to APECED, leading to the various clinical findings seen in the syndrome, including hypergonadotropic hypogonadism, type I diabetes, autoimmune thyroid diseases, and pituitary defects, along with features in the classic triad.120 More than 100 APECED-associated mutations have been reported in the AIRE gene. However, 2 mutations are found in the majority of patients.121 The cause of CMCC in APECED was unclear until in 2010 it was demonstrated to be caused by neutralizing autoantibodies against TH17 cytokines (IL-22, IL-17F, and IL-17A).122-124 Chronic Candida infection in early childhood is often the first sign of APECED. Early candidiasis often presents as persistent oral thrush. Oral infection can spread to the esophagus, causing an increased risk of squamous cell carcinoma.125 The nails are often affected, with the fingernails more commonly affected than toenails, likely due to spreading of infection from the mouth in infancy.126 Autoimmune hypoparathyroidism and adrenal failure (Addison’s disease) follow later in the disease course. The ectodermal manifestations associated with APECED include vitiligo, alopecia, keratoconjunctivitis, dental enamel hypoplasia, nail dystrophy, and tympanic membrane calcification.115,127

RORC mutation RORgt is the key transcription factor for the development of TH17 cells. Patients with RORgt deficiency, caused by autosomal recessive mutations in RORC, have been identified as a cause of chronic mucocutaneous candidiasis.112 In addition to the lack of IL-17A/F-producing cells, these patients surprisingly have diminished IFN-g production in response to mycobacteria, with associated impairments in gd-T cells, as well as absent NKT cells and mucosal associated invariant T cells. The poor IFN-g response leads to susceptibility to disseminated mycobacterial infection (bacille Calmette-Guérin infection in all patients described to date) in addition to CMCC.

Hyper-IgE syndromes and CMCC Susceptibility to mucocutaneous candidiasis, along with Aspergillus, Cryptococcus, and S. aureus infections, is seen in patients with AD-HIES due to autosomal dominant STAT3 lossof-function mutation. In 1 series, 85% of patients with AD-HIES due to STAT3 mutation exhibited CMCC.29 STAT3 is critical for RORgt induction in response to IL-6 and IL-23, leading to differentiation of TH17 lineage cells. Therefore, patients with STAT3 deficiency have decreased TH17 cells and resultant mucocutaneous candidiasis. Mucocutaneous candidiasis has been observed somewhat less often in the diseases underlying autosomal recessive hyper-IgE syndrome, DOCK8 deficiency, and PGM3 deficiency. In a series of 64 patients with DOCK8 deficiency, 64% had

FIGURE 1. Components of Candida albicans (mannans and b-glucan) are recognized by pattern recognition receptors such as Toll-like receptors (TLRs), Dectin-1 and Dectin-2, present on antigen presenting cells (APCs) and phagocytes. Dectin-1 and Dectin-2 signaling is transduced through caspase recruitment domain 9 (CARD9). This leads to transcription of proinflammatory cytokines IL-1b, IL-6, and IL-23. Cytokine-receptor interactions with IL-1R and IL-23R activate T cells via STAT3 and RORgt transcription factors to differentiate into TH17 lymphocytes. These cells produce IL-17 and IL-22 cytokines, which interact with receptors on various cell types, including neutrophils. Activation of T cells via STAT1 transcription factor alternatively impairs the differentiation of TH17 lymphocytes and promotes TH1 cell differentiation. Defects in molecules associated with chronic mucocutaneous candidiasis are labeled in red.

794

LEHMAN AND GORDON

J ALLERGY CLIN IMMUNOL PRACT MARCH 2019

TABLE I. Genetic mutations with chronic mucocutaneous candidiasis as a major feature Gene

Inheritance

AIRE

AD or AR

STAT3 (LOF)

STAT1 (GOF)

AD

AD

Infectious susceptibilities in addition to CMCC

Immunologic defect causing CMCC

Development of anti-IL-17A, IL-17F, and IL-22 neutralizing autoantibodies

Staphylococcal skin and lung infections Aspergillus lung infections

Compromised development of TH17 cells

Coccidioidomycosis

Compromised development of TH17 cells

Histoplasmosis Herpes simplex virus

IL-17F IL-17RA IL-17RC ACT1 CARD9

AD AR AR AR AR

CLEC7A/DECTIN1

AR

RORC

AR

Staphylococcal skin infections Staphylococcal skin infections Candida meningoencephalitis

Noninfectious complications

Hypoparathyroidism

Adrenal insufficiency Other autoimmune disorders Hypothyroidism Diabetes Gonadal dysfunction Alopecia areata Pernicious anemia Eczema Pneumatoceles Hyperextensible joints Osteoporosis and bone fractures Scoliosis Retention of primary teeth Cerebral and coronary aneurysms Autoimmunity Thyroid disease Cytopenias Hepatitis Vitiligo Alopecia Cerebral and aortic aneurysms

Impaired IL-17 signaling Impaired IL-17 signaling Impaired IL-17 signaling Impaired IL-17 signaling Impaired signaling downstream of Dectin-1 and Dectin-2 pattern recognition receptors

Deep dermatophytosis

Disseminated mycobacterial infection

Absent Dectin-1 pattern recognition receptor Absent TH17 cells

AD, Autosomal dominant; AR, autosomal recessive; CMCC, chronic mucocutaneous candidiasis.

mucocutaneous candidiasis,85 whereas 53% of another series of 136 DOCK8 patients reported CMCC.128 DOCK8 deficiency is typically characterized as a combined immune deficiency (CID), as it has effects on the function of T cells, B cells, and NK cells, and is associated with susceptibility to a relatively diverse group of pathogens.129 Impaired TH17 cell differentiation, with skewing instead towards TH2 cells, has been demonstrated in patients with DOCK8 deficiency,130 accounting for both predisposition to candidal infections and the presence of atopic disease. PGM3 deficiency has been reported in a small number of patients to date, with a phenotype of CID/hyper-IgE syndrome in some,44,45 versus SCID in others.43 One series of 9 patients described mucocutaneous candidiasis in two-thirds of cases,44 whereas another series of 8 patients reported no evidence of CMCC.45

Evaluation and treatment of CMCC The diagnosis of CMCC should be considered in patients with chronic or recurrent candidal or dermatophytic infections of the nails, skin, and mucous membranes as their major or exclusive infectious presentation. It is important to consider whether any other infectious or noninfectious complications are present that may point towards a specific genetic cause of CMCC. The 2015 Practice Parameter for the Diagnosis and Management of Primary Immunodeficiency recommends quantitative and functional evaluation of NK cells and assessment of lymphocyte stimulation to Candida in the evaluation of suspected CMCC.131 Decreased TH17 cell enumeration is found in certain causes of CMCC, including STAT3, CARD9, and STAT1 GOF, but TH17 cells are at normal levels with mutations in IL17RA and

J ALLERGY CLIN IMMUNOL PRACT VOLUME 7, NUMBER 3

IL17F.13,92,105,131 Abnormally prolonged STAT1 phosphorylation assays support the diagnosis of STAT1 GOF. Genetic testing is typically required to confirm a specific diagnosis in cases of CMCC. The treatment of isolated CMCC has traditionally focused on acute treatment and prophylaxis of candidal infections. Fluconazole is typically considered a first-line agent, followed by other azole antifungals. Resistance to at least 1 azole antifungal is common in patients with CMCC.132 Echinocandins and amphotericin B can be considered for patients with severe, treatment-refractory disease. Antifungal drugs have had modest success in treating dermatophyte infections. For superficial fungal infections, terbinafine or posaconazole has been recommended.133 Granulocyte-macrophage-colony stimulating factor (GMCSF) and granulocyte-colony stimulating factor have been proposed as second-line therapies for severe and invasive fungal infections. These products have been proposed as ways of augmenting TH17 cell differentiation,134,135 and GM-CSF increases Dectin-1 expression.136 However, results of these products in patients with severe CMCC have been variable.132,135 Management of patients with STAT1 GOF mutation has been challenging. Traditional immunosuppression to control autoimmunity further increases the risk of infections. HSCT has been attempted in cases of STAT1 GOF mutation with severe symptoms. However, the nature of STAT1 GOF mutations amplifies the risk for uncontrolled infections and graft-versushost disease after HSCT, resulting in the risk of secondary graft failure and mortality.137-139 Recently, novel biologic therapy with ruxolitinib, a JAK1 and JAK2 inhibitor, has shown promise in the treatment of STAT1 disease. The initial report of ruxolitinib use in CMCC described complete resolution of alopecia areata and oral candidiasis in a patient with STAT1 GOF, with return of the oral candidiasis 2 weeks after discontinuation of ruxolitinib.140 A second case experienced significant improvement, but not resolution, of ulcerative mucositis due to C. albicans with the use of ruxolitinib.141 A third case showed significant effects of ruxolitinib, with control of autoimmune cytopenias, cured mucocutaneous candidiasis and diarrhea, and improvement of pulmonary function.88 After these 3 cases of success with ruxolitinib, there was a report of ruxolitinib treatment failure in 2 further patients with STAT1 GOF mutation.142 A case series of jakinib use in 11 patients with STAT1 GOF (along with several patients with STAT3 GOF mutations) across multiple different institutions is in press and reports improvement in 10 of 11 patients with STAT1 GOF, with the case of treatment failure occurring in a patient with disseminated coccidioidomycosis that was active at the time ruxolitinib was initiated.143 The authors infer that initiation of jakinibs late in the course of disease may be too late to control severe manifestations, and they recommend consideration of jakinib therapy in STAT1 GOF or STAT3 GOF when immune dysregulation, organ-specific autoimmunity, chronic autoimmune cytopenias, lymphoproliferation, refractory CMCC, or recurrent severe infections are present. Histone deacetylase (HDAC) inhibitors have also been proposed as potential treatment for CMCC in STAT1 GOF mutations. The altered gene transcription of STAT1 and STAT3 seen in STAT1 GOF mutation has been demonstrated to be modified by inhibition of HDAC1 and HDAC2.91 The use of selective HDAC inhibitors, Entinostat and RGFP966, has been associated with decreased STAT1 phosphorylation in in vitro

LEHMAN AND GORDON

795

studies of peripheral blood mononuclear cells from patients with STAT1 GOF.144

CONCLUSIONS Cutaneous manifestations are commonly observed in PIDD and are most often a presenting feature at the time of diagnosis. Eczematous dermatitis and mucocutaneous candidiasis are 2 key manifestations of underlying immune deficiency as discussed in this review. Advances in molecular diagnostic techniques have accelerated the discovery of the biologic basis of disease. This understanding of underlying disease associations has aided in better establishment of phenotypic relationships of PIDD and associated skin and systemic manifestations. REFERENCES 1. Picard C, Bobby Gaspar H, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, et al. International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on inborn errors of immunity. J Clin Immunol 2018;38:96-128. 2. Al-Herz W, Nanda A. Skin manifestations in primary immunodeficient children. Pediatr Dermatol 2011;28:494-501. 3. Berron-Ruiz A, Berron-Perez R, Ruiz-Maldonado R. Cutaneous markers of primary immunodeficiency diseases in children. Pediatr Dermatol 2000;17: 91-6. 4. Moin A, Farhoudi A, Moin M, Pourpak Z, Bazargan N. Cutaneous manifestations of primary immunodeficiency diseases in children. Iran J Allergy Asthma Immunol 2006;5:121-6. 5. Williams H, Robertson C, Stewart A, Ait-Khaled N, Anabwani G, Anderson R, et al. Worldwide variations in the prevalence of symptoms of atopic eczema in the International Study of Asthma and Allergies in Childhood. J Allergy Clin Immunol 1999;103(Pt 1):125-38. 6. Shaw TE, Currie GP, Koudelka CW, Simpson EL. Eczema prevalence in the United States: data from the 2003 National Survey of Children’s Health. J Invest Dermatol 2011;131:67-73. 7. Deckers IA, McLean S, Linssen S, Mommers M, van Schayck CP, Sheikh A. Investigating international time trends in the incidence and prevalence of atopic eczema 1990-2010: a systematic review of epidemiological studies. PloS One 2012;7:e39803. 8. Aghamohammadi A, Moghaddam ZG, Abolhassani H, Hallaji Z, Mortazavi H, Pourhamdi S, et al. Investigation of underlying primary immunodeficiencies in patients with severe atopic dermatitis. Allergol Immunopathol (Madr) 2014;42: 336-41. 9. Davis SD, Schaller J, Wedgwood RJ. Job’s syndrome. Recurrent, “cold”, staphylococcal abscesses. Lancet 1966;1:1013-5. 10. Freeman AF, Kleiner DE, Nadiminti H, Davis J, Quezado M, Anderson V, et al. Causes of death in hyper-IgE syndrome. J Allergy Clin Immunol 2007; 119:1234-40. 11. Holland SM, DeLeo FR, Elloumi HZ, Hsu AP, Uzel G, Brodsky N, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med 2007;357: 1608-19. 12. Minegishi Y, Saito M, Tsuchiya S, Tsuge I, Takada H, Hara T, et al. Dominant-negative mutations in the DNA-binding domain of STAT3 cause hyperIgE syndrome. Nature 2007;448:1058-62. 13. Ma CS, Chew GY, Simpson N, Priyadarshi A, Wong M, Grimbacher B, et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med 2008;205:1551-7. 14. Milner JD, Brenchley JM, Laurence A, Freeman AF, Hill BJ, Elias KM, et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature 2008;452:773-6. 15. Speckmann C, Enders A, Woellner C, Thiel D, Rensing-Ehl A, Schlesier M, et al. Reduced memory B cells in patients with hyper IgE syndrome. Clin Immunol 2008;129:448-54. 16. Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol 2007;178:2623-9. 17. O’Shea JJ, Murray PJ. Cytokine signaling modules in inflammatory responses. Immunity 2008;28:477-87. 18. Ives ML, Ma CS, Palendira U, Chan A, Bustamante J, Boisson-Dupuis S, et al. Signal transducer and activator of transcription 3 (STAT3) mutations underlying autosomal dominant hyper-IgE syndrome impair human CD8(þ) T-cell memory formation and function. J Allergy Clin Immunol 2013;132:400-411. e9.

796

LEHMAN AND GORDON

19. Siegel AM, Heimall J, Freeman AF, Hsu AP, Brittain E, Brenchley JM, et al. A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory. Immunity 2011;35:806-18. 20. Avery DT, Deenick EK, Ma CS, Suryani S, Simpson N, Chew GY, et al. B cell-intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med 2010;207: 155-71. 21. Olaiwan A, Chandesris MO, Fraitag S, Lortholary O, Hermine O, Fischer A, et al. Cutaneous findings in sporadic and familial autosomal dominant hyperIgE syndrome: a retrospective, single-center study of 21 patients diagnosed using molecular analysis. J Am Acad Dermatol 2011;65:1167-72. 22. Cho C, Ferdman RM, Church JA, Ong PY. Skin-deep clues to a complex disease. Ann Allergy Asthma Immunol 2010;104:93-4. 23. Eberting CL, Davis J, Puck JM, Holland SM, Turner ML. Dermatitis and the newborn rash of hyper-IgE syndrome. Arch Dermatol 2004;140:1119-25. 24. Minegishi Y, Saito M, Nagasawa M, Takada H, Hara T, Tsuchiya S, et al. Molecular explanation for the contradiction between systemic Th17 defect and localized bacterial infection in hyper-IgE syndrome. J Exp Med 2009;206: 1291-301. 25. Mogensen TH. STAT3 and the hyper-IgE syndrome: clinical presentation, genetic origin, pathogenesis, novel findings and remaining uncertainties. JAKSTAT 2013;2:e23435. 26. Ochs HD, Oukka M, Torgerson TR. TH17 cells and regulatory T cells in primary immunodeficiency diseases. J Allergy Clin Immunol 2009;123: 977-83. quiz 84-5. 27. Hill HR, Ochs HD, Quie PG, Clark RA, Pabst HF, Klebanoff SJ, et al. Defect in neutrophil granulocyte chemotaxis in Job’s syndrome of recurrent “cold” staphylococcal abscesses. Lancet 1974;2:617-9. 28. Van Scoy RE, Hill HR, Ritts RE, Quie PG. Familial neutrophil chemotaxis defect, recurrent bacterial infections, mucocutaneous candidiasis, and hyperimmunoglobulinemia E. Ann Int Med 1975;82:766-71. 29. Chandesris MO, Melki I, Natividad A, Puel A, Fieschi C, Yun L, et al. Autosomal dominant STAT3 deficiency and hyper-IgE syndrome: molecular, cellular, and clinical features from a French national survey. Medicine (Baltimore) 2012;91:e1-19. 30. Freeman AF, Davis J, Anderson VL, Barson W, Darnell DN, Puck JM, et al. Pneumocystis jiroveci infection in patients with hyper-immunoglobulin E syndrome. Pediatrics 2006;118:e1271-5. 31. Gernez Y, Freeman AF, Holland SM, Garabedian E, Patel NC, Puck JM, et al. Autosomal dominant hyper-IgE syndrome in the USIDNET Registry. J Allergy Clin Immunol Pract 2018;6:996-1001. 32. Melia E, Freeman AF, Shea YR, Hsu AP, Holland SM, Olivier KN. Pulmonary nontuberculous mycobacterial infections in hyper-IgE syndrome. J Allergy Clin Immunol 2009;124:617-8. 33. Grimbacher B, Holland SM, Gallin JI, Greenberg F, Hill SC, Malech HL, et al. Hyper-IgE syndrome with recurrent infections—an autosomal dominant multisystem disorder. N Engl J Med 1999;340:692-702. 34. Freeman AF, Avila EM, Shaw PA, Davis J, Hsu AP, Welch P, et al. Coronary artery abnormalities in hyper-IgE syndrome. J Clin Immunol 2011;31:338-45. 35. Chu EY, Freeman AF, Jing H, Cowen EW, Davis J, Su HC, et al. Cutaneous manifestations of DOCK8 deficiency syndrome. Arch Dermatol 2012;148: 79-84. 36. Siegel AM, Stone KD, Cruse G, Lawrence MG, Olivera A, Jung MY, et al. Diminished allergic disease in patients with STAT3 mutations reveals a role for STAT3 signaling in mast cell degranulation. J Allergy Clin Immunol 2013; 132:1388-96. 37. Engelhardt KR, McGhee S, Winkler S, Sassi A, Woellner C, Lopez-Herrera G, et al. Large deletions and point mutations involving the dedicator of cytokinesis 8 (DOCK8) in the autosomal-recessive form of hyper-IgE syndrome. J Allergy Clin Immunol 2009;124:1289-1302.e4. 38. Al-Zahrani D, Raddadi A, Massaad M, Keles S, Jabara HH, Chatila TA, et al. Successful interferon-alpha 2b therapy for unremitting warts in a patient with DOCK8 deficiency. Clin Immunol 2014;153:104-8. 39. Caracciolo S, Moratto D, Giacomelli M, Negri S, Lougaris V, Porta F, et al. Expansion of CCR4þ activated T cells is associated with memory B cell reduction in DOCK8-deficient patients. Clini Immunol 2014;152:164-70. 40. Bath-Hextall FJ, Birnie AJ, Ravenscroft JC, Williams HC. Interventions to reduce Staphylococcus aureus in the management of atopic eczema: an updated Cochrane review. Br J Dermatol 2010;163:12-26. 41. Boztug H, Karitnig-Weiss C, Ausserer B, Renner ED, Albert MH, SawalleBelohradsky J, et al. Clinical and immunological correction of DOCK8 deficiency by allogeneic hematopoietic stem cell transplantation following a

J ALLERGY CLIN IMMUNOL PRACT MARCH 2019

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60. 61.

reduced toxicity conditioning regimen. Pediatr Hematol Oncol 2012;29: 585-94. Ghosh S, Schuster FR, Adams O, Babor F, Borkhardt A, Comoli P, et al. Haploidentical stem cell transplantation in DOCK8 deficiency—successful control of pre-existing severe viremia with a TCRass/CD19-depleted graft and antiviral treatment. Clin Immunol 2014;152:111-4. Stray-Pedersen A, Backe PH, Sorte HS, Morkrid L, Chokshi NY, Erichsen HC, et al. PGM3 mutations cause a congenital disorder of glycosylation with severe immunodeficiency and skeletal dysplasia. Am J Hum Genet 2014;95:96-107. Sassi A, Lazaroski S, Wu G, Haslam SM, Fliegauf M, Mellouli F, et al. Hypomorphic homozygous mutations in phosphoglucomutase 3 (PGM3) impair immunity and increase serum IgE levels. J Allergy Clin Immunol 2014; 133:1410-1419.e1-1419.e13. Zhang Y, Yu X, Ichikawa M, Lyons JJ, Datta S, Lamborn IT, et al. Autosomal recessive phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment. J Allergy Clin Immunol 2014;133:1400-9.e1-5. Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol 2012;3:211. McGinness JL, Bivens MM, Greer KE, Patterson JW, Saulsbury FT. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) associated with pemphigoid nodularis: a case report and review of the literature. J Am Acad Dermatol 2006;55:143-8. Moraes-Vasconcelos D, Costa-Carvalho BT, Torgerson TR, Ochs HD. Primary immune deficiency disorders presenting as autoimmune diseases: IPEX and APECED. J Clin Immunol 2008;28(Suppl 1):S11-9. Ozcan E, Notarangelo LD, Geha RS. Primary immune deficiencies with aberrant IgE production. J Allergy Clin Immunol 2008;122:1054-62. quiz 63-4. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, et al. Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology 2007; 132:1705-17. Tsuda M, Torgerson TR, Selmi C, Gambineri E, Carneiro-Sampaio M, Mannurita SC, et al. The spectrum of autoantibodies in IPEX syndrome is broad and includes anti-mitochondrial autoantibodies. J Autoimmun 2010;35: 265-8. Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked: forkhead box protein 3 mutations and lack of regulatory T cells. J Allergy Clin Immunol 2007;120:744-50. Gambineri E, Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T-cell homeostasis. Curr Opin Rheumatol 2003;15:430-5. Barzaghi F, Amaya Hernandez LC, Neven B, Ricci S, Kucuk ZY, Bleesing JJ, et al. Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: an international multicenter retrospective study. J Allergy Clin Immunol 2018;141:1036-1049.e5. Malinova D, Fritzsche M, Nowosad CR, Armer H, Munro PM, Blundell MP, et al. WASp-dependent actin cytoskeleton stability at the dendritic cell immunological synapse is required for extensive, functional T cell contacts. J Leukoc Biol 2016;99:699-710. Burns S, Thrasher AJ, Blundell MP, Machesky L, Jones GE. Configuration of human dendritic cell cytoskeleton by Rho GTPases, the WAS protein, and differentiation. Blood 2001;98:1142-9. Albert MH, Bittner TC, Nonoyama S, Notarangelo LD, Burns S, Imai K, et al. X-linked thrombocytopenia (XLT) due to WAS mutations: clinical characteristics, long-term outcome, and treatment options. Blood 2010;115:3231-8. Jin Y, Mazza C, Christie JR, Giliani S, Fiorini M, Mella P, et al. Mutations of the Wiskott-Aldrich syndrome protein (WASP): hotspots, effect on transcription, and translation and phenotype/genotype correlation. Blood 2004;104: 4010-9. Devriendt K, Kim AS, Mathijs G, Frints SG, Schwartz M, Van Den Oord JJ, et al. Constitutively activating mutation in WASP causes X-linked severe congenital neutropenia. Nat Genet 2001;27:313-7. Sullivan KE, Mullen CA, Blaese RM, Winkelstein JA. A multi-institutional survey of the Wiskott-Aldrich syndrome. J Pediatr 1994;125:876-85. Gismondi A, Cifaldi L, Mazza C, Giliani S, Parolini S, Morrone S, et al. Impaired natural and CD16-mediated NK cell cytotoxicity in patients with WAS and XLT: ability of IL-2 to correct NK cell functional defect. Blood 2004;104:436-43.

J ALLERGY CLIN IMMUNOL PRACT VOLUME 7, NUMBER 3

62. Humblet-Baron S, Sather B, Anover S, Becker-Herman S, Kasprowicz DJ, Khim S, et al. Wiskott-Aldrich syndrome protein is required for regulatory T cell homeostasis. J Clin Invest 2007;117:407-18. 63. Maillard MH, Cotta-de-Almeida V, Takeshima F, Nguyen DD, Michetti P, Nagler C, et al. The Wiskott-Aldrich syndrome protein is required for the function of CD4(þ)CD25(þ)Foxp3(þ) regulatory T cells. J Exp Med 2007; 204:381-91. 64. Blaese RM, Strober W, Levy AL, Waldmann TA. Hypercatabolism of IgG, IgA, IgM, and albumin in the Wiskott-Aldrich syndrome. A unique disorder of serum protein metabolism. J Clin Invest 1971;50:2331-8. 65. Mullen CA, Anderson KD, Blaese RM. Splenectomy and/or bone marrow transplantation in the management of the Wiskott-Aldrich syndrome: longterm follow-up of 62 cases. Blood 1993;82:2961-6. 66. Bassett AS, McDonald-McGinn DM, Devriendt K, Digilio MC, Goldenberg P, Habel A, et al. Practical guidelines for managing patients with 22q11.2 deletion syndrome. J Pediatr 2011;159:332-339.e1. 67. Stone CA Jr, Markert ML, Abraham RS, Norton A. A case of atypical, complete DiGeorge syndrome without 22q11 mutation. Ann Allergy Asthma Immunol 2017;118:640-642.e2. 68. Selim MA, Markert ML, Burchette JL, Herman CM, Turner JW. The cutaneous manifestations of atypical complete DiGeorge syndrome: a histopathologic and immunohistochemical study. J Cutan Pathol 2008;35:380-5. 69. Vu QV, Wada T, Toma T, Tajima H, Maeda M, Tanaka R, et al. Clinical and immunophenotypic features of atypical complete DiGeorge syndrome. Pediatr Int 2013;55:2-6. 70. de Saint-Basile G, Le Deist F, de Villartay JP, Cerf-Bensussan N, Journet O, Brousse N, et al. Restricted heterogeneity of T lymphocytes in combined immunodeficiency with hypereosinophilia (Omenn’s syndrome). J Clin Invest 1991;87:1352-9. 71. Shearer WT, Dunn E, Notarangelo LD, Dvorak CC, Puck JM, Logan BR, et al. Establishing diagnostic criteria for severe combined immunodeficiency disease (SCID), leaky SCID, and Omenn syndrome: the Primary Immune Deficiency Treatment Consortium experience. J Allergy Clin Immunol 2014;133:1092-8. 72. Anderson KC, Weinstein HJ. Transfusion-associated graft-versus-host disease. N Engl J Med 1990;323(5):315-21. 73. Le Deist F, Raffoux C, Griscelli C, Fischer A. Graft vs graft reaction resulting in the elimination of maternal cells in a SCID patient with maternofetal GVHd after an HLA identical bone marrow transplantation. J Immunol 1987;138: 423-7. 74. Chavanas S, Bodemer C, Rochat A, Hamel-Teillac D, Ali M, Irvine AD, et al. Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat Genet 2000;25:141-2. 75. Hoeger PH, Harper JI. Neonatal erythroderma: differential diagnosis and management of the “red baby”. Arch Dis Child 1998;79:186-91. 76. Sehgal VN, Srivastava G, Sardana K. Erythroderma/exfoliative dermatitis: a synopsis. Int J Dermatol 2004;43:39-47. 77. Furio L, Hovnanian A. Netherton syndrome: defective kallikrein inhibition in the skin leads to skin inflammation and allergy. Biol Chem 2014;395: 945-58. 78. Hannula-Jouppi K, Laasanen SL, Heikkila H, Tuomiranta M, Tuomi ML, Hilvo S, et al. IgE allergen component-based profiling and atopic manifestations in patients with Netherton syndrome. J Allergy Clin Immunol 2014;134: 985-8. 79. Aydin BK, Bas F, Tamay Z, Kilic G, Suleyman A, Bundak R, et al. Netherton syndrome associated with growth hormone deficiency. Pediatr Dermatol 2014; 31:90-4. 80. Machet P, Bodemer C, Lorette G, Fraitag S, Raynaud M, Willot S, et al. Exocrine pancreatic insufficiency in a child with Netherton syndrome. Eur J Dermatol 2016;26:311-2. 81. Tan X, Soualmia F, Furio L, Renard JF, Kempen I, Qin L, et al. Toward the first class of suicide inhibitors of kallikreins involved in skin diseases. J Med Chem 2015;58:598-612. 82. Di WL, Mellerio JE, Bernadis C, Harper J, Abdul-Wahab A, Ghani S, et al. Phase I study protocol for ex vivo lentiviral gene therapy for the inherited skin disease, Netherton syndrome. Hum Gene Ther Clin Dev 2013;24: 182-90. 83. Grimbacher B, Schaffer AA, Holland SM, Davis J, Gallin JI, Malech HL, et al. Genetic linkage of hyper-IgE syndrome to chromosome 4. Am J Hum Genet 1999;65:735-44. 84. Schimke LF, Sawalle-Belohradsky J, Roesler J, Wollenberg A, Rack A, Borte M, et al. Diagnostic approach to the hyper-IgE syndromes: immunologic and clinical key findings to differentiate hyper-IgE syndromes from atopic dermatitis. J Allergy Clin Immunol 2010;126:611-617.e1.

LEHMAN AND GORDON

797

85. Engelhardt KR, Gertz ME, Keles S, Schaffer AA, Sigmund EC, Glocker C, et al. The extended clinical phenotype of 64 patients with dedicator of cytokinesis 8 deficiency. J Allergy Clin Immunol 2015;136:402-12. 86. McDonald DR. TH17 deficiency in human disease. J Allergy Clin Immunol 2012;129:1429-35. quiz 36-7. 87. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 2005;6:1133-41. 88. Weinacht KG, Charbonnier LM, Alroqi F, Plant A, Qiao Q, Wu H, et al. Ruxolitinib reverses dysregulated T helper cell responses and controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation. J Allergy Clin Immunol 2017;139: 1629-1640.e2. 89. van de Veerdonk FL, Plantinga TS, Hoischen A, Smeekens SP, Joosten LA, Gilissen C, et al. STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med 2011;365:54-61. 90. Yamazaki Y, Yamada M, Kawai T, Morio T, Onodera M, Ueki M, et al. Two novel gain-of-function mutations of STAT1 responsible for chronic mucocutaneous candidiasis disease: impaired production of IL-17A and IL-22, and the presence of anti-IL-17F autoantibody. J Immunol 2014;193:4880-7. 91. Zheng J, van de Veerdonk FL, Crossland KL, Smeekens SP, Chan CM, Al Shehri T, et al. Gain-of-function STAT1 mutations impair STAT3 activity in patients with chronic mucocutaneous candidiasis (CMC). Eur J Immunol 2015; 45:2834-46. 92. Liu L, Okada S, Kong XF, Kreins AY, Cypowyj S, Abhyankar A, et al. Gainof-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med 2011;208:1635-48. 93. Sampaio EP, Bax HI, Hsu AP, Kristosturyan E, Pechacek J, Chandrasekaran P, et al. A novel STAT1 mutation associated with disseminated mycobacterial disease. J Clin Immunol 2012;32:681-9. 94. Toth B, Mehes L, Tasko S, Szalai Z, Tulassay Z, Cypowyj S, et al. Herpes in STAT1 gain-of-function mutation [corrected]. Lancet 2012;379:2500. 95. Uzel G, Sampaio EP, Lawrence MG, Hsu AP, Hackett M, Dorsey MJ, et al. Dominant gain-of-function STAT1 mutations in FOXP3 wild-type immune dysregulation-polyendocrinopathy-enteropathy-X-linked-like syndrome. J Allergy Clin Immunol 2013;131:1611-23. 96. Depner M, Fuchs S, Raabe J, Frede N, Glocker C, Doffinger R, et al. The extended clinical phenotype of 26 patients with chronic mucocutaneous candidiasis due to gain-of-function mutations in STAT1. J Clin Immunol 2016; 36:73-84. 97. Frans G, Moens L, Schaballie H, Van Eyck L, Borgers H, Wuyts M, et al. Gain-of-function mutations in signal transducer and activator of transcription 1 (STAT1): chronic mucocutaneous candidiasis accompanied by enamel defects and delayed dental shedding. J Allergy Clin Immunol 2014;134:1209-1213.e6. 98. Tanimura M, Dohi K, Hirayama M, Sato Y, Sugiura E, Nakajima H, et al. Recurrent inflammatory aortic aneurysms in chronic mucocutaneous candidiasis with a gain-of-function STAT1 mutation. Int J Cardiol 2015;196:88-90. 99. Kisand K, Peterson P. Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy and other primary immunodeficiency diseases help to resolve the nature of protective immunity against chronic mucocutaneous candidiasis. Curr Opin Pediatr 2013;25:715-21. 100. Thakur R, Anand R, Tiwari S, Singh AP, Tiwary BN, Shankar J. Cytokines induce effector T-helper cells during invasive aspergillosis; what we have learned about T-helper cells? Front Microbiol 2015;6:429. 101. Ferwerda B, Ferwerda G, Plantinga TS, Willment JA, van Spriel AB, Venselaar H, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 2009;361:1760-7. 102. Marodi L, Erdos M. Dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 2010;362:367-8. 103. Engelhardt KR, Grimbacher B. Mendelian traits causing susceptibility to mucocutaneous fungal infections in human subjects. J Allergy Clin Immunol 2012;129:294-305. quiz 6-7. 104. Drewniak A, Gazendam RP, Tool AT, van Houdt M, Jansen MH, van Hamme JL, et al. Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency. Blood 2013;121:2385-92. 105. Glocker EO, Hennigs A, Nabavi M, Schaffer AA, Woellner C, Salzer U, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med 2009;361:1727-35. 106. Lanternier F, Pathan S, Vincent QB, Liu L, Cypowyj S, Prando C, et al. Deep dermatophytosis and inherited CARD9 deficiency. N Engl J Med 2013;369: 1704-14. 107. Dorhoi A, Desel C, Yeremeev V, Pradl L, Brinkmann V, Mollenkopf HJ, et al. The adaptor molecule CARD9 is essential for tuberculosis control. J Exp Med 2010;207:777-92.

798

LEHMAN AND GORDON

108. Hsu YM, Zhang Y, You Y, Wang D, Li H, Duramad O, et al. The adaptor protein CARD9 is required for innate immune responses to intracellular pathogens. Nat Immunol 2007;8:198-205. 109. Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L, Lim HK, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 2011;332:65-8. 110. Ling Y, Cypowyj S, Aytekin C, Galicchio M, Camcioglu Y, Nepesov S, et al. Inherited IL-17RC deficiency in patients with chronic mucocutaneous candidiasis. J Exp Med 2015;212:619-31. 111. Boisson B, Wang C, Pedergnana V, Wu L, Cypowyj S, Rybojad M, et al. An ACT1 mutation selectively abolishes interleukin-17 responses in humans with chronic mucocutaneous candidiasis. Immunity 2013;39:676-86. 112. Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, et al. IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science 2015;349: 606-13. 113. Thorpe ES, Handley HE. Chronic tetany and chronic mycelial stomatitis in a child aged four and one-half years. Am J Dis Child 1929;38:328-38. 114. Leonard MF. Chronic idiopathic hypoparathyroidism with superimposed Addison’s disease in a child. J Clin Endocrinol Metabol 1946;6:493-506. 115. Ferre EM, Rose SR, Rosenzweig SD, Burbelo PD, Romito KR, Niemela JE, et al. Redefined clinical features and diagnostic criteria in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. JCI Insight 2016;1: e88782. 116. Finnish-German AC. An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat Genet 1997; 17:399-403. 117. Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, et al. Positional cloning of the APECED gene. Nat Genet 1997;17:393-8. 118. Gardner JM, Devoss JJ, Friedman RS, Wong DJ, Tan YX, Zhou X, et al. Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 2008;321:843-7. 119. Kyewski B, Derbinski J. Self-representation in the thymus: an extended view. Nature Rev Immunol 2004;4:688-98. 120. Perheentupa J. Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. The J Clin Endocrinol Metab 2006;91:2843-50. 121. Peterson P, Peltonen L. Autoimmune polyendocrinopathy syndrome type 1 (APS1) and AIRE gene: new views on molecular basis of autoimmunity. J Autoimmun 2005;25(Suppl):49-55. 122. Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, Kisand KV, et al. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. J Exp Med 2010;207: 299-308. 123. Puel A, Doffinger R, Natividad A, Chrabieh M, Barcenas-Morales G, Picard C, et al. Autoantibodies against IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. J Exp Med 2010;207:291-7. 124. Kisand K, Lilic D, Casanova JL, Peterson P, Meager A, Willcox N. Mucocutaneous candidiasis and autoimmunity against cytokines in APECED and thymoma patients: clinical and pathogenetic implications. Eur J Immunol 2011;41:1517-27. 125. Rautemaa R, Hietanen J, Niissalo S, Pirinen S, Perheentupa J. Oral and oesophageal squamous cell carcinoma—a complication or component of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED, APS-I). Oral Oncol 2007;43:607-13. 126. Collins SM, Dominguez M, Ilmarinen T, Costigan C, Irvine AD. Dermatological manifestations of autoimmune polyendocrinopathy-candidiasisectodermal dystrophy syndrome. Br J Dermatol 2006;154:1088-93.

J ALLERGY CLIN IMMUNOL PRACT MARCH 2019

127. Husebye ES, Perheentupa J, Rautemaa R, Kampe O. Clinical manifestations and management of patients with autoimmune polyendocrine syndrome type I. J Intern Med 2009;265:514-29. 128. Aydin SE, Kilic SS, Aytekin C, Kumar A, Porras O, Kainulainen L, et al. DOCK8 deficiency: clinical and immunological phenotype and treatment options—a review of 136 patients. J Clin Immunol 2015;35:189-98. 129. Kearney CJ, Randall KL, Oliaro J. DOCK8 regulates signal transduction events to control immunity. Cell Mol Immunol 2017;14:406-11. 130. Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, et al. Dedicator of cytokinesis 8-deficient CD4(þ) T cells are biased to a TH2 effector fate at the expense of TH1 and TH17 cells. J Allergy Clin Immunol 2017;139:933-49. 131. Bonilla FA, Khan DA, Ballas ZK, Chinen J, Frank MM, Hsu JT, et al. Practice parameter for the diagnosis and management of primary immunodeficiency. J Allergy Clin Immunol 2015;136:1186-1205.e1-1205.e78. 132. Gavino C, Cotter A, Lichtenstein D, Lejtenyi D, Fortin C, Legault C, et al. CARD9 deficiency and spontaneous central nervous system candidiasis: complete clinical remission with GM-CSF therapy. Clin Infect Dis 2014;59:81-4. 133. Jachiet M, Lanternier F, Rybojad M, Bagot M, Ibrahim L, Casanova JL, et al. Posaconazole treatment of extensive skin and nail dermatophytosis due to autosomal recessive deficiency of CARD9. JAMA Dermatol 2015;151:192-4. 134. Sonderegger I, Iezzi G, Maier R, Schmitz N, Kurrer M, Kopf M. GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J Exp Med 2008;205:2281-94. 135. Toubiana J, Okada S, Hiller J, Oleastro M, Lagos Gomez M, Aldave Becerra JC, et al. Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood 2016;127:3154-64. 136. van de Veerdonk FL, Netea MG, Joosten LA, van der Meer JW, Kullberg BJ. Novel strategies for the prevention and treatment of Candida infections: the potential of immunotherapy. FEMS Microbiol Rev 2010;34:1063-75. 137. Aldave JC, Cachay E, Nunez L, Chunga A, Murillo S, Cypowyj S, et al. A 1year-old girl with a gain-of-function STAT1 mutation treated with hematopoietic stem cell transplantation. J Clin Immunol 2013;33:1273-5. 138. Leiding JW, Okada S, Hagin D, Abinun M, Shcherbina A, Balashov DN, et al. Hematopoietic stem cell transplantation in patients with gain-of-function signal transducer and activator of transcription 1 mutations. J Allergy Clin Immunol 2018;141:704-717.e5. 139. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med 2015;66:311-28. 140. Higgins E, Al Shehri T, McAleer MA, Conlon N, Feighery C, Lilic D, et al. Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol 2015;135:551-3. 141. Mossner R, Diering N, Bader O, Forkel S, Overbeck T, Gross U, et al. Ruxolitinib induces interleukin 17 and ameliorates chronic mucocutaneous candidiasis caused by STAT1 gain-of-function mutation. Clin Infect Dis 2016; 62:951-3. 142. Zimmerman O, Rosler B, Zerbe CS, Rosen LB, Hsu AP, Uzel G, et al. Risks of ruxolitinib in STAT1 gain-of-function-associated severe fungal disease. Open Forum Infect Dis 2017;4:ofx202. 143. Forbes LR, Vogel TP, Cooper MA, Castro-Wagner J, Schussler E, Weinacht KG, et al. Jakinibs for the treatment of immune dysregulation in patients with gain-of-function STAT1 or STAT3 mutations. J Allergy Clin Immunol 2018;142:1665-9. 144. Rosler B, Wang X, Keating ST, Joosten LAB, Netea MG, van de Veerdonk FL. HDAC inhibitors modulate innate immune responses to microorganisms relevant to chronic mucocutaneous candidiasis. Clin Exp Immunol 2018;194:205-19.