Next Article in Journal
Complete and Incomplete Pentalogy of Cantrell
Next Article in Special Issue
Quality of Life and Disease Impact of Atopic Dermatitis and Psoriasis on Children and Their Families
Previous Article in Journal
The Hunger Vital Sign Identifies Household Food Insecurity among Children in Emergency Departments and Primary Care
Previous Article in Special Issue
Biologic Treatment Options for Pediatric Psoriasis and Atopic Dermatitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pathophysiology of Atopic Dermatitis and Psoriasis: Implications for Management in Children

by
Raj Chovatiya
1 and
Jonathan I. Silverberg
2,*
1
Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
2
Department of Dermatology, George Washington University School of Medicine, Washington, DC 20037, USA
*
Author to whom correspondence should be addressed.
Submission received: 30 August 2019 / Revised: 23 September 2019 / Accepted: 25 September 2019 / Published: 4 October 2019
(This article belongs to the Special Issue Atopic Dermatitis and Psoriasis in Children)

Abstract

:
Atopic dermatitis (AD) and psoriasis are chronic inflammatory skin diseases associated with a significant cutaneous and systemic burden of disease as well as a poor health-related quality of life. Here, we review the complex pathophysiology of both AD and psoriasis and discuss the implications for treatment with current state-of-the-art and emerging topical and systemic therapies. Both AD and psoriasis are caused by a complex combination of immune dysregulation, skin-barrier disruption, genetic factors, and environmental influences. Previous treatments for both diseases were limited to anti-inflammatory agents that broadly suppress inflammation. Emerging insights into relevant pathways, including recognition of the role of T-helper type 2 driven inflammation in AD and T-helper 1 and 17 driven inflammation in psoriasis, have led to a therapeutic revolution. There are a number of novel treatment options available for AD and psoriasis with many more currently under investigation.

1. Introduction

Atopic dermatitis (AD) and psoriasis are chronic, inflammatory skin diseases associated with considerable morbidity. Though both AD and psoriasis were once considered to be skin-limited disorders, emerging evidence suggests a substantial systemic burden of disease. AD is associated with higher rates of mental health symptoms and disorders, including depression, anxiety, and attention-deficit (hyperactivity) disorder, sleep dysregulation, other atopic disorders (e.g., asthma, hay fever), cardiovascular disease, stroke, and obesity [1,2,3,4,5,6,7]. Psoriasis is associated with rheumatologic (psoriatic arthritis), cardiovascular, metabolic, hepatic, and psychiatric disease [8,9,10,11]. Both AD and psoriasis are strongly associated with poor health-related quality of life (QOL), high direct and indirect costs of care, and significant societal cost—highlighting a need for optimal disease control [12,13,14,15].
AD and psoriasis are caused by a complex interplay between skin-barrier disruption, immune dysregulation, host genetics, and environment triggers [16,17]. Both diseases result in chronic, systemic inflammation with increased circulating populations of leukocytes, lymphocytes, cytokines, and chemokines (predominantly Th2 pathways in AD vs. Th1 and Th17 pathways in psoriasis) [18,19]. Previously, therapeutic options for AD and psoriasis, particularly extensive and/or severe disease, were limited to topical and non-targeted systemic immunosuppressants that had poor efficacy, safety and/or tolerability. In recent years, a better understanding of the underlying pathologic mechanisms in both conditions has led to a revolution in the development of novel, targeted therapies.
This manuscript reviews the pathophysiology of AD and psoriasis, current and novel therapeutics, and implications for management in children.

2. Atopic Dermatitis

Atopic dermatitis affects up to 15–20% of children and 1–10% of adults worldwide [20]. Approximately two-thirds of affected children present by two years of age, and 80% present by five years of age [21]. AD is a heterogeneous disorder associated with variable symptoms (e.g., pain, sleep disturbance, depression, anxiety), signs (e.g., papules, plaques, xerosis, erosions, oozing/weeping, lichenification, prurigo nodules, lichenoid papules, follicular accentuation), distributions (e.g., flexural, extensor, head and neck, hands, trunk), age of onset (e.g., pediatric, adult) and persistence (e.g., transient, intermittent, persistent) [4,5,22,23,24,25,26,27,28,29,30,31,32,33,34]. While some clinical characteristics are more common in children (e.g., ventral wrist dermatitis), others are more common in adults (e.g., hand and foot dermatitis, nipple eczema, thinning of the eyebrows, modification of disease course by emotional or environmental factors) [24].
AD is characterized by dysfunction in both non-immune and immune components of the skin-barrier. Despite this general framework, there is continued debate as to the exact etiology. The “outside-in” hypothesis posits that physical changes in the epidermal structure (e.g., keratinocytes, proteins, lipids, pH) drive immune dysregulation, whereas the “inside-out” hypothesis suggests that aberrant immune activity (e.g., leukocytes, lymphocytes, cytokines, antimicrobial peptides) alters the skin-barrier [35]. Despite the incongruity in the sequence of events, both models agree upon many of the same common mechanisms that result in chronic inflammation.

2.1. Pathophysiology

2.1.1. The Outside-In Hypothesis

The physical barrier function of the epidermis is crucial for protection against pathogens, allergens, toxins, and other irritants and maintenance of appropriate skin hydration. According to the outside-in hypothesis, a compromise in the complex matrix of epidermal keratinocytes, proteins, and/or lipids predisposes the epidermis to external insults, transepidermal water loss (TEWL) and reduced skin hydration, resulting in immune dysregulation and inflammation [36]. Previous studies found that lesional skin from AD patients had significant barrier dysfunction compared to healthy controls. Moreover, even previously inflamed and non-lesional skin continues to show barrier disruption [37].
Further evidence for importance of barrier dysfunction in AD is provided by loss-of-function mutations in the filaggrin gene (FLG) [38,39]. The outermost skin-barrier is composed of the cornified envelope, a highly cross-linked, flexible, insoluble structure composed of corneocytes, lipids, involucrin, loricrin, and filaggrin. Filaggrin, cleaved from its precursor profilaggrin, plays a key role in aligning and securing corneocytes, protein fibrils, and lamellar sheets [40]. Loss-of-function mutations in FLG confer increased risk of developing AD, earlier onset, persistent disease, other atopic disorders, and infection [41,42,43]. The processing of profilaggrin to filaggrin produces polycarboxylic acids (e.g., urocanic acid, pyrrolidone-5-carboxylic acid) known as natural moisturizing factors (NMFs) These NMFs promote hydration of the skin. A decrease in NMFs as a result of impaired profilaggrin cleavage decreases osmotic draw and results in increased TEWL [44].
NMFs and other acidic metabolites, including free fatty acids, function more broadly in decreasing skin surface pH [45]. Acidic skin pH (4–6) is important for appropriate expression, confirmation, and modification of epidermal proteins [46], inhibition (e.g., proteases) and activation (e.g., lipases) of enzymes that are critical for barrier function [47,48], and regulation of nonpathogenic skin flora [49]. Patients with AD have significantly higher pH in both lesional and non-lesional skin [50,51].
Tight junctions are epidermal protein complexes composed of claudins and occludins that are designed to seal off intercellular space, control permeability to solutes, water, pathogens, antigens, and allergens, and maintain cell polarity [52]. Functional impairment of tight junctions has been observed in patients with AD and results in skin-barrier dysfunction [53,54,55].
A variety of external and environmental factors have also been shown to contribute to skin-barrier impairment in AD patients, including in utero exposures [56], bacterial dysbiosis [57], harsh climate [58], water hardness [59,60], airborne pollutants [61,62,63], tobacco smoke [64], personal care products (i.e., irritants and pruritogens) [65,66,67], and, in certain cases, contact allergens [68]. Kantor and Silverberg have recently reviewed the topic of environmental risk factors in AD [69].
Thus, a variety of factors contribute to dysfunction of the physical epidermal barrier, resulting in keratinocyte damage, enhanced antigen and allergen penetration, and increased microbial signaling, ultimately resulting in immune cell activation, cytokine production, and inflammation.

2.1.2. The Inside-Out Hypothesis

According to the inside-out hypothesis of AD, dysregulation of the immune system results in disruption of the keratinocyte barrier and inflammation [70]. General support for this model is provided by the fact that many patients with AD have genetic polymorphisms in cytokines, chemokines, and receptors (including IL-4, IL-13, IL-18, IL-22, IL-31, CCL5, and CD14) [71,72,73,74], and treatment of human epidermal keratinocytes with many of these same mediators recapitulates the atopic dermatitis phenotype [75].
The innate immune system is composed of soluble and cellular effectors that utilize germline-encoded pattern recognition receptors (PRRs). Genetic polymorphisms in Toll-like receptors (TLRs), one such family of PRRs, confers increased risk of AD and staphylococcus aureus (SA) infection [76,77]. Antimicrobial peptides (AMPs), a diverse group of soluble, amphipathic, cationic proteins, are decreased in the skin of atopics and associated with increased TEWL, fatty acid loss, increased pH, and colonization with SA [78,79,80]. Various subsets of CD11c+ and CD1a+ dendritic cells (DCs), which specialize in antigen uptake and presentation to lymphocytes, are increased in number and activity in patients with AD and contribute to T-cell activation and polarization [81,82]. Type 2 innate lymphoid cells (ILCs) also show increased activity in AD patients and contribute to DC activation by producing IL-5 and IL-13 [83]. Keratinocyte-derived IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) are increased in atopic skin and contribute to activation of DCs, type 2 ILCs, and type 2 inflammation. Though eosinophils and mast cells can both be elevated in the skin and blood of AD patients, they likely play a lesser role in the pathogenesis of AD [84].
The adaptive immune system is composed of antigen-specific lymphocytes (T-cells, B-cells) that utilize long-lasting, specific memory to direct the host immune response. Previously, AD was thought to be a disease of Th1/Th2 imbalance; however, more recent data suggest a biphasic T-cell response with additional roles played by Th17 and Th22 cells [85]. All stages of AD (early through late) are dominated by Th2 cell activity, as evidenced by increased production of the type 2 cytokines and chemokines IL-4, IL-5, IL-10, IL-13, IL-31, and CCL5 [86]. This group of mediators is involved in allergic responses, B-cell class-switching to IgE, impairment of terminal keratinocyte differentiation (through inhibition of filaggrin, loricrin, and involucrin), and downregulation of AMPs, all of which lead to increased skin permeability to exogenous antigens and pathogens [75,87]. Later stages of AD show upregulation of Th1 activity, resulting in higher levels of IFN-γ and ultimately keratinocyte apoptosis [88]. Th17 cells, a subset of T-cells defined by production of IL-17 and IL-22, appear to be increased in number but decreased in activity in AD (unlike psoriasis). Their role in pathogenesis still remains unclear. Th22 cells, a more recently described subset of T-cells that show an increased number and activity in AD lesions, produce high levels of IL-22 that may induce epidermal hyperplasia and terminal keratinocyte differentiation [89].

2.2. Treatments

2.2.1. Topical Treatments

Topical therapies are typically the first-line treatment modality for AD. They are often used as monotherapy in mild to moderate disease and in combination with systemic, biologic, or phototherapy in moderate to severe cases. Moisturizers and emollients restore the dysfunctional epidermal barrier by reducing TEWL and in certain formulations, replacing and supplementing lipids [90]. Emollients include thicker, occlusive vehicles like petrolatum and thinner, more spreadable vehicles such as creams and lotions, which are generally less effective sealants. Topical corticosteroids (TCS) act broadly to suppress the activity of innate and adaptive immune cells. Numerous randomized controlled trials (RCTs) demonstrated efficacy for both reactive and proactive treatment protocols [91]. While TCS are overall safe, efficacious, and well tolerated for a wide spectrum of disease, adverse effects (AEs) of their chronic use include skin atrophy, fragility, striae, poor wound healing, local immunosuppression, and potential systemic absorption leading to adrenal insufficiency. Topical calcineurin inhibitors (TCIs), including tacrolimus and pimecrolimus, inhibit calcineurin signaling thereby inhibiting T-cell activation. TCIs are most often used in areas of thinner skin (e.g., eyelids, axillae, genitals) that have a higher risk of AEs from TCS. They are less commonly used as first-line therapy owing to high cost, activity comparable to low potency TCS [92], slower onset than TCS, application site reactions (e.g., stinging, burning) and an FDA-issued black box warning for potential malignancy. Long-term manufacturer sponsored registries for pimecrolimus and tacrolimus have found no evidence of increased malignancy to-date [92]. As such, TCIs are still recommended for topical treatment of AD in infants, children and adults [91].

2.2.2. Systemic Treatments

Systemic corticosteroids, unlike TCS, universally suppress immune activity. While effective in the short-term and as a bridge therapy, their long-term use is limited by a poor AE profile including hyperglycemia, diabetes, weight gain, insomnia, cardiovascular disease, osteoporosis and osteonecrosis, adrenal insufficiency, gastroesophageal reflux, and immune suppression [93,94]. Systemic immunosuppressants, including cyclosporine A, methotrexate, azathioprine, and to a lesser extent, mycophenolate mofetil, demonstrated efficacy for moderate to severe AD in a variety of RCTs [95]. While more specific than systemic corticosteroids, given their relatively broad effects on immune function, systemic immunosuppressants still have a broad AE profile and are often poorly tolerated and not ideal options for long-term treatment.

2.2.3. Phosphodiesterase-4 Inhibition

In 2016, the FDA approved the first topical phosphodiesterase-4 (PDE-4) inhibitor (crisaborole) for use in mild-to-moderate AD in children and adults. PDE-4 is responsible for degrading cAMP in immune cells, which promotes the expression of pro-inflammatory cytokines like IFN-γ and IL-17 and inhibits production of anti-inflammatory cytokines such as IL-10 [96]. Two phase III, double-blind, placebo controlled RCTs in adults and children showed that crisaborole was significantly more effective than vehicle with no major AEs other than application site reactions (e.g., stinging) [97]. Other topical PDE-4 inhibitors are currently in phase I, II, and II development [98]. Apremilast, an oral PDE-4 inhibitor that is approved for the treatment of moderate-to-severe psoriasis, has also been studied in AD. A recent phase II showed no difference compared to vehicle at lower doses and modest (but statistically significant) improvement in AD at higher doses [99]. However, higher doses of apremilast were associated with a high rate of treatment emergent AEs resulting in its discontinuation during the trial [99].

2.2.4. IL-4 And IL-13 Inhibition

In 2017, the FDA approved dupilumab, a fully human monoclonal antibody for the treatment of AD. Dupilumab targets IL4Ra, a receptor subunit protein shared by both type I and type II receptors for IL-4 and/or IL-13. In phase II and III double-blind, placebo controlled RCTs, patients with moderate to severe AD treated with dupilumab showed significant improvement in a number of primary and secondary outcomes including clinician reported outcomes (e.g., Eczema Area and Severity Index (EASI), SCORing Atopic Dermatitis (SCORAD), body surface area (BSA) involvement, investigators global assessment (IGA)) and patient-reported outcomes (e.g., pruritus numeric rating scale (NRS), Patient-Oriented Eczema Measure (POEM), and Dermatology Life Quality Index (DLQI)) [100,101,102,103]. Dupilumab demonstrated an excellent safety profile with mainly mild or moderate AEs, including injection site reactions and ocular AEs (dry eye, eye pruritus, blepharitis, conjunctivitis, keratitis), with no increases of infection or malignancy overall, and lower rates of bacterial skin infection compared to placebo.
There are currently ongoing studies examining the impact of monoclonal antibodies targeting IL-13 (lebrikizumab and tralokinumab), which are even more specific than targeting IL4 and IL13 for AD [104]. Lebrikizumab in combination with TCS showed promising results in a phase II study with significant improvement in the proportion of patients achieving ≥50% reduction in EASI from baseline (EASI-50) at 12 weeks compared to placebo [105]. Tralokinumab in combination with mid-potency TCS was effective in a phase II RCT in adults and showed significant improvement in EASI, DLQI, SCORAD, and NRS-itch scores with a significant proportion of patients achieving EASI-50 and EASI-75 at 12 weeks compared to placebo [106].

2.2.5. Il-31 Inhibition

Nemolizumab, a humanized monoclonal antibody targeting the IL-31 receptor (a major signaling pathway for itch in AD), is currently in development. A phase I double-blind, placebo controlled RCT showed that a single dose of nemolizumab in AD patients resulted in significant improvement in visual analog scale (VAS)-itch, sleep and decreased use of TCS compared to placebo [107]. A phase II double-blind placebo controlled RCT demonstrated that nemolizumab 0.5 mg/kg every four weeks significantly improved VAS-itch, EASI, and IGA in adults with AD [108]. The results of phase III studies are pending.

2.2.6. Janus Kinase Inhibition

IL-4, IL-13, IL-31, and other important cytokines in the pathogenesis of AD signal through extracellular receptors that activate intracellular Janus kinases (JAKs) [109,110]. Activation of JAKs results in downstream activation of mitogen-activated protein kinases (MAPKs), phosphoinositide 3-kinases (PI3Ks), and signal transducers and activators of transcription (STATs). Multiple JAK inhibitors are currently approved therapy for treatment of rheumatoid arthritis, psoriatic arthritis, and ulcerative colitis. Emerging data showed potential efficacy for AD. A case series of six adults with moderate to severe AD treated off-label with tofacitinib (an oral JAK 1 and 3 inhibitor) showed significant improvements in pruritus and mean SCORAD [111]. A phase II double-blind, placebo controlled RCT of adults with mild-to-moderate AD demonstrated that topical tofacitinib ointment versus vehicle resulted in significant reduction in EASI, BSA, and Physician’s Global Assessment (PGA). A number of other topical and/or oral JAK inhibitors are currently under investigation for treatment of AD. A phase II double-blind, placebo controlled RCT with baricitinib (an oral selective JAK 1 and 2 inhibitor) in combination with TCS showed improvement in inflammation and pruritus with significantly more patients achieving EASI-50 at 16 weeks compared to placebo [112]. A phase II double-blind, placebo controlled RCT with upadacitinib (an oral JAK 1 inhibitor) monotherapy showed a significant decrease in EASI and NRS itch scores at 16 weeks (primary endpoint) and 32 weeks (long-term extension) [113]. Additional long-term studies are underway to better understand the efficacy and safety profile of JAK inhibitors.

3. Psoriasis

Psoriasis is estimated to affect 2–3% of the global population, corresponding to >125 million individuals [114,115]. Though most affected individuals present during adulthood between 18–39 or after the age of 50, approximately one-third of patients experience their first episode of psoriasis under the age of 20 [17,114]. Psoriasis is an immune-mediated disease characterized by cycles of sustained inflammation and remission, uncontrolled keratinocyte proliferation, and impaired keratinocyte differentiation. Inflammation is driven by dysregulation in both the innate and adaptive immunity, and influenced by genetic factors. Plaque psoriasis is characterized by well-demarcated, erythematous, scaly plaques on the trunk and extensor surfaces of the limbs. Inverse psoriasis affects the intertrigenous skin with erythematous, occasionally erosive patches and plaques. Guttate psoriasis is more common in children and adolescents, and is characterized by droplet sized scaly plaques that can be triggered by group A streptococcal infection. Nearly one-half of childhood cases of psoriasis may first present in this way. Pustular psoriasis, occurring in both local and generalized forms, presents with innumerable, coalescing, sterile pustules. While psoriasis can manifest with a variety of morphologies, all subtypes share several important features: (1) hyperplasia of the epidermis (i.e., acanthosis), (2) incomplete keratinization with retention of nuclei (i.e., parakeratosis), (3) newly generated, tortuous superficial blood vessels (i.e., neovascularization), and (4) a dense inflammatory infiltrate composed of DCs, macrophages, neutrophils, and T-cells [116].

3.1. Pathophysiology

3.1.1. Dysregulated Immune Activity

The initiation phase of psoriasis consists of an external insult to the epidermis, such as trauma (e.g., Koebner phenomenon), medication (e.g., β-blocker, ACE-inhibitor, lithium), and/or infection (e.g., group A streptococcus) [115]. Following keratinocyte damage, two important events occur to activate the innate immune system: (1) keratinocytes dramatically upregulate the production of AMPs including S100, β-defensins, and LL-37 (i.e., cathelicidin), and (2) damaged keratinocytes release DNA, RNA, and other endogenous danger signals [117,118]. One proposed mechanism for the initiation of psoriasis posits that these AMPs bind to the keratinocyte DNA and RNA and activate plasmacytoid DCs (pDCs), a circulating antigen presenting cell (APC) specialized for recognition of viral pathogens. In particular, LL37 complexes with self-genetic material and binds to TLR7, TLR8, and TLR9, resulting in the secretion of type I interferons (IFN-α and IFN-β) and a break in immunologic tolerance [119,120,121]. Keratinocytes, which also function as a physical barrier and sensory cell in the innate immunity, produce pro-inflammatory cytokines in response to AMP-nucleic acid complexes, including type I IFNs, TNF-α, IL-1, and IL-6 [122]. Type I IFNs promote myeloid DC (mDC) differentiation, activation, and entry into skin-draining lymph nodes [123]. These cells also secrete a number of pro-inflammatory cytokines, such as TNF-α, IL-12, and IL-23, resulting in the differentiation and activation of activation of Th1, Th17, and Th22 cells.
Without specific therapeutic intervention, activation of adaptive immunity drives the self-sustaining, maintenance phase of inflammation in psoriasis [124]. The initial stage of the adaptive immunity is characterized by Th1 cell activity and concomitant production of type 1 cytokines such as IFN-γ, while Th17 cells (and to a lesser extent, Th22 cells) dominate the later portion of the adaptive immune response with production of IL-17, IL-23, IL-21, and IL-22 [125]. Recent studies focused extensively on the role Th17 cells in the pathogenesis of psoriasis, in part due to their high concentration in psoriatic lesions and significant reduction following anti-TNF-α treatment [126]. Th17 cells are a subset of T-lymphocytes that specialize in epithelial surveillance and defense against extracellular pathogens through production of IL-17, IL-23, and IL-22. The IL-17 family is composed of six members, two of which (IL-17A and IL-17F) appear to be most relevant in psoriasis. IL-17 activates a variety of intracellular kinase cascades in effector cells, including JAK, MAPK, NF-κB, and glycogen synthase kinase 3 β (GSK-3-β) [127]. IL-22 and IL-23 link immune activity and keratinocyte function by inducing keratinocyte proliferation and secretion of AMPs and cytokines [128].
Though TNF-α, IL-17, and IL-23 play a key role in the general pathophysiology of psoriasis, especially plaque psoriasis, there are additional relevant pathways in other variants of psoriasis. IL-1, IL-8, and IL-36 appear to be important for neutrophilic activity in pustular psoriasis [129,130], whereas guttate psoriasis may be driven by streptococcal superantigen mediated activation of TCRs and subsequent molecular mimicry of keratin proteins [131,132].

3.1.2. Genetic Factors

Early epidemiologic studies suggested a genetic component to psoriasis, as patients with psoriasis had higher incidence of disease among first and second-degree relatives compared to the general population, and monozygotic twins had two to threefold higher risk of psoriasis compared to dizygotic twins [133,134,135]. These findings were later confirmed by genome-wide linkage studies, which have identified over 70 different genetic loci associated with psoriasis [136,137]. However, variability in these loci are thought to only account for about 30% of heritability, suggesting a cumulative effect from multiple mutations with smaller individual effects and/or undetected gene-gene interactions. The nine most highly associated loci are named psoriasis susceptibility 1 through 9 (PSORS1 through PSORS9), with PSORS1 accounting for 35–50% of heritability in psoriasis [136,138]. PSORS1 is located in the major histocompatibility complex (MHC) on chromosome 6p21, spanning a 220 kb segment class I telomeric region of human leukocyte antigen B (HLA-B). This region contains several candidate genes, including HLA-C (associated variant HLA-Cw6 [HLA-C*06:02]; an MHC class I protein), CDSN (associated variant allele 5; corneodesmin, a protein expressed in the upper epidermis) [139], and CCHCR1 (associated variant WWCC; a widely expressed protein that is overexpressed in psoriasis) [140]. While exact identification has been technically challenging due to strong linkage disequilibrium, most studies agree that HLA-Cw6 is the susceptibility allele in PSORS1 [141]. Studies of HLA-Cw6 showed several important associations: overall increased risk of psoriasis, especially in white and Chinese populations [141,142]; early-onset and more severe disease, especially in those with positive family history [141,143]; guttate psoriasis and streptococcal pharyngitis, but not pustular psoriasis [143,144]. Other psoriasis susceptibility loci include PSORS2 on chromosome 17q24–q25, which spans the gene for caspase recruitment domain-containing protein 14 (CARD14), a scaffolding protein involved triggering NF-κB activation [145,146]; PSORS4 on chromosome 1q which spans the epidermal differentiation complex [147]; PSORS6 on chromosome 19p13, which spans the gene for tyrosine kinase 2 (TYK2) [148]; PSORS7 on chromosome 1p, which spans the gene for IL-23 receptor (IL-23R) [149].
Genome-wide association studies (GWAS) have largely confirmed the findings from previous linkage studies, including the importance of the PSORS1 and the MHC locus [150,151,152], while identifying additional risk loci through high resolution analyses of small nucleotide polymorphisms (SNPs) in large sample populations [153,154]. Many of these variants highlight the importance of the immune system in the pathogenesis of psoriasis [155,156]. Numerous GWAS studies identified variants in cytokines, receptors, and signaling pathways that are critical for Th17 cell function: IL-23R (the receptor that binds to IL-23 and promotes activation and expansion of Th17 cells); IL-12B (the shared p40 subunit of IL-23 and IL-12); STAT3 (a signal transducer in the JAK/STAT signaling pathway downstream of several cytokine receptors including IL-23R); and Runx1 (a transcription factor important in Th17 cell differentiation) [153,155,156,157,158]. Additional variants were identified in the NF-κB signaling pathway [159,160]. More recently, next-generation sequencing and expression profiling have been combined with linkage and association studies to identify genetic variants with high accuracy, as evidenced by the successful identification of loss-of-function mutation in IL-36 antagonist (IL-36N) as the genetic basis for generalized pustular psoriasis [161,162].

3.2. Treatments

3.2.1. Topical Treatments

As with AD, topical therapies are the first-line treatment for psoriasis, both as monotherapy in mild-to-moderate disease and as combination therapy with oral systemic, biologic, and phototherapy in moderate-to-severe disease. Historically, treatment for psoriasis was limited to topical preparations of anthralin (also known as dithranol), which induces apoptosis of keratinocytes through production of reactive oxygen species, and coal tar, which inhibits production of IL-15 and nitric oxide [163,164]. Though still used today, these therapies have largely been replaced by TCS. Numerous RCTs demonstrated clinical efficacy for both continuous and intermittent treatment of psoriasis with varying potencies of TCS [165,166]. Topical vitamin D analogs (including calcitriol, calcipotriene, and tacalcitol) have broad effects of keratinocyte proliferation, apoptosis, and immunomodulation [167]. Their activity is comparable to mid-potency TCS (without the associated AEs), and is enhanced when combined or alternated with TCS [168,169]. Topical vitamin D analogs have few AEs, and systemic effects on vitamin D, calcium, and parathyroid hormone are extremely rare [170]. The TCIs tacrolimus and pimecrolimus are used off-label to treat psoriasis, particularly in the facial, genital, and intertrigenous areas [171]. While safe and efficacious, their use is limited by their overall potency, which is rated lower than mid-potency TCS and vitamin D analogs [172]. Topical retinoids (vitamin A derivatives) bind to the retinoic acid receptor (RAR) on both keratinocytes and non-immune cells, altering expression of genes important for differentiation, proliferation, and inflammation [173]. Tazarotene was the first retinoid developed for treatment of psoriasis, and, while efficacious, its continuous use is limited by local irritation. In combination with TCS, tazarotene shows increased potency and decreased local AEs [174,175].

3.2.2. Phototherapy

Phototherapy is a therapeutic option for moderate-to-severe psoriasis involving a large BSA (>10%), both as monotherapy and combination therapy. Its utility, however, is practically limited by the requirement to travel to a phototherapy center multiple times weekly or own a home phototherapy unit. The mechanism of phototherapy is believed to be multifactorial, including induction of apoptosis in keratinocytes, APCs, and Th17 cells, and promotion of regulatory T-cell (Treg) activation [176]. Broadband UVB (290–320 nm) was used for nearly a century; however, it has largely been replaced by narrow band UVB (NB-UVB; 311 nm) and excimer laser (308 nm) [177]. NB-UVB has a good safety profile with a low risk for cutaneous malignancy. In contrast, an alternative to UVB-based therapy is psoralen combined with UVA (PUVA). Oral ingestion of 8-methoxsalen is followed by exposure to UVA light, resulting in selective apoptosis of proliferating cells. Once the gold standard for targeted phototherapy, PUVA has fallen out of favor due to its carcinogenic effects and association with squamous cell carcinoma and melanoma of the skin [178,179].

3.2.3. Systemic Therapy

As was previously discussed for AD, systemic immunosuppressants, including methotrexate (the first systemic therapy approved for psoriasis) and cyclosporine A, have shown efficacy in treating moderate-to-severe psoriasis, but are limited by their relative non-specificity and broad AE profile. Acitretin is an oral second-generation retinoid that is approved for the treatment of moderate-to-severe psoriasis and is particularly efficacious for pustular psoriasis, erythrodermic psoriasis, and as an adjunct to phototherapy [180]. Similar to the mechanism of action of tazarotene, it binds to the RAR and regulates transcription of genes important for epidermal proliferation, keratinocyte differentiation, and immune cell production of cytokines. Overall, it appears to be less effective than traditional systemic agents for plaque psoriasis, and has several important AEs including dry skin and mucous membranes, dyslipidemia, elevated liver function tests, and potential teratogenicity [181].

3.2.4. Phosphodiesterase-4 Inhibition

In 2014, the FDA approved apremilast for the treatment of moderate-to-severe psoriasis. As discussed earlier, apremilast is a small molecule inhibitor of the PDE-4 enzyme that reduces the level of pro-inflammatory Th17 cytokines and increases expression of IL-10 [182]. In multiple phase III double-blind, placebo controlled RCTs, patients with moderate-to-severe plaque or palmoplantar psoriasis treated with apremilast showed significant improvement in PASI-50 (Psoriasis Area and Severity Index, ≥50% reduction from baseline), PASI-75, and static investigators global assessment (sIGA) at week 16 [183,184,185,186]. AEs include nausea, diarrhea, and weight loss, which can be quite significant and chronic in some patients. Though apremilast does not carry the same immunosuppression risks as biologic therapy, it is overall less efficacious with only approximately 30% of patients achieving PASI-75 scores at week 16.

3.2.5. TNF-α Inhibition

TNF-α inhibitors are the first generation of biologic medications for psoriasis, and they have been successfully used to treat moderate-to-severe psoriasis for over a decade. TNF-α is a pleiotropic, pro-inflammatory cytokine involved in both the innate and adaptive arms of the immune response in psoriasis. However, given its more general role in the immune response, TNF-α inhibitors have a broad AE profile, especially with regard to increased infectious risk (e.g., conversation of tuberculosis from latent to active, reactivation of hepatitis B virus). Etanercept (a recombinant human fusion protein consisting of the TNF-α receptor protein and Fc portion of immunoglobulin IgG1) was first-in-class and approved by the FDA in 2004, followed by infliximab (a human-murine chimeric monoclonal IgG1 antibody against TNF-α) in 2006, adalimumab (the first fully human monoclonal IgG1 antibody against TNF-α) in 2008, and certolizumab (a recombinant humanized IgG1 antibody Fab fragment against TNF-α conjugated to polyethylene glycol) in 2018 [187]. Phase III double-blind, placebo controlled RCTs for etanercept (PASI-75 at week 10–16: 47–49%) [188,189,190], infliximab (75–80%) [191,192], adalimumab (53–80%) [193,194,195,196], and certolizumab (67–83%) [197] demonstrated that biologic therapy with TNF-α inhibitors is efficacious and safe, thus paving the way for additional generations of immunotherapy.

3.2.6. IL-12/IL-23 Inhibition

The FDA approved Ustekinumab for treatment of psoriasis in 2009. Representing the next generation of biologic therapy after TNF-α inhibitors, ustekinumab is a fully human IgG1 monoclonal antibody against the shared p40 subunit of IL-12 and IL-23, thus blocking two distinct pathways of T-cell activity: Th1 and Th17, respectively [187]. Th17 cells, and to a lesser extent Th1 cells, are crucial for orchestrating the chronic, maintenance phase of psoriasis, and, as such, the p40 subunit presents a more specific target for psoriasis compared to TNF-α. Phase III double-blind, placebo controlled RCTs showed a PASI-75 of 65–78% at week 12 with overall good efficacy, safety, and long-term drug survival [198,199,200]. While infectious complications were the most serious AEs associated with ustekinumab, the overall rate of infections was lower than that seen with TNF-α inhibitors [201].

3.2.7. IL-17 Inhibition

Highlighting an evolving understanding of the immunological basis of psoriasis, the third generation of biologic medications approved by the FDA was the IL-17 inhibitors: secukinumab (a fully human monoclonal IgG1 antibody against IL-17A) in 2015; ixekizumab (a humanized monoclonal IgG4 antibody against IL-17A) in 2016; brodalumab (a human monoclonal IgG2 antibody against IL-17RA) in 2017 [187]. IL-17 is one of the signature cytokines produced by Th17 cells and responsible for amplifying the inflammatory cascade during the chronic phase of psoriasis. Phase III double-blind, placebo controlled RCTs showed high PASI-75 scores at week 12 (secukinumab: 75–87%; ixekizumab: 87–90%; brodalumab: 83–86%) and even significant, measurable PASI-90 and PASI-100 responses compared to placebo, TNF-α inhibitors, and/or ustekinumab [202,203,204,205,206,207]. Interestingly, even after discontinuation of treatment with secukinumab, approximately 20% maintained their response even after one year, suggesting the possibility of altering homeostatic set points with biologic therapy. Overall, while IL-17 inhibitors were well-tolerated and safe, AEs included upper respiratory infections, nasopharyngitis, and candidal infections, underscoring the important role of Th17 cells in extracellular defense at epithelial surfaces. However, the role of IL-17 inhibition in flaring inflammatory bowel disease (IBD) in those with a personal or family history of IBD is currently under investigation [208].

3.2.8. IL-23 Inhibition

Based on clinical data from ustekinumab patients in conjunction with basic studies showing protective effects of IL-12 in psoriasis [209] and a more important role for IL-12 in Th1 mediated host defense, IL-23 was identified as a more specific target for psoriasis therapy [210]. The latest generation of biologic medications approved by the FDA for use in moderate-to-severe psoriasis are the IL-23 inhibitors: guselkumab in 2017; tildrakizumab in 2018; and risankizumab in 2019, all of which are human monoclonal IgG1 antibodies against the p19 subunit of IL-23 [187]. Primary or secondary endpoints for the phase III double-blind, placebo controlled RCTs of the IL-23 inhibitors were measured with PASI-90 (guselkumab: week 16, 70–85%; tildrakizumab: week 12, 35%; risankizumab: week 16, 72–75%) and PASI-100 scores, representing a remarkable improvement in treatment specificity [211,212,213,214,215,216]. IL-23 inhibitors showed higher efficacy compared head-to-head with TNF-α inhibitors and/or ustekinumab. As was seen with IL-17 inhibitors, about one-quarter of patients who discontinued therapy with risankizumab after 16 weeks maintained their response even after 48 weeks, again suggesting the possibility for long-term modification of cytokine signaling pathways with biologic therapy. IL-23 inhibitors were well-tolerated and safe, with most AEs related to infection (upper respiratory infection, gastroenteritis, herpes simplex virus, tinea).

3.2.9. Janus Kinase Inhibition

Many of the Th17 and Th1 cytokines involved in the pathogenesis of psoriasis signal through JAK. Oral tofacitinib is currently an FDA-approved therapy for psoriatic arthritis [217,218], and has shown promising findings for the treatment of psoriasis in phase III double-blind, placebo controlled RCTs (PASI-75 at week 16: 40–59%) [219,220]. Topical tofacitinib has also demonstrated efficacy in phase II RCTs for psoriasis [221,222]. However, systemic tofacitinib use is associated with a number of infectious AEs (several of which were observed during trials), and, in 2015, the FDA declined to approve oral tofacitinib for use in moderate-to-severe without additional safety studies. Further investigation of tofacitinib, as well as other oral JAK inhibitors including baricitinib [223] and peficitinib [224], is ongoing.

4. Management in Children

For the treatment of mild-to-moderate AD or psoriasis in children, first-line therapies include TCS and TCI, with the addition of emollients for AD and topical vitamin D analogs for psoriasis. Data supporting their use in monotherapy and/or combination therapy is derived from years of anecdotal clinical experience and studies performed in adults, as RCTs are for these agents are rare in children, especially under the age of 12. TCS use in children is sometimes limited given concern for cutaneous (e.g., atrophy, fragility, striae, infection) and systemic (e.g., hypothalamic–pituitary axis suppression) AEs. While TCIs do not cause atrophy or hypothalamic–pituitary axis suppression, they are associated with application site reactions such as burning or stinging and a theoretical black-box warning by the United States Food and Drug Association for malignancy. Despite these potential AEs, both TCS and TCIs can be used safely and efficaciously in continuous and/or intermittent fashion. Adjunctive treatment with topical vitamin D analogs in children with psoriasis is overall very well tolerated with minimal cutaneous AEs and extremely rare systemic AEs (e.g., hypercalcemia, hypercalciuria) with overuse. Crisaborole was recently added as an additional first-line topical option in AD, and the combination of its relatively specific mode of action, non-steroidal formulation, and promising long-term safety data make it an important addition to the pediatric AD arsenal [225]. However, further studies are needed to better understand crisaborole’s efficacy in children, especially in comparison with TCS and TCIs. Common AEs of crisaborole in children include application site burning and/or stinging and, rarely, application site skin infection. For psoriasis, Tazarotene is rarely used in children given the associated skin irritation, while anthralin and coal tar are limited by practical features associated with application: staining of skin and clothing (both agents) and malodor (coal tar).
For the treatment of moderate-to-severe AD or psoriasis in children, treatment options include systemic therapy (most of which are used off-label), phototherapy, and biologic agents. Current guidelines recommend a stepwise approach to treatment in children with moderate-to-severe disease with biologic agents at the top of the therapeutic ladder. Methotrexate is the most frequently used systemic agent in children and has demonstrated reasonable efficacy in prospective and retrospective analyses, though the overall quality of data is low with high inter-study variability [226,227,228,229,230,231]. Disadvantages include the need for continuous lab monitoring and potential AEs including hepatotoxicity and liver fibrosis (especially with long-term use), hematologic abnormalities, and gastrointestinal symptoms. Cyclosporine A, which is usually reserved for flaring and/or refractory AD and psoriasis, has shown good efficacy in the short-term in several uncontrolled studies; however, there are sparse data on long-term effects in children [229,232]. Furthermore, chronic use of cyclosporine A is associated with a number of severe AEs including infection, malignancy, hypertension, nephrotoxicity, and hepatotoxicity. Acitretin can be effective for children with psoriasis (especially the pustular subtype) but can cause severe dryness of the skin and mucous membranes as well as dyslipidemia [229]. Other systemic therapies have limited data to support use, including mycophenolate mofetil and azathioprine. Both are associated with similar AEs that restrict their use in children, including diarrhea, nausea, vomiting, anemia, leukopenia, immune suppression, and increased risk of infection. Systemic corticosteroids are generally contraindicated for management of AD and psoriasis despite continued inappropriate use especially in childhood AD [233], and they should only be used as bridge therapy, for immediate relief, or when other options are not available. Common side effects of chronic systemic corticosteroid use in children are similar to those in adults and include immune suppression, hyperglycemia, weight gain, insomnia, gastroesophageal reflux, osteonecrosis, and adrenal insufficiency.
For both AD and psoriasis, phototherapy with either NB-UVB or excimer laser can be preferable to systemic immunosuppressive therapy in children given the limited AEs. Uncontrolled studies support its efficacy; however, multiple sessions per week in-office can be challenging for children and caregivers, and there continues to be concern about long-term exposure to UVB light starting in childhood [234,235,236].
Dupilumab is currently the only biologic agent available for the treatment of AD, though many new biologics are under investigation. The FDA recently expanded dupilumab’s indication in 2019 for use in children ≥12 years old with AD, and investigation for use in infants and children with AD is currently ongoing. Long-term safety and efficacy data for dupilumab use in children are eagerly anticipated, with adult open-label extension data supporting its continuous, long-term use [237]. The most common treatment-related AEs observed in both adolescents and adults included injection site reactions, conjunctivitis, and nasopharyngitis. For children with psoriasis, etanercept [238], adalimumab [239], and ustekinumab [240] have been studied in phase III double-blind, placebo controlled RCTs. All three agents showed comparable efficacy and safety to the previous adult trials. Adalimumab showed higher efficacy in head-to-head comparison with methotrexate. Currently, the only FDA-approved biologics in children are etanercept (≥4 years old) and ustekinumab (≥12 years old), with ongoing studies for many of the newer treatment classes. In general, biologic treatment-associated AEs in children with psoriasis are similar to those observed in adults and more favorable compared to classic systemic therapies. Common AEs associated with TNF-α and IL-12/IL-23 inhibitors use in children include injection site reactions, nasopharyngitis, mild upper respiratory infection (similar to those normally seen in children), and headache.

5. Conclusions

Improved understanding of the pathophysiology of AD and psoriasis has led to a revolution in targeted therapy. Targeted treatments have filled many unmet needs for both of these conditions. As additional novel agents are approved, the next goal in management of AD and psoriasis is to optimize specific treatments for individual patients. The next decade of research will be dominated by extensive genetic, molecular, and clinical phenotyping of patients in order to understand which pathologic mechanisms are most relevant. The era for personalized medicine in AD and psoriasis is upon us!

Author Contributions

Data curation, R.C. and J.I.S.; Formal analysis, R.C. and J.I.S.; Writing—original draft, R.C. and J.I.S.; Writing—review and editing, R.C. and J.I.S.

Funding

This research received no external funding. Jonathan I. Silverberg is supported by a grant from the Dermatology Foundation. Raj Chovatiya is supported by a grant from the National Institutes of Health T32AR060710. The funding source had no involvement in design of the review or in curation, analysis, preparation, review, editing, or approval of the manuscript.

Conflicts of Interest

Jonathan Silverberg reports personal fees from Abbvie, Anaptysbio, Asana, EliLilly, Galderma, GlaxoSmithKline, Kiniksa, Leo, Menlo, Pfizer, Realm, Regeneron­Sanofi, and Roivant, and grants from GlaxoSmithKline, Regeneron­Sanofi, and Galderma, during the conduct of the Review. Raj Chovatiya declares no competing interests.

References

  1. Silverberg, J.I.; Silverberg, N.B.; Lee-Wong, M. Association between atopic dermatitis and obesity in adulthood. Br. J. Dermatol. 2012, 166, 498–504. [Google Scholar] [CrossRef] [PubMed]
  2. Silverberg, J.I. Association between adult atopic dermatitis, cardiovascular disease, and increased heart attacks in three population-based studies. Allergy 2015, 70, 1300–1308. [Google Scholar] [CrossRef] [PubMed]
  3. Yu, S.H.; Silverberg, J.I. Association between Atopic Dermatitis and Depression in US Adults. J. Investig. Dermatol. 2015, 135, 3183–3186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Silverberg, J.I.; Garg, N.K.; Paller, A.S.; Fishbein, A.B.; Zee, P.C. Sleep disturbances in adults with eczema are associated with impaired overall health: A US population-based study. J. Investig. Dermatol. 2015, 135, 56–66. [Google Scholar] [CrossRef]
  5. Yu, S.H.; Attarian, H.; Zee, P.; Silverberg, J.I. Burden of Sleep and Fatigue in US Adults With Atopic Dermatitis. Dermatitis 2016, 27, 50–58. [Google Scholar] [CrossRef]
  6. Silverberg, J.I.; Simpson, E.L. Association between severe eczema in children and multiple comorbid conditions and increased healthcare utilization. Pediatric Allergy Immunol. 2013, 24, 476–486. [Google Scholar] [CrossRef] [Green Version]
  7. Strom, M.A.; Fishbein, A.B.; Paller, A.S.; Silverberg, J.I. Association between atopic dermatitis and attention deficit hyperactivity disorder in U.S. children and adults. Br. J. Dermatol. 2016, 175, 920–929. [Google Scholar] [CrossRef] [Green Version]
  8. Gelfand, J.M.; Neimann, A.L.; Shin, D.B.; Wang, X.; Margolis, D.J.; Troxel, A.B. Risk of myocardial infarction in patients with psoriasis. JAMA 2006, 296, 1735–1741. [Google Scholar] [CrossRef]
  9. Ludwig, R.J.; Herzog, C.; Rostock, A.; Ochsendorf, F.R.; Zollner, T.M.; Thaci, D.; Kaufmann, R.; Vogl, T.J.; Boehncke, W.-H. Psoriasis: A possible risk factor for development of coronary artery calcification. Br. J. Dermatol. 2007, 156, 271–276. [Google Scholar] [CrossRef]
  10. Gisondi, P.; Fostini, A.C.; Fossà, I.; Girolomoni, G.; Targher, G. Psoriasis and the metabolic syndrome. Clin. Dermatol. 2018, 36, 21–28. [Google Scholar] [CrossRef]
  11. Kim, N.; Thrash, B.; Menter, A. Comorbidities in psoriasis patients. Semin. Cutan. Med. Surg. 2010, 29, 10–15. [Google Scholar] [CrossRef] [PubMed]
  12. Chiesa Fuxench, Z.C.; Block, J.K.; Boguniewicz, M.; Boyle, J.; Fonacier, L.; Gelfand, J.M.; Grayson, M.H.; Margolis, D.J.; Mitchell, L.; Silverberg, J.I.; et al. Atopic Dermatitis in America Study: A Cross-Sectional Study Examining the Prevalence and Disease Burden of Atopic Dermatitis in the US Adult Population. J. Investig. Dermatol. 2019, 139, 583–590. [Google Scholar] [CrossRef] [Green Version]
  13. Silverberg, J.I. Public Health Burden and Epidemiology of Atopic Dermatitis. Dermatol. Clin. 2017, 35, 283–289. [Google Scholar] [CrossRef] [PubMed]
  14. Gelfand, J.M.; Feldman, S.R.; Stern, R.S.; Thomas, J.; Rolstad, T.; Margolis, D.J. Determinants of quality of life in patients with psoriasis: A study from the US population. J. Am. Acad. Dermatol. 2004, 51, 704–708. [Google Scholar] [CrossRef] [PubMed]
  15. De Korte, J.; Sprangers, M.A.; Mombers, F.M.; Bos, J.D. Quality of life in patients with psoriasis: A systematic literature review. J. Investig. Dermatol. Symp. Proc. 2004, 9, 140–147. [Google Scholar] [CrossRef]
  16. Weidinger, S.; Beck, L.A.; Bieber, T.; Kabashima, K.; Irvine, A.D. Atopic dermatitis. Nat. Rev. Dis. Primers 2018, 4, 1–20. [Google Scholar] [CrossRef]
  17. Greb, J.E.; Goldminz, A.M.; Elder, J.T.; Lebwohl, M.G.; Gladman, D.D.; Wu, J.J.; Mehta, N.N.; Finlay, A.Y.; Gottlieb, A.B. Psoriasis. Nat. Rev. Dis. Primers 2016, 2, 16082. [Google Scholar] [CrossRef]
  18. Cai, Y.; Fleming, C.; Yan, J. New insights of T cells in the pathogenesis of psoriasis. Cell. Mol. Immunol. 2012, 9, 302–309. [Google Scholar] [CrossRef] [Green Version]
  19. Brunner, P.M.; Guttman-Yassky, E.; Leung, D.Y.M. The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies. J. Allergy Clin. Immunol. 2017, 139, S65–S76. [Google Scholar] [CrossRef] [Green Version]
  20. Garg, N.; Silverberg, J.I. Epidemiology of childhood atopic dermatitis. Clin. Dermatol. 2015, 33, 281–288. [Google Scholar] [CrossRef]
  21. Levy, R.M.; Gelfand, J.M.; Yan, A.C. The epidemiology of atopic dermatitis. Clin. Dermatol. 2003, 21, 109–115. [Google Scholar] [CrossRef]
  22. McKenzie, C.; Silverberg, J.I. The prevalence and persistence of atopic dermatitis in urban United States children. Ann. Allergy Asthma Immunol. 2019, 123, 173–178.e171. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, J.P.; Chao, L.X.; Simpson, E.L.; Silverberg, J.I. Persistence of atopic dermatitis (AD): A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2016, 75, 681–687.e611. [Google Scholar] [CrossRef] [PubMed]
  24. Yew, Y.W.; Thyssen, J.P.; Silverberg, J.I. A systematic review and meta-analysis of the regional and age-related differences in atopic dermatitis clinical characteristics. J. Am. Acad. Dermatol. 2019, 80, 390–401. [Google Scholar] [CrossRef]
  25. Vakharia, P.P.; Silverberg, J.I. Adult-Onset Atopic Dermatitis: Characteristics and Management. Am. J. Clin. Dermatol. 2019, 76, 958. [Google Scholar] [CrossRef]
  26. Vakharia, P.P.; Chopra, R.; Sacotte, R.; Patel, K.R.; Singam, V.; Patel, N.; Immaneni, S.; White, T.; Kantor, R.; Hsu, D.Y.; et al. Burden of skin pain in atopic dermatitis. Ann. Allergy Asthma Immunol. 2017, 119, 548–552.e543. [Google Scholar] [CrossRef]
  27. Silverberg, J.I.; Vakharia, P.P.; Chopra, R.; Sacotte, R.; Patel, N.; Immaneni, S.; White, T.; Kantor, R.; Hsu, D.Y. Phenotypical Differences of Childhood- and Adult-Onset Atopic Dermatitis. J. Allergy Clin. Immunol. Pract. 2018, 6, 1306–1312. [Google Scholar] [CrossRef]
  28. Silverberg, J.I.; Margolis, D.J.; Boguniewicz, M.; Fonacier, L.; Grayson, M.H.; Ong, P.Y.; Chiesa Fuxench, Z.C.; Simpson, E.L.; Gelfand, J.M. Distribution of atopic dermatitis lesions in United States adults. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1341–1348. [Google Scholar] [CrossRef]
  29. Silverberg, J.I.; Gelfand, J.M.; Margolis, D.J.; Boguniewicz, M.; Fonacier, L.; Grayson, M.H.; Ong, P.Y.; Chiesa Fuxench, Z.C.; Simpson, E.L. Symptoms and diagnosis of anxiety and depression in atopic dermatitis in U.S. adults. Br. J. Dermatol. 2019, 119, e3. [Google Scholar] [CrossRef]
  30. Silverberg, J.I.; Gelfand, J.M.; Margolis, D.J.; Boguniewicz, M.; Fonacier, L.; Grayson, M.H.; Chiesa Fuxench, Z.C.; Simpson, E.L.; Ong, P.Y. Pain Is a Common and Burdensome Symptom of Atopic Dermatitis in United States Adults. J. Allergy Clin. Immunol. Pract. 2019. [Google Scholar] [CrossRef]
  31. Silverberg, J.I. Adult-Onset Atopic Dermatitis. J. Allergy Clin. Immunol. Pract. 2019, 7, 28–33. [Google Scholar] [CrossRef] [PubMed]
  32. Patel, K.R.; Immaneni, S.; Singam, V.; Rastogi, S.; Silverberg, J.I. Association between atopic dermatitis, depression, and suicidal ideation: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2019, 80, 402–410. [Google Scholar] [CrossRef] [PubMed]
  33. Li, J.C.; Fishbein, A.; Singam, V.; Patel, K.R.; Zee, P.C.; Attarian, H.; Cella, D.; Silverberg, J.I. Sleep Disturbance and Sleep-Related Impairment in Adults With Atopic Dermatitis: A Cross-sectional Study. Dermatitis 2018, 29, 270–277. [Google Scholar] [CrossRef] [PubMed]
  34. Cheng, B.T.; Silverberg, J.I. Depression and psychological distress in US adults with atopic dermatitis. Ann. Allergy Asthma Immunol. 2019, 123, 179–185. [Google Scholar] [CrossRef] [PubMed]
  35. Silverberg, N.B.; Silverberg, J.I. Inside out or outside in: Does atopic dermatitis disrupt barrier function or does disruption of barrier function trigger atopic dermatitis? Cutis 2015, 96, 359–361. [Google Scholar] [PubMed]
  36. Wolf, R.; Wolf, D. Abnormal epidermal barrier in the pathogenesis of atopic dermatitis. Clin. Dermatol. 2012, 30, 329–334. [Google Scholar] [CrossRef]
  37. Seidenari, S.; Giusti, G. Objective assessment of the skin of children affected by atopic dermatitis: A study of pH, capacitance and TEWL in eczematous and clinically uninvolved skin. Acta Derm. Venereol. 1995, 75, 429–433. [Google Scholar] [CrossRef]
  38. Baurecht, H.; Irvine, A.D.; Novak, N.; Illig, T.; Bühler, B.; Ring, J.; Wagenpfeil, S.; Weidinger, S. Toward a major risk factor for atopic eczema: Meta-analysis of filaggrin polymorphism data. J. Allergy Clin. Immunol. 2007, 120, 1406–1412. [Google Scholar] [CrossRef]
  39. Palmer, C.N.A.; Irvine, A.D.; Terron-Kwiatkowski, A.; Zhao, Y.; Liao, H.; Lee, S.P.; Goudie, D.R.; Sandilands, A.; Campbell, L.E.; Smith, F.J.D.; et al. Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat. Genet. 2006, 38, 441–446. [Google Scholar] [CrossRef]
  40. O’Regan, G.M.; Sandilands, A.; McLean, W.H.I.; Irvine, A.D. Filaggrin in atopic dermatitis. J. Allergy Clin. Immunol. 2009, 124, R2–R6. [Google Scholar] [CrossRef]
  41. Mócsai, G.; Gáspár, K.; Nagy, G.; Irinyi, B.; Kapitány, A.; Bíró, T.; Gyimesi, E.; Tóth, B.; Maródi, L.; Szegedi, A. Severe skin inflammation and filaggrin mutation similarly alter the skin barrier in patients with atopic dermatitis. Br. J. Dermatol. 2014, 170, 617–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Szegedi, A. Filaggrin mutations in early- and late-onset atopic dermatitis. Br. J. Dermatol. 2015, 172, 320–321. [Google Scholar] [CrossRef] [PubMed]
  43. Irvine, A.D.; McLean, W.H.I.; Leung, D.Y.M. Filaggrin mutations associated with skin and allergic diseases. N. Engl. J. Med. 2011, 365, 1315–1327. [Google Scholar] [CrossRef] [PubMed]
  44. Riethmuller, C.; McAleer, M.A.; Koppes, S.A.; Abdayem, R.; Franz, J.; Haftek, M.; Campbell, L.E.; MacCallum, S.F.; McLean, W.H.I.; Irvine, A.D.; et al. Filaggrin breakdown products determine corneocyte conformation in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2015, 136, 1573–1580.e1572. [Google Scholar] [CrossRef]
  45. Fluhr, J.W.; Kao, J.; Jain, M.; Ahn, S.K.; Feingold, K.R.; Elias, P.M. Generation of free fatty acids from phospholipids regulates stratum corneum acidification and integrity. J. Investig. Dermatol. 2001, 117, 44–51. [Google Scholar] [CrossRef]
  46. Fluhr, J.W.; Elias, P.M. Stratum corneum pH: Formation and Function of the ‘Acid Mantle’. Exog. Dermatol. 2002, 1, 163–175. [Google Scholar] [CrossRef]
  47. Jang, H.; Matsuda, A.; Jung, K.; Karasawa, K.; Matsuda, K.; Oida, K.; Ishizaka, S.; Ahn, G.; Amagai, Y.; Moon, C.; et al. Skin pH Is the Master Switch of Kallikrein 5-Mediated Skin Barrier Destruction in a Murine Atopic Dermatitis Model. J. Investig. Dermatol. 2016, 136, 127–135. [Google Scholar] [CrossRef]
  48. Brattsand, M.; Stefansson, K.; Lundh, C.; Haasum, Y.; Egelrud, T. A proteolytic cascade of kallikreins in the stratum corneum. J. Investig. Dermatol. 2005, 124, 198–203. [Google Scholar] [CrossRef]
  49. Cork, M.J.; Danby, S.G.; Vasilopoulos, Y.; Hadgraft, J.; Lane, M.E.; Moustafa, M.; Guy, R.H.; Macgowan, A.L.; Tazi-Ahnini, R.; Ward, S.J. Epidermal barrier dysfunction in atopic dermatitis. J. Investig. Dermatol. 2009, 129, 1892–1908. [Google Scholar] [CrossRef]
  50. Sparavigna, A.; Setaro, M.; Gualandri, V. Cutaneous pH in children affected by atopic dermatitis and in healthy children: A multicenter study. Skin Res. Technol. 1999, 5, 221–227. [Google Scholar] [CrossRef]
  51. Eberlein-König, B.; Schäfer, T.; Huss-Marp, J.; Darsow, U.; Möhrenschlager, M.; Herbert, O.; Abeck, D.; Krämer, U.; Behrendt, H.; Ring, J. Skin surface pH, stratum corneum hydration, trans-epidermal water loss and skin roughness related to atopic eczema and skin dryness in a population of primary school children. Acta Derm. Venereol. 2000, 80, 188–191. [Google Scholar] [CrossRef] [PubMed]
  52. Tsukita, S.; Yamazaki, Y.; Katsuno, T.; Tamura, A. Tight junction-based epithelial microenvironment and cell proliferation. Oncogene 2008, 27, 6930–6938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yuki, T.; Tobiishi, M.; Kusaka-Kikushima, A.; Ota, Y.; Tokura, Y. Impaired Tight Junctions in Atopic Dermatitis Skin and in a Skin-Equivalent Model Treated with Interleukin-17. PLoS ONE 2016, 11, e0161759. [Google Scholar] [CrossRef] [PubMed]
  54. Yuki, T.; Komiya, A.; Kusaka, A.; Kuze, T.; Sugiyama, Y.; Inoue, S. Impaired tight junctions obstruct stratum corneum formation by altering polar lipid and profilaggrin processing. J. Dermatol. Sci. 2013, 69, 148–158. [Google Scholar] [CrossRef]
  55. De Benedetto, A.; Rafaels, N.M.; McGirt, L.Y.; Ivanov, A.I.; Georas, S.N.; Cheadle, C.; Berger, A.E.; Zhang, K.; Vidyasagar, S.; Yoshida, T.; et al. Tight junction defects in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2011, 127, 773–786.e7. [Google Scholar] [CrossRef] [Green Version]
  56. Hartwig, I.R.V.; Sly, P.D.; Schmidt, L.A.; van Lieshout, R.J.; Bienenstock, J.; Holt, P.G.; Arck, P.C. Prenatal adverse life events increase the risk for atopic diseases in children, which is enhanced in the absence of a maternal atopic predisposition. J. Allergy Clin. Immunol. 2014, 134, 160–169. [Google Scholar] [CrossRef]
  57. Nakatsuji, T.; Chen, T.H.; Two, A.M.; Chun, K.A.; Narala, S.; Geha, R.S.; Hata, T.R.; Gallo, R.L. Staphylococcus aureus Exploits Epidermal Barrier Defects in Atopic Dermatitis to Trigger Cytokine Expression. J. Investig. Dermatol. 2016, 136, 2192–2200. [Google Scholar] [CrossRef] [Green Version]
  58. Engebretsen, K.A.; Johansen, J.D.; Kezic, S.; Linneberg, A.; Thyssen, J.P. The effect of environmental humidity and temperature on skin barrier function and dermatitis. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 223–249. [Google Scholar] [CrossRef]
  59. Perkin, M.R.; Craven, J.; Logan, K.; Strachan, D.; Marrs, T.; Radulovic, S.; Campbell, L.E.; MacCallum, S.F.; McLean, W.H.I.; Lack, G.; et al. Association between domestic water hardness, chlorine, and atopic dermatitis risk in early life: A population-based cross-sectional study. J. Allergy Clin. Immunol. 2016, 138, 509–516. [Google Scholar] [CrossRef] [Green Version]
  60. McNally, N.J.; Williams, H.C.; Phillips, D.R.; Smallman-Raynor, M.; Lewis, S.; Venn, A.; Britton, J. Atopic eczema and domestic water hardness. Lancet 1998, 352, 527–531. [Google Scholar] [CrossRef]
  61. Kathuria, P.; Silverberg, J.I. Association of pollution and climate with atopic eczema in US children. Pediatric Allergy Immunol. 2016, 27, 478–485. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, J.; Han, Y.; Ahn, J.H.; Kim, S.W.; Lee, S.I.; Lee, K.H.; Ahn, K. Airborne formaldehyde causes skin barrier dysfunction in atopic dermatitis. Br. J. Dermatol. 2016, 175, 357–363. [Google Scholar] [CrossRef] [PubMed]
  63. Ahn, K. The role of air pollutants in atopic dermatitis. J. Allergy Clin. Immunol. 2014, 134, 993–999, discussion 1000. [Google Scholar] [CrossRef] [PubMed]
  64. Kantor, R.; Kim, A.; Thyssen, J.P.; Silverberg, J.I. Association of atopic dermatitis with smoking: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2016, 75, 1119–1125.e1111. [Google Scholar] [CrossRef] [PubMed]
  65. Gfatter, R.; Hackl, P.; Braun, F. Effects of soap and detergents on skin surface pH, stratum corneum hydration and fat content in infants. Dermatology 1997, 195, 258–262. [Google Scholar] [CrossRef]
  66. Angelova-Fischer, I.; Dapic, I.; Hoek, A.-K.; Jakasa, I.; Fischer, T.W.; Zillikens, D.; Kezic, S. Skin barrier integrity and natural moisturising factor levels after cumulative dermal exposure to alkaline agents in atopic dermatitis. Acta Derm. Venereol. 2014, 94, 640–644. [Google Scholar] [CrossRef] [PubMed]
  67. Nassif, A.; Chan, S.C.; Storrs, F.J.; Hanifin, J.M. Abnormal skin irritancy in atopic dermatitis and in atopy without dermatitis. Arch. Dermatol. 1994, 130, 1402–1407. [Google Scholar] [CrossRef]
  68. Thyssen, J.P.; McFadden, J.P.; Kimber, I. The multiple factors affecting the association between atopic dermatitis and contact sensitization. Allergy 2014, 69, 28–36. [Google Scholar] [CrossRef]
  69. Kantor, R.; Silverberg, J.I. Environmental risk factors and their role in the management of atopic dermatitis. Expert Rev. Clin. Immunol. 2017, 13, 15–26. [Google Scholar] [CrossRef]
  70. Czarnowicki, T.; Krueger, J.G.; Guttman-Yassky, E. Skin barrier and immune dysregulation in atopic dermatitis: An evolving story with important clinical implications. J. Allergy Clin. Immunol. Pract. 2014, 2, 371–379. [Google Scholar] [CrossRef]
  71. Schulz, F.; Marenholz, I.; Fölster-Holst, R.; Chen, C.; Sternjak, A.; Baumgrass, R.; Esparza-Gordillo, J.; Grüber, C.; Nickel, R.; Schreiber, S.; et al. A common haplotype of the IL-31 gene influencing gene expression is associated with nonatopic eczema. J. Allergy Clin. Immunol. 2007, 120, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  72. Oiso, N.; Fukai, K.; Ishii, M. Interleukin 4 receptor alpha chain polymorphism Gln551Arg is associated with adult atopic dermatitis in Japan. Br. J. Dermatol. 2000, 142, 1003–1006. [Google Scholar] [CrossRef] [PubMed]
  73. Novak, N.; Kruse, S.; Potreck, J.; Maintz, L.; Jenneck, C.; Weidinger, S.; Fimmers, R.; Bieber, T. Single nucleotide polymorphisms of the IL18 gene are associated with atopic eczema. J. Allergy Clin. Immunol. 2005, 115, 828–833. [Google Scholar] [CrossRef] [PubMed]
  74. Hanifin, J.M. Evolving concepts of pathogenesis in atopic dermatitis and other eczemas. J. Investig. Dermatol. 2009, 129, 320–322. [Google Scholar] [CrossRef]
  75. Malajian, D.; Guttman-Yassky, E. New pathogenic and therapeutic paradigms in atopic dermatitis. Cytokine 2015, 73, 311–318. [Google Scholar] [CrossRef]
  76. Niebuhr, M.; Langnickel, J.; Draing, C.; Renz, H.; Kapp, A.; Werfel, T. Dysregulation of toll-like receptor-2 (TLR-2)-induced effects in monocytes from patients with atopic dermatitis: Impact of the TLR-2 R753Q polymorphism. Allergy 2008, 63, 728–734. [Google Scholar] [CrossRef]
  77. Ahmad-Nejad, P.; Mrabet-Dahbi, S.; Breuer, K.; Klotz, M.; Werfel, T.; Herz, U.; Heeg, K.; Neumaier, M.; Renz, H. The toll-like receptor 2 R753Q polymorphism defines a subgroup of patients with atopic dermatitis having severe phenotype. J. Allergy Clin. Immunol. 2004, 113, 565–567. [Google Scholar] [CrossRef]
  78. Ong, P.Y.; Ohtake, T.; Brandt, C.; Strickland, I.; Boguniewicz, M.; Ganz, T.; Gallo, R.L.; Leung, D.Y.M. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N. Engl. J. Med. 2002, 347, 1151–1160. [Google Scholar] [CrossRef]
  79. Hata, T.R.; Gallo, R.L. Antimicrobial peptides, skin infections, and atopic dermatitis. Semin. Cutan. Med. Surg. 2008, 27, 144–150. [Google Scholar] [CrossRef]
  80. Kobayashi, T.; Glatz, M.; Horiuchi, K.; Kawasaki, H.; Akiyama, H.; Kaplan, D.H.; Kong, H.H.; Amagai, M.; Nagao, K. Dysbiosis and Staphylococcus aureus Colonization Drives Inflammation in Atopic Dermatitis. Immunity 2015, 42, 756–766. [Google Scholar] [CrossRef] [Green Version]
  81. Johnson-Huang, L.M.; McNutt, N.S.; Krueger, J.G.; Lowes, M.A. Cytokine-producing dendritic cells in the pathogenesis of inflammatory skin diseases. J. Clin. Immunol. 2009, 29, 247–256. [Google Scholar] [CrossRef] [PubMed]
  82. Guttman-Yassky, E.; Lowes, M.A.; Fuentes-Duculan, J.; Whynot, J.; Novitskaya, I.; Cardinale, I.; Haider, A.; Khatcherian, A.; Carucci, J.A.; Bergman, R.; et al. Major differences in inflammatory dendritic cells and their products distinguish atopic dermatitis from psoriasis. J. Allergy Clin. Immunol. 2007, 119, 1210–1217. [Google Scholar] [CrossRef] [PubMed]
  83. Hammad, H.; Lambrecht, B.N. Barrier Epithelial Cells and the Control of Type 2 Immunity. Immunity 2015, 43, 29–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Liu, F.-T.; Goodarzi, H.; Chen, H.-Y. IgE, mast cells, and eosinophils in atopic dermatitis. Clin. Rev. Allergy Immunol. 2011, 41, 298–310. [Google Scholar] [CrossRef]
  85. Leung, D.Y.M.; Boguniewicz, M.; Howell, M.D.; Nomura, I.; Hamid, Q.A. New insights into atopic dermatitis. J. Clin. Investig. 2004, 113, 651–657. [Google Scholar] [CrossRef]
  86. Gittler, J.K.; Shemer, A.; Suárez-Fariñas, M.; Fuentes-Duculan, J.; Gulewicz, K.J.; Wang, C.Q.F.; Mitsui, H.; Cardinale, I.; de Guzman Strong, C.; Krueger, J.G.; et al. Progressive activation of T(H)2/T(H)22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. J. Allergy Clin. Immunol. 2012, 130, 1344–1354. [Google Scholar] [CrossRef]
  87. Howell, M.D.; Fairchild, H.R.; Kim, B.E.; Bin, L.; Boguniewicz, M.; Redzic, J.S.; Hansen, K.C.; Leung, D.Y.M. Th2 cytokines act on S100/A11 to downregulate keratinocyte differentiation. J. Investig. Dermatol. 2008, 128, 2248–2258. [Google Scholar] [CrossRef]
  88. Rebane, A.; Zimmermann, M.; Aab, A.; Baurecht, H.; Koreck, A.; Karelson, M.; Abram, K.; Metsalu, T.; Pihlap, M.; Meyer, N.; et al. Mechanisms of IFN-γ-induced apoptosis of human skin keratinocytes in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2012, 129, 1297–1306. [Google Scholar] [CrossRef]
  89. Nograles, K.E.; Zaba, L.C.; Shemer, A.; Fuentes-Duculan, J.; Cardinale, I.; Kikuchi, T.; Ramon, M.; Bergman, R.; Krueger, J.G.; Guttman-Yassky, E. IL-22-producing “T22” T cells account for upregulated IL-22 in atopic dermatitis despite reduced IL-17-producing TH17 T cells. J. Allergy Clin. Immunol. 2009, 123, 1244–1252. [Google Scholar] [CrossRef]
  90. Czarnowicki, T.; Malajian, D.; Khattri, S.; Correa da Rosa, J.; Dutt, R.; Finney, R.; Dhingra, N.; Xiangyu, P.; Xu, H.; Estrada, Y.D.; et al. Petrolatum: Barrier repair and antimicrobial responses underlying this “inert” moisturizer. J. Allergy Clin. Immunol. 2016, 137, 1091–1102.e1097. [Google Scholar] [CrossRef]
  91. Eichenfield, L.F.; Tom, W.L.; Berger, T.G.; Krol, A.; Paller, A.S.; Schwarzenberger, K.; Bergman, J.N.; Chamlin, S.L.; Cohen, D.E.; Cooper, K.D.; et al. Guidelines of care for the management of atopic dermatitis: Section 2. Management and treatment of atopic dermatitis with topical therapies. J. Am. Acad. Dermatol. 2014, 71, 116–132. [Google Scholar] [CrossRef] [PubMed]
  92. Cury Martins, J.; Martins, C.; Aoki, V.; Gois, A.F.T.; Ishii, H.A.; da Silva, E.M.K. Topical tacrolimus for atopic dermatitis. Cochrane Database Syst. Rev. 2015, 36, CD009864. [Google Scholar] [CrossRef] [PubMed]
  93. Sidbury, R.; Davis, D.M.; Cohen, D.E.; Cordoro, K.M.; Berger, T.G.; Bergman, J.N.; Chamlin, S.L.; Cooper, K.D.; Feldman, S.R.; Hanifin, J.M.; et al. Guidelines of care for the management of atopic dermatitis: Section 3. Management and Treatment with Phototherapy and Systemic Agents; J. Am. Acad. Dermatol. 2014, 71, 327–349. [Google Scholar] [CrossRef] [PubMed]
  94. Yu, S.H.; Drucker, A.M.; Lebwohl, M.; Silverberg, J.I. A systematic review of the safety and efficacy of systemic corticosteroids in atopic dermatitis. J. Am. Acad. Dermatol. 2018, 78, 733–740. [Google Scholar] [CrossRef] [PubMed]
  95. Roekevisch, E.; Spuls, P.I.; Kuester, D.; Limpens, J.; Schmitt, J. Efficacy and safety of systemic treatments for moderate-to-severe atopic dermatitis: A systematic review. J. Allergy Clin. Immunol. 2014, 133, 429–438. [Google Scholar] [CrossRef]
  96. Jimenez, J.L.; Punzón, C.; Navarro, J.; Muñoz-Fernández, M.A.; Fresno, M. Phosphodiesterase 4 inhibitors prevent cytokine secretion by T lymphocytes by inhibiting nuclear factor-kappaB and nuclear factor of activated T cells activation. J. Pharmacol. Exp. Ther. 2001, 299, 753–759. [Google Scholar]
  97. Paller, A.S.; Tom, W.L.; Lebwohl, M.G.; Blumenthal, R.L.; Boguniewicz, M.; Call, R.S.; Eichenfield, L.F.; Forsha, D.W.; Rees, W.C.; Simpson, E.L.; et al. Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J. Am. Acad. Dermatol. 2016, 75, 494–503.e496. [Google Scholar] [CrossRef]
  98. Hanifin, J.M.; Ellis, C.N.; Frieden, I.J.; Fölster-Holst, R.; Stein Gold, L.F.; Secci, A.; Smith, A.J.; Zhao, C.; Kornyeyeva, E.; Eichenfield, L.F. OPA-15406, a novel, topical, nonsteroidal, selective phosphodiesterase-4 (PDE4) inhibitor, in the treatment of adult and adolescent patients with mild to moderate atopic dermatitis (AD): A phase-II randomized, double-blind, placebo-controlled study. J. Am. Acad. Dermatol. 2016, 75, 297–305. [Google Scholar] [CrossRef] [Green Version]
  99. Simpson, E.L.; Imafuku, S.; Poulin, Y.; Ungar, B.; Zhou, L.; Malik, K.; Wen, H.-C.; Xu, H.; Estrada, Y.D.; Peng, X.; et al. A Phase 2 Randomized Trial of Apremilast in Patients with Atopic Dermatitis. J. Investig. Dermatol. 2019, 139, 1063–1072. [Google Scholar] [CrossRef] [Green Version]
  100. Simpson, E.L.; Gadkari, A.; Worm, M.; Soong, W.; Blauvelt, A.; Eckert, L.; Wu, R.; Ardeleanu, M.; Graham, N.M.H.; Pirozzi, G.; et al. Dupilumab therapy provides clinically meaningful improvement in patient-reported outcomes (PROs): A phase IIb, randomized, placebo-controlled, clinical trial in adult patients with moderate to severe atopic dermatitis (AD). J. Am. Acad. Dermatol. 2016, 75, 506–515. [Google Scholar] [CrossRef]
  101. Thaçi, D.; Simpson, E.L.; Beck, L.A.; Bieber, T.; Blauvelt, A.; Papp, K.; Soong, W.; Worm, M.; Szepietowski, J.C.; Sofen, H.; et al. Efficacy and safety of dupilumab in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical treatments: A randomised, placebo-controlled, dose-ranging phase 2b trial. Lancet 2016, 387, 40–52. [Google Scholar] [CrossRef]
  102. Beck, L.A.; Thaçi, D.; Hamilton, J.D.; Graham, N.M.; Bieber, T.; Rocklin, R.; Ming, J.E.; Ren, H.; Kao, R.; Simpson, E.; et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N. Engl. J. Med. 2014, 371, 130–139. [Google Scholar] [CrossRef] [PubMed]
  103. Simpson, E.L.; Bieber, T.; Guttman-Yassky, E.; Beck, L.A.; Blauvelt, A.; Cork, M.J.; Silverberg, J.I.; Deleuran, M.; Kataoka, Y.; Lacour, J.-P.; et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N. Engl. J. Med. 2016, 375, 2335–2348. [Google Scholar] [CrossRef] [PubMed]
  104. Tsoi, L.C.; Rodriguez, E.; Degenhardt, F.; Baurecht, H.; Wehkamp, U.; Volks, N.; Szymczak, S.; Swindell, W.R.; Sarkar, M.K.; Raja, K.; et al. Atopic Dermatitis Is an IL-13-Dominant Disease with Greater Molecular Heterogeneity Compared to Psoriasis. J. Investig. Dermatol. 2019, 139, 1480–1489. [Google Scholar] [CrossRef] [PubMed]
  105. Simpson, E.L.; Flohr, C.; Eichenfield, L.F.; Bieber, T.; Sofen, H.; Taïeb, A.; Owen, R.; Putnam, W.; Castro, M.; DeBusk, K.; et al. Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). J. Am. Acad. Dermatol. 2018, 78, 863–871.e811. [Google Scholar] [CrossRef] [PubMed]
  106. Wollenberg, A.; Howell, M.D.; Guttman-Yassky, E.; Silverberg, J.I.; Kell, C.; Ranade, K.; Moate, R.; van der Merwe, R. Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J. Allergy Clin. Immunol. 2019, 143, 135–141. [Google Scholar] [CrossRef] [PubMed]
  107. Nemoto, O.; Furue, M.; Nakagawa, H.; Shiramoto, M.; Hanada, R.; Matsuki, S.; Imayama, S.; Kato, M.; Hasebe, I.; Taira, K.; et al. The first trial of CIM331, a humanized antihuman interleukin-31 receptor A antibody, in healthy volunteers and patients with atopic dermatitis to evaluate safety, tolerability and pharmacokinetics of a single dose in a randomized, double-blind, placebo-controlled study. Br. J. Dermatol. 2016, 174, 296–304. [Google Scholar] [CrossRef] [PubMed]
  108. Ruzicka, T.; Hanifin, J.M.; Furue, M.; Pulka, G.; Mlynarczyk, I.; Wollenberg, A.; Galus, R.; Etoh, T.; Mihara, R.; Yoshida, H.; et al. Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis. N. Engl. J. Med. 2017, 376, 826–835. [Google Scholar] [CrossRef]
  109. Cornelissen, C.; Lüscher-Firzlaff, J.; Baron, J.M.; Lüscher, B. Signaling by IL-31 and functional consequences. Eur. J. Cell Biol. 2012, 91, 552–566. [Google Scholar] [CrossRef]
  110. Kelly-Welch, A.E.; Hanson, E.M.; Boothby, M.R.; Keegan, A.D. Interleukin-4 and interleukin-13 signaling connections maps. Science 2003, 300, 1527–1528. [Google Scholar] [CrossRef]
  111. Levy, L.L.; Urban, J.; King, B.A. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J. Am. Acad. Dermatol. 2015, 73, 395–399. [Google Scholar] [CrossRef] [PubMed]
  112. Guttman-Yassky, E.; Silverberg, J.I.; Nemoto, O.; Forman, S.B.; Wilke, A.; Prescilla, R.; de la Peña, A.; Nunes, F.P.; Janes, J.; Gamalo, M.; et al. Baricitinib in adult patients with moderate-to-severe atopic dermatitis: A phase 2 parallel, double-blinded, randomized placebo-controlled multiple-dose study. J. Am. Acad. Dermatol. 2019, 80, 913–921.e919. [Google Scholar] [CrossRef] [PubMed]
  113. Beck, L.; Hong, C.; Hu, X.; Chen, S.; Calimlim, B.; Teixeira, H.; Guttman-Yassky, E. Upadacitinib effect on pruritus in moderate-to-severe atopic dermatitis; from a phase 2b randomized, placebo-controlled trial. Ann. Allergy Asthma Immunol. 2018, 121, S21. [Google Scholar] [CrossRef]
  114. Parisi, R.; Symmons, D.P.M.; Griffiths, C.E.M.; Ashcroft, D.M.; The Identification and Management of Psoriasis and Associated ComorbidiTy (IMPACT) Project Team. Global epidemiology of psoriasis: A systematic review of incidence and prevalence. J. Investig. Dermatol. 2013, 133, 377–385. [Google Scholar] [CrossRef] [PubMed]
  115. Lebwohl, M. Psoriasis. Lancet 2003, 361, 1197–1204. [Google Scholar] [CrossRef]
  116. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef]
  117. Nograles, K.E.; Davidovici, B.; Krueger, J.G. New insights in the immunologic basis of psoriasis. Semin. Cutan. Med. Surg. 2010, 29, 3–9. [Google Scholar] [CrossRef]
  118. Morizane, S.; Gallo, R.L. Antimicrobial peptides in the pathogenesis of psoriasis. J. Dermatol. 2012, 39, 225–230. [Google Scholar] [CrossRef]
  119. Lande, R.; Gregorio, J.; Facchinetti, V.; Chatterjee, B.; Wang, Y.-H.; Homey, B.; Cao, W.; Wang, Y.-H.; Su, B.; Nestle, F.O.; et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 2007, 449, 564–569. [Google Scholar] [CrossRef]
  120. Gregorio, J.; Meller, S.; Conrad, C.; Di Nardo, A.; Homey, B.; Lauerma, A.; Arai, N.; Gallo, R.L.; Digiovanni, J.; Gilliet, M. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J. Exp. Med. 2010, 207, 2921–2930. [Google Scholar] [CrossRef]
  121. Nestle, F.O.; Conrad, C.; Tun-Kyi, A.; Homey, B.; Gombert, M.; Boyman, O.; Burg, G.; Liu, Y.-J.; Gilliet, M. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J. Exp. Med. 2005, 202, 135–143. [Google Scholar] [CrossRef] [PubMed]
  122. Morizane, S.; Yamasaki, K.; Mühleisen, B.; Kotol, P.F.; Murakami, M.; Aoyama, Y.; Iwatsuki, K.; Hata, T.; Gallo, R.L. Cathelicidin antimicrobial peptide LL-37 in psoriasis enables keratinocyte reactivity against TLR9 ligands. J. Investig. Dermatol. 2012, 132, 135–143. [Google Scholar] [CrossRef] [PubMed]
  123. Nestle, F.O.; Turka, L.A.; Nickoloff, B.J. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. J. Clin. Investig. 1994, 94, 202–209. [Google Scholar] [CrossRef] [PubMed]
  124. Uyemura, K.; Yamamura, M.; Fivenson, D.F.; Modlin, R.L.; Nickoloff, B.J. The cytokine network in lesional and lesion-free psoriatic skin is characterized by a T-helper type 1 cell-mediated response. J. Investig. Dermatol. 1993, 101, 701–705. [Google Scholar] [CrossRef] [PubMed]
  125. Lowes, M.A.; Kikuchi, T.; Fuentes-Duculan, J.; Cardinale, I.; Zaba, L.C.; Haider, A.S.; Bowman, E.P.; Krueger, J.G. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. J. Investig. Dermatol. 2008, 128, 1207–1211. [Google Scholar] [CrossRef] [PubMed]
  126. Zaba, L.C.; Cardinale, I.; Gilleaudeau, P.; Sullivan-Whalen, M.; Suárez-Fariñas, M.; Fuentes-Duculan, J.; Novitskaya, I.; Khatcherian, A.; Bluth, M.J.; Lowes, M.A.; et al. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J. Exp. Med. 2007, 204, 3183–3194. [Google Scholar] [CrossRef]
  127. Gaffen, S.L. Structure and signalling in the IL-17 receptor family. Nat. Rev. Immunol. 2009, 9, 556–567. [Google Scholar] [CrossRef] [Green Version]
  128. Zheng, Y.; Danilenko, D.M.; Valdez, P.; Kasman, I.; Eastham-Anderson, J.; Wu, J.; Ouyang, W. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 2007, 445, 648–651. [Google Scholar] [CrossRef]
  129. Johnston, A.; Xing, X.; Wolterink, L.; Barnes, D.H.; Yin, Z.; Reingold, L.; Kahlenberg, J.M.; Harms, P.W.; Gudjonsson, J.E. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J. Allergy Clin. Immunol. 2017, 140, 109–120. [Google Scholar] [CrossRef]
  130. Tauber, M.; Bal, E.; Pei, X.-Y.; Madrange, M.; Khelil, A.; Sahel, H.; Zenati, A.; Makrelouf, M.; Boubridaa, K.; Chiali, A.; et al. IL36RN Mutations Affect Protein Expression and Function: A Basis for Genotype-Phenotype Correlation in Pustular Diseases. J. Investig. Dermatol. 2016, 136, 1811–1819. [Google Scholar] [CrossRef] [Green Version]
  131. McFadden, J.; Valdimarsson, H.; Fry, L. Cross-reactivity between streptococcal M surface antigen and human skin. Br. J. Dermatol. 1991, 125, 443–447. [Google Scholar] [CrossRef] [PubMed]
  132. Leung, D.Y.; Travers, J.B.; Giorno, R.; Norris, D.A.; Skinner, R.; Aelion, J.; Kazemi, L.V.; Kim, M.H.; Trumble, A.E.; Kotb, M. Evidence for a streptococcal superantigen-driven process in acute guttate psoriasis. J. Clin. Investig. 1995, 96, 2106–2112. [Google Scholar] [CrossRef] [PubMed]
  133. Farber, E.M.; Nall, M.L. The natural history of psoriasis in 5,600 patients. Dermatologica 1974, 148, 1–18. [Google Scholar] [CrossRef] [PubMed]
  134. Farber, E.M.; Nall, M.L.; Watson, W. Natural history of psoriasis in 61 twin pairs. Arch. Dermatol. 1974, 109, 207–211. [Google Scholar] [CrossRef]
  135. Lønnberg, A.S.; Skov, L.; Skytthe, A.; Kyvik, K.O.; Pedersen, O.B.; Thomsen, S.F. Heritability of psoriasis in a large twin sample. Br. J. Dermatol. 2013, 169, 412–416. [Google Scholar] [CrossRef]
  136. Bowcock, A.M.; Krueger, J.G. Getting under the skin: The immunogenetics of psoriasis. Nat. Rev. Immunol. 2005, 5, 699–711. [Google Scholar] [CrossRef]
  137. Gudjonsson, J.E.; Elder, J.T. Psoriasis: Epidemiology. Clin. Dermatol. 2007, 25, 535–546. [Google Scholar] [CrossRef]
  138. Trembath, R.C.; Clough, R.L.; Rosbotham, J.L.; Jones, A.B.; Camp, R.D.; Frodsham, A.; Browne, J.; Barber, R.; Terwilliger, J.; Lathrop, G.M.; et al. Identification of a major susceptibility locus on chromosome 6p and evidence for further disease loci revealed by a two stage genome-wide search in psoriasis. Hum. Mol. Genet. 1997, 6, 813–820. [Google Scholar] [CrossRef]
  139. Allen, M.H.; Veal, C.; Faassen, A.; Powis, S.H.; Vaughan, R.W.; Trembath, R.C.; Barker, J.N. A non-HLA gene within the MHC in psoriasis. Lancet 1999, 353, 1589–1590. [Google Scholar] [CrossRef]
  140. Asumalahti, K.; Laitinen, T.; Itkonen-Vatjus, R.; Lokki, M.L.; Suomela, S.; Snellman, E.; Saarialho-Kere, U.; Kere, J. A candidate gene for psoriasis near HLA-C, HCR (Pg8), is highly polymorphic with a disease-associated susceptibility allele. Hum. Mol. Genet. 2000, 9, 1533–1542. [Google Scholar] [CrossRef]
  141. Nair, R.P.; Stuart, P.E.; Nistor, I.; Hiremagalore, R.; Chia, N.V.C.; Jenisch, S.; Weichenthal, M.; Abecasis, G.R.; Lim, H.W.; Christophers, E.; et al. Sequence and haplotype analysis supports HLA-C as the psoriasis susceptibility 1 gene. Am. J. Hum. Genet. 2006, 78, 827–851. [Google Scholar] [CrossRef] [PubMed]
  142. Fan, X.; Yang, S.; Huang, W.; Wang, Z.-M.; Sun, L.-D.; Liang, Y.-H.; Gao, M.; Ren, Y.-Q.; Zhang, K.-Y.; Du, W.-H.; et al. Fine mapping of the psoriasis susceptibility locus PSORS1 supports HLA-C as the susceptibility gene in the Han Chinese population. PLoS Genet. 2008, 4, e1000038. [Google Scholar] [CrossRef] [PubMed]
  143. Allen, M.H.; Ameen, H.; Veal, C.; Evans, J.; Ramrakha-Jones, V.S.; Marsland, A.M.; Burden, A.D.; Griffiths, C.E.M.; Trembath, R.C.; Barker, J.N.W.N. The major psoriasis susceptibility locus PSORS1 is not a risk factor for late-onset psoriasis. J. Investig. Dermatol. 2005, 124, 103–106. [Google Scholar] [CrossRef] [PubMed]
  144. Asumalahti, K.; Ameen, M.; Suomela, S.; Hagforsen, E.; Michaëlsson, G.; Evans, J.; Munro, M.; Veal, C.; Allen, M.; Leman, J.; et al. Genetic analysis of PSORS1 distinguishes guttate psoriasis and palmoplantar pustulosis. J. Investig. Dermatol. 2003, 120, 627–632. [Google Scholar] [CrossRef]
  145. Tomfohrde, J.; Silverman, A.; Barnes, R.; Fernandez-Vina, M.A.; Young, M.; Lory, D.; Morris, L.; Wuepper, K.D.; Stastny, P.; Menter, A. Gene for familial psoriasis susceptibility mapped to the distal end of human chromosome 17q. Science 1994, 264, 1141–1145. [Google Scholar] [CrossRef]
  146. Jordan, C.T.; Cao, L.; Roberson, E.D.O.; Pierson, K.C.; Yang, C.-F.; Joyce, C.E.; Ryan, C.; Duan, S.; Helms, C.A.; Liu, Y.; et al. PSORS2 is due to mutations in CARD14. Am. J. Hum. Genet. 2012, 90, 784–795. [Google Scholar] [CrossRef]
  147. Capon, F.; Semprini, S.; Chimenti, S.; Fabrizi, G.; Zambruno, G.; Murgia, S.; Carcassi, C.; Fazio, M.; Mingarelli, R.; Dallapiccola, B.; et al. Fine mapping of the PSORS4 psoriasis susceptibility region on chromosome 1q21. J. Investig. Dermatol. 2001, 116, 728–730. [Google Scholar] [CrossRef]
  148. Lee, Y.A.; Rüschendorf, F.; Windemuth, C.; Schmitt-Egenolf, M.; Stadelmann, A.; Nürnberg, G.; Ständer, M.; Wienker, T.F.; Reis, A.; Traupe, H. Genomewide scan in german families reveals evidence for a novel psoriasis-susceptibility locus on chromosome 19p13. Am. J. Hum. Genet. 2000, 67, 1020–1024. [Google Scholar] [CrossRef]
  149. Veal, C.D.; Clough, R.L.; Barber, R.C.; Mason, S.; Tillman, D.; Ferry, B.; Jones, A.B.; Ameen, M.; Balendran, N.; Powis, S.H.; et al. Identification of a novel psoriasis susceptibility locus at 1p and evidence of epistasis between PSORS1 and candidate loci. J. Med Genet. 2001, 38, 7–13. [Google Scholar] [CrossRef] [Green Version]
  150. Elder, J.T. Expanded Genome-Wide Association Study Meta-Analysis of Psoriasis Expands the Catalog of Common Psoriasis-Associated Variants. J. Investig. Dermatol. Symp. Proc. 2018, 19, S77–S78. [Google Scholar] [CrossRef] [Green Version]
  151. Hirata, J.; Hirota, T.; Ozeki, T.; Kanai, M.; Sudo, T.; Tanaka, T.; Hizawa, N.; Nakagawa, H.; Sato, S.; Mushiroda, T.; et al. Variants at HLA-A, HLA-C, and HLA-DQB1 Confer Risk of Psoriasis Vulgaris in Japanese. J. Investig. Dermatol. 2018, 138, 542–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Okada, Y.; Han, B.; Tsoi, L.C.; Stuart, P.E.; Ellinghaus, E.; Tejasvi, T.; Chandran, V.; Pellett, F.; Pollock, R.; Bowcock, A.M.; et al. Fine mapping major histocompatibility complex associations in psoriasis and its clinical subtypes. Am. J. Hum. Genet. 2014, 95, 162–172. [Google Scholar] [CrossRef] [PubMed]
  153. Yin, X.; Low, H.Q.; Wang, L.; Li, Y.; Ellinghaus, E.; Han, J.; Estivill, X.; Sun, L.; Zuo, X.; Shen, C.; et al. Genome-wide meta-analysis identifies multiple novel associations and ethnic heterogeneity of psoriasis susceptibility. Nat. Commun. 2015, 6, 6916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Elder, J.T. Genome-wide association scan yields new insights into the immunopathogenesis of psoriasis. Genes Immun. 2009, 10, 201–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Tsoi, L.C.; Spain, S.L.; Knight, J.; Ellinghaus, E.; Stuart, P.E.; Capon, F.; Ding, J.; Li, Y.; Tejasvi, T.; Gudjonsson, J.E.; et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat. Genet. 2012, 44, 1341–1348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Tsoi, L.C.; Spain, S.L.; Ellinghaus, E.; Stuart, P.E.; Capon, F.; Knight, J.; Tejasvi, T.; Kang, H.M.; Allen, M.H.; Lambert, S.; et al. Enhanced meta-analysis and replication studies identify five new psoriasis susceptibility loci. Nat. Commun. 2015, 6, 7001. [Google Scholar] [CrossRef]
  157. Capon, F.; Di Meglio, P.; Szaub, J.; Prescott, N.J.; Dunster, C.; Baumber, L.; Timms, K.; Gutin, A.; Abkevic, V.; Burden, A.D.; et al. Sequence variants in the genes for the interleukin-23 receptor (IL23R) and its ligand (IL12B) confer protection against psoriasis. Hum. Genet. 2007, 122, 201–206. [Google Scholar] [CrossRef]
  158. Cargill, M.; Schrodi, S.J.; Chang, M.; Garcia, V.E.; Brandon, R.; Callis, K.P.; Matsunami, N.; Ardlie, K.G.; Civello, D.; Catanese, J.J.; et al. A large-scale genetic association study confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes. Am. J. Hum. Genet. 2007, 80, 273–290. [Google Scholar] [CrossRef]
  159. Hüffmeier, U.; Uebe, S.; Ekici, A.B.; Bowes, J.; Giardina, E.; Korendowych, E.; Juneblad, K.; Apel, M.; McManus, R.; Ho, P.; et al. Common variants at TRAF3IP2 are associated with susceptibility to psoriatic arthritis and psoriasis. Nat. Genet. 2010, 42, 996–999. [Google Scholar] [CrossRef] [Green Version]
  160. Nair, R.P.; Duffin, K.C.; Helms, C.; Ding, J.; Stuart, P.E.; Goldgar, D.; Gudjonsson, J.E.; Li, Y.; Tejasvi, T.; Feng, B.-J.; et al. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat. Genet. 2009, 41, 199–204. [Google Scholar] [CrossRef]
  161. Marrakchi, S.; Guigue, P.; Renshaw, B.R.; Puel, A.; Pei, X.-Y.; Fraitag, S.; Zribi, J.; Bal, E.; Cluzeau, C.; Chrabieh, M.; et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N. Engl. J. Med. 2011, 365, 620–628. [Google Scholar] [CrossRef] [PubMed]
  162. Onoufriadis, A.; Simpson, M.A.; Pink, A.E.; Di Meglio, P.; Smith, C.H.; Pullabhatla, V.; Knight, J.; Spain, S.L.; Nestle, F.O.; Burden, A.D.; et al. Mutations in IL36RN/IL1F5 are associated with the severe episodic inflammatory skin disease known as generalized pustular psoriasis. Am. J. Hum. Genet. 2011, 89, 432–437. [Google Scholar] [CrossRef] [PubMed]
  163. McGill, A.; Frank, A.; Emmett, N.; Turnbull, D.M.; Birch-Machin, M.A.; Reynolds, N.J. The anti-psoriatic drug anthralin accumulates in keratinocyte mitochondria, dissipates mitochondrial membrane potential, and induces apoptosis through a pathway dependent on respiratory competent mitochondria. FASEB J. 2005, 19, 1012–1014. [Google Scholar] [CrossRef] [PubMed]
  164. Arbiser, J.L.; Govindarajan, B.; Battle, T.E.; Lynch, R.; Frank, D.A.; Ushio-Fukai, M.; Perry, B.N.; Stern, D.F.; Bowden, G.T.; Liu, A.; et al. Carbazole is a naturally occurring inhibitor of angiogenesis and inflammation isolated from antipsoriatic coal tar. J. Investig. Dermatol. 2006, 126, 1396–1402. [Google Scholar] [CrossRef]
  165. Castela, E.; Archier, E.; Devaux, S.; Gallini, A.; Aractingi, S.; Cribier, B.; Jullien, D.; Aubin, F.; Bachelez, H.; Joly, P.; et al. Topical corticosteroids in plaque psoriasis: A systematic review of efficacy and treatment modalities. J. Eur. Acad. Dermatol. Venereol. 2012, 26 (Suppl. 3), 36–46. [Google Scholar] [CrossRef]
  166. Samarasekera, E.J.; Sawyer, L.; Wonderling, D.; Tucker, R.; Smith, C.H. Topical therapies for the treatment of plaque psoriasis: Systematic review and network meta-analyses. Br. J. Dermatol. 2013, 168, 954–967. [Google Scholar] [CrossRef]
  167. Trémezaygues, L.; Reichrath, J. Vitamin D analogs in the treatment of psoriasis: Where are we standing and where will we be going? Derm. Endocrinol. 2011, 3, 180–186. [Google Scholar] [CrossRef]
  168. Devaux, S.; Castela, A.; Archier, E.; Gallini, A.; Joly, P.; Misery, L.; Aractingi, S.; Aubin, F.; Bachelez, H.; Cribier, B.; et al. Topical vitamin D analogues alone or in association with topical steroids for psoriasis: A systematic review. J. Eur. Acad. Dermatol. Venereol. 2012, 26 (Suppl. 3), 52–60. [Google Scholar] [CrossRef]
  169. Lebwohl, M.; Siskin, S.B.; Epinette, W.; Breneman, D.; Funicella, T.; Kalb, R.; Moore, J. A multicenter trial of calcipotriene ointment and halobetasol ointment compared with either agent alone for the treatment of psoriasis. J. Am. Acad. Dermatol. 1996, 35, 268–269. [Google Scholar] [CrossRef]
  170. Gerritsen, M.J.; Van De Kerkhof, P.C.; Langner, A. Long-term safety of topical calcitriol 3 microg g(-1) ointment. Br. J. Dermatol. 2001, 144 (Suppl. 58), 17–19. [Google Scholar] [CrossRef]
  171. Wang, C.; Lin, A. Efficacy of topical calcineurin inhibitors in psoriasis. J. Cutan. Med. Surg. 2014, 18, 8–14. [Google Scholar] [CrossRef] [PubMed]
  172. Kreuter, A.; Sommer, A.; Hyun, J.; Bräutigam, M.; Brockmeyer, N.H.; Altmeyer, P.; Gambichler, T. 1% pimecrolimus, 0.005% calcipotriol, and 0.1% betamethasone in the treatment of intertriginous psoriasis: A double-blind, randomized controlled study. Arch. Dermatol. 2006, 142, 1138–1143. [Google Scholar] [CrossRef] [PubMed]
  173. Duvic, M.; Asano, A.T.; Hager, C.; Mays, S. The pathogenesis of psoriasis and the mechanism of action of tazarotene. J. Am. Acad. Dermatol. 1998, 39, S129–S133. [Google Scholar] [CrossRef]
  174. Gold, L.S.; Lebwohl, M.G.; Sugarman, J.L.; Pariser, D.M.; Lin, T.; Martin, G.; Pillai, R.; Israel, R.; Ramakrishna, T. Safety and efficacy of a fixed combination of halobetasol and tazarotene in the treatment of moderate-to-severe plaque psoriasis: Results of 2 phase 3 randomized controlled trials. J. Am. Acad. Dermatol. 2018, 79, 287–293. [Google Scholar] [CrossRef] [Green Version]
  175. Lebwohl, M.G.; Breneman, D.L.; Goffe, B.S.; Grossman, J.R.; Ling, M.R.; Milbauer, J.; Pincus, S.H.; Sibbald, R.G.; Swinyer, L.J.; Weinstein, G.D.; et al. Tazarotene 0.1% gel plus corticosteroid cream in the treatment of plaque psoriasis. J. Am. Acad. Dermatol. 1998, 39, 590–596. [Google Scholar] [CrossRef]
  176. Zhang, P.; Wu, M.X. A clinical review of phototherapy for psoriasis. Lasers Med. Sci. 2018, 33, 173–180. [Google Scholar] [CrossRef]
  177. Mehta, D.; Lim, H.W. Ultraviolet B Phototherapy for Psoriasis: Review of Practical Guidelines. Am. J. Clin. Dermatol. 2016, 17, 125–133. [Google Scholar] [CrossRef]
  178. Stern, R.S.; Study, P.F.-U. The risk of melanoma in association with long-term exposure to PUVA. J. Am. Acad. Dermatol. 2001, 44, 755–761. [Google Scholar] [CrossRef]
  179. Stern, R.S.; Study, P.F.-U. The risk of squamous cell and basal cell cancer associated with psoralen and ultraviolet A therapy: A 30-year prospective study. J. Am. Acad. Dermatol. 2012, 66, 553–562. [Google Scholar] [CrossRef]
  180. Sbidian, E.; Maza, A.; Montaudié, H.; Gallini, A.; Aractingi, S.; Aubin, F.; Cribier, B.; Joly, P.; Jullien, D.; Le Maître, M.; et al. Efficacy and safety of oral retinoids in different psoriasis subtypes: A systematic literature review. J. Eur. Acad. Dermatol. Venereol. 2011, 25 (Suppl. 2), 28–33. [Google Scholar] [CrossRef]
  181. Lee, C.S.; Koo, J. A review of acitretin, a systemic retinoid for the treatment of psoriasis. Expert Opin. Pharmacother. 2005, 6, 1725–1734. [Google Scholar] [CrossRef] [PubMed]
  182. Schafer, P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem. Pharmacol. 2012, 83, 1583–1590. [Google Scholar] [CrossRef] [PubMed]
  183. Reich, K.; Gooderham, M.; Green, L.; Bewley, A.; Zhang, Z.; Khanskaya, I.; Day, R.M.; Goncalves, J.; Shah, K.; Piguet, V.; et al. The efficacy and safety of apremilast, etanercept and placebo in patients with moderate-to-severe plaque psoriasis: 52-week results from a phase IIIb, randomized, placebo-controlled trial (LIBERATE). J. Eur. Acad. Dermatol. Venereol. 2017, 31, 507–517. [Google Scholar] [CrossRef] [PubMed]
  184. Bissonnette, R.; Haydey, R.; Rosoph, L.A.; Lynde, C.W.; Bukhalo, M.; Fowler, J.F.; Delorme, I.; Gagné-Henley, A.; Gooderham, M.; Poulin, Y.; et al. Apremilast for the treatment of moderate-to-severe palmoplantar psoriasis: Results from a double-blind, placebo-controlled, randomized study. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 403–410. [Google Scholar] [CrossRef] [PubMed]
  185. Papp, K.; Reich, K.; Leonardi, C.L.; Kircik, L.; Chimenti, S.; Langley, R.G.B.; Hu, C.; Stevens, R.M.; Day, R.M.; Gordon, K.B.; et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: Results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J. Am. Acad. Dermatol. 2015, 73, 37–49. [Google Scholar] [CrossRef] [PubMed]
  186. Paul, C.; Cather, J.; Gooderham, M.; Poulin, Y.; Mrowietz, U.; Ferrandiz, C.; Crowley, J.; Hu, C.; Stevens, R.M.; Shah, K.; et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate-to-severe plaque psoriasis over 52 weeks: A phase III, randomized controlled trial (ESTEEM 2). Br. J. Dermatol. 2015, 173, 1387–1399. [Google Scholar] [CrossRef]
  187. Kaushik, S.B.; Lebwohl, M.G. Review of safety and efficacy of approved systemic psoriasis therapies. Int. J. Dermatol. 2019, 58, 649–658. [Google Scholar] [CrossRef]
  188. Tyring, S.; Gottlieb, A.; Papp, K.; Gordon, K.; Leonardi, C.; Wang, A.; Lalla, D.; Woolley, M.; Jahreis, A.; Zitnik, R.; et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: Double-blind placebo-controlled randomised phase III trial. Lancet 2006, 367, 29–35. [Google Scholar] [CrossRef]
  189. Papp, K.A.; Tyring, S.; Lahfa, M.; Prinz, J.; Griffiths, C.E.M.; Nakanishi, A.M.; Zitnik, R.; van de Kerkhof, P.C.M.; Melvin, L.; Group, E.P.S. A global phase III randomized controlled trial of etanercept in psoriasis: Safety, efficacy, and effect of dose reduction. Br. J. Dermatol. 2005, 152, 1304–1312. [Google Scholar] [CrossRef]
  190. Leonardi, C.L.; Powers, J.L.; Matheson, R.T.; Goffe, B.S.; Zitnik, R.; Wang, A.; Gottlieb, A.B.; Group, E.P.S. Etanercept as monotherapy in patients with psoriasis. N. Engl. J. Med. 2003, 349, 2014–2022. [Google Scholar] [CrossRef]
  191. Reich, K.; Nestle, F.O.; Papp, K.; Ortonne, J.-P.; Evans, R.; Guzzo, C.; Li, S.; Dooley, L.T.; Griffiths, C.E.M.; EXPRESS Study Investigators. Infliximab induction and maintenance therapy for moderate-to-severe psoriasis: A phase III, multicentre, double-blind trial. Lancet 2005, 366, 1367–1374. [Google Scholar] [CrossRef]
  192. Menter, A.; Feldman, S.R.; Weinstein, G.D.; Papp, K.; Evans, R.; Guzzo, C.; Li, S.; Dooley, L.T.; Arnold, C.; Gottlieb, A.B. A randomized comparison of continuous vs. intermittent infliximab maintenance regimens over 1 year in the treatment of moderate-to-severe plaque psoriasis. J. Am. Acad. Dermatol. 2007, 56, 31.e1–31.e15. [Google Scholar] [CrossRef] [PubMed]
  193. Gordon, K.; Papp, K.; Poulin, Y.; Gu, Y.; Rozzo, S.; Sasso, E.H. Long-term efficacy and safety of adalimumab in patients with moderate to severe psoriasis treated continuously over 3 years: Results from an open-label extension study for patients from REVEAL. J. Am. Acad. Dermatol. 2012, 66, 241–251. [Google Scholar] [CrossRef] [PubMed]
  194. Menter, A.; Tyring, S.K.; Gordon, K.; Kimball, A.B.; Leonardi, C.L.; Langley, R.G.; Strober, B.E.; Kaul, M.; Gu, Y.; Okun, M.; et al. Adalimumab therapy for moderate to severe psoriasis: A randomized, controlled phase III trial. J. Am. Acad. Dermatol. 2008, 58, 106–115. [Google Scholar] [CrossRef] [PubMed]
  195. Saurat, J.H.; Stingl, G.; Dubertret, L.; Papp, K.; Langley, R.G.; Ortonne, J.P.; Unnebrink, K.; Kaul, M.; Camez, A.; Investigators, C.S. Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br. J. Dermatol. 2008, 158, 558–566. [Google Scholar] [CrossRef] [PubMed]
  196. Gordon, K.B.; Langley, R.G.; Leonardi, C.; Toth, D.; Menter, M.A.; Kang, S.; Heffernan, M.; Miller, B.; Hamlin, R.; Lim, L.; et al. Clinical response to adalimumab treatment in patients with moderate to severe psoriasis: Double-blind, randomized controlled trial and open-label extension study. J. Am. Acad. Dermatol. 2006, 55, 598–606. [Google Scholar] [CrossRef]
  197. Gottlieb, A.B.; Blauvelt, A.; Thaçi, D.; Leonardi, C.L.; Poulin, Y.; Drew, J.; Peterson, L.; Arendt, C.; Burge, D.; Reich, K. Certolizumab pegol for the treatment of chronic plaque psoriasis: Results through 48 weeks from 2 phase 3, multicenter, randomized, double-blinded, placebo-controlled studies (CIMPASI-1 and CIMPASI-2). J. Am. Acad. Dermatol. 2018, 79, 302–314.e306. [Google Scholar] [CrossRef]
  198. Kimball, A.B.; Gordon, K.B.; Fakharzadeh, S.; Yeilding, N.; Szapary, P.O.; Schenkel, B.; Guzzo, C.; Li, S.; Papp, K.A. Long-term efficacy of ustekinumab in patients with moderate-to-severe psoriasis: Results from the PHOENIX 1 trial through up to 3 years. Br. J. Dermatol. 2012, 166, 861–872. [Google Scholar] [CrossRef]
  199. Papp, K.A.; Langley, R.G.; Lebwohl, M.; Krueger, G.G.; Szapary, P.; Yeilding, N.; Guzzo, C.; Hsu, M.-C.; Wang, Y.; Li, S.; et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008, 371, 1675–1684. [Google Scholar] [CrossRef]
  200. Leonardi, C.L.; Kimball, A.B.; Papp, K.A.; Yeilding, N.; Guzzo, C.; Wang, Y.; Li, S.; Dooley, L.T.; Gordon, K.B.; PHOENIX 1 Study Investigators. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008, 371, 1665–1674. [Google Scholar] [CrossRef]
  201. Gordon, K.B.; Papp, K.A.; Langley, R.G.; Ho, V.; Kimball, A.B.; Guzzo, C.; Yeilding, N.; Szapary, P.O.; Fakharzadeh, S.; Li, S.; et al. Long-term safety experience of ustekinumab in patients with moderate to severe psoriasis (Part II of II): Results from analyses of infections and malignancy from pooled phase II and III clinical trials. J. Am. Acad. Dermatol. 2012, 66, 742–751. [Google Scholar] [CrossRef] [PubMed]
  202. Lebwohl, M.; Strober, B.; Menter, A.; Gordon, K.; Weglowska, J.; Puig, L.; Papp, K.; Spelman, L.; Toth, D.; Kerdel, F.; et al. Phase 3 Studies Comparing Brodalumab with Ustekinumab in Psoriasis. N. Engl. J. Med. 2015, 373, 1318–1328. [Google Scholar] [CrossRef] [PubMed]
  203. Papp, K.A.; Reich, K.; Paul, C.; Blauvelt, A.; Baran, W.; Bolduc, C.; Toth, D.; Langley, R.G.; Cather, J.; Gottlieb, A.B.; et al. A prospective phase III, randomized, double-blind, placebo-controlled study of brodalumab in patients with moderate-to-severe plaque psoriasis. Br. J. Dermatol. 2016, 175, 273–286. [Google Scholar] [CrossRef] [PubMed]
  204. Griffiths, C.E.M.; Reich, K.; Lebwohl, M.; van de Kerkhof, P.; Paul, C.; Menter, A.; Cameron, G.S.; Erickson, J.; Zhang, L.; Secrest, R.J.; et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): Results from two phase 3 randomised trials. Lancet 2015, 386, 541–551. [Google Scholar] [CrossRef]
  205. Gordon, K.B.; Blauvelt, A.; Papp, K.A.; Langley, R.G.; Luger, T.; Ohtsuki, M.; Reich, K.; Amato, D.; Ball, S.G.; Braun, D.K.; et al. Phase 3 Trials of Ixekizumab in Moderate-to-Severe Plaque Psoriasis. N. Engl. J. Med. 2016, 375, 345–356. [Google Scholar] [CrossRef] [PubMed]
  206. Langley, R.G.; Elewski, B.E.; Lebwohl, M.; Reich, K.; Griffiths, C.E.M.; Papp, K.; Puig, L.; Nakagawa, H.; Spelman, L.; Sigurgeirsson, B.; et al. Secukinumab in plaque psoriasis--results of two phase 3 trials. N. Engl. J. Med. 2014, 371, 326–338. [Google Scholar] [CrossRef]
  207. Blauvelt, A.; Reich, K.; Tsai, T.-F.; Tyring, S.; Vanaclocha, F.; Kingo, K.; Ziv, M.; Pinter, A.; Vender, R.; Hugot, S.; et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-to-severe plaque psoriasis up to 1 year: Results from the CLEAR study. J. Am. Acad. Dermatol. 2017, 76, 60–69.e69. [Google Scholar] [CrossRef]
  208. Hueber, W.; Sands, B.E.; Lewitzky, S.; Vandemeulebroecke, M.; Reinisch, W.; Higgins, P.D.R.; Wehkamp, J.; Feagan, B.G.; Yao, M.D.; Karczewski, M.; et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: Unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012, 61, 1693–1700. [Google Scholar] [CrossRef]
  209. Kulig, P.; Musiol, S.; Freiberger, S.N.; Schreiner, B.; Gyülveszi, G.; Russo, G.; Pantelyushin, S.; Kishihara, K.; Alessandrini, F.; Kündig, T.; et al. IL-12 protects from psoriasiform skin inflammation. Nat. Commun. 2016, 7, 13466. [Google Scholar] [CrossRef]
  210. Lee, E.; Trepicchio, W.L.; Oestreicher, J.L.; Pittman, D.; Wang, F.; Chamian, F.; Dhodapkar, M.; Krueger, J.G. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J. Exp. Med. 2004, 199, 125–130. [Google Scholar] [CrossRef]
  211. Gordon, K.B.; Strober, B.; Lebwohl, M.; Augustin, M.; Blauvelt, A.; Poulin, Y.; Papp, K.A.; Sofen, H.; Puig, L.; Foley, P.; et al. Efficacy and safety of risankizumab in moderate-to-severe plaque psoriasis (UltIMMa-1 and UltIMMa-2): Results from two double-blind, randomised, placebo-controlled and ustekinumab-controlled phase 3 trials. Lancet 2018, 392, 650–661. [Google Scholar] [CrossRef]
  212. Reich, K.; Gooderham, M.; Thaçi, D.; Crowley, J.J.; Ryan, C.; Krueger, J.G.; Tsai, T.-F.; Flack, M.; Gu, Y.; Williams, D.A.; et al. Risankizumab compared with adalimumab in patients with moderate-to-severe plaque psoriasis (IMMvent): A randomised, double-blind, active-comparator-controlled phase 3 trial. Lancet 2019. [Google Scholar] [CrossRef]
  213. Reich, K.; Papp, K.A.; Blauvelt, A.; Tyring, S.K.; Sinclair, R.; Thaçi, D.; Nograles, K.; Mehta, A.; Cichanowitz, N.; Li, Q.; et al. Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSURFACE 1 and reSURFACE 2): Results from two randomised controlled, phase 3 trials. Lancet 2017, 390, 276–288. [Google Scholar] [CrossRef]
  214. Langley, R.G.; Tsai, T.-F.; Flavin, S.; Song, M.; Randazzo, B.; Wasfi, Y.; Jiang, J.; Li, S.; Puig, L. Efficacy and safety of guselkumab in patients with psoriasis who have an inadequate response to ustekinumab: Results of the randomized, double-blind, phase III NAVIGATE trial. Br. J. Dermatol. 2018, 178, 114–123. [Google Scholar] [CrossRef]
  215. Reich, K.; Armstrong, A.W.; Foley, P.; Song, M.; Wasfi, Y.; Randazzo, B.; Li, S.; Shen, Y.-K.; Gordon, K.B. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: Results from the phase III, double-blind, placebo- and active comparator-controlled VOYAGE 2 trial. J. Am. Acad. Dermatol. 2017, 76, 418–431. [Google Scholar] [CrossRef]
  216. Blauvelt, A.; Papp, K.A.; Griffiths, C.E.M.; Randazzo, B.; Wasfi, Y.; Shen, Y.-K.; Li, S.; Kimball, A.B. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial. J. Am. Acad. Dermatol. 2017, 76, 405–417. [Google Scholar] [CrossRef]
  217. Mease, P.; Hall, S.; Fitzgerald, O.; van der Heijde, D.; Merola, J.F.; Avila-Zapata, F.; Cieślak, D.; Graham, D.; Wang, C.; Menon, S.; et al. Tofacitinib or Adalimumab versus Placebo for Psoriatic Arthritis. N. Engl. J. Med. 2017, 377, 1537–1550. [Google Scholar] [CrossRef]
  218. Gladman, D.; Rigby, W.; Azevedo, V.F.; Behrens, F.; Blanco, R.; Kaszuba, A.; Kudlacz, E.; Wang, C.; Menon, S.; Hendrikx, T.; et al. Tofacitinib for Psoriatic Arthritis in Patients with an Inadequate Response to TNF Inhibitors. N. Engl. J. Med. 2017, 377, 1525–1536. [Google Scholar] [CrossRef]
  219. Papp, K.A.; Krueger, J.G.; Feldman, S.R.; Langley, R.G.; Thaçi, D.; Torii, H.; Tyring, S.; Wolk, R.; Gardner, A.; Mebus, C.; et al. Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: Long-term efficacy and safety results from 2 randomized phase-III studies and 1 open-label long-term extension study. J. Am. Acad. Dermatol. 2016, 74, 841–850. [Google Scholar] [CrossRef] [Green Version]
  220. Papp, K.A.; Menter, M.A.; Abe, M.; Elewski, B.; Feldman, S.R.; Gottlieb, A.B.; Langley, R.; Luger, T.; Thaci, D.; Buonanno, M.; et al. Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: Results from two randomized, placebo-controlled, phase III trials. Br. J. Dermatol. 2015, 173, 949–961. [Google Scholar] [CrossRef]
  221. Papp, K.A.; Bissonnette, R.; Gooderham, M.; Feldman, S.R.; Iversen, L.; Soung, J.; Draelos, Z.; Mamolo, C.; Purohit, V.; Wang, C.; et al. Treatment of plaque psoriasis with an ointment formulation of the Janus kinase inhibitor, tofacitinib: A Phase 2b randomized clinical trial. BMC Dermatol. 2016, 16, 15. [Google Scholar] [CrossRef] [PubMed]
  222. Ports, W.C.; Khan, S.; Lan, S.; Lamba, M.; Bolduc, C.; Bissonnette, R.; Papp, K. A randomized phase 2a efficacy and safety trial of the topical Janus kinase inhibitor tofacitinib in the treatment of chronic plaque psoriasis. Br. J. Dermatol. 2013, 169, 137–145. [Google Scholar] [CrossRef] [PubMed]
  223. Papp, K.A.; Menter, M.A.; Raman, M.; Disch, D.; Schlichting, D.E.; Gaich, C.; Macias, W.; Zhang, X.; Janes, J.M. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis. Br. J. Dermatol. 2016, 174, 1266–1276. [Google Scholar] [CrossRef] [PubMed]
  224. Papp, K.; Pariser, D.; Catlin, M.; Wierz, G.; Ball, G.; Akinlade, B.; Zeiher, B.; Krueger, J.G. A phase 2a randomized, double-blind, placebo-controlled, sequential dose-escalation study to evaluate the efficacy and safety of ASP015K, a novel Janus kinase inhibitor, in patients with moderate-to-severe psoriasis. Br. J. Dermatol. 2015, 173, 767–776. [Google Scholar] [CrossRef] [PubMed]
  225. Eichenfield, L.F.; Call, R.S.; Forsha, D.W.; Fowler, J.; Hebert, A.A.; Spellman, M.; Stein Gold, L.F.; Van Syoc, M.; Zane, L.T.; Tschen, E. Long-term safety of crisaborole ointment 2% in children and adults with mild to moderate atopic dermatitis. J. Am. Acad. Dermatol. 2017, 77, 641–649.e645. [Google Scholar] [CrossRef] [PubMed]
  226. El-Khalawany, M.A.; Hassan, H.; Shaaban, D.; Ghonaim, N.; Eassa, B. Methotrexate versus cyclosporine in the treatment of severe atopic dermatitis in children: A multicenter experience from Egypt. Eur. J. Pediatrics 2013, 172, 351–356. [Google Scholar] [CrossRef]
  227. Dvorakova, V.; O’Regan, G.M.; Irvine, A.D. Methotrexate for Severe Childhood Atopic Dermatitis: Clinical Experience in a Tertiary Center. Pediatric Dermatol. 2017, 34, 528–534. [Google Scholar] [CrossRef]
  228. Purvis, D.; Lee, M.; Agnew, K.; Birchall, N.; Dalziel, S.R. Long-term effect of methotrexate for childhood atopic dermatitis. J. Paediatr. Child Health 2019, 155, 145. [Google Scholar] [CrossRef]
  229. Van Geel, M.J.; Mul, K.; de Jager, M.E.A.; van de Kerkhof, P.C.M.; de Jong, E.M.G.J.; Seyger, M.M.B. Systemic treatments in paediatric psoriasis: A systematic evidence-based update. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 425–437. [Google Scholar] [CrossRef]
  230. Collin, B.; Vani, A.; Ogboli, M.; Moss, C. Methotrexate treatment in 13 children with severe plaque psoriasis. Clin. Exp. Dermatol. 2009, 34, 295–298. [Google Scholar] [CrossRef]
  231. Kaur, I.; Dogra, S.; De, D.; Kanwar, A.J. Systemic methotrexate treatment in childhood psoriasis: Further experience in 24 children from India. Pediatric Dermatol. 2008, 25, 184–188. [Google Scholar] [CrossRef] [PubMed]
  232. Schmitt, J.; Schmitt, N.; Meurer, M. Cyclosporin in the treatment of patients with atopic eczema—A systematic review and meta-analysis. J. Eur. Acad. Dermatol. Venereol. 2007, 21, 606–619. [Google Scholar] [CrossRef] [PubMed]
  233. Totri, C.R.; Eichenfield, L.F.; Logan, K.; Proudfoot, L.; Schmitt, J.; Lara-Corrales, I.; Sugarman, J.; Tom, W.; Siegfried, E.; Cordoro, K.; et al. Prescribing practices for systemic agents in the treatment of severe pediatric atopic dermatitis in the US and Canada: The PeDRA TREAT survey. J. Am. Acad. Dermatol. 2017, 76, 281–285. [Google Scholar] [CrossRef] [PubMed]
  234. Jury, C.S.; McHenry, P.; Burden, A.D.; Lever, R.; Bilsland, D. Narrowband ultraviolet B (UVB) phototherapy in children. Clin. Exp. Dermatol. 2006, 31, 196–199. [Google Scholar] [CrossRef] [PubMed]
  235. Pavlovsky, M.; Baum, S.; Shpiro, D.; Pavlovsky, L.; Pavlotsky, F. Narrow band UVB: Is it effective and safe for paediatric psoriasis and atopic dermatitis? J. Eur. Acad. Dermatol. Venereol. 2011, 25, 727–729. [Google Scholar] [CrossRef]
  236. Zamberk, P.; Velázquez, D.; Campos, M.; Hernanz, J.M.; Lázaro, P. Paediatric psoriasis--narrowband UVB treatment. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 415–419. [Google Scholar] [CrossRef]
  237. Deleuran, M.; Thaçi, D.; Beck, L.A.; de Bruin-Weller, M.; Blauvelt, A.; Forman, S.; Bissonnette, R.; Reich, K.; Soong, W.; Hussain, I.; et al. Dupilumab shows long-term safety and efficacy in moderate-to-severe atopic dermatitis patients enrolled in a phase 3 open-label extension study. J. Am. Acad. Dermatol. 2019, 81. [Google Scholar] [CrossRef]
  238. Paller, A.S.; Siegfried, E.C.; Langley, R.G.; Gottlieb, A.B.; Pariser, D.; Landells, I.; Hebert, A.A.; Eichenfield, L.F.; Patel, V.; Creamer, K.; et al. Etanercept treatment for children and adolescents with plaque psoriasis. N. Engl. J. Med. 2008, 358, 241–251. [Google Scholar] [CrossRef]
  239. Papp, K.; Thaçi, D.; Marcoux, D.; Weibel, L.; Philipp, S.; Ghislain, P.-D.; Landells, I.; Hoeger, P.; Kotkin, C.; Unnebrink, K.; et al. Efficacy and safety of adalimumab every other week versus methotrexate once weekly in children and adolescents with severe chronic plaque psoriasis: A randomised, double-blind, phase 3 trial. Lancet 2017, 390, 40–49. [Google Scholar] [CrossRef]
  240. Landells, I.; Marano, C.; Hsu, M.-C.; Li, S.; Zhu, Y.; Eichenfield, L.F.; Hoeger, P.H.; Menter, A.; Paller, A.S.; Taïeb, A.; et al. Ustekinumab in adolescent patients age 12 to 17 years with moderate-to-severe plaque psoriasis: Results of the randomized phase 3 CADMUS study. J. Am. Acad. Dermatol. 2015, 73, 594–603. [Google Scholar] [CrossRef] [Green Version]

Share and Cite

MDPI and ACS Style

Chovatiya, R.; Silverberg, J.I. Pathophysiology of Atopic Dermatitis and Psoriasis: Implications for Management in Children. Children 2019, 6, 108. https://0-doi-org.brum.beds.ac.uk/10.3390/children6100108

AMA Style

Chovatiya R, Silverberg JI. Pathophysiology of Atopic Dermatitis and Psoriasis: Implications for Management in Children. Children. 2019; 6(10):108. https://0-doi-org.brum.beds.ac.uk/10.3390/children6100108

Chicago/Turabian Style

Chovatiya, Raj, and Jonathan I. Silverberg. 2019. "Pathophysiology of Atopic Dermatitis and Psoriasis: Implications for Management in Children" Children 6, no. 10: 108. https://0-doi-org.brum.beds.ac.uk/10.3390/children6100108

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop