Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disorder of childhood. Underlying factors that contribute to AD are impaired epithelial barrier, alterations in the lipid composition of the skin, immunological imbalance including increased Th2/Th1 ratio, proinflammatory cytokines, decreased T regulatory cells, genetic mutations, and epigenetic alterations. Atopic dermatitis is a multifactorial disease with a particularly complicated pathophysiology. Discoveries to date may be considered the tip of the iceberg, and the increasing number of studies in this field indicate that there are many points to be elucidated in AD pathophysiology. In this review, we aimed to illustrate the current understanding of the underlying pathogenic mechanisms in AD, to evaluate available treatment options with a focus on recently discovered therapeutic agents, and to determine the personal, familial, and economic burdens of the disease, which are frequently neglected issues in AD. Currently available therapies only provide transient solutions and cannot fully cure the disease. However, advances in the understanding of the pathogenic mechanisms of the disease have led to the production of new treatment options, while ongoing drug trials also have had promising results.
Keywords: Atopic dermatitis, childhood, pathogenesis, treatment
1. Introduction
Atopic dermatitis (AD), also known as atopic eczema, is the most common chronic inflammatory skin disorder of childhood and is characterized by pruritus, dryness of skin, and scratching [1]. The disease presents with eczematous, itchy lesions that show distinct distribution in different pediatric age groups, and episodes of clinical exacerbation called flares or flare-ups [1]. AD affects 5%–20% of children [1], and the prevalence differs among geographical regions [2]. The disease usually occurs in early childhood; about 85% of cases are observed during the first 5 years of life [3], and the disease alleviates substantially by the age of 7 [4]. In a small percentage of patients, the disease may begin in adulthood [5]. In addition, AD may be the first manifestation of “atopic march”, which is characterized by development of asthma and allergic rhinitis at a later age [6].
AD is a multifactorial heterogeneous disorder that results from the interaction of genetic and epigenetic factors, environmental agents, immunological defects, and epithelial barrier dysfunction [7]. As understanding underlying disease mechanisms is critical for the development of effective treatment strategies, studies focusing on the pathogenesis of AD have increased in recent years [8]. This review focuses on the pathogenesis of the disease and recently discovered novel therapies, in addition to classical treatment methods.
2. Epidemiology
Although AD lesions were well described in antique texts [9], the disease was first described and named by Wise and Sulzberger in 1933 [10]. There are many studies on the frequency and pathogenesis of this common disease; however, the International Study of Asthma and Allergies in Childhood (ISAAC) can be considered the most comprehensive and largest study on AD. According to ISAAC, prevalence of AD varies considerably in different regions [11], and the frequency of childhood AD was reported to range from 0.2% to 24.6% worldwide [1]. Phase 3 of the ISAAC study [12] showed that AD occurs at a higher, relatively stable ratio in developed countries and urban areas, whereas the prevalence is steadily rising in developing countries. Likewise, Civelek et al. reported that the frequency of AD is lower in developing rather than developed Mediterranean countries. In the same study, risk factors and progression of AD also showed differences in developing Mediterranean countries [13]. Compared to ISAAC phase 1, the ISAAC phase 3 study showed stable or decreased AD frequency in developed countries [2], while low-income countries were surprisingly shown to have an increase in frequency. This increase has been attributed to changes in lifestyle [14], as the short time period between phases 1 and 3 would have been insufficient for the emergence of substantial genetic changes [15]. Therefore, it is believed that a modern lifestyle may have caused changes in daily practices, eating habits, and the perception of hygiene among people living in developing countries. The use of household cleaning products containing strong chemicals and increased frequency of soap, shampoo, and shower gel usage are among the factors believed to have contributed to the increased prevalence of AD in these countries [2].
3. Diagnosis and scoring systems for clinical assessment and severity
AD is diagnosed by clinical examination mainly based on morphological features and distribution of lesions; however, no laboratory or pathological findings specific to the disease have been discovered to date. Therefore, strict application of standard diagnosis criteria is important and even necessary to prevent misdiagnoses of atopic dermatitis in cases of other kinds of dermatitis. Until 1960, there were no criteria for the diagnosis of AD. In 1961 Georg Rajka established the first criteria, followed by Jon Hanifin, who published 13 features for the diagnosis of AD in 1977 [16]. After these initial efforts, Hanifin and Rajka proposed modified diagnostic criteria for AD in 1980 [17]. This modified set of criteria has been shown to have high sensitivity, up to 93% to 96% according to several studies [18,19], and this set of criteria has been used as a basis for later versions of AD diagnostic criteria [20]. According to the original Hanifin and Rajka criteria, a patient was diagnosed with AD when at least 3 of 4 major and at least 3 of 23 minor features were met [16]. However, the minor criteria were not practical to use in clinical practice [21]. The Hanifin and Rajka criteria were used by Kang and Tian (1987, China), Schultz Larsen (1992, Sweden), Japanese Dermatological Association (JDA) (in 1994), Danish Allergy Research Center (DARC) (in 2005), Korean Dermatological Association (KDA) (in 2006), and American Academia of Dermatology in 2003 [22]. Unfortunately, the Hanifin and Rajka criteria were deemed unsatisfactory for epidemiological surveys [20]. Therefore, the United Kingdom Working Party diagnostic criteria (UK criteria, in 1994) [23] and the ISAAC criteria (in 1995) [24] were used for epidemiological studies. Although the UK criteria were appropriate for clinical practice, they were not applicable for small children, especially infants. The ISAAC criteria have been utilized as the gold standard diagnostic criteria in global surveys for all childhood age groups since the 1990s [20,25].
Assessment of disease severity is particularly important in clinical practice when determining appropriate treatment for the patient, identifying the time of treatment cessation, and for epidemiological surveys. For this purpose, a comprehensive scoring system was published in 1993 based on a consensus by the European Task Force Group on Atopic Dermatitis (ETFAD), with participation of over 20 experts [26]. This scoring system was named SCORAD, an acronym for “SCORing Atopic Dermatitis” [26]. This scoring system comprises both objective (A. Extent: area involved according to the rule of nines; B. Intensity: erythema, edema/papules, scratching, crust formation, lichenification, and dryness) and subjective symptoms (C. Pruritus and loss of sleep) [26], and total score is calculated with the formula A / 5 + 7B / 2 + C [26]. The final score is extremely variable and is highly dependent on the physician’s examination. Consequently, an objective SCORAD which omits the subjective C criteria was put forth in 1997 [27]. Other than these scoring systems, a simplified score named Three-Item Severity (TIS) which evaluates erythema, edema, and excoriation [28], SASSAD (Six Area, Six Sign Atopic Dermatitis) that encompasses 6 signs (erythema, exudation, excoriation, dryness, cracking, and lichenification), each on a scale of 0 (absent), 1 (mild), 2 (moderate), or 3 (severe), at each of 6 sites (arms, hands, legs, feet, head and neck, and trunk) [29]; finally, the EASI (Eczema Area and Severity Index) has been developed [30]. Nevertheless, TIS is less sensitive than SCORAD [31]; SASSAD does not have the parameters of pruritus and loss of sleep and has disadvantages in calculation of an objective score; EASI scoring is a time-consuming and rather complex system [32]. Nowadays, objective SCORAD has been widely used and is considered the most accurate scoring system for AD [32].
AD may be confused with many other skin diseases, and differential diagnosis should be made with these disorders. Several diseases, including diseases causing immunodeficiency, may present with eczematous skin rash and could mimic AD. Seborrheic dermatitis, especially in infancy, dermatitis herpetiformis, irritant contact dermatitis, nummular dermatitis, and psoriasis are common inflammatory skin disorders that are often mistaken for AD [33,34]. Other skin conditions which may be ascribed to AD include allergic contact dermatitis, scabies, molluscum contagiosum, tinea corporis and capitis, mycosis fungoides, Langerhans cell histiocytosis, and pityriasis lichenoides chronica [35]. Additionally, a long list of immunodeficiency disorders, such as hyperimmunoglobulin E syndrome, Wiskott–Aldrich syndrome, Netherton syndrome, immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX), severe combined immunodeficiency (SCID), and Omenn syndrome may be diagnosed in children who present with symptoms suggestive of AD [33,36].
4. Novelties in pathogenesis of atopic dermatitis
AD is a multifactorial and heterogeneous skin disorder resulting from genetic predisposition, environmental factors, skin barrier defects, and immunological abnormalities [8] (Figure 1). Elucidating the underlying pathogenesis and understanding the mechanism that causes itching are essential for determining the most effective treatment.
4.1. Epithelial barrier dysfunction
There are many molecules in the structure of the epidermis that are critical in preventing water loss and providing protection against environmental factors [37]. Mutations in the genes encoding for proteins responsible for normal barrier function are among the main reasons for AD development [37]. Defined mutations are located in filaggrin (FLG), desmoglein-1 (DSG1), corneodesmosin (CDSN), serine protease inhibitor Kazal-type 5 (SPINK5), Matt [38], and lymphoepithelial Kazal-type–related inhibitor (LEKTI) [37]. The SPINK5 gene encodes LEKTI which is involved in the pathogenesis of AD-like disorders, such as Netherton syndrome, a syndrome characterized by severe dermatitis, allergic diseases, and high serum IgE levels [39]. Skin barrier integrity is maintained by fillagrin via keratinocyte formation. Degraded end products of fillagrin maintain water balance, low acid pH, and barrier function of the skin [40]. Decreased or defective fillagrin molecules lead to high pH and enhance the function of serine protease kallikrein (KLK) [41]. KLK binds to its receptor and thus induces the production of thymic stromal lymphopoietin (TSLP), which promotes inflammation [42]. Inflammation causes further degradation of fillagrin end products and decreases fillagrin production [43]. As a result of decreased fillagrin and its end products, allergen penetration is enhanced [44], as well as bacterial colonization [45]. Changes in the lipid composition of the stratum corneum [46] and looseness of tight junctions are other factors that substantially contribute to AD pathology [47]. Tight junction defects and the claudin-1, claudin-23, and ZO-1 molecules have been associated with skin problems in AD [47,48]. Additionally, polymorphisms in the CLDN-1 gene which encodes claudin-1 have also been linked to AD pathology [47].
Sensitization with food allergens can occur not only via the gastrointestinal tract but also through defective and inflamed skin even before the ingestion of the allergen foods [49]. By this route, 35% of the children with AD sensitize to foods by the age of 2 years [50]. The association between inhalant allergens and AD has been demonstrated in about 65% of patients at the age of 6 years old [51].
4.2. Skin microbiota
Many bacteria and fungi colonize on the skin; however, in AD, the bacterial composition of skin completely changes due to an increased adhesion of various bacteria to the skin of those with AD [52]. Staphylococcus aureus occurs at up to 90% of lesion sites [53], while it is found at lower percentages in healthy skin. IL-4, an important cytokine of the Th2 pathway, is crucial for adhesion of Staphylococcus aureus onto skin [54]. Staphylococcal α-toxin induced keratinocyte death was shown to be increased by IL-4 and IL-13 through activation of signaling transducer and activator of transcription 6 (STAT6) [55]. It has also been shown that toxins of the bacteria increase inflammation in the skin, thereby exacerbating symptoms [56]. Fillagrin has a neutralizing effect on Staphylococcus aureus α-toxin in healthy skin [57], and fragmented filaggrin products inhibit the growth of S. aureus [58]. Additionally, enterotoxin B of the bacteria induces the production of IL-31, one of the major cytokines responsible in pruritus, amplifying the severity of symptoms [59].
Lastly, the increase in fibronectin deposition in the stratum corneum of patients with AD serves as another mechanism that enables adhesion of Staphylococcus aureus in the epidermal layer [60]. Although sphingosine prevents the colonization of the bacteria in normal healthy skin, its levels are diminished in lesion sites [61]. Furthermore, the high pH, water loss, and dryness that are characteristic of the disease are known to ease the adhesion of Staphylococcus aureus to skin [61].
4.3. Immunological mechanisms underlying AD
Immunological background is rather complicated in AD; however, cytokine profile has been shown to have an important effect on the life cycle of keratinocytes [62]. Keratinocytes express pattern recognition receptors including Toll-like receptors (TLRs), NOD-like receptors (NLRs), C-type lectin receptors (CTLRs), and RIG-I-like receptors (RLRs) [63]. These receptors recognize pathogenic antigens, subsequently causing an increase of proinflammatory cytokines [63]. Cells receive signals from their environment and transduce these inputs through intracellular signal molecules. When a relevant signal arrives to the cell surface, the cell responds by producing intracellular cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), which increase the phosphorylation of protein kinase A (PKA). This pathway leads to a decline in the levels of proinflammatory molecules such as IL-2, IL-4, IL-6, IL-10, and TNF-α, thereby suppressing innate immunity [64]. Phosphodiesterase type 4 (PDE4) inhibitors prevent cAMP degradation [65] and can therefore be used to reduce the levels of inflammatory cytokines [66]. On the other hand, keratinocytes contribute to innate immunity by producing antimicrobial peptides (AMPs) (defensin, dermicidin, cathelicidin, calprotectin) which damage microbial cellular membranes [67]. Abnormalities in the production of these molecules may contribute to worsening of AD.
In patients with AD, cytokine profile shifts from T helper 1 (Th1) to T helper 2 cells (Th2). In the acute phase of AD, there is an excess of Th2 cytokines including IL-4, IL-5, IL-9, IL-13, IL-31 [68], as well as IL-22, which is also produced by Th22 lymphocytes [69], a type of lymphocyte which is crucial in epidermal immunity. These cytokines upregulate inflammatory status and cause overproduction of immunoglobulin E; IL-22 is known to inhibit keratinocyte differentiation [70]. IL-22 is also responsible for skin barrier integrity, and its level has been shown to be correlated with AD severity [71]. TSLP, an IL-7–like cytokine, is expressed by epithelial cells and binds to antigen-presenting cells (APCs) including dendritic cells, macrophages, monocytes, and T and B lymphocytes in the epidermal layer of the skin [72]. Moreover, TSLP increases IL-4 levels, which then further increases TSLP production [73]. Keratinocytes in lesion sites express TSLP, IL-25, and IL-33, which increase Th2 cytokines by activating dendritic cells (DCs) in the skin [74]. These stimulated DCs then migrate to regional lymph nodes and are instrumental in the conversion of naïve T cells to Th2 lymphocytes [75]. Furthermore, TSLP, IL-25, and IL-33 promote the production of innate lymphocyte group 2 cells (ILC2), increase their response, and induce the production of cytokines, including IL-5 and IL-13 [74]. On the other hand, ILC2s elicit IgE isotype class switching in cells [76]. IL-4 and IL-13 are also responsible in the chronic phase through their contribution to tissue remodeling [77]. IL-5 is primarily produced by Th2 and mast cells and participates in eosinophil growth, chemotaxis, and survival [78]. Recent studies have shown that IL-31 has a substantial effect on the development of pruritus [79]. IL-31 is mainly produced by Th2 lymphocytes and to a lesser extent by dendritic cells upon activation [80]. Intriguingly, eosinophils in patients with AD express IL-31 more intensely compared to healthy individuals [81]. IL-31 binds to its receptor IL-31 receptor RA (IL31RA) on sensory nerves and induces an itching sensation, as well as facilitating the elongation and branching of sensory nerves [79].
Th17 lymphocyte is another cell type that contributes to the acute phase [62]. This cell produces IL-17 and IL-22, which increase proinflammatory molecules, S100 proteins, and other types of AMPs in keratinocytes [82]. S100 proteins act as AMP and damage-associated molecular pattern molecules, which initiate the inflammatory cascade [83]. The level of S100 protein increases in acute and chronic AD; thus, it is thought to be an important parameter in preserving enhanced inflammatory status [83]. Furthermore, increases in IL-17 lead to eosinophil- and neutrophil-mediated inflammation [84], while reduced IL-17 levels are correlated with reduced AMP, which has been reported to be associated with increased susceptibility to skin infections [85]. In the chronic phase, interferon gamma (IFNɣ), a Th1 cytokine, is believed to be more prevalent, along with Th2 and Th22 cell responses [86].
5. New insights in the mechanism of pruritus
Cytokines and other chemical molecules that cause itching (pruritogens) are released from eczematous skin areas where they activate the relevant sensory nerves and cause pruritus; thus, the patient feels the need to scratch these areas [87]. Hyperesthesia in AD is due to aberrantly elongated sensory nerves in the upper layer of the skin which are increasingly exposed to environmental factors such as dryness, irritation, and chemicals [88]. Histamine, substance P, TSLP, and IL-31 act on these elongated nerve fibers, contributing to the itching sensation [89,90]. Heat is an important aggravating factor for itching in AD [91]. Artemin (enovin, neublastin) is a substance which has been shown to increase in AD lesions and is reported to cause itching, especially in warm temperatures [92].
Pruritus has detrimental effects on sleep and may reduce the length and quality of sleep, which leads to a decrease in melatonin levels. Decreased melatonin levels have been suggested to promote further itching [93]. Additionally, decreased cortisol level and increased inflammatory status due to elevated IL-2 levels are other factors that contribute to itching at night [93]. Sweating is another factor that exacerbates itching in AD, due to the normal effects of excess sweat on the skin and also the abnormal composition of sweat in patients with AD [94]. Conversely, keratinocyte debris may obstruct sweat glands and prevent normal sweating; this also leads to raised body temperature and could increase pruritus [95].
6. Genetic and epigenetic factors
Fillagrin mutation is the most important predisposing genetic change in AD. However, it is found in only 10%–50% in patients with AD, and in 9% of the healthy population [69]. Therefore, AD cannot be explained only by Fillagrin mutation. Genome‑wide association studies (GWAS) have been initiated to elucidate the pathogenesis of AD. Many novel genes have been identified in patients with AD, especially after the first GWAS published by Esparza-Gordillo et al. in 2009 [96]. Genes responsible for the pathogenesis of AD are given in Table 1. TRAF6, RAG1, RAG2, SOCS1, NGFR, 4q27 IL2/IL21, 11p13 PRR5L, 16p13.13 CLEC16A/DEXI, and 17q21.32 ZNF652 are the novel genes that have been reported in GWAS studies on AD [97]. Additionally, several genetic polymorphisms have been found to be related to AD [98]. These include polymorphism of vitamin D pathway molecules [99] and polymorphisms of cytokines IL-4, IL-5, IL-9, IL-13, and IL-31, as well as their receptors [100]. Aside from genetic mutations, epigenetic mechanisms have been implicated in AD pathogenesis [101]. Epigenetic mechanisms regulate expression of genes without altering the DNA sequence [102]. These mechanisms are mainly DNA methylation, microRNA (miRNA), and histone tail modification [102]. It has been shown that epigenetics affect gene expression through environmental factors such as pollutants, tobacco smoke, aging, and diet [103]. DNA methylation differences between normal epidermis and AD epidermis have been shown in a study focusing on epigenetic factors [104,105]. miRNAs have fundamental roles in cell proliferation, differentiation, apoptosis, signal transduction, and organ development [106]. miRNAs are upregulated in the skin of patients with AD [107]. Children who were exposed to smoking during pregnancy have high miRNA-223 and low T regulatory (Treg) lymphocytes [108]. These children were found to have a higher tendency to develop AD during the first 3 years compared to children without exposure to smoking during pregnancy [108].
Table 1.
Gene symbol | Gene name | Chromosomal location | Genetic variation | Disease severity | References |
Epidermal differentiation | |||||
FLG | Fillagrin | 1q21.3 | R501X, 2282del4 | correlated with severity | [69] |
FLG2 | Fillagrin 2 | 1q21.3 | rs12568784, Q2053del224 rs16833974 | persistent disease | [222] |
SPINK5/LEKTI | Serine protease inhibitor Kazal type 5/Lymphoepithelial Kazal type related inhibitor | 5q31 | rs2303070 T, E420K | increased disease risk | [223,224] |
TMEM79 | Transmembrane protein 79 | 1q22 | rs6684514 | increased risk of AD | [38] |
claudin-1 | 3q28 | rs893051, rs9290929 | Mold infection | [225] | |
MHC (HLA) | Major histocompatibility complex (Human leucocyte antigen) | 6p21 | TAP1 (Val333IIe, Gly637Asp) TAP2 (IIe379Val, Thr565Ala, Ala665Thr, Gln687Stop) | [226,227] | |
Innate immunity | |||||
TLR2 | 4q32 | rs4696480, rs3804099, rs3804100, rs5713708 | severe disease | [228–230] | |
TLR4 | 9q32/33 | rs4986790, rs4986791, rs2770150 | increased risk of AD | [228,230,231] | |
TLR6 | 4p14 | rs5743810 | [148] | ||
TLR9 | 3p21.3 | C-1237T | [130] | ||
NOD1(CARD4) | Caspase recruitment domain-containing protein 4 | 7p14/15 | rs2907748, rs2907749, rs2736726, rs3030207, rs2075818 | allergen sensitization | [232,233] |
CARD 11 | Caspase recruitment domain-containing protein 11 | 7p22.2 | p.Glu57Asp (E57D) p.Leu194Pro (L194) p.Arg975Trp (R975W) p.Met183_Lys196 | severe disease | [234] |
CARD 14 | Caspase recruitment domain-containing protein 14 | 17q25.3 | rs11652075 | [235] | |
NOD2 (CARD15) | Caspase recruitment domain-containing protein 15 | 16q21 | rs1077861 | [232,236] | |
TSLP | Thymic stromal lymphopoetin | 5q22 | rs1898671 | eczema herpeticum | [237] |
DEFB1 | Human β-defensin 1 | 8p23 | rs5743399, rs5743409 | [238] | |
Adaptive immunity | |||||
IL-4 | Interleukin 4 | 5q31-33 | 590C/T, 1098G/T, 589C>T | increased risk of AD | [100,239–241] |
IL-4Ra | Interleukin 4 receptor alpha | 16 | I50V, Q576R | increased risk of disease | [100] |
IL13 | Interleukin 13 | 5q31.1 | rs12188917 | association with asthma | [98] |
IL-31 | Interleukin 31 | 12 | 1066, _2057, and ivs2 +12 | high intensity of pruritus | [242] |
IL-17A | Interleukin 17 | 152 G/A | association with asthma and severe disease | [243] | |
IL9 | Interleukin 9 | 5q31/35 | 4091G>A | [244] | |
IL9R | Interleukin 9 receptor | Xq/Yq | 1737C>T | [244] | |
IL18 | Interleukin 18 | 11q22.2/3 | 132A>G, 133C>G, 137G>C, 113T>G, 127 C>T | [245] | |
GM-CSF | Granulocyte macrophage colony stimulating factor | 5q31.1 | 677A>T, 1916T>C | [246] | |
Chemokines and related genes | |||||
CCL5 (RANTES) | Chemokine (C-Cmotif) ligand 5 (Regulated upon activation, normally T cell expressed + secreted) | 7q11.2/q12 | 28C>G, 403G>A, 2518 A>G | allergen sensitization | [247,248] |
CCL11 (eotaxin 1) | Chemokine (C-Cmotif) ligand 11 | 17q21.1/q21.2 | 426C>T, 384A>G | [249] | |
CCL17 (TARC) | Chemokine (C-Cmotif) ligand 17 (thymus and activation-regulated chemokine) | 16q.13 | 431C>T | [250] | |
Vitamin D pathway | |||||
Cyp24a1 | 20q54 | rs2248359 | severe disease | [251] | |
VDR | 20q13 | rs7975232 | severe disease with high eosinophil and IgE levels | [99] | |
Other genes | |||||
CTLA4 | Cytotoxic T lymphocyte associated antigen-4 | 2q33 | 49A>G | [252] | |
IRF2 | Interferon regulatory factor 2 | 4q35 | 829C>T, 830C>T, 684C>T | [253] |
7. Current treatment of atopic dermatitis
7.1. Moisturizers and bathing
Treatment of AD is aimed at suppressing inflammation, eliminating identified triggers, reducing pruritus, and combating the development of xerosis, a major feature of AD [109,110] (Figure 2). For this purpose, conventional therapy is the first-line treatment approach. However, new therapeutic strategies and agents are continuously being developed with the help of advances in the understanding of AD pathophysiology. Basically, topical moisturizers are the mainstay of AD treatment, as they prevent dehydration, soften the skin, and increase water retention. Above all, they serve to minimize the need for pharmacological drugs for AD [110,111]. Moisturizers are the first line of therapy in mild AD and constitute an important and primary step in moderate and severe forms of AD [112]. Moisturizing the skin with different types of emollients, occlusive agents, and humectants decrease inflammation, pruritus, and xerosis, and also reduce the requirement for topical steroids and antibiotics [110]. There are many types of products in the form of ointments, lotions, creams, emollients, oils, and gels. Even though there are some differences with regards to their contents, the most important aspect of these moisturizers are to ensure that they are perfume- and fragrance-free and do not contain harsh chemicals [110]. In addition to moisturizers, daily bathing with warm water is an extremely important step for hydration of skin, as well as removing allergens, irritants, crusts, and bacteria [113]. Applying moisturizing agents immediately after bathing has been a fundamental approach for treatment in patients with AD [113]. Otherwise, bathing without moisturizing has a negative impact on AD.
7.2. Wet-wrap and bleach therapy
Wet-wrap therapy in combination with topical agents is an effective method used in the treatment of AD flare-ups [114]. Generally, topical agents are applied on the skin followed by a layer of tubular wet bandage and an outer layer of dry bandage [110]. This technique intensifies the effect of moisturizers by providing a smooth skin texture and preventing water loss [110]. A bleach bath was thought to be effective in the treatment of AD, because some physicians reported that eczematous lesions benefited from the chlorinated water of swimming pools [110]. Additionally, Huang et al. showed the efficacy of bleach bath combined with intranasal mupirocin treatment twice a week compared to placebo [115]. Bleach baths have been considered to reduce the colonization of Staphylococcus aureus on the skin, preventing secondary skin infections [116]. In this therapy, the child is soaked in water containing a certain amount of bleach (1/4 cup of bleach into 35–40 L of water) for 5 to 10 min [117,118]. However, bleach baths were shown to be ineffective in the treatment of AD in some studies [119]. Other than bleach or sodium hypochlorite, there are antiseptic agents including triclosan, potassium permanganate, and chlorhexidine gluconate used for both the management of infected skin and the prophylactic treatment of AD. Bathing with antiseptic agents has been shown to be useful in diminishing the bacterial load on the skin of patients with AD. Furthermore, no significant effect on reducing bacterial load on inflamed skin in AD has been identified in a few studies [120]. Therefore, combination therapy of antibiotics along with antiseptic agents has been shown to be particularly effective in the treatment of clinically infected skin in AD [115].
7.3. Topical antimicrobials
AD exacerbations and acute flares are mostly associated with bacterial and viral infections, including Staphylococcus aureus, Streptococcus pyogenes, Herpes simplex, Varicella zoster viruses, papillomavirus, and molluscum contagiosum [121,122]. S. aureus mostly colonizes the skin and the nasal mucosa, and is frequently associated with worsening of the eczematous lesions [122]. Mupirocin and fusidic acid are effective antibiotics for the prevention of S. aureus colonization when used twice a day for a week [123,124]. Furthermore, application of mupirocin twice a day in both nasal orifices for 5 days in a month for 3 to 18 months has been shown effective in eradicating S. aureus [124]. Systemic antibiotics are given in the presence of clear evidence of bacterial infection, not to prevent colonization [125,126]. The first choice of antibiotic treatment is beta-lactam antibiotics for 7–14 days [127].
About 3% of AD cases are prone to severe viral infections, especially Herpes simplex and Varicella zoster viruses even after immunization [111,122]. The clinical picture caused by Herpes simplex virus is eczema herpeticum, which requires administration of systemic acyclovir for treatment [128]. Prompt initiation of acyclovir treatment significantly reduces the mortality of eczema herpeticum [128].
8. Topical antiinflammatory therapy
Antiinflammatory therapy with topical calcineurin inhibitors (TCI), including tacrolimus and pimecrolimus, as well as topical corticosteroids (TCS), is reported to be the most effective approach in the treatment of acute flares [118]. Excessive inflammatory response in the skin is minimized by antiinflammatory treatment agents during flares [118]. Both TCS and TCI suppress T lymphocytes and inhibit proinflammatory cytokine release from immune cells [110,129]. Therefore, these agents are useful in controlling inflammation, pruritus, and skin eruptions [118].
9. Topical corticosteroids
TCSs are widely used; they are the first-line treatment for acute flares in AD [110,130]. They are indicated for eczematous lesions unresponsive to daily skin care and proper usage of emollients, creams, and ointments [116]. TCSs exhibit their effect by inhibition of T lymphocytes [110], thus reducing inflammation on the skin; they are also known to abate pruritus. In addition, topical corticosteroids are not only used for acute attacks but also for prevention of relapses [110].
TCSs are divided into 7 categories according to their potency [110]. Corticosteroids with higher potency should be applied for acute flares, whereas low potency corticosteroids are highly recommended for chronic long term usage [110,131]. The optimal suggested period of application is twice daily for 2 weeks, and only on affected surfaces of the skin for protection from local adverse effects of corticosteroids [116]. Many local side effects have been reported, including hypertrichosis, striae, telangiectasia, and skin atrophy for long term skin application, as well as glaucoma and cataract after use on the periorbital region [116]. Therefore, TCIs are recommended for thin skin surfaces such as eyelids and periorbital areas [132]. Studies have demonstrated that skin atrophy is extremely rare in children [133]. Systemic adverse effects, including the suppression of the hypothalamic–pituitary–adrenal (HPA) axis, have been reported after use of high potency topical corticosteroids in children [134]. Use of low to moderate potency topical corticosteroids for 3 to 4 weeks has been demonstrated to rarely affect the HPA axis [134]. Additionally, the possibility of growth retardation in children has been researched in many studies; however, results have mostly revealed only insignificant delays in growth [135].
10. Topical calcineurin inhibitors
Tacrolimus and pimecrolimus were approved for AD treatment in 2000 and 2001, respectively [136]; they are safe for use in adults and children over 2 years old [136]. TCIs are a product of the Streptomyces genus that inhibits calcineurin-dependent T-cell activation and proinflammatory cytokines. They exert their effects by inhibiting mast cells, dendritic cells, and T lymphocytes [137]. In studies, TCIs have been shown to be more effective than mild TCSs and as effective as moderate TCSs [111,138,139]. Therefore, TCIs are suggested for severe, TCS-unresponsive AD lesions, and also for sensitive areas such as eyelids and the face, and when concerned about steroid-related adverse effects [132,140]. However, erythema, burning, and itching sensations have been frequently reported with TCIs [141]. Additionally, many carcinogenic effects were reported during trials on animals [136]. Systemic absorption of TCI has been shown in patients with AD through the disrupted skin barrier and other skin areas [136]; therefore, side effects may be seen more frequently in pediatric age groups [136]. Pimecrolimus 1% and tacrolimus 0.03% are approved for children who are between 2 and 15 years old; 0.1% ointment of tacrolimus is not allowed for pediatric patients [110,136]. Both pimecrolimus and tacrolimus are contraindicated in acute viral skin infections, suspicion of skin malignancy, or immunodeficiency [136]. Although the exact relationship between malignancy and calcineurin inhibitors cannot be demonstrated, skin tumors and lymphoma have been reported [142]. Therefore, long-term use of these agents in all age groups should be avoided. In clinical trials, tacrolimus and pimecrolimus have been shown to be effective on moderate to severe and mild to moderate AD, respectively [143]. TCI should be used twice a day over 2 to 3 weeks, then should be reduced to once a day for 2 weeks [144]. At the beginning of the treatment, burning and itching are significant problems for patients and may reduce treatment compliance. Concomitant steroid therapy may overcome this issue [144]. Although safe for use and very effective, there is a risk for rebound after the cessation of therapy with TCIs; in the event of a rebound or flare-up, TCIs may be restarted twice a day again [144].
11. Systemic treatment
Systemic therapy for AD is considered when the skin lesions do not ameliorate after usage of topical treatments [145]
11.1. Systemic corticosteroids (SCS)
SCS therapy has many side effects, and therefore it is applied for only a week, especially in adults [146]. It has little efficacy on improvement of lesions, and its adverse effects usually outweigh its benefits [147]. Cushing syndrome, hyperglycemia, osteoporosis, and peptic ulcer are primary side effects of systemic steroid therapy [148]. Due to these adverse effects, they are not recommended for routine treatment and recommended for use only for a short time until symptoms are alleviated in severe cases [145,149,150].
11.2. Cyclosporine
Cyclosporine exhibits its effect by inhibiting calcineurin receptors which induce the proliferation of IL-2 cytokine. This cytokine is vital for T helpers, T regulatory lymphocytes, Natural Killer cells, and monocytes. By blocking the production of IL-2, the activity and proliferation of T lymphocytes are hampered [151]. Patients with severe, topical therapy-resistant lesions have been shown to benefit from oral cyclosporine treatment [152]. Recommended dosage is 2.5 mg/kg/day in 2 divided doses, not exceeding a maximum dose of 5.0 mg/kg/day [153]. Serum levels of cyclosporine should be monitored routinely and managed according to clinical symptoms and serum levels of the drug. If there is clinical advancement, duration of the therapy may be extended to 12 months [151]. In case of the clinical improvement, cyclosporine therapy is reduced and terminated in 2–3 months [151]. Most frequently observed side effects are headache, hypertension, renal dysfunction, hypertrichosis, hepatotoxicity, and gingival hyperplasia in adults [152]. However, these are extremely rare in children [154]. Cyclosporine has been well tolerated by children and is particularly effective in this age group [155].
11.3. Azathioprine
Azathioprine acts as an immunosuppressant on proliferating cells. It is a purine analogue inhibiting DNA/RNA synthesis and prevents proliferation of both T and B lymphocytes [156]. Azathioprine has been used with substantial benefits for AD treatment in adults and children [156]. Azathioprine is generally used for a short period in patients who are refractory to cyclosporine [155]. Its effect arises 2–4 weeks after initiation of the therapy, and it is thus considered to be a secondary preference for treatment of AD [149]. The most substantial adverse effects of azathioprine are hepatotoxicity, increased risk of malignancy, and myelosuppression [157]. The recommended dose is 2–4 mg/kg/day, and whole blood count should be monitored for cytopenia regularly [158].
11.4. Methotrexate
Methotrexate is a folic acid antagonist which inhibits cell division and DNA/RNA synthesis and suppresses the immune system, which are similar to the effects of azathioprine [149]. The efficacy of the drug has been shown in children and it is well tolerated in the pediatric age group [159]. It has been suggested to be effective in improving skin lesions with few side effects and is considered a safe option for children [159]. However, it may cause bone marrow suppression, increase transaminase levels, and result in cytopenia. Furthermore, although extremely rare, pulmonary fibrosis has been shown to be a complication of the drug [149]. Beginning of action may be prolonged up to 3 months, as is sometimes the case with azathioprine [149]. There is little data on the application of methotrexate in children. In reports, a low dose (5–15 mg/week orally) has been demonstrated to be effective in alleviating lesions [159,160].
11.5. Mycophenolate mofetil (MMF)
MMF is an immunosuppressive drug derived from mycophenolic acid. It regulates purine synthesis and inhibits T and B lymphocytes [161]. MMF was used in adult patients for the first time in 1999 [162], and was applied to children over 2 years old with refractory AD in 2007 [163]. Pediatric results were promising and no significant undesirable effects were reported. The most frequent adverse effects seen in children were nausea, vomiting, and diarrhea [163]. In addition to these side effects, hepatotoxicity, myelosuppression, and susceptibility to infections may be seen during treatment [163]. Daily recommended dose is 600 mg/m2/day in 2 divided doses in children [164].
12. Other therapies
Extracorporeal photopheresis and intravenous immunoglobulin therapies have been applied to patients with AD [165,166]. There are limited data available about these methods, and results of studies which utilized these methods showed that they had little efficacy on AD.
12.1. Antihistamines
Itching is the most intolerable symptom of AD and it has devastating effects on social life and sleep. First-generation antihistamines may be beneficial due to their sedation properties [167], but second-generation antihistamines have less sedative effect [167]. Therefore, only first-generation drugs may be used for severe cases of pruritus and overcoming sleep disturbance.
12.2. Allergen immunotherapy
Atopy is defined in up to 70% in patients with AD and exposure to aeroallergens have been shown to cause acute flares [130,168]. Removal of allergens from patients with AD with underlying etiology of allergy is an important step in treatment, and avoidance of aeroallergens has been suggested in the literature to decrease flare-up frequency and the severity of AD [130]. There are many studies on immunotherapy and AD; however, outcomes of the reports are conflicting, and contrary to studies on allergic rhinitis, efficacy is questionable in AD [169]. Atopic march, a severe problem in children with AD, should be prevented; however, the preventive role of allergen immunotherapy has not been demonstrated yet [170]. Nevertheless, patients with severe AD and accompanying allergic rhinitis and/or asthma may be considered for allergen immunotherapy [171].
13. Biological agents and new treatment strategies
13.1. Interferon gamma (IFNɣ)
IFNɣ inhibits T helper 2 lymphocytes and reduces IgE production [172]. In clinical trials, patients with moderate to severe AD have been shown to benefit from IFNɣ therapy [173,174]. The dose used in the pediatric age group was 50 µg/m2 for 22 months; clinical improvement was observed [173,174]. However, a specific recommended dose for IFNɣ for AD has not been determined and the Food and Drug Administration (FDA) has not approved the use of IFNɣ for AD [172].
13.2. Omalizumab
Omalizumab is a recombinant humanized mAb (rhmAb) that inhibits binding of free, unbound IgE to the high-affinity IgE receptor (FcεRI) on mast cells and basophils [175]. Its first use for severe asthma was assessed in the Global Initiative for Asthma Guidelines study in 2003 [176]. Furthermore, omalizumab was initially used on children who were between 6 and 12 years old in 2009 for severe asthma [177]. Later, Barrios et al. reported the efficacy of omalizumab in their pediatric group of patients with AD in 2013 [178]. Doses were not standardized and varied from 150 mg/dose to 375 mg/dose every 2 to 4 weeks in various studies [179,180]. The drug was well tolerated in children without any particular side effects, but clinical trials on the use of omalizumab for AD are very few, and they report debatable efficacy [181,182].
13.3. Rituximab
Rituximab is an anti-CD20 monoclonal antibody. It was shown to be beneficial on severe AD lesions in 2008 [183]. However, that was followed by a report on the clinical experience of 2 patients with severe AD who had only minimal transient clinical benefits from rituximab therapy [184]. There is no standardized recommended dose for the usage in patients with AD and further studies with higher numbers of patients are required to evaluate its efficacy in AD.
13.4. Dupilumab
Dupilumab is a human monoclonal antibody against a subunit of the IL-4/13 receptor [185]. In a 2014 clinical trial, significant improvements in patients were observed at the end of the 3-month treatment period [186]. The FDA approved dupilumab for use in adults with moderate-to-severe AD in March 2017 [172]. Clinical trials on children are ongoing [172]; preliminary results indicate that results with dupilumab could be effective in the treatment of AD. The dose of the drug was 2 mg/kg in patients aged 6–11 years and 4 mg/kg in patients aged 12–17 years, or 200–300 mg at intervals of 2–4 weeks in clinical trials [187].
13.5. Mepolizumab
IL-5 is critical for eosinophil growth and differentiation [188]. Eosinophils are essential contributors to AD pathophysiology and have been shown to be elevated in the serum of patients with AD [188]. Therefore, it was suggested that mepolizumab, a human IL-5 mAb, may have an important effect on the treatment of AD [172]. However, initial studies in 2005 did not yield positive results and no clinical improvement was seen [172,189].
14. Biological agents in trials
Biological agents are new therapeutic drugs targeting the molecules responsible in the pathogenesis of disease. Omalizumab, rituximab, interferon gamma, and mepolizumab have already been tried, while new agents that function by affecting the various stages of pathogenesis are under ongoing investigation. The primary agents among these new potential treatments are nemolizumab, ustekinumab, tralokinumab, lebrikizumab, antithymic stromal lymphopoietin receptor (TSLPR), and phosphodiesterase (PDE) inhibitors.
14.1. Nemolizumab
Nemolimumab (CIM331), a humanized anti-IL-31 receptor A mAb, binds to IL-31 receptor A, therefore inhibiting IL-31 signaling [190]. This promising agent is thought to be especially effective for pruritus, and therefore may alleviate problems such as sleep disturbance and psychological concerns in both patients and parents. In clinical trials, pruritus has been reported to be reduced 2-fold compared to placebo [190,191].
14.2. Ustekinumab
Ustekinumab is a humanized mAb that binds to the p40 subunit of IL-12/IL-23 cytokines. These molecules have a pivotal role in the development of Th1 and Th17 cells [192]. Trials have demonstrated that ustekinumab reduces the infiltration of T lymphocytes, dendritic cells, and mast cells in the skin [172]. However, no meaningful clinical efficacy has been reported [193,194].
14.3. Tralokinumab
Tralokinumab is a recombinant IgG4 neutralizing mAb that binds to IL-13 and interferes with the interaction between IL-13 and its receptor [195]. In trials, the drug has not shown any particular difference from placebo [196]. However, various studies are still ongoing [172].
14.4. Lebrikizumab
Lebrikizumab is another IL-13 binding mAb, but its efficacy and safety profile have been thought to be different from other drugs that bind IL-13. Initial results on the drug’s efficacy have shown superior results compared to placebo with repeated doses [197] and no significant adverse effect has been reported [197].
14.5. Antithymic stromal lymphopoietin receptor (TSLPR)
TSLP is an IL-7–like cytokine that is primarily expressed in epithelial cells, keratinocytes, and ocular cells, and has a role in allergic inflammatory disorders [198–200]. Secretion of the cytokine can be stimulated by allergens, viruses, trauma, and smoke [201]. Myeloid dendritic cells (mDCs) are activated by TSLP, and begin a cascade of events that lead to the differentiation of naive T cells into Th2 lymphocytes [202]. Blocking of TSLPR was first explored in asthma treatment [203]; other trials comprised of patients with AD have also begun [196]. TSLP’s role in the exacerbation of pruritus via sensory neurons has been demonstrated [90]. Studies on AD have revealed significant improvement in the severity scores of the patients [196].
14.6. Phosphodiesterase (PDE) inhibitors
In the 1980s, enhanced PDE activity was determined in blood cells as well as cord blood cells of patients with AD [204]. Since then, the effects of PDE inhibitors in patients with AD have been demonstrated [205]. Topical forms of PDE inhibitors are crisaborole, OPA-15406, RVT-501, and roflumilast, while systemic (oral) forms of the drug are apremilast and roflumilast [204]. In December 2016, FDA approved crisaborole for the treatment of mild-to-moderate atopic dermatitis in patients aged ≥2 years [204].
15. Burden of atopic dermatitis
AD mostly begins in childhood and may persist throughout adulthood [206]. Therefore, it may have both economic and psychosocial impacts on patients and their families [207]. Evaluation of quality of life (QoL), social, academic, and occupational impacts, and financial costs are important in capturing the whole picture when assessing the burden of AD. Patients with AD were shown to have lower overall QoL [208], especially in the form of mental health and social function [208]. A study by Solomon et al. showed that children with AD had severe sleep difficulties and were more dependent on their caregivers than patients with other chronic disorders [209]. Chamlin et al. also showed that these children were avoided by other people, causing social isolation [210]. Many studies have shown that sleep deprivation, isolation from social activities, and feelings of embarrassment due to itching and appearance of the eczematous lesions were the major problems faced by children with AD [211,212]. In a study performed on children and adults with AD, it was reported that 87.1% of patients had difficulties falling asleep, and 73.5% reported that severe itching was the cause for waking up [213].
AD also affects parents. The parents of children with AD also reported sleep disturbance [210] and stress [214], and were found to spend a significant amount of time on daily skin care [210] and hygiene [215]. Furthermore, the time spent by parents with the siblings of children with AD was also reported to be decreased [216], suggesting that AD has detrimental effects on the entire family. Lifestyle changes including modified dietary habits and usage of cleaning products and moisturizers for the skin in patients with AD were also shown to cause a substantial burden on the family budget [217]. Additionally, regular physician visits may cause loss of worktime for parents, while spending excessive time and effort on the care of a child with AD may also cause social isolation for the parents [217].
Assessing the exact financial burden of AD on the family budget and the health system is very difficult due to the indirect costs as well as direct expenditures. Additionally, the total costs of AD are also dependent on disease severity and may differ significantly from country to country [218]. Indirect costs include loss of labor and decrease in productivity at work [207]. Direct costs consist of pharmacy expenditures, follow-up visits, applications to emergency departments, and spending on special diet programs [207]. A striking result reported by Storan et al. indicated that 86% of the children admitted to their inpatient clinic were diagnosed with AD [219]. This study demonstrates the financial strain caused by AD on the health system. According to studies on economic costs of chronic diseases, AD was in fourth place among skin diseases [220]. In 2004, the annual cost of AD was 4.228 billion dollars in the United States [220], and had increased to approximately 5.297 billion dollars in 2015 [207]. Although indirect costs are not exactly estimated, the direct costs of AD have increased significantly over the years.
16. Future perspectives
The complexity of AD pathogenesis, the lack of a definitive treatment, and the fact that available therapies only provide transient well-being are among the factors that fuel the research for a cure for AD. Mainly due to this enthusiasm, experts have explored many novel molecules, mutations, and genes responsible for the disease. New treatment modalities are reported to be promising for the treatment of AD. However, it is also important to consider that current treatments (and the majority of proposed treatments) do not aim to prevent the development of atopic march. For this reason, researchers on this field concentrate not only on antiinflammatory drugs, but also on immunological and biological agents. With our knowledge today, it is evident that AD has a significant immunological background, and if the immunological dysregulation underlying the disease can be elucidated and targeted in treatment, success in AD treatment and prevention will be achieved. Pruritus is one of the most important problems of AD and suggested treatments must address this problem. Pruritus causes exhaustion, despair, and sleep deprivation in patients. Unfortunately, current drugs are insufficient for the control of pruritus. Therefore, development of antipruritic drugs with higher effectiveness should be regarded as high priority. The current therapeutic approach to AD, the advantages of treatments, and unmet needs are summarized in Table 2.
Table 2.
Current treatment | Unmet needs | New goals |
Topical and systemic corticosteroids, calcineurine inhibitors, immunomodulators | Disease flare-ups are seen despite long term usage of today’s available drugs | Minimizing flare-ups, achieving disease free life |
Drugs are used for months or years in general | Long term application may not be safe in children (malignancy in calcineurine inhibitors) | Patients should adapt therapy easily in their lives, attaining short term using drugs |
Only for eczematous lesions, providing temporary wellbeing, not prevent recurrence and other allergic diseases | Generally proceeds to asthma and allergic rhinitis | Prevention of other allergic disorders with radical treatment strategies |
Systemic antihistamines are used for pruritus and give sedation | Pruritus does not cease and is recalcitrant, psychological exhaustion appears frequently due to pruritus | Effective antipruritic drugs |
Parents and patients should spend many hours in a day for skin care. In addition, moisturizers and drugs are expensive | Time-consuming and economic burden on families and country economy | Spending shorter time for skin care, cheap and effective alternative treatment options |
17. Conclusion
The aims of new therapies for AD are not only to procure a definite treatment but also to enhance the adherence of patients and parents to the therapy [221]. Other factors to consider as treatment goals in AD are the prevention of secondary infections, providing support to patients with psychological problems, and increasing the awareness of the population and physicians to a disease with severe consequences on the patient, family, and country.
References
- Williams H Robertson C Stewart A A t-Khaled N Anabwani G Worldwide variations in the prevalence of symptoms of atopic eczema in the International Study of Asthma and Allergies in Childhood. Journal of Allergy and Clinical Immunology. 1999;239:125. doi: 10.1016/s0091-6749(99)70536-1. [DOI] [PubMed] [Google Scholar]
- Williams H Stewart A von Mutius E Cookson W Anderson HR Is eczema really on the increase worldwide. Journal of Allergy and Clinical Immunology. 2008;121:947. doi: 10.1016/j.jaci.2007.11.004. [DOI] [PubMed] [Google Scholar]
- Nutten S Atopic dermatitis: global epidemiology and risk factors. Annals of Nutrition and Metabolism. 2015;66:8. doi: 10.1159/000370220. [DOI] [PubMed] [Google Scholar]
- Huang E Ong PY Severe Atopic Dermatitis in Children. Current Allergy and Asthma Reports. 2018;18:35. doi: 10.1007/s11882-018-0788-4. [DOI] [PubMed] [Google Scholar]
- Novak N Bieber T Allergic and nonallergic forms of atopic diseases. Journal of Allergy and Clinical Immunology. 2003;112:252. doi: 10.1067/mai.2003.1595. [DOI] [PubMed] [Google Scholar]
- Dharmage S Lowe A Matheson M Burgess J Allen K Atopic dermatitis and the atopic march revisited. Allergy. 2014;69:17. doi: 10.1111/all.12268. [DOI] [PubMed] [Google Scholar]
- Bieber T Novak N Pathogenesis of atopic dermatitis: new developments. Current Allergy and Asthma Reports. 2009;9:291. doi: 10.1007/s11882-009-0041-2. [DOI] [PubMed] [Google Scholar]
- E D'Auria Banderali G Barberi S Gualandri L Pietra B Atopic dermatitis: recent insight on pathogenesis and novel therapeutic target. Asian Pacific Journal of Allergy and Immunology. 2016;8217:98. doi: 10.12932/AP0732.34.2.2016. [DOI] [PubMed] [Google Scholar]
- Mier P Earliest description of the atopic syndrome? British Journal of Dermatology. 1975;92:359–359. [Google Scholar]
- Year Book of Dermatology and Syphilology. Chicago, IL, USA: Year Book Publishers. 1933. p. 38.
- Odhiambo JA Williams HC Clayton TO Robertson CF Asher MI Global variations in prevalence of eczema symptoms in children from ISAAC Phase Three. Journal of Allergy and Clinical Immunology. 2009;124:1251. doi: 10.1016/j.jaci.2009.10.009. [DOI] [PubMed] [Google Scholar]
- Mallol J Crane J von Mutius E Odhiambo J Keil U The International Study of Asthma and Allergies in Childhood (ISAAC) Phase Three: a global synthesis. Allergologia et Immunopathologia. 2013;41:73. doi: 10.1016/j.aller.2012.03.001. [DOI] [PubMed] [Google Scholar]
- Civelek E Sahiner U Yüksel H Boz A Orhan F Prevalence, burden, and risk factors of atopic eczema in schoolchildren aged 10-11 years: a national multicenter study. Journal of Investigational Allergology and Clinical Immunology. 2011;252:270. [PubMed] [Google Scholar]
- Epidemiology of atopic dermatitis: a review. Allergy & Asthma Proceedings. 2012;33:227. doi: 10.2500/aap.2012.33.3569. [DOI] [PubMed] [Google Scholar]
- Spergel JM Epidemiology of atopic dermatitis and atopic march in children. Immunology and Allergy Clinics of North America. 2010;30:269. doi: 10.1016/j.iac.2010.06.003. [DOI] [PubMed] [Google Scholar]
- Tada J Diagnostic standard for atopic dermatitis. Japan Medical Association Journal. 2002;45:460. [Google Scholar]
- Hanifin JM Diagnostic features of atopic dermatitis. Acta Dermato-Venereologica. 1980;92:44. [PubMed] [Google Scholar]
- De D Kanwar A Handa S Comparative efficacy of Hanifin and Rajka's criteria and the UK working party's diagnostic criteria in diagnosis of atopic dermatitis in a hospital setting in North India. Journal of the European Academy of Dermatology and Venereology. 2006;20:853. doi: 10.1111/j.1468-3083.2006.01664.x. [DOI] [PubMed] [Google Scholar]
- Williams H Burney P Pembroke A Hay RJ Validation of the UK diagnostic criteria for atopic dermatitis in a population setting. British Journal of Dermatology. 1996;135:12. [PubMed] [Google Scholar]
- Lee SC Committee of Korean Atopic Dermatitis Association for REACH. Various diagnostic criteria for atopic dermatitis (AD): A proposal of Reliable Estimation of Atopic Dermatitis in Childhood (REACH) criteria, a novel questionnaire‐based diagnostic tool for AD. Journal of Dermatology. 2016;43:376. doi: 10.1111/1346-8138.13264. [DOI] [PubMed] [Google Scholar]
- Eichenfield LF Tom WL Chamlin SL Feldman SR Hanifin JM Guidelines of care for the management of atopic dermatitis: section 1. Diagnosis and assessment of atopic dermatitis. Journal of the American Academy of Dermatology. 2014;70:338. doi: 10.1016/j.jaad.2013.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hanifin JM Cooper KD Ho VC Kang S Krafchik BR Guidelines of care for atopic dermatitis. Journal of the American Academy of Dermatology. 2004;50:391. doi: 10.1016/j.jaad.2003.08.003. [DOI] [PubMed] [Google Scholar]
- Williams H Jburney P Hay R Archer C Shipley M The UK Working Party’s Diagnostic Criteria for Atopic Dermatitis. I. Derivation of a minimum set of discriminators for atopic dermatitis. British Journal of Dermatology. 1994;131:383. doi: 10.1111/j.1365-2133.1994.tb08530.x. [DOI] [PubMed] [Google Scholar]
- Strina A Barreto ML Cunha S F tima S Moreira SC Validation of epidemiological tools for eczema diagnosis in Brazilian children: the ISAAC's and UK Working Party's criteria. BMC Dermatology. 2010;225:11. doi: 10.1186/1471-5945-10-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Odhiambo JA Williams HC Clayton TO Robertson CF Asher MI Global variations in prevalence of eczema symptoms in children from ISAAC Phase Three. Journal of Allergy and Clinical Immunology. 2009;124:1251. doi: 10.1016/j.jaci.2009.10.009. [DOI] [PubMed] [Google Scholar]
- Stalder J Taieb A Atherton D Bieber P Bonifazi E Severity scoring of atopic dermatitis: the SCORAD index: consensus report of the European Task Force on Atopic Dermatitis. Dermatology. 1993;186:23. doi: 10.1159/000247298. [DOI] [PubMed] [Google Scholar]
- Oranje AP Stalder JF Taïeb A Tasset C de Longueville M. Scoring of atopic dermatitis by SCORAD using a training atlas by investigators from different disciplines. Pediatric Allergy and Immunology. 1997;239:28. doi: 10.1111/j.1399-3038.1997.tb00139.x. [DOI] [PubMed] [Google Scholar]
- Wolkerstorfer A De Waard van der Spek F Glazenburg E Mulder P Oranje A Scoring the severity of atopic dermatitis: three item severity score as a rough system for daily practice and as a pre-screening tool for studies. Acta Dermato-Venereologica. 1999;79:356. doi: 10.1080/000155599750010256. [DOI] [PubMed] [Google Scholar]
- Charman C Venn A Williams H Reliability testing of the six area, six sign atopic dermatitis severity score. British Journal of Dermatology. 2002;146:1057. doi: 10.1046/j.1365-2133.2002.04644.x. [DOI] [PubMed] [Google Scholar]
- Hanifin J Thurston M Omoto M Cherill R Tofte S The eczema area and severity index (EASI): assessment of reliability in atopic dermatitis. Experimental Dermatology. 2001;10:11. doi: 10.1034/j.1600-0625.2001.100102.x. [DOI] [PubMed] [Google Scholar]
- Oranje AP Practical issues on interpretation of scoring atopic dermatitis: SCORAD Index, objective SCORAD, patient-oriented SCORAD and Three-Item Severity score. Current Problems in Dermatology. 2011;41:149. doi: 10.1159/000323308. [DOI] [PubMed] [Google Scholar]
- Gelmetti C Colonna C The value of SCORAD and beyond: Towards a standardized evaluation of severity? Allergy. 2004;59:61. doi: 10.1111/j.1398-9995.2004.00651.x. [DOI] [PubMed] [Google Scholar]
- Krol A Krafchik B The differential diagnosis of atopic dermatitis in childhood. Dermatologic Therapy. 2006;19:73. doi: 10.1111/j.1529-8019.2006.00058.x. [DOI] [PubMed] [Google Scholar]
- Siegfried E Hebert A Diagnosis of atopic dermatitis: mimics, overlaps, and complications. Journal of Clinical Medicine. 2015;4:884. doi: 10.3390/jcm4050884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett M Luu M Differential diagnosis of atopic dermatitis. Immunology and Allergy Clinics of North America. 2017;37:11. doi: 10.1016/j.iac.2016.08.009. [DOI] [PubMed] [Google Scholar]
- Gaudinski MR Milner JD Atopic dermatitis and allergic urticaria: cutaneous manifestations of immunodeficiency. Immunology and Allergy Clinics of North America. 2017;37:1. doi: 10.1016/j.iac.2016.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lyons JJ Milner JD Stone KD Atopic dermatitis in children: clinical features, pathophysiology, and treatment. Immunology and Allergy Clinics of North America. 2015;35:161. doi: 10.1016/j.iac.2014.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saunders SP Goh CS Brown SJ Palmer CN Porter RM Tmem79/Matt is the matted mouse gene and is a predisposing gene for atopic dermatitis in human subjects. Journal of Allergy and Clinical Immunology. 2013;132:1121. doi: 10.1016/j.jaci.2013.08.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chavanas S Bodemer C Rochat A Hamel-Teillac D Ali M Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nature Genetics. 2000;25:141. doi: 10.1038/75977. [DOI] [PubMed] [Google Scholar]
- Lee H-J Lee S-H Epidermal permeability barrier defects and barrier repair therapy in atopic dermatitis. Allergy, Asthma & Immunology Research. 2014;6:276. doi: 10.4168/aair.2014.6.4.276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brattsand M Stefansson K Lundh C Haasum Y Egelrud T A proteolytic cascade of kallikreins in the stratum corneum. Journal of Investigative Dermatology. 2005;124:198. doi: 10.1111/j.0022-202X.2004.23547.x. [DOI] [PubMed] [Google Scholar]
- Moniaga CS Jeong SK Egawa G Nakajima S Hara-Chikuma M Protease activity enhances production of thymic stromal lymphopoietin and basophil accumulation in flaky tail mice. American Journal of Pathology. 2013;182:841. doi: 10.1016/j.ajpath.2012.11.039. [DOI] [PubMed] [Google Scholar]
- Bager P Wohlfahrt J Thyssen JP Melbye M. Filaggrin genotype and skin diseases independent of atopic dermatitis in childhood. Pediatric Allergy and Immunology. 2016;27:162. doi: 10.1111/pai.12511. [DOI] [PubMed] [Google Scholar]
- Nagao K Kubo A Hata T Shimizu A Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. Journal of Allergy and Clinical Immunology. 2012;129:1538. doi: 10.1016/j.jaci.2012.01.068. [DOI] [PubMed] [Google Scholar]
- Thyssen JP Kezic S. Causes of epidermal filaggrin reduction and their role in the pathogenesis of atopic dermatitis. Journal of Allergy and Clinical Immunology. 2014;134:792. doi: 10.1016/j.jaci.2014.06.014. [DOI] [PubMed] [Google Scholar]
- Janssens M van Smeden J Gooris GS Bras W Portale G Increase in short-chain ceramides correlates with an altered lipid organization and decreased barrier function in atopic eczema patients. Journal of Lipid Research. 2012;53:2755. doi: 10.1194/jlr.P030338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Benedetto A Rafaels NM McGirt LY Ivanov AI Georas SN Tight junction defects in patients with atopic dermatitis. Journal of Allergy and Clinical Immunology. 2011;127:773. doi: 10.1016/j.jaci.2010.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;11:1. doi: 10.1371/journal.pone.0161759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lack G Fox D Northstone K Golding J Factors associated with the development of peanut allergy in childhood. New England Journal of Medicine. 2003;348:977. doi: 10.1056/NEJMoa013536. [DOI] [PubMed] [Google Scholar]
- Eigenmann PA Sicherer SH Borkowski TA Cohen BA Sampson HA Prevalence of IgE-mediated food allergy among children with atopic dermatitis. Pediatrics. 1998;101:e8–e8. doi: 10.1542/peds.101.3.e8. [DOI] [PubMed] [Google Scholar]
- Sybilski AJ Zalewska M czyk K Lipiec A Krzychta E The prevalence of sensitization to inhalant allergens in children with atopic dermatitis. Allergy & Asthma Proceedings. 2015;324:81. doi: 10.2500/aap.2015.36.3882. [DOI] [PubMed] [Google Scholar]
- Heilmann C Adhesion mechanisms of staphylococci. Advances in Experimental Medicine and Biology. 2011;715:105. doi: 10.1007/978-94-007-0940-9_7. [DOI] [PubMed] [Google Scholar]
- Abeck D Mempel M. colonization in atopic dermatitis and its therapeutic implications. British Journal of Dermatology. 1998;139:13. doi: 10.1046/j.1365-2133.1998.1390s3013.x. [DOI] [PubMed] [Google Scholar]
- Kaesler S Volz T Skabytska Y Kberle M Hein U Toll-like receptor 2 ligands promote chronic atopic dermatitis through IL-4-mediated suppression of IL-10. Journal of Allergy and Clinical Immunology. 2014;246:92. doi: 10.1016/j.jaci.2014.02.017. [DOI] [PubMed] [Google Scholar]
- Brauweiler AM Goleva E Leung DY Th2 cytokines increase alpha toxin-induced keratinocyte death through the signal transducer and activator of transcription 6 (STAT6) Journal of Investigative Dermatology. 2014;134:2114. doi: 10.1038/jid.2014.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller LS Cho JS cutaneous infections. Nature Reviews Immunology. 2011;11:505. doi: 10.1038/nri3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brauweiler AM Bin L Kim BE Oyoshi MK Geha RS Filaggrin-dependent secretion of sphingomyelinase protects against staphylococcal. Journal of Allergy and Clinical Immunology. 2013;945:421. doi: 10.1016/j.jaci.2012.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miajlovic H Fallon PG Irvine AD Foster TJ Effect of filaggrin breakdown products on growth of and protein expression by. Journal of Allergy and Clinical Immunology. 2010;126:1184. doi: 10.1016/j.jaci.2010.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raap U Wichmann K Bruder M Ständer S Wedi B Correlation of IL-31 serum levels with severity of atopic dermatitis. Journal of Allergy and Clinical Immunology. 2008;228:421. doi: 10.1016/j.jaci.2008.05.047. [DOI] [PubMed] [Google Scholar]
- Cho S-H Strickland I Tomkinson A Fehringer AP Gelfand EW Preferential binding of Staphylococcus aureus to skin sites of Th2-mediated inflammation in a murine model. Journal of Investigative Dermatology. 2001;116:658. doi: 10.1046/j.0022-202x.2001.01331.x. [DOI] [PubMed] [Google Scholar]
- Mempel M Schmidt T Weidinger S Schnopp C Ring J surface-associated proteins in the attachment to cultured HaCaT keratinocytes in a new adhesion assay. Journal of Investigative Dermatology. 1998;111:452. doi: 10.1046/j.1523-1747.1998.00293.x. [DOI] [PubMed] [Google Scholar]
- Czarnowicki T Krueger JG Guttman-Yassky E Skin barrier and immune dysregulation in atopic dermatitis: an evolving story with important clinical implications. Journal of Allergy and Clinical Immunology in Practice. 2014;2:371. doi: 10.1016/j.jaip.2014.03.006. [DOI] [PubMed] [Google Scholar]
- Bitschar K Wolz C Krismer B Peschel A Schittek B Keratinocytes as sensors and central players in the immune defense against Staphylococcus aureus in the skin. Journal of Dermatological Science. 2017;87:215. doi: 10.1016/j.jdermsci.2017.06.003. [DOI] [PubMed] [Google Scholar]
- Eyerich K Novak N Immunology of atopic eczema: overcoming the T h1/T h2 paradigm. Allergy. 2013;68:974. doi: 10.1111/all.12184. [DOI] [PubMed] [Google Scholar]
- Samrao A Berry TM Goreshi R Simpson EL A pilot study of an oral phosphodiesterase inhibitor (apremilast) for atopic dermatitis in adults. Archives of Dermatology. 2012;148:890. doi: 10.1001/archdermatol.2012.812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wittmann M Helliwell PS Phosphodiesterase 4 inhibition in the treatment of psoriasis, psoriatic arthritis and other chronic inflammatory diseases. Dermatologic Therapy. 2013;3:1. doi: 10.1007/s13555-013-0023-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aoki W Ueda M Characterization of antimicrobial peptides toward the development of novel antibiotics. Pharmaceuticals. 2013;6:1055. doi: 10.3390/ph6081055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gittler JK Shemer A Suárez-Fariñas M Fuentes-Duculan J Gulewicz KJ Progressive activation of TH2/TH22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. Journal of Allergy and Clinical Immunology. 2012;225:1344. doi: 10.1016/j.jaci.2012.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henderson J Northstone K Lee SP Liao H Zhao Y The burden of disease associated with filaggrin mutations: a population-based, longitudinal birth cohort study. Journal of Allergy and Clinical Immunology. 2008;121:872. doi: 10.1016/j.jaci.2008.01.026. [DOI] [PubMed] [Google Scholar]
- Boniface K Bernard F-X Garcia Gurney AL Lecron J-C -22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. Journal of Immunology. 2005;174:3695. doi: 10.4049/jimmunol.174.6.3695. [DOI] [PubMed] [Google Scholar]
- Czarnowicki T Gonzalez J Shemer A Malajian D Xu H Severe atopic dermatitis is characterized by selective expansion of circulating TH2/TC2 and TH22/TC22, but not TH17/TC17, cells within the skin-homing T-cell population. Journal of Allergy and Clinical Immunology. 2015;136:104. doi: 10.1016/j.jaci.2015.01.020. [DOI] [PubMed] [Google Scholar]
- Liu Y-J Thymic stromal lymphopoietin: master switch for allergic inflammation. Journal of Experimental Medicine. 2006;203:269. doi: 10.1084/jem.20051745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Omori M Ziegler S. Induction of IL-4 expression in CD4+ T cells by thymic stromal lymphopoietin. Journal of Immunology. 2007;178:1396. doi: 10.4049/jimmunol.178.3.1396. [DOI] [PubMed] [Google Scholar]
- Brandt EB Sivaprasad U Th2 cytokines and atopic dermatitis. Journal of Clinical and Cellular Immunology. 2011;2:1. doi: 10.4172/2155-9899.1000110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jakubzick C Gautier EL Gibbings SL Sojka DK Schlitzer A Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity. 2013;39:599. doi: 10.1016/j.immuni.2013.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zheng T Oh MH Oh SY Schroeder JT Glick AB Transgenic expression of interleukin-13 in the skin induces a pruritic dermatitis and skin remodeling. Journal of Investigative Dermatology. 2009;129:742. doi: 10.1038/jid.2008.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Izuhara K Nunomura S Nanri Y Ogawa M Ono J Periostin in inflammation and allergy. Cellular and Molecular Life Sciences. 2017;74:4293. doi: 10.1007/s00018-017-2648-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Namkung JH Lee JE Kim E Cho HJ Kim S IL-5 and IL-5 receptor alpha polymorphisms are associated with atopic dermatitis in Koreans. Allergy. 2007;62:934. doi: 10.1111/j.1398-9995.2007.01445.x. [DOI] [PubMed] [Google Scholar]
- Furue M Yamamura K Nakahara M Nakahara T Fukui Y Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy. 2018;8208:29. doi: 10.1111/all.13239. [DOI] [PubMed] [Google Scholar]
- Bilsborough J Leung DY Maurer M Howell M Boguniewcz M -31 is associated with cutaneous lymphocyte antigen-positive skin homing T cells in patients with atopic dermatitis. Journal of Allergy and Clinical Immunology. 2006;117:418. doi: 10.1016/j.jaci.2005.10.046. [DOI] [PubMed] [Google Scholar]
- Kunsleben N R drich U Gehring M Novak N Kapp A -31 induces chemotaxis, calcium mobilization, release of reactive oxygen species, and CCL26 in eosinophils, which are capable to release IL-31. Journal of Investigative Dermatology. 2015;252:1908. doi: 10.1038/jid.2015.106. [DOI] [PubMed] [Google Scholar]
- Noda S Krueger JG Guttman-Yassky E The translational revolution and use of biologics in patients with inflammatory skin diseases. Journal of Allergy and Clinical Immunology. 2015;135:324. doi: 10.1016/j.jaci.2014.11.015. [DOI] [PubMed] [Google Scholar]
- Schittek B The antimicrobial skin barrier in patients with atopic dermatitis. Current Problems in Dermatology. 2011;41:67–67. doi: 10.1159/000323296. [DOI] [PubMed] [Google Scholar]
- Park H Li Z Yang XO Chang SH Nurieva R A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nature Immunology. 2005;6:1069. doi: 10.1038/ni1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toda M Leung DY Molet S Boguniewicz M Taha R Polarized in vivo expression of IL-11 and IL-17 between acute and chronic skin lesions. Journal of Allergy and Clinical Immunology. 2003;111:875. doi: 10.1067/mai.2003.1414. [DOI] [PubMed] [Google Scholar]
- Esaki H Brunner PM Renert-Yuval Y Czarnowicki T Huynh T Early-onset pediatric atopic dermatitis is TH2 but also TH17 polarized in skin. Journal of Allergy and Clinical Immunology. 2016;138:1639. doi: 10.1016/j.jaci.2016.07.013. [DOI] [PubMed] [Google Scholar]
- Ikoma A Steinhoff M nder S Yosipovitch G Schmelz M. The neurobiology of itch. Nature Reviews Neuroscience. 2006;228:535. doi: 10.1038/nrn1950. [DOI] [PubMed] [Google Scholar]
- Kamo A Tominaga M Taneda K Ogawa H Takamori K. Neurotropin inhibits the increase in intraepidermal nerve density in the acetone-treated dry-skin mouse model. Clinical and Experimental Dermatology. 2013;38:665. doi: 10.1111/ced.12100. [DOI] [PubMed] [Google Scholar]
- Sonkoly E Muller A Lauerma AI Pivarcsi A Soto H -31: a new link between T cells and pruritus in atopic skin inflammation. Journal of Allergy and Clinical Immunology. 2006;117:411. doi: 10.1016/j.jaci.2005.10.033. [DOI] [PubMed] [Google Scholar]
- Wilson SR Batia LM Beattie K Katibah GE McClain SP The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell. 2013;155:285. doi: 10.1016/j.cell.2013.08.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murota H Katayama I Evolving understanding on the aetiology of thermally provoked itch. European Journal of Pain. 2016;20:47. doi: 10.1002/ejp.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murota H Izumi M El-Latif MIA Nishioka M Terao M Artemin causes hypersensitivity to warm sensation, mimicking warmth-provoked pruritus in atopic dermatitis. Journal of Allergy and Clinical Immunology. 2012;130:671. doi: 10.1016/j.jaci.2012.05.027. [DOI] [PubMed] [Google Scholar]
- Lavery MJ Stull C Kinney MO Yosipovitch G Nocturnal pruritus: the battle for a peaceful night's sleep. International Journal of Molecular Sciences. 2016;17:425–425. doi: 10.3390/ijms17030425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams J Burr M Williams H Factors influencing atopic dermatitis—a questionnaire survey of schoolchildren's perceptions. British Journal of Dermatology. 2004;150:1154. doi: 10.1111/j.1365-2133.2004.05869.x. [DOI] [PubMed] [Google Scholar]
- Shiohara T Doi T Hayakawa J Defective sweating responses in atopic dermatitis. Current Problems in Dermatology. 2011;41:68. doi: 10.1159/000323297. [DOI] [PubMed] [Google Scholar]
- Esparza-Gordillo J Weidinger S Flster-Holst R Bauerfeind A Ruschendorf F A common variant on chromosome 11q13 is associated with atopic dermatitis. Nature Genetics. 2009;246:596. doi: 10.1038/ng.347. [DOI] [PubMed] [Google Scholar]
- Bin L Leung DY Genetic and epigenetic studies of atopic dermatitis. Allergy, Asthma & Clinical Immunology. 2016;12:52–52. doi: 10.1186/s13223-016-0158-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paternoster L Standl M Chen C-M Ramasamy Bnnelykke K Meta-analysis of genome-wide association studies identifies three new risk loci for atopic dermatitis. Nature Genetics. 2012;248:187. doi: 10.1038/ng.1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang S Hon K Kong A Tang M Sy HY Eczema phenotypes are associated with multiple vitamin D pathway genes in Chinese children. Allergy. 2014;69:118. doi: 10.1111/all.12337. [DOI] [PubMed] [Google Scholar]
- Hussein YM Shalaby SM Nassar A Alzahrani SS Alharbi AS Association between genes encoding components of the IL-4/IL-4 receptor pathway and dermatitis in children. Gene. 2014;545:276. doi: 10.1016/j.gene.2014.04.024. [DOI] [PubMed] [Google Scholar]
- Amarasekera M Prescott SL Palmer DJ Nutrition in early life, immune-programming and allergies: the role of epigenetics. Asian Pacific Journal of Allergy and Immunology. 2013;31:175. [PubMed] [Google Scholar]
- Wang Y Liang Y Lu Q. MicroRNA epigenetic alterations: predicting biomarkers and therapeutic targets in human diseases. Clinical Genetics. 2008;74:307. doi: 10.1111/j.1399-0004.2008.01075.x. [DOI] [PubMed] [Google Scholar]
- Jirtle RL Skinner MK Environmental epigenomics and disease susceptibility. Nature Reviews Genetics. 2007;8:253. doi: 10.1038/nrg2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodríguez E Baurecht H Wahn AF Kretschmer A Hotze M An integrated epigenetic and transcriptomic analysis reveals distinct tissue-specific patterns of DNA methylation associated with atopic dermatitis. Journal of Investigative Dermatology. 2014;237:1873. doi: 10.1038/jid.2014.87. [DOI] [PubMed] [Google Scholar]
- Hinz D Bauer M der S Olek S Huehn J Cord blood T regs with stable FOXP3 expression are influenced by prenatal environment and associated with atopic dermatitis at the age of one year. Allergy. 2012;246:380. doi: 10.1111/j.1398-9995.2011.02767.x. [DOI] [PubMed] [Google Scholar]
- Makeyev EV Maniatis T Multilevel regulation of gene expression by microRNAs. Science. 2008;319:1789. doi: 10.1126/science.1152326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonkoly E Janson P Majuri M-L Savinko T Fyhrquist N -155 is overexpressed in patients with atopic dermatitis and modulates T-cell proliferative responses by targeting cytotoxic T lymphocyte-associated antigen 4. Journal of Allergy and Clinical Immunology. 2010;126:581. doi: 10.1016/j.jaci.2010.05.045. [DOI] [PubMed] [Google Scholar]
- Herberth G Bauer M Gasch M Hinz D R der S Maternal and cord blood miR-223 expression associates with prenatal tobacco smoke exposure and low regulatory T-cell numbers. Journal of Allergy and Clinical Immunology. 2014;246:543. doi: 10.1016/j.jaci.2013.06.036. [DOI] [PubMed] [Google Scholar]
- Oranje A Proactive therapy and emollient therapy in atopic dermatitis. Current Treatment Options in Allergy. 2014;1:365. [Google Scholar]
- Eichenfield LF Tom WL Berger TG Krol A Paller AS Guidelines of care for the management of atopic dermatitis: Section 2. Management and treatment of atopic dermatitis with topical therapies. Journal of the American Academy of Dermatology. 2014;71:116. doi: 10.1016/j.jaad.2014.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carr WW Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Pediatric Drugs. 2013;15:303. doi: 10.1007/s40272-013-0013-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draelos ZD An evaluation of prescription device moisturizers. Journal of Cosmetic Dermatology. 2009;8:40. doi: 10.1111/j.1473-2165.2009.00422.x. [DOI] [PubMed] [Google Scholar]
- Boguniewicz M Fonacier L Guttman-Yassky E Ong PY Silverberg J Atopic dermatitis yardstick: Practical recommendations for an evolving therapeutic landscape. Annals of Allergy, Asthma & Immunology. 2018;120:10. doi: 10.1016/j.anai.2017.10.039. [DOI] [PubMed] [Google Scholar]
- Dabade TS Davis DM Wetter DA Hand JL McEvoy MT Wet dressing therapy in conjunction with topical corticosteroids is effective for rapid control of severe pediatric atopic dermatitis: experience with 218 patients over 30 years at Mayo Clinic. Journal of the American Academy of Dermatology. 2012;67:100. doi: 10.1016/j.jaad.2011.06.025. [DOI] [PubMed] [Google Scholar]
- Huang JT Abrams M Tlougan B Rademaker A Paller AS colonization in atopic dermatitis decreases disease severity. Pediatrics. 2009;123:808. doi: 10.1542/peds.2008-2217. [DOI] [PubMed] [Google Scholar]
- Sayaseng KY Vernon P Pathophysiology and management of mild to moderate pediatric atopic dermatitis. Journal of Pediatric Health Care. 2018;32:2. doi: 10.1016/j.pedhc.2017.10.002. [DOI] [PubMed] [Google Scholar]
- Barnes TM Greive KA Use of bleach baths for the treatment of infected atopic eczema. Australasian Journal of Dermatology. 2013;54:251. doi: 10.1111/ajd.12015. [DOI] [PubMed] [Google Scholar]
- Eichenfield L Totri C Optimizing outcomes for paediatric atopic dermatitis. British Journal of Dermatology. 2014;170:31. doi: 10.1111/bjd.12976. [DOI] [PubMed] [Google Scholar]
- Chopra R Vakharia PP Sacotte R Silverberg JI Efficacy of bleach baths in reducing severity of atopic dermatitis: a systematic review and meta-analysis. Annals of Allergy, Asthma & Immunology. 2017;119:435. doi: 10.1016/j.anai.2017.08.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schnopp C Ring J Mempel M. The role of antibacterial therapy in atopic eczema. Expert Opinion on Pharmacotherapy. 2010;11:929. doi: 10.1517/14656561003659992. [DOI] [PubMed] [Google Scholar]
- Lebon A Labout JA Verbrugh HA Jaddoe VW Hofman A Role of Staphylococcus aureus nasal colonization in atopic dermatitis in infants: The Generation R Study. Archives of Pediatrics and Adolescent Medicine. 2009;163:745. doi: 10.1001/archpediatrics.2009.117. [DOI] [PubMed] [Google Scholar]
- Howell MD Gao P Kim BE Lesley LJ Streib JE The signal transducer and activator of transcription 6 gene (STAT6) increases the propensity of patients with atopic dermatitis toward disseminated viral skin infections. Journal of Allergy and Clinical Immunology. 2011;128:1006. doi: 10.1016/j.jaci.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonamonte D Belloni Fortina A Neri L Patrizi A Fusidic acid in skin infections and infected atopic eczema. Giornale Italiano di Dermatologia e Venereologia. 2014;149:453. [PubMed] [Google Scholar]
- Gelmetti C Local antibiotics in dermatology. Dermatologic Therapy. 2008;21:187. doi: 10.1111/j.1529-8019.2008.00190.x. [DOI] [PubMed] [Google Scholar]
- Travers JB Kozman A Yao Y Ming W Yao W Treatment outcomes of secondarily impetiginized pediatric atopic dermatitis lesions and the role of oral antibiotics. Pediatric Dermatology. 2012;29:289. doi: 10.1111/j.1525-1470.2011.01661.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boguniewicz M Sampson H Leung SB Harbeck R Leung DY Effects of cefuroxime axetil on Staphylococcus aureus colonization and superantigen production in atopic dermatitis. Journal of Allergy and Clinical Immunology. 2001;108:651. doi: 10.1067/mai.2001.118598. [DOI] [PubMed] [Google Scholar]
- Liu C Bayer A Cosgrove SE Daum RS Fridkin SK Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clinical Infectious Diseases. 2011;52:18. doi: 10.1093/cid/ciq146. [DOI] [PubMed] [Google Scholar]
- Aronson PL Yan AC Mittal MK Mohamad Z Shah SS Delayed acyclovir and outcomes of children hospitalized with eczema herpeticum. Pediatrics. 2011;128:1161. doi: 10.1542/peds.2011-0948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stuetz A Baumann K Grassberger M Wolff K Meingassner JG Discovery of topical calcineurin inhibitors and pharmacological profile of pimecrolimus. International Archives of Allergy and Immunology. 2006;141:199. doi: 10.1159/000095289. [DOI] [PubMed] [Google Scholar]
- Eichenfield LF Ahluwalia J Waldman A Borok J Udkoff J Current guidelines for the evaluation and management of atopic dermatitis: A comparison of the Joint Task Force Practice Parameter and American Academy of Dermatology Guidelines. Alergologia Polska-Polish Journal of Allergology. 2017;4:158. doi: 10.1016/j.jaci.2017.01.009. [DOI] [PubMed] [Google Scholar]
- Thomas K Williams H Armstrong S O'neill C Avery T Randomized controlled trial of short bursts of a potent topical corticosteroid versus more prolonged use of a mild preparation for children with atopic eczema. British Journal of Dermatology. 2002;8217:541. doi: 10.1136/bmj.324.7340.768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Draelos ZD Use of topical corticosteroids and topical calcineurin inhibitors for the treatment of atopic dermatitis in thin and sensitive skin areas. Current Medical Research and Opinion. 2008;24:985. doi: 10.1185/030079908x280419. [DOI] [PubMed] [Google Scholar]
- Hong E Smith S Fischer G Evaluation of the atrophogenic potential of topical corticosteroids in pediatric dermatology patients. Pediatric Dermatology. 2011;28:393. doi: 10.1111/j.1525-1470.2011.01445.x. [DOI] [PubMed] [Google Scholar]
- Ellison JA Patel L Ray DW David TJ Clayton PE Hypothalamic–pituitary–adrenal function and glucocorticoid sensitivity in atopic dermatitis. Pediatrics. 2000;8211:794. doi: 10.1542/peds.105.4.794. [DOI] [PubMed] [Google Scholar]
- Del Rosso J Friedlander SF Corticosteroids: options in the era of steroid-sparing therapy. Journal of the American Academy of Dermatology. 2005;53:50. doi: 10.1016/j.jaad.2005.04.030. [DOI] [PubMed] [Google Scholar]
- Manthripragada AD Pinheiro SP MaCurdy TE Saneinejad S Worrall CM Off-label topical calcineurin inhibitor use in children. Pediatrics. 2013;132:1327. doi: 10.1542/peds.2013-0931. [DOI] [PubMed] [Google Scholar]
- Breuer K Werfel T Kapp A Safety and efficacy of topical calcineurin inhibitors in the treatment of childhood atopic dermatitis. American Journal of Clinical Dermatology. 2005;6:65. doi: 10.2165/00128071-200506020-00001. [DOI] [PubMed] [Google Scholar]
- Svensson A Chambers C nemo A Mitchell S A systematic review of tacrolimus ointment compared with corticosteroids in the treatment of atopic dermatitis. Current Medical Research and Opinion. 2011;229:1395. doi: 10.1185/03007995.2011.582483. [DOI] [PubMed] [Google Scholar]
- El-Batawy MMY Bosseila MA Mashaly HM Hafez VSG Topical calcineurin inhibitors in atopic dermatitis: a systematic review and meta-analysis. Journal of Dermatological Science. 2009;54:76. doi: 10.1016/j.jdermsci.2009.02.002. [DOI] [PubMed] [Google Scholar]
- Hengge UR Ruzicka T Schwartz RA Cork MJ Adverse effects of topical glucocorticosteroids. Journal of the American Academy of Dermatology. 2006;54:1. doi: 10.1016/j.jaad.2005.01.010. [DOI] [PubMed] [Google Scholar]
- Chen S-L Yan J Wang F-S Two topical calcineurin inhibitors for the treatment of atopic dermatitis in pediatric patients: a meta-analysis of randomized clinical trials. Journal of Dermatological Treatment. 2010;21:144. doi: 10.3109/09546630903401470. [DOI] [PubMed] [Google Scholar]
- Ormerod A Topical tacrolimus and pimecrolimus and the risk of cancer: how much cause for concern? British Journal of Dermatology. 2005;153:701. doi: 10.1111/j.1365-2133.2005.06899.x. [DOI] [PubMed] [Google Scholar]
- Kempers S Boguniewicz M Carter E Jarratt M Pariser D A randomized investigator-blinded study comparing pimecrolimus cream 1% with tacrolimus ointment 0.03% in the treatment of pediatric patients with moderate atopic dermatitis. Journal of the American Academy of Dermatology. 2004;51:515. doi: 10.1016/j.jaad.2004.01.051. [DOI] [PubMed] [Google Scholar]
- Thaci D Reitamo S Gonzalez Ensenat M Moss C Boccaletti V Proactive disease management with 0· 03% tacrolimus ointment for children with atopic dermatitis: results of a randomized, multicentre, comparative study. British Journal of Dermatology. 2008;159:1348. doi: 10.1111/j.1365-2133.2008.08813.x. [DOI] [PubMed] [Google Scholar]
- Akdis CA Akdis M Bieber T Bindslev-Jensen C Boguniewicz M Diagnosis and treatment of atopic dermatitis in children and adults: European Academy of Allergology. Asthma and Immunology/PRACTALL Consensus Report. Allergy. 2006;61:969. doi: 10.1111/j.1398-9995.2006.01153.x. [DOI] [PubMed] [Google Scholar]
- Ring J Alomar A Bieber T Deleuran M Fink-Wagner A Guidelines for treatment of atopic eczema (atopic dermatitis) part I. Journal of the European Academy of Dermatology and Venereology. 2012;26:1045. doi: 10.1111/j.1468-3083.2012.04635.x. [DOI] [PubMed] [Google Scholar]
- La Rosa M Musarra I Ranno C Maiello N Negri L A randomized, double-blind, placebo-controlled, crossover trial of systemic flunisolide in the treatment of children with severe atopic dermatitis. Current Therapeutic Research. 1995;56:720. [Google Scholar]
- Saag KG Koehnke R Caldwell JR Brasington R Burmeister LF Low dose long-term corticosteroid therapy in rheumatoid arthritis: an analysis of serious adverse events. American Journal of Medicine. 1994;96:115. doi: 10.1016/0002-9343(94)90131-7. [DOI] [PubMed] [Google Scholar]
- Nygaard U Vestergaard C Deleuran M Systemic treatment of severe atopic dermatitis in children and adults. Current Treatment Options in Allergy. 2014;1:384. [Google Scholar]
- Eichenfield LF Hanifin JM Luger TA Stevens SR Pride HB Consensus conference on pediatric atopic dermatitis. Journal of the American Academy of Dermatology. 2003;49:1088. doi: 10.1016/s0190-9622(03)02539-8. [DOI] [PubMed] [Google Scholar]
- Griffiths C Katsambas A Dijkmans B Finlay A Ho V Update on the use of ciclosporin in immune-mediated dermatoses. British Journal of Dermatology. 2006;155:1. doi: 10.1111/j.1365-2133.2006.07343.x. [DOI] [PubMed] [Google Scholar]
- Madan V Griffiths CM Systemic ciclosporin and tacrolimus in dermatology. Dermatologic Therapy. 2007;20:239. doi: 10.1111/j.1529-8019.2007.00137.x. [DOI] [PubMed] [Google Scholar]
- Naeyaert J Lachapelle J Degreef H Brassinne Mdl Heenen M Pharmacology and treatment: cyclosporin in atopic dermatitis. Review of the Literature and Outline of a Belgian Consensus. Dermatology. 1999;198:45. doi: 10.1159/000018091. [DOI] [PubMed] [Google Scholar]
- Harper J Ahmed I Barclay G Lacour M Hoeger P Cyclosporin for severe childhood atopic dermatitis: short course versus continuous therapy. British Journal of Dermatology. 2000;142:52. doi: 10.1046/j.1365-2133.2000.03241.x. [DOI] [PubMed] [Google Scholar]
- Schmitt J Schmitt N Meurer M. Cyclosporin in the treatment of patients with atopic eczema-a systematic review and meta-analysis. Journal of the European Academy of Dermatology and Venereology. 2007;21:606. doi: 10.1111/j.1468-3083.2006.02023.x. [DOI] [PubMed] [Google Scholar]
- Darsow U Wollenberg A Simon D eb A Werfel T ETFAD/EADV eczema task force 2009 position paper on diagnosis and treatment of atopic dermatitis. Journal of the European Academy of Dermatology and Venereology. 2010;239:317. doi: 10.1111/j.1468-3083.2009.03415.x. [DOI] [PubMed] [Google Scholar]
- Berth-Jones J Takwale A Tan E Barclay G Agarwal S Azathioprine in severe adult atopic dermatitis: a double-blind, placebo-controlled, crossover trial. British Journal of Dermatology. 2002;147:324. doi: 10.1046/j.1365-2133.2002.04989.x. [DOI] [PubMed] [Google Scholar]
- Sidbury R Davis DM Cohen DE Cordoro KM Berger TG Guidelines of care for the management of atopic dermatitis: Section 3. Management and treatment with phototherapy and systemic agents. Journal of the American Academy of Dermatology. 2014;71:327. doi: 10.1016/j.jaad.2014.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El-Khalawany MA 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. European Journal of Pediatrics. 2013;172:351. doi: 10.1007/s00431-012-1893-3. [DOI] [PubMed] [Google Scholar]
- Deo M Yung A Hill S Rademaker M Methotrexate for treatment of atopic dermatitis in children and adolescents. International Journal of Dermatology. 2014;53:1037. doi: 10.1111/ijd.12314. [DOI] [PubMed] [Google Scholar]
- Hsu C-J Wang L-F Emerging treatment of atopic dermatitis. Clinical Reviews in Allergy & Immunology. 2007;33:199. doi: 10.1007/s12016-007-0043-6. [DOI] [PubMed] [Google Scholar]
- Grundmann-Kollmann M Korting H Behrens S Leiter U hn G Successful treatment of severe refractory atopic dermatitis with mycophenolate mofetil. British Journal of Dermatology. 1999;141:175–175. doi: 10.1046/j.1365-2133.1999.02952.x. [DOI] [PubMed] [Google Scholar]
- Heller M Shin H Orlow S Schaffer J Mycophenolate mofetil for severe childhood atopic dermatitis: experience in 14 patients. British Journal of Dermatology. 2007;157:127. doi: 10.1111/j.1365-2133.2007.07947.x. [DOI] [PubMed] [Google Scholar]
- Downing HJ Pirmohamed M Beresford MW Smyth RL Paediatric use of mycophenolate mofetil. British Journal of Clinical Pharmacology. 2013;75:45. doi: 10.1111/j.1365-2125.2012.04305.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ozen A Baris S Karakoc EA Yucelten D Nadir NB Experience with intravenous immunoglobulin in severe childhood atopic dermatitis. Allergologia et Immunopathologia. 2012;40:131. doi: 10.1016/j.aller.2011.04.007. [DOI] [PubMed] [Google Scholar]
- Hjuler KP Vestergaard C Deleuran M A retrospective study of six cases of severe recalcitrant atopic dermatitis treated with long-term extracorporeal photopheresis. Acta Dermato-Venereologica. 2010;90:635. doi: 10.2340/00015555-0952. [DOI] [PubMed] [Google Scholar]
- Akdis C Akdis M Bieber T Bindslev-Jensen C Boguniewicz ME Diagnosis and treatment of atopic dermatitis in children and adults: European Academy of Allergology and Clinical Immunology/American Academy of Allergy, Asthma and Immunology/PRACTALL Consensus Report. Journal of Allergy and Clinical Immunology. 2006;118:152. doi: 10.1016/j.jaci.2006.03.045. [DOI] [PubMed] [Google Scholar]
- Schäfer T The impact of allergy on atopic eczema from data from epidemiological studies. Current Opinion in Allergy and Clinical Immunology. 2008;228:418. doi: 10.1097/ACI.0b013e32830e71a7. [DOI] [PubMed] [Google Scholar]
- Burks AW Calderon MA Casale T Cox L Demoly P Update on allergy immunotherapy: American Academy of Allergy, Asthma & Immunology/European Academy of Allergy and Clinical Immunology/PRACTALL consensus report. Journal of Allergy and Clinical Immunology. 2013;131:1288. doi: 10.1016/j.jaci.2013.01.049. [DOI] [PubMed] [Google Scholar]
- Martignago I Incorvaia C Preventive actions of allergen immunotherapy: the facts and the effects in search of evidence. Clinical and Molecular Allergy. 2017;15:13. doi: 10.1186/s12948-017-0070-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vidal C del R o PR Tabar A Moreno C Allergen immunotherapy (AIT) in allergic rhinitis: long-term efficacy and the development of asthma. What do we know? . Current Treatment Options in Allergy. 2014;237:14. [Google Scholar]
- Boguniewicz M Biologic therapy for atopic dermatitis: moving beyond the practice parameter and guidelines. Journal of Allergy and Clinical Immunology in Practice. 2017;5:1477. doi: 10.1016/j.jaip.2017.08.031. [DOI] [PubMed] [Google Scholar]
- Schneider LC Baz Z Zarcone C Zurakowski D. Long-term therapy with recombinant interferon-gamma (rIFN-γ) for atopic dermatitis. Annals of Allergy, Asthma & Immunology. 1998;80:263. doi: 10.1016/S1081-1206(10)62968-7. [DOI] [PubMed] [Google Scholar]
- Stevens SR Hanifin JM Hamilton T Tofte SJ Cooper KD Long-term effectiveness and safety of recombinant human interferon gamma therapy for atopic dermatitis despite unchanged serum IgE levels. Archives of Dermatology. 1998;134:799. doi: 10.1001/archderm.134.7.799. [DOI] [PubMed] [Google Scholar]
- Presta LG Lahr S Shields R Porter J Gorman C Humanization of an antibody directed against IgE. Journal of Immunology. 1993;151:2623. [PubMed] [Google Scholar]
- Reddel HK Bateman ED Becker A Boulet L-P Cruz AA A summary of the new GINA strategy: a roadmap to asthma control. European Research Journal. 2015;46:622. doi: 10.1183/13993003.00853-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chipps BE Lanier B Milgrom H Deschildre A Hedlin G Omalizumab in children with uncontrolled allergic asthma: review of clinical trial and real-world experience. Journal of Allergy and Clinical Immunology. 2017;139:1431. doi: 10.1016/j.jaci.2017.03.002. [DOI] [PubMed] [Google Scholar]
- Lane JE Cheyney JM Lane TN Kent DE Cohen DJ Treatment of recalcitrant atopic dermatitis with omalizumab. Journal of the American Academy of Dermatology. 2006;54:68. doi: 10.1016/j.jaad.2005.09.030. [DOI] [PubMed] [Google Scholar]
- Belloni B Ziai M Lim A Lemercier B Sbornik M Low-dose anti-IgE therapy in patients with atopic eczema with high serum IgE levels. Journal of Allergy and Clinical Immunology. 2007;120:1223. doi: 10.1016/j.jaci.2007.08.060. [DOI] [PubMed] [Google Scholar]
- Amrol D Anti-immunoglobulin e in the treatment of refractory atopic dermatitis. Southern Medical Journal. 2010;103:554. doi: 10.1097/SMJ.0b013e3181de0cf6. [DOI] [PubMed] [Google Scholar]
- Ghaffari J Shahmohammadi S Ashrafi H Ranjbar AR Ghaffari N. Omalizumab (Xolair) in children above 12 years with chronic urticaria: a review of literature. Journal of Pediatrics Review. 2015;3:152. [Google Scholar]
- Wang H-H Li Efficacy of omalizumab in patients with atopic dermatitis: a systematic review and meta-analysis. Journal of Allergy and Clinical Immunology. 2016;138:1719. doi: 10.1016/j.jaci.2016.05.038. [DOI] [PubMed] [Google Scholar]
- Simon D sli S Kostylina G Yawalkar N Simon H Anti-CD20 (rituximab) treatment improves atopic eczema. Journal of Allergy and Clinical Immunology. 2008;246:122. doi: 10.1016/j.jaci.2007.11.016. [DOI] [PubMed] [Google Scholar]
- Sediva A Polouckova A Capkova S Anti-CD20 (rituximab) treatment for atopic eczema. Journal of Allergy and Clinical Immunology. 2008;121:122. doi: 10.1016/j.jaci.2008.03.007. [DOI] [PubMed] [Google Scholar]
- Hamilton JD rez-Farias M Dhingra N Cardinale I Li X Dupilumab improves the molecular signature in skin of patients with moderate-to-severe atopic dermatitis. Journal of Allergy and Clinical Immunology. 2014;225:1293. doi: 10.1016/j.jaci.2014.10.013. [DOI] [PubMed] [Google Scholar]
- Beck LA Thaçi D Hamilton JD Graham NM Bieber T Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. New England Journal of Medicine. 2014;231:130. doi: 10.1056/NEJMoa1314768. [DOI] [PubMed] [Google Scholar]
- Cork M i D DiCioccio T Davis JD Zhang Q Pharmacokinetics, safety, and efficacy of dupilumab in a pediatric population with moderate-to-severe atopic dermatitis: results from an open-label phase 2a trial. 2017;231:1. [Google Scholar]
- Cheng JF Ott NL Peterson EA George TJ Hukee MJ Dermal eosinophils in atopic dermatitis undergo cytolytic degeneration. Journal of Allergy and Clinical Immunology. 1997;99:683. doi: 10.1016/s0091-6749(97)70031-9. [DOI] [PubMed] [Google Scholar]
- Oldhoff J Darsow U Werfel T Katzer K Wulf A Anti-IL-5 recombinant humanized monoclonal antibody (mepolizumab) for the treatment of atopic dermatitis. Allergy. 2005;60:693. doi: 10.1111/j.1398-9995.2005.00791.x. [DOI] [PubMed] [Google Scholar]
- Nemoto O Furue M Nakagawa H Shiramoto M Hanada R 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. British Journal of Dermatology. 2016;174:296. doi: 10.1111/bjd.14207. [DOI] [PubMed] [Google Scholar]
- Ruzicka T Hanifin JM Furue M Pulka G Mlynarczyk I Anti–interleukin-31 receptor A antibody for atopic dermatitis. New England Journal of Medicine. 2017;8211:826. doi: 10.1056/NEJMoa1606490. [DOI] [PubMed] [Google Scholar]
- Teng MW Bowman EP McElwee JJ Smyth MJ Casanova J-L -12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nature Medicine. 2015;21:719. doi: 10.1038/nm.3895. [DOI] [PubMed] [Google Scholar]
- Khattri S Brunner PM Garcet S Finney R Cohen SR Efficacy and safety of ustekinumab treatment in adults with moderate-to-severe atopic dermatitis. Experimental Dermatology. 2017;26:28. doi: 10.1111/exd.13112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saeki H Kabashima K Tokura Y Murata Y Shiraishi A Efficacy and safety of ustekinumab in Japanese patients with severe atopic dermatitis: a randomized, double-blind, placebo-controlled, phase II study. British Journal of Dermatology. 2017;177:419. doi: 10.1111/bjd.15493. [DOI] [PubMed] [Google Scholar]
- Popovic B Breed J Rees D Gardener M Vinall L Structural characterisation reveals mechanism of IL-13-neutralising monoclonal antibody tralokinumab as inhibition of binding to IL-13Rα1 and IL-13Rα2. J Mol Biol. 2017;429:208. doi: 10.1016/j.jmb.2016.12.005. [DOI] [PubMed] [Google Scholar]
- Snast I Reiter O Hodak E Friedland R Mimouni D Are biologics efficacious in atopic dermatitis? A systematic review and meta-analysis. American Journal of Clinical Dermatology. 2018;19:145. doi: 10.1007/s40257-017-0324-7. [DOI] [PubMed] [Google Scholar]
- Patel N Strowd LC The future of atopic dermatitis treatment. Advances in Experimental Medicine and Biology. 2017;1027:185. doi: 10.1007/978-3-319-64804-0_15. [DOI] [PubMed] [Google Scholar]
- Ziegler SF Thymic stromal lymphopoietin and allergic disease. Journal of Allergy and Clinical Immunology. 2012;130:845. doi: 10.1016/j.jaci.2012.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma P Bian F Wang Z Zheng X Chotikavanich S Human corneal epithelium-derived thymic stromal lymphopoietin links the innate and adaptive immune responses via TLRs and Th2 cytokines. Investigative Ophthalmology & Visual Science. 2009;50:2702. doi: 10.1167/iovs.08-3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reche PA Soumelis V Gorman DM Clifford T Liu MR Human thymic stromal lymphopoietin preferentially stimulates myeloid cells. Journal of Immunology. 2001;167:336. doi: 10.4049/jimmunol.167.1.336. [DOI] [PubMed] [Google Scholar]
- Allakhverdi Z Comeau MR Jessup HK Yoon B-RP Brewer A Thymic stromal lymphopoietin is released by human epithelial cells in response to microbes, trauma, or inflammation and potently activates mast cells. Journal of Experimental Medicine. 2007;204:253. doi: 10.1084/jem.20062211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soumelis V Reche PA Kanzler H Yuan W Edward G Human epithelial cells trigger dendritic cell-mediated allergic inflammation by producing TSLP. Nature Immunology. 2002;3:673. doi: 10.1038/ni805. [DOI] [PubMed] [Google Scholar]
- Cheng DT Ma C Niewoehner J Dahl M Tsai A Thymic stromal lymphopoietin receptor blockade reduces allergic inflammation in a cynomolgus monkey model of asthma. Journal of Allergy and Clinical Immunology. 2013;132:455. doi: 10.1016/j.jaci.2013.05.011. [DOI] [PubMed] [Google Scholar]
- Ahluwalia J Udkoff J Waldman A Borok J Eichenfield LF Phosphodiesterase 4 inhibitor therapies for atopic dermatitis: progress and outlook. Drugs. 2017;77:1389. doi: 10.1007/s40265-017-0784-3. [DOI] [PubMed] [Google Scholar]
- Hanifin JM Chan SC Cheng JB Tofte SJ Henderson Jr Type 4 phosphodiesterase inhibitors have clinical and in vitro anti-inflammatory effects in atopic dermatitis. Journal of Investigative Dermatology. 1996;107:51. doi: 10.1111/1523-1747.ep12297888. [DOI] [PubMed] [Google Scholar]
- Margolis JS Abuabara K Bilker W Hoffstad O Margolis DJ Persistence of mild to moderate atopic dermatitis. JAMA Dermatology. 2014;150:593. doi: 10.1001/jamadermatol.2013.10271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drucker AM Wang AR Li W-Q Sevetson E Block JK The burden of atopic dermatitis: summary of a report for the National Eczema Association. Journal of Investigative Dermatology. 2017;137:26. doi: 10.1016/j.jid.2016.07.012. [DOI] [PubMed] [Google Scholar]
- Kiebert G Sorensen SV Revicki D Fagan SC Doyle JJ Atopic dermatitis is associated with a decrement in health-related quality of life. International Journal of Dermatology. 2002;41:151. doi: 10.1046/j.1365-4362.2002.01436.x. [DOI] [PubMed] [Google Scholar]
- Solomon CR Gagnon C Mother and child characteristics and involvement in dyads in which very young children have eczema. Journal of Developmental and Behavioral Pediatrics. 1987;8:213. [PubMed] [Google Scholar]
- Chamlin SL Frieden IJ Williams ML Chren M-M Effects of atopic dermatitis on young American children and their families. Pediatrics. 2004;114:607. doi: 10.1542/peds.2004-0374. [DOI] [PubMed] [Google Scholar]
- Ballardini N stblom E Wahlgren C-F Kull I. Mild eczema affects self-perceived health among pre-adolescent girls. Acta Dermato-Venereologica. 2014;94:312. doi: 10.2340/00015555-1738. [DOI] [PubMed] [Google Scholar]
- Ho RC Giam Y Ng T Mak A Goh D The influence of childhood atopic dermatitis on health of mothers, and its impact on Asian families. Pediatric Allergy and Immunology. 2010;21:501. doi: 10.1111/j.1399-3038.2009.00972.x. [DOI] [PubMed] [Google Scholar]
- Sánchez-Pérez J Daudén-Tello E Mora A Surinyac NL Impact of atopic dermatitis on health-related quality of life in Spanish children and adults: the PSEDA study. ACTAS Dermo-Sifiliograficas (English Edition) 2013;225:44. doi: 10.1016/j.ad.2012.03.008. [DOI] [PubMed] [Google Scholar]
- Su JC Kemp AS Varigos GA Nolan TM Atopic eczema: its impact on the family and financial cost. Archives of Disease in Childhood. 1997;76:159. doi: 10.1136/adc.76.2.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ring J Palos E Psychosomatic aspects of parent-child relations in atopic eczema in childhood. II. Child-rearing style, the family situation in a drawing test and structured interview. Hautarzt. 1986;37:609. [PubMed] [Google Scholar]
- Lapidus CS Kerr PE Social impact of atopic dermatitis. Medicine and Health/Rhode Island. 2001;84:294. [PubMed] [Google Scholar]
- Carroll CL Balkrishnan R Feldman SR Fleischer Jr AB Manuel JC The burden of atopic dermatitis: impact on the patient, family, and society. Pediatric Dermatology. 2005;22:192. doi: 10.1111/j.1525-1470.2005.22303.x. [DOI] [PubMed] [Google Scholar]
- Lee BW Detzel PR Treatment of childhood atopic dermatitis and economic burden of illness in Asia Pacific countries. Annals of Nutrition and Metabolism. 2015;66:18. doi: 10.1159/000370221. [DOI] [PubMed] [Google Scholar]
- Storan ER McEvoy MT Wetter DA el-Azhary RA Hand JL Pediatric hospital dermatology: experience with inpatient and consult services at the Mayo Clinic. Pediatric Dermatology. 2013;30:433. doi: 10.1111/pde.12081. [DOI] [PubMed] [Google Scholar]
- Bickers DR Lim HW Margolis D Weinstock MA Goodman C The burden of skin diseases: 2004: A joint project of the American Academy of Dermatology Association and the Society for Investigative Dermatology. Journal of the American Academy of Dermatology. 2006;55:490. doi: 10.1016/j.jaad.2006.05.048. [DOI] [PubMed] [Google Scholar]
- Patel NU D'Ambra V Feldman SR Increasing adherence with topical agents for atopic dermatitis. American Journal of Clinical Dermatology. 2017;8217:323. doi: 10.1007/s40257-017-0261-5. [DOI] [PubMed] [Google Scholar]
- Margolis DJ Gupta J Apter AJ Ganguly T Hoffstad O Filaggrin-2 variation is associated with more persistent atopic dermatitis in African-American subjects. Journal of Allergy and Clinical Immunology. 2014;133:784. doi: 10.1016/j.jaci.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lan CCE Tu HP Wu CS Ko YC Yu HS Distinct SPINK5 and IL-31 polymorphisms are associated with atopic eczema and non-atopic hand dermatitis in Taiwanese nursing population. Experimental Dermatology. 2011;20:975. doi: 10.1111/j.1600-0625.2011.01374.x. [DOI] [PubMed] [Google Scholar]
- Fortugno P Furio L Teson M Berretti M El Hachem M The 420K LEKTI variant alters LEKTI proteolytic activation and results in protease deregulation: implications for atopic dermatitis. Human Molecular Genetics. 2012;21:4187. doi: 10.1093/hmg/dds243. [DOI] [PubMed] [Google Scholar]
- Yu H-S Kang Kwon J-W Lee S-Y Lee E Claudin-1 polymorphism modifies the effect of mold exposure on the development of atopic dermatitis and production of IgE. Journal of Allergy and Clinical Immunology. 2015;135:827. doi: 10.1016/j.jaci.2014.10.040. [DOI] [PubMed] [Google Scholar]
- Ismaïl A Bousaffara R Kaziza J Zili J El Kamel A Sfar MT Polymorphism in transporter antigen peptides gene (TAP1) associated with atopy in Tunisians. Journal of Allergy and Clinical Immunology. 1997;239:216. doi: 10.1016/s0091-6749(97)70099-x. [DOI] [PubMed] [Google Scholar]
- Braff MH Di Nardo A Gallo RL Keratinocytes store the antimicrobial peptide cathelicidin in lamellar bodies. Journal of Investigative Dermatology. 2005;124:394. doi: 10.1111/j.0022-202X.2004.23443.x. [DOI] [PubMed] [Google Scholar]
- Levchenko L lova O Shlykova O Kaĭdashev I. Polymorphism 896A/G of TLR4 gene rather than 1196C/T and 2258G/A of TLR2 gene determines severe and complicated course of atopic dermatitis in children. Tsitologiia i genetika. 2013;301:46. [PubMed] [Google Scholar]
- Potaczek DP Nastalek M Okumura K Wojas-Pelc A Undas A An association of TLR2-16934A> T polymorphism and severity/phenotype of atopic dermatitis. Journal of the European Academy of Dermatology and Venereology. 2011;25:715. doi: 10.1111/j.1468-3083.2010.03812.x. [DOI] [PubMed] [Google Scholar]
- Oh DY Schumann R Hamann L Neumann K Worm M Association of the toll-like receptor 2 A-16934T promoter polymorphism with severe atopic dermatitis. Allergy. 2009;64:1608. doi: 10.1111/j.1398-9995.2009.02066.x. [DOI] [PubMed] [Google Scholar]
- Salpietro C Rigoli L Del Giudice MM Cuppari C Di Bella C et al. International Journal of Immunopathology and Pharmacology. 2011;24:33. doi: 10.1177/03946320110240S408. [DOI] [PubMed] [Google Scholar]
- Weidinger S R mmler L Klopp N Wagenpfeil S Baurecht H Association study of mast cell chymase polymorphisms with atopy. Allergy. 2005;252:1256. doi: 10.1111/j.1398-9995.2005.00879.x. [DOI] [PubMed] [Google Scholar]
- Margolis DJ Kim B Apter AJ Gupta J Hoffstad O Thymic stromal lymphopoietin variation, filaggrin loss of function, and the persistence of atopic dermatitis. JAMA Dermatology. 2014;150:254. doi: 10.1001/jamadermatol.2013.7954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma CA Stinson JR Zhang Y Abbott JK Weinreich MA Germline hypomorphic CARD11 mutations in severe atopic disease. Nature Genetics. 2017;49:1192. doi: 10.1038/ng.3898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoi LC Spain SL Knight J Ellinghaus E Stuart PE Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nature Genetics. 2012;44:1341. doi: 10.1038/ng.2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kabesch M Peters W Carr D Leupold W Weiland SK Association between polymorphisms in caspase recruitment domain containing protein 15 and allergy in two German populations. Journal of Allergy and Clinical Immunology. 2003;111:813. doi: 10.1067/mai.2003.1336. [DOI] [PubMed] [Google Scholar]
- Gao P-S Rafaels Mu D Hand T Murray T Genetic variants in thymic stromal lymphopoietin are associated with atopic dermatitis and eczema herpeticum. Journal of Allergy and Clinical Immunology. 2010;12:1403. doi: 10.1016/j.jaci.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim E Lee JE Namkung JH Kim PS Kim S Single nucleotide polymorphisms and the haplotype in the DEFB1 gene are associated with atopic dermatitis in a Korean population. Journal of Dermatological Science. 2009;54:25. doi: 10.1016/j.jdermsci.2008.12.005. [DOI] [PubMed] [Google Scholar]
- Zentsova-Jaresova I Katina S Vernerova E A prospective study in children with a severe form of atopic dermatitis: clinical outcome in relation to cytokine gene polymorphisms. Journal of Investigational Allergology and Clinical Immunology. 2012;22:92. [PubMed] [Google Scholar]
- Walley AJ Chavanas S Moffatt MF Esnouf RM Ubhi B Gene polymorphism in Netherton and common atopic disease. Nature Genetics. 2001;29:175. doi: 10.1038/ng728. [DOI] [PubMed] [Google Scholar]
- Chang Y Lee W Yu C Liu H Lin M No association of cytokine gene polymorphisms in Chinese patients with atopic dermatitis. Clinical and Experimental Dermatology. 2006;31:419. doi: 10.1111/j.1365-2230.2006.02124.x. [DOI] [PubMed] [Google Scholar]
- Sokołowska-Wojdyło M The frequencies of haplotypes J Zabłotna M Rębała K Trzeciak M The frequencies of haplotypes defined by three polymorphisms of the IL. International Journal of Dermatology. 2015;322:1066–1066. doi: 10.1111/ijd.12666. [DOI] [PubMed] [Google Scholar]
- Narbutt J Wojtczak M Zalińska A Salinski A Przybylowska-Sygut K The A/A genotype of an interleukin-17A polymorphism predisposes to increased severity of atopic dermatitis and coexistence with asthma. Clinical and Experimental Dermatology. 2015;324:11. doi: 10.1111/ced.12438. [DOI] [PubMed] [Google Scholar]
- Namkung J-H Lee J-E Kim E Park GT Yang HS An association between IL-9 and IL-9 receptor gene polymorphisms and atopic dermatitis in a Korean population. Journal of Dermatological Science. 2011;62:16. doi: 10.1016/j.jdermsci.2011.01.007. [DOI] [PubMed] [Google Scholar]
- Novak N Kruse S Potreck J Maintz L Jenneck C Single nucleotide polymorphisms of the IL18 gene are associated with atopic eczema. Journal of Allergy and Clinical Immunology. 2005;115:828. doi: 10.1016/j.jaci.2005.01.030. [DOI] [PubMed] [Google Scholar]
- Rafatpanah H Bennett E Pravica V McCoy MJ David TJ Association between novel GM-CSF gene polymorphisms and the frequency and severity of atopic dermatitis. Journal of Allergy and Clinical Immunology. 2003;112:593. doi: 10.1016/s0091-6749(03)01797-4. [DOI] [PubMed] [Google Scholar]
- Nickel RG Casolaro V Wahn U Beyer K Barnes KC Atopic dermatitis is associated with a functional mutation in the promoter of the CC chemokine RANTES. Journal of Immunology. 2000;164:1612. doi: 10.4049/jimmunol.164.3.1612. [DOI] [PubMed] [Google Scholar]
- Tanaka K Roberts M Yamamoto N Sugiura H Uehara M Upregulating promoter polymorphisms of RANTES relate to atopic dermatitis. International Journal of Immunogenetics. 2006;33:423. doi: 10.1111/j.1744-313X.2006.00635.x. [DOI] [PubMed] [Google Scholar]
- Tsunemi Y Saeki H Nakamura K Sekiya T Hirai K Eotaxin gene single nucleotide polymorphisms in the promoter and exon regions are not associated with susceptibility to atopic dermatitis, but two of them in the promoter region are associated with serum IgE levels in patients with atopic dermatitis. Journal of Dermatological Science. 2002;29:222. doi: 10.1016/s0923-1811(02)00038-5. [DOI] [PubMed] [Google Scholar]
- Gangur V Oppenheim JJ Are chemokines essential or secondary participants in allergic responses? Annals of Allergy, Asthma & Immunology. 2000;84:569. doi: 10.1016/S1081-1206(10)62403-9. [DOI] [PubMed] [Google Scholar]
- Hallau J Hamann L Schumann RR Worm M Heine G A promoter polymorphism of the vitamin D metabolism gene Cyp24a1 is associated with severe atopic dermatitis in adults. Acta Dermato-Venereologica. 2016;96:169. doi: 10.2340/00015555-2226. [DOI] [PubMed] [Google Scholar]
- Yang K Liu CA Chang JC Chuang H Ou CY Polymorphism of the immune-braking gene CTLA-4 (+ 49) involved in gender discrepancy of serum total IgE levels and allergic diseases. Clinical & Experimental Allergy. 2004;34:32. doi: 10.1111/j.1365-2222.2004.01776.x. [DOI] [PubMed] [Google Scholar]
- Nishio Y Noguchi E Ito S Ichikawa E Umebayashi Y Mutation and association analysis of the interferon regulatory factor 2 gene (IRF2) with atopic dermatitis. Journal of Human Genetics. 2001;46:664. doi: 10.1007/s100380170018. [DOI] [PubMed] [Google Scholar]
- Howell M Parker M Mustelin T Ranade K. Past future for biologic intervention in atopic dermatitis. Allergy. 2015;70:887. doi: 10.1111/all.12632. [DOI] [PubMed] [Google Scholar]
- Darsow U Wollenberg A Simon D Taïeb A Werfel T Difficult to control atopic dermatitis. World Allergy Organization Journal. 2013;239:1. doi: 10.1186/1939-4551-6-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bell DC Brown SJ Atopic eczema treatment now and in the future: Targeting the skin barrier and key immune mechanisms in human skin. World Journal of Dermatology. 2017;6:42. [Google Scholar]