Atopic dermatitis is a chronic inflammatory skin disorder affecting children and adults, with the majority presenting mild to moderate disease severity. The use of topical corticosteroids (TCSs) in combination with emollients has been the mainstay for treating mild to moderate atopic dermatitis since the 1950s, and as a supplement to systemic treatment in severe disease. However, while very effective, TCSs are often accompanied by poor adherence due to corticophobia (fear of using corticosteroids in patients or doctors), unwanted side effects, and in some cases insufficient clinical response. Topical calcineurin inhibitors (TCIs) are able to inhibit the activation of T-lymphocytes and thereby diminish inflammation. In some patients the use of TCIs has been limited due to a localized burning sensation on the first days of treatment, and also due to fear of other adverse effects. Consequently, there has been a need for the development of new topical products for atopic dermatitis. Novel topical therapies are in the pipeline and comprise both new doses and formulations of well-known pharmaceutical molecules and novel approaches targeting unique inflammatory pathways and mechanisms of disease, with a promise of higher efficacy and less harmful side effects. We review topical drugs in the pipeline for atopic dermatitis, and focus on those available in the clinicaltrials.gov database with a first received date from January 1, 2014 to May 31, 2017.

Atopic dermatitis (AD) is chronic inflammatory dermatological disease, with characteristics including pruritus, dry skin, and chronic or chronically relapsing dermatitis. It is the most common inflammatory skin disease with 10-25% of children and 2-10% of adults in affluent nations being affected. The socioeconomic impact on the patient, the family, and the society as a whole is substantial [1,2,3]. Within the AD population, severity scoring suggests that more than 70% of patients have mild disease that is dealt with in primary care. Approximately 20% have moderate and 5-10% severe AD, commonly requiring referral to a dermatologist [4]. The complexity of AD is demonstrated by a complex pathogenesis comprising a dysregulated immune system, a compromised skin barrier, and a range of trigger factors [5]. There are new risk modifiers and associated comorbidities constantly being identified for this disease, which gives rise to a need for early intervention and new disease modifying strategies [6,7,8,9,10,11,12,13].

Various treatment approaches exist for alleviating the disease, including hydration and restoration of the skin barrier with the use of emollients, avoidance of typical AD triggers, specific behavioural approaches to reduce scratching, antibacterial measures, and topical and/or systemic anti-inflammatory drugs [14,15]. During a 10-year period in the USA, 7.4 million visits of children younger than 18 years were registered at physicians' offices for the diagnosis of AD. Topical corticosteroids (TCSs) were prescribed in 25-34% and calcineurin inhibitors in 23% of visits [16]. In mild disease the regular use of emollients and either TCSs or calcineurin inhibitors are recommended [15,17]. These agents are in most situations individually or in combination able to bring relief to the patient; they are, however, rarely sufficient in more recalcitrant cases. The therapeutic approach to AD is highly dependent on several factors, e.g., patient insight into their disease, adherence, trigger factors, and psychosocial disturbances, but perhaps the most important factors when choosing a specific drug and way of administration are disease severity and any registered efficacy of previous treatments [18]. Thus, for the purpose of this review of emerging topical treatments, we focus on AD patients commonly alleviated by established topical therapies and emollients [14,15,17].

Topical Therapies

The treatment goal is to diminish pruritus, supress inflammation, and restore skin barrier function. In mild to moderate AD the use of topical therapies is advantageous in comparison with systemic treatments as they generally perform well with a minimum risk of adverse events. The most commonly used topical therapies are emollients, TCSs, and topical calcineurin inhibitors (TCIs).

Emollients or moisturisers are widely recommended, as they are effective and safe. They reduce transepidermal water loss by acting as an occlusive layer on the skin [18,19]. Moreover, advanced moisturisers are able to bind or attract water in the stratum corneum, actively hydrating the epidermal barrier [20,21], and can reduce the severity of disease and prolong the interval between flares [21]. The addition of moisturisers to topical anti-inflammatory treatment has been shown to be more effective than anti-inflammatory treatment alone and has reduced the amount of TCSs used [20,21].

For more than half a century the use of TCSs has been the backbone of AD treatment, and they are recommended by European, American, and Japanese Academies of Dermatology [17,22,23]. TCSs have anti-inflammatory effects and reduce skin inflammation and pruritus. In most cases, TCSs used correctly bring swift relief to the patient. Due to the chronic and chronically relapsing nature of AD there can be a lifelong need for treatment, with a significant risk of adverse events. These include skin atrophy, and to a lesser extent striae and telangiectasias, while the risk of a rebound phenomenon when tapering TCSs is exaggerated if used appropriately [24,25]. The risk of systemic side effects is low and correlated to the potency of the TCS, the extent and degree of skin inflammation, and the age of the patient. Infants and children have a higher degree of systemic absorption due to a less mature skin barrier [24]. The biggest challenge faced when prescribing TCSs is patient fear of TCS-related adverse events (corticophobia). In conclusion, TCSs are recommended if AD is not sufficiently controlled by emollient therapy alone; it is the mainstay anti-inflammatory treatment, and proactive use is recommended in skin areas that commonly flare [17,22].

The use of TCIs was introduced in 2000 as an alternative to TCS. The selective inhibition of T-cell activation decreases the activity of a range of transcriptional factors that control cell division and reduces the expression of pro-inflammatory cytokines which ultimately diminish inflammation. Pimecrolimus cream and a high and low concentration formulation of tacrolimus ointment are currently approved for AD in both children over 2 years of age and adults. Their widespread use is facilitated by their inability to produce skin atrophy, and despite a black box warning from the FDA in 2005 concerning increased risk of malignancy, no solid evidence has been published demonstrating such a causal relationship. TCIs are primarily used for delicate skin areas unresponsive to low-potency TCSs, in AD cases recalcitrant to steroids, and in patients with a need for continuous long-term topical anti-inflammatory therapy [17,22]. TCIs can also be used as intermittent prophylactic therapy equivalent to TCSs [26].

A systematic review and meta-analysis of TCSs versus TCIs concluded that calcineurin inhibitors and corticosteroids have comparable efficacy. However, TCIs were associated with higher costs and have more adverse events, such as skin burning and pruritus. The results provided a level-1a support for the use of corticosteroids as the first anti-inflammatory therapy of choice for AD [27].

Immunopathology of AD

The pathophysiology of AD involves genetic predispositions, skin barrier dysfunction, increased activity, and Th2/Th22 skewing of the immune system and the implication of environmental trigger factors. One possible hypothesis of the mechanisms underlying the disease is that reduced function of the skin barrier results in increased transepidermal water loss, xerosis, and subsequent penetration of allergens, pathogens, and irritants, altogether activating and modulating the immune system and producing dermatitis (outside-in hypothesis). A second approach is that of innate and adaptive immune imbalances giving rise to an abnormal T-cell response, elevated IgE levels, and increased type 2 cytokine production, consequently leading to reduced skin barrier function, xerosis, and dermatitis (inside-out hypothesis). Whether one or the other is the primary key pathophysiological mechanism, the bottom line is a state of increased immune activity and reduced skin barrier integrity. Thus, by addressing the first, the latter or both is meaningful [11,28,29,30,31,32,33]. Targeting key mediators of disease, either via narrow isolated effects or broader downstream actions, are potential means to alleviate the disease.

Selecting Studies for This Paper

Papers on emerging topical therapies comprise the body of this review in AD combined with data on current and completed clinical trials concerning new topical treatment options for AD identified at clinicaltrials.gov. We selected AD-related therapies that have been reported from randomized, controlled trials in phases II, III, and IV with a first received date from January 1, 2014 to May 31, 2017. If one agent was represented in more than one trial in the clinicaltrials.gov database, it is described in the review and tables in only one study (Table 1). We focused on the emerging drugs which are developed based on reasonable evidence of a potential effect in AD. In doing so, more speculative therapeutic approaches and alternative means of disease modification received a lower priority. Lastly, we excluded therapies that could be categorized as traditional Chinese medicine, herbal medicine, probiotics, emollients, TCSs, or TCIs.

Table 1

Details of the studies included in the review

Details of the studies included in the review
Details of the studies included in the review

Targeting Janus Kinase/Signal Transducer and Activator of Transcription Pathways

The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is utilized by numerous cytokines and growth factors for signal transduction in AD, involving augmentation of Th2 cell response, activation of eosinophils, and suppression of regulatory T cells [34]. Moreover, it downregulates the expression of structural skin proteins, and weakens the epidermal barrier function [35,36,37,38,39]. Additionally, downstream signalling in this pathway has been shown to prevent the induction of genes encoding innate immune response proteins, including β-defensins and cathelicidin in keratinocytes [40], thus raising the vulnerability of patients to both viral and bacterial skin infections. Inhibitors of the JAK/STAT signalling axis are categorized as small molecules blocking intracellular targets in comparison with anticytokine/antireceptor agents. The relevance in targeting this family of kinases is that they constitute the main signalling pathway for several cytokines, thus providing an opportunity to prevent the downstream signalling of numerous AD-typical Th2 cytokines. Several pharmaceutical agents targeting this group of tyrosine kinases (comprising TYK2, JAK1, JAK2, and JAK3) are being evaluated in patients with AD both as systemic and topical therapies. The small size of the JAK inhibitors makes them suitable for topical use, and this is in part substantiated by a recent study demonstrating great antipruritic and anti-inflammatory effects from topically administered tofacitinib and oclacitinib in a mouse model of allergic dermatitis [41].

Tofacitinib

Tofacitinib (NCT02001181) is approved for rheumatoid arthritis and has previously been investigated in a range of dermatological conditions like psoriasis, alopecia areata, and systemic lupus erythematosus. It inhibits JAK1 and JAK3 and in theory interferes to a larger extent with lymphocyte activation and Th2 skewing than the inhibitors also targeting JAK2, which are more involved in Th1 signalling [42]. Moreover, tofacitinib has been shown to directly inhibit cytokines, most importantly IL-4, thereby rapidly attenuating JAK-STAT signalling in keratinocytes [43,44]. While the results in psoriasis trials have shown quite varying results [45,46], the single published trial on topically administered tofacitinib in adults with mild to moderate AD revealed significantly better efficacy. Most importantly, for the primary analysis at week 4, the mean percentage change from baseline in the Eczema Area and Severity Index (EASI) total score was significantly greater (p < 0.001) for patients treated with tofacitinib (81.7%) versus patients treated with vehicle (29.9%). Topical tofacitinib also displayed an early onset of effect, a comparable safety profile, and local tolerability to vehicle, with the most common adverse events being mild self-limiting infections (e.g., nasopharyngitis) and application site pain and pruritus [47]. In conclusion, it seems that JAK1/JAK3 inhibition, through topical delivery, is possibly a capable treatment target for AD, though due to the nature of the trial, additional studies are warranted to address the use for long-term control.

Ruxolitinib

Ruxolitinib (INCB018424), a JAK1/JAK2 inhibitor, is approved for the treatment of adult patients with polycythemia vera and for the treatment of disease-related splenomegaly or symptoms in patients with myelofibrosis. Moreover, use of ruxolitinib in the field of dermatology is emerging, as it has already been trialled in a number of diseases e.g., psoriasis (only topical), alopecia areata (topical and oral), vitiligo (only topical), and graft-versus-host disease [48].

A phase II placebo-controlled study to evaluate the safety and efficacy in adult AD patients is planned (NCT03011892). The participants are to be randomized between the ruxolitinib cream once or twice daily compared with vehicle cream twice daily. With an estimated enrolment of 300 participants, the study would provide well-powered data on the possible benefits of topical ruxolitinib in AD.

The potential therapeutic use of phosphodiesterase-4 (PDE4) inhibitors in AD is based on the recognized intracellular role of PDE4 in keratinocytes [49,50]. Circulating leukocytes in AD patients have PDE4 activity, which has been associated with higher production of proinflammatory mediators and lower production of the anti-inflammatory mediator IL-10, in part due to hydrolization of cyclic adenosine monophosphate (cAMP) [51,52,53]. This consequently diminishes levels of cAMP, which leads to increased transcription of numerous cytokines, accelerating a number of intracellular functions involved in acute and chronic inflammation [51]. Thus, targeting PDE4 has been shown to directly attenuate inflammation due to inhibition of the breakdown of cAMP, consequently reducing the levels of tumour necrosis factor-α, IL-12, IL-23, and other signalling effectors [54,55]. Of note is that a number of topical PDE4 inhibitors have been investigated previously, but despite completion of these trials several years ago no results have been presented and the drugs are no longer in clinical trial, e.g., DRM02 (NCT0199342, completed in 2014) and LEO 29102 (NCT01037881, NCT00958516, NCT01005823, NCT01447758, NCT00891709, and NCT01423656, the last one completed in 2011). To lessen systemic exposure, a topical PDE4 inhibitor with little transdermal bioavailability might be clinically advantageous, as systemic treatment with agents from this drug class have so far been compromised by a significant rate of mechanism-associated adverse reactions, most prominently gastrointestinal discomfort and headache. Thus, it is promising that the low molecular weight of PDE4 inhibitors ensures excellent skin penetration, and safety studies have shown minimal systemic uptake. Currently, there are several trials investigating 5 different topically administered PDE4 inhibitors.

Crisaborole

The boron-based benzoxaborole named crisaborole (AN2728) is a small molecule specifically inhibiting PDE4 activity (NCT02118792) and the first in its class to be approved by the FDA. Seven studies have been conducted on the topical formulation of the compound to date, and two of them have reached phase III (NCT02118792 and NCT02118766). Data on the previous phase I and II studies showed positive results across a accumulated cohort of 189 subjects with AD as young as 2 years of age [54,56,57,58,59]. The first study conducted assessed the safety profile and pharmacokinetics in both children and adolescents under conditions with a supposed maximal use of topical crisaborole [56]. The drug showed swift absorption and minimal systemic exposure. However, circulating levels increased in parallel with the extent of the skin area treated, yet displayed no correlation with the incidence of adverse events.

Crisaborole had an acceptable safety profile, but a high percentage of participants experienced local self-limiting and mild application site adverse events. Both the pruritus score (after 5 days) and signs and symptoms (after 4 weeks) significantly improved with treatment. However, as application site adverse events were present in the majority of patients in the treatment group across all trials, varying drop-out rates directly linked to this have been reported. In two of the phase II studies both pruritus and individual scores of signs and symptoms were positively affected by crisaborole treatment in comparison with placebo vehicle, though all studies were underpowered and did not report data on the most common disease severity scores e.g., EASI and Scoring Atopic Dermatitis (SCORAD). Therefore, these conclusions on efficacy should be cautiously interpreted. In contrast, the two phase III trials completed in 2015 were published in late 2016 and evaluated in parallel [60]. The two identically designed, vehicle-controlled, double-blind studies enrolled and randomly assigned (2:1, crisaborole:vehicle) patients aged 2 years or older with an Investigator's Static Global Assessment (ISGA) score of mild or moderate for twice-daily application for 28 days. The primary end point of the ISGA score at day 29 of clear (0)/almost clear (1) with 2-grade or greater improvement from baseline was achieved in a greater percentage of crisaborole-treated patients than placebo-treated subjects. Time to success in the ISGA score was significantly shorter in the crisaborole-treated group than in those treated with vehicle. Lastly, the individual score tool (signs of AD scale) revealed a significant reduction in the severity of AD signs, and the twice-daily pruritus severity scoring showed a swift (significant after 8 days) and sustainable improvement in pruritus [60]. However, to summarize the bulk of data on topically administered crisaborole, the difference of approximately 10% in both the ISGA success rate and the improvement score of pruritus between crisaborole- and placebo-treated subjects is modest, and the minimal clinically important difference (MCID) was not defined for the reported outcome measure. Therefore, it is difficult to thoroughly assess the impact of treatment on the key domain “signs of disease” as communal severity scores were not included.

E6005

E6005 (RVT-501) is a selective PDE4 inhibitor and has shown antipruritic abilities in mouse models mimicking AD [61,62,63]. E6005 is able to suppress C-fibre depolarization and activation of the dorsal root ganglion through elevation of cAMP levels, thereby exerting an antipruritic effect [62]. An early phase I/II study evaluating safety, tolerability, and pharmacokinetics showed no application site adverse event related to treatment and systemic absorption was below the detection limit [64]. The following phase II trial of 78 adults with eczema on 5-30% of the skin showed a reasonable safety profile, with a slight increase in incidence of AD exacerbation in the E6005-treated group compared with placebo (13.5 vs. 7.7%). At the end of week 4, EASI, objective SCORAD, visual analogue scales for pruritus and sleep loss, and the severity of the targeted eczematous lesions in the topical E6005 group showed insignificant but trending improvement compared with those in the vehicle group [52]. The trial last to be completed, E6005 (NCT02094235), included a fairly low number of adult Japanese males with mild to moderate AD (n = 40) randomized 1:1:1:1:1 to either 1 of 4 doses of twice-daily E6005 or placebo vehicle [65]. The study, possibly due to being severely underpowered, was not able to produce many significant end point measures of efficacy between the E6005- and vehicle-treated subjects. Of note, however, were a few significant changes on days 5 and 11 in the SCORAD and EASI scores that lay above the defined MCID threshold for these severity measures. In summary, the highest potency formulation of E6005 seemed to have the best efficacy, though results should be interpreted with caution, as the total enrolment numbers across studies are still low. An active phase II study with an expected number of 150 participants is in the pipeline (NCT02950922), but enrolment has yet to commence.

OPA-15406

OPA-15406, another specific PDE4 inhibitor, has solely been investigated as topical treatment of AD (NCT02068352). With two phase I and one phase II trials already completed and three ongoing phase II trials (two recruiting and one active but not yet recruiting) there is extensive activity related to the drug. Published data comprise a single paper communicating the results from the completed randomized, double-blind, vehicle-controlled, phase II study of 121 patients aged 10-70 years with mild to moderate AD who received either 1 or 0.3% doses of topical OPA-15406 or vehicle twice daily for 8 weeks (randomized 1:1:1) [66]. The primary end point, a score of 0 or 1 in the Investigator's Global Assessment (IGA) scale with a greater than or equal to 2-step reduction, was seen at week 4 in the OPA-15406 1% group (p = 0.0165 vs. vehicle). Secondly, the study showed a significant mean improvement from baseline EASI score for OPA-15406 1% in week 1 (31.4 vs. 6.0% for vehicle; p = 0.0005), which was even greater in week 2, persisting for the duration of the study. The pruritus scores improved significantly within the first week in the OPA-15406 1% group (p = 0.0011), though they declined during the study and were no longer significant at week 6. Circulating OPA-15406 levels were insignificant, and the safety profile was good as the rate of adverse events was low, with most events considered mild and transient. In conclusion, the PDE4 inhibitor show somewhat promising results; however, the study size is a considerable limitation and larger studies are needed.

Roflumilast

Roflumilast is a selective, long-acting inhibitor of PDE4. It has previously been used as systemic therapy but in the context of AD solely as topical treatment. The primary proof-of-concept trial, designed to assess the safety and efficacy of 0.5% dermal roflumilast cream, was withdrawn prior to enrolment. A subsequent phase II study evaluating the effect of topical roflumilast on the reduction of AD lesions in 40 adults with moderate AD randomized 1:1 to either topical roflumilast 0.5% cream twice daily or a parallel regimen with vehicle has been completed, with results available at clinicaltrials.gov. The primary outcome measures were change from baseline to day 15 in modified local SCORAD, transepidermal water loss, and participants' assessment of pruritus. The study showed significant results only in the mean difference between day 15 pruritus score in the roflumilast-treated group compared with vehicle (difference,1.56, p = 0.013). However, this difference between roflumilast and placebo did not meet the proposed MCID for pruritus reduction measured on a visual analogue scale [67]. In summary, roflumilast showed little to no potential as topical PDE4 selective therapy in AD. However, to settle the matter completely, larger trials are to be conducted.

DRM02

DRM02 is a new PDE4 inhibitor that was investigated in three single studies for psoriasis, rosacea, and AD (NCT01993420). The double-blind, randomized, within-subject control, phase II study enrolling 21 adult subjects with stable moderate AD was designed to assess the safety, tolerability, and preliminary efficacy of DRM02. Despite all three trials being completed in 2014, no results have been communicated.

Benvitimod

Benvitimod (2-isopropyl-5-[(E)-2-phenylethenyl]-benzene-1,3-diol) (GSK-2894512, WBI-1001) is a non-steroidal, anti-inflammatory small molecule that was originally derived from the metabolites of nematodes. Benvitimod holds properties enabling a reduced expression of several proinflammatory cytokines (e.g., IFN-γ, IL-2, and TNF-α) and the inhibition of T-cell viability and infiltration, ultimately diminishing skin inflammation [68,69,70,71]. Two phase II trials have been completed and published on the safety and efficacy of topical benvitimod treatment. The first trial (NCT00837551) of 37 men and women with a baseline EASI <12 was designed with randomized (1:1:1) application twice daily of benvitimod 0.5%, benvitimod 1.0%, or vehicle for 4 weeks. Results showed that both of the benvitimod concentrations were well tolerated [71]. Both 0.5 and 1.0% benvitimod were superior to vehicle in improving AD at week 4 determined by a statistically significant reduction in the EASI, SCORAD, IGA, and body surface area (BSA) scores. The second trial (NCT01098734) was set to further test the safety and efficacy of benvitimod as a topical treatment studied over a 12-week period in 148 patients with mild to severe AD (randomized 1:1:1 to placebo, benvitimod 0.5%, or benvitimod 1.0% applied twice daily for 6 weeks). At the end of this phase, patients receiving benvitimod continued the same treatment for an additional 6 weeks. Patients receiving placebo entered into a 6-week double-blind phase with re-randomization (1:1) to benvitimod (0.5 or 1.0%). The study showed that there was a decrease of 1.3 (p < 0.001) and 1.8 (p < 0.001) in IGA at day 42 in the benvitimod 0.5 and 1.0% groups, respectively, compared with a decrease of 0.5 in the placebo group [69]. The EASI, SCORAD, BSA, and pruritus scores were significantly improved with each active treatment compared with placebo on day 42, demonstrating a swift elimination of these differences (on days 56 and 84) when the subjects initially treated with placebo switched to either 0.5 or 1.0% benvitimod treatment. Adverse events encompassed limited cases of folliculitis, contact dermatitis, and headache. A completed but not published trial (NCT02564055) that enrolled 247 AD patients could confirm the existing data and perhaps endorse benvitimod as a novel topical treatment in AD.

Less-Investigated Therapies

DGLA (dihomo-γ-linolenic acid) is a 20-carbon ω−6 fatty acid that has been investigated in mice models of AD and as both oral (NCT02211417) and topical treatment (NCT02925793) of AD for a decade or more [72,73,74]. Somewhat positive results have been observed in both of the administered formulations of the drug as communicated in press releases [75,76]. However, no crude data or scientific papers have emerged. There are ongoing studies of topical and systemic DGLA treatment with DS107, both enrolling approximately 300 AD patients.

The transient receptor potential vanilloid type 1 (TRPV1) is a cation channel activated by various stimuli like pH changes or heat. Several studies have shown that TRPV1 could be deeply associated with skin permeability barrier function, and is a likely mediator of chronic pruritus, as TRPV1 antagonists have shown positive effects in animal models of AD [77,78,79,80,81,82]. Results from previous phase I and II studies have not yet been published, and ongoing phase II and III studies (NCT02748993 + NCT02965118) of the topical TRPV1 antagonist PAC-14028 are expected to shed better light on the efficacy and safety of this therapy.

The isoprenylcysteine analogue DMT210 is a topical therapy designed to mimic the amino acid tail found at the C-terminus of G proteins, and is thus supposed to downregulate the inflammatory response via the G-protein-coupled receptor [83]. It is being trialled in various dermatological conditions, including AD (NCT02949960), but further research is needed to illuminate the properties of the drug.

Another investigational topically applied agent, SB011, contains the DNAzyme hgd40 that targets GATA-3, a key regulatory factor of Th2-driven immune responses. Hgd40 was initially designed for the treatment of allergic bronchial asthma [84] by cleaving GATA-3 mRNA capable of mitigating cytokine production, consequently reducing key features of atopic inflammation [85]. The now completed primary proof-of-concept study addresses the efficacy, safety, tolerability, pharmacokinetics, and pharmacodynamics of the topical formulation of SB011 (containing 2% hgd40) twice daily in 25 AD patients with mild to moderate disease (NCT02079688). Results are pending.

VTP-38543 is a liver X receptor (LXR) agonist. The LXR agonists have been study for years in the context of various cancers, neurodegenerative diseases, and liver disorders, as they are imperative regulators of cholesterol, fatty acids, and glucose homeostasis [86,87,88]. LXR agonists may intervene with AD typical skin inflammation [89,90]. A completed, but not yet published, phase I/II proof-of-concept study in a total of 104 adult patients with mild to moderate AD might give preliminary insights into this possible treatment modality (NCT02655679). Importantly, a press release deemed the treatment a “flop” as it failed to show a positive signal in the study [91], and it is therefore unlikely that the results will ever be issued.

Cytosolic phospholipase A2 (cPLA2) is the rate-limiting enzyme responsible for the release of arachidonic acid and the succeeding production of a range of inflammatory lipid mediators (e.g., leukotrienes, prostaglandins, and thromboxanes). ZPL-5212372 is a selective cPLA2 inhibitor that apparently exhibited long-term action and good efficacy in small and large animal models of airway and skin inflammation [92]. A dermal formulation of ZPL-5212372 has been developed to support an ongoing phase I/IIa study in moderate to severe AD patients (NCT02795832).

MRX-6, a somewhat similar drug to the aforementioned, is another non-steroidal anti-inflammatory cream working through the inhibition of secreted soluble PLA2 and via enriching cell surface glycosaminoglycans. The supposed mechanism of effect is similar to other PLA2 inhibitors [93]. The first study of the drug for allergic contact dermatitis (NCT00867607) was completed in 2015 and results were communicated as positive. However, the sole phase II study in AD patients (NCT02031445) was terminated as interim analysis showed lack of efficacy, hence the likelihood of further investigations is low.

IL-31 is a key mediator of acute and chronic itch in AD patients, and mitigating this cytokine might be beneficial in AD [30,79,94,95]. The 5-lipoxygenase inhibitor zileuton has been shown to attenuate the action of IL-31 in mice [96] as IL-31, in addition to direct action on primary sensory neurones, also induces leukotriene B4 production in keratinocytes, the synthesis of which is controlled by the enzyme 5-lipoxygenase. Topical zileuton has shown positive results as an anti-acne agent [97]. However, we are still to see whether the results of the recently completed phase II study assessing 8 weeks of twice daily Q301 (zileuton) cream versus vehicle in adult subjects with moderate to severe AD show positive effects (NCT02426359).

The literature suggests that serotonin, i.e., 5-hydroxytryptamine (5-HT), and the 5-HT2 receptor family could contribute to dermal inflammation and pruritus both in general and in AD specifically [98,99,100]. The aminoguanidine derivative and 5-HT2B receptor antagonist AM1030 significantly diminished the T-cell-dependent and the T-cell-independent inflammatory responses in in vivo mouse and rat models and in an in vitro setting with staphylococcal enterotoxin A-stimulated leukocytes [101]. AM1030 is similar to a previously investigated agent demonstrating anti-inflammatory properties [102], and has been evaluated in a phase I/II study of 36 adults with mild to severe AD (NCT02379910). The study was completed in June 2015, though results have still to be revealed.

SP14019 is a formulation of cyclosporine A (CsA) compatible with cutaneous spray administration. It has been shown that the formulation delivers CsA to the target layers in the skin, with efficacy in preclinical models of AD as presented as a poster at the 25th European Academy of Dermatology and Venereology (EADV) conference [103]. SP14019 possibly bypasses the severe adverse events commonly seen from systemic CsA treatment [14,104,105]. Moreover, topical CsA has been extensively used in the field of ophthalmology [106]. An ongoing phase IIa trial of 36 patients with AD (3 age groups) is expected to be complete in late 2017 (NCT02865356). However, it is underpowered, and should be considered no more than a proof-of-concept study to perchance substantiate future studies.

We have, as stated in the introduction, omitted a range of therapies. However, we would like to notify the readers that many other ongoing trials are investigating new topical treatments for AD. These studies assess new TCS molecules and novel formulations of well-known corticosteroids. Moreover, several new formulations of both pimecrolimus and tacrolimus ointments and creams are being explored. Lastly, a range of humectants, emollients, probiotics, eubiotics, antibacterials, and molecules with undisclosed mechanism of action are items for investigation.

Despite the use of emollients in combination with TCSs or TCIs being effective mainstay topical treatments for AD, the many concerns from clinicians and patients over their use are significant and comprise an everyday challenge in dermatology. Consequently, the need for new topical treatments in AD is important. Fortunately, novel insights into the etiopathogenesis of AD have led to promising new therapies and treatment modalities. The primary areas of interest are currently the mitigation of the JAK-STAT pathway and PDE4 enzyme inhibition, closely followed by an array of other new drugs and pharmaceutical agents where, in several cases, the mechanisms of action need to be identified.

There are several new topical therapies in the pipeline for treating mild to moderate atopic dermatitis and as a supplement to systemic treatment in more severe, recalcitrant, and chronic disease. In this review, we present emerging topical therapies in a systematic fashion by summarizing the major background information followed by an appraisal of the existing data and studies related to the drugs.

M.D. is an investigator, speaker, and/or advisor for AbbVie, Pierre Fabre Dermo-Cosmétique, Meda Pharma, Leo Pharma, Sanofi-Genzyme, and Regeneron. C.V. is an investigator for AbbVie A/S and Pierre Fabre Dermo-Cosmétique. He has served on advisory boards for Astellas Pharma, and has been a speaker for Leo Pharma, Astellas, MSD, AbbVie, Novartis, and Pfizer. U.N. declares no relevant conflicts of interest.

All authors have participated sufficiently in the work to take public responsibility for its contents as stated at http://www.publicationethics.org.uk.

1.
Weidinger S, Novak N: Atopic dermatitis. Lancet 2016;387:1109-1122.
2.
Flohr C, Mann J: New insights into the epidemiology of childhood atopic dermatitis. Allergy 2014;69:3-16.
3.
Silverberg JI, Simpson EL: Associations of childhood eczema severity: a US population-based study. Dermatitis 2014;25:107-114.
4.
Willemsen MG, van Valburg RW, Dirven-Meijer PC, Oranje AP, van der Wouden JC, Moed H: Determining the severity of atopic dermatitis in children presenting in general practice: an easy and fast method. Dermatol Res Pract 2009;2009:357046.
5.
Bieber T: Atopic dermatitis. New Engl J Med 2008;358:1483-1494.
6.
Silverberg JI, Hanifin J, Simpson EL: Climatic factors are associated with childhood eczema prevalence in the United States. J Invest Dermatol 2013;133:1752-1759.
7.
Engebretsen KA, Bager P, Wohlfahrt J, Skov L, Zachariae C, Nybo Andersen AM, et al: Prevalence of atopic dermatitis in infants by domestic water hardness and season of birth: cohort study. J Allergy Clin Immunol 2017;139:1568-1574.
8.
Irvine AD, McLean WH, Leung DY: Filaggrin mutations associated with skin and allergic diseases. N Engl J Med 2011;365:1315-1327.
9.
Nygaard U, Riis JL, Deleuran M, Vestergaard C: Attention-deficit/hyperactivity disorder in atopic dermatitis. An appraisal of the current literature. Pediatr Allergy Immunol Pulmonol 2016;29:181-188.
10.
Andersen YMF, Egeberg A, Skov L, Thyssen JP: Comorbidities of atopic dermatitis: beyond rhinitis and asthma. Curr Dermatol Rep 2017;6:35-41.
11.
Simpson EL, Chalmers JR, Hanifin JM, Thomas KS, Cork MJ, McLean WH, et al: Emollient enhancement of the skin barrier from birth offers effective atopic dermatitis prevention. J Allergy Clin Immunol 2014;134:818-823.
12.
Nygaard U, Hvid M, Johansen C, Buchner M, Folster-Holst R, Deleuran M, et al: TSLP, IL-31, IL-33 and sST2 are new biomarkers in endophenotypic profiling of adult and childhood atopic dermatitis. J Eur Acad Dermatol Venereol 2016;30:1930-1938.
13.
Riis JL, Vestergaard C, Deleuran MS, Olsen M: Childhood atopic dermatitis and risk of attention deficit/hyperactivity disorder: a cohort study. J Allergy Clin Immunol 2016;138:608-610.
14.
Nygaard U, Vestergaard C, Deleuran M: Systemic treatment of severe atopic dermatitis in children and adults. Curr Treat Options Allergy 2014;1:384-396.
15.
Proudfoot LE, Powell AM, Ayis S, Barbarot S, Baselga Torres E, Deleuran M, et al: The European TREatment of severe Atopic eczema in children Taskforce (TREAT) survey. Br J Dermatol 2013;169:901-909.
16.
Horii KA, Simon SD, Liu DY, Sharma V: Atopic dermatitis in children in the United States, 1997-2004: visit trends, patient and provider characteristics, and prescribing patterns. Pediatrics 2007;120:e527-e534.
17.
Sidbury R, Davis DM, Cohen DE, Cordoro KM, Berger TG, Bergman JN, et al: Guidelines of care for the management of atopic dermatitis. Section 3. Management and treatment with phototherapy and systemic agents. J Am Acad Dermatol 2014;71:327-349.
18.
Arkwright PD, Motala C, Subramanian H, Spergel J, Schneider LC, Wollenberg A, et al: Management of difficult-to-treat atopic dermatitis. J Allergy Clin Immunol Pract 2013;1:142-151.
19.
Boralevi F, Saint Aroman M, Delarue A, Raudsepp H, Kaszuba A, Bylaite M, et al: Long-term emollient therapy improves xerosis in children with atopic dermatitis. J Eur Acad Dermatol Venereol 2014;28:1456-1462.
20.
Catherine Mack Correa M, Nebus J: Management of patients with atopic dermatitis: the role of emollient therapy. Dermatol Res Pract 2012;2012:836931.
21.
van Zuuren EJ, Fedorowicz Z, Arents BWM: Emollients and moisturizers for eczema: abridged Cochrane systematic review including GRADE assessments. Br J Dermatol 2017 DOI: 10.1111/bjd.15602.
22.
Eichenfield LF, Tom WL, Berger TG, Krol A, Paller AS, Schwarzenberger K, et al: Guidelines of care for the management of atopic dermatitis. Section 2. Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol 2014;71:116-132.
23.
Katayama I, Aihara M, Ohya Y, Saeki H, Shimojo N, Shoji S, et al: Japanese guidelines for atopic dermatitis 2017. Allergol Int 2017;66:230-247.
24.
Hengge UR, Ruzicka T, Schwartz RA, Cork MJ: Adverse effects of topical glucocorticosteroids. J Am Acad Dermatol 2006;54:1-15; quiz 16-18.
25.
Hajar T, Leshem YA, Hanifin JM, Nedorost ST, Lio PA, Paller AS, et al: A systematic review of topical corticosteroid withdrawal (“steroid addiction”) in patients with atopic dermatitis and other dermatoses. J Am Acad Dermatol 2015;72:541-549 e542.
26.
Carr WW: Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs 2013;15:303-310.
27.
Broeders JA, Ahmed Ali U, Fischer G: Systematic review and meta-analysis of randomized clinical trials (RCTs) comparing topical calcineurin inhibitors with topical corticosteroids for atopic dermatitis: A 15-year experience. J Am Acad Dermatol 2016;75:410-419 e413.
28.
Thijs J, Strickland I, Bruijnzeel-Koomen C, Nierkens S, Giovannone B, Csomor E, et al: Moving towards endotypes in atopic dermatitis: identification of patient clusters based on serum biomarker analysis. J Allergy Clin Immunol 2017;140:730-737.
29.
Guttman-Yassky E, Krueger JG, Lebwohl MG: Systemic immune mechanisms in atopic dermatitis and psoriasis with implications for treatment. Exp Dermatol 2017 DOI: 10.1111/exd.13336.
30.
Nygaard UH, Hvid M, Johansen C, Buchner M, Folster-Holst R, Deleuran M, et al: TSLP, IL-31, IL-33, and sST2 are new biomarkers in endophenotypic profiling of adult and childhood atopic dermatitis. J Eur Acad Dermatol Venereol 2016;30:1930-1938.
31.
Thijs JL, de Bruin-Weller MS, Hijnen D: Current and future biomarkers in atopic dermatitis. Immunol Allergy Clin North Am 2017;37:51-61.
32.
Ardern-Jones MR, Bieber T: Biomarkers in atopic dermatitis: it is time to stratify. Br J Dermatol 2014;171:207-208.
33.
Bieber T: Atopic dermatitis 2.0: from the clinical phenotype to the molecular taxonomy and stratified medicine. Allergy 2012;67:1475-1482.
34.
Bao L, Zhang H, Chan LS: The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT 2013;2:e24137.
35.
Vestergaard C, Deleuran M, Gesser B, Gronhoj Larsen C: Expression of the T-helper 2-specific chemokine receptor CCR4 on CCR10-positive lymphocytes in atopic dermatitis skin but not in psoriasis skin. Br J Dermatol 2003;149:457-463.
36.
Vestergaard C, Deleuran M, Gesser B, Larsen CG: Thymus- and activation-regulated chemokine (TARC/CCL17) induces a Th2-dominated inflammatory reaction on intradermal injection in mice. Exp Dermatol 2004;13:265-271.
37.
Nygaard U, van den Bogaard EH, Niehues H, Hvid M, Deleuran M, Johansen C, et al: The “Alarmins” HMBG1 and IL-33 downregulate structural skin barrier proteins and impair epidermal growth. Acta Derm Venereol 2017;97:305-312.
38.
Howell MD, Kim BE, Gao P, Grant AV, Boguniewicz M, Debenedetto A, et al: Cytokine modulation of atopic dermatitis filaggrin skin expression. J Allergy Clin Immunol 2007;120:150-155.
39.
Thyssen JP, Kezic S: Causes of epidermal filaggrin reduction and their role in the pathogenesis of atopic dermatitis. J Allergy Clin Immunol 2014;134:792-799.
40.
Nomura I, Goleva E, Howell MD, Hamid QA, Ong PY, Hall CF, et al: Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes. J Immunol 2003;171:3262-3269.
41.
Fukuyama T, Ehling S, Cook E, Baumer W: Topically administered Janus-kinase inhibitors tofacitinib and oclacitinib display impressive antipruritic and anti-inflammatory responses in a model of allergic dermatitis. J Pharmacol Exp Ther 2015;354:394-405.
42.
O'Shea JJ, Plenge R: JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity 2012;36:542-550.
43.
Meyer DM, Jesson MI, Li X, Elrick MM, Funckes-Shippy CL, Warner JD, et al: Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690,550, in rat adjuvant-induced arthritis. J Inflamm (Lond) 2010;7:41.
44.
Krueger J, Clark JD, Suarez-Farinas M, Fuentes-Duculan J, Cueto I, Wang CQ, et al: Tofacitinib attenuates pathologic immune pathways in patients with psoriasis: a randomized phase 2 study. J Allergy Clin Immunol 2016;137:1079-1090.
45.
Ports WC, Feldman SR, Gupta P, Tan H, Johnson TR, Bissonnette R: Randomized pilot clinical trial of tofacitinib solution for plaque psoriasis: challenges of the intra-subject study design. J Drugs Dermatol 2015;14:777-784.
46.
Papp KA, Bissonnette R, Gooderham M, Feldman SR, Iversen L, Soung J, et al: Treatment of plaque psoriasis with an ointment formulation of the Janus kinase inhibitor, tofacitinib: a phase 2b randomized clinical trial. BMC Dermatol 2016;16:15.
47.
Bissonnette R, Papp KA, Poulin Y, Gooderham M, Raman M, Mallbris L, et al: Topical tofacitinib for atopic dermatitis: a phase IIa randomized trial. Br J Dermatol 2016;175:902-911.
48.
Punwani N, Scherle P, Flores R, Shi J, Liang J, Yeleswaram S, et al: Preliminary clinical activity of a topical JAK1/2 inhibitor in the treatment of psoriasis. J Am Acad Dermatol 2012;67:658-664.
49.
Dastidar SG, Rajagopal D, Ray A: Therapeutic benefit of PDE4 inhibitors in inflammatory diseases. Curr Opin Investig Drugs 2007;8:364-372.
50.
Hanifin JM, Chan SC, Cheng JB, Tofte SJ, Henderson WR Jr, Kirby DS, et al: Type 4 phosphodiesterase inhibitors have clinical and in vitro anti-inflammatory effects in atopic dermatitis. J Invest Dermatol 1996;107:51-56.
51.
Grewe SR, Chan SC, Hanifin JM: Elevated leukocyte cyclic AMP-phosphodiesterase in atopic disease: a possible mechanism for cyclic AMP-agonist hyporesponsiveness. J Allergy Clin Immunol 1982;70:452-457.
52.
Furue M, Kitahara Y, Akama H, Hojo S, Hayashi N, Nakagawa H, et al: Safety and efficacy of topical E6005, a phosphodiesterase 4 inhibitor, in Japanese adult patients with atopic dermatitis: results of a randomized, vehicle-controlled, multicenter clinical trial. J Dermatol 2014;41:577-585.
53.
Baumer W, Hoppmann J, Rundfeldt C, Kietzmann M: Highly selective phosphodiesterase 4 inhibitors for the treatment of allergic skin diseases and psoriasis. Inflamm Allergy Drug Targets 2007;6:17-26.
54.
Murrell DF, Gebauer K, Spelman L, Zane LT: Crisaborole topical ointment, 2% in adults with atopic dermatitis: a phase 2a, vehicle-controlled, proof-of-concept study. J Drugs Dermatol 2015;14:1108-1112.
55.
Nazarian R, Weinberg JM: AN-2728, a PDE4 inhibitor for the potential topical treatment of psoriasis and atopic dermatitis. Curr Opin Investig Drugs 2009;10:1236-1242.
56.
Kircik L, Call R, Tschen E: Maximal use systemic exposure (MUSE) study evaluating AN2728, a novel boron-based small molecule, for the treatment of pediatric and adolescent subjects with mild-to-moderate atopic dermatitis. Orlando Dermatology Aesthetic and Clinical Conference, January 15-18, 2016.
57.
Stein Gold LF, Spelman L, Spellman MC, Hughes MH, Zane LT: A phase 2, randomized, controlled, dose-ranging study evaluating crisaborole topical ointment, 0.5 and 2% in adolescents with mild to moderate atopic dermatitis. J Drugs Dermatol 2015;14:1394-1399.
58.
Tom WL, Van Syoc M, Chanda S, Zane LT: Pharmacokinetic profile, safety, and tolerability of crisaborole topical ointment, 2% in adolescents with atopic dermatitis: an open-label phase 2a study. Pediatr Dermatol 2016;33:150-159.
59.
Zane LT, Kircik L, Call R, Tschen E, Draelos ZD, Chanda S, et al: Crisaborole topical ointment, 2% in patients ages 2 to 17 years with atopic dermatitis: a phase 1b, open-label, maximal-use systemic exposure study. Pediatr Dermatol 2016;33:380-387.
60.
Paller AS, Tom WL, Lebwohl MG, Blumenthal RL, Boguniewicz M, Call RS, et al: Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J Am Acad Dermatol 2016;75:494-503 e496.
61.
Ishii N, Wakita H, Shirato M: Effect of the phosphodiesterase 4 inhibitor E6005 on nerve growth factor elevation in irritated skin of NC/Nga mice. J Dermatol Sci 2014;76:263-264.
62.
Wakita H, Ohkuro M, Ishii N, Hishinuma I, Shirato M: A putative antipruritic mechanism of the phosphodiesterase-4 inhibitor E6005 by attenuating capsaicin-induced depolarization of C-fibre nerves. Exp Dermatol 2015;24:215-216.
63.
Ishii N, Shirato M, Wakita H, Miyazaki K, Takase Y, Asano O, et al: Antipruritic effect of the topical phosphodiesterase 4 inhibitor E6005 ameliorates skin lesions in a mouse atopic dermatitis model. J Pharmacol Exp Ther 2013;346:105-112.
64.
Ohba F, Nomoto M, Hojo S, Akama H: Safety, tolerability and pharmacokinetics of a novel phosphodiesterase inhibitor, E6005 ointment, in healthy volunteers and in patients with atopic dermatitis. J Dermatolog Treat 2016;27:241-246.
65.
Ohba F, Matsuki S, Imayama S, Matsuguma K, Hojo S, Nomoto M, et al: Efficacy of a novel phosphodiesterase inhibitor, E6005, in patients with atopic dermatitis: an investigator-blinded, vehicle-controlled study. J Dermatolog Treat 2016;27:467-472.
66.
Hanifin JM, Ellis CN, Frieden IJ, Folster-Holst R, Stein Gold LF, Secci A, et al: OPA-15406, a novel, topical, nonsteroidal, selective phosphodiesterase-4 (PDE4) inhibitor, in the treatment of adult and adolescent patients with mild to moderate atopic dermatitis (AD): a phase-II randomized, double-blind, placebo-controlled study. J Am Acad Dermatol 2016;75:297-305.
67.
Reich A, Riepe C, Anastasiadou Z, Medrek K, Augustin M, Szepietowski JC, et al: Itch assessment with visual analogue scale and numerical rating scale: determination of minimal clinically important difference in chronic itch. Acta Derm Venereol 2016;96:978-980.
68.
Zhao L, Chen X, Cai L, Zhang C, Wang Q, Jing S, et al: Randomized, double-blind, placebo-controlled, multiple-dose study of the safety, tolerability and pharmacokinetics of benvitimod, a candidate drug for the treatment of psoriasis. J Clin Pharm Ther 2014;39:418-423.
69.
Bissonnette R, Poulin Y, Zhou Y, Tan J, Hong HC, Webster J, et al: Efficacy and safety of topical WBI-1001 in patients with mild to severe atopic dermatitis: results from a 12-week, multicentre, randomized, placebo-controlled double-blind trial. Br J Dermatol 2012;166:853-860.
70.
Bissonnette R, Bolduc C, Maari C, Nigen S, Webster JM, Tang L, et al: Efficacy and safety of topical WBI-1001 in patients with mild to moderate psoriasis: results from a randomized double-blind placebo-controlled, phase II trial. J Eur Acad Dermatol Venereol 2012;26:1516-1521.
71.
Bissonnette R, Chen G, Bolduc C, Maari C, Lyle M, Tang L, et al: Efficacy and safety of topical WBI-1001 in the treatment of atopic dermatitis: results from a phase 2A, randomized, placebo-controlled clinical trial. Arch Dermatol 2010;146:446-449.
72.
Amagai Y, Oida K, Matsuda A, Jung K, Kakutani S, Tanaka T, et al: Dihomo-γ-linolenic acid prevents the development of atopic dermatitis through prostaglandin D1 production in NC/Tnd mice. J Dermatol Sci 2015;79:30-37.
73.
Focke M, Sesztak-Greinecker G, Brannath W, Gotz M, Jarisch R, Hemmer W: Plasma levels of polyunsaturated fatty acids in children with atopic dermatitis and in atopic and nonatopic controls. Wien Klin Wochenschr 2005;117:485-491.
74.
Kawashima H, Tateishi N, Shiraishi A, Teraoka N, Tanaka T, Tanaka A, et al: Oral administration of dihomo-γ-linolenic acid prevents development of atopic dermatitis in NC/Nga mice. Lipids 2008;43:37-43.
75.
GlobeNewswire: DS107E-02 - Positive Phase II Results in Atopic Dermatitis (press release). Dublin, PRWEB, July 8, 2014.
76.
PRNewswire: DS Biopharma Announces Positive Top-Line Phase IIa Trial Results for DS107 as an Oral Treatment for Moderate to Severe Atopic Dermatitis (press release). Dublin, PRNewswire, January 7, 2016.
77.
Kittaka H, Uchida K, Fukuta N, Tominaga M: Lysophosphatidic acid-induced itch is mediated by signalling of LPA5 receptor, phospholipase D and TRPA1/TRPV1. J Physiol 2017;595:2681-2698.
78.
Shibata T, Takahashi K, Matsubara Y, Takahashi N, Mori Y, Uchida K: 15-Deoxy-?12,14-prostaglandin J2 as a potential TRPV1-dependent atopic dermatitis enhancer. Free Radic Biol Med 2014;75(suppl 1):S49.
79.
Cevikbas F, Wang X, Akiyama T, Kempkes C, Savinko T, Antal A, et al: A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: Involvement of TRPV1 and TRPA1. J Allergy Clin Immunol 2014;133:448-460.
80.
Yun JW, Seo JA, Jang WH, Koh HJ, Bae IH, Park YH, et al: Antipruritic effects of TRPV1 antagonist in murine atopic dermatitis and itching models. J Invest Dermatol 2011;131:1576-1579.
81.
Yun JW, Seo JA, Jeong YS, Bae IH, Jang WH, Lee J, et al: TRPV1 antagonist can suppress the atopic dermatitis-like symptoms by accelerating skin barrier recovery. J Dermatol Sci 2011;62:8-15.
82.
Lim KM, Park YH: Development of PAC-14028, a novel transient receptor potential vanilloid type 1 (TRPV1) channel antagonist as a new drug for refractory skin diseases. Arch Pharm Res 2012;35:393-396.
83.
Gordon JS, Wolanin PM, Gonzalez AV, Fela DA, Sarngadharan G, Rouzard K, et al: Topical N-acetyl-S-farnesyl-L-cysteine inhibits mouse skin inflammation, and unlike dexamethasone, its effects are restricted to the application site. J Invest Dermatol 2008;128:643-654.
84.
Fuhst R, Runge F, Buschmann J, Ernst H, Praechter C, Hansen T, et al: Toxicity profile of the GATA-3-specific DNAzyme hgd40 after inhalation exposure. Pulm Pharmacol Ther 2013;26:281-289.
85.
Tamauchi H, Amoh Y, Itoh M, Terashima M, Masuzawa M, Habu S, et al: GATA-3 regulates contact hyperresponsiveness in a murine model of allergic dermatitis. Immunobiology 2012;217:446-454.
86.
Marwarha G, Raza S, Hammer K, Ghribi O: 27-Hydroxycholesterol: a novel player in molecular carcinogenesis of breast and prostate cancer. Chem Phys Lipids 2017;207(pt B): 108-126.
87.
Paterniti I, Campolo M, Siracusa R, Cordaro M, Di Paola R, Calabrese V, et al: Liver X receptors activation, through TO901317 binding, reduces neuroinflammation in Parkinson's disease. PLoS One 2017;12:e0174470.
88.
Tanaka N, Aoyama T, Kimura S, Gonzalez FJ: Targeting nuclear receptors for the treatment of fatty liver disease. Pharmacol Ther 2017;179:142-157.
89.
Ouedraogo ZG, Fouache A, Trousson A, Baron S, Lobaccaro JA: Role of the liver X receptors in skin physiology: putative pharmacological targets in human diseases. Chem Phys Lipids 2017;207(pt B):59-68.
90.
Kim B, Kim JE, Kim H, Lee JD, Choi KY, Lee SH: Co-treatment with retinyl retinoate and a PPARα agonist reduces retinoid dermatitis. Int J Dermatol 2012;51:733-741.
91.
Adams B: Midstage Vitae Drug Flops in PhII, Leaving Allergan's $640M Buyout in Question (press release). Questex LLC, November 3, 2016.
92.
Roebrock K, Wolf M, Bovens S, Lehr M, Sunderkotter C: Inhibition of benzalkonium chloride-induced skin inflammation in mice by an indol-1-ylpropan-2-one inhibitor of cytosolic phospholipase A2α. Br J Dermatol 2012;166:306-316.
93.
Henderson WR Jr, Oslund RC, Bollinger JG, Ye X, Tien YT, Xue J, et al: Blockade of human group X secreted phospholipase A2 (GX-sPLA2)-induced airway inflammation and hyperresponsiveness in a mouse asthma model by a selective GX-sPLA2 inhibitor. J Biol Chem 2011;286:28049-28055.
94.
Sonkoly E, Muller A, Lauerma AI, Pivarcsi A, Soto H, Kemeny L, et al: IL-31: a new link between T cells and pruritus in atopic skin inflammation. J Allergy Clin Immunol 2006;117:411-417.
95.
Ruzicka T, Hanifin JM, Furue M, Pulka G, Mlynarczyk I, Wollenberg A, et al: Anti-interleukin-31 receptor A antibody for atopic dermatitis. N Engl J Med 2017;376:826-835.
96.
Andoh T, Harada A, Kuraishi Y: Involvement of leukotriene B4 released from keratinocytes in itch-associated response to intradermal interleukin-31 in mice. Acta Derm Venereol 2017;97:922-927.
97.
Zouboulis CC: Zileuton, a new efficient and safe systemic anti-acne drug. Dermatoendocrinol 2009;1:188-192.
98.
Rausl A, Nordlind K, Wahlgren CF: Pruritic and vascular responses induced by serotonin in patients with atopic dermatitis and in healthy controls. Acta Derm Venereol 2013;93:277-280.
99.
Leon-Ponte M, Ahern GP, O'Connell PJ: Serotonin provides an accessory signal to enhance T-cell activation by signaling through the 5-HT7 receptor. Blood 2007;109:3139-3146.
100.
Muller T, Durk T, Blumenthal B, Grimm M, Cicko S, Panther E, et al: 5-Hydroxytryptamine modulates migration, cytokine and chemokine release and T-cell priming capacity of dendritic cells in vitro and in vivo. PLoS One 2009;4:e6453.
101.
Palmqvist N, Siller M, Klint C, Sjodin A: A human and animal model-based approach to investigating the anti-inflammatory profile and potential of the 5-HT2B receptor antagonist AM1030. J Inflamm (Lond) 2016;13:20.
102.
Dambrova M, Zvejniece L, Skapare E, Vilskersts R, Svalbe B, Baumane L, et al: The anti-inflammatory and antinociceptive effects of NF-κB inhibitory guanidine derivative ME10092. Int Immunopharmacol 2010;10:455-460.
103.
Santos B, Lluch N, Armengol J, Llorente M, Esparza I, Ruiz L: Topical therapy stable, high-load topical formulation of ciclosporin for the treatment of psoriasis and atopic dermatitis, P2298. 25th EADV Conference, Vienna, 2016.
104.
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. Eur J Pediatr 2013;172:351-356.
105.
Schmitt J, Schmitt N, Meurer M: Cyclosporin in the treatment of patients with atopic eczema - a systematic review and meta-analysis. J Eur Acad Dermatol Venereol 2007;21:606-619.
106.
Levy O, Labbe A, Borderie V, Laroche L, Bouheraoua N: Topical cyclosporine in ophthalmology: Pharmacology and clinical indications (in French). J Fr Ophtalmol 2016;39:292-307.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.