+ All Categories
Home > Documents > Therapeutic strategies in extrinsic atopic dermatitis: focus on inhibition of IL-4 as a new...

Therapeutic strategies in extrinsic atopic dermatitis: focus on inhibition of IL-4 as a new...

Date post: 24-Nov-2023
Category:
Upload: uniecampus
View: 0 times
Download: 0 times
Share this document with a friend
11
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=iett20 Download by: [Vito Di Lernia] Date: 29 November 2015, At: 08:13 Expert Opinion on Therapeutic Targets ISSN: 1472-8222 (Print) 1744-7631 (Online) Journal homepage: http://www.tandfonline.com/loi/iett20 Therapeutic strategies in extrinsic atopic dermatitis: focus on inhibition of IL-4 as a new pharmacological approach Vito Di Lernia MD To cite this article: Vito Di Lernia MD (2015) Therapeutic strategies in extrinsic atopic dermatitis: focus on inhibition of IL-4 as a new pharmacological approach, Expert Opinion on Therapeutic Targets, 19:1, 87-96 To link to this article: http://dx.doi.org/10.1517/14728222.2014.965682 Published online: 04 Oct 2014. Submit your article to this journal Article views: 239 View related articles View Crossmark data Citing articles: 1 View citing articles
Transcript

Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=iett20

Download by: [Vito Di Lernia] Date: 29 November 2015, At: 08:13

Expert Opinion on Therapeutic Targets

ISSN: 1472-8222 (Print) 1744-7631 (Online) Journal homepage: http://www.tandfonline.com/loi/iett20

Therapeutic strategies in extrinsic atopicdermatitis: focus on inhibition of IL-4 as a newpharmacological approach

Vito Di Lernia MD

To cite this article: Vito Di Lernia MD (2015) Therapeutic strategies in extrinsic atopicdermatitis: focus on inhibition of IL-4 as a new pharmacological approach, Expert Opinion onTherapeutic Targets, 19:1, 87-96

To link to this article: http://dx.doi.org/10.1517/14728222.2014.965682

Published online: 04 Oct 2014.

Submit your article to this journal

Article views: 239

View related articles

View Crossmark data

Citing articles: 1 View citing articles

1. Introduction

2. Genetics

3. Role of IL-4 in AD from a

historical perspective

4. IL-4 in intrinsic versus extrinsic

AD

5. Treatments affecting IL-4

expression

6. IL-4 as a therapeutic target

7. Conclusions

8. Expert opinion: future place of

anti-IL-4-based strategies in

the treatment of AD

Review

Therapeutic strategies in extrinsicatopic dermatitis: focus oninhibition of IL-4 as a newpharmacological approachVito Di LerniaArcispedale Santa Maria Nuova-IRCCS, Dermatology Unit, Reggio Emilia, Italy

Introduction: Recent data about atopic dermatitis (AD) pathogenesis

postulate that T cells and their related cytokines and chemokines are primarily

responsible for the inflammatory responses.

Areas covered: AD, the primary complex disease associated with filaggrin

deficiency, is characterized by cutaneous inflammation driven by type 2

helper T (TH2) cells. TH2-related molecules, such as IL-4, IL-13, dominate the

immune infiltrate. Experimental evidences suggest that these cytokines may

be considered attractive therapeutic targets in AD, particularly in extrinsic

AD with IgE overproduction. Recently, a fully human monoclonal antibody

directed against the IL-4 receptor a subunit blocking IL-4 and IL-13 signaling

has been evaluated in Phase I and Phase II clinical trials in patients with mod-

erate-to-severe AD with significant improvement in disease severity. Phase III

trials are ongoing.

Expert opinion: Treatment of AD represents a therapeutic challenge. TH2

cytokine-targeted therapies represent promising treatment options that

could improve the therapeutic armamentarium for AD. These therapies are

likely to become future therapeutic options in AD, particularly in the extrinsic

AD.

Keywords: atopic dermatitis, cytokines, IL-4, IL-13, inhibition, treatment

Expert Opin. Ther. Targets (2015) 19(1):87-96

1. Introduction

Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin disease whosepathogenesis still remains to be fully elucidated. It is considered a primarily T-celldriven disorder characterized by a type 2 helper T (TH2) environment and epider-mal barrier dysfunction. Current models associate AD with progressive activation ofTH2 and type 22 helper T (TH22) cytokines, with the association of a type 1 helperT (TH1) component in patients with chronic AD and a possible contribution oftype 17 helper T (TH17) cells [1].

The prevalence of AD is high. Reports range from 10 to 20% prevalence inchildren and 1 to 3% in adults [2-4]. New diagnoses are estimated at almost 11%per year [4]. Prevalence of the disease has been found to have increased greatly insome parts of the world [5].

For the past decades, research has been focused on pathogenesis of AD with stud-ies showing a complex etiology, due to the activation of multiple immunologic andinflammatory pathways including TH1/TH2 cell dysregulation, IgE overproduction,mast cell hyperactivity and dendritic cell signaling. Subsequently, proofs emergedshowing structural abnormalities in the epidermis [6] and resulting inflammation [7].Thus, a leaky epithelial barrier as well as chronic immune activation have been con-sidered the central players of the pathophysiology of this disease [6]. In particular, a

10.1517/14728222.2014.965682 © 2015 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 87All rights reserved: reproduction in whole or in part not permitted

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

primary defect in the skin barrier followed by epiphenomenalimmunologic sequelae is considered the core of the so-called‘outside-in’ hypothesis [7]. The opposing hypothesis is the‘inside-out’ or immune-driven which advocates that AD is aprimarily immunologically driven disease caused by increasedexpression of cytokines produced by distinct T-cell subsetswith a secondary barrier defect [7-10]. However, the twopathogenic models could not be mutually exclusive and inde-pendent. As a matter of fact, a primary inherited barrierabnormality in AD could stimulate downstream paracrinemechanisms that could further compromise permeability bar-rier function, finalizing a possible ‘outside--inside--outside’pathogenic vicious circle [7].AD usually presents during early infancy and childhood. It

can disappear with age, go into remission and recur aftermany years or persist into adulthood [11]. AD may less fre-quently start around puberty or even in adulthood. In thiscase, patients continue to suffer throughout their lives [12].Various reports set the prevalence of adult onset AD atbetween 13 and 47% [13].At least two forms of AD have been delineated, the

‘extrinsic’ form associated with IgE-mediated sensitizationinvolving 70 -- 80% of the patients and the ‘intrinsic’ form,without IgE-mediated sensitization affecting 20 -- 30% ofthe patients [14]. Extrinsic AD has a pivotal role in the emer-gence of IgE-related disorders, in particular allergic rhinitisand asthma, in the so-called ‘atopic march’ [15,16]. Additionalatopic comorbidities consist of allergic eye disease, atopickeratoconjunctivitis and immediate-type food allergy [17].Beyond IgE-mediated diseases, other associated disease mayinclude skin infections, sleep disruption and mental healthdisorders [18,19].

The individual, family and public health burden of AD issignificant. The signs and symptoms of the disease negativelyaffect quality of life for patients and their families. In addi-tion, the comorbidities can amplify noticeably the negativeimpact of the disease [20]. Adults with severe AD are usuallygreatly affected by the disease. Indeed, even when the clinicalpresentation is mild, the psychosocial and economic burdenof the disease can be profound [21]. Nevertheless, moderatelylittle has been published on adult AD compared to the bodyof literature devoted to childhood.

Management of AD must consider the individual symp-tomatic variability of the disease. Basic therapy is focused ontopical glucocorticosteroids and calcineurin inhibitors [22].Adjuvant therapy includes UV irradiation, preferably withUVA1 wavelength or UVB 311 nm [22], which show variableefficacy in controlling the disease. Systemic corticosteroids arefrequently utilized for severe and acute exacerbations witheffectiveness, but long-term side effects advise against theiruse in a chronic disease such as AD. Mainstays of the manage-ment of severe or difficult-to-control patients require broadT-cell-targeting therapeutics. Depending on geographicareas, several immunosuppressive drugs, as cyclosporine A,methotrexate, azathioprine and mycophenolate mofetil areadministered to improve the clinical symptoms of severeAD [23-26]. However, with the exception of cyclosporine A,none of these drugs has a registered indication for AD(Table 1). Recently, different biologic drugs, such as mepoli-zumab, omalizumab, rituximab and tocilizumab, have beentried out for the treatment of AD [27-32].

The chronic and severe nature of AD would require moreeffective and better-tolerated therapeutic options. With theidentification of various cytokines abnormally expressed inthe skin affected by AD, a crucial challenge is to identifymolecules that are ‘driver’ cytokines. Emerging evidencenow suggests that IL-4 is central in the pathogenesis ofAD [33]. This paper reviews the rationale for targeting IL-4and then describes the preliminary results of clinical trialswith anti-IL-4 inhibitors.

2. Genetics

The association of mutations in filaggrin gene and AD is con-sidered one of the strongest between any gene and a complexdisease [34]. Overall, patients with AD show a prevalence offilaggrin (FLG) mutations of between 20 and 50% [35].Hershey et al. showed that a R576 allele of IL-4 receptor a,which is common among patients with allergic inflammatorydisorders, was strongly associated with atopy [36]. However,the frequency of these substitutions is very variable amongracial and ethnic groups. Polymorphisms have been found ingenes encoding IL-4, IL-13, signal transducer and activatorof transcription 6 (STAT-6), and the high-affinity IgEreceptor [37]. Genome-wide linkage studies identified multiplesusceptibility loci on 3q and 17q [38]. Candidate region linkagestudies discovered other susceptibility loci on 5q23-33,

Article highlights.

. Atopic dermatitis (AD) is a chronic, inflammatory skindisorder. Adults with severe AD are usually greatlyaffected by the disease with a profound psychosocialand economic burden of the disease.

. Adult patients with AD may be included in dermatologicareas with a high unmet medical need.

. AD is characterized by a type 2 helper T (TH2)environment. A TH1 component and a possiblecontribution of TH17 cells characterize chronic AD.

. TH2 cytokines can modulate and alter barrierfunction, through modulation of the expressionof filaggrin.

. IL-4 and IL-13 are key cytokines that are required forthe initiation and maintenance of theTH2 immune response.

. Clinical studies with a monoclonal antibody that targetsIL-4 and IL-13 provide concrete evidence for a key roleof TH2 activation in the pathogenesis of AD.

. The blockade of IL-4 and IL-13 signaling may be anattractive therapeutic target for AD.

This box summarizes key points contained in the article.

V. Di Lernia

88 Expert Opin. Ther. Targets (2015) 19(1)

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

11q13 and 13q12-14. Finally, although at least 28 candidategenes have to date been verified in association studies, only cor-relation with genes of IL-4, IL-13, IL-4 receptor a, mast cellchymase and serine protease inhibitor, kazal-type 5 have beenreplicated in more than two different studies [38]. Interestingly,low quantities of IFN-g and high amounts of IL-4-producingCD4+ cord blood T cells were considered to be associatedwith the risk to develop AD during the first 2 years of life [39].

3. Role of IL-4 in AD from a historicalperspective

As IL-4 was considered able to enhance IgE production andIgE-Fc receptor expression by B cells, Furue et al. hypothesizedits key role in the regulation of the immune response in ADalready in 1989 [40]. High levels of the IL-4 gene expressionwere demonstrated in peripheral blood mononuclear cellsobtained from highly atopic patients [41]. In 1991, Reinholdet al. showed that high levels of IL-4 could promote a largeexpansion of skin-infiltrating lymphocytes from AD in vitro[42]. IL-4 was proved to control the selective endothelium-driven transmigration of eosinophils from allergic individuals.IL-4 showed chemotactic properties only for eosinophils fromthe peripheral blood of patients with AD and not for eosino-phils from normal individuals [43,44]. IL-4 production andIL-4 receptor expression was enhanced in AD and modulatedby IFN-g [45]. A predominant TH2 cytokine milieu in the ini-tiating stages of acute AD lesions was explained because acutelesions are associated with a T cells expressing mRNA forIL-4 and IL-13 compared with chronic lesions [46,47]. Chroniclichenified AD skin lesions show fewer IL-4 mRNA-andIL-13 mRNA expressing cells but significantly greater num-bers of IL-5, GM-CSF, IL-12 and IFN-g mRNA-expressingcells, a TH1 component and a possible contribution of TH17cells. These cytokines substantiate the growth of eosinophilsand macrophages and promote the mixed TH1/TH2 pattern,which is more characteristic of chronic lesions of AD.

3.1 Eosinophil chemoattractionIL-4 is a potent inducer of CC chemokine eotaxin expressionin dermal fibroblasts which in turn may account for

eosinophilic infiltration of involved skin areas in AD [48].IL-4, at both the mRNA and protein levels, promotes expres-sion of the eotaxin-3/CCL26, a strong chemoattractant foreosinophils and member of the CC chemokine subfamily [49],the chemokine receptor CCR7, a marker linked to migratoryevents in adaptive immune function, and the chemokineligand 23 [50,51].

3.2 Other IL-4 regulated factorsIL-4 strongly enhances the generation of CD1a(+) Langerhanscells (LC)/dendritic cells. In addition to LC, a distinct popula-tion of inflammatory dendritic epidermal cells (IDEC) isexclusively present in inflammatory skin lesions, includingAD. They are epidermally located; express CD1a, CD11band class II molecules; and do not contain Birbeck granules.IDEC of skin lesions of AD display the highest expression ofthe high-affinity receptor for IgE (FcepsilonRI) ever detectedon human antigen-presenting cells. [52,53].

IL-4 also upregulates the expression of the skin-homingstructures E-cadherin and cutaneous lymphocyte antigen.IL-4 is able to induce also IL-31, a tissue-signaling cytokinecorrelated with the induction of pruritus in the AD skinlesions [54-56].

3.3 Filaggrin and skin barrier disruptionFLG is an epidermal protein thought to be important forcutaneous barrier integrity. FLG mutations may play a rolein the development of each of the key features of AD througha functional barrier defect and enhanced cutaneous allergeningress [57] leading to a TH2-dominant infiltrate [58]. FLGdeficiency may contribute to defective extracellular lamellaethrough a number of different mechanisms. Tight junctionsare decreased in FLG-deficient individuals who show also areduced density of corneodesmosin, the major protein com-ponent of corneodesmosomes [57]. In addition, the increaseof skin-surface pH that is detected in FLG-deficient individu-als may enhance the activities of multiple serine proteaseswhich all show neutral-to-alkaline pH optimal. Serumproteases in turn may induce the maturation and release ofprimary cytokine IL-1a and IL-1b from their precursor

Table 1. Current systemic therapeutic approaches for AD and relative limitation.

Drug Registration for AD Limitations

Cyclosporine A Yes Long-term use limited by nephrotoxicityMethotrexate No Gastrointestinal disturbances, contraception indicated also in

malesAzathioprine No Monitoring for bone marrow suppression, hepatotoxicity,

increase risk of malignancyMycophenolate mofetil No Gastrointestinal disturbances, risk of infectionsCorticosteroids - Short courses (limited to flares)PUVA therapy - Long term use limited by risk of non melanoma skin cancer risk.

Gastrointestinal disturbances. Hospital-based treatment

AD: Atopic dermatitis.

Therapeutic strategies in extrinsic atopic dermatitis

Expert Opin. Ther. Targets (2015) 19(1) 89

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

molecules and drive kallikrein 5-mediated TH2 inflammation,even in the absence of allergen priming [57,59].However, AD dermatitis develops in only ~ 42% of all

FLG heterozygotes suggesting the interplay of genetic modi-fiers and environmental factors [60]. TH2 cytokines can mod-ulate barrier function, through modulation of the expressionof FLG, other structural proteins and peptides important formicrobial barrier function [61,62]. In particular, TH2 cytokinesIL-4 and IL-13 attenuate the expression of genes that regulateepidermal cellular structures and the barrier function [63]. Inaddition, IL-4 and IL-13 may alter epidermal barrier func-tions through STAT6 decreasing expressions of FLG, loricrinand involucrin in keratinocytes [64]. IL-4 suppresses thesynthesis of ceramides, the cutaneous permeability barrierfunctions, the recovery of cutaneous permeability barrier dys-function [65] and the cohesion of stratum corneum by reduc-ing epidermal differentiation [66]. Indeed, it was observedthat IL-4 decreased the amount of corneodesmosomes anddown-regulated the expression of desmoglein 1. It enhancesexpression of kallikrein 7 which in turn degrades corneodes-mosome proteins such as desmoglein 1, desmocollin 1 andcorneodesmosin [67]. These multifaceted interactions may beconsidered a link between outside and inside hypothesesshowing the way for a ‘outside--inside--outside’ pathogenicloop [68].

3.4 Antimicrobial peptides expressionKeratinocytes produce and secrete several antimicrobial pepti-des which function as endogenous antibiotics and as signalingmolecules within the cutaneous innate immune system.Defensins are constitutively expressed or induced after exoge-nous stimuli, contact with bacteria or tissue injury [69]. Theyplay a part in the host defense by disrupting the cytoplasmicmembrane of microorganisms. In this way, the innate immunesystem quickly activates an unspecific, standardized first-linedefense against different pathogens. Human b-defensins(HBDs) are one major class of antimicrobial peptides that areprimarily expressed in the epithelial cells [69,70]. They are alsoable to recruit dendritic and T cells to inflammatory sites. Inthis way, they mobilize and amplify adaptive immune hostdefenses linking the innate and adaptive immune responses[71,72]. Defects in this system cause barrier dysfunction whichresults in increased allergen penetration through the epidermisand predispose to secondary skin infections [73,74]. AD hasbeen associated with decreased or impaired production ofHBDs [74,75]. In addition, overexpression of TH2 cytokinessuch as IL-4 and IL-13, but also IL-10, downregulate endoge-nous antimicrobial peptide expression, as HBDs and cathelici-din LL-37, in AD keratinocytes [74-76]. Further mechanismspotentially causing mechanical and chemical epidermal barrierdysfunction in AD involve the enhancement of protease activ-ity. IL-4 and IL-13 are able to enhance KLK7 expression andfunction in keratinocytes [77]. KLK7 is a chymotrypsin-likeserine protease which may degrade human cathelicidin antimi-crobial peptide LL-37 [77].

3.5 Staphylococcus aureusPatients with AD have a distinctive predisposition to coloni-zation or infection by a number of microbial organisms,especially Staphylococcus aureus and herpes simplex virus [78].Bacterial colonization is directly correlated with the severityof eczema and highest on lesional skin, but colony countsare high on clinically normal skin of AD patients aswell [79,80]. S. aureus toxins induce expansion of IL-4- andIL-5-producing TH2 clones, leading to increased IgE synthesisand eosinophil activation. An elevation in the pH of the stra-tum corneum, which is commonly observed in FLG-deficientpersons, may lead to enhanced S. aureus adhesion and multi-plication [81]. Staphylococcal-specific T cell are primarily ofthe TH17 subset. AD patients show an ineffective IL-17-dependent upregulation of HBD-2 due to a partial inhibitioninduced by the type 2 microenvironment [80,81]. Also, HBD-3has potent killing activity against S. aureus, but increasedlevels of TH2 cytokines IL-4 and IL-13 inhibit keratinocytemobilization of HBD-3 [82]. Thus, barrier disruption itselffavors bacterial colonization and secondary infections; onthe other hand, TH2 cytokines inhibit mobilization of antimi-crobial peptides (Figure 1). Barrier disruption and TH2 micro-environment act as interdependent cofactors in defectiveS. aureus killing in patients with AD [83].

4. IL-4 in intrinsic versus extrinsic AD

A subgroup of patients affected with ‘intrinsic’ AD (20% ofcases) are known to have normal total and specific IgE levels,and negative skin prick tests and Radio-Allergo-Sorbent Teststowards common environmental allergens. The incidence ofFLG gene mutations was discovered to be significantly lowerin the intrinsic or ‘nonatopic’ AD [84]. The existence of such‘nonatopic’ or intrinsic form of AD raised the questionwhether this form may be characterized by distinct immuno-logic features and cytokine pattern with respect to the classicTH2-related cytokine pattern [85,86].

A higher immune activation has been found in patientswith intrinsic compared with extrinsic AD, involving otherT-cell subsets, such as the TH17 and TH22 cells. IL-22mRNA expression is significantly increased in lesional versusnonlesional skin in the setting of both AD variants but is sig-nificantly higher in lesional intrinsic compared with extrinsicAD [87]. Both intrinsic and extrinsic AD lesions show markedTH2 activation with high IL-4/IL-13 expression, with IL-13higher levels in extrinsic AD [88].

5. Treatments affecting IL-4 expression

A significant decrease of soluble IL-4 receptor (sIL-4R) in theserum of patients with AD has been observed after successfulUVA1 phototherapy [89]. Also, cyclosporine and dexametha-sone significantly downregulate the activity of IL-4 in adose-dependent manner [90]. Phosphodiesterase inhibitors

V. Di Lernia

90 Expert Opin. Ther. Targets (2015) 19(1)

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

suppress IL-4 and IL-5 secretion by human T-helper type 2cells [91,92]. Treatment with recombinant IFN-g was assessedin patients with AD with the aim to decrease serum IgE andIL-4 levels, to shift the cytokine balance, and thereby lead toclinical improvement. After treatment with rIFN-g , serumlevels of IL-4 and IL-6 decline without affecting IgE produc-tion [93,94] and patients often showed marked improvement.However, toxicity and the short half-life of IFN-g in serumwere leading causes of reduced enthusiasm to manage it forthe treatment of AD. A pioneering experience was conductedwith a highly efficient mouse dominant negative IL-4/IL-13antagonist blocking both IL-4 and IL-13 signal transductionsin an AD model. The simultaneous suppression of both sig-nals successfully controlled TH2-type chronic dermatitis [95].

6. IL-4 as a therapeutic target

A fully human monoclonal antibody directed against theIL-4 receptor a subunit and administered via subcutaneousinjection has recently been developed. As IL-4 and IL-13signaling occurs through two different IL-4 receptors (type Iand II), which both contain a common IL-4R a subunit,this antibody inhibits both IL-4 and IL-13 signaling. It hasbeen shown to be efficacious in patients with persistent, mod-erate-to-severe asthma and elevated eosinophil levels [96]. Noclear pattern of change in blood eosinophil levels wasobserved, but reduced levels of TH2-associated inflammatorybiomarkers, in particular fractional exhaled nitric oxide level,

serum IgE, eotaxin-3 and thymus and activation-regulatedchemokine (TARC) were assessed after treatment. Interest-ingly, serum TARC levels reflect disease activity of AD aswell [97,98]. Pruritus correlates with the ratio of serum TARCwhich is actually considered the most sensitive disease markerof AD [98]. Recently, IL-4 receptor a antagonist was evaluatedin a series of four randomized, double-blind, placebo-controlled trials in adults with moderate-to-severe AD [99].The treatment was given as monotherapy in two 4-week trialsand in one 12-week trial and in combination with topicalglucocorticoids in another 4-week study. End points includedthe Eczema Area and Severity Index score, the investigator’sglobal assessment score, pruritus, safety assessments andserum biomarker levels. A significant improvement in skinlesions together with a rapid and substantial reduction inpruritus was observed in patients treated with the IL-4 recep-tor a antagonist. The treatment was associated with a decreasein serum biomarker levels. Particularly, there was adose-dependent reduction of K16, a marker of keratinocyteproliferation and innate immunity. A significant associatedreduction in the level of TARC was confirmed [96,99]. Aboutsafety, there were more mild-to-moderate adverse events inthe patients treated with the active treatment (i.e., nasophar-yngitis and headache), but there were more serious adverseevents in the placebo group, mostly due to skin infections.Therapeutics designed for targeting IL-4 are limited so farto asthma and LP; however, speculative hypothesis may beput forward for other diseases (Table 2).

IL-4IL-31induction

PruritusReduced AMP

mobilization

Eosinophilchemoattraction

Increased IgEproduction

Enhanced DCgeneration

Epidermalbarrier

alteration

DefectiveS.Aureus killing

Enhancedallergen

penetration

Figure 1. Main activities of IL-4 in atopic dermatitis.AMP: Antimicrobial peptides; DC: Dendritic cells.

Therapeutic strategies in extrinsic atopic dermatitis

Expert Opin. Ther. Targets (2015) 19(1) 91

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

7. Conclusions

The identification of driver cytokines is a priority in the devel-opment of new therapeutic concepts against AD. TH2 immu-nity seems to be the end stream of a heterogeneous group ofconditions with the clinical phenotype of AD. In acute lesionsof AD, the TH2 cells produce IL-4, IL-13 and IL-31, whichmay potentiate barrier dysfunction and contribute to pruritus.In chronic lesions, a distinct T-cell infiltrate with TH1,TH17 and TH22 is present. However, many other cells areinvolved in the pathogenesis of AD, including TH9 cells, LCand IDEC, although the precise contribution of some ofthem remains to be fully understood. Preliminary data suggestthat blockade of IL-4 and IL-13 signaling may be an attractivetherapeutic target for AD.

8. Expert opinion: future place ofanti-IL-4-based strategies in the treatmentof AD

Adult patients with AD may be included in dermatologicareas with a high unmet medical need. Systemic agents forthe treatment of moderate-to-severe AD have not been devel-oped in the last two decades. Treatment options for refractoryAD consist of phototherapy and broad-spectrum immuno-suppressant, sometimes in an off-label basis. Some of thesedrugs, such as cyclosporine, cannot often be administeredfor a long time because of organ toxicity. However, ADresearch is entering a new era. Development in definingimmunogenetics and pathomechanisms of AD takes with itthe demand to understand better the clinical spectrum ofdisease and integrate phenotype with genetics. The biologicrevolution in therapy of inflammatory diseases is going toinvolve AD, bringing with it an urgent need for rationaldrug design based on known biological targets. A concertedeffort between academia, industry and patients will take for-ward the understanding of, and therapy for, AD.

AD and some types of asthma are characterized by theinduction of an immune response that is driven byTH2 cells. Besides intrinsic mechanisms, the nature of classicalallergens might drive the immune system into TH2 direction.IL-4 and IL-13 are key cytokines that are required for theinitiation and maintenance of this TH2 immune response.Consequently, such cytokines may be considered new andappealing therapeutic targets in AD. Phase I and II studieswith a monoclonal antibody that targets a subunit of theIL-4 and IL-13 receptors provide additional proofs for a keyrole of TH2 activation in the pathogenesis of AD. Theoreti-cally, IL-4 blocking could induce more significant improve-ment in extrinsic AD with IgE overproduction. However, byblocking IL-4 receptor a, signaling of both IL-4 and IL-13cytokines is modulated. As IL-13 appears to be a key cytokinein the pathogenesis of both extrinsic and intrinsic forms ofAD, the double signaling inhibition could be beneficial inintrinsic AD as well. Whereas it is arduous to predict theeffect of IL-4 and IL-13 inhibition on chronic, lichenifiedskin lesions of AD, which show fewer IL-4 mRNA, IL-13mRNA expressing cells and a distinct T-cell infiltrate withTH1, TH17 and TH22 dominance. Results of ongoingPhase III studies will be crucial in determining the clinicalutility of the targeting of IL-4 and IL-13 as well as the long-term effectiveness and tolerability of the treatment. Theycould help to identify disease phenotypes that can predicttreatment response. The advantages and weaknesses of block-ing IL-4 and IL-13 as compared with molecular targetshaving broader impact on the immune system, as cyclosporineand methotrexate should be proved. Neutralization of IL-4and IL-13 should theoretically improve skin barrier integrityand the expression of endogenous antimicrobial peptidespreventing dermatitis flares. Thus, ancillary analysis shouldassess patients for microbial colonization before and aftertreatment.

If on-going clinical trials will confirm the efficacy andsafety results of preliminary studies, an IL-4 receptor a inhib-itor is likely to become the first biologic therapy for refractoryadult AD.

Declaration of interest

The author has no relevant affiliations or financial involve-ment with any organization or entity with a financial interestin or financial conflict with the subject matter or materialsdiscussed in the manuscript. This includes employment,consultancies, honoraria, stock ownership or options, experttestimony, grants or patents received or pending, or royalties.

Table 2. Potential therapeutic applications for

IL-4 inhibitors.

Disease Evidence

Asthma Phase II trial [96]Adult atopic dermatitis Phase II trial [99]Systemic sclerosis Hypothesis [100]

Hyper IgE syndrome Hypothesis*Cardiac fibrosis Hypothesis [101]

*Suggested by the author.

V. Di Lernia

92 Expert Opin. Ther. Targets (2015) 19(1)

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Gittler JK, Shemer A, Suarez-Farinas M,

et al. Progressive activation of T(H)2/T

(H)22 cytokines and selective epidermal

proteins characterizes acute and chronic

atopic dermatitis. J Allergy

Clin Immunol 2012;130:1344-54

.. The authors describe a model of

progressive activation of

type 2 helper T (TH2) and type 22

helper T immune axes from the

acute-to-chronic phases of atopic

dermatitis (AD).

2. Naldi L, Parazzini F, Gallus S.

Prevalence of atopic dermatitis in Italian

schoolchildren: factors affecting its

variation. Acta Derm Venereol

2009;89:122-5

3. DaVeiga SP. Epidemiology of atopic

dermatitis: a review. Allergy Asthma Proc

2012;33:227-34

4. Eichenfield LF, Ellis CN, Mancini AJ,

et al. Atopic dermatitis: epidemiology

and pathogenesis update. Semin Cutan

Med Surg 2012;31(3 suppl):S3-5

5. Deckers IA, McLean S, Linssen S, et al.

Investigating international time trends in

the incidence and prevalence of atopic

eczema. 1990-2010: a systematic review

of epidemiological studies. PLoS One

2012;7:e39803

6. Leung DY. New insights into atopic

dermatitis: role of skin barrier and

immune dysregulation. Allergol Int

2013;62:151-61

. Insights into the complex relationship

between skin barrier and

immune abnormalities.

7. Elias PM, Schmuth M. Abnormal skin

barrier in the etiopathogenesis of atopic

dermatitis. Curr Allergy Asthma Rep

2009;9:265-72

.. Review on emerging evidence that the

inflammation in AD results from

inherited and acquired insults to

the barrier.

8. Bieber T. Atopic dermatitis. N Engl

J Med 2008;358:1483-94

9. Elias PM, Feingold KR. Does the tail

wag the dog? Role of the barrier in the

pathogenesis of inflammatory dermatoses

and therapeutic implications.

Arch Dermatol 2001;137:1079-81

10. Guttman-Yassky E, Nograles KE,

Krueger JG. Contrasting pathogenesis of

atopic dermatitis and psoriasis -- part II:

immune cell subsets and therapeutic

concepts. J Allergy Clin Immunol

2011;127:1420-32

11. Leung DY, Boguniewicz M,

Howell MD, et al. New insights into

atopic dermatitis. J Clin Invest

2004;113:651-7

12. Katsarou A, Armenaka M. Atopic

dermatitis in older patients: particular

points. J Eur Acad Dermatol Venereol

2011;25(1):12-18

13. Ozkaya E. Adult-onset atopic dermatitis.

J Am Acad Dermatol 2005;52:579-82

14. Novak N, Bieber T. Allergic and

nonallergic forms of atopic diseases.

J Allergy Clin Immunol 2003;112:252-62

15. Rhodes HL, Sporik R, Thomas P, et al.

Early life risk factors for adult asthma:

a birth cohort study of subjects at risk.

J Allergy Clin Immunol 2001;108:720-5

16. Lan S, Nickel R, Niggemann B, et al.

The development of childhood asthma:

lessons from the German Multicentre

Allergy Study (MAS).

Paediatr Respir Rev 2002;3:265-72

17. Spergel JM, Paller AS. Atopic dermatitis

and the atopic march. J Allergy

Clin Immunol 2003;112:S118-27

18. Simpson EL, Eichenfield LF, Ellis CN,

et al. Current issues in atopic

comorbidities and preventing the atopic

march. Semin Cutan Med Surg

2012;31:S6-9

19. Kiken DA, Silverberg NB. Atopic

Dermatitis in Children, Part 1:

epidemiology, Clinical Features, and

Complications. Cutis 2006;78:241-7

20. de Bruin Weller MS, Rockmann H,

Knulst AC, Bruijnzeel-Koomen CA.

Evaluation of the adult patient with

atopic dermatitis. Clin Exp Allergy

2013;43:279-91

21. Ellis CN, Mancini AJ, Paller AS, et al.

Understanding and managing atopic

dermatitis in adult patients. Semin Cutan

Med Surg 2012;31:S18-22

22. Ring J, Alomar A, Bieber T, et al.

Guidelines for treatment of atopic

eczema (atopic dermatitis) Part II. J Eur

Acad Dermatol Venereol

2012;26:1176-93

23. Deleuran MS, Vestergaard C. Therapy of

severe atopic dermatitis in adults. J Dtsch

Dermatol Ges 2012;10:399-406

24. Schram ME, Roekevisch E,

Leeflang MM, et al. A randomized trial

of methotrexate versus azathioprine for

severe atopic eczema. J Allergy

Clin Immunol 2011;128:353-9

25. Haeck IM, Knol MJ, Ten Berge O, et al.

Enteric-coated mycophenolate sodium

versus cyclosporin A as long-term

treatment in adult patients with severe

atopic dermatitis: a randomized

controlled trial. J Am Acad Dermatol

2011;64:1074-84

26. Darsow U, Wollenberg A, Simon D,

et al. Difficult to control atopic

dermatitis. World Allergy Organ J

2013;6:6

27. Oldhoff JM, Darsow U, Werfel T, et al.

Anti-IL-5 recombinant humanized

monoclonal antibody (Mepolizumab) for

the treatment of atopic dermatitis.

Allergy 2005;60:693-6

28. Belloni B, Ziai M, Lim A, et al.

Low-dose anti-IgE therapy in patients

with atopic eczema with high serum IgE

levels. J All Clini Immunol

2007;120:1223-5

29. Simon D, Hosli S, Kostylina G, et al.

Anti-CD20 (rituximab) treatment

improves atopic eczema. J Allergy

Clin Immunol 2008;121:122-8

30. Ponte P, Lopes MJ. Apparent safe use of

single dose rituximab for recalcitrant

atopic dermatitis in the first trimester of

a twin pregnancy. J Am Acad Dermatol

2010;63:355-6

31. Sanchez-Ramon S, Eguıluz-Gracia I,

Rodrıguez-Mazariego ME, et al.

Sequential combined therapy with

omalizumab and rituximab: a new

approach to severe atopic dermatitis.

J Investig Allergol Clin Immunol

2013;23:190-6

32. Navarini AA, French LE, Hofbauer DF.

Interrupting IL-6-receptor signaling

improves atopic dermatitis but associates

with bacterial superinfection. J All

Clin Immunol 2011;128:1128-30

33. Bao L, Shi VY, Chan LS. IL-4 up-

regulates epidermal chemotactic,

angiogenic, and pro-inflammatory genes

and down-regulates antimicrobial genes

in vivo and in vitro: relevant in the

Therapeutic strategies in extrinsic atopic dermatitis

Expert Opin. Ther. Targets (2015) 19(1) 93

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

pathogenesis of atopic dermatitis.

Cytokine 2013;61:419-25

. The authors identify keratinocyte genes

regulated by IL-4.

34. Rodriguez E, Baurecht H, Herberich E,

et al. Meta-analysis of filaggrin

polymorphisms in eczema and asthma:

robust risk factors in atopic disease.

J Allergy Clin Immunol

2009;123:1361-70

.. Meta-analysis summarizing the strong

evidences for a high AD risk conferred

by filaggrin mutations.

35. Irvine AD. Fleshing out filaggrin

phenotypes. J Invest Dermatol

2007;127:504-7

36. Hershey GK, Friedrich MF, Esswein LA,

et al. The association of atopy with a

gain-of-function mutation in the alpha

subunit of the interleukin-4 receptor.

N Engl J Med 1997;337:1720-5

37. Ring J, M€ohrenschlager M, Weidinger S.

Molecular genetics of atopic eczema.

2012;96:24-9

38. Chien YH, Hwu WL, Chiang BL. The

genetics of atopic dermatitis. Clin Rev

Allergy Immunol 2007;33:178-90

39. Herberth G, Heinrich J, R€oder S, et al.

Reduced IFN-gamma- and enhanced

IL-4-producing CD4+ cord blood T cells

are associated with a higher risk for

atopic dermatitis Pediatr Allergy

Immunol. 2010;21:5-13

40. Furue M, Ogata F, Ootsuki M,

Ishibashi Y. Hyperresponsibility to

exogeneous interleukin 4 in atopic

dermatitis. J Dermatol 1989;16:247-50

41. Okudaira H, Mori A, Akiyoshi K, et al.

Interleukin-4 gene expression and IgE

responsiveness. Int Arch Allergy

Appl Immunol 1991;94:184-6

42. Reinhold U, Kukel S, Goeden B, et al.

Interleukin-4 promotes the expansion of

skin-infiltrating lymphocytes from atopic

dermatitis in vitro. J Invest Dermatol

1991;96:370-5

43. Dubois GR, Bruijnzeel-Koomen CA,

Bruijnzeel PL. IL-4 induces chemotaxis

of blood eosinophils from atopic

dermatitis patients, but not from normal

individuals. J Invest Dermatol

1994;102:843-6

44. Moser R, Fehr J, Bruijnzeel PL.

IL-4 controls the selective endothelium-

driven transmigration of eosinophils from

allergic individuals. J Immunol

1992;149:1432-8

45. Renz H, Jujo K, Bradley KL, et al.

Enhanced IL-4 production and IL-4

receptor expression in atopic dermatitis

and their modulation by

interferon-gamma. J Invest Dermatol

1992;99:403-8

46. Hamid Q, Boguniewicz M, Leung DY.

Differential in situ cytokine gene

expression in acute versus chronic atopic

dermatitis. J Clin Invest 1994;94:870-6

47. Hamid Q, Naseer T, Minshall EM, et al.

In vivo expression of IL-12 and IL-13 in

atopic dermatitis. J Allergy

Clin Immunol 1996;98:225-31

48. Gahr N, F€olster-Holst R,

Weichenthal M, et al. Dermal fibroblasts

from acute inflamed atopic dermatitis

lesions display increased eotaxin/

CCL11 responsiveness to inteleukin-4

stimulation. Br J Dermatol

2011;164:586-92

49. Bao L, Shi VY, Chan LS. IL-4 regulates

chemokine CCL26 in keratinocytes

through the Jak1, 2/Stat6 signal

transduction pathway: implication for

atopic dermatitis. Mol Immunol

2012;50:91-7

50. Seneviratne SL, Black AP, Jones L, et al.

Interleukin-4 promotes human CD8 T

cell expression of CCR7. Immunology

2007;120:66-72

51. Novak H, Muller A, Harrer N, et al.

CCL23 expression is induced by IL-4 in

a STAT6-dependent fashion. J Immunol

2007;177:4335-41

52. Wollenberg A, Kraft S, Hanau D,

Bieber T. Immunomorphological and

ultrastructural characterization of

Langerhans cells and a novel,

inflammatory dendritic epidermal cell

(IDEC) population in lesional skin of

atopic eczema. J Invest Dermatol

1996;106:446-53

53. Novak N, Kraft S, Haberstok J, et al.

A reducing microenvironment leads to

the generation of FcepsilonRI high

inflammatory dendritic epidermal cells

(IDEC). J Invest Dermatol

2002;119:842-9

54. Sonkoly E, Muller A, Lauerma AI, et al.

IL-31: a new link between T cells and

pruritus in atopic skin inflammation.

J Allergy Clin Immunol

2006;117:411-17

. A link among staphylococcal

colonization, T-cell recruitment and

activation, and pruritus induction in

patients with AD is provided in this

article.

55. Kasraie S, Niebuhr M, Baumert K,

Werfel T. Functional effects of

interleukin 31 in human primary

keratinocytes. Allergy 2011;66:845-52

56. Stott B, Lavender P, Lehmann S, et al.

Human IL-31 is induced by IL-4 and

promotes TH2-driven inflammation.

J Allergy Clin Immunol

2013;132:446-54

57. Irvine AD, McLean WH, Leung DY.

Filaggrin mutations associated with skin

and allergic diseases. N Engl J Med

2011;365:1315-27

58. Hudson TJ. Skin barrier function and

allergic risk. Nat Genet 2006;38:399-400

. The article underlines the importance

of the epidermal barrier in preventing

sensitization to allergens.

59. Elias PM, Hatano Y, Williams ML. Basis

for the barrier abnormality in atopic

dermatitis: outside-inside-outside

pathogenic mechanisms. J Allergy

Clin Immunol 2008;121:1337-43

.. Review assessing the emerging evidence

that inflammation in AD results from

inherited and acquired insults to

the barrier.

60. Henderson J, Northstone K, Lee SP,

et al. The burden of disease associated

with filaggrin mutations: a population-

based, longitudinal birth cohort study.

J Allergy Clin Immunol 2008;121:872-7

61. Howell MD, Kim BE, Gao P, et al.

Cytokine modulation of atopic dermatitis

filaggrin skin expression. J Allergy

Clin Immunol 2007;120:150-5

.. Article showing how underlying

inflammation can alter the expression

of genes that are involved in

epidermal-barrier function.

62. Ogg G. Role of T cells in the

pathogenesis of atopic dermatitis.

Clin Exp Allergy 2009;39:310-16

63. Omori-Miyake M, Yamashita M,

Tsunemi Y, et al. In vitro assessment of

IL-4- or IL-13-mediated changes in the

structural components of keratinocytes in

mice and humans. J Invest Dermatol

2014;134:1342-50

64. Kim BE, Leung DY, Boguniewicz M,

Howell MD. Loricrin and involucrin

expression is down-regulated by

V. Di Lernia

94 Expert Opin. Ther. Targets (2015) 19(1)

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

Th2 cytokines through STAT-6.

Clin Immunol 2008;126:332-7

. This article shows that TH2 cytokines

may inhibit expression of important

proteins for skin barrier formation

and integrity.

65. Hatano Y, Terashi H, Arakawa S,

Katagiri K. Interleukin-4 suppresses the

enhancement of ceramide synthesis and

cutaneous permeability barrier functions

induced by tumor necrosis factor-alpha

and interferon-gamma in human

epidermis. J Invest Dermatol

2005;124:786-92

. This study investigates how a balance

between TH1 and TH2 might affect the

repair of the epidermal barrier via

regulation of the production

of ceramide.

66. Hatano Y, Adachi Y, Elias PM, et al.

The Th2 cytokine, interleukin-4,

abrogates the cohesion of normal stratum

corneum in mice: implications for

pathogenesis of atopic dermatitis.

Exp Dermatol 2013;22:30-5

67. Morizane S, Yamasaki K, Kajita A, et al.

TH2 cytokines increase kallikrein

7 expression and function in patients

with atopic dermatitis. J All

Clin Immunol 2012;130:259-61

68. Elias PM, Steinhoff M.

Outside-to-Inside” (and now back to

“outside”) pathogenic mechanisms in

atopic dermatitis. J Invest Dermatol

2008;128:1067-70

69. Harder J, Bartels J, Christophers E,

Schroder JM. Isolation and

characterization of human beta -defensin-

3, a novel human inducible peptide

antibiotic. J Biol Chem

2001;276:5707-13

. The authors discovery a novel human-

inducible, epithelial antimicrobial

peptide confirming the hypothesis that

such peptides represent an integral

part in the innate immunity of

human epithelia.

70. Duits LA, Ravensbergen B,

Rademaker M, et al. Expression of beta-

defensin 1 and 2 mRNA by human

monocytes, macrophages and dendritic

cells. Immunology 2002;106:517-25

71. Yang D, Chertov O, Bykovskaia SN,

et al. Beta-defensins: linking innate and

adaptive immunity through dendritic and

T cell CCR6. Science 1999;286:525-8

.. The authors prove that b-defensins

may promote adaptive immune

responses by recruiting dendritic and T

cells to the site of microbial invasion.

72. Oppenheim JJ, Biragyn A, Kwak LW,

Yang D. Roles of antimicrobialpeptides

such as defensins in innate and adaptive

immunity. Ann Rheum Dis

2003;62(Suppl 2):ii17-21

73. Schauber J, Gallo RL. Antimicrobial

peptides and the skin immune defense

system. J Allergy Clin Immunol

2009;124:R13-18

74. Ong PY, Ohtake T, Brandt C, et al.

Endogenous antimicrobial peptides and

skin infections in atopic dermatitis.

N Engl J Med 2002;347:1151-60

75. Nomura I, Goleva E, Howell MD, et al.

Cytokine milieu of atopic dermatitis, as

compared to psoriasis, skin prevents

induction of innate immune response

genes. J Immunol 2003;171:3262-9

76. Howell MD, Novak N, Bieber T, et al.

Interleukin-10 downregulates anti-

microbial peptide expression in atopic

dermatitis. J Invest Dermatol

2005;125:738-45

77. Yamasaki K, Schauber J, Coda A, et al.

Kallikrein-mediated proteolysis regulates

the antimicrobial effects of cathelicidins

in skin. FASEB J 2006;20:2068-80

78. Boguniewicz M, Leung DY. Recent

insights into atopic dermatitis and

implications for management of

infectious complications. J Allergy

Clin Immunol 2010;125(1):4-13

79. Baker BS. The role of microorganisms in

atopic dermatitis. Clin Exp Immunol

2006;144:1-9

80. Eyerich K, Pennino D, Scarponi C, et al.

IL17 in atopic eczema: linking allergen-

specific adaptive and microbial triggered

innate immune response. J Allergy

Clin Immunol 2009;123:59-66

. This study shows interesting

relationships between IL-17 and

microbial innate immune response.

81. Miajlovic H, Fallon PG, Irvine AD,

Foster TJ. Effect of filaggrin breakdown

products on growth of and protein

expression by Staphylococcus aureus.

J Allergy Clin Immunol

2010;126:1184-90

82. Kisich KO, Carspecken CW, Fieve S,

et al. Defective killing of Staphylococcus

aureus in atopic dermatitis is associated

with reduced mobilization of human

beta-defensin-3. J Allergy Clin Immunol

2008;122:62-8

83. Aberg KM, Man MQ, Gallo RL, et al.

Co-regulation and interdependence of

the mammalian epidermal permeability

and antimicrobial barriers.

J Invest Dermatol 2008;128:917-25

84. Kabashima-Kubo R, Nakamura R,

Sakabe J, et al. A group of atopic

dermatitis without IgE elevation or

barrier impairment shows a high

Th1 frequency: possible immunological

state of the intrinsic type. J Dermatol Sci

2012;67:37-43

. This study shows that AD patients

with IgE-low have much less filaggrin

mutations and increased frequency of

TH1 cells with respect to the group of

patients with extrinsic AD.

85. Tokura Y. Extrinsic and intrinsic types of

atopic dermatitis. J Dermatol Sci

2010;58:1-7

86. Kagi MK, Wuthrich B, Montano E,

et al. Differential cytokine profiles in

peripheral blood lymphocyte supernatants

and skin biopsies from patients with

different forms of atopic dermatitis,

psoriasis and normal individuals. Allergy

2002;57:431-5

87. Suarez-Farinas M, Dhingra N, Gittler J,

et al. Intrinsic atopic dermatitis shows

similar TH2 and higher TH17 immune

activation compared with extrinsic atopic

dermatitis. J Allergy Clin Immunol

2013;132:361-70

88. Jeong CW, Ahn KS, Rho NK, et al.

Differential in vivo cytokine

mRNA expression in lesional skin of

intrinsic vs. extrinsic atopic dermatitis

patients using semiquantitative RT-PCR.

Clin Exp Allergy 2003;33:1717-24

89. von Kobyletzki G, Pieck C,

H€oxtermann S, et al. Circulating

activation markers of severe atopic

dermatitis following ultraviolet A1 cold

light phototherapy: eosinophil cationic

protein, soluble interleukin-2 receptor

and soluble interleukin-4 receptor.

Br J Dermatol 1999;140:966-8

90. Choi JJ, Park BK, Song GY, et al.

Establishment of an in vitro test system

to evaluate the down-regulatory activities

of natural products on IL-4.

Arch Pharm Res 2007;30:1102-10

91. Crocker IC, Townley RG, Khan MM.

Phosphodiesterase inhibitors suppress

proliferation of peripheral blood

mononuclear cells and interleukin-4 and

-5 secretion by human T-helper

Therapeutic strategies in extrinsic atopic dermatitis

Expert Opin. Ther. Targets (2015) 19(1) 95

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015

type 2 cells. J Invest Dermatol

1996;107:51-6

92. Hanifin JM, Chan SC, Cheng JB, et al.

Type 4 phosphodiesterase inhibitors have

clinical and in vitro anti-inflammatory

effects in atopic dermatitis.

Clin Exp Med 2012;12:241-5

93. Panahi Y, Davoudi SM, Madanchi N,

Abolhasani E. Recombinant human

interferon gamma in treatment of atopic

dermatitis. Clin Exp Med 2012;12:241-5

94. Chang TT, Stevens SR. Atopic

dermatitis: the role of recombinant

interferon-gamma therapy. Am J

Clin Dermatol 2002;3:175-83

95. Morioka T, Yamanaka K, Mori H, et al.

IL-4/IL-13 antagonist DNA vaccination

successfully suppresses Th2 type chronic

dermatitis. Br J Dermatol

2009;160:1172-9

96. Wenzel S, Ford L Pearlman D, et al.

Dupilumab in persistent asthma with

elevated eosinophil levels. New Engl

J Med 2013;368:2455-66

97. Hijnen D, De Bruin-Weller M,

Oosting B, et al. Serum thymus and

activation-regulated chemokine (TARC)

and cutaneous T-cell-attracting

chemokine (CTACK) levels in allergic

diseases: TARC and CTACK are disease-

specific markers for atopic dermatitis.

J Allergy Clin Immunol

2004;113:334-40

98. Kimura T, Sugaya M, Suga H, et al.

Variations in serum TARC and I-TAC

levels reflect minor changes in disease

activity and pruritus in atopic dermatitis.

Acta Derm Venereol 2014;94(3):331-2

99. Beck LA, Thaci D, Hamilton JD, et al.

Dupilumab treatment in adults with

moderate-to-severe atopic dermatitis.

N Engl J Med 2014;371:130-9

.. This study gives reliable evidence for a

key role of TH2 activation in the

pathogenesis of AD. Results of the

trials show a remarkable efficacy of

Dupilumab in the short-term

treatment of AD.

100. Lafyatis R. Targeting fibrosis in systemic

sclerosis. Endocr Metab Immune Disord

Drug Targets 2006;6:395-400

101. Kanellakis P, Ditiatkovski M,

Kostolias G, Bobik A. A pro-fibrotic role

for interleukin-4 in cardiac pressure

overload. Cardiovasc Res

2012;95(1):77-85

AffiliationVito Di Lernia MD

Arcispedale Santa Maria Nuova-IRCCS,

Dermatology Unit, viale Risorgimento 68,

42123 Reggio Emilia, Italy

V. Di Lernia

96 Expert Opin. Ther. Targets (2015) 19(1)

Dow

nloa

ded

by [

Vito

Di L

erni

a] a

t 08:

13 2

9 N

ovem

ber

2015


Recommended