Advances in the diagnosis and therapeutic management of atopic dermatitis

13
REVIEW ARTICLE Advances in the Diagnosis and Therapeutic Management of Atopic Dermatitis Christian Vestergaard Mette Deleuran Published online: 13 May 2014 Ó Springer International Publishing Switzerland 2014 Abstract Atopic dermatitis is a very prevalent disease that affects children as well as adults. The disease has a severe impact on quality of life for the patients and their families. The skin in atopic dermatitis patients is a site of both a severe inflammatory reaction dominated by lym- phocytes and decreased skin barrier function. The treat- ment of the disease is mainly aimed at reducing the inflammation in the skin and/or restoring the skin barrier function. However, most of the treatments used today singularly aim at reducing the inflammation in the skin. Depending on the severity of the disease, the anti-inflam- matory treatment may be topical or systemic, but basic treatment, no matter the severity, should always be emol- lients. In addition, new studies have shown good effects of psychosocial interventions, such as eczema schools, for patients and their families. This review covers the latest trends in the treatment of atopic dermatitis. Key Points Atopic dermatitis is both an inflammatory skin disease and a disease of the skin barrier function. Anti-inflammatory treatment is topical but systemic therapy may be added in severe cases. Treatment should always be combined with skin barrier regeneration (moisturizer), and occasionally with antibacterials. New emerging biological therapies for atopic dermatitis are promising. 1 Introduction Atopic dermatitis (AD) is a chronic or chronically relaps- ing eczematous disease that affects between 15 and 20 % of all children in affluent countries [1]. The classical symptoms are intense pruritus in the affected areas, often with clinically visible eczema, typically in the flexural areas of the body [2]. The lesions may be complicated with bacterial super-infection with Staphylococcus aureus or viral infections such as Herpes simplex virus [3]. In 60 % of paediatric cases, the onset of the disease is before the age of 1 year and in 85 % of the cases before the age of 5 years [4]. Apart from the impact on the patient, the disease affects the quality of life of the patient’s family to the same degree as having a child with diabetes mellitus [5]. Classically, the diagnosis of AD is made on the basis of the Hanifin and Rajka [6] criteria from 1980, and it is a clinical diagnosis. There is a long-standing discussion on whether IgE-mediated allergies are part of the pathogenesis C. Vestergaard Á M. Deleuran (&) Department of Dermatology, Aarhus University Hospital, P.P. Ørumsgade 11, 8000 Aarhus C, Denmark e-mail: [email protected] Drugs (2014) 74:757–769 DOI 10.1007/s40265-014-0219-3

Transcript of Advances in the diagnosis and therapeutic management of atopic dermatitis

REVIEW ARTICLE

Advances in the Diagnosis and Therapeutic Managementof Atopic Dermatitis

Christian Vestergaard • Mette Deleuran

Published online: 13 May 2014

� Springer International Publishing Switzerland 2014

Abstract Atopic dermatitis is a very prevalent disease

that affects children as well as adults. The disease has a

severe impact on quality of life for the patients and their

families. The skin in atopic dermatitis patients is a site of

both a severe inflammatory reaction dominated by lym-

phocytes and decreased skin barrier function. The treat-

ment of the disease is mainly aimed at reducing the

inflammation in the skin and/or restoring the skin barrier

function. However, most of the treatments used today

singularly aim at reducing the inflammation in the skin.

Depending on the severity of the disease, the anti-inflam-

matory treatment may be topical or systemic, but basic

treatment, no matter the severity, should always be emol-

lients. In addition, new studies have shown good effects of

psychosocial interventions, such as eczema schools, for

patients and their families. This review covers the latest

trends in the treatment of atopic dermatitis.

Key Points

Atopic dermatitis is both an inflammatory skin

disease and a disease of the skin barrier function.

Anti-inflammatory treatment is topical but systemic

therapy may be added in severe cases. Treatment

should always be combined with skin barrier

regeneration (moisturizer), and occasionally with

antibacterials.

New emerging biological therapies for atopic

dermatitis are promising.

1 Introduction

Atopic dermatitis (AD) is a chronic or chronically relaps-

ing eczematous disease that affects between 15 and 20 %

of all children in affluent countries [1]. The classical

symptoms are intense pruritus in the affected areas, often

with clinically visible eczema, typically in the flexural

areas of the body [2]. The lesions may be complicated with

bacterial super-infection with Staphylococcus aureus or

viral infections such as Herpes simplex virus [3]. In 60 %

of paediatric cases, the onset of the disease is before the

age of 1 year and in 85 % of the cases before the age of 5

years [4]. Apart from the impact on the patient, the disease

affects the quality of life of the patient’s family to the same

degree as having a child with diabetes mellitus [5].

Classically, the diagnosis of AD is made on the basis of

the Hanifin and Rajka [6] criteria from 1980, and it is a

clinical diagnosis. There is a long-standing discussion on

whether IgE-mediated allergies are part of the pathogenesis

C. Vestergaard � M. Deleuran (&)

Department of Dermatology, Aarhus University Hospital,

P.P. Ørumsgade 11, 8000 Aarhus C, Denmark

e-mail: [email protected]

Drugs (2014) 74:757–769

DOI 10.1007/s40265-014-0219-3

of AD and this debate is even more pertinent after the

finding of the importance of filaggrin mutations as the basis

of approximately one-third of all AD patients in Northern

Europe, but with much higher variations in the rest of the

world [7, 8].

The severity of AD dictates the level of treatment. In

mild to moderate cases, topical immunomodulators or

steroids may be used both in a proactive manner to prevent

flare-ups, and as treatment of acute eczema. In moderate to

severe cases, systemic immunomodulatory therapy may be

necessary even in children [9]. However, in all cases the

basic therapy is moisturizers to alleviate the dry skin and

the impaired skin barrier function [10].

This review will discuss the latest trends in diagnosis,

pathogenesis and treatments of AD.

2 The Diagnosis of Atopic Dermatitis (AD)

In the clinic the diagnosis of AD is made on the basis of

The Hanifin and Rajka [6] criteria from 1980 (Table 1), of

which the patient must have at least three major and three

minor criteria. The distribution of the eczema depends on

the age of the patient. In the infantile stage, the face, the

scalp and the extensor sides of the extremities are typically

affected whereas in the childhood phase the eczema is

typically located in the flexural folds of the extremities as

well as the hands and the face. In teenagers and adults the

flexural areas are still the most common sites but the hands

and feet are often involved as well as the face and neck

[11]. The latter is often described as head and neck der-

matitis and can be a sign of sensitisation to Malassezia

furfur, which is a skin commensal [12]. In severe cases, the

entire skin organ may be involved and the patients may

develop erythroderma. The affected skin shows a typical

eczema with a maculopapular rash, with or without vesi-

cles, desquamation and excoriations [1]. In certain cases,

the eczema may present itself as a nummular eczema with

sharply demarcated edges, as a pityriasiform lichenoid

eczema or as a follicular-type AD, characterized by plaque-

shaped, lichenoid desquamation eczema, which is common

in the Japanese population [13]. Post-inflammatory hypo-

pigmentation is often seen in healed skin in fair skin types

and hyperpigmentation in darker skin types.

A hallmark of AD, and one of the minor criterion in the

Hanifin and Rajka criteria, is dry skin or xerosis, though the

prevalence varies between 48 and 100 % compared with

14–40 % in healthy controls [11, 14]. Furthermore, sweat-

induced itch, skin reactions upon food intake, hand eczema,

positive skin prick testing, facial erythema and disease

activity influenced by environmental factors are more

prevalent among patients with AD than among healthy

controls [11]. Other signs of AD that are not included in the

Hanifin and Rajka criteria are earlobe rhagades (fissures at

the base of the earlobes), atopic winter feet (juvenile

plantar dermatitis), and retro-auricular fissuring.

3 Pathogenesis of AD

Until recently, AD has been perceived as a disease driven

by T helper 2 (Th2; lymphocytes that express interleukin

[IL]-4, IL-5, IL-6 and IL-10), based on the IL-4-dominated

response achieved when peripheral blood mononuclear

cells (PBMCs) from AD patients are stimulated with, for

example, lipopolysaccharide (LPS) [15]. This perception

was supported by the fact that approximately 80 % of all

AD patients had increased serum concentrations of

immunoglobulin E (IgE) [16]. However, over the last

decade this has changed immensely. The perception of IgE

as a central molecule in the pathogenesis has been chal-

lenged and it has been suggested that it may just be an

epiphenomenon to the severe inflammation taking place

[8]. The Th2 paradigm has also been questioned as a result

of animal experiments, which show that neither Th2 lym-

phocytes nor the Th2 pathway are a prerequisite for AD-

like eczema [17, 18]. Lastly, the skin barrier function has

drawn much more attention after the milestone publication

showing that lack of filaggrin, due to loss of function

mutations in the FLG gene, increased the risk of having AD

13.4-fold [19].

3.1 Inflammation of the Skin in AD

Classically, the inflammation in AD is described as a

biphasic response with an initial Th2-dominated cytokine

profile; for example, high production of IL-4, IL-5 and IL-

13 followed by a mixed Th1/Th2 response (e.g., additional

production of IL-2 and interferon-c [IFN-c]) [20]. IL-22

that originates from the Th22 lymphocytes has been

implied in the acute phase of AD as it increases the epi-

dermal growth but down-regulates the skin barrier function

[21, 22], along with IL-31, which also induces pruritus

[23]. Furthermore, the keratinocytes play an active role in

the production of inflammatory signals, as they produce the

skin-specific CC-chemokine-ligand 27 (CCL27) and the

Th2 inducing CCL17 [24, 25]. The latter can also be

induced in dermal dendritic cells by thymic stromal lym-

phopoietin (TSLP), which is also produced by the kerati-

nocytes. Another cytokine produced by the dendritic cells

in the dermis of AD patients is IL-25 (IL-17E) [26]. This

cytokine is known to induce a Th2 response and has been

found increased not only in AD patients, but also in the

airway epithelium of asthma patients. Interestingly, IL-25

is released from mast cells when the FceRI receptors are

cross-linked by antigens binding to IgE [27]. This may

758 C. Vestergaard, M. Deleuran

provide a link between food allergies and the flare-ups of

AD.

IL-33 is a member of the IL-1 family, and is described

as a member of the ‘alarmin’ family [28]. It is released

from keratinocytes upon mechanical stimuli, and binds to

the ST2 receptors that are expressed on mast cells and Th2

lymphocytes, which suggests that IL-33 may be involved in

the early events of AD [29, 30].

In the chronic stages of AD, a more mixed response can

be observed. As already mentioned, more Th1 lymphocytes

can be observed in the skin. The expression of IFN-c is

preceded by IL-12, a cytokine that can be induced in

monocytes by IL-4 [31]. However, recent results have

demonstrated that Th17 lymphocytes, which are usually

associated with psoriasis, are part of the T lymphocyte

infiltrate found in chronic lesions of AD skin [32]. IL-17 is

described as a master regulator of antimicrobial peptides

(AMPs) and may be a key to the decreased expression of

AMPs in AD skin, and the following increased numbers of

skin infections [33].

3.2 Barrier Function and Filaggrin

AD patients have a decreased skin barrier that leads to an

increased trans-epidermal water loss (TEWL) [34, 35] but

also to an increased risk for the patient to acquire allergies

[36]. The composition of the skin barrier is very complex,

but one of the major constituents of the barrier is the out-

ermost layer of the epidermis, the stratum corneum. The

keratinocytes in this layer have undergone apoptosis,

excluded their nuclei and have become completely filled

with keratin [37]. Keratin filaments are tightly cross-linked

by filaggrin molecules and thus a lack of filaggrin leads to a

poorer aggregation of keratin. Filaggrin is encoded by the

FLG gene as profilaggrin, which is made up by 8–12 fil-

aggrin repeats [38]. Heavy phosphorylation protects

Table 1 The Hanifin and Rajka

criteria for the diagnosis of

atopic dermatitis [6]

Must have three or more basic features:

Pruritus

Typical morphology and distribution:

Flexural lichenification or linearity in adults

Facial and extensor involvement in infants and children

Chronic or chronically relapsing dermatitis

Personal or family history of atopy (asthma, allergic rhinitis, atopic dermatitis)

Plus three or more minor features:

Xerosis

Ichthyosis/palmar hyperlinearity/keratosis pilaris

Immediate (type I) skin test reactivity

Elevated serum IgE

Early age of onset

Tendency towards cutaneous infections (esp. Staphylococcus aureus and Herpes simplex)/impaired cell-

mediated immunity

Tendency towards non-specific hand or foot dermatitis

Nipple eczema

Cheilitis

Recurrent conjunctivitis

Dennie–Morgan infraorbital fold

Keratoconus

Anterior subcapsular cataracts

Orbital darkening

Facial pallor/facial erythema

Pityriasis alba

Anterior neck folds

Itch when sweating

Intolerance to wool and lipid solvents

Perifollicular accentuation

Food intolerance

Course influenced by environmental/emotional factors

White dermographism/delayed blanch

Diagnosis and Management of Atopic Dermatitis 759

filaggrin from breakdown [39] until it reaches the stratum

granulosum, in which several proteases including matrip-

tase, prostasin, as well as caspase 14 are situated [40–42].

In 2006, Palmer et al. [19] described the significantly

increased risk (OR 13.4) of developing AD in patients

heterozygous for loss of function mutations in the FLG

gene. Filaggrin mutations exist globally but they vary from

population to population. Although defects in this gene to

date have the best correlation with developing AD,

between 44 and 85 % of patients with AD do not have

mutations in the filaggrin gene [34].

Inflammation itself is also able to induce functional

filaggrin defects through down-regulation of filaggrin

production or filaggrin maturation [43]. IL-4 and Il-13,

both classical Th2-type cytokines, down-regulate filaggrin

on a transcriptional level [44]. IL-25, which may be con-

sidered as a Th2-inducing cytokine, also down-regulates

filaggrin expression on a transcriptional level [26]. IL-22

that is produced by skin homing T lymphocytes (Th22) and

dendritic cells from the skin down-regulate filaggrin and

profilaggrin on both mRNA and protein levels [21]. IL-31,

which is produced mainly by Th2 lymphocytes but also

mast cells, dendritic cells and monocytes, is strongly

associated with itch, since blocking of this cytokine may

alleviate this symptom [23]. IL-31 also induces IL-20 and

IL-24 in HaCaT cells and through this down-regulates fil-

aggrin expression [45]. Cytokines typically associated with

a Th1 response, such as tumour necrosis factor (TNF)-a[46] and IL-17A, also down-regulate filaggrin [47].

4 Treatment of AD

The main goal of therapy for AD is relief from the itch and

possibly inhibition of the inflammatory reaction in the skin.

Treatment modalities can be divided into basic therapy,

topical therapy and systemic therapy. Basic therapy

includes non-pharmacological therapies such as emollients

and baths, and avoidance strategies of both specific and

non-specific provoking factors. Topical therapy includes

treatment with topical glucocorticoids and topical calci-

neurin inhibitors (TCIs) and this may be used either as

acute therapy or as proactive treatment. Systemic therapy

includes the use of immunosuppressants such as systemic

glucocorticoids, azathioprine, cyclosporine A, methotrex-

ate, and in some instances mycophenolate. Phototherapy

and coal tar baths are classic dermatological treatments and

do have an effect in AD. Biologics have so far not been

registered for the treatment of AD, as they have in psori-

asis, but early studies indicate that this may happen in the

future. Other modalities such as psychosomatic counselling

and education through eczema schools also have increasing

evidence and are recommended.

4.1 Basic Treatment

4.1.1 Emollients

Emollients are the mainstay of therapy in AD and should

be applied concomitantly with any other therapy chosen.

Although the mechanism of action for emollients is largely

unknown, several studies have shown that application of

emollients, both in the short and long term, have a steroid-

sparing effect [48]. However, the use of emollients only

without specific treatment of the inflammatory reaction

may increase the risk of disseminating skin infections [49].

Emollients containing intact proteins such as oats and

peanut may increase the risk of allergies [50, 51].

4.2 Topical Treatment

The use of topical treatment is often the first choice of

pharmacological treatment in AD, either with glucocorti-

coids or calcineurin inhibitors. The effect of the drug is

determined by three factors: the strength of the drug, the

dosage of the drug, and the application of the drug [52].

Dosing of topical treatment might seem difficult but when

using the fingertip unit (FTU) it is quite simple. One FTU

equals the amount of ointment/cream that can be expressed

from the distal crease of the index finger to the fingertip

(approximately 0.5 g of cream). This covers the body

surface area corresponding to two palms.

Topical treatment can be applied on a daily basis during

acute flares, and twice or thrice weekly as a proactive

treatment during periods of remission.

4.2.1 Topical Glucocorticoids

Glucocorticoids partly exert their action through binding to

steroid receptors in the cytosol of the cell. The receptor

complex then translocates into the nucleus of the cell,

where it inhibits the expression of inflammatory cytokines

(transrepression), induces the expression of anti-inflam-

matory cytokines (transactivation) and inhibits the pro-

duction of structural cytokines [53]. Topical steroids are

grouped according to potency into groups I–IV (Europe)

and groups I–VII (US), and are effective against inflam-

mation in the skin. In Europe, group I corresponds to the

products with the weakest potency. This is in contrast to

the US, where group I topical corticosteroids corresponds

to the products with the strongest potency. The potency

reflects both the effect of the topical treatment but also the

risk of side effects. One well performed treatment per day,

compared with two, is sufficient to obtain effect of the

treatment [54]. Once the symptoms are controlled, treat-

ment should be tapered either through decrease of appli-

cation frequency or of potency of the glucocorticoid.

760 C. Vestergaard, M. Deleuran

Proactive treatment with application of fluticasone propi-

onate twice weekly during remission periods significantly

reduced flare-ups of the eczema [55].

The use of high-potency steroids increases the risk for

systemic side effects, although the risk for hypothalamus-

pituitary-adrenal axis suppression is very low [56]. How-

ever, high-potency steroids also restore the skin barrier

significantly faster than low-potency steroids, and thus

shorter treatment periods are needed [57]. The restoration

of the skin barrier through glucocorticoids is probably due

to the inhibition of the skin-disrupting effect of inflam-

mation, as shown by betamethasone valerate, which by

itself actually inhibits rate-limiting enzymes for lipid syn-

thesis [58].

4.2.2 Topical Calcineurin Inhibitors

Tacrolimus and pimecrolimus are macrolides that exert

immunosuppression through inhibition of the calcium-

dependent dephosphorylation of the transcription factor

nuclear factor of activated T cells (NFAT) that is required for

the transcription of inflammatory cytokines such as IL-2 [59].

Both have efficacy in both long- [60, 61] and short-term [62,

63] studies, and have shown a high degree of efficacy in

proactive treatment over a 1-year period with regard to

reduction of both severity and number of flare-ups. The

efficacy of 0.1 % tacrolimus equals that of a corticosteroid of

medium potency [64], whereas pimecrolimus is weaker [65].

There have been controversies over the safety of the drugs,

especially with regard to the risk of non-melanoma skin

cancer and lymphoma. However, a follow-up study over

6 years has shown no increased risk of lymphoma after the

use of TCIs and the photocarcinogenic effect of the drugs has

also been examined and found to be non-existent when used

topically [66]. However, since the systemic use of calcineurin

inhibitors in solid organ transplant patients significantly

increases the risk for non-melanoma skin cancer, the use of

sunscreens is still recommended when using these drugs [64].

4.3 Antibacterials and Antiseptics

Most patients with AD are colonized with S. aureus on

both affected and unaffected skin [67]. A systematic review

found no clear evidence of benefit from the use of anti-

bacterial soaps, antibacterial bath additives or topical

antibacterials/antiseptics [68]. The studies included in the

review were, however, quite small. A randomized con-

trolled trial (RCT) in children with AD showed beneficial

effects of intranasal mupirocin ointment combined with

baths containing sodium hypochlorite (bleach baths) on

disease activity in children [69]. Oral antibacterials are not

recommended when the skin is not clinically infected, but

should be used as a short-term treatment for clinically

super-infected AD with oozing, crusts, pustules and/or

fissures. Cephalosporins, penicillinase-resistant penicillins,

or clindamycin are preferred for this purpose [70, 71]. In

most countries, methicillin-resistant strains of S. aureus

(MRSA) are an increasing problem, and it is very difficult

to clear the bacteria from patients with active skin disease.

In this context, bleach baths are a useful supplementary

treatment [72].

4.4 UV Phototherapy

Ultraviolet (UV) phototherapy for AD has been used for

many years. Treatments include broadband (BB)-UVB,

narrowband (NB)-UVB, UVA/B, UVA1 and psoralen (P)-

UVA. Most patients experience significant benefits. A

limitation is that the effectiveness of UV phototherapy is

short term, moderate and followed by recurrence of

symptoms within a few months, especially in patients with

severe AD.

A recent systematic review included 19 RCTs and 905

patients. The studies were clinically and qualitatively het-

erogeneous, but the authors concluded that UVA-1 and

NB-UVB appear to be the most effective treatment

modalities for the reduction of clinical signs and symptoms

of AD [73].

UV treatments should not be started when the skin is

acutely inflamed and oozing, as this will often result in a

poor outcome of the treatment. The patient should be

treated with emollients, topical corticosteroids and anti-

bacterials, if indicated, for some days before UV treatment

is started. This may not be necessary for high-dose UVA1.

The carcinogenic effects of UV phototherapy limit their

use for long-term maintenance treatment.

There are limited data on the use of phototherapy for

children with AD. Darne et al. [74] performed a prospec-

tive comparative cohort study including 55 children with

moderate to severe AD. Twenty-nine children were treated

with NB-UVB for 12 weeks and 26 children served as

controls. There was a 61 % reduction in Six Area Six Sign

Atopic Dermatitis (SASSAD) score in the treated group

compared with a 6 % increase in the unexposed cohort, and

the quality of life was improved in the treated group [74].

These data are supported by two other retrospective

studies on the use of NB-UVB in children with AD [75,

76]. Further studies are needed to evaluate the long-term

safety in treating children with phototherapy.

4.5 Systemic Immunosuppressive Therapy

4.5.1 Corticosteroids

There are very limited data documenting the effect of

systemic corticosteroids, but clinical experience shows that

Diagnosis and Management of Atopic Dermatitis 761

they are rapidly effective in controlling the symptoms of a

severe flare in AD.

If a patient has moderate to severe chronic disease, one

should consider starting another systemic immunosup-

pressive treatment in combination with topical treatment,

while tapering the systemic corticosteroid, due to the

severe long-term side effects of systemic corticosteroids,

and the risk of flare after stopping the treatment [77].

4.5.2 Cyclosporine A

Cyclosporine A is the best-documented systemic treatment

for severe AD, licensed in many countries for this indica-

tion in adults. A meta-analysis pooling data from eight

RCTs demonstrated its effectiveness compared with pla-

cebo [78], and this conclusion has been confirmed in a very

recent systematic review including 14 trials [79]. Both

short-term and long-term use may help control severe AD

[80, 81].

The advantage of cyclosporine A is its rapid onset of

action. While most patients experience very few subjective

side effects, they have to be carefully monitored due to the

risk of severe side effects, in particular nephrotoxicity,

liver impairment and hypertension. The recommended dose

is between 2.5 and 5 mg/kg/day. It is beneficial to the

patient to start with 4–5 mg/kg/day to gain good control of

the disease and then taper off to the lowest effective dose.

When the drug is discontinued, around half of the patients

experience a rapid relapse, half remain in remission for

about 3 months and a few patients experience a rebound

phenomenon [80].

4.5.3 Methotrexate

Many clinicians have used methotrexate for severe AD in

both adults and children, but until recently only a few open,

uncontrolled case series had documented its effectiveness in

adults [82–84]. In 2010, an RCT compared the use of meth-

otrexate versus azathioprine for severe AD in adults. A total

of 42 patients were included and the treatment period was

12 weeks [85]. The mean relative reduction in the eczema

severity score (SCORAD) was 42 and 39 %, respectively. No

serious adverse events were observed in the study.

A recent RCT including 40 children has also demon-

strated its effect in children between 7 and 14 years of age

[86]. Methotrexate 7.5 mg/week was compared with

cyclosporine A 2.5 mg/kg/day for 12 weeks. The mean

reduction in SCORAD was 49.3 and 44.7 %, respectively.

Both drugs were well tolerated.

The primary side effects for methotrexate are cytopenias,

liver function abnormalities and gastrointestinal side effects.

Methotrexate is teratogenic and women of childbearing age

must use a safe contraception method [87]. This also includes

men treated with methotrexate who have fertile female

partners. If well tolerated, the major advantage of metho-

trexate is that it can be used for many years in chronic cases,

with a weekly dose between 7.5 and 25 mg in adults. The

recommended therapeutic dose range for children with pso-

riasis is 0.2–0.7 mg/kg/week [88] and we use the same dose

interval in children with AD. Methotrexate treatment in

children should not exceed adult dosing.

The patients should be informed about the slow onset of

action, which is up to 12 weeks, before the maximal effect

can be expected. If gastrointestinal side effects occur, or

the patient does not respond to the oral treatment, subcu-

taneous administration may be considered.

4.5.4 Azathioprine

Azathioprine is a frequently used immunosuppressive drug

in the management of severe AD in many countries, but

hardly ever used in other countries. This difference is

primarily based on clinical tradition. Main adverse effects

are cytopenias, gastrointestinal side effects, viral infec-

tions, and a small increase in skin carcinomas, especially in

patients with marked UV exposure. A typical maintenance

dose is between 1.5 and 3.0 mg/kg/day. Like methotrexate,

it has a slow onset of action.

A few patients have a low level or lack of the enzyme

thiomethylpurine transferase (TPMT), with a resultant

increased risk of developing severe myelosuppression.

Measurement of TPMT level before prescribing the drug, or

starting with a small test dose of azathioprine, under close

weekly laboratory monitoring, is necessary. Patients with

normal levels of TPMT can also develop myelosuppression.

A few controlled studies have so far been published docu-

menting the effect in adult patients with severe AD. Berth-

Jones et al. [89] demonstrated a 26 % reduction in the SAS-

SAD score in the azathioprine-treated group compared with a

3 % reduction in the placebo group. The treatment period was

3 months. The dropout rate was quite high, but a significant

reduction in pruritus, sleep loss and fatigue in the active group

was observed. In 2006, an RCT including 63 patients was

performed. Forty-two patients were treated with azathioprine

for 12 weeks. Patients with normal levels of TPMT received

azathioprine 2.5 mg/kg/day and patients with reduced TPMT

activity received 1 mg/kg/day. A mean reduction of 37 % in

disease activity was observed in the azathioprine-treated

group compared with 20 % in the placebo group [90].

Azathioprine is also effective in controlling severe AD

in children [91].

4.5.5 Mycophenolate

Mycophenolate mofetil can be considered as a third-line

drug in the treatment of severe AD. Standard doses are

762 C. Vestergaard, M. Deleuran

between 0.5 and 1.5 g twice daily in adults. Clinical

experience, case reports and small open studies suggest

benefit in patients with severe AD [92, 93]. The most

prominent adverse events are cytopenias and gastrointes-

tinal problems, and it is very important to know that my-

cophenolate is a teratogen [94, 95]. If gastrointestinal side

effects are a problem, a small open study suggests the use

of mycophenolate sodium as an alternative [96]. The drug

is formulated in an enteric-coated form and none of the ten

patients studied discontinued the drug due to adverse

events. The effect of this drug has been confirmed in a

larger study comparing mycophenolate sodium with low-

dose cyclosporine A for adult patients with severe AD [97].

4.6 Biologics

During the last years, a limited number of studies have

been published indicating an effect of some biological

agents on AD. However, the future looks promising, as

different biological treatments for AD are in the pipeline,

but so far no specific biologic therapy for AD has been

approved by the drug agencies.

4.6.1 Interferon-c

The main adverse effects of IFNc are flu-like symptoms,

and this has limited the use of the drug. IFNc treatment for

severe, recalcitrant AD should be considered as a third-line

option for patients who do not respond to or do not tolerate

other systemic treatments.

A double-blind, placebo-controlled study including 83

patients demonstrated more than 50 % improvement in

physicians’ overall assessment in 45 % of patients treated

with daily subcutaneous injections of 50 lg/m2 IFNc for

12 weeks compared with 20 % in the placebo group [98].

Two smaller studies have shown that the treatment can be

continued for up to 2 years [99, 100].

4.6.2 Anti-CD20

Rituximab is an anti-CD20 monoclonal antibody directed

against B cells. Six patients with severe AD were treated

with rituximab 1,000 mg intravenously at day 0 and 14. All

patients experienced a significant improvement of their

eczema, and the treatment was well tolerated [101]. This

effect could not be confirmed in a case report of two

patients with severe AD [102].

4.6.3 Anti-IL-5

IL-5 is essential for eosinophil growth, differentiation and

migration, and these cells play a role in the pathogenesis of

AD. In an RCT, two single doses of 750 mg of

mepolizumab (an anti-IL-5 recombinant humanized

monoclonal antibody) were given 1 week apart, to patients

with moderate to severe AD. Forty patients participated in

the study and the effect was assessed on day 14. A decrease

in peripheral blood eosinophils and a moderate clinical

improvement (\50 %) was observed [103].

4.6.4 Anti-IgE

Omalizumab is a humanized monoclonal mouse antibody

directed against IgE. It is registered for the treatment of

severe asthma, and it has also been shown to be effective in

chronic urticaria.

Some reports describe a beneficial effect in patients with

moderate to severe AD [104–106]. However, a 2:1-ran-

domized, double-blind, placebo-controlled study including

20 patients with an investigator global assessment score C2

did not reveal a superior effect of omalizumab over placebo

in reversing pre-existing chronic atopic disease in adults.

The total treatment period was 16 weeks [107].

4.6.5 Anti-IL-4 Receptor

Dupilumab, a new human monoclonal antibody that targets

the IL-4Ra subunit and inhibits the effect of IL-14 and IL-

13, has shown very promising results. In an RCT with

dupilumab 300 mg (n = 55) or placebo (n = 54) weekly

for 12 weeks, the Eczema Area and Severity Index (EASI)

score improved significantly by 74 versus 23 % [108].

Another study showed that 100 % of patients (n = 21)

treated with dupilumab and topical corticosteroids

achieved EASI 50 compared with 50 % treated with pla-

cebo and corticosteroids (n = 10) [109].

4.6.6 Intravenous Immunoglobulins (IVIG)

IVIG treatment has been tried for both adults and children

with severe, treatment-refractory AD. Conflicting results

have been obtained, suggesting that the treatment may be

helpful in primarily paediatric cases of the disease.

A small open study including ten adult patients showed

a very limited effect of the treatment after 2 months [110].

In 2011, Jee et al. [111] published an RCT including 40

children, of whom 30 received IVIG with a 1-month

interval for 3 months compared with 10 children who

received placebo. A significant reduction in SCORAD was

observed in the IVIG-treated group compared with the

placebo group after 3 months. Five patients in the active

group did not complete the study due to adverse effects.

Another RCT compared a single dose of IVIG 2 g/kg in

six children with cyclosporine A 4 mg/kg/day in eight

children for 3 months. As expected, cyclosporine A was

significantly more effective than IVIG, due to the single

Diagnosis and Management of Atopic Dermatitis 763

dose of IVIG, but initially there was a good effect of both

treatments [112]. Concomitant treatment with topical cor-

ticosteroids, antihistamines and antibacterials were allowed

in this study, and this may have influenced the outcome.

In conclusion, IVIG may be considered as a last resort

treatment in severe treatment-refractory AD in children.

4.7 Other Immunotherapies

4.7.1 Allergen-Specific Immunotherapy (ASIT)

Patients with severe AD often suffer from complicating type I

allergies and ASIT may be a useful complementary treatment

for patients with moderate to severe chronic AD and com-

plicating allergic asthma and/or rhino-conjunctivitis.

An RCT by Werfel et al. [113] has shown that ASIT

with house dust mite (HDM) preparations significantly

improved the disease activity, measured by SCORAD, in

patients sensitized to HDM, in a dose-dependent manner

over an observation period of 1 year. These results have

been confirmed in another study over 12 weeks using birch

pollen extracts in patients sensitized to this allergen [114].

4.7.2 Immunoadsorption

A pilot study including 12 patients with severe, treatment-

refractory AD was performed. All patients had a high

SCORAD and very high IgE values, in spite of a combi-

nation of topical and systemic treatments [115]. Patients

were treated with a total of ten immunoadsorptions, in

order to reduce high titres of circulating antibodies. Serum

IgE was reduced more than 90 % after each treatment, but

returned to initial concentrations shortly after the last

treatment. In spite of this, all patients experienced a highly

significant reduction in SCORAD and pruritus. Larger

controlled trials with this promising but expensive and

time-consuming treatment are warranted.

4.8 Dietary Factors

4.8.1 Food Allergy

Food allergy is most prevalent in young children with

moderate to severe eczema. A Danish population-based

study found that 14.8 % of children suffered from food

allergy and of these, 90 % had AD [116]. An undetected

food allergy may result in severe allergic reactions in the

child, including respiratory symptoms and anaphylaxis, but

it may also worsen the AD.

It is quite rare that a teenager or adult develops food

allergy for the first time, and a substantial proportion of

children suffering from allergy against primarily milk and

egg will grow out of their allergy during the first years of

life. In the Western world the most prevalent food allergies

are against milk, egg, peanut, hazelnut and fish [117].

Children with AD should be considered for food allergy

evaluation if there is a suspicion found in the clinical his-

tory. Food provocation tests are conducted in order to

confirm or reject the diagnosis, to find the threshold of the

allergy, but also to investigate if a child has developed

tolerance to the food over time.

4.8.2 Probiotics

There have been a number of studies dealing with the effect

of probiotics on AD. Most studies have shown beneficial

effects of the use of supplementation with probiotics in

mothers and infants in preventing development and

reducing the severity of AD [118].

The results taken together are conflicting, however, due

to differences in study design, dosage and strain of the

probiotic [119].

4.8.3 c-Linolenic Acid

Studies on essential fatty acid supplementation in AD have

indicated beneficial effects in some patients with AD, but a

recent Cochrane Systematic Database review concluded

that oral borage oil and evening primrose oil, which both

contain c-linolenic acid, lack effect on eczema. Improve-

ment was similar to respective placebos used in the

included trials [120].

4.9 Psychosomatic Approaches

A recent study by Chrostowska-Plak and co-workers

evaluated the relationship between pruritus and stress,

health-related quality of life (HRQoL) and depression in

adult patients with AD, and it was shown that patients with

symptoms suggesting depression had more intense pruritus

compared with the rest of the patients [121].

Psychological stress can induce exacerbations in eczema

activity [122], and illness representations and coping are

highly associated with self-rated physical impairment in

AD patients [123].

Further, it has been shown that psychological interventions

have a positive effect on itch and scratching behaviour. Stress

management programmes may therefore be a useful addition

to standard treatment in patients with AD [122].

4.9.1 Therapeutic Patient Education and Eczema Schools

Poor adherence to the treatment is frequent in patients with

AD. This may be due to fear of unwanted side effects, but

also to the time-consuming procedures related to topical

treatment regimens.

764 C. Vestergaard, M. Deleuran

The German Atopic Dermatitis Intervention Study

(GADIS), which included 823 children and adolescents,

showed that age-related educational programmes are

effective in the long-term management of AD [124].

A position paper describing the objectives and recom-

mendations for patient education in AD has recently been

published [125], and in spite of cultural and financial dif-

ferences between countries, a recent survey has shown that

there is a consensus among experts to integrate education

into the treatment of AD [126].

5 Conclusion

AD is a multi-etiological disease. The skin is characterized

by an inflammatory reaction primarily made up of Th2/

Th22 lymphocytes as well as Tc2 lymphocytes, but other

cell types such as dendritic cells, eosinophils and mast cells

may also play an important role. It is therefore safe to

consider AD as an inflammatory skin disease. Furthermore,

the research over the past 10–15 years has shown that a

decreased barrier function in the skin also plays a signifi-

cant role in the pathogenesis of AD. Recent studies have

linked these etiologies together because the inflammatory

reaction can influence the barrier function and vice versa.

Thus, the treatment for AD should ideally inhibit the

inflammatory reaction and re-establish the skin barrier

function. From this review of the latest trends in the

treatment of AD it is clear that most, if not all, of the

treatments are aimed at the inflammatory reactions. As

recommended by the EADV/ETFAD/EFA/ESPD and

GA2LEN [52, 127], the basic therapy of AD should be

application of moisturisers and then, depending on the

severity, an anti-inflammatory treatment should be added.

In mild to moderate cases, topical treatment with either

corticosteroids or calcineurin inhibitors may be used,

whereas in moderate to severe cases systemic immuno-

suppressive drugs can be added. The evidence level for

most systemic drugs is low and the use of these varies

greatly between the countries of Europe [128]; however,

some patients do need more than topical treatment.

Several studies have demonstrated that patient education

can have a very deep impact on the quality-of-life score for

AD patients and their families, and thus psychosocial

interventions are of equal importance as the immunosup-

pressive drugs. Thus, this kind of treatment should be

instigated as soon as possible for patients with moderate to

severe AD.

No AD-specific biological treatments are registered at

the moment, but phase I and II trials are being carried out.

There is a dire need for effective biological treatments that

target this disease specifically, with fewer adverse events

than the traditional treatments we have described here,

especially for patients with severe, chronic disease.

Acknowledgments Mette Deleuran is an investigator, speaker and/

or an advisor for AbbVie A/S, MSD, Pierre Fabre Dermo-cosmetique,

Meda Pharma, Leo Pharma, and Regeneron. Christian Vestergaard is

an investigator and/or speaker for Abbvie A/S, Leo Pharma, Novartis

and Astellas. No sources of funding were used to prepare this review.

References

1. Bieber T. Atopic dermatitis. N Engl J Med.

2008;358(14):1483–94.

2. Leung DY, Bieber T. Atopic dermatitis. Lancet.

2003;361(9352):151–60.

3. Ong PY, Leung DY. The infectious aspects of atopic dermatitis.

Immunol Allergy Clin North Am. 2010;30(3):309–21.

4. Olesen AB, Ellingsen AR, Larsen FS, Larsen PO, Veien NK,

Thestrup-Pedersen K. Atopic dermatitis may be linked to whe-

ther a child is first- or second-born and/or the age of the mother.

Acta Dermato-Venereologica. 1996;76(6):457–60.

5. Lewis-Jones S. Quality of life and childhood atopic dermatitis:

the misery of living with childhood eczema. Int J Clin Pract.

2006;60(8):984–92.

6. Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis.

Acta Dermato-Venereologica. 1980;Suppl 92:44–7.

7. Flohr C, Johansson SG, Wahlgren CF, Williams H. How atopic

is atopic dermatitis? J Allergy Clin Immunol.

2004;114(1):150–8.

8. Williams H, Flohr C. How epidemiology has challenged 3

prevailing concepts about atopic dermatitis. J Allergy Clin

Immunol. 2006;118(1):209–13.

9. Deleuran MS, Vestergaard C. Therapy of severe atopic derma-

titis in adults. Journal der Deutschen Dermatologischen

Gesellschaft (J German Soc Dermatol JDDG).

2012;10(6):399–406.

10. Darsow U, Wollenberg A, Simon D, Taieb A, Werfel T, Oranje

A, et al. ETFAD/EADV eczema task force 2009 position paper

on diagnosis and treatment of atopic dermatitis. J Eur Acad

Dermatol Venereol JEADV. 2010;24(3):317–28.

11. Bohme M, Svensson A, Kull I, Wahlgren CF. Hanifin’s and

Rajka’s minor criteria for atopic dermatitis: which do 2-year-

olds exhibit? J Am Acad Dermatol. 2000;43(5 Pt 1):785–92.

12. Gaitanis G, Velegraki A, Mayser P, Bassukas ID. Skin diseases

associated with Malassezia yeasts: facts and controversies. Clin

Dermatol. 2013;31(4):455–63.

13. Sutton RL. Summertime pityriasis of the elbow and knee. In:

Sutton RLJ, editor. Disease of the skin. 2nd ed. St. Louis: CV

Mosby; 1956. p. 898.

14. Wutrich B. Minimal variants of atopic eczema. In: Ring J,

Przybilla B, Ruzicka T, editors. Handbook of atopic eczema.

2nd ed. Berlin: Springer; 2006. p. 74–83.

15. Parronchi P, Macchia D, Piccinni MP, Biswas P, Simonelli C,

Maggi E, et al. Allergen- and bacterial antigen-specific T-cell

clones established from atopic donors show a different profile of

cytokine production. Proc Natl Acad Sci USA.

1991;88(10):4538–42.

16. Wuthrich B, Schmid-Grendelmeier P. The atopic eczema/der-

matitis syndrome. Epidemiology, natural course, and immunol-

ogy of the IgE-associated (’’extrinsic’’) and the nonallergic

(’’intrinsic’’) AEDS. J Invest Allergol Clin Immunol.

2003;13(1):1–5.

Diagnosis and Management of Atopic Dermatitis 765

17. Hvid M, Johansen C, Deleuran B, Kemp K, Deleuran M,

Vestergaard C. Regulation of caspase 14 expression in kerati-

nocytes by inflammatory cytokines—a possible link between

reduced skin barrier function and inflammation? Exp Dermatol.

2011;20(8):633–6.

18. Yagi R, Nagai H, Iigo Y, Akimoto T, Arai T, Kubo M.

Development of atopic dermatitis-like skin lesions in STAT6-

deficient NC/Nga mice. J Immunol. 2002;168(4):2020–7.

19. Palmer CN, Irvine AD, Terron-Kwiatkowski A, Zhao Y, Liao H,

Lee SP, et al. Common loss-of-function variants of the epider-

mal barrier protein filaggrin are a major predisposing factor for

atopic dermatitis. Nat Genet. 2006;38(4):441–6.

20. Gittler JK, Shemer A, Suarez-Farinas M, Fuentes-Duculan J,

Gulewicz KJ, Wang CQ, 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(6):1344–54.

21. Gutowska-Owsiak D, Schaupp AL, Salimi M, Taylor S, Ogg

GS. Interleukin-22 downregulates filaggrin expression and

affects expression of profilaggrin processing enzymes. Br J

Dermatol. 2011;165(3):492–8.

22. Nograles KE, Zaba LC, Shemer A, Fuentes-Duculan J, Cardi-

nale I, Kikuchi T, et al. IL-22-producing ’’T22’’ T cells account

for upregulated IL-22 in atopic dermatitis despite reduced IL-

17-producing TH17 T cells. J Allergy Clin Immunol.

2009;123(6):1244–52, e2.

23. Grimstad O, Sawanobori Y, Vestergaard C, Bilsborough J, Ol-

sen UB, Gronhoj-Larsen C, et al. Anti-interleukin-31-antibodies

ameliorate scratching behaviour in NC/Nga mice: a model of

atopic dermatitis. Exp Dermatol. 2009;18(1):35–43.

24. Homey B, Alenius H, Muller A, Soto H, Bowman EP, Yuan W,

et al. CCL27-CCR10 interactions regulate T cell-mediated skin

inflammation. Nat Med. 2002;8(2):157–65.

25. Vestergaard C, Bang K, Gesser B, Yoneyama H, Matsushima K,

Larsen CG. A Th2 chemokine, TARC, produced by keratino-

cytes may recruit CLA? CCR4? lymphocytes into lesional

atopic dermatitis skin. J Investig Dermatol. 2000;115(4):640–6.

26. Hvid M, Vestergaard C, Kemp K, Christensen GB, Deleuran B,

Deleuran M. IL-25 in atopic dermatitis: a possible link between

inflammation and skin barrier dysfunction? J Investig Dermatol.

2011;131(1):150–7.

27. Ikeda K, Nakajima H, Suzuki K, Kagami S, Hirose K, Suto A,

et al. Mast cells produce interleukin-25 upon Fc epsilon RI-

mediated activation. Blood. 2003;101(9):3594–6.

28. Ohno T, Morita H, Arae K, Matsumoto K, Nakae S. Interleukin-

33 in allergy. Allergy. 2012;67(10):1203–14.

29. Savinko T, Karisola P, Lehtimaki S, Lappetelainen AM,

Haapakoski R, Wolff H, et al. ST2 regulates allergic airway

inflammation and T-cell polarization in epicutaneously sensi-

tized mice. J Investig Dermatol. 2013;133(11):2522–9.

30. Yang Q, Li G, Zhu Y, Liu L, Chen E, Turnquist H, et al. IL-33

synergizes with TCR and IL-12 signaling to promote the

effector function of CD8? T cells. Eur J Immunol.

2011;41(11):3351–60.

31. Grewe M, Walther S, Gyufko K, Czech W, Schopf E, Krutmann

J. Analysis of the cytokine pattern expressed in situ in inhalant

allergen patch test reactions of atopic dermatitis patients. J In-

vestig Dermatol. 1995;105(3):407–10.

32. Fischer-Stabauer M, Boehner A, Eyerich S, Carbone T, Traidl-

Hoffmann C, Schmidt-Weber CB, et al. Differential in situ

expression of IL-17 in skin diseases. Eur J Dermatol EJD.

2012;22(6):781–4.

33. Peric M, Koglin S, Dombrowski Y, Gross K, Bradac E, Buchau

A, et al. Vitamin D analogs differentially control antimicrobial

peptide/’’alarmin’’ expression in psoriasis. PloS one.

2009;4(7):e6340.

34. Irvine AD. Fleshing out filaggrin phenotypes. J Investig Der-

matol. 2007;127(3):504–7.

35. O’Regan GM, Sandilands A, McLean WH, Irvine AD. Filaggrin

in atopic dermatitis. J Allergy Clin Immunol.

2008;122(4):689–93.

36. Thyssen JP, Linneberg A, Ross-Hansen K, Carlsen BC,

Meldgaard M, Szecsi PB, et al. Filaggrin mutations are strongly

associated with contact sensitization in individuals with der-

matitis. Contact Dermat. 2013;68(5):273–6.

37. Candi E, Schmidt R, Melino G. The cornified envelope: a model

of cell death in the skin. Nat Rev Mol Cell Biol.

2005;6(4):328–40.

38. McGrath JA, Uitto J. The filaggrin story: novel insights into

skin-barrier function and disease. Trends Mol Med.

2008;14(1):20–7.

39. Resing KA, Johnson RS, Walsh KA. Characterization of pro-

tease processing sites during conversion of rat profilaggrin to

filaggrin. Biochemistry. 1993;32(38):10036–45.

40. Denecker G, Ovaere P, Vandenabeele P, Declercq W. Caspase-

14 reveals its secrets. J Cell Biol. 2008;180(3):451–8.

41. Leyvraz C, Charles RP, Rubera I, Guitard M, Rotman S, Breiden

B, et al. The epidermal barrier function is dependent on the

serine protease CAP1/Prss8. J Cell Biol. 2005;170(3):487–96.

42. List K, Szabo R, Wertz PW, Segre J, Haudenschild CC, Kim

SY, et al. Loss of proteolytically processed filaggrin caused by

epidermal deletion of Matriptase/MT-SP1. J Cell Biol.

2003;163(4):901–10.

43. Pellerin L, Henry J, Hsu CY, Balica S, Jean-Decoster C, Mechin

MC, et al. Defects of filaggrin-like proteins in both lesional and

nonlesional atopic skin. J Allergy Clin Immunol.

2013;131(4):1094–102.

44. 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(1):150–5.

45. Cornelissen C, Marquardt Y, Czaja K, Wenzel J, Frank J, Lu-

scher-Firzlaff J, et al. IL-31 regulates differentiation and filag-

grin expression in human organotypic skin models. J Allergy

Clin Immunol. 2012;129(2):426–33, 33, e1–8.

46. Kim BE, Howell MD, Guttman-Yassky E, Gilleaudeau PM,

Cardinale IR, Boguniewicz M, et al. TNF-alpha downregulates

filaggrin and loricrin through c-Jun N-terminal kinase: role for

TNF-alpha antagonists to improve skin barrier. J Investig Der-

matol. 2011;131(6):1272–9.

47. Gutowska-Owsiak D, Schaupp AL, Salimi M, Selvakumar TA,

McPherson T, Taylor S, et al. IL-17 downregulates filaggrin and

affects keratinocyte expression of genes associated with cellular

adhesion. Exp Dermatol. 2012;21(2):104–10.

48. Breternitz M, Kowatzki D, Langenauer M, Elsner P, Fluhr JW.

Placebo-controlled, double-blind, randomized, prospective study

of a glycerol-based emollient on eczematous skin in atopic

dermatitis: biophysical and clinical evaluation. Skin Pharmacol

Physiol. 2008;21(1):39–45.

49. Wollenberg A, Wetzel S, Burgdorf WH, Haas J. Viral infections

in atopic dermatitis: pathogenic aspects and clinical manage-

ment. J Allergy Clin Immunol. 2003;112(4):667–74.

50. Boussault P, Leaute-Labreze C, Saubusse E, Maurice-Tison S,

Perromat M, Roul S, et al. Oat sensitization in children with

atopic dermatitis: prevalence, risks and associated factors.

Allergy. 2007;62(11):1251–6.

51. Lack G, Fox D, Northstone K, Golding J. Avon Longitudinal

Study of P, Children Study T. Factors associated with the

development of peanut allergy in childhood. N Engl J Med.

2003;348(11):977–85.

52. Ring J, Alomar A, Bieber T, Deleuran M, Fink-Wagner A,

Gelmetti C, et al. Guidelines for treatment of atopic eczema

766 C. Vestergaard, M. Deleuran

(atopic dermatitis) part I. J Eur Acad Dermatol Venereol JE-

ADV. 2012;26(8):1045–60.

53. Barnes PJ. Corticosteroids: the drugs to beat. Eur J Pharmacol.

2006;533(1–3):2–14.

54. Charman C, Williams H. The use of corticosteroids and corti-

costeroid phobia in atopic dermatitis. Clin Dermatol.

2003;21(3):193–200.

55. Berth-Jones J, Damstra RJ, Golsch S, Livden JK, Van Hooteg-

hem O, Allegra F, et al. Twice weekly fluticasone propionate

added to emollient maintenance treatment to reduce risk of

relapse in atopic dermatitis: randomised, double blind, parallel

group study. BMJ. 2003;326(7403):1367.

56. Levin E, Gupta R, Butler D, Chiang C, Koo JY. Topical steroid

risk analysis: differentiating between physiologic and pathologic

adrenal suppression. J Dermatol Treat. 2014;25(6):501–6.

57. Walsh P, Aeling JL, Huff L, Weston WL. Hypothalamus–pitu-

itary–adrenal axis suppression by superpotent topical steroids.

J Am Acad Dermatol. 1993;29(3):501–3.

58. Jensen JM, Scherer A, Wanke C, Brautigam M, Bongiovanni S,

Letzkus M, et al. Gene expression is differently affected by

pimecrolimus and betamethasone in lesional skin of atopic

dermatitis. Allergy. 2012;67(3):413–23.

59. Liu J, Farmer JD Jr, Lane WS, Friedman J, Weissman I, Schreiber

SL. Calcineurin is a common target of cyclophilin–cyclosporin A

and FKBP-FK506 complexes. Cell. 1991;66(4):807–15.

60. Meurer M, Folster-Holst R, Wozel G, Weidinger G, Junger M,

Brautigam M, et al. Pimecrolimus cream in the long-term

management of atopic dermatitis in adults: a six-month study.

Dermatology. 2002;205(3):271–7.

61. Reitamo S, Wollenberg A, Schopf E, Perrot JL, Marks R,

Ruzicka T, et al. Safety and efficacy of 1 year of tacrolimus

ointment monotherapy in adults with atopic dermatitis. The

European Tacrolimus Ointment Study Group. Arch Dermatol.

2000;136(8):999–1006.

62. Ruzicka T, Bieber T, Schopf E, Rubins A, Dobozy A, Bos JD,

et al. A short-term trial of tacrolimus ointment for atopic der-

matitis. European Tacrolimus Multicenter Atopic Dermatitis

Study Group. N Engl J Med. 1997;337(12):816–21.

63. Van Leent EJ, Graber M, Thurston M, Wagenaar A, Spuls PI,

Bos JD. Effectiveness of the ascomycin macrolactam SDZ ASM

981 in the topical treatment of atopic dermatitis. Arch Dermatol.

1998;134(7):805–9.

64. Reitamo S, Rustin M, Ruzicka T, Cambazard F, Kalimo K,

Friedmann PS, et al. Efficacy and safety of tacrolimus ointment

compared with that of hydrocortisone butyrate ointment in adult

patients with atopic dermatitis. J Allergy Clin Immunol.

2002;109(3):547–55.

65. Chen SL, Yan J, Wang FS. Two topical calcineurin inhibitors

for the treatment of atopic dermatitis in pediatric patients: a

meta-analysis of randomized clinical trials. J Dermatol Treat.

2010;21(3):144–56.

66. Arellano FM, Wentworth CE, Arana A, Fernandez C, Paul CF.

Risk of lymphoma following exposure to calcineurin inhibitors

and topical steroids in patients with atopic dermatitis. J Investig

Dermatol. 2007;127(4):808–16.

67. Leyden JJ, Marples RR, Kligman AM. Staphylococcus aureus in

the lesions of atopic dermatitis. Brit J Dermatol.

1974;90(5):525–30.

68. Birnie AJ, Bath-Hextall FJ, Ravenscroft JC, Williams HC.

Interventions to reduce Staphylococcus aureus in the manage-

ment of atopic eczema. Cochrane Database Syst Rev.

2008(3):CD003871.

69. Huang JT, Abrams M, Tlougan B, Rademaker A, Paller AS.

Treatment of Staphylococcus aureus colonization in atopic

dermatitis decreases disease severity. Pediatrics.

2009;123(5):e808–14.

70. Abeck D, Mempel M. Staphylococcus aureus colonization in

atopic dermatitis and its therapeutic implications. Brit J Der-

matol. 1998;139(Suppl 53):13–6.

71. Niebuhr M, Mai U, Kapp A, Werfel T. Antibiotic treatment of

cutaneous infections with Staphylococcus aureus in patients

with atopic dermatitis: current antimicrobial resistances and

susceptibilities. Exp dermatol. 2008;17(11):953–7.

72. Huang JT, Rademaker A, Paller AS. Dilute bleach baths for

Staphylococcus aureus colonization in atopic dermatitis to

decrease disease severity. Arch Dermatol. 2011;147(2):246–7.

73. Garritsen FM, Brouwer MW, Limpens J, Spuls PI.

Photo(chemo)therapy in the management of atopic dermatitis:

an updated systematic review with the use of GRADE and

implications for practice and research. 2014;170(3):501–13.

74. Darne S, Leech SN, Taylor AE. Narrowband ultraviolet B

phototherapy in children with moderate-to-severe eczema: a

comparative cohort study. Brit J Dermatol. 2014;170(1):150–6.

75. Clayton TH, Clark SM, Turner D, Goulden V. The treatment of

severe atopic dermatitis in childhood with narrowband ultravi-

olet B phototherapy. Clin Exp Dermatol. 2007;32(1):28–33.

76. Pavlovsky M, Baum S, Shpiro D, Pavlovsky L, Pavlotsky F.

Narrow band UVB: is it effective and safe for paediatric pso-

riasis and atopic dermatitis? J Eur Acad Dermatol Venereol

JEADV. 2011;25(6):727–9.

77. Schmitt J, Schakel K, Folster-Holst R, Bauer A, Oertel R, Au-

gustin M, et al. Prednisolone vs. ciclosporin for severe adult

eczema. An investigator-initiated double-blind placebo-con-

trolled multicentre trial. Brit J Dermatol. 2010;162(3):661–8.

78. Hoare C, Li Wan Po A, Williams H. Systematic review of

treatments for atopic eczema. Health Technol Assess.

2000;4(37):1–191.

79. Roekevisch E, Spuls PI, Kuester D, Limpens J, Schmitt J.

Efficacy and safety of systemic treatments for moderate-to-

severe atopic dermatitis: a systematic review. J Allergy Clin

Immunol. 2014;133(2):429–38.

80. Hijnen DJ, ten Berge O, Timmer-de Mik L, Bruijnzeel-Koomen

CA, de Bruin-Weller MS. Efficacy and safety of long-term

treatment with cyclosporin A for atopic dermatitis. J Eur Acad

Dermatol Venereol JEADV. 2007;21(1):85–9.

81. Mrowietz U, Boehncke WH. Leukocyte adhesion: a suitable

target for anti-inflammatory drugs. Curr Pharm Des.

2006;12(22):2825–31.

82. Goujon C, Berard F, Dahel K, Guillot I, Hennino A, Nosbaum

A, et al. Methotrexate for the treatment of adult atopic derma-

titis. Eur J Dermatol EJD. 2006;16(2):155–8.

83. Lyakhovitsky A, Barzilai A, Heyman R, Baum S, Amichai B,

Solomon M, et al. Low-dose methotrexate treatment for mod-

erate-to-severe atopic dermatitis in adults. J Eur Acad Dermatol

Venereol JEADV. 2010;24(1):43–9.

84. Weatherhead SC, Wahie S, Reynolds NJ, Meggitt SJ. An open-

label, dose-ranging study of methotrexate for moderate-to-

severe adult atopic eczema. Brit J Dermatol.

2007;156(2):346–51.

85. Schram ME, Roekevisch E, Leeflang MM, Bos JD, Schmitt J,

Spuls PI. A randomized trial of methotrexate versus azathioprine

for severe atopic eczema. J Allergy Clin Immunol.

2011;128(2):353–9.

86. 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(3):351–6.

87. Gromnica-Ihle E, Kruger K. Use of methotrexate in young

patients with respect to the reproductive system. Clin Exp

Rheumatol. 2010;28(5 Suppl 61):S80–4.

88. Paller AS. Dermatologic uses of methotrexate in children:

indications and guidelines. Pediatr Dermatol. 1985;2(3):238–43.

Diagnosis and Management of Atopic Dermatitis 767

89. Berth-Jones J, Takwale A, Tan E, Barclay G, Agarwal S, Ahmed

I, et al. Azathioprine in severe adult atopic dermatitis: a double-

blind, placebo-controlled, crossover trial. Brit J Dermatol.

2002;147(2):324–30.

90. Meggitt SJ, Gray JC, Reynolds NJ. Azathioprine dosed by

thiopurine methyltransferase activity for moderate-to-severe

atopic eczema: a double-blind, randomised controlled trial.

Lancet. 2006;367(9513):839–46.

91. Murphy LA, Atherton D. A retrospective evaluation of azathi-

oprine in severe childhood atopic eczema, using thiopurine

methyltransferase levels to exclude patients at high risk of

myelosuppression. Brit J Dermatol. 2002;147(2):308–15.

92. Ballester I, Silvestre JF, Perez-Crespo M, Lucas A. Severe adult

atopic dermatitis: treatment with mycophenolate mofetil in 8

patients. Actas Dermosifiliogr. 2009;100(10):883–7.

93. Murray ML, Cohen JB. Mycophenolate mofetil therapy for

moderate to severe atopic dermatitis. Clin Exp Dermatol.

2007;32(1):23–7.

94. Anderka MT, Lin AE, Abuelo DN, Mitchell AA, Rasmussen

SA. Reviewing the evidence for mycophenolate mofetil as a new

teratogen: case report and review of the literature. Am J Med

Genet A. 2009;149A(6):1241–8.

95. Klieger-Grossmann C, Chitayat D, Lavign S, Kao K, Garcia-

Bournissen F, Quinn D, et al. Prenatal exposure to mycophen-

olate mofetil: an updated estimate. J Obstet Gynaecol Can.

2010;32(8):794–7.

96. van Velsen SG, Haeck IM, Bruijnzeel-Koomen CA, de Bruin-

Weller MS. First experience with enteric-coated mycophenolate

sodium (Myfortic) in severe recalcitrant adult atopic dermatitis:

an open label study. Brit J Dermatol. 2009;160(3):687–91.

97. Haeck IM, Knol MJ, Ten Berge O, van Velsen SG, de Bruin-

Weller MS, Bruijnzeel-Koomen CA. Enteric-coated myco-

phenolate 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(6):1074–84.

98. Hanifin JM, Schneider LC, Leung DY, Ellis CN, Jaffe HS, Izu

AE, et al. Recombinant interferon gamma therapy for atopic

dermatitis. J Am Acad Dermatol. 1993;28(2 Pt 1):189–97.

99. Schneider LC, Baz Z, Zarcone C, Zurakowski D. Long-term therapy

with recombinant interferon-gamma (rIFN-gamma) for atopic der-

matitis. Ann Allergy Asthma Immunol. 1998;80(3):263–8.

100. Stevens SR, Hanifin JM, Hamilton T, Tofte SJ, Cooper KD. Long-

term effectiveness and safety of recombinant human interferon

gamma therapy for atopic dermatitis despite unchanged serum IgE

levels. Arch Dermatol. 1998;134(7):799–804.

101. Simon D, Hosli S, Kostylina G, Yawalkar N, Simon HU. Anti-

CD20 (rituximab) treatment improves atopic eczema. J Allergy

Clin Immunol. 2008;121(1):122–8.

102. Sediva A, Kayserova J, Vernerova E, Polouckova A, Capkova S,

Spisek R, et al. Anti-CD20 (rituximab) treatment for atopic

eczema. J Allergy Clin Immunol. 2008;121(6):1515–6 (author

reply 6–7).

103. Oldhoff JM, Darsow U, Werfel T, Katzer K, Wulf A, Laifaoui J,

et al. Anti-IL-5 recombinant humanized monoclonal antibody

(mepolizumab) for the treatment of atopic dermatitis. Allergy.

2005;60(5):693–6.

104. Forman SB, Garrett AB. Success of omalizumab as mono-

therapy in adult atopic dermatitis: case report and discussion of

the high-affinity immunoglobulin E receptor, FcepsilonRI.

Cutis. 2007;80(1):38–40.

105. Sheinkopf LE, Rafi AW, Do LT, Katz RM, Klaustermeyer WB.

Efficacy of omalizumab in the treatment of atopic dermatitis: a

pilot study. Allergy Asthma Proc. 2008;29(5):530–7.

106. Vigo PG, Girgis KR, Pfuetze BL, Critchlow ME, Fisher J,

Hussain I. Efficacy of anti-IgE therapy in patients with atopic

dermatitis. J Am Acad Dermatol. 2006;55(1):168–70.

107. Heil PM, Maurer D, Klein B, Hultsch T, Stingl G. Omalizumab

therapy in atopic dermatitis: depletion of IgE does not improve

the clinical course - a randomized, placebo-controlled and

double blind pilot study. Journal der Deutschen Dermatologis-

chen Gesellschaft (J German Soc Dermatol JDDG).

2010;8(12):990–8.

108. Bieber TRM, Thaci D, Graham N, Pirozzi G, Teper A, Ren H,

et al. Dupilumab monotherapy in adults with moderate-to-severe

atopic dermatitis: a 12-week, Randomized, Double-Blind, Pla-

cebo-Controlled Study. J Allergy Clin Immunol. 2014;133(2

Supplement):AB404.

109. Thaci D, Worm M, Ren H, Weinstein S, Graham N, Pirozzi G,

et al. Safety and Efficacy Of Dupilumab Versus Placebo For

Moderate-To-Severe Atopic Dermatitis In Patients Using Top-

ical Corticosteroids (TCS): Greater Efficacy Observed With

Concomitant Therapy Compared To TCS Alone. Journal of

Allergy and Clinical Immunology. 2014;133(2,

Supplement):AB192.

110. Paul C, Lahfa M, Bachelez H, Chevret S, Dubertret L. A ran-

domized controlled evaluator-blinded trial of intravenous

immunoglobulin in adults with severe atopic dermatitis. Brit J

Dermatol. 2002;147(3):518–22.

111. Jee SJ, Kim JH, Baek HS, Lee HB, Oh JW. Long-term efficacy

of intravenous immunoglobulin therapy for moderate to severe

childhood atopic dermatitis. Allergy Asthma Immunol Res.

2011;3(2):89–95.

112. Bemanian MH, Movahedi M, Farhoudi A, Gharagozlou M,

Seraj MH, Pourpak Z, et al. High doses intravenous immuno-

globulin versus oral cyclosporine in the treatment of severe

atopic dermatitis. Iran J Allergy Asthma Immunol.

2005;4(3):139–43.

113. Werfel T, Breuer K, Rueff F, Przybilla B, Worm M, Grewe M,

et al. Usefulness of specific immunotherapy in patients with

atopic dermatitis and allergic sensitization to house dust mites: a

multi-centre, randomized, dose–response study. Allergy.

2006;61(2):202–5.

114. Novak N, Thaci D, Hoffmann M, Folster-Holst R, Biedermann

T, Homey B, et al. Subcutaneous immunotherapy with a de-

pigmented polymerized birch pollen extract—a new therapeutic

option for patients with atopic dermatitis. Int Arch Allergy

Immunol. 2011;155(3):252–6.

115. Kasperkiewicz M, Schmidt E, Frambach Y, Rose C, Meier M,

Nitschke M, et al. Improvement of treatment-refractory atopic

dermatitis by immunoadsorption: a pilot study. J Allergy Clin

Immunol. 2011;127(1):267–70, 70, e1–6.

116. Eller E, Kjaer HF, Host A, Andersen KE, Bindslev-Jensen C.

Food allergy and food sensitization in early childhood: results

from the DARC cohort. Allergy. 2009;64(7):1023–9.

117. Sampson HA. The evaluation and management of food allergy

in atopic dermatitis. Clin Dermatol. 2003;21(3):183–92.

118. Foolad N, Brezinski EA, Chase EP, Armstrong AW. Effect of

nutrient supplementation on atopic dermatitis in children: a

systematic review of probiotics, prebiotics, formula, and fatty

acids. JAMA Dermatol. 2013;149(3):350–5.

119. Folster-Holst R. Probiotics in the treatment and prevention of

atopic dermatitis. Ann Nutr Metab. 2010;57(Suppl):16–9.

120. Bamford JT, Ray S, Musekiwa A, van Gool C, Humphreys R,

Ernst E. Oral evening primrose oil and borage oil for eczema.

Cochrane Database Syst Rev. 2013;4:CD004416.

121. Chrostowska-Plak D, Reich A, Szepietowski JC. Relationship

between itch and psychological status of patients with atopic der-

matitis. J Eur Acad Dermatol Venereol JEADV. 2013;27(2):e239–42.

122. Schut C, Weik U, Tews N, Gieler U, Deinzer R, Kupfer J.

Psychophysiological effects of stress management in patients

with atopic dermatitis: a randomized controlled trial. Acta

Dermato-Venereol. 2013;93(1):57–61.

768 C. Vestergaard, M. Deleuran

123. Schut C, Felsch A, Zick C, Hinsch KD, Gieler U, Kupfer J. Role

of illness representations and coping in patients with atopic

dermatitis: a cross-sectional study. J Eur Acad Dermatol

Venereol JEADV. 2013 [epub ahead of print].

124. Staab D, Diepgen TL, Fartasch M, Kupfer J, Lob-Corzilius T,

Ring J, et al. Age related, structured educational programmes for

the management of atopic dermatitis in children and adoles-

cents: multicentre, randomised controlled trial. BMJ.

2006;332(7547):933–8.

125. Barbarot S, Bernier C, Deleuran M, De Raeve L, Eichenfield L,

El Hachem M, et al. Therapeutic patient education in children

with atopic dermatitis: position paper on objectives and rec-

ommendations. Pediatr Dermatol. 2013;30(2):199–206.

126. Stalder JF, Bernier C, Ball A, De Raeve L, Gieler U, Deleuran

M, et al. Therapeutic patient education in atopic dermatitis:

worldwide experiences. Pediatr Dermatol. 2013;30(3):329–34.

127. Ring J, Alomar A, Bieber T, Deleuran M, Fink-Wagner A,

Gelmetti C, et al. Guidelines for treatment of atopic eczema

(atopic dermatitis) Part II. J Eur Acad Dermatol Venereol JE-

ADV. 2012;26(9):1176–93.

128. 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. Brit J Dermatol.

2013;169(4):901–9.

Diagnosis and Management of Atopic Dermatitis 769