You are on page 1of 19

Allergy 2006: 61: 969–987  2006 American Academy of Allergy, Asthma and Immunology

and the European Academy of Allergology and Clinical Immunology


DOI: 10.1111/j.1398-9995.2006.01153.x

Review article

Diagnosis and treatment of atopic dermatitis in children and


adults: European Academy of Allergology and Clinical
Immunology/American Academy of Allergy, Asthma and
Immunology/PRACTALL Consensus Report

There are remarkable differences in the diagnostic and therapeutic management C. A. Akdis1, M. Akdis1, T. Bieber2,
of atopic dermatitis practiced by dermatologists and pediatricians in different C. Bindslev-Jensen3,
countries. Therefore, the European Academy of Allergy and Clinical Immuno- M. Boguniewicz4,5, P. Eigenmann6,
logy and the American Academy of Allergy, Asthma and Immunology nomin- Q. Hamid7, A. Kapp8, D. Y. M. Leung4,
ated expert teams who were given the task of finding a consensus to serve as a J. Lipozencic9, T. A. Luger10,
guideline for clinical practice in Europe as well as in North America. The con- A. Muraro11, N. Novak2, T. A. E.
sensus report is part of the PRACTALL initiative, which is endorsed by both Platts-Mills12, L. Rosenwasser4,
academies. A. Scheynius13, F. E. R. Simons14,
J. Spergel15, K. Turjanmaa16,
U. Wahn17, S. Weidinger18, T. Werfel19,
T. Zuberbier17 for the European
Academy of Allergology and Clinical
Immunology/American Academy of
Allergy, Asthma and Immunology/
PRACTALL Consensus Group*
1
The Swiss Institute of Allergy and Asthma
Research, Davos, Switzerland; 2The University of
Bonn, Bonn, Germany; 3The Odense University
Hospital, Odense, Denmark; 4The National Jewish
Medical and Research Center, Denver, CO, USA;
5
The University of Colorado School of Medicine,
Denver, CO, USA; 6The University Children's
Hospital Geneva, Geneva, Switzerland; 7The McGill
University, Montreal, QC, Canada; 8The Hannover
Medical University, Hannover, Germany; 9The
Zagreb Hospital Center and School of Medicine
University of Zagreb, Zagreb, Croatia; 10The
Munster University, Munster, Germany; 11The
University of Padua, Padua, Italy; 12The Asthma and
Allergic Diseases Center, Charlottesville, VA, USA;
13
The Karolinska University Hospital Solna,
Stockholm, Sweden; 14The University of Manitoba,
MB, Canada; 15The Children's Hospital of
Philadelphia and the University of Pennsylvania
School of Medicine, USA; 16The Tampere University
Hospital, Finland; 17The ChariteÕ University of Berlin,
Berlin, Germany; 18The Technical University Munich,
Munich, Italy; 19The Hannover Medical School,
Abbreviations: AD, atopic dermatitis; APT, atopy patch test; CyA, cyclosporine A; DC: Hannover, Germany
dendritic cell; IDEC, inflammatory dendritic epidermal cell; LC, langerhans cell; MnSOD,
manganese superoxide dismutase; pDC, plasmacytoid dendritic cell; SPT, skin prick test; TCI, Key words: atopic dermatitis; children; adults; risk
factors; immunopathology; diagnosis; systemic
topical calcineurin inhibitors; TReg, T regulatory.
treatment; topical treatment.
*The EAACI/AAAAI/PRACTALL Consensus Group was chaired by Professor Ulrich Wahn. Ulrich Wahn, MD
ChariteÕ–Universittsmedizin Berlin
The PRACTALL program is a common initiative of both academies, focusing on practical Augustenburger Platz 1
issues of allergology. It is supported by an unrestricted educational grant from Novartis. D-13353 Berlin
Germany
This article is being copublished by The Journal of Allergy and Clinical Immunology and
Allergy. Accepted for publication 31 March 2006.

969
Akdis et al.

Atopic dermatitis (AD) is a chronic inflammatory children. The circumstance by which the genetics of AD
pruritic skin disease that affects a large number of might play a role in the level of natural history and
children and adults in industrialized countries. The development has been reflected in 2 forms of genetic
12-month prevalence in 11-year-old children, as studied studies:
in the Global International Study of Asthma and
• genome-wide screens that identify broad regions of
Allergies in Childhood trial, ranged from 1% to 20%,
the genome linked with AD and
with the highest prevalence typically found in Northern
• candidate gene studies that examine a presumed
Europe (1).
contribution of genetic variants of disease-process
In 45% of children, the onset of AD occurs during the
genes in case-control association studies.
first 6 months of life, during the first year of life in 60%,
and before the age of 5 years in at least 85% of affected Both approaches highlight the search for disease specific
individuals (2). In those children with onset before the AD alleles, as well as identifying overlapping genes
age of 2 years, 20% will have persisting manifestations associated with other allergic characteristics and disorders.
of the disease, and an additional 17% will have This process has been recently reviewed (6). A European
intermittent symptoms by the age of 7 years (Fig. 1) study of about 200 families with affected sibs looked at
(3). In adults with AD, only 16.8% had onset after phenotypes for AD, as well as allergic sensitization, and
adolescence (4, 5). found a region of highest linkage at human chromosome
The clinical pattern of AD varies with age. Infants 3q21 (7). Another study of 148 nuclear families in which
typically present with erythematous papules and vesicles AD, as well as other intermediate phenotypes, including
on the cheeks, forehead, or scalp, which are intensely asthma phenotype and total serum IgE level, were exam-
pruritic. The childhood phase typically occurs from ined identified association with 5 regions, including 1q21,
2 years of age to puberty. Children are less likely to have 17q25, 20p, 16q, and 5q31 (8). A Swedish study of 197
the exudative lesions of infancy and instead exhibit more affected sib pairs identified 4 phenotypes, as well as 11
lichenified papules and plaques representing the more locations, associated with all of the different phenotypes
chronic disease and involving the hands, feet, wrists, that ranged from severity of AD, specific IgE, and direct
ankles, and antecubital and popliteal regions. The adult diagnosis of AD (9). Finally, a Danish study looked at a
phase of AD begins at puberty and frequently continues small number of affected sib pairs and found association
into adulthood. Predominant areas of involvement with 3 locations within the genome (10). Not all of these
include the flexural folds, the face and neck, the upper locations are highly robust, and most of these associations
arms and back, and the dorsa of the hands, feet, fingers, from genome-wide screens might not yield effective iden-
and toes. The eruption is characterized by dry, scaling tification of specific genes when examined in more detail;
erythematous papules and plaques and the formation of these are summarized in Table 1 (7–10). Table 2 (11–13)
large lichenified plaques from lesional chronicity. summarizes the findings of candidate gene studies and their
association with atopy and asthma.
Among nongenetic determinants for the development
of AD, the role of infantile feeding has been investigated
Genetic and other risk factors for AD
(14). Arecent meta-analysis suggests that the incidence of
Parental atopy, in particular AD, is significantly associ- infantile AD is reduced by breast-feeding for a least
ated with the manifestation and severity of early AD in 4 months (15), but this effect is most probably transient
and tends to disappear after 3 years of age.
Environmental factors also play a role in the develop-
Age with AD Course of AD
ment of AD. In contrast to asthma, the role of passive
1–2 years
tobacco smoke exposure in AD is inconclusive. However,
exposure to aeroallergens (pets, mites, and pollen) has
3 years been clearly shown to increase the risk factors for AD and
AD severity (16–18). In addition, aeroallergens are a
4 years
trigger for exacerbations in adult AD. Sensitization to
5 years food allergens (cow’s milk and hen’s eggs) is associated
with infantile AD and related to disease severity. Food
6 years allergen sensitization is also predictive for persistence of
symptoms throughout childhood (3). Only in a minority
7 years
of those with food sensitization (up to 33% of patients
Pattern of AD: Intermittent Complete remission
with moderate-to-severe disease of all age groups) are
Persistent
food allergens of clinical relevance, as demonstrated by
Figure 1. The natural history of AD from infancy to childhood food challenge studies (19). Another risk factor for
obtained from the prospective birth cohort study Multicenter persistent AD symptoms is the severity of disease in
Atopy Study (3). infancy.

970
PRACTALL Consensus Report

Table 1. Genome-wide screens AD

Reference Study population Sample Phenotypes Regions of highest linkage

Lee et al. (7) European 199 families with two affected siblings AD 3q21
(2000) Allergic sensitization 3q21
Cookson et al. (8) British 148 nuclear families recruited via children with active AD AD 1q21
(2001) 17q25
AD plus asthma 20p
Serum IgE 16q-tel
5q31
Bradley et al. (9) Swedish 109 families (197 affected full sib-pairs, nine affected half sib-pairs) AD 3p24-22
(2002) 5p13
6q16
10p13-12
AD plus specific IgE 18q21
4q24-26
6p
1p32
Extreme AD 18p
21q21
7p14
Severity score of AD 13q14
15q14-15
17q21
3q14
Haagerup et al. (10) Danish 23 affected sib-pair families AD plus specific IgE 3p26-24
(2004) 4p15-14
18q11-12

Children with AD are at high risk of allergic asthma


Histopathology
and allergic rhinitis. Of those with AD during the first
2 years of life, 50% will have asthma during subsequent Clinically unaffected skin in AD is not normal. It is
years (20). The severity of AD, including early sensi- frequently dry and has a greater irritant skin response
tization to food, increases the risk of asthma and than normal healthy skin. Microscopic studies reveal a
allergic rhinitis (3, 21). The exact mechanism for the sparse perivascular T-cell infiltrate in unaffected AD skin
progression of the disease in children withADis that is not seen in normal healthy skin (Fig. 2) (25).
unknown; however, it appears to be a complex inter- Acute AD skin lesions present to the physician as
action of genetics, environmental exposure, and sensi- intensely pruritic, erythematous papules associated with
tization. For children with a family history of atopy, excoriation and serous exudation. There is a marked
early AD, and sensitization, almost all are expected to infiltration of CD41 activated memory T cells in acute
have asthma. AD. Antigen-presenting cells (eg, Langerhans cells [LCs],
In murine models of AD, epicutaneous sensitization inflammatory dendritic epidermal cells [IDECs], and
leads to systemic allergic responses, increased IgE levels, macrophages) in lesional and, to a lesser extent, in
airway eosinophilia, airway sensitization, and airway nonlesional skin bear IgE molecules (26). Mast cell
hyperresponsiveness similar to that seen in human asthma degranulation can be observed.
and allergy (22). In human subjects a recent report Chronic AD skin lesions have undergone tissue
suggests that in selected individuals sensitization to remodeling caused by chronic inflammation. These skin
peanut allergen might occur through the skin (23). lesions are associated with thickened plaques with
increased skin markings (lichenification), increased colla-
gen deposition in the dermis, and dry fibrotic papules.
Macrophages dominate the dermal mononuclear cell
Immunopathology
infiltrate. Eosinophils also contribute to the inflammatory
The pathophysiology of AD is the product of a complex response, and T cells remain present, although in smaller
interaction between various susceptibility genes, host numbers than seen in acute AD.
environments, infectious agents, defects in skin barrier
function, and immunologic responses (24). Activation of
Cytokines and chemokines
T lymphocytes, dendritic cells (DCs), macrophages,
keratinocytes, mast cells, and eosinophils are character- AD skin lesions are orchestrated by the local tissue
istic of AD skin inflammatory responses. expression of proinflammatory cytokines and chemokines

971
Akdis et al.

Table 2. Candidate gene studies in AD

Gene Gene name Location Variant Phenotype Population Association

TLR2 Toll-like receptor 2 4q32 Arg753Gln Severe AD German Yes


AD German No
IRF2 Interferon regulatory factor 2 4q35 )467 G/A AD Japanese Yes
CSF2 Colony-stimulating factor 2 5q31 )677 A/C AD British Yes
3606 T/C, 3928 C/T AD at 12 and 24 months Canadian Yes
IL4 IL-4 5q31 )590 C/T AD Japanese Yes
Extrinsic AD German Yes
AD Australian No
Intrinsic AD Japanese No
IL13 IL-13 5q31 Arg130Gln AD Canadian Yes
AD Japanese Yes
AD German Yes
)111C/T AD Dutch Yes
AD Japanese No
IL5 IL-5 5q31 )703 C/T Blood eosinophilia in AD Japanese Yes
IL18 (11) IL-18 11q22 )137 G/C, )133 C/G AD German Yes
113 T/G, 127 G/T
CARD4 (12) Caspase recruitment 7p14-p15- rs2736726 AD German Yes
domain-containing protein 4 rs2075817
rs2975632
rs2075822
rs2907749
rs2907748
CD14 Monocyte differentiation antigen CD14 5q31 )159 C/T AD (interaction with dog American Yes
ownership)
AD German No
IL12B Interleukin-12B 5q31-33 1188 A/C AD, Psoriasis Japanese Yes
SPINK5 Serine protease inhibitor, Kazal-type 5 Glu420Lys AD British Yes
AD Japanese Yes
AD Japanese Yes
Asthma with AD German Yes
Disease severity and food Japanese Yes
allergy in AD
FCER1B High-affinity IgE receptor, b-chain 11q13 RsaIin2, RsaIex7 AD British Yes
GSTP1 Glutathione S-transferase, PI 11q13 Ile105Val AD Russian Yes
Haplotypes AD Russian Yes
PHF11 Plant homeodomain zink finger 13q14 T/C intron3, G/A 3 UTR Childhood AD Australian Yes
11 protein
CMA1 Mast cell chymase 14q11 BstXI AD Japanese Yes
AD Japanese Yes
AD Japanese No
AD Italian No
)1903 G/A IgE levels in AD British Yes
AD German (13) Yes
IL4RA Interleukin 4 Receptor chain 16p12 Gln551Arg Severe AD American Yes
Adult AD Japanese Yes
Ad (interaction with infection) British Yes
Intrinsic AD Japanese No
)3223 C/T Extrinsic AD German Yes
AD Japanese Yes
CARD15 Caspase recruitment domain- 16q12 2722 G/C AD German Yes
containing protein 15 AD German (11) Yes
RANTES Regulated on activation, normally 17q11-q12 )403 A/G AD German Yes
T cell expressed plus secreted AD Hungarian No
EOTAXIN Eotaxin 17q21 )426 C/T, )384 A/G IgE levels in AD Japanese Yes
TGFb1 TGF-b1 19q13.1 Arg25Pro AD British Yes
SCCE Stratum corneum chymotryptic enzyme 19q13 AACCins AD British Yes

(27). Cytokines, such as TNF-a and IL-1 from resident cells including the nuclear factor (NF) jB pathway, and
(keratinocytes, mast cells, and DCs), binds to receptors on inducing expression of vascular endothelial cell adhesion
the vascular endothelium, activating cellular signaling, molecules. These events initiate the process of tethering,

972
PRACTALL Consensus Report

Figure 2. Immunologic pathway involved in the progression of AD. Updated from Leung DY. Atopic dermatitis: new insights and
opportunities for therapeutic intervention. J Allergy Clin Immunol 2000;105:860–76. LC, Langerhans cell; MC, mast cell; TSLP,
human thymic stromal lymphopoietin; Ag, antigen; SAg, superantigen; AICD, activation-induced cell death; CLA, cutaneous lym-
phocyte antigen; MO, monocyte.

activation, and adhesion to the endothelium, followed by cells into acute and chronic AD skin lesions (31). Cutane-
extravasation of inflammatory cells. Once the inflamma- ous T cell–attracting chemokine (CCL27) is highly upreg-
tory cells have infiltrated into the tissue, they respond to ulated in AD and preferentially attracts cutaneous
chemotactic gradients established by chemoattractant lymphocyte antigen–positive T cells into the skin. Finally,
cytokines and chemokines, which emanate from sites of selective recruitment of CCR4-expressing TH2 cells is
injury or infection. These molecules play a central role in mediated by macrophage-derived chemokine and thymus
defining the nature of the inflammatory infiltrate in AD. and activation-regulated cytokine, levels of which are
The onset of acute AD is strongly associated with the increased in patients with AD (27). Severity of AD has been
production of TH2-produced cytokines, notably IL-4 and linked to magnitude of thymus and activation-regulated
IL-13, levels of which are significantly higher in AD cytokine levels (32). In addition, chemokines, such as
individuals compared with control subjects (28). Medi- fractalkine (33), IFN-c–inducible protein 10, monokine
ating isotype switching to IgE synthesis and upregulating induced by IFN-c, and IFN-c–inducible a chemoattract-
expression of adhesion molecules on endothelial cells, IL- ant, are strongly upregulated in keratinocytes (34) and
4 and IL-13 are implicated in the initial phase of tissue contribute to TH1 cell migration toward the epidermis.
inflammation, whereas the TH2 cytokine IL-5, which is
involved in eosinophil development and survival, pre-
IgE and IgE receptors
dominates in the chronic form of the disease (28).
Increased production of GM-CSF in patients with AD In about 80% of adult patients with AD, the disease is
is reported to inhibit apoptosis of monocytes, thereby associated with increased serum IgE levels (>150 kU/L),
contributing to the chronicity of this condition (29). The sensitization against aeroallergens and food allergens,
maintenance of chronic AD also involves production of and/or concomitant allergic rhinitis and asthma (35, 36).
the TH1-like cytokines IL-12 and IL-18, as well as several In contrast, 20% of adult patients with AD have normal
remodeling-associated cytokines, such as IL-11 and TGF- serum IgE levels. This subtype of AD often has a late
b1, which are expressed preferentially in chronic forms of onset (>20 years of life) and a lack of IgE sensitization
the disease (30). against inhalant or food allergens (35, 36). However,
Increased expression of C-C chemokines (monocyte some of these patients might have IgE sensitization
chemoattractant protein 4, eotaxin, and RANTES) con- against microbial antigens, such as Staphylococcus aureus
tributes to infiltration of macrophages, eosinophils, and T enterotoxins and Candida albicans or Malassezia sympo-

973
Akdis et al.

dialis (formally known as Pityrosporum ovale) (37, 38). In genetically engineered to overexpress IL-4 in their skin
addition, some of these patients have positive reactions have inflammatory pruritic skin lesions similar to those
on the atopy patch test (APT) (39). In children a transient seen in patients with AD, suggesting that local skin
form of AD with low IgE serum levels and without any expression of TH2 cytokines plays a critical role in AD
detectable sensitizations has been shown, which develops (49). Allergen-sensitized skin from IL-5–deficient mice
into the extrinsic variant of AD with increasing IgE has been found to have no detectable eosinophils and
serum levels and developing sensitizations against aero- exhibits decreased thickening, whereas skin from
allergens and food allergens later in life (40). IL-4–deficient mice displays normal thickening of the
Expression of IgE receptors (i.e., the high-affinity skin layers but has a reduction in eosinophil counts (47).
receptor for IgE [FceRI]) can be found in the epidermal In patients with AD, activated T cells with skin-homing
skin lesions of patients with AD. The reason for a higher properties, which express high levels of IFN-c, predom-
IgE-binding capacity of DCs in the skin and in the inantly undergo apoptosis in the circulation, skewing the
peripheral blood of patients with AD is that FceRI is immune response to surviving TH2 cells as a mechanism
regulated distinctly on DCs of atopic and nonatopic for TH2 predominance (50). In the affected skin these
subjects (41). T cells switch on effector cytokines and induce the
activation and apoptosis of keratinocytes (51).
Recently, T regulatory (TReg) cells have been described
Skin barrier dysfunction
as a further subtype of T cells, with immunosuppressive
AD is characterized by dry skin, even involving nonle- function and cytokine profiles distinct from those of
sional skin and increased transepidermal water loss. In either TH1 or TH2 cells (52–54). TReg cells are able to
particular, ceramides serve as the major water-retaining inhibit the development of both TH1 and TH2 responses.
molecules in the extracellular space of the cornified Mutations in a nuclear factor expressed in TReg cells,
envelope, and the barrier function of these complex Foxp3, result in immune dysregulation polyendocrinop-
structures is provided by a matrix of structural proteins, athy enteropathy X-linked syndrome characterized by
which are bound to ceramides (42, 43). A reduced content hyper-IgE, food allergy, and eczema (55). In addition,
of ceramides has been reported in the cornified envelope staphylococcal superantigens subvert TReg cell function
of both lesional and nonlesional skin in patients with AD. and might thereby augment skin inflammation (56).
Changes in stratum corneum pH levels have been found
in patients with AD and might impair lipid metabolism in DCs. Two types of FceRI-bearing myeloid DCs have
the skin (44). Overexpression of stratum corneum chym- been found in the lesional skin of patients with atopic
otryptic enzyme is also likely to contribute to the eczema, namely LCs and IDECs. Both display a different
breakdown of the AD epidermal barrier (45). This would function in the pathophysiologic network of AD. LCs
allow penetration of irritants and allergens, which trigger play a predominant role in the initiation of the allergic
an inflammatory response, thus contributing to the immune response and prime naive T cells into T cells of
cutaneous hyperreactivity characteristic of AD. The the TH2 type with high IL-4–producing capacity, which
increased susceptibility to irritants in patients with AD predominate in the initial phase of AD (57). Further on,
might therefore represent a primary defect of epidermal stimulation of FceRI on the surface of LCs by allergens
differentiation compounded by the presence of inflam- induces the release of chemotactic signals and recruitment
mation-induced skin damage. of precursor cells of IDECs and T cells in vitro.
Stimulation of FccRI on IDECs leads to the release of
high amounts of proinflammatory signals, which contrib-
Key cells in AD
ute to the amplification of the allergic immune response.
T cells. The key role of immune effector T cells in AD is In contrast to other inflammatory skin diseases, such as
supported by the observation that primary T-cell immu- allergic contact dermatitis or psoriasis vulgaris, only very
nodeficiency disorders frequently have increased serum low numbers of plasmacytoid DCs (pDCs), which play a
IgE levels and eczematous skin lesions, which clear after major role in the defense against viral infections, can be
successful bone marrow transplantation (24, 46). In detected within the epidermal skin lesions of AD (58).
animal models of AD, the eczematous rash does not pDCs in the peripheral blood of patients with AD have
occur in the absence of T cells (47). In addition, treatment been shown to bear the trimeric variant of FceRI on their
with topical calcineurin inhibitors (TCIs), which specif- cell surface, which is occupied with IgE molecules. The
ically target activated T cells, significantly reduces the modified immune function of pDCs in patients with AD
clinical skin rash present in AD (48). after FceRI-mediated allergen stimulation might contrib-
The important role that TH1 and TH2 cytokines play in ute to the deficiency of pDCs in patients with AD to
the skin’s inflammatory response has been demonstrated produce type I IFNs and thereby contribute to the high
in experimental models of allergen-induced allergic skin susceptibility of patients with AD toward viral skin
inflammation in mice with targeted deletions or overex- infections, such as herpes simplex–induced eczema her-
pression of these cytokines. In this regard transgenic mice peticum (59).

974
PRACTALL Consensus Report

Keratinocytes. Keratinocytes play a role in innate immu- after exposure to allergens, changes in humidity, excessive
nity by expressing Toll-like receptors and producing sweating, and low concentrations of irritants (73).
antimicrobial peptides in response to invading microbes Although pruritus can occur throughout the day, it is
(60). AD keratinocytes secrete a unique profile of usually worse at night, frequently disrupting the patient’s
hemokines and cytokines after exposure to proinflamma- sleep and overall quality of life (74). The mechanisms of
tory cytokines. This includes high levels of RANTES pruritus in AD are complex and poorly understood.
after stimulation with TNF-a and IFN-c (61). They are Allergen-induced release of histamine from skin mast cells
also an important source of thymic stromal lymphopoie- is not an exclusive cause of pruritus in AD because
tin, which activates DCs to prime naive T cells to produce antihistamines are not effective in controlling the itch of
IL-4 and IL-13 (62). AD unless there is associated urticaria (75). The observa-
There is growing evidence to incriminate the epidermis tion that TCIs and corticosteroids are effective therapeutic
as both target and enhancer of the inflammatory response agents at reducing pruritus suggests that the inflammatory
in AD (34, 63). Apoptosis of keratinocytes induced by T cells play an important role in driving pruritus (76, 77).
cells and mediated by Fas is a crucial event in the Other substances that have been implicated in pruritus
formation of eczema/spongiosis in AD (63). IFN-c include cytokines, neuropeptides, proteases, eicosanoids,
released from T cells upregulates Fas and several cytok- and eosinophil-derived proteins (78–81).
ines, such as IL-1a, IL-1 receptor agonist, TNF-a, and
GM-CSF, and chemokines on keratinocytes (34, 64).
Triggers of AD
There is evidence that cleavage of E-cadherin and
sustained desmosomal cadherin contacts between kera- Stress. Stress-induced immunomodulation is altered in
tinocytes that are undergoing apoptosis result in spong- patients with AD, but the exact mechanisms are not well
ioform morphology in the epidermis as a hallmark of understood (69). This phenomenon might be mediated by
eczematous lesions (65). Suppression of keratinocyte neuroimmunologic factors, such as neuropeptides, which
activation and apoptosis remains a potential target for can be found in the blood and within the epidermal nerve
the treatment of AD (34, 62, 66). fibers in close association with epidermal LCs. Increased
levels of nerve growth factor and substance P can be
Eosinophils. Studies over the past 2 decades have shown found in the plasma of patients with AD and correlate
that eosinophils play a major role in AD, characterized by positively with the disease activity (82). Enhanced levels
activated eosinophils in the peripheral blood and in the of brain-derived growth factor can be detected in the sera
lesional skin (67, 68). Interestingly, psychosocial stress and plasma of patients with AD. Brain-derived growth
represents an important trigger for the increase of eosino- factor has been shown to reduce eosinophil apoptosis
phil counts in the peripheral blood (69). Inhibition of while enhancing chemotaxis of eosinophils in vitro (72).
eosinophil apoptosis in AD, probably mediated by an
autocrine release of IL-5 and GM-CSF, appears to be a Allergens. Placebo-controlled food challenge studies have
relevant mechanism for the eosinophil accumulation demonstrated that food allergens can induce eczematoid
observed in AD (70). Several lines of investigation indicate skin rashes in a subset of infants and children with AD
that eosinophils are recruited to, and activated at, tissue (83, 84). In some patients urticarial reactions can trigger
sites by TH2 cytokines, such as IL-5 and IL-13 (68). In the itch-scratch cycle that flares this skin condition.
addition, chemokines (eotaxin and RANTES) also con- Children with food allergy have positive immediate skin
tribute to eosinophil chemotaxis and activation. Moreover, test responses or serum IgE directed to various foods,
interaction of eosinophil surface molecules and the endot- particularly egg, milk, wheat, soy, and peanut. Food
helial cells vascular cell adhesion molecule 1 and intercel- allergen– specific T cells have been cloned from the skin
lular adhesion molecule 1 are important for eosinophil lesions of patients with AD, providing direct evidence
extravasation and activation. Eosinophils might also play a that foods can contribute to skin immune responses. In
role in switching the TH1 response in AD through addition, it is well established that food can exacerbate
production of significant amounts of IL-12 on activation AD both through allergic and nonallergic hypersensitivity
(71). Once activated, the eosinophil is capable of releasing reactions. Furthermore, direct contact with the skin (e.g.,
an armory of potent cytotoxic granule proteins and in the preparation of meals or when feeding infants)
chemical mediators contributing to tissue inflammation, might be an important factor for the aggravation of
as shown by the deposition of eosinophil products in the eczema.
inflamed skin (67, 68). Moreover, eosinophils might have a Beyond the age of 3 years, food allergy is frequently
relevant role in neuroimmunologic interactions (72). outgrown, but sensitization to inhalant allergens is com-
mon. Pruritus and skin lesions can develop after intrana-
sal or bronchial inhalation challenge with aeroallergens in
Pathophysiology of pruritus in AD
patients with AD. Epicutaneous application of aero-
Patients with AD have a reduced threshold for pruritus allergens (e.g., house dust mites, weeds, animal danders,
manifested as cutaneous hyperreactivity and scratching and molds) by means of the APT on uninvolved skin of

975
Akdis et al.

patients withADelicits eczematoid reactions in a subset of induce the proliferation of autoreactive T cells (98).
patients with AD (85). A combination of effective house Recently, it has been shown that IgE against manganese
dust mite reduction measures has been reported to superoxide dismutase (MnSOD) from the skin-colonizing
improve AD. The isolation from AD skin lesions and yeast M sympodialis cross-reacts with human MnSOD
allergen patch test sites of T cells that selectively respond (99). Because patients reacting to human MnSOD were
to Dermatophagoides pteronyssinus (Der p 1) and other sensitized against the M sympodialis MnSOD, sensitiza-
aeroallergens supports the concept that immune responses tion most likely occurs primarily by exposure to the
in AD skin can be elicited by inhalant allergens. environmental fungal MnSOD.

Microorganisms. Most patients with AD are colonized Irritant factors. Frequently, rough or woolly clothing
with S. aureus and experience exacerbation of their skin leads to mechanical irritation and exacerbation of AD
disease after infection with this organism (86). In patients and eczema. Chemical irritants like skin-cleansing agents
with AD with bacterial infection, treatment with anti- should also be considered but can only be satisfactorily
staphylococcal antibiotics can result in reduction of skin identified by means of avoidance.
disease. An important strategy by which S. aureus
exacerbates AD is by secreting toxins called superanti-
gens, which stimulate activation of T cells and macroph- Diagnosis
ages. Most patients with AD make specific IgE antibodies
Symptoms and signs and diagnostic criteria
directed against staphylococcal superantigens, which
correlate with skin disease severity (87). Superantigens The use of well-defined diagnostic criteria is important in
also induce corticosteroid resistance, thereby complica- the diagnosis of AD, especially for those patients who
ting their response to therapy (88). lack the typical phenotype of the disease, and the
Binding of S. aureus to skin is enhanced by AD skin diagnostic criteria developed by Hanifin and Rajka are
inflammation. This is supported by clinical studies widely accepted (Fig. 3) (100). Other criteria have been
demonstrating that treatment with topical corticosteroids developed (101) that correlate well with those of Hanifin
or tacrolimus reduces S. aureus counts in AD. AD skin and Rajka, although use of only visible eczema as a
has also been found to be deficient in antimicrobial criterion might lead to overdiagnosis of the disease. Skin
peptides needed for host defense against bacteria, fungi, biopsies are not essential for the diagnosis but might be
and viruses (89, 90). This constellation of genes is required to exclude other diagnoses, particularly in
underexpressed because of the significant upregulation adults.
of TH2 cytokines in AD. Along with lower levels of
proinflammatory cytokines, such as TNF-a and IFN-c,
Differential diagnosis
the decrease in antimicrobial defenses within patients
with AD might explain their increased susceptibility to The most important differential diagnoses are other forms
skin infections compared with that seen in patients with of eczema. Especially in adulthood, combination forms
psoriasis (90). are prevalent with components of atopic, contact, and
Patients with AD have an increased propensity toward irritative eczema. Atopic eczema of the hands and feet
disseminated infections with herpes simplex or vaccinia must be differentiated from psoriasis in the palms and
virus. Susceptibility to severe viral infections, such as
eczema herpeticum or eczema vaccinatum, might be
linked to the severity of atopy (91). As such, smallpox Hanifin & Rajka
UK Working Party Symptoms and Differential diagnoses
vaccination is contraindicated in patients with AD unless Concomitant diagnoses
there is imminent danger of exposure to smallpox (92).
Schultz Larsen signs
DARC
There is increasing evidence that the opportunistic Visible eczema
yeast Malassezia species represents a contributing factor
in AD (37, 93). Several studies have demonstrated the
Scorad Assessment of Fluctuations
Easi Exacerbating factors
presence of specific serum IgE, a positive skin prick test severity
(SPT) response, and a positive APT response against
Malassezia species in adults with AD (37). IgE sensitiza- Food allergy
Inhalant allergy
Complicating or Relevance to eczema
tion to Malassezia species is specific for patients with AD Infections exacerbating Diagnostic work up
but is not seen in patients with asthma or allergic rhinitis Milieu factors
(94, 95). Other

Autoantigens. Autoreactivity of patients with AD to Diagnosis


human proteins might contribute to the pathophysiology
of this condition (96, 97). IgE against autoantigens could Figure 3. Diagnostic approach to patients presenting with
stimulate type 1 hypersensitivity reactions and DCs and symptoms and signs of atopic dermatitis.

976
PRACTALL Consensus Report

soles and from tinea. Scabies infection must always be Decision points can be helpful in making the decision to
considered. The differential diagnosis of acute AD with perform oral challenges. However, the need for challenges
intense erythema of the skin, together with exudation or has to be decided on an individual basis.
blistering, for example, differs from differential diagnoses Other invalidated tests, such as lymphocyte cytotoxic-
of the chronic lichenified forms. Other, more rare diseases ity tests, the basophil degranulation test, or measurement
should be suspected, especially in recalcitrant cases: in of serum IgG (or subclasses), should not be used.
children genodermatoses, such as Netherton syndrome, APT is primarily a tool to investigate the mechanisms
including the recently described immune dysregulation of eczema in the skin. However, APT can also reveal
polyendocrinopathy enteropathy X-linked syndrome sensitization in patients with AD and might identify a
(102), should be considered, and in both children and subgroup of such patients. An elimination diet should not
adults vitamin deficiencies and malignancies, especially be recommended for a patient solely on the basis of a
cutaneous T cell lymphoma/mycosis fungoides, should be positive APT response to a food (108).
considered. All the abovementioned tests require specialist know-
ledge in their performance and interpretation.
Standardized, physician-supervised food challenges
Diagnostic work-up
provide the most accurate diagnostic tool (103). How-
The investigation of exacerbating factors in AD involves ever, it should be noted that patients can present with
a patient history, specific skin and blood tests, and reactions at least 24 hours after a food challenge, and the
challenge tests, depending on the degree of the disease challenge settings and protocol should be appropriately
severity and on the suspected factors involved (Fig. 4). designed; for example, in case of a negative challenge
response, the skin of the patient should be examined the
following day (104). After a diagnosis has been estab-
Food
lished, a tailor-made education program for the patient
There is no universally recommended diet for patients should be initiated.
with AD. Dietary restrictions should only be recommen-
ded in cases of an established diagnosis of food hyper-
Inhalant allergy
sensitivity. International guidelines for the diagnosis of
food hypersensitivity have been published (83, 103). As Sensitization to inhalant allergens is often seen in patients
regards food-induced eczema, it is important to note that with AD. Allergens can exacerbate AD either by means of
the predictive value of a positive case history is lower than inhalation, direct contact with the skin, or ingestion.
that for food-induced immediate reactions (104). Sensitization can be detected by means of SPTs (if the
Both SPTs and measurement of specific IgE can be skin is free from eczema) or by measurement of specific
used to assess for sensitization to a food at any age. IgE antibodies. In addition, APTs can be used to assess
Diagnostic sensitivity and specificity varies considerably the response in the skin. Most important allergens include
among different foods, reading systems, and age groups. dust mite, animal dander, and pollen confirmed by
A decision point discriminating between clinical relevance clinical trials and avoidance measures. The role of dust
of sensitization (with challenge as the gold standard) has mite allergen exposure is supported by patch tests,
been developed with regard to specific IgE and SPTs to avoidance studies, and the very high titers of IgE
egg, milk, peanut, and others foods in children (105–107). antibodies to mite proteins in a large proportion of
adults, as well as children older than 7 years with AD (94,
109–111). The positive effect of house dust mite avoid-
ance with special encasings has been shown in various
studies (110).

Contact allergy
In patients with AD, contact sensitization to topical
medications frequently occurs, especially in adults. In
cases of worsening eczema despite treatment, the possi-
bility of contact dermatitis needs to be ruled out by means
of patch testing. Patients with AD are no more likely to
be sensitized than normal.

Systemic and topical treatment


Figure 4. Relative significance of complicating or exacerbating
factors in patients with AD from infancy to adulthood. The management of AD presents a clinical challenge.

977
Akdis et al.

Because different emollients are available, selection cri-


Basic treatment
teria, such as the individual skin status, seasonal and
Basic therapy of AD should comprise optimal skin care, climatic conditions, and the time of day, should be
addressing the skin barrier defect with regular use of considered for optimizing the patientsÕ basic treatment.
emollients and skin hydration, along with identification ÔÔWater-in-oilÕÕ or ÔÔoil-in-waterÕÕ emulsions might be sub-
and avoidance of specific and nonspecific trigger factors. stituted to support the skin barrier function. Emollients
Nonspecific irritants include contactants, such as clothing containing polidocanol are effective in reducing pruritic
made from occluding or irritating synthetic or wool symptoms. Adjuvant application of topical preparations
material (112). Further irritating factors are soaps and hot with urea allows for intensive hydration of the skin,
water temperature during showering or bathing. Contacts whereas salicyl acid can be added to an emollient for the
with water should be minimized, moderately heated water treatment of chronic hyperkeratotic lesions (119).
should be used, and mild syndets with an adjusted pH
value (acidified to pH 5.5–6.0 in order to protect the acid Topical glucocorticosteroids. Topical glucocorticosteroids
mantle of the skin) should be used for cleansing (113). are still an important tool for the treatment of acute flare-
Other specific provocation factors and airborne and food ups (120, 121). Over recent years, the risk of adverse
allergens have to be considered (see the section on effects induced by topical steroids could effectively be
diagnosis for how to identify allergens) (108, 114). reduced by optimizing application protocols and using
Further treatment, on the basis of disease severity, new steroid preparations with improved risk/benefit
includes the addition of multiple therapeutic agents in a ratios and lower atrophogenic potential, such as predni-
step-wise fashion (Fig. 5). A combination of different carbate, mometasone furoate, fluticasone, and methyl-
topical agents might be indicated. In cases of severe AD prednisolone aceponate (77, 122, 123). For the topical use
that cannot be controlled with topical treatment, systemic of glucocorticosteroids, different therapeutic schemes
treatment options might need to be considered. For have been established: intermittent use might be as
optimal disease management, regular medical supervision, effective as an initial therapy with a high potent steroid
together with education of the patient or care providers and followed by a time-dependent dose reduction or change
appropriate psychosocial support, is needed. In selected over to a lower potent preparation (124). The choice of an
patients hospitalization might be of great benefit, especially adequate vehicle is important to achieve the optimal
in centers with a multidisciplinary team approach. therapeutic effect. Recent data indicate that in children
and adults an application of corticosteroids (fluticasone)
on unaffected skin twice weekly prevents further flare-ups
Topical treatment
of AD (125). Aside from an anti-inflammatory effect,
Emollients. A key feature of AD is severe dryness of the treatment with topical steroids contributes to a reduction
skin caused by a dysfunction of the skin barrier with of skin colonization with S. aureus and therefore might
increased transepidermal water loss (115). This is typically affect a further trigger factor of AD (126, 127).
accompanied by intense pruritus and inflammation. The The side effects of uncontrolled topical steroid use,
regular use of emollients is important for addressing this particularly on delicate skin areas, are well documented,
problem, and together with skin hydration, it represents and therefore topical steroid preparations should be
the mainstay of the general management of AD (116, 117). applied no more than twice daily as short-term therapy
Emollients should be applied continuously, even if no for acute eczematous lesions. Only mild to moderately
actual inflammatory skin lesions are obvious (118). potent preparations should be used on genital, facial, or
intertriginous skin areas. In children only mild to
moderately potent steroid preparations should be used
(128). In general, during acute flares, steroids should be
used in combination with baseline emollient skin care to
Systemic therapy
Recalcitrant, severe AD Step 4 (e.g. CyA) or UV therapy avoid steroid overuse and steroid-related side effects.

TCIs. The TCIs pimecrolimus and tacrolimus allow a


ase
se

Moderate to severe AD Step 3 Mid-high potency TCS and/or TCI* steroid-free, anti-inflammatory topical therapy of AD. In
di
of

both animal and human studies, both molecules demon-


y
sit

strated an immunomodulatory activity (129). In the


en

Mild to moderate AD Step 2 Low-mid potency TCS and/or TCI*


t
In

United States and Europe pimecrolimus cream (1%)


Basic treatment: and tacrolimus ointment (0.03%) are approved for the
Dry skin only Step 1 Skin hydration, emollients, avoidance of irritants,
identification and addressing of specific trigger factors treatment of AD in children aged 2 years and older (130)
and in adults (131). Tacrolimus ointment (0.1%) is only
Figure 5. Stepwise management of patients with atopic derma- approved for use in adults.
titis (AD). TCS, topical corticosteroids; TCI, topical calcineurin The anti-inflammatory potency of 0.1% tacrolimus
inhibitors; CyA, cyclosporine A. *Over the age of 2 years. ointment is similar to a corticosteroid with moderate

978
PRACTALL Consensus Report

potency (132), whereas 1% pimecrolimus cream is less observed (148). The use of silvercoated textiles and silk
active (133). Thus far, no trials have been published fabric with a durable antimicrobial finish can reduce
comparing pimecrolimus 1% with a mild corticosteroid. S. aureus colonization and eczema severity (149, 150).
Both agents proved to be effective, with a good safety pro- These new options are still under investigation.
file for a treatment period of up to 2 years with pime- The addition of a topical antimicrobial agent to a topical
crolimus (134) and up to 4 years with tacrolimus (135). steroid preparation has been shown to result in greater
A frequently observed side effect with TCIs is a clinical improvement than a topical steroid alone (151).
transient burning sensation of the skin. In a comparative Interestingly, AD seems not to be associated with a higher
study of the local side effects of 0.03% tacrolimus risk of sensitization against topical antimicrobials (152).
ointment versus 1% pimecrolimus cream in children, Because of deficient skin barrier function, patients with
pimecrolimus achieved better local tolerability than AD are exposed to a higher risk of recurrent bacterial
tacrolimus (136). superinfections of the skin. For the treatment of mild and
Preliminary studies indicate that treatment with TCIs is localized forms of this secondary infection, a topical
not associated with a risk of skin atrophy (137). Therefore antibiotic treatment might be beneficial.
they are a useful alternative for the treatment of sensitive Although erythromycin and fusidic acid have been
skin areas, such as the face and intertriginous regions. widely used in Europe, high resistance rates of S. aureus
Generalized viral infections, such as eczema herpeticum to erythromycin have resulted in a preferential use of
or eczema molluscatum, have been observed during TCI fusidic acid (153). Topical fusidic acid has proved to be
treatment (138). It is unclear whether a trend for very effective against S. aureus because of its low minimal
increased frequency of viral superinfections with use of inhibitory concentration and good tissue penetration
TCIs really exists (134). (154). However, long-term therapy with fusidic acid is
Although there is no evidence of a causal link of cancer suspected to be responsible for increasing resistance (155).
and the use of TCIs, the United States Food and Drug Therefore a restricted topical application for only short
Administration has issued a black-box warning for periods of about 2 weeks is advisable (156). For intrana-
pimecrolimus (Elidel; Novartis, Basel, Switzerland) and sal eradication of methicillin-resistant S. aureus, topical
tacrolimus (Protopic; Astellas, Deerfield, Ill) because of a mupirocin has been shown to be effective (157).
lack of long-term safety data (139, 140). Furthermore, the Other secondary infections caused by yeasts, dermat-
new labeling states that these drugs are recommended as ophytes, or streptococci have also been implicated as
second-line treatments and that their use in children trigger factors in AD (158). In general, signs of secondary
younger than 2 years of age is currently not recommen- infections should only be treated if present.
ded. Longterm safety studies with TCIs in patients with
AD, including infants and children, are ongoing (134).
Systemic treatment
Wet-wrap therapy. A wet layer of cotton dressing, which
Antimicrobial treatment
is then covered with tubular bandages applied over
emollients (141) in combination with antiseptics or Systemic antibiotic treatment is indicated for widespread
topical steroids (142), has been shown to be beneficial bacterial secondary infection, (primarily S. aureus). First-
in cases of exacerbated AD skin lesions (143). A more or second-generation cephalosporins or semisynthetic
practical alternative approach using clothing rather than penicillins for 7 to 10 days are usually effective. Eryth-
bandages has also been described in detail (144). romycin-resistant organisms are fairly common, making
macrolides less useful alternatives (159). In cases of
Topical antimicrobial therapy. The skin of patients with penicillin or cephalosporin allergy, clindamycin or oral
AD is heavily colonized with S. aureus, even at unin- fusidic acid are possible alternatives. Unfortunately,
volved sites. Toxins secreted by the majority of S. aureus recolonization after a course of antistaphylococcal ther-
on the skin behave as superantigens and, as discussed in apy occurs rapidly (160). Maintenance antibiotic therapy,
the pathophysiology section, can directly influence the however, should be avoided because it might result in
disease activity, although clinical signs of bacterial colonization by methicillin-resistant organisms.
superinfection might be absent (145, 146). Topical Infection of the skin with the herpes simplex virus in
antiseptics, such as triclosan (2,4,4Õ-trichloro-2Õ-hydrox- the form of an eczema herpeticum (Kaposi’s varicelliform
ydiphenyl ether) or chlorhexidine, offer the advantage of eruption) represents a severe and possibly life-threatening
a low sensitizing potential and low resistance rate. They complication of AD, requiring a systemic antiviral
can be used in emollients or as part of an additional wet- treatment with acyclovir or other antiviral agents (e.g.,
wrap dressing therapy (146). The topical use of triclosan valacyclovir) (138).
has been shown to be effective in significantly reducing Recent findings underline the pathogenetic importance
skin colonization with S. aureus and skin symptoms of a fungal colonization as a trigger factor (37, 96, 99).
(147). An irritative, photoallergenic, phototoxic, muta- Contradictory data have been published about the
genic, or carcinogenic potential of triclosan has not been efficacy of a systemic treatment of AD with ketoconazole

979
Akdis et al.

(161, 162), and although it is assumed that selected pression, hepatotoxicity, gastrointestinal disturbances,
patients with AD might benefit from a topical or systemic increased susceptibility for infections, and possible devel-
antimycotic therapy (163), the effect of a therapeutic opment of skin cancer. Because azathioprine is metabo-
intervention needs to be better defined by further studies. lized by the thiopurine methyltransferase, a deficiency of
this enzyme should be excluded before starting oral
immunosuppression with azathioprine (174). The recom-
Systemic corticosteroids
mended dosage of azathioprine for dermatologic indica-
Although oral corticosteroids are commonly used for tions is 1 to 3 mg/kg daily but should be determined
many different skin diseases, few randomized clinical based on thiopurine methyltransferase levels (175). Regu-
trials have been performed in patients with AD thus far lar blood tests must be performed throughout treatment
(164). It is well known that relapse after the discontinu- with azathioprine (173). The onset of action is usually
ation of oral steroids is often observed. Corticosteroids in slow, and benefit might not be apparent until 2 to
the form of a long-term oral therapy are associated with a 3 months after starting treatment (176).
series of well-documented side effects (e.g., disturbance of
growth, osteoporosis, cataracts, and development of
Antihistamines
lymphopenia). In cases of acute flare-up, patients might
benefit from a short course of systemic therapy with The therapeutic value of antihistamines seems to reside
corticosteroids, but long-term use and use in children principally in their sedative properties, and they are useful
should be avoided. as a short-term adjuvant to topical treatment during
relapses associated with severe pruritus. Nonsedating
antihistamines seem to have only very modest value in
Cyclosporin A
atopic eczema (177, 178). Although there are no large
As with TCIs, cyclosporin A (CyA) inhibits calcineurin- controlled studies to date, newer nonsedating antihista-
dependent pathways, resulting in reduced levels of pro- mines seem to have little or no value in atopic eczema
inflammatory cytokines, such as IL-2 and IFN-c. (177).
Multiple clinical trials have shown CyA to be an effective
treatment for adult and childhood AD, and although
Phototherapy
relapse after discontinuation of therapy is often observed,
posttreatment disease severity often does not return to The treatment of AD with phototherapy is well estab-
baseline levels (165–167). lished and represents a standard second-line treatment for
Despite the effectiveness of oral CyA in the treatment of adults (179). In phases of acute flares, a combination with
AD, because of the possible side effects, particularly renal corticosteroids is often practiced.
toxicity, the use of CyA should be limited to patients with The following therapy options can be used for AD:
severe refractory disease, contraindications must be exclu- broad-band UVB (280–320 nm), narrow-band UVB
ded, and blood pressure and laboratory parameters must (311–313 nm), UVA (320–400 nm), UVA1 (340–
be monitored closely. The treatment can be performed in 400 nm), PUVA, and Balneo-PUVA. In addition, com-
the form of a short- or long-term therapy with high-dose binations of UVB and topical glucocorticosteroids and
(3–5 mg/kg/d) or low-dose (2.5 mg/kg/d) administration, UVB with UVA, as well as UVA1 medium- and high-
depending on the patientsÕ individual medical conditions dose therapy, showed favorable results (180–182).
(168, 169). The principle of treatment should be to aim for In children UV therapy should be restricted to
the lowest effective dose and the shortest treatment period adolescents older than 12 years, except in exceptional
because toxicity is related to both of these factors. In cases. It must be emphasized that to date, information
children it should be considered that vaccinations might about the longterm side effects of UV therapy is still not
not be effective during immunosuppression. available.

Azathioprine Immunotherapy
Azathioprine is a long-known systemic immunosuppres- To date, hyposensitization is not an established instru-
sive agent affecting purine nucleotide synthesis and ment for the treatment of AD. Although a number of case
metabolism, which has been shown to be efficient for reports suggest clinical benefit from allergen-specific
many dermatologic conditions (170). It is also used quite desensitization in AD, double-blinded controlled trials
frequently as monotherapy in nonlicensed indications, have failed to show consistent efficacy of immunotherapy
including AD (171). Although most reports on the use of in the treatment of AD (183). A recent randomized
azathioprine in AD have been uncontrolled, open, and multicenter trial investigated the efficacy of an allergen-
retrospective studies, there is accumulating evidence for specific immunotherapy of house dust mite preparations
its efficacy in severe recalcitrant AD (172–174). Azathiop- in patients with AD sensitized to house dustmite allergens
rine has a number of side effects, including myelosup- for 1 year and revealed a dose-dependent effect on disease

980
PRACTALL Consensus Report

symptoms (184). However, further well-controlled studies patients with moderate and severe chronic AD and their
are needed to determine the future role of immunothera- parents. Structured patient education should enable both
py for AD. the patient and the parent to have realistic short-term
goals, enter a process of problem solving, accept living
with their disease, appropriately use available social
Additional treatment options and future perspectives
support, and enhance their own motivation for therapy.
The development of new, targeted therapeutic approaches
is based on an increasing knowledge of the cellular and
molecular aspects in atopic diseases. Most of the new
Potential approaches for primary and secondary prevention
approaches aim at inhibiting components of the allergic
of AD
inflammatory response, including cytokine modulation
(eg, TNF inhibitors) (185, 186), blockade of inflammatory Based on the idea of diet as modulatory, a number of
cell recruitment (chemokine receptor antagonists and controlled interventions have tested this hypothesis of
cutaneous lymphocyte antigen inhibitors) (187), and inhi- primary prevention through the nutritional route. Hydro-
bition of T-cell activation (alefacept and efalizumab) (86). lyzed cow’s milk formula consists of predigested peptides
of whey and casein. The formulas have equivalent
nutritional values but a reduced capacity to induce
IgEmediated reactions (194–197). A large controlled
Education
study in high-risk infants using different partially and
The goal of the patientsÕ education should be living with extensively hydrolyzed formulas for the first 6 months of
atopic dermatitis by means of an empowered patient or, life demonstrated that extensively hydrolyzed casein
in the case of infants and young children, a caregiver who formula has the capacity to reduce AD by 50% in the
can work as a partner with the doctor in selfmanaging first year of life (198, 199). A different approach for
their own or their children’s disease. primary prevention is suggested by the introduction of
Education to enhance disease knowledge, psychologic probiotics (Lactobacillus GG) into the maternal and
improvement in disease perception, and scratch control infantile diet. One study reported a decreased incidence of
behavior modification, together with regular daily treat- AD but had no effect on allergic sensitization (200).
ment, will lead to better skin care. This improvement in Elimination diets in the mother are not recommended
disease control will restore family dynamics, and the because of their limited success. Other potential avenues
patient and family will cope better and have an overall that are being explored include adding bacterial, myco-
improvement in quality of life. Additionally, education bacterial, and parasitic materials into the infant diet.
should be aimed at reducing doctor shopping, facilitating A secondary prevention trial with cetirizine for
a better partnership between the doctor and the patient- infants with AD and a positive family history of
parent, and decreasing the long-termcosts of chronic asthma failed to demonstrate an effect for the whole
disease treatment. group. In a subset of patients with dust mite or grass
Many preliminary studies have been single nurse-led pollen sensitization, the incidence of asthma was
interventions that were usually not controlled to assess reduced by 50% (20). This concept is currently being
outcomes (188). From the recent controlled studies, there is investigated in a second trial of 500 children (20). Trials
the general impression that positive outcomes are depend- using environmental control measures have shown
ant on the time spent with parents and the qualification of potential effects in AD severity in children, although
the trainer (189–191). Sharing personal experiences in not in adults (16). Another secondary prevention trial is
managing AD was helpful in 80% of those parents under investigation based on the early treatment of AD
attending the program conducted by Staab et al. (192). with topical pimecrolimus to prevent the progression of
A 12-lesson educational program(192) described pos- AD to asthma based on the concept that the skin is the
itive outcomes after 1 year, including diminished fear of primary site of sensitization.
topical corticosteroid cream use. In a recent German This document represents a consensus of an interna-
multicenter study 820 children with AD were randomized tional panel of experts from the European Academy of
into an intervention group (n ¼ 443) and a control group Allergology and Clinical Immunology and the American
(n ¼ 377) (193) The intervention group underwent a 12- Academy of Allergy, Asthma and Immunology. These
hour education program on an outpatient basis. After common proposals were developed to aid in the diagnosis
1 year, the overall Severity Score for Atopic Dermatitis and treatment of AD on both sides of the Atlantic.
(SCORAD) measure, quality of life, scratching index, and
adherence to treatment showed statistically significant
improvement.
Conflict of interest
Fundamentally, each patient with AD should be
educated on various aspects of the disease. For economic T. Bieber has consultant arrangements with Novartis and
and practical reasons, structured education will target Schering. C. Bindslev-Jensen serves on the advisory board

981
Akdis et al.

for Schering-Plough. A. Kapp has received grants/ research support from Novartis, Astellas, and Sinclair
research support from Novartis, Astellas, UCB, ALK, and has received lecture honoraria from Novartis and
and DPO and served on the speakersÕ bureau for Astellas. D. Y. M. Leung has received grants/research
Novartis, Astellas, UCB, and ALK. K. Turjanmaa a support from Novartis and has served on the speakersÕ
grant and research support from Ansell. U. Wahn has bureau for Novartis and Astellas. L. Rosenwasser has
received grants and lecture honoraria from Novartis, consultant arrangements with Novartis and Genentech. J.
MSD, GSK, UCB-Pharma, ALK, and Stallergenes. T. Spergel has received grants/research support from Nov-
Zuberbier has served on the speakersÕ bureau for Nov- artis and the NIH and served on the speakersÕ bureau for
artis, Schering-Plough, UCB, Schering, MSD, Staller- GSK and Astellas. The rest of the authors have declared
genes, and Leti. M. Boguniewicz has received grants/ that they have no conflict of interest.

References
1. Anonymous. Worldwide variation in 11. Novak N, Kruse S, Potreck J, 20. Warner JOETAC Study Group. A
prevalence of symptoms of asthma, Weidinger S, Fimmers R, Bieber T. double-blinded, randomized, placebo-
allergic rhinoconjunctivitis, and atopic Single nucleotide polymorphisms of the controlled trial of cetirizine in prevent-
eczema: ISAAC. The International IL18 gene are associated with atopic ing the onset of asthma in children with
Study of Asthma and Allergies in eczema. J Allergy Clin Immunol atopic dermatitis: 18 monthsÕ treatment
Childhood (ISAAC) Steering Commit- 2005;115:828–33. and 18 monthsÕ posttreatment follow-
tee. Lancet 1998;351:1225–32. 12. Weidinger S, Klopp N, Rümmler L, up. J Allergy Clin Immunol
2. Kay J, Gawkrodger DJ, Mortimer MJ, Wagenpfeil S, Novak N, Baurecht HJ 2001;108:929–37.
Jaron AG. The prevalence of childhood et al. Association of NOD1 polymor- 21. Gustafsson D, Sjoberg O, Foucard T.
atopic eczema in a general population. J phisms with atopic eczema and related Development of allergies and asthma in
Am Acad Dermatol 1994;30:35–9. phenotypes. J Allergy Clin Immunol infants and young children with atopic
3. Illi S, von Mutius E, Lau S, Nickel R, 2005;116:177–84. dermatitis—a prospective follow-up to
Gruber C, Niggemann B et al. The 13. Weidinger S, Rümmler L, Klopp N, 7 years of age. Allergy 2000;55:240–5.
natural course of atopic dermatitis from Wagenpfeil S, Baurecht HJ, Fischer G 22. Spergel JM, Mizoguchi E, Brewer JP,
birth to age 7 years and the association et al. Association study of mast cell Martin TR, Bhan AK, Geha RS. Epi-
with asthma. J Allergy Clin Immunol chymase polymorphisms with atopy. cutaneous sensitization with protein
2004;113:925–31. Allergy 2005;60:1256–61. antigen induces localized allergic der-
4. Williams HC, Strachan DP. The natural 14. Laubereau B, Brockow I, Zirngibl A, matitis and hyperresponsiveness to
history of childhood eczema: observa- Koletzko S, Gruebl A, von Berg A et al. methacholine after single exposure to
tions from the British 1958 birth cohort Effect of breast-feeding on the devel- aerosolized antigen in mice. J Clin In-
study. Br J Dermatol 1998;139:834–9. opment of atopic dermatitis during the vest 1998;101:1614–22.
5. Ozkaya E. Adult-onset atopic dermatitis. first 3 years of life – results from the 23. Lack G, Fox D, Northstone K, Golding
J Am Acad Dermatol 2005;52:579–82. GINI-birth cohort study. J Pediatr J, Avon Longitudinal Study of Parents
6. Hoffjan S, Epplen JT. The genetics of 2004;144:602–7. and Children Study Team. Factors
atopic dermatitis: recent findings and fu- 15. Schafer T. [Prevention of atopic eczema. associated with the development of
ture options. J Mol Med 2005;83:682–92. Evidence based guidelines]. Hautarzt peanut allergy in childhood. N Eng J
7. Lee YA, Wahn U, Kehrt R, Tarani L, 2005;56:232–40 Med 2003;348:977–85.
Businco L, Gustafsson D et al. A major 16. Capristo C, Romei I, Boner AL. Envi- 24. Novak N, Bieber T, Leung DY. Im-
susceptibility locus for atopic dermatitis ronmental prevention in atopic eczema mune mechanisms leading to atopic
maps to chromosome 3q21. Nat Genet dermatitis syndrome (AEDS) and asth- dermatitis. J Allergy Clin Immunol
2000;26:470–3. ma: avoidance of indoor allergens. Al- 2003;112(suppl):S128–39.
8. Cookson WO, Ubhi B, Lawrence R, lergy 2004;59(Suppl. 78):53–60. 25. Leung DYM, Bhan AK, Schneeberger
Abecasis GR, Walley AJ, Cox HE et al. 17. Purvis DJ, Thompson JM, Clark PM, EE, Geha RS. Characterization of the
Genetic linkage of childhood atopic Robinson E, Black PN, Wild CJ et al. mononuclear cell infiltrate in atopic
dermatitis to psoriasis susceptibility Risk factors for atopic dermatitis in dermatitis using monoclonal antibodies.
loci. Nat Genet 2001;27:372–3. New Zealand children at 3.5 years of J Allergy Clin Immunol 1983;71:47–56.
9. Bradley M, Soderhall C, Luthman H, age. Br J Dermatol 2005;152:742–9. 26. Novak N, Kraft S, Bieber T. IgE
Wahlgren CF, Kockum I, Nordenskjold 18. Schafer T, Heinrich J, Wjst M, Adam receptors. Curr Opin Immunol
M. Susceptibility loci for atopic der- H, Ring J, Wichmann HE. Association 2001;13:721–6.
matitis on chromosomes 3, 13, 15, 17 between severity of atopic eczema and 27. Leung DY, Boguniewicz M, Howell
and 18 in a Swedish population. Hum degree of sensitization to aeroallergens MD, Nomura I, Hamid QA. New in-
Mol Genet 2002;11:1539–48. in schoolchildren. J Allergy Clin sights into atopic dermatitis. J Clin In-
10. Haagerup A, Bjerke T, Schiotz PO, Immunol 1999;104:1280–4. vest 2004;113:651–7.
Dahl R, Binderup HG, Tan Q et al. 19. Eigenmann PA, Sicherer SH, 28. Hamid Q, Boguniewicz M, Leung DY.
Atopic dermatitis—a total genome-scan Borkowski TA, Cohen BA, Sampson Differential in situ cytokine gene
for susceptibility genes. Acta Derm HA. Prevalence of IgE-mediated food expression in acute versus chronic der-
Venereol 2004;84:346–52. allergy among children with atopic matitis. J Clin Invest 1994;94:870–6.
dermatitis. Pediatrics 1998;101:e8.

982
PRACTALL Consensus Report

29. Bratton DL, Hamid Q, Boguniewicz M, 39. Kerschenlohr K, Decard S, Darsow U, 49. Chan LS, Robinson N, Xu L. Expres-
Doherty DE, Kailey JM, Leung DY. Ollert M, Wollenberg A. Clinical and sion of interleukin-4 in the epidermis of
Granulocyte macrophage colony-sti- immunologic reactivity to aeroallergens transgenic mice results in a pruritic
mulating factor contributes to enhanced in ÔÔintrinsicÕÕ atopic dermatitis patients. inflammatory skin disease: an experi-
monocyte survival in chronic atopic J Allergy Clin Immunol 2003;111:195–7. mental animal model to study atopic
dermatitis. J Clin Invest 1995;95:211–8. 40. Novembre E, Cianferoni A, Lombardi dermatitis. J Invest Dermatol
30. Toda M, Leung DY, Molet S, E, Bernardini R, Pucci N, Vierucci A. 2001;117:977–83.
Boguniewicz M, Taha R, Natural history of ÔÔintrinsicÕÕ atopic 50. Akdis M, Trautmann A, Klunker S,
Christodoulopoulos P et al. Polarized in dermatitis. Allergy 2001;56:452–3. Daigle I, Kucuksezer UC, Deglmann W
vivo expression of IL-11 and Il-17 be- 41. Novak N, Tepel C, Koch S, Brix K, et al. T helper (Th) 2 predominance in
tween acute and chronic skin lesions. J Bieber T, Kraft S. Evidence for a dif- atopic diseases is due to preferential
Allergy Clin Immunol 2003;111:875–81. ferential expression of the apoptosis of circulating memory/effector
31. Taha RA, Minshall EM, Leung DY, FcepsilonRIgamma chain in dendritic Th1 cells. FASEB J 2003;17:1026–35.
Boguniewicz M, Luster A et al. Evi- cells of atopic and nonatopic donors. J 51. Akdis CA, Blaser K, Akdis M. Apop-
dence for increased expression of eo- Clin Invest 2003;111:1047–56. tosis in tissue inflammation and allergic
taxin and monocyte chemotactic 42. Sator PG, Schmidt JB, Honigsmann H. disease. Curr Opin Immunol
protein-4 in atopic dermatitis. J Allergy Comparison of epidermal hydration 2004;16:717–23.
Clin Immunol 2000;105:1002–7. and skin surface lipids in healthy indi- 52. Umetsu DT, Akbari O, Dekruyff RH.
32. Hijnen D, De Bruin-Weller M, Oosting viduals and in patients with atopic der- Regulatory T cells control the develop-
B, Lebre C, De Jong E, Bruijnzeel- matitis. J Am Acad Dermatol ment of allergic disease and asthma. J
Koomen C et al. Serum thymus and 2003;48:352–8. Allergy Clin Immunol 2003;112:480–7.
activation-regulated chemokine 43. Arikawa J, Ishibashi M, Kawashima M, 53. Akdis M, Verhagen J, Taylor A,
(TARC) and cutaneous T cell–attract- Takagi Y, Ichikawa Y, Imokawa G. Karamloo F, Karagiannidis C, Crameri
ing chemokine (CTACK) levels in Decreased levels of sphingosine, a nat- R et al. Immune responses in healthy
allergic diseases: TARC and CTACK ural antimicrobial agent, may be asso- and allergic individuals are character-
are disease-specific markers for atopic ciated with vulnerability of the stratum ized by a fine balance between allergen-
dermatitis. J Allergy Clin Immunol corneum from patients with atopic der- specific T regulatory 1 and T helper 2
2004;113:334–40. matitis to colonization by Staphylococ- cells. J Exp Med 2004;199:1567–75.
33. Echigo T, Hasegawa M, Shimada Y, cus aureus. J Invest Dermatol 54. Carneiro R, Reefer A, Wilson B,
Takehara K, Sato S. Expression of 2002;119:433–9. Hammer J, Platts-Mills J, Custis N
fractalkine and its receptor, CX3CR1, 44. Rippke F, Schreiner V, Doering T, et al. T cell epitope-specific defects in
in atopic dermatitis: possible contribu- Maibach HI. Stratum corneum pH in the immune response to cat allergen in
tion to skin inflammation. J Allergy atopic dermatitis: impact on skin barrier patients with atopic dermatitis. J Invest
Clin Immunol 2004;113:940–8. function and colonization with Staphy- Dermatol 2004;122:927–36.
34. Klunker S, Trautmann A, Akdis M, lococcus aureus. Am J Clin Dermatol 55. Lin W, Truong N, Grossman WJ,
Verhagen J, Schmid-Grendelmeier P, 2004;5:217–23. Haribhai D, Williams CB, Wang J et al.
Blaser K et al. A second step of chemo- 45. Vasilopoulos Y, Cork MJ, Murphy R, Allergic dysregulation and hyperimmu-
taxis after transendothelial migration: Williams HC, Robinson DA, Duff GW noglobulinemia E in Foxp3 mutant
keratinocytes undergoing apoptosis re- et al. Genetic association between an mice. J Allergy Clin Immunol
lease IFN-gamma-inducible protein 10, AACC insertion in the 3ÕUTR of the 2005;116:1106–15.
monokine induced by IFN-gamma, and stratum corneum chymotryptic enzyme 56. Ou LS, Goleva E, Hall C, Leung DY. T
IFN-gamma-inducible alpha-chemoat- gene and atopic dermatitis. J Invest regulatory cells in atopic dermatitis and
tractant for T cell chemotaxis toward Dermatol 2004;123:62–6. subversion of their activity by superan-
epidermis in atopic dermatitis. J Immu- 46. Baud O, Goulet O, Canioni D, Le Deist tigens. J Allergy Clin Immunol
nol 2003;171:1078–84. F, Radford I, Rieu D et al. Treatment 2004;113:756–63.
35. Novak N, Bieber T. Allergic and non- of the immune dysregulation, poly- 57. Novak N, Valenta R, Bohle B, Laffer S,
allergic forms of atopic diseases. J Al- endocrinopathy, enteropathy, X-linked Haberstok J, Kraft S et al. Fcepsilon RI
lergy Clin Immunol 2003;112:252–62. syndrome (IPEX) by allogeneic bone engagement of Langerhans cell-like
36. Schmid-Grendelmeier P, Simon D, marrow transplantation. N Engl J Med dendritic cells and inflammatory dend-
Simon HU, Akdis CA, Wuthrich B. 2001;344:1758–62. ritic epidermal cell-like dendritic cells
Epidemiology, clinical features, and 47. Spergel JM, Mizoguchi E, Oettgen H, induces chemotactic signals and differ-
immunology of the ÔÔintrinsicÕÕ (non-IgE- Bhan AK, Geha RS. Roles of Th1 and ent T-cell phenotypes in vitro. J Allergy
mediated) type of atopic dermatitis Th2 cytokines in a murine model of Clin Immunol 2004;113:949–57.
(constitutional dermatitis). Allergy allergic dermatitis. J Clin Invest 58. Wollenberg A, Wagner M, Gunther S,
2001;56:841–9. 1999;103:1103–11. Towarowski A, Tuma E, Moderer M
37. Scheynius A, Johansson C, Buentke E, 48. Hoetzenecker W, Ecker R, Kopp T, et al. Plasmacytoid dendritic cells: a new
Zagari A, Linder MT. Atopic eczema/ Stuetz A, Stingl G, Elbe-Burger A. Pi- cutaneous dendritic cell subset with dis-
dermatitis syndrome and Malassezia. mecrolimus leads to an apoptosis-in- tinct role in inflammatory skin diseases. J
Int Arch Allergy Immunol duced depletion of T cells, but not Invest Dermatol 2002;119:1096–102.
2002;127:161–9. Langerhans cells in patients with atopic
38. Novak N, Allam JP, Bieber T. Allergic dermatitis. J Allergy Clin Immunol
hyperreactivity to microbial compo- 2005;115:1276–83.
nents: a trigger factor of ÔÔintrinsicÕÕ
atopic dermatitis? J Allergy Clin
Immunol 2003;112:215–6.

983
Akdis et al.

59. Novak N, Allam JP, Hagemann T, 69. Schmid-Ott G, Jaeger B, Adamek C, 80. Bilsborough J, Leung DY, Maurer M,
Jenneck C, Laffer S, Valenta R et al. Koch H, Lamprecht F, Kapp A et al. Howell M, Boguniewcz M, Yao L et al.
Characterization of FcepsilonRI-bear- Levels of circulating CD8+ T-lympho- IL-31 is associated with cutaneous
ing CD123 blood dendritic cell antigen- cytes, natural killer cells and eosinophils lymphocyte antigen-positive skin hom-
2 plasmacytoid dendritic cells in atopic increase upon acute psychosocial stress ing T cells in patients with atopic der-
dermatitis. J Allergy Clin Immunol in patients with atopic dermatitis. J Al- matitis. J Allergy Clin Immunol
2004;114:364–70. lergy Clin Immunol 2001;107:171–7. 2006;117:418–25.
60. Sayama K, Komatsuzawa H, Yamasaki 70. Wedi B, Raap U, Lewrick H, Kapp A. 81. Sonkoly E, Muller A, Lauerma AI,
K, Shirakata Y, Hanakawa Y, Ouhara Delayed eosinophil programmed cell Pivarcsi A, Soto H, Kemeny L et al. IL-
K et al. New mechanisms of skin innate death in vitro: a common feature of 31: a new link between T cells and
immunity: ASK1-mediated keratinocyte inhalant allergy and extrinsic and pruritus in atopic skin inflammation. J
differentiation regulates the expression intrinsic atopic dermatitis. J Allergy Allergy Clin Immunol 2006;117:411–7.
of b-defensins, LL37, and TLR2. Eur J Clin Immunol 1997;100:536–43. 82. Toyoda M, Nakamura M, Makino T,
Immunol 2005;35:1886–95. 71. Grewe M, Czech W, Morita A, Werfel Hino T, Kagoura M, Morohashi M.
61. Giustizieri ML, Mascia F, Frezzolini A, T, Klammer M, Kapp A et al. Human Nerve growth factor and substance P
De Pita O, Chinni LM, Giannetti A eosinophils produce biologically active are useful plasma markers of disease
et al. Keratinocytes from patients with IL-12: implications for control of T cell activity in atopic dermatitis. Br J Der-
atopic dermatitis and psoriasis show a responses. J Immunol 1998;161:415–20. matol 2002;147:71–9.
distinct chemokine production profile in 72. Raap U, Goltz C, Deneka N, Bruder M, 83. Sampson HA. Update on food allergy. J
response to T cell-derived cytokines. J Renz H, Kapp A et al. Brain-derived Allergy Clin Immunol 2004;113:805–19.
Allergy Clin Immunol 2001;107:871–7. neurotrophic factor is increased in 84. Werfel T, Breuer K. Role of food al-
62. Yoo J, Omori M, Gyarmati D, Zhou B, atopic eczema and modulates eosinophil lergy in atopic dermatitis. Curr Opin
Aye T, Brewer A et al. Spontaneous functions compared with that seen in Allergy Clin Immunol 2004;4:379–85.
atopic dermatitis in mice expressing an nonatopic subjects. J Allergy Clin 85. Darsow U, Laifaoui J, Kerschenlohr K,
inducible thymic stromal lymphopoietin Immunol 2005;115:1268–75. Wollenberg A, Przybilla B, Wuthrich B
transgene specifically in the skin. J Exp 73. Leung DY, Bieber T. Atopic dermatitis. et al. The prevalence of positive reac-
Med 2005;202:541–9. Lancet 2003;361:151–60. tions in the atopy patch test with aero-
63. Trautmann A, Akdis M, Kleeman D, 74. Leo HL, Bender BG, Leung SB, Tran allergens and food allergens in subjects
Altznauer F, Simon H-U, Graeve T ZV, Leung DY. Effect of pimecrolimus with atopic eczema: a European multi-
et al. T cell-mediated Fas-induced ker- cream 1% on skin condition and sleep center study. Allergy 2004;59:1318–25.
atinocyte apoptosis plays a key patho- disturbance in children with atopic 86. Leung DY. Infection in atopic derma-
genetic role in eczematous dermatitis. J dermatitis. J Allergy Clin Immunol titis. Curr Opin Pediatr 2003;15:399–
Clin Invest 2000;106:25–35. 2004;114:691–93. 404.
64. Pastore S, Corinti S, La Placa M, 75. Klein PA, Clark RA. An evidence- 87. Leung DYM, Harbeck H, Bina P,
Didona B, Girolomoni G. Interferon- based review of the efficacy of antihis- Reiser RF, Yang E, Norris AD et al.
gamma promotes exaggerated cytokine tamines in relieving pruritus in atopic Presence of IgE antibodies to staphylo-
production in keratinocytes cultured dermatitis. Arch Dermatol coccal enterotoxins on the skin of pa-
from patients with atopic dermatitis. J 1999;135:1522–25. tients with atopic dermatitis: evidence
Allergy Clin Immunol 1998;101:538–44. 76. Eichenfield LF, Lucky AW, for a new group of allergens. J Clin
65. Trautmann A, Altznauer F, Akdis M, Boguniewicz M, Langley RG, Cherill R, Invest 1993;92:1374–80.
Simon H-U, Disch R, Bröcker E-B Marshall K et al. Safety and efficacy of 88. Hauk PJ, Leung DYM. Tacrolimus
et al. The differential fate of cadherins pimecrolimus (ASM 981) cream 1% in (FK506): new treatment approach in
during T cell-induced keratinocyte the treatment of mild and moderate superantigen-associated diseases like
apoptosis leads to spongiosis in ecze- atopic dermatitis in children and ado- atopic dermatitis? J Allergy Clin
matous dermatitis. J Invest Derm lescents. J Am Acad Dermatol Immunol 2001;107:391–2.
2001;117:927–34. 2002;46:495–504. 89. Ong PY, Ohtake T, Brandt C,
66. Trautmann A, Akdis M, Schmid- 77. Van Der Meer JB, Glazenburg EJ, Strickland I, Boguniewicz M, Ganz T
Grendelmeier P, Disch R, Bröcker E-B, Mulder PG, Eggink HF, Coenraads PJ. et al. Endogenous antimicrobial pep-
Blaser K et al. Targeting keratinocyte The management of moderate to severe tides and skin infections in atopic der-
apoptosis in the treatment of atopic atopic dermatitis in adults with topical matitis. N Engl J Med 2002;347:1151–
dermatitis and allergic contact dermati- fluticasone propionate. The Nether- 60.
tis. J Allergy Clin Immunol lands Adult Atopic Dermatitis Study 90. Nomura I, Goleva E, Howell MD,
2001;108:839–46. Group. Br J Dermatol 1999;140:1114– Hamid QA, Ong PY, Hall CF et al.
67. Kapp A. The role of eosinophils in the 21. Cytokine milieu of atopic dermatitis, as
pathogenesis of atopic dermatitis – eo- 78. Dillon SR, Sprecher C, Hammond A, compared to psoriasis, skin prevents
sinophil granule proteins as markers of Bilsborough J, Rosenfeld-Franklin M, induction of innate immune response
disease activity. Allergy 1993;48:1–5. Presnell SR et al. Interleukin 31, a cy- genes. J Immunol 2003;171:3262–9.
68. Simon D, Braathen LR, Simon HU. tokine produced by activated T cells, 91. Howell MD, Wollenberg A, Gallo RL,
Eosinophils and atopic dermatitis. induces dermatitis in mice. Nat Immu- Flaig M, Streib JE, Wong C et al.
Allergy 2004;59:561–70. nol 2004;5:752–60. Cathelicidin deficiency predisposes to
79. Takaoka A, Arai I, Sugimoto M, eczema herpeticum. J Allergy Clin
Yamaguchi A, Tanaka M, Nakaike S. Immunol 2006;117:836–41.
Expression of IL-31 gene transcripts in 92. Plaut M, Tinkle SS. Risks of smallpox
NC/Nga mice with atopic dermatitis. vaccination: 200 years after Jenner. J
Eur J Pharmacol 2005;516:180–1. Allergy Clin Immunol 2003;112:683–5.

984
PRACTALL Consensus Report

93. Faergemann J. Pityrosporum species as 105. Sampson HA. Utility of food-specific 118. Ellis C, Luger Ton behalf of the ICCAD
a cause of allergy and infection. Allergy IgE concentrations in predicting symp- II Faculty. International consensus
1999;54:413–9. tomatic food allergy. J Allergy Clin conference on atopic dermatitis II (IC-
94. Scalabrin DM, Bavbek S, Perzanowski Immunol 2001;107:891–6. CAD II*): clinical update and current
MS, Wilson BB, Platts-Mills TA, 106. Osterballe M, Bindslev-Jensen C. treatment strategies. Br J Dermatol
Wheatley LM et al. Use of specific IgE Threshold levels in food challenge and 2003;148:3–10.
in assessing the relevance of fungal and specific IgE in patients with egg allergy: 119. Rudolph R, Kownatzki E. Corneomet-
dust mite allergens to atopic dermatitis: is there a relationship? J Allergy Clin ric, sebumetric and TEWL measure-
a comparison with asthmatic and non- Immunol 2003;112:196–201. ments following the cleaning of atopic
asthmatic control subjects. J Allergy 107. Celik-Bilgili S, Mehl A, Verstege A, skin with a urea emulsion versus a
Clin Immunol 1999;104:1273–9. Staden U, Nocon M, Beyer K et al. The detergent cleanser. Contact Dermatitis
95. Schmid-Grendelmeier P, Scheynius A, predictive value of specific immuno- 2004;50:354–8.
Crameri R. The role of sensitization to globulin E levels in serum for the out- 120. Darsow U, Lubbe J, Taieb A, Seidenari
Malassezia sympodialis in atopic ecze- come of oral food challenges. Clin Exp S, Wollenberg A, Calza AM et al.
ma. Chem Immunol Allergy Allergy 2005;35:268–73. Position paper on diagnosis and treat-
2006;91:98–109. 108. Turjanmaa K. The role of atopy patch ment of atopic dermatitis. J Eur Acad
96. Mothes N, Niggemann B, Jenneck C, tests in the diagnosis of allergy in atopic Dermatol Venereol 2005;19:286–95.
Hagemann T, Weidinger S, Bieber T dermatitis. Curr Opin Allergy Clin 121. Hanifin JM, Cooper KD, Ho VC, Kang
et al. The cradle of IgE hyperreactivity Immunol 2005;5:425–8. S, Krafchik BR, Margolis DJ et al.
in atopic eczema lies in infancy. J Al- 109. Mitchell EB, Crow J, Chapman MD, Guidelines of care for atopic dermatitis,
lergy Clin Immunol 2005;116:706–9. Jouhal SS, Pope FM, Platts-Mills TA. developed in accordance with the
97. Natter S, Seiberler S, Hufnagl P, Binder Basophils in allergen-induced patch test American Academy of Dermatology
BR, Hirschl AM, Ring J et al. Isolation sites in atopic dermatitis. Lancet (AAD)/American Academy of Derma-
of cDNA clones coding for IgE auto- 1982;1:127–30. tology Association ÔÔAdministrative
antigens with serum IgE from atopic 110. Tan BB, Weald D, Strickland I, Regulations for Evidence-Based Clin-
dermatitis patients. FASEB J Friedmann PS. Double-blind controlled ical Practice GuidelinesÕÕ. J Am Acad
1998;12:1559–69. trial of effect of housedust-mite allergen Dermatol 2004;50:391–404.
98. Valenta R, Seiberler S, Natter S, Mahler avoidance on atopic dermatitis. Lancet 122. Korting HC, Kerscher MJ, Schafer-
V, Mossabeb R, Ring J et al. Autoal- 1996;347:15–8. Korting M. Topical glucocorticoids
lergy – a pathogenetic factor in atopic 111. Platts-Mills TA, Mitchell EB, Rowntree with improved benefit/risk ratio: do
dermatitis. J Allergy Clin Immunol S, Chapman MD, Wilkins SR. The role they exist? J Am Acad Dermatol
2000;105:432–7. of dust mite allergens in atopic derma- 1992;27:87–92.
99. Schmid-Grendelmeier P, Fluckiger S, titis. Clin Exp Dermatol 1983;8:233–47. 123. Kerscher MJ, Hart H, Korting HC,
Disch R, Trautmann A, Wuthrich B, 112. Morren MA, Przybilla B, Bamelis M, Stalleicken D. In vivo assessment of the
Blaser K et al. IgE-mediated and T cell- Heykants B, Reynaers A, Degreef H. atrophogenic potency of mometasone
mediated autoimmunity against man- Atopic dermatitis: triggering factors. J furoate, a newly developed chlorinated
ganese superoxide dismutase in atopic Am Acad Dermatol 1994;31:467–73. potent topical glucocorticoid as com-
dermatitis. J Allergy Clin Immunol 113. Subramanyan K. Role of mild cleansing pared to other topical glucocorticoids
2005;115:1068–75. in the management of patient skin. old and new. Int J Clin Pharmacol Ther
100. Hanifin JM, Rajka G. Diagnostic fea- Dermatol Ther 2004;17:26–34. 1995;33:187–9.
tures of atopic eczema. Acta Dermatol 114. Sampson HA. Food allergy – accurately 124. Thomas KS, Armstrong S, Avery A, Po
Venereol (Stockh) 1980;92:44–7. identifying clinical reactivity. Allergy AL, O’Neill C, Young S et al. Rand-
101. Johnke H, Vach W, Norberg LA, 2005;60(Suppl. 79):19–24. omised controlled trial of short bursts of
Bindslev-Jensen C, Host A, Andersen 115. Seidenari S, Giusti G. Objective assess- a potent topical corticosteroid versus
KE. A comparison between criteria for ment of the skin of children affected by prolonged use of a mild preparation for
diagnosing atopic eczema in infants. Br atopic dermatitis: a study of pH, capa- children with mild or moderate atopic
J Dermatol 2005;153:352–8. citance and TEWL in eczematous and eczema. BMJ 2003;30:768.
102. Chatila TA. Role of regulatory T cells clinically uninvolved skin. Acta Derm 125. Berth-Jones J, Damstra RJ, Golsch S,
in human diseases. J Allergy Clin Venereol 1996;75:429–33. Livden JK, Van Hooteghem O, Allegra F
Immunol 2005;116:949–59. 116. Loden M. Role of topical emollients et al. Twice weekly fluticasone propionate
103. Bindslev-Jensen C, Ballmer-Weber BK, and moisturizers in the treatment of dry added to emollient maintenance treatment
Bengtsson U, Blanco C, Ebner C, skin barrier disorders. Am J Clin Der- to reduce risk of relapse in atopic derma-
Hourihane J et al. Standardization of matol 2003;4:771–88. titis: randomised, double blind, parallel
food challenges in patients with imme- 117. McHenry PM, Williams HC, Bingham group study. BMJ 2003;326:1367.
diate reactions to foods—position paper EA. Management of atopic eczema: 126. Stalder JF, Fleury M, Sourisse M,
from the European Academy of Al- Joint Workshop of the British Associ- Rostin M, Pheline F, Litoux P. Local
lergology and Clinical Immunology. ation of Dermatologists and the Re- steroid therapy and bacterial skin flora
Allergy 2004;59:690–7. search Unit of the Royal College of in atopic dermatitis. Br J Dermatol
104. Breuer K, Heratizadeh A, Wulf A, Physicians of London. BMJ 1994;131:536–40.
Baumann U, Constien A, Tetau D et al. 1995;310:843–7. 127. Nilsson EJ, Henning CG, Magnusson J.
Late eczematous reactions to food in Topical corticosteroids and Staphylo-
children with atopic dermatitis. Clin coccus aureus in atopic dermatitis. J Am
Exp Allergy 2004;34:817–24. Acad Dermatol 1992;27:29–34.

985
Akdis et al.

128. Ainley-Walker PF, Patel L, David TJ. 139. Elidel [package insert]. Basel, Switzer- 154. Verbist I. The antimicrobial activity of
Side to side comparison of topical land: Novartis Pharmaceuticals Corp.; fucidic acid. J Antimicrob Chemother
treatment in atopic dermatitis. Arch Dis 2006. 1990;25(Suppl. B):1–5.
Child 1998;79:149–52. 140. Protopic [package insert. Deerfield, Ill: 155. Ravenscroft JC, Layton A, Barnham
129. Hultsch T, Kapp A, Spergel J. Immu- Astellas Pharma Manufacturing Inc.; M. Observations on high levels of fusi-
nomodulation and safety of topical 2006. dic acid resistant Staphylococcus aureus
calcineurin inhibitors for the treatment 141. Mallon E, Powell S, Bridgman A. ÔÔWet- in Harrogate, North Yorkshire, UK.
of atopic dermatitis. Dermatology wrapÕÕ dressings for the treatment of Clin Exp Dermatol 2000;25:327–30.
2005;211:174–87. atopic eczema in the community. J 156. Peeters KABM, Mascini EM, Sanders
130. Wahn U, Bos JD, Goodfield M, Caputo Dermatolog Treat 1994;5:97–8. CJG. Resistance of Staphylococcus
R, Papp K, Manjra A et al. Efficacy and 142. Oranje AP, Wolkerstorfer A, de Waard- aureus to fusidic acid. Int J Dermatol
safety of pimecrolimus cream in the long- van der Spek FB. Treatment of eryth- 2004;43:235–7.
term management of atopic dermatitis in rodermic atopic dermatitis with ÔÔwet- 157. Ravenscroft JC, Layton AM, Eady EA,
children. Pediatrics 2002;110:e2. wrapÕÕ fluticasone propionate 0.05% Murtagh MS, Coates P, Walker M et al.
131. Harper J, Green A, Scott G, Gruendl E, cream/emollient 1:1 dressings. J Der- Short-term effects of topical fusidic acid
Dorobek B, Cardno B et al. First matolog Treat 1999;10:73–4. or mupirocin on the prevalence of fusi-
experience of topical SDZ ASM 981 in 143. Foelster-Holst R, Nagel F, Zoellner P, dic acid resistant (FusR) Staphylococcus
children with atopic dermatitis. Br J Spaeth D. Efficacy of crisis intervention aureus in atopic eczema. Br J Dermatol
Dermatol 2001;144:781–7. treatment with topical corticosteroid 2003;148:1010–7.
132. Ashcroft DM, Dimmock P, Garside R, prednicarbat with and without partial 158. Lubbe J. Secondary infections in pa-
Stein K, Williams HC. Efficacy and wet-wrap dressing in atopic dermatitis. tients with atopic dermatitis. Am J Clin
tolerability of topical pimecrolimus and Dermatology 2006;212:66–9. Dermatol 2003;4:641–54.
tacrolimus in the treatment of atopic 144. Boguniewicz M, Nicol N. Conventional 159. Hoeger PH. Antimicrobial susceptibility
dermatitis: meta-analysis of randomised therapy. Immunol Allergy Clinics N of skin-colonizing S. aureus strains in
controlled trials. BMJ 2005;330:516. Am 2002;22:107–24. children with atopic dermatitis. Pediatr
133. Luger T, Van Leent EJ, Graeber M, 145. Breuer K, Kapp A, Werfel T. Bacterial Allergy Immunol 2004;15:474–7.
Hedgecock S, Thurston M, Kandra A infections and atopic dermatitis. Allergy 160. Boguniewicz M, Sampson H, Harbeck
et al. SDZ ASM 981: an emerging safe 2001;56:1034–41. R, Leung DYM. Effects of cefuroxime
and effective treatment for atopic der- 146. Breuer K, HAussler S, Kapp A, Werfel axetil on S. aureus colonization and
matitis. Br J Dermatol 2001;144:788–94. T. Staphylococcus aureus: colonizing superantigen production in atopic der-
134. Paul C, Cork M, Rossi AB, Papp KA, features and influence of an antibacte- matitis. J Allergy Clin Immunol
Barbier N, de Prost Y. Safety and tol- rial treatment in adults with atopic 2001;108:651–2.
erability of 1% pimecrolimus cream dermatitis. Br J Dermatol 2002;147:55– 161. Lintu P, Savolainen J, Kortekangas-
among infants: experience with 1133 61. Savolainen O, Kalimo K. Systemic ke-
patients treated for up to 2 years. Pe- 147. Sporik R, Kemp AS. Topical triclosan toconazole is an effective treatment of
diatrics 2006;117:118–28. treatment of atopic dermatitis. J Allergy atopic dermatitis with IgE-mediated
135. Breuer K, Werfel T, Kapp A. Safety Clin Immunol 1997;99:861. hypersensitivity to yeasts. Allergy
and efficacy of topical calcineurin 148. Bhargava HN, Leonhard PA. Triclosan: 2001;56:512–7.
inhibitors in the treatment of childhood application and safety. Am J Infect 162. Baeck O, Bartosik J. Systemic ketoc-
atopic dermatitis. Am J Clin Dermatol Control 1996;24:209–18. onazole for yeast allergic patients with
2005;6:65–77. 149. Gauger A, Mempel M, Schekatz A, atopic dermatitis. J Eur Acad Derm
136. Kempers S, Boguniewicz M, Carter E, Schafer T, Ring J, Abeck D. Silver- Venereol 2001;15:34–8.
Jarratt M, Pariser D, Stewart D et al. A coated textiles reduce Staphylococcus 163. Nikkels AF, Piérard GE. Framing the
randomized inverstigator-blinded study aureus colonization in patients with future of antifungals in atopic dermati-
comparing pimecrolimus cream 1% atopic eczema. Dermatology tis. Dermatology 2003;206:398–400.
with tacrolimus 0.03% in the treatment 2003;207:15–21. 164. Aylett SE, Atherton DJ, Preece MA.
of pediatric patients with moderate 150. Ricci G, Patrizi A, Bendandi B, Menna The treatment of difficult atopic der-
atopic dermatitis. J Am Acad Dermatol G, Varotti E, Masi M. Clinical effect- matitis in childhood with oral beclo-
2004;51:515–25. iveness of a silk fabric in the treatment methasone dipropionate. Acta Derm
137. Queille-Roussel C, Paul C, Duteil L, of atopic dermatitis. Br J Dermatol Venereol Suppl 1992;176:123–5.
Lefebvre MC, Rapatz G, Zagula M 2004;150:127–31. 165. Sowden JM, Berth-Jones J, Ross JS,
et al. The new topical ascomycin deri- 151. Leyden JJ, Kligman AM. The case for Motley RJ, Marks R, Finlay AY et al.
vate SDZ ASM 981 does not induce steroid-antibiotic combination. Br J Double-blind, controlled, crossover
skin atrophy when applied to normal Dermatol 1977;96:179–87. study of cyclosporin in adults with se-
skin for 4 weeks: a randomized, double- 152. Jappe U, Schnuch A, Uter W. Fre- vere refractory atopic dermatitis. Lancet
blind controlled study. Br J Dermatol quency of sensitization to antimicrobi- 1991;338:137–40.
2001;144:507–13. als in patients with atopic eczema 166. Berth-Jones J, Finlay AY, Zaki I, Tan
138. Wollenberg A, Zoch C, Wetzel S, compared with nonatopic individuals: B, Goodyear H, Lewis-Jones S et al.
Plewig G, Przybilla B. Predisposing analysis of multicentre surveillance Cyclosporine in severe childhood atopic
factors and clinical features of eczema data, 1995–1999. Br J Dermatol dermatitis: a multicenter study. J Am
herpeticum: a retrospective analysis of 2003;149:87–93. Acad Dermatol 1996;34:1016–21.
100 cases. J Am Acad Dermatol 153. Abeck D, Mempel M. Staphylococcus 167. Zaki I, Emerson R, Allen BR. Treat-
2003;49:198–205. aureus colonisation in atopic dermatitis ment of severe atopic dermatitis in
and its therapeutic implications. Br J childhood with cyclosporin. Br J Der-
Dermatol 1998;139:13–16. matol 1996;135(Suppl. 48):21–4.

986
PRACTALL Consensus Report

168. Berth-Jones J, Graham-Brown RA, 180. Abeck D, Schmidt T, Fesq H, Strom K, 192. Staab D, Ruden U, Kehrt R, Wahn U.
Marks R, Camp RD, English JS, Mempel M, Brockow K et al. Long- The impact of childhood atopic derma-
Freeman K et al. Long-term efficacy term efficacy of medium-dose UVA1 titis on quality of life of family. Dema-
and safety of cyclosporin in severe adult phototherapy in atopic dermatitis. J Am tol Psychosom 2000;1:173–8.
atopic dermatitis. Br J Dermatol Acad Dermatol 2000;42:254–7. 193. Staab D, Diepgen T, Fartasch M,
1997;136:76–81. 181. Krutmann J, Diepgen TL, Luger TA, Kupfer J, Lob-Corzilius T, Ring J et al.
169. Akhavan A, Rudikoff D. The treatment Grabbe S, Meffert H, Sonnichsen N Age-related, structured education pro-
of atopic dermatitis with systemic et al. High-dose UVA1 therapy for grammes improve the management of
immunosuppressive agents. Clin Der- atopic dermatitis: results of a multicen- atopic dermatitis in children and ado-
matol 2003;21:225–40. ter trial. J Am Acad Dermatol lescents: results of the German Atopic
170. Anstey AV, Wakelin S, Reynolds NJ. 1998;38:589–93. Dermatitis Intervention Study (GA-
British Association of Dermatologists 182. Reynolds NJ, Franklin V, Gray JC, DIS). BMJ 2006;332:933–8.
Therapy, Guidelines and Audit Sub- Diffey BL, Farr PM. Narrow-band 194. Halken S, Hansen KS, Jacobsen HP,
committee. Guidelines for prescribing ultraviolet B and broad-band ultraviolet Estmann A, Faelling AE, Hansen LG
azathioprine in dermatology. Br J Der- A phototherapy in adult atopic eczema: et al. Comparison of a partially hydro-
matol 2004;151:1123–32. a randomised controlled trial. Lancet lyzed infant formula with two exten-
171. Tan BB, Lear JT, Gwakrdger DJ, 2001;357:2012–6. sively hydrolyzed formulas for allergy
English JSC. Azathioprine in derma- 183. Mastrandrea F. The potential role of prevention: a prospective, randomized
tology: a survey of current practice in allergen-specific sublingual immuno- study. Pediatr Allergy Immunol
the UK. Br J Dermatol 1997;136:351–5. therapy in atopic dermatitis. Am J Clin 2000;11:149–61.
172. Meggitt SJ, Reynolds NJ. Azathioprine Dermatol 2004;5:281–94. 195. Halken S. Prevention of allergic disease
for atopic dermatitis. Clin Exp Derma- 184. Werfel T, Breuer K, Rueff F, Przybilla in childhood: clinical and epidemiolog-
tol 2001;26:369–75. B, Worm M, Grewe M et al. Usefulness ical aspects of primary and secondary
173. Berth-Jones J, Takwale A, Tan E, of specific immunotherapy in patients allergy prevention. Pediatr Allergy
Barclay G, Agarwal S, Ahmed I et al. with atopic dermatitis and allergic sen- Immunol 2004;15(Suppl. 16):4–5,9–32.
Azathioprine in severe adult atopic sitization to house dust mites: a multi- 196. Mimouni Bloch A, Mimouni D,
dermatitis: a double-blind, placebo- centre, randomized, dose–response Mimouni M, Gdalevich M. Does
controlled, crossover trial. Br J Der- study. Allergy 2006;61:202–5. breastfeeding protect against allergic
matol 2002;147:324–30. 185. Jacobi A, Antoni C, Manger B, Schuler rhinitis during childhood? A meta-ana-
174. Lear JT, English JSC, Jones P, Smith G, Hertl M. Infliximab in the treatment lysis of prospective studies. Acta Pae-
AG. Retrospective review of the use of of moderate to severe atopic dermatitis. diatr 2002;91:275–9.
azathioprine in severe atopic dermatitis. J Am Acad Dermatol 2005;52:522–6. 197. Muraro A, Dreborg S, Halken S, Host
J Am Acad Dermatol 1996;35:642–3. 186. Buka RL, Resh B, Roberts B, A, Niggemann B, Aalberse R et al.
175. Snow JL, Gibson LE. The role of gen- Cunningham BB, Friedlander S. Etan- Dietary prevention of allergic diseases
etic variation in thiopurine methyl- ercept is minimally effective in 2 chil- in infants and small children. Part III:
transferase activity and the efficacy and/ dren with atopic dermatitis. J Am Acad critical review of published peer-re-
or side effects of azathioprine therapy in Dermatol 2005;53:358–9. viewed observational and interventional
dermatologic patients. Arch Dermatol 187. Dimitroff CJ, Kupper TS, Sackstein R. studies and final recommendations.
1995;131:193–7. Prevention of leukocyte migration to Pediatr Allergy Immunol 2004;15:
176. Wojnarowska F, Kirtschig G, Highet inflamed skin with a novel fluorosugar 291–307.
AS, Venning VA, Khumalo NP. British modifier of cutaneous lymphocyte- 198. von Berg A, Koletzko S, Grubl A,
Association of Dermatologists. Guide- associated antigen. J Clin Invest Filipiak-Pittroff B, Wichmann HE,
lines for the management of bullous 2004;112:1008–18. Bauer CP et al. The effect of hydrolyzed
pemphigoid. Br J Dermatol 188. Chinn DJ, Poyner T, Sibley G. Rand- cow’s milk formula for allergy preven-
2002;147:214–21. omized controlled trial of a single der- tion in the first year of life: the German
177. Wahlgren C-F, Hagermark O, matology nurse consultation in primary Infant Nutritional Intervention Study, a
Bergstrom R. The antipruritic effect of a care on the quality of life of children randomized double-blind trial. J Allergy
sedative and a non-sedative antihista- with atopic eczema. Br J Dermatol Clin Immunol 2003;111:533–40.
mine in atopic dermatitis. Br J Derma- 2002;146:432–9. 199. Muraro A, Dreborg S, Halken S, Host
tol 1990;122:545–51. 189. McSkimming J, Gleeson L, Sinclair M. A, Niggemann B, Aalberse R et al.
178. Diepgen TL. Early Treatment of the A pilot study of a support group for Dietary prevention of allergic diseases
Atopic Child Study Group. Long-term parents of children with eczema. Aus- in infants and small children. Part II.
treatment with cetirizine of infants with tralas J Dermatol 1984;25:8–11. Evaluation of methods in allergy pre-
atopic dermatitis: a multi-country, 190. Koblenzer CS, Koblenzer PJ. Chronic vention studies and sensitization mark-
double-blind, randomized, placebo- intractable atopic eczema. Arch Der- ers. Definitions and diagnostic criteria
controlled trial (the ETAC trial) over matol 1988;124:1673–7. of allergic diseases. Pediatr Allergy
18 months. Pediatr Allergy Immunol 191. Broberg A, Kalimo K, Lindblad B, Immunol 2004;15:196–205.
2002;13:278–86. Swanbeck G. Parental education in the 200. Isolauri E, Arvola T, Sutas Y, Moilanen
179. Scheinfeld NS, Tutrone WD, Weinberg treatment of childhood atopic eczema. E, Salminen S. Probiotics in the man-
JM, DeLeo VA. Phototherapy of atopic Acta Derm Venereol 1990;70:495–9. agement of atopic eczema. Clin Exp
dermatitis. Clin Dermatol 2003;21:241– Allergy 2000;30:1604–10.
48.

987

You might also like