Nothing Special   »   [go: up one dir, main page]

Atopic Dermatitis in Children

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

HHS Public Access

Author manuscript
Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Author Manuscript

Published in final edited form as:


Curr Treat Options Allergy. 2017 September ; 4(3): 355–369. doi:10.1007/s40521-017-0140-6.

Atopic Dermatitis: Early Treatment in Children


Amy Huang, MD1, Christine Cho, MD2, Donald Y.M. Leung, MD, PhD2, and Kanwaljit Brar,
MD2
1Department of Dermatology, State University of New York Downstate Medical Center, Brooklyn,
NY
2Department of Pediatrics, National Jewish Health, Denver, CO
Author Manuscript

OPINION STATEMENT
Therapeutic regimens for the treatment and long-term management of AD traditionally had a two-
fold objective of decreasing skin inflammation and repairing the defective skin barrier. Essential
treatments for AD in children should include topical moisturizers for skin hydration and
prevention of flares, topical anti-inflammatory medications (e.g. corticosteroids, calcineurin
inhibitors, PDE4 inhibitor), allergen/irritant avoidance, and treatment of skin infections. Treatment
regimens should be severity-based, and implemented in a stepwise approach tailored to the
individual patient. This stepwise approach includes initial use of emollients, gentle skin care, and
escalating to more potent anti-inflammatory treatments as the disease severity increases. Currently
available systemic medications should be reserved for the presence of recalcitrance to topical
therapies due to associated toxicities.
Author Manuscript

We believe that early treatment of AD is not only essential in treating the skin disease, but also in
preventing the development of additional atopic diseases, such as food allergy, asthma and allergic
rhinitis. The defective skin barrier of AD permits a route of entry for food and environmental
allergens, and upon exposure, keratinocytes secrete TSLP, which activates the TH2 pathway. This
TH2 differentiation sets off the atopic march and the subsequent diseases that are seen. This review
highlights treatment options and strategies in pediatric AD therapy with an emphasis on early
therapy. Supporting evidence on the efficacy and safety of each intervention will be discussed.

Keywords
atopic dermatitis; eczema; atopic march; allergy; corticosteroids; immunosuppression
Author Manuscript

Corresponding author: Kanwaljit Brar, MD: brark@njhealth.org.


COMPLIANCE WITH ETHICS GUIDELINES
Conflict of Interest:
Dr. Amy Huang, Dr. Christine Cho, Dr. Kanwaljit Brar, and Dr. Donald Y.M. Leung declares that they have no conflict of interest.
Donald Y.M. Leung is supported by the National Institutes of Health under grant number R01AR41256. The content is solely the
responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Human and Animal Rights and Informed Consent:
This article does not contain any studies with human or animal subjects performed by any of the authors.
Huang et al. Page 2

INTRODUCTION
Author Manuscript

Atopic dermatitis (AD), or atopic eczema, is the most common inflammatory skin condition
of childhood. It affects 15–30% of children and it is particularly common in industrialized
countries worldwide [1]. In the United States, there were 7.4 million visits of children
younger than 18 years to physicians for AD [2]. The disease is characterized by chronic and
relapsing pruritic skin lesions that generally develop in early childhood, usually between 3
and 6 months of age. Approximately 60% of patients develop eczematous lesions in the first
year of life and 90% by 5 years of age [3].

The distribution of AD lesions also differs with age, as neonates and infants (0–2 years)
generally have pruritic, erythematous, weeping patches on the cheeks, scalp, and extensor
surfaces of extremities. These lesions progress to xerosis, thickened plaques, papules, and
excoriations on the wrists and flexural surfaces of extremities in childhood (2–12 years).
Author Manuscript

There are currently 12 various diagnostic criteria for AD, the first of which is the Hanifin
and Rajka criteria, which was developed in 1980 [4]. The Hanifin and Rajka criteria is
composed of four major and 23 minor clinical criteria, and is often used in clinical trials for
the diagnosis of AD.

AD often heralds the onset of the atopic march, a natural progression of atopic disorders that
begins with AD in infancy and leads to allergic rhinitis, food allergies, and asthma in later
childhood [5••]. A recent systematic review showed a strong association between AD, food
sensitization, and food allergy, especially in AD of increased severity and chronicity. In
addition, the study also found evidence that AD precedes the development of food
sensitization and allergy [6]. AD and progressive atopy have a complex pathogenesis that
can involve environmental stressors, mutations in the filaggrin gene and the epidermal
Author Manuscript

differentiation complex that encodes epidermal structure and immune effectors, as well as
dysfunctional immune responses [5]. These dysregulated immune responses include a
selective decrease in skin-homing TH1 cells and an increase in TH2 cells, especially in
patients with severe AD [7•]. A study by Czarnowicki et al. found that patients with severe
AD have significantly lower CLA + (skin homing) IFN-γ producing T cells than control
subjects indicating a significant role for IFN-γ, a regulatory cytokine in the pathogenesis of
AD [7•].

The impaired epidermal barrier in AD contributes to transepidermal water loss, leading to


xerosis and promoting colonization and secondary infection with Staphylococcus aureus,
which can enhance type 2 inflammation resulting in downregulation of FLG expression [8].
Patients colonized with S. aureus have higher total serum IgE levels, as well as higher
peripheral eosinophilia [9]. Children with early-onset, severe, persistent AD, and elevated
Author Manuscript

levels of total and specific IgE antibodies, are at increased risk of developing asthma and
allergic rhinitis later in life [10, 11]. This may be due to the defective skin barrier in AD
permitting epicutaneous exposure of environmental antigens to a local skin milieu primed
toward type 2 immune responses [12, 13]. Cytokines, such as thymic stromal lymphopoietin
(TSLP) and IL-33, are released by keratinocytes when the skin barrier is disrupted,
activating dendritic cells to trigger an aberrant TH2-mediated immune response [5••].

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 3

There is also evidence that early sensitization to foods or aeroallergens in the first year of
Author Manuscript

life increase the risk of persistent AD and asthma [14–16]. In fact, food-induced AD flares
occur in one-third of infants and young children with moderate-to-severe AD, but are
uncommon in adults [17]. Infants with moderate-to-severe AD are also at high risk of food
allergy at 2 years of age, with studies demonstrating that percutaneous exposure to food
proteins is allergenic. In contrast, enteral exposure is tolerogenic [18–20]. As such, it is
believed that early optimal treatment of AD, would prevent epicutaneous sensitization,
which may halt or attenuate the atopic march including food allergies, though there is no
data to date to support this hypothesis.

It is important to note the timing of solid food introduction or withholding of allergenic


foods in both maternal and infant diets does not seem to have a protective effect against AD
[21]. However, clinical trials have shown that hydrolyzed protein formula, probiotic
supplementation, and early introduction of allergenic foods can be beneficial in preventing
Author Manuscript

and improving AD in high-risk infants and children [22–28]. The Learning Early About
Peanut Allergy (LEAP) Study showed that food allergy can be avoided in this high-risk
population with moderate to severe AD [29••] by early introduction of peanut. This is an
effective and feasible method to prevent peanut allergy in high-risk atopic infants, without
negatively affecting nutrition and growth.

TREATMENT
Non-Pharmacologic Interventions
Topical Moisturizers—Moisturizers are the cornerstone of all AD regimens. Xerosis is
one of the main clinical features of AD, and results from a dysfunctional epidermal barrier
that leads to increased transepidermal water loss. Topical moisturizers combat xerosis
Author Manuscript

through a combination of ingredients that maintain skin hydration, such as emollients (e.g.
glyceryl stearate, soy sterols) that lubricate the skin, occlusive agents (e.g. petrolatum,
dimethicone, mineral oil) that prevent water evaporation, and humectants (e.g. lactic acid,
urea, glycerol) that attract and hold water into the stratum corneum [30••].

There has been an abundance of evidence supporting emollient therapy in preventing and
treating pediatric AD. The predictions of one mathematical model of AD confirm that
emollient therapies reduce the ability of environmental stressors to cause TH2 sensitization
[31]. This was defined by a two-fold increase in minimum stress load needed to trigger
systemic TH2 sensitization and subsequent AD flares. Numerous clinical trials have
demonstrated efficacy of emollient therapy in preventing and decreasing the clinical
manifestations of AD, including pruritus, erythema, fissuring, and lichenification, in
Author Manuscript

neonates, infants, and children [32–35•] and in adults [36–38]. In neonates, early moisturizer
intervention resulted in a decrease in the cumulative incidence of AD, with a relative risk
reduction of 50% [32•]. Moisturizers have a steroid-sparing effect on treatment of AD. This
was shown in three randomized controlled trials [34, 39, 40], and should be a component in
the regimen for moderate-to-severe disease. Moisturizers should also be used as
maintenance therapy in any AD regimen. There are currently no studies that define an
optimal amount or frequency of moisturizer application, although current guidelines from
the American Academy of Dermatology suggest liberal daily use of moisturizers [30••].

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 4

Special considerations should be made in the treatment of neonates and infants, compared to
Author Manuscript

that of adults. The skin of infants under the age of 2 years is characterized by a thinner
epidermis and stratum corneum, higher water content, increased transepidermal water loss,
high skin pH, and high desquamation and proliferation rates [41]. Infants have a high ratio of
body surface area to body weight, resulting in increased absorption of topical medications.
This translates to an increased need for moisturizers to combat skin water loss and lower
doses of topical medications.

Newer formulations of topical moisturizers containing ingredients that mimic components of


the epidermal barrier in distinct ratios have also been effective in treating pediatric AD.
These prescription emollient devices (PEDs) contain ingredients such as ceramide,
palmitoylethanolamide, glycyrrhetinic acid, and other hydrolipids and filaggrin breakdown
products that are reduced in levels in the skin of patients with AD. PEDs have been found to
be just as effective as over-the-counter moisturizers in reducing the clinical signs and
Author Manuscript

symptoms of AD, in limited studies in children [42–47], and can be useful adjuncts to
topical corticosteroid therapy. However, they can be cost-prohibitive, and are mostly
available by prescription.

Bathing Practices and Additives—Daily bathing with warm water is beneficial in AD


therapy by hydrating the skin and removing serous crusts, allergens, and irritants [30••].
Cleansers should be hypoallergenic, fragrance-free, and neutral to low pH. This should be
followed by quick towel-drying by patting, and application of moisturizers to prevent
transepidermal water loss [48]. There is a paucity of evidence to determine the best bathing
practices, as most recommendations stem from personal experience. The “soak and smear”
technique of soaking in plain water for 20 minutes, followed by immediate and direct
application of topical corticosteroids, can be beneficial in severely inflamed lesions [49].
Author Manuscript

There are no studies to support the use of bath additives, such as oils, emollients, and salts,
in treating AD.

Bleach Baths—The addition of dilute sodium hypochlorite in bath water, or bleach baths,
has demonstrated efficacy in clinically improving moderate-to-severe AD in children [50–
52] and has been recommended as a low-cost and effective adjuvant therapy in these patients
by the American Academy of Dermatology (AAD) and the American Academy of Allergy,
Asthma and Immunology (AAAAI). Bleach baths are thought to reduce skin inflammation
and thereby decrease colonization of Staphylococcus aureus bacteria on the skin. This can
be beneficial, as staphylococcal exotoxins are known to exacerbate AD [53]. However, one
recent study noted that bleach baths did not reduce S. aureus colonization/infection or
improve AD [54]. Common side-effects of bleach baths include exacerbation of xerosis and
Author Manuscript

skin and nasal irritation.

Wet Wrap Therapy—Wet wrap therapy (WWT) is used to reduce disease severity in
children with significant AD flares and/or refractory disease. After a soaking bath, a wetted
layer of bandages, gauze, or a cotton suit is applied over a layer of topical corticosteroid or
emollient, followed by a dry outer layer [30••]. WWT serves to increase penetration of the
medication by occlusion, prevents patient scratching, and decreases epidermal water loss. In
our experience, a cotton suit, which covers hands and feet, works best as an outer layer, as

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 5

this physically prevents scratching. In children with refractory AD, WWT improved AD
Author Manuscript

severity in a number of studies, especially when used with corticosteroids versus emollients
[55, 56]. A recent systematic review revealed low evidence that WWT is more effective than
conventional treatment with topical corticosteroids in AD [57]. However, the review
included all ages and severities of AD.

Pharmacological Interventions
Topical Corticosteroids—Topical corticosteroids (TCS) are essential anti-inflammatory
agents in the management of AD. They reduce the production of pro-inflammatory
cytokines, interfere with antigen processing, and reduce the activity of immune effector
cells, thus lowering skin inflammation [30••]. TCS are typically administered when the skin
appears inflamed as evidenced by erythema, oozing, crusting, and/or lichenification. TCS
can also be used as maintenance therapy for prevention of relapses. TCS have been used to
Author Manuscript

treat AD for over 60 years, and there are more than 110 different randomized-controlled
trials performed to date [30••]. Newer types of TCS are constantly being developed, each
with different potencies, vehicles, and excipients. Patient preference (vehicle, cost, and
availability), lesion site, and disease severity often drive selection in prescribing and
selecting potency of TCS. TCS are well-studied in pediatric patients, and have proven to be
safe and effective in reducing the clinical signs and symptoms of AD [58]. Fluticasone
propionate 0.05% cream, desonide 0.05% gel and foam, and hydrocortisone butyrate 0.1%
lotion are the only topical corticosteroids that are U.S. Food and Drug Administration (FDA)
approved for use in infants as young as three months of age.

TCS range in potencies and are grouped accordingly into seven classes, from very low/
lowest potency (VII) to very high potency (I) (Table 1) [59]. There are currently no
guidelines in optimal dosing and quantity of TCS application [30••]. Low potency (Class
Author Manuscript

VII) TCS are generally applied to sensitive and thin areas, such as the face, skin folds, and
genitalia. Special considerations are needed when using topical therapies for children with
AD. Children have a proportionately greater body surface area to weight ratio, resulting in a
higher degree of absorption of topical agents. Higher potency TCS can be used in short-term
courses to rapidly control significant flares, but should be followed by a stepwise decrease in
potency and then tapered to the lowest effective potency for long-term management. This
minimizes the adverse effects of skin atrophy, telangiectasia, acne, and striae, which are
major concerns of parents of children with AD.

There have been multiple studies addressing the short and long-term safety of TCS in infants
and children. A recent systematic review of AD patients less than 12 years of age found that
the evidence supporting long-term TCS use is limited only to low- to mid-potency agents,
Author Manuscript

and there was a lack of data supporting the use of long-term monotherapy with mid- to high-
potency TCS in pediatric AD [60]. In fact, continuous, long-term application of high- and
very high-potency agents can lead to significant systemic absorption and increased risk of
systemic adverse effects, such as hypothalamic-pituitary-axis (HPA) suppression, especially
in children concurrently receiving other forms of steroids for asthma [61]. Some
observational studies reported growth delay and abnormal bone turnover in children treated
long-term with TCS for AD, but others have not [62–64]. Sustained, long-term use of a mid-

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 6

potency TCS with one to twice weekly application did not demonstrate adverse effects in a
Author Manuscript

clinical trial [65], and one systematic review concluded a good overall safety profile of TCS
[66]. A recent cross-sectional observational study of children with AD on long-term TCS
(weak, moderate, and potent) showed no rates of skin atrophy, both in the TCS treatment
group and in the control group [67]. Nonetheless, allaying parental fears of TCS use in
children is essential in maintaining adherence and appropriate use, which decreases the risks
of adverse effects and relapse [68, 69].

Topical Calcineurin Inhibitors—Topical calcineurin inhibitors (TCIs) are anti-


inflammatory agents and bacteria-derived macrolides that bind intracellular protein
macrophilin-12 (FK-binding protein) and prevents translocation of the nuclear factor of
activated T cells (NFAT), which reduces the expression of T cell activation cytokines, in
particular IL-2 [70]. TCIs do not cause skin atrophy, telangiectasia, and striae, as do potent
TCS, and are effective both as steroid-sparing agents and agents in the treatment of AD
Author Manuscript

lesions on the eyelids, face, and skin folds. They are the only FDA-approved treatment for
chronic AD, and are typically prescribed to patients where long-term use of TCS increases
the risk of steroid-related side-effects. Two TCIs are available: Tacrolimus (Protopic) is
available as a 0.03% (approved for children 2 years of age and older) or 0.1% ointment
(approved for children over 16 years of age), and Pimecrolimus (Elidel) is available as a 1%
cream (approved for use in children 2 years of age and older). However, both agents have
been shown in studies to effectively treat AD in infants [71]. Tacrolimus is indicated for
moderate-to-severe AD, although tacrolimus 0.03% has shown efficacy and safety in
pediatric patients with mild-to-moderate AD [72]. Pimecrolimus is indicated for mild-to-
moderate AD [30••]. The FDA’s Pediatrics Advisory Committee issued a black box warning
of cancer risk for topical tacrolimus and pimecrolimus. However, this is mostly a theoretical
risk of malignancy, as most cancers have been seen only with oral tacrolimus use in
Author Manuscript

transplant patients [73]. The FDA recommended the medications be avoided for children
younger than age 2. However, since this warning, a number of studies have failed to
demonstrate this causation, and incidence of malignancy in the treated population is similar
to that of the general population [73]. In infants and children with active lesions, both agents
have been shown to be more effective than vehicle in short-term (3 to 12 weeks) and long-
term (up to 12 months) studies [74–79]. Two systematic reviews have shown that TCIs and
TCS have similar efficacy [80, 81••] , though TCIs had a higher incidence of skin irritation
and pruritus, and may cause stinging and burning. TCIs are also often combined with TCS to
control flares, prevent relapse, and spare topical steroid use. TCIs used concomitantly with
TCS have been shown to be more effective than TCI and vehicle or use of either agent alone
[82].
Author Manuscript

PDE4 Inhibitor—Crisaborole ointment, a non-steroidal phosphodiesterase 4 (PDE4)


inhibitor, was recently approved by the FDA to treat mild-to-moderate AD in patients 2
years of age and older. PDE4 is a regulator of the cyclic adenosine monophosphate (cAMP)
pathway at an intracellular level to reduce cytokine production of immune effector cells
[83•] . Crisaborole has been shown to be effective and with a favorable safety profile in two
phase III AD studies [83•] . Common side-effects included stinging and burning upon
application.

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 7

Systemic Immunosuppressants
Author Manuscript

Systemic immunosuppressants, such as cyclosporine, azathioprine, mycophenolate, and


methotrexate, are administered off-label in the management of chronic and/or moderate-to-
severe AD refractory to topical conventional treatments. These therapies are not FDA
approved for use in AD; and have multiple associated toxicities. However, they may be
effective in children with severe AD, in conjunction with aggressive use of moisturizers and
topical agents.

Cyclosporine—Cyclosporine A (CSA) is a calcineurin inhibitor that inhibits T cell


activation and IL-2 production. CSA can be administered as both short-term and long-term
therapy in severe pediatric AD (ages 2–16 years) [84]. However, CSA has been linked to
lower bone mineral density in children [85]. Dosing guidelines in the pediatric population
recommend 3–6 mg/kg/day, and 150–300mg/day in adults. Patients should be monitored for
Author Manuscript

side-effects, which include infection, nephrotoxicity, hypertension, tremor, hypertrichosis,


headache, gingival hyperplasia, and increased risk of skin cancer and lymphoma [86••].

Azathioprine—Azathioprine (AZA) is a purine analog that inhibits DNA production,


affecting cells with high proliferation rates, such as immune effector cells during active
inflammation. Although it is FDA-approved for rheumatoid arthritis and renal transplant
rejection prophylaxis, AZA has also been used off-label for severe recalcitrant AD. In a
retrospective study of AZA use in severe pediatric AD, 28/48 children had an excellent
response to treatment and 13/48 had a good response [87]. AZA is metabolized by
thiopurine methyltransferase (TPMT), and baseline TPMT activity levels should be
measured in children before administration of the drug. The same study showed that children
with higher TPMT levels responded less well to treatment and had a greater risk of
Author Manuscript

hepatotoxicity, while children with lower TPMT levels responded more favorably, but had an
increased risk of myelosuppression [87]. More common adverse effects include nausea,
vomiting, and gastrointestinal symptoms, which may cause the child emotional distress and
result in medication non-compliance. Pediatric dosing varies, but most studies recommend
2.5 mg/kg/day with a maximum of 4 mg/kg/day [86••] . The dose is gradually titrated up to
minimize nausea and vomiting.

Methotrexate—Methotrexate (MTX) is an anti-folate metabolite that blocks DNA


synthesis. It is used in a variety of inflammatory dermatological diseases, such as psoriasis
and cutaneous lupus erythematosus. There was only one prospective study on MTX use in
children with severe AD, and it compared MTX to CSA [88]. The same considerations in
safety and dosing apply for pediatric patients, as in adult patients. Dosing varies greatly with
the patient, and is based on the psoriasis dose regimen (0.2–0.7 mg/kg/week) [86••] .
Author Manuscript

Common side-effects include nausea, vomiting, and stomatitis. Patients should be monitored
for severe and potentially irreversible side-effects, such as bone marrow suppression and
pulmonary fibrosis.

Mycophenolate Mofetil—Mycophenolate mofetil (MMF) is an immunosuppressant drug


that inhibits B and T cell growth and proliferation. MMF has been used as monotherapy in
children with severe refractory AD aged 2 years and older, without significant adverse

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 8

effects [89]. Dosing in children (600–1200 mg/m2) is adjusted for increased hepatic
Author Manuscript

metabolism in the pediatric population. Although there are currently no long-term efficacy
and safety studies examining the use of MMF in pediatric AD; in one study, use in children
for up to 24 consecutive months did not demonstrate significant adverse effects. Common
side-effects include nausea, vomiting, and abdominal cramping [86••] .

Systemic Corticosteroids—Systemic corticosteroids (prednisone, prednisolone, and


methylprednisolone) should generally be avoided in children with AD, as the risks of
treatment outweigh the benefits [86••]. Risks include severe rebound flares and increased
disease severity when systemic corticosteroids are weaned or stopped. Additionally,
hypertension, glucose intolerance, weight gain, adrenal suppression, increased infections,
and decreased bone density are also common with chronic use. Pediatric patients are at risk
of decreased linear growth, which can be long-term [90]. There is little evidence to support
use of systemic steroids in AD; in one double-blind, placebo-controlled trial, only one
Author Manuscript

patient out of 27 taking prednisolone achieved durable remission after two weeks of oral
steroid therapy [91]. The study was also prematurely discontinued due to significant rebound
flaring in the steroid group. Dosing is generally 0.5–1.0 mg/kg, and steroids must be
carefully tapered to decrease the risk of adrenal suppression and severe AD flare [86••].
Pediatric patients on systemic steroids require blood pressure monitoring, growth-velocity
measurements, ophthalmologic examination, and hypothalamic-pituitary-adrenal axis
suppression testing [86••].

Interventional Procedures
Phototherapy—The usefulness of ultraviolet (UV) light plus oral psoralen in the treatment
of AD has been documented since the late 1970’s [92]. Since then, a variety of light therapy
Author Manuscript

have been used as short-term treatment of AD, including natural sunlight, narrow-band
ultraviolet light B (NB-UVB), broad-band ultraviolet light B (BB-UVB), ultraviolet light A
(UVA), topical and systemic psoralen plus UVA (PUVA), and ultraviolet light A and B
(UVAB) [86••] . NB-UVB is the most commonly recommended light therapy, due to its
efficacy, availability, and low risk of side-effects. Phototherapy is generally recommended in
the treatment of AD refractory to conventional topical medications, and can be used as
monotherapy or in combination with TCS. Use with TCIs is not recommended. In children,
phototherapy has demonstrated efficacy and safety in multiple studies [93–95]. Various
lasers (e.g. excimer, diode, and pulse dye) and extracorporeal photochemotherapy (ECP) are
additional modes of treatment for AD; the latter is used in generalized and erythrodermic
AD patients [86••] . There is a theoretical increased risk of skin cancer with long-term
phototherapy, due to exposure to UV light. Although one large study found no significant
association between NB-UVB therapy and skin cancer, treatment with PUVA was found to
Author Manuscript

have a slight increase in incidence of basal cell carcinoma [96].

Emerging Therapies
Several promising agents are currently under investigation in the treatment of AD. These
agents, including biologics, are not currently FDA-approved for use in children, though there
are ongoing studies examining their use in pediatric populations.

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 9

Topical Lipoxin—Lipoxins are endogenous, anti-inflammatory molecules derived from


Author Manuscript

the metabolic breakdown of arachidonic acid. These molecules are activated during
inflammation and inhibit the production of pro-inflammatory cytokines, such as IL-12,
IL-13, and various leukotrienes [97]. In a study of infantile AD, topical 15(R/S)-methyl-
lipoxin A4 (LXA4) significantly reduced disease severity, induced remission, and improved
quality of life. This was a randomized, double-blind, placebo-controlled, parallel-group trial
of patients 1–12 months of age with varying disease severity that compared the lipoxin
cream with mometasone furoate cream, a mid-potency topical steroid [98]. Extent and rate
of recovery were similar to that of patients in the 0.1% mometasone furoate cohort.

Omalizumab—Another biologic that has been used off-label in pediatric AD is


omalizumab, a humanized monoclonal antibody that binds to free IgE and surface-bound
IgE on mast cells and basophils. A case series of seven patients (aged 6–19 years) with
severe AD demonstrated clinical improvement of symptoms after 3 to 6 months of treatment
Author Manuscript

[99]. A recent systematic review, however, stated no concrete evidence of the effectiveness
of omalizumab as a treatment for AD [100].

Recombinant Interferon Gamma—Several studies have examined the use of


subcutaneous recombinant interferon gamma (rIFN-γ) in AD. IFN-γ is a cytokine that is
involved in innate and adaptive immunity. In the first double-blind, placebo-controlled trial
of rIFN-γ in moderate-to-severe AD in children and adults, rIFN-γ was found to have an
age-related, improved clinical response in the pediatric cohort versus adults. 67% of children
ages 3 to 20 reported greater than 50% improvement, compared to 56% of adults ages 21–40
and 44% of adults ages 41–65 [101]. Side-effects included flu-like symptoms, transient
transaminase elevation, and granulocyte suppression [102].
Author Manuscript

Dupilumab—Dupilumab, a fully human monoclonal antibody against interleukin-4


receptor alpha, is a biologic that was recently FDA approved for use in adults with
moderate-to-severe AD. Dupilumab blocks signaling from two key cytokines in the TH2
inflammatory pathway, IL-4 and IL-13. In Phase III trials, significantly more patients treated
with 300 mg bi-weekly showed 75% improvement in Eczema Area and Severity Index
(EASI) from baseline compared with the placebo group [103]. In addition, treatment
resulted in improvement of other endpoints, including pruritus, symptoms of anxiety, and
depression, and quality of life. A Phase 2a, open-label trial of 78 children and adolescents,
ages 6–18, with moderate-to-severe AD (NCT02407756) demonstrated mean improvement
of pruritus and EASI scores, especially in the younger cohort, at increasing subcutaneous
doses (from 2–4mg/kg) [104•]. Dupilumab is a promising therapy in AD and is expected to
alter future management of the disease towards a more personalized approach. Though its
Author Manuscript

use is currently only approved in adults, it may benefit the younger pediatric population once
studied and found to be safe in children.

CONCLUSION
Early treatment of AD in children can delay or prevent the atopic march. Although the
pathogenesis of AD is multivariate and complex, therapeutic interventions target two major
areas of dysfunction: 1) the defective skin barrier leading to early sensitization to allergens

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 10

and 2) the dysfunctional skin immune response to allergens and irritants. Topical
Author Manuscript

moisturizers and topical anti-inflammatory medications are the cornerstones of AD


treatment regimens. Although there are a wide variety of both moisturizers and topical
corticosteroids, any combination of both with gentle skin care can effectively control most
mild-to-moderate AD lesions. Moderate-to-severe and recalcitrant AD, however, may
require the addition of phototherapy or systemic medications. Special considerations are
made for pediatric patients, as severe adverse effects of systemic medications and high-dose
topical corticosteroids must be weighed against the benefits of treatment. Emerging
therapies, such as crisaborole and dupilumab, are welcome additions to the current arsenal of
treatments for AD.

References
Papers of particular interest, published recently, have been highlighted as:
Author Manuscript

• Of importance

•• Of major importance

1. Baron SE, et al. Guidance on the diagnosis and clinical management of atopic eczema. Clin Exp
Dermatol. 2012; 37(Suppl 1):7–12. [PubMed: 22486763]
2. Horii KA, et al. Atopic dermatitis in children in the United States, 1997–2004: visit trends, patient
and provider characteristics, and prescribing patterns. Pediatrics. 2007; 120(3):e527–34. [PubMed:
17766497]
3. Kay J, et al. The prevalence of childhood atopic eczema in a general population. J Am Acad
Dermatol. 1994; 30(1):35–9. [PubMed: 8277028]
4. Lee SC. Committee of Korean Atopic Dermatitis Association for R. Various diagnostic criteria for
atopic dermatitis (AD): A proposal of Reliable Estimation of Atopic Dermatitis in Childhood
(REACH) criteria, a novel questionnaire-based diagnostic tool for AD. J Dermatol. 2016; 43(4):
Author Manuscript

376–84. [PubMed: 26813749]


5••. Leung DY, Guttman-Yassky E. Deciphering the complexities of atopic dermatitis: shifting
paradigms in treatment approaches. J Allergy Clin Immunol. 2014; 134(4):769–79. This paper is
an educational discussion on the complex pathogenesis of atopic dermatitis, covering the
immunological, microbiological, genetic, and environmental etiologies of the disease. [PubMed:
25282559]
6••. Tsakok T, et al. Does atopic dermatitis cause food allergy? A systematic review. J Allergy Clin
Immunol. 2016; 137(4):1071–8. This systematic review provides evidence of the causal
relationship of atopic dermatitis and food allergies/sensitization in the atopic march. [PubMed:
26897122]
7•. Czarnowicki T, et al. Severe atopic dermatitis is characterized by selective expansion of circulating
TH2/TC2 and TH22/TC22, but not TH17/TC17, cells within the skin-homing T-cell population. J
Allergy Clin Immunol. 2015; 136(1):104–15. e7. This scientific study demonstrates the
immunologic etiology of atopic dermatitis, in relation to aberrant T cell response. [PubMed:
25748064]
Author Manuscript

8. van Drongelen V, et al. Reduced filaggrin expression is accompanied by increased Staphylococcus


aureus colonization of epidermal skin models. Clin Exp Allergy. 2014; 44(12):1515–24. [PubMed:
25352374]
9. Warner JA, et al. Biomarkers of Th2 polarity are predictive of staphylococcal colonization in
subjects with atopic dermatitis. Br J Dermatol. 2009; 160(1):183–5. [PubMed: 19016695]
10. Saunes M, et al. Early eczema and the risk of childhood asthma: a prospective, population-based
study. BMC Pediatr. 2012; 12:168. [PubMed: 23095804]
11. von Kobyletzki LB, et al. Eczema in early childhood is strongly associated with the development of
asthma and rhinitis in a prospective cohort. BMC Dermatol. 2012; 12:11. [PubMed: 22839963]

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 11

12•. Bantz SK, et al. The Atopic March: Progression from Atopic Dermatitis to Allergic Rhinitis and
Asthma. J Clin Cell Immunol. 2014; 5(2) This comprehensive review provides further insight
Author Manuscript

into the pathogenesis of the atopic march.


13. Akei HS, et al. Epicutaneous aeroallergen exposure induces systemic TH2 immunity that
predisposes to allergic nasal responses. J Allergy Clin Immunol. 2006; 118(1):62–9. [PubMed:
16815139]
14. Filipiak-Pittroff B, et al. Predictive value of food sensitization and filaggrin mutations in children
with eczema. J Allergy Clin Immunol. 2011; 128(6):1235–41. e5. [PubMed: 22030464]
15. Rhodes HL, et al. Early life risk factors for adult asthma: a birth cohort study of subjects at risk. J
Allergy Clin Immunol. 2001; 108(5):720–5. [PubMed: 11692095]
16. Gustafsson D, et al. Development of allergies and asthma in infants and young children with atopic
dermatitis--a prospective follow-up to 7 years of age. Allergy. 2000; 55(3):240–5. [PubMed:
10753014]
17. Forbes LR, et al. Food allergies and atopic dermatitis: differentiating myth from reality. Pediatr
Ann. 2009; 38(2):84–90. [PubMed: 19263783]
18. Kelleher MM, et al. Skin barrier impairment at birth predicts food allergy at 2 years of age. J
Author Manuscript

Allergy Clin Immunol. 2016; 137(4):1111–6. e1–8. [PubMed: 26924469]


19. Strid J, et al. Epicutaneous exposure to peanut protein prevents oral tolerance and enhances allergic
sensitization. Clin Exp Allergy. 2005; 35(6):757–66. [PubMed: 15969667]
20. Strid J, et al. A novel model of sensitization and oral tolerance to peanut protein. Immunology.
2004; 113(3):293–303. [PubMed: 15500615]
21. Greer FR, et al. Effects of early nutritional interventions on the development of atopic disease in
infants and children: the role of maternal dietary restriction, breastfeeding, timing of introduction
of complementary foods, and hydrolyzed formulas. Pediatrics. 2008; 121(1):183–91. [PubMed:
18166574]
22. Kramer MS, Kakuma R. Maternal dietary antigen avoidance during pregnancy or lactation, or both,
for preventing or treating atopic disease in the child. Evid Based Child Health. 2014; 9(2):447–83.
[PubMed: 25404609]
23. Yang YW, et al. Exclusive breastfeeding and incident atopic dermatitis in childhood: a systematic
review and meta-analysis of prospective cohort studies. Br J Dermatol. 2009; 161(2):373–83.
Author Manuscript

[PubMed: 19239469]
24. Osborn DA, Sinn J. Formulas containing hydrolysed protein for prevention of allergy and food
intolerance in infants. Cochrane Database Syst Rev. 2006; (4):CD003664. [PubMed: 17054180]
25. Alexander DD, Cabana MD. Partially hydrolyzed 100% whey protein infant formula and reduced
risk of atopic dermatitis: a meta-analysis. J Pediatr Gastroenterol Nutr. 2010; 50(4):422–30.
[PubMed: 20216095]
26. Pelucchi C, et al. Probiotics supplementation during pregnancy or infancy for the prevention of
atopic dermatitis: a meta-analysis. Epidemiology. 2012; 23(3):402–14. [PubMed: 22441545]
27. Kuitunen M, et al. Probiotics prevent IgE-associated allergy until age 5 years in cesarean-delivered
children but not in the total cohort. J Allergy Clin Immunol. 2009; 123(2):335–41. [PubMed:
19135235]
28. Rosenfeldt V, et al. Effect of probiotic Lactobacillus strains in children with atopic dermatitis. J
Allergy Clin Immunol. 2003; 111(2):389–95. [PubMed: 12589361]
29••. Du Toit G, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. N
Engl J Med. 2015; 372(9):803–13. The LEAP Study is an important clinical trial that showed that
Author Manuscript

early, regular introduction of peanut in infants may decrease the frequency of peanut allergy in at-
risk infants. [PubMed: 25705822]
30••. Eichenfield LF, et al. Guidelines of care for the management of atopic dermatitis: section 2.
Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol.
2014; 71(1):116–32. This is a comprehensive set of guidelines from the American Academy of
Dermatology on the topical treatments of atopic dermatitis. [PubMed: 24813302]
31. Dominguez-Huttinger E, et al. Mathematical modeling of atopic dermatitis reveals “double-switch”
mechanisms underlying 4 common disease phenotypes. J Allergy Clin Immunol. 2016

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 12

32•. Simpson EL, et al. Emollient enhancement of the skin barrier from birth offers effective atopic
dermatitis prevention. J Allergy Clin Immunol. 2014; 134(4):818–23. This study showed that
Author Manuscript

early and consistent use of emollients in neonates at increased risk of atopic dermatitis effectively
prevents the appearance of atopic dermatitis. Emollient therapy is essential to treating and
preventing atopic dermatitis in at-risk children. [PubMed: 25282563]
33•. Horimukai K, et al. Application of moisturizer to neonates prevents development of atopic
dermatitis. J Allergy Clin Immunol. 2014; 134(4):824–30. e6. This is another study
demonstrating the efficacy of emollient therapy in preventing atopic dermatitis in children.
[PubMed: 25282564]
34. Grimalt R, et al. The steroid-sparing effect of an emollient therapy in infants with atopic dermatitis:
a randomized controlled study. Dermatology. 2007; 214(1):61–7. [PubMed: 17191050]
35. Korting HC, et al. Efficacy and tolerability of pale sulfonated shale oil cream 4% in the treatment
of mild to moderate atopic eczema in children: a multicentre, randomized vehicle-controlled trial.
J Eur Acad Dermatol Venereol. 2010; 24(10):1176–82. [PubMed: 20236198]
36. Czarnowicki T, et al. Petrolatum: Barrier repair and antimicrobial responses underlying this “inert”
moisturizer. J Allergy Clin Immunol. 2016; 137(4):1091–102. e1–7. [PubMed: 26431582]
Author Manuscript

37. Akerstrom U, et al. Comparison of Moisturizing Creams for the Prevention of Atopic Dermatitis
Relapse: A Randomized Double-blind Controlled Multicentre Clinical Trial. Acta Derm Venereol.
2015; 95(5):587–92. [PubMed: 25594845]
38. Breternitz M, et al. Placebo-controlled, double-blind, randomized, prospective study of a glycerol-
based emollient on eczematous skin in atopic dermatitis: biophysical and clinical evaluation. Skin
Pharmacol Physiol. 2008; 21(1):39–45. [PubMed: 18025807]
39. Tan WP, et al. A randomized double-blind controlled trial to compare a triclosan-containing
emollient with vehicle for the treatment of atopic dermatitis. Clin Exp Dermatol. 2010;
35(4):e109–12. [PubMed: 19843084]
40. Msika P, et al. New emollient with topical corticosteroid-sparing effect in treatment of childhood
atopic dermatitis: SCORAD and quality of life improvement. Pediatr Dermatol. 2008; 25(6):606–
12. [PubMed: 19067864]
41. Folster-Holst R. Management of atopic dermatitis: are there differences between children and
adults? J Eur Acad Dermatol Venereol. 2014; 28(Suppl 3):5–8. [PubMed: 24702444]
42. Sugarman JL, Parish LC. Efficacy of a lipid-based barrier repair formulation in moderate-to-severe
Author Manuscript

pediatric atopic dermatitis. J Drugs Dermatol. 2009; 8(12):1106–11. [PubMed: 20027938]


43. Chamlin SL, et al. Ceramide-dominant barrier repair lipids alleviate childhood atopic dermatitis:
changes in barrier function provide a sensitive indicator of disease activity. J Am Acad Dermatol.
2002; 47(2):198–208. [PubMed: 12140465]
44. Kircik LH, Del Rosso JQ. Nonsteroidal treatment of atopic dermatitis in pediatric patients with a
ceramide-dominant topical emulsion formulated with an optimized ratio of physiological lipids. J
Clin Aesthet Dermatol. 2011; 4(12):25–31.
45. Marseglia A, et al. Local rhamnosoft, ceramides and L-isoleucine in atopic eczema: a randomized,
placebo controlled trial. Pediatr Allergy Immunol. 2014; 25(3):271–5. [PubMed: 24750568]
46. Hon KL, et al. Acceptability and efficacy of an emollient containing ceramide-precursor lipids and
moisturizing factors for atopic dermatitis in pediatric patients. Drugs R D. 2013; 13(1):37–42.
[PubMed: 23456759]
47. Miller DW, et al. An over-the-counter moisturizer is as clinically effective as, and more cost-
effective than, prescription barrier creams in the treatment of children with mild-to-moderate
Author Manuscript

atopic dermatitis: a randomized, controlled trial. J Drugs Dermatol. 2011; 10(5):531–7. [PubMed:
21533301]
48. Chiang C, Eichenfield LF. Quantitative assessment of combination bathing and moisturizing
regimens on skin hydration in atopic dermatitis. Pediatr Dermatol. 2009; 26(3):273–8. [PubMed:
19706087]
49. Gutman AB, et al. Soak and smear: a standard technique revisited. Arch Dermatol. 2005; 141(12):
1556–9. [PubMed: 16365257]

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 13

50. Wong SM, et al. Efficacy and safety of sodium hypochlorite (bleach) baths in patients with
moderate to severe atopic dermatitis in Malaysia. J Dermatol. 2013; 40(11):874–80. [PubMed:
Author Manuscript

24111816]
51. Huang JT, et al. Treatment of Staphylococcus aureus colonization in atopic dermatitis decreases
disease severity. Pediatrics. 2009; 123(5):e808–14. [PubMed: 19403473]
52. Huang JT, et al. Dilute bleach baths for Staphylococcus aureus colonization in atopic dermatitis to
decrease disease severity. Arch Dermatol. 2011; 147(2):246–7. [PubMed: 21339459]
53. Ong PY, Leung DY. The infectious aspects of atopic dermatitis. Immunol Allergy Clin North Am.
2010; 30(3):309–21. [PubMed: 20670815]
54. Hon KL, et al. Efficacy of sodium hypochlorite (bleach) baths to reduce Staphylococcus aureus
colonization in childhood onset moderate-to-severe eczema: A randomized, placebo-controlled
cross-over trial. J Dermatolog Treat. 2016; 27(2):156–62. [PubMed: 26270469]
55. Nicol NH, et al. Wet wrap therapy in children with moderate to severe atopic dermatitis in a
multidisciplinary treatment program. J Allergy Clin Immunol Pract. 2014; 2(4):400–6. [PubMed:
25017527]
56. Janmohamed SR, et al. The proactive wet-wrap method with diluted corticosteroids versus
Author Manuscript

emollients in children with atopic dermatitis: a prospective, randomized, double-blind, placebo-


controlled trial. J Am Acad Dermatol. 2014; 70(6):1076–82. [PubMed: 24698702]
57. Gonzalez-Lopez G, et al. Efficacy and safety of wet wrap therapy for patients with atopic
dermatitis: a systematic review and meta-analysis. Br J Dermatol. 2016
58. Torrelo A. Methylprednisolone aceponate for atopic dermatitis. Int J Dermatol. 2017
59. Eichenfield LF, et al. Translating Atopic Dermatitis Management Guidelines Into Practice for
Primary Care Providers. Pediatrics. 2015; 136(3):554–65. [PubMed: 26240216]
60. Siegfried EC, et al. Systematic review of published trials: long-term safety of topical
corticosteroids and topical calcineurin inhibitors in pediatric patients with atopic dermatitis. BMC
Pediatr. 2016; 16:75. [PubMed: 27267134]
61. Ellison JA, et al. Hypothalamic-pituitary-adrenal function and glucocorticoid sensitivity in atopic
dermatitis. Pediatrics. 2000; 105(4 Pt 1):794–9. [PubMed: 10742322]
62. McGowan R, et al. Short-term growth and bone turnover in children undergoing occlusive steroid
(‘Wet-Wrap’) dressings for treatment of atopic eczema. J Dermatolog Treat. 2003; 14(3):149–52.
Author Manuscript

[PubMed: 14522624]
63. Kristmundsdottir F, David TJ. Growth impairment in children with atopic eczema. J R Soc Med.
1987; 80(1):9–12.
64. Aalto-Korte K, Turpeinen M. Bone mineral density in patients with atopic dermatitis. Br J
Dermatol. 1997; 136(2):172–5. [PubMed: 9068726]
65. Schmitt J, et al. Efficacy and tolerability of proactive treatment with topical corticosteroids and
calcineurin inhibitors for atopic eczema: systematic review and meta-analysis of randomized
controlled trials. Br J Dermatol. 2011; 164(2):415–28. [PubMed: 20819086]
66. Callen J, et al. A systematic review of the safety of topical therapies for atopic dermatitis. Br J
Dermatol. 2007; 156(2):203–21. [PubMed: 17223859]
67. Hong E, et al. Evaluation of the atrophogenic potential of topical corticosteroids in pediatric
dermatology patients. Pediatr Dermatol. 2011; 28(4):393–6. [PubMed: 21507057]
68. Charman CR, et al. Topical corticosteroid phobia in patients with atopic eczema. Br J Dermatol.
2000; 142(5):931–6. [PubMed: 10809850]
Author Manuscript

69. Cork MJ, et al. Comparison of parent knowledge, therapy utilization and severity of atopic eczema
before and after explanation and demonstration of topical therapies by a specialist dermatology
nurse. Br J Dermatol. 2003; 149(3):582–9. [PubMed: 14510993]
70. Gutfreund K, et al. Topical calcineurin inhibitors in dermatology. Part I: Properties, method and
effectiveness of drug use. Postepy Dermatol Alergol. 2013; 30(3):165–9. [PubMed: 24278069]
71. Papp KA, et al. Long-term control of atopic dermatitis with pimecrolimus cream 1% in infants and
young children: a two-year study. J Am Acad Dermatol. 2005; 52(2):240–6. [PubMed: 15692468]

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 14

72. Chapman MS, et al. Tacrolimus ointment 0.03% shows efficacy and safety in pediatric and adult
patients with mild to moderate atopic dermatitis. J Am Acad Dermatol. 2005; 53(2 Suppl 2):S177–
Author Manuscript

85. [PubMed: 16021173]


73. Siegfried EC, et al. Topical calcineurin inhibitors and lymphoma risk: evidence update with
implications for daily practice. Am J Clin Dermatol. 2013; 14(3):163–78. [PubMed: 23703374]
74. Boguniewicz M, et al. A randomized, vehicle-controlled trial of tacrolimus ointment for treatment
of atopic dermatitis in children. Pediatric Tacrolimus Study Group. J Allergy Clin Immunol. 1998;
102(4 Pt 1):637–44. [PubMed: 9802373]
75. Eichenfield LF, et al. Safety and efficacy of pimecrolimus (ASM 981) cream 1% in the treatment of
mild and moderate atopic dermatitis in children and adolescents. J Am Acad Dermatol. 2002;
46(4):495–504. [PubMed: 11907497]
76. Ho VC, et al. Safety and efficacy of nonsteroid pimecrolimus cream 1% in the treatment of atopic
dermatitis in infants. J Pediatr. 2003; 142(2):155–62. [PubMed: 12584537]
77. Kang S, et al. Long-term safety and efficacy of tacrolimus ointment for the treatment of atopic
dermatitis in children. J Am Acad Dermatol. 2001; 44(1 Suppl):S58–64. [PubMed: 11145796]
78. Paller A, et al. A 12-week study of tacrolimus ointment for the treatment of atopic dermatitis in
Author Manuscript

pediatric patients. J Am Acad Dermatol. 2001; 44(1 Suppl):S47–57. [PubMed: 11145795]


79. Schachner LA, et al. Tacrolimus ointment 0.03% is safe and effective for the treatment of mild to
moderate atopic dermatitis in pediatric patients: results from a randomized, double-blind, vehicle-
controlled study. Pediatrics. 2005; 116(3):e334–42. [PubMed: 16140675]
80. Ashcroft DM, et al. Efficacy and tolerability of topical pimecrolimus and tacrolimus in the
treatment of atopic dermatitis: meta-analysis of randomised controlled trials. BMJ. 2005;
330(7490):516. [PubMed: 15731121]
81. Broeders JA, et al. Systematic review and meta-analysis of randomized clinical trials (RCTs)
comparing topical calcineurin inhibitors with topical corticosteroids for atopic dermatitis: A 15-
year experience. J Am Acad Dermatol. 2016; 75(2):410–9. e3. [PubMed: 27177441]
82. Hebert AA, et al. Desoximetasone 0.25% and tacrolimus 0.1% ointments versus tacrolimus alone
in the treatment of atopic dermatitis. Cutis. 2006; 78(5):357–63. [PubMed: 17186796]
83. Paller AS, et al. Efficacy and safety of crisaborole ointment, a novel, nonsteroidal
phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in
Author Manuscript

children and adults. J Am Acad Dermatol. 2016; 75(3):494–503. e4. [PubMed: 27417017]
84. Harper JI, et al. Cyclosporin for severe childhood atopic dermatitis: short course versus continuous
therapy. Br J Dermatol. 2000; 142(1):52–8. [PubMed: 10651694]
85. Pedreira CC, et al. Oral cyclosporin plus topical corticosteroid therapy diminishes bone mass in
children with eczema. Pediatr Dermatol. 2007; 24(6):613–20. [PubMed: 18035982]
86••. Sidbury R, et al. Guidelines of care for the management of atopic dermatitis: section 3.
Management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;
71(2):327–49. This is a comprehensive set of guidelines from the American Academy of
Dermatology on the systemic treatments of atopic dermatitis. [PubMed: 24813298]
87. Murphy LA, Atherton D. A retrospective evaluation of azathioprine in severe childhood atopic
eczema, using thiopurine methyltransferase levels to exclude patients at high risk of
myelosuppression. Br J Dermatol. 2002; 147(2):308–15. [PubMed: 12174104]
88. El-Khalawany MA, et al. Methotrexate versus cyclosporine in the treatment of severe atopic
dermatitis in children: a multicenter experience from Egypt. Eur J Pediatr. 2013; 172(3):351–6.
[PubMed: 23229188]
Author Manuscript

89. Heller M, et al. Mycophenolate mofetil for severe childhood atopic dermatitis: experience in 14
patients. Br J Dermatol. 2007; 157(1):127–32. [PubMed: 17489974]
90. Daley-Yates PT, Richards DH. Relationship between systemic corticosteroid exposure and growth
velocity: development and validation of a pharmacokinetic/pharmacodynamic model. Clin Ther.
2004; 26(11):1905–19. [PubMed: 15639702]
91. Schmitt J, et al. Prednisolone vs. ciclosporin for severe adult eczema. An investigator-initiated
double-blind placebo-controlled multicentre trial. Br J Dermatol. 2010; 162(3):661–8. [PubMed:
19863501]

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 15

92. Morison WL, et al. Oral psoralen photochemotherapy of atopic eczema. Br J Dermatol. 1978;
98(1):25–30. [PubMed: 626712]
Author Manuscript

93. Tay YK, et al. Experience with UVB phototherapy in children. Pediatr Dermatol. 1996; 13(5):406–
9. [PubMed: 8893243]
94. Clayton TH, et al. The treatment of severe atopic dermatitis in childhood with narrowband
ultraviolet B phototherapy. Clin Exp Dermatol. 2007; 32(1):28–33. [PubMed: 17305905]
95. Jury CS, et al. Narrowband ultraviolet B (UVB) phototherapy in children. Clin Exp Dermatol.
2006; 31(2):196–9. [PubMed: 16487089]
96. Hearn RM, et al. Incidence of skin cancers in 3867 patients treated with narrow-band ultraviolet B
phototherapy. Br J Dermatol. 2008; 159(4):931–5. [PubMed: 18834483]
97. Levy BD. Lipoxins and lipoxin analogs in asthma. Prostaglandins Leukot Essent Fatty Acids.
2005; 73(3–4):231–7. [PubMed: 16046112]
98. Wu SH, et al. Efficacy and safety of 15(R/S)-methyl-lipoxin A(4) in topical treatment of infantile
eczema. Br J Dermatol. 2013; 168(1):172–8. [PubMed: 22834636]
99. Lacombe Barrios J, et al. Anti-IgE therapy and severe atopic dermatitis: a pediatric perspective. J
Am Acad Dermatol. 2013; 69(5):832–4. [PubMed: 24124824]
Author Manuscript

100. Wang HH, et al. Efficacy of omalizumab in patients with atopic dermatitis: A systematic review
and meta-analysis. J Allergy Clin Immunol. 2016; 138(6):1719–22. e1. [PubMed: 27543070]
101. Hanifin JM, et al. Recombinant interferon gamma therapy for atopic dermatitis. J Am Acad
Dermatol. 1993; 28(2 Pt 1):189–97. [PubMed: 8432915]
102•. Brar K, Leung DY. Recent considerations in the use of recombinant interferon gamma for
biological therapy of atopic dermatitis. Expert Opin Biol Ther. 2016; 16(4):507–14. Recombinant
interferon gamma is a therapeutic agent that has shown efficacy in treatment of atopic dermatitis
patients. [PubMed: 26694988]
103. Simpson EL, et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N Engl
J Med. 2016; 375(24):2335–48. [PubMed: 27690741]
104. Brunner PM, et al. The immunology of atopic dermatitis and its reversibility with broad-spectrum
and targeted therapies. J Allergy Clin Immunol. 2017; 139(4S):S65–S76. [PubMed: 28390479]
Author Manuscript
Author Manuscript

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.
Huang et al. Page 16

Table 1

Topical Corticosteroid Potencies, Strengths, and Formulations,


Author Manuscript

Class I: Superpotent/Very High Potency Class IV: Mid-Strength/Medium Potency

Betamethasone dipropionate (ointment) Desoximetasone, 0.05% (cream, ointment)


Clobetasol propionate, 0.05% (cream, ointment, solution, spray, shampoo) Fluocinolone acetonide, 0.03% (ointment)
Clobetasol propionate (foam) Flurandrenolide, 0.05% (ointment)
Desoximetasone, 0.25% (spray) Hydrocortisone valerate, 0.2% (ointment)
Diflorasone diacetate, 0.05% (ointment) Mometasone furoate, 0.1% (cream)
Fluocinonide, 0.1% (cream) Triamcinolone acetonide, 0.1% (cream, spray)
Flurandrenolide, 0.05% (Cordran Tape)
Halobetasol propionate, 0.05% (cream, ointment, lotion)

Class II: Potent/High Potency Class V: Lower Mid-Strength/Lower-Medium Potency


Author Manuscript

Betamethasone dipropionate, 0.05% (cream) Desonide, 0.05% (lotion)


Desoximetasone, 0.25% (cream, ointment) Fluocinolone acetonide, 0.03%/0.01% (cream)
Desoximetasone, 0.05% (gel) Fluocinolone acetonide, 0.01% (shampoo)
Diflorasone diacetate, 0.05% (cream, ointment) Flurandrenolide, 0.05% (cream, lotion, tape)
Fluocinonide, 0.05% (cream, gel, ointment) Fluticasone propionate, 0.05% (cream, lotion)
Halcinonide, 0.1% (ointment, cream) Hydrocortisone, 0.1% (cream, lotion, ointment, solution)
Mometasone furoate, 0.1% (ointment) Hydrocortisone valerate, 0.2% (cream)
Prednicarbate, 0.1% (cream)

Class III: Upper Mid-Strength/Medium Potency Class VI: Mild/Low Potency

Betamethasone valerate, 0.12% (foam) Alclometasone dipropionate, 0.05% (cream, ointment)


Author Manuscript

Fluocinonide, 0.05% (cream) Desonide, 0.05% (gel, foam)


Fluticasone propionate, 0.005% (ointment) Fluocinolone acetonide, 0.01% (cream, solution, oil)

Class VII: Least Potent/Lowest Potency

Hydrocortisone, 0.5%/1% (lotion)


Hydrocortisone, 1%/2.5% (cream, lotion)
Hydrocortisone, 2%/2.5% (cream)
Author Manuscript

Curr Treat Options Allergy. Author manuscript; available in PMC 2018 September 01.

You might also like