You are on page 1of 22

Immunopharmacology and Immunotoxicology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iipi20

Atopic dermatitis: new insight into the etiology,


pathogenesis, diagnosis and novel treatment
strategies

Deepa S. Mandlik & Satish K. Mandlik

To cite this article: Deepa S. Mandlik & Satish K. Mandlik (2021) Atopic dermatitis: new insight
into the etiology, pathogenesis, diagnosis and novel treatment strategies, Immunopharmacology
and Immunotoxicology, 43:2, 105-125, DOI: 10.1080/08923973.2021.1889583

To link to this article: https://doi.org/10.1080/08923973.2021.1889583

Published online: 28 Feb 2021.

Submit your article to this journal

Article views: 181

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iipi20
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY
2021, VOL. 43, NO. 2, 105–125
https://doi.org/10.1080/08923973.2021.1889583

REVIEW

Atopic dermatitis: new insight into the etiology, pathogenesis, diagnosis and
novel treatment strategies
Deepa S. Mandlika and Satish K. Mandlikb
a
Bharat Vidyapeeth, Deemed to be University, Poona College of Pharmacy, Pune, India; bSinhgad College of Pharmacy, Vadgaon,
Pune, India

ABSTRACT ARTICLE HISTORY


Atopic dermatitis (AD) is the long-lasting chronic inflammatory skin condition associated with cutane- Received 14 August 2020
ous hyper-reactivity and triggered by environmental factors. The attributes of AD include dry skin, Accepted 5 February 2021
pruritus, lichenification and frequent eczematous abrasions. This has a strong heritable aspect and typ-
KEYWORDS
ically occurs with asthma and allergic rhinitis. The complex pathological mechanism behind AD eti-
Atopic dermatitis;
ology is epidermal barrier destruction resulting in the lack of filaggrin protein that can induce pathogenesis; glucocorticos-
inflammation and T-cell infiltration. T-helper 2 cell-mediated pathways also bear the responsibility of teroids; systemic therapy;
damage to the epidermal barrier. Certain causative factors for AD include microbial imbalance of skin phototherapy; adjuvant
microbiota, immunoglobulin-E-induced sensitization and neuro-inflammation. Numerous beneficial top- treatment; natural products;
ical and oral treatments have been available to patients and there are even more drugs in the pipeline nonadherence
for the treatment of AD. Topical moisturizers, corticosteroids, anti-inflammatory agents such as calci-
neurin inhibitors, phototherapy, cAMP-specific 3, 5 half-cyclic phosphodiesterase 4 inhibitors and sys-
temic immunosuppressants are widely available for AD treatments. Different positions and pathways
inside the immune system including JAK-STAT, phosphodiesterase 4, aryl hydrocarbon receptor and T-
helper 2 cytokines are targeted by above-mentioned drug treatments. Instead of the severe side
effects of topical steroids and oral antihistamines, herbal plants and their derived phytoconstituents
are commonly used for the treatment of AD. A clear understanding of AD’s cellular and molecular
pathogenesis through substantial advancement in genetics, skin immunology and psychological fac-
tors resulted in advancement of AD management. Therefore, the review highlights the recent advance-
ments in the understanding of clinical features, etiology, pathogenesis, treatment and management
and non-adherence to AD treatment.

Introduction as per the various clinical measurement scales, the most


commonly employed of which are the Investigator Global
Scientists, Wise and Sulzberger first invented the word AD to
Evaluation (IGA), the Eczema Region and Frequency Rating
explain puzzling kinds of local or general skin lichenification
(EASI) and the SCORing Atopic Dermatitis (SCORAD) scale [5].
[1]. AD is a long-lasting, inflammatory and pruritus skin dis-
AD shows a varied range of clinical skin features. The clinical
ease generally observed in early childhood but may also con-
features of AD are given in Table 1. Present research indi-
tinue in adulthood [2]. AD is distinguished by a hyperactive
cates that AD is a key skin barrier deficiency that causes cer-
immune reaction to environmental influences that results in
dry, itchy skin. Skin lesions can cause significant emotional tain atopic disorders to evolve [6]. Also, AD is always the first
stress and a vivid burden on the quality of life of patients. stage in the ‘atopic march’ (the concurrent progression of
Patients are not only impacted by the common perception symptoms of allergic infection in infancy), developing bron-
of a prominent skin disorder, but the extreme scratching chial asthma and/or allergic rhinitis in most affected patients
characteristic of the diseases also contributes to skin discom- [7]. Interestingly, initial AD might be an etiological factor in
fort and severe sleep disruptions. Skin lesions may be caused food allergy progression [8]. New findings into AD indicate
by stress, interaction with allergens, scrabbling and many that both skin structure defects and immune dysregulation
other reasons [3]. The incidence of AD is projected to be 10 play significant roles in the disease’s pathophysiology. Of this
to 15 percent in children and two to 10 percent in adults in cause, effective treatment of AD involves a multidimensional
the Western population. Two forms of AD were primarily approach to repair and maintain the skin barrier and resolv-
established, the extrinsic form linked to IgE intermediate sen- ing the multifarious immunopathogenesis of the diseases [9].
sitization and the intrinsic form without IgE intermediate sen- A breakthrough in translational science in recent years has
sitization [4]. AD is characterized for its variations, potential broadened understanding of AD pathogenesis and contrib-
reversibility and irregular progression in the patient life. uted to the creation of novel molecules that target core
Twenty percent cases of AD will be graded as mild to severe inflammatory features of the disease. The review article offers

CONTACT Deepa S. Mandlik deepa_ingawale@yahoo.com Department of Pharmacology, Poona College of Pharmacy, Pune, India
ß 2021 Informa UK Limited, trading as Taylor & Francis Group
106 D. S. MANDLIK AND S. K. MANDLIK

Table 1. Clinical features of AD.


Clinical sign Description
Pruritus The unpleasant sensation of the skin that provokes the urge to scratch
Xerosis Dry skin in areas without clinically apparent inflammation
Ichthyosis vulgaris Fish-like dry scales that can often look extremely thick and dry
Keratosis pilaris Appearance of thick scale and redness around the hair follicles that may be
surrounded by a patchy erythema
Follicular prominence Follicles have a goose-bump appearance
Palmar & plantar hyperlinearity Patients more often have exaggerated palmar hand creases than
plantar creases.
Dennie-Morgan lines Dark, symmetric, double horizontal folds below the lower eyelids
Periorbital darkening Gray to violet-brown discoloration and swelling around the eyes
Anterior neck folds Horizontal folds or lines across the middle of the anterior neck
Hertoghe sign Loss of the lateral third of the eyebrows because of constant scratching
White dermatographism Blanching response as a result of stroking of the skin with the back of a
fingernail that leads to white streaks
Pityriasis alba Consists of multiple ill-defined light (hypopigmented) patches with fine
scaling that are often located on the face and neck and occasionally
appear on the shoulders and arms

a summary of existing research on etiology, disease patho- Environment


physiology with possible use for therapeutic targets, diagno-
Several different environmental risk factors for AD have been
sis and effective treatment of AD.
considered potentially causative, only a few are widely recog-
nized. There is significant evidence, for example, that our
western lifestyle has contributed to some of the recorded
rise in the incidence of eczema over the past few years but
Etiology of AD
this did not point to particular environmental risk factors or
In those family whose members are affected, the risk of explicitly translate into functional preventive measures [13].
developing AD is substantially greater. For example, in Some support the hygiene hypothesis when describing the
monozygotic twins, the concordance rate of AD is about 75 dramatic rise in the prevalence of eczema [14]. This hypoth-
percent, which means that if the cotwin is infected, the risk esis notes that decreased early childhood exposure to proto-
of disease in the twin sibling is 75 percent [10]. The risk in typical infections like hepatitis A and tuberculosis has
dizygotic twins, on the other hand, is just 30%. It indicates enhanced atopic disease susceptibility [15]. The theory is
that genetic factors play a part in AD vulnerability. Because backed by the findings that the youngest sibling has the
there is no full agreement between monozygotic twins, how- lowest risk of AD and that children growing up in a typical
ever, who share all of their genes, environmental and devel- agricultural community where they are exposed to a range
opmental factors may also play a role. As such, AD is a of microflora, such as unpasteurized cow’s milk, livestock and
complex genetic disorder that results from many interactions poultry, are to some degree shielded against disease devel-
between the gene-gene and gene-environment. opment and allergic disease [16]. Conversely, the incidence
of the disease is likely to be positively associated with the
period of breastfeeding [17], although several studies have
linked the parents’ high social status to an enhanced risk of
Genetics AD in the infant. While these findings are not easy to under-
stand, they may also support the hygiene hypothesis or at
Several genes have been linked to AD, mainly genes that least the widely established theory that eczema arises in
encode epidermal structural proteins and genes that encode genetically susceptible individuals that are subjected to a
the main elements of the immune system. The reported certain detrimental climate [18].
strong association between AD and mutations in the filag-
grin gene (a genetic risk factor for AD), located on chromo-
some 1 is a recent and important genetic discovery [11]. Pathophysiology of AD
Approximately 10 percent of people from western cultures AD’s pathophysiology is dynamic and multifactorial, contain-
have mutations in the filaggrin gene, while about 50 percent ing aspects of barrier failure, shifts in cell-mediated immune
of all patients with AD have those mutations. Mutations of responses, IgE-induced hypersensitivity and environmental
the filaggrin gene cause functional impairments in the filag- influences. It also occurs due to a complicated interface
grin protein and thus weaken the skin barrier. The clinical between accountable genes, a climate of the host, pharma-
manifestation of these impairments is dry skin and an ele- cological abnormalities and immunological factors [19].
vated risk of eczema. Not all patients with AD have these
mutations, and there have also been other genetic variations
Skin barrier dysfunction
incriminated. This is the cumulative action of both of these
genetic variations along with AD causing environmental and The skin serves as a physicochemical shield to numerous
developmental risk factors [12]. physical, chemical and biological influences that are
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 107

Table 2. Elements of skin barrier dysfunction in AD.


Component Description
Filaggrin gene expression Filaggrin is an essential skin protein that is responsible for cross-linkage of
keratin filaments into tight bundles and stratum corneum hydration.
Around 50 percent of AD patients display mutation in the filaggrin
gene [11].
Skin Ceramide Phospholipid is essential for preservation of water in the outermost layer of
the skin (stratum corneum). In the stratum corneum of AD patient, there
is an inverse relationship between the ceramide levels and the
transepidermal water loss [22].
Epidermal proteases The proteases responsible for desquamation of corneal desmosomes are
Kallikrein (KLK)-5, KLK-7, and KLK-14. Lymphoepithelial Kazal-type 5 serine
protease inhibitor is expressed by serine protease inhibitor Kazal type 5
genes. The mutation in SPINK5 was related to the exacerbation of AD
symptoms [23].

responsible for causing AD. Proksch et al., have examined In response to allergens invasion, skin-inhabitant keratino-
core facets of skin barrier failure in AD [20]. A variety of gen- cytes and Langerhans cells (LCs) produce cytokines like IL-10,
etic vulnerability loci responsible for skin barrier activity is IL-12, IL-18 and chemokines that cause stimulation of other
noted by genome-wide linkage studies [21]. The role of these inflammatory cells like neutrophils, eosinophils, basophils
individual genes in AD pathogenesis requires additional and T cells. Cytokines, such as IL-10 and IL-23, respectively,
exploration but at best three main motives for barrier dys- stimulate differentiation of naïve T cells into Th2 and Th17.
function have been identified in AD patients: (a) reduced The Th17 cells stimulate the release of IL-17A, IL-17E and IL-
gene expression of filaggrin, (b) deficiency of ceramides and 22 cytokines. The change from an acute to a chronic inflam-
(c) over manifestation of epidermal proteases (Table 2). matory phase is triggered by intensified IL-12 and IL-18 pro-
duced from inflammatory epidermal dendritic cells (IDECs) or
eosinophils or both. During chronic AD, dendritic cells
Immunological factors release IL-12 and IL-18 lead to activation of the Th1 cells.
There has been much discussion in the literature about the IFN- c released from Th1 cells induces keratinocyte apoptosis
pathophysiology of IgE mediated or extrinsic AD. The pri- while IL-22 promotes skin modification and chronic AD skin
mary passageway leading to the inflammation of the skin is thickness. A recent study further concludes that stimulation
still not clear. This may occur due to scratching of the skin of Toll-like receptors 2 (TLR2) on DCs converts Th2 dermatitis
that induces the keratinocytes to stimulate pro-inflammatory to chronic skin inflammation in mouse models [29]. Both the
cytokines or may trigger due to contact between T-cell and stages of AD have been reported to contain a substantial
infiltrated allergens from interrupted skin barrier [24]. level of Th2 cytokines compared to normal skin; however,
Togetherly adaptive and non-adaptive immunity have an chronic skin lesions are associated to improved IL-5 and IL-
important role in the pathophysiology of AD. The adaptive 12 and decreased IL-4 and IL-13 release [30].
immune responses are greatly specific to a precise antigen
while non-adaptive immune responses are unspecific. For Environmental and microbial factors
any repeated antigen exposure, the adaptive immune
responses get stronger [25]. Multiple cells which reside in AD occurs due to the interaction between hereditary and
the skin like dendritic cells (DCs), keratinocytes, macro- environmental influences. Many environmental factors like
phages, mast cells (MCs), and innate lymphoid cells (ILCs) bacteria, viruses, airborne allergens, food, smoking, soaps
play a significant part in skin inflammation of AD patients and detergents can trigger the inflammatory pathway in AD
[26]. Rather than skin resident cells, T cells, granulocytes, and cause the interruption of the epidermal barrier. The
monocytes, and plasmacytoid dendritic (pDCs) from blood essential environmental factors have been mentioned in
circulation, also play an imperative role in the progress of Figure 1 and Table 3.
AD skin lesions [27]. After the presentation of antigen from
DCs into the skin, the naive T cell is divided into T helper
Diagnosis of AD
(Th) 1 and Th2-cells. It is commonly believed that, regardless
of the relative disruption in the equilibrium between Th1/ There are no screening measures to identify AD. Disorder
Th2 cells, AD occurs, with total Th2 cell superiority [28]. The evaluation is based upon the patient history and clinical
inflammatory reaction is a two-step process, original, acute, symptoms of the disease’s condition [38]. Figure 2 discusses
Th2 dominant process accompanied by a chronic Th1 like the diagnostic criteria of AD. Through these benchmarks, the
profile. In the acute AD stage, allergens through disrupted finding of AD includes the existence of a scratchy skin dis-
skin barrier stimulate the degranulation of mast cells (MC) order or report of itching or rubbing of the skin by the par-
triggering the release of histamine, interleukin (IL)-6, IL-8, ental/caregiver in a child plus three or more minor criteria,
prostaglandins D2 and tumor necrosis factor (TNF)-a along which differ based on the age of the patient. Irrespective of
with IL-23 and IL-31. Along with these mediators, damaged age, the scratching associated with AD usually persists dur-
epithelial cells release thymic stromal lymphopoietin (TSLP) ing the day and worsens at night, contributing to lack of
which results in increased Th2 type inflammatory response. sleep and severe impairments in quality of life.
108 D. S. MANDLIK AND S. K. MANDLIK

Differentiating AD from certain skin disorders such as contact Prevention of AD


dermatitis, seborrheic dermatitis, scabies and psoriasis is
often difficult; nevertheless, in certain instances, a familial Though there are currently no proven primary preventive
history of atopy and lesions scattering is beneficial in deter- methods for AD, the efficacy of early, reliable application of
mining the diagnosis. Nutritional abnormalities, malignancies emollients to infants at elevated risk has been shown in
and keratinization or immunodeficiency diseases correlated recent clinical trial studies. This quick and cost-effective
with skin symptoms are other factors that need to be method has culminated in a decrease of 30–50 percent of
addressed in the differential diagnosis of AD [9]. AD diagnosis at 6 months [39].

Treatments of AD
AD treatment should be aimed at repairing the skin barrier
that involves moisturizing and maintenance of the skin, pre-
venting scratching, and reducing inflammation when needed.
Hence, proper treatment of AD includes a multidimensional
strategy including patient and caregiver awareness, appropri-
ate skin maintenance procedures, treatment with anti-inflam-
matory drugs like topical corticosteroids (first-line) and/or
topical calcineurin inhibitors (TCIs) and skin infection man-
agement [40]. For extreme situations, systemic immunosup-
pressive agents can also be used that cannot be managed
with adequate skincare and topical therapy. Though first-
generation antihistamines are not widely prescribed for AD
treatment due to their sedative and damaging side effects,
acute usage of such drugs can be beneficial in certain per-
sons with serious AD flares, mainly if such flares are corre-
lated with major sleep disorders [41]. Physicians will
routinely track patient improvement and progression of the
illness, and assess treatment efficacy and tolerability. The
treatment pattern of AD and treatment in adult and children
Figure 1. Different risk factors affecting on AD. are mentioned in Figure 3 and Table 4.

Table 3. Environmental factors in AD.


Factors Description
Airborne allergens The inflammatory reaction in sensitized atopic skin can be triggered by
airborne allergens. House dust mites (HDM) obtained from the species
Dermatophagoides pteronyssinus and D. farina are the commonest source
of airborne allergens. The HDM can destroy the skin barrier by abolishing
the tight epithelial junctions by enzymatic activity [31].
Food Food allergy in children is well recognized in around one-third of cases of
AD. Milk, fish, for example, protein, soya, peanut and nuts are mostly seen
among food allergens to exacerbate AD conditions.
Microbial exposure S. aureus colonization of the atopic skin influenced by dysfunction of the skin
barrier, elevated skin pH, elevated content of fibronectin, and reduced
ceramides and low antimicrobial peptide levels [32]. S. aureus also
documented to secrets enterotoxin A and B and toxin-1, playing a major
function in aggravating AD [33]. In addition, S. aureus secretes proteinases
capable of breaking down corneodesmosomes [34]. AD patients are more
prone to skin infections like Verruca vulgaris, Molluscum contagiosum and
Herpes simplex than non-AD patients.
Soap and Detergents The acidic pH of human skin greatly leads to barrier activity against multiple
antigens. This serves as an antibacterial agent that regulates desquamation
of corneodesmosomes. The change in skin pH to greater side can
aggravate lesions of AD skin. Soaps induce skin pH rise, lipid
emulsification on skin surface, skin protease enhancement and subsequent
thinning of stratum corneum [35].
Weather factor and Smoking Humidity, temperature and UV sensitivity affect the production of AD skin
lesions. In AD incidence, the combination of high UV intensity and
temperature tends to have a shielding effect, while high humidity and
precipitation are correlated with progressive impact on AD [36]. Active
smoking behavior was not considered as a risk in AD but maternal
smoking was identified as a danger in children with AD [37].
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 109

Figure 2. Diagnostic criteria for AD.

Figure 3. Treatment pattern of AD.

Table 4. Treatment of AD in adults and children [5].


Severity of AD AD management in adult and children
Baseline AD Educational program, Avoidance of allergens, Emollients, Bath oils
Mild AD Topical glucocorticosteroids, Topical calcineurin inhibitors, Antiseptics
containing silver
Moderate AD Tacrolimus or Class II or Class III topical glucocorticosteroids, UV therapy, Wet
wrap therapy, Psychosomatic counseling
Severe AD Hospitalization, Systemic immunosuppressive agents like Oral
glucocorticosteroids, Cyclosporine A, Mycophenolate mofetil, Duplimab,
Methotrexate, Azathioprin

Education
with inadequate adherence, in particular with topical treat-
Patients and/or their carers should be informed regarding ments, arising from unreasonable concerns about the side
the progressive existence of the disease’s condition, the effects and inadequate diseases knowledge [9]. This has
necessity for consistent commitment to appropriate skincare been found that time invested in discussing these concerns
procedures, and the correct usage of topical treatments for and training patients and carers have a significant impact on
effective disease control. Poor care results are also correlated the outcomes of the disease. For better understanding,
110 D. S. MANDLIK AND S. K. MANDLIK

Table 5. Skin care in AD.


Methods Recommendations
Cleansing and bathing Smooth and thorough cleaning is done to avoid surface microbial contamination.
Nonirritating cleansers like syndets, aqueous solutions can be used with or without antiseptics.
In case of infants bathing, use of baby oil is advised in the last stage of bathing.
For antibacterial action, sodium hypochlorite (0.005 %) may be used as an antiseptic in bathwater [44].
Emollient treatment It is recommended to apply the emollients twice a day.
Tannins and ichthammol emollients are advised for the treatment of AD.
Emollient formulations without protein allergens and haptens can be used.
Non-medicated emollients containing riboflavins, flavonoids and saponins were suggested for the AD treatment [45].

Topical anti-inflammatory therapy


Topical anti-inflammatory AD therapy relies on three funda-
mental principles; appropriate dose, sufficient intensity and
proper usage. There are currently three types of topical anti-
inflammatory drug therapy approved by USFDA for the treat-
ment of AD. Those involve topical corticosteroids, calcineurin
inhibitors and phosphodiesterase 4-inhibitor (PDE4 inhibitor).
Topical anti-inflammatory treatment is usually performed
only on skin lesions and minimized as the flares are
decreased. Nowadays, this type of therapy is more structured
Figure 4. Examples of flare factors in AD.
and recognized as preventive therapy that is triggered by a
systemic anti-inflammatory treatment after the effective heal-
patients may also provide with detailed instructions/informa- ing of lesions [46]. Regardless of advancements in the pro-
tion on proper drug usage, skincare, and flare prevention. duction of systemic medications, topical therapies appear to
be important for both the renovation and distribution of
anti-inflammatory drugs in barrier functions. Furthermore,
ointments, calcineurin inhibitors and corticosteroids newer
drugs have been established for the topical use. Various top-
Skin care
ical drugs under different phases of clinical trials are men-
Good routine skincare is a crucial function in AD manage- tioned in Table 6.
ment. While bathing the frequency is a little uncertain, most
experts recommend bathing every day [42]. Bathing in warm
water one or two times in a day (based on the intensity of Glucocorticosteroids
AD) for ten minutes is advised to help hydrate and cleaning Topical glucocorticosteroids are the first alternative for the
of the skin, help debride diseased skin and increase the per- treatment of inflammatory skin in AD. In case of moderate
meation of topical drugs. Moisturizing cleansing agents are AD, topical applications of corticosteroids two or three times
advised whereas avoiding heavily fragrant soaps because in a week following the usage of emollients aids ineffective
they can irritate the skin. The skin of the patient should be treatment. The corticosteroids were categorized into a cat-
patted dry with a towel after bathing and moisturizers egory I (mild) to group IV (super potent) by Niedner based
should be used to avoid further loss of moisture and drying on their potency [47] (Figure 5). Super potent corticosteroids
of the skin. In the treatment of AD creams and ointments are not recommended to babies and children. The adrenal
are more powerful than lotions for nourishing the skin [43] gland functions are usually suppressed by group III (Potent)
(Table 5). and group IV (super potent) than group I and group II corti-
costeroids, but their systemic effects will reduce rapidly due
to immediate repair of the skin barrier [48]. Group I and II
corticosteroids would be used for the treatment of face
lesions, particularly the eyelid area. According to the US-
Identification and elimination of flare factor
American classification, the corticosteroids are classified from
Possible flare factors of AD may be recognized depending group VII (mild) to the group I (super potent). The long term
on the background and detailed allergy testing. The sensitiv- use of glucocorticosteroids produces numerous adverse
ity should be documented by in vivo tests (skin prick test effects include skin atrophy, spontaneous scars, ecchymosis,
and prick-prick test) and in vitro tests (serum specific IgE). stretch marks, dirty neck, hypertrichosis and perioral derma-
Food sensitivity can be tested by in vitro allergy studies, such titis [49]. In children, chronic topical usage of corticosteroids
as double-blind placebo-controlled food challenges, avoid- in the diaper area may induce granuloma gluteale infantum
ance of diets or patch checks for atopy. In nonspecific and or even iatrogenic Cushing ’s disease. Ocular adverse effects
specific flare factors are distinguished from each other and like cataract or glaucoma have been identified after systemic
examples are mentioned in Figure 4. glucocorticosteroids treatment [50].
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 111

Table 6. New topical treatments under development for the treatment of AD.
Biological agent Therapeutic target Severity Phase Status
Tofacitinib JAK 1, JAK 3 Mild to moderate II Completed
Ruxolitinib JAK 1, JAK 2 Mild to moderate II Completed
Delgocitinib PAN- JAK Mild to moderate II Completed
Crisaborol PDE4 Mild to moderate III Completed
MM36 PDE4 Mild to moderate II Completed
III Active
AN2898 PDE4 Mild to moderate II Completed
Rofiumilast PDE4 Mild to moderate IIa Completed
E6005 PDE4 Mild to moderate II Completed
Tapinarof Aryl hydrocarbon receptor agonist Mild to moderate II Completed
Omiganan Anti-microbial peptide Mild to moderate II Completed
VTP-38543 Liver X receptors receptor Mild to moderate I/II Completed
Q301/Zyleuton PG 2 trans-membrane receptor Mild to moderate II Completed
PR022 Hypochlorous acid Mild to moderate II Active
SP14019/Cyclato Ciclosporin Mild to moderate II Completed
JAK: Janus kinase; PDE4: Phosphodiesterase-4; PG: Prostaglandin.

not account for an elevated lymphoma hazard in a newly


released article [54].

Phosphodiesterase 4-inhibitors
In 2016, the FDA licensed a new topical anti-inflammatory
drug category that inhibits the cAMP-specific 3, 5-cyclic
phosphodiesterase 4 (PDE4) enzyme. Crisaborole ointment is
licensed by USFDA for the care of patients aged up to
2 years with mild to moderate AD. By inhibiting PDE4, crisa-
borole promotes second messenger cAMP activity, contribu-
ting to a decrease in pro-inflammatory cytokine production.
The reason for this therapy emerged from multiple experi-
ments that have shown that leukocytes from AD patients
had a blunted reaction to cAMP owing to elevated PDE4
activity [51]. Many investigational PDE4 antagonists are OPA-
15406 and E6005. In phase III trials, two years age patients
with mild to moderate AD, 32% of patients treated with cri-
saborole displayed nearly clear skin as compared to patients
Figure 5. Examples of common topical corticosteroid therapies.
treated with the vehicle no severe adverse effects have been
reported, and the main adverse effect at the application site
was burning or stinging [55]. Crisaborole crucial role in AD
Topical calcineurin inhibitors management is yet to be decided; however, it can provide
In the year 2000 and 2001, the FDA approved the topical cal-
another topical non-steroidal alternative for use in individuals
cineurin inhibitors, that is, tacrolimus and pimecrolimus cor-
with mild to severe AD [56].
respondingly [51]. Both the drugs act by inhibiting the
calcineurin phosphatase enzyme thus; impeding the activa-
Tapinarof. It is a nonsteroidal anti-inflammatory drug that
tion of T cells and mast cells with the secretion of cytokines.
functions as an aryl hydrocarbon receptor agonist, an activity
Additionally, both medications have been documented to
that may boost the barrier role and reduce the Th2 immune
have an increased impact on the skin AD. Kyllonen et al.
response. The usage of one per cent tapinarof two times in a
found that the application of 0.1% tacrolimus exhibited a sig-
nificant improvement in collagen production resulting in daily in a phase-II trial demonstrated substantial improve-
increased skin thickness and reversal of skin atrophy from ments in investigator global assessment score 0–1 and
the usage of corticosteroids [52]. Murrell and colleagues reduction of eczematous area and severity index, scoring AD
stated that pimecrolimus therapy results in substantial and body surface area relative to vehicle control [57].
changes in AD lesions, skin thickening of the neck and the
head area along with the eyelids [53]. Topical calcineurin Cyclatop (SP14019). It is a topical medication developed as
inhibitors offer a healthy substitute to corticosteroids for a five percent spray of cyclosporine under investigation in
treating vulnerable areas of the face and back skin. In March patients over 2 years of age in phase-II randomized clinical
2006, the FDA issued both pimecrolimus and tacrolimus a trial (NCT02865356). The findings, reported at the 2018
blanket black box warning over an elevated incidence of European dermatology congress, revealed substantial advan-
malignancies like lymphoma with topical calcineurin inhibi- tages of strong tolerability and minimal systemic absorption
tors. A meta-analysis of preclinical and clinical evidence does in eczematous area, severity index and investigator’s global
112 D. S. MANDLIK AND S. K. MANDLIK

assessment scale as compared to vehicles for four with 40 and 80 mg doses [65]. Topically administered JAK
weeks [58]. inhibitors are a successful treatment but additional clinical
trials are required in patients with AD.
Hypochlorous acid (PR022). Study of AD patients resulted
in a reduction of pruritus was correlated with the usage of
Phototherapy
hypochlorous acid (0.008 and 0.002 per cent) topically in a
hydrogel formulation. Hypochlorous acid is believed to Artificial UV radiation revelation has been documented as an
reduce the concentrations of various like TNF-a, histamine, effective treatment for atopic skin improvement. A new
interferon (IFN) - c, IL-2 [59]. study found that mild to moderate AD was entirely resolved
in 74 percent of patients, 16 percent showed marginal
Ominagan. It is a gel formulation of an antimicrobial pep- improvement and nine percent showed no change in AD at
tide that is being tested in phase-II randomized clinical trial; all. This concludes the infection’s seasonal variability, with
while there are no efficacy results, good tolerability was improvement during summer season and degradation in the
documented. The reduced antimicrobial peptide develop- other season. Furthermore, seaside holidays in sunshine
ment in AD promotes microbial invasion and infection and exhibited substantial change relative to holidays spent in
enhances inflammatory response [59]. mountain regions, with considerable reversal of AD situations
[66]. These findings do not fully explain the function of ultra-
violet (UV) exposure in AD but they encourage the medical
Janus associated kinase-signal transducer and activator group to further investigate the effect of UV radiation on AD.
of transcription (JAK-STAT) inhibitors Phototherapy enhances the atopic skin circumstances by
Most of the signaling cytokines in AD utilizes the JAK path- modifying the immune system by inflammatory cell apop-
way. Tofacitinib, a JAK inhibitor, has been shown to decrease tosis, inhibition of LCs and variability in cytokine production
inflammation in AD due to inhibition of interleukin-4 [60]. [67]. Additionally, UV radiations act as antimicrobials and
Already there are pending trials on its usage in AD, psoriasis lessen staphylococcus aureus colonization on atopic skin.
and alopecia areata. A Phase-IIa clinical trial found that two UVA1 treatment has also revealed the immunosuppression of
percent tofacitinib topical administration two times a daily IL-5, IL-13 and IL-31 [68]. Recent research supports the
had substantial-effectiveness when match with vehicle con- importance of vitamin-D during phototherapy in healing
trol ointment [61]. Currently, topical JAK/STAT receptor inhib- atopic lesions [69]. Phototherapy for two weeks exhibited a
itors are reportedly under review. These involve 2% topical marked change in the balance of vitamin D with an enlarged
tofacitinib and 1.5% ruxolitinib, which tend to be effective in level of serum calcidiol contributing to an effective cure of
minimizing EASI and pruritus [62]. A PAN-JAK inhibitor in a AD lesions. Reduction and functional loss of DCs in the skin’s
Japanese RCT also demonstrated EASI gains that were epidermal and dermal regions help immunosuppression
greater than with the vehicle alone, but no major harmful through the source of UV light [70]. Narrowband UVB (311-
effects [63]. Four orally administered JAK inhibitors are cur- 313 nm) performs more efficiently than UVB broadband.
rently under investigation. Baricitinib antagonizes JAK 1 and Medium UVA1 dose (340-400 nm) affects close to that of UVB
JAK2 that is linked to modulation of different cytokines. In narrowband. It is possible to use a combination of UVA and
phase-II clinical trial, oral administration of 4 mg/day bariciti- UVB for the treatment of AD lesions. Photochemotherapy
nib in patients with moderate to extreme AD was associated was also prescribed in conjunction with a photosensitizing
with a higher proportion of patients attaining Eczema Area drug given orally, that is, psoralens (PUVA: psoralen and
and Severity Index (EASI-50) compared to placebo with bet- ultraviolet A). Chronic use of all UV treatments, particularly in
ter tolerability. The reported adverse effects included asymp- PUVA, poses the risk of skin aging and cancer. Blue visible
tomatic creatine kinase elevation but no cases of thrombosis light has also been observed in AD for some promising
or herpes zoster were recorded [64]. The active inhibitors of results and is being investigated as other phototherapy
JAK1 are Upadacitinib (ABT-494) and Abrocitinib (PF- modes [71]. Photopheresis in selected cases of extreme
04965842) and show early progress with these medications. refractory AD has also shown a positive response. Other
A phase-IIb randomized clinical trial assessed the usage of excimer-308 nm laser monochromatic tools extend the thera-
Upadacitinib for 16 weeks at doses of 7.5, 15 and 30 mg peutic options in patients with localized and therapy-resist-
when compared with placebo. Several phase-III randomized ant AD [72]. Pulsed-dye lasers are currently under rigorous
clinical trial are underway with Abrocitinib at doses of 100 review for the treatment of chronic AD [73].
and 200 mg compared to placebo and a phase-III RCT as a
comparator with Dupilumab. In addition, ASN002 is a dual
Systemic therapy
inhibitor of JAK and spleen tyrosine kinase (SYK) pathways.
Inhibition of PAN-JAK inhibits the expression of several AD- Numerous types of medications, that is, antihistamines, cyclo-
implied cytokines such as IL-4, IL-13, IL-31 and IL-33, whereas sporine A, mycophenolate mofetil, selective serotonin
inhibition of SYK suppresses the signals of proinflammatory reuptake inhibitor and opioid receptor antagonists have
cytokines such as IL-1b, IL-10 and IL-17. In the first phase-Ib been systemically approved for immunosuppression.
randomized clinical trial with ASN002 the number of patients Different proof exists as to the effectiveness of antihist-
who reached EASI-50 at 28 days was higher than in placebo amines (H1 antagonists) for pruritus treatment in AD, since
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 113

most investigations exhibited a poor or no response to prur- Apremilast


itus [74]. The naltrexone opioid receptor antagonists have It is an oral PDE4 inhibitor approved for obstructive pulmon-
been found to alleviate pruritus in AD patients which can ary disease, psoriatic arthritis and plaque psoriasis. Its safety
have frequent side effects such as dizziness, headache, vom- and efficacy investigation in adult AD patients in an open-
iting, weakness, agitation, arthralgia and sleepiness [75]. The label pilot study revealed a substantial reduction in itching
drugs from selective serotonin reuptake inhibitors category and enhancement in quality of life [83]. Then, its beneficial
like paroxetine and fluvoxamine may be effective for the use has been documented in AD and other inflammatory
management of pruritus but are associated with several side skin conditions but more comprehensive randomized con-
effects such as dizziness, diarrhea, insomnia, constipation, trolled trials are needed [84].
erectile and ejaculatory dysfunction and diminished libido
[76]. Cyclosporine suppresses cytokine-stimulating transcrip-
Nemolizumab
tion of T cells. The drug interacts with intracellular cyclophi-
It is a monoclonal antibody that affects the IL-31 receptor. A
lin and thus acts in a similar way as tacrolimus and
phase II, placebo-controlled trial assessing safety and efficacy
pimecrolimus. Short duration cyclosporine with an enhanced
in patients with mild to serious AD reported a substantial
quality of life can support conventional drug therapy for AD
enhancement in AD conditions. Treatment in a dose-depend-
patients [77]. Furthermore, termination of treatment typically
ent manner provided noticeable relief in itching [85].
occurs in periods of relapse. The treatment of cyclosporine
Therefore, further work is needed to better understand the
associated with specific side effects like hypertension and
therapeutic impact of skin lesions and the history of the
renal inefficiency. For the treatment of refractory inflamma-
adverse effects.
tory skin diseases, mycophenolate mofetil, an antimetabolite
that inhibits biosynthesis of purine has been recommended
[78]. The daily dose of two gram has cleansed the adult AD Omalizumab
lesions that were resistant to steroid or PUVA therapy. It is a recombinant humanized monoclonal antibody that
Suppression of bone marrow was the main side effect identi- affects the IgE antibodies and has been approved by USFDA
fied with the mycophenolate mofetil treatment. Various sys- for allergic asthma and urticaria treatment [86]. It is a deriva-
temic drugs under different phases of clinical trials are tive of a monoclonal murine antibody, which is humanized
mentioned in Table 7. Many emerging types of medications to produce omalizumab [87]. Several reported studies sug-
are under review for the management of acute AD. gest that the omalizumab is effective in atopic dermatitis
and is well tolerated too. In a case study conducted by Holm
and colleagues, out of nine omalizumab-treated AD patients,
Duplimab 50% of the patient’s revealed excellent response, 12.5% dis-
In 2017, the FDA has approved Duplimab to treat mild to played moderate response, and 37.5% presented no effect to
severe AD. Duplimab is a fully humanized, monoclonal anti- the treatment. Researchers also carried out a systematic
body that affects the IL-4 receptor alpha subunit and thus review of 26 separate studies conducted to assess the thera-
inhibits IL-4 and IL-13 signaling [51]. Duplimab obstructs the peutic effectiveness of omalizumab in AD [88]. The research
mRNA expression that activates T cells, inflammatory cas- focused on large scale randomized clinical trials is still
cade, eosinophils, dendritic cells, Th2 cytokines and sup- awaited to support its clinical application in AD.
presses epidermal hyperplasia in AD [79]. It has also been
proposed that inhibition of IL-4 and IL-13 can not only min-
imize skin lesion severity but may also restore skin barrier Rituximab
function due to increased levels of claudine, loricrin, filaggrin It is an anti-CD20 antibody that causes reduction of circulat-
and lipid products. The suggested dose of duplimab is ing B cells. Rituximab therapy enhanced skin signs in
300 mg two times in a day followed subcutaneously by severely AD patients, indicating its possible function on B
300 mg every other week. Duplimab has less reported serious cells in the AD pathogenesis [89].
adverse reactions such as conjunctivitis, upper respiratory
tract infection, injection-site reactions and nasopharyngi- Pitrakinra
tis [80]. It is an inhibitor of IL-4 receptor signaling that obstructs the
binding of IL-4 and IL-13 to IL-4 a receptor complexes. It has
been proposed that the biologic material enhances the
Lebrikizumab and tralokinumab
asthma symptoms [90].
Lebrikizumab and Tralokinumab are the human monoclonal
antibody was stated to have substantial efficacy in moderate
to severe AD due to the blockade of IL-13 [81]. However, Infliximab
outcomes for both medications were deemed questionable It is a TNF antagonist was tested in a pilot study containing
because results were obtained in the clinical trials with sig- nine patients with moderate to extreme AD. The treatment
nificant use of topical corticosteroids. The drugs are under with infliximab has enhanced the signs but the effect
phase II clinical trial, for regulatory approval [82]. through maintenance therapy was not maintained [91].
114 D. S. MANDLIK AND S. K. MANDLIK

Table 7. New systemic treatments under development for the treatment of AD.
Biological agent Therapeutic target Route of administration Severity Phase Status
Dupilumab IL-4, IL-13 SC Moderate to severe III Completed
Mepolizumab IL-5 SC Moderate to severe II Discontinued
Lebrikizumab IL-13 SC Moderate to severe II Completed
Tralokinumab IL-13 SC Moderate to severe IIb Completed
III
KHK4083 OX40 IV Moderate to severe I Completed
II
GBR 830 OX40 IV Moderate to severe IIa Completed
SC IIb
Tezepelumab TSLP SC Moderate to severe IIa Completed
IIb
MK-8226 TSLP -R IV Moderate to severe I Discontinued
Secukinumab IL-17A SC Moderate to severe II Completed
MOR106 IL-17C IV Moderate to severe II Completed
Fezakinumab IL-22 IV Moderate to severe II Completed
Ustekinumab IL-12, SC Moderate to severe II Completed
IL-23
BMS-981164 IL-31 SC Moderate to severe I Discontinued
Nemolizumab IL-31RA SC Moderate to severe II Completed
Etokinab IL-33 SC Moderate to severe II Active
Omalizumab IL-E IV Moderate to severe II Completed
Ligelizumab IL-E SC Moderate to severe II Completed
KPL-716 OSMRb IV Moderate to severe Ia Completed
SC
Upadacitinib JAK 1 Oral Moderate to severe IIb Completed
III Active
Baricitinib JAK 1, JAK 2 Oral Moderate to severe I Completed
III Active
Abrocitinib JAK 1 Oral Moderate to severe IIb Completed
III Active
ASN002 JAK 1, JAK 2, JAK 3; SYK Oral Moderate to severe I Completed
II
Apremilast PDE 4 Oral Moderate to severe II Completed
ZPL389 HT 4 receptor Oral Moderate II Completed
JNJ-39758979 HT 4 receptor Oral Moderate II Discontinued
Tradipitant NK-1receptor Oral Mild, moderate or severe II Completed
II Completed
III Active
Serlopitant NK-1receptor Oral Mild, moderate or severe II Completed
DS107 Dihomo-c-linoleic acid Oral Moderate to severe II Completed
Asimadolin Kappa opioid receptor agonist Oral Mild, moderate or severe II Completed
IL: Interleukin; TSLP: Thymic stromal lymphopoietin; OSMRb: Deficiency of oncostatin M receptor b; SYK: Spleen tyrosine kinase; PDE 4: Phosphodiesterase-4;
HT4: Histamine 4; NK-1: Neurokinin 1 receptor.

Mepolizumab inflammation of all kinds. AMG 157 is a human anti-TSLP


It is a humanized monoclonal antibody binding to the IL-5 monoclonal immunoglobulin which binds to human TSLP
receptor, did not cause clinical progress in AD patients follow- and prevents interaction between receptors. Treatment with
ing a substantial reduction in serum eosinophilic count [92]. anti-TSLP antibody in patients with mild allergic asthma with
Furthermore, a recent double-blind study found that, in patients elevated allergen-induced early and late asthma responses
with extreme eosinophilic asthma, mepolizumab had a major [95]. Phase-I clinical trial was conducted with an anti-TSLP
glucocorticoid-sparing effect [93]. Additional research is needed antibody in a healthy individual and moderate to severe AD
to decide whether anti-IL5 therapy can be used to treat AD. patients [96].

Tocilizumab
It is an IL-6 receptor humanized monoclonal antibody used Treatment of skin infections
for the management of rheumatoid arthritis. Current research The skin of AD patient gets severely populated with the S.
demonstrated the possible efficacy of disturbing activation of Aureus bacterial infection. Short duration topical and oral
IL-6-receptors in AD patients [94]. Nevertheless, the correl- antibiotic treatment is required when there is an apparent
ation of bacterial superinfections with the therapy was also
bacterial infection. Suitable systemic antibiotics are recom-
identified. More research is required to examine the effi-
mended for extensive secondary infections. First- or second-
ciency and safety of IL-6 receptor antagonists.
generation cephalosporin or anti-staphylococcal penicillin for
seven to 10 days are usually efficacious in controlling the
Anti-TSLP bacterial infection. Macrolides antibiotics are less effective
It is a cytokine obtained from epithelial cells that performs alternatives for erythromycin-resistant organism’s infection in
the main part in the maturation of T-cell by activating anti- AD patients. Often, AD patients are vulnerable to recurrent
gen-presenting cells. Production of TSLP can cause viral infections. Eczema herpeticum (a serious disseminated
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 115

herpes infection that typically occurs at skin damage sites), is Evening primrose oil, black currant seed oil and borage oil:
a significant risk in patients with widespread AD and can be Due to its gamma-linoleic acid content, primrose oil at night
certainly misdiagnosed as a bacterial infection, requiring sys- was thought to be helpful in AD patients. Even though a cur-
temic antiviral treatment with acyclovir or other antiviral rent meta-analysis has revealed that primrose oil has unsuc-
agents [97]. Among children with AD, molluscum contagio- cessful to substantially recover AD, it is nevertheless utilized
sum (a serious viral cutaneous infection caused by as an adjuvant treatment in AD with a limited danger of side
Molluscipox genus poxvirus) is repeatedly seen. Although the effects [102].
infection is typically self-limited, the lesions also gradually Sunflower seed oil: Both anti-inflammatory and membrane
resolve and continue to spread in AD patients. Severe, per- restorative properties have been reported with the sunflower
sistent infection with the molluscum contagiosum can require seed oil. Its main lipid is linoleic acid that is supposed to
laser and/or antiviral therapy. It is also recommended that reduce skin inflammation by stimulating the peroxisome pro-
diluted baths with bleach help raising the amount of S. aur- liferative-activated receptor-alpha. The oleodistillate form of
eus skin infections, and the necessity for systemic antibiotics the sunflower seed oil is believed to strengthen the skin bar-
is highly colonized skin patients. Diluted bleach baths require rier [103]. Sunflower seed oil was shown to be equivalent to
soaking the patient in a tub full of lukewarm water for Aquaphor in decreasing child mortality and sepsis levels by
around ten minutes, combined with 60–120 ml of chlorine growing the skin barrier recovery [104].
bleach. The patient is then exhaustively washed with fresh Olive oil: It was found to be toxic to the skin barrier, while
water and immediate application of a moisturizer or emolli- sunflower oleodistillate maintained the integrity of stratum
ent to avoid dehydration and dryness of the skin. Several corneum, did not lead to erythema and increased skin hydra-
authors have suggested diluted bleach baths for three tion than the olive oil [105].
months for two times in a week [98]. Virgin coconut oil: It has demonstrated benefits in animal
models of both acute and chronic inflammation as an out-
standing emollient, natural antibacterial and anti-inflamma-
Anti-histamines tory agent [106]. A latest randomized double-blind study in
117 pediatric AD patients tested the impact of virgin coconut
Even though antihistamines of the first generation such as
oil on SCORAD index and transepidermal water loss for
chlorpheniramine, hydroxyzine, diphenhydramine do not spe-
eight-weeks as compared to mineral oil. Mean SCORAD indi-
cifically affect AD-related itching, the sedative effects of these
ces in the virgin coconut oil decreased from baseline to the
drugs have been shown to enhance sleep pattern in AD
mineral oil, and it was found to be superior to mineral
patients. Furthermore, these drugs lessen rapid eye movement
oil [107].
sleep, diminish work performance, and impair learning, there-
Honey: It has customarily been used in the diagnosis of
fore are not regularly recommended for AD patients. They can
inflammatory skin disorders like psoriasis and AD. Fingleton
be recommended for the acute adjuvant care of serious AD
et al. performed an open-label, single-blind, randomized clin-
patients who have trouble sleeping or frequently scratch dur-
ical study of 15 adults who had bilateral lesions identified
ing sleep. Due to their sedative effects, chronic and daytime
with AD on their limbs [108]. Some patients were treated
use of first-generation antihistamines should be circumvented.
with Kanuka honey added topically. Honey’s clinical efficacy
Among AD patients with allergic causes, nonsedating second- was evaluated by measuring AD scores and severity of skin
generation antihistamines tend to be of moderate benefit, lesions. Honey has also been demonstrated to be therapeut-
and thus, a therapeutic trial of these agents could be sug- ically beneficial when used in adult patients with AD in con-
gested in some clinical circumstances [99]. The histamine 4 junction with beeswax and olive oil [109].
receptor (H4R) antihistamines are other long-acting, oral, small Omega-3 fatty acid: Currently, it has been proposed that
molecules which represent an added auspicious treatment the increased frequency of AD could be linked with improve-
option for AD. In comparison to the H1R-blocking antihist- ments in the Western diet (decreased consumption of
amines traditionally used as antipruritic agents, the effects in omega-3 polyunsaturated fatty acids and improved intake of
patients with AD are primarily based on their sedative proper- omega-3 polyunsaturated fatty acids. A pilot study found
ties. H4R antihistamines have effects related to the pathogen- that dietary consumption of omega-3 polyunsaturated fatty
esis of AD. H4R is vastly expressed on keratinocytes in AD acids may have a beneficial impact on the AD symp-
patients with lesional skin, and its activation encourages the toms [110].
proliferation of keratinocytes and prevents their differentiation, Probiotics: These are live microbial foods additives that are
impairs the skin barrier and causes pruritus [100,101]. ZPL- indigestible carbohydrates that promote the production of
3893787, an H4R antagonist, is an ongoing phase II clinical probiotic bacteria in the intestine. These act as an immuno-
trial (NCT02424253). modulator by acting on T-helper cell cytokine activation
[111]. Recently, Baquerizo et al., reported that probiotics and
prebiotics tend to be effective in decreasing AD incidence in
Adjuvant treatment
the infant [112]. Yet their role in treating AD remains debat-
Adjuvant therapy should be provided in situations where AD able. More studies are required to determine if, by modulat-
symptoms are unrestrained without sufficient ing immunological dysfunction of AD, these agents may play
basic treatment. a role in AD treatment.
116 D. S. MANDLIK AND S. K. MANDLIK

Adherence to treatment as a hurdle in AD adverse effects. In AD trials in which patients were given the
drug and decided to contribute, however, adherence was
Since AD is a long-lasting skin illness, treatment schedules
low, indicating that costs and worries about side effects are
that are often formed precisely for individual patients require
not the only significant restrictions to adherence in AD.
regular management. It can be a daunting hurdle to sustain
Other explanations include oblivion, confusing orders, and
strong adherence over long periods of time. It is assessed
difficulty of time for medication, corticosteroid phobia, or
that 30% to 40% of all drugs used for chronic conditions are
lack of knowledge of the disease or treatment [120].
not taken as prescribed [113]. Electronic surveillance of der-
Assistance between parents, disapproval of treatment,
matologic patients indicates that patients are frequently
inability to bathe, late bedtime, absence of social support,
poorly adherent to topical therapy and that their use of
concern about eczema, feelings of discrimination and per-
medication in care logs could be overestimated by patients
ceived seriousness of the disease were factors linked to
[114]. Numerous studies have demonstrated varying grades
adherence in a group of parents of children specifically with
of nonadherence directly to AD care. Primary nonadherence
AD [121]. Factors such as patient preferences, past experi-
defines the first stage of a patient’s nonadherence when
ence with interventions, and mode of drug administration
their original prescription at a pharmacy is not redeemed. If
can also influence adherence, as topical procedures may be
the patient does not adequately obey their care plan or use
more problematic for patients [122]. Even short-term use of
their medicine as prescribed, secondary nonadherence hap-
topical fluorescent test cream applications is difficult [123].
pens at home. In Denmark, the primary adherence activity of
Topical drug adherence can also be affected by the preferen-
322 patients with outpatient dermatology was examined by
ces of patients for particular formulations of vehicles
means of a national pharmacy registry, showing that pre-
to others.
scriptions were redeemed within four weeks of their appoint-
Although there are a multitude of treatments for AD and
ment for consultation. 31.8% of patients with AD did not
more are being developed, topical therapies for AD, such as
receive their medications within 4 weeks, which indicates
corticosteroids, remain the first-line treatment. Adherence to
that nearly one-third of such patients showed primary non-
these therapies can vary, with anxiety regarding the use of
adherence [115]. While the emphasis on enhancing second-
corticosteroids, also called as ‘steroid phobia,’ playing a role
ary non-adherence is often greater, primary nonadherence is
in the use of these drugs by patients. Despite their safety
just as significant and should not be discounted. It is not
and effectiveness in treating AD, patients often show fright
likely to be successful if the drug is not filled in.
and worry about topical steroid usage [124]. Topical cortico-
If the drugs are filled out by patients, secondary adher-
steroids may also be confused with anabolic steroids by
ence can still be poor. In research secondary adherence has
been assessed in which patients are given medication using patients or caregivers, causing more misunderstandings
electronic monitors. Sometimes, patients exaggerate their about the usage of these drugs. The social media may also
use of therapy. For 5 days of treatment with fluocinonide spread this steroid phobia, along with the popular miscon-
cream (0.1 percent), the median daily adherence was 40 per- ception that topical steroids may have the similar side effects
cent per electronic monitoring, whereas subjects reported as oral or systemic steroids [125]. In France, 80.7 percent of
100 percent adherence to drug diaries. On day 1 of treat- 208 patients or parents of AD patients reported fears of top-
ment, adherence was higher in six out of nine patients using ical steroids and 36 percent admitted that they did nonad-
their treatment regimen, while on day 5, only 1 out of 9 herence to their medication. 47.8 percent of those surveyed
patients was adherent [116]. Mean adherence to twice-daily were not aware of the adverse effects of specific drugs, but
desonide hydrogel decreased from 81 percent adherence on also showed fear of using these drugs. Fear of topical corti-
day 1 to 50 percent by day 27 over a 4-week period out of costeroids was substantially associated with the belief in 300
41 adult patients with mild to moderate AD [117]. Italian patients with AD that treatment benefits do not over-
At the end of 8 weeks of treatment, electronic monitoring weigh the drawbacks and the belief that TCS could be risky
of 26 pediatric AD patients showed that average adherence regardless of the specific side effect [126]. Likewise, 72.5 per-
to topical triamcinolone was 32 percent. At the date of the cent of the 200 AD patients surveyed in the UK expressed
initial visit, adherence was highest, then fell steadily, dou- concern about topical steroids, and 24 percent registered
bling before the date of the return visit and declining again failure to comply with their care because of these concerns
subsequently. In Japan, adherence was usually lower in AD [127]. The perceived thinning of the skin, systemic absorp-
patients compared to psoriasis or tinea patients [118]. In a tion, skin inflammation, nonspecific long-term effects and a
small pilot study of 25 patients, 92% of patients indicated negative influence on growth and development are the most
that they often failed to use their drugs, 88 percent stated common concerns among patients [128].
that they sometimes stopped their AD treatment when their
skin indications upgraded, and 33.3 percent described that
Role of natural products in the treatment of AD
when their skin signs deteriorated, they stopped their AD
treatment [119]. Natural products extracted from medicinal plants have been
There may be several explanations for the diminished used since ancient times for the management of various dis-
commitment to care seen in AD. For parents of children with eases. The earliest use in Mesopotamia of natural resources
skin disorders, the utmost common reasons identified for as medicine dates back to 2600 BC. Also, the ‘Ebers Papyrus’
nonadherence were the cost of drug and unease about records of over 700 drugs back in 1550 BC are well kept
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 117

[129]. Similarly, the Indian Ayurveda philosophy was intro- remedies. In Europe, the Middle East [135], Africa, India (Behl
duced after the first century BC [130]. The production of and Srivastava 2002), China, Japan, Australia, and the
drugs based on natural products is fraught with certain prob- Americas, herbal usage systems have evolved regionally.
lems, such as consistency, identification of bioactive substan- Ayurvedic herbs in India [136] and herb combinations, devel-
ces, difficulties in obtaining wild samples and unsuitability oped as part of traditional Chinese medicine (TCM) in China,
with high-performance screening of natural products [131]. are two well-known systems still in use [137]. For the follow-
To further establish the detailed mechanism of action of nat- ing reasons, the use of herbs has become resurgent in recent
ural products is a challenging task. The pharmaceutical firms years: the side effects of chemical medicines have become
have shifted their prime focus on new product innovations evident, there has been a demand for a return to nature,
to synthetic substances because of these challenges. Yet the natural remedies have become part of the green revolution,
results recorded from the recently launched synthetic medi- and there has been a return to organic produce. Herbal rem-
cines during the 1990s do not satisfy expectations [132]. edies are currently gaining popularity among patients and to
USFDA approval levels have been poor for such medicines. a lesser degree among doctors, including those for
Given these reasons, it has revived the trust in the develop- skin disorders.
ment of medicines depending on natural ingredients. For Natural drugs from plant origin are getting more accept-
drug development and study, many natural phytoconstitu- ance due to many benefits such as having fewer side effects,
ents used for the management of a number of diseases, improved patient tolerance, being comparatively less costly
including AD, are becoming important [133]. A significant and appropriate due to a long history of use. Besides herbal
number of herbs and phytoconstituents are used for the medicines, other diseases which are obstinate and incurable
treatment of chronic hepatic disorders worldwide due to in other medicine systems are treated rationally. Given the
budget effectiveness, high protection constraints, long-term profound side effects caused by topical steroids and oral
beneficial action and less side effects (Figure 6). antihistamines, herbal medicines and their derived phytocon-
For thousands of years, herbal medicine for skin condi- stituents are widely used for treating AD [138]. Therefore,
tions has been used. The great apes, and our biologically efforts have been made to identify safer and more powerful
close relatives, use herbal self-medication [134]. Regional pro- plant-derived compounds that can modulate AD’s patho-
duction of specific herbs and their uses, based on locally logical mechanism(s) such as antihistaminic effects, inhibition
available plants, and through the trade in ethnobotanical of Th2 responses and production of IgE [139]. The review

Figure 6. Mechanism of action of herbal plants and phytoconstituents in the management of AD.
118 D. S. MANDLIK AND S. K. MANDLIK

Table 8. The effect of various plants on AD with respect to their mechanism of action.
No. Plant name & Family Nature of Extract Inducing agent Mechanism of action References
1 Actinidia arguta Water DNCB Decrease inflammatory cells, lymphocyte, [140]
Actinidiaceae elevate Treg-related & Foxp3 expression
2 Amomum xanthioides Water DNCB Reduce immunoglobulin E (IgE), CCL-17, [141]
Zingiberaceae CCL22, pro-inflammatory cytokines levels
3 Angelicae Water DNCB Reduce pro-inflammatory cytokines levels, [142]
dahuricae Apiaceae decrease inflammatory cells, mast cells &
CD4þ cells
4 Artemisia argyi Ethanol DNCB Reduce IgE, histamine, pro-inflammatory [143]
Asteraceae cytokines; Inhibit IjBa & PI3K/Akt
phosphorylation
5 Artemisia argyi Methyl alcohol DNFB Inhibit the release of NO & PGE2 in [144]
Asteraceae macrophages, pro-inflammatory cytokines
6 Artemisia scoparia Butanol DNFB Inhibit TSLP, histamine, IgE, pro-inflammatory [145]
Asteraceae cytokines levels & suppress caspase-1
7 Bambusae caulis Ethanol D. farina Reduce TEWL, inhibit IgE, eosinophil, pro- [146]
Poaceae inflammatory cytokines levels
8 Bambusae caulis Bamboo stem 2,4-DNFB Suppression of IgE, IL-4, IL-13 & TNF-a levels [146]
Poaceae
9 Betula Aqueous Picryl chloride Suppresses T-helper 2 cell response & serum [147]
platyphylla Betulaceae IgE level
10 Broussonetia kazinoki Ethanol D. farina Reduce IgE, IL-4 & skin mast cells levels [148]
Moraceae
11 Calophyllum inophyllum Methanol DNCB Decrease in pro-inflammatory cytokines and [149]
Calophyllaceae scratch behavior
12 Cervus Nippon Water DNCB Reduce mast cells, CD4 þ T, downregulate IgE, [150]
Cervidae pro-inflammatory cytokines levels
13 Cinnamomum cassia Ethanol HDM antigen Reduce IgE, histamine, pro-inflammatory [151]
Lauraceae cytokines & TARC mRNA expressions
14 Cordyceps bassiana Butanol DNFB Inhibition of release of histamine, IgE, IL-4 & [152]
Cordycipitaceae IFN-c production
15 Cynanchum atratum Water DNCB Ameliorate total IgE; reduce mast cell & pro- [153]
Apocynaceae inflammatory cytokines levels
16 Dictamnus dasycarpus Methyl alcohol Oxazolone Reduction of pro-inflammatory cytokines [154]
Rutaceae
17 Forsythia suspense Ethanol HDM antigen Suppression of expression of chemokines, [155]
Oleaceae cytokines & adhesion molecules in
keratinocytes
18 Gardenia jasminoides Water DNCB Suppressed COX-2, TNF-a expression, NF-jB [156]
Rubiaceae activation, IgE level & pro-
inflammatory cytokines
19 Hypsizigus marmoreus Ethanol 1-chloro 2, 4, 6- Decrease in dermatitis score, thickness of [157]
Tricholomataceae trinitrobenzene epidermis & proinflammatory cytokines
expressions
20 Morus alba Ethanol Biostir-AD Decrease PGE2 & NO production; suppress IgE, [158]
Moraceae TARC & histamine levels
21 Panax ginseng Water TNCB Decrease IFN-c, TNF-a, IgE, substance P, TSLP [159]
Araliaceae & TNF-a levels
22 Perilla frutescens Water DNFB Alleviating expressions of MMP-9 and IL-31 [160]
23 Platycodon grandiflorum Water DNFB Downregulation of IgE levels through [161]
Campanulaceae modulation of TH1 & TH2 immune
responses, decrease in TH2-
mediated cytokines
24 Pseudostellaria Water DNCB Suppress mast cells, IgE, pro-inflammatory [162]
heterophylla cytokines, MAPKs & NF-jB expression
Caryophyllaceae
25 Pyrus ussuriensis Ethanol DNCB Reduce IgE, pro-inflammatory cytokines; [163]
Rosaceae Reduction of scratching tendency &
erythema; Improved skin hydration
26 Rheum tanguticum Ethanol Hapten Decrease WBC, IgE & 5-LOX levels [164]
Polygonaceae
27 Saussurea lappa Methanol Biostir-AD Downregulate MDC, IL-8, IgE, TARC, RANTES, [165]
Asteraceae histamine & TARC levels
28 Schizandra chinensis Water DNCB Inhibit histamine, IgE, IgM, mast cell, IL-4, IL- [166]
Schisandraceae 5 expression
29 Scutellariae radix Aqueous Environmental stimuli Decrease IL-5 expression [167]
Lamiaceae
30 Tribulus terrestris Ethanol Oxazolone Reduce transepidermal water loss (TEWL), [168]
Zygophyllaceae Orai-1 & TRPV3 activation, inhibit
inflammatory cell
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 119

Table 9. The effect of phytoconstituents on AD with respect to their mechanism of action.


No. Phytoconstituents Plant source Inducing agent Mechanism of action Reference
1 7, 8, 40 - Glycine max DNCB Decrease eosinophils count, lower pro- [169]
trihydroxyisoflavone inflammatory cytokines levels
2 Capsaicin Capsicum annuum DNFB Suppressed activation of CD4þ T cells & mast [170]
cells, expression of pro-
inflammatory cytokines
3 Chlorogenic acid Artemisia capilliaris D. farina Reduce histamine, NO & IgE levels [171]
4 Chrysophanol Rumex crispus DNFB Reduce histamine, IgE, TSLP, ICAM-1, IL-4, IL-6 [172]
Cordyceps militaris & TNF-a, MIP-2
5 Cordycepic acid Cordyceps sinensis DNFB Inhibit IL-4, INF-c, histamine & IgE [152]
6 Crocin Gardenia jasminoides D. farina Inhibit activation of IL-4, IL-13, [173]
STAT6 & NF-jB
7 Cupressuflavone Juniperus rigida OXA/DNCB Inhibit IL-4, IgE & b-hexosaminidase release [174]
8 Daidzein, Genistein Glycine max Merr D. farina Reduce IL-12, TNF-a, IL-4, PKC, MMP-9, IL-4, [175]
substance P, iNOS & p-IjB levels
9 Decursin Angelica sinensis DNCB Reduce pro-inflammatory cytokines, NF-jB, [176]
IgE, MAPKs levels
10 Ergosterol Poria cocos DNCB Increase aryl hydrocarbon receptor expression [177]
11 Forsythiaside, Phillyrin, Forsythia suspensa D. farina Reduce VCAM-1, ICAM-1, IgE, histamine, mast [155]
Phylligenin Pinoresinol cell, TARC, MDC, RANTES, IL-4 levels
12 Gallic acid Cortex Moutan OXA Suppress pro-inflammatory cytokine, CCL7 [178]
& CCL8
13 Geniposide Gardenia jasminoides D. farina Reduce INF-c, IgE, histamine, mast cells, IL-4, [179]
IL-6 & TNF-a levels
14 GinsenosideRh2 Panax ginseng DNCB Inhibit TARC, TAR, TSLP, IL-4, IL-10, IL-6, TNF- [180,181]
Ginsenoside Rg3 a & MAPKs
15 Gintonin Panax ginseng DNFB Inhibit microglial activation & reduce IL-4, IgE, [182]
histamine, ]INF-c levels
16 Indirubin Polygonum tinctorium DNFB Inhibit TSLP, IgE, IL-4, IL-6, caspase-1 & [183]
histamine level
17 Lancerin Polygala tenuifolia Trimellitic anhydride Reduce IgE & histamine levels, Inhibit [184]
degranulation of tryptase, HMC-1 cells
& PKA
18 Linolenic acid Coriandrum sativum DNCB Reduce Th-1 & Th2-mediated cytokines [185]
19 Liquiritigenin, Liquritin Coptidis Rhizoma DNFB Inhibit Substance P, MMP-9; reduce COX-2, [186]
Glycyrrhiza uralensis TNF-a, p-IjB, iNOS & levels
20 Momordin Kochia scoparia DNCB Reduce expression of NF-jB, IL-1b, TNF-a [187]
& MAPKs
21 Naringenin Citrus fruits, tomatoes, DNFB Suppressed IFN-c, serum IgE levels, Reduced [188]
cherries, grapefruit infiltration of infiltration of eosinophils,
mast cells
22 Naringin Drynaria fortune D. farina Decrease IgG, IgE, IL-4, IL-6 & TNF-a [189]
expressions
23 Oregonin Alnus japonica Biostir AD Decrease the eosinophil count; Reduce IgE, IL- [190]
4, IL-5, IL-13 levels
24 Polysaccharide Ericerus pela DNCB Decrease IgE & mast cell; Inhibit Th1, Th17 [191]
activation, IL-6, IL-17A & INF-c
25 Quercetin Dendrobium tosaense TNCB Upregulate INF-c; Downregulate IgG, IL-4 [192]
& IgE,
26 Resveratrol Skin of red grapes DNFB Inhibition of proinflammatory cytokines [193]
27 Rutin Broussonetia kazinoki D. farina Inhibit TARC, MDC & RANTES production; [194]
Reduce IgE, IL-4, IL-5, IL-13
28 Valencene Cyperus rotundus DNCB Inhibit TARC, GM-CSF & I-CAM levels; Reduce [195]
IgE, IL-1b, IL-6, IL-13 levels
29 Ziyuglycoside I Sanguisorba officinalis DNCB Reduce IgE & b-HEX levels [196]
30 b-pinene Alpinia intermedia HDM Inhibit mast cell & TLSP expression [197]

paper presents the study findings of the anti-inflammatory and their families. The elevated incidence of AD in develop-
role of the medicinal plants and phytoconstituents used in ing countries has been a major public health problem as it
the treatment of AD (Tables 8 and 9). Table 10 depict the substantially reduces the patient’s quality of life. It also leads
available marketed products (like cream, ointment, lotion) to hereditary allergy and asthma if not treated properly. AD
used in the management of atopic dermatitis. Table 11 illus- may be triggered by genetic conditions and can be affected
trate the list of patented drug along with mechanism of by environmental factors. Optimum skin care activities and
action and patent number for the treatment of AD. topical corticosteroids treatment remains the keystone of the
disease. Topical calcineurin inhibitors in suitable patients vul-
nerable to regular flare-ups are an efficient, alternative treat-
Conclusion
ment to topical corticosteroids. In severe cases, systemic
In conclusion, AD is a complex, multifactorial disorder with a immunosuppressive agents may also be considered for the
substantial effect on the quality of life of affected patients treatment of AD. Enhanced knowledge of physiology and
120 D. S. MANDLIK AND S. K. MANDLIK

immunoregulatory pathway can contribute to effective drug appear to be implemented as step-up medication in the pro-
discovery and management therapy reducing flares recur- tocols for the treatment of AD. Since the introduction of
rence and increased quality of life. Monoclonal antibodies dupilumab several other monoclonal antibodies is currently
being explored as possible therapeutic alternatives for AD,
Table 10. Marketed products for the treatment of AD [198]. albeit with conflicting outcomes. Also, detailed clinical trials
No. Brand name Generic name Dosage form are needed to establish a successful treatment regimen. As a
1 Cortizone Hydrocortisone Cream/Ointment chronic disease, long-term adherence, a long-term care bur-
2 Kenalog Triamcinolone Aerosol den for the patient, is expected for AD. By enhancing adher-
solution
3 Vanos Fluocinonide Cream ence to treatment regimens through instructional
4 Protopic Tacrolimus Ointment approaches, earlier follow-up visits and better contact with
5 Verdeso Desonide Foam patients and their caregivers, clinicians may enhance patient
6 Elidel Pimecrolimus Cream
7 Elocon Mometasone Cream outcomes. Natural herbal remedies and active constituents
8 Cutivate Fluticasone Cream extracted from plants have been commonly used for the pre-
9 Diprolene Betamethasone Cream
dipropionate
vention, remedy or diagnosis of different forms of acute,
10 Embeline and Temovate/Clobex Clobetasol Cream/Lotion chronic moderate to serious atopic diseases for several deca-
11 Topicort and Topicort LP Desoximetasone Cream des. Literature survey has reported that the usage of natural
12 No brand Fluocinolone Cream
herbal medicinal in the management of AD is safe, cost-

Table 11. List of patented drug for the treatment of AD [199].


No. Category Drug Mechanism of action Patent No.
1 GPR142 agonists N-quinolylbenzamide Filaggrin production promoters WO2014017319
2 GPR142 agonists phenylalanine amide derivatives Filaggrin production promoters WO2014017319
3 Tomethamol-based pseudoceramide Pseudoceramides Accelerated barrier repair WO2014084676
4 Ceramide and sphingosine kinase inhibitor Guanidino- sphingosine or ceramide Inhibitory effects on ceramidases & sphingosine kinases US20120035268
5 PDE4/PDE7 inhibitors Pyrazolol [3,4-b] pyridnes Suppress release of inflammatory mediators US8420666
6 PDE4/PDE7 inhibitors 1-oxa-2-azaspirocycles Suppress release of inflammatory mediators US20110021473
7 PDE4 inhibitors Isoxazoline spirocycles Suppress release of inflammatory mediators US20090048247
8 PDE4 inhibitors Benzo [4, 5] furo [3,2-c] pyridines Suppress release of inflammatory mediators US7943634
9 PDE4 inhibitors Dibenzofuranederivatives Suppress release of inflammatory mediators WO2009115874
10 CRTH2 inhibitors Phenoxy acetic acid derivatives Anti-inflammatory activity WO2010092043
11 CRTH2 inhibitors Indol-1-yl acetic acid derivatives Anti-inflammatory activity WO2011006936
12 CRTH2 inhibitors Indol-1-yl acetic acid derivatives Anti-inflammatory activity EP2492268
13 CRTH2 inhibitors OC000459 (Indol-1-yl acetic acid) Anti-inflammatory activity US20110124683
US20130052190
14 CRTH2 inhibitors Indol-1-yl acetic acid derivatives Anti-inflammatory activity US20140039012
15 CRTH2 inhibitors Quinazoline derivatives Anti-inflammatory activity WO201205036
16 CRTH2 inhibitors Pyrazol-4-yl acetic acid derivatives Anti-inflammatory activity WO2012101043
17 CRTH2 inhibitors Pyrazol-4-yl acetic acid derivatives Anti-inflammatory activity WO2012069175
18 CRTH2 inhibitors Pyrimidine-5-yl acetic acid derivatives Anti-inflammatory activity WO2012013566
19 CCR3 antagonists Benzenesulfonamides Anti-inflammatory activity US20140005176
20 CCR3 antagonists Arylsulfonamides Anti-inflammatory activity US20100204213
21 CCR3 antagonists Arylsulfonamides Anti-inflammatory activity NZ620092
22 CCR3 antagonists 2,5-disubstituted arylsulfonamides Anti-inflammatory activity NZ620048
23 CCR2 antagonists Piperazinyl 3-aminopyrrolidines Anti-inflammatory activity US8575173
24 CCR5 antagonists Bipiperidinyl derivatives Anti-inflammatory activity US7652142
25 CCR2 and CCR5 agonists 3-aminocyclopentanecarboxamides Anti-inflammatory activity US8293903
26 CCR2 modulators 3-aminocyclopentane derivatives Anti-inflammatory activity US20100234409
27 CCR3 antagonists Substituted piperazines Anti-inflammatory activity WO2009020534
28 Chymase inhibitors Imidazopyridines Anti-inflammatory and anti-pruritic activity US20120094944
29 Chymase inhibitors Diazepane derivatives Anti-inflammatory and anti-pruritic activity US8846660
30 TLR7 agonists 2, 4-diaminopyrimidines Immune response modifiers WO2009067081
31 TLR7 agonists Pidotimod Immunomodulators WO2014094839
32 Raft modulator Miltefosine Anti-inflammatory activity US20110263531
33 Vitamin D receptor antagonist ZK 168281 US2010144683
34 SYK inhibitors Pyrimidine-5-carboxamides Anti-inflammatory US2015232470
35 TRPV1 channel agonist Palvanil Antipruritic US20140066399
36 TRPM8 agonist Menthoxypropanediol Antipruritic WO2014060150
37 Opioid antagonist Nalmefene salts Antipruritic US20090093509
38 5-HT7 receptor antagonist SB-269970 Antipruritic WO2013154513
39 H4 receptor antagonist Tricyclic heterocycles Antipruritic US20140315888
40 H1, H2 and H4 receptor antagonist Alcaftadin Antipruritic WO2014130526
GPR142 agonists: Agonists of G protein-coupled receptor 142; CRTH2: Chemoattractant receptor-homologous molecule expressed on Th2 cells; PDE4:
Phosphodiesterase-4; PDE7: Phosphodiesterase-7; TLR7: Toll-like receptor 7; SYK: spleen tyrosine kinase; TRPV1: Transient receptor potential cation channel sub-
family V member 1; TRPM8: Transient receptor potential cation channel subfamily M (melastatin) member 8; 5HT7: 5-hydroxytryptamine 7; H1 receptor:
Histamine 1 receptor; H2: Histamine 2 receptor; H4: Histamine 4 receptor.
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 121

efficient, systemically nontoxic, patient-compliant and effi- [20] Proksch E, Fo€lster-Holst R, Jensen JM. Skin barrier function, epi-
cient following traditional therapeutic protocols. dermal proliferation and differentiation in eczema. J Dermatol
Sci. 2006;43:159–169.
[21] Tamari M, Hirota T. Genome-wide association studies of atopic
dermatitis. J Dermatol 2014;41:213–220.
Declaration
[22] Imokawa G. Ceramides as natural moisturizing factors and their
The work is original and has not been submitted for publica- efficacy in dry skin. Cosmetic Sci Technol Series. 2002;267–302
[23] Moffatt MF. SPINK5: a gene for atopic dermatitis and asthma.
tion elsewhere in the journal.
Clin Exp Allergy. 2004;34:325–327.
[24] Bieber T. Atopic dermatitis. N Engl J Med 2008;358:1483–1494.
[25] Male D, Roitt I. Introduction to the immune system.
Disclosure statement Immunology. 5th ed. London: Mosby; 1998, 1–12.
No potential conflict of interest was reported by the author(s). [26] Tay SS, Roediger B, Tong PL, et al. The skin-resident immune
network. Curr Dermatol Rep. 2013;3:13–22.
[27] Peng W, Novak N. Pathogenesis of atopic dermatitis. Clin Exp
Allergy. 2015;45:566–574.
References €pf E, et al. A role for Th1
[28] Grewe M, Bruijnzeel-Koomen CA, Scho
[1] Kramer ON, Strom MA, Ladizinski B, et al. The history of atopic and Th2 cells in the immunopathogenesis of atopic dermatitis.
dermatitis. Clin Dermatol. 2017;35: 344–348. Immunol Today. 1998;19:359–361.
[2] Wollenberg A, Oranje A, Deleuran M, et al. ETFAD/EADV Eczema [29] Kaesler S, Volz T, Skabytska Y, et al. Toll-like receptor 2 ligands
task force 2015 position paper on diagnosis and treatment of promote chronic atopic dermatitis through IL-4- mediated sup-
atopic dermatitis in adult and paediatric patients. J Eur Acad pression of IL-10. J Allergy Clin Immunol. 2014;134:92–99.
Dermatol Venereol. 2016;30: 729–747. [30] Hamid Q, Naseer T, Minshall EM, et al. In vivo expression of IL-
[3] Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for 12 and IL-13 in atopic dermatitis. J Allergy Clin Immunol. 1996;
the management of atopic dermatitis: section 1. Diagnosis and 98: 225–231.
assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70: [31] Takai T, Ikeda S. Barrier dysfunction caused by environmental
338–351. proteases in the pathogenesis of allergic diseases. Allergol Int.
[4] Novak N, Bieber T, Leung DYM. Immune mechanisms leading to 2011;60:25–35.
atopic dermatitis. J Allergy Clin Immunol. 2003;112:S128–S139. [32] Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin
[5] Wollenberg A, Barbarot S, Bieber T, et al. Consensus-based microbiome associated with disease flares and treatment in chil-
European guidelines for treatment of atopic eczema (atopic dren with atopic dermatitis. Genome Res. 2012;22:850–859.
dermatitis) in adults and children: part I. J Eur Acad Dermatol [33] Kobayashi T, Glatz M, Horiuchi K, et al. Dysbiosis and
Venereol. 2018;32: 657–682. Staphylococcus aureus colonization drives inflammation in atopic
[6] Nomura T, Kabashima K. Advances in atopic dermatitis in 2015. dermatitis. Immunity. 2015;42:756–766.
J Allergy Clin Immunol. 2016;138: 1548–1555. [34] Cork MJ, Danby SG, Vasilopoulos Y, et al. Epidermal barrier dys-
[7] Spergel JM, Paller AS. Atopic dermatitis and the atopic march. J function in atopic dermatitis. J Invest Dermatol. 2009;129:
Allergy Clin Immunol. 2003;112: S128–S139. 1892–1908.
[8] Tsakok T, Marrs T, Mohsin M, et al. Does atopic dermatitis cause [35] Cork MJ, Robinson DA, Vasilopoulos Y, et al. New perspectives
food allergy? A systematic review. J Allergy Clin Immunol. 2016; on epidermal barrier dysfunction in atopic dermatitis: gene-
137: 1071–1078. environment interactions. J Allergy Clin Immunol. 2006;118:
[9] Weidinger S, Novak N. Atopic dermatitis. Lancet. 2016;387: 3–21.
1109–1122. [36] Silverberg JI, Hanifin J, Simpson EL. Climatic factors are associ-
[10] Thomsen SF, Ulrik CS, Kyvik KO, et al. Importance of genetic fac- ated with childhood eczema prevalence in the United States. J
tors in the etiology of atopic dermatitis: a twin study. Allergy & Invest Dermatol. 2013;133:1752–1759.
Asthma Proc. 2007;28: 535–539. [37] Murray AB, Morrison BJ. It is children with atopic dermatitis who
[11] Palmer CN, Irvine AD, Terron-Kwiatkowski A, et al. Common develop asthma more frequently if the mother smokes. J Allergy
loss-of-function variants of the epidermal barrier protein filag- Clin Immunol. 1990;86(5):732–739.
grin are a major predisposing factor for atopic dermatitis. Nat [38] Gu H, Chen XS, Chen K, et al. Evaluation of diagnostic criteria
Genet. 2006;38: 441–446. for atopic dermatitis: validity of the criteria of Williams et al in a
[12] Irvine AD, McLean WHI, Leung DYM. Filaggrin mutations associ- hospital-based setting. Br J Dermatol. 2001;145:428–433.
ated with skin and allergic diseases. N Engl J Med. 2011;365: [39] Xu S, Immaneni S, Hazen GB, et al. Cost effectiveness of prophy-
1315–1327. lactic moisturization for atopic dermatitis. JAMA Pediatr. 2017;
[13] Douwes J, Pearce N. Asthma and the westernization ‘package’. 171: e163909.
Int J Epidemiol. 2002;31:1098–1102. [40] Krakowski AC, Eichenfield LF, Dohil MA. Management of atopic
[14] Strachan DP. Hay fever, hygiene, and household size. Brit Med J. dermatitis in the pediatric population. Pediatrics. 2008;122:
1989;299:1259–1260. 812–824.
[15] Bach JF. The effect of infections on susceptibility to auto- [41] Lee JH, Son SW, Cho SH. A comprehensive review of the treat-
immune and allergic diseases. N Engl J Med. 2002;347:911–920. ment of atopic eczema. Allergy Asthma Immunol Res. 2016;8:
[16] von Mutius E. Maternal farm exposure/ingestion of unpasteur- 181–190.
ized cow’s milk and allergic disease. Curr Opin Gastroenterol. [42] Gittler JK, Wang JF, Orlow SJ. Bathing and associated treatments
2012;28:570–576. in atopic dermatitis. Am J Clin Dermatol. 2017;18: 45–57.
[17] Hong S, Choi WJ, Kwon HJ, et al. Effect of prolonged breast- [43] Leung DYM, Bieber T. Atopic dermatitis. Lancet. 2003;361:
feeding on risk of atopic dermatitis in early childhood. Allergy 151–160.
Asthma Proc. 2014;35:66–70. [44] Wong SM, Ng TG, Baba R. Efficacy and safety of sodium hypo-
[18] Hammer-Helmich L, Linneberg A, Thomsen SF, et al. Association chlorite (bleach) baths in patients with moderate to severe
between parental socioeconomic position and prevalence of atopic dermatitis in Malaysia. J Dermatol. 2013;40:874–880.
asthma, atopic eczema and hay fever in children. Scand J Public [45] Mandeau A, Aries MF, Bo e JF, et al. RhealbaV R oat plantlet

Health Suppl. 2014;42:120–127. extract: evidence of protein-free content and assessment of


[19] Egawa G, Weninger W. Pathogenesis of atopic dermatitis: A regulatory activity on immune inflammatory mediators. Planta
short review. Cogent Biol. 2015;1:1–10. Med. 2011;77:900–906.
122 D. S. MANDLIK AND S. K. MANDLIK

[46] Wollenberg A, Frank R, Kroth J, et al. Proactive therapy of atopic [66] Patrizi A, Savoia F, Giacomini F, et al. The effect of summer holi-
eczema–an evidence-based concept with a behavioral back- days and sun exposure on atopic dermatitis. G Ital Dermatol
ground. J Dtsch Dermatol Ges. 2009;7:117–121. Venereol. 2009;144: 463–466.
[47] Niedner R. Therapy with systemic glucocorticoids. Hautarzt. [67] Gambichler T, Kreuter A, Tomi NS, et al. Gene expression of
2001;52(11):1062–1071. cytokines in atopic eczema before and after ultraviolet A1
[48] Walsh P, Aeling JL, Huff L, et al. Hypothalamus pituitary- adrenal phototherapy. Br J Dermatol 2008;158: 1117–1120.
axis suppression by superpotent topical steroids. J Am Acad [68] Dotterud LK, Wilsgaard T, Vorland LH, et al. The effect of UVB
Dermatol. 1993;29:501–503. radiation on skin microbiota in patients with atopic dermatitis
[49] Hajar T, Leshem YA, Hanifin JM, et al. A systematic review of and healthy controls. Int J Circumpolar Health. 2008;67:
topical corticosteroid withdrawal (“steroid addiction”) in patients 254–260.
with atopic dermatitis and other dermatoses. J Am Acad [69] V€ah€avihu K, Ylianttila L, Salmelin R, et al. Heliotherapy improves
Dermatol. 2015;72:541–549. vitamin D balance and atopic dermatitis. Br J Dermatol. 2008;
[50] Haeck IM, Rouwen TJ, Timmer-de Mik L, et al. Topical corticoste- 158: 1323–1328.
roids in atopic dermatitis and the risk of glaucoma and cata- [70] Grabbe J, Welker P, Humke S, et al. High-dose ultraviolet A1
racts. J Am Acad Dermatol. 2011;64:275–281. (UVA1), but not UVA/UVB therapy, decreases IgE binding cells in
[51] Papier A, Strowd LC. Atopic dermatitis: a review of topical non- lesional skin of patients with atopic eczema. J Invest Dermatol.
steroid therapy. Drugs Context. 2018;7:212521. 1996;107: 419–422.
[52] Kyllo€nen H, Remitz A, Mandelin JM, et al. Effects of 1-year inter- [71] Becker D, Langer E, Seemann M, et al. Clinical efficacy of blue
mittent treatment with topical tacrolimus monotherapy on skin light full body irradiation as treatment option for severe atopic
collagen synthesis in patients with atopic dermatitis. Br J dermatitis. PLoS One. 2011;6: e20566.
Dermatol. 2004;150:1174–1181. [72] Wollenschl€ager I, Hermann J, Ockenfels HM. [Targeted UVB-
[53] Murrell DF, Calvieri S, Ortonne JP, et al. A randomized controlled 308 nm (NUVB) therapy with excimer laser in the treatment of
trial of pimecrolimus cream 1% in adolescents and adults with atopic dermatitis and other inflammatory dermatoses]. Hautarzt.
head and neck atopic dermatitis and intolerant of, or dependent 2009;60: 898–906.
on, topical corticosteroids. Br J Dermatol. 2007;157:954–959. [73] Syed S, Weibel L, Kennedy H, et al. A pilot study showing
[54] Legendre L, Barnetche T, Mazereeuw-Hautier J, et al. Risk of pulsed-dye laser treatment improves localized areas of chronic
lymphoma in patients with atopic dermatitis and the role of atopic dermatitis. Clin Exp Dermatol. 2008;33: 243–248.
topical treatment: A systematic review and meta-analysis. J Am [74] Kawakami T, Kaminishi K, Soma Y, et al. Oral antihistamine ther-
apy influences plasma tryptase levels in adult atopic dermatitis.
Acad Dermatol. 2015;72:992–1002.
[55] Eichenfield LF, Call RS, Forsha DW, et al. Long-term safety of cri- J Dermatol Sci. 2006;43: 127–134.
[75] Malekzad F, Arbabi M, Mohtasham N, et al. Efficacy of oral nal-
saborole ointment 2% in children and adults with mild to mod-
trexone on pruritus in atopic eczema: a double blind, placebo-
erate atopic dermatitis. J Am Acad Dermatol. 2017;77:641–649.
controlled study. J Eur Acad Dermatol Venereol. 2009;23:
[56] Ahmed A, Solman L, Williams HC. Magnitude of benefit for top-
948–950.
ical crisaborole in the treatment of atopic dermatitis in children
[76] St€ander S, Bo
€ckenholt B, Sch€ urmeyer-Horst F, et al. Treatment of
and adults does not look promising: a critical appraisal. Br J
chronic pruritus with the selective serotonin re-uptake inhibitors
Dermatol. 2018;178:659–662.
paroxetine and fluvoxamine: results of an open-labelled, two-
[57] Peppers J, Paller AS, Maeda-Chubachi T, et al. A phase 2,
arm proof-of-concept study. Acta Derm Venereol. 2009;89:
randomized dose-finding study of tapinar of (GSK2894512
45–51.
cream) for the treatment of atopic dermatitis. J Am Acad
[77] Sowden JM, Berth-Jones J, Ross JS, et al. Double-blind, con-
Dermatol. 2019;80:89–98.
trolled, crossover study of cyclosporin in adults with severe
[58] SP14019-Cyclatop. Pilot study to assess efficacy and safety of
refractory atopic dermatitis. Lancet. 1991;338: 137–140.
5% topical cyclosporine A in atopic dermatitis. In: 27th Congress
[78] Grundmann-Kollmann M, Podda M, Ochsendorf F, et al.
of the European Academy of Dermatology and Venereology. Mycophenolate mofetil is effective in the treatment of atopic
2018, 12–16. dermatitis. Arch Dermatol. 2001;137: 870–873.
[59] Vakharia PP, Silverberg JI. New therapies for atopic dermatitis: [79] Simpson EL, Bieber T, Guttman-Yassky E, et al. Two phase 3 tri-
additional treatment classes. J Am Acad Dermatol. 2018;78: als of dupilumab versus placebo in atopic dermatitis. N Engl J
S76–S83. Med. 2016;375: 2335–2348.
[60] Hajar T, Gontijo JRV, Hanifin JM. New and developing therapies [80] Hamilton JD, Suarez-Farin ~as M, Dhingra N, et al. Dupilumab
for atopic dermatitis. An Bras Dermatol. 2018;93: 104–107. improves the molecular signature in skin of patients with mod-
[61] Bissonnette R, Papp KA, Poulin Y, et al. Topical tofacitinib for erate-to-severe atopic dermatitis. J Allergy Clin Immunol. 2014;
atopic dermatitis: a phase IIa randomized trial. Br J Dermatol. 134(6): 1293–1300.
2016;175: 902–911. [81] Kim BE, Leung DY, Boguniewicz M, et al. Loricrin and involucrin
[62] He H, Guttman-Yassky E. JAK inhibitors for atopic dermatitis: an expression is down-regulated by Th2 cytokines through STAT-6.
update. Am J Clin Dermatol. 2019;20: 181–192. Clin Immunol. 2008;126: 332–337.
[63] Nakagawa H, Nemoto O, Igarashi A, et al. Efficacy and safety of [82] Eichenfield L, Flohr C, Simpson E, et al. Lebrikizumab improves
topical JTE-052, a Janus kinase inhibitor, in Japanese adult patient-reported outcomes (PROs) in a phase 2 study in patients
patients with moderate-to-severe atopic dermatitis: a phase II, with atopic dermatitis. J Am Acad Dermatol. 2017;76:AB423.
multicentre, randomized, vehicle-controlled clinical study. Br J [83] Samrao A, Berry TM, Goreshi R, et al. A pilot study of an oral
Dermatol. 2018;178: 424–432. phosphodiesterase inhibitor (apremilast) for atopic dermatitis in
[64] Guttman-Yassky E, Silverberg JI, Nemoto O, et al. Baricitinib in adults. Arch Dermatol. 2012;148:890–897.
adult patients with moderate-to-severe atopic dermatitis: a [84] Abrouk M, Farahnik B, Zhu TH, et al. Apremilast treatment of
phase 2 parallel, double-blinded, randomized placebo-controlled atopic dermatitis and other chronic eczematous dermatoses. J
multiple-dose study. J Am Acad Dermatol. 2019;80: 913–921. Am Acad Dermatol. 2017;77:177–180.
[65] Bissonnette R, Maari C, Forman S, et al. The oral Janus kinase/ [85] Ruzicka T, Hanifin JM, Furue M, et al. Anti-interleukin-31 recep-
spleen tyrosine kinase inhibitorASN002 demonstrates efficacy tor A antibody for atopic dermatitis. N Engl J Med. 2017;376:
and improves associated systemic inflammation in patients with 826–835.
moderate-to-severe atopic dermatitis: results from a randomized [86] Holm JG, Agner T, Sand C, et al. Omalizumab for atopic derma-
double-blind placebo-controlled study. Br J Dermatol. 2019;181: titis: case series and systematic review of the literature. Int J
733–742. Dermatol. 2016;1:1–9.
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 123

[87] Godse K, Mehta A, Patil S, et al. Omalizumab- A Review. Indian J [108] Fingleton J, Sheahan D, Corin A, et al. A randomised controlled
Dermatol. 2015;60: 381–384. trial of topical Kanuka honey for the treatment of psoriasis.
[88] Holm JG, Thomsen SF. Omalizumab for atopic dermatitis: evi- JRSM Open. 2014;5: 2042533313518913.
dence for and against its use. In: G Ital Deramtol Venereol. 2019; [109] Al-Waili NS. Topical application of natural honey, beeswax and
154:480–487. olive oil mixture for atopic dermatitis or psoriasis: partially con-
[89] Simon D, Ho €sli S, Kostylina G, et al. Anti-CD20 (rituximab) treat- trolled, single-blinded study. Complement Ther Med. 2003;11:
ment improves atopic eczema. J Allergy Clin Immunol. 2008;121: 226–234.
122–128. [110] Koch C, Do €lle S, Metzger M, et al. Docosahexaenoic acid (DHA)
[90] Wenzel S, Wilbraham D, Fuller R, et al. Effect of an interleukin-4 supplementation in atopic eczema: a randomized, double-blind,
variant on late phase asthmatic response to allergen challenge controlled trial. Br J Dermatol. 2008;158: 786–792.
in asthmatic patients: results of two phase 2a studies. Lancet. [111] Sudo N, Sawamura S, Tanaka K, et al. The requirement of intes-
2007;370:1422–1431. tinal bacterial flora for the development of an IgE production
[91] Jacobi A, Antoni C, Manger B, et al. Infliximab in the treatment system fully susceptible to oral tolerance induction. J Immunol.
of moderate to severe atopic dermatitis. J Am Acad Dermatol. 1997;159: 1739–1745.
2005;52: 522–526. [112] Baquerizo Nole KL, Yim E, Keri JE. Probiotics and prebiotics in
[92] Oldhoff JM, Darsow U, Werfel T, et al. Anti-IL-5 recombinant dermatology. J Am Acad Dermatol. 2014;71: 814–821.
humanized monoclonal antibody (mepolizumab) for the treat- [113] Richards HL, Fortune DG, O’Sullivan TM, Main CJ, Griffiths CE.
ment of atopic dermatitis. Allergy. 2005;60:693–696. Patients with psoriasis and their compliance with medication. J
[93] Bel EH, Wenzel SE, Thompson PJ, et al. Oral glucocorticoid-spar- Am Acad Dermatol. 1999;41(4):581–583.
ing effect of mepolizumab in eosinophilic asthma. N Engl J [114] Carroll CL, Feldman SR, Camacho FT, Manuel JC, Balkrishnan R.
Med. 2014;371:1189–1197. Adherence to topical therapy decreases during the course of an
[94] Navarini AA, French LE, Hofbauer GF. Interrupting IL-6-receptor 8-week psoriasis clinical trial: commonly used methods of meas-
signaling improves atopic dermatitis but associates with bacter- uring adherence to topical therapy overestimate actual use. J
ial superinfection. J Allergy Clin Immunol. 2011;128:1128–1130. Am Acad Dermatol. 2004;51(2):212–216.
[95] Gauvreau GM, O’Byrne PM, Boulet LP, et al. Effects of an anti- [115] Storm A, Andersen SE, Benfeldt E, Serup J. One in 3 prescrip-
TSLP antibody on allergen-induced asthmatic responses. N Engl tions are never redeemed: primary nonadherence in an out-
J Med. 2014;370:2102–2110. patient clinic. J Am Acad Dermatol. 2008;59(1):27–33.
[96] www.clinicaltrials.gov [116] Hix E, Gustafson CJ, O’Neill JL, et al. Adherence to a five day
[97] Akdis CA, Akdis M, Bieber T, et al.; European Academy of treatment course of topical fluocinonide 0.1% cream in atopic
Allergology and Clinical Immunology/American Academy of dermatitis. Dermatol Online J. 2013;19(10):20029.
Allergy, Asthma and Immunology. Diagnosis and treatment of [117] Yentzer BA, Camacho FT, Young T, Fountain JM, Clark AR,
atopic dermatitis in children and adults: European Academy of Feldman SR. Good adherence and early efficacy using desonide
Allergology and Clinical Immunology/American Academy of hydrogel for atopic dermatitis: results from a program address-
Allergy, Asthma and Immunology/PRACTALL Consensus Report. ing patient compliance. J Drugs Dermatol. 2010; 9(4):324–329.
J Allergy Clin Immunol. 2006;118: 152–169. [118] Furue M, Onozuka D, Takeuchi S, et al. Poor adherence to oral
[98] Huang JT, Abrams M, Tlougan B, et al. Treatment of and topical medication in 3096 dermatological patients as
Staphylococcus aureus colonization in atopic dermatitis assessed by the Morisky Medication Adherence Scale-8. Br J
decreases disease severity. Pediatrics. 2009;123: e808–e814. Dermatol. 2015; 172(1):272–275.
[99] Church MK, Maurer M, Simons FE, et al.; Global Allergy and [119] Pena-Robichaux V, Kvedar JC, Watson AJ. Text messages as a
Asthma European Network. Risk of first-generation H(1)-antihist- reminder aid and educational tool in adults and adolescents
amines: a GA(2)LEN position paper. Allergy. 2010;65: 459–466. with atopic dermatitis: a pilot study. Dermatol Res Pract. 2010;
[100] Glatzer F, Gschwandtner M, Ehling S, et al. Histamine induces 2010;894258.
proliferation in keratinocytes from patients with atopic derma- [120] Ellis RM, Koch LH, McGuire E, Williams JV. Potential barriers to
titis through the histamine 4 receptor. J Allergy Clin Immunol. adherence in pediatric dermatology. Pediatr Dermatol. 2011;
2013;132(6): 1358–1367. 28(3):242–244.
[101] De Benedetto A, Yoshida T, Fridy S, et al. Histamine and skin [121] Ohya Y, Williams H, Steptoe A, et al. Psychosocial factors and
barrier: are histamine antagonists useful for the prevention or adherence to treatment advice in childhood atopic dermatitis. J
treatment of atopic dermatitis? J Clin Med. 2015;4(4):741–755. Invest Dermatol. 2001;117(4):852–857.
[102] Bamford JT, Ray S, Musekiwa A, et al. Oral evening primrose oil [122] Serup J, Lindblad AK, Maroti M, et al. To follow or not to follow
and borage oil for eczema. Cochrane Database Syst Rev. 2013;4: dermatological treatment. Acta Derm Venereol. 2006;86(3):
CD004416. 193–197.
[103] Eichenfield LF, McCollum A, Msika P. The benefits of sunflower [123] Ulff E, Maroti M, Kettis-Lindblad Å, et al. Single application of a
oleodistillate (SOD) in pediatric dermatology. Pediatr Dermatol. fluorescent test cream by healthy volunteers: assessment of
2009;26: 669–675. treated and neglected body sites. Br J Dermatol. 2007;156(5):
[104] Darmstadt GL, Saha SK, Ahmed AN, et al. Effect of skin barrier 974–978.
therapy on neonatal mortality rates in preterm infants in [124] Aubert-Wastiaux H, Moret L, Le Rhun A, et al. Topical cortico-
Bangladesh: a randomized, controlled, clinical trial. Pediatrics. steroid phobia in atopic dermatitis: a study of its nature, origins
2008;121: 522–529. and frequency. Br J Dermatol. 2011; 165(4):808–814.
[105] Danby SG, AlEnezi T, Sultan A, et al. Effect of olive and sun- [125] Hon K-LE, Kam W-YC, Leung T-F, et al. Steroid fears in children
flower seed oil on the adult skin barrier: implications for neo- with eczema. Acta Paediatr. 2006; 95(11):1451–1455.
natal skin care. Pediatr Dermatol. 2013;30: 42–50. [126] El Hachem M, Gesualdo F, Ricci G, et al. Topical corticosteroid
[106] Verallo-Rowell VM, Dillague KM, Syah-Tjundawan BS. Novel anti- phobia in parents of pediatric patients with atopic dermatitis: a
bacterial and emollient effects of coconut and virgin olive oils multicentre survey. Ital J Pediatr. 2017; 43(1):22.
in adult atopic dermatitis. Dermatitis. 2008;19: 308–315. [127] Charman CR, Morris AD, Williams HC. Topical corticosteroid pho-
[107] Evangelista MTP, Abad-Casintahan F, Lopez-Villafuerte L. The bia in patients with atopic eczema. Br J Dermatol. 2000;142(5):
effect of topical virgin coconut oil on SCORAD index, transepi- 931–936.
dermal water loss, and skin capacitance in mild to moderate [128] Lee JY, Her Y, Kim CW, Kim SS. Topical corticosteroid phobia
pediatric atopic dermatitis: a randomized, double blind, clinical among parents of children with atopic eczema in Korea. Ann
trial. Int J Dermatol. 2014;53: 100–108. Dermatol. 2015; 27(5):499–506.
124 D. S. MANDLIK AND S. K. MANDLIK

[129] Cragg GM, Newman DJ. Natural products: a continuing source induced atopic dermatitis in BALB/c mice and inflammatory
of novel drug leads. Biochim Biophys Acta. 2013;1830(6): effects in mast cells. Int J Mol Med. 2018;42:2961–2971.
3670–3695. [151] Sung YY, Yoon T, Jang JY, et al. Inhibitory effects of
[130] Parasuraman S, Thing GS, Dhanaraj SA. Polyherbal formulation: Cinnamomum cassia extract on atopic dermatitis-like skin lesions
concept of ayurveda. Pharmacog Rev. 2014;8(16):73–80. induced by mite antigen in NC/Nga mice. J Ethnopharmacol.
[131] Atanasov AG, Waltenberger B, Pferschy-Wenzig E-M, Linder T, 2011;133:621–628.
Wawrosch C, Uhrin P, Temml V, Wang L, Schwaiger S, Heiss EH, [152] Wu G, Li L, Sung GH, et al. Inhibition of 2, 4-dinitrofluoroben-
et al. Discovery and resupply of pharmacologically active plant- zene-induced atopic dermatitis by topical application of the
derived natural products: a review. Biotechnol Adv. 2015;33(8): butanol extract of Cordyceps bassiana in NC/Nga mice. J
1582–1614. Ethnopharmacol. 2011;134:504–509.
[132] Scannell JW, Blanckley A, Boldon H, Warrington B. Diagnosing [153] Choi YY, Kim MH, Lee H, et al. Cynanchum atratum inhibits the
the decline in pharmaceutical R&D efficiency. Nat Rev Drug development of atopic dermatitis in 2,4-dinitrochlorobenzene-
Discov. 2012;11(3):191–200. induced mice. Biomed Pharmacother. 2017;90:321–327.
[133] Saha S, Sadhukhan P, Mahalanobish S, Dutta S, Sil PC. [154] Yang B, Lyu JH, Kim S, et al. Dictamnus dasycarpus Turcz., root
Ameliorative role of genistein against age dependent chronic bark alleviates oxazolone-induced atopy-like dermatitis in mice.
arsenic toxicity in murine brains via the regulation of oxidative Pharmacog Mag. 2019;15:219–255.
stress and inflammatory signaling cascades. J Nutritional [155] Sung YY, Yoon T, Jang S, et al. Forsythia suspensa suppresses
Biochem. 2018;55:26–40. house dust mite extract-induced atopic dermatitis in NC/Nga
[134] Huffman MA. Self-medicative behavior in the African great apes: mice. PLoS One. 2016;11:e0167687.
An evolutionary perspective into the origins of human trad- [156] Debnath T, Lee YM, Lim JH, et al. Anti-allergic and anti-atopic
itional medicine. Bioscience. 2001;51(8):651–661.2.0.CO;2] dermatitis effects of Gardenia Fructus extract. Food Agr
[135] Ghazanfar SA. Handbook of Arabian medicinal plants. Boca Immunol. 2018;29:665–674.
Raton, FL: CRC Press; 1994. [157] Kim T, Park K, Jung HS, et al. Evaluation of anti-atopic dermatitis
[136] Kapoor LD. CRC handbook of Ayurvedic medicinal plants. Boca activity of Hypsizigus marmoreus extract. Phytother Res. 2014;28:
Raton, FL: CRC Press; 1990. 1539–1546.
[137] Xu Y. Dermatology in Traditional Chinese Medicine. St. Albans, [158] Lim HS, Ha H, Lee H, et al. Morus alba L. suppresses the devel-
UK: Donica Publishing Ltd.; 2004 opment of atopic dermatitis induced by the house dust mite in
[138] Wollenberg A, Reitamo S, Atzori F, et al. Proactive treatment of
NC/Nga mice. BMC Complement Altern Med. 2014;14:1–8.
atopic dermatitis in adults with 0.1% tacrolimus ointment. [159] Lee JH, Cho SH. Korean red ginseng extract ameliorates skin
Allergy Eur J Allergy Clin Immunol. 2008;63:742–750.
lesions in NC/Nga mice: an atopic dermatitis model. J
[139] Leung DYM. Atopic dermatitis: new insights and opportunities
Ethnopharmacol. 2011;133:810–817.
for therapeutic intervention. J Allergy Clin Immunol. 2000;105:
[160] Heo JC, Nam DY, Seo MS, et al. Alleviation of atopic dermatitis-
860–876.
related symptoms by Perilla frutescens Britton. Int J Mol Med.
[140] Bae MJ, Lim S, Lee DS, et al. Water soluble extracts from
2011;28:733–737.
Actinidia arguta, PG102, attenuates house dust mite-induced
[161] Kim MS, Hur YG, Kim WG, et al. Inhibitory effect of Platycodon
murine atopic dermatitis by inhibiting the mTOR pathway with
grandiflorum on TH1 and TH2 immune responses in a murine
Treg generation. J Ethnopharmacol. 2016;193:96–106.
model of 2, 4-dinitrofluorobenzene–induced atopic dermatitis–-
[141] Choi Y, Choi JK, Jang YH, et al. Antiinflammatory effect of
like skin lesions. Ann Allergy Asthma Immunol. 2011;106:54–61.
Amomum xanthioides in a mouse atopic dermatitis model. Mol
[162] Choi YY, Kim MH, Ahn KS, et al. Immunomodulatory effects of
Med Rep. 2017;16:8964–8972.
Pseudostellaria heterophylla (Miquel) Pax on regulation of Th1/
[142] Ku JM, Hong SH, Kim HI, et al. Effects of Angelicae dahuricae
Th2 levels in mice with atopic dermatitis. Mol Med Rep. 2017;
Radix on 2, 4-dinitrochlorobenzene-induced atopic dermatitis-
15:649–656.
like skin lesions in mice model. BMC Complement Altern Med.
[163] Cho K, Parveen A, Kang MC, et al. Pyrus ussuriensis Maxim.
2017;17:1–8.
[143] Han HM, Kim SJ, Kim JS, et al. Ameliorative effects of Artemisia leaves extract ameliorates DNCB-induced atopic dermatitis-like
argyi Folium extract on 2, 4dinitrochlorobenzeneinduced atopic symptoms in NC/Nga mice. Phytomed. 2018;48:76–83.
[164] Jin JH, Ngoc TM, Bae K, et al. Inhibition of experimental atopic
dermatitislike lesions in BALB/c mice. Mol Med Rep. 2016;14:
3206–3214. dermatitis by rhubarb (rhizomes of Rheum tanguticum) and 5-
[144] Yun C, Jung Y, Chun W, et al. Anti-inflammatory effects of lipoxygenase inhibition of its major constituent, emodin.
Artemisia leaf extract in mice with contact dermatitis in vitro Phytother Res. 2011;25:755–759.
and in vivo. Mediat Inflamma. 2016;2016:8027537. [165] Lim HS, Ha H, Lee MY, et al. Saussurea lappa alleviates inflam-
[145] Ryu KJ, Yoou MS, Seo Y, et al. Therapeutic effects of Artemisia matory chemokine production in HaCaT cells and house dust
scoparia Waldst. et Kitaib in a murine model of atopic derma- mite-induced atopic-like dermatitis in Nc/Nga mice. Food Chem
titis. Clin Exp Dermatol. 2018;43:798–805. Toxicol. 2014;63:212–220.
[146] Qi XF, Kim DH, Yoon YS, et al. Effects of Bambusae caulis in [166] Kang TH, Shin HM. Inhibitory effects of Schizandra chinensis
Liquamen on the development of atopic dermatitis-like skin extract on atopic dermatitis in NC/Nga mice. Immunopharmacol
lesions in hairless mice. J Ethnopharmacol. 2009; 123: 195–200. Immunotoxicol. 2012;34;292–298.
[147] Kim EC, Lee HS, Kim SK, et al. The bark of Betula platyphylla var. [167] Kim J, Seok Lee I, Park S, et al. Effects of Scutellariae radix and
japonica inhibits the development of atopic dermatitis-like skin Aloe vera gel extracts on immunoglobulin E and cytokine levels
lesions in NC/Nga mice. J Ethnopharmacol. 2008;116:270–278. in atopic dermatitis NC/Nga mice. J Ethnopharmacol 2010;132:
[148] Lee JK, Ha H, Lee HY, et al. Inhibitory effects of heartwood 529–532.
extracts of Broussonetia kazinoki sieb on the development of [168] Kang SY, Jung HW, Nam JH, et al. Effects of the fruit extract of
atopic dermatitis in NC/Nga mice. Biosci Biotechnol Biochem. Tribulus terrestris on skin inflammation in mice with oxazolone-
2010;74:1802–1806. induced atopic dermatitis through regulation of calcium chan-
[149] Um YL, Jo YW. Inhibitory effects of calophyllum inophyllum nels, Orai-1 and TRPV3, and mast cell activation. Evid
extract on atopic dermatitis induced by DNCB in mouse. Am J Complement Alternat Med. 2017;2017:8312946.
Phytomed Clin Thera. 2016;4:165–173. [169] Kim H, Kim JR, Kang H, et al. 7, 8, 40 -Trihydroxyisoflavone
[150] Hong SH, Ku JM, Kim HI, et al. Oral administration of Cervus nip- attenuates DNCB-induced atopic dermatitis-like symptoms in
pon mantchuricus extract suppresses 2, 4-dinitrochlorobenzene- NC/Nga mice. PLoS One. 2014;9:e104938.
IMMUNOPHARMACOLOGY AND IMMUNOTOXICOLOGY 125

[170] Lee JH, Lee YS, Lee EJ, et al. Capsiate inhibits DNFB-induced modulation of protein Kinase A and p38 mitogen-activated pro-
atopic dermatitis in NC/Nga mice through mast cell and CD4þ tein Kinase signaling pathway. Int J Mol Sci. 2017;18:190.
T-cell inactivation. J Investig Dermatol. 2015;135:1977–1985. [185] Park G, Kim HG, Lim S, et al. Coriander alleviates 2, 4-dinitro-
[171] Ha H, Lee H, Seo CS, et al. Artemisia capillaris inhibits atopic chlorobenzene-induced contact dermatitis-like skin lesions in
dermatitis-like skin lesions in Dermatophagoides farinae-sensi- mice. J Med Food. 2014; 17: 862–868.
tized Nc/Nga mice, BMC Complement Altern Med. 2014; 14: 100. [186] Cha HY, Ahn SH, Cheon JH, et al. Hataedock treatment has pre-
[172] Han NR, Moon PD, Kim HM, et al. Cordycepin ameliorates skin ventive therapeutic effects in atopic dermatitis-induced NC/Nga
inflammation in a DNFB-challenged murine model of atopic mice under high-fat diet conditions. Evid Complement Alternat
dermatitis. Immunopharmacol Immunotoxicol. 2018;40:401–407. Med. 2016;2016:1739760.
[173] Sung YY, Kim HK. Crocin ameliorates atopic dermatitis symp- [187] Choi YY, Kim MH, Lee JY, et al. Topical application of Kochia sco-
toms by down regulation of Th2 response via blocking of NF- paria inhibits the development of contact dermatitis in mice. J
kappaB/STAT6 signaling pathways in mice. Nutrients. 2018; 10: Ethnopharmacol. 2014;154:380–385.
1625. [188] Kim TH, Kim GD, Ahn HJ, et al. The inhibitory effect of naringe-
[174] Lee S, Park NJ, Bong SK, et al. Ameliorative effects of Juniperus nin on atopic dermatitis induced by DNFB in NC/Nga mice. Life
rigida fruit on oxazolone-and 2, 4-dinitrochlorobenzene-induced Sci. 2013;93:516–524.
atopic dermatitis in mice. J Ethnopharmacol. 2018;214:160–167. [189] Sung YY, Kim DS, Yang WK, et al. Inhibitory effects of Drynaria
[175] Jung AR, Ahn SH, Park IS, et al. Douchi (fermented Glycine max fortunei extract on house dust mite antigen-induced atopic
Merr.) alleviates atopic dermatitis-like skin lesions in NC/Nga dermatitis in NC/Nga mice. J Ethnopharmacol. 2012;144:94–100.
mice by regulation of PKC and IL-4. BMC Complement Altern [190] Choi SE, Park KH, Jeong MS, et al. Effect of Alnus japonica
Med. 2016;16:1–4. extract on a model of atopic dermatitis in NC/Nga mice. J
[176] Lee J, Choi YY, Kim MH, et al. Topical application of Angelica Ethnopharmacol. 2011;136:406–413.
[191] Lin L, Zhou Y, Li H, et al. Polysaccharide extracted from Chinese
sinensis improves pruritus and skin inflammation in mice with
white wax scale ameliorates 2, 4-dinitrochlorobenzene-induced
atopic dermatitis-like symptoms. J Med Food 2016;19:98–105.
atopic dermatitis-like symptoms in BALB/c mice. Saudi Pharm J.
[177] Bae MJ, See HJ, Choi G, et al. Regulatory T cell induced by Poria
2017;25:625–632.
cocos bark exert therapeutic effects in murine models of atopic
[192] Wu CT, Huang KS, Yang CH, et al. Inhibitory effects of cultured
dermatitis and food allergy. Mediators Inflamm. 2016;2016:
Dendrobium tosaense on atopic dermatitis murine model. Int J
3472608.
Pharm. 2014;463:193–200.
[178] Tsang MS, Jiao D, Chan BC, et al. Anti-inflammatory activities of
[193] Sozmen SC, Karaman M, Micili SC, et al. Resveratrol ameliorates
pentaherbs formula, berberine, gallic acid and chlorogenic acid
2, 4-dinitrofluorobenzene-induced atopic dermatitis-like lesions
in atopic dermatitis-like skin inflammation. Molecules. 2016; 21:
through effects on the epithelium. Peer J. 2016;4:e1889.
519. [194] Lee H, Ha H, Lee JK, et al. The leaves of Broussonetia kazinoki
[179] Sung YY, Lee AY, Kim HK. The Gardenia jasminoides extract and siebold inhibit atopic dermatitis-like response on mite allergen-
its constituent, geniposide, elicit anti-allergic effects on atopic treated Nc/Nga mice. Biomol Ther. 2014;22:438–444.
dermatitis by inhibiting histamine in vitro and in vivo. J [195] Yang IJ, Lee DU, Shin HM. Inhibitory effect of valencene on the
Ethnopharmacol. 2014;156:33–40. development of atopic dermatitis-like skin lesions in NC/Nga
[180] Choi JH, Jin SW, Park BH, et al. Cultivated ginseng inhibits 2, 4- mice. Evid Complement Alternat Med. 2016;2016:9370893.
dinitrochlorobenzene-induced atopic dermatitis-like skin lesions [196] Yang JH, Yoo JM, Cho WK, et al. Ethanol extract of sanguisorbae
in NC/Nga mice and TNF-a/IFN-c-induced TARC activation in radix inhibits mast cell degranulation and suppresses 2, 4-dini-
HaCaT cells. Food Chem Toxicol. 2013;56:195–203. trochlorobenzene-induced atopic dermatitis-like skin lesions.
[181] Park JH, Ahn EK, Ko HJ, et al. Korean red ginseng water extract Mediators Inflamm. 2016;2016:2947390.
alleviates atopic dermatitis-like inflammatory responses by nega- [197] Amagai Y, Katsuta C, Nomura Y, et al. Amelioration of atopic-
tive regulation of mitogen-activated protein kinase signaling like skin conditions in NC/Tnd mice by topical application with
pathway in vivo. Biomed Pharmacother. 2019;117:109066. distilled Alpinia intermedia Gagnep extracts. J Dermatol. 2017;44:
[182] Lee BH, Kim HK, Jang M, et al. Effects of gintonin-enriched frac- 1238–1247.
tion in an atopic dermatitis animal model: involvement of auto- [198] Akhtar N, Verma A, Pathak K. Exploring preclinical and clinical
taxin regulation. Biol Pharm Bull. 2017;40:1063–1070. effectiveness of nanoformulations in the treatment of atopic
[183] Han NR, Park JY, Jang JB, et al. A natural dye, Niram improves dermatitis: Safety aspects and patent reviews. Bull Fac Pharm
atopic dermatitis through down-regulation of TSLP. Environ Cairo Univ. 2017;55:121.
Toxicol Pharmacol. 2014;38:982–990. [199] Vavrova K. Emerging small-molecule compounds for treatment
[184] Sur B, Lee B, Yoon YS, et al. Extract of polygala tenuifolia allevi- of atopic dermatitis: a review. Expert Opin Ther Pat. 2016;26:
ates stress-exacerbated atopy-like skin dermatitis through the 21–34.

You might also like