Sie sind auf Seite 1von 7

Pharmacology and therapeutics

Methylprednisolone aceponate for atopic dermatitis


Antonio Torrelo, MD

Department of Dermatology, Hospital Abstract


~o Jesu
Infantil Nin s, Madrid, Spain Background The 4th generation topical corticosteroids (TCS) have demonstrated a most
favorablerisk–benefit ratio. Methylprednisolone aceponate (MPA) is a non-halogenated
Correspondence
Antonio Torrelo, MD
corticosteroid with a methyl group at C6, which confers higher intrinsic activity. MPA is
Department of Dermatology, Hospital included in the group of potent TCS (category III/IV).
~o Jesu
Infantil Nin s Methods A literature review is carried out of the clinical efficacy, pharmacokinetics, and
Mene ndez Pelayo 65, 28009-Madrid adverse effects of MPA, especially for the treatment of atopic dermatitis (AD).
Spain
Results Several clinical studies support the use of MPA in infants and children, with
E-mail: atorrelo@aedv.es
minimal local or systemic adverse effects reported. The pharmacokinetic profile and the
Conflicts of interest: The author has acted low rate of adverse effects of MPA are most suitable for the treatment of atopic dermatitis
and has been paid as a consultant and (AD), a chronic disease with frequent flaring that can involve extensive areas of the skin.
lecturer for Bayer AG in the last 3 years, a Conclusions Most patients with AD can be easily brought into control with the use of only
company manufacturing the product being
TCS. Achieving a complete healing of eczema is key in AD, and once the skin is clinically
studied, and on issues related and
unrelated to the product. The author has
healthy, emollients can be used according to the physician and patient preferences.
not received any payment for writing this Physicians should be trained in the recognition of early or subtle manifestations of active
article. eczema that are most suitably treated with topical TCS to achieve a most rapid and
satisfactory control of the disease. If the whole area with eczema is not treated, active
doi: 10.1111/ijd.13485
eczema will remain and treatment will be ineffective. Insufficient use of TCS will lead to
inefficiency and frustration.

1980s, new TCS molecules that could retain the high potency
Introduction
of action and had a much lower profile of adverse effects were
In this article, the author will review the most important features researched, and hence the so-called 4th generation TCS, were
regarding the treatment of atopic dermatitis (AD) with topical marketed in the late 80s and early 90s. They include mometa-
corticosteroids (TCS), with special emphasis on the 4th genera- sone furoate, hydrocortisone-17-butyrate-21-propionate, predni-
tion TCS methylprednisolone aceponate (MPA). Recent carbate, and methylprednisolone aceponate (MPA), and they
advances in the pathogenesis of AD justify a shift in the para- demonstrate a most favorable risk-benefit ratio.1 Interestingly,
digm of therapy of the disease with the most potent and safe 25 years after the marketing of the 4th generation TCS, no
available first-line agents such as MPA. improvement in the synthesis of new corticosteroid molecules
has been produced, which gives an idea that the available 4th
generation TCS meet a high standard of efficacy and safety.
A Brief History of Topical Corticosteroids

Hydrocortisone was the first topical corticosteroid (TCS) avail-


Pharmacological Properties of MPA
able for the treatment of cutaneous diseases, and its use meant
a revolution in dermatology in the 1950s. Patients with condi- Methylprednisolone aceponate is a nonhalogenated corticos-
tions such as eczema, which was mostly untreatable, obtained teroid with a methyl group at C6, which confers higher intrinsic
a relief never achieved before. After the introduction of this low- activity.2 The ester groups in C17 and C21 render the molecule
potency agent, the pharmaceutical companies synthetized highly lipophilic, convenient for more efficient penetration within
higher potency TCS during the 60s and 70s, usually by includ- the stratum corneum and low serum concentrations2,3 (Fig. 1).
ing a halogen atom (halogenation) or methylation in the basic In the skin, MPA is hydrolyzed by skin esterases into methyl-
molecule of the steroid scaffold. An extensive use of these prednisolone 17-propionate (MPP), the active metabolite of
agents was associated with the apparition of adverse skin and MPA with a higher affinity for glucocorticosteroid receptors.4
less frequently systemic reactions that partially occluded the This bioactivation is enhanced in inflamed skin because of
therapeutic effects of the TCS and led to a generalized opinion higher levels of esterases,4,5 which further concentrates MPP at
that TCS were even more harmful than beneficial. During the the therapeutic site. Once in the skin, MPP undergoes migration
1

ª 2017 The International Society of Dermatology International Journal of Dermatology 2017


2 Pharmacology and therapeutics Methylprednisolone aceponate for atopic dermatitis Torrelo et al.

eczematous disorders,1,15,18 comparable to twice-daily


betamethasone valerate.
Methylprednisolone aceponate efficacy studies have included
mostly patients with varied eczematous conditions including ato-
pic dermatitis, contact dermatitis, and others. Furthermore,
patients with age ranges from 2 months to 87 years have been
included, thus showing age-independent efficacy.1

Safety and Tolerability of MPA


Whereas the efficacy of a potent TCS such as MPA can be
easily demonstrated and generally affected, safety and tolerabil-
ity of a TCS must be thoroughly proven. Because TCS of high
and ultra-high potency are available, the physician’s greatest
concern about TCS is not efficacy but rather a favorable profile
Figure 1 Molecular structure of methylprednisolone aceponate of adverse effects. In the past, efficacy and adverse effects
were invariably linked, but 4th generation TCS showed that a
of the propionate group from C-17 to C-21, to form methylpred- high therapeutic index (i.e., high potency with low adverse
nisolone-21-propionate, which is in turn hydrolyzed and ren- effects) is possible, thus dissociating the concepts of ‘potent’
dered relatively inactive.4 Any MPP reaching systemic and ‘harmful’ TCS.19–22 Overall, studies involving animal mod-
absorption is rapidly inactivated by conjugation with glucuronic els, healthy volunteers, and patients have demonstrated that
acid to an inactive metabolite that is mainly excreted in the adverse effects with MPA are significantly lower than other
urine.4 potent TCS and are similar to those of less potent TCS.8,10,17
The adverse effects of TCS are well known. They include
local effects from topical application and systemic effects from
Efficacy of MPA
percutaneous absorption of the TCS reaching significant blood
Methylprednisolone aceponate is included in the group of potent levels. The latter resemble the adverse effects of orally adminis-
TCS (category III/IV); triamcinolone acetonide and bethameta- tered corticosteroids.
sone valerate are reference TCS also included in this group.6
Multiple double-blind studies, both placebo-controlled and com- Local adverse effects
pared to reference TCS, have shown a high clinical efficacy of The most common and largely studied adverse effect of TCS is
MPA in the treatment of eczema after 1–4 weeks of application skin atrophy. The antiproliferative action of corticosteroids on
once daily.7–17 Of note, once daily MPA is at least equally or keratinocytes and fibroblasts leads to epidermal and dermal
more effective than twice daily betamethasone valerate, a refer- thinning.23 Epidermal thinning is partly reversible, but atrophy of
ence TCS, with treatment success superior to 90% of the dermis and even subcutaneous tissue is often permanent.
patients.13,14 Direct comparison of MPA with other 4th genera- Striae and telangiectasias are a reflection of permanent atrophy
tion TCS showed similar efficacy index than mometasone furo- and are often motive of concern for patients and physi-
ate17 and prednicarbate.8 Most double-blind studies for efficacy cians.6,21,24 Skin atrophy is mostly an effect of long-term use of
of MPA were carried out before its marketing and during mole- TCS, but the epidermal and stratum corneum atrophy can be
cule development, more than 25 years ago. However, the induced even in short-term treatments.
extensive use of MPA in Europe in the postmarketing period Animal models indicate a low atrophogenic potential of
after gaining experience for more than 25 years indicates that it MPA,25,26 higher than vehicle but significantly less than
is a very effective therapy. It is generally accepted that potent mometasone furoate.27 The same was true for the incidence of
TCS are effective molecules that have been helping patients telangiectasias. In humans, although MPA is a potent topical
with skin diseases for more than 60 years. corticosteroid,16 it has been proven to show a low atrophogenic
Most physicians generally believe that application of TCS potential,25,27 possibly lower than betamethasone valerate
twice daily is superior to once daily, but this has been rarely cream and ointment, and similar to hydrocortisone butyrate. In
tested nor proven. Studies with MPA demonstrated that once- 20 healthy volunteers, once-daily MPA cream did not cause any
daily application achieved similar efficacy to twice-daily (92% significant reduction in skin thickness in comparison to vehicle,
vs. 90% of patients treated successfully).2,13,14 whereas betamethasone valerate significantly reduced skin
Different presentations of TCS can show different efficacy thickness.2,25 Both under occlusive and nonocclusive conditions,
rates. MPA once-daily was tested in cream and ointment pre- treatment with clobetasol propionate induced significantly more
sentations, and both showed identical success rates in pronounced atrophy and telangiectasias.28 In 20 healthy

International Journal of Dermatology 2017 ª 2017 The International Society of Dermatology


Torrelo et al. Methylprednisolone aceponate for atopic dermatitis Pharmacology and therapeutics 3

volunteers, a double-blind, 6-week, occlusive, randomized trial Safety in children


showed that treatment with mometasone furoate resulted in a Children, especially infants, present a higher rate of skin sur-
higher incidence and severity of telangiectasias compared with face to volume ratio8 and thus are subject to a possible higher
MPA.17 amount of absorption of topical corticosteroids. Because of the
Visible signs of atrophy were observed in only two of more low profile of topical absorption, MPA cream, milk/emulsion, and
than 1900 patients in clinical trials treated with MPA for ointment have been approved for the treatment of children of all
eczema,2 and it is even likely that these two patients had pre- ages. The milk/emulsion formulation of MPA is especially very
existing atrophy unmasked by the successful action of MPA.13 acceptable cosmetically and can be used safely and effectively
Common local adverse effects observed with MPA include in infants aged 4 months and over.1,2
application site burning and pruritus.6 Although less common, Several clinical studies support the use of MPA in infants and
the following have occurred as well: application site dryness, children,7,8,29,30 with no local or systemic adverse effects
eczema, skin exfoliation, skin fissures, application site pain, reported.8 Trials in children aged 4 months to 15 years with AD
application site vesicles, application site pustules, application reveal that adverse events were mostly absent after the applica-
site erosion, application site papules, application site erythema, tion of 0.1% MPA ointment once daily for 21 days. No effect on
peripheral edema, skin atrophy, ecchymosis, impetigo, greasy plasma cortisol levels was seen after 7 days using nonocclusive
skin, and telangiectasia.6,10,14,15,25,26,28 In addition, it should be dressings in 20 children over 6 months of age with atopic der-
noted that a specific therapy is required in bacterially infected matitis. Mild burning sensation, usually attributed to the vehicle,
skin diseases and/or in fungal infections as local skin infections and infectious skin diseases were the most common adverse
can be potentiated by topical glucocorticoid use. Other con- effects.7,8,31
traindications include tuberculous or syphilitic processes in the
area to be treated, viral diseases (e.g., varicella, herpes zoster),
MPA for the Treatment of Atopic Dermatitis
rosacea, perioral dermatitis, ulcera, acne vulgaris, atrophic skin
diseases, and postvaccination skin reactions in the area to be Corticosteroids (CCS) bind to specific glucocorticoid receptors
treated. (GR) in the cytoplasm and are translocated into the nucleus,
where they exert actions on DNA by activating (transactivation)
Systemic adverse effects and repressing (transrepression) different genes.32,33 Transacti-
These are related to absorption through the skin and are equal vation is usually linked to the metabolic properties of CCS,
to the effects of systemic corticosteroids. They include hyper- whereas transrepression of inflammatory genes leads to thera-
glycemia, hypertension, leukocytosis, and others.24 Because peutic effects.33 The result is a decreased production of many
MPA has a rapid action and suffers inactivation of its metabo- proinflammatory cytokines (including TNFa, IL-1a, IL-3, IL-5,
lites after absorption, a low risk of systemic effects is expected. and granulocyte-macrophage colony stimulating factor) and
None of the effects mentioned above were observed in patients mediators (such as prostaglandins and leukotrienes).6,32
treated with 0.1% MPA ointment for 3 months,10 although it Furthermore, CCS inhibit function and maturation of den-
cannot be excluded that systemic effects due to absorption may dritic cells and leukocyte migration, suppress eosinophil matura-
occur when topical preparations containing corticoids are tion, reduce vascular leakage, and reduce collagen
applied. synthesis.6,19,34–37 With all these anti-inflammatory actions,
Systemic corticosteroids can suppress the hypothalamic– MPA exerts a fast and effective action on the relief of AD, and
pituitary–adrenal (HPA) axis, and there is a concern for topi- this has been demonstrated in randomized trials in children and
cal corticosteroids producing this effect. The measurement of adults,7,15,38 as well as accumulated clinical experience for
cortisol levels reflects HPA axis function.6 MPA did not sup- more than 25 years. Hairy areas of the scalp treated for
press cortisol after 43 days of treatment in rats.6,28 In 100 2 weeks with once daily 0.1% MPA solution also experienced
healthy volunteers, after daily application of 40 g of topical complete remission or significant improvement by patients’ and
0.1% MPA over 60% of the body surface, no changes in physicians’ global assessment.39
cortisol levels were observed over 8 days.6,10,13 In adults Sixty-seven percent of patients with severe AD flares were
with atopic dermatitis involving 40–60% of the skin surface clear of or experienced significant improvement in Investigator’s
treated with 30 g of 0.1% MPA ointment for 7 days, no Global Assessment scores after 3 weeks of once-daily treat-
effects on the HPA axis were noted.6 In 45 adults with ment with 0.1% MPA ointment, a similar proportion as with
chronic dermatoses, application of MPA ointment 1–3 times 0.03% tacrolimus ointment twice daily.9 However, MPA was
daily for 4 months, showed no suppression of endogenous superior to tacrolimus in terms of itching, EASI score, and sleep
cortisol secretion.6,10 These studies demonstrate that MPA quality.9 The study concluded that MPA is recommended as a
has a minimal effect on the HPA axis, in contrast with other first line treatment in AD.
corticosteroids such as clobetasol propionate and betametha- The pharmacokinetic profile and the low rate of adverse effects
sone valerate.2,13 of MPA are most suitable for the treatment of AD, a chronic

ª 2017 The International Society of Dermatology International Journal of Dermatology 2017


4 Pharmacology and therapeutics Methylprednisolone aceponate for atopic dermatitis Torrelo et al.

disease with frequent flaring that can involve extensive areas of defined, and the term ‘dry’ skin is applied when fine scaling is
the skin.14,15,40 Although most physicians apply twice daily TCS present. It should be remembered that scaling is an intrinsic
to AD, MPA permits once-daily administration.8,26,41 On the other component in the definition of eczema and that many patients
hand, physicians use different formulations according to exten- with mild AD have a finely scaling process with slight or minimal
sion, area of the skin involved, exudation, and chronicity of erythema that can be confused with simply ‘dry’ skin.48 The use
lesions, and thus the availability of MPA in formulations including of emollients in active eczema can reduce scaling temporarily
ointment, fatty ointment, cream, lotion and milk/emulsion, all with but will not improve the disease and can cause irritation and fur-
equal efficacy, make it convenient for any patient age, skin condi- ther worsening of eczema. Physicians should be trained in the
tion, location of lesions, and disease severity.6 recognition of early or subtle manifestations of active eczema
that are most suitably treated with TCS to achieve a most rapid
and satisfactory control of the disease. In the author’s experi-
Utilities of MPA According to the New
ence, patients who are instructed to treat minor flares of AD
Discoveries in AD
with TCS have better control of the disease, better quality of
For many years, most experienced dermatologists felt that AD life, and use much less topical treatment than when flare-ups
was primarily a skin disorder, most likely related to skin barrier and mild eczema are left untreated until they worsen or spread,
deficiency. However, extensive study of immunologic pathome- thus necessitating longer and more extensive therapy.
chanisms has led many researchers to consider immune sys-
tem imbalance as the primary abnormality in AD. The recent Achieving control of AD
discovery that filaggrin null mutations lead to genetic suscepti- Because AD is a chronic disease, both physicians and patients
bility to AD and can be responsible for at least a significant pro- may have feelings of frustration and surrendering. In children,
portion of AD patients (up to 40% in certain European the expectation for a remission when the child grows up may
populations) has shifted the paradigm of understanding and lead to a wait-and-see policy that can cause damage to children
management of AD.42,43 Filaggrin null mutations are also a first- in the form of continuous sensitization through damaged skin,
line risk factor for developing asthma and other allergic dis- mood changes, disturbance of academic achievements, and
eases.44 Because filaggrin is mostly expressed in the skin, the frustration from chronic suffering.
current opinion is that primary skin barrier defects lead to It is the author’s opinion and experience that every patient
increased exposure to environmental irritants and allergens, with AD should be brought into complete control, no matter if
causing irritant and allergic eczema.45 Eczema itself further the disease is mild or severe. The reasons are simple: (i) we
impairs skin barrier that becomes fully inefficient to prevent the have good and effective therapies that can be used safely; (ii)
entry of external molecules that are massively presented to the good control leads to a better quality of life; and (iii) control of
skin immune system, thus worsening the patient’s condition and the disease decreases the risk of further overexposure to aller-
leading to a circle of eczema and sensitization. gens and infectious agents. We can definitely change the life of
our patients with an appropriate approach to AD.
Combining emollient and pharmacologic treatment in AD Most patients with AD can be easily brought into control
The use of emollients has been recommended in an extensive with the use of only TCS. Because we have effective and
liberal use for all patients with AD. This seems reasonable in safe 4th generation TCS, their use for any flare-up, as early
view of the recent knowledge that primarily skin barrier defects as possible, on the whole affected area and for as many days
are the origin of AD. However, whereas the use of emollients is as necessary to achieve complete healing is encouraged. An
beneficial for patients with controlled AD and in the areas not insufficient use of TCS will lead to inefficiency and frustration.
affected by eczema, it is the author’s experience, as well as If the whole area with eczema is not treated, active eczema
others’, that the direct application of emollients on the skin will remain and treatment will be ineffective; it is important for
affected with eczema is poorly tolerated and causes itch and physicians and patients to recognize eczema and use 4th
irritation.46 In the author’s opinion, atopic eczema should be generation TCS on every affected area, whether severe or
brought into control before emollients are liberally used, and for mild. If TCS treatment is withdrawn before eczema is under
this, pharmacological therapy is necessary. The 4th generation complete control, the risk of rebound is very high; rebound is
topical CCS are the first-line drugs that should be used in order usually more linked to an insufficient treatment than to TCS
to achieve a complete healing of eczema because of their effec- themselves.
tiveness and safety profile. Once the skin is clinically healthy, Patients must be reassured that using 4th generation TCS is
emollients can be used according to the physician and patient safe and will not cause any harm when properly used. Cortico-
preferences. phobia49 will lead to the use of noneffective remedies, simply
It is generally accepted that the skin of the patients with AD on the basis that they will not cause any harm. In the author’s
is dry, and the concept of ‘dry’ skin has been equivalent to ‘ato- opinion, using ineffective remedies is harmful because AD is
pic’ skin.47 From a clinical point of view, ‘dryness’ is not well harmful itself in terms of affecting quality of life, inducing further

International Journal of Dermatology 2017 ª 2017 The International Society of Dermatology


Torrelo et al. Methylprednisolone aceponate for atopic dermatitis Pharmacology and therapeutics 5

exposure to allergens and the likelihood of serious infectious prevention of the development of the atopic phase of atopic
complications, and should not be left untreated. dermatitis, and possibly other manifestations of the atopic
disease.
Use of MPA for AD in maintenance therapy
Because of the risk of relapse of AD, strategies for maintenance QUESTIONS (answers found after references)
or preventive treatment have been proposed. In one study, 221
adult patients with AD were treated once daily with 0.1% MPA 1 The first available topical corticosteroid was:
cream until control of the disease and were then randomized to A Hydrocortisone
twice weekly 0.1% MPA cream (weekends) on the previously B Methylprednisolone aceponate
affected areas + emollients vs. emollients alone for 16 weeks.50 C Clobetasol propionate
Patients in the MPA branch had a 3.5-fold lower risk of relapse D Betamethasone valerate
with maintenance therapy than with emollient alone and had 2 Which of the following is not a halogenated topical corticos-
lower EASI scores throughout the study.50 During 16 weeks of teroid:
maintenance therapy with MPA, no signs of skin atrophy, striae, A Betamethasone valerate
telangiectasias, irritation, or infection were observed.50 Similar B Methylprednisolone aceponate
studies have been carried out with other TCS, emphasizing C Clobetasol-17-propionate
their effectiveness and safety.51–54 A similar study with intermit- D Betamethasone dipropionate
tent tacrolimus treatment in children and adults with AD also 3 In which group of potency of topical corticosteroids is methyl-
indicated that maintenance therapy can reduce the incidence of prednisolone aceponate included:
flares.55,56 A I/IV (mild)
B II/IV (moderate)
Use of MPA in infants with AD C III/IV (potent)
AD may start in infancy, as early as 2 or 3 months of age. MPA D IV/IV (ultra-potent)
is approved for children from the age of 4 months, which is 4 Which is the main active metabolite of methylprednisolone
unique for this young age. MPA is suitable for the treatment of aceponate:
AD in infants because of its effectiveness, safety profile, and A Methylprednisolone aceponate itself
the convenience of the milk/emulsion formulation. B Methylprednisolone-6-propionate
Following the most currently accepted model for the devel- C Methylprednisolone-17-propionate
opment of AD,57 it is suspected that skin barrier defects in D Methylprednisolone-21-propionate
infants lead to increased passage through the skin of external 5 Which of the following topical corticosteroids shows a lower
agents that reach the upper dermis, thus causing a primary atrophogenic potential on the skin:
irritant eczema. This early, nonatopic eczema further impairs A Betamethasone valerate cream
the skin barrier, thus allowing for more and more external B Betamethasone valerate ointment
molecules, both irritants and allergens, to pass through the C Clobetasol-17-propionate ointment
skin and gain contact with the immune system that, because D Methylprednisolone aceponate ointment
of overexposure to these substances and under genetic 6 Which of the following is a common systemic effect of the
unknown circumstances, will shift towards a Th2-enhanced topical application of methylprednisolone aceponate:
response. Immunologic sensitization will then cause truly ato- A Hyperglycemia
pic dermatitis and other allergic diseases according to the B Hypertension
‘atopic march’. If this situation could be overcome in early C Suppression of the HPA axis
infancy, by restoring the epidermal skin barrier, it is likely that D None of the above
AD will not develop. Certainly, double-blind studies have 7 At which age can methylprednisolone aceponate be used
demonstrated that early intervention with strict use of safely:
emollients in infants at risk of AD permits a reduction in the A 12 years and over
incidence of AD in almost 50% of children.58 In practice, B 6 years and over
however, most infants with AD are brought to pediatricians C 2 years and over
and dermatologists when early, active eczema is already D 6 months and over
present. In such cases, in the author’s experience, application 8 Which is the most commonly known gene mutation responsi-
of emollients will be poorly tolerated and can even ble for atopic dermatitis:
worsen eczema. It is very important to bring early AD under A SPINK5
control before emollients can be used. Maintaining the infants D Corneodesmosine
free of eczema and under appropriate use of emollients will C Filaggrin
lead to full control of the disease and most likely to D Desmoglein 1

ª 2017 The International Society of Dermatology International Journal of Dermatology 2017


6 Pharmacology and therapeutics Methylprednisolone aceponate for atopic dermatitis Torrelo et al.

9 Which are the first-line drugs for the treatment of the atopic corticoid therapy with few side effects. Jahrb Dermatol 1992; 3:
dermatitis flares: 247–263.
14 Fritsch P. Clinical experience with methylprednisolone
A Topical corticosteroids
aceponate (MPA) in eczema. J Dermatolog Treat 1992; 3
B Topical calcineurin inhibitors (Suppl. 2): 17–19.
C Emollients 15 Mensing H, Lorenz B. Experience with methylprednisolone
D Oral antihistamines aceponate (MPA) in patients suffering from acute and chronic
10 Which of the following is false for the management of atopic eczema. Results of a large observational study. Z Hautkr 1998;
73: 281–285.
dermatitis:
16 Kecske s A, Jahn P, Wendt H, et al. Activity of topically applied
A Safe and effective therapies are available for atopic der- methylprednisolone aceponate in relation to other topical
matitis glucocorticosteroids in healthy volunteers. Arzneimittelforschung
B Control of atopic dermatitis leads to a better quality of life 1993; 43: 144–147.
C Control of atopic dermatitis decreases further exposure to 17 Kecske s A, Heger-Mahn D, Kuhlmann RK, et al. Comparison of
the local and systemic side effects of methylprednisolone
allergens
aceponate and mometasone furoate applied as ointments with
D Atopic dermatitis should be left untreated because most equal anti-inflammatory activity. J Am Acad Dermatol 1993; 29:
cases will eventually resolve 576–580.
18 Kungurov NV, Kokhan MM, Keniksfest JV, et al. Difference
topical therapy of the atopic eczema in children and adults
References (poster 2–3). J Eur Acad Dermatol Venereol 2001; 15(Suppl. 2):
107.
1 Blume-Peytavi U, Wahn U. Optimizing the treatment of atopic
19 Brazzini B, Pimpinelli N. New and established topical
dermatitis in children: a review of the benefit/risk ratio of
corticosteroids in dermatology: clinical pharmacology and
methylprednisolone aceponate. J Eur Acad Dermatol Venereol
therapeutic use. Am J Clin Dermatol 2002; 3: 47–58.
2011; 25: 508–515. €uber U. Dermatocorticosteroids: structure, activity,
20 Ta
2 Ruzicka T. Methylprednisolone aceponate in eczema and other
pharmacokinetics. Eur J Dermatol 1994; 4: 419–429.
inflammatory skin disorders – a clinical update. Int J Clin Pract
21 Wozniacka A, Sysa-Jedrzejowska A. Topical steroids – a new
2006; 60: 85–92.
approach after 50 years. Med Sci Monit 2001; 7: 539–544.
3 Toepert M, Olivar A, Opitz D. New developments in
22 Luger T, Loske K, Elsner P, et al. Topical skin therapy with
corticosteroid research. J Dermatolog Treat 1990; 1(Suppl. 3):
glucocorticosteroids – therapeutic index. J Deutsch Dermatol
S5–S9.
Ges 2004; 7: 629–634.
4 Ta€uber U. Pharmacokinetics and bioactivation of MPA. J Eur
23 Mori M, Pimpinelli N, Giannotti B. Topical corticosteroids and
Acad Dermatol Venereol 1994; 3(Suppl. 1): 23–31.
unwanted local effects. Improving the benefit/risk ratio. Drug Saf
5 Zentl HJ, Toepert M. Preclinical evaluation of a new topical
1994; 10: 406–412.
corticosteroid methylprednisolone aceponate. J Eur Acad
24 Hengge UR, Ruzicka T, Schwartz RA, et al. Adverse effects of
Dermatol Venereol 1994; 3(Suppl. 1): 32–38.
topical glucocorticosteroids. J Am Acad Dermatol 2006; 54: 1–15.
6 Luger TA. Balancing efficacy and safety in the management of
25 Ortonne J-P. Skin atrophogenic potential of methylprednisolone
atopic dermatitis: the role of methylprednisolone aceponate.
aceponate (MPA). J Eur Acad Dermatol Venereol 1994; 3
J Eur Acad Dermatol Venereol 2011; 25: 251–258.
(Suppl. 1): S13–S18.
7 Korotky NG, Taganov AV, Shimanovsky NL. Use of Advantan in s A, Ta€uber U, et al. Methylprednisolone
26 Zaumseil R-P, Kecske
the treatment of atopic dermatitis in children. Pediatriya 2000; 5:
aceponate (MPA) – a new therapeutic for eczema: a
75–78.
pharmacological overview. J Dermatol Treat 1992; 3(Suppl. 2):
8 Rampini E. Methylprednisolone aceponate (MPA) – use and
3–7.
clinical experience in children. J Dermatol Treat 1992; 3(Suppl. €cke W-D, Merbold U, et al. Superior
27 Mirshahpanah P, Do
2): 27–29.
nuclear receptor selectivity and therapeutic index of
9 Bieber T, Vick K, Fo €lster-Holst R, et al. Efficacy and safety
methylprednisolone aceponate versus mometasone furoate. Exp
of methylprednisolone aceponate ointment 0.1% compared
Dermatol 2007; 16: 753–761.
to tacrolimus 0.03% in children and adolescents with an s A, Jahn P, Lange L. Local tolerability of topically
28 Kecske
acute flare of severe atopic dermatitis. Allergy 2007; 62:
applied methylprednisolone aceponate. J Am Acad Dermatol
184–189.
1993; 28: 786–788.
10 Ortonne JP. Safety aspects of topical methylprednisolone
29 Korotky NG, Taganov AV. The use of steroid Advantan
aceponate (MPA) treatment. J Dermatol Treat 1992; 3(Suppl.
(methylprednisolone aceponate) for management of
2): 21–25.
allergodermatoses in children. Vestn Dermatol Venereol 2000;
11 Grebenyuk VN, Balabolkin II. Progress in topical corticosteroid
3: 61–63.
therapy of atopic dermatitis in children. Pediatriya 1998; 5: 88–
30 Soennichsen N, Mekbib K. Die behandlung des atopischen
91.
ekzems (neurodermitis) im kindesalter mit 6-alpha-
12 Niedner R, Zaumseil R-P. Advantan milk ⁄ cream.ointment in
methylprednisoloneaceponat [in German]. Der Deutsche
children with atopic dermatitis and other inflammatory or
Dermatologe 1996; 44: 8.
eczematous dermatoses – an observational study in 558
31 Alchorne MM, Da CP, Cestari S, et al. A multicenter,
children. Aktuelle Derm 2004; 30: 200–203.
comparative, open-labelled, randomized study of tolerability and
13 Zaumseil R-P, Fuhrmann H, Kecske0 s A, et al.
efficacy of methylprednisolone aceponate and mometasone
Methylprednisolone aceponate (Advantan) – an effective topical

International Journal of Dermatology 2017 ª 2017 The International Society of Dermatology


Torrelo et al. Methylprednisolone aceponate for atopic dermatitis Pharmacology and therapeutics 7

furoate in children from 2 to14 years of age suffering from dermatitis outpatients and their parents. PLoS ONE 2013; 8:
atopic dermatitis. Ped Mod 2003; 39: 275–280. e76493.
32 Nawata H, Okabe T, Yanase T, et al. Mechanism of action and 50 Peserico A, Sta €dtler G, Sebastian M, et al. Reduction of
resistance to glucocorticoid and selective glucocorticoid receptor relapses of atopic dermatitis with methylprednisolone aceponate
modulator to overcome glucocorticoid-related adverse effects. cream twice weekly in addition to maintenance treatment with
Clin Exp Allergy Rev 2008; 8: 53–56. emollient: a multicentre, randomized, double-blind, controlled
33 Newton R, Holden NS. Separating transrepression and study. Br J Dermatol 2008; 158: 801–807.
transactivation: a distressing divorce for the glucocorticoid 51 Van Der Meer JB, Glazenburg EJ, Mulder PG, et al. The
receptor? Mol Pharmacol 2007; 72: 799–809. management of moderate to severe atopic dermatitis in adults
34 Hoetzenecker W, Meingassner JG, Ecker R, et al. with topical fluticasone propionate. The Netherlands Adult Atopic
Corticosteroids but not pimecrolimus affect viability, maturation DermatitisStudy Group. Br J Dermatol 1999; 140: 1114–1121.
and immune function of murine epidermal Langerhans cells. J 52 Hanifin J, Gupta AK, Rajagopalan R. Intermittent dosing of
Invest Dermatol 2004; 122: 673–684. fluticasone propionate cream for reducing the risk of relapse in
35 Schwiebert LM, Beck LA, Stellato C, et al. Glucocorticosteroid atopic dermatitis patients. Br J Dermatol 2002; 147: 528–537.
inhibition of cytokine production: relevance to antiallergic 53 Berth-Jones J, Damstra RJ, Golsch S, et al. ; Multinational
actions. J Allergy Clin Immunol 1996; 97: 143–152. Study Group. Twice weekly fluticasone propionate added to
36 Boschetto P, Rogers DF, Fabbri LM, et al. Corticosteroid emollient maintenance treatment to reduce risk of relapse in
inhibition of airway microvascular leakage. Am Rev Respir Dis atopic dermatitis: randomised, double blind, parallel group study.
1991; 143: 605–609. BMJ 2003; 326: 1367.
37 Haapasaari KM, Risteli J, Koivukangas V, et al. Comparison of 54 Glazenburg EJ, Wolkerstorfer A, Gerretsen AL, et al. Efficacy
the effect of hydrocortisone, hydrocortisone-17-butyrate and and safety of fluticasone propionate 0.005% ointment in the
betamethasone on collagen synthesis in human skin in vivo. long-term maintenance treatment of children with atopic
Acta Derm Venereol 1995; 75: 269–271. dermatitis: differences between boys and girls? Pediatr Allergy
38 Albrecht G. Clinical comparison of methylprednisolone Immunol 2009; 20: 59–66.
aceponate and prednicarbate in chronic eczema. J Eur Acad 55 Thacßi D, Reitamo S, Gonzalez Ensenat MA, et al. Proactive
Dermatol Venereol 1994; 3(Suppl. 1): S42–S48. disease management with 0.03% tacrolimus ointment for
39 Sonnichsen N, Zaumseil R-P. Efficacy and tolerability of children with atopic dermatitis: results of a randomized,
methylprednisolone aceponate solution in eczematous diseases multicentre, comparative study. Br J Dermatol 2008; 159: 1348–
of the hairy scalp. Dermatology 1999; 5: 317–324. 1356.
40 Haria M, Balfour JA. Methylprednisolone aceponate. A review of 56 Wollenberg A, Reitamo S, Atzori F, et al. Proactive treatment of
its pharmacological properties and therapeutic potential in the atopic dermatitis in adults with 0.1% tacrolimus ointment. Allergy
topical treatment of eczema. Clin Immunother 1995; 3: 241–253. 2008; 63: 742–750.
41 Haneke E. The treatment of atopic dermatitis with 57 Bieber T. Atopic dermatitis. N Engl J Med 2008; 358: 1483–
methylprednisolone aceponate (MPA), a new topical 1494.
corticosteroid. J Dermatol Treat 1992; 3(Suppl. 2): 13–15. 58 Simpson EL, Chalmers JR, Hanifin JM, et al. Emollient
42 Palmer CN, Irvine AD, Terron-Kwiatkowski A, et al. Common enhancement of the skin barrier from birth offers effective atopic
loss-of-function variants of the epidermal barrier protein filaggrin dermatitis prevention. J Allergy Clin Immunol 2014; 134: 818–
are a major predisposing factor for atopic dermatitis. Nat Genet 823.
2006; 38: 441–446.
43 Irvine AD, McLean WH, Leung DY. Filaggrin mutations
associated with skin and allergic diseases. N Engl J Med 2011;
365: 1315–1327. Answers to Questions
44 McAleer MA, Irvine AD. The multifunctional role of filaggrin in
allergic skin disease. J Allergy Clin Immunol 2013; 131: 280–
1 A
291.
45 O’Regan GM, Irvine AD. The role of filaggrin in the atopic 2 B
diathesis. Clin Exp Allergy 2010; 40: 965–972. 3 C
46 Gelmetti C, Wollenberg A. Atopic dermatitis – all you can do 4 C
from the outside. Br J Dermatol 2014; 170(Suppl. 1): 19–24. 5 D
47 Boralevi F, Saint Aroman M, Delarue A, et al. Long-term
6 D
emollient therapy improves xerosis in children with atopic
dermatitis. J Eur Acad Dermatol Venereol 2014; 28: 1456–1462. 7 D
48 Uehara M, Miyauchi H. The morphologic characteristics of dry 8 C
skin in atopic dermatitis. Arch Dermatol 1984; 120: 1186–1190. 9 A
49 Moret L, Anthoine E, Aubert-Wastiaux H, et al. TOPICOP©: a 10 D
new scale evaluating topical corticosteroid phobia among atopic

ª 2017 The International Society of Dermatology International Journal of Dermatology 2017

Das könnte Ihnen auch gefallen