ArticlePDF AvailableLiterature Review

Targeting Interleukin 13 for the Treatment of Atopic Dermatitis

Authors:

Abstract and Figures

Atopic dermatitis (AD) is a common chronic inflammatory skin condition that has a significant impact on a patient’s quality of life and requires ongoing management. Conventional topical and systemic therapies do not target specific components of AD pathogenesis and, therefore, have limited efficacy and may be associated with long-term toxicity. Thus, AD management is challenging, with a significant proportion of patients not achieving clear skin or a reduction in pruritus. There remains a large unmet need for effective therapeutic strategies with favorable safety profiles that can be used long-term in patients with refractory AD. The emergence of targeted biological and small molecule therapies has effectively broadened available treatment options for moderate-to-severe AD. Most recently, interleukin 13 (IL-13) inhibitors were shown to be efficacious and well-tolerated, with tralokinumab already approved for use in this patient population. It is important for dermatologists to be aware of the evidence behind this emerging class of biologic agents to guide treatment choices and improve outcomes in patients with AD. The main objective of this paper is to review the current literature regarding the efficacy and safety of current and emerging anti-IL-13 monoclonal antibodies, including tralokinumab, lebrikizumab, cendakimab, and eblasakimab, for the treatment of moderate-to-severe AD.
Content may be subject to copyright.
Citation: Lytvyn, Y.; Gooderham, M.
Targeting Interleukin 13 for the
Treatment of Atopic Dermatitis.
Pharmaceutics 2023,15, 568.
https://doi.org/10.3390/
pharmaceutics15020568
Academic Editor: Bozena B.
Michniak-Kohn
Received: 23 November 2022
Revised: 24 January 2023
Accepted: 6 February 2023
Published: 8 February 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
pharmaceutics
Review
Targeting Interleukin 13 for the Treatment of Atopic Dermatitis
Yuliya Lytvyn 1and Melinda Gooderham 2,3,4,*
1Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
2SKiN Centre for Dermatology, Peterborough, ON K9J 5K2, Canada
3Probity Medical Research, Waterloo, ON N2J 1C4, Canada
4Department of Family Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
*Correspondence: mgooderham@centrefordermatology.com
Abstract:
Atopic dermatitis (AD) is a common chronic inflammatory skin condition that has a
significant impact on a patient’s quality of life and requires ongoing management. Conventional
topical and systemic therapies do not target specific components of AD pathogenesis and, therefore,
have limited efficacy and may be associated with long-term toxicity. Thus, AD management is
challenging, with a significant proportion of patients not achieving clear skin or a reduction in
pruritus. There remains a large unmet need for effective therapeutic strategies with favorable safety
profiles that can be used long-term in patients with refractory AD. The emergence of targeted
biological and small molecule therapies has effectively broadened available treatment options for
moderate-to-severe AD. Most recently, interleukin 13 (IL-13) inhibitors were shown to be efficacious
and well-tolerated, with tralokinumab already approved for use in this patient population. It is
important for dermatologists to be aware of the evidence behind this emerging class of biologic
agents to guide treatment choices and improve outcomes in patients with AD. The main objective
of this paper is to review the current literature regarding the efficacy and safety of current and
emerging anti-IL-13 monoclonal antibodies, including tralokinumab, lebrikizumab, cendakimab, and
eblasakimab, for the treatment of moderate-to-severe AD.
Keywords:
interleukin 13; lebrikizumab; tralokinumab; cendakimab; eblasakimab; atopic dermatitis
1. Introduction
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin con-
ditions which impacts 15–20% of people in developed countries [
1
4
], with 80% of cases
typically onsetting in infancy or childhood [
5
]. AD is characterized by localized or dissem-
inated pruritic, xerotic, and erythematous lesions which are frequently accompanied by
sleep disturbances, reduced productivity, decreased self-esteem, social isolation, depres-
sion, and suicidal ideation in severe cases [
6
10
]. Thus, the relapsing chronic symptoms
of AD have a significant impact on quality of life and require ongoing long-term manage-
ment [
6
,
11
,
12
]. Moderate-to-severe AD constitutes about 20% of the cases and requires
treatment with phototherapy and/or conventional systemic immunosuppressive agents,
such as corticosteroids, methotrexate, mycophenolate mofetil, cyclosporin A, and aza-
thioprine [
11
,
13
16
]. These conventional therapies do not target specific components of
AD pathogenesis and, therefore, often have limited efficacy and may be associated with
long-term toxicity, posing challenges in AD management [7,8,15,17,18].
In recent years, the emergence of targeted biological and small molecule therapies
has effectively broadened the available treatment options for moderate-to-severe AD. The
first available biological agent for this patient population was dupilumab, an inhibitor of
interleukin-4 (IL-4) receptor-
α
, which blocks IL-4 and IL-13 signaling and prevents the
downstream inflammatory cascade [
19
21
]. Despite the demonstrated efficacy and safety
of dupilumab in AD treatment, only 40% of patients were shown to achieve clear or almost
clear skin in clinical trials [
22
24
], and real-world data suggest about a 70% improvement
Pharmaceutics 2023,15, 568. https://doi.org/10.3390/pharmaceutics15020568 https://www.mdpi.com/journal/pharmaceutics
Pharmaceutics 2023,15, 568 2 of 18
in the clinical severity score (Eczema Area and Severity Index, EASI) after 3 months
of treatment [
25
]. Moreover, dupilumab’s use may be limited by potential associated
adverse effects in 8–38% of patients, such as conjunctivitis, injection site reactions, and
persistent head and neck erythema [
25
27
]. It is believed that inhibition of IL-13 is the
dominant mechanism of dupilumab’s effectiveness in treating AD [
28
,
29
]. JAK-STAT
pathway inhibitors were also shown to be safe and efficacious for use in AD, and, thus,
abrocitinib and upadacitinib have both been approved by the European Commission,
Health Canada, and the FDA for use in moderate-to-severe AD [
30
]. Most recently, IL-13
inhibitors have been studied for the management of AD. The main objective of this paper
is to review the current literature regarding the efficacy and safety of current and emerging
IL-13 monoclonal antibodies, including tralokinumab, lebrikizumab, cendakimab, and
eblasakimab, for the treatment of moderate-to-severe AD.
2. Methods
A search was conducted from the study’s inception up until 30 October 2022, in
the OVID PubMed, Medline, and Google Scholar databases and on ClinicalTrials.gov.
The following specific keywords present in the title, abstract, or body were used for the
search: “interleukin 13,” “lebrikizumab”, “tralokinumab”, “cendakimab”, “eblasakimab”,
“atopic dermatitis,” and “eczema.” Review articles and clinical trials were included in
our summary of the literature. The reference lists of included articles were reviewed for
retrieval of relevant studies not identified in the original search. Only studies involving
human patients published in the English language were included.
3. The Role of Interleukin 13 in the Pathogenesis of Atopic Dermatitis
The pathogenesis of AD is multifactorial, driven by an interplay of environmental
and genetic factors that trigger inflammation, dysbiosis, and immune dysregulation of
the cutaneous epidermal layer [
18
,
31
,
32
]. This markedly increases transdermal water loss
and facilitates permeation by irritants, microbes, and allergens [
32
35
]. An aberrant type
2 immune response is triggered by these antigens stimulating naïve T cells to commit to
the Th2 lineage [
20
,
36
,
37
]. This ultimately leads to the overproduction of cytokines central
to atopic manifestations of AD and pruritus, namely IL-4, IL-5, IL-13, and IL-31 [
38
,
39
].
IL-4 and IL-13 contribute to epidermal barrier dysfunction by stimulating the production
of immunoglobulin E (IgE), recruiting eosinophils, amplifying Th2 cell differentiation,
and reducing filaggrin expression [
18
,
37
,
40
]. IL-4 and IL-13 also decrease anti-microbial
peptide (AMP) production by keratinocytes, which predisposes the skin to Staphylococcus
aureus colonization [
20
]. This may explain the altered skin microbiome in AD, whereby an
abundance of S. aureus and a decrease in bacterial diversity further disrupt the epidermal
barrier [33,4150].
IL-4 and IL-13 are produced predominantly by activated Th2 cells, type 2 innate lym-
phoid cells (ILC2s), and, to a lesser extent, by type 2 CD8(+) T cells (Tc2), basophils,
eosinophils, and mast cells. IL-4 and IL-13 trigger a signaling cascade via a shared
heterodimeric receptor formed by IL-4 receptor
α
(IL-4R
α
) and IL-13 receptor
α
1 (IL-
13R
α
1) [
20
,
39
,
51
]. The binding of either IL-4 or IL-13 to IL-13R
α
1 recruits IL-4R
α
, caus-
ing dimerization of the receptors, activation of Janus kinase 1 (JAK1) and tyrosine ki-
nase 2 (TYK2), and phosphorylation of a signal transducer and activator of transcription
6 (STAT6), which promotes Th2 differentiation [
8
,
20
,
52
]. IL-13 also has a high affinity
for the IL-13R
α
2 receptor, which plays a role in the endogenous regulation of IL-13 and
in the itch–scratch cycle, collagen deposition, and fibrotic tissue remodeling [
34
,
53
,
54
].
Thus, IL-13 may contribute to histamine-independent stimulation of afferent nerve endings
and pruritus [
34
]. Interestingly, skin biopsy samples of patients with AD demonstrate
significant overexpression of IL-13 in lesional and non-lesional skin, and only a mild IL-4
overexpression is detectable in 40% of AD lesions [
28
,
55
]. Moreover, IL-13 overexpression
in peripheral blood T cells was shown to correlate with disease severity, while a decrease
in its concentration correlates with improved clinical outcomes [
55
61
]. Overall, IL-13 is
Pharmaceutics 2023,15, 568 3 of 18
emerging to play an increasingly prominent role in the epidermal barrier dysfunction and
inflammatory processes associated with AD [20,62].
4. Therapies Targeting Interleukin-13 for Management of Atopic Dermatitis
IL-13 is a promising drug target for the management of AD, with a great potential for
efficacy and limited toxicity [
28
]. Therefore, four selective IL-13 inhibitors have emerged
which are currently at various stages of development or approval. Tralokinumab has
recently been approved for use in patients with moderate-to-severe AD by Health Canada,
the FDA, and the European Commission. Lebrikizumab is currently in the late stages of
clinical development, while cendakimab and eblasakimab are being investigated in phase
II trials. See Figure 1for the mechanisms of action of these agents.
Pharmaceutics 2023, 15, x FOR PEER REVIEW 3 of 17
peripheral blood T cells was shown to correlate with disease severity, while a decrease in
its concentration correlates with improved clinical outcomes [5561]. Overall, IL-13 is
emerging to play an increasingly prominent role in the epidermal barrier dysfunction and
inflammatory processes associated with AD [20,62].
4. Therapies Targeting Interleukin-13 for Management of Atopic Dermatitis
IL-13 is a promising drug target for the management of AD, with a great potential for
efficacy and limited toxicity [28]. Therefore, four selective IL-13 inhibitors have emerged
which are currently at various stages of development or approval. Tralokinumab has re-
cently been approved for use in patients with moderate-to-severe AD by Health Canada,
the FDA, and the European Commission. Lebrikizumab is currently in the late stages of
clinical development, while cendakimab and eblasakimab are being investigated in phase
II trials. See Figure 1 for the mechanisms of action of these agents.
Figure 1. Mechanisms of IL-13 inhibition by dupilumab, lebrikizumab, tralokinumab, and eblasa-
kimab in atopic dermatitis [18,20,63]. Dupilumab binds to the IL-4Rα subunit of IL-4 and IL-13 re-
ceptor complexes to reduce inflammatory signaling. Lebrikizumab is a fully human IgG4 that binds
to IL-13 and prevents the formation of the IL-13Rα1/IL-4Rα heterodimer receptor signaling com-
plex. Tralokinumab is fully human IgG4λ that binds to IL-13 preventing binding to the IL-13 recep-
tor. Eblasakimab is a monoclonal antibody that targets IL13Rα1 to block IL-13 signal transduction.
It, therefore, interferes with IL-4 signaling elicited via the type 2 receptor, but not the type 1 receptor.
This figure was created with Biorender at www.biorender.com.
5. Use of Tralokinumab to Manage Patients with Atopic Dermatitis
5.1. Overview of Tralokinumab
Tralokinumab is approved for use in patients 18 years of age or older with moderate-
to-severe AD refractory to topical therapies. It is a fully humanized IgG4λ anti-IL-13 mon-
oclonal antibody that competitively blocks the binding of IL-13 to both IL-13Rα1 and IL-
13Rα2 receptor chains (Figure 1) [18,53,63–65]. In vitro studies confirmed the suppression
of inflammation and restoration of the skin barrier by tralokinumab [65]. In these studies,
tralokinumab treatment normalized the upregulated type 2 inflammatory markers and
the downregulated genes related to terminal keratinocyte differentiation (i.e., filaggrin
and loricrin) in primary human epidermal keratinocytes and human dermal fibroblasts
pre-treated with IL-13 [65]. Recent investigations of the skin of patients with AD treated
with tralokinumab showed increased microbial diversity, decreased Staphylococcus au-
reus, and increased coagulase-negative Staphylococci with treatment [66]. Therefore,
Figure 1.
Mechanisms of IL-13 inhibition by dupilumab, lebrikizumab, tralokinumab, and
eblasakimab in atopic dermatitis [
18
,
20
,
63
]. Dupilumab binds to the IL-4R
α
subunit of IL-4 and IL-13
receptor complexes to reduce inflammatory signaling. Lebrikizumab is a fully human IgG4 that binds
to IL-13 and prevents the formation of the IL-13R
α
1/IL-4R
α
heterodimer receptor signaling complex.
Tralokinumab is fully human IgG4
λ
that binds to IL-13 preventing binding to the IL-13 receptor.
Eblasakimab is a monoclonal antibody that targets IL13R
α
1 to block IL-13 signal transduction. It,
therefore, interferes with IL-4 signaling elicited via the type 2 receptor, but not the type 1 receptor.
This figure was created with Biorender at www.biorender.com.
5. Use of Tralokinumab to Manage Patients with Atopic Dermatitis
5.1. Overview of Tralokinumab
Tralokinumab is approved for use in patients 18 years of age or older with moderate-
to-severe AD refractory to topical therapies. It is a fully humanized IgG4
λ
anti-IL-13
monoclonal antibody that competitively blocks the binding of IL-13 to both IL-13R
α
1
and IL-13R
α
2 receptor chains (Figure 1) [
18
,
53
,
63
65
].
In vitro
studies confirmed the
suppression of inflammation and restoration of the skin barrier by tralokinumab [
65
]. In
these studies, tralokinumab treatment normalized the upregulated type 2 inflammatory
markers and the downregulated genes related to terminal keratinocyte differentiation
(i.e., filaggrin and loricrin) in primary human epidermal keratinocytes and human dermal
fibroblasts pre-treated with IL-13 [
65
]. Recent investigations of the skin of patients with AD
treated with tralokinumab showed increased microbial diversity, decreased Staphylococcus
aureus, and increased coagulase-negative Staphylococci with treatment [
66
]. Therefore,
tralokinumab may improve the dysbiosis of bacterial growth observed in the skin of patients
with AD. The bioavailability of tralokinumab in human studies is 61%, with peak serum
concentrations reached after 3–9 days (median: 5 days) and a mean half-life of 19.3 days
when administered subcutaneously and 21.4 days when administered intravenously [67].
Pharmaceutics 2023,15, 568 4 of 18
5.2. Clinical Efficacy of Tralokinumab in Patients with Atopic Dermatitis
There were significant clinical benefits observed with the use of tralokinumab to man-
age patients with AD in phase II and phase III trials (Table 1, Table 2). There have been three
randomized, double-blind, placebo-controlled, 52-week, phase III trials completed with
tralokinumab in patients with AD: ECZTRA 1 (NCT03131648), ECZTRA 2 (NCT03160885),
and ECZTRA 3 (NCT03363854). ECZTRA 1 and 2 included 807 and 794 patients with AD,
respectively. Patients were randomized to receive subcutaneous tralokinumab monother-
apy with a 600 mg loading dose followed by 300 mg every other week (Q2W) or a placebo
for 16 weeks [
68
]. The eligibility criteria included patients with AD suitable for systemic
therapy with an Investigator’s Global Assessment (IGA) score of
3, an EASI score of
12,
a body surface area (BSA) of
10%, and a pruritus numeric rating score (NRS) of
4 [
68
].
In both trials, tralokinumab showed superiority to the placebo in reaching the co-primary
endpoints of achieving an IGA of 0 or 1 and an EASI of 75 by week 16 of treatment [
68
].
In the ECZTRA 1 trial, 15.8% of patients treated with tralokinumab achieved IGA scores
of 0 or 1, compared to 7.1% of patients treated with a placebo (p= 0.002). EASI 75 was
achieved in 25.0% of patients treated with tralokinumab compared to 12.7% of patients
receiving a placebo (p= 0.001) [
68
]. Similarly, in the ECZTRA 2 trial, significantly more
patients treated with tralokinumab achieved IGA 0 or 1 (22.2% vs. 10.9%, p< 0.001) and
EASI 75 (33.2% vs. 11.4%, p< 0.001) compared to placebo [
68
]. Finally, a significantly larger
proportion of patients achieved EASI 50 or EASI 90 in the tralokinumab group compared
to the placebo group by week 16 [68].
Patients treated with tralokinumab who achieved primary endpoints by week 16 were
re-randomized into three groups for the following 36 weeks: tralokinumab 300 mg Q2W,
tralokinumab 300 mg Q4W, and placebo [
68
]. The patients that had a clinical response
to placebos were maintained on placebos; however, they were not incorporated into the
analysis after week 16 [
68
]. Patients that did not obtain a clinical response with placebos
were treated with open-label tralokinumab 300 mg Q2W with optional topical corticos-
teroids [
68
]. If patients experienced a loss of effect between weeks 16 and 52, they were
switched to the open-label tralokinumab group [
68
]. Rescue treatment was used to con-
trol unbearable symptoms, and patients continued in the open-label or randomized arm;
however, they were considered non-responders in the final analysis [
68
]. By week 52, in
the ECZTRA 1 trial, there were no significant differences observed in the percentage of
patients reaching IGA scores of 0 or 1 without rescue medication (51% tralokinumab Q2W
vs. 47% placebo, p= 0.68) or in the percentage of patients maintaining or reaching EASI
75 (60% tralokinumab Q2W vs. 33% placebo, p= 0.056) [
68
]. Responses in the IGA and
EASI 75 in the tralokinumab group improved further at week 52 compared to week 16,
suggesting peak efficacy occurs at a later time point [
68
]. In the ECZTRA 2 trial, IGA
scores of 0 or 1 were reported in 59% of patients that continued tralokinumab Q2W treat-
ment compared, to 25% that were re-randomized from tralokinumab Q2W to the placebo
(p= 0.004) [
68
]. Similarly, an EASI 75 score was maintained in 56% of patients continu-
ing tralokinumab Q2W treatment and 21% of patients that were switched to the placebo
(p= 0.001) [
68
]. The long-term outcomes of tralokinumab treatment showed greater differ-
ences in the tralokinumab group compared to the placebo group in the ECZTRA 2 trial,
which could be due to the greater use of topical corticosteroids in ECZTRA 1 by 35.8% of
patients compared to 22.8% of patients in the ECZTRA 2 trial [
68
]. Patients treated with
Q4W tralokinumab had lower frequencies of IGA 0 or 1 and EASI 75 at 52 weeks compared
to the Q2W treatment schedule in both the ECZTRA 1 and ECZTRA 2 trials [
68
]. Greater
improvements in eczema-related sleep loss and pruritus were reported in the tralokinumab
group compared to the placebo group in both trials in the secondary endpoint analysis [
68
].
The tralokinumab group had a greater proportion of patients with pruritus NRS reductions
of
4 (ECZTRA 1: 20.0% vs. 10.3%, p= 0.002; ECZTRA 2: 25.0% vs. 9.5%, p< 0.001),
reductions in SCORAD (ECZTRA 1:
25.2 vs. 14.7, p< 0.001; ECZTRA 2:
28.1 vs.
14.0,
p< 0.001), and reductions in the Dermatology Life Quality Index (DLQI) (ECZTRA 1:
7.1 vs. 5.0, p= 0.002; ECZTRA 2: 8.8 vs. 4.9, p< 0.001) [68].
Pharmaceutics 2023,15, 568 5 of 18
Table 1.
Summary of primary and select secondary outcomes for phase II clinical trials completed with tralokinumab treatment in patients with moderate-to-severe
atopic dermatitis.
Clinical Trial Eligibility
(Sample Size)
Treatment Groups
(Duration) EASI EASI 50 EASI 75 IGA 0 or 1 SCORAD 50 NRS DLQI
Tralokinumab
NCT02347176
[69]
18–75 years
SCORAD 25
EASI 12
BSA 10% IGA 3
(N = 204)
45 mg tralokinumab
Q2W + TCS
(12 weeks)
10.78
(p= 0.143) 54.3% N/A 11.6%
(p= 0.974) 26.9% 1.80 N/A
150 mg tralokinumab
Q2W + TCS
(12 weeks)
15.14
(p= 0.027) 67.3% N/A 19.5%
(p= 0.281) 44.2% 1.59 N/A
300 mg tralokinumab
Q2W + TCS
(12 weeks)
15.72
(p= 0.011)
73.4%
(p= 0.03)
42.5%
(p= 0.003)
26.7%
(p= 0.061) 44.1% 2.17 N/A
Placebo + TCS
(12 weeks) 10.78 51.9% 15.5% 11.8% 19.5% 1.03 N/A
Lebrikizumab
TREBLE
(NCT02340234)
[70]
18–75 years
EASI 14
BSA 10% IGA 3
Pruritis VAS 3
(N = 212)
Lebrikizumab 125 mg
single dose N/A 69.2% 38.5% 21.2% N/A N/A N/A
Lebrikizumab 250 mg
single dose N/A 69.5% 49.1% 28.3% N/A N/A N/A
Lebrikizumab 125 mg
Q4W + TCS
(12 weeks)
N/A 82.4%
(p= 0.026)
54.9%
(p= 0.036) 33.3% N/A N/A N/A
Placebo + TCS
(12 weeks) N/A 62.3% 34.0% 18.9% N/A N/A N/A
Pharmaceutics 2023,15, 568 6 of 18
Table 1. Cont.
Clinical Trial Eligibility
(Sample Size)
Treatment Groups
(Duration) EASI EASI 50 EASI 75 IGA 0 or 1 SCORAD 50 NRS DLQI
NCT03443024
[26]
18–75 years
EASI 16
BSA 10% IGA 3
(N = 280)
Lebrikizumab 250 mg
loading dose + 125 mg
Q4W (16 weeks)
62.34%
(p= 0.0165)
66.4%
(p= 0.0554)
43.3%
(p= 0.0610)
26.6%
(p= 0.1917) N/A 35.94% (p
= 0.0047) N/A
Lebrikizumab 500 mg
loading dose + 250 mg
lebrikizumab Q4W
(16 weeks)
69.21%
(p= 0.0022)
77.0%
(p= 0.0037)
56.1%
(p= 0.0021)
33.7%
(p= 0.0392) N/A 49.6%
(p= 0.0002) N/A
Lebrikizumab 500 mg
loading dose + 250 mg
lebrikizumab Q2W
(16 weeks)
72.09%
(p= 0.0005)
81.0%
(p= 0.0008)
60.6%
(p= 0.0005)
44.6%
(p= 0.0023) N/A 60.63% (p
< 0.0001) N/A
Placebo (16 weeks) 41.12% 45.8% 24.3% 15.3% N/A 4.26 N/A
EASI: Eczema Area and Severity Index; IGA: Investigator’s Global Assessment; N/A: not applicable; NRS: numerical rating scale; TCS: topical corticosteroid. p-values are all compared
to the respective placebo group.
Table 2.
Summary of primary and select secondary outcomes for phase III clinical trials completed with tralokinumab and lebrikizumab treatment in patients with
moderate-to-severe atopic dermatitis.
Clinical Trial Eligibility
(Sample Size)
Treatment Groups
(Duration) EASI EASI 90 EASI 75 IGA 0 or 1 SCORAD
Change NRS Change DLQI
Change
Tralokinumab
ECZTRA 1
(NCT03131648)
18 years
BSA 10%
(N = 802)
Tralokinumab 300 mg
Q2W (16 weeks)
15.5
(p< 0.001)
14.5%
(p< 0.001)
25.0%
(p< 0.001)
15.8%
(p= 0.002)
25.2
(p< 0.001)
2.6
(p< 0.001)
7.1
(p= 0.002)
Placebo (16 weeks) 9.0 4.1% 12.7% 7.1% 14.7 1.7 5.0
Tralokinumab 300 mg
Q2W (52 weeks) N/A N/A 59.6%
(p= 0.056)
51.3%
(p= 0.68) N/A N/A N/A
Tralokinumab 300 mg
Q4W (52 weeks) N/A N/A 49.1%
(p= 0.27)
38.95%
(p= 0.50) N/A N/A N/A
Placebo (52 weeks) N/A N/A 33.3% 47.4% N/A N/A N/A
Pharmaceutics 2023,15, 568 7 of 18
Table 2. Cont.
Clinical Trial Eligibility
(Sample Size)
Treatment Groups
(Duration) EASI EASI 90 EASI 75 IGA 0 or 1 SCORAD
Change NRS Change DLQI
Change
ECZTRA 2
(NCT03160885)
18 years
BSA 10%
(N = 794)
Tralokinumab 300 mg
Q2W (16 weeks)
16.9
(p< 0.001)
18.3%
(p< 0.001)
33.2%
(p< 0.001)
22.2%
(p< 0.001)
28.1
(p< 0.001)
2.9
(p< 0.001)
8.8
(p< 0.001)
Placebo (16 weeks) 7.0 5.5% 11.4% 10.9% 14.0 1.6 4.9
Tralokinumab 300 mg
Q2W (52 weeks) N/A N/A 55.8%
(p< 0.001)
59.3%
(p= 0.004) N/A N/A N/A
Tralokinumab 300 mg
Q4W (52 weeks) N/A N/A 51.4%
(p= 0.001)
44.9%
(p= 0.084) N/A N/A N/A
Placebo (52 weeks) N/A N/A 21.4% 25.0% N/A N/A N/A
ECZTRA 3
(NCT03363854)
18 years
BSA 10%
(N = 380)
Tralokinumab 600 mg
loading dose + 300 mg
Q2W + TCS
(16 weeks)
21.0
(p< 0.001)
32.9%
(p= 0.022)
56.0%
(p< 0.001)
38.9%
(p= 0.015)
37.7
(p< 0.001)
4.1
(p< 0.001)
11.7
(p< 0.001)
Placebo + TCS
(16 weeks) 15.6 21.4% 35.7% 26.2% 26.8 2.9 8.8
Tralokinumab 600 mg
loading dose + 300 mg
Q2W + TCS
(32 weeks)
N/A N/A 92.5% 89.6% N/A N/A N/A
Tralokinumab 600 mg
loading dose + 300 mg
Q4W + TCS
(32 weeks)
N/A N/A 90.8% 77.6% N/A N/A N/A
Pharmaceutics 2023,15, 568 8 of 18
Table 2. Cont.
Clinical Trial Eligibility
(Sample Size)
Treatment Groups
(Duration) EASI EASI 90 EASI 75 IGA 0 or 1 SCORAD
Change NRS Change DLQI
Change
Lebrikizumab
ADVOCATE 1
(NCT04146363)
12 years
EASI 16
BSA 10% IGA 3
(N = 424)
Lebrikizumab 500 mg
loading dose + 250 mg
Q2W (16 weeks)
64.75%
(p< 0.001)
38.2%
(p< 0.001)
59.3%
(p< 0.001)
43.0%
(p< 0.001)
47.26
(p< 0.001)
45.75%
(p< 0.001)
8.78
(p< 0.001)
Placebo (16 weeks) 26.16% 9.1% 16.4% 12.8% 16.79 15.24% 2.94
Lebrikizumab Q2W
(52 weeks) N/A N/A 79.2%
(p< 0.05)
75.8%
(p< 0.001) N/A N/A N/A
Lebrikizumab Q4W
(52 weeks) N/A N/A 77.4%
(p< 0.05)
74.2%
(p< 0.001) N/A N/A N/A
Lebrikizumab
Withdrawal
(52 weeks)
N/A N/A 61.3% 46.5% N/A N/A N/A
ADVOCATE 2
(NCT04178967)
12 years
EASI 16
BSA 10% IGA 3
(N = 445)
Lebrikizumab 500 mg
loading dose + 250 mg
Q2W (16 weeks)
60.61%
(p< 0.001)
30.2%
(p< 0.001)
50.8%
(p< 0.001)
33.1%
(p< 0.001)
43.85%
(p< 0.001)
35.7%
(p< 0.001)
6.99%
(p< 0.001)
Placebo
(16 weeks) 28.22% 9.4% 18.2% 10.9% 13.87% 8.91% 2.47%
Lebrikizumab Q2W
(52 weeks) N/A N/A 79.2%
(p< 0.05)
64.6%
(p< 0.001) N/A N/A N/A
Lebrikizumab Q4W
(52 weeks) N/A N/A 84.7%
(p< 0.05)
80.6%
(p< 0.001) N/A N/A N/A
Lebrikizumab
Withdrawal
(52 weeks)
N/A N/A 72.0% 49.8%
(p< 0.001) N/A N/A N/A
ADhere
(NCT04250337)
12 years
EASI 16
BSA 10% IGA 3
(N = 228)
Lebrikizumab 500 mg
loading dose + 250 mg
Q2W + TCS
(16 weeks)
76.76%
(p< 0.001)
41.2%
(p= 0.008)
69.5%
(p< 0.001)
41.2%
(p= 0.011)
55.04%
(p< 0.001)
50.68%
(p= 0.017263)
9.79
(p= 0.001031)
Placebo + TCS
(16 weeks) 53.12% 21.7% 42.2% 22.1% 37.35% 35.47% 6.46
EASI: Eczema Area and Severity Index; IGA: Investigator’s Global Assessment; N/A: not applicable; NRS: numerical rating scale; TCS: topical corticosteroid. p-values are all compared
to the respective placebo group.
Pharmaceutics 2023,15, 568 9 of 18
ECZTRA 3 (NCT03363854) was another randomized, double-blind, placebo-controlled,
52-week, phase III trial designed to assess the efficacy and safety of tralokinumab in com-
bination with topical corticosteroids (TCS) in patients with moderate-to-severe AD [
71
].
This trial included 380 patients with AD for
1 year with an unsatisfactory response to
topical therapies or with a history of systemic medication therapy in the past year [
71
].
Eligibility criteria were similar to the monotherapy studies. Patients were provided with
TCS throughout the study to use as needed and were randomized into a 16-week treat-
ment with tralokinumab 300 mg Q2W after a 600 mg loading dose or a placebo [
71
].
At 16 weeks, significantly more tralokinumab + TCS-treated patients achieved an IGA
score of 0 or 1 (38.9% vs. 26.2%, p= 0.015) and an EASI 75 response (56.0% vs. 35.7%,
p< 0.001
) compared to patients receiving placebo + TCS [
71
]. Tralokinumab + TCS was
also superior to
placebo + TCS
in EASI 50, EASI 90, DLQI, and pruritus NRS [
71
]. Patients
in the tralokinumab-treated group required about 50% less cumulative TCS and rescue
medications compared to the placebo group (p= 0.004) [71].
Similar to the ECZTRA 1 and 2 trials, if patients obtained a clinical response of IGA
0 or 1 or EASI 75 at week 16 (responders) in ECZTRA 3, they were re-randomized to
either continue tralokinumab 300 mg Q2W or receive tralokinumab 300 mg Q4W for
another 16 weeks [
71
]. The remaining patients who did not achieve a clinical response
(non-responders) in the placebo or tralokinumab groups initiated tralokinumab Q2W [
71
].
All patients continued to receive TCS as needed throughout the trial [
71
]. By week 32,
89.6% of patients treated with tralokinumab Q2W maintained IGA scores of 0 or 1, and
92.5% maintained the EASI 75 response without needing rescue therapy [
71
]. The response
was maintained in the tralokinumab Q4W group in 77.6% and 90.8% of patients for IGA
0 or 1 and EASI 75, respectively [
71
]. There was no significant increase in TCS use in either
treatment group [
71
]. An EASI 90 response, which was observed in approximately 60% of
week 16 responders, was improved over the 32 weeks of treatment and was observed in
72.5% and 63.8% of patients treated with Q2W and Q4W tralokinumab, respectively [
71
].
Furthermore, a reduction of
4 points in the NRS pruritus score was achieved by 45.4%
of the tralokinumab + TCS patients compared to 34.1% of patients in the placebo + TCS
group (p< 0.001), as well as an improvement in the total DLQI score (
11.7 vs.
8.8,
respectively, p< 0.001), and SCORAD scores (37.7 vs. 26.8, respectively, p< 0.001) [71].
Patients who were non-responders at week 16 continued to improve with tralokinumab
Q2W, with 30.5% more responders for IGA 0 or 1 and 55.8% more responders for EASI
75 at week 32 [
71
]. A post hoc analysis of data pooled from all patients on tralokinumab
Q2W, independent from the response achieved at week 16, showed that at week 32, the
EASI 50 response was maintained from week 16 to 32 in 81% of patients, while EASI 75 and
EASI 90 rates progressively increased over time to 70.2% and 50.4%, respectively [
72
].
Finally, preliminary results of the ECZTEND trial were recently presented at the 2022
Annual Meeting of the American Academy of Dermatology. In this trial, a longer-term,
2-year continuous treatment with tralokinumab Q2W and optional TCS was studied in
1442 patients with AD [
73
]. A high level of response rates was sustained after 2 years of
treatment, where 85.1% of treated patients achieved EASI 75, 65% achieved EASI 90, 50.5%
had clear or almost clear skin measured via the IGA, 60.6% had pruritus NRS
3, and
76.4% had DLQI 5 [73].
5.3. Safety of Tralokinumab in Patients with Atopic Dermatitis
In the phase III trials ECZTRA 1 and ECZTRA 2, tralokinumab exhibited a similar
safety profile to the phase IIb study over 16 and 52 weeks of treatment, with a similar
incidence of mild to moderate adverse events in the tralokinumab and placebo groups. In
ECZTRA 1, 23% of patients treated with tralokinumab reported upper respiratory tract
infections (URTI; vs. 21% in the placebo group), and 7% of patients reported conjunctivitis
(vs. 2% in the placebo group) [
68
]. Similarly, in the ECZTRA 2 trial, 10% of patients
treated with tralokinumab reported URTI (vs. 9% in the placebo group) and 3% of patients
reported conjunctivitis (vs. 2% in the placebo group) [
68
]. All cases of conjunctivitis
Pharmaceutics 2023,15, 568 10 of 18
were mild and resolved by the end of the treatment period, except for one patient who
discontinued the study treatment [
68
]. In contrast, worsening of AD, incidents of eczema
herpeticum, and skin infections were more frequently seen in the placebo group [
68
]. In
the maintenance period, AEs occurred more frequently in the tralokinumab Q2W group
than in the tralokinumab Q4W group, with a low number of events leading to permanent
interruption [
68
]. The safety profile, including the frequency and severity of adverse events
that emerged from the ECZTRA 3 trial, was comparable with that of the ECZTRA 1 and
2 trials. The tralokinumab safety profile did not appear to differ with the addition of
a TCS. In ECZTRA 3, URTI was again the most common AE in 19% of patients treated
with tralokinumab (vs. 11% in the placebo group), followed by conjunctivitis in 11% of
tralokinumab-treated patients (vs. 3% in the placebo group) [
71
]. Headache was reported in
9% of tralokinumab patients compared to 5% in the placebo group [
71
]. Skin infections that
required systemic management were more frequent in the placebo group [
71
]. AEs were
less frequent and less severe in the tralokinumab Q4W group compared to the tralokinumab
Q2W groups [
71
]. Four patients discontinued due to adverse events; however, none of these
AEs were severe: two were due to AD worsening, one was due to herpetic eczema, and
one was due to a prostate cancer diagnosis [
71
]. A total of 13 serious adverse events (SAEs)
were recorded, with no difference between groups [
71
]. A recently presented analysis from
ECZTRA 1, 2, 3, 5, and phase IIb trials of safety data pooled from patients with AD treated
with tralokinumab Q2W with TCS for 16 weeks showed a greater proportion of patients
experiencing conjunctivitis compared to placebo groups (5.4% vs. 1.9%) and a similar rate of
all adverse events (65.7% vs. 67.2%, respectively), serious AEs (
2.1% vs. 2.8%
, respectively),
mild AEs (53.2% vs. 49.0%, respectively), moderate AEs (31.5% vs. 39.0%, respectively),
severe AEs (4.6% vs. 6.3%, respectively), and AEs leading to drug withdrawal (2.3% vs.
2.8%, respectively) [
73
]. The most frequently reported adverse events in the pooled data
were viral URTIs (15.7% vs. 12.2%, respectively) and AD (15.4% vs. 26.2%, respectively) [
73
].
Finally, the recently presented safety analysis set from the 2-year ECZTEND trial showed
78.2% of patients reporting AEs, with 7.0% being serious AEs, 66.3% being mild, 46.3%
being moderate, and 7.1% being severe [
73
]. Only 2.4% of patients reported AEs that
led to drug withdrawal. The most frequently reported AEs were viral URTI in 20.5% of
patients, AD in 17.8% of patients, and conjunctivitis in 5.3% of patients; the proportion of
patients experiencing the latter was similar to previously reported trials [
73
]. Although
tralokinumab use is associated with an increased incidence of conjunctivitis, cases reported
in clinical trials with 16 weeks of treatment were mostly mild and transient [74].
6. Use of Lebrikizumab to Manage Patients with Atopic Dermatitis
6.1. Overview of Lebrikizumab
Lebrikizumab is a humanized monoclonal antibody that binds soluble IL-13 at the
non-receptor binding domain with a high affinity [
18
]. A bound IL-13 is able to form a
complex with IL-13R
α
1; however, it prevents heterodimerization with IL-4R
α
and prevents
signal transduction [
18
]. Therefore, lebrikizumab inhibits the IL-4R
α
–IL-13R
α
1 signal-
ing complex while continuing to regulate endogenous IL-13 via stimulation of IL-13R
α
2
(Figure 1) [
75
]. Pharmacokinetic data for lebrikizumab are available from a large meta-
analysis of 11 studies, which pooled data from 2148 patients receiving either 37.5 mg or
125 mg of lebrikizumab Q4W [
76
]. The bioavailability of lebrikizumab was 85.6% with a
0.156 L/day clearance, and an elimination half-life of 25.7 days [76].
6.2. Clinical Efficacy of Lebrikizumab in Patients with Atopic Dermatitis
Results from the first induction period of two monotherapy phase III studies for
lebrikizumab management of AD have recently been presented and are consistent with
those observed in the phase II trials (Table 1, Table 2) [
77
]. ADVOCATE 1 (NCT04146363)
and ADVOCATE 2 (NCT04178967) are randomized, double-blind, placebo-controlled,
parallel-group, 52-week trials that included 851 patients aged 12 years or older with
moderate-to-severe AD, with inadequate responses to topical treatments, IGA scores of
Pharmaceutics 2023,15, 568 11 of 18
3, and who were naïve to dupilumab and tralokinumab treatments [
77
]. Patients were
randomized to receive a lebrikizumab 500 mg loading dose followed by 250 mg Q2W
or placebo [
77
]. After 16 weeks of treatment in the ADVOCATE 1 trial, the co-primary
endpoint IGA response of 0 or 1 was reached in 43.0% of patients in the lebrikizumab
group compared to 12.8% in the placebo group (p< 0.001), and EASI 75 in 59.3% of patients
compared to 16.4%, respectively (p< 0.001) [
77
]. Similarly, after 16 weeks of treatment
in the ADVOCATE 2 trial, 33.1% of patients in the lebrikizumab group reached IGA
0 or 1 compared to 10.9% in the placebo group (p< 0.001), and EASI 75 was reached
by 50.8% compared to 18.2%, respectively (p< 0.001) [
77
]. In both trials, a significantly
greater proportion of patients achieved secondary endpoints in the lebrikizumab treatment
group compared to the placebo group, such as EASI 90 (ADVOCATE 1: 38.2% vs. 9.1%,
respectively, p< 0.001; ADVOCATE 2: 30.2% vs. 9.4%, respectively, p< 0.001), pruritus
NRS
4 point improvement (ADVOCATE 1: 46.3% vs. 12.7%, respectively, p< 0.001;
ADVOCATE 2: 38.3% vs. 11.3%, respectively, p< 0.001), sleep-loss scale score
4 point
improvement (ADVOCATE 1: 38.7% vs. 5.1%, respectively, p< 0.001; ADVOCATE 2:
26.5% vs. 7.8%, respectively, p< 0.001), and DLQI
4 point improvement (ADVOCATE 1:
75.5% vs. 33.8%, respectively, p< 0.001; ADVOCATE 2: 64.4% vs. 34.6%, respectively,
p< 0.001) [77].
After week 16, patients were re-randomized to either continue receiving 250 mg lebrik-
izumab Q2W or receive 250 mg lebrikizumab Q4W or a placebo for another 36 weeks [
78
].
Participants that required rescue therapy in the first 16 weeks or did not maintain an
EASI
50 after 16 weeks of treatment received open-label lebrikizumab Q2W for the
next 36 weeks [
78
]. The maintenance period (weeks 16 to 52) of the ADVOCATE 1 and
ADVOCATE 2 trials has been completed and the preliminary results were presented at
the European Academy of Dermatology and Venereology 2022 meeting [
78
]. The efficacy
achieved by week 16 with the lebrikizumab treatment was reported to be maintained at
52 weeks [
78
]. By week 52 of treatment, clear or almost clear skin measured via IGA was
achieved in a greater proportion of patients treated with Q2W and Q4W lebrikizumab
regimens compared to the withdrawal group (ADVOCATE 1: 75.8% and 74.2% vs. 46.5%,
respectively; ADVOCATE 2: 64.6% and 80.6% vs. 49.8%, respectively) [
78
]. EASI 75 was
maintained in a greater proportion of patients treated with Q2W and Q4W lebrikizumab
regimens compared to the withdrawal group (ADVOCATE 1: 79.2% and 79.2% vs. 61.3%,
respectively; ADVOCATE 2: 77.4% and 84.7% vs. 72.0%, respectively) [
78
]. Overall, 81.2%
and 90.3% of patients treated with lebrikizumab Q2W in ADVOCATE 1 and 2, respectively,
and 80.4% and 88.1% in the Q4W group achieved a
4-point improvement in pruritus NRS
compared to 65.4% and 67.6% of patients in the lebrikizumab withdrawal group [78].
ADhere (NCT04250337) is a randomized, double-blind, parallel-group, placebo-controlled
combination trial that recruited 228 adolescents and adults aged 12 years or older with
moderate-to-severe AD for 1 or more years with inadequate responses to topical or systemic
treatment, an IGA score of
3, an EASI score
16, and a BSA
10 [
79
]. Patients were
randomized to receive a lebrikizumab 500 mg loading dose followed by 250 mg Q2W or a
placebo Q2W, where both treatment groups were treated with concomitant TCS [
79
]. After
16 weeks of treatment, the co-primary endpoint IGA response of 0 or 1 was reached in 41.2%
of patients in the lebrikizumab + TCS group compared to 22.1% in the placebo + TCS group
(p= 0.011), and EASI 75 was reached in 69.5% compared to 42.2% of patients, respectively
(p< 0.001) [
79
]. Similarly, a significantly greater proportion of patients achieved secondary
endpoints in the lebrikizumab + TCS treatment group compared to the
placebo + TCS
group, such as EASI 90 (41.2% vs. 21.7%, respectively, p= 0.008), pruritus
NRS 4
point
improvement (50.6% vs. 31.9%, respectively, p= 0.017), percent improvement in pruri-
tus NRS (50.68% vs. 35.47%, respectively, p= 0.017), and DLQI
4 point improvement
(77.4% vs. 58.7%, respectively, p< 0.036) [79].
Pharmaceutics 2023,15, 568 12 of 18
6.3. Safety of Lebrikizumab in Patients with Atopic Dermatitis
Lebrikizumab is well tolerated, with a similar safety profile reported in phase II and
phase III trials to date. While only preliminary long-term data are available from the phase
III trials, lebrikizumab safety data appear to be similar to that of a placebo when treated
for up to 16 weeks [
77
]. In the ADVOCATE 1 and 2 trials, the proportion of patients
experiencing AEs was similar in the lebrikizumab-treated and placebo groups, with the
majority of AEs being mild or moderate in severity (ADVOCATE 1: 45.4% vs. 51.5%,
respectively; ADVOCATE 2: 53.0% vs. 66.2%, respectively) [
77
]. The proportion of patients
reporting conjunctivitis in the treatment group appears to be greater compared to the
placebo group in both studies (ADVOCATE 1: 7.4% vs. 2.8%, respectively; ADVOCATE 2:
7.8% vs. 2.1%, respectively) [
77
]. However, all conjunctivitis treatment-emergent AEs were
mild-to-moderate in severity and did not lead to treatment discontinuation [
77
]. Infections
with herpes were similar among the treatment and placebo groups (ADVOCATE 1: 3.2%
vs. 4.3%, respectively; ADVOCATE 2: 2.8% vs. 4.1%, respectively) [
77
]. Preliminary data
at 52 weeks of treatment indicated that 58.1% and 68.1% of lebrikizumab-treated patients
in ADVOCATE 1 and 2, respectively, reported AEs, the majority of which were mild to
moderate in severity [
78
]. None of the severe AEs which were reported by 3.3% and 2.6%
of patients in ADVOCATE 1 and 2, respectively, were related to the study drug, as was
assessed by the study investigators [
78
]. The most common AEs in the lebrikizumab-treated
patients in ADVOCATE 1 and 2 were AD (7.8 and 10.1%, respectively), nasopharyngitis
(6.8% and 9.6%, respectively), conjunctivitis (8.3% and 8.1%, respectively), herpes infections
(5.0 and 4.8%, respectively), and skin infections (3.0% and 4.9%, respectively) [78].
ADhere phase III study safety data are only available up to week 28 of treatment, with
43.1% of patients in the lebrikizumab group and 34.7% of patients in the placebo group
reporting AEs [
79
]. Conjunctivitis was reported in 4.6% of patients in the treatment arm
and none in the placebo arm to date [
79
]. Safety data from the maintenance phases of both
phase III trials will be important to improve our understanding of the safety profile of
lebrikizumab longer term [
79
]. Nevertheless, the safety data available to date in patients
with AD are consistent with those previously reported in several asthma trials of over
2000 patients since 2011 [8084].
6.4. Future IL-13 Inhibitors
Eblasakimab and cendakimab are two more IL-13 inhibitors being developed in the
pipeline and are currently being investigated in phase II trials (NCT04800315). Eblasakimab
is a monoclonal antibody that targets the IL-13R
α
1, a subunit of the type 2 receptor,
interfering with the signaling of IL-13 and IL-4 (Figure 1). Early data presented at the
Annual Meeting of the American Academy of Dermatology 2022 showed significant efficacy
after 8 weeks of treatment [
85
]. These were preliminary reported data in a small sample
size of patients with AD that were randomized to receive a placebo (n = 16) or eblasakimab
at one of three doses: 200 mg (n = 4), 400 mg (n = 7), or 600 mg (n = 22) [
85
]. A significant
reduction in EASI was reported: 50% in the 200 mg group, 63% in the 400 mg group, and
61% in the 600 mg group, as opposed to 32% in the placebo group (p= 0.023) [
85
]. EASI
50 was achieved in 50% of patients in the 200 mg group, 71% in the 400 mg group, and
77% in the 600 mg group, compared to 38% in the placebo group (p= 0.016) [
85
]. EASI
75 was achieved in 50% of patients in the 200 mg group, 57% in the 400 mg group, and 50%
in the 600 mg group, as opposed to 13% in the placebo group (p= 0.018) [
85
]. Finally, the
peak pruritus NRS decreased by 37% in the group of patients treated with eblasakimab
600 mg, which was a significant improvement in comparison to the 16% decrease in the
group treated with placebos (p= 0.032) [
85
]. Although eblasakimab shows great promise in
efficacy for the treatment of AD, this preliminary study is limited by a lack of available long-
term and safety data as well as a small sample size. Thus, further trial data for eblasakimab
and cendakimab are awaited.
Pharmaceutics 2023,15, 568 13 of 18
7. Discussion
For many decades, conventional therapy for AD comprised topical and oral immuno-
suppression as well as phototherapy. Approval of dupilumab triggered a significant leap
forward into an era of rapid development of targeted biological therapy for AD manage-
ment. The next targeted therapies approved were the JAK inhibitors abrocitinib, baricitinib,
and upadacitinib, as well as an IL-13 inhibitor, tralokinumab. It is important to carefully
study each biological and small molecule class to define how the nuanced differences can
be used to benefit each individual patient.
Overall, growing evidence suggests that targeted IL-13 inhibitors offer a great advan-
tage in developing efficacious and safe management strategies for patients with moderate-
to-severe AD. The two most studied IL-13 inhibitors, tralokinumab and lebrikizumab,
were shown to be efficacious in phase III trials, with the response being maintained over
time. Treatment with these agents also results in significant improvements in pruritus and
quality of life. The detailed mechanisms of IL-13 inhibition in tralokinumab, lebrikizumab,
and eblasakimab are distinct (Figure 1). Tralokinumab binds IL-13 in the IL-13R
α
bind-
ing site and blocks IL-13 interaction with both IL- 13R
α
1 and IL-13R
α
2. Lebrikizumab
binds IL-13 at the IL-4R
α
- binding site and inhibits IL-4R
α
and IL-13R
α
1 receptors’ sig-
naling. It is unknown whether differences in the mechanisms of IL-13 inhibition result
in clinical implications, given that there are no available direct comparison studies and
different study design, duration, and TCS use are present in the available phase III studies.
While the mechanistic differences between tralokinumab and lebrikizumab are more subtle,
dupilumab differs by targeting the IL-4R
α
receptor subunit and inhibiting the binding of
both IL-4 and IL13. Thus, results of current clinical programs investigating these agents
will help elucidate important new insights into the roles of IL-13, IL-13R
α
2, IL-13R
α
1, type
1 and type 2 receptors in the regulation of inflammatory skin conditions. Moreover, direct
head-to-head comparison trials are needed to elucidate the differences in the efficacy of
dupilumab, selective IL-13 and JAK 1 inhibitors. While these targeted therapies offer a
great advantage for an improved efficacy and safety profile, there remains a proportion of
patients that do not respond to treatment, which is likely a consequence of the heterogeneity
in the pathogenesis of AD. Thus, the identification of biomarkers that help predict the
response to treatment is an area of unmet need. For example, in phase II tralokinumab
trials, periostin and DPP-4 were used as biomarkers for IL-13 activity and were shown to
be associated with a response to treatment [
69
]. Further development of such biomarkers
will help advance the era of personalized medicine. Finally, despite all the benefits of
targeted therapy that were discussed, there remain challenges with subcutaneous injection
administration of biological agents for patients with needle phobia, the need for cold chain
delivery, and the long half-life of these agents.
In terms of differences in safety among different biologic agents and small molecule
therapies, JAK inhibitors have a class warning for thrombosis, major cardiovascular events,
and malignancies that have not been reported in trials of dupilumab and selective IL-13
inhibitors. In addition, JAKs have a side effect profile that includes an increased risk of
infections, such as viral infections including eczema herpeticum and herpes zoster [
86
].
Although head-to-head data comparing dupilumab to JAK inhibitors have been pub-
lished, there are no direct head-to-head comparison trials available with the current or
emerging IL-13 inhibitors. In phase III trials, all agents that inhibit IL-13, specifically
dupilumab, tralokinumab, lebrikizumab, cendakimab, and eblasakimab, have been re-
ported to have a considerably increased risk of conjunctivitis, injection site reactions, and
head and neck erythema. Interestingly, there appears to be a larger risk of conjunctivitis
with dupilumab (up to 22% in phase III trials) compared to tralokinumab (~11%) or lebrik-
izumab (~3%) [
22
,
23
,
87
,
88
]. Real-world analyses and direct head-to-head comparison trials
are needed to confirm these observations. While the pathogenesis of conjunctivitis in this
setting is not completely understood, it is hypothesized to be the consequence of Demodex
mite proliferation, direct IL-13-mediated reduction in goblet cells, or OX40-related inflam-
mation [
87
,
88
]. Biopsies obtained from AD patients that developed conjunctivitis while
Pharmaceutics 2023,15, 568 14 of 18
being treated with dupilumab confirmed a substantial decrease in the number of intraep-
ithelial goblet cells [
89
]. Similar to dupilumab-associated conjunctivitis, this association
with selective IL-13 inhibitors could be initially managed with warm compresses, artificial
tears, sodium hyaluronate, or antihistamine drops while continuing the use of the biological
agent. A consultation with ophthalmology is warranted if a patient develops eye pain,
vision changes, purulent discharge, conjunctival scarring, or corneal involvement. Resistant
or severe conjunctivitis cases may require anti-inflammatory steroid drops, calcineurin
inhibitors, or cyclosporine [90].
8. Conclusions
Improving the knowledge of the complex inflammatory mechanisms involved in
AD pathogenesis has led to an increase in the use of targeted biological therapies for
the effective and safe management of this chronic skin condition. IL-13 is thought to
be the main mediator implicated in the inflammation, epidermal barrier dysfunction,
and pruritus associated with AD. Thus, selective IL-13 inhibitors, such as tralokinumab,
lebrikizumab, and eblasakimab, have shown good efficacy in the treatment of moderate-to-
severe AD. While these agents have favorable safety profiles, there remains an increased
risk of conjunctivitis, requiring monitoring. Although access to biological agents remains
to be a challenge for some patients, the emergence of these therapies significantly increased
the effective options available to manage patients with this skin condition. Further long-
term studies are ongoing to continue investigating how the use of IL-13 inhibitors can be
further utilized to therapeutically manage patients with moderate-to-severe AD.
Author Contributions:
Y.L. and M.G. conducted the search and wrote and reviewed the manuscript.
All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest:
Melinda Gooderham has been an investigator, speaker and/or advisor for:
abbvie, Amgen, Akros, Arcutis, Aristea, AnaptysBio, Bausch Health, BMS, Boehringer Ingelheim,
Celgene, Dermira, Dermavant, Eli Lilly, Galderma, GSK, Incyte, Janssen, Kyowa Kirin, LEO Pharma,
MedImmune, Merck, Moonlake, Meiji, Nimbus, Novartis, Pfizer, Regeneron, Reistone, Roche, Sanofi
Genzyme, Sun Pharma, and UCB. The company had no role in the design of the study; in the
collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to
publish the results.
References
1.
Deckers, I.A.; McLean, S.; Linssen, S.; Mommers, M.; van Schayck, C.P.; Sheikh, A. Investigating international time trends in the
incidence and prevalence of atopic eczema 1990-2010: A systematic review of epidemiological studies. PLoS ONE
2012
,7, e39803.
[CrossRef] [PubMed]
2.
Hay, R.J.; Johns, N.E.; Williams, H.C.; Bolliger, I.W.; Dellavalle, R.P.; Margolis, D.J.; Marks, R.; Naldi, L.; Weinstock, M.A.; Wulf,
S.K.; et al. The global burden of skin disease in 2010: An analysis of the prevalence and impact of skin conditions. J. Investig.
Dermatol. 2014,134, 1527–1534. [CrossRef] [PubMed]
3.
Nygaard, U.; Vestergaard, C.; Deleuran, M. Emerging Treatment Options in Atopic Dermatitis: Systemic Therapies. Dermatology
2017,233, 344–357. [CrossRef] [PubMed]
4.
Fabbrocini, G.; Napolitano, M.; Megna, M.; Balato, N.; Patruno, C. Treatment of Atopic Dermatitis with Biologic Drugs. Dermatol.
Ther. 2018,8, 527–538. [CrossRef]
5. Bieber, T. Atopic dermatitis. N. Engl. J. Med. 2008,358, 1483–1494. [CrossRef]
6.
Rønnstad, A.T.M.; Halling-Overgaard, A.S.; Hamann, C.R.; Skov, L.; Egeberg, A.; Thyssen, J.P. Association of atopic dermatitis
with depression, anxiety, and suicidal ideation in children and adults: A systematic review and meta-analysis. J. Am. Acad.
Dermatol. 2018,79, 448–456.e30. [CrossRef]
7. Renert-Yuval, Y.; Guttman-Yassky, E. What’s New in Atopic Dermatitis. Dermatol. Clin. 2019,37, 205–213. [CrossRef]
8.
Silverberg, J.I.; Kantor, R. The Role of Interleukins 4 and/or 13 in the Pathophysiology and Treatment of Atopic Dermatitis.
Dermatol. Clin. 2017,35, 327–334. [CrossRef]
Pharmaceutics 2023,15, 568 15 of 18
9.
Yaghmaie, P.; Koudelka, C.W.; Simpson, E.L. Mental health comorbidity in patients with atopic dermatitis. J. Allergy Clin. Immunol.
2013,131, 428–433. [CrossRef]
10.
Drucker, A.M.; Wang, A.R.; Li, W.Q.; Sevetson, E.; Block, J.K.; Qureshi, A.A. The Burden of Atopic Dermatitis: Summary of a
Report for the National Eczema Association. J. Investig. Dermatol. 2017,137, 26–30. [CrossRef]
11.
Ferreira, S.; Torres, T. Dupilumab for the Treatment of Atopic Dermatitis. Actas Dermosifiliogr. (Engl. Ed.)
2018
,109, 230–240.
[CrossRef] [PubMed]
12.
Lewis-Jones, S. Quality of life and childhood atopic dermatitis: The misery of living with childhood eczema. Int. J. Clin. Pract.
2006,60, 984–992. [CrossRef] [PubMed]
13.
Guttman-Yassky, E.; Thaçi, D.; Pangan, A.L.; Hong, H.C.; Papp, K.A.; Reich, K.; Beck, L.A.; Mohamed, M.F.; Othman, A.A.;
Anderson, J.K.; et al. Upadacitinib in adults with moderate to severe atopic dermatitis: 16-week results from a randomized,
placebo-controlled trial. J. Allergy Clin. Immunol. 2020,145, 877–884. [CrossRef]
14.
Ring, J.; Alomar, A.; Bieber, T.; Deleuran, M.; Fink-Wagner, A.; Gelmetti, C.; Gieler, U.; Lipozencic, J.; Luger, T.; Oranje, A.P.;
et al. Guidelines for treatment of atopic eczema (atopic dermatitis) part I. J. Eur. Acad. Dermatol. Venereol.
2012
,26, 1045–1060.
[CrossRef] [PubMed]
15. Roekevisch, E.; Spuls, P.I.; Kuester, D.; Limpens, J.; Schmitt, J. Efficacy and safety of systemic treatments for moderate-to-severe
atopic dermatitis: A systematic review. J. Allergy Clin. Immunol. 2014,133, 429–438. [CrossRef] [PubMed]
16.
Baghoomian, W.; Na, C.; Simpson, E.L. New and Emerging Biologics for Atopic Dermatitis. Am. J. Clin. Dermatol.
2020
,21,
457–465. [CrossRef]
17.
Silverberg, J.I. Atopic dermatitis treatment: Current state of the art and emerging therapies. Allergy Asthma Proc.
2017
,38, 243–249.
[CrossRef]
18.
Moyle, M.; Cevikbas, F.; Harden, J.L.; Guttman-Yassky, E. Understanding the immune landscape in atopic dermatitis: The era of
biologics and emerging therapeutic approaches. Exp. Dermatol. 2019,28, 756–768. [CrossRef] [PubMed]
19.
Silverberg, J.I.; Hanifin, J.M. Adult eczema prevalence and associations with asthma and other health and demographic factors: A
US population-based study. J. Allergy Clin. Immunol. 2013,132, 1132–1138. [CrossRef]
20. Bieber, T. Interleukin-13: Targeting an underestimated cytokine in atopic dermatitis. Allergy 2020,75, 54–62. [CrossRef]
21. Chang, H.Y.; Nadeau, K.C. IL-4RαInhibitor for Atopic Disease. Cell 2017,170, 222. [CrossRef] [PubMed]
22.
Simpson, E.L.; Bieber, T.; Guttman-Yassky, E.; Beck, L.A.; Blauvelt, A.; Cork, M.J.; Silverberg, J.I.; Deleuran, M.; Kataoka, Y.;
Lacour, J.P.; et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N. Engl. J. Med.
2016
,375, 2335–2348.
[CrossRef] [PubMed]
23.
Blauvelt, A.; de Bruin-Weller, M.; Gooderham, M.; Cather, J.C.; Weisman, J.; Pariser, D.; Simpson, E.L.; Papp, K.A.; Hong, H.C.;
Rubel, D.; et al. Long-term management of moderate-to-severe atopic dermatitis with dupilumab and concomitant topical
corticosteroids (LIBERTY AD CHRONOS): A 1-year, randomised, double-blinded, placebo-controlled, phase 3 trial. Lancet
2017
,
389, 2287–2303. [CrossRef] [PubMed]
24.
Thaçi, D.; Simpson, E.L.; Deleuran, M.; Kataoka, Y.; Chen, Z.; Gadkari, A.; Eckert, L.; Akinlade, B.; Graham, N.M.H.;
Pirozzi, G.; et al. Efficacy and safety of dupilumab monotherapy in adults with moderate-to-severe atopic dermatitis: A pooled
analysis of two phase 3 randomized trials (LIBERTY AD SOLO 1 and LIBERTY AD SOLO 2). J. Dermatol. Sci.
2019
,94, 266–275.
[CrossRef] [PubMed]
25.
Uchida, H.; Kamata, M.; Mizukawa, I.; Watanabe, A.; Agematsu, A.; Nagata, M.; Fukaya, S.; Hayashi, K.; Fukuyasu, A.;
Tanaka, T.; et al
. Real-world effectiveness and safety of dupilumab for the treatment of atopic dermatitis in Japanese patients: A
single-centre retrospective study. Br. J. Dermatol. 2019,181, 1083–1085. [CrossRef]
26.
Guttman-Yassky, E.; Blauvelt, A.; Eichenfield, L.F.; Paller, A.S.; Armstrong, A.W.; Drew, J.; Gopalan, R.; Simpson, E.L. Efficacy and
Safety of Lebrikizumab, a High-Affinity Interleukin 13 Inhibitor, in Adults With Moderate to Severe Atopic Dermatitis: A Phase
2b Randomized Clinical Trial. JAMA Dermatol. 2020,156, 411–420. [CrossRef]
27.
Soria, A.; Du-Thanh, A.; Seneschal, J.; Jachiet, M.; Staumont-Sallé, D.; Barbarot, S. Development or Exacerbation of Head and
Neck Dermatitis in Patients Treated for Atopic Dermatitis With Dupilumab. JAMA Dermatol. 2019,155, 1312–1315. [CrossRef]
28.
Tsoi, L.C.; Rodriguez, E.; Degenhardt, F.; Baurecht, H.; Wehkamp, U.; Volks, N.; Szymczak, S.; Swindell, W.R.; Sarkar, M.K.;
Raja, K.; et al
. Atopic Dermatitis Is an IL-13-Dominant Disease with Greater Molecular Heterogeneity Compared to Psoriasis.
J. Investig. Dermatol. 2019,139, 1480–1489. [CrossRef]
29.
Bao, K.; Reinhardt, R.L. The differential expression of IL-4 and IL-13 and its impact on type-2 immunity. Cytokine
2015
,75, 25–37.
[CrossRef]
30.
Oetjen, L.K.; Mack, M.R.; Feng, J.; Whelan, T.M.; Niu, H.; Guo, C.J.; Chen, S.; Trier, A.M.; Xu, A.Z.; Tripathi, S.V.; et al. Sensory
Neurons Co-opt Classical Immune Signaling Pathways to Mediate Chronic Itch. Cell 2017,171, 217–228.e13. [CrossRef]
31.
Napolitano, M.; Marasca, C.; Fabbrocini, G.; Patruno, C. Adult atopic dermatitis: New and emerging therapies. Expert Rev. Clin.
Pharm. 2018,11, 867–878. [CrossRef] [PubMed]
32. Misery, L.; Huet, F.; Gouin, O.; Ständer, S.; Deleuran, M. Current pharmaceutical developments in atopic dermatitis. Curr. Opin.
Pharm. 2019,46, 7–13. [CrossRef]
33.
Fujii, M. Current Understanding of Pathophysiological Mechanisms of Atopic Dermatitis: Interactions among Skin Barrier
Dysfunction, Immune Abnormalities and Pruritus. Biol. Pharm. Bull. 2020,43, 12–19. [CrossRef]
Pharmaceutics 2023,15, 568 16 of 18
34.
Furue, K.; Ito, T.; Tsuji, G.; Ulzii, D.; Vu, Y.H.; Kido-Nakahara, M.; Nakahara, T.; Furue, M. The IL-13-OVOL1-FLG axis in atopic
dermatitis. Immunology 2019,158, 281–286. [CrossRef]
35.
Zaniboni, M.C.; Samorano, L.P.; Orfali, R.L.; Aoki, V. Skin barrier in atopic dermatitis: Beyond filaggrin. Bras. Dermatol.
2016
,91,
472–478. [CrossRef]
36. Bonness, S.; Bieber, T. Molecular basis of atopic dermatitis. Curr. Opin. Allergy Clin. Immunol. 2007,7, 382–386. [CrossRef]
37.
Loh, T.Y.; Hsiao, J.L.; Shi, V.Y. Therapeutic Potential of Lebrikizumab in the Treatment of Atopic Dermatitis. J. Asthma Allergy
2020,13, 109–114. [CrossRef] [PubMed]
38.
Garcovich, S.; Maurelli, M.; Gisondi, P.; Peris, K.; Yosipovitch, G.; Girolomoni, G. Pruritus as a Distinctive Feature of Type 2
Inflammation. Vaccines 2021,9, 303. [CrossRef]
39.
Dubin, C.; Del Duca, E.; Guttman-Yassky, E. The IL-4, IL-13 and IL-31 pathways in atopic dermatitis. Expert Rev. Clin. Immunol.
2021,17, 835–852. [CrossRef]
40.
Gooderham, M.J.; Hong, H.C.; Eshtiaghi, P.; Papp, K.A. Dupilumab: A review of its use in the treatment of atopic dermatitis.
J. Am. Acad. Dermatol. 2018,78, S28–S36. [CrossRef]
41.
Omori-Miyake, M.; Yamashita, M.; Tsunemi, Y.; Kawashima, M.; Yagi, J.
In vitro
assessment of IL-4- or IL-13-mediated changes in
the structural components of keratinocytes in mice and humans. J. Investig. Dermatol. 2014,134, 1342–1350. [CrossRef]
42.
Howell, M.D.; Kim, B.E.; Gao, P.; Grant, A.V.; Boguniewicz, M.; DeBenedetto, A.; Schneider, L.; Beck, L.A.; Barnes, K.C.;
Leung, D.Y
. Cytokine modulation of atopic dermatitis filaggrin skin expression. J. Allergy Clin. Immunol.
2009
,124, R7–R12.
[CrossRef] [PubMed]
43.
Howell, M.D.; Fairchild, H.R.; Kim, B.E.; Bin, L.; Boguniewicz, M.; Redzic, J.S.; Hansen, K.C.; Leung, D.Y. Th2 cytokines act on
S100/A11 to downregulate keratinocyte differentiation. J. Investig. Dermatol. 2008,128, 2248–2258. [CrossRef] [PubMed]
44.
Berdyshev, E.; Goleva, E.; Bronova, I.; Dyjack, N.; Rios, C.; Jung, J.; Taylor, P.; Jeong, M.; Hall, C.F.; Richers, B.N.; et al. Lipid
abnormalities in atopic skin are driven by type 2 cytokines. JCI Insight 2018,3, e98006. [CrossRef] [PubMed]
45.
Agrawal, R.; Woodfolk, J.A. Skin barrier defects in atopic dermatitis. Curr. Allergy Asthma Rep.
2014
,14, 433. [CrossRef] [PubMed]
46.
Nomura, I.; Goleva, E.; Howell, M.D.; Hamid, Q.A.; Ong, P.Y.; Hall, C.F.; Darst, M.A.; Gao, B.; Boguniewicz, M.;
Travers, J.B.; et al
.
Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes.
J. Immunol. 2003,171, 3262–3269. [CrossRef] [PubMed]
47.
Nakatsuji, T.; Chen, T.H.; Two, A.M.; Chun, K.A.; Narala, S.; Geha, R.S.; Hata, T.R.; Gallo, R.L. Staphylococcus aureus Exploits
Epidermal Barrier Defects in Atopic Dermatitis to Trigger Cytokine Expression. J. Investig. Dermatol.
2016
,136, 2192–2200.
[CrossRef] [PubMed]
48.
Nograles, K.E.; Suárez-Fariñas, M.; Shemer, A.; Fuentes-Duculan, J.; Chiricozzi, A.; Cardinale, I.; Zaba, L.C.; Kikuchi, T.;
Ramon, M.
; Bergman, R.; et al. Atopic dermatitis keratinocytes exhibit normal T(H)17 cytokine responses. J. Allergy Clin. Immunol.
2010,125, 744–746. [CrossRef]
49.
Kim, J.; Kim, B.E.; Leung, D.Y.M. Pathophysiology of atopic dermatitis: Clinical implications. Allergy Asthma Proc.
2019
,40, 84–92.
[CrossRef]
50.
Woo, T.E.; Sibley, C.D. The emerging utility of the cutaneous microbiome in the treatment of acne and atopic dermatitis. J. Am.
Acad. Dermatol. 2020,82, 222–228. [CrossRef]
51.
Tubau, C.; Puig, L. Therapeutic targeting of the IL-13 pathway in skin inflammation. Expert Rev. Clin. Immunol.
2021
,17, 15–25.
[CrossRef] [PubMed]
52.
McCormick, S.M.; Heller, N.M. Commentary: IL-4 and IL-13 receptors and signaling. Cytokine
2015
,75, 38–50. [CrossRef]
[PubMed]
53.
Hussein, Y.M.; Ahmad, A.S.; Ibrahem, M.M.; Elsherbeny, H.M.; Shalaby, S.M.; El-Shal, A.S.; Sabbah, N.A. Interleukin 13 receptors
as biochemical markers in atopic patients. J. Investig. Allergol. Clin. Immunol. 2011,21, 101–107.
54.
Kwak, E.J.; Hong, J.Y.; Kim, M.N.; Kim, S.Y.; Kim, S.H.; Park, C.O.; Kim, K.W.; Lee, C.G.; Elias, J.A.; Jee, H.M.; et al. Chitinase
3-like 1 drives allergic skin inflammation via Th2 immunity and M2 macrophage activation. Clin. Exp. Allergy
2019
,49, 1464–1474.
[CrossRef] [PubMed]
55.
Tazawa, T.; Sugiura, H.; Sugiura, Y.; Uehara, M. Relative importance of IL-4 and IL-13 in lesional skin of atopic dermatitis. Arch.
Dermatol. Res. 2004,295, 459–464. [CrossRef] [PubMed]
56.
La Grutta, S.; Richiusa, P.; Pizzolanti, G.; Mattina, A.; Pajno, G.B.; Citarrella, R.; Passalacqua, G.; Giordano, C. CD4(+)IL-13(+)
cells in peripheral blood well correlates with the severity of atopic dermatitis in children. Allergy 2005,60, 391–395. [CrossRef]
57.
Hijnen, D.; Knol, E.F.; Gent, Y.Y.; Giovannone, B.; Beijn, S.J.; Kupper, T.S.; Bruijnzeel-Koomen, C.A.; Clark, R.A. CD8(+) T cells in
the lesional skin of atopic dermatitis and psoriasis patients are an important source of IFN-
γ
, IL-13, IL-17, and IL-22. J. Investig.
Dermatol. 2013,133, 973–979. [CrossRef]
58.
Bangert, C.; Rindler, K.; Krausgruber, T.; Alkon, N.; Thaler, F.M.; Kurz, H.; Ayub, T.; Demirtas, D.; Fortelny, N.; Vorstandlechner, V.;
et al. Persistence of mature dendritic cells, T(H)2A, and Tc2 cells characterize clinically resolved atopic dermatitis under IL-4R
α
blockade. Sci. Immunol. 2021,6, eabe2749. [CrossRef]
59.
Mashiko, S.; Mehta, H.; Bissonnette, R.; Sarfati, M. Increased frequencies of basophils, type 2 innate lymphoid cells and Th2 cells
in skin of patients with atopic dermatitis but not psoriasis. J. Dermatol. Sci. 2017,88, 167–174. [CrossRef]
60.
Szegedi, K.; Lutter, R.; Res, P.C.; Bos, J.D.; Luiten, R.M.; Kezic, S.; Middelkamp-Hup, M.A. Cytokine profiles in interstitial fluid
from chronic atopic dermatitis skin. J. Eur. Acad. Dermatol. Venereol. 2015,29, 2136–2144. [CrossRef]
Pharmaceutics 2023,15, 568 17 of 18
61.
Choy, D.F.; Hsu, D.K.; Seshasayee, D.; Fung, M.A.; Modrusan, Z.; Martin, F.; Liu, F.T.; Arron, J.R. Comparative transcriptomic
analyses of atopic dermatitis and psoriasis reveal shared neutrophilic inflammation. J. Allergy Clin. Immunol.
2012
,130,
1335–1343.e5. [CrossRef] [PubMed]
62.
Ungar, B.; Garcet, S.; Gonzalez, J.; Dhingra, N.; Correa da Rosa, J.; Shemer, A.; Krueger, J.G.; Suarez-Farinas, M.; Guttman-
Yassky, E. An Integrated Model of Atopic Dermatitis Biomarkers Highlights the Systemic Nature of the Disease. J. Investig.
Dermatol. 2017,137, 603–613. [CrossRef] [PubMed]
63.
Popovic, B.; Breed, J.; Rees, D.G.; Gardener, M.J.; Vinall, L.M.; Kemp, B.; Spooner, J.; Keen, J.; Minter, R.; Uddin, F.; et al. Structural
Characterisation Reveals Mechanism of IL-13-Neutralising Monoclonal Antibody Tralokinumab as Inhibition of Binding to
IL-13Rα1 and IL-13Rα2. J. Mol. Biol. 2017,429, 208–219. [CrossRef]
64. May, R.D.; Fung, M. Strategies targeting the IL-4/IL-13 axes in disease. Cytokine 2015,75, 89–116. [CrossRef] [PubMed]
65.
Tollenaere, M.A.X.; Litman, T.; Moebus, L.; Rodriguez, E.; Stölzl, D.; Drerup, K.; Werfel, T.; Schmitt, J.; Norsgaard, H.;
Weidinger, S
.
Skin Barrier and Inflammation Genes Associated with Atopic Dermatitis are Regulated by Interleukin-13 and Modulated by
Tralokinumab In vitro. Acta Dermatol. Venereol. 2021,101, adv00447. [CrossRef]
66.
Beck, L.A.; Bieber, T.; Weidinger, S.; Tauber, M.; Saeki, H.; Irvine, A.D.; Eichenfield, L.F.; Werfel, T.; Arlert, P.; Jiang, L.; et al.
Tralokinumab treatment improves the skin microbiota by increasing the microbial diversity in adults with moderate-to-severe
atopic dermatitis: Analysis of microbial diversity in ECZTRA 1, a randomized controlled trial. J. Am. Acad. Dermatol.
2022
.
[CrossRef]
67.
Oh, C.K.; Faggioni, R.; Jin, F.; Roskos, L.K.; Wang, B.; Birrell, C.; Wilson, R.; Molfino, N.A. An open-label, single-dose bioavailability
study of the pharmacokinetics of CAT-354 after subcutaneous and intravenous administration in healthy males. Br. J. Clin. Pharm.
2010,69, 645–655. [CrossRef]
68.
Wollenberg, A.; Blauvelt, A.; Guttman-Yassky, E.; Worm, M.; Lynde, C.; Lacour, J.P.; Spelman, L.; Katoh, N.; Saeki, H.;
Poulin, Y.; et al
. Tralokinumab for moderate-to-severe atopic dermatitis: Results from two 52-week, randomized, double-blind,
multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2). Br. J. Dermatol. 2021,184, 437–449. [CrossRef]
69.
Wollenberg, A.; Howell, M.D.; Guttman-Yassky, E.; Silverberg, J.I.; Kell, C.; Ranade, K.; Moate, R.; van der Merwe, R. Treatment of
atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J. Allergy Clin. Immunol. 2019,143, 135–141. [CrossRef]
70.
Simpson, E.L.; Flohr, C.; Eichenfield, L.F.; Bieber, T.; Sofen, H.; Taïeb, A.; Owen, R.; Putnam, W.; Castro, M.; DeBusk, K.; et al.
Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis
inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). J. Am. Acad.
Dermatol. 2018,78, 863–871.e11. [CrossRef]
71.
Silverberg, J.I.; Toth, D.; Bieber, T.; Alexis, A.F.; Elewski, B.E.; Pink, A.E.; Hijnen, D.; Jensen, T.N.; Bang, B.; Olsen, C.K.; et al.
Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: Results from the double-blind,
randomized, multicentre, placebo-controlled phase III ECZTRA 3 trial. Br. J. Dermatol.
2021
,184, 450–463. [CrossRef] [PubMed]
72.
Silverberg, J.I.; Adam, D.N.; Zirwas, M.; Kalia, S.; Gutermuth, J.; Pinter, A.; Pink, A.E.; Chiricozzi, A.; Barbarot, S.;
Mark, T.; et al
.
Tralokinumab Plus Topical Corticosteroids as Needed Provides Progressive and Sustained Efficacy in Adults with Moderate-to-
Severe Atopic Dermatitis Over a 32-Week Period: An ECZTRA 3 Post Hoc Analysis. Am. J. Clin. Dermatol.
2022
,23, 547–559.
[CrossRef] [PubMed]
73.
Blauvelt, A. Atopic dermatitis: Therapy. In Proceedings of the Annual Meeting of the American Academy of Dermatology (AAD
2022), Boston, MA, USA, 28 March 2022.
74.
Wollenberg, A.; Beck, L.A.; de Bruin Weller, M.; Simpson, E.L.; Imafuku, S.; Boguniewicz, M.; Zachariae, R.; Olsen, C.K.;
Thyssen, J.P
. Conjunctivitis in adult patients with moderate-to-severe atopic dermatitis: Results from five tralokinumab clinical
trials. Br. J. Dermatol. 2022,186, 453–465. [CrossRef] [PubMed]
75.
Gandhi, N.A.; Bennett, B.L.; Graham, N.M.; Pirozzi, G.; Stahl, N.; Yancopoulos, G.D. Targeting key proximal drivers of type 2
inflammation in disease. Nat. Rev. Drug Discov. 2016,15, 35–50. [CrossRef]
76.
Zhu, R.; Zheng, Y.; Dirks, N.L.; Vadhavkar, S.; Jin, J.Y.; Peng, K.; Holweg, C.T.J.; Olsson, J.; Matthews, J.G.; Putnam, W.S.
Model-based clinical pharmacology profiling and exposure-response relationships of the efficacy and biomarker of lebrikizumab
in patients with moderate-to-severe asthma. Pulm Pharm. Ther. 2017,46, 88–98. [CrossRef]
77.
Simpson, E.L. Efficacy And Safety of Lebrikizumab in Moderate-to-Severe Atopic Dermatitis: Results from Two Phase 3,
Randomized, Double-Blinded, Placebo-Controlled Trials. In Proceedings of the American Academy of Dermatology Annual
Meeting, Boston, MA, USA, 26 March 2022.
78.
Blauvelt, B.; Thyssen, J.P.; Guttman-Yassky, E.; Bieber, T.; Serra-Baldrich, E.; Simpson, E.; Rosmarin, D.; Elmaraghy, H.; Meski-
men, E.; Natalie, C.R.; et al. Efficacy and Safety of Lebrikizumab in Moderate-to-Severe Atopic Dermatitis: 52-Week Results of
Two Randomized, Double-Blinded, Placebo-Controlled Phase 3 Trials (ADvocate1 and ADvocate2). In Proceedings of the EADV
Congress, Milan, Italy, 7–10 September 2022.
79.
Safety and Efficacy of Lebrikizumab (LY3650150) in Combination With Topical Corticosteroid in Moderate-to-Severe Atopic Der-
matitis. (ADhere). Available online: https://clinicaltrials.gov/ct2/show/study/NCT04250337 (accessed on 9 November 2022).
80.
Noonan, M.; Korenblat, P.; Mosesova, S.; Scheerens, H.; Arron, J.R.; Zheng, Y.; Putnam, W.S.; Parsey, M.V.; Bohen, S.P.;
Matthews, J.G
. Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. J. Allergy. Clin. Immunol.
2013,132, 567–574.e12. [CrossRef]
Pharmaceutics 2023,15, 568 18 of 18
81.
Hanania, N.A.; Noonan, M.; Corren, J.; Korenblat, P.; Zheng, Y.; Fischer, S.K.; Cheu, M.; Putnam, W.S.; Murray, E.;
Scheerens, H.; et al
. Lebrikizumab in moderate-to-severe asthma: Pooled data from two randomised placebo-controlled studies.
Thorax 2015,70, 748–756. [CrossRef]
82.
Korenblat, P.; Kerwin, E.; Leshchenko, I.; Yen, K.; Holweg, C.T.J.; Anzures-Cabrera, J.; Martin, C.; Putnam, W.S.;
Governale, L
.;
Olsson, J.; et al. Efficacy and safety of lebrikizumab in adult patients with mild-to-moderate asthma not receiving inhaled
corticosteroids. Respir. Med. 2018,134, 143–149. [CrossRef]
83.
Scheerens, H.; Arron, J.R.; Zheng, Y.; Putnam, W.S.; Erickson, R.W.; Choy, D.F.; Harris, J.M.; Lee, J.; Jarjour, N.N.; Matthews, J.G.
The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge. Clin. Exp. Allergy
2014
,44,
38–46. [CrossRef]
84.
Hanania, N.A.; Korenblat, P.; Chapman, K.R.; Bateman, E.D.; Kopecky, P.; Paggiaro, P.; Yokoyama, A.; Olsson, J.; Gray, S.;
Holweg, C.T.; et al
. Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II):
Replicate, phase 3, randomised, double-blind, placebo-controlled trials. Lancet Respir. Med. 2016,4, 781–796. [CrossRef]
85.
Blauvelt, A. Eblasakimab, a human anti-IL-13 receptor monoclonal antibody, in adult patients with moderate-to-severe atopic
dermatitis: A randomized, double-blinded, placebo-controlled, proof-of-concept study. In Proceedings of the American Academy
of Dermatology Annual Meeting, Boston, MA, USA, 26 March 2022.
86.
Blauvelt, A.; Teixeira, H.D.; Simpson, E.L.; Costanzo, A.; De Bruin-Weller, M.; Barbarot, S.; Prajapati, V.H.; Lio, P.; Hu, X.;
Wu, T.; et al
. Efficacy and Safety of Upadacitinib vs Dupilumab in Adults With Moderate-to-Severe Atopic Dermatitis: A
Randomized Clinical Trial. JAMA Dermatol. 2021,157, 1047–1055. [CrossRef] [PubMed]
87.
Akinlade, B.; Guttman-Yassky, E.; de Bruin-Weller, M.; Simpson, E.L.; Blauvelt, A.; Cork, M.J.; Prens, E.; Asbell, P.; Akpek, E.;
Corren, J.; et al. Conjunctivitis in dupilumab clinical trials. Br. J. Dermatol. 2019,181, 459–473. [CrossRef] [PubMed]
88.
Ferreira, S.; Torres, T. Conjunctivitis in patients with atopic dermatitis treated with dupilumab. Drugs Context
2020
,9. [CrossRef]
[PubMed]
89.
Bakker, D.S.; Ariens, L.F.M.; van Luijk, C.; van der Schaft, J.; Thijs, J.L.; Schuttelaar, M.L.A.; van Wijk, F.; Knol, E.F.; Balak, D.M.W.;
van Dijk, M.R.; et al. Goblet cell scarcity and conjunctival inflammation during treatment with dupilumab in patients with atopic
dermatitis. Br. J. Dermatol. 2019,180, 1248–1249. [CrossRef]
90.
Agnihotri, G.; Shi, K.; Lio, P.A. A Clinician’s Guide to the Recognition and Management of Dupilumab-Associated Conjunctivitis.
Drugs R D 2019,19, 311–318. [CrossRef]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Lebrikizumab is a humanized mAb that binds soluble IL-13 at the non-receptorbinding domain with a high affinity [84]. Bound IL-13 is able to form a complex with IL-13Rα1, whereas it prevents heterodimerization with IL-4Rα and prevents signal transduction. ...
... Bound IL-13 is able to form a complex with IL-13Rα1, whereas it prevents heterodimerization with IL-4Rα and prevents signal transduction. Therefore, lebrikizumab inhibits the IL-4Rα-IL-13Rα1 signaling complex while continuing to regulate endogenous IL-13 via the stimulation of IL-13Rα2 [84]. Lebrikizumab is effective against atopic dermatitis and can be used clinically. ...
... Tralokinumab is a humanized mAb that competitively blocks the binding of IL-13 to both the IL-13Rα1 and IL-13Rα2 receptor chains [84]. Tralokinumab is also effective against atopic dermatitis and can be used clinically. ...
Article
Full-text available
Bronchial asthma is characterized by airway inflammation, airway hyperresponsiveness, and reversible airway obstruction. Eosinophils contribute to the pathogenesis of airway disease mainly by releasing eosinophil-specific granules, lipid mediators, superoxide anions, and their DNA. Type-2 cytokines such as interleukin (IL)-4 and IL-13 also play roles in the development of bronchial asthma. Among these cytokines, IL-4 is involved in T-cell differentiation, B-cell activation, B-cell differentiation into plasma cells, and the production of immunoglobulin E. Although IL-13 has similar effects to IL-4, IL-13 mainly affects structural cells, such as epithelial cells, smooth muscle cells, and fibroblasts. IL-13 induces the differentiation of goblet cells that produce mucus and induces the airway remodeling, including smooth muscle hypertrophy. IL-4 and IL-13 do not directly activate the effector functions of eosinophils; however, they can induce eosinophilic airway inflammation by upregulating the expression of vascular cell adhesion molecule-1 (for adhesion) and CC chemokine receptor 3 ligands (for migration). Dupilumab, a human anti-IL-4 receptor α monoclonal antibody that inhibits IL-4 and IL-13 signaling, decreases asthma exacerbations and mucus plugs and increases lung function in moderate to severe asthma. In addition, dupilumab is effective for chronic rhinosinusitis with nasal polyps and for atopic dermatitis, and IL-4/IL-13 blocking is expected to suppress allergen sensitization, including transcutaneous sensitization and atopic march.
... This activation leads to the phosphorylation of signal transducer and activator of transcription 6 (STAT6), which promotes the polarization of T-helper cells toward the Th2 phenotype. Additionally, IL-13 has the ability to bind to a decoy receptor known as IL-13Rα2, which has been studied for its involvement in collagen deposition, the itch-scratch cycle, fibrotic tissue remodeling, and the endogenous regulation of IL-13 [76]. The action of IL-13 can be summarized in Figure 3a. ...
... The action of IL-13 can be summarized in Figure 3a. Among the pathogenic factors of IL-13 are the following: keratinocyte alteration, reduction in antimicrobial peptide production, promotion of eosinophil recruitment, and stimulation of IgE [76]. ...
... The study demonstrated that EASI-75 was achieved in 13%, 50%, 57%, and 50% in the placebo, 200 mg group, 400 mg group, and 600 mg group, respectively. A decrease in the peak pruritus NRS by 37% in the 600 mg groups compared with a 16% decrease in the placebo group was statistically significant [76]. Currently, a randomized, double-blind, placebo-controlled, multicenter trial to evaluate the efficacy and safety of eblasakimab in 75 adult participants with moderate-to-severe AD previously treated with dupilumab is being conducted. ...
Article
Full-text available
Atopic dermatitis represents a complex and multidimensional interaction that represents potential fields of preventive and therapeutic management. In addition to the treatment armamentarium available for atopic dermatitis, novel drugs targeting significant molecular pathways in atopic dermatitis biologics and small molecules are also being developed given the condition’s complex pathophysiology. While most of the patients are expecting better efficacy and long-term control, the response to these drugs would still depend on numerous factors such as complex genotype, diverse environmental triggers and microbiome-derived signals, and, most importantly, dynamic immune responses. This review article highlights the challenges and the recently developed pharmacological agents in atopic dermatitis based on the molecular pathogenesis of this condition, creating a specific therapeutic approach toward a more personalized medicine.
... IL-13 is considered a major mediator involved in the inflammation, epidermal barrier dysfunction, and pruritus associated with AD. Selective IL-13 inhibitors such as tralokinumab, lebrikizumab, and eblasakimab have shown promising efficacy in the treatment of moderate to severe AD (102). While their safety profiles are generally favorable, there is a heightened risk of conjunctivitis, necessitating monitoring (102). ...
... Selective IL-13 inhibitors such as tralokinumab, lebrikizumab, and eblasakimab have shown promising efficacy in the treatment of moderate to severe AD (102). While their safety profiles are generally favorable, there is a heightened risk of conjunctivitis, necessitating monitoring (102). These findings collectively affirm the pivotal role of IL-4/13 in human AD. ...
Article
Full-text available
The pathogenesis of atopic dermatitis (AD) is understood to be crucially influenced by three main factors: dysregulation of the immune response, barrier dysfunction, and pruritus. In the lesional skin of AD, various innate immune cells, including Th2 cells, type 2 innate lymphoid cells (ILC2s), and basophils, produce Th2 cytokines [interleukin (IL)-4, IL-5, IL-13, IL-31]. Alarmins such as TSLP, IL-25, and IL-33 are also produced by epidermal keratinocytes, amplifying type 2 inflammation. In the chronic phase, not only Th2 cells but also Th22 and Th17 cells increase in number, leading to suppression of filaggrin expression by IL-4, IL-13, and IL-22, which further deteriorates the epidermal barrier function. Dupilumab, which targets IL-4 and IL-13, has shown efficacy in treating moderate to severe AD. Nemolizumab, targeting IL-31RA, effectively reduces pruritus in AD patients. In addition, clinical trials with fezakinumab, targeting IL-22, have demonstrated promising results, particularly in severe AD cases. Conversely, in murine models of AD, several cytokines, initially regarded as promising therapeutic targets, have not demonstrated sufficient efficacy in clinical trials. IL-33 has been identified as a potent activator of immune cells, exacerbating AD in murine models and correlating with disease severity in human patients. However, treatments targeting IL-33 have not shown sufficient efficacy in clinical trials. Similarly, thymic stromal lymphopoietin (TSLP), integral to type 2 immune responses, induces dermatitis in animal models and is elevated in human AD, yet clinical treatments like tezepelumab exhibit limited efficacy. Therapies targeting IL-1α, IL-5, and IL-17 also failed to achieve sufficient efficacy in clinical trials. It has become clear that for treating AD, IL-4, IL-13, and IL-31 are relevant therapeutic targets during the acute phase, while IL-22 emerges as a target in more severe cases. This delineation underscores the necessity of considering distinct pathophysiological aspects and therapeutic targets in AD between mouse models and humans. Consequently, this review delineates the distinct roles of cytokines in the pathogenesis of AD, juxtaposing their significance in human AD from clinical trials against insights gleaned from AD mouse models. This approach will improve our understanding of interspecies variation and facilitate a deeper insight into the pathogenesis of AD in humans.
... Cendakimab has recently completed a phase two clinical trial in atopic dermatitis, but the results have not yet been published. Eblasakimab is a monoclonal antibody targeting the IL-13Rα1 fragment, i.e., a component of the IL-13Rα1/IL-4Rα receptor complex, thus inhibiting IL-13 and IL-4 signaling [21,53]. A study based on a small group of patients suggests that the drug may be efficacious in reducing pruritus intensity: after eight weeks of therapy, a significant reduction in PP-NRS in patients receiving eblasakimab was noted in comparison to placebo [53]. ...
... Eblasakimab is a monoclonal antibody targeting the IL-13Rα1 fragment, i.e., a component of the IL-13Rα1/IL-4Rα receptor complex, thus inhibiting IL-13 and IL-4 signaling [21,53]. A study based on a small group of patients suggests that the drug may be efficacious in reducing pruritus intensity: after eight weeks of therapy, a significant reduction in PP-NRS in patients receiving eblasakimab was noted in comparison to placebo [53]. ...
Article
Full-text available
Atopic dermatitis is a heterogenous inflammatory disease with high variety in terms of clinical symptoms and etiopathogenesis, occurring both in pediatric and adult populations. The clinical manifestation of atopic dermatitis varies depending on the age of patients, but all age groups share certain common features, such as a chronic and recurrent course of disease, pruritus, and a co-occurrence of atopic diseases in personal or family medical history. Treating pruritus is a high priority due to its incidence rate in atopic dermatitis and substantial impact on quality of life. In recent years, treatments with biological drugs have increased the range of therapeutic possibilities in atopic dermatitis. The aim of the study is to present the safety profile, efficacy, and effectiveness of various biological treatment methods for the therapy of pruritus in the course of atopic dermatitis.
... In 2017, dupilumab, an anti-interleukin (IL)-4 receptor antibody [11,12], became available for the treatment of AD, followed by the Janus kinase (JAK) inhibitors, baricitinib in 2020 [13] and upadacitinib [14,15] and abrocitinib [16] in 2021. Nemolizumab [17], an anti-IL-31 receptor antibody, was introduced in 2022, and anti-IL-13 and anti-OX40 antibodies will become available in the near future [18][19][20]. However, the abundance of new drugs makes it difficult for clinicians to decide which one to choose because there is yet to be sufficient real-world evidence regarding the effectiveness of these new systemic therapies. ...
Article
Full-text available
Introduction It remains unclear which therapy contributes to atopic dermatitis (AD) remission and to what extent. We aimed to clarify which therapy contributes to the treatment of AD by investigating the time-to-remission and remission hazard ratios for each therapy using real-world data. Methods This retrospective cohort study included 110 patients diagnosed with AD after their first visit to the Department of Dermatology at Fukuoka University Hospital between 2016 and 2022. The patients were categorized into six treatment groups: 1) topical treatment alone or topical treatment plus 2) ultraviolet light, 3) oral steroids, 4) oral cyclosporine, 5) dupilumab, and 6) oral Janus kinase inhibitors (JAKi). The topical therapy alone group served as the control, and the hazard ratios for remission (Investigator’s Global Assessment [IGA] 0/1) were calculated. Results Forty patients achieved remission, while 70 did not (IGA ≥2) with the first treatment regimen. A multivariate Cox proportional hazards analysis adjusted for age, sex, and severity at the first visit (IGA) revealed that the hazard ratios for remission were 4.2 (95% confidence interval (C.I.): 1.28–13.83, p = 0.018) for the oral cyclosporine group, 5.05 (95% C.I.: 1.96–13, p = 0.001) for the dupilumab group, and 67.56 (95% C.I.: 12.28–371.68, p < .0001) for the oral JAKi group. The median time to remission was 3 months for JAKi, cyclosporine, and steroid was shorter than 6 months for dupilumab. No serious adverse events were observed. Conclusion Oral therapy with small molecules requires a shorter duration to achieve remission. However, long-term safety and recurrence are important indicators.
... The half-maximal inhibitory concentration for inhibition of IL-4einduced TARC secretion by NM26-2198 ranged from 328 to 490 pM and averaged 409 AE 81.1 pM. Inhibition of IL-13einduced TARC secretion by NM26-2198 had a similar potency (286 AE 142 pM, data not shown), which is important because of evidence that IL-13 has a strong role in AD pathology (Lytvyn and Gooderham, 2023;Tazawa et al, 2004;Tsoi et al, 2019;Zhang et al, 2022). These results are consistent with clinical trial results in which dupilumab treatment reduced serum TARC levels in adult patients with AD (Guttman-Yassky et al, 2019) and suggest that NM26-2198 may have a similar effect in patients. ...
Article
Full-text available
Inhibition of IL-4/IL-13 signaling has dramatically improved the treatment of atopic dermatitis (AD). However, in many patients, clinical responses are slow to develop and remain modest. Indeed, some symptoms of AD are dependent on IL-31, which is only partially reduced by IL-4/IL-13 inhibition. Thus, there is an unmet need for AD treatments that concomitantly block IL-4/IL-13 and IL-31 pathways. We engineered NM26-2198, a bispecific tetravalent antibody designed to accomplish this task. In reporter cell lines, NM26-2198 concomitantly inhibited IL-4/IL-13 and IL-31 signaling with a potency comparable with that of the combination of an anti–IL-4Rα antibody (dupilumab) and an anti–IL-31 antibody (BMS-981164). In human PBMCs, NM26-2198 inhibited IL-4–induced upregulation of CD23, demonstrating functional binding to FcγRII (CD32). NM26-2198 also inhibited the secretion of the AD biomarker thymus and activation-regulated chemokine (TARC) in blood samples from healthy human donors. In male cynomolgus monkeys, NM26-2198 exhibited favorable pharmacokinetics and significantly inhibited IL-31–induced scratching at a dose of 30 mg/kg. In a repeat-dose, good laboratory practice toxicology study in cynomolgus monkeys, no adverse effects of NM26-2198 were observed at a weekly dose of 125 mg/kg. Together, these results justify the clinical investigation of NM26-2198 as a treatment for moderate-to-severe AD.
... The dysregulation of type 2 helper T cells, which produce cytokines such as interleukins 4, 13, and 31, plays an essential role in the pathogenesis of AD [8,9]. Recent evidence suggests that interleukin-13 may be the key cytokine in AD and serum levels of interleukin-13 correlate with disease severity [10][11][12][13][14][15][16][17][18][19][20][21][22][23][24]. Consequently, targeting the signalling of interleukins is an attractive approach to treatment of AD. ...
Article
Full-text available
Lebrikizumab and dupilumab are monoclonal antibodies approved for treating moderate-to-severe atopic dermatitis (AD). Both have demonstrated efficacy and safety over the 16-week SOLOs and ADvocate trials. However, AD is a chronic and relapsing inflammatory disease, and the long-term maintenance of efficacy is critical for achieving disease control from the perspective of patients, physicians, and regulatory agencies. This study aims to compare the long-term efficacy and safety of lebrikizumab every 4 weeks (Q4W) and dupilumab every week or every 2 weeks (QW/Q2W) among adult patients who have achieved treatment efficacy following the induction period of 16 weeks. Lebrikizumab’s efficacy was assessed using individual patient data (IPD) from the ADvocate 1 and 2 monotherapy trials. Dupilumab’s efficacy was evaluated using aggregate data from the adult-exclusive SOLO-CONTINUE trial. Due to the absence of a common comparator trial arm, we employed an unanchored matching-adjusted indirect comparison (MAIC), a robust methodology widely accepted by health technology assessment (HTA) agencies. This re-weights ADvocate IPD to align with SOLO-CONTINUE’s prognostic factors and effect modifiers. We compared lebrikizumab’s adjusted outcomes with dupilumab outcomes at week 52, focusing on 75% improvement in the Eczema Area and Severity Index from baseline (EASI-75), Investigator’s Global Assessment (IGA) score of 0 or 1, and overall adverse event (AE) rates. Sensitivity analyses were conducted to test various combinations of matching variables. Adults on lebrikizumab Q4W were more likely to maintain IGA 0/1 through the 36-week maintenance period (weeks 16–52) compared with those on dupilumab QW/Q2W [risk ratio (RR) 1.334; 95% confidence interval (CI) 1.02–1.74; p = 0.035]. Both treatments demonstrated comparable efficacy in terms of EASI-75 maintenance (RR 0.937; 95% CI 0.78–1.13; p = 0.490) and similar AE rates (RR 1.052; 95% CI 0.90–1.23; p = 0.526). Sensitivity analyses substantiated these findings. Our findings suggest that lebrikizumab Q4W may provide equal or superior long-term maintenance of efficacy measured with EASI-75 and IGA 0/1 compared with dupilumab QW/Q2W, with the advantage of requiring less frequent doses.
Article
Globally, nearly 400 million persons have COPD, and COPD is one of the leading causes of hospitalisation and mortality across the world. While it has been long-recognised that COPD is an inflammatory lung disease, dissimilar to asthma, type 2 inflammation was thought to play a minor role. However, recent studies suggest that in approximately one third of patients with COPD, type 2 inflammation may be an important driver of disease and a potential therapeutic target. Importantly, the immune cells and molecules involved in COPD-related type 2 immunity may be significantly different from those observed in severe asthma. Here, we identify the important molecules and effector immune cells involved in type 2 airway inflammation in COPD, discuss the recent therapeutic trial results of biologicals that have targeted these pathways and explore the future of therapeutic development of type 2 immune modulators in COPD.
Article
Full-text available
Background: Lebrikizumab is a novel, high-affinity monoclonal antibody that selectively binds to interleukin (IL)-13. Objectives: To evaluate the efficacy and safety of lebrikizumab monotherapy in adolescent and adult patients with moderate-to-severe atopic dermatitis (AD) over 52 weeks of treatment in ADvocate1 (NCT04146363) and ADvocate2 (NCT04178967). Methods: Patients who responded to lebrikizumab 250 mg every 2 weeks (Q2W) at the end of the 16-week induction period were re-randomized 2 : 2 : 1 to receive lebrikizumab Q2W, lebrikizumab 250 mg every 4 weeks (Q4W) or placebo Q2W (lebrikizumab withdrawal) for 36 additional weeks. Response at week 16 was defined as achieving a 75% reduction in Eczema Area Severity Index (EASI 75) or an Investigator's Global Assessment (IGA) of 0 or 1, with a ≥ 2-point improvement and no rescue medication use. Multiple imputation was used to handle missing data. Intermittent use of topical therapy was permitted during the maintenance period. Results: After 52 weeks, an IGA of 0 or 1 with a ≥ 2 point improvement was maintained by 71.2% of patients treated with lebrikizumab Q2W, 76.9% of patients treated with lebrikizumab Q4W and 47.9% of patients in the lebrikizumab withdrawal arm. EASI 75 was maintained by 78.4% of patients treated with lebrikizumab Q2W, 81.7% of patients treated with lebrikizumab Q4W and 66.4% of patients in the lebrikizumab withdrawal arm at week 52. Across treatment arms, proportions of patients using any rescue therapy were 14.0% (ADvocate1) and 16.4% (ADvocate2). During the combined induction and maintenance periods of ADvocate1 and ADvocate2, 63.0% of lebrikizumab-treated patients reported any treatment emergent adverse event, with most events (93.1%) being mild or moderate in severity. Conclusions: After a 16-week induction period with lebrikizumab Q2W, lebrikizumab Q2W and Q4W maintained similar improvement of the signs and symptoms of moderate-to-severe AD, with a safety profile consistent with previously published data.
Article
Full-text available
Background Atopic dermatitis (AD) is characterized by microbial dysbiosis, immune dysregulation, and an impaired skin barrier. Microbial dysbiosis in AD involves a reduction in diversity primarily driven by an increased abundance of Staphylococcus aureus. Tralokinumab, an approved treatment for adults with moderate-to-severe AD, improves the skin barrier and immune abnormalities by specifically targeting the interleukin (IL)-13 cytokine, but its impact on the skin microbiome is unknown. Objective To investigate how tralokinumab affects the skin microbiome by examining the lesional skin of adults with moderate-to-severe AD from the Phase 3 ECZTRA 1 trial (NCT03131648). Methods Microbiome profiling, S. aureus abundance, and biomarker data were assessed in a subset of ECZTRA 1 participants (S. aureus abundance at baseline and Week 16; microbiome profiling at baseline, Week 8/16; and serum sampling at pre-dose, Week 4/8/16/28/52). Results Tralokinumab treatment led to increased microbial diversity, reduced S. aureus abundance, and increased abundance of the commensal coagulase-negative Staphylococci. Limitations Limitations include a lack of S. aureus abundance data at Week 8, sampling site variation between participants, and possible influence from concomitant systemic anti-infectives. Conclusion Our findings indicate specific targeting of the IL-13 cytokine with tralokinumab can directly and/or indirectly improve microbial dysbiosis seen in AD skin.
Article
Full-text available
Background: The efficacy and safety of tralokinumab, a fully human monoclonal antibody that specifically neutralizes interleukin-13, plus topical corticosteroids (TCS) as needed were evaluated over 32 weeks in the phase III ECZTRA 3 trial. Significantly more tralokinumab- versus placebo-treated patients achieved the primary endpoints of Investigator's Global Assessment (IGA) score of 0/1 and 75% improvement in Eczema Area and Severity Index (EASI-75) and all confirmatory endpoints at Week 16. Objective: This post hoc analysis investigated the impact of tralokinumab plus TCS on atopic dermatitis (AD) severity, symptoms, and health-related quality of life (QoL) over the entire 32-week treatment period of ECZTRA 3, including all patients initiated on tralokinumab irrespective of the response achieved at Week 16. Methods: Patients were randomized 2:1 to receive subcutaneous tralokinumab 300 mg or placebo every 2 weeks (q2w) with TCS as needed for an initial 16 weeks. At Week 16, patients who achieved the clinical response criteria (IGA 0/1 and/or EASI-75) with tralokinumab were re-randomized 1:1 to tralokinumab q2w or every 4 weeks (q4w), with TCS as needed, for another 16 weeks. Patients not achieving the clinical response criteria with tralokinumab received tralokinumab q2w plus TCS from Week 16. All patients randomized to tralokinumab in the initial treatment period were pooled for this analysis, irrespective of response at Week 16 or dosing regimen beyond Week 16. Results: Continued tralokinumab (q2w, N = 164; q4w, N = 69) plus TCS treatment provided progressive improvements from Week 16 onwards in AD signs, with 70.2% (177/252) of patients achieving EASI-75 and 50.4% (127/252) achieving EASI-90 at Week 32. Improvements in patient-reported outcomes were observed within the first few weeks of tralokinumab q2w plus TCS treatment and were sustained throughout the 32-week period. At Week 32, patients initiated on tralokinumab q2w plus TCS achieved a relative improvement versus baseline of 70.8% (standard error (SE), 2.4) in eczema-related sleep interference numeric rating scale (NRS) and 66.8% (SE, 3.1) in Dermatology Life Quality Index (DLQI). Mean TCS use during Weeks 16-32 ranged from 9.2 to 13.6 g (SE, 1.2-2.0) q2w. Most patients (89.9% (222/247)) initiated on tralokinumab q2w plus TCS achieved a meaningful improvement in at least one of the three disease domains, including AD signs (EASI-50), symptoms (pruritus NRS improvement ≥ 3), and QoL (DLQI improvement ≥ 4) at Week 16. Of patients initiated on tralokinumab q2w plus TCS, 53.4% (132/247) achieved a clinically meaningful improvement in all three domains at Week 16 (vs. placebo, 28.5% (35/123); p < 0.001). Conclusions: Continued tralokinumab treatment plus TCS as needed provides progressive and sustained improvements in AD signs, symptoms, and health-related QoL over 32 weeks. Clinical trial registration: NCT03363854; study start date: 22 February 2018; primary completion date: 8 March 2019; study completion date: 26 September 2019.
Article
Full-text available
Background: Tralokinumab, a fully human immunoglobulin G4 monoclonal antibody that specifically binds to the interleukin-13 cytokine with high affinity, effectively reduces moderate-to-severe atopic dermatitis when given every 2 weeks. The incidence of conjunctivitis is elevated compared to placebo, but severity and etiology have not been examined. Objective: To analyze conjunctivitis data recorded in five randomized, placebo-controlled trials of tralokinumab in adult patients with moderate-to-severe atopic dermatitis. Methods: Overall, 2285 adults with atopic dermatitis were studied up to 16 weeks. Cochran-Mantel-Haenszel weights were applied to calculate adjusted adverse-event incidences. Results: Incidence of conjunctivitis was higher (7.5%) with tralokinumab compared to placebo (3.2%). Most events were mild or moderate in severity and 78.6% and 73.9% of events resolved during the trial in the tralokinumab and placebo groups, respectively. Two (1.4%) events led to permanent discontinuation of tralokinumab. An increased incidence of conjunctivitis, regardless of treatment group, was associated with more severe baseline atopic dermatitis, and history of allergic conjunctivitis/atopic keratoconjunctivitis, as well as the number of atopic comorbidities. Limitation This analysis reports events up to Week 16 only, with limited confirmation of conjunctivitis and its etiology by an ophthalmologist and insufficient reporting of ophthalmic treatments. Conclusions: Treatment with tralokinumab was associated with increased incidence of conjunctivitis compared to placebo, but these cases were mostly mild and transient.
Article
Full-text available
Importance Atopic dermatitis (AD) is a chronic, recurrent, inflammatory skin disease with an unmet need for treatments that provide rapid and high levels of skin clearance and itch improvement. Objective To assess the safety and efficacy of upadacitinib vs dupilumab in adults with moderate-to-severe AD. Design, Setting, and Participants Heads Up was a 24-week, head-to-head, phase 3b, multicenter, randomized, double-blinded, double-dummy, active-controlled clinical trial comparing the safety and efficacy of upadacitinib with dupilumab among 692 adults with moderate-to-severe AD who were candidates for systemic therapy. The study was conducted from February 21, 2019, to December 9, 2020, at 129 centers located in 22 countries across Europe, North and South America, Oceania, and the Asia-Pacific region. Efficacy analyses were conducted in the intent-to-treat population. Interventions Patients were randomized 1:1 and treated with oral upadacitinib, 30 mg once daily, or subcutaneous dupilumab, 300 mg every other week. Main Outcomes and Measures The primary end point was achievement of 75% improvement in the Eczema Area and Severity Index (EASI75) at week 16. Secondary end points were percentage change from baseline in the Worst Pruritus Numerical Rating Scale (NRS) (weekly average), proportion of patients achieving EASI100 and EASI90 at week 16, percentage change from baseline in Worst Pruritus NRS at week 4, proportion of patients achieving EASI75 at week 2, percentage change from baseline in Worst Pruritus NRS (weekly average) at week 1, and Worst Pruritus NRS (weekly average) improvement of 4 points or more at week 16. End points at week 24 included EASI75, EASI90, EASI100, and improvement of 4 points or more in Worst Pruritus NRS from baseline (weekly average). Safety was assessed as treatment-emergent adverse events in all patients receiving 1 or more dose of either drug. Results Of 924 patients screened, 348 (183 men [52.6%]; mean [SD] age, 36.6 [14.6] years) were randomized to receive upadacitinib and 344 were randomized to receive dupilumab (194 men [56.4%]; mean [SD] age, 36.9 [14.1] years); demographic and disease characteristics were balanced among treatment groups. At week 16, 247 patients receiving upadacitinib (71.0%) and 210 patients receiving dupilumab (61.1%) achieved EASI75 (P = .006). All ranked secondary end points also demonstrated the superiority of upadacitinib vs dupilumab, including improvement in Worst Pruritus NRS as early as week 1 (mean [SE], 31.4% [1.7%] vs 8.8% [1.8%]; P < .001), achievement of EASI75 as early as week 2 (152 [43.7%] vs 60 [17.4%]; P < .001), and achievement of EASI100 at week 16 (97 [27.9%] vs 26 [7.6%]; P < .001). Rates of serious infection, eczema herpeticum, herpes zoster, and laboratory-related adverse events were higher for patients who received upadacitinib, whereas rates of conjunctivitis and injection-site reactions were higher for patients who received dupilumab. Conclusions and Relevance During 16 weeks of treatment, upadacitinib demonstrated superior efficacy vs dupilumab in patients with moderate-to-severe AD, with no new safety signals. Trial Registration ClinicalTrials.gov Identifier: NCT03738397
Article
Full-text available
Pruritus is a common symptom of several skin diseases, both inflammatory and neoplastic. Pruritus might have a tremendous impact on patients’ quality of life and strongly interfere with sleep, social, and work activities. We review the role of type-2 inflammation and immunity in the pathogenesis of chronic pruritic conditions of the skin. Type 2 cytokines, including IL-4, IL-13, thymic stromal lymphopoietin, periostin, IL-31, IL-25, and IL-33 are released by mast cells, innate lymphoid cells 2, keratinocytes, and type 2 T lymphocytes, and are master regulators of chronic itch. These cytokines might act as direct pruritogen on primary sensory neurons (pruriceptors) or alter the sensitivity to other itch mediators Type 2 inflammation- and immunity-dominated skin diseases, including atopic dermatitis, prurigo nodularis, bullous pemphigoid, scabies, parasitic diseases, urticaria, and Sézary syndrome are indeed conditions associated with most severe pruritus. In contrast, in other skin diseases, such as scleroderma, lupus erythematosus, hidradenitis suppurativa, and acne, type 2 inflammation is less represented, and pruritus is milder or variable. Th2 inflammation and immunity evolved to protect against parasites, and thus, the scratching response evoked by pruritus might have developed to alert about the presence and to remove parasites from the skin surface.
Conference Paper
Re: SPEAKER INVITATION TO THE 23RD WORLD CONGRESS OF DERMATOLOGY The 23RD World Congress of Dermatology (WCD) will take place from June 8 to 13, 2015 in Vancouver, Canada. On behalf of the 23RD WCD and the International League of Dermatological Societies (ILDS), it is our honor and pleasure to invite you to deliver a presentation at the following session: Session: SY01-Atopic Dermatitis Session Day / Time: Tuesday June 9, 08:00-10:30 Session Chair: Dhar Sandipan, India drsandipan@gmail.com Co-Chairs: Thomas Bieber, Germany thomas.bieber@ukb.uni-bonn.de Michael J. Cork, United Kingdom m.j.cork@sheffield.ac.uk; l.hunter@sheffield.ac.uk Presentation Title: Topical Therapy of Atopic Dermatitis
Article
Lebrikizumab (LEB) is a novel, high-affinity monoclonal antibody that selectively binds to interleukin (IL)-13. To evaluate the efficacy and safety of LEB monotherapy in patients with moderate-to-severe atopic dermatitis (AD) in two identical phase 3 trials ADvocate1 (ADv1) and ADvocate2 (ADv2). Patients who responded to LEB 250 mg every 2 weeks (LEB Q2W) at the end of the 16-week induction period were re-randomized in a 2 : 2 : 1 ratio to receive LEB Q2W, LEB 250 mg every 4 weeks (LEB Q4W) or placebo (LEB withdrawal) for an additional 36 weeks. Response, at week 16, was defined as achieving an IGA (0, 1) with a ≥2-point improvement or EASI75 and no use of rescue medication. Efficacy outcomes reported at week 52 included IGA (0, 1), EASI 75, ≥4-point reduction in Pruritis Numeric Rating Scale (NRS), EASI 90 and DLQI ≥4-point. Safety analysis was conducted on all patients who received ≥1 dose of LEB. Patients maintained IGA (0, 1) in LEB Q2W (ADv1, 75.8%; ADv2, 64.6%), LEB Q4W (ADv1, 74.2%; ADv2, 80.6%) and LEB withdrawal (ADv1, 46.5%; ADv2, 49.8%). Maintenance of EASI75 was, in LEB Q2W (ADv1, 79.2%; ADv2, 77.4%), LEB Q4W (ADv1, 79.2%; ADv2, 84.7%) and LEB withdrawal (ADv1, 61.3%; ADv2, 72.0%). For Pruritus NRS ≥4-point improvement from baseline, patients-maintained improvement in the LEB Q2W (ADv1, 81.2%; ADv2, 90.3%), LEB Q4W (ADv1, 80.4%; ADv2, 88.1%) and LEB withdrawal (ADv1, 65.4%; ADv2, 67.6%). Maintenance of EASI90 was, in LEB Q2W (ADv1, 66.1%; ADv2, 61.5%), LEB Q4W (ADv1, 66.6%; ADv2, 67.4%) and LEB withdrawal (ADv1, 45.5%; ADv2, 36.9%). DLQI ≥4-point improvement from baseline was LEB Q2W (ADv1, 64.0%; ADv2, 59.0%), LEB Q4W (ADv1, 62.7%; ADv2, 73.0%) and LEB withdrawal (ADv1, 57.7%; ADv2, 45.5%). TEAEs were reported by 58.1% (ADv1) and 67.8% (ADv2) LEB-treated patients at week 52. Serious adverse events were reported by 3.3% of ADv1 patients and 2.7% of ADv2 patients. In ADv1 and ADv2, 2.3% and 3.9% of patients reported an adverse event leading to treatment discontinuation, respectively. Both LEB Q2W and LEB Q4W maintained improvement in all reported outcomes for the treatment of moderate-to-severe AD through 52 weeks. The safety profile was consistent with previously published data.
Article
Introduction Atopic dermatitis (AD) is the most common inflammatory skin disease. It has a complex pathophysiology, with a combination of immune dysregulation and intrinsic barrier defects driving cutaneous inflammation and allergic symptomatology. The IL-4, IL-13, and IL-31 inflammatory pathways have been identified as hallmark features in the pathogenesis of the disease, contributing uniquely and synergistically to immune and barrier abnormalities as well as the key symptoms, such as pruritis. Novel therapeutics that target these pathways have been under development to find treatments for AD. Areas covered This review discusses the IL-4, IL-13, and IL-31 pathways in AD. We will also detail novel targeted therapeutics that have recently been or are currently in clinical trials for AD. A literature search was conducted by querying Scopus, Google Scholar, PubMed, and Clinicaltrials.gov up to January 2021 using combinations of the search terms “IL-4” “IL-13” “IL-31” “atopic dermatitis” “immune pathway” “biologics” “novel therapeutics” “JAK/STAT inhibitors.” Expert opinion The complex pathophysiology of AD advocates for innovation. Novel minimally invasive sampling modalities such as tape stripping will allow for a broader characterization of the immunomechanisms behind AD pathophysiology. This will allow for the continued development of a personalized medicine approach to treat AD.