Next Article in Journal
Multivessel versus Culprit-Only Percutaneous Coronary Intervention in Patients with Non-ST-Elevation Acute Coronary Syndrome
Next Article in Special Issue
Elevated Level of Serum Neurotrophin-4, but Not of Brain-Derived Neurotrophic Factor, in Patients with Chronic Kidney Disease-Associated Pruritus
Previous Article in Journal
A Clot Waveform Analysis of Thrombin Time Using a Small Amount of Thrombin Is Useful for Evaluating the Clotting Activity of Plasma Independent of the Presence of Emicizumab
Previous Article in Special Issue
The Impact of Pruritus on the Quality of Life and Sleep Disturbances in Patients Suffering from Different Clinical Variants of Psoriasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Dawn of a New Era in Atopic Dermatitis Treatment

by
Kazuhiko Yamamura
1,* and
Takeshi Nakahara
1,2
1
Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
2
Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka 812-8582, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(20), 6145; https://doi.org/10.3390/jcm11206145
Submission received: 15 September 2022 / Revised: 12 October 2022 / Accepted: 17 October 2022 / Published: 18 October 2022
(This article belongs to the Special Issue 10th Anniversary of JCM – New Era in Dermatology)

Abstract

:
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases, and the condition is typified by barrier dysfunction and immune dysregulation. Recent studies have characterized various phenotypes and endotypes of AD and elucidated the mechanism. Numerous topical and systemic narrow targeting therapies for AD have been developed according to these findings. Topical medications, including Janus kinase (JAK) inhibitors, phosphodiesterase 4 inhibitors, and the aryl hydrocarbon receptor agonist tapinarof, are effective and safe for AD compared to topical corticosteroids. Oral JAK inhibitors and monoclonal antibodies targeting interleukin (IL)-4, IL-13, IL-31, IL-33, OX40, thymic stromal lymphopoietin, and sphingosine 1-phosphate signaling have displayed outstanding efficacy against moderate-to-severe AD. We are currently in a new era of AD treatment.

1. Introduction

Atopic dermatitis (AD) is a chronic, relapsing, inflammatory skin disease characterized by persistent pruritus with barrier dysfunction, microbial dysbiosis, and immune dysregulation [1]. The estimated prevalence of AD is 15–20% in children and 6–10% in adults, among whom 40% are classified as having moderate-to-severe disease [2,3,4,5]. In recent decades, patients have been treated with topical corticosteroids/calcineurin inhibitors, phototherapy, and systemic immunosuppressants. However, many patients require frequent laboratory monitoring during systemic immunosuppressant therapy, and they are undertreated because of concerns regarding adverse effects [6]. Patients with moderate-to-severe AD harbor systemic inflammation/immune abnormalities such as strong Th2 activation, expansion of T cell subsets, and increased levels of pro-inflammatory cytokines, including interleukin (IL)-4, IL-13, and IL-31 [7,8,9,10,11,12,13,14]. Therefore, new agents have been developed to target these cytokines, and they have displayed outstanding efficacy for patients with moderate-to-severe AD. Interestingly, topical phosphodiesterase (PDE) 4 inhibitors and aryl hydrocarbon receptor (AhR) agonists are also effective for AD skin lesions in terms of the restoration of skin barrier function and the regulation of inflammatory cytokine production [15,16,17]. This review discusses the molecular mechanisms and therapeutic targets involved in the pathogenesis of AD.

2. Emerging Systemic/Topical Agents

In the past few years, numerous systemic (Table 1) and topical (Table 2) emerging agents have been developed for the treatment of patients with AD.

3. Monoclonal Antibodies for Moderate-to-Severe AD

3.1. Targeting Th2 and Th2-Associated Cytokines: IL-4, IL-13, IL-33, Thymic Stromal Lymphopoietin, and OX40

Narrow targeting agents for AD have been developed based on its pathogenesis (Figure 1). Accumulating evidence indicates that AD features multiple abnormalities in terms of epidermal barrier dysfunction, immunologic dysregulation, and microbial dysbiosis (e.g., increased abundance of Staphylococcus aureus and loss of commensal bacterial species) [1]. AD is considered a disease of Th2 predominance, and blockade of Th2 signaling is highly effective in treatment [37]. Dupilumab, an emerging narrow targeting agent that blocks both IL-4 and IL-13 signaling, has exhibited significant clinical benefits in patients with AD [18,38]. Skin IL-13 expression is correlated with disease severity in patients with AD [39,40]. Furthermore, recent studies have illustrated that the IL-13-specific antagonists tralokinumab and lebrikizumab have similar effects as dupilumab [19,20]. These data indicate that IL-13 acts as a critical cytokine in moderate-to-severe AD [41]. Traditionally, CD4+ helper T cells have been implicated as the source of Th2 cytokines. However, group 2 innate lymphoid cells (ILC2s) recently emerged as important contributors to AD through their production of IL-5 and IL-13 [42]. ILC2s, which belong to the larger ILC family, also include group 1 and group 3 ILCs [43]. At the cell surface, ILC2s express receptors for the cytokines IL-25, IL-33, thymic stromal lymphopoietin (TSLP), IL-2, IL-9, and IL-7 [44,45,46,47]. IL-33, an alarmin belonging to the IL-1 family, is mainly produced by keratinocytes in skin after cell death or in response to various stimuli, such as antigen challenges and scratches [48]. Human ILC2s in steady-state skin respond to IL-33 and IL-25 but not to TSLP [49]. Etokimab, a human monoclonal IgG1 antibody that neutralizes the activity of IL-33, proved efficacious for AD in a phase 2a trial [21,22]. TSLP is highly expressed in the skin of patients with AD, similar to IL-33, and it activates human myeloid dendritic cells to induce an inflammatory Th2 response [50,51]. However, tezepelumab, the monoclonal antibody targeting TSLP, did not provide significant improvements in patients with moderate-to-severe AD compared to the effects of placebo in a phase 2a trial [23]. These results indicate that IL-33 might contribute to AD aggravation by being more closely associated with ILC-mediated IL-13 production than TSLP. Conversely, the ligand for OX40 (OX40L, also known as CD134L and CD252) is primarily induced by TSLP [50,51]. OX40L is mainly expressed on antigen-presenting cells, such as activated B cells, dendritic cells, monocytes, and Langerhans cells [52,53,54,55]. OX40 (CD134), the receptor for OX40L, transiently expresses after antigen recognition [56]. It is predominantly expressed on activated/memory CD4+ T cells and Tregs, whereas it displays lower expression on CD8+ T, NK, and NKT cells [56]. The OX40–OX40L interaction is crucial for Th2 responses generating memory T cells by promoting the survival of effector T cells after antigen priming [57,58,59,60,61]. The OX40L–OX40 axis is a novel therapeutic target in autoimmune and inflammatory diseases, as it directly targets antigen-specific T cells responsible for clinical phenotypes without causing widespread immunosuppression [52,56]. A recent phase 2a clinical trial demonstrated that GBR 830, a humanized monoclonal antibody against OX40 that inhibits OX40–OX40L binding, induced significant progressive tissue and clinical changes in patients with moderate-to-severe AD [24].

3.2. Targeting Pruritus and the Th2-Associated Cytokine IL-31

IL-31, a four-helix bundle cytokine belonging to the IL-6 cytokine family, is preferentially produced by activated Th2 cells [62,63,64]. It transmits signals via a heterodimeric receptor composed of IL-31 receptor A (IL31RA) and oncostatin M receptor [64,65]. These receptors are expressed on various cell types, including cutaneous peripheral neurons and dorsal root ganglia neurons [65,66,67,68,69]. IL-31 is considered a major pruritogen in AD, and serum IL-31 levels are correlated with disease severity [14,70]. The humanized monoclonal antibody nemolizumab, which targets IL-31 receptor alfa, displayed an apparent anti-pruritic effect in a phase 3 clinical trial [25]. Although the precise mechanism of IL-31 production has not been fully elucidated, the transcription factor endothelial PAS domain protein 1 (EPAS1) plays a key role in IL-31 induction in AD skin inflammation [71]. Further, 4-(2-(4-isopropylbenzylidene)hydrazineyl)benzoic acid, as a small-molecule inhibitor, suppresses EPAS1-driven IL-31 induction [72]. In addition, DOCK8 has been identified as a negative regulator of IL-31 production linked to EPAS1 nuclear translocation [71]. It is well known that homozygous and compound heterozygous mutations in DOCK8 cause combined immunodeficiency characterized by recurrent viral infections, early-onset malignancy, and AD [73,74,75,76,77]. Consistent with this, DOCK8 polymorphism is associated with serum IL-31 levels in patients with moderate-to-severe AD [78].

3.3. Targeting Th17-Associated Cytokine IL-17

Psoriasis, along with AD, is one of the most common inflammatory skin diseases. While AD has a strong Th2 component associated with IL-4 and IL-13 over-production, psoriasis is largely driven by Th17 T cells and associated IL-17 activation [79]. IL-17 expression is also enhanced in acute lesions in AD skin compared to uninvolved skin [80], and a correlation between the number of Th17 cells in peripheral blood and acute AD severity has been reported [81]. However, secukinumab, the monoclonal antibody targeting IL-17, did not provide significant improvements in patients with moderate-to-severe AD compared to the effects of placebo in a phase 2 trial [26].

4. Targeting Immunomodulatory Effects and Sphingosine 1-Phosphate (S1P) Receptors (S1PRs)

S1P, a bioactive lipid mediator, regulates various cell activities, including cell growth, differentiation, apoptosis, migration, inflammation, metabolism, and angiogenesis [82,83,84]. S1P is secreted by red blood cells, endothelial cells, and platelets into the extracellular environment, and it contributes to several cardiovascular, autoimmune, inflammatory, neurological, oncologic, and fibrotic diseases [85]. In patients with AD, it has been reported that serum S1P levels are elevated and associated with severity [86]. Five subtypes of S1PRs (S1PR1–5) have been identified as seven-membrane-spanning proteins, a characteristic feature of G protein-coupled receptors. S1PR1, S1PR2, and S1PR3 are widely expressed in various tissues, including the brain, lungs, spleen, heart, and kidneys [87]. Unlike S1PR1–3, S1PR4 is expressed in the lungs and lymphoid tissues, and S1PR5 expresses in the brain and skin [87]. Igawa et al. reported that the expression of S1PR1 and S1PR2 is increased in impetigo, a common bacterial skin infection mostly caused by Staphylococcus aureus [88]. S1PRs are considered therapeutic targets for patients with AD because agents targeting S1PRs have displayed immunomodulatory effects [89]. In addition, a study using mice reported that S1PR3–TRPA1 signaling contributes to the onset of itches in sensory nerves [90]. Currently, the safety and efficacy of systemic treatment with etrasimod, which targets S1PR1, S1PR4, and S1PR5, has been illustrated in patients with moderate-to-severe AD in a phase 2 clinical trial (NCT04162769), opening the door for this compound to enter phase 3 development.

5. Small-Molecule Inhibitors

5.1. Janus Kinase (JAK) Inhibitors

IL-4, IL-13, IL-31, and TSLP require downstream JAK-signal transducer and activator of transcription (STAT) signaling [91]. The involvement of all four JAK family members (JAK1–3 and TYK) has been observed in AD, mediating downstream inflammation [92,93]. Phosphorylation of JAK following the binding of a cytokine to its cognate receptor induces the phosphorylation and dimerization of STAT proteins [94]. These STAT proteins regulate target genes after translocating to the nucleus [94,95]. JAK inhibitors inhibit the activity of one or more JAKs, thereby interfering with the JAK–STAT signaling pathway (Figure 2). IL-4 and IL-13 induce JAK1 and JAK3, which activate STAT6 [96]. TSLP and IL-31 induce JAK1 and JAK2 expression, which activates STAT5 [91]. The oral JAK inhibitors baricitinib (JAK1/2), abrocitinib (JAK1-selective), and upadacitinib (JAK1-selective) have been approved for the treatment of AD. All three met primary and secondary endpoints across numerous trials in moderate-to-severe AD [94]. Of patients receiving baricitinib at doses of 1, 2, and 4 mg, EASI-75 scores were significantly higher with the 2 and 4 mg dosages (17% and 21%) than placebo (6%) at week 16 in a phase 3 trial (BREEZE-AD2) [27]. Of patients receiving abrocitinib at doses of 100 and 200 mg, EASI-75 scores were significantly higher with both dosages (45% and 61%) than placebo (10%) at week 12 in a phase 3 trial (JADE-MONO2) [28]. Of patients receiving upadacitinib at doses of 15 and 30 mg, EASI-75 scores were significantly higher with both dosages (60% and 73%) than placebo (13%) at week 16 in a phase 3 trial [29]. These results highlight the importance of Th2 signaling in the pathogenesis of AD. In addition, topical JAK inhibitors such as ruxolitinib (JAK1/2) and delgocitinib (a JAK1/2/3 and Tyk2 inhibitor, i.e., pan-JAK) have also been approved. Ruxolitinib, a first-generation small molecule-inhibitor approved by the FDA, was well tolerated and associated with a low frequency of treatment-emergent adverse events in patients with mild-to-moderate AD [31,32]. Delgocitinib, the world’s first approved topical JAK inhibitor, has been studied in Japan, where it was approved for treating AD in adults and children based on long-term efficacy and safety data [33,34,35].

5.2. PDE4 Inhibitors

PDE4 is a key regulator of inflammatory cytokine production in AD through the degradation of cyclic adenosine monophosphate [97,98]. PDE4 inhibitors increase the levels of cyclic adenosine monophosphate in patients with AD and thereby reduce the expression of pro-inflammatory cytokines [99]. The systemic PDE4 inhibitor apremilast did not meet its primary endpoint for patients with moderate-to-severe AD in a double-blind, placebo-controlled PoC trial (NCT02087943) [30]. Conversely, the topical agents crisaborole and difamilast were approved for treating AD in adults and children based on long-term efficacy and safety data in phase 3 trials [15,16,36].

6. AhR-Modulating Agent

Tapinarof (GSK2894512, previously WBI-1001) is a naturally derived small molecule produced by bacterial symbionts of entomopathogenic nematodes [100]. It directly binds AhR and activates signaling in multiple cell types, including CD4+ T cells and keratinocytes [101]. The ligation of tapinarof and AhR improves the expression of skin barrier genes, regulates the expression of Th2 cytokines, and protects against inflammation-associated oxidative damage [101]. A phase 2b trial revealed that topical tapinarof improved both eczema area and severity index and itch numerical rating scale scores in patients with moderate-to-severe AD with largely mild adverse events [17].

7. Conclusions

Emerging topical and systemic targeted agents have been developed on the basis of expanding knowledge of the pathogenesis of AD. These specific cytokine/receptor-targeted agents have displayed safety and efficacy. Moreover, upcoming trials will provide additional therapeutic options for patients with AD. These new therapies also raise problems, such as the long-term socioeconomic burden associated with monoclonal antibody treatments. Thus, we need to choose more appropriate treatments, including combinations of existing therapies. We are currently at the dawn of a new era in the treatment of AD.

Author Contributions

Conceptualization, K.Y. and T.N.; writing—original draft preparation, K.Y.; writing—review and editing, K.Y. and T.N.; supervision, T.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors state no conflict of interest.

References

  1. Czarnowicki, T.; Krueger, J.G.; Guttman-Yassky, E. Skin barrier and immune dysregulation in atopic dermatitis: An evolving story with important clinical implications. J. Allergy Clin. Immunol. Pract. 2014, 2, 371–379, quiz 380–371. [Google Scholar] [CrossRef] [PubMed]
  2. Sacotte, R.; Silverberg, J.I. Epidemiology of adult atopic dermatitis. Clin. Dermatol. 2018, 36, 595–605. [Google Scholar] [CrossRef] [PubMed]
  3. Garg, N.; Silverberg, J.I. Epidemiology of childhood atopic dermatitis. Clin. Dermatol. 2015, 33, 281–288. [Google Scholar] [CrossRef] [PubMed]
  4. Chiesa Fuxench, Z.C.; Block, J.K.; Boguniewicz, M.; Boyle, J.; Fonacier, L.; Gelfand, J.M.; Grayson, M.H.; Margolis, D.J.; Mitchell, L.; Silverberg, J.I.; et al. Atopic Dermatitis in America Study: A Cross-Sectional Study Examining the Prevalence and Disease Burden of Atopic Dermatitis in the US Adult Population. J. Investig. Dermatol. 2019, 139, 583–590. [Google Scholar] [CrossRef] [Green Version]
  5. Hanifin, J.M.; Reed, M.L. A population-based survey of eczema prevalence in the United States. Dermatitis 2007, 18, 82–91. [Google Scholar] [CrossRef]
  6. Puar, N.; Chovatiya, R.; Paller, A.S. New treatments in atopic dermatitis. Ann. Allergy Asthma Immunol. 2021, 126, 21–31. [Google Scholar] [CrossRef] [PubMed]
  7. He, H.; Del Duca, E.; Diaz, A.; Gay-Mimbrera, J.; Zhang, N.; Wu, J.; Beaziz, J.; Estrada, Y.; Krueger, J.G.; Pavel, A.B.; et al. Mild atopic dermatitis lacks systemic inflammation and shows reduced nonlesional skin abnormalities. J. Allergy Clin. Immunol. 2020, 147, 1369–1380. [Google Scholar] [CrossRef] [PubMed]
  8. Czarnowicki, T.; Gonzalez, J.; Shemer, A.; Malajian, D.; Xu, H.; Zheng, X.; Khattri, S.; Gilleaudeau, P.; Sullivan-Whalen, M.; Suárez-Fariñas, M.; et al. Severe atopic dermatitis is characterized by selective expansion of circulating TH2/TC2 and TH22/TC22, but not TH17/TC17, cells within the skin-homing T-cell population. J. Allergy Clin. Immunol. 2015, 136, 104–115.e7. [Google Scholar] [CrossRef]
  9. Czarnowicki, T.; Gonzalez, J.; Bonifacio, K.M.; Shemer, A.; Xiangyu, P.; Kunjravia, N.; Malajian, D.; Fuentes-Duculan, J.; Esaki, H.; Noda, S.; et al. Diverse activation and differentiation of multiple B-cell subsets in patients with atopic dermatitis but not in patients with psoriasis. J. Allergy Clin. Immunol. 2016, 137, 118–129.e5. [Google Scholar] [CrossRef]
  10. Brunner, P.M.; Suárez-Fariñas, M.; He, H.; Malik, K.; Wen, H.C.; Gonzalez, J.; Chan, T.C.; Estrada, Y.; Zheng, X.; Khattri, S.; et al. The atopic dermatitis blood signature is characterized by increases in inflammatory and cardiovascular risk proteins. Sci. Rep. 2017, 7, 8707. [Google Scholar] [CrossRef]
  11. Thijs, J.L.; Strickland, I.; Bruijnzeel-Koomen, C.A.F.M.; Nierkens, S.; Giovannone, B.; Knol, E.F.; Csomor, E.; Sellman, B.R.; Mustelin, T.; Sleeman, M.A.; et al. Serum biomarker profiles suggest that atopic dermatitis is a systemic disease. J. Allergy Clin. Immunol. 2018, 141, 1523–1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pavel, A.B.; Zhou, L.; Diaz, A.; Ungar, B.; Dan, J.; He, H.; Estrada, Y.D.; Xu, H.; Fernandes, M.; Renert-Yuval, Y.; et al. The proteomic skin profile of moderate-to-severe atopic dermatitis patients shows an inflammatory signature. J. Am. Acad. Dermatol. 2020, 82, 690–699. [Google Scholar] [CrossRef]
  13. He, H.; Li, R.; Choi, S.; Zhou, L.; Pavel, A.; Estrada, Y.D.; Krueger, J.G.; Guttman-Yassky, E. Increased cardiovascular and atherosclerosis markers in blood of older patients with atopic dermatitis. Ann. Allergy Asthma Immunol. 2020, 124, 70–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Raap, U.; Wichmann, K.; Bruder, M.; Ständer, S.; Wedi, B.; Kapp, A.; Werfel, T. Correlation of IL-31 serum levels with severity of atopic dermatitis. J. Allergy Clin. Immunol. 2008, 122, 421–423. [Google Scholar] [CrossRef] [PubMed]
  15. Saeki, H.; Imamura, T.; Yokota, D.; Tsubouchi, H. Difamilast Ointment in Japanese Adult and Pediatric Patients with Atopic Dermatitis: A Phase III, Long-Term, Open-Label Study. Dermatol. Ther. 2022, 12, 1589–1601. [Google Scholar] [CrossRef]
  16. Saeki, H.; Baba, N.; Ito, K.; Yokota, D.; Tsubouchi, H. Difamilast, a selective phosphodiesterase 4 inhibitor, ointment in paediatric patients with atopic dermatitis: A phase III randomized double-blind, vehicle-controlled trial. Br. J. Dermatol. 2022, 186, 40–49. [Google Scholar] [CrossRef]
  17. Paller, A.S.; Stein Gold, L.; Soung, J.; Tallman, A.M.; Rubenstein, D.S.; Gooderham, M. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J. Am. Acad. Dermatol. 2021, 84, 632–638. [Google Scholar] [CrossRef]
  18. 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. [Google Scholar] [CrossRef]
  19. Freitas, E.; Guttman-Yassky, E.; Torres, T. Tralokinumab for the Treatment of Atopic Dermatitis. Am. J. Clin. Dermatol. 2021, 22, 625–638. [Google Scholar] [CrossRef] [PubMed]
  20. 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. [Google Scholar] [CrossRef]
  21. Chen, Y.L.; Gutowska-Owsiak, D.; Hardman, C.S.; Westmoreland, M.; MacKenzie, T.; Cifuentes, L.; Waithe, D.; Lloyd-Lavery, A.; Marquette, A.; Londei, M.; et al. Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Sci. Transl. Med. 2019, 11, eaax2945. [Google Scholar] [CrossRef]
  22. van de Veen, W.; Akdis, M. The use of biologics for immune modulation in allergic disease. J. Clin. Investig. 2019, 129, 1452–1462. [Google Scholar] [CrossRef] [Green Version]
  23. Simpson, E.L.; Parnes, J.R.; She, D.; Crouch, S.; Rees, W.; Mo, M.; van der Merwe, R. Tezepelumab, an anti-thymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: A randomized phase 2a clinical trial. J. Am. Acad. Dermatol. 2019, 80, 1013–1021. [Google Scholar] [CrossRef] [Green Version]
  24. Guttman-Yassky, E.; Pavel, A.B.; Zhou, L.; Zhou, L.; Estrada, Y.D.; Zhang, N.; Xu, H.; Peng, X.; Wen, H.C.; Govas, P.; et al. GBR 830, an anti-OX40, improves skin gene signatures and clinical scores in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2019, 144, 482–493.e7. [Google Scholar] [CrossRef] [Green Version]
  25. Kabashima, K.; Matsumura, T.; Komazaki, H.; Kawashima, M. Trial of Nemolizumab and Topical Agents for Atopic Dermatitis with Pruritus. N. Engl. J. Med. 2020, 383, 141–150. [Google Scholar] [CrossRef]
  26. Ungar, B.; Pavel, A.B.; Li, R.; Kimmel, G.; Nia, J.; Hashim, P.; Kim, H.J.; Chima, M.; Vekaria, A.S.; Estrada, Y.; et al. Phase 2 randomized, double-blind study of IL-17 targeting with secukinumab in atopic dermatitis. J. Allergy Clin. Immunol. 2021, 147, 394–397. [Google Scholar] [CrossRef]
  27. Simpson, E.L.; Lacour, J.P.; Spelman, L.; Galimberti, R.; Eichenfield, L.F.; Bissonnette, R.; King, B.A.; Thyssen, J.P.; Silverberg, J.I.; Bieber, T.; et al. Baricitinib in patients with moderate-to-severe atopic dermatitis and inadequate response to topical corticosteroids: Results from two randomized monotherapy phase III trials. Br. J. Dermatol. 2020, 183, 242–255. [Google Scholar] [CrossRef]
  28. Silverberg, J.I.; Simpson, E.L.; Thyssen, J.P.; Gooderham, M.; Chan, G.; Feeney, C.; Biswas, P.; Valdez, H.; DiBonaventura, M.; Nduaka, C.; et al. Efficacy and Safety of Abrocitinib in Patients With Moderate-to-Severe Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2020, 156, 863–873. [Google Scholar] [CrossRef]
  29. Guttman-Yassky, E.; Teixeira, H.D.; Simpson, E.L.; Papp, K.A.; Pangan, A.L.; Blauvelt, A.; Thaç, D.; Chu, C.-Y.; Hong, H.C.; Katoh, N.; et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): Results from two replicate double-blind, randomised controlled phase 3 trials. Lancet 2021, 397, 2151–2168. [Google Scholar] [CrossRef]
  30. Samrao, A.; Berry, T.M.; Goreshi, R.; Simpson, E.L. A pilot study of an oral phosphodiesterase inhibitor (apremilast) for atopic dermatitis in adults. Arch. Dermatol. 2012, 148, 890–897. [Google Scholar] [CrossRef]
  31. Kim, B.S.; Howell, M.D.; Sun, K.; Papp, K.; Nasir, A.; Kuligowski, M.E. Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2 inhibitor) or triamcinolone cream. J. Allergy Clin. Immunol. 2020, 145, 572–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kim, B.S.; Sun, K.; Papp, K.; Venturanza, M.; Nasir, A.; Kuligowski, M.E. Effects of ruxolitinib cream on pruritus and quality of life in atopic dermatitis: Results from a phase 2, randomized, dose-ranging, vehicle- and active-controlled study. J. Am. Acad. Dermatol. 2020, 82, 1305–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Nakagawa, H.; Nemoto, O.; Igarashi, A.; Saeki, H.; Kabashima, K.; Oda, M.; Nagata, T. Delgocitinib ointment in pediatric patients with atopic dermatitis: A phase 3, randomized, double-blind, vehicle-controlled study and a subsequent open-label, long-term study. J. Am. Acad. Dermatol. 2021, 85, 854–862. [Google Scholar] [CrossRef] [PubMed]
  34. Nakagawa, H.; Nemoto, O.; Igarashi, A.; Saeki, H.; Kaino, H.; Nagata, T. Delgocitinib ointment, a topical Janus kinase inhibitor, in adult patients with moderate to severe atopic dermatitis: A phase 3, randomized, double-blind, vehicle-controlled study and an open-label, long-term extension study. J. Am. Acad. Dermatol. 2020, 82, 823–831. [Google Scholar] [CrossRef] [Green Version]
  35. Nakagawa, H.; Nemoto, O.; Igarashi, A.; Saeki, H.; Murata, R.; Kaino, H.; Nagata, T. Long-term safety and efficacy of delgocitinib ointment, a topical Janus kinase inhibitor, in adult patients with atopic dermatitis. J. Dermatol. 2020, 47, 114–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Paller, A.S.; Tom, W.L.; Lebwohl, M.G.; Blumenthal, R.L.; Boguniewicz, M.; Call, R.S.; Eichenfield, L.F.; Forsha, D.W.; Rees, W.C.; Simpson, E.L.; et al. Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J. Am. Acad. Dermatol. 2016, 75, 494–503.e496. [Google Scholar] [CrossRef] [Green Version]
  37. Gittler, J.K.; Shemer, A.; Suárez-Fariñas, M.; Fuentes-Duculan, J.; Gulewicz, K.J.; Wang, C.Q.; Mitsui, H.; Cardinale, I.; de Guzman Strong, C.; Krueger, J.G.; et al. Progressive activation of T(H)2/T(H)22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. J. Allergy Clin. Immunol. 2012, 130, 1344–1354. [Google Scholar] [CrossRef] [Green Version]
  38. Boguniewicz, M.; Fonacier, L.; Guttman-Yassky, E.; Ong, P.Y.; Silverberg, J.; Farrar, J.R. Atopic dermatitis yardstick: Practical recommendations for an evolving therapeutic landscape. Ann. Allergy Asthma Immunol. 2018, 120, 10–22.e2. [Google Scholar] [CrossRef] [Green Version]
  39. 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. [Google Scholar] [CrossRef] [Green Version]
  40. 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. [Google Scholar] [CrossRef]
  41. Akdis, C.A.; Arkwright, P.D.; Brüggen, M.C.; Busse, W.; Gadina, M.; Guttman-Yassky, E.; Kabashima, K.; Mitamura, Y.; Vian, L.; Wu, J.; et al. Type 2 immunity in the skin and lungs. Allergy 2020, 75, 1582–1605. [Google Scholar] [CrossRef] [PubMed]
  42. Roediger, B.; Kyle, R.; Le Gros, G.; Weninger, W. Dermal group 2 innate lymphoid cells in atopic dermatitis and allergy. Curr. Opin. Immunol. 2014, 31, 108–114. [Google Scholar] [CrossRef] [PubMed]
  43. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.; Mebius, R.E.; et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef] [PubMed]
  44. Moro, K.; Yamada, T.; Tanabe, M.; Takeuchi, T.; Ikawa, T.; Kawamoto, H.; Furusawa, J.; Ohtani, M.; Fujii, H.; Koyasu, S. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells. Nature 2010, 463, 540–544. [Google Scholar] [CrossRef] [PubMed]
  45. Hoyler, T.; Klose, C.S.; Souabni, A.; Turqueti-Neves, A.; Pfeifer, D.; Rawlins, E.L.; Voehringer, D.; Busslinger, M.; Diefenbach, A. The transcription factor GATA-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity 2012, 37, 634–648. [Google Scholar] [CrossRef] [Green Version]
  46. Neill, D.R.; Wong, S.H.; Bellosi, A.; Flynn, R.J.; Daly, M.; Langford, T.K.; Bucks, C.; Kane, C.M.; Fallon, P.G.; Pannell, R.; et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 2010, 464, 1367–1370. [Google Scholar] [CrossRef] [Green Version]
  47. Price, A.E.; Liang, H.E.; Sullivan, B.M.; Reinhardt, R.L.; Eisley, C.J.; Erle, D.J.; Locksley, R.M. Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc. Natl. Acad. Sci. USA 2010, 107, 11489–11494. [Google Scholar] [CrossRef] [Green Version]
  48. Imai, Y. Interleukin-33 in atopic dermatitis. J. Dermatol. Sci. 2019, 96, 2–7. [Google Scholar] [CrossRef] [Green Version]
  49. Salimi, M.; Barlow, J.L.; Saunders, S.P.; Xue, L.; Gutowska-Owsiak, D.; Wang, X.; Huang, L.C.; Johnson, D.; Scanlon, S.T.; McKenzie, A.N.; et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 2013, 210, 2939–2950. [Google Scholar] [CrossRef]
  50. Liu, Y.J. Thymic stromal lymphopoietin: Master switch for allergic inflammation. J. Exp. Med. 2006, 203, 269–273. [Google Scholar] [CrossRef]
  51. Liu, Y.J. Thymic stromal lymphopoietin and OX40 ligand pathway in the initiation of dendritic cell-mediated allergic inflammation. J. Allergy Clin. Immunol. 2007, 120, 238–244, quiz 245–236. [Google Scholar] [CrossRef] [PubMed]
  52. Webb, G.J.; Hirschfield, G.M.; Lane, P.J. OX40, OX40L and Autoimmunity: A Comprehensive Review. Clin. Rev. Allergy Immunol. 2016, 50, 312–332. [Google Scholar] [CrossRef] [PubMed]
  53. Stüber, E.; Neurath, M.; Calderhead, D.; Fell, H.P.; Strober, W. Cross-linking of OX40 ligand, a member of the TNF/NGF cytokine family, induces proliferation and differentiation in murine splenic B cells. Immunity 1995, 2, 507–521. [Google Scholar] [CrossRef] [Green Version]
  54. Sato, T.; Ishii, N.; Murata, K.; Kikuchi, K.; Nakagawa, S.; Ndhlovu, L.C.; Sugamura, K. Consequences of OX40-OX40 ligand interactions in langerhans cell function: Enhanced contact hypersensitivity responses in OX40L-transgenic mice. Eur. J. Immunol. 2002, 32, 3326–3335. [Google Scholar] [CrossRef]
  55. Ohshima, Y.; Tanaka, Y.; Tozawa, H.; Takahashi, Y.; Maliszewski, C.; Delespesse, G. Expression and function of OX40 ligand on human dendritic cells. J. Immunol. 1997, 159, 3838–3848. [Google Scholar]
  56. Sugamura, K.; Ishii, N.; Weinberg, A.D. Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40. Nat. Rev. Immunol. 2004, 4, 420–431. [Google Scholar] [CrossRef]
  57. Gramaglia, I.; Jember, A.; Pippig, S.D.; Weinberg, A.D.; Killeen, N.; Croft, M. The OX40 costimulatory receptor determines the development of CD4 memory by regulating primary clonal expansion. J. Immunol. 2000, 165, 3043–3050. [Google Scholar] [CrossRef] [Green Version]
  58. Maxwell, J.R.; Weinberg, A.; Prell, R.A.; Vella, A.T. Danger and OX40 receptor signaling synergize to enhance memory T cell survival by inhibiting peripheral deletion. J. Immunol. 2000, 164, 107–112. [Google Scholar] [CrossRef] [Green Version]
  59. Rogers, P.R.; Song, J.; Gramaglia, I.; Killeen, N.; Croft, M. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 2001, 15, 445–455. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, A.I.; McAdam, A.J.; Buhlmann, J.E.; Scott, S.; Lupher, M.L., Jr.; Greenfield, E.A.; Baum, P.R.; Fanslow, W.C.; Calderhead, D.M.; Freeman, G.J.; et al. Ox40-ligand has a critical costimulatory role in dendritic cell: T cell interactions. Immunity 1999, 11, 689–698. [Google Scholar] [CrossRef] [Green Version]
  61. Murata, K.; Ishii, N.; Takano, H.; Miura, S.; Ndhlovu, L.C.; Nose, M.; Noda, T.; Sugamura, K. Impairment of antigen-presenting cell function in mice lacking expression of OX40 ligand. J. Exp. Med. 2000, 191, 365–374. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, Q.; Putheti, P.; Zhou, Q.; Liu, Q.; Gao, W. Structures and biological functions of IL-31 and IL-31 receptors. Cytokine Growth Factor Rev. 2008, 19, 347–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bilsborough, J.; Leung, D.Y.; Maurer, M.; Howell, M.; Boguniewicz, M.; Yao, L.; Storey, H.; LeCiel, C.; Harder, B.; Gross, J.A. IL-31 is associated with cutaneous lymphocyte antigen-positive skin homing T cells in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2006, 117, 418–425. [Google Scholar] [CrossRef] [PubMed]
  64. Dillon, S.R.; Sprecher, C.; Hammond, A.; Bilsborough, J.; Rosenfeld-Franklin, M.; Presnell, S.R.; Haugen, H.S.; Maurer, M.; Harder, B.; Johnston, J.; et al. Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat. Immunol. 2004, 5, 752–760. [Google Scholar] [CrossRef] [PubMed]
  65. Cornelissen, C.; Lüscher-Firzlaff, J.; Baron, J.M.; Lüscher, B. Signaling by IL-31 and functional consequences. Eur. J. Cell Biol. 2012, 91, 552–566. [Google Scholar] [CrossRef]
  66. Cevikbas, F.; Wang, X.; Akiyama, T.; Kempkes, C.; Savinko, T.; Antal, A.; Kukova, G.; Buhl, T.; Ikoma, A.; Buddenkotte, J.; et al. A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: Involvement of TRPV1 and TRPA1. J. Allergy Clin. Immunol. 2014, 133, 448–460. [Google Scholar] [CrossRef] [Green Version]
  67. Sonkoly, E.; Muller, A.; Lauerma, A.I.; Pivarcsi, A.; Soto, H.; Kemeny, L.; Alenius, H.; Dieu-Nosjean, M.C.; Meller, S.; Rieker, J.; et al. IL-31: A new link between T cells and pruritus in atopic skin inflammation. J. Allergy Clin. Immunol. 2006, 117, 411–417. [Google Scholar] [CrossRef]
  68. Raap, U.; Gehring, M.; Kleiner, S.; Rüdrich, U.; Eiz-Vesper, B.; Haas, H.; Kapp, A.; Gibbs, B.F. Human basophils are a source of-and are differentially activated by-IL-31. Clin. Exp. Allergy 2017, 47, 499–508. [Google Scholar] [CrossRef] [Green Version]
  69. Kato, A.; Fujii, E.; Watanabe, T.; Takashima, Y.; Matsushita, H.; Furuhashi, T.; Morita, A. Distribution of IL-31 and its receptor expressing cells in skin of atopic dermatitis. J. Dermatol. Sci. 2014, 74, 229–235. [Google Scholar] [CrossRef]
  70. Furue, M.; Yamamura, K.; Kido-Nakahara, M.; Nakahara, T.; Fukui, Y. Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy 2018, 73, 29–36. [Google Scholar] [CrossRef] [Green Version]
  71. Yamamura, K.; Uruno, T.; Shiraishi, A.; Tanaka, Y.; Ushijima, M.; Nakahara, T.; Watanabe, M.; Kido-Nakahara, M.; Tsuge, I.; Furue, M.; et al. The transcription factor EPAS1 links DOCK8 deficiency to atopic skin inflammation via IL-31 induction. Nat. Commun. 2017, 8, 13946. [Google Scholar] [CrossRef]
  72. Kamikaseda, Y.; Uruno, T.; Kunimura, K.; Harada, A.; Saiki, K.; Oisaki, K.; Sakata, D.; Nakahara, T.; Kido-Nakahara, M.; Kanai, M.; et al. Targeted inhibition of EPAS1-driven IL-31 production by a small-molecule compound. J. Allergy Clin. Immunol. 2021, 148, 633–638. [Google Scholar] [CrossRef] [PubMed]
  73. Engelhardt, K.R.; Gertz, M.E.; Keles, S.; Schäffer, A.A.; Sigmund, E.C.; Glocker, C.; Saghafi, S.; Pourpak, Z.; Ceja, R.; Sassi, A.; et al. The extended clinical phenotype of 64 patients with dedicator of cytokinesis 8 deficiency. J. Allergy Clin. Immunol. 2015, 136, 402–412. [Google Scholar] [CrossRef] [PubMed]
  74. Freeman, A.F.; Holland, S.M. Clinical manifestations of hyper IgE syndromes. Dis. Markers 2010, 29, 123–130. [Google Scholar] [CrossRef] [PubMed]
  75. Yong, P.F.; Freeman, A.F.; Engelhardt, K.R.; Holland, S.; Puck, J.M.; Grimbacher, B. An update on the hyper-IgE syndromes. Arthritis Res. Ther. 2012, 14, 228. [Google Scholar] [CrossRef] [Green Version]
  76. Zhang, Q.; Davis, J.C.; Lamborn, I.T.; Freeman, A.F.; Jing, H.; Favreau, A.J.; Matthews, H.F.; Davis, J.; Turner, M.L.; Uzel, G.; et al. Combined immunodeficiency associated with DOCK8 mutations. N. Engl. J. Med. 2009, 361, 2046–2055. [Google Scholar] [CrossRef] [Green Version]
  77. Zhang, Q.; Davis, J.C.; Dove, C.G.; Su, H.C. Genetic, clinical, and laboratory markers for DOCK8 immunodeficiency syndrome. Dis. Markers 2010, 29, 131–139. [Google Scholar]
  78. Kunimura, K.; Yamamura, K.; Nakahara, T.; Kido-Nakahara, M.; Uruno, T.; Fukui, Y. Identification of a functional DOCK8 gene polymorphism associated with atopic dermatitis. Allergy 2022. epub ahead of print. [Google Scholar] [CrossRef]
  79. Guttman-Yassky, E.; Krueger, J.G. Atopic dermatitis and psoriasis: Two different immune diseases or one spectrum? Curr. Opin. Immunol. 2017, 48, 68–73. [Google Scholar] [CrossRef]
  80. Toda, M.; Leung, D.Y.; Molet, S.; Boguniewicz, M.; Taha, R.; Christodoulopoulos, P.; Fukuda, T.; Elias, J.A.; Hamid, Q.A. Polarized in vivo expression of IL-11 and IL-17 between acute and chronic skin lesions. J. Allergy Clin. Immunol. 2003, 111, 875–881. [Google Scholar] [CrossRef]
  81. Koga, C.; Kabashima, K.; Shiraishi, N.; Kobayashi, M.; Tokura, Y. Possible pathogenic role of Th17 cells for atopic dermatitis. J Investig. Dermatol. 2008, 128, 2625–2630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Coant, N.; Sakamoto, W.; Mao, C.; Hannun, Y.A. Ceramidases, roles in sphingolipid metabolism and in health and disease. Adv. Biol. Regul. 2017, 63, 122–131. [Google Scholar] [CrossRef] [PubMed]
  83. Nema, R.; Vishwakarma, S.; Agarwal, R.; Panday, R.K.; Kumar, A. Emerging role of sphingosine-1-phosphate signaling in head and neck squamous cell carcinoma. Onco Targets Ther. 2016, 9, 3269–3280. [Google Scholar]
  84. Henkel, F.D.R.; Friedl, A.; Haid, M.; Thomas, D.; Bouchery, T.; Haimerl, P.; de Los Reyes Jiménez, M.; Alessandrini, F.; Schmidt-Weber, C.B.; Harris, N.L.; et al. House dust mite drives proinflammatory eicosanoid reprogramming and macrophage effector functions. Allergy 2019, 74, 1090–1101. [Google Scholar]
  85. Proia, R.L.; Hla, T. Emerging biology of sphingosine-1-phosphate: Its role in pathogenesis and therapy. J. Clin. Investig. 2015, 125, 1379–1387. [Google Scholar] [CrossRef] [Green Version]
  86. Sakai, T.; Herrmann, N.; Maintz, L.; Nümm, T.J.; Welchowski, T.; Claus, R.A.; Gräler, M.H.; Bieber, T. Serum sphingosine-1-phosphate is elevated in atopic dermatitis and associated with severity. Allergy 2021, 76, 2592–2595. [Google Scholar] [CrossRef] [PubMed]
  87. Brinkmann, V. Sphingosine 1-phosphate receptors in health and disease: Mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol. Ther. 2007, 115, 84–105. [Google Scholar] [CrossRef]
  88. Igawa, S.; Choi, J.E.; Wang, Z.; Chang, Y.L.; Wu, C.C.; Werbel, T.; Ishida-Yamamoto, A.; Di Nardo, A. Human Keratinocytes Use Sphingosine 1-Phosphate and its Receptors to Communicate Staphylococcus aureus Invasion and Activate Host Defense. J. Investig. Dermatol. 2019, 139, 1743–1752.e5. [Google Scholar] [CrossRef]
  89. Cartier, A.; Hla, T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019, 366, eaar5551. [Google Scholar] [CrossRef]
  90. Hill, R.Z.; Morita, T.; Brem, R.B.; Bautista, D.M. S1PR3 Mediates Itch and Pain via Distinct TRP Channel-Dependent Pathways. J. Neurosci. 2018, 38, 7833–7843. [Google Scholar] [CrossRef] [Green Version]
  91. Howell, M.D.; Kuo, F.I.; Smith, P.A. Targeting the Janus Kinase Family in Autoimmune Skin Diseases. Front. Immunol. 2019, 10, 2342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Villarino, A.V.; Kanno, Y.; O’Shea, J.J. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat. Immunol. 2017, 18, 374–384. [Google Scholar] [CrossRef] [PubMed]
  93. Bao, L.; Zhang, H.; Chan, L.S. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. Jak-Stat 2013, 2, e24137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Chovatiya, R.; Paller, A.S. JAK inhibitors in the treatment of atopic dermatitis. J. Allergy Clin. Immunol. 2021, 148, 927–940. [Google Scholar] [CrossRef]
  95. Choy, E.H. Clinical significance of Janus Kinase inhibitor selectivity. Rheumatology 2019, 58, 953–962. [Google Scholar] [CrossRef] [Green Version]
  96. Rodrigues, M.A.; Torres, T. JAK/STAT inhibitors for the treatment of atopic dermatitis. J. Dermatol. Treat. 2020, 31, 33–40. [Google Scholar] [CrossRef]
  97. Jimenez, J.L.; Punzón, C.; Navarro, J.; Muñoz-Fernández, M.A.; Fresno, M. Phosphodiesterase 4 inhibitors prevent cytokine secretion by T lymphocytes by inhibiting nuclear factor-kappaB and nuclear factor of activated T cells activation. J. Pharmacol. Exp. Ther. 2001, 299, 753–759. [Google Scholar]
  98. Bäumer, W.; Hoppmann, J.; Rundfeldt, C.; Kietzmann, M. Highly selective phosphodiesterase 4 inhibitors for the treatment of allergic skin diseases and psoriasis. Inflamm. Allergy Drug Targets 2007, 6, 17–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Zane, L.T.; Chanda, S.; Jarnagin, K.; Nelson, D.B.; Spelman, L.; Gold, L.S. Crisaborole and its potential role in treating atopic dermatitis: Overview of early clinical studies. Immunotherapy 2016, 8, 853–866. [Google Scholar] [CrossRef] [Green Version]
  100. Richardson, W.H.; Schmidt, T.M.; Nealson, K.H. Identification of an anthraquinone pigment and a hydroxystilbene antibiotic from Xenorhabdus luminescens. Appl. Environ. Microbiol. 1988, 54, 1602–1605. [Google Scholar] [CrossRef] [Green Version]
  101. Smith, S.H.; Jayawickreme, C.; Rickard, D.J.; Nicodeme, E.; Bui, T.; Simmons, C.; Coquery, C.M.; Neil, J.; Pryor, W.M.; Mayhew, D.; et al. Tapinarof Is a Natural AhR Agonist that Resolves Skin Inflammation in Mice and Humans. J. Investig. Dermatol. 2017, 137, 2110–2119. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Therapies based on the pathogenesis of atopic dermatitis. Disrupted epidermal barrier function and microbial dysbiosis induce the production of pro-inflammatory mediators. Keratinocyte-produced TSLP and IL-33 enhance type 2 inflammatory responses through the activation of Th2 cells and ILC2s. Th2 cells and ILC2s produce the key inflammatory cytokines (IL-4 and/or IL-13) of AD. The ligation of OX40L and OX40 augments Th2 immune responses. IL-31 is a T cell-derived cytokine associated with pruritus. IL-31 transmits itch sensations via IL-31R in peripheral neurons. The lipid mediator S1P regulates various cell activities, including cell growth, differentiation, apoptosis, migration, inflammation, metabolism, and angiogenesis, through S1PRs. TSLP, thymic stromal lymphopoietin; IL, interleukin; Th, T-helper cells; ILC2s, group 2 innate lymphoid cells; OX40L, OX40 ligand; IL-31R, IL-31 receptor; S1P, sphingosine 1-phosphate.
Figure 1. Therapies based on the pathogenesis of atopic dermatitis. Disrupted epidermal barrier function and microbial dysbiosis induce the production of pro-inflammatory mediators. Keratinocyte-produced TSLP and IL-33 enhance type 2 inflammatory responses through the activation of Th2 cells and ILC2s. Th2 cells and ILC2s produce the key inflammatory cytokines (IL-4 and/or IL-13) of AD. The ligation of OX40L and OX40 augments Th2 immune responses. IL-31 is a T cell-derived cytokine associated with pruritus. IL-31 transmits itch sensations via IL-31R in peripheral neurons. The lipid mediator S1P regulates various cell activities, including cell growth, differentiation, apoptosis, migration, inflammation, metabolism, and angiogenesis, through S1PRs. TSLP, thymic stromal lymphopoietin; IL, interleukin; Th, T-helper cells; ILC2s, group 2 innate lymphoid cells; OX40L, OX40 ligand; IL-31R, IL-31 receptor; S1P, sphingosine 1-phosphate.
Jcm 11 06145 g001
Figure 2. JAK–STAT signaling and oral/topical JAK inhibitors in atopic dermatitis. The JAK protein family (JAK1, JAK2, JAK3, and TYK2) mediates IL-4, IL-13, and IL-31 cytokine signaling via cognate receptors. Activation of JAKs results in the phosphorylation of downstream STAT proteins, followed by their nuclear translocation and activation of target genes. JAK inhibitors inhibit the activity of one or more JAKs, thereby interfering with the JAK–STAT signaling pathway. JAK, Janus kinase; STAT, signal transducer and activator of transcription; TYK2, tyrosine kinase 2; IL, interleukin.
Figure 2. JAK–STAT signaling and oral/topical JAK inhibitors in atopic dermatitis. The JAK protein family (JAK1, JAK2, JAK3, and TYK2) mediates IL-4, IL-13, and IL-31 cytokine signaling via cognate receptors. Activation of JAKs results in the phosphorylation of downstream STAT proteins, followed by their nuclear translocation and activation of target genes. JAK inhibitors inhibit the activity of one or more JAKs, thereby interfering with the JAK–STAT signaling pathway. JAK, Janus kinase; STAT, signal transducer and activator of transcription; TYK2, tyrosine kinase 2; IL, interleukin.
Jcm 11 06145 g002
Table 1. Emerging Systemic Agents.
Table 1. Emerging Systemic Agents.
AgentTargetStudy TypeStudy DurationDoseReference
BiologicsDupilumabIL-4RaPhase 316 weeks300 mg[18]
TralokinumabIL-13Phase 2b16 weeks150, 300 mg[19]
LebrikizumabIL-13Phase 2b16 weeks125, 250 mg[20]
EtokimabIL-33Phase 2a20 weeks300 mg[21,22]
TezepelumabTSLPPhase 2a12 weeks280 mg[23]
GBR 830OX40Phase 2a16 weeks10 mg/kg[24]
NemolizumabIL-31RaPhase 316 weeks60 mg[25]
Secukinumab IL-17APhase 216 weeks300 mg[26]
EtrasimodS1PR 1/4/5Phase 216 weeks1, 2 mgNCT04162769
Small-molecule inhibitorsBaricitinibJAK 1/2Phase 316 weeks1, 2, 4 mg[27]
AbrocitinibJAK1Phase 312 weeks100, 200 mg[28]
UpadacitinibJAK1Phase 316 weeks15, 30 mg[29]
ApremilastPDE4Phase 212 weeks30, 40 mg[30]
Table 2. Emerging Topical Agents.
Table 2. Emerging Topical Agents.
AgentTargetStudy TypeStudy DurationDoseReference
RuxolitinibJAK1/2Phase 212 weeks0.15, 0.5, 1.5%[31,32]
DelgocitinibPan-JAKPhase 328 weeks0.25, 0.5%[33,34,35]
CrisaborolePDE4Phase 34 weeks2.00%[36]
DifamilastPDE4Phase 34, 52 weeks0.3, 1.0%[15,16]
TapinarofAhRPhase 2b12 weeks0.5, 1.0%[17]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamamura, K.; Nakahara, T. The Dawn of a New Era in Atopic Dermatitis Treatment. J. Clin. Med. 2022, 11, 6145. https://doi.org/10.3390/jcm11206145

AMA Style

Yamamura K, Nakahara T. The Dawn of a New Era in Atopic Dermatitis Treatment. Journal of Clinical Medicine. 2022; 11(20):6145. https://doi.org/10.3390/jcm11206145

Chicago/Turabian Style

Yamamura, Kazuhiko, and Takeshi Nakahara. 2022. "The Dawn of a New Era in Atopic Dermatitis Treatment" Journal of Clinical Medicine 11, no. 20: 6145. https://doi.org/10.3390/jcm11206145

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop