• Open Access
    Review

    Revisiting immunotherapeutic strategies for the management of atopic dermatitis

    Gael Tchokomeni Siwe 1,2*
    Emmanuel Adebowale Fajemisin 1,2
    Masala Mugeri 1,2
    Krupa Naran 1,2
    Stefan Barth 1,2

    Explor Asthma Allergy. 2024;2:373–398 DOI: https://doi.org/10.37349/eaa.2024.00052

    Received: May 03, 2024 Accepted: June 18, 2024 Published: August 09, 2024

    Academic Editor: Somasundaram Arumugam, National Institute of Pharmaceutical Education and Research, India

    This article belongs to the special issue Atopic Dermatitis – Pathology and Treatment modalities

    Abstract

    Atopic dermatitis (AD) represents the most common inflammatory skin disease with a highly intricated immune fingerprint. Until recently, AD management mostly relied on topical corticosteroids, calcineurin inhibitors, and systemic immunosuppressants, with a range of safety and tolerability concerns including toxicity, drug interactions, and contraindications. With the onset of biologics, safer and more targeted therapeutics have become available, displaying various degrees of success in treating AD, but not yet able to meet all the needs of AD patients. Some of the challenges encountered included variability of responses among patients, long-term safety, and limited access due to prohibitive costs. As the pathophysiology of AD has been increasingly understood within the last years, new approaches are explored, leading to an unprecedented diversification of therapeutic options to address these hurdles. This review highlights current immunotherapeutic strategies developed towards AD, whether already in the clinical pipeline or still in preclinical exploration.

    Keywords

    Atopic dermatitis, chronic inflammation, immunotherapy, precision medicine

    Introduction

    Atopic dermatitis (AD), also referred to as atopic eczema, is a skin disease that is a very diverse in terms of ethnic-based phenotypic variability, age of onset, dynamic course of disease, risk of various cutaneous infections, atopic and non-atopic comorbidities [1]. AD, characterized by eosinophilic/spongiotic inflammation of the skin with characteristic age-dependent distribution patterns and morphology of lesions, is a chronically relapsing and intensely pruritic, Th2-driven skin disease often occurring in families with atopic diseases (AD, food allergy, bronchial asthma or allergic rhinoconjunctivitis). Although the etiology of AD is yet to be fully understood, it is commonly accepted that the pathophysiology of AD is driven by a combination of factors comprising epidermal barrier defects, genetic predisposition, immune dysregulation, imbalance of microbiome, and extrinsic stimuli like allergens, irritants, and microbes [2, 3]. According to the latest Global Burden of Disease study [4], AD represents 36.17% of all new immune-mediated inflammatory diseases reported across the world.

    The current therapeutic regimen of AD incorporates different pharmacological approaches including topical anti-inflammatory drugs (topical corticosteroids, topical calcineurin inhibitors, phosphodiesterase-4 inhibitors), immunosuppressants (ciclosporin A, azathioprine, methotrexate, mycophenolate mofetil), targeted monoclonal antibodies and oral Janus kinase (JAK) inhibitors [5, 6]. Clinical management of AD is generally guided by disease severity (based on scoring); mild AD can usually be controlled with topical treatments, whereas systemic immunomodulatory therapies (with topicals as adjuvants) are recommended for moderate-to-severe forms of AD. For AD patients whose disease course is not satisfactory or whose quality of life is substantially lowered despite appropriate topical therapy, ultraviolet phototherapy might be considered alone or in combination with systemic therapy [79]. While substantial progress has been made to meet the therapeutic needs of AD patients, considerable limitations remain associated to these conventional approaches. For instance, the action of existing systemic immunomodulators is hampered by poor tolerability, the need for ongoing bloodwork monitoring, and transient relief [10]. The limitations of biologics as a class include difficulty in personalizing the dosage, repeated injections, conjunctivitis following interleukin IL-13 or IL-4/IL-13 blockade, possible anti-drug antibody formation, and decreased potency over time [1, 11]. As for phototherapy, major concerns have arisen including long-term cumulative adverse effects (photodamage), skin carcinogenesis, and melanoma induction, particularly for ultraviolet A1 radiations [12].

    Over the last decade, novel immunotherapeutic strategies have been developed based on upraised understanding of AD mechanisms [1318]. Furthermore, a broad range of immunotherapeutic options is currently under investigation for the management of other skin or immune-mediated diseases, with increasing evidence of their applicability in AD [19, 20]. This review provides insights upon current immunotherapy options, including the latest developments on biologics and targeted therapies by small molecules (JAK inhibitors, phosphodiesterase-4 inhibitors, etc.), with emphasis towards emerging therapies either at their exploratory phase (antibody-drug conjugates, antibody-cytokine fusion proteins), or in clinical development (allergen immunotherapy and cell-based therapy) (Figure 1). The literature search was conducted during the month of April 2024 using PubMed, PubMed Central, Embase, MEDLINE, Web of Science, and ClinicalTrials.gov. The search terms were specifically related to the fields of “immunotherapy of atopic dermatitis”, “allergen immunotherapy”, “cell therapy” and included studies published in English from 2000 to 2024.

    Immunotherapeutics developed for atopic dermatitis (AD) treatment. AD is characterized by chronic inflammation, immune dysfunction, and severe skin impairment. Several immunotherapeutics developed towards an inflammation/allergic reaction resolution include allergen immunotherapy, monoclonal antibodies, antibody-drug conjugates, immunocytokines, small molecule inhibitors, and cell-based therapies. These immunotherapy approaches have shown positive preclinical and clinical efficacy in AD. Created with Biorender.com

    Immunotherapeutics in preclinical development

    Antibody-drug conjugates

    Antibody-drug conjugates (ADCs) represent a special class of immunotherapeutics composed of a cell-specific monoclonal antibody linked to cytotoxic small molecules (toxins or drugs) via a chemical (cleavable or non-cleavable) linker [21]. They have been extensively applied in cancer and inflammatory disease therapy [21, 22]. To date, 14 ADCs have been approved by the United States Food and Drug Administration (US FDA) and approximately 300 ADCs are undergoing preclinical/clinical evaluations for various cancers [21, 23]. Aside from cancers, there is a growing interest in the use of ADCs for the treatment of non-oncological conditions, especially chronic inflammatory and allergic diseases [22] like AD.

    ADCs with chemical moieties

    In a preclinical study, anti-inflammatory phosphodiesterase 4 (PDE4) inhibitor ADC was generated to target CD11a antigen on primary human monocytes and mouse peritoneal cells. The results showed both human and mouse αCD11a-PDE4 ADCs significantly inhibit tumor necrosis factor-alpha (TNF-α) secretion in dysfunctional immune cells in vitro and in vivo [24]. In another preclinical in vitro study targeting membrane immunoglobulin E B-cell receptor (IgE-BCR) fragment, an ADC containing a 15c112 antibody conjugated to monomethyl auristatin E (2:1) demonstrated highly selective binding and cytotoxicity to BCR-positive immune cells, which provides evidence for therapeutic potential in allergic conditions like AD, via IgE secretory cells depletion [25]. Furthermore, anti-E Selectin Dex (Dexa-AbhEsel) ADC, anti-TNF-α glucocorticoid ADC, and anti-CD163 Dex (Cymac-001) ADC, were each used to deliver dexamethasone as an anti-inflammatory payload in vitro and in vivo [23, 26, 27], with all studies demonstrating positive preclinical outcomes in depletion of dysfunctional immune cells and proinflammatory cytokines.

    ADCs with biological moieties

    Aside from chemical moieties, cytotoxic molecules like toxins and cytolytic proteins can be chemically or genetically fused to cell-specific antibodies or antibody fragments for the development of recombinant immunotoxins (rITs) [28], and targeted human cytolytic fusion proteins (hCFPs) [29]. So far, these cytotoxic agents have been explored for cancer immunotherapy and treatment of cutaneous inflammation and lesions, including AD [2931].

    A study by Thepen et al. [32] reported the therapeutic potential of a rIT for the treatment of cutaneous inflammation in transgenic mice expressing human CD64 receptors. The immunotoxin consisted of an anti-CD64 single-chain variable fragment [H22(scFv)] chemically conjugated to ricin A and was used to treat sodium lauryl sulfate (SLS)-induced chronic skin inflammation in human CD64+ transgenic (hCD64tg) mice. The results from intradermal injection at the site of chronic inflammation showed selective elimination of activated M1 pro-inflammatory macrophages, downregulation of pro-inflammatory cytokines like IL-1 and IL-6, and subsequent resolution of chronic cutaneous inflammation after 24 hours. Also, clinical symptoms of inflammation, such as local skin temperature and capillary vasodilation, drastically decreased [32]. Another study from the same group later reported that a rIT containing H22(scFv), genetically fused to truncated Pseudomonas aeruginosa exotoxin A (ETA’) [H22(scFv)-ETA’], demonstrated selective elimination of dysregulated CD64+ pro-inflammatory macrophages and resolution of cutaneous inflammation in hCD64tg mice and patient biopsies [33]. In a different study that took advantage of the ability of cross-linked CD64 to rapidly internalize IgG, bivalent H22(scFv)2-ETA’ was constructed and evaluated for increased uptake and cytotoxicity in CD64+ monocytic cells in cutaneous inflammation. The results demonstrated that H22(scFv)2-ETA’ exerts more cytotoxicity against IFN-γ stimulated CD64+ monocytes compared to monovalent H22(scFv)-ETA’, hence providing evidence that increased valency could remarkably improve the efficacy of anti-CD64 rIT therapy in cutaneous inflammation [31]. Jiemy et al. [34] also developed a rIT containing an anti-CD64 (scFv) antibody fused to the cytolysin HALT-1 (sourced from Hydra magnipapillata) at the C- and T- terminus generating both HALT-1-scFv and scFv-HALT-1 rITs. The study showed that both rITs demonstrate selective binding and significant cytotoxic killing/potency on CD64+ in vitro and M1-like macrophages irrespective of HALT-1 positioning, which provides preliminary evidence for HALT-1 potential as a toxin moiety in therapeutic cell targeting for CD64+ chronic inflammatory disease models [34].

    Besides rITs, humanized enzymes have been used to develop hCFPs that overcome the limitations of immunogenicity associated with toxins. A full human microtubule-associated protein (MAP tau) fused to an H22(scFv) antibody [H22(scFv)-MAP-tau] efficiently killed proliferating HL-60 pro-monocytic cells in vitro and specifically eliminated polarized M1 macrophages in hCD64tg mice in a cutaneous model of chronic inflammation [35]. This result further provides evidence that hCFPs specifically targeted CD64+ M1 proinflammatory macrophages, which are major players in chronic inflammatory diseases, including AD.

    Taken together, these preliminary results clearly demonstrate the anti-inflammatory potency of these ADCs, making some of them serious candidates for first-in-human clinical trials.

    Cell-based therapies

    Cell-based therapy explores the use of living cells as therapeutics to combat hard-to-treat diseases. It extensively involves the transfer of autologous and/or allogeneic cellular materials into a patient for medical purposes [36]. In parallel with pharmacological agents, cell-based therapies are now developed for AD treatment. A novel and effective cell-based therapy for AD treatment is mesenchymal stem cells (MSCs) therapy [3]. MSCs are multipotent stem cells able to differentiate into different cell lineages. MSCs are isolated from different sources and include human umbilical cord blood MSCs (hUCB-MSCs), human adipocytes MSCs (hAD-MSCs), and human bone marrow MSCs (hBM-MSCs) [37]. They express cell surface markers (CD73, CD90, CD105) but lack CD45 and CD34 available in hematopoietic stem cells (HSCs) [37].

    Allogeneic MSCs function as an immunomodulatory therapy (immunosuppressive and immunotolerant), hence their use for the treatment of immunopathological disorders like AD [3, 38]. In AD treatment, MSC-based therapy inhibited T-cell and B-cell activation, and therefore upregulated the production of anti-inflammatory cytokines (IL-10 and TGF-β) and downregulation of proinflammatory cytokines (IL-4 and IFN-γ), as well as allergic IgE levels [39]. Some MSC-based therapies investigated in preclinical AD models are summarized below (Table 1).

    MSC-based therapies in mouse models of AD

    MSC sourceAD modelMouse strainRouteMechanisms of actionReferences
    hBM-MSCsOvalbumin-induced ADBALB/c or C3H/HeNIVSuppression of T cells function via NO and suppression of B-cell function via class switch recombination[40]
    hUCB-MSCsDermatophagoides farinae-induced ADNc/NgaSCInhibition of mast cells degranulation through PGE2 and TGF-β1[41]
    hAD-MSCsDermatophagoides farinae-induced ADNc/NgaIVB-cell suppression via cyclooxygenase 2 regulation[42]
    hAD-MSCsDinitrochlorobenzene-induced ADBALB/cIVRegulation of MIP-2 and miR-122a-SOCS1 expression, as well as Th1/Th2 responses[43]
    hSOD3-MSCsOvalbumin-induced ADBALB/cSCSuppression of response elicited by keratinocytes, mast cells, neutrophils, dendritic cells, and T cells through multiple mechanisms[44]
    hUCB-MSCsDermatophagoides farinae-induced ADNc/NgaSCPre-conditioning of MSC with mast cells granules optimizes suppression of mast and B cells[45]
    hUCB-MSCsDermatophagoides farinae-induced ADBALB/cSCControl of both eosinophil-associated Th2 immunity and neutrophil-related Th17[46]
    Primed T-MSCsDNFB-induced ADC57BL/6JSCRegulation of B cell-mediated inflammatory responses, which are dependent on CD40 expression on primed T-MSCs mediated through the non-canonical NF-κB pathway[47]
    HA-SH/hAD-MSCs (3D-Hydrogel)1-chloro-DNCB-induced ADBALB/cSCImmunomodulatory action on immune cells via downregulation of major inflammatory cytokines (IL-13, CCL11, and CCL24), reduced epidermal thickness, and mast cell penetration[48]
    hAD-MFSCEDermatophagoides farinae-induced ADNC/NgaTopicalDecrease of IgE and inflammatory cytokines levels. Inhibition of epidermal thickness, mast cell infiltration, and expression of expression of IL-4, IL-10, IFN-γ, TNF-α, activation-regulated chemokines[49]
    hAD-MSCsOvalbumin-induced ADBALB/cSCDecrease of serum IgE levels, and mast cells infiltration skin lesions. Suppression of Th17 cells proliferation and proinflammatory cytokines (IL-17A and RORγT) expression. Upregulation of PD-L1, TGF-β and PGE2 levels[50]
    hUCB-MSCs1-chloro-DNCB-induced ADBALB/cSC + IVAnti-inflammatory and Immunomodulatory function resulting in amelioration of skin lesions, through inhibition of JAK-STAT pathway and different cytokines (IL-4, IL-13, IL-17, and IgE) receptors[51]
    Display full size

    CCL11: eotaxin 1; CCL24: eotaxin 2; DNCB: 2,4-dinitrobenzene; DNFB: 2,4-dinitrofluorobenzene; IL: interleukin; IV: intravenous; MIP-2: macrophage inflammatory protein 2; NF-κB: nuclear factor kappa B; PGE2: prostaglandin E2; SC: subcutaneous; TGF-β1: transforming growth factor beta 1

    Immunocytokines

    Immunocytokines, also referred to as antibody-cytokine fusion proteins, are molecules that consist of a cytokine moiety fused to a monoclonal antibody or an antibody fragment [52], with antibody and cytokine serving as vehicle and payload, respectively. The development of antibody-cytokine fusion proteins emerged as a novel immunotherapy strategy to circumvent the detrimental effects associated with the systemic administration of cytokines as a monotherapy [53]. In fact, while cytokines may display a potent therapeutic activity in preclinical models of cancer and other conditions, their clinical use is often limited by severe systemic toxicities that prevent a dose escalation to therapeutically active regimes. The antibody-mediated delivery of cytokines to the site of disease might be a step forward in increasing therapeutic activity, while reducing side effects [54]. Immunocytokines have been extensively investigated for cancer treatment and a recent review describes their engineering formats and clinical benefits [55]. Immunocytokines also attract huge interest in the field of chronic inflammatory conditions, including inflammatory skin diseases.

    In inflamed tissue, the lymphatic vasculature undergoes extensive remodeling and expansion, including lymphangiogenesis (formation of new lymphatic vessels) and enlargement of preexisting vessels. Vascular endothelial growth factor C (VEGF-C) was found to potently reduce inflammation in a mouse model of oxazolone-induced psoriasis-like skin inflammation [56]. Later on, Schwager et al. [57] generated and intravenously administered a human VEGF-C fused to the F8 diabody (F8-VEGF-C) to two other mouse models of psoriasis-like skin inflammation (induced by oxazolone in transgenic VEGF-A overexpressing animals or repeated application of imiquimod). F8 antibody is highly specific for the angiogenesis-marking extradomain A (EDA) of fibronectin, a protein selectively expressed in inflamed tissues. F8-VEGF-C treatment significantly reduced leukocyte infiltration in inflamed tissue, including CD4+ and γδ T cells. In addition, the treatment reduced ear skin edema and significantly improved lymphatic drainage function [57]. These results were further confirmed by the same group, who investigated the long-term effects of F8-VEGF-C using the oxazolone-mediated contact hypersensitivity (CHS) model in mice overexpressing VEGF-A in their epidermis. Authors reported that F8-VEGF-C treatment resulted in a long-term anti-inflammatory activity that persisted for more than 70 days, by reducing the severity of edema and immune cell (CD4+ T cells, macrophages, and dendritic cells) infiltration upon hapten re-challenge [58]. As psoriasis and AD are two inflammatory skin diseases sharing some common features, this promising approach, though at its early stage, could be exploited in the near future for AD treatment.

    Immunotherapeutics in the clinical pipeline

    Monoclonal antibodies

    Targeted monoclonal antibodies were first introduced in dermatology for the treatment of psoriasis [59] before their extension to AD.

    Th-2 mediated axis

    AD involves predominately Th2-mediated inflammation and a key feature of the disease is the upregulation of IL-13 and IL-4 in lesional and non-lesional skin [60]. The two key Th2 cytokines, IL-4/IL-13 are suggested to drive important pathologic processes in AD including; keratinocyte differentiation, tight junction formation, recruitment of inflammatory cells to skin by upregulating Th2-associated chemokines (CCL17 and CCL26), antimicrobial peptides activity, and inhibition of important physiological processes like lipid synthesis [61]. Dupilumab is a monoclonal antibody targeting both IL-4 and IL-13 receptors, through IL-4Rα, and has been the first FDA-approved (March 2017) monoclonal antibody for use in AD [62]. Dupilumab’s success paved the way in exploring the individual contribution of IL-4 and IL-13 in AD pathophysiology, leading to the development of two monoclonal antibodies targeting IL-13, namely tralokinumab and lebrikizumab. Tralokinumab acts by binding to IL-4Rα and IL-13Rα1 (the two subunits of the IL-13 receptor), as well as to the IL-13Rα2 decoy receptor [63], whereas, lebrikizumab specifically binds soluble IL-13 at an epitope that strongly overlaps with the binding site of IL-4Rα [64]. Thymic stromal lymphopoietin (TSLP) is a cytokine produced by keratinocytes that has recently been postulated to play a key pathogenic role in AD. In proinflammatory states, damaged keratinocytes produce TSLP that acts on myeloid dendritic cells and Langerhans cells, leading to a Th2-skewed inflammatory response with the release of IL-4, IL-5, and IL-13 [65]. In a recent phase 2b clinical trial (NCT03809663), tezepelumab (an anti-TSLP monoclonal antibody) was investigated for its use as monotherapy in moderate-to-severe AD patients but could not achieve the primary endpoints for this patient population.

    Th22-mediated axis

    AD is not only Th2-centered among various disease subtypes, it also overexpresses Th22-related cytokines and chemokines, such as IL-22 which was found to positively correlate with disease severity [66]. IL-22 was proven to increase epidermal barrier dysfunction, induce epidermal hyperplasia, downregulate key genes (such as filaggrin and loricrin) involved in epidermal differentiation, and drive the epidermal acanthosis characterizing chronic AD lesions [67]. Fezakinumab (a fully human monoclonal antibody directed against IL-22) was investigated in adults with uncontrolled AD for 20 weeks in a phase 2a clinical trial and SCORAD 50 (proportion of patients achieving scoring atopic dermatitis improvement of ≥ 50% from the baseline) endpoint was achieved, at week 12, in severe AD subpopulation [68].

    Th17-related and other axis

    Th17-related cytokines, specifically IL-17 and IL-23, are the key drivers of the inflammatory process in psoriasis, and their potential use as therapeutic targets in the context of AD is supported by several studies [61]. IL-17 synergizes with IL-22 to promote tissue inflammation and barrier defects, while IL-23 activates both Th17 and Th22 pathways [69]. Ustekinumab is a fully human monoclonal antibody directed against the p40 subunit of IL-12 and IL-23, preventing them from binding to T cell-surface receptors and causing differentiation and clonal expansion of naïve T cells into specific Th subtypes [70]. A phase 2 double-blinded and placebo-controlled study revealed that ustekinumab administration only resulted in a numerical improvement of AD clinical scores, without statistical significance [71]. A summary of clinical trials investigating monoclonal antibody therapy in AD, irrespective of disease subtypes or immune axis explored, has been tabulated (see Table 2).

    Monoclonal antibodies in advanced clinical trials for AD

    TargetDrugRouteStatusGlobal endpoints achievedReported adverse effectsReferences
    IL-1αBermekimabSCPhase IIEASI-75, SCORAD-50Wheezing, injection-site reaction, nauseaNCT03496974
    IL-4αStapokibartSCPhase IIIOngoingOngoingNCT06277765
    NCT05265923
    RademikibartSCPhase IIIGA 0/1Headache, vomiting, injection-site reaction, eye pruritus[72]
    611SCPhase IIIOngoingOngoingNCT06173284
    AK120SCPhase I/IIEASI-75Upper respiratory tract infection, headache, abdominal pain, mouth ulceration, injection-site reaction[73]
    NCT05048056
    MG-K10SCPhase IIIOngoingOngoingNCT06026891
    DupilumabSCApprovedEASI-75; EASI-90Conjunctivitis, injection-site reaction, nasopharyngitis, herpes viral infections, viral gastroenteritis, dental caries[62, 7479]
    IL-13TralokinumabSCApprovedIGA 0/1; EASI-50; EASI-75; EASI-90Viral upper respiratory tract infection, injection-site reaction, conjunctivitis[8082]
    LebrikizumabSCApprovedIGA 0/1; EASI-75; EASI-90Headache, oral herpes, conjunctivitis, nasopharyngitis, dry eye, folliculitis, upper respiratory tract infection[83, 84]
    EblasakimabSCPhase IIbEASI-75Not availableNCT05158023
    IL-5RαBenralizumabSCPhase IIEASI-90Bronchitis, conjunctivitis, Covid-19, nasopharyngitis, upper respiratory tract infectionNCT04605094
    IL-12/IL-23p40UstekinumabSCPhase IINo significant efficacyUpper respiratory tract infection, musculoskeletal pain[71]
    IL-17ASecukinumabSCPhase IINo efficacyOrbital cellulitis, upper respiratory infection, streptococcal pharyngitis[85]
    IL-22FezakinumabSCPhase IIaSCORAD-50 in severe AD subpopulationViral upper respiratory tract infections, facial cellulitis[68]
    LEO138559SCPhase IIbOngoingOngoingNCT05923099
    IL-23RizankisumabSCPhase II No efficacyCellulitis, nasopharyngitis, pruritus[86]
    IL-31NemolizumabSCApproved (Japan)EASI-90; IGA 0/1Upper respiratory tract infection, nasopharyngitis[8791]
    IL-33AstegolimabSCPhase IINo efficacyNone recorded[92]
    EtokimabIVPhase IIaEASI-75Headache, upper respiratory tract infection, conjunctivitis[93]
    TozorakimabSCPhase IIaNo efficacyOral herpes, cellulitis, conjunctivitis, nasopharyngitis, urinary tract infectionNCT04212169
    REGN3500SCPhase IIbNo efficacyPeripheral oedema, nasopharyngitisNCT03738423
    IL-36RSpesolimabIVPhase IIaNo significant efficacyUpper respiratory tract infection, nasopharyngitis, depression[94]
    OX40GBR830SCPhase IIbNo significant efficacyUpper respiratory tract, myalgia, headache, postprocedural infection, fatigue[95]
    AmlitelimabIVPhase IIa/bEASI-75; IGA 0/1Nasopharyngitis, Covid-19, headache, hyperhidrosis, upper respiratory tract infection, pyrexia, increased aspartate aminotransferase and iron deficiency anemia[96, 97]
    RocatinlimabSCPhase IIbEASI-90Atopic cataract, small intestine ulcer, pyrexia, vertigo, aphthous ulcer, constipation, diarrhea, vomiting, nausea, chills, nasopharyngitis, arthralgia, oropharyngeal painNCT03703102
    IgEOmalizumabSCPhase IINo significant efficacyViral infection, stomach pain, sore throat, toothache, urticaria, headache[98100]
    mIgEFB825IVPhase IINot availableNot availableNCT04413942
    TSLPTezepelumabSCPhase IIbNo significant efficacyHeadache, diarrhea, injection-site erythema[101]
    NCT03809663
    SCFOpSCFSCPhase IIaOngoingOngoingNCT06101823
    Display full size

    EASI: eczema area and severity index; EASI 50/75/90: ≥ 50/75/90% improvement in EASI from baseline; IGA: investigator global assessment; IGA 0/1: “clear or almost clear” with ≥ 2-point improvement in IGA from baseline; IgE: immunoglobulin E; ILα: alpha chain of interleukin; ILRα: alpha chain of interleukin receptor; IV: intravenous; mIgE: membrane-bound immunoglobulin E; NCT: national clinical trial identifier consultable on www.clinicaltrials.gov (website consulted last time as of April 30, 2024), only clinical trials in advanced phases (Phase II onwards) are listed; SCF: stem cell factor; SC: subcutaneous; SCORAD: scoring atopic dermatitis; SCORAD 50: ≥ 50% improvement in SCORAD from baseline; TSLP: thymic stromal lymphopoietin

    Small molecules

    Besides antibody-based therapies that usually target cytokines or their receptors, small molecules exert their effects by interfering with specific intracellular signaling pathways. Various small molecules are being evaluated as targeted therapeutics for AD and given their potential in blocking several immune pathways, these drugs significantly contribute to the expanding toolbox for AD management. Moreover, their action can be further potentiated when fused to antibodies targeting specific receptors.

    Janus kinase inhibitors

    JAK inhibitors are intracellular enzymes mediating the signaling cascade from a cytokine receptor into the cell [102]. The JAKs are a family of tyrosine kinases, including JAK1, JAK2, JAK3, and TYK2. The intracellular domain of various cytokine receptors is phosphorylated by JAKs, which results in binding and activation of the signal transducer and activator of transcription (STAT). The JAK-STAT signaling then mediates a range of intracellular immune dysregulations, including robust Th2 responses [61, 103]. Upadacitinib and abrocitinib are two recent FDA-approved oral JAK inhibitors with high potency and good safety profiles.

    Phosphodiesterase 4 inhibitors

    Cyclic adenosine monophosphate (cAMP) was reported to play a role in the regulation of inflammatory and immune responses, with increased levels of cAMP being associated with monocytes and T cell suppression. PDEs are enzymes that break down cAMP or cyclic guanosine monophosphate (cGMP), with certain isoenzymes expressed only in selected tissues [104, 105]. PDE4 is found in immune and inflammatory cells (basophils, mast cells, eosinophils, B and T cells, monocytes, macrophages, neutrophils and endothelial cells) and PDE4 isoforms (PDE4A, PDE4B, PDE4C and PDE4D) have been observed to be increased in dermal fibroblasts of AD patients skin samples compared to dermal fibroblasts from healthy skin [106]. PDE4 inhibitors impair cytokine production by elevating cAMP levels, thereby directly or indirectly blocking the NFκB pathway and the subsequent cytokine and chemokine synthesis. Moreover, this mechanism of immunologic suppression has been suggested to also occur in keratinocytes, indicating a role in skin barrier protection [107]. To date, crisaborole and difamilast are the only approved PDE4 inhibitors for mild-to-moderate AD in adults and children, and the FDA recently accepted the supplemental new drug application (sNDA) for roflumilast cream 0.15% to treat AD in patients aged 6 and up.

    Other small molecules

    Molecules targeting histamine 4 receptor (H4R) could be exploited for AD treatment, as histamine, in addition to inducing pruritus, was found to inhibit the terminal differentiation of keratinocytes and impair the skin barrier in AD [108]. Sphingosine1-phosphate (S1P) is a bioactive lipid mediator that impacts immunological processes by regulating migration of lymphocytes out of lymphatic tissues and dendritic cells migration to lymph nodes [109]. Therefore, S1P receptors (S1PRs) are a promising therapeutic target, with several clinical trials evaluating S1PRs modulators for the treatment of multiple inflammatory disorders, including AD. Etrasimod is an oral selective S1PR1/4/5 modulator in development for multiple immune-mediated inflammatory disorders. Its efficacy and safety have been confirmed in patients with moderate-to-severe AD in a Phase 2b clinical trial [110] and Pfizer® has recently announced ongoing Phase III clinical development of etrasimod for AD. Other immune pathways still in their exploratory phase for small molecules usage include neurokinin 1 receptor (NK1R), liver X receptor (LXR), purinergic receptor P2X (P2X3) or kappa opioid receptor (KOR). A comprehensive overview of small molecules in clinical trials is captured in Table 3.

    Small molecules in advanced clinical trials for AD

    TargetDrugRouteStatusGlobal endpoints achievedReported adverse effectsReferences
    JAK1UpadacitinibOralApprovedEASI-75; EASI-90; EASI-100; IGA 0/1Nasopharyngitis, acne, headache, oral herpes, diarrhea, eczema, herpes zoster[111114]
    AbrocitinibOralApprovedIGA 0/1; EASI-50; EASI-75Transient decreased platelet count, acne, dizziness, nasopharyngitis, acne, headache, oral herpes, vomiting, diarrhoea, eczema, herpes zoster, nausea, folliculitis[115120]
    LNK01001OralPhase IIIOngoingOngoingNCT06277245
    SHR0302OralPhase II; Phase II/IIIIGA 0/1; EASI-75Blood pressure increased, upper respiratory tract infection, folliculitis, urinary tract infection, hyperlipidemia, hyperuricemia, headache[121]
    NCT04717310
    JAK1/2BaricitinibOralApprovedIGA 0/1; EASI-75Headache, increased blood CPK, nasopharyngitis, herpes simplex, influenza, diarrhea, nausea, folliculitis, back pain, acne[122128]
    RuxolitinibTopicalApprovedIGA 0/1; EASI-75; EASI-90Nasopharyngitis, headache[129131]
    JAK1/3TofacitinibTopicalPhase IIaEASI-75Viral upper respiratory tract infection, gastroenteritis, bronchitis, nausea[132]
    ATI-1777TopicalPhase IIa/bmEASI-75Blood CPK increased, headache[133]
    NCT05432596
    ATI-502TopicalPhase IIbNo significant efficacyNone reported[134]
    NCT03585296
    JAK1/TYK2BrepocitinibTopicalPhase IIbEASI-90Nasopharyngitis, erythema[135]
    JAK1/2/3/TYK2DelgocitinibTopicalApproved (Japan)mEASI-50; mEASI-75; mEASI-90Acne, folliculitis, gastroenteritis, herpes simplex, paronychia, nasopharyngitis[136140]
    Pan-JAKJaktinibOralPhase IIIOngoingOngoingNCT05526222
    NCT05676242
    PDE4CrisaboroleTopicalApprovedIGA 0/1Application-site pain, upper respiratory tract infection, nasopharyngitis, headache, oropharyngeal pain, cough[141143]
    PDE4LotamilastTopicalPhase IINot availableNot availableNCT03394677
    PDE4DRM02TopicalPhase IINot availableNot availableNCT01993420
    PDE4DifamilastTopicalApproved (Japan)IGA 0/1; EASI-75; EASI-90Nasopharyngitis, folliculitis, gastroenteritis[144146]
    NCT05571943
    NCT05372653
    PDE4RoflumilastTopicalPhase IIIIGA 0/1; EASI-75Headache, nausea, application-site pain, nasopharyngitis, Covid-19, upper respiratory tract infection, diarrhea, vomiting[147]
    NCT04773587
    NCT04773600
    P2X3BLU-5937OralPhase IINot availableNot availableNCT04693195
    NK1RSerlopitantOralPhase IINo significant efficacyNasopharyngitis, urinary tract infectionNCT02975206
    NK1RTradipitantOralPhase IIIWI-NRS ≥ 4; IGA 0/1; SCORAD 50Not available[148]
    NCT04140695
    KORDifelikefalinOralPhase III-NRS ≥ 4Abdominal pain/discomfort, nausea, dry mouth, headache, dizziness, hypertension, hyponatremia, nephrolithiasis, costochondritis[149]
    S1PR1VibozilimobOralPhase IIOngoingOngoingNCT04684485
    S1PR1UdifitimodOralPhase IINo significant efficacySkin mass, depression, upper respiratory tract infection, pyrexiaNCT05014438
    S1PR1/S1PR4/S1PR5EtrasimodOralPhase IIbEASI-50Nausea, constipation, back pain, dizziness[110]
    H4RAdriforantOralPhase IIEASI-75Headache, somnolence, nasopharyngitis, eczema, urinary tract infection[150]
    GPCR19HY209TopicalPhase IINot availableNot availableNCT04530643
    LXRVTP-38543TopicalPhase I/IINo significant efficacyUpper respiratory tract infection, fatigue, cellulitis, dizziness, pruritus[151]
    LXRALX-101TopicalPhase IIbNot availableNot availableNCT03859986
    Display full size

    CPK: creatine phosphokinase; EASI: eczema area and severity index; EASI 50/75/90: ≥ 50/75/90% improvement in EASI from baseline; GPCR19: G protein-coupled receptor 19; H4R: type 4 histamine receptor; IGA: investigator global assessment; IGA 0/1: “clear or almost clear” with ≥ 2-point improvement in IGA from baseline; I-NRS: itch numeric rating scale; JAK: janus kinase; KOR: kappa opioid receptor; LXR: liver X receptor; mEASI: modified eczema area and severity index; mEASI 50/75/90: ≥ 50/75/90% improvement in mEASI from baseline; NCT: national clinical trial identifier consultable on www.clinicaltrials.gov (website consulted last time as of April 30, 2024), only clinical trials in advanced phases (Phase II onwards) are listed; NK1R: neurokinin 1 receptor; PDE4: phosphodiesterase 4; Pan-JAK: multiple janus kinases inhibitor; P2X3: purinergic receptor P2X; SCORAD: scoring atopic dermatitis; SCORAD 50: ≥ 50% improvement in SCORAD from baseline; S1PR1/4/5: sphingosine 1-phosphate receptor 1/4/5; TYK: tyrosine kinase; WI-NRS: worst itch numeric rating scale; WI-NRS/I-NRS ≥ 4: ≥ 4-point change in WI-NRS/I-NRS from baseline

    Cell-based therapies

    Over the last decade, MSCs therapy has grown to a position of serious alternative treatment for AD, following successful proof-of-concept preclinical studies.

    In 2011, Ra et al. [152] reported case studies where intravenous injections of autologous human adipose-derived MSCs (hAdMSCs) where used as a compassionate treatment for 4 AD patients with exhausted therapeutic options. Patients were administered 6–10 × 108 hAdMSCs in total and followed up for 2 to 5 and half months after treatment. The treatment resulted in significant AD score improvement in treated patients with no adverse events recorded during the follow-up period, providing thus, the first evidence for clinical benefit of hAdMSCs in AD. The first-in-class clinical trial was published in 2016 [153], from a study that enrolled 34 moderate-to-severe AD adult patients with symptoms not adequately controlled by topical corticosteroids or systemic immunosuppressants. Patients received subcutaneous injections of low dose (2.5 × 107) or high dose (5 × 107) of hUCB-MSCs fortnightly for 4 weeks in Phase 1 trial, and 12 weeks in Phase 2. Half of the patients receiving the high dose of hUCB-MSCs experienced a 50% reduction of EASI score, while the investigator global assessment (IGA) and SCORAD index decreased by 33% and 50%, respectively. The treatment also reduced serum IgE and blood eosinophils levels, while appearing to be globally safe. In another study, conditioned media from hUCB-MSCs was investigated for its beneficial effects in 34 patients suffering from mild AD [154]. Topical application of conditioned media (contained in a cream formulation) twice daily for 4 weeks on patients’ lesions strengthened the skin barrier and reduced the trans-epidermal water loss. This outcome was attributed to the anti-inflammatory potency of the conditioned media, as it was found to inhibit the expression of TNF-α and IL-6 cytokines in HaCaT cells, as well as IL-4 and IL-13 levels in Th2 cells in vitro. Shin et al. [155] reported the use of allogenic bone marrow-derived clonal MSCs (bmMSCs), in a single-center investigator-initiated clinical trial, in five adult patients with moderate to severe AD. Patients received intravenous injections of bmMSCs (1 × 106 cells/kg) three times every 2 weeks during a 4 weeks period and were followed up the next 12 months. At week 16, 4 out of the 5 patients (80%) achieved EASI-50 after one or two treatment cycles and two patients maintained good clinical response for over 84 weeks, with significant improvement of IGA, SCORAD, and BSA. Minor adverse reactions such as cellulitis, otitis, and acute upper respiratory tract infection were recorded, but were only transient with full recovery of patients with, and often, without symptomatic treatment. Authors surmised that efficacy of bmMSCs would be related to increased levels of IL-17, associated to lower blood levels of CCL-17, IL-13, and IL-22. Other human clinical trials still in progress or which results are not yet published involve umbilical cord-derived MSCs (NCT01927705; NCT05004324; NCT03269773), adipose tissue-derived MSCs (NCT04137562; NCT02888704), bone marrow-derived MSCs (NCT04179760), or induced pluripotent stem cell-derived exosomes (NCT05969717).

    Not only humans are affected by AD but dogs as well, with a prevalence of 10–15% worldwide [156]. The therapeutic use of MSCs has also been investigated in canine AD and to date, a handful of studies involving adipose tissue-derived MSCs treatment in canine AD (cAD) have been documented [38, 157161]. Notwithstanding the positive outcomes conveyed, large scale placebo-controlled, long-term studies are necessary to further validate these results.

    Allergen immunotherapy

    Also known as specific desensitization, hyposensitization or allergen-specific immunotherapy, allergen immunotherapy (AIT) involves the administration of gradually increasing quantities of relevant allergens in order to induce immunologic tolerance. This therapeutic option has yielded evidenced benefits in allergen-induced conditions like allergic asthma, allergic rhinitis and Hymenoptera venom allergy [162, 163]. Since AD is characterized by varied clinical presentations and heterogeneous phenotypes, AIT in AD is a controversial issue. Allergen-specific immunotherapy for AD has several indications, including exacerbation of symptoms following aeroallergen exposure, aeroallergen sensitization along with positive skin prick test (SPT) and/or sIgE production, moderate to severe SCORAD [25] and atopy patch testing (APT) positivity against the aeroallergen [164, 165]. Allergen extracts used for AIT primarily consist of allergenic proteins derived from grass/tree pollen, pet dander, dust mites, insect venom, and mold. House dust mite (HDM) extract, whose main trigger agents are Dermatophagoides pteronyssinus and Dermatophagoides farinae, is by far the most used allergen in AIT of AD [166]. Roughly, the proposed mechanism of long-term tolerance induction in AIT, even upon treatment discontinuation, involves desensitization of effector cells (basophils, mast cells, innate lymphoid cells, dendritic cells) through activation of allergen-specific regulatory T and B cells, downregulation of effector type 2 responses, decrease in the production of IgE and increase in production of allergen-specific blocking antibodies such as IgG2 and IgG4 [167]. AIT administration includes subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT), both being well established as safe and effective treatments to address allergies to aeroallergens for allergic rhinitis and allergic asthma [2]. Since SCIT requires an injection-based administration, SLIT may offer a viable alternative to reluctant patients. Furthermore, SLIT reduces the utilization of healthcare resources and staff time, does not necessitate specialized expertise or facilities, and can be administered at home, making it a suitable option even for young children [166, 168]. However, both SCIT and SLIT can induce local and systemic reactions, requiring physicians and patients to be aware of the potential occurrence of such adverse events. Fortunately, manifestation of systemic life-threatening reactions, such as severe anaphylactic reactions, remains very uncommon [169]. Lately, the clinical efficacy of AIT in AD patients has been extensively investigated and the main outcomes of some recent studies are summarized in Table 4.

    AIT studies in AD

    Study typeAllergenTreatment durationNumber of patients enrolledComplementary medicationRouteGlobal endpoints achievedAdverse reactions reportedReferences
    Open-label, controlled, randomized trialHDM12 months60Moisturisers, topical and systemic drugsSLSignificant increase of allergen-specific Ig4Oropharyngeal itching, localized lip swelling[170]
    Randomized controlled trialHDM36 months440Topical glucocorticoids, skin moisturizers, oral antihistaminesSLSignificant improvement of SCORADLip swelling, numbness, pruritus[171]
    Randomized controlled trialHDM6 months14Moisturizers, antihistamine and mild-potency anti-inflammatory topical treatmentsSLSignificant improvement of SCORAD and TEWLNot available[172]
    Randomized, double-blind, placebo-controlled trialHDM18 months66Topical corticosteroids, topical immunosuppressors, and systemic immunosuppressantsSLSignificant improvement of SCORADVomiting, abdominal pain, aphthous ulcer, alopecia, dyspnea[173]
    Randomized, placebo-controlled, double-blind trialHDM12 months37Oral antihistamines, topical medicationsSCSignificant improvement of EASIMild local reaction, transient urticaria[174]
    Multi-centre, randomized, double-blind, placebo-controlled studyHDM36 weeks239Mometasone furoate cream, levocetirizine hydrochloride tablets, mupirocin ointment, clarithromycin tabletsSLSignificant improvement of SCORAD and SLASwelling in tough and lips, throat pain, diarrhoea, itching, numb in mouth[175]
    Randomized controlled studyHDM24 months96Oral levocetirizine hydrochloride tablets, topical fluticasoneSLSignificant reduction of SCORADTransient oral itching, gastrointestinal discomfort[176]
    Randomized controlled studyHDM12 months107Oral antihistamines and topical medicationsSLSignificant increase of serum-specific IgG4 levelTransient oral itching, gastrointestinal discomfort[177]
    Randomized, double-blind, placebo-controlled studyHDM18 months56Short term use (3 days) of topical fluticasone propionate and/or oral hydroxyzineSLSignificant reduction of SCORAD in mild-moderate patientsTiredness, oral itching, headache, mouth, lips, and face swelling[178]
    Observational studyHDM; cat/dog dander2–58 months19Topical treatment and/or oral antihistaminesSLSignificant improvement of IGA and EASINot available[179]
    Multi-centre randomized, double-blind, placebo-controlled parallel group trialHDM18 months168Topical and (as necessary) systemic medicationSCSignificant reduction of SCORAD in severe AD patientsFlare-ups of eczematous and urticarial lesions, symptoms of rhinitis, pruritus, transient headache, and asthma[180]
    Display full size

    AD: atopic dermatitis; AIT: allergen immunotherapy; EASI: eczema area and severity index; HDM: house dust mite; IGA: investigator global assessment; SC: subcutaneous; SL: sublingual; SLA: skin lesion area; SCORAD: scoring atopic dermatitis; TEWL: transepithelial water loss

    Strategies for precision healthcare

    Besides immunotherapeutic strategies, harnessing improved formulations and delivery systems of existing molecules could go a long way to improve their clinical efficacy. Nanocarrier-based formulations for enhanced delivery of AD treatments have recently arisen as a promising approach prompted by positive deliveries in vivo. These alternate drug delivery systems include nanoemulsions (oil-in-water, water-in-oil, oil-in-water-in-oil, water-in-oil-in-water), liposomes, transfersomes, ethosomes, lipid nanoparticles (nanostructured lipid carrier, solid lipid nanoparticles), nanocrystals, polymeric nanoparticles (nanocapsules, nanospheres) and polymeric micelles [181, 182]. These nanocarriers have significant advantages over traditional formulations such as improved skin penetration and retention, controlled release, and reduced off-target effects, largely driven by optimized physicochemical features such as shape, size, structural design, and surface charge. Nevertheless, major drawbacks, including large-scale production, the scarcity of global regulatory guidelines, intellectual property rights, cost effectiveness [183], and lack of models that emulate the entire AD complexity, are still to be addressed on the path to clinical translation and commercialization.

    A better stratification strategy of AD patients is also needed in achieving patient-specific treatments (precision medicine), by taking into account phenotypical features (as AD varies among different subpopulations), disease triggers, genetic hallmarks, and specific biomarkers. This would enable medical practitioners to identify AD endotypes more amenable to certain treatments (most probably in the form of combinatory therapies), consequently improving therapeutic efficacy while simultaneously limiting the issues of adverse side effects. This approach could be exemplified by a genuine use of the patient’s molecular profiles to identify appropriate therapeutic approaches for each patient. For instance, fezakinumab (an IL-22 antagonist) revealed significant clinical improvement, compared with placebo, especially in patients with higher IL-22 levels, through downregulation of Th1, Th2, and Th17-related biomarkers [184]. Developing easy-to-use and non-invasive techniques, such as skin tape strip sampling, would therefore facilitate diagnosis and then treatment based on the patient’s specific immune pathway [185]. Building such a sharp stratification strategy would further require the development of new and robust tools able to analyze a tremendous amount of highly complex data. Recent advances in artificial intelligence (AI) have shown promise in the diagnosis, classification, and predictive treatment of AD. Ghosh et al. [186], identified 89 genes expressed in AD to build a “signature” model, using a support vector machine, and achieved an AD predictive accuracy of 98%, compared to normal skin [186]. Furthermore, a recent machine learning tool successfully classified infant AD patients into various clusters, based on several chemokines and cytokines expression profiles [187]. Beyond diagnostic, scoring, and staging purposes, AI-based tools have been very recently applied in AD for drug repurposing, new drug eligibility, and prediction of the therapeutic response to biologics [188].

    Conclusions

    Understanding a complex disorder such as AD in its full phenotypic and mechanistic array remains a daunting goal, however, with the ever-increasing pathophysiology insights, improved patient management and outcomes are expected. Notwithstanding the limited armamentarium of approved targeted biological therapies at present, current trends in alternative diseases, particularly cancer, are certain to pave the way to address this unmet need for more well tolerated and narrow-targeted therapeutics. In addition to monoclonal antibodies which seem to offer safer effective systemic therapies, compared with the off-label use of broad-acting immunosuppressants, small molecules, AIT and cell-based therapy bring additional options addressing different causative factors within AD context. ADCs and immunocytokines, albeit still at their preclinical exploratory phase, also hold the promise of a possible breakthrough in AD therapeutic arsenal.

    The advent of immunotherapy in the context of AD has marked a paradigm shift towards personalized medicine and has led to remarkable successes. However, major limitations remain, and ongoing efforts to further understand the complete picture of the complex pathophysiology of AD are expected to yield more effective immunotherapeutic agents. Integration of these immunotherapeutic strategies into a holistic approach involving optimized drug delivery systems and AI-based technologies has the potential to profoundly change the landscape of AD healthcare, with the promise of more customized treatments and a better quality of life for AD patients.

    Abbreviations

    AD:

    atopic dermatitis

    ADC:

    antibody-drug conjugate

    AI:

    artificial intelligence

    AIT:

    allergen immunotherapy

    bmMSC:

    bone marrow-derived clonal mesenchymal stem cell

    cAMP:

    cyclic adenosine monophosphate

    ETA’:

    exotoxin A

    FDA:

    Food and Drug Administration

    hAdMSC:

    human adipose-derived mesenchymal stem cell

    hCFP:

    human cytolytic fusion protein

    hUCB-MSC:

    human umbilical cord blood mesenchymal stem cell

    IgE:

    immunoglobulin E

    JAK:

    janus kinase

    MSCs:

    mesenchymal stem cells

    PDE4:

    phosphodiesterase 4

    rIT:

    recombinant immunotoxin

    scFv:

    single-chain variable fragment

    SCIT:

    subcutaneous immunotherapy

    SCORAD:

    scoring atopic dermatitis

    SLIT:

    sublingual immunotherapy

    S1PR:

    sphingosine1-phosphate receptors

    TNF-α:

    tumor necrosis factor-alpha

    TSLP:

    thymic stromal lymphopoietin

    VEGF-C:

    vascular endothelial growth factor C

    Declarations

    Author contributions

    GTS: Conceptualization, Writing—original draft, Writing—review & editing. EAF and MM: Writing—original draft. KN: Writing—review & editing. SB: Conceptualization, Writing—review & editing, Supervision, Funding acquisition. All authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    This research was funded by the South African Research Chairs Initiative of the Department of Science and Technology (DST) and the National Research Foundation (NRF) [47904] of South Africa. The NRF and the South African Research Chair in Cancer Biotechnology have provided bursaries to support students contributing to this work. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

    Copyright

    © The Author(s) 2024.

    References

    Butala S, Castelo-Soccio L, Seshadri R, Simpson EL, O’Shea JJ, Bieber T, et al. Biologic Versus Small Molecule Therapy for Treating Moderate to Severe Atopic Dermatitis: Clinical Considerations. J Allergy Clin Immunol Pract. 2023;11:136173. [DOI] [PubMed] [PMC]
    Yepes-Nuñez JJ, Guyatt GH, Gómez-Escobar LG, Pérez-Herrera LC, Chu AWL, Ceccaci R, et al. Allergen immunotherapy for atopic dermatitis: Systematic review and meta-analysis of benefits and harms. J Allergy Clin Immunol. 2023;151:14758. [DOI] [PubMed]
    Najera J, Hao J. Recent advance in mesenchymal stem cells therapy for atopic dermatitis. J Cell Biochem. 2023;124:1817. [DOI] [PubMed]
    GBD 2019 IMID Collaborators. Global, regional, and national incidence of six major immune-mediated inflammatory diseases: findings from the global burden of disease study 2019. EClinicalMedicine. 2023;64:102193. [DOI] [PubMed] [PMC]
    Zhou S, Qi F, Gong Y, Zhang J, Zhu B. Biological Therapies for Atopic Dermatitis: A Systematic Review. Dermatology. 2021;237:54252. [DOI] [PubMed]
    Müller S, Maintz L, Bieber T. Treatment of atopic dermatitis: Recently approved drugs and advanced clinical development programs. Allergy. 2024;79:150115. [DOI] [PubMed]
    Simpson EL, Bruin-Weller M, Flohr C, Ardern-Jones MR, Barbarot S, Deleuran M, et al. When does atopic dermatitis warrant systemic therapy? Recommendations from an expert panel of the International Eczema Council. J Am Acad Dermatol. 2017;77:62333. [DOI] [PubMed]
    Sidbury R, Davis DM, Cohen DE, Cordoro KM, Berger TG, Bergman JN, et al. Guidelines of care for the management of atopic dermatitis: section 3. Management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:32749. [DOI] [PubMed] [PMC]
    Eichenfield LF, Tom WL, Berger TG, Krol A, Paller AS, Schwarzenberger K, et al. Guidelines of care for the management of atopic dermatitis: section 2. Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:11632. [DOI] [PubMed] [PMC]
    Moyle M, Cevikbas F, Harden JL, Guttman-Yassky E. Understanding the immune landscape in atopic dermatitis: The era of biologics and emerging therapeutic approaches. Exp Dermatol. 2019;28:75668. [DOI] [PubMed] [PMC]
    Akinlade B, Guttman-Yassky E, de Bruin-Weller M, Simpson EL, Blauvelt A, Cork MJ, et al. Conjunctivitis in dupilumab clinical trials. Br J Dermatol. 2019;181:45973. [DOI] [PubMed] [PMC]
    Rodenbeck DL, Silverberg JI, Silverberg NB. Phototherapy for atopic dermatitis. Clin Dermatol. 2016;34:60713. [DOI] [PubMed]
    Shi C, Pei S, Ding Y, Tao C, Zhu Y, Peng Y, et al. Exosomes with overexpressed miR 147a suppress angiogenesis and infammatory injury in an experimental model of atopic dermatitis. Sci Rep. 2023;13:8904. [DOI] [PubMed] [PMC]
    Yang Z, Zeng B, Wang C, Wang H, Huang P, Pan Y. MicroRNA-124 alleviates chronic skin inflammation in atopic eczema via suppressing innate immune responses in keratinocytes. Cell Immunol. 2017;319:5360. [DOI] [PubMed]
    Zeng Y, Nguyen GH, Jin H. MicroRNA-143 inhibits IL-13-induced dysregulation of the epidermal barrier-related proteins in skin keratinocytes via targeting to IL-13Rα1. Mol Cell Biochem. 2016;416:6370. [DOI] [PubMed]
    Rebane A, Runnel T, Aab A, Maslovskaja J, Rückert B, Zimmermann M, et al. MicroRNA-146a alleviates chronic skin inflammation in atopic dermatitis through suppression of innate immune responses in keratinocytes. J Allergy Clin Immunol. 2014;134:83647, e11. [DOI] [PubMed]
    Yan C, Ying J, Lu W, Changzhi Y, Qihong Q, Jingzhu M, et al. MiR-1294 suppresses ROS-dependent inflammatory response in atopic dermatitis via restraining STAT3/NF-κB pathway. Cell Immunol. 2022;371:104452. [DOI] [PubMed]
    Mitamura Y, Nunomura S, Nanri Y, Ogawa M, Yoshihara T, Masuoka M, et al. The IL-13/periostin/IL-24 pathway causes epidermal barrier dysfunction in allergic skin inflammation. Allergy. 2018;73:188191. [DOI] [PubMed]
    Chan OM, Xu W, Cheng NS, Leung ASY, Ching JYL, Fong BLY, et al. A novel infant microbiome formula (SIM03) improved eczema severity and quality of life in preschool children. Sci Rep. 2024;14:3168. [DOI] [PubMed] [PMC]
    Gürtler A, Rades T, Heinz A. Electrospun fibers for the treatment of skin diseases. J Control Release. 2023;363:62140. [DOI] [PubMed]
    Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct Target Ther. 2022;7:93. [DOI] [PubMed] [PMC]
    Pal LB, Bule P, Khan W, Chella N. An Overview of the Development and Preclinical Evaluation of Antibody-Drug Conjugates for Non-Oncological Applications. Pharmaceutics. 2023;15:1807. [DOI] [PubMed] [PMC]
    Dragovich PS. Antibody-Drug Conjugates for Immunology. J Med Chem. 2022;65:44969. [DOI] [PubMed]
    Yu S, Pearson AD, Lim RK, Rodgers DT, Li S, Parker HB, et al. Targeted Delivery of an Anti-inflammatory PDE4 Inhibitor to Immune Cells via an Antibody-drug Conjugate. Mol Ther. 2016;24:207889. [DOI] [PubMed] [PMC]
    Rodak A, Stadlbauer K, Bobbili MR, Smrzka O, Rüker F, Knopp GW. Development of a Cytotoxic Antibody-Drug Conjugate Targeting Membrane Immunoglobulin E-Positive Cells. Int J Mol Sci. 2023;24:14997. [DOI] [PubMed] [PMC]
    Everts M, Kok RJ, Asgeirsdóttir SA, Melgert BN, Moolenaar TJM, Koning GA, et al. Selective intracellular delivery of dexamethasone into activated endothelial cells using an E-selectin-directed immunoconjugate. J Immunol. 2002;168:8839. [DOI] [PubMed]
    Graversen JH, Svendsen P, Dagnæs-Hansen F, Dal J, Anton G, Etzerodt A, et al. Targeting the hemoglobin scavenger receptor CD163 in macrophages highly increases the anti-inflammatory potency of dexamethasone. Mol Ther. 2012;20:15508. [DOI] [PubMed] [PMC]
    Barth S. Recombinant immunotoxins--the next generation. Curr Pharm Des. 2009;15:26501. [DOI] [PubMed]
    Mungra N, Jordaan S, Hlongwane P, Naran K, Chetty S, Barth S. Targeted human cytolytic fusion proteins at the cutting edge: harnessing the apoptosis-inducing properties of human enzymes for the selective elimination of tumor cells. Oncotarget. 2019;10:897915. [DOI] [PubMed] [PMC]
    Akinrinmade OA, Chetty S, Daramola AK, Islam M, Thepen T, Barth S. CD64: An Attractive Immunotherapeutic Target for M1-type Macrophage Mediated Chronic Inflammatory Diseases. Biomedicines. 2017;5:56. [DOI] [PubMed] [PMC]
    Ribbert T, Thepen T, Tur MK, Fischer R, Huhn M, Barth S. Recombinant, ETA’-based CD64 immunotoxins: improved efficacy by increased valency, both in vitro and in vivo in a chronic cutaneous inflammation model in human CD64 transgenic mice. Br J Dermatol. 2010;163:27986. [DOI] [PubMed]
    Thepen T, van Vuuren AJ, Kiekens RC, Damen CA, Vooijs WC, van De Winkel JG. Resolution of cutaneous inflammation after local elimination of macrophages. Nat Biotechnol. 2000;18:4851. [DOI] [PubMed]
    Hristodorov D, Mladenov R, von Felbert V, Huhn M, Fischer R, Barth S, et al. Targeting CD64 mediates elimination of M1 but not M2 macrophages in vitro and in cutaneous inflammation in mice and patient biopsies. MAbs. 2015;7:85362. [DOI] [PubMed] [PMC]
    Jiemy WF, Hiew LF, Sha HX, In LLA, Hwang JS. Evaluation of Hydra HALT-1 as a toxin moiety for recombinant immunotoxin. BMC Biotechnol. 2020;20:31. [DOI] [PubMed] [PMC]
    Hristodorov D, Mladenov R, Fischer R, Barth S, Thepen T. Fully human MAP-fusion protein selectively targets and eliminates proliferating CD64+ M1 macrophages. Immunol Cell Biol. 2016;94:4708. [DOI] [PubMed]
    El-Kadiry AE, Rafei M, Shammaa R. Cell Therapy: Types, Regulation, and Clinical Benefits. Front Med (Lausanne). 2021;8:756029. [DOI] [PubMed] [PMC]
    Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med. 2019;4:22. [DOI] [PubMed] [PMC]
    de Oliveira Ramos F, Malard PF, Brunel HDSS, Paludo GR, de Castro MB, da Silva PHS, et al. Canine atopic dermatitis attenuated by mesenchymal stem cells. J Adv Vet Anim Res. 2020;7:55465. [DOI] [PubMed] [PMC]
    Dias IE, Pinto PO, Barros LC, Viegas CA, Dias IR, Carvalho PP. Mesenchymal stem cells therapy in companion animals: useful for immune-mediated diseases? BMC Vet Res. 2019;15:358. [DOI] [PubMed] [PMC]
    Na K, Yoo HS, Zhang YX, Choi M, Lee K, Yi TG, et al. Bone marrow-derived clonal mesenchymal stem cells inhibit ovalbumin-induced atopic dermatitis. Cell Death Dis. 2014;5:e1345. [DOI] [PubMed] [PMC]
    Kim H, Yun J, Shin T, Lee S, Lee B, Yu K, et al. Human umbilical cord blood mesenchymal stem cell-derived PGE2 and TGF-β1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells. 2015;33:125466. [DOI] [PubMed]
    Shin T, Lee B, Choi SW, Shin J, Kang I, Lee JY, et al. Human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis via regulation of B lymphocyte maturation. Oncotarget. 2017;8:51222. [DOI] [PubMed] [PMC]
    Kim M, Lee S, Kim Y, Kwon Y, Park Y, Lee H, et al. Human Adipose Tissue-Derived Mesenchymal Stem Cells Attenuate Atopic Dermatitis by Regulating the Expression of MIP-2, miR-122a-SOCS1 Axis, and Th1/Th2 Responses. Front Pharmacol. 2018;9:1175. [DOI] [PubMed] [PMC]
    Sah SK, Agrahari G, Nguyen CT, Kim Y, Kang K, Kim T. Enhanced therapeutic effects of human mesenchymal stem cells transduced with superoxide dismutase 3 in a murine atopic dermatitis-like skin inflammation model. Allergy. 2018;73:236476. [DOI] [PubMed]
    Lee B, Kim J, Lee JY, Kang I, Shin N, Lee S, et al. Disease-specific primed human adult stem cells effectively ameliorate experimental atopic dermatitis in mice. Theranostics. 2019;9:360821. [DOI] [PubMed] [PMC]
    Park A, Park H, Yoon J, Kang D, Kang M, Park Y, et al. Priming with Toll-like receptor 3 agonist or interferon-gamma enhances the therapeutic effects of human mesenchymal stem cells in a murine model of atopic dermatitis. Stem Cell Res Ther. 2019;10:66. [DOI] [PubMed] [PMC]
    Kim C, Hong S, Kim S, Yu JI, Jung S, Bang CH, et al. Skin repair and immunoregulatory effects of myeloid suppressor cells from human cord blood in atopic dermatitis. Front Immunol. 2024;14:1263646. [DOI] [PubMed] [PMC]
    Lee H, Lee TW, Chandrasekharan A, Sung S, Yim S, Kim S, et al. Injectable Self-Crosslinkable Thiolated Hyaluronic Acid for Stem Cell Therapy of Atopic Dermatitis. ACS Biomater Sci Eng. 2022;8:161322. [DOI] [PubMed]
    Pang QQ, Noh BW, Park HS, Kim YS, Kim JH, Cho EJ. Improvement Effect of Membrane-Free Stem Cell Extract on Atopic Dermatitis in NC/Nga Mice. Appl Sci. 2023;13:113. [DOI]
    Guan J, Li Y, Lu F, Feng J. Adipose-derived stem cells ameliorate atopic dermatitis by suppressing the IL-17 expression of Th17 cells in an ovalbumin-induced mouse model. Stem Cell Res Ther. 2022;13:98. [DOI] [PubMed] [PMC]
    Hua C, Liang Q, Chen S, Zhu J, Tang Y, Chen X, et al. Human umbilical cord mesenchymal stem cell treatment alleviates symptoms in an atopic dermatitis-like mouse model. Stem Cell Res Ther. 2023;14:147. [DOI] [PubMed] [PMC]
    Murer P, Neri D. Antibody-cytokine fusion proteins: A novel class of biopharmaceuticals for the therapy of cancer and of chronic inflammation. N Biotechnol. 2019;52:4253. [DOI] [PubMed] [PMC]
    Zhang J, An J. Cytokines, inflammation, and pain. Int Anesthesiol Clin. 2007;45:2737. [DOI] [PubMed] [PMC]
    Hemmerle T, Zgraggen S, Matasci M, Halin C, Detmar M, Neri D. Antibody-mediated delivery of interleukin 4 to the neo-vasculature reduces chronic skin inflammation. J Dermatol Sci. 2014;76:96103. [DOI] [PubMed]
    Rybchenko VS, Aliev TK, Panina AA, Kirpichnikov MP, Dolgikh DA. Targeted Cytokine Delivery for Cancer Treatment: Engineering and Biological Effects. Pharmaceutics. 2023;15:336. [DOI] [PubMed] [PMC]
    Huggenberger R, Ullmann S, Proulx ST, Pytowski B, Alitalo K, Detmar M. Stimulation of lymphangiogenesis via VEGFR-3 inhibits chronic skin inflammation. J Exp Med. 2010;207:225569. [DOI] [PubMed] [PMC]
    Schwager S, Renner S, Hemmerle T, Karaman S, Proulx ST, Fetz R, et al. Antibody-mediated delivery of VEGF-C potently reduces chronic skin inflammation. JCI Insight. 2018;3:e124850. [DOI] [PubMed] [PMC]
    Cousin N, Bartel S, Scholl J, Tacconi C, Egger A, Thorhallsdottir G, et al. Antibody-Mediated Delivery of VEGF-C Promotes Long-Lasting Lymphatic Expansion That Reduces Recurrent Inflammation. Cells. 2022;12:172. [DOI] [PubMed] [PMC]
    Chiricozzi A, Krueger JG. IL-17 targeted therapies for psoriasis. Expert Opin Investig Drugs. 2013;22:9931005. [DOI] [PubMed]
    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;295:45964. [DOI] [PubMed]
    Renert-Yuval Y, Guttman-Yassky E. Monoclonal antibodies for the treatment of atopic dermatitis. Curr Opin Allergy Clin Immunol. 2018;18:35664. [DOI] [PubMed]
    Zhao Y, Wu L, Lu Q, Gao X, Zhu X, Yao X, et al. The efficacy and safety of dupilumab in Chinese patients with moderate-to-severe atopic dermatitis: a randomized, double-blind, placebo-controlled study. Br J Dermatol. 2022;186:63341. [DOI] [PubMed] [PMC]
    Popovic B, Breed J, Rees DG, Gardener MJ, Vinall LMK, Kemp B, 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;429:20819. [DOI] [PubMed]
    Ultsch M, Bevers J, Nakamura G, Vandlen R, Kelley RF, Wu LC, et al. Structural basis of signaling blockade by anti-IL-13 antibody Lebrikizumab. J Mol Biol. 2013;425:13309. [DOI] [PubMed]
    Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3:67380. [DOI] [PubMed]
    Gittler JK, Shemer A, Suárez-Fariñas M, Fuentes-Duculan J, Gulewicz KJ, Wang CQF, et al. Progressive activation of Th2/Th22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. J Allergy Clin Immunol. 2012;130:134454. [DOI] [PubMed] [PMC]
    Nograles KE, Zaba LC, Shemer A, Fuentes-Duculan J, Cardinale I, Kikuchi T, et al. IL-22-producing “T22” T cells account for upregulated IL-22 in atopic dermatitis despite reduced IL-17-producing TH17 T cells. J Allergy Clin Immunol. 2009;123:124452, e2. [DOI] [PubMed] [PMC]
    Guttman-Yassky E, Brunner PM, Neumann AU, Khattri S, Pavel AB, Malik K, et al. Efficacy and safety of fezakinumab (an IL-22 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by conventional treatments: A randomized, double-blind, phase 2a trial. J Am Acad Dermatol. 2018;78:87281, e6. [DOI] [PubMed] [PMC]
    Zheng Y, Danilenko DM, Valdez P, Kasman I, Eastham-Anderson J, Wu J, et al. Interleukin-22, a TH17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature. 2007;445:64851. [DOI] [PubMed]
    Vakharia PP, Silverberg JI. Monoclonal Antibodies for Atopic Dermatitis: Progress and Potential. BioDrugs. 2017;31:40922. [DOI] [PubMed]
    Khattri S, Brunner PM, Garcet S, Finney R, Cohen SR, Oliva M, et al. Efficacy and safety of ustekinumab treatment in adults with moderate-to-severe atopic dermatitis. Exp Dermatol. 2017;26:2835. [DOI] [PubMed] [PMC]
    Silverberg JI, Strober B, Feinstein B, Xu J, Guttman-Yassky E, Simpson EL, et al. Efficacy and safety of rademikibart (CBP-201), a next-generation mAb targeting IL-4Rα, in adults with moderate to severe atopic dermatitis: A phase 2 randomized trial (CBP-201-WW001). J Allergy Clin Immunol. 2024;153:10409, e12. [DOI] [PubMed]
    Wynne CJ, Cole A, Lemech C, Wang G, Zhang Y, Chen B, et al. Safety, Pharmacokinetics and Preliminary Efficacy of IL4-Rα Monoclonal Antibody AK120 in Both Healthy and Atopic Dermatitis Subjects: A Phase I, Randomized, Two-Part, Double-Blind, Placebo-Controlled, Dose-Escalation, First-In-Human Clinical Study. Dermatol Ther (Heidelb). 2023;13:235773. [DOI] [PubMed] [PMC]
    Paller AS, Simpson EL, Siegfried EC, Cork MJ, Wollenberg A, Arkwright PD, et al. Dupilumab in children aged 6 months to younger than 6 years with uncontrolled atopic dermatitis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2022;400:90819. [DOI] [PubMed]
    Beck LA, Thaçi D, Hamilton JD, Graham NM, Bieber T, Rocklin R, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371:1309. [DOI] [PubMed]
    Simpson EL, Bieber T, Guttman-Yassky E, Beck LA, Blauvelt A, Cork MJ, et al.; SOLO 1 and SOLO 2 Investigators. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N Engl J Med. 2016;375:233548. [DOI] [PubMed]
    Blauvelt A, de Bruin-Weller M, Gooderham M, Cather JC, Weisman J, Pariser 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;389:2287303. [DOI] [PubMed]
    Wu JJ, Spelman L, Tan JL, Etoh T, Zhang H, Shumel B, et al. Dupilumab Maintains Long-Term Disease Control in Adults with Moderate-to-Severe Atopic Dermatitis as Measured by Well-Controlled Weeks: Results From the LIBERTY AD CHRONOS Clinical Trial. Dermatol Ther (Heidelb). 2021;11:32730. [DOI] [PubMed] [PMC]
    de Bruin-Weller M, Thaçi D, Smith CH, Reich K, Cork MJ, Radin A, et al. Dupilumab with concomitant topical corticosteroid treatment in adults with atopic dermatitis with an inadequate response or intolerance to ciclosporin A or when this treatment is medically inadvisable: a placebo-controlled, randomized phase III clinical trial (LIBERTY AD CAFÉ). Br J Dermatol. 2018;178:1083101. [DOI] [PubMed]
    Silverberg JI, Adam DN, Zirwas M, Kalia S, Gutermuth J, Pinter A, 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;23:54759. [DOI] [PubMed] [PMC]
    Simpson EL, Blauvelt A, Silverberg JI, Cork MJ, Katoh N, Mark T, et al. Tralokinumab Provides Clinically Meaningful Responses at Week 16 in Adults with Moderate-to-Severe Atopic Dermatitis Who Do Not Achieve IGA 0/1. Am J Clin Dermatol. 2024;25:13948. [DOI] [PubMed] [PMC]
    Paller AS, Flohr C, Cork M, Bewley A, Blauvelt A, Hong HC, et al. Efficacy and Safety of Tralokinumab in Adolescents With Moderate to Severe Atopic Dermatitis: The Phase 3 ECZTRA 6 Randomized Clinical Trial. JAMA Dermatol. 2023;159:596605. [DOI] [PubMed] [PMC]
    Paller AS, Flohr C, Eichenfield LF, Irvine AD, Weisman J, Soung J, et al. Safety and Efficacy of Lebrikizumab in Adolescent Patients with Moderate-to-Severe Atopic Dermatitis: A 52-Week, Open-Label, Phase 3 Study. Dermatol Ther (Heidelb). 2023;13:151734. [DOI] [PubMed] [PMC]
    Blauvelt A, Thyssen JP, Guttman-Yassky E, Bieber T, Serra-Baldrich E, Simpson E, et al. Efficacy and safety of lebrikizumab in moderate-to-severe atopic dermatitis: 52-week results of two randomized double-blinded placebo-controlled phase III trials. Br J Dermatol. 2023;188:74048. [DOI] [PubMed]
    Ungar B, Pavel AB, Li R, Kimmel G, Nia J, Hashim P, et al. Phase 2 randomized, double-blind study of IL-17 targeting with secukinumab in atopic dermatitis. J Allergy Clin Immunol. 2021;147:39497. [DOI] [PubMed]
    Tyring SK, Rich P, Tada Y, Beeck S, Messina I, Liu J, et al. Risankizumab in Patients with Moderate-to-Severe Atopic Dermatitis: A Phase 2, Randomized, Double-Blind, Placebo-Controlled Study. Dermatol Ther (Heidelb). 2023;13:595608. [DOI] [PubMed] [PMC]
    Kabashima K, Matsumura T, Komazaki H, Kawashima M; Nemolizumab JP01 and JP02 Study Group. Nemolizumab plus topical agents in patients with atopic dermatitis (AD) and moderate-to-severe pruritus provide improvement in pruritus and signs of AD for up to 68 weeks: results from two phase III, long-term studies. Br J Dermatol. 2022;186:64251. [DOI] [PubMed] [PMC]
    Kabashima K, Matsumura T, Komazaki H, Kawashima M; Group NS; Abe M, et al.; Nemolizumab-JP01 Study Group. Trial of Nemolizumab and Topical Agents for Atopic Dermatitis with Pruritus. N Engl J Med. 2020;383:14150. [DOI] [PubMed]
    Ruzicka T, Hanifin JM, Furue M, Pulka G, Mlynarczyk I, Wollenberg A, et al.; XCIMA Study Group. Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis. N Engl J Med. 2017;376:82635. [DOI] [PubMed]
    Silverberg JI, Pinter A, Alavi A, Lynde C, Bouaziz J, Wollenberg A, et al. Nemolizumab is associated with a rapid improvement in atopic dermatitis signs and symptoms: subpopulation (EASI ≥ 16) analysis of randomized phase 2B study. J Eur Acad Dermatol Venereol. 2021;35:156268. [DOI] [PubMed]
    Igarashi A, Katsunuma T, Matsumura T, Komazaki H; Group NS; Takahashi H, et al.; Nemolizumab-JP04 Study Group. Efficacy and safety of nemolizumab in paediatric patients aged 6-12 years with atopic dermatitis with moderate-to-severe pruritus: results from a phase III, randomized, double-blind, placebo-controlled, multicentre study. Br J Dermatol. 2023;190:208. [DOI] [PubMed]
    Maurer M, Cheung DS, Theess W, Yang X, Dolton M, Guttman A, et al. Phase 2 randomized clinical trial of astegolimab in patients with moderate to severe atopic dermatitis. J Allergy Clin Immunol. 2022;150:151724. [DOI] [PubMed]
    Chen Y, Gutowska-Owsiak D, Hardman CS, Westmoreland M, MacKenzie T, Cifuentes L, 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. [DOI] [PubMed]
    Bissonnette R, Abramovits W, Proulx C, Lee P, Yassky EG, Zovko E, et al. Spesolimab, an anti-interleukin-36 receptor antibody, in patients with moderate-to-severe atopic dermatitis: Results from a multicentre, randomized, double-blind, placebo-controlled, phase II a study. J Eur Acad Dermatology Venereol. 2023;37:54957. [DOI] [PubMed]
    Guttman-Yassky E, Pavel AB, Zhou L, Estrada YD, Zhang N, Xu H, et al. GBR 830, an anti-OX40, improves skin gene signatures and clinical scores in patients with atopic dermatitis. J Allergy Clin Immunol. 2019;144:48293, e7. [DOI] [PubMed]
    Late-breaking amlitelimab Phase 2b data presented at EADV show potential best-in-class profile in atopic dermatitis [Internet]. Sanofi; c2004–2024 [cited 2023 Oct 13]. Available from: https://www.sanofi.com/en/media-room/press-releases/2023/2023-10-13-14-00-00-2760021/
    Weidinger S, Bieber T, Cork MJ, Reich A, Wilson R, Quaratino S, et al. Safety and efficacy of amlitelimab, a fully human nondepleting, noncytotoxic anti-OX40 ligand monoclonal antibody, in atopic dermatitis: results of a phase IIa randomized placebo-controlled trial. Br J Dermatol. 2023;189:5319. [DOI] [PubMed]
    Iannelli M, Caminiti L, Vaccaro M, Marafioti I, Spinuzza A, Panasiti I, et al. Omalizumab for treatment of refractory severe atopic dermatitis. A pediatric perspective. Dermatol Ther. 2020;33:e13519. [DOI] [PubMed]
    Chan S, Cornelius V, Cro S, Harper JI, Lack G. Treatment Effect of Omalizumab on Severe Pediatric Atopic Dermatitis: The ADAPT Randomized Clinical Trial. JAMA Pediatr. 2020;174:2937. [DOI] [PubMed] [PMC]
    Chan S, Cornelius V, Chen T, Radulovic S, Wan M, Jahan R, et al. Atopic Dermatitis Anti-IgE Paediatric Trial (ADAPT): the role of anti-IgE in severe paediatric eczema: study protocol for a randomised controlled trial. Trials. 2017;18:136. [DOI] [PubMed] [PMC]
    Simpson EL, Parnes JR, She D, Crouch S, Rees W, Mo M, et al. 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;80:101321. [DOI] [PubMed]
    Worm M, Francuzik W, Kraft M, Alexiou A. Modern therapies in atopic dermatitis: biologics and small molecule drugs. J Dtsch Dermatol Ges. 2020;18:108592. [DOI] [PubMed]
    Bao L, Zhang H, Chan LS. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT. 2013;2:e24137. [DOI] [PubMed] [PMC]
    Guttman-Yassky E, Hanifin JM, Boguniewicz M, Wollenberg A, Bissonnette R, Purohit V, et al. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. Exp Dermatol. 2019;28:310. [DOI] [PubMed]
    Raker VK, Becker C, Steinbrink K. The cAMP Pathway as Therapeutic Target in Autoimmune and Inflammatory Diseases. Front Immunol. 2016;7:123. [DOI] [PubMed] [PMC]
    Schafer PH, Truzzi F, Parton A, Wu L, Kosek J, Zhang L, et al. Phosphodiesterase 4 in inflammatory diseases: Effects of apremilast in psoriatic blood and in dermal myofibroblasts through the PDE4/CD271 complex. Cell Signal. 2016;28:75363. [DOI] [PubMed]
    Wittmann M, Helliwell PS. Phosphodiesterase 4 inhibition in the treatment of psoriasis, psoriatic arthritis and other chronic inflammatory diseases. Dermatol Ther (Heidelb). 2013;3:115. [DOI] [PubMed] [PMC]
    Benedetto AD, Yoshida T, Fridy S, Park JS, Kuo I, Beck LA. Histamine and Skin Barrier: Are Histamine Antagonists Useful for the Prevention or Treatment of Atopic Dermatitis? J Clin Med. 2015;4:74155. [DOI] [PubMed] [PMC]
    Rathinasamy A, Czeloth N, Pabst O, Förster R, Bernhardt G. The origin and maturity of dendritic cells determine the pattern of sphingosine 1-phosphate receptors expressed and required for efficient migration. J Immunol. 2010;185:407281. [DOI] [PubMed]
    Silverberg JI, Bissonnette R, Kircik L, Murrell DF, Selfridge A, Liu K, et al. Efficacy and safety of etrasimod, a sphingosine 1-phosphate receptor modulator, in adults with moderate-to-severe atopic dermatitis (ADVISE). J Eur Acad Dermatol Venereol. 2023;37:136674. [DOI] [PubMed]
    Silverberg JI, de Bruin-Weller M, Bieber T, Soong W, Kabashima K, Costanzo A, et al. Upadacitinib plus topical corticosteroids in atopic dermatitis: Week 52 AD Up study results. J Allergy Clin Immunol. 2022;149:97787, e14. [DOI] [PubMed]
    Guttman-Yassky E, Teixeira HD, Simpson EL, Papp KA, Pangan AL, Blauvelt A, 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;397:215168. [DOI] [PubMed]
    Reich K, Teixeira HD, de Bruin-Weller M, Bieber T, Soong W, Kabashima K, et al. Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD Up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2021;397:216981. [DOI] [PubMed]
    Guttman-Yassky E, Thaçi D, Pangan AL, Hong HC, Papp KA, Reich 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;145:87784. [DOI] [PubMed]
    Shi VY, Bhutani T, Fonacier L, Deleuran M, Shumack S, Valdez H, et al. Phase 3 efficacy and safety of abrocitinib in adults with moderate-to-severe atopic dermatitis after switching from dupilumab (JADE EXTEND). J Am Acad Dermatol. 2022;87:3518. [DOI] [PubMed]
    Simpson EL, Silverberg JI, Thyssen JP, Viguier M, Thaçi D, de Bruin-Weller M, et al. Efficacy and Safety of Abrocitinib in Patients with Severe and/or Difficult-to-Treat Atopic Dermatitis: A Post Hoc Analysis of the Randomized Phase 3 JADE COMPARE Trial. Am J Clin Dermatol. 2023;24:60921. [DOI] [PubMed] [PMC]
    Bieber T, Simpson EL, Silverberg JI, Thaçi D, Paul C, Pink AE, et al.; JADE COMPARE Investigators. Abrocitinib versus Placebo or Dupilumab for Atopic Dermatitis. N Engl J Med. 2021;384:110112. [DOI] [PubMed]
    Silverberg JI, Simpson EL, Thyssen JP, Gooderham M, Chan G, Feeney C, et al. Efficacy and Safety of Abrocitinib in Patients With Moderate-to-Severe Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2020;156:86373. [DOI] [PubMed] [PMC]
    Simpson EL, Sinclair R, Forman S, Wollenberg A, Aschoff R, Cork M, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2020;396:25566. [DOI] [PubMed]
    Gooderham MJ, Forman SB, Bissonnette R, Beebe JS, Zhang W, Banfield C, et al. Efficacy and Safety of Oral Janus Kinase 1 Inhibitor Abrocitinib for Patients With Atopic Dermatitis: A Phase 2 Randomized Clinical Trial. JAMA Dermatol. 2019;155:137179. [DOI] [PubMed] [PMC]
    Zhao Y, Zhang L, Ding Y, Tao X, Ji C, Dong X, et al. Efficacy and Safety of SHR0302, a Highly Selective Janus Kinase 1 Inhibitor, in Patients with Moderate to Severe Atopic Dermatitis: A Phase II Randomized Clinical Trial. Am J Clin Dermatol. 2021;22:87789. [DOI] [PubMed] [PMC]
    Simpson EL, Forman S, Silverberg JI, Zirwas M, Maverakis E, Han G, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: Results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J Am Acad Dermatol. 2021;85:6270. [DOI] [PubMed]
    Simpson EL, Lacour J, Spelman L, Galimberti R, Eichenfield LF, Bissonnette R, 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;183:24255. [DOI] [PubMed]
    Guttman-Yassky E, Silverberg JI, Nemoto O, Forman SB, Wilke A, Prescilla R, et al. Baricitinib in adult patients with moderate-to-severe atopic dermatitis: A phase 2 parallel, double-blinded, randomized placebo-controlled multiple-dose study. J Am Acad Dermatol. 2019;80:91321, e9. [DOI] [PubMed]
    King B, Maari C, Lain E, Silverberg JI, Issa M, Holzwarth K, et al. Extended Safety Analysis of Baricitinib 2 mg in Adult Patients with Atopic Dermatitis: An Integrated Analysis from Eight Randomized Clinical Trials. Am J Clin Dermatol. 2021;22:395405. [DOI] [PubMed] [PMC]
    Wollenberg A, Nakahara T, Maari C, Peris K, Lio P, Augustin M, et al. Impact of baricitinib in combination with topical steroids on atopic dermatitis symptoms, quality of life and functioning in adult patients with moderate-to-severe atopic dermatitis from the BREEZE-AD7 Phase 3 randomized trial. J Eur Acad Dermatol Venereol. 2021;35:154352. [DOI] [PubMed] [PMC]
    Reich K, Kabashima K, Peris K, Silverberg JI, Eichenfield LF, Bieber T, et al. Efficacy and Safety of Baricitinib Combined With Topical Corticosteroids for Treatment of Moderate to Severe Atopic Dermatitis: A Randomized Clinical Trial. JAMA Dermatol. 2020;156:133343. [DOI] [PubMed] [PMC]
    Silverberg JI, Simpson EL, Wollenberg A, Bissonnette R, Kabashima K, DeLozier AM, et al. Long-term Efficacy of Baricitinib in Adults With Moderate to Severe Atopic Dermatitis Who Were Treatment Responders or Partial Responders: An Extension Study of 2 Randomized Clinical Trials. JAMA Dermatol. 2021;157:6919. [DOI] [PubMed] [PMC]
    Papp K, Szepietowski JC, Kircik L, Toth D, Eichenfield LF, Leung DYM, et al. Efficacy and safety of ruxolitinib cream for the treatment of atopic dermatitis: Results from 2 phase 3, randomized, double-blind studies. J Am Acad Dermatol. 2021;85:86372. [DOI] [PubMed]
    Kim BS, Sun K, Papp K, Venturanza M, Nasir A, Kuligowski ME. 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;82:130513. [DOI] [PubMed]
    Kim BS, Howell MD, Sun K, Papp K, Nasir A, Kuligowski ME, et al.; INCB 18424-206 Study Investigators. Treatment of atopic dermatitis with ruxolitinib cream (JAK1/JAK2 inhibitor) or triamcinolone cream. J Allergy Clin Immunol. 2020;145:57282. [DOI] [PubMed]
    Bissonnette R, Papp KA, Poulin Y, Gooderham M, Raman M, Mallbris L, et al. Topical tofacitinib for atopic dermatitis: a phase IIa randomized trial. Br J Dermatol. 2016;175:90211. [DOI] [PubMed]
    Changelian P, Xu C, Mnich S, Hope H, Kostecki K, Hirsch J, et al. ATI-1777, a Topical Jak1/3 Inhibitor, May Benefit Atopic Dermatitis without Systemic Drug Exposure: Results from Preclinical Development and Phase 2a Randomized Control Study ATI-1777-AD-201. JID Innov. 2023;4:100251. [DOI] [PubMed] [PMC]
    Smith S, Bhatia N, Shanler SD, Demoor R, Schnyder J. 16089 Safety of ATI-502, a novel topical JAK1/3 inhibitor, in adults with moderate to severe atopic dermatitis: Results from a phase 2a open-label trial. J Am Acad Dermatol. 2020;83:AB170. [DOI]
    Landis MN, Arya M, Smith S, Draelos Z, Usdan L, Tarabar S, et al. Efficacy and safety of topical brepocitinib for the treatment of mild-to-moderate atopic dermatitis: a phase IIb, randomized, double-blind, vehicle-controlled, dose-ranging and parallel-group study. Br J Dermatol. 2022;187:87887. [DOI] [PubMed] [PMC]
    Nakagawa H, Nemoto O, Yamada H, Nagata T, Ninomiya N. Phase 1 studies to assess the safety, tolerability and pharmacokinetics of JTE-052 (a novel Janus kinase inhibitor) ointment in Japanese healthy volunteers and patients with atopic dermatitis. J Dermatol. 2018;45:70109. [DOI] [PubMed] [PMC]
    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;82:82331. [DOI] [PubMed]
    Nakagawa H, Nemoto O, Igarashi A, Saeki H, Oda M, Kabashima K, et al. Phase 2 clinical study of delgocitinib ointment in pediatric patients with atopic dermatitis. J Allergy Clin Immunol. 2019;144:157583. [DOI] [PubMed]
    Nakagawa H, Nemoto O, Igarashi A, Nagata T. Efficacy and safety of topical JTE-052, a Janus kinase inhibitor, in Japanese adult patients with moderate-to-severe atopic dermatitis: a phase II, multicentre, randomized, vehicle-controlled clinical study. Br J Dermatol. 2018;178:42432. [DOI] [PubMed]
    Nakagawa H, Nemoto O, Igarashi A, Saeki H, Murata R, Kaino H, et al. Long-term safety and efficacy of delgocitinib ointment, a topical Janus kinase inhibitor, in adult patients with atopic dermatitis. J Dermatol. 2020;47:11420. [DOI] [PubMed] [PMC]
    Callender VD, Alexis AF, Gold LFS, Lebwohl MG, Paller AS, Desai SR, et al. Efficacy and Safety of Crisaborole Ointment, 2%, for the Treatment of Mild-to-Moderate Atopic Dermatitis Across Racial and Ethnic Groups. Am J Clin Dermatol. 2019;20:71123. [DOI] [PubMed] [PMC]
    Eichenfield LF, Gower RG, Xu J, Alam MS, Su JC, Myers DE, et al. Once-Daily Crisaborole Ointment, 2%, as a Long-Term Maintenance Treatment in Patients Aged ≥ 3 Months with Mild-to-Moderate Atopic Dermatitis: A 52-Week Clinical Study. Am J Clin Dermatol. 2023;24:62335. [DOI] [PubMed] [PMC]
    Paller AS, Tom WL, Lebwohl MG, Blumenthal RL, Boguniewicz M, Call RS, 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;75:494503, e6. [DOI] [PubMed]
    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 (Heidelb). 2022;12:1589601. [DOI] [PubMed] [PMC]
    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;186:409. [DOI] [PubMed] [PMC]
    Saeki H, Ito K, Yokota D, Tsubouchi H. Difamilast ointment in adult patients with atopic dermatitis: A phase 3 randomized, double-blind, vehicle-controlled trial. J Am Acad Dermatol. 2022;86:60714. [DOI] [PubMed]
    Eichenfield L, Boguniewicz M, Simpson E, Blauvelt A, Gooderham M, Lain E, et al. Once-daily roflumilast cream 0.15% for atopic dermatitis: pooled results: from Integument-1/2 phase 3 trials. Ann Allergy, Asthma Immunol. 2023;131:S91. [DOI]
    Welsh SE, Xiao C, Kaden AR, Brzezynski JL, Mohrman MA, Wang J, et al. Neurokinin-1 receptor antagonist tradipitant has mixed effects on itch in atopic dermatitis: results from EPIONE, a randomized clinical trial. J Eur Acad Dermatol Venereol. 2021;35:e33840. [DOI] [PubMed] [PMC]
    Guttman-Yassky E, Facheris P, Rosa JCD, Rothenberg-Lausell C, Duca ED, David E, et al. Oral difelikefalin reduces moderate to severe pruritus and expression of pruritic and inflammatory biomarkers in subjects with atopic dermatitis. J Allergy Clin Immunol. 2023;152:91626. [DOI] [PubMed]
    Werfel T, Layton G, Yeadon M, Whitlock L, Osterloh I, Jimenez P, et al. Efficacy and safety of the histamine H4 receptor antagonist ZPL-3893787 in patients with atopic dermatitis. J Allergy Clin Immunol. 2019;143:183037, e4. [DOI] [PubMed]
    Czarnowicki T, Dohlman AB, Malik K, Antonini D, Bissonnette R, Chan TC, et al. Effect of short-term liver X receptor activation on epidermal barrier features in mild to moderate atopic dermatitis: A randomized controlled trial. Ann Allergy Asthma Immunol. 2018;120:63140, e11. [DOI] [PubMed]
    Ra JC, Kang SK, Shin IS, Park HG, Joo SA, Kim JG, et al. Stem cell treatment for patients with autoimmune disease by systemic infusion of culture-expanded autologous adipose tissue derived mesenchymal stem cells. J Transl Med. 2011;9:181. [DOI] [PubMed] [PMC]
    Kim H, Lee JH, Roh K, Jun HJ, Kang K, Kim T. Clinical Trial of Human Umbilical Cord Blood-Derived Stem Cells for the Treatment of Moderate-to-Severe Atopic Dermatitis: Phase I/IIa Studies. Stem Cells. 2017;35:24855. [DOI] [PubMed]
    Kim Y, Ahn H, Lee S, Lee M, Kang K. Effects of conditioned media from human umbilical cord blood-derived mesenchymal stem cells in the skin immune response. Biomed Pharmacother. 2020;131:110789. [DOI] [PubMed]
    Shin H, Lee SH, Yoon HS, Heo JH, Lee SB, Byun JW, et al. Long-term efficacy and safety of intravenous injection of clonal mesenchymal stem cells derived from bone marrow in five adults with moderate to severe atopic dermatitis. J Dermatol. 2021;48:123642. [DOI] [PubMed]
    Hillier A, Griffin CE. The ACVD task force on canine atopic dermatitis (I): incidence and prevalence. Vet Immunol Immunopathol. 2001;81:14751. [DOI] [PubMed]
    Villatoro AJ, Hermida-Prieto M, Fernández V, Fariñas F, Alcoholado C, Rodríguez-García MI, et al. Allogeneic adipose-derived mesenchymal stem cell therapy in dogs with refractory atopic dermatitis: clinical efficacy and safety. Vet Rec. 2018;183:654. [DOI] [PubMed]
    Enciso N, Amiel J, Pando J, Enciso J. Multidose intramuscular allogeneic adipose stem cells decrease the severity of canine atopic dermatitis: A pilot study. Vet World. 2019;12:174754. [DOI] [PubMed] [PMC]
    Kaur G, Ramirez A, Xie C, Clark D, Dong C, Maki C, et al. A double-blinded placebo-controlled evaluation of adipose-derived mesenchymal stem cells in treatment of canine atopic dermatitis. Vet Res Commun. 2022;46:25160. [DOI] [PubMed]
    Black L, Zacharias S, Hughes M, Bautista R, Taechangam N, Sand T. The effect of uterine-derived mesenchymal stromal cells for the treatment of canine atopic dermatitis: A pilot study. Front Vet Sci. 2022;9:1011174. [DOI] [PubMed] [PMC]
    Kim S, Lim K, Cho S, Ryu B, Kim C, Park SY, et al. Efficacy of Allogeneic and Xenogeneic Exosomes for the Treatment of Canine Atopic Dermatitis: A Pilot Study. Animals (Basel). 2024;14:282. [DOI] [PubMed] [PMC]
    Rostaher A, Fischer NM, Vigani A, Steblaj B, Martini F, Brem S, et al. Hymenoptera Venom Immunotherapy in Dogs: Safety and Clinical Efficacy. Animals (Basel). 2023;13:3002. [DOI] [PubMed] [PMC]
    Fritzsching B, Contoli M, Porsbjerg C, Buchs S, Larsen JR, Elliott L, et al. Long-term real-world effectiveness of allergy immunotherapy in patients with allergic rhinitis and asthma: Results from the REACT study, a retrospective cohort study. Lancet Reg Health Eur. 2021;13:100275. [DOI] [PubMed] [PMC]
    Darsow U. Allergen-specific immunotherapy for atopic eczema: updated. Curr Opin Allergy Clin Immunol. 2012;12:6659. [DOI] [PubMed]
    Shah D, Hales J, Cooper D, Camp R. Recognition of pathogenically relevant house dust mite hypersensitivity in adults with atopic dermatitis: a new approach? J Allergy Clin Immunol. 2002;109:10128. [DOI] [PubMed]
    Guo B, Wu K, Chen C, Lin W, Chang Y, Lin M, et al. Advancements in Allergen Immunotherapy for the Treatment of Atopic Dermatitis. Int J Mol Sci. 2024;25:1316. [DOI] [PubMed] [PMC]
    Satitsuksanoa P, Angelina A, Palomares O, Akdis M. Mechanisms in AIT: Insights 2021. Allergol Select. 2022;6:25966. [DOI] [PubMed] [PMC]
    Rizk P, Rodenas M, Benedetto AD. Allergen Immunotherapy and Atopic Dermatitis: the Good, the Bad, and the Unknown. Curr Allergy Asthma Rep. 2019;19:57. [DOI] [PubMed]
    James C, Bernstein DI. Allergen immunotherapy: an updated review of safety. Curr Opin Allergy Clin Immunol. 2017;17:559. [DOI] [PubMed] [PMC]
    Kim M, Lee E, Yoon J, Jung S, Song KB, Choi EJ, et al. Sublingual immunotherapy may be effective in reducing house dust mite allergies in children with atopic dermatitis. Acta Paediatr. 2022;111:21428. [DOI] [PubMed]
    Huang C, Tang J. Sublingual immunotherapy with Dermatophagoides farinae drops for pediatric atopic dermatitis. Int J Dermatol. 2022;61:24651. [DOI] [PubMed]
    Hajdu K, Kapitány A, Dajnoki Z, Soltész L, Baráth S, Hendrik Z, et al. Improvement of clinical and immunological parameters after allergen-specific immunotherapy in atopic dermatitis. J Eur Acad Dermatol Venereol. 2021;35:135761. [DOI] [PubMed]
    Langer SS, Cardili RN, Melo JML, Ferriani MPL, Moreno AS, Dias MM, et al. Efficacy of House Dust Mite Sublingual Immunotherapy in Patients with Atopic Dermatitis: A Randomized, Double-Blind, Placebo-Controlled Trial. J Allergy Clin Immunol Pract. 2022;10:53949, e7. [DOI] [PubMed]
    Bogacz-Piaseczyńska A, Bożek A. The Effectiveness of Allergen Immunotherapy in Adult Patients with Atopic Dermatitis Allergic to House Dust Mites. Medicina (Kaunas). 2022;59:15. [DOI] [PubMed] [PMC]
    Liu L, Chen J, Xu J, Yang Q, Gu C, Ni C, et al. Sublingual immunotherapy of atopic dermatitis in mite-sensitized patients: a multi-centre, randomized, double-blind, placebo-controlled study. Artif Cells Nanomed Biotechnol. 2019;47:354047. [DOI] [PubMed]
    Yu N, Luo H, Liang D, Lu N. Sublingual immunotherapy in mite-sensitized patients with atopic dermatitis: a randomized controlled study. Postepy Dermatol Alergol. 2021;38:6974. [DOI] [PubMed] [PMC]
    Qin Y, Mao J, Sang Y, Li W. Clinical efficacy and compliance of sublingual immunotherapy with Dermatophagoides farinae drops in patients with atopic dermatitis. Int J Dermatol. 2014;53:6505. [DOI] [PubMed]
    Pajno GB, Caminiti L, Vita D, Barberio G, Salzano G, Lombardo F, et al. Sublingual immunotherapy in mite-sensitized children with atopic dermatitis: a randomized, double-blind, placebo-controlled study. J Allergy Clin Immunol. 2007;120:16470. [DOI] [PubMed]
    Chu H, Park KH, Kim SM, Lee J, Park J, Lee KH, et al. Allergen-specific immunotherapy for patients with atopic dermatitis sensitized to animal dander. Immun Inflamm Dis. 2020;8:1659. [DOI] [PubMed] [PMC]
    Novak N, Bieber T, Hoffmann M, Fölster-Holst R, Homey B, Werfel T, et al. Efficacy and safety of subcutaneous allergen-specific immunotherapy with depigmented polymerized mite extract in atopic dermatitis. J Allergy Clin Immunol. 2012;130:92531, e4. [DOI] [PubMed]
    Paiva-Santos AC, Gama M, Peixoto D, Sousa-Oliveira I, Ferreira-Faria I, Zeinali M, et al. Nanocarrier-based dermopharmaceutical formulations for the topical management of atopic dermatitis. Int J Pharm. 2022;618:121656. [DOI] [PubMed]
    Garrós N, Bustos-Salgados P, Domènech Ò, Rodríguez-Lagunas MJ, Beirampour N, Mohammadi-Meyabadi R, et al. Baricitinib Lipid-Based Nanosystems as a Topical Alternative for Atopic Dermatitis Treatment. Pharmaceuticals (Basel). 2023;16:117. [DOI] [PubMed] [PMC]
    Campos EVR, Proença PLDF, Doretto-Silva L, Andrade-Oliveira V, Fraceto LF, de Araujo DR. Trends in nanoformulations for atopic dermatitis treatment. Expert Opin Drug Deliv. 2020;17:161530. [DOI] [PubMed]
    Brunner PM, Pavel AB, Khattri S, Leonard A, Malik K, Rose S, et al. Baseline IL-22 expression in patients with atopic dermatitis stratifies tissue responses to fezakinumab. J Allergy Clin Immunol. 2019;143:14254. [DOI] [PubMed]
    Leung DYM. Evolving atopic dermatitis toward precision medicine. Ann Allergy Asthma Immunol. 2024;132:1078. [DOI] [PubMed]
    Ghosh D, Ding L, Sivaprasad U, Geh E, Myers JB, Bernstein JA, et al. Multiple Transcriptome Data Analysis Reveals Biologically Relevant Atopic Dermatitis Signature Genes and Pathways. PLoS One. 2015;10:e0144316. [DOI] [PubMed] [PMC]
    Yamamoto-Hanada K, Kawakami E, Saito-Abe M, Sato M, Mitsubuchi H, Oda M, et al. Exploratory analysis of plasma cytokine/chemokine levels in 6-year-old children from a birth cohort study. Cytokine. 2020;130:155051. [DOI] [PubMed]
    Pomi FL, Papa V, Borgia F, Vaccaro M, Pioggia G, Gangemi S. Artificial Intelligence: A Snapshot of Its Application in Chronic Inflammatory and Autoimmune Skin Diseases. Life (Basel). 2024;14:516. [DOI] [PubMed] [PMC]