Introduction

Among dermatological disorders, psoriasis (PsO) and atopic dermatitis (AD) represent the two most prominent immune-mediated inflammatory skin diseases—one characterized primarily by Th17 dysregulation, and the other by Th2 imbalance. With the advent of biologic therapies, both diseases have entered a new era of targeted treatment, leading to remarkable clinical improvement.1–3 However, this deliberate modulation of the immune system also raises important concerns regarding potential systemic risks, which dermatologists should remain vigilant about.

Interestingly, targeting the TNF/IL-17/23 axis in psoriasis or the IL-4/13 axis in AD can sometimes result in a phenotypic shift toward the opposite disease.4 Such paradoxical transitions may necessitate treatment interruption or switching, thereby increasing the risk of disease exacerbation.

This review systematically summarizes the underlying mechanisms of both diseases and, for the first time, compiles all currently reported cases of paradoxical reactions. It further elucidates the potential causes of these phenomena and highlights how advanced sequencing technologies have deepened our understanding of their immunopathogenesis.

Contrasting Immunopathogenesis
Immunology of Psoriasis

Psoriasis is a chronic inflammatory disease driven by immune dysregulation, characterized by excessive proliferation and abnormal differentiation of keratinocytes. The inflammation in psoriasis is closely related to both innate and acquired immune responses, with its pathogenesis showing a significant genetic background.5–7

In the initial stages of psoriasis, antimicrobial peptide LL-37, released by keratinocytes under stress, forms complexes with DNA/RNA liberated from damaged cells,8–10 The complex activates plasmacytoid dendritic cells (pDC) through Toll-like receptors (TLR9/TLR7), inducing them to produce large amounts of type I interferons (such as IFN α).11,12 Subsequently, pDC activate myeloid dendritic cells (mDC).13

The axis of IL-23/Th17 cells is a central hub in the inflammatory loop of psoriasis.8,14 IL-23, the most important upstream signaling factor driving the production of key effector molecules IL-17A and IL-17F, not only regulates the proliferation and maturation of Th17 cells but also plays a pivotal role in their function.15–19 It is important to note that Th17 cells are not the only source of IL-17. Other immune cells, especially γδ T cells and type 3 innate lymphoid cells, are important sources of IL-17 in skin lesions. These cells, which can respond rapidly to IL-23, may play a “pioneer” role in the early initiation and acute exacerbation of the disease, making the inflammatory response more rapid and powerful.20 Th17 cells, through surface homing receptors such as CCR6, recognize chemotactic signals like CCL20 produced in inflamed skin, migrating in a directed manner and infiltrating the dermis and epidermis, further exacerbate the inflammatory response.21–25

In the pathogenesis of psoriasis, an inflammatory environment centered on the IL-23/Th17 axis is formed under the reactivation of local antigens by skin-resident memory T cells and immune cells recruited to the site.26 IL-17A has been confirmed as a key factor that directly causes the abnormal proliferation of keratinocytes, the production of chemokines, and the accumulation of neutrophils in the epidermis.27 TNF-α and IFN-γ, on the other hand, amplify the inflammatory response, promoting the dilation of dermal blood vessels, congestion, and lymphocyte infiltration.28 IL-22 is the key factor driving the excessive proliferation and abnormal differentiation of keratinocytes, leading to epidermal thickening and the formation of numerous silver-white scales.29–32 while the self-amplifying inflammatory loop formed between these cytokines and keratinocytes causes the skin lesions to persist stubbornly, become chronic, and well-demarcated.33,34

This process, sustained by cytokines such as IL-23,35 forms a self-amplifying inflammatory loop.36 Immune cells in the lesions activate keratinocytes, while abnormal keratinocytes, in turn, sustain and amplify the immune response. The existence of this vicious cycle is considered the fundamental immunological basis for the chronic development and frequent relapse of psoriasis.35,36

Immunology of Atopic Dermatitis

Atopic dermatitis, which is commonly referred to as eczema, is a prevalent, chronic, and relapsing inflammatory skin disorder.37,38 Similar to psoriasis, it is also an immune-mediated disease, but its immunological mechanisms are distinctly different.39 The core of atopic dermatitis lies in a vicious cycle between skin barrier defects and immune dysregulation, involving multiple immune pathways that dynamically change across different stages and subtypes.40

The immune mechanism of AD, with the main characteristic being type 2 immune response, especially evident in the acute phase and in Asian populations or early-onset types.41,42 After the damage to the epithelial barrier, alerting factors such as thymic stromal lymphopoietin (TSLP), IL-25, and IL-33 are first released by keratinocytes and others.43 These cytokines directly activate type 2 innate lymphoid cells (ILC2s), causing them to rapidly produce IL-5, IL-9, and IL-13, The dendritic cells, which can also be activated, subsequently drive the differentiation of naive T cells into Th2 cells.44–46 Finally, the activated ILC2s and Th2 cells work together to release a series of cytokines, including IL-4, IL-5, IL-9, IL-13, and IL-31, in large amounts.40,47–49 This leads to an increase in eosinophils, the large production of immunoglobulin E (IgE), and associated inflammation, thereby causing the typical pathological changes of AD.46,50

IL-4 and IL-13, which have synergistic and complementary core functions in the Th2 immune response.45,51 Specifically, IL-4 is the key inducer factor that drives the differentiation of naive T cells into Th2 cells.52 At the same time, it is also the major cytokine that promotes class switching in B cells and the production of large amounts of IgE.51 IL-13, on the other hand, primarily acts on tissue cells, directly damaging the integrity of the skin barrier by downregulating the expression of key barrier proteins, such as filaggrin.53 These two cytokines together form a self-reinforcing inflammatory loop: IL-4 ensures the continuous activation and proliferation of Th2 cells, while IL-13, by disrupting the epithelial barrier, creates conditions for the invasion of external allergens, thereby continuously stimulating the Th2 immune pathway.54

IL-31 plays a unique role in linking the immune system and the nervous system. IL-31, primarily produced by activated Th2 cells, directly activates itch-related neural pathways on sensory neurons,41,44,55–57 This intense pruritic effect leads to the characteristic scratching behavior in patients, which further damages the already fragile skin barrier.58 This forms a vicious cycle of Th2 immune activation-IL-31 production-itching-scratching-barrier disruption-further Th2 activation.59–62 It is worth noting that the production of IL-31 itself is also positively regulated by IL-4 and IL-13.63

The immune response in patients with AD can also manifest as the upregulation of Th1, Th17, and Th22 pathways, with Th1 particularly associated with chronic skin lesions, Th17 has more prominent characteristics in Asian populations.64–66

Antagonistic Mechanism
The Mutual Inhibition Between Th1/Th17 and Th2

Psoriasis and AD are two of the most common chronic inflammatory skin diseases, which exhibit significant differences in clinical presentation, immunological characteristics, and histopathology. Traditionally, psoriasis is defined as a disease dominated by Th1/Th17-type immune responses, while atopic dermatitis is defined as a disease dominated by Th2-type. A large body of clinical and basic research evidence suggests that there exists an antagonistic relationship between the immune pathways of these two diseases. A bidirectional inhibitory immune network is formed between Th1/Th17 cells and Th2 cells, along with their characteristic cytokines. There exists a dynamic balance between the Th1/Th17 and Th2 immune axes, with suppression of one side leading to dominance of the other.67–70

In this dynamic immune network with balance, IFN-γ, as one of the major cytokines of Th1 cells, not only directly inhibits the occurrence of Th2 immune responses but also further strengthens the immune activity of Th1/Th17.71 The inhibitory effect is primarily manifested in the transcriptional level, IFN-γ, by activating the STAT1 signaling pathway, suppresses the expression of GATA3, a key transcription factor essential for Th2 differentiation, thereby preventing the differentiation of naïve T cells into Th2 cells at the source.72,73 IFN-γ can also suppress the production of Th2 characteristic cytokines, thereby weakening the core components of Th2 inflammation.74–77 IFN-γ even further enhances the Th1 immune response.78 In addition, IFN-γ can also shape a local immune microenvironment unfavorable to Th2 responses by regulating the functions of macrophages and dendritic cells.79–81

Research has shown that IL-4 and IL-13, as typical Th2 cytokines, have a classic mutual inhibitory relationship with the Th17 pathway. IL-4 and IL-13 can inhibit the differentiation and function of Th17 cells through activating STAT6. It had been found in models of AD, psoriasis, and infections.82–84 Further studies had shown that IL-4 can prevent the differentiation of naive T cells into Th17 cells by inhibiting STAT3 and the expression of RORγt.85–88 Moreover, some scholars have hypothesized that in Th2-dominated diseases such as AD, IL-4 and IL-13 may indirectly alter the local cytokine milieu by suppressing the expression of keratinocyte terminal differentiation–related genes (eg, FLG, LOR, CDSN). This suppression could attenuate the tissue’s responsiveness to IL-17 signaling. However, this assumption remains to be further validated through experimental evidence.89 IL-13 induced by antigens like IPSE/α-1 may inhibit the Th17 pathway, though its pathological relevance in human diseases remains unclear.86

The Differences in Keratinocyte Response to Various Cytokines

In the classic mutual antagonism model, while the interaction between immune cells is central, keratinocytes can interpret and respond to various cytokine signals, thereby amplifying and accentuating the distinct immune features. In psoriasis, keratinocytes exhibit a proliferative response, promoting cell proliferation, inhibiting differentiation, and releasing chemokines through IL-17/IL-22.90–93 In AD, keratinocytes exhibit a barrier-disruptive response to Th2 signaling, downregulating barrier proteins with IL-4/IL-13, impairing skin defense functions.94–96 This differentiated response mechanism makes keratinocytes not only participants in immune responses but play a decisive role in the immunological opposition between the two diseases.97

Shared Pathways May Act as a Bridge

Although PsO and AD exhibit opposing immune responses, multiple shared immune pathways between them. The key bridges connect the two diseases. These overlapping mechanisms highlight the complexity of their pathogenesis and provide valuable insights into potential therapeutic targets.

The immune mechanism of AD is not a singular Th2 model. There are an immune pathway overlap driven by factors such as the IL-23/IL-17 axis and TSLP, similar to PsO. This contributes to disease heterogeneity and variability in treatment responses, which are particularly pronounced in Asian populations.98 Moreover, TSLP secreted by keratinocytes not only drives Th2 responses but, in certain contexts, can also interact with IL-17–driven inflammation, thereby modulating the direction of the immune response.99–101 Additionally, the JAK-STAT pathway, a downstream integration point for these signals, is capable of integrating various cytokine signals from psoriasis and AD.102–104 IL-36, a potent pro-inflammatory factor, is also upregulated in severe subtypes of AD. It serves as a key link between skin barrier disruption and amplified inflammatory responses (Figure 1).

Figure 1 Psoriasis (left) is driven by IL-23/Th17 cytokines that cause keratinocyte excessive proliferation and abnormal differentiation. Atopic dermatitis (right) is mediated by Th2 cytokines (IL-4, IL-13, IL-31) leading to skin inflammation and pruritus. The center shows shared JAK-STAT signaling and mutual inhibition between Th17/Th1 and Th2 pathways.

In summary, these shared immune pathways indicate that the immunological opposition between psoriasis and atopic dermatitis is not entirely independent. Rather, a complex and dynamic interplay exists between them, providing a new theoretical foundation for the development of cross-disease, broad-spectrum therapeutic strategies.105

Clinical Presentations and Epidemiological Support

AD and PsO, both common chronic inflammatory skin diseases, show an opposing trend in their pathogenesis, as demonstrated by extensive clinical observations and epidemiological studies. This negative correlation provides strong demographic and clinical support.

The Coexistence of Psoriasis and Atopic Dermatitis in a Single Patient is Exceedingly Rare

In clinical practice, it is relatively rare to encounter a patient simultaneously diagnosed with classic AD and PsO, a clinical observation supported by data from large-scale epidemiological investigations.106 For instance, a cohort study demonstrated a significantly reduced risk of psoriasis among patients with AD compared to the general population.107 And conversely, his inverse correlation suggests that the Th2-dominated immune microenvironment driving AD may suppress the establishment or development of the characteristic Th17 immune response in PsO, and, conversely, this immunological antagonism or deviation represents a key factor that constitutes their seemingly contradictory relationship.108

Epidemiological Data Showing a Negative Correlation in Patient Populations

This negative correlation becomes more evident when observed at the population level. Mendelian randomization analysis of genome-wide association studies (GWAS) provides strong evidence at the genetic level. Multiple studies have found that genetic loci associated with AD exhibit a slight protective effect against the development of PsO.109–111 This suggests that the individual’s innate immune genetic background may predispose them to develop a specific type of inflammatory pattern. It may reduce the risk of developing immune diseases of the opposite type.112

Case Reports of Special Cases

Despite their exclusivity, there are indeed case reports of AD and PsO occurring concurrently or alternately in the same patient. These special cases provide valuable insights for understanding the complex interactions of their immune pathways.

Some patients experience AD in childhood that remits with age, followed by psoriasis appearing in adolescence or adulthood.113,114 Biologic agents targeting the Th2 pathway, such as dupilumab (anti–IL-4Rα antibody), have also been reported to induce psoriasis-like eruptions or unmask previously undiagnosed psoriasis in a small subset of patients treated for AD. The underlying mechanism is thought to involve a rebound dominance of the Th1/Th17 immune response once Th2 inflammation is strongly suppressed, thereby inducing a psoriasis-like phenotype.115–117 This proves the existence of a dynamic balance between the two pathways.69,118

In a very small number of patients, the lesions may appear typical AD and PsO lesions in different sites.113,119–121 Such cases suggest the presence of a highly complex immune disturbance.122

Disparities Between Pediatric and Adult Populations

AD occurs predominantly in infancy and early childhood, while the peak onset of PsO occurs in adulthood.123–125 However, the differential diagnosis in pediatric cases can be more challenging. Chronic and severe AD can lead to latensification which may clinically resemble the plaque-like lesions of PsO.126 In addition, guttate psoriasis triggered by infections in children sometimes requires differentiation from AD exacerbated by infections.127–129 These clinical overlaps highlight the importance of distinguishing based on the immunological nature.

In summary, clinical and epidemiological evidence strongly supports the existence of an immunological contradiction between AD and PsO. The low comorbidity rate and negative genetic correlation between them underscore their distinct innate immune predispositions, whereas the rare comorbid or alternating cases vividly illustrate the delicate equilibrium and reciprocal restraint between the two immune pathways within an individual. These findings are of significant clinical relevance, particularly for therapeutic choices and risk surveillance in the context of biologic treatments.

Antagonistic Responses from a Therapeutic Perspective

AD and psoriasis are two immune-pathologically opposing diseases. Clinical studies have found that highly effective biologics targeting a specific immune pathway may triggering or exacerbating another disease. This phenomenon is referred to as antagonistic responses in therapeutics.

Suppression of the Th2 Pathway and Induction of Psoriasis

Currently, most therapeutic approaches for atopic dermatitis primarily focus on suppressing the Th2-mediated immune response. However, Th2 cytokines such as IL-4 exert a physiological inhibitory effect on the Th1 and Th17 pathways. When the Th2 axis is strongly suppressed, this inhibition is lifted, potentially resulting in the reactivation or amplification of the previously restrained Th1/Th17 responses. This phenomenon, known as immune drift, represents a characteristic immune rebalancing side effect observed during biologic therapy.130–132

We summarized all currently reported cases in which biologic therapy for atopic dermatitis led to psoriasis-like manifestations, as shown in Table 1.

Table 1 The Contradictory Reactions That Occur During the Treatment of AD

Inhibition of the Th17/Th1 Pathway and the Induction of Atopic Dermatitis

Similar phenomena have also been observed in psoriasis treatment, mirroring those seen in atopic dermatitis. Increasing clinical evidence indicates that inhibition of the Th17/Th1 pathway may disrupt immune homeostasis, resulting in a predominance of Th2 responses that can induce new-onset AD or exacerbate previously controlled disease—a typical paradoxical reaction.

We summarized all currently reported cases in which biologic therapy for psoriasis resulted in atopic dermatitis–like manifestations, as presented in Table 2.

Table 2 The Contradictory Reactions That Occur During the Treatment of Psoriasis

JAK Inhibitors, as Intermediate Modulators, May Simultaneously Act on Th1/Th2

The JAK–STAT signaling pathway, a central hub mediating the actions of numerous cytokines, plays a pivotal role in regulating multiple immune axes, including Th1, Th2, and Th17 responses. JAK inhibitors can block the activation of downstream STAT proteins through targeting JAK family proteins (such as JAK1, JAK2, JAK3, TYK2). This mechanism of action endows it with the ability to simultaneously modulate both Th1 and Th2 immune responses, making it a promising option for correcting immune imbalances induced by targeted therapies, such as paradoxical reactions (Table 3).

Table 3 The Efficacy of JAK Inhibitors in Managing Paradoxical Reactions Induced by Biologic Therapies

In summary, JAK inhibitors act on the common downstream of multiple cytokines signaling pathways through their unique mechanism of action. By doing so, they serve as versatile immune modulators capable of rebalancing dysregulated immune responses. Clinical practice has demonstrated that JAK inhibitors offer an effective, rapid, and relatively safe approach for managing paradoxical reactions induced by biologic therapies that disrupt immune balance.

Molecular and Transcriptomics Evidence

The specific mechanisms underlying these paradoxical reactions remain poorly understood by clinicians. Single-cell RNA sequencing offers a powerful approach to decoding the immunological signatures of inflammatory diseases. By integrating skin transcriptomic data with clinical phenotypes, it becomes possible to delineate local inflammatory processes and broader immune interactions in greater detail. Therefore, we have summarized the current genomic and transcriptomic studies investigating paradoxical reactions to provide an updated overview of this emerging field.

A recently published single-cell sequencing study compared PBMCs from psoriasis patients who developed dermatitis after biologic therapy with those from psoriasis patients without such adverse reactions. Compared with the psoriasis control group, the case clusters exhibited a global upregulation of TNF and IFN-γ expression. In addition, IFN-α expression was increased within both B-cell and T-cell populations, whereas Th2 cytokine signaling was not enriched. These findings suggest that the pathogenesis of paradoxical eczema does not simply represent an inflammatory shift from Th1/Th17 to Th2 polarization.70

Another study conducted proteomic and genomic analyses comparing serum samples from psoriasis patients who developed dermatitis after biologic therapy with those from psoriasis patients without such changes. The study found that STAMBP expression was reduced in cases of paradoxical eczema and demonstrated that, at the gene set level, the systemic inflammatory profile of patients with paradoxical reactions tended to resemble that of AD.175

Although genomic data on paradoxical reactions remain limited at present, we believe that this emerging approach will help elucidate the underlying mechanisms in the future. As more sequencing studies are conducted, the molecular landscape of these reactions will become clearer, paving the way for targeted experimental validation and mechanistic exploration.

Future Research Directions and Clinical Implications
From Genetic Background to Immune Endotype

Although PsO and AD are generally considered mutually exclusive diseases in most populations, there are still a few clinical cases of patients suffering from both conditions simultaneously. This phenomenon suggests that the two may share certain susceptible genetic backgrounds. Studies indicate that the HLA gene, FLG mutations, and GWAS may play key roles in the pathogenesis of both diseases.176,177 Especially in the immune genetic factors that regulate the balance between Th17 and Th2 responses.178–180 In addition, FLG mutations have also been identified in some psoriasis patients, suggesting a shared genetic basis underlying the barrier dysfunction observed in both diseases.181

Emerging Therapeutic Target: Dual-Pathway Regulation

Emerging therapies are moving from single-pathway blockade toward dual or upstream regulation to more precisely modulate the shared immune network of PsO and AD. TSLP inhibitors can inhibit both Th2 and IL-23/IL-17 pathways, proving particularly effective for severe mixed-type patients.182 IL-36R inhibitors, show potential in the AD subtype with IL-36 signaling activation.183,184 JAK inhibitors have become representatives of dual-pathway regulation through blocking multiple inflammatory pathways. In the future, targeting skin-resident memory T cells (TRM) and innate lymphoid cells (ILCs) may offer new opportunities for achieving sustained remission or even cure.185–187

From the Perspective of Precision Medicine: Treatment Selection Guided by the Patient’s Immune Phenotype

Future dermatologic therapies are expected to move beyond the traditional disease name–based approach toward precision models defined by immune subtyping. At the core of this transition is biomarker-driven, stratified treatment tailored to individual immune profiles.188 Patients can be stratified into distinct immune endotypes by analyzing cytokine profiles in serum, skin transcriptomic signatures, or immune cell subset compositions. For instance, in patients with AD who show inadequate responses to Th2-targeted therapies, improved outcomes may be achieved with JAK inhibitors or combination approaches.112 This immune phenotype-guided treatment strategy will maximize treatment efficacy and promote the development of precision medicine.

The Significance of Basic Research in Skin Immunology and Clinical Management

The research of the contradictory phenomenon is significant for skin immunology and clinical management. It is a challenges for the traditional Th1/Th17 and Th2 dichotomy model, highlighting the dynamics and plasticity of the skin immune system,189 It provides a new perspective for understanding immune tolerance and immune memory. In clinical practice, this understanding can drive innovation in diagnostic and treatment strategies. It remind doctors to pay attention to atypical dermatitis presentations and make individually adjust treatment plans.188 Collectively, this research expands current knowledge in skin immunology and fosters the translation of mechanistic discoveries into precision-based clinical applications.

Consent for Publication

All authors consented to publication of the manuscript in its current form.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

YZ is funded by the Chinese Scholarship Council (YZ: CSC no. 202308210137). YZ is funded by the Jinzhou Medical University Teacher Reserve Program.

Disclosure

The authors declare that they have no competing interests.

References

1. David Boothe W, Tarbox JA, Tarbox MB. Atopic dermatitis: pathophysiology. Adv Exp Med Biol. 2017;1027:21–37.

2. Boehncke WH, Schön MP. Psoriasis. Lancet. 2015;386(9997):983–994. doi:10.1016/S0140-6736(14)61909-7

3. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. Int J Mol Sci. 2019;20(6):1475. doi:10.3390/ijms20061475

4. Kamiya K, Kishimoto M, Sugai J, Komine M, Ohtsuki M. Risk factors for the development of psoriasis. Int J Mol Sci. 2019;20(18):4347. doi:10.3390/ijms20184347

5. Griffiths CEM, Armstrong AW, Gudjonsson JE, Barker JNWN. Psoriasis. Lancet. 2021;397(10281):1301–1315. doi:10.1016/S0140-6736(20)32549-6

6. Mrowietz U, Lauffer F, Sondermann W, Gerdes S, Sewerin P. Psoriasis as a systemic disease. Dtsch Arztebl Int. 2024. doi:10.3238/arztebl.m2024.0064

7. Perry M. Psoriasis: an overview. Br J Nurs. 2024;33(15):686–692. doi:10.12968/bjon.2024.0112

8. Lwin SM, Clements CM, Mukherjee N. Curing psoriasis. J Invest Dermatol. 2024;145(1):144. doi:10.1016/j.jid.2024.04.027

9. Morizane S, Yamasaki K, Mühleisen B, et al. Cathelicidin antimicrobial peptide LL-37 in psoriasis enables keratinocyte reactivity against TLR9 ligands. J Invest Dermatol. 2012;132(1):135–143. doi:10.1038/jid.2011.259

10. Honma M, Hayashi K. Psoriasis: recent progress in molecular‐targeted therapies. J Dermatol. 2021;48(6):761–777. doi:10.1111/1346-8138.15727

11. Sugumaran D, Yong ACH, Stanslas J. Advances in psoriasis research: from pathogenesis to therapeutics. Life Sci. 2024;355:122991. doi:10.1016/j.lfs.2024.122991

12. Georgescu SR, Tampa M, Caruntu C, et al. Advances in understanding the immunological pathways in psoriasis. IJMS. 2019;20(3):739. doi:10.3390/ijms20030739

13. Conrad C, Gilliet M. Psoriasis: from pathogenesis to targeted therapies. Clin Rev Allergy Immunol. 2018;54(1):102–113. doi:10.1007/s12016-018-8668-1

14. Singh R, Koppu S, Perche PO, Feldman SR. The cytokine mediated molecular pathophysiology of psoriasis and its clinical implications. Int J Mol Sci. 2021;22(23):12793. doi:10.3390/ijms222312793

15. Krzysztofik M, Brzewski P, Kulbat A, Masajada M, Richter K, Wysocki WM. The IL-23/Th17 pathway inhibitors in the treatment of psoriasis and the risk of skin malignancies: a review. Adv Dermatol Allergol. 2024;41(6):552–559. doi:10.5114/ada.2024.143428

16. Petit RG, Cano A, Ortiz A et al. Psoriasis: From Pathogenesis to Pharmacological and Nano-Technological-Based Therapeutics. Int J Mol Sci. 2021;22(9):4893. doi:10.3390/ijms22094983

17. Guo J, Zhang H, Lin W, Lu L, Su J, Chen X. Signaling pathways and targeted therapies for psoriasis. Signal Transduct Target Ther. 2023;8(1):437. doi:10.1038/s41392-023-01655-6

18. Sánchez-Rodríguez G, Puig L. pathogenic role of IL-17 and therapeutic targeting of IL-17F in psoriatic arthritis and spondyloarthropathies. IJMS. 2023;24(12):10305. doi:10.3390/ijms241210305

19. Schinocca C, Rizzo C, Fasano S, et al. Role of the IL-23/IL-17 pathway in rheumatic diseases: an overview. Front Immunol. 2021;12:637829. doi:10.3389/fimmu.2021.637829

20. Bugaut H, Aractingi S. Major role of the IL17/23 axis in psoriasis supports the development of new targeted therapies. Front Immunol. 2021;12:621956. doi:10.3389/fimmu.2021.621956

21. Shi ZR, Mabuchi T, Riutta SJ, et al. The chemokine, CCL20, and its receptor, CCR6, in the pathogenesis and treatment of psoriasis and psoriatic arthritis. J Psoriasis Psoriatic Arthritis. 2023;8(3):107–117. doi:10.1177/24755303231159106

22. Meitei HT, Jadhav N, Lal G. CCR6-CCL20 axis as a therapeutic target for autoimmune diseases. Autoimmunity Rev. 2021;20(7):102846. doi:10.1016/j.autrev.2021.102846

23. Gómez-Melero S, Caballero-Villarraso J. CCR6 as a potential target for therapeutic antibodies for the treatment of inflammatory diseases. Antibodies. 2023;12(2):30. doi:10.3390/antib12020030

24. Francis L, Capon F, Smith CH, Haniffa M, Mahil SK. Inflammatory memory in psoriasis: from remission to recurrence. J Allergy Clin Immunol. 2024;154(1):42–50. doi:10.1016/j.jaci.2024.05.008

25. Lowes MA, Suárez-Fariñas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–255. doi:10.1146/annurev-immunol-032713-120225

26. Gaffen SL, Jain R, Garg AV, Cua DJ. IL-23-IL-17 immune axis: discovery, mechanistic understanding, and clinical testing. Nat Rev Immunol. 2014;14(9):585–600. doi:10.1038/nri3707

27. Krueger JG, Fretzin S, Suárez-Fariñas M, et al. IL-17A is essential for cell activation and inflammatory gene circuits in psoriasis. J Allergy Clin Immunol. 2012;130(1):145–54.e9. doi:10.1016/j.jaci.2012.04.024

28. Magliocco M, Krueger JG, Abello MV, et al. Psoriasis plaques multiple proinflammatory pathways in TNF Inhibition rapidly down-regulates.

29. Wolk K, Witte E, Wallace E, et al. IL‐22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol. 2006;36(5):1309–1323. doi:10.1002/eji.200535503

30. Lopez DV, Kongsbak‐Wismann M. Role of IL‐22 in homeostasis and diseases of the skin. APMIS. 2022;130(6):314–322. doi:10.1111/apm.13221

31. Guilloteau K, Paris I, Pedretti N, et al. Skin inflammation induced by the synergistic action of IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α recapitulates some features of psoriasis. J Immunol. 2010;184(9):5263–5270. doi:10.4049/jimmunol.0902464

32. Gao J, Chen F, Fang H, Mi J, Qi Q, Yang M. Daphnetin inhibits proliferation and inflammatory response in human HaCaT keratinocytes and ameliorates imiquimod-induced psoriasis-like skin lesion in mice. Biol Res. 2020;53:48. doi:10.1186/s40659-020-00316-0

33. Hu P, Wang M, Gao H, et al. The role of helper T cells in psoriasis. Front Immunol. 12:788940. PMID: 34975883; PMCID: PMC8714744. doi:10.3389/fimmu.2021.788940

34. Akhter S, Tasnim FM, Islam MN, et al. Role of Th17 and IL-17 cytokines on inflammatory and auto-immune diseases. CPD. 2023;29(26):2078–2090. doi:10.2174/1381612829666230904150808

35. Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129(6):1339–1350. doi:10.1038/jid.2009.59

36. Hawkes JE, Chan TC, Krueger JG. Psoriasis pathogenesis and the development of novel, targeted immune therapies. J Allergy Clin Immunol. 2017;140(3):645–653. doi:10.1016/j.jaci.2017.07.004

37. Guttman-Yassky E, Renert-Yuval Y, Brunner PM. Atopic dermatitis. Lancet. 2025;405(10478):583–596. doi:10.1016/S0140-6736(24)02519-4

38. Afshari M, Kolackova M, Rosecka M, Čelakovská J, Krejsek J. Unraveling the skin; a comprehensive review of atopic dermatitis, current understanding, and approaches. Front Immunol. 2024;15:1361005. doi:10.3389/fimmu.2024.1361005

39. Chen WY, Chen SC, Hsu SY, et al. Annoying psoriasis and atopic dermatitis: a narrative review. IJMS. 2022;23(9):4898. doi:10.3390/ijms23094898

40. Kamata Y, Tominaga M, Takamori K. Mechanisms of itch in atopic dermatitis. Juntendo Med J. 2025;71(1):43–50. doi:10.14789/ejmj.JMJ24-0036-R

41. Goh MS, Yun JS, Su JC. Management of atopic dermatitis: a narrative review. Med j Aust. 2022;216(11):587–593. doi:10.5694/mja2.51560

42. Gomułka K, Tota M, Laska J, Gojny K, Sędek Ł. Serum concentration of IL-5 Receptor (IL-5R) and associations with disease severity in patients with chronic spontaneous urticaria (CSU) and atopic dermatitis (AD). Int J Mol Sci. 2024;25(14):7598. doi:10.3390/ijms25147598

43. Tsuji G, Yamamura K, Kawamura K, Kido-Nakahara M, Ito T, Nakahara T. Novel therapeutic targets for the treatment of atopic dermatitis. Biomedicines. 2023;11(5):1303. doi:10.3390/biomedicines11051303

44. Missaoui S, Gaadi A, Oussama K, Aadam Z, Bousfiha AA, Hali F. The primary mechanisms underlying atopic dermatitis Les principaux mécanismes sous-jacents à la dermatite atopique. Tunis Med. 2025;103:65–72.

45. Facheris P, Jeffery J, Del Duca E, Guttman-Yassky E. The translational revolution in atopic dermatitis: the paradigm shift from pathogenesis to treatment. Cell Mol Immunol. 2023;20(5):448–474. doi:10.1038/s41423-023-00992-4

46. Ogulur I, Mitamura Y, Yazici D, et al. Type 2 immunity in allergic diseases. Cell Mol Immunol. 2025;22(3):211–242. doi:10.1038/s41423-025-01261-2

47. Szymański Ł, Cios A, Ciepielak M, Stankiewicz W. Cytokines and apoptosis in atopic dermatitis. pdia. Adv Dermatol Allergol. 2021;38(1):1–13.

48. Laska J, Tota M, Łacwik J, Sędek Ł, Gomułka K. IL-22 in atopic dermatitis. Cells. 2024;13(16):1398. doi:10.3390/cells13161398

49. Li H, Zhang Z, Zhang H, Guo Y, Yao Z. Update on the pathogenesis and therapy of atopic dermatitis. Clin Rev Allerg Immunol. 2021;61(3):324–338. doi:10.1007/s12016-021-08880-3

50. Torres T, Ferreira EO, Gonçalo M, Mendes-Bastos P, Selores M, Filipe P. Update on atopic dermatitis. Acta Med Port. 2019;32(9):606–613. doi:10.20344/amp.11963

51. Commonality of the IL-4/IL-13 pathway in atopic diseases [Internet]. 2025. Available from: https://jlylos.jlss.vip/%E5%A4%96%E5%88%8A%E5%85%A8%E6%96%87%2F2017%2F1744-666X%2F13%2F5%2F31037238.pdf?X-Tos-Algorithm=TOS4-HMAC-SHA256&X-Tos-Credential=AKLTNGZhYzU5ODhjOGY0NGNhMjhkMDYyY2UzOTM1Y2NhYTg%2F20251008%2Fcn-guangzhou%2Ftos%2Frequest&X-Tos-Expires=3600&X-Tos-Date=20251008T115914Z&X-Tos-Signature=188b6ec7dc92a9ace28a8c16b311458bbca6f6f9f498c414ae565dc69ca21aaa&X-Tos-SignedHeaders=host. Accessed December 25, 2025.

52. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations. Annu Rev Immunol. 2010;28:445–489. doi:10.1146/annurev-immunol-030409-101212

53. Bao L, Zhang H, Chan LS. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT. 2013;2(3):e24137. doi:10.4161/jkst.24137

54. Beck LA, Graham NM, Ming JE, et al. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. n Engl J Med. 2014;(2):130–139. doi:10.1056/NEJMoa1314768

55. Santamaria-Babi L. Atopic dermatitis pathogenesis: lessons from immunology. Dermatol Pract Concept. 2022;12(1):e2022152. doi:10.5826/dpc.1201a152

56. Schuler CF, Billi AC, Maverakis E, Tsoi LC, Gudjonsson JE. Novel insights into atopic dermatitis. J Allergy Clin Immunol. 2023;151(5):1145–1154. doi:10.1016/j.jaci.2022.10.023

57. Cevikbas F, Steinhoff M. IL-33: a novel danger signal system in atopic dermatitis. J Invest Dermatol. 2012;132(5):1326–1329. doi:10.1038/jid.2012.66

58. Savva M, Papadopoulos NG, Gregoriou S, et al. Recent advancements in the atopic dermatitis mechanism. Front Biosci. 2024;29(2):84. doi:10.31083/j.fbl2902084

59. Tsakok T, Woolf R, Smith CH, Weidinger S, Flohr C. Atopic dermatitis: the skin barrier and beyond. Br J Dermatol. 2019;180(3):464–474. doi:10.1111/bjd.16934

60. Tominaga M, Takamori K. Peripheral itch sensitization in atopic dermatitis. Allergol Int. 2022;71(3):265–277. doi:10.1016/j.alit.2022.04.003

61. Ständer S, Luger T, Kim B, et al. Cutaneous components leading to pruritus, pain, and neurosensitivity in atopic dermatitis: a narrative review. Dermatol Ther. 2024;14(1):45–57. doi:10.1007/s13555-023-01081-0

62. Escalante A, Serra-Baldrich E. Pathogenic mechanisms underlying itch in atopic dermatitis: the emerging role of neuroimmune interactions. Eur J Dermatol. 2023;33(4):343–349. doi:10.1684/ejd.2023.4514

63. Cornelissen C, Marquardt Y, Czaja K. IL-31 regulates differentiation and filaggrin expression in human organotypic skin models. J Allergy Clin Immunol. 2012;129(2). doi:10.1016/j.jaci.2011.10.042

64. Yue C, Zhou H, Wang X, et al. Atopic dermatitis: pathogenesis and therapeutic intervention. MedComm. 2024;5(12):e70029. doi:10.1002/mco2.70029

65. Armario-Hita J, Galán-Gutiérrez M, Dodero-Anillo J, Carrascosa J, Ruiz-Villaverde R. Updated review on treatment of atopic dermatitis. J Investig Allergol Clin. 2023;33(3):158–167. doi:10.18176/jiaci.0906

66. Tokura Y, Hayano S. Subtypes of atopic dermatitis: from phenotype to endotype. Allergol Int. 2022;71(1):14–24. doi:10.1016/j.alit.2021.07.003

67. Belpaire A, van Geel N, Speeckaert R. From IL-17 to IFN-g in inflammatory skin disorders: is transdifferentiation a potential treatment target? Front Immunol. 2022;13. doi:10.3389/fimmu.2022.932265

68. Mesas‐Fernández A, Bodner E, Hilke FJ, Meier K, Ghoreschi K, Solimani F. Interleukin‐21 in autoimmune and inflammatory skin diseases. Eur J Immunol. 2023;53(4):2250075. doi:10.1002/eji.202250075

69. Tsai YC, Tsai TF. Overlapping features of psoriasis and atopic dermatitis: from genetics to immunopathogenesis to phenotypes. IJMS. 2022;23(10):5518. doi:10.3390/ijms23105518

70. Al-Janabi A, Martin P, Simpson C, et al. Blood single-cell transcriptomic and proteomic signatures of paradoxical eczema in patients with psoriasis treated with biologics. J Invest Dermatol. 2025;145(10):2535–2548. doi:10.1016/j.jid.2025.02.153

71. Kanoh H, Ishitsuka A, Fujine E, et al. IFN- γ reduces epidermal barrier function by affecting fatty acid composition of ceramide in a mouse atopic dermatitis model. J Immunol Res. 2019;2019:1–10. doi:10.1155/2019/3030268

72. Agnello D, Lankford CSR, Bream J, et al. Cytokines and transcription factors that regulate t helper cell differentiation: new players and new insights. J Clin Immunol. 2003;23(3):147–161. doi:10.1023/A:1023381027062

73. Finotto S, Glimcher L. T cell directives for transcriptional regulation in asthma. Springer Semin Immunopathol. 2004;25(3–4):281–294. doi:10.1007/s00281-003-0143-1

74. Lee JW, Wu Q, Jang YP, Choung SY. Pinus densiflora bark extract ameliorates 2,4‐dinitrochlorobenzene‐induced atopic dermatitis in NC/Nga mice by regulating Th1/Th2 balance and skin barrier function. Phytother Res. 2018;32(6):1135–1143. doi:10.1002/ptr.6061

75. Yamada K, Matsushita K, Wang J, Kanekura T. Topical glucose induces claudin-1 and filaggrin expression in a mouse model of atopic dermatitis and in keratinocyte culture, exerting anti-inflammatory effects by repairing skin barrier function. Acta Derm Venerol. 2018;98(1):19–25. doi:10.2340/00015555-2807

76. Choi JK, Kim SH. Inhibitory effect of galangin on atopic dermatitis-like skin lesions. Food Chem Toxicol. 2014;68:135–141. doi:10.1016/j.fct.2014.03.021

77. Krupka-Olek M, Bozek A, Drewienkowska M, Dryjka K, Medrala A. Immunological differences between atopic dermatitis, psoriasis, and their combination in adult patients. Allergologia et Immunopathologia. 2022;50(4):143–146. doi:10.15586/aei.v50i4.557

78. Ylikoski E, Lund R, Kyläniemi M, et al. IL-12 up-regulates T-bet independently of IFN-c in human CD4+ T cells. Eur J Immunol. 2005;35(11):3297–3306. doi:10.1002/eji.200526101

79. Bullens DMA, Kasran A, Thielemans K, Bakkus M, Ceuppens JL. CD40L‐induced IL‐12 production is further enhanced by the Th2 cytokines IL‐4 and IL‐13. Scand J Immunol. 2001;53(5):455–463. doi:10.1046/j.1365-3083.2001.00900.x

80. Dieu-Nosjean MC, Massacrier C, Vanbervliet B, Fridman WH, Caux C. IL-10 induces CCR6 expression during langerhans cell development while IL-4 and IFN-γ suppress it. J Immunol. 2001;167(10):5594–5602. doi:10.4049/jimmunol.167.10.5594

81. Hatano Y, Elias PM. “Outside-to-inside”, “inside-to-outside”, and “intrinsic” endogenous pathogenic mechanisms in atopic dermatitis: keratinocytes as the key functional cells involved in both permeability barrier dysfunction and immunological alterations. Front Immunol. 2023;14. doi:10.3389/fimmu.2023.1239251

82. Harb H, Chatila TA. Mechanisms of Dupilumab. Clin Exp Allergy. 2020;50(1):5–14. doi:10.1111/cea.13491

83. Furue M. Regulation of skin barrier function via competition between AHR axis versus IL-13/IL-4‒JAK‒STAT6/STAT3 axis: pathogenic and therapeutic implications in atopic dermatitis. JCM. 2020;9(11):3741. doi:10.3390/jcm9113741

84. Dardalhon V, Awasthi A, Kwon H, et al. Interleukin 4 inhibits TGF-β-induced-Foxp3+T cells and generates, in combination with TGF-β, Foxp3− effector T cells that produce interleukins 9 and 10. 2010.

85. Niehues T, Hardenberg SV, Velleuer E. Rapid identification of primary atopic disorders (PAD) by a clinical landmarkguided, upfront use of genomic sequencing. ALS. 2024;8(01):304–323. doi:10.5414/ALX02520E

86. Schramm G, Suwandi A, Galeev A, et al. Schistosome eggs impair protective Th1/Th17 immune responses against salmonella infection. Front Immunol. 2018;9:2614. doi:10.3389/fimmu.2018.02614

87. He R, Kim HY, Yoon J, et al. Exaggerated IL-17 response to epicutaneous sensitization mediates airway inflammation in the absence of IL-4 and IL-13. J Allergy Clin Immunol. 2009;124(4):761–770.e1. doi:10.1016/j.jaci.2009.07.040

88. Ghoreschi K, Laurence A, Yang XP, Hirahara K, O’Shea JJ. T helper 17 cell heterogeneity and pathogenicity in autoimmune disease. Trend Immunol. 2011;32(9):395–401. doi:10.1016/j.it.2011.06.007

89. Guttman-Yassky E, Suárez-Fariñas M, Chiricozzi A, et al. Broad defects in epidermal cornification in atopic dermatitis identified through genomic analysis. J Allergy Clin Immunol. 2009;124(6):1235–1244.e58. doi:10.1016/j.jaci.2009.09.031

90. Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol. 2009;129(9):2175–2183. doi:10.1038/jid.2009.65

91. Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 2006;203(10):2271–2279. doi:10.1084/jem.20061308

92. Albanesi C, Scarponi C, Cavani A, Federici M, Nasorri F, Girolomoni G. Interleukin-17 is produced by both Th1 and Th2 lymphocytes, and modulates interferon-γ- and interleukin-4-induced activation of human keratinocytes. J Invest Dermatol. 2000;115(1):81–87. doi:10.1046/j.1523-1747.2000.00041.x

93. Johnson-Huang LM, McNutt NS, Krueger JG, Lowes MA. Cytokine-producing dendritic cells in the pathogenesis of inflammatory skin diseases. J Clin Immunol. 2009;29(3):247–256. doi:10.1007/s10875-009-9278-8

94. Cabanillas B, Novak N. Atopic dermatitis and filaggrin. Curr Opin Immunol. 2016;42:1–8. doi:10.1016/j.coi.2016.05.002

95. Howell MD, Kim BE, Gao P, et al. Cytokine modulation of atopic dermatitis filaggrin skin expression. J Allergy Clin Immunol. 2009;124(3):R7–12. doi:10.1016/j.jaci.2009.07.012

96. Guttman-Yassky E, Lowes MA, Fuentes-Duculan J, et al. Low expression of the IL-23/Th17 pathway in atopic dermatitis compared to psoriasis. J Immunol. 2008;181(10):7420–7427. doi:10.4049/jimmunol.181.10.7420

97. Albanesi C, Madonna S, Gisondi P, Girolomoni G. The interplay between keratinocytes and immune cells in the pathogenesis of psoriasis. Front Immunol. 2018;9:1549. doi:10.3389/fimmu.2018.01549

98. Noda S, Suárez-Fariñas M, Ungar B, et al. The Asian atopic dermatitis phenotype combines features of atopic dermatitis and psoriasis with increased TH17 polarization. J Allergy Clin Immunol. 2015;136(5):1254–1264. doi:10.1016/j.jaci.2015.08.015

99. Kim BS, Siracusa MC, Saenz SA, et al. TSLP elicits IL-33–independent innate lymphoid cell responses to promote skin inflammation. Sci Transl Med. 2013;5(170). doi:10.1126/scitranslmed.3005374

100. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nat Immunol. 2002;3(7):673–680. doi:10.1038/ni805

101. Divekar R, Kita H. Recent advances in epithelium-derived cytokines (IL-33, IL-25 and TSLP) and allergic inflammation. Curr Opin Allergy Clin Immunol. 2015;15(1):98–103. doi:10.1097/ACI.0000000000000133

102. Simpson EL, Papp KA, Blauvelt A, et al. Efficacy and safety of upadacitinib in patients with moderate to severe atopic dermatitis: analysis of follow-up data from the measure up 1 and measure up 2 randomized clinical trials. JAMA Dermatol. 2022;158(4):404. doi:10.1001/jamadermatol.2022.0029

103. O’Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–328. doi:10.1146/annurev-med-051113-024537

104. Sano S, Chan KS, DiGiovanni J. Impact of Stat3 activation upon skin biology: a dichotomy of its role between homeostasis and diseases. J Dermatological Sci. 2008;50(1):1–14. doi:10.1016/j.jdermsci.2007.05.016

105. Iznardo H, Puig L. IL-1 family cytokines in inflammatory dermatoses: pathogenetic role and potential therapeutic implications. IJMS. 2022;23(16):9479. doi:10.3390/ijms23169479

106. Egeberg A, Skov L, Gislason G, Thyssen J, Mallbris L. Incidence and prevalence of psoriasis in Denmark. Acta Derm Venerol. 2017;97(7):808–812. doi:10.2340/00015555-2672

107. Christophers E, Henseler T. Contrasting disease patterns in psoriasis and atopic dermatitis. Arch Dermatol Res. 1987;279(S1):S48–S51. doi:10.1007/BF00585919

108. Eyerich S, Onken AT, Weidinger S, et al. mutual antagonism of T cells causing psoriasis and atopic eczema. N Engl J Med. 2011;365(3):231–238. doi:10.1056/NEJMoa1104200

109. Zhao D, Wu S, Wang Y, Zheng H, Zhu M. Association between allergic diseases and both psoriasis and psoriatic arthritis: a bidirectional 2-sample mendelian randomization study. Arch Dermatol Res. 2024;316(5):181. doi:10.1007/s00403-024-02978-2

110. Baurecht H, Hotze M, Brand S, et al. Genome-wide comparative analysis of atopic dermatitis and psoriasis gives insight into opposing genetic mechanisms. Am J Hum Genet. 2015;96(1):104–120. doi:10.1016/j.ajhg.2014.12.004

111. Wang Z, Nie H, Fan X, et al. Causal associations between psoriasis and atopic dermatitis: a bidirectional mendelian randomization study. PLoS One. 2025;20(9):e0322602. doi:10.1371/journal.pone.0322602

112. Guttman-Yassky E, Krueger JG. Atopic dermatitis and psoriasis: two different immune diseases or one spectrum? Curr Opin Immunol. 2017;48:68–73. doi:10.1016/j.coi.2017.08.008

113. Radtke MA, Glaeske G, Reich K, Christophers E, Schaefer I, Jacobi A. Epidemiology and comorbidity in children with psoriasis and atopic eczema. Dermatology. 2015;231:35–40.

114. Griffiths CEM, van de Kerkhof P, Czarnecka-Operacz M. Psoriasis and atopic dermatitis. Dermatol Ther. 2017;7(1):31–41. doi:10.1007/s13555-016-0167-9

115. Dai YX, Tai YH, Chang YT, Chen TJ, Chen MH. Bidirectional association between psoriasis and atopic dermatitis: a nationwide population-based cohort study. Dermatology. 2021;237(4):521–527. doi:10.1159/000514581

116. Ma L, Chen X, Aziz MAA, Chen A, Cai T, Chen S. Dual biologic therapy for psoriasis in a patient with atopic dermatitis. PTT. 2025;15:159–161. doi:10.2147/PTT.S516268

117. Parker JJ, Sugarman JL, Silverberg NB, et al. Psoriasiform dermatitis during dupilumab treatment for moderate‐to‐severe atopic dermatitis in children. Pediatr Dermatol. 2021;38(6):1500–1505. doi:10.1111/pde.14820

118. Paolino G, Di Nicola MR, Brianti P, Bianchi VG, Mercuri SR. New onset atopic dermatitis and psoriasis in the same patients under biologic treatments: the role of systemic treatments as a possible trigger. Dermatol Ther. 2022;35(11). doi:10.1111/dth.15814

119. Kouwenhoven TA, Bronckers IM, Van De Kerkhof PC, Kamsteeg M, Seyger MM. Psoriasis dermatitis: an overlap condition of psoriasis and atopic dermatitis in children. J Eur Acad Dermatol Venereol. 2019;33:e74–6.

120. Callea M. Pathophysiology, management and quality of life in atopic dermatitis and psoriasis—a challenge for patients and their families. Children. 2022;9(5):592. doi:10.3390/children9050592

121. Docampo A, Sánchez‐Pujol MJ, Belinchón I, et al. Response to Letter to the editor: ‘Psoriasis dermatitis: an overlap condition of psoriasis and atopic dermatitis in children. Acad Dermatol Venereol. 2019;33. doi:10.1111/jdv.15716

122. Bozek A, Zajac M, Krupka M. Atopic dermatitis and psoriasis as overlapping syndromes. Mediators Inflamm. 2020;2020:7527859. doi:10.1155/2020/7527859

123. Tollefson MM, Crowson CS, McEvoy MT, Kremers HM. Incidence of psoriasis in children: a population-based study. J Am Acad Dermatol. 2010;62(6):979–987. doi:10.1016/j.jaad.2009.07.029

124. Andrade DDBC, Nzundu R, Rodrigues RRD, et al. The epidemiology of atopic dermatitis and psoriasis in the indigenous people of Brazil. Acad Dermatol Venereol. 2024;38(10). doi:10.1111/jdv.19984

125. Patra P, Harrison T, Khoury M. Challenges in diagnosis and treatment of pediatric psoriasis and atopic dermatitis. Arch Dermatol Res. 2025;317(1):393. doi:10.1007/s00403-025-03862-3

126. Beer WE, Smith AE, Kassab JY, Smith PHS, Rowland Payne CME. Concomitance of psoriasis and atopic dermatitis. Dermatology. 1992;184(4):265–270. doi:10.1159/000247564

127. Chalmers RJG, Colman G. The role of streptococcal infection in the initiation of guttate psoriasis. Arch Dermatol. 1992;128:39–42.

128. Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22(5):850–859. doi:10.1101/gr.131029.111

129. Fry L, Baker BS. Triggering psoriasis: the role of infections and medications. Clin Dermatol. 2007;25(6):606–615. doi:10.1016/j.clindermatol.2007.08.015

130. Tsai JH, Tsai TF. A review of dupilumab-induced adverse events to dermatologists and the potential pathogenesis in the treatment of atopic dermatitis. Dermatitis. 2024;35(1):24–42. doi:10.1089/derm.2022.0096

131. Li Y, Liu H, Zhang F. Biologics-induced immunophenotypic cross-switching in patients with psoriasis and atopic dermatitis. Indian J Dermatol. 2023;68(2):186–191. doi:10.4103/ijd.ijd_871_22

132. Johnson MC. Concurrent atopic dermatitis and psoriasis vulgaris: implications for targeted biologic therapy. Cutis. 2022;109(2). doi:10.12788/cutis.0453

133. Liu L, Chen J, Tang K, Li F, Li S, Ding X. Pustular psoriasis in a patient treated with dupilumab for atopic dermatitis: a case report. CCID. 2023;16:2217–2221. doi:10.2147/CCID.S420904

134. Ali K, Wu L, Qiu Y, Li M. Case report: clinical and histopathological characteristics of psoriasiform erythema and de novo IL-17A cytokines expression on lesioned skin in atopic dermatitis children treated with dupilumab. Front Med. 2022;9:932766. doi:10.3389/fmed.2022.932766

135. Jiao Y, Li L, Zhao A, et al. Dupilumab-induced follicular psoriasis and hypercytokinemia. Dermatitis. 2024;35(6):668–670. doi:10.1089/derm.2023.0388

136. Liu Y, Liu L, Zhou H, Chen G, Wen C, Wu R. Pustular psoriasis induced by dupilumab: a case report. JIR. 2024;17:6389–6394. doi:10.2147/JIR.S476297

137. Maiolini VM, Sousa NA, Marsillac PFD, Bressan AL. Alopecia areata-like and psoriasis after dupilumab use for atopic dermatitis. Anais Brasileiros de Dermatologia. 2021;96(5):634–636. doi:10.1016/j.abd.2021.03.005

138. Jia X, Li C, Wu J, Liu Q Pustular psoriasis appearing induced by Dupilumab therapy in a patient with atopic dermatitis. J Drugs Dermatol. 21(3):311–312. PMID: 35254760. doi:10.36849/JDD.6271

139. Cirone KD, Lovegrove FE. Dupilumab-induced psoriasis in a patient with atopic dermatitis successfully treated with Upadacitinib: a case report. SAGE Open MedCase Rep. 2025;13:2050313X251317811. doi:10.1177/2050313X251317811

140. Safa G, Paumier V Psoriasis induced by dupilumab therapy. Clin Exp Dermatol. 2019;44(3):e49–e50. Epub 2019 Jan 17. PMID: 30656722. doi:10.1111/ced.13901

141. D’Ambra I, Babino G, Fulgione E, et al. Psoriasis onset under dupilumab treatment in two patients affected by atopic dermatitis and one patient affected by alopecia areata: clinical and dermoscopic patterns. Dermatol Ther. 2020;33(6). Epub 2020 Sep 8. PMID: 32779323. doi:10.1111/dth.14169

142. Psoriasis onset under dupilumab treatment in two patients affected by atopic dermatitis and one patient affected by alopecia areata: clinical and dermoscopic patterns.

143. Varma A, Levitt J. Dupilumab-induced phenotype switching from atopic dermatitis to psoriasis. JAAD Case Reports. 2020;6(3):217–218. doi:10.1016/j.jdcr.2020.01.012

144. Gori N, Caldarola G, Pirro F, De Simone C, Peris K A case of guttate psoriasis during treatment with dupilumab. Dermatol Ther. 2019;32(5). Epub 2019 Jun 30. PMID: 31215726. doi:10.1111/dth.12998

145. Flanagan KE, Pupo Wiss IM, Pathoulas JT, Walker CJ, Senna MM Dupilumab-induced psoriasis in a patient with atopic dermatitis and alopecia totalis: a case report and literature review. Dermatol Ther. 2022;35(2). Epub 2021 Dec 17. PMID: 34877768. doi:10.1111/dth.15255

146. Fan J, Zhang L, Lu Y, Zhou P A case of dupilumab-induced reverse psoriasis in a patient with atopic dermatitis. Dermatol Ther. 2022;35(4). Epub 2022 Feb 9. PMID: 35094480. doi:10.1111/dth.15345

147. A case of dupilumab‐induced reverse psoriasis in a patient with atopic dermatitis.

148. Park JW, Park SJ, Koh YG, Park KY, Seo SJ. Dupilumab-induced palmoplantar psoriasis: is it true psoriasis? Dermatitis. 2022;33(3):e33–e35. doi:10.1097/DER.0000000000000824

149. Colonna C, Bortoluzzi P, Cavalli R. Dupilumab treatment for severe atopic dermatitis in children and SARS‐CoV‐2 infection: a combination of triggers for psoriasis. J Eur Acad Dermatol Venereol. 2023;37.

150. Dupilumab‐induced psoriasis in a patient with atopic dermatitis and alopecia totalis: a case report and literature review.

151. Trave I, Salvi I, Burlando M, Cozzani E, Parodi A. “De Novo” psoriasis and relapse of psoriasis induced by dupilumab: three new cases and review of the literature. J Clin Med. 2023;12(19):6291. doi:10.3390/jcm12196291

152. Dang N, Zheng H, Ren Y. A pediatric case of dupilumab-induced pustular psoriasis. J Dermatolog Treat. 2024;35(1). doi:10.1080/09546634.2024.2333016

153. Quattrini L, Caldarola G, Falco GM, Pinto LM, Peris K. Successful treatment with tralokinumab in patients with atopic dermatitis and dupilumab‐induced psoriasis. Acad Dermatol Venereol. 2023;37(12). doi:10.1111/jdv.19351

154. Balakirski G, Burmann SN, Hofmann SC, Kreuter A. Paradoxical tralokinumab-induced psoriasis in a patient with atopic dermatitis. J Dermatological Treat. 2023;34(1):2258240. doi:10.1080/09546634.2023.2258240

155. Chiei Gallo A, Tavoletti G, Termini D, Marzano AV, Ferrucci SM. Tralokinumab‐induced psoriasis relapse in a patient with atopic dermatitis. Int J Dermatol. 2025;64(3):580–581. doi:10.1111/ijd.17397

156. Speeckaert R, Belpaire A, Lambert J, Speeckaert M, Van Geel N. Th pathways in immune-mediated skin disorders: a guide for strategic treatment decisions. Immune Netw. 2024;24(5):e33. doi:10.4110/in.2024.24.e33

157. Ma X, Li T, Han G. Two cases of immune drift phenomena caused by biologic agents for treating psoriasis and atopic dermatitis. Clin Cosmet Invest Dermatol. 2023;16:3521–3525. doi:10.2147/CCID.S445468

158. Galluzzo M, D’Adamio S, Massaro A, Piccolo A, Bianchi L, Talamonti M. Spotlight on brodalumab in the treatment of plaque psoriasis: the evidence to date. Clin Cosmet Invest Dermatol. 2019;12:311–321. doi:10.2147/CCID.S165605

159. Sulich-Moore C, Altman D. Phenotypic shift during treatment of plaque psoriasis with ixekizumab. J Clin Aesthet Dermatol. 2024;17(11):32–33.

160. Mendes Roncada EV, Brambilla VR, Freitas Filitto B, Genta MP, Morgado De Abreu M. Atopic dermatitis as a paradoxical effect of secukinumab for the treatment of psoriasis. Case Rep Dermatol. 2021;13(2):336–339. doi:10.1159/000513467

161. Abe F, Mitsuyama S, Nagao E, Kimura M, Higuchi T. Atopic dermatitis-like eruption induced by two different biologics in a patient with psoriatic arthritis. Acta Derm Venereol. 2019;99(13):1291–1292. doi:10.2340/00015555-3321

162. Burlando M, Cozzani E, Russo R, Parodi A. Atopic-like dermatitis after secukinumab injection: a case report. Dermatol Ther. 2019;32(1):e12751. doi:10.1111/dth.12751

163. Caldarola G, Pirro F, Di Stefani A. Clinical and histopathological characterization of eczematous eruptions occurring in course of anti IL-17 treatment: a case series and review of the literature. Exp Opin Biol Ther. 2020;20(6):665–672. doi:10.1080/14712598.2020.1727439

164. Li Q, Li X, Li W, et al. Development of IL‐17A inhibitor‐induced atopic dermatitis‐like rash in psoriasis patients: insights into immune shift. Exp Dermatol. 2024;33(1):e14958. doi:10.1111/exd.14958

165. Gambardella A, Licata G, De Rosa A, et al. Concurrent atopic dermatitis and psoriasis successfully treated with brodalumab. Dermatitis. 2021;32(1S):e86–8. doi:10.1097/DER.0000000000000645

166. Fu J, Lu J, Wu W, Wang P. Successful treatment of ixekizumab-induced paradoxical eczematous reaction with JAK inhibitors: a case report. Clin Cosmetic Invest Dermatol. 2025;Volume 18:2133–2139. doi:10.2147/CCID.S541725

167. Weiss L, Marbet CP, Branca LB, Mühleisen B, Navarini A. Janus kinase inhibitors as successful treatment alternative in dupilumab-induced psoriasis. Case Rep Dermatol. 2024;16(1):144–148. doi:10.1159/000539124

168. Salvi I, Parodi A, Cozzani E, Burlando M. Case report: psoriasiform eczema with immune-mediated comorbidities treated with upadacitinib. Front Immunol. 2024;15. doi:10.3389/fimmu.2024.1432233

169. Luo N, Zhong L, Luo N, Wang Q, Li T, Hao P. Tofacitinib for the treatment of psoriasiform dermatitis caused by IL-17 inhibitors: a case report. J Inflamm Res. 2023: 2167–72.

170. Tsunoda M, Adachi T, Nakajima Y, Yatomi Y, Shimizu T, Nakasute K. Successful treatment of the dupilumab-induced psoriatic dermatitis/arthritis and atopic dermatitis with a JAK inhibitor: a case report and literature review. J Allergy Clin Immunol. 2024;3(4):100323. doi:10.1016/j.jacig.2024.100323

171. Patruno C, Fabbrocini G, De Lucia M, Picone V, Genco L, Napolitano M. Psoriasiform dermatitis induced by dupilumab successfully treated with upadacitinib. Dermatol Ther. 2022;35(11). doi:10.1111/dth.15788

172. Kamata M, Sun DI, Paller AS. Deciding which patients with atopic dermatitis to prioritize for biologics and Janus kinase inhibitors. J Allergy Clin Immunol Pract. 2025;13(8):1901–1910.e1. doi:10.1016/j.jaip.2025.04.042

173. De Stefano L, Bobbio-Pallavicini F, Montecucco C, Bugatti S. Dupilumab-induced enthesoarthritis and refractory atopic dermatitis successfully treated with baricitinib. Rheumatology. 2022;61(3):e64–6. doi:10.1093/rheumatology/keab771

174. Ferrucci SM, Buffon S, Marzano AV, Maronese CA. Phenotypic switch from atopic dermatitis to psoriasis during treatment with upadacitinib. Clin Experimental Derm. 2022;47(5):986–987. doi:10.1111/ced.15104

175. Al-Janabi A, Martin P, Khan AR, et al. Integrated proteomics and genomics analysis of paradoxical eczema in psoriasis patients treated with biologics. J Allergy Clin Immunol. 2023;152(5):1237–1246. doi:10.1016/j.jaci.2023.07.011

176. Henseler T, Christophers E. Psoriasis of early and late onset: characterization of two types of psoriasis vulgaris. J Am Acad Dermatol. 1985;13(3):450–456. doi:10.1016/S0190-9622(85)70188-0

177. Arakawa A, Siewert K, Stöhr J, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med. 2015;212(13):2203–2212. doi:10.1084/jem.20151093

178. Tsoi LC, Spain SL, Knight J, et al. Identification of fifteen new psoriasis susceptibility loci highlights the role of innate immunity. Nat Genet. 2012;44(12):1341–1348. doi:10.1038/ng.2467

179. Paternoster L, Standl M, Waage J, et al. Multi-ethnic genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis. Nat Genet. 2015;47(12):1449–1456.

180. Bergboer JGM, Tjabringa GS, Kamsteeg M. Psoriasis risk genes of the late cornified envelope-3 group are distinctly expressed compared with genes of other LCE groups. Am J Pathol. 2011;178(4):1470–1477. doi:10.1016/j.ajpath.2010.12.017

181. Brown SJ, McLean WHI, Dd D. One remarkable molecule: filaggrin. J Invest Dermatol. 2012;132(3):751–762. doi:10.1038/jid.2011.393

182. Menzies-Gow A, Corren J, Bourdin A, et al. Tezepelumab in adults and adolescents with severe, uncontrolled asthma. N Engl J Med. 2021;384(19):1800–1809. doi:10.1056/NEJMoa2034975

183. Bachelez H, Choon SE, Marrakchi S, et al. Trial of spesolimab for generalized pustular psoriasis. New Engl J Med. 2021;385(26):2431–2440. doi:10.1056/NEJMoa2111563

184. Patrick GJ, Liu H, Alphonse MP, et al. Epicutaneous Staphylococcus aureus induces IL-36 to enhance IgE production and ensuing allergic disease. J Clin Invest. 2021;131(5). doi:10.1172/JCI143334

185. Emmanuel T, Mistegård J, Bregnhøj A, Johansen C, Iversen L. Tissue-resident memory t cells in skin diseases: a systematic review. IJMS. 2021;22(16):9004. doi:10.3390/ijms22169004

186. Morita H, Moro K, Koyasu S. Innate lymphoid cells in allergic and nonallergic inflammation. J Allergy Clin Immunol. 2016;138(5):1253–1264. doi:10.1016/j.jaci.2016.09.011

187. Niu C, Xie H, Aisa HA. Janus kinase inhibitors: a review of their application in the vitiligo. MRMC. 2021;21(20):3203–3218. doi:10.2174/1389557521666210325120233

188. Bieber T. Atopic dermatitis: an expanding therapeutic pipeline for a complex disease. Nat Rev Drug Discov. 2022;21(1):21–40. doi:10.1038/s41573-021-00266-6

189. Guttman-Yassky E, Lowes MA, Fuentes-Duculan J, et al. Major differences in inflammatory dendritic cells and their products distinguish atopic dermatitis from psoriasis. J Allergy Clin Immunol. 2007;119(5):1210–1217. doi:10.1016/j.jaci.2007.03.006