Your new experience awaits. Try the new design now and help us make it even better

MINI REVIEW article

Front. Immunol., 12 December 2025

Sec. Inflammation

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1698816

Clinical updates of JAK inhibitors in cutaneous granulomatous diseases

Jiaxu Gu,Jiaxu Gu1,2Xinglin He,Xinglin He1,2Bingcheng Lu,Bingcheng Lu1,2Jieyi Wang,,Jieyi Wang1,2,3Kexin Chen,Kexin Chen1,2Qiaofen WangQiaofen Wang4Xingling Jian,Xingling Jian1,2Cong Huang,*Cong Huang1,2*Bo Yu,*Bo Yu1,2*
  • 1Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
  • 2Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
  • 3Department of Dermatology, Shenzhen Xinhua Hospital, Shenzhen, China
  • 4Department of Rheumatology, Hainan Provincial People’s Hospital, Haikou, China

Cutaneous granulomatous diseases, characterized by persistent granuloma formation, often exhibit chronic and relapsing courses that are challenging to manage with conventional therapies. The Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway plays a central role in mediating key cytokines involved in granuloma initiation and maintenance, such as IFN-γ, IL-6, IL-12, and IL-23. JAK inhibitors, by targeting this pathway, offer a promising therapeutic strategy for refractory cases. This review synthesizes current evidence supporting the efficacy of JAK inhibitors—including tofacitinib, ruxolitinib, baricitinib, upadacitinib, and abrocitinib—in conditions such as sarcoidosis, granuloma annulare, granulomatous rosacea, and adverse reactions to cosmetic injectables. Clinical studies and case reports have demonstrated that JAK inhibitors significantly improve lesion outcomes and effectively control symptoms in these conditions, highlighting their potential as targeted treatments. However, further large-scale trials are needed to establish optimal dosing, long-term safety, and predictive biomarkers for personalized therapy.

1 Introduction

Cutaneous granulomatous diseases represent a heterogeneous group of disorders unified by the common pathological feature of granuloma formation (1). Their etiologies are diverse, encompassing infections, autoimmune dysfunctions, foreign body reactions, and idiopathic factors (2). These conditions manifest with complex and varied clinical presentations, often following a chronic and relapsing course that significantly impairs patients’ quality of life (3). Conventional therapeutic strategies, including corticosteroids, immunosuppressants (such as methotrexate), and biological agents, often yield suboptimal outcomes for some patients, characterized by inadequate efficacy, high relapse rates, or significant adverse effects (46). Consequently, there is a pressing clinical need to explore novel and more targeted treatment approaches.

The Janus kinase (JAK) - signal transducer and activator of transcription (STAT) signaling pathway has emerged as a major research focus in immunology and dermatology in recent years. It plays a pivotal role in the signal transduction of numerous cytokines—such as Interferon-gamma (IFN-γ), IL-2, IL-4, IL-6, IL-12, IL-15, and IL-23—which are key mediators driving granuloma formation and sustaining inflammatory processes (7, 8). JAK inhibitors are a class of small-molecule targeted therapeutics that attenuate the signaling of multiple pro-inflammatory cytokines by inhibiting the JAK-STAT pathway, either orally or topically (9). Mounting evidence from recent case reports, case series, and early-phase clinical trials indicates that JAK inhibitors exhibit promising therapeutic potential in managing refractory cutaneous granulomatous diseases, such as sarcoidosis and granuloma annulare (1012).

This review provides a clinically oriented appraisal of the potential of JAK inhibitors in this challenging disease spectrum. Rather than presenting a comprehensive mechanistic treatise, the present review aims to synthesize and critically evaluate the current clinical evidence—comprising case reports, series, and early trials—that supports the use of various JAK inhibitors. It will candidly discuss the significant limitations and biases inherent in this low-level evidence base and, most importantly, provide a forward-looking roadmap for future clinical research and trial design, outlining the necessary steps to translate this preliminary promise into robust, evidence-based clinical practice.

2 JAK-STAT signaling

The JAK-STAT signaling pathway is a central intracellular transduction mechanism mediating the signals of numerous cytokines, growth hormones, and interferons. Pathway activation is initiated by the binding of extracellular cytokines to their cognate receptors, which induces transphosphorylation and activation of the associated Janus kinases (JAKs), including JAK1, JAK2, JAK3, and TYK2 (13). The activated JAKs then phosphorylate the intracellular domains of the receptors, creating docking sites for STAT proteins (STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6) (1416). Upon phosphorylation, STAT proteins form dimers, translocate to the nucleus, and function as transcription factors to regulate the expression of target genes, thereby playing broad roles in immune responses, inflammatory reactions, cell survival, and proliferation (17, 18).

It is noteworthy that the JAK-STAT pathway demonstrates both specificity and redundancy in its function. For instance, IFN-γ signaling primarily involves JAK1 and JAK2 to activate STAT1, whereas IL-6 signaling can engage various combinations such as JAK1, JAK2, and/or TYK2 to ultimately activate STAT3 (19). This characteristic implies that broad inhibition of JAK proteins can simultaneously block the effects of multiple cytokines, providing a therapeutic target for autoimmune diseases (2024). However, selective inhibition of specific JAK subtypes may yield more targeted therapeutic effects and influence the safety profile of such interventions (2527).

3 Metabolic reprogramming in granulomatous inflammation: the interplay of immune signaling and metabolic dysregulation

Emerging evidence underscores that metabolic reprogramming is a fundamental driver of immune cell function in chronic inflammatory diseases. In cutaneous granulomatous disorders, recent findings have revealed a critical axis in which immune signaling directly orchestrates metabolic shifts to sustain the granulomatous response.

A seminal study by Nakamizo et al. has identified the transcription factor c-Maf as a key regulator of macrophage metabolism within granulomas (28). Their work demonstrated that c-Maf, which is upregulated in granulomatous macrophages, acts as a metabolic switch by suppressing glycolysis and mitochondrial respiration while concurrently enhancing the pentose phosphate pathway (PPP). This specific reprogramming is essential for supporting the unique biosynthetic and redox demands of chronic macrophage activation and directly contributes to granuloma formation. The upregulation of the PPP, a branch of glucose metabolism, is particularly crucial. The PPP yields two key products: ribose-5-phosphate and NADPH. Ribose-5-phosphate serves as a precursor for de novo nucleotide synthesis, which is indispensable for the clonal expansion of antigen-specific T cells and the localized proliferation of macrophages within the granuloma structure (29, 30). Concurrently, NADPH plays a dual role: it maintains cellular redox balance by regenerating glutathione, thereby protecting macrophages from oxidative stress-induced apoptosis in the inflammatory microenvironment, and it serves as an essential cofactor for NADPH oxidase (NOX2) to generate reactive oxygen species (ROS) for microbial killing (31, 32). The c-Maf-driven metabolic shift ensures a steady supply of these metabolites, thereby maintaining the viability and pro-inflammatory function of granulomatous macrophages (33, 34).

This metabolic reprogramming is intricately linked to and potentiated by canonical pro-inflammatory signaling. Key cytokines implicated in granuloma pathogenesis, such as IFN-γ and IL-6, signal through their respective JAK-STAT axes (JAK1/JAK2-STAT1 and JAK1/JAK2/TYK2-STAT3) (35, 36). These pathways can further amplify the PPP, creating a feed-forward loop that sustains inflammation. For example, STAT activation can regulate the expression of rate-limiting enzymes in the PPP, such as glucose-6-phosphate dehydrogenase (G6PD) (37, 38). The resulting metabolic state—characterized by PPP dependency—is a hallmark of the classically activated (M1) macrophage phenotype and is critical for sustaining the high ROS output and biosynthetic capacity required for granuloma maintenance (37, 39, 40).

Overall, the pathogenesis of granulomatous diseases is no longer viewed solely through an immunological lens but is now understood as an integrated network of immune and metabolic circuits. The JAK-STAT pathway and transcription factors like c-Maf converge to rewire core metabolic pathways, such as the PPP, to fuel the persistent inflammatory state.

3.1 The relationship between JAK-STAT signaling and skin granuloma formation

Granuloma formation is a complex immune response to persistent stimuli such as infection, foreign bodies, or autoantigens, characterized by the organized aggregation of macrophages and other immune cells (1). In this process, the JAK-STAT signaling pathway plays a central role in the initiation, maintenance, and progression of the disease by mediating the signal transduction of multiple key cytokines (Table 1).

Table 1
www.frontiersin.org

Table 1. Key Cytokine-JAK-STAT axes and their pathogenic roles in cutaneous granuloma formation.

The process is initiated by the release of key cytokines. IFN-γ, by activating the JAK1/JAK2–STAT1 signaling axis in macrophages, drives their polarization toward the pro-inflammatory M1 phenotype (41). This enhances MHC class II expression and antigen presentation capacity, while also upregulating chemokines such as CXCL9 and CXCL10, which recruit T cells to the inflammatory site (4244). Simultaneously, IL-6 signals through the JAK1/JAK2/TYK2–STAT3 pathway, promoting the expression of anti-apoptotic genes like Bcl-2, thereby extending macrophage survival and sustaining the local inflammatory state (45, 46).

These early innate immune responses pave the way for the activation of adaptive immunity. Recruited CD4+ T cells differentiate into effector subsets under the influence of specific cytokines (47). IL-12, signaling via the TYK2/JAK2–STAT4 pathway, directs Th1 cell differentiation, while IL-23, through the TYK2/JAK2–STAT3 pathway, promotes the generation and maintenance of Th17 cells (48, 49). Differentiated Th1 cells secrete large amounts of IFN-γ, establishing a positive feedback loop with macrophages that exacerbates inflammation (50). Th17 cells, in turn, produce IL-17 and IL-22, further amplifying the inflammatory response and recruiting neutrophils (51).

The interactions between macrophages and T cells, mediated by JAK-STAT pathways, collectively establish a self-amplifying inflammatory microenvironment essential for the formation and maintenance of granulomatous structures. In the chronic phase, persistent antigenic stimulation leads to repeated activation of STAT1 and STAT3 (52). This not only sustains elevated expression of pro-inflammatory mediators and chemokines but also upregulates the transcription of pro-fibrotic genes such as TGF-β1 and collagen, ultimately resulting in tissue damage and fibrosis (53, 54). Similarly, STAT3 activation via IL-6 and other gp130-family cytokines promotes the expression of anti-apoptotic genes such as Bcl-2 and survivin, thereby extending the lifespan of macrophages and maintaining granuloma architecture (55, 56). Furthermore, negative regulatory mechanisms of the JAK-STAT pathway, such as suppressors of cytokine signaling (SOCS) proteins, are often dysregulated in chronic granulomatous diseases, preventing the proper termination of inflammatory signaling (57, 58).

4 JAK inhibitors

JAK inhibitors are a class of small-molecule therapeutics designed to selectively inhibit one or more members of the Janus kinase family (JAK1, JAK2, JAK3, TYK2), thereby modulating the JAK-STAT signaling pathway. Therapeutic targeting of JAK-STAT components with inhibitors (e.g., tofacitinib, ruxolitinib) can disrupt this vicious cycle by blocking downstream phosphorylation and nuclear translocation of STATs, thereby reducing cytokine production and immune cell activation, as evidenced in preclinical and clinical studies of sarcoidosis and granuloma annulare. By blocking the phosphorylation and activation of STAT proteins, these agents reduce the downstream effects of multiple cytokines implicated in inflammation, immune activation, and granuloma formation. Their oral and topical formulations offer flexible administration routes, making them suitable for both systemic and localized diseases. Beyond their well-established use in rheumatoid arthritis and myeloproliferative disorders, JAK inhibitors are increasingly applied in dermatology – particularly in conditions driven by dysregulated innate and adaptive immune responses (5961). Their ability to interfere with key pro-inflammatory cytokines such as IFN-γ, IL-4, IL-6, IL-12, IL-13, and IL-23 underpins their therapeutic potential in a range of inflammatory and autoimmune skin diseases (Table 2).

Table 2
www.frontiersin.org

Table 2. JAK Inhibitors Used in dermatological practice.

The efficacy of JAK inhibitors has garnered particular interest in the management of granulomatous skin disorders, including sarcoidosis, granuloma annulare, adverse reactions to cosmetic injectables and granulomatous rosacea. These conditions are characterized by the formation of immune cell aggregates and persistent activation of macrophage and T-cell pathways, largely mediated by dysregulated JAK-STAT signaling. Early preclinical and clinical reports suggest that both topical and oral JAK inhibitors can improve granuloma formation and cutaneous inflammation with a favorable safety profile (Refs). As research advances, these targeted agents hold promise as novel therapeutic strategies for refractory granulomatous dermatoses.

5 Clinical application of JAK inhibitors in cutaneous granulomatous diseases

5.1 Sarcoidosis

Sarcoidosis is a classic non-infectious granulomatous disorder, most commonly manifesting as pulmonary nodules. However, approximately 30% of patients develop cutaneous symptoms, which may present as plaques, nodules, or lupus pernio (62). Sarcoid granulomas are composed of macrophages and T cells that secrete IL-6 and IFN-γ. Within these lesions, STAT1 is primarily activated in macrophages, whereas STAT3 shows activation among lymphocytic aggregates surrounding the granulomas (63). Molecular analyses have demonstrated that oral tofacitinib reduces levels of proinflammatory cytokines in patients with cutaneous sarcoidosis (64). Additionally, there have been reports of successful treatment of cutaneous sarcoidosis, including cases with comorbidities, using ruxolitinib and baricitinib (65, 66). These findings collectively highlight the considerable potential of JAK inhibitors in the management of sarcoidosis.

5.2 Granuloma annulare

In granuloma annulare, a Th1-polarized immune response drives aberrant activation of the JAK-STAT pathway, leading to the development and progression of the granulomatous lesions. Activated T cells secrete large amounts of IFN-γ, which binds to receptors on the surface of macrophages and activates associated JAK1 and JAK2 kinases (67). This in turn phosphorylates and activates the transcription factor STAT1. Activated STAT1 translocates into the nucleus, where it initiates the transcription of a series of pro-inflammatory genes (68). This process directly promotes macrophage activation, proliferation, and M1-polarization, and stimulates the secretion of cytokines such as TNF-α (69). These events ultimately lead to collagen degradation and lymphocytic aggregation, resulting in the formation of the characteristic palisaded granulomatous structure. Clinical evidence indicates that the use of JAK inhibitors, such as upadacitinib and abrocitinib, can effectively block this signaling axis and significantly improve GA skin lesions, thereby providing therapeutic confirmation of the central role of the JAK-STAT pathway in the pathogenesis of granuloma annulare (7072).

5.3 Granulomatous rosacea

Rosacea is a chronic inflammatory disorder whose pathogenesis involves the interaction between genetic and environmental factors, dysregulation of the innate immune system, neurovascular modifications, and their interplay with the skin (73). Among the subtypes of rosacea, granulomatous rosacea represents a distinct variant, primarily affecting the perinasal area and characterized histopathologically by typical granulomatous lesions (74). The JAK/STAT signaling pathway plays a significant role in the pathogenesis of granulomatous rosacea. Previous studies have demonstrated that in an LL-37-induced rosacea-like mouse model, topical application of tofacitinib significantly improved rosacea-like phenotypes, reduced CD4+ T cell and mast cell infiltration, and suppressed dermal angiogenesis. RT-qPCR analysis revealed decreased mRNA expression levels of STAT1, STAT4, and STAT5a in the lesions after topical tofacitinib treatment (75). In addition to basic research, clinical applications have also confirmed these findings. A 53-year-old woman underwent a skin biopsy, which revealed granulomatous inflammation in the lesion area. Both acid-fast staining and periodic acid–Schiff (PAS) staining yielded negative results. Based on clinical manifestations and laboratory findings, the patient was diagnosed with granulomatous rosacea. After 20 weeks of treatment with the JAK-1 inhibitor abrocitinib, the skin rash and associated burning sensation substantially improved. Subsequent follow-up indicated no adverse reactions or recurrence (76).

5.4 Adverse reactions to cosmetic injectables

In the field of modern aesthetic medicine, cosmetic injectables are increasingly chosen by a wide range of beauty seekers due to their simplicity, minimally invasive nature, and relatively affordable cost (77). However, the incidence of adverse reactions has been rising as a result of issues such as substandard materials, improper injection techniques, and inadequate sterilization practices. Among all adverse reactions, chronic and disfiguring nodules (or granulomas) and swelling following cosmetic injections are one of the most common. A study showed that in their statistical analysis of histological examinations of skin lesions following adverse reactions to injections, 87.1% of patients had foreign body granulomas, and 3% had inflammatory granulomas (78). In the context of foreign body granuloma (FBG), JAK inhibitors exert therapeutic effects through multi-level immunomodulatory mechanisms. Their actions primarily include: disrupting T cell-macrophage homeostasis, inhibiting STAT6-dependent multinucleated giant cell (MGC) formation; suppressing pro-fibrotic cytokines such as TGF-β1 and IL-13 at the transcriptional level, while attenuating macrophage polarization and T cell-mediated inflammatory pathways; and downregulating M1 macrophage markers (TNF-α, CXCL10) to block key inflammatory mediators (7982). Ultimately, by inhibiting the IFN-γ-driven JAK-STAT signaling pathway, they comprehensively intervene in multiple pathophysiological aspects of granulomatous inflammation. The emergence of JAK inhibitors provides an additional treatment option for this condition with its increasing incidence. Many studies have demonstrated that JAK inhibitors, such as tofacitinib and abrocitinib, play a significant role in the treatment of post-injection granulomas (8385). Their ability to selectively suppress key inflammatory pathways offers a targeted therapeutic strategy for lesions resistant to conventional treatments.

5.5 Other types of cutaneous granulomatous diseases

In addition to the four major granulomatous skin diseases discussed above, JAK inhibitors have also shown promising therapeutic potential in a broader spectrum of granulomatous dermatoses. For instance, granulomatous cheilitis and necrobiotic xanthogranuloma—conditions characterized by persistent granulomatous inflammation and often resistant to conventional therapies—may also benefit from JAK inhibition due to their shared reliance on Th1-mediated immune activation (8689). Even in rare entities such as granulomatous mycosis fungoides or palisaded neutrophilic and granulomatous dermatitis, early evidence points to the central role of dysregulated cytokine signaling, thereby positioning JAK inhibitors as rational therapeutic options (9092). These observations collectively highlight the expanding relevance of JAK-STAT pathway inhibition across diverse granulomatous skin disorders, supporting its role as a versatile strategy for conditions with limited treatment alternatives.

6 Limitations of the current evidence base

Despite the encouraging therapeutic potential of JAK inhibitors in cutaneous granulomatous diseases, the current evidence base—primarily composed of case reports and small case series—must be interpreted with caution. For instance, open-label studies of tofacitinib in cutaneous sarcoidosis and granuloma annulare have involved only approximately 10 patients, while evidence supporting the use of upadacitinib and abrocitinib similarly derives from sporadic reports of a comparable scale (9395). Although these preliminary findings are valuable for hypothesis generation and proof-of-concept demonstration, they lack the robustness required to confirm efficacy and safety. A significant gap remains in the form of large-scale, randomized, placebo-controlled trials, which fundamentally limits the clinical persuasiveness of claims regarding the effectiveness of JAK inhibitors. Therefore, it is imperative to conduct well-designed controlled clinical trials to definitively establish efficacy, determine long-term safety, optimize dosing strategies, and identify predictive biomarkers for personalized therapy. Until such high-quality evidence is available, the use of JAK inhibitors in this context should be considered investigational and reserved for carefully selected, treatment-refractory cases. Additionally, treatment responses are heterogeneous, and not all patients achieve satisfactory clinical improvement, potentially due to disease heterogeneity, redundant signaling pathways, or pharmacokinetic variations (96). Relapse following drug discontinuation is another notable challenge, suggesting that JAK inhibition may suppress but not eradicate the underlying inflammatory process. Furthermore, the high cost of these therapies and their off-label use in many granulomatous conditions pose substantial barriers to widespread clinical adoption (97).

7 Prospects and outlook

Looking ahead, JAK inhibitors represent a promising frontier in the management of refractory cutaneous granulomatous diseases. The development of topical JAK formulations may offer a targeted approach for localized disease, minimizing systemic exposure and reducing adverse effects (98). Moreover, exploring combination therapies with conventional immunosuppressants or biologic agents may yield synergistic effects, enhancing therapeutic outcomes while allowing dose reduction of individual drugs (99).

The promising yet preliminary nature of the current evidence base underscores an urgent need for high-quality, prospective clinical trials to definitively establish the role of JAK inhibitors in cutaneous granulomatous diseases. Moving beyond anecdotal reports requires meticulously designed studies that can generate robust, generalizable data. A key consideration involves refining patient selection by focusing on those with well-defined, refractory disease and exploring the integration of predictive biomarkers—such as tissue phospho-STAT expression or serum cytokine profiles—to identify subgroups most likely to benefit from treatment. This stratification strategy would enhance trial efficiency and improve the chances of detecting a meaningful clinical signal (100, 101).

The choice of primary endpoints requires careful validation to ensure they capture clinically relevant improvement. While early-phase trials may utilize clinical response rates, definitive Phase III studies should aim to develop and implement a standardized disease activity index that incorporates lesion characteristics, physician assessments, and patient-reported outcomes like quality-of-life measures (102). Furthermore, the field would benefit from adopting innovative trial designs to overcome the challenges of studying rare diseases. A randomized withdrawal design, for instance, is particularly well-suited for demonstrating efficacy maintenance after an initial open-label induction phase. Alternatively, adaptive designs allow for pre-planned modifications based on interim data, making the development process more efficient and flexible.

Finally, establishing a comprehensive safety profile is paramount, given the chronic relapsing nature of these conditions and the known class-level risks of JAK inhibitors. Future trials must incorporate prospective, long-term monitoring protocols for serious infections, major adverse cardiovascular events, thromboembolism, and malignancy. Parallel long-term patient registries will be essential to gather robust real-world safety data that extends beyond the typical duration of controlled clinical trials (103, 104). By systematically addressing these aspects of trial design—patient stratification, endpoint validation, statistical innovation, and safety surveillance—the field can transition from off-label use to evidence-based, personalized therapy for these challenging dermatoses.

Author contributions

JG: Methodology, Visualization, Writing – review & editing, Writing – original draft. XH: Writing – review & editing. BL: Writing – review & editing. JW: Writing – review & editing. KC: Writing – review & editing. QW: Writing – review & editing. XJ: Funding acquisition, Data curation, Writing – review & editing. CH: Visualization, Validation, Resources, Supervision, Funding acquisition, Writing – review & editing. BY: Supervision, Investigation, Resources, Visualization, Funding acquisition, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by Guangdong Basic and Applied Basic Research Foundation (2025A1515010947, 2024A1515220018), Shenzhen Sanming Project (No.SZSM202311029), Shenzhen Key Medical Discipline Construction Fund (SZXK040), Shenzhen High-level Hospital Construction Fund and Peking University Shenzhen Hospital Scientific Research Fund (KYQD2024378).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Pagán AJ and Ramakrishnan L. The formation and function of granulomas. Annu Rev Immunol. (2018) 36:639–65. doi: 10.1146/annurev-immunol-032712-100022

PubMed Abstract | Crossref Full Text | Google Scholar

2. Asai J. What is new in the histogenesis of granulomatous skin diseases? J Dermatol. (2017) 44:297–303. doi: 10.1111/1346-8138.13662

PubMed Abstract | Crossref Full Text | Google Scholar

3. Joshi TP, Chen V, Dong JL, Golden K, Iacobucci A, Lim Y, et al. Psychiatric comorbidities associated with granuloma annulare: A case-control study in the All of Us database. J Am Acad Dermatol. (2022) 87:e119–e20. doi: 10.1016/j.jaad.2022.05.056

PubMed Abstract | Crossref Full Text | Google Scholar

4. Oscherwitz ME, Hrin ML, Neuberg ZJ, and Jorizzo JL. Should systemic corticosteroids be relatively contraindicated in granuloma annulare? J Eur Acad Dermatol Venereol. (2024) 38:e910–e3. doi: 10.1111/jdv.19990

PubMed Abstract | Crossref Full Text | Google Scholar

5. Hrin ML, Bowers NL, Feldman SR, and Huang WW. Methotrexate for generalized granuloma annulare: A 60% response rate in a retrospective case series of 15 patients. J Am Acad Dermatol. (2022) 87:201–3. doi: 10.1016/j.jaad.2021.07.037

PubMed Abstract | Crossref Full Text | Google Scholar

6. Adler BL, Wang CJ, Bui TL, Schilperoort HM, and Armstrong AW. Anti-tumor necrosis factor agents in sarcoidosis: A systematic review of efficacy and safety. Semin Arthritis Rheumatol. (2019) 48:1093–104. doi: 10.1016/j.semarthrit.2018.10.005

PubMed Abstract | Crossref Full Text | Google Scholar

7. Damsky W, Thakral D, Emeagwali N, Galan A, and King B. Tofacitinib treatment and molecular analysis of cutaneous sarcoidosis. N Engl J Med. (2018) 379:2540–6. doi: 10.1056/NEJMoa1805958

PubMed Abstract | Crossref Full Text | Google Scholar

8. Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, et al. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol. (2020) 80:106210. doi: 10.1016/j.intimp.2020.106210

PubMed Abstract | Crossref Full Text | Google Scholar

9. Kameda H. JAK inhibitors ∼ overview∼. Immunol Med. (2023) 46:108–11. doi: 10.1080/25785826.2023.2183594

PubMed Abstract | Crossref Full Text | Google Scholar

10. Chen A, Harview CL, Rand SE, and Harms JL. Refractory granuloma faciale successfully treated with adjunct topical JAK inhibitor. JAAD Case Rep. (2023) 33:91–4. doi: 10.1016/j.jdcr.2023.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

11. Özkoca D and Vural S. JAK inhibitors for granuloma annulare: hope for refractory cases? Int J Dermatol. (2025) 64:1335–6. doi: 10.1111/ijd.17813

PubMed Abstract | Crossref Full Text | Google Scholar

12. De Greef A, Benjelloun G, Harkemanne E, and Baeck M. Successful treatment of disseminated granuloma annulare with upadacitinib. Dermatol Ther (Heidelb). (2024) 14:813–7. doi: 10.1007/s13555-024-01117-z

PubMed Abstract | Crossref Full Text | Google Scholar

13. Villarino AV, Kanno Y, and O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. (2017) 18:374–84. doi: 10.1038/ni.3691

PubMed Abstract | Crossref Full Text | Google Scholar

14. Samra S, Bergerson JRE, Freeman AF, and Turvey SE. JAK-STAT signaling pathway, immunodeficiency, inflammation, immune dysregulation, and inborn errors of immunity. J Allergy Clin Immunol. (2025) 155:357–67. doi: 10.1016/j.jaci.2024.09.020

PubMed Abstract | Crossref Full Text | Google Scholar

15. Yu H, Pardoll D, and Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. (2009) 9:798–809. doi: 10.1038/nrc2734

PubMed Abstract | Crossref Full Text | Google Scholar

16. Xu P, Shen P, Yu B, Xu X, Ge R, Cheng X, et al. Janus kinases (JAKs): The efficient therapeutic targets for autoimmune diseases and myeloproliferative disorders. Eur J Med Chem. (2020) 192:112155. doi: 10.1016/j.ejmech.2020.112155

PubMed Abstract | Crossref Full Text | Google Scholar

17. Gao Q, Liang X, Shaikh AS, Zang J, Xu W, and Zhang Y. JAK/STAT signal transduction: promising attractive targets for immune, inflammatory and hematopoietic diseases. Curr Drug Targets. (2018) 19:487–500. doi: 10.2174/1389450117666161207163054

PubMed Abstract | Crossref Full Text | Google Scholar

18. Agashe RP, Lippman SM, and Kurzrock R. JAK: not just another kinase. Mol Cancer Ther. (2022) 21:1757–64. doi: 10.1158/1535-7163.MCT-22-0323

PubMed Abstract | Crossref Full Text | Google Scholar

19. Woznicki JA, Saini N, Flood P, Rajaram S, Lee CM, Stamou P, et al. TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis. (2021) 12:864. doi: 10.1038/s41419-021-04151-3

PubMed Abstract | Crossref Full Text | Google Scholar

20. Simon LS, Taylor PC, Choy EH, Sebba A, Quebe A, Knopp KL, et al. The Jak/STAT pathway: A focus on pain in rheumatoid arthritis. Semin Arthritis Rheumatol. (2021) 51:278–84. doi: 10.1016/j.semarthrit.2020.10.008

PubMed Abstract | Crossref Full Text | Google Scholar

21. Chovatiya R and Paller AS. JAK inhibitors in the treatment of atopic dermatitis. J Allergy Clin Immunol. (2021) 148:927–40. doi: 10.1016/j.jaci.2021.08.009

PubMed Abstract | Crossref Full Text | Google Scholar

22. Liu Y, Wang W, Zhang J, Gao S, Xu T, and Yin Y. JAK/STAT signaling in diabetic kidney disease. Front Cell Dev Biol. (2023) 11:1233259. doi: 10.3389/fcell.2023.1233259

PubMed Abstract | Crossref Full Text | Google Scholar

23. Liu J, Wang F, and Luo F. The role of JAK/STAT pathway in fibrotic diseases: molecular and cellular mechanisms. Biomolecules. (2023) 13:119. doi: 10.3390/biom13010119

PubMed Abstract | Crossref Full Text | Google Scholar

24. Shao S, Chen C, Shi G, Zhou Y, Wei Y, Wu L, et al. JAK inhibition ameliorated experimental autoimmune encephalomyelitis by blocking GM-CSF-driven inflammatory signature of monocytes. Acta Pharm Sin B. (2023) 13:4185–201. doi: 10.1016/j.apsb.2023.07.026

PubMed Abstract | Crossref Full Text | Google Scholar

25. Singh JA. Filgotinib, a JAK1 inhibitor, for treatment-resistant rheumatoid arthritis. Jama. (2019) 322:309–11. doi: 10.1001/jama.2019.9056

PubMed Abstract | Crossref Full Text | Google Scholar

26. Mease P, Helliwell P, Silwinska-Stanczyk P, Miakisz M, Ostor A, Peeva E, et al. Efficacy and safety of the TYK2/JAK1 inhibitor brepocitinib for active psoriatic arthritis: A phase IIb randomized controlled trial. Arthritis Rheumatol. (2023) 75:1370–80. doi: 10.1002/art.42519

PubMed Abstract | Crossref Full Text | Google Scholar

27. Tavoletti G, Avallone G, Conforti C, Roccuzzo G, Maronese CA, Mattioli MA, et al. Topical ruxolitinib: A new treatment for vitiligo. J Eur Acad Dermatol Venereol. (2023) 37:2222–30. doi: 10.1111/jdv.19162

PubMed Abstract | Crossref Full Text | Google Scholar

28. Nakamizo S, Sugiura Y, Ishida Y, Ueki Y, Yonekura S, Tanizaki H, et al. Activation of the pentose phosphate pathway in macrophages is crucial for granuloma formation in sarcoidosis. J Clin Invest. (2023) 133:e171088. doi: 10.1172/JCI171088

PubMed Abstract | Crossref Full Text | Google Scholar

29. Wang F, Zhang S, Jeon R, Vuckovic I, Jiang X, Lerman A, et al. Interferon gamma induces reversible metabolic reprogramming of M1 macrophages to sustain cell viability and pro-inflammatory activity. EBioMedicine. (2018) 30:303–16. doi: 10.1016/j.ebiom.2018.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

30. Sun B, Long Y, Xu G, Chen J, Wu G, Liu B, et al. Acute hypoxia modulate macrophage phenotype accompanied with transcriptome re-programming and metabolic re-modeling. Front Immunol. (2025) 16:1534009. doi: 10.3389/fimmu.2025.1534009

PubMed Abstract | Crossref Full Text | Google Scholar

31. Liu Y, Zhang M, Wang T, and Zhang J. Reactive oxygen species in asthma: regulators of macrophage polarization and therapeutic implications: A narrative review. J Asthma Allergy. (2025) 18:1129–46. doi: 10.2147/JAA.S529371

PubMed Abstract | Crossref Full Text | Google Scholar

32. Tanino T, Ueda Y, Nagai N, Ishihara Y, Saijo M, and Funakami Y. In vivo upstream factors of mouse hepatotoxic mechanism with sustained hepatic glutathione depletion: Acetaminophen metabolite-erythrocyte adducts and splenic macrophage-generated reactive oxygen species. Chem Biol Interact. (2024) 398:111091. doi: 10.1016/j.cbi.2024.111091

PubMed Abstract | Crossref Full Text | Google Scholar

33. Lim PN, Cervantes MM, Pham LK, Doherty SR, Tufts A, Dubey D, et al. Absence of c-Maf and IL-10 enables type I IFN enhancement of innate responses to LPS in alveolar macrophages. J Immunol. (2025) 214:551–64. doi: 10.1093/jimmun/vkae029

PubMed Abstract | Crossref Full Text | Google Scholar

34. Yang L, Song Y, Wang T, Cui Z, Wu J, Shi Y, et al. Transcription factor c-Maf drives macrophages to promote hypertrophic scar formation. J Cosmet Dermatol. (2024) 23:639–47. doi: 10.1111/jocd.15952

PubMed Abstract | Crossref Full Text | Google Scholar

35. Dasgupta S, Choudhury P, Patidar S, Joshi M, Dhar R, Roychowdhury S, et al. Integrative analysis of transcriptome and metabolome profiles reveals immune-metabolic alterations in pulmonary sarcoidosis. Metabolomics. (2025) 21:131. doi: 10.1007/s11306-025-02325-0

PubMed Abstract | Crossref Full Text | Google Scholar

36. Xie L, Feng J, Gao Q, Qu W, Shao S, Sun J, et al. The autoimmune profiles in the etiopathogenesis of granulomatous lobular mastitis. Immunobiology. (2025) 230:152878. doi: 10.1016/j.imbio.2025.152878

PubMed Abstract | Crossref Full Text | Google Scholar

37. Liu X, Wu XP, Zhu XL, Li T, and Liu Y. IRG1 increases MHC class I level in macrophages through STAT-TAP1 axis depending on NADPH oxidase mediated reactive oxygen species. Int Immunopharmacol. (2017) 48:76–83. doi: 10.1016/j.intimp.2017.04.012

PubMed Abstract | Crossref Full Text | Google Scholar

38. Sánchez-Bayuela T, Peral-Rodrigo M, Parra-Izquierdo I, López J, Gómez C, Montero O, et al. Inflammation via JAK-STAT/HIF-1α Drives metabolic changes in pentose phosphate pathway and glycolysis that support aortic valve cell calcification. Arterioscler Thromb Vasc Biol. (2025) 45:e232–e49. doi: 10.1161/ATVBAHA.124.322375

PubMed Abstract | Crossref Full Text | Google Scholar

39. Hu L, Liu Y, Yuan Z, Guo H, Duan R, Ke P, et al. Glucose-6-phosphate dehydrogenase alleviates epileptic seizures by repressing reactive oxygen species production to promote signal transducer and activator of transcription 1-mediated N-methyl-d-aspartic acid receptors inhibition. Redox Biol. (2024) 74:103236. doi: 10.1016/j.redox.2024.103236

PubMed Abstract | Crossref Full Text | Google Scholar

40. Xiao Q, Fang T, Peng R, Zhou M, Liu K, Qiu W, et al. Dual-layered metal nanorod-integrated carboxymethyl chitosan and hyaluronic acid hydrogels for accelerated wound healing via ROS scavenging, antibacterial activity, and macrophage reprogramming. Int J Biol Macromol. (2025) 330:148064. doi: 10.1016/j.ijbiomac.2025.148064

PubMed Abstract | Crossref Full Text | Google Scholar

41. Günther C, Ruder B, Stolzer I, Dorner H, He GW, Chiriac MT, et al. Interferon lambda promotes paneth cell death via STAT1 signaling in mice and is increased in inflamed ileal tissues of patients with crohn’s disease. Gastroenterology. (2019) 157:1310–22.e13. doi: 10.1053/j.gastro.2019.07.031

PubMed Abstract | Crossref Full Text | Google Scholar

42. Zhang Y, Xu D, Nie Q, Wang J, Fang D, Xie Y, et al. Macrophages exploit the mannose receptor and JAK-STAT1-MHC-II pathway to drive antigen presentation and the antimycobacterial immune response after BCG vaccination. Acta Biochim Biophys Sin (Shanghai). (2024) 56:1130–44. doi: 10.3724/abbs.2024100

PubMed Abstract | Crossref Full Text | Google Scholar

43. Maeng JE, Son SW, Lee SJ, Kim JC, Kim S, Kang HY, et al. Pulsed-dye laser as an effective treatment for recalcitrant granulomatous rosacea and a potential regulator of CXCL9 expression. J Dermatol. (2024) 51:722–6. doi: 10.1111/1346-8138.17051

PubMed Abstract | Crossref Full Text | Google Scholar

44. Snyder JD, Yoon TW, Lee S, Halder P, Fitzpatrick EA, and Yi AK. Protein kinase D1 in myeloid lineage cells contributes to the accumulation of CXCR3(+)CCR6(+) nonconventional Th1 cells in the lungs and potentiates hypersensitivity pneumonitis caused by S. rectivirgula. Front Immunol. (2024) 15:1403155. doi: 10.3389/fimmu.2024.1403155

PubMed Abstract | Crossref Full Text | Google Scholar

45. Li Q, Wang C, Gou J, Kitanovski S, Tang X, Cai Y, et al. Deciphering lung granulomas in HIV & TB co-infection: unveiling macrophages aggregation with IL6R/STAT3 activation. Emerg Microbes Infect. (2024) 13:2366359. doi: 10.1080/22221751.2024.2366359

PubMed Abstract | Crossref Full Text | Google Scholar

46. Johnson DE, O’Keefe RA, and Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. (2018) 15:234–48. doi: 10.1038/nrclinonc.2018.8

PubMed Abstract | Crossref Full Text | Google Scholar

47. Huang S, Liu M, Zhang H, Song W, Guo W, Feng Y, et al. HIV-MTB co-infection reduces CD4+ T cells and affects granuloma integrity. Viruses. (2024) 16:1335. doi: 10.3390/v16081335

PubMed Abstract | Crossref Full Text | Google Scholar

48. Ryzhakov G, West NR, Franchini F, Clare S, Ilott NE, Sansom SN, et al. Alpha kinase 1 controls intestinal inflammation by suppressing the IL-12/Th1 axis. Nat Commun. (2018) 9:3797. doi: 10.1038/s41467-018-06085-5

PubMed Abstract | Crossref Full Text | Google Scholar

49. Tolomeo M and Cascio A. The STAT signaling pathway in HIV-1 infection: roles and dysregulation. Int J Mol Sci. (2025) 26:9123. doi: 10.3390/ijms26189123

PubMed Abstract | Crossref Full Text | Google Scholar

50. Willemsen L, Lee J, Shinde P, Soldevila F, Aoki M, Orfield S, et al. Th1 polarization in Bordetella pertussis vaccine responses is maintained through a positive feedback loop. Nat Commun. (2025) 16:3132. doi: 10.1038/s41467-025-58460-8

PubMed Abstract | Crossref Full Text | Google Scholar

51. Ma Z, Sun X, Lin Y, Wang Z, Nie Q, Yu J, et al. The dynamic expression changes of neutrophil extracellular traps in mouse apical periodontitis: A potential correlation with IL-17. J Immunol Res. (2025) 2025:8039031. doi: 10.1155/jimr/8039031

PubMed Abstract | Crossref Full Text | Google Scholar

52. Damsky W, Thakral D, McGeary MK, Leventhal J, Galan A, and King B. Janus kinase inhibition induces disease remission in cutaneous sarcoidosis and granuloma annulare. J Am Acad Dermatol. (2020) 82:612–21. doi: 10.1016/j.jaad.2019.05.098

PubMed Abstract | Crossref Full Text | Google Scholar

53. Koçak A, Ural C, Cavdar Z, Sarioğlu S, Akdoğan G, and Birlik M. Selective JAK-1 inhibitor Upadacitinib and peptide PD29 modulate the JAK and TGF-β/Smad signaling pathways reducing experimental dermal fibrosis. Toxicol In Vitro. (2025) 107:106078. doi: 10.1016/j.tiv.2025.106078

PubMed Abstract | Crossref Full Text | Google Scholar

54. Ruscitti P, Liakouli V, Panzera N, Angelucci A, Berardicurti O, Di Nino E, et al. Tofacitinib may inhibit myofibroblast differentiation from rheumatoid-fibroblast-like synoviocytes induced by TGF-β and IL-6. Pharm (Basel). (2022) 15:622. doi: 10.3390/ph15050622

PubMed Abstract | Crossref Full Text | Google Scholar

55. Zanders L, Kny M, Hahn A, Schmidt S, Wundersitz S, Todiras M, et al. Sepsis induces interleukin 6, gp130/JAK2/STAT3, and muscle wasting. J Cachexia Sarcopenia Muscle. (2022) 13:713–27. doi: 10.1002/jcsm.12867

PubMed Abstract | Crossref Full Text | Google Scholar

56. Reece MD, Song C, Hancock SC, Pereira Ribeiro S, Kulpa DA, and Gavegnano C. Repurposing BCL-2 and Jak 1/2 inhibitors: Cure and treatment of HIV-1 and other viral infections. Front Immunol. (2022) 13:1033672. doi: 10.3389/fimmu.2022.1033672

PubMed Abstract | Crossref Full Text | Google Scholar

57. Sakai Y, Uchida K, and Nakayama H. Histopathological features and expression profiles of cytokines, chemokines and SOCS family proteins in trehalose 6,6’-dimycolate-induced granulomatous lesions. Inflammation Res. (2011) 60:371–8. doi: 10.1007/s00011-010-0280-7

PubMed Abstract | Crossref Full Text | Google Scholar

58. Durham GA, Williams JJL, Nasim MT, and Palmer TM. Targeting SOCS proteins to control JAK-STAT signalling in disease. Trends Pharmacol Sci. (2019) 40:298–308. doi: 10.1016/j.tips.2019.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

59. Kubo S, Nakayamada S, and Tanaka Y. JAK inhibitors for rheumatoid arthritis. Expert Opin Investig Drugs. (2023) 32:333–44. doi: 10.1080/13543784.2023.2199919

PubMed Abstract | Crossref Full Text | Google Scholar

60. King BA and Craiglow BG. Janus kinase inhibitors for alopecia areata. J Am Acad Dermatol. (2023) 89:S29–s32. doi: 10.1016/j.jaad.2023.05.049

PubMed Abstract | Crossref Full Text | Google Scholar

61. Keenan C, Nichols KE, and Albeituni S. Use of the JAK inhibitor ruxolitinib in the treatment of hemophagocytic lymphohistiocytosis. Front Immunol. (2021) 12:614704. doi: 10.3389/fimmu.2021.614704

PubMed Abstract | Crossref Full Text | Google Scholar

62. Drent M, Crouser ED, and Grunewald J. Challenges of sarcoidosis and its management. N Engl J Med. (2021) 385:1018–32. doi: 10.1056/NEJMra2101555

PubMed Abstract | Crossref Full Text | Google Scholar

63. Kraaijvanger R, Janssen Bonás M, Vorselaars ADM, and Veltkamp M. Biomarkers in the diagnosis and prognosis of sarcoidosis: current use and future prospects. Front Immunol. (2020) 11:1443. doi: 10.3389/fimmu.2020.01443

PubMed Abstract | Crossref Full Text | Google Scholar

64. Kerkemeyer KL, Meah N, and Sinclair RD. Tofacitinib for cutaneous and pulmonary sarcoidosis: A case series. J Am Acad Dermatol. (2021) 84:581–3. doi: 10.1016/j.jaad.2020.10.016

PubMed Abstract | Crossref Full Text | Google Scholar

65. Wei JJ, Kallenbach LR, Kreider M, Leung TH, and Rosenbach M. Resolution of cutaneous sarcoidosis after Janus kinase inhibitor therapy for concomitant polycythemia vera. JAAD Case Rep. (2019) 5:360–1. doi: 10.1016/j.jdcr.2019.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

66. Scheinberg M, Maluf F, and Wagner J. Steroid-resistant sarcoidosis treated with baricitinib. Ann Rheum Dis. (2020) 79:1259–60. doi: 10.1136/annrheumdis-2020-217271

PubMed Abstract | Crossref Full Text | Google Scholar

67. Chen A, Truong AK, and Worswick S. The role of biologics in the treatment of chronic granuloma annulare. Int J Dermatol. (2019) 58:622–6. doi: 10.1111/ijd.14350

PubMed Abstract | Crossref Full Text | Google Scholar

68. Wang A, Rahman NT, McGeary MK, Murphy M, McHenry A, Peterson D, et al. Treatment of granuloma annulare and suppression of proinflammatory cytokine activity with tofacitinib. J Allergy Clin Immunol. (2021) 147:1795–809. doi: 10.1016/j.jaci.2020.10.012

PubMed Abstract | Crossref Full Text | Google Scholar

69. Chu CY. New targets in treating granuloma annulare. J Allergy Clin Immunol. (2021) 147:1646–7. doi: 10.1016/j.jaci.2021.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

70. Michels A, Heiland R, Hammerschmidt S, Farcas A, Voigt TP, Braun SA, et al. Successful treatment of recalcitrant generalized granuloma annulare with the JAK inhibitor abrocitinib. J Dtsch Dermatol Ges. (2024) 22:841–3. doi: 10.1111/ddg.15426

PubMed Abstract | Crossref Full Text | Google Scholar

71. Liu W, Chen W, Tian X, Yu Y, Zhu J, Liang J, et al. Oral abrocitinib in the treatment of granuloma annulare: a case report. J Dermatolog Treat. (2024) 35:2313090. doi: 10.1080/09546634.2024.2313090

PubMed Abstract | Crossref Full Text | Google Scholar

72. Stratman S, Amara S, Tan KJ, George-Washburn EA, Shokrian N, Lau W, et al. Systemic Janus kinase inhibitors in the management of granuloma annulare. Arch Dermatol Res. (2025) 317:743. doi: 10.1007/s00403-025-04248-1

PubMed Abstract | Crossref Full Text | Google Scholar

73. van Zuuren EJ, Arents BWM, van der Linden MMD, Vermeulen S, Fedorowicz Z, and Tan J. Rosacea: new concepts in classification and treatment. Am J Clin Dermatol. (2021) 22:457–65. doi: 10.1007/s40257-021-00595-7

PubMed Abstract | Crossref Full Text | Google Scholar

74. Jiang Y, Huang Y, Ma G, Liu T, Li Q, Wu H, et al. Granulomatous rosacea in Chinese patients: Clinical-histopathological analysis and pathogenesis exploration. J Dermatol. (2023) 50:856–68. doi: 10.1111/1346-8138.16767

PubMed Abstract | Crossref Full Text | Google Scholar

75. Sun R, Fan H, Liu J, Gao G, Liu C, Zhang D, et al. The treatment of Tofacitinib for rosacea through the inhibition of the JAK/STAT signaling pathway. Arch Dermatol Res. (2024) 316:566. doi: 10.1007/s00403-024-03314-4

PubMed Abstract | Crossref Full Text | Google Scholar

76. Ren M, Yang X, Teng Y, Lu W, Ding Y, and Tao X. Successful treatment of granulomatous rosacea by JAK inhibitor abrocitinib: A case report. Clin Cosmet Investig Dermatol. (2023) 16:3369–74. doi: 10.2147/CCID.S440138

PubMed Abstract | Crossref Full Text | Google Scholar

77. Triana L, Palacios Huatuco RM, Campilgio G, and Liscano E. Trends in surgical and nonsurgical aesthetic procedures: A 14-year analysis of the international society of aesthetic plastic surgery-ISAPS. Aesthetic Plast Surg. (2024) 48:4217–27. doi: 10.1007/s00266-024-04260-2

PubMed Abstract | Crossref Full Text | Google Scholar

78. MaChado RA, Oliveira LQ, Martelli-Júnior H, Pires FR, Carvas JB, Rogerio VE, et al. Adverse reactions to the injection of face and neck aesthetic filling materials: a systematic review. Med Oral Patol Oral Cir Bucal. (2023) 28:e278–e84. doi: 10.4317/medoral.25713

PubMed Abstract | Crossref Full Text | Google Scholar

79. Fortelny N, Farlik M, Fife V, Gorki AD, Lassnig C, Maurer B, et al. JAK-STAT signaling maintains homeostasis in T cells and macrophages. Nat Immunol. (2024) 25:847–59. doi: 10.1038/s41590-024-01804-1

PubMed Abstract | Crossref Full Text | Google Scholar

80. Stark GR and Darnell JE Jr. The JAK-STAT pathway at twenty. Immunity. (2012) 36:503–14. doi: 10.1016/j.immuni.2012.03.013

PubMed Abstract | Crossref Full Text | Google Scholar

81. Nyirenda MH, Nijjar JS, Frleta-Gilchrist M, Gilchrist DS, Porter D, Siebert S, et al. JAK inhibitors disrupt T cell-induced proinflammatory macrophage activation. RMD Open. (2023) 9:e002671. doi: 10.1136/rmdopen-2022-002671

PubMed Abstract | Crossref Full Text | Google Scholar

82. Lescoat A, Lelong M, Jeljeli M, Piquet-Pellorce C, Morzadec C, Ballerie A, et al. Combined anti-fibrotic and anti-inflammatory properties of JAK-inhibitors on macrophages in vitro and in vivo: Perspectives for scleroderma-associated interstitial lung disease. Biochem Pharmacol. (2020) 178:114103. doi: 10.1016/j.bcp.2020.114103

PubMed Abstract | Crossref Full Text | Google Scholar

83. Mansouri P and Farshi S. The new frontier of JAK inhibitors: significant therapeutic response to tofacitinib in a patient with granulomatous reaction to filler in the buttocks. J Cosmet Dermatol. (2024) 23:3924–6. doi: 10.1111/jocd.16578

PubMed Abstract | Crossref Full Text | Google Scholar

84. Wang J, Chen Z, Wu L, Liao Y, and Yu B. Tofacitinib for managing granuloma formation after dermal filler injection: three case reports and literature review. J Dermatolog Treat. (2024) 35:2338281. doi: 10.1080/09546634.2024.2338281

PubMed Abstract | Crossref Full Text | Google Scholar

85. Li Z, Lu J, Wu M, Yu B, and Yang Z. Successful treatment of delayed onset nodules after dermal fillers injection with abrocitinib: A case report. Clin Cosmet Investig Dermatol. (2025) 18:955–9. doi: 10.2147/CCID.S524045

PubMed Abstract | Crossref Full Text | Google Scholar

86. De Greef A, Peeters C, Dewit O, de Montjoye L, and Baeck M. Upadacitinib for treatment of granulomatous cheilitis. JAMA Dermatol. (2024) 160:1001–3. doi: 10.1001/jamadermatol.2024.2378

PubMed Abstract | Crossref Full Text | Google Scholar

87. Miller AC, Harding T, Houpe J, Jacox E, Schaffer A, Jain A, et al. Treatment of necrobiotic xanthogranuloma with upadacitinib: A case report. Int J Dermatol. (2025) 64:2315–8. doi: 10.1111/ijd.17873

PubMed Abstract | Crossref Full Text | Google Scholar

88. Ramassamy S, Van HTA, Chuang JY, and Wu YH. Pathological and immunohistochemical characteristics of granuloma and lymphatics in cheilitis granulomatosa. Am J Dermatopathol. (2022) 44:83–91. doi: 10.1097/DAD.0000000000001952

PubMed Abstract | Crossref Full Text | Google Scholar

89. Ding X, Cao Y, Xing Y, Ge S, Lin M, and Li J. TIMP-1 mediates inflammatory and immune response to IL-6 in adult orbital xanthogranulomatous disease. Ocul Immunol Inflamm. (2020) 28:288–97. doi: 10.1080/09273948.2019.1581227

PubMed Abstract | Crossref Full Text | Google Scholar

90. Quadri I, Reneau JC, Hanel W, and Chung CG. Advancements in the treatment of mycosis fungoides and Sézary syndrome: monoclonal antibodies, immunotherapies, and Janus kinase inhibitors. Front Immunol. (2023) 14:1291259. doi: 10.3389/fimmu.2023.1291259

PubMed Abstract | Crossref Full Text | Google Scholar

91. Moskowitz AJ, Ghione P, Jacobsen E, Ruan J, Schatz JH, Noor S, et al. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas. Blood. (2021) 138:2828–37. doi: 10.1182/blood.2021013379

PubMed Abstract | Crossref Full Text | Google Scholar

92. Chung WH, Chen CB, and Chan TM. Baricitinib treatment for palisaded neutrophilic granulomatous dermatitis: A new paradoxical reaction to tocilizumab. Dermatitis. (2023) 34:466–7. doi: 10.1097/DER.0000000000000879

PubMed Abstract | Crossref Full Text | Google Scholar

93. Damsky W, Wang A, Kim DJ, Young BD, Singh K, Murphy MJ, et al. Inhibition of type 1 immunity with tofacitinib is associated with marked improvement in longstanding sarcoidosis. Nat Commun. (2022) 13:3140. doi: 10.1038/s41467-022-30615-x

PubMed Abstract | Crossref Full Text | Google Scholar

94. Dev A, Keshavamurthy V, and Chatterjee D. Therapeutic success of tofacitinib in granuloma annulare: A retrospective case series of 15 patients. Indian J Dermatol Venereol Leprol. (2025) 91:65–70. doi: 10.25259/IJDVL_215_2024

PubMed Abstract | Crossref Full Text | Google Scholar

95. Coican A, Meckley A, Sagasser N, Greenfield M, Song EJ, and El-Bahri J. Successful treatment of refractory generalized granuloma annulare with upadacitinib. Case Rep Dermatol Med. (2024) 2024:8859178. doi: 10.1155/2024/8859178

PubMed Abstract | Crossref Full Text | Google Scholar

96. Yuan J, Lu H, Zuo X, Yin L, Pu Y, and Zhang J. Adverse event assessment of upadacitinib: A pharmacovigilance study based on the FAERS database. Pharmacoepidemiol Drug Saf. (2024) 33:e70030. doi: 10.1002/pds.70030

PubMed Abstract | Crossref Full Text | Google Scholar

97. Li GX, Sun HY, Pham JP, Tatian AH, Polcz M, Chee SN, et al. Cost-utility analysis of upadacitinib vs. dupilumab for atopic dermatitis in Australia. Clin Exp Dermatol. (2025) 50:1551–60. doi: 10.1093/ced/llaf097

PubMed Abstract | Crossref Full Text | Google Scholar

98. Kang C. Ruxolitinib cream 1.5%: A review in non-segmental vitiligo. Drugs. (2024) 84:579–86. doi: 10.1007/s40265-024-02055-y

PubMed Abstract | Crossref Full Text | Google Scholar

99. Ahmed W, Galati J, Kumar A, Christos PJ, Longman R, Lukin DJ, et al. Dual biologic or small molecule therapy for treatment of inflammatory bowel disease: A systematic review and meta-analysis. Clin Gastroenterol Hepatol. (2022) 20:e361–e79. doi: 10.1016/j.cgh.2021.03.034

PubMed Abstract | Crossref Full Text | Google Scholar

100. Hao L, Imran QM, and Ullah N. CD39 dynamics in tuberculosis: a potential biomarker of immune dysregulation and T cell exhaustion. Front Immunol. (2025) 16:1601637. doi: 10.3389/fimmu.2025.1601637

PubMed Abstract | Crossref Full Text | Google Scholar

101. van Wijck RTA, Sharma HS, Swagemakers SMA, Dik WA, IJ H, Dalm V, et al. Bioinformatic meta-analysis reveals novel differentially expressed genes and pathways in sarcoidosis. Front Med (Lausanne). (2024) 11:1381031. doi: 10.3389/fmed.2024.1381031

PubMed Abstract | Crossref Full Text | Google Scholar

102. Wechsler ME, Silver J, Wolff G, Price RG, Verghis R, Weller PF, et al. Long-term safety and efficacy of mepolizumab in eosinophilic granulomatosis with polyangiitis. Arthritis Rheumatol. (2025) 77:1052–62. doi: 10.1002/art.43146

PubMed Abstract | Crossref Full Text | Google Scholar

103. Altınöz Güney C and Koç HA. Investigation of infection risk associated with janus kinase inhibitors: A pharmacovigilance analysis using the food and drug administration’s adverse event reporting system database. Clin Exp Dermatol. (2025). doi: 10.1093/ced/llaf427

PubMed Abstract | Crossref Full Text | Google Scholar

104. Lin J, Cheng Y, Chen L, Chen M, and Shen Z. A real-world analysis of adverse event signals of cognitive and communication disorder in patients treated with Janus kinase inhibitors based on the FAERS database. J Am Acad Dermatol. (2025) 93:1562–4. doi: 10.1016/j.jaad.2025.08.003

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: JAK inhibitors, cutaneous granulomatous diseases, JAK-STAT pathway, targeted therapy, clinical practice

Citation: Gu J, He X, Lu B, Wang J, Chen K, Wang Q, Jian X, Huang C and Yu B (2025) Clinical updates of JAK inhibitors in cutaneous granulomatous diseases. Front. Immunol. 16:1698816. doi: 10.3389/fimmu.2025.1698816

Received: 04 September 2025; Accepted: 27 November 2025; Revised: 06 October 2025;
Published: 12 December 2025.

Edited by:

Fabio Candotti, Centre Hospitalier Universitaire Vaudois (CHUV), Switzerland

Reviewed by:

Toshiaki Kogame, Kyoto University, Japan

Copyright © 2025 Gu, He, Lu, Wang, Chen, Wang, Jian, Huang and Yu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Cong Huang, Y29uZ2h1YW5ncGhkMTk4OEAxNjMuY29t; Bo Yu, ZHJib3l1X2Rlcm1AMTI2LmNvbQ==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.