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

MINI REVIEW article

Front. Immunol., 03 December 2025

Sec. Autoimmune and Autoinflammatory Disorders: Autoinflammatory Disorders

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1715107

This article is part of the Research TopicEarly Diagnosis and Intervention in Psoriatic Arthritis: Unraveling Clinical and Immunoinflammatory MysteriesView all 5 articles

Beyond the joints: mechanistic insights and multidisciplinary strategies for spondyloarthritis-associated uveitis

  • 1Department of Rheumatology, Fifth People’s Hospital Affiliated to Chengdu University of Traditional Chinese Medicine, Chengdu, China
  • 2Department of Radiology, Fifth People’s Hospital Affiliated to Chengdu University of Traditional Chinese Medicine, Chengdu, China
  • 3Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China

Spondyloarthritis (SpA) is a group of diseases characterized by chronic inflammation and extra-articular involvement of tissues and organs. Acute anterior uveitis (AAU), the most common extra-articular manifestation of SpA, typically presents as a unilateral acute disruption of the blood-aqueous barrier, characterized by photophobia, redness of the eye, pain, and blurred vision. The underlying pathogenesis is not fully understood but is thought to involve genetic factors, gut microbiota, and dysregulation of the immune system. Corticosteroids remain the cornerstone of AAU treatment. However, alternative therapies such as nonsteroidal anti-inflammatory drugs (NSAIDs), disease modifying antirheumatic drugs (DMARDs), and biologics have also demonstrated utility. In recent years, biologics have gained increasing attention due to their efficacy and safety. This review summarizes recent research findings and advancements in the diagnosis and management of spondyloarthritis-associated uveitis.

1 Introduction

Spondyloarthritis (SpA) is a group of systemic chronic inflammatory rheumatic immune diseases characterized by significant clinical heterogeneity. The global prevalence of SpA varies significantly, ranging from 0.20% in Southeast Asia to 1.61% in the Arctic regions (1). Common subtypes include ankylosing spondylitis (AS), psoriatic arthritis (PsA), reactive arthritis (ReA), inflammatory bowel disease-associated arthritis (IBD-A), and undifferentiated spondyloarthritis (uSpA) (2). These conditions typically affect sacroiliac joints, spine, and peripheral joints, causing inflammatory low back pain and/or asymmetric peripheral arthritis. They are often accompanied by extra-articular multisystem involvement (3). Ocular involvement is a common extra-articular manifestation in SpA patients, with uveitis being the most common type (4). Early recognition, diagnosis and systematic treatment of the SpA complicated by uveitis are critical for improving patient outcomes. This article reviews the clinical manifestations, etiology, pathogenesis, and treatment of SpA-associated uveitis.

2 Methods

2.1 Search strategy

In this review, a systematic search strategy was employed to identify relevant peer-reviewed articles from 2020 to 2025 in the electronic databases of PubMed, Embase, China National Knowledge Infrastructure (CNKI), and Wanfang, without any language restrictions applied. The search terms comprised "Spondyloarthritis", "Uveitis", "Ankylosing Spondylitis", "Psoriatic Arthritis" ,"Enteropathic Arthritis", "Targeted Therapy" ,"HLA-B27", and "Spondylitis". Additionally, the reference lists of retrieved articles were examined to identify other relevant studies for inclusion.

2.2 Inclusion criteria and exclusion criteria

Inclusion Criteria: (1) Adult patients (age ≥18 years) with a definitive diagnosis of spondyloarthritis (including axial SpA, ankylosing spondylitis, reactive arthritis, psoriatic arthritis, or enteropathic arthritis) accompanied by uveitis of any type (e.g., anterior, intermediate, posterior, or panuveitis), regardless of race or sex. (2) Studies investigating any aspect of SpA-associated uveitis, including but not limited to epidemiology, pathogenesis, clinical manifestations, diagnosis, efficacy and safety of treatments.

Exclusion Criteria: (1) Pediatric patients (age <18 years). (2) Patients with uveitis secondary to other definite causes (e.g., infection, trauma, or other autoimmune diseases such as sarcoidosis or Behçet’s disease). (3) Case reports (with fewer than 2 cases) and studies from which data could not be extracted.

All retrieved records were imported into a reference management software (e.g., EndNote) for automatic removal of duplicates. Two authors (S-YP and LX) conducted the electronic search independently, and any discrepancies were resolved by a third author (ZC).

3 Clinical manifestations and characteristics

Extra-articular manifestations of SpA are often overlooked and primarily involve multiple systems, including eyes, skin, gastrointestinal tract, bone, heart, lung and kidney (5). Among these, uveitis is regarded as one of the most prevalent extra-articular manifestations of SpA, with acute anterior uveitis (AAU) representing the predominant form, accounting for approximately 60% to 90% of cases (5). The prevalence of AAU varies in different regions, with higher rates observed in in Europe and North America compared to Asia (6). In a study by Rupali et al. (7), 225 SpA patients were evaluated and 42 patients (18.7%) were found to have uveitis. Uveitis was more common in males than in females, with ratio of 2.5:1. Furthermore, the incidence of uveitis increases with the duration of SpA. At the time of diagnosis, the incidence is approximately 10.5%, rising to 46.6% after 30 years of disease (8). The prevalence of uveitis also varies among different subtypes of SpA: AS is reported to have the highest prevalence (32.2%), followed by IBD-A at 36.9% and PsA at 25.1% (9).

Uveitis refers to the inflammation of the uveal tract of the eye, which includes the iris, ciliary body and choroid. It can be classified based on the anatomical location of the inflammation into anterior, intermediate, posterior, or panuveitis (involving all three segments) (10). In Western countries, anterior uveitis is the most common form, while in developing countries, posterior uveitis and panuveitis are more frequently observed (11). Clinically, uveitis is further categorized into acute (lasting less than 3 months), chronic (more than 3 months), or recurrent uveitis (recurring after resolution from a prior episode) (12). SpA-associated uveitis most commonly presents as AAU. When uveitis persists, severe disruption of the blood-aqueous barrier can occur and lead to significant infiltration of inflammatory cells in the anterior chamber. This process is usually accompanied by fibrin exudate and hypopyon, resulting in adhesion between the iris and lens (13). In addition, severe and/or recurrent AAU may lead to structural complications, such as cataract, elevated intraocular pressure, glaucoma, and cystoid macular edema, which can progress to chronic anterior uveitis (14, 15).

AAU primarily affects the anterior segment structures of the eye, including the iris and ciliary body, but it can also involve the posterior segment through the dissemination of immune mediators. SpA-associated uveitis is typically non-granulomatous, acute, and unilateral (16). On the one hand, the prognosis for AS-related uveitis is generally good, with most cases resolving spontaneously within 4 to 6 weeks, although recurrence is common. Patients with AS-related uveitis are often young adults, and while the majority experience some degree of visual impairment, severe vision loss is rare (17). In the early stages of the disease, ciliary muscle spasm can cause photophobia and localized ocular discomfort. As the disease progresses, patient may develop characteristic symptoms such as photophobia, red eye, lacrimation, pain, and blurred vision. On the other hand, uveitis associated with IBD-A and PsA more commonly affect both eyes. It tends to have a more insidious onset, slow progression and prolonged duration. They may present as vitritis, affecting the middle and posterior segments of the eye, and can lead to various severe ocular complications (18, 19).

Risk factors associated with secondary uveitis in patients with SpA include age, smoking history, delayed diagnosis, long disease duration, HLA-B27 positivity, family history of SpA, and radiographic damage (20). These factors provide valuable guidance for clinical practice. For those patients with SpA risk factors, such as arthritis and family history, ophthalmologists and rheumatologists should maintain a high level of vigilance and timely carry out the necessary examinations and evaluations (Figure 1). Uveitis associated with SpA is generally a form of non-infectious inflammation. During an acute episode of SpA-associated AAU, intraocular pressure in the affected eye is typically lower than that of the contralateral eye due to the release of prostaglandins. This characteristic can help differentiate SpA-associated AAU from viral anterior uveitis and other types of uveitis (21). The prognosis of SpA-associated uveitis largely depends on the timeliness of diagnosis and treatment. If diagnosed and treated early, most patients experience a favorable outcome. Therefore, detailed eye examination and systematic evaluation are particularly important (22). Eye examinations mainly include vision assessment, intraocular pressure measurement, slit lamp examination, optical coherence tomography (OCT), and fundus examination. Slit lamp examination provides high magnification images of the eye, enabling observation of the distribution of inflammatory cells, fibrin exudates, and provide overall evaluation of intraocular inflammation. OCT is a valuable monitoring tool that can identify macular edema early and assist in management.

Figure 1
Flowchart illustrating the referral and evaluation process between SpA and AAU with a question mark at the top. SpA involves medical history, physical, laboratory, and radiological examinations by a Rheumatologist. AAU includes medical history, laboratory, and ophthalmic examinations by an Ophthalmologist. The central diamond highlights “Timely bidirectional referral and evaluation.

Figure 1. Diagnostic flowchart for AAU in spondyloarthritis.

Diagnosis of SpA-associated uveitis should be targeted based on thorough review of medical history, symptoms, uveitis course, ocular signs, and laboratory data (23). The Standardization of Uveitis Nomenclature Working Group has released the main classification criteria for SpA/HLA-B27-related AAU (24). The first step in classifying is to exclude infectious causes of uveitis. Then the following situations can be classified as SpA/HLA-B27-AU: For patients presenting with acute or recurrent acute, unilateral or unilateral alternating AAU, accompanied by spinal arthritis or positive HLA-B27 test; or patients with a history of recurrent acute, unilateral, or unilateral alternating in the context of a chronic disease, along with spinal arthritis or positive HLA-B27 test; or patients with AAU accompanied by spondyloarthritis and positive HLA-B27. In addition, the Dublin uveitis assessment tool (Duet) (25)recommends that AAU patients who present with inflammatory back pain or HLA-B27 positivity, regardless of the presence of axial or peripheral SpA features, should be referred to a rheumatology specialist for a comprehensive evaluation. Such interdisciplinary collaboration is crucial for the early screening, identification, and treatment of potential SpA cases (26).

4 Research progress on etiology and pathogenesis

The exact etiology and pathogenesis of SpA-associated uveitis remain complex and are not completely understood. Current research shows that it may be related to genetic factors, gut microbiota, infection factors and dysregulation of immune system (Table 1). We summarized the research in understanding the mechanism underlying AAU.

Table 1
www.frontiersin.org

Table 1. Overview of the pathological mechanisms of spondyloarthritis-associated uveitis.

4.1 Genetic factors

Genetic factors play a significant role in SpA, with the strongest association being with the human leukocyte antigen HLA-B27. Approximately 50% of AAU patients are HLA-B27 positive (27). HLA-B27 increases the risk of AAU in patients with SpA, and for HLA-B27 positive individuals without SpA, the risk of developing AAU is 23 times higher than that of the general population (28). Studies have shown that as the duration of SpA increases, the incidence of uveitis also rises, particularly among HLA-B27 positive patients, with an odds ratio of 4.2 (29). This highlights the importance of routine HLA-B27 testing in patients with AAU, as it may influence disease severity and prognosis. A retrospective study conducted in Sydney further supported routine testing, showing that HLA-B27 positive patients had relatively poorer visual outcomes after review of 241 AAU cases (30). Currently, there are three main hypotheses explain the role of HLA-B27 in the pathogenesis of SpA-associated uveitis: Arthritis/uveal peptide hypothesis, HLA-B27 misfolding hypothesis and innate immune recognition of abnormal HLA-B27 (31).

According to the Arthritis/uveal peptide hypothesis, HLA-B27, as a major histocompatibility complex (MHC) class I molecule, presents processed self-peptides or antigens to the surface of cytotoxic CD8+T cells. Upon recognition, this process can lead to auto-reactivity, inducing inflammation in the joints or eyes, resulting in arthritis and uveitis (5). The fundamental mechanism of this process revolves around the recognition of HLA-B27 bound self-peptide complexes by the T-cell receptor (TCR), which subsequently triggers a specific autoimmune response. Hermann et al. (32)tested the synovial fluid of four patients with ReA and two patients with AS. From these samples, they obtained 354 αβ-TCR CD8+T lymphocyte clones (TLCs). Autoreactive cytotoxic T cells were identified in five of the six patients, with five demonstrating HLA-B27 restricted killing of uninfected cell lines. Yang et al. (33) identified a clonally expanded TCR (TRBV9/TRAV21) associated with AS and AAU. They propose that this TCR interacts with a common HLA-B27-peptide complex, thereby contributing to the pathogenesis of both conditions. To date, over 200 HLA-B27 subtypes have been reported based on nucleotide sequence polymorphisms. Among these, HLA-B27*04 and HLA-B27*05 subtypes are associated with SpA and AAU, while HLA-B27*09 shows no association (34, 35). A comprehensive analysis of the eight most common HLA-B27 subtypes revealed considerable overlap among the peptidomes, suggesting that it may be the quantity rather than the quality of peptides that contribute to disease occurrence – a discovery of considerable significance in understanding the pathogenesis of SpA-associated uveitis (36). Despite advancements in comprehending HLA-B27 antigen presentation, critical questions regarding the specific self-peptides involved and the mechanisms of immune activation remain unanswered, necessitating further investigation.

The HLA-B27 misfolding hypothesis posits that the inherent instability of the HLA-B27 molecule makes it prone to misfolding. This can trigger endoplasmic reticulum (ER) stress, thereby activating the unfolded protein response (UPR) (37). In some cases, the upregulation of UPR genes can further lead to the activation of the NF-κB signaling pathway, resulting in the production of pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6, causing inflammation (38). It has also been reported that UPR may participate in the development of the disease by activating the IL-23/IL-17 axis (39). These pathways could all contribute to the development of SpA-associated uveitis.

In SpA-associated uveitis, the HLA-B27 molecule may directly participate in immune activation by forming dimers or free heavy chains (FHCs). The formation of dimers, which occurs in the absence of β2-microglobulin (β2m), is dependent on the Cys67 residue on its molecule (40). These “abnormal” cell surface forms can be recognized by the innate immune system, including natural killer (NK) cells or the killer cell immunoglobulin-like receptors KIR3DL1, KIR3DL2, and the immunoglobulin-like transcript 4 (ILT4) on T helper cells (41), thereby stimulating CD4+T lymphocytes to produce IL-17, initiating an immune response (42, 43). In a study observing KIR gene polymorphisms, HLA-B27 positive AS patients had higher levels of KIR2DL1 and KIR2DL5 compared to those without AS (44). Furthermore, research conducted by Vendelbosch et al. (45)indicated that the severity of AS in patients appears to be influenced by KIR genotypes. However, there were no significant differences between patients and controls in terms of KIR haplotypes, genes, or allele distributions.

In recent years, genome-wide association studies (GWAS) have identified numerous genetic links associated with AAU and SpA. Apart from HLA-B27, HLA-DR13 is also closely related to uveitis in PsA patients (46). Researchers have discovered that genetic variations in non-HLA regions may also be associated with SpA-associated uveitis. These including ERAP1, intergenic region 2p15, IL23R, IL-10-19, and IL-18R1-IL-1R1 genes, which predispose to uveitis and inflammatory bowel disease. In contrast genes such as IL-6R, KIF21B, and EYS appear to specifically predispose individuals to AAU (47). Furthermore, associations have also been found at previously unrecognized loci for AS and AU, including MERTK, KIFAP3, CLCN7, ACAA2 (48). Among these genes, the endoplasmic reticulum aminopeptidases ERAP1 and ERAP2 have genetic associations with SpA that are second only to HLA-B27. These enzymes regulate the length of peptides by trimming N-terminal amino acids, enabling the peptides to fit with MHC class I molecules and present them on the cell surface to CD8+ T cells or NK cells (49).

4.2 Gut microbiota

The human body, gut microbiota, host organism, and environment are interdependent mutually influence one another. Gut microbes are thought to play a crucial role in the pathogenesis of SpA-associated uveitis, but the exact mechanisms are not yet clear.

The diversity and composition of gut microbiota vary in different diseases. Studies have found that patients with AS have increased abundance of Lachnospiraceae, Ruminococcaceae, Rikenellaceae, Porphyromonadaceae, and Bacteroidaceae in the terminal ileum, while Veillonellaceae and Prevotellaceae are reduced (50).The Firmicutes/Bacteroidetes (F/B) ratio is critical for maintaining normal gut homeostasis, and an increase in Firmicutes species abundance has been linked to AS (51). Compared with healthy controls, uveitis patients showed enrichment of Prevotella and Streptococcus, and decreased diversity of Faecalibacterium, Bacteroides, Lachnospira, Ruminococcus, Lachnospiraceae and Ruminococcaceae (52). Recently, studies have shown that patients with AAU and patients with SpA and Crohn’s disease have increased Collinsella and decreased Lachnospiraceae, Blautia and Fusicatenibacter (53). However, it remains unclear whether dysbiosis is a cause or consequence of inflammation, and methodological variations limit the comparability and generalizability of microbiota findings.

Animal models provide a valuable tool for investigating the role of gut microbiota in the pathogenesis of SpA-associated uveitis. Experiments have shown that HLA-B27 transgenic rats raised in a sterile environment have a reduced incidence of intestinal and joint inflammatory diseases, confirming the significant role of microbiota in disease development (54). Oral antibiotic treatment can significantly reduce ocular inflammation compared to intraperitoneal administration, suggesting that the modulation of gut microbiota has a direct impact on ocular inflammation (55). Furthermore, a retrospective study from Taiwan has demonstrated a clear association between the progression of SpA and the severity of intestinal inflammation, further underscoring the interconnectedness of gut and systemic immune responses (56).

Under normal conditions, the gut microbiota maintains the integrity of the mucosa and supports the homeostasis of the immune system. However, dysbiosis may alter the intestinal permeability, allowing bacterial products or metabolites to cross the intestinal barrier and enter the bloodstream and tissues, triggering immune responses and inflammation (57). This is also associated with an elevation in the levels of intestinal inflammation markers, such as calprotectin (58). Medications aimed at reducing intestinal permeability may therefore help decrease the occurrence of uveitis. Additionally, dysbiosis of the gut microbiota can activate Paneth cells (PCs), causing release of pro-inflammatory cytokines IL-23 and IL-7, thereby inducing the differentiation and aggregation of type 3 innate lymphoid cells (ILC3) and mucosal-associated invariant T cells (MAIT) (51). This process results in increased levels of IL-17A and IL-22, further compromising the intestinal barrier and perpetuating inflammation (59).

4.3 Infectious factors

In recent years, it has become increasingly evident that infectious factors play a significant role in immune-mediated diseases. Infections of the genitourinary system (e.g. Chlamydia trachomatis) and gastrointestinal system (e.g. Campylobacter, Salmonella, Shigella, Yersinia) can lead to development of ReA (60). In patients with ReA, the presence of microbial antigens in the synovial fluid can be detected, and these patients exhibit a positive clinical response to antibiotic therapy (61), indicating that infectious agents play a direct role in disease onset. Further research has found that the serum levels of antibodies against Gram-negative bacteria are elevated in patients with HLA-B27 positive AAU and SpA, providing additional evidence of microbial involvement in these conditions (62). Helicobacter pylori has also been identified as a potential pathogen associated with the pathogenesis of AAU and SPA (62), although its exact mechanism of action remains unclear. These findings provide strong evidence for the role of microbial triggers in the pathogenesis of AAU and other immune-mediated diseases.

Infectious factors may interact with individual genetic susceptibility to jointly promote the occurrence of disease. Specifically, bacterial antigens can cross-react with self-antigens through molecular simulation to trigger autoimmune reactions (63). This mimicry enables the polypeptides of some microorganisms to resemble the structure of self-antigens, thereby reacting with host immune cells. This mechanism was further supported by a study conducted by bodis (64) and others, which demonstrated that peptides from various microorganisms can bind HLA-B27 molecules, thereby inducing immune responses in the eyes and joints, leading to local inflammation. However, microorganisms have not been detected in the aqueous humor of AAU patients, suggesting that direct microbial invasion of the eye is unlikely. Instead, studies propose that the presence of an unidentified microbiome may regulate the inflammatory response in the eye, contributing to disease pathogenesis (65).

4.4 Dysregulation of immune system

Uveitis is an intraocular inflammatory disease, and the dysregulation of immune system is an important factor in the development of AAU. The pathogenesis of uveitis involves a variety of immune cells and complex molecular mechanisms, including T cells, NK cells, macrophages and dendritic cells. IHLA-B27 plays a pivotal role in this process. It has been shown that HLA-B27 can form homodimers that present on the surface of T cells, monocytes and NK cells. These homodimers bind to killer cell immunoglobulin like receptor (KIR) which are displayed on peripheral blood mononuclear cells and synoviocytes of patients with SPA, thereby activating innate immunity (66). Specifically (67), the expression of killer cell Ig like receptor KIR3DL2 is increased in HLA-B27 AAU patients. This increased KIR3DL2 may stimulate CD4+T cell activation, leading to the secretion of a large number of pro-inflammatory cytokines. B cell infiltration can be observed in ocular tissues of patients with juvenile idiopathic arthritis associated uveitis, suggesting that B cells are also involved in the pathogenesis of uveitis (68). The activation of both the innate and adaptive immune systems leads to the release of various pro0inflammatory cytokines such as TNF-α, IL-17, and IL-23, which mediate the occurrence and progression of SpA through multiple signaling pathways including Wnt, NF-kB, and Jak-STAT (69, 70).

TNF is a 233-amino-acid protein that exists in two forms: transmembrane TNF (tmTNF) and soluble TNF (sTNF). After binding to the two receptors TNFR1 and TNFR2, it can activate multiple signaling pathways such as Wnt and NF-κB, initiating immune responses (71).In a prospective study, it was observed that the levels of TNF-α in the serum and aqueous humor of patients with uveitis were elevated, with a significantly higher concentration of TNF-α in the aqueous humor of HLA-B27 positive patients compared to HLA-B27 negative patients (72).

IL-17 is produced by various immune cells, including helper T cells (Th17), γδT cells, ILC3, and MAIT cells, with Th17 being the primary source (73). IL-17 can promote the recruitment and activation of neutrophils (74), increase angiogenesis and vascular permeability (75), thereby exacerbating the inflammatory response at the site of inflammation. Furthermore, it synergizes with TNF-α to release cytokines such as IL-1, TNF-α, and IL-6, forming a positive feedback loop that further amplifies the inflammatory response (76). This inflammatory cascade reaction may also be triggered by molecular simulations of invading microbial antigens and self-antigens (77).

Antigen presenting cells (APCs) such as dendritic cells (DCs), monocytes and macrophages produce IL-23. IL-23 is a key proinflammatory cytokine composed of P19 and P40 subunits (73). Sustained production of IL-17 is supported by IL-23, which promotes the proliferation and terminal differentiation of Th17 cells (78). Research indicates that the IL-23/IL-17 immunological axis plays a critical role in SpA-associated uveitis (79). Upon binding to cell surface or intracellular receptors of Th17 cells, γδT cells, ILC3, mast cells, and other cells, IL-23 activates the Jak2-STAT3 intracellular signaling pathway, inducing effector cells such as Th17 to produce pro-inflammatory cytokines like IL-17 and IL-22, while also disrupting the barrier function of retinal pigment epithelial cells, thereby promoting the occurrence and progression of uveitis (80),Therefore, modulating the activity of the IL-23/IL-17 immunological axis may offer a potential therapeutic strategy for the treatment of SpA-associated uveitis.

5 Advancements in treatment strategies

The choice of initial treatment for uveitis depends on many factors, including the degree of inflammation, laterality, anatomical location (anterior, middle, posterior uveitis), and the presence and extent of systemic disease (81). Current treatment strategies generally fall into two categories: local treatment and systemic treatment. A stepwise approach is typically employed, starting with less intensive measures and escalating to more powerful interventions as needed. The primary goals are to control inflammation, prevent recurrence, and restore or preserve vision.

5.1 Conventional treatment plan

The first-line treatment for most acute attacks of uveitis typically involves the local use of corticosteroid eye drops or ointments combined with dilators. These treatments rapidly control inflammation of the ocular surface and anterior chamber, reduce ciliary muscle spasm, prevent the formation of posterior adhesions, and offer a cost-effective therapeutic profile. At the beginning of treatment, patients are advised to apply the eye drops frequently (every half an hour to an hour), then gradually reduce the frequency of eye drops to 3–5 times a day for 1–2 weeks. The dosage is then tapered slowly to avoid the “rebound phenomenon” caused by stopping treatment too quickly (81). When choosing local eye drops, consideration should be given to their anti-inflammatory efficacy and permeability in the anterior chamber (82). If the control of anterior chamber inflammation is poor or if the uveitis involves the middle, posterior, or the entire eye, additional interventions may be necessary. These include periocular (lateral or subconjunctival) or intravitreal corticosteroid injections (83), which can supplement eye drop treatment. Studies indicate that intravitreal injection of corticosteroids are effectively against uveitic macular edema (84).

Systemic treatment includes hormone, nonsteroidal anti-inflammatory drugs (NSAIDs), disease-modifying antirheumatic drugs (DMARDs) and immunosuppressant therapy. When local treatment is ineffective or in case of refractory, chronic, posterior, and/or bilateral uveitis, systemic corticosteroid therapy is required. Treatment typically starts at an oral dose of 1 mg/kg per day and gradually decreasing over a course of 6–12 weeks (85). Although effective against both ocular and axial/peripheral joint inflammation, the long-term use of systemic corticosteroids can lead to adverse effects such as elevated blood sugar, systemic hypertension, decreased bone density, depression, and increased body weight, which limit their prolonged use (86). To mitigate these risks, the current recommended approach is to prioritize corticosteroids for initial inflammation control and transition to alternative treatment methods, such as immunosuppressants or biologics, to minimize long-term corticosteroid exposure (87).

NSAIDs are effective in controlling inflammation and are commonly used to manage postoperative inflammation and macular edema following cataract surgery (88). A study by Fiorelli et al. (89)indicated that oral NSAIDs may serve as a therapeutic option for patients with recurrent acute anterior uveitis, offering a potential transitional strategy between local therapy and long-term immunosuppression. However, the definitive efficacy of this approach requires validation through more high-quality studies. Additionally, long-term use of NSAIDs carries significant risks, including an increased incidence of gastrointestinal bleeding and perforation. Given the unclear therapeutic benefits and definite safety concerns, NSAIDs are not currently recommended for the routine management of SpA-associated uveitis.

DMARDs are not first-line but can be used as steroid-sparing therapy. Among them, sulfasalazine (SSZ) is favored for its safety and cost, and is effective mainly against peripheral joint involvement in SpA. In a 3-year prospective study, SSZ significantly reduced the frequency of recurrent AAU flare-ups (90). A recent study also confirmed that SSZ significantly and sustainably reduced the recurrence rate of AAU in NIU patients over a three-year period (91). Meanwhile, adverse effects were observed in approximately 11-40% of patients receiving SSZ, primarily including nausea, vomiting, diarrhea, headache, and loss of appetite (92). The overall safety profile of SSZ is favorable, with most adverse effects being mild. However, due to the risk of rare but severe reactions, routine safety monitoring is essential. Methotrexate (MTX) and azathioprine (AZP) are not routinely recommended for SpA-associated AAU due to insufficient evidence. Although MTX reduces uveitis relapse and complications, such as cataracts, glaucoma, and macular edema (93), and AZP has been proven to be an effective steroid-sparing therapy for chronic non-inflammatory eye diseases (94), the existing studies in SpA-AAU are limited by small samples and low evidence grade. Their definitive efficacy and safety require confirmation from large-scale, randomized controlled trials.

5.2 Progress in targeted therapy with biologics

In recent years, biologics have received increasing attention and shown promising therapeutic prospects. By precisely targeting specific inflammatory mediators or signaling pathways, they can rapidly control inflammation, reduce uveitis relapses, and minimize the side effects associated with traditional treatments, making therapy safer and more effective. Consequently, biological agents have emerged as a vital option in the clinical management of uveitis (Table 2).

Table 2
www.frontiersin.org

Table 2. Targeted therapeutics for spondyloarthritis-associated uveitis.

As discussed earlier in the pathogenesis section, the pro-inflammatory cytokine TNF-α is a key factor in the development of uveitis, and its levels are upregulated in both the patient’s aqueous humor and serum. Numerous studies have confirmed that tumor necrosis factor inhibitors (TNFi) have significant therapeutic effects in treating SpA-associated uveitis. Currently, five different types of TNFi are available: monoclonal antibodies such as adalimumab (ADA), Infliximab (IFX), Golimumab (GOL), and Certolizumab pegol (CZP), as well as fusion protein etanercept (ETA).

While these agents are often used off-label for the treatment of AAU, ADA is currently the only TNFi approved for the treatment of non-infectious intermediate, posterior, or panuveitis. The VISUAL I trial included 217 participants with active non-infectious intermediate, posterior, or panuveitis. The findings demonstrated that adalimumab (40 mg administered biweekly following a loading dose) significantly decreased the risk of visual impairment in comparison to placebo (95). The subsequent VISUAL II trial in patients with quiescent, steroid-dependent disease showed a significantly lower treatment failure rate with ADA (39%) than with placebo (55%) (96). These findings were further corroborated by a subsequent multicenter study, which confirmed that long-term treatment with ADA promotes and sustains disease quiescence while also reducing dependence on systemic corticosteroids (97). Additionally, a study by Rudwaleit et al. (98) showed that ADA can reduce the attacks of SpA-associated uveitis by 51%. A meta-analysis supports that ADA can decrease the frequency of uveitis attacks, reduce recurrences, and lower the use of corticosteroids (99). A subsequent meta-analysis by Li et al. (100) further confirmed the efficacy and safety of ADA. Therefore, ADA is recommended for patients with NIU who are corticosteroid-dependent or have an inadequate response to conventional therapy. Recent studies also recommend routine ADA monitoring during adalimumab treatment to enable the maintenance of consistent therapeutic drug levels (101).

Liu et al. (102) reported that INF and ADA have similar effects in terms of therapeutic efficacy and corticosteroid-sparing. A recent retrospective nationwide cohort study by Kwon et al. (103) indicated that ADA and IFX are comparable, both effectively reducing the recurrence and incidence of AAU, and both are significantly superior to ETA. In contrast, ETA has even been reported to be ineffective and potentially associated with an increased risk of uveitis flares in some cases (104). Therefore, for patients with SpA-associated recurrent, refractory uveitis, or those for whom conventional treatment has failed, ADA or IFX are recommended as the preferred choice, while ETA should be avoided (105).

Results from a prospective study by Van Bentum et al. (106)demonstrated that GOL significantly reduced the incidence of NIU in AS patients. Other studies have also suggested that GOL is effective for refractory NIU (107). The VIEW study demonstrated that treatment with certolizumab pegol significantly reduced the incidence of AAU flares in patients with axial spondyloarthritis who have a history of AAU, suggesting it as a viable treatment option (108). However, robust evidence supporting the comparative efficacy and safety of GOL and CZP is still lacking, and further studies are needed to provide a more substantial evidence base. In summary, the role of TNFi in managing SpA-associated uveitis is considerable. Nevertheless, in clinical practice, serious attention must be paid to their potential side effects and adverse reactions.

IL-17A inhibitors (IL-17Ai) work by targeting IL-17A to reduce inflammation and improve clinical symptoms. However, their role in preventing or reducing AAU recurrence is not yet clear. Secukinumab, a widely used IL-17Ai, has been trialed for its efficacy in uveitis. A nationwide observational cohort study of SpA patients suggested that those using secukinumab may have higher risk of AAU compared with monoclonal TNFi (109). Additionally, the results of three randomized controlled trials suggested that secukinumab failed to meet its primary efficacy endpoints in the treatment of non-infectious uveitis, indicating that its efficacy in this area may be limited (110). In a post-hoc analysis of 11 trials, secukinumab demonstrated a numerically lower AAU incidence than placebo in axSpA (1.29 vs. 1.72), though not significant (111). In summary, IL-17A inhibitors play a limited role in preventing spondyloarthritis-associated uveitis and are less effective than TNFi. Consequently, they are not recommended as the first-choice biologic treatment for patients with a history of recurrent AAU. Bimekizumab (BKZ), a dual IL-17A/F inhibitor, has been shown to significantly reduce the rate of uveitis recurrence, indicating its potential therapeutic value (112). However, its efficacy and safety necessitate further validation.

Targeting the IL-12/IL-23 pathway with biologic agents is another potential therapeutic strategy for patients who experience failure or intolerance to TNFi and/or IL-17A inhibitors (18). Ustekinumab is a monoclonal antibody that blocks the IL-12/IL-23 pathway, primarily used in the treatment of autoimmune diseases such as psoriatic arthritis and Crohn’s disease. Studies have demonstrated its therapeutic efficacy on SpA-associated uveitis, such as psoriatic arthritis related uveitis (113) and inflammatory bowel disease-related uveitis (114). However, these conclusions are predominantly based on small-sample or retrospective studies and lack validation from randomized controlled trials. Therefore, the therapeutic role of ustekinumab in SpA-associated uveitis requires confirmation through more high-quality studies.

Janus kinase inhibitors (JAKi) inhibit intracellular signaling by blocking the JAK pathway, thereby reducing the production of inflammatory cytokines and effectively mitigating the inflammatory response. Tofacitinib is a representative drug of this class, selectively inhibiting the activity of JAK1 and JAK3 (115). It has been reported that tofacitinib is effective in controlling refractory ocular inflammation after failed steroid-sparing treatment (116). Studies have shown that tofacitinib can also treat juvenile idiopathic arthritis-related uveitis without significant side effects (117). The phase 2 HUMBOLDT trial (118) found filgotinib significantly reduced treatment failure by 30.1% vs placebo in active non-infectious uveitis, though adverse events were higher. Furthermore, compared to TNFi or IL-17Ai, the clinical cohort for JAKi in SpA-associated uveitis remains small, with limited long-term safety data. Thus, further evaluation through more extensive studies is urgently needed. As clinical trials progress and treatment experience accumulates, JAKi hold promise as a novel therapeutic strategy for the management of SpA-associated uveitis.

To achieve optimal management, ophthalmologists and rheumatologists need to collaborate closely in key areas such as diagnosis, early referral, decision-making, and follow-up care. Additionally, the personalization of treatment plans is crucial, with strategies tailored to the specific conditions of each patient. Through this individualized and comprehensive treatment approach, the negative impact of uveitis on patients’ vision and quality of life can be minimized to the greatest extent.

6 Summary and outlook

SpA-associated uveitis is a significant extra-articular manifestation that greatly impact the visual health of patients. Early diagnosis and timely treatment are crucial to prevent serious complications and protect vision. Detailed ophthalmic examinations and comprehensive systemic assessments should be conducted for SpA patients, and vigilance should be maintained regarding the potential impact of associated risk factors. Future research should focus on further understanding the pathogenesis of SpA-associated uveitis in order to develop more effective prevention and treatment strategies. While most patients have a good response to local corticosteroid therapy, recurrent or refractory patients often require a more comprehensive treatment approach, including systemic corticosteroids, NSAIDs, DMARDS, or biologics.

Biological agents, especially those targeting specific inflammatory pathways, have shown unique advantages in long-term control of joint and ocular inflammation. However, their efficacy and safety need to be further validated through extensive research. JAK inhibitors, as an emerging treatment option, provide a new treatment approach for SpA-associated uveitis. Although their efficacy and safety remain under investigation, preliminary studies highlight their potential in regulating immune responses and reducing inflammation.

In this review, we discussed in detail the clinical characteristics and popular hypothesis for the pathogenesis of SpA-associated uveitis, as well as provided an overview of current treatment approaches, including novel treatment strategies targeting immune components. Effective management of SpA-associated uveitis requires close collaboration between ophthalmologists and rheumatologists, providing patients with personalized and comprehensive treatment strategies through multidisciplinary teams. This collaborative approach not only improve treatment effectiveness, but also reduce risk of missed or incorrect diagnosis, thereby minimizing the impact of the disease on patients’ vision and quality of life.

Author contributions

S-YP: Conceptualization, Writing – original draft. LX: Data curation, Writing – original draft. ZC: Formal analysis, Writing – original draft, Visualization. ZY: Conceptualization, Writing – review & editing. R-JC: Conceptualization, Supervision, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

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. Sieper J and Poddubnyy D. Axial spondyloarthritis. Lancet. (2017) 390:73–84. doi: 10.1016/S0140-6736(16)31591-4

PubMed Abstract | Crossref Full Text | Google Scholar

2. Robinson PC, Van Der Linden S, Khan MA, and Taylor WJ. Axial spondyloarthritis: concept, construct, classification and implications for therapy. Nat Rev Rheumatol. (2021) 17:109–18. doi: 10.1038/s41584-020-00552-4

PubMed Abstract | Crossref Full Text | Google Scholar

3. Dougados M and Baeten D. Spondyloarthritis. Lancet. (2011) 377:2127–37. doi: 10.1016/S0140-6736(11)60071-8

PubMed Abstract | Crossref Full Text | Google Scholar

4. Stolwijk C, Van Tubergen A, Castillo-Ortiz JD, and Boonen A. Prevalence of extra-articular manifestations in patients with ankylosing spondylitis: a systematic review and meta-analysis. Ann Rheum Dis. (2015) 74:65–73. doi: 10.1136/annrheumdis-2013-203582

PubMed Abstract | Crossref Full Text | Google Scholar

5. Rademacher J, Poddubnyy D, and Pleyer U. Uveitis in spondyloarthritis. Ther Adv Musculoskelet Dis. (2020) 12:1759720x20951733. doi: 10.1177/1759720X20951733

PubMed Abstract | Crossref Full Text | Google Scholar

6. Bittar M, Yong WC, Magrey M, and Khan MA. Worldwide differences in clinical phenotype of axial spondyloarthritis. Curr Rheumatol Rep. (2021) 23:76. doi: 10.1007/s11926-021-01043-5

PubMed Abstract | Crossref Full Text | Google Scholar

7. Yasmin MR, Islam MN, Pannu ZR, Azad MK, and Uddin MSG. Prevalence and risk factors for uveitis in spondyloarthritis. Int J Rheum Dis. (2022) 25:517–22. doi: 10.1111/1756-185X.14303

PubMed Abstract | Crossref Full Text | Google Scholar

8. Maldonado-Ficco H, López-Medina C, Perez-Alamino R, Waimann CA, Maldonado-Cocco JA, Moltó A, et al. Prevalence and incidence of uveitis in patients with spondyloarthritis: the impact of the biologics era. Data from International ASAS-COMOSPA Study. Rheumatol (Oxford). (2024) 64(5):2618–24. doi: 10.1093/rheumatology/keae536

PubMed Abstract | Crossref Full Text | Google Scholar

9. Rudwaleit M, Walscheid K, and Heiligenhaus A. Uveitis in spondyloarthritis. Z Rheumatol. (2017) 76:682–92. doi: 10.1007/s00393-017-0357-6

PubMed Abstract | Crossref Full Text | Google Scholar

10. Abdulla D, Ali Y, Menezo V, and Taylor SRJ. The use of sustained release intravitreal steroid implants in non-infectious uveitis affecting the posterior segment of the eye. Ophthalmol Ther. (2022) 11:479–87. doi: 10.1007/s40123-022-00456-4

PubMed Abstract | Crossref Full Text | Google Scholar

11. Biggioggero M, Crotti C, Becciolini A, Miserocchi E, and Favalli EG. The management of acute anterior uveitis complicating spondyloarthritis: present and future. BioMed Res Int. (2018) 2018:9460187. doi: 10.1155/2018/9460187

PubMed Abstract | Crossref Full Text | Google Scholar

12. Khan MA, Haroon M, and Rosenbaum JT. Acute anterior uveitis and spondyloarthritis: more than meets the eye. Curr Rheumatol Rep. (2015) 17:59. doi: 10.1007/s11926-015-0536-x

PubMed Abstract | Crossref Full Text | Google Scholar

13. Accorinti M, Iannetti L, Liverani M, Caggiano C, and Gilardi M. Clinical features and prognosis of HLA B27-associated acute anterior uveitis in an Italian patient population. Ocul Immunol Inflammation. (2010) 18:91–6. doi: 10.3109/09273941003597268

PubMed Abstract | Crossref Full Text | Google Scholar

14. Wakefield D, Clarke D, and Mccluskey P. Recent developments in HLA B27 anterior uveitis. Front Immunol. (2020) 11:608134. doi: 10.3389/fimmu.2020.608134

PubMed Abstract | Crossref Full Text | Google Scholar

15. Gritz DC, Schwaber EJ, and Wong IG. Complications of uveitis: the northern california epidemiology of uveitis study. Ocul Immunol Inflammation. (2018) 26:584–94. doi: 10.1080/09273948.2016.1247174

PubMed Abstract | Crossref Full Text | Google Scholar

16. Pandey A and Ravindran V. Ocular manifestations of spondyloarthritis. Mediterr J Rheumatol. (2023) 34:24–9. doi: 10.31138/mjr.34.1.24

PubMed Abstract | Crossref Full Text | Google Scholar

17. Loh AR and Acharya NR. Incidence rates and risk factors for ocular complications and vision loss in HLA-B27-associated uveitis. Am J Ophthalmol. (2010) 150:534–542.e2. doi: 10.1016/j.ajo.2010.04.031

PubMed Abstract | Crossref Full Text | Google Scholar

18. Jacquot R, Kodjikian L, Chapurlat R, and Sève P. Targeted therapies for uveitis in spondyloarthritis: A narrative review. Joint Bone Spine. (2024) 91:105697. doi: 10.1016/j.jbspin.2024.105697

PubMed Abstract | Crossref Full Text | Google Scholar

19. Rosenbaum JT. Uveitis in spondyloarthritis including psoriatic arthritis, ankylosing spondylitis, and inflammatory bowel disease. Clin Rheumatol. (2015) 34:999–1002. doi: 10.1007/s10067-015-2960-8

PubMed Abstract | Crossref Full Text | Google Scholar

20. Yaşar Bilge N, Kalyoncu U, Atagündüz P, Dalkılıç E, Pehlivan Y, Küçükşahin O, et al. Uveitis-related factors in patients with spondyloarthritis: TReasure real-life results. Am J Ophthalmol. (2021) 228:58–64. doi: 10.1016/j.ajo.2021.03.026

PubMed Abstract | Crossref Full Text | Google Scholar

21. Ma KS, Lee YH, Lin CJ, Shih PC, and Wei JC. Management of extra-articular manifestations in spondyloarthritis. Int J Rheum Dis. (2023) 26:183–6. doi: 10.1111/1756-185X.14485

PubMed Abstract | Crossref Full Text | Google Scholar

22. Chao YJ, Hung JH, Lin CP, Kuo HK, Chen SN, Hwang YS, et al. Diagnosis, treatment, and prevention of noninfectious acute anterior uveitis with or without human leukocyte antigen B27 in adults - expert consensus in Taiwan. Ocul Immunol Inflammation. (2024) 32:226–33. doi: 10.1080/09273948.2023.2165113

PubMed Abstract | Crossref Full Text | Google Scholar

23. Hwang DK, Hung JH, Chang YC, Chen CL, Chen SN, Cheng CK, et al. Step-wise diagnostic approach for patients with uveitis - Experts consensus in Taiwan. J Microbiol Immunol Infect. (2022) 55:573–80. doi: 10.1016/j.jmii.2022.02.003

PubMed Abstract | Crossref Full Text | Google Scholar

24. Group S O U N S W. Classification criteria for spondyloarthritis/HLA-B27-associated anterior uveitis. Am J Ophthalmol. (2021) 228:117–25. doi: 10.1016/j.ajo.2021.03.049

PubMed Abstract | Crossref Full Text | Google Scholar

25. Haroon M, O'rourke M, Ramasamy P, Murphy CC, and Fitzgerald O. A novel evidence-based detection of undiagnosed spondyloarthritis in patients presenting with acute anterior uveitis: the DUET (Dublin Uveitis Evaluation Tool). Ann Rheum Dis. (2015) 74:1990–5. doi: 10.1136/annrheumdis-2014-205358

PubMed Abstract | Crossref Full Text | Google Scholar

26. Mitulescu CT, Popescu CC, Oprea CL, Predețeanu D, Istrate S, Ciuluvică R, et al. A referable clinical pattern of spondyloarthritis-associated uveitis. Rom J Ophthalmol. (2018) 62:155–61. doi: 10.22336/rjo.2018.23

PubMed Abstract | Crossref Full Text | Google Scholar

27. Khan MA. HLA-B27 and its pathogenic role. J Clin Rheumatol. (2008) 14:50–2. doi: 10.1097/RHU.0b013e3181637a38

PubMed Abstract | Crossref Full Text | Google Scholar

28. Robinson PC, Leo PJ, Pointon JJ, Harris J, Cremin K, Bradbury LA, et al. The genetic associations of acute anterior uveitis and their overlap with the genetics of ankylosing spondylitis. Genes Immun. (2016) 17:46–51. doi: 10.1038/gene.2015.49

PubMed Abstract | Crossref Full Text | Google Scholar

29. Zeboulon N, Dougados M, and Gossec L. Prevalence and characteristics of uveitis in the spondyloarthropathies: a systematic literature review. Ann Rheum Dis. (2008) 67:955–9. doi: 10.1136/ard.2007.075754

PubMed Abstract | Crossref Full Text | Google Scholar

30. Karaconji T, Maconochie Z, and Mccluskey P. Acute anterior uveitis in Sydney. Ocul Immunol Inflammation. (2013) 21:108–14. doi: 10.3109/09273948.2012.745882

PubMed Abstract | Crossref Full Text | Google Scholar

31. Navid F, Chen L, Bowness P, and Colbert RA. HLA-B27 and spondyloarthritis: at the crossroads of innate and adaptive immunity. Nat Rev Rheumatol. (2025) 21:77–87. doi: 10.1038/s41584-024-01189-3

PubMed Abstract | Crossref Full Text | Google Scholar

32. Hermann E, Yu DT, Meyer Zum Büschenfelde KH, and Fleischer B. HLA-B27-restricted CD8 T cells derived from synovial fluids of patients with reactive arthritis and ankylosing spondylitis. Lancet. (1993) 342:646–50. doi: 10.1016/0140-6736(93)91760-J

PubMed Abstract | Crossref Full Text | Google Scholar

33. Yang X, Garner LI, Zvyagin IV, Paley MA, Komech EA, Jude KM, et al. Autoimmunity-associated T cell receptors recognize HLA-B*27-bound peptides. Nature. (2022) 612:771–7. doi: 10.1038/s41586-022-05501-7

PubMed Abstract | Crossref Full Text | Google Scholar

34. Khan MA. An update on the genetic polymorphism of HLA-B*27 with 213 alleles encompassing 160 subtypes (and still counting). Curr Rheumatol Rep. (2017) 19:9. doi: 10.1007/s11926-017-0640-1

PubMed Abstract | Crossref Full Text | Google Scholar

35. Cauli A, Shaw J, Giles J, Hatano H, Rysnik O, Payeli S, et al. The arthritis-associated HLA-B*27:05 allele forms more cell surface B27 dimer and free heavy chain ligands for KIR3DL2 than HLA-B*27:09. Rheumatol (Oxford). (2013) 52:1952–62. doi: 10.1093/rheumatology/ket219

PubMed Abstract | Crossref Full Text | Google Scholar

36. Schittenhelm RB, Sian TC, Wilmann PG, Dudek NL, and Purcell AW. Revisiting the arthritogenic peptide theory: quantitative not qualitative changes in the peptide repertoire of HLA-B27 allotypes. Arthritis Rheumatol. (2015) 67:702–13. doi: 10.1002/art.38963

PubMed Abstract | Crossref Full Text | Google Scholar

37. Schröder M and Kaufman RJ. ER stress and the unfolded protein response. Mutat Res. (2005) 569:29–63. doi: 10.1016/j.mrfmmm.2004.06.056

PubMed Abstract | Crossref Full Text | Google Scholar

38. Colbert RA. HLA-B27 misfolding: a solution to the spondyloarthropathy conundrum? Mol Med Today. (2000) 6:224–30. doi: 10.1016/s1357-4310(00)01699-3

PubMed Abstract | Crossref Full Text | Google Scholar

39. Colbert RA, Navid F, and Gill T. The role of HLA-B*27 in spondyloarthritis. Best Pract Res Clin Rheumatol. (2017) 31:797–815. doi: 10.1016/j.berh.2018.07.012

PubMed Abstract | Crossref Full Text | Google Scholar

40. Allen RL, Bowness P, and Mcmichael A. The role of HLA-B27 in spondyloarthritis. Immunogenetics. (1999) 50:220–7. doi: 10.1007/s002510050596

PubMed Abstract | Crossref Full Text | Google Scholar

41. Kollnberger S, Bird L, Sun MY, Retiere C, Braud VM, Mcmichael A, et al. Cell-surface expression and immune receptor recognition of HLA-B27 homodimers. Arthritis Rheum. (2002) 46:2972–82. doi: 10.1002/art.10605

PubMed Abstract | Crossref Full Text | Google Scholar

42. Bowness P. HLA-B27. Annu Rev Immunol. (2015) 33:29–48. doi: 10.1146/annurev-immunol-032414-112110

PubMed Abstract | Crossref Full Text | Google Scholar

43. Wong-Baeza I, Ridley A, Shaw J, Hatano H, Rysnik O, Mchugh K, et al. KIR3DL2 binds to HLA-B27 dimers and free H chains more strongly than other HLA class I and promotes the expansion of T cells in ankylosing spondylitis. J Immunol. (2013) 190:3216–24. doi: 10.4049/jimmunol.1202926

PubMed Abstract | Crossref Full Text | Google Scholar

44. Jiao YL, Zhang BC, You L, Li JF, Zhang J, Ma CY, et al. Polymorphisms of KIR gene and HLA-C alleles: possible association with susceptibility to HLA-B27-positive patients with ankylosing spondylitis. J Clin Immunol. (2010) 30:840–4. doi: 10.1007/s10875-010-9444-z

PubMed Abstract | Crossref Full Text | Google Scholar

45. Vendelbosch S, Heslinga SC, John M, Van Leeuwen K, Geissler J, De Boer M, et al. Study on the protective effect of the KIR3DL1 gene in ankylosing spondylitis. Arthritis Rheumatol. (2015) 67:2957–65. doi: 10.1002/art.39288

PubMed Abstract | Crossref Full Text | Google Scholar

46. Queiro R, Torre JC, Belzunegui J, González C, De Dios JR, Unanue F, et al. Clinical features and predictive factors in psoriatic arthritis-related uveitis. Semin Arthritis Rheum. (2002) 31:264–70. doi: 10.1053/sarh.2002.28798

PubMed Abstract | Crossref Full Text | Google Scholar

47. Robinson PC, Claushuis TA, Cortes A, Martin TM, Evans DM, Leo P, et al. Genetic dissection of acute anterior uveitis reveals similarities and differences in associations observed with ankylosing spondylitis. Arthritis Rheumatol. (2015) 67:140–51. doi: 10.1002/art.38873

PubMed Abstract | Crossref Full Text | Google Scholar

48. Huang XF, Li Z, De Guzman E, Robinson P, Gensler L, Ward MM, et al. Genomewide association study of acute anterior uveitis identifies new susceptibility loci. Invest Ophthalmol Vis Sci. (2020) 61:3. doi: 10.1167/iovs.61.6.3

PubMed Abstract | Crossref Full Text | Google Scholar

49. López De Castro JA. How ERAP1 and ERAP2 shape the peptidomes of disease-associated MHC-I proteins. Front Immunol. (2018) 9:2463. doi: 10.3389/fimmu.2018.02463

PubMed Abstract | Crossref Full Text | Google Scholar

50. Costello ME, Ciccia F, Willner D, Warrington N, Robinson PC, Gardiner B, et al. Brief report: intestinal dysbiosis in ankylosing spondylitis. Arthritis Rheumatol. (2015) 67:686–91. doi: 10.1002/art.38967

PubMed Abstract | Crossref Full Text | Google Scholar

51. Song ZY, Yuan D, and Zhang SX. Role of the microbiome and its metabolites in ankylosing spondylitis. Front Immunol. (2022) 13:1010572. doi: 10.3389/fimmu.2022.1010572

PubMed Abstract | Crossref Full Text | Google Scholar

52. Kalyana Chakravarthy S, Jayasudha R, Sai Prashanthi G, Ali MH, Sharma S, Tyagi M, et al. Dysbiosis in the gut bacterial microbiome of patients with uveitis, an inflammatory disease of the eye. Indian J Microbiol. (2018) 58:457–69. doi: 10.1007/s12088-018-0746-9

PubMed Abstract | Crossref Full Text | Google Scholar

53. Essex M, Rios Rodriguez V, Rademacher J, Proft F, Löber U, Markó L, et al. Shared and distinct gut microbiota in spondyloarthritis, acute anterior uveitis, and crohn's disease. Arthritis Rheumatol. (2024) 76:48–58. doi: 10.1002/art.42658

PubMed Abstract | Crossref Full Text | Google Scholar

54. Taurog JD, Richardson JA, Croft JT, Simmons WA, Zhou M, Fernández-Sueiro JL, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med. (1994) 180:2359–64. doi: 10.1084/jem.180.6.2359

PubMed Abstract | Crossref Full Text | Google Scholar

55. Nakamura YK, Metea C, Karstens L, Asquith M, Gruner H, Moscibrocki C, et al. Gut microbial alterations associated with protection from autoimmune uveitis. Invest Ophthalmol Vis Sci. (2016) 57:3747–58. doi: 10.1167/iovs.16-19733

PubMed Abstract | Crossref Full Text | Google Scholar

56. Feng HY, Chan CH, Chu YC, Qu XM, Wang YH, Wei JC, et al. Patients with ankylosing spondylitis have high risk of irritable bowel syndrome: a long-term nationwide population-based cohort study. Postgrad Med. (2022) 134:290–6. doi: 10.1080/00325481.2022.2041338

PubMed Abstract | Crossref Full Text | Google Scholar

57. Rosenbaum JT and Asquith M. The microbiome and HLA-B27-associated acute anterior uveitis. Nat Rev Rheumatol. (2018) 14:704–13. doi: 10.1038/s41584-018-0097-2

PubMed Abstract | Crossref Full Text | Google Scholar

58. Gazim CC, Borba AA, Castro GKP, Simioni J, Gehlen ML, Ramos Júnior O, et al. Fecal calprotectin levels in acute anterior uveitis in patients with spondyloarthritis. Arq Bras Oftalmol. (2023) 86:33–7. doi: 10.5935/0004-2749.20230008

PubMed Abstract | Crossref Full Text | Google Scholar

59. Gracey E, Vereecke L, Mcgovern D, Fröhling M, Schett G, Danese S, et al. Revisiting the gut-joint axis: links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol. (2020) 16:415–33. doi: 10.1038/s41584-020-0454-9

PubMed Abstract | Crossref Full Text | Google Scholar

60. Parthasarathy R, Santiago F, Mccluskey P, Kaakoush NO, Tedla N, Wakefield D, et al. The microbiome in HLA-B27-associated disease: implications for acute anterior uveitis and recommendations for future studies. Trends Microbiol. (2023) 31:142–58. doi: 10.1016/j.tim.2022.08.008

PubMed Abstract | Crossref Full Text | Google Scholar

61. Colmegna I, Cuchacovich R, and Espinoza LR. HLA-B27-associated reactive arthritis: pathogenetic and clinical considerations. Clin Microbiol Rev. (2004) 17:348–69. doi: 10.1128/CMR.17.2.348-369.2004

PubMed Abstract | Crossref Full Text | Google Scholar

62. Otasevic L, Zlatanovic G, Stanojevic-Paovic A, Miljkovic-Selimovic B, Dinic M, Djordjevic-Jocic J, et al. Helicobacter pylori: an underestimated factor in acute anterior uveitis and spondyloarthropathies? Ophthalmologica. (2007) 221:6–13. doi: 10.1159/000096515

PubMed Abstract | Crossref Full Text | Google Scholar

63. Rojas M, Restrepo-Jiménez P, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, et al. Molecular mimicry and autoimmunity. J Autoimmun. (2018) 95:100–23. doi: 10.1016/j.jaut.2018.10.012

PubMed Abstract | Crossref Full Text | Google Scholar

64. Bodis G, Toth V, and Schwarting A. Role of human leukocyte antigens (HLA) in autoimmune diseases. Rheumatol Ther. (2018) 5:5–20. doi: 10.1007/s40744-018-0100-z

PubMed Abstract | Crossref Full Text | Google Scholar

65. Wen X, Hu X, Miao L, Ge X, Deng Y, Bible PW, et al. Epigenetics, microbiota, and intraocular inflammation: New paradigms of immune regulation in the eye. Prog Retin Eye Res. (2018) 64:84–95. doi: 10.1016/j.preteyeres.2018.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

66. Kuon W, Holzhütter HG, Appel H, Grolms M, Kollnberger S, Traeder A, et al. Identification of HLA-B27-restricted peptides from the Chlamydia trachomatis proteome with possible relevance to HLA-B27-associated diseases. J Immunol. (2001) 167:4738–46. doi: 10.4049/jimmunol.167.8.4738

PubMed Abstract | Crossref Full Text | Google Scholar

67. Bowness P, Ridley A, Shaw J, Chan AT, Wong-Baeza I, Fleming M, et al. Th17 cells expressing KIR3DL2+ and responsive to HLA-B27 homodimers are increased in ankylosing spondylitis. J Immunol. (2011) 186:2672–80. doi: 10.4049/jimmunol.1002653

PubMed Abstract | Crossref Full Text | Google Scholar

68. Zhu L, Chen B, and Su W. A review of the various roles and participation levels of B-cells in non-infectious uveitis. Front Immunol. (2021) 12:676046. doi: 10.3389/fimmu.2021.676046

PubMed Abstract | Crossref Full Text | Google Scholar

69. Egwuagu CE, Alhakeem SA, and Mbanefo EC. Uveitis: molecular pathogenesis and emerging therapies. Front Immunol. (2021) 12:623725. doi: 10.3389/fimmu.2021.623725

PubMed Abstract | Crossref Full Text | Google Scholar

70. Su Y, Tao T, Liu X, and Su W. JAK-STAT signaling pathway in non-infectious uveitis. Biochem Pharmacol. (2022) 204:115236. doi: 10.1016/j.bcp.2022.115236

PubMed Abstract | Crossref Full Text | Google Scholar

71. Hreggvidsdottir HS, Noordenbos T, and Baeten DL. Inflammatory pathways in spondyloarthritis. Mol Immunol. (2014) 57:28–37. doi: 10.1016/j.molimm.2013.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

72. Pérez-Guijo V, Santos-Lacomba M, Sánchez-Hernández M, Castro-Villegas Mdel C, Gallardo-Galera JM, Collantes-Estévez E, et al. Tumour necrosis factor-alpha levels in aqueous humour and serum from patients with uveitis: the involvement of HLA-B27. Curr Med Res Opin. (2004) 20:155–7. doi: 10.1185/030079903125002847

PubMed Abstract | Crossref Full Text | Google Scholar

73. Tsukazaki H and Kaito T. The role of the IL-23/IL-17 pathway in the pathogenesis of spondyloarthritis. Int J Mol Sci. (2020) 21(17):6401. doi: 10.3390/ijms21176401

PubMed Abstract | Crossref Full Text | Google Scholar

74. Dragon S, Saffar AS, Shan L, and Gounni AS. IL-17 attenuates the anti-apoptotic effects of GM-CSF in human neutrophils. Mol Immunol. (2008) 45:160–8. doi: 10.1016/j.molimm.2007.04.027

PubMed Abstract | Crossref Full Text | Google Scholar

75. Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. (2003) 101:2620–7. doi: 10.1182/blood-2002-05-1461

PubMed Abstract | Crossref Full Text | Google Scholar

76. Ruddy MJ, Wong GC, Liu XK, Yamamoto H, Kasayama S, Kirkwood KL, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem. (2004) 279:2559–67. doi: 10.1074/jbc.M308809200

PubMed Abstract | Crossref Full Text | Google Scholar

77. Lin P, Suhler EB, and Rosenbaum JT. The future of uveitis treatment. Ophthalmology. (2014) 121:365–76. doi: 10.1016/j.ophtha.2013.08.029

PubMed Abstract | Crossref Full Text | Google Scholar

78. Mcgeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, Blumenschein WM, et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol. (2009) 10:314–24. doi: 10.1038/ni.1698

PubMed Abstract | Crossref Full Text | Google Scholar

79. Schinocca C, Rizzo C, Fasano S, Grasso G, La Barbera L, Ciccia F, 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

PubMed Abstract | Crossref Full Text | Google Scholar

80. Wu X, Tao M, Zhu L, Zhang T, and Zhang M. Pathogenesis and current therapies for non-infectious uveitis. Clin Exp Med. (2023) 23:1089–106. doi: 10.1007/s10238-022-00954-6

PubMed Abstract | Crossref Full Text | Google Scholar

81. Balasubramaniam B, Chong YJ, Azzopardi M, Logeswaran A, and Denniston AK. Topical anti-inflammatory agents for non-infectious uveitis: current treatment and perspectives. J Inflammation Res. (2022) 15:6439–51. doi: 10.2147/JIR.S288294

PubMed Abstract | Crossref Full Text | Google Scholar

82. Werkl P, Rademacher J, and Pleyer U. HLA-B27-positive anterior uveitis: Clinical aspects, diagnostics, interdisciplinary management, and treatment. Ophthalmologie. (2024) 121:12–22. doi: 10.1007/s00347-023-01960-z

PubMed Abstract | Crossref Full Text | Google Scholar

83. Gaudio PA. A review of evidence guiding the use of corticosteroids in the treatment of intraocular inflammation. Ocul Immunol Inflammation. (2004) 12:169–92. doi: 10.1080/092739490500192

PubMed Abstract | Crossref Full Text | Google Scholar

84. Fung AT, Tran T, Lim LL, Samarawickrama C, Arnold J, Gillies M, et al. Local delivery of corticosteroids in clinical ophthalmology: A review. Clin Exp Ophthalmol. (2020) 48:366–401. doi: 10.1111/ceo.13702

PubMed Abstract | Crossref Full Text | Google Scholar

85. Touhami S, Diwo E, Sève P, Trad S, Bielefeld P, Sène D, et al. Expert opinion on the use of biological therapy in non-infectious uveitis. Expert Opin Biol Ther. (2019) 19:477–90. doi: 10.1080/14712598.2019.1595578

PubMed Abstract | Crossref Full Text | Google Scholar

86. Thorne JE, Sugar EA, Holbrook JT, Burke AE, Altaweel MM, Vitale AT, et al. Periocular Triamcinolone vs. Intravitreal Triamcinolone vs. Intravitreal Dexamethasone Implant for the Treatment of Uveitic Macular Edema: The PeriOcular vs. INTravitreal corticosteroids for uveitic macular edema (POINT) Trial. Ophthalmology. (2019) 126:283–95. doi: 10.1016/j.ophtha.2018.08.021

PubMed Abstract | Crossref Full Text | Google Scholar

87. Carnahan MC and Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. Curr Opin Ophthalmol. (2000) 11:478–83. doi: 10.1097/00055735-200012000-00016

PubMed Abstract | Crossref Full Text | Google Scholar

88. Burkholder BM and Jabs DA. Uveitis for the non-ophthalmologist. BMJ. (2021) 372:m4979. doi: 10.1136/bmj.m4979

PubMed Abstract | Crossref Full Text | Google Scholar

89. Fiorelli VM, Bhat P, and Foster CS. Nonsteroidal anti-inflammatory therapy and recurrent acute anterior uveitis. Ocul Immunol Inflammation. (2010) 18:116–20. doi: 10.3109/09273941003587558

PubMed Abstract | Crossref Full Text | Google Scholar

90. Muñoz-Fernández S, Hidalgo V, Fernández-Melón J, Schlincker A, Bonilla G, Ruiz-Sancho D, et al. Sulfasalazine reduces the number of flares of acute anterior uveitis over a one-year period. J Rheumatol. (2003) 30:1277–9.

PubMed Abstract | Google Scholar

91. Avello-Gorostidi A, Hernanz I, Robles-Barrena I, Olivas-Vergara O, Borges PE, Arconada-López C, et al. Efficacy of sulfasalazine on recurrent acute anterior uveitis: a three-year follow-up. Int Ophthalmol. (2025) 45:36. doi: 10.1007/s10792-024-03405-1

PubMed Abstract | Crossref Full Text | Google Scholar

92. Rogler G. Gastrointestinal and liver adverse effects of drugs used for treating IBD. Best Pract Res Clin Gastroenterol. (2010) 24:157–65. doi: 10.1016/j.bpg.2009.10.011

PubMed Abstract | Crossref Full Text | Google Scholar

93. Zu Hoerste MM, Walscheid K, Tappeiner C, Zurek-Imhoff B, Heinz C, Heiligenhaus A, et al. The effect of methotrexate and sulfasalazine on the course of HLA-B27-positive anterior uveitis: results from a retrospective cohort study. Graefes Arch Clin Exp Ophthalmol. (2018) 256:1985–92. doi: 10.1007/s00417-018-4082-x

PubMed Abstract | Crossref Full Text | Google Scholar

94. Pacheco PA, Taylor SR, Cuchacovich MT, and Diaz GV. Azathioprine in the management of autoimmune uveitis. Ocul Immunol Inflammation. (2008) 16:161–5. doi: 10.1080/09273940802204519

PubMed Abstract | Crossref Full Text | Google Scholar

95. Jaffe GJ, Dick AD, Brézin AP, Nguyen QD, Thorne JE, Kestelyn P, et al. Adalimumab in patients with active noninfectious uveitis. N Engl J Med. (2016) 375:932–43. doi: 10.1056/NEJMoa1509852

PubMed Abstract | Crossref Full Text | Google Scholar

96. Nguyen QD, Merrill PT, Jaffe GJ, Dick AD, Kurup SK, Sheppard J, et al. Adalimumab for prevention of uveitic flare in patients with inactive non-infectious uveitis controlled by corticosteroids (VISUAL II): a multicentre, double-masked, randomised, placebo-controlled phase 3 trial. Lancet. (2016) 388:1183–92. doi: 10.1016/S0140-6736(16)31339-3

PubMed Abstract | Crossref Full Text | Google Scholar

97. Suhler EB, Jaffe GJ, Fortin E, Lim LL, Merrill PT, Dick AD, et al. Long-term safety and efficacy of adalimumab in patients with noninfectious intermediate uveitis, posterior uveitis, or panuveitis. Ophthalmology. (2021) 128:899–909. doi: 10.1016/j.ophtha.2020.10.036

PubMed Abstract | Crossref Full Text | Google Scholar

98. Rudwaleit M, Rødevand E, Holck P, Vanhoof J, Kron M, Kary S, et al. Adalimumab effectively reduces the rate of anterior uveitis flares in patients with active ankylosing spondylitis: results of a prospective open-label study. Ann Rheum Dis. (2009) 68:696–701. doi: 10.1136/ard.2008.092585

PubMed Abstract | Crossref Full Text | Google Scholar

99. Ming S, Xie K, He H, Li Y, and Lei B. Efficacy and safety of adalimumab in the treatment of non-infectious uveitis: a meta-analysis and systematic review. Drug Des Devel Ther. (2018) 12:2005–16. doi: 10.2147/DDDT.S160431

PubMed Abstract | Crossref Full Text | Google Scholar

100. Li B, Li H, Zhang L, and Zheng Y. Efficacy and safety of adalimumab in noninfectious uveitis: A systematic review and meta-analysis of randomized controlled trials. Front Pharmacol. (2021) 12:673984. doi: 10.3389/fphar.2021.673984

PubMed Abstract | Crossref Full Text | Google Scholar

101. Zur Bonsen LS, Knecht VA, Rübsam A, Pohlmann D, and Pleyer U. Adalimumab autoantibodies in uveitis patients: do we need routine drug monitoring? Biomedicines. (2024) 12. doi: 10.3390/biomedicines12122782

PubMed Abstract | Crossref Full Text | Google Scholar

102. Liu W, Bai D, and Kou L. Comparison of infliximab with adalimumab for the treatment of non-infectious uveitis: a systematic review and meta-analysis. BMC Ophthalmol. (2023) 23:240. doi: 10.1186/s12886-023-02987-1

PubMed Abstract | Crossref Full Text | Google Scholar

103. Kwon OC, Lee HS, Yang J, and Park M-C. Comparative risk of incident and recurrent acute anterior uveitis across different biological agents in patients with ankylosing spondylitis. Rheumatol (Oxford England). (2025) 64:588–96. doi: 10.1093/rheumatology/keae003

PubMed Abstract | Crossref Full Text | Google Scholar

104. Sieper J, Koenig A, Baumgartner S, Wishneski C, Foehl J, Vlahos B, et al. Analysis of uveitis rates across all etanercept ankylosing spondylitis clinical trials. Ann Rheum Dis. (2010) 69:226–9. doi: 10.1136/ard.2008.103192

PubMed Abstract | Crossref Full Text | Google Scholar

105. Ramiro S, Nikiphorou E, Sepriano A, Ortolan A, Webers C, Baraliakos X, et al. ASAS-EULAR recommendations for the management of axial spondyloarthritis: 2022 update. Ann Rheum Dis. (2023) 82:19–34. doi: 10.1136/ard-2022-223296

PubMed Abstract | Crossref Full Text | Google Scholar

106. Van Bentum RE, Heslinga SC, Nurmohamed MT, Gerards AH, Griep EN, Koehorst C, et al. Reduced occurrence rate of acute anterior uveitis in ankylosing spondylitis treated with golimumab - the GO-EASY study. J Rheumatol. (2019) 46:153–9. doi: 10.3899/jrheum.180312

PubMed Abstract | Crossref Full Text | Google Scholar

107. Jin Y, Lu S, Lin Y, and Mou X. The efficacy and safety of TNF inhibitor (golimumab) as salvage treatment in patients with refractory noninfectious uveitis. Inflammopharmacology. (2022) 30:1363–8. doi: 10.1007/s10787-022-01019-6

PubMed Abstract | Crossref Full Text | Google Scholar

108. Van Der Horst-Bruinsma IE, Van Bentum RE, Verbraak FD, Deodhar A, Rath T, Hoepken B, et al. Reduction of anterior uveitis flares in patients with axial spondyloarthritis on certolizumab pegol treatment: final 2-year results from the multicenter phase IV C-VIEW study. Ther Adv Musculoskelet Dis. (2021) 13:1759720x211003803. doi: 10.1177/1759720X211003803

PubMed Abstract | Crossref Full Text | Google Scholar

109. Lindström U, Bengtsson K, Olofsson T, Di Giuseppe D, Glintborg B, Forsblad-D'elia H, et al. Anterior uveitis in patients with spondyloarthritis treated with secukinumab or tumour necrosis factor inhibitors in routine care: does the choice of biological therapy matter? Ann Rheum Dis. (2021) 80:1445–52. doi: 10.1136/annrheumdis-2021-220420

PubMed Abstract | Crossref Full Text | Google Scholar

110. Dick AD, Tugal-Tutkun I, Foster S, Zierhut M, Melissa Liew SH, Bezlyak V, et al. Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology. (2013) 120:777–87. doi: 10.1016/j.ophtha.2012.09.040

PubMed Abstract | Crossref Full Text | Google Scholar

111. Brandt-Jürgens J, Rudwaleit M, Behrens F, Ritchlin C, Peterlik D, Quebe-Fehling E, et al. Uveitis in patients with axial spondyloarthritis or psoriatic arthritis: a post hoc analysis from placebo-controlled phase III studies with secukinumab. Ther Adv Musculoskelet Dis. (2025) 17:1759720x251340255. doi: 10.1177/1759720X251340255

PubMed Abstract | Crossref Full Text | Google Scholar

112. Brown MA, Rudwaleit M, Van Gaalen FA, Haroon N, Gensler LS, Fleurinck C, et al. Low uveitis rates in patients with axial spondyloarthritis treated with bimekizumab: pooled results from phase 2b/3 trials. Ann Rheum Dis. (2024) 83:1722–30. doi: 10.1136/ard-2024-225933

PubMed Abstract | Crossref Full Text | Google Scholar

113. Mugheddu C, Atzori L, Del Piano M, Lappi A, Pau M, Murgia S, et al. Successful ustekinumab treatment of noninfectious uveitis and concomitant severe psoriatic arthritis and plaque psoriasis. Dermatol Ther. (2017) 30. doi: 10.1111/dth.12527

PubMed Abstract | Crossref Full Text | Google Scholar

114. Chateau T, Angioi K, and Peyrin-Biroulet L. Two cases of successful ustekinumab treatment for non-infectious uveitis associated with crohn's disease. J Crohns Colitis. (2020) 14:571. doi: 10.1093/ecco-jcc/jjz167

PubMed Abstract | Crossref Full Text | Google Scholar

115. Gaggiano C, Sota J, Gentileschi S, Caggiano V, Grosso S, Tosi GM, et al. The current status of biological treatment for uveitis. Expert Rev Clin Immunol. (2020) 16:787–811. doi: 10.1080/1744666X.2020.1798230

PubMed Abstract | Crossref Full Text | Google Scholar

116. Paley MA, Karacal H, Rao PK, Margolis TP, and Miner JJ. Tofacitinib for refractory uveitis and scleritis. Am J Ophthalmol Case Rep. (2019) 13:53–5. doi: 10.1016/j.ajoc.2018.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

117. Miserocchi E, Giuffrè C, Cornalba M, Pontikaki I, and Cimaz R. JAK inhibitors in refractory juvenile idiopathic arthritis-associated uveitis. Clin Rheumatol. (2020) 39:847–51. doi: 10.1007/s10067-019-04875-w

PubMed Abstract | Crossref Full Text | Google Scholar

118. Srivastava SK, Watkins TR, Nguyen QD, Sharma S, Scales DK, Dacey MS, et al. Filgotinib in active noninfectious uveitis: the HUMBOLDT randomized clinical trial. JAMA Ophthalmol. (2024) 142:789–97. doi: 10.1001/jamaophthalmol.2024.2439

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: spondyloarthritis, uveitis, HLA-B27, extra articular manifestations, biologics

Citation: Pan S, Xiang L, Cao Z, Yang Z and Cheng R-J (2025) Beyond the joints: mechanistic insights and multidisciplinary strategies for spondyloarthritis-associated uveitis. Front. Immunol. 16:1715107. doi: 10.3389/fimmu.2025.1715107

Received: 28 September 2025; Accepted: 14 November 2025; Revised: 30 October 2025;
Published: 03 December 2025.

Edited by:

Emanuele Bizzi, Vita-Salute San Raffaele University, Italy

Reviewed by:

Eshwa Dar, Islam College of Pharmacy, Pakistan
Jose Alexandre Mendonça, Pontifical Catholic University of Campinas, Brazil

Copyright © 2025 Pan, Xiang, Cao, Yang and Cheng. 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: Zhi Yang, eWFuZ3poaWNkd3lAcXEuY29t; Rui-Juan Cheng, b3JhbmdlNDExQHFxLmNvbQ==

These authors have contributed equally to this work and share first authorship

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.