Uveitis in Juvenile Idiopathic Arthritis


Inflammatory eye diseases comprise some of the most devastating complications of childhood rheumatic diseases, especially juvenile idiopathic arthritis (JIA). Chronic (initially asymptomatic) uveitis accompanying JIA is one of the most common causes of uveitis in childhood. It is predominantly anterior, nongranulomatous inflammation affecting the iris and ciliary body (iridocyclitis) of insidious onset ( Fig. 22.1 ). Acute (symptomatic) anterior uveitis is characteristic of human leukocyte antigen-B27 (HLA-B27)–associated diseases such as enthesitis-related arthritis (ERA). The posterior uveal tract (the choroid) is less often affected in rheumatic diseases of childhood.

Fig. 22.1
Schematic view of a sagittal section of the eye. Chronic anterior uveitis (iridocyclitis) involves the iris and ciliary body primarily, but secondary effects occur in the cornea, anterior chamber, lens, vitreous, and (rarely) retina.

Classification of Uveitis

Uveitis is classified in terms of its anatomy and time course according to the Standardization of Uveitis Nomenclature (SUN) international working group ( Tables 22.1 and 22.2 ). Anatomically it can be anterior, intermediate, posterior, or panuveitis. The onset can be insidious or sudden and its duration limited (3 months or less) or persistent (more than 3 months). The term acute refers to cases in which the onset was sudden and the duration limited (as seen in HLA-B27–associated acute anterior uveitis); recurrent, in which there are repeated episodes separated by periods of inactivity without treatment greater than 3 months in duration; and chronic, in which there is persistent disease with prompt relapses (within 3 months) after discontinuation of therapy ( Table 22.2 ). Most uveitis in children with oligoarticular, rheumatoid factor (RF)-negative polyarticular, or psoriatic JIA is anterior with an insidious onset and has a chronic or frequently recurrent course. Intermediate uveitis with inflammation predominantly in the vitreous humor is occasionally described in JIA.

TABLE 22.1
The SUN Working Group Classification of Uveitis
Type Primary Site of Inflammation Includes
Anterior uveitis Anterior chamber Iritis
Iridocyclitis
Anterior cyclitis
Intermediate uveitis Vitreous Pars planitis
Posterior cyclitis
Hyalitis
Posterior uveitis Retina or choroid Focal, multifocal, or diffuse choroiditis
Chorioretinitis
Retinochoroiditis
Retinitis
Neuroretinitis
SUN, Standardization of Uveitis Nomenclature.

TABLE 22.2
The SUN Working Group Descriptors of Uveitis
Category Descriptor Comment
Onset Sudden
Insidious
Duration Limited <3 months’ duration
Persistent >3 months’ duration
Course Acute Episode of sudden onset and limited duration
Recurrent Repeated episodes separated by periods of inactivity without treatment >3 months in duration
Chronic Persistent uveitis with relapses in <3 months after discontinuing treatment
SUN, Standardization of Uveitis Nomenclature.

Uveitis in Juvenile Idiopathic Arthritis

History: The Association of Arthritis and Uveitis

Ohm first described chronic uveitis and band keratopathy in 1910 in a child with arthritis. The association of ocular disease and juvenile arthritis was confirmed by several authors. In Sury’s large series of children with chronic arthritis, chronic uveitis was found in 15% of the total, and two-thirds of those patients with uveitis had band keratopathy. The majority of his patients had an insidious onset of uveitis with little or no early disturbance of vision; diagnosis was often delayed until slit-lamp examination was performed. The occurrence of “chronic, asymptomatic, nongranulomatous anterior uveitis” became recognized as an important complication of what was called juvenile rheumatoid arthritis , particularly the limited joint disease referred to as pauciarthritis , equivalent to oligoarthritis, a subset of JIA in the International League of Associations for Rheumatology (ILAR) classification, as well as RF-negative polyarthritis and psoriatic arthritis.

Epidemiology

The uveitis associated with JIA accounts for 15% to 67% of cases of uveitis in children reported in large series from tertiary care ophthalmology centers in Europe, North America, and Israel. The documented frequency of chronic uveitis in children with chronic arthritis has varied considerably from 2% in Costa Rica to 11.6% in the United States, 13% in Canada, and 16% in the Nordic countries. , It appears to be particularly uncommon in Asian and African populations but has been reported worldwide. A worldwide observational cohort study confirmed disparities in prevalence of JIA-associated uveitis being highest in northern Europe (19.1%) and southern Europe (18.8%) and lowest in Latin America (6.4%), Africa and the Middle East (5.9%), and Southeast Asia (5.0%).

A longitudinal prospective cohort study of 435 children with JIA in Nordic countries identified uveitis in 20.5%. The frequency of uveitis was related to JIA subtype: 35.7% in juvenile psoriatic arthritis, 22.5% in RF-negative polyarticular, 20.5% in extended oligoarticular, 19.1% in persistent oligoarticular, 19.0% in undifferentiated, and 8.3% in ERA. This series reported a very high rate of uveitis among patients with juvenile psoriatic arthritis, but the cohort included only 14 patients with this condition. No patients with systemic (n = 18) or RF-positive JIA (n = 3) developed uveitis, consistent with earlier studies that reported low incidence in these subtypes , ( Table 22.3 )

TABLE 22.3
Prevalence of Uveitis According to JIA Subtype
Sim Saurenmann Heiligenhaus Nordal
Year 2006 2007 2007 2017
Total cohort 202 1080 3271 435
Number with uveitis (% of total) 23 (11.4) 142 (13.1) 406 (12.4) 89 (20.5)
Percentage of each ILAR subtype with uveitis
Persistent oligoarticular 15.6 18 16 19.1
Extended oligoarticular 25 30 25 20.5
Polyarticular (RF-negative) 6 14.1 4 22.5
Polyarticular (RF-positive) 0 0 2 0
Psoriatic arthritis 8 9.8 10 35.7
Enthesitis-related arthritis 4 7.8 7 25.0
Systemic arthritis 0 0.6 1 0
Undifferentiated 0 7.1 11 20.6
ILAR, International League of Associations for Rheumatology; RF, rheumatoid factor.

The large Canadian ReACCH-Out (Research in Arthritis in Canadian Children Emphasizing Outcomes) cohort study has reported on the incidence of uveitis in the years after diagnosis of JIA. Of 1183 patients enrolled between 2005 and 2010 with a median follow-up 35.2 months, 87 (7.4%) developed uveitis. The 5-year cumulative incidence calculated by Kaplan-Meier analysis and including patients with uveitis prior to JIA diagnosis was 13.9%. The incidence of new-onset uveitis was 2.8% per year and decreased during the first 5 years but remained at 2.1% in the fifth year. Although decreasing each year, the risk is never entirely absent , ( Fig. 22.2 ). This highlights the importance of regular screening for uveitis for at least several years, as discussed in more detail in the Clinical Manifestations section. Uveitis may precede the onset of arthritis in 3% to 7% of children with JIA. In almost half of all patients with uveitis, it occurs just before arthritis is diagnosed, at the time of diagnosis, or shortly thereafter. , , In the Nordic study discussed earlier, uveitis developed at a median interval of 0.8 years (range: 4.7 to 9.4 years) after the onset of arthritis. Another study, from Atlanta, Georgia, in the United States, found that 24% of 52 children were diagnosed with uveitis prior to arthritis, 22% within the first year after onset of arthritis and 86% cumulatively within 4 years. A Finnish study of children with JIA and uveitis documented the appearance of uveitis within the first 3 months after onset of arthritis in 49%; 90% developed uveitis within the first 4 years after onset of arthritis. There is some evidence that, although the prevalence of JIA may be increasing, there may be a decreasing frequency of uveitis in children with JIA. A prospective, observational, cross-sectional study from Germany found a small but significant decrease in uveitis prevalence from 2002 to 2013 (13.0% to 11.6%, odds ratio [OR] 0.98, P = 0.015). The authors suggest this may be associated with increased use of synthetic and biological disease-modifying antirheumatic drugs (DMARDs) during this period. Uveitis is bilateral in 70% to 80% of children. , Patients with unilateral disease are unlikely to develop bilateral involvement after the first year of disease; however, there are exceptions, and unilateral uveitis may persist for many years in a few children before the other eye is involved.

Fig. 22.2, Graphs showing the temporal relationships between arthritis and uveitis in children with juvenile idiopathic arthritis. A, The distribution of age at onset of arthritis in a series of 38 children who developed uveitis. B, The distribution of age at onset of uveitis in the same children. Note that in four patients, uveitis began after their 15th birthday: at 15 ½, 18, 31, and 39 years of age. C, Interval between onset of arthritis and diagnosis of uveitis in these patients. Note that for one patient the interval was 29 years, and for another, 34 years.

Risk Factors for JIA-Associated Uveitis

Several risk factors for development of uveitis in JIA have been identified including age of onset, gender, JIA subtype, and antinuclear antibody (ANA) and HLA-B27 positivity. , , Early studies have consistently shown that the frequency of chronic anterior uveitis in JIA is highest in girls who have oligoarthritis with an early age at onset (younger than 7 years of age) and who are ANA-positive by immunofluorescence. , , The interaction between individual risk factors is likely to be interdependent and complex. In a study of 1047 patients with JIA, the age-associated risk of uveitis was seen only in young girls but not in young boys. This finding appeared to be confirmed in a Nordic longitudinal study in which the median age at onset of arthritis in girls with uveitis was 3.4 years versus 6.8 years in girls without uveitis ( P < 0.001). The difference in age of onset in boys was not significant. The same study also identified antihistone antibodies (AHAs) greater than or equal to 15 units/mL as a significant predictor of uveitis only in girls. A multivariable analysis in the ReACCH-Out cohort found only young age at JIA diagnosis and ANA positivity were independent predictors of uveitis. These findings support the earlier results in a large cohort study from Germany. Salek et al. described a group of six children with uveitis in association with psoriatic arthritis and noted that the uveitis was especially severe in those with an onset at or before age six. Reports from India document a low frequency of ANA positivity in children with JIA, including those with uveitis.

A further predictor of increased risk of uveitis appears to be elevated erythrocyte sedimentation rate (ESR) at the time of diagnosis of JIA. In a retrospective study of patients with oligoarticular JIA who were recruited at the time of diagnosis, ESR was significantly higher in the group who later developed uveitis versus those who did not (52 mm/hr vs. 24 mm/hr, respectively; OR 1.05, 95% confidence interval [CI], 1.01 to 1.09). Multivariate analysis in this study suggested that ESR greater than 22 mm/hr and patient age less than 3 years at onset of arthritis were associated with ORs of 5.28 and 3.80, respectively, for the development of uveitis. Elevated ESR (≥20 mm/hr) at baseline (hazard ratio [HR] 2.36, 95% CI, 1.38 to 4.02), and moderate or high disease activity during follow-up (based on 10-joint clinical Juvenile Arthritis Disease Activity Score) (HR 4.30, 95% CI, 2.51 to 7.37) were also found to be predictors of the development of uveitis in a cohort of 954 patients with JIA. Elevated serum levels of S100A12 greater than or equal to 250 ng/mL at baseline were significantly associated with the risk of uveitis (HR 2.10, 95% CI, 1.15 to 3.85). In the same study, treatment of arthritis with methotrexate alone (HR 0.18, 95% CI, 0.12 to 0.29) or in combination with etanercept (HR 0.10, 95% CI, 0.04 to 0.23) or with adalimumab (HR 0.09, 95% CI, 0.01 to 0.61) all reduced the risk of uveitis onset.

Etiology and Pathogenesis

The pathogenesis of chronic uveitis and the basis of its association with JIA are not fully understood. It is evident that both B and T lymphocytes are participants in the process, and that the innate immune system plays a role. It is thought that an immune response is triggered against intraocular antigens including S-arrestin (also known as retinal S-antigen ), retinol-binding protein 3 (RBP3), and tyrosinase-related proteins.

Immunohistochemistry of eye biopsies from patients with JIA-associated uveitis showed a predominance of CD4 + T cells compared with CD8 + T cells and variable levels of CD20 + B cells. CD4 + T cells of T helper type 1 (Th1) and Th17 subsets produce interferon (IFN)-γ and interleukin (IL)-17, respectively. , The proinflammatory effects of these cells are counterbalanced by CD4 + , CD25 + , FOXP3 + , regulatory T cells (Tregs), and inducible Treg cells. Although there is evidence for a role of Th1, Th17, and Treg cells in uveitis pathogenesis, the exact roles of the T-cell subsets in the course of disease have not been fully defined. , , Analysis of peripheral blood samples has shown an increased Th1/Th2 ratio and increased Th17 numbers in patients with JIA-associated uveitis compared with healthy controls.

Evidence of the role of B lymphocytes in JIA-associated uveitis has been supported by transcriptomic and proteomic analysis of iris tissue and aqueous humor. Compared with patients with primary open-angle glaucoma, those with JIA-associated uveitis had significantly upregulated expression of B-lymphocyte–related genes including CD19 and CD20. At the protein level, there was higher expression of immunoglobulin components and the B-lymphocyte–associated protein MZB1. BAFF, APRIL, and IL-6 were also higher in the aqueous humor of JIA-associated uveitis patients. The same group also showed significantly higher levels of serum amyloid A (SAA), IL-8, transforming growth factor beta (TGFβ)-1, TGFβ-3, and tumor necrosis factor (TNF)-α in aqueous humor of patients with JIA-associated uveitis without glaucoma compared with a control group with senile cataracts.

Ooi et al. have reviewed the evidence that IL-1β, IL-2, IL-6, IFN-γ, and TNF-α are present in ocular fluids and tissues of adults with ankylosing spondylitis, and that higher levels of these cytokines are associated with more severe uveitis. Levels of TNF-α were especially elevated in the aqueous of patients with ankylosing spondylitis–associated uveitis. In another study that included children with JIA-associated uveitis, levels of IL-2, IL-6, IL-13, IL-18, IFN-γ, TNF-α, soluble ICAM-1 (also known as CD54 ), CCL5, and CXCL10 in the aqueous humor were considerably higher than in controls without uveitis. The clinical responsiveness of uveitis to the administration of some, but not all, anti–TNF-α agents provides strong support for the premise that TNF-α is at least in part responsible for the inflammation.

Keino et al. demonstrated that the oral administration of an inhibitor of IL-12/IL-23 prevented autoimmune uveoretinitis in animals by reducing the generation of IL-17–producing cells. Other animal studies have shown that anti-Th17 can block the development of uveitis in uveitis-susceptible mice, but that administration of the cytokine itself into uveitis-susceptible rats has a mitigating effect on disease development. Anti–IL-17 treatment of human uveitis (principally posterior disease) was not effective in three randomized controlled trials. However, a subsequent trial using a greater dosage of neutralizing antibody indicated that anti–IL-17 could reduce the inflammation characteristic of uveitis. Despite the implication of IL-17 in uveitis, some models involve either Th1 or Th2 cells exclusively. Animal models may not be entirely relevant to understanding human disease, however. In addition, in the most widely studied T-cell–dependent mouse model of uveitis, experimental autoimmune uveitis (EAU) is predominantly a chorioretinitis whose pathogenesis might differ significantly from anterior uveitis.

As noted previously, ANA positivity is a risk factor for JIA-associated uveitis, which raises the question of whether autoantibodies are involved in the pathogenesis. Although a correlation between ANA and plasma cell infiltration in anterior uveitis has been reported, it is not clear whether ANAs are actually pathogenic and the specifically recognized antigens are not known. Potential targets of autoantibodies have been studied using immunofluorescence of tissue sections from eyes incubated with sera from patients with JIA. When using patient sera compared with controls, there appeared to be an increased frequency of antibodies against the iris and retina, but not the ciliary body. However, because blood samples were taken after uveitis was well established, it is not clear whether the antiocular antibodies are part of the cause or a consequence of disease. In the future, serial serum samples taken from JIA patients before and after onset of uveitis could enable the potential prognostic significance of antibody binding to be investigated.

The basis of the association between inflammatory joint disease and inflammatory ocular disease is unexplained, but it is likely that a shared genetic predisposition plays a part.

Genetic Background

Genetic susceptibility to uveitis in JIA is complex. A few case reports have documented the occurrence of oligoarthritis and chronic anterior uveitis in siblings. However, monogenic or Mendelian patterns of inheritance of JIA-associated uveitis have not been seen.

A number of sometimes contradictory studies have reported an array of HLA alleles associated with chronic anterior uveitis in JIA. The strongest and most consistent associations appear to be with genes in the class II region. Studies from the United States , have documented a strong association with DRB1∗1104 (formerly called DR5 ), and associations with DQA1∗0501 and DQB1∗0301, which are in linkage disequilibrium with DRB1∗1104. The combination of HLA-DRB1∗1104 and HLA-DPB1∗0201 is associated with a 7.7-fold increased risk of chronic uveitis. The association of uveitis in JIA with DRB1∗11 noted in American children was confirmed in one Italian study but not in another. An association with DRB1∗13 has been reported in British and Greek children. An increased prevalence of HLA-DR9 (DRB1∗09) (OR 2.33) was also noted in one study. In a European cohort of patients with JIA, the presence of a serine residue at position 11 of HLA-DRB1 was associated with an increased risk of uveitis (OR 2.60) specifically in girls. Acute anterior uveitis is associated with the class I gene HLA-B27 . ,

Genetic polymorphisms of non-HLA genes have been less well studied. TNF gene polymorphisms (−238 and −308 GA) have been linked with HLA-B27–associated uveitis. In patients with acute anterior uveitis, an association was found with another single nucleotide polymorphism (SNP) (857T) in the TNF gene. The TNF locus is located in the HLA/major histocompatibility complex (MHC), and large studies are needed to determine whether TNF associations reflect linkage disequilibrium with class I or class II loci.

Clinical Manifestations

Insidious Onset, Chronic Uveitis

Chronic anterior uveitis, which is characteristic of oligoarticular JIA (persistent or extended) and most children with psoriatic or RF-negative polyarticular JIA (see Table 22.3 ), is usually completely asymptomatic with an insidious onset. Later in the disease course, and if untreated, some children have symptoms attributable to the uveitis such as pain, redness, headache, photophobia, and change in vision, although these are more typical features of acute anterior uveitis ( Table 22.4 ).

TABLE 22.4
Ocular Signs and Symptoms in Children with Chronic Uveitis and Arthritis
Data summarized from published studies.
Characteristic Percent Affected (Range)
Bilateral uveitis 25–89
Symptoms
Ocular pain and or redness 0–25
Change in vision 0–30
Photophobia 0–8
Headache 0–6
None 51–97

Regular screening for uveitis by slit-lamp biomicroscopy in patients with JIA is essential to detect clinically silent but potentially vision-threatening disease. Younger children in particular may be unable to report reliably any visual changes, so the need for formal assessment of vision and the education of families of the importance of this is vital. The early detection of chronic uveitis requires slit-lamp biomicroscopy, which should be performed in every child at the time diagnosis iof JIA is suspected rather than waiting for confirmation of the diagnosis at a pediatric rheumatology specialist center. Ophthalmological examination should be repeated at prescribed intervals during the first few years of the disease. Screening guidelines have been published from several countries including the United Kingdom, Germany, and the United States. The recommended frequency of ophthalmological examinations is influenced by the level of risk of uveitis ( Table 22.5 ). U.S. guidelines recommend that slit-lamp examinations be performed every 3 months for the first 4 years in children in the high-risk group (early age at onset, oligoarthritis or polyarthritis, and ANA positivity) and every 6 months thereafter for a period of 7 years at a minimum. In children with ANA-negative disease, slit-lamp examinations should be done initially at 6-month intervals. Children with psoriatic arthritis are also at considerable risk for the development of uveitis and should be followed at the same frequencies as children with oligoarticular or polyarticular JIA. In children with systemic-onset JIA, examinations once a year are probably sufficient. Subsequent to the publication of these guidelines, there have been suggestions for their modification. , Heiligenhaus et al. proposed guidelines that conformed to the ILAR classification of JIA ( Table 22.6 ).

TABLE 22.5
American Academy of Pediatrics Guidelines for the Ophthalmological Screening of Children with JIA
Adapted from Cassidy J, et al., American Academy of Pediatrics Section on Rheumatology and Section on Ophthalmology. Ophthalmologic examinations in children with juvenile rheumatoid arthritis. Pediatrics. 2006;117(5):1843-1845.
JIA Onset Type ANA Screening Schedule
Onset ≤6 Years Onset >6 Years
Oligoarthritis Positive Every 3 months Every 6 months
Oligoarthritis Negative Every 6 months Every 12 months
Polyarthritis Positive Every 3 months Every 6 months
Polyarthritis Negative Every 6 months Every 12 months
Systemic Negative or positive Every 12 months Every 12 months
High risk: Screen every 3 months
Moderate risk: Screen every 6 months
Low risk: Screen every 12 months
ANA, Antinuclear antibody; JIA, juvenile idiopathic arthritis.

All patients are considered to be at low risk 7 years after onset of arthritis; they should have yearly ophthalmological examinations indefinitely.

All patients are considered to be at low risk 4 years after onset of arthritis; they should have yearly ophthalmological examinations indefinitely.

All high-risk patients are considered to be at medium risk 4 years after onset of arthritis.

TABLE 22.6
Recommendations for Screening Based on the ILAR Classification of JIA
Adapted from Heiligenhaus A, Niewerth M, Ganser G, et al. German Uveitis in Childhood Study Group. Prevalence and complications of uveitis in juvenile idiopathic arthritis in a population-based nation-wide study in Germany: suggested modification of the current screening guidelines. Rheumatology (Oxford). 2007;46(6):1015–1019.
JIA Subgroup ANA Age At Onset of JIA Duration of JIA Screening
Oligoarthritis + <7 years <5 years 3 months
RF-negative polyarthritis
Psoriatic arthritis
Undifferentiated arthritis
+ <7 years >4 years 6 months
+ <7 years >7 years 12 months
+ >6 years <3 years 6 months
+ >6 years >2 years 12 months
<7 years <5 years 6 months
<7 years >4 years 12 months
>6 years NA 12 months
Enthesitis-related arthritis NA NA 12 months
RF-positive polyarthritis NA NA 12 months
Systemic arthritis NA NA 12 months
Patients in any category with uveitis NA NA According to uveitis course
ANA, Antinuclear antibody; ILAR, International League of Associations for Rheumatology; JIA, juvenile idiopathic arthritis; NA, not applicable; RF, rheumatoid factor.

The 2019 American College of Rheumatology (ACR)/Arthritis Foundation guideline recommends that, for children and adolescents with JIA and controlled uveitis who are tapering or discontinuing topical glucocorticoids, ophthalmic monitoring should take place within 1 month after each change in topical treatment. The British Society for Paediatric and Adolescent Rheumatology and the Royal College of Ophthalmologists in the United Kingdom recommend two monthly screenings for 6 months after stopping systemic immunosuppression. European consensus guidelines suggest that all patients with JIA-associated uveitis who stop systemic treatment should have three times monthly ophthalmology monitoring for at least 3 years off all forms of treatment. Any child who has had uveitis should be considered to be at high risk, even if it has remitted, and continued surveillance is essential.

The diagnostic signs of anterior uveitis on slit-lamp examination are the presence of inflammatory cells (“cells”) and increased protein concentration (“flare”) in the aqueous humor of the anterior chamber (AC) of the eye ( Fig. 22.3 ). Deposition of inflammatory cells on the inner surface of the cornea (keratic precipitates) may be detected at presentation or develop later. Terms used to describe uveitis and its complications are listed in Table 22.7 .

Fig. 22.3, A slit-lamp examination shows “flare” in the fluid of the anterior chamber (caused by increased protein content) and keratic precipitates on the posterior surface of the cornea, representing small collections of inflammatory cells.

TABLE 22.7
Terms Used by Ophthalmologists to Describe Abnormalities in Uveitis
Term Definition
Anterior uveitis Ocular inflammation that is predominantly anterior to the lens. Subsets of anterior uveitis include iritis and iridocyclitis; the latter term is preferred if the ciliary body is involved.
Intermediate uveitis Ocular inflammation in which the predominant manifestation is leukocytes in the vitreous humor.
Panuveitis Ocular inflammation involving the anterior chamber, the vitreous humor, and the retina and/or choroid.
Mutton-fat precipitates Large concretions of leukocytes attached to the endothelial surface of the cornea.
Granulomatous uveitis Uveitis usually characterized by large concretions of cells on the corneal endothelium (granulomatous keratic precipitates) or iris nodules. Although these findings are more common in a granulomatous disease such as sarcoidosis or tuberculosis, the histology of these lesions is not a granuloma.
Nodules Cellular nodules in a patient with uveitis including Koeppe nodule at the pupillary margin and Busacca nodules in the iris stroma.
Pars planitis A subset of intermediate uveitis in which the inflammation is predominantly over the pars plana, an anatomical region just posterior to the ciliary body.
Cystoid macular edema Collection of fluid in the macula, a common cause of vision loss in patients with uveitis.
Vitritis Inflammation in the vitreous humor.
Cyclitic membrane Fibrotic tissue that forms posterior to the lens capsule and impedes the visual axis.
Cataract Opacification of the lens as occurs from aging, chronic oral or topical corticosteroid use, or chronic inflammation.
Glaucoma Optic nerve injury usually resulting from a chronic elevation of intraocular pressure.
Hypotony Excessively low intraocular pressure.
Posterior synechiae Adhesions between the iris and the anterior lens capsule.
Anterior synechiae Adhesions between the iris and the cornea.
Iris bombé Circumferential posterior synechiae resulting in impaired flow of aqueous humor and a marked elevation of intraocular pressure.
Keratic precipitates Concretions of cells that deposit on the corneal endothelium during uveitis.
Band keratopathy Deposition of calcium in the corneal epithelium as can occur from chronic anterior uveitis in JIA.
Phthisis The irreversible end stage of chronic inflammation or infection.
Snowbanking The cellular exudate over the pars plana, a characteristic of pars planitis.
JIA, Juvenile idiopathic arthritis.

Intraocular inflammation is graded according to the SUN criteria, which take into account AC cells, AC flare, vitreous cells, and vitreous haze or debris ( Tables 22.8 and 22.9 ). The criteria also provide definitions of improvement and worsening of the condition ( Table 22.10 ) allowing reproducible assessment and monitoring of uveitis activity. It is important to note that 0.5+ AC cells should not be considered inactive uveitis.

TABLE 22.8
Standardization of Uveitis Nomenclature Grading Scheme for Anterior Chamber Cells
Grade Cells in Field
0 <1
0.5+ 1–5
1+ 6–15
2+ 16–25
3+ 26–50
4+ >50

Field size is 1 mm by 1 mm slit beam

TABLE 22.9
Standardization of Uveitis Nomenclature Grading Scheme for Anterior Chamber Flare
Grade Description
0 None
1+ Faint
2+ Moderate (iris and lens details clear)
3+ Marked (iris and lens details hazy)
4+ Intense (fibrin or plastic aqueous)

TABLE 22.10
Standardization of Uveitis Nomenclature Activity of Uveitis Terminology
Term Definition
Inactive Grade 0 cells
Worsening activity Two-step increase in level of inflammation (e.g., anterior chamber cells, vitreous haze) or increase from grade 3+ to grade 4+
Improved activity Two-step decrease in level of inflammation (e.g., anterior chamber cells, vitreous haze) or decrease to grade 0
Remission Inactive disease for >3 months after discontinuing all treatments for eye disease

Applies to anterior chamber inflammation

Measurement of visual acuity (VA) is an important part of assessment and gives an indication of both disease activity and damage resulting from both chronic inflammation and failure or complications of treatment. Guidelines for measuring outcome in JIA-associated uveitis, which include assessment of VA as a key component, have been developed. This should be best corrected visual acuity (BCVA) using age-appropriate tests recorded monocularly and binocularly and converted to logarithm of the minimal angle of resolution (logMAR). LogMAR is calculated using the following formula: logMAR = –log 10 (VA fraction).

Complications of chronic anterior uveitis are frequent and increase with increasing duration of active disease. Earlier systemic treatment is likely to have resulted in a lower frequency of complications in later series. Posterior synechiae, inflammatory adhesions between the iris and anterior surface of the lens, result in an irregular or poorly reactive pupil ( Table 22.11 ; Fig. 22.4 ). This abnormality may be the first obvious clue to the presence of uveitis on ophthalmoscopic examination, but it is frequently a sign of long-standing or severe disease. Synechiae that are circumferential prevent the free flow of aqueous humor between the posterior and anterior chambers, resulting in bulging of the iris (iris bombé) and increased intraocular pressure.

TABLE 22.11
Frequency of Complications of Chronic Uveitis in Reported Cases
Complication Reported Range (%)
Synechiae 37–75
Band keratopathy 11–56
Cataract 6–75
Glaucoma 8–25
Hypotony 5–10
Phthisis bulbi 0–14
Cystoid macular edema 3–6

Fig. 22.4, Posterior synechiae. A, Iris is tethered to the lens at a single point revealed only after mydriatics were given in this 4-year-old girl with oligoarthritis. B, The eye of a 7-year-old boy shows an irregular pupil that resulted from multiple adhesions of the iris to the anterior surface of the lens.

Band keratopathy is caused by deposition of calcium in the corneal epithelium and tends to be a late occurrence ( Fig. 22.5 ). Initially, crescentic gray-to-brown depositions are seen at the corneal limbus. With time, the band progresses centrally at the equator of the eye and impairs light entry through the pupil. Cataracts result either from the inflammatory disease or the use of topical corticosteroids. Changes in intraocular pressure can result in serious complications including blindness: Raised pressure is associated with glaucoma and low pressure (hypotony). , , Chronic ocular inflammation or persistent hypotony may rarely result in phthisis bulbi, irreversible end-stage disease characterized by a shrunken, nonfunctional eye. Cystoid macular edema, which affects central vision, is reported in some series ( Fig. 22.6 ). ,

Fig. 22.5, A, Early band keratopathy is noted as a semilunar band just inside the limbus medially and laterally. It does not extend across the pupil. B, The semiopaque band extends across the midplane of the cornea in this example of more advanced band keratopathy.

Fig. 22.6, Cystoid macular edema. Optical coherence tomography (OCT) images the retina and its layers by reflectance. The normal macula is concave. The convex appearance of the macula in this example is the result of cystoid edema.

Rare ocular conditions associated with childhood rheumatic diseases include papillitis, scleritis, episcleritis, and keratoconjunctivitis sicca. Keenan et al. suggest that granulomatous uveitis may be more common than has been thought. Using a definition of granulomatous uveitis as the presence of Busacca or angle nodules, mutton-fat keratic precipitates (KPs), or hyalinized ghost KPs, they observed granulomatous uveitis in 28% of 71 children with JIA. Granulomatous uveitis may be more common in black patients (67% of those patients with granulomatous disease in the series) than white patients (25%). Many of these patients had only ghost KP as evidence of disease; this may account for the higher incidence of granulomatous uveitis than in other studies of juvenile arthritis. It should be noted that “granulomatous” in this context does not indicate the presence of granulomata as seen in tuberculosis or sarcoidosis.

Sudden-Onset (Acute) Uveitis

Sudden-onset (acute) uveitis differs in several ways from the more common insidious-onset chronic uveitis and is strongly associated with HLA-B27 and ERA. Among 142 patients with JIA-associated uveitis in one study, acute anterior uveitis was seen in 16.2%. In a large series reported by Nordal et al., 16.7% of children with ERA (n = 48) had acute uveitis. It is much more common in boys (as is ERA), is more often unilateral, and is characterized by a painful, red, photophobic eye. Until the patient is evaluated by an ophthalmologist, the symptoms are often mistakenly attributed to a foreign body, an infection, or an allergy. Slit-lamp examination reveals the presence of cells and flare in the anterior chamber. Because of the symptomatic nature of this type of anterior uveitis, the process is usually identified and appropriately treated soon after onset; as a result, long-term sequelae are uncommon and visual prognosis is excellent. The reasons for the differences in clinical appearance between sudden-onset acute uveitis and insidious-onset chronic uveitis are unknown.

Differential Diagnosis

Anterior uveitis without evidence of joint or systemic involvement probably accounts for most children with anterior uveitis. , Such children should be evaluated for the presence of occult inflammatory joint disease (including leg length inequality and muscle wasting) and monitored for the possible development of arthritis or other systemic disease over time.

Uveitis and arthritis occur together with high frequency in a number of diseases other than JIA. Inflammation of the anterior uveal tract may complicate the arthropathy of inflammatory bowel disease and reactive arthritis. Uveitis also occurs in chronic infantile neurological cutaneous and articular syndrome (CINCA)/neonatal-onset multisystem inflammatory disease (NOMID), sarcoidosis, Blau syndrome, Behçet disease, and Kawasaki disease. Uveitis that occurs in these disorders is discussed in the respective chapters. Uveitis rarely occurs in systemic lupus erythematosus, polyarteritis nodosa, or Henoch–Schönlein purpura. Syndromes associated with uveitis are listed in Box 22.1 .

BOX 22.1
Diseases Most Often Associated with Anterior Uveitis in Children

Rheumatic Diseases

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here