Skip to main content

Mimickers of anterior uveitis, scleritis and misdiagnoses- tips and tricks for the cornea specialist

Abstract

Background

Anterior uveitis, inflammation of the anterior chamber and related structures, is a cohort of diseases that can present to almost any general or sub-specialty Ophthalmology practice. Its features classically involve anterior chamber cell and flare. Below the surface of these two signs exist a panoply of diagnoses.

Body

The purpose of this review is to provide a general framework for diagnoses of anterior uveitis that are often missed as well as non-uveitic pathologies that often mimic anterior uveitis. Diagnostic deviation in either direction can have vision-threatening and rarely life-threatening consequences for patients. Using a comprehensive literature review we have collected a broad spectrum of etiologies of anterior uveitis that are easily missed and non-uveitic pathologies that can masquerade as anterior uveitis.

Conclusions

We present a focused review on specific misdiagnosed anterior uveitis pathologies and some of the conditions that can masquerade as anterior uveitis and scleritis.

Background

Anterior uveitis (AU) and anterior scleritis/ episcleritis, inflammations of the anterior chamber, sclera and related structures, are a cohort of diseases that can present to almost any general or sub-specialty ophthalmology practice. Their features classically involve anterior chamber cell, flare for anterior uveitis; and an inflammatory disorder of the sclera that may also involve the cornea, adjacent episclera, and underlying uveal tract for scleritis. Below the surface of these signs exist a panoply of diagnoses. Broadly speaking, the etiology for uveitis can be divided into infectious, inflammatory and autoimmune [1, 2].

The purpose of this review is to provide a general framework for diagnoses of AU/ scleritis that are often missed as well as non-uveitic pathologies that often mimic AU/ scleritis. Thus, the field of ocular inflammation possesses a significant challenge as many diseases may present as masqueraders and represent obscure diagnoses. Actually, when using the term “Uveitis Masqueraders”, most of these are simply different conditions with similar phenotypes or clinical appearance that mimic a chronic ocular inflammation. We must exclude neoplastic and non neoplastic ocular and systemic diseases uveitis that mimic chronic ocular inflammation. Among them, oncologic conditions such as lymphoma, hematologic malignancy, and paraneoplastic syndrome can present with ophthalmic manifestations. Infectious conditions, like Lyme disease, tuberculosis, syphilis, bartonella, Herpes and zoster virus have diverse ocular presentations, and treatment modalities. Patients who are immune compromised, either as a result of HIV infecting, cancer, or the treatment thereof, are at increased risk of infectious uveitis with atypical presentations. The growing field of drug-related uveitis and are also important considerations. The correct diagnosis often requires a comprehensive rheumatologic and ophthalmological evaluation, including: clinical history, multimodal imaging and systemic workup. Diagnostic deviation in either direction can have vision-threatening and rarely life-threatening consequences for patients. We reviewed the most common conditions that present as mimickers of AU, scleritis and conversely, inflammatory conditions that present as mimickers of AU, scleritis, and uveitis conditions with rare and diagnostically challenging etiologies.

Their typical and atypical presentations are reviewed as outlined below. Diagnostic strategies among the entities described below are also presented and discussed. (1.) Common vascular and neoplastic conditions that may present as anterior uveitis masqueraders; (2.) Misdiagnoses among uveitis/anterior segment inflammation entities (3.) Drug Related Uveitis. (4.) Masquerade presentations of anterior segment inflammation: e.g. Peripheral Ulcerative Keratitis (PUK) misdiagnosed for an infectious bacterial ulcer, (5.) Conditions that may present as scleritis masqueraders. (6.) Misdiagnoses among patients with scleritis.

Method of literature search

The study was conducted according to the Preferred Reporting Items of Systematic Reviews (PRISMA) guidelines [3].

A literature search and subsequent screening of articles was conducted in 2022 by four authors (MPE, SK, GK, MHE). PubMed served as the primary database for the electronic literature search, although EBSCO and Cochrane were also surveyed. We systematically reviewed the available literature on neoplastic and nonneoplastic inflammatory masquerade syndromes. Literature searches were performed using electronic medical databases of the following keywords: Anterior uveitis OR anterior scleritis OR Neoplastic Uveitis OR Masquerader syndrome OR Uveitis mimickers OR medication induced uveitis OR Post-Vaccination Uveitis. The search timeframe was not limited by a specific date, but rather by the results of the articles retrieved.

The retrieved articles were initially screened by title and abstract, and articles with the relevant titles were then screened by full text using predefined inclusion and exclusion criteria. Inclusion criteria included 1) the paper must be written in or available in English and 2) the paper discussed the presentation and management of masquerade syndromes, inflammatory and infectious ocular diseases and uveitis. Exclusion criteria included 1) the paper concerned patients only with other inflammatory ocular diseases (episcleritis, scleritis) and AU, intermediate uveitis. 2) the paper did not clearly diagnose the patient with a masquerade syndrome. 3) citations were from grey literature. The full article was screened in cases where the relevance was unclear from the abstract. Relevant articles were ultimately compiled into a database and removed of duplicates. A total of 214 papers were finally selected.

No research ethics approval was needed for this study, as there were no human or animal participants included. The study protocol complied with the tenets of the Declaration of Helsinki.

Common vascular and neoplastic conditions that may present as anterior uveitis masqueraders

Anterior uveitis (AU) has a variety of infectious, inflammatory and autoimmune etiologies. However, data show that up to 38-88% of all acute anterior uveitis cases are diagnosed as idiopathic, depending on geographic location [4, 5]. While there may be certainly a variety of uveitic presentations that are idiopathic in nature, there are several AU etiologies that are frequently misdiagnosed as idiopathic and are highlighted here.

Neoplastic conditions masquerading as anterior uveitis

Although rare, leukemic infiltration of the anterior segment can mimic anterior uveitis. One study found only this type of leukemic infiltration in 0.5%–2.5% cases of leukemic relapses [6].

Beyond leukemia, other malignancies have been reported: metastasis in the case of lung, gastrointestinal and breast carcinoma, primary intraocular lymphoma (PIOL) [7, 8], systemic non-Hodgkin lymphoma metastatic to uveal structures within the eye, primary uveal melanoma (ciliary body), and retinoblastoma [9,10,11,12,13,14,15,16].

The most common type of lymphoma is Non-Hodgkin lymphoma (NHL). If it involves the eye, it usually localizes to the adnexal structures or the posterior segment, including the vitreous cavity, retina and uveal tissue [13].

Other lymphomas, like Burkitt lymphoma (BL), are associated with Epstein-Barr virus (EBV), with few reports of BL masquerading as anterior uveitis. Patients often present with orbital invasion, cranial neuropathy, optic neuropathy, and cavernous sinus infiltration with ophthalmoplegia. When it presents as anterior uveitis; eye redness, pain, photophobia and blurry vision are common complaints [17, 18]. Early in the disease course, dilated episcleral vessels, anterior chamber hyphema or hypopyon and circumferential engorgement of the iris can be seen [18, 19]. Examples in literature show: one eye with a non-granulomatous AU with vitritis and cystoid macular edema secondary to a mixed cellularity Hodgkin lymphoma, one eye with recurrent hyphema secondary to an iris mass from a mantle cell lymphoma, initially thought to be a UGH syndrome (ie., Uveitis-Glaucoma-Hyphema syndrome), and one case of peripheral non Hodgkin lymphoma presenting with pseudohypopion and iris nodules. Biopsies of the anterior segment lesions showed the intraocular tumor involvement [11,12,13].

In our experience, patients with hematologic malignancies can rarely present with anterior uveitis. In a case series we published of ocular involvement in six patients with hematological malignancies, we described one patient who had iris plasmacytoma and developed an anterior uveitis as a secondary presentation. The other patients in our series presented with a posterior ophthalmic involvement as pseudo-panuveitis or scleritis [20].

Diagnostic implications

Involvement of the iris and anterior chamber is a recognized but unusual manifestation of leukemia. Although vitreal, uveal, and orbital involvement of NHL are widely recognized, anterior chamber involvement is less frequent. The differentiating features between hypopyon of inflammatory origin and tumoral pseudohypopyon have been very interestingly outlined by Evereklioglu et al. [21] In a pseudohypopion, the perilimbal area of the eye is not dusky red but rather white. Macroscopic signs to the naked eye shows that the anterior chamber meniscus is heaped up at its edges. These anterior chamber collections contain neoplastic cells, such as the authors also find in their unique case upon an aqueous tab, which revealed the presence of atypical lymphoblast cells [22]. Tumoral pseudohypopyon contains a lower concentration of fibrinous exudate which is mobile in the direction of leaning which is a pathognomonic feature of “tumoral pseudohypopyon”. Finally, a pseudohypopyon is “blood-streaked pinkish” and not white-colored [21].

Ocular biopsies (anterior chamber, vitreous or orbital), depending on the location of the malignant ocular lesion, and imaging of the eye by brain MRI enables the diagnosis. Of note, we would recommend discussing with the hematology services, who can perform biopsy of the bone marrow for final leukemia diagnosis since diagnosis involves integrating the clinical features, sites of involvement (predominantly peripheral blood, bone marrow, liver, and spleen), and cellular characteristics, including morphology and immunophenotyping. Biopsy of the conjunctival lesion when associated with cells in anterior and posterior segments can show a nonspecific, mixed inflammatory infiltrate and a granuloma but no malignant cells [23]. Chronic myeloid leukemia can be confirmed by hematological investigations, i.e., blood smear revealing increased leucocyte count with presence of abnormal cells (myelocytes, band forms, and promyelocytes) and subsequent bone marrow trephine biopsy [24] rather than by cytopathology of the hypopyon (predominance of lymphocytes and few plasma cells) [25]. Pseudo-hypopyon with isolated anterior segment involvement is a rare clinical presentation of intraocular B-cell lymphoma [26]. A high index of suspicion along with insisting on anterior chamber sampling for cytology and flow cytometry analysis are essential to establish the diagnosis [26]. As discussed by Ramani et al., cytological examination of rapidly transported, unfixed vitreous specimens is considered the gold standard in exclusion of intraocular lymphoma in patients with idiopathic steroid resistant chronic uveitis. The majority of vitreoretinal lymphomas are of a DLBCL histologic subtype, though occasionally T-cell lymphomas can occur. They usually express CD19, CD20, CD22, PAX5, BOB.1, and OCT2 [27].

Vascular disease: Ocular Ischemic syndrome

Ocular Ischemic Syndrome (OIS) describes a constellation of clinical sequelae due to chronic, severe hypoperfusion in the setting of significant ipsilateral carotid or ophthalmic artery disease [28,29,30,31,32,33]. The classic etiology for this syndrome is vascular occlusion in the setting of severe atherosclerosis, typically greater than 90% occlusion of the ipsilateral carotid artery [29,30,31]. Other less common occlusive etiologies include Moyamoya [34, 35], giant cell arteritis [36,37,38], Takayasu arteritis [37, 39,40,41,42,43,44], optic disc melanoma [38] and radiation damage [44]. Patients may have orbital pain typified as a dull aching pain which can mislead the ophthalmologist for an inflammatory eye condition [30, 31, 45]. The posterior manifestations of mid-peripheral intraretinal hemorrhages and fluorescein angiography frequently displays delayed or patchy choroidal filling, retinal vascular staining and prolonged retinal arteriovenous transit time [34]. OIS can present as anterior uveitis in 18% of the patients [28, 30,31,32, 45,46,47,48]. In the anterior segment, rubeosis iridis and, neovascular glaucoma can be seen [32, 34]. Patients can present with anterior chamber cell and flare [34, 36, 46, 49]. Though it is important to note that cells and flare might co-occur in patients with OIS that also have rubeosis iridis [30]. The crux of its masquerading presentation lies in initial symptoms of ocular pain with anterior chamber cell and flare. However, given the cerebrovascular implications of severe carotid stenosis, a delay in diagnosis can have significant ramifications [28].

Diagnostic implications

Atherosclerotic risk factors including hypertension, diabetes, prior cerebrovascular accident (CVA), coronary artery disease increase suspicion to a diagnosis of OIS when subacute to acute vision loss. Iridis rubeosis in the setting of posterior pole ischemia as represented by retinal artery attenuation with venous dilation and scattered dot hemorrhages can differentiate OIS from classic anterior uveitis.

Infectious: syphilis

Ocular manifestations of ocular syphilis vary widely, but it most commonly presents as uveitis. It can present as any anatomic classification of uveitis with published reports documenting cases of anterior, intermediate, posterior and pan- uveitis. Despite the commonly held association between posterior uveitis and syphilis, it is important to note that syphilis is known as the great imitator and there are documented cases of anterior uveitis as well [50]. One case series found that 16.4% of ocular syphilis presented as anterior uveitis, 90% of which were in HIV positive patients [51]. We studied 21 cases (29 eyes) of ocular syphilis, finding only one patient that was affected with anterior uveitis representing 3.5% of the uveitis presentations [52] (Fig. 1). Most of the patients presented instead with a posterior uveitis in the form of posterior placoid chorioretinitis (58%) [53]. In our experience with a larger retrospective multicentric analysis of 95 patients treated for syphilic uveitis, 10 eyes, representing 14% of the cases, showed features of isolated anterior uveitis at presentation [54].

Fig. 1
figure 1

Slit-lamp photograph of a syphilitic gumma of the iris, a rare manifestation of tertiary syphilis

Diagnostic implications

In the non-specific AU patient, consider syphilis testing.

Post surgery: differential diagnosis of endophthalmitis after cataract surgery

Toxic Anterior Segment Syndrome (TASS)

Toxic Anterior Segment Syndrome (TASS), delayed-onset (chronic) endophthalmitis in the post-operative setting, retained lens material or lens-induced uveitis, de-hemoglobinized vitreous hemorrhage, and post-operative migration of triamcinolone in the anterior chamber are among the differential diagnosis for post-operative inflammation, presenting diagnostic and therapeutic challenges [55, 56].

Toxic anterior segment syndrome (TASS) is a syndrome that describes sterile anterior segment inflammation. It is most commonly associated with cataract surgery though there have been associations with other anterior and posterior segment surgeries [57,58,59,60,61]. The pathophysiology of TASS centers on the breakdown of the blood-aqueous barrier due to inflammation, specifically a breakdown in the tight junctions in the ciliary body and iris epithelium that separate the anterior chamber from the vasculature. Specific insults related to chemicals used in instrument processing cause an immune response resulting in anterior chamber inflammation [62]. Classically, it is known to present within 12-48 hours of recent intraocular surgery. Patients with TASS classically exhibit diffuse, “limbus-to-limbus” corneal edema. Anterior chamber inflammation with cell, flare, KPs and fibrin deposition are also found. In the acute setting, patients with TASS often exhibit a fixed and dilated pupil secondary to iris ischemia. This is also associated with decreased intraocular pressure due to decreased aqueous humor production.

Generally, TASS is a clinical diagnosis, made in the setting of a history of recent intraocular surgery, pertinent exam findings and laboratory data supporting a sterile inflammatory process.

Delayed-onset (chronic) endophthalmitis

While also typically infectious in etiology, it is distinct from acute infectious endophthalmitis in that it presents with recurrent episodes of low grade inflammation, 6 weeks or more after surgery [63]. Classically, a white plaque is seen between the intraocular lens and the posterior capsule. Over time, patients can develop hypopyon and vitritis. It can have both indolent bacterial or fungal causative organisms, with Propionibacterium species accounting for the majority of cases (41 to 63%) followed by coagulase-negative Staphylococcus, Corynebacterium, cutibacterium acnes, and others. Aspergillus species, Candida species, and Curvularia lunata are among the implicated fungal etiologies [63, 64]. Delayed-onset endophthalmitis cases are often managed by intraocular cultures (aqueous humor, and or vitreous) followed by repeated intravitreal antibiotics. Options for treatment of bacterial chronic bacterial endophthalmitis include diagnostic and therapeutic combined vitrectomy (PPV), intraocular antibiotics injection (IOAB), total capsulectomy, and removal or exchange of the intraocular lens (IOL). Current approach for chronic fungal postoperative endophthalmitis includes diagnostic and therapeutic PPV, intravitreal amphotericin or voriconazole, and a prolonged systemic antifungal drug [64].

Retained lens material or lens-induced uveitis

Lens associated uveitis represents another cause of post-operative inflammation. Disruption of the lens capsule results in a phacoantigenic uveitis, in which there is a granulomatous immune reaction centering on the site of lenticular injury and disruption. Leakage of lens protein often occurs in the setting of a hyper-mature cataract and results in a phacolytic uveitis as macrophages react to the dispersed protein. The resulting macrophage response results in a clogged trabecular meshwork and an acute increase in intraocular pressure.

In phacoantigenic uveitis, patients typically present with sudden or insidious ocular pain, redness, decreased visual acuity, and photophobia. In phacolytic uveitis patients present with acute onset of ocular pain, redness, and worsening vision [65,66,67,68].

Patients with phacoantigenic uveitis will present with an anterior uveitis with KPs, mild to moderate anterior chamber reaction and anterior vitreous inflammation without fundus involvement. Elevated IOP and posterior synechiae are also commonly present. Patients with phacolytic uveitis have conjunctival hyperemia, corneal edema, protein deposits (frequently mistaken for KPs) and anterior chamber cell and flare.

Diagnostic implications

Differentiating causes of post-operative inflammation in patients can be difficult with specific ramifications to the direction of treatment. TASS can, in many ways, mimic infectious endophthalmitis, specifically chronic endophthalmitis, whose time course of weeks to months overlaps with TASS [2, 63, 64, 69]. Of the two diagnoses, it is reasonable to initially rule out chronic post-operative endophthalmitis, especially given the likely surgical management required. Careful follow-up for resolution can parse TASS from chronic endophthalmitis, which will not typically resolve with corticosteroids over time [62,63,64, 69,70,71,72]. Lens-associated uveitis related inflammation can be triggered in the setting of mature cataract lenticular leakage or disruption of lens capsule intraoperatively. In some instances, inflammation from severe AU may cause a sterile hypopyon that mimics the appearance of an infectious endophthalmitis. Management entails a thorough work-up to exclude an infectious etiology prior to initial treatment with topical and/or systemic steroids [73]. Because of the post-operative clinical context and the common presenting ocular findings, the use of microbiological samples is a key differentiator. In other words, chronic postoperative endophthalmitis should always be in the differential of recurrent postoperative inflammation and vitreous or aqueous samples should be sent for analysis early enough.

Below we describe some entities that are uveitis/ anterior segment inflammation/ scleritis related but are commonly misdiagnosed.

Misdiagnoses among Uveitis/ Anterior segment inflammation entities

Tubulointerstitial Nephritis and Uveitis (TINU) syndrome

Tubulointerstitial Nephritis (TIN) and Uveitis (TINU) is a multi-system pathology of unknown cause that commonly presents as an acute-onset bilateral, non-granulomatous AU with accompanied acute nephritis [74]. Eye inflammation can manifest in different anatomical forms. The most common exam findings include anterior chamber cell (65%) and flare, and conjunctival injection [75]. Occasionally keratic precipitates and vitreous cell can be seen as well. Complications of TINU include posterior synechiae, optic disc swelling and cystoid macular edema [76] and involvement of the vitreous or choroid may also be present [77,78,79]. We have recently published a case of AU and bilateral papillitis associated with TINU showing that it remains a common diagnosis among children and adolescents [80].

Key criteria for TINU have been updated by the SUN working group in 2021 and includes anterior chamber inflammation and evidence of tubulointerstitial nephritis with either (1) a positive renal biopsy or (2) evidence of TIN and an elevated urine β-2 microglobulin [81].

Since patients with TIN may be asymptomatic or exhibit nonspecific symptoms (fever, abdominal pain) that do not lead to kidney function tests being performed, a diagnosis of TINU may be significantly delayed or unrecognized even after the onset of uveitis symptoms and ophthalmological assessment [82,83,84]. Mandeville et al. noted that ocular symptoms were concurrent with systemic symptoms in only 15% of cases; in 21% of cases, uveitis occurred before systemic symptoms, occurring up to two months beforehand; in 65% of cases, uveitis occurred after systemic symptoms with a median time of onset being 3 months, though it was noted to extend up to 14 months [76].

It is often suggested that most incidence and prevalence figures of TINU are likely to be under-estimates. For example, Mackensen et al. identified that in a cohort of 1985 patients, 26 had been diagnosed with TINU during routine care (prevalence of 1.3%). However, on subsequent analysis, a further 7 patients who had been labeled idiopathic were found to have features consistent with TINU based on the criteria of typical bilateral sudden onset AU with renal dysfunction (total prevalence of 1.7%). They also identified that there were a further 18 ‘idiopathic’ pediatric cases in which the uveitis was typical for TINU but in whom there had not been adequate laboratory investigations to rule in or rule out the diagnosis, leading to the possibility that the real prevalence is even higher [76, 82].

Diagnostic implications

The multi-system presentation of TINU can mimic other auto-immune pathologies including sarcoidosis, Sjögren’s syndrome, systemic lupus erythematosus, granulomatous polyangiitis, Behcet’s disease, tuberculosis and syphilis [76, 85]. The inconsistency of presentation timeline among cases can compound the diagnostic difficulty, more than half of the patients develop ocular symptoms after the acute interstitial nephritis [76]. This diagnosis should be promptly suspected in young patients presenting with either uveitis or TIN. A definite diagnosis is confirmed through renal biopsy.

Immune reconstitution inflammatory syndrome

Immune Reconstitution inflammatory syndrome (IRIS) is observed in patients who have a diagnosis of AIDS with opportunistic infections in the setting of Acquired Immunodeficiency Syndrome (AIDS) with recovering CD4+ T cell counts on Highly Active Antiretroviral Therapy (HAART). While IRIS generally a broad spectrum of presentation, its ocular manifestation is most classically seen in patients with CMV retinitis and presents as uveitis. Ocular presentations of IRIS are termed immune recovery uveitis. The most common presenting signs are cystoid macular edema, epiretinal membrane, papillitis, and vitritis. However, there have notably been published cases of anterior segment inflammation. A review of Jabs et al. on ocular complications in patients with AIDS found that a significant proportion of those with immune recovery uveitis (IRU) had some degree of anterior inflammation, with up to 6.6% having posterior synechiae [86]. There were also a small proportion of patients with keratic precipitates on exam.

Diagnostic implications

Patients with AIDS are at a high risk for a diverse range of opportunistic infections. Consideration of IRU in HIV+ positive patients with uveitis is critical in managing CMV-related therapy and appropriate initiation of topical corticosteroids.

Cytomegalovirus (CMV)- related anterior uveitis

In immunocompetent patients, CMV presents as recurrent, acute or chronic, occasionally bilateral, AU often with associated ocular hypertension, and corneal endotheliitis [2, 87, 88], however it can have a broad spectrum of presentation. It has been documented to present as Posner-Schlossman Syndrome (PSS), an acute, recurrent hypertensive AU with few granulomatous keratic precipitates [88], and Fuchs uveitis syndrome (FHI), a chronic hypertensive AU with moderate anterior chamber reaction and diffuse, fine stellate keratic precipitates often involving the entire endothelial surface (Fig. 2). This later presentation appears to be more common in older patients, with a mean age of 65 years old [89]. Other forms include a chronic hypertensive AU with few, inferiorly-located, brown keratic precipitates and corneal endotheliitis with coin-shaped lesions (Fig. 3) or linear keratic precipitates (KPs) [87, 90].

Fig. 2
figure 2

Slit-lamp photograph of an eye with Fuchs' heterochromic iridocyclitis. A Low magnification showing the typical stellate keratic precipitates, which are better delineated in high magnification in B) and retro-illumination in C)

Fig. 3
figure 3

A Slit-lamp photograph of a coin-shaped keratic precipitate in the left eye of a 72-year-old female with Cytomegalovirus anterior uveitis. Diagnosis was confirmed by real-time PCR (RT-PCR) on aqueous humor samples. B Magnified image of the coin- shaped keratic precipitate. C Note in her right eye, few coin-shaped keratic precipitates

Of note, Fuchs heterochromic iridocyclitis was once thought to be a non-infectious, idiopathic entity. Infection with herpes simplex virus, ocular toxoplasmosis, cytomegalovirus, rubella virus, and other viruses have been implicated in the pathogenesis of the disease [91,92,93].

With regards to the relative prevalence of CMV- related AU presentations, we showed in our own series of 38 patients that features of Posner-Schlossman syndrome were observed in 50% of the eyes, Fuchs heterochromic iridocyclitis in 13% of the eyes, chronic nonspecific AU in 21% of the eyes, and corneal endotheliitis in 18% of the eyes [94]. Diagnosis usually relies on aqueous fluid viral PCR or Goldmann-Witmer coefficients positive for CMV [2, 87, 88, 95].

Typically, the differential for AU with ocular hypertension includes herpes simplex virus (HSV), and varicella zoster virus (VZV). There are, however, several signs that can differentiate AU caused by these three herpesviridae. Demographically, patients with CMV-AU are predominately male and older in age. Conversely, mutton-fat KPs are found predominantly in patients with HSV and VZV. With regards to the overall clinical severity of AU and viral load, the highest of both is typically in VZV-AU, followed by HSV-AU and lastly, CMV-AU. Iris atrophy is often observed in HSV-AU and VZV-AU typically with round type and sector type morphology, respectively [96,97,98].

Diagnostic implications

Parsing the diagnosis often relies on the distinct differences in anterior chamber reaction. In CMV patients, KPs are common between the entities and difficult to use as a discerning feature. In case of Fuchs heterochromic iridocyclitis, differentials include multiple infectious etiologies among them cytomegalovirus (CMV), and rubella virus (RV). Diagnostic anterior chamber paracentesis typically can be analyzed to differentiate among CMV, HSV, VZV (by Reverse Transcriptase Polymerase Chain Reaction (RT-PCR; Goldmann-Witmer coefficient (GWc)) and rubella (PCR).

HLA-B27 anterior uveitis mimicking infectious endogenous endophthalmitis

One of the most prevalent etiologies for acute AU is acute unilateral AU related to human leukocyte antigen B27 (HLA-B27) [99, 100] a product of the major histocompatibility complex (MHC). It is a genotype associated with a variety of spondylarthropathies. Classically, patients with HLA-B27 AU present with either an acute unilateral episode, or recurrent, unilateral and alternating episodes [101,102,103] or less typical presentations of chronic or persistent AU, episcleritis and scleritis [104]. The presence of a fibrinous reaction or hypopion due to severe uveitis in acute HLA-B27 AU obscures the vitreous and retina, and when taken in isolation, can be diagnostically difficult to discern from other uveitis or pseudo-uveitis causes like Behcet’s disease, lymphoma, leukemia or importantly, endogenous endophthalmitis [105, 106]. A Bscan ultrasound can be helpful in these instances and if endogenous endophthalmitis or other infection is suspected, an anterior fluid paracentesis and aqueous fluid analysis should be performed to rule out the differentials.

In our experience, and as validated by the literature, HLA-B27 AU with severe anterior chamber inflammation can be misdiagnosed for endogenous endophthalmitis especially in cases where fibrin and/or hypopyon obscure the fundus view (Fig. 4) [107, 108].

Fig. 4.
figure 4

A Slit-lamp photograph of an eye with HLA-B27 associated uveitis. Note the fibrin and sterile hypopyon in anterior segment. B-D Slit-lamp of an eye post-trabeculectomy associated endophthalmitis. Note the presence of ciliary flush and conjunctival injection

Endogenous endophthalmitis describes endophthalmitis secondary to hematogenous spread. It represents roughly 2-20% of all cases of endophthalmitis [109,110,111]. It classically affects the posterior chamber with findings including retinitis, chorioretinitis, vitreous haze and cell, retinal detachment and retinal periphlebitis. It also involves patients with a history of intravenous drug use that self-inoculate, causing a Gram positive bacteremia. Other etiologies for endogenous endophthalmitis include indwelling catheters, endocarditis or immunosuppression, i.e. HIV/AIDS, diabetes mellitus, chronic corticosteroid use, and malignancy [109,110,111,112,113,114].

Diagnostic implications

A patient’s comorbidities can help in differentiating HLA-B27 from endogenous endophthalmitis. Age of onset differs between both uveitidies, with the median age for patients with endogenous endophthalmitis ranging from early fifties to mid-sixties, which is older than for patients with uveitis related to HLAB-27 [109, 111, 113, 115,116,117]. The symptoms are the same between both uveitis entities. Decreased visual acuity, pain and injection are common, though relatively non-specific symptoms for patients with endophthalmitis [118]. However, definitive diagnosis is typically made with culture from intravitreal specimens [108, 118,119,120].

Tattoo associated uveitis

Tattoo-Associated Uveitis is a rare entity that can be often overlooked by ophthalmologists. It is seen in patients with cutaneous tattoos in any area of the body with subsequent bilateral, granulomatous AU. Both the site of the tattoo and the eye are concomitantly inflamed. Currently, the disease is postulated to either be a form of delayed hypersensitivity or systemic sarcoidosis. The overall prevalence is low with its presence documented only in the form of case series [121,122,123,124,125,126,127,128].

Although bilateral, anterior granulomatous uveitis is a more common presentation, it is important to acknowledge that panuveitis with retinochoroiditis, and retinal vasculitis have been documented; along with cases of macular and optic disc edema [126].

Diagnostic implications

Bilateral anterior granulomatous uveitis immediately prompts a differential of sarcoidosis, tuberculosis, VKH, Lyme and syphilis. In these patients, it is important to elicit a history of recent tattooing and to perform a detailed dermatologic exam.

Drug related uveitis

Moorthy et al. have provided a comprehensive review of medications associated with uveitis [129] after intraocular injection of antibiotics, urokinase, plasmin/ microplasmin, and antibodies (ranibizumab, bevacizumab, etc.). As such, a non-granulomatous AU was noted in 17% to 89% of cases and hypotony in approximately 10% of cases after the use of cidofovir for the treatment of CMV [129]. There have been several reports of anterior chamber inflammation associated with intraocular injection of anti-VEGF: pegaptanib sodium, ranibizumab (0.3-1.5%) [130,131,132,133,134] and bevacizumab (0.5-1.1%) [132, 135].

Anti- VEGF medications

Brolucizumab (Beovu) is a novel anti-VEGF molecule, a humanized single- chain variable antibody fragment (scFv). Compared to aflibercept in eyes with diabetic macular edema (DME). KITE, KESTREL and KINGFISHER, phase III studies found an intraocular inflammation in 2.2% to 4.7% of cases with brolucizumab (3 - 6 mg) versus 0.5% to 1.7% with aflibercept (2mg). These studies report a posterior involvement in the form of a retinal vasculitis in up to 1.6% [136,137,138]. According to the approved label, brolucizumab is contraindicated in eyes with active intraocular inflammation [139]. Phase III trials of brolucizumab for neovascular age macular degeneration (AMD) have also reported higher frequencies of intraocular inflammation [140] (4.6%), including retinal vasculitis (2.1%) and retinal vein occlusion, compared to the aflibercept [140]. Although the cause of ocular inflammation with brolucizumab is unknown, the delayed onset (30-53 days) seems to signal an immune (Type III/IV hypersensitivity), rather than a toxic or infectious cause [140, 141]. Approximately 90% of the uveitis and iritis cases related to brolucizumab were mild to moderate and treated with a course of topical corticosteroids/anti-infectives in phase 3 trials (HAWK and HARRIER) [142]. Subclinical anterior chamber inflammation may occur at rates as high as 20% after intravitreal anti-VEGF injection. Rates of acute onset sterile inflammation have been reported to range from 0.05–2.1%, 0.05–1.1%, and 0.005–1.9% in aflibercept, bevacizumab, and ranibizumab, respectively with clinical manifestations of anterior chamber and/or vitreous cavity inflammation [143]. Many mechanisms have already been put forward to explain the pathogenesis of inflammation following the injection of anti-VEGF medications. These mechanisms can be divided into 3 causes: patient-specific susceptibility, medication specific impurities, non-human proteins, the formulation in which the drug is administered, the immunogenic properties of the actual anti-VEGF antibody itself and protein aggregates may play a role in inflammation caused by these agents [143].

Recently, intravitreal injections of pegcetacoplan (15 mg) to slow progression of atrophy in AMD have shown an incidence of 3% intraocular inflammation, including anterior chamber cells, iritis, and anterior chamber flare, in the DERBY and OAKS phase 3 randomized controlled clinical trials [138].

It is current knowledge that a severe inflammation following intravitreal injection of triamcinolone acetonide, also called sterile endophthalmitis can happen (Fig. 5). From several reports, Moorthy et al. highlight the substantial anterior chamber cell involvement, often with hypopyon and they evaluated its incidence as occurring in 0.5% to 9.7% of injections [129].

Fig. 5
figure 5

A Slit-lamp photograph of an eye with sterile hypopyon following the injection of intravitreal triamcinolone. B-C Slit-lamp photograph showing the triamcinolone acetonide crystals in the anterior chamber of a pseudophakic eye following intravitreal triamcinolone injection in another patient

Topical glaucoma medications

The same authors have also reviewed uveitis associated with topical instillation of the following: prostaglandin analogs, metipranolol, cholinomimetics, antibiotics, betaxolol, cholinesterase inhibitors. Topical prostaglandin analogs latanoprost, travoprost, and bimatoprost are used to lower the intraocular pressure primarily by increasing uveoscleral outflow have been associated with conjunctival hyperemia, reactivation of herpes simplex keratitis, AU along with cystoid macular edema, iris and periocular skin pigmentation, and iris cyst formation [129]. The mechanism by which prostaglandin analogs might cause AU may involve the downstream stimulation of proinflammatory eicosanoids [144].

Brimonidine tartrate is a selective alpha2-adrenergic receptor agonist that is used for the treatment of ocular hypertension. Of note, there are few case reports describing the occurrence of a granulomatous AU with elevated intraocular pressure associated with chronic use of brimonidine [129].

Anti-neoplastic medications

Antineoplastics, both specific targeted therapies and immunotherapies can involve all parts of the uvea from anterior to posterior [145]. BRAF/MEK inhibitor therapies are associated with a significant increase in the risk of uveitis, either anterior, intermediate uveitis with or without macular edema, papilledema. The mean probability of developing uveitis during 1 year of treatment has been reported between 0.8% and 5% [145,146,147]. Similarly, erlotinib, a first-generation small-molecule tyrosine kinase inhibitor which reversibly inhibits the kinase domain of epithelial growth factor receptor (EGFR) and is used in treatment of advanced non-small cell lung cancer has been reported as the cause of AU in a small number of cases [148, 149]. Anterior uveitis alone or with posterior segment involvement including vitritis, cystoid macular edema, sub-retinal fluid, serous retinal detachment or papillitis have been reported secondary to immune checkpoint inhibitor cytotoxic T-Lymphocyte antigen (CTLA-4) and anti PD-1 therapy, each separate or in combination anti-CTLA-4/anti-PD-1 therapy [150,151,152,153].

Others

In their review of medications associated with uveitis, Moorthy et al. implicated the systemic use of sulfonamides, diethylcarbamazine, fluoroquinolones, oral contraceptives, topiramates, rifluoperazine, quinidine, ibuprofen, reserpine, sildenafil, and clomiphene citrate [129]. Among uveitis associated with systemic fluoroquinolone therapy, the most commonly implicated agent is moxifloxacin. The associated AU presents as fine, pigmented KPs with prominent pigment in the anterior chamber with only minimal non-pigmented cell, diffuse iris transillumination defects with atonic pupils and ocular hypertension or glaucoma in up to 50% of patients [153,154,155]. The pathophysiology of fluoroquinolone-associated uveitis is unknown but Moorthy et al. have hypothesized phototoxicity, autoimmune predisposition, and/or concurrent viral infection [129].

Rifabutin is an oral bactericidal antibiotic used as prophylaxis against Mycobacterium avium complex. Rifabutin-associated uveitis is more commonly an AU with hypopyon [129]. The risk factors associated with its occurrence are dosage and duration of rifabutin therapy, low body weight, and use of concomitant medications, including clarithromycin and ritonavir through inhibition of hepatic cytochrome P-450 [156,157,158]. Moreover, bisphosphonates have been noted to cause conjunctivitis, acute non-granulomatous AU, and scleritis/episcleritis in some patients. Intravenous pamidronate sodium is the bisphosphonate most frequently associated with uveitis [129].

Paradoxical inflammation is more common with etanercept and is much less common with monoclonal antibody type anti-TNF agents [159].

Post vaccine

Multiple case reports have implicated the following vaccines in the development of acute bilateral AU Bacille Calmette-Guérin (BCG), influenza, Hepatitis B, measles, mumps, and rubella (MMR), diphtheria, tetanus, and pertussis (DPT), varicella, and smallpox [129].

Recent post mRNA COVID-19 vaccinations

Numerous reports have been made of cases of anterior, episcleritis, scleritis, but also posterior/ panuveitis after mRNA COVID-19 vaccinations [160]. According to the US Food and Drug Administration Vaccine Adverse Event Reporting System, anterior chamber inflammation (1), conjunctivitis unspecified and viral (6), corneal endotheliitis (1), episcleritis (3), eye inflammation (16), eye edema (1), eye swelling (7), herpes ophthalmic (2), iridocyclitis (33), iritis (13), ocular hyperemia (36), uveitis (155), 851 cases of uveitis after COVID-19 vaccination involved mRNA or adenovirus vector vaccines from the reports processed as of June 24, 2022 [161]. A large multinational case series including 70 patients has shown that the most common events were AU (58.6%), followed by posterior uveitis (12.9%) and scleritis (10.0%). The mean time to event was 5 days and 6 days (range, 1-14 days) after the first and second dose of vaccine, respectively. Among all patients, 36 (54.1%) had a previous history of ocular inflammatory event [162].

In 2022, Haseeb et al. did a retrospective review of 58 studies describing adverse ocular manifestations following any vaccination against COVID-19 between December 2020 and December 2021 for a total of 94 patients included. Of the 87 cases in which vaccine information were present, BNT162b2 mRNA SARS-CoV-2 (BioNTech/Pfizer, Mainz, Germany) was reported 55 (63.2%) times, AZD1222 ChAdO×1 nCoV-19 (AstraZeneca, Cambridge, UK, also marketed as the CoviShield Serum Institute of India vaccine) was reported 20 (22.9%) times, Moderna COVID-19 Vaccine (ModernaTX, Inc., Cambridge, MA, USA) was reported 6 (6.9%) times, BBIBP-CorV (Sinopharm, Beijing, China) was reported 3 (3.4%) times, Corona Vac (Sinovac Biotech Ltd., Beijing, China) was reported 2 (2.3%) times, and Gam-COVID-Vac/Sputnik V (Gamaleya Institute, Moscow, Russia) was reported once (1.1%) [163].

There has been also reports of reactivated uveitis and kerato-uveitis related to underlying infectious disease, herpetic disease, or pre-existing uveitis-related diseases (including ankylosing spondylitis, psoriasis, Crohn’s disease, and herpes zoster (VZV) ophthalmicus) after COVID-19 vaccines [164,165,166,167,168,169,170,171].

Several reports have described complications in the form of conjunctival or ciliary injection, corneal graft edema, Descemet’s folds, cells in anterior segment and scattered KPs involving unilateral and bilateral corneal transplant rejections occurring 2 days to 2 weeks following vaccination to COVID-19 with mRNA vaccine BNT162b2, AZD1222, Moderna anti-SARS-CoV-2 or ChAdOx1 nCoV-19 Corona Virus Vaccine Recombinant, COVISHIELD™ [172,173,174,175,176,177]. Nearly all types of anterior segment transplantation have been linked with failure in the setting of recent vaccination, including penetrating keratoplasty, descemet membrane endothelial keratoplasty, descemet stripping automated endothelial keratoplasty, and living-related conjunctival-limbal allograft [178, 179].

Diagnostic implications

The occurrence of AU and less frequently of posterior uveitis or scleritis especially if previous history of ocular inflammatory event (i.e., herpetic, herpes zoster (VZV) ophthalmicus, ankylosing spondylitis, psoriasis, Crohn’s disease) is suspicious of a COVID-19 related uveitis if it happens within a time range of 5 days to 14 days after the first or second dose of vaccine. One might think also of largery prescribed topical antiglaucoma eye drops when AU occurrence. BRAF/MEK/ CHECK inhibitor therapies are associated with a significant increase in the risk of uveitis, either anterior, intermediate uveitis with or without macular edema, papilledema.

Recently, intravitreal injections of brolizumab for exudative AMD and pegcetaplan for atrophic AMD have been involved in uveitis.

Masquerade presentation of anterior segment inflammation: peripheral ulcerative keratitis misdiagnosed for an infectious bacterial ulcer

Peripheral Ulcerative Keratitis (PUK) is a destructive, corneal inflammatory process. Importantly, it can be either an infectious or sterile process. Infectious etiologies include bacteria such at Staphylococcus, and Streptococcus, viruses such as HSV and VZV, and fungi. Non-infectious PUK is typically tied to autoimmune systemic pathology such as rheumatoid arthritis, systemic lupus erythematosus, granulomatosis with polyangiitis and polyarteritis nodosa. Corneal ulcers secondary to herpetic infection more commonly mimic autoimmune PUK. Thus, a unilateral PUK in an elderly individual should raise suspicion of herpetic keratitis, prompting the search for other signs indicative of herpetic keratitis, such as endotheliitis or loss of corneal sensitivity. Bacterial keratitis typically occurs in contact lens wearers, with a usually intact cornea and limbus. Fungal keratitis should be suspected in contact lens wearers, patients with chronic ocular surface conditions, or those undergoing prolonged local corticosteroid therapy.

Patients classically present with a unilateral, crescent shaped ulcer in the peripherally cornea, though bilateral presentations are not uncommon. There is often stromal thinning and associated scleritis. Much of the morbidity centers around incomplete treatment which can lead to stromal melting, especially in patients with PUK secondary to systemic autoimmune disease. In patients with PUK and associated scleritis it is also not uncommon to find AU as well [180,181,182].

Diagnostic implications

Both PUK and bacterial keratitis can present as peripheral ulcer with patient complaints of redness, pain and photophobia. On exam, corneal ulcers present with conjunctival injection, corneal thinning, Descemet’s folds and stromal edema. Clinical suspicion for PUK is warranted in patients with a history of autoimmune disease, especially rheumatoid arthritis. Patients will often have concurrent systemic symptoms associated with their autoimmune pathology. Contextualizing exam findings with a thorough history inclusive of extraocular symptoms can parse PUK from bacterial keratitis, as well as scraping of corneal ulcer for culture and sensitivity and special stains to rule out infection.

Conditions that may present as scleritis masqueraders

Drug induced: biphosphonates

Specific drugs have been shown to induce scleritis. An investigation leveraging data from the National Registry and the World Health Organization found a significant amount of case reports with bisphosphonate induced scleritis [183]. Bisphosphonates have been circumstantially implicated in a diverse range of inflammatory presentations beyond scleritis that are also important to consider [184,185,186,187,188].

Neoplastic conditions masquerading as scleritis: squamous cell carcinoma, choroidal melanoma, adenocarcinoma

Though largely limited to case reports, neoplastic conditions like choroidal melanoma or undifferentiated carcinoma, conjunctival lymphomas, and ocular surface squamous neoplasia can masquerade as scleritis [186, 189,190,191]. Corticosteroid-resistant scleritis should prompt a scleral biopsy to investigate for neoplasia. Herpetic origin is also a differential diagnosis for scleritis.

Conjunctival lymphoma has been classically described as a painless, nodular lesion with salmon-pink or fleshy patches at the fornix or bulbar conjunctiva. It is slow-growing, and feeder vessels are not typically seen. Other clinical manifestations such as ptosis, diplopia, or a palpable mass have been reported. Systemic involvement was observed in 20% to 31% of conjunctival cases [192]. The relatively asymptomatic presentation and indolent course of the disease often result in a delay in diagnosis.

Conjunctival epidermoid carcinoma is a rare tumor affecting mainly the perilimbal region of the bulbar conjunctiva that can mimic a Mooren pseudo-ulcer. Conjunctival epidermoid carcinomas typically have a raised and avascular appearance.

Parasitic ocular infections

Helminth species (e.g., Brugia spp., Thelazia spp., Dirofilaria spp., and Wuchereria spp.) may infest human eyelids, conjunctival sacs, lacrimal glands and, in some cases, the ocular globe. Human ocular filariasis, caused by Onchocerca species, have been found in fibrous tissue masses underneath conjunctiva mimicking scleritis [193, 194].

Inflammatory systemic disease: scleritis and intraocular involvement in IgG4-related ophthalmic disease (IgG4-ROD)

IgG4-ROD is a rare progressive disease hallmarked by chronic immune activation across many organ systems including the orbit with resultant tissue fibrosis. The chronic inflammation and fibrosis often cause diffuse enlargement of affected tissues mimicking a neoplastic etiology. IgG4-ROD orbital manifestations include lacrimal gland enlargement, extraocular muscle enlargement and it most commonly presents as inflammation of orbital and scleral tissues, but is also a rare cause of uveitis [195,196,197,198,199,200,201,202].

Foreign bodies

Non-absorbable sutures from strabismus surgery in childhood have been reported to result in inflamed episcleral granulomas mimicking severe nodular anterior scleritis [203]. Episcleral nodules in the form of lipid granulomas caused by silicone oil leakage near entry sites of vitrectomy have been also reported [204].

Diagnostic implication

Cases of scleritis non-responsive to treatment should be re-evaluated for a possible neoplastic etiology. A detailed medication review should be performed in initial evaluations of uveitis patients.

In patients with significant conjunctival infiltration, nodular development or chronic relapsing scleritis, IgG4 related disease should be elevated to diagnostic consideration. Diagnosis requires biopsy and histopathologic confirmation.

Misdiagnosed among scleritis

Necrotizing scleritis without inflammation (scleromalacia perforans) (differential with scleral hyaline plaque)

Scleromalacia perforans is a rare type III hypersensitivity reaction characterized by the presence of anterior necrotizing scleritis without inflammation. It presents as scleral plaques without significant vascular congestion or injection. It is commonly associated with rheumatoid arthritis [205]. It has also reported in Behcet’s disease [206, 207], ulcerative colitis [208] and graft versus host disease [209]. It can progress rarely to a staphyloma in cases of elevated intraocular pressure. The progressive thinning can lead to eventual spontaneous perforation. Medical management of patients with necrotizing scleritis require systemic therapy with steroids and immunosuppression (often cyclophosphamide or biologic agents), as well as control of associated systemic disease. In severe cases with exposure of underlying uvea, tectonic patch grafting may be required [210, 211].

Diagnostic implications

The difficulty in diagnosis of scleromalacia perforans centers on its indolent appearance. The lack of inflammation allows cases of scleromalacia perforans to be mistaken for senile scleral hyaline plaques. Keys to appropriate diagnosis include consideration of systemic comorbidities such as rheumatoid arthritis. Monitoring of scleral plaques for progression and coalescence can also point towards scleromalacia perforans as opposed to a diffuse scleral hyaline plaque.

Surgically induced necrotizing scleritis

Surgically induced necrotizing scleritis (SINS) can occur after ocular surgeries with a variable latency period and is diagnosed clinically when ischemia or vascular nonperfusion accompanies scleritis with or without active inflammation. It is thought to be a immune-complex hypersensitivity reaction towards an antigen that is either revealed or altered by ocular surgery [212]. Importantly, cases of SINS have been reported as infectious, predominantly in pterygium surgeries [213]. Although SINS can be a harbinger of underlying systemic disease after cataract/lens procedures, a search for contributing underlying autoimmunity will most often prove unrevealing with SINS following other surgeries, particularly following pterygium surgery [213].

SINS is more commonly associated with the use of antimetabolites, for example, mitomycin C in pterygium surgery or trabeculectomy. However, SINS has been reported after a variety of procedures, including cataract surgery, strabism surgery, corneal grafting, vitrectomy and pterygium excision with conjunctival autograft [207,208,209,210,211,212,213,214,215,216,217].

Diagnostic implications

Although SINS has been reported after a many surgical procedures, SINS is more commonly associated with the use of metabolites in pterygium surgery or trabeculectomy. Treatment decisions should be informed, therefore, both by the low rate of underlying systemic autoimmunity and the high risk of infection, particularly by Pseudomonas and fungal organisms. An infectious origin is the primary etiology to consider in the case of scleritis following ocular surgery. Samples should be taken, and antibiotic therapy initiated.

Conclusion

In this review, we described the most representative ophthalmologic diseases that manifest with intraocular inflammation that mimic AU/ scleritis, including those belonging to the group of immune-mediated or infectious uveitis entities, classically named as “Uveitis Masquerade Syndromes” and we also focused on misdiagnoses among uveitis/scleritis/ anterior segment inflammation entities. In the case of recurrent or persisting ocular inflammation, it is critical to prove the underlying diagnosis if neoplastic (i.e., lymphoma, leukemia or metastatic solid tumors) in order to initiate the appropriate therapy and avoid a systemic spread and consecutive deterioration of the prognosis. Anterior segment infiltration of the eye is an uncommon complication of acute lymphoblastic leukemia and portends a poor prognosis. Systematic reviews underscore the rarity of such cases of anterior segment infiltration.

Early recognition, systemic work-up, and prompt treatment of patients with malignancies with ocular relapse is likely an essential step in improving survival outcomes in these aggressive diseases.

However, other post surgical conditions have clinical features that can also mimic AU or scleritis. If an endogenous endophthalmitis is suspected, the diagnosis must be confirmed by obtaining intraocular (aqueous, vitreous) specimens. Delay of treatment with trial of topical corticosteroids can result in disease progression and worsening of symptoms.

We also showed that previous studies have also reported that HLA B27-associated panuveitis could mimic an endophthalmitis.

The recent reports of cases of AU, after COVID-19 vaccines are particularly worrisome since apart from being potentially sight threatening, this can represent a contraindication on future COVID-19 vaccinations.

A high index of suspicion must be maintained for masqueraders of AU and scleritis, which can include serious inflammatory, infectious, and neoplastic disorders. Scleritis has a broad differential that should be worked through systematically. The differential diagnosis for anterior scleritis includes episcleritis and severe microbial conjunctivitis.

Careful review of possible past uveitis history, current medications and recent vaccinations, detailed examination of signs of past or present inflammation and multimodal retinal imaging are required for the correct diagnosis.

Making the correct diagnosis for AU, scleritis and its masqueraders can be challenging even for the experienced uveitis or cornea specialist, and one must maintain a broad differential initially in order to catch rare entities. The above outline provides insights into distinct clinical features and specific findings on ocular biopsies and anterior chamber paracentesis for cytopathologic confirmation. If anterior segment infiltration is suspected in a patient with an history of malignancy or hemopathy, cytological examination should be performed. Moreover, the use of topical corticosteroids before the first anterior chamber paracentesis may delay cytopathologic diagnosis by reducing the number of tumor cells in the aqueous humor.

Availability of data and materials

Not applicable.

Abbreviations

AU:

Anterior uveitis

OIS:

ocular ischemic syndrome

PIOL:

Primary intraocular lymphoma

NHL:

Non-Hodgkin Lymphoma

BL:

Burkitt Lymphoma

EBV:

Epstein-Barr Virus

CVA:

Cerebrovascular accident

HIV:

Humman Immunodeficiency Virus

TASS:

Toxic anterior segment syndrome

KP:

Keratic precipitate

TIN:

Tubulointerstitial Nephritis

TINU:

Tubulointerstitial Nephritis and Uveitis

UWFFA:

Ultra-widefield fluorescein angiography

OCT:

Optical Coherence Tomography

IRIS:

Immune reconstitution Inflammatory syndrome

AIDS:

Acquired immunodeficiency syndrome

HAART:

Highly active antiretroviral therapy

CMV:

Cytomeglaovirus

FHI:

Fuchs heterochromic iridocyclitis

PSS:

Posner-Schlossman syndrome

HSV:

Herpes simplex virus

VZV:

Varicella zoster virus

HLA-B27:

to human leukocyte antigen B27

MCH:

Major histocompatibility complex

VKH:

Vogt-Koyanagi-Harada disease

Anti-VEGF:

Anti-Vascular Endothelial Growth Factor

EGFR:

Epithelial Growth factor receptor

BCG:

Bacille Calmette-Guérin

COVID-19:

Coronavirus Disease of 2019

PUK:

Peripheral Ulcerative Keratitis

SINS:

Surgically Induced necrotizing scleritis

References

  1. Jabs DA, Nussenblatt RB, Rosenbaum JT, Standardization of Uveitis Nomenclature Working, G (2005) Standardization of uveitis nomenclature for reporting clinical data. Results of the First International Workshop. Am J Ophthalmol 140:509–516

    Article  PubMed  Google Scholar 

  2. Russell W. Read, N.A., Ralph D. Levinson, P. Kumar Rao, H. Nida Sen, Jonathan D. Walker, Daniel V. Vasconcelos-Santos. 2020-2021 BASIC AND CLINICAL SCIENCE COURSE (BCSC), SECTION 09: Uveitis and Ocular inflammation, (American Academy of Ophthalmology, 2017)

  3. Moher D, Liberati A, Tetzlaff J, Altman DG, Group, P (2010) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Int J Surg 8:336–341

    Article  PubMed  Google Scholar 

  4. Wakefield D, Chang JH (2005) Epidemiology of uveitis. Int Ophthalmol Clin 45:1–13

    Article  PubMed  Google Scholar 

  5. Tsirouki T et al (2018) A focus on the epidemiology of uveitis. Ocul Immunol Inflamm 26:2–16

    Article  PubMed  Google Scholar 

  6. Badeeb O et al (1992) Leukemic infiltrate versus anterior uveitis. Ann Ophthalmol 24:295–298

    CAS  PubMed  Google Scholar 

  7. Carbonell D et al (2021) Consensus recommendations for the diagnosis of vitreoretinal lymphoma. Ocul Immunol Inflamm 29:507–520

    Article  PubMed  Google Scholar 

  8. Takase H et al (2022) Challenges in the diagnosis and management of vitreoretinal lymphoma - clinical and basic approaches. Prog Retin Eye Res 90:101053

    Article  CAS  PubMed  Google Scholar 

  9. Nguyen QL, Reynolds SB, Piri N, Rivas Perez HL (2021) Ophthalmic anterior segment metastasis masquerading as uveitis. BMJ Case Rep 14

  10. Choi JY, Kafkala C, Foster CS (2006) Primary intraocular lymphoma: a review. Seminars Ophthalmol 21:125–133

    Article  Google Scholar 

  11. Ayhan Z, YildirimKarabag R, Alacacioglu I, Kaynak S, Kocak N (2017) Rare clinical sign of hodgkin’s lymphoma: ocular involvement. Turk J Ophthalmol 47:110–112

    Article  PubMed  PubMed Central  Google Scholar 

  12. Verity DH, Graham EM, Carr R, van der Walt JD, Stanford MR (2000) Hypopyon uveitis and iris nodules in non-Hodgkin’s lymphoma: ocular relapse during systemic remission. Clin Oncol (R Coll Radiol) 12:292–294

    CAS  PubMed  Google Scholar 

  13. Gauthier AC, Nguyen A, Munday WR, Xu ML, Materin MA (2016) Anterior Chamber Non-Hodgkin Lymphoma of the Iris Masquerading as Uveitis-Glaucoma-Hyphema Syndrome. Ocular Oncol Pathol 2:230–233

    Article  Google Scholar 

  14. Touhami S et al (2019) Neoplasia and intraocular inflammation: from masquerade syndromes to immunotherapy-induced uveitis. Prog Retin Eye Res 72:100761

    Article  PubMed  Google Scholar 

  15. Shields CL, Ghassemi F, Tuncer S, Thangappan A, Shields JA (2008) Clinical spectrum of diffuse infiltrating retinoblastoma in 34 consecutive eyes. Ophthalmology 115:2253–2258

    Article  PubMed  Google Scholar 

  16. Richards WW (1968) Retinoblastoma simulating uveitis. Am J Ophthalmol 65:427–431

    Article  CAS  PubMed  Google Scholar 

  17. Monsalvo S et al (2017) Unilateral uveitis masquerade syndrome caused by diffuse large B-cell lymphoma diagnosed using multiparametric flow cytometry of the aqueous humor. Cytometry Part B, Clinical Cytometry 92:286–290

    Article  CAS  PubMed  Google Scholar 

  18. Bell J et al (2016) Concurrent uveal and vitreoretinal lymphoma masquerading as anterior uveitis. Ocul Immunol Inflamm 24:120–123

    Article  PubMed  Google Scholar 

  19. Batis V, Shuttleworth J, Shuttleworth G, Williams GS (2019) Diagnostic dilemma of ocular lymphoma. BMJ CaseRep 12

  20. Denier M et al (2022) Hidden in the eyes-recurrence of systemic hemopathies reportedly “in remission”: six cases and review of literature. Medicina (Kaunas) 58:456

    Article  PubMed  Google Scholar 

  21. Evereklioglu C et al (2023) Comments on: Relapse of acute lymphoblastic leukemia presenting as masquerade uveitis with hypopyon in a child. Indian J Ophthalmol 71:2314–2315

    Article  PubMed  PubMed Central  Google Scholar 

  22. Rehman O et al (2022) Relapse of acute lymphoblastic leukemia presenting as masquerade uveitis with hypopyon in a child. Indian J Ophthalmol 70:2732–3

    Article  PubMed  PubMed Central  Google Scholar 

  23. Hung YC, Wilkinson B, Chong CF (2022) A case of aggressive natural killer cell leukemia. Digit J Ophthalmol 28:116–118

    Article  PubMed  PubMed Central  Google Scholar 

  24. Tyagi M et al (2017) Bilateral Hypopyon Uveitis in Chronic Myeloid Leukemia. Ocul Oncol Pathol 4:12–15

    Article  PubMed  PubMed Central  Google Scholar 

  25. Sudharshan S, Kumari A, Biswas J (2008) Bilateral hypopyon as the presenting feature of chronic myeloid leukemia. Ocular Immunol Inflamm 16:244–246

    Article  Google Scholar 

  26. Zamani G et al (2022) Intraocular lymphoma masquerading as unilateral hypopyon anterior uveitis: a case report. J Ophthalmic Inflamm Infect 12:25

    Article  PubMed  PubMed Central  Google Scholar 

  27. Li S, Young KH, Medeiros LJ (2018) Diffuse large B-cell lymphoma. Pathology 50:74–87

    Article  PubMed  Google Scholar 

  28. Mizener JB, Podhajsky P, Hayreh SS (1997) Ocular ischemic syndrome. Ophthalmology 104:859–864

    Article  CAS  PubMed  Google Scholar 

  29. Malhotra R, Gregory-Evans K (2000) Management of ocular ischaemic syndrome. Br J Ophthalmol 84:1428–1431

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gary C. Brown, S.S., Melissa M. Brown. Ocular Ischemic Syndrome. in Ryan's Retina, Vol. 6 (ed. Andrew P. Schachat, S.R.S., David R. Hinton, C.P. Wilkinson, Peter Wiedermann) 1227-1239 (Elsevier, 2018)

  31. Brown GC, Magargal LE (1988) The ocular ischemic syndrome. Clinical, fluorescein angiographic and carotid angiographic features. Int Ophthalmol 11:239–251

    Article  CAS  PubMed  Google Scholar 

  32. Brown GC, Brown MM, Magargal LE (1986) The ocular ischemic syndrome and neovascularization. Trans Pa Acad Ophthalmol Otolaryngol 38:302–306

    CAS  PubMed  Google Scholar 

  33. Casson RJ, Fleming FK, Shaikh A, James B (2001) Bilateral ocular ischemic syndrome secondary to giant cell arteritis. Arch Ophthalmol 119:306–307

    CAS  PubMed  Google Scholar 

  34. Papavasileiou E, Sobrin L, Papaliodis GN (2015) Ocular ischemic syndrome presenting as retinal vasculitis in a patient with moyamoya syndrome. Retinal Cases Brief Rep 9:170–172

    Article  Google Scholar 

  35. Wang YY et al (2020) Moyamoya disease associated with morning glory disc anomaly and other ophthalmic findings: a mini-review. Front Neurol 11:338

    Article  PubMed  PubMed Central  Google Scholar 

  36. Hwang JM et al (1999) Bilateral ocular ischemic syndrome secondary to giant cell arteritis progressing despite corticosteroid treatment. Am J Ophthalmol 127:102–104

    Article  CAS  PubMed  Google Scholar 

  37. Worrall M, Atebara N, Meredith T, Mann ES (2001) Bilateral ocular ischemic syndrome in Takayasu disease. Retina 21:75–76

    Article  CAS  PubMed  Google Scholar 

  38. Salinas-La Rosa CM (2017) Malignant Transformation of Optic Nerve Melanocytoma into Melanoma Associated with Ocular Ischemic Syndrome and Oculocardiac Reflex: Case Report and Review of the Literature. Seminars Ophthalmol 32:253–256

    Article  Google Scholar 

  39. Peter J et al (2011) Ocular manifestations of Takayasu arteritis: a cross-sectional study. Retina 31:1170–1178

    Article  PubMed  Google Scholar 

  40. Peter J, David S, Joseph G, Horo S, Danda D (2010) Hypoperfusive and hypertensive ocular manifestations in Takayasu arteritis. Clin Ophthalmol 4:1173–1176

    Article  PubMed  PubMed Central  Google Scholar 

  41. Larrazabal RB, Chiu HHC, Magbitang-Santiago AT (2020) Takayasu arteritis presenting as bilateral ocular ischemic syndrome. Vasc Special Int 36:163–169

    Article  Google Scholar 

  42. Koz OG et al (2007) Bilateral ocular ischemic syndrome as an initial manifestation of Takayasu’s arteritis associated with carotid steal syndrome. Rheumatol Int 27:299–302

    Article  PubMed  Google Scholar 

  43. Anguita R, Nazar C, Kobus R, Salinas A, Astete M (2019) Bilateral ocular ischemic syndrome as a manifestation of Takayasu arteritis in children. Can J Ophthalmol 54:e105–e108

    Article  PubMed  Google Scholar 

  44. Tang Y, Luo D, Peng W, Huang F, Peng Y (2010) Ocular ischemic syndrome secondary to carotid artery occlusion as a late complication of radiotherapy of nasopharyngeal carcinoma. J Neuro-ophthalmology 30:315–320

    Article  Google Scholar 

  45. Dhooge M, de Laey JJ (1989) The ocular ischemic syndrome. Bulletin de la Societe belge d’ophtalmologie 231:1–13

    CAS  PubMed  Google Scholar 

  46. Akinsoji E, Goldhardt R, Galor A (2018) A glimpse into uveitis in the aging eye: pathophysiology, clinical presentation and treatment considerations. Drugs Aging 35:399–408

    Article  PubMed  PubMed Central  Google Scholar 

  47. Gonder JR (1986) The ocular ischemic syndrome. Can J Ophthalmol 21:1–2

    CAS  PubMed  Google Scholar 

  48. Rothova A, Ooijman F, Kerkhoff F, Van Der Lelij A, Lokhorst HM (2001) Uveitis masquerade syndromes. Ophthalmology 108:386–399

    Article  CAS  PubMed  Google Scholar 

  49. Jacobs NA, Ridgway AE (1985) Syndrome of ischaemic ocular inflammation: six cases and a review. Br J Ophthalmol 69:681–687

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Karaconji T, Maconochie Z, McCluskey P (2013) Acute anterior uveitis in Sydney. Ocul Immunol Inflamm 21:108–114

    Article  CAS  PubMed  Google Scholar 

  51. Moradi A et al (2015) Clinical features and incidence rates of ocular complications in patients with ocular syphilis. Am J Ophthalmol 159:334–343 e331

    Article  PubMed  Google Scholar 

  52. Mathew RG, Goh BT, Westcott MC (2014) British Ocular Syphilis Study (BOSS): 2-year national surveillance study of intraocular inflammation secondary to ocular syphilis. Invest Ophthalmol Vis Sci 55:5394–5400

    Article  PubMed  Google Scholar 

  53. Pratas AC et al (2018) Increase in Ocular Syphilis Cases at Ophthalmologic Reference Center, France, 2012–2015. Emerg Infect Dis 24:193–200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoogewoud F et al (2017) Prognostic Factors in Syphilitic Uveitis. Ophthalmology 124:1808–1816

    Article  PubMed  Google Scholar 

  55. Berrocal AM, Davis JL (2002) Uveitis following intraocular surgery. Ophthalmol Clin North Am 15:357–364, vii

    Article  PubMed  Google Scholar 

  56. Nguyen JK, Fung AE, Flynn HW Jr, Scott IU (2006) Hypopyon and pseudoendophthalmitis associated with chronic vitreous hemorrhage. Ophthalmic Surgery Lasers Imaging 37:317–319

    Article  Google Scholar 

  57. Shunmugam M, Hugkulstone CE, Wong R, Williamson TH (2013) Consecutive toxic anterior segment syndrome in combined phaco-vitrectomy. Int Ophthalmol 33:289–290

    Article  PubMed  Google Scholar 

  58. Moisseiev E, Barak A (2012) Toxic anterior segment syndrome outbreak after vitrectomy and silicone oil injection. Eur J Ophthalmol 22:803–807

    Article  PubMed  Google Scholar 

  59. Sevimli N et al (2016) Toxic anterior segment syndrome following deep anterior lamellar keratoplasty. Arquivos Brasileiros de Oftalmologia 79:330–332

    Article  PubMed  Google Scholar 

  60. Sorkin N, Varssano D (2012) Toxic anterior segment syndrome following a triple descemet’s stripping automated endothelial keratoplasty procedure. Case Rep Ophthalmol 3:406–409

    Article  PubMed  PubMed Central  Google Scholar 

  61. Maier P, Birnbaum F, Bohringer D, Reinhard T (2008) Toxic anterior segment syndrome following penetrating keratoplasty. Arch Ophthalmol 126:1677–1681

    Article  PubMed  Google Scholar 

  62. Hernandez-Bogantes E et al (2019) Toxic anterior segment syndrome: a review. Surv Ophthalmol 64:463–476

    Article  PubMed  Google Scholar 

  63. Samson CM, Foster CS (2000) Chronic postoperative endophthalmitis. Int Ophthalmol Clin 40:57–67

    Article  CAS  PubMed  Google Scholar 

  64. Maalouf F, Abdulaal M, Hamam RN (2012) Chronic postoperative endophthalmitis: a review of clinical characteristics, microbiology, treatment strategies, and outcomes. Int J Inflamm 2012:313248

    Google Scholar 

  65. Nche EN, Amer R (2020) Lens-induced uveitis: an update. Graefes Arch Clin Exp Ophthalmol 258:1359–1365

    Article  PubMed  PubMed Central  Google Scholar 

  66. Rosenbaum JT, Samples JR, Seymour B, Langlois L, David L (1987) Chemotactic activity of lens proteins and the pathogenesis of phacolytic glaucoma. Arch Ophthalmol 105:1582–1584

    Article  CAS  PubMed  Google Scholar 

  67. Chandler PA (1958) Problems in the diagnosis and treatment of lens-induced uveitis and glaucoma. AMA Arch Ophthalmol 60:828–841

    Article  CAS  PubMed  Google Scholar 

  68. Marak GE Jr (1992) Phacoanaphylactic endophthalmitis. Surv Ophthalmol 36:325–339

    Article  PubMed  Google Scholar 

  69. Meisler DM, Mandelbaum S (1989) Propionibacterium-associated endophthalmitis after extracapsular cataract extraction. Review of reported cases. Ophthalmology 96:54–61

    Article  CAS  PubMed  Google Scholar 

  70. Oshika T, Eguchi S, Goto H, Ohashi Y (2017) Outbreak of subacute-onset toxic anterior segment syndrome associated with single-piece acrylic intraocular lenses. Ophthalmology 124:519–523

    Article  PubMed  Google Scholar 

  71. Mamalis N et al (2006) Toxic anterior segment syndrome. J Cataract Refractive Surg 32:324–333

    Article  Google Scholar 

  72. Bodnar Z, Clouser S, Mamalis N (2012) Toxic anterior segment syndrome: update on the most common causes. J Cataract Refractive Surg 38:1902–1910

    Article  Google Scholar 

  73. Ophthalmology, A.A.o. Differential Diagnosis. (2020)

  74. Dobrin RS, Vernier RL, Fish AL (1975) Acute eosinophilic interstitial nephritis and renal failure with bone marrow-lymph node granulomas and anterior uveitis. A new syndrome. Am J Med 59:325–333

    Article  CAS  PubMed  Google Scholar 

  75. Regusci A et al (2022) Tubulointerstitial nephritis and uveitis syndrome: a systematic review. Nephrol Dial Transplant 37:876–886

    Article  PubMed  Google Scholar 

  76. Mandeville JT, Levinson RD, Holland GN (2001) The tubulointerstitial nephritis and uveitis syndrome. Surv Ophthalmol 46:195–208

    Article  CAS  PubMed  Google Scholar 

  77. Okafor LO, Hewins P, Murray PI, Denniston AK (2017) Tubulointerstitial nephritis and uveitis (TINU) syndrome: a systematic review of its epidemiology, demographics and risk factors. Orphanet J Rare Dis 12:128

    Article  PubMed  PubMed Central  Google Scholar 

  78. Aguilar MC, Lonngi M, de-la-Torre A (2016) Tubulointerstitial nephritis and uveitis syndrome: case report and review of the literature. Ocul Immunol Inflamm 24:415–421

    Article  PubMed  Google Scholar 

  79. Caplash S et al (2018) Treatment challenges in an atypical presentation of tubulointerstitial nephritis and uveitis (TINU). Am J Ophthalmol Case Rep 10:253–256

    Article  PubMed  PubMed Central  Google Scholar 

  80. Kong CL, B.B., Loewen R, Conner E, Zambrano EV, Khatib L, Errera MH (2022) Case report: severe atypical presentation of uveitis associated with tubulointerstitial nephritis. J Ophthalmol Res

  81. Standardization of Uveitis Nomenclature Working, G (2021) Classification criteria for tubulointerstitial nephritis with uveitis syndrome. Am J Ophthalmol 228:255–261

    Article  Google Scholar 

  82. Mackensen F, Smith JR, Rosenbaum JT (2007) Enhanced recognition, treatment, and prognosis of tubulointerstitial nephritis and uveitis syndrome. Ophthalmology 114:995–999

    Article  PubMed  Google Scholar 

  83. Mackensen F, Billing H (2009) Tubulointerstitial nephritis and uveitis syndrome. Curr Opin Ophthalmol 20:525–531

    Article  PubMed  Google Scholar 

  84. Levinson RD (2008) Tubulointerstitial nephritis and uveitis syndrome. Int Ophthalmol Clin 48:51–59

    Article  PubMed  Google Scholar 

  85. Pakzad-Vaezi K, Pepple KL (2017) Tubulointerstitial nephritis and uveitis. Curr Opin Ophthalmol 28:629–635

    Article  PubMed  PubMed Central  Google Scholar 

  86. Jabs DA et al (2007) Longitudinal study of the ocular complications of AIDS: 2. Ocular examination results at enrollment. Ophthalmology 114:787–793

    Article  PubMed  Google Scholar 

  87. Chan NS, Chee SP, Caspers L, Bodaghi B (2018) Clinical Features of CMV-Associated Anterior Uveitis. Ocul Immunol Inflamm 26:107–115

    Article  CAS  PubMed  Google Scholar 

  88. Chee SP et al (2008) Clinical features of cytomegalovirus anterior uveitis in immunocompetent patients. Am J Ophthalmol 145:834–840

    Article  PubMed  Google Scholar 

  89. Chee SP, Jap A (2008) Presumed fuchs heterochromic iridocyclitis and Posner-Schlossman syndrome: comparison of cytomegalovirus-positive and negative eyes. Am J Ophthalmol 146:883–889 e881

    Article  PubMed  Google Scholar 

  90. Koizumi N et al (2015) Clinical features and management of cytomegalovirus corneal endotheliitis: analysis of 106 cases from the Japan corneal endotheliitis study. Br J Ophthalmol 99:54–58

    Article  PubMed  Google Scholar 

  91. Touhami S et al (2019) Predictive factors of intraocular pressure level evolution over time and glaucoma severity in fuchs’ heterochromic iridocyclitis. Invest Ophthalmol Vis Sci 60:2399–2405

    Article  CAS  PubMed  Google Scholar 

  92. de Groot-Mijnes JD et al (2006) Rubella virus is associated with fuchs heterochromic iridocyclitis. Am J Ophthalmol 141:212–214

    Article  PubMed  Google Scholar 

  93. Quentin CD, Reiber H (2004) Fuchs heterochromic cyclitis: rubella virus antibodies and genome in aqueous humor. Am J Ophthalmol 138:46–54

    Article  PubMed  Google Scholar 

  94. Leleu I et al (2021) Clinical features and diagnosis of anterior segment inflammation related to cytomegalovirus in immunocompetent African, Asian, and caucasian patients. Ocul Immunol Inflamm 29:160–168

    Article  CAS  PubMed  Google Scholar 

  95. van Boxtel LA, van der Lelij A, van der Meer J, Los LI (2007) Cytomegalovirus as a cause of anterior uveitis in immunocompetent patients. Ophthalmology 114:1358–1362

    Article  PubMed  Google Scholar 

  96. Sakai JI, Usui Y, Suzuki J, Kezuka T, Goto H (2019) Clinical features of anterior uveitis caused by three different herpes viruses. Int Ophthalmol 39:2785–2795

    Article  PubMed  Google Scholar 

  97. Terada Y et al (2021) Distinguishing features of anterior uveitis caused by herpes simplex virus, varicella-zoster virus, and cytomegalovirus. Am J Ophthalmol 227:191–200

    Article  CAS  PubMed  Google Scholar 

  98. Takase H et al (2014) Comparison of the ocular characteristics of anterior uveitis caused by herpes simplex virus, varicella-zoster virus, and cytomegalovirus. Jpn J Ophthalmol 58:473–482

    Article  CAS  PubMed  Google Scholar 

  99. Bodaghi B et al (2001) Chronic severe uveitis: etiology and visual outcome in 927 patients from a single center. Medicine (Baltimore) 80:263–270

    Article  CAS  PubMed  Google Scholar 

  100. Bajwa A et al (2015) Epidemiology of uveitis in the mid-Atlantic United States. Clin Ophthalmol 9:889–901

    Article  PubMed  PubMed Central  Google Scholar 

  101. D’Ambrosio EM et al (2017) Clinical features and complications of the HLA-B27-associated acute anterior uveitis: a metanalysis. Seminars Ophthalmol 32:689–701

    Article  Google Scholar 

  102. Wakefield D, Yates W, Amjadi S, McCluskey P (2016) HLA-B27 Anterior Uveitis: Immunology and Immunopathology. Ocul Immunol Inflamm 24:450–459

    Article  CAS  PubMed  Google Scholar 

  103. Wakefield D, Montanaro A, McCluskey P (1991) Acute anterior uveitis and HLA-B27. Surv Ophthalmol 36:223–232

    Article  CAS  PubMed  Google Scholar 

  104. Pathanapitoon K, Dodds EM, Cunningham ET Jr, Rothova A (2017) Clinical Spectrum of HLA-B27-associated Ocular Inflammation. Ocul Immunol Inflamm 25:569–576

    Article  CAS  PubMed  Google Scholar 

  105. Yoshida A, Obata H, Kawashima H (2015) Cytomegalovirus uveitis with hypopyon mimicking bacterial endophthalmitis. Case Rep Ophthalmological Med 2015:489813

    Google Scholar 

  106. Ganapathy PS, Lowder CY, Srivastava SK (2018) Aggressive initial presentation of HLA-B27 uveitis in older individuals: a case series. Ocul Immunol Inflamm 26:425–427

    Article  PubMed  Google Scholar 

  107. Toh ZH, Agrawal R (2020) Diagnostic dilemma: unilateral panuveitis mimicking endophthalmitis in very severe HLA B27-associated uveitis. Am J Ophthalmol Case Rep 17:100589

    Article  PubMed  PubMed Central  Google Scholar 

  108. Garg P, Roy A, Sharma S (2017) Endophthalmitis after cataract surgery: epidemiology, risk factors, and evidence on protection. Curr Opin Ophthalmol 28:67–72

    Article  PubMed  Google Scholar 

  109. Durand ML (2017) Bacterial and fungal endophthalmitis. Clin Microbiol Rev 30:597–613

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Okada AA, Johnson RP, Liles WC, D’Amico DJ, Baker AS (1994) Endogenous bacterial endophthalmitis. Report of a ten-year retrospective study. Ophthalmology 101:832–838

    Article  CAS  PubMed  Google Scholar 

  111. Jenkins TL et al (2020) Endogenous bacterial endophthalmitis: a five-year retrospective review at a tertiary care academic center. Ocul Immunol Inflamm 28:975–983

    Article  PubMed  Google Scholar 

  112. Connell PP et al (2011) Endogenous endophthalmitis: 10-year experience at a tertiary referral centre. Eye (Lond) 25:66–72

    Article  CAS  PubMed  Google Scholar 

  113. Cho H et al (2018) Endogenous endophthalmitis in the American and Korean population: an 8-year retrospective study. Ocul Immunol Inflamm 26:496–503

    PubMed  Google Scholar 

  114. Jackson TL, Paraskevopoulos T, Georgalas I (2014) Systematic review of 342 cases of endogenous bacterial endophthalmitis. Surv Ophthalmol 59:627–635

    Article  PubMed  Google Scholar 

  115. Chersich MF et al (2018) Diagnosis and treatment of listeria monocytogenes endophthalmitis: a systematic review. Ocul Immunol Inflamm 26:508–517

    PubMed  Google Scholar 

  116. Dadia SD et al (2018) Salmonella typhi associated endogenous endophthalmitis: a case report and a review of literature. Ocul Immunol Inflamm 26:527–532

    PubMed  Google Scholar 

  117. Silpa-Archa S, Ponwong A, Preble JM, Foster CS (2018) Culture-positive endogenous endophthalmitis: an eleven-year retrospective study in the Central Region of Thailand. Ocul Immunol Inflamm 26:533–542

    PubMed  Google Scholar 

  118. Kresloff MS, Castellarin AA, Zarbin MA (1998) Endophthalmitis. Surv Ophthalmol 43:193–224

    Article  CAS  PubMed  Google Scholar 

  119. Results of the Endophthalmitis Vitrectomy Study (1995) A randomized trial of immediate vitrectomy and of intravenous antibiotics for the treatment of postoperative bacterial endophthalmitis. Endophthalmitis vitrectomy study group. Arch Ophthalmol 113:1479–1496

    Article  Google Scholar 

  120. Gokce G, Sobaci G, Ozgonul C (2015) Post-traumatic endophthalmitis: a mini-review. Sem Ophthalmol 30:470–474

    Article  Google Scholar 

  121. Saliba N, Owen ME, Beare N (2010) Tattoo-associated uveitis. Eye (Lond) 24:1406

    Article  CAS  PubMed  Google Scholar 

  122. Reddy AK, Shildkrot Y, Newman SA, Thorne JE (2015) T-Lymphocyte predominance and cellular atypia in tattoo-associated uveitis. JAMA Ophthalmol 133:1356–1357

    Article  PubMed  Google Scholar 

  123. Pandya VB, Hooper CY, Essex RW, Cook M (2014) Tattoo-associated uveitis. Am J Ophthalmol 158:1355–1356

    Article  PubMed  Google Scholar 

  124. Ostheimer TA et al (2014) Tattoo-associated uveitis. Am J Ophthalmol 158:637–643 e631

    Article  PubMed  Google Scholar 

  125. Mansour AM, Chan CC (2015) Tattoo-associated uveitis. Am J Ophthalmol 159:408–409

    Article  PubMed  Google Scholar 

  126. Kluger N (2018) Tattoo-associated uveitis with or without systemic sarcoidosis: a comparative review of the literature. J Eur Acad Dermatol Venereol 32:1852–1861

    Article  CAS  PubMed  Google Scholar 

  127. Adam MK, Rahimy E, Dunn JP (2015) Tattoo-associated uveitis. JAMA Ophthalmol 133:e144543

    Article  PubMed  Google Scholar 

  128. Pandya VB, Hooper CY, Merani R, McCluskey P (2017) Tattoo-associated uveitis with choroidal granuloma: a rare presentation of systemic sarcoidosis. Retinal Cases Brief Rep 11:272–276

    Article  Google Scholar 

  129. Moorthy RS, London NJ, Garg SJ, Cunningham ET Jr (2013) Drug-induced uveitis. Curr Opin Ophthalmol 24:589–597

    Article  PubMed  Google Scholar 

  130. Campochiaro PA et al (2011) Sustained benefits from ranibizumab for macular edema following central retinal vein occlusion: twelve-month outcomes of a phase III study. Ophthalmology 118:2041–2049

    Article  PubMed  Google Scholar 

  131. Brown DM et al (2009) Ranibizumab versus verteporfin photodynamic therapy for neovascular age-related macular degeneration: Two-year results of the ANCHOR study. Ophthalmology 116:57–65 e55

    Article  PubMed  Google Scholar 

  132. Comparison of Age-related Macular Degeneration Treatments Trials Research, G et al (2020) Ranibizumab and Bevacizumab for Treatment of Neovascular Age-related Macular Degeneration. Ophthalmology 127:S135–S145

    Article  Google Scholar 

  133. Regillo CD et al (2008) Randomized, double-masked, sham-controlled trial of ranibizumab for neovascular age-related macular degeneration: PIER Study year 1. Am J Ophthalmol 145:239–248

    Article  CAS  PubMed  Google Scholar 

  134. Brown DM et al (2011) Sustained benefits from ranibizumab for macular edema following branch retinal vein occlusion: 12-month outcomes of a phase III study. Ophthalmology 118:1594–1602

    Article  PubMed  Google Scholar 

  135. Wickremasinghe SS et al (2008) Acute intraocular inflammation after intravitreous injections of bevacizumab for treatment of neovascular age-related macular degeneration. Ophthalmology 115:1911–1915

    Article  PubMed  Google Scholar 

  136. Kuo BL, Singh RP (2022) Brolucizumab for the treatment of diabetic macular edema. Curr Opin Ophthalmol 33:167–173

    Article  PubMed  Google Scholar 

  137. Brown DM et al (2022) KESTREL and KITE: 52-Week Results From Two Phase III Pivotal Trials of Brolucizumab for Diabetic Macular Edema. Am J Ophthalmol 238:157–172

    Article  CAS  PubMed  Google Scholar 

  138. Biarnes M et al (2023) Methodological appraisal of phase 3 clinical trials in geographic atrophy. Biomedicines 11:1548

    Article  PubMed  PubMed Central  Google Scholar 

  139. Beovu® [US prescribing information]. (ed. Novartis) ( East Hanover, NJ (2019))

  140. Singer M et al (2022) Clinical characteristics and outcomes of eyes with intraocular inflammation after brolucizumab: post hoc analysis of HAWK and HARRIER. Ophthalmol Retina 6:97–108

    Article  PubMed  Google Scholar 

  141. Iyer PG et al (2020) Brolucizumab-related retinal vasculitis with exacerbation following ranibizumab retreatment: a clinicopathologic case study. Am J Ophthalmol Case Rep 20:100989

    Article  PubMed  PubMed Central  Google Scholar 

  142. Dugel PU et al (2020) HAWK and HARRIER: phase 3, multicenter, randomized, double-masked trials of brolucizumab for neovascular age-related macular degeneration. Ophthalmology 127:72–84

    Article  PubMed  Google Scholar 

  143. Anderson WJ et al (2021) Mechanisms of sterile inflammation after intravitreal injection of antiangiogenic drugs: a narrative review. Int J Retina Vitreous 7:37

    Article  PubMed  PubMed Central  Google Scholar 

  144. Fechtner RD et al (1998) Anterior uveitis associated with latanoprost. Am J Ophthalmol 126:37–41

    Article  CAS  PubMed  Google Scholar 

  145. Li D, Yang L, Bai F, Zeng S, Liu X (2022) Clinical manifestations, diagnosis, treatment and prognosis of uveitis induced by anticancer drugs: a review of literature. Brain Sci 12

  146. Dimitriou F et al (2021) The association between immune checkpoint or BRAF/MEK inhibitor therapy and uveitis in patients with advanced cutaneous melanoma. Eur J Cancer 144:215–223

    Article  CAS  PubMed  Google Scholar 

  147. Choe CH, McArthur GA, Caro I, Kempen JH, Amaravadi RK (2014) Ocular toxicity in BRAF mutant cutaneous melanoma patients treated with vemurafenib. Am J Ophthalmol 158:831–837 e832

    Article  CAS  PubMed  Google Scholar 

  148. Klein KA, Azzoli CG, Rifkin LM (2017) Bilateral acute simultaneous onset anterior uveitis presumed secondary to erlotinib: a report of two cases. Am J Ophthalmol Case Rep 6:21–23

    Article  PubMed  Google Scholar 

  149. Chan S, Ahern E, Chaudhry S, Hughes B (2019) Bilateral acute anterior uveitis: a rare ocular side effect of erlotinib. BMJ Case Rep 12

  150. Kiratli H, Mocan MC, Irkec M (2016) In vivo confocal microscopy in differentiating ipilimumab-induced anterior uveitis from metastatic uveal melanoma. Case Rep Ophthalmol 7:126–131

    Article  PubMed  PubMed Central  Google Scholar 

  151. Sun Mm Md P et al (2020) Uveitis in patients treated with CTLA-4 and PD-1 checkpoint blockade inhibition. Ocul Immunol Inflamm 28:217–227

    Article  Google Scholar 

  152. Papavasileiou E, Prasad S, Freitag SK, Sobrin L, Lobo AM (2016) Ipilimumab-induced ocular and orbital inflammation–a case series and review of the literature. Ocul Immunol Inflamm 24:140–146

    CAS  PubMed  Google Scholar 

  153. Venkat AG et al (2020) Local therapy for cancer therapy-associated uveitis: a case series and review of the literature. Br J Ophthalmol 104:703–711

    Article  PubMed  Google Scholar 

  154. Wefers Bettink-Remeijer M et al (2009) Uveitis-like syndrome and iris transillumination after the use of oral moxifloxacin. Eye (Lond) 23:2260–2262

    Article  CAS  PubMed  Google Scholar 

  155. Willermain F et al (2010) Uveitis-like syndrome and iris transillumination after the use of oral moxifloxacin. Eye (Lond) 24:1419 author reply 1419-1420

    Article  CAS  PubMed  Google Scholar 

  156. Kuper JI, D’Aprile M (2000) Drug-Drug interactions of clinical significance in the treatment of patients with Mycobacterium avium complex disease. Clin Pharmacokinet 39:203–214

    Article  CAS  PubMed  Google Scholar 

  157. Skinner MH, Blaschke TF (1995) Clinical pharmacokinetics of rifabutin. Clin Pharmacokinet 28:115–125

    Article  CAS  PubMed  Google Scholar 

  158. Shafran SD et al (1998) Determinants of rifabutin-associated uveitis in patients treated with rifabutin, clarithromycin, and ethambutol for Mycobacterium avium complex bacteremia: a multivariate analysis. Canadian HIV Trials Network Protocol 010 Study Group. J Infect Dis 177:252–255

    Article  CAS  PubMed  Google Scholar 

  159. Lim LL, Fraunfelder FW, Rosenbaum JT (2007) Do tumor necrosis factor inhibitors cause uveitis? A registry-based study. Arthritis Rheum 56:3248–3252

    Article  CAS  PubMed  Google Scholar 

  160. Bostanci Ceran B, Ozates S (2020) Ocular manifestations of coronavirus disease 2019. Graefes Arch Clin Exp Ophthalmol 258:1959–1963

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. United States Department of Health and Human Services (DHHS) (Vaccine Adverse Event Reporting System). (2022)

  162. Testi I, Brandao-de-Resende C, Agrawal R, Pavesio C, Group, C.-V.O.I.E.S. (2022) Ocular inflammatory events following COVID-19 vaccination: a multinational case series. J Ophthalmic Inflamm Infect 12:4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Haseeb AA, Solyman O, Abushanab MM, Abo Obaia AS, Elhusseiny AM (2022) Ocular complications following vaccination for COVID-19: a one-year retrospective. Vaccines (Basel) 10

  164. Bolletta E et al (2021) Uveitis and other ocular complications following COVID-19 vaccination. J Clin Med 10

  165. Murgova S, Balchev G (2022) Ophthalmic manifestation after SARS-CoV-2 vaccination: a case series. J Ophthalmic Inflamm Infect 12:20

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Ortiz-Egea JM, Sanchez CG, Lopez-Jimenez A, Navarro OD (2022) Herpetic anterior uveitis following Pfizer-BioNTech coronavirus disease 2019 vaccine: two case reports. J Med Case Rep 16:127

    Article  PubMed  PubMed Central  Google Scholar 

  167. Arora A et al (2022) Recurrence of tubercular choroiditis following anti-SARS-CoV-2 vaccination. Eur J Ophthalmol 11206721221088439

  168. Goyal M, Murthy SI, Annum S (2021) Bilateral Multifocal Choroiditis following COVID-19 Vaccination. Ocul Immunol Inflamm 29:753–757

    Article  CAS  PubMed  Google Scholar 

  169. Mishra SB, Mahendradas P, Kawali A, Sanjay S, Shetty R (2021) Reactivation of varicella zoster infection presenting as acute retinal necrosis post COVID 19 vaccination in an Asian Indian male. Eur J Ophthalmol 11206721211046485

  170. Rabinovitch T et al (2021) UVEITIS AFTER THE BNT162b2 mRNA VACCINATION AGAINST SARS-CoV-2 INFECTION: A Possible Association. Retina 41:2462–2471

    Article  CAS  PubMed  Google Scholar 

  171. Herbort CP Jr, Papasavvas I (2021) Effect of SARS-CoV-2 mRNA vaccination on ocular herpes simplex and varicella-zoster virus reactivation: should preventive antiviral treatment be given in known herpes patients. J Ophthalmic Inflamm Infect 11:33

    Article  PubMed  PubMed Central  Google Scholar 

  172. Balidis M et al (2021) Acute corneal graft rejection after anti-severe acute respiratory syndrome-coronavirus-2 vaccination: a report of four cases. Eur J Ophthalmol 11206721211064033

  173. Parmar DP, Garde PV, Shah SM, Bhole PK (2021) Acute graft rejection in a high-risk corneal transplant following COVID-19 vaccination: a case report. Indian J Ophthalmol 69:3757–3758

    Article  PubMed  PubMed Central  Google Scholar 

  174. Crnej A, Khoueir Z, Cherfan G, Saad A (2021) Acute corneal endothelial graft rejection following COVID-19 vaccination. J Fr Ophtalmol 44:e445–e447

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Rallis KI, Ting DSJ, Said DG, Dua HS (2022) Corneal graft rejection following COVID-19 vaccine. Eye (Lond) 36:1319–1320

    Article  CAS  PubMed  Google Scholar 

  176. Wasser LM, Roditi E, Zadok D, Berkowitz L, Weill Y (2021) Keratoplasty rejection after the BNT162b2 messenger RNA vaccine. Cornea 40:1070–1072

    Article  PubMed  PubMed Central  Google Scholar 

  177. Singh S et al (2022) SARS-CoV-2 and its beta variant of concern infect human conjunctival epithelial cells and induce differential antiviral innate immune response. Ocul Surf 23:184–194

    Article  PubMed  Google Scholar 

  178. Phylactou M, Li JO, Larkin DFP (2021) Characteristics of endothelial corneal transplant rejection following immunisation with SARS-CoV-2 messenger RNA vaccine. Br J Ophthalmol 105:893–896

    Article  PubMed  Google Scholar 

  179. Fujio K et al (2022) Characteristics and clinical ocular manifestations in patients with acute corneal graft rejection after receiving the COVID-19 vaccine: a systematic review. J Clin Med 11

  180. Sainz de la Maza M, Foster CS, Jabbur NS, Baltatzis S (2002) Ocular characteristics and disease associations in scleritis-associated peripheral keratopathy. Arch Ophthalmol 120:15–19

    Article  PubMed  Google Scholar 

  181. Sainz de la Maza M, Jabbur NS, Foster CS (1994) Severity of scleritis and episcleritis. Ophthalmology 101:389–396

    Article  CAS  PubMed  Google Scholar 

  182. Galor A, Thorne JE (2007) Scleritis and peripheral ulcerative keratitis. Rheumatic Dis Clin North Am 33:835–854, vii

    Article  Google Scholar 

  183. Fraunfelder FW, Fraunfelder FT (2004) Adverse ocular drug reactions recently identified by the National Registry of Drug-Induced Ocular Side Effects. Ophthalmology 111:1275–1279

    Article  CAS  PubMed  Google Scholar 

  184. Wolpert LE, Watts AR (2021) Zoledronate-induced anterior uveitis, scleritis and optic neuritis: a case report. N Z Med J 134:91–94

    PubMed  Google Scholar 

  185. Moore MM, Beith JM (2008) Acute unilateral anterior uveitis and scleritis following a single infusion of zoledronate for metastatic breast cancer. Med J Aust 188:370–371

    Article  PubMed  Google Scholar 

  186. Tabbara KF (2008) Nodular scleritis following alendronate therapy. Ocul Immunol Inflamm 16:99–101

    Article  PubMed  Google Scholar 

  187. Leung S, Ashar BH, Miller RG (2005) Bisphosphonate-associated scleritis: a case report and review. South Med J 98:733–735

    Article  PubMed  Google Scholar 

  188. Provost C, Sene T, Lecler A (2021) Bisphosphonate-induced posterior scleritis. Ophthalmology 128:371

    Article  PubMed  Google Scholar 

  189. Kafkala C, Daoud YJ, Paredes I, Foster CS (2005) Masquerade scleritis. Ocul Immunol Inflamm 13:479–482

    Article  PubMed  Google Scholar 

  190. Sharma M et al (2017) Invasive Ocular Surface Squamous Neoplasia Masquerading as Nodular Scleritis. Ophthalmic Plast Reconstr Surg 33:e45–e47

    Article  PubMed  Google Scholar 

  191. Putri CA, Salvi SM (2020) Necrotic choroidal melanoma masquerading as scleritis. Indian J Ophthalmol 68:1979–1981

    Article  PubMed  PubMed Central  Google Scholar 

  192. Knowles DM, Jakobiec FA, McNally L, Burke JS (1990) Lymphoid hyperplasia and malignant lymphoma occurring in the ocular adnexa (orbit, conjunctiva, and eyelids): a prospective multiparametric analysis of 108 cases during 1977 to 1987. Hum Pathol 21:959–973

    Article  CAS  PubMed  Google Scholar 

  193. Pattnaik M, Tripathy D, Mittal R (2021) Subconjunctival dirofilariasis mimicking infectious scleritis. J Pediatr Ophthalmol Strabismus 58:136

    Article  PubMed  Google Scholar 

  194. Hasanreisoglu M et al (2021) Case report: a human case of onchocerca lupi mimicking nodular scleritis. Am J Trop Med Hyg 105:1782–1785

    Article  PubMed  PubMed Central  Google Scholar 

  195. Chen JL, Men M, Naini BV, Tsui E (2022) IgG4-related hypertensive granulomatous anterior uveitis. Am J Ophthalmol Case Rep 26:101465

    Article  PubMed  PubMed Central  Google Scholar 

  196. Philippakis E et al (2015) IgG4-related disease masquerading as recurrent scleritis and chronic conjunctivitis. Ocul Immunol Inflamm 23:168–172

    Article  CAS  PubMed  Google Scholar 

  197. Mase Y et al (2022) Posterior scleritis with choroidal detachments and periaortitis associated with IgG4-related disease: a case report. Medicine (Baltimore) 101:e29611

    Article  PubMed  Google Scholar 

  198. Kim SJ et al (2021) IgG4-related disease presenting as recurrent scleritis combined with optic neuropathy. BMC Ophthalmol 21:5

    Article  PubMed  PubMed Central  Google Scholar 

  199. Karim F et al (2017) IgG4-related disease as an emerging cause of scleritis. Acta Ophthalmol 95:e795–e796

    Article  PubMed  Google Scholar 

  200. Herwig-Carl MC, Fischer HP, Holz FG, Loeffler KU (2022) Scleritis in IgG4-related disease. Ophthalmologie 119:767–768

    Article  PubMed  Google Scholar 

  201. Berkowitz E, Arnon E, Yaakobi A, Cohen Y, Tiosano B (2017) IgG4-Related Disease Presenting as Isolated Scleritis. Case Rep Ophthalmological Med 2017:4876587

    Google Scholar 

  202. Maritati F, Peyronel F, Vaglio A (2020) IgG4-related disease: a clinical perspective. Rheumatology (Oxford) 59:iii123–iii131

    Article  CAS  PubMed  Google Scholar 

  203. Khoo LW, Srinivasan S, Roberts F (2020) Foreign body episcleral suture granulomas mimicking nodular anterior scleritis. BMJ Case Rep 13

  204. Srinivasan S, Singh AK, Desai SP, Talbot JF, Parsons MA (2003) Foreign body episcleral granulomas complicating intravitreal silicone oil tamponade: a clinicopathological study. Ophthalmology 110:1837–1840

    Article  PubMed  Google Scholar 

  205. Yangzes S, Sharma VK, Singh SR, Ram J (2019) Scleromalacia perforans in rheumatoid arthritis. QJM 112:459–460

    Article  CAS  PubMed  Google Scholar 

  206. Sakellariou G, Berberidis C, Vounotrypidis P (2005) A case of Behcet’s disease with scleromalacia perforans. Rheumatology (Oxford) 44:258–260

    Article  CAS  PubMed  Google Scholar 

  207. Chan AY, Liu DT (2005) A case of Behcet’s disease with scleromalacia perforans. Rheumatology 44:1077

    Article  CAS  PubMed  Google Scholar 

  208. Tesar PJ, Burgess JA, Goy JA, Lazell RW (1981) Scleromalacia perforans in ulcerative colitis. Gastroenterology 81:153–155

    Article  CAS  PubMed  Google Scholar 

  209. Hon C, Law RW, Au WY (2005) Scleromalacia perforans complicating graft-versus-host disease. Br J Haematol 129:1

    Article  PubMed  Google Scholar 

  210. Kachmaryk M, Bouchard CS, Duffner LA (1996) Bilateral fascia lata patch grafts in a patient with progressive scleromalacia perforans. Ophthalmic Surg Lasers 27:397–400

    Article  CAS  PubMed  Google Scholar 

  211. Droutsas K et al (2021) Treatment of progressive scleromalacia perforans by tectonic enhancement with lyophilized equine pericardium. Cornea 40:648–651

    Article  PubMed  Google Scholar 

  212. Riono WP, Hidayat AA, Rao NA (1999) Scleritis: a clinicopathologic study of 55 cases. Ophthalmology 106:1328–1333

    Article  CAS  PubMed  Google Scholar 

  213. Doshi RR, Harocopos GJ, Schwab IR, Cunningham ET Jr (2013) The spectrum of postoperative scleral necrosis. Surv Ophthalmol 58:620–633

    Article  PubMed  Google Scholar 

  214. Salamon SM, Mondino BJ, Zaidman GW (1982) Peripheral corneal ulcers, conjunctival ulcers, and scleritis after cataract surgery. Am J Ophthalmol 93:334–337

    Article  CAS  PubMed  Google Scholar 

  215. Alsagoff Z, Tan DT, Chee SP (2000) Necrotising scleritis after bare sclera excision of pterygium. Br J Ophthalmol 84:1050–1052

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Vagefi MR, Hollander DA, Seitzman GD, Margolis TP (2005) Bilateral surgically induced necrotising scleritis with secondary superinfection. Br J Ophthalmol 89:124–125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Lyne AJ, Lloyd-Jones D (1979) Necrotizing scleritis after ocular surgery. Trans Ophthalmol Soc U K 1962(99):146–149

    Google Scholar 

Download references

Acknowledgements

None.

Funding

This work was supported by grants P30 EY08098 and UL1 TR001857 (NIH, Bethesda, MD) and an unrestricted grant from Research to Prevent Blindness (New York, NY). The sponsor or funding organizations had no role in the design or conduct of this research. The authors have no financial disclosures to declare.

Author information

Authors and Affiliations

Authors

Contributions

SC: Conceptualization, Investigation, Writing- Original Draft, Writing-Review and editing. MPE: Visualization, Writing- Review and editing. MW: Writing- Review and Editing Kunal K. Dansingani: CI: Visualization. NK: Writing- Review and Editing. EF: Supervision, Writing- Review and Editing. JAS: Supervision, Writing- Review and Editing. BB Supervision, Writing- Review and Editing. VJ: Conceptualization, Supervision, Writing- Review and Editing. MHE: Conceptualization, Supervision, Writing- Review and Editing, Project Administration. All authors reviewed the manuscript.

Corresponding author

Correspondence to Marie-Helene Errera.

Ethics declarations

Ethics approval and consent to participate

Consent was obtained for all figures presented in this manuscript.

Consent for publication

Not applicable.

Competing interests

No authors have any competing interests to declare.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caplash, S., Paez-Escamilla, M., Westcott, M. et al. Mimickers of anterior uveitis, scleritis and misdiagnoses- tips and tricks for the cornea specialist. J Ophthal Inflamm Infect 14, 14 (2024). https://doi.org/10.1186/s12348-024-00396-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12348-024-00396-z

Keywords