Next Article in Journal
Effects of High-Biotin Sample Interference on Antibody Concentrations in Sandwich Immunoassays
Next Article in Special Issue
Regional Differences in Uptake of Vaccination against COVID-19 and Influenza in Germany: Results from the DigiHero Cohort
Previous Article in Journal
Comparison of Post-Vaccination Response between mRNA and Vector Vaccines against SARS-CoV-2 in Terms of Humoral Response after Six Months of Observation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ocular Inflammation Post-Vaccination

Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
*
Author to whom correspondence should be addressed.
Vaccines 2023, 11(10), 1626; https://doi.org/10.3390/vaccines11101626
Submission received: 29 September 2023 / Revised: 19 October 2023 / Accepted: 20 October 2023 / Published: 23 October 2023
(This article belongs to the Special Issue Vaccination Coverage and Vaccine Hesitancy)

Abstract

:
The association between vaccines and ocular disorders has attracted significant attention in scientific research. Numerous mainstream vaccines are associated with a range of uveitis types, including anterior, intermediate, and posterior uveitis. Additionally, they are associated with distinct ocular diseases such as multifocal choroiditis, Vogt–Koyanagi–Harada (VKH) disease, acute posterior multifocal placoid pigment epitheliopathy (APMPPE), and multiple evanescent white dot syndrome (MEWDS). These ocular conditions are often transient, with a vast majority of patients experiencing improvement after steroid intervention. To date, numerous cases of vaccine-induced uveitis have been reported. This study analyzed the correlation between antiviral vaccines, including the hepatitis B virus (HBV), human papillomavirus (HPV), measles–mumps–rubella (MMR), varicella zoster virus (VZV), and influenza vaccines, and different manifestations of uveitis. This is the first comprehensive study to offer a detailed analysis of uveitis types induced by antiviral vaccines. Through an extensive database search, we found a particularly strong link between influenza vaccines, followed by VZV and HPV vaccines. While anterior uveitis is common, conditions such as APMPPE, MEWDS, and VKH are particularly notable and merit careful consideration in clinical practice. Corticosteroid treatment was effective; however, half of the observed patients did not achieve full recovery, indicating potentially prolonged effects of the vaccine.

1. Introduction

Vaccines play a pivotal role in the prevention and control of infectious diseases and significantly affect global health [1]. By preventing the spread of infectious diseases and reducing the burden on health-care systems, vaccines can save countless lives and improve community well-being. According to World Health Organization (WHO) data, immunization currently prevents 3.5–5 million fatalities annually by targeting diseases such as pertussis, influenza, measles, diphtheria, and tetanus [2]. As biological agents, vaccines contain attenuated or inactivated pathogens or their derivatives, prompting the immune system to produce antibodies, memory cells, and other immune responses that recognize and target specific pathogens [3]. Vaccines can be classified into two categories based on the type of pathogen: (a) viral (b) bacterial. A more detailed classification includes live attenuated vaccines, such as the measles–mumps–rubella (MMR) vaccine [4]; inactivated vaccines, such as influenza and hepatitis A vaccines; subunit vaccines, such as recombinant hepatitis B virus surface antigen vaccines and adjuvants [5,6]; and mRNA vaccines, such as those developed for COVID-19 [7].
Vaccines offer significant benefits, but also have potential side effects. Uveitis and ocular inflammation are relatively prevalent among these complications [8], with annual post-vaccine uveitis incidence ranging from 8 to 13 per 100,000 cases/year [9]. Uveitis involves inflammation of the uvea and the middle layer of the eye, including the iris, ciliary body, and choroid. Based on its anatomical location, it can be classified as anterior, intermediate, posterior, or pan-uveitis [10]. Uveitis often has a chronic relapsing course, leading to temporary or permanent vision impairment, making it one of the leading global causes of blindness [11]. According to the etiology, uveitis can be divided into two categories: infectious and noninfectious. Infectious uveitis includes bacterial, fungal, viral, spirochete, and parasitic diseases; infectious uveitis includes idiopathic, traumatic, autoimmune, and rheumatic immune diseases [12]. Viruses are a major cause of infectious uveitis. Half of the human herpes viruses, including herpes simplex virus (HSV) [12,13], varicella zoster virus (VZV) [14], and cytomegalovirus (CMV) [15], account for 5–10% of all uveitis cases [16,17,18]. These include rubella virus [19] and human T-lymphotropic virus 1 (HTLV-1) [20]. However, there is no clear evidence of a causal link between antiviral vaccination and uveitis. This study aimed to review the literature on the possible relationship between uveitis and antiviral vaccines.

2. Methods of Literature Search

The Medline [PubMed], Embase, and Cochrane Library databases (from 1983 to June 2023) were used to search the relevant publications that included case reports and series. All the sourced articles had their full text reviewed to ensure that the contents were relevant to the study.

3. Results

3.1. General Analysis Regarding Cases of Uveitis Post-Vaccination

After conducting a comprehensive review, a total of 51 reported cases (involving 61 patients), spanning from 1978 to 2023, have been associated with uveitis following various vaccinations [21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71] (Table 1), including 4 case reports (5 patients) related to hepatitis B virus (HBV) vaccination [28,29,30,31]; 7 case reports (8 patients) related to human papillomavirus (HPV) vaccination [21,22,23,32,33,34,35]; 16 cases (18 patients) regarding uveitis after influenza vaccination [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]; 3 cases (4 patients) related to measles–mumps–rubella (MMR) vaccination [52,53,54]; 12 cases (16 patients) related to varicella zoster virus (VZV) vaccination [24,25,26,27,55,56,57,58,59,60,61,62]; 4 cases (5 patients) for yellow fever [63,64,65,66]; one each for hepatitis A virus (HAV) [67] and rabies virus vaccination [68]; and 3 case reports involving mixed vaccine administrations [69,70,71].
The average age of the affected individuals was 38.5 years (range, 1–86 years), with a female predominance (female:male = 35:26). Based on the site of infection, this study included 11 cases of anterior uveitis [27,35,37,45,52,54,56,58,63], 1 case of intermediate uveitis [63], 8 cases of posterior uveitis [21,22,27,28,32,42], 3 cases of uveitis involving both anterior and intermediate segments [27,55,64], 1 case of anterior and posterior uveitis [38], and 7 cases of pan-uveitis [23,34,40,43,48,53]. Distinct types of uveitis, such as acute posterior multifocal placoid pigment epitheliopathy (APMPPE) [36,41,46,47,57], Vogt–Koyanagi–Harada syndrome (VKH) [31,39,49,51,65,66], multiple evanescent white dot syndrome (MEWDS) [30,33,50,67,68,69,70], acute retinal necrosis (ARN) [24,25,26,59,61,62], and uveitis sarcoidosis [60] were also included. Most patients presented with bilateral involvement (bilateral:unilateral = 36:25), and left eye involvement was more common among patients with unilateral involvement (left:right = 16:9). In addition to cases lacking specific timeframes, uveitis typically occurs within days to years after vaccination (range, 24 h to 3 years), with some cases showing symptoms within 24–48 h, suggesting a potential association between vaccines and uveitis.
Of the reported cases, 32 patients presented systemic symptoms [24,27,28,31,33,34,35,36,37,38,39,41,42,45,46,47,48,49,50,51,53,55,56,57,60,63,64,65,66,71]. Most patients achieved full recovery following anti-inflammatory medications, intravenous and/or oral corticosteroids, intraocular injections, and periocular administration. However, it is noteworthy that a subset of patients exhibited varying degrees of residual symptoms even after systematic and targeted treatment during the follow-up period (range, 1 month to 5 years).

3.2. Detailed Analysis by Vaccine

3.2.1. Human Papillomavirus (HPV) Vaccine

HPV is a sexually transmitted virus that, if persistently infected with high-risk genotypes (such as HPV 16 and 18), is strongly associated with cervical cancer [72,73]. Currently, there is no virus-specific treatment for HPV infections; however, preventive measures are available. Six prophylactic HPV vaccines have been licensed using recombinant DNA and cell culture techniques to produce purified L1 structural proteins that self-assemble into virus-like particles (VLPs) [74]. As of the end of 2022, the estimated global coverage rate for the first dose of HPV vaccination among girls was 21% [75]. It is noted that all types of HPV vaccines should be used in females aged 9 and above. They are licensed for use by people up to the age of 26 or 45 [76]. As awareness of HPV vaccines grows among women, monitoring of adverse reactions should also increase.
By 2023, numerous adverse events related to HPV have been reported, including autoimmune diseases such as scleroderma and rheumatoid arthritis [77]. However, ocular side effects are limited. Our study specifically focused on uveitis and compiled the reported cases of HPV-related uveitis to date (Table 1). Seven cases involving eight female patients (average age = 21.75 years) [21,22,23,32,33,34,35] developed uveitis at various locations following HPV vaccination (two anterior [35], one posterior [32], two pan-uveitis [23,34], one MEWDS [33], and two Harada-like uveitis [21,22]). Symptoms manifested within weeks, ranging from 4 days to 10 weeks after vaccination, with the majority (four out of eight) occurring after the third vaccine dose [21,22,34,35]. Except for one unreported case [35], approximately 42.9% (three out of seven) of patients exhibited systemic symptoms such as fever, hearing loss, sore throat, headache, and joint pain [33,34,35]. With an average follow-up period of 1.2 years, vision completely recovered in five patients following corticosteroid treatment [21,22,23,34,35], including one patient who recovered with eyedrops alone [21]. However, some patients experience prolonged effects, such as recurrent leakage [33] (Figure 1) and extensive macular scarring [32]. Furthermore, one patient required long-term medication to manage uveitis [35]. These enduring side effects are likely to persist throughout a patient’s lifetime and should therefore be treated with the utmost seriousness.
Three patients developed Harada-like uveitis [21,22,23], two of whom tested positive for HLA-DRB1*0405 [23] and DR04 and 07 suballeles [22]. In a recent meta-analysis, the odds ratios (OR) of developing VKH was 10.3 for HLA-DRB1*0405 [78]. However, rapid and significant recovery after a relatively short course of corticosteroid treatment is an atypical finding in Harada’s disease, which typically requires corticosteroid immunosuppressive therapy for at least 3–6 months [79]. Their clinical course more closely resembles that of vaccine-induced uveitis than that of coincidental autoimmune diseases. Tubulointerstitial nephritis and uveitis (TINU) syndrome is an autoimmune disorder widely suspected to be triggered by infections or drug hypersensitivity reactions [80]. Two patients with TINU syndrome following HPV vaccination have been reported [35], who exhibited clinical manifestations of interstitial nephritis, including glucosuria, proteinuria with low molecular weight, and elevated serum creatinine levels, and subsequently developed eye-related symptoms, such as redness and photophobia. Given the similarities between these two cases and their HPV vaccination histories, it is speculated that this could be related to HPV vaccine administration.
The WHO guidelines have acknowledged a “possible” association between HPV vaccination and uveitis [81]. Potential mechanisms include broad nonspecific immune responses or molecular mimicry. Comparative computer analysis of HPV 16 oncogenic proteins and human self-proteins has revealed molecular mimicry and demonstrated high and widespread similarity in numerous critical regulatory processes. Causality assessment is typically based on data regarding the administration, development of uveitis, and timing of rechallenge. However, existing case reports do not definitively establish a causal relationship and necessitate further investigation of the underlying mechanisms [82].
Considering these findings, health-care practitioners should be vigilant about the potential association between HPV vaccination and uveitis. When obtaining medical histories from patients with suspected uveitis, detailed inquiries about vaccination history and extraneous ocular manifestations should be made. Intravitreal injections may serve as a potential therapeutic option, especially for patients with contraindications to systemic corticosteroids. Extended follow-ups and proactive interventions are recommended for patients at risk of long-term adverse reactions.

3.2.2. Hepatitis B Virus (HBV) Vaccine

As a DNA virus [83], HBV is the most common cause of liver cancer worldwide [84]. The widespread adoption of the hepatitis B vaccine has significantly reduced the global prevalence of the virus [85]. By the end of 2022, hepatitis B vaccination for infants had been implemented nationwide in 190 member countries. The global coverage for the three-dose hepatitis B vaccine was estimated to reach 84% [75]. This vaccine is produced through recombinant DNA technology, utilizing purified HBsAg antigen, and is typically administered in three doses: within 24 h of birth, at 1 month, and at 6 months [86].
Research indicates that uveitis is the most frequent ocular complication following HBV vaccine administration [87]. Our study encompassed four reports (involving five patients) of HBV-related uveitis [28,29,30,31], ranging in age from 20 to 43 years, with a male-to-female ratio of 2:3. The ocular lesions were bilateral and included two patients diagnosed with APMPPE [29], one with MEWDS [30], one with VKH syndrome [31], and one with bilateral posterior uveitis [28]. Therefore, HBV vaccine-induced uveitis presents with diverse manifestations, primarily bilateral and posterior uveitis. Some patients report systemic symptoms such as headaches, hearing loss, and skin changes [28,31]. The average duration was 4.8 days (range, 1–14 days). During the follow-up period ranging from 3 to 9 months, two patients still exhibited residual paracentral scotomas and potential persistence [29], while one patient responded to oral and topical steroids, but required prolonged treatment [31].
A potential mechanism underlying the development of uveitis may involve a hypersensitivity reaction. Fried et al. speculated that the vaccine surface forms immune complexes with hepatitis B antibodies, leading to uveitis [28]. While there is an ongoing debate about the role of this mechanism [88,89], evidence suggests immune complex deposition diseases, such as cryoglobulinemia and glomerulonephritis, during chronic hepatitis B infection [90,91,92]. Aguirre et al. conducted a study reinforced this perspective by discovering type III hypersensitive antigen-antibody complexes in the aqueous humor of dogs after adenovirus vaccination [90]. Another potential mechanism may involve the adjuvant effects of the vaccine vectors. Adjuvants, which are often combined with vaccines to enhance immunogenic activity, have been implicated in the development of uveitis in animal models [93]. Other autoimmune diseases, including multiple sclerosis and dermatomyositis, have been reported after hepatitis B vaccination, with a potential role in the molecular mimicry between retinal pigment epithelial protein and hepatitis B surface antigen [94,95,96,97,98]. A case report concerning the development of APMPPE after immunization with a recombinant hepatitis B virus vaccine suggested that the hepatitis B surface antigen may trigger immune-mediated retinal pigment epithelium disruption or choroidal vascular occlusion [29]. To some extent, these mechanisms contribute to establishing a causal link between hepatitis B vaccination and uveitis.
In summary, patients typically develop uveitis-related ocular symptoms shortly after vaccine administration. Clinicians should be attentive to such cases and consider uveitis-related examinations and targeted treatments for affected individuals.

3.2.3. Influenza Virus Vaccine

Influenza is a widespread febrile illness that causes millions of severe cases, resulting in 290,000 to 650,000 deaths annually [99]. Vaccination is a pivotal strategy for preventing and controlling the spread of influenza A and B viruses, which are the primary causes of human seasonal influenza [100]. According to statistics, vaccination coverage for seasonal influenza among high-risk groups varies significantly across countries. The vaccination rates for chronic disease patients and health-care workers in most countries fall below 40% [101]. Various types of influenza vaccines, including live attenuated influenza vaccines (LAIV4), inactivated influenza vaccines (IIV4s), and recombinant influenza vaccines (RIV4) are currently available [102]. Owing to continuous antigenic changes in influenza viruses, health authorities engage in annual surveillance to identify the strains most likely to cause illness in the upcoming flu season [103]. In general, influenza vaccines are safe and effective, with minor side effects, such as local swelling, joint pain, and low-grade fever. Severe adverse events are extremely rare. However, in recent years, ocular reactions attributed to influenza vaccination have been reported, including, reactivation of herpes simplex keratitis, corneal transplant rejection [104], “ocular respiratory syndrome” (conjunctivitis and respiratory symptoms) [105], and bilateral optic neuropathy [106]. Uveitis is a relatively common ocular manifestation of influenza vaccination.
Sixteen cases (involving eighteen patients) following influenza vaccination have been reported [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. Most cases have been associated with live attenuated vaccines [36,42,43,45,48,49]. The final diagnoses varied and included conditions such as APMPPE [36,41,47,50], MEWDS [44,50], VKH disease [39,49,51], uveitis involving both anterior and posterior segments [38], anterior uveitis [37,45] and posterior uveitis [42] to pan-uveitis [39,40,43,48]. The sex distribution among the patients was balanced, with an average age of 42.7 years (range, 10–78 years), indicating that influenza vaccines may affect individuals of all ages without sex disparity. Nearly all the patients exhibited flu-like systemic symptoms. On average, ocular symptoms started 12.7 days after vaccination, with some cases manifesting within 24 h after the first dose [40]. When the initial flu-like symptoms fail to resolve, most patients are treated with topical or systemic corticosteroids and show significant improvement, suggesting that this is an autoimmune response rather than an infection. The follow-up period ranged from 1 month to 5 years, and 8 patients experienced significant symptom alleviation [38,39,40,42,44,45,46]. Patients with APMPPE and MEWDS often exhibit extensive white lesions on fundus examination and OCT, with residual retinal pigment epithelium (RPE) changes observed during long-term follow-up (Figure 2). Severe vision loss occurred in three patients, with one patient’s left eye remaining devoid of light perception even after one year of follow-up following systemic corticosteroid therapy [48]. Tao et al. reported the case of a 47-year-old female developed systemic fever, headache, flu-like symptoms, and ocular manifestations two days after vaccination [42]. The diagnoses included bilateral posterior uveitis and exudative retinal detachment (ERD). Although the exudative retinal detachment recovered after steroid therapy, visual acuity remained poor (0.01), possibly due to a prolonged disease course. Therefore, if not managed promptly and effectively, patients with suspected vaccine-induced uveitis may be at risk of blindness.
Uveitis can result from immune factors triggered by molecular mimicry between vaccine peptide fragments and retinal self-antigens, delayed hypersensitivity reactions [4,107,108]. It has been proposed that in live attenuated vaccines, inflammation might arise from viral peptide-induced reactions [4], whereas inactivated or subunit/conjugate vaccines can trigger inflammation from adjuvants, such as aluminum salts [107].
In summary, clinicians should monitor patients with a history of influenza vaccination who present with flu-like symptoms shortly after vaccination, and conduct comprehensive ocular examinations to assess the risk of uveitis. Absence of response to antibiotic treatment and negative test results. The absence of response to antibiotic treatment and negative test results can provide vital cues. Moreover, prompt initiation of judicious corticosteroid therapy upon diagnosis is imperative because delayed treatment may lead to adverse outcomes, including blindness. Considering the evolving composition of influenza vaccines each year to combat rapidly mutating viruses, we recommend annual influenza vaccination.

3.2.4. Measles–Mumps–Rubella (MMR) Vaccine

The MMR vaccine, a combination of three live attenuated viruses, was licensed in 1971 [54]. Currently, 83% of children receive the first dose of measles vaccine before turning two, and 74% receive the second dose. The global coverage for the rubella vaccine is estimated at 68%, and the mumps vaccine has been implemented in 123 member countries worldwide [75]. The Advisory Committee on Immunization Practices recommends administration of the first dose at the age of 12–15 months and the second dose at 4–6 years. Rubella vaccine can be administered alone or in combination with measles and mumps vaccines [109], reducing the incidence of severe congenital rubella syndrome, which is characterized by deafness, ocular abnormalities, and congenital heart defects [110].
Our study included four cases of MMR vaccine-associated uveitis, with an average age of 11 years old and a male-to-female ratio of 3:1. Almost all the patients achieved complete recovery during the follow-up period after local or systemic steroid treatment. Surprisingly, case reports since 2004 have used polymerase chain reaction (PCR) and Goldmann–Witmer coefficient (GWC) analysis of anterior chamber fluid to identify a correlation between rubella virus (RV) and Fuchs’s heterochromic iridocyclitis syndrome (FHI) [16,111,112,113]. In a retrospective study involving 127 patients, all Fuchs’s heterochromic iridocyclitis cases were RV-positive except for two (5%) [113]. This aligns with a case report we collected [54], detailing a 12-month-old Caucasian girl who developed conjunctival redness, photophobia, heterochromia, and redness of the iris three months after receiving the MMR-attenuated live vaccine. Serological analysis revealed RV immunoglobulin G (IgG) positivity, underscoring the potential pathogenic role of the MMR vaccine.
Multiple factors may contribute to the development of severe uveitis following MMR vaccine administration: (a). Antigenic mimicry: evidence suggests that immune activation pathways induced by antigenic mimicry may lead to uveitis following administration of attenuated live MMR vaccine [52]. (b). Vaccine contamination: contamination of the vaccine with toxins, antigens, or vaccine modifications due to improper storage may play a role in uveitis induction. (c). Nature of attenuated live vaccines: the survival of viruses within attenuated live vaccines may contribute to the occurrence of uveitis. (d). Coincidence: although rare, the possibility of pure coincidence cannot be ruled out.
In conclusion, most patients achieved a complete recovery shortly after steroid treatment. However, refractory uveitis and other severe consequences may occur in specific instances. Our findings underscore the importance of increased ocular awareness following the MMR vaccination. Close monitoring, particularly among adolescents, is recommended for all recipients. Furthermore, for young patients exhibiting signs of Fuchs’s syndrome, such as heterochromia and posterior synechiae, aqueous humor examination is recommended to assess the risk of RV infection.

3.2.5. Varicella Zoster Virus (VZV) Vaccine

VZV can cause herpes zoster following primary infection [114], primarily affecting individuals aged 70 years and older, potentially leading to severe complications such as postherpetic neuralgia [115]. These challenges underscore the compelling rationale for developing effective immunization strategies against VZV in the elderly population [116]. Subcutaneously administered VZV vaccines include Varivax (Merck, Whitehouse Station, NJ, USA), a live attenuated varicella vaccine for infants; Zostavax (Merck), approved for the secondary prevention of herpes zoster reactivation in individuals 75 years and older; and the recombinant subunit vaccine (RZV, Shingrix), introduced in 2017 [117].
Our study consists of 12 cases comprising 16 patients with VZV-associated uveitis [24,25,26,27,55,56,57,58,59,60,61,62]. Patients had an average age of 55.8 years and a 1:1 sex distribution. Nearly all patients exhibited primary viral infection symptoms, with the onset of symptoms varying from 24 h to 3 years, possibly because of the latent nature of the virus. Underlying conditions were present in 12 patients, including metabolic disorders such as diabetes [24], impaired glucose tolerance [59], insulin-dependent diabetes [62], and cirrhosis [59]; necessitating immunosuppressive treatment for primary conditions, including kidney transplantation [24], chronic lymphocytic leukemia [62], inflammatory bowel disease [25], autoimmune diabetes [26], and multiple myeloma [61]; ocular disorders, such as herpes zoster ophthalmicus (HZO) with anterior uveitis [58], multifocal choroiditis [27], and viral keratouveitis [27]. Most cases have been treated with antiviral drugs and/or corticosteroids. An unfavorable prognosis includes significant visual acuity decline [62], requiring prolonged medication for secondary choroidal neovascularization and residual scars [26,56].
Zostavax, which contains the Oka strain of VZV, has been linked to some cases, particularly in immunocompromised individuals (Table 1). In three documented cases, patients with different medical histories who received immunosuppressive therapy developed ARN shortly after VZV vaccination [24,25,26]. Two of these patients tested positive for the Oka strain of the VZV vaccine [25,26], suggesting a potential link between vaccine strain infection and ARN. However, VZV DNA tests conducted in seven cases revealed that five patients had wild-type VZV, indicating that most ARN cases may have resulted from VZV reactivation rather than vaccine reactivation. Moreover, the majority of patients had systemic metabolic disorders, such as diabetes, cirrhosis, and inflammatory gastrointestinal diseases, which may contribute to the occurrence of ARN after post-VZV vaccination.
Recently, a safer alternative to live-strain vaccines has emerged with the approval of the recombinant subunit vaccine RZV (Shingrix, GlaxoSmithKline, Research Triangle Park, NC, USA). This vaccine, which contains the novel adjuvant AS01B, stimulates robust immune responses, potentially inducing persistent cell-mediated immunity [118]. However, adjuvant vaccines such as RZV can increase the likelihood of immune-mediated events, particularly in patients with known inflammatory conditions. To date, only five cases of RZV-associated uveitis have been reported. Heydari-Kamjani et al. reported a case of uveitis sarcoidosis in a 53-year-old female following RZV injection, with initial ocular symptoms occurring only four days after Shingrix vaccination [60]. Chen et al. documented a 65-year-old female with multiple myeloma who developed ARN in her left eye six weeks after RZV vaccination but recovered with valacyclovir and intravitreal foscarnet [61]. More recently, Richards et al. documented three cases of reactivation of previously controlled uveitis after receiving RZV, with various degrees of improvement following antiviral or corticosteroid treatment [27] (Figure 3).
In contrast to the uveitis pathogenesis caused by direct infection from attenuated but still active live attenuated vaccines, there may be two potential mechanisms: (1) the adjuvant AS01B, demonstrated to induce uveitis in various in vitro studies, such as monophosphorylate lipid A (MPL) [118]. (2) Shingrix vaccine production, which involves Chinese hamster ovary (CHO) cells, may result in trace amounts of host cell proteins (HCPs) in the final vaccine product, potentially triggering autoimmune responses [118].
Consequently, vaccination suitability should be rigorously evaluated in patients with underlying inflammatory conditions and those receiving immunosuppressive agents. Live attenuated vaccines may not be the optimal choice for patients with compromised immune function or immunosuppressants. Where possible, PCR testing to determine the VZV DNA type could aid in identifying potential causes of infection and guide targeted therapeutic interventions. Antiviral drugs and corticosteroids have demonstrated efficacy, and vitrectomy may be performed when necessary.

3.2.6. Yellow Fever Vaccine/Hepatitis A Virus (HAV) Vaccine/Co-Administration

Although rarely reported as a uveitis trigger, the yellow fever vaccine, a live attenuated vaccine, has been associated with autoimmune disorders such as neuromyelitis optica spectrum disorder [119]. Only four reports (involving five patients) were included [64,65,66,67], with an average age of 34.4 years and a male-to-female ratio of 3:2. All patients developed symptoms within 3 weeks post-vaccination and initially presented with systemic manifestations. Treatment with local and/or systemic corticosteroids led to improvements. In rare severe adverse reactions, such as yellow fever vaccine-associated viscerotropic and neurological diseases, particular attention is needed. Volkov et al. reported a patient with bilateral acute anterior uveitis and intermediate uveitis secondary to viscerotropic disease after post-vaccination [64]. Initially, severe systemic symptoms included fever, cough, dyspnea, malaise, sore throat, non-bloody diarrhea, trunk maculopapular rash, and yellow fever virus RNA (+). After corticosteroid treatment, the patient improved but experienced persistent fatigue for several months. The mechanism of uveitis after yellow fever vaccination is unknown, with some studies suggesting that immune responses disrupt prior tolerance [65], whereas others propose molecular mimicry-triggered immune cross-reactivity and subsequent autoimmune disease [63].
The hepatitis A virus vaccine (HAVV) effectively prevents hepatitis A, a virus typically transmitted through contaminated water or shellfish consumption. Our study included only one patient diagnosed with left eye MEWDS post-vaccination who recovered spontaneously within six weeks without treatment [67].
Yellow fever and hepatitis A vaccines are frequently administered as prophylaxis in travelers. Our study includes two cases of concurrent hepatitis A and yellow fever vaccine administration. A 33-year-old male received both vaccines, and typhoid vaccine before a trip to Panama (having previously received a single dose of hepatitis A vaccine) [71], he experienced intermittent scotomas and flashes in the right eye, along with systemic symptoms such as fever, headache, and rash. Another patient experienced rapid progressive painless vision loss in the left eye 10 days after co-administration [69]. Both patients showed spontaneous improvement without treatment. Neither report distinctly leans towards any specific vaccine as the causative factor. In conclusion, the use of yellow fever and hepatitis A vaccines as travelers’ immunization choices warrants clinicians’ awareness of the potential for co-administration to induce uveitis.

3.3. Different Types of Uveitis Induced by Antiviral Vaccines

In our investigation, anterior uveitis was predominant among patients, most of whom responded favorably to corticosteroid therapy, mirroring findings from a previous meta-analysis [87]. However, the possibility of posterior uveitis could not be ruled out. Our data included 31 patients presenting with posterior uveitis, encapsulating specific syndromes such as APMPPE, MEWDS, ARN, and Harada-like diseases.

3.3.1. Acute Posterior Multifocal Placoid Pigment Epitheliopathy (AMPEE) and Multiple Evanescent White Dot Syndrome (MEWDS)

Both APMPPE and MEWDS are categorized as primary inflammatory diseases of the choroid and retinal vasculature diseases (PICCP) [120,121], typically manifesting as unilateral or bilateral posterior uveitis. We documented seven cases of APMPPE [36,41,46,47,57] and eight cases of MEWDS [30,33,50,67,68,69,70] after vaccination. The age at onset varied between 11 and 31 years, with a male-to-female ratio of 4:3. All cases revealed bilateral involvement, consistent with previous multicenter retrospective studies [122]. Notably, five patients presented with flu-like symptoms [36,41,46,47,57] a few days before visual symptoms. Whether these symptoms directly contribute to APMPPE or are associated with an increased susceptibility remains unclear.
Patients with APMPPE frequently report acute or subacute visual blurring, scotoma, and metamorphopsia. Dilated fundus examination and optical coherence tomography (OCT). alterations in RPE and deep yellow–white placoid lesions were discerned. Indocyanine green angiography commonly reveals hypofluorescent spots [123]. Fluorescein angiography (FA) showed multifocal early hypofluorescence, followed by late hyperfluorescence. After the resolution of active lesions, permanent changes in the RPE pigmentation dispersion led to projection defects; however, late staining or leakage disappeared with time. Among the cases we included, 71.4% of patients exhibited residual RPE changes during the follow-up period [29,36,41,47]. In some cases, choroidal vessels can be observed at the center of the dark areas [120,124].
MEWDS is an idiopathic, self-limiting retinitis that predominantly affects young women and often presents unilaterally. Among the eight patients, ages ranged from 16 to 53 years, with a male-to-female ratio of 2:6, and the majority occurred in females. Common features include multiple small white-yellow lesions in the outer retina and RPE. Fluorescein angiography indicates early multifocal high-fluorescence spots, followed by late staining of the fovea, usually presenting with orange granularity [122]. Subjective symptoms include decreased visual acuity, photosensitivity, scotomas, and visual field defects. Both APMPPE and MEWDS often show widespread white lesions on fundus examination and persistent RPE changes on OCT.

3.3.2. Vogt–Koyanagi–Harada (VKH)

Harada disease is characterized by bilateral posterior uveitis with optic disc swelling, macular edema, and retinal exudates. When combined with Vogt–Koyanagi disease, which primarily involves anterior segment inflammation, it is classified as a VKH disease [125]. VKH is a rare granulomatous inflammatory condition that targets melanocyte-rich tissues such as the choroid, meninges, skin, and hair follicles. Ocular manifestations include bilateral pan-uveitis, serous retinal detachment, and optic disc hyperemia. The disease comprises four consecutive stages: prodromal, acute uveitis, convalescent, and chronic recurrence [126].
Our analysis covered eight relevant studies on Harada-like diseases with a median gap of 11 days between vaccination and symptom emergence, strongly suggesting a correlation between post-influenza vaccination and VKH. All the patients demonstrated bilateral involvement. The pathogenesis of VKH is theorized to involve a T cell-driven autoimmune response against melanocyte antigens, which is closely associated with the HLA-DR4/HLA-DRB104 alleles [78]. We identified three patients exhibiting allele positivity: HLA-DRB10405 positive [23], HLA-DR04 and 07 positive [22], and HLA-DR4 positive [51]. A significant majority of the patients reported relief after corticosteroid therapy.

3.3.3. Acute Retinal Necrosis (ARN)

ARN is rare. According to the American Uveitis Society, it manifests as over one zone of retinal necrosis, progresses gradually if left untreated with antivirals, encompasses occlusive arterial vascular involvement, and induces a pronounced inflammatory response in the anterior chamber and vitreous cavity [127]. VZV is the main cause of ARN, followed by HSV [128].
This review discusses eight cases of ARN, all post-VZV vaccination. The median age was 65 years (range 20–80 years), highlighting ARN’s potential of ARN to affect all age groups, with a predilection for the elderly population. The male-to-female ratio was 5:3. Unlike earlier reports [129,130], bilateral disease was not as common; 75% (six of eight) of patients had unilateral involvement, possibly because of prompt detection and efficient antiviral intervention.

4. Discussion

Although uncommon and cause being difficult to prove, there are several reports of adverse ocular reactions to various vaccines [131,132,133]. A recent review of vaccine-induced ocular adverse events for 2010–2020 highlighted ocular adverse effects associated with multiple vaccines, including optic neuritis, uveitis, and retinal inflammations [131]. Additionally, several reports in recent years have explored the link between vaccines and uveitis [87,134,135,136,137,138,139]. A review published by Benage et al. in 2016 identified hepatitis B vaccine (administered alone or with other vaccines) as the leading cause of vaccine-induced uveitis, followed by human papillomavirus virus (HPV), and influenza vaccine [87]. In this systematic review, we summarize the various types of uveitis triggered by different vaccines and provide detailed descriptions of specific uveitis entities. Our findings suggest that post-vaccination uveitis can affect individuals of all age groups, with a potentially higher susceptibility among women. However, due to the varying indications and circumstances of immunization, it is challenging to draw specific conclusions regarding patient sex and age. It is important to note that the vaccines used in these cases included a variety of formulations, reflecting considerations that may differ depending on the year and underlying conditions of the patient.
In contrast to the study by Benage et al. [87], 18 patients with post-vaccination uveitis were recruited from the influenza vaccine group, which appeared to be the most likely cause of uveitis, followed by vaccination with varicella zoster virus and human papillomavirus (HPV). Patients with influenza vaccine-associated uveitis often present with systemic flu-like symptoms. Factors contributing to these adverse effects may include residual activity from attenuated vaccines and molecular mimicry of immune responses. Therefore, when patients resist antiviral therapy and the test results are negative, clinicians should consider the possibility of vaccine-induced immune-related uveitis. Patients with uveitis following VZV vaccination often have severe systemic disease and poor prognosis. Some patients even succumbed to primary diseases during the follow-up period [59], suggesting that individuals with severe underlying or immune system-related diseases may be more susceptible to adverse reactions to VZV vaccination. This may be due to potential susceptibility to attenuated VZV vaccines. Currently, a novel recombinant VZV vaccine (Shingrix) that greatly benefits patients with underlying conditions is available. However, there are still reports of uveitis after vaccination. Future studies should focus on investigating post-Shingrix uveitis cases. In 2014, Holt et al. reported 24 cases of HPV vaccine-related uveitis in females with a median age of 17 years [139]. Similarly, in our study, all patients with HPV-related uveitis were women, suggesting the need to monitor systematic and ocular manifestations following HPV vaccination. Fraunfelder et al. reported 32 cases of HBV-related uveitis in 2010, with 72.7% of the patients experiencing symptoms after the first administration [138]. Unlike our findings, nearly all the patients in their study recovered after treatment without evidence of long-term vaccine-related adverse effects. This discrepancy may be related to variations in follow-up time and vaccine type. Moreover, definitively attributing causality to a specific vaccine in patients receiving combinations or a series of vaccines is challenging. These adverse effects could potentially be related to adjuvant-containing HAV vaccines [71].
A deeper understanding of “rechallenge” can offer compelling clinical evidence. One patient experienced bilateral posterior uveitis three days after the second dose of the HBV vaccine [29]. Improvement occurred following systemic treatment, yet the patient disregarded the medical advice and received a third dose, leading to recurrence of posterior uveitis. This case provides evidence for a causal relationship between uveitis and vaccine administration. Furthermore, the “exacerbation” and “recurrence” of preexisting uveitis following vaccination warrant attention. A patient with anterior uveitis associated with herpes zoster exhibited worsening of the pre-existing condition three weeks after receiving the VZV vaccine [58]. In 2021, Richards et al. [27] reported on three patients with a history of controlled ocular inflammation of varying degrees and locations who experienced disease recurrence after receiving the VZV vaccine. Potential causes include cell-mediated responses to recombinant zoster vaccines (RZV), reactions to residual viral DNA, vaccine failure, and use of immunosuppressants.
Our review also highlights specific uveitis entities, including MEWDS, AMPMEE, VKH, and ARN. MEWDS and APMPPEE are generally associated with favorable prognoses, given their self-limiting course, which often requires no treatment. While there is no evidence suggesting that corticosteroid therapy affects the final visual acuity, it is still recommended in cases of significant macular involvement or systemic complications. Fiorce et al. analyzed the visual prognosis in patients with APMPPEE and found that the probability of achieving a final visual acuity of 20/25 was less than 40% in cases with foveal involvement, whereas the likelihood of visual recovery beyond 20/25 was nearly 90% in patients without foveal damage [140]. The prognosis for most patients with VKH is good, but if left untreated, it can lead to complications such as glaucoma, cataracts, choroidal neovascularization, and retinal atrophy [141]. Patients diagnosed with ARN often have a history of metabolic disease or immunosuppressant use. One study found that nearly 30% of cases exhibited immunodeficiency, correlating with a more severe disease [142]. In addition, all patients with ARN in our review developed this condition after VZV vaccination; however, the mechanism underlying VZV vaccine-related uveitis remains unclear. All patients tested positive for VZV DNA, although only a few studies have investigated genotyping owing to equipment variations. For patients testing positive for wild-type VZV DNA, post-vaccination immunosuppression might reactivate latent VZV. For those testing positive for the VZV Oka strain, this may be related to the attenuated VZV vaccine. However, it is essential to emphasize that this review provides a direction for future research, and that precise conclusions necessitate further validation through fundamental research.
Interestingly, some patients did not fully recover during the posttreatment follow-up period. These cases included those with residual retinal pigment epithelium (RPE) changes [29,36,41,47,50,71], the need for long-term medical treatment due to incomplete recovery from uveitis [31,35], extensive macular scars [32], documented recurrent leakage on fluorescein angiography (FA) [33], no light perception in the left eye [48], irreversible blind spot enlargement [121], severe drops in visual acuity [42,48,62], and the presence of a sunset glow fundus [51,66]. In future studies, we will analyze the long- and short-term side effects of the vaccine in detail.
The mechanisms underlying post-vaccination uveitis remain unclear, but they are believed to share a pathogenesis similar to that of post-infectious uveitis. The four proposed mechanisms for vaccine-induced uveitis include the following. (1) Direct infection: live attenuated vaccines, such as poliovirus, MMR, and yellow fever, may introduce direct infection with attenuated yet active viral strains [134]. (2) Adjuvant-induced syndrome: adjuvant vaccines (especially aluminum-containing vaccines) such as HPV, HBV, and influenza may trigger a syndrome entitled ASIA (autoimmune/inflammatory syndrome induced by adjuvants; Shoenfeld syndrome). Upon injection, these vaccines do not readily dissolve in the extracellular space but instead aggregate at the injection site, forming aluminum conglomerates. This delayed dissolution allows injected aluminum particles to be rapidly captured by immune system cells and transported to various organs, including the brain, where they can induce systemic symptoms [143]. (3) Delayed hypersensitivity reactions: recombinant vaccines such as HBV and HPV may induce delayed hypersensitivity reactions, and the deposition of immune complexes with subsequent complement activation causes uveitis [144]. (4) Cross-reactivity: this occurs when peptide fragments presented to T cells closely resemble conformationally similar peptides of the uvea, triggering an immune response against uveal antigens [145]. Experimental evidence suggests that melanoma-associated molecules present in the choroid can induce ocular inflammation [146].
Despite these findings, our study has certain limitations. While uveitis has been observed after vaccination, it is possible that the international literature predominantly reports only severe cases. This might introduce a reporting bias. Nevertheless, the overall benefits of vaccines remain paramount for global health. Additionally, our study encompasses a significant period, from 1978 to 2023. It is plausible that updates in vaccine products and disparities in vaccine quality among different companies could influence the types of uveitis. However, our study did not delve into these specific details. This limitation presents a pathway for future research, enabling a more in-depth exploration. Further investigation into these specific variables is warranted to gain a nuanced understanding of their impact on uveitis epidemiology.

5. Conclusions

This is the first comprehensive study to date that provides a detailed analysis of the various types of uveitis triggered by antiviral vaccines. Through an extensive database search, we identified a strong link with influenza vaccines, followed by VZV and HPV vaccines. Although anterior uveitis is common, conditions such as APMPPE, MEWDS, and VKH are prominent and deserve attention in clinical practice. The efficacy of corticosteroid treatment was evident; however, half of the observed patients did not achieve full recovery, suggesting possible prolonged effects of the vaccine. Future studies should prioritize long-term monitoring and delve more deeply into the vaccine-induced responses.

Author Contributions

Conceptualization, K.K.; writing—original draft preparation, Y.Z. (Yaru Zou); writing—review and editing, K.K., Y.Z. (Yuan Zong), J.Z., M.Y. and K.O.-M.; funding acquisition, K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI (grant JP 20K09824), a Grant on Rare and Intractable Diseases from the Ministry of Health, Labour, and Welfare of Japan (grant 22FC0201), and a Research Program on Emerging and Re-emerging Infectious Diseases grant from the Japan Agency for Medical Research and Development, AMED (grant 23fk0108671h0001, 23fk0108672h0001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data related to this study are presented and published here.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maldonado, Y.A. Current Controversies in Vaccination: Vaccine Safety. JAMA 2002, 288, 3155–3158. [Google Scholar] [CrossRef] [PubMed]
  2. Vaccines and Immunization. Available online: https://www.who.int/health-topics/vaccines-and-immunization (accessed on 28 July 2023).
  3. Coffman, R.L.; Sher, A.; Seder, R.A. Vaccine Adjuvants: Putting Innate Immunity to Work. Immunity 2010, 33, 492–503. [Google Scholar] [CrossRef] [PubMed]
  4. Minor, P.D. Live Attenuated Vaccines: Historical Successes and Current Challenges. Virology 2015, 479–480, 379–392. [Google Scholar] [CrossRef] [PubMed]
  5. Pérez, O.; Romeu, B.; Cabrera, O.; González, E.; Batista-Duharte, A.; Labrada, A.; Pérez, R.; Reyes, L.M.; Ramírez, W.; Sifontes, S.; et al. Adjuvants Are Key Factors for the Development of Future Vaccines: Lessons from the Finlay Adjuvant Platform. Front. Immunol. 2013, 4, 407. [Google Scholar] [CrossRef]
  6. Smith, J.; Lipsitch, M.; Almond, J.W. Vaccine Production, Distribution, Access, and Uptake. Lancet 2011, 378, 428–438. [Google Scholar] [CrossRef]
  7. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. MRNA Vaccines—A New Era in Vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef]
  8. Agarwal, M.; Majumder, P.D.; Babu, K.; Konana, V.K.; Goyal, M.; Touhami, S.; Stanescu-Segall, D.; Bodaghi, B. Drug-Induced Uveitis: A Review. Indian J. Ophthalmol. 2020, 68, 1799–1807. [Google Scholar] [CrossRef]
  9. Kuniyoshi, K.; Hatsukawa, Y.; Kimura, S.; Fujino, T.; Ohguro, H.; Nakai, R.; Sunami, K.; Mishima, S.; Sato, T.; Kusaka, S.; et al. Acute Bilateral Photoreceptor Degeneration in an Infant After Vaccination Against Measles and Rubella. JAMA Ophthalmol. 2017, 135, 478–482. [Google Scholar] [CrossRef]
  10. Standardization of Uveitis Nomenclature (SUN) Working Group. Classification Criteria for Multiple Evanescent White Dot Syndrome. Am. J. Ophthalmol. 2021, 228, 198–204. [Google Scholar] [CrossRef]
  11. Ronday, M.J.; Stilma, J.S.; Barbe, R.F.; Kijlstra, A.; Rothova, A. Blindness from Uveitis in a Hospital Population in Sierra Leone. Br. J. Ophthalmol. 1994, 78, 690–693. [Google Scholar] [CrossRef]
  12. Gritz, D.C.; Wong, I.G. Incidence and Prevalence of Uveitis in Northern California; the Northern California Epidemiology of Uveitis Study. Ophthalmology 2004, 111, 491–500; discussion 500. [Google Scholar] [CrossRef] [PubMed]
  13. Sancho-Shimizu, V.; Zhang, S.-Y.; Abel, L.; Tardieu, M.; Rozenberg, F.; Jouanguy, E.; Casanova, J.-L. Genetic Susceptibility to Herpes Simplex Virus 1 Encephalitis in Mice and Humans. Curr. Opin. Allergy Clin. Immunol. 2007, 7, 495. [Google Scholar] [CrossRef] [PubMed]
  14. Kinchington, P.; Leger, A.; Guedon, J.; Hendricks, R. Herpes Simplex Virus and Varicella Zoster Virus, the House Guests Who Never Leave. Herpesviridae 2012, 3, 5. [Google Scholar] [CrossRef] [PubMed]
  15. Crough, T.; Khanna, R. Immunobiology of Human Cytomegalovirus: From Bench to Bedside. Clin. Microbiol. Rev. 2009, 22, 76–98. [Google Scholar] [CrossRef] [PubMed]
  16. Chee, S.-P.; Bacsal, K.; Jap, A.; Se-Thoe, S.-Y.; Cheng, C.L.; Tan, B.H. Clinical Features of Cytomegalovirus Anterior Uveitis in Immunocompetent Patients. Am. J. Ophthalmol. 2008, 145, 834–840. [Google Scholar] [CrossRef] [PubMed]
  17. Miserocchi, E.; Fogliato, G.; Bianchi, I.; Bandello, F.; Modorati, G. Clinical Features of Ocular Herpetic Infection in an Italian Referral Center. Cornea 2014, 33, 565–570. [Google Scholar] [CrossRef]
  18. Pleyer, U.; Chee, S.-P. Current Aspects on the Management of Viral Uveitis in Immunocompetent Individuals. Clin. Ophthalmol. 2015, 9, 1017–1028. [Google Scholar] [CrossRef]
  19. De Groot-Mijnes, J.D.F.; de Visser, L.; Rothova, A.; Schuller, M.; van Loon, A.M.; Weersink, A.J.L. Rubella Virus Is Associated with Fuchs Heterochromic Iridocyclitis. Am. J. Ophthalmol. 2006, 141, 212–214.e1. [Google Scholar] [CrossRef]
  20. Kamoi, K.; Mochizuki, M. HTLV-1 Uveitis. Front. Microbiol. 2012, 3, 270. [Google Scholar] [CrossRef]
  21. Kong, K.; Ding, X.; Ni, Y. Resolution of Harada Disease-like Uveitis after Quadrivalent Human Papillomavirus Vaccination: A Case Report. Hum. Vaccines Immunother. 2022, 18, 1–4. [Google Scholar] [CrossRef]
  22. Ye, H.; Feng, H.; Zhao, P.; Fei, P. Case Report: Posterior Uveitis after Divalent Human Papillomavirus Vaccination in an Asian Female. Optom. Vis. Sci. 2020, 97, 390–394. [Google Scholar] [CrossRef] [PubMed]
  23. Dansingani, K.K.; Suzuki, M.; Naysan, J.; Samson, C.M.; Spaide, R.F.; Fisher, Y.L. Panuveitis with Exudative Retinal Detachments After Vaccination Against Human Papilloma Virus. Ophthalmic Surg. Lasers Imaging Retin. 2015, 46, 967–970. [Google Scholar] [CrossRef] [PubMed]
  24. Charkoudian, L.D.; Kaiser, G.M.; Steinmetz, R.L.; Srivastava, S.K. Acute Retinal Necrosis after Herpes Zoster Vaccination. Arch. Ophthalmol. 2011, 129, 1495–1497. [Google Scholar] [CrossRef] [PubMed]
  25. Gonzales, J.A.; Levison, A.L.; Stewart, J.M.; Acharya, N.R.; Margolis, T.P. Retinal Necrosis Following Varicella-Zoster Vaccination. Arch. Ophthalmol. 2012, 130, 1355–1356. [Google Scholar] [CrossRef] [PubMed]
  26. Heath, G.; Depledge, D.P.; Brown, J.R.; Hale, A.D.; Tutil, H.; Williams, R.; Breuer, J. Acute Retinal Necrosis Caused by the Zoster Vaccine Virus. Clin. Infect. Dis. 2017, 65, 2122–2125. [Google Scholar] [CrossRef] [PubMed]
  27. Richards, P.J.; Wingelaar, M.J.; Armbrust, K.R.; Kopplin, L.J. Uveitis Reactivation Following Recombinant Zoster Vaccination. Am. J. Ophthalmol. Case Rep. 2021, 23, 101115. [Google Scholar] [CrossRef]
  28. Fried, M.; Conen, D.; Conzelmann, M.; Steinemann, E. Uveitis after hepatitis B vaccination. Lancet 1987, 330, 631–632. [Google Scholar] [CrossRef]
  29. Brézin, A.P.; Massin-Korobelnik, P.; Boudin, M.; Gaudric, A.; LeHoang, P. Acute Posterior Multifocal Placoid Pigment Epitheliopathy after Hepatitis B Vaccine. Arch. Ophthalmol. 1995, 113, 297–300. [Google Scholar] [CrossRef]
  30. Baglivo, E.; Safran, A.B.; Borruat, F.X. Multiple Evanescent White Dot Syndrome after Hepatitis B Vaccine. Am. J. Ophthalmol. 1996, 122, 431–432. [Google Scholar] [CrossRef]
  31. Sood, A.B.; O’Keefe, G.; Bui, D.; Jain, N. Vogt-Koyanagi-Harada Disease Associated with Hepatitis B Vaccination. Ocul. Immunol. Inflamm. 2019, 27, 524–527. [Google Scholar] [CrossRef]
  32. Khalifa, Y.M.; Monahan, P.M.; Acharya, N.R. Ampiginous Choroiditis Following Quadrivalent Human Papilloma Virus Vaccine. Br. J. Ophthalmol. 2010, 94, 137–139. [Google Scholar] [CrossRef]
  33. Ogino, K.; Kishi, S.; Yoshimura, N. Multiple Evanescent White Dot Syndrome after Human Papillomavirus Vaccination. Case Rep. Ophthalmol. 2014, 5, 38–43. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, Y.-H.; Chang, Y.-H.; Lee, Y.-C. Panuveitis Following Administration of Quadrivalent Human Papillomavirus Vaccine. Tzu Chi Med. J. 2014, 26, 44–46. [Google Scholar] [CrossRef]
  35. Sawai, T.; Shimizu, M.; Sakai, T.; Yachie, A. Tubulointerstitial Nephritis and Uveitis Syndrome Associated with Human Papillomavirus Vaccine. J. Pediatr. Ophthalmol. Strabismus 2016, 53, 190–191. [Google Scholar] [CrossRef] [PubMed]
  36. Hector, R.E. Acute Posterior Multifocal Placoid Pigment Epitheliopathy. Am. J. Ophthalmol. 1978, 86, 424–425. [Google Scholar] [CrossRef]
  37. Blumberg, S.; Bienfang, D.; Kantrowitz, F.G. A Possible Association Between Influenza Vaccination and Small-Vessel Vasculitis. Arch. Intern. Med. 1980, 140, 847–848. [Google Scholar] [CrossRef]
  38. Blanche, P.; Decrette, C.; Sicard, D. Development of Uveitis Following Vaccination for Influenza. Clin. Infect. Dis. 1994, 19, 979. [Google Scholar] [CrossRef]
  39. Gallagher, M.J.; Yilmaz, T.; Foster, C.S. Vogt-Koyanagi-Harada Syndrome Associated with Bilateral Serous Macular Detachments Responsive to Immunomodulatory Therapy. Ophthalmic Surg. Lasers Imaging Retin. 2009, 40, 345–347. [Google Scholar] [CrossRef]
  40. Wells, M.B.; Garg, S. Bilateral panuveitis after influenza vaccination. Retin. Cases Brief Rep. 2009, 3, 386. [Google Scholar] [CrossRef]
  41. Mendrinos, E.; Baglivo, E. Acute Posterior Multifocal Placoid Pigment Epitheliopathy Following Influenza Vaccination. Eye 2010, 24, 180–181. [Google Scholar] [CrossRef]
  42. Tao, Y.; Chang, L.-B.; Zhao, M.; Li, X.-X. Two Cases of Exudative Retina Detachment and Uveitis Following H1N1 Influenza Vaccination. Chin. Med. J. 2011, 124, 3838–3840. [Google Scholar] [PubMed]
  43. Rothova, A.; de Groot, J.D.F.; Mudrikova, T. Reactivation of Acute Retinal Necrosis after Flu H1N1 Vaccination. Br. J. Ophthalmol. 2011, 95, 291. [Google Scholar] [CrossRef] [PubMed]
  44. Goyal, S.; Nazarian, S.M.; Thayi, D.R.; Hammond, F.; Petrovic, V. Multiple Evanescent White Dot Syndrome Following Recent Influenza Vaccination. Can. J. Ophthalmol. 2013, 48, e115–e116. [Google Scholar] [CrossRef] [PubMed]
  45. Williams, G.S.; Evans, S.; Yeo, D.; Al-bermani, A. Retinal Artery Vasculitis Secondary to Administration of Influenza Vaccine. BMJ Case Rep. 2015, 2015, bcr2015211971. [Google Scholar] [CrossRef]
  46. Gonome, T.; Suzuki, Y.; Metoki, T.; Takahashi, S.; Nakazawa, M. Acute Posterior Multifocal Placoid Pigment Epitheliopathy and Granulomatous Uveitis Following Influenza Vaccination. Am. J. Ophthalmol. Case Rep. 2016, 4, 60–63. [Google Scholar] [CrossRef]
  47. Branisteanu, D.; Bilha, A. Acute posterior multifocal placoid pigment epitheliopathy following influenza vaccination. Rom. J. Ophthalmol. 2015, 59, 52–58. [Google Scholar]
  48. Manusow, J.S.; Rai, A.; Yeh, S.; Mandelcorn, E.D. Two Cases of Panuveitis with Orbital Inflammatory Syndrome after Influenza Vaccination. Can. J. Ophthalmol. 2015, 50, e71–e74. [Google Scholar] [CrossRef]
  49. Kim, M. Vogt-Koyanagi-Harada Syndrome Following Influenza Vaccination. Indian J. Ophthalmol. 2016, 64, 98. [Google Scholar] [CrossRef]
  50. Abou-Samra, A.; Tarabishy, A.B. Multiple Evanescent White Dot Syndrome Following Intradermal Influenza Vaccination. Ocul. Immunol. Inflamm. 2019, 27, 528–530. [Google Scholar] [CrossRef]
  51. Murtaza, F.; Pereira, A.; Mandelcorn, M.S.; Kaplan, A.J. Vogt-Koyanagi-Harada Disease Following Influenza Vaccination. Am. J. Ophthalmol. Case Rep. 2022, 26, 101516. [Google Scholar] [CrossRef]
  52. Islam, S.M.M.; El-Sheikh, H.F.; Tabbara, K.F. Anterior Uveitis Following Combined Vaccination for Measles, Mumps and Rubella (MMR): A Report of Two Cases. Acta Ophthalmol. Scand. 2000, 78, 590–592. [Google Scholar] [CrossRef]
  53. Sedaghat, M.; Zarei-Ghanavati, S.; Shokoohi, S.; Ghasemi, A. Panuveitis and Dermal Vasculitis Following MMR Vaccination. East. Mediterr. Health J. 2007, 13, 470–474. [Google Scholar]
  54. Ferrini, W.; Aubert, V.; Balmer, A.; Munier, F.L.; Abouzeid, H. Anterior Uveitis and Cataract after Rubella Vaccination: A Case Report of a 12-Month-Old Girl. Pediatrics 2013, 132, e1035–e1038. [Google Scholar] [CrossRef]
  55. Esmaeli-Gutstein, B.; Winkelman, J.Z. Uveitis Associated with Varicella Virus Vaccine. Am. J. Ophthalmol. 1999, 127, 733–734. [Google Scholar] [CrossRef]
  56. Naseri, A.; Good, W.V.; Cunningham, E.T. Herpes Zoster Virus Sclerokeratitis and Anterior Uveitis in a Child Following Varicella Vaccination. Am. J. Ophthalmol. 2003, 135, 415–417. [Google Scholar] [CrossRef]
  57. Fine, H.F.; Kim, E.; Flynn, T.E.; Gomes, N.L.; Chang, S. Acute Posterior Multifocal Placoid Pigment Epitheliopathy Following Varicella Vaccination. Br. J. Ophthalmol. 2010, 94, 282–283. [Google Scholar] [CrossRef]
  58. Sham, C.W.; Levinson, R.D. Uveitis Exacerbation After Varicella-Zoster Vaccination in an Adult. Arch. Ophthalmol. 2012, 130, 793–794. [Google Scholar] [CrossRef]
  59. Weinlander, E.J.; Wang, A.L.; Jaru-Ampornpan, P.; Altaweel, M.M.; Nork, T.M. Two Cases of Acute Retinal Necrosis Due to Varicella Zoster Despite Prior Shingles Vaccination. Retin. Cases Brief Rep. 2019, 13, 241. [Google Scholar] [CrossRef]
  60. Heydari-Kamjani, M.; Vante, I.; Uppal, P.; Demory Beckler, M.; Kesselman, M.M. Uveitis Sarcoidosis Presumably Initiated After Administration of Shingrix Vaccine. Cureus 2019, 11, e4920. [Google Scholar] [CrossRef]
  61. Chen, R.I.; Deaner, J.D.; Srivastava, S.K.; Lowder, C.Y. Acute Retinal Necrosis Following Recombinant Subunit Varicella-Zoster Virus Vaccine. Am. J. Ophthalmol. Case Rep. 2020, 20, 100962. [Google Scholar] [CrossRef]
  62. Menghini, M.; Raja, V.; Raiter, J.; Balaratnasingam, C.; Constable, I.J. Acute retinal necrosis associated with Herpes Zoster vaccination. Retin. Cases Brief Rep. 2021, 15, 166–168. [Google Scholar] [CrossRef] [PubMed]
  63. Biancardi, A.L.; Moraes, H.V. de Anterior and Intermediate Uveitis Following Yellow Fever Vaccination with Fractional Dose: Case Reports. Ocul. Immunol. Inflamm. 2019, 27, 521–523. [Google Scholar] [CrossRef] [PubMed]
  64. Volkov, L.; Grard, G.; Bollaert, P.-E.; Durand, G.A.; Cravoisy, A.; Conrad, M.; Nace, L.; Courte, G.; Marnai, R.; Leparc-Goffart, I.; et al. Viscerotropic Disease and Acute Uveitis Following Yellow Fever Vaccination: A Case Report. BMC Infect. Dis. 2020, 20, 116. [Google Scholar] [CrossRef] [PubMed]
  65. Campos, W.R.; Cenachi, S.P.F.; Soares, M.S.; Gonçalves, P.F.; Vasconcelos-Santos, D.V. Vogt-Koyanagi-Harada-like Disease Following Yellow Fever Vaccination. Ocul. Immunol. Inflamm. 2021, 29, 124–127. [Google Scholar] [CrossRef]
  66. Pereima, R.R.; Bonatti, R.; Crotti, F.; Furtado, J.M.; Lopes, M.H.; Yamamoto, J.H.; Kreuz, A.C. Ocular Adverse Events Following Yellow Fever Vaccination: A Case Series. Ocul. Immunol. Inflamm. 2022, 30, 1425–1429. [Google Scholar] [CrossRef]
  67. Fine, L.; Fine, A.; Cunningham, E.T. Multiple Evanescent White Dot Syndrome Following Hepatitis a Vaccination. Arch. Ophthalmol. 2001, 119, 1856–1858. [Google Scholar] [CrossRef]
  68. Yang, J.; Chen, C.; Hu, Y.; Zeng, R. Multiple Evanescent White Dot Syndrome Following Rabies Vaccination: A Case Report. BMC Ophthalmol. 2018, 18, 312. [Google Scholar] [CrossRef]
  69. Stangos, A.; Zaninetti, M.; Petropoulos, I.; Baglivo, E.; Pournaras, C. Multiple Evanescent White Dot Syndrome Following Simultaneous Hepatitis-A and Yellow Fever Vaccination. Ocul. Immunol. Inflamm. 2006, 14, 301–304. [Google Scholar] [CrossRef]
  70. Cohen, S.M. Multiple Evanescent White Dot Syndrome After Vaccination for Human Papilloma Virus and Meningococcus. J. Pediatr. Ophthalmol. Strabismus 2010, 47, 1–3. [Google Scholar] [CrossRef]
  71. Escott, S.; Tarabishy, A.B.; Davidorf, F.H. Multifocal Choroiditis Following Simultaneous Hepatitis A, Typhoid, and Yellow Fever Vaccination. Clin. Ophthalmol. 2013, 7, 363–365. [Google Scholar] [CrossRef]
  72. Schiffman, M.; Castle, P.E.; Jeronimo, J.; Rodriguez, A.C.; Wacholder, S. Human Papillomavirus and Cervical Cancer. Lancet 2007, 370, 890–907. [Google Scholar] [CrossRef]
  73. Schiffman, M.; Doorbar, J.; Wentzensen, N.; de Sanjosé, S.; Fakhry, C.; Monk, B.J.; Stanley, M.A.; Franceschi, S. Carcinogenic Human Papillomavirus Infection. Nat. Rev. Dis. Prim. 2016, 2, 16086. [Google Scholar] [CrossRef] [PubMed]
  74. WHO. Human Papillomavirus Vaccines: WHO Position Paper, December 2022. Available online: https://www.who.int/publications-detail-redirect/who-wer9750-645-672 (accessed on 28 July 2023).
  75. WHO. Immunization Coverage. Available online: https://www.who.int/news-room/fact-sheets/detail/immunization-coverage (accessed on 18 October 2023).
  76. WHO. List of Prequalified Vaccines. Available online: https://extranet.who.int/pqweb/vaccines/list-prequalified-vaccines (accessed on 28 July 2023).
  77. Slade, B.A.; Leidel, L.; Vellozzi, C.; Woo, E.J.; Hua, W.; Sutherland, A.; Izurieta, H.S.; Ball, R.; Miller, N.; Braun, M.M.; et al. Postlicensure Safety Surveillance for Quadrivalent Human Papillomavirus Recombinant Vaccine. JAMA 2009, 302, 750–757. [Google Scholar] [CrossRef] [PubMed]
  78. Shi, T.; Lv, W.; Zhang, L.; Chen, J.; Chen, H. Association of HLA-DR4/HLA-DRB1*04 with Vogt-Koyanagi-Harada Disease: A Systematic Review and Meta-Analysis. Sci. Rep. 2014, 4, 6887. [Google Scholar] [CrossRef] [PubMed]
  79. Attia, S.; Khochtali, S.; Kahloun, R.; Ammous, D.; Jelliti, B.; Ben Yahia, S.; Zaouali, S.; Khairallah, M. Clinical and Multimodal Imaging Characteristics of Acute Vogt–Koyanagi–Harada Disease Unassociated with Clinically Evident Exudative Retinal Detachment. Int. Ophthalmol. 2016, 36, 37–44. [Google Scholar] [CrossRef] [PubMed]
  80. Mackensen, F.; Billing, H. Tubulointerstitial Nephritis and Uveitis Syndrome. Curr. Opin. Ophthalmol. 2009, 20, 525. [Google Scholar] [CrossRef]
  81. Edwards, I.R.; Biriell, C. Harmonisation in Pharmacovigilance. Drug Saf. 1994, 10, 93–102. [Google Scholar] [CrossRef]
  82. Natale, C.; Giannini, T.; Lucchese, A.; Kanduc, D. Computer-Assisted Analysis of Molecular Mimicry between Human Papillomavirus 16 E7 Oncoprotein and Human Protein Sequences. Immunol. Cell Biol. 2000, 78, 580–585. [Google Scholar] [CrossRef]
  83. Sonneveld, M.J.; Zoutendijk, R.; Janssen, H.L.A. Hepatitis B Surface Antigen Monitoring and Management of Chronic Hepatitis B. J. Viral Hepat. 2011, 18, 449–457. [Google Scholar] [CrossRef]
  84. Hadziyannis, S.J.; Papatheodoridis, G.V. Hepatitis B e Antigen-Negative Chronic Hepatitis B: Natural History and Treatment. Semin. Liver Dis. 2006, 26, 130–141. [Google Scholar] [CrossRef]
  85. Ocama, P.; Opio, C.K.; Lee, W.M. Hepatitis B Virus Infection: Current Status. Am. J. Med. 2005, 118, 1413. [Google Scholar] [CrossRef] [PubMed]
  86. Shepard, C.W.; Simard, E.P.; Finelli, L.; Fiore, A.E.; Bell, B.P. Hepatitis B Virus Infection: Epidemiology and Vaccination. Epidemiol. Rev. 2006, 28, 112–125. [Google Scholar] [CrossRef] [PubMed]
  87. Benage, M.; Fraunfelder, F.W. Vaccine-Associated Uveitis. Mo. Med. 2016, 113, 48–52. [Google Scholar] [PubMed]
  88. Grob, P.J.; Martenet, A.C.; Witmer, R. Nonspecific Immune Parameters and Hepatitis B Antigens in Patients with Uveitis. Mod. Probl. Ophthalmol. 1976, 16, 254–258. [Google Scholar] [PubMed]
  89. Murray, P.I.; Prasad, J.; Rahi, A.H. Status of Hepatitis B Virus in the Aetiology of Uveitis in Great Britain. Br. J. Ophthalmol. 1983, 67, 685–687. [Google Scholar] [CrossRef] [PubMed]
  90. Aguirre, G.; Carmichael, L.; Bistner, S. Corneal Endothelium in Viral Induced Anterior Uveitis. Ultrastructural Changes Following Canine Adenovirus Type 1 Infection. Arch. Ophthalmol. 1975, 93, 219–224. [Google Scholar] [CrossRef]
  91. London, W.T. Hepatitis B Virus and Antigen-Antibody Complex Diseases. N. Engl. J. Med. 1977, 296, 1528–1529. [Google Scholar] [CrossRef]
  92. Farthing, C.F.; Howard, R.S.; Thin, R.N. Papillitis and Hepatitis B. Br. Med. J. Clin. Res. Ed. 1986, 292, 1712. [Google Scholar] [CrossRef]
  93. Yokochi, T.; Fujii, Y.; Nakashima, I.; Asai, J.; Kiuchi, M.; Kojima, K.; Kato, N. A Murine Model of Experimental Autoimmune Lens-Induced Uveitis Using Klebsiella O3 Lipopolysaccharide as a Potent Immunological Adjuvant. Int. J. Exp. Pathol. 1993, 74, 573–582. [Google Scholar]
  94. Geier, D.A.; Geier, M.R. A Case-Control Study of Serious Autoimmune Adverse Events Following Hepatitis B Immunization. Autoimmunity 2005, 38, 295–301. [Google Scholar] [CrossRef]
  95. Verstraeten, T.; Descamps, D.; David, M.-P.; Zahaf, T.; Hardt, K.; Izurieta, P.; Dubin, G.; Breuer, T. Analysis of Adverse Events of Potential Autoimmune Aetiology in a Large Integrated Safety Database of AS04 Adjuvanted Vaccines. Vaccine 2008, 26, 6630–6638. [Google Scholar] [CrossRef]
  96. Altman, A.; Szyper-Kravitz, M.; Shoenfeld, Y. HBV Vaccine and Dermatomyositis: Is There an Association? Rheumatol. Int. 2008, 28, 609–612. [Google Scholar] [CrossRef] [PubMed]
  97. Mikaeloff, Y.; Caridade, G.; Rossier, M.; Suissa, S.; Tardieu, M. Hepatitis B Vaccination and the Risk of Childhood-Onset Multiple Sclerosis. Arch. Pediatr. Adolesc. Med. 2007, 161, 1176–1182. [Google Scholar] [CrossRef] [PubMed]
  98. Mikaeloff, Y.; Caridade, G.; Suissa, S.; Tardieu, M. Hepatitis B Vaccine and the Risk of CNS Inflammatory Demyelination in Childhood. Neurology 2009, 72, 873–880. [Google Scholar] [CrossRef] [PubMed]
  99. Centers for Disease Control. Influenza Vaccine: Recommendation of the Public Health Service Advisory Committee on Immunization Practices. Ann. Intern. Med. 1978, 89, 657–659. [Google Scholar] [CrossRef]
  100. World Health Organization. Influenza (Seasonal). Available online: https://www.who.int/news-room/fact-sheets/detail/influenza-(seasonal) (accessed on 31 July 2023).
  101. World Health Organization. Influenza Vaccination Coverage and Effectiveness. Available online: https://www.who.int/europe/news-room/fact-sheets/item/influenza-vaccination-coverage-and-effectiveness (accessed on 18 October 2023).
  102. Grohskopf, L.A.; Alyanak, E.; Ferdinands, J.M.; Broder, K.R.; Blanton, L.H.; Talbot, H.K.; Fry, A.M. Prevention and Control of Seasonal Influenza with Vaccines: Recommendations of the Advisory Committee on Immunization Practices, United States, 2021–2022 Influenza Season. MMWR Recomm. Rep. 2021, 70, 1–28. [Google Scholar] [CrossRef]
  103. Thurairajan, G.; Hope-Ross, M.W.; Situnayake, R.D.; Murray, P.I. Polyarthropathy, Orbital Myositis and Posterior Scleritis: An Unusual Adverse Reaction to Influenza Vaccine. Br. J. Rheumatol. 1997, 36, 120–123. [Google Scholar] [CrossRef]
  104. Solomon, A.; Siganos, C.S.; Frucht-Pery, J. Adverse Ocular Effects Following Influenza Vaccination. Eye 1999, 13, 381–382. [Google Scholar] [CrossRef]
  105. Skowronski, D.M.; Strauss, B.; De Serres, G.; MacDonald, D.; Marion, S.A.; Naus, M.; Patrick, D.M.; Kendall, P. Oculo-Respiratory Syndrome: A New Influenza Vaccine-Associated Adverse Event? Clin. Infect. Dis. 2003, 36, 705–713. [Google Scholar] [CrossRef]
  106. Walter, R.; Hartmann, K.; Fleisch, F.; Reinhart, W.H.; Kuhn, M. Reactivation of Herpesvirus Infections after Vaccinations? Lancet 1999, 353, 810. [Google Scholar] [CrossRef]
  107. Watad, A.; Quaresma, M.; Brown, S.; Cohen Tervaert, J.W.; Rodríguez-Pint, I.; Cervera, R.; Perricone, C.; Shoenfeld, Y. Autoimmune/Inflammatory Syndrome Induced by Adjuvants (Shoenfeld’s Syndrome)—An Update. Lupus 2017, 26, 675–681. [Google Scholar] [CrossRef] [PubMed]
  108. Cunningham, E.T.; Moorthy, R.S. Vaccine-Associated Posterior Uveitis. Retina 2020, 40, 595–598. [Google Scholar] [CrossRef] [PubMed]
  109. Rec, W.E. Rubella Vaccines: WHO Position Paper. Wkly Epidemiol. Rec. 2000, 86, 301–316. [Google Scholar]
  110. Birnbaum, A.D.; Tessler, H.H.; Schultz, K.L.; Farber, M.D.; Gao, W.; Lin, P.; Oh, F.; Goldstein, D.A. Epidemiologic Relationship between Fuchs Heterochromic Iridocyclitis and the United States Rubella Vaccination Program. Am. J. Ophthalmol. 2007, 144, 424–428. [Google Scholar] [CrossRef]
  111. De Groot-Mijnes, J.D.F.; de Visser, L.; Zuurveen, S.; Martinus, R.A.; Völker, R.; ten Dam-van Loon, N.H.; de Boer, J.H.; Postma, G.; de Groot, R.J.; van Loon, A.M.; et al. Identification of New Pathogens in the Intraocular Fluid of Patients with Uveitis. Am. J. Ophthalmol. 2010, 150, 628–636. [Google Scholar] [CrossRef]
  112. Quentin, C.D.; Reiber, H. Fuchs Heterochromic Cyclitis: Rubella Virus Antibodies and Genome in Aqueous Humor. Am. J. Ophthalmol. 2004, 138, 46–54. [Google Scholar] [CrossRef]
  113. Groen-Hakan, F.; van de Laar, S.; van der Eijk-Baltissen, A.A.; van Loon, N.T.D.; de Boer, J.; Rothova, A. Clinical Manifestations, Prognosis, and Vaccination Status of Patients with Rubella Virus–Associated Uveitis. Am. J. Ophthalmol. 2019, 202, 37–46. [Google Scholar] [CrossRef]
  114. Johnson, R.W.; Wasner, G.; Saddier, P.; Baron, R. Herpes Zoster and Postherpetic Neuralgia: Optimizing Management in the Elderly Patient. Drugs Aging 2008, 25, 991–1006. [Google Scholar] [CrossRef]
  115. Johnson, R.W.; Rice, A.S.C. Pain Following Herpes Zoster: The Influence of Changing Population Characteristics and Medical Developments. Pain 2007, 128, 3–5. [Google Scholar] [CrossRef]
  116. Sanford, M.; Keating, G.M. Zoster Vaccine (Zostavax): A Review of Its Use in Preventing Herpes Zoster and Postherpetic Neuralgia in Older Adults. Drugs Aging 2010, 27, 159–176. [Google Scholar] [CrossRef]
  117. Hesse, E.M. Postlicensure Safety Surveillance of Recombinant Zoster Vaccine (Shingrix)—United States, October 2017–June 2018. MMWR Morb. Mortal. Wkly. Rep. 2019, 68, 91–94. [Google Scholar] [CrossRef]
  118. Bharucha, T.; Ming, D.; Breuer, J. A Critical Appraisal of ‘Shingrix’, a Novel Herpes Zoster Subunit Vaccine (HZ/Su or GSK1437173A) for Varicella Zoster Virus. Hum. Vaccines Immunother. 2017, 13, 1789–1797. [Google Scholar] [CrossRef] [PubMed]
  119. Schöberl, F.; Csanadi, E.; Eren, O.; Dieterich, M.; Kümpfel, T. NMOSD Triggered by Yellow Fever Vaccination—An Unusual Clinical Presentation with Segmental Painful Erythema. Mult. Scler. Relat. Disord. 2017, 11, 43–44. [Google Scholar] [CrossRef] [PubMed]
  120. Hedges, T.R.; Sinclair, S.H.; Gragoudas, E.S. Evidence for Vasculitis in Acute Posterior Multifocal Placoid Pigment Epitheliopathy. Ann. Ophthalmol. 1979, 11, 539–542. [Google Scholar] [PubMed]
  121. Wong, M.; Campos-Baniak, M.G.; Colleaux, K. Acute Idiopathic Blind Spot Enlargement Syndrome Following Measles, Mumps and Rubella Vaccination. Can. J. Ophthalmol. 2019, 54, e199–e203. [Google Scholar] [CrossRef] [PubMed]
  122. Abu-Yaghi, N.E.; Hartono, S.P.; Hodge, D.O.; Pulido, J.S.; Bakri, S.J. White Dot Syndromes: A 20-Year Study of Incidence, Clinical Features and Outcomes. Ocul. Immunol. Inflamm. 2011, 19, 426–430. [Google Scholar] [CrossRef]
  123. Dhaliwal, R.S.; Maguire, A.M.; Flower, R.W.; Arribas, N.P. Acute Posterior Multifocal Placoid Pigment Epitheliopathy. An Indocyanine Green Angiographic Study. Retina 1993, 13, 317–325. [Google Scholar] [CrossRef]
  124. Jones, N.P. Acute Posterior Multifocal Placoid Pigment Epitheliopathy. Br. J. Ophthalmol. 1995, 79, 384–389. [Google Scholar] [CrossRef]
  125. Du, L.; Kijlstra, A.; Yang, P. Vogt-Koyanagi-Harada Disease: Novel Insights into Pathophysiology, Diagnosis and Treatment. Prog. Retin. Eye Res. 2016, 52, 84–111. [Google Scholar] [CrossRef]
  126. Imai, Y.; Sugita, M.; Nakamura, S.; Toriyama, S.; Ohno, S. Cytokine Production and Helper T Cell Subsets in Vogt-Koyanagi-Harada’s Disease. Curr. Eye Res. 2001, 22, 312–318. [Google Scholar] [CrossRef]
  127. Holland, G.N. Standard Diagnostic Criteria for the Acute Retinal Necrosis Syndrome. Executive Committee of the American Uveitis Society. Am. J. Ophthalmol. 1994, 117, 663–667. [Google Scholar] [CrossRef] [PubMed]
  128. Hillenkamp, J.; Nölle, B.; Bruns, C.; Rautenberg, P.; Fickenscher, H.; Roider, J. Acute Retinal Necrosis: Clinical Features, Early Vitrectomy, and Outcomes. Ophthalmology 2009, 116, 1971–1975.e2. [Google Scholar] [CrossRef] [PubMed]
  129. Young, N.J.; Bird, A.C. Bilateral Acute Retinal Necrosis. Br. J. Ophthalmol. 1978, 62, 581–590. [Google Scholar] [CrossRef]
  130. Palay, D.A.; Sternberg, P.; Davis, J.; Lewis, H.; Holland, G.N.; Mieler, W.F.; Jabs, D.A.; Drews, C. Decrease in the Risk of Bilateral Acute Retinal Necrosis by Acyclovir Therapy. Am. J. Ophthalmol. 1991, 112, 250–255. [Google Scholar] [CrossRef] [PubMed]
  131. Cheng, J.Y.; Margo, C.E. Ocular Adverse Events Following Vaccination: Overview and Update. Surv. Ophthalmol. 2022, 67, 293–306. [Google Scholar] [CrossRef]
  132. Scalabrin, S.; Becco, A.; Vitale, A.; Nuzzi, R. Ocular Effects Caused by Viral Infections and Corresponding Vaccines: An Overview of Varicella Zoster Virus, Measles Virus, Influenza Viruses, Hepatitis B Virus, and SARS-CoV-2. Front. Med. 2022, 9, 999251. [Google Scholar] [CrossRef] [PubMed]
  133. Ness, T.; Hengel, H. Impfschäden am Auge. Ophthalmologe 2016, 113, 615–622. [Google Scholar] [CrossRef]
  134. Cunningham, E.T.; Moorthy, R.S.; Fraunfelder, F.W.; Zierhut, M. Vaccine-Associated Uveitis. Ocul. Immunol. Inflamm. 2019, 27, 517–520. [Google Scholar] [CrossRef]
  135. Cordero-Coma, M.; Salazar-Méndez, R.; Garzo-García, I.; Yilmaz, T. Drug-Induced Uveitis. Expert Opin. Drug Saf. 2015, 14, 111–126. [Google Scholar] [CrossRef]
  136. Moorthy, R.S.; Moorthy, M.S.; Cunningham, E.T.J. Drug-Induced Uveitis. Curr. Opin. Ophthalmol. 2018, 29, 588. [Google Scholar] [CrossRef]
  137. Cunningham, E.T.; London, N.J.S.; Moorthy, R.; Garg, S.J.; Zierhut, M. Drugs, Inflammation, and the Eye. Ocul. Immunol. Inflamm. 2016, 24, 125–127. [Google Scholar] [CrossRef] [PubMed]
  138. Fraunfelder, F.W.; Suhler, E.B.; Fraunfelder, F.T. Hepatitis B Vaccine and Uveitis: An Emerging Hypothesis Suggested by Review of 32 Case Reports. Cutan. Ocul. Toxicol. 2010, 29, 26–29. [Google Scholar] [CrossRef] [PubMed]
  139. Holt, H.D.; Hinkle, D.M.; Falk, N.S.; Fraunfelder, F.T.; Fraunfelder, F.W. Human Papilloma Virus Vaccine Associated Uveitis. Curr. Drug Saf. 2014, 9, 65–68. [Google Scholar] [CrossRef]
  140. Fiore, T.; Iaccheri, B.; Androudi, S.; Papadaki, T.G.; Anzaar, F.; Brazitikos, P.; D’Amico, D.J.; Foster, C.S. Acute Posterior Multifocal Placoid Pigment Epitheliopathy: Outcome and Visual Prognosis. Retina 2009, 29, 994–1001. [Google Scholar] [CrossRef] [PubMed]
  141. Lavezzo, M.M.; Sakata, V.M.; Morita, C.; Rodriguez, E.E.C.; Abdallah, S.F.; da Silva, F.T.G.; Hirata, C.E.; Yamamoto, J.H. Vogt-Koyanagi-Harada Disease: Review of a Rare Autoimmune Disease Targeting Antigens of Melanocytes. Orphanet J. Rare Dis. 2016, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  142. Cochrane, T.F.; Silvestri, G.; McDowell, C.; Foot, B.; McAvoy, C.E. Acute Retinal Necrosis in the United Kingdom: Results of a Prospective Surveillance Study. Eye 2012, 26, 370–377; quiz 378. [Google Scholar] [CrossRef]
  143. Gherardi, R.K.; Crépeaux, G.; Authier, F.-J. Myalgia and Chronic Fatigue Syndrome Following Immunization: Macrophagic Myofasciitis and Animal Studies Support Linkage to Aluminum Adjuvant Persistency and Diffusion in the Immune System. Autoimmun. Rev. 2019, 18, 691–705. [Google Scholar] [CrossRef]
  144. Read, R.W. Uveitis: Advances in Understanding of Pathogenesis and Treatment. Curr. Rheumatol. Rep. 2006, 8, 260–266. [Google Scholar] [CrossRef]
  145. Waisbren, B.A. Acquired Autoimmunity after Viral Vaccination Is Caused by Molecular Mimicry and Antigen Complimentarity in the Presence of an Immunologic Adjuvant and Specific HLA Patterns. Med. Hypotheses 2008, 70, 346–348. [Google Scholar] [CrossRef]
  146. Broekhuyse, R.M.; Kuhlmann, E.D.; Winkens, H.J. Experimental Autoimmune Anterior Uveitis (EAAU). III. Induction by Immunization with Purified Uveal and Skin Melanins. Exp. Eye Res. 1993, 56, 575–583. [Google Scholar] [CrossRef]
Figure 1. Fundus images of a patient with multiple evanescent white dot syndrome after human papillomavirus vaccination. FA revealed leakage from the mid-peripheral vasculature at 7 months (A), FA was performed again and revealed a marked increase in vascular leakage at 1.5 years (B), 1 week after steroid pulse therapy, FA showed a drastic reduction in leakage from the vasculature (C), FA revealed recurrent leakage after 2 years (D) (adapted with permission from [33] 2014, Ogino et al.).
Figure 1. Fundus images of a patient with multiple evanescent white dot syndrome after human papillomavirus vaccination. FA revealed leakage from the mid-peripheral vasculature at 7 months (A), FA was performed again and revealed a marked increase in vascular leakage at 1.5 years (B), 1 week after steroid pulse therapy, FA showed a drastic reduction in leakage from the vasculature (C), FA revealed recurrent leakage after 2 years (D) (adapted with permission from [33] 2014, Ogino et al.).
Vaccines 11 01626 g001
Figure 2. Fundus images of a patient with uveitis following influenza vaccination. Multiple white dots (black arrow) at the level of outer retina are observed in patient with MEWDS (A). A whitish spot (white arrow) found in optical coherence tomography in patient with APMPPE (B). ((A) Adapted with permission from [44] 2013, Goyal et al.; (B) Adapted with permission from [46] 2016, Gonome et al.).
Figure 2. Fundus images of a patient with uveitis following influenza vaccination. Multiple white dots (black arrow) at the level of outer retina are observed in patient with MEWDS (A). A whitish spot (white arrow) found in optical coherence tomography in patient with APMPPE (B). ((A) Adapted with permission from [44] 2013, Goyal et al.; (B) Adapted with permission from [46] 2016, Gonome et al.).
Vaccines 11 01626 g002
Figure 3. Fundus images of a patient with reactivation of previously controlled uveitis after receiving RZV. Colored fundus photographs of right (A) and left (B) eyes with chorioretinal scarring due to prior inflammation before RZV vaccination. Photos of stability of the right eye (C) and a new active inflammatory lesion in the temporal macula of the left eye after RZV vaccination ((D), white arrow). (Adapted with permission from Ref. [27] 2021, Richards et al.).
Figure 3. Fundus images of a patient with reactivation of previously controlled uveitis after receiving RZV. Colored fundus photographs of right (A) and left (B) eyes with chorioretinal scarring due to prior inflammation before RZV vaccination. Photos of stability of the right eye (C) and a new active inflammatory lesion in the temporal macula of the left eye after RZV vaccination ((D), white arrow). (Adapted with permission from Ref. [27] 2021, Richards et al.).
Vaccines 11 01626 g003
Table 1. Case reports of uveitis following vaccination.
Table 1. Case reports of uveitis following vaccination.
ReferenceDiagnosisVaccine TypeAge */GenderSymptoms/Signs/Lab Tests/Medical HistoryInterval Post-Vaccination ^TreatmentOutcome/Follow-Up
Hepatitis B virus (HBV) vaccine
Fried 1987 [28]Bilateral posterior uveitisRecombinant20/FHeadache1st: 3 days after the second dose
2nd: 4 days after the third dose
NRRecurrence due to re-exposure/not reported
Brézin 1995 [29]Bilateral APMPPERecombinant31/MNone3 days after the fourth doseNRA residual paracentral scotoma/9 m
Bilateral APMPPERecombinant30/MNone14 days after the third doseNRA residual paracentral scotoma OS 4 m
Baglivo 1996 [30]Bilateral MEWDSRecombinant23/FNone24 h after the third doseNRNone/3 m
Sood 2019 [31]Bilateral VKH (pan-uveitis)Recombinant43/MHearing loss, tinnitus, integumentary changes.3 days after the first dosepo/io steroidsVKH with long-term steroids treatment/5 m
Human papillomavirus (HPV) vaccine
Khalifa 2010 [32]Bilateral ampiginous choroiditisQuadrivalent/recombinant17/FNone3 weeks after the first dosepo steroidsExtensive macular scarring remained/3 m
Ogino 2014 [33]MEWDS (left eye)Bivalent/recombinant16/FThroat pain, headache2 weeks after the second doseiv steroids and anti-histamineFA revealed recurrent leakage/2 y
Chen, Y.-H. 2014 [34]Bilateral pan-uveitisQuadrivalent/recombinant27/FBilateral knee pain, erythematous papules vertigo, and hearing impairment4 days after the third doseio/po steroidsNone/2 y
Dansingani 2015 [23]Bilateral pan-uveitis and ERD resembling VKHQuadrivalent/recombinant20/FHLA-DRB1*0405(+)3 weeks after the second dosepo steroidsNone/5 m
Sawai 2016 [35]Bilateral anterior uveitis/TINU Recombinant14/FFever, general malaise, low back pain4 days after the first doseio/gtt steroidlong-term steroids treatment for uveitis/3 y
Bilateral anterior uveitis/TINU Recombinant14/FNR10 weeks after the third doseio/gtt steroidNone/NR
Ye, H. 2020 [22]Bilateral posterior uveitis resembling Harada diseaseDivalent/recombinant29/FHLA-DR04 and 07(+)7 days after the third doseio/po steroidsNone/4 m
Kong 2022 [21]Bilateral Harada disease-like uveitis(posterior)Quadrivalent/recombinant37/FNone10 days after the third dosegtt steroidsNone/3 m
Influenza virus vaccine
Hector 1978 [36]Bilateral APMPPELive attenuated21/MFever, chills, headache48 h after the first doseNoneResidual RPE changes/6 m
Blumberg 1980 [37]Iritis (OD) and optic neuritis (OS)Inactivated27/MFever, arthralgias, and myalgias14 days after vaccinationSystematic/gtt steroidsC3: 91 mg/dL; ESR: 40 mm/h/1 m
Blanche 1994 [38]Bilateral anterior and posterior uveitisInactivated68/FFever48 h after the first dosegtt steroidsNone/3 m
Gallagher 2009 [39]Bilateral VKH (pan-uveitis)NR44/FTinnitus1 month after vaccinationiv/po steroids with steroids-sparing and long-term immunomodulationNone/NR
Wells 2009 [40]Bilateral pan-uveitisNR70/MNR1 days after the first doseio/gtt steroidsNone/3 m
Mendrinos 2010 [41]Bilateral APMPPENR27/MFlu-like symptoms14 days after the vaccinationpo steroidsResidual RPE changes/3 m
Tao 2011 [42]Posterior uveitis and ERD (right)Live attenuated (H1N1)10/MNone10 days after vaccinationiv/po steroidsNone/1 m
Bilateral posterior uveitis and ERDLive attenuated (H1N1)47/FHigh fever, bilateral headache2 days after vaccinationiv/po steroidsVisual acuity: 0.01/NR
Rothova 2011 [43]Bilateral VZV-associated pan-uveitisLive attenuated (H1N1)60/MNR4 days after vaccinationsystematic/io steroidsThe intraocular inflammation slowly subsided/NR
Goyal 2013 [46]MEWDS (right eye)NR53/MNR10 days after vaccinationNoneA paracentral scotoma/1 m
Williams 2015 [45]Retinal artery vasculitis (right eye; anterior uveitis)Live attenuated78/FRight-sided headache8 weeks after vaccinationgtt steroidsNone/9 m
Branisteanu 2015 [47]Bilateral APMPEENR18/FIntermittent headaches14 days after vaccinationpo steroidsResidual RPE changes/5 y
Manusow 2015 [48]Bilateral pan-uveitis with OISLive attenuated49/FPolyarthritis, fever, tender cervical lymphadenopathy4 days after vaccinationiv/po steroidsNo light perception OS/1 y
Pan-uveitis with OIS (right)Live attenuated57/MMild jaundice, confusion and disorientation to place and time/ESR-84(+)3 days after vaccinationiv/po steroidsLP, 20/50/3 m
Gonome 2016 [46]Bilateral AMPEE and granulomatous uveitis (pan-uveitis)NR30/FFever, cough, and nausea17 days after vaccinationInitiate iv/gtt NSAIDs; After the granulomatous uveitis appearance; gtt steroidsNone/1 m
Kim 2016 [49]Bilateral VKH (pan-uveitis)Live attenuated52/FTinnitus1 month after vaccinationiv/po steroidNone/NR
Abou-Samra 2019 [50]MEWDS (right eye)NR27/FFever, rash, oral ulcers, arthralgias, headache, or vertigo.14 days after vaccinationNoneResidual RPE changes/8 w
Murtaza 2022 [51]Bilateral VKH (pan-uveitis)Inactivated30/MHeadache, tinnitus, and HLA-DR4(+)2 days after vaccinationio/po steroidsSunset glow fundus from choroidal depigmentation/6 m
Measles–Mumps–Rubella (MMR) vaccine
Islam 2000 [52]Bilateral anterior uveitisLive attenuated12/FNR6 weeks after vaccinationgtt steroidsNone/1 y
Bilateral anterior uveitisLive attenuated14/MNR4 weeks after vaccinationpo/gtt steroidsNone/NR
Sedaghat 2007 [53]Bilateral pan-uveitis and dermal vasculitisLive attenuated17/FFever, chills, skin rash and knee arthritis5 days after vaccinationpo/gtt steroidsNone/6 m
Ferrini 2013 [54]Anterior uveitis (left eye) with iris heterochromia and cataractLive attenuated12
month/F
HLA-B51, rubella IgG (+)3 months after vaccinationSystematic/gtt/io steroids cataract extractionNone/3 m
Varicella zoster virus (VZV) vaccine
Esmaeli-Gutstein 1999 [55]Anterior and intermediate uveitis (left eye)Live attenuated16/FGeneralized vesicular rash1 week after vaccinationpo acyclovir, gtt steroidsNone/NR
Naseri 2003 [56]Herpes zoster virus sclerokeratitis and anterior uveitis (left eye)Live attenuated9/Mrash in left face, wild-type VZV DNA (+)3 years after vaccinationpo acyclovir/gtt steroidsNR/NR
Fine 2010 [57]Bilateral APMPPELive attenuated11/FSevere headaches and tinnitus/VZV Ab (+)10 days after vaccinationpo steroidsNR/1 y
Charkoudian 2011 [24]ARN (left eye)Live attenuated77/FVZV DNA (+)/diabetes mellitus6 days after vaccinationpo/iv antiviral drugs, vitrectomyNR/NR
Bilateral ARNLive attenuated80/Mrash and fever/VZV DNA (+), immunosuppressant use for renal transplantation2 months after vaccinationpo/iv antiviral drugs, io foscarnet, bilateral vitrectomyNR/NR
Gonzales 2012 [25]Bilateral ARNLive attenuated20/MOka strain VZV DNA (+)/
immunosuppressant for an inflammatory gastroenteropathy
1 month after vaccinationio foscarnet, antiviral drugs, pars plana vitrectomyNR/NR
Sham 2012 [58]Exacerbation of anterior uveitis (right eye)Live attenuated86/MMedical history of HZO with anterior uveitis3 weeks after vaccinationpo Valacyclovir, gtt steroidsNone/NR
Heath 2017 [26]ARN (left eye)Live attenuated78/FOka strain VZV DNA (+)/immunosuppressant for autoimmune diabetes6 weeks after vaccinationpo valaciclovir, gtt steroids, pars plana vitrectomyA pigmented scar/NR
Weinlander 2019 [59]ARN (left eye)Live attenuated64/MWild-type VZV DNA (+)/metabolic syndrome and impaired glucose tolerance16 months after vaccinationpo Valacyclovir, po/gtt steroidsNone/6 m
ARN (left eye)Live attenuated62/MWild-type VZV DNA (+)/Cirrhosis and diabetes mellitus type 27 months after vaccinationpo Valacyclovir, gtt steroidsDied from complications of his cirrhosis/6 m
Heydari-Kamjani 2019 [60]Bilateral uveitis sarcoidosisRecombinant zoster vaccine53/FHeadaches4 days after vaccinationgtt steroidsNone/NR
Chen R.I. 2020 [61]ARN (left eye)Recombinant zoster vaccine65/FImmunomodulator for multiple myeloma/wild-type VZV DNA (+)6 weeks after vaccinationio foscarnet, iv/po antiviral drugsNone/19 w
Menghini 2021 [62]ARN with obliterative angiopathy (left eye)Live attenuated76/MInsulin-dependent diabetes mellitus, chronic lymphocytic leukemia/wild-type VZV DNA (+)2 days after vaccinationio foscarnet, iv/po/iv antiviral drugs, iv/po steroidsLeft eye visual acuity dropped to perception only/NR
Richards 2021 [27]Recurrent bilateral multifocal choroiditisRecombinant zoster vaccine57/Farm swelling at the injection site, chills, malaise, subjective fever, and tinnitus/immunosuppressant for multifocal choroiditis24 h after the first dosepo steroids and continued methotrexateIntravitreal bevacizumab for a secondary choroidal neovascular membrane/2 m
Recurrent bilateral anterior and mild intermediate uveitisRecombinant zoster vaccine69/MHeadache/gtt steroids for uveitis1 month after the second dosepo valacyclovir/gtt steroidsNone/1 m
Recurrent anterior uveitis (left eye)Recombinant zoster vaccine70/Fgtt steroids and po valacyclovir for viral keratouveitis2 weeks after the first dosepo valacyclovir, po/gtt steroidsNone/6 w
Yellow Fever virus vaccine
Biancardi 2019 [63]Anterior uveitis (right eye)Live attenuated35/FNone10 days after vaccinationgtt steroidsNone/NR
Intermediate uveitis (left eye)Live attenuated21/Flow fever, body ache, and mild headache14 days after vaccinationpo steroidsNone/6 w
Volkov 2020 [64]Viscerotropic disease followed by bilateral acute anterior and intermediate uveitisLive attenuated37/MFever, cough, dyspnea, malaise, sore throat, non-bloody diarrhea, and morbilliform skin rash of the chest/YFV RNA (+)2–3 weeks after vaccinationio/gtt steroidsPersistent fatigue for few month/NR
Campos 2021 [65]Bilateral VKH (pan-uveitis)Live attenuated34/Mtinnitus, headache12 days after a booster doseIv/po steroidsNone/2 y
Pereima 2022 [66]Bilateral Acute VKH (pan-uveitis)Live attenuated45/Mtinnitus, headache2 weeks after vaccinationIv/po steroidsSunset glow fundus and dark dots/30 m
Hepatitis A virus (HAV) vaccine
Fine 2001 [67]MEWDS (left eye)Inactivated30/MNot reported13 days after booster vaccinationNoneNone/6 w
Rabies vaccine
Yang 2018 [68]MEWDS (left eye)Inactivated33/FNone7 days after the third doseio steroidsNone/3 y
Co-administration
Stangos 2006 [69]MEWDS (left eye)HAV/yellow fever50/FNone1 week after vaccinationNoneNone/6 w
Cohen 2010 [70]MEWDS (left eye)HPV/Meningococcus17/FHLA-B27(+)1 months after vaccinationNoneNone/2 m
Escott 2013 [71]Acute multifocal choroiditis (right eye)HAV/typhoid/yellow fever33/Mfever, rash, oral ulcers, arthralgias, headache, vertigo3 weeks after vaccinationNoneRPE atrophy/8 w
* In years unless otherwise listed. ^ Time between last vaccination and initial ocular symptom/sign. HBV = hepatitis B virus; APMPPE = acute posterior multifocal placoid pigment epitheliopathy; MEWDS = multiple evanescent white dot syndrome; VKH = Vogt–Koyanagi–Harada; HPV = human papillomavirus; TINU = tubulointerstitial nephritis and uveitis; MMR = measles–mumps–rubella; VZV = varicella zoster virus; HAV = hepatitis A virus; OIS = orbital inflammatory syndrome; OS = left eye; OD = right eye; ERD = exudative retinal detachment; ESR = erythrocyte sedimentation rate; ARN: acute retinal necrosis; NSAIDs = non-steroid anti-inflammatory drugs; RPE = retinal pigment epithelium; HLA = human leukocyte antigen; io = intra-orbital/retrobulbar; iv = intravenous; gtt = eyedrops; po = orally; NR = not reported; Ab = antibody; HZO = herpes zoster ophthalmic; FA = fluorescein angiography.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zou, Y.; Kamoi, K.; Zong, Y.; Zhang, J.; Yang, M.; Ohno-Matsui, K. Ocular Inflammation Post-Vaccination. Vaccines 2023, 11, 1626. https://doi.org/10.3390/vaccines11101626

AMA Style

Zou Y, Kamoi K, Zong Y, Zhang J, Yang M, Ohno-Matsui K. Ocular Inflammation Post-Vaccination. Vaccines. 2023; 11(10):1626. https://doi.org/10.3390/vaccines11101626

Chicago/Turabian Style

Zou, Yaru, Koju Kamoi, Yuan Zong, Jing Zhang, Mingming Yang, and Kyoko Ohno-Matsui. 2023. "Ocular Inflammation Post-Vaccination" Vaccines 11, no. 10: 1626. https://doi.org/10.3390/vaccines11101626

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop