Next Article in Journal
Engaging Broader Stakeholders to Accelerate Group A Streptococcus Vaccine Development
Previous Article in Journal
Factors Associated with Acceptance of Vaccination Against Human Papillomavirus in eThekwini District of South Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Retinal Vascular Occlusion Following COVID-19 Vaccination: A Comprehensive Review of Observational Study and Pathophysiological Mechanisms

1
Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
2
National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
3
Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou 350000, China
4
Department of Ophthalmology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350000, China
5
Harvard Retinal Imaging Lab, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02115, USA
*
Authors to whom correspondence should be addressed.
Vaccines 2025, 13(7), 733; https://doi.org/10.3390/vaccines13070733
Submission received: 1 May 2025 / Revised: 27 June 2025 / Accepted: 2 July 2025 / Published: 7 July 2025
(This article belongs to the Section COVID-19 Vaccines and Vaccination)

Abstract

Background: Retinal vascular occlusion (RVO) and retinal artery occlusion (RAO) have been reported as rare adverse events following COVID-19 vaccination, raising concerns about vaccine safety. This review synthesizes cohort and case–control studies assessing the association between COVID-19 vaccines and RVO/RAO, while exploring potential pathophysiological mechanisms. Methods: We analyzed large-scale population-based studies from South Korea, Europe, and the TriNetX database, focusing on odds ratios (OR), hazard ratios (HR), and relative risks (RR) across mRNA and adenoviral vector vaccines. Pathological processes were hypothesized based on molecular and clinical evidence. Results: Studies investigating the association between COVID-19 vaccination and retinal vascular occlusion show conflicting results; some studies report no association (e.g., OR 0.93, 95% CI 0.60–1.45), others suggest reduced risk (e.g., OR 0.80, 95% CI 0.64–0.99), and one indicates increased risk over two years (HR 2.19, 95% CI 2.00–2.39). Adenoviral vector vaccines, particularly ChAdOx1, show higher RAO incidence in specific cohorts. Proposed mechanisms include vaccine-induced immune thrombotic thrombocytopenia (VITT) via anti-PF4 antibodies, spike protein-mediated endothelial dysfunction, and adjuvant-driven inflammation. Conclusions: While causality remains unproven, temporal heterogeneity and vaccine type-specific risks warrant further investigation. Longitudinal studies with robust controls are needed to clarify these associations in the post-pandemic context.

1. Research Background

1.1. Prevalence and Hazards of COVID-19

Initiated in March 2020, the World Health Organization (WHO) classified COVID-19 as a global public health emergency of international concern (PHEIC), resulting in considerable morbidity and mortality. SARS-CoV-2 is a β-subclass, single-stranded, positive-stranded RNA virus with an envelope and round or oval particles [1] that employs its S protein as a critical structure for binding to host cells via the angiotensin-converting enzyme 2 (ACE2) receptor, leading to a spectrum of respiratory symptoms and disease severity [2]. Ocular manifestations of COVID-19 primarily include inflammation of the conjunctiva, cornea, and sclera, along with uveitis and retinal and optic nerve damage. Retinal vascular occlusion has garnered significant attention from the research community, yet no definitive conclusions have been reached [3,4,5]. A literature review [6] of cases reporting severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pneumonia reported before January 2022 suggested it as a rare but possible manifestation of the disease. A retrospective cohort study [7] of 1,460,634 individuals found that patients with new-onset COVID-19 had a significantly higher risk of branch retinal vein occlusion (BRVO) compared to uninfected controls (hazard ratio [HR] 1.27, 95% confidence interval [CI] 1.04–1.52). Contrasting epidemiological patterns emerged from South Korean surveillance [8]; a modest decline in retinal vein occlusion rates contrasted with rising retinal artery occlusion frequency during COVID-19 surges, though confirmed SARS-CoV-2 infection exhibited no causal association with either condition.

1.2. Vaccine Development, Rationale, and Vaccination Status

The genetic sequence of SARS-CoV-2 was first published on 11 January 2020 [9]. Vaccines against SARS-CoV-2 are the most effective means for controlling the COVID-19 pandemic, developed in a remarkably short timeframe. These include mRNA vaccines, which deliver the genetic code via lipid nanoparticles (e.g., Pfizer/BioNTech (Pfizer Inc. & BioNTech SE, New York, NY, USA, Modern (Moderna, Inc., Cambridge, MA, USA))); adenoviral vector vaccines, which use genetically modified adenoviruses to insert the spiky protein genes (e.g., Janssen (Janssen Vaccines, Leiden, The Netherlands), Oxford—AstraZeneca (AstraZeneca PLC, Cambridge, UK))); and whole virus vaccines, produced by inactivating the virus chemically or physically (e.g., Sinopharm (China National Pharmaceutical Group (Sinopharm), Beijing, China, CoronoVac (Sinovac Biotech Ltd., Beijing, China))). Extensive multicenter clinical trials confirmed their safety prior to market authorization. As of December 2022, over 13 billion doses of the COVID-19 vaccine had been administered globally. However, reports of thrombocytopenia and thromboembolic events, particularly with adenoviral vector vaccines, have raised safety concerns [10,11,12]. Despite the low incidence of these adverse events, European countries suspended the use of the Oxford–AstraZeneca vaccine due to its association with high mortality rate [13]. This has heightened public apprehension. Healthcare professionals have since intensified efforts to closely monitor and report the post-vaccination adverse effects.

1.3. Retinal Vein Occlusion and Retinal Artery Occlusion

Retinal vein occlusion (RVO), the second most prevalent retinal vascular disease, is a leading cause of vision impairment. In 2015, RVO affected over 28.06 million people worldwide, with a prevalence rate of 0.77% among those aged 30–89 [14]. In China, the prevalence of retinal vein occlusion (RVO) significantly increased from 0.099% (95% CI, 0.073–0.126) in 2019 to 0.188% (95% CI, 0.142–0.244) in 2021 [15]. Fundus findings in RVO patients include disk swelling, retinal hemorrhages, tortuous dilated veins, and cotton wool spots. Vison loss may result from complications such as macular edema and macular ischemia, which are persistent and challenging to treat [16,17]. RVO is often associated with systemic inflammation or localized inflammatory ocular disorders, including but not limited to systemic vasculitis, Behçet’s disease, systemic lupus erythematosus, Takayasu’s aortitis, polyarteritis nodosa, and antiphospholipid syndrome. Among localized inflammatory ocular conditions, idiopathic retinal phlebitis is the most common. Non-inflammatory predisposing factors encompass systemic diseases (e.g., hyperhomocysteinemia, hypertension, diabetes mellitus, dyslipidemia, smoking, renal disease) and localized ocular diseases (e.g., glaucoma, hypertensive retinopathy) [18]. Primary RVO, lacking an identifiable etiology beyond these factors, is attributed to the Virchow triad (vascular wall damage, blood stasis, and hypercoagulability) [17,19].
Retinal artery occlusion (RAO) denotes sudden cessation of arterial retinal blood flow due to thromboembolism or vasospasm, causing acute monocular vision loss. This spectrum includes branch (BRAO) and central (CRAO) subtypes. CRAO incidence ranges 1–2/100,000 annually with male predominance, while BRAO constitutes ~33% of RAO cases [20]. Visual deficit severity correlates with occluded vessel caliber and ischemic duration. RAO pathogenesis parallels ischemic stroke, principally involving atherosclerosis, cardiogenic/aortic embolism, vasculitides (e.g., giant-cell arteritis), or hypercoagulable states [16].

2. Cohort and Case–Control Studies: Is RVO or RAO Associated with COVID-19 Vaccines?

Few cohort studies provide odds ratio (OR) on the association between COVID-19 vaccines and retinal vascular occlusion (Table 1). Results vary widely; most studies concluded that there was no association, some suggested the vaccine significantly reduced the risk, and others indicated potential risk.
This causal diagram (Figure 1) illustrates pathways linking COVID-19 vaccines to retinal vascular occlusion.
Three South Korean studies analyzed national databases (NHIS/KDCA) covering millions of participants, assessing 60-day post-exposure outcomes. Vaccines comprised adenoviral (ChAdOx1, Ad26.COV2.S) and mRNA platforms (BNT162b2, mRNA-1273).
The nationwide population-based cohort study of Kim et al. included 2,742,065 individuals aged  ≥  20 years who received SARS-CoV-2 vaccination, matched 1:10 with unvaccinated controls by gender and age. Their findings established that vaccination lowered the risk of retinal vascular occlusion, with an OR of 0.80 (95% CI, 0.64–0.99; p  =  0.039). For those under 40, the protection was even stronger (OR 0.35 for age 20–39, 0.83 for age 40–64, 0.81 for age  ≥  65; p for interaction  =  0.028) [21]. Hwang et al.’s self-controlled case series analyzed 6590 RAO and 70,120 RVO cases, reporting incidence rate ratios (IRRs) (95% CI) of 0.95 (0.88–1.01) and 0.96 (0.94–0.98) during the early risk period (0–28 days), and 0.96 (0.89–1.03) and 0.93 (0.91–0.96) in the late risk period (29–56 days), respectively. They found no significant association [22]. Park et al. [23] stratified cohorts into unvaccinated/uninfected (G1), COVID-19-positive (G2), and vaccinated (G3). Within 60 days, RVO incidence was lowest in G1 (0.0072%, 195/2,680,438), intermediate in G2 (0.0114%, 66/577,471), and highest in G3 (0.0159%, 822/5,160,681) (G1 vs. G2: p = 0.001; G1 vs. G3: p < 0.001; G2 vs. G3: p = 0.009). Notably, vaccinated males exhibited significantly reduced RVO risk versus G1 (aHR 0.77, 0.61–0.99; p = 0.042).
Table 1. This table systematically presents the current evidence base through big data analyses conducted to date investigating the potential association between retinal vascular occlusion and COVID-19 vaccine administration.
Table 1. This table systematically presents the current evidence base through big data analyses conducted to date investigating the potential association between retinal vascular occlusion and COVID-19 vaccine administration.
PaperResearch MethodPopulation Interval Between Onset of IllnessDatabaseGroup DesignConfounders AdjustedBaseline
Characteristics
ExcludedVaccineNumber of Events/Sample Size (Cumulative
Incidence %;
95% CI)
Cumulative Incidence of Vaccine TypeOdds Ratio OR
Confidence Interval (95% CI)
Li JX et al. [24]Nationwide
population-based retrospective
cohort study
4,619,499
745,041 vaccinated and 3,874,458 unvaccinated
12 weeks or 2 yearsTriNetX global
network
Employed
multivariable-adjusted Cox proportional hazards models after performing a 1:1 propensity score matching between the
vaccinated and unvaccinated
cohorts.
age, sex, race, comorbidities, medications and
previous hospitalization
vaccinated group Age: 52.5  ±  18.5 years;
unvaccinated group Age:
52.2  ±  18.2 years.
1. Any diagnosis of retinal vascular occlusion six months prior to the index date.
2. Cases with
confirmation of COVID-19 diagnosis.
3. Cases with the use of any antiplatelets, anticoagulants,
diuretics,
contraceptives, or
antihemorrhages 4 weeks prior to the index date.
BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2.S (Janssen)In 2 years
Vaccinated: 3213/743,505 (0.432%; 95% CI, 0.417–0.447%)
Unvaccinated: 1599/743,505 (0.215%; 95% CI, 0.205–0.226%)
2 years: First dose of BNT162b2: 120/111,491 (0.108%; 95% CI, 0.089–0.127%)
Second dose of BNT162b2: 116/96,135 (0.121%; 95% CI,
0.100–0.143%)
First dose of mRNA-1273: 114/50,382 (0.226%; 95% CI, 0.186–0.267%)
Second dose of BNT162b2: 106/47,536 (0.223%; 95% CI,
0.182–0.265%)
12 weeks: First dose of BNT162b2: 40/111,491 (0.036%; 95% CI, 0.025–0.047%)
Second dose of BNT162b2: 40/96,135 (0.042%; 95% CI,
0.030–0.055%)
First dose of mRNA-1273: 32/50,382 (0.064%; 95% CI,
0.043–0.086%)
Second dose of BNT162b2: 33/47,536 (0.069%; 95% CI,
0.047–0.093%)
HR 2 years 2.19 (95% CI, 2.00–2.39)
HR 12 weeks 3.54 (95% CI, 3.03–4.11)
Dorney, I. et al. [25]Nationwide
population-based retrospective
cohort study
3,108,829 patients receiving the mRNA
COVID-19
vaccine
718,400 Tdap
vaccine
1,470,351 Influenza vaccine
21 daysTriNetX global
network
Group 1
received the first dose of mRNA COVID-19 from 15 December 2020 to 15 June 2022; group 2
received the
second dose of mRNA COVID-19 vaccine from 15 December 2020 to 15 June 2022; group 3
received
influenza
vaccine from 1 June 2018 to 31 December 2019; and group 4 received Tdap vaccine from 1 June 2018 to 31 December 2019.
demographic characteristics (age, sex, race and
ethnicity) and comorbidities
(diabetes, hypertension, and hyperlipidemia)
Age at
vaccination:
50.7 ± 20.4 years; Sex: 56.4% female
Any history of
encounter diagnoses of RVO any time
before the vaccination event
mRNA COVID-19 vaccinationVaccinated: 104/3,108,829 (0.003%; 95% CI, 0.003–0.004%)21 days
First dose of mRNA vaccine: 45/1,180,006 (0.004%; 95% CI, 0.003–0.005%)
Second dose of BNT162b2: 93/1,180,006 (0.006%; 95% CI, 0.008–0.010%)
First dose of COVID-19 mRNA vaccination vs.
influenza
vaccination (RR, 0.74; 95% CI,
0.54–1.01)
First dose of COVID-19
vaccination vs. Tdap vaccination (RR, 0.78; 95% CI, 0.44–1.38),
First dose of COVID-19
vaccination vs.
second dose of COVID-19
vaccination (RR, 2.25; 95% CI,
1.33–3.81)
Kim, Y. et al. [21]Nationwide
population-based retrospective
cohort study
2,742,065
vaccinated
262,603
unvaccinated
60 daysThe
National Health
Insurance Service (NHIS)–COVID-19 cohort
database
Vaccinated
individuals against
SARS-CoV-2.
unvaccinated
individuals matched at a
ratio of
approximately 1:10 by gender and age.
sex, age, income, residence, disability, and CCI scores.age: 48.7 ± 17.1 years; sex: 395,211
(50.9%) male
1. A history of retinal vascular occlusion
2. Aged < 20 years
ChAdOx1 (Astr-Zeneca), BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2.S (Janssen)Vaccinated: 93/262,603 (0.035%; 95% CI,
0.029–0.043%)
Unvaccinated: 725/2,479,462 (0.029%; 95% CI, 0.027–0.031%)
ChAdOx1 503/710,004
(0.071%; 95% CI, 0.065–0.077%)
BNT162b2 162/1,292,421
(0.013%; 95% CI, 0.011–0.015%)
mRNA-1273 44/388,831
(0.011%; 95% CI, 0.008–0.015%)
Ad26.COV2.S 16/88,206
(0.018%; 95% CI, 0.011–0.029)
1. Vaccination
lowered the risk of retinal vascular
occlusion. OR = 0.80 (95% CI,
0.64–0.99; p = 0.039)
2. For individuals aged < 40 years, the vaccination lowered the risk of retinal vascular occlusion occurrence
significantly
compared with those over the age of 40. Age 20–39, OR = 0.35; Age
40–64 OR = 0.83; Age ≥ 65 OR = 0.81; p for interaction = 0.028)
Park, H.S. et al. [23]Nationwide
population-based cohort study.
8,418,59060 daysThe Korea Disease Control and
Prevention Agency (KDCA) and the Korean National Health
Insurance Service (NHIS)
Group 1
included
patients who neither received a diagnosis of COVID-19 nor were vaccinated against
COVID-19
(n = 2,680,438) before 1
September 2021. Group 2
included
patients who
received a
diagnosis of COVID-19
(n = 577 471) once or more
before 31
January 2022. Group 3
included
patients
vaccinated at least once
(n = 5,160,681) before 1 September 2021.
age, sex, and the presence of hypertension, DM, or dyslipidemiagroup 1 age:
33.5 ± 13.3 years; group 2 age:
41.7 ± 21.6 years; group 3 age:
54.2 ± 17.1 years.
The population of group 1 was younger, was
predominantly male, and had fewer
comorbidities,
including
hypertension and DM, compared with that of groups 2 and 3.
1. A diagnosis of RAO or RVO before the
preset index date of each group.
2. Patients with
incomplete data regarding the first
vaccination
(n = 32 269) or
demographic data (such as sex and age;
n = 943)
ChAdOx1 (AstraZeneca), BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2 S (Janssen)60 days RAO
group 1: 13/2,680,438 (0.0005%; 95% CI, 0.0003–0.0008%)
group 2: 4/577,471 (0.0007%; 95% CI, 0.0002–0.0018%)
group 3: 73/5,160,681 (0.0014%; 95% CI, 0.0011–0.0018%)
60 days RVO
group 1: 195/2,680,438 (0.0072%; 95% CI, 0.0063–0.0083%)
group 2: 66/577,471 (0.01143%; 95% CI, 0.0089–0.0144%)
group 3: 822/5,160,681 (0.0159%; 95% CI, 0.0149–0.0170%)
In women who received
mRNA-1273 vaccines, who showed a higher RAO HR (4.65; 95% CI, 1.27–17.03; p = 0.021).
Using incidence rates of group 1 as a reference
RAO: Vaccination male group, HR 0.82 (95% CI, 0.53–1.17, p-value = 0.644)
Vaccination female group, HR 1.37 (95% CI, 0.42–4.44, p-value = 0.602)
RVO: Vaccination male group, HR 0.77 (95% CI, 0.61–0.99, p-value = 0.042)
Vaccination female group, HR 1.03 (95% CI,
0.79–1.35,
p-value = 0.807)
Hwang, S. et al. [22]Nationwide
population-based self-controlled case series
6590 cases of
incident RAO 70,120 cases of
incident RVO
early risk period (0 to 28 days from vaccination)
late risk period (29 to 56 days from vaccination)
Korea
National Health
Insurance Service (NHIS) and Korea Disease Control and
Prevention Agency (KDCA)
Any period
immediately
following
vaccination was designated as a risk period.
Any other time during the study period was
considered as an unexposed
control period.
Pre-risk period
(−28 to −1 days from
vaccination)
Risk period, early (0 to 28 days from
vaccination)
Risk period, late (29 to 56 days from
vaccination)
/RAO: Age
67.69 ± 13.28; Sex: 4025 (61.08%) male. Income level: Low 1730 (26.25%);
Previous disease history:
Cerebro-cardiovascular 2237 (33.95%),
Rheumatic
autoimmune 1056 (16.02%).
RVO: Age:
63.76 ± 13.88;
Sex: 32,318 (46.09%) male;
Income level Low 19,395 (27.66%); Previous disease history
Cerebro-cardiovascular 16,288 (23.23%),
Rheumatic
autoimmune 9759 (13.92%).
1. A history of the
disease within the 6 years before the
observation period
2. Passed away during the study period
3. Missing vaccination or demographic data.
ChAdOx1 (AstraZeneca), BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2 S (Janssen)RAO: Control
period: 3885/1,711,590 (0.227%; 95% CI, 0.220–0.234%)
Pre-risk period: 819/380,402 (0.215%; 95% CI,
0.201–0.230%)
Risk period, early: 1040/497,916 (0.209%; 95% CI, 0.196–0.222%) Risk period, late: 846/408,542 (0.207%; 95% CI,
0.193–0.221%)
RVO: Control
period: 40,916/17,900,627 (0.229%; 95% CI, 0.227–0.231%)
Pre-risk period: 8399/4,110,437 (0.204%; 95% CI, 0.200–0.208%)
Risk period, early: 11,572/5,422,603 (0.213%; 95% CI, 0.209–0.217%)
Risk period, late: 9233/4, 470,933 (0.207%; 95% CI, 0.203–0.211%)
/0–28 days
postvaccination, RAO IRR 0.95
(0.88–1.01), RVO IRR0.96 (0.94–0.98)
29–56 days RAO IRR 0.96 (0.89–1.03), RVO IRR 0.93
(0.91–0.96)
Pellegrini, M. et al. [26]multicenter
self-controlled case series
210 RVOs
patients
28 daysFive
tertiary
referral centers in Italy
Days 1–14 after first dose
days 14–28 after first dose
days 1–28 after first dose
days 1–14 after second dose
days 14–28 after second dose
days 1–28 after second dose
all other
observed time
/Age: 68.8 ± 12.3 years; Sex: 111 (52.9%) males1. Diagnosis of RVO prior to the study
period
2. Unavailability of data regarding COVID-19 vaccination status.
ChAdOx1 (AstraZeneca), BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2 S (Janssen)//Days 1–14 after first dose, IRR 0.87
(0.41–1.85)
days 14–28 after first dose, IRR 1.01 (0.50–2.04)
days 1–28 after first dose, IRR 0.94
(0.55–1.58)
days 1–14 after
second dose, IRR 1.21 (0.62–2.37)
days 14–28 after
second dose, IRR 1.08 (0.53–2.20)
days 1–28 after
second dose, IRR 1.16 (0.70–1.90)
PaperResearch MethodPopulation Interval Between Onset of IllnessDatabaseGroup DesignConfounders AdjustedBaseline
Characteristics
ExcludedVaccineNumber of Events/Sample Size (Cumulative
Incidence %;
95% CI)
Cumulative Incidence of Vaccine TypeOdds Ratio OR
Confidence
Interval (95% CI)
Feltgen, N. et al. [27]Case-by-case analysis
(descriptive case-only study)
population-based Case–control study cohort study
421 retinal
vascular disease patients
28 daysan
adjusted conditional logistic
regression analysis was conducted
Gutenberg Health Study
In this analysis, we compared the odds of being vaccinated in the last four weeks among patients with RVOD (cases) to
controls from the general
population
recruited by the Gutenberg Health Study (GHS) (age ±5 years and sex-matched).
adjusted for
obesity (BMI ≥ 30),
diabetes, arterial hypertension, smoking, and use of anticoagulation.
Age: 67.6 ± 14.6 years; Sex: 51.8% female; 49.0%
Eye (OS).
A substantial
cardiovascular risk profile in the cohort.
/ChAdOx1 (AstraZeneca), BNT162b2 (Pfizer), mRNA-1273 (Moderna), Ad26.COV2 S (Janssen)//OR = 0.93; 95%
CI: 0.60–1.45,
p = 0.75
CI calculated using Clopper–Pearson exact method for binomial proportions.
Two European studies from Germany and Italy conducted multicenter self-controlled retinal vascular occlusion case series that may be associated with the COVID-19 vaccine and integrated baseline data from local populations. Feltgen et al. found no increased RVO risk 4 weeks post-vaccination (OR 0.93; 95% CI 0.60–1.45, p = 0.75) [27]. Pellegrini et al. reported no significant risk of RVO after the first dose (1–28 days IRR: 0.94, 95% CI 0.55–1.58) or second dose of the four vaccines (1–28 days IRR: 1.16, 95% CI 0.70–1.90) [26]. Both analyzed adenovirus vector vaccines (ChAdOx1 nCoV-19, Ad26.COV2.S) and mRNA vaccines (BNT162b2, mRNA-1273), finding no vaccine type-specific associations. However, their limited sample sizes (hundreds of cases) reduced statistical power.
Given RAO’s lower prevalence, few studies focus solely on RAO, often combining it with RVO analyses. For instance, Feltgen et al. included 118 RAO cases [27] and Hwang et al. included 6590 [22], both concluding no association with vaccination. However, Park et al. reported a higher cumulative RAO incidence with ChAdOx1 (0.0018%) [23]. Notably, the HR for RAO in women vaccinated with mRNA-1273 was 4.65 (95% CI 1.27–17.03; p = 0.02), warranting further investigation.
It is noteworthy that two studies using the TriNetX global network database reached opposing conclusions regarding mRNA vaccines. Li et al. [24], after excluding cases with a confirmed COVID-19 diagnosis or the use of specific medications (antiplatelet, anticoagulant, diuretic, contraceptive, antihemorrhagics), analyzed 4,619,499 individuals, matching vaccinated (BNT162b2 or mRNA-1273) and unvaccinated cohorts 1:1 by age and RVO risk factors. Using Cox multivariate and Kaplan–Meier analyses, they reported a 2.19-fold higher risk of retinal vascular occlusion in the vaccinated cohort over 2 years (95% Cl 2.00–2.39), with significantly higher cumulative incidence at 12 weeks and 2 years (log rank p < 0.001). Meanwhile, Dorney et al. [25] examined over 3 million individuals vaccinated with mRNA COVID-19 vaccines (Group 1), propensity score-matching them with influenza (Group 2) and Tdap (Group 3) vaccine recipients from 2018 to 2019 to assess the relative risk (RR) of new RVO diagnoses within 21 days. They drew the conclusion that there was no significant difference in RR post-COVID-19 vaccination compared to influenza (RR 0.74, 95% CI 0.54–1.01) or Tdap (RR 0.78, 95% CI 0.44–1.38) vaccination.
These conflicting findings have sparked debate. Both studies used the TriNetX database for 2020–2022 RVO cases and applied propensity score matching via the nearest neighbor greedy algorithm. However, control group selection, exclusion criteria and time windows selection differed significantly. Li et al. chose unvaccinated individuals from the same period, while Dorney et al. selected influenza and Tdap recipients from 2018 to 2019. COVID-19 infection’s impact on coagulation is a known risk factor for RVO. Dorney et al. did not exclude infected cases, despite the U.S. Centers for Disease Control and Prevention estimating a 43.9% infection rate. Li et al., relying solely on positive test records in TriNetX, may not have fully excluded asymptomatic or untested cases. Utilizing linked health claims and vaccination registries, Hashimoto et al. [28] assessed post-vaccination ocular events in Japan while excluding COVID-19-infected individuals. Their matched cohort analysis identified elevated RVO and composite outcome risks post-dose two, whereas self-controlled case series (SCCS) revealed no significant association for any ocular event. Additionally, Dorney et al. included patients on anticoagulants, antiplatelets, contraceptives, diuretics, and other medications without matching for these factors, potentially confounding coagulation status. Their observation windows also differed; Dorney used 21 days, while Li examined 12 weeks and 2 years. A shorter window aligns with case reports averaging a 2-week diagnostic interval. Longer periods during a pandemic may introduce confounding from repeated COVID-19 exposures, vaccination updates, or interim health changes. Extended windows may also detect delayed autoimmune responses, which clinical evidence suggests manifest after six weeks or more.
These studies face limitations due to RVO and RAO’s multifactorial nature. Firstly, accounting for all contributing factors is challenging, with key variables like ethnicity and ocular comorbidities often omitted. Secondly, the rarity of these events limits study power, particularly in smaller European cohorts. Thirdly, despite propensity score matching, residual imbalances in cardiovascular risk factors (e.g., age, hypertension, dyslipidemia) may bias results. Designing rigorous cohort studies during a pandemic with rapid viral transmission and complex clinical scenarios remains difficult.
Evidence shows temporal heterogeneity in RVO risk post-vaccination. Studies with ≤4-week windows typically report no association. A 60-day analysis found higher RVO incidence in vaccinated vs. unvaccinated cohorts (median diagnosis latency: 38 days). Li et al.’s extended observation (12 weeks to 2 years) uniquely identified vaccination as an RVO risk factor. This raises the question, do vaccine side effects emerge over time, increasing RVO incidence, as Li et al. suggest? Further research is needed.
Vaccine platforms exhibit safety profile heterogeneity for RVO risk. Kim et al. [21] reported a 4–6-fold higher RVO risk with ChAdOx1 compared to mRNA vaccines in a case–control analysis. Park et al. [23] reported significant sex-based differential risks; mRNA-1273-vaccinated females had a markedly elevated RAO hazard (aHR 4.65, 1.27–17.03; p = 0.02), whereas BNT162b2-vaccinated males showed reduced RVO incidence (aHR 0.66, 0.49–0.88; p = 0.004). However, most population-based studies found no significant platform-specific differences.
Emerging data suggest that most RVO cases occur after the first dose. Dorney et al. reported an RR of 2.25 (95% CI 1.33–3.81) for RVO after the first vs. second dose [25]. Park et al. found that 77% (633/822) of cases emerged post-first dose [23]. Günalp Uzun et al. noted a low VITT recurrence risk (3%) with mRNA boosters after ChAdOx1 priming [29]. Pellegrini et al. [26] demonstrated distinct dose–response patterns; adenovirus-vectored vaccines showed higher first-dose IRR at 4 weeks (1.61, 95% CI 0.57–4.55) versus second dose, while mRNA vaccines exhibited elevated second-dose IRR (1.12, 95% CI 0.52–2.38) compared to initial administration. This may reflect differences in the pathogenic principles of the two types of vaccines.
For patients with potential vaccine-related RVO, decisions on revaccination should consider systemic inflammatory markers (ESR/CRP), coagulation markers (D-dimer, anti-PF4 antibodies), fundus perfusion, and visual recovery.

3. Pathological Processes

The mechanisms linking COVID-19 vaccines to retinal vascular occlusion remain incompletely understood. For delayed central retinal vascular occlusion, vaccine-induced immune thrombotic thrombocytopenia (VITT) may be implicated. Peripheral occlusion within two weeks of vaccination may stem from immune responses to spike proteins or adjuvants, cytokine storms from allergic reactions, or vasculitis, mimicking the Virchow triad (vascular wall damage, blood stasis, hypercoagulability). We propose the following mechanisms based on observed blood abnormalities and pathophysiological evidence (Figure 2).

3.1. PF4 and VITT

Platelet factor 4 (PF4), or chemokine (C—X—C motif) ligand 4 (CXCL4), is abundant in platelets (20 μg/109 platelets) with plasma levels of 2–10 ng/mL [30,31]. At physiological pH, PF4 exists as a highly cationic tetramer with extensive positive charge [32]. Pre-pandemic research on heparin-induced thrombocytopenia (HIT) showed that heparin, a polyanion, binds PF4, triggering anti-PF4-heparin antibodies that activate platelets, monocytes, neutrophils, and endothelial cells, leading to thrombocytopenia [33]. During mass vaccination, adenoviral vector vaccines, particularly ChAdOx1, have been linked to VITT [34]. Laboratory evidence suggests that adenoviral DNA may act as a polyanion, initiating VITT in susceptible individuals.
The VITT process may unfold as follows. Within a week of vaccination, endothelial injury from needle puncture or disruption of tight junctions by vaccine components (e.g., EDTA) may allow vaccine entry into the bloodstream [35]. Platelets may be activated by injection-related vascular injury, adenoviral binding to platelet surfaces, or immune complexes of host cell proteins and IgG antibodies. Adenoviral particles bind PF4 from activated platelets or the endothelial matrix [36,37]. These PF4-vaccine complexes are transported to the spleen, where B cells with PF4-specific Ig receptors proliferate, releasing high-titer anti-PF4 IgG within a week. FcRγIIA-mediated signaling of PF4-anti-PF4 IgG complexes induces pro-coagulant thrombocytosis, platelet/neutrophil aggregation, and NETosis. NETosis-released DNA amplifies immune damage, activating complement deposition in the endothelium. Activated endothelial cells express tissue factor and release von Willebrand factor (VWF), which binds activated platelets and clotting factors, further promoting neutrophil activation and thrombin generation, culminating in thrombosis [38].
An alternative hypothesis posits that circulating PF4 interacts with negatively charged extracellular DNA at the vaccination site, forming immune complexes that hyperactivate plasmacytoid dendritic cells (pDCs) via TLR9, boosting interferon-α production. RNA elicits a weaker effect. Memory B cells in regional lymph nodes then increase autoantibody production against PF4, perpetuating a cycle of FcRγII-mediated platelet activation and severe thrombosis.
The primary initiator of these mechanisms remains unclear, but adenoviral vaccines, containing both DNA and protein components, are the most implicated in VITT. ChAdOx1’s high electronegative surface potential enhances its interaction with oppositely charged proteins like PF4 [36]. Nucleic acids bind PF4 when plasma concentrations exceed 200 ng/mL, though vaccine-derived nucleic acids typically remain below this threshold [39]. Electron and super-resolution microscopy indicate that PF4-anti-PF4 VITT antibody complexes primarily bind nonstructural vaccine components (e.g., host cell proteins, unassembled hexons), amplified by DNA [40].
Thrombosis and thrombocytopenia in VITT are mediated by distinct mechanisms. Anti-PF4-polyanion antibodies drive platelet and neutrophil activation, with thrombocytopenia potentially resulting from direct platelet binding [38]. VWF knockout mice showed no significant thrombocytopenia post-adenovirus, suggesting VWF and P-selectin mediate platelet clearance [37]. NETosis, driven by neutrophil activation, is a key contributor to VITT thrombosis, with inhibition of NETosis (via GSK484 or PAD4 knockdown) preventing thrombosis but not thrombocytopenia. In platelet absence, PF4 immune complexes can directly activate neutrophils. Despite its prominence, VITT remains rare, with anti-PF4-polyanion antibodies typically low in optical density and not inducing abnormal platelet activation in most vaccinated individuals [41].

3.2. Spike Protein

SARS-CoV-2, an RNA virus, completes its lifecycle in the cytoplasm, lacking evolutionary pressure for splice donor (SD) and acceptor (SA) sites. Vaccines based on mRNA technology, whose functional activity is limited to the cytoplasm, differ fundamentally from adenoviral vector vaccines, where transcription occurs in the nucleus and undergoes RNA splicing [42]. Experimental data show that adenoviral vector vaccines can produce soluble spike protein variants due to intranuclear splicing, allowing these proteins to enter the bloodstream [42]. However, free spike antigen was detected in the blood of adolescents and young adults who developed post-mRNA vaccine myocarditis [43]. Soluble spike proteins may trigger signaling pathway abnormalities through direct binding to receptors or through immune-mediated cell injury, ultimately leading to microangiitis and microthrombosis.
We hypothesize that soluble spike proteins bind angiotensin-converting enzyme 2 (ACE2) via their receptor-binding domains (RBDs), with anti-spike antibodies recognizing these variants on endothelial cells [44,45]. This may trigger local inflammation through (1) cytokine/chemokine induction (e.g., IL-6, IL-1β, TNFα, CXCL1, CXCL2, CCL2); (2) antibody-dependent cell-mediated cytotoxicity (ADCC), where NK cells target endothelial cells via Fcγ receptors [46,47]; and (3) complement-dependent cytotoxicity (CDC), where complement activation promotes membrane attack complex MAC formation, platelet activation, and thrombin release [48].
Spike proteins impair endothelial function by reducing nitric oxide synthase activity and mitochondrial function, increasing redox stress, and destabilizing ACE2, potentially exacerbating endothelial dysfunction via the renin-angiotensin system [49]. Although genetically modified to enhance immunity and prevent ACE2 binding, soluble spike proteins may still induce inflammatory responses in endothelial cells, mimicking the thromboembolic effects of wild-type SARS-CoV-2 or pseudoviruses [50].
These variants may explain rare adverse events post-Vaxzevria (ChAdOx1) vaccination. Unlike Ad5.S and ChAdOx1.S, Ad26.COV2. S lacks key splice sites (SD506, SD3614), with no significant spliced RNA detected, suggesting lower risk [42]. However, evidence indicates platelets and megakaryocytes lack ACE2, implying alternative entry mechanisms (e.g., TLR7) for SARS-CoV-2 or spike proteins, requiring further investigation [50], such as through transferrin receptor [51].

3.3. Allergic Reactions or Vasculitis Due to Adjuvants

Adjuvants in vaccines enhance stability and efficacy but may contribute to rare thrombotic events. Though RVO reports linked to adjuvants are scarce, hypotheses suggest immune/allergic reactions or small-vessel vasculitis. Polyethylene glycol (PEG) in mRNA vaccines (BNT162b2, mRNA-1273) may bind anti-PEG antibodies (prevalence 25% vs. 0.2% historically [52,53]), activating complement (C3a, C5a) and inflammatory cells (macrophages, basophils, mast cells), releasing mediators like histamine and platelet-activating factor [54,55]. The ChAdOx1 nCov-19 and Ad26. COV. 2 vaccines contain polysorbate 80, a synthetic nonionic surfactant and emulsifier that is widely utilized to fabricate nanoparticle drug delivery platforms. Structurally, polysorbate 80 resembles PEG, featuring repetitive ethylene oxide sidechains that may elicit analogous allergic reactions [56]. In preclinical studies, polysorbate 80 enhances membrane permeability and disrupts the blood–brain and intestinal barriers [57]. However, no empirical evidence currently suggests analogous effects on the blood-retinal barrier, which is structurally similar. EDTA in ChAdOx1 may activate platelets and cause capillary infiltration, though its low dose (500 µL intramuscularly) makes systemic effects unlikely [58].
Lipid nanoparticles (LNPs) in mRNA vaccines, comprising phospholipids, cholesterol, PEG-lipids, and ionizable lipids, exhibit adjuvant activity, protecting mRNA and aiding delivery [59]. In mice, LNPs trigger neutrophil infiltration, inflammatory pathways, and cytokine/chemokine production (e.g., IL-1β, IL-6) [60]. Studies show that Pfizer’s Comirnaty and Moderna’s Spikevax increase C5a, sC5b-9, and Bb in serum, with Comirnaty inducing pro-inflammatory cytokines (IL-1α < IFN-γ < IL-1β < TNF-α < IL-6 < IL-8) in PBMC cultures [61]. These responses may cause transient endothelial dysfunction and inflammation, particularly after the second dose [62], predisposing vessels to thrombosis [63,64,65,66].

3.4. Molecular Mimicry

Molecular mimicry, where vaccine components resemble human proteins, may trigger autoimmunity in susceptible individuals [67]. Computational analysis identified 5693 epitopes in BNT-162b2 corresponding to 21 viral and human proteins, some linked to immune diseases via HLA alleles [68]. However, no direct evidence connects molecular mimicry to retinal vascular occlusion.

3.5. Why Thrombosis Occurs in Retinal Vessels

Retinal vessels may be particularly susceptible to vaccine-related thrombosis due to their unique anatomy and physiology. The retina’s high metabolic demand and limited collateral circulation amplify the impact of vascular occlusion. Endothelial cells in retinal capillaries express ACE2, potentially interacting with soluble spike proteins or immune complexes [49]. The Virchow triad—exacerbated by vaccine-induced endothelial damage, stasis from inflammation, and hypercoagulability from VITT or adjuvants—may preferentially manifest in these small-caliber vessels. Additionally, the retina’s immune-privileged status may delay clearance of inflammatory mediators, prolonging thrombotic risk [69]. Further studies on retinal-specific factors are needed (Figure 3).
Figure 2. The schematic diagram illustrates currently proposed hypotheses of pathophysiological mechanisms associated with thromboembolic events or vasculopathic responses following administration of adenoviral vector-based and mRNA COVID-19 vaccines. This image was drawn using Figdraw.
Figure 2. The schematic diagram illustrates currently proposed hypotheses of pathophysiological mechanisms associated with thromboembolic events or vasculopathic responses following administration of adenoviral vector-based and mRNA COVID-19 vaccines. This image was drawn using Figdraw.
Vaccines 13 00733 g002
Figure 3. This figure delineates the pathophysiological mechanisms underlying hemodynamic disturbances and neurovascular architecture deconstruction in the delicate retinal tissue, mediated by systemic vascular risk factors, culminating in vision loss. This image was drawn using Figdraw.
Figure 3. This figure delineates the pathophysiological mechanisms underlying hemodynamic disturbances and neurovascular architecture deconstruction in the delicate retinal tissue, mediated by systemic vascular risk factors, culminating in vision loss. This image was drawn using Figdraw.
Vaccines 13 00733 g003
Due to data volume limitations, big data studies do not visually analyze retinal conditions through fundus imaging; they often use codes in the system to confirm diagnoses. In this regard, there are small cohort studies that have analyzed retinal perfusion after vaccination [70,71,72]. Using the optical coherence tomography angiography (OCTA) in individuals vaccinated with the Pfizer-BioNTech vaccine, Birumut Gedik et al. [70] found a significant decrease in vessel density and a significant increase in retinal thickness. However, in patients vaccinated with inactivated virus vaccine, no such changes in retinal structure were observed.

4. Discussion

In the post-pandemic era, researchers are exploring whether reinfection or multiple infections post-vaccination elevate complication risks. Current focus examines if residual vaccine components (nucleic acids, adenoviral vectors, spike proteins) amplify immune responses during subsequent SARS-CoV-2 infections. Notably, VITT-related anti-PF4 antibodies do not cross-react with viral spike proteins [73], and COVID-19-associated thrombosis operates distinctly from VITT pathways [67]. Park et al.’s subgroup analyses [23] showed higher RAO and RVO risks in individuals vaccinated before COVID-19 diagnosis compared to those vaccinated post-diagnosis or unvaccinated, though risks remained lower than in unvaccinated, uninfected individuals. This suggests no excessive concern for vaccine-COVID-19 interactions is warranted. In contrast, other analysis [5] indicates a significant increase in the incidence of retinal vein occlusions (RVOs) within 6 months post-COVID-19 diagnosis (adjusted IRR, 1.54; 95% CI, 1.05–2.26; p = 0.03). Although retinal artery occlusions (RAOs) showed a nominally elevated incidence (adjusted IRR, 1.35; 95% CI, 0.64–2.85; p = 0.44), this did not reach statistical significance. Critically, COVID-19 vaccination demonstrates well-established protective effects against severe respiratory complications (e.g., acute respiratory distress syndrome) and cardiovascular sequelae following SARS-CoV-2 infection. While the link between vaccination and fundus blood flow issues remains inconclusive, COVID-19 itself poses a far greater risk than vaccines.
The clear conclusion we can draw is that cases of retinal vascular occlusion following vaccination are rare. In our review, we found that most of the cases had common risk factors for retinal vascular occlusion such as hypertension and hyperlipidemia. Individuals at particularly high risk should be informed of this risk before vaccination. At the same time, these individuals should be protected by routine visual acuity examinations after vaccination.
Case reports noting short intervals between COVID-19 vaccination and RVO onset likely reflect reporting bias, with clinicians attributing proximate events to vaccination absent robust pathological evidence. Molecular and animal studies suggest adenoviral vector-induced VITT manifests > 14 days post-vaccination. Many studies fail to distinguish vaccine types, despite their differing compositions and mechanisms. Future case reports should document inflammatory markers and anti-PF4 antibodies, analyzing onset intervals and the presence or absence of COVID-19 infection meticulously.
Based on compositional and pathogenic mechanisms, the immunogenicity of mRNA vaccine components—specifically spike proteins and LNP carriers—may induce vascular tissue damage and trigger thrombosis. However, as noted previously, ChAdOx1 contains adenoviral vectors with higher pathogenic potential and more frequent RNA intranuclear splicing, potentially leading to greater clinical incidence. At the level of patient-derived antibodies, the striking similarity in autoantibody fingerprints between these two conditions strongly indicates that VITT and adenovirus infection-associated anti-PF4 disorders represent a distinct category of adverse immune responses targeting adenoviral structures [74]. While minimal differences exist for retinal vascular occlusion, ChAdOx1 demonstrates comparatively higher risks in other immune-mediated and thrombotic diseases versus mRNA vaccines [75].
Four years after the COVID-19 vaccination campaigns, sporadic reports describe patients experiencing prolonged symptoms lasting 12 months or longer following acute post-vaccination reactions [76,77,78]. These observations suggest potential long-term vaccine-associated effects, though establishing causality faces significant challenges. This diagnostic uncertainty underscores the need for internationally accepted criteria to define “long post-COVID-19 vaccination syndrome”. Standardized enrollment protocols would enable systematic global data collection and analysis.
In addition to this, we need more high-quality studies to analyze the incidence of long-term disease after COVID-19 vaccination, such as the article by Li et al. [24]. Such studies will clarify whether vaccine-related complications have the potential for delayed onset. Concurrently, we must assess if biological mechanisms exist for delayed-onset vaccine-related manifestations. A novel disease entity termed VITT-Like Monoclonal Gammopathy of Thrombotic Significance (VITT-like MGTS) has been proposed. This condition is characterized by M-protein production and the presence of VITT-like autoantibodies in serum/plasma. Crucially, the M-protein itself exhibits platelet-activating properties analogous to those in classical VITT [79]. Patients present with chronic, progressive, or intermittent thrombocytopenia and develop a chronic prothrombotic hypercoagulable state, leading to recurrent systemic thrombosis. The disease course may persist for years. Whether retinal vascular thrombosis represents a clinical manifestation of VITT-like MGTS remains unestablished and warrants further investigation [80].
Investigating the association between retinal vascular occlusion (RVO) and COVID-19 vaccines extends beyond observational studies, with prospective interventional trials representing a critical methodological avenue—though none have been published to date. Given RVO’s multifactorial pathophysiology and heterogeneous progression, adaptive trial designs offer distinct advantages. By actively assigning interventions (e.g., vaccine platform, dosing intervals, sample size re-estimation) while controlling variables, such designs directly quantify causal effects and minimize residual confounding.
Within retrospective frameworks, propensity score weighting—particularly inverse probability treatment weighting (IPTW) coupled with machine learning algorithms—has emerged as a key technical approach for handling high-dimensional confounders. IPTW assigns each subject a weight to construct a pseudo-randomized population where measured covariates achieve inter-group balance, simulating randomized trial conditions. Machine learning techniques (e.g., LASSO regression, random forests, gradient boosting machines) enable robust propensity score estimation through effective processing of numerous covariates—including nonlinear relationships and interactions—while performing variable selection to reduce noise.
The self-controlled case series (SCCS) method may provide longitudinal advantages for pathophysiological timeline investigation. However, this design carries limitations; it may fail to address within-individual temporal confounding from time-varying factors (e.g., healthcare utilization, medication changes, or disease progression), potentially leading to vaccine-attribution bias. Consequently, SCCS application requires presuming transient, acute vaccine effects on RVO. Prolonged risk windows amplify time-varying confounding, making precise definition of risk/control window durations challenging yet essential.
In the context of the post-epidemic era, this paper systematically reviews the association between COVID-19 vaccination and the risk of retinal vascular occlusion (RVO and RAO) with the aim of integrating the existing evidence, dissecting the underlying thrombotic mechanisms. Building upon prior studies, our study reveals the mechanisms that may cause vascular occlusion by direct comparative analysis of the biological components of adenoviral vectors and mRNA vaccine platforms. These findings directly inform clinical protocols for monitoring high-risk subgroups, such as individuals with anti-PF4 antibody profiles. These findings do not go to blame the vaccines that have made a significant contribution to controlling the COVID-19 pandemic, but rather to inform approaches for next-generation vaccine development [81]—including the rational design of low-immunogenicity delivery vectors and adjuvant systems [82,83]—to optimize the safety and efficacy of vaccination programs in preparation for possible future pandemics.

5. Conclusions

Similarly to other systemic vasculopathies, current evidence does not establish a definitive association between COVID-19 vaccines and retinal vascular occlusions in the short term. Due to the lack of research evidence, we cannot jump to conclusions about the effects of vaccines within a long window of time. For patients with predisposing factors such as hyperlipidemia or hypertension, comprehensive hemodynamic evaluations should precede vaccination to mitigate potential risks associated with Virchow’s triad. Post-pandemic surveillance of visual acuity changes remains imperative for early detection of vaccine-related or COVID-19-related ophthalmic sequelae.
Big data analytics face inherent challenges in investigating retinal vascular occlusions due to their low incidence rates, multifactorial etiology, and confounding from widespread COVID-19 infections. Given the near-universal exposure to COVID-19 vaccines and the challenges of studying rare outcomes like retinal vascular occlusion (RVO) with multiple confounders, no single methodological approach is flawless. We must strategically combine complementary study designs to mitigate their respective limitations. Longitudinal investigations of pathophysiological timelines, either after COVID-19 infection or after vaccination, will optimize critical observation window selection for causal inference.

Author Contributions

Conceptualization and funding acquisition, F.C. and C.C.; literature/data curation, H.Z., K.L., and X.L.; pathology analysis and visualization, Y.Z., C.C., and H.C.; original draft preparation (cohort analysis and mechanistic synthesis), Y.Z.; writing—review and editing (clinical/mechanistic rigor), H.C. and C.C.; final validation and supervision, F.C. and C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (grant number: 82271096).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
RVORetinal vascular occlusion
RAORetinal artery occlusion
OROdds ratios
HRHazard ratio
RRRelative risks
VITTVaccine-induced immune thrombotic thrombocytopenia
PF4Platelet Factor 4
ACE2Angiotensin-converting enzyme 2
SARS-CoV-2Syndrome-coronavirus-2
CIConfidence interval
EDTAEthylenediaminetetraacetic acid
VWFVon Willebrand factor
pDCsPlasmacytoid dendritic cells
SDSplice donor
SASplice acceptor
RBDsReceptor-binding domains
ADCCAntibody-dependent cell-mediated cytotoxicity
CDCComplement-dependent cytotoxicity
MACMembrane attack complex
OCTAOptical coherence tomography angiography

References

  1. Attaway, A.H.; Scheraga, R.G.; Bhimraj, A.; Biehl, M.; Hatipoğlu, U. Severe covid-19 pneumonia: Pathogenesis and clinical management. BMJ 2021, 372, n436. [Google Scholar] [CrossRef] [PubMed]
  2. Leung, N.H.L. Transmissibility and transmission of respiratory viruses. Nat. Rev. Microbiol. 2021, 19, 528–545. [Google Scholar] [CrossRef] [PubMed]
  3. Badawi, A.E.; Elsheikh, S.S.; Addeen, S.Z.; Soliman, M.A.; Abd-Rabu, R.; Abdella, W.S.; Gad, E.A. An Ophthalmic Insight into Novel Coronavirus 2019 Disease: A Comprehensive Review of the Ocular Manifestations and Clinical Hazards. J. Curr. Ophthalmol. 2020, 32, 315–328. [Google Scholar] [CrossRef]
  4. Ng, H.W.; Scott, D.A.R.; Danesh-Meyer, H.V.; Smith, J.R.; McGhee, C.N.; Niederer, R.L. Ocular manifestations of COVID-19. Prog. Retin. Eye Res. 2024, 102, 101285. [Google Scholar] [CrossRef]
  5. Modjtahedi, B.S.; Do, D.; Luong, T.Q.; Shaw, J. Changes in the Incidence of Retinal Vascular Occlusions After COVID-19 Diagnosis. JAMA Ophthalmol. 2022, 140, 523–527. [Google Scholar] [CrossRef]
  6. Yeo, S.; Kim, H.; Lee, J.; Yi, J.; Chung, Y.R. Retinal vascular occlusions in COVID-19 infection and vaccination: A literature review. Graefes Arch. Clin. Exp. Ophthalmol. 2023, 261, 1793–1808. [Google Scholar] [CrossRef] [PubMed]
  7. Li, J.X.; Wei, J.C.; Wang, Y.H.; Bair, H.; Hsu, S.B.; Lin, C.J. RETINAL VASCULAR OCCLUSION AND COVID-19 DIAGNOSIS: A Multicenter Population-Based Study. Retina 2024, 44, 345–352. [Google Scholar] [CrossRef]
  8. Park, H.S.; Kim, S.; Lee, C.S.; Byeon, S.H.; Kim, S.S.; Lee, S.W.; Kim, Y.J. Retinal vascular occlusion risks during the COVID-19 pandemic and after SARS-CoV-2 infection. Sci. Rep. 2023, 13, 16851. [Google Scholar] [CrossRef]
  9. Le, T.T.; Andreadakis, Z.; Kumar, A.; Gómez Román, R.; Tollefsen, S.; Saville, M.; Mayhew, S. The COVID-19 vaccine development landscape. Nat. Rev. Drug Discov. 2020, 19, 305–306. [Google Scholar] [CrossRef]
  10. Pottegård, A.; Lund, L.C.; Karlstad, Ø.; Dahl, J.; Andersen, M.; Hallas, J.; Lidegaard, Ø.; Tapia, G.; Gulseth, H.L.; Ruiz, P.L.; et al. Arterial events, venous thromboembolism, thrombocytopenia, and bleeding after vaccination with Oxford-AstraZeneca ChAdOx1-S in Denmark and Norway: Population based cohort study. BMJ 2021, 373, n1114. [Google Scholar] [CrossRef]
  11. Bayas, A.; Menacher, M.; Christ, M.; Behrens, L.; Rank, A.; Naumann, M. Bilateral superior ophthalmic vein thrombosis, ischaemic stroke, and immune thrombocytopenia after ChAdOx1 nCoV-19 vaccination. Lancet 2021, 397, e11. [Google Scholar] [CrossRef] [PubMed]
  12. Hippisley-Cox, J.; Patone, M.; Mei, X.W.; Saatci, D.; Dixon, S.; Khunti, K.; Zaccardi, F.; Watkinson, P.; Shankar-Hari, M.; Doidge, J.; et al. Risk of thrombocytopenia and thromboembolism after covid-19 vaccination and SARS-CoV-2 positive testing: Self-controlled case series study. BMJ 2021, 374, n1931. [Google Scholar] [CrossRef]
  13. Wise, J. Covid-19: European countries suspend use of Oxford-AstraZeneca vaccine after reports of blood clots. BMJ 2021, 372, n699. [Google Scholar] [CrossRef] [PubMed]
  14. Song, P.; Xu, Y.; Zha, M.; Zhang, Y.; Rudan, I. Global epidemiology of retinal vein occlusion: A systematic review and meta—Analysis of prevalence, incidence, and risk factors. J. Glob. Health 2019, 9, 010427. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, R.; Dong, L.; Fu, X.; Hua, L.; Zhou, W.; Li, H.; Wu, H.; Yu, C.; Li, Y.; Shi, X.; et al. Trends in the Prevalence of Common Retinal and Optic Nerve Diseases in China: An Artificial Intelligence Based National Screening. Transl. Vis. Sci. Technol. 2024, 13, 28. [Google Scholar] [CrossRef]
  16. Scott, I.U.; Campochiaro, P.A.; Newman, N.J.; Biousse, V. Retinal vascular occlusions. Lancet 2020, 396, 1927–1940. [Google Scholar] [CrossRef]
  17. Romano, F.; Lamanna, F.; Gabrielle, P.H.; Teo, K.Y.C.; Battaglia Parodi, M.; Iacono, P.; Fraser-Bell, S.; Cornish, E.E.; Nassisi, M.; Viola, F.; et al. Update on Retinal Vein Occlusion. Asia Pac. J. Ophthalmol. 2023, 12, 196–210. [Google Scholar] [CrossRef]
  18. Ørskov, M.; Vorum, H.; Bjerregaard Larsen, T.; Vestergaard, N.; Lip, G.Y.H.; Bek, T.; Skjøth, F. A review of risk factors for retinal vein occlusions. Expert Rev. Cardiovasc. Ther. 2022, 20, 761–772. [Google Scholar] [CrossRef]
  19. Rehak, M.; Wiedemann, P. Retinal vein thrombosis: Pathogenesis and management. J. Thromb. Haemost. 2010, 8, 1886–1894. [Google Scholar] [CrossRef]
  20. Park, S.J.; Choi, N.K.; Seo, K.H.; Park, K.H.; Woo, S.J. Nationwide incidence of clinically diagnosed central retinal artery occlusion in Korea, 2008 to 2011. Ophthalmology 2014, 121, 1933–1938. [Google Scholar] [CrossRef]
  21. Kim, Y.; Han, K.; Kim, J.H. Retinal Vascular Occlusions After COVID-19 Vaccination in South Korea: A Nation-Wide Population-Based Study. Ophthalmic Epidemiol. 2025, 32, 403–411. [Google Scholar] [CrossRef] [PubMed]
  22. Hwang, S.; Kang, S.W.; Choi, J.; Park, K.A.; Lim, D.H.; Shin, J.Y.; Kang, D.; Cho, J.; Kim, S.J. COVID-19 Vaccination and Ocular Adverse Events: A Self-Controlled Case Series Study From the Entire South Korean Population. Am. J. Ophthalmol. 2025, 269, 69–77. [Google Scholar] [CrossRef]
  23. Park, H.S.; Lee, N.K.; Lee, C.S.; Byeon, S.H.; Kim, S.S.; Lee, S.W.; Kim, Y.J. Retinal Artery and Vein Occlusion Risks after Coronavirus Disease 2019 or Coronavirus Disease 2019 Vaccination. Ophthalmology 2024, 131, 322–332. [Google Scholar] [CrossRef] [PubMed]
  24. Li, J.X.; Wang, Y.H.; Bair, H.; Hsu, S.B.; Chen, C.; Wei, J.C.; Lin, C.J. Risk assessment of retinal vascular occlusion after COVID-19 vaccination. npj Vaccines 2023, 8, 64. [Google Scholar] [CrossRef]
  25. Dorney, I.; Shaia, J.; Kaelber, D.C.; Talcott, K.E.; Singh, R.P. Risk of New Retinal Vascular Occlusion After mRNA COVID-19 Vaccination Within Aggregated Electronic Health Record Data. JAMA Ophthalmol. 2023, 141, 441–447. [Google Scholar] [CrossRef]
  26. Pellegrini, M.; Carnevali, A.; Fiore, T.; Cagini, C.; De Palma, A.; Fontana, L.; Lupardi, E.; Cassini, F.; Bacherini, D.; Giansanti, F.; et al. Risk of retinal vein occlusion following COVID-19 vaccination: A self-controlled case series. Eye 2023, 37, 3000–3003. [Google Scholar] [CrossRef]
  27. Feltgen, N.; Ach, T.; Ziemssen, F.; Quante, C.S.; Gross, O.; Abdin, A.D.; Aisenbrey, S.; Bartram, M.C.; Blum, M.; Brockmann, C.; et al. Retinal Vascular Occlusion after COVID-19 Vaccination: More Coincidence than Causal Relationship? Data from a Retrospective Multicentre Study. J. Clin. Med. 2022, 11, 5101. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Hashimoto, Y.; Yamana, H.; Iwagami, M.; Ono, S.; Takeuchi, Y.; Michihata, N.; Uemura, K.; Yasunaga, H.; Aihara, M.; Kaburaki, T. Ocular Adverse Events after Coronavirus Disease 2019 mRNA Vaccination: Matched Cohort and Self-Controlled Case Series Studies Using a Large Database. Ophthalmology 2023, 130, 256–264. [Google Scholar] [CrossRef]
  29. Uzun, G.; Ringelmann, T.; Hammer, S.; Zlamal, J.; Luz, B.; Wolf, M.E.; Henkes, H.; Bakchoul, T.; Althaus, K. Subsequent Vaccination against SARS-CoV-2 after Vaccine-Induced Immune Thrombotic Thrombocytopenia. J. Clin. Med. 2024, 13, 5462. [Google Scholar] [CrossRef]
  30. Cai, Z.; Yarovoi, S.V.; Zhu, Z.; Rauova, L.; Hayes, V.; Lebedeva, T.; Liu, Q.; Poncz, M.; Arepally, G.; Cines, D.B.; et al. Atomic description of the immune complex involved in heparin-induced thrombocytopenia. Nat. Commun. 2015, 6, 8277. [Google Scholar] [CrossRef]
  31. Files, J.C.; Malpass, T.W.; Yee, E.K.; Ritchie, J.L.; Harker, L.A. Studies of human platelet α—Granule release in vivo. Blood 1981, 58, 607–618. [Google Scholar] [CrossRef]
  32. Mayo, K.H.; Ilyina, E.; Roongta, V.; Dundas, M.; Joseph, J.; Lai, C.K.; Maione, T.; Daly, T.J. Heparin binding to platelet factor-4. An NMR and site-directed mutagenesis study: Arginine residues are crucial for binding. Biochem. J. 1995, 312, 357–365. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, Z.; Li, L.; Zhang, H.; Pang, X.; Qiu, Z.; Xiang, Q.; Cui, Y. Platelet factor 4(PF4) and its multiple roles in diseases. Blood Rev. 2024, 64, 101155. [Google Scholar] [CrossRef]
  34. Schultz, N.H.; Sørvoll, I.H.; Michelsen, A.E.; Munthe, L.A.; Lund-Johansen, F.; Ahlen, M.T.; Wiedmann, M.; Aamodt, A.H.; Skattør, T.H.; Tjønnfjord, G.E.; et al. Thrombosis and Thrombocytopenia after ChAdOx1 nCoV-19 Vaccination. N. Engl. J. Med. 2021, 384, 2124–2130. [Google Scholar] [CrossRef]
  35. Greinacher, A.; Schönborn, L.; Siegerist, F.; Steil, L.; Palankar, R.; Handtke, S.; Reder, A.; Thiele, T.; Aurich, K.; Methling, K.; et al. Pathogenesis of vaccine-induced immune thrombotic thrombocytopenia (VITT). Semin. Hematol. 2022, 59, 97–107. [Google Scholar] [CrossRef] [PubMed]
  36. Baker, A.T.; Boyd, R.J.; Sarkar, D.; Teijeira-Crespo, A.; Chan, C.K.; Bates, E.; Waraich, K.; Vant, J.; Wilson, E.; Truong, C.D.; et al. ChAdOx1 interacts with CAR and PF4 with implications for thrombosis with thrombocytopenia syndrome. Sci. Adv. 2021, 7, eabl8213. [Google Scholar] [CrossRef]
  37. Othman, M.; Labelle, A.; Mazzetti, I.; Elbatarny, H.S.; Lillicrap, D. Adenovirus-induced thrombocytopenia: The role of von Willebrand factor and P-selectin in mediating accelerated platelet clearance. Blood 2007, 109, 2832–2839. [Google Scholar] [CrossRef] [PubMed]
  38. Nicolai, L.; Leunig, A.; Pekayvaz, K.; Esefeld, M.; Anjum, A.; Rath, J.; Riedlinger, E.; Ehreiser, V.; Mader, M.; Eivers, L.; et al. Thrombocytopenia and splenic platelet-directed immune responses after IV ChAdOx1 nCov-19 administration. Blood 2022, 140, 478–490. [Google Scholar] [CrossRef]
  39. Mittra, I.; Nair, N.K.; Mishra, P.K. Nucleic acids in circulation: Are they harmful to the host? J. Biosci. 2012, 37, 301–312. [Google Scholar] [CrossRef]
  40. Greinacher, A.; Selleng, K.; Palankar, R.; Wesche, J.; Handtke, S.; Wolff, M.; Aurich, K.; Lalk, M.; Methling, K.; Völker, U.; et al. Insights in ChAdOx1 nCoV-19 vaccine-induced immune thrombotic thrombocytopenia. Blood 2021, 138, 2256–2268. [Google Scholar] [CrossRef]
  41. Thiele, T.; Ulm, L.; Holtfreter, S.; Schönborn, L.; Kuhn, S.O.; Scheer, C.; Warkentin, T.E.; Bröker, B.M.; Becker, K.; Aurich, K.; et al. Frequency of positive anti-PF4/polyanion antibody tests after COVID-19 vaccination with ChAdOx1 nCoV-19 and BNT162b2. Blood 2021, 138, 299–303. [Google Scholar] [CrossRef] [PubMed]
  42. Kowarz, E.; Krutzke, L.; Külp, M.; Streb, P.; Larghero, P.; Reis, J.; Bracharz, S.; Engler, T.; Kochanek, S.; Marschalek, R. Vaccine-induced COVID-19 mimicry syndrome. eLife 2022, 11, e74974. [Google Scholar] [CrossRef]
  43. Yonker, L.M.; Swank, Z.; Bartsch, Y.C.; Burns, M.D.; Kane, A.; Boribong, B.P.; Davis, J.P.; Loiselle, M.; Novak, T.; Senussi, Y.; et al. Circulating Spike Protein Detected in Post-COVID-19 mRNA Vaccine Myocarditis. Circulation 2023, 147, 867–876. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, H.; Penninger, J.M.; Li, Y.; Zhong, N.; Slutsky, A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS—CoV—2 receptor: Molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020, 46, 586–590. [Google Scholar] [CrossRef]
  45. Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Del Pozo, C.H.; Prosper, F.; et al. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell 2020, 181, 905–913.e7. [Google Scholar] [CrossRef] [PubMed]
  46. Hagemann, K.; Riecken, K.; Jung, J.M.; Hildebrandt, H.; Menzel, S.; Bunders, M.J.; Fehse, B.; Koch-Nolte, F.; Heinrich, F.; Peine, S.; et al. Natural killer cell-mediated ADCC in SARS-CoV-2-infected individuals and vaccine recipients. Eur. J. Immunol. 2022, 52, 1297–1307. [Google Scholar] [CrossRef]
  47. Zedan, H.T.; Smatti, M.K.; Al-Sadeq, D.W.; Al Khatib, H.A.; Nicolai, E.; Pieri, M.; Bernardini, S.; Hssain, A.A.; Taleb, S.; Qotba, H.; et al. SARS-CoV-2 infection triggers more potent antibody-dependent cellular cytotoxicity (ADCC) responses than mRNA-, vector-, and inactivated virus-based COVID-19 vaccines. J. Med. Virol. 2024, 96, e29527. [Google Scholar] [CrossRef]
  48. Lamerton, R.E.; Marcial-Juarez, E.; Faustini, S.E.; Perez-Toledo, M.; Goodall, M.; Jossi, S.E.; Newby, M.L.; Chapple, I.; Dietrich, T.; Veenith, T.; et al. SARS-CoV-2 Spike- and Nucleoprotein-Specific Antibodies Induced After Vaccination or Infection Promote Classical Complement Activation. Front. Immunol. 2022, 13, 838780. [Google Scholar] [CrossRef]
  49. Terentes-Printzios, D.; Gardikioti, V.; Solomou, E.; Emmanouil, E.; Gourgouli, I.; Xydis, P.; Christopoulou, G.; Georgakopoulos, C.; Dima, I.; Miliou, A.; et al. The effect of an mRNA vaccine against COVID-19 on endothelial function and arterial stiffness. Hypertens. Res. 2022, 45, 846–855. [Google Scholar] [CrossRef] [PubMed]
  50. Manne, B.K.; Denorme, F.; Middleton, E.A.; Portier, I.; Rowley, J.W.; Stubben, C.; Petrey, A.C.; Tolley, N.D.; Guo, L.; Cody, M.; et al. Platelet gene expression and function in patients with COVID-19. Blood 2020, 136, 1317–1329. [Google Scholar] [CrossRef] [PubMed]
  51. Liao, Z.; Wang, C.; Tang, X.; Yang, M.; Duan, Z.; Liu, L.; Lu, S.; Ma, L.; Cheng, R.; Wang, G.; et al. Human transferrin receptor can mediate SARS-CoV-2 infection. Proc. Natl. Acad. Sci. USA 2024, 121, e2317026121. [Google Scholar] [CrossRef]
  52. Armstrong, J.K. The occurrence, induction, specificity and potential effect of antibodies against poly(ethylene glycol). In PEGylated Protein Drugs: Basic Science and Clinical Applications; Veronese, F.M., Ed.; Birkhäuser Verlag: Basel, Switzerland, 2009. [Google Scholar]
  53. Richter, A.W.; Akerblom, E. Polyethylene glycol reactive antibodies in man: Titer distribution in allergic patients treated with monomethoxy polyethylene glycol modified allergens or placebo, and in healthy blood donors. Int. Arch. Allergy Appl. Immunol. 1984, 74, 36–39. [Google Scholar] [CrossRef]
  54. Reinke, S.; Thakur, A.; Gartlan, C.; Bezbradica, J.S.; Milicic, A. Inflammasome-mediated immunogenicity of clinical and experimental vaccine adjuvants. Vaccines 2020, 8, 554. [Google Scholar] [CrossRef]
  55. Teijaro, J.R.; Farber, D.L. COVID-19 vaccines: Modes of immune activation and future challenges. Nat. Rev. Immunol. 2021, 21, 195–197. [Google Scholar] [CrossRef]
  56. Zhao, X.; Cheng, J.; Gui, S.; Jiang, M.; Qi, D.; Huang, J.; Fu, L.; Liu, S.; Ma, Y.; Shi, J.; et al. Amifostine-Loaded Nanocarrier Traverses the Blood-Brain Barrier and Prevents Radiation-Induced Brain Injury. ACS Appl. Mater. Interfaces 2023, 15, 15203–15219. [Google Scholar] [CrossRef]
  57. Greenhawt, M.; Shaker, M.; Golden, D.B.K.; Abrams, E.M.; Blumenthal, K.G.; Wolfson, A.R.; Stone, C.A., Jr.; Krantz, M.S.; Chu, D.K.; Dwamena, B.A. Diagnostic accuracy of vaccine and vaccine excipient testing in the setting of allergic reactions to COVID-19 vaccines: A systematic review and meta-analysis. Allergy 2023, 78, 71–83. [Google Scholar] [CrossRef]
  58. Azzarone, B.; Veneziani, I.; Moretta, L.; Maggi, E. Pathogenic Mechanisms of Vaccine-Induced Immune Thrombotic Thrombocytopenia in People Receiving Anti-COVID-19 Adenoviral-Based Vaccines: A Proposal. Front. Immunol. 2021, 12, 728513. [Google Scholar] [CrossRef]
  59. Li, Z.; Guo, J.; Bi, L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed. Pharmacother. 2020, 130, 110542. [Google Scholar] [CrossRef]
  60. Cabanillas, B.; Akdis, C.A.; Novak, N. Allergic reactions to the first COVID-19 vaccine: A potential role of polyethylene glycol? Allergy 2021, 76, 1617–1618. [Google Scholar] [CrossRef]
  61. Bakos, T.; Mészáros, T.; Kozma, G.T.; Berényi, P.; Facskó, R.; Farkas, H.; Dézsi, L.; Heirman, C.; de Koker, S.; Schiffelers, R.; et al. mRNA-LNP COVID-19 Vaccine Lipids Induce Complement Activation and Production of Proinflammatory Cytokines: Mechanisms, Effects of Complement Inhibitors, and Relevance to Adverse Reactions. Int. J. Mol. Sci. 2024, 25, 3595. [Google Scholar] [CrossRef] [PubMed]
  62. Leung, H.M.; Au, S.C. Retinal Vein Occlusion after COVID-19 Vaccination—A Review. Vaccines 2023, 11, 1281. [Google Scholar] [CrossRef] [PubMed]
  63. Krantz, M.S.; Liu, Y.; Phillips, E.J.; Stone, C.A., Jr. COVID-19 vaccine anaphylaxis: PEG or not? Allergy 2021, 76, 1934–1937. [Google Scholar] [CrossRef] [PubMed]
  64. Borgsteede, S.D.; Geersing, T.H.; Tempels-Pavlica, Ž. Other excipients than PEG might cause serious hypersensitivity reactions in COVID-19 vaccines. Allergy 2021, 76, 1941–1942. [Google Scholar] [CrossRef]
  65. Ndeupen, S.; Qin, Z.; Jacobsen, S.; Bouteau, A.; Estanbouli, H.; Igyarto, B.Z. The mRNA-LNP platform’s lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. iScience 2021, 24, 103479. [Google Scholar] [CrossRef]
  66. Springer, J.; Villa-Forte, A. Thrombosis in vasculitis. Curr. Opin. Rheumatol. 2013, 25, 19–25. [Google Scholar] [CrossRef]
  67. Safary, A.; Akbarzadeh-Khiavi, M.; Barar, J.; Omidi, Y. SARS-CoV-2 vaccine-triggered autoimmunity: Molecular mimicry and/or bystander activation of the immune system. BioImpacts BI 2023, 13, 269–273. [Google Scholar] [CrossRef]
  68. Talotta, R. Molecular Mimicry and HLA Polymorphisms May Drive Autoimmunity in Recipients of the BNT-162b2 mRNA Vaccine: A Computational Analysis. Microorganisms 2023, 11, 1686. [Google Scholar] [CrossRef]
  69. Du, Y.; Yan, B. Ocular immune privilege and retinal pigment epithelial cells. J. Leukoc. Biol. 2023, 113, 288–304. [Google Scholar] [CrossRef] [PubMed]
  70. Gedik, B.; Erol, M.K.; Suren, E.; Yavuz, S.; Kucuk, M.F.; Bozdogan, Y.C.; Ekinci, R.; Akidan, M. Evaluation of retinal and optic disc vascular structures in individuals before and after Pfizer-BioNTech vaccination. Microvasc. Res. 2023, 147, 104500. [Google Scholar] [CrossRef] [PubMed]
  71. Gedik, B.; Bozdogan, Y.C.; Yavuz, S.; Durmaz, D.; Erol, M.K. The assesment of retina and optic disc vascular structures in people who received CoronaVac vaccine. Photodiagn. Photodyn. Ther. 2022, 38, 102742. [Google Scholar] [CrossRef] [PubMed]
  72. Sarıtaş, Ö.; Yorgun, M.A.; Gökpınar, E. Effects of Sinovac-Coronavac and Pfizer-BioNTech mRNA vaccines on choroidal and retinal vascular system. Photodiagn. Photodyn. Ther. 2023, 43, 103702. [Google Scholar] [CrossRef]
  73. Greinacher, A.; Selleng, K.; Mayerle, J.; Palankar, R.; Wesche, J.; Reiche, S.; Aebischer, A.; Warkentin, T.E.; Muenchhoff, M.; Hellmuth, J.C.; et al. Anti-platelet factor 4 antibodies causing VITT do not cross-react with SARS-CoV-2 spike protein. Blood 2021, 138, 1269–1277. [Google Scholar] [CrossRef]
  74. Wang, J.J.; Schönborn, L.; Warkentin, T.E.; Chataway, T.; Grosse, L.; Simioni, P.; Moll, S.; Greinacher, A.; Gordon, T.P. Antibody Fingerprints Linking Adenoviral Anti-PF4 Disorders. N. Engl. J. Med. 2024, 390, 1827–1829. [Google Scholar] [CrossRef] [PubMed]
  75. Faksova, K.; Walsh, D.; Jiang, Y.; Griffin, J.; Phillips, A.; Gentile, A.; Kwong, J.C.; Macartney, K.; Naus, M.; Grange, Z.; et al. COVID-19 vaccines and adverse events of special interest: A multinational Global Vaccine Data Network (GVDN) cohort study of 99 million vaccinated individuals. Vaccine 2024, 42, 2200–2211. [Google Scholar] [CrossRef] [PubMed]
  76. Weirauch, T.; Schüttfort, G.; Vehreschild, M.J.G.T. Syncopes, paresis and loss of vision after COVID-19 mRNA-based vaccination and SARS-CoV-2 infection. Infection 2025, 53, 741–746. [Google Scholar] [CrossRef]
  77. Kamoi, K.; Ohno-Matsui, K. Long Vax in the Eye: Long Post-COVID Vaccination Syndrome Presenting with Frosted Branch Angiitis. Diseases 2024, 12, 36. [Google Scholar] [CrossRef]
  78. Finsterer, J. A Case Report: Long Post-COVID Vaccination Syndrome During the Eleven Months After the Third Moderna Dose. Cureus 2022, 14, e32433. [Google Scholar] [CrossRef]
  79. Wang, J.J.; Warkentin, T.E.; Schönborn, L.; Wheeler, M.B.; Geerts, W.H.; Costedoat-Chalumeau, N.; Gendron, N.; Ene, G.; Lozano, M.; Langer, F.; et al. VITT-like Monoclonal Gammopathy of Thrombotic Significance. N. Engl. J. Med. 2025, 392, 995–1005. [Google Scholar] [CrossRef]
  80. Warkentin, T.E. Classification of Platelet-Activating Anti-Platelet Factor 4 Disorders. Int. J. Lab. Hematol, early view. 2025. [Google Scholar]
  81. Borriello, F.; Poli, V.; Shrock, E.; Spreafico, R.; Liu, X.; Pishesha, N.; Carpenet, C.; Chou, J.; Di Gioia, M.; McGrath, M.E.; et al. An adjuvant strategy enabled by modulation of the physical properties of microbial ligands expands antigen immunogenicity. Cell 2022, 185, 614–629.e21. [Google Scholar] [CrossRef]
  82. Vakili, B.; Karami-Darehnaranji, M.; Mirzaei, E.; Hosseini, F.; Nezafat, N. Graphene oxide as novel vaccine adjuvant. Int. Immunopharmacol. 2023, 125, 111062. [Google Scholar] [CrossRef]
  83. Rahman, T.; Das, A.; Abir, M.H.; Nafiz, I.H.; Mahmud, A.R.; Sarker, M.R.; Emran, T.B.; Hassan, M.M. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine 2023, 169, 156268. [Google Scholar] [CrossRef]
Figure 1. Arrows denote causal directions (exposure→mediator→outcome). Four confounder groups (demographics/pre-existing conditions, indicated by purple boxes) affect both vaccine exposure (indicated by blue boxes) and RVO risk. Unmeasured variables (dashed boxes) indicate residual confounding. Green boxes represent possible pathologicalmechanisms.Excluded factors were intentionally controlled in some of studies. DAG guides bias adjustment and mechanistic analysis.
Figure 1. Arrows denote causal directions (exposure→mediator→outcome). Four confounder groups (demographics/pre-existing conditions, indicated by purple boxes) affect both vaccine exposure (indicated by blue boxes) and RVO risk. Unmeasured variables (dashed boxes) indicate residual confounding. Green boxes represent possible pathologicalmechanisms.Excluded factors were intentionally controlled in some of studies. DAG guides bias adjustment and mechanistic analysis.
Vaccines 13 00733 g001
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

Zhang, Y.; Zhang, H.; Lv, K.; Lin, X.; Chen, F.; Cao, H.; Chen, C. Retinal Vascular Occlusion Following COVID-19 Vaccination: A Comprehensive Review of Observational Study and Pathophysiological Mechanisms. Vaccines 2025, 13, 733. https://doi.org/10.3390/vaccines13070733

AMA Style

Zhang Y, Zhang H, Lv K, Lin X, Chen F, Cao H, Chen C. Retinal Vascular Occlusion Following COVID-19 Vaccination: A Comprehensive Review of Observational Study and Pathophysiological Mechanisms. Vaccines. 2025; 13(7):733. https://doi.org/10.3390/vaccines13070733

Chicago/Turabian Style

Zhang, Yuchen, Haoliang Zhang, Kangjia Lv, Xin Lin, Feng’e Chen, Hui Cao, and Chong Chen. 2025. "Retinal Vascular Occlusion Following COVID-19 Vaccination: A Comprehensive Review of Observational Study and Pathophysiological Mechanisms" Vaccines 13, no. 7: 733. https://doi.org/10.3390/vaccines13070733

APA Style

Zhang, Y., Zhang, H., Lv, K., Lin, X., Chen, F., Cao, H., & Chen, C. (2025). Retinal Vascular Occlusion Following COVID-19 Vaccination: A Comprehensive Review of Observational Study and Pathophysiological Mechanisms. Vaccines, 13(7), 733. https://doi.org/10.3390/vaccines13070733

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