Next Article in Journal
Exploratory Evaluation of Pre-Treatment Inflammation Profiles in Patients with Colorectal Cancer
Previous Article in Journal
Collateral Damage in the Placenta during Viral Infection in Pregnancy: A Possible Mechanism for Vertical Transmission and an Adverse Pregnancy Outcome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clinical Research into Central Nervous System Inflammatory Demyelinating Diseases Related to COVID-19 Vaccines

1
Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine, Taoyuan 333, Taiwan
2
Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
3
Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine, Taoyuan 333, Taiwan
4
Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, Chang Gung University, New Taipei 236, Taiwan
5
Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, Taipei 110, Taiwan
6
Brain and Consciousness Research Center, Shuang Ho Hospital, New Taipei 235, Taiwan
7
Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 300, Taiwan
*
Author to whom correspondence should be addressed.
Diseases 2024, 12(3), 60; https://doi.org/10.3390/diseases12030060
Submission received: 4 February 2024 / Revised: 15 March 2024 / Accepted: 18 March 2024 / Published: 20 March 2024

Abstract

:
Various vaccines have been developed in response to the SARS-CoV-2 pandemic, and the safety of vaccines has become an important issue. COVID-19 vaccine-related central nervous system inflammatory demyelinating diseases (CNS IDDs) have been reported recently. We present one case of AstraZeneca vaccine-related myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease and a literature review of another 78 patients published from January 2020 to October 2022. Patients were divided into three vaccine types (viral vector, mRNA, and inactivated vaccines) for further analyses. Among 79 patients with COVID-19 vaccine-related CNS IDDs, 49 (62%) cases received viral vector vaccines, 20 (25.3%) received mRNA vaccines, and 10 (12.7%) received inactivated vaccines. Twenty-seven cases (34.2%) were confirmed with autoantibodies, including fifteen patients (19%) with anti-MOG, eleven (13.9%) with anti-aquaporin 4 (AQP4), and one (1.3%) with both antibodies. Significantly, more males developed CNS IDDs post viral vector vaccines compared to mRNA and inactivated vaccines. Patients receiving mRNA vaccines were older than those receiving other types. Furthermore, mRNA and inactivated vaccines correlated more with anti-AQP4 antibodies, while viral vector vaccines showed higher MOG positivity. This research suggests potential associations between COVID-19 vaccine-related CNS IDDs and gender, age, and autoantibodies, contingent on vaccine types. Protein sequence analysis implies similarities between the S protein and AQP4/MOG. Further studies may elucidate the mechanisms of CNS IDDs, aiding vaccine selection for specific types.

1. Introduction

Diseases of myelin sheaths in the central nervous system (CNS) can be divided into two categories: genetic dysmyelinating diseases with abnormal myelin formation and acquired inflammatory demyelinating diseases, or so-called central nervous system inflammatory demyelinating diseases (CNS IDDs). CNS IDDs include multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSDs), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), acute disseminated encephalomyelitis (ADEM), optic neuritis (ON), and transverse myelitis (TM) [1].
NMOSDs and MOGAD are both related to autoantibodies, which target aquaporin 4 (AQP4) and myelin oligodendrocyte glycoprotein (MOG), respectively. The incidence and prevalence of NMOSDs is about 0.28–0.73 per 100,000 person-years and 0.52–10 per 100,000 people, respectively [2]. The variability in epidemiological data on NMOSDs may be attributed to differences in study designs, geographical regions, and ethnicities. NMOSDs are more common in East Asian, African, and Latin American populations compared to other Western populations, and the prevalence is 2.3 to 7.6 times higher in women than in men [3]. Previous studies in Europe estimated that the incidence of MOGAD is around 0.16–0.34 per 100,000 person-years, and the prevalence is 2 per 100,000 people [4,5]. The median age of onset is from the early- to mid-thirties with a slight predominance in females (57–68%) [6,7,8]. The clinical manifestations of MOGAD are diverse and can include one or a combination of the following diseases: ON, TM, and ADEM [9]. ON was the most common symptom (54–68.5%), followed by TM (27–30%) and ADEM or ADEM-like presentation (18–25%) [6,8,10]. The initial presentation of MOGAD in children and adults was also found to be different: ADEM was most common in children and ON was primarily found in adults [7].
ADEM is characterized by monophasic multifocal neurologic symptoms, and its diagnosis requires exclusion of MS, NMOSDs, MOGAD, or other demyelinating diseases [11]. ADEM is primarily regarded as a post-infectious disease, whereas vaccine-related ADEM is a rare condition. According to a prior systematic review, determining the incidence of CNS IDDs in confirmed COVID-19 infections posed challenges [12]. In the Taiwan Centers for Disease Control and Prevention (CDC) reporting system, it is also not possible to obtain the exact incidence of CNS IDD after COVID infection. Furthermore, there is a need for clarification regarding the severity and treatment response of CNS IDDs following COVID-19 infection compared to those without the virus [12]. It has been published that <5% of ADEM cases are related to vaccination for diseases such as rabies, measles, mumps, smallpox, or Japanese B encephalitis [13]. However, a recent study in the United States reported that there were no statistically significantly increased risks of ADEM after vaccination for 5–28 days, except for tetanus, reduced diphtheria, and acellular pertussis (Tdap) [14]. Another study provided additional evidence indicating no association between ADEM and different vaccine types, including Tdap [15]. The controversial relationship between vaccination and ADEM remains ambiguous, and large-scale epidemiologic data and clinical studies are required to confirm their association.
During the outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, vaccines were one of the most effective ways to prevent infection. The mechanisms of COVID-19 vaccines include viral vectors (AstraZeneca, Janssen, Sputnik V, CanSino), mRNA (Pfizer-BioNTech and Moderna), inactivated vaccines (Sinovac, Sinopharm, Covaxin), and protein subunit vaccines (Medigen). Various side effects of COVID-19 vaccines have been widely reported recently. Although the distinct mechanism has not yet been elucidated, some adverse events may be related to specific types of vaccines. For example, vaccine-induced immune thrombotic thrombocytopenia (VITT) and Guillain–Barre syndrome are generally considered to be related to viral vector COVID-19 vaccines [16,17,18,19]. An elevated risk of myocarditis was observed in a population of young men who received mRNA COVID-19 vaccines [20]. CNS IDDs, such as TM, NMOSD, ADEM, or ON, have been reported after receiving AstraZeneca [21,22,23,24,25,26,27,28], Janssen [29], Sputnik V [30], Pfizer-BioNTech [31,32,33,34,35], Moderna [36,37,38,39,40], Sinovac [41,42], and Sinopharm [43,44] vaccines and an unknown inactivated vaccine [45].
We speculate that COVID-19 vaccines may induce CNS IDDs in a small subset of the population. Additionally, different types of COVID-19 vaccines may lead to different clinical manifestations, laboratory, or imaging characteristics of CNS IDDs. We now present a patient who developed MOGAD following COVID-19 vaccination and recruitment of relevant cases from the updated literature for comparison and analysis.

2. Materials and Methods

2.1. Case Report

Clinical features, brain and spinal MRIs, and laboratory data were obtained from the index case’s medical chart records.

2.2. Literature Review

To analyze the clinical presentations and MRI findings of patients with CNS IDDs after COVID-19 vaccination, a literature review was conducted using PubMed, EMBASE, Google Scholar, Ovid, and SCOPUS. Published articles and preprints of cases between January 2020 and October 2022 were included to obtain sufficient data, particularly brain and spinal MR imaging. The Medical Subject Heading (MeSH) terms “acute disseminated encephalomyelitis (ADEM)”, “myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD)”, “neuromyelitis optica spectrum disorders (NMOSDs)”, “optic neuritis”, and “myelitis” with “COVID-19 vaccines” were used to retrieve all accessible articles. We aimed to identify cases with first onset of CNS IDDs related to COVID-19 vaccination. Those diagnosed with relapses of MS, SARS-CoV-2 infection, Guillain–Barre syndrome, and CNS infection were excluded. We recorded the clinical features, past history, laboratory findings, brain/spinal MR imaging, treatment, and outcomes of our index patient in addition to previously reported cases of CNS IDDs after receiving COVID-19 vaccines. The lesions on brain MRIs were classified as cortical, deep gray matters, white matters (periventricular, subcortical, periaqueductal), brainstem, and with or without gadolinium enhancement. The lesions on spinal MRIs were divided into long-cord (≥3 segments) or short-cord (<3 segments), and with or without gadolinium enhancement.
In total, 79 cases (including our index case of MOGAD) diagnosed with CNS IDDs after COVID-19 vaccination were identified across 45 articles [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] (Table 1). Among them, the proportion of females (54.4%) was higher than that of males (45.6%), with average ages of 46.2 and 48.5 (p = 0.524), respectively. The majority of patients developed symptoms after the first dose (77.2%) or second dose (21.5%) of the COVID-19 vaccine, while one patient (1.3%) developed CNS IDDs after the third dose. The mean time of onset from vaccination to clinical symptoms was approximately 12.2 days (range 1–42 days). Twenty-one patients (26.6%) presented with ADEM or ADEM-like symptoms, nineteen cases (24.1%) with myelitis, fifteen (19%) with MOGAD, thirteen (16.5%) with NMOSDs, ten (12.7%) with ADEM with myelitis, and one (1.3%) with ON. A total of 27 patients (34.2%) had autoantibodies, including 15 patients (19%) with anti-MOG antibodies, 11 patients (13.9%) with anti-AQP4 antibodies, and 1 patient (1.3%) with both anti-MOG and anti-AQP4 antibodies. In the CSF examination of the 79 cases, the average WBC was 63.5 cells/µL with lymphocyte predominant, and the average total protein was 69.5 mg/dL. Additionally, oligoclonal bands were positive in 13 cases (16.5%).

2.3. Statistical Analysis

SPSS 26.0 (IBM Corp., Armonk, NY, USA) was used for statistical analyses. Categorical variables were compared using Pearson’s chi-square test or Fisher’s exact test. One-way analysis of variance (ANOVA) was used to compare unpaired groups. An independent samples t-test was applied for continuous variables. The level of significance was set at p < 0.05. All tests were two-tailed.

3. Results

3.1. Case Report

Our report describes a 50-year-old man who has chronic hepatitis B virus (HBV) disease and with no other comorbidities (Case No. 1 in Table 1). After receiving the first dose of the COVID-19 AZD1222 vaccine (AstraZeneca) via a 0.5 mL intramuscular injection into the deltoid muscle [66], the patient experienced a mild headache and general weakness that persisted for 2 days and then spontaneously subsided. On the 13th day following vaccination, he experienced an acute onset of a headache which was exacerbated upon waking up in the morning. In addition, fever, bilateral lower limb numbness, low back pain, confusion, slow response, unsteady gait, constipation, and urine retention requiring insertion of a Foley catheter developed over the following 4 days. Neurologic examination revealed Glasgow coma scale E4V5M6, but with episodic lethargy and obtundation. Impaired proprioception in the bilateral lower limbs leading to an ataxic gait and decreased pin-prick sensation below the T4 level were observed. Motor function and deep tendon reflexes were normal, and the Babinski reflex was a bilateral plantar flexion. Autonomic dysfunction led to constipation and urine retention. Cervical and thoracic spinal MRI revealed T2 hyperintensity in the spinal cord at the T3 to T4 levels without contrast enhancement, resembling focal myelitis (Figure 1a,c, 19th day post-vaccination). Brain MRI revealed symmetric, poorly demarcated hyperintensities involving the brainstem, bilateral pulvinar thalami, putamen, and centrum semiovale, with subcortical white matter fluid-attenuated inversion recovery (FLAIR) and T2-weighted imaging (T2WI) without gadolinium enhancement (Figure 2, 20th day post-vaccination). Blood tests revealed leukocytosis (11,000 cells/mL) with neutrophil predominance (85%) and elevated C-reactive protein (CRP) (48.49 mg/L). Urinalysis for intoxication was negative. Cerebral spinal fluid (CSF) analysis showed lymphocytic predominant pleocytosis (175 cells/µL, 99% lymphocytes), elevated total protein (78.1 mg/dL), and low CSF/serum glucose ratio (0.47).
He was administered empiric antibiotics (ceftriaxone and vancomycin) and antiviral (acyclovir) treatment initially, which were discontinued because results for bacterial culture and viral examination of CSF and blood were negative. HBV DNA was positive (439 IU/mL) and HBsAg was reactive (1484 IU/mL). Other laboratory results, including COVID-19 (nasopharyngeal swab for SARS-CoV-2-RT-PCR), Japanese encephalitis virus (JEV), herpes simplex virus (HSV), cytomegalovirus (CMV), Enterovirus, Epstein–Barr virus (EBV), varicella zoster virus (VZV), human immunodeficiency virus (HIV), cryptococcus, tuberculosis, syphilis, hepatitis C, and mycoplasma, were all negative. Further tests for vitamin B12 deficiency, vasculitis, connective tissue diseases, and tumor markers were unremarkable. Anti-AQP4 antibodies and oligoclonal bands were not found, but anti-MOG antibodies were detected in the serum by cell-based assays (CBA) using the commercially available kit (NMOSD Screen 1 EUROPattern, Euroimmun, Luebeck, Germany) (Figure 3). An indirect immunofluorescence test on HEK-293 cells transfected with plasmids containing myelin oligodendrocyte glycoprotein (MOG) was conducted. The initial 1:10 diluted serum was applied to transfected HEK-293 cells with plasmids containing MOG. Following a 30 min incubation, FITC-labeled secondary anti-human IgG antibody was added to the reaction field and incubated for an additional 30 min. Evaluation was performed using a fluorescence microscope. A positive antibody result was indicated by corresponding cytoplasmic and/or cell membrane smooth-to-fine-granular fluorescence in transfected HEK-293 cells (Figure 3b).
Pulse therapy with methylprednisolone (1 g per day for 5 consecutive days) was administered [67], followed by oral prednisolone (1 mg/kg/day) with gradual tapering. His consciousness and response became clear, and proprioception and pin-prick sensation gradually improved from the 2nd day after pulse therapy. The patient was discharged with total recovery of proprioception, pin-prick sensory level receding to T10 level, and improvement in constipation. The Foley catheter was successfully removed on the 38th day post-vaccination. Spinal MRI 5 months after vaccination revealed total resolution of T3 to T4 myelitis (Figure 1b,d). All neurological deficits have fully recovered. Anti-MOG antibodies in serum remained positive after 7 months of follow-up.

3.2. Literature Review

COVID-19 vaccines with different mechanisms of action may lead to different manifestations of CNS IDDs. These numbers and percentages reported here are derived solely from our index case and reviewed case reports, and do not represent the true incidence percentages in recipients of each type of vaccine. Among the 79 cases, we categorized the vaccines into three major vaccine types, including viral vector (49 cases, 62%), mRNA (20 cases, 25.3%), and inactivated vaccines (10 cases, 12.7%), to analyze their clinical presentations and laboratory examinations of CNS IDDs (Table 2). Compared to viral vector vaccines, CNS IDDs were more commonly observed in female patients receiving mRNA or inactivated vaccines (p = 0.027). The mean age of onset of patients receiving mRNA vaccines was much older than those receiving viral vector or inactivated vaccines (p = 0.002). The vast majority of patients in the viral vector vaccine type developed CNS IDDs after the first dose. Approximately two-thirds of the mRNA vaccine type experienced CNS IDDs after the first dose, and one-third after the second dose. In the inactivated vaccine type, there was one patient who developed CNS IDDs after the third dose. The time interval between vaccination and clinical presentations of CNS IDDs in patients receiving mRNA vaccines appears to be shorter than the other two types, although it is not statistically significant. There was no significant difference in the clinical presentations of CNS IDDs among these three types. Patients receiving mRNA or inactivated vaccines were more commonly found to have anti-AQP4 antibodies, while those receiving viral vector vaccines were frequently associated with anti-MOG antibodies (p = 0.044). On the other hand, a higher rate of CSF pleocytosis with lymphocyte predominant was observed in patients administered viral vector and mRNA vaccines (p = 0.004). Spinal cord lesions with gadolinium enhancement were most commonly found in patients who received mRNA vaccines (p = 0.015). There was no significant difference in the distribution of brain lesions or LETM (≥3 contiguous vertebral segments) between the three types. Among the 79 patients, 23 had documented past history, with hypertension and diabetes being the most common, each affecting patients (Table 1). The next most common diseases were hyperlipidemia and autoimmune diseases, each with four patients. Nevertheless, there was no statistically significant correlation between past history and the occurrence of CNS IDDs due to receiving different types of vaccines (data not presented in Table 2).
The majority of patients (86%) received first-line immunotherapy, including steroid pulse therapy, plasmapheresis/plasma exchange, or IVIG. Ten patients (13%) underwent additional second-line immunotherapy, such as cyclophosphamide or Rituximab. Among the 50 patients with available prognosis data, 3 individuals died (3.8%), while the remaining patients either fully recovered (20 patients) or showed improvement (27 patients).

3.3. Protein Sequence Analysis

By aligning proteins using protein sequence databases, it is possible that the S protein shares similar protein sequences with AQP4 and MOG (Figure 4).

4. Discussion

During the global SARS-CoV-2 pandemic, universal vaccination was a key factor in preventing infection and avoiding critical illness [68]. The overall safety and tolerability of COVID-19 vaccines has been shown to be generally acceptable in patients with underlying CNS IDDs, but there were still some patients (16.7%) who reported worsening of their symptoms after vaccination during the first week, with rapid resolution within 3 days [69]. The cases we reviewed in our study resulted from exposure to COVID-19 vaccines and they did not have a past history of CNS IDDs. The decision whether to receive the second or booster shots of COVID-19 vaccines, or to shift to another kind of vaccine, are important clinical issues.
According to statistics from the Centers for Disease Control in Taiwan [70], as of April 2023, the COVID-19 vaccine coverage rate was 94% for the first dose, 89% for the second dose, and 76.7% for additional doses. Most people received mRNA or viral vector vaccines, including Moderna (42.7%), Pfizer–BioNTech (29.2%), AstraZeneca (22.5%), Medigen (4.5%, a type of protein subunit vaccine produced in Taiwan), and Novavax (0.9%). Various adverse effects were reported. Six cases of ADEM (not included our index case), two cases of NMOSD, thirteen cases of ON, and four myelitis cases suspected to be related to COVID-19 vaccination were reported on the Taiwan CDC website, but not formally published [70]. Thrombosis with thrombocytopenia syndrome (TTS, also known as VITT) was suspected in 74 cases (86% of them received AstraZeneca, 8% received Moderna, 4% received Pfizer, and 1% received Medigen) [70]. In Taiwan, the first case of VITT after AstraZeneca vaccination was diagnosed in May 2021 at Chang Gung Memorial Hospital, a tertiary medical center. The patient presented with severe headaches, thrombocytopenia, and abdominal pain without neurological deficit. Brain CT revealed lacunar infarction in the right centrum semiovale. His platelet count normalized after treatment with high-dose IVIG (2 g/kg for two consecutive days) and he was discharged without further thrombotic or hemorrhagic events [71].
Our study provides a detailed case presentation of typical MOGAD clinical manifestations after receiving the AstraZeneca COVID-19 vaccine. A comprehensive comparison was conducted with previously published case reports. All 79 cases were excluded for concurrent COVID-19 infection. Since this complication of CNS IDDs following COVID-19 vaccination remains rare, the precise incidence rate has not yet been specified in pharmaceutical product documentation or registries, such as the Vaccine Adverse Event Report System (VAERS). The number of CNS IDDs related to COVID-19 vaccines may be underestimated. Factors contributing to this uncertainty include the widespread administration of COVID-19 vaccines, patients displaying relatively mild symptoms, lack of relevant diagnostic tools in resource-limited areas, the requirement for case reports to undergo peer review, and the unknown efforts made to contact the authors of published cases for additional information.
The pathophysiology of vaccine-induced CNS IDDs remains unclear. Many studies have suggested the molecular mimicry theory. For example, amino acid similarities between small HBV surface antigen (SHBsAg, the target of HBV non-infectious viral subunit vaccine) and MOG have been analyzed, displaying 66–100% homology in several sequences [72]. This finding suggests the possibility of immunological cross-reactivity. H1N1 and HPV vaccines have been identified with suspicious overlapping targets, which may result in certain autoimmune diseases [73]. While there is currently no evidence of molecular mimicry related to COVID-19 vaccines, the detection of anti-MOG antibodies and anti-AQP4 antibodies after receiving COVID-19 vaccines suggests potential similarities in the amino acid sequences of MOG, AQP4, and the spike (S) protein of SARS-CoV-2 virus or other unidentified immunological and inflammatory mechanisms. Articles discussing the similarity in protein sequences between the S protein and MOG or AQP4 have not been previously proposed. Although the S protein shares similar protein sequences with AQP4 and MOG (Figure 4), there are still some critical factors that may affect the protein structure, including hydropathy, global charge, and volume modifications. These factors have the potential to induce variations in the three-dimensional structures during protein folding, thereby altering the binding affinity with antibodies targeting the S protein [74]. Further clinical studies and experiments are needed to clarify the association and pathogenic mechanisms between COVID-19 vaccines and CNS IDDs. Once the definite pathological mechanisms have been elucidated, CNS IDDs induced by COVID-19 vaccines may be avoided by the development of safer vaccines. A larger database is required for further investigation into the relationship between demographics and vaccine types. The occurrence of CNS IDDs may be reduced by employing more precise indications for each kind of COVID-19 vaccines.
There are several limitations in our study, including a small number of cases, selection bias, difficulty in accessing complete original patient data, inability to confirm pre-onset levels of AQP4 and MOG antibodies, and lack of long-term follow-up for recurrence. It is challenging to confirm a causal relationship between COVID-19 vaccines and CNS IDDs. Factors such as past history, other autoimmune comorbidities, genetics, adaptive and innate immunity, and brain lesions may influence the autoimmune response after receiving the vaccines.
The rare complication of CNS IDDs after vaccination should not restrain the use of vaccines during the COVID-19 pandemic. For those who develop vaccine-related neuroimmunological adverse effects, further treatment strategies should be considered. Although the short-term outcomes were primarily ideal among the patients listed in our review following appropriate treatment, the possibility of long-term sequelae or recurrence of CNS IDDs still exists. To our knowledge, NMOSD and MOGAD patients have a higher risk of relapse if seropositivity persists, and prolonged immunotherapy should be considered to prevent relapse [9,75]. Therefore, patients initially positive for autoantibodies are recommended for regular follow-up MRI and antibody testing. Attention is also required for seronegative cases because of the possibility of seroconversion or the existence of other yet undefined autoantibodies.

5. Conclusions

We observed that there were more male patients with CNS IDDs in the viral vector vaccine type and they were often accompanied by anti-MOG antibodies. Patients receiving mRNA vaccines were older and more commonly positive for anti-AQP4 antibodies. The current consensus is that the rare occurrence of CNS IDDs is not a contraindication to vaccination. More extensive studies with larger cohorts are necessary to elucidate the pathological mechanisms of vaccine-related CNS IDDs and can enable physicians to select safer and more appropriate vaccines for each individual type to reduce the risk of adverse effects.

Author Contributions

All authors contributed to the study conception and design. Data collection and analysis were performed by M.-Y.C., H.-C.H., J.-L.H., S.-N.L. and M.-F.L. Protein sequence analysis was undertaken by Y.W. and L.C. The first draft of the manuscript was written by M.-Y.C. and H.-C.H. and L.-S.R. was responsible for the analysis and interpretation of data, as well as editing and revising the draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by Grant #113QF004E1 from National Tsing Hua University, Taiwan, awarded to Linyi Chen and Mei-Yun Cheng.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (No: 202200651B0) of Chang Gung Memorial Hospital for studies involving humans.

Informed Consent Statement

Patient consent was waived due to retrospective medical chart reviews.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author.

Acknowledgments

The cell-based immunofluorescence assay was performed and photographed by Yang Hsuan at Chang-Gung Memorial Hospital in Linkou, Taiwan.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hegen, H.; Reindl, M. Recent developments in MOG-IgG associated neurological disorders. Ther. Adv. Neurol. Disord. 2020, 13, 1756286420945135. [Google Scholar] [CrossRef] [PubMed]
  2. Jarius, S.; Aktas, O.; Ayzenberg, I.; Bellmann-Strobl, J.; Berthele, A.; Giglhuber, K.; Häußler, V.; Havla, J.; Hellwig, K.; Hümmert, M.W.; et al. Update on the diagnosis and treatment of neuromyelits optica spectrum disorders (NMOSD)—Revised recommendations of the Neuromyelitis Optica Study Group (NEMOS). Part I: Diagnosis and differential diagnosis. J. Neurol. 2023, 270, 3341–3368. [Google Scholar] [CrossRef]
  3. Papp, V.; Magyari, M.; Aktas, O.; Berger, T.; Broadley, S.A.; Cabre, P.; Jacob, A.; Kira, J.I.; Leite, M.I.; Marignier, R.; et al. Worldwide Incidence and Prevalence of Neuromyelitis Optica: A Systematic Review. Neurology 2021, 96, 59–77. [Google Scholar] [CrossRef] [PubMed]
  4. de Mol, C.L.; Wong, Y.; van Pelt, E.D.; Wokke, B.; Siepman, T.; Neuteboom, R.F.; Hamann, D.; Hintzen, R.Q. The clinical spectrum and incidence of anti-MOG-associated acquired demyelinating syndromes in children and adults. Mult. Scler. 2020, 26, 806–814. [Google Scholar] [CrossRef] [PubMed]
  5. O’Connell, K.; Hamilton-Shield, A.; Woodhall, M.; Messina, S.; Mariano, R.; Waters, P.; Ramdas, S.; Leite, M.I.; Palace, J. Prevalence and incidence of neuromyelitis optica spectrum disorder, aquaporin-4 antibody-positive NMOSD and MOG antibody-positive disease in Oxfordshire, UK. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1126–1128. [Google Scholar] [CrossRef] [PubMed]
  6. Wynford-Thomas, R.; Jacob, A.; Tomassini, V. Neurological update: MOG antibody disease. J. Neurol. 2019, 266, 1280–1286. [Google Scholar] [CrossRef] [PubMed]
  7. Ramanathan, S.; Mohammad, S.; Tantsis, E.; Nguyen, T.K.; Merheb, V.; Fung, V.S.C.; White, O.B.; Broadley, S.; Lechner-Scott, J.; Vucic, S.; et al. Clinical course, therapeutic responses and outcomes in relapsing MOG antibody-associated demyelination. J. Neurol. Neurosurg. Psychiatry 2018, 89, 127–137. [Google Scholar] [CrossRef] [PubMed]
  8. Jurynczyk, M.; Messina, S.; Woodhall, M.R.; Raza, N.; Everett, R.; Roca-Fernandez, A.; Tackley, G.; Hamid, S.; Sheard, A.; Reynolds, G.; et al. Clinical presentation and prognosis in MOG-antibody disease: A UK study. Brain 2017, 140, 3128–3138. [Google Scholar] [CrossRef]
  9. López-Chiriboga, A.S.; Majed, M.; Fryer, J.; Dubey, D.; McKeon, A.; Flanagan, E.P.; Jitprapaikulsan, J.; Kothapalli, N.; Tillema, J.M.; Chen, J.; et al. Association of MOG-IgG Serostatus With Relapse After Acute Disseminated Encephalomyelitis and Proposed Diagnostic Criteria for MOG-IgG-Associated Disorders. JAMA Neurol. 2018, 75, 1355–1363. [Google Scholar] [CrossRef]
  10. Cobo-Calvo, A.; Ruiz, A.; Maillart, E.; Audoin, B.; Zephir, H.; Bourre, B.; Ciron, J.; Collongues, N.; Brassat, D.; Cotton, F.; et al. Clinical spectrum and prognostic value of CNS MOG autoimmunity in adults: The MOGADOR study. Neurology 2018, 90, e1858–e1869. [Google Scholar] [CrossRef]
  11. Pohl, D.; Alper, G.; Van Haren, K.; Kornberg, A.J.; Lucchinetti, C.F.; Tenembaum, S.; Belman, A.L. Acute disseminated encephalomyelitis: Updates on an inflammatory CNS syndrome. Neurology 2016, 87, S38–S45. [Google Scholar] [CrossRef] [PubMed]
  12. Lotan, I.; Nishiyama, S.; Manzano, G.S.; Lydston, M.; Levy, M. COVID-19 and the risk of CNS demyelinating diseases: A systematic review. Front. Neurol. 2022, 13, 970383. [Google Scholar] [CrossRef] [PubMed]
  13. Bennetto, L.; Scolding, N. Inflammatory/post-infectious encephalomyelitis. J. Neurol. Neurosurg. Psychiatry 2004, 75 (Suppl. S1), i22–i28. [Google Scholar] [CrossRef] [PubMed]
  14. Baxter, R.; Lewis, E.; Goddard, K.; Fireman, B.; Bakshi, N.; DeStefano, F.; Gee, J.; Tseng, H.F.; Naleway, A.L.; Klein, N.P. Acute Demyelinating Events Following Vaccines: A Case-Centered Analysis. Clin. Infect. Dis. 2016, 63, 1456–1462. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, Y.; Ma, F.; Xu, Y.; Chu, X.; Zhang, J. Vaccines and the risk of acute disseminated encephalomyelitis. Vaccine 2018, 36, 3733–3739. [Google Scholar] [CrossRef] [PubMed]
  16. Woo, E.J.; Mba-Jonas, A.; Dimova, R.B.; Alimchandani, M.; Zinderman, C.E.; Nair, N. Association of Receipt of the Ad26.COV2.S COVID-19 Vaccine With Presumptive Guillain-Barré Syndrome, February–July 2021. JAMA 2021, 326, 1606–1613. [Google Scholar] [CrossRef] [PubMed]
  17. MacNeil, J.R.; Su, J.R.; Broder, K.R.; Guh, A.Y.; Gargano, J.W.; Wallace, M.; Hadler, S.C.; Scobie, H.M.; Blain, A.E.; Moulia, D.; et al. Updated Recommendations from the Advisory Committee on Immunization Practices for Use of the Janssen (Johnson & Johnson) COVID-19 Vaccine After Reports of Thrombosis with Thrombocytopenia Syndrome Among Vaccine Recipients—United States, April 2021. MMWR Morb. Mortal. Wkly. Rep. 2021, 70, 651–656. [Google Scholar] [CrossRef]
  18. 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]
  19. Su, S.C.; Lyu, R.K.; Chang, C.W.; Tseng, W.J. The First Guillain-Barr? Syndrome after SARS-CoV-2 Vaccination in Taiwan. Acta Neurol. Taiwan 2022, 31, 46–51. [Google Scholar]
  20. Gargano, J.W.; Wallace, M.; Hadler, S.C.; Langley, G.; Su, J.R.; Oster, M.E.; Broder, K.R.; Gee, J.; Weintraub, E.; Shimabukuro, T.; et al. Use of mRNA COVID-19 Vaccine After Reports of Myocarditis Among Vaccine Recipients: Update from the Advisory Committee on Immunization Practices—United States, June 2021. MMWR Morb. Mortal. Wkly. Rep. 2021, 70, 977–982. [Google Scholar] [CrossRef]
  21. Rinaldi, V.; Bellucci, G.; Romano, A.; Bozzao, A.; Salvetti, M. ADEM after ChAdOx1 nCoV-19 vaccine: A case report. Mult. Scler. 2021, 28, 13524585211040222. [Google Scholar] [CrossRef] [PubMed]
  22. Permezel, F.; Borojevic, B.; Lau, S.; de Boer, H.H. Acute disseminated encephalomyelitis (ADEM) following recent Oxford/AstraZeneca COVID-19 vaccination. Forensic Sci. Med. Pathol. 2021, 18, 74–79. [Google Scholar] [CrossRef] [PubMed]
  23. Pagenkopf, C.; Südmeyer, M. A case of longitudinally extensive transverse myelitis following vaccination against COVID-19. J. Neuroimmunol. 2021, 358, 577606. [Google Scholar] [CrossRef] [PubMed]
  24. Tan, W.Y.; Yusof Khan, A.H.K.; Mohd Yaakob, M.N.; Abdul Rashid, A.M.; Loh, W.C.; Baharin, J.; Ibrahim, A.; Ismail, M.R.; Inche Mat, L.N.; Wan Sulaiman, W.A.; et al. Longitudinal extensive transverse myelitis following ChAdOx1 nCOV-19 vaccine: A case report. BMC Neurol. 2021, 21, 395. [Google Scholar] [CrossRef]
  25. Notghi, A.A.; Atley, J.; Silva, M. Lessons of the month 1: Longitudinal extensive transverse myelitis following AstraZeneca COVID-19 vaccination. Clin. Med. 2021, 21, e535–e538. [Google Scholar] [CrossRef]
  26. Hsiao, Y.T.; Tsai, M.J.; Chen, Y.H.; Hsu, C.F. Acute Transverse Myelitis after COVID-19 Vaccination. Medicina 2021, 57, 1010. [Google Scholar] [CrossRef]
  27. Vegezzi, E.; Ravaglia, S.; Buongarzone, G.; Bini, P.; Diamanti, L.; Gastaldi, M.; Prunetti, P.; Rognone, E.; Marchioni, E. Acute myelitis and ChAdOx1 nCoV-19 vaccine: Casual or causal association? J. Neuroimmunol. 2021, 359, 577686. [Google Scholar] [CrossRef]
  28. Singh Malhotra, H.; Gupta, P.; Prabhu, V.; Garg, R.K.; Dandu, H.; Agarwal, V. COVID-19 vaccination-associated myelitis. QJM 2021, 114, 591–593. [Google Scholar] [CrossRef]
  29. Tahir, N.; Koorapati, G.; Prasad, S.; Jeelani, H.M.; Sherchan, R.; Shrestha, J.; Shayuk, M. SARS-CoV-2 Vaccination-Induced Transverse Myelitis. Cureus 2021, 13, e16624. [Google Scholar] [CrossRef] [PubMed]
  30. Badrawi, N.; Kumar, N.; Albastaki, U. Post COVID-19 vaccination neuromyelitis optica spectrum disorder: Case report & MRI findings. Radiol. Case Rep. 2021, 16, 3864–3867. [Google Scholar] [CrossRef] [PubMed]
  31. Shimizu, M.; Ogaki, K.; Nakamura, R.; Kado, E.; Nakajima, S.; Kurita, N.; Watanabe, M.; Yamashiro, K.; Hattori, N.; Urabe, T. An 88-year-old woman with acute disseminated encephalomyelitis following messenger ribonucleic acid-based COVID-19 vaccination. eNeurologicalSci 2021, 25, 100381. [Google Scholar] [CrossRef]
  32. Vogrig, A.; Janes, F.; Gigli, G.L.; Curcio, F.; Negro, I.D.; D’Agostini, S.; Fabris, M.; Valente, M. Acute disseminated encephalomyelitis after SARS-CoV-2 vaccination. Clin. Neurol. Neurosurg. 2021, 208, 106839. [Google Scholar] [CrossRef]
  33. Khayat-Khoei, M.; Bhattacharyya, S.; Katz, J.; Harrison, D.; Tauhid, S.; Bruso, P.; Houtchens, M.K.; Edwards, K.R.; Bakshi, R. COVID-19 mRNA vaccination leading to CNS inflammation: A case series. J. Neurol. 2021, 269, 1093–1106. [Google Scholar] [CrossRef]
  34. Alshararni, A. Acute Transverse Myelitis Associated with COVID-19 vaccine: A Case Report. Int. J. Res. Pharm. Sci. 2021, 12, 5. [Google Scholar] [CrossRef]
  35. McLean, P.; Trefts, L. Transverse myelitis 48 hours after the administration of an mRNA COVID-19 vaccine. Neuroimmunol. Rep. 2021, 1, 100019. [Google Scholar] [CrossRef]
  36. Kania, K.; Ambrosius, W.; Tokarz Kupczyk, E.; Kozubski, W. Acute disseminated encephalomyelitis in a patient vaccinated against SARS-CoV-2. Ann. Clin. Transl. Neurol. 2021, 8, 2000–2003. [Google Scholar] [CrossRef]
  37. Fujikawa, P.; Shah, F.A.; Braford, M.; Patel, K.; Madey, J. Neuromyelitis Optica in a Healthy Female after Severe Acute Respiratory Syndrome Coronavirus 2 mRNA-1273 Vaccine. Cureus 2021, 13, e17961. [Google Scholar] [CrossRef]
  38. Gao, J.J.; Tseng, H.P.; Lin, C.L.; Shiu, J.S.; Lee, M.H.; Liu, C.H. Acute Transverse Myelitis Following COVID-19 Vaccination. Vaccines 2021, 9, 1008. [Google Scholar] [CrossRef] [PubMed]
  39. Khan, E.; Shrestha, A.K.; Colantonio, M.A.; Liberio, R.N.; Sriwastava, S. Acute transverse myelitis following SARS-CoV-2 vaccination: A case report and review of literature. J. Neurol. 2022, 269, 1121–1132. [Google Scholar] [CrossRef] [PubMed]
  40. Fitzsimmons, W.; Nance, C.S. Sudden Onset of Myelitis after COVID-19 Vaccination: An Under-Recognized Severe Rare Adverse Event. SSRN 2021, 7, 3841558. [Google Scholar] [CrossRef]
  41. Ozgen Kenangil, G.; Ari, B.C.; Guler, C.; Demir, M.K. Acute disseminated encephalomyelitis-like presentation after an inactivated coronavirus vaccine. Acta Neurol. Belg. 2021, 121, 1089–1091. [Google Scholar] [CrossRef]
  42. Erdem, N.; Demirci, S.; Özel, T.; Mamadova, K.; Karaali, K.; Çelik, H.T.; Uslu, F.I.; Özkaynak, S.S. Acute transverse myelitis after inactivated COVID-19 vaccine. Ideggyogy. Sz. 2021, 74, 273–276. [Google Scholar] [CrossRef]
  43. Cao, L.; Ren, L. Acute disseminated encephalomyelitis after severe acute respiratory syndrome coronavirus 2 vaccination: A case report. Acta Neurol. Belg. 2021, 122, 793–795. [Google Scholar] [CrossRef] [PubMed]
  44. Sepahvand, M.; Yazdi, N.; Rohani, M.; Emamikhah, M. Cervical longitudinally extensive myelitis after vaccination with inactivated virus-based COVID-19 vaccine. Radiol. Case Rep. 2022, 17, 303–305. [Google Scholar] [CrossRef]
  45. Chen, S.; Fan, X.R.; He, S.; Zhang, J.W.; Li, S.J. Watch out for neuromyelitis optica spectrum disorder after inactivated virus vaccination for COVID-19. Neurol. Sci. 2021, 42, 3537–3539. [Google Scholar] [CrossRef] [PubMed]
  46. Corrêa, D.G.; Cañete, L.A.Q.; Dos Santos, G.A.C.; de Oliveira, R.V.; Brandão, C.O.; da Cruz, L.C.H., Jr. Neurological symptoms and neuroimaging alterations related with COVID-19 vaccine: Cause or coincidence? Clin. Imaging 2021, 80, 348–352. [Google Scholar] [CrossRef]
  47. Matsumoto, Y.; Ohyama, A.; Kubota, T.; Ikeda, K.; Kaneko, K.; Takai, Y.; Warita, H.; Takahashi, T.; Misu, T.; Aoki, M. MOG Antibody-Associated Disorders Following SARS-CoV-2 Vaccination: A Case Report and Literature Review. Front. Neurol. 2022, 13, 845755. [Google Scholar] [CrossRef]
  48. Dams, L.; Kraemer, M.; Becker, J. MOG-antibody-associated longitudinal extensive myelitis after ChAdOx1 nCoV-19 vaccination. Mult. Scler. 2022, 28, 1159–1162. [Google Scholar] [CrossRef] [PubMed]
  49. Mumoli, L.; Vescio, V.; Pirritano, D.; Russo, E.; Bosco, D. ADEM anti-MOG antibody-positive after SARS-CoV2 vaccination. Neurol. Sci. 2022, 43, 763–766. [Google Scholar] [CrossRef]
  50. Sehgal, V.; Bansal, P.; Arora, S.; Kapila, S.; Bedi, G.S. Myelin Oligodendrocyte Glycoprotein Antibody Disease After COVID-19 Vaccination—Causal or Incidental? Cureus 2022, 14, e27024. [Google Scholar] [CrossRef]
  51. Garg, R.K.; Malhotra, H.S.; Kumar, N.; Pandey, S.; Patil, M.R.; Uniyal, R.; Rizvi, I. Tumefactive Demyelinating Brain Lesion Developing after Administration of Adenovector-Based COVID-19 Vaccine: A Case Report. Neurol. India 2022, 70, 409–411. [Google Scholar] [CrossRef] [PubMed]
  52. Ballout, A.A.; Babaie, A.; Kolesnik, M.; Li, J.Y.; Hameed, N.; Waldman, G.; Chaudhry, F.; Saba, S.; Harel, A.; Najjar, S. A Single-Health System Case Series of New-Onset CNS Inflammatory Disorders Temporally Associated With mRNA-Based SARS-CoV-2 Vaccines. Front. Neurol. 2022, 13, 796882. [Google Scholar] [CrossRef]
  53. Raknuzzaman, D.M.; Jannaty, T.; Hossain, D.M.B.; Saha, D.B.; Dey, D.S.K.; Shahidullah, D.M. Post COVID-19 Vaccination Acute Disseminated Encephalomyelitis: A Case Report in Bangladesh. Int. J. Med. Sci. And. Clin. Res. Stud. 2021, 1, 31–36. [Google Scholar]
  54. Nagaratnam, S.A.; Ferdi, A.C.; Leaney, J.; Lee, R.L.K.; Hwang, Y.T.; Heard, R. Acute disseminated encephalomyelitis with bilateral optic neuritis following ChAdOx1 COVID-19 vaccination. BMC Neurol. 2022, 22, 54. [Google Scholar] [CrossRef] [PubMed]
  55. Yazdanpanah, F.; Iranpour, P.; Haseli, S.; Poursadeghfard, M.; Yarmahmoodi, F. Acute disseminated encephalomyelitis (ADEM) after SARS- CoV-2 vaccination: A case report. Radiol. Case Rep. 2022, 17, 1789–1793. [Google Scholar] [CrossRef] [PubMed]
  56. Doi, K.; Ohara, Y.; Ouchi, T.; Sasaki, R.; Maki, F.; Mizuno, J. Cervical Transverse Myelitis Following COVID-19 Vaccination. NMC Case Rep. J. 2022, 9, 145–149. [Google Scholar] [CrossRef]
  57. Ahmad, H.R.; Timmermans, V.M.; Dakakni, T. Acute Disseminated Encephalomyelitis After SARS-CoV-2 Vaccination. Am. J. Case Rep. 2022, 23, e936574. [Google Scholar] [CrossRef]
  58. Maramattom, B.V.; Lotlikar, R.S.; Sukumaran, S. Central nervous system adverse events after ChAdOx1 vaccination. Neurol. Sci. 2022, 43, 3503–3507. [Google Scholar] [CrossRef]
  59. Al-Quliti, K.; Qureshi, A.; Quadri, M.; Abdulhameed, B.; Alanazi, A.; Alhujeily, R. Acute Demyelinating Encephalomyelitis Post-COVID-19 Vaccination: A Case Report and Literature Review. Diseases 2022, 10, 13. [Google Scholar] [CrossRef]
  60. Mousa, H.; Patel, T.H.; Meadows, I.; Ozdemir, B. Acute Disseminated Encephalomyelitis (ADEM) After Consecutive Exposures to Mycoplasma and COVID Vaccine: A Case Report. Cureus 2022, 14, e26258. [Google Scholar] [CrossRef]
  61. Motahharynia, A.; Naghavi, S.; Shaygannejad, V.; Adibi, I. Fulminant neuromyelitis optica spectrum disorder (NMOSD) following COVID-19 vaccination: A need for reconsideration? Mult. Scler. Relat. Disord. 2022, 66, 104035. [Google Scholar] [CrossRef] [PubMed]
  62. Anamnart, C.; Tisavipat, N.; Owattanapanich, W.; Apiwattanakul, M.; Savangned, P.; Prayoonwiwat, N.; Siritho, S.; Rattanathamsakul, N.; Jitprapaikulsan, J. Newly diagnosed neuromyelitis optica spectrum disorders following vaccination: Case report and systematic review. Mult. Scler. Relat. Disord. 2022, 58, 103414. [Google Scholar] [CrossRef] [PubMed]
  63. Kuntz, S.; Saab, G.; Schneider, R. Antibody-Positive Neuromyelitis Optica Spectrum Disorder After Second COVID-19 Vaccination: A Case Report. SN Compr. Clin. Med. 2022, 4, 130. [Google Scholar] [CrossRef]
  64. Caliskan, I.; Bulus, E.; Afsar, N.; Altintas, A. A Case With New-Onset Neuromyelitis Optica Spectrum Disorder Following COVID-19 mRNA BNT162b2 Vaccination. Neurologist 2022, 27, 147–150. [Google Scholar] [CrossRef] [PubMed]
  65. Netravathi, M.; Dhamija, K.; Gupta, M.; Tamborska, A.; Nalini, A.; Holla, V.V.; Nitish, L.K.; Menon, D.; Pal, P.K.; Seena, V.; et al. COVID-19 vaccine associated demyelination & its association with MOG antibody. Mult. Scler. Relat. Disord. 2022, 60, 103739. [Google Scholar] [CrossRef] [PubMed]
  66. World Health Organization. Interim Recommendations for Use of the ChAdOx1-S [Recombinant] Vaccine against COVID-19 (AstraZeneca COVID-19 Vaccine AZD1222 Vaxzevria™, SII COVISHIELD™): Interim Guidance, First Issued 10 February 2021, Updated 21 April 2021, Updated 30 July 2021, Latest Update 15 March 2022; World Health Organization: Geneva, Switzerland, 2022.
  67. Marignier, R.; Hacohen, Y.; Cobo-Calvo, A.; Pröbstel, A.K.; Aktas, O.; Alexopoulos, H.; Amato, M.P.; Asgari, N.; Banwell, B.; Bennett, J.; et al. Myelin-oligodendrocyte glycoprotein antibody-associated disease. Lancet Neurol. 2021, 20, 762–772. [Google Scholar] [CrossRef]
  68. Haque, A.; Pant, A.B. Efforts at COVID-19 Vaccine Development: Challenges and Successes. Vaccines 2020, 8, 739. [Google Scholar] [CrossRef]
  69. Lotan, I.; Romanow, G.; Levy, M. Patient-reported safety and tolerability of the COVID-19 vaccines in persons with rare neuroimmunological diseases. Mult. Scler. Relat. Disord. 2021, 55, 103189. [Google Scholar] [CrossRef]
  70. Taiwan Food and Drug Administration. The Report of Adverse Effects after COVID-19 Vaccines in Taiwan. Available online: https://www.fda.gov.tw/tc/includes/GetFile.ashx?id=f637814781667990040&type=2&cid=39989 (accessed on 10 May 2023).
  71. Huang, C.T.; Hsu, S.Y.; Wang, C.H.; Tseng, W.J.; Yang, C.Y.; Ng, C.J.; Warkentin, T.E.; Cheng, M.H. Double high-dose immunoglobulin for ChAdOx1 nCov-19 vaccine-induced immune thrombotic thrombocytopenia. Thromb. Res. 2021, 206, 14–17. [Google Scholar] [CrossRef]
  72. Bogdanos, D.P.; Smith, H.; Ma, Y.; Baum, H.; Mieli-Vergani, G.; Vergani, D. A study of molecular mimicry and immunological cross-reactivity between hepatitis B surface antigen and myelin mimics. Clin. Dev. Immunol. 2005, 12, 217–224. [Google Scholar] [CrossRef]
  73. Segal, Y.; Shoenfeld, Y. Vaccine-induced autoimmunity: The role of molecular mimicry and immune crossreaction. Cell. Mol. Immunol. 2018, 15, 586–594. [Google Scholar] [CrossRef] [PubMed]
  74. Machado, R.S.; Gomes-Neto, F.; Aguiar-Oliveira, M.L.; Burlandy, F.M.; Tavares, F.N.; da Silva, E.E.; Sousa, I.P., Jr. Analysis of Coxsackievirus B5 Infections in the Central Nervous System in Brazil: Insights into Molecular Epidemiology and Genetic Diversity. Viruses 2022, 14, 899. [Google Scholar] [CrossRef] [PubMed]
  75. Weinshenker, B.G.; Wingerchuk, D.M.; Vukusic, S.; Linbo, L.; Pittock, S.J.; Lucchinetti, C.F.; Lennon, V.A. Neuromyelitis optica IgG predicts relapse after longitudinally extensive transverse myelitis. Ann. Neurol. 2006, 59, 566–569. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Spinal MRI: On the 6th day after onset, an intramedullary spotty lesion with T2 hyperintensity at T3 to T4 was shown (red arrows: (a) sagittal view. (c) axial view). Five months later, the lesion over T3 to T4 was totally resolved on T2 FLAIR images (red arrows: (b) sagittal view. (d) axial view).
Figure 1. Spinal MRI: On the 6th day after onset, an intramedullary spotty lesion with T2 hyperintensity at T3 to T4 was shown (red arrows: (a) sagittal view. (c) axial view). Five months later, the lesion over T3 to T4 was totally resolved on T2 FLAIR images (red arrows: (b) sagittal view. (d) axial view).
Diseases 12 00060 g001
Figure 2. Brain MRI: Bilateral poorly demarcated T2 FLAIR hyperintensities at brainstem (ac), pulvinar thalami (d), putamen (d), centrum semiovale (e), and subcortical white matters (f) are marked by red arrows in the image.
Figure 2. Brain MRI: Bilateral poorly demarcated T2 FLAIR hyperintensities at brainstem (ac), pulvinar thalami (d), putamen (d), centrum semiovale (e), and subcortical white matters (f) are marked by red arrows in the image.
Diseases 12 00060 g002
Figure 3. Cell-based immunofluorescence assay (Euroimmun, Lübeck, Germany): Indirect immunofluorescence test was used on HEK-293 cells transfected with plasmids containing MOG. Compared to the negative control (a), positive fluorescence was observed over the cell membrane and cytoplasm in transfected HEK-293 cells after applying the diluted (1:10) patient’s serum (b) and FITC-labeled secondary anti-human IgG antibody at 20× magnification.
Figure 3. Cell-based immunofluorescence assay (Euroimmun, Lübeck, Germany): Indirect immunofluorescence test was used on HEK-293 cells transfected with plasmids containing MOG. Compared to the negative control (a), positive fluorescence was observed over the cell membrane and cytoplasm in transfected HEK-293 cells after applying the diluted (1:10) patient’s serum (b) and FITC-labeled secondary anti-human IgG antibody at 20× magnification.
Diseases 12 00060 g003
Figure 4. S-Covid 19 refers to the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), retrieved from the Protein Data Bank (PDB) under the accession number 6X29. Aquaporin 4 (AQP4) has a well-documented 3D structure with established structural domains, sourced from the PDB with the accession number 3GD8 (a). Notably, the extracellular loop of the AQP4 transmembrane protein can function as an antigen or be recognized by antibodies. The 3D structure of MOG is unknown (b). The Expasy SIM protein sequence alignment tool was utilized for analysis (https://web.expasy.org/sim/ accessed on 6 June 2023). Additionally, we employed PyMOL for protein structural analysis. The red-highlighted structure represents regions of high similarity between the two proteins.
Figure 4. S-Covid 19 refers to the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), retrieved from the Protein Data Bank (PDB) under the accession number 6X29. Aquaporin 4 (AQP4) has a well-documented 3D structure with established structural domains, sourced from the PDB with the accession number 3GD8 (a). Notably, the extracellular loop of the AQP4 transmembrane protein can function as an antigen or be recognized by antibodies. The 3D structure of MOG is unknown (b). The Expasy SIM protein sequence alignment tool was utilized for analysis (https://web.expasy.org/sim/ accessed on 6 June 2023). Additionally, we employed PyMOL for protein structural analysis. The red-highlighted structure represents regions of high similarity between the two proteins.
Diseases 12 00060 g004
Table 1. Demographic, clinical features, and laboratory data of 79 patients with CNS IDDs after COVID-19 vaccines.
Table 1. Demographic, clinical features, and laboratory data of 79 patients with CNS IDDs after COVID-19 vaccines.
NoAge/Sex/PHVaccine/
Interval (D)
DxSerum CSFMRITxOutcomeRef.
AQP4MOGWBCLym (%)Neut (%)TPGlu RatioOCBBrainSpine (Myelitis)
150M/chronic HBV 1st AZ a/13MOGAD+17599078.10.47NABil thalami, pu, subcortical WM, brainstemT3–T4 PTRIndex case
245M1st AZ/12ADEM + SSM44PNANNA+Pons, R MCP, R thalamus; C+Cervical, thoracic, conus medullaris; C+PTR[21]
363M/DM, HL, IHD, Af1st AZ/12ADEM2NANA69N+Bil WM, bil CC, L thalamus, IC, L midbrain, lower pons, R MCPNPT + PPE(D20)[22]
445M/atopic dermatitis1st AZ/8LETM481NA671400.43NC3–T2PTI[23]
525F1st AZ/12LETMNANANA54.60.55NT3–T5, T7–T8, T11–L1; C+ in T7–T8PTI[24]
658M/DM, pulmonary sarcoidosis1st AZ/7LETM1110001620.54+NAC1–T10; C+ in T3–T4, T9–T10PT + PEI[25]
741M/DM1st AZ/14LETMNA11100044.3NANANT1–T6; C+PTR[26]
844F1st AZ/4SSMPNA76.7NANT7–T8, T10–T11; C+ in T7–T8PTR[27]
936M1st AZ/8SSM NANANA54NANANC6–C7; C+PTR[28]
1044F1st Janssen b/10LETMNA22796043NNC2 to upper thoracic PT + PER[29]
1134M2nd Sputnik V c/21NMOSD+PNANA3rd, 4th PV, thalamus, CC, optic chiasmaNPPI[30]
1288F/DM, AD2nd Pfizer d/29ADEMNANANANANANANABil MCPsNAPTR[31]
1356F/Post-infectious rhombencephalitis1st Pfizer/14ADEM-like iNNANANNL cerebellar peduncle and L centrum semiovale NAOral steroidR[32]
1464M1st Pfizer/18NMOSD+NANNANANNCC, L frontal and parietal WM; C−Cervical to the conus; C+PT + PE + RTXI[33]
1538M1st Pfizer/2SSMNANANANANA62.1NNANAT11–T12; C+NANA[34]
1669F/cervical ca, HL, hypothyroidism1st Pfizer/2LETMNNANANN+NC3–C4 to T2–T3 PTI[35]
1719F/atopic dermatitis, depression1st Moderna e/14ADEM +
LETM
29491164.8NABil hemispheres, pons, medulla, cerebellum; C+Medulla to T11; C+PT + PER[36]
1846F/vit B12 deficiency1st Moderna/2NMOSDNANANANANANANANC6–T2PTR[37]
1976F/HTN, vit B12 deficiency1st Moderna/2LETMNA15NA7357.2NANC2–C5; C+ in C3 PTI[38]
2067F/CAD, CKD, neuropathy1st Moderna/1LETM2NANA560.61+Nonspecific WM C1–C3; C+PT + PPI[39]
2163M2nd Moderna/1SSM3NANA37NNANonspecific bil corona radiata Conus medullaris; C+IVIG + PTI[40]
2246F/Hashimoto’s thyroiditis2nd Sinovac f/30ADEM-like00045NAL thalamus, bil corona radiata, L diencephalon, R parietal cortexNAPTR[41]
2378F/DM, HTN, breast ca2nd Sinovac/21LETM2NANA560.69NC1–T3PTI[42]
2424F1st Sinopharm g/14ADEM51NANANANABil temporalNAIVIGI[43]
2571M/DM, HTN, IHD1st Sinopharm/5LETM000NNNCervico–medullary junction to C3PTI[44]
2650F1st inactivated/3NMOSD+31NANANNArea postrema, bil hypothalamusNPTI[45]
2765M1st AZ/8LETMNNANA70NANAC4–C6PTR[46]
2868F/HTN, pancreatic ca2nd Moderna/14MOGAD+00032NA+R lateral pons, trigeminal nerve, MCP NAPTI[47]
2959M1st AZ/14MOGAD+110NANA625N+NT7–L1PT + PEI[48]
3045M/allergic asthma1st AZ/7MOGAD+43NANA40.6NBil subcortical, gray–white matterT10–conus PTI[49]
3126M1st AZ/20MOGAD+184NANA88NABil MCPs, ponsC3–C6PTI[50]
3256F/HTN1st AZ/2ADEM-likeNANANANANANANANAL parietal WM, CCNAOral steroidI[51]
3381M1st Moderna/13ADEMNA6983%NA52NNAR dorsal medulla, L pons, midbrain, thalamiNAPT + IVIG + PPE(D26)[52]
3463F/HL, hypothyroidism1st Pfizer/7NMOSD+3391%NA57NAL thalamusT6–T12PT + PPR
3554F/ITP2nd Moderna/3NMOSD+2686%NA71NANT2–T9PTI
3655M1st mRNA/21ADEMNANA20095%NA75NNABil WMNAPTR[53]
3736F1st AZ/14ADEM59NANA40N+Subcortical WM, PIC, pons, L MCPNPTR[54]
3837M1st Sinopharm/30ADEMNANA2NANA560.61L cerebral peduncle, bil pons, medullaNPTR[55]
3927F/Rectovaginal fistula2nd Pfizer/4LETM7NANA43NNC5–C7 PTI[56]
4061F/HTN, anxiety1st Pfizer/5ADEMNANNANA61NDeep WM, brainstem, cerebellumNPT + IVIGI[57]
4164M1st AZ/10ADEM25PNANANNABil mesial temporal, hippocampus, MCPsNAPT + PP + RTXR[58]
4264M2nd AZ/20ADEM + SSMNNANANNNABil perirolandic cortex, corona radiataT8–T9 dorsal PT + IVIG + RTXI
4346M1st AZ/5ADEM + LETM63NANA52NNABil MCP, pons, R paramedian medulla, L thalamocapsular LETMPT + PEI
4442F1st AZ/5ADEM-likeNNANANNNAR temporal NAOral steroidR
4556F1st AZ/10ADEM-likeNANA116%20%NNASubcortical WM, basal gangliaNAPTR[59]
4644F/HL, hypothyroidism, renal stone, anxiety1st mRNA/6ADEM +
LETM
NA105NANA98NMultifocal PV lesions; C+ in L frontal WMC3–C4 to thoracic with sparing C5–C6; C+ in T7–T8PT + PPI[60]
4770F3rd Sinovac/
7
NMOSD+NANNNNNNAC1–C7 and T1–T3PT + PE + CPE(M2)[61]
4826F1st Sinovac/10NMOSD+NANNNNNNC4–C5PT + PE + RTXI[62]
4946F1st AZ/10NMOSD+NANNNNNR lateral medulla, PV C2–C3PT +AZTI
5080M2nd Pfizer/2NMOSD++3993%NANNNT3–T10PT + PE + MMFI[63]
5143F2nd Pfizer/1NMOSD+6NANA40.1N+R ON, R periatrium, L crus cerebriC1 to mid-thoracicPT + PE + RTXR[64]
5229F1st AZ/11MOGADNA+0NANA18NLong intraorbital segment of R ONNAPT + PPNA[65]
5326F1st Covaxin h/11LETM207NAP95.8NNANAC2–L1PT + PPNA
5454F1st AZ/14ADEM-like8PNA77NNACC, PV, subcortical WM, infratentorialNAPT + PPNA
5544M1st AZ/7MOGADNA+130PNA38NNANACervical and dorsal cord, conus PT + PPNA
5650F1st AZ/28SSM2PNA28NNANAC6PTNA
5739M1st AZ/14MOGADNA+NANANANANANALong intraorbital segment of R ONNAPTNA
5854M1st AZ/14MOGADNA+NANANANANANAR ponsNPTNA
5934M1st AZ/1ON2PNA26NNAR ONNAPTNA
6035M1st AZ/9MOGADNA+58PNA47.4NNAMidbrain, pons, L MCP, PICs, thalamus, bil centrum semiovaleCervical to conusPTNA
6120F1st AZ/3ADEM-likeNANANANANANAPericallosal, callososeptal, PV, fronto-parietalNAPTNA
6231M1st AZ/14LETM370NAP174NNANACervico–dorsal long segmentPT + PP + RTXNA
6320F1st Covaxin/1ADEM +
SSM
8PNA24.9NJuxtacortical C5PT + PPNA
6445F1st AZ/21MOGADNA+2PNA52.3N+Bil ONNPT + PPNA
6533F1st AZ/14MOGADNA+105PNA28.12NNABil fronto-parietalNAPTNA
6653F2nd AZ/1ADEM +
LETM
6PNA54.2NNABil subcortical, PV, insular, cerebellum, brainstem C5–C7 and T6–T7PTNA
6738M2nd AZ/6ADEM-like6NANA67.8NNAL MCP, R corona radiataNAPTNA
6830M1st AZ/14ADEM + ON450NA26.8N+Bil subcortical, bil ONNAPT + PP + RTXNA
6930F1st AZ/15ADEM +
SSM
4NANA36N+CCC3PT + PP + MMFNA
7036M2nd AZ/32MOGADNA+72080NA144.4NNABil trigeminal n, pons Obex to conusPT + PPNA
7127F1st AZ/8ADEM-likeClearNANA27.7NNABil PV NPTNA
7260M2nd AZ/14ADEM990NA68.3NR pons, midbrain, temporal, parietal, CC NAPT + MMFNA
7323F2nd AZ/7ADEM +
LETM
NANANANANAR frontal horn and bil lateral ventricles C2–C5 and T4 myelitisPTNA
7440M1st AZ/10MOGADNA+8100032N+Pons, bil thalami, and R frontal cortexC4–T3PT + MMFNA
7545M1st AZ/10MOGAD+4444NA90.9NNABrainstem, supratentorial Cervicodorsal cordPT + PPNA
7634F2nd AZ/36NMOSD+1NANA15.3NDorsal aspect of medullaNAPT + PP + RTXNA
7731M1st AZ/42ADEM +
LETM
32100049.2NNACervico-medullary junction, R frontal subcortical C2–C5PT + PP + MMFNA
7852F1st AZ/35ADEM-like2NANA40.5NNAL frontal, insular, midbrainNAPT + PP + RTXNA
7965F1st AZ/42NMOSD+17NANA49NNAFrontal subcortical WMT2–T11PT + PP + MMFNA
Abbreviations: ↑: means elevated without specifying exact values. Af: atrial fibrillation. AD: Alzheimer’s disease. AZT: azathioprine. C+: with contrast enhancement. CC: corpus callosum. CKD: chronic kidney disease. CP: cyclophosphamide. DM: diabetes mellitus. E: expired. HBV: hepatitis B virus. HL: hyperlipidemia. HTN: hypertension. I: improvement. IC: internal capsule. IHD: ischemic heart disease. ITP: immune thrombocytopenic purpura. MCP: middle cerebellar peduncle. MMF: Mycophenolate mofetil. N: normal. NA: not available. ON: optic neuritis. P: predominant. PIC: posterior limb of internal capsule. PP: plasmapheresis. PT: steroid pulse therapy. PU: putamen. PV: periventricular. R: full recovery. RTX: rituximab. WM: white matter. a Oxford–AstraZeneca, marketed as Covishield. A kind of viral vector (chimpanzee adenovirus) vaccine. b Johnson & Johnson’s Janssen, a kind of viral vector (human adenovirus) vaccine. c A kind of viral vector (human adenovirus) vaccine. d Pfizer–BioNTech, marketed as Comirnaty. A kind of messenger RNA vaccine. e A kind of messenger RNA vaccine. f Marketed as CoronaVac. A kind of inactivated vaccine. g Also known as BBIBP-CorV. A kind of inactivated vaccine. h Also known as BBV 152. A kind of inactivated vaccine. i Clinical and MRI features are compatible with ADEM, but without presentation of encephalopathy.
Table 2. Differences between three major vaccine types.
Table 2. Differences between three major vaccine types.
Vaccine Types
Viral Vector (n = 49)mRNA (n = 20)Inactivated (n = 10)p Value
Sex, n (%) 0.027 *
 Male 28 (57)6 (30)2 (20)
 Female 21 (43)14 (70)8 (80)
Mean age of onset (S.D.) 44.3 (12.2)58.1 (17.9)44.8 (21.8)0.002 #*
Doses (1st/2nd/3rd) 41/8/013/7/07/2/10.042 *
Post-vaccination onset time (days) 13.68.113.60.086
Clinical presentations, n (%) 0.203
 ADEM 13 (27)5 (25)3 (30)
 Pure myelitis 10 (20)6 (30)3 (30)
  MOGAD 14 (29)1 (5)0 (0)
 NMOSD 4 (8)6 (30)3 (30)
  ADEM with myelitis 7 (14)2 (10)1 (10)
 ON 1 (2)0 (0)0 (0)
Serum Autoantibodies , n 0.044 *
 Negative 31147
 MOG 1410
 AQP4 443
 MOG + AQP4 010
CSF, n
 WBC count 74.257.130.10.605
 Lym predominant (yes/no) 23/36/21/40.004 *
 Elevated total protein (yes/no) @24/1910/84/50.817
 CSF/serum glu ratio (<0.6/>0.6) 4/330/130/80.295
 Oligoclonal bands (+/−) 9/134/110/90.071
Brain MRI lesions, n
 Cortex (+/−) 7/331/162/50.329
 Deep grey matters (+/−) 8/322/152/50.598
 Subcortical white matters (+/−) 18/225/122/50.454
 Periventricular white matters (+/−) 16/246/111/60.424
 Periaqueductal white matters (+/−) 3/370/170/70.389
 Brainstem (+/−) 20/207/102/50.532
  Gadolinium enhanced (+/−) 1/92/90/50.562
Spine MRI, n
 Segments of cord lesions (≥3/<3) 20/812/24/20.143
  Gadolinium enhanced (+/−) 6/67/10/40.015 *
Treatment, n 0.754
  First-line immunotherapy 43178
  First- and second-line immunotherapy 622
Outcome, n 0.762
  Recovery 1172
  Improved 11115
  Expired 111
* Pearson chi-squared test, p < 0.05; # Independent t-test, p < 0.05; @ Elevated total protein defined as total protein > 45 mg/dL.
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

Cheng, M.-Y.; Ho, H.-C.; Hsu, J.-L.; Wang, Y.; Chen, L.; Lim, S.-N.; Liao, M.-F.; Ro, L.-S. Clinical Research into Central Nervous System Inflammatory Demyelinating Diseases Related to COVID-19 Vaccines. Diseases 2024, 12, 60. https://doi.org/10.3390/diseases12030060

AMA Style

Cheng M-Y, Ho H-C, Hsu J-L, Wang Y, Chen L, Lim S-N, Liao M-F, Ro L-S. Clinical Research into Central Nervous System Inflammatory Demyelinating Diseases Related to COVID-19 Vaccines. Diseases. 2024; 12(3):60. https://doi.org/10.3390/diseases12030060

Chicago/Turabian Style

Cheng, Mei-Yun, Hsuan-Chen Ho, Jung-Lung Hsu, Yi Wang, Linyi Chen, Siew-Na Lim, Ming-Feng Liao, and Long-Sun Ro. 2024. "Clinical Research into Central Nervous System Inflammatory Demyelinating Diseases Related to COVID-19 Vaccines" Diseases 12, no. 3: 60. https://doi.org/10.3390/diseases12030060

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