ArticlePDF Available

A Case Report: Multifocal Necrotizing Encephalitis and Myocarditis after BNT162b2 mRNA Vaccination against COVID-19

MDPI
Vaccines
Authors:

Abstract and Figures

The current report presents the case of a 76-year-old man with Parkinson’s disease (PD) who died three weeks after receiving his third COVID-19 vaccination. The patient was first vaccinated in May 2021 with the ChAdOx1 nCov-19 vector vaccine, followed by two doses of the BNT162b2 mRNA vaccine in July and December 2021. The family of the deceased requested an autopsy due to ambiguous clinical signs before death. PD was confirmed by post-mortem examinations. Furthermore, signs of aspiration pneumonia and systemic arteriosclerosis were evident. However, histopathological analyses of the brain uncovered previously unsuspected findings, including acute vasculitis (predominantly lymphocytic) as well as multifocal necrotizing encephalitis of unknown etiology with pronounced inflammation including glial and lymphocytic reaction. In the heart, signs of chronic cardiomyopathy as well as mild acute lympho-histiocytic myocarditis and vasculitis were present. Although there was no history of COVID-19 for this patient, immunohistochemistry for SARS-CoV-2 antigens (spike and nucleocapsid proteins) was performed. Surprisingly, only spike protein but no nucleocapsid protein could be detected within the foci of inflammation in both the brain and the heart, particularly in the endothelial cells of small blood vessels. Since no nucleocapsid protein could be detected, the presence of spike protein must be ascribed to vaccination rather than to viral infection. The findings corroborate previous reports of encephalitis and myocarditis caused by gene-based COVID-19 vaccines.
Content may be subject to copyright.
Citation: Mörz, M. A Case Report:
Multifocal Necrotizing Encephalitis
and Myocarditis after BNT162b2
mRNA Vaccination against
COVID-19. Vaccines 2022,10, 1651.
https://doi.org/10.3390/
vaccines10101651
Academic Editor: Sung Ryul Shim
Received: 31 August 2022
Accepted: 27 September 2022
Published: 1 October 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the author.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Case Report
A Case Report: Multifocal Necrotizing Encephalitis and Myocarditis
after BNT162b2 mRNA Vaccination against COVID-19
Michael Mörz
Institute of Pathology ’Georg Schmorl’, The Municipal Hospital Dresden-Friedrichstadt, Friedrichstrasse 41,
01067 Dresden, Germany; michael.moerz@klinikum-dresden.de
Abstract:
The current report presents the case of a 76-year-old man with Parkinson’s disease (PD)
who died three weeks after receiving his third COVID-19 vaccination. The patient was first vaccinated
in May 2021 with the ChAdOx1 nCov-19 vector vaccine, followed by two doses of the BNT162b2
mRNA vaccine in July and December 2021. The family of the deceased requested an autopsy
due to ambiguous clinical signs before death. PD was confirmed by post-mortem examinations.
Furthermore, signs of aspiration pneumonia and systemic arteriosclerosis were evident. However,
histopathological analyses of the brain uncovered previously unsuspected findings, including acute
vasculitis (predominantly lymphocytic) as well as multifocal necrotizing encephalitis of unknown
etiology with pronounced inflammation including glial and lymphocytic reaction. In the heart, signs
of chronic cardiomyopathy as well as mild acute lympho-histiocytic myocarditis and vasculitis were
present. Although there was no history of COVID-19 for this patient, immunohistochemistry for
SARS-CoV-2 antigens (spike and nucleocapsid proteins) was performed. Surprisingly, only spike
protein but no nucleocapsid protein could be detected within the foci of inflammation in both the
brain and the heart, particularly in the endothelial cells of small blood vessels. Since no nucleocapsid
protein could be detected, the presence of spike protein must be ascribed to vaccination rather than
to viral infection. The findings corroborate previous reports of encephalitis and myocarditis caused
by gene-based COVID-19 vaccines.
Keywords:
COVID-19 vaccination; necrotizing encephalitis; myocarditis; detection of spike protein;
detection of nucleocapsid protein; autopsy
1. Introduction
The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
in 2019 with the subsequent worldwide spread of COVID-19 gave rise to a perceived need
for halting the progress of the COVID-19 pandemic through the rapid development and
deployment of vaccines. Recent advances in genomics facilitated gene-based strategies
for creating these novel vaccines, including DNA-based nonreplicating viral vectors, and
mRNA-based vaccines, which were furthermore developed on an aggressively shortened
timeline [14].
The WHO Emergency Use Listing Procedure (EUL), which determines the acceptabil-
ity of medicinal products based on evidence of quality, safety, efficacy, and performance [
5
],
permitted these vaccines to be marketed as soon as 1–2 years after development had begun.
Published results of the phase 3 clinical trials described only a few severe side effects [
2
,
6
8
].
However, it has since become clear that severe and even fatal adverse events may occur;
these include in particular cardiovascular and neurological manifestations [
9
13
]. Clini-
cians should take note of such case reports for the sake of early detection and management
of such adverse events among their patients. In addition, a thorough post-mortem ex-
amination of deaths in connection with COVID-19 vaccination should be considered in
ambiguous circumstances, including histology. This report presents the case of a senior aged
76 years old, who had received three doses overall of two different COVID-19 vaccines,
Vaccines 2022,10, 1651. https://doi.org/10.3390/vaccines10101651 https://www.mdpi.com/journal/vaccines
Vaccines 2022,10, 1651 2 of 17
and who died three weeks after the second dose of the mRNA-BNT162b-vaccine. Autopsy
and histology revealed unexpected necrotizing encephalitis and mild myocarditis with
pathological changes in small blood vessels. A causal connection of these findings to
the preceding COVID-19 vaccination was established by immunohistochemical demon-
stration of SARS-CoV-2 spike protein. The methodology introduced in this study should
be useful for distinguishing between causation by COVID-19 vaccination or infection in
ambiguous cases.
2. Materials and Methods
2.1. Routine Histology
Formalin-fixed tissues were routinely processsed and paraffin-embedded tissues were
cut into 5
µ
m sections and stained with hematoxylin and eosin (H&E) for histopathologi-
cal examination.
2.2. Immunohistochemistry
Immunohistochemical staining was performed on the heart and brain, using a fully
automated immunostaining system (Ventana Benchmark, Roche). An antigen retrieval
(Ultra CC1, Roche Ventana) was used for every antibody. The target antigens and dilution
factors for the antibodies used are summarized in Table 1. Incubation with the primary
antibody was carried out for 30 min in each case. Tissues from SARS-CoV-2-positive
COVID-19 patients were used as a control for the antibodies against SARS-CoV-2-spike
and nucleocapsid (Figure 1). Cultured cells that had been transfected
in vitro
(see hereafter)
served as a positive control for the detection of vaccine-induced spike protein expression
and as a negative control for the detection of nucleocapsid protein. The slides were
examined with a light microscope (Nikon ECLIPSE 80i) and representative images were
captured by the camera system Motic®Europe Motic MP3.
Table 1.
Primary antibodies used for immunohistochemistry. Tissue sections were incubated 30 min
with the antibody in question, diluted as stated in the table.
Target Antigen Manufacturer Clone Dilution Incubation Time
CD3 (expressed by T-Lymphocytes) cytomed ZM-45 1:200 30 min
CD68 (expressed by monocytic cells) DAKO PG-M1 1:100 30 min
SARS-CoV-2-Spike subunit 1 ProSci 9083 1:500 30 min
SARS-CoV-2-Nucleocapsid ProSci 35–720 1:500 30 min
Vaccines 2022, 10, 1651 2 of 17
tion, a thorough post-mortem examination of deaths in connection with COVID-19 vac-
cination should be considered in ambiguous circumstances, including histology. This
report presents the case of a senior aged 76 years old, who had received three doses
overall of two different COVID-19 vaccines, and who died three weeks after the second
dose of the mRNA-BNT162b-vaccine. Autopsy and histology revealed unexpected ne-
crotizing encephalitis and mild myocarditis with pathological changes in small blood
vessels. A causal connection of these findings to the preceding COVID-19 vaccination
was established by immunohistochemical demonstration of SARS-CoV-2 spike protein.
The methodology introduced in this study should be useful for distinguishing between
causation by COVID-19 vaccination or infection in ambiguous cases.
2. Materials and Methods
2.1. Routine Histology
Formalin-fixed tissues were routinely processsed and paraffin-embedded tissues
were cut into 5 μm sections and stained with hematoxylin and eosin (H&E) for histo-
pathological examination.
2.2. Immunohistochemistry
Immunohistochemical staining was performed on the heart and brain, using a fully
automated immunostaining system (Ventana Benchmark, Roche). An antigen retrieval
(Ultra CC1, Roche Ventana) was used for every antibody. The target antigens and dilu-
tion factors for the antibodies used are summarized in Table 1. Incubation with the pri-
mary antibody was carried out for 30 min in each case. Tissues from
SARS-CoV-2-positive COVID-19 patients were used as a control for the antibodies
against SARS-CoV-2-spike and nucleocapsid (Figure 1). Cultured cells that had been
transfected in vitro (see hereafter) served as a positive control for the detection of vac-
cine-induced spike protein expression and as a negative control for the detection of nu-
cleocapsid protein. The slides were examined with a light microscope (Nikon ECLIPSE
80i) and representative images were captured by the camera system Motic® Europe Motic
MP3.
Table 1. Primary antibodies used for immunohistochemistry. Tissue sections were incubated 30
min with the antibody in question, diluted as stated in the table.
Target Antigen
Manufacturer
Clone
Dilution
Incubation Time
CD3 (expressed by T-Lymphocytes)
cytomed
ZM-45
1:200
30 min
CD68 (expressed by monocytic cells)
DAKO
PG-M1
1:100
30 min
SARS-CoV-2-Spike subunit 1
ProSci
9083
1:500
30 min
SARS-CoV-2-Nucleocapsid
ProSci
35720
1:500
30 min
Figure 1.
Nasal smear from a person with acute symptomatic SARS-CoV-2-infection (confirmed by
PCR). Note the presence of ciliated epithelium. Immunohistochemistry for two SARS-CoV-2 antigens
(spike and nucleocapsid protein) revealed a positive reaction for both as to be expected after infection.
(
a
) Detection of the spike protein. Positive control for spike subunit 1 SARS-CoV-2 protein detection. Several
Vaccines 2022,10, 1651 3 of 17
ciliated epithelia of the nasal mucosa show brownish granular deposits of DAB (red arrow). Com-
pared to nucleocapsid, the DAB-granules are fewer and less densely packed granular deposits of
DAB. (
b
) Detection of nucleocapsid protein. Positive control for nucleocapsid SARS-CoV-2 protein
detection. Several ciliated epithelia of the nasal mucosa show dense brownish granular deposits of
DAB in immunohistochemistry (examples red arrows). Compared to spike detection, the granules of
DAB are finer and more densely packed. Magnification: 400x.
2.3. Preparation of Positive Control Samples for the Immunohistochemical Detection of the
Vaccine-Induced Spike Protein
Cell culture and transfection: Ovarian cancer cell lines (OVCAR-3 and SK-OV3, CSL
cell Lines Service, Heidelberg, Germany) were grown to 70% confluence in flat bottom
75 cm
2
cell culture flasks (Cell star) in DMEM/HAMS-F12 medium supplemented with
Glutamax (Sigma-Aldrich, St. Louis, MO, USA), 10% FCS (Gibco, Shanghai, China) and
Gentamycin (final concentration 20
µ
g/mL, Gibco), at 37
C, 5% CO
2
in a humidified cell
incubator. For transfection, the medium was completely removed, and cells were incubated
for 1 h with 2 mL of fresh medium containing the injection solutions directly from the
original bottles, diluted 1:500 in the case of BNT162b2 (Pfizer/Biotech), and 1:100 in cases of
mRNA-1273 (Moderna), Vaxzevria (AstraZeneca), and Jansen (COVID-19 vaccine Jansen).
Then, another 15 mL of fresh medium was added to the cell cultures and cells were grown
to confluence for another 3 days.
Preparation of tissue blocks from transfected cells: The cell culture medium was
removed from transfected cells, and the monolayer was washed twice with PBS, then
trypsinized by adding 1 mL of 0.25% Trypsin-EDTA (Gibco), harvested with 10 mL of
PBS/10% FCS, and washed 2
×
with PBS and centrifugation at 280
×
gfor 10 min each.
Cell pellets were fixed overnight in 2 mL in PBS/4% Formalin at 8
C and then washed
in PBS once. The cell pellets remaining after centrifugation were suspended in 200
µ
L
PBS each, mixed with 400
µ
L 2% agarose in PBS solution (precooled to around 40
C),
and immediately transferred to small (1 cm) dishes for fixation. The fixed and agarose-
embedded cell pellets were stored in 4% Formalin/PBS till subjection to routine paraffin
embedding in parallel to tissue samples.
2.4. Case Presentation and Description
2.4.1. Clinical History
This report presents the case of a 76-year-old male with a history of Parkinson’s
disease (PD) who passed away three weeks after his third COVID-19 vaccination. On the
day of his first vaccination in May 2021 (ChAdOx1 nCov-19 vector vaccine), he experienced
pronounced cardiovascular side effects, for which he repeatedly had to consult his doctor.
After the second vaccination in July 2021 (BNT162b2 mRNA vaccine/Comirnaty), the family
noted obvious behavioral and psychological changes (e.g., he did not want to be touched
anymore and experienced increased anxiety, lethargy, and social withdrawal even from
close family members). Furthermore, there was a striking worsening of his PD symptoms,
which led to severe motor impairment and a recurrent need for wheelchair support. He
never fully recovered from these side effects after the first two vaccinations but still got
another vaccination in December 2021. Two weeks after the third vaccination (second
vaccination with BNT162b2), he suddenly collapsed while taking his dinner. Remarkably,
he did not show coughing or any signs of food aspiration but just fell down silently. He
recovered from this more or less, but one week later, he again suddenly collapsed silently
while taking his meal. The emergency unit was called, and after successful, but prolonged
resuscitation attempts (over one hour), he was transferred to the hospital and directly put
into an artificial coma but died shortly thereafter. The clinical diagnosis was death due
to aspiration pneumonia. According to his family, there was no history of a clinical or
laboratory diagnosis of COVID-19 in the past.
Vaccines 2022,10, 1651 4 of 17
2.4.2. Autopsy
The autopsy was requested and consented to by the family of the patient because of the
ambiguity of symptoms before his death. The autopsy was performed according to standard
procedures including macroscopic and microscopic investigation. Gross brain tissue was
prepared for histological examination including the brain (frontal cortex, Substantia nigra,
and Nucleus ruber) as well as the heart (left and right ventricular cardiac tissue).
3. Results
3.1. Autopsy Findings
Anatomical Specifications: Body weight, height, and specifications of body organs
were summarized in Table 2.
Table 2. Anatomical Specifications.
Item Measure
Body weight 60 kg
Hight 175 cm
Heart weight 410 g
Brain weight 1560 g
Liver weight 1500 g
Brain: A macroscopic examination of brain tissue revealed a circumscribed segmen-
tal cerebral parenchymal necrosis at the site of the right hippocampus. Substantia nigra
showed a loss of pigmented neurons. Microscopically, several areas with lacunar necrosis
were detected with inflammatory debris reaction on the left frontal side (Figure 2). Stain-
ing of Nucleus ruber with H&E showed neuronal cell death, microglia, and lymphocyte
infiltration (Figure 3). Furthermore, there were microglial and lymphocytic reactions as
well as predominantly lymphocytic vasculitis, sometimes with mixed infiltrates includ-
ing neutrophilic granulocytes (Figure 4) in the frontal cortex, paraventricular, Substantia
nigra, and Nucleus ruber on both sides. In some places with inflammatory changes in
brain capillaries, there were also signs of apoptotic cell death within the endothelium
(Figure 4). Meninges’ findings were unremarkable. The collective findings were sugges-
tive of multifocal necrotizing encephalitis. Furthermore, chronic arteriosclerotic lesions of
varying degrees were noted in large brain vessels, which are described in detail in section
“Vascular system”.
Parkinson’s disease (PD): Macroscopic and histological examination of brain tis-
sue revealed bilateral pallor of the substantia nigra with loss of pigmented neurons.
In addition, pigment-storing macrophages as well as scattered neuronal necrosis with
glial debris reaction were noted. These findings were suggestive of PD, confirming the
clinical diagnosis.
Thoracic cavity: An examination of the chest showed a funnel-shaped chest with
serial rib fractures (extending from the second to fifth ribs on the right, and from the
second to sixth ribs on the left); which is a common picture of a patient who underwent
cardiopulmonary resuscitation. An endotracheal tube was properly inserted. There was
evidence of regular placement of a central venous catheter in the left femoral vein. There
was evidence of regular placement of an arterial catheter in the left radial artery. The
urinary catheter was inserted as well. There was a 9 cm long skin scar on the front of the
right shoulder.
Vaccines 2022,10, 1651 5 of 17
Figure 2.
Frontal brain. Already in the overview image (
a
), prominent vacuolations with in-
creased parenchymal cellularity are evident, indicative of degenerative and inflammatory processes.
At higher magnification (
b
), acute brain damage is visible with diffuse and zonal neuronal and glial
cell death, activation of microglia, and inflammatory infiltration by granulocytes and lymphocytes.
1: neuronal deaths (cells with red cytoplasm); 2: microglial proliferation; 3: lymphocytes. H&E stain.
Magnification 40×(a) and 200×(b).
Vaccines 2022, 10, 1651 6 of 17
Figure 2. Frontal brain. Already in the overview image (a), prominent vacuolations with increased
parenchymal cellularity are evident, indicative of degenerative and inflammatory processes. At
higher magnification (b), acute brain damage is visible with diffuse and zonal neuronal and glial
cell death, activation of microglia, and inflammatory infiltration by granulocytes and lymphocytes.
1: neuronal deaths (cells with red cytoplasm); 2: microglial proliferation; 3: lymphocytes. H&E
stain. Magnification 40× (a) and 200× (b).
Figure 3. Brain, Nucleus ruber. In the overview image (a), note pronounced focal necrosis with
increased cellularity, indicative of ongoing inflammation and glial reaction. At higher
magnification (b), death of neuronal cells is evident and associated with an increased number of
glial cells. Note activation of microglia and presence of inflammatory cell infiltrates, predominantly
lymphocytic. 1: neuronal death with hypereosinophilia and destruction of cell nucleus with signs
of karyolysis (nuclear content being distributed into the cytoplasm); 2: microglia (example); 3:
lymphocyte (example). H&E stain. Magnification 40× (a) and 400× (b).
Figure 3.
Brain, Nucleus ruber. In the overview image (
a
), note pronounced focal necrosis with
increased cellularity, indicative of ongoing inflammation and glial reaction. At higher magnification
(
b
), death of neuronal cells is evident and associated with an increased number of glial cells. Note
activation of microglia and presence of inflammatory cell infiltrates, predominantly lymphocytic.
1: neuronal death with hypereosinophilia and destruction of cell nucleus with signs of karyolysis
(nuclear content being distributed into the cytoplasm); 2: microglia (example); 3: lymphocyte
(example). H&E stain. Magnification 40×(a) and 400×(b).
Vaccines 2022,10, 1651 6 of 17
Vaccines 2022, 10, 1651 7 of 17
Figure 4. Brain, periventricular vasculitis. Cross section through a capillary vessel showing
prominent signs of vasculitis. The endothelial cells (5) show swelling and vacuolation and are
increased in number with enlargement of nuclei, indicative for activation. Furthermore, presence of
mixed inflammatory cell infiltrates within the endothelial layer, consisting of lymphocytes (1),
granulocytes (2), and histiocytes (4). The adjacent brain tissue also shows signs of inflammation
(encephalitis) with presence of lymphocytes as well and activated microglia (3). H&E.
Magnification: 200× (a) and 400× (b).
Figure 4.
Brain, periventricular vasculitis. Cross section through a capillary vessel showing prominent
signs of vasculitis. The endothelial cells (5) show swelling and vacuolation and are increased in
number with enlargement of nuclei, indicative for activation. Furthermore, presence of mixed
inflammatory cell infiltrates within the endothelial layer, consisting of lymphocytes (1), granulocytes
(2), and histiocytes (4). The adjacent brain tissue also shows signs of inflammation (encephalitis)
with presence of lymphocytes as well and activated microglia (3). H&E. Magnification: 200
×
(
a
) and
400×(b).
Lungs: Macroscopical lung examination revealed cloudy secretion and purulent spots
with notably brittle parenchyma. The pleura showed bilateral serous effusion, amounting
to 450 mL of fluid on the right side and 400 mL on the left side. Bilateral mucopurulent
tracheobronchitis was evident with copious purulent secretion in the trachea and bronchi.
Bilateral chronic destructive pulmonary emphysema was detected. Bilateral bronchopneu-
monia was noted in the lower lung lobes at multiple stages of development and lobe-filling
with secretions and fragile parenchyma. Furthermore, chronic arteriosclerotic lesions of
varying degrees were noted, which are described in detail in the section “Vascular system”.
Heart: Macroscopic cardiac examination revealed manifestations of acute and chronic
cardiovascular insufficiency, including ectasia of the atria and ventricles. Furthermore,
left ventricular hypertrophy was noted (wall thickness: 18 mm, heart weight: 410 g, body
weight: 60 kg, height: 1.75 m). There was evidence of tissue congestion (presumably
due to cardiac insufficiency) in the form of pulmonary edema, cerebral edema, brain
congestion, chronic hepatic congestion, renal tissue edema, and pituitary tissue edema.
Moreover, there was evidence of shock kidney disorder. Histological examination of
the heart revealed mild myocarditis with fine-spotted fibrosis and lympho-histiocytic
infiltration (Figure 5). Furthermore, there were chronic arteriosclerotic lesions of varying
degrees, which are described in detail under “Vascular system”. In addition to these, there
were more acute myocardial and vascular changes in the heart. They consisted of mild
signs of myocarditis, characterized by infiltrations with foamy histiocytes and lymphocytes
as well as hypereosinophilia and some hypercontraction of cardiomyocytes. Furthermore,
mild acute vascular changes were observed in the capillaries and other small blood vessels
of the heart. They consisted of mild lympho-histiocytic infiltrates, prominent endothelial
swelling and vacuolation, multifocal myocytic degeneration and coagulation necrosis as
well as karyopyknosis of single endothelial cells and vascular muscle cells (Figure 5).
Occasionally, adhering plasma coagulates/fibrin clots were present on the endothelial
surface, indicative of endothelial damage (Figure 5).
Vaccines 2022,10, 1651 7 of 17
Vaccines 2022, 10, 1651 7 of 17
Figure 4. Brain, periventricular vasculitis. Cross section through a capillary vessel showing
prominent signs of vasculitis. The endothelial cells (5) show swelling and vacuolation and are
increased in number with enlargement of nuclei, indicative for activation. Furthermore, presence of
mixed inflammatory cell infiltrates within the endothelial layer, consisting of lymphocytes (1),
granulocytes (2), and histiocytes (4). The adjacent brain tissue also shows signs of inflammation
(encephalitis) with presence of lymphocytes as well and activated microglia (3). H&E.
Magnification: 200× (a) and 400× (b).
Figure 5.
Heart left ventricle. (
a
): Mild lympho-histiocytic myocarditis.Pronounced interstitial
edema (7) and mild lympho-histiocytic infiltrates (2 + 4). Signs of cardiomyocytic degeneration
(5) with cytoplasmic hypereosinophilia and single contraction bands. (
d
): Arteriole with signs of
acute degeneration and associated inflammation, associated by lymphocytic infiltrates (2) within
the vascular wall, endothelial swelling and vacuolation (3), and vacuolation of vascular myocytes
with signs of karyopyknosis (1). Within the vascular lumen (
d
), note plasma coagulation/fibrin
clots adhering to the endothelial surface, indicative of endothelial damage. 1: pyknotic vascular
myocytes, 2: lymphocytes, 3: swollen endothelial cells, 4: macrophages, 5: necrotic cardiomyocytes,
6: eosinophilic granulocytes, 7 (blue line): interstitial edema. H&E stain. Magnification: 200
×
(
a
) and
(c), 40×(b), and detailed enlargement (d).
Vascular system (large blood vessels): The pulmonary arteries showed ectasia and
lipidosis. The kidney showed slight diffuse glomerulosclerosis and arteriosclerosis with
renal cortical scars (up to 10 mm in diameter). The findings are suggestive of generalized
atherosclerosis and systemic hypertension. Major arteries including the aorta and its
branches as well as the coronary arteries showed variable degrees of arteriosclerosis and
mild to moderate stenosis. Furthermore, examination revealed mild nodular arteriosclerosis
of cervical arteries. Ascending aorta, aortic arch, and thoracic aorta showed moderate,
nodular, and partially calcified arteriosclerosis. The cerebral basilar artery showed mild
arteriosclerosis. Nodular and calcified arteriosclerosis were of high grade in the abdominal
aorta and iliac arteries and moderate grade with moderate stenosis in the right coronary
arteries. Coronary artery examination showed variable degrees of arteriosclerosis and
stenosis more on the left coronary arteries. The left anterior descending coronary artery (the
anterior interventricular branch of the left coronary artery; LAD) showed high-grade and
moderately stenosed arteriosclerosis. The arteriosclerosis and stenosis of the left circumflex
artery (the circumflex branch of the left coronary artery) were mild. Mild cerebral basal
artery sclerosis. High-grade nodular and calcified arteriosclerosis of the abdominal aorta
and the iliac arteries. Moderate stenosed arteriosclerosis of the right coronary artery.
Lymphocytic periarteritis was detected as well.
Vaccines 2022,10, 1651 8 of 17
3.2. Other Findings
-
Oral cavity: tongue bite was detected with bleeding under the tongue muscle (tongue
bite is common with epileptic seizures).
- Adrenal glands: bilateral mild cortical hyperplasia.
- Colon: the elongated sigmoid colon was elongated with fecal impaction.
-
Kidneys: slight diffuse glomerulosclerosis and arterio-sclerosis, renal cortical scars
(up to 10 mm in diameter), bilateral mild active nephritis and urocystitis as well as
evidence of shock kidney disorder.
- Liver: slight lipofuscinosis.
- Spleen: mild acute splenitis.
- Stomach: mild diffuse gastric mucosal bleeding.
-
Thyroid gland: bilateral nodular goiter with chocolate cysts (up to 0.5 cm in diameter).
-
Prostate gland: benign nodular prostatic hyperplasia and chronic persistent prostatitis.
3.3. Immunohistochemical Analyses
Immunohistochemical staining for the presence of SARS-CoV-2 antigens (spike protein
and nucleocapsid) was studied in the brain and heart. In the brain, SARS-CoV-2 spike
protein subunit 1 was detected in the endothelia, microglia, and astrocytes in the necrotic
areas (Figures 6and 7). Furthermore, spike protein could be demonstrated in the areas of
lymphocytic periarteritis, present in the thoracic and abdominal aorta and iliac branches,
as well as a cerebral basal artery (Figure 8). The SARS-CoV-2 subunit 1 was found in
macrophages and in the cells of the vessel wall, in particular the endothelium (Figure 9),
as well as in the Nucleus ruber (Figure 10). In contrast, the nucleocapsid protein of SARS-
CoV-2 could not be detected in any of the corresponding tissue sections (Figures 11 and 12).
In addition, SARS-CoV-2 spike protein subunit 1 was detected in the cardiac endothelial
cells that showed lymphocytic myocarditis (Figure 13). Immunohistochemical staining did
not detect the SARS-CoV-2 nucleocapsid protein (Figure 14).
Vaccines 2022, 10, 1651 9 of 17
Figure 6. Frontal brain. Immunohistochemistry for CD68 (expressed by monocytic cells). Note
map-like tissue destruction with the presence of CD68-positive microglial cells. Furthermore zonal
activation of microglia (brown granules). Activation of the microglia means that tissue destruction
has taken place in the brain, which is cleared/removed by macrophages (called microglia in the
brain). Brown granules: macrophages/microglia. Magnification: 40×.
Figure 7. Brain. Nucleus ruber. Immunohistochemistry for CD68 (expressed by monocytic cells)
shows abundant positive cells, indicative of zonal activation of microglia (brown granules). Mag-
nification: 40×.
Figure 6.
Frontal brain. Immunohistochemistry for CD68 (expressed by monocytic cells). Note
map-like tissue destruction with the presence of CD68-positive microglial cells. Furthermore zonal
activation of microglia (brown granules). Activation of the microglia means that tissue destruction
has taken place in the brain, which is cleared/removed by macrophages (called microglia in the
brain). Brown granules: macrophages/microglia. Magnification: 40×.
Vaccines 2022,10, 1651 9 of 17
Vaccines 2022, 10, 1651 9 of 17
Figure 6. Frontal brain. Immunohistochemistry for CD68 (expressed by monocytic cells). Note
map-like tissue destruction with the presence of CD68-positive microglial cells. Furthermore zonal
activation of microglia (brown granules). Activation of the microglia means that tissue destruction
has taken place in the brain, which is cleared/removed by macrophages (called microglia in the
brain). Brown granules: macrophages/microglia. Magnification: 40×.
Figure 7. Brain. Nucleus ruber. Immunohistochemistry for CD68 (expressed by monocytic cells)
shows abundant positive cells, indicative of zonal activation of microglia (brown granules). Mag-
nification: 40×.
Figure 7.
Brain. Nucleus ruber. Immunohistochemistry for CD68 (expressed by monocytic cells)
shows abundant positive cells, indicative of zonal activation of microglia (brown granules). Magnifi-
cation: 40×.
Vaccines 2022, 10, 1651 10 of 17
Figure 8. Frontal brain. Immunohistochemistry for CD3 (expressed by T-Lymphocytes) shows
numerous CD3-positive lymphocytes (brown granules, red arrow highlights an example), partic-
ularly within the endothelium, but also in the brain tissue, indicative of lymphocytic vasculitis and
encephalitis. Blue dotted lines: blood vessels. Magnification: 200×.
Figure 9. Frontal brain. Positive reaction for SARS-CoV-2 spike protein. Cross section through a
capillary vessel (same vessel as shown in Figure 11, serial sections of 5 to 20 µ m). Immunohisto-
chemical reaction for SARS-CoV-2 spike subunit 1 detectable as brown granules in capillary en-
dothelial cells (red arrow) and individual glial cells (blue arrow). Magnification: 200×.
Figure 8.
Frontal brain. Immunohistochemistry for CD3 (expressed by T-Lymphocytes) shows
numerous CD3-positive lymphocytes (brown granules, red arrow highlights an example), particu-
larly within the endothelium, but also in the brain tissue, indicative of lymphocytic vasculitis and
encephalitis. Blue dotted lines: blood vessels. Magnification: 200×.
Vaccines 2022,10, 1651 10 of 17
Vaccines 2022, 10, 1651 10 of 17
Figure 8. Frontal brain. Immunohistochemistry for CD3 (expressed by T-Lymphocytes) shows
numerous CD3-positive lymphocytes (brown granules, red arrow highlights an example), partic-
ularly within the endothelium, but also in the brain tissue, indicative of lymphocytic vasculitis and
encephalitis. Blue dotted lines: blood vessels. Magnification: 200×.
Figure 9. Frontal brain. Positive reaction for SARS-CoV-2 spike protein. Cross section through a
capillary vessel (same vessel as shown in Figure 11, serial sections of 5 to 20 µm). Immunohisto-
chemical reaction for SARS-CoV-2 spike subunit 1 detectable as brown granules in capillary en-
dothelial cells (red arrow) and individual glial cells (blue arrow). Magnification: 200×.
Figure 9.
Frontal brain. Positive reaction for SARS-CoV-2 spike protein. Cross section through a cap-
illary vessel (same vessel as shown in Figure 11, serial sections of 5 to 20
µ
m). Immunohistochemical
reaction for SARS-CoV-2 spike subunit 1 detectable as brown granules in capillary endothelial cells
(red arrow) and individual glial cells (blue arrow). Magnification: 200×.
Vaccines 2022, 10, 1651 11 of 17
Figure 10. Brain, Nucleus ruber. The abundant presence of SARS-CoV-2 spike protein in swollen
endothelium of a capillary vessel shows acute signs of inflammation with sparse mononuclear in-
flammatory cell infiltrates (same vessel as shown in Figure 12, serial sections of 5 to 20 µm). Im-
munohistochemical demonstration for SARS-CoV-2 spike protein subunit 1 visible as brown
granules in capillary endothelial cells (red arrow) and individual glial cells (blue arrow). Magnifi-
cation: 200×.
Figure 11. Frontal brain. Negative immunohistochemical reaction for SARS-CoV-2 nucleocapsid
protein. Cross section through a capillary vessel (same vessel as shown in Figure 9, serial sections
of 5 to 20 µm). Magnification: 200×.
Figure 10.
Brain, Nucleus ruber. The abundant presence of SARS-CoV-2 spike protein in swollen
endothelium of a capillary vessel shows acute signs of inflammation with sparse mononuclear
inflammatory cell infiltrates (same vessel as shown in Figure 12, serial sections of 5 to 20
µ
m). Im-
munohistochemical demonstration for SARS-CoV-2 spike protein subunit 1 visible as brown granules
in capillary endothelial cells (red arrow) and individual glial cells (blue arrow). Magnification: 200
×
.
Vaccines 2022,10, 1651 11 of 17
Vaccines 2022, 10, 1651 11 of 17
Figure 10. Brain, Nucleus ruber. The abundant presence of SARS-CoV-2 spike protein in swollen
endothelium of a capillary vessel shows acute signs of inflammation with sparse mononuclear in-
flammatory cell infiltrates (same vessel as shown in Figure 12, serial sections of 5 to 20 µm). Im-
munohistochemical demonstration for SARS-CoV-2 spike protein subunit 1 visible as brown
granules in capillary endothelial cells (red arrow) and individual glial cells (blue arrow). Magnifi-
cation: 200×.
Figure 11. Frontal brain. Negative immunohistochemical reaction for SARS-CoV-2 nucleocapsid
protein. Cross section through a capillary vessel (same vessel as shown in Figure 9, serial sections
of 5 to 20 µm). Magnification: 200×.
Figure 11.
Frontal brain. Negative immunohistochemical reaction for SARS-CoV-2 nucleocapsid
protein. Cross section through a capillary vessel (same vessel as shown in Figure 9, serial sections of
5 to 20 µm). Magnification: 200×.
Vaccines 2022, 10, 1651 12 of 17
Figure 12. Brain, Nucleus ruber. Negative immunohistochemical reaction for SARS-CoV-2 nucle-
ocapsid protein. Cross section through a capillary vessel (same vessel as shown in Figure 11, serial
sections of 5 to 20 µm). Magnification: 200×.
Figure 13. Heart left ventricle. Positive reaction for SARS-CoV-2 spike protein. Cross section
through a capillary vessel (same vessel as shown in Figure 14, serial sections of 5 to 20 µm). Im-
munohistochemical demonstration of SARS-CoV-2 spike subunit 1 as brown granules. Note the
abundant presence of spike protein in capillary endothelial cells (red arrow) associated with
prominent endothelial swelling and the presence of a few mononuclear inflammatory cells. Mag-
nification: 400×.
Figure 12.
Brain, Nucleus ruber. Negative immunohistochemical reaction for SARS-CoV-2 nucleo-
capsid protein. Cross section through a capillary vessel (same vessel as shown in Figure 11, serial
sections of 5 to 20 µm). Magnification: 200×.
Vaccines 2022,10, 1651 12 of 17
Vaccines 2022, 10, 1651 12 of 17
Figure 12. Brain, Nucleus ruber. Negative immunohistochemical reaction for SARS-CoV-2 nucle-
ocapsid protein. Cross section through a capillary vessel (same vessel as shown in Figure 11, serial
sections of 5 to 20 µm). Magnification: 200×.
Figure 13. Heart left ventricle. Positive reaction for SARS-CoV-2 spike protein. Cross section
through a capillary vessel (same vessel as shown in Figure 14, serial sections of 5 to 20 µ m). Im-
munohistochemical demonstration of SARS-CoV-2 spike subunit 1 as brown granules. Note the
abundant presence of spike protein in capillary endothelial cells (red arrow) associated with
prominent endothelial swelling and the presence of a few mononuclear inflammatory cells. Mag-
nification: 400×.
Figure 13.
Heart left ventricle. Positive reaction for SARS-CoV-2 spike protein. Cross section through
a capillary vessel (same vessel as shown in Figure 14, serial sections of 5 to 20
µ
m). Immunohistochem-
ical demonstration of SARS-CoV-2 spike subunit 1 as brown granules. Note the abundant presence of
spike protein in capillary endothelial cells (red arrow) associated with prominent endothelial swelling
and the presence of a few mononuclear inflammatory cells. Magnification: 400×.
Vaccines 2022, 10, 1651 13 of 17
Figure 14. Heart left ventricle. Negative immunohistochemical reaction for SARS-CoV-2 nucle-
ocapsid protein. Cross section through a capillary vessel (same vessel as shown in Figure 13, serial
sections of 5 to 20 µm). Magnification: 400×.
3.4. Autopsy-Based Diagnosis
The 76-year-old deceased male patient had PD, which corresponded to typical
post-mortem findings. The main cause of death was recurrent aspiration pneumonia. In
addition, necrotizing encephalitis and vasculitis were considered to be major contribu-
tors to death. Furthermore, there was mild lympho-histiocytic myocarditis with fi-
ne-spotted myocardial fibrosis as well as systemic arteriosclerosis, which will have also
contributed to the deterioration of the physical condition of the senior.
The final diagnosis was abscedating bilateral bronchopneumonia (J18.9), Parkin-
son’s disease (G20.9), necrotic encephalitis (G04.9), and myocarditis (I40.9).
Immunohistochemistry for SARS-CoV-2 antigens (spike protein and nucleocapsid)
revealed that the lesions with necrotizing encephalitis as well as the acute inflammatory
changes in the small blood vessels (brain and heart) were associated with abundant de-
posits of the spike protein SARS-CoV-2 subunit 1. Since the nucleocapsid protein of
SARS-CoV-2 was consistently absent, it must be assumed that the presence of spike pro-
tein in affected tissues was not due to an infection with SARS-CoV-2 but rather to the
transfection of the tissues by the gene-based COVID-19-vaccines. Importantly, spike
protein could be only demonstrated in the areas with acute inflammatory reactions
(brain, heart, and small blood vessels), in particular in endothelial cells, microglia, and
astrocytes. This is strongly suggestive that the spike protein may have played at least a
contributing role to the development of the lesions and the course of the disease in this
patient.
4. Discussion
This is a case report of a 76-year-old patient with Parkinson’s disease (PD) who died
three weeks after his third COVID-19 vaccination. The stated cause of death appeared to
be a recurrent attack of aspiration pneumonia, which is indeed common in PD [14,15].
However, the detailed autopsy study revealed additional pathology, in particular ne-
crotizing encephalitis and myocarditis. While the histopathological signs of myocarditis
were comparatively mild, the encephalitis had resulted in significant multifocal necrosis
and may well have contributed to the fatal outcome. Encephalitis often causes epileptic
seizures, and the tongue bite found at the autopsy suggests that it had done so in this
Figure 14.
Heart left ventricle. Negative immunohistochemical reaction for SARS-CoV-2 nucleocapsid
protein. Cross section through a capillary vessel (same vessel as shown in Figure 13, serial sections of
5 to 20 µm). Magnification: 400×.
3.4. Autopsy-Based Diagnosis
The 76-year-old deceased male patient had PD, which corresponded to typical post-
mortem findings. The main cause of death was recurrent aspiration pneumonia. In
addition, necrotizing encephalitis and vasculitis were considered to be major contributors
to death. Furthermore, there was mild lympho-histiocytic myocarditis with fine-spotted
myocardial fibrosis as well as systemic arteriosclerosis, which will have also contributed to
the deterioration of the physical condition of the senior.
Vaccines 2022,10, 1651 13 of 17
The final diagnosis was abscedating bilateral bronchopneumonia (J18.9), Parkinson’s
disease (G20.9), necrotic encephalitis (G04.9), and myocarditis (I40.9).
Immunohistochemistry for SARS-CoV-2 antigens (spike protein and nucleocapsid)
revealed that the lesions with necrotizing encephalitis as well as the acute inflammatory
changes in the small blood vessels (brain and heart) were associated with abundant deposits
of the spike protein SARS-CoV-2 subunit 1. Since the nucleocapsid protein of SARS-CoV-2
was consistently absent, it must be assumed that the presence of spike protein in affected
tissues was not due to an infection with SARS-CoV-2 but rather to the transfection of
the tissues by the gene-based COVID-19-vaccines. Importantly, spike protein could be
only demonstrated in the areas with acute inflammatory reactions (brain, heart, and small
blood vessels), in particular in endothelial cells, microglia, and astrocytes. This is strongly
suggestive that the spike protein may have played at least a contributing role to the
development of the lesions and the course of the disease in this patient.
4. Discussion
This is a case report of a 76-year-old patient with Parkinson’s disease (PD) who died
three weeks after his third COVID-19 vaccination. The stated cause of death appeared
to be a recurrent attack of aspiration pneumonia, which is indeed common in PD [
14
,
15
].
However, the detailed autopsy study revealed additional pathology, in particular necrotiz-
ing encephalitis and myocarditis. While the histopathological signs of myocarditis were
comparatively mild, the encephalitis had resulted in significant multifocal necrosis and may
well have contributed to the fatal outcome. Encephalitis often causes epileptic seizures, and
the tongue bite found at the autopsy suggests that it had done so in this case. Several other
cases of COVID-19 vaccine-associated encephalitis with status epilepticus have appeared
previously [1618].
The clinical history of the current case showed some remarkable events in correlation
to his COVID-19 vaccinations. Already on the day of his first vaccination in May 2021
(ChAdOx1 nCov-19 vector vaccine), he experienced cardiovascular symptoms, which
needed medical care and from which he recovered only slowly. After the second vaccination
in July 2021 (BNT162b2 mRNA vaccine), the family recognized remarkable behavioral and
psychological changes and a sudden onset of marked progression of his PD symptoms,
which led to severe motor impairment and recurrent need for wheelchair support. He
never fully recovered from this but still was again vaccinated in December 2021. Two weeks
after this third vaccination (second vaccination with BNT162b2), he suddenly collapsed
while taking his dinner. Remarkably, he did not show any coughing or other signs of food
aspiration but just fell from his chair. This raises the question of whether this sudden
collapse was really due to aspiration pneumonia. After intense resuscitation, he recovered
from this more or less, but one week later, he again suddenly collapsed silently while taking
his meal. After successful but prolonged resuscitation attempts, he was transferred to the
hospital and directly set into an artificial coma but died shortly thereafter. The clinical
diagnosis was death due to aspiration pneumonia. Due to his ambiguous symptoms after
the COVID-vaccinations the family asked for an autopsy.
Based on the alteration pattern in the brain and heart, it appeared that the small blood
vessels were especially affected, in particular, the endothelium. Endothelial dysfunction is
known to be highly involved in organ dysfunction during viral infections, as it induces a
pro-coagulant state, microvascular leak, and organ ischemia [
19
,
20
]. This is also the case for
severe SARS-CoV-2 infections, where a systemic exposure to the virus and its spike protein
elicits a strong immunological reaction in which the endothelial cells play a crucial role,
leading to vascular dysfunction, immune-thrombosis, and inflammation [21].
Although there was no history of COVID-19 for this patient, immunohistochemistry
for SARS-CoV-2 antigens (spike and nucleocapsid proteins) was performed. Spike protein
could be indeed demonstrated in the areas of acute inflammation in the brain (particularly
within the capillary endothelium) and the small blood vessels of the heart. Remarkably,
however, the nucleocapsid was uniformly absent. During an infection with the virus, both
Vaccines 2022,10, 1651 14 of 17
proteins should be expressed and detected together. On the other hand, the gene-based
COVID-19 vaccines encode only the spike protein and therefore, the presence of spike
protein only (but no nucleocapsid protein) in the heart and brain of the current case can
be attributed to vaccination rather than to infection. This agrees with the patient’s history,
which includes three vaccine injections, the third one just 3 weeks before his death, but no
positive laboratory or clinical diagnosis of the infection.
Discrimination of vaccination response from natural infection is an important question
and had been addressed already in clinical immunology, where the combined application
of anti-spike and anti-nucleocapsid protein-based serology was proven as a useful tool [
22
].
In histology, however, this immunohistochemical approach has not yet been described, but
it is straightforward and appears to be very useful for identifying the potential origin of
SARS-CoV-2 spike protein in autopsy or biopsy samples. Where additional confirmation is
required, for instance in a forensic context, rt-PCR methods might be used to ascertain the
presence of the vaccine mRNA in the affected tissues [23,24].
Assuming that, in the current case, the presence of spike protein was indeed driven
by the gene-based vaccine, then the question arises whether this was also the cause the
accompanying acute tissue alterations and inflammation. The stated purpose of the gene-
based vaccines is to induce an immune response against the spike protein. Such an immune
response will, however, not only results in antibody formation against the spike protein but
also lead to direct cell- and antibody-mediated cytotoxicity against the cells expressing this
foreign antigen. In addition, there are indications that the spike protein on its own can elicit
distinct toxicity, in particular, on pericytes and endothelial cells of blood vessels [25,26].
While it is widely held that spike protein expression, and the ensuing cell and tissue
damage will be limited to the injection site, several studies have found the vaccine mRNA
and/or the spike protein encoded by it at a considerable distance from the injection site
for up to three months after the injection [
23
,
24
,
27
29
]. Biodistribution studies in rats with
the mRNA-COVID-19 vaccine BNT162b2 also showed that the vaccine does not stay at the
injection site but is distributed to all tissues and organs, including the brain [
30
]. After the
worldwide roll-out of COVID-19 vaccinations in humans, spike protein has been detected
in humans as well in several tissues distant from the injection site (deltoid muscle): for
instance in heart muscle biopsies from myocarditis patients [
28
], within the skeletal muscle
of a patient with myositis [
23
] and within the skin, where it was associated with a sudden
onset of Herpes zoster lesions after mRNA-COVID-19 vaccination [29].
The underlying diagnosis in this patient was Parkinson’s disease, and one may ask
what role, if any, this condition had played in the causation of the encephalitis, and the
myocarditis detected at post-mortem examination. PD had been long-standing in the cur-
rent case, whereas the encephalitis was acute. Conversely, there is no plausible mechanism
and no case report of PD causing secondary necrotizing encephalitis. On the other hand,
numerous cases have been reported of autoimmune encephalitis and encephalomyelitis
after COVID-19 vaccination [
12
,
31
]. Autoimmune diseases in organs other than the CNS
have been reported as well, for example, a striking case of a patient who after mRNA
vaccination suffered multiple autoimmune disorders all at once—acute disseminated en-
cephalomyelitis, myasthenia gravis, and thyroiditis [
32
]. In the case reported here, it may
be noted that the spike protein was primarily detected in the vascular endothelium and
sparsely in the glial cells but not in the neurons. Nevertheless, neuronal cell death was
widespread in the encephalitic foci, which suggests some contribution of immunological
bystander activation, i.e., autoimmunity, to the observed cell and tissue damage.
A contributory role of PD in the development of cardiomyopathy is indeed docu-
mented and cannot be ruled out with absolute certainty. However, inflammatory myocar-
dial changes with pathological alterations in small blood vessels as seen in the current case
are uncommon. Instead, the most prominent cause of cardiac failure in PD patients is rather
due to cardiac autonomic dysfunction [
33
,
34
]. PD seems well to be significantly associated
with increased left ventricular hypertrophy and diastolic dysfunction [
34
]. In the current
case, ventricular dilatation and hypertrophy were present but seem rather related to mani-
Vaccines 2022,10, 1651 15 of 17
fest signs of chronic hypertension. In contrast, myocardial inflammatory reactions had been
well-linked to gene-based COVID-19 vaccinations in numerous cases [
9
,
35
37
]. In one case,
the spike protein of SARS-CoV-2 could also be demonstrated by immunohistochemistry in
the heart of vaccinated individuals [28].
5. Conclusions
Numerous cases of encephalitis and encephalomyelitis have been reported in connec-
tion with the gene-based COVID-19 vaccines, with many being considered causally related
to vaccination [
31
,
38
,
39
]. However, this is the first report to demonstrate the presence of
the spike protein within the encephalitic lesions and to attribute it to vaccination rather
than infection. These findings corroborate a causative role of the gene-based COVID-19
vaccines, and this diagnostic approach is relevant to potentially vaccine-induced damage
to other organs as well.
Funding: This research received no specific funding.
Institutional Review Board Statement:
According to the Saxonian State Chamber of Medicine
(Ethikkommission Landesärztekammer Sachsen), no explicit ethical approval is required for autopsy
case reports as long as informed consent was obtained from the entitled person and all data has
been anonymized.
Informed Consent Statement:
The informed consent was obtained from the entitled person for the
subject involved in this case report.
Data Availability Statement: Data are available upon request.
Acknowledgments:
The author wishes to thank Hany A. Salem and David O. Fischer for supporting
the preparation of this paper with valuable comments and suggestions.
Conflicts of Interest: The author declares he has no conflict of interest.
References
1.
Chen, W.-H.; Strych, U.; Hotez, P.J.; Bottazzi, M.E. The SARS-CoV-2 Vaccine Pipeline: An Overview. Curr. Trop. Med. Rep.
2020, 1–4. [CrossRef] [PubMed]
2.
Sultana, J.; Mazzaglia, G.; Luxi, N.; Cancellieri, A.; Capuano, A.; Ferrajolo, C.; de Waure, C.; Ferlazzo, G.; Trifirò, G. Potential
effects of vaccinations on the prevention of COVID-19: Rationale, clinical evidence, risks, and public health considerations. Expert
Rev. Vaccines 2020,19, 919–936. [CrossRef] [PubMed]
3.
WHO. COVID-19 Vaccine Tracker and Landscape. Available online: https://www.who.int/publications/m/item/draft-
landscape-of-covid-19-candidate-vaccines (accessed on 2 June 2022).
4.
Lurie, N.; Saville, M.; Hatchett, R.; Halton, J. Developing COVID-19 Vaccines at Pandemic Speed. N. Engl. J. Med.
2020
,382,
1969–1973. [CrossRef]
5.
World Health Organization (WHO). Diagnostics Laboratory Emergency Use Listing. Available online: https://www.who.int/
teams/regulation-prequalification/eul (accessed on 2 June 2022).
6.
Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al.
Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021,384, 403–416. [CrossRef] [PubMed]
7. O’Reilly, P. A phase III study to investigate a vaccine against COVID-19. ISRCTN 2020. [CrossRef]
8.
Polak, S.B.; Van Gool, I.C.; Cohen, D.; von der Thüsen, J.H.; van Paassen, J. A systematic review of pathological findings in
COVID-19: A pathophysiological timeline and possible mechanisms of disease progression. Mod. Pathol.
2020
,33, 2128–2138.
[CrossRef] [PubMed]
9.
Choi, S.; Lee, S.; Seo, J.-W.; Kim, M.-J.; Jeon, Y.H.; Park, J.H.; Lee, J.K.; Yeo, N.S. Myocarditis-induced Sudden Death after
BNT162b2 mRNA COVID-19 Vaccination in Korea: Case Report Focusing on Histopathological Findings. J. Korean Med. Sci.
2021
,
36, e286. [CrossRef]
10.
Fillon, A.; Sautenet, B.; Barbet, C.; Moret, L.; Thillard, E.M.; Jonville-Béra, A.P.; Halimi, J.M. De novo and relapsing necrotizing
vasculitis after COVID-19 vaccination. Clin. Kidney J. 2022,15, 560–563. [CrossRef]
11.
Gao, J.-J.; Tseng, H.-P.; Lin, C.-L.; Hsu, R.-F.; Lee, M.-H.; Liu, C.-H. Acute encephalitis after COVID-19 vaccination: A case report
and literature review. Hum. Vaccines Immunother. 2022, 2082206. [CrossRef]
12.
Jarius, S.; Bieber, N.; Haas, J.; Wildemann, B. MOG encephalomyelitis after vaccination against severe acute respiratory syndrome
coronavirus type 2 (SARS-CoV-2): Case report and comprehensive review of the literature. J. Neurol.
2022
,269, 5198–5212.
[CrossRef] [PubMed]
Vaccines 2022,10, 1651 16 of 17
13.
Oshida, S.; Akamatsu, Y.; Matsumoto, Y.; Suzuki, T.; Sasaki, T.; Kondo, Y.; Fujiwara, S.; Kashimura, H.; Kubo, Y.; Ogasawara, K.
Intracranial aneurysm rupture within three days after receiving mRNA anti-COVID-19 vaccination: Three case reports. Surg.
Neurol. Int. 2022,13, 117. [CrossRef] [PubMed]
14.
Won, J.H.; Byun, S.J.; Oh, B.-M.; Park, S.J.; Seo, H.G. Risk and mortality of aspiration pneumonia in Parkinson’s disease:
A nationwide database study. Sci. Rep. 2021,11, 6597. [CrossRef] [PubMed]
15.
Matsumoto, H.; Sengoku, R.; Saito, Y.; Kakuta, Y.; Murayama, S.; Imafuku, I. Sudden death in Parkinson’s disease: A retrospective
autopsy study. J. Neurol. Sci. 2014,343, 149–152. [CrossRef] [PubMed]
16.
Fan, H.-T.; Lin, Y.-Y.; Chiang, W.-F.; Lin, C.-Y.; Chen, M.-H.; Wu, K.-A.; Chan, J.-S.; Kao, Y.-H.; Shyu, H.-Y.; Hsiao, P.-J. COVID-19
vaccine-induced encephalitis and status epilepticus. QJM Mon. J. Assoc. Physicians 2022,115, 91–93. [CrossRef] [PubMed]
17.
Liu, B.D.; Ugolini, C.; Jha, P. Two Cases of Post-Moderna COVID-19 Vaccine Encephalopathy Associated with Nonconvulsive
Status Epilepticus. Cureus 2021,13, e16172. [CrossRef]
18.
Werner, J.; Brandi, G.; Jelcic, I.; Galovic, M. New-onset refractory status epilepticus due to autoimmune encephalitis after
vaccination against SARS-CoV-2: First case report. Front. Neurol. 2022,13, 946644. [CrossRef]
19. Kim, D.; Bresette, C.; Liu, Z.; Ku, D.N. Occlusive thrombosis in arteries. APL Bioeng. 2019,3, 041502. [CrossRef]
20.
Ludewig, B.; Zinkernagel, R.M.; Hengartner, H. Arterial inflammation and atherosclerosis. Trends Cardiovasc. Med.
2002
,
12, 154–159. [CrossRef]
21.
Pons, S.; Fodil, S.; Azoulay, E.; Zafrani, L. The vascular endothelium: The cornerstone of organ dysfunction in severe SARS-CoV-2
infection. Crit. Care Lond. Engl. 2020,24, 353. [CrossRef]
22.
Dörschug, A.; Frickmann, H.; Schwanbeck, J.; Yilmaz, E.; Mese, K.; Hahn, A.; Groß, U.; Zautner, A.E. Comparative Assessment of
Sera from Individuals after S-Gene RNA-Based SARS-CoV-2 Vaccination with Spike-Protein-Based and Nucleocapsid-Based
Serological Assays. Diagnostics 2021,11, 426. [CrossRef]
23.
Magen, E.; Mukherjee, S.; Bhattacharya, M.; Detroja, R.; Merzon, E.; Blum, I.; Livoff, A.; Shlapobersky, M.; Baum, G.; Talisman,
R.; et al. Clinical and Molecular Characterization of a Rare Case of BNT162b2 mRNA COVID-19 Vaccine-Associated Myositis.
Vaccines 2022,10, 1135. [CrossRef] [PubMed]
24.
Röltgen, K.; Nielsen, S.C.A.; Silva, O.; Younes, S.F.; Zaslavsky, M.; Costales, C.; Yang, F.; Wirz, O.F.; Solis, D.; Hoh,
R.A.; et al. Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection
and vaccination. Cell 2022,185, 1025–1040.e14. [CrossRef] [PubMed]
25.
Lei, Y.; Zhang, J.; Schiavon, C.R.; He, M.; Chen, L.; Shen, H.; Zhang, Y.; Yin, Q.; Cho, Y.; Andrade, L.; et al. SARS-CoV-2 Spike
Protein Impairs Endothelial Function via Downregulation of ACE2. bioRxiv 2020. bioRxiv:2020.12.04.409144. [CrossRef]
26. Avolio, E.; Carrabba, M.; Milligan, R.; Kavanagh Williamson, M.; Beltrami, A.P.; Gupta, K.; Elvers, K.T.; Gamez, M.; Foster, R.R.;
Gillespie, K.; et al. The SARS-CoV-2 Spike protein disrupts human cardiac pericytes function through CD147 receptor-mediated
signalling: A potential non-infective mechanism of COVID-19 microvascular disease. Clin. Sci.
2021
,135, 2667–2689. [CrossRef]
27.
Bansal, S.; Perincheri, S.; Fleming, T.; Poulson, C.; Tiffany, B.; Bremner, R.M.; Mohanakumar, T. Cutting Edge: Circulating
Exosomes with COVID Spike Protein Are Induced by BNT162b2 (Pfizer-BioNTech) Vaccination prior to Development of
Antibodies: A Novel Mechanism for Immune Activation by mRNA Vaccines. J. Immunol. Baltim. Md. 1950
2021
,207, 2405–2410.
[CrossRef]
28.
Baumeier, C.; Aleshcheva, G.; Harms, D.; Gross, U.; Hamm, C.; Assmus, B.; Westenfeld, R.; Kelm, M.; Rammos, S.; Wenzel,
P.; et al. Intramyocardial Inflammation after COVID-19 Vaccination: An Endomyocardial Biopsy-Proven Case Series. Int. J. Mol.
Sci. 2022,23, 6940. [CrossRef]
29.
Yamamoto, M.; Kase, M.; Sano, H.; Kamijima, R.; Sano, S. Persistent varicella zoster virus infection following mRNA COVID-19
vaccination was associated with the presence of encoded spike protein in the lesion. J. Cutan. Immunol. Allergy 2022. [CrossRef]
30.
TGA Therapeutic Goods Administration. Australian Government 2021: Non Clinical Evaluation Report of BNT162b2 [mRNA]
COVID-19 vaccine (COMIRNATY
) Submission No: PM-2020-05461-1-2 Sponsor: Pfizer Australia Pty Ltd. Available online:
https://www.tga.gov.au/sites/default/files/foi-2389-06.pdf (accessed on 2 June 2022).
31.
Ancau, M.; Liesche-Starnecker, F.; Niederschweiberer, J.; Krieg, S.M.; Zimmer, C.; Lingg, C.; Kumpfmüller, D.; Ikenberg, B.; Ploner,
M.; Hemmer, B.; et al. Case Series: Acute Hemorrhagic Encephalomyelitis After SARS-CoV-2 Vaccination. Front. Neurol.
2021
,
12, 820049. [CrossRef]
32.
Poli, K.; Poli, S.; Ziemann, U. Multiple Autoimmune Syndromes Including Acute Disseminated Encephalomyelitis, Myasthenia
Gravis, and Thyroiditis Following Messenger Ribonucleic Acid-Based COVID-19 Vaccination: A Case Report. Front. Neurol.
2022
,
13, 913515. [CrossRef]
33.
Scorza, F.A.; Fiorini, A.C.; Scorza, C.A.; Finsterer, J. Cardiac abnormalities in Parkinson’s disease and Parkinsonism. J. Clin.
Neurosci. 2018,53, 1–5. [CrossRef]
34.
Piqueras-Flores, J.; López-García, A.; Moreno-Reig, Á.; González-Martínez, A.; Hernández-González, A.; Vaamonde-Gamo,
J.; Jurado-Románet, A. Structural and functional alterations of the heart in Parkinson’s disease. Neurol. Res.
2018
,40, 53–61.
[CrossRef] [PubMed]
35.
Burstein, B.; Barbosa, R.S.; Samuel, M.; Kalfon, E.; Philippon, F.; Birnie, D.; Mangat, I.; Redfearn, D.; Sandhu, R.; Macle, L.; et al.
Prevention of venous thrombosis after electrophysiology procedures: A survey of national practice. J. Interv. Card. Electrophysiol.
2018,53, 357–363. [CrossRef] [PubMed]
Vaccines 2022,10, 1651 17 of 17
36.
Kang, H.K.; Rhodes, C.; Rivers, E.; Thornton, C.P.; Rodney, T. Prevalence of Mental Health Disorders Among Undergraduate
University Students in the United States: A Review. J. Psychosoc. Nurs. Ment. Health Serv. 2021,59, 17–24. [CrossRef] [PubMed]
37.
Sung, J.G.; Sobieszczyk, P.S.; Bhatt, D.L. Acute Myocardial Infarction Within 24 Hours After COVID-19 Vaccination. Am. J. Cardiol.
2021,156, 129–131. [CrossRef] [PubMed]
38.
Cao, J.; Li, S.; Ma, Y.; Li, Z.; Liu, G.; Liu, Y.; Jiao, J.; Zhu, C.; Song, B.; Jin, J.; et al. Risk factors associated with deep venous
thrombosis in patients with different bed-rest durations: A multi-institutional case-control study. Int. J. Nurs. Stud.
2021
,
114, 103825. [CrossRef]
39.
Walter, A.; Kraemer, M. A neurologist’s rhombencephalitis after comirnaty vaccination. A change of perspective. Neurol. Res.
Pract. 2021,3, 56. [CrossRef] [PubMed]
... However, this argument was not based on data. Contrary to initial expectations, it was found that genes and proteins from genetic vaccines persist in the blood of vaccine recipients for prolonged periods of time [22,28,[40][41][42][43][44], and a variety of adverse events resulting from genetic vaccines are now being reported worldwide. Roubinian et al. reported that transfusions of plasma and platelet blood components collected before and after COVID-19 vaccination were not associated with increased adverse outcomes in transfusion recipients who did not develop COVID-19 [39]. ...
... However, compared to conventional inactivated vaccines, genetic vaccines, which produce an antigen within the body, are expected to prolong the period of exposure to the same antigen, and as a result, the risk of immune imprinting may be higher than with conventional vaccines. It is not actually known how long the vaccine components remain in the body after a person has received a genetic vaccine [22,40,43], but it is expected that they will remain in the body for a longer period than originally thought, in part because spike protein has been detected in the bodies of people several months after vaccination ( Table 1, point 1) [22,28,41,42]. In addition, since long-term exposure to a specific identical antigen (in this case, spike protein) causes immunoglobulins to become IgG4 [68,70] and some of the 4 B cells that produce them are likely to differentiate into memory B cells that survive in the body for a sustained period [70,89], the immune dysfunction of genetic vaccine recipients is expected to be prolonged (Table 1, point 3 & 6). ...
... Potential candidates include nucleocapsid. [4,5,41,128] 9 ...
Preprint
Full-text available
The coronavirus pandemic was declared by the World Health Organization (WHO) in 2020, and a global genetic vaccination program has been rapidly implemented as a fundamental solution. However, many countries around the world have reported that so-called genetic vaccines, such as those using modified mRNA encoding the spike protein and lipid nanoparticles as the drug delivery system, have resulted in post-vaccination thrombosis and subsequent cardiovascular damage, as well as a wide variety of diseases involving all organs and systems, including the nervous system. In this article, based on these circumstances and the volume of evidence that has recently come to light, we call the attention of medical professionals to the various risks associated with blood transfusions using blood products derived from people who have suffered from long COVID and from genetic vaccine recipients, including those who have received mRNA vaccines, and we make proposals regarding specific tests, testing methods, and regulations to deal with these risks. We expect that this proposal will serve as a basis for discussion on how to address post-vaccination syndrome and its consequences following these genetic vaccination programs.
... Thus, the Pfizer vaccine reduces the risk of infection (not death or serious illness) by 0.785%. But what Pfizer did: they compared 9 infected people in the vaccinated group with 169 in the placebo group and calculated a 95% efficacy [1,[10][11][12][13]. Relatively seen this is correct, but absolutely calculated on 40 000 test persons it is not [7,8,10,12]. ...
... The mRNA vaccines against the spike protein of COVID-19 were not designed, constructed and brought to market in a very short time [6,11,14]. The pharmaceutical companies knew and know that the search for a vaccine against coronaviruses has been unsuccess-ful for decades [14][15][16]. ...
... Was the COVID-19 pandemic deliberately hyped up in order to finally bring the expensive mRNA vaccines and with them the mRNA technology, even if deliberately flawed, onto the market? Billions had been invested in this research for years -now came the opportunity to collect this money worldwide [4,6,11,17]. The COVID-19 pandemic, a welcome "cash cow" for vaccine manufacturers? ...
... Various reports of VZV infection [12][13][14] or VZV encephalitis [15][16][17] have been reported after COVID-19 infection [2]. As such, cases of HSV encephalitis [18] and VZV meningitis [19] have also been reported after COVID-19 vaccination. ...
Article
Full-text available
The varicella zoster virus (VZV) is a latent viral infection and its reactivation has been reported following different conditions such as immunosuppression. This study presents a confirmed case of VZV encephalitis following the first dose administration of the Sinopharm COVID-19 vaccine. A 63-year-old immunocompetent woman who developed VZV encephalitis after first dose administration of Sinopharm COVID-19 vaccine. A final diagnosis of VZV encephalitis was made based on positive CSF PCR results for VZV infection. Treatment was administered with acyclovir and she returned to normal life without any neurological sequelae. In this report, VZV reactivation and VZV encephalitis have been observed after COVID-19 vaccination; however, the results of this report should be considered with some caution, and continued post-vaccine surveillance of adverse events is recommended to explore whether any causal association with VZV reactivation is biologically plausible in this context, or if it is just a coincidence.
... Altogether 18 articles meeting the search criteria were included (Table 1) [4,5,[8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23]. These articles reported altogether 21 patients with SC2VIE. ...
Article
Background: The rapid development of COVID-19 vaccines, combined with a high number of adverse event reports, have led to concerns over possible mechanisms of injury including systemic lipid nanoparticle (LNP) and mRNA distribution, Spike protein-associated tissue damage, thrombogenicity, immune system dysfunction, and carcinogenicity. The aim of this systematic review is to investigate possible causal links between COVID-19 vaccine administration and death using autopsies and post-mortem analysis. Methods: We searched PubMed and ScienceDirect for all published autopsy and necropsy reports relating to COVID-19 vaccination up until May 18th, 2023. All autopsy and necropsy studies that included COVID-19 vaccination as an antecedent exposure were included. Because the state of knowledge has advanced since the time of the original publications, three physicians independently reviewed each case and adjudicated whether or not COVID-19 vaccination was the direct cause or contributed significantly to death. Results: We initially identified 678 studies and, after screening for our inclusion criteria, included 44 papers that contained 325 autopsy cases and one necropsy case. The mean age of death was 70.4 years. The most implicated organ system among cases was the cardiovascular (49%), followed by hematological (17%), respiratory (11%), and multiple organ systems (7%). Three or more organ systems were affected in 21 cases. The mean time from vaccination to death was 14.3 days. Most deaths occurred within a week from last vaccine administration. A total of 240 deaths (73.9%) were independently adjudicated as directly due to or significantly contributed to by COVID-19 vaccination, of which the primary causes of death include sudden cardiac death (35%), pulmonary embolism (12.5%), myocardial infarction (12%), VITT (7.9%), myocarditis (7.1%), multisystem inflammatory syndrome (4.6%), and cerebral hemorrhage (3.8%). Conclusions: The consistency seen among cases in this review with known COVID-19 vaccine mechanisms of injury and death, coupled with autopsy confirmation by physician adjudication, suggests there is a high likelihood of a causal link between COVID-19 vaccines and death. Further urgent investigation is required for the purpose of clarifying our findings.
Article
Full-text available
According to the CDC, both Pfizer and Moderna COVID‐19 vaccines contain nucleoside‐modified messenger RNA (mRNA) encoding the viral spike glycoprotein of severe acute respiratory syndrome caused by corona virus (SARS‐CoV‐2), administered via intramuscular injections. Despite their worldwide use, very little is known about how nucleoside modifications in mRNA sequences affect their breakdown, transcription and protein synthesis. It was hoped that resident and circulating immune cells attracted to the injection site make copies of the spike protein while the injected mRNA degrades within a few days. It was also originally estimated that recombinant spike proteins generated by mRNA vaccines would persist in the body for a few weeks. In reality, clinical studies now report that modified SARS‐CoV‐2 mRNA routinely persist up to a month from injection and can be detected in cardiac and skeletal muscle at sites of inflammation and fibrosis, while the recombinant spike protein may persist a little over half a year in blood. Vaccination with 1‐methylΨ (pseudouridine enriched) mRNA can elicit cellular immunity to peptide antigens produced by +1 ribosomal frameshifting in major histocompatibility complex‐diverse people. The translation of 1‐methylΨ mRNA using liquid chromatography tandem mass spectrometry identified nine peptides derived from the mRNA +1 frame. These products impact on off‐target host T cell immunity that include increased production of new B cell antigens with far reaching clinical consequences. As an example, a highly significant increase in heart muscle 18‐flourodeoxyglucose uptake was detected in vaccinated patients up to half a year (180 days). This review article focuses on medical biochemistry, proteomics and deutenomics principles that explain the persisting spike phenomenon in circulation with organ‐related functional damage even in asymptomatic individuals. Proline and hydroxyproline residues emerge as prominent deuterium (heavy hydrogen) binding sites in structural proteins with robust isotopic stability that resists not only enzymatic breakdown, but virtually all (non)‐enzymatic cleavage mechanisms known in chemistry.
Article
Full-text available
Introduction Excess mortality during the COVID-19 pandemic has been substantial. Insight into excess death rates in years following WHO’s pandemic declaration is crucial for government leaders and policymakers to evaluate their health crisis policies. This study explores excess mortality in the Western World from 2020 until 2022. Methods All-cause mortality reports were abstracted for countries using the ‘Our World in Data’ database. Excess mortality is assessed as a deviation between the reported number of deaths in a country during a certain week or month in 2020 until 2022 and the expected number of deaths in a country for that period under normal conditions. For the baseline of expected deaths, Karlinsky and Kobak’s estimate model was used. This model uses historical death data in a country from 2015 until 2019 and accounts for seasonal variation and year-to-year trends in mortality. Results The total number of excess deaths in 47 countries of the Western World was 3 098 456 from 1 January 2020 until 31 December 2022. Excess mortality was documented in 41 countries (87%) in 2020, 42 countries (89%) in 2021 and 43 countries (91%) in 2022. In 2020, the year of the COVID-19 pandemic onset and implementation of containment measures, records present 1 033 122 excess deaths (P-score 11.4%). In 2021, the year in which both containment measures and COVID-19 vaccines were used to address virus spread and infection, the highest number of excess deaths was reported: 1 256 942 excess deaths (P-score 13.8%). In 2022, when most containment measures were lifted and COVID-19 vaccines were continued, preliminary data present 808 392 excess deaths (P-score 8.8%). Conclusions Excess mortality has remained high in the Western World for three consecutive years, despite the implementation of containment measures and COVID-19 vaccines. This raises serious concerns. Government leaders and policymakers need to thoroughly investigate underlying causes of persistent excess mortality.
Article
Full-text available
Lutein, a plant-derived xanthophyl-carotenoid, is an exceptional antioxidant and anti-inflammatory constituent found in food. High dietary intake of lutein is beneficial against eye disease, improves cardiometabolic health, protects from neurodegenerative diseases, and is beneficial for liver, kidney, and respiratory health. Lutein protects against oxidative and nitrosative stress, both of which play a major role in post-COVID and mRNA vaccination injury syndromes. Lutein is an important natural agent for therapeutic use against oxidative and nitrosative stress in chronic illnesses such as cardiovascular and neurodegenerative diseases and cancer. It can also potentially inhibit spike protein-induced inflammation. Rich dietary supplementation of lutein, naturally derived in non-biodegradable Extra Virgin Olive Oil (EVOO), can most optimally be used against oxidative and nitrosative stress during post-COVID and mRNA vaccination injury syndromes. Due to its high oleic acid (OA) content, EVOO supports optimal absorption of dietary lutein. The main molecular pathways by which the SARS-CoV-2 spike protein induces pathology, nuclear factor kappa-light-chain-enhancer activated B cells (NF-κB) and activated protein (AP)-1, can be suppressed by lutein. Synergy with other natural compounds for spike protein detoxification is likely.
Preprint
Full-text available
Exposure to vaccine lipid nanoparticles, mRNA, adenoviral DNA, and or Spike protein from one of the approved Covid-19 vaccines, or through secondary exposure, as through blood transfusion, is a potential source of harm. Blood reactions are an acknowledged side-effect of Covid-19 vaccination, not limited to hemolysis, paroxysmal nocturnal hemoglobinuria, chronic cold agglutinin disease, immune thrombocytopenia, haemophagocytosis, hemophagocytic lymphohistiocytosis, and many other blood related conditions. The observation of adverse events has motivated investigation into the cardiovascular mechanisms of harm by Covid-19 vaccines, and the biodistribution of vaccine contents. Biodistribution may not be limited to the body of the vaccine recipient, as a growing body of evidence demonstrates the possibility of secondary exposure to vaccine particles. These can be via bodily fluids and include the following routes of exposure: blood transfusion, organ transplantation, breastfeeding, and possibly other means. As covid-19 vaccines are associated with an increased risk of stroke, the persistence of vaccine artifacts in the blood presents a possible threat to a recipient of a blood donation from a vaccinated donor who suffered from vaccine induced thrombosis or thrombocytopenia. (VITT) We assess the feasibility and significance of these risks through an overview of the case report literature of blood disorders in vaccinated individuals, pharmacovigilance reports from the US Vaccine Adverse Events Reporting System (VAERS) and a meta-analysis of the available literature on organ transplants from vaccinated organ donors. Our analysis establishes biological mechanistic plausibility, a coherent safety signal in pharmacovigilance databases for secondary vaccine contents exposure (for the cases of blood transfusion and breastfeeding) and also an elevated level of adverse events in organ transplants from VITT-deceased donors, echoing increases in organ transplantation related complications seen in national statistics for some countries. Secondary exposure to vaccine artifacts is a potential explanation for some of the cases put forth, and requires a deeper investigation.
Article
Full-text available
Background Since the campaign of vaccination against COVID‐19 was started, a wide variety of cutaneous adverse effects after vaccination has been documented worldwide. Varicella zoster virus (VZV) reactivation was reportedly the most frequent cutaneous reaction in men after administration of mRNA COVID‐19 vaccines, especially BNT162b2. Aims A patient, who had persistent skin lesions after BNT162b2 vaccination for such a long duration over 3 months, was investigated for VZV virus and any involvement of vaccine‐derived spike protein. Materials & Methods Immunohistochemistry for detection of VZV virus and the spike protein encoded by mRNA COVID‐19 vaccine. PCR analysis for VZV virus. Results The diagnosis of VZV infection was made for these lesions using PCR analyses and immunohistochemistry. Strikingly, the vaccine‐encoded spike protein of the COVID‐19 virus was expressed in the vesicular keratinocytes and endothelial cells in the dermis. Discussion mRNA COVID‐19 vaccination might induce persistent VZV reactivation through perturbing the immune system, although it remained elusive whether the expressed spike protein played a pathogenic role. Conclusion We presented a case of persistent VZV infection following mRNA COVID‐19 vaccination and the presence of spike protein in the affected skin. Further vigilance of the vaccine side effect and investigation for the role of SP is warranted.
Article
Full-text available
Background Vaccination against SARS-CoV-2 has been conducted frequently to limit the pandemic but may rarely be associated with postvaccinal autoimmune reactions or disorders. Case presentation We present a 35-year-old woman who developed fever, skin rash, and headache 2 days after the second SARS-CoV-2 vaccination with BNT162b2 (Pfizer/Biontech). Eight days later, she developed behavioral changes and severe recurrent seizures that led to sedation and intubation. Cerebral magnetic resonance imaging showed swelling in the (para-) hippocampal region predominantly on the left hemisphere and bilateral subcortical subinsular FLAIR hyperintensities. Cerebrospinal fluid analysis revealed a lymphocytic pleocytosis of 7 cells/μl and normal protein and immunoglobulin parameters. Common causes of encephalitis or encephalopathy such as viral infections, autoimmune encephalitis with well-characterized autoantibodies, paraneoplastic diseases, and intoxications were ruled out. We made a diagnosis of new-onset refractory status epilepticus (NORSE) due to seronegative autoimmune encephalitis. The neurological deficits improved after combined antiepileptic therapy and immunomodulatory treatment including high-dose methylprednisolone and plasma exchange. Conclusions Although a causal relationship cannot be established, the onset of symptoms shortly after receiving the SARS-CoV-2 vaccine suggests a potential association between the vaccination and NORSE due to antibody-negative autoimmune encephalitis. After ruling out other etiologies, early immunomodulatory treatment may be considered in such cases.
Article
Full-text available
Initial clinical trials and surveillance data have shown that the most commonly administered BNT162b2 COVID-19 mRNA vaccine is effective and safe. However, several cases of mRNA vaccine-induced mild to moderate adverse events were recently reported. Here, we report a rare case of myositis after injection of the first dose of BNT162b2 COVID-19 mRNA vaccine into the left deltoid muscle of a 34-year-old, previously healthy woman who presented progressive proximal muscle weakness, progressive dysphagia, and dyspnea with respiratory failure. One month after vaccination, BNT162b2 vaccine mRNA expression was detected in a tissue biopsy of the right deltoid and quadriceps muscles. We propose this case as a rare example of COVID-19 mRNA vaccine-induced myositis. This study comprehensively characterizes the clinical and molecular features of BNT162b2 mRNA vaccine-associated myositis in which the patient was severely affected.
Article
Full-text available
Myocarditis in response to COVID-19 vaccination has been reported since early 2021. In particular, young male individuals have been identified to exhibit an increased risk of myocardial inflammation following the administration of mRNA-based vaccines. Even though the first epidemiological analyses and numerous case reports investigated potential relationships, endomyocardial biopsy (EMB)-proven cases are limited. Here, we present a comprehensive histopathological analysis of EMBs from 15 patients with reduced ejection fraction (LVEF = 30 (14-39)%) and the clinical suspicion of myocarditis following vaccination with Comirnaty® (Pfizer-BioNTech) (n = 11), Vaxzevria® (AstraZenica) (n = 2) and Janssen® (Johnson & Johnson) (n = 2). Immunohistochemical EMB analyses reveal myocardial inflammation in 14 of 15 patients, with the histopathological diagnosis of active myocarditis according the Dallas criteria (n = 2), severe giant cell myocarditis (n = 2) and inflammatory cardiomyopathy (n = 10). Importantly, infectious causes have been excluded in all patients. The SARS-CoV-2 spike protein has been detected sparsely on cardiomyocytes of nine patients, and differential analysis of inflammatory markers such as CD4+ and CD8+ T cells suggests that the inflammatory response triggered by the vaccine may be of autoimmunological origin. Although a definitive causal relationship between COVID-19 vaccination and the occurrence of myocardial inflammation cannot be demonstrated in this study, data suggest a temporal connection. The expression of SARS-CoV-2 spike protein within the heart and the dominance of CD4+ lymphocytic infiltrates indicate an autoimmunological response to the vaccination.
Article
Full-text available
Background In around 20% of cases, myelin oligodendrocyte glycoprotein (MOG) immunoglobulin (IgG)-associated encephalomyelitis (MOG-EM; also termed MOG antibody-associated disease, MOGAD) first occurs in a postinfectious or postvaccinal setting. Objective To report a case of MOG-EM with onset after vaccination with the Pfizer BioNTech COVID-19 mRNA vaccine BNT162b2 (Comirnaty®) and to provide a comprehensive review of the epidemiological, clinical, radiological, electrophysiological and laboratory features as well as treatment outcomes of all published patients with SARS-CoV-2 vaccination-associated new-onset MOG-EM. Methods Case report and review of the literature. Results In our patient, MOG-IgG-positive (serum 1:1000, mainly IgG1 and IgG2; CSF 1:2; MOG-specific antibody index < 4) unilateral optic neuritis (ON) occurred 10 days after booster vaccination with BNT162b2, which had been preceded by two immunizations with the vector-based Oxford AstraZeneca vaccine ChAdOx1-S/ChAdOx1-nCoV-19 (AZD1222). High-dose steroid treatment with oral tapering resulted in complete recovery. Overall, 20 cases of SARS-CoV2 vaccination-associated MOG-EM were analysed (median age at onset 43.5 years, range 28–68; female to male ratio = 1:1.2). All cases occurred in adults and almost all after immunization with ChAdOx1-S/ChAdOx1 nCoV-19 (median interval 13 days, range 7–32), mostly after the first dose. In 70% of patients, more than one CNS region (spinal cord, brainstem, supratentorial brain, optic nerve) was affected at onset, in contrast to a much lower rate in conventional MOG-EM in adults, in which isolated ON is predominant at onset and ADEM-like phenotypes are rare. The cerebrospinal fluid white cell count (WCC) exceeded 100 cells/μl in 5/14 (36%) patients with available data (median peak WCC 58 cells/μl in those with pleocytosis; range 6–720). Severe disease with tetraparesis, paraplegia, functional blindness, brainstem involvement and/or bladder/bowel dysfunction and a high lesion load was common, and treatment escalation with plasma exchange (N = 9) and/or prolonged IVMP therapy was required in 50% of cases. Complete or partial recovery was achieved in the majority of patients, but residual symptoms were significant in some. MOG-IgG remained detectable in 7/7 cases after 3 or 6 months. Conclusions MOG-EM with postvaccinal onset was mostly observed after vaccination with ChAdOx1-S/ChAdOx1 nCoV-19. Attack severity was often high at onset. Escalation of immunotherapy was frequently required. MOG-IgG persisted in the long term.
Article
Full-text available
Vaccine-related immune responses are one of the causes of encephalitis. Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) have been administered worldwide due to the ongoing global pandemic; cases of SARS-CoV-2 vaccination-related encephalitis were scarcely reported. An 82-year-old female was diagnosed with acute encephalitis following her first dose of vaccination with mRNA-1273 against SARS-CoV-2. The patient presented with fever and headache five days after vaccination, followed by behavior change 17 days after vaccination. Electroencephalographic recordings revealed focal slow waves in the right frontoparietal regions. Brain MRI revealed the signal change in the right middle and posterior temporal lobe. Cerebrospinal fluid analysis showed mildly elevated protein. She responded well to steroid pulse therapy and made a full recovery. The severity of the immune response following COVID-19 vaccination may be alleviated if adequate treatment is achieved. Physicians must be alert for encephalitis after vaccination to help ensure a favorable outcome.
Article
Full-text available
The global pandemic has resulted from the emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causing coronavirus disease 2019 (COVID-19). To control the spread of the pandemic, SARS-CoV-2 vaccines have been developed. Messenger ribonucleic acid (mRNA)-based COVID-19 vaccines have been the most widely used. We present the case of a 65-year-old patient, who was diagnosed with acute disseminated encephalomyelitis, ocular myasthenia gravis, and autoimmune thyroiditis, following his third mRNA COVID-19 vaccination. On admission, the patient showed mild left-sided hemiparesis, contralateral dissociated sensory loss, dizziness, and right-sided deafness. Brain MRI revealed multiple acute inflammatory contrast-enhancing periventricular and brainstem lesions with involvement of vestibulo-cerebellar tract and cochlear nuclei. Despite steroid pulse and intravenous immunoglobulin therapy, clinical symptoms and MRI lesions worsened, and additional signs of ocular myasthenia gravis and elevated but asymptomatic thyroid antibodies developed. After repeated plasma exchange, all clinical symptoms resolved. This is, to the best of our knowledge, the first case report of multiple autoimmune syndromes triggered by COVID-19 vaccination. The rare occurrence of such treatable autoimmune complications should not question the importance of vaccination programs during the COVID-19 pandemic.
Article
Full-text available
Background Although neurological adverse events have been reported after receiving coronavirus disease 2019 (COVID-19) vaccines, associations between COVID-19 vaccination and aneurysmal subarachnoid hemorrhage (SAH) have rarely been discussed. We report here the incidence and details of three patients who presented with intracranial aneurysm rupture shortly after receiving messenger ribonucleic acid (mRNA) COVID-19 vaccines. Case Description We retrospectively reviewed the medical records of individuals who received a first and/ or second dose of mRNA COVID-19 vaccine between March 6, 2021, and June 14, 2021, in a rural district in Japan, and identified the occurrences of aneurysmal SAH within 3 days after mRNA vaccination. We assessed incidence rates (IRs) for aneurysmal SAH within 3 days after vaccination and spontaneous SAH for March 6–June 14, 2021, and for the March 6–June 14 intervals of a 5-year reference period of 2013–2017. We assessed the incidence rate ratio (IRR) of aneurysmal SAH within 3 days after vaccination and spontaneous SAH compared to the crude incidence in the reference period (2013–2017). Among 34,475 individuals vaccinated during the study period, three women presented with aneurysmal SAH (IR: 1058.7/100,000 person-years), compared with 83 SAHs during the reference period (IR: 20.7/100,000 persons-years). IRR was 0.026 (95% confidence interval [CI] 0.0087–0.12; P < 0.001). A total of 28 spontaneous SAHs were verified from the Iwate Stroke Registry database during the same period in 2021 (IR: 34.9/100,000 person-years), and comparison with the reference period showed an IRR of 0.78 (95%CI 0.53–1.18; P = 0.204). All three cases developed SAH within 3 days (range, 0–3 days) of the first or second dose of BNT162b2 mRNA COVID-19 vaccine by Pfizer/BioNTech. The median age at the time of SAH onset was 63.7 years (range, 44– 75 years). Observed locations of ruptured aneurysms in patients were the bifurcations of the middle cerebral artery, internal carotid-posterior communicating artery, and anterior communicating artery, respectively. Favorable outcomes (modified Rankin scale scores, 0–2) were obtained following microsurgical clipping or intra-aneurysm coiling. Conclusion Although the advantages of COVID-19 vaccination appear to outweigh the risks, pharmacovigilance must be maintained to monitor potentially fatal adverse events and identify possible associations.
Article
Full-text available
We present three cases fulfilling diagnostic criteria of hemorrhagic variants of acute disseminated encephalomyelitis (acute hemorrhagic encephalomyelitis, AHEM) occurring within 9 days after the first shot of ChAdOx1 nCoV-19. AHEM was diagnosed using magnetic resonance imaging, cerebrospinal fluid analysis and brain biopsy in one case. The close temporal association with the vaccination, the immune-related nature of the disease as well as the lack of other canonical precipitating factors suggested that AHEM was a vaccine-related adverse effect. We believe that AHEM might reflect a novel COVID-19 vaccine-related adverse event for which physicians should be vigilant and sensitized.
Article
Full-text available
During the SARS-CoV-2 pandemic, novel and traditional vaccine strategies have been deployed globally. We investigated whether antibodies stimulated by mRNA vaccination (BNT162b2), including 3rd dose boosting, differ from those generated by infection or adenoviral (ChAdOx1-S and Gam-COVID-Vac) or inactivated viral (BBIBP-CorV) vaccines. We analyzed human lymph nodes after infection or mRNA vaccination for correlates of serological differences. Antibody breadth against viral variants is less after infection compared to all vaccines evaluated, but improves over several months. Viral variant infection elicits variant-specific antibodies, but prior mRNA vaccination imprints serological responses toward Wuhan-Hu-1 rather than variant antigens. In contrast to disrupted germinal centers (GCs) in lymph nodes during infection, mRNA vaccination stimulates robust GCs containing vaccine mRNA and spike antigen up to 8 weeks post-vaccination in some cases. SARS-CoV-2 antibody specificity, breadth and maturation are affected by imprinting from exposure history, and distinct histological and antigenic contexts in infection compared to vaccination.