ArticlePDF AvailableLiterature Review

Immunotherapies and COVID-19 related Neurological manifestations: A Comprehensive Review Article

Authors:
  • Koç Üniversitesi

Abstract and Figures

In December 2019, an outbreak of pandemic severe respiratory distress syndrome coronavirus disease 2019 (COVID-19) initially occurred in China, has spread the world resulted in serious threats to human public health. Uncommon neurological manifestations with pathophysiological symptoms were observed in infected patients including headache, seizures, and neuroimmunological disorders. Regardless of whether these neurological symptoms are direct or indirect casual infection relationship, this novel viral infection has a relevant impact on the neuroimmune system that requires a neurologist’s careful assessment. Recently, the use of immunotherapy has been emerged in fighting against COVID-19 infection despite the uncertain efficiency in managing COVID-19 related disorders or even its proven failure by increasing its severity. Herein, the author is addressing the first approaches in using immunotherapies in controlling COVID-19 viral impact on the brain by highlighting their role in decreasing or increasing infection risks among subjects. This point of view review article supports the use of immunotherapies in managing COVID-19 neurological disorders but in optimal timing and duration to ensure the maximum therapeutic outcome by reducing morbidity and mortality rate. Based on recently published data, the current review article highlights the beneficial effects and drawbacks of using immunotherapies to combat COVID-19 and its neurological symptoms.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ljii20
Journal of Immunoassay and Immunochemistry
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ljii20
Immunotherapies and COVID-19 related
Neurological manifestations: A Comprehensive
Review Article
Mai M. Anwar
To cite this article: Mai M. Anwar (2021): Immunotherapies and COVID-19 related Neurological
manifestations: A Comprehensive Review Article, Journal of Immunoassay and Immunochemistry,
DOI: 10.1080/15321819.2020.1865400
To link to this article: https://doi.org/10.1080/15321819.2020.1865400
Published online: 03 Jan 2021.
Submit your article to this journal
View related articles
View Crossmark data
Immunotherapies and COVID-19 related Neurological
manifestations: A Comprehensive Review Article
Mai M. Anwar
Department of Biochemistry, National Organization for Drug Control and Research (Nodcar)/egyptian
Drug Authority, Cairo, Egypt
ABSTRACT
In December 2019, an outbreak of pandemic severe respiratory
distress syndrome coronavirus disease 2019 (COVID-19) initially
occurred in China, has spread the world resulted in serious threats
to human public health. Uncommon neurological manifestations
with pathophysiological symptoms were observed in infected
patients including headache, seizures, and neuroimmunological
disorders. Regardless of whether these neurological symptoms
are direct or indirect casual infection relationship, this novel viral
infection has a relevant impact on the neuroimmune system that
requires a neurologist’s careful assessment. Recently, the use of
immunotherapy has been emerged in ghting against COVID-19
infection despite the uncertain eciency in managing COVID-
19 related disorders or even its proven failure by increasing its
severity. Herein, the author is addressing the rst approaches in
using immunotherapies in controlling COVID-19 viral impact on the
brain by highlighting their role in decreasing or increasing infection
risks among subjects. This point of view review article supports the
use of immunotherapies in managing COVID-19 neurological dis-
orders but in optimal timing and duration to ensure the maximum
therapeutic outcome by reducing morbidity and mortality rate.
Based on recently published data, the current review article high-
lights the benecial eects and drawbacks of using immunothera-
pies to combat COVID-19 and its neurological symptoms.
KEYWORDS
COVID-19; cytokines storm;
inflammation;
immunotherapies;
neurological disorders;
vaccines
Introduction
The new pandemic threatening infection COVID-19 is a worldwide outbreak
previously declared as a public health emergency by the WHO and was officially
named as SARS-CoV-2. The uncontrolled COVID-19 outbreak throughout the
world resulted in several economic, social, and financial drawbacks in addition to
extreme lockdown measures.
[1,2]
Coronavirus is a single-stranded envelope crown-
like shape RNA virus that can spread via personal contact and respiratory
droplets.
[3]
The most common COVID-19 respiratory infection symptoms include
cough, fever, and shortness of breath. Additionally, COVID-19 may also develop
several related neurological disorders ranging from severe headache to neurode-
generative diseases.
[4–6]
It has been previously reported that COVID-19 associated
CONTACT Mai M. Anwar mainwr@hotmail.com Lecturer at the Department of Biochemistry, National
Organization for Drug Control and Research (Nodcar)/egyptian Drug Authority, Cairo, Egypt.
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY
https://doi.org/10.1080/15321819.2020.1865400
© 2020 Taylor & Francis
acute respiratory failure is directly related to brain stem damage caused by SARS-
CoV-2 brain infection.
[7,8]
Several questions regarding the hypothetical mechan-
ism and routes via which the COVID-19 can penetrate the brain have been recently
declared with the real need for more distinctive clarifications on the appropriate
prophylaxis, and treatment of COVID-19 complications.
[4,5]
Table 1.
Review literature methods
A relevant search was conducted using google scholar, PubMed, Scopus,
Science Direct, and Web of Science to find out published articles mainly
related to COVID-19 and neurological aspects. Type of articles used in the
current review included research articles, review articles, case series, case
reports, letter to editors, randomized controlled trials, and clinical trials
using relevant keywords in search strategy methods. Inclusion search criteria
were any published English language articles from reputable journals with
a publication date starting from January 2020 to December 2020 with exclu-
sion to any non-relevant topics and non-English language articles.
The neurotropic potentials of COVID-19 on the central nervous system (CNS)
SARS-CoV-2 is considered as the seventh type of coronavirus family with about
82% similarity to SARS coronavirus (SARS-CoV) and 60% to the Middle East
respiratory syndrome (MERS-CoV). Recently, SARS-CoV-2 viral nucleic acid was
observed in the cerebrospinal fluid of several infected patients along with its
presence among brain autopsy which can be directly justified to its neurotropic
penetration action using both hematogenous and/or non-hematogenous
routes.
[4,9–11]
Several epidemiological studies reported various COVID-19 neuro-
logical manifestations commonly found among infected persons such as headache,
loss of consciousness, and delirium.
[4,12]
The COVID-19 neuropenetration
hypothesis was initially suggested based on the temporary or permanent anosmia
symptom mostly found among infected patients along with the novel approach
toward highlighting that neurological symptoms might be the second drawback of
respiratory infection.
[4–6,11,13,14]
Multiples routes via which the COVID-19 can
penetrate the brain
[15]
[Figure 1] include (a) direct infection injury via nasal
olfactory bulb route;
[16]
(b) through ACE-2 functional receptors;
[10]
(c) neuronal
pathways and nerve ending dissemination;
[16,17]
(d) synapse conjugated route;
[17]
and (e) hypoxic metabolic injury.
[5]
COVID-19 and CNS immune-mediated system injuries
COVID-19 brain infection can results in severe CNS damages as
a drawback of disturbances in brain homeostasis with direct polarization
toward microglia pro-inflammatory M1 type resulting in the release of
2M. M. ANWAR
Table 1. Brief of immunomodulatory agents and recommended doses for the management of COVID-19 patients with neurological disorders.
Treatment Mode of action Common adverse
events
Recommendations Recommended
doses
References
Corticosteroids -Act as anti-inflammatory
to prevent exaggerated
cytokine release.
-Water and sodium retention,
lymphopenia,
myasthenia gravis, and
infections.
-To be used with high caution in elderly and
immunocompromised COVID-19 patients using the
smallest recommended dose for the shortest duration of
time.
-General health advice and monitoring for patients on
immunotherapies.
- Dexamethasone IV:
6 mg daily dose for
10 days.
- Dexamethasone
orally:
20 mg for 5 days
followed by 10 mg for
other 5 days.
- Methylprednisolone
orally:
0.5–1 mg/kg daily or
60–125 mg (high dose)
every 6 hr for 3 days in
case of sever cytokines
storm.
25–28
Tocilizumab -Anti-cytokine, IL-6 antagonist. -Upper respiratory
tract infections,
hypertension,
and elevated liver enzymes.
-General health advice and monitoring for the probability
of secondary infection.
-Not recommended in pregnant and Lactating women.
-4–8 mg/kg
or
single maximum
dose 800 mg which
may be repeated after
12 hr.
30–32
Teriflunomide -Teriflunomide selectively
inhibits dihydroorotate
dehydrogenase (DHODH),
a key enzyme in the novo
pyrimidine synthesis
pathway.
-Lymphopenia with a mild
decreases in lymphocyte count
Probability of increasing the risk
of severe COVID-19 symptoms
due to known risk of
lymphopenia.
-In case of initiating teriflunomide in COVID-19 patients
with previous neurological disorders, subjects will
undego a required weekly blood monitoring tests to
screen for any uncomman drawbacks.
-General health advice for high probability of other risk
severe infections.
-14 mg daily
recommended dose in
COVID-19 patients with
neurological disorders.
35–37
IVIg -Antibodiesto provide short-
termpassive type of immunity
by Pleiotropic immune-
modulation actions.
-Anaphylactic shock
or severe systemic
hypersensitivity
action.
-Use with caution in elderly COVID-19 patients, with a high
attention to immunocompromised patients.
-0.5 g/kg daily for
5 consequent days IV.
40–43
Treatment Mode of action Common adverse
events
Recommendations Recommended
doses
References
(Continued)
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 3
Table 1. (Continued).
Cladribine -Type of synthetic purine
nucleoside analog by
inhibiting DNA synthesis via
depleting B and
T cells.
-Risk of increased COVID-19 viral
infection symptoms with
a decreased lymphocyte counts.
-General health advice and monitoring for COVID-19
patients with neurological disorders on treatment
initiation.
-3.5 mg/kg for 1 to
2 weeks.
44–48
Rituximab -Anti-cytokine and act as JAK1/
JAK2 inhibitor.
-Thrombocytopenia,
neutropenia,
severe infections,
headache, anemia, and
dizziness.
-Not recommended in pregnant and
lactating women.
-Dose monitoring and reduction in renal and hepatic
impairment.
-5 mg bid for 14 days/
5 mg bid for 3 days
followed by 10 mg bid
for other 7 days.
49–54
IFN-β -Antiviral and
immunomodulatory action.
-Severe peripheral edema,
skin rash, leukopenia,
neutropenia, ataxia,
hypertonia, and
neurological symptoms.
-To be used with caution in patients with bone
marrow suppression, cardiac patients
, and hepatic impairment.
-0.25 mg (8 million
IU) for 3 days.
55–58
Remdesivir -Antiviral nucleotide analog. -Nausea, acute respiratory failure,
hypokalemia, hypotension,
acute kidney injury, and
elevated liver enzymes.
-The neuropsychiatric adverse effect should be monitored
following treatment initiation for patients infected with
COVID-19.
-200 mg loading dose for
1 day, followed by
100 mg daily for other
9 days.
59–65
Natalizumab -Monoclonal antibodies (mAbs)
that act by blocking the
migration of lymphocytes
into the brain.
-Rarely brain and respiratory tract
infection.
-General health monitoring. -300 mg for2-4 weeks.
66–69
4M. M. ANWAR
associated inflammatory cascades and cytokines including Interleukin-6
(IL-6) defined as the syndrome of cytokine storm
[18,19]
[Figure 2]. The
consequences of COVID-19 penetration to the CNS directly activate
CD4+ immune cells, IL-6, and tumor necrosis factor-alpha (TNF-α) pro-
inflammatory factors resulting in Neurodegeneration and CNS comor-
bidities. Consequently, IL-6 is the main predominant component of
COVID-19 cytokine syndrome along with TNF-α, and interferon-
gamma (IFNγ) leading to several organ damages including the
brain.
[9,18,19]
It was also reported that the peripheral nervous system
(PNS) is mainly being vulnerable during mediated immune infectious
diseases such as COVID-19 resulting in prolonged immobilization.
[20]
Figure 1. Hypothetical illustration of COVID-19 possible routes of entry into the CNS resulting in
severe nerves damage along with post-infection neurological manifestation.
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 5
Recently, attention has been given to COVID-19 patients suffering from
neuroimmunological diseases including multiple sclerosis (MS) which
mainly results from severe dysfunction in the immune regulatory net-
works and necessitating a continual administration of immunomodula-
tory or immunosuppressive agents.
[5]
The action of Immunomodulatory
agents mainly depends on adjusting the immune system or amplifying
the host immune responses while the immunosuppressive agent sup-
presses the immune responses by limiting the immune competence
activity. The beneficial rational use of immunotherapies in the manage-
ment of COVID-19 complications such as neurological disorders and
acute respiratory distress syndrome (ARDS) requires causative attention
to avoid no deleterious effect and to ensure an efficient action.
[11,20,21]
Figure 2. Hypothetical illustration of cytokine storm syndrome following COVID-19 brain penetra-
tion resulting in neurodegeneration and several CNS comorbidities.
6M. M. ANWAR
The impact of immunotherapies in managing neurological manifestations
of COVID-19
Regardless of the exact mechanism of how COVID-19 penetrates the brain,
direct or indirect post-infection neurological manifestations require onset
management and assessment to avoid case progression and irreversible neu-
rological damage. Further attention and considerations must be given to
COVID-19 patients with previous neurological disorders who require the
administration of immunotherapies that could result in increasing the severity
of the viral infection.
[11,20,21]
However, some recent reports suggest their
efficiency in managing COVID-19 neurological symptoms.
[19–21]
Recently,
physicians are using various types of immunotherapies in managing COVID
infection due to their known pharmacokinetics and safety profile. Many
immune-modulating agents previously used to regulate different aspects of
inflammatory cascades are being used lately among physicians in the manage-
ment of COVID-19 severe immune imbalance.
[19,20]
Since hyperinflammation
is the determinant factor of COVID-19 infection, immunosuppressive agents
are also considered to be beneficial in reducing the mortality rate of hospita-
lized COVID-19 patients.
[18–20,22]
It is highly required to monitor and balance
the risk to benefit ratio before immunotherapy initiation and optimization for
COVID-19 patients with neurological manifestations.
[23]
Glucocorticoids and their synthetic analogs were widely used in managing
rheumatic disease-related autoimmune disorders by suppressing cytokine release
and T-cell activation. Due to their rapid immunosuppressive actions, glucocorti-
coids are mainly used in the management of severe hyperinflammatory syndromes
including acute respiratory distress syndrome (ARDS). Lately, corticosteroid ther-
apy has been effectively used in managing SARS and MERS acquired pneumonia
infection associated with some hazard drawbacks. Clear evidence regarding the use
of corticosteroids in managing COVID-19 is still unclear in addition to the huge
scientific debate on the announced recommendations for its vital usage in severe
hospitalized ARDS COVID-19 patients.
[24–26]
Although the use of corticosteroid
therapy in the case of COVID-19 related neurological disorders has not been well
addressed, relevant positive feedback was observed related to its efficiency in
controlling elevated CSF cytokines suggesting its efficiency in managing
COVID-19-related encephalitis disorders.
[26–28]
However, corticosteroids may
also increase the risk of infection due to short-term lymphopenia but this is mainly
associated with long-term doses of steroids, not with short-term use.
[25–28]
IL-6 is a key point inflammatory cytokine that acts as the main regulator for
the cytokine storm syndrome and was found to be elevated in COVID-19
patients. Tocilizumab is categorized as a recombinant monoclonal antibody
acting as an IL-6 blocking agent widely used in the treatment of autoimmune
diseases such as rheumatoid arthritis.
[29,30]
A significant relevant increase in
the production of IL-6-with simultaneous elevated release of CD14+ and
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 7
CD16+ inflammatory monocytes were observed among COVID-19-infected
subjects. The hyperactivated T helper cells (Th1) suggested being found
among COVID-19 patients potentially provoke the elevation of IFNγ in the
lung along with IL-6, suggesting that blocking the IL-6 signaling cascade might
be potential as therapeutic agents in COVID-19 patients.
[5,18,19]
Tocilizumab
act as the first-line treatment for managing cytokine release syndrome in
severely infected subjects. Repeated observational studies previously con-
ducted in China and Italy reported the efficacy of tocilizumab treatment in
hindering severe COVID-19 neurological disorders.
[31,32]
Based on these
approaches, blocking the IL-6 signaling pathway represents a major therapeu-
tic potential for the treatment of neuroinflammatory and neurodegenerative
diseases as a drawback of COVID-19 infection.
[33,34]
Recent reports demon-
strated that the administration of tocilizumab was associated with a high rate
of secondary infection. These findings raised a lot of concerns about the use of
anti-IL-6 tocilizumab therapy to attenuate cytokine storm syndrome in
COVID-19 patients.
[30]
Teriflunomide selectively inhibits the mitochondrial enzyme Dihydroorotate
dehydrogenase (DHODH) in the de novo pyrimidine pathway. It selectively
results in relevant immunosuppression by inhibiting the division and growth of
replicating cells, including activated B and T cells along with decreasing lym-
phocyte counts.
[35,36]
Huge scientific debates among neurologists on the ability
of teriflunomide to increase the severity of COVID-19 which is considered
heterogeneous, due to its lymphopenia associated risk occurring within the
first few weeks of treatment initiation. On the other hand, several other recom-
mendations suggest that teriflunomide may not increase the severity of COVID-
19 manifestation indicating its safety but these recommendations require more
potential monitoring and justifications especially in the first 2 weeks of treat-
ment initiation.
[35–37]
Intravenous immunoglobulins (IVIg) are an efficient treatment for several
patients with antibody deficiencies. IVIg therapy contains immunoglobulin
G obtained from pooled type healthy donor plasma that can act on both body
initiative and adaptive immune type.
[38,39]
Additionally, IVIg was previously
indicated in managing neuroimmunological diseases such as MS and myasthe-
nia gravis.
[38,39]
Although the use of IVIg on COVID-19 patients is somehow
limited, the risk of increasing disease-related severity including neurological
symptoms is considered mild. It was reported that IVIg is effective against
thromboembolism and elevated D-dimer levels in hospitalized COVID-19
patients. Moreover, IVIg may have promising effects on reducing the antiviral
neutralizing antibody level by activating both natural killer cells (NK) and
macrophages in addition to its TNF-α inhibiting actions.
[40–43]
Recently,
several COVID-19 cases have been reported to administrate IVIg therapy
with observed anaphylactic shock and hypersensitivity symptoms following
treatment initiations.
[41–43]
8M. M. ANWAR
Cladribine is characterized as a synthetic purine analog that directly disrupts the
DNA synthesis process and repair. Upon using cladribine for more than 4 months,
a relevant decrease in circulating CD4 + T, NK cells, B cells, and T cells resulting in
lymphopenia which makes COVID-19 patients more susceptible to infection
severity and secondary infection.
[44–47]
The observed relevant immunosuppression
actions following cladribine administration are mainly associated with higher
infectious risk.
[46,47]
Several studies also suggest that cladribine treatments can be
initiated in COVID-19 patients with demandable general health monitoring, but
the risk-to-benefit ratio should be taken into consideration especially for aged
immunocompromised patients.
[48]
Rituximab is one of the main B cells targeting monoclonal antibody agents
(mAB) frequently used in managing neuroinflammatory disorders by acting as
anti-CD20 mAB.
[49,50]
The expression of CD20 molecule is associated with B cell
maturation, rituximab works effectively by depleting pre-B and memory B cells.
[50]
The use of rituximab can be considered as one of the highest risk agents to be used
in hindering COVID-19 pandemic infection due to its toxic and dangerous side
effects with a recommendation on considering the delay of rituximab treatment
initiation in managing COVID-19 neurological symptoms.
[51–54]
Human interferon beta (IFN-β) was the first disease-modifying agent previously
used in MS by modulating the cytokines immune responsive action through
shifting cytokine toward the production of anti-inflammatory B cell types. No
observed drawbacks were associated with IFN-β administration except
lymphopenia.
[55,56]
Studies regarding the IFN-β antiviral COVID-19 replication
are suggested to be effective through activating Janus kinases (JAKs) pathways
leading to activated antiviral and antiproliferative actions.
[57,58]
It was severally
reported an increase in COVID-19 neurological manifestations as a drawback of
IFN-β administration in infected patients.
[55–57]
Remdesivir is a promising nucleoside antiviral analog drug currently used
in controlling COVID-19 delirium complications.
[59,60]
Although Remdesivir
has been widely used in COVID-19 treatment protocols, no significant clinical
or antiviral effective actions were observed.
[61,62]
Its safety is still under
evaluation as numerous studies reported neuropsychiatric adverse effects
and neurological complications associated with remdesivir administration in
COVID-19 patients.
[63–65]
Natalizumab is a humanized recombinant mAb that acts by blocking leukocyte
alpha 4-integrin molecules along with transferring peripheral immune cells into
the CNS by acting as prophylaxis against COVID-19 infection and its related
neurological disorders.
[66]
The administration of natalizumab results in a relevant
exaggerated increase in NK, B, and T cells in the peripheral blood tissues.
[66,67]
The
higher the elevated B cell-mediated inflammatory level, the better would be
favorable against viral infectious disease resulting in an observed decrease in
lymphocyte migration in COVID-19-infected cases indicating its efficacy as
a protective agent against COVID-19 viral infection.
[66–68]
Some reports suggested
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 9
the possible role of natalizumab administration in preventing COVID-19 from
binding to some of its receptors with a recommendation toward interval dosing to
avoid rebound.
[69]
Since the spread of this sudden pandemic viral infection and the world has been
calling scientists for the discovery of the COVID-19 vaccine. Although we are a few
steps away from the release of more the one type of vaccine, many questions have
been raised regarding whether it would be efficient for immunocompromised and
neurologically impaired COVID-19 patients. Unfortunately, till now no clear
results have been declared. Successful vaccination with minor side effects depends
mainly on immune reconstitution factors following cell depletion.
[70,71]
On the
other hand, a suggested increase in the immune responsive action against vaccine’s
different types including live attenuated, inactive viruses, and adjuvant-containing
type could result in triggering aggressive autoimmune responsive actions along
with increasing the incidence of relapse. Furthermore, restrictions on using live
attenuated vaccines along with immunotherapy agents must be taken into
consideration.
[70–72]
The impact of immunotherapies in the pandemic era of COVID-19
infection
Although relevant data are still lacking, the author suggests that the use of
immunotherapy agents might be associated with higher infection risk and severe
neurological manifestations as a result of long-lasting lymphopenia. On the other
hand, the justification for using immunotherapy agents in the era of pandemic
COVID-19 despite their various drawbacks and severe side-effects, all can all be
related to the unavailable alternatives and rarely conducted clinical studies.
[73,74]
Thus, immunotherapy treatment risks in severe COVID-19 patients with neuro-
logical symptoms might be relieved by attentive hospitalization and great precision
regarding the initiation and optimization of therapy protocol. Features of using
suitable immunotherapy mainly depend on patient health status and other related
conditions as misleading usage may lead to severe neurological disorders and
persistent irreversible disability.
[73,74]
Conclusion
In conclusion, the observed neurological manifestations of COVID-19 are severe,
but neither their clear reasons for incidence nor their causative symptoms have
been clearly demonstrated. However, COVID-19 directly affects the assessments
and monitoring of patients with neurological disorders in several ways, but
evidence concerning the use of immunotherapy in their management is still
unclear due to fluctuations in drawbacks among subjects participated in research
studies. Conduction of clinical studies is urgently required to attain more accurate
data on neuroimmunological disease and COVID-19 infection for better future
10 M. M. ANWAR
management approaches. These clinical studies should answer whether immu-
notherapy agents can control COVID-19 neurological manifestations and respira-
tory distress syndrome along with estimating the probability of resulting in
comorbidities. The careful risk-to-benefit balance along with initiation and dis-
continuation administration time has to be taken into consideration while practi-
cing immunotherapy among hospitalized infected subjects till obtaining unbiased
answers. Patients’ inclusion and exclusion criteria mainly depend on the response
of subject host phase biomarkers. These findings may help the upcoming future
research on the accurate diagnosis, treatment, and prophylaxis of COVID-19
severe symptoms.
Data accessibility
This article has no further additional data.
Competing interests
I declare no competing interest with others.
ORCID
Mai M. Anwar http://orcid.org/0000-0002-1991-2945
References
[1] Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.;
et al. Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan,
China. Lancet. 2020, 395(10223), 497–506.
[2] Wang, C.; Horby, P. W.; Hayden, F. G.; Gao, G. F. A Novel Coronavirus Outbreak of
Global Health Concern. Lancet. 2020, 395(10223), 470–473. DOI: 10.1016/S0140-
6736(20)30185-9.
[3] Chen, Y.; Chen, L.; Deng, Q.; Zhang, G.; Wu, K.; Ni, L.; Yang, Y.; Liu, B.; Wang, W.;
Wei, C.; et al. The Presence of SARS-CoV-2 RNA in the Feces of COVID-19 Patients. J
Med Virol. 2020, 92(7), 833–840. DOI: 10.1002/jmv.25825.
[4] Anwar, M. M.; Badawi, A. M.; Eltablawy, N. A. Can the Coronavirus Infection Penetrates
the Brain Resulting in Sudden Anosmia Followed by Severe Neurological Disorders?
eNeurologicalSci. 2020, 21, 100290. DOI: 10.1016/j.ensci.2020.100290.
[5] Mao, L.; Jin, H.; Wang, M.; Hu, Y.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.;
Wang, D.; et al. Neurologic Manifestations of Hospitalized Patients with Coronavirus
Disease 2019 in Wuhan, China. JAMA neurol. 2020, 77(6), 683–690.
[6] Li, Y. C.; Bai, W. Z.; Hashikawa, T. The Neuroinvasive Potential of SARS-CoV2 May
Play a Role in the Respiratory Failure of COVID-19 Patients. J. Med. Virol. 2020, 92(6),
552–555. DOI: 10.1002/jmv.25728.
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 11
[7] Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.;
Zhu, L.; et al. Pathological Findings of COVID-19 Associated with Acute Respiratory
Distress Syndrome. Lancet Respir Med. 2020, 8(4), 420–422.
[8] Lu, H.; Stratton, C. W.; Tang, Y. W. Outbreak of Pneumonia of Unknown Etiology in
Wuhan, China: The Mystery and the Miracle. J Med Virol. 2020, 92(4), 401–402. DOI:
10.1002/jmv.25678.
[9] Fu, Y.; Cheng, Y.; Wu, Y. Understanding SARS-CoV-2-Mediated Inflammatory
Responses: From Mechanisms to Potential Therapeutic Tools. Virologica Sin. 2020, 35
(3), 266–271. DOI: 10.1007/s12250-020-00207-4.
[10] Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural Basis for the Recognition
of SARS-CoV-2 by Full-length Human ACE2. Science (New York, NY). 2020, 367(6485),
1444–1448. DOI: 10.1126/science.abb2762.
[11] Zou, L.; Ruan, F.; Huang, M.; Liang, L.; Huang, H.; Hong, Z.; Yu, J.; Kang, M.; Song, Y.;
Xia, J.; et al. SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected
Patients. N. Engl. J. Med. 2020, 382(12), 1177–1179.
[12] Liguori, C.; Pierantozzi, M.; Spanetta, M.; Sarmati, L.; Cesta, N.; Iannetta, M.; Ora, J.;
Mina, G. G.; Puxeddu, E.; Balbi, O.; et al. Subjective Neurological Symptoms Frequently
Occur in Patients with SARS-CoV2 Infection. Brain Behav. Immun. 2020, 88, 11–16.
DOI: 10.1016/j.bbi.2020.05.037.
[13] Asadi-Pooya, A. A.; Simani, L. Central Nervous System Manifestations of COVID-19:
A Systematic Review. J. Neurol. Sci. 2020, 413, 116832. DOI: 10.1016/j.jns.2020.116832.
[14] Tsai, S. T.; Lu, M. K.; San, S.; Tsai, C. H. The Neurologic Manifestations of Coronavirus
Disease 2019 Pandemic: A Systemic Review. Front. Neurol. 2020, 11, 498. DOI: 10.3389/
fneur.2020.00498.
[15] Xydakis, M. S.; Dehgani-Mobaraki, P.; Holbrook, E. H.; Geisthoff, U. W.; Bauer, C.;
Hautefort, C.; Herman, P.; Manley, G. T.; Lyon, D. M.; Hopkins, C.; et al. Smell and Taste
Dysfunction in Patients with COVID-19. Lancet Infect Dis. 2020, 20(9), 1015–1016.
[16] Zhou, L.; Zhang, M.; Wang, J.; Gao, J. Sars-Cov-2: Underestimated Damage to Nervous
System. Travel Med Infectious Dis. 2020, 36, 101642. DOI: 10.1016/j.tmaid.2020.101642.
[17] Zubair, A. S.; McAlpine, L. S.; Gardin, T.; Farhadian, S.; Kuruvilla, D. E.; Spudich, S.
Neuropathogenesis and Neurologic Manifestations of the Coronaviruses in the Age of
Coronavirus Disease 2019: A Review. JAMA neurol. 2020, 77(8), 1018–1027. DOI:
10.1001/jamaneurol.2020.2065.
[18] Chen, C.; Zhang, X. R.; Ju, Z. Y.; He, W. F. [Advances in the Research of Mechanism and
Related Immunotherapy on the Cytokine Storm Induced by Coronavirus Disease 2019].
Zhonghua Shao Shang Za Zhi = Zhonghua Shaoshang Zazhi = Chin J Burns. 2020, 36(6),
471–475. DOI: 10.3760/cma.j.cn501120-20200224-00088.
[19] Wan S, Yi Q, Fan S, et al. Characteristics of Lymphocyte Subsets and Cytokines in
Peripheral Blood of 123 Hospitalized Patients with 2019 Novel Coronavirus Pneumonia
(NCP). medRxiv. 2020, 2020.02.10.20021832.
[20] Mehta, P.; McAuley, D. F.; Brown, M.; Sanchez, E.; Tattersall, R. S.; Manson, J. J.
COVID-19: Consider Cytokine Storm Syndromes and Immunosuppression. Lancet.
2020, 395(10229), 1033–1034. DOI: 10.1016/S0140-6736(20)30628-0.
[21] Sellner, J.; Taba, P. The Need for Neurologists in the Care of COVID-19 Patients.Eur J
Neurol. 2020, 27(9), e31–e2. DOI: 10.1111/ene.14257.
[22] Pereira, A.;. Long-Term Neurological Threats of COVID-19: A Call to Update the
Thinking about the Outcomes of the Coronavirus Pandemic. Front. Neurol. 2020, 11,
308. DOI: 10.3389/fneur.2020.00308.
12 M. M. ANWAR
[23] Guan, W.-J.; Ni, Z.-Y.; Hu, Y.; Liang, W.-H.; Ou, C.-Q.; He, J.-X.; Liu, L.; Shan, H.;
Lei, C.-L.; Hui, D. S. C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in
China. N. Engl. J. Med. 2020, 382(18), 1708–1720.
[24] Hardy, R. S.; Raza, K.; Cooper, M. S. Therapeutic Glucocorticoids: Mechanisms of
Actions in Rheumatic Diseases. Nat Rev Rheumatol. 2020, 16(3), 133–144. DOI:
10.1038/s41584-020-0371-y.
[25] Alhazzani, W.; Møller, M. H.; Arabi, Y. M.; Loeb, M.; Gong, M. N.; Fan, E.;
Oczkowski, S.; Levy, M. M.; Derde, L.; Dzierba, A.; et al. Surviving Sepsis Campaign:
Guidelines on the Management of Critically Ill Adults with Coronavirus Disease 2019
(COVID-19). Crit. Care Med. 2020, 48(6), e440–e69.
[26] Horby, P.; Lim, W. S.; Emberson, J.; Mafham, M.; Bell, J.; Linsell, L.; et al. Effect of
Dexamethasone in Hospitalized Patients with COVID-19: Preliminary Report. medRxiv.
2020, 2020.06.22.20137273.
[27] Villar, J.; Ferrando, C.; Martínez, D.; Ambrós, A.; Muñoz, T.; Soler, J. A.; Aguilar, G.;
Alba, F.; González-Higueras, E.; Conesa, L. A.; et al. Dexamethasone Treatment for the
Acute Respiratory Distress Syndrome: A Multicentre, Randomised Controlled Trial.
Lancet Respir Med. 2020, 8(3), 267–276.
[28] Russell, C. D.; Millar, J. E.; Baillie, J. K. Clinical Evidence Does Not Support
Corticosteroid Treatment for 2019-nCoV Lung Injury. Lancet. 2020, 395(10223),
473–475. DOI: 10.1016/S0140-6736(20)30317-2.
[29] Zhou Y, Fu B, Zheng X, et al. Aberrant Pathogenic GM-CSF+ T Cells and Inflammatory
CD14+CD16+ Monocytes in Severe Pulmonary Syndrome Patients of a New
Coronavirus. bioRxiv. 2020, 2020.02.12.945576.
[30] Kimmig LM, Wu D, Gold M, Pettit NN, Pitrak D, Mueller J, et al. IL-6 Inhibition in
Critically Ill COVID-19 Patients Is Associated With Increased Secondary Infections.
Front Med (Lausanne). 2020;7:583897.
[31] Xu, X.; Han, M.; Li, T.; Sun, W.; Wang, D.; Fu, B.; Zhou, Y.; Zheng, X.; Yang, Y.; Li, X.;
et al. Effective Treatment of Severe COVID-19 Patients with Tocilizumab. Proc. National
Academy Sci. 2020, 117(20), 10970–10975.
[32] De Luna, G.; Habibi, A.; Deux, J. F.; Colard, M.; Pham Hung d’Alexandry
d’Orengiani, A. L.; Schlemmer, F.; Joher, N.; Kassasseya, C.; Pawlotsky, J. M.;
Ourghanlian, C.; et al. Rapid and Severe Covid-19 Pneumonia with Severe Acute
Chest Syndrome in a Sickle Cell Patient Successfully Treated with Tocilizumab. Am.
J. Hematol. 2020, 95(7), 876–878.
[33] Cellina, M.; Orsi, M.; Bombaci, F.; Sala, M.; Marino, P.; Oliva, G. Favorable Changes of
CT Findings in a Patient with COVID-19 Pneumonia after Treatment with Tocilizumab.
Diagnostic and Interventional Imaging. 2020, 101(5), 323–324. DOI: 10.1016/j.
diii.2020.03.010.
[34] Zhang, C.; Wu, Z.; Li, J. W.; Zhao, H.; Wang, G. Q. Cytokine Release Syndrome in Severe
COVID-19: Interleukin-6 Receptor Antagonist Tocilizumab May Be the Key to Reduce
Mortality. Int. J. Antimicrob. Agents. 2020, 55(5), 105954. DOI: 10.1016/j.
ijantimicag.2020.105954.
[35] Vermersch P, Oh J, Cascione M, Oreja-Guevara C, Gobbi C, Travis LH, et al.
Teriflunomide Vs Injectable Disease Modifying Therapies for Relapsing Forms of MS.
Mult. Scler. Relat. Disord. 2020, 43, 102158.
[36] Comi, G.; Miller, A. E.; Benamor, M.; Truffinet, P.; Poole, E. M.; Freedman, M. S.
Characterizing Lymphocyte Counts and Infection Rates with Long-term
Teriflunomide Treatment: Pooled Analysis of Clinical Trials. Mult. Scler. 2020, 26(9),
1083–1092. DOI: 10.1177/1352458519851981.
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 13
[37] Maghzi, A. H.; Houtchens, M. K.; Preziosa, P.; Ionete, C.; Beretich, B. D.;
Stankiewicz, J. M.; Tauhid, S.; Cabot, A.; Berriosmorales, I.; Schwartz, T. H. W.; et al.
COVID-19 in Teriflunomide-treated Patients with Multiple Sclerosis. J. Neurol. 2020,
267(10), 2790–2796.
[38] Galeotti, C.; Kaveri, S. V.; Bayry, J. Intravenous Immunoglobulin Immunotherapy for
Coronavirus Disease-19 (COVID-19). Clin. Transl. Immunol. 2020, 9(10), e1198. DOI:
10.1002/cti2.1198.
[39] Nierhaus, A.; Berlot, G.; Kindgen-Milles, D.; Müller, E.; Girardis, M. Best-practice IgM-
and IgA-enriched Immunoglobulin Use in Patients with Sepsis. Ann Intens Care. 2020,
10(1), 132. DOI: 10.1186/s13613-020-00740-1.
[40] Muccioli, L.; Pensato, U.; Bernabè, G.; Ferri, L.; Tappatà, M.; Volpi, L.; et al.
Intravenous Immunoglobulin Therapy in COVID-19-related Encephalopathy.
J. Neurol. 2020, 1–5.
[41] Klok, F. A.; Kruip, M.; van der Meer, N. J. M.; Arbous, M. S.; Gommers, D.; Kant, K. M.;
Kaptein, F. H. J.; van Paassen, J.; Stals, M. A. M.; Huisman, M. V.; et al. Confirmation of
the High Cumulative Incidence of Thrombotic Complications in Critically Ill ICU
Patients with COVID-19: An Updated Analysis. Thromb. Res. 2020, 191, 148–150.
DOI: 10.1016/j.thromres.2020.04.041.
[42] Chiarini, M.; Paghera, S.; Moratto, D.; Rossi, N.; Giacomelli, M.; Badolato, R.; Capra, R.;
Imberti, L.; et al. Immunologic Characterization of a Immunosuppressed Multiple
Sclerosis Patient that Recovered from SARS-CoV-2 Infection. J. Neuroimmunol. 2020,
345, 577282. DOI: 10.1016/j.jneuroim.2020.577282.
[43] Tzilas, V.; Manali, E.; Papiris, S.; Bouros, D. Intravenous Immunoglobulin for the
Treatment of COVID-19: A Promising Tool. Respiration. 2020, 1–3. DOI: 10.1159/
000512727.
[44] Dersch, R.; Wehrum, T.; Fähndrich, S.; Engelhardt, M.; Rauer, S.; Berger, B. COVID-19
Pneumonia in a Multiple Sclerosis Patient with Severe Lymphopenia Due to Recent
Cladribine Treatment. Mult. Scler. 2020, 26(10), 1264–1266.
[45] Schweitzer, F.; Laurent, S.; Fink, G. R.; Barnett, M. H.; Hartung, H. P.; Warnke, C. Effects
of Disease-modifying Therapy on Peripheral Leukocytes in Patients with Multiple
Sclerosis. J. Neurol. 2020. DOI: 10.1007/s00415-019-09690-6.
[46] Jack, D.; Nolting, A.; Galazka, A. Favorable Outcomes after COVID-19 Infection in
Multiple Sclerosis Patients Treated with Cladribine Tablets. Mult. Scler. Relat. Disord.
2020, 46, 102469. DOI: 10.1016/j.msard.2020.102469.
[47] Moser, T.; Schwenker, K.; Seiberl, M.; Feige, J.; Akgün, K.; Haschke-Becher, E.;
Ziemssen, T.; Sellner, J.; et al. Long-term Peripheral Immune Cell Profiling Reveals
Further Targets of Oral Cladribine in MS. Ann. Clin. Transl. Neurol. 2020, 7(11),
2199–2212.
[48] De Angelis, M.; Petracca, M.; Lanzillo, R.; Brescia Morra, V.; Moccia, M. Mild or No
COVID-19 Symptoms in Cladribine-treated Multiple Sclerosis: Two Cases and
Implications for Clinical Practice. Mult. Scler. Relat. Disord. 2020, 45, 102452. DOI:
10.1016/j.msard.2020.102452.
[49] Garcia-Montoya, L.; Villota-Eraso, C.; Yusof, M. Y. M.; Vital, E. M.; Emery, P. Lessons
for Rituximab Therapy in Patients with Rheumatoid Arthritis. Lancet Rheumatol. 2020,
2(8), e497–e509. DOI: 10.1016/S2665-9913(20)30033-3.
[50] Luna, G.; Alping, P.; Burman, J.; Fink, K.; Fogdell-Hahn, A.; Gunnarsson, M.; et al.
Infection Risks among Patients with Multiple Sclerosis Treated with Fingolimod,
Natalizumab, Rituximab, and Injectable Therapies. JAMA neurol. 2020, 77(2), 184–191.
14 M. M. ANWAR
[51] Kondratiuk, A.; Muraro, P.; Johnson, M.; Sing-Curry, V.; Pritchard, J.; Dorsey, R.; et al.
The Use of Rituximab in Neurological Diseases; Real World Data from a Neuroscience
Centre (2575). Neurology. 2020, 94(15 Supplement), 2575.
[52] Wurm, H.; Attfield, K.; Iversen, A. K. N.; Gold, R.; Fugger, L.; Haghikia, A. Recovery
from COVID-19 in a B-cell-depleted Multiple Sclerosis Patient. Mult. Scler. J. 2020, 26
(10), 1261–1264. DOI: 10.1177/1352458520943791.
[53] Mehta, P.; Porter, J. C.; Chambers, R. C.; Isenberg, D. A.; Reddy, V. B-cell Depletion with
Rituximab in the COVID-19 Pandemic: Where Do We Stand? Lancet Rheumatol. 2020,
2(10), e589–e90. DOI: 10.1016/S2665-9913(20)30270-8.
[54] Guilpain, P.; Le Bihan, C.; Foulongne, V.; Taourel, P.; Pansu, N.; Maria, A. T. J.; et al.
Rituximab for Granulomatosis with Polyangiitis in the Pandemic of Covid-19: Lessons
from a Case with Severe Pneumonia. Ann. Rheumatic Dis. 2020, annrheumdis-2020-
217549.
[55] Goldschmidt, C. H.; Hua, L. H. Re-Evaluating the Use of IFN-β and Relapsing Multiple
Sclerosis: Safety, Efficacy and Place in Therapy. Degener Neurol Neuromuscul Dis. 2020,
10, 29–38. DOI: 10.2147/DNND.S224912.
[56] Severa, M.; Farina, C.; Salvetti, M.; Coccia, E. M. Three Decades of Interferon-β in
Multiple Sclerosis: Can We RepurposeThis Information for the Management of SARS-
CoV2 Infection? Front. Immunol. 2020, 11, 1459. DOI: 10.3389/fimmu.2020.01459.
[57] Alhazzani, W.; Møller, M. H.; Arabi, Y. M.; Loeb, M.; Gong, M. N.; Fan, E.; et al.
Surviving Sepsis Campaign: Guidelines on the Management of Critically Ill Adults
with Coronavirus Disease 2019 (COVID-19). Crit. Care Med. 2020, 48(6), e440–e69.
[58] Sallard, E.; Lescure, F.-X.; Yazdanpanah, Y.; Mentre, F.; Peiffer-Smadja, N. Type 1
Interferons as a Potential Treatment against COVID-19. Antiviral Res. 2020, 178,
104791. DOI: 10.1016/j.antiviral.2020.104791.
[59] Hsu, J.;. Covid-19: What Now for Remdesivir? BMJ. 2020, 371, m4457. DOI: 10.1136/
bmj.m4457.
[60] Grein, J.; Ohmagari, N.; Shin, D.; Diaz, G.; Asperges, E.; Castagna, A.; Feldt, T.;
Green, G.; Green, M. L.; Lescure, F.-X.; et al. Compassionate Use of Remdesivir for
Patients with Severe Covid-19. N. Engl. J. Med. 2020, 382(24), 2327–2336.
[61] Wang, L.-Y.; Cui, -J.-J.; Ouyang, Q.-Y.; Zhan, Y.; Guo, C.-X.; Yin, J.-Y. Remdesivir and
COVID-19. Lancet. 2020, 396(10256), 953–954. DOI: 10.1016/S0140-6736(20)32019-5.
[62] Wang, Y.; Zhang, D.; Du, G.; Du, R.; Zhao, J.; Jin, Y.; Fu, S.; Gao, L.; Cheng, Z.; Lu, Q.;
et al. Remdesivir in Adults with Severe COVID-19: A Randomised, Double-blind,
Placebo-controlled, Multicentre Trial. Lancet. 2020, 395(10236), 1569–1578.
[63] Gulati, G.; Kelly, B. D. Does Remdesivir Have Any Neuropsychiatric Adverse Effects?
Irish J Psychol Med. 2020, 1–2.
[64] Sheahan, T. P.; Sims, A. C.; Leist, S. R.; Schäfer, A.; Won, J.; Brown, A. J.; Montgomery, S. A.;
Hogg, A.; Babusis, D.; Clarke, M. O.; et al. Comparative Therapeutic Efficacy of Remdesivir
and Combination Lopinavir, Ritonavir, and Interferon Beta against MERS-CoV. 2020, 11(1),
222. DOI: 10.1038/s41467-019-13940-6.
[65] Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.;
Xiao, G.; et al. Remdesivir and Chloroquine Effectively Inhibit the Recently Emerged
Novel Coronavirus (2019-ncov) in Vitro. Nat Commun, 2020, 30(3), 269–271. DOI:
10.1038/s41422-020-0282-0.
[66] Butzkueven, H.; Kappos, L.; Wiendl, H.; Trojano, M.; Spelman, T.; Chang, I.; et al. Long-
term Safety and Effectiveness of Natalizumab Treatment in Clinical Practice: 10 Years of
Real-world Data from the Tysabri Observational Program (TOP). J Neurol Neurosurg
Psychiatry. 2020, 91(6), 660–668.
JOURNAL OF IMMUNOASSAY AND IMMUNOCHEMISTRY 15
[67] Abraham, A.; Nicholson, L.; Dick, A.; Rice, C.; Atan, D. Intermediate Uveitis Associated
with MS. Diagn. Clin. Features Pathogen. Mech. Recommendations Manage. 2021, 8(1),
e909.
[68] Borriello, G.; Ianniello, A. COVID-19 Occurring during Natalizumab Treatment: A Case
Report in a Patient with Extended Interval Dosing Approach. Multiple sclerosis and
related disorders. 2020;41:102165.
[69] Aguirre, C.; Meca-Lallana, V.; Barrios-Blandino, A.; Del Río, B.; Vivancos, J. Covid-19 in
a Patient with Multiple Sclerosis Treated with Natalizumab: May the Blockade of
Integrins Have a Protective Role? Mult. Scler. Relat. Disord. 2020, 44, 102250. DOI:
10.1016/j.msard.2020.102250.
[70] Jeyanathan, M.; Afkhami, S.; Smaill, F.; Miller, M. S.; Lichty, B. D.; Xing, Z.
Immunological Considerations for COVID-19 Vaccine Strategies. Nat. Rev. Immunol.
2020, 20(10), 615–632. DOI: 10.1038/s41577-020-00434-6.
[71] Yang, L.; Tian, D.; Liu, W. [Strategies for Vaccine Development of COVID-19]. Sheng
Wu Gong Cheng Xue Bao = Chin J Biotechnol. 2020, 36(4), 593–604. DOI: 10.13345/j.
cjb.200094.
[72] Bennet, B. M.; Wolf, J.; Laureano, R.; Sellers, R. S. Review of Current Vaccine
Development Strategies to Prevent Coronavirus Disease 2019 (COVID-19). Toxicol
Pathol. 2020, 48(7), 800–809. DOI: 10.1177/0192623320959090.
[73] Bonam, S. R.; Kaveri, S. V.; Sakuntabhai, A.; Gilardin, L.; Bayry, J. Adjunct
Immunotherapies for the Management of Severely Ill COVID-19 Patients. Cell Rep
Med. 2020, 1(2), 100016. DOI: 10.1016/j.xcrm.2020.100016.
[74] Mansourabadi, A. H.; Sadeghalvad, M.; Mohammadi-Motlagh, H.-R.; Rezaei, N. The
Immune System as a Target for Therapy of SARS-CoV-2: A Systematic Review of the
Current Immunotherapies for COVID-19. Life Sci. 2020, 258, 118185. DOI: 10.1016/j.
lfs.2020.118185.
16 M. M. ANWAR
... Cytokines released during a cytokine storm include interleukins such as IL-6 and IL-1β, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor (GMCSF) and tumor necrosis factor. The release of proinflammatory cytokines, including IL-6, is among the main reasons for mortality associated with the disease [9]. CD4+ T lymphocytes are rapidly activated and differentiated into T helper cells and generate GMCSF. ...
... Tocilizumab (a human form of anti-IL-6 receptor antibody), which was originally used for the treatment of rheumatoid arthritis and systemic juvenile idiopathic arthritis, has recently been suggested for the treatment of severe COVID-19 [9]. Tocilizumab selectively and competitively binds to soluble IL-6 receptors and subsequently blocks the IL-6 signaling, which inhibits the IL-6 receptors that lead to a cytokine storm. ...
... Tocilizumab selectively and competitively binds to soluble IL-6 receptors and subsequently blocks the IL-6 signaling, which inhibits the IL-6 receptors that lead to a cytokine storm. Tocilizumab can be applied as first-line therapy to inhibit cytokine release, especially in severe COVID-19 cases [9]. Treatment with tocilizumab has been reported previously, especially in cases of new-onset refractory SE [10], and has been demonstrated to be useful in the treatment of resistant autoimmune encephalitis [11]. ...
Article
Background: COVID-19 is a novel infectious disease caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), in which, neurological complications have been increasingly recognized. Acute symptomatic epileptic seizures and status epilepticus (SE) is a frequently reported neurological complication associated with this infection. The nervous system damage caused by SARS-CoV-2 may be mediated by the immune system. Interleukin 6 (IL-6), an important component of the cytokine storm, is directly correlated with the severity of symptoms. Tocilizumab is an inhibitor of IL-6 receptors, which blocks IL-6-mediated signal transduction and is used in the treatment of COVID-19 and status epilepticus. Case report: We present a patient with the Unverricht-Lundborg Disease (ULD) who had developed refractory recurrent status epilepticus during COVID-19 infection, which was finally controlled by treatment with tocilizumab. Discussion: Tocilizumab, an IL-6 inhibitor, may be considered as a treatment option in patients with SE and refractory seizures.
... The cytokine storm phenomenon is an increase of pro-inflammatory cytokines levels in the serum, such as IL-2, IL-6 and IL-1β, IL-17, IL-8, G-CSF, GM-CSF, IP10, MCP1, MIP1α (also known as CCL3), and TNF [33], and causes acute respiratory distress syndrome (ARDS) in COVID-19 severe cases [106]. It has indicated that increased cytokines concentration, especially IL-6, TNFα, and IL-1β 9 of 15 has a strong effect on working memory and attention. ...
Preprint
Full-text available
As the name implies, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a single-stranded RNA virus and a member of the corona virus family, primarily affecting the upper respiratory system and the lungs. Like many other respiratory viruses, SARS-CoV-2 can spread to other organ systems. Apart from causing diarrhea, another most common but debilitating complication caused by the SARS-CoV-2 is neurological symptoms and cognitive difficulties, which occur in up to two thirds of hospitalized covid patients and ranging from shortness of concentration, overall declined cognitive speed to executive or memory function impairment. Neuro-cognitive dysfunction and “brain fog” are frequently present in COVID-19 cases, which can last several months after the infection, leading to disruption of daily life. Cumulative evidence suggests that SARS-CoV-2 affects vasculature in the extra pulmonary systems directly or indirectly, leading to impairment of endothelial function and even multi-organ damage. The post COVID-19 long-lasting neurocognitive impairments have not been studied fully; and the underlying mechanism remains elusive. In this review, we summarize the current understanding of the effects of COVID-19 on vascular dysfunction and how vascular dysfunction leads to cognitive impairment in patients.
... The cytokine storm phenomenon is an increase in pro-inflammatory cytokine levels in the serum, such as IL-2, IL-6 and IL-1β, IL-17, IL-8, G-CSF, GM-CSF, IP10, MCP1, MIP1α (also known as CCL3), and TNF [36], and causes acute respiratory distress syndrome (ARDS) in severe COVID-19 cases [117]. It has been indicated that increased cytokine concentration, especially IL-6, TNFα, and IL-1β, has a strong effect on working memory and attention. ...
Article
Full-text available
Simple Summary Although SARS-CoV-2 primarily affects the upper respiratory system and the lungs, it can also affect vasculature, leading to the impairment of endothelial and multi-organ function. SARS-CoV-2 can trigger the release of pro-inflammatory cytokines, leading to disruption of tight junction proteins between endothelial cells and impairment of the blood-brain barrier (BBB). This process further allows the infiltration of immune cells and other particles into the brain, worsening brain injury. Prolonged neuro-inflammation and disruption of the BBB have been postulated as the potential primary causes of both acute and chronic cognitive dysfunction in COVID-19 patients. This review explores the effects of COVID-19 on vascular dysfunction and consequent cognitive impairment in patients. Abstract Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a single-stranded RNA virus and a member of the corona virus family, primarily affecting the upper respiratory system and the lungs. Like many other respiratory viruses, SARS-CoV-2 can spread to other organ systems. Apart from causing diarrhea, another very common but debilitating complication caused by SARS-CoV-2 is neurological symptoms and cognitive difficulties, which occur in up to two thirds of hospitalized COVID-19 patients and range from shortness of concentration and overall declined cognitive speed to executive or memory function impairment. Neuro-cognitive dysfunction and “brain fog” are frequently present in COVID-19 cases, which can last several months after the infection, leading to disruption of daily life. Cumulative evidence suggests that SARS-CoV-2 affects vasculature in the extra-pulmonary systems directly or indirectly, leading to impairment of endothelial function and even multi-organ damage. The post COVID-19 long-lasting neurocognitive impairments have not been studied fully and their underlying mechanism remains elusive. In this review, we summarize the current understanding of the effects of COVID-19 on vascular dysfunction and how vascular dysfunction leads to cognitive impairment in patients.
... The pathophysiological mechanisms of headache attributed to vaccination against COVID-19 remain unclear. Whether the S glycoprotein of SARS-CoV-2, synthesized intracellularly using the gene supplied by the vaccine, is itself responsible for the headache or whether it is due to the resulting immune response triggered by that protein [8,[26][27][28][29][30][31][32] must remain open at present. The intracellular formation of the spike protein and the triggered immune response could be directly related to the development of the headache phenotype described, including the accompanying symptoms of fatigue, chills, exhaustion, weakness, joint pain, and dizziness. ...
Article
Full-text available
IntroductionThe most frequently reported neurological adverse event of ChAdOx1 nCoV-19 (AZD1222) vaccine is headache in 57.5%. Several cases of cerebral venous thrombosis (CVT) have developed after vaccination. Headache is the leading symptom of CVT. For the differential diagnosis of headaches attributed to this vaccine and headaches attributed to CVT, it is of central clinical importance whether and, if so, how the phenotypes and course of these headaches can be differentiated. The study aims to examine in detail the phenotype of headache attributed to this vaccine.Methods Data on the clinical features and corresponding variables were recorded using a standardized online questionnaire in this multicenter observational cohort study. The primary outcomes of this study are the clinical features of headaches after vaccination.FindingsA total of 2464 participants reported headaches after vaccination with the ChAdOx1 nCoV-19 (AZD1222) vaccine. On average, headaches occurred 14.5 ± 21.6 h after vaccination and lasted 16.3 ± 30.4 h. A bilateral location was described by 75.8% of participants. This is most often found on the forehead (40.0%) and temples (31.4%); 50.4% reported a pressing and 37.7% a dull pain character. Headache intensity was most often severe (38.7%), moderate (35.2%), or very severe (15.5%). Accompanying symptoms were most commonly fatigue (44.8%), chills (36.1%), exhaustion (34.9%), and fever (30.4%).Conclusion Headaches attributed to COVID-19 vaccination with the ChAdOx1 nCoV-19 (AZD1222) vaccine demonstrate an extensive and characteristic complex of symptoms. The findings have several important clinical implications for the differentiation of post-vaccinal headache and other primary as well as secondary headaches.
... It must remain open at present, whether the spike protein, synthesized intracellularly using the mRNA supplied by the vaccine, is itself responsible for the headache or whether it is due to the resulting immune response triggered by that protein. 5,[22][23][24][25][26][27] The intracellular formation of the spike protein and the immune response triggered by it could be directly related to the development of the headache phenotype described, including the accompanying symptoms of tiredness, exhaustion, muscle pain, dizziness, poor concentration, chills and fever. It is speculated that microorganisms can activate anti-inflammatory substances, such as nitric oxide, prostaglandins and cytokines. ...
Article
Full-text available
The novel coronavirus SARS-CoV-2 causes the infectious disease Covid-19. Newly developed mRNA vaccines can prevent the spread of the virus. Headache is the most common neurological symptom in over 50% of those vaccinated. Detailed information about the clinical characteristics of this form of headache has not yet been described. The aim of the study is to examine in detail the clinical characteristics of headaches occurring after vaccination against Covid-19 with the BNT162b2 mRNA Covid-19 vaccine for the first time. In a multicenter observational cohort study data on the clinical features and corresponding variables were recorded using a standardized online questionnaire. The questionnaire was circulated to 12,000 residential care homes of the elderly as well as tertiary university hospitals in Germany and the United Arab Emirates. The primary outcomes of this study are the clinical features of headache after vaccination. Comorbidities, treatment with medication and sociodemographic variables are also analyzed. A total of 2349 participants reported headaches after vaccination with the BNT162b2 mRNA Covid-19 vaccine. Headaches occur an average of 18.0 ± 27.0 hours after vaccination and last an average duration of 14.2 ± 21.3 hours. Only 9.7% of those affected also report headaches resulting from previous vaccinations. In 66.6% of the participants headache occurs as a single episode. A bilateral location is indicated by 73.1% of the participants. This is most often found on the forehead (38.0%) and temples (32.1%). A pressing pain character is indicated by 49.2% and 40.7% report a dull pain character. The pain intensity is most often moderate (46.2%), severe (32.1%) or very severe (8.2%). The most common accompanying symptoms are fatigue (38.8%), exhaustion (25.7%) and muscle pain (23.4%). Headaches after Covid-19 vaccination show an extensive complex of symptoms. The constellation of accompanying symptoms together with the temporal and spatial headache characteristics delimit a distinctive headache phenotype.
... The medical research community had dedicated a lot of effort and resources to find about the causes of these complications, treatment options that were available to treat these complications, and methods to prevent these complications (Hus, Salomon-Perzyński, Tomasiewicz, & Robak, 2020;Kaseda & Levine, 2020;Mosaddeghi, Shahabinezhad, Dorvash, Goodarzi, & Negahdaripour, 2020;Saleh et al., 2020;Taherifard & Taherifard, 2020;Wagner et al., 2021). Similarly, several studies had been conducted to analyze the complications of the COVID-19 pandemic worldwide including (Ai et al., 2020;Akacha et al., 2020;Anwar, 2020;Bartel, Sherry, & Stewart, 2020;Collins & Levenson, 2020;. Further, it is important to mention here that several studies has been conducted employing different methods including qualitative/quantitative survey (Anwar, Shoaib, & Javed, 2013;Shoaib, Khan, & Shaukat, 2012;Shoaib, Latif, & Usmani, 2013;Shoaib, Saeed, & Cheema, 2012;Shoaib & Ullah, 2019, 2021a, 2021b and the bibliometric or scientometric analysis techniques to show the trend of data and data visualization (Ali, Shoaib, & Asad, 2021;Shoaib, Abdullah, & Ali, 2020;Shoaib, Rasool, & Anwar, 2021;Ullah & Shoaib, 2021). ...
Article
This study focused to evaluate the published articles on PubMed whose focus was to study the different relations between COVID 19, and different complications caused due to it. The main complications that were used to formulate this study are Deep Venous Thrombosis (DVT), Stroke, Pulmonary Embolism, Disseminated Intravascular Coagulation (DIC), Myocardial Infarction (MI), anosmia, and pneumonia. The data was extracted from the PubMed database on March 20, 2021at 03:20 PM, PST with the period of 2020 to March 20, 2021. We used Biblioshiny, VOSviewer, and MS Excel software to analyze data and found 2375 published documents. The results revealed that Pneumonia and COVID-19 was the top topic out of 2375 published documents, journal articles as the top type, and the top year was 2020. Similarly, study findings also asserted that Guaraldi, Giovanni was the top author, China was a top country, COVID-19 top keyword, and Stroke was the top source of published documents. Further, the results were illustrated in tables and figures to show the trend of the published document.
... Besides, in stage 2, the percentage of monocytes and macrophages is increased following viral infection, which leads to the release of a large amount of proinflammatory cytokines, referred to as cytokine storm [55,56]. This phenomenon is an enhancement of serum levels of pro-inflammatory cytokines including IL-6 and IL-1β, as well as IL-2, IL-8, IL-17, G-CSF, GM-CSF, IP10, MCP1, MIP1α (also known as CCL3), and TNF, which has found in most patients with severe COVID-19 [57]. Accordingly, a dysfunctional and disharmonic immune response in severe COVID-19 cases initiates an extensive lung and systemic inflammation by triggering cytokine storm, that possibly worsens infection in the brain [58]. ...
Article
The coronavirus disease of 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus-2 (SARS CoV-2), that already appeared as a global pandemic. Presentation of the disease often includes upper respiratory symptoms like dry cough, dyspnea, chest pain, and rhinorrhea that can develop to respiratory failure, needing intubation. Furthermore, the occurrence of acute and subacute neurological manifestations such as stroke, encephalitis, headache, and seizures are frequently stated in patients with COVID-19. One of the reported neurological complications of severe COVID-19 is the demolition of the myelin sheath. Indeed, the complex immunological dysfunction provides a substrate for the development of demyelination. Nevertheless, few published reports in the literature describe demyelination in subjects with COVID-19. In this short narrative review, we discuss probable pathological mechanisms that may trigger demyelination in patients with SARS‐CoV‐2 infection and summarize the clinical evidence, confirming SARS-CoV-2 condition as a risk factor for the destruction of myelin.
Article
Full-text available
Despite decades of repeated and intense research, the etiology of sudden Alzheimer's disease (AD) symptoms is still unclear. AD progressive pathology mainly involves neuron damage, depositions of amyloid‐beta (Aβ), and hyperphosphorylated tau protein. All these defects are manifested by exaggerated cytokine storm and neuroinflammation leading to irreversible brain damage in the long term. Despite the numerous risks and drawbacks associated with AD, it is believed that there is a hidden unknown causative and predisposing factors for AD. Extracellular vesicles (EVs) are small vesicles released by cells as a type of intercellular communication. Several pieces of evidence support the inclusion of viral components within EVs facilitating their penetration into the blood–brain barrier leading to neuroinflammation. In light of the SARS‐CoV‐19 pandemic and its related neurological complications, it is mandatory to highlight the possibility and viability of viral infections such as varicella‐zoster virus (VZV) and herpes simplex virus (HSV) on the onset of AD. Herein, the author is investigating the potential role of VZV and HSV along with highlighting the suggested route of pathogenesis entry resulting in AD manifestations. Additionally, this review aims to summarize the role of EVs in mediating the central nervous system viral infections leading to AD.
Article
Full-text available
Background: Anti-inflammatory therapies such as IL-6 inhibition have been proposed for COVID-19 in a vacuum of evidence-based treatment. However, abrogating the inflammatory response in infectious diseases may impair a desired host response and pre-dispose to secondary infections. Methods: We retrospectively reviewed the medical record of critically ill COVID-19 patients during an 8-week span and compared the prevalence of secondary infection and outcomes in patients who did and did not receive tocilizumab. Additionally, we included representative histopathologic post-mortem findings from several COVID-19 cases that underwent autopsy at our institution. Results: One hundred eleven patients were identified, of which 54 had received tocilizumab while 57 had not. Receiving tocilizumab was associated with a higher risk of secondary bacterial (48.1 vs. 28.1%; p = 0.029 and fungal (5.6 vs. 0%; p = 0.112) infections. Consistent with higher number of infections, patients who received tocilizumab had higher mortality (35.2 vs. 19.3%; p = 0.020). Seven cases underwent autopsy. In three cases who received tocilizumab, there was evidence of pneumonia on pathology. Of the four cases that had not been given tocilizumab, two showed evidence of aspiration pneumonia and two exhibited diffuse alveolar damage. Conclusions: Experimental therapies are currently being applied to COVID-19 outside of clinical trials. Anti-inflammatory therapies such as anti-IL-6 therapy have the potential to impair viral clearance, pre-dispose to secondary infection, and cause harm. We seek to raise physician awareness of these issues and highlight the need to better understand the immune response in COVID-19.
Article
Full-text available
Serious of unpredictable drawbacks of Coronavirus 2019 (COVID-19) infectious disease caused by SARS-COV-2 on the nervous system, have been widely noticed among the huge number of infected people. It was found that this type of newly revolving pandemic infection mainly infects the human respiratory tract causing mild to moderate symptoms, however, the hidden door side of COVID-19 is via penetrating the brain, revealing a huge threat especially to elderly people who are more susceptible to its severe side effects and even death to more extent. Almost 80% of COVID-19 patients suffer from severe neurological manifestations including dizziness, headache, unconscious, irritability, dysfunction in smell, and taste accompanied by muscle fatigue. Herein, we are trying to address the direct neuroinvasive pathway of COVID-19 into human brain cells which is mainly through the olfactory route leading to long-term neurological complications. In addition to highlighting the ability of COVID-19 infection to intensify a pre-existing AD to a more prominent severe stage. The other thing to emphasize is whether AD patients with a highly prominent activation of local immune responses are more or less exposed to getting infected with COVID-19. Along with underlying the hypothesis that the susceptibility to COVID-19 infection may lead to a future risk for neurodegenerative diseases including Alzheimer’s disease (AD).
Article
Full-text available
Intravenous immunoglobulin (IVIG), a pooled normal IgG from several thousand healthy donors and one of the commonly used immunotherapeutic molecules for the management of autoimmune and inflammatory diseases, has been explored for the treatment of coronavirus disease‐19 (COVID‐19). Although placebo‐controlled, double‐blind randomised clinical trials are lacking, current data from either retrospective, case series or open‐label randomised controlled trials provide an indicator that IVIG immunotherapy could benefit severe and critically ill COVID‐19 patients.
Article
Full-text available
Background: Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Despite treatment being in line with current guidelines, mortality remains high in those with septic shock. Intravenous immunoglobulins represent a promising therapy to modulate both the pro- and anti-inflammatory processes and can contribute to the elimination of pathogens. In this context, there is evidence of the benefits of immunoglobulin M (IgM)- and immunoglobulin A (IgA)-enriched immunoglobulin therapy for sepsis. This manuscript aims to summarize current relevant data to provide expert opinions on best practice for the use of an IgM- and IgA-enriched immunoglobulin (Pentaglobin) in adult patients with sepsis. Main text: Sepsis patients with hyperinflammation and patients with immunosuppression may benefit most from treatment with IgM- and IgA-enriched immunoglobulin (Pentaglobin). Patients with hyperinflammation present with phenotypes that manifest throughout the body, whilst the clinical characteristics of immunosuppression are less clear. Potential biomarkers for hyperinflammation include elevated procalcitonin, interleukin-6, endotoxin activity and C-reactive protein, although thresholds for these are not well-defined. Convenient biomarkers for identifying patients in a stage of immune-paralysis are still matter of debate, though human leukocyte antigen-antigen D related expression on monocytes, lymphocyte count and viral reactivation have been proposed. The timing of treatment is potentially more critical for treatment efficacy in patients with hyperinflammation compared with patients who are in an immunosuppressed stage. Due to the lack of evidence, definitive dosage recommendations for either population cannot be made, though we suggest that patients with hyperinflammation should receive an initial bolus at a rate of up to 0.6 mL (30 mg)/kg/h for 6 h followed by a continuous maintenance rate of 0.2 mL (10 mg)/kg/hour for ≥ 72 h (total dose ≥ 0.9 g/kg). For immunosuppressed patients, dosage is more conservative (0.2 mL [10 mg]/kg/h) for ≥ 72 h, without an initial bolus (total dose ≥ 0.72 g/kg). Conclusions: Two distinct populations that may benefit most from Pentaglobin therapy are described in this review. However, further clinical evidence is required to strengthen support for the recommendations given here regarding timing, duration and dosage of treatment.
Article
Full-text available
Objective: To report on efficacy and safety of intravenous immunoglobulin (IVIg) therapy in a case series of patients with COVID-19-related encephalopathy. Methods: We retrospectively collected data on all patients with COVID-19 hospitalized at two Italian hospitals who developed encephalopathy during disease course and were treated with IVIg. Results: Five patients (two females, mean age 66.8 years) developed encephalopathy after a mean of 12.6 days, since the onset of respiratory/constitutional symptoms related to COVID-19. Four patients suffered severe respiratory distress, three of which required invasive mechanical ventilation. Neurological manifestations included impaired consciousness, agitation, delirium, pyramidal and extrapyramidal signs. EEG demonstrated diffuse slowing in all patients. Brain MRI showed non-specific findings. CSF analysis revealed normal cell count and protein levels. In all subjects, RT-PCR for SARS-CoV-2 in CSF tested negative. IVIg at 0.4 g/kg/die was commenced 29.8 days (mean, range: 19-55 days) after encephalopathy onset, leading to complete electroclinical recovery in all patients, with an initial improvement of neuropsychiatric symptoms observed in 3.4 days (mean, range: 1-10 days). No adverse events related to IVIg were observed. Conclusions: Our preliminary findings suggest that IVIg may represent a safe and effective treatment for COVID-19-associated encephalopathy. Clinical efficacy may be driven by the anti-inflammatory action of IVIg, associated with its anti-cytokine qualities.
Article
Full-text available
Objectives: To expand the knowledge about the immunological consequences of cladribine (CLAD), a pulsed immune reconstitution therapy approved for active multiple sclerosis (MS), beyond the known short-term effects on peripheral immune cell subsets. Methods: In this study, we characterized depletion and restitution kinetics as well as cytokine profiles of peripheral immune cell subsets in 18 patients with MS following treatment with oral CLAD. The methods involved blood collection prior to CLAD and every three months over a period of 24 months, and extensive characterization of various immune cells subsets by multiparametric flow cytometry. Results: We found a selectivity of CLAD towards central memory T cells and memory B cells and detected a hyper-repopulation of maturing B cells. Counts of classical (-65%) and various nonclassical TH17 cells (-84% to -87%) were markedly reduced 24 months after treatment start, and were comparable with depletion rates of class-switched memory B-cell phenotypes (-87% to -95%). The nadir of TH cells was more pronounced in the second treatment year. We observed a proportional surge of CD20 T-cell subsets and an expansion of regulatory T, B and NK cells. Natural killer T cells (NKT) were only depleted in year two and did not recover. Interpretation: Peripheral immune cell profiling revealed more differentiated insights into the immunological effects of CLAD. While some immune cell subsets expanded, we also observed additive depleting effects after the second treatment course. Further studies are required to elucidate whether these changes are paramount for the consistent and prolonged disease-modifying effect of CLAD.
Article
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) outbreak that started in Wuhan, China, in 2019 resulted in a pandemic not seen for a century, and there is an urgent need to develop safe and efficacious vaccines. The scientific community has made tremendous efforts to understand the disease, and unparalleled efforts are ongoing to develop vaccines and treatments. Toxicologists and pathologists are involved in these efforts to test the efficacy and safety of vaccine candidates. Presently, there are several SARS-CoV-2 vaccines in clinical trials, and the pace of vaccine development has been highly accelerated to meet the urgent need. By 2021, efficacy and safety data from clinical trials are expected, and potentially a vaccine will be available for those most at risk. This review focuses on the ongoing SARS-CoV-2 vaccine development efforts with emphasis on the nonclinical safety assessment and discusses emerging preliminary data from nonclinical and clinical studies. It also provides a brief overview on vaccines for other coronaviruses, since experience gained from these can be useful in the development of SARS-CoV-2 vaccines. This review will also explain why, despite this unprecedented pace of vaccine development, rigorous standards are in place to ensure nonclinical and clinical safety and efficacy. [Box: see text]