ArticlePDF Available

USING AN ANTAGONIST OF CHIMERIC RECEPTOR ANTIGEN T CELL THERAPY TO PREVENT CYTOKINE STORM IN COVID-19: A HYPOTHESIS

Authors:

Abstract

SARS COV2 pandemic is almost 3 years old and remains a case production of pro clearance but also promotes paradoxically hyper unfortunate event leading to organ failure death. With the advent of new variants, the interaction between vaccinated and unvaccinated people and also the interaction between persons with different variants or between the healthy and the asymptomatic; all of these achieved remarkable clinical results CAR T and SARS COV 2 share one thing in common; the cytokine storm treatment associated complication and the latter is an exaggerated host response. Hence, we propose using Immunopharmacology (which targets amongst others, pathologies in which inflammation main component), by using anti objective to help alleviate these lymphocytes as living drugs equipped with a CAR construct directed against TH2 or viral epitopes. We hope to curb the severe complications and the poor outcomes(mortalities) associated with this pathology.
USING AN ANTAGONIST OF CHIMERIC RECEPTOR ANTIGEN T CELL THERAPY TO PREVENT
CYTOKINE STORM IN COVID
Zilefac Brian NGOKWE1,2*
,Tamoh
Stephane1,2, Cheboh Cho Fon1,2
, Kouamou Tchiekou Audrey
Audrey Sandra
1
Department of Oral Surgery, Maxillofacial Surgery and Periodontology, Faculty of Medicine and Biomedical
Sciences,
2
ARTICLE INFO
ABSTRACT
SARS COV2 pandemic is almost 3 years old and remains a case
production of pro
clearance but also promotes paradoxically hyper
unfortunate event leading to organ failure
death. With the advent of new variants, the interaction between vaccinated and unvaccinated people
and also the interaction between persons with different variants or between the healthy and the
asymp
tomatic; all of these
achieved remarkable clinical results
CAR T and SARS COV 2 share one thing in common; the cytokine storm
treatment associated complication and the latter is an exaggerated host response. Hence, we propose
using Immunopharmacology (which targets amongst others, pathologies in which inflammation
main component), by using anti
objective to help alleviate these
lymphocytes as living drugs equipped with a CAR construct directed against TH2 or viral epitopes.
We hope to curb the severe complications and the poor outcomes(mortalities) associated with this
pathology
Copyright©2023, Zilefac Brian NGOKWE et al.
This
unrestricted use, distribution, and reproduction in any
medium,
INTRODUCTION
Coronaviruses in the last two decades have caused serious infection
and mortality in humans (1,2). SARSCoV-2,
is a
similar to two other
coronaviruses causing deadly infections
differences in sequences in the spike protein in SARS
in its enhanced binding to angiotensin-
converting enzyme 2 (ACE2)
in human lung cells (4). This
zoonotic disease, COVID
affected people's physical health, but also destroyed mental health and
floundered economic growth.(5,6)
Globally, as of February
there have been 757 264 511 confirmed cases of COVID
including 6 850 594de
aths, reported to WHO and a total of 13 222
459 780 vaccine doses have been administered
(
has been critical during
this pandemic, both for general guidelines and
for new scientific information; however, there is a fine line between
scie
ntifically accurate information and the brevity and simplicity with
which it should/could be communicated; sometimes trading accuracy
for simplicity may result in a message that is more confusing rather
than clarifying (8).
ISSN
: 0975
-
833X
Article History:
Received 14th January, 2023
Received in revised form
17th February, 2023
Accepted 105th March, 2023
Published online 18th April, 2023
Citation: Zilefac Brian NGOKWE,
Tamoh Stive Fokam, Ntep Ntep David Bienvenue, Nokam Kamdem Stephane, Cheboh Cho Fon
Audrey, Elage Epie Macbrain
Noubissie Audrey Sandra
cell therapy to prevent cytokine storm in COVID-
19: A hypothesis
Key words:
SARS COV2, CAR T, Cytokine Storm,
anti-CAR, TH2, Viral Epitopes, Poor
Outcomes.
*Corresponding Author:
Zilefac Brian NGOKWE
RESEARCH ARTICLE
USING AN ANTAGONIST OF CHIMERIC RECEPTOR ANTIGEN T CELL THERAPY TO PREVENT
CYTOKINE STORM IN COVID
-19: A HYPOTHESIS
,Tamoh
Stive Fokam1, Ntep
Ntep David Bienvenue
, Kouamou Tchiekou Audrey
1,2, Elage
Epie Macbrain
Audrey Sandra
1 and
Tamgnoue Guillaume Arthur
Department of Oral Surgery, Maxillofacial Surgery and Periodontology, Faculty of Medicine and Biomedical
Sciences,
University of Yaoundé I, Cameroon
Post graduate school for Life, Health and Environmental Sciences,
University of Yaoundé I
ABSTRACT
SARS COV2 pandemic is almost 3 years old and remains a case
production of pro
-
inflammatory cytokines in the context of COVID 19, not only impairs viral
clearance but also promotes paradoxically hyper
inflammation including cytokine storm; an
unfortunate event leading to organ failure
following long term damage due to inflammation and even
death. With the advent of new variants, the interaction between vaccinated and unvaccinated people
and also the interaction between persons with different variants or between the healthy and the
tomatic; all of these
possibly leading to the formation of new variants. CAR T therapy
achieved remarkable clinical results
treatment of relapsed/refractory B
CAR T and SARS COV 2 share one thing in common; the cytokine storm
treatment associated complication and the latter is an exaggerated host response. Hence, we propose
using Immunopharmacology (which targets amongst others, pathologies in which inflammation
main component), by using anti
-CAR-
engineered T to target the cytokine storm in COVID 19 with
objective to help alleviate these
symptoms or as a possible solution. Specifically, by using T
lymphocytes as living drugs equipped with a CAR construct directed against TH2 or viral epitopes.
We hope to curb the severe complications and the poor outcomes(mortalities) associated with this
pathology
.
This
is an open access article distributed under the Creative
Commons
medium,
provided the original work is properly cited.
Coronaviruses in the last two decades have caused serious infection
is a
beta-coronavirus,
coronaviruses causing deadly infections
(3) but
differences in sequences in the spike protein in SARS
-CoV-2 results
converting enzyme 2 (ACE2)
zoonotic disease, COVID
-19 not only
affected people's physical health, but also destroyed mental health and
Globally, as of February
21, 2023,
there have been 757 264 511 confirmed cases of COVID
-19,
aths, reported to WHO and a total of 13 222
(
7). Communication
this pandemic, both for general guidelines and
for new scientific information; however, there is a fine line between
ntifically accurate information and the brevity and simplicity with
which it should/could be communicated; sometimes trading accuracy
for simplicity may result in a message that is more confusing rather
Interestingly, there is
the principle of the iceberg in COVID 19, with
symptomatic patients consisting the visible part of the iceberg.
Different countries exhibit varying results and this could be due to
poor diagnostic tests, limited access to diagnostic tests to identify
affec
ted people in different countries and different percentages of
asymptomatic people with COVID 19 in various communities. Hence
the interaction between these groups of people could increase the
creation of new recombinant viruses which could be more harmfu
and more difficult to treat.
This possible interaction has been
demonstrated in a uniquely closed environment
interaction can be seen by the variants of concern (variants that have
been associated with an increase in the transmission or
COVID-
19) with the latest being variant B.1.1.529, commonly known
as Omicron (7,10–12)
. There are reports that vaccinated people made
up 42% of fatalities in January and February during omicron. thats
compared with 23% at the peak of the
testament to this viewpoint in our opinion. However recent data
suggests that people who are vaccinated are 20 times less likely to be
hospitalized (13).
International Journal of Current Research
Vol. 15, Issue, 04, pp.24215-24219, April, 2023
DOI: https://doi.org/10.24941/ijcr.
44965
.04.2023
Tamoh Stive Fokam, Ntep Ntep David Bienvenue, Nokam Kamdem Stephane, Cheboh Cho Fon
Noubissie Audrey Sandra
and Tamgnoue Guillaume Arthur. 2023.
Using an antagonist of Chimeric Receptor Antigen T
19: A hypothesis
.”. International Journal of Current Research, 15, (0
4
Available online at http://www.journalcra.com
z
USING AN ANTAGONIST OF CHIMERIC RECEPTOR ANTIGEN T CELL THERAPY TO PREVENT
Ntep David Bienvenue
1, Nokam Kamdem
Epie Macbrain
1, Noubissie
Tamgnoue Guillaume Arthur
1
Department of Oral Surgery, Maxillofacial Surgery and Periodontology, Faculty of Medicine and Biomedical
University of Yaoundé I
SARS COV2 pandemic is almost 3 years old and remains a case
for concern. An overwhelming
inflammatory cytokines in the context of COVID 19, not only impairs viral
inflammation including cytokine storm; an
following long term damage due to inflammation and even
death. With the advent of new variants, the interaction between vaccinated and unvaccinated people
and also the interaction between persons with different variants or between the healthy and the
possibly leading to the formation of new variants. CAR T therapy
has
treatment of relapsed/refractory B
-cell-derived malignancies.
CAR T and SARS COV 2 share one thing in common; the cytokine storm
even though the former is a
treatment associated complication and the latter is an exaggerated host response. Hence, we propose
using Immunopharmacology (which targets amongst others, pathologies in which inflammation
is the
engineered T to target the cytokine storm in COVID 19 with
symptoms or as a possible solution. Specifically, by using T
lymphocytes as living drugs equipped with a CAR construct directed against TH2 or viral epitopes.
We hope to curb the severe complications and the poor outcomes(mortalities) associated with this
Commons
Attribution License, which permits
the principle of the iceberg in COVID 19, with
symptomatic patients consisting the visible part of the iceberg.
Different countries exhibit varying results and this could be due to
poor diagnostic tests, limited access to diagnostic tests to identify
ted people in different countries and different percentages of
asymptomatic people with COVID 19 in various communities. Hence
the interaction between these groups of people could increase the
creation of new recombinant viruses which could be more harmfu
l
This possible interaction has been
demonstrated in a uniquely closed environment
(9). Also, this
interaction can be seen by the variants of concern (variants that have
been associated with an increase in the transmission or
mortality of
19) with the latest being variant B.1.1.529, commonly known
. There are reports that vaccinated people made
up 42% of fatalities in January and February during omicron. that’s
compared with 23% at the peak of the
delta wave.(13) This could be
testament to this viewpoint in our opinion. However recent data
suggests that people who are vaccinated are 20 times less likely to be
INTERNATIONAL JOURNAL
OF
CURRENT
RESEARCH
Tamoh Stive Fokam, Ntep Ntep David Bienvenue, Nokam Kamdem Stephane, Cheboh Cho Fon
, Kouamou Tchiekou
Using an antagonist of Chimeric Receptor Antigen T
4
), 24215-24219.
We strongly believe that primary prevention would have been the best
option in an ideal situation but we are far from been in one; due to
socio-economic difficulties (which could explain the disparity in
vaccinations between countries) and the enormous human interaction
given that the world is now a global village.
Figure 1. Iceberg principle in COVID 19
SARS-CoV-2 invades the host by virtue of angiotensin-converting
enzyme 2 (ACE2) receptors broadly distributed on various tissues and
immune cells (Yang, 2021; Kumar, 2022).(1,14) The virus can cause
a wide array of clinical manifestations ranging from mild to severe
forms with fatal outcomes. Evidence has demonstrated that
deterioration of COVID 19 may be due immunopathological damage.
Particularly, separate studies have reported that highly elevated levels
of pro-inflammatory cytokines are produced during the crosstalk
between epithelial cells and immune cells in COVID-19, which has
linked the cytokine storm (CS) with the severe complications and
poor outcomes in this infection. Due to their cytotoxic capacity, T
cells emerged as attractive candidates for specific immunotherapy of
cancer. A promising approach is the genetic modification of T cells
with chimeric antigen receptors (CARs) (15). The CAR-mediated
recognition induces cytokine production and tumor-directed
cytotoxicity of T cells (16) . Chimeric antigen receptor (CAR) T cell
therapy represents a paradigm shift in the management of pediatric B-
cell acute lymphoblastic leukemia (ALL) and adult B-cell non-
Hodgkin lymphomas (NHL) (17). There has been enormous progress
in genetic engineering in the last decades paving the way for the
development of CAR T cells.
CARs typically comprise an extracellular antigen recognition moiety
fused via a flexible hinge and transmembrane region to an
intracellular signaling unit, thereby combining the virtues of
antibodies (high antigen-binding specificity) and immune cells (potent
anti-tumor effector mechanisms) within one single fusion molecule
(1). Unfortunately, severe treatment-associated toxicities still restrain
the widespread application of this promising technology. The most
frequent side effects following CAR T-cell administration include
cytokine release syndrome (4–7), Given that organ injury in COVID
19 could be due to direct viral infection and immune overactivation,
we intend to prevent these by using anti-CAR T engineered T cells.
COVID-19 and cytokine storm release: Cytokine storm is the most
life-threatening complication associated with COVID-19,
characterized by a hyperactivated and proinflammatory state of the
immune system(18–20). Highlighting the importance of the immune
system in the physiopathology of this disease (20–22)
Sufficient evidence has revealed the components and characteristics
of CS in the patients with severe COVID-19, which are composed of
an array of cytokines (1). The initiation of COVID-CS induction
during infection and the predominant causative cytokine in COVID-
19 immunopathology remain largely unknown. Despite the lack of
definite pathogen associated molecule pattern (PAMP) of SARS-
CoV-2, in analogy with SARS-CoV and MERS-CoV, it can be
speculated that upon cellular entry of SARS-CoV-2 via its ACE2
receptor, viral genomic single-stranded RNA or other RNA
compositions (double-stranded RNA) as PAMPs can be sensed by the
related pattern recognition receptors (PRRs), in host cells.(1)
However, the protective IFN-I response is quickly and selectively
abrogated by SARS-CoV-2 via different mechanisms. This is
accompanied by an overwhelming production of pro-inflammatory
cytokines in the context of COVID-19, which not only impairs viral
clearance but also promotes paradoxical hyperinflammation including
CS. Therefore, from the immunology perspective, COVID-CS may be
an unfortunate event whereby the intended host immune response
combating the SARS-CoV-2 has lost control and transformed into an
inflammatory type. In SARS-CoV-2 infection, the virus infects the
respiratory epithelial tissue and activates local innate immune cells to
release inflammatory cytokines and other chemokines, which then
recruit more innate immune cells and activate adaptive immune cells
(CD4+ and CD8+ T cells) from the peripheral tissues to produce
sustained inflammatory which induce myelopoiesis and emergency
granulopoiesis that further aggravate lung and epithelial damage. In
addition, overproduction of systemic cytokines gives rise to anemia
through erythro-phagocytosis and macrophage activation as well as
causes perturbation of coagulation and vascular hemostasis, resulting
in capillary leak syndrome, thrombosis, and DIC. These events
together lead to ARDS, multiorgan failure, and death. It is worth
noting that, the host immunoregulatory system is usually capable of
retaining and fine-tuning the protective inflammation to an
appropriate level. Regulatory cells such as Tregs, can produce
regulatory cytokines like IL-10 and tumor growth factor-β to
antagonize overactivated immune responses. However, aggressive
inflammatory conditions such as CS cannot be calmed by the
regulatory system’s natural ability.
COVID-CS is a complicated and dynamic inflammatory process
caused by a group of cytokines from initiation, immune cell
hyperactivation, to organ dysfunction. The development of precise
therapeutic intervention in appropriate time is required to effectively
control COVID-CS. In principle, the treatment strategy is to control
ongoing inflammatory response by specifically or nonspecifically
targeting inflammatory cytokines or related signaling pathways and to
resume the host immunoregulatory system (1). Excessive local release
of cytokines is considered to be the determinant of pathological
alterations and the clinical manifestation of ARDS. Overall, the
primary pathological manifestations in the lung tissue are viral
cytopathic-like changes, infiltration of inflammatory cells, and the
presence of viral particles. Thus, severe lung injury in COVID-19
patients is considered as the result of both direct viral infection and
immune overactivation.(23)
CAR biochemistry and CAR therapy in current medicine:
Clinical research on adoptive transfer of T lymphocytes for infectious
and malignant disorders is quite active. The use of cells transduced
with T cell receptors (TCRs), in which tumor antigen recognition
occurs through presentation on cell surface human leukocyte antigens
(HLA), and the use of chimeric antigen receptors (CARs), which are
typically specified by a single-chain variable region domain of an
antibody and directed to a cell surface tumor-associated antigen, are
fundamental approaches to live T cell immune therapy (24). Chimeric
antigen receptor T cells (CAR T) are a promising type of
immunotherapy that uses genetically modified T cells to target cancer
cells. Several CAR T cell products have recently demonstrated
impressive efficacy against previously difficult-to-treat cancers.
Additionally, second-generation CARs include a CD28 or 4-1BB co-
stimulatory endodomain (25). Over the last few decades, there has
been tremendous progress in genetic engineering, paving the way for
the development of CAR T cells. CARs typically consist of an
extracellular antigen recognition moiety fused to an intracellular
signaling unit via a flexible hinge and transmembrane region,
combining the benefits of antibodies (high antigen-binding
specificity) and immune cells (potent anti-tumor effector
mechanisms) within a single fusion molecule (1). Immunotherapies
24216 Zilefac Brian NGOKWE et al. Using an antagonist of Chimeric receptor antigen t cell therapy to prevent cytokine storm in covid-19: A hypothesis
have showed promise, but they are far from flawless as evidenced
with certain serious treatment-associated toxicities. Unlike
conventional cytotoxic chemotherapy or small molecule inhibitors,
immunotherapies have specific toxicities that might be fatal, such as
cytokine release syndrome (CRS) and CAR-related encephalopathy
syndrome (CRES).(25)with cytokine release syndrome being the most
common side event after CAR T-cell administration (4–7). Several
strategies have been tested to counteract these effects, from non-
specific immunosuppression to selective CAR-engineered T cell
ablation. The latter technique is currently under huge research and is
based on the transgenic introduction of either suicide genes or
elimination marker genes. Following the introduction of these
therapeutic agents, selective CAR T-cell depletion is achieved
through complement-dependent cytotoxicity (CDC) and antibody-
dependent cellular cytotoxicity (ADCC). However, each method has
inherent limits that could prevent it from having a wide range of
therapeutic applications. These procedures also have serious
toxicities.
These include immunogenicity (21) the proposed depletion marker's
enormous size (more than 130 amino acids), the reliance on the
patients' immune systems (ADCC, CDC), and the occurrence of on-
target adverse effects brought on by mAb (monoclonal antibodies)
recognizing healthy tissue (22). The term CRS (cytokine release
syndrome) here refers to the excessive release of cytokines (IL-1, IL-
6, IFN-, and IL-10) by CAR-modified immune cells or bystander
innate immune cells (macrophages, monocytes, dendritic cells, and
other immune cells). Excess cytokines can result in vascular leakage,
respiratory failure, coagulopathy, and multi-organ system
dysfunction. The production of a significant amount of interferon
gamma (IFN-) and/or tumor necrosis factor alpha (TNF-) by CAR-T
activated cells, which in turn activates macrophages, dendritic cells,
and endothelial cells, is one possible cause causing CRS. These cells
can release more proinflammatory cytokines after being stimulated by
IFN- or TNF (8)
The perspective of CAR-T antagonist: This genetically modified T
cells that constitute the CAR T cells can induce a cytokine storm.
Control and reversal of toxicity have emerged as crucial components
of CAR T-cell treatment due to the extremely long-term survival and
proliferation capacity of genetically altered T cells (26). Bachmann et
al. suggested integrating a particular peptide epitope (E7B6) into the
CAR architecture that can be utilized as an intrinsic elimination tag as
a way to address this. This E-tag into the CAR's extracellular spacer
region should stop CAR T-cell escape, reducing the therapy's
cytotoxic effects while also increasing its safety (26). Therefore, for
selective and rigorous CAR T-cell elimination, Bachmann et al used a
short peptide epitope (E-tag) directly inserted into the CAR
architecture (26–28). Additionally, Bachmann et al sought to avoid
reliance on pharmacological drugs whose therapeutic effect invariably
declines due to their short half-life, which is a significant drawback,
by using T lymphocytes as living drugs that are equipped with a CAR
construct directed against a targetable portion of the therapeutic
CAR.(26)
With the other experiments, the cloning and structural characteristics
of conventional CARs as well as the universal chimeric antigen
receptor (UniCAR) 28/ζ construct with the incorporation of the
peptide epitope E7B6 (E-tag) as a targetable moiety in the
extracellular spacer region are described in detail (26,29–31). In the
hinge region of CARs, a peptide tag called E7B6 is recognized by the
αE-tag CAR construct. When an antigen is recognized, these αE-tag
CAR effector cells cross-link with target cells, which should cause the
elimination of the latter. Furthermore, Bachman et al demonstrated
that the cytotoxic effect was caused by the incorporated peptide's
specific recognition and binding. (αE-tag CAR). The anti-CAR
therapy has been demonstrated to prevent the cytotoxic effects due to
CAR T therapy and has been described in detail by Bachmann et
al.(26). Our hypothesis could act by targeting impaired viral clearance
and/or lymphocytes
Against viral infections, the adaptive immune system, particularly T
cells, plays an important role (32). T cell responses can be divided
into two types: effector cytotoxic cells (CTLs) and T helper cells (Th)
(20,33,34). Data suggest that cytokine storms may contribute to the
pathophysiology of severe COVID-19 disease together with reduced
Th1 antiviral adaptive responses.(35). Adults with COVID-19
experience acute peripheral T cell depletion, the degree of which is
positively correlated with the severity of the disease, whereas
asymptomatic patients and children typically retain peripheral T cell
numbers (36). The progression of the disease in COVID-19 has been
associated with the Th1 / Th2 balance. An appropriate Th1 immune
response, once a viral infection has been recognized, can eradicate it.
The cytokine storm that results from an overactive immune response
and increased cytokine production, however, leads to a Th2-type
response and a poor prognosis for COVID-19(36–38). In the same
light, research has demonstrated that elevated IL-15 levels and a
strong Th2 response are linked to the disease's deadly prognosis and
that senescent Th2 cell percentage was an independent risk factor for
death(20)
Also, COVID-19 patients had significantly fewer Th1 and more
highly activated Th2 cells than the reference population (20) . Th1
coordinated immune response during SARS-CoV-2 infection has been
associated with a good prognosis and resolution of COVID-19. The
type of Th response influenced disease outcome, as 78% of patients
who died had an over reactive Th2 response (20,39,40). Because their
cytokines stimulate antibody production, Th2 hyper activation could
explain why severe patients have higher antibody titers than mild or
asymptomatic patients (20,39,40). Moreover, within the cell, M.
Bacillus Calmette-Guerin (BCG) vaccination stimulates a Th1
response, guarding against the development of tuberculosis in
humans. With incredibly unexpected results, the effectiveness of the
BCG vaccine has been researched in COVID-19 patients. Patients
who received the vaccine and developed Th1 responses had lower
death and infection rates(41). A recent research, has emphasized the
significance of Th1 hypoactivation and Th2 overreaction, followed by
exhaustion, as they are related to a worse prognosis (20).
Hence by directing genetically modified anti-CAR T cells against
these lymphocytes (Th2) we hypothesize that this could limit or
prevent the cytokine storm. Also, could we direct these genetically
modified anti-CAR T cells against the viral epitopes to help limit or
prevent this cytokine storm. The SARS-CoV-2 virus is a single-
stranded enveloped positive-sense RNA virus that belongs to the β-
coronavirus (42,43). The proteins encoded by the genome of SARS-
CoV-2 are comprised of structural (SPs) and nonstructural proteins
(NSPs), as well as accessory proteins(44,45) . SPs mainly include
spike (S), membrane (M), envelope (E), and nucleocapsid (N)
proteins (42,46). The virus receptor-binding domain (RBD) contains
several antigenic epitopes. Those antigenic epitopes, also known as
antigenic determinants, are the binding sites of host antibodies. The
antigenic epitope plays an important role in activating the host CD4
and CD8 T cell immune response (47). Therefore, the S protein, the
RBD domain, and antigenic epitopes pave the road for the therapeutic
strategies (42). So by targeting these viral epitopes especially of the S
protein, we might to control the interactions and underlying
mechanisms between the host and virus infection. More so, research
has shown that SARS-CoV-2 uses the structural function of the S
protein to weaken or escape host immune surveillance (42). Hence by
creating a genetically modified CAR T antagonist, could help curb the
cytokine storm, given that thus aggressive inflammatory condition
(the cytokine storm) cannot be calmed by the regulatory system’s
natural ability and also given the non-precise nature of general
immunosuppression. We propose an antagonist for CAR-T (Chimeric
Receptor Antigen T cell) therapy as a solution to prevent cytokine
storm in patients with COVID-19 and hence reduce the morbidity and
mortality due to COVID 19.
CONCLUSION
The SARS-CoV-2 virus can cause a wide range of manifestations
from mild to severe/critical forms with fatal outcomes; fatal outcomes
with increased morbidities and mortalities which we aim to prevent.
24217 International Journal of Current Research, Vol. 15, Issue, 04, pp.
24215-24219, April, 2023
Cytokine storm during the COVID 19 infection is
This unfortunate event causes deaths due to ARDS and multi
failure CAR-
T therapies has as one of its effects inducing a cytokine
storm. So, by using immunopharmacology more precisely, a
genetically modified antagonist of this t
herapy; we intend to prevent
or curb the adverse effects caused by this cytokine storm which can
be helpful in severe SARS-CoV-2
to reduce the mortalities and
morbidities associated with this pandemic.
Figure 2.
Target; CAR T cells
Anti-CAR targets
selectively and precisely eliminates CAR T
with the help of its short peptide epitope.
Figure 3.
Target; Th2 lymphocytes or viral epitopes
We propose using a genetically modified anti-
CAR T cells against
Th2lymphocytes to prevent cytokine storm
or viral epitopes
Acknowledgements
: Dr Ronit Sionov (Hebrew University of
Jerusalem)
REFERENCES
1.
Yang L, Xie X, Tu Z, Fu J, Xu D, Zhou Y. The signal pathways
and treatment of cytokine storm in COVID
Transduct Target Ther (Internet)
. Springer US; 2021;6:1
Available from:
http://dx.doi.org/10.1038/s41392
2.
Patil AM, Göthert JR, Khairnar V. Emergence, Transmission,
and Potential Therapeutic Targets for the COVID
Associated with the SARS-CoV-
2. Cellular Physiology and
Biochemistry. 2020;54:767–90.
3.
Soy M, Tabak F, Kayhan S. Cytokine storm in COVID
pathogenesis and overview of anti-
inflammatory agents used in
treatment. 2020;
4. Asrani P, Tiwari K, Eap
en MS, McAlinden KD, Haug G,
Johansen MD, et al. Clinical features and mechanistic insights
into drug repurposing for combating COVID
Cell Biol. Elsevier Ltd; 2021;142:106114.
5.
Sarıışık M, Usta S. Global effect of COVID
t
he Hospitality and Tourism Industry: A Research Companion.
2021;41–59.
6.
Belitski M, Guenther C, Kritikos AS, Thurik R. Economic
effects of the COVID-
19 pandemic on entrepreneurship and
small businesses. Small Business Economics. Springer US;
2022;58:593–609.
7. Coronavirus disease (COVID-19) (
Internet
26). Available from:
https://www.who.int/
diseases/novel-coronavirus-2019
24218 Zilefac Brian NGOKWE et al.
Using an antagonist of Chimeric receptor antigen t cell therapy to prevent cytokine storm in covid
Cytokine storm during the COVID 19 infection is
fatal to the patients.
This unfortunate event causes deaths due to ARDS and multi
-organ
T therapies has as one of its effects inducing a cytokine
storm. So, by using immunopharmacology more precisely, a
herapy; we intend to prevent
or curb the adverse effects caused by this cytokine storm which can
to reduce the mortalities and
Target; CAR T cells
selectively and precisely eliminates CAR T
-cell
Target; Th2 lymphocytes or viral epitopes
CAR T cells against
or viral epitopes
.
: Dr Ronit Sionov (Hebrew University of
Yang L, Xie X, Tu Z, Fu J, Xu D, Zhou Y. The signal pathways
and treatment of cytokine storm in COVID
-19. Signal
. Springer US; 2021;6:1
–20.
http://dx.doi.org/10.1038/s41392
-021-00679-0
Patil AM, Göthert JR, Khairnar V. Emergence, Transmission,
and Potential Therapeutic Targets for the COVID
-19 Pandemic
2. Cellular Physiology and
Soy M, Tabak F, Kayhan S. Cytokine storm in COVID
-19 :
inflammatory agents used in
en MS, McAlinden KD, Haug G,
Johansen MD, et al. Clinical features and mechanistic insights
into drug repurposing for combating COVID
-19. Int J Biochem
Sarıışık M, Usta S. Global effect of COVID
-19. COVID-19 and
he Hospitality and Tourism Industry: A Research Companion.
Belitski M, Guenther C, Kritikos AS, Thurik R. Economic
19 pandemic on entrepreneurship and
small businesses. Small Business Economics. Springer US;
Internet
). (cited 2023Feb
https://www.who.int/
emergencies/
8.
Centers US, Control D. Nailin
2022;377.
9.
Abe H, Ushijima Y, Amano M, Sakurai Y, Yoshikawa R,
Kinoshita T, et al.
Unique Evolution of SARS
Second Large Cruise Ship Cluster in Japan. Microorganisms.
2022;10:1–16.
10.
Augusto G, Mohsen MO, Zinkhan S, Liu X, Vogel M,
Bachmann MF. In vitro data suggest that Indian delta variant
B.1.617 of SARS-CoV-
2 escapes neutralization by both
receptor affinity and immune evasion. Allergy: European
Journal of Allergy and Clinical Immun
Sons Inc; 2022;77:111–7.
11.
Akkız H. The Biological Functions and Clinical Significance of
SARS-CoV-
2 Variants of Corcern. Front Med (Lausanne)
(Internet)
. Frontiers Media SA; 2022
26)
;9:849217. Available from:
http://www.ncbi.nlm.nih.gov/pubmed/35669924
12.
Emerging Variants of SARS
Against Coronavirus (COVID
2022 Jul 26
https://pubmed.ncbi.nlm.nih.gov/34033342/
13.
Where We Are in the Pandemic | Johns Hopkins | Bloomberg
School of Public Health
Available from:
https://publichealth.jhu.edu/2022/where
are-in-the-pandemic
14.
Kumar S, Thambiraja TS, Karuppanan K, Subramaniam G.
Omicron and Delta variant of SARS
computatio
nal study of spike protein. J Med Virol.
2022;94:1641–9.
15.
Bachmann M, Cartellieri M, Feldmann A, Bippes C, Stamova S,
Wehner R, et al. Chimeric antigen receptor
for immunotherapy of cancer. J Biomed Biotechnol. 2010;2010.
16. Bachmann M, Ca
rtellieri M, Feldmann A, Bippes C, Stamova S,
Wehner R, et al. Chimeric antigen receptor
for immunotherapy of cancer. J Biomed Biotechnol. 2010;2010.
17.
Bachanova V, Bishop MR, Dahi P, Dholaria B, Grupp SA,
Hayes-
lattin B, et al. Biology o
Transplantation Chimeric Antigen Receptor T Cell Therapy
During the COVID-
19 Pandemic. Biology of Blood and
Marrow Transplantation
2020;26:1239–
46. Available from:
https://doi.org/10.1016/j.bbmt.2020.04.008
18.
Buszko M, Park JH, Verthelyi D, Sen R, Young HA, Rosenberg
AS. The dynamic changes in cytokine responses in COVID
a snapshot of the current state of knowledge. Nat Immunol.
Nature Research; 2020;21:
1146
19.
Schett G, Sticherling M, Neurath MF. COVID
cytokine targeting in chronic inflammatory diseases? Nat Rev
Immunol. Nature Research; 2020. p. 271
20. Gil-Etayo FJ, Suàrez-
Fernández P, Cabrera
D, Garcinuño S, Naranjo L,
Response Is a Determining Factor in COVID
Front Cell Infect Microbiol. Frontiers Media S.A.; 2021;11.
21.
Chen Z, John Wherry E. T cell responses in patients with
COVID-
19. Nat Rev Immunol. Nature Research; 2020;20
36.
22.
Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, et al.
Lymphopenia predicts disease severity of COVID
descriptive and predictive study. Signal Transduct Target Ther.
Springer Nature; 2020.
23.
Hu B, Huang S, Yin L. The cytokine storm and COV
Med Virol (Internet)
. John Wiley & Sons, Ltd; 2:0
from:
http://dx.doi.org/10.1002/jmv.26232
24.
Krebs S, Dacek MM, Carter LM, Scheinberg DA, Larson SM.
CAR Chase: Where Do Engineered Cells
Oncol. 2020;10:1–7.
25.
Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment
of chimeric antigen receptor T cell
syndrome. Expert Rev Clin Immunol. 2019;15:813
26. Koristka S, Ziller-
Walter P, Bergmann R
A, Kegler A, et al. Anti-
CAR
based elimination of autologous CAR T cells. Cancer
Using an antagonist of Chimeric receptor antigen t cell therapy to prevent cytokine storm in covid
Centers US, Control D. Nailin
g the nuance on COVID-19.
Abe H, Ushijima Y, Amano M, Sakurai Y, Yoshikawa R,
Unique Evolution of SARS
-CoV-2 in the
Second Large Cruise Ship Cluster in Japan. Microorganisms.
Augusto G, Mohsen MO, Zinkhan S, Liu X, Vogel M,
Bachmann MF. In vitro data suggest that Indian delta variant
2 escapes neutralization by both
receptor affinity and immune evasion. Allergy: European
Journal of Allergy and Clinical Immun
ology. John Wiley and
Akkız H. The Biological Functions and Clinical Significance of
2 Variants of Corcern. Front Med (Lausanne)
. Frontiers Media SA; 2022
(cited 2022 Jul
;9:849217. Available from:
http://www.ncbi.nlm.nih.gov/pubmed/35669924
Emerging Variants of SARS
-CoV-2 And Novel Therapeutics
Against Coronavirus (COVID
-19) - PubMed (Internet). (cited
2022 Jul 26
). Available from:
https://pubmed.ncbi.nlm.nih.gov/34033342/
Where We Are in the Pandemic | Johns Hopkins | Bloomberg
School of Public Health
(Internet). (cited 2022 Jul 26).
https://publichealth.jhu.edu/2022/where
-we-
Kumar S, Thambiraja TS, Karuppanan K, Subramaniam G.
Omicron and Delta variant of SARS
-CoV-2: A comparative
nal study of spike protein. J Med Virol.
Bachmann M, Cartellieri M, Feldmann A, Bippes C, Stamova S,
Wehner R, et al. Chimeric antigen receptor
-engineered T cells
for immunotherapy of cancer. J Biomed Biotechnol. 2010;2010.
rtellieri M, Feldmann A, Bippes C, Stamova S,
Wehner R, et al. Chimeric antigen receptor
-engineered T cells
for immunotherapy of cancer. J Biomed Biotechnol. 2010;2010.
Bachanova V, Bishop MR, Dahi P, Dholaria B, Grupp SA,
lattin B, et al. Biology o
f Blood and Marrow
Transplantation Chimeric Antigen Receptor T Cell Therapy
19 Pandemic. Biology of Blood and
Marrow Transplantation
(Internet). Elsevier Inc.;
46. Available from:
https://doi.org/10.1016/j.bbmt.2020.04.008
Buszko M, Park JH, Verthelyi D, Sen R, Young HA, Rosenberg
AS. The dynamic changes in cytokine responses in COVID
-19:
a snapshot of the current state of knowledge. Nat Immunol.
1146
–51.
Schett G, Sticherling M, Neurath MF. COVID
-19: risk for
cytokine targeting in chronic inflammatory diseases? Nat Rev
Immunol. Nature Research; 2020. p. 271
–2.
Fernández P, Cabrera
-Marante O, Arroyo
D, Garcinuño S, Naranjo L,
et al. T-Helper Cell Subset
Response Is a Determining Factor in COVID
-19 Progression.
Front Cell Infect Microbiol. Frontiers Media S.A.; 2021;11.
Chen Z, John Wherry E. T cell responses in patients with
19. Nat Rev Immunol. Nature Research; 2020;20
:529–
Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, et al.
Lymphopenia predicts disease severity of COVID
-19: a
descriptive and predictive study. Signal Transduct Target Ther.
Hu B, Huang S, Yin L. The cytokine storm and COV
ID-19. J
. John Wiley & Sons, Ltd; 2:0
–2. Available
http://dx.doi.org/10.1002/jmv.26232
Krebs S, Dacek MM, Carter LM, Scheinberg DA, Larson SM.
CAR Chase: Where Do Engineered Cells
Go in Humans? Front
Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment
of chimeric antigen receptor T cell
-induced cytokine release
syndrome. Expert Rev Clin Immunol. 2019;15:813
–22.
Walter P, Bergmann R
, Arndt C, Feldmann
CAR
-engineered T cells for epitope-
based elimination of autologous CAR T cells. Cancer
Using an antagonist of Chimeric receptor antigen t cell therapy to prevent cytokine storm in covid
-19: A hypothesis
Immunology, Immunotherapy. Springer Berlin Heidelberg;
2019;68:1401–15.
27. Cartellieri M, Koristka S, Arndt C, Feldmann A, Stamova S,
Von Bonin M, et al. A novel Ex Vivo isolation and expansion
procedure for chimeric antigen receptor engrafted human T
cells. PLoS One. 2014;9:1–12.
28. Cartellieri M, Feldmann A, Koristka S, Arndt C, Loff S,
Ehninger A, et al. Switching CAR T cells on and off: a novel
modular platform for retargeting of T cells to AML blasts.
Blood Cancer J. Nature Publishing Group; 2016;6:e458.
29. Feldmann A, Arndt C, Bergmann R, Loff S, Cartellieri M,
Bachmann D, et al. Retargeting of T lymphocytes to PSCA- or
PSMA positive prostate cancer cells using the novel modular
chimeric antigen receptor platform technology “UniCAR.”
Oncotarget. 2017;8:31368–85.
30. Cartellieri M, Feldmann A, Koristka S, Arndt C, Loff S,
Ehninger A, et al. Switching CAR T cells on and off: a novel
modular platform for retargeting of T cells to AML blasts.
Blood Cancer J. Nature Publishing Group; 2016;6:e458.
31. Cartellieri M, Koristka S, Arndt C, Feldmann A, Stamova S,
von Bonin M, et al. A novel Ex Vivo isolation and expansion
procedure for chimeric antigen receptor engrafted human T
cells. PLoS One. 2014;9:1–12.
32. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al.
Reduction and Functional Exhaustion of T Cells in Patients
With Coronavirus Disease 2019 (COVID-19). Front Immunol.
Frontiers Media S.A.; 2020;11.
33. Mescher MF, Curtsinger JM, Agarwal P, Casey KA, Gerner M,
Hammerbeck CD, et al. Signals required for programming
effector and memory development by CD8 + T cells.
34. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4+ T
cell populations. Annu Rev Immunol. 2010. p. 445–89.
35. Kuppalli K, Rasmussen AL. A glimpse into the eye of the
COVID-19 cytokine storm. EBioMedicine. Elsevier B.V.;
2020;55:4–5.
36. Aleebrahim-Dehkordi E, Molavi B, Mokhtari M, Deravi N,
Fathi M, Fazel T, et al. T helper type (Th1/Th2) responses to
SARS-CoV-2 and influenza A (H1N1) virus: From cytokines
produced to immune responses. Transpl Immunol (Internet).
Elsevier B.V.; 2022;70:101495. Available from:
https://doi.org/10.1016/j.trim.2021.101495
37. Roncati L, Nasillo V, Lusenti B, Riva G. Signals of Th2
immune response from COVID-19 patients requiring intensive
care. Ann Hematol. Annals of Hematology; 2020. p. 1419–20.
38. Neidleman J, Luo X, Frouard J, Xie G, Gill G, Stein ES, et al.
SARS-CoV-2-Specific T Cells Exhibit Phenotypic Features of
Helper Function, Lack of Terminal Differentiation, and High
Proliferation Potential. Cell Rep Med. ElsevierCompany.;
2020;1:100081.
39. Spellberg B, Edwards JE. Type 1/Type 2 Immunity in
Infectious Diseases (Internet). Available from:
https://academic.oup.com/cid/article/32/1/76/311106
40. Infante-Duarto C, Kamradt T. Springer Seminars in
Immunopathology Thl/Th2 balance in infection. Springer
SeminImmunopathol. 1999.
41. Ozdemir C, Kucuksezer UC, Tamay ZU. Is BCG vaccination
affecting the spread and severity of COVID-19? Allergy
(Internet). John Wiley & Sons, Ltd; 2020 (cited 2022 Sep
8);75:1824–7. Available from: https://onlinelibrary.wiley. com/
doi/full/10.1111/all.14344
42. Zhang C, Yang M. Newly Emerged Antiviral Strategies for
SARS-CoV-2: From Deciphering Viral Protein Structural
Function to the Development of Vaccines, Antibodies, and
Small Molecules. Int J Mol Sci. 2022;23:6083.
43. Machhi J, Herskovitz J, Senan AM, Dutta D, Nath B, Oleynikov
MD, et al. The Natural History, Pathobiology, and Clinical
Manifestations of SARS-CoV-2 Infections. Journal of
Neuroimmune Pharmacology. Journal of Neuroimmune
Pharmacology; 2020;15:359–86.
44. Yadav R, Chaudhary JK, Jain N, Chaudhary PK. Role of
Structural and Non-Structural Proteins and Therapeutic.
2021;1–16.
45. Malik YA. Properties of coronavirus and SARS-CoV-2.
Malaysian Journal of Pathology. 2020;42:3–11.
46. Wu C, Liu Y, Yang Y, Zhang P, Zhong W, Wang Y, et al.
Analysis of therapeutic targets for SARS-CoV-2 and discovery
of potential drugs by computational methods. Acta Pharm Sin
B. Elsevier Ltd; 2020;10:766–88.
47. Dakal TC. Antigenic sites in SARS-CoV-2 spike RBD show
molecular similarity with pathogenic antigenic determinants and
harbors peptides for vaccine development. Immunobiology.
Elsevier GmbH; 2021;226:152091.
*******
24219 International Journal of Current Research, Vol. 15, Issue, 04, pp.
24215-24219, April, 2023
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Coronavirus disease 2019 (COVID-19) caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the most severe health crisis, causing extraordinary economic disruption worldwide. SARS-CoV-2 is a single-stranded RNA-enveloped virus. The process of viral replication and particle packaging is finished in host cells. Viral proteins, including both structural and nonstructural proteins, play important roles in the viral life cycle, which also provides the targets of treatment. Therefore, a better understanding of the structural function of virus proteins is crucial to speed up the development of vaccines and therapeutic strategies. Currently, the structure and function of proteins encoded by the SARS-CoV-2 genome are reviewed by several studies. However, most of them are based on the analysis of SARS-CoV-1 particles, lacking a systematic review update for SARS-CoV-2. Here, we specifically focus on the structure and function of proteins encoded by SARS-CoV-2. Viral proteins that contribute to COVID-19 infection and disease pathogenesis are reviewed according to the most recent research findings. The structure-function correlation of viral proteins provides a fundamental rationale for vaccine development and targeted therapy. Then, current antiviral vaccines are updated, such as inactive viral vaccines and protein-based vaccines and DNA, mRNA, and circular RNA vaccines. A summary of other therapeutic options is also reviewed, including monoclonal antibodies such as a cross-neutralizer antibody, a constructed cobinding antibody, a dual functional monoclonal antibody, an antibody cocktail, and an engineered bispecific antibody, as well as peptide-based inhibitors, chemical compounds, and clustered regularly interspaced short palindromic repeats (CRISPR) exploration. Overall, viral proteins and their functions provide the basis for targeted therapy and vaccine development.
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuing to evolve, emerging novel variants with spike protein mutations. Although most mutations emerged in the SARS-CoV-2 genome are neutral or mildly deleterious, a small number of mutations can affect virus phenotype that confers the virus a fitness advantage. These mutations can enhance viral replication, raise the risk of reinfection and blunt the potency of neutralizing antibodies triggered by previous infection and vaccination. Since December 2020, the SARS-CoV-2 has emerged five quickly spreading strains, designated variants of concern (VOCs), including the Alpha (B.1.1.7) variant, the Beta (B.1.351) variant, the Gamma (P.1) variant, the Delta (B.1.617.2) variant and the Omicron (B.1.1.529) variant. These variants have a high number of the mutations in the spike protein that promotes viral cell entry through the angiotensin-converting enzyme -2 (ACE2). Mutations that have arisen in the receptor binding domain (RBD) of the spike protein are of great concern due to their potential to evade neutralizing antibodies triggered by previous infection and vaccines. The Alpha variant emerged in the United Kingdom in the second half of 2020 that has spread quickly globally and acquired the E484K mutation in the United Kingdom and the United States. The Beta and Gamma variants emerged in South Africa and Brazil, respectively, that have additional mutations at positions E484 and K417 in the RBD. SARS-CoV-2 variants containing the combination of N501Y, E484K, and K417N/T mutations exhibit remarkably decreased sensitivity to neutralizing antibodies mediated by vaccination or previous infection. The Gamma variant may result in more severe disease than other variants do even in convalescent individuals. The Delta variant emerged in India in December 2020 and has spread to many countries including the United States and the United Kingdom. The Delta variant has 8 mutations in the spike protein, some of which can influence immune responses to the key antigenic regions of RBD. In early November 2021, the Omicron (B.1.1.529) variant was first detected in Botswana and South Africa. The Omicron variant harbors more than 30 mutations in the spike protein, many of which are located within the RBD, which have been associated with increased transmissibility and immune evasion after previous infection and vaccination. Additionally, the Omicron variant contains 3 deletions and one insertion in the spike protein. Recently, the Omicron variant has been classified into three sublineages, including BA.1, BA.2, and BA.3, with strikingly different genetic characteristics. The Omicron BA.2 sublineage has different virological landscapes, such as transmissibility, pathogenicity and resistance to the vaccine-induced immunity compared to BA.1 and BA.3 sublineages. Mutations emerged in the RBD of the spike protein of VOCs increase viral replication, making the virus more infectious and more transmissible and enable the virus to evade vaccine-elicited neutralizing antibodies. Unfortunately, the emergence of novel SARS-CoV-2 VOCs has tempered early optimism regarding the efficacy of COVID-19 vaccines. This review addresses the biological and clinical significance of SARS-CoV-2 VOCs and their impact on neutralizing antibodies mediated by existing COVID-19 vaccines.
Article
Full-text available
In the initial phase of the novel coronavirus disease (COVID-19) pandemic, a large-scale cluster on the cruise ship Diamond Princess (DP) emerged in Japan. Genetic analysis of the DP strains has provided important information for elucidating the possible transmission process of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on a cruise ship. However, genome-based analyses of SARS-CoV-2 detected in large-scale cruise ship clusters other than the DP cluster have rarely been reported. In the present study, whole-genome sequences of 94 SARS-CoV-2 strains detected in the second large cruise ship cluster, which emerged on the Costa Atlantica (CA) in Japan, were characterized to understand the evolution of the virus in a crowded and confined place. Phylogenetic and haplotype network analysis indicated that the CA strains were derived from a common ancestral strain introduced on the CA cruise ship and spread in a superspreading event-like manner, resulting in several mutations that might have affected viral characteristics, including the P681H substitution in the spike protein. Moreover, there were significant genetic distances between CA strains and other strains isolated in different environments, such as cities under lockdown. These results provide new insights into the unique evolution patterns of SARS-CoV-2 in the CA cruise ship cluster.
Article
Full-text available
Cytokines produced by T helper cells (Th cells) have essential roles in the body's defense against viruses. Type 1 T helper (Th1) cells are essential for the host defense toward intracellular pathogens while T helper type 2 (Th2) cells are considered to be critical for the helminthic parasites' elimination swine-origin influenza A (H1N1) virus, a disease led to an epidemic in 2009 and rapidly spread globally via human-to-human transmission. Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a global pandemic in 2020 and is a serious threat to the public health. Pulmonary immunopathology is the leading cause of death during influenza and SARS-CoV-2 epidemics and pandemics. Influenza and SARS-CoV-2 cause high levels of cytokines in the lung. Both inadequate levels and high levels of specific cytokines can have side effects. In this literature review article, we want to compare the Th1 and Th2 cells responses in SARS-CoV-2 and H1N1.
Article
Full-text available
Plain English Summary Responding to COVID-19 involves not just shielding small business jobs, supporting entrepreneurship, and raising government debt but also creating productive entrepreneurship and resilient location-specific entrepreneurial ecosystems. The COVID-19 pandemic is an unprecedented challenge for small businesses that also brings new market opportunities. The papers in this special issue of Small Business Economics Journal aim to shed light on the economic effects of the COVID-19 pandemic by looking at the macro- and microeconomic effects on entrepreneurship and small businesses as well as the role of financial support policies and well-being in both developed and developing countries. Future research should focus on the role of digitization and financial mechanisms supporting small businesses during crises.
Article
Full-text available
Background Emerged mutations can be attributed to increased transmissibility of the B.1.617 and B.1.36 Indian delta variants of SARS‐CoV‐2, most notably substitutions L452R/E484Q and N440K, respectively, which occur in the receptor‐binding domain (RBD) of the Spike (S) fusion glycoprotein. Objective We aimed to assess the effects of mutations L452R/E484Q and N440K (as well as the previously studied mutation E484K present in variants B.1.351 and P.1) on the affinity of RBD for ACE2, SARS‐CoV‐2 main receptor. We also aimed to assess the ability of antibodies induced by natural infection or by immunization with BNT162b2 mRNA vaccine to recognize the mutated versions of the RBD, as well as blocking the interaction RBD‐ACE2, an important surrogate readout for virus neutralization. Methods To this end, we produced recombinant wild‐type RBD, as well as RBD containing each of the mutations L452R/E484Q, N440K, or E484K (the latest present in variants of concern B.1.351 and P.1), as well as the ectodomain of ACE2. Using Biolayer Interferometry (BLI), we measured the binding affinity of RBD for ACE2 and the ability of sera from COVID‐19 convalescent donors or subjects immunized with BNT162b2 mRNA vaccine to block this interaction. Finally, we correlated these results with total anti‐RBD IgG titers measured from the same sera by direct ELISA. Results The binding assays showed L452R/E484Q double‐mutant RBD to interact with ACE2 with higher affinity (KD = 4.6 nM) than wild‐type (KD = 21.3 nM) or single mutants N440K (KD = 9.9 nM) and E484K (KD = 19.7 nM) RBDs. Meanwhile, the anti‐RBD IgG titration resulted in lower recognition of mutants E484K and L452R/E484Q by infection‐induced antibodies, whereas only mutant E484K was recognized less by antibodies induced by vaccination. More interestingly, sera from convalescent as well as immunized subjects showed reduced ability to block the interaction between ACE2 and RBD mutants E484K and L452R/E484Q, as shown by the inhibition assays. Conclusion Our data suggest that the newly emerged SARS‐CoV‐2 variant B.1.617, as well as the better‐studied variants B.1.351 and P.1 (all containing a mutation at position E484) display increased transmissibility both due to their higher affinity for the cell receptor ACE2 and their ability to partially bypass immunity generated against the wild‐type virus. For variant B.1.36 (with a point mutation at position N440), only increased affinity seems to play a role.
Article
Full-text available
The Coronavirus Disease 2019 (COVID-19) pandemic has become a global crisis and is more devastating than any other previous infectious disease. It has affected a significant proportion of the global population both physically and mentally, and destroyed businesses and societies. Current evidence suggested that immunopathology may be responsible for COVID-19 pathogenesis, including lymphopenia, neutrophilia, dysregulation of monocytes and macrophages, reduced or delayed type I interferon (IFN-I) response, antibody-dependent enhancement, and especially, cytokine storm (CS). The CS is characterized by hyperproduction of an array of pro-inflammatory cytokines and is closely associated with poor prognosis. These excessively secreted pro-inflammatory cytokines initiate different inflammatory signaling pathways via their receptors on immune and tissue cells, resulting in complicated medical symptoms including fever, capillary leak syndrome, disseminated intravascular coagulation, acute respiratory distress syndrome, and multiorgan failure, ultimately leading to death in the most severe cases. Therefore, it is clinically important to understand the initiation and signaling pathways of CS to develop more effective treatment strategies for COVID-19. Herein, we discuss the latest developments in the immunopathological characteristics of COVID-19 and focus on CS including the current research status of the different cytokines involved. We also discuss the induction, function, downstream signaling, and existing and potential interventions for targeting these cytokines or related signal pathways. We believe that a comprehensive understanding of CS in COVID-19 will help to develop better strategies to effectively control immunopathology in this disease and other infectious and inflammatory diseases.
Article
Emerging SARS-CoV-2 variants, especially those of concern, may have an impact on the virus's transmissibility and pathogenicity, as well as diagnostic equipment performance and vaccine effectiveness. Even though the SARS-CoV-2 Delta variant (B.1.617.2) emerged during India's second wave of infections, Delta variants have grown dominant internationally and are still evolving. On November 26, 2021, WHO identified the variant B.1.1.529 as a variant of concern, naming it Omicron, based on evidence that Omicron contains numerous mutations that may influence its behaviour. However, the mode of transmission and severity of the Omicron variant remains unknown. We used computational studies to examine the Delta and Omicron variants in this work and found that the Omicron variant had a higher affinity for human ACE2 than the Delta variant due to a significant number of mutations in the SARS-CoV-2 receptor binding domain, indicating a higher potential for transmission. Based on docking studies, the Q493R, N501Y, S371L, S373P, S375F, Q498R, and T478K mutations contribute significantly to high binding affinity with human ACE2. In comparison to the Delta variant, both the entire spike protein and the receptor-binding domain (RBD) in Omicron include a high proportion of hydrophobic amino acids such as leucine and phenylalanine. These amino acids are located within the protein's core and are required for structural stability. We observed a disorder-order transition in the Omicron variant between spike protein RBD regions 468-473, and it may be significant in the influence of disordered residues/regions on spike protein stability and binding to ACE2. A future study might investigate the epidemiological and biological consequences of the Omicron variant. This article is protected by copyright. All rights reserved.
Article
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) emerged from Wuhan in China before it spread to the entire globe. It causes coronavirus disease of 2019 (COVID-19) where mostly individuals present mild symptoms, some remain asymptomatic and some show severe lung inflammation and pneumonia in the host through the induction of a marked inflammatory ‘cytokine storm’. New and efficacious vaccines have been developed and put into clinical practice in record time, however, there is a still a need for effective treatments for those who are not vaccinated or remain susceptible to emerging SARS-CoV-2 variant strains. Despite this, effective therapeutic interventions against COVID-19 remain elusive. Here we review potential drugs for COVID-19 classified on the basis of their mode of action. The mechanisms of action of each are discussed in detail to highlight the therapeutic targets that may help in reducing the global pandemic. The review was done up to July 2021 and the data was assessed through the official websites of WHO and CDC for collecting the information on the clinical trials. Moreover, the recent research papers were also assessed for the relevant data. The search was made based on keywords like Coronavirus, SARS-C0V-2, drugs (specific name of the drugs), COVID-19, clinical efficiency, safety profile, side-effects etc.This review outlines potential areas for future research into COVID-19 treatment strategies.
Article
The spike protein of coronavirus is key target for drug development and other pharmacological interventions. In current study, we performed an integrative approach to predict antigenic sites in SARS-CoV-2 spike receptor binding domain and found nine potential antigenic sites. The predicted antigenic sites were then assessed for possible molecular similarity with other known antigens in different organisms. Out of nine sites, seven sites showed molecular similarity with 54 antigenic determinants found in twelve pathogenic bacterial species (Mycobacterium tuberculosis, Mycobacterium leprae, Bacillus anthracis, Borrelia burgdorferi, Clostridium perfringens, Clostridium tetani, Helicobacter Pylori, Listeria monocytogenes, Staphylococcus aureus, Streptococcus pyogenes, Vibrio cholera and Yersinia pestis), two malarial parasites (Plasmodium falciparum and Plasmodium knowlesi) and influenza virus A. Most of the bacterial antigens that displayed molecular similarity with antigenic sites in SARS-CoV-2 RBD (receptor binding domain) were toxins and virulent factors. Antigens from Mycobacterium that showed similarity were mainly involved in modulating host cell immune response and ensuring persistence and survival of pathogen in host cells. Presence of a large number of antigenic determinants, similar to those in highly pathogenic microorganisms, not merely accounts for complex etiology of the disease but also provides an explanation for observed pathophysiological complications, such as deregulated immune response, unleashed or dysregulated cytokine secretion (cytokine storm), multiple organ failure etc., that are more evident in aged and immune-compromised patients. Over-representation of antigenic determinants from Plasmodium and Mycobacterium in all antigenic sites suggests that anti-malarial and anti-TB drugs can prove to be clinical beneficial for COVID-19 treatment. Besides this, anti-leprosy, anti-lyme, anti-plague, anti-anthrax drugs/vaccine etc. are also expected to be beneficial in COVID-19 treatment. Moreover, individuals previously immunized/vaccinated or had previous history of malaria, tuberculosis or other disease caused by fifteen microorganisms are expected to display a considerable degree of resistance against SARS-CoV-2 infection. Out of the seven antigenic sites predicted in SARS-CoV-2, a part of two antigenic sites were also predicted as potent T-cell epitopes (KVGGNYNYL⁴⁴⁴⁻⁴⁵² and SVLYNSASF³⁶⁶⁻³⁷⁴) against MHC class I and three (KRISNCVADYSVLYN³⁵⁶⁻³⁷⁰, DLCFTNVYADSFVI³⁸⁹⁻⁴⁰², and YRVVVLSFELLHA⁵⁰⁸⁻⁵²⁰) against MHC class II. All epitopes possessed significantly lower predicted IC50 value which is a prerequisite for a preferred vaccine candidate for COVID-19.