Cells: Thymoquinone: A Tie-Breaker in Sars-Cov2-Infected Cancer Patients?

Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

cells

Review
Thymoquinone: A Tie-Breaker in SARS-CoV2-Infected
Cancer Patients?
Sawsan Elgohary 1,† , Aya A. Elkhodiry 1,† , Nada S. Amin 1,† , Ulrike Stein 2,3,4 and Hend M. El Tayebi 1, *

1 Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology,


Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835 Cairo, Egypt;
[email protected] (S.E.); [email protected] (A.A.E.); [email protected] (N.S.A.)
2 Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
[email protected]
3 Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
4 German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
* Correspondence: [email protected]; Tel.: +20-100-5566415; Fax: +20-2-27581041
† These authors have contributed equally to the manuscript.

Abstract: Since the beginning of the SARS-CoV-2(severe acute respiratory syndrome-coronavirus-


2) pandemic, a race to develop a vaccine has been initiated, considering the massive and rather
significant economic and healthcare hits that this virus has caused. The pathophysiology occurring
following COVID-19(coronavirus disease-2019) infection has given hints regarding the supportive
and symptomatic treatments to establish for patients, as no specific anti-SARS-CoV-2 is available
yet. Patient symptoms vary greatly and range from mild symptoms to severe fatal complications.
Supportive treatments include antipyretics, antiviral therapies, different combinations of broad-
spectrum antibiotics, hydroxychloroquine and plasma transfusion. Unfortunately, cancer patients
 are at higher risk of viral infection and more likely to develop serious complications due to their

immunocompromised state, the fact that they are already administering multiple medications, as
Citation: Elgohary, S.; Elkhodiry, well as combined comorbidity compared to the general population. It may seem impossible to find
A.A.; Amin, N.S.; Stein, U.; El Tayebi,
a drug that possesses both potent antiviral and anticancer effects specifically against COVID-19
H.M. Thymoquinone: A Tie-Breaker
infection and its complications and the existing malignancy, respectively. Thymoquinone (TQ) is
in SARS-CoV2-Infected Cancer
the most pharmacologically active ingredient in Nigella sativa seeds (black seeds); it is reported to
Patients? Cells 2021, 10, 302.
have anticancer, anti-inflammatory and antioxidant effects in various settings. In this review, we
https://doi.org/10.3390/
cells10020302
will discuss the multiple effects of TQ specifically against COVID-19, its beneficial effects against
COVID-19 pathophysiology and multiple-organ complications, its use as an adjuvant for supportive
Academic Editor: Chae-OK Yun COVID-19 therapy and cancer therapy, and finally, its anticancer effects.
Received: 20 December 2020
Accepted: 28 January 2021 Keywords: SARS-CoV-2; COVID-19; cancer; thymoquinone; apoptosis; COVID-19 treatments; anti-
Published: 2 February 2021 cancer drugs; COVID-19 complications; Nrf2; GRP78; RAS

Publisher’s Note: MDPI stays neutral


with regard to jurisdictional claims in
published maps and institutional affil- 1. Introduction
iations.
In March 2020, the WHO declared coronavirus disease -2019 (COVID-19) as a pan-
demic; this served as the spark that ignited the global race for the development of a vaccine
against the novel coronavirus, severe acute respiratory syndrome-coronavirus-2(SARS-
CoV-2). This was evidently important amidst the sudden and substantial increase in
Copyright: © 2021 by the authors. hospitalizations for pneumonia associated with multi-organ disease. The pathophysiol-
Licensee MDPI, Basel, Switzerland. ogy of COVID-19 patients gave clues to suitable supportive and symptomatic treatment
This article is an open access article of this pandemic; however, no specific treatment against COVID-19 has been found to
distributed under the terms and
date. COVID-19 patients have many symptoms that vary in severity, including dry cough,
conditions of the Creative Commons
fever [1], sore throat, fatigue, diarrhea, shortness of breath, myalgia, in addition to ra-
Attribution (CC BY) license (https://
diographic and laboratory or biochemical abnormalities [2]. Moreover, cardiovascular
creativecommons.org/licenses/by/
manifestations include. Acute cardiac injury, myocarditis, arrhythmia and cardiovascular
4.0/).

Cells 2021, 10, 302. https://doi.org/10.3390/cells10020302 https://www.mdpi.com/journal/cells


Cells 2021, 10, 302 2 of 29

thromboembolism havebeen frequently reported in COVID-19 patients [3]. Furthermore,


dizziness, headache, taste and smell dysfunctions, or impaired consciousness has been
frequently shown among neurological manifestations in COVID-19 patients [4,5]. Cases in-
volving either viral co-infection or co-infection with both viral and bacterial pathogens that
cause pneumonia have been described, particularly in the period following the initial phase
of viral respiratory infection [6]. In severe cases, acute lung injury (ALI), acute respiratory
distress syndrome (ARDS), respiratory failure, heart failure, sepsis, multiple organ dys-
function, and sudden cardiac arrest occur within a few days [7]. The supportive treatments
include antipyretics, antiviral therapies, different combinations of broad-spectrum antibi-
otics, hydroxychloroquine and plasma transfusion [8]. Cancer patients are at higher risk
(two-folds) for COVID-19 and are more likely to develop severe outcomes compared to the
general population [9–11]. This is greatly attributed to immunosuppressive conditions, con-
sumption of multiple medications, combined comorbidity, and are more likely to require
ventilatory support or ICU admission [12–15]. They also have significantly higher fatality
rates (28.6% compared to 2.3% for all COVID-19 patients in China) [12]. Patients who have
received cancer treatment within 14 days of getting a COVID-19 diagnosis are considered
to have a risk factor for developing adverse events, such as ARDS, septic shock, and
acute myocardial infarction [12]. Despite this observation, delaying cancer treatment for
patients also has well-documented complications [12]. It may seem rather challenging
to discover a drug that shows not only antiviral effects specifically against SARS-CoV-2
infection and entry but also antimicrobial (antiviral, -bacterial, -fungal) that protects against
SARS-CoV-2 co-infection induced pneumonia, ameliorate respiratory symptoms, as well
as acts as an antioxidant thereby protecting against COVID-19 associated multiple organ
dysfunction (i.e., cardio-protective, hepato-protective, gastro-protective and protects the
kidney from injury). Moreover, this drug should also ideally have anti-inflammatory effects
that are sufficient enough to ameliorate COVID-19 induced cytokine storm and whose
overall pharmacological effects are consistent with the pathophysiology of COVID-19 in
cancer patients.
TQ shows all the above-mentioned effects [16–26]. TQ is the most pharmacologically
active ingredient in Nigella sativa seeds (black seeds) extract [27,28]. In this review, we
will be discussing the potential effects of TQ as a SARS-CoV-2 antiviral drug, its beneficial
effects against COVID-19 pathophysiology with a focus on cancer patients, as well as
some of its anticancer effects and its use as an adjuvant besides supportive COVID-19
therapy and cancer therapy. To achieve the purpose ofthe review, research was conducted
at the States National Library of Medicine (PubMed). For the search in databases, the
descriptors used were: “thymoquinone” and “COVID-19”/“SARS-CoV-2” or “cancer”,
“COVID-19”/“SARS-CoV-2” and “cancer” or “apoptosis”, “thymoquinone” and various
COVID-19 complications and key molecular pathways. Research papers, books, and
published data were reviewed for their relevance to the aim of the review and summarized.
Criteria for inclusion were complete, relevant publication, available online, in English,
published between 2010 and 2020, with detailed information about participants, methods,
and analyses. Data collection was done during August 2020, and data abstracted was in
the form of descriptive information, covering the type of samples used, techniques, and
findings or effects reported. Bias was limited through the evaluation of the studies through
their internal validity rather than the conclusion.

2. Sars-Cov-2 Structural Features Involved in Entry


Understanding the structural features of SARS-CoV-2 and its replication is pivotal to
focus on molecular targets lessening the viral entry, replication and to fully understand the
possible treatment options. SARS-CoV-2 belongs to the family of beta-corona viruses (β-
CoVs) [29] that have the crown-shaped spike (S) trans-membrane glycoprotein. The human
serine proteases participate in viral attachment and RNA entry into the cell. The transmem-
brane serine protease 9 (TMPRSS2) and endogenous serine proteases, such as furin, prime
and cleave the S spike into S1 and S2; this separation is essential for the attachment of
Cells 2021, 10, x FOR PEER REVIEW tor
entry (S1-part
transmembrane
and of theofdelivery
spike)
serine and of theRNA
protease
its cell 9membrane
(TMPRSS2)
into (S2-part of
and endogenous
theangiotensin-converting
cytoplasm spike).where
[30,31], The attachment
serineit isproteases,
ready of be
to th
s
tachment
spikes to both thethe SARS-CoV-2
ACE2 receptor to both and the
the cell membrane is necessaryenzyme for 2successfu
(ACE2)
furin,
lated prime
as mRNA. and cleave the S spike into S1 and S2; this separation is essential for
Cells 2021, 10, x FOR PEER REVIEW tor entry (S1-part
transmembrane
and of theofdelivery
spike)
serine and theRNA
protease
of its cell 9membrane
(TMPRSS2)
into (S2-part of
and endogenous
theangiotensin-converting
cytoplasm spike).where
[30,31], The attachment
serineit isproteases,
ready of be
to th
s
tachment
spikes Since to both thethe
viral SARS-CoV-2
spike
ACE2 attachment
receptor to both to
and the
ACE2
the receptors
cell membrane is key is for its
necessaryenzyme
entry for 2successfu
and(ACE2)
infect
furin,
lated
tor prime
as
(S1-part mRNA. and
ofdelivery
spike)cleaveand the
the S spike into
cell 9membrane S1 and
(S2-part S2; this
of separation
aspike). The is essential
attachment for
ofent
th
Cells 2021, 10, x FOR PEER REVIEW cells, entry the
andlocation
transmembrane the of these
serine of particular
protease
its RNA into receptors
(TMPRSS2) explains
and endogenous
theangiotensin-converting
cytoplasm lot about
[30,31], where SARS-CoV-2
serineit isproteases,
ready to bes
tachment
spikes Since
subsequent to of
both thethe
viral
COVID-19SARS-CoV-2
spike
ACE2 attachment
receptor
symptoms. to both to
and
ACE2 the
ACE2
the receptors
cell
receptorsmembrane are ishighly
keyis for
necessaryenzyme
its entry
expressed for 2successfu
and
in (ACE2)
the infect
lun
Cells 2021, 10, 302 furin,
lated
tor
cells, prime
as
(S1-part
the mRNA. and
oftype
spike)cleaveand the
the S spike into
cell 9membrane S1 and
(S2-part S2; this
of separation
aspike). The is 3essential
of
attachment
29 for
ofent
th
Cells 2021, 10, x FOR PEER REVIEW example, entry
tachment andlocation
transmembrane
Since the
on
of the
viral
of
serine
delivery these
2 alveolar
SARS-CoV-2
spike of particular
protease
its
attachment RNA
cells
to both into receptors
(TMPRSS2)
(AT2)),
to the
ACE2 nose, explains
and
theangiotensin-converting
cytoplasm endogenous
oropharynx,
receptors
lot kidney
[30,31],
ishighly
key
about
where
for its
SARS-CoV-2
serineit isproteases,
proximal
enzyme
entry ready to
tubule
2successfu
and(ACE2) be
infect s
spikes
subsequent
furin,
lated
bladder as to
prime both
mRNA.
urothelial the
COVID-19
and ACE2
cleave
cells, receptor
symptoms.
the
cells S ofspike
the and
ACE2
into the S1 cell
receptors
gastrointestinal and membrane
S2; are
this
tract is necessary
separation
[12] andexpressedis for in
essential
myocardial the
cellslun
for [
21, 10, x FOR PEER REVIEW tor
cells,
entry (S1-part
example, the
andlocation
transmembrane the
on oftype
spike)of
serine
delivery and
these
2 COVID-19of
alveolar the
its cell 9membrane
particular
protease RNA
cells into receptors
(TMPRSS2)
(AT2)), the nose, (S2-part
explains
and
cytoplasm of
3 a spike).
oflot
endogenous
oropharynx, [30,31], The
about
where
30 kidney attachment
SARS-CoV-2
serineit isproteases,
proximalready ofent
to
tubule th
bes
tachment
This
spikes Since
explains
subsequent of
tomRNA.
both the
viral why SARS-CoV-2
spike
thecleave
COVID-19 ACE2 attachment
receptor
symptoms. to both
to
patients
and
ACE2 the
ACE2 do
theS1 angiotensin-converting
receptors
not
cell
receptors only
membrane is key
experience
are for enzyme
its entry
respiratory
is necessary
highly expressed 2
and(ACE2)
infect
problem
forinsuccessfu
the lun[
the SARS-CoV-2 furin,
lated
bladder
tor
cells, prime
as
to(S1-part
both
the theof and
urothelial
location cells, the
cells
angiotensin-converting
spike)of and
these the Scell
spike
ofARDS
particular the into
gastrointestinal
membraneenzyme
receptors and S2;
2explains
(ACE2)
(S2-part this
tract
of a separation
[12]
receptor
spike).
lot andThe
about is essential
myocardial
(S1-part
attachment
SARS-CoV-2 of and cellsfor
of th
21, 10, x FOR PEER REVIEW pneumonia
transmembrane
entry
example,
tachment andon
Since the
of
that
type
the
viral
might
serine
delivery
2 COVID-19
spike
lead
alveolar
SARS-CoV-2 protease
of
attachment
to
its RNA
cells
to into
both
to
but
9(AT2)),
(TMPRSS2)
the
the
ACE2
may
nose, also
and
cytoplasm experience
endogenous
oropharynx,
angiotensin-converting
receptors [30,31],
3
is of
key30 heart,
where
kidney
for serinekidneys
it isproteases,
proximal
enzyme
its entry ready
2
and toent
tubule
(ACE2)
dig
be
infect s
spike) and the This cell
spikes explains
membrane
subsequent
tract tomRNA.
disorders. both why
(S2-part
the
COVID-19
The ACE2 of spike).
receptor
symptoms.
susceptibility patients
The
and
ACE2
and thedo
attachment not
cell
receptors
vulnerability only of
membrane experience
the
are viral
of highly respiratory
spikes
is necessary
cancer expressed
patientsto both
for problem
toinsuccessfu
the
COVID- lun[
furin,
lated
bladder
tor prime
as and
urothelial cleave
cells, the
cells S ofspike
the into S1
gastrointestinal and S2; this
tract separation
[12] and is essential
myocardial cellsfor
21, 10, x FOR PEER REVIEW transmembranethe ACE2
serine receptor
protease entry9(S1-part
cells,
pneumonia
example, the
andandlocation
(TMPRSS2)
multi-factorial
tachment
This Since
explains
the
the
on
of
of
that
type
the
viral
spike)
cell
why
of
might and
these
membrane
and
delivery
2 COVID-19
issue. lead
alveolar
Cancer
SARS-CoV-2
spike
the
endogenous
of its
attachment
cell
particular
tois
RNA ARDS
cells
patientsmembrane
necessary
into receptors
but
serine
(AT2)),
topatients
both
to the
tend
the
ACE2
may
for (S2-part
explains
also
successful
proteases,
cytoplasm
nose,
to oropharynx,
have
doangiotensin-converting
receptors
not only
of
3 a
experience
such spike).
lot
viral
[30,31],
higher of as
ishighly
key
experience
The
about
heart,
entry
where
30 kidney
levels
for of attachment
SARS-CoV-2
kidneys
and
it isthe
proximal
ACE2
enzyme
its entry
respiratory ready and ofent
to
tubule
expression
2successfu
and(ACE2)
th
dig
be
infect
problem
delivery
furin, prime and cleave the S bladder of its spikes
subsequent
tract
RNA
spike
lated into
as to
disorders.
into both
the
mRNA. S1
urothelial the
COVID-19
The
cytoplasm
and ACE2
S2;
cells, receptor
symptoms.
susceptibility
[30,31],
thiscellsseparation and
where
ofARDS
the ACE2
and the
it is cell
receptors
vulnerability
ready
is essential
gastrointestinal membrane
to
forare
be of
the
tract is
cancer
translatednecessary
expressed
patients
at-outcomes
[12] and as mRNA. for
myocardial to in the
COVID-
cellslun
21, 10, x FOR PEER REVIEW transmembrane serine protease
are
tor
cells,
entry
therefore
9(S1-part
pneumonia
example, the
(TMPRSS2)
and
multi-factorial the
on of
location
that
type
more
spike)of
might
and
delivery
likely
and
theselead the
endogenous
2 COVID-19
issue. of
alveolar its
tocell
particular
to
RNA
cells
experience
membrane
receptors
but
serine
into
(AT2)), the may more
(S2-part
explains
also
proteases,
cytoplasm
nose,
adverse
of
3 a
experience
such
oropharynx, spike).
oflot
[30,31], as The
about
heart,
where
kidney
[13]. Additio
attachment
SARS-CoV-2
kidneys
it is ready
proximal and to
tubule th[
ofent
dig
be
tachment of the SARS-CoV-2 Since viralto spike
This both
TMPRSS Since attachment
the
explains andviral why spike
cathepsin toCancer
ACE2
angiotensin-converting
attachment
L patients
receptors
(CTSL), to
patients
two tend
enzyme
ACE2 is
do
key keyto2 have
for
(ACE2)
receptors
not
enzymesonly higher
its entry
recep-
is key
experience
that levels
30
and
formfor of
its ACE2
infection
entry
respiratory
essential ofexpression
and infect
problem
interaction
furin, primeofand cleave the spikes
subsequent
tract to
disorders. both the
COVID-19
The ACE2 receptor
symptoms.
susceptibility and
ACE2
and isthe cell
receptors
vulnerability membrane areof isoutcomes
highly
cancer necessary
expressed
patients fortoinsuccessfu
the
COVID- lun[
21, 10, x FOR PEER REVIEW tor (S1-part
transmembrane
cells,
spike)
serine
the
and theScell
location
protease
spike
lated
bladder
are
cells,
the
entry9
of as
viral
example,
into
therefore
membrane
pneumonia the mRNA.
these
(TMPRSS2)
and
multi-factorial spike
S1
urothelial
the
on
and
more
particular
location
that (S2-part
of
mightS2;
cells,
proteins,
and
delivery
type
this
likely
these
2 COVID-19
issue. of cells
were
endogenous
ofLACE2
alveolar
Cancer its
separation
receptors
lead toof
spike).
particular
to
RNA
the
ARDS
found
cells
patients
explains
The
into but
to
serine
(AT2)), be
the
tend
essential
gastrointestinal
experience attachment
receptors may amore
lot
also
upregulated
proteases,
cytoplasm
nose,
to
for
about
of
explains thethe
tract
adverse
experience
such
oropharynx,
have in
at-
[12]
certain
[30,31],
higher
3 of as
and
SARS-CoV-2
viral
a lot about
heart,
where
kidney
levels
30
myocardial
types
of
[13].
entry
SARS-CoV-2
kidneys
of
itlungs
is ready
proximal
ACE2
cells
Additio
and
cancer, toent
tubule
expression dig
as
bes
tachment
spikes to of
boththetheSARS-CoV-2
and subsequent
ACE2 receptor to
This both
TMPRSS Since
COVID-19
tract
in and
subsequent the
explains and
the
disorders.
Table 1 angiotensin-converting
viral why spike
cathepsin
symptoms.
cell
COVID-19
[34]. membrane
The attachment
(CTSL),
symptoms.
susceptibility is to
patients enzyme
two
receptors
necessaryACE2
and ACE2 do
key are
for 2
not (ACE2)
receptors
enzymesonly
highly
successful
receptors
vulnerability recep-
is key
experience
that
expressed
areof viralform
highly
cancer for in its
theentry
respiratory
essential
expressed
patients to and
in the infect
problem
interaction
COVID- lun
furin, primeofand cleave the Son spike
lated
bladder
are as2into
thereforemRNA. S1
urothelial and
more S2;
cells, this
likelycellsseparation
toof the is essential
gastrointestinal
experience more for the
tract
adverse at-outcomes
aof[12] and myocardial
[13]. cells
Additio [s
21, 10, x FOR PEER REVIEW tor (S1-part
transmembrane
entry and of thethespike)
(for
serine
delivery and
example,
of theRNA
protease
its cell
the 9membrane
cells,
pneumonia
type
example, the
viral
(TMPRSS2)
into
multi-factorial location
alveolar
spike
the
on that (S2-part
cytoplasm
type of
might
cells
proteins,
and theseof
lead
(AT2)),
werespike).
particular
endogenous
2 COVID-19
issue. alveolar[30,31],
Cancer to ARDS
nose,
found
cells The
where
patients toattachment
receptors
but
oropharynx,
serine
(AT2)), itbe
tend ismay also
kidney
upregulated
proteases,
ready
nose,
to have of
explains
to the
such
be
oropharynx, viral
experience
in lot
proximal about
certain
3trans-
higher as heart,
30 kidney
levels tubuleSARS-CoV-2
types
of kidneys
cells,
of
proximal
ACE2 and
cancer, ent
dig
as
tubule
expression
tachment
spikes tomRNA.
bothandtheSARS-CoV-2
ACE2urothelial
bladder receptor to
This
tractboth
TMPRSS Since
and
subsequent
cells, the
explains and
1the
disorders. angiotensin-converting
viral
cells why
cell
COVID-19
of spike
cathepsin
membrane
Thethe attachment
L (CTSL),
symptoms. is
gastrointestinal
susceptibility to
patients
necessary enzyme
two
ACE2ACE2
tract do
key for 2
not
[12] (ACE2)
receptors
enzymesonly
successful
receptorsand are recep-
is key
experience
thatviralform
highly
myocardial for its entry
respiratory
essential
expressed
cells [32,33]. and infect
problem
interaction
toinCOVID-
the lun[
furin,
lated prime
as cleave the in
Table
Scell Table
spike
bladder 1.into
The [34].
differential
S1
urothelial and S2;
cells, expression
thiscellsseparation
toof theof and
key vulnerability
isenzymes
essential
gastrointestinal essential
for of
the
tract cancer
forat-viral patients
spike proteins interac
tor (S1-part
transmembrane
entry and ofdelivery
the spike)
serine
This and theRNA
protease
explains
of its
are
cells,
why 9
therefore
membrane
pneumonia
the
example, the
viral
(TMPRSS2)
COVID-19
into
multi-factorial location
spike
the
on that more
(S2-part
cytoplasm
type of
might
proteins,
and
patients
2
issue.
likely
theseof
lead
werespike).
particular
endogenous
alveolar do
[30,31],
Cancer to
not
cells
experience
ARDS
found The
only
where
patients attachment
receptors
but
to
serine
(AT2)), beismay
experience
it
tend
more
proteases,
ready
nose,
to of
explains
also
upregulated to
adverse
the
such
respiratory
be
oropharynx,
have higher a [12]
viral
experience
in lot
trans- as
and
outcomes
about
certain heart,
kidney
levels
myocardial
types
problems
of
[13].
SARS-CoV-2
kidneys
of
proximal
ACE2 cancer,
like
cells
Additio
and ent
digs
as
tubule
expression
tachment
Since ofviral
thetheSARS-CoV-2
spike attachment different
to
This both
TMPRSS to
explainscancer
the
ACE2 and types.COVID-19
angiotensin-converting
receptors
why
cathepsin is key
L (CTSL), for
patients enzyme
its
two entrydo
key 2successful
and
not (ACE2)
enzymes infection
only recep-
experience
that of expressed
form respiratory
essential problem
interaction
spikes
furin, to
prime both ACE2
pneumonia receptor
that tract
in
Table and
subsequent
Table
might 1.into1the
disorders.
The
lead cell
COVID-19
[34]. membrane
The
differential
toandARDS symptoms.
susceptibility
expression
but may is necessary
also ACE2
and
of key for
isenzymes
experience receptors
vulnerability are
essential
heart, of viral
highly
cancer
for
kidneys at-viral
andpatients
spike to
proteins
digestive in the
COVID- lun
interac
lated
tor as
(S1-part
cells, the
transmembrane
entry and
mRNA. ofand
location
the spike)cleave
serine
delivery and
of theseof
the
the
protease
its
Scell
RNA
spike
bladder
are
particular
the 9
therefore
membrane
pneumonia
viral
example, receptors
(TMPRSS2)
into
multi-factorial
Cancer spike
the
Type on
S1
urothelial
that more
(S2-partS2;likely
cells,
explains
might
proteins,
cytoplasm
type and
2
issue.
this
of
alveolar
cells
lead
were
endogenous
separation
[30,31],
Cancer
toof
aspike).
lot
to the
about
ARDS
found
cells The
where
patients to
serine
(AT2)), be
it
tend
essential
gastrointestinal
experience attachment
SARS-CoV-2
but ismay
TMPRSS
more
also
upregulated
proteases,
ready
nose,
to have to
for
of the
entrythe
tract
adverse
such
be
oropharynx,
higher in [12]
viral
experience and
certain
trans- as
and
outcomes
heart,
kidney
levels
myocardial
types
of
[13].
kidneys
of
proximal
ACE2CTSL
cells
Additio
and
cancer, tubule
expression dig
as [s
tachment
Since ofviral tract
thethespike disorders.
SARS-CoV-2 attachment different
to
ThisThe
both
to cancer
susceptibility
the
ACE2
explains types.
receptors
why and
angiotensin-converting
COVID-19 is vulnerability
key for enzyme
its
patients entryof
do cancer
2successful
and (ACE2) patients
infection recep- to COVID-19
of essential is
spikes
subsequent
furin,
lated tomRNA.
prime
as bothCOVID-19
and ACE2
cleave receptor
symptoms.
the
TMPRSS
tract
in
Table
Scell
spike
bladder
are and
ACE2
disorders.
Table 1.into
thereforeThe
and
1the cathepsin
cell
receptors
[34]. membrane
The
differential
S1
urothelial and
more cells,are Lhighly
(CTSL),
S2;susceptibility
expression
this
likelycells is
separation
toof theof
two
necessary
expressed
and
key
key fornot
isenzymes
enzymes
in
vulnerability
essential
gastrointestinal
experience more
only
theessential
for
experience
lungsthat
of
the
tract
adverse viral
for
form
(for
cancer
at-viral
[12] and
respiratory
patients
spike
outcomes to
proteins
myocardial
[13].
problem
interaction
COVID-
interac
cells
Additio [s
tor (S1-part
cells, the of
location a multi-factorial
spike)of and
these the DLBC
particular issue.
membrane
pneumonia
the viral Cancer
receptors
spike that patients
(S2-part
explains
might
proteins, of
lead a
were tend
spike).
lot
to to
about
ARDS
found Thehave higher
attachment
SARS-CoV-2
but
to be may also
upregulated levels
of the
entry of
experience
in ACE2
viral
and
certain expression;
heart,
types kidneys
of and
cancer, dig
as
entry
example,
tachment and
Since the
on
of delivery
type
the
viral 2
SARS-CoV-2
spike of
alveolar its
attachment RNA
cells
to
This into
(AT2)),
multi-factorial
Cancer
different
both
TMPRSS to the
ACE2
explains the
Type
cancer
and cytoplasm
nose, oropharynx,
issue.
types.
angiotensin-converting
receptors
why
cathepsin COVID-19[30,31],
Canceris
L key
(CTSL), where
kidney
patients
for
patients it
enzyme
its
two entry is
proximal
tend
TMPRSS
do
key ready
to2
and
not have
(ACE2)
enzymes to
tubule be
higher
infection
only trans-
cells,
recep-
experience
that levels
of
form of ACE2CTSL
respiratory
essential expression
problem
interaction
spikesastomRNA.
subsequent both theyACE2
the
COVID-19 aresymptoms.
therefore
receptor ESCA
tract
in
Table more
and
ACE2 1the
disorders.
Table 1. The likely
cell
receptors
[34]. to
The
differential experience
membrane are is
highly
susceptibility
expression more
necessary adverse
expressed
of and
key forin
vulnerability
enzymes outcomes
successful
the lungs
essential [13].
ofviral
(for
cancer
for viralAdditionally,
patients
spike proteins to COVID-
interac
lated
bladder
tor (S1-part
cells, the urothelial
oftype
locationspike) cells,
of and
these cells
the of
are
particularthe
DLBC
cell
pneumonia
the gastrointestinal
therefore
membrane
viral receptors
spike that more
(S2-part
explains
might
proteins, tract
likely
of
lead
were [12]
aspike).
lot
to and
tofound
about
ARDS Themyocardial
experience attachment
SARS-CoV-2
but
to be may more
also
upregulated cells [32,33].
adverse
ofentry
the viral
experience
in and outcomes
certain heart,
types [13].
kidneys
of Additio
and as
cancer, digs
entry
example, and the
on TMPRSS
delivery
2 of
alveolar and
its cathepsin
RNAGBM
cells
Cancer into
(AT2)),
multi-factorial
different Type L
the
cancer(CTSL),
cytoplasm
nose, two
oropharynx,
issue.
types. Cancer key
[30,31], enzymes
where
kidney
patients that
it is
proximal
tend
TMPRSS form
ready
to have essential
to
tubule be
highertrans-interactions
cells,
levels of ACE2 with
CTSL expression
This
spikes Since
explains
subsequentto viral
both why
thespike
COVID-19 ACE2 attachment
COVID-19 receptor
symptoms. TMPRSS
ESCA
tract
in to
patients
and
ACE2 ACE2 do
and
the
disorders.
Table 1 receptors
not
cell
receptors
[34]. only
cathepsin
membrane
The are is key
experience
L (CTSL),
is
highly
susceptibility for its
necessary
expressed
and entry
respiratory
two key for and
in infection
problems
enzymes
successful
vulnerability the lungsthat
of viralof
like
form
(for
cancer essential
patients interaction
to COVID-
lated
bladder as mRNA.
urothelialthe cells,
viral spike
cells proteins,
Table
of the 1. The were
gastrointestinal found
differential to be upregulated
expression
tract of key in certain
enzymes types
essential of
forcancer,
viral as shown
spike proteins interac
cells,
entry
example,the
pneumoniaand location
that
the
on of
might
delivery
type 2 theselead
of
alveolar its to
RNA
are
HNSC
DLBC
particular
ARDS
the
GBM
cells
Cancer
therefore
viral
into
(AT2)),receptors
but
multi-factorial the
Type may
spike moreexplains
also
proteins,
cytoplasm
nose,
likely
experience
oropharynx,
issue. a [12]
were
[30,31],
Cancer lot and
tofound
about
heart,
where
kidney
patients
myocardial
experienceSARS-CoV-2
to kidneys
be
it is
proximal
tend
TMPRSS
more
ready
to and
upregulated
have
cells
to
tubule be
[32,33].
adverse
entry
digestive
higher in and outcomes
certain
trans-
cells,
levels types
of ACE2
[13].
of
CTSL
Additio
cancer,
expression as s
This Since
explains viral in
why Table
spikeCOVID-191 [34].
attachment different
to
patients
TMPRSS cancer
ACE2 do
and nottypes.
receptors
only are
cathepsin is key forexpressed
experience
Lhighly
(CTSL), its
twoentry
respiratory and
keytoenzymes infection
problems ofa essential interaction
like
that(for form
subsequent
tract disorders. COVID-19
The symptoms.
susceptibility LGG
ESCA
in
Table ACE2
and
Table 1. 1
The receptors
vulnerability
[34].
differential of cancer
expression patients
of key enzymes in the
COVID-19 lungs
essential is
for viral spike proteins interac
lated
bladder as mRNA.
cells, the
pneumonia urothelial
location
that cells,
of
might these cells
lead to of
arethe
HNSC
DLBC
particular
ARDS
the gastrointestinal
therefore
viral receptors
but may
spike moreexplains
also
proteins, tract
likely
experiencea [12]
were lot and
tofound
about
heart, myocardial
experienceSARS-CoV-2
to kidneys
be moreand
upregulated cells [32,33].
adverse
entry
digestive
in and outcomes
certain typesCTSL [13].
of Additio
cancer, as s
example,
multi-factorial
Since on type
viral Table2 COVID-19
issue.
spike alveolar
1.Cancer
attachment PADD
GBM
cells
patients
Cancer(AT2)),
different
The differential to tend
Type
cancernose,
expression
ACE2 to oropharynx,
have
types. of
receptors higher
key enzymes
is key kidney
levelsfor of
essential
its proximal
ACE2 TMPRSS
entry for viral
and tubule
expression; spike
infection cells,
they
proteins
of interaction in
This
tract explains
subsequent
disorders. why
COVID-19
The symptoms.
susceptibility inpatients
TMPRSS
LGG
ESCA ACE2
and
Table 1 do
and not
receptors
vulnerability
[34]. only
cathepsin experience
are L
of (CTSL),
highly
cancer respiratory
two
expressed
patients key toin problems
enzymes
the
COVID-19 lungsthat like
form
(for
is a essential interaction
bladder Table 1. The differential expression of key enzymes essential for viral spike proteins interac
are
example,theurothelial
cells,therefore
pneumonia
multi-factoriallocation
onthat
more
type
cells,
different
of
might these
2 COVID-19
issue. alveolar
cells
likely
cancer
lead
Cancer
toof
to thethe
HNSC
DLBC
types.
particular
ARDS
PADD
GBM
cells
patients
Cancer
gastrointestinal
experience
SKCM viral
(AT2)),receptors
but may
spike
tend
Type nose,
to
more explains
also
proteins, tract
adverse
experience
oropharynx,
have higher a [12]
were lot and
outcomes
about
heart,
found
kidney
levels
myocardial
ofto kidneys
be [13].
SARS-CoV-2
proximal
ACE2 TMPRSS and
upregulated cells
Additionally,
tubule
expression;
[32,33].
entry
digestive
in and
certain typesCTSL
cells,
they of cancer, as s
This
TMPRSS explains and why
cathepsin L (CTSL),different
patients
STAD two cancer
do
key nottypes.
enzymesonly experience
that form respiratory
essential problems
interactions like
with
subsequent
tract disorders. COVID-19
The symptoms.
susceptibility LGG
ESCA
in
Table ACE2
and
Table 1 receptors
vulnerability
[34].
1. The differential areof highly
cancer
expression expressed
patients
of key enzymes toin the
COVID-19 lungs
essential (for
is a
for viral spike proteins interac
bladder
are
pneumonia
the viral urothelial
therefore that
spike more cells,
Cancer
might
proteins, cellsto
likely
Type
lead
were toof the gastrointestinal
experience
SKCM
HNSC
DLBC
ARDS
found to but bemay more TMPRSS
also
upregulated tract
adverse in [12]
experience certain and
outcomes
heart, myocardial
types kidneys[13].
of cells
Additionally,
CTSL
and
cancer, [32,33].
digestive
as shown
example,
multi-factorial on type 2
issue. alveolar
Cancer THYM
PADD
GBM
cells
patients
Cancer(AT2)),
different tend
Type
cancernose,
to oropharynx,
have
types. higher kidney
levels of proximal
ACE2 TMPRSS tubule
expression; cells,
they CTSL
This
TMPRSS
tract
in explains
1and
disorders.
Table why
[34].cathepsin
The COVID-19 L (CTSL),
susceptibility patients
STAD
LGG
ESCA two
and do not
key enzymes
vulnerability only experience
that
of cancer respiratory
form essential
patients problemswith
interactions
to COVID-19 like
is aviral spike proteins interac
bladder
are urothelial
therefore more cells,
DLBC cells
likely to Table
CESC
of the 1. The
experience
SKCM
HNSC
DLBC differential
gastrointestinal more expression
tract
adverse [12] andof
outcomes key enzymes
myocardial [13]. essential
cells
Additionally, for
[32,33].
pneumonia
the viral spike
multi-factorial that might
proteins,
issue. lead
were
Cancer topatients
ARDSto
found
THYM
PADD
GBM but bemay
tend also experience
upregulated in certain heart,types kidneys
of cancer, and as digestive
shown
This
TMPRSS
tract explains and
disorders. why
cathepsin
The COVID-19 L (CTSL),
susceptibility
Cancer
different
patients
UCS
STAD
LGG
ESCA two
and
Type do to
cancer
key not have
enzymes
vulnerability onlyhigher
types. experience
that
of
levels
form
cancer
of ACE2
essential
patients
TMPRSS
respiratory expression;
problemswith
interactions
to COVID-19
they
like CTSL
in
areTable
Tabletherefore1 [34].
1. The differential
moreESCAlikelyexpression to CESC of key enzymes
experience
SKCM
HNSC
DLBC more essential
adverse for viral spike proteins
outcomes [13]. interactionisina
Additionally,
pneumonia
the viral spike
multi-factorial that might
proteins,
issue. lead
were
Cancer topatients
ARDSto
found
KICH
THYM
PADD
GBM
Cancer but bemay
tend
Type to also experience
upregulated
have higher in certain
levels heart,types
of kidneys
ACE2 of cancer,
TMPRSS and as
expression; digestive
shownthey CTSL
different cancer
TMPRSS and types.
cathepsin L (CTSL), UCS
STAD two key enzymes that form essential interactions with
tract
in disorders.
Table 1 [34]. The susceptibility LGG
ESCA and vulnerability of cancer patients to COVID-19 isina
Table
are 1. The differential
therefore moreGBM likelyexpression to PRAD
CESC of key
experience
SKCM enzymes more essential
adverse for viral
HNSCto be upregulated in certain types of cancer, as shown
DLBC spike
outcomes proteins
[13]. interaction
Additionally,
the viralType
multi-factorialspike proteins,
issue. Cancer werepatients
found
KICH tend
THYM
PADD
GBM
Cancer
different
TMPRSS
in Table
cancer
1 and
[34].
types.
cathepsin L (CTSL),READ
UCS
STAD
LGG
ESCA two TMPRSS keyto have higher
enzymes that formlevelsessential
of ACE2 CTSL expression; with
interactions they
Table
are
DLBC 1. The
therefore differential
more expression
HNSClikely toHNSC PRAD
CESC of key enzymes
experienceupregulated
SKCM essential
more adverse for viral spike
outcomes proteins
[13]. interaction
Additionally, in
the viral
Cancer spike proteins,
Type were found PADDto be
UCEC
KICH
THYM
GBM TMPRSS in certain typesCTSL of cancer, as shown
different
TMPRSS cancer types.
ESCA
in Table
Table 1and
1. The [34].cathepsin L (CTSL),
differential
LGG expression
READ
UCS
STAD
LGG
PRAD
CESC
SKCM
HNSC
two key enzymes that form essential interactions with
Abbreviations:
of key enzymes TMPRSS,essential transmembrane
for viral spikeserine proteins protease;
interactionCTSL;incathepsin L; DLBC, lympho
DLBC
the viral spike proteins, were found
UCEC
KICH to be upregulated in lymphoma;
certain types of cancer, as shown
GBM
Cancer Type
different cancer types. THYM
PADD
neoplasm TMPRSS
diffuse large B-cell ESCA,CTSL esophageal carcinoma; GBM, glioblastoma
ESCA
in Table 1 [34]. READ
UCS
STAD
LGG
Table
HNSC
DLBC 1. The differential
PADD expression Abbreviations:
PRAD
CESC of
forme;
SKCM keyHNSC, enzymes TMPRSS,
head essential
and transmembrane
neck for viral
squamous spike serine
cell proteins protease;
carcinoma; interactionCTSL;
KICH, incathepsin
kidney L; DLBC, lympho
chromophobe; LGG,
GBM
Cancer Type
different cancer types. UCEC
KICH
THYM
PADD
neoplasm
lower grade TMPRSS
diffuse
glioma; large B-cell
PAAD, lymphoma;
pancreatic ESCA,CTSL
adenocarcinoma; esophageal PRAD, carcinoma;
prostate GBM, glioblastoma
adenocarcinoma; R
LGG
ESCA READ
UCS
STAD
Table
HNSC1. The differential
DLBC SKCMexpression Abbreviations:
PRAD
CESC of key
forme;
SKCM
rectum HNSC, enzymes TMPRSS,
head
adenocarcinoma; essential
and neck transmembrane
SKCM, for viral
skinspike
squamous serine
cell
cutaneous proteins protease;
carcinoma; interaction
melanoma; CTSL;
KICH, STAD,incathepsin
kidney L;adenocarcinom
DLBC, lympho
chromophobe;
stomach LGG,
PADD Type
GBM
Cancer UCEC
KICH
THYM
neoplasm TMPRSS
diffuse large B-cell lymphoma; ESCA,CTSL esophageal carcinoma;
different
LGG
ESCA
cancer types. lower
READgrade
THYM,
UCS
STAD thymoma; glioma; PAAD,
UCEC, pancreatic
uterine corpus adenocarcinoma;
endometrial PRAD,
carcinoma [34]. GBM,
prostate glioblastomaR
adenocarcinoma;
SKCM
HNSC
DLBC STAD Abbreviations:
PRADHNSC,
CESC
forme;
rectum TMPRSS,
head
adenocarcinoma; and neck transmembrane
SKCM, squamous
skin cutaneous serine
cell protease;
carcinoma;
melanoma; CTSL;
KICH, STAD, cathepsin
kidney L;adenocarcinom
DLBC, lympho
chromophobe;
stomach LGG,
PADD Type
GBM
Cancer UCEC
KICH
THYM
neoplasm
lower grade TMPRSS
diffuse
glioma; large B-cell
PAAD, lymphoma;
pancreatic ESCA,CTSL
adenocarcinoma; esophageal PRAD, carcinoma;
prostate GBM, glioblastomaR
adenocarcinoma;
STAD
LGG
ESCA READ
THYM,
UCS
3. thymoma;
Benefits ofhead UCEC,
Thymoquinone uterine corpus endometrial
as an Adjuvant carcinoma
in COVID-19 [34].
SKCM
HNSC
DLBC THYM Abbreviations:
PRAD
CESC
forme;
rectumHNSC, TMPRSS,
adenocarcinoma; and neck transmembrane
SKCM, squamous serine
cell
skin cutaneous protease;
carcinoma;
melanoma; CTSL;
KICH, STAD, cathepsin
kidney L;adenocarcinom
DLBC, lympho
chromophobe;
stomach LGG,
THYM
PADD
GBM UCEC
KICH
neoplasm
lower Asgrade diffuse
mentioned
glioma; large B-cell
previously,
PAAD, lymphoma;
pancreatic there ESCA,
is no
adenocarcinoma; esophageal
targeted carcinoma;
therapy
PRAD, for
prostate GBM,
SARS-CoV-2; glioblastoma
adenocarcinoma; supp R
STAD
LGG
ESCA READ
THYM,
UCS
3. Benefits thymoma; ofhead UCEC,
Thymoquinone uterine corpus endometrial
as an Adjuvant carcinoma
in COVID-19 [34].
CESC
SKCM
HNSC CESC Abbreviations:
PRAD
forme;
rectum
treatments HNSC, TMPRSS,
adenocarcinoma;
against and
the transmembrane
neck
SKCM,
disease squamous
skin serine
cell
cutaneous
manifestations protease;
carcinoma;
melanoma;
are CTSL;
KICH, STAD,
undertaken cathepsin
kidneystomach
as L;adenocarcinom
reviewed DLBC,
chromophobe; in lympho
LGG,
[35,36
THYM
PADD
GBM UCEC
KICH
neoplasm
lower Asgrade diffuse
mentioned
glioma; large B-cell
previously,
PAAD, lymphoma;
pancreatic there ESCA,
is esophageal
no targeted carcinoma;
therapy GBM,
for SARS-CoV-2; glioblastoma supp
UCS
STAD
LGG READ
THYM,
portive
3. Benefits
Abbreviations:
thymoma;
therapyofhead UCEC,
includes
Thymoquinone
TMPRSS,
uterine
steroids
transmembrane
corpus
as an toadenocarcinoma;
endometrial
alleviate
Adjuvant
serine protease;
PRAD,
incarcinoma
the inflammatory
COVID-19
CTSL;
prostate
[34].
cathepsin
adenocarcinoma;
status; literature
L;adenocarcinom
DLBC, lympho shR
CESC
SKCM
HNSC UCS PRAD
forme;
rectum
treatments HNSC,
adenocarcinoma;
against and
the neck
SKCM,
disease squamous
skin cell
cutaneous
manifestations carcinoma;
melanoma;
are KICH, STAD,
undertaken kidney chromophobe;
stomach
as reviewed in LGG,
[35,36
KICH
THYM
PADD UCEC
that
neoplasm
lower
TQ
Asgrade
improved
diffuse
mentioned
glioma; largeoxygenation
B-cell
previously,
PAAD, lymphoma;
pancreatic
when
there iscombined
ESCA, esophageal
no targeted
adenocarcinoma;
with PRAD,
steroids
carcinoma;
therapy forand
prostate GBM,showed
SARS-CoV-2; protect
glioblastoma
adenocarcinoma; supp R
UCS
STAD
LGG READ
THYM,
portive
3. Benefits thymoma;
therapyof UCEC,
includes
Thymoquinone uterine
steroids corpus
as to
an endometrial
alleviate
Adjuvant the
in carcinoma
inflammatory
COVID-19 [34]. status; literature sh
PRAD
CESC
SKCM KICH fects
forme;
rectum inHNSC,
Abbreviations:
treatments the lungs head
adenocarcinoma;
against [37].
TMPRSS, and
the Non-steroidal
transmembrane
neck
SKCM,
disease squamous
skin anti-inflammatory
serine
cell
cutaneous
manifestations protease;
carcinoma;
melanoma;
are CTSL;
KICH, drugs
STAD,
undertaken kidney like
cathepsin diclofenac
L;adenocarcinom
DLBC,
chromophobe;
stomach
as reviewed in are
lympho
LGG,
[35,36als
KICH
THYM
PADD UCEC
that
neoplasm
lower
TQ
Asgrade
improved
diffuse
mentioned
glioma; largeoxygenation
B-cell
previously,
PAAD, lymphoma;
pancreatic
when
there combined
ESCA,
is esophageal
no targeted with steroids
carcinoma;
therapy forand GBM,showed
SARS-CoV-2; protect
glioblastoma supp
READ
UCS
STAD THYM,
portive
3. Benefits
fects in
Abbreviations:
thymoma;
therapy
the ofhead
lungs
UCEC,
includes
Thymoquinone
[37].
TMPRSS,
uterine
steroids
Non-steroidal
transmembrane
corpus
as an toadenocarcinoma;
endometrial
alleviate
Adjuvant
anti-inflammatory
serine the
protease;
PRAD,
incarcinoma
inflammatory
COVID-19
CTSL; drugs
prostate
[34].
like
cathepsin
adenocarcinoma;
status;
diclofenac
L; literature
DLBC, are
lympho sh
als
R
PRAD
CESC
SKCM PRAD forme;
rectum
treatments HNSC,
adenocarcinoma;
against and
the neck squamous
SKCM, skin
disease cell carcinoma;
cutaneous melanoma;
manifestations are KICH, kidney
STAD, stomach
undertaken as chromophobe;
adenocarcinom
reviewed in LGG,
[35,36
UCEC
KICH
THYM that
neoplasm
lower
TQ
Asgrade
improved
diffuse
mentioned
glioma; largeoxygenation
B-cell
previously,
PAAD, lymphoma;
pancreatic
when
there combined
ESCA,
is esophageal
no targeted with steroids
carcinoma;
therapy forand GBM,showed
SARS-CoV-2; protect
glioblastoma supp
READ
UCS
STAD
Abbreviations: TMPRSS, transmembrane
THYM,
portive
3. Benefits
fects in
thymoma;
therapy
the
serine of
lungs
UCEC,
includes
Thymoquinone
[37].
protease;
uterine
steroids
Non-steroidal
CTSL;
corpus
as
cathepsin toadenocarcinoma;
an endometrial
alleviate
Adjuvant
anti-inflammatory
L; DLBC, the
in
lymphoid
PRAD,
carcinoma
inflammatory
COVID-19 drugs
prostate
[34].
like
adenocarcinoma;
status;
diclofenacliterature
are sh
als
R
PRAD
CESC READ forme; HNSC,
rectum
treatments head and
adenocarcinoma;
against the neck
SKCM,
disease squamous
skin cell carcinoma;
cutaneous
manifestations melanoma;
are KICH, STAD,
undertaken kidney chromophobe;
stomach
as adenocarcinom
reviewed in LGG,
[35,36
UCEC
KICH
THYM diffuse large B-cell lymphoma;
neoplasm that
lower
TQ
Asgrade
improved
ESCA,
mentioned
glioma;
oxygenation
esophageal carcinoma;
previously,
PAAD, pancreatic
when
there GBM,iscombined
no glioblastoma
adenocarcinoma; targeted with steroids
multi-
therapy
PRAD, forand
prostate
showed protect
SARS-CoV-2;
adenocarcinoma; supp R
READ
UCS THYM,
portive
3. Benefits thymoma;
therapyof UCEC,
includes
Thymoquinone uterine
steroids corpus
as to
an endometrial
alleviate
Adjuvant the
in carcinoma
inflammatory
COVID-19 [34]. status; literature sh
Abbreviations:
PRADHNSC, head
CESC
forme; TMPRSS,
UCEC transmembrane
and neck fects
squamous
rectum in
treatments the
serine
cell lungs
carcinoma;
adenocarcinoma;
against [37].
protease;the Non-steroidal
CTSL;
KICH,
SKCM,
disease cathepsin
kidney
skin anti-inflammatory
L;
chromophobe;
cutaneous
manifestations DLBC, lymphoid
are LGG,
melanoma; drugs
brain
STAD,
undertaken like
stomach
as diclofenac
adenocarcinom
reviewed in are
[35,36als
UCEC
KICH
neoplasm
lower diffuse large B-cellpancreaticthat TQ
lymphoma; As improved
ESCA,
mentioned oxygenation
esophageal carcinoma;
previously, when
there GBM,iscombined
no glioblastoma
targeted with steroids
multi-
therapy forand showed protect
SARS-CoV-2; supp
UCS grade
READ glioma; PAAD,
Abbreviations: THYM,
portive
3.
TMPRSS,
adenocarcinoma;
Benefits thymoma;
therapy
transmembrane UCEC,
includes
of protease;
Thymoquinone
PRAD,
serineCTSL;uterine
protease;
prostate
steroids corpus
as an
CTSL;
adenocarcinoma;
endometrial
to anti-inflammatory
alleviate
Adjuvant
cathepsin L;the
in
DLBC,
READ,
carcinoma
inflammatory
COVID-19
lymphoid [34]. status;
neoplasm literature
diffuse sh
Abbreviations:
PRAD
forme; HNSC,
rectum adenocarcinoma;TMPRSS,
head and transmembrane
neck
SKCM, fects
squamous in
skin cutaneous
treatments the
serine
cell lungs
carcinoma; [37].
melanoma;
against the Non-steroidal
KICH,
disease cathepsin
kidney
STAD,GBM, L;
chromophobe;
stomach
manifestations DLBC,
adenocarcinoma;lymphoid
are LGG, drugs
brain
undertaken like
as diclofenac
reviewed in are
[35,36als
UCEC
KICH large B-cell lymphoma; that ESCA,
TQ esophageal
improved carcinoma;
oxygenation when glioblastoma
combined multiforme;
with HNSC,
steroids head
and and
showedneck protect
neoplasm
lower
READgrade
THYM, diffuse
thymoma;glioma;large
PAAD,
UCEC,
squamous B-cell lymphoma;
pancreatic
uterine
cell corpus
carcinoma; As ESCA,
mentioned
adenocarcinoma;
endometrial
KICH, esophageal PRAD,
carcinoma carcinoma;
previously, there
prostate
[34]. GBM,is no glioblastoma
adenocarcinoma;targeted multi- PAAD,
therapy
READ, for SARS-CoV-2; supp
portive
3. Benefits
fects therapy
inprostate
the ofkidney
lungs
chromophobe;
includes
Thymoquinone
[37]. steroids
Non-steroidal asLGG,
an brain
Adjuvant lower
to anti-inflammatory
alleviate the
ingrade glioma;
inflammatory
COVID-19 drugs
pancreatic
status; literature sh
Abbreviations:
PRADHNSC,
forme;
rectum TMPRSS,
head
adenocarcinoma; transmembrane
and neckSKCM,
adenocarcinoma; squamous
skin cutaneous
treatments
PRAD, serine
cell protease;
carcinoma;
melanoma;
against the
adenocarcinoma; CTSL;
KICH, STAD,
disease cathepsin
kidney
READ, stomach
manifestations
rectum L;adenocarcinoma;
chromophobe; DLBC, lymphoid
are LGG,
adenocarcinoma; brain
undertaken SKCM, likeas
skin diclofenac
cutaneousin are
reviewed [35,36als
UCEC
neoplasm
lower grade diffuse
glioma;large
PAAD,
melanoma; B-cell that
lymphoma;
pancreatic
STAD, stomach
TQ
As improved
ESCA,
mentioned
adenocarcinoma;
adenocarcinoma;
oxygenation
esophageal carcinoma;
previously,
PRAD,
THYM,
when
there
prostate
thymoma; GBM, combined
is no glioblastoma
adenocarcinoma;targeted with
corpustherapy
steroids
READ,
and
multi- forcarcinoma showed
SARS-CoV-2; protect
supp
READ
THYM,
3. Benefitsthymoma;
Abbreviations: ofhead UCEC,
Thymoquinone
TMPRSS,
uterine
transmembrane
corpus
portive
as an
fects in
endometrial
therapy
Adjuvant
the
serinelungs incarcinoma
includes
[37].
protease; COVID-19
Non-steroidal
CTSL;
[34].
steroids
cathepsin toUCEC,
alleviate uterine
anti-inflammatory
L; DLBC, the
lymphoid
endometrial
inflammatory
drugs like status;
diclofenac
[34].
literature
are sh
als
forme;
rectumHNSC,
adenocarcinoma; and neckSKCM, squamous
skin cell
cutaneous
treatments carcinoma;
melanoma; KICH,
against oxygenation STAD, kidney chromophobe;
stomach
the disease manifestations adenocarcinoma; LGG,
arewith brain
undertaken asandreviewed inprotect
[35,36
UCEC
neoplasm
lower As
grade diffuse
mentioned
glioma;large B-cell
previously,
PAAD,
that
lymphoma;
pancreatic
TQ
there improved
ESCA,
is esophageal
no targeted
adenocarcinoma; carcinoma;
therapy
PRAD,
when
for
prostate GBM, combined
SARS-CoV-2; glioblastoma
adenocarcinoma; supportive steroids
multi-
READ,
showed
THYM, thymoma; 3. UCEC,
Benefits of Thymoquinone
uterine corpus
portive endometrial
therapy as inan Adjuvant
carcinoma
includes [34].
steroids into COVID-19
alleviate the inflammatory
3. Benefits
Abbreviations:
forme;
rectum
treatmentsHNSC,
ofhead
Thymoquinone
TMPRSS,
adenocarcinoma;
against and
the transmembrane
neck
SKCM,
disease
as an
fects
squamous
skin inAdjuvant
the
serine
cell
cutaneous
manifestations lungs
carcinoma; [37].
protease;
melanoma;
are
COVID-19
Non-steroidal
CTSL;
KICH,
undertakenSTAD, cathepsin
kidney anti-inflammatory
L;adenocarcinoma;
chromophobe;
stomach
as reviewed DLBC, in lymphoid
LGG, brain
[35,36]. drugs likestatus;
Sup- diclofenacliterature
are als sh
neoplasm
lower As
grade diffuse
mentioned
glioma;large As mentioned
B-cell
previously,
PAAD,
that
lymphoma;
pancreatic therepreviously,
TQ improved
ESCA,
is
adenocarcinoma;
there
esophageal
no targeted is
oxygenation no targeted
carcinoma;
therapy
PRAD,
when therapy
GBM,
for SARS-CoV-2;
prostate
combined for
glioblastoma
adenocarcinoma;
SARS-CoV-2;
with steroids
multi-
supportive
READ,
supportive
and showed protect
THYM,
portive
3. Benefitsthymoma;
therapy
ofhead UCEC,
includes
Thymoquinone uterine
steroids corpus
as an to endometrial
alleviate incarcinoma
the inflammatory [34]. status; literature showed
forme;
rectum
treatmentsHNSC, treatments
adenocarcinoma;
against and
the neck
SKCM,
diseaseagainstfects
squamous
skin inAdjuvant
the disease
the
cell
cutaneous
manifestations lungs
carcinoma; [37].
melanoma;
are
COVID-19
manifestations
Non-steroidal
KICH,
undertakenSTAD, kidney are undertaken
anti-inflammatory
chromophobe;
stomach
as reviewed adenocarcinoma;
in as
LGG, reviewed
[35,36]. drugs
brain
Sup- inlike
[35,36]. Sup- are als
diclofenac
that
lower
TQ
As
grade
improved
mentioned
glioma;
oxygenation
previously,
PAAD, pancreatic
when
there combined
is no targeted with steroids
therapy for and showed
SARS-CoV-2; protective
supportive ef-
THYM,
portive
3. Benefits
fects
thymoma;
therapy UCEC,
includes
of Thymoquinone
inadenocarcinoma;
the lungs [37].
uterine
steroids
Non-steroidal
corpus
as an toadenocarcinoma;
endometrial
alleviate
Adjuvant
anti-inflammatory
PRAD, prostate
incarcinoma
the inflammatory
COVID-19 [34]. status;
drugsstomachlike
adenocarcinoma;
diclofenac literatureare
READ,
showed
also used
rectum
treatments against the SKCM, skin
disease cutaneous
manifestations melanoma;
are undertakenSTAD, as reviewed adenocarcinoma;
in [35,36]. Sup-
that TQAs improved oxygenation
mentioned previously, wheniscombined
there no targeted with steroids
therapy for and showed protective
SARS-CoV-2; supportive ef-
THYM,
portive
3. Benefitsthymoma;
therapy
of UCEC,
includes
Thymoquinone uterine
steroids corpus
as to
an endometrial
alleviate
Adjuvant the
in carcinoma
inflammatory
COVID-19 [34]. status; literature showed
fects in
treatments the lungs [37]. Non-steroidal
against oxygenation
the disease manifestations anti-inflammatory arewithundertaken drugs like diclofenac
as reviewed are also used
that TQ improved
As mentioned previously, there wheniscombined no targeted steroids
therapy forand showedinprotective
SARS-CoV-2;
[35,36]. Sup-
supportive ef-
Cells 2021, 10, 302 4 of 29

portive therapy includes steroids to alleviate the inflammatory status; literature showed
that TQ improved oxygenation when combined with steroids and showed protective ef-
fects in the lungs [37]. Non-steroidal anti-inflammatory drugs like diclofenac are also
used to counteract COVID-19-induced fever and myalgia [38]. Furthermore, COVID-19
and cancer patients are at high-risk of acetaminophen-induced hepatotoxicity as reviewed
in [39], diclofenac-induced gastrointestinal side effects as reviewed in [40], along with renal
toxicity [41]; luckily, TQ administration protects against acetaminophen-induced hepatotox-
icity [42]. In a recent study, TQ protected against diclofenac-induced kidney injury [42,43]
and ameliorated GIT toxicity with proton pump inhibition activity [44,45]. Moreover,
TQ counteracted the toxic effects of other supportive therapies like gentamicin [46,47],
chloroquine [48] and vancomycin [25] and showed a synergistic effect when administered
with ranitidine [44,45]. Furthermore, tocilizumab, an IL-6 receptor antagonist, has been
used in various cancers, such as multiple myeloma and solid tumors, including renal,
prostate, lung, colorectal and ovarian cancers, to decrease cancer-associated inflammation.
It is used alone or in combination with conventional chemotherapy, as reviewed in [49].
In COVID-19 patients, tocilizumab is given to decrease the viral-induced IL-6 elevation,
cytokine storm, and inflammation [50]. TQ is reported as a potent anti-inflammatory drug
that decreases IL-6 expression and inhibits NFkB [51]. Hence, giving TQ with tocilizumab
could give double beneficial effects in COVID-19 patients. It has been reported that even a
single dose of tocilizumab caused gut ulceration in COVID-19 patients [52], and since TQ
showed gastroprotective effects, it would be beneficial to investigate the GIT protective
effects of TQ in combination with tocilizumab against gut ulceration. Table 2 shows details
about the use of TQ as a potential adjuvant therapy to minimize COVID-19 supportive
treatments’ side effects.

Table 2. Thymoquinone (TQ) as a potential adjuvant therapy to minimize SARS-CoV-2(severe acute respiratory syndrome-
coronavirus-2) treatments’ side effects.

Drug Beneficial Effect of Thymoquinone as Adjuvant Reference


-Reversal of acetaminophen-induced lipid peroxide increase in ALT, total nitrate/nitrite,
and a decrease in GSH and ATP.
Acetaminophen -TQ is effective in protecting mice against acetaminophen-induced hepatotoxicity, liver [23,53]
cirrhosis and fibrosis via increased resistance to oxidative and nitrosative stress and
decreased serum biomarker enzymes (SGOT, SGPT and ALP)
-TQ could ameliorate gastrointestinal and renal toxicity induced by high dose diclofenac
treatment.
Diclofenac [42,43]
-TQ administration, with Diclofenac treatment, alleviated the buildup of poly-unsaturated
fatty acids (PUFAs) and diclofenac-induced cell death in kidney cells.
Gentamicin (severe -TQ showed a protective role against Gentamicin ototoxicity
secondary bacterial -TQ significantly prevented the Gentamicin-induced elevations of serum AST, ALT and [46,47]
infections) LDH activities and also tumor necrosis factor-alpha (TNF-α) and total bilirubin levels
-TQ provides cardio-protective effects by reducing myocardial enzymes following
Chloroquine [48]
chloroquine induced cardiotoxicity.
-TQ reversed the vancomycin induced elevated serum blood urea nitrogen, creatinine and
Vancomycin [25]
MDA and showed protective effects against injury
Steroids -TQ improved oxygenation and protected lung tissue from hazardous effects of human
[37]
(methylprednisolone) gastric juice (pH 1.2)
-TQ and montelukast have dose-dependent effects on cilia beat frequency (CBF),
Montelukast [54]
extending their mechanism of action in respiratory diseases.
Ranitidine, H2 -TQ+ ranitidine showed synergistic effects and a more significant decrease in ulcer index.
blockers, Proton pump -TQ has novel gastroprotective mechanisms via inhibiting proton pump, acid secretion [44,45]
inhibitors and neutrophil infiltration while enhancing mucin secretion, and nitric oxide production
Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; GSH, glutathione; ALP, alkaline phosphatase; SGOT,
serum glutamic oxaloacetic transaminase, SGPT, serum glutamic pyruvic transaminase.
Cells 2021, 10, 302 5 of 29

Thus, far, this literature review has attempted to highlight how TQ could represent
a potential adjuvant molecule to COVID-19 standard supportive treatments. To create a
full picture, and in order to visualize the true value of TQ in COVID-19 cancer patients,
the potential anticancer effects of TQ will be reviewed in the next section, as well as its
potential benefit as an adjuvant to chemotherapy.

4. Anticancer Effects of Thymoquinone


For more than 50 years, TQ has been known for its anti-neoplastic and chemo-
preventive effects that have been proven in various types of cancer [55]. Even though
TQ is not an FDA-approved drug for cancer therapy, several in vitro and in vivo studies
showed that the anticancer activity of TQ is mediated through different pathways involved
in proliferation, cell cycle regulation of apoptosis, angiogenesis and cancer metastasis.
Apoptosis or programmed cell death is considered to be the most powerful shield against
cancer, as it maintains body homeostasis by eliminating aged or diseased cells from the
body [56]. This is why mutations or dysregulations in genes regulating apoptosis, such
as members of the Bcl-2 family, caspases, p53 and PTEN, are usually present in human
cancers [57–60]. The antiproliferative and apoptotic effects of TQ are modulated through
several mediators and transcription factors, along with its anti-angiogenic, antimetastatic
and chemo, radio-sensitizing effects.
TQ and apoptosis: the downregulation of the antiapoptotic proteins Bcl-2, Bcl-xl, XIAP,
survivin, and the upregulation of proapoptotic proteins Bax, p21 and p53 have been
associated with TQ-induced apoptosis in various cancers [61–63]. TQ upregulating effect on
the proapoptotic transcription factor, p53, was clearly emphasized in numerous in vitro and
in vivo studies on cancer models, where TQ induced apoptosis through the upregulation of
p53 in breast cancer [64–66], renal cell carcinoma [67], leukemia [68], glioblastoma [69,70],
squamous cell carcinoma [71], cervical cancer [72], lung cancer [73], and osteosarcoma [74].
The downstream protein of p53, p21 [75–77], was shown to be upregulated in pancreatic
ductal adenocarcinoma as well [78]. The diminished expression of Bcl-2, Bcl-xl, survivin, c-
FLIP and XIAP following TQ treatment has been established in several cell lines and animal
models [79–84]. This diminished expression of Bcl-2 is usually coupled to increased Bax
expression as reported in liver cancer [85], leukemia [83], bladder cancer [84], squamous
cell carcinoma [86], ovarian cancer [82], and renal cancer [87]. Consequently, the disruption
in the Bcl-2/Baxratio, accompanied by reactive oxygen species (ROS) generation induced
by TQ inside cancer cells, damages mitochondrial membrane potential, leading to the
release of cytochrome c. This triggers the intrinsic pathway and activates procaspases 9, 7, 3
and PARP enzyme cleavage. This cascade of events is seen in leukemia/lymphoma [88,89],
glioblastoma [79,90], renal cell carcinoma [91], colon [92], brain [93], bladder [84] and
ovarian cancers [82]. TQ has been reported to exert a dual effect in which it acts as both
pro-oxidant and antioxidant in a dose-dependent manner; the antioxidant effect occurs at
low concentration (<5 µM) whereas, at higher concentrations (>20 µM), it possesses pro-
oxidant property [94]. TQ-induced cell death was mediated through caspase-3 activation
in glioblastoma [79], liver [95,96] and cervical cancer [72], while caspases 9 and 3 mediated
apoptosis wasseen in lymphoma [88], lung [73], and liver cancer [97]. Moreover, the
induction of apoptosis in leukemia was mediated by caspases 8 and 3 involving more than
one apoptotic pathway [83].
TQ and cell cycle arrest: since microtubule assembly is important in cell cycle progres-
sion, microtubule targeting is a potential mechanism for chemotherapy. TQ was reported
to induce cell cycle arrest through depolymerizing microtubule network and disrupting
the mitotic spindle organization in A549 lung cancer cells [98]. Several studies reported
TQ’s cell cycle arrest action through inhibition of cyclin D, cyclin E and cyclin-dependent
kinase 2 (Cdk-2) in various cancers [77,92,99,100]. Additionally, a death receptor-mediated-
apoptosis was seen with TQ treatment, as TQ caused re-localization of CD95/Fas from
cytoplasm to the membrane, enhancing apoptosis in multiple myeloma [101] and enhancing
TRAIL/TRAILR expression in medulloblastoma [93], liver [96,97] and lung cancers [76].
Cells 2021, 10, 302 6 of 29

TQ vs. carcinogen activation: One of TQ’s anticancer mechanisms is the suppression


of carcinogen activation through inhibiting metabolizing enzymes that are responsible
for the biotransformation process of various pro-carcinogens into highly active carcino-
gens. These reactive carcinogens lead to oxidative damage, or covalent modification of
DNA, as well as changes in signaling networks [102–104]. Of particular interest in the
cytochrome p450 family are CYP1 members, specifically CYP1A1 phase I enzyme, that
was found to be subject to inhibition by TQ, accompanied byan elevation in glutathione
and phase II GST enzyme in HepG2 cells [85]. Another anticancer mechanism lies in the
anti-inflammatory effects of TQ through inhibiting or modulating cyclooxygenase-2 (COX-
2) enzyme, which is known to catalyze the formation of the proinflammatory mediators,
prostaglandins (PGs). TQ was reported to inhibit COX-2 and PGE in colorectal cancer [105]
and cholangiocarcinoma cell lines [80].
TQ vs. angiogenesis and metastasis: TQ is thought to inhibit tumor angiogenesis by
inhibiting the expression of vascular endothelial growth factor (VEGF) and its down-
stream signaling on cholangiocarcinoma [80], stomach [106] and triple-negative breast
cancer (TNBC) cells [107]. Moreover, TQ is known to inhibit tumor migration, invasion
and consequently tumor metastasis through modulating genes, transcription factors, and
proteins responsible for facilitating the migration process [108]. A study on TNBC con-
cluded that TQ exhibited its effect through inhibiting eEF-2K-signaling pathway with its
downstream targets, Src kinase, focal adhesion kinase (FAK) and AKT following NF-kB
inhibition [109]. Other studies reported that the antimetastatic effect of TQ is exhibited
through the inhibition of PI3K/AKT/mTOR pathways, leading to the reversal of endothe-
lial mesenchymal transformation (EndMT). This results in the upregulation of endothelial
markers E-cadherin and cytokeratin-19, and downregulation of mesenchymal markers
N-cadherin, vimentin, Twist, Slug, Snail, ZEB1 in renal cell carcinoma [110], gastric [111],
prostate [112], cervical [113,114], bladder [115] and breast cancer [107,116]. A study on
renal cancer cells concluded that the antimetastatic effect of TQ, facilitated by autophagy, is
regulated by the AMPK/mTOR pathway [117]. Furthermore, the anti-migratory action of
TQ is also exerted by the inhibition of MMP-2, MMP-9 along with integrin B1, urokinase-
type plasminogen activator (u-PA), fibronectin, RhoA, and suppression of the activation
of JNK, p38, EGFR/IKKa/b/NF-kB-signaling, paxillin and Gαi2. This was observed in
cancers like TNBC [107], lymphoma [118], glioblastoma [119], renal cell carcinoma [120],
colon [121,122] and ovarian cancers [123]. It was also reported that TQ causes down-
regulation in the TGF-β/Smad2/3-signaling pathway and reduction in CXCR4/CXCL12
pathway mediated by NF-kB expression [112,124,125]. Finally, TQ caused inhibition in
Wnt/B-catenin signaling with a consequent reduction in β-catenin target genes, Myc,
Axin-2, MMP-7, cyclin D1 and Met in bladder cancer cells [126], and inhibiting ERK1/2
phosphorylation in glioblastoma [127] and lung cancer [128].
TQ vs. gene transcription: the effects of TQ are related and can possibly be linked
together by the transcription factors involved. TQ treatment inhibits signal transducer and
activator of transcription-3 (STAT3) phosphorylation, causing downregulation of STAT3
and its related genes, Janus activated kinase-2 (JAK2), c-Src, Bcl-2, cyclin D, survivin, VEGF
and caspases 3, 7 and 9 in gastric cancer [129]. The downregulation of the PI3K/Akt path-
way and NF-kB leads to changes in various related genes, NF-kB p65, GSK3B, XIAP, Bcl-2,
VEGF, COX-2, PGE2, inducing apoptosis and inhibiting metastasis in cancer cells [130–132].
Another TQ-affected signaling pathway is the tumor necrosis factor (TNF) pathway, which
usually induces NF-kB-signaling, hence modulating apoptosis and inflammation. This path-
way was found to be modulated following the treatment of HeLa cells with TQ [133].
Following NF-kB inhibition, NLRP3 inflammasome is inhibited, causing inactivation of
caspase-1 and inhibition of IL-1 and IL-18 in melanoma cells [134]. Moreover, TQ binds
to PAK1, causing conformational changes, affecting RAF/MEK/ERK1/2-signaling path-
way [105]. Figure 1 summarizes the signaling pathways contributing to carcinogenesis and
how TQ inhibits them through its anticancer mechanisms.
Cells 2021, 10, 302 7 of 29
Cells 2021, 10, x FOR PEER REVIEW 7 of 30

Figure 1. Molecular mechanisms of TQ in cancer development (A) pathways involved in cancer development; (B) path-
Figureways
1. Molecular mechanisms of TQ in cancer development (A) pathways involved in cancer development; (B) path-
inhibited by TQ that appear transparent in the figure while opaque molecules are activated.
ways inhibited by TQ that appear transparent in the figure while opaque molecules are activated.
TQ as a chemo-adjuvant: adding to the previously mentioned anticancer effects of TQ,
TQ asstudies
several a chemo-adjuvant: adding to effect
reported the potentiating the previously
of combining mentioned anticancer effects of
TQ with chemotherapeutic
TQ,agents
several
andstudies reported
radiation. the potentiating
It was shown effect
to enhance their of combining
anticancer activity,TQ with chemo-
causing chemothera-
peutic agents and radiation.
or radio-sensitization, It was[135–137].
respectively shown to It enhance
also reducedtheir
oranticancer
attenuated activity,
some treat-causing
chemo- or radio-sensitization,
ment-induced toxicities, such asrespectively
nephropathy[135–137]. It also reduced
or hepatotoxicity, seen with or attenuated
cisplatin treat- some
ment [138–141]. Table
treatment-induced 3 discusses
toxicities, suchsome of the chemo-modulating
as nephropathy effects ofseen
or hepatotoxicity, TQ in combi-
with cisplatin
nation with known anticancer drugs. However, as with any other drug,
treatment [138–141]. Table 3 discusses some of the chemo-modulating effects of TQ in further studies
should be done
combination withtoknown
investigate the standardization
anticancer of TQ administration
drugs. However, as with anywhen otherused with
drug, further
cancer drugs, as literature showed that CYP1A2, CYP2C9, CYP2D6 and CYP3A4
studies should be done to investigate the standardization of TQ administration when used were
subject to inhibition by TQ, hinting at various potential drug–drug interactions [142].
with cancer drugs, as literature showed that CYP1A2, CYP2C9, CYP2D6 and CYP3A4 were
subject to inhibition by TQ, hinting at various potential drug–drug interactions [142].

Table 3. The chemomodulatory/chemosensitizing and protective effects of thymoquinone in combination with chemothera-
peutic agents.

Drugs Effects Mechanism Experimental Model Reference


Downregulating Twist1
Breast cancer T47D and
Paclitaxel Chemomodulatory Increase apoptotic cell death and [143]
MCF-7 cells
autophagy
Blocking PI3K/AKT pathways
Prostate cancer DU145
Docetaxel Chemomodulatory Enhance Bax, Bid, caspase 3 and [144,145]
and C4-2B cells
inhibiting Bcl-XL
Upregulate PTEN Inhibiting AKT
Breast cancer MCF-7
Chemomodulatory Increasing Bax/Bcl2 ratio [146,147]
cells
Increase ROS formation
Doxorubicin
ROS generation T cell leukemia cells
Chemomodulatory and
Higher mitochondrial disruption Lymphoblastic [148–150]
protective
Maintain cardiac myocyte survival leukemia
Cells 2021, 10, 302 8 of 29

Table 3. Cont.

Drugs Effects Mechanism Experimental Model Reference


Inactivating AKT, XIAP, bcl-xl and
Bcl-2
Chemomodulatory and Activating caspase-3, Bax, cytochrome
Tamoxifen Breast cancer cells [151–153]
protective c and p27
Hepatoprotective via antioxidant and
anti-inflammatory actions
Downregulate Bcl-2, Wnt/β-catenin,
NF-kB, Cox2, iNOS, VEGF and Colorectal cancer stem
5-fluorouracil Chemomodulatory PI3k/AKT cells [154–156]
Upregulate Bax, Bcl-2, TGF-β1 and Gastric cancer cells
Samd4
Block JAK2/STAT3 pathway
Inhibition of PI3K/AKT pathway Esophageal cancer cells
Chemomodulatory and Activating Bax, cytochrome c and Gastric cancer cells
Cisplatin [138,139,157–160]
protective caspases Oral squamous cell
Reduced hepatorenal toxicity through carcinoma
antioxidant effect

5. Thymoquinone’s Double Hits in COVID-19 Infected Cancer Patients


First of all, a recent study showed that TQ could work as an antiviral against SARS-
CoV-2 since it revealed that TQ mighthave inhibitory activities against its viral protease in
silico [161], and several other studies are also working on proving the same effect through
insilico computational analysis. This rather interesting observation prompted the analysis
of TQ’s reported roles in regulating proteins that are involved in COVID-19 as well as in
cancer pathogenesis. This review, therefore, aims to summarize in the following sections
the common protein targets that are subject to regulation by TQ and whose roles are
furthermore crucial in COVID-19 cancer patients.
Heat shock protein A5 (HSPA5)/GRP78 is also known as immunoglobulin heavy
chain-binding protein (BiP) or glucose-regulated protein (GRP78). GRP78 overexpression
can occur under a variety of stressful conditions. This overexpression can cause the protein
to be, in turn, highly abundant on the cell surface, where it can further potentiate the
stressful conditions under which its expression was increased. For instance, overexpression
of GRP78 on the cell membranes can increase viral entry via recognition and binding to
the substrate-binding domain (SBD) of GRP78. This is true for several viruses [162], and
molecular docking also showed that SARS-CoV-2 spike protein could bind to the host
cell surface GRP78 [163]. Furthermore, the literature confirmed the in vitro presence of
GRP78 protein in airway epithelial cells and in situ protein expression of GRP78 in the
respiratory mucosa [164]. Higher serum GRP78 concentrations were found in COVID-19
patients compared to patients with pneumonia and the control group [165]. In another
example, GRP78 can be overexpressed in the setting of a malignancy, where its abundance
on cancer cell membranes endows antiapoptotic properties to the tumor cell by increasing
the levels of antiapoptotic proteins such as Bcl-2 and reducing the levels of proapoptotic
proteins, such as Bax. Ultimately, this results in promoting tumor survival, progression,
angiogenesis, invasion, metastasis, as well as resistance to therapy [166]. This has been
observed in various types of cancers [167–171].
HSPA5/GRP78 and TQ: TQ decreases the expression of HSPA5/GRP78 and improved
mitochondrial function [172]. Furthermore, molecular docking showed that TQ might in-
terfere with SARS-CoV-2 attachment to HSPA5 by tightly binding to this protein on the cell
surface [173]. Collectively, TQ produces a dual hit, where it tightly binds to HSPA5/GRP78,
reducing its expression. Thus both reduces the risk of SARS-CoV-2 infection and decrease
chemotherapy resistance, cancer invasion, metastasis, and survival, as illustrated briefly in
Figure 2 [173].
CoV-2 infection, inhibiting NF-ĸB[205], thus ameliorating the COVID-19-associated cyto-
kine storm and inflammation. This would indeed also be beneficial in cancer patients who
suffer from elevated cytokines and inflammation. On the other hand, Nrf2 appears to have
contrasting roles in cancer patients, where it protects against chemical and radiation-in-
duced carcinogenesis [180–182] but also leads to tumor-promoting effects. It would be
Cells 2021, 10, 302 beneficial to investigate the effects of TQ on Nrf2/HO-1/NF-ĸB in cancer patients with 9 of 29
COVID-19.

Figure 2. Effect of TQ on Nrf2, GRP78 and SARS-CoV-2 infection. A diagram showing the effect of TQ on Nrf2; it activates
Nrf2 by phosphorylation, causing its translocation inside the nucleus, binding to ARE (antioxidant response element) and
Maf. This binding results in the reduction of NF-kB, cytokine production, inflammation, oxidative damage and an increase
Figure 2. Effect of TQ on Nrf2, GRP78 and SARS-CoV-2 infection. A diagram showing the effect of TQ on Nrf2; it acti-
in detoxifying cytoprotective genes and enzymes such as the HO-1 enzyme. Moreover, TQ decreases GRP78 expression,
vates Nrf2 by phosphorylation, causing its translocation inside the nucleus, binding to ARE(antioxidant response element)
angiotensin-converting enzyme
and Maf. This binding 2 (ACE)-receptor
results expression
in the reduction of and hence
NF-kB, cytokine decreases
production, viral entry.oxidative damage and an
inflammation,
increase in detoxifying cytoprotective genes and enzymes such as the HO-1 enzyme. Moreover, TQ decreases GRP78 ex-
Nrf2
pression, angiotensin-converting (nuclear
enzyme factor erythroid-derived
2 (ACE)-receptor 2 related
expression and hence factor
decreases viral2) is an important transcription
entry.
factor that counteracts oxidative stress, where it acts as a sensor of oxidative stress, pre-
venting genomic instability. It regulates about 250 genes involved in cellular homeostasis,
including detoxifying enzymes, antioxidant proteins, and cytoprotective proteins [174].
Under normal physiological conditions, Nrf2 is sequestered by the cytoplasmic keap1
(Kelch-like ECH-associated protein 1), which maintains the Nrf2 at low levels through
targeting it for proteasomal degradation (ubiquitination) [175]. Disruption of Nrf2 home-
ostasis can be seen in various settings, including viral infection and cancer. During a viral
infection, intracellular expression of viral proteins leads to an increase in the oxidative stress
of a cell. This leads to the dissociation of Nrf2 from keap1, which consequently prevents
its ubiquitination [176,177]. Nrf2 is now able to translocate to the nucleus and activate the
transcription of detoxifying, cytoprotective genes such as heme oxygenase (HO-1) [178,179].
This mechanism is also involved in the setting of cancer, where Nrf2 is additionally able
to protect a cell from chemical and radiation-induced carcinogenesis [180–183]. Moreover,
literature revealed that Nrf2 is able to enhance innate immune system activity, as well as
participate in the inhibition of inflammatory cytokine expression, including IL-1β, IL-6,
and NF-kB, ultimately decreasing inflammation [184–186]. Although some studies show
that the continuous activation of Nrf2, as a result of excessive levels of ROS, could have
deleterious effects on the host cell [187,188], it usually does not happen in viral infections
since the virus needs to keep optimal oxidative stress levels allowing it to maintain viral
metabolism without causing death in the host cell [189]. A recent study in 2020 involving 40
patients showed that the severity of COVID-19 is inversely associated with Nrf2 expression
and directly linked to age and intensity of inflammatory response [190]. Interestingly,
recent studies showed that Nrf2 deficiency upregulates ACE2 receptors, while activation
of Nrf2 downregulates ACE2 receptors, with Nrf2 knock-out mice showing enhanced
ACE2 expression. In cultured immortalized renal proximal tubule cells, treatment with
Nrf2 inhibitor (Trigonelline) or transfection with Nrf2 small interfering RNA led to an
increase in ACE2 transcription [191]. The exact mechanism of how Nrf2 downregulates the
Cells 2021, 10, 302 10 of 29

ACE2 receptor remains unclear. Additionally, Nrf2 activators downregulate the mRNA
expression of TMPRSS2 [192,193] via upregulating TMPRSS2 inhibitors PAI-1 plasminogen
activator inhibitor-1 encoded by the SERPINE gene (SERPINE/PAI-1) [192] and secretory
leukocyte protease inhibitor (SLPI) [193]. This may highlight the important role of Nrf2
in downregulating TMPRSS2, ACE2 receptor expression and subsequently decreasing
SARS-CoV-2 infection load. Despite the numerous cytoprotective mechanisms of Nrf2, it
still appears to be a double-edged molecule since there is cumulative evidence establishing
the fact that Nrf2 is one of the pathways that drive cancer progression, spread or metastasis,
and chemo-resistance [194–203], and hence further dedicated studies may still be required.
Nrf2 and TQ: in keratinocytes, TQ works as a pro-oxidant that activates Nrf2 and
increases its nuclear accumulation, leading to increased mRNA and protein expression of
HO-1 [204], enhancing its binding to ARE, thus decreasing ACE2 expression, as demon-
strated in Figure 2 [205]. During the course of COVID-19, white blood cells become acti-
vated, releasing excessive, prolonged inflammatory cytokines, leading to what is called the
cytokine storm, acute respiratory distress syndrome, multiple organ dysfunction, and even
death [206,207]. In addition, SARS-CoV-2 might invade the central nervous system induc-
ing neurological diseases [208] manifested as headache, nausea, and vomiting. TQ showed
inhibitory effects on NF-kB mediated neuroinflammation, as mentioned in Table 4, report-
ing other TQ effects in COVID-19-induced neurological disease. Altogether, TQ activates
Nrf2 [204] that decreases ACE2 expression [205], potentially decreasing SARS-CoV-2 infec-
tion, inhibiting NF-kB [205], thus ameliorating the COVID-19-associated cytokine storm
and inflammation. This would indeed also be beneficial in cancer patients who suffer
from elevated cytokines and inflammation. On the other hand, Nrf2 appears to have con-
trasting roles in cancer patients, where it protects against chemical and radiation-induced
carcinogenesis [180–182] but also leads to tumor-promoting effects. It would be beneficial
to investigate the effects of TQ on Nrf2/HO-1/NF-kB in cancer patients with COVID-19.

Table 4. The beneficial effects of TQ against COVID-19 pathophysiological effects.

COVID-19
Thymoquinone References
Complications
-Protects against hyperlipidemia, cyclophosphamide, doxorubicin and diesel
exhaust particle-induced cardiac damage or changes
Cardiac damage -Counteracts sepsis-induced elevation in p62 and increases beclin-1 expression [21,48,209–212]
-Reduced infarct size, improved cardiac function, reduced myocardial enzyme LDH
and CK-MB activities and inhibited oxidative stress
-Inhibited nuclear factor kappa-B (NF-kB) in lung tissue
-Ameliorates the pathological changes of chronic asthma, inhibits inflammatory
changes by antagonizing IL-4/5, and a single dose prevented asthma in the guinea
pig model
-Showed anti-neoangiogenic effects through inhibition of expression of
VEGFR2/PI3K/Akt-signaling pathway
-Decreased the inflammatory and apoptotic index levels in rats exposed to cigarette
smoke and showed anti-inflammatory and cytoprotective effects against cigarette
smoke-induced COPD and LPS induced acute lung injury by COX inhibition
-Downregulated pro-fibrosis genes and decreased oxidative stress in lung fibrosis
Lung injury, ARDS,
-Counteracted emphysema in air alveoli, lymphoid hyperplastic cells activation
COPD, pulmonary [22,213–223]
surrounding the bronchioles, inflammatory cell infiltration
fibrosis
-Ameliorated pulmonary arterial hypertension, Induced relaxation of the
precontracted pulmonary artery and caused a decrease in the tension of pulmonary
arterial rings precontracted with phenylephrine
-Reduced alterations in lungs and inflammatory markers induced by
cyclophosphamide and toluene, decreased lipid peroxidation and restored
antioxidants
-Showed inhibitory effects on the aggregation of inflammatory cells in
bronchoalveolar lavage (BAL) fluid and in lung tissues
-Significantly decreased serum IgE and showed superior inhibitory effects on iNOS
and TGF-β1
Cells 2021, 10, 302 11 of 29

Table 4. Cont.

COVID-19
Thymoquinone References
Complications
-Effectively improved the plasma and liver antioxidant capacity and enhanced the
expression of liver antioxidant genes of hypercholesterolemic rats
-Protects against oxidative liver damage and ductular proliferation
-Reduced liver injury and tumor markers expressions;thus its beneficial in the treatment
of hepatocellular carcinoma
-Protected against cyclophosphamide, tamoxifen, cypermethrin, cadmium, anti-TB
Liver injury, drugs, aflatoxinB1, acetaminophen and paracetamol-induced
[20,23,24,53,153,
elevated liver hepatotoxicity/necrosis/cirrhosis and fibrosis by decreasing the elevated ALT, AST and
224–232]
enzymes ALP, enhanced regeneration after tissue damage, decreased oxidative protein damage
and increased SOD expression and GSH
-Ameliorated liver fibrosis via blocking TLR4 expression and PI3K phosphorylation on
the activated hepatic stellate cells
-Reduced thioacetamide-induced liver fibrosis and inflammation by decreasing
proinflammatory cytokines, inhibiting PI3K phosphorylation and enhanced p-AMPK
and liver kinase B (LKB-1)
-Attenuated oxidative stress and inflammation-reducing renal ischemia-induced
damage and several drugs-induced nephrotoxicities
-Reversed the vancomycin and doxorubicin-induced nephrotoxicity, reversed elevated
serum blood urea nitrogen, creatinine, MDA and urinary albumin excretion [24,25,226,233,
Kidney damage
-Protected kidney against urinary tract infections (Escherichia coli) induced 234]
pyelonephritis
-Showed protective action against cypermethrin induced shrinkage of glomeruli and
necrosis of renal tubules in kidneys (in mice)
-Induced a significant increase in expression of neuroprotective proteins such as
biliverdin reductase-A, 3-mercaptopyruvate sulfurtransferase, glutaredoxin-3, and
mitochondrial Lon protease, a significant decrease in expression of inflammatory
cytokines, and NF-κB pathway
-Decreased malondialdehyde and nitric oxide metabolites in the brain
Neurological -Enhanced the thiol content and superoxide dismutase and catalase activities and serum
disease, cognitive T4 level [51,235–237]
decline -Protected against PTU-induced memory impairments in rats
-Reversed learning and memory impairments, brain tissue-oxidative damage in
hypothyroid juvenile rats and alleviated changes in the hippocampal lipid peroxide
level and SOD and AChE activities
-Have a protective effect on learning and memory function. It significantly increased the
expression of Nrf2, HO-1 proteins and SOD in the hippocampus
-Showed significant antimicrobial activity against anaerobic bacteria, thus can be used
against diarrhea
-Prevented and significantly reduced the appearance of diarrhea and body weight loss
in mice and ameliorates dextran sulfate sodium-induced colitis
-Inhibited proton pump, acid secretion and neutrophil infiltration, while enhancing
GIT [18,26,44,45,238]
mucin secretion, reduces ulcer index, markers of oxidative stress and nitric oxide
production
-TQ+ ranitidine showed synergistic effects and a more significant decrease in ulcer index
-Suppressed spontaneously contracting rabbit jejunum and also relaxed high K induced
contractions in the jejunum and guinea-pig ileum, thus can be used in colic and diarrhea
-Inhibited histamine production
Allergy, allergic -Decreases the symptoms of allergic conjunctivitis almost the same as dexamethasone
[239,240]
conjunctivitis -Suppressed the Total IgE, OVA-specific IgE and Recruitment of Inflammatory cells
-Alleviated allergic inflammation and may be valuable for treating allergic rhinitis
-Decreased lipase\amylase ratio
Acute pancreatitis -Decreased oxidative stress markers, pancreatic 4-hydroxynonenal (4-HNE), protein
[16,241]
and elevated lipase carbonyl content and protected pancreatic acinar cells from oxidative stress
-Attenuated GSH depletion and increased the activity of GST
Abbreviations: LDH: lactate dehydrogenase; CK-MB: creatininekinase-MB; ARDS: acute respiratory distress syndrome; COPD: chronic
obstructive pulmonary disease.
Cells 2021, 10, 302 12 of 29

The Renin-Angiotensin System (RAS) is a homeostatic loop that begins when the
hepatic angiotensinogen is converted into angiotensin I (ATI) by the renal renin enzyme.
This loop then involves two arms; the ACE enzyme (from the lungs) converts ATI to
angiotensin II (ATII), increasing its circulating levels. This mediator is implicated in
vasoconstriction, fibrosis, hypertension, and inflammation. The second arm involves
the conversion of ATII to AT1-7 by ACE2, which carries out the opposite effects of its
precursor, ATII [242,243]. Many of the cardiovascular symptoms, as well as multiple
organ damage seen with COVID-19, can be linked to RAS-induced dysregulation [244].
In fact, patients with COVID-19 tend to have significantly higher levels of circulating ATII
compared to healthy individuals, and the findings of various studies that were conducted
on autopsies [245] suggest that SARS-CoV-2 may cause downregulation of ACE2 receptors.
This finding, along with receptor internalization upon viral binding, further contribute to
the increase in ATII levels and the imbalance between the pro- and anti-inflammatory roles
of the RAS, ultimately causing the proinflammatory role of the RAS to predominate [13],
endowing procoagulant properties to endothelial cells [245].
The increased ATII enhances the activity of NADPH oxidase, increasing ROS, leading
to induction of vascular smooth muscle cell (VSMC) proliferation, migration, atheroscle-
rosis [246], and hypertension via increased oxidative stress, inflammatory reactions and
production of vasoconstrictors while decreasing the vasodilator nitric oxide (NO). Thus, in-
hibition of VSMC proliferation and migration would be beneficial in preventing atheroscle-
rosis and avoiding cardiovascular complications [247]. Peroxisome proliferator-activated
receptor γ-coactivator 1α (PGC-1 α) was shown to attenuate ROS generation, thereby lim-
iting ATII-induced rat VSMC proliferation [248]. Moreover, AMPK activation attenuates
ATII induced VSMC proliferation, and PPARγ has been reported to have antiproliferative
effects in multiple cancers [249,250]. The ramifications of the prolonged basal inflamma-
tory state observed in the setting of malignancy can be closely mirrored in other statesof
inflammation, including hypertension, obesity, diabetes, and even in COVID-19 patients
with the aforementioned comorbidities. This surge in inflammatory cytokines increases
the production of local and systemic ATII [251]. Increased levels of ATII in cancer patients
has been associated with shortened survival [252]. It is therefore hypothesized that two
hits contribute to the hyper-inflammatory response that is observed in COVID-19 patients
with comorbidities; the first hit was resulting from the chronic state of inflammation, which
increases the activity of the ATII proinflammatory axis, and the second hit resulting from
SARS-CoV-2 itself, via the internalization of ACE2 receptors, and the decrease in the activity
of the anti-inflammatory ACE2 axis [253]. Together, these two hits increase tissue damage
as well as the risk for multiple organ failure.
TQ and RAS/ATII: the effect of TQ on ATII can be seen when TQ (40 mg/kg) is
injected intraperitoneally 30 min before ATII (300 mg/kg) intravenous injection. This
significantly blunted the rise in systolic blood pressure, mean arterial pressure and heart
rate that wasinduced by ATII. Thus, the antagonistic effect of TQ on ATII appears to be
beneficial in both COVID-19 patients and cancer patients [254]. TQ markedly inhibited ATII-
induced VSMC proliferation, migration, and oxidative stress. It also reversed the elevated
NADPH oxidase activity and ROS induced by ATII [255]. In addition, TQ enhanced the
expression of p-AMPK, PPARγ, and PGC-1 α that were inhibited by ATII. These particular
effects of TQ were abolished by AMPK inhibitor or PGC-1 α siRNA transfection [255].
Furthermore, TQ reduced oxidative stress, lipid peroxidation (specifically renal MDA),
renal TNF-α in unilateral ureteral obstruction (UUO), and increased antioxidant enzymes
(SOD, catalase). Meanwhile, it significantly decreased obstruction-induced apoptotic
cell death and decreased ATII renal expression [256]. Thus, collectively, TQ improves
renal oxidative damage, apoptotic cell death, TNF-α expression and also prevents the
upregulation of ATII in UUO injured rats [257]. Looking at the bigger picture, TQ can be
a promising drug in ameliorating COVID-19-induced RAS dysregulation, counteracting
multiple organ failure in COVID-19 cancer patients.
Cells 2021, 10, 302 13 of 29

6. The Significance of Thymoquinone in Other Pathological Processes in


COVID-Cancer Patients
Platelet hyper-reactivity, the novel coronavirus, promotes a state of hypercoagula-
tion [13] and additionally predisposes some patients to thromboembolic events through
altering platelet function. Literature showed that resting platelets from COVID-19 patients
had increased basal P-selectin expression, aggregated faster, and showed increased spread-
ing on both fibrinogen and collagen. The increase in platelet activation and aggregation
could partially be attributed to increased MAPK pathway activation and thromboxane gen-
eration [258,259]. Interestingly enough, TQ is able to ameliorate platelet hyper-reactivity
by suppressing COVID-19-induced MAPK activation, and another study showed that TQ
triggers apoptosis of blood platelets in a PI3K-dependent manner. Hypercoagulation in
COVID-19 is typically characterized by elevated levels of fibrinogen and D-dimer, increased
prothrombin time (PT), activated partial thromboplastin time, mild thrombocytopenia,
elevated factor 8 and Von Willebrand Factor [12]. D-dimer levels are especially increased
in critically ill patients who are administered into the ICU and is used as a prognostic
factor [13]. Evidence suggests that SARS-CoV-2-induced release of proinflammatory cy-
tokines of the innate immunity, either in close proximity to the affected organ (alveolar
cells of the lung) or in the bloodstream, represents the culprit of the inflammatory pro-
cess [12,206]. Concerning cancer patients, studies showed that multiple myeloma (MM)
patients have an increased risk for venous thromboembolism (VTE) [258,259]. In this re-
gard, TQ has been shown to reverse cancer-associated thrombosis and thus can be used as
a preventive anti-coagulant and/or as a supplement to existing chemotherapies, producing
a double protective effect in COVID-19 infected cancer patients, counteracting the viral-
and cancer-induced platelet hyperreactivity [260–262].
With regard to inflammation and multiple organ failure; treatment approaches of COVID-
19 that aim at targeting the viral replication cycle may not be enough for promoting host
survival, as it is also necessary to target the virus-induced cytokine release syndrome
(CRS) that leads to multiple organ damage and ARDS [263]. Some of the proinflammatory
cytokines that are locally and systemically elevated include IL-1β, interferon-γ (IFN-γ),
IL-1, IL-6, and monocyte chemoattractant protein 1 (MCP-1). They have been detected
in critically ill patients, and they were found to correlate to disease severity [264,265].
Furthermore, upon SARS-CoV-2 entry into the host cell, the disruption of the intracellular
environment via ion redistribution results in the activation of NLRP3 inflammasome that
increases the secretion of proinflammatory cytokines: IL-1β, IL-18, IL-6 and TNF, further
contributing to the imminent cytokine storm and tissue inflammation during respiratory
illness [266]. In this context, TQ could have a promising anti-inflammatory role, where
it significantly decreases the mRNA expression of the aforementioned proinflammatory
cytokines, IL-18, IL-1β and TNF-α, and inhibits IL-6-signaling [16,17,267–269]. In cancer
patients, inflammasomes are also overexpressed, leading to an increased inflammatory
status [270,271]. The disastrous effect of NLRP3 inflammasome in melanoma is owed
to its suppression of NK cell activation, necessary for the release of IFN-γ and killing
of tumor cells, therefore, ultimately increasing lung metastasis [272,273]. Closely exam-
ining the molecular mechanisms of inflammasome complexes in different cancer types
revealed that the overexpression of IL-1β causes mobilization of myeloid-derived sup-
pressor cells (MDSCs) and induces gastric cancer [274], while signaling of IL-1 drives
the accumulation of MDSCs, promoting primary and metastatic mammary tumors [275].
Moreover, the inflammatory mediator IL-18 is known to accumulate in cancer patients with
the ability to fine-tune the activation status of NK cells, depending on the amount of IL-
18 [12,276]. It is worth noting that inflammasomes do not always show tumor-promoting
effects; in fact, they showed protective roles in some cancer types [277]. Since TQ is an
NLRP3 inhibitor, this would consequently decrease the secreted IL-1β, IL-18 and IL-6 and
ameliorate pain and inflammation in COVID-19 cancer patients. In addition to NLRP3,
eicosanoids play a critical role in the progression of inflammation, fever, allergies, can-
cer, and pain [206,273,275]. Although the role of eicosanoids in COVID-19 is not well
Cells 2021, 10, 302 14 of 29

characterized, it is believed that cell debris is able to initiate an eicosanoid and cytokine
storm in the context of inflammatory diseases. TQ is also able to target arachidonic acid
metabolism [278]. Targeting the upstream portion of the eicosanoid storm will potently
inhibit the cytokine storm formation, protecting against inflammation-mediated multiple
organ damage in COVID-19 patients.
Oxidative stress: oxidative damage is an important aspect in which COVID-19 has a
notable impact on cancer patients. The analysis showed that interactions between viral
proteins and some of the components of the ROS pathway had been implicated in different
cancer types. These interactions may be exploited by SARS-CoV-2 to disrupt the basic pro-
cesses that modulate mitochondrial respiration, metabolism, and oxidative stress, including
certain pathways that are known to be important for the development of tumors [279].
TQ happens to demonstrate potent antioxidant properties, where it significantly attenuates
glutathione (GSH) depletion and increases the activity of the glutathione-S-transferase
(GST) enzyme. It significantly decreases oxidative stress markers, including thiobarbituric
acid reactive substance (TBARS), which is elevated in several cancer types like colon and
cervical cancer [280,281]. Furthermore, TQ induces the expression of several detoxifying
enzymes, including glutathione reductase, superoxide dismutase 1 (SOD1), catalase, and
glutathione peroxidase 2 (GPX) [16,19,20]. The reduction of oxidative stress, therefore,
gives a protective effect against multiple organ failure associated with COVID-19 and
cancer patients.
Myocardial injury: cardiac injury is significantly associated with fatal outcomes in
COVID-19. In sepsis, elevated plasma levels of troponin-T (c-Tnt) are an important indicator
of cardiac damage. Severe cases of COVID-19 showed elevated plasma levels of c-Tnt
and were in higher need of mechanical ventilation compared to those with normal Tnt
levels. They also had more frequent malignant arrhythmias and needed glucocorticoid
therapy [282]. Increased inflammatory cytokines can directly or indirectly cause cardiac
damage, but the precise mechanism of myocardial dysfunction-induced sepsis is still
unclear. This is also common in cancer patients, where cardiac injury is a risk factor
for mortality and may be indicated by elevated levels of troponin and BNP biomarkers,
which could result from either the malignancy or the treatment regimen [13]. For example,
certain anticancer medications cause patients to suffer from cardiotoxicity presenting as
ischemia or vasospasm. An example of this is CAR-T therapy, which may result in CRS,
and proteasome inhibitors, which may cause heart failure [283]. Literature showed that TQ
significantly decreased TnT levels and markedly reduced cardiac tissue-inflammatory cell
infiltration in the cecal ligation and puncture (CLP)-induced-sepsis group. Additionally,
P62 and beclin1 are two major proteins in autophagy, and previous literature showed
that P62 expression is increased, while beclin1 is decreased in cardiac damage and in
sepsis [284–286]. TQ significantly decreased P62 and increased expression of beclin1 in
CLP mice, thus decreasing sepsis-induced cardiac damage.
Lung injury: COVID-19 causes respiratory symptoms including breathlessness, pneu-
monia, severe ARDS, lung fibrosis and injury [287]. Studies show that TQ has promising
effects in lung protection; it protects against injury and fibrosis induced by LPS, toluene,
and cyclo-phosphamide. It also induces the relaxation of the precontracted pulmonary
artery [213,214,216–220]. Moreover, TQ inhibits inflammation, neoangiogenesis, and vas-
cular remodeling in various asthmatic models [215,221–223]. It also counteracts several
processes, including emphysema in air alveoli, activation of hyperplastic lymphoid cells
surrounding the bronchioles, and inflammatory cell infiltration [216]. According to Jaber
S. Alqahtani et al., 2020, chronic obstructive pulmonary disease (COPD) patients were at
a higher risk of more severe COVID-19, especially in smokers, as they were seen to have
higher expression of ACE2 in their airways [288,289]. TQ decreases the inflammatory and
apoptotic index levels in rats exposed to cigarette smoke and shows anti-inflammatory and
cytoprotective effects, as summarized in Table 4 [22].
Cells 2021, 10, 302 15 of 29

Liver injury: liver injury is a critical, life-threatening COVID-19 complication [290].


It is common in cancer patients, as some chemotherapeutic agents like cyclophosphamide
[291,292] and tamoxifen cause hepatotoxicity [293], as reviewed in De La Vega et al. [294].
This demonstrates the potentially fatal ramifications of COVID-19 in cancer patients.
TQ shows several promising hepatoprotective effects against toxicities and fibrosis
[20,23,24,53,224,226–228,230–232], where it protects against chemotherapy-induced hep-
atotoxicity through reducing liver injury and tumor markers expression, hinting atthe
use of TQ in the treatment of hepatocellular carcinoma [153,225,229]. Altogether, us-
ing TQ provides a dual benefit, where it protects against both COVID-19-induced and
chemotherapy-induced hepatotoxicity.
Kidney injury: this is another COVID-19 related serious complication as reviewed
in [295]. Certain chemotherapeutic regimens make cancer patients more susceptible to
nephrotoxicity [296]. TQ shows protective effects in the kidneys through ameliorating
oxidative stress and reversing the nephrotoxic effects of various drugs and infections
[24,25,226,233,234], as summarized in Table 4. Moreover, TQ shows reno-protective ef-
fects in sepsis-induced AKI. Since AKI is majorly mediated by dysregulated activation of
inflammasomes and proinflammatory cytokines, the aforementioned anti-inflammatory
properties of TQ also appear to be of significant benefit in this context [17], where TQ de-
creases apoptosis of kidney cells and alleviates AKI. Finally, NFκB is the main transcription
regulator of inflammatory genes and has crucial roles in inflammation and the pathophys-
iology of sepsis. TQ reverses the increased NFκB expression in the kidney from septic
mice. Collectively, TQ contributes to the alleviation of sepsis-induced AKI by regulating
pyroptosis, proinflammatory cytokines, and apoptosis-related expression [17].
Neurologic/cognitive manifestations associated with COVID-19: these manifestations in-
clude headache, memory loss, mood changes, vision changes, hearing loss, loss of smell,
loss of taste, impaired mobility, limb numbness, tremor, fatigue and myalgia [297]. This is
along with cases of encephalitis, necrotizing hemorrhagic encephalopathy, stroke, and
epileptic seizures as reviewed in [298]. TQ shows several neuroprotective effects, as sum-
marized in Table 4 [191,236].
TQ and gastrointestinal (GIT) manifestations associated with COVID-19: these include
diarrhea, loss of appetite, nausea/vomiting and abdominal pain as reviewed in [299].
TQ shows a promising effect against anaerobic bacteria, thus would alleviate diarrhea.
It also acts as a gastroprotective agent, proton pump inhibitor and enhances mucin se-
cretion, which would be additionally beneficial in cancer patients suffering from GIT
symptoms [18,26,44,45,238].
TQ and ocular manifestations and conjunctivitis associated with COVID-19 [300]: literature
showed that TQ decreases the symptoms of allergic conjunctivitis nearly as effectively as
dexamethasone [239], possibly ameliorating COVID-19 associated conjunctivitis.
Pancreatitis: pancreatic inflammation, along with increased lipases associated with
COVID-19, can be reduced with TQ since it has been shown to decrease lipases and protect
the pancreas against oxidative stress [16,241].
Collectively, TQ showed protective effects in the heart, lungs, kidney, liver, GIT, pancreas
and alleviated oxidative stress as summarized in Figure 3 and reported in detail in Table 4.
Cells 2021, 10,
Cells 2021, 10, 302
x FOR PEER REVIEW 16 of
17 of 29
30

Figure 3. The multifunctional effects of TQ on COVID-19-associated pathophysiology. The diagram summarizes the
Figure 3. The multifunctional effects of TQ on COVID-19-associated pathophysiology. The diagram summarizes the
main organ-related and organ protective effects of TQ on the main body organs. In the heart, TQ decreases cardiac enzymes,
main organ-related and organ protective effects of TQ on the main body organs. In the heart, TQ decreases cardiac en-
cholesterol, triglycerides
zymes, cholesterol, and inflammatory
triglycerides cytokines.
and inflammatory In the In
cytokines. liver,
theTQ increases
liver, antioxidant
TQ increases enzymes
antioxidant and decreases
enzymes and de-
proinflammatory genes and
creases proinflammatory cytokines,
genes elevated liver
and cytokines, enzymes
elevated liver and liver fibrosis.
enzymes and liverIn fibrosis.
the pancreas,
In theTQ increasesTQ
pancreas, antioxidant
increases
capacity by increasing
antioxidant capacity byglutathione-S-transferase (GST) enzyme
increasing glutathione-S-transferase (GST)andenzyme
glutathione (GSH) levels
and glutathione whilelevels
(GSH) decreasing lipase and
while decreasing
oxidative
lipase andstress. In thestress.
oxidative lungs,InTQ
theinhibits
lungs, cyclooxygenase (COX) enzyme,
TQ inhibits cyclooxygenase IL-4/5
(COX) levels,IL-4/5
enzyme, NF-kB,levels,
vascular endothelial
NF-ĸB, vasculargrowth
endo-
thelial(VEGF),
factor growth transforming
factor (VEGF), transforming
growth factor-β1 growth
(TGF-β1),factor-β1
oxidative (TGF-β1), oxidativenitric
stress, inducible stress,
oxideinducible nitric oxide
synthase(iNOS) syn-
enzyme,
thase(iNOS)
lipid enzyme,
peroxidation, lipid peroxidation,
pro-fibrosis genes andpro-fibrosis
pulmonary genes
arterialand pulmonary arterial
hypertension;in hypertension;in
the kidney, TQ decreases thekidney
kidney,markers,
TQ de-
creasesurea
blood kidney markers,
nitrogen blood
(BUN), urea nitrogen
creatinine, (BUN), creatinine,
malondialdehyde (MDA) malondialdehyde
and albumin and (MDA) andantioxidant
increases albumin and increases
activity. an-
In the
tioxidant activity. In the gastrointestinal tract (GIT), TQ inhibits proton pump, acid secretion, ulcer index, neutrophil in-
gastrointestinal tract (GIT), TQ inhibits proton pump, acid secretion, ulcer index, neutrophil infiltration and oxidative stress
filtration and oxidative stress markers and increases mucin secretion and nitric oxide (NO) production.
markers and increases mucin secretion and nitric oxide (NO) production.

7. Conclusions
7. Conclusions
The clinical spectrum of COVID-19 can generally range from patients who are pauci-
symptomatic to those
The clinical experiencing
spectrum of COVID-19 severecan respiratory failurefrom
generally range and patients
ARDS, and whofinally,
are pau- to
those who suffertofrom
cisymptomatic thosesystemic manifestations
experiencing and multiple
severe respiratory organ
failure and failure.
ARDS,Cancer patients
and finally, to
are ofwho
those particular interest
suffer from in light
systemic of this pandemic;
manifestations they areorgan
and multiple morefailure.
liable Cancer
to experience
patientsa
more
are ofsevere clinical
particular course
interest inof the of
light disease, owing to multiple
this pandemic; they areconfounding
more liable to factors, such as
experience a
more severe clinical course of the disease, owing to multiple confounding
increased basal inflammatory state, increased oxidative stress, as well as the very likely factors, such as
increasedofbasal
presence inflammatory
existing comorbiditiesstate, increased
other than the oxidative stress,
malignancy as well
itself. Amidstas thethevery likely
eagerness
presence of existing
to vaccinate comorbidities
populations other than the
against COVID-19, themalignancy
aforementioneditself.points
Amidst thejustify
still eagerness to
the ra-
vaccinate populations
ther meticulous hunt foragainst
a drugCOVID-19,
or an agentthe thataforementioned
would ideally points
provide still justify the
COVID-19 rather
infected
meticulous
cancer huntwith
patients for adouble
drug orbenefits,
an agentpositively
that would ideally provide
affecting the courses COVID-19
of bothinfected can-
their malig-
cer patients with double benefits, positively affecting the courses of both
nancies and the viral infection. In this review, we have hoped to shed light on TQ from their malignancies
and thesativa
Nigella viralseeds
infection. In this
and how review,
it has been we
shownhavethroughout
hoped to shed light on
numerous TQ from
studies Nigella
to have po-
sativa seeds and how it has been shown throughout numerous studies
tential beneficial and protective effects in general and in COVID-19-infected cancer pa- to have potential
beneficial
tients and protective
in particular, effects in general
with significantly lowerand in COVID-19-infected
incidences of side effects. First,cancer patientsthe
it reduces in
particular,
probabilitywith significantly entry
of SARS-CoV-2 lowerinto
incidences of side effects.
cells. Second, it helpsFirst, it reduces
ameliorate the the probability
deleterious ef-
of SARS-CoV-2
fects of CRS in entry into cells. Second,
COVID-19-infected it helps
cancer ameliorate
patients, therebythe deleterious
protecting effectsmultiple
against of CRS
in COVID-19-infected cancer patients, thereby protecting against multiple
organ damage. Additionally, it is particularly beneficial for cancer patients because it organ damage.
Cells 2021, 10, 302 17 of 29
Cells 2021, 10, x FOR PEER REVIEW 18 of 30

Additionally, it is particularly beneficial for cancer patients because it could potentially alle-
could potentially alleviate multiple chemotherapy-induced toxicities, exhibit anticancer
viate multiple chemotherapy-induced toxicities, exhibit anticancer effects, as well as exhibit
effects, as well as exhibit chemo- and radio-sensitizing effects. Figure 4 summarizes the
chemo- and radio-sensitizing effects. Figure 4 summarizes the common molecular targets
common molecular targets and effects of TQ that are beneficial in both cancer and COVID-
and effects of TQ that are beneficial in both cancer and COVID-19 patients. Altogether, TQ
19 patients. Altogether, TQ is a promising solution that may win the battle against COVID-
is a promising solution that may win the battle against COVID-19 and cancer. While TQ is
19 and cancer. While TQ is not an FDA approved drug for the treatment of cancer or
not an FDA approved drug for the treatment of cancer or COVID-19; during the past year,
COVID-19; during the past year, onephase 3 clinical trial was done while threephase 2
onephase 3 clinical trial was done while threephase 2 clinical trials are currently studying
clinical trials are currently studying the effect of Nigella sativa extracts on immunity and
the effect of Nigella sativa extracts on immunity and clinical outcomes of COVID-19 patients
clinical outcomes of COVID-19 patients as per clinicaltrials.gov (Identifier number:
as per clinicaltrials.gov (Identifier number: NCT04347382, NCT04472585, NCT04401202
NCT04347382, NCT04472585, NCT04401202 and NCT04553705). It is clear that further
and NCT04553705). It is clear that further clinical trials investigating the use of TQ in
clinical
numerous trials investigating
cancers need to the use of TQ in numerous cancers need to be implemented.
be implemented.

Figure
Figure 4. TQ
TQ common
common effects
effects in
in SARS-CoV-2 infection and cancer.
cancer. The figure summarizes TQ’s actions on molecular
molecular
targets
targets that cause beneficial outcomes in both SARS-CoV-2 infection and cancer pathogenesis.
and cancer pathogenesis.

1.
1. Increase
IncreaseininNrf2
Nrf2that
thatdecreases
decreasesACE2
ACE2expression
expression lessening
lessening the
the viral
viral entry
entry and
and inhibits
inhibits
NF-kB
NF-kBthat
thatdownregulates
downregulatesinflammatory
inflammatory genes
genes and
and cytokine
cytokine storm
storm beneficial
beneficial in
in viral
viral
infection and cancer pathogenesis;
infection and cancer pathogenesis;
2.
2. Reduction
Reductioninininflammatory
inflammatorycytokines
cytokinesthrough
throughNLRP3
NLRP3inhibition,
inhibition,which
whichininturn
turnin-in-
hibits inflammasome formation and all inflammasome components
hibits inflammasome formation and all inflammasome components and inhibits and inhibits
COX-2
COX-2enzyme
enzymehence
hencedecreasing
decreasinginflammatory
inflammatoryPGE2 PGE2levels;
levels;
3.
3. Decrease
Decreasein inRAS
RASactivity
activityby
byattenuating
attenuatingangiotensin
angiotensinIIIIlevels
levels (ATII)
(ATII) which
which abolishes
abolishes
its
its detrimental effects such as vasoconstriction (VC), fibrosis, hypertension (HTN)
detrimental effects such as vasoconstriction (VC), fibrosis, hypertension (HTN)
and
andinflammation;
inflammation;
4. Decrease in expression of HSPA5/GRP78, which halts viral entry into the cells and
decreases cancer metastasis while enhancing apoptosis.
Cells 2021, 10, 302 18 of 29

4. Decrease in expression of HSPA5/GRP78, which halts viral entry into the cells and
decreases cancer metastasis while enhancing apoptosis.
5. Inhibition of MAPK, which decreases dysregulated platelet activity reducing risks of
clotting/coagulopathy.
6. Decrease in oxidative stress by upregulating antioxidant enzymes such as superoxide
dismutase (SOD1) and enhancing glutathione (GSH) levels.

Author Contributions: S.E., A.A.E. and N.S.A. contributed toreviewing the literature, writing the
review, and designing figures. H.M.E.T. and U.S. were responsible for designing, editing, and revising
the review. All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not Applicable.
Data Availability Statement: Not Applicable.
Conflicts of Interest: The authors declare that they have no conflict of interest.

References
1. Grant, M.C.; Geoghegan, L.; Arbyn, M.; Mohammed, Z.; McGuinness, L.; Clarke, E.L.; Wade, R.G. The prevalence of symptoms in
24,410 adults infected by the novel coronavirus (SARS-CoV-2; COVID-19): A systematic review and meta-analysis of 148 studies
from 9 countries. PLoS ONE 2020, 15, e0234765. [CrossRef] [PubMed]
2. Baj, J.; Karakuła-Juchnowicz, H.; Teresiński, G.; Buszewicz, G.; Ciesielka, M.; Sitarz, E.; Forma, A.; Karakuła, K.; Flieger, W.;
Portincasa, P.; et al. COVID-19: Specific and Non-Specific Clinical Manifestations and Symptoms: The Current State of Knowledge.
J. Clin. Med. 2020, 9, 1753. [CrossRef] [PubMed]
3. Thakkar, S.; Arora, S.; Kumar, A.; Jaswaney, R.; Faisaluddin, M.; Ammad Ud Din, M.; Shariff, M.; Barssoum, K.; Patel, H.P.; Nirav,
A.; et al. A Systematic Review of the Cardiovascular Manifestations and Outcomes in the Setting of Coronavirus-19 Disease.
Clin. Med. Insights Cardiol. 2020, 14, 1179546820977196. [CrossRef]
4. Chen, X.; Laurent, S.; Onur, O.A.; Kleineberg, N.N.; Fink, G.R.; Schweitzer, F.; Warnke, C.A. systematic review of neurological
symptoms and complications of COVID-19. J. Neurol. 2020, 1–11. [CrossRef] [PubMed]
5. Pennisi, M.; Lanza, G.; Falzone, L.; Fisicaro, F.; Ferri, R.; Bella, R. Sars-cov-2 and the nervous system: From clinical features to
molecular mechanisms. Int. J. Mol. Sci. 2020, 21, 5475. [CrossRef]
6. Wu, C.P.; Adhi, F.; Highland, K. Recognition and management of respiratory coinfection and secondary bacterial pneumonia in
patients with COVID-19. Clevel. Clin. J. Med. 2020, 87, 1–5. [CrossRef]
7. Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, Transmission, Diagnosis, and Treatment
of Coronavirus Disease 2019 (COVID-19): A Review. JAMA J. Am. Med. Assoc. 2020, 324, 782–793. [CrossRef]
8. Jin, Y.H.; Cai, L.; Cheng, Z.S.; Cheng, H.; Deng, T.; Fan, Y.P.; Fang, C.; Huang, D.; Huang, L.Q.; Huang, Q.; et al. A rapid
advice guideline for the diagnosis and treatment of 2019 novel coronavirus (2019-nCoV) infected pneumonia (standard version).
Mil. Med. Res. 2020, 7, 4. [CrossRef]
9. ElGohary, G.M.; Hashmi, S.; Styczynski, J.; Kharfan-Dabaja, M.A.; Alblooshi, R.M.; de la Cámara, R.; Mohmed, S.; Alshaibani, A.;
Cesaro, S.; Abd El-Aziz, N.; et al. The risk and prognosis of COVID-19 infection in cancer patients: A systematic review and
meta-analysis. Hematol. Oncol. Stem Cell. Ther. 2020. [CrossRef]
10. Carreira, H.; Strongman, H.; Peppa, M.; McDonald, H.I.; Dos-Santos-Silva, I.; Stanway, S.; Smeeth, L.; Bhaskaran, K. Prevalence of
COVID-19-related risk factors and risk of severe influenza outcomes in cancer survivors: A matched cohort study using linked
English electronic health records data. EClinicalMedicine 2020, 29–30, 100656. [CrossRef]
11. Vivarelli, S.; Falzone, L.; Torino, F.; Scandurra, G.; Russo, G.; Bordonaro, R.; Pappalardo, F.; Spandidos, D.; Raciti, G.; Libra, M.
Immune-checkpoint inhibitors from cancer to COVID-19: A promising avenue for the treatment of patients with COVID-19
(Review). Int. J. Oncol. 2020, 58, 145–157. [CrossRef] [PubMed]
12. Vaninov, N. In the eye of the COVID-19 cytokine storm. Nat. Rev. Immunol. 2020, 20, 277. [CrossRef] [PubMed]
13. Labò, N.; Ohnuki, H.; Tosato, G. Vasculopathy and Coagulopathy Associated with SARS-CoV-2 Infection. Cells 2020, 9, 1583.
[CrossRef] [PubMed]
14. Liang, W.; Guan, W.; Chen, R.; Wang, W.; Li, J.; Xu, K.; Li, C.; Ai, Q.; Lu, W.; Liang, H.; et al. Cancer patients in SARS-CoV-2
infection: A nationwide analysis in China. Lancet Oncol. 2020, 21, 335–337. [CrossRef]
15. Wang, H.; Zhang, L. Risk of COVID-19 for patients with cancer. Lancet Oncol. 2020, 21, e181. [CrossRef]
16. Periyanayagam, S.; Arumugam, G.; Ravikumar, A.; Ganesan, V.S. Thymoquinone ameliorates NLRP3-mediated inflammation in
the pancreas of albino Wistar rats fed ethanol and high-fat diet. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 623–632. [CrossRef]
17. Guo, L.P.; Liu, S.X.; Yang, Q.; Liu, H.Y.; Xu, L.L.; Hao, Y.H.; Zhang, X.Q. Effect of Thymoquinone on Acute Kidney Injury Induced
by Sepsis in BALB/c Mice. BioMed Res. Int. 2020, 2020, 1594726. [CrossRef]
Cells 2021, 10, 302 19 of 29

18. Ghayur, M.N.; Gilani, A.H.; Janssen, L.J. Intestinal, airway, and cardiovascular relaxant activities of thymoquinone. Evid. Based
Complement. Altern. Med. 2012, 2012, 305319. [CrossRef]
19. Elbarbry, F.; Ragheb, A.; Marfleet, T.; Shoker, A. Modulation of hepatic drug metabolizing enzymes by dietary doses of
thymoquinone in female New Zealand white rabbits. Phyther Res. 2012, 26, 1726–1730. [CrossRef]
20. Ismail, M.; Al-Naqeep, G.; Chan, K.W. Nigella sativa thymoquinone-rich fraction greatly improves plasma antioxidant capacity
and expression of antioxidant genes in hypercholesterolemic rats. Free Radic. Biol. Med. 2010, 48, 664–672. [CrossRef]
21. Xu, J.; Zhu, L.; Liu, H.; Li, M.; Liu, Y.; Yang, F.; Pei, Z. Thymoquinone reduces cardiac damage caused by hypercholesterolemia in
apolipoprotein E-deficient mice. Lipids Health Dis. 2018, 17, 173. [CrossRef] [PubMed]
22. Yetkin, N.A.; Büyükoğlan, H.; Sönmez, M.F.; Tutar, N.; Gülmez, I.; Yilmaz, I. The protective effects of thymoquinone on lung
damage caused by cigarette smoke. Biotech. Histochem. 2020, 95, 268–275. [CrossRef] [PubMed]
23. Nagi, M.N.; Almakki, H.A.; Sayed-Ahmed, M.M.; Al-Bekairi, A.M. Thymoquinone supplementation reverses acetaminophen-
induced oxidative stress, nitric oxide production and energy decline in mice liver. Food Chem. Toxicol. 2010, 48, 2361–2365.
[CrossRef] [PubMed]
24. Ince, S.; Kucukkurt, I.; Demirel, H.H.; Turkmen, R.; Sever, E. Thymoquinone attenuates cypermethrin induced oxidative stress in
Swiss albino mice. Pestic. Biochem. Physiol. 2012, 104, 229–235. [CrossRef]
25. Basarslan, F.; Yilmaz, N.; Ates, S.; Ozgur, T.; Tutanc, M.; Motor, V.K.; Arica, V.; Yilmaz, C.; Inci, M.; Buyukbas, S. Protective effects
of thymoquinone on vancomycin-induced nephrotoxicity in rats. Hum. Exp. Toxicol. 2012, 31, 726–733. [CrossRef] [PubMed]
26. Lei, X.; Liu, M.; Yang, Z.; Ji, M.; Guo, X.; Dong, W. Thymoquinone prevents and ameliorates dextran sulfate sodium-induced
colitis in mice. Dig. Dis. Sci. 2012, 57, 2296–2303. [CrossRef]
27. Butt, A.S.; Nisar, N.; Mughal, T.A.; Ghani, N.; Altaf, I. Anti-oxidative and anti-proliferative activities of extracted phytochemical
compound thymoquinone. J. Pak. Med. Assoc. 2019, 69, 1479–1485. [CrossRef]
28. Hossen, M.J.; Yang, W.S.; Kim, D.; Aravinthan, A.; Kim, J.H.; Cho, J.Y. Thymoquinone: An IRAK1 inhibitor with in vivo and
in vitro anti-inflammatory activities. Sci. Rep. 2017, 7, 42995. [CrossRef]
29. Guo, Y.R.; Cao, Q.D.; Hong, Z.S.; Tan, Y.Y.; Chen, S.D.; Jin, H.J.; Tan, K.; Wang, D.Y.; Yan, Y. The origin, transmission and clinical
therapies on coronavirus disease 2019 (COVID-19) outbreak—An update on the status. Mil. Med. Res. 2020, 7, 11. [CrossRef]
30. 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 2020, 367, 1444–1448. [CrossRef]
31. Loganathan, S.K.; Schleicher, K.; Malik, A.; Quevedo, R.; Langille, E.; Teng, K.; Oh, R.H.; Rathod, B.; Tsai, R.; Samavarchi-Tehrani,
P.; et al. Rare driver mutations in head and neck squamous cell carcinomas converge on NOTCH signaling. Science 2020, 367,
1264–1269. [CrossRef] [PubMed]
32. Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the
epithelial cells of oral mucosa. Int. J. Oral. Sci. 2020, 12, 8. [CrossRef] [PubMed]
33. Perlot, T.; Penninger, J.M. ACE2—From the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect. 2013, 15,
866–873. [CrossRef] [PubMed]
34. Katopodis, P.; Anikin, V.; Randeva, H.S.; Spandidos, D.A.; Chatha, K.; Kyrou, I.; Karteris, E. Pan-cancer analysis of transmembrane
protease serine 2 and cathepsin L that mediate cellular SARS.CoV.2 infection leading to COVID-19. Int. J. Oncol. 2020, 57, 533–539.
[CrossRef] [PubMed]
35. Jean, S.S.; Lee, P.I.; Hsueh, P.R. Treatment options for COVID-19: The reality and challenges. J. Microbiol. Immunol. Infect. 2020, 53,
436–443. [CrossRef]
36. Xu, X.; Ong, Y.K.; Wang, D.Y. Role of adjunctive treatment strategies in COVID-19 and a review of international and national
clinical guidelines. Mil. Med. Res. 2020, 7, 22. [CrossRef]
37. Isik, A.F.; Kati, I.; Bayram, I.; Ozbek, H. A new agent for treatment of acute respiratory distress syndrome: Thymoquinone.
An experimental study in a rat model. Eur. J. Cardio Thorac. Surg. 2005, 28, 301–305. [CrossRef]
38. Ekström, M.; Dahlander, B. Palliativ farmakologisk behandling vid svår covid-19. Lakartidningen 2020, 117, covidwho-68125.
39. Tittarelli, R.; Pellegrini, M.; Scarpellini, M.G.; Marinelli, E.; Bruti, V.; Di Luca, N.M.; Busardò, F.P.; Zaami, S. Hepatotoxicity of
paracetamol and related fatalities. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 95–101.
40. Bjarnason, I. Gastrointestinal safety of NSAIDs and over-the-counter analgesics. Int. J. Clin. Pract. 2013, 67, 37–42. [CrossRef]
41. Hellms, S.; Gueler, F.; Gutberlet, M.; Schebb, N.H.; Rund, K.; Kielstein, J.T.; VoChieu, V.D.; Rauhut, S.; Greite, R.; Martirosian,
P.; et al. Single-dose diclofenac in healthy volunteers can cause decrease in renal perfusion measured by functional magnetic
resonance imaging. J. Pharm. Pharmacol. 2019, 71, 1262–1270. [CrossRef] [PubMed]
42. Aslan, M.; Kırımlıoğlu, E.; Afşar, E.; Çeker, T.; Yılmaz, Ç. Increased PUFA levels in kidney epithelial cells in the course of
diclofenac toxicity. Toxicol. Vitr. 2020, 66, 104836. [CrossRef] [PubMed]
43. Aycan, İ.Ö.; Elpek, Ö.; Akkaya, B.; Kıraç, E.; Tuzcu, H.; Kaya, S.; Coşkunfırat, N.; Aslan, M. Diclofenac induced gastrointestinal
and renal toxicity is alleviated by thymoquinone treatment. Food Chem. Toxicol. 2018, 118, 795–804. [CrossRef] [PubMed]
44. Ahmad, S.S.; Najmi, A.K.; Kaundal, M.; Akhtar, M. Gastroprotective Effect of Thymoquinone on Water Immersion Restraint
Stress Induced Ulceration in Rats. Drug Res. (Stuttgart) 2017, 67, 366–372. [CrossRef] [PubMed]
45. Magdy, M.A.; Hanan, E.A.; Nabila, E.M. Thymoquinone: Novel gastroprotective mechanisms. Eur. J. Pharmacol. 2012, 697,
126–131. [CrossRef] [PubMed]
Cells 2021, 10, 302 20 of 29

46. Sagit, M.; Korkmaz, F.; Gürgen, S.G.; Kaya, M.; Akcadag, A.; Ozcan, I. The protective role of thymoquinone in the prevention of
gentamicin ototoxicity. Am. J. Otolaryngol. Head Neck Med. Surg. 2014, 35, 603–609. [CrossRef]
47. Galaly, S.R.; Ahmed, O.M.; Mahmoud, A.M. Thymoquinone and curcumin prevent gentamicin-induced liver injury by attenuating
oxidative stress, inflammation and apoptosis. J. Physiol. Pharmacol. 2014, 65, 823–832.
48. Xiao, J.; Ke, Z.P.; Shi, Y.; Zeng, Q.; Cao, Z. The cardioprotective effect of thymoquinone on ischemia-reperfusion injury in isolated
rat heart via regulation of apoptosis and autophagy. J. Cell. Biochem. 2018, 119, 7212–7217. [CrossRef]
49. Kampan, N.C.; Xiang, S.D.; McNally, O.M.; Stephens, A.N.; Quinn, M.A.; Plebanski, M. Immunotherapeutic Interleukin-6 or
Interleukin-6 Receptor Blockade in Cancer: Challenges and Opportunities. Curr. Med. Chem. 2017, 25, 4785–4806. [CrossRef]
50. Luo, P.; Liu, Y.; Qiu, L.; Liu, X.; Liu, D.; Li, J. Tocilizumab treatment in COVID-19: A single center experience. J. Med. Virol. 2020,
92, 814–818. [CrossRef]
51. Cobourne-Duval, M.K.; Taka, E.; Mendonca, P.; Soliman, K.F.A. Thymoquinone increases the expression of neuroprotective
proteins while decreasing the expression of pro-inflammatory cytokines and the gene expression NFκB pathway signaling targets
in LPS/IFNγ -activated BV-2 microglia cells. J. Neuroimmunol. 2018, 320, 87–97. [CrossRef] [PubMed]
52. Bruce-Hickman, D.; Sajeed, S.M.; Pang, Y.H.; Seow, C.S.; Chen, W.; Gulati Kansal, M. Bowel ulceration following tocilizumab
administration in a COVID-19 patient. BMJ Open Gastroenterol. 2020, 7, e000484. [CrossRef] [PubMed]
53. Singh, A.; Ahmad, I.; Akhter, S.; Jain, G.K.; Iqbal, Z.; Talegaonkar, S.; Ahmad, F.J. Nanocarrier based formulation of Thymoquinone
improves oral delivery: Stability assessment, in vitro and in vivo studies. Colloids Surf. B Biointerfaces 2013, 102, 822–832.
[CrossRef] [PubMed]
54. Uz, U.; Chen, B.; Palmer, J.N.; Cingi, C.; Unlu, H.; Cohen, N. A Effects of thymoquinone and montelukast on sinonasal ciliary
beat frequency. Am. J. Rhinol. Allergy 2014, 28, 122–125. [CrossRef] [PubMed]
55. Schneider-Stock, R.; Fakhoury, I.H.; Zaki, A.M.; El-Baba, C.O.; Gali-Muhtasib, H.U. Thymoquinone: Fifty years of success in the
battle against cancer models. Drug Discov. Today 2014, 19, 18–30. [CrossRef]
56. Sun, S.Y.; Hail, N.; Lotan, R. Apoptosis as a novel target for cancer chemoprevention. J. Natl. Cancer Inst. 2004, 96,
662–672. [CrossRef]
57. Levine, A.J. P53, the Cellular Gatekeeper for Growth and Division. Cell 1997, 88, 323–331. [CrossRef]
58. Cantley, L.C.; Neel, B.G. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phospho-
inositide 3-kinase/AKT pathway. Proc. Natl. Acad. Sci. USA 1999, 96, 4240–4245. [CrossRef]
59. Reed, J.C. Mechanisms of apoptosis avoidance in cancer. Curr. Opin. Oncol. 1999, 11, 68–75. [CrossRef]
60. Olsson, M.; Zhivotovsky, B. Caspases and cancer. Cell Death Differ. 2011, 18, 1441–1449. [CrossRef]
61. Dastjerdi, M.N.; Mehdiabady, E.M.; Iranpour, F.G.; Bahramian, H. Effect of thymoquinone on P53 gene expression and conse-
quence apoptosis in breast cancer cell line. Int. J. Prev. Med. 2016, 7, 66. [CrossRef] [PubMed]
62. Woo, C.C.; Hsu, A.; Kumar, A.P.; Sethi, G.; Tan, K.H.B. Thymoquinone Inhibits Tumor Growth and Induces Apoptosis in a Breast
Cancer Xenograft Mouse Model: The Role of p38 MAPK and ROS. PLoS ONE 2013, 8, e75356. [CrossRef] [PubMed]
63. Woo, C.C.; Loo, S.Y.; Gee, V.; Yap, C.W.; Sethi, G.; Kumar, A.P.; Tan, K.H.B. Anticancer activity of thymoquinone in breast cancer
cells: Possible involvement of PPAR-γ pathway. Biochem. Pharmacol. 2011, 82, 464–475. [CrossRef] [PubMed]
64. Dehghani, H.; Hashemi, M.; Entezari, M.; Mohsenifar, A. The comparison of anticancer activity of thymoquinone and nanothy-
moquinone on human breast adenocarcinoma. Iran. J. Pharm Res. 2015, 14, 539–546.
65. Motaghed, M.; Al-Hassan, F.M.; Hamid, S.S. Cellular responses with thymoquinone treatment in human breast cancer cell line
MCF-7. Pharmacogn. Res. 2013, 5, 200–206.
66. Linjawi, S.A.A.; Khalil, W.K.B.; Hassanane, M.M.; Ahmed, E.S. Evaluation of the protective effect of Nigella sativa extract and
its primary active component thymoquinone against DMBA-induced breast cancer in female rats. Arch. Med. Sci. 2015, 11,
220–229. [CrossRef]
67. Chae, I.G.; Song, N.Y.; Kim, D.H.; Lee, M.Y.; Park, J.M.; Chun, K.S. Thymoquinone induces apoptosis of human renal carcinoma
Caki-1 cells by inhibiting JAK2/STAT3 through pro-oxidant effect. Food Chem. Toxicol. 2020, 139, 111253. [CrossRef]
68. Diab-Assaf, M.; Semaan, J.; El-Sabban, M.; Al Jaouni, S.K.; Azar, R.; Kamal, M.A.; Harakeh, S. Inhibition of Proliferation and
Induction of Apoptosis by Thymoquinone via Modulation of TGF Family, p53, p21 and Bcl-2α in Leukemic Cells. Anticancer Agents
Med. Chem. 2018, 18, 210–215. [CrossRef]
69. Subburayan, K.; Thayyullathil, F.; Pallichankandy, S.; Rahman, A.; Galadari, S. Par-4-dependent p53 up-regulation plays a critical
role in thymoquinone-induced cellular senescence in human malignant glioma cells. Cancer Lett. 2018, 426, 80–97. [CrossRef]
70. Paramasivam, A.; Raghunandhakumar, S.; Priyadharsini, J.V.; Jayaraman, G. In vitro anti-neuroblastoma activity of thymoquinone
against neuro-2a cells via cell-cycle arrest. Asian Pac. J. Cancer Prev. 2016, 16, 8313–8319. [CrossRef]
71. Park, J.E.; Kim, D.H.; Ha, E.; Choi, S.M.; Choi, J.S.; Chun, K.S.; Joo, S.H. Thymoquinone induces apoptosis of human epidermoid
carcinoma A431 cells through ROS-mediated suppression of STAT3. Chem. Biol. Interact. 2019, 312, 108799. [CrossRef] [PubMed]
72. Ichwan, S.J.A.; Al-Ani, I.M.; Bilal, H.G.; Suriyah, W.H.; Taher, M.; Ikeda, M.A. Apoptotic activities of thymoquinone, an active
ingredient of black seed (Nigella sativa), in cervical cancer cell lines. Chin. J. Physiol. 2014, 57, 249–255. [CrossRef] [PubMed]
73. Samarghandian, S.; Azimi-Nezhad, M.; Farkhondeh, T. Thymoquinone-induced antitumor and apoptosis in human lung
adenocarcinoma cells. J. Cell. Physiol. 2019, 234, 10421–10431. [CrossRef] [PubMed]
74. Peng, L.; Liu, A.; Shen, Y.; Xu, H.Z.; Yang, S.Z.; Ying, X.Z.; Liao, W.; Liu, H.X.; Lin, Z.Q.; Chen, Q.Y.; et al. Antitumor and
anti-angiogenesis effects of thymoquinone on osteosarcoma through the NF-κB pathway. Oncol. Rep. 2013, 29, 571–578. [PubMed]
Cells 2021, 10, 302 21 of 29

75. Connelly, L.; Barham, W.; Onishko, H.M.; Sherrill, T.; Chodosh, L.A.; Blackwell, T.S.; Yull, F.E. Inhibition of NF-kappa B activity in
mammary epithelium increases tumor latency and decreases tumor burden. Oncogene 2011, 30, 1402–1412. [CrossRef] [PubMed]
76. Ulasli, S.S.; Celik, S.; Gunay, E.; Ozdemir, M.; Hazman, O.; Ozyurek, A.; Koyuncu, T.; Unlu, M. Anticancer effects of thymoquinone,
caffeic acid phenethyl ester and resveratrol on A549 non-small cell lung cancer cells exposed to benzo(a)pyrene. Asian Pac. J.
Cancer Prev. 2013, 14, 6159–6164. [CrossRef] [PubMed]
77. Ke, X.; Zhao, Y.; Lu, X.; Wang, Z.; Liu, Y.; Ren, M.; Lu, G.; Zhang, D.; Sun, Z.; Xu, Z.; et al. TQ inhibits hepatocellular carcinoma
growth in vitro and in vivo via repression of Notch signaling. Oncotarget 2015, 6, 32610–32621. [CrossRef]
78. Relles, D.; Chipitsyna, G.I.; Gong, Q.; Yeo, C.J.; Arafat, H.A. Thymoquinone Promotes Pancreatic Cancer Cell Death and Reduction
of Tumor Size through Combined Inhibition of Histone Deacetylation and Induction of Histone Acetylation. Adv. Prev. Med. 2016,
2016, 1–9. [CrossRef]
79. Paramasivam, A.; Sambantham, S.; Shabnam, J.; Raghunandhakumar, S.; Anandan, B.; Rajiv, R.; Priyadharsini, J.V.; Jayaraman, G.
Anti-cancer effects of thymoquinone in mouse neuroblastoma (Neuro-2a) cells through caspase-3 activation with down-regulation
of XIAP. Toxicol. Lett. 2012, 213, 151–159. [CrossRef]
80. Xu, D.; Ma, Y.; Zhao, B.; Li, S.; Zhang, Y.; Pan, S.; Wu, Y.; Wang, J.; Wang, D.; Pan, H.; et al. Thymoquinone induces G2/M arrest,
inactivates PI3K/Akt and nuclear factor-κB pathways in human cholangiocarcinomas both in vitro and in vivo. Oncol. Rep. 2014,
31, 2063–2070. [CrossRef]
81. Park, E.J.; Chauhan, A.K.; Min, K.J.; Park, D.C.; Kwon, T.K. Thymoquinone induces apoptosis through downregulation of c-FLIP
and Bcl-2 in Renal carcinoma Caki cells. Oncol. Rep. 2016, 36, 2261–2267. [CrossRef] [PubMed]
82. Taha, M.M.E.; Sheikh, B.Y.; Salim, L.Z.A.; Mohan, S.; Khan, A.; Kamalidehghan, B.; Ahmadipour, F.; Abdelwahab, S.I. Thymo-
quinone induces apoptosis and increase ROS in ovarian cancer cell line. Cell. Mol. Biol. 2016, 62, 97–101. [PubMed]
83. Salim, L.Z.A.; Mohan, S.; Othman, R.; Abdelwahab, S.I.; Kamalidehghan, B.; Sheikh, B.Y.; Ibrahim, M.Y. Thymoquinone
induces mitochondria-mediated apoptosis in acute lymphoblastic leukaemia in vitro. Molecules 2013, 18, 11219–11240.
[CrossRef] [PubMed]
84. Zhang, M.; Du, H.; Huang, Z.; Zhang, P.; Yue, Y.; Wang, W.; Liu, W.; Zeng, J.; Ma, J.; Chen, G.; et al. Thymoquinone induces
apoptosis in bladder cancer cell via endoplasmic reticulum stress-dependent mitochondrial pathway. Chem. Biol. Interact. 2018,
292, 65–75. [CrossRef]
85. Elkhoely, A.; Hafez, H.F.; Ashmawy, A.M.; Badary, O.; Abdelaziz, A.; Mostafa, A.; Shouman, S.A. Chemopreventive and
therapeutic potentials of thymoquinone in HepG2 cells: Mechanistic perspectives. J. Nat. Med. 2015, 69, 313–323. [PubMed]
86. Das, S.; Dey, K.K.; Dey, G.; Pal, I.; Majumder, A.; MaitiChoudhury, S.; Kundu, S.C.; Mandal, M. Antineoplastic and Apoptotic
Potential of Traditional Medicines Thymoquinone and Diosgenin in Squamous Cell Carcinoma. PLoS ONE 2012, 7, e46641.
[CrossRef] [PubMed]
87. Dera, A.; Rajagopalan, P. Thymoquinone attenuates phosphorylation of AKT to inhibit kidney cancer cell proliferation.
J. Food Biochem. 2019, 43, e12793. [CrossRef] [PubMed]
88. Hussain, A.R.; Ahmed, M.; Ahmed, S.; Manogaran, P.; Platanias, L.C.; Alvi, S.N.; Al-Kuraya, K.S.; Uddin, S. Thymoquinone
suppresses growth and induces apoptosis via generation of reactive oxygen species in primary effusion lymphoma. Free Radic.
Biol. Med. 2011, 50, 978–987. [CrossRef] [PubMed]
89. Dergarabetian, E.M.; Ghattass, K.I.; El-Sitt, S.B.; Al Mismar, R.M.; El-Baba, C.O.; Itani, W.S.; Melhem, N.M.; El-Hajj, H.A.;
Bazarbachi, A.A.H.; Schneider-Stock, R.; et al. Thymoquinone induces apoptosis in malignant T-cells via generation of ROS.
Front. Biosci. Elit. 2013, 5, 706–719. [CrossRef]
90. Krylova, N.G.; Drobysh, M.S.; Semenkova, G.N.; Kulahava, T.A.; Pinchuk, S.V.; Shadyro, O.I. Cytotoxic and antiproliferative
effects of thymoquinone on rat C6 glioma cells depend on oxidative stress. Mol. Cell. Biochem. 2019, 462, 195–206. [CrossRef]
91. Liou, Y.F.; Chen, P.N.; Chu, S.C.; Kao, S.H.; Chang, Y.Z.; Hsieh, Y.S.; Chang, H.R. Thymoquinone suppresses the proliferation of
renal cell carcinoma cells via reactive oxygen species-induced apoptosis and reduces cell stemness. Environ. Toxicol. 2019, 34,
1208–1220. [CrossRef] [PubMed]
92. Kundu, J.; Choi, B.Y.; Jeong, C.H.; Kundu, J.K.; Chun, K.S. Thymoquinone induces apoptosis in human colon cancer HCT116 cells
through inactivation of STAT3 by blocking JAK2- and Src-mediated phosphorylation of EGF receptor tyrosine kinase. Oncol. Rep.
2014, 32, 821–828. [CrossRef] [PubMed]
93. Ashour, A.E.; Ahmed, A.F.; Kumar, A.; Zoheir, K.M.A.; Aboul-Soud, M.A.; Ahmad, S.F.; Attia, S.M.; Abd-Allah, A.R.A.;
Cheryan, V.T.; Rishi, A.K. Thymoquinone inhibits growth of human medulloblastoma cells by inducing oxidative stress and
caspase-dependent apoptosis while suppressing NF-κB signaling and IL-8 expression. Mol. Cell. Biochem. 2016, 416, 141–155.
[CrossRef] [PubMed]
94. Koka, P.S.; Mondal, D.; Schultz, M.; Abdel-Mageed, A.B.; Agrawal, K.C. Studies on molecular mechanisms of growth inhibitory
effects of thymoquinone against prostate cancer cells: Role of reactive oxygen species. Exp. Biol. Med. 2010, 235, 751–760.
[CrossRef] [PubMed]
95. Bashir, A.O.; El-Mesery, M.E.; Anwer, R.; Eissa, L.A. Thymoquinone potentiates miR-16 and miR-375 expressions in hepatocellular
carcinoma. Life Sci. 2020, 254, 117794. [CrossRef]
96. Helmy, S.A.; El-Mesery, M.; El-Karef, A.; Eissa, L.A.; El Gayar, A.M. Thymoquinone upregulates TRAIL/TRAILR2 expression
and attenuates hepatocellular carcinoma in vivo model. Life Sci. 2019, 233, 116673. [CrossRef]
Cells 2021, 10, 302 22 of 29

97. Ashour, A.E.; Abd-Allah, A.R.; Korashy, H.M.; Attia, S.M.; Alzahrani, A.Z.; Saquib, Q.; Bakheet, S.A.; Abdel-Hamied, H.E.;
Jamal, S.; Rishi, A.K. Thymoquinone suppression of the human hepatocellular carcinoma cell growth involves inhibition of IL-8
expression, elevated levels of TRAIL receptors, oxidative stress and apoptosis. Mol. Cell. Biochem. 2014, 389, 85–98. [CrossRef]
98. Acharya, B.R.; Chatterjee, A.; Ganguli, A.; Bhattacharya, S.; Chakrabarti, G. Thymoquinone inhibits microtubule polymerization
by tubulin binding and causes mitotic arrest following apoptosis in A549 cells. Biochimie 2014, 97, 78–91. [CrossRef]
99. Rajput, S.; Kumar, B.N.P.; Dey, K.K.; Pal, I.; Parekh, A.; Mandal, M. Molecular targeting of Akt by thymoquinone promotes G1
arrest through translation inhibition of cyclin D1 and induces apoptosis in breast cancer cells. Life Sci. 2013, 93, 783–790. [CrossRef]
100. Li, F.; Rajendran, P.; Sethi, G. Thymoquinone inhibits proliferation, induces apoptosis and chemosensitizes human multiple
myeloma cells through suppression of signal transducer and activator of transcription 3 activation pathway. Br. J. Pharmacol.
2010, 161, 541–554. [CrossRef]
101. Badr, G.; Mohany, M.; Abu-Tarboush, F. Thymoquinone decreases F-actin polymerization and the proliferation of human
multiple myeloma cells by suppressing STAT3 phosphorylation and Bcl2/Bcl-XL expression. Lipids Health Dis. 2011, 10,
236. [CrossRef] [PubMed]
102. Attoub, S.; Sperandio, O.; Raza, H.; Arafat, K.; Al-Salam, S.; Al Sultan, M.A.; Al Safi, M.; Takahashi, T.; Adem, A. Thymoquinone
as an anticancer agent: Evidence from inhibition of cancer cells viability and invasion in vitro and tumor growth in vivo. Fundam.
Clin. Pharmacol. 2013, 27, 557–569. [CrossRef] [PubMed]
103. Ashley, R.E.; Osheroff, N. Natural products as topoisomerase II poisons: Effects of thymoquinone on DNA cleavage mediated by
human topoisomerase IIα. Chem. Res. Toxicol. 2014, 27, 787–793. [CrossRef] [PubMed]
104. Hatiboglu, M.A.; Kocyigit, A.; Guler, E.M.; Akdur, K.; Nalli, A.; Karatas, E.; Tuzgen, S. Thymoquinone Induces Apoptosis in
B16-F10 Melanoma Cell Through Inhibition of p-STAT3 and Inhibits Tumor Growth in a Murine Intracerebral Melanoma Model.
World Neurosurg. 2018, 114, e182–e190. [CrossRef] [PubMed]
105. El-Baba, C.; Mahadevan, V.; Fahlbusch, F.B.; Suma Mohan, S.; Rau, T.T.; Gali-Muhtasib, H.; Schneider-Stock, R. Thymoquinone-
induced conformational changes of PAK1 interrupt prosurvival MEK-ERK signaling in colorectal cancer. Mol. Cancer 2014, 13,
201. [CrossRef] [PubMed]
106. Rashid, M.; Sanjarin, F.; Sabouni, F. Thymoquinone Effects on Cell Viability, Apoptosis and VEGF-A Gene Expression Level in
AGS(CRL-1739) Cell Line. Anticancer Agents Med. Chem. 2019, 19, 820–826. [CrossRef]
107. Ünal, T.D.; Hamurcu, Z.; Delibaşı, N.; Çınar, V.; Güler, A.; Gökçe, S.; Nurdinov, N.; Ozpolat, B. Thymoquinone Inhibits
Proliferation and Migration of MDA-MB-231 Triple Negative Breast Cancer Cells by Suppressing Autophagy and Beclin-1 and
LC3. Anticancer Agents Med. Chem. 2020, 21, 355–364. [CrossRef]
108. Costa, J.G.; Keser, V.; Jackson, C.; Saraiva, N.; Guerreiro Almeida, N.; Camões, S.P.; Manguinhas, R.; Castro, M.; Miranda, J.P.;
Fernandes, A.S.; et al. A multiple endpoint approach reveals potential in vitro anticancer properties of thymoquinone in human
renal carcinoma cells. Food Chem. Toxicol. 2020, 136, 111076. [CrossRef]
109. Kabil, N.; Bayraktar, R.; Kahraman, N.; Mokhlis, H.A.; Calin, G.A.; Lopez-Berestein, G.; Ozpolat, B. Thymoquinone inhibits
cell proliferation, migration, and invasion by regulating the elongation factor 2 kinase (eEF-2K) signaling axis in triple-negative
breast cancer. Breast Cancer Res. Treat. 2018, 171, 593–605. Available online: http://www.ncbi.nlm.nih.gov/pubmed/29971628
(accessed on 11 August 2020). [CrossRef]
110. Kou, B.; Kou, Q.; Ma, B.; Zhang, J.; Sun, B.; Yang, Y.; Li, J.; Zhou, J.; Liu, W. Thymoquinone inhibits metastatic phenotype and
epithelial-mesenchymal transition in renal cell carcinoma by regulating the LKB1/AMPK signaling pathway. Oncol. Rep. 2018, 40,
1443–1450. [CrossRef]
111. Feng, L.M.; Wang, X.F.; Huang, Q.X. Thymoquinone induces cytotoxicity and reprogramming of EMT in gastric cancer cells by
targeting PI3K/Akt/mTOR pathway. J. Biosci. 2017, 42, 547–554. [CrossRef] [PubMed]
112. Kou, B.; Liu, W.; Zhao, W.; Duan, P.; Yang, Y.; Yi, Q.; Guo, F.; Li, J.; Zhou, J.; Kou, Q. Thymoquinone inhibits epithelial-
mesenchymal transition in prostate cancer cells by negatively regulating the TGF-β/Smad2/3 signaling pathway. Oncol. Rep.
2017, 38, 3592–3598. [CrossRef] [PubMed]
113. Li, J.; Asaduzzaman Khan, M.; Wei, C.; Cheng, J.; Chen, H.; Yang, L.; Ijaz, I.; Fu, J. Thymoquinone inhibits the migration and
invasive characteristics of cervical cancer cells siha and caski in vitro by targeting epithelial to mesenchymal transition associated
transcription factors twist1 and zeb1. Molecules 2017, 22, 2105. [CrossRef] [PubMed]
114. Ng, W.K.; Yazan, L.S.; Ismail, M. Thymoquinone from Nigella sativa was more potent than cisplatin in eliminating of SiHa cells
via apoptosis with down-regulation of Bcl-2 protein. Toxicol. Vitr. 2011, 25, 1392–1398. [CrossRef] [PubMed]
115. Iskender, B.; Izgi, K.; Hizar, E.; Jauch, J.; Arslanhan, A.; Yuksek, E.H.; Canatan, H. Inhibition of epithelial-mesenchymal transition
in bladder cancer cells via modulation of mTOR signalling. Tumor Biol. 2016, 37, 8281–8291. [CrossRef] [PubMed]
116. Khan, M.A.; Tania, M.; Wei, C.; Mei, Z.; Fu, S.; Cheng, J.; Xu, J.; Fu, J. Thymoquinone inhibits cancer metastasis by downregulating
TWIST1 expression to reduce epithelial to mesenchymal transition. Oncotarget 2015, 6, 19580–19591. [CrossRef] [PubMed]
117. Zhang, Y.; Fan, Y.; Huang, S.; Wang, G.; Han, R.; Lei, F.; Luo, A.; Jing, X.; Zhao, L.; Gu, S.; et al. Thymoquinone inhibits
the metastasis of renal cell cancer cells by inducing autophagy via AMPK/mTOR signaling pathway. Cancer Sci. 2018, 109,
3865–3873. [CrossRef]
118. Hussain, A.R.; Uddin, S.; Ahmed, M.; Al-Dayel, F.; Bavi, P.P.; Al-Kuraya, K.S. Phosphorylated IκBα Predicts Poor Prognosis
in Activated B-Cell Lymphoma and Its Inhibition with Thymoquinone Induces Apoptosis via ROS Release. PLoS ONE 2013, 8,
e60540. [CrossRef] [PubMed]
Cells 2021, 10, 302 23 of 29

119. Arumugam, P.; Subramanian, R.; Priyadharsini, J.V.; Gopalswamy, J. Thymoquinone inhibits the migration of mouse neuroblas-
toma (Neuro-2a) cells by down-regulating MMP-2 and MMP-9. Chin. J. Nat. Med. 2016, 14, 904–912. [CrossRef]
120. Liou, Y.F.; Hsieh, Y.S.; Hung, T.W.; Chen, P.N.; Chang, Y.Z.; Kao, S.H.; Lin, S.W.; Chang, H.R. Thymoquinone inhibits metastasis
of renal cell carcinoma cell 786-o-si3 associating with downregulation of MMP-2 and u-pa and suppression of PI3K/src signaling.
Int. J. Med. Sci. 2019, 16, 686–695. [CrossRef]
121. Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Hsieh, D.J.Y.; Lin, Y.M.; Chen, L.M.; Kuo, W.W.; Huang, C.Y. Thymoquinone
induces caspase-independent, autophagic cell death in cpt-11-resistant LoVo colon cancer via mitochondrial dysfunction and
activation of JNK and p38. J. Agric. Food Chem. 2015, 63, 1540–1546. [CrossRef] [PubMed]
122. Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Chen, R.J.; Lin, Y.M.; Viswanadha, V.P.; Kuo, W.W.; Huang, C.Y. Inhibition of
NF-κB and metastasis in irinotecan (CPT-11)-resistant LoVo colon cancer cells by thymoquinone via JNK and p38. Environ. Toxicol.
2017, 32, 669–678. [CrossRef] [PubMed]
123. Ha, J.H.; Jayaraman, M.; Radhakrishnan, R.; Gomathinayagam, R.; Yan, M.; Song, Y.S.; Isidoro, C.; Dhanasekaran, D.N. Differential
effects of thymoquinone on lysophosphatidic acid-induced oncogenic pathways in ovarian cancer cells. J. Tradit. Complement.
Med. 2020, 10, 207–216. [CrossRef] [PubMed]
124. Shanmugam, M.K.; Ahn, K.S.; Hsu, A.; Woo, C.C.; Yuan, Y.; Tan, K.H.B.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Koh,
A.P.F.; et al. Thymoquinone Inhibits Bone Metastasis of Breast Cancer Cells Through Abrogation of the CXCR4 Signaling Axis.
Front. Pharmacol. 2018, 9, 1294. [CrossRef] [PubMed]
125. Badr, G.; Lefevre, E.A.; Mohany, M. Thymoquinone inhibits the CXCL12-induced chemotaxis of multiple myeloma cells and
increases their susceptibility to fas-mediated apoptosis. PLoS ONE 2011, 6, e23741. [CrossRef] [PubMed]
126. Zhang, M.; Du, H.; Wang, L.; Yue, Y.; Zhang, P.; Huang, Z.; Lv, W.; Ma, J.; Shao, Q.; Ma, M.; et al. Thymoquinone suppresses
invasion and metastasis in bladder cancer cells by reversing EMT through the Wnt/β-catenin signaling pathway. Chem. Biol.
Interact. 2020, 320, 109022. [CrossRef] [PubMed]
127. Kolli-Bouhafs, K.; Boukhari, A.; Abusnina, A.; Velot, E.; Gies, J.P.; Lugnier, C.; Rondé, P. Thymoquinone reduces migration and
invasion of human glioblastoma cells associated with FAK, MMP-2 and MMP-9 down-regulation. Investig. New Drugs. 2012, 30,
2121–2131. [CrossRef]
128. Yang, J.; Kuang, X.R.; Lv, P.T.; Yan, X.X. Thymoquinone inhibits proliferation and invasion of human nonsmall-cell lung cancer
cells via ERK pathway. Tumor Biol. 2014, 36, 259–269. [CrossRef]
129. Zhu, W.Q.; Wang, J.; Guo, X.F.; Liu, Z.; Dong, W.G. Thymoquinone inhibits proliferation in gastric cancer via the STAT3 pathway
in vivo and in vitro. World J. Gastroenterol. 2016, 22, 4149–4159. [CrossRef]
130. Hsu, H.H.; Chen, M.C.; Day, C.H.; Lin, Y.M.; Li, S.Y.; Tu, C.C.; Padma, V.V.; Shih, H.N.; Kuo, W.W.; Huang, C.Y. Thymoquinone
suppresses migration of LoVo human colon cancer cells by reducing prostaglandin E2 induced COX-2 activation. World J.
Gastroenterol. 2017, 23, 1171–1179. [CrossRef]
131. Lang, M.; Borgmann, M.; Oberhuber, G.; Evstatiev, R.; Jimenez, K.; Dammann, K.W.; Jambrich, M.; Khare, V.; Campregher, C.;
Ristl, R.; et al. Thymoquinone attenuates tumor growth in ApcMin mice by interference with Wnt-signaling. Mol. Cancer 2013,
12, 41. [CrossRef] [PubMed]
132. Ren, X.; Luo, W. Exploration of pro-apoptotic effect of Thymoquinone on oral squamous cell carcinoma cells through PI3K/Akt
signaling pathway. Cell. Mol. Biol. 2019, 65, 61–64. [CrossRef] [PubMed]
133. Sakalar, C.; Yuruk, M.; Kaya, T.; Aytekin, M.; Kuk, S.; Canatan, H. Pronounced transcriptional regulation of apoptotic and
TNF-NF-kappa-B signaling genes during the course of thymoquinone mediated apoptosis in HeLa cells. Mol. Cell. Biochem. 2013,
383, 243–251. [CrossRef] [PubMed]
134. Ahmad, I.; Muneer, K.M.; Tamimi, I.A.; Chang, M.E.; Ata, M.O.; Yusuf, N. Thymoquinone suppresses metastasis of melanoma
cells by inhibition of NLRP3 inflammasome. Toxicol. Appl. Pharmacol. 2013, 270, 70–76. [CrossRef] [PubMed]
135. Kotowski, U.; Heiduschka, G.; Kadletz, L.; Fahim, T.; Seemann, R.; Schmid, R.; Schneider, S.; Mitterbauer, A.; Thurnher, D. Effect
of thymoquinone on head and neck squamous cell carcinoma cells in vitro: Synergism with radiation. Oncol. Lett. 2017, 14,
1147–1151. [CrossRef]
136. Rajput, S.; Kumar, B.N.P.; Banik, P.; Parida, S.; Mandal, M. Thymoquinone restores radiation-induced TGF-β expression and
abrogates EMT in chemoradiotherapy of breast cancer cells. J. Cell. Physiol. 2015, 230, 620–629. [CrossRef]
137. Velho-Pereira, R.; Kumar, A.; Pandey, B.N.; Jagtap, A.G.; Mishra, K.P. Radiosensitization in human breast carcinoma cells by
thymoquinone: Role of cell cycle and apoptosis. Cell. Biol. Int. 2011, 35, 1025–1029. [CrossRef]
138. Farooqui, Z.; Shahid, F.; Khan, A.A.; Khan, F. Oral administration of Nigella sativa oil and thymoquinone attenuates long term
cisplatin treatment induced toxicity and oxidative damage in rat kidney. Biomed. Pharmacother. 2017, 96, 912–923. [CrossRef]
139. Al-Malki, A.L.; Sayed, A.A.R. Thymoquinone attenuates cisplatin-induced hepatotoxicity via nuclear factor kappa-β. BMC
Complement. Altern. Med. 2014, 14, 282. [CrossRef]
140. Liu, X.; Dong, J.; Cai, W.; Pan, Y.; Li, R.; Li, B. The effect of thymoquinone on apoptosis of SK-OV-3 ovarian cancer cell by
regulation of Bcl-2 and Bax. Int. J. Gynecol. Cancer 2017, 27, 1596–1601. [CrossRef]
141. Wilson, A.J.; Saskowski, J.; Barham, W.; Yull, F.; Khabele, D. Thymoquinone enhances cisplatin-response through direct tumor
effects in a syngeneic mouse model of ovarian cancer. J. Ovarian Res. 2015, 8, 46. [CrossRef] [PubMed]
142. Albassam, A.A.; Ahad, A.; Alsultan, A.; Al-Jenoobi, F.I. Inhibition of cytochrome P450 enzymes by thymoquinone in human liver
microsomes. Saud. Pharm J. 2018, 26, 673–677. [CrossRef] [PubMed]
Cells 2021, 10, 302 24 of 29

143. Bashmail, H.A.; Alamoudi, A.A.; Noorwali, A.; Hegazy, G.A.; Ajabnoor, G.M.; Al-Abd, A.M. Thymoquinone enhances paclitaxel
anti-breast cancer activity via inhibiting tumor-associated stem cells despite apparent mathematical antagonism. Molecules 2020,
25, 426. [CrossRef] [PubMed]
144. Singh, S.K.; Apata, T.; Gordetsky, J.B.; Singh, R. Docetaxel combined with thymoquinone induces apoptosis in prostate cancer
cells via inhibition of the PI3K/AKT signaling pathway. Cancers 2019, 11, 1390. [CrossRef]
145. Dirican, A.; Atmaca, H.; Bozkurt, E.; Erten, C.; Karaca, B.; Uslu, R. Novel combination of docetaxel and thymoquinone induces
synergistic cytotoxicity and apoptosis in DU-145 human prostate cancer cells by modulating PI3K–AKT pathway. Clin. Transl.
Oncol. 2014, 17, 145–151. [CrossRef]
146. Arafa, E.S.A.; Zhu, Q.; Shah, Z.I.; Wani, G.; Barakat, B.M.; Racoma, I.; El-Mahdy, M.A.; Wani, A.A. Thymoquinone up-regulates
PTEN expression and induces apoptosis in doxorubicin-resistant human breast cancer cells. Mutat. Res. Fundam. Mol. Mech.
Mutagen. 2011, 706, 28–35. [CrossRef]
147. Effenberger-Neidnicht, K.; Schobert, R. Combinatorial effects of thymoquinone on the anti-cancer activity of doxorubicin. Cancer
Chemother. Pharmacol. 2011, 67, 867–874. [CrossRef]
148. Fatfat, M.; Fakhoury, I.; Habli, Z.; Mismar, R.; Gali-Muhtasib, H. Thymoquinone enhances the anticancer activity of doxorubicin
against adult T-cell leukemia in vitro and in vivo through ROS-dependent mechanisms. Life Sci. 2019, 232, 116628. [CrossRef]
149. Brown, R.K.; Wilson, G.; Tucci, M.; Benghuzzi, H. The effects of thymoquinone and doxorubicin on leukemia and cardiomyocyte
cell lines. Biomed. Sci. Instrum. 2014, 50, 391–396.
150. Soltani, A.; Pourgheysari, B.; Shirzad, H.; Sourani, Z. Antiproliferative and Apoptosis-Inducing Activities of Thymoquinone in
Lymphoblastic Leukemia Cell Line. Indian J. Hematol. Blood Transfus. 2017, 33, 516–524. [CrossRef]
151. Ganji-Harsini, S.; Khazaei, M.; Rashidi, Z.; Ghanbari, A. Thymoquinone could increase the efficacy of tamoxifen induced apoptosis
in human breast cancer cells: An in vitro study. Cell J. 2016, 18, 245–254. [PubMed]
152. Rajput, S.; Kumar, B.N.P.; Sarkar, S.; Das, S.; Azab, B.; Santhekadur, P.K.; Das, S.K.; Emdad, L.; Sarkar, D.; Fisher, P.B.; et al.
Targeted Apoptotic Effects of Thymoquinone and Tamoxifen on XIAP Mediated Akt Regulation in Breast Cancer. PLoS ONE
2013, 8, e61342. [CrossRef]
153. Suddek, G.M. Protective role of thymoquinone against liver damage induced by tamoxifen in female rats. Can. J. Physiol.
Pharmacol. 2014, 92, 640–644. [CrossRef] [PubMed]
154. Ndreshkjana, B.; Çapci, A.; Klein, V.; Chanvorachote, P.; Muenzner, J.K.; Huebner, K.; Steinmann, S.; Erlenbach-Wuensch, K.;
Geppert, C.I.; Agaimy, A.; et al. Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal
cancer cells. Cell Death Dis. 2019, 10, 379. [CrossRef] [PubMed]
155. Kensara, O.A.; El-Shemi, A.G.; Mohamed, A.M.; Refaat, B.; Idris, S.; Ahmad, J. Thymoquinone subdues tumor growth and
potentiates the chemopreventive effect of 5-fluorouracil on the early stages of colorectal carcinogenesis in rats. Drug Des. Dev. Ther.
2016, 10, 2239–2253.
156. Lei, X.; Lv, X.; Liu, M.; Yang, Z.; Ji, M.; Guo, X.; Dong, W. Thymoquinone inhibits growth and augments 5-fluorouracil-induced
apoptosis in gastric cancer cells both in vitro and in vivo. Biochem. Biophys. Res. Commun. 2012, 417, 864–868. [CrossRef]
157. Abuzinadah, M.F.; Ahmad, A. Pharmacological studies on the efficacy of a thymoquinone-containing novel polyherbal formula-
tion against cisplatin-induced hepatorenal toxicity in rats. J. Food Biochem. 2020, 44, e13131. [CrossRef] [PubMed]
158. Hu, X.; Ma, J.; Vikash, V.; Li, J.; Wu, D.; Liu, Y.; Zhang, J.; Dong, W. Thymoquinone Augments Cisplatin-Induced Apoptosis on
Esophageal Carcinoma Through Mitigating the Activation of JAK2/STAT3 Pathway. Dig. Dis. Sci. 2018, 63, 126–134. [CrossRef]
159. Ma, J.; Hu, X.; Li, J.; Wu, D.; Lan, Q.; Wang, Q.; Tian, S.; Dong, W. Enhancing conventional chemotherapy drug cisplatin-induced
anti-tumor effects on human gastric cancer cells both in vitro and in vivo by Thymoquinone targeting PTEN gene. Oncotarget
2017, 8, 85926–85939. [CrossRef] [PubMed]
160. Alaufi, O.M.; Noorwali, A.; Zahran, F.; Al-Abd, A.M.; Al-Attas, S. Cytotoxicity of thymoquinone alone or in combination with
cisplatin (CDDP) against oral squamous cell carcinoma in vitro. Sci. Rep. 2017, 7, 13131. [CrossRef]
161. Kadil, Y.; Mouhcine, M.; Filali, H. In Silico Investigation of the SARS CoV2 Protease with Thymoquinone Major Constituent of
Nigella Sativa. Curr. Drug. Discov. Technol. 2020, 17. [CrossRef] [PubMed]
162. Ibrahim, I.M.; Abdelmalek, D.H.; Elfiky, A.A. GRP78: A cell’s response to stress. Life Sci. 2019, 226, 156–163. [CrossRef] [PubMed]
163. Ibrahim, I.M.; Abdelmalek, D.H.; Elshahat, M.E.; Elfiky, A.A. COVID-19 spike-host cell receptor GRP78 binding site prediction.
J. Infect. 2020, 80, 554–562. [CrossRef] [PubMed]
164. Aguiar, J.; Tremblay, B.J.-M.; Mansfield, M.; Woody, O.; Lobb, B.; Banerjee, A.; Chandiramohan, A.; Tiessen, N.; Dvorkin-Gheva,
A.; Revill, S.; et al. Gene expression and in situ protein profiling of candidate SARS-CoV-2 receptors in human airway epithelial
cells and lung tissue. Eur. Respir. J. 2020, 56, 2001123. [CrossRef] [PubMed]
165. Köseler, A.; Sabirli, R.; Gören, T.; Türkçüer, I.; Kurt, Ö. Endoplasmic reticulum stress markers in SARS-COV-2 infection and
pneumonia: Case-control study. In Vivo (Brooklyn) 2020, 34, 1645–1650.
166. Xie, J.; Tao, Z.-H.; Zhao, J.; Li, T.; Wu, Z.-H.; Zhang, J.-F.; Zhang, J.; Hu, X.-C. Glucose regulated protein 78 (GRP78) inhibits
apoptosis and attentinutes chemosensitivity of gemcitabine in breast cancer cell via AKT/mitochondrial apoptotic pathway.
Biochem. Biophys. Res. Commun. 2016, 474, 612–619. [CrossRef]
167. Kuroda, K.; Horiguchi, A.; Asano, T.; Ito, K.; Asakuma, J.; Sato, A.; Yoshii, H.; Hayakawa, M.; Sumitomo, M.; Asano, T.
Glucose-regulated protein 78 positivity as a predictor of poor survival in patients with renal cell carcinoma. Urol. Int. 2011, 87,
450–456. [CrossRef]
Cells 2021, 10, 302 25 of 29

168. Teng, Y.; Ai, Z.; Wang, Y.; Wang, J.; Luo, L. Proteomic identification of PKM2 and HSPA5 as potential biomarkers for predicting
high-risk endometrial carcinoma. J. Obstet. Gynaecol. Res. 2013, 39, 317–325. [CrossRef]
169. Zhang, J.; Jiang, Y.; Jia, Z.; Li, Q.; Gong, W.; Wang, L.; Wei, D.; Yao, J.; Fang, S.; Xie, K. Association of elevated GRP78 expression
with increased lymph node metastasis and poor prognosis in patients with gastric cancer. Clin. Exp. Metastasis 2006, 23,
401–410. [CrossRef]
170. Xing, X.; Lai, M.; Wang, Y.; Xu, E.; Huang, Q. Overexpression of glucose-regulated protein 78 in colon cancer. Clin. Chim Acta
2006, 364, 308–315. [CrossRef]
171. Wu, C.T.; Wang, W.C.; Chen, M.F.; Su, H.Y.; Chen, W.Y.; Wu, C.H.; Chang, Y.J.; Liu, H.H. Glucose-regulated protein 78 mediates
hormone-independent prostate cancer progression and metastasis through maspin and COX-2 expression. Tumor Biol. 2014, 35,
195–204. [CrossRef] [PubMed]
172. Bouhlel, A.; Mosbah, B.; Abdallah, H.; Ribault, C.; Viel, R.; Mannaï, S.; Corlu, A.; Abdennebi, B. Thymoquinone prevents
endoplasmic reticulum stress and mitochondria-induced apoptosis in a rat model of partial hepatic warm ischemia reperfusion.
Biomed. Pharmacother. 2017, 94, 964–973. [CrossRef] [PubMed]
173. Elfiky, A.A. Natural products may interfere with SARS-CoV-2 attachment to the host cell. J. Biomol. Struct. Dyn. 2020,
1–16. [CrossRef]
174. O’Connell, M.A.; Hayes, J.D. The Keap1/Nrf2 pathway in health and disease: From the bench to the clinic. Biochem. Soc. Trans.
2015, 43, 687–689. [CrossRef]
175. Su, C.; Zhang, P.; Song, X.; Shi, Q.; Fu, J.; Xia, X.; Bai, H.; Hu, L.; Xu, D.; Song, E.; et al. Tetrachlorobenzoquinone activates NRF2
signaling by keap1 cross-linking and ubiquitin translocation but not keap1-cullin3 complex dissociation. Chem. Res. Toxicol. 2015,
28, 765–774. [CrossRef]
176. Lewis, K.N.; Mele, J.; Hayes, J.D.; Buffenstein, R. Nrf2, a guardian of healthspan and gatekeeper of species longevity. Integr. Comp.
Biol. 2010, 50, 829–843. [CrossRef]
177. Niture, S.K.; Kaspar, J.W.; Shen, J.; Jaiswal, A.K. Nrf2 signaling and cell survival. Toxicol. Appl. Pharmacol. 2010, 244,
37–42. [CrossRef]
178. Das, B.N.; Kim, Y.W.; Keum, Y.S. Mechanisms of Nrf2/keap1-dependent phase II cytoprotective and detoxifying gene expression
and potential cellular targets of chemopreventive isothiocyanates. Oxid. Med. Cell. Longev. 2013, 2013, 839409. [CrossRef]
179. Prawan, A.; Kundu, J.K.; Surh, Y.J. Molecular basis of heme oxygenase-1 induction: Implications for chemoprevention and
chemoprotection. Antioxid. Redox Signal. 2005, 7, 1688–1703. [CrossRef]
180. Denicola, G.M.; Karreth, F.A.; Humpton, T.J.; Gopinathan, A.; Wei, C.; Frese, K.; Mangal, D.; Yu, K.H.; Yeo, C.J.; Calhoun, E.S.; et al.
Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 2011, 475, 106–110. [CrossRef]
181. Satoh, H.; Moriguchi, T.; Takai, J.; Ebina, M.; Yamamoto, M. Nrf2 prevents initiation but accelerates progression through the kras
signaling pathway during lung carcinogenesis. Cancer Res. 2013, 73, 4158–4168. [CrossRef] [PubMed]
182. Tao, S.; Rojo de la Vega, M.; Chapman, E.; Ooi, A.; Zhang, D.D. The effects of NRF2 modulation on the initiation and progression
of chemically and genetically induced lung cancer. Mol. Carcinog. 2018, 57, 182–192. [CrossRef] [PubMed]
183. Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43.
[CrossRef] [PubMed]
184. Kobayashi, E.H.; Suzuki, T.; Funayama, R.; Nagashima, T.; Hayashi, M.; Sekine, H.; Tanaka, N.; Moriguchi, T.; Motohashi, H.;
Nakayama, K.; et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription.
Nat. Commun. 2016, 7, 11624. [CrossRef] [PubMed]
185. Singh, N.; Saha, L.; Kumari, P.; Singh, J.; Bhatia, A.; Banerjee, D.; Chakrabarti, A. Effect of dimethyl fumarate on neuroinflammation
and apoptosis in pentylenetetrazol kindling model in rats. Brain Res. Bull. 2019, 144, 233–245. [CrossRef]
186. Battino, M.; Giampieri, F.; Pistollato, F.; Sureda, A.; de Oliveira, M.R.; Pittalà, V.; Fallarino, F.; Nabavi, S.F.; Atanasov, A.G.; Nabavi,
S.M. Nrf2 as regulator of innate immunity: A molecular Swiss army knife! Biotechnol. Adv. 2018, 36, 358–370. [CrossRef] [PubMed]
187. Chhunchha, B.; Kubo, E.; Singh, D.P. Sulforaphane-Induced Klf9/Prdx6 Axis Acts as a Molecular Switch to Control Redox
Signaling and Determines Fate of Cells. Cells 2019, 8, 1159. [CrossRef]
188. Zucker, S.N.; Fink, E.E.; Bagati, A.; Mannava, S.; Bianchi-Smiraglia, A.; Bogner, P.N.; Wawrzyniak, J.A.; Foley, C.; Leonova, K.I.;
Grimm, M.J.; et al. Nrf2 amplifies oxidative stress via induction of Klf9. Mol. Cell. 2014, 53, 916–928. [CrossRef]
189. Lee, C. Therapeutic modulation of virus-induced oxidative stress via the Nrf2-dependent antioxidative pathway. Oxid. Med. Cell.
Longev. 2018, 2018, 6208067. [CrossRef]
190. Lomeli, N.; Bota, D.A.; Davies, K.J.A. Diminished stress resistance and defective adaptive homeostasis in age-related diseases.
Clin. Sci. 2017, 131, 2573–2599. [CrossRef]
191. Zhao, S.; Ghosh, A.; Lo, C.S.; Chenier, I.; Scholey, J.W.; Filep, J.G.; Ingelfinger, J.R.; Zhang, S.L.; Chan, J.S.D. Nrf2 deficiency
upregulates intrarenal angiotensin-converting enzyme-2 and angiotensin 1-7 receptor expression and attenuates hypertension
and nephropathy in diabetic mice. Endocrinology 2018, 159, 836–852. [CrossRef] [PubMed]
192. McCord, J.M.; Hybertson, B.M.; Cota-Gomez, A.; Geraci, K.P.; Gao, B. Nrf2 activator pb125® as a potential therapeutic agent
against covid-19. Antioxidants 2020, 9, 518. [CrossRef] [PubMed]
193. Hassan, S.M.; Jawad, M.J.; Ahjel, S.W.; Singh, R.B.; Singh, J.; Awad, S.M.; Hadi, N.R. The Nrf2 Activator (DMF) and Covid-19: Is
there a Possible Role? Med. Arch. (Sarajevo) 2020, 74, 134–138. [CrossRef] [PubMed]
Cells 2021, 10, 302 26 of 29

194. Ramos-Gomez, M.; Kwak, M.K.; Dolan, P.M.; Itoh, K.; Yamamoto, M.; Talalay, P.; Kensler, T.W. Sensitivity to carcinogenesis is
increased and chemoprotective efficacy of enzyme inducers is lost in nrf2 transcription factor-deficient mice. Proc. Natl. Acad. Sci.
USA 2001, 98, 3410–3415. [CrossRef] [PubMed]
195. Bauer, A.K.; Cho, H.Y.; Miller-DeGraff, L.; Walker, C.; Helms, K.; Fostel, J.; Yamamoto, M.; Kleeberger, S.R. Targeted deletion of
Nrf2 reduces urethane-induced lung tumor development in mice. PLoS ONE 2011, 6, e26590. [CrossRef]
196. Long, M.; Tao, S.; De La Vega, M.R.; Jiang, T.; Wen, Q.; Park, S.L.; Zhang, D.D.; Wondrak, G.T. Nrf2-dependent suppression of
azoxymethane/dextran sulfate sodium-induced colon carcinogenesis by the cinnamon-derived dietary factor cinnamaldehyde.
Cancer Prev. Res. 2015, 8, 444–454. [CrossRef]
197. Shen, T.; Jiang, T.; Long, M.; Chen, J.; Ren, D.M.; Wong, P.K.; Chapman, E.; Zhou, B.; Zhang, D.D. A Curcumin Derivative That
Inhibits Vinyl Carbamate-Induced Lung Carcinogenesis via Activation of the Nrf2 Protective Response. Antioxid. Redox Signal.
2015, 23, 651–664. [CrossRef]
198. Sekhar, K.R.; Freeman, M.L. Nrf2 promotes survival following exposure to ionizing radiation. Free Radic. Biol. Med. 2015, 88,
268–274. [CrossRef]
199. Tao, S.; Justiniano, R.; Zhang, D.D.; Wondrak, G.T. The Nrf2-inducers tanshinone I and dihydrotanshinone protect human skin
cells and reconstructed human skin against solar simulated UV. Redox Biol. 2013, 1, 532–541. [CrossRef]
200. Tao, S.; Park, S.L.; De La Vega, M.R.; Zhang, D.D.; Wondrak, G.T. Systemic administration of the apocarotenoid bixin protects
skin against solar UV-induced damage through activation of NRF2. Free Radic. Biol. Med. 2015, 89, 690–700. [CrossRef]
201. Knatko, E.V.; Ibbotson, S.H.; Zhang, Y.; Higgins, M.; Fahey, J.W.; Talalay, P.; Dawe, R.S.; Ferguson, J.; Huang, J.T.J.; Clarke, R.; et al.
Nrf2 activation protects against solar-simulated ultraviolet radiation in mice and humans. Cancer Prev. Res. 2015, 8, 475–486.
[CrossRef] [PubMed]
202. Wang, H.; Liu, X.; Long, M.; Huang, Y.; Zhang, L.; Zhang, R.; Zheng, Y.; Liao, X.; Wang, Y.; Liao, Q.; et al. NRF2 activation by
antioxidant antidiabetic agents accelerates tumor metastasis. Sci. Transl. Med. 2016, 8, 334ra51. [CrossRef] [PubMed]
203. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [PubMed]
204. Kundu, J.; Kim, D.H.; Kundu, J.K.; Chun, K.S. Thymoquinone induces heme oxygenase-1 expression in HaCaT cells via Nrf2/ARE
activation: Akt and AMPKα as upstream targets. Food Chem. Toxicol. 2014, 65, 18–26. [CrossRef] [PubMed]
205. Velagapudi, R.; Kumar, A.; Bhatia, H.S.; El-Bakoush, A.; Lepiarz, I.; Fiebich, B.L.; Olajide, O.A. Inhibition of neuroinflammation
by thymoquinone requires activation of Nrf2/ARE signalling. Int. Immunopharmacol. 2017, 48, 17–29. [CrossRef]
206. Ye, Q.; Wang, B.; Mao, J. The pathogenesis and treatment of the ‘Cytokine Storm” in COVID-19. J. Infect. 2020, 80,
607–613. [CrossRef]
207. Cheng, J.L.; Huang, C.; Zhang, G.J.; Liu, D.W.; Li, P.; Lu, C.Y.; Li, J. Epidemiological characteristics of novel coronavirus
pneumonia in Henan. Zhonghua Jie He He Hu Xi Za Zhi 2020, 43, 327–331.
208. 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, 552–555. [CrossRef]
209. Liu, H.; Sun, Y.; Zhang, Y.; Yang, G.; Guo, L.; Zhao, Y.; Pei, Z. Role of Thymoquinone in Cardiac Damage Caused by Sepsis from
BALB/c Mice. Inflammation 2019, 42, 516–525. [CrossRef]
210. Pei, Z.; Hu, J.; Bai, Q.; Liu, B.; Cheng, D.; Liu, H.; Na, R.; Yu, Q. Thymoquinone protects against cardiac damage from
doxorubicin-induced heart failure in Sprague-Dawley rats. RSC Adv. 2018, 8, 14633–14639. [CrossRef]
211. Nagi, M.N.; Al-Shabanah, O.A.; Hafez, M.M.; Sayed-Ahmed, M.M. Thymoquinone supplementation attenuates cyclophosphamide-
induced cardiotoxicity in rats. J. Biochem. Mol. Toxicol. 2011, 25, 135–142. [CrossRef] [PubMed]
212. Nemmar, A.; Al-Salam, S.; Zia, S.; Marzouqi, F.; Al-Dhaheri, A.; Subramaniyan, D.; Dhanasekaran, S.; Yasin, J.; Ali, B.H.; Kazzam,
E.E. Contrasting actions of diesel exhaust particles on the pulmonary and cardiovascular systems and the effects of thymoquinone.
Br. J. Pharmacol. 2011, 164, 1871–1882. [CrossRef] [PubMed]
213. Çolak, M.; Kalemci, S.; Alpaydın, A.Ö.; Karaçam, V.; Meteoğlu, I.; Yılmaz, O.; Itil, B.O. Efficacy of thymoquinone in the treatment
of experimental lipopolysaccharide-induced acute lung injury. Kardiochir. Torakochir. Pol. 2020, 17, 65–69. [CrossRef] [PubMed]
214. Pourgholamhossein, F.; Sharififar, F.; Rasooli, R.; Pourgholi, L.; Nakhaeipour, F.; Samareh-Fekri, H.; Iranpour, M.; Mandegary, A.
Thymoquinone effectively alleviates lung fibrosis induced by paraquat herbicide through down-regulation of pro-fibrotic genes
and inhibition of oxidative stress. Environ. Toxicol. Pharmacol. 2016, 45, 340–345. [CrossRef]
215. Keyhanmanesh, R.; Pejman, L.; Omrani, H.; Mirzamohammadi, Z.; Shahbazfar, A.A. The effect of single dose of thymoquinone,
the main constituents of Nigella sativa, in guinea pig model of asthma. BioImpacts 2014, 4, 75–81.
216. El-Khouly, D.; El-Bakly, W.M.; Awad, A.S.; El-Mesallamy, H.O.; El-Demerdash, E. Thymoquinone blocks lung injury and
fibrosis by attenuating bleomycin-induced oxidative stress and activation of nuclear factor Kappa-B in rats. Toxicology 2012, 302,
106–113. [CrossRef]
217. Zhu, N.; Zhao, X.; Xiang, Y.; Ye, S.; Huang, J.; Hu, W.; Lv, L.; Zeng, C. Thymoquinone attenuates monocrotaline-induced
pulmonary artery hypertension via inhibiting pulmonary arterial remodeling in rats. Int. J. Cardiol. 2016, 221, 587–596. [CrossRef]
218. Kanter, M. Thymoquinone attenuates lung injury induced by chronic toluene exposure in rats. Toxicol. Ind. Health 2011, 27,
387–395. [CrossRef]
219. Suddek, G.M.; Ashry, N.A.; Gameil, N.M. Thymoquinone attenuates cyclophosphamide-induced pulmonary injury in rats.
Inflammopharmacology 2013, 21, 427–435.
220. Suddek, G.M. Thymoquinone-induced relaxation of isolated rat pulmonary artery. J. Ethnopharmacol. 2010, 127, 210–214. [CrossRef]
Cells 2021, 10, 302 27 of 29

221. Ammar, E.S.M.; Gameil, N.M.; Shawky, N.M.; Nader, M.A. Comparative evaluation of anti-inflammatory properties of thymo-
quinone and curcumin using an asthmatic murine model. Int. Immunopharmacol. 2011, 11, 2232–2236. [CrossRef] [PubMed]
222. Su, X.; Ren, Y.; Yu, N.; Kong, L.; Kang, J. Thymoquinone inhibits inflammation, neoangiogenesis and vascular remodeling in
asthma mice. Int. Immunopharmacol. 2016, 38, 70–80. [CrossRef] [PubMed]
223. Kalemci, S.; Micili, S.C.; Acar, T.; Senol, T.; Dirican, N.; Omeroglu, G.; Bagriyanik, A.; Kamaci, G.; Yilmaz, O. Effectiveness of
thymoquinone in the treatment of experimental asthma. Clin. Ter. 2013, 164. [CrossRef]
224. Oguz, S.; Kanter, M.; Erboga, M.; Erenoglu, C. Protective effects of thymoquinone against cholestatic oxidative stress and hepatic
damage after biliary obstruction in rats. J. Mol. Histol. 2012, 43, 151–159. [CrossRef]
225. Raghunandhakumar, S.; Paramasivam, A.; Senthilraja, S.; Naveenkumar, C.; Asokkumar, S.; Binuclara, J.; Jagan, S.; Anan-
dakumar, P.; Devaki, T. Thymoquinone inhibits cell proliferation through regulation of G1/S phase cell cycle transition in
N-nitrosodiethylamine-induced experimental rat hepatocellular carcinoma. Toxicol. Lett. 2013, 223, 60–72. [CrossRef]
226. Awad, A.S.; Kamel, R.; Sherief, M.A.E. Effect of thymoquinone on hepatorenal dysfunction and alteration of CYP3A1 and
spermidine/spermine N-1-acetyl-transferase gene expression induced by renal ischaemia-reperfusion in rats. J. Pharm. Pharmacol.
2011, 63, 1037–1042. [CrossRef]
227. Bai, T.; Lian, L.H.; Wu, Y.L.; Wan, Y.; Nan, J.X. Thymoquinone attenuates liver fibrosis via PI3K and TLR4 signaling pathways in
activated hepatic stellate cells. Int. Immunopharmacol. 2013, 15, 275–281. [CrossRef]
228. Zafeer, M.F.; Waseem, M.; Chaudhary, S.; Parvez, S. Cadmium-induced hepatotoxicity and its abrogation by thymoquinone. J.
Biochem. Mol. Toxicol. 2012, 26, 199–205. [CrossRef]
229. Alenzi, F.Q.; El-Sayed El-Bolkiny, Y.; Salem, M.L. Protective effects of Nigella sativa oil and thymoquinone against toxicity
induced by the anticancer drug cyclophosphamide. Br. J. Biomed. Sci. 2010, 67, 20–28. [CrossRef]
230. Jaswal, A.; Sinha, N.; Bhadauria, M.; Shrivastava, S.; Shukla, S. Therapeutic potential of thymoquinone against anti-tuberculosis
drugs induced liver damage. Environ. Toxicol. Pharmacol. 2013, 36, 779–786. [CrossRef]
231. Nili-Ahmadabadi, A.; Tavakoli, F.; Hasanzadeh, G.R.; Rahimi, H.R.; Sabzevari, O. Protective effect of pretreatment with
thymoquinone against Aflatoxin B1 induced liver toxicity in mice. DARU. J. Pharm. Sci. 2011, 19, 282–287.
232. Bai, T.; Yang, Y.; Wu, Y.L.; Jiang, S.; Lee, J.J.; Lian, L.H.; Nan, J.X. Thymoquinone alleviates thioacetamide-induced hepatic
fibrosis and inflammation by activating LKB1-AMPK signaling pathway in mice. Int. Immunopharmacol. 2014, 19, 351–357.
[CrossRef] [PubMed]
233. Elsherbiny, N.M.; El-Sherbiny, M. Thymoquinone attenuates Doxorubicin-induced nephrotoxicity in rats: Role of Nrf2 and NOX4.
Chem. Biol. Interact. 2014, 223, 102–108. [CrossRef] [PubMed]
234. Evirgen, O.; Gökçe, A.; Ozturk, O.H.; Nacar, E.; Onlen, Y.; Ozer, B.; Motor, V.K. Effect of Thymoquinone on Oxidative Stress in
Escherichia coli-Induced Pyelonephritis in Rats. Curr. Ther. Res. Clin. Exp. 2011, 72, 204–215. [CrossRef] [PubMed]
235. Shao, Y.Y.; Li, B.; Huang, Y.M.; Luo, Q.; Xie, Y.M.; Chen, Y.H. Thymoquinone attenuates brain injury via an antioxidative pathway
in a status epilepticus rat model. Transl. Neurosci. 2017, 8, 9–14. [CrossRef]
236. Fanoudi, S.; Alavi, M.S.; Hosseini, M.; Sadeghnia, H.R. Nigella sativa and thymoquinone attenuate oxidative stress and cognitive
impairment following cerebral hypoperfusion in rats. Metab. Brain Dis. 2019, 34, 1001–1010. [CrossRef]
237. Baghcheghi, Y.; Hosseini, M.; Beheshti, F.; Salmani, H.; Anaeigoudari, A. Thymoquinone reverses learning and memory
impairments and brain tissue oxidative damage in hypothyroid juvenile rats. Arq. Neuropsiquiatr. 2018, 76, 32–40. [CrossRef]
238. Randhawa, M.A.; Alenazy, A.K.; Alrowaili, M.G.; Basha, J. An active principle of Nigella sativa L.; thymoquinone, showing
significant antimicrobial activity against anaerobic bacteria. J. Intercult. Ethnopharmacol. 2017, 6, 97–101. [CrossRef]
239. Hayat, K.; Asim, M.R.; Nawaz, M.; Li, M.; Zhang, L.; Sun, N. Ameliorative effect of thymoquinone on ovalbumin-induced allergic
conjunctivitis in Balb/c mice. Curr. Eye Res. 2011, 36, 591–598. [CrossRef]
240. Yurttaş, V.; Şereflican, M.; Erkoçoǧlu, M.; Dagli, M.; Terzi, E.H.; Flrat, T.; Seyhan, S. Comparison of histopathological effects of
thymoquinone and local nasal corticosteroids in allergic rhinitis in a rabbit model. ORL 2016, 78, 55–60. [CrossRef]
241. McNabb-Baltar, J.; Jin, D.X.; Grover, A.S.; Redd, W.D.; Zhou, J.C.; Hathorn, K.E.; McCarty, T.R.; Bazarbashi, A.N.; Shen, L.; Chan,
W.W. Lipase Elevation in Patients With COVID-19. Am. J. Gastroenterol. 2020, 115, 1286–1288. [CrossRef] [PubMed]
242. Satou, R.; Penrose, H.; Navar, L.G. Inflammation as a Regulator of the Renin-Angiotensin System and Blood Pressure. Curr.
Hypertens Rep. 2018, 20, 100. [CrossRef] [PubMed]
243. Donoghue, M.; Hsieh, F.; Baronas, E.; Godbout, K.; Gosselin, M.; Stagliano, N.; Donovan, M.; Woolf, B.; Robison, K.; Jeyaseelan,
R.; et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ.
Res. 2000, 87, E1–E9. [CrossRef] [PubMed]
244. Nishiga, M.; Wang, D.W.; Han, Y.; Lewis, D.B.; Wu, J.C. COVID-19 and cardiovascular disease: From basic mechanisms to clinical
perspectives. Nat. Rev. Cardiol. 2020, 17, 543–558. [CrossRef]
245. Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-
Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary
of the Discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [CrossRef]
246. Berk, B.C.; Haendeler, J.; Sottile, J. Angiotensin II, atherosclerosis, and aortic aneurysms. J. Clin. Investig. 2000, 105,
1525–1526. [CrossRef]
247. Bruemmer, D.; Law, R.E. Thiazolidinedione regulation of smooth muscle cell proliferation. Am. J. Med. 2003, 115, 87–92. [CrossRef]
Cells 2021, 10, 302 28 of 29

248. Zhao, Q.; Zhang, J.; Wang, H. PGC-1α limits angiotensin II-induced rat vascular smooth muscle cells proliferation via attenuating
NOX1-mediated generation of reactive oxygen species. Biosci. Rep. 2015, 35, e00252. [CrossRef]
249. Wang, J.; Lv, X.W.; Shi, J.P.; Hu, X.S. Mechanisms involved in ceramide-induced cell cycle arrest in human hepatocarcinoma cells.
World J. Gastroenterol. 2007, 13, 1129–1134. [CrossRef]
250. Kim, J.E.; Choi, H.C. Losartan inhibits vascular smooth muscle cell proliferation through activation of AMP-activated protein
kinase. Korean J. Physiol. Pharmacol. 2010, 14, 299–304. [CrossRef]
251. Wang, K.; Gheblawi, M.; Oudit, G.Y. Angiotensin Converting Enzyme 2: A Double-Edged Sword. Circulation 2020, 142, 426–428.
[CrossRef] [PubMed]
252. Penafuerte, C.A.; Gagnon, B.; Sirois, J.; Murphy, J.; Macdonald, N.; Tremblay, M.L. Identification of neutrophil-derived proteases
and angiotensin II as biomarkers of cancer cachexia. Br. J. Cancer 2016, 114, 680–687. [CrossRef] [PubMed]
253. Mahmudpour, M.; Roozbeh, J.; Keshavarz, M.; Farrokhi, S.; Nabipour, I. COVID-19 cytokine storm: The anger of inflammation.
Cytokine 2020, 133, 155151. [CrossRef] [PubMed]
254. Enayatfard, L.; Mohebbati, R.; Niazmand, S.; Hosseini, M.; Shafei, M.N. The standardized extract of Nigella sativa and its major
ingredient, thymoquinone, ameliorates angiotensin II-induced hypertension in rats. J. Basic Clin. Physiol. Pharmacol. 2019, 30,
51–58. [CrossRef]
255. Pei, X.; Li, X.; Chen, H.; Han, Y.; Fan, Y. Thymoquinone Inhibits Angiotensin II-Induced Proliferation and Migration of Vascular
Smooth Muscle Cells Through the AMPK/PPARγ/PGC-1α Pathway. DNA Cell Biol. 2016, 35, 426–433. [CrossRef]
256. Idris-Khodja, N.; Schini-Kerth, V. Thymoquinone improves aging-related endothelial dysfunction in the rat mesenteric artery.
Naunyn Schmiedebergs Arch. Pharmacol. 2012, 385, 749–758. [CrossRef]
257. Hosseinian, S.; Rad, A.K.; Bideskan, A.E.; Soukhtanloo, M.; Sadeghnia, H.; Shafei, M.N.; Motejadded, F.; Mohebbati, R.; Shahraki,
S.; Beheshti, F. Thymoquinone ameliorates renal damage in unilateral ureteral obstruction in rats. Pharmacol. Rep. 2017, 69,
648–657. [CrossRef]
258. Carsana, L.; Sonzogni, A.; Nasr, A.; Rossi, R.S.; Pellegrinelli, A.; Zerbi, P.; Rech, R.; Colombo, R.; Antinori, S.; Corbellino, M.; et al.
Pulmonary post-mortem findings in a series of COVID-19 cases from northern Italy: A two-centre descriptive study. Lancet Infect.
Dis. 2020, 20, 1135–1140. [CrossRef]
259. Wichmann, D.; Sperhake, J.P.; Lütgehetmann, M.; Steurer, S.; Edler, C.; Heinemann, A.; Heinrich, F.; Mushumba, H.; Kniep, I.;
Schröder, A.S.; et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study.
Ann. Intern. Med. 2020, 173, 268–277. [CrossRef]
260. Towhid, S.T.; Schmidt, E.M.; Schmid, E.; Münzer, P.; Qadri, S.M.; Borst, O.; Lang, F. Thymoquinone-induced platelet apoptosis.
J. Cell. Biochem. 2011, 112, 3112–3121. [CrossRef]
261. Thummuri, D.; Jeengar, M.K.; Shrivastava, S.; Nemani, H.; Ramavat, R.N.; Chaudhari, P.; Naidu, V.G.M. Thymoquinone prevents
RANKL-induced osteoclastogenesis activation and osteolysis in an in vivo model of inflammation by suppressing NF-KB and
MAPK Signalling. Pharmacol. Res. 2015, 99, 63–73. [CrossRef] [PubMed]
262. Muralidharan-Chari, V.; Kim, J.; Abuawad, A.; Naeem, M.; Cui, H.; Mousa, S.A. Thymoquinone modulates blood coagulation
in vitro via its effects on inflammatory and coagulation pathways. Int. J. Mol. Sci. 2016, 17, 474. [CrossRef] [PubMed]
263. Panigrahy, D.; Gilligan, M.M.; Huang, S.; Gartung, A.; Cortés-Puch, I.; Sime, P.J.; Phipps, R.P.; Serhan, C.N.; Hammock, B.D.
Inflammation resolution: A dual-pronged approach to averting cytokine storms in COVID-19? Cancer Metastasis. Rev. 2020, 39,
337–340. [CrossRef] [PubMed]
264. Wang, D.; Dubois, R.N. Eicosanoids and cancer. Nat. Rev. Cancer. 2010, 10, 181–193. [CrossRef]
265. Serhan, C.N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014, 510, 92–101. [CrossRef]
266. Shah, A. Novel Coronavirus-Induced NLRP3 Inflammasome Activation: A Potential Drug Target in the Treatment of COVID-19.
Front. Immunol. 2020, 11, 1021. [CrossRef]
267. Umar, S.; Zargan, J.; Umar, K.; Ahmad, S.; Katiyar, C.K.; Khan, H.A. Modulation of the oxidative stress and inflammatory cytokine
response by thymoquinone in the collagen induced arthritis in Wistar rats. Chem. Biol. Interact. 2012, 197, 40–46. [CrossRef]
268. Arjumand, S.; Shahzad, M.; Shabbir, A.; Yousaf, M.Z. Thymoquinone attenuates rheumatoid arthritis by downregulating TLR2,
TLR4, TNF-α IL-1, and NFκB expression levels. Biomed. Pharmacother. 2019, 111, 958–963. [CrossRef]
269. Liu, Y.; Liao, R.; Qiang, Z.; Zhang, C. Pro-inflammatory cytokine-driven PI3K/Akt/Sp1 signalling and H2S production facilitates
the pathogenesis of severe acute pancreatitis. Biosci. Rep. 2017, 37, BSR20160483. [CrossRef]
270. Hirota, S.A.; Ng, J.; Lueng, A.; Khajah, M.; Parhar, K.; Li, Y.; Lam, V.; Potentier, M.S.; Ng, K.; Bawa, M.; et al. NLRP3 inflammasome
plays a key role in the regulation of intestinal homeostasis. Inflamm. Bowel Dis. 2011, 17, 1359–1372. [CrossRef]
271. Zaki, M.H.; Vogel, P.; Body-Malapel, M.; Lamkanfi, M.; Kanneganti, T.-D. IL-18 Production Downstream of the Nlrp3 Inflamma-
some Confers Protection against Colorectal Tumor Formation. J. Immunol. 2010, 185, 4912–4920. [CrossRef] [PubMed]
272. Chow, M.T.; Sceneay, J.; Paget, C.; Wong, C.S.F.; Duret, H.; Tschopp, J.; Möller, A.; Smyth, M.J. NLRP3 suppresses NK cell-mediated
responses to carcinogen-induced tumors and metastases. Cancer Res. 2012, 72, 5721–5732. [CrossRef] [PubMed]
273. Elinav, E.; Strowig, T.; Kau, A.L.; Henao-Mejia, J.; Thaiss, C.A.; Booth, C.J.; Peaper, D.R.; Bertin, J.; Eisenbarth, S.C.; Gordon, J.I.;
et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011, 145, 745–757. [CrossRef] [PubMed]
274. Tu, S.; Bhagat, G.; Cui, G.; Takaishi, S.; Kurt-Jones, E.A.; Rickman, B.; Betz, K.S.; Penz-Oesterreicher, M.; Bjorkdahl, O.; Fox, J.G.;
et al. Overexpression of Interleukin-1β Induces Gastric Inflammation and Cancer and Mobilizes Myeloid-Derived Suppressor
Cells in Mice. Cancer Cell. 2008, 14, 408–419. [CrossRef] [PubMed]
Cells 2021, 10, 302 29 of 29

275. Bunt, S.K.; Yang, L.; Sinha, P.; Clements, V.K.; Leips, J.; Ostrand-Rosenberg, S. Reduced inflammation in the tumor microen-
vironment delays the accumulation of myeloid-derived suppressor cells and limits tumor progression. Cancer Res. 2007, 67,
10019–10026. [CrossRef]
276. Yang, Y.; Shen, C.; Li, J.; Yuan, J.; Yang, M.; Wang, F.; Li, G.; Li, Y.; Xing, L.; Peng, L.; et al. Exuberant elevation of IP-10, MCP-3
and IL-1ra during SARS-CoV-2 infection is associated with disease severity and fatal outcome. medRxiv 2020. [CrossRef]
277. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al.
Activation of the NLRP3 inflammasome in dendritic cells induces IL-1B-dependent adaptive immunity against tumors. Nat. Med.
2009, 15, 1170–1178. [CrossRef]
278. Houghton, P.J.; Zarka, R.; De Las Heras, B.; Hoult, J.R.S. Fixed oil of Nigella sativa and derived thymoquinone inhibit eicosanoid
generation in leukocytes and membrane lipid peroxidation. Planta Med. 1995, 61, 33–36. [CrossRef]
279. Liu, X.; Chen, Z. The pathophysiological role of mitochondrial oxidative stress in lung diseases. J. Transl. Med. 2017,
15, 207. [CrossRef]
280. Nayak, B.S.; Pinto, S. Protein thiols and thiobarbituric acid reactive substance status in colon cancer patients. Scand. J. Gastroenterol.
2007, 42, 848–851. [CrossRef]
281. Gonçalves, T.L.; Erthal, F.; Corte, C.L.D.; Müller, L.G.; Piovezan, C.M.; Nogueira, C.W.; Rocha, J.B.T. Involvement of ox-
idative stress in the pre-malignant and malignant states of cervical cancer in women. Clin. Biochem. 2005, 38, 1071–1075.
[CrossRef] [PubMed]
282. Guo, T.; Fan, Y.; Chen, M.; Wu, X.; Zhang, L.; He, T.; Wang, H.; Wan, J.; Wang, X.; Lu, Z. Cardiovascular Implications of Fatal
Outcomes of Patients with Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 811–818. [CrossRef] [PubMed]
283. Shah, K.S.; Yang, E.H.; Maisel, A.S.; Fonarow, G.C. The Role of Biomarkers in Detection of Cardio-toxicity. Curr. Oncol. Rep. 2017,
19, 42. [CrossRef] [PubMed]
284. Zilinyi, R.; Czompa, A.; Czegledi, A.; Gajtko, A.; Pituk, D.; Lekli, I.; Tosaki, A. The cardioprotective effect of metformin in
doxorubicin-induced cardiotoxicity: The role of autophagy. Molecules 2018, 23, 1184. [CrossRef]
285. Zheng, Y.; Gu, S.; Li, X.; Tan, J.; Liu, S.; Jiang, Y.; Zhang, C.; Gao, L.; Yang, H.T. Berbamine postconditioning protects the heart
from ischemia/reperfusion injury through modulation of autophagy. Cell Death Dis. 2017, 8, e2577. [CrossRef]
286. Su, Y.J.; Qu, Y.; Zhao, F.Y.; Li, H.F.; Mu, D.Z.; Li, X.H. Regulation of autophagy by the nuclear factor κB signaling pathway in the
hippocampus of rats with sepsis. J. Neuroinflammation. 2015, 12, 116. [CrossRef]
287. Singhal, T. A Review of Coronavirus Disease-2019 (COVID-19). Indian J. Pediatr. 2020, 87, 281–286. [CrossRef]
288. Alqahtani, J.S.; Oyelade, T.; Aldhahir, A.M.; Alghamdi, S.M.; Almehmadi, M.; Alqahtani, A.S.; Almehmadi, M.; Alqahtani, A.S.;
Quaderi, S.; Mandal, S.; et al. Prevalence, severity and mortality associated with COPD and smoking in patients with COVID-19:
A rapid systematic review and meta-analysis. PLoS ONE 2020, 15, e0233147. [CrossRef]
289. Leung, J.M.; Yang, C.X.; Tam, A.; Shaipanich, T.; Hackett, T.L.; Singhera, G.K.; Dorscheid, D.R.; Sin, D.D. ACE-2 expression in the
small airway epithelia of smokers and COPD patients: Implications for COVID-19. Eur. Respir. J. 2020, 55, 2000688. [CrossRef]
290. Parohan, M.; Yaghoubi, S.; Seraji, A. Liver injury is associated with severe coronavirus disease 2019 (COVID-19) infection:
A systematic review and meta-analysis of retrospective studies. Hepatol. Res. 2020, 50, 924–935.
291. Subramaniam, S.R.; Cader, R.A.; Mohd, R.; Yen, K.W.; Ghafor, H.A. Low-dose cyclophosphamide-induced acute hepatotoxicity.
Am. J. Case Rep. 2013, 14, 345–349. [PubMed]
292. Ming, Z.; Yongqiang, Z.; Zijin, Z.; Yan, X.; Di, C.; Xiaoxin, T. Severe and prolonged cyclophosphamide-induced hepatotoxicity in
a breast cancer patient carrying a CYP2B67 variant. Pharmacogenomics 2019, 20, 1119–1124. [CrossRef] [PubMed]
293. Elefsiniotis, I.S.; Pantazis, K.D.; Ilias, A.; Pallis, L.; Mariolis, A.; Glynou, I.; Kada, H.; Moulakakis, A. 586 Tamoxifen (TMX)-
induced hepatotoxicity in breast cancer (BC) patients with pre-existing liver steatosis (LS). The role of glucose intolerance. J.
Hepatol. 2004, 40, 171–172. [CrossRef]
294. De la Vega, M.C.L.; Zapater, P.; Such, J.; Sola-Vera, J.; Payá, A.; Horga, J.F.; Pérez-Mateo, M. Hepatitis tóxica asociada al uso de
tamoxifeno. Presentación de un caso y revisión bibliográfica. Gastroenterol. Hepatol. 2002, 25, 247–250. [CrossRef]
295. Gabarre, P.; Dumas, G.; Dupont, T.; Darmon, M.; Azoulay, E.; Zafrani, L. Acute kidney injury in critically ill patients with
COVID-19. Intensive Care Med. 2020, 46, 1339–1348. [CrossRef]
296. Li, A.; Zhang, W.; Zhang, L.; Liu, Y.; Li, K.; Du, G.; Qin, X. Elucidating the time-dependent changes in the urinary metabolome
under doxorubicin-induced nephrotoxicity. Toxicol. Lett. 2020, 319, 204–212. [CrossRef]
297. Lu, Y.; Li, X.; Geng, D.; Mei, N.; Wu, P.Y.; Huang, C.C.; Jia, T.; Zhao, Y.; Wang, D.; Xiao, A.; et al. Cerebral Micro-Structural Changes
in COVID-19 Patients—An MRI-based 3-month Follow-up Study: A brief title: Cerebral Changes in COVID-19. EClin. Med. 2020,
25, 100484.
298. Filatov, A.; Sharma, P.; Hindi, F.; Espinosa, P.S. Neurological Complications of Coronavirus Disease (COVID-19): Encephalopathy.
Cureus 2020, 12, e7352. [CrossRef]
299. Su, S.; Shen, J.; Zhu, L.; Qiu, Y.; He, J.S.; Tan, J.Y.; Iacucci, M.; Ng, S.C.; Ghosh, S.; Mao, R.; et al. Involvement of diges-
tive system in COVID-19: Manifestations, pathology, management and challenges. Therap. Adv. Gastroenterol. 2020, 13,
1756284820934626. [CrossRef]
300. Ozturker, Z.K. Conjunctivitis as sole symptom of COVID-19: A case report and review of literature. Eur. J. Ophthalmol.
2020. [CrossRef]

You might also like