ArticlePDF AvailableLiterature Review

Interleukin-6 Perpetrator of the COVID-19 Cytokine Storm

Authors:

Abstract and Figures

COVID-19 has emerged as a global pandemic. It is mainly manifested as pneumonia which may deteriorate into severe respiratory failure. The major hallmark of the disease is the systemic inflammatory immune response characterized by Cytokine Storm (CS). CS is marked by elevated levels of inflammatory cytokines, mainly interleukin-6 (IL-6), IL-8, IL-10, tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). Of these, IL-6 is found to be significantly associated with higher mortality. IL-6 is also a robust marker for predicting disease prognosis and deterioration of clinical profile. In this review, the pivotal role played by IL-6 in the immuno-pathology of COVID-19 has been illustrated. The role of IL-6 as a pleiotropic cytokine executing both pro and anti-inflammatory activities has been reviewed. ADAM 10, a metalloproteinase switches the anti-inflammatory pathway of IL-6 to pro inflammatory hence blocking the action of ADAM 10 could be a new therapeutic strategy to mitigate the proinflammatory action of IL-6. Furthermore, we explore the role of anti-IL6 agents, IL-6 receptor antibodies which were being used for autoimmune diseases but now are being repurposed for the therapy of COVID-19.
Content may be subject to copyright.
REVIEW ARTICLE
Interleukin-6 Perpetrator of the COVID-19 Cytokine Storm
Jyoti Shekhawat
1
Kavya Gauba
1
Shruti Gupta
1
Purvi Purohit
1
Prasenjit Mitra
1
Mahendra Garg
2
Sanjeev Misra
3
Praveen Sharma
1
Mithu Banerjee
1
Received: 19 January 2021 / Accepted: 8 June 2021
ÓAssociation of Clinical Biochemists of India 2021
Abstract COVID-19 has emerged as a global pandemic. It
is mainly manifested as pneumonia which may deteriorate
into severe respiratory failure. The major hallmark of the
disease is the systemic inflammatory immune response
characterized by Cytokine Storm (CS). CS is marked by
elevated levels of inflammatory cytokines, mainly inter-
leukin-6 (IL-6), IL-8, IL-10, tumour necrosis factor-a
(TNF-a) and interferon-c(IFN-c). Of these, IL-6 is found
to be significantly associated with higher mortality. IL-6 is
also a robust marker for predicting disease prognosis and
deterioration of clinical profile. In this review, the pivotal
role played by IL-6 in the immuno-pathology of COVID-
19 has been illustrated. The role of IL-6 as a pleiotropic
cytokine executing both pro and anti-inflammatory activi-
ties has been reviewed. ADAM 10, a metalloproteinase
switches the anti-inflammatory pathway of IL-6 to pro
inflammatory hence blocking the action of ADAM 10
could be a new therapeutic strategy to mitigate the proin-
flammatory action of IL-6. Furthermore, we explore the
role of anti-IL6 agents, IL-6 receptor antibodies which
were being used for autoimmune diseases but now are
being repurposed for the therapy of COVID-19.
Keywords Interleukin-6 SARS-CoV-2 Cytokine storm
ACE2 Receptor Monoclonal antibodies
Introduction
Coronavirus Disease 2019 (COVID-19) was first reported
in Wuhan, China, as a cluster of pneumonia cases with
unknown etiology [1]. Since then, it has emerged as an
outbreak and caused global havoc. On 30th January 2020,
the disease had been declared as a Public Health Emer-
gency of International Concern (PHEIC) by the World
Health Organization (WHO) [2]. The etiological agent of
the disease was soon identified as a member of the coro-
navirus family called Severe Acute Respiratory Syndrome
Coronavirus 2 (SARS-CoV-2). Preceding the COVID-19
pandemic, two other outbreaks were witnessed in the
twenty-first century, namely the Severe Acute Respiratory
Syndrome (SARS) and the Middle East Respiratory Syn-
drome (MERS) caused by different pathogenic members of
the coronavirus family (SARS-CoV and MERS-CoV) [3].
&Mithu Banerjee
mithu.banerjee.3@gmail.com
Jyoti Shekhawat
jyotikanwar1000@gmail.com
Kavya Gauba
kavyagauba17@gmail.com
Shruti Gupta
drshrutiga@gmail.com
Purvi Purohit
dr.purvipurohit@gmail.com
Prasenjit Mitra
prasy4u@gmail.com
Mahendra Garg
mkgargs@gmail.com
Sanjeev Misra
misralko@gmail.com
Praveen Sharma
praveensharma55@gmail.com
1
Department of Biochemistry, All India Institute of Medical
Sciences, Jodhpur 342005, India
2
Department of Endocrinology, All India Institute of Medical
Sciences, Jodhpur 342005, India
3
Department of Surgical Oncology, All India Institute of
Medical Sciences, Jodhpur 342005, India
123
Ind J Clin Biochem
https://doi.org/10.1007/s12291-021-00989-8
The immune pathogenesis, genome sequences and evolu-
tionary phylogenetic relationship of SARS-CoV-2 with
other coronaviruses have been fully elucidated [4]. The
mortality rate of COVID-19 infection is 6.7% which is
comparatively lower than for both SARS (10%) and MERS
(35%) [5]. The disease presents itself similar to common
flu with symptoms including fever, cough, nausea, head-
ache, myalgia, dyspnea, pneumonia etc., some are
asymptomatic while others may experience ARDS (actute
respiratory distress syndrome) [6]. However, individuals
with co-morbidities such as diabetes, hypertension or old
age may experience exacerbated symptoms [7]. Inter-
leukin-6 is a multifactorial cytokine which plays role in
both pro-inflammatory pathways as well as anti-inflam-
matory pathways. In COVID-19 it has been reported to be
elevated in from mild to critically ill patients. It is also
correlated with disease patheogenesis and its progression
[8].
COVID-19 Pathogenesis
The transmission of SARS-CoV-2 is primarily via aerosol
droplets or contact with an infected person. Upon entry of
the viral particle inside the lungs, it attaches itself to cells
of the respiratory tract after which it is endocytosed into the
host pulmonary cell. The crystallised structure of SARS-
CoV-2 shows the presence of surface spike (S) proteins,
similar to those found in SARS-CoV, which binds to the
Angiotensin-converting enzyme-2 (ACE-2) receptor pre-
sent on the alveolar epithelial cells and it is responsible for
conversion of Ang-II (angiotensin-II) to angiotensin 1–7
(Ang 1–7) in the renin angiotensin system [9]. The ACE2
receptor is crucial for the virulence of the virus as cells
lacking them are found to be resistant to infection by
SARS-CoV-2 infection [10]. ACE2 are not only expressed
in both upper and lower respiratory tract but also in cell of
small intestine, pancreas, kidneys, heart, oesophagus,
bladder and brain making them all vulnerable to infection
by SARS-CoV-2 [11]. It is noteworthy that normal ACE2
expression is not very high in upper respiratory tract and is
only transiently enhanced in response to infection with
SARS-CoV-2 for increased transmissibility of the virus
[12]. Also, ACE2 expression in other tissues explains the
development of multi-organ failure that often accompanies
severe COVID-19 infection [13,14]. After viral entry,
spike protein is cleaved by transmembrane serine protease
2 (TMPRSS2) and then released by another protease called
Furin, thus promoting viral entry through endosomal
pathway as depicted in Fig. 1[15]. The spike fusion pep-
tide released by Furin then executes the virus-cell fusion
contributing to viral spread and persistence. The low pH of
the endosomal vesicle favours the delivery of viral genome
into the cytoplasm allowing it to replicate and assemble
into virions which are then released from the cell [16]. The
infected cells are then subjected to apoptosis or necrosis,
triggering an inflammatory response which is characterised
by production of pro-inflammatory cytokines and simulta-
neous recruitment of macrophages and Th cells. Th1
(CD4 ?) cells regulate antigen presentation and Interferon-
cproduction while Th17 cells induce secretion of IL-17,
IL-21 and IL-22. This leads to further recruitment of other
inflammatory cells such as neutrophils and CD8 cells [17].
Spread of infection into circulating immune cells causing
their apoptosis, results in lymphocytopenia. A reduction in
the total number of T cells results in a hyperinflammatory
state in the body due to over secretion of cytokines and
chemokines, termed as CS. CS, if untreated, may aggra-
vated to other life-threatening scenarios such as Acute
Respiratory Distress Syndrome (ARDS), Macrophage
Activation Syndrome (MAS) and Secondary Hemophago-
cytic Lymphohistiocytosis (sHLH).
Cytokine Storm Associated with SARS-CoV-2
Infection
Hyper-cytokinemia or ‘Cytokine storm (CS)’ in COVID-
19 is marked by an aggressive hyperinflammatory immune
response and is the major hallmark of the disease [18]. It is
a conglomeration of inflammatory symptoms induced by
the activation of T-cells, macrophages and subsequent
release of cytokines, which further potentiates recruitment
and activation of other immune cells [19]. It is hypothe-
sised that immune responses associated with MAS/HLH
might be the driving force for COVID-19 related CS (20).
CS is characterised by elevated serum levels of pro-in-
flammatory cytokines and chemokines, namely, IL-1, IL-6,
IL-12, IFN-cand TNF-a[8,20,21]. Elevated levels of
these pro-inflammatory cytokines in severe patients might
be useful as biomarkers of the disease prognosis and can be
used as potential targets for therapy [22]. Several studies
have suggested that CS, directly or indirectly, correlates
with lung injury, ARDS, multi-organ failure, need for
mechanical ventilation and unfavourable disease prognosis
in COVID-19 [22,23].
Along with cytokines, elevated levels of serum crea-
tinine, lactate dehydrogenase, C-reactive protein, procal-
citonin, ferritin, D-dimer and White Blood Cell count are
the lab parameters that portend impending respiratory
failure and need for supplementary oxygen in COVID-19
patients [23].
Of all the cytokines mediating the CS, IL-6 has been
proposed as the most accurate predictor of disease course
as well as mortality in SARS-CoV-2 infected patients.
Moreover, therapeutic implications targeting IL-6 or its
Ind J Clin Biochem
123
signalling have shown success in treatment of COVID-19
patients.
IL-6 in Viral Immunity: A Double-Edged Sword
Interleukin-6 is a pleiotropic cytokine produced in response
to tissue damage by viral infections. The IL-6 signaling
cascade is a highly regulated and dynamic process with
well-defined pro and anti-inflammatory effects mediated by
the Trans and Classical signal transduction respectively
[24]. IL-6 homeostasis governs the outcomes of immuno-
protection versus immunopathology of viral infections.
As a part of pro-inflammatory functions, Th17 activation
and differentiation are dependent on IL-6. Rapid induction
of granzyme B and perforin expression in CD8
?
T cells
which eliminates the viral pathogen is also dependant on
IL-6 trans-signalling along with IL-15 [25]. The activation
of these Cytotoxic T Lymphocytes (CTLs) then stimulates
the release of TNF-aand IFN-cwhich modulates
neutrophil migration [26]. Furthermore, IL-6 serves as an
endogenous pyrogenic cytokine serving as a thermoregu-
lator and amplifying the immune surveillance in viral
infections [27]. Early IL-6 signalling is known to promote
IL-27 dependant maturation of regulatory T cells in lungs
and limit viral immunopathology. Removal of either IL-6
or IL-27 is known to enhance viral infection. During res-
piratory viral infections, IL-6 acts as a driver of virus-
induced immunopathology by inducing the production of
IL-27 by monocytes and macrophages in the respiratory
tract, promoting the local maturation of T cells [28].
Studies on IL-6 deficient mice infected with H1N1 influ-
enza virus have shown that IL-6 is required to heal acute
lung injury caused by influenza A virus [29]. Mice lacking
IL-6 displayed reduced ability to mount an anti-viral
immune response which was further associated with
enhanced proliferation and migration of lung fibroblasts.
These mice exhibited a compromised recruitment of mac-
rophages in the lungs leading to reduced phagocytosis of
viruses in lung macrophages [30].
Fig. 1 SARS-CoV-2 entry into target cell. The virus binds to ACE2
receptor followed by cleavage of spike protein by a serine protease,
TRMPSS2. This cleavage triggers the internalization of the virion
particle through endocytosis. Then another protease called Furin acts
to release the spike fusion peptide facilitating viral entry into host.s
Ind J Clin Biochem
123
On the other hand, as part of anti-inflammatory func-
tions, IL-6 is involved in the progression of viral diseases,
ultimately favouring viral persistence in infected hosts. IL-
6 promotes Th2 response while inhibiting Th1 cell prolif-
eration [31]. It does so by two mechanisms: Firstly, by
stimulating CD4 ?T cells to secrete IL-4 thus directing
response to Th2 and secondly, by suppressing IFN-c
expression by CD4 ?T cells which normally promotes
Th1 polarization and is essential for an effective antiviral
response [32]. IL-6 promotes Th17 cell differentiation
leading to the production of IL-17 which upregulates the
expression of anti-apoptotic molecules enhancing the sur-
vival of the virus in infected cells. IL-17 also blocks the
destruction of target cells by CTLs. Neutralization of IL-17
augments virus clearance and enhances lysis by CTLs
leading to elimination of virus-infected cells [33]. IL-17
also promotes migration of neutrophils to the lungs leading
to tissue damage during inflammation (35). This infiltration
by neutrophils leads to the development of Neutrophil
Extracellular Traps (NETs) in the lungs which contributes
to organ damage and COVID-19 related ARDS [34]. IL-6
produced by TLR-mediated signalling counter-regulates
the CD8 ?T cell responses leading to T-cell dysfunction
in chronic viral infections [35]. It works synergistically
with IL-1band TNFato induce trypsin upregulation in
influenza virus-cytokine-trypsin cycle and this helps virus
to replicate by activating matrix metalloproteinases that
causes degradation of extracellular matrix. This IL-6
mediated trypsin upregulation may account for multi-organ
failure in influenza virus infection [36].
IL-6 binds to its membrane-bound receptor to initiate the
classic-type signalling which adds to anti-inflammatory
processes as illustrated in Fig. 2. Cells lacking expression
of membrane-bound IL-6 receptors are part of trans-sig-
nalling, mediated by soluble type receptor of IL-6 (sIL-6R)
and gp130, responsible for pro-inflammatory functions of
IL-6 [38]. sIL-6R is generated by the action of metallo-
proteinases known as ADAM10 and ADAM17 and alter-
native splicing mechanism [37]. Thus, therapeutic
approaches need to be designed in a way that would
attenuate the pro-inflammatory response without affecting
the classic (anti-inflammatory) signalling process of IL-6
[38,39]. Since ADAM10 plays a key role in switching
from the classical (anti-inflammatory) to trans signalling
pathway (pro-inflammatory), strategies to block its action
holds a lot of promise in preventing the cytokine storm
[37]. A fusion protein Sgp130-Fc has been developed from
the conjugation of sgp130, a natural inhibitor of trans
signalling cascade, and Fc region of IgG human antibody
[40,41]. This molecule specifically blocks trans signalling
without effecting classic signalling pathway [42].
Therefore, the debatable role of IL-6 as an anti- and pro-
inflammatory cytokine requires vigilant evaluation for it to
be considered as an effective therapeutic target in SARS-
CoV-2 infection.
Fig. 2 The classic and the
trans-signalling pathways are
mediated by the membrane-
bound form of IL-6 receptor
(mIL6R) and soluble form of
IL-6 receptor (sIL6-R)
respectively. Monoclonal
antibodies against IL-6 and IL-
6R can inhibit these pathways.
Similarly, Janus Kinase
Inhibitors (JAKinibs) inhibit
phosphorylation of STAT (a
transcription factor), thus
blocking the downstream
signalling of IL-6. ADAM10
plays a key role in switching
from the classical to trans
signalling pathway by
converting sIL-6R to mIL-6R
Ind J Clin Biochem
123
IL-6: The Sidekick to ACE -2 Immunopathology
in SARS-CoV-2 Infection
There is evidence that suggests a strong association
between circulating IL-6 levels and blood pressure. An
increased IL-6 expression directly correlates with hyper-
tension [43]. Therefore, hypertensive patients with
COVID-19 who have elevated levels of IL-6 are highly
susceptible to severe respiratory failure [44]. The Renin-
Angiotenstin Aldosterone System (RAAS) maintains the
blood pressure and electrolyte balance by two major
mechanisms which are in a state of dynamic equilibrium.
These are the Angiotensin II–Angiotensin 1 Receptor axis
and the Angiotensin 1–7-Mas receptor axis as illustrated in
Fig. 3a. The former causes an inflammatory response and
vasoconstriction while the latter suppresses inflammatory
responses and supports vasodilation [45]. Disruption of the
equilibrium between these two axes disturbs the home-
ostasis maintained by RAAS thus regulating systemic
inflammation.
As already discussed, sars cov2 gains entry into the host
lungs via ACE2 receptor binding, thus rendering the
receptor unavailable for maintenance of RAAS. An
imbalance between the two axes of RAAS results in a shift
in equilibrium towards the pro-inflammatory pathway
mediated by Angiotensin 1 receptor axis, as depicted in
Fig. 3b[46]. Many studies have also reported increased
levels of angiotensin II in covid 19 patients when compared
to healthy individuals, and is found to be in linear associ-
ation with viral load and lung injury [47]. Also, angiotensin
Fig. 3 a In healthy state, the
Angiotensin II–Angiotensin 1
Receptor axis and the
Angiotensin 1–7-Mas receptor
axis are in a state of dynamic
equilibrium to maintain the
blood pressure. The former
causes an inflammatory
response while the latter
suppresses inflammatory
responses. bIn SARS-CoV-2
infected state, viral binding to
ACE2 renders it unavailable to
bind to Angiotensin II causing
an imbalance between the two
axes and a shift towards the pro-
inflammatory functions
Ind J Clin Biochem
123
II is known to significantly increase the expression of IL-6
mRNA and protein in a dose-dependent manner [48].
Angiotensin II binds to angiotensin I receptor which
then initiates a downstream signaling that leads to oxida-
tive stress, formation of reactive oxygen species (ROS) and
upregulated expression of IL-6 [49]. Conversely, IL-6
released by macrophages promotes enhanced expression of
Angiotensin 1 receptor, which attaches itself to walls of
endothelial cells leading to cell and vascular inflammation
[50]. This, in turn, creates a redox imbalance, causing an
increase in oxidative burden and IL-6 expression. Hence,
IL-6 and Angiotensin II activate each other by exerting a
positive feedback control. Treatment of COVID-19
patients with RAAS inhibitors such as ACE inhibitors
(ACEIs) and angiotensin II type 1 receptor blockers
(ARBs) led to attenuation of the inflammatory response, as
observed through the inhibition of IL-6 levels, increased
CD3 and CD8 T cell counts in peripheral blood and
decreased viral load [46,51].
IL-6: Predictor of Disease Prognosis
and Associated Risk Factors in COVID-19 Patients
Elevated IL-6 serum levels are linked to poorer prognosis
and deteriorating clinical outcomes in COVID-19 patients.
It has been indicated that moderately elevated levels of IL-
6 above 80 pg/ml were adequate to identify COVID-19
patients with a high risk of respiratory failure [23]. Fur-
thermore, RNAaemia (SARS CoV2 nucleic acid) in serum
was strongly associated with cytokine storm and an
immensely high IL-6 serum levels [52]. Old age, high
SOFA score (Sequential Organ Failure Assessment), high
BMI, elevated D-dimers and high IL-6 levels were proven
risk factors for increase in mortality & poor prognosis in
adult patients [53]. A retrospective study conducted on ICU
patients revealed significantly high levels of IL-6 in non-
survivor group compared to survivor with AUROC of 0.73
[54]. Another, two hospital based retrospective study also
revealed higher baseline IL-6 levels in severe cases [55]. A
study generated two predictive models based on high flow
oxygen requirement which also demonstrated that patients
with higher IL-6 values are early identification markers of
COVID-19 severity [56]. It was also significantly related
with elevated levels of AST, ALT and GGT during hos-
pitalization of COVID-19 patients [57]. Higher IL-6 levels
has also been linked to long term care need in elderly
patients and in-hospital mortality rates [61]. A study con-
ducted on 225 COVID-19 patients in Southwest quaternary
United states hospital found that elevated levels of IL-6
positively correlates with adverse cardiac events or deaths
[58].
Among the predisposing conditions, diabetes emerged
as the most important association in COVID-19 patients
with mortality rate of 16–35% [59]. Recent data from
COVID-19 patients with diabetes have shown elevated
levels of systemic IL-6 compared to patients without dia-
betes [60]. SARS-CoV-2 infection may exacerbate pre-
existing diabetes, as ACE2 expression in pancreatic cells
contributes to impaired insulin secretion as well as insulin
resistance [61]. Thus, IL-6 may govern COVID-19 disease
severity in diabetics. Similarly, obese patients with
COVID-19 are more likely to develop severe respiratory
distress syndrome [62]. They have higher concentration of
pro-inflammatory cytokines both in steady state and in
diseases [63]. Further, obese patients tend to have height-
ened IL-6 production in adipocytes leading to decreased
anti-viral immune response by neutrophils and hence,
uncontrolled viral replication at early stages of infection
[63]. Age is another risk factor for poorer prognosis in
COVID-19 [53]. This age-associated vulnerability is
probably due to increased expression of ACE2 with age
accounting for higher mortality in elderly patients [64].
Smoking has also been identified as a risk factor for
SARS-CoV-2 infection. It has already been stated that
active smokers are more susceptible to influenza virus
infections and are more likely to develop chronic
obstructive pulmonary disease (COPD) [65]. They have
increased ACE2 expression in the lungs, which may
potentially explain a fourfold increased risk of develop-
ment of severe COVID-19 in smokers [66,67]. During
evaluation of risk factors associated with COVID-19 in a
Chinese cohort, it was found that 19.2% of patients were
smokers [68].
COVID-19 mortality rates in males are twice that of
females [69]. Similar data on association of male gender
with poorer prognosis were obtained earlier for SARS-CoV
and MERS-CoV infections [70,71]. The reason behind this
gender difference lies in the ACE2 gene which is an
X-linked gene. During X-chromosome inactivation, a sig-
nificant number of genes undergo dosage compensation.
However, ACE2, escapes X chromosome inactivation, thus
contributing to gender disparity in disease susceptibility
[72]. Endocrine factors may further contribute to gender
disparity in immune responses [73,74]. Viral load in
COVID-19 females is lower in comparison to males, with
concomitant higher CD4
?
T cells [79]. A comparative
analysis depicted that course of COVID-19 is more severe
in males. The number of men who died from COVID-19 is
2.4 times that of females (70.3% vs. 29.7%). This sug-
gested that male patients are at increased risk of worse
outcomes and death compared to female patients, inde-
pendent of age [75]. A study also concluded that levels of
IL-6 were significantly elevated in males compared to
females. Also, a higher number of males were reported to
Ind J Clin Biochem
123
experience lymphopenia [76]. Therefore, immune-modu-
latory effects of hormones such as estrogens contribute to
decrease susceptibility and better prognosis in women
infected with SARS-CoV-2.
Anti-IL-6 Agents: Therapeutic Targets
for COVID-19 Infection
A diverse spectrum of pharmaceutical agents is presently
being employed for the treatment and management of
COVID-19 infection. Monoclonal antibody Tocilizumab
an IL-6 receptor antibody has been advocated for treatment
[7779].
While antivirals like remdesivir specifically target the
viral replication by inhibiting the RNA-dependant-RNA
polymerase [80], other drugs like Hydroxychloroquine
inhibit the CS but have adverse effects like gastrointestinal
complications, retinopathy and QT interval prolongation in
treated COVID-19 patients [81]. Thus, in addition to
antiviral drugs, ameliorating the CS would prove to be an
efficient treatment strategy to successfully combat the
disease.
Plant-derived natural immunosuppressant compounds,
such as curcumin, luteolin, piperine, resveratrol, allicin,
colchicines, eugenol etc. have been known to inhibit the
pro-inflammatory cytokines and chemokines. This specific
targeting of cytokines is achieved by the inhibitory action
on specific signalling cascades like NF-jB, JAK/STAT,
MAPK/ERK [82]. Use of these plants sources with
enhanced bioavailability and safety profiles is a ray of hope
and opens up a novel approach to mitigate the CS in
COVID-19.
The latest treatment molecules for combating the CS
have been the monoclonal antibodies (MoAbs) emerging as
the versatile class of biotherapeutics for passive
immunotherapy. The use of monoclonal antibodies against
viral infections like influenza and rheumatoid arthritis have
already shown promising results [83]. Since IL-6 is found
to be significantly upregulated in COVID-19 infection,
MoAbs that can neutralize its effects may serve as a
potential treatment option as depicted in Fig. 2.
Tocilizumab is a recombinant humanised monoclonal
antibody of the IgG1 class, which is directed against both
the soluble and membrane-bound forms of the interleukin-
6 (IL-6) receptor.
Small retrospective cohort studies on patients affected
by severe COVID-19 demonstrated that treatment with
Tocilizumab improved the clinical profile of COVID-19
patients [84]. Patients with severe respiratory failure dis-
playing macrophage activation syndrome (MAS) showed
decreased expression of human leucocyte antigen-D (HLA-
DR) which was mediated by IL-6 expression. Treatment of
these patients with Tocilizumab partially restored HLA-DR
expression [85]. A case study of 64 year old male treated
with tocilizumab for hemophagocytic lymphohistocytosis
syndrome and COVID-19 has also shown decreased levels
of inflammatory markers including IL-6 [86]. Tocilizumab
has been recommended for use in severe or critically ill
patients with confirmed elevated levels of IL-6 in the
‘Diagnosis and Treatment Protocol for Novel Coronavirus
Pneumonia (Trial Version 7)’ issued by the National
Health Commission of China [87]. Several clinical trials on
the use of Tocilizumab have been attempted in different
COVID-19 population groups ranging from patients with
pneumonia to life-threatening CS associated condition
(available on ClinicalTrials.gov.in).
Sarilumab, a human MoAb against IL-6 receptor, is
presently in use for the treatment of RA patients [88].
Addition of Sarilumab in the treatment regimen of eight
patients diagnosed positive for SARS-CoV-2 was done and
a significant improvement in respiratory function (30%
reduction in oxygen requirement compared to baseline)
was observed leading to an early discharge, merely 14 days
after hospital admission to hospital [89].
Siltuximab, an anti-IL-6 antibody, has been evaluated in
the Siltuximab in Severe COVID-19 (SISCO) study.
Mortality rates were compared in two cohorts consisting of
patients on treatment regimen with or without siltuximab
and it was found that siltuximab receiving patients exhib-
ited a decreased mortality rate. Another anti- IL-6 MoAb
which is presently suggested as an effective treatment
option for RA and psoriatic arthritis is Clazakizumab
[90,91]. Significant improvement has been observed in
respiratory functions, inflammatory markers and oxygen
requirements in COVID-19 patients with significantly
raised levels of CRP and IL-6 after treatment with Claza-
kizumab [92].
The effects of cytokines involved in COVID-19 induced
CS are mediated via the JAK/STAT cascade as illustrated
in Fig. 2. Thus, CS in SARS-CoV-2 can also be alleviated
using JAK inhibitors (JAKi). Ruxolitinib is the first FDA
approved JAK inhibitor that inhibits both JAK1 and 2 [93].
It was observed that patients who received Ruxolitinib had
a faster clinical and chest CT improvement and displayed
significantly decreased levels of cytokines compared to
control group. Moreover, no deaths were recorded in the
Ruxolitinib receiving group (n = 20) while three patients
died due to respiratory failure in the control group (n = 21)
[94]. Another JAKi, Baricitinib has been suggested to be of
therapeutic use against SARS-CoV-2 [95]. Baricitinib-
treated COVID-19 patients achieve greater improvements
in all clinical characteristics (fever, cough and dyspnea)
and respiratory function parameters compared to baseline.
A substantial number of trials on anti-IL6 agents that have
Ind J Clin Biochem
123
been presently undertaken will pave the path for treatment
strategies in the days to come.
Conclusion and Future Perspectives
In conclusion, the role of IL-6 in the immunopathology of
COVID-19 is pivotal. IL-6 occupies the centre stage in
initiating and potentiating the dreaded CS. It also helps in
predicting disease severity & mortality in COVID-19.
Raised IL-6 levels were associated with ARDS, increased
requirement of mechanical ventilation, prolonged hospital
stay, worse SOFA score, multiple organ impairment and
intensive care unit admission. This review elaborated IL-6
levels in preference to other cytokines raised in cytokine
storm because anti IL-6 antibodies and IL-6 receptor
inhibitors have already been in vogue for the treatment of
autoimmune diseases and now have been repurposed for
the treatment of COVID-19 with some success. In addition,
JAK inhibitors are also being tried in clinical trials for the
treatment of COVID-19.
Molecules that block ADAM 10 hold a lot of promise
since they tilt the inflammatory balance from pro-inflam-
matory to anti-inflammatory pathway thus abetting the
cytokine storm which is the main pathological event that
propels SARS-COVID-19 patients into a downhill course.
Further studies on genetic polymorphisms in various ethnic
groups which affect IL-6 levels need to be conducted for
stratification of COVID patients into mild, moderate and
severe. Delineating such genetic polymorphisms may also
pave the path for pharmacogenomic database for the
exhibition of anti-IL-6 antibodies.
Author contributions JS: Data acquisition, original draft, Editing.
KG: Data creation, resources, writing, original draft. SG: Review and
editing, data creation. PP: Review and editing. MR: Review and
editing. PM: Review and editing. MG: Data creation, review and
editing. SM: Supervision. PS: Review and editing, supervision. MB:
Conceptualization, supervision, review and editing.
Funding This review article did not receive any specific grant from
funding agencies in the public, commercial sectors.
Declarations
Conflict of interest The authors declare that they have no conflict of
interest to disclose.
References
1. WHO Pneumonia of unknown cause China [Internet]. WHO.
World Health Organization; [cited 2020 Aug 14]. http://www.
who.int/csr/don/05-january-2020-pneumonia-of-unkown-cause-
china/en/.
2. COVID-19 Public Health Emergency of International Concern
(PHEIC) Global research and innovation forum [Internet]. [cited
2020]. https://www.who.int/publications/m/item/covid-19-public-
health-emergency-of-international-concern-(pheic)-global-
research-and-innovation-forum.
3. de Wit E, van Doremalen N, Falzarano D, Munster VJ. SARS and
MERS: recent insights into emerging coronaviruses. Nat Rev
Microbiol. 2016;14(8):523–34.
4. Li X, Geng M, Peng Y, Meng L, Lu S. Molecular immune
pathogenesis and diagnosis of COVID-19. J Pharm Anal.
2020;10(2):102–8.
5. Lu L, Zhong W, Bian Z, Li Z, Zhang K, Liang B, et al. A
comparison of mortality-related risk factors of COVID-19,
SARS, and MERS: a systematic review and meta-analysis. J In-
fect. 2020;81(4):e18-25.
6. Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L, Zhang W, et al.
A pneumonia outbreak associated with a new coronavirus of
probable bat origin. Nature. 2020;579(7798):270–3.
7. Valencia DN. Brief review on COVID-19: the 2020 pandemic
caused by SARS-CoV-2. Cureus [Internet]. 2020. https://doi.org/
10.7759/cureus.7386.
8. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical
features of patients infected with 2019 novel coronavirus in
Wuhan China. Lancet. 2020;395(10223):497–506.
9. Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, et al. Structure of the
SARS-CoV-2 spike receptor-binding domain bound to the ACE2
receptor. Nature. 2020;581(7807):215–20.
10. Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, et al. Characterization of
spike glycoprotein of SARS-CoV-2 on virus entry and its
immune cross-reactivity with SARS-CoV. Nat Commun.
2020;11(1):1620.
11. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor
H. Tissue distribution of ACE2 protein, the functional receptor
for SARS coronavirus. a first step in understanding SARS
pathogenesis. J Pathol. 2004;203(2):631–7.
12. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA,
Muley T, et al. SARS -CoV-2 receptor ACE 2 and TMPRSS 2 are
primarily expressed in bronchial transient secretory cells. EMBO
J. 2020. https://doi.org/10.15252/embj.20105114.
13. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, et al.
Kidney impairment is associated with in-hospital death of
COVID-19 patients [Internet]. Nephrology. 2020. https://doi.org/
10.1101/2020.02.18.20023242.
14. Robba C, Battaglini D, Pelosi P, Rocco PRM. Multiple organ
dysfunction in SARS-CoV-2: MODS-CoV-2. Expert Rev Respir
Med. 2020;22:1–4.
15. Hoffmann M, Kleine-Weber H, Schroeder S, Kru
¨ger N, Herrler
T, Erichsen S, et al. SARS-CoV-2 cell entry depends on ACE2
and TMPRSS2 and Is blocked by a clinically proven protease
inhibitor. Cell. 2020;181(2):271-280.e8.
16. Muniyappa R, Gubbi S. COVID-19 pandemic, coronaviruses, and
diabetes mellitus. Am J Physiol-Endocrinol Metab.
2020;318(5):E736–41.
17. Gubernatorova EO, Gorshkova EA, Polinova AI, Drutskaya MS.
IL-6: relevance for immunopathology of SARS-CoV-2. Cytokine
Growth Factor Rev. 2020;53:13–24.
18. Moore JB, June CH. Cytokine release syndrome in severe
COVID-19. Science. 2020;368(6490):473–4.
19. Shimabukuro-Vornhagen A, Go
¨del P, Subklewe M, Stemmler HJ,
Schlo
¨ßer HA, Schlaak M, et al. Cytokine release syndrome. J
immuno cancer. 2018;6(1):56.
20. Shimizu M. Clinical features of cytokine storm syndrome. In:
Cron RQ, Behrens EM, editors. Cytokine storm syndrome. Cham:
Springer International Publishing; 2019. p. 31–41. https://doi.org/
10.1007/978-3-030-22094-5_3.
Ind J Clin Biochem
123
21. Du F, Liu B, Zhang S. COVID-19: the role of excessive cytokine
release and potential ACE2 down-regulation in promoting
hypercoagulable state associated with severe illness. J Thromb
Thrombolysis. 2020. https://doi.org/10.1007/s11239-020-02224-
2.
22. Picchianti Diamanti A, Rosado MM, Pioli C, Sesti G, Lagana
`B.
Cytokine release syndrome in COVID-19 patients, a new scenario
for an old concern: the fragile balance between infections and
autoimmunity. IJMS. 2020;21(9):3330.
23. Herold T, Jurinovic V, Arnreich C, Lipworth BJ, Hellmuth JC,
von Bergwelt-Baildon M, et al. Elevated levels of IL-6 and CRP
predict the need for mechanical ventilation in COVID-19. J Al-
lergy Clin Immunol. 2020;146(1):128-136.e4.
24. Rose-John S. Interleukin-6 biology is coordinated by membrane-
bound and soluble receptors: role in inflammation and cancer.
J Leukoc Biol. 2006;80(2):227–36.
25. Bo
¨ttcher JP, Schanz O, Garbers C, Zaremba A, Hegenbarth S,
Kurts C, et al. IL-6 trans-signaling-dependent rapid development
of cytotoxic CD8?T cell function. Cell Rep. 2014;8(5):1318–27.
26. Cox MA, Kahan SM, Zajac AJ. Anti-viral CD8 T cells and the
cytokines that they love. Virology. 2013;435(1):157–69.
27. Evans SS, Repasky EA, Fisher DT. Fever and the thermal reg-
ulation of immunity: the immune system feels the heat. Nat Rev
Immunol. 2015;15(6):335–49.
28. Pyle CJ, Uwadiae FI, Swieboda DP, Harker JA. Early IL-6 sig-
nalling promotes IL-27 dependent maturation of regulatory T
cells in the lungs and resolution of viral immunopathology. PLoS
Pathog. 2017;13(9):e1006640.
29. Yang M-L, Wang C-T, Yang S-J, Leu C-H, Chen S-H, Wu C-L,
et al. IL-6 ameliorates acute lung injury in influenza virus
infection. Sci Rep. 2017;7(1):43829.
30. Lauder SN, Jones E, Smart K, Bloom A, Williams AS, Hindley
JP, et al. Interleukin-6 limits influenza-induced inflammation and
protects against fatal lung pathology: immunity to infection. Eur J
Immunol. 2013;43(10):2613–25.
31. Velazquez-Salinas L, Verdugo-Rodriguez A, Rodriguez LL,
Borca MV. The role of interleukin 6 during viral infections. Front
Microbiol. 2019;10(10):1057.
32. Dienz O, Rincon M. The effects of IL-6 on CD4 T cell responses.
Clin Immunol. 2009;130(1):27–33.
33. Hou W, Kang HS, Kim BS. Th17 cells enhance viral persistence
and inhibit T cell cytotoxicity in a model of chronic virus
infection. J Exp Med. 2009;206(2):313–28.
34. Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-
Lartigue J, Crawford JM, et al. Targeting potential drivers of
COVID-19: neutrophil extracellular traps. J Exp Med.
2020;217(6):e20200652.
35. Wu W, Dietze KK, Gibbert K, Lang KS, Trilling M, Yan H, et al.
TLR ligand induced IL-6 counter-regulates the anti-viral CD8?
T cell response during an acute retrovirus infection. Sci Rep.
2015;5(1):10501.
36. Indalao IL, Sawabuchi T, Takahashi E, Kido H. IL-1bis a key
cytokine that induces trypsin upregulation in the influenza virus–
cytokine–trypsin cycle. Arch Virol. 2017;162(1):201–11.
37. Schumacher R-J. ADAM17 activity and IL-6 trans-signaling in
inflammation and cancer. Cancers. 2019;11(11):1736.
38. Chalaris A, Garbers C, Rabe B, Rose-John S, Scheller J. The
soluble Interleukin 6 receptor: generation and role in inflamma-
tion and cancer. Eur J Cell Biol. 2011;90(6–7):484–94.
39. Rose-John S. IL-6 trans-signaling via the soluble IL-6 receptor:
importance for the pro-inflammatory activities of IL-6. Int J Biol
Sci. 2012;8(9):1237–47.
40. Tenhumberg S, Waetzig GH, Chalaris A, Rabe B, Seegert D,
Scheller J, et al. Structure-guided optimization of the interleukin-
6 trans-signaling antagonist sgp130. J Biol Chem.
2008;283(40):27200–7.
41. Jostock T, Mu
¨llberg J, O
¨zbek S, Atreya R, Blinn G, Voltz N,
et al. Soluble gp130 is the natural inhibitor of soluble interleukin-
6 receptor transsignaling responses: soluble gp130 inhibits
transsignaling responses. Eur J Biochem. 2001;268(1):160–7.
42. Schett G. Physiological effects of modulating the interleukin-6
axis. Rheumatology. 2018;57(suppl_2):ii43–50.
43. Furuya Y, Satoh T, Kuwana M. Interleukin-6 as a potential
therapeutic target for pulmonary arterial hypertension. Int J
Rheumatol. 2010;2010:1–8.
44. Zheng Y-Y, Ma Y-T, Zhang J-Y, Xie X. COVID-19 and the
cardiovascular system. Nat Rev Cardiol. 2020;17(5):259–60.
45. Hampl V, Herget J,
´bova
´J, Ban
ˇasova
´A, Huskova
´Z,
Van
ˇourkova
´Z, et al. Intrapulmonary activation of the angio-
tensin-converting enzyme type 2/angiotensin 1–7/G-protein-
coupled mas receptor axis attenuates pulmonary hypertension in
Ren-2 transgenic rats exposed to chronic hypoxia. Physiol Res.
2015;27:25–38.
46. Meng J, Xiao G, Zhang J, He X, Ou M, Bi J, et al. Renin-
angiotensin system inhibitors improve the clinical outcomes of
COVID-19 patients with hypertension. Emerg Microb Infect.
2020;9(1):757–60.
47. Liu Y, Yang Y, Zhang C, Huang F, Wang F, Yuan J, et al.
Clinical and biochemical indexes from 2019-nCoV infected
patients linked to viral loads and lung injury. Sci China Life Sci.
2020;63(3):364–74.
48. Funakoshi Y, Ichiki T, Ito K, Takeshita A. Induction of inter-
leukin-6 expression by angiotensin II in rat vascular smooth
muscle cells. Hypertension. 1999;34(1):118–25.
49. Gomolak JR, Didion SP. Angiotensin II-induced endothelial
dysfunction is temporally linked with increases in interleukin-6
and vascular macrophage accumulation. Front Physiol. 2014.
https://doi.org/10.3389/fphys.2014.00396/abstract.
50. Liu PP, Blet A, Smyth D, Li H. The science underlying COVID-
19: implications for the cardiovascular system. Circulation.
2020;142(1):68–78.
51. Yang G, Tan Z, Zhou L, Yang M, Peng L, Liu J, et al. Angio-
tensin II receptor blockers and angiotensin-converting enzyme
inhibitors usage is associated with improved inflammatory status
and clinical outcomes in COVID-19 patients with hypertension
[Internet]. Infect Dis (Except HIV/AIDS). 2020. https://doi.org/
10.1101/2020.03.31.20038935.
52. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, et al.
Detectable serum severe acute respiratory syndrome coronavirus
2 viral load (RNAemia) is closely correlated with drastically
elevated interleukin 6 level in critically Ill patients with coron-
avirus disease 2019. Clin Infect Dis. 2020;71:449.
53. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course
and risk factors for mortality of adult inpatients with COVID-19
in Wuhan, China: a retrospective cohort study. Lancet.
2020;395(10229):1054–62.
54. Gorham J, Moreau A, Corazza F, Peluso L, Ponthieux F, Tala-
monti M, et al. Interleukine-6 in critically ill COVID-19 patients:
a retrospective analysis. PLOS ONE. 2020;15(12):e0244628.
55. Zhu W, Wu Y, Liu Y, Li B, Xiong P, Cui D, et al. Baseline
interleukin-6 level predicts risk of severe COVID-19: a two-
center, retrospective study [Internet]. In Review; 2020 Nov [cited
2021 Jan 11]. https://www.researchsquare.com/article/rs-68501/
v1.
56. Riveiro-Barciela M, Labrador-Horrillo M, Camps-Relats L,
Gonza
´lez-Sans D, Ventura-Cots M, Terrones-Peinador M, et al.
Simple predictive models identify patients with COVID-19
pneumonia and poor prognosis. PLOS ONE.
2020;15(12):e0244627.
57. Xiang D, Ren X, Chen Q, Yu H, Li X, Liu D. Association of
ACEI/ARB, inflammatory cytokines, and antiviral drugs with
Ind J Clin Biochem
123
liver dysfunction in patients with hypertension and COVID-19.
Clin Exp Hyper. 2020;1–6.
58. Nicholas Nguyen, Hao Nguyen, Chirag Patel, Farukh Ikram,
Priyanka Acharya, Anmol Dhillion, et al. (2020) Abstract 17279:
elevated interleukin-6 levels in COVID-19-infected patients are
associated with major adverse cardiac events and/or mortality.
Circulation. 2020;142(Suppl_3):A17279–A17279.
59. Guo W, Li M, Dong Y, Zhou H, Zhang Z, Tian C, et al. Diabetes
is a risk factor for the progression and prognosis of COVID-19.
Diabetes Metab Res Rev. 2020, e3319.
60. Zhu L, She Z-G, Cheng X, Qin J-J, Zhang X-J, Cai J, et al.
Association of blood glucose control and outcomes in patients
with COVID-19 and pre-existing type 2 diabetes. Cell Metab.
2020;31(6):1068-1077.e3.
61. Bindom SM, Lazartigues E. The sweeter side of ACE2: physio-
logical evidence for a role in diabetes. Mol Cell Endocrinol.
2009;302(2):193–202.
62. Banerjee M, Gupta S, Sharma P, Shekhawat J, Gauba K. Obesity
and COVID-19: a fatal alliance. Ind J Clin Biochem.
2020;35(4):410–7.
63. Richard C, Wadowski M, Goruk S, Cameron L, Sharma AM,
Field CJ. Individuals with obesity and type 2 diabetes have
additional immune dysfunction compared with obese individuals
who are metabolically healthy. BMJ Open Diab Res Care.
2017;5(1):e000379.
64. Baker SA, Kowk S, Berry GJ, Montine TJ. Angiotensin-con-
verting enzyme 2 (ACE2) expression increases with age in
patients requiring mechanical ventilation [Internet]. Pathology.
2020. https://doi.org/10.1101/2020.07.05.20140467.
65. Lawrence H, Hunter A, Murray R, Lim WS, McKeever T.
Cigarette smoking and the occurrence of influenza: systematic
review. J Infect. 2019;79(5):401–6.
66. Leung JM, Yang CX, Tam A, Shaipanich T, Hackett T-L, Sin-
ghera GK, et al. ACE-2 expression in the small airway epithelia
of smokers and COPD patients: implications for COVID-19. Eur
Respir J. 2020;55(5):2000688.
67. Brake SJ, Barnsley K, Lu W, McAlinden KD, Eapen MS, Sohal
SS. Smoking upregulates angiotensin-converting enzyme-2
receptor: a potential adhesion site for novel coronavirus SARS-
CoV-2 (Covid-19). JCM. 2020;9(3):841.
68. Zhao Q, Meng M, Kumar R, Wu Y, Huang J, Lian N, et al. The
impact of COPD and smoking history on the severity of COVID-
19: a systemic review and meta-analysis. J Med Virol.
2020;92(10):1915–21.
69. Wu Z, McGoogan JM. Characteristics of and important lessons
from the coronavirus disease 2019 (COVID-19) outbreak in
China: summary of a report of 72 314 cases from the chinese
center for disease control and prevention. JAMA.
2020;323(13):1239.
70. Karlberg J. Do men have a higher case fatality rate of severe
acute respiratory syndrome than women do? Am J Epidemiol.
2004;159(3):229–31.
71. Badawi A, Ryoo SG. Prevalence of comorbidities in the Middle
East respiratory syndrome coronavirus (MERS-CoV): a system-
atic review and meta-analysis. Int J Infect Dis. 2016;49:129–33.
72. Burrell LM, Harrap SB, Velkoska E, Patel SK. The ACE2 gene:
its potential as a functional candidate for cardiovascular disease.
Clin Sci. 2013;124(2):65–76.
73. Klein SL, Flanagan KL. Sex differences in immune responses.
Nat Rev Immunol. 2016;16(10):626–38.
74. La Vignera S, Cannarella R, Condorelli RA, Torre F, Aversa A,
Calogero AE. Sex-specific SARS-CoV-2 mortality: among hor-
mone-modulated ACE2 expression, risk of venous thromboem-
bolism and hypovitaminosis D. IJMS. 2020;21(8):2948.
75. Jin J-M, Bai P, He W, Wu F, Liu X-F, Han D-M, et al. Gender
differences in patients with COVID-19: focus on severity and
mortality. Front Public Health. 2020;29(8):152.
76. Lv Z, Cheng S, Le J, Huang J, Feng L, Zhang B, et al. Clinical
characteristics and co-infections of 354 hospitalized patients with
COVID-19 in Wuhan, China: a retrospective cohort study.
Microb Infect. 2020;22(4–5):195–9.
77. Antiviral Therapy [Internet]. COVID-19 treatment guidelines.
[cited 2021 Jun 3]. https://www.covid19treatmentguidelines.nih.
gov/antiviral-therapy/.
78. Sethi A, Bach H. Evaluation of current therapies for COVID-19
treatment. Microorganisms. 2020;8(8):1097.
79. Tobaiqy M, Qashqary M, Al-Dahery S, Mujallad A, Hershan AA,
Kamal MA, et al. Therapeutic management of patients with
COVID-19: a systematic review. Infect Prev Pr.
2020;2(3):100061.
80. Kokic G, Hillen HS, Tegunov D, Dienemann C, Seitz F, Sch-
mitzova J, et al. Mechanism of SARS-CoV-2 polymerase stalling
by remdesivir. Nat Commun. 2021;12(1):279.
81. Satarker S, Ahuja T, Banerjee M, Balaji EV, Dogra S, Agarwal T,
et al. Hydroxychloroquine in COVID-19: potential mechanism of
action against SARS-CoV-2. Curr Pharmacol Rep.
2020;6(5):203–11.
82. Peter AE, Sandeep BV, Rao BG, Kalpana VL. Calming the
storm: natural immunosuppressants as adjuvants to target the
cytokine storm in COVID-19. Front Pharmacol. 2021;11:583777.
83. Perspectives on monoclonal antibody therapy as potential thera-
peutic intervention for Coronavirus disease-19 (COVID-19).
Asian Pac J Allergy Immunol [Internet]. 2020 [cited 2020 May
26]. http://apjai-journal.org/wp-content/uploads/2020/03/2.pdf.
84. Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective
treatment of severe COVID-19 patients with tocilizumab. Proc
Natl Acad Sci USA. 2020;117(20):10970–5.
85. Giamarellos-Bourboulis EJ, Netea MG, Rovina N, Akinosoglou
K, Antoniadou A, Antonakos N, et al. Complex immune dys-
regulation in COVID-19 patients with severe respiratory failure.
Cell Host Microbe. 2020. https://doi.org/10.1016/j.chom.2020.04.
009.
86. Eroglu A, Kartal S, Saral OB. Helmet mask and tocilizumab for a
patient with hemophagocytic lymphohistiocytosis syndrome and
COVID-19: a case report. Braz J Anesthesiol (English Edition)
[Internet]. 2020 Dec 26 [cited 2021 Jan 11]. http://www.science
direct.com/science/article/pii/S0104001420302153.
87. (Released by National Health Commission & National Admin-
istration of Traditional Chinese Medicine on March 3, 2020).
Diagnosis and treatment protocol for novel coronavirus pneu-
monia (Trial Version 7): Chinese Medical Journal. 2020.
133(9):1087–95.
88. Lamb YN, Deeks ED. Sarilumab: a review in moderate to severe
rheumatoid arthritis. Drugs. 2018;78(9):929–40.
89. Benucci M, Giannasi G, Cecchini P, Gobbi FL, Damiani A,
Grossi V, et al. COVID-19 pneumonia treated with sarilumab: a
clinical series of eight patients. J Med Virol. 2020. jmv.26062.
90. Weinblatt ME, Mease P, Mysler E, Takeuchi T, Drescher E,
Berman A, et al. The efficacy and safety of subcutaneous
clazakizumab in patients with moderate-to-severe rheumatoid
arthritis and an inadequate response to methotrexate: results from
a multinational, phase IIb, randomized, double-blind, placebo/
active-controlled, dose-Ra: clazakizumab in moderate-to-severe
Ra. Arthritis Rheumatol. 2015;67(10):2591–600.
91. Bilal J, Riaz IB, Kamal MU, Elyan M, Sudano D, Khan MA.
A Systematic review and meta-analysis of efficacy and safety of
novel interleukin inhibitors in the management of psoriatic
arthritis. JCR: J Clin Rheumatol. 2018;24(1):6–13.
92. Vaidya G, Czer LSC, Kobashigawa J, Kittleson M, Patel J, Chang
D, et al. Successful Treatment of severe COVID-19 pneumonia
Ind J Clin Biochem
123
with clazakizumab in a heart transplant recipient: a case report.
transplantation proceedings. 2020. S0041134520325756.
93. Bjørn ME, Hasselbalch HC. The impact of ruxolitinib treatment
on inflammation-mediated comorbidities in myelofibrosis and
related neoplasms. Clin Case Rep. 2015;3(6):499–503.
94. Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L, et al. Ruxolitinib
in treatment of severe coronavirus disease 2019 (COVID-19): A
multicenter, single-blind, randomized controlled trial. J Allergy
Clin Immunol. 2020. https://doi.org/10.1016/j.jaci.2020.05.019.
95. Winthrop KL. The emerging safety profile of JAK inhibitors in
rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–43.
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Ind J Clin Biochem
123
... Again, the exact characterization of the leukocyte subsets involved here would require performing specific immune assays and has to be addressed in future research. With regard to IL-6, a well-known marker of the inflammatory response caused by SARS-CoV-2, 45 there was a trend toward a positive association between IL-6 and choroid plexus volume. ...
Article
Full-text available
Objective To investigate whether choroid plexus volumes in subacute coronavirus disease 2019 (COVID‐19) patients with neurological symptoms could indicate inflammatory activation or barrier dysfunction and assess their association with clinical data. Methods Choroid plexus volumes were measured in 28 subacute COVID‐19 patients via cerebral magnetic resonance imaging (MRI), compared with those in infection‐triggered non‐COVID‐19 encephalopathy patients (n = 25), asymptomatic individuals after COVID‐19 (n = 21), and healthy controls (n = 21). Associations with inflammatory serum markers (peak counts of leukocytes, C‐reactive protein [CRP], interleukin 6), an MRI‐based marker of barrier dysfunction (CSF volume fraction [V‐CSF]), and clinical parameters like olfactory performance and cognitive scores (Montreal Cognitive Assessment) were investigated. Results COVID‐19 patients showed significantly larger choroid plexus volumes than control groups ( p < 0.001, η ² = 0.172). These volumes correlated significantly with peak leukocyte levels ( p = 0.001, Pearson's r = 0.621) and V‐CSF ( p = 0.009, Spearman's rho = 0.534), but neither with CRP nor interleukin 6. No significant correlations were found with clinical parameters. Interpretation In patients with subacute COVID‐19, choroid plexus volume is a marker of central nervous system inflammation and barrier dysfunction in the presence of neurologic symptoms. The absence of plexus enlargement in infection‐triggered non‐COVID‐19 encephalopathy suggests a specific severe acute respiratory syndrome coronavirus 2 effect. This study also documents an increase in choroid plexus volume for the first time as a parainfectious event. ANN NEUROL 2024
... By stimulating T-cell and M1 macrophages, IL-6 likely exerts a protective function against viral replication in asymptomatic infection and also against initial tissue destruction by altering granzyme and cytokine release in mild-moderate illness. Of note, IL-6 may be more pathogenic in severe disease due to the induction of proinflammatory cells (components of so-called cytokines storms) (Kang and Kishimoto, 2021;Shekhawat et al., 2021). This will bear careful study in future clinical trials in various stages of infection. ...
... IL-6's value extends beyond the scope of COVID-19, enriching the evaluation of cytokine release syndrome linked to CAR-T cell therapy and other diseases [71]. Specifically, in COVID-19, IL-6 is considered the most accurate marker for tracking disease progression and predicting mortality among cytokines associated with cytokine storms [72]. IL-6 often demonstrates enhanced predictive power for disease progression over non-cytokine inflammatory markers like C-reactive protein, highlighting its critical role in cytokine storm evaluation [73]. ...
Article
Full-text available
Background As a fatal disease, cytokine storm has garnered research attention in recent years. Nonetheless, as the body of related studies expands, a thorough and impartial evaluation of the current status of research on cytokine storms remains absent. Consequently, this study aimed to thoroughly explore the research landscape and evolution of cytokine storm utilizing bibliometric and knowledge graph approaches. Methods Research articles and reviews centered on cytokine storms were retrieved from the Web of Science Core Collection database. For bibliometric analysis, tools such as Excel 365, CiteSpace, VOSviewer, and the Bibliometrix R package were utilized. Results This bibliometric analysis encompassed 6647 articles published between 2004 and 2022. The quantity of pertinent articles and citation frequency exhibited a yearly upward trend, with a sharp increase starting in 2020. Network analysis of collaborations reveals that the United States holds a dominant position in this area, boasting the largest publication count and leading institutions. Frontiers in Immunology ranks as the leading journal for the largest publication count in this area. Stephan A. Grupp, a prominent researcher in this area, has authored the largest publication count and has the second-highest citation frequency. Research trends and keyword evaluations show that the connection between cytokine storm and COVID-19, as well as cytokine storm treatment, are hot topics in research. Furthermore, research on cytokine storm and COVID-19 sits at the forefront in this area. Conclusion This study employed bibliometric analysis to create a visual representation of cytokine storm research, revealing current trends and burgeoning topics in this area for the first time. It will provide valuable insights, helping scholars pinpoint critical research areas and potential collaborators.
... It has been proven that as a result of COVID-19 infection, the epithelium is damaged while IL-1α and IL-1β concentration increases. Elevated levels of IL-6, TNF-α and IFN-γ in COVID-19 patient could be significant in forming an overacting immune response, especially IL-6 (22). Despite this connection in elevated values of IL-6, IL-1α, TNF-α and IFN-γ the pathophysiological mechanism remains not fully elucidated, because their values are elevated in many other diseases. ...
... Moreover, the patients in the ketamine group had shorter overall hospital LOS. This might be explained by the interesting properties of ketamine as an immunomodulator and its ability to reduce inflammatory cytokines such as IL-6 (De Kock et al., 2013), which is known to be elevated during COVID-19 illness (cytokine storming) and was found to be associated with an increased risk of mortality (Shekhawat et al., 2021). Another factor that might have contributed to our findings of shorter length of hospital stay could be related to reduced incidence of delirium with low doses of ketamine (Perbet et al., 2018). ...
Article
Full-text available
Backgrounds Ketamine possesses analgesia, anti-inflammation, anticonvulsant, and neuroprotection properties. However, the evidence that supports its use in mechanically ventilated critically ill patients with COVID-19 is insufficient. The study's goal was to assess ketamine's effectiveness and safety in critically ill, mechanically ventilated (MV) patients with COVID-19. Methods Adult critically ill patients with COVID-19 were included in a multicenter retrospective-prospective cohort study. Patients admitted between March 1, 2020, and July 31, 2021, to five ICUs in Saudi Arabia were included. Eligible patients who required MV within 24 hours of ICU admission were divided into two sub-cohort groups based on their use of ketamine (Control vs. Ketamine). The primary outcome was the length of stay (LOS) in the hospital. P/F ratio differences, lactic acid normalization, MV duration, and mortality were considered secondary outcomes. Propensity score (PS) matching was used (1:2 ratio) based on the selected criteria. Results In total, 1,130 patients met the eligibility criteria. Among these, 1036 patients (91.7 %) were in the control group, whereas 94 patients (8.3 %) received ketamine. The total number of patients after PS matching, was 264 patients, including 88 patients (33.3 %) who received ketamine. The ketamine group's LOS was significantly lower (beta coefficient (95 % CI): −0.26 (−0.45, −0.07), P = 0.008). Furthermore, the PaO2/FiO2 ratio significantly improved 24 hours after the start of ketamine treatment compared to the pre-treatment period (6 hours) (124.9 (92.1, 184.5) vs. 106 (73.1, 129.3; P = 0.002). Additionally, the ketamine group had a substantially shorter mean time for lactic acid normalization (beta coefficient (95 % CI): −1.55 (−2.42, −0.69), P 0.01). However, there were no significant differences in the duration of MV or mortality. Conclusions Ketamine-based sedation was associated with lower hospital LOS and faster lactic acid normalization but no mortality benefits in critically ill patients with COVID-19. Thus, larger prospective studies are recommended to assess the safety and effectiveness of ketamine as a sedative in critically ill adult patients.
... Severe SARS-CoV-2 infection is characterized primarily by elevated levels of IL-6, but also IL-8, IL-10, TNF-a and interferon-g (IFN-g). In particular, IL-6 has been associated with a significantly higher mortality and may serve as an indicator for disease prognosis and the severity of in COVID-19 (8). ...
Article
Full-text available
Background and aims Several factors, such as hypertension and diabetes mellitus, are known to influence the course of coronavirus disease 2019 (COVID-19). However, there is currently little information on genetic markers that influence the severity of COVID-19. In this study, we specifically investigated the single nucleotide polymorphism (SNP) rs4986790 in the TLR4 gene to identify a universal marker for preclinical prediction of COVID-19 disease progression. Methods We analyzed the influence of demographics, pre-existing conditions, inflammatory parameters at the time of hospitalization, and TLR4 rs4986790 genotype on the outcome of COVID-19 in a comprehensive cohort (N = 1570). We performed multivariable analysis to investigate the impact of each factor. Results We confirmed that younger patient age and absence of pre-existing conditions were protective factors against disease progression. Furthermore, when comparing patients with mild SARS-CoV-2 infection with patients who required hospitalization or intensive care or even died due to COVID-19, the AG/GG genotype of TLR4 rs4986790 was found to be a protective factor against COVID-19 disease progression (OR: 0.51, 95% CI: 0.34 - 0.77, p = 0.001). In addition, we demonstrated that low levels of interleukin-6 (IL-6) and procalcitonin (PCT) had a favorable effect on COVID-19 disease severity. In the subsequent multivariable analysis, we confirmed the absence of cardiovascular disease, low levels of IL-6 and PCT, and TLR4 rs4986790 AG/GG genotypes as independent predictors of potential hospitalization and reduction of severe or fatal disease course. Conclusion In this study, we identified an additional genetic factor that may serve as an invariant predictor of COVID-19 outcome. The TLR4 rs4986790 AG/GG genotype reduced by half the risk of COVID-19 patients requiring hospitalization, intensive care or to have a fatal outcome. In addition, we were able to confirm the influence of previously known factors such as pre-existing conditions and inflammatory markers upon the onset of disease on the course of COVID-19. Based on these observations, we hereby provide another prognostic biomarker that could be used in routine diagnostics as a predictive factor for the severity of COVID-19 prior to SARS-CoV-2 infection.
... Furthermore, IL-17 facilitates the migration of neutrophils to the lungs, resulting in tissue damage during inflammation. This influx of neutrophils leads to the formation of Neutrophil Extracellular Traps (NETs) in the lungs, contributing to organ damage and the development of COVID-19-related ARDS [22,42,49]. ...
Article
The COVID-19 pandemic has underscored the critical importance of understanding the intricate relationship between micronutrient levels and disease outcomes. This study explores the impact of Vitamin D, calcium, phosphorus, magnesium, and alkaline phosphatase (ALP) on COVID-19 severity and mortality. The study involves 200 participants (100 COVID-19 patients, 100 controls), we meticulously analyzed micronutrient dynamics. Calcium, phosphorus, magnesium and ALP was measured spectrophotometrically. Vitamin D was measured using Chemiluminescent method. The study reveals that diminished levels of calcium, phosphorus, magnesium, and with elevated ALP, are significantly associated with COVID-19 cases. Whereas the Vitamin D levels in severe group was increased when compared to mild cases but decreased than control group. Disease severity correlated with declining calcium (r = − 0.35, p < 0.01), phosphorus (r = − 0.26, p < 0.05), and magnesium (r = − 0.21, p < 0.05), and increased ALP (r = 0.42, p < 0.001). Post-discharge, calcium (p < 0.05) and phosphorus (p < 0.01) showed positive trends, while ALP (p < 0.001) decreased. Notably, calcium (OR = 0.63, p < 0.05) and ALP (OR = 1.87, p < 0.001) emerged as significant predictors of disease severity. The findings not only illuminate potential therapeutic avenues but also emphasize the need to optimize nutrient levels, including magnesium, for COVID-19 prevention and management. Given the complexities of these relationships, further rigorous exploration, including well-designed trials and understanding underlying mechanisms, is imperative to unravel the dynamics of these nutrient interactions in the context of COVID-19.
Article
Full-text available
Mortality due to Covid-19 is highly associated with advanced age, owing in large part to severe lower respiratory tract infection. SARS-CoV-2 utilizes the host ACE2 receptor for infection. Whether ACE2 abundance in the lung contributes to age-associated vulnerability is currently unknown. We set out to characterize the RNA and protein expression profiles of ACE2 in aging human lung in the context of phenotypic parameters likely to affect lung physiology. Examining publicly available RNA sequencing data, we discovered that mechanical ventilation is a critical variable affecting lung ACE2 levels. Therefore, we investigated ACE2 protein abundance in patients either requiring mechanical ventilation or spontaneously breathing. ACE2 distribution and expression were determined in archival lung samples by immunohistochemistry (IHC). Tissues were selected from the specimen inventory at a large teaching hospital collected between 2010–2020. Twelve samples were chosen from patients receiving mechanical ventilation for acute hypoxic respiratory failure (AHRF). Twenty samples were selected from patients not requiring ventilation. We compared samples across age, ranging from 40–83 years old in the ventilated cohort and 14–80 years old in the non-ventilated cohort. Within the alveolated parenchyma, ACE2 expression is predominantly observed in type II pneumocytes (or alveolar type II / AT2 cells) and alveolar macrophages. All 12 samples from our ventilated cohort showed histologic features of diffuse alveolar damage including reactive, proliferating AT2 cells. In these cases, ACE2 was strongly upregulated with age when normalized to lung area (p = 0.004) or cellularity (p = 0.003), associated with prominent expression in AT2 cells. In non-ventilated individuals, AT2 cell reactive changes were not observed and ACE2 expression did not change with age when normalized to lung area (p = 0.231) or cellularity (p = 0.349). In summary, ACE2 expression increases with age in the setting of alveolar damage observed in patients on mechanical ventilation, providing a potential mechanism for higher Covid-19 mortality in the elderly.
Article
Full-text available
The COVID-19 pandemic has caused a global health crisis, with no specific antiviral to treat the infection and the absence of a suitable vaccine to prevent it. While some individuals contracting the SARS-CoV-2 infection exhibit a well coordinated immune response and recover, others display a dysfunctional immune response leading to serious complications including ARDS, sepsis, MOF; associated with morbidity and mortality. Studies revealed that in patients with a dysfunctional immune response, there is a massive cytokine and chemokine release, referred to as the ‘cytokine storm’. As a result, such patients exhibit higher levels of pro-inflammatory/modulatory cytokines and chemokines like TNFα, INFγ, IL-1β, IL-2, IL-4, IL-6, IL-7, IL-9, IL-10, IL-12, IL-13, IL-17, G-CSF, GM-CSF, MCSF, HGF and chemokines CXCL8, MCP1, IP10, MIP1α and MIP1β. Targeting this cytokine storm is a novel, promising treatment strategy to alleviate this excess influx of cytokines observed at the site of infection and their subsequent disastrous consequences. Natural immunosuppressant compounds, derived from plant sources like curcumin, luteolin, piperine, resveratrol are known to inhibit the production and release of pro-inflammatory cytokines and chemokines. This inhibitory effect is mediated by altering signal pathways like NF-κB, JAK/STAT, MAPK/ERK that are involved in the production and release of cytokines and chemokines. The use of these natural immunosuppressants as adjuvants to ameliorate the cytokine storm; in combination with antiviral agents and other treatment drugs currently in use presents a novel, synergistic approach for the treatment and effective cure of COVID-19. This review briefly describes the immunopathogenesis of the cytokine storm observed in SARS-CoV-2 infection and details some natural immunosuppressants that can be used as adjuvants in treating COVID-19 disease.
Article
Full-text available
Remdesivir is the only FDA-approved drug for the treatment of COVID-19 patients. The active form of remdesivir acts as a nucleoside analog and inhibits the RNA-dependent RNA polymerase (RdRp) of coronaviruses including SARS-CoV-2. Remdesivir is incorporated by the RdRp into the growing RNA product and allows for addition of three more nucleotides before RNA synthesis stalls. Here we use synthetic RNA chemistry, biochemistry and cryo-electron microscopy to establish the molecular mechanism of remdesivir-induced RdRp stalling. We show that addition of the fourth nucleotide following remdesivir incorporation into the RNA product is impaired by a barrier to further RNA translocation. This translocation barrier causes retention of the RNA 3ʹ-nucleotide in the substrate-binding site of the RdRp and interferes with entry of the next nucleoside triphosphate, thereby stalling RdRp. In the structure of the remdesivir-stalled state, the 3ʹ-nucleotide of the RNA product is matched and located with the template base in the active center, and this may impair proofreading by the viral 3ʹ-exonuclease. These mechanistic insights should facilitate the quest for improved antivirals that target coronavirus replication.
Article
Full-text available
Introduction Coronavirus disease 2019 (COVID-19) appeared in China in December 2019 and has spread around the world. High Interleukin-6 (IL-6) levels in COVID-19 patients suggest that a cytokine storm may play a major role in the pathophysiology and are considered as a relevant parameter in predicting most severe course of disease. The aim of this study was to assess repeated IL-6 levels in critically ill COVID-19 patients admitted to our Intensive Care Unit (ICU) and to evaluate their relationship with patient’s severity and outcome. Methods We conducted a retrospective study on patients admitted to the ICU with a diagnosis of COVID-19 between March 10 (i.e. the date of the first admitted patients) and April 30, 2020. Demographic, clinical and laboratory data were collected at admission. On the day of IL-6 blood concentration measurement, we also collected results of D-Dimers, C-Reactive Protein, white blood cells and lymphocytes count, lactate dehydrogenase (LDH) and ferritin as well as microbiological samples, whenever present. Results Of a total of 65 patients with COVID-19 admitted to our ICU we included 41 patients with repeated measure of IL-6. There was a significant difference in IL-6 levels between survivors and non-survivors over time (p = 0.001); moreover, non survivors had a significantly higher IL-6 maximal value when compared to survivors (720 [349–2116] vs. 336 [195–646] pg/mL, p = 0.01). The IL-6 maximal value had a significant predictive value of ICU mortality (AUROC 0.73 [95% CI 0.57–0.89]; p = 0.01). Conclusions Repeated measurements of IL-6 can help clinicians in identifying critically ill COVID-19 patients with the highest risk of poor prognosis.
Article
Full-text available
Background and aims Identification of SARS-CoV-2-infected patients at high-risk of poor prognosis is crucial. We aimed to establish predictive models for COVID-19 pneumonia severity in hospitalized patients. Methods Retrospective study of 430 patients admitted in Vall d’Hebron Hospital (Barcelona) between 03-12-2020 and 04-28-2020 due to COVID-19 pneumonia. Two models to identify the patients who required high-flow-oxygen-support were generated, one using baseline data and another with also follow-up analytical results. Calibration was performed by a 1000-bootstrap replication model. Results 249 were male, mean age 57.9 years. Overall, 135 (31.4%) required high-flow-oxygen-support . The baseline predictive model showed a ROC of 0.800 based on: SpO2/FiO2 (adjusted Hazard Ratio-aHR = 8), chest x-ray (aHR = 4), prior immunosuppressive therapy (aHR = 4), obesity (aHR = 2), IL-6 (aHR = 2), platelets (aHR = 0.5). The cut-off of 11 presented a specificity of 94.8%. The second model included changes on the analytical parameters: ferritin (aHR = 7.5 if ≥200ng/mL) and IL-6 (aHR = 18 if ≥64pg/mL) plus chest x-ray (aHR = 2) showing a ROC of 0.877. The cut-off of 12 exhibited a negative predictive value of 92%. Conclusions SpO2/FiO2 and chest x-ray on admission or changes on inflammatory parameters as IL-6 and ferritin allow us early identification of COVID-19 patients at risk of high-flow-oxygen-support that may benefit from a more intensive disease management.
Article
Full-text available
The management of acute hypoxemic respiratory failure and the effect of antiviral drugs in patients with severe COVID-19 have been debated. This case presents the management of a 64-year-old man COVID-19 patient admitted to the Intensive Care Unit with fever, fatigue, shortness of breath and hemophagocytic lymphohistiocytosis syndrome. Helmet mask was successfully used to treat his hypoxemic respiratory failure without any aerosol problems. Tocilizumab, an antagonist interleukin-6, was intravenously infused as an alternative drug. After administration, the high level of IL-6, CRP, ferritin, D-dimer, triglyceride, and H-scores decreased, and the patient observed good clinical and laboratory improvements. In this case report, we describe the effect of noninvasive ventilation delivered by helmet mask and antiviral drugs, and the intravenous administration of tocilizumab in a patient with hemophagocytic lymphohistiocytosis syndrome and COVID-19.
Article
Full-text available
Background: Currently, SARS-CoV-2 liver invasion, inflammatory cytokines, and antiviral drugs are widely thought to be associated with liver dysfunction in COVID-19 patients. Besides, previous studies indicated that ACEI/ARB drugs can increase the expression of hepatic ACE2, a cell entry receptor for SARS-CoV-2. This study aims to investigate whether ACEI/ARB aggravates liver injury and the association of inflamma-tory cytokines and antiviral drugs with liver dysfunction in patients with hypertension and COVID-19. Method: This retrospective study included 127 hypertensive patients with long-term use or nonuse of ACEI/ARBs hospitalized for COVID-19 from January 30 to April 7, 2020, in Tongji hospital of Wuhan, China. Demographic, clinical, laboratory, treatment, and outcome data were collected. Results: Of the 127 patients with COVID-19 and hypertension, 43 taking long-term of ACEI/ARBs and 84 without using ACEI/ARBs. Abnormal liver function was observed in part of ACEI/ARB and non-ACEI/ARB users but without significant differences between these two groups. Serum inflammatory cytokines, IL-6, IL-8, and TNFα, as well as inflammation-related markers, ferritin, procalcitonin, and C-reactive protein, were significantly elevated in patients with liver dysfunction. IL-6 level was positively correlated with liver function tests on admission and highly consistent with the changes of abnormal ALT, AST, and GGT during hospitalization, but the correlations of other inflammatory cytokines were low. There was no significant association between the use of antiviral drugs and liver dysfunction in these patients. Conclusion: The elevation of inflammatory cytokine, IL-6, but not ACEI/ARB and antiviral drugs, is closely associated with liver dysfunction in patients with hypertension and COVID-19.
Preprint
Full-text available
Background:The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic has affected almost every country. Interleukin-6 (IL-6), a cytokine secreted by CD4+ T cell, has been shown to be a reliable marker of disease severity and a useful parameter for monitoring progression of coronavirus disease-2019 (COVID-19). However its value as a predictor of severe disease has not been assessed. Methods:A total of 160 laboratory-confirmed COVID-19 patients admitted to two hospitals were enrolledand separated into two groups according to whether or not they progressed to develop severe illness. Demographic and clinical characteristics at admission were compared between the groups. Results: Patients who developed severe COVID-19 had significantly higher baseline IL-6 levels than patients who had mild disease course in hospital (P< 0.001). Patients were further grouped according to quartiles of IL-6 level. The cumulative incidence of severe illnesswas significantly higher in the third and fourth quartiles groups than in the first quartile group (55% vs. 15% and 80% vs. 15%, respectively;bothP< 0.001). In multivariate logistic regression analysis, the risk for developing severe disease was markedly higher in the highest IL-6 quartile than in the lowest quartile (odds ratio: 14.95; 95% confidence interval: 3.65–61.30; P< 0.001). Receiver operating characteristic curve analysis of potential predictive variables showed the area under the curve to be largest for baseline IL-6, with the value of 5.20 pg/mL having the best balance of sensitivity and specificity for predicting risk of severe COVID-19. Conclusion: Serum baseline IL-6 appears to be a reliable predictor of risk of severe COVID-19. Early intervention may be advisable in patients with serum IL-6 levels >5.20 pg/mL, even if initial symptoms are mild.
Article
Full-text available
Purpose of review: The rapid spread of virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has turned out to be a global emergency. Symptoms of this viral infection, coronavirus disease 2019 (COVID-19), include mild infections of the upper respiratory tract, viral pneumonia, respiratory failure, multiple organ failure and death. Till date, no drugs have been discovered to treat COVID-19 patients, and therefore, a considerable amount of interest has been shown in repurposing the existing drugs. Recent findings: Out of these drugs, chloroquine (CQ) and hydroxychloroquine (HCQ) have demonstrated positive results indicating a potential antiviral role against SARS-CoV-2. Its mechanism of action (MOA) includes the interference in the endocytic pathway, blockade of sialic acid receptors, restriction of pH mediated spike (S) protein cleavage at the angiotensin-converting enzyme 2 (ACE2) binding site and prevention of cytokine storm. Unfortunately, its adverse effects like gastrointestinal complications, retinopathy and QT interval prolongation are evident in treated COVID-19 patients. Yet, multiple clinical trials have been employed in several countries to evaluate its ability in turning into a needed drug in this pandemic. Summary: This review attempts to summarize the MOA of CQ/HCQ and its side effects. The existing literature hints that till date, the role of CQ/HCQ in COVID-19 may be sceptical, and further studies are warranted for obtaining a therapeutic option that could be effectively used across the world to rise out from this pandemic.
Article
Introduction: Interleukin-6 (IL-6) is an inflammatory cytokine marker produced in response to infection and tissue injury. As the COVID-19 pandemic continues to thrive, uncertainties remain regarding COVID-19-related cardiovascular outcomes, specifically major adverse cardiovascular events (MACE). High IL-6 levels correlate with disease severity in COVID-19 patients; however, studies have not shown an association between elevated IL-6 levels and MACE and/or mortality. Methods: Two hundred twenty-five patients tested positive for COVID-19 at a major Southwest quaternary United States hospital between March 2020 and May 2020. Chart review of these patients was used to identify cases of MACE/ mortality and their respective IL-6 levels. This data was extracted for clinical analysis to identify any correlation between IL-6 levels and incidence of MACE and/or death. MACE was defined as a composite of myocardial infarction, stroke, deep venous thrombosis/ pulmonary embolism, or shock requiring vasopressor support. Results: Of the 225 patients with COVID-19, 112 patients had an IL-6 level drawn. Patients with elevated IL-6 levels had a higher incidence of MACE compared to patients with normal IL-6 levels (81.8% vs. 48.5%, p=0.0004, OR=4.77, 95% CI=1.81-13.50). There was a significant difference between the percentage of patients with elevated IL-6 levels who expired and patients with elevated IL-6 levels who did not expire (90.4% vs. 53.9%, p=0.0023, OR=8.11, 95% CI=1.76-74.86). IL-6 levels were significantly higher in patients who developed MACE and expired compared to levels in patients that did not (81.6% vs. 46%, p=0.0001, OR=5.21, 95% CI=2.03-14.13). Conclusions: MACE and/or death in COVID-19 patients were associated with elevated IL-6 levels. It is advisable to obtain IL-6 levels in all COVID-19 infected patients to help risk stratify these patients and treat IL-6 positive patients more aggressively.