ArticlePDF AvailableLiterature Review

Advances in the development of therapeutic strategies against COVID-19 and perspectives in the drug design for emerging SARS-CoV-2 variants

Authors:

Abstract

Since Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was identified in late 2019, the coronavirus disease 19 (COVID-19) pandemic has challenged public health around the world. Currently, there is an urgent need to explore antiviral therapeutic targets and effective clinical drugs. In this study, we systematically summarized two main therapeutic strategies against COVID-19, namely drugs targeting the SARS-CoV-2 life cycle and SARS-CoV-2-induced inflammation in host cells. The development of above two strategies is implemented by repurposing drugs and exploring potential targets. A comprehensive summary of promising drugs, especially cytokine inhibitors and Chinese traditional medicine (CTM), provides recommendations for clinicians as evidence-based medicine in the actual clinical COVID-19 treatment. Considering the emerging SARS-CoV-2 variants greatly impact the effectiveness of drugs and vaccines, we reviewed the apperance and details of SARS-CoV-2 variants for further perspectives in drug design, which brings update clues to develop therapeutical agents against the variants. Based on this, the development of broadly antiviral drugs, combined with immunomodulatory or holistic therapy in the host, is prior to be considered for therapeutic interventions on mutant strains of SARS-CoV-2. Therefore, it is highly acclaimed the requirements of the concerted efforts from multi-disciplinary basic studies and clinical trials, which improves the accurate treatment of COVID-19 and optimizes the contingency measures to emerging SARS-CoV- 2 variants.
Review
Advances in the development of therapeutic strategies against COVID-19
and perspectives in the drug design for emerging SARS-CoV-2 variants
Jialing Yin
a,1
, Chengcheng Li
a,1
, Chunhong Ye
a
, Zhihui Ruan
a,b
, Yicong Liang
a
, Yongkui Li
a
, Jianguo Wu
a,b,
,
Zhen Luo
a,b,
a
Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, PR China
b
Foshan Institute of Medical Microbiology, Foshan 528315, PR China
article info
Article history:
Received 10 November 2021
Received in revised form 18 January 2022
Accepted 27 January 2022
Available online 31 January 2022
Keywords:
SARS-CoV-2
COVID-19 pandemic
Therapeutic strategies
Drug target
SARS-CoV-2 variants
abstract
Since Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was identified in late 2019, the
coronavirus disease 2019 (COVID-19) pandemic has challenged public health around the world.
Currently, there is an urgent need to explore antiviral therapeutic targets and effective clinical drugs.
In this study, we systematically summarized two main therapeutic strategies against COVID-19, namely
drugs targeting the SARS-CoV-2 life cycle and SARS-CoV-2-induced inflammation in host cells. The devel-
opment of above two strategies is implemented by repurposing drugs and exploring potential targets. A
comprehensive summary of promising drugs, especially cytokine inhibitors, and traditional Chinese med-
icine (TCM), provides recommendations for clinicians as evidence-based medicine in the actual clinical
COVID-19 treatment. Considering the emerging SARS-CoV-2 variants greatly impact the effectiveness
of drugs and vaccines, we reviewed the appearance and details of SARS-CoV-2 variants for further per-
spectives in drug design, which brings updating clues to develop therapeutical agents against the vari-
ants. Based on this, the development of broadly antiviral drugs, combined with immunomodulatory, or
holistic therapy in the host, is prior to being considered for therapeutic interventions on mutant strains
of SARS-CoV-2. Therefore, it is highly acclaimed the requirements of the concerted efforts from multi-
disciplinary basic studies and clinical trials, which improves the accurate treatment of COVID-19 and
optimizes the contingency measures to emerging SARS-CoV-2 variants.
Ó2022 The Author(s). Published by Elsevier B.V. on behalf of Research Network of Computational and
Structural Biotechnology. This is an open access article under the CC BY-NC-ND license (http://creative-
commons.org/licenses/by-nc-nd/4.0/).
Contents
1. Introduction . . . ...................................................................................................... 825
2. The structure of SARS-CoV-2 . . . . . . . . . ................................................................................... 825
3. Life cycle of SARS-CoV-2 . . . . . . . . . . . . ................................................................................... 825
4. Drugs targeting the SARS-CoV-2 life cycle . . . . . . . . . . . . . . . . . ................................................................ 827
4.1. Anti-viral entry . . . . . . . . ....... .................................................................................. 827
4.1.1. Inhibition of key enzymes in the binding process . . . ........................................................... 827
4.1.2. Competitive binding to ACE2 and SARS-CoV-2 S protein. . . . . . . . . . . . . . . . . ........................................ 828
4.2. Viral protease inhibitors . ....... .................................................................................. 828
https://doi.org/10.1016/j.csbj.2022.01.026
2001-0370/Ó2022 The Author(s). Published by Elsevier B.V. on behalf of Research Network of Computational and Structural Biotechnology.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Abbreviations: SARS-CoV-2, Severe Acute Respiratory Syndrome Coronavirus 2; COVID-19, coronavirus disease 2019; ACE2, Angiotensin-converting enzyme 2; RBD,
receptor-binding domain; CTD, C-terminal domain; NTD, N-terminal domain; RdRp, RNA dependent RNA polymerase; ERGIC, endoplasmic reticulum-Golgi intermediate
compartment; ARDS, acute respiratory distress syndrome; CRS, cytokine release syndrome; MODS, multiple organ dysfunction syndrome; mAb, monoclonal antibody; Nbs,
nanobodies; JAK, Janus kinase; STAT, Signal Transducer and Activator of Transcription; TCZ, Tocilizumab;
a
1AT, alpha-1 antitrypsin; CEP, Cepharanthine; TCM, traditional
Chinese medicine; EMA, European Medicines Agency; FDA, U.S. Food and Drug Administration; NMPA, National Medical Products Administration; VOI, variants of interest;
VOC, variants of concern; VUM, variants under monitoring.
Corresponding authors at: Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, PR China.
E-mail addresses: jwu898@jnu.edu.cn (J. Wu), zhluo18@jnu.edu.cn (Z. Luo).
1
Jialing Yin and Chengcheng Li contributed equally to the work.
Computational and Structural Biotechnology Journal 20 (2022) 824–837
journal homepage: www.elsevier.com/locate/csbj
4.3. Anti-viral RNA polymerase . . . . . . . . . . . . . . . ......................................................... ................ 829
4.4. Anti-viral release . . . . . . . ....... .................................................................................. 829
5. Drugs targeting SARS-CoV-2-induced inflammation in host cells . . . . . . . . . . . . . . . . . . ............................................. 829
5.1. Cytokine inhibitors . . . . . ....... .................................................................................. 829
5.2. Signaling pathway inhibitors . . . . . . . . . . . . . ......................................................................... 830
5.3. Steroids treatment. . . . . . ......................................................................................... 830
5.4. Holistic therapy and traditional Chinese medicine treatment . . . . . . .......... ............................................ 831
6. Emerging SARS-CoV-2 variants . . . . . . . ................................................................................... 832
6.1. Mutations in structural proteins . . . . . . . . . . ................................ ......................................... 832
6.2. Mutations in non-structural proteins. . . . . . . .... ..................................................................... 832
7. Perspectives in the drug design for SARS-CoV-2 variants . . . . . ................................................................ 833
8. Discussion. . . . . ...................................................................................................... 833
CRediT authorship contribution statement . . . . . . . . . . . . . . . . . . ................................................................ 833
Declaration of Competing Interest . . . . ................................................................................... 833
Acknowledgements . . . . . . . . . . . . . . . . ................................................................................... 833
References . . . . ...................................................................................................... 833
1. Introduction
The new coronavirus, namely 2019-nCoV, was first identified on
December 31, 2019. Due to being highly homologous to Severe
Acute Respiratory Syndrome Coronavirus (SARS-CoV), 2019-nCoV
was listed as Severe Acute Respiratory Syndrome Coronavirus 2
(SARS-CoV-2) by the International Commission for Classification
of Viruses (ICTV) on February 11, 2020 [1]. Among seven coron-
aviruses infecting humans, three coronaviruses can cause serious
diseases, including severe acute respiratory syndrome coronavirus
(SARS-CoV) emerging in 2003, Middle East respiratory syndrome
coronavirus (MERS-CoV) in 2012, and Severe Acute Respiratory
Syndrome Coronavirus 2 (SARS-CoV-2) in 2019 [2].
The outbreak of SARS-CoV-2 causes an acute respiratory disease
called coronavirus disease 2019 (COVID-19) and leads to a severe
epidemic worldwide. Typical symptoms of COVID-19 include fever,
sore throat, fatigue, cough, and difficulty breathing [3,4]. Some of
the clinical symptoms are similar to those of respiratory infections
and cannot be accurately diagnosed. The approach to quickly diag-
nose and develop related drugs and vaccines has become a top pri-
ority. At present, the WHO has approved the use of a variety of
vaccines [5]. As of January 9, 2022, the cumulative number of
reported cases worldwide has exceeded 304 million, and the
cumulative death toll has been more than 5.4 million [6]. As of Jan-
uary 13, 2022, a total of 9.2 billion doses of vaccine have been
administrated globally (Fig. 1).
Till now, there have been candidate drugs in COVID-19 treat-
ment at different stages and levels available for selection and fur-
ther research and development (Table 1). These small-molecule
drugs can exert therapeutic effects against COVID-19 by prevent-
ing SARS-CoV-2 from entering cells, inhibiting viral proteases or
RNA-dependent RNA polymerase activities, reducing virus-
induced inflammation, and balancing immunomodulatory effect
in host [7,8]. Fortunately, some antiviral drugs have been used
in clinical practice and play certain roles in the treatment of
COVID-19 patients, such as Remdesivir and Molnupinavir [9–11].
Currently, hundreds of clinical trials of COVID-19 drugs are under-
way to obtain satisfactory clinical results [12]. A large number of
promising clinical trials will determine which drugs or vaccines
are suitable for the treatment or prevention of COVID-19 (Table 2).
The effective medicine is expected to be approved for COVID-19
treatment, however, due to the rapid mutation of SARS-CoV-2 gen-
ome, the present drugs or vaccines may lose their control on SARS-
CoV-2 spread with the increased infectivity [13,14]. The emerging
SARS-CoV-2 variants bring severe challenges to COVID-19 surveil-
lance and control. In this review, we systematically summarized
the advances in the therapeutic strategies against COVID-19 tar-
geting the structure of SARS-CoV-2 in viral life cycle and immune
response in virus-host interaction. More importantly, we presented
the details and clues of SARS-CoV-2 variants for further perspec-
tives in drug design, which inspires follow-up researchers to
develop therapeutic agents against pandemic COVID-19 and
emerging SARS-CoV-2 variants.
2. The structure of SARS-CoV-2
SARS-CoV-2 is a kind of enveloped virus at a diameter ranging
from 80 to 220 nm with a positive single-stranded RNA inside its
shell, belonging to the b-CoV class of human coronaviruses [2].
The entire SARS-CoV-2 particle is mainly composed of 4 structural
proteins, a fragile lipid envelop, and genomic RNA. The four struc-
tural proteins are the membrane (M), nucleocapsid (N), envelop
(E), and spike (S) protein (Fig. 2A).
The M protein plays a central role in virus assembly. Its pres-
ence enables viruses and host factors to gather on the cell mem-
brane to form progeny virus particles [15]. The complex formed
by N protein and genomic RNA plays an important role in viral
transcription and assembly. The N protein is divided into N-
terminal domain, C-terminal domain, and disordered central
domain (NTD, CTD, and RNA binding domain) [16]. The E protein
is a small and complete membrane protein, and its functions run
through the life cycle of SARS-CoV-2, including assembly and
pathogenicity [17]. The S protein is the key to the invasion of cells
by SARS-CoV-2 and it exists on the surface of the virus membrane
in the form of trimers [18]. It is composed of S1 and S2 subunits
and the latter one is the most conserved region in spike protein
[19]. The E protein and the M protein are alternately arranged on
the surface of the virus membrane, together with the S protein
forming the virus shell, and the N protein interacts with the viral
RNA to form the core of the virus particle [2]. During viral replica-
tion, a polyprotein 1ab is translated through ORF1ab in viral gen-
ome, and subsequently cleaved into 16 non-structural proteins
by protease (Fig. 2B).
The proteins of SARS-CoV-2 are highly glycosylated [20]. For
example, about 40% surface of S protein is covered by glycans
[21], which helps the virus hide epitopes and avoid antibody recog-
nition [22]. SARS-CoV-2 S protein comprises 22 N-linked glycosyla-
tion sequons per protomer [23]. Because glycosylation is the key to
virus invasion, the glycosylation of S protein needs to be taken into
consideration when designing vaccines and antibodies [24].
3. Life cycle of SARS-CoV-2
SARS-CoV-2 can bind to Angiotensin-converting enzyme 2
(ACE2), a functional receptor of SARS-CoV [25], on the cell surface
with its S protein to enter the cell through membrane fusion and
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
825
endocytosis [19] (Fig. 3A). Proteins from host cells, such as the
serine protease TMPRSS2 [26] and high-density lipoprotein
(HDL) scavenger receptor type B (SR-B1) [27], facilitate the
SARS-CoV-2 invasion processes (Fig. 3B). Transmembrane serine
protease 4 (TMPRSS4) is found to be most significantly related
to ACE2 [28]. SARS-CoV-2 has its unique Furin cleavage site never
found before in other coronaviruses, is required for the virus to
enter cells lacking cathepsin protease [29]. Furin can cleave the
SARS-CoV-2 spike protein at the S1/S2 site [2], resulting in active
S1 and S2 subunits. The cutting process occurs during the virus
packaging process [30].
Once entering the host cell, the virus particle releases the viral
genome, and utilizes host ribosome to translate the viral polypro-
teins pp1a and pp1ab [31]. Then the viral protease 3CLpro [32] and
PLpro [33] cleave the polyprotein into a variety of active proteins
(Fig. 3C). Viral replication is dominated by a replication/transcrip-
tion complex composed of non-structural proteins [34]. For exam-
ple, nsp7, nsp8, and nsp12 together form the virus-independent
RNA polymerase structure (RdRp) for viral genome replication
[35] (Fig. 3D). The translation of nsp14 can shut down the host pro-
tein synthesis and inhibit the innate immune response, while the
combination of nsp10 enhances this effect [36].
After four structural protein is produced by transcription, the N
protein binds to the genome, and the remaining three (S, E, and M)
are integrated into the endoplasm and then sent to the endoplas-
mic reticulum-Golgi intermediate compartment (ERGIC) for fur-
ther process, such as furin-mediated cleavage. Subsequently, the
assembled progeny virus was released by exocytosis or budding
[37,38] for the next round of invasion to host cell (Fig. 3E). In
ERGIC, E protein can form pores to cause Ca
2+
to leak and activate
NLRP3 inflammasome to achieve pro-inflammatory effects [39]
(Fig. 3F).
Fig. 1. Update of COVID-19 pandemic worldwide. As of January 9, 2022, the cumulative number of reported cases worldwide has exceeded 304 million, while the
cumulative death toll has exceeded 5.4 million. Till to January 13, 2022, 9.2 billion doses of vaccine have been administered globally.
Table 1
Summary of approved and developed drugs against COVID-19.
Approved drugs
Product Developer Therapeutic
class/drug type
Status Approving
Authority
Kineret (Anakinra) Sobi Immunomodulator Marketing authorization granted: 17/12/2021 EMA
Regkirona (Regdanvimab) Celltrion Healthcare Monoclonal antibody Marketing authorization granted: 12/11/2021 EMA
RoActemra Roche Immunomodulator Marketing authorization for COVID-19 indication
granted: 07/12/2021
EMA
Ronapreve (Casirivimab/
Imdevimab)
Roche & Regeneron Monoclonal antibody Marketing authorization granted: 12/11/2021 EMA
Veklury (Remdesivir) Gilead Sciences Nucleotide analogs Conditional marketing authorization granted: 03/07/
2020
EMA
Marketing authorization granted: 22/10/2020 FDA
Xevudy (Sotrovimab) GlaxoSmithKline & Vir
Biotechnology
Monoclonal antibody Marketing authorization granted: 17/12/2021 EMA
Developed drugs
Product Developer Therapeutic
class/drug type
Status Data source
Sarilumab Sanofi Aventis Immunomodulator Clinical phase EMA
Canakinumab Novartis Monoclonal antibody Clinical phase Clinical trials
Anakinra Swedish Orphan Biovitrum
AB (SOBI)
Immunomodulator Clinical phase EMA
Tocilizumab Roche Immunomodulator Clinical phase EMA
Pidotimod / Immunomodulator / /
Cepharanthine / Immunomodulator / /
The information is summarized from the available data online at sites https://www.fda.gov/drugs/emergency-preparedness-drugs/coronavirus-covid-19-drugs,https://www.
ema.europa.eu/en/human-regulatory/overview/public-health-threats/coronavirus-disease-covid-19/treatments-vaccines/treatments-covid-19/covid-19-treatments-
research-development, and https://clinicaltrials.gov/ct2/results?cond=COVID-19&term = anakinra&cntry=&state=&city=&dist=. EMA: European Medicines Agency; FDA: the
U.S. Food and Drug Administration.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
826
4. Drugs targeting the SARS-CoV-2 life cycle
The process of SARS-CoV-2 entering the human body and self-
replicating to release progeny virus can be divided into different
stages. According to the characteristics of each dynamic stage, dif-
ferent drug targets to these specialized processes are formulated to
block the life activities of the virus (Fig. 3).
4.1. Anti-viral entry
4.1.1. Inhibition of key enzymes in the binding process
Alpha-1 antitrypsin (
a
1AT) inhibits the protease activity of
TMPRSS2 at physiological concentrations to restrain the entry
and replication of SARS-CoV-2 in cell lines and primary cells [40].
A statistical study based on 500,000 people shows that mild
a
1-
Antitrypsin deficiency (AATD) genotypes were not associated with
increased SARS-CoV-2 infection rates or fatalities [41]. For the lack
of
a
1AT makes it easier to activate TMPRSS2, severe AATD patients
may be susceptible to SARS-CoV-2 [42,43]. There have been four
trials recruited or completed to test whether
a
1AT can be used
for the treatment of COVID-19 patients [44].
Halofuginone can reduce endogenous TMPRSS2 expression at
sub-micromolar concentrations, which showed significant resis-
tance to SARS-CoV-2 infection in vitro in both live- and pseudo-
virus models [45]. Halofuginone is an oral anti-fibrosis [46] and
anti-inflammatory [47] drug. A study in mice reveals that the dis-
tribution of halofuginone can be detected in various organs, includ-
ing kidneys and lungs [48]. ACE2 is widely expressed in these
organs [49], making these organs vulnerable to the SARS-CoV-2.
Therefore, the antiviral activity and wide distribution of halofugi-
none in vivo repurpose it as a promising anti-SARS-CoV-2 drug.
Platycodon grandiflorum D (PD), a natural component of Platy-
codon grandiflorum [50], and pan-coronavirus fusion inhibitor EK1
derived lipopeptide EK1C4 [51] can effectively block SARS-CoV2-
mediated membrane fusion to combat viral infections. Since
TMPRSS2 is induced by androgens (AR), the AR anti-caking agent
Table 2
Summary of approved COVID-19 vaccines.
Name of Vaccine NRA of
record
Type
Comirnaty (developed by Pfizer and
BioNTech)
EMA/FDA mRNA vaccine
COVID-19 Vaccine Janssen EMA Adenovirus vector
vaccine
Nuvaxovid EMA Recombinant protein
vaccine
Spikevax (previously COVID-19 Vaccine
Moderna)
EMA mRNA vaccine
Vaxzevria (previously COVID-19
Vaccine AstraZeneca)
EMA Adenovirus vector
vaccine
Moderna COVID-19 Vaccine FDA mRNA vaccine
Janssen COVID-19 Vaccine FDA Adenovirus vector
vaccine
SARS-CoV-2 Vaccine (Vero Cell),
Inactivated (InCoV)
NMPA Inactivated vaccine
COVID-19 Vaccine (Vero Cell),
Inactivated/Coronavac
TM
NMPA Inactivated vaccine
Ad5-nCoV NMPA Recombinant protein
vaccine
Recombinant Novel Coronavirus
Vaccine (CHO Cell)
NMPA Recombinant protein
vaccine
SARS-CoV-2 Vaccine, Inactivated (Vero
Cell)
NMPA Inactivated vaccine
NRA: National Regulatory Authority; EMA: European Medicines Agency; FDA: the U.
S. Food and Drug Administration; NMPA: National Medical Products
Administration.
Fig. 2. The morphologic and genomic structure of SARS-CoV-2. (A) SARS-CoV-2 is an enveloped virus consisting of spike protein (S), envelope protein (E), membrane
protein (M), nucleic acid protein (N), and single-stranded RNA in the virion. (B) The genomic RNA of SARS-CoV2 is linear, positive-sense, and single-stranded with the length
of approximately 30 kb, which contains a 5
0
cap and 5
0
UTR cap, open read frame (ORF), followed by 3
0
UTR and poly(A) tail. The largest gene, ORF1ab, encodes the pp1ab
protein containing 15 nsps (nsp1 to nsp10 and nsp12 to nsp16). The pp1a protein encoded by ORF1a gene also contains 10 nsps (nsp1 to nsp10). Structural protein is encoded
by 4 structural genes, including S protein, E protein, M protein, and N protein encoding genes. Accessory genes, encoding accessory proteins such as nsp3a and nsp6p, are
distributed in structural genes.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
827
Proxalutamide (GT0918) can significantly accelerate virus clear-
ance on day 7 in patients with mild to moderate COVID-19 [52].
4.1.2. Competitive binding to ACE2 and SARS-CoV-2 S protein
Catechin and curcumin have a strong binding affinity to the
virus S protein and host receptor ACE2 and its complex, which
can trigger local structural fluctuations of the protein through their
binding with RBD/ACE2 complex [53]. In a study, using protein
engineering technology, the self-assembled tetramerization
domain from p53 protein produces a super tetravalent form of
ACE2 coupled to the Fc region of human immunoglobulin
c
1, the
high-molecular-weight Quad protein (ACE2-Fc-TD) retains the
binding to the ACE2 and its binding SARS-CoV-2 S protein and
can form a complex with the S protein and antiviral antibodies
as a bait protein [54]. Similarly, antibodies that bind to viral RBD
epitopes (such as REGE-CoV) [55,56] or that target the ACE2 recep-
tor (such as h11B11) [57] can also effectively prevent and reduce
the symptoms of viral infections. The viral load in the SARS-CoV-
2 infected animals is significantly reduced after the single-dose
h11B11 treatment, while the animals showed less interstitial
pneumonia and limited pathological features under the preventive
treatment conditions [57], which makes it a potential therapeutical
agent in COVID-19.
4.2. Viral protease inhibitors
The organic selenium compound Ebselen and its structural ana-
logs can inhibit the activity of the SARS-CoV-2 protein PLpro in the
nanomolar range [45]. Two clinical trials of Ebselen are now
recruiting moderate and severe COVID-19 patients [44]. There is
also evidence that Catechin binds to the S1 ubiquitin-binding site
of PLpro, which may inhibit its protease function and cancel the
inhibitory function of SARS-CoV-2 on the ubiquitin–proteasome
system and the interferon-stimulated gene system [58].
The viral main protease Mpro is also a widely explored drug tar-
get. Two kinds of small molecule compounds with indole structure
named GRL-1720 and 5 h inhibit the main protein Mpro of the
SARS-CoV-2, which can effectively block viral infections in vitro
[59]. Another Mpro inhibitor, GC-376, is also a promising main
candidate for further development of the treatment of SARS-CoV-
2 infection [60]. GC-376 exhibits no toxicity in K18-hACE2 mice
experiment, showing moderate benefits in terms of clinical symp-
toms, weight change, and survival rate. Under low-dose virus
attack, GC-376 can prevent the virus from reaching the brain and
reduce inflammatory cell distribution and virus staining [60]. How-
ever, the potential anti-inflammatory effect needs to be further
developed.
Fig. 3. The life cycle of SARS-CoV-2. The entry of SARS-CoV-2 is initiated through that TMPRSS2 acts on the S protein to activate the S1 and S2 subunits. The S1 subunit binds
to the ACE2 receptor to occur endocytosis (A), and S2 mediates membrane fusion (B). Subsequently, the virus releases its genomic RNA, which is translated into viral
polyprotein by the ribosome of the host cell. Under the action of lysosomal cathepsin, the virus releases genomic RNA and then is translated into viral polyprotein,
subsequently cleaved into nonstructural proteins (nsp) with viral proteases including 3CLpro and PLpro (C). SARS-CoV-2 RdRp replicates and amplifies a large number of viral
genomes, and then are transcribed and translated into four structural proteins (D). The progeny genome and structural proteins are assembled on endoplasmic reticulum-
Golgi intermediate compartment (ERGIC), which are transported and released to the outside of host cell (E). The furin on the Golgi apparatus cleavages and pre-activates the S
protein. Infected cells can fuse with neighboring cells to form syncytia. In addition, E protein in ERGIC can form pores to cause Ca
2+
to leak to achieve pro-inflammatory effects
(F).
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
828
4.3. Anti-viral RNA polymerase
The active form of Remdesivir, which has been approved for
COVID-19 treatment, acts as a nucleoside analog [61] and inhibits
RNA-dependent RNA polymerase (RdRp). Remdesivir is incorpo-
rated into the growing RNA product by RdRp [62], and the translo-
cation barrier causes RNA 3
0
-nucleotide to remain at the substrate
binding site of RdRp and interfere with the entry of the next nucle-
oside triphosphate, thereby stagnating RdRp [63]. Multiple trials
have confirmed the effectiveness of Remdesivir treatment
[64,65]. Through binding to viral RdRp, other nucleoside analogs
such as Ribavirin, Sofosbuvir, Galidesivir, Setrobuvir, and Tenofovir
behave as potent drugs against SARS-CoV-2[66,67]. Nevertheless, it
is still a long way for these FDA-approved RdRp inhibitors to apply
in COVID-19 from bench to bed.
So far, it has always been the top priority of scientists to find a
convenient, effective, and low-cost oral drug against COVID-19
because of the expensive monoclonal antibodies. On December
22, 2021, the FDA issued an emergency use authorization for Pfi-
zer’s Paxlovid, and on the following day, it urgently approved Mer-
ck’s Molnupiravir [68]. Paxlovid is composed of Nimarivir (PF-
07321332) and low-dose Ritonavir. Pfizer claims that compared
with placebo treatment group, the risk of hospitalization or death
in non-hospitalized adults at high risk of COVID-19 treated with
Paxlovid was reduced by 89%. Nimarivir is a SARS-CoV-2 3CLpro
inhibitor and can reduce virus replication [69]. Ritonavir can alle-
viate the degradation of Nimarivir and keep it at a higher concen-
tration for a long duration as possible [70]. Recently, the oral
antiviral drug Molnupiravir, an inhibitor of RdRp leading to viral
fatal mutations or premature termination of replication [71], has
attracted attention and expectation. Although Molnupirivir is
reported host mutation activity in animal cell culture experiments
[72], the safety of molnupiravir was confirmed in a drug safety
assessment [73].
4.4. Anti-viral release
The assembled progeny of SARS-CoV-2 is released by exocytosis
or budding [37,38]. Oseltamivir is a prodrug against neuraminidase
inhibitor, which has been approved for the treatment and prophy-
laxis of influenza A by inhibiting the release of progeny virion by
budding from the infected cells[74]. In both in vitro and in vivo
studies, Oseltamivir is ineffective against SARS-CoV-2 and fails to
improve the patients’ symptoms and signs in the clinic [75].
SARS-CoV-2 infected cells express Spike protein (S) on their sur-
face and fuse with ACE2-positive neighboring cells. The expression
of SARS-CoV-2 S protein in the absence of any other viral proteins
triggers the formation of syncytia [76] and determines syncytium-
mediated lymphocyte elimination [77]. The anti-helminth drug
Niclosamide was found to significantly reduce the calcium oscilla-
tion and membrane conductance in cells expressing spikes [78],
which interferes with the syncytial formation process induced by
S protein. A trial proved that Niclosamide has relatively safe clini-
cal benefits in COVID-19 management. Compared with the control
group, the cure rate of the Niclosamide treatment group increased
[79]. Niclosamide has poor oral bioavailability due to its limited
water solubility. Therefore, considering the economic issues of
treatment, it is necessary to explore its effective use.
5. Drugs targeting SARS-CoV-2-induced inflammation in host
cells
Inflammation is an early response triggered by harmful stimuli
and conditions to restore homeostasis. SARS-CoV-2 could evoke
the immune system and induce pro-inflammatory factors such as
IL-6, IL-1, TNF-
a
, and IFN [80–82] overproduction called a cytokine
storm, which brings catastrophic damage to cells and then causes
dysfunction and failure of tissues and organs [83]. The levels of
cytokines in critical COVID-19 patients are significantly higher
than those in mild conditions [84]. SARS-CoV-2 infection can cause
cytokine release syndrome (CRS) [85], which then produces a ser-
ies of consequences such as multiple organ dysfunction syndrome
(MODS), acute respiratory distress syndrome (ARDS), and even
death [86 87]. The intervention and treatment of cytokine storm
are necessary means to reduce COVID-19 mortality (Fig. 4).
5.1. Cytokine inhibitors
Overdose of interleukin-6 (IL-6) is one of the causes of cytokines
storm in COVID-19 [88]. Tocilizumab (TCZ) is the first IL-6 receptor
inhibitor discovered, which reduces the immune damage caused
by IL-6 to target cells by competitively binding to IL-6 receptors
[89]. Shreds of evidence have shown that in a limited number of
patients, symptoms, hypoxemia, and changes in chest tomography
(CT) opacity in patients treated with TCZ are immediately
improved [90]. Results from a primary intention-to-treat (ITT)
phase 2 population shows that TCZ may reduce lethality rates at
30 days [91]. In a meta-analysis, however, there was no difference
in mortality between the TCZ treatment group and the placebo
group. TCZ can reduce the length of hospital stay while the impact
on survival has not been statistically proven, but the combined use
of corticosteroids is found enable to optimize the proven effective-
ness of corticosteroids [92]. Hence, to better use TCZ in COVID-19
treatment, the combination of medication can be taken into
consideration.
Anakinra is a recombinant interleukin-1 (IL-1) receptor antago-
nist with anti-inflammatory and immunomodulatory effects [93].
A study has revealed that Anakinra significantly reduces the mor-
tality and the ICU invasive mechanical ventilation needs of
COVID-19 patients [94]. An updated meta-analysis shows that
Anakinra can reduce the 50% risk of death in hospitalized patients
with moderate to severe COVID-19 compared with patients
untreated with Anakinra [95]. Chloroquine owns an immunomod-
ulatory effect, inhibiting the production and release of TNF-
a
and
IL-6 [96], which may modulate cytokine storm in COVID-19
[97,98]. However, it has not been shown to benefit COVID-19
patients in randomized controlled trials [99].
Sarilumab is another IL-6 blocker [100,101], a fully human
immunoglobulin G1 monoclonal antibody with a high affinity for
membranes and IL-6 receptors [102]. A retrospective case from
an institution in southern Italy showed that 10 COVID-19 patients
(67%) observed rapid improvement in their respiratory parameters
after using Sarilumab [103]. In the results of the Sarilumab at a safe
dose of 400 mg and placebo groups, the COVID-19 patient mortal-
ity rates were 23% and 27%, while the hospital discharge rates were
53% and 41%, respectively [102].
Canakinumab is an IL-1 blocker developed by Novartis for the
treatment of inflammatory diseases [104]. In a study of 48 patients
with moderate COVID-19-related pneumonia, Canakinumab was
found to reduce the need for invasive mechanical ventilation.
63% of patients in the Canakinumab group were discharged within
21 days while the control group was 0% [105]. For mild or severe
COVID-19 pneumonia, a study has observed a decrease in the
inflammation index in the Canakinumab group, which can lead
to a rapid and lasting improvement in oxygenation levels [106].
With IL-12 or IL-15, IL-8 can be an effective inducer of IFN-
c
in
certain kinds of cells [107]. The combination of TNF-
a
and IFN-
c
can strongly induce cell death characterized by inflammatory cell
death [108]. In COVID-19 patients, these uncontrolled expressions
of cytokines can cause excessive damage to tissues and organs
[109]. As a result, inhibitors of abnormally increased levels of
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
829
cytokines can be used as potential drugs for COVID-19 treatment.
Meanwhile, due to the existence of synergistic effects, combination
drugs or multi-targets can be considered view of drug develop-
ment. However, the combinate use of corticosteroids is still contro-
versial. While inhibiting inflammation, it also stops the elimination
of the virus and causes a series of side effects [110,111].
5.2. Signaling pathway inhibitors
Janus Kinase (JAK)-Signal Transducer and Activator of Tran-
scription (STAT) pathway is essential for the development and
function of the immune system [112], which are involved in the
various pro-inflammatory factors [113]. Therefore, the use of JAK
inhibitors is one of the ideas to reduce the cytokine storm in
COVID-19. Baricitinib and Ruxolitinib are potent and selective
JAK inhibitors approved for indications such as rheumatoid arthri-
tis and myelofibrosis [114], which are currently reused for the
treatment of SARS-CoV-2 infected patients. Studies have confirmed
the safety and effectiveness of these drugs in reducing cytokine
levels in the treatment of COVID-19 as antiviral drugs [115–117].
In addition, Baricitinib has the effect of reducing the endocytosis
of the SARS-CoV-2 [118]. The combined use of Baritinib and
Remdesivir can shorten the recovery time and accelerate the
improvement of clinical status, especially for patients receiving
high-flow oxygen or non-invasive ventilation [119].
The nuclear factor NF-
j
B pathway is a classic pro-inflammatory
signaling pathway [120]. The inhibition of NF-
j
B signaling has
therapeutic applications in cancer and inflammatory diseases
[121], as well as virus- and LPS- induced cytokine storms [122].
The use of NF-
j
B inhibitors Caffeic acid phenethyl ester (CAPE)
and Parthenolide can improve the survival rate of mice infected
with SARS-CoV [123]. Considering the spike protein of coronavirus
upregulates IL-6 and TNF-
a
in murine macrophages through the
NF-
j
B pathway [124], the exploration of NF-
j
B inhibitors may
be an effective measure to reduce the cytokine storm upon SARS-
CoV-2 infection. Although some studies have found potential
new drugs that can be used to inhibit NF-
j
B to inhibit SARS-
CoV-2 infection, [125,126], whether they can be applied to the
treatment of COVID-19 remains to be investigated.
In a trial to screen drugs with SARS-CoV-2-related pangolin
coronavirus model, the Cepharanthine (CEP) showed a significant
antiviral effect [127]. CEP inhibits viral replication by modulating
signaling pathways [128,129], among which it exhibits anti-
inflammatory effects through AMPK activation and NF-
j
B inhibi-
tion[130]. Another study also confirmed the antiviral effect of
CEP, and the combination of CEP and Nelfinavir can enhance their
efficacy [131]. Existing evidence shows that CEP has important
potential value in the treatment of COVID-19 [132], it still needs
further verification by in vivo experiments and clinical trials.
5.3. Steroids treatment
Pidotimod is a synthetic dipeptide molecule, which has biolog-
ical and immunological activity on both the adaptive and the
innate immune responses [133]. Pidotimod can increase the
expression of NF-
j
B protein without an increase of IL-8 expression
[134]. Pidotimod has been evaluated as an immunostimulant for
respiratory infections (RTIs) [135] and the treatment of many other
diseases [136]. A controlled experiment involving 20 COVID-19
patients showed that Pidotimod can effectively improve the fever
of patients and prevent the activation of the cytokine cascade
[137].
Viral infections usually cause excessive hyperinflammation
[138]. The anti-inflammatory effect of steroids can stabilize hemo-
dynamics and shorten the stay time of the intensive care unit (ICU)
and the duration of mechanical ventilation. The use of steroids has
always been controversial. Steroids can significantly reduce anti-
inflammatory factors and may also delay virus clearance [139].
Long-term use of steroids can cause many adverse events, such
as secondary infections [140]. In addition, The correlation between
steroids and mortality remains unclear [141]. Steroids are cur-
rently used in critically ill patients to reduce clinical deterioration.
One meta-analysis shows that the use of steroids is associated with
higher mortality [142], whereas another prospective meta-analysis
Fig. 4. Summary of therapeutic targets to SARS-CoV-2-induced inflammation in host cells. SARS-CoV-2 could evoke the immune system and trigger cytokine release
syndrome (CRS) and cytokine storm, leading to multiple organ dysfunction syndrome (MODS), acute respiratory distress syndrome (ARDS), and even death. The therapuetic
inventations are introduced to alleviate inflammation to reduce the severity of COVID-19.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
830
of seven randomized trials showed that the use of systemic corti-
costeroids was associated with lower all-cause mortality from
COVID-19 [143]. The contradictory results indicate that there is
still uncertainty in the role of corticosteroids, and more random-
ized controlled trials are needed to prove its effectiveness.
It is undeniable that corticosteroids have been widely used in
the treatment of severe COVID-19 cases [144]. A trial showed that
the 28-day mortality is reduced in COVID-19 patients receiving
respiratory support after receiving up to ten days of dexametha-
sone treatment, which is useless for patients who do not require
respiratory support [145]. The use of cedemethasone can reduce
lung damage in COVID-19 patients [146], which is recognized by
WHO in the clinical trial [147]. Whether other steroids have the
same effects as dexamethasone is still unknown [148]. The combi-
nation of other drugs may alleviate the side effects of steroid use.
An observational cohort study found that the combination of
Barectinib and corticosteroids in the COVID-19 treatment can sig-
nificantly improve lung function compared with corticosteroids
alone [149]. Nonetheless, how to maximize the drug effect with a
lower dose and shorter treatment time is another urgent problem
that needs to be solved.
5.4. Holistic therapy and traditional Chinese medicine treatment
Rehabilitation plasma therapy can be taken in the early stage of
COVID-19 by using antibodies in the convalescent serum to neu-
tralize the virus to reduce the body damage caused by the immune
system [150]. After the occurrence of cytokine storm, patients will
have a variety of symptoms, such as MODS and ARDS. At this stage,
anti-shock treatment is required to maintain the patient’s body
homeostasis and protect important organ functions.
In addition to conventional treatment, traditional Chinese med-
icine (TCM) plays an important role as an auxiliary method in clin-
ical COVID-19 treatment, which has been confirmed to have
antiviral activity against various coronavirus strains [151]. TCM
treatment can improve clinical efficacy and alleviate COVID-19
patients’ severe conditions [152,153]. Since TCM recipes were
according to the Diagnosis and Treatment Protocol for Novel
Coronavirus Pneumonia (Trial Version 7) [154], 92% of all of the
confirmed cases in China had taken TCM in COVID-19 treatment
[155]. Different recipes are recommended for different severity of
conditions in COVID-19 treatment. Namely, Lianhua Qingwen
(LHQW), Jinhua Qinggan (JHQG), Hua Shibaidu Granules (HSBD),
Xuanfeibaidu Granules (XFBD), Xuebijing Injection (XBJ), and Qing-
fei Paidu Decoction are common TCM recipes [155].
In Vero cells, LHQW reduced the mRNA expression of
pro-inflammatory cytokines TNF-
a
, IL-6, CCL-2/MCP-1, and
CXCL-10/IP-10 [156]. A comparative study of multiple combination
medications shows that the quadruple combination with LHQW
can be the first choice for the treatment of patients in critical
condition [157]. In detail, Rhein, forsythoside A, forsythoside I,
neochlorogenic acid and its isomers may be the effective active
ingredients of LHQW against SARS-CoV-2 [158]. A retrospective
analysis of JHQG treatment promotes the absorption of pneumonia
permeate with a higher 7-day viral clearance rate compared with
the control group [159]. HSBD is suitable for early COVID-19
treatment [160], while XBJ is mainly used for patients in critical
conditions [161]. The XBJ may relieve systemic inflammation by
inhibiting the secretion of pro-inflammatory cytokines mediated
by the HMGB1/RAGE axis to reduce mortality [162,163].
Traditional Chinese medicine has played a huge role in China’s
anti-COVID-19 epidemic battle. However, there are many
unsolved issues for Chinese scientists and researchers to precise
the administration of TCM in COVID-19 treatment in the future.
The concerning characteristics of TCM treatment are that the
types and dosages of medications often change due to the
different conditions of different patients. The molecular mecha-
nisms of most TCMs are unclear, which raises the limitations in
the global promotion of TCM against COVID-19 pandemic. Nota-
bly, the standardization behind different kinds of traditional Chi-
nese medicines with various functions is highlighted and needs to
be concerned.
Table 3
Detailed information on emerging SARS-CoV-2 variants.
VOC
WHO
Label
Pango lineage Earliest
Documented
Samples
Key Mutation Sites
Alpha B.1.1.7 United
Kingdom in
Sep-2020
DEL69/70; DEL144/145; N501Y;
A570D; D614G; P681H; T716I;
S982A; D1118H
Beta B.1.351 South Africa
in May-2020
D80A; D215G; DEL241/243;
K417N; E484K; N501Y; D614G;
A701V
B.1.351.2 L18F; D80A; D215G; DEL241/
243; K417N; E484K; N501Y;
D614G; A701V
B.1.351.3
Gamma P.1 Brazil in
Nov-2020
L18F; T20N; P26S; D138Y;
R190S; K417T; E484K; N501Y;
D614G; H655Y; T1027I; V1176F
Delta B.1.617.2 India in Oct-
2020
T19R; G142D; E156G; DEL157/
158; L452R; T478R; T478K;
D614G; P681R; D950N
AY.1 T19R; W258L; K417N; L452R;
T478K; D614G; P681R; D950N
AY.2 T19R; V70F; E156G; DEL157/
158; A222V; K417N; L452R;
T478K; D614G; P681R; D950N
AY.3 T19R; E156G; DEL157/158;
L452R; T478K; D614G; P681R;
D950N
Omicron B.1.1.529 South Africa
in Nov-2020
A67V; DEL69/70; T95I; G142D;
DEL143/145; T547K; D614G;
H655Y; N679K; P681H; D796Y;
N856K; Q954H; N969K; L981F
BA.1 A67V; DEL69/70; T95I; G142D;
DEL143/145; N211I; DEL212/
212; G339D; S371L; S373P;
S375F; S477N; T478K; E484A;
Q493R; G496S; Q498R; N501Y;
Y505H; T547K; D614G; H655Y;
N679K; P681H; D796Y; N856K;
Q954H; N969K; L981F
BA.2 T19I; L24S; DEL25/27; G142D;
V213G; G339D; S371F; S373P;
S375F; T376A; D405N; R408S;
K417N; N440K; S477N; T478K;
E484A; Q493R; Q498R; N501Y;
Y505H; D614G; H655Y; N679K;
P681H; N764K; D796Y; Q954H;
N969K
BA.3 A67V; DEL69/70; T95I; G142D;
DEL143/145; N211I; DEL212/
212; G339D; S371F; S373P;
S375F; D405N; S477N; T478K;
E484A; Q493R; Q498R; N501Y;
Y505H; D614G; H655Y; N679K;
P681H; N764K; D796Y; Q954H;
N969K
VOI
Lambda C.37 Peru in Dec-
2020
G75V; T76I; R246N; DEL247/
253; L452Q; F490S; D614G;
T859N
Mu B.1.621 Columbia
Oct-2020
T95I; Y144S; Y145N; R346K;
E484K; N501Y; D614G; P681H;
D950N
The mutations of amino acid in SARS-CoV-2 S protein are presented. The summa-
rized information represented in the table was derived from the public data
(https://outbreak.info/situation-reports#custom-report, as of 26 November 2021).
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
831
6. Emerging SARS-CoV-2 variants
SARS-CoV-2 has been continuing to evolve, posing higher infec-
tivity efficiency and faster transmission, leading to a greater risk to
global public health. To better assess the consequences of different
variants and facilitate prevention measures or medical counter-
measures, WHO divides them into variants of interest (VOI) and
variants of concern (VOC) [164]. There are currently two VOIs:
Kappa and Mu; and five VOCs: Alpha, Beta, Gamma, Delta, and
Omicron (Table 3). On November 24, 2021, a new SARS-CoV-2 vari-
ant B.1.1.529 named Omicron was discovered in South Africa. Pre-
viously, the SARS-CoV-2 Delta variant has become the main
epidemic strain in many countries [165]. Now, the emergence of
omicron has aroused new attention and vigilance. The mutations
in Omicron are concentrated in the S protein, and there seems to
be a tendency to collect mutations that are beneficial to immune
escape [166,167]. A model predicts and calculates that Omicron’s
infectivity is about ten times that of the original virus or twice that
of the Delta variant. Omicron may greatly undermine the efficacy
of the Eli Lilly monoclonal antibody (mAb) approved by the FDA,
and may also reduce the efficacy of mAbs from Celltrion and Rock-
efeller University [168]. The main SARS-CoV-2 variants reported in
different places worldwide create new concerns about anti-viral
drugs and vaccinations against COVID-19 pandemic (Fig. 5). The
following critical mutation sites in SARS-CoV-2 genome determine
the virulence and spread of the SARS-CoV-2 (Table 4), which pro-
vides fresh ideas in the drug design for the main emerging variants.
6.1. Mutations in structural proteins
D614G mutation in S protein will not significantly change the
neutralizing properties of antibodies against SARS-CoV-2, and the
currently developed vaccine against wildtype is still effective
against the D614G strain [169]. The presence of the E484K muta-
tion located in the receptor-binding domain (RBD) of the S protein
will reduce the neutralizing power of a variety of effective mAbs
that change the receptor-binding motif on RBD [170]. The intro-
duction of E484K mutations into other variant strains will also lead
to a decrease in the neutralization of antibodies and monoclonal
antibodies caused by the vaccine [171]. N501Y mutation found in
the British variant strain B.1.1.7 has almost no effect on the neu-
tralizing effect of neutralizing nanobodies (Nbs) [172]. However,
the mutant strain often accompanies other key amino acid muta-
tions that affect the binding of S protein to ACE2. L18F mutation
occurs in the NTD region and can cancel the binding of S2L28 mon-
oclonal antibody to NTD [173] and the L18F mutation is positively
correlated with mortality [174]. K417N mutation, close to the ACE2
binding site, is slightly detrimental to ACE2 binding [171], but it
can promote the process of variants effectively avoiding antibodies
by eliminating the buried interface salt bridge between RBD and
neutralizing antibody CB6 [175]. L452R mutation locates in the
RBD hydrophobic plaques of the spike protein [176]. It will
increase the affinity of the spike protein to ACE2 and is kind of
destructive to NAb binding [177] and make the virus evade the
monoclonal antibody LY-CoV555 [178]. P681R is a mutation near
the furin-cleavage site [179], which enhances the cleavage of S1
and S2 by the full-length spike, resulting in increased infection
through the cell surface [180].
6.2. Mutations in non-structural proteins
Among the SARS-CoV-2 nonstructural proteins (nsp), virus-
associated enzymes such as RdRp and 3C are important drug tar-
gets [181]. Mutations at these sites may increase virus resistance
to related drugs [182,183]. Mutations in RdRp, for example, may
Fig. 5. The appearance of mutant strains of SARS-CoV-2 around the world. The
main emerging SARS-CoV-2 variant strains are classified as VOC (variants of
concern), VOI (variants of interest), and VUM (variants under monitoring). VOC
includes Alpha, Beta, Gamma, Delta, and Omicron; VOI contains Lambda and Mu,
while VUM includes Theta, Eta, Iota, and Kappa from the public data (https://
outbreak.info/situation-reports#custom-report, as of 26 November 2021). The time
of appearance is expressed as yy/mm.
Table 4
Mutations of concern and interest reports in SARS-CoV-2 variants.
Mutation of Concern Reports
Mutation Prominent in VOC Prominent in
VOI
E484K B.1.351; B.1.351.2; B.1.351.3; P.1 B.1.621
Mutation of Interest Reports
Mutation Prominent in VOC Prominent in
VOI
L18F B.1.351.2; B.1.351.3; P.1 /
K417N B.1.351; B.1.351.2; B.1.351.3; AY.1; AY.2; BA.2 /
K417T P.1 /
L452R B.1.617.2; AY.1; AY.2; AY.3 /
N501Y B.1.1.7; B.1.351; B.1.351.2; B.1.351.3; P.1; BA.1;
BA.2; BA.3
B.1.621
P681R B.1.617.2; AY.1; AY.2; AY.3 /
The mutations of amino acid in SARS-CoV-2 S protein are presented. The summa-
rized information represented in the table was derived from the public data
(https://outbreak.info/situation-reports#custom-report, as of 26 November 2021).
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
832
reduce the effect of Remdesivir. Studies have shown that the
14,408 C>T mutation increases the viral mutation rate, while
15324 C>T has a reduced effect [184]. Mutations in nsp may corre-
late with the degree of symptoms caused by SARS-CoV-2. A study
found that mutations located in the nsp6 coding region were sig-
nificantly associated with asymptomatic COVID-19 [185], which
could make it difficult to initially screen COVID-19 patients. Differ-
ent nsps have their structures and functions, which can be changed
by the mutations [186]. For example, the V121D mutation in nsp1
may have a disruptive effect on it, and the G1691C in Nsp3 reduces
the flexibility of the protein [187]. Therefore, these mutations in
nsps should be taken into account in drug and vaccine develop-
ment or treatment against COVID-19.
7. Perspectives in the drug design for SARS-CoV-2 variants
The current SARS-CoV-2 genome site mutations are all subject
to natural selection and drug screening, which reflect the adapta-
tion of the virus to the treatments. Some mutations make the virus
increase the infectivity and transmission, however, in terms of
treatment, the previous drugs are still effective against SARS-
CoV-2 variants [188]. Other mutations render part of the antibody
ineffective against SARS-CoV-2 [189], and the virus escapes from
immunity, which brings new challenges to treatment. Therefore,
the exploration of broadly antiviral drugs is prior to being concen-
trated and developed. More importantly, the immunomodulatory
and holistic therapy in host, including anti-inflammatory strategies
and Chinese traditional medicine treatment, should be concerned
in the drug design for SARS-CoV-2 variants.
The binding of SARS-CoV-2 S protein to the ACE2 receptor is a
key activity for the virus to invade the human body, and most of
the mutation sites that are currently being studied are located in
the S protein [190]. Researchers need to test one by one to deter-
mine the impact of specific mutation sites on the life of the
SARS-CoV-2 and then test whether the previous treatments such
as monoclonal antibodies are still effective for the emerging vari-
ants. However, a SARS-CoV-2 variant often carries a combination
of different mutation sites and becomes a multiple mutant strain,
and the speed of mutation is rapid [177]. It is of high concern that
the effects from different combinations of mutation sites will have
on viral infection activities. As a result, the highly conservative
sites of the SARS-CoV-2 genome as drug targets could be paid more
attention to maintaining antiviral activities against variants.
8. Discussion
SARS-CoV-2 has caused a global pandemic since late 2019. With
the increase of experience and knowledge, defensive measures and
clinical treatment plans are adopted immediately according to
observation and management of COVID-19 pandemic. Meanwhile,
researchers are exploring more possibilities of repurposing drugs
against SARS-CoV-2 [191]. However, the development of specific
drugs and vaccines requires fundamental studies on SARS-CoV-2,
such as the interaction between the virus and the host, the epi-
demiology and molecular virology, the host immune responses to
viral infection, to uncover the mechanism underlying the infection,
transmission, and pathogenesis of the virus and explore effective
drugs.
Traditional Chinese medicine has played a huge role in China’s
anti-epidemic process [192]. Research on the mechanism of action
of traditional Chinese medicine may provide new ideas for the
development of anti-SARS-CoV-2 drugs. At the same time, it is nec-
essary to pullulate holistic treatments to deal with the systemic
pathological imbalance caused by the SARS-CoV-2.
Owing to the error-prone RNA replicase of SARS-CoV-2 [193],
the continuous emergence of mutant strains has undoubtedly
brought huge difficulties and challenges to the control of the global
COVID-19 epidemic. The appearance of SARS-CoV-2 variants
means that the virus adapts to manual interventions and natural
selection, which may eliminate the effectiveness of previous drugs
and vaccines. In this case, we must maintain epidemiological
surveillance of COVID-19 timely and focus on the evolution of
the virus, especially the emergence of drug-resistant strains.
The pathogenesis of COVID-19 can be mainly divided into two
phases [194]. In the early stage, the COVID-19 patients may begin
with a series of mild symptoms including fever, cough, fatigue,
hemoptysis, headache, or diarrhea, and then will develop pneumo-
nia [155,195]. Accordingly, antiviral drugs can be given to clear the
virus from the body of patients infected with SARS-CoV-2. Later on,
as the viral load increases and spreads in the human body, which
triggers excessive production of cytokines, a cytokine storm occurs
in the COVID-19 patients to deteriorate to moderate and severe
symptoms with respiratory distress syndrome and organ failure
[196,197]. It should be necessary to adopt antiviral drugs with
immunomodulatory treatment, and also encouraged to introduce
holistic therapy and traditional Chinese medicine treatment, and
even external mechanical equipment and maintain the normal
operation of the patient’s body [198].
In sum, SARS-CoV-2 mutation is an urgent issue that needs to be
explored and solved, which requires concerted efforts from the
fields of structural biology, medicine, virology, pharmacy, public
health, epidemiology, chemistry, and other disciplines, to jointly
deal with the treatment and control of COVID-19 pandemic.
CRediT authorship contribution statement
Jialing Yin: Conceptualization, Data curation, Investigation,
Writing – original draft. Chengcheng Li: Conceptualization, Data
curation, Investigation, Writing – original draft. Chunhong Ye:
Data curation, Investigation. Zhihui Ruan: Data curation, Formal
analysis, Investigation. Yicong Liang: Data curation, Formal analy-
sis, Methodology, Investigation. Yongkui Li: Formal analysis,
Methodology, Validation. Jianguo Wu: Conceptualization, Valida-
tion, Writing – review & editing. Zhen Luo: Conceptualization, Val-
idation, Funding acquisition, Supervision, Writing – original draft,
Writing – review & editing.
Declaration of Competing Interest
The authors declare that they have no known competing finan-
cial interests or personal relationships that could have appeared
to influence the work reported in this paper.
Acknowledgements
This study was supported by the National Natural Science Foun-
dation of China [32070148], the Guangdong Basic and Applied
Basic Research Foundation [2019A1515011073], and Guangzhou
Basic Research Program - Basic and Applied Basic Research Project
[202102020260].
References
[1] Coronaviridae Study Group of the International Committee on Taxonomy of V.
The species Severe acute respiratory syndrome-related coronavirus:
classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol
2020;5:536–44.
[2] Malik YA. Properties of Coronavirus and SARS-CoV-2. Malays J Pathol
2020;42:3–11.
[3] Velavan TP, Meyer CG. The COVID-19 epidemic. Trop Med Int Health
2020;25:278–80.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
833
[4] Li H, Liu Z, Ge J. Scientific research progress of COVID-19/SARS-CoV-2 in the
first five months. J Cell Mol Med 2020;24:6558–70.
[5] W.H. Organization, Status of COVID-19 Vaccines within WHO EUL/PQ
evaluation process, (2021) (https://www.who.int/publications/m/item/draft-
landscape-of-covid-19-candidate-vaccines).
[6] W.H. Organization, COVID-19 Weekly Epidemiological Update, (2022) Edition
74.
[7] Beeraka NM, Sadhu SP, Madhunapantula SV, Rao Pragada R, Svistunov AA,
Nikolenko VN, et al. Strategies for targeting SARS CoV-2: small molecule
inhibitors-the current status. Front Immunol 2020;11:552925.
[8] Zhou Y-W, Xie Y, Tang L-S, Pu D, Zhu Y-J, Liu J-Y, et al. Therapeutic targets and
interventional strategies in COVID-19: mechanisms and clinical studies.
Signal Transduct Target Ther 2021;6(1).
[9] McKee DL, Sternberg A, Stange U, Laufer S, Naujokat C. Candidate drugs
against SARS-CoV-2 and COVID-19. Pharmacol Res 2020;157:104859.
[10] Habler K, Brugel M, Teupser D, Liebchen U, Scharf C, Schonermarck U, et al.
Simultaneous quantification of seven repurposed COVID-19 drugs remdesivir
(plus metabolite GS-441524), chloroquine, hydroxychloroquine, lopinavir,
ritonavir, favipiravir and azithromycin by a two-dimensional isotope dilution
LC-MS/MS method in human serum. J Pharm Biomed Anal 2021;196:113935.
[11] Fischer W, Eron JJ, Holman W, M.S. Cohen, L. Fang, L.J. Szewczyk, T.P. Sheahan,
R. Baric, K.R. Mollan, C.R. Wolfe, E.R. Duke, M.M. Azizad, K. Borroto-Esoda, D.A.
Wohl, A.J. Loftis, P. Alabanza, F. Lipansky, W.P. Painter, Molnupiravir, an oral
antiviral treatment for COVID-19, medRxiv, (2021), 10.1101/
2021.06.17.21258639v1.
[12] Venkadapathi J, Govindarajan VK, Sekaran S, Venkatapathy S. A minireview of
the promising drugs and vaccines in pipeline for the treatment of COVID-19
and current update on clinical trials. Front Mol Biosci 2021;8:637378.
[13] Zahradník J, Marciano S, Shemesh M, Zoler E, Harari D, Chiaravalli J, et al.
SARS-CoV-2 variant prediction and antiviral drug design are enabled by RBD
in vitro evolution. Nat Microbiol 2021;6(9):1188–98.
[14] Liu C, Ginn HM, Dejnirattisai W, Supasa P, Wang B, Tuekprakhon A, et al.
Reduced neutralization of SARS-CoV-2 B.1.617 by vaccine and convalescent
serum. Cell 2021;184(16):4220–4236.e13.
[15] Neuman BW, Kiss G, Kunding AH, Bhella D, Baksh MF, Connelly S, et al. A
structural analysis of M protein in coronavirus assembly and morphology. J
Struct Biol 2011;174(1):11–22.
[16] McBride R, van Zyl M, Fielding B. The coronavirus nucleocapsid is a
multifunctional protein. Viruses 2014;6(8):2991–3018.
[17] Schoeman D, Fielding BC. Coronavirus envelope protein: current knowledge.
Virol J 2019;16:69.
[18] Song HC, Seo M-Y, Stadler K, Yoo BJ, Choo Q-L, Coates SR, et al. Synthesis and
characterization of a native, oligomeric form of recombinant severe acute
respiratory syndrome coronavirus spike glycoprotein. J Virol 2004;78
(19):10328–35.
[19] Belouzard S, Millet JK, Licitra BN, Whittaker GR. Mechanisms of coronavirus
cell entry mediated by the viral spike protein. Viruses 2012;4(6):1011–33.
[20] Gong Y, Qin S, Dai L, Tian Z. The glycosylation in SARS-CoV-2 and its receptor
ACE2. Signal Transduct Target Ther 2021;6:396.
[21] Grant OC, Montgomery D, Ito K, Woods RJ. Analysis of the SARS-CoV-2 spike
protein glycan shield reveals implications for immune recognition. Sci Rep
2020;10:14991.
[22] Watanabe Y, Berndsen ZT, Raghwani J, Seabright GE, Allen JD, Pybus OG, et al.
Vulnerabilities in coronavirus glycan shields despite extensive glycosylation.
Nat Commun 2020;11:2688.
[23] Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure,
function, and antigenicity of the SARS-CoV-2 Spike glycoprotein. Cell
2020;181:281–292 e286.
[24] Reis CA, Tauber R, Blanchard V. Glycosylation is a key in SARS-CoV-2
infection. J Mol Med (Berl) 2021;99:1023–31.
[25] Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, et al. Angiotensin-
converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature
2003;426:450–4.
[26] Majumder J, Minko T. Recent developments on therapeutic and diagnostic
approaches for COVID-19. AAPS J 2021;23:14.
[27] Wei C, Wan L, Yan Q, Wang X, Zhang J, Yang X, et al. HDL-scavenger receptor B
type 1 facilitates SARS-CoV-2 entry. Nat Metab 2020;2(12):1391–400.
[28] Wruck W, Adjaye J. SARS-CoV-2 receptor ACE2 is co-expressed with genes
related to transmembrane serine proteases, viral entry, immunity and
cellular stress. Sci Rep 2020;10:21415.
[29] Zhai X, Sun J, Yan Z, Zhang J, Zhao J, Zhao Z, et al. Comparison of severe acute
respiratory syndrome coronavirus 2 spike protein binding to ACE2 receptors
from human, pets, farm animals, and putative intermediate hosts. J Virol
2020;94(15). https://doi.org/10.1128/JVI.00831-20.
[30] Shang J, Wan Y, Luo C, Ye G, Geng Q, Auerbach A, et al. Cell entry mechanisms
of SARS-CoV-2. Proc Natl Acad Sci U S A 2020;117(21):11727–34.
[31] Ziebuhr J, Siddell SG. Processing of the human coronavirus 229E replicase
polyproteins by the virus-encoded 3C-like proteinase: identification of
proteolytic products and cleavage sites common to pp1a and pp1ab. J Virol
1999;73:177–85.
[32] Ziebuhr J, Herold J, Siddell SG. Characterization of a human coronavirus
(strain 229E) 3C-like proteinase activity. J Virol 1995;69:4331–8.
[33] Harcourt BH, Jukneliene D, Kanjanahaluethai A, Bechill J, Severson KM, Smith
CM, et al. Identification of severe acute respiratory syndrome coronavirus
replicase products and characterization of papain-like protease activity. J
Virol 2004;78:13600–12.
[34] Wang MY, Zhao R, Gao LJ, Gao XF, Wang DP, Cao JM. SARS-CoV-2: structure
biology, and structure-based therapeutics development. Front Cell Infect
Microbiol 2020;10:587269.
[35] Hillen HS, Kokic G, Farnung L, Dienemann C, Tegunov D, Cramer P. Structure
of replicating SARS-CoV-2 polymerase. Nature 2020;584:154–6.
[36] Hsu JC, Laurent-Rolle M, Pawlak JB, Wilen CB, Cresswell P. Translational
shutdown and evasion of the innate immune response by SARS-CoV-2 NSP14
protein. Proc Natl Acad Sci U S A 2021;118.
[37] Zhu N, Wang W, Liu Z, Liang C, Wang W, Ye F, et al. Morphogenesis and
cytopathic effect of SARS-CoV-2 infection in human airway epithelial cells.
Nat Commun 2020;11:3910.
[38] Obukhov AG, Stevens BR, Prasad R, Li Calzi S, Boulton ME, Raizada MK, et al.
SARS-CoV-2 infections and ACE2: clinical outcomes linked with increased
morbidity and mortality in individuals with diabetes. Diabetes
2020;69:1875–86.
[39] Nieto-Torres JL, Verdia-Baguena C, Jimenez-Guardeno JM, Regla-Nava JA,
Castano-Rodriguez C, Fernandez-Delgado R, et al. Severe acute respiratory
syndrome coronavirus E protein transports calcium ions and activates the
NLRP3 inflammasome. Virology 2015;485:330–9.
[40] Harrison AG, Lin T, Wang P. Mechanisms of SARS-CoV-2 transmission and
pathogenesis. Trends Immunol 2020;41:1100–15.
[41] Schneider CV, Strnad P. SARS-CoV-2 infection in alpha1-antitrypsin
deficiency. Respir Med 2021;184:106466.
[42] Yang C, Chapman KR, Wong A, Liu M. alpha1-Antitrypsin deficiency and the
risk of COVID-19: an urgent call to action. Lancet Respir Med 2021;9:337–9.
[43] Faria N, Ines Costa M, Gomes J, Sucena M. Alpha-1 antitrypsin deficiency
severity and the risk of COVID-19: a Portuguese cohort. Respir Med
2021;181:106387.
[44] clinicaltrials.gov (https://clinicaltrials.gov/ct2/results?term=Alpha1-
Proteinase+Inhibitor&cond=COVID-19&draw=2&rank=1#rowId0).
[45] Weglarz-Tomczak E, Tomczak JM, Talma M, Burda-Grabowska M, Giurg
M, Brul S. Identification of ebselen and its analogues as potent covalent
inhibitors of papain-like protease from SARS-CoV-2. Sci Rep
2021;11:3640.
[46] Sandoval DR, Clausen TM, Nora C, Cribbs AP, Denardo A, Clark AE, et al., The
Prolyl-tRNA synthetase inhibitor halofuginone inhibits SARS-CoV-2 Infection,
bioRxiv, (2021) (https://www.biorxiv.org/content/10.1101/
2021.03.22.436522v3).
[47] Pines M, Spector I. Halofuginone – the multifaceted molecule. Molecules
2015;20:573–94.
[48] Stecklair KP, Hamburger DR, Egorin MJ, Parise RA, Covey JM, Eiseman JL.
Pharmacokinetics and tissue distribution of halofuginone (NSC 713205) in
CD2F1 mice and Fischer 344 rats. Cancer Chemother Pharmacol
2001;48:375–82.
[49] Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between
ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med 2020;76:14–20.
[50] Kim TY, Jeon S, Jang Y, Gotina L, Won J, Ju YH, et al. Platycodin D, a natural
component of Platycodon grandiflorum, prevents both lysosome- and
TMPRSS2-driven SARS-CoV-2 infection by hindering membrane fusion. Exp
Mol Med 2021;53:956–72.
[51] Xia S, Liu M, Wang C, Xu W, Lan Q, Feng S, et al. Inhibition of SARS-CoV-2
(previously 2019-nCoV) infection by a highly potent pan-coronavirus fusion
inhibitor targeting its spike protein that harbors a high capacity to mediate
membrane fusion. Cell Res 2020;30:343–55.
[52] Cadegiani FA, McCoy J, Gustavo Wambier C, Vano-Galvan S, Shapiro J, Tosti A,
et al. Proxalutamide significantly accelerates viral clearance and reduces time
to clinical remission in patients with mild to moderate COVID-19: results
from a randomized, double-blinded, placebo-controlled trial. Cureus
2021;13:e13492.
[53] Jena AB, Kanungo N, Nayak V, Chainy GBN, Dandapat J. Catechin and
curcumin interact with S protein of SARS-CoV2 and ACE2 of human cell
membrane: insights from computational studies. Sci Rep 2021;11:2043.
[54] Miller A, Leach A, Thomas J, McAndrew C, Bentley E, Mattiuzzo G, et al. A
super-potent tetramerized ACE2 protein displays enhanced neutralization of
SARS-CoV-2 virus infection. Sci Rep 2021;11:10617.
[55] Copin R, Baum A, Wloga E, Pascal KE, Giordano S, Fulton BO, et al. The
monoclonal antibody combination REGEN-COV protects against SARS-CoV-2
mutational escape in preclinical and human studies. Cell
2021;184:3949–3961 e3911.
[56] O’Brien MP, Forleo-Neto E, Sarkar N, Isa F, Hou P, Chan KC, et al.,
Subcutaneous REGEN-COV Antibody Combination in Early SARS-CoV-2
Infection, medRxiv, (2021) (https://www.medrxiv.org/content/10.1101/
2021.06.14.21258569v2).
[57] Du Y, Shi R, Zhang Y, Duan X, Li L, Zhang J, et al. A broadly neutralizing
humanized ACE2-targeting antibody against SARS-CoV-2 variants. Nat
Commun 2021;12:5000.
[58] Chourasia M, Koppula PR, Battu A, Ouseph MM, Singh AK. EGCG, a green tea
catechin, as a potential therapeutic agent for symptomatic and asymptomatic
SARS-CoV-2 infection. Molecules 2021;26:1200.
[59] Hattori SI, Higashi-Kuwata N, Hayashi H, Allu SR, Raghavaiah J, Bulut H, et al.
A small molecule compound with an indole moiety inhibits the main
protease of SARS-CoV-2 and blocks virus replication. Nat Commun
2021;12:668.
[60] Caceres CJ, Cardenas-Garcia S, Carnaccini S, Seibert B, Rajao DS, Wang J, et al.
Efficacy of GC-376 against SARS-CoV-2 virus infection in the K18 hACE2
transgenic mouse model. Sci Rep 2021;11:9609.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
834
[61] Hashemian SM, Farhadi T, Velayati AA. A review on Remdesivir: a possible
promising agent for the treatment of COVID-19. Drug Des Devel Ther
2020;14:3215–22.
[62] Jorgensen SCJ, Kebriaei R, Dresser LD. Remdesivir: review of pharmacology,
pre-clinical data, and emerging clinical experience for COVID-19.
Pharmacotherapy 2020;40:659–71.
[63] Kokic G, Hillen HS, Tegunov D, Dienemann C, Seitz F, Schmitzova J, et al.
Mechanism of SARS-CoV-2 polymerase stalling by remdesivir. Nat Commun
2021;12:279.
[64] Rosenberg K. Remdesivir in the treatment of COVID-19. Am J Nurs
2021;121:55.
[65] Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al.
Remdesivir for the treatment of Covid-19 – final report. N Engl J Med
2020;383:1813–26.
[66] Elfiky AA. SARS-CoV-2 RNA dependent RNA polymerase (RdRp) targeting: an
in silico perspective. J Biomol Struct Dyn 2021;39:3204–12.
[67] Elfiky AA. Ribavirin, remdesivir, sofosbuvir, galidesivir, and tenofovir against
SARS-CoV-2 RNA dependent RNA polymerase (RdRp): a molecular docking
study. Life Sci 2020;253:117592.
[68] T.U.S.F.a.D. Administration, Press Announcements (https://www.fda.gov/
news-events/fda-newsroom/press-announcements).
[69] Macchiagodena M, Pagliai M, Procacci P. Characterization of the non-covalent
interaction between the PF-07321332 inhibitor and the SARS-CoV-2 main
protease. J Mol Graph Model 2022;110:108042.
[70] Pfizer, Pfizer’s Novel COVID-19 Oral Antiviral Treatment Candidate Reduced
Risk of Hospitalization or Death by 89% in Interim Analysis of Phase 2/3 EPIC-
HR Study, (2021) (https://www.pfizer.com/news/press-release/press-release-
detail/pfizers-novel-covid-19-oral-antiviral-treatment-candidate).
[71] Imran M, Kumar Arora M, Asdaq SMB, Khan SA, Alaqel SI, Alshammari MK,
et al., Discovery, Development, and patent trends on molnupiravir: a
prospective oral treatment for COVID-19, Molecules, 26 (2021) 5795.
[72] Zhou S, Hill CS, Sarkar S, Tse LV, Woodburn BMD, Schinazi RF, et al. beta-d-
N4-hydroxycytidine Inhibits SARS-CoV-2 through lethal mutagenesis but is
also mutagenic to mammalian cells. J Infect Dis 2021;224:415–9.
[73] Hiremath CN. Abbreviated Profile of Drugs (APOD): modeling drug safety
profiles to prioritize investigational COVID-19 treatments. Heliyon 2021;7:
e07666.
[74] Langlois RA, Meyerholz DK, Coleman RA, Cook RT, Waldschmidt TJ, Legge KL.
Oseltamivir treatment prevents the increased influenza virus disease severity
and lethality occurring in chronic ethanol consuming mice. Alcohol Clin Exp
Res 2010;34:1425–31.
[75] Tan Q, Duan L, Ma Y, Wu F, Huang Q, Mao K, et al. Is oseltamivir suitable for
fighting against COVID-19: In silico assessment, in vitro and retrospective
study. Bioorg Chem 2020;104:104257.
[76] Buchrieser J, Dufloo J, Hubert M, Monel B, Planas D, Rajah MM, et al. Syncytia
formation by SARS-CoV-2-infected cells. EMBO J 2020;39(23).
[77] Zhang Z, Zheng Y, Niu Z, Zhang B, Wang C, Yao X, et al. SARS-CoV-2 spike
protein dictates syncytium-mediated lymphocyte elimination. Cell Death
Differ 2021;28:2765–77.
[78] Braga L, Ali H, Secco I, Chiavacci E, Neves G, Goldhill D, et al. Drugs that inhibit
TMEM16 proteins block SARS-CoV-2 spike-induced syncytia. Nature
2021;594:88–93.
[79] Abdulamir AS, Gorial FI, Saadi SJ, Maulood MF, Hashim HA, Alnuaimi AS, et al.
A randomised controlled trial of effectiveness and safety of Niclosamide as
add on therapy to the standard of care measures in COVID-19 management.
Ann Med Surg (Lond) 2021;69:102779.
[80] Lee JS, Park S, Jeong HW, Ahn JY, Choi SJ, Lee H, et al. Immunophenotyping of
COVID-19 and influenza highlights the role of type I interferons in
development of severe COVID-19. Sci Immunol 2020;5(49).
[81] Wong CK, Lam CW, Wu AK, Ip WK, Lee NL, Chan IH, et al. Plasma
inflammatory cytokines and chemokines in severe acute respiratory
syndrome. Clin Exp Immunol 2004;136:95–103.
[82] Smits SL, de Lang A, van den Brand JMA, Leijten LM, van IJcken WF, Eijkemans
MJC, et al. Exacerbated innate host response to SARS-CoV in aged non-human
primates. PLoS Pathog 2010;6(2):e1000756.
[83] Wang S, Le TQ, Kurihara N, Chida J, Cisse Y, Yano M, et al. Influenza virus-
cytokine-protease cycle in the pathogenesis of vascular hyperpermeability in
severe influenza. J Infect Dis 2010;202:991–1001.
[84] Tang Y, Liu J, Zhang D, Xu Z, Ji J, Wen C. Cytokine storm in COVID-19: the
current evidence and treatment strategies. Front Immunol 2020;11:1708.
[85] Pasrija R, Naime M. The deregulated immune reaction and cytokines release
storm (CRS) in COVID-19 disease. Int Immunopharmacol 2021;90:107225.
[86] Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19
cytokine storm; what we know so far. Front Immunol 2020;11:1446.
[87] Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. novel coronavirus in Wuhan,
China. Lancet 2019;395(2020):497–506.
[88] 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 coronavirus disease 2019. Clin Infect Dis 2020;71:1937–42.
[89] Sebba A. Tocilizumab: the first interleukin-6-receptor inhibitor. Am J Health
Syst Pharm 2008;65(15):1413–8.
[90] 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 U S A
2020;117:10970–5.
[91] Perrone F, Piccirillo MC, Ascierto PA, Salvarani C, Parrella R, Marata AM, et al.
Tocivid-19 investigators, Tocilizumab for patients with COVID-19
pneumonia. The single-arm TOCIVID-19 prospective trial. J Transl Med
2020;18:405.
[92] Richier Q, Placais L, Lacombe K, Hermine O. COVID-19: Still a place for
tocilizumab? Rev Med Interne 2021;42:73–8.
[93] Anakinra, LiverTox: Clinical and Research Information on Drug-Induced Liver
Injury, Bethesda (MD), 2012.
[94] Huet T, Beaussier H, Voisin O, Jouveshomme S, Dauriat G, Lazareth I, et al.
Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol
2020;2(7):e393–400.
[95] Kyriakoulis KG, Kollias A, Poulakou G, Kyriakoulis IG, Trontzas IP, Charpidou
A, et al. The effect of anakinra in hospitalized patients with COVID-19: an
updated systematic review and meta-analysis. J Clin Med 2021;10:4462.
[96] Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine
on viral infections: an old drug against today’s diseases? Lancet Infect Dis
2003;3(11):722–7.
[97] Sly LM, Braun P, Woodcock BG. COVID-19: Cytokine storm modulation/
blockade with oral polyvalent immunoglobulins (PVIG, KMP01D): A potential
and safe therapeutic agent (Primum nil nocere). Int J Clin Pharmacol Ther
2020;58:678–86.
[98] Fatima U, Rizvi SSA, Fatima S, Hassan MI. Impact of hydroxychloroquine/
chloroquine in COVID-19 therapy: two sides of the coin. J Interferon Cytokine
Res 2020;40(10):469–71.
[99] Mitja O, Corbacho-Monne M, Ubals M, Tebe C, Penafiel J, Tobias A, et al. A
randomized, controlled trial. Clin Infect Dis 2019;73(2021):e4073–81.
[100] Hsu JY, Mao YC, Liu PY, Lai KL. Pharmacology and adverse events of
emergency-use authorized medication in moderate to severe COVID-19.
Pharmaceuticals (Basel) 2021;14:955.
[101] Angriman F, Ferreyro BL, Burry L, Fan E, Ferguson ND, Husain S, et al.
Interleukin-6 receptor blockade in patients with COVID-19: placing clinical
trials into context, Lancet. Respir Med 2021;9(6):655–64.
[102] Khiali S, Rezagholizadeh A, Entezari-Maleki T. A comprehensive review on
sarilumab in COVID-19. Expert Opin Biol Ther 2021;21(5):615–26.
[103] Montesarchio V, Parella R, Iommelli C, Bianco A, Manzillo E, Fraganza F, et al.
Outcomes and biomarker analyses among patients with COVID-19 treated
with interleukin 6 (IL-6) receptor antagonist sarilumab at a single institution
in Italy. J Immunother Cancer 2020;8(2):e001089.
[104] Dhimolea E. Canakinumab. MAbs 2010;2(1):3–13.
[105] Generali D, Bosio G, Malberti F, Cuzzoli A, Testa S, Romanini L, et al.
Canakinumab as treatment for COVID-19-related pneumonia: a prospective
case-control study. Int J Infect Dis 2021;104:433–40.
[106] Katia F, Myriam DP, Ucciferri C, Auricchio A, Di Nicola M, Marchioni M, et al.
Efficacy of canakinumab in mild or severe COVID-19 pneumonia. Immun
Inflamm Dis 2021;9(2):399–405.
[107] Wawrocki S, Druszczynska M, Kowalewicz-Kulbat M, Rudnicka W.
Interleukin 18 (IL-18) as a target for immune intervention. Acta Biochim
Pol 2016;63:59–63.
[108] Karki R, Sharma BR, Tuladhar S, Williams EP, Zalduondo L, Samir P, et al.
Synergism of TNF-alpha and IFN-gamma triggers inflammatory cell death,
tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock
syndromes. Cell 2021;184:149–168 e117.
[109] Luo Z, Zhu C, Ruan Z, Cui X, Shereen MA, Pan P, et al. Longitudinal
characterization of cytokine overproduction: a case report in critically ill
COVID-19 patients with hyperinflammation in bronchoalveolar lavage. Front
Med (Lausanne) 2021;8. https://doi.org/10.3389/fmed.2021.690523.
[110] Sun X, Wang T, Cai D, Hu Z, Chen J, Liao H, et al. Cytokine storm intervention
in the early stages of COVID-19 pneumonia. Cytokine Growth Factor Rev
2020;53:38–42.
[111] Russell CD, Millar JE, Baillie JK. Clinical evidence does not support
corticosteroid treatment for 2019-nCoV lung injury. Lancet 2020;395:473–5.
[112] Liongue C, O’Sullivan LA, Trengove MC, Ward AC. Evolution of JAK-STAT
pathway components: mechanisms and role in immune system
development. PLoS ONE 2012;7:e32777.
[113] Pavel MS, Schulz H, Behnke S. Object class segmentation of RGB-D video using
recurrent convolutional neural networks. Neural Netw 2017;88:105–13.
[114] Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D, et al. COVID-19:
combining antiviral and anti-inflammatory treatments. Lancet Infect Dis
2020;20:400–2.
[115] El Khouli RH, Jacobs MA. Use of MRI for personalized treatment of more
aggressive tumors. Radiology 2020;295:527–8.
[116] Cantini F, Niccoli L, Nannini C, Matarrese D, Natale MED, Lotti P, et al.
Beneficial impact of Baricitinib in COVID-19 moderate pneumonia;
multicentre study. J Infect 2020;81:647–79.
[117] 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;146:137–146 e133.
[118] Richardson P, Griffin I, Tucker C, Smith D, Oechsle O, Phelan A, et al.
Baricitinib as potential treatment for 2019-nCoV acute respiratory disease.
Lancet 2020;395(10223):e30–1.
[119] Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, et al.
Baricitinib plus Remdesivir for hospitalized adults with Covid-19. N Engl J
Med 2021;384:795–807.
[120] Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold
Spring Harb Perspect Biol 2009;1:a001651.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
835
[121] Yu H, Lin L, Zhang Z, Zhang H, Hu H. Targeting NF-kappaB pathway for the
therapy of diseases: mechanism and clinical study. Signal Transduct Target
Ther 2020;5:209.
[122] Kiron V, Park Y, Siriyappagouder P, Dahle D, Vasanth GK, Dias J, et al.
Intestinal transcriptome analysis reveals soy derivative-linked changes in
atlantic salmon. Front Immunol 2020;11:596514.
[123] DeDiego ML, Nieto-Torres JL, Regla-Nava JA, Jimenez-Guardeno JM,
Fernandez-Delgado R, Fett C, et al. Inhibition of NF-kappaB-mediated
inflammation in severe acute respiratory syndrome coronavirus-infected
mice increases survival. J Virol 2014;88:913–24.
[124] Wang W, Ye L, Ye L, Li B, Gao B, Zeng Y, et al. Up-regulation of IL-6 and TNF-
alpha induced by SARS-coronavirus spike protein in murine macrophages via
NF-kappaB pathway. Virus Res 2007;128:1–8.
[125] Roshdy WH, Rashed HA, Kandeil A, Mostafa A, Moatasim Y, Kutkat O, et al. An
immunomodulatory herbal extract with potent antiviral activity against
SARS-CoV-2. PLoS ONE 2020;15:e0241739.
[126] Masih A, Agnihotri AK, Srivastava JK, Pandey N, Bhat HR, Singh UP. Discovery
of novel pyrazole derivatives as a potent anti-inflammatory agent in
RAW264.7 cells via inhibition of NF-kB for possible benefit against SARS-
CoV-2. J Biochem Mol Toxicol 2021;35:e22656.
[127] Fan HH, Wang LQ, Liu WL, An XP, Liu ZD, He XQ, et al. in a 2019-novel
coronavirus-related coronavirus model. Chin Med J (Engl) 2019;133
(2020):1051–6.
[128] Liu Y, Chen L, Liu W, Li D, Zeng J, Tang Q, et al. Cepharanthine suppresses
herpes simplex virus type 1 replication through the downregulation of the
PI3K/Akt and p38 MAPK signaling pathways. Front Microbiol
2021;12:795756.
[129] Yang C, Zuo Q, Liu X, Zhao Q, Pu H, Gao L, et al. Small molecule screening
identified cepharanthine as an inhibitor of porcine reproductive and
respiratory syndrome virus infection in vitro by suppressing integrins/ILK/
RACK1/PKCalpha/NF-kappaB signalling axis. Vet Microbiol 2021;255:109016.
[130] Bailly C. Cepharanthine: an update of its mode of action, pharmacological
properties and medical applications. Phytomedicine 2019;62:152956.
[131] Ohashi H, Watashi K, Saso W, Shionoya K, Iwanami S, Hirokawa T, et al.
Potential anti-COVID-19 agents, cepharanthine and nelfinavir, and their
usage for combination treatment. iScience 2021;24:102367.
[132] Rogosnitzky M, Okediji P, Koman I. Cepharanthine: a review of the antiviral
potential of a Japanese-approved alopecia drug in COVID-19. Pharmacol Rep
2020;72:1509–16.
[133] Riboldi P, Gerosa M, Meroni PL. Pidotimod: a reappraisal. Int J Immunopathol
Pharmacol 2009;22(2):255–62.
[134] Carta S, Silvestri M, Rossi GA. Modulation of airway epithelial cell functions
by Pidotimod: NF-kB cytoplasmatic expression and its nuclear translocation
are associated with an increased TLR-2 expression. Ital J Pediatr 2013;39:29.
[135] Puggioni F, Alves-Correia M, Mohamed M-F, Stomeo N, Mager R, Marinoni M,
et al. Immunostimulants in respiratory diseases: focus on Pidotimod.
Multidiscip Respir Med 2019;14(1). https://doi.org/10.1186/s40248-019-
0195-2.
[136] Mahashur A, Thomas PK, Mehta P, Nivangune K, Muchhala S, Jain R.
Pidotimod: In-depth review of current evidence. Lung India 2019;36
(5):422. https://doi.org/10.4103/lungindia.lungindia_39_19.
[137] Ucciferri C, Mirko B, Vecchiet J, Falasca K. Pidotimod in paucisymptomatic
SARS-CoV2 infected patients. Mediterr J Hematol Infect Dis 2020;12(1):
e2020048. https://doi.org/10.4084/mjhid.2020.048.
[138] Iwasaki M, Saito J, Zhao H, Sakamoto A, Hirota K, Ma D. Inflammation
triggered by SARS-CoV-2 and ACE2 augment drives multiple organ failure of
severe COVID-19: molecular mechanisms and implications. Inflammation
2021;44:13–34.
[139] Arabi YM, Mandourah Y, Al-Hameed F, Sindi AA, Almekhlafi GA, Hussein MA,
et al. Saudi critical care trial, corticosteroid therapy for critically ill patients
with middle east respiratory syndrome. Am J Respir Crit Care Med
2018;197:757–67.
[140] Rice JB, White AG, Scarpati LM, Wan G, Nelson WW. Long-term systemic
corticosteroid exposure: a systematic literature review. Clin Ther
2017;39:2216–29.
[141] Li H, Chen C, Hu F, Wang J, Zhao Q, Gale RP, et al. Impact of corticosteroid
therapy on outcomes of persons with SARS-CoV-2, SARS-CoV, or MERS-CoV
infection: a systematic review and meta-analysis. Leukemia
2020;34:1503–11.
[142] Ni YN, Chen G, Sun J, Liang BM, Liang ZA. The effect of corticosteroids on
mortality of patients with influenza pneumonia: a systematic review and
meta-analysis. Crit Care 2019;23:99.
[143] Group WHOREAfC-TW, Sterne JAC, Murthy S, Diaz JV, Slutsky AS, Villar J,
Angus DC, et al. Association between administration of systemic
corticosteroids and mortality among critically ill patients with COVID-19: a
meta-analysis. JAMA 2020;324:1330–41.
[144] Gibbison B, Lopez-Lopez JA, Higgins JP, Miller T, Angelini GD, Lightman SL,
et al. Corticosteroids in septic shock: a systematic review and network meta-
analysis. Crit Care 2017;21:78.
[145] Group RC, Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, et al.
Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med
2021;384:693–704.
[146] Tomazini BM, Maia IS, Cavalcanti AB, Berwanger O, Rosa RG, Veiga VC, et al.
Effect of dexamethasone on days alive and ventilator-free in patients with
moderate or severe acute respiratory distress syndrome and COVID-19: The
CoDEX randomized clinical trial. JAMA 2020;324:1307–16.
[147] W.H. Organization, WHO welcomes preliminary results about
dexamethasone use in treating critically ill COVID-19 patients, (2020).
(https://www.who.int/zh/news/item/16-06-2020-who-welcomes-
preliminary-results-about-dexamethasone-use-in-treating-critically-ill-
covid-19-patients).
[148] Andreakos E, Papadaki M, Serhan CN. Dexamethasone, pro-resolving lipid
mediators and resolution of inflammation in COVID-19. Allergy
2021;76:626–8.
[149] Rodriguez-Garcia JL, Sanchez-Nievas G, Arevalo-Serrano J, Garcia-Gomez C,
Jimenez-Vizuete JM, Martinez-Alfaro E. Baricitinib improves respiratory
function in patients treated with corticosteroids for SARS-CoV-2
pneumonia: an observational cohort study. Rheumatology (Oxford)
2021;60(1):399–407.
[150] Rojas M, Rodríguez Y, Monsalve DM, Acosta-Ampudia Y, Camacho B, Gallo JE,
et al. Convalescent plasma in Covid-19: Possible mechanisms of action.
Autoimmun Rev 2020;19(7):102554. https://doi.org/10.1016/j.
autrev.2020.102554.
[151] Xian Y, Zhang J, Bian Z, Zhou H, Zhang Z, Lin Z, et al. Bioactive natural
compounds against human coronaviruses: a review and perspective. Acta
Pharm Sin B 2020;10(7):1163–74.
[152] Wu X, Li W, Qin Z, Xue L, Huang G, Luo Z, et al. Traditional Chinese medicine
as an adjunctive therapy for mild and common COVID-19: a systematic
review and network meta-analysis. Medicine (Baltimore) 2021;100:e27372.
[153] Luo H, Gao Y, Zou J, Zhang S, Chen H, Liu Q, et al. Reflections on treatment of
COVID-19 with traditional Chinese medicine. Chin Med 2020;15:94.
[154] Diagnosis and Treatment Protocol for Novel Coronavirus Pneumonia (Trial
Version 7), Chin Med J (Engl), 133 (2020) 1087–1095.
[155] Huang K, Zhang P, Zhang Z, Youn JY, Wang C, Zhang H, et al. Traditional
Chinese Medicine (TCM) in the treatment of COVID-19 and other viral
infections: Efficacies and mechanisms. Pharmacol Ther 2021;225:107843.
https://doi.org/10.1016/j.pharmthera.2021.107843.
[156] Runfeng L, Yunlong H, Jicheng H, Weiqi P, Qinhai M, Yongxia S, et al.
Lianhuaqingwen exerts anti-viral and anti-inflammatory activity against
novel coronavirus (SARS-CoV-2). Pharmacol Res 2020;156:104761.
[157] Li X, Yang Y, Liu L, Yang X, Zhao X, Li Y, et al. Effect of combination antiviral
therapy on hematological profiles in 151 adults hospitalized with severe
coronavirus disease 2019. Pharmacol Res 2020;160:105036.
[158] Perrotti G, Baccaglione G, Clauser T, Testarelli L, Del Fabbro M, Testori T. Total
Face Approach (TFA): a novel 3D approach to describe the main
cephalometric craniomaxillofacial parameters. Methods Protoc 2021;4:15.
[159] Liu Z, Li X, Gou C, Li L, Luo X, Zhang C, et al. Effect of Jinhua Qinggan granules
on novel coronavirus pneumonia in patients. J Tradit Chin Med
2020;40:467–72.
[160] Zhao C, Li L, Yang W, Lv W, Wang J, Guo J, et al. Chinese medicine formula
huashibaidu granule early treatment for mild COVID-19 patients: an
unblinded, cluster-randomized clinical trial. Front Med (Lausanne)
2021;8:696976.
[161] Song Y, Yao C, Yao Y, Han H, Zhao X, Yu K, et al. XueBiJing injection versus
placebo for critically ill patients with severe community-acquired
pneumonia: a randomized controlled trial. Crit Care Med 2019;47:e735–43.
[162] Wang Q, Wu X, Tong X, Zhang Z, Xu B, Zhou W. Xuebijing Ameliorates Sepsis-
Induced Lung Injury by Downregulating HMGB1 and RAGE Expressions in
Mice. Evid Based Complement Alternat Med 2015;2015:860259.
[163] Li C, Wang P, Zhang L, Li M, Lei X, Liu S, et al. Efficacy and safety of Xuebijing
injection (a Chinese patent) for sepsis: A meta-analysis of randomized
controlled trials. J Ethnopharmacol 2018;224:512–21.
[164] Ramesh S, Govindarajulu M, Parise RS, Neel L, Shankar T, Patel S, et al.
Emerging SARS-CoV-2 variants: a review of its mutations, its implications
and vaccine efficacy. Vaccines (Basel) 2021;9:1195.
[165] Bian L, Gao Q, Gao F, Wang Q, He Q, Wu X, et al. Impact of the Delta variant on
vaccine efficacy and response strategies. Expert Rev Vaccines
2021;20:1201–9.
[166] Science Brief: Omicron (B.1.1.529) Variant, CDC COVID-19 Science Briefs,
Atlanta (GA), 2020 (https://www.ncbi.nlm.nih.gov/books/NBK575856/).
[167] Thakur V, Ratho RK, OMICRON (B.1.1.529): A new SARS-CoV-2 variant of
concern mounting worldwide fear, J Med Virol, (2021) 27541 (https://
onlinelibrary.wiley.com/doi/10.1002/jmv.27541).
[168] Chen J, Wang R, Gilby NB, Wei G-W. Omicron variant (B.1.1.529): infectivity,
vaccine breakthrough, and antibody resistance. J Chem Inf Model 2022;62
(2):412–22.
[169] Dampalla CS, Zheng J, Perera KD, Wong LR, Meyerholz DK, Nguyen HN, et al.
Postinfection treatment with a protease inhibitor increases survival of mice
with a fatal SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2021;118.
[170] Wang P, Casner RG, Nair MS, Wang M, Yu J, Cerutti G, et al. Increased
resistance of SARS-CoV-2 variant P.1 to antibody neutralization. Cell Host
Microbe 2021;29:747–751 e744.
[171] Collier DA, De Marco A, Ferreira I, Meng B, Datir R, Walls AC, et al., SARS-CoV-
2 B.1.1.7 sensitivity to mRNA vaccine-elicited, convalescent and monoclonal
antibodies, medRxiv, (2021) (https://www.medrxiv.org/content/10.1101/
2021.01.19.21249840v4).
[172] Sun D, Sang Z, Kim YJ, Xiang Y, Cohen T, Belford AK, et al. Potent neutralizing
nanobodies resist convergent circulating variants of SARS-CoV-2 by targeting
diverse and conserved epitopes. Nat Commun 2021;12:4676.
[173] McCallum M, De Marco A, Lempp FA, Tortorici MA, Pinto D, Walls AC, et al. N-
terminal domain antigenic mapping reveals a site of vulnerability for SARS-
CoV-2. Cell 2021;184:2332–2347 e2316.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
836
[174] Jawabrah Al Hourani B, Swedan S, Nimer R, Alzoughool F, J.A.-A. H, E.A.T. S, R.
Alashqar, O. Al Bawareed, et al., Case Incidence and Case Fatality Ratio among
the Continents for the Period of 1 December 2020 to 15 March 2021, Genes
(Basel), 12 (2021) 1061.
[175] Luan B, Huynh T. Insights into SARS-CoV-2’s mutations for evading human
antibodies: sacrifice and survival. J Med Chem 2021. https://doi.org/10.1021/
acs.jmedchem.1c00311.
[176] Deng X, Garcia-Knight MA, Khalid MM, Servellita V, Wang C, Morris MK, et al.
Transmission, infectivity, and neutralization of a spike L452R SARS-CoV-2
variant. Cell 2021;184:3426–3437 e3428.
[177] Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM,
Ludden C, Reeve R, Rambaut A, et al. SARS-CoV-2 variants, spike mutations
and immune escape. Nat Rev Microbiol 2021;19:409–24.
[178] Starr TN, Greaney AJ, Dingens AS, Bloom JD. Complete map of SARS-CoV-2
RBD mutations that escape the monoclonal antibody LY-CoV555 and its
cocktail with LY-CoV016. Cell Rep Med 2021;2:100255.
[179] Planas D, Veyer D, Baidaliuk A, Staropoli I, Guivel-Benhassine F, Rajah MM,
et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody
neutralization. Nature 2021;596:276–80.
[180] Liu Y, Liu J, Johnson BA, Xia H, Ku Z, Schindewolf C, Widen SG, Z. An, S.C.
Weaver, V.D. Menachery, X. Xie, P.Y. Shi, Delta spike P681R mutation
enhances SARS-CoV-2 fitness over Alpha variant, bioRxiv, (2021) (https://
www.biorxiv.org/content/10.1101/2021.08.12.456173v3).
[181] Faheem BK, Kumar K, Sekhar S, Kunjiappan J, Jamalis R, Balana-Fouce BL, et al.
Druggable targets of SARS-CoV-2 and treatment opportunities for COVID-19.
Bioorg Chem 2020;104:104269.
[182] Goldhill DH, Te Velthuis AJW, Fletcher RA, Langat P, Zambon M, Lackenby A,
et al. The mechanism of resistance to favipiravir in influenza. Proc Natl Acad
Sci U S A 2018;115:11613–8.
[183] Pachetti M, Marini B, Benedetti F, Giudici F, Mauro E, Storici P, et al. Emerging
SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA
polymerase variant. J Transl Med 2020;18:179.
[184] Eskier D, Karakülah G, Suner A, Oktay Y. RdRp mutations are associated with
SARS-CoV-2 genome evolution. PeerJ 2020;8:e9587.
[185] Aiewsakun P, Nilplub P, Wongtrakoongate P, Hongeng S, Thitithanyanont A.
SARS-CoV-2 genetic variations associated with COVID-19 pathogenicity.
Microb Genom 2021;7:000734.
[186] Banerjee S, Seal S, Dey R, Mondal KK, Bhattacharjee P. Mutational spectra of
SARS-CoV-2 orf1ab polyprotein and signature mutations in the United States
of America. J Med Virol 2021;93:1428–35.
[187] Hossain MU, Bhattacharjee A, Emon MTH, Chowdhury ZM, Ahammad I,
Mosaib MG, et al. Novel mutations in NSP-1 and PLPro of SARS-CoV-2 NIB-
1 genome mount for effective therapeutics. J Genet Eng Biotechnol
2021;19:52.
[188] Awadasseid A, Wu Y, Tanaka Y, Zhang W. Effective drugs used to combat
SARS-CoV-2 infection and the current status of vaccines. Biomed
Pharmacother 2021;137:111330.
[189] Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, et al. Antibody Resistance of
SARS-CoV-2 Variants B.1.351 and B.1.1.7. Nature 2021;593:130–5.
[190] Majumdar P, Niyogi S. SARS-CoV-2 mutations: the biological trackway
towards viral fitness. Epidemiol Infect 2021;149:e110.
[191] Han N, Hwang W, Tzelepis K, Schmerer P, Yankova E, MacMahon M,
et al. Identification of SARS-CoV-2-induced pathways reveals drug
repurposing strategies. Sci Adv 2021;7(27). https://doi.org/10.1126/
sciadv.abh3032.
[192] Yang Y, Islam MS, Wang J, Li Y, Chen X. Traditional Chinese Medicine
in the Treatment of Patients Infected with 2019-New Coronavirus
(SARS-CoV-2): A Review and Perspective. Int J Biol Sci 2020;16
(10):1708–17.
[193] Castro C, Arnold JJ, Cameron CE. Incorporation fidelity of the viral RNA-
dependent RNA polymerase: a kinetic, thermodynamic and structural
perspective. Virus Res 2005;107:141–9.
[194] Tian W, Zhang N, Jin R, Feng Y, Wang S, Gao S, et al. Immune suppression in
the early stage of COVID-19 disease. Nat Commun 2020;11:5859.
[195] 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;63:364–74.
[196] Xie J, Ding C, Li J, Wang Y, Guo H, Lu Z, et al. Characteristics of patients with
coronavirus disease (COVID-19) confirmed using an IgM-IgG antibody test. J
Med Virol 2020;92(10):2004–10.
[197] Ucciferri C, Vecchiet J, Falasca K. Role of monoclonal antibody drugs in the
treatment of COVID-19. World J Clin Cases 2020;8(19):4280–5.
[198] Pham T, Brochard LJ, Slutsky AS. Mechanical ventilation: state of the art.
Mayo Clin Proc 2017;92(9):1382–400.
J. Yin, C. Li, C. Ye et al. Computational and Structural Biotechnology Journal 20 (2022) 824–837
837
...  Drug-Drug Interactions: High bioavailability may increase the potential for drugdrug interactions [88]. When multiple drugs are co-administered, their pharmacokinetics and pharmacodynamics may be affected, leading to unpredictable outcomes. ...
Chapter
Full-text available
This book series invites all the Specialists, Professors, Doctors, Scientists, Academicians, Healthcare professionals, Nurses, Students, Researchers, Business Delegates, and Industrialists across the globe to publish their insights and convey recent developments in the field of Nursing, Pharmaceutical Research and Innovations in Pharma Industry. Book series on Pharmacy and Nursing covers research work in a set of clinical sciences and medicine.
...  Drug-Drug Interactions: High bioavailability may increase the potential for drugdrug interactions [88]. When multiple drugs are co-administered, their pharmacokinetics and pharmacodynamics may be affected, leading to unpredictable outcomes. ...
Chapter
Bioavailability refers to the fraction of administered drug that reaches the systemic circulation in an unchanged form and is available to exert its therapeutic effects.
...  It was discovered that brains were smaller after developing COVID. The normal aging process causes us to lose between 0.2 and 0.3% of our grey matter annually (Yin et al, 2022). The study found that all subjects with COVID, regardless of how mild their symptoms, lost anywhere from 0.2% to 2.0% in their olfactory cortex during scans and had disrupted connections between multiple brain regions when compared to uninfected individuals (Huang, 2022). ...
Chapter
Full-text available
The COVID-19 pandemic has caused significant challenges, -2 infection (PASC).Physiotherapists have been instrumental in informing the public and medical professionals about the value of exercise, pulmonary exercises, and methods of rehabilitation to improve respiratory function in COVID-19 patients. In order to ensure continuity of care and reduce the danger of virus transmission, telehealth solutions have emerged as useful tools for physiotherapists. These platforms allow them to conduct remote consultations and provide patients with instructions for performing exercises at home. Physiotherapists have been essential components of interdisciplinary teams in acute care settings, helping to optimize airway health, mobility, and general functional outcomes for COVID-19 patients. Furthermore, the identification of PASC has brought attention to the enlarged role that physiotherapy plays in the management of residual symptoms in patients who are recuperating from the acute stage of the illness, including exhaustion, dyspnea, and musculoskeletal problems. In light of the COVID-19 pandemic, this abstract highlights how physiotherapy is changing and emphasizes its critical role in improving health, reducing complications, and assisting in the rehabilitation of those afflicted by the virus. Physiotherapists continue to play a critical role in modifying their practices to meet the changing healthcare demands brought on by the COVID-19 pandemic as our understanding of the virus continues to grow.
... [7,8] Key targets for therapies against SARS-CoV-2 are structural and functional proteins which play important roles in viral transcription and replication, such as the main protease or 3 C-like protease (Mpro or 3CLpro), papain-like protease (PLpro), non-structural protein 12 (nsp12), and RNAdependent RNA polymerase (RdRP). [9,10,11,12] Mpro is an attractive drug target for the development of general inhibitors of SARSlike coronaviruses, due to its importance in the viral life cycle. It also has high conservation among different SARS-type coronaviruses: with 96 % sequence identity among SARS-CoV, MERS, and SARS-CoV-2 main proteases. ...
Article
Full-text available
Coronaviruses are continually evolving, as evidenced by the emergence of three closely related pathogenic variants over the past two decades: SARS (2002), MERS (2012), and SARS‐CoV‐2 (2019); and the appearance of many new SARS‐CoV‐2 strains during the COVID‐19 pandemic. Hence, COVID‐19 has now become a pandemic disease; with a real possibility for the emergence of new deadly SARS‐related coronaviruses. The SARS‐CoV‐2 main protease, Mpro is an attractive drug target for the development of inhibitors against SARS type and other coronaviruses because it is essential for the viral life cycle and highly conserved among a wide range of pathogenic coronaviruses. This motivated the synthesis of novel inhibitors of Mpro, which could be the basis for COVID‐19 drug development. The synthesis of new 5,6 : 16,17‐diseco and 6,7 : 16,17‐disecoandrostane 16,17a‐dinitriles is reported. In silico ADMET analyses suggest that the synthesized compounds possess drug‐like properties with no predicted toxicities. Structural similarity analyses indicates that synthesized B,D‐diseco 16,17a‐dinitrile derivatives have potential as protease inhibitors. Molecular docking simulations suggest that B,D‐diseco 16,17a‐dinitriles could inhibit substrate binding by Mpro. The present study suggests that newly synthesized androstane 5,6 : 16,17‐diseco‐5‐keto‐6‐oic acid and 6,7 : 16,17‐diseco‐6,7‐dioic acid 16,17a‐dinitriles 8 and 11 could represent a starting point for the design of Mpro inhibitors against SARS type coronaviruses.
... Methods for preventing SARS-CoV-2 developing mutations[63]. ...
Article
Full-text available
The ongoing COVID-19 pandemic has seen the evolution of the SARS-CoV-2 virus, resulting in the emergence of various concerning variants with unique biological characteristics. As the pandemic continues, it will be crucial to promptly evaluate the potential of any new variant to cause severe illness. The severity of the latest Omicron sublineages, including BA.5, XBB, BQ.1.18, BA.2, BA.2.75, and EG.5.1, is currently under assessment. This system provides valuable and essential information for rapidly assessing the threat posed by new versions of the virus.
Article
Drug discovery involving covalent drugs has a rich history dating back to the 20th century. Despite their longevity, concerns about safety have relegated covalent drug development to the periphery of research focus. The emergence of the groundbreaking concept of Targeted Covalent Inhibition (TCI) has catalyzed a renewed emphasis on covalent medications, elevating their status within the field. This review delves into the landscape of covalent inhibitors, comprehensively examining those developed to date and those currently under investigation. Covalent inhibitors are categorized based on their target proteins, simplifying the understanding of their diverse applications. Notable targets include disease-related proteins such as EGFR, BTK, and SARS-COV-2, signifying the broad therapeutic potential of covalent drugs across various medical domains. One of the most significant aspects discussed is the potential of covalent drugs to overcome the drawbacks associated with conventional therapy, contingent upon the effective consideration of toxicity-related factors. These drugs present a range of advantages, including heightened selectivity, the ability to administer lower doses, and the degradation of previously deemed undruggable proteins. The trajectory of covalent drugs appears promising for drug discovery. By targeting different proteins, these compounds hold the potential to provide effective treatment options in the years to come. The comprehensive exploration of covalent drugs in this review not only contributes to our understanding of their historical evolution and contemporary applications but also underscores their importance as a focal point for advancing therapeutic options and addressing the limitations of current treatment modalities.
Article
Full-text available
The emergence of SARS-CoV-2 variants diminished the efficacy of current antiviral drugs and vaccines. Hence, identifying highly conserved sequences and potentially druggable pockets for drug development was a promising strategy against SARS-CoV-2 variants. In viral infection, the receptor-binding domain (RBD) proteins are essential in binding to the host receptor. Others, Heparan sulfate (HS), widely distributed on the surface of host cells, is thought to play a central role in the viral infection cycle of SARS-CoV-2. Therefore, it might be a reasonable strategy for antiviral drug design to interfere with the RBD in the HS binding site. In this study, we used computational approaches to analyze multiple sequences of coronaviruses and reveal important information about the binding of HS to RBD in the SARS-CoV-2 spike protein. Our results showed that the potential hot-spots, including R454 and E471, in RBD, exhibited strong interactions in the HS-RBD binding region. Therefore, we screened different compounds in the natural product database towards these hot-spots to find potential antiviral candidates using LibDock, Autodock vina and furthermore applying the MD simulation in AMBER20. The results showed three potential natural compounds, including Acetoside (ACE), Hyperoside (HYP), and Isoquercitrin (ISO), had a strong affinity to the RBD. Our results demonstrate a feasible approach to identify potential antiviral agents by evaluating the binding interaction between viral glycoproteins and host receptors. The present study provided the applications of the structure-based computational approach for designing and developing of new antiviral drugs against SARS-CoV-2 variants.
Article
Objectives: The purpose of this study was to evaluate the effectiveness and safety of traditional herbal medicine as a stand-alone treatment group through major English databases due to the lack of RCTs in Korea, and to provide a review of the herbal interventions used.Methods: Using four databases (Pubmed, EMBASE, OASIS, RISS), combination of words such as “Coronavirus” “RCT” “Herb” “Decoction” “TCM” were used. RCTs using herbal medicines to treat coronavirus were searched. Final 4 studies were selected by two authors according to inclusion and exclusion criteria.Results: A total of 1,435 patients were studied. The Chinese herbs used in the treatment group were Shengmai Yin, JingYinGuBiao granules, Jinhua Qinggan granules, and Bufei Huoxue capsules. The intervention group showed greater attenuation of pneumonia lesions on CT. Also, improvement in 6-min walk distance (6MWD), and negative conversion rate in treatment group were reported. Furthermore, scores on the Fatigue Assessment Inventory (FAI) were lower in the herbal group than in the placebo group. The median time to recovery of COVID-19 related symptoms was shorter in TCM group compared to the control group. Reported adverse effects were diarrhea, liver dysfunction, and excessive menstruation, and two papers did not mention side effects in detail.Conclusion: Herbal medicine alone can increase the conversion rate of viral negativity and relieve COVID-19 related symptoms without significant adverse effects.
Article
Full-text available
Cepharanthine (CEP) is a naturally occurring isoquinoline alkaloid extracted from Stephania cepharantha Hayata. Although its underlying molecular mechanism is not fully understood, this compound is reported as a promising antiviral drug. In the present study, we explore the anti-HSV-1 effects and the underlying molecular mechanisms of CEP in vitro. Our results show that CEP could significantly inhibit the formation of plaque and the expression of viral proteins and exhibit a general suppression of replication-associated genes. Whereas HSV-1 infection increases the expressions of phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), and p38 mitogen-activated protein kinase (p38 MAPK) in host cells, CEP was effective indirectly inhibiting phosphorylation levels of the targets in PI3K/Akt and p38 MAPK signaling pathways. Moreover, CEP markedly decreased G0/G1 phase and increased G2/M phase cells and decreased the expression of cyclin-dependent kinase1 (CDK1) and cyclinB1 in a dose-dependent manner. Additionally, CEP increased apoptosis in infected cells, reduced B cell lymphoma-2 (Bcl-2) protein levels, and increased the protein levels of Bcl-associated X protein (Bax), cleaved-caspase3, and nuclear IκB kinaseα (IκBα). Collectively, CEP could arrest the cell cycle in the G2/M phase and induce apoptosis in infected cells by inhibiting the PI3K/Akt and p38 MAPK signaling pathways, hence further reducing HSV-1 infection and subsequent reproduction.
Article
Full-text available
In this study, we performed genome-wide association analyses on SARS-CoV-2 genomes to identify genetic mutations associated with pre-symptomatic/asymptomatic COVID-19 cases. Various potential covariates and confounding factors of COVID-19 severity, including patient age, gender and country, as well as virus phylogenetic relatedness were adjusted for. In total, 3021 full-length genomes of SARS-CoV-2 generated from original clinical samples and whose patient status could be determined conclusively as either ‘pre-symptomatic/asymptomatic’ or ‘symptomatic’ were retrieved from the GISAID database. We found that the mutation 11 083G>T, located in the coding region of non-structural protein 6, is significantly associated with asymptomatic COVID-19. Patient age is positively correlated with symptomatic infection, while gender is not significantly correlated with the development of the disease. We also found that the effects of the mutation, patient age and gender do not vary significantly among countries, although each country appears to have varying baseline chances of COVID-19 symptom development.
Article
Full-text available
Coronavirus disease 2019 (COVID-19), a highly infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected more than 235 million individuals and led to more than 4.8 million deaths worldwide as of October 5 2021. Cryo-electron microscopy and topology show that the SARS-CoV-2 genome encodes lots of highly glycosylated proteins, such as spike ( S ), envelope ( E ), membrane ( M ), and ORF3a proteins, which are responsible for host recognition, penetration, binding, recycling and pathogenesis. Here we reviewed the detections, substrates, biological functions of the glycosylation in SARS-CoV-2 proteins as well as the human receptor ACE2, and also summarized the approved and undergoing SARS-CoV-2 therapeutics associated with glycosylation. This review may not only broad the understanding of viral glycobiology, but also provide key clues for the development of new preventive and therapeutic methodologies against SARS-CoV-2 and its variants.
Article
Full-text available
Background: Tocilizumab blocks pro-inflammatory activity of interleukin-6 (IL-6), involved in pathogenesis of pneumonia the most frequent cause of death in COVID-19 patients. Methods: A multicenter, single-arm, hypothesis-driven trial was planned, according to a phase 2 design, to study the effect of tocilizumab on lethality rates at 14 and 30 days (co-primary endpoints, a priori expected rates being 20 and 35%, respectively). A further prospective cohort of patients, consecutively enrolled after the first cohort was accomplished, was used as a secondary validation dataset. The two cohorts were evaluated jointly in an exploratory multivariable logistic regression model to assess prognostic variables on survival. Results: In the primary intention-to-treat (ITT) phase 2 population, 180/301 (59.8%) subjects received tocilizumab, and 67 deaths were observed overall. Lethality rates were equal to 18.4% (97.5% CI: 13.6–24.0, P = 0.52) and 22.4% (97.5% CI: 17.2–28.3, P < 0.001) at 14 and 30 days, respectively. Lethality rates were lower in the validation dataset, that included 920 patients. No signal of specific drug toxicity was reported. In the exploratory multivariable logistic regression analysis, older age and lower PaO2/FiO2 ratio negatively affected survival, while the concurrent use of steroids was associated with greater survival. A statistically significant interaction was found between tocilizumab and respiratory support, suggesting that tocilizumab might be more effective in patients not requiring mechanical respiratory support at baseline. Conclusions: Tocilizumab reduced lethality rate at 30 days compared with null hypothesis, without significant toxicity. Possibly, this effect could be limited to patients not requiring mechanical respiratory support at baseline.
Article
Full-text available
The widespread increase in multiple severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants is causing a significant health concern in the United States and worldwide. These variants exhibit increased transmissibility, cause more severe disease, exhibit evasive immune properties, impair neutralization by antibodies from vaccinated individuals or convalescence sera, and reinfection. The Centers for Disease Control and Prevention (CDC) has classified SARS-CoV-2 variants into variants of interest, variants of concern, and variants of high consequence. Currently, four variants of concern (B.1.1.7, B.1.351, P.1, and B.1.617.2) and several variants of interests (B.1.526, B.1.525, and P.2) are characterized and are essential for close monitoring. In this review, we discuss the different SARS-CoV-2 variants, emphasizing variants of concern circulating the world and highlight the various mutations and how these mutations affect the characteristics of the virus. In addition, we discuss the most common vaccines and the various studies concerning the efficacy of these vaccines against different variants of concern.
Article
Full-text available
Background Coronavirus disease 2019 (COVID-19) in many countries is still very serious. At present, there is no specific and effective drug for this disease. Traditional Chinese medicine (TCM) has played a great role in fighting against COVID-19. However, their effectiveness and safety are still obscure and deserve further investigation. The aim of the study was to evaluate the efficacy and safety of TCM assisted in conventional treatment in the treatment of mild and common COVID-19. Methods PubMed, EMbase, MEDLINE, China National Knowledge Infrastructure Database, WANFANG DATA, and VIP Chinese Science and Technology Periodical Database were searched for randomized controlled trials (RCTs) and non-randomized controlled trials of TCM assisted in conventional treatment. The RCT research quality was evaluated by Cochrane 5.1.0 bias risk scale and the non-randomized controlled trial research quality was evaluated by Newcastle Ottawa scale, and the statistical analysis was conducted by Revman 5.3 and R software. The bias and sensitivity of the statistical results were analyzed by STATA 14.0. Registration number: CRD42020210619. Results Fifteen studies were included with 7 RCT studies and 8 retrospective cohort studies, involving a total of 1623 patients. Compared with the control group, TCM can improve the main index clinical effective rate (odds ratio [OR] = 2.64, 95% Confidence interval (CI) [1.94,3.59], P < .00001). The results of Begg test (Pr > z = 0.266) and sensitivity analysis showed that the results were relatively stable. Toujie Quwen (OR = 4.9, 95%CI [1.9,14.0]), Shufeng Jiedu (OR = 2.9, 95%CI [1.5,5.7]), and Lianhua Qingwen (OR = 2.4, 95%CI [1.6,3.6]) were with the best. It can also improve the main clinical symptoms (fever, cough, fatigue, and the regression time of the 3 symptoms), severe conversion rate, and computed tomography improvement rate. Its safety was not significantly compared with conventional treatment. However, in terms of safety of a single TCM, Shufeng Jiedu (OR = –0.86, 95%CI [–1.89,0.09]) and Lianhua Qingwen (OR = –0.49, 95%CI[–0.94,–0.05]) were lower than those of conventional treatment. Conclusion TCM as an adjuvant therapy combined with conventional treatment has good curative effect on mild and common type of COVID-19 patients. Its advantages lie in clinical efficacy and improvement of symptom group, and can prevent patients from transforming to severe disease. In terms of clinical efficacy and safety, Shufeng Jiedu and Lianhua Qingwen have obvious advantages, which are worthy of clinical promotion.
Article
Full-text available
We have studied the non-covalent interaction between PF-07321332 and SARS-CoV2 main protease at the atomic level using a computational approach based on extensive molecular dynamics simulations with explicit solvent. PF-07321332, whose chemical structure has been recently disclosed, is a promising oral antiviral clinical candidate with well-established anti-SARS-CoV-2 activity in vitro. The drug, currently in phase III clinical trials in combination with ritonavir, relies on the electrophilic attack of a nitrile warhead to the catalytic cysteine of the protease. Nonbonded interaction between the inhibitor and the residues of the binding pocket, as well as with water molecules on the protein surface, have been characterized using two different force fields and the two possible protonation states of the main protease catalytic dyad HIS41-CYS145. When the catalytic dyad is in the neutral state, the non-covalent binding is likely to be stronger. Molecular dynamics simulations seems to lend support for an inhibitory mechanism in two steps: a first non-covalent addition with the dyad in neutral form and then the formation of the thiolate-imidazolium ion pair and the ligand relocation for finalising the electrophilic attack.
Article
The latest severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant Omicron (B.1.1.529) has ushered panic responses around the world due to its contagious and vaccine escape mutations. The essential infectivity and antibody resistance of the SARS-CoV-2 variant are determined by its mutations on the spike (S) protein receptor-binding domain (RBD). However, a complete experimental evaluation of Omicron might take weeks or even months. Here, we present a comprehensive quantitative analysis of Omicron's infectivity, vaccine breakthrough, and antibody resistance. An artificial intelligence (AI) model, which has been trained with tens of thousands of experimental data and extensively validated by experimental results on SARS-CoV-2, reveals that Omicron may be over 10 times more contagious than the original virus or about 2.8 times as infectious as the Delta variant. On the basis of 185 three-dimensional (3D) structures of antibody-RBD complexes, we unveil that Omicron may have an 88% likelihood to escape current vaccines. The U.S. Food and Drug Administration (FDA)-approved monoclonal antibodies (mAbs) from Eli Lilly may be seriously compromised. Omicron may also diminish the efficacy of mAbs from AstraZeneca, Regeneron mAb cocktail, Celltrion, and Rockefeller University. However, its impacts on GlaxoSmithKline's sotrovimab appear to be mild. Our work calls for new strategies to develop the next generation mutation-proof SARS-CoV-2 vaccines and antibodies.
Article
Recent emergence of the SARS-CoV-2 variant as OMICRON has become a global concern. This short note highlights the identification and global spread of OMICRON which has occupied more than 77 nations by now, which resulted in many hypotheses about its origin and degree of infectivity. The detection of mutations in the RBD region of Spike protein is a concern by passing vaccine immunity. The ahead will speak about its transmission potentiality, infectivity, disease morbidity or mortality as well as its effect on COVID-19 vaccines. This article is protected by copyright. All rights reserved.