Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2021 Oct 25;193:1165–1200. doi: 10.1016/j.ijbiomac.2021.10.144

COVID-19: A review of newly formed viral clades, pathophysiology, therapeutic strategies and current vaccination tasks

Chandran Murugan a, Sharmiladevi Ramamoorthy b, Guruprasad Kuppuswamy b, Rajesh Kumar Murugan b, Yuvaraj Sivalingam b, Anandhakumar Sundaramurthy a,c,
PMCID: PMC8545698  PMID: 34710479

Abstract

Today, the world population is facing an existential threat by an invisible enemy known as severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) or COVID-19. It is highly contagious and has infected a larger fraction of human population across the globe on various routes of transmission. The detailed knowledge of the SARS-CoV-2 structure and clinical aspects offers an important insight into the evolution of infection, disease progression and helps in executing the different therapies effectively. Herein, we have discussed in detail about the genome structure of SARS-CoV-2 and its role in the proteomic rational spread of different muted species and pathogenesis in infecting the host cells. The mechanisms behind the viral outbreak and its immune response, the availability of existing diagnostics techniques, the treatment efficacy of repurposed drugs and the emerging vaccine trials for the SARS-CoV-2 outbreak also have been highlighted. Furthermore, the possible antiviral effects of various herbal products and their extracted molecules in inhibiting SARS-CoV-2 replication and cellular entry are also reported. Finally, we conclude our opinion on current challenges involved in the drug development, bulk production of drug/vaccines and their storage requirements, logistical procedures and limitations related to dosage trials for larger population.

Keywords: SARS-CoV-2, Spike protein mutation, Variants, Phytocomponent, Vaccines, And drugs

1. Introduction

Coronavirus (CoV) is a major pathogen predominantly affecting the human respiratory system [1]. The Latin etymology of the term corona hails from its crown form [2]. The CoVs belong to the coronavirinae subfamily and are categorized into four genera based on genomic architecture and evolutionary relationships: (i) alphacoronavirus (αCoV), (ii) betacoronavirus (βCoV), (iii) gammacoronavirus (γCoV) and (iv) deltacoronavirus (δCoV) [3]. Among the six human-infecting CoVs, 229E and NL63 come under the α-CoV genus, whereas, HKU1, OC43, Severe Acute Respiratory Syndrome-related CoV (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) are classified under the β-CoV genus. Each genus is then subdivided into 4 lineage subgroups: A, B, C, and D [4]. In the end of 2019, many patients had been severely affected with pneumonia by the causative pathogen known as SARS-CoV-2 in Wuhan, China. Upon this taxonomy background, phylogeny, and existing practice, the CoV Research Group of the International Committee on Virus Taxonomy officially announced that this virus is related to extreme SARS-CoV and called it as ‘SARS-CoV-2’, which falls under the B type lineage [5]. The spherical/pleomorphic-shaped SARS-CoV-2 is about 80–120 nm in size and is classified under the order of Nidovirales. The coiled nucleocapsid of SARS-CoV-2 is formed by complexing a single-stranded positive-sense genomic RNA (+ssRNA) with a protective helical nucleocapsid (N) protein shell. Further, it is encased by three fundamental proteins: spike (S), matrix (M) membrane and a highly hydrophobic small envelope (E) protein [6], as shown in Fig. 1 . Most of the CoVs family cause pathogenic enzootic infections that affect humans, birds and many animals, and often chronically instigate enteric, respiratory, and neuronal diseases [7], [8], [9], [10], [11]. The World Health Organization (WHO) has announced SARS-CoV-2 as most highly pathogenic human CoVs when compared to SARS-CoV and MERS-CoV [12]. By 17th October 2021, the total number of infected cases of SARS-CoV-2 worldwide was reported to be as 241,170,384 including 4,910,066 deaths, in major countries such as the United State of America, India, Brazil, United Kingdom, Russia, Turkey, France, Iran, Argentina, and Spain. It has spread to >216 countries worldwide and declared as ‘pandemic' [13], as shown in Fig. 2 . The major symptoms of SARS-CoV-2 infection include cough, loss of appetite, high fever, headache, breath shortness, vomiting, dyspnea, sore throat, rhinorrhea, diarrhea and abdominal pain. The infected patients also displayed increased numbers in leukocytes, plasma pro-inflammatory cytokines and abnormal respiratory findings [14], [15], [16]. A communicable disease, SARS-CoV-2 is spreading from human to human through two main routes: virion-laden in respiratory fluid droplets (less than 1–2000 μm) arising from infected coughs or sneezes, and also from coming in contact with virus-contaminated surfaces (e.g. skin-to-skin and touching infected objects and surfaces). The infection is likely to spread in crowds and in areas where people are within 1–2 m, [17], [18], [19] as shown in Fig. 3 . Another possible transmission mode is through infected fecal matter due to the virus's ability to survive in stool samples for 4 days [20]. The disease severity in patients with SARS-CoV-2 is based on chronic conditions, including coronary heart disease, diabetes and hypertension [21]. The incubation time duration is estimated to be between 4 and 14 days before the onset of the disease. Recent studies suggest that certain patients with SARS-CoV-2 display damage to other organs such as the brain (encephalitis), kidney, heart and eye (conjunctivitis) [22]. The SARS-CoV-2 can live for extended periods on the exterior of sterile sponges, aluminum and sterile surgical gloves, which increases the risk of transmission through contact. These components can help as a defense matrix for the SARS-CoV-2 virions, making therapies for sanitization inefficient or exploited for enhanced inactivation [23], [24]. The enveloped virus of SARS-CoV-2 inserts its nucleic acid into host epithelial cells and keeps its lipid bilayer intact throughout pathways for its infectivity. Besides, the enveloped virus has a dynamic protein capsid that satisfactorily confines the elastically-strained genomic coil, sustains osmotic pressure in the surroundings and readily disassemble to release their genome into the host cell [25]. Physical therapies such as desiccation, heating, UV irradiation and chemical decontamination with different agents (e.g. alcohols, oxidants, acids and few specialized surfactants) can induce the rupture in SARS-CoV-2 envelope and capsid. They can be highly successful in lowering or even stopping the virus's propagation and transmission [26], [27], [28], [29], [30]. It is worth noting that the severity of pathogenic SARS-CoV-2 was profoundly underestimated by society at the outset of the viral outbreak. As a result, SARS-CoV-2 has caused tremendous social harm, brought global fear, medium mortality rates and high transmission than other deadly viral outbreaks. Hence, the current state of affairs may be different, and have shown modified methodologies to deal with these kind of viruses. By using various molecular medicine based tools and techniques, the research communities from various countries have developed host-based and virus-based targets/vaccines to fight SARS-CoV-2 infections. In this review, we have comprehensively summarized the physiochemical features, mutation, clades, and infectivity of SARS-CoV-2. Further, the key performances of the innate immune system towards the viral infection, possible therapeutic targets with activity rely on host/virus and diagnostics methods have been discussed in detail. The recent progress in the development of drugs, approved vaccines in multiple phases of clinical trials across the countries and possibilities of herbal production have also been highlighted. The discussion on vaccine hesitancy, list of approved vaccines and global vaccine distribution plans (Covax) has been incorporated into this review to provide a complete information about the SARS-CoV-2.

Fig. 1.

Fig. 1

Diagrammatic depictions reveal the SARS-CoV-2 virion structure and morphology.

Fig. 2.

Fig. 2

Statistical reports of the SARS-CoV-2 pandemic. (A) Worldwide report of SARS-CoV-2 infection and (B) topmost leading countries with SARS-CoV-2 infection as on October 17, 2021.

Fig. 3.

Fig. 3

Key routes of human-human transmission of SARS-CoV-2.

2. Genome and structure of SARS-CoV-2

2.1. SARS-CoV-2 genome structure

A SARS-CoV-2 genome +ssRNA composed of 29,891 nucleotides, 11 protein-coding genes, 38% of the guanine-cytosine (GC) content and encodes 9860 amino acids (aa). The sequenced SARS-CoV-2 has ~ 95% of the same genomic structure as SARS-CoV, identified in a cave from Yunnan Province, China, in 2013 [31]. The 5′-3′ gene lineup noted as 5′-replicase (open reading frame (ORF1/ab))-structural proteins [S, E, M and N] −3′ untranslated regions (UTRs) poly (A) tail, is shown in Fig. 4 . The length of 3′ and 5′-UTRs contains 358 and 265 nucleotides, respectively [32]. At the 3′ end, the six accessory genes (3a, 6, 7a, 7b, 8, and 9a) are scattered among the structural genes, in which some genes play a vital role in SARS-CoV-2 pathogenesis. At 5′ terminal, a region of approximately 20 kb coincides with two ORFs (ORF1a and ORF1b), in which ORF1a formulates a huge polyprotein (pp1ab, about 790 kDa). Then, two proteases such as 3C-like protease or main protease (3CLpro) and papain-like protease (PLpro) cleave the pp1ab and playing a key role in maturing them into non-structural proteins (nsps). The dimeric 3CLpro consists of two parts, namely intracellular N-terminal catalytic and the extracellular C-terminal domains in each of their monomers. The similarity of 3CLpro sequence between SARS-CoV and newly emerged SARS-CoV-2 was noticed to be 96%. However, the minimal differences noted were only observed on their surface proteins [33], [34], [35], [36]. PLpro enzymes consist of 4 distinct domains: a single SARS-unique (SUD), an ADP-ribose-1d-phosphatase (ADRP), a transmembrane (TM), and a fold-like ubiquitin (UB1) domain [37]. The genomic RNA consists of a well-organized ribosomal frameshift signal, in which both heptanucleotide “slip site” (slippery sequence) and RNA pseudoknot structure are arranged one after another. The slippery sequence occasionally causes a frameshift at the end of ORF1a and consecutively translate them into a joint ORF1a and 1b polyprotein [38], [39]. Notably, SARS-CoV-2 pp1ab encompasses 14 exact proteolytic cleavage sites for PLpro and 3CLpro. While PLpro can cleave the N-terminus of amino acid (aa) sequences at three sites, namely 181–182, 818–819 and 2763–2764 aa, 3CLpro cleaves C-terminus of aa sequence at 11 different sites. After completion of cleavage process, the formed 15 nsps play a pivotal role in host cell infection and RNA synthesis. Besides viral proteins, the disparate cellular proteins of the host such as poly(A)-binding protein, mitochondrial aconitase, heterogeneous nuclear ribonucleoprotein A1 and polypyrimidine-track-binding (PTB) protein are also interacting with the critical cis-acting elements of SARS-CoV-2 replication [40], [41].

Fig. 4.

Fig. 4

Genomic distribution profile of SARS-CoV and SARS-CoV-2. (Adapted and reprinted with permission from ref. [34]), copyrights © 2020 Cell press.

2.2. Structural and accessory proteins of SARS-CoV-2

The SARS-CoV-2 structural proteins, named as S, E, M, and N proteins are produced by ORF-2, ORF-4, ORF-5 and ORF-9, respectively (Table 1 ) [62]. These structural proteins share a huge sequence closeness to the same protein that present in SARS-CoV and MERS-CoV [63]. The exterior of the SARS-CoV-2 virion is guarded by the trimeric S glycoprotein (180/90-kDa) shaped as club-like projections. It has a diverging sequence with similarity of <75% to other SARS-related CoVs [64]. S protein is frequently formed by two subunits S1 and S2, which are crucial for viral entry as they bind to angiotensin-converting enzyme 2 (ACE2) receptors on the host cell plasma membrane surface [65]. The S1 subunit receptor-binding domains (RBD) undergoes transient hinge-like conformational motions (receptor-inaccessible states/receptor-accessible) with the host-cell ACE2 receptor [66], [67]. The attraction between the S protein RBD (394 glutamine residues) and ACE2 receptor (31 lysine residues) creates a “Van der Waals” forces that reinforce a binding between viral-host cell [68], [69], [70].

Table 1.

The functions of structural and assessory proteins in SARS-CoV-2.

Protein Genes coding Functions References
Spike (S) protein ORF-2 Viral entry, binding with host receptor and membrane fusion, main component for giving the virus crown like structure. [42], [43], [44], [45]
Envelope (E) protein ORF-6 Virion entry, viral mutation and viral production [46]
Membrane (M) glycoprotein ORF-7 Leaving a short NH2 and a long COOH terminus on outside and indoor of virus (cytoplasmic domain), attach with other structural proteins and determine the shape of viral envelope. [47], [48]
Nucleocapsid (N) protein ORF-8a Viral assembly, viral replication cycle and viral-host cellular infection [49], [50]
pp1a ORF-1a Express the virus exploit a slippery sequence as 5′-UUUAAAC-3′, contain the nsps 1–11 [51]
pp1ab ORF-1b Express the virus exploit a slippery sequence as 5′-UUUAAAC-3′, contain the nsps 1–16 [51]
7a ORF-7a Increases the expression of JNK, NF-κβ, and p38 MAP kinase. It involves in cell cycle arrest, inhibits host translation and stimulate apoptosis. [52]
3a ORF-3a Increases the expression of JNK, IL-8, NF-κβ, and RANTES. It involves in cell cycle arrest, ion-channel activity and stimulate apoptosis. [52]
3b ORF-3b Stimulate type-I IFN production, and prevents cell signaling, cell cycle arrest and induces apoptosis. [52]
Region of nsp1coding nsp1 Antiviral host response and trigger mRNA degradation [50], [51]
Unknown nsp2 Unknown function and often bind to prohibiting proteins [51]
Papain-like protease nsp3 Involves in viral polyprotein cleavage. It Interacts with N protein and can block the host immune response. [52]
Transmembrane domain nsp4 DMV formation and combines with nsp3 and nsp6. It also involves in proliferation of cellular membrane and more important for DMVs structure. [52], [53]
3CLpro nsp5 Involves in viral polyprotein cleavage. [53]
Transmembrane domain nsp6 DMV formation and complex with nsp3 and nsp4. [54]
Unknown nsp7 Primase, hexadecameric complex with nsp8 [52], [55]
Primase nsp8 Primase activity, hexadecameric complex with nsp7 [55], [56]
Unknown nsp9 Activity of RNA/DNA binding [57]
Unknown nsp10 complex with nsp14, proliferation of cellular membrane and maintaining the activity of 2′-O-MTase activity [52], [58]
Unknown nsp11 short peptide at ORF-1a end [54]
RNA dependent RNA polymerase nsp12 Viral replication [54], [59]
Superfamily 1- helicase nsp13 Helicase, viral replication, virulence and tropism affection [50], [60]
3′–5′- Exonuclease nsp14 Exonuclease 3′–5′ activity and viral replication [50], [60]
N7-methyltransferase Nidoviral endoribonuclease specific for U nsp15 Viral replication and poly(U)-specific endoribonuclease [61]
S-adenosylmethinonine dependent nbose 2′ - O- methyltransferase nsp16 Viral replication, 2’-O-ribose methyltransferase inhibition and IFN antagonism [60], [61]

JNK-c - Jun N-terminal kinases, DMV-Double-membrane vesicle, NF-κB-Nuclear factor-κB, IL-8- Interleukin 8, RANTES-Regulated on Activation, Normal T Cell Expressed and Secreted, IFN-Interferon.

The most bountiful structural constituent of the viral envelope is the M glycoprotein produced by the M gene, which contains 222 aa (23 kDa). It extends the membrane bilayer three times, flees a concise NH2-terminal domain on the lateral side of the virus, and a long COOH terminus (cytoplasmic domain) on indoors of the virion [71]. It delineates the shape of the viral envelope by forming the homotypic interaction between them. It often interacts with the envelope E protein present in the host cell membrane budding compartment, produces the viral envelope and accountable for constructing virus-like particles (VLPs) [72], [73]. During the assembly and budding process, M protein is performing a dominant aspect in retaining the S protein by its interaction. It interacts with N protein, assists in RNA packaging and functions in viral immunoevasion [74], [75]. The envelope (E) protein is a smallest single-pass type ΙΙΙ transmembrane protein (8 kDa), consists of approximately 76–109 aa that are prevailing in tiny amounts on the viral envelope [76], as shown in Fig. 5 . The E protein is an exceedingly perpetuate protein across β-CoV as only 3 variants have been raised until now. Its homopentamer facet supports virus assembly, budding, envelope formation and pathogenesis [77], [78], [79]. It also serves as viroporins that self-assemble into the host membrane, emerging pentameric protein-lipid pores to participate in ion transport and the apoptosis induction by persuading the pore formation of the host cell membrane [80]. While an extensive amount of E protein is let out during the viral replication process, only a minimal amount is unified into the viral envelope. However, much of the protein is found at the site of intercellular trafficking [81].

Fig. 5.

Fig. 5

The structure of E protein pentameric ion channel predicted by nuclear magnetic resonance (NMR) spectroscopy (Adapted, redrawn and reprinted with permission from ref. [82], copyrights © 2018 Elsevier).

The N protein (50–60 kDa) are phosphoproteins that interact with viral genetic material in a beads-on-a-string pattern, producing the helically symmetric nucleocapsid (Fig. 6 ) [83]. During the replication and transcription process, the N-terminal domain (NTD) of N protein interacts with nsp3 of replicase-transcriptase complex (RTC) to tether with genomic RNA, regulating the viral RNA synthesis and altering the metabolism in infected host [84], [85]. After translation, a helical ribonucleocapsid (RNP) plays a prominent role in virion assembly [86].

Fig. 6.

Fig. 6

The Structure of N protein predicted by NMR (Adapted, redrawn and reprinted with permission from ref. [87], copyrights © 2020 Elsevier).

In addition to these four proteins, SARS-CoV-2 encodes at least six or more accessory proteins (3a, 6, 7a, 7b, 8b, and 9b) that are produced by ORF-3a, ORF-6, ORF-7a, ORF-7b, ORF-8 and ORF-10, respectively. Notably, all of them are translated from the subgenomic (sg) RNA [88]. The ORF-3a is an ion channel protein, triggers pattern recognition receptor (PRRs) superfamily members by interacting with apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-TNF-receptor-associated factor (TRAF3)-dependent ubiquitination. This contributes to viral spreading and infection [89]. Using a yeast two-hybrid system, ORF-6 associates with nsp8 and promoting RNA polymerase activity [90], [91]. Besides, ORF-7a, a type I transmembrane protein situated within the endoplasmic reticulum (ER) and Golgi network, is linked to the trafficking of the protein. Both ORF-6 and ORF-7 show protruding performance in SARS-CoV-2 pathogenesis [92]. While SARS-CoV has two ORF-8 proteins (ORF-8a and ORF-8b), SARS-CoV-2 has only a single ORF-8 protein with 366 nucleotides that encodes a protein with 121 aa [93]. A recent report has shown that ORF-8 interacts with ORF-6 and N protein, whereby it serves as a possible type I IFN signaling pathway inhibitors. IFN is the chief component of the host innate immune system protecting the host from the viral infection [94]. ORF-10 is the tiny accessory protein that consists of a 38-residue peptide with the highest number of immunogenic epitopes that could alter SARS-CoV-2 pathogenicity [95].

3. Mutation and clades of SARS-CoV-2

3.1. Mutations of SARS-CoV-2

Insights into the entire genome sequence of reference (Wuhan genome) and mutated SARS-CoV-2 species will succor in prospective vaccine and drug development. In general, RNA viruses, except for Nidoviruses, are susceptible to random mutations due to the scarcity of exonuclease proofreading activity of the virus-encoded RNA polymerases [96]. The first complete SARS-CoV-2 genome sequence was available on January 5, 2020, in the National Center for Biotechnology Information (NCBI) Genbank, and thousands of genomes have been sequenced till date [97]. The Global Initiative on Sharing All Influenza Data (GISAID) has made available those genomic data shared from all over the world in its database. There are currently about 4,383,873 million SARS-CoV-2 genome sequences (as of 16th October 2021) available on the GISAID data base (https://www.gisaid.org/hcov19-variants/). It has been useful to detect the viral mutations and track the virus movement around the world. SARS-CoV-2 mutations are caused due to errors like base substitution, insertion or deletion during the replication of genome [98], causing changes in viral characteristics and virulence. Hence they need to be studied for designing new vaccines, antiviral drugs and diagnostic assays as they could be used to determine changes in the viral immune escape, drug resistance and pathogenesis associated mechanisms. The first SARS-CoV-2 genome has about 80% sequence congruence with that of SARS-CoV, and it has undergone more than 10,000 documented single mutations announced on January 5, 2020 [99], [100]. Nevertheless, noted with SARS-CoV, SARS-CoV-2 S glycoprotein has 725 mutations over its 1255 residues, and their sequence closeness is only 76%. Among 725 mutations, 89 were on the RBD, which has 194 aa residues, proposing that the RBD is a concern to higher mutations [101]. RBD pivotal aa (5 out of 6) were disparate between SARS-CoV and newly emerged SARS-CoV-2, hence, it has a robust binding affinity to ACE2 than SARS-CoV [102]. The existence of S protein with plenty of mutations suggests the infectivity alteration of SARS-CoV-2 subtypes. It revealed that the existing mutations trigger the SARS-CoV-2 infectivity alteration and predict the future infection tendency. A recent report by Koyama et al. described that 10,022 SARS-CoV-2 samples were acquired from 68 countries, including the USA, UK, Australia and Netherland. Among them, 5775 distinct genome variants were noted, which comprises a total number of 2969 missense mutations, 142 non-coding deletions, 484 mutations in the non-coding regions, 1965 synonymous mutations, 66 non-coding insertions, 11 frameshift deletions, 36 stop-gained variants, 100 in-frame deletions and 2 in-frame insertions. It demonstrates the diverse in genomic variants involving structural transmission with the prospect of several debuts into the population [103]. One more bulletin from Mercatelli et al. divulged that the acquired 48,635 SARS-CoV-2 samples worldwide have a total of 3,53,341 mutations than the Wuhan reference genome (NC_045512.2). Notably, 48,379 samples consist of at least one mutation and an average of 7.23 mutations per sample. It is worth noting that few samples had more than 15 mutation occurrences. Notably, C > T transition, A > G transition and G > T transversions were estimated to be 55.1, 14.8 and 12%, respectively. Among them, A>G transition is most widespread in Europe, Africa and the America while G>T transversion is the second leading occurrence in Oceania and Asia [104]. One mutation event in the gene encoding of spike protein at the 614th position in the amino aspartic acid (D) is substituted by acid glycine (G), which is most widespread and named as D614G. The D614G was first noticed in Germany and China, eventually spread to all of Europe, Canada, USA, Australia and India [105]. Spike proteins are made of three smaller peptides arranged in open and closed orientations. When more peptides are in open orientations, they are easily accessible for binding of host proteins, as shown in Fig. 7 . The D614G mutation incites a more transmissible form of SARS-CoV-2 by changing a single amino acid in the viral RNA code, which relaxes the connection between peptides and results in open conformations to increase the chances of infection [106].

Fig. 7.

Fig. 7

(A-B) Structural representation of the smaller peptides in a spike protein RBD domain occurs as ‘open’ or ‘closed’ orientations (C) (Adapted and reprinted with permission from ref. [34], copyrights © 2020 Cell press).

3.2. Clades of SARS-CoV-2

A clade is a name for a group of a virus with genetic variations. It is also named as subtypes, genotypes or groups that all arise from a common ancestor [107]. SARS-CoV-2 is itself a clade inside the family Coronaviridae and the genus betacoronavirus. SARS-CoV-2 genome has changed by numerous mutations in the past few months, as they moved across the globe. A recent report displayed 11 major mutation occurrences, which are defined in five major clades according to its respective amino acid mutations: D392 (ORF1ab, G392D), S84 (ORF8, L84S), L378 (ORF1ab, V378I), V251 (ORF3a, G251V) and G614 (S, D614G). The percentage of individual clades is as follows: clade G614 (71.14%), S84 (10.85%), V251 (7.66%), D392 (1.03%) and L378 (1.70%) of all the sequenced viral genome.

3.2.1. Clade D614G

The clade G614 has been widespread globally with the non-coding variant 241C > T along with 3037C > T and ORF1ab P4715L [108]. The most prevalent mutation of transversion altered 23,403rd nucleotide adenosine into guanosine (A23403G) in the spike protein of SARS-CoV-2 genome G-clade, which is widespread in Europe, Oceania, South America and Africa. The clade G and its two derivative GH and GR having four mutations, namely C3037T, C241T, A23403G and C14408T. The ORF3a further characterizes GH's derived Q57H mutation and GR having an N gene trinucleotide mutation, named RG203KR mutation [109]. The G clade and its offspring (GH and GR) are most familiar worldwide and accounting for 74% among the sequenced SARS-CoV-2 genome [110]. Phylogenetic studies displayed that the mutations in ORF8:L84S and ORF3a: G251V regions of SARS-CoV-2 are associated with the formation of new clades S and V, respectively. The nsp6: L37F mutation is also occasionally observed in combination with the above mentioned clades. While G and GR clades notably exist in Europe, S and GH clades have been mostly widespread across the globe [111], [112]. The clade L was the first SARS-CoV-2 type (reference genome NC_045512.2) to appear in Wuhan in 2019 December. It mutated into clade S in the following month, and it gave rise to clade V around mid-January 2020. The clade G also appeared around the same period. By the end of February 2020, clade G gave rise to GR and GH clades and started to spread across the globe. Notably, L and V clades have gradually disappeared over the same time. Similarly, the clade S is also declining but still could be seen in US and Spain [104]. Apart from these clades, few infrequent mutations are also seen, and grouped under clade O. The characteristics of each clade have been given in the table along with their names based on different nomenclature systems [Table 2 ]. Despite these mutations, SARS-CoV-2 has limited variability; hence the virus structure remains the same even with different variants. The average pairwise difference between any two genomes is 9.6 single nucleotide polymorphisms (SNPs), which show that SARS-CoV-2 has attained only a moderate genetic diversity [113]. This also means that vaccines developed or currently under development have a high chance of succeeding the spread of the virus. There are two commonly used nomenclature systems called Year-Letter nomenclature and PANGOLIN (Phylogenetic Assignment of Named Global Outbreak LINeages) nomenclature. Year-Letter nomenclature names clades that persist for at least several months with a significant geographic spread. Their frequency should exceed 20% in a global sample with a variation in at-least 2 positions compared to its parent clade. The clades such as 19A, 19B, 20A, 20B, and 20C are the currently named clades in this system (https://nextstrain.org/blog/2020-06-02-SARSCoV2-clade-naming). PANGOLIN lineages offer elaborated outbreak cluster information, whereas other two nomenclatures served a huge scale entire view of clade trends [114].

Table 2.

Year-Letter Nomenclature names clades that persists for at least few months with important SARS-CoV-2 geographic spread.

GISAID Clade Year Number PANGOLIN Lineage Genomic Coordinate Effect on Protein sequence Class Effect on protein References
L 19A B NC_045512.2 Wuhan reference genome [97]
S 19B A C8782T S76S Silent SNP nsp 4 [104]
T28144C L84S aa-changing SNP ORF8 protein
V 19A B.2 G11083T L37F aa-changing SNP nsp 6 (transmembrane protein) [137]
G26144T G251V aa-changing SNP ORF3a protein
G 20A B.1 A23403G D614G aa-changing SNP Spike protein [138]
C14408T P314L aa-changing SNP nsp12, post-ribosomal frameshift (RNA-dependent RNA polymerase)
C3037T F106F Silent SNP nsp 3 (predicted phosphoesterase)
C241T 5’ UTR SNP 5’ Untranslated Region
GH 20C B.1.* G25563T Q57H aa-changing SNP ORF3a protein [137]
In addition to the 4 mutations of clade G
GR 20B B.1.1 GGG28881AAC RG203KR aa-changing SNP triplet Nucleocapsid protein [139]
In addition to the 4 mutations of clade G

3.2.2. B.1.1.7 lineage in UK

Given the fact that the replication of SARS-CoV-2 to produce more of their copies inside the host cells results in mistakes and generates an erroneous reproduction of its genetic material termed as mutation [109]. The genetic variant assessment in SARS-CoV-2 provides an expanding knowledge on new clade or lineage formation and adaptation to contain its outbreak. Similar like other RNA viruses getting mutated over a time, the fundamental aspects of viral biology reveal different mutations of SARS-CoV-2. The recent estimates specify that the nucleotide mutations are acquired by circulating SARS-CoV-2 lineages at a rate of about 1–2 mutations per month [115]. In this context, the SARS-CoV-2 Genomics UK (COG-UK) consortium surveillance dataset identified and reported a distinct phylogenetic cluster to the WHO on December 14, 2020. After the genomic sequencing, it is termed as VUI 202012/01 (Variant Under Investigation) or “B.1.1.7 lineage” [116]. The B.1.1.7 refers to a mutation in the SARS-CoV-2 RNA that renders the virus more contagious up to 70% with a vast number of genetic variations in the S protein. The B.1.1.7 RNA differs from oldest RNA by 23 mutations with at least 8 lying in the RNA portion corresponding to the S protein [117]. Notably, two deletions differ in new B.1.1.7 when compared to previously documented mutations and is significantly reducing the sensitivity of human antibodies against SARS-CoV-2. The 23 specific deletions/insertion of the reference Wuhan SARS-CoV-2 virus altered into the B.1.1.7 S variant, includes 8 spike protein mutations associated with high infectivity. These deletions may also be accompanying with diverse mutations in the CoV S protein binding region, including those stated in farmed mink infections and mutations that have been exposed to play a crucial role in the virus's ability to evade human immune systems. A truncated ORF8 gene is also present in B.1.1.7 and the deletions in this region is associated with a decreased severity of the disease as per previous observations [118]. As a result, it is most prominant to determine the functional effects of these mutations and deletions, especially the combination that is present in B.1.1.7. As the number of cases and regions that documented the infections with B.1.1.7 is rising with each passing day, a detailed investigation on number of mutations, the cause for this particular variant and the different biological assets of mutation candidates is urgently needed [119]. Two earliest sampled genomes belonging to the B.1.1.7 lineage were obtained in Kent on 20-Sept-2020 and Greater London on 21-Sept-2020. Since then, the infections of B.1.1.7 are on the rise in the UK and it has 1623 genomes as of 15 December 2020. Of all these, 519 samples from Greater London, 555 in Kent, 545 in other parts of the United Kingdom (including Scotland and Wales) and 4 in other nations have shown B.1.1.7 infections. Based on epidemiological and genetic studies, several research teams have concluded that B.1.1.7 is spreading more efficiently than other strains [117]. One of the most notable modifications in VUI-202012/01 seems to be N501Y, a transition from asparagine (N) to tyrosine (Y) at amino-acid site of 501. This is due to its location within the RBD of the spike glycoprotein, more precisely within the RBM, a component of the RBD used to bind with human ACE2. The RBD mutations can affect the recognition of antibodies and the specificity of ACE2-binding. It may also contribute to the virus becoming more contagious [120]. It is highly likely that N501Y affects the receptor binding affinity of the spike protein. As a result, this mutation alone or in combination with a deletion in the N-terminal domain at 69/70 provides high transmissibility for the virus. Separately, another variant having the same N501Y change was also detected in South Africa [116]. Though it has a separate lineage from the UK variant, it also showed higher transmissibility for the virus.

The N501Y change was also detected simultaneously in Australia and US (June–July 2019) and Brazil (April 2019). Hence, there was no clarity whether it was imported from UK. As 501 position is in RBD, this mutation significantly reduces the neutralizing ability of antibodies and leads to high transmissibility [121]. Various studies on different monoclonal antibodies showed that one antibody (LYCoV016) exhibited decreased efficacy against SARS-CoV-2 variants having mutation at 501 position. Presently, there are no neutralization data for N501Y mutation obtained from polyclonal sera of natural infections. Recently, Dr. Shibo Jiang, Fudan University, Shanghai, reported the SARS-CoV-2 with 501Y mutation linked with high infectivity and virulence in mouse models. It displayed that N501Y mutation triggers to increase the SARS-CoV-2 binding affinity with the mouse ACE2 receptor and also increase virulence [122].

3.2.3. B.1.351 variant

It is also known as 501Y.V2 in the GH clade, emerged in late 2020 in Eastern Cape, South Africa, and found to be high transmissibility [123]. B.1.351 contains 9 spike mutations (in addition to D614G) in various domains such as NTD (e.g. a cluster of mutations at 242-244del and R246I), RBD (three mutations at K417N, E484K and N501Y) and one mutation (A701V) near the furin cleavage site [124]. The E484K mutation plays a crucial role in the loss of neutralizing activity of some monoclonal antibodies as well as most convalescent and vaccine sera against variant B.1.351. There is a growing concern that these new variants could impair the efficacy of current mAb therapies and vaccines as these mutations reside either in the antigenic supersite of NTD or in the ACE2-binding site (also known as the receptor-binding motif (RBM), which is a major target of potent virus-neutralizing antibodies) [123], [125]. The receptor-binding domain mutations are largely driven by E484K, provide tighter ACE2 binding and widespread escape from monoclonal antibody neutralization. Further, K417N and N501Y mutations act together against some important antibody classes [125]. As a result, the newly emerging strains have picked up multiple changes (e.g. deletions and substitutions) in the spike protein. For instance, the identified B.1.351 variant has acquired mutations in the ACE2 interaction surface of the RBD that led to increased transmissibility [126]. Thus, it has rapidly expanded and became the dominant strain in the regions where they were first identified, and posing a serious challenge to spread across the globe.

3.2.4. B.1.427/B.1.429 variant

The new variant of concern (VOC) named as CAL.20C (B.1.427/B.1.429) was originally detected in California and is currently spreading throughout the US and other countries. The variant has five mutations as S13I, W152C and L452R in S protein and I4205V and D1183Y in the ORF1a. It has two mutations (S13I and W152C) in the NTD while having L452R mutation in the RBD [127]. As of March 26, 2021, the sequenced genomes reported in GISAID for the B.1.427 and B.1.429 lineages are 4292 and 10,934, respectively. B.1.429 is less susceptible to neutralizing antibodies and may be linked to worse outcomes of disease [128]. In fact, two recent vaccine trials have shown lower protective efficacy against B.1.351 in South Africa. As the above said spike mutations are specifically targeting the neutralizing antibodies, there is a growing concern about possible immune escape, which could compromise vaccine efficacy and increase the risk of reinfection [129], [130].

3.2.5. B.1. 526 variant

B.1.526 variant has unique set of spike mutations and mostly scattered in the Northeast of the USA. Nearly all of the newly identified B.1.526 variants have a set of common mutations in the spike protein such as L5F, T95I, D253G, E484K, D614G and A701V [131]. The B.1.526 variant showed the ability to neutralize antibodies and making current SARS-CoV-2 treatments less effective when tested against 4 monoclonal antibodies, 10 convalescent plasma and 10 vaccine sera. Notably, the decrease in neutralizing activities were found to be 7.7 and 3.4-fold for convalescent plasma and vaccine sera against E484K variant, respectively. Recently, several versions of B.1.526 have emerged with different mutations on the spike protein's receptor-binding domain. While some versions have E484K mutation similar to B.1.351 and P1, other versions have S477N mutation on the spike protein instead of E484K mutation [132]. As a result, it increases the affinity for the ACE2 receptor. Though these versions have both D614G and A701V mutations, L5F, T95I and D253G mutations are not observed in previously reported variants. It has been reported that the B.1.526 variant with S477N mutation had neutralizing titers similar to SARS-CoV-2 with D614G mutation. The B.1.526 variant with E484K mutation shows decrease in neutralizing activity against convalescent sera by 3.8-fold [124].

3.2.6. P.1 Lineage and Double mutant’ SARS-CoV-2 variant

The P.1 or Brazilian variant, also known as 20 J/501Y·V3, is one of the SARS-CoV-2 variant emerged from Brazil. It was first identified by the National Institute of Infectious Diseases (NIID), Japan, on 6 January 2021 in four people who had arrived in Tokyo after visiting Amazonas, Brazil. The genome sequencing revealed that the emergence of Lineage P.1 contains 10-lineage-defining amino acid mutations in the virus spike protein (L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, H655Y and T1207I) when compared to its immediate ancestor (B.1.1.28). Three key mutations present in P.1, N501Y, K417T and E484K, are observed only in the spike protein RBD [133]. In addition, it has been understood that E484K helps the virus to evade the antibodies generated by previous infections and make it less vulnerable to antibody drugs. The P.1 (the shortened form of B.1.1.28.1) has mutation at the 484 position of the spike protein that might decrease the susceptibility of the virus to immunization through vaccination or prior infection [134].

Recently, the Indian SARS-CoV-2 Consortium on Genomics (INSACOG) identified a new “double mutant” variant of the SARS-CoV-2, wherein two mutations, including E484Q and L452R rise together in the same virus [135]. These mutations may confer immune escape and increased infectivity for the virus. The E484Q mutation is known to be similar to E484K-a mutation seen in the B.1.351 (South Africa) and P.1 (Brazil) variants, which have emerged independently several times. However, the L452R mutation (which is also found in the “double mutation”) was first identified as part of B.1.427/B.1.429 lineage in the US, hence, it is also called as the “California variant” [136].

4. Mode of entry, infection and replication of SARS-CoV-2

4.1. Mode of entry and infection of SARS-CoV-2

As mentioned earlier, SARS-CoV-2 easily enters the host body through droplet infection and makes its way into the respiratory tract. Homotrimers transmembrane S glycoprotein made by 1273 amino acids protruding the exterior of SARS-CoV-2 facilitates viral entry into host epithelial cells [140], [141], [142]. SARS-CoV-2 S glycoprotein is a type I viral fusion protein that require protease cleavage for its activation and subsequent fusion. Subsequently, two subunits (S1 and S2) are participating in viral fusion with the host cell membrane [143], as shown in Fig. 8 . While the S1 receptor subunit consists of three domains, namely a single peptide, an extracellular N-terminal domain (14–305 aa) and RBD (319–541 aa), only the single peptide is used for tethering SARS-CoV-2 to host cells. The S2 subunit consists of a well-maintained fusion peptide (788–806 aa) and double heptad repeats (HR1 (912–984 aa) and HR2 (1163–1213 aa)) followed by a transmembrane region (1214–1237 aa) and a cytoplasm domain (1238–1273 aa) [144]. The S2 subunits interact with ACE2 receptor of host cells and initiate the viral-host cell membrane fusion process. Both S protein domains (S1 and S2) are separated from each other through a flexible loop comprising a cleavage spot accessible to host cell proteases [145]. Two-step proteolytic cleavages are occurring for the activation of S protein: 1) the cleavage at the site between S1 and S2 and 2) the activation cleavage at the site of S2 [146]. The S protein of SARS virus is activated by endosomal host proteases, namely transmembrane protease and serine 2 (TMPRSS2) [147]. While the S2 cleavage position in both viruses is identical, the cleavage site in S1/S2 differs. The S1/S2 cleavage site of SARS-CoV-2 is Arginine (Arg) 815, whereas it is Arg 797 for SARS-CoV [148], [149]. The different host proteases such as trypsin, cathepsin L, furin, TMPRSS-4, TMPRSS-2 and human airway trypsin-like protease (HAT) are required depending upon the cell types for the cleavage of S protein [150], [151]. ACE2 is an integral membrane glycoprotein known to have the highest expression in organs such as kidneys, lungs and heart [152]. The ACE2 is strongly expressed in cells such as kidney proximal tubule cells, absorptive enterocytes in the ileum and colon, bladder urothelial cells, cholangiocytes, myocardial cells, lungs AT2 (type II alveolar) cells, upper esophagus and stratified epithelial cells [153], [154], [155]. ACE2 has three domains, namely N-terminal signal peptide, C-terminal collectrin-like domain (CLD) and HEXXH zinc-binding metalloprotease motif bound peptidase domain (PD). The C-terminal domain consists of a ferredoxin-like fold “Neck” domain engaged with small extracellular domain, the single transmembrane hydrophobic helix and an intracellular segment [156]. The S1 domain of SARS-CoV-2 is attached to a helix 1 (Lys31 and Tyr 41) and b5 region (Lys353) of the PD domain of ACE2. Subsequently, the cleavage occurs at C-terminal (aa 697 to 716) by the activity of TMPRSS2 that enhances the S-protein-driven viral entry, as shown in Fig. 8. The ACE2 binding affinity with SARS-CoV-2 S protein is higher (~15–40 nM) than SARS-CoV S protein [157], [158]. Also, the expression of ACE2 protein at the lung alveolar epithelial cell exterior permits the respiratory tract infection by SARS-CoV-2. Recent reports displayed that men have overexpression of ACE2 in lung alveolar cells than females, and ACE2 expression was higher in Asian people than Caucasian and African American people [159], [160]. The renin-angiotensin system (RAS) has a major role in regulating blood pressure, electrolytes and fluid balance in the human body. ACE2 forms angiotensin (1-7) by interacting and cleaving angiotensin II, and the resulting complex of ACE2/angiotensin-(1-7)/MAX axis counteracts the negative impact of RAS. This complex causes inflammation, hypercoagulation, major adverse cardiovascular event, insulin resistance, endothelial dysfunction and respiratory problems [161], [162], [163]. SARS-CoV-2 is entering into the cells of central nervous system, gastrointestinal tract and respiratory tract including the pancreas via the ACE2 receptor, and thus, causing adverse tissue damages [164]. Recent reports displayed that the SARS-CoV-2 causes acute pancreatitis, which induces the self-digestion of pancreas, secretion deficiency and formation of large endocytic vacuoles in acinar cells [165], [166]. After the viral attachment, SARS-CoV-2 enters into host cells through endocytosis, thereafter enters into endosomes, and finally, the membrane fusion occurs between viral and lysosomal membranes [167].

Fig. 8.

Fig. 8

The cellular interaction between the host cell ACE2 and SARS-CoV-2 S protein.

4.2. Replication and progeny assembly of SARS-CoV-2

The viral genome is discharged into the host cell cytosol, wherein its replicase gene (ORF1a and ORF1ab) is translated to formulate replicase pp1a and pp1ab. The translated polyprotein cleaves and gets transformed to 16 nsps by PL-pro and 3CLpro [168], [169], [170]. PL-pro cleaves nsp1, nsp2 and nsp3 from the polyprotein N terminal, whereas 3-CL pro cleaves the remaining nsps from the C terminal. These non-structural proteins then carry the viral replication and transcription processes. The viral proteins such as nsp3, nsp4 and nsp6 subunits, are altered in the endoplasmic reticulum (ER) to direct the formation of viral replication organelles (ROs) [171], [172]. The viral genome replication takes place in the DMV of these viral ROs. This helps in immune evasion as they protect the viral RNA from innate immune responses [173]. The RNA-dependent RNA polymerase (RdRp), also called as nsp12, forms either RNA replicase-transcriptase complex (RTC) or RNA polymerase complex with nsp7-nsp8 heterodimer and nsp8 as cofactors in DMVs [174]. Then, both nsp7 and nsp8 proteins increase the poor RNA processivity by lowering the dissociation rate of nsp12 and RNA. The nsp8 protein also acts as a primase enzyme synthesizing short oligonucleotides primers for subsequent extension by the nsp12. The helicase enzyme (nsp13) unwinds the double-strand RNA for nsp12 polymerase [55]. The RTC complex plays a major role in replication process as it generates negative sense genomic RNAs, which would act as a template for positive-sense genomic RNA (gRNA) and subgenomic RNA (sgRNA) (by discontinuous replication) [175]. The nascent RNA strands synthesized are then proofread for misincorporated nucleotides by nsp14 exonuclease enzymes. The genomic and sgRNAs are then polyadenylated at their 3′ end and capped at their 5′ end as it protects them from the host antiviral response and degradation by cellular exonucleases [176]. Then, the capping process is performed by a GTPase, nsp13, nsp16-2’-O-methyltransferase/nsp10 complex and nsp14-N7-methyltransferase. While the sgRNA is translated into respective structure in ER, the full-length positive-sense genomic RNA released from DMVs combines with N proteins and forms a helical ribonucleoprotein (RNP) complex in the host cell cytoplasm [177]. Later, the structural proteins (S, M and E) and ribonucleoprotein (RNP) complexes (in encapsulated budding form) are separately entering into the endoplasmic Reticulum-Golgi intermediate compartment (ERGIC), a budding compartment. The RNP interacts initially with M protein to form inner structures and generates the basic viral structure by interacting further with S and E proteins. The matured viral budding is released later from the Golgi apparatus. After completing the viral replication cycle, the assembled SARS-CoV-2 progeny is released from host cells via exocytosis [178], [179], [180], as shown in Fig. 9 .

Fig. 9.

Fig. 9

Scheme reveals the SARS-CoV-2 life cycle in host epithelial cells. The cellular interaction between ACE2 receptor and S-protein facilitates SARS-CoV-2 entry into host cells. After completing the endocytosis and uncoating process, the SARS-CoV-2 components can be reused to formulate new viruses by host cells. Lastly, the new progeny viruses are released from host cells by exocytosis process.

5. Host-responses to SARS-CoV-2 infection

The international spread of SARS-CoV-2 is correlated with the host immunological naivety, accessibility of social dynamics, global communication and subdued innate immune responses [181]. The innate immunity is the preliminary virus removal system in the human body that induces adaptive immunity through the secretion of chemokines and cytokines [182]. The first line of innate immunity is activated by the binding of viral S protein with alveolar lung cells, which causes the activation of Pattern Recognition Receptors (PRRS), a local immune response that induces co-stimulatory signals for T lymphocytes (adaptive immune cells) [183].

Previous reports displayed that the lung epithelial cells, macrophages, and dendritic cells are bearing PRRs subfamily proteins such as nucleotide-binding oligomerization domain (NOD)-like receptor, endosomal and extracellular and toll-like receptors (TLRs) and cytosolic RIG-I like receptors on their cell surface. They are activated by the internalized single or double-strand viral RNAs. The activated PRR receptor proteins subsequently stimulate cytokine secretion, including tumor necrosis factor-alpha (TNF-a), type I/III IFNs, IL-6, Interleukin-18 and IL-1. Among them, type I/III IFNs are prominent cytokines for inducing the defense mechanism against the virus [184]. The secreted cytokines carry the primary adaptive immune response in target cells against the virus [185]. The second line of adaptive immune response is performed by T and B-cell responses. While CD8+ and CD4+ T cells are involved in triggering the production of antibody against the virus and killing the virus-infected host cells, respectively, the plasma cells (produced from B-cells secreted antibody) inhibit the viral infection and activate the alveolar macrophages to engulf apoptotic cells and neutralized viruses [186]. These kinds of adaptive immune responses can stop viral load into host cells and enable the recovery of patients from SARS-CoV-2 infection with minimal lung damage [187], as shown in Fig. 10 .

Fig. 10.

Fig. 10

Overview of innate immune response and interferon signaling between the recovery and severely affected patients by SARS-CoV-2.

Notably, severely affected patients with SARS-CoV-2 validate remarkably impaired IFN-I cytokine production than mild or moderately infected patients [188]. The increased production of cytokines in severely infected SARS-CoV-2 patients such as IL-2, TNFα, IL-6, IL-10, monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory proteins (MIP1α), interferon-γ-inducible protein 10 (IP-10) and granulocyte colony-stimulating factor (G-CSF), leads to severe lung and even systemic pathology [Table 3 ]. The high-level of cytokine production causes increased vascular permeability, plasma leakage and accumulation into the alveolar cavity. These kinds of events cause pneumonia, tissue damage in vital organs, respiratory failure and even multiple organ failure [189], [190]. The pro-inflammatory phase is followed by the immune suppression stage, which is characterized by a reduction in peripheral lymphocyte count. Lymphopenia is commonly seen in severe cases as they have reduced lymphocytes counts such as CD8+ T cells, B cells, CD4+ T cells and natural killer (NK) cells [191]. Thus, the adaptive immune response cannot be effectively initiated due to the reduction and dysfunction of these lymphocytes. Hence, the degree of lymphopenia could be used to predict the prognosis even at an early stage. The uncontrolled viral infection, inefficient viral clearance and weak antibody production stimulate the activation of more macrophages and resulting in severe cytokine storms that lead to death [192]. The patients with underlying medical conditions like chronic kidney disease, asthma, serious heart conditions, diabetes, severe obesity and immunocompromised (due to cancer treatment, smoking, bone marrow or organ transplant and prolonged use of corticosteroids) are severely affected by SARS-CoV-2 infection and worst treatment outcomes [193].

Table 3.

Abnormal secretion of the blood parameters and inflammatory cytokines in SARS-CoV-2 infection.

S. No Biomarker Function Ref.
1 Counts of predominant immune cells such as CD4+, CD8+ and NK cell The decreased level of CD4+, CD8+ and NK cell in peripheral blood mononuclear cell (PBMC) associated with severity of SARS-CoV-2 [210], [211]
2 MCP-3, IP-10 and IL-1Ra Among 48 cytokines, MCP-3, IP-10 and IL-1Ra were closely linked with SARS-CoV-2 disease severity and outcome. [212]
3 Serum amyloid A (SAA) The elevated level of SAA was identified in 80% SARS-CoV-2 patients and utilized as an auxiliary index for diagnosis [213]
4 Platelet count High platelet-to-lymphocyte and thrombocytopenia (TC) ratio was correlated with poor outcome and increased TC was associated with the incidence of myocardial injury in SARS-CoV-2 [214], [215]
5 C-reactive protein (CRP) Expressed as a biomarker at early stage of disease progression, CRP level was positively linked with disease severity, lung lesions and risk of acute myocardial injury. [216], [217], [218]
6 GM-CSF The increased level of CD14+, CD16+, GM-CSF+, monocytes and GM-CSF+IFN- γ+ T cells are correlated with disease severity. [219], [220]
7 IL-2, IFN-γ, IL-6, IL-8 and IL-10 Correlated with the severity of SARS-CoV-2 [IL-6, IL-8 and IL-10]. The increased levels of IL-2 and IFN-γ were correlated with the higher risk of respiratory failure. [221], [222], [223]
8 IL-18 The occurrence of IL-18 was to be main in antibodies producing B cell that is most important in the recovery. [224]
9 Lymphocyte count Lymphocyte count is correlated with disease severity. [225]
10 IL-4 Some reports displayed IL-4 was found to have potential mediator effect or correlated with impaired lung lesions. [226]

5.1. Brain injury

The brain glial cells and neurons express ACE2 receptors on their cell surface, which are a potential target for cellular infection by SARS-CoV-2. The SARS-CoV-2 can pass from the general circulation into the cerebral circulation by rupturing the endothelial lining and infect the neurons. It also leads to bleeding within the cerebral tissue [194]. About 88% of severe SARS-CoV-2 patients had impaired consciousness with hyposmia and acute cerebrovascular diseases at the early stages [195].

5.2. Acute cardiac injury

Acute cardiac injury is a major complication in SARS-CoV-2 infection caused by various mechanisms such as direct viral infection, cytokine storm, respiratory dysfunction and hypoxemia [196], as shown Fig. 11 . The elevated myocardial injury biomarkers (e.g., higher cardiac troponin and creatine kinase) were often seen in patients treated in intensive care units (ICU), showing its severity [197]. The elevated levels of troponin T, C-reactive protein and N-terminal pro-brain natriuretic peptide are linking myocardial injury to the severity of inflammation and ventricular dysfunction induced mortality [198]. The meta-analysis, a quantitative, formal and epidemiological study, revealed that the mortality rate of patients without myocardial injury (11.2%) was lower than patients with myocardial injury (67.1%) [199]. The cytokine storm and increased D-dimer level directly activates coagulopathy in severe cases (20–30%) of SARS-CoV-2 infection [200], [201]. The coagulation pathway is activated by multiple mechanisms. For instance, the SARS-CoV-2 binding with alveolar ACE2 receptors induces inflammation and reduces the pulmonary vasoconstriction, which causes the depletion of blood oxygen (O2) level. The reduced O2 level is described as a hypoxia condition. The vascular response to hypoxia is controlled by hypoxia-inducible transcription factors (HIF), facilitated by thrombus formation, leading to blood clots [202]. In other words, antiphospholipid antibodies (produced from immune cells) cause severe tissue damage due to coagulation [203]. Besides, the destruction of alveolar and capillary endothelial cells causes more inflammation and blood clots. Further, the increased rate of inflammatory cytokines, including TNF-α, IL-6 and IL-8, can cause venous thromboembolism (VTE) by activating the blood coagulation. The abnormal blot coagulation is associated with poor treatment outcomes and reduced survival rate in SARS-CoV-2 infected patients [204].

Fig. 11.

Fig. 11

Host responses during the viral infection. SARS-CoV-2 infects ACE2 expressing target cells in (a) brain, (b) lung, (c) kidney and (d) heart.

5.3. Acute renal injury

Recent reports have found that the lymphocyte infiltration and severe acute tubular necrosis in kidney tubules are caused by SARS-CoV-2 infection. The detection of a cluster of virus-like particles with distinctive spike proteins in podocytes and the tubular epithelium of the kidney under electron microscopy revealed the direct viral infection of the kidney in SARS-CoV-2 infected patients [205]. Various biological factors such as systemic hypoxia, possible drug or hyperventilation-relevant rhabdomyolysis and abnormal coagulation contribute to acute renal injury. The optical microscopic analysis also displayed the diffuse proximal tubule injury with Frank necrosis, non-isometric vacuolar degeneration and brush border loss. Also, the hemosiderin granules and pigmented casts are observed in acute renal injury, which has obstructing capillaries lumen without fibrinoid material or platelet and prominent erythrocyte aggregates [206]. In a recent report, a clinical investigation on kidney injury of SARS-CoV-2 patients showed high serum creatinine, new-onset proteinuria, the infiltration of lymphocytes like CD68+ macrophage into tubulointerstitium and the enhanced deposition of complement, C5b-9 on the tubules [207]. Further, it showed the direct viral infection on kidney organoids for shedding viral progeny, which further confirms its ability to cause acute renal failure in humans [208].

5.4. Rhabdomyolysis

The patients with SARS-CoV-2 were reported to have developed rhabdomyolysis with symptoms of muscle pain and weakness. It could have resulted from the direct viral infection as severe immune response to the viral infection causes cytokine storm and muscle tissue damage. Further, the circulating viral toxins could directly destroy muscle cell membranes [209].

5.5. Neurological manifestations

Neurological manifestations of SARS-CoV-2 are neurological and other extra respiratory symptoms that occur in patients as a direct result of the virus's neuroinvasive features or as an indirect outcome of downstream multi-organ dysfunction and abnormal biochemistry.

5.5.1. General neurological symptoms

The virus causes neuropsychiatric problems in some patients, including altered consciousness and encephalopathy. More serious neurological consequences such as cerebrovascular accidents and seizures, were uncommon, however occurring in only 3 and 0.5% of cases, respectively. Neurological problems were shown to be more common in those who had “severe” SARS-CoV-2 infections [227], [228]. A recent report discovered that the majority of neurological manifestations occurred early in the disease, which could be a strong indicator of future clinical worsening. Severe neurological diseases such as bleeding of cerebral vein thrombosis, ischemic stroke etc. were more important and potentially enduring neurological sequelae by SARS-CoV-2 in patients that lead to a significant proportion of death [229], [230].

5.5.2. Stroke

The percentage of thromboembolic complications in patients with SARS-CoV-2 revealed the incidence of ischemic stroke to be 1.6% and 2.5%, respectively. Beyond regular cardiovascular and metabolic comorbidities and those associated to a prolonged stay in intensive care amenities, there are obviously added risk factors that predispose individuals with SARS-CoV-2 to develop thromboembolic stroke [231], [232]. The SARS-COV-2 patients with thrombo-inflammatory conditions showed increase in concentration levels of platelet (62%), interleukin-6 (IL-6) (100%), D-dimer (100%) and fibrinogen (94%). They offer an association between inflammation and consequent coagulopathy induced by IL-6 and fibrinogen [233]. An inflammatory state is ‘on’ when the alveoli is damaged that triggers the release of inflammatory cytokines such as IL-6. The downstream effects can be classified into two parts: (i) the release of procoagulant factors, and (ii) the damage to the capillary endothelium that leads to a dysregulation of its antithrombotic capabilities. Thus, both of these factors lead to the formation of microvascular thrombosis, which has the ability to embolize the entire body [233].

5.5.3. Guillain-Barre syndrome (GBS)

GBS is a significant neurological complication associated with SARS-CoV-2 infection. Both the onset of GBS symptoms and the SARS-CoV-2-specific respiratory symptoms are exceedingly different. A mild temperature, apparent lower limb weakness, upper limb weakness, loss of deep tendon reflexes and a variety of sensory abnormalities, are all signs of GBS [234]. GBS has been linked to recent inoculation with a variety of bacteria, which could explain the disease's clinical heterogeneity. Several mechanisms have been hypothesized to explain the role of viruses in causing an acute areflexic state in GBS. The antibodies against surface glycoproteins are most likely produced in response to a pathogen that also interacts to native protein structures found on the surface of neurones, resulting in the clinical characteristics seen in GBS [235].

6. SARS-CoV-2 presence in sewage and public health concerns

In recent years, the investigation of waterborne infections is considered as an important tool to know more about the localized infections and disease origins [236]. It helps us in controlling viral infections and devising a better preventive strategy. Similar investigations show the evidence of presence of SARSCoV-2 in surface/ground water, sewage stream lines and contaminated drinking water [237], [238], [239]. SARS-CoV-2 has been proposed as a waterborne virus that enters and contaminates water sources via infected faeces [240]. Although the fecal-oral transmission of SARS-CoV-2 has not been confirmed yet, a detailed investigation is necessary to determine the impact of water and related sanitation initiatives in the spreading of COVID-19 by this route. The investigation on the presence of SARS-CoV-2 in faeces and its persistence in the environment suggests that it could stay for a longer time in the water and pollute the environment [241]. While the respiratory droplet infection by close contact is the predominant and most common mode of SARS-CoV-2 transmission, the possibility of fomite-based vertical and faeco-oral transmission can indeed be ruled out given the growing body of evidence [242]. However, SARS-CoV-2 needs to be explored for its ability to sustain in water, transmissibility through water and potential to infect humans. When compared to non-enveloped viruses, the enveloped viruses such as coronaviruses (CoVs) have various structural and survival features to stay alive for longer time in water [243], [244]. Hence, wastewater management is seen to be a viable method for tracking the spread of CoVs in a particular locality, i.e. viral clusters. The monitoring of critical reservoir on a regular basis will aid in detecting increasing viral concentrations or indicators, which might be used as early warning signs of an epidemic [245], [246]. Such reports can reveal the actual virus burden in communities, thus allowing proper control measures to be implemented to prevent further spread of SARS-CoV-2. Furthermore, when handling the stools of SARS-CoV-2 positive individuals, stringent personal hygiene (hand hygiene) and preventive measures must be followed. The appropriate disinfection strategies have to be followed for waste and sewage water coming from healthcare settings, aged homes, quarantine facilities and containment areas. Based on this, a detailed investigation to study the involvement of the fecal contamination route in SARS-CoV-2 transmission must be urgently performed to investigate the role of environmental parameters in keeping them alive and aiding them further transmission. To gain further information about the relation between SARS-CoV-2 RNA concentrations in faeces samples, disease severity and gastrointestinal symptoms, a complete investigation of enteric involvement and viral shedding in the faeces is urgently required.

7. Diagnostic methods for SARS-CoV-2 infection

The high specificity and accurate clinical diagnostic methods for SARS-CoV-2 infection are desirable for dealing with the earlier diagnosis and appropriate treatment (Fig. 12 and Table 4 ). The symptoms observed from SARS-CoV-2 affected patients are non-specific and could associate with various other respiratory infections [247]. As per the initial report by Guan et al., the patients' data collected from china showed that the most common symptom was fever in 43.8% of patients and 88.7% patients had developed fever after hospitalization. The other symptoms, cough and diarrhea, were observed in 67.8 and 3.8% patients, respectively [248]. In advanced molecular biotechnology, the nucleic acid detection method based on Polymerase Chain Reaction (PCR) has been referred as the gold standard for detecting the virus [249]. Conversely, computed tomography (CT) scan could also be used to examine any abnormalities present in the lungs, and hence, the diagnosis of the virus at earlier stage is possible [250], [251], [252]. Besides, molecular tests employing non-PCR based methods and novel emerging diagnostic methods (e.g., isothermal amplification, protein testing, Point of Care (POC) detection and SHERLOCK) are also under development. Following this, we have briefly reviewed and discussed various diagnostic methods used for the detection of SARS-CoV-2.

Fig. 12.

Fig. 12

Various diagnostic methods for SARS-CoV-2 identification in the given biological samples.

Table 4.

Various emerging diagnosis techniques for SARS-CoV-2 infection.

Platform Techniques Clinical sample Ref.
CRISPR RPA Serum [295]
CRISPR RT-RPA Nasopharyngeal swabs [290]
LAMP LAMP Throat swabs [296]
RPA RPA Fecal and Nasal Swabs [297]
NASBA Real Time- NASBA Nasal Swabs [298]
RCA RCA Serum [299]
RT-LAMP LAMP Nasopharyngeal aspirates [300]
ELISA ELISA Serum [301]
SIMOA Digital ELISA Serum [302]
Biobarcode Assay Immunoassay assisted by DNA Serum [303]
MCLIA Enzyme assisted immunoassay Serum [275]
Field Effect Transistor (FET) FET Nasopharyngeal swab [294]

CRISPR-Clustered Regularly Interspaced Short Palindromic Repeats, RPA- Recombinase Polymerase Amplification, RT- RPA -Real Time-Recombinase Polymerase Amplification, RCA-Rolling Circle Amplification, NASBA-Nucleic Acid Sequence-based Amplification.

7.1. PCR based methods

PCR is an enzymatic method separating two DNA strands to produce multiple gene copies, and primer marks the location of the gene segment present in DNA strands. Further, DNA polymerase starts formulating a new DNA strand by adding nucleotides, forming two identical DNA copy from a short RNA segment [219]. This method is mainly used to obtain more DNA copies from minimal quantity of biological samples to make it adequate for laboratory study. In general, the viral genomic RNA is transferred into cDNA using reverse transcription [253], [254]. Then, PCR is carried out and the virus is detected using specialized methods. Among them, the sequencing and gel visualization techniques are conventional for the detection of CoVs. The designing of kits generally involves two main steps [255].

  • Aligning of sequence and designing the primer

  • Optimization of assay and testing

Corman et al. disclosed various SARS-CoV-2 genomes to identify sets of primers and probe through which they have identified three regions having conserved sequence, namely RdRP gene in the ORF1 ab region, E gene and N gene. Notably, the attempts to detect RdRP gene and E gene have shown high sensitivity as the technical limitations for the detection are 3.6 and 3.2 copies per reaction, respectively. However, N-gene resulted in 8.3 copies per reaction, and hence, exhibited poor sensitivity [256]. The assay has been made as a dual-target system with 2 sets of primers, wherein one set detects various coronaviruses and another set of primers specifically detects SARS-CoV-2 [255]. After designing suitable probes and primers, the assay conditions (e.g., incubation times, temperature and reagent conditions) were optimized prior to PCR testing. Though RT-PCR can be executed through one step or two-step assays, combining the reverse transcription and PCR amplification in a single step provides rapid and reproducible results. Despite such advantages, the difficulties in optimizing the reverse transcription process and PCR amplification (mostly due to simultaneous occurrence) result in lower amplification than the desired target. Since the reaction is performed sequentially in two distinct tubes, two-step assay is highly sensitive than one-step assay. However, it is time consuming and slow that results in low number of tests per unit time.

RT-PCR is analytically specific but not reliable due to its high false-negative rate [257]. Unfortunately, the positivity rate with RT-PCR is only 30–50% for laboratory confirmed SARS-CoV-2 patients, particularly in the early stage of the infection and if the samples were collected from upper respiratory tract [258]. It was shown in another report that 3% people confirmed for infection in chest computed tomography (CT) were given false-negative report after RT-PCR testing [259]. Later they were confirmed for viral infection in repeated swab tests. Further, it can amplify spurious nucleic acid contaminations and can provide false-positive results. Hence, a single negative-result from RT-PCR does not rule out SARS-CoV-2 infection in clinical diagnosis.

7.2. Computed Tomography (CT) scan

CT of the chest uses X-ray equipment to examine the abnormalities and helps in diagnosing the cause of unknown cough, fever, shortness of breath and other respiratory symptoms [251], [252]. The high-resolution CT test could be utilized for earlier diagnosis and to understand the SARS-CoV-2 infection severity in patients. Bernheim et al. observed virus-positive CT scan reports for 56% of patients in their early stages of disease within 0 to 2 days [250]. The CT images of the SARS-CoV-2 infected cases show bilateral occurrence of uneven ground-glass opacities, which can coalesce into the spherical shaped randomly distributed deep-clustered lesions along the periphery of the lung [260]. In addition to this, an irregular paving pattern (Irregular shaped paved stone pattern) was also observed along with an increase in consolidation in the lungs [261]. It is worth noting that the ground-glass opacities are prominent in the initial period of 0 to 4 days after symptom onset. However, the drawback of CT scan is its specificity for SARS-CoV-2 infection (~ 25%) as its inability to distinguish SARS-CoV-2 infection from normal pneumonia virus affects its specificity [262].

7.3. Isothermal amplification based method

The isothermal amplification techniques includes recombinase loop-mediated isothermal amplification (LAMP), polymerase amplification and helicase-dependent amplification [263]. LAMP retains some fundamental advantages such as exclusion of a thermal cycler, amplification at a constant temperature, faster test results and potentially a larger diagnostic capacity, while maintaining similar sensitivity and specificity, thus making it more suitable than the RT-PCR for monitoring a pandemic. The LAMP technique amplifies DNA under isothermal conditions, wherein the amplification is directly related to the turbidity caused by the production of magnesium pyrophosphate. It is extensively utilized for SARS-CoV-2 detection [264], [265], [266], [267]. RT LAMP reaction performed at 60–65 °C resulting in DNA polymerase with strand displacement activity to initiate the DNA synthesis. A primer set used for a typical LAMP assay consists of a four constructed primers (two inner and two outers), which can attach with six disparate sequences in the target genome [268]. The inner primes are termed as a backward inner primer (BIP) and forward inner primer (FIP). Each primer having two unique sense and antisense sequences is matching to the target DNA. The mechanism of LAMP involves three stages: (1) starting material production (2) cycling amplification and elongation and (3) recycling. As the LAMP uses a more number of primers, it is considered highly specific. Notably, the results are easily distinguished by naked eye by visualizing a color change (pH-sensitive dye addition), turbidity change (the reaction of a by-product) and fluorescence coming out of the sample (double-stranded DNA binds to the added fluorescent dye). The above mentioned changes reveal the amplification of DNA in the samples. Further, the procedure is simple, cheaper and faster, and hence, handling of a large number of samples is possible without any difficulty [269]. The reaction time is less than 1 h and the detection limit is nearly 75 copies/μL. Overall, this procedure is simple to follow, easy for visualizing the detection, has less background noise and doesn't need a thermocycler. Optimizing the primers and reaction conditions are the challenges in the LAMP technique.

7.4. Microarray-based method

The microarray detection method is a rapid and high throughput technique in which coronavirus RNA produces labeled cDNA with probes through reverse transcription [219]. This cDNA is loaded into the well and gets hybridized with the microarray fixed solid oligonucleotides. Further, free DNAs are removed by washing steps and SARS-CoV-2 RNA detection is performed by a specific probe detection. Guo et al. displayed a 24 single nucleotide polymorphism mutations predicting among the SARS-CoV S gene by microarray technique [270]. Since CoVs outbreak can happen in large numbers, the assays with an ability to detect an extensive viral range are being developed. In this regard, Luna et al. have attempted to increase the low-density oligonucleotide array to detect the entire CoVs genus whose sensitivity is comparable to RT-PCR technique [271]. Hardick et al. assessed a portable and microarray chip-based Mobile Analysis Platform (MAP) and near-POC diagnostic platform to detect the virus [272].

7.5. Protein testing method

SARS-CoV-2 antibodies or antigens produced in response to infection can be used for the diagnosis. However, the viral protein detection is difficult due to variation in viral load during the infection period. Wang To et al. reported that the SARS-CoV-2 viral load in infected person is decreased as a function of time [273]. The antibodies generated after the infection may provide additional time for the secondary detection of SARS-CoV-2. Further, the antibody test also utilized for close observation of virus. Due to antibody cross-reactivity produced for SARS-CoV-2 with other CoVs, developing serological tests with accuracy is challenging. When Lv et al. investigated cross-reactivity for spike protein of SARS-CoV and SARS-CoV-2 in 15 patients, it showed high cross-reactivity [274]. Long et al. demonstrated the detection of SARS-CoV-2-specific antibodies by magnetic chemiluminescence enzyme immunoassay (MCLIA) [275]. Though there was no cross-binding with S1 subunit of SARS-CoV S antigen, some cross-reactivity was noted with SARS-CoV N antigens when it was added with the serum sample of SARS-CoV-2 patients.

7.6. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)

Recently developed CRISPR has taken molecular diagnosis to the next level due to its benefits such as speed, precision, specificity, strength, efficiency, versatility, portable and inexpensive [276]. The CRISPR system enables researcher to alter gene function by making changes in genomic sequence. It closely resembles the pair of molecular scissors that can precisely cut the DNA strands. This system has a family of DNA sequences found in archae and bacteria, and is composed of two major parts: (i) Cas endonuclease and (ii) guide RNA. The former is responsible to break the target genomic site, whereas the latter is used to identify and lead the Cas endonuclease to the target [277], [278], [279]. The CRISPR system is further divided into two main classes and six types. The first class is composed of Type I, III and IV while the second class is consisting of Type II, V and VI. In first class, Cas 3 nuclease and Cas 10 nuclease of Type I and III are used to cut DNA and RNA, respectively. In the second class, type II and type V systems use Cas 9 endonuclease and Cas 12 nuclease to cut the DNA, respectively. Cas 13 nuclease is used in Type VI of second class to make cuts in target RNA. Notably, the class II system is generally adopted to diagnose the disease and used for genomic manipulation [280], [281], [282], [283], [284], [285]. For instance, multiplex diagnostic system was developed by Kellner et al., wherein the nucleic acid preamplification is incorporated with CRISPR/Cas to identify the targeted nucleic acid sequences [286]. DNA endonuclease targeted CRISPR trans reporter (DETECTR) is CRISPR based inexpensive diagnostic method, mostly relying on Cas 12 to detect the infection within ~30 mins [283]. Another technique called specific high sensitivity enzymatic reporter unlocking (SHERLOCK) can detect the sequence of nucleic acids in clinical samples [286]. Both these techniques are comparable to PCR in the performance wise, however they are cheaper than PCR. Recenlty, these techniques, -SHERLOCK and DETECTR, are approved and commercially available for the detection of SARS CoV-2 [283], [286].

7.7. Specific High Sensitivity Enzymatic Reporter Unlocking (SHERLOCK)

SHERLOCK is a nucleic acid detection approach, wherein the viral RNA sensing is carried out by Cas13a ribonuclease enzyme [287]. In this technique, the viral RNA is converted to cDNA by reverse transcription, followed by isothermal amplification of cDNA using Reverse Polymerase Amplification (RPA) [288]. The Cas13a forms a complex with RNA guide sequence and gets attached with the amplified RNA. When it binds with the target, Cas13a gets activated and produces a fluorescent signal by breaking and releasing the surrounding fluorophore quencher probes. It is highly sensitive and specific as it can detect a single molecule in 1 μL sample volume of DNA and RNA targets [287]. It is also reported that the scaling up of preamplification content can detect even a single-molecule in large sample volumes [289]. In terms of specificity, two similar viruses can easily be distinguished from one another (e.g., dengue and zika virus) [290]. The reason is that Cas13 doesn't get activated when there are more than one mismatches in crRNA target duplex. In the detection of cancer-associated mutations, human genotyping and single base distinction, the specificity can also be increased by introducing mismatchs into the crRNA. Another promising feature of SHERLOCK is rapid detection. However, the SHERLOCK technique is limited due to multi-step nucleic acid amplification that affects the target quantification [287].

7.8. Point of care diagnostics

Pont of care diagnostics is the method of detection in which the sensing is carried out without any need for laboratory or centralized facilities. Among these techniques, the lateral flow assays are made of paper-based membrane strip with two lines marked on it [291]. The first line is filled with gold nanoparticle (AuNPs)-antibody conjugates and another line is filled by capture antibodies. When the blood and urine samples from SARS-CoV-2 infected person is dropped onto strips, the antibodies in the samples spread over the strip. The SARS-CoV-2 antibody present in the sample forms a colourful complex by interacting with AuNPs-antibody conjugates and capturing antibody that can be visualized by naked eye. In the previous report, the lateral flow assay is combined with RT-LAMP to detect MERS-CoV effectively. Another such point of care diagnosis method is the microfluidic device, wherein palm-sized chips consisting of micrometer range channels and reaction chambers are used for the detection of viruses. On the chip, the liquid samples are made to mix with antibody specific reagents and separated via electrokinetic force, vacuum, capillary action, etc. [292]. The advantages of microfluidic devices are compactness, rapid detection, portability and miniaturization. The previous report has demonstrated these microfluidics devices' use as an attachment with mobile to detect sexually transmitted disease antibodies by sequentially moving reagent pre-stored in cassette [293].

7.9. Field Effect Transistor (FET) based biosensor

Seo et al. have demonstrated FET-based biosensor to detect SARS-CoV-2 virus using graphene-based sensing layer [294]. They prepared a FET based biosensing device with the dimension of 100 × 100 μm2 (L × W). Initially, a layer of poly (methyl methacrylate) (PMMA) over graphene whole supported on Copper (Cu) foil was spin-coated. The Cu foil was later etched by CE-100 (a copper etchant) and the obtained PMMA/graphene film was transported to SiO2/Si substrate. After dried at room temperature, the PMMA layer was removed using acetone and the remaining graphene layer was washed with isopropyl alcohol and dried under N2 atmosphere. Later, the photolithography followed by reactive ion etching was used to pattern the graphene layers. Before detection, the prepared device is functionalized with 1-pyrene butyric acid N-hydroxysuccinimide ester (PBASE) in methanol for 1 h. Among four structural proteins of the SARS-CoV-2 virus (i.e., spike, envelope, matrix and nucleocapsid), the spike protein was selected because it is a major transmembrane protein, highly immunogenic and shows better specificity due to amino acid sequence diversity among the family of coronavirus [290]. The electrical measurement using current and voltage before and after the attachment of the antibody was carried out. The slope variation indicates the SARS-CoV-2 spike antibody occurrence. The viral antigen detected by FET based biosensor is 1 fg/mL, i.e., the limit of detection. They have also carried out the real-time cultured SARS-CoV-2 detection from clinical samples. Interestingly, the device also showed no cross-reactivity with MERS-CoV antigen.

8. Therapeutic targets for SARS-CoV-2

The unfortunate pandemic of SARS-CoV-2 in early 2020 has caused a challenge to all researchers to find the potential therapeutic agents for the treatment. However, there was no reliable vaccines or drugs available for either treating or controlling SARS-COV-2 virus. Hence, the scientific communities were actively involved in the examination of approved/existing drugs for other diseases for drug repurposing efforts for the treatment of SARS-COV-2. In the initial period of pandemic, the drugs utilized in clinical trials were based on empiric data, which were actually developed for other viruses or parasites [304]. The antiviral therapies were developed to induce direct effect on SARS-CoV-2 either by blocking the viral entry to host cells or controlling the viral enzymes having significant contribution for genome replication (Table 5) [305]. Alternatively, other therapeutic agents were also developed with the aim of boosting the innate immunity towards viruses or hindering the inflammatory response that cause lung injury [306]. Besides, the acquired plasma or hyper immunoglobulin from recovered patients of SARS-CoV-2 infection was also tried to treat SARS-CoV-2 infections. The studies have shown that it can effectively prevent the viral replication and spread. Owing to continued efforts in the past 12–18 months, 23 vaccines have been approved by WHO as of 14th October 2021 for emergency use across the globe. Further, about 300 vaccines are either in different stages of clinical trials or in development stages. Hence, this section discusses the availability of therapies for SARS-CoV-2 infection, their advantages and limitations, as shown in Fig. 13 .

Table 5.

Mode of action and target of repurposed drugs for SARS-CoV-2 treatment.

S. No Drug name Target Mechanism of action Adverse effect Status Ref.
1 Chloroquine and Hydroxychloroquine ACE2
viral cell entry
Lysosome
Glycosylate inhibition of host cell receptors to block viral entry, acidification of endosomal and proteolytic process.
Immunomodulatory effects through inhibition of cytokine production, autophagy and lysosomal activity in host cells.
Overdose may present with respiratory arrest, cardiac arrest and hypokalemia and patients may also be given epinephrine Phase 4
Completed
[348], [349]
2 Lopinavir/Ritonavir Enzyme
3CL protease
Inhibition of 3CL protease.
Resists viral cell entry
Risk to Pediatric Patients Phase 4
Active, Not Recruiting
[350]
3 Ribavirin Nucleotide analogue remdesivi (RDV)
Inhibits HVC polymerase
RNA dependent
RNA polymerase inhibitor
Viral RNA synthesis and mRNA capping inhibitor
Hepatotoxicity and
hypersensitivity
Phase 4
Active, Not Recruiting
[351], [352]
4 Remdesivir RNA polymerase RNA dependent
RNA polymerase inhibitor.
Block viral replication
Over dose - remdesivir are not readily available. Phase 4
Not yet recruiting
[353]
5 Favipiravir RNA-dependent RNA polymerase (RdRp) RNA polymerase inhibitor which prevents RNA replication and transcription Not recommended, if the pregnancy is confirmed or suspected Phase 4
Not Yet Recruiting
[354]
6 Tocilizumab Cytokine storm Inhibition IL-6 receptor.
Block cytokine storm reduction
Monoclonal antibody
Over dose - neutropenia Phase 4
Not yet recruiting
[355], [356]
7 Sarilumab interleukin-6 inhibitor
monoclonal antibodies
Inhibition of IL-6 Receptor Decrease in neutophil count and a reversible decrease in fibrinogen Phase 4
Completed
[357]
8 Fingolimod Enzyme dihydroorotate dehydrogenase inactivating IL-6/STAT3 pathway Cardiac effects (bradycardia and heart block) Phase 4
Completed
[358]
9 Canakinumab interleukin-1 (IL-1) monoclonal antibody which acts against IL-1-β Adverse reaction Influenza Phase 3
Active, not Recruiting
[359]
10 Anakinra interleukin-1 (IL-1) monoclonal antibody which acts against IL-1 Receptor Rheumatoid arthritis
AEs (incidence > 10%)
Phase 4
Completed
[360]
11 Gimsilumab granulocyte macrophage-colony stimulating factor (GM-CSF) monoclonal antibody which acts against GM-CSF Not available Phase 2
Completed
[361]
12 Heparin Vero E6 cells Anticoagulant and anti-inflammatory Platelet counts usually do not fall until between days 5 and 12 Phase 4
Active, not Recruiting
[362], [363], [364]
13 Baricitinib JAK1/2 Antiviral activity
Clathrin-mediated endocytosis inhibitor
Janus kinases 1 and 2 (JAK1/2 inhibitor
Many adverse reactions Phase 4
Active, not Recruiting
[365], [366]
14 Ruxolitinib JAK1/2 Inhibition of Janus kinase (JAK) Myelosuppression, including leukopenia, anemia, and thrombocytopenia. Phase 4
Completed
[367]
15 Umifenovir Spike protein /ACE2
Cell membrane
Host cell membrane fusion, DNA and RNA inhibitor Pathological changes Phase 4
Completed
[368]
16 Pirfenidone Spike protein/ACE2 Inhibition of collagen synthesis, down-regulates profibrotic cytokines and decreases fibroblast proliferation
Inhibition of TNF – α and IL – 1 β
Photosensitivity rash and gastrointestinal symptoms. Phase 4
Completed
[369]
17 Camostat Mesilate Protease inhibitor TMPRSS2 inhibitor which prevents the viral replication
Block viral mutation
Rash, pruritus, nausea, abnormal laboratory test values, and diarrhea Phase 4
Withdrawn
[370]
18 Darunavir/Cobicistat Protease Viral entry inhibitor
Inhibits HIV replication
Jaundice (13%), ocular icterus (15%), and nausea (12%). Phase 4
Active, not Recruiting/ Phase 4
Active, not Recruiting
[370]
19 Aerosolized interferon -α cytokines Stimulate antiviral immunity [371]
20 Oseltamivir Neuraminidase Viral replication inhibitor No adverse reactions Phase 4
Completed
[372]
21 Baloxivir marboxil Viral endonuclease Viral multiplication inhibitor [373]
22 The SARS-Cov-2 specific protease drug candidate Protease inhibitor Prevent the viral infectivity
Inhibitor of ACE2, S protein and TMPRSS2 serine protease
[374], [375]
23 SARS-Cov-2 specific antibodies Antibody Bind with viral cell, and inhibit the viral entry and improve immune system [372], [376]

Fig. 13.

Fig. 13

Disparate treatment options for treating SARS-CoV-2 infection.

8.1. Therapeutic agents to target RNA-dependent RNA polymerase (RdRp)

The SARS-CoV-2 replication and transcription is strongly depending on RdRp to produce new RNA. The RdRp consists of a 500–600 residue catalytic module consists of fingers, palm and thumb domains [307]. The mutation induction in RdRp amino acid residue causes a complete loss or lowering of RNA polymerase activity, modifies the metal cofactor requirements [308]. A class of nucleoside analogs (NAs) and small molecular drugs are utilized as RdRp inhibitors that metabolize intracellularly into their active ribonucleoside 5′-triphosphate (RTP) form and get incorporated into the nascent viral RNA. They can cause chain termination and induce deleterious mutation in the viral RNA without any impact on other proteins in host cells. Conventionally, the viral RdRp is a low-fidelity enzyme due to a lack of proofreading functions. Recently, it has been demonstrated that the FDA approved HIV and HBV drug tenofovir-diphosphate (TFV-DP), a nucleotide inhibitor, effectively suppresses the SARS-CoV-2 RdRp activity [309].

8.1.1. Remdesivir

Remdesivir (RDV) is a most prominent monophosphate nucleotide analog consists of a 1′-cyano modification sugar (formally known as GS-5734) that displays the broad-spectrum of antiviral effects against numerous RNA viruses, including MERS-CoV and SARS-CoV [310]. RDV has structural resemblance with adenosine and had a half-life of about 1 h in plasma. It gets incorporated into nascent viral RNA and quickly turned into intermediate monophosphate and nucleoside metabolites [311]. The inactive form of monophosphate converts active adenosine triphosphate and inhibits the viral RNA polymerases. The nucleoside triphosphate is behaving as an adenosine triphosphate (ATP) analog ended with a natural ATP substrate to inhibit selective RdRp inside the virally infected cells [312]. RDV was originally developed to inhibit the spread of Ebola virus, however it was unsuccessful in clinical trials. Therefore, it was allowed immediately to enter into clinical trials for SARS-CoV-2 infection [313]. The antiviral activity of RDV (IC50 value) was found to be 0.99 uM in E6 cell model of SARS-CoV-2 [314]. The human trial of RDV with adults and pediatric SARS-CoV-2 patients (~12 years old and weighing 40 kg or more) in 5 and 10 days' courses showed that only the 5 days course of RDV demonstrated a statistically significant improvement when compared to the 10 days course and conventional standard of care [315], [316]. Further, the efficiency evaluation of RDV with SARS-CoV-2 patients through randomized placebo-controlled trial demonstrated that RDV was superior to placebo in shortening the recovery time from the viral infection. Based on the clinical trials that clearly evaluated the clinical efficiency of RDV inpatients with SARS-CoV-2 (mild-to-severe infection), the U.S. Food and Drug Administration (U.S. FDA) has approved RDV for the treatment of adults and pediatric hospitalized patients with SARS-CoV-2 (over age 12 years and weighing at least 40 kgs or more). It was also recommended for the treatment of severely suspected or laboratory confirmed infection of SARS-CoV-2 and patient with severity being described as SpO2 ≤ 94% on room air with additional requirement of oxygen, mechanical ventilation and extracorporeal membrane oxygenation [317], [318]. However, later it was proved that there is no evidence of RVD improving the survival and outcomes of therapies. Based on this, WHO has insisted and issued a conditional recommendation against the use of RDV as per November 20, 2020 report [319], [320].

8.1.2. Favipiravir

Favipiravir (FPV) is a modified pyrazine analog (also known as 6-fluoro-3-hydroxy-2-pyrazine carboxamide, T-705). It was actually approved for antiviral resistant influenza in Japan. It could successfully inhibit the replication of A and B influenzas and avian influenza. The cellular enzymes convert inactive FPV to an active form of favipiravir-ribofuranosyl-5′-triphosphate (FPV-RTP) by ribosylation phosphorylation. As it is recognized as a substrate by RdRp, it prevents the elongation of RNA strand and viral proliferation by inhibiting viral replication and transcription [321], [322], [323]. The interesting fact that FPV shows excellent antiviral effects against other RNA viruses (e.g. bunyaviruses, filoviruses and arenaviruses), all of which are identified to cause fatal hemorrhagic fever. This unique antiviral profile makes FPV as a prominent potential drug for specific untreatable RNA viral disease. Both the purine nucleosides and FPV has a conflict for the same binding site, hence, the existence of purine nucleoside analogs as an antiviral agent that could prominantly inhibit FPV [324], [325], [326]. The EC50 value was noted to be 61.88 μM/L for FPV towards Vero E6 cells infected with SARS-CoV-2. The FPV bioavailability is very high at about 98% in human. Its metabolites are mostly renally cleared, and its half-life is calculated to be 2 to 5.5 h [327]. The clinical trials were executed for FPV towards SARS-CoV-2 in China and Italy in Mid-2020 as both the countries were severely affected in that time period. The FPV doses were started with 1600 mg/day (twice) for a few days, followed by 600 mg twice a day for the next 9 days. It acts as a mutagen and has shown 3-fold increase in total mutation than control [328], [329]. Recently, the repeat dose toxicity studies in rats, monkeys and dogs showed the notable adverse effects of oral favipravir on hematopoietic system such as the reduction of red blood cell (RBC) production and increase of the liver function parameters (e.g., alanine aminotransferase, total bilirubin, aspartate aminotransferase, alkaline phosphatase and increased vacuolization in hepatocytes). Besides, testis toxicity was also noted. Hence, the administration of favipiravir should not be recommended for women suspected or confirmed for pregnancy [330].

8.1.3. Sofosbuvir (2'-F, Me-UTP)

Sofosbuvir (SFV) is a pyrimidine nucleoside analog with a hydrophobic masked phosphate group used for SARS-CoV-2 treatment due to its potential advantages such as low toxicity and high stability of the active molecule [331]. The hydrophobic masked phosphate group of SFV enables it to uptake into the infected cells, and then, it gets changed into its active triphosphate form by cellular enzymes [332]. The activated drug (2'-F, Me-UTP) attaches with RdRp active site using its fluoro and methyl groups at 2' position and prevents RNA chain extension. Furthermore, it performs as a RNA polymerase inhibitor by competing with natural ribonucleotides. Further, it has lower incorporation activity into SARS-CoV-2 polymerase than Uridine-5′-triphosphate (UTP) [333]. It is worth noting that SFV as a single agent, has mild toxicity. The common adverse effects of SFV are headache and fatigue. Currently, the Food and Drug Administration (FDA) Label warns of a risk of symptomatic bradycardia such as low blood pressure, shortness of breath, chest discomfort and pulmonary edema when Epclusa (sofosbuvir (400 mg)/velpatasvir (10 mg)) is used in combination with amiodarone [334].

8.2. Therapeutic agents to target viral proteases

The viral replication depends on the proteolytic cleavage of either one or several viral polyproteins encoded by the virus genetic information. These proteolytic processes are essential for the functions of polyproteins and cleavage of the host protein, facilitating infection, and for the cellular entry and viral replication of SARS-CoV-2. Therefore, viral proteases are a major target in the development of SARS-CoV-2 drug. The diverse range of repurposed drugs and their targets are mentioned in Fig. 14 .

Fig. 14.

Fig. 14

Schematic illustration showing the role of different repurposed drugs in targeting SARS-CoV-2.

8.2.1. Ivermectin

Ivermectin (IVM) is a familiar anti-helmintic agent used as an antiviral agent against flaviviruses such as Japanese encephalitis, dengue fever, chikungunya and tick-borne encephalitis [335], [336]. It is worth noting that there are no approved indications by FDA for the above-mentioned applications. However, its anti-inflammatory properties make it ideal for veterinary use for diseases caused by parasitic worms such as onchocerciasis and intestinal strongyloidiasis [337], [338]. In SARS-CoV-2 infection, the nuclear transport of viral protein is essential for viral replication. IVM targets importin α/β1 heterodimer mediated transport of viral proteins by dissociating IMPa/b1 heterodimer. It also prevents the transport of viral protein into the nucleus [339]. Hence, IVM causes hyperpolarization by triggering gamma amino butyric acid (GABA)-gated-Cl channels and paralyses the infesting organism [340]. Besides, IVM affects the host response immunomodulation by increasing IL-6 and C-reactive protein levels and activating the neutrophils [341]. It has been reported that 5 μM of IVM could curtail SARS-CoV-2 RNA up to 5000-fold at 48 h without any toxicity. The IC50 value of IVM was calculated to be ~2 μM towards SARS-CoV-2 [342]. Notably, the over dosage (higher than recommended dose) with orally administered formulation found to be lethal in mice models with death preceded by significant ataxia, ptosis, bradypnea, emesis, tremors, decreased activity and mydriasis [341], [342].

8.2.2. Lopinavir/ritonavir

Ritonavir (RV) is a small peptide molecule developed by Abbott Laboratories in 1996 that can inhibit HIV protease. Then, an enhanced second-generation human immunodeficiency virus 1 (HIV) protease inhibitor, known as lopinavir (LPV) was developed [343]. Retropepsin is a HIV-1 aspartyl protease enzyme liable for cleaving the structural viral Gag polyprotein, which performs a crucial role in HIV viral life cycle [344]. Peptidomimetic LPV drug consists of a hydroxyl ethylene scaffold that mimics the normal peptide linkage cleaved by HIV protease, resulting in the inhibition of HIV protease [345]. It causes the production of immature and non-infectious virus particles and proteolysis of Gag protein. In other words, LPV delivered alone causes low human bioavailability of ~25%, but in the case of co-administration with ritonavir, it improves LPVs bioavailability and reduces the drug metabolism [343]. The previously published Lancet report discloses that the amalgamations of lopinavir, interferon, ritonavir, and ribavirin have found to be very effective in targeting SARS-CoV-2 infection at the earlier stage. The combinational antiviral therapy divulged superior effects and shortened hospital duration in both combination and control groups and the duration was calculated to be 7 and 12 days (p = 0.001), respectively [346]. Further, the oral Kaletra solution is highly concentrated (contains approximately 42% ethanol) and posing a potential risk of overdose in children and infants. The common side effects of overdose in infants are cardiogenic shock, complete AV block, cardiomyopathy, lactic acidosis and acute renal failure. Further, the drug-drug interactions present in the formulations may also induce adverse side effects. The other effects observed are hepatotoxicity, pancreatitis and allergic reactions/hypersensitivity [347].

8.3. Nasal spray

Nasal drugs /spray have been developed for disinfection and treatment for SARS-CoV-2. To the obvious fact that the viral loads are expelled out of nasal cavities from infected patients, disinfecting nasal cavities can help to decrease the risk of infection in uninfected person as well as to reduce the viral load in infected person [377]. Few of developed products are given in Table 6 along with their working mechanism.

Table 6.

Developed nasal drugs, its mechanism and current status.

Product Name Developer Mechanism Current Status Ref
Nitric Oxide Nasal Spray (NONS) SaNOtize Reduces Viral RNA generation and fusion of S protein Phase-III completed [378]
Nebulizer suspension powder APEPTICO By restoring endothelial-epithelial barrier of pulmonary alveoli Phase II [379]
Inhaled mRNA antibodies Neurimmune and Ethris Encoded with mRNA and helps in neutralizing the anti SARS-CoV-2 antibodies Phase I [380]
Alvesco Covis Pharma Restrains viral replication Phase III [381]
Neuroactive nasal spray VistaGen Therapeutics By reducing anxiety through enabling synaptic pathways Phase III [382]
Interferon beta for nebulization Synairgen Enhances pulmonary antiviral defenses Phase II [383]
Leukine
sargramostim
nebulizer
Partner therapeutics Treating acute hypoxemia Phase II [384]

8.4. Therapeutic agents for blocking the virus–cell membrane fusion

8.4.1. Recombinant human angiotensin-converting enzyme 2 (rhACE2)

The rhACE2 is also known as APN01, can block the SARS-CoV-2 entry into the host cell by interfering with the interaction between S protein and the ACE2 receptor of host cells [385]. A recent report stated that SARS-CoV-2 infected human kidney and blood vessel organoids treated with rhACE2 could block early entery of SARS-CoV-2 infection in host cells by a factor of 1000–5000 times [386]. The rhACE2 could preclude further ACE2 receptor activation, thereby perpetuating pulmonary vascular integrity and inhibiting acute respiratory distress syndrome (ARDS) [387].

8.4.2. Hydroxychloroquine and chloroquine

Hydroxychloroquine (HCQ) and chloroquine (CQ) are long-standing oral drugs specifically used to treat malaria and chronic inflammation. These two drugs have unique properties such as lipophilic weak bases and superior diffusion into organelles membrane (e.g. cell membranes, lysosomes, endosomes and Golgi vesicles). Both drugs become protonated, trapped in the organelles and upsurging the pH in the cell interior [388]. It is postulated that the raise in endosomal pH prevents the viral particles fusion and their cellular entry [389]. Also, they are known to interfere in the glycosylation of ACE2. This can make spike protein-ACE2 binding less efficient and impedes the virus's entry into the cells [390]. HCQ and CQ can prevent the antigen processing, T-cell activation, TLR-cyclic GMP-AMP Synthase (cGAS), CD154 expression and down-regulate the pro-inflammatory genes [391]. The EC50 values for HCQ and CQ towards SARS-CoV-2 were estimated to be 6.14 and 23.90 μM, respectively [392]. However, both HCQ and QC trigger severe side effects, including neuropsychiatric effects, hypoglycemia, retinopathy and QTc prolongation [393], [394].

8.5. Peptidomimetics inhibitors

8.5.1. Therapeutic efficiency of peptidomimetics inhibitors for SARS-CoV-2

Peptidomimetics are compounds with pharmacophore as an essential element that mimics natural compounds such as peptides and proteins to interact with biological targets. This alternative can overcome the limitations of naturally available peptides, i.e., limited stability against proteolysis and poor bioavailability. Notably, they improve the receptor selectivity on the one hand while lowering the degradation rate by peptidases on the other hand. These special properties make them potential candidates for targeting SARS-CoV-2 [395]. Previous investigations demonstrate the potential of peptidomimetics in developing peptide-based therapeutics. Yan et al. have shown the structural image of human ACE2 with the SARS-CoV-2 RBD using cryo-electron microscopy. Notably, RBD was bound to ACE2 extracellular peptidase domain [396]. In an another investigation, a detailed study on targeting membrane fusion revealed that S2 site of the spike (S) protein facilitates the fusion, which includes the fusion peptide (FP), heptad region, namely HR1 and HR2, transmembrane site and a cytoplasmic tail. While S2 creates the hydrophobic α helix interface due to HR1 and HR2 sites' interaction following the receptor binding process [397], the membrane fusion begins as soon as the hydrophobic FB gets inserted into the host cell. Similarly, M. Hoffmann et al. reported that SARS-CoV-2 entry into the human cell is mainly dependant on ACE2 and TMPRSS2 [137]. This type of study and other research reports could provide a fundamental outline for the development of therapeutics.

8.5.2. Therapeutic efficiency of inhibitors for HR-1 site of SARS CoV-2 as target

Recently, Xia et al. carried out a study related to derivatives of SARS-CoV-2 HR2 peptides [398]. They prepared HR1 and HR2 based peptides called as 2019-nCoV-HR1P and 2019-nCoV-HR2P, respectively. The derivatives had been designed in combination with pan-CoV fusion inhibitor (EK1 peptide) and their biological properties were studied [399]. A similar peptide derived from HR2 site was successfully used against SARS-CoV to inhibit the fusion process. It inhibited the process of binding of HR1 region with HR2 region and prevented the pore formation. While the authors reported that 2019-nCoV-HR2P and EK1 displayed inhibitory activity with IC50 of 0.18 and 0.19 μM, respectively. Interestingly, 2019-nCoV-HR1P did not show any such effects up to 40 μM of concentration. Additionally, 2019-to-HR2P and EK1 showed prominent inhibitory effect on SARS-CoV-2 infection in ACE2 expressed 293 T cells with IC50 values of 0.98 and 2.38 μM, respectively [400].

As peptide-based therapeutics have half-life timeless in in-vivo, the lipid conjugation can increase the antiviral potency and improve the pharmacokinetics. To get more detailed information, Xia et al. developed a series of lipopeptides to ‘C' end of EK1 by connecting the cholesterol through polyethylene glycol (PEG) and/or an amino acid (GSG) [401]. This design (also termed as EK1C4) is very effective against pseudotyped SARS-CoV-2 and S-mediated cell–cell fusion of SARS CoV-2 when compared to EK1 therapy. The IC50 values were estimated to be 1.3 and 15.8 nM for pseudotyped SARS-CoV-2 and S-mediated cell–cell fusion of SARS CoV-2, respectively. It also demonstrates a good inhibitory effect against SARS CoV and MERS CoV. In vivo studies showed better prevention against HCoV-OC43 infection in mice models. The investigations on pre and post-infection showed that EK1C4 could effectively inhibit SARS-CoV-2 activity. Similarly, Zhu et al. developed a lipopeptide fusion inhibitor based on HR2 sequence called as IPB02, which displayed a good response in preventing SARS-CoV-2 infection [402]. The inhibition effect was determined by an assay of dual split protein with an IC50 of 0.025 μM whereas single-cycle infection assay exhibited the IC50 value of 0.08 μM.

8.5.3. Inhibitors for RBD of spike (S) protein as target

Peptide-based inhibitors can prevent the SARS-CoV-2 initiation to host cell entry by binding to ACE2 receptor through RBD of S protein. A report by Zhang et al. shows ACE2 peptidase domain (PD) α1 helix as an essential component for attaching the SARS-CoV-2-RBD using molecular dynamic simulations [403]. The authors had prepared a natural 23-mer peptide from the human ACE2 (hACE2) α1 helix (called as SBP1) and investigated the peptide-protein binding processes using kinetic binding assays. It showed that SBP1 binding with SARS-CoV-2 was strong with a dissociation constant (KD) of 47 nm and equivalent with that of full-length hACE2. Then, the biotinylated SBP1 prevents the contact between host cell receptor (ACE2) and RBD domain of SARS-CoV-2, and inhibiting the viral entry into host cells. However, a real-time investigation is needed to prove its efficiency against SARS-CoV-2. Recently Han et al. have screened compounds that could mimic ACE2 using computer modelling [404]. The XRD crystal structure investigations revealed that 15 amino acids from the ACE2 receptor can bind with the RBD of viral S protein during the cellular entry into host cells [405]. This report also suggests four peptides with this region were considered and involved with two α-helices extracted from PD of ACE2. Among four peptides, two have shown stable structure and interacted strongly with RBD of SARS-CoV-2. From all these studies and findings, it has been understood that at least one peptide could bound to the spike protein and block the interaction of SARS-CoV-2 with ACE2. Through computational findings, Barh et al. state that many anti-S protein peptides can inhibit the viral entry [404]. The interaction of spike protein of SARS-CoV-2 with peptides was identified by HPEPDOCK protein-peptide docking server. After extensive investigations, researchers have identified 10 peptides that could be effectively used against SARS-CoV-2. Similarly, Huang et al. have designed many peptides through computational approaches and showed their interaction with SARS-CoV-2 [406]. Researchers have also remade the peptide designs using the structural bioinformatics and logo analyses to enhance the binding affinity to RBD of SARS-CoV-2 [404]. Thus, these computational approaches in combination with in-vitro and in-vivo experiments could lead to the design of appropriate therapeutics [407].

8.6. Small molecules

8.6.1. Therapeutic efficiency of inhibitors for TMPRSS2 as host target

Nafamostat, a serine protease inhibitor used for disseminated intravascular coagulation (DIC), has an ability to prevent SARS-CoV-2 host cell entry by preventing the pathway through TMPRSS2 [408]. By performing dual split protein (DSP) reporter assay using spike protein of SARS-CoV-2, Yamamoto et al. have shown that nafamostat inhibits the fusion activity and blocks the SARS CoV-2 replication in pulmonary Calu-3 cells with EC50 of 10 nM after pretreatment [409]. The same group similarly used DSP to identify nafamostat as an inhibitor for MERS' fusion spike protein. It effectively impacted the terminating viral replication up to 100-fold at a minimal concentration of 1 nM. The viral activity decreased by 300-fold when added during the infection suggesting good antiviral activity. Nafamostat showed inhibitory effect towards TMPRSS2 serine proteases to prevent SARS-CoV-2 S protein-mediated cell fusion in the range of 10–1000 nM concentration. From previous reports and outcomes, it has been understood that camostat-mediated viral entry inhibition never surpassed 65% even at high drug concentration of upto 100 M. It revealed 35% of the SARS-CoV-2 used endosomal cathepsins for cell entry [410]. Camostat could also inhibit SARS-CoV-2 by 50 to 60% in TMPRSS2+ cell lines with no side effects, and the inhibition efficacy is increased to 100% by the addition of E64d [147]. Through animal studies, it was found that camostat mesylate controlled the viral entry by 10-fold in calu 3 airway epithelial cells and interestingly increased the mice's survival rate [411], [412]. Camostat is known to inhibit an in-vivo infection caused by a virus that utilizes TMPRSS2, and could be a suitable component of a drug combination against SARS-CoV-2.

8.6.2. Therapeutic efficiency of inhibitors for Cat/BL as host target

Oxocarbazate inhibitor CID23631927 has revealed better anti-viral effect and selectivity towards CatL of SARS-CoV [413], [414]. It is noteworthy that oxocarbazate inhibited CatL with IC50 and inhibitor constant (Ki) value of 0.4 and 0.29 nM, respectively. The CID23631927 has successfully inhibited SARS-CoV entry in human embryonic kidney 293 T cells, and there was no toxicity up to the concentration of 100 μM observed in human aortic endothelial cells [415]. Based on this, oxocarbazate inhibitor can be a potential therapeutic agent for treating SARS-CoV-2.

8.7. Plant-based molecules for targeting SARS-CoV-2 proteins

Most of the plants have the potential to mitigate the new SARS-CoV-2 infection. Numerous traditional herbal remedies also inhibited SARS-CoV-2 infection in a healthy person and enhanced patient health state with severe or mild symptoms [416]. For example, Runfeng et al. reported an herbal mixture described as Lianhuaqingwen that consists of a mineral medicine (gypsum and menthol) and a mixture of 11 medicinal species [416]. It showed strong anti-inflammatory and inhibitory effects towards Vero E6 cells infected with SARS-CoV-2. The herbal mixture also restricted SARS-CoV-2 replication, chemokine (C—C motif) ligand 2/monocyte chemoattractant protein-1 (CCL-2/MCP-1) and pro-inflammatory cytokines such as IL-6, C–X–C motif chemokine 10/ interferon-γ-inducible protein 10 (CXCL-10/IP-10) and TNF-α in a dose-dependent manner. The IC50 value of lianhuaqingwen was noted to be 411.2 μg/mL against SARS-CoV-2 [416], [417]. By virtually screening 83 compounds from traditional medicines, Lung et al. identified that theaflavin, an antioxidant polyphenol, showed inhibitory activity against SARS-CoV-2 RdRp [418]. Similarly, Zhang et al. virtually screened 115 traditional medicines and highlighted 13 medicinal plants consisting of naturally occurring polyphenolic compounds such as kaempferol and quercetin, which are of considerable interest in the treatment of SARS-CoV-2 [419]. Recent reports showed that phytochemicals such as 3,5,7,3′,4′,5′-hexahydroxy flavanone-3-O-β-D-glucopyranoside, amaranthin, methyl rosmarinate, licoleafol, calceolarioside B, myricetin3-O-β-D-glucopyranoside, myricitrin, (2S)-eriodictyol 7-O-(6′′-O-galloyl)-β-D-glucopyranoside and 5,7,3′,4′-tetrahydroxy-2′-(3,3-dimethylallyl) isoflavone could be potential anti-viral agents against SARS-CoV-2 [420].

The plant-based chemical components have high potential to inhibit the viral proteins effectively and block the replication/transcription complexes. The major targets of SARS-CoV-2 are PLpro, 3CLpro, RdRP and S protein. PLpro enzyme provides a significant contribution in the SARS-CoV-2 replication and maturation processes [421]. Similarly, 3CLpro enzyme plays a crucial role in SARS-CoV-2 life cycle by directly triggering the nsps maturation [422]. The essential viral enzyme 3CLpro monomer consists of three domains, termed as domain I (8-101aa residues), domain II (102–184 aa residues) and domain III (201–303 aa residues), and a long loop (185–200 aa residues) that attaches domains II and III. The 3CLpro active site has a Cys-His catalytic dyad (Cys145 and His41) that placed in the space between domains I and II. Hence, a detailed investigation on the 3CLpro structure and its catalytic mechanism might provide more information for the anti-SARS-CoV-2 drug development. The RdRp, also named nsp12 is a conserved protein needs to form SARS-CoV-2 replication/transcription complex by catalyzing the RNA replication from a RNA template [174]. Therefore, nsp-12 has high potential for use as prominent therapeutic target. In other words, the S protein cleavage activation and its structural integrity performs a prominent role in SARS-CoV-2 virulence and invasion. Notably, the blocking of viral entry into host cells by targeting either S proteins or the host cell surface receptor is valuable for developing the suitable therapeutic strategies. Thus, these four structural and functional proteins make attractive targets for SARS-CoV-2 drug development [55].

Recently, the molecule-protein docking was carried out between different molecules of plants and reported targets of SARS-COV-2. The use of plant-derived phytochemicals with proven antiviral activities such as quercetins, flavonoids and gallates are effectively able to inhibit the proliferation of SARS-CoV-2 [423]. More specifically, it is believed that the plants with high phenolic compounds such as Satureja hortensis, Zataria multiflora, Rosmarinus officinalis, Thymus spp., Eucalyptus caesia, Mentha spp., and Artemisia kermanensis could be very effective in controlling SARS-CoV-2 infections [424], [425]. Recently, an aqueous extract of Senna alata (a large genus of flowering plants in the sub family of Caeslpinioideae and family of Fabaceae) was demonstrated to show better relief from the virus symptoms in SARS-CoV-2 patients. Currently, it has been used in the treatment of malaria, flu, fever and other medical conditions [426]. Similarly, Nicotiana benthamiana has a significant place in plant based vaccine preparation. It is a SARS-CoV-2 RBD based vaccine and has shown good ability to induce positive SARS-CoV-2-specific immunity when formulated with CpG adjuvant in pre-clinical trials (status trial phase I/II, NCT04473690) via intramuscular immunization [427], [428]. It is being developed by British American Tobacco company through Kentucky BioProcessing unit (KBP, biotech subsidiary in the US). Besides, Medicago Inc. (Quebec City, QC, Canada) is also developing virus like particles (VLPs) (NCT04636697) to combat SARS-COV-2 using N. benthamiana [429]. They have successfully developed VLPs having structural similarity with actual corona virus using Covid-19 spike protein and lipid membrane of Nicotiana benthamiana plant. As it is lacking nucleic acid, it is obviously noninfectious. The clinical study with volunteer people has shown an enhanced ability to induce the antibiotic response in human [430], [431]. Further, the VLPs developed with influenza virus have also demonstrated good biocompatibility, safety and efficacy in animal models and human clinical trials [432]. The vaccine developed from tomato and low nicotine tobacco plants have also shown stable expression of S protein (S1) against SARS. Notably, it exhibited a significant increase in amount of SARS-CoV-specific antibodies after immunization in mice model. It can be concluded that the plant based vaccines developed so for have shown promising results in pre-clinical trials [433], [434]. Hence, any continued efforts in this direction might result in plant based safe vaccines for SARS-CoV-2 in near future (Table 7).

Table 7.

Plant based SARS-CoV-2 vaccine candidates.

Vaccine Plant Host Vaccine Antigen Trial status/ID References
CoVLP Transient expression in N. benthamiana SARS-CoV-2 S protein/ Viral like particle based vaccine Phase 3
NCT04636697
[435]
KBP-201 Transient expression in N. benthamiana SARS-CoV-2 RBD-based vaccine Phase 1/2
NCT04473690
[436]
IBIO-200 Transient expression in N. benthamiana SARS-CoV-2 Viral like particle based vaccine Pre-clinical study [437]
IBIO-201 Transient expression in N. benthamiana SARS-CoV-2 Spike-based sub-unit vaccine Pre-clinical study [437]
IBIO-202 Transient expression in N. benthamiana SARS-CoV-2 nucleocapsid protein-based sub-unit vaccine Pre-clinical study [437]
Recombinant SARS-CoV-2 and RBD protein Transient expression in N. benthamiana RBD Research [438]
Recombinant SARS-CoV-2 and N protein Transient expression in N. benthamiana N protein Research [438]
N/M protein based vaccine Transient expression in N. benthamiana Recombinant SARS-CoV N and M protein Research [439]
Baiya SARS-CoV-2 Vax 1 Transient expression in N. benthamiana Protein subunit vaccine/ SARS-CoV N and M protein Pre-clinical study [440]
Nucleocapsid SARS-CoV Transient expression in N. benthamiana SARS-CoV nucleocapsid protein Pre-clinical study [441]
SARS-CoV S1 protein fusion Transient expression in N. benthamiana SARS-CoV S1-GFP fusion protein Research [442]
SARS-CoV S1 protein based vaccine Stable expression in tomato SARS-CoV S1 protein Pre-clinical study [443]

RBD - receptor binding domain (RBD), GFP - green fluorescent protein.

8.8. Stem cell therapy for COVID-19

Stem cells (SCs) have the ability to differentiate into a variety of diverse cell types in the body. Self-regenerative and differentiation ability of certain types stem cells (e.g. mesenchymal stromal cells (MSCs)) plays an important role in stimulating the regeneration of alveolar epithelial type II cells by secreting vascular endothelial and hepatocyte growth factors [444], [445]. Alternatively, various kinds of chemokines present at the site of Inflammation can also attract MSCs and its secretion of immunoregulatory cytokines to modify the functioning of various immunocytes such as dendritic cells, NK cells, T cells, B cells, macrophages and neutrophils [446]. Notably, the factors such as the transforming growth factor β, prostaglandin E2, Indoleamine 2,3-dioxygenase and human leukocyte antigen isoform have been recognized as the major effectors in the above-mentioned processes [447]. Thus, MSCs may provide a therapeutic alternative for patients with severe or critical COVID-19 symptoms either by repairing the lung damage or inhibiting the over-activated inflammatory response and influencing the progression of pulmonary fibrosis. The MSC therapy has been shown to minimise pulmonary lesions and limit the inflammatory response generated by influenza virus infection in both human and animal models [448], [449]. The efficacy and safety of MSC treatment in patients with acute respiratory distress syndrome (ARDS) have also been studied. The intravenous transfusion of MSCs in moderate or severe COVID-19 patients was proven to be safe and well tolerated in recent phase-1 clinical trials [451]. For the first time, the intravenous MSC therapy has improved the clinical outcome of COVID-19 patients while displaying good immunological tolerance in critically ill patients [451]. Similarly, menstrual blood-derived MSCs (MB-MSCs) were used in a clinical trial for severe patients, and it was discovered that MSC transplantation could help in treating COVID-19, particularly in ICU patients. In a recent study, the infusion of umbilical cord MSCs (UC-MSCs) into COVID-19 patients with moderate or severe disease was found to be safe in phase 2 and 3 studies with 96-week follow-up course [452]. When the influence of high dose of MSCs (upto 200 × 106 cells) and exosomes produced from allogeneic MSCs were tested for therapy efficacy, the treatment was found to be well tolerated and showed prospective improvement in some clinical measures [453]. Further, COVID-19 patients have also been treated using stem cells other than MSCs such as human embryonic stem cell-derived immunity- and matrix-regulatory cells (hESC-IMRCs) [450].

8.9. Molecular docking against 3CLpro, PLpro and RdRp

The docking score was recently assessed between 3CLpro crystalline structure (5R7Y) and medicinal plant compounds by Glide XP protocol. The top three scoring compounds are rutin, rocymosin-B and verbascoside, which are mostly extracted from various medicinal plants such as Glycyrrhiza glabra, Allium myrianthum, Anastatica hierochuntica and Marrubium vulgare. These compounds are binding to Cys-His catalytic dyad (Cys145 and His41) of 3CLpro. The plant compounds are also binding with 3CLpro amino acids' active site via hydrogen bonding and polar contributions [453], [454]. The phytochemical, verbascoside was bound to the crystal structure of the PLpro (PDB ID: 6W9C) active site [455]. The extracted luteolin-7-rutinoside from Cynara scolymus is an active compound, easily interacts with PLpro amino acid residues by hydrogen bonds, π–π stacking and negatively charged interactions. A phytochemical component of Cichorium intybus, Olea europaea and Marrubium vulgare called as caftaric acid that displayed an inhibitory efficacy against the crystal structure of RdRp with cofactors (PDB ID: 6M71). Notably, it has the docking score of −10.664 kcal mol−1 and interacts with RdRp amino acid residue by π–π stacking interactions and polar interactions. Another compound, named fenugreekine isolated from Trigonella foenum graecum, possessed a docking score of −9.894 kcal mol−1. It interacts with RdRP active site via Van der Waals interactions [456]. Similarly, hesperidin extracted from Citrus aurantium L provides a significant contribution in targeting the binding boundary between ACE2 and S protein by laying on the RBD surface middle shallow pit. By superimposing ACE2-RBD complex to hesperidin-RBD complex, hesperidin may inhibit the interaction of ACE2 with RBD. Hence, the plant derived components such as luteolin 7-rutinoside B, verbascoside, rocymosin rutin, caftaric acid, fenugreekine and cyanidin 3-(6′′-malonylglucoside) have shown promising potential for further drug development.

In a recent report, the antiviral potential of PF-00835231 for the inhibition of SARS-CoV-2 was investigated. The study revealed that it is the active component of the first-in-class 3CLpro-targeting regimen in clinical trials performed with 3D in vitro models of the human airway epithelium [457]. Similarly, three real-time reverse transcription-PCR (RT-PCR) assays targeting the RdRp/helicase (Hel), spike (S) and nucleocapsid (N) genes of SARS-CoV-2 have also been reported. Among these assays, the COVID-19-RdRp/Hel assay exhibited lower limit of detection in in-vitro (1.8, 50% tissue culture infective doses [TCID50]/mL with genomic RNA and 11.2 RNA copies/reaction with in vitro RNA transcripts). Notably, the RdRp-P2 assay cross-reacted with SARS-CoV in cell culture experiments. Thus, the highly sensitive and specific COVID-19-RdRp/Hel assay might help us in improving the laboratory diagnosis of COVID-19 [458]. Further molecular dynamic simulation reports may substantiate the on-going investigations on anti-SARS-CoV-2 therapeutic agent’s development from the natural origin [455].

9. Development of SARS-CoV-2 vaccines and ongoing clinical trials

Vaccines are described as substances of biological preparation contain weakened or killed agents resembling to disease-causing microorganisms or their surface protein/toxin. It can trigger the production of disease specific antibodies without prompting the disease by specifically acting as an antigen. As a consequence, it enhances the immunity against one or more viral diseases. In recent years, vaccine technology has significantly evolved for developing various kinds of vaccines ranging from cell-cultured vaccines, DNA and RNA vaccines and recombinant protein vaccines to licensed vectored vaccines for controlling and curing various deadly viral diseases [459]. Specifically, the SARS-CoV-2 spreading has created huge stress on the global population, both financially and emotionally. In normal circumstances, the average time to bring a vaccine to the international market is more than 10 years [460]. However, WHO had reduced this time duration to a matter of few months during the SARS-CoV-2 pandemic by providing the guidance to accelerate all clinical trials in short periods and simplifying the logistical procedure. Due to coordinated efforts from various governing bodies and countries across the globe, we are successful in getting many WHO approved vaccines for the treatment of SARS-CoV-2 in the end of 2020. However, the developed vaccines don't ensure safety and effectiveness against different variants of SARS-CoV-2. Further, the applicability of developed vaccines for the wider human population is also under investigation. Hence, a large scale screening on different phases of various vaccines are being performed extensively to develop a reliable, safer and effective SARS-CoV-2 vaccines for people of different age groups ranging from elders, middle age and kids to pregnant woman. In this section, vaccines prepared from different viruses (whole) or virus parts and their status in clinical trials are highlighted briefly [461].

9.1. Vaccines administered via intramuscular routes

9.1.1. Viral like particles (VLPs)

VLPs are protein mimicking structures having similar size, shape and morphology of real virus. As they lack nucleic acid/genetic property, they are noninfectious in nature. They can undergo self-assembly with different types of proteins and form chimeras, known as cVLPs. The immune system detects VLPs in the same way as real viruses, thereby by inducing immune responses [462], [463]. Upon entry into the body, they trigger B- and T-cell immune responses by simulating the antigen presenting cells via antigen presenting cell mediated activation of B- and T-cells. They are also involved in developing CD8+ cytotoxic T-cell mediated destroying of pathogen cells. One such example for licensed vaccines based on this approach is the use of human papillomavirus. At present, many SARS-CoV-2 vaccine candidates developed using VLPs are in different stages of clinical evaluation, as shown in Table 8 [464].

Table 8.

Overview of the vaccine production for SARS-CoV-2 treatment. The major companies around the world are participating in developing a vaccine for SARS-CoV-2 infection [472].

Vaccine Name Characteristics of vaccine Development status Details of clinical trials Developer Trial ID
NVX-CoV2373 Prefusion protein nanoparticle vaccine Not actively recruiting
Phase 3
Phase 1–130 participants
Phase 2b- 2665 healthy,
240 HIV positive participants
Doses - 5 μg or 25 μg
Intramuscular injection
Novavax NCT04368988
mRNA-1273 LNP encapsulated mRNA based vaccine encoding S Protein Active and not recuiting
Phase 3
Phase 1 trial (NCT04283461) – 120 participants
Phase 2 trial (NCT04405076) - 600 participants
Phase 3 (NCT04470427)
Dose levels - 50, 100, 200 and 250 μg.
Intramuscular injection
Moderna
Kaise Permanente Washington Health Research Institute
NCT04470427
COVAX19 Protein sub unit
Recombinant of covid-19 S protein with adjuvant of Advax-SM
Completed
Phase 1
40 participants
Dose – 2 μg and saline
Intramuscular injection
Vaxine Pty/Medytox NCT04453852
V591 Measles virus vector Completed
Phase 1
90 participants
Dose – 2 different doses
Intramuscular injection
Institute Pasteur NCT04497298
CVnCoV, CureVac mRNA based vaccine Active and not recruiting
Phase 1
280 participants
Doses – 2, 4, 6, 8, and 12 μg
Intramuscular injection
Curevac, CEPI NCT04449276
BCG Vaccine live-attenuated strain derived from Mycobacterium bovis Recruiting
Phase 4
700 participants
Dose - single dose in 0.1 mL saline
Intradermal injection
Andrew Dinardo
Texas A&M University
NCT04348370
VPM1002 Further development of BCG Active and not recruiting
Phase 3
1200 participants
Dose- 1 μg
Intradermal injection
University Health Network, Toronto
Vakzine Projekt Management GmbH
NCT04439045
INO-4800 DNA plasmid encoded S protein delivered by electroporation Active and not recruiting
Phase 3
120 participants
Dose – 1.0 mg
Intradermal injection
Inovio Pharmaceuticals
NCT04336410
BNT162 mRNA based vaccine Recruiting
Phase 3
Phase 1/2–200 participants
Phase 2/3–32,000 participants
Dose - 10 to 100 μg
Intramuscular injection
Pfizer, BioNTech
Multiple study sites in Europe and North America
NCT04368728
BNT162, BioNTech BioNTech mRNA vaccine Recruiting
Phase 3
200 participants
Dose – 1 to 100 μg (escalating levels of dose)
Intramuscular injection
Pharmaceuticals GmbH + Pfizer Inc.
BioNTech RNA Pharmaceuticals GmbH
NCT04380701
CoronaVac Inactivated SARS-CoV-2 Active and not recruiting
Phase 2
Phase 1–144 participants
Phase 2–600 participants
Doses – 3 or 6 μg / 0.5 mL
Intramuscular injection
Sinovac Research & Development Co., Ltd NCT04352608
Ad5-nCoV Adenovirus Type 5 Vector/Non replicating Viral Vector that express S protein Completed
Phase 1
Phase 1–108 participants
Dose – 3 different doses
Low - 5E10 vp Ad5-nCoV
middle - 1E11 vp Ad5-nCoV
high - 1.5E11vp Ad5-nCoV
Intramuscular injection
CanSino Biologics Inc. NCT04313127
RBD-Dimer Adjuvanted recombinant protein/ protein subunit Active and not recruiting
Phase 1
50 participants
Dose – 25 and 50 μg / 0.5 mL
Intramuscular injection
Anhui Zhifei Longcom Biologic Pharmacy Co., Ltd. NCT04445194
Gam-COVID-Vac Lyo, Gamaleya Non replicating viral vector/adenoviral based vaccine combined (rAd26-S + rAd5-S) expressing S protein Completed
Phase 3
Phase 1–138 participants
Dose: 2 dose levels
Intramuscular injection
Gamaleya Research Institute of Epidemiology and Microbiology, Health Ministry of the Russian Federation NCT04437875
EpiVacCorona Peptide antigens based vaccine Active and not recruiting
Phase 2
100 participants
Dose – a dose of 1 mL
Intramuscular injection
Federal Budgetary Research Institution, State Research Center of Virology and Biotechnology “Vector” NCT04527575
DNA plasmid vaccine - ZyCoV-D Electroporation to deliver S protein Active and not recruiting
Phase 2
1048 participants
Dose – 3 doses
Intramuscular injection
Cadila Healthcare Limited NCT04336410
BBV152 Whole virion based inactivated vaccine Active and not recruiting
Phase 2
755 participants
Dose – 0.5 mL
Intramuscular injection
Bharat Biotech International Limited NCT04471519
CTRI/2020/07/026300
LV-SMENP-DC Lentiviral vector with DCs modification express the synthetic minigene based on domains of selected viral proteins; administered with low antigen specific cytotoxic t lymphocytes Recruiting
Phase 2
100 participants
5E6 and 1E10 LV- DC
Subcutaneous injection
Shenzhen Geno-Immune Medical Institute NCT04276896
AZD1222 ChAdOx1 vector that expresses S protein Active and not recruiting
Phase 3
32,459 participants
Dose – 2
Intramuscular injection of 5E10 viral particles
Intramuscular injection
AstraZeneca/ Oxford University NCT04516746
ARCT-021 LNP-encapsulated self-replicating mRNA that encodes the prefusion S protein Completed
Phase 2
106 participants
1 dose in 0.5 mL
Intramuscular injection
Arcturus Therapeutics, Inc. NCT04480957
Inactivated
SARS-CoV-2 vaccine
Covid-19 inactivated vaccine Enrolling by invitation
No longer being studied
Phase 2
471 participants
Dose - 50–150 μg/0.5 mL
Intramuscular injection
Institute of Medical Biology, Chinese Academy of Medical Sciences NCT04470609

9.1.2. Protein-based vaccines

The purified recombinant proteins from different etiologic agents are most prominent candidates under the investigation for vaccines. The protein-based vaccines are formulated by using the harmless protein fragment or protein shells that mimic the SARS-CoV-2 virus to stimulate the human immune response [459]. Notably, there are several examples for recombinant protein-based vaccines being used in humans, for example, hepatitis B vaccine (HBV). As discussed in the previous sections, S protein is playing an important role in host cell receptor binding and cell membrane fusion, thus, S protein based SARS-CoV-2 vaccines might improve antibody production and virus neutralization efficacy. As a result, S protein is mostly used as a recombinant protein subunit in many cell-based systems to induce the protein expression [465]. This approach has shown better protection in immunized animal cells in in-vivo experiments. However, there is a clear risk of generation of polarized (TH2 over TH1) immune response, which is usually prevented by the use of suitable adjuvants. In this category, a saponin based matrix-M adjuvant vaccine known as Novavax has been recently reported, which has demonstrated 89% of efficacy against SARS-COV-2 in clinical trials conducted in UK [466]. It should be noted that there are many protein subunit-based vaccines in the development stage and clinical trials, however none of them are authorized for the use [https://jamanetwork.com/journals/jama/fullarticle/2777059].

9.1.3. Viral-vector vaccines

Viral vector vaccines use replication-deficient viruses to express the genetic sequence of the antigen in host cells. The genetically modified viruses such as adenovirus and measles are used as vectors to produce SARS-CoV-2 proteins in the human body because it reduces their virulence and cannot cause disease. It is divided into two types of vectors: one type is still replicated within human cells, and another one is disabled to divide due to the absence of key genes. These types of genetically modified vectors stimulate antibody production in the human body [467]. Currently, two viral vector vaccines, namely adenovirus serotype 26 vector vaccine (Ad26.CoV2.S) and chimpanzee adenovirus vector (ChAdOX), have shown promising results and considered as potential candidates for the treatment of SARS-CoV-2 [468].

9.1.4. Nucleic-acid vaccines

Nucleic acids such as DNA and RNA are inserted into human cells as a genetic instruction, which can induce an immune response [467]. DNA-based vaccines are non-replicating, non-infectious and provide long term immunogenicity to the host. Further, they are stable, less expensive, prepared in short time duration and easily getting degraded in host models. However, it has poor immunogenicity when used in humans [466]. Due to this, RNA-based vaccines are preferred over DNA-based vaccines. Often, mRNA based vaccines are directly injected into the host cell and allowed for translation in the cytoplasm. Currently, two types of mRNA based vaccines are established, namely self-amplifying mRNA and non-amplifying mRNA based vaccines. The self-amplifying mRNA-based vaccine technology has the capability to ramp-up vaccine production to meet the increase in demand for vaccines [469], [470], [471].

9.1.5. Inactivated vaccine

They are generally produced either by completely killing or inactivating the pathogen. When they are injected into the host, they induce protective antigen against epitopes that are present on the surface of real virus. However, these vaccines tend to produce the weaker immune response, thus it requires the adjuvants to provide an effective immune response [472], [473]. While the inactivated polio vaccine is the better example for the completely killed (whole) pathogen, the tetanus and diphtheria vaccination is the example for subunit formulation [474]. Sinopharm and Sinovac are among the manufacturers farthest along in the development of this type of vaccine. They have also successfully completed phase 3 clinical trials for their vaccines and obtained international authorization for emergency use [466].

9.2. Intranasal vaccination

Nasal vaccination is considered to be a reliable and influencing method to prevent SARS-CoV-2 infection as the viral invasion mainly occurs via nasal mucosa. Among the available vaccines, adenovirus vector-based vaccines are considered as reliable vectors of antigens to hosts owing to its potential to induce both adaptive and innate responses [476]. They are safer, less expensive and produced in higher quantities to meet the demands [475]. Alternatively, SARS-COV-2-M2sr categorized under M2SR is another type of vaccine which is produced upon deletion of M2 gene that activates the immune response (innate and cellular) in the host [481], [482], [483]. The advantage of such vaccine is that it delivers multiple antigen targets to the immune system without producing progeny virus. Furthermore, DelNS1-nCoV-RBD LAIV categorized under live attenuated vaccine is another type of vaccine comprises of H1N1, H3N2 and B with genetic segments of S protein of SARS CoV-2, hence mimicking the infection to induce the immune response [484], [485]. Mv-014-212 vaccine candidate developed by meissa vaccines is also undergoing phase 1 clinical trials. An inhaled therapy, named as CROWNase, developed by Illinois Institute of Technology, Chicago, USA is currently in preclinical trial. The S protein of SARS-CoV-2 makes the virus to interact with the hACE2 and causes infection. The S protein has human derived molecule coating which helps in overcoming the immune system of human being which leads to spread of infection easily. CROWNase works by removing the human derived molecule coating [486] . Similarly, CovMOV developed by Intravacc embeds viral S antigen in bacterial outer membrane vesicles and is in the preclinical trial [488]. AuraVax therapeutics has also proposed a liposomal stimulator of interferon genes or STING agonist to use as an adjuvant in vaccine for SARS-CoV-2 [489]. Notably, it triggers mucosal immune response and provides better protection against the virus. The list of aforementioned vaccines along with their current state is given in Table 9.

Table 9.

Recently developed intranasal vaccines and their respective current stages.

Vaccine Name Type Developer Details of clinical trials Ref
ChAdOx1 nCoV-19 Adenovirus vector-based vaccines University of Oxford in collaboration with AstraZeneca Phase I trial [476]
ChAd-SARS-CoV-2-S Adenovirus vector-based vaccines Washington University school of medicine in St. Louis, USA Phase I trial [477], [478]
Ad5-nCoV and ZF2001 vaccines Adenovirus vector-based vaccines CanSino Biologics Inc., China Phase II trial [479]
AdCOVID Adenovirus vector-based vaccines USA-based Altimmune Phase I trial [480]
SARS-COV2-M2sr M2SR (single replication) vaccines The University of Wisconsin-Madison in collaboration with FluGen and Bharat Biotech To initiate Preclinical trial [481], [482], [483]
DelNS1-nCoV-RBD LAIV Live-attenuated vaccines Beijing Wantai Biological Pharmacy Enterprise, USA Phase I trial [484], [485]
Mv-014-212 Live-attenuated vaccines Meissa Vaccines, Inc., USA Phase I trial [486]
CROWNase Subunit vaccines Illinois Institute of Technology, Chicago, USA Preclinical trial [487]
CovOMV Subunit vaccines Intravacc, Netherlands Preclinical trial [488]
STINGa-liposomes Nanoparticle-based subunit vaccines AuraVax Therapeutics, USA Preclinical trial [489]

10. Challenges in vaccines development and commercialization

The introduction of the vaccine into the human body is described as vaccination that protects the body from specific infection by developing the resistance and strengthening the immune system against pathogens [490]. It is considered as a harmless and effective route to control or kill pathogens by triggering the immune cells to produce antibodies that react against the exposed disease. The immunization process saves several millions of people from more than 20 life-threatening diseases and makes them live longer and healthier by building resistance against specific diseases [491]. Up to 180 vaccines including inactivated or live-virus vaccines, vectored vaccines, recombinant protein vaccines, DNA and RNA vaccines have been developed against SARS-CoV-2. Most vaccine designs target the receptor binding proteins and membrane fusion process because of their key responsibilities in SARS-CoV-2 infection and pathogenesis. These events are mainly depending on the large viral surface protein, namely S protein. Specifically, most vaccines target S proteins or its domain protein, including RBD, to avert its attachment to the host cell surface protein to neutralize the SARS-CoV-2 virus. Interestingly, the greatness of the vaccines towards the SARS-CoV-2 S protein is somewhat different when compared to other vaccines. After confirming their activity and safety profiles in in-vitro experiments, the vaccines are being tested on human volunteers in a phased manner in clinical trials by increasing the size of volunteer population to investigate their safety in human population. Later, they are being administered to health care workers, older adults and people with underlying disease conditions such as diabetes and heart disease [492]. It could also induce life-threatening allergies for people who are in treatment for chemotherapy and chronic illnesses as it severely affects the immune system. Interestingly, it is very safe and effective in people with underlying conditions such as liver or kidney disease, asthma, diabetes and hypertension. The vaccine ingredients may stabilize and control these diseases [493]. However, these impacts may differ for every vaccine.

The major ingredients of vaccines such as weakened or killed antigen, adjuvants, preservatives and stabilizers play a crucial role in encountering the virus and boosting the host immune response. For instance, the added preservatives ensure a vaccine stays effective, and stabilizers are protecting the vaccine during storage and transportation [494]. After satisfying the potential benefit of immunization, the approval for commercial use of vaccines will be obtained from study investigators, regulatory agencies and overseeing ethical committees. The time duration from preclinical trial to manufacturing in large scale often takes over a decade. However, the clinical trials of SARS-CoV-2 vaccines are performed in parallel for different phases to complete them in a short period. Common challenges faced by newly developed SARS-CoV-2 vaccines are listed in the following sections.

10.1. Ensuring the safety and effectiveness of vaccines

Most vaccines for SARS-CoV-2 disease have been developed in a short time span (within 10–16 months) in order to control the rate of infection and casualties around the globe. Hence, the concerns over the safety, effectiveness and side effects of newly developed vaccines need to be addressed in detail [495]. For example, the Strategic Advisory Group of Experts (SAGE) announced that the developed Pfizer-BioNTech COVID-19 mRNA vaccine is safe and effective to people. As of 14th October 2021, 23 vaccines have been approved for emergency use across the globe, as shown in Table 10. However, the vaccination is not recommended for some specific populations and it may be either due to lack of supply or limited data and contraindications [496]. Further, a certain fraction of population who had received the COVID 19 vaccine developed severe headache, abdominal pain, leg pain, shortness of breath and severe type of blood clot called cerebral venous sinus thrombosis (or CVST) combined with low levels of blood platelets (thrombocytopenia) [497]. As a result, the regular medical assessments and post-approval clinical studies are urgently needed to confirm their safety and effectiveness.

Table 10.

List of approved vaccines across the globe [498].

Vaccine name Vaccine type Number of countries approved (Nos.) Developer Ongoing trials in different number of countries
mRNA-1273 RNA 76 Moderna 30 trials in 2 countries
BNT162b2 RNA 101 Pfizer/BioNtech 37 trials in 2 countries
TAK-919 RNA 1 Takeda (Moderna formulation) 2 trials in 1 country
ZyCoV-D DNA 1 Zydus Cadila 5 trials in 1 country
Ad26.COV2·S Non Replicating Viral Vector 71 Janssen (Johnson & Johnson) 13 trials in 2 countries
AZD1222 Non Replicating Viral Vector 123 Oxford/AstraZeneca 44 trials in 2 countries
Covishield Non Replicating Viral Vector 46 Serum Institute of India
(Oxford/AstraZeneca formulation)
2 trials in 1 country
Ad5-nCoV Non Replicating Viral Vector 9 CanSino 11 trials in 6 countries
Sputnik Light Non Replicating Viral Vector 18 Gamaleya 4 trials in 2 countries
Sputnik V Non Replicating Viral Vector 72 Gamaleya 21 trials in 7 countries
BBIBP-CorV (Vero Cells) Inactivated 66 Sinopharm (Beijing) 11 trials in 9 countries
CoronaVac Inactivated 40 Sinovac 22 trials in 1 country
Inactivated (Vero Cells) Inactivated 2 Sinopharm (Wuhan) 8 trials in 5 countries
COVID-19 Inactivated Vaccine Inactivated 11 Shifa Pharmed Industrial Co 4 trials in 1 country
QazVac Inactivated 2 Kazakhstan RIBSP 3 trials in 1 country
SARS-CoV-2 Vaccine (Vero Cells) Inactivated 1 Minhai Biotechnology Co 5 trials in 1 country
KoviVac Inactivated 1 Chumakov Center 2 trials in 1 country
Covaxin Inactivated 9 Bharat Biotech 7 trials in 1 country
ZF2001 Protein subunit 3 Anhui Zhifei Longcom 7 trials in 5 countries
CIGB-66 Protein subunit 4 Center for Genetic Engineering and Biotechnology (CIGB) 5 trials in 1 country
EpiVacCorona Protein subunit 2 FBRI 3 trials in 1 country
MVC-COV1901 Protein subunit 1 Medigen 7 trials in 2 countries
COVAX-19 Protein subunit 1 Vaxine/CinnaGen Co. 4 trials in 2 countries

10.2. Long-term protection

Given the fact that the long-time protection of vaccine from specific disease is the main thing in preventing the infections, their ability of long-term protection should be fully investigated for newly developed SARS-CoV-2 vaccines. At present, most SARS-CoV-2 vaccines are given to people in two dose regimens. As reinfection of cases raise concerns over the immunity after vaccination, the additional studies need to be performed urgently to give further direction for the people who recuperate from SARS-CoV-2 disease in order to suppress the subsequent wave of infections. A detailed investigation on reinfections (even after vaccination) and the ability of vaccines against new mutations will shed more light on long-term protection of vaccines against SARS-CoV-2 [499].

10.3. Storage and transportation of vaccines

The storage and transportation mechanisms are also pose great challenges for the commercialization of vaccines as the existing installations are not able to cope up with the demands caused by larger vaccination schedules across the globe. The prepared vaccines are packed carefully in glass vials due to its ability to withstand extreme temperatures for safe cold vaccine storage and transport globally [500]. When a vaccine is too hot or cold, it becomes less effective or even inactive. If stored at the incorrect temperature, the vaccines can be ruined or unsafe for use. Most vaccines require refrigerated storage at temperature in the range between 2 and 8 °C [501]. Some vaccines even require ultra-cold temperature from −20 to −70 °C [502]. For frozen vaccines, the storage temperature is maintained at 2–8 °C. Since the regular refrigerators cannot consistently maintain these low temperatures, specialized medical refrigerators are required for these precious products. The messenger RNA (mRNA) based vaccines (mRNA-1273-Moderna and BNT162b2-Pfizer-BioNTech) are developed using the strands of mRNA held together within lipid particles. The prepared vaccines are vulnerable to degradation at room temperature and need doses to be frozen for transportation, then thawed for the use [503]. Therefore, the concerns regarding the storage temperature could slow down the rollout of SARS-CoV-2 vaccines. Similarly, the mRNA-1273 vaccine is stored at temperature in the range between −25 and −15 °C. As it is stable and active at −20 °C, it can be stored in standard −20 °C freezers meant for hospitals and pharmacies [504]. The Indian vaccines developed can be stored at a temperature of −20 °C, hence they can be stored at normal hospital refrigerators. However, the efficacy and other temperature related data is yet to be declared officially. For instance, Pfizer-BioNTech announced that BNT162b2 vaccine displayed 90% efficacy when stored at −70 °C. Similarly, the Sputnik V vaccine liquid form must be stored at −18 °C or below to maintain its stated 92% efficacy [505].

10.4. Mass production of plastic syringes, vials and needles

Up to 275 vaccines are being developed worldwide till date and 23 of them are being currently used in different countries. As most of the countries are facing frequent waves of infections and there is no sign for the end of Covid-19 pandemic, the need for vaccines along with syringes, needles and plastic vials for effective administration and storage of vaccines is growing with each passing day. Based on the available data, the USA has ordered about 850 million of syringes and needles for their two doses of vaccination [506]. Hence, the requirement across the globe has to be taken care while a series of vaccines are being rolled out. In addition, further developments are required on the design and mechanical integrity of glass vials for efficient storage and transport of vaccines. For instance, the parameters such as mechanical integrity, resistant to breakage, chemical stability, mechanical durability, thermal stability and compact designs/dimensions are not only playing a key role in maintaining the activity of vaccines but also preventing the loss of doses during the transportation and handling of vaccines. It is worth noting that they should be stable in the temperature range between −196 °C to 121 °C while having the chemical stability to handle liquids with pH range of 3–14 [507].

10.5. Vaccine hesitancy

Worldwide acceptance of vaccine is necessary to prevent further spread of SARS CoV-2 and improving the immune system. Vaccine hesitancy is predominant in upper middle/high-income countries (UMIC) than low and middle-income countries (LMIC). As per the recent report in LMIC (Asia, Africa and South America) and UMIC (Russia and US) with 44,260 participants, the willingness of people to take vaccine were found to be 80.3, 30.4 and 64.6% for LMIC, Russia and US, respectively. Even though large-scale production and availability of vaccine is limited in LMIC, the willingness to take vaccine seems to be comparatively higher. The personal interest for protection against SARS CoV-2 is the main reason for such huge difference between LMIC and high income countries. However, the hesitance towards vaccine is the fear of having side effects post vaccination. It is worth to mention that apart from fearful thoughts, misleading information from untrusted web sources could be the another reason of vaccine hesitancy [508]. The probable solution is seeking advice from the health expert even for very common doubts and not trusting any untrusted web-sources. It also seems to be the common practice of assuming the same side effects of post vaccination of one with other vaccinated individual. However, it is important to know that the side effects may vary among different individuals. The willingness to take the vaccine with trust is the key determinant in the successful vaccination campaign [509].

10.6. Global vaccine distribution plans (Covax)

COVID-19 vaccines global access (COVAX) acts as a pillar of the Access to COVID-19 Tools (ACT) Accelerator. ACT accelerator collaboration is done globally for the development, production and equitable access to SARS CoV-2 diagnostic kit, treatment and vaccines. Gavi, WHO and Coalition for Epidemic Preparedness Innovations (CEPI) in collaboration with COVAX and UNICEF (Key Delivery Partner) uphold the responsibility for the development, manufacturing of vaccine and equitable access for every country [510], [511]. WHO had estimated that US$1.7 billion was required in 2020 for SARS CoV-2 response in addition to government need for their respective national action plan and strategic preparedness and response plans (SPRP). The SPRP 2021 was approximated to be US$ 1.96 billion which will allow WHO to continuously support countries in terms of reducing transmission, countering misinformation, morbidity rates, equitable access and protecting the riskier groups. CEPI also focuses on accelerating the development of vaccines against emerging infectious threats and enabling equitable access to people. Similarly, UNICEF procures 2 billion doses of vaccine annually for approximately 100 countries. Further, Gavi, UNICEF and WHO are also heavily investing on reliable infrastructure, technical support and delivery of vaccines [512], [513], [514].

11. Future outlooks

Currently, SARS-CoV-2 is spreading rapidly as the third severely contagious human disease and has caused serious threat globally. Beyond doubts, it has now been widely accepted that the prevention of public gatherings and following quarantine strategies and standard operating procedures is the way to move forward for controlling SARS-CoV-2 infections. As it has coexisted with people for a long time, it has developed a niche in human beings. As a result, continuous monitoring of the gene alterations in new SARS-CoV-2 infections is required to promptly recognize any deletion or insertion in the genomic sequence. It will shed more light not only on the phenotypic related disease severity but also the development of new vaccines and medicines with required modifications in the structure. As the originating source of SARS-CoV-2 and the mode of cross-species infection is yet to be discovered, a detailed investigation needs to be done on the origination and transmission pathways of SARS-CoV-2 via the extra-respiratory and respiratory routes for further understanding on the rapid spreading of the virus. Further, the investigations on the molecular mechanisms of viral-host interactions and pathogenesis along with the molecular modulation of the host immune response during SARS-CoV-2 infection is urgently needed to determine the outcomes of medical therapies. These details will also help in the identification of specific biomarkers to describe the immune correlation of the disease. We believe that any support comes from theoretical studies can substantially improve the understanding in the above mentioned investigations and positively contribute to the development of different medical therapies.

Recently, various diagnostic methodologies have emerged for effective identification, monitoring and control of SARS-CoV-2 infections. However, most of the detection techniques are still relying on PCR and antibody based techniques that help in identifying and isolating the virus strains. Notably, other detection techniques such as RT-LAMP, CRISPR, LAMP, electrochemical sensing etc. are currently under development, and further experiments on sensitivity and repeatability are urgently needed. Further, the above mentioned methods have the capability to handle only the nasopharyngeal samples that limit their potential in the detection of SARS-CoV-2. As a result, any breakthrough in directly using blood and oral samples in the upcoming technologies may improve the reliability, sensitivity and reproducibility for SARS-CoV-2 detection. Various vaccines and therapeutic agents are being developed to fight against the SARS-CoV-2. They are most widely used to overcome the dysfunctional immune responses and suppress the viral infection to synergize and block pathologies at several steps. However, continuously evolving SARS-CoV-2 and mutating itself into different forms and safety concerns related to vaccines available in the markets demands alternative vaccines with good safety profiles. Hence, a rapid development of therapeutic agents/vaccines especially for targeting 3CLpro, RdRp and PLpro in combination with high-throughput and virtual screening tools could help us in developing effective and safer anti-SARS-CoV-2 therapy. Finally, a long-term efficient vaccine development against SARS-CoV-2 is also one of the important task as it provides an active acquired immunity to the host. At the same time, the correlation between the disease severity and immune dysfunction in patients with SARS-CoV-2, should serve as a note of caution in vaccine development and evaluation. At present, it seems that we are in the middle of never ending war against SARS-CoV-2.

Acknowledgements

We acknowledge financial support from the Department of Science and Technology (DST)-Science and Engineering Research Board (SERB), Government of India (File Nos. YSS/2015/000771 and CRG/2020/004175).

References

  • 1.Rothan H.A., Byrareddy S.N. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J. Autoimmun. May 2020;109 doi: 10.1016/j.jaut.2020.102433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cascella M., Rajnik M., Aleem A., Dulebohn S.C., Di Napoli R. Features, evaluation, and treatment of coronavirus (COVID-19) StatPearls. 2021 https://www.ncbi.nlm.nih.gov/books/NBK554776/ [PubMed] [Google Scholar]
  • 3.Li H., Liu S.-M., Yu X.-H., Tang S.-L., Tang C.-K. Coronavirus disease 2019 (COVID-19): current status and future perspectives. Int. J. Antimicrob. Agents. May 2020;55(5) doi: 10.1016/j.ijantimicag.2020.105951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yin Y., Wunderink R.G. MERS, SARS and other coronaviruses as causes of pneumonia. Respirology. Feb. 2018;23(2):130–137. doi: 10.1111/resp.13196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Letko M., Marzi A., Munster V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat. Microbiol. 2020;5(4):562–569. doi: 10.1038/s41564-020-0688-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pal M., Berhanu G., Desalegn C., Kandi V. Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2): an update. Cureus. 2020;12(3):1–9. doi: 10.7759/cureus.7423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Narayanan K., Maeda A., Maeda J., Makino S. Characterization of the coronavirus M protein and nucleocapsid interaction in infected cells. J. Virol. Sep. 2000;74(17):8127–8134. doi: 10.1128/jvi.74.17.8127-8134.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Alluwaimi A.M., Alshubaith I.H., Al-Ali A.M., Abohelaika S. The coronaviruses of animals and birds: their zoonosis, vaccines, and models for SARS-CoV and SARS-CoV2. Front. Vet. Sci. Sep. 2020;7 doi: 10.3389/fvets.2020.582287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Belhaouari D.Brahim, A. Fontanini, J.-P. Baudoin, G. Haddad, M. Le Bideau, J.Y. Bou Khalil, D. Raoult, B. La Scola. The strengths of scanning electron microscopy in deciphering SARS-CoV-2 infectious cycle. Front. Microbiol. 2020;11 doi: 10.3389/fmicb.2020.02014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ye Z.-W., Yuan S., Yuen K.-S., Fung S.-Y., Chan C.-P., Jin D.-Y. Zoonotic origins of human coronaviruses. Int. J. Biol. Sci. 2020;16(10):1686–1697. doi: 10.7150/ijbs.45472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wu Y., W. Ho, Y. Huang, D.-Y. Jin, S. Li, S.-L. Liu, X. Liu, J Qiu, Y. Sang, Q. Wang. SARS-CoV-2 is an appropriate name for the new coronavirus. Lancet (London, England) 2020;395(10228):949–950. doi: 10.1016/S0140-6736(20)30557-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cui J., Li F., Shi Z.-L. Origin and evolution of pathogenic coronaviruses. Nat. Rev. Microbiol. 2019;17(3):181–192. doi: 10.1038/s41579-018-0118-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Khan I., Haleem A., Javaid M. Analysing COVID-19 pandemic through cases, deaths, and recoveries. J. Oral Biol. Craniofacial Res. 2020;10(4):450–469. doi: 10.1016/j.jobcr.2020.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zheng J. SARS-CoV-2: an emerging coronavirus that causes a global threat. Int. J. Biol. Sci. Mar. 2020;16(10):1678–1685. doi: 10.7150/ijbs.45053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lai C.-C., Shih T.-P., Ko W.-C., Tang H.-J., Hsueh P.-R. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): the epidemic and the challenges. Int. J. Antimicrob. Agents. Mar. 2020;55(3) doi: 10.1016/j.ijantimicag.2020.105924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hajifathalian K., et al. SARS-COV-2 infection (coronavirus disease 2019) for the gastrointestinal consultant. World J. Gastroenterol. Apr. 2020;26(14):1546–1553. doi: 10.3748/wjg.v26.i14.1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Peeri N.C., et al. The SARS, MERS and novel coronavirus (COVID-19) epidemics, the newest and biggest global health threats: what lessons have we learned? Int. J. Epidemiol. Jun. 2020;49(3):717–726. doi: 10.1093/ije/dyaa033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jayaweera M., Perera H., Gunawardana B., Manatunge J. Transmission of COVID-19 virus by droplets and aerosols: a critical review on the unresolved dichotomy. Environ. Res. Sep. 2020;188 doi: 10.1016/j.envres.2020.109819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Seminara G., Carli B., Forni G., Fuzzi S., Mazzino A., Rinaldo A. Biological fluid dynamics of airborne COVID-19 infection. Rend. Lincei. Sci. Fis. e Nat. Aug. 2020:1–33. doi: 10.1007/s12210-020-00938-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Heller L., Mota C.R., Greco D.B. COVID-19 faecal-oral transmission: are we asking the right questions? Sci. Total Environ. Aug. 2020;729 doi: 10.1016/j.scitotenv.2020.138919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hussain A., Bhowmik B., do Vale Moreira N.C. COVID-19 and diabetes: Knowledge in progress. Diabetes Res. Clin. Pract. Apr. 2020;162:108142. doi: 10.1016/j.diabres.2020.108142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Patrikar S.R., A. Kotwal, V. Bhatti, A. Banerjee, K. Chatterjee, R. Kunte, M. Tambe. Incubation period and reproduction number for novel coronavirus (COVID-19) infections in India. medRxiv. 2020 doi: 10.1101/2020.06.27.20141424. [DOI] [PubMed] [Google Scholar]
  • 23.Al-Khalifa K.S. Pandemic preparedness of dentists against coronavirus disease: A Saudi Arabian experience. PLoS One. 2020;15(8 August):1–13. doi: 10.1371/journal.pone.0237630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bauchner H., Fontanarosa P.B., Livingston E.H. Conserving supply of personal protective equipment—a call for ideas. JAMA. May 2020;323(19):1911. doi: 10.1001/jama.2020.4770. [DOI] [PubMed] [Google Scholar]
  • 25.Fung T.S., Liu D.X. Human coronavirus: host-pathogen interaction. Annu. Rev. Microbiol. Sep. 2019;73:529–557. doi: 10.1146/annurev-micro-020518-115759. [DOI] [PubMed] [Google Scholar]
  • 26.Stannard C.J., Abbiss J.S., Wood J.M. Efficiency of treatments involving ultraviolet irradiation for decontaminating packaging Board of Different Surface Compositions. J. Food Prot. Sep. 1985;48(9):786–789. doi: 10.4315/0362-028X-48.9.786. [DOI] [PubMed] [Google Scholar]
  • 27.Fejer S.N. Minimalistic coarse-grained modeling of viral capsid assembly. Prog. Mol. Biol. Transl. Sci. 2020;170:405–434. doi: 10.1016/bs.pmbts.2019.12.003. [DOI] [PubMed] [Google Scholar]
  • 28.Sun L., et al. Design, synthesis, and mechanism study of benzenesulfonamide-containing phenylalanine derivatives as novel HIV-1 capsid inhibitors with improved antiviral activities. J. Med. Chem. May 2020;63(9):4790–4810. doi: 10.1021/acs.jmedchem.0c00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Faris M., Jahrami H., Bragazzi N.L., Buheji M., Naja F. Dietary intakes as potential risk factors for COVID-19 mortality: a multicountry ecological Bayesian Regression Analysis. SSRN Electron. J. 2020 doi: 10.2139/ssrn.3658660. [DOI] [Google Scholar]
  • 30.Cadnum J.L., et al. Evaluation of an electrostatic spray disinfectant technology for rapid decontamination of portable equipment and large open areas in the era of SARS-CoV-2. Am. J. Infect. Control. Aug. 2020;48(8):951–954. doi: 10.1016/j.ajic.2020.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hu B., et al. Discovery of a rich gene pool of bat SARS-related coronaviruses provides new insights into the origin of SARS coronavirus. PLoS Pathog. 2017;13(11):e1006698. doi: 10.1371/journal.ppat.1006698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chan J.F.-W., et al. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg. Microbes Infect. Jan. 2020;9(1):221–236. doi: 10.1080/22221751.2020.1719902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Graham R.L., Sparks J.S., Eckerle L.D., Sims A.C., Denison M.R. SARS coronavirus replicase proteins in pathogenesis. Virus Res. Apr. 2008;133(1):88–100. doi: 10.1016/j.virusres.2007.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wang Y.T., et al. Spiking pandemic potential: structural and immunological aspects of SARS-CoV-2. Trends Microbiol. 2020;28(8):605–618. doi: 10.1016/j.tim.2020.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ulferts R., Imbert I., Canard B., Ziebuhr J. Expression and functions of SARS coronavirus replicative proteins. Mol. Biol. SARS-Coronavirus. 2010:75–98. doi: 10.1007/978-3-642-03683-5_6. [DOI] [Google Scholar]
  • 36.Ullrich S., Nitsche C. The SARS-CoV-2 main protease as drug target. Bioorg. Med. Chem. Lett. Sep. 2020;30(17) doi: 10.1016/j.bmcl.2020.127377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Michalska K., Y. Kim, R. Jedrzejczak, N.I. Maltseva, L. Stols, M. Endres, A. Joachimiak. Crystal structures of SARS-CoV-2 ADP-ribose phosphatase (ADRP): from the apo form to ligand complexes. bioRxiv. 2020;7:814–824. doi: 10.1101/2020.05.14.096081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Khailany R.A., Safdar M., Ozaslan M. Genomic characterization of a novel SARS-CoV-2. Gene Rep. Jun. 2020;19 doi: 10.1016/j.genrep.2020.100682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Mousavizadeh L., Ghasemi S. Genotype and phenotype of COVID-19: their roles in pathogenesis. J. Microbiol. Immunol. Infect. 2020;54(2):159–163. doi: 10.1016/j.jmii.2020.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Báez-Santos Y.M., John S.E.St, Mesecar A.D. The SARS-coronavirus papain-like protease: structure, function and inhibition by designed antiviral compounds. Antiviral Res. 2015;115:21–38. doi: 10.1016/j.antiviral.2014.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wu C., Y. Liu, Y. Yang, P. Zhang, W. Zhong, Y. Wang, Q. Wang, Y. Xu, M. Li, X. Li, M. Zheng, L. Chen, H. Li. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B. May 2020;10(5):766–788. doi: 10.1016/j.apsb.2020.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Delmas B., Laude H. Assembly of coronavirus spike protein into trimers and its role in epitope expression. J. Virol. Nov. 1990;64(11):5367–5375. doi: 10.1128/JVI.64.11.5367-5375.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li F. Structure, function, and evolution of coronavirus spike proteins. Annu. Rev. Virol. Sep. 2016;3(1):237–261. doi: 10.1146/annurev-virology-110615-042301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Walls A.C., Park Y.-J., Tortorici M.A., Wall A., McGuire A.T., Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. Apr. 2020;181(2):281–292. doi: 10.1016/j.cell.2020.02.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhai Y., F. Sun, X. Li, H. Pang, X. Xu, M. Bartlam, Z. Rao. Insights into SARS-CoV transcription and replication from the structure of the nsp7-nsp8 hexadecamer. Nat. Struct. Mol. Biol. Nov. 2005;12(11):980–986. doi: 10.1038/nsmb999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tai W., L. He, X. Zhang, J. Pu, D. Voronin, S. Jiang, Y. Zhou, L. Du. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020;17(6):613–620. doi: 10.1038/s41423-020-0400-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Neuman B.W., et al. A structural analysis of M protein in coronavirus assembly and morphology. J. Struct. Biol. 2011;174(1):11–22. doi: 10.1016/j.jsb.2010.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nieto-Torres J.L., et al. Subcellular location and topology of severe acute respiratory syndrome coronavirus envelope protein. Virology. Jul. 2011;415(2):69–82. doi: 10.1016/j.virol.2011.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Astuti I., Ysrafil Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): an overview of viral structure and host response. Diabetes Metab. Syndr. 2020;14(4):407–412. doi: 10.1016/j.dsx.2020.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zeng W., W. Zeng, G. Liu, H. Ma, D. Zhao, Y. Yang, M. Liu, A. Mohammed, C. Zhao, Y. Yang, J. Xie. Biochemical characterization of SARS-CoV-2 nucleocapsid protein. Biochem. Biophys. Res. Commun. 2020;527(3):618–623. doi: 10.1016/j.bbrc.2020.04.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gildenhuys S. Expanding our understanding of the role polyprotein conformation plays in the coronavirus life cycle. Biochem. J. Apr. 2020;477(8):1479–1482. doi: 10.1042/BCJ20200223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Fehr A.R., S. Perlman, H.J. Maier, E. Bickerton, P. Britton. Coronaviruses: an overview of their replication and pathogenesis. Methods Mol. Biol. 2015;1282:1–23. doi: 10.1007/978-1-4939-2438-7_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kopecky-Bromberg S.A., Martínez-Sobrido L., Frieman M., Baric R.A., Palese P. Severe acute respiratory syndrome coronavirus open reading frame (ORF) 3b, ORF 6, and nucleocapsid proteins function as interferon antagonists. J. Virol. Jan. 2007;81(2):548–557. doi: 10.1128/JVI.01782-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.He F., Deng Y., Li W. Coronavirus disease 2019: what we know? J. Med. Virol. 2020;92(7):719–725. doi: 10.1002/jmv.25766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kirchdoerfer R.N., Ward A.B. Structure of the SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-factors. Nat. Commun. 2019;10(1):2342. doi: 10.1038/s41467-019-10280-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Q. Peng, R. Peng, B. Yuan, J. Zhao, M. Wang, X. Wang, Q. Wang, Y. Sun, Z. Fan, J. Qi. Structural and biochemical characterization of the nsp12-nsp7-nsp8 Core polymerase complex from SARS-CoV-2. Cell Rep. 2020;31(11) doi: 10.1016/j.celrep.2020.107774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.I. Imbert, J. Guillemot, J. Bourhis, C. Bussetta, B. Coutard, M. Egloff, F. Ferron, A.E. Gorbalenya, B. Canard. A second, non-canonical RNA-dependent RNA polymerase in SARS coronavirus. EMBO J. Oct. 2006;25(20):4933–4942. doi: 10.1038/sj.emboj.7601368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.M.-P. Egloff, F. Ferron, V. Campanacci, S. Longhi, C. Rancurel, H. Dutartre, E.J. Snijder, A.E. Gorbalenya, C. Cambillau, B. Canard. The severe acute respiratory syndrome-coronavirus replicative protein nsp9 is a single-stranded RNA-binding subunit unique in the RNA virus world. Proc. Natl. Acad. Sci. U. S. A. 2004;101(11):3792–3796. doi: 10.1073/pnas.0307877101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.E. Decroly, C. Debarnot, F. Ferron, M. Bouvet, B. Coutard, I. Imbert, L. Gluais, N. Papageorgiou, A. Sharff, G. Bricogne. Crystal structure and functional analysis of the SARS-coronavirus RNA cap 2′-o-methyltransferase nsp10/nsp16 complex. PLoS Pathog. 2011;7(5) doi: 10.1371/journal.ppat.1002059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Xu X., Liu Y., Weiss S., Arnold E., Sarafianos S.G., Ding J. Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug design. Nucleic Acids Res. 2003;31(24):7117–7130. doi: 10.1093/nar/gkg916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Malle L. A map of SARS-CoV-2 and host cell interactions. Nat. Rev. Immunol. 2020;20(6):351. doi: 10.1038/s41577-020-0318-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.R. Züst, L. Cervantes-Barragan, M. Habjan, R. Maier, B.W. Neuman, J. Ziebuhr, K.J. Szretter, S.C. Baker, W. Barchet, M.S. Diamond. Ribose 2′-O-methylation provides a molecular signature for the distinction of self and non-self mRNA dependent on the RNA sensor Mda5. Nat. Immunol. 2011;12(2):137–143. doi: 10.1038/ni.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Tan Y.-J., Lim S.G., Hong W. Characterization of viral proteins encoded by the SARS-coronavirus genome. Antivir. Res. 2005;65(2):69–78. doi: 10.1016/j.antiviral.2004.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.A.A.T. Naqvi, K. Fatima, T. Mohammad, U. Fatima, I.K. Singh, A. Singh, S.M. Atif, G. Hariprasad, G.M. Hasan, M.I. Hassan. Insights into SARS-CoV-2 genome, structure, evolution, pathogenesis and therapies: Structural genomics approach. Biochim. Biophys. Acta Mol. Basis Dis. 2020;1866(10) doi: 10.1016/j.bbadis.2020.165878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Huang Y., Yang C., Xu X., Xu W., Liu S. Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID-19. Acta Pharmacol. Sin. 2020;41(9):1141–1149. doi: 10.1038/s41401-020-0485-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wan Y., Shang J., Graham R., Baric R.S., Li F. Receptor recognition by the novel coronavirus from Wuhan: an analysis based on decade-long structural studies of SARS coronavirus. J. Virol. 2020;94(7) doi: 10.1128/JVI.00127-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Y.X. Gue, R. Kanji, V. Markides, D.A. Gorog. Angiotensin converting enzyme 2 may mediate disease severity in COVID-19. Am. J. Cardiol. 2020;130:161–162. doi: 10.1016/j.amjcard.2020.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.N.K. Dutta, K. Mazumdar, J.T. Gordy. The nucleocapsid protein of SARS-CoV-2: a target for vaccine development. J. Virol. 2020;94(13) doi: 10.1128/JVI.00647-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.M.A. Shereen, S. Khan, A. Kazmi, N. Bashir, R. Siddique. COVID-19 infection: origin, transmission, and characteristics of human coronaviruses. J. Adv. Res. 2020;24:91–98. doi: 10.1016/j.jare.2020.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.S. Bertram, I. Glowacka, M.A. Müller, H. Lavender, K. Gnirss, I. Nehlmeier, D. Niemeyer, Y. He, G. Simmons, C. Drosten. Cleavage and activation of the severe acute respiratory syndrome coronavirus spike protein by human airway trypsin-like protease. J. Virol. 2011;85(24):13363–13372. doi: 10.1128/JVI.05300-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.J. Shang, Y. Wan, C. Luo, G. Ye, Q. Geng, A. Auerbach, F. Li. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. U. S. A. 2020;117(21):11727–11734. doi: 10.1073/pnas.2003138117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Artika I.M., Dewantari A.K., Wiyatno A. Molecular biology of coronaviruses: current knowledge. Heliyon. 2020;6(8) doi: 10.1016/j.heliyon.2020.e04743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.A. Kumar, N. Garg, R. Giri. An insight into SARS-CoV-2 membrane protein interaction with spike, envelope, and nucleocapsid proteins. bioRxiv. 2020 doi: 10.1101/2020.10.30.363002. [DOI] [PubMed] [Google Scholar]
  • 74.Q. Wan, D. Song, H. Li, M. He. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Signal Transduct. Target. Ther. 2020;5:1–40. doi: 10.1038/s41392-020-00233-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Z. Wang, Y. Li, X. Yang, J. Zhao, Y. Cheng, J. Wang. Mechanism and complex roles of HSC70 in viral infections. Front. Microbiol. 2020;11:1–8. doi: 10.3389/fmicb.2020.01577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.M. Nampoothiri. Structural proteins in severe acute respiratory syndrome Coronavirus-2. Arch. Med. Res. 2020;51(6):482–491. doi: 10.1016/j.arcmed.2020.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gupta M.K., Vemula S., Donde R., Gouda G., Behera L., Vadde R. In-silico approaches to detect inhibitors of the human severe acute respiratory syndrome coronavirus envelope protein ion channel. J. Biomol. Struct. Dyn. 2020;39(7):2617–2627. doi: 10.1080/07391102.2020.1751300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.C. McClenaghan, A. Hanson, S.J. Lee, C.G. Nichols. Coronavirus proteins as ion channels: current and potential research. Front. Immunol. 2020;11:1–11. doi: 10.3389/fimmu.2020.573339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.J.K. Stodola, G. Dubois, A. Le Coupanec, M. Desforges, P.J. Talbot. The OC43 human coronavirus envelope protein is critical for infectious virus production and propagation in neuronal cells and is a determinant of neurovirulence and CNS pathology. Virology. 2018;515:134–149. doi: 10.1016/j.virol.2017.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kuo L., Hurst-Hess K.R., Koetzner C.A., Masters P.S. Analyses of Coronavirus Assembly Interactions with Interspecies Membrane and Nucleocapsid Protein Chimeras. J. Virol. 2016;90(9):4357–4368. doi: 10.1128/JVI.03212-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.S. Mukherjee, D. Bhattacharyya, A. Bhunia. Host-membrane interacting interface of the SARS coronavirus envelope protein: immense functional potential of C-terminal domain. Biophys. Chem. 2020;266 doi: 10.1016/j.bpc.2020.106452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Surya W., Li Y., Torres J. Structural model of the SARS coronavirus E channel in LMPG micelles. Biochim. Biophys. Acta - Biomembr. 2018;1860(6):1309–1317. doi: 10.1016/j.bbamem.2018.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lin S.-M., et al. Structure-based stabilization of non-native protein-protein interactions of coronavirus nucleocapsid proteins in antiviral drug design. J. Med. Chem. Mar. 2020;63(6):3131–3141. doi: 10.1021/acs.jmedchem.9b01913. [DOI] [PubMed] [Google Scholar]
  • 84.Mariano G., Farthing R.J., Lale-farjat S.L.M. Vol. 7. December, 2020. Structural Characterization of SARS-CoV-2 : Where We Are , and Where We Need to Be. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen Y., Liu Q., Guo D. Emerging coronaviruses: genome structure, replication, and pathogenesis. J. Med. Virol. Apr. 2020;92(4):418–423. doi: 10.1002/jmv.25681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tian X., et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg. Microbes Infect. 2020;9(1):382–385. doi: 10.1080/22221751.2020.1729069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kang S., et al. Crystal structure of SARS-CoV-2 nucleocapsid protein RNA binding domain reveals potential unique drug targeting sites. Acta Pharm. Sin. B. 2020;10(7):1228–1238. doi: 10.1016/j.apsb.2020.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yin C. Genotyping coronavirus SARS-CoV-2: methods and implications. Genomics. Sep. 2020;112(5):3588–3596. doi: 10.1016/j.ygeno.2020.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Siu K.-L. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019;33(8):8865–8877. doi: 10.1096/fj.201802418R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hass E.P., Zappulla D.C. A CRISPR-based Yeast Two-hybrid System for Investigating RNA-protein Interactions. bioRxiv. 2017;410:139600. doi: 10.1101/139600. [DOI] [Google Scholar]
  • 91.Hillen H.S., Kokic G., Farnung L., Dienemann C., Tegunov D., Cramer P. Structure of replicating SARS-CoV-2 polymerase. Nature. 2020;584(7819):154–156. doi: 10.1038/s41586-020-2368-8. [DOI] [PubMed] [Google Scholar]
  • 92.E.J. Snijder, P.J. Bredenbeek, J.C. Dobbe, V. Thiel, J. Ziebuhr, L.L.M. Poon, Y. Guan, M. Rozanov, W.J.M. Spaan, A.E. Gorbalenya. Unique and conserved features of genome and proteome of SARS-coronavirus, an early split-off from the coronavirus group 2 lineage. J. Mol. Biol. 2003;331(5):991–1004. doi: 10.1016/s0022-2836(03)00865-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pereira F. Evolutionary dynamics of the SARS-CoV-2 ORF8 accessory gene. Infect. Genet. Evol. J. Mol. Epidemiol. Evol. Genet. Infect. Dis. Nov. 2020;85 doi: 10.1016/j.meegid.2020.104525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Li J.-Y., et al. The ORF6, ORF8 and nucleocapsid proteins of SARS-CoV-2 inhibit type I interferon signaling pathway. Virus Res. Sep. 2020;286 doi: 10.1016/j.virusres.2020.198074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kim D., Lee J.-Y., Yang J.-S., Kim J.W., Kim V.N., Chang H. The architecture of SARS-CoV-2 transcriptome. Cell. 2020;181(4):914–921.e10. doi: 10.1016/j.cell.2020.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.F. Wu, et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020;579(7798):265–269. doi: 10.1038/s41586-020-2008-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Pathan R.K., Biswas M., Khandaker M.U. Time series prediction of COVID-19 by mutation rate analysis using recurrent neural network-based LSTM model. Chaos. Solitons. Fractals. 2020;138 doi: 10.1016/j.chaos.2020.110018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sanjuán R., Domingo-Calap P. Mechanisms of viral mutation. Cell. Mol. Life Sci. 2016;73(23):4433–4448. doi: 10.1007/s00018-016-2299-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chen J., Wang R., Wang M., Wei G.-W. Mutations strengthened SARS-CoV-2 infectivity. J. Mol. Biol. 2020;432(19):5212–5226. doi: 10.1016/j.jmb.2020.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.H. Wang, X. Li, T. Li, S. Zhang, L. Wang, X. Wu, J. Liu. The genetic sequence, origin, and diagnosis of SARS-CoV-2. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2020;39(9):1629–1635. doi: 10.1007/s10096-020-03899-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.J. Shang, Y. Wan, C. Liu, B. Yount, K. Gully, Y. Yang, A. Auerbach, G. Peng, R. Baric, F. Li. Structure of mouse coronavirus spike protein complexed with receptor reveals mechanism for viral entry. PLoS Pathog. 2020;16(3) doi: 10.1371/journal.ppat.1008392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.S.K. Samrat, A.M. Tharappel, Z. Li, H. Li. Prospect of SARS-CoV-2 spike protein: potential role in vaccine and therapeutic development. Virus Res. 2020;288 doi: 10.1016/j.virusres.2020.198141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Koyama T., Platt D., Parida L. Variant analysis of SARS-CoV-2 genomes. Bull. World Health Organ. Jul. 2020;98(7):495–504. doi: 10.2471/BLT.20.253591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mercatelli D., Giorgi F.M. Geographic and genomic distribution of SARS-CoV-2 mutations. Front. Microbiol. Jul. 2020;11:1800. doi: 10.3389/fmicb.2020.01800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Korber B. Spike mutation pipeline reveals the emergence of a more transmissible form of SARS-CoV-2. bioRxiv. 2020 doi: 10.1101/2020.04.29.069054. (p. 2020.04.29.069054) [DOI] [Google Scholar]
  • 106.Plante J.A., et al. Spike mutation D614G alters SARS-CoV-2 fitness. Nature. 2021;592(7852):116–121. doi: 10.1038/s41586-020-2895-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Eden J.-S. An emergent clade of SARS-CoV-2 linked to returned travellers from Iran. Virus Evol. Jan. 2020;6(1) doi: 10.1093/ve/veaa027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Guan Q. A genetic barcode of SARS-CoV-2 for monitoring global distribution of different clades during the COVID-19 pandemic. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. Nov. 2020;100:216–223. doi: 10.1016/j.ijid.2020.08.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Andersen K.G., Rambaut A., Lipkin W.I., Holmes E.C., Garry R.F. The proximal origin of SARS-CoV-2. Nat. Med. 2020;26(4):450–452. doi: 10.1038/s41591-020-0820-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Joshi M., et al. Genomic variations in SARS-CoV-2 genomes from Gujarat: underlying role of variants in disease epidemiology. bioRxiv. 2020 doi: 10.1101/2020.07.10.197095. (p. 2020.07.10.197095) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Ceraolo C., Giorgi F.M. Genomic variance of the 2019-nCoV coronavirus. J. Med. Virol. May 2020;92(5):522–528. doi: 10.1002/jmv.25700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Forster P., Forster L., Renfrew C., Forster M. Phylogenetic network analysis of SARS-CoV-2 genomes. Proc. Natl. Acad. Sci. U. S. A. Apr. 2020;117(17):9241–9243. doi: 10.1073/pnas.2004999117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.van Dorp L., et al. Emergence of genomic diversity and recurrent mutations in SARS-CoV-2. Infect. Genet. Evol. Sep. 2020;83 doi: 10.1016/j.meegid.2020.104351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Rambaut A., et al. A dynamic nomenclature proposal for SARS-CoV-2 to assist genomic epidemiology. bioRxiv. 2020 doi: 10.1101/2020.04.17.046086. (p. 2020.04.17.046086) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Duchene S., Featherstone L., Haritopoulou-sinanidou M., Rambaut A., Lemey P., Baele G. Temporal signal and the phylodynamic threshold of SARS-CoV-2. Virus Evolution. 2020;6(2):1–8. doi: 10.1093/ve/veaa061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Kemp S.A., et al. Recurrent emergence and transmission of a SARS-CoV-2 Spike deletion ΔH69/ΔV70. bioRxiv. 2020 doi: 10.1101/2020.12.14.422555. (p. 2020.12.14.422555) [DOI] [Google Scholar]
  • 117.Santos J.C., Passos G.A. The high infectivity of SARS-CoV-2 B.1.1.7 is associated with increased interaction force between Spike-ACE2 caused by the viral N501Y mutation. bioRxiv. 2021 doi: 10.1101/2020.12.29.424708. (p. 2020.12.29.424708) [DOI] [Google Scholar]
  • 118.Davies N.G., et al. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science. Apr. 2021;372(6538):eabg3055. doi: 10.1126/science.abg3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Conti P., Caraffa Al, Gallenga C E, Kritas S K, Frydas I, Younes A, Emidio P Di, Tetè G, Pregliasco F, Ronconi G. The British variant of the new coronavirus-19 (Sars-Cov-2) should not create a vaccine problem. Journal of biological regulators and homeostatic agents. 2021;35(1):1–4. doi: 10.23812/21-3-E. Italy. [DOI] [PubMed] [Google Scholar]
  • 120.Volz E., et al. Transmission of SARS-CoV-2 Lineage B.1.1.7 in England: Insights from linking epidemiological and genetic data. medRxiv. 2021 doi: 10.1101/2020.12.30.20249034. (p. 2020.12.30.20249034) [DOI] [Google Scholar]
  • 121.F. Fratev. The SARS-CoV-2 S1 spike protein mutation N501Y alters the protein interactions with both hACE2 and human derived antibody: A Free energy of perturbation study. bioRxiv. 2020 doi: 10.1101/2020.12.23.424283. [DOI] [PubMed] [Google Scholar]
  • 122.Gu H. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy. Science. Sep. 2020;369(6511):1603–1607. doi: 10.1126/science.abc4730. 80- [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lemmermann N.A., Lieb B., Laufs T., Renzaho A., Runkel S., Kohnen W., Linke M., Gerber S., Schweiger S., Michel A., Bikar S.E. SARS-CoV-2 genome surveillance in Mainz, Germany, reveals convergent origin of the N501Y spike mutation in a hospital setting. medRxiv. 2021 doi: 10.1101/2021.02.11.21251324. [DOI] [Google Scholar]
  • 124.Zhou D., Dejnirattisai W., Supasa P., Liu C., Mentzer A.J., Ginn H.M., Zhao Y., Duyvesteyn H.M., Tuekprakhon A., Nutalai R., Wang B. Evidence of escape of SARS-CoV-2 variant B. 1.351 from natural and vaccine-induced sera. Cell. 2021;184(9):2348–2361. doi: 10.1016/j.cell.2021.02.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wang P., Nair M.S., Liu L., Iketani S., Luo Y., Guo Y., Wang M., Yu J., Zhang B., Kwong P.D., Graham B.S. Antibody resistance of SARS-CoV-2 variants B. 1.351 and B. 1.1. 7. Nature. 2021:1–6. doi: 10.1101/2021.01.25.428137. [DOI] [PubMed] [Google Scholar]
  • 126.Burki T. Understanding variants of SARS-CoV-2. Lancet. 2021;397(10273):462. doi: 10.1016/S0140-6736(21)00298-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Jacobson K.B., Pinsky B.A., Rath M.E.M., Wang H., Miller J.A., Skhiri M., Shepard J., Mathew R., Lee G., Bohman B., Parsonnet J., Holubar M. Post-vaccination SARS-CoV-2 infections and incidence of the B. 1.427/B. 1.429 variant among healthcare personnel at a northern California academic medical center. medRxiv. 2021 doi: 10.1101/2021.04.14.21255431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Long S.W., Olsen R.J., Christensen P.A., Subedi S., Olson R., Davis J.J., Saavedra M.O., Yerramilli P., Pruitt L., Reppond K., Shyer M.N. Sequence analysis of 20,453 SARS-CoV-2 genomes from the Houston Metropolitan Area identifies the emergence and widespread distribution of multiple isolates of all major variants of concern. Am J Pathol . 2021;191(6):983–992. doi: 10.1016/j.ajpath.2021.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Garcia-Beltran W.F., Lam E.C., Denis K.S., Nitido A.D., Garcia Z.H., Hauser B.M., Feldman J., Pavlovic M.N., Gregory D.J., Poznansky M.C., Sigal A. Circulating SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. MedRxiv. 2021 doi: 10.1016/j.cell.2021.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Harvey W.T., Carabelli A.M., Jackson B, Gupta R.K., Thomson E.C., Harrison E.M., Ludden C., Reeve R., Rambaut A., Peacock S.J. SARS-CoV-2 variants, spike mutations and immune escape. Nat. Rev. Microbiol. 2021;19:409–424. doi: 10.1038/s41579-021-00573-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.West A.P., Jr., Barnes C.O., Jr., Yang Z., Jr., Bjorkman P.J., Jr. bioRxiv; 2021. SARS-CoV-2 lineage B. 1.526 emerging in the New York region detected by software utility created to query the spike mutational landscape. [DOI] [Google Scholar]
  • 132.Lasek-Nesselquist E., Lapierre P., Schneider E., George K.S., Pata J. The localized rise of a B. 1.526 variant containing an E484K mutation in New York State. medRxiv. 2021 doi: 10.1101/2021.02.26.21251868. [DOI] [Google Scholar]
  • 133.Hirotsu Y., Omata M. Discovery of a SARS-CoV-2 variant 1 from the P.1 lineage harboring K417T/E484K/N501Y mutations in Kofu, Japan. J. Infect. 2021;82(6):276–316. doi: 10.1016/j.jinf.2021.03.013. (pp. S0163-4453(21)00130–4) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Faria N.R. Genomics and epidemiology of a novel SARS-CoV-2 lineage in Manaus, Brazil. medRxiv. 2021 doi: 10.1101/2021.02.26.21252554. (p. 2021.02.26.21252554) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Murhekar M.V., Bhatnagar T., Wesley J., Thangaraj V. SARS-CoV-2 seroprevalence among the general population and healthcare workers in India, December 2020–January 2021. International Journal of Infectious Diseases. 2021;108:145–155. doi: 10.1016/j.ijid.2021.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kirby T. Country in focus new variant of SARS-CoV-2 in UK causes surge of COVID-19. Lancet Respir. 2021;9(2):e20–e21. doi: 10.1016/S2213-2600(21)00005-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Issa E., Merhi G., Panossian B., Salloum T., Tokajian S. SARS-CoV-2 and ORF3a: non-synonymous mutations and polyproline regions. bioRxiv. 2020 doi: 10.1101/2020.03.27.012013. (p. 2020.03.27.012013) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lorenzo-redondo R., et al. A clade of SARS-CoV-2 viruses associated with lower viral loads in patient upper airways. EBioMedicine. 2020;62 doi: 10.1016/j.ebiom.2020.103112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ayub M.I. Reporting Two SARS-CoV-2 Strains Based on A Unique Trinucleotide-Bloc Mutation and Their Potential Pathogenic Difference. Preprints. 2020;April:2020040337. doi: 10.20944/preprints202004.0337.v1. [DOI] [Google Scholar]
  • 140.Liu Z., et al. Composition and divergence of coronavirus spike proteins and host ACE2 receptors predict potential intermediate hosts of SARS-CoV-2. J. Med. Virol. 2020;92:595–601. doi: 10.1002/jmv.25726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lu R., et al. Articles genomic characterisation and epidemiology of 2019 novel coronavirus : implications for virus origins and receptor binding. Lancet. 2020;395(10224):565–574. doi: 10.1016/S0140-6736(20)30251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ou X., et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020;11(1):1620. doi: 10.1038/s41467-020-15562-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wrapp D., Wang N., Corbett K.S., Goldsmith J.A., Hsieh C.-L., Abiona O., Graham B.S., McLellan J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. Mar. 2020;367(6483):1260–1263. doi: 10.1126/science.abb2507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Duan L., Zheng Q., Zhang H., Niu Y., Lou Y., Wang H. The SARS-CoV-2 spike glycoprotein biosynthesis, structure, function, and antigenicity: implications for the design of spike-based vaccine immunogens. Front. Immunol. 2020;11 doi: 10.3389/fimmu.2020.576622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tang T., Bidon M., Jaimes J.A., Whittaker G.R., Daniel S. Coronavirus membrane fusion mechanism offers a potential target for antiviral development. Antivir. Res. Jun. 2020;178 doi: 10.1016/j.antiviral.2020.104792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Bertram S., et al. TMPRSS2 activates the human coronavirus 229E for cathepsin-independent host cell entry and is expressed in viral target cells in the respiratory epithelium. J. Virol. Jun. 2013;87(11):6150–6160. doi: 10.1128/JVI.03372-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hoffmann M., et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. Apr. 2020;181(2):271–280.e8. doi: 10.1016/j.cell.2020.02.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Coutard B., Valle C., de Lamballerie X., Canard B., Seidah N.G., Decroly E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. Apr. 2020;176 doi: 10.1016/j.antiviral.2020.104742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Cheng Y.-W. Furin inhibitors block SARS-CoV-2 spike protein cleavage to suppress virus production and cytopathic effects. Cell Rep. Oct. 2020;33(2):108254. doi: 10.1016/j.celrep.2020.108254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Millet J.K., Whittaker G.R. Host cell proteases: critical determinants of coronavirus tropism and pathogenesis. Virus Res. Apr. 2015;202:120–134. doi: 10.1016/j.virusres.2014.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Zhang H., Penninger J.M., Li Y., Zhong N., Slutsky A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46(4):586–590. doi: 10.1007/s00134-020-05985-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Zhao Y., Zhao Z., Wang Y., Zhou Y., Ma Y., Zuo W. Single-cell RNA expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov. bioRxiv. 2020 doi: 10.1101/2020.01.26.919985. (p. 2020.01.26.919985) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zou X., Chen K., Zou J., Han P., Hao J., Han Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. Apr. 2020;14(2):185–192. doi: 10.1007/s11684-020-0754-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhang H., Kang Z., Gong H., Xu D., Wang J., Li Z., Cui X. The digestive system is a potential route of 2019-nCov infection: a bioinformatics analysis based on single-cell transcriptomes. Gut. 2020;69(6) doi: 10.1101/2020.01.30.927806. [DOI] [Google Scholar]
  • 155.Chai X., et al. Specific ACE2 expression in cholangiocytes may cause liver damage after 2019-nCoV infection. bioRxiv. 2020 doi: 10.1101/2020.02.03.931766. [DOI] [Google Scholar]
  • 156.Mary D., et al. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1–9. Circ. Res. Sep. 2000;87(5):e1–e9. doi: 10.1161/01.RES.87.5.e1. [DOI] [PubMed] [Google Scholar]
  • 157.Xu H., et al. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020;12(1):8. doi: 10.1038/s41368-020-0074-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Lan J., et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 2020;581(7807):215–220. doi: 10.1038/s41586-020-2180-5. [DOI] [PubMed] [Google Scholar]
  • 159.Vinciguerra M., Greco E. Sars-CoV-2 and black population: ACE2 as shield or blade? Infect. Genet. Evol. Oct. 2020;84 doi: 10.1016/j.meegid.2020.104361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Verdecchia P., Cavallini C., Spanevello A., Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur. J. Intern. Med. Jun. 2020;76:14–20. doi: 10.1016/j.ejim.2020.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Keidar S., Kaplan M., Gamliel-Lazarovich A. ACE2 of the heart: from angiotensin I to angiotensin (1–7) Cardiovasc. Res. Feb. 2007;73(3):463–469. doi: 10.1016/j.cardiores.2006.09.006. [DOI] [PubMed] [Google Scholar]
  • 162.M. C.R., Zhi-Qin W., M. S.H. Role of the angiotensin type 2 receptor in the regulation of blood pressure and renal function. Hypertension. Jan. 2000;35(1):155–163. doi: 10.1161/01.HYP.35.1.155. [DOI] [PubMed] [Google Scholar]
  • 163.Santos R.A.S., et al. The ACE2/Angiotensin-(1–7)/MAS Axis of the renin-angiotensin system: focus on angiotensin-(1–7) Physiol. Rev. Dec. 2017;98(1):505–553. doi: 10.1152/physrev.00023.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Gheblawi M., et al. Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ. Res. May 2020;126(10):1456–1474. doi: 10.1161/CIRCRESAHA.120.317015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bonney G.K., Gao Y., Chew C.A., Windsor J.A. SARS-COV-2 associated acute pancreatitis: cause, consequence or epiphenomenon? Pancreatology. Jul. 2020;20(5):1017–1018. doi: 10.1016/j.pan.2020.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Rawla P., Bandaru S.S., Vellipuram A.R. Review of infectious etiology of acute pancreatitis. Gastroenterol. Res. Jun. 2017;10(3):153–158. doi: 10.14740/gr858w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wang H., et al. SARS coronavirus entry into host cells through a novel clathrin- and caveolae-independent endocytic pathway. Cell Res. Feb. 2008;18(2):290–301. doi: 10.1038/cr.2008.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Oostra M., Hagemeijer M.C., Gent M.V., Bekker C.P.J., Lintelo E.G.T., Rottier P.J.M., Haan C.A.M.D. Topology and Membrane Anchoring of the Coronavirus Replication Complex: Not All Hydrophobic Domains of nsp3 and nsp6 Are Membrane Spanning. J. Virol. Dec. 2008;82(24):12392–12405. doi: 10.1128/JVI.01219-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Ziebuhr J. Molecular biology of severe acute respiratory syndrome coronavirus. Curr. Opin. Microbiol. Aug. 2004;7(4):412–419. doi: 10.1016/j.mib.2004.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Mommaas A.M. SARS-coronavirus replication / transcription complexes are membrane-protected and need a host factor for activity in vitro. 2008;4(5) doi: 10.1371/journal.ppat.1000054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Benvenuto D., et al. Evolutionary analysis of SARS-CoV-2: how mutation of non-structural protein 6 (NSP6) could affect viral autophagy. J. Infect. 2020;81(1):e24–e27. doi: 10.1016/j.jinf.2020.03.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Prajapat M., et al. Drug targets for corona virus: a systematic review. Indian J. Pharmacol. 2020;52(1):56–65. doi: 10.4103/ijp.IJP_115_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.E.J. Snijder, Y. Van Der Meer, J. Zevenhoven-Dobbe, J.J.M. Onderwater, J. Van Der Meulen, H.K. Koerten, A.M. Mommaas. Ultrastructure and origin of membrane vesicles associated with the severe acute respiratory syndrome coronavirus replication complex. J. Virol. 2006;80(12):5927–5940. doi: 10.1128/JVI.02501-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Y. Gao, L. Yan, Y. Huang, F. Liu, Y. Zhao, L. Cao, T. Wang, Q. Sun, Z. Ming, L. Zhang. Structure of the RNA-dependent RNA polymerase from COVID-19 virus. Science. 2020;368(6492):779–782. doi: 10.1126/science.abb7498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.I. Sola, F. Almazán, S. Zúñiga, L. Enjuanes. Continuous and discontinuous RNA synthesis in coronaviruses. Annu. Rev. Virol. 2015;2(1):265–288. doi: 10.1146/annurev-virology-100114-055218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.F. Robson, K.S. Khan, T.K. Le, C. Paris, S. Demirbag, P. Barfuss, P. Rocchi, W.-L. Ng. Coronavirus RNA proofreading: molecular basis and therapeutic targeting. Mol. Cell. 2020;79(5):710–727. doi: 10.1016/j.molcel.2020.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Y. Wang, Y. Sun, A. Wu, S. Xu, R. Pan, C. Zeng, X. Jin, X. Ge, Z. Shi, T. Ahola. Coronavirus nsp10/nsp16 methyltransferase can be targeted by nsp10-derived peptide in vitro and in vivo to reduce replication and pathogenesis. J. Virol. 2015;89(16):8416–8427. doi: 10.1128/JVI.00948-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Q. Ye, A.M.V. West, S. Silletti, K.D. Corbett. Architecture and self-assembly of the SARS-CoV-2 nucleocapsid protein. Protein Sci. 2020;29(9):1890–1901. doi: 10.1002/pro.3909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.M. Ke, Y. Chen, A. Wu, Y. Sun, C. Su, H. Wu, X. Jin, J. Tao, Y. Wang, X. Ma. Short peptides derived from the interaction domain of SARS coronavirus nonstructural protein nsp10 can suppress the 2’-O-methyltransferase activity of nsp10/nsp16 complex. Virus Res. 2012;167(2):322–328. doi: 10.1016/j.virusres.2012.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.O.A. Olwenyi, S.R. Dyavar, A. Acharya, A.T. Podany, C. V Fletcher, C.L. Ng, S.P. Reid, S.N. Byrareddy. Immuno-epidemiology and pathophysiology of coronavirus disease 2019 (COVID-19) J. Mol. Med. (Berl) 2020;98(10):1369–1383. doi: 10.1007/s00109-020-01961-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sokol C.L., Luster A.D. The chemokine system in innate immunity. Cold Spring Harb. Perspect. Biol. 2015;7(5) doi: 10.1101/cshperspect.a016303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.A. Angelopoulou, N. Alexandris, E. Konstantinou, K. Mesiakaris, C. Zanidis, K. Farsalinos, K. Poulas. Imiquimod - a toll like receptor 7 agonist - is an ideal option for management of COVID 19. Environ. Res. 2020;188 doi: 10.1016/j.envres.2020.109858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.R. Channappanavar, A.R. Fehr, R. Vijay, M. Mack, J. Zhao, D.K. Meyerholz, S. Perlman. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe. 2016;19(2):181–193. doi: 10.1016/j.chom.2016.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.T. Kawai, S. Akira. TLR signaling. Cell Death Differ. 2006;13(5):816–825. doi: 10.1038/sj.cdd.4401850. [DOI] [PubMed] [Google Scholar]
  • 185.A. Iwasaki, R. Medzhitov. Control of adaptive immunity by the innate immune system. Nat. Immunol. 2015;16(4):343–353. doi: 10.1038/ni.3123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.J. Geginat, P. Larghi, M. Paroni, G. Nizzoli, A. Penatti, M. Pagani, N. Gagliani, P. Meroni, S. Abrignani, R.A. Flavell. The light and the dark sides of Interleukin-10 in immune-mediated diseases and cancer. Cytokine Growth Factor Rev. 2016;30:87–93. doi: 10.1016/j.cytogfr.2016.02.003. [DOI] [PubMed] [Google Scholar]
  • 187.J. Hadjadj, N. Yatim, L. Barnabei, A. Corneau, J. Boussier, N. Smith, H. Péré, B. Charbit, V. Bondet, C. Chenevier-Gobeaux. Impaired type I interferon activity and exacerbated inflammatory responses in severe Covid-19 patients. medRxiv. 2020 doi: 10.1101/2020.04.19.20068015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Huang C. Articles Clinical features of patients infected with 2019 novel coronavirus in Wuhan , China. 2020. pp. 497–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Monteleone G., Sarzi-Puttini P.C., Ardizzone S. Preventing COVID-19-induced pneumonia with anticytokine therapy. Lancet Rheumatol. May 2020;2(5):e255–e256. doi: 10.1016/S2665-9913(20)30092-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.F. Wang, H. Hou, Y. Luo, G. Tang, S. Wu, M. Huang, W. Liu, Y. Zhu, Q. Lin, L. Mao. The laboratory tests and host immunity of COVID-19 patients with different severity of illness. JCI insight. 2020;5(10) doi: 10.1172/jci.insight.137799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Liu P.P., Blet A., Smyth D., Li H. The science underlying COVID-19: implications for the cardiovascular system. Circulation. 2020;142(1):68–78. doi: 10.1161/CIRCULATIONAHA.120.047549. [DOI] [PubMed] [Google Scholar]
  • 192.Hernández-Galdamez D.R., et al. Increased risk of hospitalization and death in patients with COVID-19 and pre-existing noncommunicable diseases and modifiable risk factors in Mexico. Arch. Med. Res. Oct. 2020;51(7):683–689. doi: 10.1016/j.arcmed.2020.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.B. Li, J. Yang, F. Zhao, L. Zhi, X. Wang, L. Liu, Z. Bi, Y. Zhao. Prevalence and impact of cardiovascular metabolic diseases on COVID-19 in China. Clin. Res. Cardiol. 2020;109(5):531–538. doi: 10.1007/s00392-020-01626-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.L. Mao, H. Jin, M. Wang, Y. Hu, S. Chen, Q. He, J. Chang, C. Hong, Y. Zhou, D. Wang. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol. 2020;77(6):683–690. doi: 10.1001/jamaneurol.2020.1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Rahman A., Niloofa R., Zoysa I.M.De, Cooray A.D., Kariyawasam J. 2020. Neurological manifestations in COVID-19 : A narrative review. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.D. Wang, B. Hu, C. Hu, F. Zhu, X. Liu, J. Zhang, B. Wang, H. Xiang, Z. Cheng, Y. Xiong. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–1069. doi: 10.1001/jama.2020.1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.T. Guo, Y. Fan, M. Chen, X. Wu, L. Zhang, T. He, H. Wang, J. Wan, X. Wang, Z. Lu. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19) JAMA Cardiol. 2020;5(7):811–818. doi: 10.1001/jamacardio.2020.1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.M.-J. Shao, L.-X. Shang, J.-Y. Luo, J. Shi, Y. Zhao, X.-M. Li, Y.-N. Yang. Myocardial injury is associated with higher mortality in patients with coronavirus disease 2019: a meta-analysis. J. Geriatr. Cardiol. 2020;17(4):224–228. doi: 10.11909/j.issn.1671-5411.2020.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Klok F.A., et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020;191:145–147. doi: 10.1016/j.thromres.2020.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Poissy J., et al. Pulmonary embolism in patients with COVID-19: awareness of an increased prevalence. Circulation. 2020;142(2):184–186. doi: 10.1161/CIRCULATIONAHA.120.047430. Italy. [DOI] [PubMed] [Google Scholar]
  • 201.Iba T., Levy J.H., Levi M., Connors J.M., Thachil J. Coagulopathy of coronavirus disease 2019. Crit. Care Med. 2020;48(9):1358–1364. doi: 10.1097/CCM.0000000000004458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Merad M., Martin J.C. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat. Rev. Immunol. 2020;20(6):355–362. doi: 10.1038/s41577-020-0331-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Shi W., Lv J., Lin L. Coagulopathy in COVID-19: focus on vascular thrombotic events. J. Mol. Cell. Cardiol. 2020;146:32–40. doi: 10.1016/j.yjmcc.2020.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Eljilany I., Elzouki A.-N. D-dimer, fibrinogen, and IL-6 in COVID-19 patients with suspected venous thromboembolism: a narrative review. Vasc. Health Risk Manag. 2020;16:455–462. doi: 10.2147/VHRM.S280962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.H. Su, M. Yang, C. Wan, L.-X. Yi, F. Tang, H.-Y. Zhu, F. Yi, H.-C. Yang, A.B. Fogo, X. Nie. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020;98(1):219–227. doi: 10.1016/j.kint.2020.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.P. J.R., Jr., Parnia S., Jr., Harris E.Q., Jr., Varney J., Jr. An Atypical Case of COVID-19 Induced Rhabdomyolysis and Acute Kidney Injury. J Nephrol Kidney Dis; 2020. p. 4. [DOI] [Google Scholar]
  • 207.Yang Y., et al. Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J. Allergy Clin. Immunol. 2020;146(1):119–127. doi: 10.1016/j.jaci.2020.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Ji W., Bishnu G., Cai Z., Shen X. Analysis clinical features of COVID-19 infection in secondary epidemic area and report potential biomarkers in evaluation. medRxiv. 2020 doi: 10.1101/2020.03.10.20033613. [DOI] [Google Scholar]
  • 209.Baig A.M., Khaleeq A., Ali U., Syeda H. Evidence of the COVID-19 virus targeting the CNS: tissue distribution, host-virus interaction, and proposed neurotropic mechanisms. ACS Chem. Neurosci. 2020;11(7):995–998. doi: 10.1021/acschemneuro.0c00122. [DOI] [PubMed] [Google Scholar]
  • 210.R. Qu, Y. Ling,, Y. Zhang,, L. Wei,, X. Chen,, X. Li,, X. Liu, ,, H. Liu,, Z. Guo, H. Ren. Platelet-to-lymphocyte ratio is associated with prognosis in patients with coronavirus disease-19. J. Med. Virol. 2020;92(9):1533–1541. doi: 10.1002/jmv.25767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Shi S., et al. Association of Cardiac Injury with Mortality in hospitalized patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5(7):802–810. doi: 10.1001/jamacardio.2020.0950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Yang Y., . Shen, ,, J. Li, J. Yuan, M. Yang, F. Wang, G. Li, Y. Li, L. Xing, L. Peng. Exuberant elevation of IP-10, MCP-3 and IL-1ra during SARS-CoV-2 infection is associated with disease severity and fatal outcome. medRxiv. 2020 doi: 10.1101/2020.03.02.20029975. (p. 2020.03.02.20029975) [DOI] [Google Scholar]
  • 213.Song J.-W., et al. Immunological and inflammatory profiles in mild and severe cases of COVID-19. Nat. Commun. 2020;11(1):3410. doi: 10.1038/s41467-020-17240-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Fu S., et al. Virologic and clinical characteristics for prognosis of severe COVID-19: a retrospective observational study in Wuhan, China. medRxiv. 2020 doi: 10.1101/2020.04.03.20051763. (p. 2020.04.03.20051763) [DOI] [Google Scholar]
  • 215.Xu H., et al. Acute Myocardial Injury of Patients with Coronavirus Disease 2019. medRxiv. 2020 doi: 10.1101/2020.03.05.20031591. (p. 2020.03.05.20031591) [DOI] [Google Scholar]
  • 216.Zhou Y., Fu B., Zheng X., Wang X, Qi Y., Sun R., Tian Z., Xu X., Wei H., Zhao C. Aberrant pathogenic GM-CSF+ T cells and inflammatory CD14+CD16+ monocytes in severe pulmonary syndrome patients of a new coronavirus. bioRxiv p. 2020.02.12.945576. Jan. 2020 doi: 10.1101/2020.02.12.945576. [DOI] [Google Scholar]
  • 217.Liu J., et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. May 2020;55 doi: 10.1016/j.ebiom.2020.102763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Chen X., ZhaO B., Qu Y., Chen Y., Xiong J., Feng Y., Men D., Huang Q., Liu Y., Yang B., Ding J., Li F. 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. an Off. Publ. Infect. Dis. Soc. Am. Nov. 2020;71(8):1937–1942. doi: 10.1093/cid/ciaa449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Gong J., Dong H., Xia Q., Huang Z., Wang D., Zhao Y., Liu W., Tu S., Zhang M., Wang Q., Lu F. Correlation analysis between disease severity and inflammation-related parameters in patients with COVID-19 pneumonia. medRxiv p. 2020.02.25.20025643. 2020 doi: 10.1101/2020.02.25.20025643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Ma C., Gu J., Hou P., Zhang L., Bai Y., Guo Z., Wu H., Zhang H.B., Li P., Zhao X. Incidence, clinical characteristics and prognostic factor of patients with COVID-19: a systematic review and meta-analysis. medRxiv p. 2020.03.17.20037572. 2020 doi: 10.1101/2020.03.17.20037572. [DOI] [Google Scholar]
  • 221.Liu W., Liu J., Liu K., Shang J., Deng Y., wei S. Analysis of clinical features of 29 patients with 2019 novel coronavirus pneumonia. Zhonghua jie he he hu xi za zhi = Zhonghua jiehe he huxi zazhi = Chinese J. Tuberc. Respir. Dis. Feb. 2020;43(0):E005. doi: 10.3760/cma.j.issn.1001-0939.2020.0005. [DOI] [PubMed] [Google Scholar]
  • 222.Wen W., et al. Immune cell profiling of COVID-19 patients in the recovery stageby single-cell sequencing. Cell Discov. 2020;6(1):31. doi: 10.1038/s41421-020-0168-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Costela-Ruiz V.J., Illescas-Montes R., Puerta-Puerta J.M., Ruiz C., Melguizo-Rodríguez L. SARS-CoV-2 infection: the role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev. Aug. 2020;54:62–75. doi: 10.1016/j.cytogfr.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Diao B., et al. Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. medRxiv p. 2020.03.04.20031120. 2020 doi: 10.1101/2020.03.04.20031120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Liu Y., Liao W., Wan L., Xiang T., Zhang W. Correlation between Relative Nasopharyngeal Virus RNA load and lymphocyte count disease severity in Patients with COVID-19. Viral Immunol. 2021;34:330–335. doi: 10.1089/vim.2020.0062. [DOI] [PubMed] [Google Scholar]
  • 226.Mukherjee A., Ghosh R., Aftab G. Rhabdomyolysis in a patient with coronavirus disease 2019. Cureus. 2020;12(7):e8956. doi: 10.7759/cureus.8956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Yin R., Feng W., Wang T., et al. Concomitant neurological symptoms observed in a patient diagnosed with coronavirus disease 2019. J. Med. Virol. 2020;92(10):1782–1784. doi: 10.1002/jmv.25888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Filatov A., Sharma P., Hindi F., Espinosa P.S. Neurological Complications of Coronavirus Disease (COVID-19): Encephalopathy. Cureus. 2020;12(3):e7352. doi: 10.7759/cureus.7352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.N. Chen, M. Zhou, X. Dong, J. Qu, F. Gong, Y. Han. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:P507–513. doi: 10.1016/S0140-6736(20)30211-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Li Y., Wang M., Zhou Y., Chang J., Xian Y., Mao L. Acute cerebrovascular disease following COVID-19: a single center, retrospective, observational study. Stroke Vasc Neurol . 2020;5(3):279–284. doi: 10.1136/svn-2020-000431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Lauren A. Beslow, et al. Pediatric Ischemic Stroke: An Infrequent Complication of SARS-CoV-2. Ann. Neurol. 2021;89(4):657–665. doi: 10.1002/ana.25991. [DOI] [PubMed] [Google Scholar]
  • 232.Lodigiani C., Lapichino G., Carenzo L., Cecconi M., Ferrazzi P., Sebastian T. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in MilanItaly. Thrombosis Research. 2020;191:9–14. doi: 10.1016/j.thromres.2020.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Connors J., Levy J. Thromboinflammation and hypercoagulability of COVID-19. J. Thromb. Haemost. 2020;18(7):1559–1561. doi: 10.1111/jth.14849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Alberti P., Beretta S., Piatti M., et al. Guillain-Barré syndrome related to COVID-19 infection. Neurol Neuroimmunol Neuroinflamm. 2020;7(4) doi: 10.1212/NXI.0000000000000741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Virani A., Rabold E., Hanson T., Haag A., Elrufay R., Cheema T. Guillain- Barré syndrome associated with SARS-CoV-2 infection. IDCases. 2020;20 doi: 10.1016/j.idcr.2020.e00771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.La Rosa G., Bonadonna L., Lucentini L., Kenmoe S., Suffredini E. Coronavirus in water environments: occurrence, persistence and concentration methods – a scoping review. Water Res. 2020;179 doi: 10.1016/j.watres.2020.115899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Chen Y., Chen L., Deng Q., Zhang G., Wu K., Ni L., Yang Y., Liu B., Wang W., Wei C., Yang J., Ye G., Cheng Z. The presence of SARS-CoV-2 RNA in the feces of COVID-19 patients. J. Med. Virol. 2020;92(7):833–840. doi: 10.1002/jmv.25825. [DOI] [PubMed] [Google Scholar]
  • 238.D’Amico F., Baumgart D.C., Danese S., Peyrin-Biroulet L. Clinical Gastroenterology and Hepatology; 2020. Diarrhea during COVID-19 infection: pathogenesis, epidemiology, prevention and management; pp. 1663–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Pan Y., Zhang D., Yang P., Poon L.L.M., Wang Q. Viral load of SARS-CoV-2 in clinical samples. Lancet Infect. Dis. 2020;20(4):411–412. doi: 10.1016/S1473-3099(20)30113-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Kampf G., Todt D., Pfaender S., Steinmann E. Persistence of coronaviruses on inanimate surfaces and their inactivation with biocidal agents. J. Hosp. Infect. 2020;104(3):246–251. doi: 10.1016/j.jhin.2020.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Cai J., Sun W., Huang J., Gamber M., Wu J., He G. Indirect virus transmission in cluster of COVID-19 cases, Wenzhou, China, 2020. Emerg. Infect. Dis. 2020;26(6):1343–1345. doi: 10.3201/eid2606.200412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Gu J., Han B., Wang J. COVID-19: gastrointestinal manifestations and potential. FecalOral Transmission Gastroenterol. 2020;158(6):1518–1519. doi: 10.1053/j.gastro.2020.02.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Amirian E.S. Potential fecal transmission of SARS-CoV-2: current evidence and implications for public health [published online ahead of print, 2020 Apr 23] Int. J. Infect. Dis. 2020;95:363–370. doi: 10.1016/j.ijid.2020.04.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Hart O.E., Halden R.U. Computational analysis of SARS-CoV-2/COVID-19 surveillance by wastewater-based epidemiology locally and globally: feasibility, economy, opportunities and challenges. Sci. Total Environ. 2020;730 doi: 10.1016/j.scitotenv.2020.138875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Lodder W., de Roda Husman A.M. SARS-CoV-2 in wastewater: potential health risk, but also data source. Lancet Gastroenterol. Hepatol. 2020;5(6):P533–534. doi: 10.1016/S2468-1253(20)30087-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Bogler A., et al. Rethinking wastewater risks and monitoring in light of the COVID-19 pandemic. Nat. Sustain. 2020;729(3):981–990. doi: 10.1038/s41893-020-00605-2. [DOI] [Google Scholar]
  • 247.Liu J., Yu H., Zhang S. The indispensable role of chest CT in the detection of coronavirus disease 2019 (COVID-19) Eur. J. Nucl. Med. Mol. Imaging. Jul. 2020;47(7):1638–1639. doi: 10.1007/s00259-020-04795-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Guan W., et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. Feb. 2020;382(18):1708–1720. doi: 10.1056/NEJMoa2002032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Shen M., et al. Recent advances and perspectives of nucleic acid detection for coronavirus. J. Pharm. Anal. Apr. 2020;10(2):97–101. doi: 10.1016/j.jpha.2020.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Bernheim A., et al. Chest CT findings in coronavirus Disease-19 (COVID-19): relationship to duration of infection. Radiology. Feb. 2020;295(3) doi: 10.1148/radiol.2020200463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Lee E.Y.P., Ng M.Y., Khong P.L. COVID-19 pneumonia: what has CT taught us? Lancet Infect. Dis. 2020;20(4):384–385. doi: 10.1016/S1473-3099(20)30134-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Whiting P., Singatullina N., Rosser J.H. Computed tomography of the chest: I. Basic principles. BJA Educ. 2015;15(6):299–304. doi: 10.1093/bjaceaccp/mku063. [DOI] [Google Scholar]
  • 253.Freeman W.M., Walker S.J., Vrana K.E. Quantitative RT-PCR: pitfalls and potential. Biotechniques. Jan. 1999;26(1):112–122. doi: 10.2144/99261rv01. (124-125) [DOI] [PubMed] [Google Scholar]
  • 254.Kageyama T., et al. Broadly reactive and highly sensitive assay for Norwalk-like viruses based on real-time quantitative reverse transcription-PCR. J. Clin. Microbiol. Apr. 2003;41(4):1548–1557. doi: 10.1128/jcm.41.4.1548-1557.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Udugama B., et al. Diagnosing COVID-19: the disease and tools for detection. ACS Nano. Apr. 2020;14(4):3822–3835. doi: 10.1021/acsnano.0c02624. [DOI] [PubMed] [Google Scholar]
  • 256.Corman V.M., et al. Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Euro Surveill. Jan. 2020;25(3):2000045. doi: 10.2807/1560-7917.ES.2020.25.3.2000045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Yang W., Yan F. Patients with RT-PCR-confirmed COVID-19 and normal chest CT. Radiology. May 2020;295(2):E3. doi: 10.1148/radiol.2020200702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Zhang Q., Zhao Q. Inactivating porcine coronavirus before nuclei acid isolation with the temperature higher than 56 °C damages its genome integrity seriously. bioRxiv p. 2020.02.20.958785. 2020 doi: 10.1101/2020.02.20.958785. [DOI] [Google Scholar]
  • 259.Xie X., Zhong Z., Zhao W., Zheng C., Wang F., Liu J. Chest CT for typical coronavirus disease 2019 (COVID-19) pneumonia: relationship to negative RT-PCR testing. Radiology. Aug. 2020;296(2):E41–E45. doi: 10.1148/radiol.2020200343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Pan F., et al. Time course of lung changes at chest CT during recovery from coronavirus disease 2019 (COVID-19) Radiology. Jun. 2020;295(3):715–721. doi: 10.1148/radiol.2020200370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Kobayashi Y., Mitsudomi T. Management of ground-glass opacities: should all pulmonary lesions with ground-glass opacity be surgically resected? Transl. Lung Cancer Res. Oct. 2013;2(5):354–363. doi: 10.3978/j.issn.2218-6751.2013.09.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Ai T., et al. Correlation of chest CT and RT-PCR testing for coronavirus disease 2019 (COVID-19) in China: a report of 1014 cases. Radiology. Feb. 2020;296(2):32–40. doi: 10.1148/radiol.2020200642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Craw P., Balachandran W. Isothermal nucleic acid amplification technologies for point-of-care diagnostics: a critical review. Lab Chip. Jul. 2012;12(14):2469–2486. doi: 10.1039/c2lc40100b. [DOI] [PubMed] [Google Scholar]
  • 264.Lamb L.E., Bartolone S.N., Ward E., Chancellor M.B. Rapid detection of novel coronavirus (COVID-19) by reverse transcription-loop-mediated isothermal Amplification. medRxiv p. 2020.02.19.20025155. 2020 doi: 10.1101/2020.02.19.20025155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Yu L. Rapid colorimetric detection of COVID-19 coronavirus using a reverse transcriptional loop-mediated isothermal amplification (RT-LAMP) diagnostic platform: iLACO. medRxiv p. 2020.02.20.20025874. 2020 doi: 10.1101/2020.02.20.20025874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Zhang Y. Rapid molecular detection of SARS-CoV-2 (COVID-19) virus RNA using colorimetric LAMP. medRxiv p. 2020.02.26.20028373. 2020 doi: 10.1101/2020.02.26.20028373. [DOI] [Google Scholar]
  • 267.Yang W. Rapid detection of SARS-CoV-2 using reverse transcription RT-LAMP method. medRxiv p. 2020.03.02.20030130. 2020 doi: 10.1101/2020.03.02.20030130. [DOI] [Google Scholar]
  • 268.Mori Y., Nagamine K., Tomita N., Notomi T. Detection of loop-mediated isothermal amplification reaction by turbidity derived from magnesium pyrophosphate formation. Biochem. Biophys. Res. Commun. Nov. 2001;289(1):150–154. doi: 10.1006/bbrc.2001.5921. [DOI] [PubMed] [Google Scholar]
  • 269.Notomi T., Okayama H., Masubuchi H., Yonekawa T., Watanabe K., Amino N., Hase T. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. Jun. 2000;28(12):E63. doi: 10.1093/nar/28.12.e63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Guo X., Geng P., Wang Q., Cao B., Liu B. Development of a single nucleotide polymorphism DNA microarray for the detection and genotyping of the SARS coronavirus. J. Microbiol. Biotechnol. Oct. 2014;24(10):1445–1454. doi: 10.4014/jmb.1404.04024. [DOI] [PubMed] [Google Scholar]
  • 271.de Souza Luna L.K., et al. Generic detection of coronaviruses and differentiation at the prototype strain level by reverse transcription-PCR and nonfluorescent low-density microarray. J. Clin. Microbiol. Mar. 2007;45(3):1049–1052. doi: 10.1128/JCM.02426-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Hardick J., et al. Initial performance evaluation of a spotted array Mobile Analysis Platform (MAP) for the detection of influenza A/B, RSV, and MERS coronavirus. Diagn. Microbiol. Infect. Dis. 2018;91:245–247. doi: 10.1016/j.diagmicrobio.2018.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.To K.K.W., et al. Temporal profiles of viral load in posterior oropharyngeal saliva samples and serum antibody responses during infection by SARS-CoV-2: an observational cohort study. Lancet Infect. Dis. 2020;20(5):565–574. doi: 10.1016/S1473-3099(20)30196-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Lv H., et al. Cross-reactive antibody response between SARS-CoV-2 and SARS-CoV infections. Cell Rep. 2020 doi: 10.1101/2020.03.15.993097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Long Q.-X., et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat. Med. 2020;26(6):845–848. doi: 10.1038/s41591-020-0897-1. [DOI] [PubMed] [Google Scholar]
  • 276.Rahimi H., Salehiabar M., Barsbay M., Ghaffarlou M., Kavetskyy T., Sharafi A., Davaran S., Chauhan S.C., Danafar H., Kaboli S., Nosrati H. CRISPR systems for COVID-19 diagnosis. ACS Sensors. 2021;6(4):1430–1445. doi: 10.1021/acssensors.0c02312. [DOI] [PubMed] [Google Scholar]
  • 277.Mohammadinejad R., Biagioni A., Arunkumar G., Shapiro R., Chang K.-C., Sedeeq M., Taiyab A., Hashemabadi M., Pardakhty A., Mandegary A., Thiery J.-P., Aref A.R., Azimi I. EMT signaling: potential contribution of CRISPR/Cas gene editing. Cell. Mol. Life Sci. 2020;77:2701. doi: 10.1007/s00018-020-03449-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Mohammadinejad R., Sassan H., Pardakhty A., Hashemabadi M., Ashrafizadeh M., Dehshahri A., Mandegary A. ZEB1 and ZEB2 gene editing mediated by CRISPR/Cas9 in A549 cell line. Bratisl. Lek. Listy. 2020;121(1):31–36. doi: 10.4149/BLL_2020_005. [DOI] [PubMed] [Google Scholar]
  • 279.Rahimi H., Salehiabar M., Charmi J., Barsbay M., Ghaffarlou M., Roohi Razlighi M., Davaran S., Khalilov R., Sugiyama M., Nosrati H., Kaboli S., Danafar H., Webster T.J. Harnessing nanoparticles for the efficient delivery of the CRISPR/Cas9 system. Nano Today. 2020;34 [Google Scholar]
  • 280.Sasano Y., Nagasawa K., Kaboli S., Sugiyama M., Harashima S. CRISPR-PCS: a powerful new approach to inducing multiple chromosome splitting in Saccharomyces cerevisiae. Sci. Rep. 2016;6(1):1–11. doi: 10.1038/srep30278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Makarova K.S., Wolf Y.I., Alkhnbashi O.S., Costa F., Shah S.A., Saunders S.J., Barrangou R., Brouns S.J.J., Charpentier E., Haft D.H., Horvath P., Moineau S., Mojica F.J.M., Terns R.M., Terns M.P., White M.F., Yakunin A.F., Garrett R.A., van derOost J., Backofen R., Koonin E.V. An updated evolutionary classification of CRISPR−Cas systems. Nat. Rev. Microbiol. 2015;13(11):722–736. doi: 10.1038/nrmicro3569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Shmakov S., Smargon A., Scott D., Cox D., Pyzocha N., Yan W., Abudayyeh O.O., Gootenberg J.S., Makarova K.S., Wolf Y.I., Severinov K., Zhang F., Koonin E.V. Diversity and evolution of class 2 CRISPR−Cas systems. Nat. Rev. Microbiol. 2017;15(3):169–182. doi: 10.1038/nrmicro.2016.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Chen J.S., Ma E., Harrington L.B., Da Costa M., Tian X., Palefsky J.M., Doudna J.A. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science. 2018;360(6387):436–439. doi: 10.1126/science.aar6245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.East-Seletsky A., O’Connell M.R., Knight S.C., Burstein D., Cate J.H., Tjian R., Doudna J.A. Two distinct RNase activities ofCRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature. 2016;538(7624):270–273. doi: 10.1038/nature19802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Mohammadinejad R., Dehshahri A., Sassan H., Behnam B., Ashrafizadeh M., Samareh Gholami A., Pardakhty A., Mandegary A. Preparation of carbon dot as a potential CRISPR/Cas9 plasmid delivery system for lung cancer cells. Minerva Biotecnol. 2020;32(3):106–113. [Google Scholar]
  • 286.Kellner M.J., Koob J.G., Gootenberg J.S., Abudayyeh O.O., Zhang F. SHERLOCK: nucleic acid detection with CRISPR nucleases. Nat. Protoc. 2019;14(10):2986–3012. doi: 10.1038/s41596-019-0210-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Joung J., et al. Point-of-care testing for COVID-19 using SHERLOCK diagnostics. MedRxiv Prepr. Serv. Heal. Sci. 2020 doi: 10.1101/2020.05.04.20091231. [DOI] [Google Scholar]
  • 288.O’Connell M.R. Molecular mechanisms of RNA targeting by Cas13-containing type VI CRISPR-cas systems. J. Mol. Biol. Jan. 2019;431(1):66–87. doi: 10.1016/j.jmb.2018.06.029. [DOI] [PubMed] [Google Scholar]
  • 289.Gootenberg J.S., Abudayyeh O.O., Kellner M.J., Joung J., Collins J.J., Zhang F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science. Apr. 2018;360(6387):439–444. doi: 10.1126/science.aaq0179. 80- [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Gootenberg J.S., et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science. Apr. 2017;356(6336):438–442. doi: 10.1126/science.aam9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Xiang J., et al. Evaluation of enzyme-linked immunoassay and colloidal gold-immunochromatographic assay kit for detection of novel coronavirus (SARS-Cov-2) causing an outbreak of pneumonia (COVID-19) medRxiv p. 2020.02.27.20028787. Jan. 2020 doi: 10.1101/2020.02.27.20028787. [DOI] [Google Scholar]
  • 292.Foudeh A.M., Fatanat Didar T., Veres T., Tabrizian M. Microfluidic designs and techniques using lab-on-a-chip devices for pathogen detection for point-of-care diagnostics. Lab Chip. 2012;12(18):3249–3266. doi: 10.1039/C2LC40630F. [DOI] [PubMed] [Google Scholar]
  • 293.Laksanasopin T. A smartphone dongle for diagnosis of infectious diseases at the point of care. Sci. Transl. Med. 2015;7(273):273re1. doi: 10.1126/scitranslmed.aaa0056. [DOI] [PubMed] [Google Scholar]
  • 294.Seo G., et al. Rapid detection of COVID-19 causative virus (SARS-CoV-2) in human nasopharyngeal swab specimens using field-effect transistor-based biosensor. ACS Nano. Apr. 2020;14(4):5135–5142. doi: 10.1021/acsnano.0c02823. [DOI] [PubMed] [Google Scholar]
  • 295.Wang X., et al. Clustered regularly interspaced short palindromic Repeats/Cas9-mediated lateral flow nucleic acid assay. ACS Nano. Feb. 2020;14(2):2497–2508. doi: 10.1021/acsnano.0c00022. [DOI] [PubMed] [Google Scholar]
  • 296.Imai M., et al. Rapid diagnosis of H5N1 avian influenza virus infection by newly developed influenza H5 hemagglutinin gene-specific loop-mediated isothermal amplification method. J. Virol. Methods. May 2007;141(2):173–180. doi: 10.1016/j.jviromet.2006.12.004. [DOI] [PubMed] [Google Scholar]
  • 297.Amer H.M., Abd El Wahed A., Shalaby M.A., Almajhdi F.N., Hufert F.T., Weidmann M. A new approach for diagnosis of bovine coronavirus using a reverse transcription recombinase polymerase amplification assay. J. Virol. Methods. Nov. 2013;193(2):337–340. doi: 10.1016/j.jviromet.2013.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Wat D., et al. The role of respiratory viruses in cystic fibrosis. J. Cyst. Fibros. Jul. 2008;7(4):320–328. doi: 10.1016/j.jcf.2007.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Martel N., Gomes S.A., Chemin I., Trépo C., Kay A. Improved rolling circle amplification (RCA) of hepatitis B virus (HBV) relaxed-circular serum DNA (RC-DNA) J. Virol. Methods. Nov. 2013;193(2):653–659. doi: 10.1016/j.jviromet.2013.07.045. [DOI] [PubMed] [Google Scholar]
  • 300.Shirato K., et al. Diagnosis of human respiratory syncytial virus infection using reverse transcription loop-mediated isothermal amplification. J. Virol. Methods. Jan. 2007;139(1):78–84. doi: 10.1016/j.jviromet.2006.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Rowe T., et al. Detection of antibody to avian influenza a (H5N1) virus in human serum by using a combination of serologic assays. J. Clin. Microbiol. Apr. 1999;37(4):937–943. doi: 10.1128/JCM.37.4.937-943.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Rissin D.M., et al. Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol. 2010;28(6):595–599. doi: 10.1038/nbt.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Thaxton C.S., et al. Nanoparticle-based bio-barcode assay redefines ‘undetectable’ PSA and biochemical recurrence after radical prostatectomy. Proc. Natl. Acad. Sci. U. S. A. Nov. 2009;106(44):18437–18442. doi: 10.1073/pnas.0904719106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Harrison C. Coronavirus puts drug repurposing on the fast track. Nature biotechnology. Apr. 2020;vol. 38(4):379–381. doi: 10.1038/d41587-020-00003-1. United States. [DOI] [PubMed] [Google Scholar]
  • 305.Prasad A., Muthamilarasan M., Prasad M. Synergistic antiviral effects against SARS-CoV-2 by plant-based molecules. Plant Cell Rep. Sep. 2020;39(9):1109–1114. doi: 10.1007/s00299-020-02560-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Florindo H.F., et al. Immune-mediated approaches against COVID-19. Nat. Nanotechnol. 2020;15(8):630–645. doi: 10.1038/s41565-020-0732-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Jia H., Gong P. A structure-function diversity survey of the RNA-dependent RNA polymerases from the positive-strand RNA Viruses. Frontiers in Microbiology. 2019;10:1945. doi: 10.3389/fmicb.2019.01945. https://www.frontiersin.org/article/10.3389/fmicb.2019.01945 [Online]. Available. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Sexton N.R., Smith E.C., Blanc H., Vignuzzi M., Peersen O.B., Denison M.R. Homology-based identification of a mutation in the coronavirus RNA-dependent RNA polymerase that confers resistance to multiple mutagens. J. Virol. Jul. 2016;90(16):7415–7428. doi: 10.1128/JVI.00080-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Ayerdi O., et al. Preventive efficacy of tenofovir/emtricitabine against severe acute respiratory syndrome coronavirus 2 among pre-exposure prophylaxis users. Open Forum Infect. Dis. Nov. 2020;7(11):455. doi: 10.1093/ofid/ofaa455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Wang Y., et al. Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2020;395(10236):1569–1578. doi: 10.1016/S0140-6736(20)31022-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Eastman R.T., et al. Remdesivir: a review of its discovery and development leading to emergency use authorization for treatment of COVID-19. ACS Cent. Sci. 2020;6(5):672–683. doi: 10.1021/acscentsci.0c00489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Mehellou Y., Rattan H.S., Balzarini J. The ProTide prodrug technology: from the concept to the clinic. J. Med. Chem. Mar. 2018;61(6):2211–2226. doi: 10.1021/acs.jmedchem.7b00734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Jacobs M., et al. Late ebola virus relapse causing meningoencephalitis: a case report. Lancet. 2016;388(10043):498–503. doi: 10.1016/S0140-6736(16)30386-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Pizzorno A., Padey B., Dubois J., Julien T., Traversier A., Dulière V., Brun P., Lina B., Rosa-Calatrava M., Terrier O. In vitro evaluation of antiviral activity of single and combined repurposable drugs against SARS-CoV-2. Antivir. Res. 2020;181 doi: 10.1016/j.antiviral.2020.104878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Spinner C.D., Gottlieb R.L., Criner G.J., López J.R.A., Cattelan A.M., Viladomiu A.S., Ogbuagu O., Malhotra P., Mullane K.M., Castagna A., Chai L.Y.A. Effect of remdesivir vs standard care on clinical status at 11 days in patients with moderate COVID-19: a randomized clinical trial. JAMA. 2020;324(11):1048–1057. doi: 10.1001/jama.2020.16349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Beigel J.H., Tomashek K.M., Dodd L.E., Mehta A.K., Zingman B.S., Kalil A.C., Hohmann E., Chu H.Y., Luetkemeyer A., Kline S., Lopez de Castilla D. Remdesivir for the treatment of Covid-19—preliminary report. N. Engl. J. Med. 2020;383(19):1813–1826. doi: 10.1056/NEJMoa2007764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Hongchao P., Peto R., Henao-Restrepo A.M., Preziosi M.P., Sathiyamoorthy V., Karim Q.A., Alejandria M.M., Hernández García C., Kieny M.P., Malekzadeh R., Murthy S. 2020. Repurposed antiviral drugs for COVID-19: interim WHO solidarity trial results (Preprint) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Aleem A., Kothadia J. Remdesivir. StatPearls. 2021 [PubMed] [Google Scholar]
  • 319.https://www.who.int/news-room/feature-stories/detail/who-recommends-against-the-use-of-remdesivir-in-covid-19-patients
  • 320.https://go.drugbank.com/drugs/DB14761
  • 321.Furuta Y., Komeno T., Nakamura T. Favipiravir (T-705), a broad spectrum inhibitor of viral RNA polymerase. Proc. Jpn. Acad. Ser. B. Phys. Biol. Sci. 2017;93(7):449–463. doi: 10.2183/pjab.93.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.H. Sangawa, T. Komeno, H. Nishikawa, A. Yoshida, K. Takahashi, N. Nomura, Y. Furuta. Mechanism of action of T-705 ribosyl triphosphate against influenza virus RNA polymerase. Antimicrob. Agents Chemother. 2013;57(11):5202–5208. doi: 10.1128/AAC.00649-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Y. Furuta, K. Takahashi, Y. Fukuda, M. Kuno, T. Kamiyama, K. Kozaki, N. Nomura, H. Egawa, S. Minami, Y. Watanabe. In vitro and in vivo activities of anti-influenza virus compound T-705. Antimicrob. Agents Chemother. 2002;46(4):977–981. doi: 10.1128/aac.46.4.977-981.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Y. Furuta, K. Takahashi, K. Shiraki, K. Sakamoto, D.F. Smee, D.L. Barnard, B.B. Gowen, J.G. Julander, J.D. Morrey. T-705 (favipiravir) and related compounds: novel broad-spectrum inhibitors of RNA viral infections. Antivir. Res. 2009;82(3):95–102. doi: 10.1016/j.antiviral.2009.02.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325.V’kovski P., et al. Coronavirus biology and replication: implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021;19:155–170. doi: 10.1038/s41579-020-00468-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.S. Joshi, J. Parkar, A. Ansari, A. Vora, D. Talwar, M. Tiwaskar, S. Patil, H. Barkate. Role of favipiravir in the treatment of COVID-19. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2021;102:501–508. doi: 10.1016/j.ijid.2020.10.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.E. Koshi, S. Saito, M. Okazaki, Y. Toyama, T. Ishimoto, T. Kosugi, H. Hiraiwa, N. Jingushi, T. Yamamoto, M. Ozaki. Efficacy of favipiravir for an end stage renal disease patient on maintenance hemodialysis infected with novel coronavirus disease 2019. CEN Case Rep. 2021;10(1):126–131. doi: 10.1007/s13730-020-00534-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.A. Shannon, B. Selisko, N. Le, J. Huchting, F. Touret, G. Piorkowski, V. Fattorini, F. Ferron, E. Decroly, C. Meier. Favipiravir strikes the SARS-CoV-2 at its Achilles heel, the RNA polymerase. bioRxiv Prepr. Serv. Biol. 2020 doi: 10.1101/2020.05.15.098731. [DOI] [Google Scholar]
  • 329.Q. Cai, M. Yang, D. Liu, J. Chen, D. Shu, J. Xia, X. Liao, Y. Gu, Y. Yang. Experimental treatment with favipiravir for COVID-19: an open-label control study. Engineering (Beijing) 2020;6(10):1192–1198. doi: 10.1016/j.eng.2020.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.https://go.drugbank.com/drugs/DB12466
  • 331.Appleby T.C., et al. Viral replication. Structural basis for RNA replication by the hepatitis C virus polymerase. Science. Feb. 2015;347(6223):771–775. doi: 10.1126/science.1259210. [DOI] [PubMed] [Google Scholar]
  • 332.Jácome R., Campillo-Balderas J.A., Ponce de León S., Becerra A., Lazcano A. Sofosbuvir as a potential alternative to treat the SARS-CoV-2 epidemic. Sci. Rep. 2020;10(1):9294. doi: 10.1038/s41598-020-66440-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.A.A. Elfiky. Ribavirin, remdesivir, sofosbuvir, galidesivir, and tenofovir against SARS-CoV-2 RNA dependent RNA polymerase (RdRp): a molecular docking study. Life Sci. 2020;253 doi: 10.1016/j.lfs.2020.117592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.https://go.drugbank.com/drugs/DB08934
  • 335.Götz V., et al. Influenza a viruses escape from MxA restriction at the expense of efficient nuclear vRNP import. Sci. Rep. 2016;6(1):23138. doi: 10.1038/srep23138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Tay M.Y.F., et al. Nuclear localization of dengue virus (DENV) 1–4 non-structural protein 5; protection against all 4 DENV serotypes by the inhibitor ivermectin. Antivir. Res. Sep. 2013;99(3):301–306. doi: 10.1016/j.antiviral.2013.06.002. [DOI] [PubMed] [Google Scholar]
  • 337.Henriquez‐Camacho C., et al. Ivermectin versus albendazole or thiabendazole for Strongyloides stercoralis infection. Cochrane Database Syst. Rev. 2016;(1) doi: 10.1002/14651858.CD007745.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Buonfrate D., Salas-Coronas J., Muñoz J., Maruri B.T., Rodari P., Castelli F., Zammarchi L., Bianchi L., Gobbi F., Cabezas-Fernández T., Requena-Mendez A. Multiple-dose versus single-dose ivermectin for strongyloides stercoralis infection (Strong treat 1 to 4): a multicentre, open-label, phase 3, randomised controlled superiority trial. Lancet Infect. Dis. 2019;19(11):1181–1190. doi: 10.1016/S1473-3099(19)30289-0. [DOI] [PubMed] [Google Scholar]
  • 339.Wagstaff K.M., Sivakumaran H., Heaton S.M., Harrich D., Jans D.A. Ivermectin is a specific inhibitor of importin α/β-mediated nuclear import able to inhibit replication of HIV-1 and dengue virus. Biochem. J. May 2012;443(3):851–856. doi: 10.1042/BJ20120150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Chen I.-S., Kubo Y. Ivermectin and its target molecules: shared and unique modulation mechanisms of ion channels and receptors by ivermectin. J. Physiol. May 2018;596(10):1833–1845. doi: 10.1113/JP275236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Zhang X. Ivermectin inhibits LPS-induced production of inflammatory cytokines and improves LPS-induced survival in mice. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. Nov. 2008;57(11):524–529. doi: 10.1007/s00011-008-8007-8. [DOI] [PubMed] [Google Scholar]
  • 342.Caly L., Druce J.D., Catton M.G., Jans D.A., Wagstaff K.M. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antivir. Res. 2020;178 doi: 10.1016/j.antiviral.2020.104787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Sham H.L., et al. ABT-378, a highly potent inhibitor of the human immunodeficiency virus protease. Antimicrob. Agents Chemother. Dec. 1998;42(12):3218–3224. doi: 10.1128/AAC.42.12.3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Lv Z., Chu Y., Wang Y. HIV protease inhibitors: a review of molecular selectivity and toxicity. HIV. AIDS. (Auckl) Apr. 2015;7:95–104. doi: 10.2147/HIV.S79956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Sundquist W.I., Kräusslich H.-G. HIV-1 assembly, budding, and maturation. Cold Spring Harb. Perspect. Med. 2012;2(7) doi: 10.1101/cshperspect.a006924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Hung I.F.N., et al. Triple combination of interferon beta-1b, lopinavir–ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet. 2020;395(10238):1695–1704. doi: 10.1016/S0140-6736(20)31042-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.https://go.drugbank.com/drugs/DB01601
  • 348.Chen Z. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial. Biomed. Pharmacother. 2020 doi: 10.1101/2020.03.22.20040758. [DOI] [Google Scholar]
  • 349.Uzunova K., et al. Insights into antiviral mechanisms of remdesivir, lopinavir/ritonavir and chloroquine/hydroxychloroquine affecting the new SARS-CoV-2. Biomed. Pharmacother. 2020;131 doi: 10.1016/j.biopha.2020.110668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Cao B., et al. A trial of lopinavir-ritonavir in adults hospitalized with severe Covid-19. N. Engl. J. Med. Mar. 2020;382(19):1787–1799. doi: 10.1056/NEJMoa2001282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Khalili J.S., Zhu H., Mak N.S.A., Yan Y., Zhu Y. Novel coronavirus treatment with ribavirin: groundwork for an evaluation concerning COVID-19. J. Med. Virol. Jul. 2020;92(7):740–746. doi: 10.1002/jmv.25798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Tong S., et al. Ribavirin therapy for severe COVID-19: a retrospective cohort study Agents. Int. J. Antimicrob. Sep. 2020;56(3) doi: 10.1016/j.ijantimicag.2020.106114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Gordon C.J., et al. Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency. J. Biol. Chem. Sep. 2020;295(20):6785–6797. doi: 10.1074/jbc.RA120.013679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Du Y.-X., Chen X.-P. Favipiravir: pharmacokinetics and concerns about clinical trials for 2019-nCoV infection. Clin. Pharmacol. Ther. Aug. 2020;108(2):242–247. doi: 10.1002/cpt.1844. [DOI] [PubMed] [Google Scholar]
  • 355.Coomes E.A., Haghbayan H. Interleukin-6 in COVID-19: a systematic review and meta-analysis. medRxiv p. 2020.03.30.20048058. Jan. 2020 doi: 10.1101/2020.03.30.20048058. [et al.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Giamarellos-Bourboulis E.J., et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe. Jun. 2020;27(6):992–1000.e3. doi: 10.1016/j.chom.2020.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Sanders J.M., Monogue M.L., Jodlowski T.Z., Cutrell J.B. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA. May 2020;323(18):1824–1836. doi: 10.1001/jama.2020.6019. [DOI] [PubMed] [Google Scholar]
  • 358.Bollo L., et al. Seroconversion and indolent course of COVID-19 in patients with multiple sclerosis treated with fingolimod and teriflunomide. J. Neurol. Sci. Sep. 2020;416 doi: 10.1016/j.jns.2020.117011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Ucciferri C. Canakinumab in a subgroup of patients with COVID-19. Lancet. Rheumatol. Aug. 2020;2(8):457–458. doi: 10.1016/S2665-9913(20)30167-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Khan A.R., Soneja M., George N., Wig N. Anakinra for severe forms of COVID-19. Lancet Rheumatol. Oct. 2020;2(10):586–587. doi: 10.1016/S2665-9913(20)30273-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.Zhang Y., et al. Coagulopathy and antiphospholipid antibodies in patients with Covid-19. N. Engl. J. Med. Apr. 2020;382(17):38. doi: 10.1056/NEJMc2007575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Fogarty H., et al. COVID19 coagulopathy in caucasian patients. Br. J. Haematol. Jun. 2020;189(6):1044–1049. doi: 10.1111/bjh.16749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Tang N., Li D., Wang X., Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. Apr. 2020;18(4):844–847. doi: 10.1111/jth.14768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Gozzo L., Viale P., Longo L., Vitale D.C., Drago F. The potential role of heparin in patients with COVID-19: beyond the anticoagulant effect. A review. Front. Pharmacol. Aug. 2020;11:1307. doi: 10.3389/fphar.2020.01307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Cantini F., Niccoli L., Matarrese D., Nicastri E., Stobbione P., Goletti D. Baricitinib therapy in COVID-19: a pilot study on safety and clinical impact. J. Infect. Aug. 2020;81(2):318–356. doi: 10.1016/j.jinf.2020.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Richardson P., et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020;395(10223):30–31. doi: 10.1016/S0140-6736(20)30304-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Cao Y., et al. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID-19): a multicenter, single-blind, randomized controlled trial. J. Allergy Clin. Immunol. Jul. 2020;146(1):137–146. doi: 10.1016/j.jaci.2020.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Lian N., Xie H., Lin S., Huang J., Zhao J., Lin Q. Umifenovir treatment is not associated with improved outcomes in patients with coronavirus disease 2019: a retrospective study. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. Jul. 2020;26(7):917–921. doi: 10.1016/j.cmi.2020.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Seifirad S. Pirfenidone: a novel hypothetical treatment for COVID-19. Med. Hypotheses. Nov. 2020;144 doi: 10.1016/j.mehy.2020.110005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Uno Y. Camostat mesilate therapy for COVID-19. Intern. Emerg. Med. Nov. 2020;15(8):1577–1578. doi: 10.1007/s11739-020-02345-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Chen J., et al. Antiviral activity and safety of darunavir/cobicistat for the treatment of COVID-19. Open Forum Infect. Dis. 2020;7(7):1–5. doi: 10.1093/ofid/ofaa241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Fu W., et al. A clinical pilot study on the safety and efficacy of aerosol inhalation treatment of IFN-κ plus TFF2 in patients with moderate COVID-19. EClinicalMedicine. 2020;25 doi: 10.1016/j.eclinm.2020.100478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Tan Q., et al. Is oseltamivir suitable for fighting against COVID-19: in silico assessment, in vitro and retrospective study. Bioorg. Chem. Nov. 2020;104 doi: 10.1016/j.bioorg.2020.104257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Lou Y., et al. Clinical outcomes and plasma concentrations of baloxavir marboxil and favipiravir in COVID-19 patients: an exploratory randomized, controlled trial. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. Feb. 2021;157 doi: 10.1016/j.ejps.2020.105631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Dai W. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science. 2020;368(6497):1331–1335. doi: 10.1126/science.abb4489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Prediction of the SARS-CoV-2 (2019-nCoV) 3C-like protease (3CLpro) structure: Virtual screening reveals velpatasvir, ledipasvir, and other drug repurposing candidates. F1000Research. 2020;9:1–17. doi: 10.12688/f1000research.22457.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Intranasal Vaccines for SARS-CoV-2: From Challenges to Potential in COVID-19 Management, Vivek P. Chavda , Lalitkumar K. Vora , Anjali K. Pandya , Vandana B. Patravale. [DOI] [PMC free article] [PubMed]
  • 378.SaNOtize NORSTMas An Upper Airway ‘Disinfectant.’. https://1236sanotize.com/covid-19/
  • 379.APEPTICO Forschung und Entwicklung GmbH . APEPTICO’s global survey on incidence of ARDS and outcomes in hospitalized patients with COVID-19 has been published by Critical Care. www.apeptico.com/index-news [accessed July 1240 2, 2021].
  • 380.F. Buller Neurimmune and Ethris Sign Collaboration Agreement to Rapidly Develop Inhaled mRNA-based Antibody Therapy for the Treatment of Covid-19. Neurimmune AG. www.neurimmune.com/news/neurimmune-and-ethris-sign-collaboration-agreement-to-rapidly-develop-inhaled-mrna-based-antibody-therapy-for-the-treatment-of-covid-19.
  • 381.ClinicalTrials.gov NCT04330586. A Trial of ciclesonide in adults with mild-to- moderate COVID-19. https://clinicaltrials.gov/ct2/show/NCT04330586
  • 382.Liebowitz Michael R., Ester Salman N.R., Nicolini Humberto. Effect of an acute intranasal aerosol dose of PH94B on social and performance anxiety in women with social anxiety disorder. Am. J. Psychiatry. 2014;171(6):675–682. doi: 10.1176/appi.ajp.2014.12101342. [DOI] [PubMed] [Google Scholar]
  • 383.EudraCT: 2020-001023-14 A randomized double-blind placebo-controlled trial 1266 to determine the safety and efficacy of inhaled SNG001 (IFNb-1a for 1267 nebulization) for the treatment of patients with confirmed SARS-CoV-2 1268 infection (COVID-19) www.clinicaltrialsregister.eu/ctr-search/trial/2020-1269001023-14/GB
  • 384.Therapeutics Partner. Partner Therapeutics initiates patient enrollment in U.S. 1253 clinical trial evaluating Leukine (rhuGM-CSF, sargramostim) in COVID-19 1254 patients. www.partnertx.com/partner-therapeutics-initiates-patient-enrollment-1255in-u-s-clinical-trial-evaluating-leukine-rhugm-csf-sargramostim-in-covid-19-1256patients
  • 385.Haschke M., Kadhiresan P., Kozlowski H.N., Malekjahani A., Osborne M., Li V.Y.C., Chen H., Mubareka S., Gubbay J.B., Chan W.C.W. Pharmacokinetics and pharmacodynamics of recombinant human angiotensin-converting enzyme 2 in healthy human subjects. Clin. Pharmacokinet. 2013;52:783–792. doi: 10.1007/s40262-013-0072-7. [DOI] [PubMed] [Google Scholar]
  • 386.Monteil V., Kwon H., Prado P., Hagelkrüys A., Wimmer R.A., Stahl M., Leopoldi A., Garreta E., Pozo C.H.D., Prosper F., Romero G.P., Wirnsberger G., Zhang H., Slutsky A.S., Conder R., Montserrat N., Mirazimi A., Penninger J.M. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2, 181 (2020) 905–913. Cell. 2020;181(4):905–913. doi: 10.1016/j.cell.2020.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387.Khan A., Benthin C., Zeno B., Albertson T.E, Boyd J.D., Christie J., Hall R., Poirier G., Poirier J.J., Tidswell M., Hardes K., Powley W.M., Wright T.J., Siederer S.K., Fairman D.A., Lipson D.A., Bayliffe A.I., Lazaar A.L. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit. Care. 2017;21:1–9. doi: 10.1186/s13054-017-1823-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Savarino A., Boelaert J.R., Majori G., Cauda V., et al. Effects of chloroquine on viral infections: an old drug against today’s diseases. Lancet Infect Dis. 2003;3:722–727. doi: 10.1016/s1473-3099(03)00806-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Wang M., Cao R., Zhang X., Yang L., Liu J., Xu M., Shi Z., Hu Z., Zhong W., Xhio G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30:269–271. doi: 10.1038/s41422-020-0282-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390.Vincent M.J., Bergeron E., Benjannet S., Erickson B.R., Rollin P.E., Ksiazek T.G., Seidah N.G., Nichol S.T. Chloroquine is a potent inhibitor of SARS coronavirus infection and spread. Virol. J. 2005;2:1–10. doi: 10.1186/1743-422X-2-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Chen L., et al. The effects of chloroquine and hydroxychloroquine on ACE2-related coronavirus pathology and the cardiovascular system: an evidence-based review. Function. 2020;1(2) doi: 10.1093/function/zqaa012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Roldan E.Q., Biasiotto G., Magro P., Zanella I., Pharmacol. Res. The possible mechanisms of action of 4-aminoquinolines (chloroquine/hydroxychloroquine) against Sars-Cov-2 infection (COVID-19): A role for iron homeostasis? 158 (2020) 104904. Radiology. 2020;158:104904. doi: 10.1016/j.phrs.2020.104904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Yao X., Ye F., Zhang M., Huang B., Niu P., Liu L., Zhao X., Song C, Zhan S, Lu R, Li H, Tan W, Liu D. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Clin. Infect. Dis. 2020;71(15):732–739. doi: 10.1093/cid/ciaa237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394.Rosenberg, E.S., Baillie C. K, Herold H. M, Volkwein W., Guertler P., Eberle U., Nikolaus N., Sing A., Pavlovic M., Goerlich O., Busch U., Wassill L., Huber I., Baiker A. Association of treatment with hydroxychloroquine or azithromycin with in-hospital mortality in patients with COVID-19 in New York State. Jama. 2020;323(24):2493–2502. doi: 10.1001/jama.2020.8630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 395.Vagner J., Qu H., Hruby V.J. Peptidomimetics, a synthetic tool of drug discovery. Curr. Opin. Chem. Biol. 2008;12(3):292–296. doi: 10.1016/j.cbpa.2008.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396.Yan, R., Zhang, Y., Li, Y., Guo, L., Zhuo, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367:1444–1448. doi: 10.1126/science.abb2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Millet J.K., Whittaker G.R. Physiological and molecular triggers for SARS-CoV membrane fusion and entry into host cells. Virology. 2018;517:3–8. doi: 10.1016/j.virol.2017.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Xia, S., Zhu Y., Liu M., Lan Q., Xu W., Wu Y., Ying T., Liu S., Shi Z., Jiang S., Lu L. Fusion mechanism of 2019-nCoV and fusion inhibitors targeting HR1 domain in spike protein. Cell. Mol. Immunol. 2020;17:765–767. doi: 10.1038/s41423-020-0374-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Xiu S., Dick A., Ju H., Mirzaie S., Abdi F., Cocklin S., Zhan X., Liu P. Inhibitors of SARS-CoV-2 entry: current and future opportunities. J. Med. Chem. 2020;63:12256–12274. doi: 10.1021/acs.jmedchem.0c00502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400.Xia S., et al. A pan-coronavirus fusion inhibitor targeting the HR1 domain of human coronavirus spike. Sci. Adv. 2019;5(4):4580. doi: 10.1126/sciadv.aav4580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401.Xia S., Liu M., Wang C., Xu W., Lan Q., Qi F., Bao L., Du L., Liu S., Sun C., Sun F., Shi Z., Zhu Y., Jiang S., Lu L. 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;30:343–355. doi: 10.1038/s41422-020-0305-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Y. Zhu, Yu Danwei, Yan Hongxia, Chong Huihui, He Yuxian. Design of potent membrane fusion inhibitors against SARS-CoV-2, an emerging coronavirus with high fusogenic activity. J. Virol. 2020;94:e00635–20. doi: 10.1128/JVI.00635-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.G. Zhang, S. Pomplun, A.R. Loftis, A. Loas, B.L. Pentelute. The first-in-class peptide binder to the SARS-CoV-2 spike protein. BioRxiv. 2020 doi: 10.1101/2020.03.19.999318. [DOI] [Google Scholar]
  • 404.Y. Han, P. Král. Computational design of ACE2-based peptide inhibitors of SARS-CoV-2. ACS Nano. 2020;14(4):5143–5147. doi: 10.1021/acsnano.0c02857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 405.D. Barh, S. Tiwari, B.S. Andrade, M. Giovanetti, R. Kumavath, P. Ghosh, A. Góes Neto, L.C.J. Alcantara, V. Azevedo. Potential chimeric peptides to block the SARS-CoV-2 spike RBD. F1000Res. 2020;576(9) doi: 10.12688/f1000research.24074.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406.X. Huang, R. Pearce, Y. Zhang. Computational design of peptides to block binding of the SARS-CoV-2 spike protein to human ACE2. ACS Nano. 2020;14(4):5143–5147. doi: 10.1021/acsnano.0c02857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407.S. VanPatten, M. He, A. Altiti, K. F Cheng, M.H. Ghanem, Y. Al-Abed. Evidence supporting the use of peptides and peptidomimetics as potential SARS-CoV-2 (COVID-19) therapeutics. Future Med. Chem. 2020;12:1647–1656. doi: 10.4155/fmc-2020-0180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408.R. Cannalire, I. Stefanelli, C. Cerchia, A.R. Beccari, S. Pelliccia, V. Summa. SARS-CoV-2 entry inhibitors: Small molecules and peptides targeting virus or host cells. Int. J. Mol. Sci. 2020;21(16):5707–5787. doi: 10.3390/ijms21165707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409.M. Yamamoto, S. Matsuyama, X. Li, M. Takeda, Y. Kawaguchi, J. Inoue, Z. Matsuda. Identification of nafamostat as a potent inhibitor of Middle East respiratory syndrome coronavirus S protein-mediated membrane fusion using the split-protein-based cell-cell fusion assay. Science. 2016;60(11):6532–6539. doi: 10.1128/AAC.01043-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410.M. Kawase, K. Shirato, L. van der Hoek, F. Taguchi, S. Matsuyama. Simultaneous treatment of human bronchial epithelial cells with serine and cysteine protease inhibitors prevents severe acute respiratory syndrome coronavirus entry. J. Virol. 2012;86:6537–6545. doi: 10.1128/JVI.00094-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.A. Shulla, T. Heald-Sargent, G. Subramanya, J. Zhao, S. Perlman, T. Gallagher. A transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entry. J. Virol. 2011:873–882. doi: 10.1128/JVI.02062-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412.Zhou Y., et al. Protease inhibitors targeting coronavirus and filovirus entry. Antiviral Res. 2015:76–84. doi: 10.1016/j.antiviral.2015.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Shah P.P., et al. A small-molecule oxocarbazate inhibitor of human cathepsin L blocks severe acute respiratory syndrome and ebola pseudotype virus infection into human embryonic kidney 293T cells. Mol. Pharmacol. 2010;78:319–324. doi: 10.1124/mol.110.064261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Myers M.C., et al. Design, synthesis, and evaluation of inhibitors of cathepsin L: Exploiting a unique thiocarbazate chemotype. Bioorg. Med. Chem. Lett. 2008;18(4):3646–3651. doi: 10.1016/j.bmcl.2008.04.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.M.C. Myers, P.P. Shah, M.P. Beavers, A.D. Napper, S.L. Diamond, A.B. Smith III, D.M. Huryn. Molecular docking of cathepsin L inhibitors in the binding site of papain. J. Chem. Inf. Model. 2008;48:1464–1472. doi: 10.1021/ci800085c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Runfeng L., et al. Lianhuaqingwen exerts anti-viral and anti-inflammatory activity against novel coronavirus (SARS-CoV-2) Pharmacol. Res. 2020;156 doi: 10.1016/j.phrs.2020.104761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.S. Rastogi, D.N. Pandey, R.H. Singh. COVID-19 pandemic: A pragmatic plan for ayurveda intervention, (2020). J. Ayurveda Integr. Med. 2020:337–340. doi: 10.1016/j.jviromet.2013.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.J. Lung, et al. The potential chemical structure of anti‐SARS‐CoV‐2 RNA‐dependent RNA polymerase. J. Med. Virol. 2020;92:693–697. doi: 10.1016/j.jcf.2007.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.D. Zhang, K. Wu, X. Zhang, S. Deng, B. Peng. In silico screening of Chinese herbal medicines with the potential to directly inhibit 2019 novel coronavirus. J. Integr. Med. 2020;18:152–158. doi: 10.1016/j.jviromet.2013.07.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420.M.T. ul Qamar, S.M. Alqahtani, M.A. Alamri, L.-L. Chen. Structural basis of SARS-CoV-2 3CLpro and anti-COVID-19 drug discovery from medicinal plants. J. Pharm. Anal. 2020;10:313–319. doi: 10.1016/j.jpha.2020.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.I. Jahan, O. Ahmet. Potentials of plant-based substance to inhabit and probable cure for the COVID-19. Turkish J. Biol. 2020;44(0):228–241. doi: 10.1128/JCM.37.4.937-943.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.E. Shawky, A.A. Nada, R.S. Ibrahim. Potential role of medicinal plants and their constituents in the mitigation of SARS-CoV-2: identifying related therapeutic targets using network pharmacology and molecular docking analyses. RSC Adv. 2020;10:27961–27983. doi: 10.1039/D0RA05126H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Chojnacka K., Witek-Krowiak A., Skrzypczak D., Mikula K., Młynarz P. Phytochemicals containing biologically active polyphenols as an effective agent against Covid-19-inducing coronavirus. J. Funct. Foods. 2020 doi: 10.1016/j.jff.2020.104146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Tohidi B., Rahimmalek M., Arzani A. Essential oil composition, total phenolic, flavonoid contents, and antioxidant activity of thymus species collected from different regions of Iran. Food Chem. 2017;220:153–161. doi: 10.1016/j.foodchem.2016.09.203. [DOI] [PubMed] [Google Scholar]
  • 425.Ma L., Yao L. Antiviral effects of plant-derived essential oils and their components: an updated review. Molecules. 2020;25(11):2627. doi: 10.3390/molecules25112627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 426.Mahmood N., Nasir S.B., Hefferon K. Plant-based drugs and vaccines for COVID-19. Vaccines. 2021;9(1):15. doi: 10.3390/vaccines9010015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Gretler, C., Tobacco-Based Coronavirus Vaccine Poised for Human Tests Bloomberg, May 15.
  • 428.Palca J. 2020. Tobacco Plants Contribute Key Ingredient For COVID-19 Vaccine. [Google Scholar]
  • 429.Shanmugaraj B., Siriwattananon K., Malla A., Phoolcharoen W. Potential for developing plant-derived candidate vaccines and biologics against emerging coronavirus infections. Pathogens. 2021;10(8):1051. doi: 10.3390/pathogens10081051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 430.Krenek P., Samajova O., Luptovciak I., Doskocilova A., Komis G., Samaj J. Transient plant transformation mediated by agrobacterium tumefaciens: principles, methods and applications. Biotechnol. Adv. 2015;33(6):1024–1042. doi: 10.1016/j.biotechadv.2015.03.012. [DOI] [PubMed] [Google Scholar]
  • 431.Rosales-Mendoza S., Márquez-Escobar V.A., González-Ortega O., Nieto-Gómez R., Arévalo-Villalobos J.I. What does plant-based vaccine technology offer to the fight against COVID-19? Vaccines. 2020;8(2):183. doi: 10.3390/vaccines8020183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Pillet S., Couillard J., Trépanier S., Poulin J.F., Yassine-Diab B., Guy B., Ward B.J., Landry N. Immunogenicity and safety of a quadrivalent plant-derived virus like particle influenza vaccine candidate—Two randomized phase II clinical trials in 18 to 49 and≥ 50 years old adults. PloS one. 2019;14(6) doi: 10.1371/journal.pone.0216533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433.Wang C. Preparation and characterization of polyclonal antibody against severe acute respiratory syndrome-associated coronavirus spike protein., Hybridoma (Larchmt) 2010;29:511–516. doi: 10.1089/hyb.2010.0044. [DOI] [PubMed] [Google Scholar]
  • 434.Pogrebnyak N., Golovkin M., Andrianov V., Spitsin S., Smirnov Y., Egolf R., Koprowski H. Severe acute respiratory syndrome (SARS) S protein production in plants: development of recombinant vaccine. Proc. Natl. Acad. Sci. 2005;102(25):9062–9067. doi: 10.1073/pnas.0503760102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 435.https://www.cbc.ca/news/health/medicago-covid-19-vaccine-phase-1-1.5796411
  • 436.ClinicalTrial - KBP-201 COVID-19 Vaccine. https://clinicaltrials.gov/ct2/show/NCT04473690
  • 437.https://ibioinc.com/vaccines/ibio-202/
  • 438.Diego-Martin B., González B., Vazquez-Vilar M., Selma S., Mateos-Fernández R., Gianoglio S., Fernández-del-Carmen A., Orzáez D. Pilot production of SARS-CoV-2 related proteins in plants: a proof of concept for rapid repurposing of indoor farms into biomanufacturing facilities. Front. Plant Sci. 2020;11:2101. doi: 10.3389/fpls.2020.612781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 439.Mamedov T., et al. Engineering, production and characterization of Spike and Nucleocapsid structural proteins of SARS–CoV-2 in Nicotiana benthamiana as vaccine candidates against COVID-19. BioRxiv. (2020) 2020.12.29.424779. 2020 doi: 10.1101/2020.12.29.424779. [DOI] [Google Scholar]
  • 440.BPP - COVID-19 Vaccine Development. https://baiyaphytopharm.com/covid-19
  • 441.Zheng N., Xia R., Yang C., Yin B., Li Y., Duan C., Liang L., Guo H., Xie Q. Boosted expression of the SARS-CoV nucleocapsid protein in tobacco and its immunogenicity in mice. Vaccine. 2009;27(36):5001–5007. doi: 10.1016/j.vaccine.2009.05.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 442.Siriwattananon K., et al. Plant-Produced Receptor-Binding Domain of SARS-CoV-2 Elicits Potent Neutralizing Responses in Mice and Non-human Primates. Front. Plant Sci. 2021;12 doi: 10.3389/fpls.2021.682953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Li H.Y., Ramalingam S., Chye M.L. Accumulation of recombinant SARS-CoV spike protein in plant cytosol and chloroplasts indicate potential for development of plant-derived oral vaccines. Exp. Biol. Med. 2006;231(8):1346–1352. doi: 10.1177/153537020623100808. [DOI] [PubMed] [Google Scholar]
  • 444.Pittenger M.F., et al. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen. Med. 2019;4:22. doi: 10.1038/s41536-019-0083-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Kyurkchiev D., Bochev I., Ivanova-Todorova E., Mourdjeva M., Oreshkova T., Belemezova K., Kyurkchiev S. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J. Stem Cells. 2014;6(5):552. doi: 10.4252/wjsc.v6.i5.552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 446.Nie W.B., Zhang D., Wang L.S. Growth factor gene-modified mesenchymal stem cells in tissue regeneration. Drug Des. Dev. Ther. 2020;14:1241. doi: 10.2147/DDDT.S243944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 447.Jian Y., Yang K., Sun X., Zhao J., Huang K., Aldanakh A., Xu Z., Wu H., Xu Q., Zhang L., Xu C. Current advance of immune evasion mechanisms and emerging immunotherapies in renal cell carcinoma. Front. Immunol. 2021;12:502. doi: 10.3389/fimmu.2021.639636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 448.Chan M.C.W., Kuok D.I.T., Leung C.Y.H., Hui K.P.Y., Valkenburg S.A., Lau E.H.Y., Nicholls J.M., Fang X., Guan Y., Lee J.W., Chan R.W.Y., Webster R.G., Matthay M.A., Peiris J.S.M., et al. Human mesenchymal stromal cells reduce influenza a H5N1-associated acute lung injury in vitro and in vivo. Proc. Natl. Acad. Sci. U. S. A. 2016;113:3621–3626. doi: 10.1073/pnas.1601911113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Chen J., Hu C., Chen L., Tang L., Zhu Y., Xu X., Chen L., Gao, H., Lu X., Yu L., Dai X., Xiang C., Lia L. Clinical Study of Mesenchymal Stem Cell Treatment for Acute Respiratory Distress Syndrome Induced by Epidemic Influenza A (H7N9) Infection: a Hint for COVID-19 Treatment. Engineering (Beijing) 2020;6(10):1153–1161. doi: 10.1016/j.eng.2020.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 450.Matthay M.A., et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomized phase 2a safety trial. Lancet Respir. Med. 2019;7:154–162. doi: 10.1016/S2213-2600(18)30418-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 451.Tang L., et al. Clinical study using mesenchymal stem cells for the treatment of patients with severe COVID-19. Front. Med. 2020;14(5):664–673. doi: 10.1007/s11684-020-0810-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 452.Meng F., et al. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trial. Signal Transduct. Target. Ther. 2020;5:172. doi: 10.1038/s41392-020-00286-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 453.Hashemian S.-M.R., et al. Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case series. Stem Cell Res Ther. 2021;12(1):91. doi: 10.1186/s13287-021-02165-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 454.Mosquera-Yuqui F., et al. Targeting the 3CLpro and RdRp of SARS-CoV-2 with phytochemicals from medicinal plants of the Andean Region: molecular docking and molecular dynamics simulations. J. Biomol. Struct. Dyn. 2020:1–14. doi: 10.1080/07391102.2020.1835716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 455.Bosken Y.K. Insights Into Dynamics of Inhibitor and Ubiquitin-Like Protein Binding in SARS-CoV-2 Papain-Like Protease. Front. Mol. Biosci. 2020;7 doi: 10.3389/fmolb.2020.00174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 456.Chu C.K., et al. Antiviral activity of nucleoside analogues against SARS-coronavirus (SARS-coV) Antivir. Chem. Chemother. 2006;17(5):285–289. doi: 10.1177/095632020601700506. [DOI] [PubMed] [Google Scholar]
  • 457.De Vries M., Mohamed A.S., Prescott R.A., Valero-Jimenez A.M., Desvignes L., O’Connor R., Steppan C., Devlin J.C., Ivanova E., Herrera A., Schinlever A. A comparative analysis of SARS-CoV-2 antivirals characterizes 3CLpro inhibitor PF-00835231 as a potential new treatment for COVID-19. J. Virol. 2021;95(10):e01819–e01820. doi: 10.1128/JVI.01819-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 458.Chan J.F.W., Yip C.C.Y., To K.K.W., Tang T.H.C., Wong S.C.Y., Leung K.H., Fung A.Y.F., Ng A.C.K., Zou Z., Tsoi H.W., Choi G.K.Y. Improved molecular diagnosis of COVID-19 by the novel, highly sensitive and specific COVID-19-RdRp/Hel real-time reverse transcription-PCR assay validated in vitro and with clinical specimens. J. Clin. Microbiol. 2020;58(5):e00310–e00320. doi: 10.1128/JCM.00310-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 459.Burton D.R. What are the Most powerful immunogen design vaccine Strategies? Reverse vaccinology 2.0 shows great promise. Cold Spring Harb. Perspect. Biol. 2017;9(11):Nov. doi: 10.1101/cshperspect.a030262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 460.Graham B.S. Advances in antiviral vaccine development. Immunol. Rev. Sep. 2013;255(1):230–242. doi: 10.1111/imr.12098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 461.Amanat F., Krammer F. SARS-CoV-2 vaccines: status report. Immunity. Apr. 2020;52(4):583–589. doi: 10.1016/j.immuni.2020.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 462.Roldão A., Mellado M.C.M., Castilho L.R., Carrondo M.J., Alves P.M. Virus-like particles in vaccine development. Expert Rev. Vaccines. 2010;9(10):1149–1176. doi: 10.1586/erv.10.115. [DOI] [PubMed] [Google Scholar]
  • 463.Novak M.A., Hamel A.F., Kelly B.J., Dettmer A.M., Meyer J.S. Stress, the HPA axis, and nonhuman primate well-being: a review. Appl. Anim. Behav. Sci. 2013;143(2–4):135–149. doi: 10.1016/j.applanim.2012.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464.Zhao Q., Li S., Yu H., Xia N., Modis Y. Virus-like particle-based human vaccines: quality assessment based on structural and functional properties. Trends Biotechnol. 2013;31(11):654–663. doi: 10.1016/j.tibtech.2013.09.002. [DOI] [PubMed] [Google Scholar]
  • 465.Rawat K., Kumari P., Saha L. COVID-19 vaccine: A recent update in pipeline vaccines, their design and development strategies. European journal of pharmacology. 2020;892:173751. doi: 10.1016/j.ejphar.2020.173751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 466.Creech C.B., Walker S.C., Samuels R.J. SARS-CoV-2 vaccines. JAMA. 2021;325(13):1318–1320. doi: 10.1001/jama.2021.3199. [DOI] [PubMed] [Google Scholar]
  • 467.Kaur S.P. COVID-19 Vaccine: A comprehensive status report. Virus Res. 2020 doi: 10.1016/j.virusres.2020.198114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 468.Graham S.P. Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19. Npj Vaccines. 2020;5:1–6. doi: 10.1038/s41541-020-00221-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 469.Kim J.H., Jacob J. Vaccines for Pandemic Influenza. 2009. DNA vaccines against influenza viruses; pp. 197–210. [DOI] [PubMed] [Google Scholar]
  • 470.Xu S., et al. Mrna vaccine era—mechanisms, drug platform and clinical prospection. Int J Mol Sci . 2020;18 doi: 10.3390/ijms21186582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 471.Hekele A., Bertholet S., Archer J., Gibson D.G., Palladino G., Brito L.A., Otten G.R., Brazzoli M., Buccato S., Bonci A., Casini D. Rapidly produced SAM® vaccine against H7N9 influenza is immunogenic in mice. Emerg. Microbes Infect. 2013;2(1):1–7. doi: 10.1038/emi.2013.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 472.“draft-landscape-of-covid-19-candidate-vaccines @ www.who.int.” [Online]. Available: https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines.
  • 473.Kumar A., Meldgaard T.S., Bertholet S. Novel platforms for the development of a universal influenza vaccine. Front. Immunol. 2018;9:600. doi: 10.3389/fimmu.2018.00600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 474.Nascimento I.P., Leite L.C.C. Recombinant vaccines and the development of new vaccine strategies. Braz. J. Med. Biol. Res. 2012;45(12):1102–1111. doi: 10.1590/S0100-879X2012007500142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 475.Afkhami S., Yao Y., Xing Z. Methods and clinical development of adenovirus- vectored vaccines against mucosal pathogens. Mol. Ther. Methods Clin. Dev. 2016;3:16030. doi: 10.1038/mtm.2016.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476.University of Oxford University of Oxford to study nasal administration of COVID-19 vaccine. www.ox.ac.uk/news/2021-03-25-university-oxford-study-nasal-administration-covid-19-vaccine
  • 477.A.O. Hassan N.M. Kafai I.P. Dmitriev J.M. Fox B.K. Smith I.B. Harvey , et al., A single-dose intranasal ChAd vaccine protects upper and Lower Respiratory Tracts against SARS-CoV-2. [DOI] [PMC free article] [PubMed]
  • 478.Biotech Bharat. Intranasal Vaccine For Covid-19. www.bharatbiotech.com/1104intranasal-vaccine.html
  • 479.CanSino Biologics Inc A randomized, double-blind, placebo-controlled Phase I/ II clinical trial to evaluate the safety and immunogenicity of Ad5-NCoV for 1112 inhalation in adults 18 years of age and older. https://clinicaltrials.gov/ct2/1113 show/NCT04840992 [accessed July 2, 2021]. 1114.
  • 480.Inc Altimmune Phase 2, double-blind, randomized, placebo-controlled study of 1120 NasoVAX in the prevention of clinical worsening in patients with early Coronavirus infectious disease 2019 (COVID-19). https://clinicaltrials.gov/ct2/1122 show/NCT04442230 [accessed July 2, 2021].
  • 481.Inc FluGen. Focused on efficacy – M2SR:The Vaccine. http://flugen.com/m2sr/1130
  • 482.Clinical Research and Development Company – Bharat Biotech. www.1132bharatbiotech.com/r&d_pipeline.html 80-
  • 483.Bilsel P. M2SR, a single replication universal flu vaccine. Virol-Mycol. 2015;4:2. [Google Scholar]
  • 484.The University of Hong Kong. A Phase 1, randomized, double-blinded, placebo- 1135 controlled, dose-escalation and dose-expansion study to evaluate the safety and 1136 immunogenicity of DelNS1-NCoV-RBD LAIV for COVID-19 in healthy adults. 1137 https://clinicaltrials.gov/ct2/show/NCT04809389 [accessed July 2, 2021]. 1138. [DOI] [PMC free article] [PubMed]
  • 485.Arena Clinical Trials. Nasal spray vaccine for Covid-19. www.clinicaltrialsarena.com/comment/nasal-spray-vaccine-covid-19/
  • 486.<collab>Meissa Vaccines Inc.</collab>. Phase 1, open-label, dose-escalation study to evaluate 1145 tolerability, safety, and immunogenicity of an intranasal live attenuate respiratory syncytial virus vaccine expressing spike protein of SARS-CoV-2 in healthy adults ages 18–69 years. https://clinicaltrials.gov/ct2/show/1148NCT04798001
  • 487.Illinois Institute of Technology Promising new COVID-19 treatment in 1150 development at Illinois Tech. www.iit.edu/news/promising-new-covid-19-1151treatment-development-illinois-tech
  • 488.Intravacc Intravacc announces positive pre-clinical data for its SARS-CoV-2 nose 1153 spray vaccine. www.intravacc.nl/news/intravacc-announces-positive-pre-clinical-data-intranasal-sars-cov-2-candidate-vaccine/
  • 489.Koshy S.T., Cheung A.S., Gu L., Graveline A.R., Mooney D.J. Liposomal deliver enhances immune activation by STING agonists for cancer immunotherapy. Adv. Biosyst. 2017;1(1–2):1600013. doi: 10.1002/adbi.201600013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 490.Tannock G.A., Kim H., Xue L. Why are vaccines against many human viral diseases still unavailable; an historic perspective? J. Med. Virol. Feb. 2020;92(2):129–138. doi: 10.1002/jmv.25593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 491.Bharati K., Vrati S. Viral vaccines in India: an overview. Proc. Natl. Acad. Sci. India. Sect. B. 2012;82(1):181–198. doi: 10.1007/s40011-011-0014-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 492.Mario P., Padaon A., Schievano F., Vecchiato E., Lippi G., Cascio G.L., Porru S., Palù G. SARS-CoV-2 serosurvey in health care workers of the Veneto region. medRxiv, p. 2020.07.23.20160457. Jan. 2020 doi: 10.1101/2020.07.23.20160457. [DOI] [PubMed] [Google Scholar]
  • 493.Ulmer J.B., Valley U., Rappuoli R. Vaccine manufacturing: challenges and solutions. Nat. Biotechnol. Nov. 2006;24(11):1377–1383. doi: 10.1038/nbt1261. [DOI] [PubMed] [Google Scholar]
  • 494.Pulendran B., Ahmed R. Immunological mechanisms of vaccination. Nat. Immunol. 2011;12(6):509–517. doi: 10.1038/ni.2039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 495.Hodgson S.H., Mansatta K., Mallett G., Harris V., Emary K.R.W., Pollard A.J. What defines an efficacious COVID-19 vaccine? A review of the challenges assessing the clinical efficacy of vaccines against SARS-CoV-2. Lancet Infect. Dis. Feb. 2021;21(2):e26–e35. doi: 10.1016/S1473-3099(20)30773-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 496.Banerji A., et al. mRNA vaccines to prevent COVID-19 disease and reported allergic reactions: current evidence and suggested approach. J. Allergy Clin. Immunol. Pract. Apr. 2021;9(4):1423–1437. doi: 10.1016/j.jaip.2020.12.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 497.“joint-cdc-and-fda-statement-johnson-johnson-covid-19-vaccine @ www.fda.gov.” [Online]. Available: https://www.fda.gov/news-events/press-announcements/joint-cdc-and-fda-statement-johnson-johnson-covid-19-vaccine.
  • 498.https://www.who.int/initiatives/act-accelerator/covax/covid-19-vaccine-country-readiness-and-delivery
  • 499.Koirala A., Joo Y.J., Khatami A., Chiu C., Britton P.N. Vaccines for COVID-19: the current state of play. Paediatr. Respir. Rev. Sep. 2020;35:43–49. doi: 10.1016/j.prrv.2020.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 500.Papania M.J., Zehrung D., Jarrahian C. Plotkin’s Vaccines. 2018. Technologies to Improve Immunization; pp. 1320–1353.e17. [DOI] [Google Scholar]
  • 501.Dong Y., Dai T., Wei Y., Zhang L., Zheng M., Zhou F. A systematic review of SARS-CoV-2 vaccine candidates. Signal Transduct. Target. Ther. Oct. 2020;5(1):237. doi: 10.1038/s41392-020-00352-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 502.Thielmann A., Puth M.T., Weltermann B. Visual inspection of vaccine storage conditions in general practices: a study of 75 vaccine refrigerators. PLoS One. 2019;14(12):1–13. doi: 10.1371/journal.pone.0225764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 503.Kartoglu U.H., Moore K.L., Lloyd J.S. Logistical challenges for potential SARS-CoV-2 vaccine and a call to research institutions, developers and manufacturers. Vaccine. Jul. 2020;38(34):5393–5395. doi: 10.1016/j.vaccine.2020.06.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 504.Traynor K. COVID-19 vaccination campaigns take shape. Am. J. Heal. Pharm. AJHP Off. J. Am. Soc. Heal. Pharm. Feb. 2021;78(4):282–284. doi: 10.1093/ajhp/zxab019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 505.Bell K.N., Hogue C.J., Manning C., Kendal A.P. Risk factors for improper vaccine storage and handling in private provider offices. Pediatrics. Jun. 2001;107(6):E100. doi: 10.1542/peds.107.6.e100. [DOI] [PubMed] [Google Scholar]
  • 506.Beirne P.V., Hennessy S., Cadogan S.L., Shiely F., Fitzgerald T., MacLeod F. Needle size for vaccination procedures in children and adolescents. Cochrane database Syst. Rev. Aug. 2018;8(8) doi: 10.1002/14651858.CD010720.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 507.Feinmann J. Why aren’t covid-19 vaccines being manufactured in standard prefilled syringes? BMJ. Feb. 2021;372 doi: 10.1136/bmj.n263. [DOI] [PubMed] [Google Scholar]
  • 508.https://www.who.int/emergencies/diseases/novel-coronavirus-2019/donors-and-partners/funding
  • 509.https://cepi.net/about/whyweexist/
  • 510.https://www.unicef.org/research-and-reports
  • 511.https://www.gavi.org/covax-facility#governance
  • 512.Solís Arce J.S., et al. COVID-19 vaccine acceptance and hesitancy in low- and middle-income countries. Nat. Med. 2021;27:1385–1394. doi: 10.1038/s41591-021-01454-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 513.de Figueiredo A., Simas C., Karafillakis E., Paterson P., Larson H.J. Mapping global trends in vaccine confidence and investigating barriers to vaccine uptake: a large-scale retrospective temporal modelling study. Lancet. 2020;396(10255):898–908. doi: 10.1016/S0140-6736(20)31558-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 514.McGill COVID19 Vaccine Tracker Team. https://covid19.trackvaccines.org/vaccines/

Articles from International Journal of Biological Macromolecules are provided here courtesy of Elsevier

RESOURCES