Next Article in Journal
Photosynthetic Responses, Growth, Production, and Tolerance of Traditional Varieties of Cowpea under Salt Stress
Next Article in Special Issue
Chemical Composition of Various Nepeta cataria Plant Organs’ Methanol Extracts Associated with In Vivo Hepatoprotective and Antigenotoxic Features as well as Molecular Modeling Investigations
Previous Article in Journal
Isolation and Comprehensive in Silico Characterisation of a New 3-Hydroxy-3-Methylglutaryl-Coenzyme A Reductase 4 (HMGR4) Gene Promoter from Salvia miltiorrhiza: Comparative Analyses of Plant HMGR Promoters
Previous Article in Special Issue
Novel Neuroprotective Potential of Bunchosia armeniaca (Cav.) DC against Lipopolysaccharide Induced Alzheimer’s Disease in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Main Protease of SARS-CoV-2 as a Target for Phytochemicals against Coronavirus

Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
*
Author to whom correspondence should be addressed.
Plants 2022, 11(14), 1862; https://doi.org/10.3390/plants11141862
Submission received: 21 June 2022 / Revised: 12 July 2022 / Accepted: 15 July 2022 / Published: 17 July 2022
(This article belongs to the Special Issue Pharmacological and Toxicological Study of Medicinal Plants)

Abstract

:
In late December 2019, the first cases of COVID-19 emerged as an outbreak in Wuhan, China that later spread vastly around the world, evolving into a pandemic and one of the worst global health crises in modern history. The causative agent was identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although several vaccines were authorized for emergency use, constantly emerging new viral mutants and limited treatment options for COVID-19 drastically highlighted the need for developing an efficient treatment for this disease. One of the most important viral components to target for this purpose is the main protease of the coronavirus (Mpro). This enzyme is an excellent target for a potential drug, as it is essential for viral replication and has no closely related homologues in humans, making its inhibitors unlikely to be toxic. Our review describes a variety of approaches that could be applied in search of potential inhibitors among plant-derived compounds, including virtual in silico screening (a data-driven approach), which could be structure-based or fragment-guided, the classical approach of high-throughput screening, and antiviral activity cell-based assays. We will focus on several classes of compounds reported to be potential inhibitors of Mpro, including phenols and polyphenols, alkaloids, and terpenoids.

1. Introduction

In late December 2019, a viral pneumonia outbreak emerged in Wuhan, China caused by a new strain of coronavirus that was identified as SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) [1,2,3,4]. Soon after, the outbreak was declared a public health emergency of international concern by the WHO, and later in March it was declared a global pandemic, named COVID-19 (coronavirus disease 2019) [5,6]. The virus spread vastly all around the world, causing, to date, more than 500 million confirmed cases and millions of deaths in one of the worst global health crises in modern history [7]. Although many vaccines have been approved worldwide, so far there is still no treatment for COVID-19, and only supportive and preventive measures are being applied to reduce the disease’s complications [8,9,10]. Moreover, in trying to adapt to changing environments, the virus has developed a number of mutations that could strongly affect its transmissibility and infectivity. These mutations are also prone to increasing and spreading worldwide, due to natural selection [11,12,13,14]. Therefore, considering the constantly emerging viral mutants and the absence of approved, fully effective medications, there is an urgent need for developing an efficient treatment for COVID-19.

2. SARS-CoV-2 Structure and the Main Protease of SARS-CoV-2 as a Potential Protein Target

Similar to other viruses in the Coronaviridae family, SARS-CoV-2 has a single-stranded, positive-sense RNA (+ssRNA) genome of approximately 29 kb [15,16]. The viral RNA is composed of more than six open reading frames (ORFs), the first one of which (ORF1) serves as a template for producing two polyproteins essential for viral replication and transcription: pp1a and pp1ab [17,18]. These two polyproteins undergo extensive processing by the viral main protease (Mpro) and another protease known as papain-like protease (PLP), producing 16 nonstructural proteins (NSPs) [3,19]. The other ORFs encode at least four main structural proteins: the spike (S), membrane (M), envelope (E), and nucleocapsid (N) proteins [17,20,21].
Mpro, also known as 3-chymotrypsin-like protease (3CLpro), is a 33.8 kDa, three-domain cysteine protease, essential for proteolytic maturation and viral replication [9,18,22,23]. Mpro was found to be conserved among coronaviruses (CoVs), along with some common features of its substrates in different CoVs [18,24]. In addition to its vital role in the SARS-CoV-2 life cycle, the absence of any closely related human homologous Mpro makes it an ideal protein target for potential antiviral drugs, as its inhibitors are unlikely to be toxic in humans [25]. Furthermore, vaccines, as we have learned from previous viruses, can represent a selection pressure resulting in the evolution of novel resistant viral mutants, which again highlights SARS-CoV-2 Mpro as a good drug target, as it is less subject to such selection pressure caused by vaccines targeting the viral spike protein [26,27].

3. Standard Approaches That Could Be Applied in the Search for Mpro Potential Inhibitors

3.1. Virtual In Silico Screening: (The Data-Driven Approach)

Enabled by the development of bioinformatics tools along with eased access to protein databases, virtual screening has proven to be a fundamental tool in drug design and drug repurposing research [28,29]. In the virtual screening approach, automated molecular docking tools are usually used to predict the best possible variant for binding one molecule to another, considering the best orientation with the best binding affinity [30]. These tools enable the screening of large numbers of candidates against a specific studied target, at a very low cost [31]. Virtual screening is a data-driven approach that can be either target-based, where a library of candidate ligands is docked against the target and analyzed, or ligand-based, where a similarity search or a machine learning strategy can be applied [32,33,34,35].
Since the beginning of the COVID-19 pandemic, a large number of studies around the world have used this approach to search for potential inhibitors of SARS-CoV-2 [22,36,37,38]. Joshi R.S. et al. used this approach in their study conducted in 2020 to scan over 7000 compounds from different origins against SARS-CoV-2 Mpro [39]. Another study conducted by Tallei E.T. et al. in 2020 used this approach to evaluate the potency of plant-derived bioactive compounds against Mpro, resulting in the identification of pectolinarin, hesperidin, nabiximols, rhoifolin, and epigallocatechin gallate as potential antiviral phytochemicals [40]. Research by Tahir Ul Qamar et al. also resulted in the identification of 5,7,3′,4′-Tetrahydroxy-2′-(3,3-dimethylallyl) isoflavone, amaranthin, licoleafol, calceolarioside B, and methyl rosmarinate as potential inhibitors of the target, using this approach [41]. Khaerunnisa S. et al. extended the list with kaempferol, quercetin, luteolin-7-glucoside, demetoxycurcumine, naringenin, apigenin-7-glucoside, oleuropein, catechin, curcumin, zingerol, gingerol, and allicin [42]. Essential oils have also shown their effectiveness against SARS-CoV-2 Mpro in silico [43,44,45]. Therefore, using this approach, multiple natural compounds have been identified as strong binders of SARS-CoV-2 Mpro, and some of them were also identified as multi-target inhibitors that could be applied in COVID-19 management approaches [36,37,38,39,40,41,42,43,44,45].

3.2. The Classical Approach of High-Throughput Screening (HTS)

High-throughput screening (HTS) is a method for automated testing of thousands to millions of compounds for their biological activity against specific targets on model systems [46]. The development of robotics, laboratory equipment, laboratory methods, and software for the control of sample preparation, incubation, results detection, and data processing has allowed the HTS approach to be used to quickly search for lead compounds. Therefore, it is possible to quickly and inexpensively test large libraries of chemical compounds for their biochemical activity [47].
In practice, HTS is implemented in the form of a large number of miniature in vitro assays to identify molecules that can modulate the activity of a biological target. These reactions are run in 96-well, 386-well, or 1536-well plates [48]. Most often, the results of such biochemical analyses are obtained using various fluorescence detection methods [49], for example, direct measurement of fluorescence, fluorescence polarization, fluorescence resonance energy transfer (FRET), fluorescence quenching energy transfer (QFRET), or time-resolved fluorescence [46].
Since the early stage of COVID-19 pandemic, a large number of HTS assays have been developed worldwide to screen huge libraries of either previously approved drugs or potential inhibitors against SARS-CoV-2 [50,51,52,53]. Using this approach in their study to screen a library of 10,755 potential inhibitory compounds against SARS-CoV-2 Mpro, along with drugs previously approved for other viruses, Zhu W. et al. identified 23 potential inhibitors with different half-maximal inhibitory concentration values (IC50), the efficacy of 7 of which was confirmed in a later cytopathic effect assay [54]. Given the high safety level required of laboratories studying and manipulating the live SARS-CoV-2 virus (BSL-3 laboratories), Zhang, Q.Y. et al. proposed a new HTS assay to enable potential antiviral testing in a BSL-2 research facility, where they constructed a reporter replicon of the virus using Renilla luciferase (Rluc) reporter gene and validated it later using hit natural compounds [52]. Froggatt H. M. et al. also developed a fluorescence-based HTS assay using a protein derived from green fluorescent protein (GFP) to serve as a target for SARS-CoV-2 Mpro, and hence a reporter for the enzyme’s inhibition and activation, enabling rapid screening of libraries and identification of lead compounds [55]. A further improvement of HTS can be achieved by combining it with the previous in silico approach to yield an ultra-high-throughput virtual screening approach, where huge libraries can be tested against multiple viral targets efficiently and rapidly [56]. Gorgulla C. et al. conducted a study to search for SARS-CoV-2 inhibitors using this large-scale HTS screening approach and were able to screen over one billion candidate molecules against 40 different target sites on 17 potential targets, both in the virus and the host [56]. Although the results were obtained from computational data and have not all been tested with experimental analyses yet, this filtration of candidates could narrow down research targets for later more detailed and efficient analyses.

3.3. Antiviral Activity Cell-Based Assays

Cell-based assays offer an advantage over virtual or biochemical screening assays, as they provide a whole physiological environment, reflecting the complexity of a living system rather than focusing on a specific isolated target and thereby enabling a more accurate evaluation of the biological activity and potential toxicity of screened compounds [57,58,59]. Due to practical considerations, it is important to develop and test drug compounds that exhibit inhibitory activity at various stages of the virus life cycle. Therefore, test systems have been developed to evaluate the effectiveness of inhibitors of entry, uncoating, replication, assembly (in which viral proteases are active), and maturation of viruses [60]. However, the whole variety of such systems can be reduced to two main mechanisms for their implementation: cytopathic and reporter mechanisms [61]. In the first case, the activity of antiviral agents is assessed by reducing the formation of plaques due to the accumulation of coloring or luminescent agents in living cells [62,63]. In the second, viruses and cells with report inserts are used, and the activity of inhibitors helps in reducing the expression of the reporter protein [64].
Since work with a live virus is accompanied by significant organizational restrictions, approaches have been developed for evaluating the effectiveness of antiviral agents that model various stages of the life cycle of viruses in cells of HeLa [65], Escherichia coli [66], and Saccharomyces cerevisiae [61]. Moreover, cell-based assays are nowadays increasingly integrated into HTS assays to accomplish rapid screens in a relevant physiological environment [58].
Several recent studies have used antiviral activity cell-based assays, either after or combined with the previously described approaches, to investigate previously approved drugs and herbal medicines for their potential inhibition potency against SARS-CoV-2 [9,67,68,69,70]. Applying this methodology followed by a further in vivo validation, Jan J.T. et al. screened a 3000-candidate library of both pharmaceuticals and herbal medicines to test their effectiveness against SARS-CoV-2 Mpro and RNA polymerase and proposed multiple herbal extracts as potential herbal inhibitors against the targeted viral enzymes [68]. Another recent study conducted by Qiao J. et al. also applied this approach to investigate 32 different inhibitors against SARS-CoV-2 Mpro, 6 of which were found to have a high inhibition potency and were used to select candidates for further in vivo investigation [71].
The phased use of these three approaches makes it possible to identify those that exhibit the targeted therapeutic activity from the variety of known plant secondary metabolites. Among these compounds, there may be those that have not previously exhibited such properties. Furthermore, compounds for which hypothetical activity is found can be quickly tested for their effectiveness on cell-free and then on cellular systems. Such a screening strategy has shown to be effective in the search for inhibitors of SARS-CoV-2 Mpro, which indicates its potential in the search for drugs against new pathogens [9].
Therefore, at each stage of applying these methods, it is possible to significantly narrow the range of compounds under study, which facilitates a significant simplification and accelerates the search for molecules with clinical potential, therefore enabling moving on to the next stage of preclinical trials as soon as possible.

4. Phytochemicals as a Reservoir to Search for SARS-CoV-2 Mpro Potential Inhibitors

As mother nature has always provided an infinite library of natural products and chemicals, the use of herbal medicines and their derivatives to combat diseases dates back to more than 60,000 years ago in ancient history [72,73,74]. A study by Fabricant D. S. and Farnsworth N. R. in 2001 estimated that there were more than 250,000 species of higher plants on our planet, of which only 6% had been tested and evaluated for their biologic activity at the time [72]. Today, advanced screening techniques and assays have led to phytochemicals composing a significant part of the pharmaceutical market [75,76,77,78,79,80,81,82]. Plants can be used as sources of medicinal active compounds using several methodologies. In some cases, the whole plant or parts of the plant could be used as herbal remedy, e.g., garlic or curcumin [72,78,83]. Another methodology uses the plant as a direct source of bioactive compounds such as digoxin or morphine [72,84]. Sometimes, plants can provide compounds that could be used later as a starting point for producing highly effective, less toxic, easy-to-obtain, semisynthetic or synthetic compounds, as in the case of narcotic analgesics [72,85]. With such knowledge, thousands of studies all over the world have been conducted on searching for anti-SARS-CoV-2 treatments from plant origins, with Mpro being one of the most targeted viral components in this research, and the in silico data-driven approach being the most frequently applied [23,73,86,87].
Several classes of bioactive phytochemicals have been shown to be potential inhibitors of SARS-CoV-2 Mpro, including phenols [88,89], polyphenols [90,91], terpenoids [92], etc. (Figure 1). The main classes of SARS-CoV-2 Mpro inhibitors and their specific representatives are summarized in Table 1.
Most often, SARS-CoV-2 Mpro inhibitors are found among flavonoids and terpenoids. A common feature of compounds of these classes that exhibit SARS-CoV-2 Mpro inhibitory activity is an alpha-beta-unsaturated ketone group conjugated to an aromatic ring. It has also been shown that the presence of bicyclic aromatic rings in the structure and the presence of hydroxyl groups on all rings, especially on the B-ring of flavonoids, increase their respective activities [37,123].
Flavonoids, which are widespread in plants, are represented by compounds of various structures, often present together, which can lead to synergistic effects, including antiviral properties, for example, in tea, garlic, fruits, vegetables, etc. [124,125].
A study conducted by Nguyen T. et al. investigated the potential inhibitory effects of plant-derived polyphenols on SARS-CoV-2 Mpro, mainly those derived from black garlic extract prepared by heating raw garlic (Allium sativum) to high temperatures [91]. The studied extract contained many polyphenols, including both phenolic acids and flavonoids [126], of which several were found to have inhibitory effects against SARS-CoV-2 Mpro and were selected for further determination of their IC50 values (Table 1) [91].
Salvadora persica contains 10 flavonoid metabolites that were found to have substantially stable binding affinities for the SARS-CoV2 Mpro, including glycosides of kaempferol and its O-methylated derivatives [104]. Another study, conducted by Jo S. et al., used a fluorescence resonance energy transfer assay (FRET) to screen a flavonoids library against SARS-CoV2 Mpro, resulting in the identification of three flavonoids as potential inhibitors: herbacetin, rhoifolin, and pectolinarin [107]. Plants from Indian traditional medicine have also been found to contain potential inhibitors of SARS-CoV-2 Mpro [23,87]. One example is tulsi (Ocimum sanctum), with its derived polyphenols vicenin and isorientin 4′-O-glucoside 2″-O-p-hydroxybenzoate [87]. Another example of flavonoids derived from Indian medicinal plants showing such inhibitory effects was also presented in a recent study conducted by Saravanan K. et al. in 2020 [23]. In this study, 41 compounds from different plants were docked against SARS-CoV-2 Mpro. Several flavonoids of the candidate compounds showed relatively high binding affinity values, with the highest value being that of amentoflavone, a flavonoid derived from Torreya nucifera that has previously been shown to have in vitro antiviral properties [23,127].
Catechins are another group of phytochemicals representing a subclass of polyphenolic compounds found in a variety of plants and plant-derived dietary supplements such as green tea, cocoa, vinegar, wine, and garlic [91,128]. Due to the 3-galloyl and 5′-OH groups in their structure [129], catechins from green tea, mainly the previously mentioned epigallocatechin-3-O-gallate (EGCG), were found to exhibit antiviral properties against SARS-CoV-2 and specifically against its Mpro [88]. The same compounds were found in black garlic extract [91] and the petals of Himalayan Rhododendron arboreum [130], with similar potential anti-SARS-CoV-2-Mpro properties, using both in silico and in vitro analyses. In green tea (Camellia sinensis), three polyphenols were found to have good binding affinities for SARS-CoV-2 Mpro: EGCG, epicatechingallate (ECG), and gallocatechin-3-gallate (GCG). They were proven to be highly stable, similarly compacted, and subject to low conformational variability [110]. EGCG, found in different plant sources including Camellia sinensis, Vitis vinifera, and black garlic extract, was further investigated in several in vitro and in vivo studies to test its effectiveness against COVID-19 [131,132,133,134]. EGCG and its oxidized form were found to inhibit Mpro in vitro [133,134], to directly inhibit an early infection [132], and to reduce viral replication in mouse lung cells [132].
Lactones derived from plants have also shown potential inhibitory anti-SARS-CoV-2-Mpro properties [89]. An example of such lactones is withaferin A, a steroidal lactone derived from the well-known Indian medicinal plant Ashwagandha (Withania somnifera), which has shown inhibitory potencies against the targeted protease in a molecular docking study conducted by Sudeep H.V. et al. in 2020, with a binding score of −9.83 Kcal/mol [89].
In addition, plant tannins have been reported to have antibacterial, antifungal, and antiviral properties [135], and accordingly have been screened in several studies to investigate their potential inhibitory effects against SARS-CoV-2 Mpro [121]. A study by Wang S.C. et al. tested the effectiveness of tannic acid, a tannin found abundantly in red wine and in berries, grapes, pomegranate, and other fruits [136], against SARS-CoV-2 Mpro both in silico and in vitro, using pseudotyped viral particles. The obtained data suggested that tannic acid was a potential inhibitor of the targeted enzyme, as it was found to form a thermodynamically stable complex with Mpro. Cinnamtannin-B is another naturally occurring tannin that has been reported as a top hit against SARS-CoV-2 Mpro [137]. Cinnamtannin-B is derived from the cinnamon plant (Cinnamomum zeylanicum) and can only be isolated from a limited number of plants such as Linderae umbellatae and bay laurel (Laurus nobilis) [138].
Alkaloids represent another group of natural phytochemicals that have a broad spectrum of biological activities, mainly antiviral [139]. One example is capsaicin, a plant-derived alkaloid that is derived mainly from the fruit of the Capsicum genus [140] and has been found by in silico research to be a potential inhibitor of SARS-CoV-2 Mpro, similar to another plant-derived alkaloid named psychotrine [112]. Achyranthine is another alkaloid derived mainly from Achyranthes aspera [141] that was found to bind three sites of Mpro, with binding scores ranging between −4.1 and −4.7 [113].
Terpenes and their modified class of terpenoids represent a huge group of phytochemicals that have also been found to have antiviral properties in general and inhibitory potential effectiveness against SARS-CoV-2 Mpro in particular [92]. Plant terpenoids with medicinal potential are estimated to include more than 100,000 compounds on our planet, with more than 12,000 belonging to the diterpenoid group alone [142]. Of the ayurvedic medicinal plants, two terpenoids were suggested by in silico research as potential inhibitors of SARS-CoV-2 Mpro: ursolic acid from tulsi (Ocimum sanctum) and withanoside V from ashwagandha (Withania somnifera) [87]. Other examples of terpenes from Indian medicinal plants with inhibitory properties against SARS-CoV-2 are listed in Table 1. Some of these terpenes, e.g., curcumin, have already been proven to show antiviral activity in humans, protecting against acute and chronic lung diseases including pneumonia [143,144], and accordingly have been further suggested for application in clinical use as a prophylactic measure against COVID-19 [145]. From Citrus limon, two cyclic monoterpenes have been proven to interact with SARS-CoV-2 Mpro, including limonene and sabinene [113]. Glycyrrhizic acid (glycyrrhizin), another plant-derived triterpenoid saponin found mainly in Glycyrrhiza glabra, was also proven to have in vitro anti-SARS-CoV-2-Mpro potential [116]. Another main source of terpenes, mainly monoterpenes, is plant essential oils [146], which have been proven in several studies to have a wide variety of antimicrobial properties [147,148,149]. One example of such a plant-essential-oil-derived monoterpene with antiviral properties is pinene, a bioactive compound of black pepper (Piper nigrum), that was proven to bind Mpro in silico [113,150]. Similarly, the diterpenoid andrographolide from Andrographis paniculate was successfully docked against SARS-CoV-2 Mpro in two different molecular docking studies and was therefore suggested as a potential inhibitor to be evaluated in further in vitro analyses [89,151]. From medicinal Arabic plants, betulinic acid, a pentacyclic triterpenoid derived from the Christ’s thorn plant (Ziziphus spina-christi), was found by in silico research to successfully bind SARS-CoV-2 Mpro [152]. Roots of the plant Maprounea africana are also considered to be a source of several bioactive compounds, including triterpenes [153] such as 1β-hydroxyaleuritolic acid 3-p-hydroxybenzoate that have shown in silico inhibitory potential against Mpro [119].

5. Perspectives of Large-Scale Synthesis of Anti-SARS-CoV-2 Compounds

The large-scale production of newly discovered compounds with anti-SARS-CoV-2 activity can proceed in several ways. Some compounds can be obtained relatively easily by means of chemical synthesis. This applies to many of the flavonoids. Others are preferably obtained using cell cultures. A number of anti-SARS-CoV-2 compounds have attracted interest in the past due to their wide range of biological activities. For this reason, methods for their production in cell cultures have been developed and constantly optimized. For example, compounds such as tanshinones and rosmarinic acid can be effectively produced in hairy root cultures of Salvia miltiorrhiza Bunge [154] and catechins are produced in hairy roots of Camellia sinensis (L.) O. Kuntze [155].
In addition, there are several approaches for increasing the yield of specialized metabolites, including metabolic engineering of tanshinones [154,155,156], phenolic acids [155,157], flavonoids [158], and diterpenoids [159] or varying the cultivation conditions [155,157].
Finally, natural compounds can be precursors for subsequent chemical modification. For example, chemical synthesis based on chalcones allowed the development of more effective anti-SARS-CoV-2 compounds [160].

6. Conclusions

Plants have been used for a long time as a resource for bioactive compounds and phytochemicals to be applied in therapeutic approaches for different diseases. Since the COVID-19 pandemic is still ongoing, phytochemicals could be used to find effective and safe treatments for the disease. To date, using computer modeling of cell-free and cell-based screening approaches, some progress has been made in the search for potential drugs aimed at inhibiting the main protease of the coronavirus. They are represented by phytochemicals from several classes, including polyphenols, terpenoids, catechins, lactones, and tannins. Some plants containing promising compounds can be used as food directly, e.g., garlic, and others can serve as sources of pure substances for pharmacology. Future studies should shift their focus towards assessing possible toxic effects on cells, since even the most promising protease inhibitors will not be able to find application if they are found to have a toxic effect.

Author Contributions

T.V.M. suggested the structure of the paper; S.S.I. wrote the first draft of the paper; S.S.I., R.R.Z., S.V.S. and T.V.M. participated in writing and correcting different sections of the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Science and Higher Education of the Russian Federation, in accordance with agreement no. 075-15-2022-322, dated 22 April 2022 on providing a grant in the form of subsidies from the Federal budget of the Russian Federation. The grant was provided for state support for the creation and development of a world-class scientific center “Agrotechnologies for the Future”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Lee, P.I.; Hsueh, P.R. Emerging threats from zoonotic coronaviruses-from SARS and MERS to 2019-nCoV. J. Microbiol. Immunol. Infect. 2020, 53, 365–367. [Google Scholar] [CrossRef] [PubMed]
  2. Li, Q.; Guan, X.; Wu, P.; Wang, X.; Zhou, L.; Tong, Y.; Feng, Z. Early Transmission Dynamics in Wuhan, China, of Novel Coronavirus-Infected Pneumonia. N. Engl. J. Med. 2020, 382, 1199–1207. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.; Wang, W.; Song, Z.; Hu, Y.; Tao, Z.; Tian, J.; Pei, Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [Green Version]
  4. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
  5. Rodríguez-Morales, A.J.; MacGregor, K.; Kanagarajah, S.; Patel, D.; Schlagenhauf, P. Going global—Travel and the 2019 novel coronavirus. Travel Med. Infect. Dis. 2020, 33, 101578. [Google Scholar] [CrossRef] [PubMed]
  6. Silveira, M.M.; Moreira, G.M.S.G.; Mendonça, M. DNA vaccines against COVID-19: Perspectives and challenges. Life Sci. 2020, 267, 118919. [Google Scholar] [CrossRef] [PubMed]
  7. WHO. COVID-19 Dashboard. 2022. Available online: https://covid19.who.int/ (accessed on 5 May 2022).
  8. Chen, P.; Shi, X.; He, W.; Zhong, G.; Tang, Y.; Wang, H.; Zhang, P. mRNA vaccine-a desirable therapeutic strategy for surmounting COVID-19 pandemic. Hum. Vaccines Immunother. 2022, 18, 2040330. [Google Scholar] [CrossRef]
  9. Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Yang, H. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289–293. [Google Scholar] [CrossRef] [Green Version]
  10. Yamamoto, K.Z.; Yasuo, N.; Sekijima, M. Screening for Inhibitors of Main Protease in SARS-CoV-2: In Silico and In Vitro Approach Avoiding Peptidyl Secondary Amides. J. Chem. Inf. Model. 2022, 62, 350–358. [Google Scholar] [CrossRef]
  11. Hossain, M.J.; Rabaan, A.A.; Mutair, A.A.; Alhumaid, S.; Emran, T.B.; Saikumar, G.; Dhama, K. Strategies to tackle SARS-CoV-2 Mu, a newly classified variant of interest likely to resist currently available COVID-19 vaccines. Hum. Vaccin. Immunother. 2022, 18, 2027197. [Google Scholar] [CrossRef]
  12. Konings, F.; Perkins, M.D.; Kuhn, J.H.; Pallen, M.J.; Alm, E.J.; Archer, B.N.; Van Kerkhove, M.D. SARS-CoV-2 Variants of Interest and Concern naming scheme conducive for global discourse. Nat. Microbiol. 2021, 6, 821–823. [Google Scholar] [CrossRef] [PubMed]
  13. Sharun, K.; Tiwari, R.; Dhama, K.; Emran, T.B.; Rabaan, A.A.; Al Mutair, A. Emerging SARS-CoV-2 variants: Impact on vaccine efficacy and neutralizing antibodies. Hum. Vaccin. Immunother. 2021, 17, 3491–3494. [Google Scholar] [CrossRef] [PubMed]
  14. Van Egeren, D.; Novokhodko, A.; Stoddard, M.; Tran, U.; Zetter, B.; Rogers, M.; Chakravarty, A. Risk of rapid evolutionary escape from biomedical interventions targeting SARS-CoV-2 spike protein. PLoS ONE 2021, 16, e0250780. [Google Scholar] [CrossRef] [PubMed]
  15. Brant, A.C.; Tian, W.; Majerciak, V.; Yang, W.; Zheng, Z.M. SARS-CoV-2: From its discovery to genome structure, transcription, and replication. Cell Biosci. 2021, 11, 136. [Google Scholar] [CrossRef]
  16. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Chen, Y.; Liu, Q.; Guo, D. Emerging coronaviruses: Genome structure, replication, and pathogenesis. J. Med. Virol. 2020, 92, 418–423. [Google Scholar] [CrossRef] [PubMed]
  18. Dai, W.; Zhang, B.; Jiang, X.M.; Su, H.; Li, J.; Zhao, Y.; Liu, H. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science 2020, 368, 1331–1335. [Google Scholar] [CrossRef] [Green Version]
  19. Ramajayam, R.; Tan, K.-P.; Liang, P.-H. Recent development of 3C and 3CL protease inhibitors for anti-coronavirus and anti-picornavirus drug discovery. Biochem. Soc. Trans. 2011, 39, 1371–1375. [Google Scholar] [CrossRef]
  20. Cao, Y.; Yang, R.; Lee, I.; Zhang, W.; Sun, J.; Wang, W.; Meng, X. Characterization of the SARS-CoV-2 E Protein: Sequence, Structure, Viroporin, and Inhibitors. Protein Sci. 2021, 30, 1114–1130. [Google Scholar] [CrossRef]
  21. 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, 914–921.e10. [Google Scholar] [CrossRef]
  22. Reiner, Ž.; Hatamipour, M.; Banach, M.; Pirro, M.; Al-Rasadi, K.; Jamialahmadi, T.; Radenkovic, D.; Montecucco, F.; Sahebkar, A. Statins and the COVID-19 main protease: In silico evidence on direct interaction. Arch. Med. Sci. 2020, 16, 490–496. [Google Scholar] [CrossRef] [PubMed]
  23. Saravanan, K.M.; Zhang, H.; Senthil, R.; Vijayakumar, K.K.; Sounderrajan, V.; Wei, Y.; Shakila, H. Structural basis for the inhibition of SARS-CoV2 main protease by Indian medicinal plant-derived antiviral compounds. J. Biomol. Struct. Dyn. 2022, 40, 1970–1978. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, H.; Xie, W.; Xue, X.; Yang, K.; Ma, J.; Liang, W.; Rao, Z. Design of wide-spectrum inhibitors targeting coronavirus main proteases. PLoS Biol. 2005, 3, e324. [Google Scholar]
  25. Rocha, R.E.; Chaves, E.J.; Fischer, P.H.; Costa, L.S.; Grillo, I.B.; da Cruz, L.E.; de Lima, L.H. A higher flexibility at the SARS-CoV-2 main protease active site compared to SARS-CoV and its potentialities for new inhibitor virtual screening targeting multi-conformers. J. Biomol. Struct. Dyn. 2021, 10, 1–21. [Google Scholar] [CrossRef] [PubMed]
  26. Luo, R.; Delaunay-Moisan, A.; Timmis, K.; Danchin, A. SARS-CoV-2 biology and variants: Anticipation of viral evolution and what needs to be done. Environ. Microbiol. 2021, 23, 2339–2363. [Google Scholar] [CrossRef]
  27. Mótyán, J.A.; Mahdi, M.; Hoffka, G.; Tőzsér, J. Potential Resistance of SARS-CoV-2 Main Protease (Mpro) against Protease Inhibitors: Lessons Learned from HIV-1 Protease. Int. J. Mol. Sci. 2022, 23, 3507. [Google Scholar] [CrossRef] [PubMed]
  28. Mohan, A.; Rendine, N.; Mohammed MK, S.; Jeeva, A.; Ji, H.F.; Talluri, V.R. Structure-based virtual screening, in silico docking, ADME properties prediction and molecular dynamics studies for the identification of potential inhibitors against SARS-CoV-2 M(pro). Mol. Divers. 2021, 26, 1645–1661. [Google Scholar] [CrossRef]
  29. Morris, G.M.; Lim-Wilby, M. Molecular docking. Methods Mol. Biol. 2008, 443, 365–382. [Google Scholar]
  30. Sari, S.; Avci, A.; Aslan, E.K. In silico Repurposing of Drugs for pan-HDAC and pan-SIRT Inhibitors: Consensus Structure-based Virtual Screening and Pharmacophore Modeling Investigations. Turk. J. Pharm. Sci. 2021, 18, 730–737. [Google Scholar] [CrossRef]
  31. Pinzi, L.; Rastelli, G. Molecular Docking: Shifting Paradigms in Drug Discovery. Int. J. Mol. Sci. 2019, 20, 4331. [Google Scholar] [CrossRef] [Green Version]
  32. Bonanno, E.; Ebejer, J.-P. Applying Machine Learning to Ultrafast Shape Recognition in Ligand-Based Virtual Screening. Front. Pharmacol. 2020, 10, 1675. [Google Scholar] [CrossRef] [PubMed]
  33. Muegge, I.; Mukherjee, P. An overview of molecular fingerprint similarity search in virtual screening. Expert Opin. Drug Discov. 2015, 11, 137–148. [Google Scholar] [CrossRef] [PubMed]
  34. Pinto, G.P.; Hendrikse, N.M.; Stourac, J.; Damborsky, J.; Bednar, D. Virtual screening of potential anticancer drugs based on microbial products. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2021. [Google Scholar]
  35. Pinto, G.P.; Vavra, O.; Filipovic, J.; Stourac, J.; Bednar, D.; Damborsky, J. Fast Screening of Inhibitor Binding/Unbinding Using Novel Software Tool CaverDock. Front. Chem. 2019, 7, 709. [Google Scholar] [CrossRef] [PubMed]
  36. Islam, M.T.; Sarkar, C.; El-Kersh, D.M.; Jamaddar, S.; Uddin, S.J.; Shilpi, J.A.; Mubarak, M.S. Natural products and their derivatives against coronavirus: A review of the non-clinical and pre-clinical data. Phytother. Res. 2020, 34, 2471–2492. [Google Scholar] [CrossRef] [PubMed]
  37. Olubiyi, O.O.; Olagunju, M.; Keutmann, M.; Loschwitz, J.; Strodel, B. High Throughput Virtual Screening to Discover Inhibitors of the Main Protease of the Coronavirus SARS-CoV-2. Molecules 2020, 25, 3193. [Google Scholar] [CrossRef]
  38. Zhang, D.-H.; Wu, K.-L.; Zhang, X.; Deng, S.-Q.; Peng, B. In silico screening of Chinese herbal medicines with the potential to directly inhibit 2019 novel coronavirus. J. Integr. Med. 2020, 18, 152–158. [Google Scholar] [CrossRef]
  39. Joshi, R.S.; Jagdale, S.S.; Bansode, S.B.; Shankar, S.S.; Tellis, M.B.; Pandya, V.K.; Kulkarni, M.J. Discovery of potential multi-target-directed ligands by targeting host-specific SARS-CoV-2 structurally conserved main protease. J. Biomol. Struct. Dyn. 2021, 39, 3099–3114. [Google Scholar] [CrossRef] [Green Version]
  40. Tallei, T.E.; Tumilaar, S.G.; Niode, N.J.; Kepel, B.J.; Idroes, R.; Effendi, Y.; Emran, T.B. Potential of Plant Bioactive Compounds as SARS-CoV-2 Main Protease (Mpro) and Spike (S) Glycoprotein Inhibitors: A Molecular Docking Study. Scientifica 2020, 2020, 6307457. [Google Scholar] [CrossRef]
  41. Ul Qamar, M.T.; Alqahtani, S.M.; Alamri, M.A.; Chen, L.L. Structural basis of SARS-CoV-2 3CL(pro) and anti-COVID-19 drug discovery from medicinal plants. J. Pharm. Anal. 2020, 10, 313–319. [Google Scholar] [CrossRef]
  42. Khaerunnisa, S.; Kurniawan, H.; Awaluddin, R.; Suhartati, S.; Soetjipto, S. Potential Inhibitor of COVID-19 Main Protease (Mpro) from Several Medicinal Plant Compounds by Molecular Docking Study. Preprints 2020, 2020, 2020030226. [Google Scholar]
  43. da Silva JK, R.; Figueiredo PL, B.; Byler, K.G.; Setzer, W.N. Essential Oils as Antiviral Agents, Potential of Essential Oils to Treat SARS-CoV-2 Infection: An In-Silico Investigation. Int. J. Mol. Sci. 2020, 21, 3426. [Google Scholar] [CrossRef] [PubMed]
  44. Mohamed, M.E.; Tawfeek, N.; Elbaramawi, S.S.; Fikry, E. Agathis robusta Bark Essential Oil Effectiveness against COVID-19: Chemical Composition, In Silico and In Vitro Approaches. Plants 2022, 11, 663. [Google Scholar] [CrossRef] [PubMed]
  45. My, T.T.A.; Loan, H.T.P.; Hai, N.T.T.; Hieu, L.T.; Hoa, T.T.; Thuy, B.T.P.; Quang, D.T.; Triet, N.T.; Van Anh, T.T.; Dieu, N.T.X.; et al. Evaluation of the Inhibitory Activities of COVID-19 of Melaleuca cajuputi Oil Using Docking Simulation. ChemistrySelect 2020, 5, 6312–6320. [Google Scholar] [CrossRef] [PubMed]
  46. Attene-Ramos, M.S.; Austin, C.P.; Xia, M. High Throughput Screening. In Encyclopedia of Toxicology, 3rd ed.; Wexler, P., Ed.; Academic Press: Oxford, UK, 2014; pp. 916–917. [Google Scholar]
  47. Ismail, F.M.; Nahar, L.; Sarker, S.D. High-Throughput Screening of Phytochemicals: Application of Computational Methods. Comput. Phytochem. 2018, 165–192. [Google Scholar]
  48. Lundblad, R.L. Drug Design. In Encyclopedia of Cell Biology; Elsevier: Amsterdam, The Netherlands, 2016. [Google Scholar]
  49. Hajare, A.A.; Salunkhe, S.S.; Mali, S.S.; Gorde, S.S.; Nadaf, S.J.; Pishawikar, S.A. Review on: High-throughput screening is an approach to drug discovery. Am. J. Pharm. Tech. Res. 2013, 4, 112–129. [Google Scholar]
  50. Liu, J.; Li, K.; Cheng, L.; Shao, J.; Yang, S.; Zhang, W.; Zhou, G.; de Vries, A.A.; Yu, Z. A high-throughput drug screening strategy against coronaviruses. Int. J. Infect. Dis. 2020, 103, 300–304. [Google Scholar] [CrossRef]
  51. Xu, T.; Zheng, W.; Huang, R. High-throughput screening assays for SARS-CoV-2 drug development: Current status and future directions. Drug Discov. Today 2021, 26, 2439–2444. [Google Scholar] [CrossRef]
  52. Zhang, Q.Y.; Deng, C.L.; Liu, J.; Li, J.Q.; Zhang, H.Q.; Li, N.; Ye, H.Q. SARS-CoV-2 replicon for high-throughput antiviral screening. J. Gen. Virol. 2021, 102, 001583. [Google Scholar] [CrossRef]
  53. Zhao, Y.; Du, X.; Duan, Y.; Pan, X.; Sun, Y.; You, T.; Yang, H. High-throughput screening identifies established drugs as SARS-CoV-2 PLpro inhibitors. Protein Cell 2021, 12, 877–888. [Google Scholar] [CrossRef]
  54. Zhu, W.; Xu, M.; Chen, C.Z.; Guo, H.; Shen, M.; Hu, X.; Zheng, W. Identification of SARS-CoV-2 3CL Protease Inhibitors by a Quantitative High-Throughput Screening. ACS Pharmacol. Transl. Sci. 2020, 3, 1008–1016. [Google Scholar] [CrossRef]
  55. Froggatt, H.M.; Heaton, B.E.; Heaton, N.S. Development of a Fluorescence-Based, High-Throughput SARS-CoV-2 3CL(pro) Reporter Assay. J. Virol. 2020, 94, e01265-20. [Google Scholar] [CrossRef] [PubMed]
  56. Gorgulla, C.; Das KM, P.; Leigh, K.E.; Cespugli, M.; Fischer, P.D.; Wang, Z.F.; Arthanari, H. A multi-pronged approach targeting SARS-CoV-2 proteins using ultra-large virtual screening. iScience 2021, 24, 102021. [Google Scholar] [CrossRef] [PubMed]
  57. Feng, Y.; Mitchison, T.J.; Bender, A.; Young, D.W.; Tallarico, J.A. Multi-parameter phenotypic profiling: Using cellular effects to characterize small-molecule compounds. Nat. Rev. Drug Discov. 2009, 8, 567–578. [Google Scholar] [CrossRef]
  58. Nierode, G.; Kwon, P.S.; Dordick, J.S.; Kwon, S.-J. Cell-Based Assay Design for High-Content Screening of Drug Candidates. J. Microbiol. Biotechnol. 2016, 26, 213–225. [Google Scholar] [CrossRef] [Green Version]
  59. Zaman, G.J. Cell-based screening. Comb. Chem. High Throughput Screen 2008, 11, 494. [Google Scholar] [CrossRef] [PubMed]
  60. Rumlová, M.; Ruml, T. In vitro methods for testing antiviral drugs. Biotechnol. Adv. 2017, 36, 557–576. [Google Scholar] [CrossRef]
  61. Green, N.; Ott, R.D.; Isaacs, R.J.; Fang, H. Cell-based assays to identify inhibitors of viral disease. Expert Opin. Drug Discov. 2008, 3, 671–676. [Google Scholar] [CrossRef] [Green Version]
  62. McLaren, C.; Ellis, M.; Hunter, G. A colorimetric assay for the measurement of the sensitivity of herpes simplex viruses to antiviral agents. Antivir. Res. 1983, 3, 223–234. [Google Scholar] [CrossRef]
  63. Severson, W.E.; Shindo, N.; Sosa, M.; Fletcher, I.T.; White, E.L.; Ananthan, S.; Jonsson, C.B. Development and Validation of a High-Throughput Screen for Inhibitors of SARS-CoV and Its Application in Screening of a 100,000-Compound Library. SLAS Discov. Adv. Sci. Drug Discov. 2007, 12, 33–40. [Google Scholar] [CrossRef] [Green Version]
  64. Westby, M.; Nakayama, G.; Butler, S.; Blair, W. Cell-based and biochemical screening approaches for the discovery of novel HIV-1 inhibitors. Antivir. Res. 2005, 67, 121–140. [Google Scholar] [CrossRef]
  65. Lindsten, K.; Uhlíková, T.; Konvalinka, J.; Masucci, M.; Dantuma, N.P. Cell-Based Fluorescence Assay for Human Immunodeficiency Virus Type 1 Protease Activity. Antimicrob. Agents Chemother. 2001, 45, 2616–2622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Cheng, Y.-S.E.; Lo, K.-H.; Hsu, H.-H.; Shao, Y.-M.; Yang, W.-B.; Lin, C.-H.; Wong, C.-H. Screening for HIV protease inhibitors by protection against activity-mediated cytotoxicity in Escherichia coli. J. Virol. Methods 2006, 137, 82–87. [Google Scholar] [CrossRef]
  67. Bertolin, A.P.; Weissmann, F.; Zeng, J.; Posse, V.; Milligan, J.C.; Canal, B.; Diffley, J.F. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of nsp12/7/8 RNA-dependent RNA polymerase. Biochem. J. 2021, 478, 2425–2443. [Google Scholar] [CrossRef] [PubMed]
  68. Jan, J.T.; Cheng TJ, R.; Juang, Y.P.; Ma, H.H.; Wu, Y.T.; Yang, W.B.; Wong, C.H. Identification of existing pharmaceuticals and herbal medicines as inhibitors of SARS-CoV-2 infection. Proc. Natl. Acad. Sci. USA 2021, 118, e2021579118. [Google Scholar] [CrossRef]
  69. Van Damme, E.; De Meyer, S.; Bojkova, D.; Ciesek, S.; Cinatl, J.; De Jonghe, S.; Van Loock, M. In vitro activity of itraconazole against SARS-CoV-2. J. Med. Virol. 2021, 93, 4454–4460. [Google Scholar] [CrossRef]
  70. Zhao, J.; Guo, S.; Yi, D.; Li, Q.; Ma, L.; Zhang, Y.; Cen, S. A cell-based assay to discover inhibitors of SARS-CoV-2 RNA dependent RNA polymerase. Antiviral Res. 2021, 190, 105078. [Google Scholar] [CrossRef] [PubMed]
  71. Qiao, J.; Li, Y.S.; Zeng, R.; Liu, F.L.; Luo, R.H.; Huang, C.; Yang, S. SARS-CoV-2 M(pro) inhibitors with antiviral activity in a transgenic mouse model. Science 2021, 371, 1374–1378. [Google Scholar] [CrossRef] [PubMed]
  72. Fabricant, D.S.; Farnsworth, N.R. The value of plants used in traditional medicine for drug discovery. Environ. Health Perspect. 2001, 109 (Suppl. S1), 69–75. [Google Scholar]
  73. Mandal, A.; Jha, A.K.; Hazra, B. Plant Products as Inhibitors of Coronavirus 3CL Protease. Front. Pharmacol. 2021, 12, 583387. [Google Scholar] [CrossRef]
  74. Solecki, R.S. Shanidar IV, a Neanderthal Flower Burial in Northern Iraq. Science 1975, 190, 880–881. [Google Scholar] [CrossRef]
  75. Cragg, G.M.; Newman, D.J. Natural product drug discovery in the next millennium. Pharm. Biol. 2001, 39 (Suppl. S1), 8–17. [Google Scholar] [PubMed] [Green Version]
  76. Denaro, M.; Smeriglio, A.; Barreca, D.; De Francesco, C.; Occhiuto, C.; Milano, G.; Trombetta, D. Antiviral activity of plants and their isolated bioactive compounds: An update. Phytother. Res. 2019, 34, 742–768. [Google Scholar] [CrossRef]
  77. Khursheed, A.; Jain, V.; Rasool, A.; Rather, M.A.; Malik, N.A.; Shalla, A.H. Molecular scaffolds from mother nature as possible lead compounds in drug design and discovery against coronaviruses: A landscape analysis of published literature and molecular docking studies. Microb. Pathog. 2021, 157, 104933. [Google Scholar] [CrossRef] [PubMed]
  78. Lin, L.-T.; Hsu, W.-C.; Lin, C.-C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complement. Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Nuraskin, C.A.; Marlina; Idroes, R.; Soraya, C. Djufri Activities inhibition methanol extract Laban Leaf (Vitex pinnata) on growth of bacteria S. mutans Atcc 31987. IOP Conf. Series Mater. Sci. Eng. 2019, 523, 012008. [Google Scholar] [CrossRef] [Green Version]
  80. Pourkhosravani, E.; Nayeri, F.D.; Bazargani, M.M. Decoding antibacterial and antibiofilm properties of cinnamon and cardamom essential oils: A combined molecular docking and experimental study. AMB Express 2021, 11, 143. [Google Scholar] [CrossRef]
  81. Rahmad, R.; Earlia, N.; Nabila, C.; Inayati, I.; Amin, M.; Prakoeswa, C.R.S.; Khairan, K.; Idroes, R. Antibacterial cream formulation of ethanolic Pliek U extracts and ethanolic residue hexane Pliek U extracts against Staphylococcus aureus. IOP Conf. Ser. Mater. Sci. Eng. 2019, 523, 012011. [Google Scholar] [CrossRef]
  82. Sardi, J.C.O.; Scorzoni, L.; Bernardi, T.; Fusco-Almeida, A.M.; Mendes Giannini, M.J.S. Candida species: Current epidemiology, pathogenicity, biofilm formation, natural antifungal products and new therapeutic options. J. Med. Microbiol. 2013, 62, 10–24. [Google Scholar] [CrossRef]
  83. El-Saber Batiha, G.; Magdy Beshbishy, A.; Wasef, L.G.; Elewa, Y.H.; Al-Sagan, A.A.; El-Hack, A.; Taha, M.E.; Abd-Elhakim, Y.M.; Prasad Devkota, H. Chemical Constituents and Pharmacological Activities of Garlic (Allium sativum L.): A Review. Nutrients 2020, 12, 872. [Google Scholar] [CrossRef] [Green Version]
  84. Liana, D.; Phanumartwiwath, A. Leveraging knowledge of Asian herbal medicine and its active compounds as COVID-19 treatment and prevention. J. Nat. Med. 2021, 76, 20–37. [Google Scholar] [CrossRef]
  85. Grigore, A.; Cord, D.; Tanase, C.; Albulescu, R. Herbal medicine, a reliable support in COVID therapy. J. Immunoass. Immunochem. 2020, 41, 976–999. [Google Scholar] [CrossRef] [PubMed]
  86. Gao, L.Q.; Xu, J.; Chen, S.D. In Silico Screening of Potential Chinese Herbal Medicine Against COVID-19 by Targeting SARS-CoV-2 3CLpro and Angiotensin Converting Enzyme II Using Molecular Docking. Chin. J. Integr. Med. 2020, 26, 527–532. [Google Scholar] [CrossRef]
  87. Shree, P.; Mishra, P.; Selvaraj, C.; Singh, S.K.; Chaube, R.; Garg, N.; Tripathi, Y.B. Targeting COVID-19 (SARS-CoV-2) main protease through active phytochemicals of ayurvedic medicinal plants—Withania somnifera (Ashwagandha), Tinospora cordifolia (Giloy) and Ocimum sanctum (Tulsi)—A molecular docking study. J. Biomol. Struct. Dyn. 2022, 40, 190–203. [Google Scholar] [CrossRef] [PubMed]
  88. Shaik, F.B.; Swarnalatha, K.; Mohan, M.; Thomas, A.; Chikati, R.; Sandeep, G.; Maddu, N. Novel antiviral effects of chloroquine, hydroxychloroquine, and green tea catechins against SARS-CoV-2 main protease (Mpro) and 3C-like protease for COVID-19 treatment. Clin. Nutr. Open Sci. 2022, 42, 62–72. [Google Scholar] [CrossRef]
  89. Sudeep, H.V.; Gouthamchandra, K.; Shyamprasad, K. Molecular docking analysis of Withaferin A from Withania somnifera with the Glucose regulated protein 78 (GRP78) receptor and the SARS-CoV-2 main protease. Bioinformation 2020, 16, 411–417. [Google Scholar] [CrossRef]
  90. 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, 73, 104146. [Google Scholar] [CrossRef] [PubMed]
  91. Nguyen TT, H.; Jung, J.H.; Kim, M.K.; Lim, S.; Choi, J.M.; Chung, B.; Kim, D. The Inhibitory Effects of Plant Derivate Polyphenols on the Main Protease of SARS Coronavirus 2 and Their Structure-Activity Relationshi. Molecules 2021, 26, 1924. [Google Scholar] [CrossRef]
  92. Khwaza, V.; Oyedeji, O.O.; Aderibigbe, B.A. Antiviral Activities of Oleanolic Acid and Its Analogues. Molecules 2018, 23, 2300. [Google Scholar] [CrossRef] [Green Version]
  93. Ghosh, R.; Chakraborty, A.; Biswas, A.; Chowdhuri, S. Depicting the inhibitory potential of polyphenols from Isatis indigotica root against the main protease of SARS-CoV-2 using computational approaches. J. Biomol. Struct. Dyn. 2020, 40, 4110–4121. [Google Scholar] [CrossRef]
  94. Hu, X.; Cai, X.; Song, X.; Li, C.; Zhao, J.; Luo, W.; Zhang, Q.; Ekumi, I.O.; He, Z. Possible SARS-coronavirus 2 inhibitor revealed by simulated molecular docking to viral main protease and host toll-like receptor. Futur. Virol. 2020, 15, 359–368. [Google Scholar] [CrossRef]
  95. Zhu, Y.; Scholle, F.; Kisthardt, S.C.; Xie, D.Y. Flavonols and dihydroflavonols inhibit the main protease activity of SARS-CoV-2 and the replication of human coronavirus 229E. Virology 2022, 571, 21–33. [Google Scholar] [CrossRef] [PubMed]
  96. Agrawal, P.K.; Agrawal, C.; Blunden, G. Naringenin as a possible candidate against SARS-CoV-2 infection and in the pathogenesis of COVID-19. Nat. Prod. Commun. 2021, 16, 1934578X211066723. [Google Scholar] [CrossRef]
  97. Mishra, A.; Rath, S.C.; Baitharu, I.; Bag, B.P. Millet Derived Flavonoids as Potential SARS-CoV-2 Main Protease Inhibitors: A Computational Approach; Cambridge Open Engage: Cambridge, UK, 2020. [Google Scholar]
  98. Su, H.X.; Yao, S.; Zhao, W.F.; Li, M.J.; Liu, J.; Shang, W.J.; Xu, Y.C. Anti-SARS-CoV-2 activities in vitro of Shuanghuanglian preparations and bioactive ingredients. Acta Pharmacol. Sin. 2020, 41, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
  99. Adem, Ş.; Eyupoglu, V.; Sarfraz, I.; Rasul, A.; Zahoor, A.F.; Ali, M.; Elfiky, A.A. Caffeic acid derivatives (CAFDs) as inhibitors of SARS-CoV-2: CAFDs-based functional foods as a potential alternative approach to combat COVID-19. Phytomedicine 2021, 85, 153310. [Google Scholar] [CrossRef] [PubMed]
  100. Bahun, M.; Jukić, M.; Oblak, D.; Kranjc, L.; Bajc, G.; Butala, M.; Ulrih, N.P. Inhibition of the SARS-CoV-2 3CLpro main protease by plant polyphenols. Food Chem. 2022, 373, 131594. [Google Scholar] [CrossRef] [PubMed]
  101. Shahab, S.; Kaviani, S.; Sheikhi, M.; Almodarresiyeh, H.A.; Al Saud, S. Dft calculations and in silico study of chlorogenic, ellagic and quisqualic acids as potential inhibitors of SARS-CoV-2 main protease mpro. Biointerface Res. Appl. Chem. 2022, 12, 61–73. [Google Scholar]
  102. Ngwe Tun, M.M.; Luvai, E.; Nwe, K.M.; Toume, K.; Mizukami, S.; Hirayama, K.; Morita, K. Anti-SARS-CoV-2 activity of various PET-bottled Japanese green teas and tea compounds in vitro. Arch. Virol. 2022, 167, 1547–1557. [Google Scholar] [CrossRef]
  103. Khan, A.; Heng, W.; Wang, Y.; Qiu, J.; Wei, X.; Peng, S.; Wei, D.Q. In silico and in vitro evaluation of kaempferol as a potential inhibitor of the SARS-CoV-2 main protease (3CLpro). Phytother. Res. 2021, 35, 2841–2845. [Google Scholar] [CrossRef]
  104. Owis, A.I.; El-Hawary, M.S.; El Amir, D.; Aly, O.M.; Abdelmohsen, U.R.; Kamel, M.S. Molecular docking reveals the potential of Salvadora persica flavonoids to inhibit COVID-19 virus main protease. RSC Adv. 2020, 10, 19570–19575. [Google Scholar] [CrossRef]
  105. Shahhamzehei, N.; Abdelfatah, S.; Efferth, T. In Silico and In Vitro Identification of Pan-Coronaviral Main Protease Inhibitors from a Large Natural Product Library. Pharmaceuticals 2022, 15, 308. [Google Scholar] [CrossRef]
  106. Choudhry, N.; Zhao, X.; Xu, D.; Zanin, M.; Chen, W.; Yang, Z.; Chen, J. Chinese therapeutic strategy for fighting COVID-19 and potential small-molecule inhibitors against severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2). J. Med. Chem. 2020, 63, 13205–13227. [Google Scholar] [CrossRef] [PubMed]
  107. Jo, S.; Kim, S.; Shin, D.H.; Kim, M.S. Inhibition of SARS-CoV 3CL protease by flavonoids. J. Enzyme Inhib. Med. Chem. 2020, 35, 145–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Liao, Q.; Chen, Z.; Tao, Y.; Zhang, B.; Wu, X.; Yang, L.; Wang, Z. An integrated method for optimized identification of effective natural inhibitors against SARS-CoV-2 3CLpro. Sci. Rep. 2021, 11, 22796. [Google Scholar] [CrossRef] [PubMed]
  109. Hariono, M.; Hariyono, P.; Dwiastuti, R.; Setyani, W.; Yusuf, M.; Salin, N.; Wahab, H. Potential SARS-CoV-2 3CLpro Inhibitors from Chromene, Flavonoid and Hydroxamic Acid Compound based on FRET Assay, Docking and Pharmacophore Studies. Results Chem. 2021, 3, 100195. [Google Scholar] [CrossRef] [PubMed]
  110. Ghosh, R.; Chakraborty, A.; Biswas, A.; Chowdhuri, S. Evaluation of green tea polyphenols as novel corona virus (SARS-CoV-2) main protease (Mpro) inhibitors—An in silico docking and molecular dynamics simulation study. J. Biomol. Struct. Dyn. 2021, 39, 4362–4374. [Google Scholar] [CrossRef]
  111. Rehman MF, U.; Akhter, S.; Batool, A.I.; Selamoglu, Z.; Sevindik, M.; Eman, R.; Aslam, M. Effectiveness of Natural Antioxidants against SARS-CoV-2? Insights from the In-Silico World. Antibiotics 2021, 10, 1011. [Google Scholar] [CrossRef]
  112. Alrasheid, A.A.; Babiker, M.Y.; Awad, T.A. Evaluation of certain medicinal plants compounds as new potential inhibitors of novel corona virus (COVID-19) using molecular docking analysis. Silico Pharmacol. 2021, 9, 10. [Google Scholar] [CrossRef]
  113. Emon, N.U.; Alam, M.M.; Akter, I.; Akhter, S.; Sneha, A.A.; Irtiza, M.; Hossain, S. Virtual screenings of the bioactive constituents of tea, prickly chaff, catechu, lemon, black pepper, and synthetic compounds with the main protease (Mpro) and human angiotensin-converting enzyme 2 (ACE 2) of SARS-CoV-2. Futur. J. Pharm. Sci. 2021, 7, 121. [Google Scholar] [CrossRef]
  114. Tripathi, M.K.; Singh, P.; Sharma, S.; Singh, T.P.; Ethayathulla, A.S.; Kaur, P. Identification of bioactive molecule from Withania somnifera (Ashwagandha) as SARS-CoV-2 main protease inhibitor. J. Biomol. Struct. Dyn. 2021, 39, 5668–5681. [Google Scholar] [CrossRef]
  115. Zígolo, M.A.; Goytia, M.R.; Poma, H.R.; Rajal, V.B.; Irazusta, V.P. Virtual screening of plant-derived compounds against SARS-CoV-2 viral proteins using computational tools. Sci. Total Environ. 2021, 781, 146400. [Google Scholar] [CrossRef]
  116. van de Sand, L.; Bormann, M.; Alt, M.; Schipper, L.; Heilingloh, C.S.; Steinmann, E.; Krawczyk, A. Glycyrrhizin Effectively Inhibits SARS-CoV-2 Replication by Inhibiting the Viral Main Protease. Viruses 2021, 13, 609. [Google Scholar] [CrossRef] [PubMed]
  117. Antonopoulou, I.; Sapountzaki, E.; Rova, U.; Christakopoulos, P. Inhibition of the main protease of SARS-CoV-2 (Mpro) by repurposing/designing drug-like substances and utilizing nature’s toolbox of bioactive compounds. Comput. Struct. Biotechnol. J. 2022, 20, 1306–1344. [Google Scholar] [CrossRef] [PubMed]
  118. Veerasamy, R.; Karunakaran, R. Molecular docking unveils the potential of andrographolide derivatives against COVID-19: An in silico approach. J. Genet. Eng. Biotechnol. 2022, 20, 58. [Google Scholar] [CrossRef] [PubMed]
  119. Shady, N.H.; Youssif, K.A.; Sayed, A.M.; Belbahri, L.; Oszako, T.; Hassan, H.M.; Abdelmohsen, U.R. Sterols and Triterpenes: Antiviral Potential Supported by In-Silico Analysis. Plants 2020, 10, 41. [Google Scholar] [CrossRef] [PubMed]
  120. Falade, V.A.; Adelusi, T.I.; Adedotun, I.O.; Abdul-Hammed, M.; Lawal, T.A.; Agboluaje, S.A. In silico investigation of saponins and tannins as potential inhibitors of SARS-CoV-2 main protease (Mpro). Silico Pharmacol. 2021, 9, 1–15. [Google Scholar] [CrossRef]
  121. Wang, S.C.; Chen, Y.; Wang, Y.C.; Wang, W.J.; Yang, C.S.; Tsai, C.L.; Hung, M.C. Tannic acid suppresses SARS-CoV-2 as a dual inhibitor of the viral main protease and the cellular TMPRSS2 protease. Am. J. Cancer Res. 2020, 10, 4538–4546. [Google Scholar]
  122. Sisakht, M.; Mahmoodzadeh, A.; Darabian, M. Plant-derived chemicals as potential inhibitors of SARS-CoV-2 main protease (6LU7), a virtual screening study. Phytother. Res. 2021, 35, 3262–3274. [Google Scholar] [CrossRef]
  123. Kaul, R.; Paul, P.; Kumar, S.; Büsselberg, D.; Dwivedi, V.D.; Chaari, A. Promising antiviral activities of natural flavonoids against SARS-CoV-2 targets: Systematic review. Int. J. Mol. Sci. 2021, 22, 11069. [Google Scholar] [CrossRef]
  124. Fraga, C.G.; Croft, K.D.; Kennedy, D.O.; Tomás-Barberán, F.A. The effects of polyphenols and other bioactives on human health. Food Funct. 2019, 10, 514–528. [Google Scholar] [CrossRef] [Green Version]
  125. Tsao, R. Chemistry and Biochemistry of Dietary Polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
  126. Kim, J.-S.; Kang, O.-J.; Gweon, O.-C. Comparison of phenolic acids and flavonoids in black garlic at different thermal processing steps. J. Funct. Foods 2013, 5, 80–86. [Google Scholar] [CrossRef]
  127. Li, F.; Song, X.; Su, G.; Wang, Y.; Wang, Z.; Jia, J.; Qing, S.; Huang, L.; Wang, Y.; Zheng, K.; et al. Amentoflavone Inhibits HSV-1 and ACV-Resistant Strain Infection by Suppressing Viral Early Infection. Viruses 2019, 11, 466. [Google Scholar] [CrossRef] [Green Version]
  128. Bernatoniene, J.; Kopustinskiene, D.M. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 23, 965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Kaihatsu, K.; Yamabe, M.; Ebara, Y. Antiviral Mechanism of Action of Epigallocatechin-3-O-gallate and Its Fatty Acid Esters. Molecules 2018, 23, 2475. [Google Scholar] [CrossRef] [Green Version]
  130. Lingwan, M.; Shagun, S.; Pahwa, F.; Kumar, A.; Verma, D.K.; Pant, Y.; Masakapalli, S.K. Phytochemical rich Himalayan Rhododendron arboreum petals inhibit SARS-CoV-2 infection in vitro. J. Biomol. Struct. Dyn. 2021, 28, 1–11. [Google Scholar] [CrossRef]
  131. Zhu, Y.; Xie, D.-Y. Docking Characterization and in vitro Inhibitory Activity of Flavan-3-ols and Dimeric Proanthocyanidins against the Main Protease Activity of SARS-CoV-2. Front. Plant Sci. 2020, 11, 601316. [Google Scholar] [CrossRef] [PubMed]
  132. Park, R.; Jang, M.; Park, Y.-I.; Park, Y.; Jung, W.; Park, J.; Park, J. Epigallocatechin Gallate (EGCG), a Green Tea Polyphenol, Reduces Coronavirus Replication in a Mouse Model. Viruses 2021, 13, 2533. [Google Scholar] [CrossRef]
  133. Zhao, Z.; Feng, M.; Wan, J.; Zheng, X.; Teng, C.; Xie, X.; Pan, W.; Hu, B.; Huang, J.-A.; Liu, Z.-H.; et al. Research progress of epigallocatechin-3-gallate (EGCG) on anti-pathogenic microbes and immune regulation activities. Food Funct. 2021, 12, 9607–9619. [Google Scholar] [CrossRef]
  134. Maiti, S.; Banerjee, A.; Kanwar, M. Effects of theaflavin-gallate in-silico binding with different proteins of SARS-CoV-2 and host inflammation and vasoregulations referring an experimental rat-lung injury. Phytomed. Plus 2022, 2, 100237. [Google Scholar] [CrossRef]
  135. Chung, K.-T.; Wong, T.Y.; Wei, C.-I.; Huang, Y.-W.; Lin, Y. Tannins and Human Health: A Review. Crit. Rev. Food Sci. Nutr. 1998, 38, 421–464. [Google Scholar] [CrossRef]
  136. Chen, C.N.; Lin, C.P.; Huang, K.K.; Chen, W.C.; Hsieh, H.P.; Liang, P.H.; Hsu, J.T.A. Inhibition of SARS-CoV 3C-like Protease Activity by Theaflavin-3,3′-digallate (TF3). Evid. Based Complement. Alternat. Med. 2005, 2, 209–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Selvaraj, J.; Rajan, L.; Selvaraj, D.; Palanisamy, D.; Pk, K.N.; Mohankumar, S.K. Identification of (2R,3R)-2-(3,4-dihydroxyphenyl)chroman-3-yl-3,4,5-trihydroxy benzoate as multiple inhibitors of SARS-CoV-2 targets; a systematic molecular modelling approach. RSC Adv. 2021, 11, 13051–13060. [Google Scholar] [CrossRef] [PubMed]
  138. Li, M.; Hao, L.; Liu, L.; Chen, G.; Jiang, W.; Xu, W.; Zhu, C.; Yao, G.; Fang, S. Cinnamtannin B-1 Prevents Ovariectomy-Induced Osteoporosis via Attenuating Osteoclastogenesis and ROS Generation. Front. Pharmacol. 2020, 11, 1023. [Google Scholar] [CrossRef]
  139. Abookleesh, F.L.; Al-Anzi, B.S.; Ullah, A. Potential Antiviral Action of Alkaloids. Molecules 2022, 27, 903. [Google Scholar] [CrossRef]
  140. Reyes-Escogido, M.D.L.; Gonzalez-Mondragon, E.G.; Vazquez-Tzompantzi, E. Chemical and Pharmacological Aspects of Capsaicin. Molecules 2011, 16, 1253–1270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Abhaykumar, K.J.W.S.N. Phytochemical studies on Achyranthes aspera. World Sci. News 2018, 100, 16–34. [Google Scholar]
  142. Bergman, M.E.; Davis, B.; Phillips, M.A. Medically Useful Plant Terpenoids: Biosynthesis, Occurrence, and Mechanism of Action. Molecules 2019, 24, 3961. [Google Scholar] [CrossRef] [Green Version]
  143. Venkatesan, N.; Punithavathi, D.; Babu, M. Protection from Acute and Chronic Lung Diseases By Curcumin. Mol. Targets Ther. Uses Curcumin Health Dis. 2007, 595, 379–405. [Google Scholar]
  144. Lelli, D.; Sahebkar, A.; Johnston, T.P.; Pedone, C. Curcumin use in pulmonary diseases: State of the art and future perspectives. Pharmacol. Res. 2016, 115, 133–148. [Google Scholar] [CrossRef]
  145. Thimmulappa, R.K.; Mudnakudu-Nagaraju, K.K.; Shivamallu, C.; Subramaniam, K.; Radhakrishnan, A.; Bhojraj, S.; Kuppusamy, G. Antiviral and immunomodulatory activity of curcumin: A case for prophylactic therapy for COVID-19. Heliyon 2021, 7, e06350. [Google Scholar] [CrossRef]
  146. Salehi, B.; Upadhyay, S.; Erdogan Orhan, I.; Kumar Jugran, A.; LDJayaweera, S.; ADias, D.; Sharifi-Rad, J. Therapeutic Potential of α- and β-Pinene: A Miracle Gift of Nature. Biomolecules 2019, 9, 738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Alviano, D.S.A.A.C.S.; Alviano, C.S. Plant Extracts: Search for New Alternatives to Treat Microbial Diseases. Curr. Pharm. Biotechnol. 2009, 10, 106–121. [Google Scholar] [CrossRef] [PubMed]
  148. Koudou, J.; Abena, A.; Ngaissona, P.; Bessière, J. Chemical composition and pharmacological activity of essential oil of Canarium schweinfurthii. Fitoterapia 2005, 76, 700–703. [Google Scholar] [CrossRef] [PubMed]
  149. Loizzo, M.R.; Saab, A.; Tundis, R.; Statti, G.A.; Lampronti, I.; Menichini, F.; Gambari, R.; Cinatl, J.; Doerr, H.W. Phytochemical analysis and in vitro evaluation of the biological activity against herpes simplex virus type 1 (HSV-1) of Cedrus libani A. Rich. Phytomedicine 2008, 15, 79–83. [Google Scholar] [CrossRef]
  150. da Silva Rivas, A.C.; Lopes, P.M.; de Azevedo Barros, M.M.; Costa Machado, D.C.; Alviano, C.S.; Alviano, D.S. Biological Activities of a-Pinene and β-Pinene Enantiomers. Molecules 2012, 17, 6305–6316. [Google Scholar]
  151. Enmozhi, S.K.; Raja, K.; Sebastine, I.; Joseph, J. Andrographolide as a potential inhibitor of SARS-CoV-2 main protease: An in silico approach. J. Biomol. Struct. Dyn. 2021, 39, 3092–3098. [Google Scholar] [CrossRef]
  152. Alsaffar, D.F.; Yaseen, A. In Silico Molecular Docking Studies of Medicinal Arabic Plant-Based Bioactive Compounds as a Promising Drug Candidate against COVID-19. Int. J. Innov. Sci. Res. Technol. 2020, 5, 876–896. [Google Scholar]
  153. Chaudhuri, S.K.; Fullas, F.; Brown, D.M.; Wani, M.C.; Wall, M.E.; Cai, L.; Mar, W.; Lee, S.K.; Luo, Y.; Zaw, K.; et al. Isolation and Structural Elucidation of Pentacyclic Triterpenoids from Maprounea africana. J. Nat. Prod. 1995, 58, 1–9. [Google Scholar] [CrossRef]
  154. Kai, G.; Xu, H.; Zhou, C.; Liao, P.; Xiao, J.; Luo, X.; You, L.; Zhang, L. Metabolic engineering tanshinone biosynthetic pathway in Salvia miltiorrhiza hairy root cultures. Metab. Eng. 2011, 13, 319–327. [Google Scholar] [CrossRef]
  155. Deng, C.; Hao, X.; Shi, M.; Fu, R.; Wang, Y.; Zhang, Y.; Zhou, W.; Feng, Y.; Makunga, N.P.; Kai, G. Tanshinone production could be increased by the expression of SmWRKY2 in Salvia miltiorrhiza hairy roots. Plant Sci. 2019, 284, 1–8. [Google Scholar] [CrossRef]
  156. Zhou, W.; Wang, S.; Shen, Y.; Liu, Y.; Maoz, I.; Gao, X.; Chen, C.; Liu, T.; Wang, C.; Kai, G. Overexpression of SmSCR1 Promotes Tanshinone Accumulation and Hairy Root Growth in Salvia miltiorrhiza. Front. Plant Sci. 2022, 13, 475. [Google Scholar] [CrossRef] [PubMed]
  157. Xie, Y.; Ding, M.; Yin, X.; Wang, G.; Zhang, B.; Chen, L.; Dong, J. MAPKK2/4/5/7-MAPK3-JAZs modulate phenolic acid biosynthesis in Salvia miltiorrhiza. Phytochemistry 2022, 199, 113177. [Google Scholar] [CrossRef] [PubMed]
  158. Wang, Y.; Chen, S.; Yu, O. Metabolic engineering of flavonoids in plants and microorganisms. Appl. Microbiol. Biotechnol. 2011, 91, 949–956. [Google Scholar] [CrossRef]
  159. Rai, A.; Smita, S.S.; Singh, A.K.; Shanker, K.; Nagegowda, D.A. Heteromeric and Homomeric Geranyl Diphosphate Synthases from Catharanthus roseus and Their Role in Monoterpene Indole Alkaloid Biosynthesis. Mol. Plant 2013, 6, 1531–1549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Duran, N.; Polat, M.F.; Aktas, D.A.; Alagoz, M.A.; Ay, E.; Cimen, F.; Tek, E.; Anil, B.; Burmaoglu, S.; Algul, O. New chalcone derivatives as effective against SARS-CoV-2 agent. Int. J. Clin. Pract. 2021, 75, e14846. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Basic flavonoid structures.
Figure 1. Basic flavonoid structures.
Plants 11 01862 g001
Table 1. Phytochemicals reported to have potential inhibitory properties against SARS-CoV-2 Mpro.
Table 1. Phytochemicals reported to have potential inhibitory properties against SARS-CoV-2 Mpro.
Class of CompoundCompoundDistributed inIncluding Food Plants and Spices *Inhibition (%), with IC50 Value, μMBinding Energy ** (kcal/mol)Ref.
IsoflavoneDaidzeinStreptomyces, and predominantly in Fabaceae plant familyVigna radiata,
Glycine max
56−6.5[93]
IsoflavonePuerarinPredominantly in Fabaceae plant familyCicer arietinum,
Glycine max,
Glycyrrhiza glabra,
Phaseolus vulgaris,
Pisum sativum,
Vigna radiata
42 ± 2−6.63[94]
FlavonolMyricetinWidely distributed in various plant familiesBuchanania lanzan,
Mangifera indica,
Asparagus officinalis,
Davidsonia pruriens,
Hippophae rhamnoides,
Vicia faba,
Salvia hispanica,
Thymus capitatus,
Punica granatum,
Hibiscus sabdariffa L.,
Moringa oleifera,
Eugenia jambolana,
Pimenta dioica,
Plinia pinnata,
Syzygium aromaticum,
Syzygium cumini,
Syzygium samarangense,
Diploknema butyracea,
Ampelopsis grossedentata,
Morella rubra
43 ± 1−22.13[41,91]
FlavonolQuercetinMucor hiemalis, and widely distributed in various plant familiesAllium cepa,
Allium Sativum,
Allium ascalonicum,
Mangifera indica,
Annona muricata,
Asparagus officinalis,
Capparis spinosa,
Carica papaya,
Garcinia cowa,
Garcinia dulcis,
Brassica oleracea var. gongylodes,
Raphanus sativus,
Momordica charantia,
Ceratonia siliqua,
Vicia faba,
Crocus sativus,
Punica granatum,
Toona sinensis,
Moringa stenopetala,
Musa acuminata,
Psidium guajava,
Phyllanthus emblica,
Zea mays,
Nigella sativa,
Eriobotrya japonica,
Prunus avium,
Kadsura heteroclita,
Capsicum annuum,
Zingiber officinale
93 ± 5−7.6[23,91]
FlavonolQuercetagetin (Quercetagenin)Asteraceae, Eriocaulaceae, Fabaceae plant familiesAcacia catechu,
Leucaena glauca
145 ± 6−15.2[91]
FlavanonolAmpelopsin (dihydromyricetin)Widely distributed in various plant familiesAsparagus officinalis,
Punica granatum L.,
Pimenta dioica,
Zea mays,
Syzygium cumini,
Capsicum annuum,
Vitis rotundifolia,
Manilkara zapota,
128 ± 5−7.5[91,95]
FlavanonolAmpelopsin-4′-O-α-d-glucopyranosideWidely distributed in various plant families-195 ± 57.4[91]
FlavanoneNaringeninWidely distributed in various plant families Camellia sinensis,
Prunus cerasus,
Prunus persica,
Citrus grandi, etc.
150 ± 10−7.7[91,96]
Flavan-3-olEpigallocatechin gallate (EGCG)Widely distributed in various plant familiesVitis vinifera171 ± 5−7.6
−8.2
[88,91,93]
FlavoneVitexinWidely distributed in various plant families Pisum sativum180 ± 6−7.6[97]
Hydrocinnamic acidChlorogenic acidWidely distributed in various plant families Lactuca sativa39.48 ± 5.51−12.98[91,98]
Dihydroxycinnamic acidCaffeic acidWidely distributed in various plant familiesActinidia deliciosa,
Allium Sativum,
Mangifera indica,
Ilex paraguariensis,
Carica papaya,
Beta vulgaris,
Terminalia catappa,
Terminalia chebula,
Brassica oleracea var. gongylodes,
Raphanus sativus,
Momordica charantia,
Arachis hypogaea,
Cicer arietinum,
Glycine max,
Phaseolus vulgaris,
Pisum sativum,
Tetrapleura tetraptera,
Ocimum basilicum,
Rosmarinus officinalis,
Thymus capitatus,
Punica granatum,
Triticum aestivum,
Zea mays,
Crataegus pinnatifida,
Prunus avium,
Coffea arabica,
Citrus limon,
Citrus sinensis,
Solanum lycopersicum,
Solanum phureja,
Solanum pimpinellifollium,
Solanum tuberosum,
Curcuma longa,
Bergera koenigii
197 ± 1−12.4985[91,99]
PolyphenolEllagic acidWidely distributed in various plant families Mangifera indica,
Terminalia chebula,
Punica granatum,
Moringa oleifera,
Moringa peregrine,
Moringa stenopetala,
Eugenia jambolana,
Myrciaria cauliflora,
Syzygium aromaticum,
Syzygium cumini,
Emblica officinalis,
Rubus idaeus L.,
11.8 ± 5.7−15.955[100,101]
PhenylpropanoidChicoric acidAlliaceae, Asteraceae, and Labiatae plant families Lactuca sativa,
Ocimum basilicum,
Cichorium intybus
-−8.2[93]
PolyphenolGallocatechin gallate (GCG)Cistaceae, Elaeagnaceae, Ericaceae, Polygonaceae, Theaceae, and Vitaceae plant familiesHippophae rhamnoides,
Camellia sinensis,
Vitis vinifera
5.774 ± 0.805−9[93]
Flavan-3-olEpicatechin gallate (ECG)Cistaceae, Elaeagnaceae, Ericaceae, Polygonaceae, Theaceae, and Vitaceae plant familiesHippophae rhamnoides,
Camellia sinensis,
Vitis vinifera,
Rheum sp.
12.5−8.2[93,102]
FlavonoidsKaempferol glycosidesWidely distributed in various plant familiesPrunus avium,
Allium cepa
125.00−7.4,
−8.1
[103,104]
FlavonoidsIsorhamnetin glycosidesWidely distributed in various plant families Brassica oleracea,
Allium ascalonicum
13.13−6.6,
−8.2
[104,105]
FlavonoidsPectolinarinLabiatae, Plantaginaceae, and Verbenaceae plant families 37.7−8.2[40,106,107]
FlavonoidHerbacetinAsteraceae, Chenopodiaceae, Crassulaceae, Ephedraceae, Equisetaceae, Linaceae, Malvaceae, Papaveraceae, Phrymaceae, Primulaceae, Rosaceae, Rutaceae, and Taxaceae plant familiesLinum usitatissimum,
Citrus limon
33.1−7.2[40,106]
FlavonoidRhoifolin (apigenin-7-O-rhamnoglucoside)Acanthaceae, Anacardiaceae, Apocynaceae, Fabaceae, Lythraceae, Oleaceae, Rutaceae, and Caprifoliaceae plant familiesVicia faba,
Hordeum vulgare L
27.4−8.2[40,106]
Flavonoid metaboliteViceninRare compoundTrigonella foenum-graecum38.856−8.97[87,108]
FlavoneIsorientin 4′-O-glucoside 2″-O-p-hydroxybenzoateGentianaceae and
Lamiaceae
Ocimum sanctum-−8.55[87]
BiflavonoidAmentoflavoneVarious plant species: yew, juniper, oak, and willowGarcinia brasiliensis,
Garcinia dulcis,
Garcinia hombroniana,
Garcinia indica,
Garcinia intermedia,
Garcinia livingstonei,
Garcinia madruno,
Garcinia mangostana,
Garcinia morella,
Garcinia wightii,
Garcinia xanthochymus,
-−10.0[23]
FlavonoidSilymarin (silibinin)AsteraceaeSilybum marianum46.88−7.6 [23]
IsoflavoneTorvanol ASolanaceae -−7.5[23]
FlavoneScutellareinAsphodelaceae,
Asteraceae,
Fabaceae, Bignoniaceae, Labiatae, Plantaginaceae, Polygonaceae, and Verbenaceae plant families
Oroxylum indicum,
Garcinia andamanica
Origanum majorana
-−7.4[23]
FlavoneApigeninWidely distributed in various plant families, mainly in LabiataeArtemisia diffusa.,
Ocimum americanum var. pilosum
Ocimum basilicum,
Ocimum x citriodorum,
Rosmarinus officinalis,
Salvia officinalis,
Thymus piperella,
Passiflora foetida,
Piper peepuloides,
Kaempferia parviflora
925−7.1[23,109]
Isoflavone5,7,3′,4′-Tetrahydroxy-2′-(3,3-dimethylallyl) isoflavoneFabaceae -−29.57 [41]
PolyphenolMethyl rosmarinateLabiataeRosmarinus officinalis21.32−20.62[41,110]
FlavonoidBaicalinMainly in LabiataeOroxylum indicum6.41 ± 0.95 μM in vitro
27.87 ± 0.04 μM in cells
−8.85[99,111]
FlavonoidBaicaleinMainly in LabiataeOroxylum indicum0.94 ± 0.20 μM in vitro
2.94 ± 1.19 μM in cells
[98]
AlkaloidCapsaicinSolanaceaeCapsicum annuum-−13.90[112]
AlkaloidPsychotrine Mainly in Rubiaceae -−13.5[112]
AlkaloidAchyranthineAmaranthaceae -4.1[113]
TerpenoidWithanoside VSolanaceae 5.774 ± 0.805 −10.32[114]
TriterpenoidUrsolic acidApocynaceae, Asteraceae, Boraginaceae, Dryopteridaceae, Ericaceae, Gesneriaceae, Labiatae, Lamiaceae, Lythraceae, Moraceae, Myricaceae, Nothofagaceae, Oleaceae, Rosaceae, Rubiaceae, Solanaceae, Stilbaceae, and Ulmaceae plant familiesVaccinium macrocarpon,
Punica granatum,
Olea europaea,
Prunus avium,
Pyrus spp.
12.6−8.2[114]
TriterpenoidGlycyrrhizic acidAsteraceae and FabaceaeStevia rebaudiana,
Glycyrrhiza glabra
-−8.03[115,116]
Pentacyclic triterpenoidTorvoside HSolanaceae--−8.4[23]
Pentacyclic triterpenoidLupeolIn Coprinaceae, and widely distributed in various plant familiesCichorium intybus,
Zanthoxylum armatum,
Olea europaea,
Myrica rubra,
Morus alba,
Ficus carica,
Carica papaya
-−7.6[116]
DiterpeneScopadulcic acid BPlantaginaceae (Scoparia dulcis)--−8.5[23]
DiterpeneOvatodiolideLamiaceae--−6.9[23]
TerpeneCurcuminMainly in ZingiberaceaeCurcuma longa,
Curcuma mangga,
Zingiber officinale
11.9−6.5[23,117]
TerpeneParthenolideAsteraceae, Magnoliaceae, and Celastraceae plant families--−6.0[23]
MeroterpenoidIllicinone AIlliaceaeIlliciumverum-−5.0[23]
MeroterpenoidPiperitenoneLabiatae and PoaceaeMentha spp.-−4.3[23]
Cyclic monoterpeneLimonene Widely distributed in various plant familiesCitrus aurantium,
Citrus aurantifolia,
Citrus bergamia,
Citrus grandis,
Citrus junos,
Citrus latifolia,
Citrus limettioides,
Citrus limon,
Citrus medica,
Citrus paradisi,
Citrus reticulata,
Citrus sinensis,
Zanthoxylum armatum,
Allium sativum,
Anacardium occidentale,
Mangifera indica,
Monodora myristica,
Xylopia aethiopica,
Cuminum cyminum.
Foeniculum vulgare,
Petroselinum crispum,
Porophyllum ruderale,
Beta vulgaris,
Ocimum basilicum,
Thymus piperella,
Acca sellowiana,
Psidium guajava,
Averrhoa carambola,
Piper nigrum
Prunus avium,
Coffea arabica,
Coffea canephora,
Citrus aurantifolia,
Curcuma amada,
Curcuma longa
-−5.2[113]
Cyclic monoterpeneSabineneLabiatae, Cupressaceae, Myristicaceae, and Pinaceae plant families--−4.8[113]
Bicyclic monoterpenePineneWidely distributed in various plant familiesPiper nigrum,
Allium sativum, Anacardium occidentale,
Mangifera indica,
Pistacia vera,
Monodora myristica,
Cuminum cyminum,
Foeniculum vulgare,
Petroselinum crispum,
Ocimum basilicum,
Origanum vulgare,
Rosmarinus officinalis,
Myristica fragrans,
Eriobotrya japonica,
Fragaria vesca,
Citrofortunella mitis,
Citrus aurantifolia,
Citrus spp.,
Curcuma mangga,
Curcuma amada,
Aframomum melegueta,
Solanum lycopersicum,
Zanthoxylum armatum,
-−4.6[113]
Labdane diterpenoidAndrographolideAcanthaceae--−6.6[118]
Triterpene1β-hydroxyaleuritolic acid 3-p-hydroxybenzoateEuphorbiaceae--−8.5[119]
Steroidal lactoneWithaferin ASolanaceae--−9.83[89]
TanninTannic acidEphedraceae and Geraniaceae-13.4−7.5[120,121]
* KNApSAcK Core System (KNApSAcK DB group (skanayagtc.naist.jp). ** The binding energy of Mpro to Lopinavir (−9.1 Kcal/mol) or Nelfinavir (−8.4 Kcal/mol) is given for comparison of this value for herbal compounds [122].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Issa, S.S.; Sokornova, S.V.; Zhidkin, R.R.; Matveeva, T.V. The Main Protease of SARS-CoV-2 as a Target for Phytochemicals against Coronavirus. Plants 2022, 11, 1862. https://doi.org/10.3390/plants11141862

AMA Style

Issa SS, Sokornova SV, Zhidkin RR, Matveeva TV. The Main Protease of SARS-CoV-2 as a Target for Phytochemicals against Coronavirus. Plants. 2022; 11(14):1862. https://doi.org/10.3390/plants11141862

Chicago/Turabian Style

Issa, Shaza S., Sofia V. Sokornova, Roman R. Zhidkin, and Tatiana V. Matveeva. 2022. "The Main Protease of SARS-CoV-2 as a Target for Phytochemicals against Coronavirus" Plants 11, no. 14: 1862. https://doi.org/10.3390/plants11141862

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop