Next Article in Journal
Effects of Fasting on the Physiological and Psychological Responses in Middle-Aged Men
Next Article in Special Issue
Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection
Previous Article in Journal
Effect of Dietary Patterns on Inflammatory Bowel Disease: A Machine Learning Bibliometric and Visualization Analysis
Previous Article in Special Issue
Cardamonin as a p38 MAPK Signaling Pathway Activator Inhibits Human Coronavirus OC43 Infection in Human Lung Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection

1
Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
2
School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus, Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
3
School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, China
4
Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
*
Authors to whom correspondence should be addressed.
Nutrients 2023, 15(15), 3443; https://doi.org/10.3390/nu15153443
Submission received: 19 July 2023 / Revised: 28 July 2023 / Accepted: 2 August 2023 / Published: 3 August 2023

Abstract

:
The exploration of non-toxic and cost-effective dietary components, such as epigallocatechin 3-gallate and myricetin, for health improvement and disease treatment has recently attracted substantial research attention. The recent COVID-19 pandemic has provided a unique opportunity for the investigation and identification of dietary components capable of treating viral infections, as well as gathering the evidence needed to address the major challenges presented by public health emergencies. Dietary components hold great potential as a starting point for further drug development for the treatment and prevention of SARS-CoV-2 infection owing to their good safety, broad-spectrum antiviral activities, and multi-organ protective capacity. Here, we review current knowledge of the characteristics—chemical composition, bioactive properties, and putative mechanisms of action—of natural bioactive dietary flavonoids with the potential for targeting SARS-CoV-2 and its variants. Notably, we present promising strategies (combination therapy, lead optimization, and drug delivery) to overcome the inherent deficiencies of natural dietary flavonoids, such as limited bioavailability and poor stability.

1. Introduction

The outbreak of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the deadliest virus since the 1918 influenza virus, has posed a serious threat to global health security [1]. Tremendous research efforts have been undertaken, aiming at controlling and/or treating SARS-CoV-2 infection [2,3,4]. To date, several small-molecule antivirals (remdesivir, ritonavir-boosted nirmatrelvir, and molnupiravir), vaccines, and monoclonal antibodies have been approved or authorized by the Food and Drug Administration (FDA) of the United States of America for the treatment of COVID-19 [5,6,7,8]. Although the pandemic appears to be on a downward trend, the potential emergence of new SARS-CoV-2 variants still represents a threat to humans, given their intrinsic transmissibility, immune escape, virulence, and susceptibility to available treatments [9,10,11]. Taking virulence as an example, if it is assumed that the mortality rate among SARS-CoV-2-infected individuals is similar to that for seasonal influenza, we can expect the annual burden of future influenza to be twice that of previous influenza. The additional burden associated with “long COVID” (e.g., respiratory symptoms and cognitive dysfunction) may also be non-negligible [12,13]. Accordingly, an inexpensive, convenient, and rapidly up-scalable response model is required to address future coronavirus pandemics.
Natural products (including herbal medicine) play an irreplaceable role in the treatment of SARS-CoV-2 infection [14,15]. Increasing evidence supports that many functional foods and nutraceuticals have potential for use in the prevention and treatment of viral infections [16]. In recent years, flavonoids have attracted much attention from pharmaceutical chemists and organic chemists due to their efficiency and low toxicity for health improvement and disease treatment. Their active components, such as epigallocatechin 3-gallate (EGCG) and myricetin (Figure 1), have drawn considerable attention as potential agents for COVID-19 treatment owing to their multitargeting potential (SARS-CoV-2 Mpro, angiotensin-converting enzyme 2 [ACE2, the primary target of SARS-CoV-2 in host cells], and RNA-dependent RNA polymerase [RdRp, an essential enzyme in RNA viruses, which is a key player driving the viral replication and transcription machinery], among other targets), broad-spectrum activities, and low toxicity [17,18]. In this review, we summarize the characteristics of natural dietary flavonoids, including their bioactive properties and potential mechanisms of action, associated with the prevention and treatment of COVID-19, and discuss strategies aiming at improving their bioavailability, chemical stability, and delivery. Finally, we present promising strategies (combination therapy and lead optimization) for overcoming the inherent shortcomings (limited bioavailability and poor chemical stability) of natural dietary flavonoids as therapeutics for SARS-CoV-2 infection.

2. Epigallocatechin 3-Gallate—A Green Tea-Derived, Multitargeting, Anti-SARS-CoV-2 Therapeutic Candidate

Epigallocatechin 3-gallate (EGCG), a nutritional supplement with promising health-beneficial effects isolated from green tea (Camellia sinensis) (Figure 2a), has long been investigated for its potential as supplementation therapy for the prevention of numerous disorders, including cancer [19] and cardiovascular [20], metabolic [21], neurodegenerative [22], and infectious diseases [23]. For instance, Polyphenon E®, comprising >65% EGCG, is a standardized preparation of green tea catechins approved by the US FDA in 2006 for the treatment of external genital and perianal warts [24]. Polyphenon E has an excellent safety and tolerability profile, an essential characteristic allowing for the extensive use of EGCG [24]. The green tea catechin palmitate (comprising 50% EGCG), an oil-soluble green tea extract, was approved by the US FDA in 2019 as a safe dietary ingredient [25]. In vitro, EGCG has highly promising broad-spectrum antiviral activity, including against Zika virus (half-maximal effective concentration [EC50] = 21.4 μM) [26], hepatitis B virus (half-maximal inhibitory concentration [IC50] = 0.11 μM) [27], Japanese encephalitis virus (IC50 = 7.0 μM) [28], human coronavirus (HCoV) 229E (IC50 = 0.77 μM) [29], human coronavirus OC43 (IC50 = 0.49 μM) [29], Middle East respiratory syndrome (MERS)-CoV (IC50 = 8.4 μM) [30], and SARS-CoV (IC50 = 1.5 μM) [30]. Given its excellent safety and broad-spectrum antiviral activities, EGCG may contribute to immediate clinical solutions for COVID-19 treatment.
Many studies have reported the impressive effects of EGCG against SARS-CoV-2 in vitro (Figure 2b). For example, Hurst et al. [17] demonstrated that EGCG blocks SARS-CoV-2 infection in Vero 76 cells (EC50 = 0.59 µM), while displaying only mild toxicity (selectivity index [SI] = 8.5). The 3C-like protease (3CLpro, also known as Mpro) is highly conserved among coronaviruses, including SARS-CoV-2 [31,32]. Given its essential role in viral replication and transcription, Mpro represents a promising therapeutic target against coronavirus infection [33]. Du et al. [34] showed that EGCG is a potent inhibitor of Mpro, with an IC50 of 0.87 μM. Surface plasmon resonance binding experiments demonstrated that EGCG has a high binding affinity for Mpro, with a dissociation constant (KD) of 6.17 μM. Similarly, Zhu et al. [35] reported that EGCG inhibits Mpro activity, with an IC50 value of 7.51 µM. Furthermore, Ngwe Tun et al. [36] indicated that EGCG is highly effective at inhibiting SARS-CoV-2 replication (IC50 = 6.5 μM) in Vero E6 cells and with minimal toxicity (SI >154). Mechanistically, the authors further demonstrated that EGCG blocks SARS-CoV-2 replication at both the entry and post-entry stages of infection, and also inhibits SARS-CoV-2 Mpro activity.
Figure 2. Epigallocatechin 3-gallate is a green tea-derived, multitargeting, anti-SARS-CoV-2 therapeutic candidate. (a) Epigallocatechin 3-gallate (EGCG), isolated from Camellia sinensis, has potential for development as a therapeutic throat spray for Omicron infection. (b) Important discoveries relating to the multi-target effects of EGCG against SARS-CoV-2. Data from references [17,34,35,37,38,39,40,41,42,43].
Figure 2. Epigallocatechin 3-gallate is a green tea-derived, multitargeting, anti-SARS-CoV-2 therapeutic candidate. (a) Epigallocatechin 3-gallate (EGCG), isolated from Camellia sinensis, has potential for development as a therapeutic throat spray for Omicron infection. (b) Important discoveries relating to the multi-target effects of EGCG against SARS-CoV-2. Data from references [17,34,35,37,38,39,40,41,42,43].
Nutrients 15 03443 g002
Meanwhile, Kato et al. [37] showed that EGCG strongly inhibits the activity of Mpro (IC50 = 0.4 μM) via the formation of a covalent bond between Cys145 of the enzyme and the 2′-position of EGCG (Figure 3). Tsvetkov et al. [38] showed that partial EGCG treatment is highly effective at suppressing viral replication (IC50 = 4.0 μM, SI = 6) by interfering with the binding between ACE2 and SARS-CoV-2 spike (S) protein. Similarly, Henss et al. [39] reported that EGCG inhibits SARS-CoV-2 infection (IC50 = 3.1 µM, SI > 11.6) in Vero E6 cells through binding at the SARS-CoV-2 S–ACE2 interface. SARS-CoV-2 endoribonuclease NendoU (NSP15), a uridine-specific endoribonuclease used by the virus to avoid the innate immune response, is considered a compelling drug target [44]. Hong et al. [40] showed that EGCG strongly inhibits the activity of NSP15, with an IC50 value of 1.6 μM. In the same study, the authors investigated the neutralizing effect of EGCG against SARS-CoV-2 and obtained a promising result (half neutralization effect concentration [PRNT50] = 0.2 μM). The above findings regarding the efficacy of EGCG appear to be generalized, indicative of the therapeutic potential of EGCG for the treatment of COVID-19. Meanwhile, combination drug therapy may offer additional advantages [45]. Rabezanahary et al. [41] revealed that the combination of EGCG (15.6 µM) and remdesivir (1.25 µM), the first FDA-approved inhibitor of SARS-CoV-2 RdRp, exerts a significant synergistic effect (3.1-fold reduction in the EC50 of EGCG for RdRp) in Vero E6 cells through multitargeting activity.
Bettuzzi et al. [42] conducted a 15-day, proof-of-principle study to evaluate the anti-SARS-CoV-2 efficacy of EGCG and catechins (two sessions of inhalation plus three capsules daily; total EGCG: 595 mg; total catechins: 840 mg) in 10 non-hospitalized SARS-CoV-2 swab-positive patients. All patients were asymptomatic within 7 to 15 days of starting treatment, while the levels of inflammation markers significantly decreased. No observable adverse events with the EGCG treatment were reported. Additionally, compared with wild-type or Delta strains, Omicron strains have greater replicative capacity in the upper respiratory tract, increasing the likelihood of viral release during breathing; this characteristic may help explain the enhanced transmission of Omicron strains via airborne routes [46]. Yang et al. [47] demonstrated that after drinking two to three cups of green tea, the levels of EGCG in saliva ranged from 4.8 to 22 µg/mL (equivalent to 8.7–39.9 µM), which was two orders of magnitude higher than those in plasma. For cancer prevention, it is recommended that humans consume six cups of green tea daily [47]; accordingly, high doses of EGCG (up to 79.8 μM in saliva) are likely to be safe and may prove highly effective in controlling Omicron infection. Similarly, Furushima et al. [48] investigated the oral retention of catechins in healthy adults after the intake of a beverage (40 mL) containing 73.4 mg of catechins. They found that the average concentrations of EGCG in the oral cavity were approximately 156.3, 58.4, and 50.5 μM at 10, 40, and 60 min, respectively. These findings support the potential value of EGCG as a supplementation therapy for patients infected with an Omicron variant.
SARS-CoV-2 infection can have long-term effects on the lungs as well as on multiple extrapulmonary tissues and organs, while EGCG exerts unique multi-organ protective effects. For example, EGCG plays an important neuroprotective role following traumatic brain injury (through the activation of the adenosine monophosphate-activated protein kinase pathway), [49] ameliorates liver injury secondary to Pseudomonas aeruginosa pneumonia (via upregulating nuclear receptor activation), [50] protects cardiomyocytes against hypoxia–reperfusion injury (via potently inhibiting the self-cleavage of OMA1), [51] and alleviates SARS-CoV-2-triggered cytokine storm, sepsis, thrombosis, and lung fibrosis [52] (Figure 3). In addition, EGCG decreases the severity of Omicron-related COVID-19 symptoms in both elderly patients and patients with metabolic syndrome by downregulating GRP78 expression or promoting hyperinsulinemia remission [43].
Despite its broad-spectrum antiviral activity, favorable safety profile, and multi-organ protective effects, EGCG demonstrated poor oral bioavailability (F) in both rats (F = 0.1%) and humans (F = 0.3%) [53]. Accordingly, the development of an EGCG throat spray as a potential therapeutic strategy targeting Omicron infection should be further explored in the clinical setting.

3. Myricetin—A Waxberry-Derived Covalent Mpro Inhibitor Suitable for Lead Optimization

Myricetin is a well-known nutritional supplement that can be isolated from “medicine food homology” plants, such as Myrica rubra, Ampelopsis grossedentata, Malus domestica, and Cistus monspeliensis [54]. Specifically, vine tea (A. grossedentata), which has myricetin as the main bioactive ingredient, received approval as a functional food ingredient in 2013 and is traditionally consumed worldwide owing to its health-promoting effects and pleasurable taste [55]. Myricetin, a natural dietary flavonol, has numerous pharmacological effects, including improving bleomycin-induced pulmonary fibrosis via the targeting of HSP90β [56], combating methicillin-resistant Staphylococcus aureus-related lethal pneumonia by inhibiting caseinolytic peptidase P [57], ameliorating brain injury and neurological deficits via nuclear factor erythroid 2-related factor 2 activation [58], enhancing immunomodulatory functions [59], and mitigating hepatic fibrosis via the inhibition of the TREM-1-TLR2/4-MyD88 signaling pathway [60]. Myricetin is also an antiviral drug with low toxicity that can treat a wide variety of viral infections in vitro, including Ebola virus (IC50 = 2.7 μM) [61], Marburg virus (IC50 = 25.5 μM) [62], infectious bronchitis virus (IC50 = 10.6 μM) [63], HIV-1 virus (IC50 = 7.6 μM) [64], African swine fever virus (IC50 = 8.4 μM) [65], Bourbon virus (IC50 = 2.2 μM) [66], and herpes simplex virus (IC50 = 1.6 μM) infections [67].
Myricetin is an ideal candidate for research targeting SARS-CoV-2 infection. SARS-CoV-2 helicase (NSP13), a highly conserved non-structural protein possessing RNA helicase and 5′-triphosphatase activities, is a promising target for the development of novel anti-SARS-CoV-2 drugs [68]. Corona et al. [69] showed that myricetin inhibits NSP13 helicase-associated activity, with an IC50 value of 0.41 µM. Moreover, Xiao et al. [70] reported that myricetin effectively inhibits SARS-CoV-2 replication in vitro by targeting Mpro (IC50 = 3.68 μM; no cytotoxicity was detected with concentrations of up to 50 μM). Further analysis revealed that the 3′- and 4′-hydroxyl groups of myricetin form hydrogen bonds with Phe140 and Glu166 of Mpro, while the chromone ring of myricetin forms π–π stacking interactions with His41, which stabilizes its binding in the catalytic center of Mpro. COVID-19 is primarily an inflammatory disease [71]. In a different study, the same authors [70] revealed that myricetin can effectively inhibit lung inflammation by suppressing inflammatory cell infiltration and the secretion of inflammatory factors (IL-6, IL-1α, TNF-α, and IFN-γ). Similarly, Kato et al. [37] reported that myricetin inhibits the synthesis of SARS-CoV-2 Mpro, with an IC50 value of 0.90 μM, while Kuzikov et al. [72] revealed that myricetin displays excellent anti-SARS-CoV-2 potency in vitro (IC50 = 0.22 μM). The authors [72] further reported the X-ray crystal structure of Mpro complexed with myricetin at a resolution of 1.77 Å (PDB ID: 7B3E), which unambiguously revealed that the mechanism of action involves the formation of a covalent bond between Cys145 and the 2′ position of myricetin.
Although myricetin has broad-spectrum antiviral potential without serious adverse effects, its use is limited due to its poor solubility and low oral bioavailability (<10%) [73,74]. Consequently, the development of myricetin prodrugs or oral derivatives, with enhanced bioavailability and membrane permeability, has been proposed as an alternative tactic for drug design (Figure 4). The pyrogallol group of the myricetin B ring, acting as an electrophile, is covalently bound to Cys145, helping to maintain a strong anti-SARS-CoV-2 potential [18]. Chaves et al. [75] evaluated a series of structurally similar natural flavonoids, including myricetin, and found that the presence of fewer hydroxyl groups in ring B of these flavonols (myricetin, three hydroxyl groups; quercetin, two hydroxyl groups; and kaempferol, one hydroxyl group) was correlated with reduced anti-SARS-CoV-2 activity in Calu-3 cells, with EC50 values of approximately 0.91, 2.40, and 3.02 μM, respectively. Notably, when the pyrogallol group of the myricetin B ring was transferred to the A ring, the binding mode of myricetin to Mpro was fundamentally changed.
Myricetin and another Mpro inhibitor, baicalein, possess pyrogallol groups, but their modes of action and their structural determinants of protease binding are different [18]. An examination of the crystal structure of the myricetin-Mpro complex (2.1 Å, PDB ID: 7DPP) revealed the presence of a covalent bond between the sulfur atom of Cys145 of Mpro and the C6′ atom of the pyrogallol group of myricetin (Figure 4) [18]. Several other interactions were identified, such as hydrogen bonding between the hydroxyl groups of myricetin and Gly143, Ser144, Cys145, and Thr26; π–π stacking interactions between the chromone moiety of myricetin and His41; and the formation of hydrogen bonds between the chromone moiety and Glu189, His164, His41, and Asp187 of Mpro. In contrast, observation of the crystal structure of baicalein complexed with Mpro (2.2 Å, PDB ID: 6M2N) revealed that baicalein forms multiple interactions (π–π stacking, hydrogen bonds, and hydrophobic interactions) with specific residues of Mpro, rather than covalently blocking the catalytic Cys145 residue (Figure 4) [18].
In terms of structure, the pyrogallol group of the B rings of flavanols can be easily oxidized to form orthoquinone, which covalently binds to Cys145 of Mpro (Figure 5) [73]. These studies present a starting point for structure-based lead identification and optimization of flavanol-based compounds.
Furthermore, Xiong et al. [76] found that myricetin (IC50 = 1.2 μM) and its glycoside myricitrin (IC50 = 14.2 μM) can inhibit SARS-CoV-2 replication by covalently binding to the biothiols of Mpro in a dose- and time-dependent manner. Nevertheless, the antiviral activity of myricitrin (IC50 = 14.2 μM) is 11.8-fold weaker than that of myricetin (IC50 = 1.2 μM), which demonstrates the pivotal role of the free C3 hydroxyl group in promoting the binding of myricetin to Mpro through hydrogen bonding. Besides that, dihydromyricetin, a trans-conformation of myricetin (hydrogenation of its C2=C3 double bond), displays weaker inhibitory activity compared to that of myricetin (IC50: 1.14 vs. 0.63 μM; EC50: 13.56 vs. 8.00 μM) [18]. This is likely because the presence of the C2=C3 bond increases electron delocalization in the A–C ring of myricetin and enhances the stability of π-conjugation with His41. Together, these results demonstrate that the 3-OH and 3′,4′,5′-OH moieties of myricetin are essential for its potent anti-SARS-CoV-2 activity (Figure 6) [18].
Lipophilicity (cLogP) is an important physicochemical parameter influencing oral absorption and pharmacokinetic properties [77,78]. Su et al. [18] found that the addition of an alkyl group (methyl, ethyl, isoamyl, or cyclopentylmethyl) to the 7-OH of myricetin can increase the lipophilicity of the resulting compound relative to that of myricetin, thereby enhancing the inhibition of the enzymatic activity of SARS-CoV-2 Mpro. An analysis of the structure–activity relationship of the derived compounds suggested that the smaller methyl group may bind more strongly to a specific but small sub-pocket within Mpro compared to the other, larger alkyl groups. Subsequently, a methyl group was introduced to the 7-OH of dihydromyricetin, yielding compound 7, which could significantly inhibit viral replication (IC50, 0.26 μM; EC50, 11.5 μM) (Figure 4) [18]. When administered orally to mice (30 mg/kg compound 7 per day), this compound showed an improved pharmacokinetic profile compared to that of myricetin (Tmax, 1.74 vs. 0.50 h; Cmax, 724 vs. 8.59 ng/mL; AUClast, 510 vs. 6.07 ng·h/mL; MRT, 1.89 vs. 0.84 h; and T1/2, 1.74 vs. 0.44 h for compound 7 and myricetin, respectively). Compound 7 displayed acceptable oral bioavailability (F = 18.1%), given that compounds with an oral bioavailability greater than 10% have potential for development as oral drugs [18]. The current data support the further optimization of 7-O-methylmyricetin-based oral inhibitors for COVID-19 treatment. Prodrugs have better pharmacokinetic properties, and their hidden pharmacological activities can be recovered after biotransformation in vivo, thereby representing an excellent option for the design of COVID-19-targeting drugs [79,80]. To improve the aqueous solubility and the membrane permeability of myricetin, compound 9 was produced via the introduction of diphenyl phosphate at the 7-OH moiety of myricetin (Figure 4) [18]. Compared to myricetin, compound 9 displayed significantly increased lipophilicity (cLogP, 3.89 vs. 0.84) and better inhibitory activity against SARS-CoV-2 replication (EC50, 3.15 vs. 8.00 μM). Similarly, the same diphenyl phosphate group was added to the 7-OH of dihydromyricetin, yielding compound 10, with an EC50 against SARS-CoV-2 replication superior to that of dihydromyricetin (EC50, 9.03 vs. 13.6 μM) [18]. Myricetin prodrugs are still experimental, and further studies are needed to demonstrate their safety and efficacy.

4. Other Anti-SARS-CoV-2 Natural Dietary Flavonoids in Development for Treating SARS-CoV-2 Infection

Natural dietary flavonoids can make a substantial contribution to mitigating the effects of the COVID-19 pandemic given their good safety profile and antiviral activities. In addition to the abovementioned bioactive compounds, other natural dietary flavonoids, shown in Table 1, have demonstrated strong anti-SARS-CoV-2 activity in vitro, and thus can serve as a starting point for further drug development for the treatment of COVID-19.

5. Conclusions and Future Directions

The COVID-19 pandemic represented an unprecedented global health crisis. Functional foods and nutritional supplements are excellent complements to vaccines and therapeutics. They encompass a large and rich library of natural bioactive products, some of which are likely to exhibit anti-COVID-19 therapeutic potential. Natural dietary flavonoids are conceptually attractive as treatment options in response to outbreaks; however, their implementation is challenging. To obtain maximal benefits, several aspects should be considered to facilitate the development of natural dietary-bioactive-product-based drugs.
First, additional research directly related to SARS-CoV-2 and its variants is urgently needed to clarify the effectiveness of the above-mentioned flavonoids. Natural dietary bioactive flavonoids must be rigorously evaluated in in vitro, animal model, and clinical studies rather than relying only on virtual screening, network pharmacological prediction, or machine learning models, especially when their targets or mechanisms of action are unknown. For instance, designing controlled clinical trials may help elucidate any additional effects of these compounds. Natural dietary flavonoids may make a marked contribution toward controlling hyperinflammatory responses and preventing lung injury. Nevertheless, the underlying mechanisms require further exploration and systematic clarification.
Secondly, combination therapy could contribute to addressing potential drug resistance associated with emerging variants. Evolution and the associated increase in selection pressures may yield SARS-CoV-2 variants that are resistant to antiviral therapies. While resistant viruses could rapidly emerge in the presence of a single agent, the appearance of escape mutations against a combination of compounds, displaying different mechanisms of action and resistance profiles, is less likely. Multi-target therapeutic modalities (mixtures of natural dietary flavonoids, similar to drug cocktails) with broad variant activity could further improve the effectiveness against globally circulating SARS-CoV-2 variants and mitigate the emergence of new escape mutants. Importantly, however, monitoring for potential drug–drug interactions is essential in the development of combination therapies. In parallel, lead optimization of multi-target dietary compounds (e.g., EGCG targeting SARS-CoV-2 NSP15, S protein, and Mpro) could potentially provide candidates for COVID-19 treatment.
Thirdly, alternative routes of administration (e.g., oral or inhalation) have the potential to maximize clinical benefit. Compared to the intravenous route, oral or inhalation administration can help address compliance issues given that the associated medications are less likely to require refrigeration, while also avoiding the use of needles. Oral administration can maximize clinical benefits by shortening the duration of COVID-19 and reducing acute post-sequelae symptoms of SARS-CoV-2 infection. Inhalation administration facilitates the direct delivery of antiviral agents to the primary site of infection, reducing systemic exposure to drugs and their metabolites, thereby minimizing systemic side effects.
In summary, to address current limitations associated with the use of dietary components for the treatment of SARS-CoV-2 infection, a mixture of factors, including combination therapy, lead optimization, and drug delivery, must be considered.

Author Contributions

Z.W.: conceptualization, data curation, writing original draft, writing—review and editing, visualization, and funding acquisition. L.Y.: conceptualization, writing—review and editing, and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Shandong Provincial Natural Science Foundation (ZR2022MH162, ZR2022QE202), and the PhD Research Start-up Foundation of Qufu Normal University (614901, 615201).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data were used for the research described in the article.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Petersen, E.; Koopmans, M.; Go, U.; Hamer, D.H.; Petrosillo, N.; Castelli, F.; Storgaard, M.; Khalili, S.A.; Simonsen, L. Comparing SARS-CoV-2 with SARS-CoV and influenza pandemics. Lancet Infect. Dis. 2020, 20, e238–e244. [Google Scholar] [CrossRef] [PubMed]
  2. Toussi, S.S.; Hammond, J.L.; Gerstenberger, B.S.; Anderson, A.S. Therapeutics for COVID-19. Nat. Microbiol. 2023, 8, 771–786. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, Z.; Wang, N.; Yang, L.; Song, X.Q. Bioactive natural products in COVID-19 therapy. Front. Pharmacol. 2022, 13, 926507. [Google Scholar] [CrossRef] [PubMed]
  4. Wahl, A.; Gralinski, L.E.; Johnson, C.E.; Yao, W.; Kovarova, M.; Dinnon, K.H., III; Liu, H.; Madden, V.J.; Krzystek, H.M.; De, C.; et al. SARS-CoV-2 infection is effectively treated and prevented by EIDD-2801. Nature 2021, 591, 451–457. [Google Scholar] [CrossRef]
  5. Yang, L.; Wang, Z. Bench-to-bedside: Innovation of small molecule anti-SARS-CoV-2 drugs in China. Eur. J. Med. Chem. 2023, 257, 115503. [Google Scholar] [CrossRef]
  6. Levin, M.J.; Ustianowski, A.; De Wit, S.; Launay, O.; Avila, M.; Templeton, A.; Yuan, Y.; Seegobin, S.; Ellery, A.; Levinson, D.J.; et al. Intramuscular AZD7442 (tixagevimab–cilgavimab) for prevention of COVID-19. N. Engl. J. Med. 2022, 386, 2188–2200. [Google Scholar] [CrossRef]
  7. Connors, M.; Graham, B.S.; Lane, H.C.; Fauci, A.S. SARS-CoV-2 vaccines: Much accomplished, much to learn. Ann. Intern. Med. 2021, 174, 687–690. [Google Scholar] [CrossRef]
  8. Wang, Z.; Yang, L.; Song, X.Q. Oral GS-441524 derivatives: Next-generation inhibitors of SARS-CoV-2 RNA-dependent RNA polymerase. Front. Immunol. 2022, 13, 1015355. [Google Scholar] [CrossRef]
  9. Bushman, M.; Kahn, R.; Taylor, B.P.; Lipsitch, M.; Hanage, W.P. Population impact of SARS-CoV-2 variants with enhanced transmissibility and/or partial immune escape. Cell 2021, 184, 6229–6242. [Google Scholar] [CrossRef]
  10. Carabelli, A.M.; Peacock, T.P.; Thorne, L.G.; Harvey, W.T.; Hughes, J.; Peacock, S.J.; Barclay, W.S.; de Silva, T.; Towers, G.J.; Robertson, D.L. SARS-CoV-2 variant biology: Immune escape, transmission and fitness. Nat. Rev. Microbiol. 2023, 21, 162–177. [Google Scholar] [CrossRef]
  11. Uriu, K.; Ito, J.; Zahradnik, J.; Fujita, S.; Kosugi, Y.; Schreiber, G.; The Genotype to Phenotype Japan (G2P-Japan) Consortium; Sato, K. Enhanced transmissibility, infectivity and immune resistance of the SARS-CoV-2 Omicron XBB. 1.5 variant. Lancet Infect. Dis. 2023, 23, 280–281. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, Z.; Yang, L. Post-acute sequelae of SARS-CoV-2 infection: A neglected public health issue. Front. Public Health 2022, 10, 908757. [Google Scholar] [CrossRef] [PubMed]
  13. Davis, H.E.; McCorkell, L.; Vogel, J.M.; Topol, E.J. Long COVID: Major findings, mechanisms and recommendations. Nat. Rev. Microbiol. 2023, 21, 133–146. [Google Scholar]
  14. Wang, Z.; Yang, L. Turning the tide: Natural products and natural-product-inspired chemicals as potential counters to SARS-CoV-2 infection. Front. Pharmacol. 2020, 11, 1013. [Google Scholar] [CrossRef]
  15. Wang, Z.; Yang, L. Chinese herbal medicine: Fighting SARS-CoV-2 infection on all fronts. J. Ethnopharmacol. 2021, 270, 113869. [Google Scholar]
  16. Banerjee, A.; Somasundaram, I.; Das, D.; Jain Manoj, S.; Banu, H.; Mitta Suresh, P.; Paul, S.; Bisgin, A.; Zhang, H.; Sun, X.F.; et al. Functional Foods: A promising strategy for restoring gut microbiota diversity impacted by SARS-CoV-2 variants. Nutrients 2023, 15, 2631. [Google Scholar] [CrossRef] [PubMed]
  17. Hurst, B.L.; Dickinson, D.; Hsu, S. Epigallocatechin-3-gallate (EGCG) inhibits SARS-CoV-2 infection in primate epithelial cells: (A short communication). Microbiol. Infect. Dis. 2021, 5, 1–6. [Google Scholar] [CrossRef]
  18. Su, H.; Yao, S.; Zhao, W.; Zhang, Y.; Liu, J.; Shao, Q.; Wang, Q.; Li, M.; Xie, H.; Shang, W.; et al. Identification of pyrogallol as a warhead in design of covalent inhibitors for the SARS-CoV-2 3CL protease. Nat. Commun. 2021, 12, 3623. [Google Scholar] [CrossRef]
  19. Wang, L.; Li, P.; Feng, K. EGCG adjuvant chemotherapy: Current status and future perspectives. Eur. J. Med. Chem. 2023, 250, 115197. [Google Scholar] [CrossRef]
  20. Cai, Y.; Yu, S.S.; He, Y.; Bi, X.Y.; Gao, S.; Yan, T.D.; Zheng, G.D.; Chen, T.T.; Ye, J.T.; Liu, P.Q. EGCG inhibits pressure overload-induced cardiac hypertrophy via the PSMB5/Nmnat2/SIRT6-dependent signalling pathways. Acta Physiol. 2021, 231, e13602. [Google Scholar] [CrossRef]
  21. Wen, J.J.; Li, M.Z.; Chen, C.H.; Hong, T.; Yang, J.R.; Huang, X.J.; Geng, F.; Hu, J.L.; Nie, S.P. Tea polyphenol and epigallocatechin gallate ameliorate hyperlipidemia via regulating liver metabolism and remodeling gut microbiota. Food Chem. 2023, 404, 134591. [Google Scholar] [CrossRef]
  22. Kuo, Y.C.; Wang, I.H.; Rajesh, R. Use of leptin-conjugated phosphatidic acid liposomes with resveratrol and epigallocatechin gallate to protect dopaminergic neurons against apoptosis for Parkinson’s disease therapy. Acta Biomater. 2021, 119, 360–374. [Google Scholar] [CrossRef] [PubMed]
  23. Cheong, Y.; Kim, M.; Ahn, J.; Oh, H.; Lim, J.; Chae, W.; Yang, S.W.; Kim, M.S.; Yu, J.E.; Byun, S.; et al. Epigallocatechin-3-gallate as a novel vaccine adjuvant. Front. Immunol. 2021, 12, 4803. [Google Scholar] [CrossRef]
  24. Hara, Y. Tea catechins and their applications as supplements and pharmaceutics. Pharmacol. Res. 2011, 64, 100–104. [Google Scholar] [CrossRef]
  25. Menegazzi, M.; Campagnari, R.; Bertoldi, M.; Crupi, R.; Di Paola, R.; Cuzzocrea, S. Protective effect of epigallocatechin-3-gallate (EGCG) in diseases with uncontrolled immune activation: Could such a scenario be helpful to counteract COVID-19? Int. J. Mol. Sci. 2020, 21, 5171. [Google Scholar] [CrossRef] [PubMed]
  26. Carneiro, B.M.; Batista, M.N.; Braga, A.C.S.; Nogueira, M.L.; Rahal, P. The green tea molecule EGCG inhibits Zika virus entry. Virology 2016, 496, 215–218. [Google Scholar] [CrossRef] [PubMed]
  27. Pang, J.Y.; Zhao, K.J.; Wang, J.B.; Ma, Z.J.; Xiao, X.H. Green tea polyphenol, epigallocatechin-3-gallate, possesses the antiviral activity necessary to fight against the hepatitis B virus replication in vitro. J. Zhejiang Univ.-Sci. B 2014, 15, 533–539. [Google Scholar] [CrossRef] [Green Version]
  28. Wang, C.Y.; Hour, M.J.; Lai, H.C.; Chen, C.H.; Chang, P.J.; Huang, S.H.; Lin, C.W. Epigallocatechin-3-gallate inhibits the early stages of Japanese encephalitis virus infection. Virus Res. 2018, 253, 140–146. [Google Scholar] [CrossRef]
  29. LeBlanc, E.V.; Colpitts, C.C. The green tea catechin EGCG provides proof-of-concept for a pan-coronavirus attachment inhibitor. Sci. Rep. 2022, 12, 12899. [Google Scholar] [CrossRef]
  30. Dinda, B.; Dinda, S.; Dinda, M. Therapeutic potential of green tea catechin, (-)-epigallocatechin-3-O-gallate (EGCG) in SARS-CoV-2 infection: Major interactions with host/virus proteases. Phytomed. Plus 2022, 3, 100402. [Google Scholar]
  31. Wang, Z.; Yang, L.; Zhao, X.E. Co-crystallization and structure determination: An effective direction for anti-SARS-CoV-2 drug discovery. Comput. Struct. Biotechnol. J. 2021, 19, 4684–4701. [Google Scholar]
  32. Brier, L.; Hassan, H.; Hanoulle, X.; Landry, V.; Moschidi, D.; Desmarets, L.; Rouillé, Y.; Dumont, J.; Herledan, A.; Warenghem, S.; et al. Novel dithiocarbamates selectively inhibit 3CL protease of SARS-CoV-2 and other coronaviruses. Eur. J. Med. Chem. 2023, 250, 115186. [Google Scholar] [CrossRef]
  33. Pang, X.; Xu, W.; Liu, Y.; Li, H.; Chen, L. The research progress of SARS-CoV-2 main protease inhibitors from 2020 to 2022. Eur. J. Med. Chem. 2023, 257, 115491. [Google Scholar] [CrossRef] [PubMed]
  34. Du, A.; Zheng, R.; Disoma, C.; Li, S.; Chen, Z.; Li, S.; Liu, P.; Zhou, Y.; Shen, Y.; Liu, S.; et al. Epigallocatechin-3-gallate, an active ingredient of Traditional Chinese Medicines, inhibits the 3CLpro activity of SARS-CoV-2. Int. J. Biol. Macromol. 2021, 176, 1–12. [Google Scholar] [CrossRef] [PubMed]
  35. 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, 1884. [Google Scholar] [CrossRef]
  36. Ngwe Tun, M.M.; Luvai, E.; Nwe, K.M.; Toume, K.; Mizukami, S.; Hirayama, K.; Komatsu, 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] [PubMed]
  37. Kato, Y.; Higashiyama, A.; Takaoka, E.; Nishikawa, M.; Ikushiro, S. Food phytochemicals, epigallocatechin gallate and myricetin, covalently bind to the active site of the coronavirus main protease in vitro. Adv. Redox Res. 2021, 3, 100021. [Google Scholar] [CrossRef] [PubMed]
  38. Tsvetkov, V.; Varizhuk, A.; Kozlovskaya, L.; Shtro, A.; Lebedeva, O.; Komissarov, A.; Vedekhina, T.; Manuvera, V.; Zubkova, O.; Eremeev, A.; et al. EGCG as an anti-SARS-CoV-2 agent: Preventive versus therapeutic potential against original and mutant virus. Biochimie 2021, 191, 27–32. [Google Scholar] [CrossRef]
  39. Henss, L.; Auste, A.; Schürmann, C.; Schmidt, C.; von Rhein, C.; Mühlebach, M.D.; Schnierle, B.S. The green tea catechin epigallocatechin gallate inhibits SARS-CoV-2 infection. J. Gen. Virol. 2021, 102, 1574. [Google Scholar] [CrossRef]
  40. Hong, S.; Seo, S.H.; Woo, S.J.; Kwon, Y.; Song, M.; Ha, N.C. Epigallocatechin gallate inhibits the uridylate-specific endoribonuclease Nsp15 and efficiently neutralizes the SARS-CoV-2 strain. J. Agric. Food Chem. 2021, 69, 5948–5954. [Google Scholar] [CrossRef]
  41. Rabezanahary, H.; Badr, A.; Checkmahomed, L.; Pageau, K.; Desjardins, Y.; Baz, M. Epigallocatechin Gallate and Isoquercetin synergize with Remdesivir to reduce SARS-CoV-2 replication in vitro. Front. Virol. 2022, 2, 62. [Google Scholar] [CrossRef]
  42. Bettuzzi, S.; Gabba, L.; Cataldo, S. Efficacy of a polyphenolic, standardized green tea extract for the treatment of COVID-19 syndrome: A proof-of-principle study. COVID 2021, 1, 2–12. [Google Scholar] [CrossRef]
  43. Zhang, Z.; Hao, M.; Zhang, X.; He, Y.; Chen, X.; Taylor, E.W.; Zhang, J. Potential of green tea EGCG in neutralizing SARS-CoV-2 Omicron variant with greater tropism toward the upper respiratory tract. Trends Food Sci. Technol. 2023, 132, 40–53. [Google Scholar] [CrossRef] [PubMed]
  44. Pillon, M.C.; Frazier, M.N.; Dillard, L.B.; Williams, J.G.; Kocaman, S.; Krahn, J.M.; Perera, L.; Hayne, C.K.; Gordon, J.; Stewart, Z.D.; et al. Cryo-EM structures of the SARS-CoV-2 endoribonuclease Nsp15 reveal insight into nuclease specificity and dynamics. Nat. Commun. 2021, 12, 636. [Google Scholar] [CrossRef]
  45. Yang, L.; Wang, Z. Natural products, alone or in combination with FDA-approved drugs, to treat COVID-19 and lung cancer. Biomedicines 2021, 9, 689. [Google Scholar] [CrossRef]
  46. Tian, D.; Sun, Y.; Xu, H.; Ye, Q. The emergence and epidemic characteristics of the highly mutated SARS-CoV-2 Omicron variant. J. Med. Virol. 2022, 94, 2376–2383. [Google Scholar] [CrossRef]
  47. Yang, C.S.; Lee, M.J.; Chen, L. Human salivary tea catechin levels and catechin esterase activities: Implication in human cancer prevention studies. Cancer Epidem. Biomar. Prev. 1999, 8, 83–89. [Google Scholar]
  48. Furushima, D.; Otake, Y.; Koike, N.; Onishi, S.; Mori, T.; Ota, N.; Yamada, H. Investigation of the oral retention of tea catechins in humans: An exploratory interventional study. Nutrients 2021, 13, 3024. [Google Scholar] [CrossRef]
  49. Wu, Y.; Cui, J. (−)-Epigallocatechin-3-gallate provides neuroprotection via AMPK activation against traumatic brain injury in a mouse model. N-S Arch. Pharmacol. 2020, 393, 2209–2220. [Google Scholar] [CrossRef]
  50. Wang, Y.; Jian, S.; Li, W.; Zhao, L.; Ye, G.; Shi, F.; Li, L.; Zou, Y.; Song, X.; Zhao, X.; et al. Epigallocatechin-3-gallate ameliorates liver injury secondary to Pseudomonas aeruginosa pneumonia. Int. Immunopharmacol. 2022, 112, 109239. [Google Scholar] [CrossRef]
  51. Nan, J.; Nan, C.; Ye, J.; Qian, L.; Geng, Y.; Xing, D.; Rahman, M.S.U.; Huang, M. EGCG protects cardiomyocytes against hypoxia-reperfusion injury through inhibition of OMA1 activation. J. Cell Sci. 2019, 132, jcs220871. [Google Scholar]
  52. Zhang, Z.; Zhang, X.; Bi, K.; He, Y.; Yan, W.; Yang, C.S.; Zhang, J. Potential protective mechanisms of green tea polyphenol EGCG against COVID-19. Trends Food Sci. Technol. 2021, 114, 11–24. [Google Scholar] [CrossRef] [PubMed]
  53. Pervin, M.; Unno, K.; Takagaki, A.; Isemura, M.; Nakamura, Y. Function of green tea catechins in the brain: Epigallocatechin gallate and its metabolites. Int. J. Mol. Sci. 2019, 20, 3630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zhang, D.; Jiang, X.; Xiao, L.; Lu, Y.; Sang, S.; Lv, L.; Dong, W. Mechanistic studies of inhibition on acrolein by myricetin. Food Chem. 2020, 323, 126788. [Google Scholar] [CrossRef] [PubMed]
  55. Carneiro, R.C.; Ye, L.; Baek, N.; Teixeira, G.H.; O’Keefe, S.F. Vine tea (Ampelopsis grossedentata): A review of chemical composition, functional properties, and potential food applications. J. Funct. Foods 2021, 76, 104317. [Google Scholar] [CrossRef]
  56. Li, X.; Yu, H.; Liang, L.; Bi, Z.; Wang, Y.; Gao, S.; Wang, M.; Li, H.; Miao, Y.; Deng, R.; et al. Myricetin ameliorates bleomycin-induced pulmonary fibrosis in mice by inhibiting TGF-β signaling via targeting HSP90β. Biochem. Pharmacol. 2020, 178, 114097. [Google Scholar] [CrossRef] [PubMed]
  57. Jing, S.; Wang, L.; Wang, T.; Fan, L.; Chen, L.; Xiang, H.; Shi, Y.; Wang, D. Myricetin protects mice against MRSA-related lethal pneumonia by targeting ClpP. Biochem. Pharmacol. 2021, 192, 114753. [Google Scholar] [CrossRef]
  58. Wu, S.; Yue, Y.; Peng, A.; Zhang, L.; Xiang, J.; Cao, X.; Ding, H.; Yin, S. Myricetin ameliorates brain injury and neurological deficits via Nrf2 activation after experimental stroke in middle-aged rats. Food Funct. 2016, 7, 2624–2634. [Google Scholar] [CrossRef]
  59. Bhat RA, H.; Rehman, S.; Tandel, R.S.; Dash, P.; Bhandari, A.; Ganie, P.A.; Shah, T.K.; Pant, K.; Yousuf, D.J.; Bhat, I.A.; et al. Immunomodulatory and antimicrobial potential of ethanolic extract of Himalayan Myrica esculanta in Oncorhynchus mykiss: Molecular modelling with Aeromonas hydrophila functional proteins. Aquaculture 2021, 533, 736213. [Google Scholar] [CrossRef]
  60. Yao, Q.; Li, S.; Li, X.; Wang, F.; Tu, C. Myricetin modulates macrophage polarization and mitigates liver inflammation and fibrosis in a murine model of nonalcoholic steatohepatitis. Front. Med. 2020, 7, 71. [Google Scholar] [CrossRef] [Green Version]
  61. Daino, G.L.; Frau, A.; Sanna, C.; Rigano, D.; Distinto, S.; Madau, V.; Esposito, F.; Fanunza, E.; Bianco, G.; Taglialatela-Scafati, O.; et al. Identification of myricetin as an ebola virus VP35–double-stranded RNA interaction inhibitor through a novel fluorescence-based assay. Biochemistry 2018, 57, 6367–6378. [Google Scholar] [CrossRef] [Green Version]
  62. Zinzula, L.; Mereu, A.M.; Orsini, M.; Seeleitner, C.; Bracher, A.; Nagy, I.; Baumeister, W. Ebola and Marburg virus VP35 coiled-coil validated as antiviral target by tripartite split-GFP complementation. iScience 2022, 25, 105354. [Google Scholar] [CrossRef] [PubMed]
  63. Peng, S.; Fang, C.; He, H.; Song, X.; Zhao, X.; Zou, Y.; Li, L.; Jia, R.; Yin, Z. Myricetin exerts its antiviral activity against infectious bronchitis virus by inhibiting the deubiquitinating activity of papain-like protease. Poultry Sci. 2022, 101, 101626. [Google Scholar] [CrossRef]
  64. Ortega, J.T.; Suárez, A.I.; Serrano, M.L.; Baptista, J.; Pujol, F.H.; Rangel, H.R. The role of the glycosyl moiety of myricetin derivatives in anti-HIV-1 activity in vitro. AIDS Res. Ther. 2017, 14, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Jo, S.; Kim, S.; Shin, D.H.; Kim, M.S. Inhibition of African swine fever virus protease by myricetin and myricitrin. J. Enzym. Inhib. Med. Chem. 2020, 35, 1045–1049. [Google Scholar] [CrossRef] [PubMed]
  66. Hao, S.; Ning, K.; Wang, X.; Wang, J.; Cheng, F.; Ganaie, S.S.; Tavis, J.E.; Qiu, J. Establishment of a replicon reporter of the emerging tick-borne bourbon virus and use it for evaluation of antivirals. Front. Microbiol. 2020, 11, 572631. [Google Scholar] [CrossRef]
  67. Li, W.; Xu, C.; Hao, C.; Zhang, Y.; Wang, Z.; Wang, S.; Wang, W. Inhibition of herpes simplex virus by myricetin through targeting viral gD protein and cellular EGFR/PI3K/Akt pathway. Antivir. Res. 2020, 177, 104714. [Google Scholar] [CrossRef] [PubMed]
  68. V’kovski, P.; Kratzel, A.; Steiner, S.; Stalder, H.; Thiel, V. Coronavirus biology and replication: Implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021, 19, 155–170. [Google Scholar] [CrossRef]
  69. Corona, A.; Wycisk, K.; Talarico, C.; Manelfi, C.; Milia, J.; Cannalire, R.; Esposito, F.; Gribbon, P.; Zaliani, A.; Iaconis, D.; et al. Natural compounds inhibit SARS-CoV-2 nsp13 unwinding and ATPase enzyme activities. ACS Pharmacol. Transl. Sci. 2022, 5, 226–239. [Google Scholar] [CrossRef]
  70. Xiao, T.; Cui, M.; Zheng, C.; Wang, M.; Sun, R.; Gao, D.; Bao, J.; Ren, S.; Yang, B.; Lin, J.; et al. Myricetin inhibits SARS-CoV-2 viral replication by targeting Mpro and ameliorates pulmonary inflammation. Front. Pharmacol. 2021, 12, 669642. [Google Scholar] [CrossRef]
  71. Yeoh, Y.K.; Zuo, T.; Lui, G.C.Y.; Zhang, F.; Liu, Q.; Li, A.Y.; Chung, A.C.; Cheung, C.P.; Tso, E.Y.; Fung, K.S.; et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698–706. [Google Scholar] [CrossRef] [PubMed]
  72. Kuzikov, M.; Costanzi, E.; Reinshagen, J.; Esposito, F.; Vangeel, L.; Wolf, M.; Ellinger, B.; Claussen, C.; Geisslinger, G.; Corona, A.; et al. Identification of inhibitors of SARS-CoV-2 3CL-pro enzymatic activity using a small molecule in vitro repurposing screen. ACS Pharmacol. Transl. Sci. 2021, 4, 1096–1110. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, M.; Hong, C.; Yao, Y.; Shen, H.; Ji, G.; Li, G.; Xie, Y. Development of a pharmaceutical cocrystal with solution crystallization technology: Preparation, characterization, and evaluation of myricetin-proline cocrystals. Eur. J. Pharm. Biopharm. 2016, 107, 151–159. [Google Scholar] [CrossRef] [PubMed]
  74. Qian, J.; Meng, H.; Xin, L.; Xia, M.; Shen, H.; Li, G.; Xie, Y. Self-nanoemulsifying drug delivery systems of myricetin: Formulation development, characterization, and in vitro and in vivo evaluation. Colloid. Surface. B 2017, 160, 101–109. [Google Scholar] [CrossRef] [PubMed]
  75. Chaves, O.A.; Fintelman-Rodrigues, N.; Wang, X.; Sacramento, C.Q.; Temerozo, J.R.; Ferreira, A.C.; Mattos, M.; Pereira-Dutra, F.; Bozza, P.T.; Castro-Faria-Neto, H.C.; et al. Commercially available flavonols are better SARS-CoV-2 inhibitors than isoflavone and flavones. Viruses 2022, 14, 1458. [Google Scholar] [CrossRef]
  76. Xiong, Y.; Zhu, G.H.; Zhang, Y.N.; Hu, Q.; Wang, H.N.; Yu, H.N.; Qin, X.Y.; Guan, X.Q.; Xiang, Y.W.; Tang, H.; et al. Flavonoids in Ampelopsis grossedentata as covalent inhibitors of SARS-CoV-2 3CLpro: Inhibition potentials, covalent binding sites and inhibitory mechanisms. Int. J. Biol. Macromol. 2021, 187, 976–987. [Google Scholar] [CrossRef]
  77. Morak-Młodawska, B.; Jeleń, M.; Martula, E.; Korlacki, R. Study of lipophilicity and ADME properties of 1,9-diazaphenothiazines with anticancer action. Int. J. Mol. Sci. 2023, 24, 6970. [Google Scholar] [CrossRef]
  78. Johnson, T.W.; Gallego, R.A.; Edwards, M.P. Lipophilic efficiency as an important metric in drug design. J. Med. Chem. 2018, 61, 6401–6420. [Google Scholar] [CrossRef]
  79. Wang, Z.; Yang, L. Broad-spectrum prodrugs with anti-SARS-CoV-2 activities: Strategies, benefits, and challenges. J. Med. Virol. 2022, 94, 1373–1390. [Google Scholar] [CrossRef]
  80. Walther, R.; Rautio, J.; Zelikin, A.N. Prodrugs in medicinal chemistry and enzyme prodrug therapies. Adv. Drug Deliv. Rev. 2017, 118, 65–77. [Google Scholar] [CrossRef]
  81. Huang, Y.; Zhou, W.; Sun, J.; Ou, G.; Zhong, N.S.; Liu, Z. Exploring the potential pharmacological mechanism of hesperidin and glucosyl hesperidin against COVID-19 based on bioinformatics analyses and antiviral assays. Am. J. Chin. Med. 2022, 50, 351–369. [Google Scholar] [CrossRef] [PubMed]
  82. Bellavite, P.; Donzelli, A. Hesperidin and SARS-CoV-2: New light on the healthy function of citrus fruits. Antioxidants 2020, 9, 742. [Google Scholar] [CrossRef] [PubMed]
  83. Chiou, W.C.; Lu, H.F.; Hsu, N.Y.; Chang, T.Y.; Chin, Y.F.; Liu, P.C.; Lo, J.M.; Wu, Y.B.; Yang, J.M.; Huang, C. Ugonin J acts as a SARS-CoV-2 3C-like protease inhibitor and exhibits anti-inflammatory properties. Front. Pharmacol. 2021, 12, 720018. [Google Scholar] [CrossRef]
  84. Klein, C.F.; Petek, B.J.; Moulson, N.; Baggish, A.L.; Churchill, T.W.; Harmon, K.G.; Kliethermes, S.A.; Patel, M.R.; Drezner, J.A. Non-COVID-19 cardiovascular pathology from return-to-play screening in college athletes after COVID-19. Heart, 2023; in press. [Google Scholar] [CrossRef]
  85. Mahmud, S.; Biswas, S.; Paul, G.K.; Mita, M.A.; Promi, M.M.; Afrose, S.; Afrose, S.; Hasan, M.R.; Zaman, S.; Uddin, M.S.; et al. Plant-based phytochemical screening by targeting main protease of SARS-CoV-2 to design effective potent inhibitors. Biology 2021, 10, 589. [Google Scholar] [CrossRef] [PubMed]
  86. Gogoi, M.; Borkotoky, M.; Borchetia, S.; Chowdhury, P.; Mahanta, S.; Barooah, A.K. Black tea bioactives as inhibitors of multiple targets of SARS-CoV-2 (3CLpro, PLpro and RdRp): A virtual screening and molecular dynamic simulation study. J. Biomol. Struct. Dyn. 2022, 40, 7143–7166. [Google Scholar] [CrossRef] [PubMed]
  87. Jeon, S.; Ko, M.; Lee, J.; Choi, I.; Byun, S.Y.; Park, S.; Shum, D.; Kim, S. Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Antimicrob. Agents Chemother. 2020, 64, e00819-20. [Google Scholar] [CrossRef] [PubMed]
  88. Kousar, K.; Majeed, A.; Yasmin, F.; Hussain, W.; Rasool, N. Phytochemicals from selective plants have promising potential against SARS-CoV-2: Investigation and corroboration through molecular docking, MD simulations, and quantum computations. BioMed Res. Int. 2020, 2020, 6237160. [Google Scholar] [CrossRef]
  89. Xiao, T.; Cui, M.; Zheng, C.; Zhang, P.; Ren, S.; Bao, J.; Gao, D.; Sun, R.; Wang, M.; Lin, J.; et al. Both baicalein and gallocatechin gallate effectively inhibit SARS-CoV-2 replication by targeting Mpro and sepsis in Mice. Inflammation 2022, 45, 1076–1088. [Google Scholar] [CrossRef]
  90. Zhang, Y.N.; Zhu, G.H.; Liu, W.; Xiong, Y.; Hu, Q.; Zhuang, X.Y.; Jia, G.H.; Zhang, W.D.; Ge, G.B. Discovery and characterization of the covalent SARS-CoV-2 3CLpro inhibitors from Ginkgo biloba extract via integrating chemoproteomic and biochemical approaches. Phytomedicine 2023, 114, 154796. [Google Scholar] [CrossRef]
  91. Abdallah, H.M.; El-Halawany, A.M.; Sirwi, A.; El-Araby, A.M.; Mohamed, G.A.; Ibrahim, S.R.; Koshak, A.E.; Asfour, H.Z.; Awan, Z.A.; A. Elfaky, M. Repurposing of some natural product isolates as SARS-CoV-2 main protease inhibitors via in vitro cell free and cell-based antiviral assessments and molecular modeling approaches. Pharmaceuticals 2021, 14, 213. [Google Scholar] [CrossRef] [PubMed]
  92. Boufissiou, A.; Abdalla, M.; Sharaf, M.; Al-Resayes, S.I.; Imededdine, K.; Alam, M.; Yagi, S.; Azam, M.; Yousfi, M. In-Silico investigation of phenolic compounds from leaves of Phillyrea angustifolia L. as a potential inhibitor against the SARS-CoV-2 main protease (Mpro PDB ID: 5R83) using a virtual screening method. J. Saudi Chem. Soc. 2022, 26, 101473. [Google Scholar] [CrossRef]
  93. Aleebrahim-Dehkordi, E.; Ghoshouni, H.; Koochaki, P.; Esmaili-Dehkordi, M.; Aleebrahim, E.; Chichagi, F.; Jafari, A.; Hanaei, S.; Heidari-Soureshjani, E.; Rezaei, N. Targeting the vital non-structural proteins (NSP12, NSP7, NSP8 and NSP3) from SARS-CoV-2 and inhibition of RNA polymerase by natural bioactive compound naringenin as a promising drug candidate against COVID-19. J. Mol. Struct. 2023, 1287, 135642. [Google Scholar] [CrossRef]
  94. Liu, S.Y.; Wang, W.; Ke, J.P.; Zhang, P.; Chu, G.X.; Bao, G.H. Discovery of Camellia sinensis catechins as SARS-CoV-2 3CL protease inhibitors through molecular docking, intra and extra cellular assays. Phytomedicine 2022, 96, 153853. [Google Scholar] [CrossRef] [PubMed]
  95. Kandeil, A.; Mostafa, A.; Kutkat, O.; Moatasim, Y.; Al-Karmalawy, A.A.; Rashad, A.A.; Kayed, A.E.; Kayed, A.E.; El-Shesheny, R.; Kayali, G.; et al. Bioactive polyphenolic compounds showing strong antiviral activities against severe acute respiratory syndrome coronavirus 2. Pathogens 2021, 10, 758. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, Z.; Yang, L.; Cui, S.; Liang, Y.; Zhang, X. Synthesis and anti-hypertensive effects of the twin drug of nicotinic acid and quercetin tetramethyl ether. Molecules 2014, 19, 4791–4801. [Google Scholar] [CrossRef] [Green Version]
  97. Yang, M.; Lin, L.; Scartelli, C.; Chen, D.Y.; Patel, A.; Bekendam, R.; Sun, L.; Saeed, M.; Flaumenhaft, R. Inhibition of SARS-CoV-2 viral replication and in vivo thrombus formation by a novel plant flavonoid. Blood 2021, 138, 3144. [Google Scholar] [CrossRef]
  98. 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]
  99. Zhu, J.; Yan, H.; Shi, M.; Zhang, M.; Lu, J.; Wang, J.; Chen, L.; Wang, Y.; Li, L.; Miao, L.; et al. Luteolin inhibits spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) binding to angiotensin-converting enzyme 2. Phytother. Res. 2023; in press. [Google Scholar] [CrossRef]
  100. Xie, Y.Z.; Peng, C.W.; Su, Z.Q.; Huang, H.T.; Liu, X.H.; Zhan, S.F.; Huang, X.F. A practical strategy for exploring the pharmacological mechanism of luteolin against COVID-19/asthma comorbidity: Findings of system pharmacology and bioinformatics analysis. Front. Immunol. 2022, 12, 769011. [Google Scholar] [CrossRef]
  101. Spiegel, M.; Ciardullo, G.; Marino, T.; Russo, N. Computational investigation on the antioxidant activities and on the Mpro SARS-CoV-2 non-covalent inhibition of isorhamnetin. Front. Chem. 2023, 11, 1122880. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, H.; Ye, F.; Sun, Q.; Liang, H.; Li, C.; Li, S.; Lu, R.; Huang, B.; Tan, W.; Lai, L. Scutellaria baicalensis extract and baicalein inhibit replication of SARS-CoV-2 and its 3C-like protease in vitro. J. Enzym. Inhib. Med. Chem. 2021, 36, 497–503. [Google Scholar] [CrossRef]
  103. Zandi, K.; Musall, K.; Oo, A.; Cao, D.; Liang, B.; Hassandarvish, P.; Lan, S.; Slack, R.L.; Kirby, K.A.; Bassit, L.; et al. Baicalein and baicalin inhibit SARS-CoV-2 RNA-dependent-RNA polymerase. Microorganisms 2021, 9, 893. [Google Scholar] [CrossRef] [PubMed]
  104. Jin, Y.H.; Lee, J.; Jeon, S.; Kim, S.; Min, J.S.; Kwon, S. Natural polyphenols, 1, 2, 3, 4, 6-O-pentagalloyglucose and proanthocyanidins, as broad-spectrum anticoronaviral inhibitors targeting Mpro and RdRp of SARS-CoV-2. Biomedicines 2022, 10, 1170. [Google Scholar] [CrossRef]
  105. Chen, H.F.; Wang, W.J.; Chen, C.Y.; Chang, W.C.; Hsueh, P.R.; Peng, S.L.; Wu, C.S.; Chen, Y.; Huang, H.Y.; Shen, W.J. The natural tannins oligomeric proanthocyanidins and punicalagin are potent inhibitors of infection by SARS-CoV-2 in vitro. bioRxiv, 2023; preprint. [Google Scholar] [CrossRef]
  106. Chauhan, M.; Bhardwaj, V.K.; Kumar, A.; Kumar, V.; Kumar, P.; Enayathullah, M.G.; Thomas, J.; George, J.; Kumar, B.K.; Purohit, R.; et al. Theaflavin 3-gallate inhibits the main protease (Mpro) of SARS-CoV-2 and reduces its count in vitro. Sci. Rep. 2022, 12, 13146. [Google Scholar] [CrossRef]
  107. Tsai, M.S.; Yang, Y.H.; Lin, Y.S.; Chang, G.H.; Hsu, C.M.; Yeh, R.A.; Shu, L.H.; Cheng, Y.C.; Liu, H.T.; Wu, Y.H.; et al. GB-2 blocking the interaction between ACE2 and wild type and mutation of spike protein of SARS-CoV-2. Biomed. Pharmacother. 2021, 142, 112011. [Google Scholar] [CrossRef] [PubMed]
  108. Wahab, G.A.; Aboelmaaty, W.S.; Lahloub, M.F.; Sallam, A. In vitro and in silico studies of SARS-CoV-2 main protease Mpro inhibitors isolated from Helichrysum bracteatum. RSC Adv. 2022, 12, 18412–18424. [Google Scholar] [CrossRef] [PubMed]
  109. Khan, A.; Heng, W.; Wang, Y.; Qiu, J.; Wei, X.; Peng, S.; Saleem, S.; Khan, M.; Ali, S.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. [Google Scholar] [CrossRef]
  110. Youssef, F.S.; Altyar, A.E.; Omar, A.M.; Ashour, M.L. Phytoconstituents, in vitro anti-infective activity of Buddleja indica Lam., and in silico evaluation of its SARS-CoV-2 inhibitory potential. Front. Pharmacol. 2021, 12, 619373. [Google Scholar] [CrossRef]
  111. Li, L.; Ma, L.; Hu, Y.; Li, X.; Yu, M.; Shang, H.; Zou, Z. Natural biflavones are potent inhibitors against SARS-CoV-2 papain-like protease. Phytochemistry 2022, 193, 112984. [Google Scholar] [CrossRef]
  112. Miroshnychenko, K.V.; Shestopalova, A.V. Combined use of the hepatitis C drugs and amentoflavone could interfere with binding of the spike glycoprotein of SARS-CoV-2 to ACE2: The results of a molecular simulation study. J. Biomol. Struct. Dyn. 2022, 40, 8672–8686. [Google Scholar] [CrossRef] [PubMed]
  113. Mandal, S.K.; Rehman, M.U.; Katyal, A.; Rajvanshi, K.; Kannan, M.; Garg, M.; Murugesan, S.; Deepa, P.R. In silico anti-viral assessment of phytoconstituents in a traditional (Siddha Medicine) polyherbal formulation—Targeting Mpro and pan-coronavirus post-fusion Spike protein. J. Tradit. Complement. Med. 2023; in press. [Google Scholar] [CrossRef]
  114. Bouback, T.A.; Aljohani, A.M.; Albeshri, A.; Al-Talhi, H.; Moatasim, Y.; GabAllah, M.; Badierah, R.; Albiheyri, R.; Al-Sarraj, F.; Ali, M.A. Antiviral activity of Humulus lupulus (HOP) aqueous extract against MERS-CoV and SARS-CoV-2: In-vitro and in-silico study. Biotechnol. Biotechnol. Equip. 2023, 37, 167–179. [Google Scholar] [CrossRef]
  115. Mukherjee, S.; Sharma, D.; Sharma, A.K.; Jaiswal, S.; Sharma, N.; Borah, S.; Kaur, G. Flavan-based phytoconstituents inhibit Mpro, a SARS-CoV-2 molecular target, in silico. J. Biomol. Struct. Dyn. 2022, 40, 11545–11559. [Google Scholar] [CrossRef] [PubMed]
  116. Yi, Y.; Zhang, M.; Xue, H.; Yu, R.; Bao, Y.O.; Kuang, Y.; Chai, Y.; Ma, W.; Wang, J.; Shi, X.; et al. Schaftoside inhibits 3CLpro and PLpro of SARS-CoV-2 virus and regulates immune response and inflammation of host cells for the treatment of COVID-19. Acta Pharm. Sin. B 2022, 12, 4154–4164. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, Y.; Li, W.; Hu, Y.; Ding, T.; Zafar, M.M.; Jia, X.; Zhang, L.; Ren, M.; Li, F.; Wang, W. Cotton flower metabolites inhibit SARS-CoV-2 main protease. FEBS Open Bio 2022, 12, 1886–1895. [Google Scholar] [CrossRef]
  118. Vicidomini, C.; Roviello, V.; Roviello, G.N. In silico investigation on the interaction of chiral phytochemicals from opuntia ficus-indica with SARS-CoV-2 Mpro. Symmetry 2021, 13, 1041. [Google Scholar] [CrossRef]
  119. Chaves, O.A.; Lima, C.R.; Fintelman-Rodrigues, N.; Sacramento, C.Q.; de Freitas, C.S.; Vazquez, L.; Temerozo, J.R.; Rocha, M.E.N.; Dias, S.S.G.; Carels, N.; et al. Agathisflavone, a natural biflavonoid that inhibits SARS-CoV-2 replication by targeting its proteases. Int. J. Biol. Macromol. 2022, 222, 1015–1026. [Google Scholar] [CrossRef]
  120. Farhat, A.; Ben Hlima, H.; Khemakhem, B.; Ben Halima, Y.; Michaud, P.; Abdelkafi, S.; Fendri, I. Apigenin analogues as SARS-CoV-2 main protease inhibitors: In-silico screening approach. Bioengineered 2022, 13, 3350–3361. [Google Scholar] [CrossRef]
  121. Ngwe Tun, M.M.; Toume, K.; Luvai, E.; Nwe, K.M.; Mizukami, S.; Hirayama, K.; Komatsu, K.; Morita, K. The discovery of herbal drugs and natural compounds as inhibitors of SARS-CoV-2 infection in vitro. J. Nat. Med. 2022, 76, 402–409. [Google Scholar] [CrossRef] [PubMed]
  122. Jo, S.; Kim, S.; Kim, D.Y.; Kim, M.S.; Shin, D.H. Flavonoids with inhibitory activity against SARS-CoV-2 3CLpro. J. Enzym. Inhib. Med. Chem. 2020, 35, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
  123. Lu, L.; Peng, Y.; Yao, H.; Wang, Y.; Li, J.; Yang, Y.; Lin, Z. Punicalagin as an allosteric NSP13 helicase inhibitor potently suppresses SARS-CoV-2 replication in vitro. Antivir. Res. 2022, 206, 105389. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structures of epigallocatechin 3-gallate (EGCG) and myricetin.
Figure 1. The chemical structures of epigallocatechin 3-gallate (EGCG) and myricetin.
Nutrients 15 03443 g001
Figure 3. Proposed multi-target (S protein, Nsp15, and Mpro) mechanism of action of EGCG against SARS-CoV-2: inhibition of oxidative stress, cytokine storm, lung fibrosis, thrombosis, and sepsis injury in SARS-CoV-2 infection. Oxidized EGCG is first recognized by the catalytic site of Mpro, which is followed by the covalent bonding between the α,β-unsaturated carbonyl moiety of EGCG (serves as an electrophile) and Cys145 of Mpro (serves as a nucleophile).
Figure 3. Proposed multi-target (S protein, Nsp15, and Mpro) mechanism of action of EGCG against SARS-CoV-2: inhibition of oxidative stress, cytokine storm, lung fibrosis, thrombosis, and sepsis injury in SARS-CoV-2 infection. Oxidized EGCG is first recognized by the catalytic site of Mpro, which is followed by the covalent bonding between the α,β-unsaturated carbonyl moiety of EGCG (serves as an electrophile) and Cys145 of Mpro (serves as a nucleophile).
Nutrients 15 03443 g003
Figure 4. Myricetin-based lead discovery and optimization. Myricetin is a waxberry-derived covalent Mpro inhibitor suitable for lead optimization.
Figure 4. Myricetin-based lead discovery and optimization. Myricetin is a waxberry-derived covalent Mpro inhibitor suitable for lead optimization.
Nutrients 15 03443 g004
Figure 5. Possible mechanism of myricetin oxidation.
Figure 5. Possible mechanism of myricetin oxidation.
Nutrients 15 03443 g005
Figure 6. Structure–activity relationships of myricetin derivatives as leads for SARS-CoV-2 treatment.
Figure 6. Structure–activity relationships of myricetin derivatives as leads for SARS-CoV-2 treatment.
Nutrients 15 03443 g006
Table 1. Other natural dietary flavonoids for treating SARS-CoV-2 infection in vitro.
Table 1. Other natural dietary flavonoids for treating SARS-CoV-2 infection in vitro.
No.NameSpeciesStructureEC50 or IC50 (μM)Target or
Mechanism
Refs.
1HesperidinCitrus sinensisNutrients 15 03443 i00113.25ACE2, M, S, and RBD proteins[81,82]
2Ugonin JHelminthostachys zeylanicaNutrients 15 03443 i0022.38Mpro[83]
3Epicatechin-3-O-gallateCamellia sinensis var. sinensisNutrients 15 03443 i0035.21Mpro[84,85]
4Catechin-3-O-gallateSenegalia catechuNutrients 15 03443 i0042.98Mpro[84]
5Procyanidin B2Punica granatumNutrients 15 03443 i00575.3Mpro[84,86]
6OsajinMaclura pomiferaNutrients 15 03443 i0063.87N protein, nsp16, and nsp13[87,88]
7(+)-GallocatechinMusa CavendishNutrients 15 03443 i00713.14Mpro[89,90]
8Apigenin-7-O-glucosideAchillea millefolium L.Nutrients 15 03443 i0080.074Mpro[91,92]
9NaringeninCitrus reticulataNutrients 15 03443 i0090.092Mpro, NSP12, NSP7, NSP8, and NSP3[91,93]
10etc-pyrrolidinone C and DCamellia sinensisNutrients 15 03443 i0100.90Mpro[94]
11(−)-epicatechin 3-O-caffeoateCamellia sinensisNutrients 15 03443 i0111.58Mpro[94]
12QuercetinCitrus reticulata BlancoNutrients 15 03443 i01218.2Mpro[95,96]
133,8′-biapigeninForsythia suspensaNutrients 15 03443 i01313.0Mpro, protein disulfide isomerase[97]
14PGHGPenthorum chinense PurshNutrients 15 03443 i0146.5Mpro, protein disulfide isomerase[97]
15LuteolinTaraxacum antungense KitagNutrients 15 03443 i01511.81Mpro, RBD-ACE2[98,99,100]
16IsorhamnetinSea buckthornNutrients 15 03443 i0168.42/2.51Mpro[98,101]
17BaicaleinScutellaria baicalensis GeorgiNutrients 15 03443 i0170.39Mpro, RdRp[102,103]
18ScutellareinScutellaria baicalensis GeorgiNutrients 15 03443 i0185.8Mpro[102]
19ProanthocyanidinGrape seedNutrients 15 03443 i01925.90/21.02Mpro, and RdRp[104,105]
20Theaflavin 3-gallateBlack teaNutrients 15 03443 i02018.48Mpro, S protein[106,107]
21TheaflavinBlack teaNutrients 15 03443 i02122.22Mpro[106]
223′,5′,5,7-tetrahydroxy-6-methoxyflavanoneHelichrysum bracteatumNutrients 15 03443 i0225.565Mpro[108]
23KaempferolCanavalia ensiformis L.Nutrients 15 03443 i02334.46Mpro, PLpro[109,110]
24AmentoflavoneNandina domesticaNutrients 15 03443 i02413.0PLpro, RBD-ACE2[111,112]
25ScutellareinScutellaria baicalensisNutrients 15 03443 i0255.80Mpro[102,113]
26Epicatechin gallateFagopyrum esculentumNutrients 15 03443 i02612.5Mpro[36,114,115]
27SchaftosideProsopis alba cotyledonsNutrients 15 03443 i02711.83Mpro and PLpro[116]
28AstilbinSmilax glabra Roxb.Nutrients 15 03443 i0287.92Mpro[117]
29AstragalinNelumbo nuciferaNutrients 15 03443 i0290.13Mpro[117,118]
30ApigeninApium Graveolens L.Nutrients 15 03443 i0305.21Mpro[119,120]
31BaicalinScutellaria baicalensisNutrients 15 03443 i0318.8RdRp and Mpro[121,122]
32RhodiosinRhodiola roseaNutrients 15 03443 i0320.48NSP13[123]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, Z.; Yang, L. The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection. Nutrients 2023, 15, 3443. https://doi.org/10.3390/nu15153443

AMA Style

Wang Z, Yang L. The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection. Nutrients. 2023; 15(15):3443. https://doi.org/10.3390/nu15153443

Chicago/Turabian Style

Wang, Zhonglei, and Liyan Yang. 2023. "The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection" Nutrients 15, no. 15: 3443. https://doi.org/10.3390/nu15153443

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