Next Article in Journal
Revealing Edible Bird Nest as Novel Functional Foods in Combating Metabolic Syndrome: Comprehensive In Silico, In Vitro, and In Vivo Studies
Next Article in Special Issue
Plant-Derived Epi-Nutraceuticals as Potential Broad-Spectrum Anti-Viral Agents
Previous Article in Journal
Urolithins: A Prospective Alternative against Brain Aging
Previous Article in Special Issue
The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 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
General Surgery Department, Baoan Central Hospital, Affiliated Baoan Central Hospital of Guangdong Medical University, Shenzhen 518000, China
4
School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, China
*
Authors to whom correspondence should be addressed.
Nutrients 2023, 15(18), 3885; https://doi.org/10.3390/nu15183885
Submission received: 25 August 2023 / Revised: 31 August 2023 / Accepted: 5 September 2023 / Published: 6 September 2023

Abstract

:
The COVID-19 pandemic has stimulated collaborative drug discovery efforts in academia and the industry with the aim of developing therapies and vaccines that target SARS-CoV-2. Several novel therapies have been approved and deployed in the last three years. However, their clinical application has revealed limitations due to the rapid emergence of viral variants. Therefore, the development of next-generation SARS-CoV-2 therapeutic agents with a high potency and safety profile remains a high priority for global health. Increasing awareness of the “back to nature” approach for improving human health has prompted renewed interest in natural products, especially dietary polyphenols, as an additional therapeutic strategy to treat SARS-CoV-2 patients, owing to its good safety profile, exceptional nutritional value, health-promoting benefits (including potential antiviral properties), affordability, and availability. Herein, we describe the biological properties and pleiotropic molecular mechanisms of dietary polyphenols curcumin, resveratrol, and gossypol as inhibitors against SARS-CoV-2 and its variants as observed in in vitro and in vivo studies. Based on the advantages and disadvantages of dietary polyphenols and to obtain maximal benefits, several strategies such as nanotechnology (e.g., curcumin-incorporated nanofibrous membranes with antibacterial-antiviral ability), lead optimization (e.g., a methylated analog of curcumin), combination therapies (e.g., a specific combination of plant extracts and micronutrients), and broad-spectrum activities (e.g., gossypol broadly inhibits coronaviruses) have also been emphasized as positive factors in the facilitation of anti-SARS-CoV-2 drug development to support effective long-term pandemic management and control.

1. Introduction

The large-scale outbreak of coronavirus disease 2019 (COVID-19), caused by the highly transmissible and pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), placed a significant burden on the economies and healthcare sector globally [1,2]. Remarkably, several vaccines (e.g., Comirnaty®, Spikevax®, Vaxzevria, Sinopharm®, and Nuvaxovid™) [3], herbal medicines (e.g., Lianhuaqingwen capsules) [4,5], and small-molecule therapies (e.g., Veklury® [6], azvudine [7], VV116 [8], Lagevrio® [9], and Paxlovid® [10]) targeting SARS-CoV-2 were approved and deployed in the clinic in a relatively short time frame. On 5 May 2023, the World Health Organization declared the end of the global health emergency, with a final death toll of at least 20 million [11]. However, SARS-CoV-2 remains a threat as it continues to spread globally. In addition, many people also continue to suffer from non-negligible long-term effects of COVID-19 (long COVID; post-acute sequelae of COVID-19), and SARS-CoV-2 variants/subvariants (e.g., Omicron variants BA.2.75, XBB.1.5, XBB.1.16, BQ.1, CH.1.1, and FE.1) are still emerging [12,13]. Long COVID is a multi-systemic disease with differing pathology in numerous organs (more than 200 symptoms were identified) [14]. Furthermore, long COVID occurs in at least 10% of SARS-CoV-2 infections, including non-hospitalized cases (10–30%), hospitalized cases (50–70%), and vaccinated cases (10–12%) [14]. In parallel, although SARS-CoV-2 vaccination significantly reduces mortality (similar to seasonal influenza), it has little effect on the viral transmission rate, thus contributing to the burden on the healthcare sector and economies of countries globally. A conservative estimate of the annual burden of SARS-CoV-2 infection suggests that it will be twice that of previous influenza seasons [15]. Therefore, there is an urgent need for a highly effective antiviral agent.
Natural dietary products have exceptional nutritional value, good safety profiles, health-promoting properties, and are abundantly available [16]. Fruits, vegetables, and spices are potential sources of dietary polyphenols that provide important benefits to human health, partly attributed to their anti-inflammatory, antioxidant, and antiviral properties [17,18,19,20]. Repurposing existing dietary polyphenols is an attractive approach for preventing or treating SARS-CoV-2 infection [21]. Numerous studies, both in in vitro and in vivo, have shown that dietary polyphenols (e.g., curcumin [22], resveratrol [23], and gossypol [24]; Figure 1) are beneficial for COVID-19 treatment via targeting of the SARS-CoV-2 papain-like protease (PLpro), main protease (Mpro, 3CLpro), RNA-dependent RNA polymerase (RdRp), and spike (S) glycoprotein [25]. Building on our previously published work [15,26], the biological properties and pleiotropic molecular mechanisms of natural dietary polyphenols as inhibitors against SARS-CoV-2 and its variants, observed in in vitro and in vivo studies, are described to support anti-SARS-CoV-2 drug discovery and development. Nonetheless, the development of dietary polyphenol-based next-generation therapies for SARS-CoV-2 still faces multiple potential challenges. Here in this review, we discuss future representative directions, combination therapies, nanotechnology, drug delivery, lead optimization, and broad-spectrum activities of dietary polyphenols targeting SARS-CoV-2 and its variants.

2. Curcumin—A Turmeric-Derived Complementary Drug against COVID-19

Curcumin, a health-promoting polyphenol isolated from the dietary spice turmeric (Curcuma longa L.) (Figure 2a), has several well-known pharmacological properties, including antioxidant [27], anti-inflammatory [28], antifungal [29], neuroprotective [30], anticancer [31], and wound-healing effects [32]. Curcumin also has broad-spectrum antiviral activity in vitro with low-micromolar efficacy. Targeted viruses include the Zika virus (IC50 = 1.9 μM) [33], HIV-1 virus (IC50 = 12 μM) [34], and SARS-CoV (IC50 = 5.7 μM) [35]. Curcumin is particularly effective in treating respiratory diseases, including acute lung injury [36], pulmonary fibrosis [37], allergic asthma [38], pulmonary infections [39], and chronic obstructive pulmonary disease [40]. Furthermore, there are no reports of curcumin treatment-related toxicity in humans when administered at doses of up to 8 g/day for up to 3 months [41]. Additionally, more than 300 clinical trials have demonstrated the protective effects of curcumin against a variety of conditions, including respiratory, liver, inflammatory, and metabolic diseases [42,43]. Curcumin has shown good safety profiles, broad-spectrum antiviral activities, and protective effects in multiple organs, making it a promising candidate for complementary treatment of SARS-CoV-2 infection.
Curcumin represents an ideal scaffold for COVID-19 drug discovery, and impressive progress has been achieved in research associated with the development of curcumin-related anti-SARS-CoV-2 drugs [44,45]. For example, Bormann et al. [22] demonstrated that pure curcumin effectively neutralizes SARS-CoV-2 in Vero E6 cells with an EC50 of 21.2 µM. In the same study, curcumin reduced SARS-CoV-2 RNA levels in Vero E6 cells with an EC50 of 38 µM. Notably, both curcumin-containing nutritional supplement capsules and turmeric root extract are shown to completely neutralize SARS-CoV-2 in vitro. Marín-Palma et al. [46] reported that curcumin (10 µg/mL) exerted anti-SARS-CoV-2 effects of 99.0% and 99.8% against the DG614 strain and the Delta variant in Vero E6 cells, respectively. Mechanistic studies have revealed that curcumin can inactivate cellular enzymes involved in viral fusion with host membranes, thus blocking viral entry. Importantly, curcumin is shown to prevent the production and release of IL-1β, IL-6, MCP-1, and IL-8. Furthermore, Bahun et al. [47] reported that curcumin effectively inhibited SARS-CoV-2 Mpro replication in vitro (IC50 = 11.9 μM). Meanwhile, in silico molecular dynamics studies indicated that the Mpro active sites Gln192 and Arg188 participate in hydrogen bonding interactions with curcumin [47].
Several trials have tentatively investigated the therapeutic effect of curcumin (Figure 2b). For example, Ujjan et al. [48] conducted an open-label, randomized, controlled clinical trial (ClinicalTrials.gov: NCT04603690) in Pakistan to evaluate the efficacy and safety of an oral curcumin-quercetin supplement plus standard of care vs. standard of care alone in outpatients with early-stage, mild to moderately symptomatic COVID-19. In total, 50 patients were enrolled, 25 of which were assigned to receive a curcumin-quercetin supplement (daily intake of 168 mg of curcumin and 260 mg of quercetin, twice a day for 2 weeks) alongside standard of care, while the other 25 were assigned to receive only standard of care (control group). Viral clearance was significantly higher in the curcumin-quercetin group (18/25 [72.0%]) vs. 6/25 [24.0%], respectively; p = 0.0008). Furthermore, the percentage of acute COVID-19-associated symptoms was lower in the curcumin-quercetin group compared to the control group (complete symptom resolution: 40.0% [10/25] vs. 16.0% [4/25], respectively; p = 0.061). No treatment-emergent adverse effects were observed in the curcumin-quercetin group. Several other studies have obtained results consistent with these findings. Khan et al. [49] conducted another open-label, randomized, controlled trial (ClinicalTrials.gov: NCT05130671) in Pakistan to assess the efficacy of a 14-day treatment regimen comprising a daily oral co-supplementation of curcumin (168 mg), quercetin (260 mg), and vitamin D3 (9 µg) as adjuvant therapy in 25 patients with mild to moderate COVID-19. The authors reported that patients in the oral co-supplementation group exhibited accelerated negativization of the SARS-CoV-2 RT-PCR test compared to those in the control group (60.0% [15/25] vs. 20.0% [5/25], respectively; p = 0.009). Furthermore, Hellou et al. [50] conducted a double-blinded, multicenter, placebo-controlled Phase II clinical trial in Israel (ClinicalTrials.gov: NCT04382040) to evaluate the efficacy and safety of ArtemiC, an oral spray containing curcumin (40 mg), artemisinin (12 mg), frankincense (30 mg), and vitamin C (120 mg), for 15 days in hospitalized patients with symptomatic COVID-19 (N = 33 vs. N = 17 for the placebo group). The results showed that 91% of patients receiving ArtemiC oral spray displayed a significant improvement in the National Early Warning Score 2. Compared with the placebo, ArtemiC treatment shortened the duration of oxygen supplementation, hospital admission time, and abnormal oxygen saturation. Overall, accumulating clinical evidence supports the hypothesis that curcumin exerts beneficial effects in the treatment of COVID-19.
However, the clinical application of curcumin is greatly limited by its poor chemical stability and oral bioavailability, short half-life, and lack of target specificity [51,52]. These limitations may potentially be overcome via the application of nanotechnology (Figure 2c,d) and lead optimization (Figure 2e). For example, Gunathilake et al. [53] developed a curcumin-loaded, inhalable, nanotherapeutic (nanocellulose/polyvinyl alcohol/curcumin) for use in COVID-19. The curcumin in these nanoparticles had good water solubility (313.61 mg/L) and a high loading capacity (8.90 mg/g) and represented a promising alternative strategy for the treatment of COVID-19. De Maio et al. [54] fabricated personal protective equipment coated with graphene and curcumin. These coatings interacted with the SARS-CoV-2 surface, thereby trapping the virus and inhibiting further transmission. Importantly, the authors suggested that the combination of the mechanical and chemical actions of the two antimicrobials had the potential to limit the emergence and spread of resistant SARS-CoV-2 strains. Organic nanofibrous membranes with ultrafine particle filtration and high gas permeance show great potential in preventing SARS-CoV-2 infection [55]. Rao et al. [56] developed a biocompatible curcumin-incorporated composite membrane for pathogen sterilization and isolation via antibacterial-antiviral functionalization and controllable heating lamination. This filter exhibited excellent gas permeability (3423.6 m3/[m2·h·kPa]), a high ultrafine particle rejection rate (>98.79%), and good SARS-CoV-2 capture and kill efficiency (99.90% for 5 min). Moreover, the curcumin-incorporated filter also showed high antibacterial activity against a variety of bacteria (Escherichia coli [99.84%], Bacillus subtilis [99.02%]) and fungi (Aspergillus niger [93.60%], Penicillium [95.23%]) in vitro. More significantly, the filter showed good stability after 10 heating cycles, which was indicative of its reusability. This material could be widely used in the design of respirator masks. Moreover, Sharma et al. [57] prepared curcumin-encapsulated polysaccharide nanoparticles (Cur-PS-NPs) with monodisperse, spherical morphologies; diameters of 43 and 22 nm as measured using dynamic light scattering and transmission electron microscopy, respectively; a low polydispersity index (0.52); a high entrapment efficiency (25%); and excellent bioavailability. The Cur-PS-NPs attenuated the interaction between the angiotensin-converting enzyme 2 (ACE2) receptor and the SARS-CoV-2 S protein. Mechanistically, Cur-PS-NPs suppressed SARS-CoV-2 S protein-induced cytokine storms in liver Huh7.5 and lung A549 epithelial cells by inhibiting the NF-κB/MAPK signaling pathway, which in turn decreased the SARS-CoV-2 S protein-mediated phosphorylation of p38 MAPK, p42/44 MAPK, and p65/NF-κB, as well as the expression of p65/NF-κB. Importantly, treatment with Cur-PS-NPs had almost no effect on S protein-naive (unstimulated) Huh7.5 and A549 cells. The results of this study provided a foundation for the development of curcumin-based nanotherapeutics with improved curcumin bioavailability to treat COVID-19 by mitigating hyperinflammatory responses and preventing lung and liver injuries.
Figure 2. Curcumin, a turmeric-derived complementary drug, may be effective against COVID-19 with the assistance of combination therapies, nanotechnology, and lead optimization. (a) The chemical structure of curcumin isolated from Curcuma longa L. (b) Clinical trials of curcumin in the form of oral co-supplementation and throat spray. (c) Schematic representation of the CUR-PS-NP preparation process and its use in preventing lung and liver injuries associated with SARS-CoV-2 spike protein-mediated cytokine storms. (Adapted with permission from TOC and Figure 2 of Sharma et al.; ACS Appl. Bio Mater. 2022, 5, 483–491 [57]. Copyright © 2022 American Chemical Society). (d) The process involved in the preparation of curcumin-incorporated nanofibrous membranes (A–A mask) and a schematic representation of the SARS-CoV-2 inactivation mechanism. (Adapted with permission [56]. Copyright © 2022 Elsevier B.V.) (e) Curcumin-based lead optimization.
Figure 2. Curcumin, a turmeric-derived complementary drug, may be effective against COVID-19 with the assistance of combination therapies, nanotechnology, and lead optimization. (a) The chemical structure of curcumin isolated from Curcuma longa L. (b) Clinical trials of curcumin in the form of oral co-supplementation and throat spray. (c) Schematic representation of the CUR-PS-NP preparation process and its use in preventing lung and liver injuries associated with SARS-CoV-2 spike protein-mediated cytokine storms. (Adapted with permission from TOC and Figure 2 of Sharma et al.; ACS Appl. Bio Mater. 2022, 5, 483–491 [57]. Copyright © 2022 American Chemical Society). (d) The process involved in the preparation of curcumin-incorporated nanofibrous membranes (A–A mask) and a schematic representation of the SARS-CoV-2 inactivation mechanism. (Adapted with permission [56]. Copyright © 2022 Elsevier B.V.) (e) Curcumin-based lead optimization.
Nutrients 15 03885 g002
Lead optimization of curcumin provides another promising strategy for anti-SARS-CoV-2 drug development. For example, dimethoxycurcumin, a methylated analog of curcumin, can acidify endolysosomes and inhibit SARS-CoV-2 entry [58]. Compared to curcumin, dimethoxycurcumin has greater metabolic stability, as well as superior bioavailability and anti-inflammatory properties. (1E,6E)-1,7-Bis(4-(dimethylamino)phenyl)hepta-1,6-diene-3,5-dione (C-NMe2), a photodynamic dimethyl amino derivative of curcumin, exhibits high stability in an aqueous environment and improved interactions with two enzymes, which are important for SARS-CoV-2 replication, namely, PLpro and ADP ribose phosphatase [59]. Dhaka et al. [60] revealed that bisdemethoxycurcumin, a natural demethoxy analog of curcumin, can effectively inhibit SARS-CoV-2 replication by targeting nucleocapsid protein with an EC50 value of 1.64 µM and with high selectivity (SI = 15.24).
These results support further clinical evaluation of curcumin as an alternative to existing targeted therapies for COVID-19. Current information on completed or ongoing clinical trials of curcumin for COVID-19 treatment is summarized in Table 1. Taken together, the above studies highlight that the delivery, efficacy, and bioavailability of curcumin-based COVID-19 therapies can be improved with the assistance of nanotechnology, combination therapies, and lead optimization.

3. Resveratrol and Its Natural Derivatives—Grape-Derived ACE2 Inhibitors That Can Limit SARS-CoV-2 Infection

Resveratrol (RSV), a polyphenolic nutraceutical mainly found in grapes (Vitis vinifera L.) (Figure 3), demonstrates health-promoting properties, including antioxidative [61], immunomodulatory [62], and anti-aging effects [63]. Since its association with the “French paradox” in 1992, resveratrol has received increasing research interest owing to its diverse pharmacological activities and multi-organ protective effects (e.g., acute lung injury [64], cerebral ischemia/reperfusion injury [65], myocardial cell apoptosis [66], and hepatic injury [67]). Resveratrol is classified as a food supplement with an excellent safety record and no serious adverse events even when administered in large doses (up to 600 mg per day) [68]. This nutraceutical is recognized as a promising therapeutic against a variety of viruses, including influenza A virus [69], respiratory syncytial virus [70], SARS-CoV [71], MERS-CoV [72], and HCoV-229E [73].
Numerous studies, both in vitro and in vivo, have shown that resveratrol is beneficial for SARS-CoV-2 treatment. Yang et al. [23] demonstrated that resveratrol significantly inhibits the replication of SARS-CoV-2 in Vero cells with an EC50 of 4.48 μM. Concomitantly, Pasquereau et al. [73] reported that resveratrol displays inhibitory activity against SARS-CoV-2 in Vero E6 cells with an EC90 and EC50 of 11.42 and 10.66 μM, respectively. Bahun et al. [47] found that resveratrol inhibited Mpro activity with an IC50 value of 16.9 µM. SARS-CoV-2 open reading frame-3a (ORF3a), an accessory protein implicated in autophagy inhibition, inflammasome activation, and apoptosis, is a potential therapeutic target against COVID-19 [74,75] (Figure 4b). Fam et al. [76] indicated that resveratrol shows promise as an ORF3a inhibitor, with an IC50 of 6.73 μM (Figure 4c). The post-acute sequelae of SARS-CoV-2 infection include long-term effects on tissues and organs [77]. Notably, adjunctive therapy with resveratrol helps reduce SARS-CoV-2 infection-induced inflammation, thereby improving patient outcomes. For example, in Brazil, de Souza Andrade et al. [78] evaluated the in vitro effect of resveratrol on neutrophil extracellular trap (NET) production in 190 hospitalized patients with moderate, severe, or critical COVID-19. The results clearly showed that resveratrol significantly attenuated NET formation and increased neutrophil viability in patients with severe disease. Idiopathic pulmonary fibrosis is a key feature in many post-COVID-19 patients [78]. Sheng et al. [79] demonstrated that piceatannol, a naturally occurring hydroxylated resveratrol analog, protects against bleomycin-induced pulmonary fibrosis by targeting the Smad3/ERK/p38 signaling pathway.
Combination therapy has been proposed as a potential strategy for treating SARS-CoV-2 infection [80]. Goc et al. [81,82] evaluated the in vitro efficacy of a specific combination of plant extracts and micronutrients (a mixture composed of resveratrol, curcumin, quercetin, baicalin, vitamin C, theaflavin, N-acetylcysteine, naringenin, and broccoli extract) against SARS-CoV-2 and its Alpha, Beta, Gamma, Delta, Kappa, Mu, and Omicron variants. They found that this combination exerted significant inhibitory effects against SARS-CoV-2 and its variants in vitro via pleiotropic mechanisms, including the targeting of viral RdRp, furin, and cathepsin L activity. At a concentration of 10 μg/mL, combination therapy of resveratrol with naturally occurring compounds enhanced treatment efficacy compared to a single compound (i.e., inhibited RBD binding to the human ACE2 receptor by 90% compared to the control treatment). These findings have important implications for ensuring the effective treatment of SARS-CoV-2 and emerging sarbecovirus clades [81,82]. Polydatin and pterostilbene, natural precursors of resveratrol, have stronger antioxidant effects and better bioavailability than resveratrol [83,84]. De Angelis et al. [85] reported that a mixture containing these polyphenols plus ellagic acid, honokiol, zinc, selenium, and chromium showed greater efficacy in inhibiting SARS-CoV-2 infection than polydatin or pterostilbene treatment alone.
Resveratrol oligomers (condensation ≥ 2), with unique structures and pleiotropic biological activities, have attracted substantial attention as potential food additives for COVID-19 treatment (Figure 3). A library composed of 512 compounds derived from natural products was screened using a high-throughput RBD/ACE2 binding assay and three resveratrol oligomers—hopeaphenol, vatalbinoside A, and vaticanol B—were identified as inhibiting both RBD/ACE2 binding (IC50 values of 0.11, 0.24, and 0.067 μM, respectively) and Mpro activity (IC50 values of 42.5, 68.7, and 47.6 μM, respectively) [86]. These results suggested that resveratrol oligomers are effective at inhibiting RBD/ACE2 binding (Figure 4a). Notably, no evidence of cytotoxicity was observed with these compounds. Additionally, Tietjen et al. [86] reported that hopeaphenol can inhibit the cellular entry of USA-WA1/2020, Alpha, and Beta variants of SARS-CoV-2 with EC50s of 23.4, 7.8, and 7.5 μM, respectively. These results further underscore the potential of these three resveratrol oligomers as attractive candidates for broad-spectrum antiviral therapy with the ability to respond to emerging SARS-CoV-2 variants. Gangadevi et al. [87] demonstrated that kobophenol A, a resveratrol tetramer, effectively blocks the interaction between the host ACE2 receptor and S1-RBD in vitro with an IC50 of 1.81 μM, and inhibits SARS-CoV-2 infection in VeroE6 cells with an EC50 of 71.6 μM. The protease cathepsin L is essential for SARS-CoV-2 infection [88]. Wang et al. [89] found that two resveratrol oligomers, miyabenol C (IC50 = 3.08 μM) and trans-ε-viniferin (IC50 = 40.4 μM), specifically inhibited the entry of SARS-CoV-2 by inhibiting cathepsin L activity. Resveratrol oligomers show a better pharmacokinetic profile than resveratrol itself. Accordingly, non-toxic, natural-product resveratrol oligomers may be promising lead compounds for COVID-19 treatment.
Resveratrol exhibits low bioavailability owing to its poor solubility and rapid metabolism and must be administered in very high oral doses to achieve therapeutic efficacy [90]. Given these critical limitations, efforts have been made in the field of nanomedicines to exploit other therapeutic agents capable of targeting SARS-CoV-2. Zakaria et al. [91] prepared spherical resveratrol-loaded nano-bilosomes (F5) with a mean diameter of 228.9 nm, a zeta potential of −39.8 mV, a high drug entrapment efficiency (86.1%), and superior cellular uptake (~4.7-fold greater than that for resveratrol in Caco-2 cells). F5 exhibited an IC50 of 0.24 μg/mL against SARS-CoV-2, a 6.6-fold improvement compared with an IC50 of 1.6 μg/mL for resveratrol. Interestingly, F5 has a good safety profile, with an SI of 139.5 for F5 and 2.9 for resveratrol. Importantly, F5 could overcome the extensive first-pass liver metabolism and degradation associated with oral resveratrol administration. This study provides a basis for the development of nanoparticles containing resveratrol as oral remedies for COVID-19.

4. Gossypol—A Cotton Plant-Derived RNA-Dependent RNA Polymerase Inhibitor with Broad-Spectrum Anti-Coronavirus Activity

RdRp, an essential therapeutic target that catalyzes the replication of RNA from RNA template, is highly conserved in positive-sense single-stranded RNA viruses, including SARS-CoV-2 [92]. Plant viruses cause devastating diseases in many important agriculture systems worldwide, yet studies show that Gossypium spp. (cotton plant) display strong resistance to single-stranded RNA viruses [93], which provide proactive drug design strategies to minimize the impact of antiviral drug resistance.
An enantiomeric mixture (atropisomerism) of natural polyphenol gossypol (Figure 5), an extraction commonly derived from the cotton plant (stems, leaves, roots, bolls, and seeds) has been shown to exhibit antioxidant [94], antivirus [95], antimicrobial [96], and anticancer properties [97]. Gossypol (GOS) is reported to be effective at treating lung diseases, including non-small cell lung cancer [98], lung injury [99], and pulmonary fibrosis [100]. GOS exists as enantiomers due to the restricted rotation around the internal binaphthyl bond. Interestingly, (−)-gossypol [(−)-GOS] is shown to be active as a male contraceptive, whereas (+)-gossypol [(+)-GOS] has previously been reported to be inactive [101]. Furthermore, its main therapeutic form, gossypol acetate (GOSAc) (Figure 5), an equimolar crystalline complex of racemic gossypol with acetic acid, is a clinically approved gynecological drug used to treat uterine leiomyoma in China [102]. Wang et al. [24] demonstrated that GOS, a promising lead compound, is beneficial for SARS-CoV-2 treatment (Table 2). To be specific, firstly, an antiviral library consisting of 881 cotton natural compounds was screened initially in vitro as potential counters to SARS-CoV-2 infection. Among these candidates, GOS (IC50 = 14.15 μM), (−)-GOS (IC50 = 15.17 μM), and GOSAc (IC50 = 14.83 μM) were identified, displaying more potent inhibitory effects against SARS-CoV-2 RdRp than the nucleoside RdRp inhibitor, remdesivir triphosphate (IC50 = 37.67 μM), and the non-nucleoside RdRp inhibitor, baicalein (IC50 = 62.55 μM), in vitro. In addition, GOS, GOSAc, and (−)-GOS were also effective dose-dependent inhibitors of SARS-CoV-2 replication in vitro in Vero E6 cells with EC50 values of 0.31 μM, 0.84 μM, and 0.72 μM, respectively, and no significant cellular cytotoxicity. In addition, the therapeutic indexes were 116.71, 42.17, and 61.82, respectively, suggesting that the antiviral effect of GOS was not affected by its optical activity [24].
Additionally, Wang et al. [24] revealed that GOS administered via the intramuscular or intranasal route can enhance anti-SARS-CoV-2 activity in a mouse model, resulting in significantly reduced SARS-CoV-2 replication in nasal turbinate in a dose-dependent manner. To elucidate the underlying inhibitory mechanism at a molecular level, the authors [24] determined the 3.36 Å crystal structure of GOS in complex with SARS-CoV-2 RdRp (PDB ID: 7BV2). This structure indicates that two GOS molecules ([(−)-GOS] and [(+)-GOS]) occupy the center of the central cavity and jointly reduce the cavity opening size (Figure 6). Further analysis of the complex structure revealed that GOS inhibits RdRp activity by occupying the binding site for the RNA template [(−)-GOS] and primer [(+)-GOS]), consequentially inhibiting the catalytic activity of RdRp (Figure 6). Specifically, in vitro, data suggest that RdRp mutants of SARS-CoV-2 variants (including Omicron, the RdRpP323L mutation [IC50 = 11.37 μM]; Delta, RdRpP323L; G671S mutation [IC50 = 13.23 μM]) remain susceptible to GOS [24]. Furthermore, GOS exerted significant suppressive effects on the SARS-CoV-2 Delta variant in Vero E6 cells (EC50 = 0.23 μM, SI = 157.3). In addition to SARS-CoV-2, GOS exhibits broad-spectrum antiviral effects in vitro, including against porcine epidemic diarrhea virus (PEDV, alpha-CoV, EC50 = 0.99 μM, SI = 36.55), SADS-CoV (alpha-CoV, EC50 = 2.55 μM, SI = 14.19), infectious bronchitis virus (IBV, gamma-CoV, EC50 = 1.02 μM, SI = 35.47), and porcine deltacoronavirus (PDCoV, delta-CoV, EC50 = 1.06 μM, SI = 19.35), suggesting that GOS can act as a pan-coronavirus inhibitor [24]. A molecular docking model revealed that GOS can recognize diverse coronaviruses by targeting the highly conserved RdRp. The binding energies of GOS with RdRps of PEDV, SADS-CoV, IBV, and PDCoV were −7.6, −8.4, −7.9, and 8.8 kcal/mol, respectively [24].
It is important to note that high concentrations of GOS may cause several side effects, including respiratory distress [103]. As the lung is the primary target tissue of SARS-CoV-2, maintaining an effective lung concentration of antiviral drugs is required. Previous studies have demonstrated that cells are more sensitive to (−)-GOS. Considering that the antiviral effect of GOS were not affected by its optical activity, the development of (+)-GOS-derived antiviral drugs may be promising for further lead optimization.

5. Other Natural Dietary Polyphenols Anti-SARS-CoV-2 Candidates in Development

Numerous studies, both in in vitro and in vivo, have shown that curcumin, resveratrol, and gossypol are beneficial for COVID-19 treatment via targeting of Mpro, the S protein, and RdRp of SARS-CoV-2 and its variants. In addition, other dietary polyphenols have shown promising potential for SARS-CoV-2 treatment and prevention that provide more options for controlling COVID-19 infection. As shown in Table 3, these polyphenols can inhibit SARS-CoV-2 replication and demonstrate strong in vitro activity, but additional in-depth research and further optimization are still required.

6. Conclusions and Future Directions

The COVID-19 pandemic highlights the need for novel strategies to rapidly develop therapies and vaccines against emerging viral diseases. Natural dietary polyphenols (e.g., curcumin, resveratrol, and gossypol) are essential complements to vaccines and small-molecule therapies and have significant potential for use in the prevention and therapy of SARS-CoV-2 and emerging variants. However, some limitations (e.g., poor stability, weak oral bioavailability, short half-life, and lack of target specificity) cannot be ignored. Multiple measures must be considered if dietary polyphenols are to be used to provide broad-spectrum or universal protection against emerging variants.
First, developing personal protective equipment (e.g., reusable, antiviral, and antibacterial masks) containing natural dietary polyphenols is a promising inexpensive and convenient option for preparing for future pandemics. For example, De Maio et al. [54] fabricated personal protective equipment coated with graphene and curcumin. This graphene/curcumin-based antiviral coating interacted with the SARS-CoV-2 surface, trapping the virus, thus preventing further transmission. Notably, the combination of two antimicrobials could limit the emergence and spread of resistant SARS-CoV-2 strains. In parallel, Rao et al. [56] developed a biocompatible curcumin-incorporated composite membrane that exhibited excellent gas permeability (3423.6 m3/[m2·h·kPa]), a high ultrafine particle rejection rate (>98.79%), and good SARS-CoV-2 capture and kill efficiency (99.9% for 5 min). The identification of efficient ways to deliver polyphenols to the site of infection (mainly targets respiratory cells) is critical for future drug development. Optimized delivery devices provide new options for delivering dietary anti-SARS-CoV-2 polyphenols.
Second, several measures (e.g., nanotechnology, combination therapies, and lead optimization) must be considered to develop dietary polyphenol-based next-generation therapeutic agents with improved pharmacokinetic, resistance, and adverse event profiles. For example, spherical resveratrol-loaded nano-bilosomes (F5) exhibited a higher activity (IC50 of 0.24 μg/mL for F5 and 1.6 μg/mL for resveratrol) and an improved safety profile (SI of 139.5 for F5 and 2.9 for resveratrol) [91]. The availability of curcumin-loaded inhalable nanocellulose (nanocellulose/polyvinyl alcohol/curcumin), with its improved water solubility (313.61 mg/L) and high loading capacity (8.90 mg/g) is also key for future development [53]. Importantly, inhaled nanomedicines should be made with food-grade materials and should not affect normal respiratory functions. Regarding combination therapies, the literature shows that a specific combination of plant extracts and micronutrients (including resveratrol, curcumin, quercetin, vitamin C, theaflavin, naringenin, and broccoli extract) exert significant inhibitory effects against SARS-CoV-2 and its Alpha, Beta, Gamma, Delta, Kappa, Mu, and Omicron variants in vitro via pleiotropic mechanisms, including the targeting of viral RdRp, furin, and cathepsin L activity [81,82]. Lead optimization provides another promising strategy for further drug development. For instance, bisdemethoxycurcumin, a natural demethoxy analog of curcumin, can effectively inhibit SARS-CoV-2 replication by targeting nucleocapsid protein with an EC50 value of 1.64 µM and with high selectivity (SI = 15.24) [60]. Nanotechnology, combination therapies, and lead optimization may be potential strategies to solve this problem, but further research and optimization are required.
Third, more validation studies with high-quality clinical data for natural dietary polyphenols are urgently needed to assess their effects and potential. Although some preliminary clinical trial results have demonstrated their potential, the sample sizes are small, and further validation is required. We must gain an improved understanding of the metabolism of dietary polyphenols in the human body and further evaluate the potential risks that may arise from their use. At the same time, individual differences, dose adjustments, and possible adverse reactions should also be considered in clinical trials. Nonetheless, we believe that dietary polyphenols can be regarded as a first-line option for managing SARS-CoV-2 infection via well-formulated in-depth research and rigorous monitoring.

Author Contributions

Z.W.: conceptualization, data curation, writing original draft, writing—review and editing, visualization, and funding acquisition. X.-q.S.: data curation, review, and visualization. W.X., S.L. and H.Z.: Data curation. 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 National Natural Science Foundation of China (No. 22309103, L.Y.), the Shandong Provincial Natural Science Foundation (No. ZR2022MH162, Z.W.; ZR2022QE202, L.Y.), and the PhD Research Start-up Foundation of Qufu Normal University (No. 614901, L.Y.; 615201, Z.W.).

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 interest or personal relationship that could have appeared to influence the work reported in this paper.

References

  1. Li, G.; Hilgenfeld, R.; Whitley, R.; De Clercq, E. Therapeutic strategies for COVID-19: Progress and lessons learned. Nat. Rev. Drug Discov. 2023, 22, 449–475. [Google Scholar] [CrossRef] [PubMed]
  2. 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] [PubMed]
  3. Firouzabadi, N.; Ghasemiyeh, P.; Moradishooli, F.; Mohammadi-Samani, S. Update on the effectiveness of COVID-19 vaccines on different variants of SARS-CoV-2. Int. Immunopharmacol. 2023, 117, 109968. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, Z.; Yang, L. Chinese herbal medicine: Fighting SARS-CoV-2 infection on all fronts. J. Ethnopharmacol. 2021, 270, 113869. [Google Scholar] [CrossRef]
  5. Houeze, E.A.; Wang, Y.; Zhou, Q.; Zhang, H.; Wang, X. Comparison study of Beninese and Chinese herbal medicines in treating COVID-19. J. Ethnopharmacol. 2023, 308, 116172. [Google Scholar] [CrossRef]
  6. Wang, Z.; Yang, L. GS-5734: A Potentially Approved Drug by FDA against SARS-CoV-2. New J. Chem. 2020, 44, 12417–12429. [Google Scholar] [CrossRef]
  7. Yu, B.; Chang, J. The first Chinese oral anti-COVID-19 drug Azvudine launched. Innovation 2022, 3, 100321. [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. Syed, Y.Y. Molnupiravir: First approval. Drugs 2022, 82, 455–460. [Google Scholar] [CrossRef]
  10. Wang, Z.; Yang, L. In the age of Omicron variant: Paxlovid raises new hopes of COVID-19 recovery. J. Med. Virol. 2022, 94, 1766–1767. [Google Scholar] [CrossRef]
  11. Wise, J. COVID-19: WHO declares end of global health emergency. BMJ 2023, 381, 1041. [Google Scholar] [CrossRef]
  12. Ma, K.C.; Shirk, P.; Lambrou, A.S.; Hassell, N.; Zheng, X.Y.; Payne, A.B.; Ali, A.R.; Batra, D.; Caravas, J.; Chau, R.; et al. Genomic Surveillance for SARS-CoV-2 Variants: Circulation of Omicron Lineages-United States, January 2022–May 2023. MMWR Morb. Mortal. Wkly. Rep. 2023, 72, 651–656. [Google Scholar] [CrossRef]
  13. Kaku, C.I.; Starr, T.N.; Zhou, P.; Dugan, H.L.; Khalifé, P.; Song, G.; Champney, E.R.; Mielcarz, D.W.; Geoghegan, J.C.; Burton, D.R.; et al. Evolution of antibody immunity following omicron BA.1 breakthrough infection. Cell Rep. 2023, 42, 112443. [Google Scholar] [CrossRef]
  14. 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] [CrossRef] [PubMed]
  15. Wang, Z.; Yang, L. The Therapeutic Potential of Natural Dietary Flavonoids against SARS-CoV-2 Infection. Nutrients 2023, 15, 3443. [Google Scholar] [CrossRef] [PubMed]
  16. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Z.; Yang, L.; Li, Y.; Song, S.; Qu, J.; He, R.; Ren, S.; Gong, P. An activatable, carrier-free, triple-combination nanomedicine for ALK/EGFR-mutant non-small cell lung cancer highly permeable targeted chemotherapy. New J. Chem. 2022, 46, 17673–17677. [Google Scholar] [CrossRef]
  18. 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]
  19. Czlapka-Matyasik, M.; Gramza-Michalowska, A. The total dietary antioxidant capacity, its seasonal variability, and dietary sources in cardiovascular patients. Antioxidants 2023, 12, 292. [Google Scholar] [CrossRef]
  20. Wang, Z.L.; Yang, L.Y.; Yang, X.W.; Zhang, X.H. Advances in the first total synthesis of natural flavonoids. Synthetic. Commun. 2013, 23, 3093–3114. [Google Scholar] [CrossRef]
  21. Kicker, E.; Tittel, G.; Schaller, T.; Pferschy-Wenzig, E.M.; Zatloukal, K.; Bauer, R. SARS-CoV-2 neutralizing activity of polyphenols in a special green tea extract preparation. Phytomedicine 2022, 98, 153970. [Google Scholar] [CrossRef] [PubMed]
  22. Bormann, M.; Alt, M.; Schipper, L.; van de Sand, L.; Le-Trilling, V.T.K.; Rink, L.; Heinen, N.; Madel, R.J.; Otte, M.; Wuensch, K.; et al. Turmeric Root and Its Bioactive Ingredient Curcumin Effectively Neutralize SARS-CoV-2 In Vitro. Viruses 2021, 13, 1914. [Google Scholar] [CrossRef]
  23. Yang, M.; Wei, J.; Huang, T.; Lei, L.; Shen, C.; Lai, J.; Yang, M.; Liu, L.; Yang, Y.; Liu, G. Resveratrol inhibits the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in cultured Vero cells. Phytother. Res. 2021, 35, 1127–1129. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, W.; Li, W.; Wen, Z.; Wang, C.; Liu, W.; Zhang, Y.; Liu, J.; Ding, T.; Shuai, L.; Zhong, G.; et al. Gossypol Broadly Inhibits Coronaviruses by Targeting RNA-Dependent RNA Polymerases. Adv. Sci. 2022, 9, 2203499. [Google Scholar] [CrossRef] [PubMed]
  25. Mehany, T.; Khalifa, I.; Barakat, H.; Althwab, S.A.; Alharbi, Y.M.; El-Sohaimy, S. Polyphenols as Promising Biologically Active Substances for Preventing SARS-CoV-2: A Review with Research Evidence and Underlying Mechanisms. Food Biosci. 2021, 40, 100891. [Google Scholar] [CrossRef]
  26. 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]
  27. Purushothaman, A.; Rose, K.T.; Jacob, J.M.; Varatharaj, R.; Shashikala, K.; Janardanan, D. Curcumin analogues with improved antioxidant properties: A theoretical exploration. Food Chem. 2022, 373, 131499. [Google Scholar] [CrossRef]
  28. Hosseini-Zare, M.S.; Sarhadi, M.; Zarei, M.; Thilagavathi, R.; Selvam, C. Synergistic effects of curcumin and its analogs with other bioactive compounds: A comprehensive review. Eur. J. Med. Chem. 2021, 210, 113072. [Google Scholar] [CrossRef]
  29. Zhang, D.; Yang, Y.; Yao, B.; Hu, T.; Ma, Z.; Shi, W.; Ye, Y. Curcumin inhibits Aspergillus flavus infection and aflatoxin production possibly by inducing ROS burst. Food Res. Int. 2023, 167, 112646. [Google Scholar] [CrossRef]
  30. Wu, S.; Rao, G.; Wang, R.; Pang, Q.; Zhang, X.; Huang, R.; Li, T.; Tang, Z.; Hu, L. The neuroprotective effect of curcumin against ATO triggered neurotoxicity through Nrf2 and NF-κB signaling pathway in the brain of ducks. Ecotoxicol. Environ. Saf. 2021, 228, 112965. [Google Scholar] [CrossRef]
  31. Ming, T.; Tao, Q.; Tang, S.; Zhao, H.; Yang, H.; Liu, M.; Ren, S.; Xu, H. Curcumin: An epigenetic regulator and its application in cancer. Biomed. Pharmacother. 2022, 156, 113956. [Google Scholar] [CrossRef] [PubMed]
  32. Das, R.P.; Gandhi, V.V.; Verma, G.; Ajish, J.K.; Singh, B.G.; Kunwar, A. Gelatin-lecithin-F127 gel mediated self-assembly of curcumin vesicles for enhanced wound healing. Int. J. Biol. Macromol. 2022, 210, 403–414. [Google Scholar] [CrossRef] [PubMed]
  33. Mounce, B.C.; Cesaro, T.; Carrau, L.; Vallet, T.; Vignuzzi, M. Curcumin inhibits Zika and chikungunya virus infection by inhibiting cell binding. Antivir. Res. 2017, 142, 148–157. [Google Scholar] [CrossRef] [PubMed]
  34. Kumari, N.; Kulkarni, A.A.; Lin, X.; McLean, C.; Ammosova, T.; Ivanov, A.; Hipolito, M.; Nekhai, S.; Nwulia, E. Inhibition of HIV-1 by curcumin A, a novel curcumin analog. Drug Des. Dev. 2015, 9, 5051–5060. [Google Scholar]
  35. Ryu, Y.B.; Park, S.J.; Kim, Y.M.; Lee, J.Y.; Seo, W.D.; Chang, J.S.; Park, K.H.; Rho, M.C.; Lee, W.S. SARS-CoV 3CLpro inhibitory effects of quinone-methide triterpenes from Tripterygium regelii. Bioorg. Med. Chem. Lett. 2010, 20, 1873–1876. [Google Scholar] [CrossRef]
  36. Chai, Y.S.; Chen, Y.Q.; Lin, S.H.; Xie, K.; Wang, C.J.; Yang, Y.Z.; Xu, F. Curcumin regulates the differentiation of naïve CD4+ T cells and activates IL-10 immune modulation against acute lung injury in mice. Biomed. Pharmacother. 2020, 125, 109946. [Google Scholar] [CrossRef]
  37. Miao, Y.M.; Zhang, Y.J.; Qiao, S.M.; Xia, Y.F.; Wei, Z.F.; Dai, Y. Oral administration of curcumin ameliorates pulmonary fibrosis in mice through 15d-PGJ2-mediated induction of hepatocyte growth factor in the colon. Acta Pharmacol. Sin. 2021, 42, 422–435. [Google Scholar] [CrossRef]
  38. Wu, Y.; Chen, Y.; Li, Q.; Ye, X.; Guo, X.; Sun, L.; Zou, J.; Shen, Y.; Mao, Y.; Li, C.; et al. Tetrahydrocurcumin alleviates allergic airway inflammation in asthmatic mice by modulating the gut microbiota. Food Funct. 2021, 12, 6830–6840. [Google Scholar] [CrossRef]
  39. Liu, Y.; Ma, Y.; Xue, L.; Guan, W.; Wang, Y. Pulmonary multidrug codelivery of curcumin nanosuspensions and ciprofloxacin with N-acetylcysteine for lung infection therapy. J. Drug Deliv. Sci. Technol. 2023, 84, 104474. [Google Scholar] [CrossRef]
  40. Li, Q.; Sun, J.; Mohammadtursun, N.; Wu, J.; Dong, J.; Li, L. Curcumin inhibits cigarette smoke-induced inflammation via modulating the PPARγ-NF-κB signaling pathway. Food Funct. 2019, 10, 7983–7994. [Google Scholar] [CrossRef]
  41. Dadhaniya, P.; Patel, C.; Muchhara, J.; Bhadja, N.; Mathuria, N.; Vachhani, K.; Soni, M.G. Safety assessment of a solid lipid curcumin particle preparation: Acute and subchronic toxicity studies. Food Chem. Toxicol. 2011, 49, 1834–1842. [Google Scholar] [CrossRef] [PubMed]
  42. Kunnumakkara, A.B.; Harsha, C.; Banik, K.; Vikkurthi, R.; Sailo, B.L.; Bordoloi, D.; Gupta, S.C.; Aggarwal, B.B. Is curcumin bioavailability a problem in humans: Lessons from clinical trials. Expert Opin. Drug Metab. Toxicol. 2019, 15, 705–733. [Google Scholar] [CrossRef] [PubMed]
  43. Saber-Moghaddam, N.; Salari, S.; Hejazi, S.; Amini, M.; Taherzadeh, Z.; Eslami, S.; Rezayat, S.M.; Jaafari, M.R.; Elyasi, S. Oral nano-curcumin formulation efficacy in management of mild to moderate hospitalized coronavirus disease-19 patients: An open label nonrandomized clinical trial. Phytother. Res. 2021, 35, 2616–2623. [Google Scholar] [CrossRef]
  44. Soni, V.K.; Mehta, A.; Ratre, Y.K.; Tiwari, A.K.; Amit, A.; Singh, R.P.; Sonkar, S.C.; Chaturvedi, N.; Shukla, D.; Vishvakarma, N.K. Curcumin, a traditional spice component, can hold the promise against COVID-19? Eur. J. Pharmacol. 2020, 886, 173551. [Google Scholar] [CrossRef] [PubMed]
  45. Zupin, L.; Fontana, F.; Clemente, L.; Borelli, V.; Ricci, G.; Ruscio, M.; Crovella, S. Optimization of Anti-SARS-CoV-2 Treatments Based on Curcumin, Used Alone or Employed as a Photosensitizer. Viruses 2022, 14, 2132. [Google Scholar] [CrossRef] [PubMed]
  46. Marín-Palma, D.; Tabares-Guevara, J.H.; Zapata-Cardona, M.I.; Flórez-Álvarez, L.; Yepes, L.M.; Rugeles, M.T.; Zapata-Builes, W.; Hernandez, J.C.; Taborda, N.A. Curcumin inhibits in vitro SARS-CoV-2 infection in Vero E6 cells through multiple antiviral mechanisms. Molecules 2021, 26, 6900. [Google Scholar] [CrossRef]
  47. Bahun, M.; Jukič, M.; Domen, O.; Kranjc, L.; Bajc, G.; Butala, M.; Bozovičar, K.; Bratkovič, T.; Podlipnik, C.; Ulrih, N.P. Inhibition of the SARS-CoV-2 3CLpro main protease by plant polyphenols. Food Chem. 2022, 373, 131594. [Google Scholar] [CrossRef]
  48. Ujjan, I.D.; Khan, S.; Nigar, R.; Ahmed, H.; Ahmad, S.; Khan, A. The possible therapeutic role of curcumin and quercetin in the early-stage of COVID-19—Results from a pragmatic randomized clinical trial. Front. Nutr. 2023, 9, 1023997. [Google Scholar] [CrossRef]
  49. Khan, A.; Iqtadar, S.; Mumtaz, S.U.; Heinrich, M.; Pascual-Figal, D.A.; Livingstone, S.; Abaidullah, S. Oral co-supplementation of curcumin, quercetin, and vitamin D3 as an adjuvant therapy for mild to moderate symptoms of COVID-19—Results from a pilot open-label, randomized controlled trial. Front. Pharmacol. 2022, 13, 898062. [Google Scholar] [CrossRef]
  50. Hellou, E.; Mohsin, J.; Elemy, A.; Hakim, F.; Mustafa-Hellou, M.; Hamoud, S. Effect of ArtemiC in patients with COVID-19: A phase II prospective study. J. Cell. Mol. Med. 2022, 26, 3281–3289. [Google Scholar] [CrossRef]
  51. Suresh, K.; Nangia, A. Curcumin: Pharmaceutical solids as a platform to improve solubility and bioavailability. CrystEngComm 2018, 20, 3277–3296. [Google Scholar] [CrossRef]
  52. Anand, P.; Kunnumakkara, A.B.; Newman, R.A.; Aggarwal, B.B. Bioavailability of curcumin: Problems and promises. Mol. Pharm. 2007, 4, 807–818. [Google Scholar] [CrossRef]
  53. Gunathilake, T.M.S.U.; Ching, Y.C.; Uyama, H.; Hai, N.D.; Chuah, C.H. Enhanced curcumin loaded nanocellulose: A possible inhalable nanotherapeutic to treat COVID-19. Cellulose 2022, 29, 1821–1840. [Google Scholar] [CrossRef] [PubMed]
  54. De Maio, F.; Santarelli, G.; Palmieri, V.; Perini, G.; Salustri, A.; Palucci, I.; Carpini, G.D.; Augello, A.; Sanguinetti, M.; De Spirito, M.; et al. Graphene-curcumin coatings resistant to SARS-CoV-2 and mycobacteria for the production of personal protective equipment. J. Nat. Fibers 2023, 20, 2156020. [Google Scholar] [CrossRef]
  55. Lu, T.; Cui, J.; Qu, Q.; Wang, Y.; Zhang, J.; Xiong, R.; Ma, W.; Huang, C. Multistructured electrospun nanofibers for air filtration: A review. ACS Appl. Mater. Interfaces 2021, 13, 23293–23313. [Google Scholar] [CrossRef] [PubMed]
  56. Rao, Y.; Feng, S.; Low, Z.X.; Wu, J.; Ju, S.; Zhong, Z.; Xing, W. Biocompatible curcumin coupled nanofibrous membrane for pathogens sterilization and isolation. J. Membr. Sci. 2022, 661, 120885. [Google Scholar] [CrossRef]
  57. Sharma, V.K.; Prateeksha; Singh, S.P.; Singh, B.N.; Rao, C.V.; Barik, S.K. Nanocurcumin potently inhibits SARS-CoV-2 spike protein-induced cytokine storm by deactivation of MAPK/NF-κB signaling in epithelial cells. ACS Appl. Bio Mater. 2022, 5, 483–491. [Google Scholar] [CrossRef]
  58. Khan, N.; Afghah, Z.; Baral, A.; Geiger, J.D.; Chen, X. Dimethoxycurcumin acidifies endolysosomes and inhibits SARS-CoV-2 entry. Front. Virol. 2022, 2, 923018. [Google Scholar] [CrossRef]
  59. Archana, V.P.; Armaković, S.J.; Armaković, S.; Celik, I.; Bhagyasree, J.B.; Babu, K.D.; Rudrapal, M.; Divya, I.S.; Pillai, R.R. Exploring the structural, photophysical and optoelectronic properties of a diaryl heptanoid curcumin derivative and identification as a SARS-CoV-2 inhibitor. J. Mol. Struct. 2023, 1281, 135110. [Google Scholar] [CrossRef]
  60. Dhaka, P.; Singh, A.; Choudhary, S.; Kumar, P.; Sharma, G.K.; Tomar, S. Discovery of anti-SARS-CoV-2 molecules using structure-assisted repurposing approach targeting N-protein. bioRxiv 2022. [Google Scholar] [CrossRef]
  61. Jin, S.; Pang, Q.; Yang, H.; Diao, X.; Shan, A.; Feng, X. Effects of dietary resveratrol supplementation on the chemical composition, oxidative stability and meat quality of ducks (Anas platyrhynchos). Food Chem. 2021, 363, 130263. [Google Scholar] [CrossRef]
  62. Dhir, H.; Choudhury, M.; Patil, K.; Cheung, C.; Bodlak, A.; Pardo, D.; Adams, A.; Travaglino, S.; Rojas, J.A.; Pai, S.B. Interception of signaling circuits of esophageal adenocarcinoma cells by resveratrol reveals molecular and immunomodulatory signatures. Cancers 2021, 13, 5811. [Google Scholar] [CrossRef] [PubMed]
  63. Hao, Y.; Shao, L.; Hou, J.; Zhang, Y.; Ma, Y.; Liu, J.; Xu, C.; Chen, F.; Cao, L.H.; Ping, Y. Resveratrol and Sir2 reverse sleep and memory defects induced by amyloid precursor protein. Neurosci. Bull. 2023, 39, 1117–1130. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, Y.; Fu, W.; Wei, K.; Liu, L.; Wu, S.; Tang, W. The therapeutic efficacy of resveratrol for acute lung injury—A meta–analysis of preclinical trials. Front. Pharmacol. 2022, 13, 963245. [Google Scholar] [CrossRef] [PubMed]
  65. Xue, R.; Gao, S.; Zhang, Y.; Cui, X.; Mo, W.; Xu, J.; Yao, M. A meta-analysis of resveratrol protects against cerebral ischemia/reperfusion injury: Evidence from rats studies and insight into molecular mechanisms. Front. Pharmacol. 2022, 13, 988836. [Google Scholar] [CrossRef] [PubMed]
  66. Yu, D.; Xiong, J.; Gao, Y.; Li, J.; Zhu, D.; Shen, X.; Sun, L.; Wang, X. Resveratrol activates PI3K/AKT to reduce myocardial cell apoptosis and mitochondrial oxidative damage caused by myocardial ischemia/reperfusion injury. Acta Histochem. 2021, 123, 151739. [Google Scholar] [CrossRef]
  67. Wang, Y.; Sun, X.; Han, X.; Sun, J.; Li, L.; Zhang, D.; Sun, G. Resveratrol improves hepatic ischemia-reperfusion injury by inhibiting neutrophils via the ERK signaling pathway. Biomed. Pharmacother. 2022, 160, 114358. [Google Scholar] [CrossRef]
  68. Shen, Y.; Jiang, Y.; Zhang, S.; Zou, J.; Gao, X.; Song, Y.; Zhang, Y.; Hu, Y.; Huang, Y.; Jiang, Q. The effect of dietary supplementation with resveratrol on growth performance, carcass and meat quality, blood lipid levels and ruminal microbiota in fattening goats. Foods 2022, 11, 598. [Google Scholar] [CrossRef]
  69. Palamara, A.T.; Nencioni, L.; Aquilano, K.; De Chiara, G.; Hernandez, L.; Cozzolino, F.; Ciriolo, M.R.; Garaci, E. Inhibition of influenza A virus replication by resveratrol. J. Infect. Dis. 2005, 191, 1719–1729. [Google Scholar] [CrossRef]
  70. Liu, T.; Zang, N.; Zhou, N.; Li, W.; Xie, X.; Deng, Y.; Ren, L.; Long, X.; Li, S.; Zhou, L.; et al. Resveratrol inhibits the TRIF-dependent pathway by upregulating sterile alpha and armadillo motif protein, contributing to anti-inflammatory effects after respiratory syncytial virus infection. J. Virol. 2014, 88, 4229–4236. [Google Scholar] [CrossRef]
  71. Horne, J.R.; Vohl, M.C. Biological plausibility for interactions between dietary fat, resveratrol, ACE2, and SARS-CoV illness severity. Am. J. Physiol.-Endoc. M. 2020, 318, E830–E833. [Google Scholar] [CrossRef] [PubMed]
  72. Zakaria, M.Y.; Zaki, I.; Alhomrani, M.; Alamri, A.S.; Abdulaziz, O.; Abourehab, M.A. Boosting the anti MERS-CoV activity and oral bioavailability of resveratrol via PEG-stabilized emulsomal nano-carrier: Factorial design, in-vitro and in-vivo assessments. Drug Deliv. 2022, 29, 3155–3167. [Google Scholar] [CrossRef] [PubMed]
  73. Pasquereau, S.; Nehme, Z.; Haidar Ahmad, S.; Daouad, F.; Van Assche, J.; Wallet, C.; Schwartz, C.; Rohr, O.; Morot-Bizot, S.; Herbein, G. Resveratrol inhibits HCoV-229E and SARS-CoV-2 coronavirus replication in vitro. Viruses 2021, 13, 354. [Google Scholar] [CrossRef]
  74. Shariq, M.; Malik, A.A.; Sheikh, J.A.; Hasnain, S.E.; Ehtesham, N.Z. Regulation of autophagy by SARS-CoV-2: The multifunctional contributions of ORF3a. J. Med. Virol. 2023, 95, e28959. [Google Scholar] [CrossRef] [PubMed]
  75. Miao, G.; Zhao, H.; Li, Y.; Ji, M.; Chen, Y.; Shi, Y.; Bi, Y.; Wang, P.; Zhang, H. ORF3a of the COVID-19 virus SARS-CoV-2 blocks HOPS complex-mediated assembly of the SNARE complex required for autolysosome formation. Dev. Cell 2021, 56, 427–442.e5. [Google Scholar] [CrossRef] [PubMed]
  76. Fam, M.S.; Sedky, C.A.; Turky, N.O.; Breitinger, H.G.; Breitinger, U. Channel activity of SARS-CoV-2 viroporin ORF3a inhibited by adamantanes and phenolic plant metabolites. Sci. Rep. 2023, 13, 5328. [Google Scholar] [CrossRef]
  77. 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]
  78. de Souza Andrade, M.M.; Leal, V.N.C.; Fernandes, I.G.; Gozzi-Silva, S.C.; Beserra, D.R.; Oliveira, E.A.; Teixeira, F.M.E.; Yendo, T.M.; Sousa, M.d.G.T.; Teodoro, W.R.; et al. Resveratrol downmodulates neutrophil extracellular trap (NET) generation by neutrophils in patients with severe COVID-19. Antioxidants 2022, 11, 1690. [Google Scholar] [CrossRef]
  79. Sheng, H.; Lin, G.; Zhao, S.; Li, W.; Zhang, Z.; Zhang, W.; Yun, L.; Yan, X.; Hu, H. Antifibrotic mechanism of piceatannol in bleomycin-induced pulmonary fibrosis in Mice. Front. Pharmacol. 2022, 13, 771031. [Google Scholar] [CrossRef]
  80. 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]
  81. Goc, A.; Niedzwiecki, A.; Ivanov, V.; Ivanova, S.; Rath, M. Inhibitory effects of specific combination of natural compounds against SARS-CoV-2 and its Alpha, Beta, Gamma, Delta, Kappa, and Mu variants. Eur. J. Microbiol. Immunol. 2021, 11, 87–94. [Google Scholar] [CrossRef] [PubMed]
  82. Goc, A.; Rath, M.; Niedzwiecki, A. Composition of naturally occurring compounds decreases activity of Omicron and SARS-CoV-2 RdRp complex. Eur. J. Microbiol. Immunol. 2022, 12, 39–45. [Google Scholar] [CrossRef] [PubMed]
  83. Chan, E.W.C.; Wong, C.W.; Tan, Y.H.; Foo, J.P.Y.; Wong, S.K.; Chan, H.T. Resveratrol and pterostilbene: A comparative overview of their chemistry, biosynthesis, plant sources and pharmacological properties. J. Appl. Pharm. Sci. 2019, 9, 124–129. [Google Scholar]
  84. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed]
  85. De Angelis, M.; Della-Morte, D.; Buttinelli, G.; Di Martino, A.; Pacifici, F.; Checconi, P.; Ambrosio, L.; Stefanelli, P.; Palamara, A.T.; Garaci, E.; et al. Protective role of combined polyphenols and micronutrients against influenza A virus and SARS-CoV-2 infection in vitro. Biomedicines 2021, 9, 1721. [Google Scholar] [CrossRef]
  86. Tietjen, I.; Cassel, J.; Register, E.T.; Zhou, X.Y.; Messick, T.E.; Keeney, F.; Lu, L.D.; Beattie, K.D.; Rali, T.; Tebas, P.; et al. The natural stilbenoid (–)-hopeaphenol inhibits cellular entry of SARS-CoV-2 USA-WA1/2020, B.1.1.7, and B.1.351 variants. Antimicrob. Agents Chemother. 2021, 65, e00772-21. [Google Scholar] [CrossRef]
  87. Gangadevi, S.; Badavath, V.N.; Thakur, A.; Yin, N.; De Jonghe, S.; Acevedo, O.; Jochmans, D.; Leyssen, P.; Wang, K.; Neyts, J.; et al. Kobophenol A inhibits binding of host ACE2 receptor with spike RBD domain of SARS-CoV-2, a lead compound for blocking COVID-19. J. Phys. Chem. Lett. 2021, 12, 1793–1802. [Google Scholar] [CrossRef]
  88. 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] [CrossRef]
  89. Wang, C.; Ye, X.; Ding, C.; Zhou, M.; Li, W.; Wang, Y.; You, Q.; Zong, S.; Peng, Q.; Duanmu, D.; et al. Two Resveratrol Oligomers Inhibit Cathepsin L Activity to Suppress SARS-CoV-2 Entry. J. Agric. Food Chem. 2023, 71, 5535–5546. [Google Scholar] [CrossRef]
  90. Wang, L.; Lai, C.; Li, D.; Luo, Z.; Liu, L.; Jiang, Y.; Li, L. Lecithin-Polysaccharide Self-Assembled Microspheres for Resveratrol Delivery. Antioxidants 2022, 11, 1666. [Google Scholar] [CrossRef]
  91. Zakaria, M.Y.; Abd El-Halim, S.M.; Beshay, B.Y.; Zaki, I.; Abourehab, M.A. ‘Poly phenolic phytoceutical loaded nano-bilosomes for enhanced caco-2 cell permeability and SARS-CoV-2 antiviral activity’: In-vitro and in silico studies. Drug Deliv. 2023, 30, 2162157. [Google Scholar] [CrossRef] [PubMed]
  92. 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] [PubMed]
  93. Wagner, T.A.; Cai, Y.; Bell, A.A.; Puckhaber, L.S.; Magill, C.; Duke, S.E.; Liu, J. RNAi suppression of CYP82D P450 hydroxylase, an enzyme involved in gossypol biosynthesis, enhances resistance to Fusarium wilt in cotton. J. Phytopathol. 2020, 168, 103. [Google Scholar] [CrossRef]
  94. Kumar, M.; Potkule, J.; Patil, S.; Saxena, S.; Patil, P.G.; Mageshwaran, V.; Punia, S.; Varghese, E.; Mahapatra, A.; Ashtaputre, N.; et al. Extraction of uultra-low gossypol protein from cottonseed: Characterization based on antioxidant activity, structural morphology and functional group analysis. LWT 2021, 140, 110692. [Google Scholar] [CrossRef]
  95. Gao, Y.; Tai, W.; Wang, N.; Li, X.; Jiang, S.; Debnath, A.K.; Du, L.; Chen, S. Identification of novel natural products as effective and broad-spectrum anti-Zika virus inhibitors. Viruses 2019, 11, 1019. [Google Scholar] [CrossRef] [PubMed]
  96. Du, R.L.; Chow, H.Y.; Chen, Y.W.; Chan, P.H.; Daniel, R.A.; Wong, K.Y. Gossypol acetate: A natural polyphenol derivative with antimicrobial activities against the essential cell division protein FtsZ. Front. Microbiol. 2023, 13, 1080308. [Google Scholar] [CrossRef] [PubMed]
  97. Mayer, M.; Berger, A.; Leischner, C.; Renner, O.; Burkard, M.; Böcker, A.; Noor, S.; Weiland, T.; Weiss, T.S.; Busch, C.; et al. Preclinical efficacy and toxicity analysis of the pan-histone deacetylase inhibitor gossypol for the therapy of colorectal cancer or hepatocellular carcinoma. Pharmaceuticals 2022, 15, 438. [Google Scholar] [CrossRef]
  98. Xu, J.; Zhu, G.Y.; Cao, D.; Pan, H.; Li, Y.W. Gossypol overcomes EGFR-TKIs resistance in non-small cell lung cancer cells by targeting YAP/TAZ and EGFRL858R/T790M. Biomed. Pharmacother. 2019, 115, 108860. [Google Scholar] [CrossRef]
  99. Huo, M.; Gao, R.; Jiang, L.; Cui, X.; Duan, L.; Deng, X.; Guan, S.; Wei, J.; Soromou, L.W.; Feng, H.; et al. Suppression of LPS-induced inflammatory responses by gossypol in RAW 264.7 cells and mouse models. Int. Immunopharmacol. 2013, 15, 442–449. [Google Scholar] [CrossRef]
  100. Judge, J.L.; Lacy, S.H.; Ku, W.Y.; Owens, K.M.; Hernady, E.; Thatcher, T.H.; Williams, J.P.; Phipps, R.P.; Sime, P.J.; Kottmann, R.M. The lactate dehydrogenase inhibitor gossypol inhibits radiation-induced pulmonary fibrosis. Radiat. Res. 2017, 188, 35–43. [Google Scholar] [CrossRef]
  101. Stipanovic, R.D.; Puckhaber, L.S.; Bell, A.A.; Percival, A.E. Occurrence of (+)- and (−)-gossypol in seed of wild species of cotton and in Gossypium hirsutum var. marie-galante (Watt) Hutchinson. J. Agric. Food Chem. 2005, 53, 6266–6271. [Google Scholar] [CrossRef] [PubMed]
  102. Yuan, J.; Zhou, M.; Xin, X.; Yao, J.; Chang, J. Comparison of the efficacy of gossypol acetate enantiomers in rats with uterine leiomyoma. J. Nat. Med. 2023, 77, 41–52. [Google Scholar] [CrossRef] [PubMed]
  103. Gadelha, I.C.N.; Fonseca, N.B.S.; Oloris, S.C.S.; Melo, M.M.; Soto-Blanco, B. Gossypol toxicity from cottonseed products. Sci. World J. 2014, 2014, 231635. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, M.; Liu, L.; Zhao, Y.; Cao, Y.; Zhu, Y.; Han, L.; Yang, Q.; Wang, Y.; Wang, C.; Zhang, H.; et al. Discovery and evaluation of active compounds from Xuanfei Baidu formula against COVID-19 via SARS-CoV-2 Mpro. Chin. Med. 2023, 18, 94. [Google Scholar] [CrossRef] [PubMed]
  105. Hu, Y.; Liu, M.; Qin, H.; Lin, H.; An, X.; Shi, Z.; Song, L.; Yang, X.; Fan, H.; Tong, Y. Artemether, Artesunate, Arteannuin B, Echinatin, Licochalcone B and andrographolide effectively inhibit SARS-CoV-2 and related viruses in vitro. Front. Cell. Infect. Microbiol. 2021, 11, 680127. [Google Scholar] [CrossRef]
  106. Yang, Y.; Yang, P.; Huang, C.; Wu, Y.; Zhou, Z.; Wang, X.; Wang, S. Inhibitory effect on SARS-CoV-2 infection of neferine by blocking Ca2+-dependent membrane fusion. J. Med. Virol. 2021, 93, 5825–5832. [Google Scholar] [CrossRef]
  107. Marahatha, R.; Shrestha, A.; Sharma, K.; Regmi, B.P.; Sharma, K.R.; Poudel, P.; Basnyat, R.C.; Parajuli, N. In silico study of alkaloids: Neferine and berbamine potentially inhibit the SARS-CoV-2 RNA-dependent RNA polymerase. J. Chem. 2022, 2022, e7548802. [Google Scholar] [CrossRef]
  108. Zhong, B.; Peng, W.; Du, S.; Chen, B.; Feng, Y.; Hu, X.; Lai, Q.; Liu, S.; Zhou, Z.W.; Fang, P.; et al. Oridonin inhibits SARS-CoV-2 by targeting its 3C-like protease. Small Sci. 2022, 13, 2100124. [Google Scholar] [CrossRef]
  109. Yin, X.; Chen, L.; Yuan, S.; Liu, L.; Gao, Z. A robust high-throughput fluorescent polarization assay for the evaluation and screening of SARS-CoV-2 fusion inhibitors. Bioorg. Chem. 2021, 116, 105362. [Google Scholar] [CrossRef]
  110. Zhang, D.; Hamdoun, S.; Chen, R.; Yang, L.; Ip, C.K.; Qu, Y.; Li, R.; Jiang, H.; Yang, Z.; Chung, S.K.; et al. Identification of natural compounds as SARS-CoV-2 entry inhibitors by molecular docking-based virtual screening with bio-layer interferometry. Pharm. Res. 2021, 172, 105820. [Google Scholar] [CrossRef]
  111. Yang, L.J.; Chen, R.H.; Hamdoun, S.; Coghi, P.; Ng, J.P.L.; Zhang, D.W.; Guo, X.; Xia, C.; Law, B.Y.K.; Wong, V.K.W. Corilagin prevents SARS-CoV-2 infection by targeting RBD-ACE2 binding. Phytomedicine 2021, 87, 153591. [Google Scholar] [CrossRef] [PubMed]
  112. Binette, V.; Côté, S.; Haddad, M.; Nguyen, P.T.; Bélanger, S.; Bourgault, S.; Ramassamy, C.; Gaudreault, R.; Mousseau, N. Corilagin and 1,3,6-tri-O-galloy-β-d-glucose: Potential inhibitors of SARS-CoV-2 variants. Phys. Chem. Chem. Phys. 2021, 23, 14873–14888. [Google Scholar] [CrossRef] [PubMed]
  113. Li, Q.; Yi, D.; Lei, X.; Zhao, J.; Zhang, Y.; Cui, X.; Xiao, X.; Jiao, T.; Dong, X.; Zhao, X.; et al. Corilagin inhibits SARS-CoV-2 replication by targeting viral RNA-dependent RNA polymerase. Acta Pharm. Sin. B 2021, 11, 1555–1567. [Google Scholar] [CrossRef] [PubMed]
  114. 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]
  115. David, A.B.; Diamant, E.; Dor, E.; Barnea, A.; Natan, N.; Levin, L.; Chapman, S.; Mimran, L.C.; Epstein, E.; Zichel, R.; et al. Identification of SARS-CoV-2 receptor binding inhibitors by in vitro screening of drug libraries. Molecules 2021, 26, 3213. [Google Scholar] [CrossRef] [PubMed]
  116. Du, R.; Cooper, L.; Chen, Z.; Lee, H.; Rong, L.; Cui, Q. Discovery of chebulagic acid and punicalagin as novel allosteric inhibitors of SARS-CoV-2 3CLpro. Antivir. Res. 2021, 190, 105075. [Google Scholar] [CrossRef]
  117. Wang, Z.; Wang, N.; Yang, L.; Song, X.Q. Bioactive natural products in COVID-19 therapy. Front. Pharmacol. 2022, 13, 926507. [Google Scholar] [CrossRef]
  118. Rudrapal, M.; Celik, I.; Khan, J.; Ansari, M.A.; Alomary, M.N.; Yadav, R.; Sharma, T.; Tallei, T.E.; Pasala, P.K.; Sahoo, R.K.; et al. Identification of bioactive molecules from Triphala (Ayurvedic herbal formulation) as potential inhibitors of SARS-CoV-2 main protease (Mpro) through computational investigations. J. King Saud Univ. Sci. 2022, 34, 101826. [Google Scholar] [CrossRef]
  119. 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]
  120. Mahmud, S.; Biswas, S.; Paul, G.K.; Mita, M.A.; Promi, M.M.; Afrose, S.; Hasan, M.R.; Zaman, S.; Uddin, M.S.; Dhama, K.; 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]
  121. Li, Y.T.; Yang, C.; Wu, Y.; Lv, J.J.; Feng, X.; Tian, X.; Zhou, Z.; Pan, X.; Liu, S.; Tian, L.W. Axial chiral binaphthoquinone and perylenequinones from the stromata of hypocrella bambusae are SARS-CoV-2 entry inhibitors. J. Nat. Prod. 2021, 84, 436–443. [Google Scholar] [CrossRef] [PubMed]
  122. Zhao, J.; Ma, Q.; Zhang, B.; Guo, P.; Wang, Z.; Liu, Y.; Meng, M.; Liu, A.; Yang, Z.; Du, G. Exploration of SARS-CoV-2 3CLpro inhibitors by virtual screening methods, FRET detection, and CPE assay. J. Chem. Inf. Model. 2021, 61, 5763–5773. [Google Scholar] [CrossRef] [PubMed]
  123. Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; et al. Structure of M(pro) from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289–293. [Google Scholar] [CrossRef]
  124. Das, R.; Habiba, S.U.; Dash, R.; Seo, Y.; Woo, J. Unveiling the Potentiality of Shikonin Derivatives Inhibiting SARS-CoV-2 Main Protease by Molecular Dynamic Simulation Studies. Int. J. Mol. Sci. 2023, 24, 3100. [Google Scholar] [CrossRef] [PubMed]
  125. Cui, J.; Jia, J. Discovery of juglone and its derivatives as potent SARS-CoV-2 main proteinase inhibitors. Eur. J. Med. Chem. 2021, 225, 113789. [Google Scholar] [CrossRef]
  126. Ma, Q.; Li, R.; Pan, W.; Huang, W.; Liu, B.; Xie, Y.; Wang, Z.; Li, C.; Jiang, H.; Huang, J.; et al. Phillyrin (KD-1) exerts anti-viral and anti-inflammatory activities against novel coronavirus (SARS-CoV-2) and human coronavirus 229E (HCoV-229E) by suppressing the nuclear factor kappa B (NF-κB) signaling pathway. Phytomedicine 2020, 78, 153296. [Google Scholar] [CrossRef]
  127. Yu, J.W.; Wang, L.; Bao, L.D. Exploring the active compounds of traditional Mongolian medicine in intervention of novel coronavirus (COVID-19) based on molecular docking method. J. Funct. Foods 2020, 71, 104016. [Google Scholar] [CrossRef]
  128. Su, H.X.; Yao, S.; Zhao, W.F.; Li, M.J.; Liu, J.; Shang, W.J.; Xie, H.; Ke, C.Q.; Hu, H.C.; Gao, M.N.; et al. Anti-SARS-CoV-2 activities in vitro of Shuanghuanglian preparations and bioactive ingredients. Acta. Pharmacol. Sin. 2020, 41, 1167–1177. [Google Scholar] [CrossRef]
  129. Bibi, S.; Khan, M.S.; El-Kafrawy, S.A.; Alandijany, T.A.; El-Daly, M.M.; Yousafi, Q.; Fatima, D.; Faizo, A.A.; Bajrai, L.H.; Azhar, E.I. Virtual screening and molecular dynamics simulation analysis of Forsythoside A as a plant-derived inhibitor of SARS-CoV-2 3CLpro. Saudi Pharm. J. 2022, 30, 979–1002. [Google Scholar] [CrossRef]
  130. Abdallah, H.M.; El-Halawany, A.M.; Sirwi, A.; El-Araby, A.M.; Mohamed, G.A.; Ibrahim, S.R.M.; Koshak, A.E.; Asfour, H.Z.; Awan, Z.A.; Elfaky, M.A. 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]
  131. Kanjanasirirat, P.; Suksatu, A.; Manopwisedjaroen, S.; Munyoo, B.; Tuchinda, P.; Jearawuttanakul, K.; Seemakhan, S.; Charoensutthivarakul, S.; Wongtrakoongate, P.; Rangkasenee, N.; et al. High-content screening of Thai medicinal plants reveals Boesenbergia rotunda extract and its component Panduratin A as anti-SARS-CoV-2 agents. Sci. Rep. 2020, 10, 19963. [Google Scholar] [CrossRef] [PubMed]
  132. Vergoten, G.; Bailly, C. Interaction of panduratin A and derivatives with the SARS-CoV-2 main protease (mpro): A molecular docking study. J. Biomol. Struct. Dyn. 2022, 41, 6834–6844. [Google Scholar] [CrossRef] [PubMed]
  133. 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]
  134. 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]
  135. 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]
  136. Delcanale, P.; Uriati, E.; Mariangeli, M.; Mussini, A.; Moreno, A.; Lelli, D.; Cavanna, L.; Bianchini, P.; Diaspro, A.; Abbruzzetti, S.; et al. The interaction of hypericin with SARS-CoV-2 reveals a multimodal antiviral activity. ACS Appl. Mater. Interfaces 2022, 14, 14025–14032. [Google Scholar] [CrossRef]
  137. 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 (M(pro)) of SARS-CoV-2 and reduces its count in vitro. Sci. Rep. 2022, 12, 13146. [Google Scholar] [CrossRef]
  138. 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]
  139. 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]
  140. Akdad, M.; Moujane, S.; Bouadid, I.; Benlyas, M.; Eddouks, M. Phytocompounds from Anvillea radiata as promising anti-COVID-19 drugs: In silico studies and in vivo safety assessment. J. Environ. Sci. Health A 2021, 56, 1512–1523. [Google Scholar] [CrossRef]
  141. Kim, Y.S.; Chung, H.-S.; Noh, S.G.; Lee, B.; Chung, H.Y.; Choi, J.-G. Geraniin inhibits the entry of SARS-CoV-2 by blocking the interaction between spike protein RBD and human ACE2 receptor. Int. J. Mol. Sci. 2021, 22, 8604. [Google Scholar] [CrossRef] [PubMed]
  142. Yu, W.D.; Jin, Q.Y.; Zeng, M.S.; Liu, J.Y.; Xu, P.P. Geraniin as a potential inhibitor of SARS-CoV-2 3CLpro. Nat. Prod. Res. 2022, 36, 6060–6063. [Google Scholar] [CrossRef] [PubMed]
  143. Boadu, A.; Agoni, C.; Karpoormath, R.; Soliman, M.; Nlooto, M. Repurposing antiviral phytochemicals from the leaf extracts of Spondias mombin (Linn) towards the identification of potential SARS-CoV-2 inhibitors. Sci. Rep. 2022, 12, 10896. [Google Scholar] [CrossRef] [PubMed]
  144. Hu, S.; Wang, J.; Zhang, Y.; Bai, H.; Wang, C.; Wang, N.; He, L. Three salvianolic acids inhibit 2019-nCoV spike pseudovirus viropexis by binding to both its RBD and receptor ACE2. J. Med. Virol. 2021, 93, 3143–3151. [Google Scholar] [CrossRef]
  145. Ibrahim, M.A.A.; Abdelrahman, A.H.M.; Hussien, T.A.; Badr, E.A.A.; Mohamed, T.A.; El-Seedi, H.R.; Pare, P.W.; Efferth, T.; Hegazy, M.F. In silico drug discovery of major metabolites from spices as SARS-CoV-2 main protease inhibitors. Comput. Biol. Med. 2020, 126, 104046. [Google Scholar] [CrossRef]
  146. Elmaaty, A.A.; Darwish, K.M.; Khattab, M.; Elhady, S.S.; Salah, M.; Hamed, M.I.A.; Al-Karmalawy, A.A.; Saleh, M.M. In a search for potential drug candidates for combating COVID-19: Computational study revealed salvianolic acid B as a potential therapeutic targeting 3CLpro and spike proteins. J. Biomol. Struct. Dyn. 2022, 40, 8866–8893. [Google Scholar] [CrossRef]
  147. Yang, C.; Pan, X.; Xu, X.; Cheng, C.; Huang, Y.; Li, L.; Jiang, S.; Xu, W.; Xiao, G.; Liu, S. Salvianolic acid C potently inhibits SARS-CoV-2 infection by blocking the formation of six-helix bundle core of spike protein. Signal Transduct. Target. Ther. 2020, 5, 220. [Google Scholar] [CrossRef]
  148. Elebeedy, D.; Elkhatib, W.F.; Kandeil, A.; Ghanem, A.; Kutkat, O.; Alnajjar, R.; Saleh, M.A.; El Maksoud, A.I.A.; Badawy, I.; Al-Karmalawy, A.A. Anti-SARS-CoV-2 activities of tanshinone IIA, carnosic acid, rosmarinic acid, salvianolic acid, baicalein, and glycyrrhetinic acid between computational and in vitro insidghts. RSC Adv. 2021, 11, 29267–29286. [Google Scholar] [CrossRef]
  149. Patel, U.; Desai, K.; Dabhi, R.C.; Maru, J.J.; Shrivastav, P.S. Bioprospecting phytochemicals of Rosmarinus officinalis L. for targeting SARS-CoV-2 main protease (Mpro): A computational study. J. Mol. Model. 2023, 29, 161. [Google Scholar] [CrossRef]
  150. He, C.L.; Huang, L.Y.; Wang, K.; Gu, C.J.; Hu, J.; Zhang, G.J.; Xu, W.; Xie, Y.H.; Tang, N.; Huang, A.L. Identification of bis-benzylisoquinoline alkaloids as SARS-CoV-2 entry inhibitors from a library of natural products. Signal Transduct. Target. Ther. 2021, 6, 131. [Google Scholar] [CrossRef]
  151. Khamto, N.; Utama, K.; Tateing, S.; Sangthong, P.; Rithchumpon, P.; Cheechana, N.; Saiai, A.; Semakul, N.; Punyodom, W.; Meepowpan, P. Discovery of natural bisbenzylisoquinoline analogs from the library of Thai traditional plants as SARS-CoV-2 3CLpro inhibitors: In silico molecular docking, molecular dynamics, and in vitro enzymatic activity. J. Chem. Inf. Model. 2023, 63, 2104–2121. [Google Scholar] [CrossRef] [PubMed]
  152. Tan, H.; Ma, C.; Wang, J. Invalidation of dieckol and 1,2,3,4,6-pentagalloylglucose (PGG) as SARS-CoV-2 main protease inhibitors and the discovery of PGG as a papain-like protease inhibitor. Med. Chem. Res. 2022, 31, 1147–1153. [Google Scholar] [CrossRef] [PubMed]
  153. Chiou, W.C.; Chen, J.C.; Chen, Y.T.; Yang, J.M.; Hwang, L.H.; Lyu, Y.S.; Yang, H.Y.; Huang, C. The inhibitory effects of PGG and EGCG against the SARS-CoV-2 3C-like protease. Biochem. Biophys. Res. Commun. 2022, 591, 130–136. [Google Scholar] [CrossRef] [PubMed]
  154. 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]
  155. 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]
  156. Kim, Y.S.; Kim, B.; Kwon, E.-B.; Chung, H.-S.; Choi, J.-G. Mulberrofuran G, a mulberry component, prevents SARS-CoV-2 infection by blocking the interaction between SARS-CoV-2 Spike protein S1 receptor-binding domain and human angiotensin-converting enzyme 2 receptor. Nutrients 2022, 14, 4170. [Google Scholar] [CrossRef]
  157. Tao, Y.; Qu, H.; Wang, S.; Yan, F.; Wang, C.; Zhang, M. Computational assessment of herbal medicine-derived compounds as potential inhibitors of SARS-CoV-2 main protease. J. Biomol. Struct. Dyn. 2022. [Google Scholar] [CrossRef]
  158. 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]
  159. Choe, J.; Har Yong, P.; Xiang Ng, Z. The efficacy of traditional medicinal plants in modulating the main protease of SARS-CoV-2 and cytokine storm. Chem. Biodiv. 2022, 19, e202200655. [Google Scholar] [CrossRef]
  160. Yan, G.; Li, D.; Lin, Y.; Fu, Z.; Qi, H.; Liu, X.; Zhang, J.; Si, S.; Chen, Y. Development of a simple and miniaturized sandwich-like fluorescence polarization assay for rapid screening of SARS-CoV-2 main protease inhibitors. Cell Biosci. 2021, 11, 199. [Google Scholar] [CrossRef]
  161. Aatif, M.; Muteeb, G.; Alsultan, A.; Alshoaibi, A.; Khelif, B.Y. Dieckol and its derivatives as potential inhibitors of SARS-CoV-2 spike protein (UK strain: VUI 202012/01): A computational study. Mar. Drugs 2021, 19, 242. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, S.C.; Chen, Y.; Wang, Y.C.; Wang, W.J.; Yang, C.S.; Tsai, C.L.; Hou, M.H.; Chen, H.F.; Shen, Y.C.; 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] [PubMed]
  163. Ghamry, H.I.; Belal, A.; El-Ashrey, M.K.; Tawfik, H.O.; Alsantali, R.I.; Obaidullah, A.J.; El-Mansi, A.A.; Abdelrahman, D. Evaluating the ability of some natural phenolic acids to target the main protease and AAK1 in SARS-CoV-2. Sci. Rep. 2023, 13, 7357. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structures of curcumin, resveratrol, and gossypol. Gossypol exists as enantiomers due to the restricted rotation around the internal binaphthyl bond.
Figure 1. The chemical structures of curcumin, resveratrol, and gossypol. Gossypol exists as enantiomers due to the restricted rotation around the internal binaphthyl bond.
Nutrients 15 03885 g001
Figure 3. Chemical structure of resveratrol and its natural derivatives. Chemical structure of resveratrol, which can be isolated from several dietary sources such as grapes, peanuts, pistachios, blueberries, etc.; Chemical structures of piceatannol (hydroxylated analog) and pterostilbene (methoxylated analog); Chemical structures of trans-ε-viniferin (dimer), miyabenol C (trimer), and hopeaphenol (tetramer), vatalbinoside A (tetramer), vaticanol B (tetramer), and kobophenol A (tetramer).
Figure 3. Chemical structure of resveratrol and its natural derivatives. Chemical structure of resveratrol, which can be isolated from several dietary sources such as grapes, peanuts, pistachios, blueberries, etc.; Chemical structures of piceatannol (hydroxylated analog) and pterostilbene (methoxylated analog); Chemical structures of trans-ε-viniferin (dimer), miyabenol C (trimer), and hopeaphenol (tetramer), vatalbinoside A (tetramer), vaticanol B (tetramer), and kobophenol A (tetramer).
Nutrients 15 03885 g003
Figure 4. Possible mechanism of action of resveratrol and its derivatives for SARS-CoV-2 treatment. (a) Possible mechanism of action of resveratrol and its derivatives in the treatment of SARS-CoV-2 involving the inhibition of RBD/ACE2 binding. (b) A model showing how SARS-CoV-2 ORF3a impairs autophagosome maturation by disrupting the HOPS-mediated assembly of the SNARE complex. (Adapted with permission [75]. Copyright © 2020 Elsevier B.V.) (c) Resveratrol inhibits SARS-CoV-2 ORF3a. ORF3a, an essential contributor to infection and propagation of SARS-CoV-2, is a potential therapeutic target against COVID-19.
Figure 4. Possible mechanism of action of resveratrol and its derivatives for SARS-CoV-2 treatment. (a) Possible mechanism of action of resveratrol and its derivatives in the treatment of SARS-CoV-2 involving the inhibition of RBD/ACE2 binding. (b) A model showing how SARS-CoV-2 ORF3a impairs autophagosome maturation by disrupting the HOPS-mediated assembly of the SNARE complex. (Adapted with permission [75]. Copyright © 2020 Elsevier B.V.) (c) Resveratrol inhibits SARS-CoV-2 ORF3a. ORF3a, an essential contributor to infection and propagation of SARS-CoV-2, is a potential therapeutic target against COVID-19.
Nutrients 15 03885 g004
Figure 5. The chemical structures of gossypol and acetate gossypol.
Figure 5. The chemical structures of gossypol and acetate gossypol.
Nutrients 15 03885 g005
Figure 6. Schematic illustration of two gossypol (GOS) molecules acting as SARS-CoV-2 inhibitors via blocking the highly conserved RNA-dependent RNA polymerase. GOS inhibits RdRp activity by occupying the binding site for the RNA template [(−)-GOS, named GOS1] and primer [(+)-GOS, named GOS2]), consequentially inhibiting the catalytic activity of RdRp [24].
Figure 6. Schematic illustration of two gossypol (GOS) molecules acting as SARS-CoV-2 inhibitors via blocking the highly conserved RNA-dependent RNA polymerase. GOS inhibits RdRp activity by occupying the binding site for the RNA template [(−)-GOS, named GOS1] and primer [(+)-GOS, named GOS2]), consequentially inhibiting the catalytic activity of RdRp [24].
Nutrients 15 03885 g006
Table 1. Curcumin-related clinical trials based on a systematic search of ClinicalTrials.gov (https://clinicaltrials.gov/, accessed 15 August 2023).
Table 1. Curcumin-related clinical trials based on a systematic search of ClinicalTrials.gov (https://clinicaltrials.gov/, accessed 15 August 2023).
InterventionsPrincipal InvestigatorIdentifier (Year)ParticipantsDetails
Curcumin, quercetin, and
vitamin D3
Liaquat University Hospital
Sindh, Pakistan
NCT04603690 (2020)50Oral curcumin, quercetin, and vitamin D3 supplements for mild to moderate symptoms of COVID-19
Micellar ArtemiC, comprising curcumin, artemisinin, frankincense, and vitamin CMGC Pharmaceuticals d.o.oNCT04382040 (2020)50A phase II, controlled clinical study designed to evaluate the effect of ArtemiC in COVID-19 patients
Curcumin and palmitoylethanolamideArizona Biomedical Collaborative
Arizona, United States
NCT04912921 (2021)115Effect of palmitoylethanolamide on proinflammatory markers in adults diagnosed with COVID-19
Curcumin and quercetinKing Edward Medical University, Punjab, PakistanNCT05130671 (2021)50Nutritional supplementation of quercetin and curcumin for early mild symptoms of COVID-19
Nanoparticular CimertrA, comprising curcumin, artemisinin, boswellia, and vitamin CMGC Pharmaceuticals d.o.oNCT04802382 (2021)252A phase III clinical study to evaluate the effect of CimetrA in patients diagnosed with COVID-19
Curcumin, boswellia serrata, and ascorbic acidPhysioMetricsNCT05150782 (2021)32To examine the effect of a mixture of micellized curcumin, boswellia serrata, and ascorbic acid on patients with long COVID
Nanoparticular CimertA, comprising curcumin, boswellia, and vitamin CMGC Pharmaceuticals d.o.oNCT05037162 (2021)240A phase II, multi-center study in Israel, Brazil, Spain, and South Africa to evaluate the effect of CimetrA on COVID-19 patients
NASAFYTOL® Capsules, containing curcumin, turmeric extract, quercetin, and vitamin D3Tilman S.A.NCT04844658 (2021)51To evaluate the effect and safety of NASAFYTOL® on COVID-19-positive hospitalized patients
Nutritional powder (including Curcumin)Shanghai Tongji Hospital, Tongji University School of Medicine, ChinaNCT05629975 (2022)150Oral nutritional supplements in the treatment of elderly mild to moderate COVID-19
Table 2. Broad-spectrum antiviral activity of gossypol and related derivatives in vitro.
Table 2. Broad-spectrum antiviral activity of gossypol and related derivatives in vitro.
CompoundVirusEC50 or IC50 (μM)Therapeutic Indexes
Gossypol
(GOS)
SARS-CoV-2IC50 = 14.15 (RdRp)-
EC50 = 0.31 (Vero E6 cells)116.71
EC50 = 0.76 (Calu-3 cells)52.07
SARS-CoV-2 Delta variant (RdRpP323L; G671S mutation)IC50 = 13.23 μM (RdRp)-
EC50 = 0.23 (Vero E6 cells)157.3
SARS-CoV-2 Omicron variant (RdRpP323L mutation)IC50 = 11.37 (RdRp)-
PEDVEC50 = 0.99 (Vero E6 cells)36.55
SADS-CoVEC50 = 2.55 (Vero E6 cells)14.19
IBVEC50 = 1.02 (Vero E6 cells)35.47
PDCoVEC50 = 1.06 (Vero E6 cells)19.35
(−)-GOSSARS-CoV-2IC50 = 15.17 (RdRp)-
EC50 = 0.84 (Vero E6 cells)42.17
GOSAcSARS-CoV-2IC50 = 14.83 (RdRp)-
EC50 = 0.72 (Vero E6 cells)61.82
Table 3. Other natural dietary polyphenols for treating SARS-CoV-2 infection in vitro.
Table 3. Other natural dietary polyphenols for treating SARS-CoV-2 infection in vitro.
NameSpeciesStructureEC50 or IC50 (μM)Target or MechanismRefs.
Licochalcone BGlycyrrhiza uralensis FischNutrients 15 03885 i00115.53Mpro[104,105]
EchinatinGlycyrrhiza inflataNutrients 15 03885 i0027.86Mpro[104,105]
NeferineNelumbinis plumulaNutrients 15 03885 i0030.36Inhibiting Ca2+-dependent membrane fusion and suppressing virus entry; RdRp[106,107]
Salvianolic acid ASalvia miltiorrhizaNutrients 15 03885 i0042.49Mpro[108,109,110]
CorilaginPhmllanthi FructusNutrients 15 03885 i0050.13RBD-ACE2[111,112,113]
Ellagic acidPunica granatumNutrients 15 03885 i00611.8RBD-ACE2, Mpro[114,115]
PunicalaginPunica granatumNutrients 15 03885 i0076.19Mpro[116]
Chebulagic acidTerminalia chebula RetzNutrients 15 03885 i0089.76Mpro[116,117,118]
Epicatechin-3-O-gallateCamellia sinensis var. sinensisNutrients 15 03885 i0095.21Mpro[119,120]
Catechin-3-O-gallateSenegalia catechuNutrients 15 03885 i0102.98Mpro[119]
Hypocrellin AHypocrella bambusaeNutrients 15 03885 i0110.038S protein[121]
BinaphthoquinoneHypocrella bambusaeNutrients 15 03885 i0120.17S protein[121]
Shiraiachrome AHypocrella bambusaeNutrients 15 03885 i0130.12S protein[121]
(+)-ShikoninLithospermum erythrorhizonNutrients 15 03885 i0144.38Mpro[122]
ShikoninLithospermum erythrorhizonNutrients 15 03885 i01515.75Mpro[123,124,125]
PhillyrinForsythia suspensaNutrients 15 03885 i0161.13Inhibiting virus proliferation[126,127]
IsoforsythiasideForsythia suspensaNutrients 15 03885 i0175.85Mpro[128]
Forsythoside AForsythia suspensaNutrients 15 03885 i0183.18Mpro[128,129]
Forsythoside BForsythia suspensaNutrients 15 03885 i0192.88Mpro[128]
ActeosideLippia triphyllaNutrients 15 03885 i0200.043Mpro[130]
Panduratin ABoesenbergia rotundaNutrients 15 03885 i0210.078Mpro[131,132]
etc-pyrrolidinone C and DCamellia sinensisNutrients 15 03885 i0220.90Mpro[133]
PGHGPenthorum chinense PurshNutrients 15 03885 i0236.5Mpro, protein disulfide isomerase[134]
HypericinHypericum perforatum L.Nutrients 15 03885 i02420.3Mpro[135,136]
Theaflavin 3-gallateblack teaNutrients 15 03885 i02518.48Mpro, S protein[137,138]
Theaflavinblack teaNutrients 15 03885 i02622.22Mpro[137]
3,5-Dicaffeoylquinic acidHelichrysum bracteatumNutrients 15 03885 i0274.74Mpro[139,140]
simplexoside (piperitol-O-β-D-glucoside)Helichrysum bracteatumNutrients 15 03885 i0280.610Mpro[139]
GeraniinCaryocar brasilienseNutrients 15 03885 i0294.2Mpro, RBD-ACE2[141,142,143]
Salvianolic acid ASalvia miltiorrhizaNutrients 15 03885 i03011.31RBD-ACE2, Mpro[144,145]
Salvianolic acid BSalvia miltiorrhizaNutrients 15 03885 i0316.22RBD-ACE2, Mpro[144,146]
Salvianolic acid CSalvia miltiorrhizaNutrients 15 03885 i03210.14 RBD-ACE2[144,147]
Rosmarinic acidSalvia miltiorrhizaNutrients 15 03885 i0339.43S protein, Mpro[148,149]
LiensinineNelumbo nucifera Gaertn.Nutrients 15 03885 i0341.00Mpro[150,151]
Forsythoside IForsythia suspensaNutrients 15 03885 i0355.47Mpro[128]
Forsythoside HForsythia suspensaNutrients 15 03885 i03610.17Mpro[128]
1,2,3,4,6-pentagalloylglucose (PGG)Toona sinensisNutrients 15 03885 i0373.90Mpro[152,153]
Epicatechin gallateFagopyrum esculentumNutrients 15 03885 i03812.5Mpro[154,155]
Mulberrofuran GBombyx mori L.Nutrients 15 03885 i0391.55S Protein, Mpro[156,157]
GlabridinGlycyrrhiza glabraNutrients 15 03885 i0402.5Mpro[158,159]
DieckolEisenia bicyclisNutrients 15 03885 i0414.5Mpro, S protein[160,161]
Tannic acidGalla chinensisNutrients 15 03885 i0422.31Mpro, TMPRSS2[162,163]
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.; Song, X.-q.; Xu, W.; Lei, S.; Zhang, H.; Yang, L. Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nutrients 2023, 15, 3885. https://doi.org/10.3390/nu15183885

AMA Style

Wang Z, Song X-q, Xu W, Lei S, Zhang H, Yang L. Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nutrients. 2023; 15(18):3885. https://doi.org/10.3390/nu15183885

Chicago/Turabian Style

Wang, Zhonglei, Xian-qing Song, Wenjing Xu, Shizeng Lei, Hao Zhang, and Liyan Yang. 2023. "Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection" Nutrients 15, no. 18: 3885. https://doi.org/10.3390/nu15183885

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