Next Article in Journal
A Triboelectric Nanogenerator Based on Sodium Chloride Powder for Self-Powered Humidity Sensor
Next Article in Special Issue
Biocompatibility Testing of Liquid Metal as an Interconnection Material for Flexible Implant Technology
Previous Article in Journal
Ligand Effects on Intramolecular Configuration, Intermolecular Packing, and Optical Properties of Metal Nanoclusters
Previous Article in Special Issue
Targeting the “Sweet Side” of Tumor with Glycan-Binding Molecules Conjugated-Nanoparticles: Implications in Cancer Therapy and Diagnosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanotechnology Innovations to Enhance the Therapeutic Efficacy of Quercetin

by
Rúben G. R. Pinheiro
1,
Marina Pinheiro
1 and
Ana Rute Neves
1,2,*
1
LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
2
CQM—Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
*
Author to whom correspondence should be addressed.
Nanomaterials 2021, 11(10), 2658; https://doi.org/10.3390/nano11102658
Submission received: 6 September 2021 / Revised: 1 October 2021 / Accepted: 5 October 2021 / Published: 9 October 2021

Abstract

:
Quercetin is a flavonol present in many vegetables and fruits. Generally, quercetin can be found in aglycone and glycoside forms, mainly in leaves. The absorption of this compound occurs in the large and small intestine, where it suffers glucuronidation, sulfidation, and methylation to improve hydrophilicity. After metabolization, which occurs mainly in the gut, it is distributed throughout the whole organism and is excreted by feces, urine, and exhalation of carbon dioxide. Despite its in vitro cytotoxicity effects, in vivo studies with animal models ensure its safety. This compound can protect against cancer, cardiovascular diseases, chronic inflammation, oxidative stress, and neurodegenerative diseases due to its radical scavenging and anti-inflammatory properties. However, its poor bioavailability dampens the potential beneficial effects of this flavonoid. In that sense, many types of nanocarriers have been developed to improve quercetin solubility, as well as to design tissue-specific delivery systems. All these studies manage to improve the bioavailability of quercetin, allowing it to increase its concentration in the desired places. Collectively, quercetin can become a promising compound if nanotechnology is employed as a tool to enhance its therapeutic efficacy.

1. Introduction

1.1. Quercetin as a Promising Compound

Quercetin (3,3′,4′,5,7-pentahydroxyflavone) is a flavonol that was isolated and identified for the first time by Szent-Gyorgyi in 1936 (Figure 1) [1]. In the past, and similarly to what happened with other flavonoids, quercetin was classified as a vitamin shown to be important in maintaining capillary wall integrity and capillary resistance, as well as antihypertensive and antiarrhythmic activity. In subsequent studies, it was proven to have anti-inflammatory and antiallergic properties, hypocholesterolemic activity, platelet and mast cell stabilization, antihepatotoxic activity, and antitumor action [2,3]. Lastly, more in depth analysis demonstrated that the beneficial effects of quercetin were related with its antioxidant (free-radical scavenging) and anti-inflammatory activity [4]. Additionally, some reports also attest its neuroprotective properties. Thus, quercetin emerged as a promising candidate for the treatment of many diseases, such as cancer and neurodegenerative diseases. Nevertheless, in order to explore the potentialities of this flavanol, it is necessary to increase its bioavailability that is very reduced due to its hydrophobicity.
Nanotechnology is an appealing option to overcome this limitation since it can increase quercetin bioavailability at the desired site of action and, consequently, improve the therapeutic effectiveness. Hence, various kinds of NPs, namely liposomes, lipid nanoparticles, polymeric nanoparticles, cyclodextrins, silica nanoparticles, magnetic nanoparticles and gold nanoparticles, have been used to tackle this issue. Despite the fact that some authors have covered specific aspects of quercetin delivery, such as a particular class of nanosystems (polymeric vehicles) or medical applications (tumor therapy) [5,6], a comprehensive review summarizing all the data available regarding nanovehicles used for quercetin delivery is still needed. Intending to fill this gap, we aim to summarize the current literature regarding quercetin (e.g., chemical structure, natural sources, biosynthesis, pharmacokinetics, and health benefits) and compile an extensive analysis of how nanotechnology have been employed to maximize the therapeutic potential of this flavanol in its wide range of medical applications.

1.2. Natural Sources of Quercetin and Biosynthesis

Quercetin is one of the most abundant flavonoids in vegetables and fruits, being found mainly in onions, chilis, berries, and apples (Table 1) [7,8,9]. In leaves, it appears predominantly as aglycones or glycosides (3-position or/and 4′-position), with glucose being the most common sugar group. Nonetheless, lactose and rhamnose can also bind to phenolic groups of quercetin [7]. It is important to mention that, in onion, the best quercetin natural source, quercetin-4′-O-β-glucoside and quercetin-3,4′-O-β-diglucoside represent 80% of the total content of quercetin flavonoid types, being present in the highest quantities in the 28 vegetables and 9 fruits studied [10]. In a different study, quercetin was detected in all 25 eatable berries studied and the highest concentration was found in bog whortleberry (158 mg/kg, fresh weight) [11]. In a different work, quercetin-3-O-β-glucoside was discovered in a considerable amount in apple, pear peels, and Hypericum perforatum leaves or flowers [7,12]. Moreover, other studies have shown that black tea, red wine, and various fruit juices are also good alternatives as natural sources for quercetin [13].
The biosynthesis of quercetin shares almost the same steps in terms of metabolic pathway as other flavonoids. The three-ring structure (A, B, and C) with a diphenyl propane skeleton (C-6-C-3-C-6) is a hallmark of quercetin (Figure 2) [3]. The A and B are benzene rings linked by oxygen containing pyrene ring (C) [14]. The A ring is biosynthesized by the condensation of three moles of malonyl-coenzyme A (CoA) originated from the metabolism of glucose [3]. The B and C rings are also derived from glucose metabolism through the shikimic acid pathway to produce cinnamic acid and its reduced product, coumaric acid [3]. As CoA derivatives, this C-9 p-coumaroyl-CoA condenses with three moles of C-3 malonyl-CoA to form a C-15 chalcone. In the final step of the biosynthesis pathway, the ring closes and the hydration gives rise to quercetin (Figure 2) [15].

1.3. Chemical Structure and Properties of Quercetin

The IUPAC name of quercetin is 2-(3,4-dihydroxyphenyl)-3,5,7-trihydroxy-4H-chromen-4-one. Quercetin is composed of two benzene rings and one oxygen containing pyrene ring (Figure 1) [3]. Quercetin has amphipathic behavior due to the phenyl rings (hydrophobic part) and the hydroxyl groups (hydrophilic part) [16,17]. Nevertheless, quercetin presents low water solubility. There is some controversy in the literature concerning its solubility value; however, most studies indicate approximately 0.01 mg/mL (25 °C) [18,19,20]. Another important parameter while working with this flavonoid is its photodegradation. A photostability study has been conducted with quercetin alcoholic solutions, and the results revealed the appearance of some products of degradation measured by spectrophotometry under the exposure to UVB and UVA radiation, indicating the degradation of this polyphenol compound [21]. Another essential property relates with quercetin stability at different pH values. One study has demonstrated that quercetin is degraded at weak basic pH 8, while, at pH 5, almost 75% of quercetin remains in solution, indicating that quercetin is more stable in a protic medium [22]. Furthermore, a magnetic study of quercetin indicates that this compound displays diamagnetic properties [16]. This feature shows that valence electrons are paired, and, consequently, the compound seems to be stable. Still, the study of LUMO (lowest unoccupied molecular orbital) and HOMO (highest occupied molecular orbital) showed a little energy gap between those orbitals, indicating that an electron can transit from LUMO to HOMO orbitals and, by this way, can easily react [16]. Additionally, the potential ionization study points out to modest requirements in terms of energy to take away an electron from the valence orbital [16]. These two properties may explain the antioxidant capacity of quercetin.

2. Pharmacokinetics of Quercetin

2.1. Absorption, Distribution, Metabolism, and Elimination

Quercetin is a highly hydrophobic compound, so, when it reaches the small intestine, it can be absorbed by the epithelial cells, traversing through cellular membranes (phospholipid bilayers) by a simple diffusion pathway [23]. Chen et al. performed some absorption experiments in Sprague–Dawley rats and found that almost 60% of quercetin orally administered was absorbed [24]. This result was comparable to the work developed by Walle et al., where 53% of quercetin administered was absorbed [25]. Inside the enterocytes, the compound suffers glucuronidation and sulfidation at one of the hydroxyl groups by glucuronosyltransferases and sulfotransferases (phase II enzymes) in order to confer hydrophilicity [26,27,28,29]. Quercetin can also be O-methylated, primarily resulting in the formation of 3′-O-methylquercetin (isorhamnetin) and, to a smaller extent, 4′-O-methylquercetin (tamaraxetin) [28,29]. When quercetin is conjugated with sugars, the first step is to remove this group by β-glucosidase present in enterocytes and intestinal flora, and, after that, the quercetin can be conjugated as previously mentioned [26]. However, some molecules of quercetin can enter the circulation without suffering any conjugation. These molecules will reach the liver through the portal vein, and metabolization by glucuronosyltransferases and sulfotransferases will occur there, which are largely expressed enzymes [30]. Moreover, the catechol-O-methyltransferase (COMT) enzymes present in the liver and kidney can methylate quercetin [29,31,32]. If quercetin conjugates are excreted for bile, they flow through small intestine until they reach the hindgut where they can be hydrolyzed by the β-glucuronidase and sulfatase activities of the microflora, which allows the enterohepatic cycling, increasing the circulation time [30]. Although the liver is the critical organ for quercetin metabolization, a study showed that 90% of quercetin absorbed was metabolized in the gut [24]. All these enzymatic changes transform quercetin in a more soluble compound, allowing its blood circulation freely or bound to blood proteins, such as albumin [33,34]. The entrance of quercetin in blood circulation will result in its tissue distribution. It is also important to refer to the fact that quercetin can be absorbed from the gastrointestinal tract to the lymphatic system [28]. The regular ingestion of the compound will lead to the accumulation in many organs (i.e., lung, kidney, thymus, heart, liver) with the highest concentrations of quercetin and its methylated derivatives, particularly isorhamnetin, found in the pulmonary tissue [35]. Once ingested, quercetin remains in the organism for an extended period (20–72 h), most likely due to its enterohepatic recycling [25]. Nevertheless, quercetin can be degraded by microflora in the colon in phenolic acids and carbon dioxide, which is expelled in breath [36]. Contrarily, the phenolic acids can be excreted in feces. In addition, some quercetin may be also eliminated in urine [37,38,39]. All these routes are valid for quercetin elimination. In accordance to that, a study conducted by Ueno et al. showed that quercetin was excreted as expired CO2 (35%), or via the feces (45%) and urine (10%) as glucuronide or sulfate conjugates following oral administration [40]. However, in a more recent study, only a small amount of absorbed quercetin was eliminated in urine (3.3–5.7%) and feces (0.21–4.6%) [25]. The majority of quercetin was eliminated under carbon dioxide (41.8–63.9%) [25].

2.2. Toxicity of Quercetin

In terms of toxicity, there are still some contradictory results. The in vitro studies revealed that quercetin induces SOS activity and reverse mutations and DNA single strand breaks in bacteria (Salmonella typhimurium strains and Escherichia coli) and in eukaryotic cells, including yeast cells, at relatively high concentrations (up to 10 mg/incubation mixture) in the last case [41,42,43,44]. In hamster and mouse cells and human lymphocytes, harmful effects were reported as chromosomal aberrations, DNA single strand breaks, and micronucleus formation [45,46]. However, all these studies were not conducted in in vivo models. To further explore this issue, mice and rats were orally administered with quercetin and did not exhibit any significant changes in several mutagenicity/genotoxicity endpoints, i.e., micronuclei, chromosomal aberrations, sister chromatid exchange, unscheduled DNA synthesis, and alkali-labile DNA damage [46,47,48]. Based on the toxicity founded by in vitro assays, the carcinogenicity of quercetin was questioned using animal models. F344 rats receiving quercetin as 0.1% of the diet (50 mg/kg body weight/day) for 540 days did not differ from the control rats in terms of tumor incidence, except for lung adenoma and one jejunal adenocarcinoma [49]. In order to test the association between quercetin and lung cancer, quercetin was administered in the diet of A/JJms mice at 5% (7500 mg/kg body weight/day) for 23 weeks, and this compound did not induce a significant difference in the incidence of lung tumors, discarding the possibility of quercetin to induce lung cancer [50]. Therefore, all studies conducted in vivo with animal models indicate that quercetin is a safe compound.

3. Health Benefits of Quercetin

3.1. Cancer Chemoprevention

Quercetin has been reported as a cancer chemopreventive compound. This feature is associated to its ability to inhibit carcinogenesis via antimutagenic and antioxidant activity, anti-inflammatory mechanisms, modulation of signal transduction pathways, and apoptosis-inducing and anti-proliferative activity [51]. The Aryl hydrocarbon receptor (AhR), which binds to polycyclic aromatic hydrocarbons (PAHs), and halogenated aromatic hydrocarbons (HAHs) can activate the expression of CYP1A1, CYP1A2, and CYP1B1; consequently, these enzymes have the capacity to activate procarcinogens resulting in lung cancer [52]. Quercetin seems to naturally bind to AhR, being capable of preventing this signaling cascade and, consequently, cancer [52,53]. Quercetin has also the capacity to induce the expression of death receptor 5 (DR5) in lung cancer cells, which binds to TNF-related apoptosis-inducing ligand (TRAIL) and promotes apoptosis [54]. Moreover, quercetin can interfere with ErbB signaling pathway, reducing the expression of ErbB2 and ErbB3 in HT-29 colon cancer cells resulting in the inhibition of cell growth and the induction of apoptosis [55]. Another critical example relates to prostate cancer prevention. Quercetin demonstrates the capacity to reduce the expression of androgen receptor (AR) in human prostate cancer cell lines, LNCaP, and/or LAPC-4, slowing down cancer progression [56]. Overall, these studies clearly show the capacity of quercetin not only to prevent cancer but also to impair its progression.

3.2. Cardiovascular Protection

Quercetin can protect the cardiovascular system by multiple pathways. Galindo et al. conducted a study with rats and showed that regular intake of quercetin reduces the systolic blood pressure, normalizes the heart rate, reduces heart hypertrophy, and allows aortic relaxation by increasing nitric oxide and reducing some subunits expression of NADPH oxidase [57,58]. Furthermore, quercetin can activate fibrinolytic proteins (t-PA, u-PA, PAI-1, u-PAR, and Annexin-II) in mice, which disrupt fibrin clots in blood vessels, contributing to eliminate the thrombi and, consequently, lowering the risk of coronary heart diseases (CHD) [59]. Despite several studies showing the decrease of blood pressure in rats, there is the need to transpose these studies into humans. With that in mind, Edwards et al. proved that quercetin can lower blood pressure in stage 1 hypertensive patients, suggesting that the same principle also applies to humans [60].

3.3. Anti-Inflammatory Action

Currently, many studies point out to the anti-inflammatory capacity of this polyphenol compound. Mamani-Matsuda and colleagues have worked with rat models of arthritis, which correlates well to what happens in humans in terms of macrophage markers, and demonstrated that quercetin reduced the production of nitric oxide (NO), tumor necrosis factor (TNF-α), monocyte chemoattractant protein 1 (MCP-1), and interleukin 1 beta (IL-1β), which are the primary inflammatory and pro-arthritic mediators of macrophages [61]. Additionally, Rogerio et al. used murine models of asthma to prove the capacity of quercetin to lower the number of white blood cells and eosinophil in the bronchoalveolar lavage fluid, blood, and lung parenchyma [62]. Besides these studies in animal models, the anti-inflammatory ability of quercetin was also tested in human cells. Human mast cell lines were stimulated with phorbol 12-myristate 13-acetate (PMA) and calcium ionophore. Quercetin decreased the gene expression and production of TNF-α, interleukin-1β, IL-6, and IL-8 by reducing the activation of NF-kB and p38 mitogen-activated protein kinase [63]. In another study with human mast cells, Kimata et al. demonstrated the capacity of quercetin to impair the release of histamine, leukotrienes, prostaglandin D2, and granulocyte macrophage-colony stimulating factor [64]. Collectively, these studies gather strong evidences of the anti-inflammatory capacity of quercetin.

3.4. Antioxidant Activity

Compelling evidence shows the antioxidant activity of quercetin. This natural compound participates in many protective mechanisms, such as scavenging reactive oxygen species (ROS) and preventing ROS formation by chelating transition metal ions, such as iron and copper [65,66]. The radical scavenging ability of this flavonol relies on its chemical structure, particularly the hydroxyl (-OH) substitutions and the catechol-type B-ring [67]. Many studies attest this capacity, namely the works developed by Kim and Jang, who showed that quercetin had the ability to protect against the oxidative stress provoked by hydrogen peroxide in HepG2 cell line [68]. Moreover, Sánchez et al. proved that quercetin downregulates NADPH oxidase, increases endothelial nitric oxide synthase (eNOS) activity, and prevents endothelial dysfunction in spontaneously hypertensive rats, also emphasizing its cardiovascular protection through the oxidative stress reduction [58]. Besides that, Chen et al. demonstrated the inhibition of iNOS gene expression by quercetin in mouse BV-2 microglia [69]. Therefore, the literature provides several evidence regarding quercetin antioxidant activity.

3.5. Neuroprotection Effects

Several studies have focused their attention in the neuroprotection activity of quercetin. In fact, it can protect nerve cells from oxidative stress, increasing the survival of neurons [70,71,72]. Simultaneously, it can induce neuron differentiation contributing to maintain the balance of neuron number [73]. Not only to prevent but also to the treatment of neurodegenerative diseases, quercetin can be an interesting natural compound to attenuate the progressive degradation and loss of neuron cells. In this context, quercetin may play an important role in the attenuation of the massive oxidative stress caused by the accumulation of aggregates. For example, in the case of Alzheimer’s disease, Ansari et al. showed that quercetin was capable of reducing protein oxidation, lipid peroxidation, and apoptosis caused by amyloid beta-protein, Aβ(1–42), in primary hippocampal cultures [74]. Moreover, this flavonol has also shown the ability to inhibit the fibril formation of Aβ in a study conducted by Kim et al. [75]. Besides that, quercetin shows a varied spectrum of action regarding brain protection, such as increased mitochondrial biogenesis [76]. Indeed, this is very important because impaired mitochondrial activity seems to be correlated with neurodegenerative diseases [51]. Simultaneously, this natural compound is a potent anti-inflammatory and, therefore, can reduce the expression of proinflammatory molecules, contributing to reduction of the damage associated with this destructive inflammation process [77]. Overall, these studies show that quercetin might contribute to the health of the brain by preventing neurodegenerative diseases, as well as attenuating their deleterious effects.

4. Nanoparticles for Quercetin Delivery

Today, the challenge in drug delivery field consists of transporting drugs to their target places, allowing to increase compounds bioavailability, thereby lowering the amount of administered substances and, consequently, minimizing their side-effects while enhancing their therapeutic efficacy. This question is particularly important while dealing with hydrophobic molecules, as it is critical to design the best vehicles to increase their solubility and bioavailability in order to reach their target areas. Quercetin is one of these examples, and several studies reported in the literature devote significant attention to the development of several nanotechnological approaches which try to establish the best strategy to encapsulate and deliver quercetin for different applications. Table 2 summarizes some parameters that characterize different quercetin delivery systems, namely their composition, particle size, zeta potential, entrapment efficiency, administration route, and in vitro/in vivo results.

4.1. Liposomes

Liposomes can be described as nanocarriers which mimic the cellular phospholipid bilayers. The phospholipids used to produce liposomes have hydrophilic and hydrophobic portions allowing the spontaneous formation of spherical lipid bilayers in water formulations. The type of lipids chosen influences the properties of liposomes, such as charge, size, rigidity, etc. [118]. Liposomes composed of natural phospholipids are biologically inert and weakly immunogenic, which is crucial to biological applications [119]. Taking in consideration these advantages, liposomes have been used to encapsulate quercetin for many purposes [78,79,80,81,82,83,84,120]. Liu et al. have developed liposomes composed of phosphatidylcholine, cholesterol, and tween 80 to encapsulate quercetin in order to protect the hairless skin of Kun Ming mice against photodamage provoked by UVB [82]. Shaji et al. used multi-lamellar vesicles (MLV) with phosphatidylcholine and cholesterol in ratio of 9:1 to encapsulate quercetin and showed hepatoprotective activity in rats [79]. Thinking of tumoral therapy, Yuan et al. worked with a tumor-bearing mice model using lecithin/cholesterol/PEG4000/quercetin in 13:4:1:6 ratio and showed tumors growth inhibition [80]. Long et al. used PEGylated liposomes composed of lecithin and cholesterol to encapsulate quercetin, showing anti-tumor and anti-angiogenesis properties in ovarian cancer mouse model [81]. Interestingly, the delivery of quercetin in liposomes might be used to potentiate the drugs used to treat cancer. With this in mind, Wong et al. used liposomes to encapsulate vincristine and quercetin for treatment in a trastuzumab-insensitive breast tumor xenograft model [83]. This formulation was capable of increasing the circulation time of both drugs in plasma and inhibiting tumor growth [83]. For brain applications, Priprem et al. used quercetin liposomes in rats, with a mixture of egg phosphatidylcholine, cholesterol, and quercetin (2:1:1), dispersed in 50% polyethylene glycol in water. They managed to reduce the anxiety and verified cognitive-enhancing in rats [78]. At the same time, Phachonpai et al. also developed egg phosphatidylcholine/cholesterol liposomes to encapsulate quercetin and showed promising results via intracerebroventricular route administration, reducing degeneration of cholinergic neurons in hippocampus [120]. Finally, in human cell lines, Goniotaki et al. demonstrated that quercetin encapsulated in egg phosphatidylcholine liposomes can inhibit the growth of many human cancer cells [84].

4.2. Lipid Nanoparticles

Lipid nanoparticles can be classified as nanostructured lipid carriers (NLC) and solid lipid nanoparticles (SLN) [121]. The last one is composed of one or more solid lipids, which form a solid matrix, being an excellent vehicle for drug delivery because of their physical stability, protection of the incorporated drug from degradation, controlled release and low cytotoxicity [122]. Despite these advantages, their lipid matrix may suffer recrystallization while stored and form more perfect matrices (β-modifications) that can prematurely release the encapsulated compound [123]. In this context, NLC have been developed to overcome this problem because they blend liquid lipids with solid ones, creating an imperfect structure with more cavities and capacity to encapsulate drugs and avoiding premature release of the encapsulated compounds [123]. Several approaches using lipid nanoparticles have been developed in order to increase quercetin bioavailability and target specific places [86,87,88,89,90]. Li et al. produced SLN with glyceryl monostearate and soya lecithin, registering increased quercetin gastrointestinal absorption in rats [86]. However, it is also feasible the topical administration of quercetin in the skin to protect against oxidative stress caused by multiple factors (radiation, stress, among others). Bose et al. have performed some permeation studies using Franz diffusion cells with human skin and SLN composed of precirol and compritol in 3:2 ratio increased the content of quercetin inside skin, demonstrating that the lipid nanoparticles have great capacities as nanocarriers for topical delivery [88]. This idea was confirmed by Chen-yu et al. using glyceryl monostearate, stearic acid, and media chain triglyceride to prepare NLC for topical administration in ear edema-induced rats. The results showed a suppression of edema in the animals [87]. This capacity to protect against oxidative stress can be used in cancer therapy because cancer formation and progression are frequently associated with multiple mutations that can be caused by ROS. In a study conducted by Sun et al., it was possible to induce apoptosis of MCF-7 and MDA-MB-231 breast cancer cells using NLC composed of 2.7% quercetin, 9.4% soy lecithin, 23.6% glyceryltridecanoate, 6.7% glyceryl tripalmitate, 13.4% vitamin E acetate, and 44.2% Kolliphor HS15 [89]. Besides that, taking advantage of its neuroprotection properties, Dhawan et al. encapsulated quercetin in SLN composed of compritol and tween 80 [90]. They tested this formulation in rats chronically administered with aluminum chloride which causes an oxidative stress responsible for brain damaged [90]. The results showed that quercetin-loaded SLN ameliorated memory retention in rats with aluminum-induced dementia compared to quercetin alone and empty nanoparticles, indicating that this nanosystem may be efficient to target the brain [90]. Besides that, our team has also developed transferrin-functionalized and RVG-29-fuctionalized SLN and NLC made of cetyl palmitate, mygliol-812, and tween 80 to encapsulate quercetin for neuroprotection. The results showed that both types of functionalization enhanced the permeability of quercetin through hCMEC/D3 cells monolayers, and these nanoparticles seemed to be suitable for brain applications due to inhibition of amyloid-beta aggregation [92,93].

4.3. Polymeric Nanoparticles

Polymeric nanoparticles are made of biodegradable polymers which may offer multiple advantages, such as being stable in blood, biodegradable, non-toxic, nonthrombogenic, nonimmunogenic, and noninflammatory, not activating neutrophils, avoiding reticuloendothelial system, and being applicable to various molecules, such as drugs, proteins, peptides, or nucleic acids [124,125]. The versatility of these nanoparticles is based in the capacity to select the most appropriate polymer to the desired application. In this way, these nanomedicines have been used in many approaches [94,95,96,97,98,99,100,101]. Kumari et al. synthesized polylactic acid (PLA) nanoparticles with high encapsulation efficiency and controlled release of quercetin, making them promising for new therapy approaches [94]. Khoee et al. used methacrylated poly(lactic-co-glycolic acid) (mPLGA) as a lipophilic domain, acrylated methoxy poly(ethylene glycol) (aMPEG) as hydrophilic part and N-2-[(tert-butoxycarbonyl)amino] ethyl methacrylamide (Boc-AEMA) as pH-responsive part. They have proven the capacity of these polymeric nanoparticles to release their content in acidic environment, showing that they might be suitable for cancer therapy due to the acidic environment of tumor site [95]. El-Gogary et al. produced PEGylated PLGA nanoparticles conjugated with folic acid and demonstrated the capacity of this quercetin-loaded nanosystem to increase the apoptosis in HeLa cells compared to quercetin alone [96]. At the same time, the tumor uptake was confirmed in injected tumor-bearing mice [96]. Finally, Bishayee et al. used gold-quercetin into PLGA nanoparticles as a special system to escape the immune recognition [97]. The results showed that these nanoparticles had the ability to control proliferation and induce apoptosis in hepatocarcinoma cells [97].

Biopolymeric Nanoparticles

Interestingly, the sub-class of biopolymers are particularly attractive since they are non-toxic, biodegradable and biocompatible. This feature is majorly attributed to its monomer composition consisting essentially of nucleic acids, amino acids, and saccharides from sugars. Indeed, there are reports in the literature describing biopolymeric nanoparticles to deliver quercetin. In one of these studies, Wang et al. developed zein and dextran sulfate sodium binary complex to deliver quercetin. This nanovehicle exhibited almost no toxicity, as well as a sustained released, making it a promising candidate for future applications [104]. In another study, Lozano-Pérez et al. synthesized silk fibroin nanoparticles loaded with quercetin and observed a controlled released under the conditions of pH characteristic of intestinal fluid, which makes them suitable for gastrointestinal delivery [105]. In a follow up study, quercetin-loaded silk fibroin nanoparticles administered in a model mouse of colitis improved the pathological inflammation and preserved the normal crypt architecture, confirming the previously reported capacity for gastrointestinal delivery [106]. Another interesting biopolymer is chitosan, a cationic polysaccharide well known for its biological properties, making it a good candidate when developing drug delivery systems [126]. Pedro et al. developed pH-responsive amphiphilic chitosan nanoparticles to encapsulate quercetin for a sustained release in cancer therapy. The results indicated a good hemocompatibility of the nanoparticles and suggested they could inhibit cancer cell growth in vitro. Therefore, these chitosan nanoparticles may help in reducing the side effects upon systematic administration of quercetin as anti-cancer therapy [107].

4.4. Magnetic Nanoparticles

Magnetic nanoparticles are very promising nanosystems whose magnetic properties allow for guiding them to the target place, applying an external magnetic field [127]. It is also interesting to notice that, below a certain range of size (10–20 nm), magnetic nanoparticles behave as a giant paramagnetic atom with a fast response to applied magnetic fields with negligible remanence (residual magnetism) and coercivity (the field required to bring the magnetization to zero) [128]. The absence of residual magnetism is crucial because agglomeration can be prevented [128]. So far, there are not so many applications using magnetic nanoparticles to encapsulate quercetin; however, their use has now started to take its first steps [108,109,110,111]. For instance, magnetic nanoparticles are very promising for cancer therapy because external magnetic fields can direct them to the tumor site. Taking this in consideration and thinking on the cancer chemotherapeutic properties of quercetin, magnetic nanoparticles must be considered as potential and promising nanosystems for delivering quercetin to the tumor cells. In a preliminary study, Barreto et al. synthesized Fe3O4 nanoparticles and showed that these magnetic nanoparticles had a controlled releasing time, making this vehicle promising for cancer chemotherapy [108]. This study was followed by some studies of concrete applications of magnetic nanoparticles to deliver quercetin. Verma et al. used Fe3O4 magnetic core-shell nanoparticles protected against oxidation by PLGA and tested this formulation in the human lung carcinoma cell line A549 [109]. The results showed that quercetin-loaded PLGA-MNPs had no toxicity after injection in mice, and, at the same time, they were able to reduce the number of A549 viable cells, demonstrating anti-cancer activity [109]. In another study conducted by Kumar et al., quercetin superparamagnetic Fe3O4 nanoparticles were tested in vitro to analyze the effects in breast cancer cell lines [110]. Fluorescent microscopy demonstrated changes in cellular morphology of MCF7-treated cells, indicating cytotoxicity for cancer cells and consequently potential for cancer therapy [110]. Additionally, for targeting brain cancer, Akal et al. designed superparamagnetic iron oxide (SPION), which was functionalized with APTES ((3-aminopropyl) triethoxysilane), polyethylene glycol (PEG), and folic acid [111]. Folic acid was strategically used in order to target brain adenocarcinoma cells (U87), which overexpress folic acid receptors [111]. Furthermore, the MTT analysis after cellular uptake of SPION loaded with quercetin demonstrated decreased cancer cell viability [111]. Together, these results showed clear evidence that magnetic nanoparticles are suitable vehicles to deliver quercetin and to be used for cancer therapy.

4.5. Mesoporous silica Nanoparticles

Mesoporous silica nanoparticles (MSN) are becoming an appealing strategy since they are biocompatible, stable, have a tunable size pore, high drug encapsulation, slow drug release, and are suitable for being functionalized [129,130,131,132,133]. In the literature, there are some works using MSN as vehicles for quercetin delivery [112,113]. For example, Sapino et al. used MSN functionalized with aminopropyl for topical delivery of quercetin [112]. The results have shown that MSN increase the penetration of quercetin in the skin and, at the same time, inhibit the proliferation of R8 human melanoma cells [112]. In another work, MSN functionalized with folate, for targeting breast cancer cells, managed to induce cell cycle arrest and apoptosis in breast cancer cells through the regulation of Akt and Bax signaling pathways [113].

4.6. Cyclodextrins

Cyclodextrins (CDs) resemble a truncated cone with a hydrophobic cavity and a hydrophilic surface. In the cavity, it is possible to accommodate lipophilic drugs and deliver them to their target place [134]. Chemically, CDs are cyclic oligosaccharides with six, seven, or eight glucose residues linked by glycosidic bonds [135]. There are a few applications of CDs as nanodelivey systems to encapsulate quercetin [114,115,116,117]. The inclusion of the quercetin inside β-cyclodextrin (β-CD), hydroxypropyl-β-cyclodextrin (HP-β-CD), and sulfobutyl ether-β-cyclodextrin (SBE-β-CD) has been studied [114,115]. The results demonstrate that SBE-β-CD can encapsulate more quercetin than the other CDs tested and, at the same time, may increase its antioxidant properties [114,115]. Moreover, Aytac et al. used quercetin/β-cyclodextrin inclusion complex, demonstrating the slow release of quercetin [116]. All of these promising features as nanocarriers for quercetin were confirmed in a study conducted by Kale et al. [117]. They used ether-7β-cyclodextrin/quercetin complex in melanoma mouse models and verified the decrease of microvessels and, consequently, the reduction of tumor cell proliferation [117]. Although scarce, the studies here indicated show that CDs holds a great potential for future applications.

5. Clinical Trials and Marketed Formulations

The potentialities of quercetin are well documented in many in vitro and in vivo studies, as cited above. With this in mind, many clinical trials are being made to prove the realistic capacity of quercetin as a tool for new therapies (Table 3) [136]. Ninety-three clinical trials were founded related with quercetin [137]. In a generic view, most of the studies are centered in the United States of America. These clinical trials mainly explore the anticancer, antioxidant, anti-inflammatory, cardiovascular protection, and the anti-infectious properties of quercetin, including against SARS-CoV-2 [137,138]. Although many studies have been completed, only a few have published the results, perhaps due to inconclusive results. One of these studies was focused on the treatment of chronic obstructive pulmonary disease, where quercetin was able to decrease lung inflammation and prevent disease progression in a patient group of 40 to 65 years in age [139]. In the cancer area, quercetin is being studied as a therapeutic agent in colon, pancreas, and prostate cancer [140]. Moreover, some studies are concentrated on the capacity of quercetin to regulate blood pressure and the maintenance of an efficient cardiovascular system [141]. At last, other clinical trials are currently exploring the anti-inflammatory and antioxidant capacity of quercetin, for example, in the treatment of idiopathic pulmonary fibrosis and sarcoidosis [142]. None of the clinical trials mentioned used nanoparticles technology yet. However, compelling evidence demonstrate its benefit in terms of increased drug half-life and solubility, improved drug accumulation on the target site, and reduction of side effects. Due to the aforementioned beneficial properties, quercetin is being used in many market formulations (Table 4). In general, it is possible to find quercetin in many dietary supplements to complement the diet regimen with a potent antioxidant and anti-inflammatory molecule, especially as capsules or tablets [143]. At the same time, this flavonoid can be found in many cosmetics [144]. Brands are creating new formulas to fight the problem of aging [144]. Additionally, some make-up products also contain quercetin in their composition, as indicated in Table 4. Therefore, quercetin is widely used in many commercially available products.

6. Conclusions

The chemical reactivity of quercetin relies on its radical scavenging properties that enable reducing the oxidative stress of cells. At the same time, quercetin presents a strong anti-inflammatory capacity which, together with its antioxidant activity, gives rise to remarkable potentialities, such as cancer chemoprevention, cardiovascular protection, and neurodegeneration attenuation. However, this compound has low water solubility, chemical instability, rapid metabolism, and poor bioavailability, which compromises its therapeutic efficacy. Therefore, new approaches are necessary to take advantage of quercetin beneficial effects, while enhancing its potential application for treating and preventing several disorders. In this context, nanoparticles are particularly interesting when used to accomplish the task of delivering quercetin and enhancing its bioavailability. Furthermore, the capacity of functionalization with specific ligands which target specific organs or cells is also very important because one can increase quercetin concentration on the desired target site, while reducing side effects. Thinking of the beneficial effects of quercetin, a wide range of strategies that use nanoparticles have been developed to deliver quercetin in a specific and controlled way. The revised nanosystems described in the literature revealed promising approaches with a great capacity for quercetin encapsulation and controlled release. Moreover, several strategies of nanoparticle functionalization have allowed the tissue-specific delivery of quercetin to tumor microenvironments, as well as to increase its blood brain barrier permeation or the penetration within skin layers. However, there is a lack of clinical trials employing nanodelivery systems for quercetin administration, considering all the advantages mentioned in this review. Therefore, we identify here a potential area to be explored in future reviews, trying to understand why such promising nanosystems are not yet being widely used in clinical practice and how we can enhance their future use by taking advantage of the therapeutic properties of such a promising compound—quercetin. This strategy has great potential to be explored in a short period of time. Hence, the beneficial effects of quercetin may be further enhanced using nanotechnology, thereby helping to improve the current application of this compound with so great therapeutic potential.

Author Contributions

R.G.R.P.: methodology, writing—original draft preparation; M.P.: writing—review and editing; A.R.N.: conceptualization, methodology, writing—original draft preparation, Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work received financial support from the European Union (FEDER funds) and National Funds (FCT/MEC, Fundação para a Ciência e a Tecnologia and Ministério da Educação e Ciência) under the Partnership Agreement PT2020 UID/MULTI/04378/2013—POCI/01/0145/FEDER/007728. ARN thanks her previous post-Doc grant under the project NORTE-01-0145-FEDER-000011. ARN also acknowledges ARDITI for her current post-Doc grant (ARDITI-CQM_2017_011-PDG) under the project M1420-01-0145-FEDER-000005-CQM+ and the CQM strategic program PEst-OE/QUI/UI0674/2019. MP thanks FCT for funding through program DL 57/2016—Norma transitória.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harborne, J.B. Flavonoids in the environment: Structure-activity relationships. Prog. Clin. Biol. Res. 1988, 280, 17–27. [Google Scholar] [PubMed]
  2. Havsteen, B. Flavonoids, a class of natural products of high pharmacological potency. Biochem. Pharmacol. 1983, 32, 1141–1148. [Google Scholar] [CrossRef]
  3. Formica, J.; Regelson, W. Review of the biology of quercetin and related bioflavonoids. Food Chem. Toxicol. 1995, 33, 1061–1080. [Google Scholar] [CrossRef]
  4. Murota, K.; Terao, J. Antioxidative flavonoid quercetin: Implication of its intestinal absorption and metabolism. Arch. Biochem. Biophys. 2003, 417, 12–17. [Google Scholar] [CrossRef]
  5. Mukhopadhyay, P.; Prajapati, A.K. Quercetin in anti-diabetic research and strategies for improved quercetin bioavailability using polymer-based carriers—A review. RSC Adv. 2015, 5, 97547–97562. [Google Scholar] [CrossRef]
  6. Zang, X.; Cheng, M.; Zhang, X.; Chen, X. Quercetin nanoformulations: A promising strategy for tumor therapy. Food Funct. 2021, 12, 6664–6681. [Google Scholar] [CrossRef]
  7. Wach, A.; Pyrzyńska, K.; Biesaga, M. Quercetin content in some food and herbal samples. Food Chem. 2007, 100, 699–704. [Google Scholar] [CrossRef]
  8. Harnly, J.M.; Doherty, R.F.; Beecher, G.R.; Holden, J.M.; Haytowitz, D.B.; Bhagwat, S.; Gebhardt, S. Flavonoid Content of U.S. Fruits, Vegetables, and Nuts. J. Agric. Food Chem. 2006, 54, 9966–9977. [Google Scholar] [CrossRef]
  9. Day, A.J.; Williamson, G. Human metabolism of dietary quercetin glycosides. Basic Life Sci. 1999, 66, 415–434. [Google Scholar] [CrossRef]
  10. Bonaccorsi, P.; Caristi, C.; Gargiulli, C.; Leuzzi, U. Flavonol Glucoside Profile of Southern Italian Red Onion (Allium cepaL.). J. Agric. Food Chem. 2005, 53, 2733–2740. [Google Scholar] [CrossRef] [PubMed]
  11. Häkkinen, S.H.; Kärenlampi, S.O.; Heinonen, I.M.; Mykkänen, H.M.; Törrönen, A.R. Content of the Flavonols Quercetin, Myricetin, and Kaempferol in 25 Edible Berries. J. Agric. Food Chem. 1999, 47, 2274–2279. [Google Scholar] [CrossRef]
  12. Urbánek, M.; Blechtová, L.; Pospíšilová, M.; Polášek, M. On-line coupling of capillary isotachophoresis and capillary zone electrophoresis for the determination of flavonoids in methanolic extracts of Hypericum perforatum leaves or flowers. J. Chromatogr. A 2002, 958, 261–271. [Google Scholar] [CrossRef]
  13. Sampson, L.; Rimm, E.; Hollman, P.C.; de Vries, J.H.; Katan, M.B. Flavonol and Flavone Intakes in US Health Professionals. J. Am. Diet. Assoc. 2002, 102, 1414–1420. [Google Scholar] [CrossRef]
  14. Erlund, I. Review of the flavonoids quercetin, hesperetin, and naringenin. Dietary sources, bioactivities, bioavailability, and epidemiology. Nutr. Res. 2004, 24, 851–874. [Google Scholar] [CrossRef]
  15. Brown, J. A review of the genetic effects of naturally occurring flavonoids, anthraquinones and related compounds. Mutat. Res. Genet. Toxicol. 1980, 75, 243–277. [Google Scholar] [CrossRef]
  16. Mendoza-Wilson, A.M.; Glossman-Mitnik, D. CHIH-DFT study of the electronic properties and chemical reactivity of quercetin. J. Mol. Struct. THEOCHEM 2005, 716, 67–72. [Google Scholar] [CrossRef]
  17. Codorniu-Hernández, E.; Mesa-Ibirico, A.; Montero-Cabrera, L.A.; Martínez-Luzardo, F.; Borrmann, T.; Stohrer, W.-D. Theoretical study of flavonoids and proline interactions. Aqueous and gas phases. J. Mol. Struct. THEOCHEM 2003, 623, 63–73. [Google Scholar] [CrossRef]
  18. Kim, M.K.; Park, K.-S.; Yeo, W.-S.; Choo, H.; Chong, Y. In vitro solubility, stability and permeability of novel quercetin–amino acid conjugates. Bioorganic Med. Chem. 2009, 17, 1164–1171. [Google Scholar] [CrossRef]
  19. Althans, D.; Schrader, P.; Enders, S. Solubilisation of quercetin: Comparison of hyperbranched polymer and hydrogel. J. Mol. Liq. 2014, 196, 86–93. [Google Scholar] [CrossRef]
  20. Gao, L.; Liu, G.; Wang, X.; Liu, F.; Xu, Y.; Ma, J. Preparation of a chemically stable quercetin formulation using nanosuspension technology. Int. J. Pharm. 2011, 404, 231–237. [Google Scholar] [CrossRef]
  21. Dall’Acqua, S.; Miolo, G.; Innocenti, G.; Caffieri, S. The Photodegradation of Quercetin: Relation to Oxidation. Molecules 2012, 17, 8898–8907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Buchner, N.; Krumbein, A.; Rohn, S.; Kroh, L.W. Effect of thermal processing on the flavonols rutin and quercetin. Rapid Commun. Mass Spectrom. 2006, 20, 3229–3235. [Google Scholar] [CrossRef]
  23. Murota, K.; Nakamura, Y.; Uehara, M. Flavonoid metabolism: The interaction of metabolites and gut microbiota. Biosci Biotechnol Biochem. 2018, 82, 600–610. [Google Scholar] [CrossRef] [Green Version]
  24. Chen, X.; Yin, O.Q.P.; Zuo, Z.; Chow, M.S.S. Pharmacokinetics and Modeling of Quercetin and Metabolites. Pharm. Res. 2005, 22, 892–901. [Google Scholar] [CrossRef] [PubMed]
  25. Walle, T.; Walle, U.K.; Halushka, P.V. Carbon Dioxide Is the Major Metabolite of Quercetin in Humans. J. Nutr. 2001, 131, 2648–2652. [Google Scholar] [CrossRef]
  26. Petri, N.; Tannergren, C.; Holst, B.; Mellon, F.A.; Bao, Y.; Plumb, G.W.; Bacon, J.; O’Leary, K.A.; Kroon, P.; Knutson, L.; et al. Absorption/Metabolism of Sulforaphane and Quercetin, and Regulation of Phase Ii Enzymes, in human jejunum In Vivo. Drug Metab. Dispos. 2003, 31, 805–813. [Google Scholar] [CrossRef] [Green Version]
  27. Rechner, A.R.; Kuhnle, G.; Bremner, P.; Hubbard, G.P.; Moore, K.P.; Rice-Evans, C.A. The metabolic fate of dietary polyphenols in humans. Free Radic. Biol. Med. 2002, 33, 220–235. [Google Scholar] [CrossRef]
  28. Murota, K.; Terao, J. Quercetin appears in the lymph of unanesthetized rats as its phase II metabolites after administered into the stomach. FEBS Lett. 2005, 579, 5343–5346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Graf, B.A.; Ameho, C.; Dolnikowski, G.; Milbury, P.E.; Chen, C.-Y.; Blumberg, J.B. Rat Gastrointestinal Tissues Metabolize Quercetin. J. Nutr. 2006, 136, 39–44. [Google Scholar] [CrossRef] [Green Version]
  30. Crespy, V.; Morand, C.; Manach, C.; Besson, C.; Demigne, C.; Remesy, C. Part of quercetin absorbed in the small intestine is conjugated and further secreted in the intestinal lumen. Am. J. Physiol. 1999, 277, G120–G126. [Google Scholar] [CrossRef]
  31. O’Leary, K.A.; Day, A.J.; Needs, P.W.; Mellon, F.A.; O’Brien, N.M.; Williamson, G. Metabolism of quercetin-7- and quercetin-3-glucuronides by an in vitro hepatic model: The role of human β-glucuronidase, sulfotransferase, catechol-O-methyltransferase and multi-resistant protein 2 (MRP2) in flavonoid metabolism. Biochem. Pharmacol. 2003, 65, 479–491. [Google Scholar] [CrossRef]
  32. De Santi, C.; Pietrabissa, A.; Mosca, F.; Pacifici, G. Methylation of quercetin and fisetin, flavonoids widely distributed in edible vegetables, fruits and wine, by human liver. Int. J. Clin. Pharmacol. Ther. 2002, 40, 207–212. [Google Scholar] [CrossRef]
  33. Boulton, D.W.; Walle, U.K.; Walle, T. Extensive Binding of the Bioflavonoid Quercetin to Human Plasma Proteins. J. Pharm. Pharmacol. 2011, 50, 243–249. [Google Scholar] [CrossRef] [PubMed]
  34. Moon, J.-H.; Nakata, R.; Oshima, S.; Inakuma, T.; Terao, J. Accumulation of quercetin conjugates in blood plasma after the short-term ingestion of onion by women. Am. J. Physiol. Integr. Comp. Physiol. 2000, 279, R461–R467. [Google Scholar] [CrossRef] [PubMed]
  35. De Boer, V.; Dihal, A.A.; Van Der Woude, H.; Arts, I.; Wolffram, S.; Alink, G.M.; Rietjens, I.; Keijer, J.; Hollman, P.C.H. Tissue Distribution of Quercetin in Rats and Pigs. J. Nutr. 2005, 135, 1718–1725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Abrahamse, S.L.; Kloots, W.J.; Van Amelsvoort, J.M. Absorption, distribution, and secretion of epicatechin and quercetin in the rat. Nutr. Res. 2005, 25, 305–317. [Google Scholar] [CrossRef]
  37. Gugler, R.; Leschik, M.; Dengler, H.J. Disposition of quercetin in man after single oral and intravenous doses. Eur. J. Clin. Pharmacol. 1975, 9, 229–234. [Google Scholar] [CrossRef]
  38. De Vries, J.H.; Hollman, P.C.; Meyboom, S.; Buysman, M.N.; Zock, P.; Van Staveren, W.A.; Katan, M.B. Plasma concentrations and urinary excretion of the antioxidant flavonols quercetin and kaempferol as biomarkers for dietary intake. Am. J. Clin. Nutr. 1998, 68, 60–65. [Google Scholar] [CrossRef] [Green Version]
  39. Young, J.F.; Nielsen, S.E.; Haraldsdottir, J.; Daneshvar, B.; Lauridsen, S.T.; Knuthsen, P.; Crozier, A.; Sandstrom, B.; Dragsted, L.O. Effect of fruit juice intake on urinary quercetin excretion and biomarkers of antioxidative status. Am. J. Clin. Nutr. 1999, 69, 87–94. [Google Scholar] [CrossRef] [Green Version]
  40. Ueno, I.; Nakano, N.; Hirono, I. Metabolic fate of [14C] quercetin in the ACI rat. Jpn. J. Exp. Med. 1983, 53, 41–50. [Google Scholar]
  41. Bjeldanes, L.; Chang, G. Mutagenic activity of quercetin and related compounds. Science 1977, 197, 577–578. [Google Scholar] [CrossRef]
  42. Hardigree, A.; Epler, J. Comparative mutagenesis of plant flavonoids in microbial systems. Mutat. Res. Toxicol. 1978, 58, 231–239. [Google Scholar] [CrossRef]
  43. Brown, J.P.; Dietrich, P.S. Mutagenicity of plant flavonols in the Salmonella/mammalian microsome test: Activation of flavonol glycosides by mixed glycosidases from rat cecal bacteria and other sources. Mutat. Res. Toxicol. 1979, 66, 223–240. [Google Scholar] [CrossRef]
  44. Rueff, J.; Laires, A.; Gaspar, J.; Borba, H.; Rodrigues, A.S. Oxygen species and the genotoxicity of quercetin. Mutat. Res. Mol. Mech. Mutagen. 1992, 265, 75–81. [Google Scholar] [CrossRef]
  45. Yoshida, M.A.; Sasaki, M.; Sugimura, K.; Kawachi, T. Cytogenetic Effects of Quercetin on Cultured Mammalian Cells. Proc. Jpn. Acad. Ser. B 1980, 56, 443–447. [Google Scholar] [CrossRef] [Green Version]
  46. Meltz, M.L.; MacGregor, J.T. Activity of the plant flavanol quercetin in the mouse lymphoma L5178Y TK+/− mutation, DNA single-strand break, and Balb/c 3T3 chemical transformation assays. Mutat. Res. Toxicol. 1981, 88, 317–324. [Google Scholar] [CrossRef]
  47. Cierniak, A.; Papiez, M.; Kapiszewska, M. Modulatory effect of quercetin on DNA damage, induced by etoposide in bone marrow cells and on changes in the activity of antioxidant enzymes in rats. Rocz. Akad. Med. Bialymst. 2004, 49 (Suppl. 1), 167–169. [Google Scholar]
  48. Ngomuo, A.J.; Jones, R.S. Genotoxicity studies of quercetin and shikimate in vivo in the bone marrow of mice and gastric mucosal cells of rats. Vet. Hum. Toxicol. 1996, 38, 176–180. [Google Scholar] [PubMed]
  49. Takanashi, H.; Aiso, S.; Hirono, I.; Matsushima, T.; Sugimura, T. Carcinogenicity Test of Quercetin and Kaempferol in Rats by Oral Administration. J. Food Saf. 1983, 5, 55–60. [Google Scholar] [CrossRef]
  50. Hosaka, S.; Hirono, I. Carcinogenicity test of quercetin by pulmonary-adenoma bioassay in strain A mice. Gan 1981, 72, 327–328. [Google Scholar]
  51. Davis, J.M.; Murphy, E.A.; Carmichael, M.D. Effects of the Dietary Flavonoid Quercetin Upon Performance and Health. Curr. Sports Med. Rep. 2009, 8, 206–213. [Google Scholar] [CrossRef] [PubMed]
  52. Murakami, A.; Ashida, H.; Terao, J. Multitargeted cancer prevention by quercetin. Cancer Lett. 2008, 269, 315–325. [Google Scholar] [CrossRef]
  53. Ashida, H.; Fukuda, I.; Yamashita, T.; Kanazawa, K. Flavones and flavonols at dietary levels inhibit a transformation of aryl hydrocarbon receptor induced by dioxin. FEBS Lett. 2000, 476, 213–217. [Google Scholar] [CrossRef]
  54. Chen, W.; Wang, X.; Zhuang, J.; Zhang, L.; Lin, Y. Induction of death receptor 5 and suppression of survivin contribute to sensitization of TRAIL-induced cytotoxicity by quercetin in non-small cell lung cancer cells. Carcinogenesis 2007, 28, 2114–2121. [Google Scholar] [CrossRef] [Green Version]
  55. Kim, W.K.; Bang, M.H.; Kim, E.S.; Kang, N.E.; Jung, K.C.; Cho, H.J.; Park, J.H. Quercetin decreases the expression of ErbB2 and ErbB3 proteins in HT-29 human colon cancer cells. J. Nutr. Biochem. 2005, 16, 155–162. [Google Scholar] [CrossRef]
  56. Xing, N.; Chen, Y.; Mitchell, S.H.; Young, C.Y.F. Quercetin inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Carcinogenesis 2001, 22, 409–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Galindo, P.; Manzano, S.G.; Zarzuelo, M.J.; Gómez-Guzmán, M.; Quintela, A.M.; González-Paramás, A.; Santos-Buelga, C.; Perez-Vizcaino, F.; Duarte, J.; Jimenez, R. Different cardiovascular protective effects of quercetin administered orally or intraperitoneally in spontaneously hypertensive rats. Food Funct. 2012, 3, 643. [Google Scholar] [CrossRef] [PubMed]
  58. Sánchez, M.; Galisteo, M.; Vera, R.; Villar, I.C.; Zarzuelo, A.; Tamargo, J.; Perez-Vizcaino, F.; Duarte, J. Quercetin downregulates NADPH oxidase, increases eNOS activity and prevents endothelial dysfunction in spontaneously hypertensive rats. J. Hypertens. 2006, 24, 75–84. [Google Scholar] [CrossRef] [Green Version]
  59. Booyse, F.M.; Pan, W.; Grenett, H.E.; Parks, D.A.; Darley-Usmar, V.; Bradley, K.M.; Tabengwa, E.M. Mechanism by which Alcohol and Wine Polyphenols Affect Coronary Heart Disease Risk. Ann. Epidemiol. 2007, 17, S24–S31. [Google Scholar] [CrossRef]
  60. Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin Reduces Blood Pressure in Hypertensive Subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef]
  61. Mamani-Matsuda, M.; Kauss, T.; Al-Kharrat, A.; Rambert, J.; Fawaz, F.; Thiolat, D.; Moynet, D.; Coves, S.; Malvy, D.; Mossalayi, M.D. Therapeutic and preventive properties of quercetin in experimental arthritis correlate with decreased macrophage inflammatory mediators. Biochem. Pharmacol. 2006, 72, 1304–1310. [Google Scholar] [CrossRef]
  62. Rogerio, A.D.P.; Kanashiro, A.; Fontanari, C.; Da Silva, E.V.G.; Lucisano-Valim, Y.M.; Soares, E.G.; Faccioli, L.H. Anti-inflammatory activity of quercetin and isoquercitrin in experimental murine allergic asthma. Inflamm. Res. 2007, 56, 402–408. [Google Scholar] [CrossRef]
  63. Min, Y.-D.; Choi, C.-H.; Bark, H.; Son, H.-Y.; Park, H.-H.; Lee, S.; Park, J.-W.; Park, E.-K.; Shin, H.-I.; Kim, S.-H. Quercetin inhibits expression of inflammatory cytokines through attenuation of NF-κB and p38 MAPK in HMC-1 human mast cell line. Inflamm. Res. 2007, 56, 210–215. [Google Scholar] [CrossRef]
  64. Kimata, M.; Shichijo, M.; Miura, T.; Serizawa, I.; Inagaki, N.; Nagai, H. Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin. Exp. Allergy 2000, 30, 501–508. [Google Scholar] [CrossRef] [PubMed]
  65. Afanas’Ev, I.B.; Dcrozhko, A.I.; Brodskii, A.V.; Kostyuk, V.; Potapovitch, A.I. Chelating and free radical scavenging mechanisms of inhibitory action of rutin and quercetin in lipid peroxidation. Biochem. Pharmacol. 1989, 38, 1763–1769. [Google Scholar] [CrossRef]
  66. Van Acker, S.A.; van Balen, G.P.; Berg, D.-J.V.D.; Bast, A.; van der Vijgh, W.J. Influence of iron chelation on the antioxidant activity of flavonoids. Biochem. Pharmacol. 1998, 56, 935–943. [Google Scholar] [CrossRef]
  67. Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.; Flamm, G.; Williams, G.; Lines, T. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem. Toxicol. 2007, 45, 2179–2205. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, G.-N.; Jang, H.-D. Protective Mechanism of Quercetin and Rutin Using Glutathione Metabolism on H2O2-induced Oxidative Stress in HepG2 Cells. Ann. N. Y. Acad. Sci. 2009, 1171, 530–537. [Google Scholar] [CrossRef]
  69. Chen, J.-C.; Ho, F.-M.; Chao, P.-D.L.; Chen, C.-P.; Jeng, K.-C.G.; Hsu, H.-B.; Lee, S.-T.; Wu, W.T.; Lin, W.W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IκB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 2005, 521, 9–20. [Google Scholar] [CrossRef] [PubMed]
  70. Heo, H.J.; Lee, C.Y. Protective Effects of Quercetin and Vitamin C against Oxidative Stress-Induced Neurodegeneration. J. Agric. Food Chem. 2004, 52, 7514–7517. [Google Scholar] [CrossRef]
  71. Zhang, Z.J.; Cheang, L.C.; Wang, M.W.; Lee, S.M. Quercetin exerts a neuroprotective effect through inhibition of the iNOS/NO system and pro-inflammation gene expression in PC12 cells and in zebrafish. Int. J. Mol. Med. 2011, 27, 195–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Dok-Go, H.; Lee, K.H.; Kim, H.J.; Lee, E.H.; Lee, J.; Song, Y.S.; Lee, Y.H.; Jin, C.; Lee, Y.S.; Cho, J. Neuroprotective effects of antioxidative flavonoids, quercetin, (+)-dihydroquercetin and quercetin 3-methyl ether, isolated from Opuntia ficus-indica var. saboten. Brain Res. 2003, 965, 130–136. [Google Scholar] [CrossRef]
  73. Sagara, Y.; Vanhnasy, J.; Maher, P. Induction of PC12 cell differentiation by flavonoids is dependent upon extracellular signal-regulated kinase activation. J. Neurochem. 2004, 90, 1144–1155. [Google Scholar] [CrossRef]
  74. Ansari, M.A.; Abdul, H.M.; Joshi, G.; Opii, W.O.; Butterfield, D.A. Protective effect of quercetin in primary neurons against Aβ(1–42): Relevance to Alzheimer’s disease. J. Nutr. Biochem. 2009, 20, 269–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kim, H.; Park, B.-S.; Lee, K.-G.; Choi, C.Y.; Jang, S.S.; Kim, Y.-H.; Lee, S.-E. Effects of Naturally Occurring Compounds on Fibril Formation and Oxidative Stress of β-Amyloid. J. Agric. Food Chem. 2005, 53, 8537–8541. [Google Scholar] [CrossRef] [PubMed]
  76. Davis, J.M.; Murphy, E.A.; Carmichael, M.D.; Davis, B. Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am. J. Physiol. Integr. Comp. Physiol. 2009, 296, R1071–R1077. [Google Scholar] [CrossRef] [Green Version]
  77. Sharma, V.; Mishra, M.; Ghosh, S.; Tewari, R.; Basu, A.; Seth, P.; Sen, E. Modulation of interleukin-1β mediated inflammatory response in human astrocytes by flavonoids: Implications in neuroprotection. Brain Res. Bull. 2007, 73, 55–63. [Google Scholar] [CrossRef]
  78. Priprem, A.; Watanatorn, J.; Sutthiparinyanont, S.; Phachonpai, W.; Muchimapura, S. Anxiety and cognitive effects of quercetin liposomes in rats. Nanomedicine 2008, 4, 70–78. [Google Scholar] [CrossRef]
  79. Shaji, J.; Iyer, S. Preparation, optimization and in-vivo hepatoprotective evaluation of quercetin liposomes. Int. J. Curr. Pharm. Res. 2012, 4, 24–32. [Google Scholar]
  80. Yuan, Z.-P.; Chen, L.-J.; Fan, L.-Y.; Tang, M.-H.; Yang, G.-L.; Yang, H.-S.; Du, X.-B.; Wang, G.-Q.; Yao, W.-X.; Zhao, Q.-M.; et al. Liposomal Quercetin Efficiently Suppresses Growth of Solid Tumors in Murine Models. Clin. Cancer Res. 2006, 12, 3193–3199. [Google Scholar] [CrossRef] [Green Version]
  81. Long, Q.; Xiel, Y.; Huang, Y.; Wu, Q.; Zhang, H.; Xiong, S.; Liu, Y.; Chen, L.; Wei, Y.; Zhao, X.; et al. Induction of Apoptosis and Inhibition of Angiogenesis by PEGylated Liposomal Quercetin in Both Cisplatin-Sensitive and Cisplatin-Resistant Ovarian Cancers. J. Biomed. Nanotechnol. 2013, 9, 965–975. [Google Scholar] [CrossRef] [PubMed]
  82. Liu, D.; Hu, H.; Lin, Z.; Chen, D.; Zhu, Y.; Hou, S.; Shi, X. Quercetin deformable liposome: Preparation and efficacy against ultraviolet B induced skin damages in vitro and in vivo. J. Photochem. Photobiol. B 2013, 127, 8–17. [Google Scholar] [CrossRef]
  83. Wong, M.-Y.; Chiu, G.N. Liposome formulation of co-encapsulated vincristine and quercetin enhanced antitumor activity in a trastuzumab-insensitive breast tumor xenograft model. Nanomedicine 2011, 7, 834–840. [Google Scholar] [CrossRef]
  84. Goniotaki, M.; Hatziantoniou, S.; Dimas, K.; Wagner, M.; Demetzos, C. Encapsulation of naturally occurring flavonoids into liposomes: Physicochemical properties and biological activity against human cancer cell lines. J. Pharm. Pharmacol. 2010, 56, 1217–1224. [Google Scholar] [CrossRef]
  85. Caddeo, C.; Gabriele, M.; Fernàndez-Busquets, X.; Valenti, D.; Fadda, A.M.; Pucci, L.; Manconi, M. Antioxidant activity of quercetin in Eudragit-coated liposomes for intestinal delivery. Int. J. Pharm. 2019, 565, 64–69. [Google Scholar] [CrossRef]
  86. Li, H.; Zhao, X.; Ma, Y.; Zhai, G.; Li, L.; Lou, H. Enhancement of gastrointestinal absorption of quercetin by solid lipid nanoparticles. J. Control. Release 2009, 133, 238–244. [Google Scholar] [CrossRef] [PubMed]
  87. Chen-Yu, G.; Chun-Fen, Y.; Qi-Lu, L.; Qi, T.; Yan-Wei, X.; Wei-Na, L.; Guang-Xi, Z. Development of a Quercetin-loaded nanostructured lipid carrier formulation for topical delivery. Int. J. Pharm. 2012, 430, 292–298. [Google Scholar] [CrossRef] [PubMed]
  88. Bose, S.; Du, Y.; Takhistov, P.; Michniak-Kohn, B. Formulation optimization and topical delivery of quercetin from solid lipid based nanosystems. Int. J. Pharm. 2013, 441, 56–66. [Google Scholar] [CrossRef]
  89. Sun, M.; Nie, S.; Pan, X.; Zhang, R.; Fan, Z.; Wang, S. Quercetin-nanostructured lipid carriers: Characteristics and anti-breast cancer activities in vitro. Colloids Surf. B Biointerfaces 2014, 113, 15–24. [Google Scholar] [CrossRef]
  90. Dhawan, S.; Kapil, R.; Singh, B. Formulation development and systematic optimization of solid lipid nanoparticles of quercetin for improved brain delivery. J. Pharm. Pharmacol. 2011, 63, 342–351. [Google Scholar] [CrossRef]
  91. Rishitha, N.; Muthuraman, A. Therapeutic evaluation of solid lipid nanoparticle of quercetin in pentylenetetrazole induced cognitive impairment of zebrafish. Life Sci. 2018, 199, 80–87. [Google Scholar] [CrossRef] [PubMed]
  92. Pinheiro, R.; Granja, A.; Loureiro, J.; Pereira, M.; Pinheiro, M.; Neves, A.; Reis, S. Quercetin lipid nanoparticles functionalized with transferrin for Alzheimer’s disease. Eur. J. Pharm. Sci. 2020, 148, 105314. [Google Scholar] [CrossRef]
  93. Pinheiro, R.; Granja, A.; Loureiro, J.; Pereira, M.; Pinheiro, M.; Neves, A.; Reis, S. RVG29-Functionalized Lipid Nanoparticles for Quercetin Brain Delivery and Alzheimer’s Disease. Pharm. Res. 2020, 37, 1–12. [Google Scholar] [CrossRef] [PubMed]
  94. Kumari, A.; Yadav, S.K.; Pakade, Y.B.; Singh, B.; Yadav, S.C. Development of biodegradable nanoparticles for delivery of quercetin. Colloids Surf. B Biointerfaces 2010, 80, 184–192. [Google Scholar] [CrossRef] [PubMed]
  95. Khoee, S.; Rahmatolahzadeh, R. Synthesis and characterization of pH-responsive and folated nanoparticles based on self-assembled brush-like PLGA/PEG/AEMA copolymer with targeted cancer therapy properties: A comprehensive kinetic study. Eur. J. Med. Chem. 2012, 50, 416–427. [Google Scholar] [CrossRef]
  96. Elgogary, R.; Rubio, N.; Wang, T.-W.; Al-Jamal, W.T.; Bourgognon, M.; Kafa, H.; Naeem, M.; Klippstein, R.; Abbate, V.; Leroux, F.; et al. Polyethylene Glycol Conjugated Polymeric Nanocapsules for Targeted Delivery of Quercetin to Folate-Expressing Cancer Cells in Vitro and in Vivo. ACS Nano 2014, 8, 1384–1401. [Google Scholar] [CrossRef]
  97. Bishayee, K.; Khuda-Bukhsh, A.R.; Huh, A.S.-O. PLGA-Loaded Gold-Nanoparticles Precipitated with Quercetin Downregulate HDAC-Akt Activities Controlling Proliferation and Activate p53-ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells. Mol. Cells 2015, 38, 518–527. [Google Scholar] [CrossRef] [Green Version]
  98. Pandey, S.K.; Patel, D.K.; Thakur, R.; Mishra, D.P.; Maiti, P.; Haldar, C. Anti-cancer evaluation of quercetin embedded PLA nanoparticles synthesized by emulsified nanoprecipitation. Int. J. Biol. Macromol. 2015, 75, 521–529. [Google Scholar] [CrossRef] [PubMed]
  99. Sun, D.; Li, N.; Zhang, W.; Zhao, Z.; Mou, Z.; Huang, D.; Liu, J.; Wang, W. Design of PLGA-functionalized quercetin nanoparticles for potential use in Alzheimer’s disease. Colloids Surf. B Biointerfaces 2016, 148, 116–129. [Google Scholar] [CrossRef]
  100. Suksiriworapong, J.; Phoca, K.; Ngamsom, S.; Sripha, K.; Moongkarndi, P.; Junyaprasert, V.B. Comparison of poly(ε-caprolactone) chain lengths of poly(ε-caprolactone)-co-d-α-tocopheryl-poly(ethylene glycol) 1000 succinate nanoparticles for enhancement of quercetin delivery to SKBR3 breast cancer cells. Eur. J. Pharm. Biopharm. 2016, 101, 15–24. [Google Scholar] [CrossRef]
  101. Zhu, X.; Zeng, X.; Zhang, X.; Cao, W.; Wang, Y.; Chen, H.; Wang, T.; Tsai, H.-I.; Zhang, R.; Chang, D.; et al. The effects of quercetin-loaded PLGA-TPGS nanoparticles on ultraviolet B-induced skin damages in vivo. Nanomedicine 2016, 12, 623–632. [Google Scholar] [CrossRef] [PubMed]
  102. Sunoqrot, S.; Abujamous, L. pH-sensitive polymeric nanoparticles of quercetin as a potential colon cancer-targeted nanomedicine. J. Drug Deliv. Sci. Technol. 2019, 52, 670–676. [Google Scholar] [CrossRef]
  103. Pedro, R.D.O.; Goycoolea, F.M.; Pereira, S.; Schmitt, C.C.; Neumann, M.G. Synergistic effect of quercetin and pH-responsive DEAE-chitosan carriers as drug delivery system for breast cancer treatment. Int. J. Biol. Macromol. 2018, 106, 579–586. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, T.-X.; Li, X.-X.; Chen, L.; Li, L.; Janaswamy, S. Carriers Based on Zein-Dextran Sulfate Sodium Binary Complex for the Sustained Delivery of Quercetin. Front. Chem. 2020, 8, 662. [Google Scholar] [CrossRef] [PubMed]
  105. Lozano-Pérez, A.A.; Rivero, H.C.; Hernández, M.D.C.P.; Pagán, A.; Montalbán, M.G.; Víllora, G.; Cénis, J.L. Silk fibroin nanoparticles: Efficient vehicles for the natural antioxidant quercetin. Int. J. Pharm. 2017, 518, 11–19. [Google Scholar] [CrossRef]
  106. Diez-Echave, P.; Ruiz-Malagón, A.J.; Molina-Tijeras, J.A.; Hidalgo-García, L.; Vezza, T.; Cenis-Cifuentes, L.; Rodríguez-Sojo, M.J.; Cenis, J.L.; Rodríguez-Cabezas, M.E.; Rodríguez-Nogales, A.; et al. Silk fibroin nanoparticles enhance quercetin immunomodulatory properties in DSS-induced mouse colitis. Int. J. Pharm. 2021, 606, 120935. [Google Scholar] [CrossRef]
  107. Pedro, R.D.O.; Pereira, S.; Goycoolea, F.M.; Schmitt, C.C.; Neumann, M.G. Self-aggregated nanoparticles of N -dodecyl,N ′-glycidyl(chitosan) as pH-responsive drug delivery systems for quercetin. J. Appl. Polym. Sci. 2018, 135, 45678. [Google Scholar] [CrossRef]
  108. Barreto, A.C.H.; Santiago, V.R.; Mazzetto, S.E.; Denardin, J.C.; Lavín, R.; Mele, G.; Ribeiro, M.E.N.P.; Vieira, I.G.P.; Gonçalves, T.; Ricardo, N.M.P.S.; et al. Magnetic nanoparticles for a new drug delivery system to control quercetin releasing for cancer chemotherapy. J. Nanoparticle Res. 2011, 13, 6545–6553. [Google Scholar] [CrossRef]
  109. Verma, N.K.; Crosbie-Staunton, K.; Satti, A.; Gallagher, S.; Ryan, K.B.; Doody, T.; McAtamney, C.; MacLoughlin, R.; Galvin, P.; Burke, C.S.; et al. Magnetic core-shell nanoparticles for drug delivery by nebulization. J. Nanobiotechnol. 2013, 11, 1. [Google Scholar] [CrossRef] [Green Version]
  110. Kumar, S.R.; Priyatharshni, S.; Babu, V.; Mangalaraj, D.; Viswanathan, C.; Kannan, S.; Ponpandian, N. Quercetin conjugated superparamagnetic magnetite nanoparticles for in-vitro analysis of breast cancer cell lines for chemotherapy applications. J. Colloid Interface Sci. 2014, 436, 234–242. [Google Scholar] [CrossRef]
  111. Akal, Z.; Alpsoy, L.; Baykal, A. Superparamagnetic iron oxide conjugated with folic acid and carboxylated quercetin for chemotherapy applications. Ceram. Int. 2016, 42, 9065–9072. [Google Scholar] [CrossRef]
  112. Sapino, S.; Ugazio, E.; Gastaldi, L.; Miletto, I.; Berlier, G.; Zonari, D.; Oliaro-Bosso, S. Mesoporous silica as topical nanocarriers for quercetin: Characterization and in vitro studies. Eur. J. Pharm. Biopharm. 2015, 89, 116–125. [Google Scholar] [CrossRef]
  113. Sarkar, A.; Ghosh, S.; Chowdhury, S.; Pandey, B.; Sil, P.C. Targeted delivery of quercetin loaded mesoporous silica nanoparticles to the breast cancer cells. Biochim. Biophys. Acta BBA 2016, 1860, 2065–2075. [Google Scholar] [CrossRef] [PubMed]
  114. Jullian, C.; Moyano, L.; Yañez, C.; Olea-Azar, C. Complexation of quercetin with three kinds of cyclodextrins: An antioxidant study. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2007, 67, 230–234. [Google Scholar] [CrossRef]
  115. Liu, M.; Dong, L.; Chen, A.; Zheng, Y.; Sun, D.; Wang, X.; Wang, B. Inclusion complexes of quercetin with three β-cyclodextrins derivatives at physiological pH: Spectroscopic study and antioxidant activity. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2013, 115, 854–860. [Google Scholar] [CrossRef]
  116. Aytac, Z.; Kusku, S.I.; Durgun, E.; Uyar, T. Quercetin/β-cyclodextrin inclusion complex embedded nanofibres: Slow release and high solubility. Food Chem. 2016, 197, 864–871. [Google Scholar] [CrossRef]
  117. Kale, R.; Saraf, M.; Juvekar, A.; Tayade, P. Decreased B16F10 melanoma growth and impaired tumour vascularization in BDF1 mice with quercetin-cyclodextrin binary system. J. Pharm. Pharmacol. 2010, 58, 1351–1358. [Google Scholar] [CrossRef]
  118. Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Immordino, M.L.; Dosio, F.; Cattel, L. Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 2006, 1, 297–315. [Google Scholar]
  120. Phachonpai, W.; Wattanathorn, J.; Muchimapura, S.; Tong-Un, T.; Preechagoon, D. Neuroprotective Effect of Quercetin Encapsulated Liposomes: A Novel Therapeutic Strategy against Alzheimer’s Disease. Am. J. Appl. Sci. 2010, 7, 480–485. [Google Scholar] [CrossRef] [Green Version]
  121. Weber, S.; Zimmer, A.; Pardeike, J. Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) for pulmonary application: A review of the state of the art. Eur. J. Pharm. Biopharm. 2014, 86, 7–22. [Google Scholar] [CrossRef] [PubMed]
  122. Wissing, S.; Kayser, O.; Müller, R. Solid lipid nanoparticles for parenteral drug delivery. Adv. Drug Deliv. Rev. 2004, 56, 1257–1272. [Google Scholar] [CrossRef] [PubMed]
  123. Mehnert, W. Solid lipid nanoparticles Production, characterization and applications. Adv. Drug Deliv. Rev. 2001, 47, 165–196. [Google Scholar] [CrossRef]
  124. Rieux, A.D.; Fievez, V.; Garinot, M.; Schneider, Y.-J.; Préat, V. Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach. J. Control. Release 2006, 116, 1–27. [Google Scholar] [CrossRef] [PubMed]
  125. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [Green Version]
  126. Quiñones, J.P.; Peniche, H.; Peniche, C. Chitosan Based Self-Assembled Nanoparticles in Drug Delivery. Polymers 2018, 10, 235. [Google Scholar] [CrossRef] [Green Version]
  127. Alexiou, C.; Schmid, R.J.; Jurgons, R.; Kremer, M.; Wanner, G.; Bergemann, C.; Huenges, E.; Nawroth, T.; Arnold, W.; Parak, F.G. Targeting cancer cells: Magnetic nanoparticles as drug carriers. Eur. Biophys. J. 2006, 35, 446–450. [Google Scholar] [CrossRef] [PubMed]
  128. Lu, A.-H.; Salabas, E.-L.; Schüth, F. Magnetic Nanoparticles: Synthesis, Protection, Functionalization, and Application. Angew. Chem. Int. Ed. 2007, 46, 1222–1244. [Google Scholar] [CrossRef]
  129. Cauda, V.; Mühlstein, L.; Onida, B.; Bein, T. Tuning drug uptake and release rates through different morphologies and pore diameters of confined mesoporous silica. Microporous Mesoporous Mater. 2009, 118, 435–442. [Google Scholar] [CrossRef]
  130. Vallet-Regí, M.; Balas, F.; Arcos, D. Mesoporous Materials for Drug Delivery. Angew. Chem. Int. Ed. 2007, 46, 7548–7558. [Google Scholar] [CrossRef]
  131. Manzano, M.; Colilla, M.; Vallet-Regí, M. Drug delivery from ordered mesoporous matrices. Expert Opin. Drug Deliv. 2009, 6, 1383–1400. [Google Scholar] [CrossRef]
  132. Di Pasqua, A.J.; Sharma, K.K.; Shi, Y.-L.; Toms, B.B.; Ouellette, W.; Dabrowiak, J.C.; Asefa, T. Cytotoxicity of mesoporous silica nanomaterials. J. Inorg. Biochem. 2008, 102, 1416–1423. [Google Scholar] [CrossRef]
  133. Lu, J.; Liong, M.; Li, Z.; Zink, J.I.; Tamanoi, F. Biocompatibility, Biodistribution, and Drug-Delivery Efficiency of Mesoporous Silica Nanoparticles for Cancer Therapy in Animals. Small 2010, 6, 1794–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wang, W.; Sun, C.; Mao, L.; Ma, P.; Liu, F.; Yang, J.; Gao, Y. The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci. Technol. 2016, 56, 21–38. [Google Scholar] [CrossRef]
  135. Lucas-Abellán, C.; Fortea, I.; López-Nicolás, J.M.; Núñez-Delicado, E. Cyclodextrins as resveratrol carrier system. Food Chem. 2007, 104, 39–44. [Google Scholar] [CrossRef]
  136. Han, M.K.; Barreto, T.A.; Martinez, F.J.; Comstock, A.T.; Sajjan, U.S. Randomised clinical trial to determine the safety of quercetin supplementation in patients with chronic obstructive pulmonary disease. BMJ Open Respir. Res. 2020, 7, e000392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 8 October 2021).
  138. Saeedi-Boroujeni, A.; Mahmoudian-Sani, M.-R. Anti-inflammatory potential of Quercetin in COVID-19 treatment. J. Inflamm. 2021, 18, 1–9. [Google Scholar] [CrossRef]
  139. Beneficial Effects of Quercetin in Chronic Obstructive Pulmonary Disease (COPD) (Quercetin). Available online: https://clinicaltrials.gov/show/NCT01708278 (accessed on 8 October 2021).
  140. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Fallahi, F.; Taghavipour, M.; Ghasemi, Y.; Akbari, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; et al. Quercetin and cancer: New insights into its therapeutic effects on ovarian cancer cells. Cell Biosci. 2020, 10, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Ferenczyova, K.; Kalocayova, B.; Bartekova, M. Potential Implications of Quercetin and its Derivatives in Cardioprotection. Int. J. Mol. Sci. 2020, 21, 1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Boots, A.W.; Veith, C.; Albrecht, C.; Bartholome, R.; Drittij, M.-J.; Claessen, S.M.H.; Bast, A.; Rosenbruch, M.; Jonkers, L.; van Schooten, F.-J.; et al. The dietary antioxidant quercetin reduces hallmarks of bleomycin-induced lung fibrogenesis in mice. BMC Pulm. Med. 2020, 20, 112. [Google Scholar] [CrossRef]
  143. Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, Inflammation and Immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef] [PubMed]
  144. Chondrogianni, N.; Kapeta, S.; Chinou, I.; Vassilatou, K.; Papassideri, I.; Gonos, E.S. Anti-ageing and rejuvenating effects of quercetin. Exp. Gerontol. 2010, 45, 763–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Chemical structure of quercetin.
Figure 1. Chemical structure of quercetin.
Nanomaterials 11 02658 g001
Figure 2. Schematic representation of quercetin biosynthesis.
Figure 2. Schematic representation of quercetin biosynthesis.
Nanomaterials 11 02658 g002
Table 1. Quercetin natural sources [1,2,3].
Table 1. Quercetin natural sources [1,2,3].
Sources Total Quercetin (mg/Kg)
White onion bulbs2604
Onion dry outer skin960
Spring onion leaves450
Chili powder400
Bog whortleberry158
Lingonberry146
Cranberry121
Kale110
Chokeberry89
Sweet rowan85
Rowanberry63
Sea buckthorn berry62
Apples red delicious58
Crowberry56
Broccoli30
Green beans25
Apple peel21
Tomato13
Table 2. Properties of different quercetin-loaded carriers.
Table 2. Properties of different quercetin-loaded carriers.
NPs TypeCompositionMorphologyParticle Size (nm)Zeta Potential (mV)Encapsulation Efficiency (EE) (%)Administration RouteIn Vitro/In Vivo ResultsReference
LiposomesPC/Chol; EPC/Chol/PEG; lecithin/Chol/PEG; ESM/Chol/PEG
P90G/STA/Eudragit S100
spherical90 to 450+7 to −3065 to 95oral, intranasal, intravenous, topical- anxiolytic and cognitive-enhancing effects in rats (in vivo);
- hepatoprotective effects in rodents (in vivo);
- attenuation of edema and inflammation in irradiated mice (in vivo);
- inhibition of tumor angiogenesis, inhibition of tumor cell growth and induction of tumor cell apoptosis (cell lines and mice) (in vitro and in vivo);
- protection against intestinal oxidative stress in intestinal cell lines (in vitro);
[78,79,80,81,82,83,84,85]
Lipid nanoparticlesGMS/soya lecithin/PEG;
GMS/SA/MCT/soya lecithin;
Compritol;
GT/TG/soya lecithin
SA/P-188;
cetyl palmitate, mygliol-812 and tween 80
spherical35 to 550−10 to −3575 to 95oral, intravenous, topical, intraperitoneal- enhancement of the oral absorption in rats (in vivo);
- increase of drug penetration in skin and anti-oxidation and anti-inflammation effect (in vivo);
- improvement of memory retention in rats with aluminum-induced dementia (in vivo);
- induction of tumor cell apoptosis in breast cancer cell lines (in vitro).
- improvements of cognition and memory impairments in zebrafish model of neurodegenerative disorders (in vivo).
- inhibition of amyloid-beta aggregation and fibril formation in vitro, being suitable for brain applications (in vitro).
[86,87,88,89,90,91,92,93]
Polymeric nanoparticlesPLA; PLGA; PLGA/PEG/AEMA;
PLGA/PEG/FA;
PCL/TPGS; PLGA/TPGS
Eudragit S100
L-CS/DEAE
spherical, bean-like shape30 to 415+42 to −4040 to 98intravenous, topical- enhancement of cancer cell uptake in tumor-bearing mice (in vivo);
- increase of cytotoxicity and induction of apoptosis in breast, colon and liver hepatocellular carcinoma cell lines (in vitro);
- block/reduce of histological alterations in irradiated mice (in vivo);
- enhancement of neuron cells viability due to inhibition of aβ-42 peptide neurotoxicity in cell lines (in vitro);
- improvements of cognition and memory impairments in APP/PS1 mice model of Alzheimer’s disease (in vivo).
[94,95,96,97,98,99,100,101,102,103]
Biopolymeric nanoparticlesZein-Dextran sulfate sodium; Skin Fibroin; Chitosanspherical140 to 300−16 to −39;
+14 to +30
20 to 78oral, intravenous- promising favorable and sustained delivery of quercetin (in vitro);
- controlled release in the pH of intestinal fluid; suitable for gastrointestinal delivery (in vitro);
- improving pathological inflammation and preserving normal crypt architecture in model mouse of colitis (in vivo);
- pH-responsive sustained release for cancer therapy; reducing side effects upon systematic administration (in vitro).
[104,105,106,107]
Magnetic nanoparticlesFe3O4;
Fe3O4/E137S18E137;
Fe3O4/APTS/PEG/FA; Fe3O4/PLGA
spherical10 to 300around +6around 80intranasal- induction of cytotoxicity in human lung carcinoma, breast cancer and glioblastoma cell lines (in vitro).[108,109,110,111]
Mesoporous silica nanoparticlesTEOS/APTS;
TEOS/FA
spherical200 to 250−25 to +13around 99n.i.- inhibition of tumor cell proliferation and induction of tumor cell apoptosis (cell lines) (in vitro).[112,113]
CyclodextrinsβCD; HP-βCD;
SBE-βCD; SBE-7βCD
truncated cone; fibersaround 270n.i.n.i.oral- increase of quercetin solubility and photostability;
- enhancement of quercetin antioxidant capacity (in vivo);
- inhibition of tumor cell proliferation in cell lines (in vitro);
- impairment of tumor growth in B16F10 mouse melanoma model (in vivo).
[114,115,116,117]
Table 3. Quercetin clinical trials conducted around the world (reported at http://clinicaltrials.gov/, last access on 8 October 2021).
Table 3. Quercetin clinical trials conducted around the world (reported at http://clinicaltrials.gov/, last access on 8 October 2021).
CountryStudyConditionInterventionPhaseStatusResults
U.S.—MichiganPhase I Study to Determine the Safety of Quercetin in COPD Patients Chronic Obstructive Pulmonary Disease Quercetin1Completed with resultsDecreased lung inflammation and prevented progression
U.S.—Massachusetts Hypoxia-inducible Transcription Factor 1 (HIF-1) in Vascular Aging Stroke;
Problem of Aging
QuercetinEarly phase 1completedNot Indicated
U.S.—Ohio Quercetin in Children with Fanconi Anemia; a Pilot Study Fanconi Anemia Quercetin1RecruitingThere are no results
U.S.—North Carolina Can Quercetin Increase Claudin-4 and Improve Esophageal Barrier Function in GERD? Gastroesophageal Reflux Disease
(GERD);
Acid Reflux;
Reflux
quercetin1completedNot indicated
U.S.—California A Phase 1 Study of Quercetin in Patients with Hepatitis C Chronic Hepatitis C quercetin1completedNot indicated
The Netherlands The Effect of Quercetin on the Increased Inflammatory and Decreased Antioxidant Status in Sarcoidosis Sarcoidosis quercetin-completedNot indicated
The Netherlands Study on the Effects of Epicatechin and Quercetin Supplementation on Vascular Function and Blood Pressure in Untreated (Pre)Hypertensive Subjects Hypertension
Endothelial Dysfunction
Epicatechin; quercetin -completedNot indicated
Islamic Republic of IranEffect of Quercetin in Prevention and Treatment of Chemotherapy Induced Oral Mucositis in Blood Dyscrasias Chemotherapy Induced Oral Mucositis oral quercetin capsules1,2completedNot indicated
U.S.—New Mexico Effect of Combined Exercise, Heat, and Quercetin Supplementation on Whole Body Stress Response Heat Acclimation and Thermotolerance Quercetin-completedNot indicated
U.S.—California Phase I Randomized, Double-Blind, Placebo-Controlled Two-Arm Study of Quercetin and Green Tea to Enhance the Bioavailability of Green Tea Polyphenols in Men Scheduled for Prostatectomy Adenocarcinoma of the Prostate;
Recurrent Prostate Cancer;
Stage I Prostate Cancer;
Stage IIA Prostate Cancer;
Stage IIB Prostate Cancer;
Stage III Prostate Cancer;
Stage IV Prostate Cancer;
Dietary Supplement: green tea extract
Drug: quercetin
Other: placebo
Procedure: therapeutic conventional surgery
Other: laboratory biomarker analysis
1 Active, not recruiting There are no results
U.S.—Massachusetts Pharmacokinetic and Pharmacodynamic Study of Oral Quercetin and Isoquercetin in Healthy Adults and Patients with Elevated Anti-phospholipid Antibodies Healthy Drug: isoquercetin or quercetin- Active, not recruiting There are no results
U.S.—Washington Evaluation of Quercetin in Type 2 Diabetes: Impact on Glucose Tolerance and Postprandial Endothelial Function. Diabetes Mellitus, Type 2 Dietary Supplement: Quercetin
Drug: Acarbose
Drug: placebo
2completedNot indicated
Islamic Republic of IranTherapeutic Effect of Quercetin and the Current Treatment of Erosive and Atrophic Oral Lichen Planus Atrophic Oral Lichen Planus;
Erosive Oral Lichen Planus
Drug: placebo
Drug: Quercetin
1unknowThere is no information
Germany Clinical Trial on the Effectiveness of the Flavonoids Genistein and Quercetin in Men with Rising Prostate-specific Antigen Primary Prevention of Prostate Cancer Dietary Supplement: Quercetin supplement
Dietary Supplement: Genistein supplement
Dietary Supplement: Placebo
-unknownThere is no information
U.S.—Texas Prospective Open Labeled Pilot Trial of Quercetin in the Treatment and Prevention of Chemotherapy-induced Neuropathic Pain in Cancer Patients Polyneuropathies and Other Disorders of the Peripheral Nervous System Chemotherapy Induced Neuropathic Pain Behavioral: Questionnaires
Drug: Quercetin
Early Phase 1Not yet recruitingThere are no results
U.S.—Maryland Inhibition of Intestinal Glucose Absorption by the Bioflavonoid Quercetin in the Obese and in Obese Type 2 Diabetics Diabetes Mellitus;
Obesity
Procedure: Oral glucose tolerance test;
coadministration of 1 or 2 g of quercetin with glucose
2recruitingThere are no results
United Kingdom A Double-Blind, Cross-Over, Placebo-Controlled Study Evaluating the Effect of Quercetin 500 mg Tablets on Blood Uric Acid in Healthy Males Hyperuricemia;
Gout;
Kidney Calculi;
Diabetes;
Cardiovascular Disease
Dietary Supplement: Treatment
Dietary Supplement: Control
Early Phase 1 completedThere are no results
U.S.—Pennsylvania Advancing Niacin by Inhibiting FLUSHing: (ANTI-FLUSH) Flushing Dietary Supplement: Quercetin
Dietary Supplement: Placebo
4Completed, has resultsNot indicated
Greece Effects of the Anti-inflammatory Flavonoid Luteolin on Behavior in Children with Autism Spectrum Disorders Autism Spectrum Disorders Dietary Supplement: Luteolin, Quercetin and Rutin combined in a capsule2completedThere are no results
U.S.—Alabama Nasal Potential Studies Utilizing CFTR Modulators (UAB Center for Clinical and Translational Science) Cystic Fibrosis Other: quercetin2completedThere are no results
U.S.—North Carolina Targeted Removal of Pro-Inflammatory Cells: An Open Label Human Pilot Study in Idiopathic Pulmonary Fibrosis Idiopathic Pulmonary Fibrosis (IPF) Drug: Dasatinib + Quercetin1recruitingThere are no results
U.S.—Minnesota Senescence, Frailty, and Mesenchymal Stem Cell Functionality in Chronic Kidney Disease: Effect of Senolytic Agents Chronic Kidney Disease Drug: Group 2: Dasatinib
Drug: Group 2: Quercetin
2recruitingThere are no results
U.S.—Minnesota Hematopoietic Stem Cell Transplant Survivors Study (HTSS Study) Stem Cell Transplant Drug: Quercetin
Other: Standard of Care—Observation Only Drug: Dasatinib
-recruitingThere are no results
U.S.—Utah A Double-Blind, Placebo-Controlled, Crossover Evaluation of a Grape Seed Extract and Quercetin Supplement (Provex CV) to Reduce Markers of Inflammatory Cytokines and Blood Pressure in Subjects with Metabolic Syndrome Blood Pressure Drug: Provex CV
Other: placebo
1CompletedThere are no results
U.S.—New York The Effect of Plant Phenolic Compounds on Human Colon Epithelial Cells Colorectal Cancer Dietary Supplement: curcumin
Dietary Supplement: rutin Drug: quercetin
Drug: sulindac
-terminatedThere are no results
U.S.—Massachusetts Absorption Kinetics of Polyphenols in Angel’s Plant (Angelica Keiskei) Oxidative Stress Dietary Supplement: angel’s plant (Angelica keiskei) Dietary Supplement: Angel’s plant (Angelica keiskei) Early Phase 1 unknownThere is no information
U.S.—Michigan Three New Ideas to Protect Special Forces from the Stress of High Altitude Mountain Sickness Dietary Supplement: Quercetin
Drug: Nifedipine extended release Drug: Methazolamide
Drug: Metformin
Drug: Placebo
Drug: Nitrite
4recruitingThere are no results
The NetherlandsThe Inflammatory and Antioxidant Status in Pulmonary Sarcoidosis, Idiopathic Pulmonary Fibrosis and COPD: a Potential Role for Antioxidants Interstitial Lung Diseases;
Sarcoidosis;
Idiopathic Pulmonary Fibrosis;
COPD;
quercetin-completedThere are no results
Republic of KoreaEffect of Onion Peel Extract on Endothelial Function and Endothelial Progenitor Cells in Overweight and Obese SubjectsHealthy
Overweight
Obese
Drug: Placebo
Drug: Onion peel extract
4completedThere are no results
United Kingdom The Impact of Isoquercetin and Aspirin on Platelet Function Cardiovascular Disease Drug: Vehicle control
Drug: Isoquercetin
Drug: Aspirin
Drug: Isoquercetin plus Aspirin
- withdrawn There are no results
U.S.—California Randomized, Placebo-controlled, Double-blind Phase II/III Trial of Oral Isoquercetin to Prevent Venous Thromboembolic Events in Cancer Patients. Thromboembolism of Vein VTE in Colorectal Cancer;
Thromboembolism of Vein in Pancreatic Cancer;
Thromboembolism of Vein in Non-small Cell Lung Cancer
Drug: Isoquercetin 2,3recruitingThere are no results
Germany Pilot Study Evaluating Broccoli Sprouts in Advanced Pancreatic Cancer [POUDER Trial]
Pancreatic Ductal Adenocarcinoma Dietary Supplement: Verum, broccoli sprout grain
Dietary Supplement: placebo
-recruitingThere are no results
- An Open, Uncontrolled Trial in Healthy Volunteers to Explore the Plasma and Urinary Pharmacokinetics of a Single Oral Dose of 1800 mg Red Vine Leaf Extract (Antistax®) Healthy Drug: Red Vine Leaf Extract 1completedThere are no results
U.S.—North Carolina Influence of Ingesting a Flavonoid-rich Supplement on the Human Metabolome and Concentration of Urine Phenolics Inflammation Dietary Supplement: Flavonoid
Dietary Supplement: Placebo
-Active, not recruitingThere are no results
Italy Isoquercetin as an Adjunct Therapy in Patients with Kidney Cancer Receiving First-line Sunitinib: a Phase I/II TrialRenal Cell Carcinoma;
Kidney Cancer
Drug: Sunitinib
Drug: Isoquercetin
Drug: Placebo
1,2RecruitingThere are no results
U.S.—Illinois, New Jersey and OhioOpen-Label Study to Evaluate the Effect of Elimune Capsules on Biomarkers in Patients with Plaque Psoriasis Plaque Psoriasis Dietary Supplement: Elimune Capsules 1completedThere are no results
FranceRandomized Controlled Double-blind Clinical Trial for the Effect of Yoghurts Enriched in XXS vs without XXS on the Evolution of Weight in Overweight Subjects Aged 50 to 65 Years.Obesity
Overweight
Dietary Supplement: yoghurts enriched with XXS
Dietary Supplement: yoghurts non enriched with XXS
-completedThere are no results
Brazil Evaluation of Clinical Efficacy Capsules Containing Standardized Extract of Bauhinia Forficata (Pata- De-vaca) in Diabetic Patients Diabetes Drug: B. forficata
Drug: Placebo
-recruitingThere are no results
Egypt??Effects of Oral Ginkgo Biloba Extract on Pregnancy Complicated by Asymmetrically Intrauterine Growth Restriction: a Double-blinded Randomized Placebo-controlled Trial Intrauterine Growth Restriction (IUGR) Drug: Ginkgo Biloba Extract
Other: Placebo
2completedThere are no results
ItalyPhysiological Effects of New Polyphenol-enriched Foods in Healthy Humans Healthy Other: free curcumin
Other: encapsulated curcumin
Other: encapsulated curcumin + PQG
Other: free cocoa polyphenol
Other: encapsulated cocoa polyphenols
Other: control nut cream
Encapsulada a quercetina
-Completed
Has Results
There are no results
U.S.—Missouri Can Fish Oil and Phytochemical Supplements Mimic Anti-Aging Effects of Calorie Restriction? Healthy Volunteers Dietary Supplement: Supplement -completedThere are no results
U.S.—MichiganGRAPe Seed Extract and Ventriculovascular Investigation in Normal Ejection-Fraction Heart FailureDiastolic Heart Failure;
Hypertensive;
Heart Disease;
;Heart Failure with Preserved Ejection Fraction;
Hypertension;
Oxidative Stress
Drug: grape seed extract (MegaNatural BP, Polyphenolics, Inc.) 1 Active, not recruiting There are no results
U.S.—South Carolina Effects of Short-term Curcumin and Multi-polyphenol Supplementation on the Anti-inflammatory Properties of HDL (PSI)Inflammation;
Atherosclerosis;
Cardiovascular Disease
Dietary Supplement: PolyResveratrol Supplementation
Dietary Supplement: Curcumin Supplementation
2RecruitingThere are no results
Canada, Alberta Orthomolecular Treatment as add-on Therapy for Childhood Asthma Asthma Dietary Supplement: Orthomolecular Therapy or Placebo Comparator 2unknownThere are no results
U.S.—CaliforniaEffects of Plant Concentrate Blend on Oxidative Stress in Healthy Humans Healthy Dietary Supplement: AOX blend
Dietary Supplement: Placebo
-completedThere are no results
U.S.—Ohio Phytochemical Release Rate from Black Raspberry Confections Alters Gene Expression and Chemical Profiles Relevant to Inhibition of Oral Carcinogenesis Healthy Volunteers Other: Fast release BRB confection
Other: Intermed release BRBconfect
Other: Prolong release BRB confect
1 Active, not recruiting There are no results
Norway Dietary Intervention in Stage III/IV Follicular Lymphoma. Impact on Markers of Cell Proliferation, Apoptosis, Host Immune Cell Infiltrate and Oxidative Stress Follicular Lymphoma Drug: Omega 3 fatty acids (EPA (eicosapentaenoic acid) and DHA (docosahexaenoic acid))
Drug: Selenium (L-Selenomethionine),
Drug: Garlic extract (Allicin)
Drug: Pomegranate juice (ellagic acid)
Drug: Grape juice (resveratrol, quercetin)
Drug: Green Tea (Epigallocathechin gallate
2unknownThere are no results
U.S.—Florida Open Label, Crossover, Pilot Study to Assess the Efficacy & Safety of Perispinal Admin. of Etanercept(Enbrel®) in Comb. w/Nutritional Supplements vs. Nutritional Supplements Alone in Subj. w/Mild to Mod. Alzheimer’s Disease Receiving Std. Care Alzheimer’s Disease Biological: Etanercept
Dietary Supplement: Curcum. Luteol. Theaflav. Lip. Acid, FishOil, Quercet, Resveratr.
1Completed There are no results
U.S.—California Proof of Concept Study to Evaluate the Effect of Oxidative Stress Response of Plant Concentrate Blends in Healthy Men Healthy Dietary Supplement: Plant concentrate A
Dietary Supplement: Plant concentrate B
Dietary Supplement: Plant concentrate C
Dietary Supplement: Plant concentrate D
-completedThere are no results
U.S.—OhioQuercetin chemoprevention for squamous cell carcinoma in patients with Fanconi anemiaFanconi anemia
Squamous cell carcinoma
Dietary supplementPhase 2RecruitingThere are no results
U.S.—PennsylvaniaImpact of quercetin on inflammatory and oxidative stress markers in COPDChronic obstructive pulmonary diseaseQuercetinPhase 1/Phase 2Not yet recruiting There are no results
Not indicated Serum concentration and gene expression of sirtuin-1 and advanced glycation End-products in postmenopausal women with atherosclerotic coronary disease after administration of atorvastatin and supplementation with quercetin: Randomized trial sirtuin-1 and advanced glycation End-products in postmenopausal women with coronary diseaseCoronary artery disease progressionQuercetinPhase 3Not yet recruiting There are no results
Table 4. Quercetin marketed formulations.
Table 4. Quercetin marketed formulations.
BrandDosagePharmaceutical FormApplication
Quercetin 500 mg (Nature’s Best)500 mgTabletsSupplement
Rutin and vitamin c 500 mg (Nature’s Best)50 mgTabletssupplement
Glucosamine Plus (Nature’s Best)25 mgTabletssupplement
Glucosamine Gold (Nature’s Best)20 mgTabletssupplement
Quercetin with bromelain (Biovea)500 mgCapsules Vegan supplement
Neuroprotek Low Phenol (algonot)70 mgcapsulesDietary supplement
Neuroprotek (algonot)70 mgcapsulesDietary supplement
Algonot plus (algonot)150 mgcapsulesDietary supplement
ArthroSoft (algonot)150 mgcapsulesDietary supplement
CystoProtek (algonot)150 mgcapsulesDietary supplement
FibroProtek (algonot)85 mgcapsulesDietary supplement
ProstaProtek (algonot)-capsulesDietary supplement
Quercetin (Natrol)250 mgcapsulesDietary supplement
Quercetin (Desert Harvest)250 mgcapsulesDietary supplements
Quercetin and vitamin (Vitacost)250 mgcapsulesDietary supplements
Aller-Aid with quercetin (Wild Harvest)137,5 mgcapsulesHerbal supplement
Advan-c (Vitacost)100 mgcapsulesVitamin supplement
Quercetin and Bromelain (Vitacost)250 mgcapsulesDietary supplement
Quercetin (Jarrow formulas)500 mgcapsulesDietary supplement
Quercetin (solaray)500 mgcapsulesDietary supplement
Quercetin and Bromelain (Now)400 mgcapsulesVegan supplement
Activated quercetin complex (Vitacost)333 mgcapsulesDietary supplement
Quercetin complex (solgar)250 mgcapsulesDietary supplement
Mega quercetin (solaray)600 mgcapsulesDietary supplement
Quercetin strength (megaFood)500 mgcapsulesDietary supplement
Activated quercetin (source naturals333 mgcapsulesDietary supplement
Optimized quercetin LifeExtension250 mgcapsulesDietary supplement
Nettle quercetin (eclectic institute)175 mgcapsulesDietary supplement
Quercetin (natural factors)235 mgcapsulesDietary supplement
Quercetin (nature’s life)400 mgcapsulesAntioxidant supplement
Quercetin (Pure encapsulations)250 mgcapsulesDietary supplement
Quercetin bioflavonoids (NutriCology)50 mgcapsulesDietary supplement
Quercetin (MRM)500 mgcapsulesDietary supplement
Quercetin (Kal)1000 mgtabletsDietary supplement
Quercetin plus (Olympian labs)500 mgcapsulesDietary supplements
ALLER-C (Vital Nutrients)250 mgcapsulesDietary supplements
Quercetin Bromelain (Doctor’s BEST)250 mgcapsulesDietary supplement
Rose Plus Anti-Ageing Face (The organic Pharmacy)n.iCream anti-ageing cream
Flash Defense Anti-Pollution Mist (REN)n.icreamskincare
Antiwrinkle Firming and Lifting serum (Korres)n.iserumskincare
Tightening eye contour gel (Alchimie forever)n.igelMake up
Firming gel for neck and bust (Alchimie forever)n.igelskincare
Kantic calming cream (Alchimie forever)n.icreamskincare
Rejuvenating eye balm (Alchimie forever)n.icreamskincare
Antioxidant skin repair gel (Alchimie forever)n.igelskincare
Gentle cream cleanser (Alchimie forever)n.icreamskincare
Resist advanced replenishing toner (Paula’s choice)n.icreamskincare
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pinheiro, R.G.R.; Pinheiro, M.; Neves, A.R. Nanotechnology Innovations to Enhance the Therapeutic Efficacy of Quercetin. Nanomaterials 2021, 11, 2658. https://doi.org/10.3390/nano11102658

AMA Style

Pinheiro RGR, Pinheiro M, Neves AR. Nanotechnology Innovations to Enhance the Therapeutic Efficacy of Quercetin. Nanomaterials. 2021; 11(10):2658. https://doi.org/10.3390/nano11102658

Chicago/Turabian Style

Pinheiro, Rúben G. R., Marina Pinheiro, and Ana Rute Neves. 2021. "Nanotechnology Innovations to Enhance the Therapeutic Efficacy of Quercetin" Nanomaterials 11, no. 10: 2658. https://doi.org/10.3390/nano11102658

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