Back to Journals » International Journal of Nanomedicine » Volume 20
Quercetin and Nano-Derivatives: Potential and Challenges in Cancer Therapy
Authors Li XR, Qi L, Zhang XW, Wei C, Yu B, Pei TL
Received 2 December 2024
Accepted for publication 28 April 2025
Published 24 May 2025 Volume 2025:20 Pages 6701—6720
DOI https://doi.org/10.2147/IJN.S509877
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 5
Editor who approved publication: Dr Krishna Nune
Xin-Ru Li,1 Lin Qi,2 Xi-Wen Zhang,3 Chao Wei,1 Bin Yu,1 Tian-Li Pei4
1College of Integrated Chinese and Western Medicine, Jining Medical University, Jining, Shandong Province, 272000, People’s Republic of China; 2Affiliated Hospital of Jining Medical University, Jining, 272000, People’s Republic of China; 3College of The Second Clinical Medical, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan Province, 450003, People’s Republic of China; 4Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, 550000, People’s Republic of China
Correspondence: Tian-Li Pei, Email [email protected]
Abstract: Quercetin, a prevalent flavonol compound, has gained attention for its multifaceted mechanisms of action against various cancers, highlighting its potential as an adjunctive therapy in cancer treatments. This review aims to systematically evaluate the structural optimization, mechanisms of action, and clinical applications of quercetin and its nano-derivatives in cancer treatment. Employing a bibliometric analysis of 6231 articles from the Web of Science Core Collection, we observed a notable increase in annual publications, particularly from the USA and China, indicating a growing interest in quercetin’s therapeutic potential. Our findings reveal that quercetin enhances the efficacy of conventional therapies by modulating critical signaling pathways, thereby increasing cancer cell sensitivity while simultaneously protecting normal tissues from therapy-induced damage. Structural modifications, including glycosylation, methylation, sulfation, and glucuronidation, alongside nanoparticle formulation, significantly improve the stability, solubility, and bioavailability of quercetin, enabling targeted drug delivery. Despite the promising preclinical outcomes, the clinical translation of quercetin remains nascent, necessitating further rigorous research to validate its safety and efficacy in human subjects. In conclusion, while quercetin exhibits substantial anticancer properties and therapeutic potential, future studies should focus on expanding sample sizes, elucidating metabolic pathways, and conducting comprehensive clinical trials to inform its application in oncology.
Keywords: quercetin, nano-derivatives, cancer therapy, mechanisms of action, drug safety
Introduction
In 1857, the flavonol compound quercetin was named after Quercus as a natural plant compound. It is one of the most abundant flavonoids among flavonol compounds.1 Quercetin (molecular formula: C15H10O7) consists of two benzene rings and a benzopyran ring. The International Union of Pure and Applied Chemistry (IUPAC) chemical name of Quercetin (QUE) is 3,3′,4′,5,7-pentahydroxyflavone. QUE is widely found in botanical drugs, such as Ginkgo biloba, Lonicerae Japonicae Flos, Scutellariae Radix, Puerariae Radix, and Buglossoides Radix, with the highest content found in sophora flower.2 (Figure 1). The medical active structures of quercetin include QUE Glycosides, Methylated QUE, Sulfated QUE, QUE glucuronides, and QUE-nanoparticle composites such as Metal-QUE Complexes, QUE Nanoparticles, QUE Liposomes, and QUE Nano-micelles.3–5 In recent years, researchers have created innovative intelligent systems that integrate synthetic materials with flavonoid compounds,6–9 like QUE-magnetoliposome (Q-MLs), for use in magnetic hyperthermia and smart drug delivery for breast cancer. In vitro experiments indicate that Q-MLs have significant cytotoxic effects on MCF-7 breast cancer cells at various concentrations. Additionally, the application of a magnetic field significantly increases both the release rate of QUE and the cell death rate. Q-MLs effectively generate heat when subjected to an alternating magnetic field, thereby enhancing the anticancer effect of quercetin. However, because quercetin is a polyphenolic compound, it often produces non-specific effects. Existing in vivo studies have demonstrated that quercetin and its nano-derivatives can serve as adjuvant therapies for cancer, yielding significant therapeutic effects. However, the reliability of these studies requires further verification.10–12 This review analyzes the research progress on QUE and its nano-derivatives in cancer treatment worldwide from 2004 to 2024 through bibliometric. Based on the latest research hotspots, it explores the mechanism of action of QUE and its nano-derivatives in cancer treatment, and evaluates their safety and effectiveness as adjuvant therapy. QUE nano-derivatives have enhanced the bioavailability and targeted delivery of quercetin, exhibiting significant clinical application potential. It is recommended to expand the sample size in in vivo studies in future research, comprehensively explore the metabolic mechanisms and biological activities of QUE under various physiological conditions, and rigorously assess its long-term efficacy and safety.
![]() |
Figure 1 QUE molecular structure and medicinal plants. Adapted from Institute of Botany, Chinese Academy of Sciences. Plant photo bank of china (PPBC). https://ppbc.iplant.cn. Accessed Dec 15, 2024.13 |
The Physiochemical Properties of QUE
QUE has hydroxyl groups at positions 3, 5, 7, 3′ and 4′. Both QUE and its isomeric molecules belong to the flavonoid compounds with the basic nucleus of 2-phenylchromen. The molecule of QUE is a pentahydroxyflavone obtained by substituting the hydrogen atoms at the 3, 5, 7, 3′, and 4′ positions of the skeleton structure with hydroxyl groups (–OH). The various phenolic hydroxyl groups on this skeleton structure have certain activity and belong to active sites. The activity of –OH groups in the structure of QUE follows the sequence of 4′-, 3′-, 7-, 3-. Changing the positions of phenolic hydroxyl groups on QUE molecule alters the properties of the generated 14 isomeric molecules.14 Lower total energy of the molecule is associated with more stable structure. However, the B-ring of QUE molecule exhibits the strongest activity and the highest medicinal value.
QUE possesses certain inherent characteristics, such as low water-solubility (2.2 µg/mL), extensive first-pass metabolism, and low oral bioavailability.15 Previous research has revealed that QUE primarily binds to plasma proteins in the body, with albumin accounting for 99.4% of this binding, thereby reducing its cellular bioavailability.16 Additionally, QUE and its derivatives demonstrate stability in gastric acid and are absorbed in the upper segment of the small intestine. Following absorption, they undergo metabolism in various organs, including the small intestine, colon, liver, and kidneys. The highest accumulation of QUE’s metabolites has been observed in the lungs (in rats) and the liver and kidneys (in pigs), with the kidneys serving as a major excretory organ.17 Therefore, to optimize the clinical application of QUE, it is necessary to make structural modifications to enhance its efficacy and reduce potential toxic side effects.
Common Structural Modifications of QUE
QUE Glycosides
QUE glycosides, also known as flavonoid glycosides, can be isolated from a variety of plants, including Scutellaria Radix,18,19 Astragalus membranaceus,20,21 Salvia miltiorrhiza Hedyotis diffusa,22,23 and Pueraria lobata.24,25 Common isolation techniques encompass solvent extraction and chromatographic separation methods.
Hydroxyl groups (–OH) in QUE are replaced by glycosyl groups (–O–glycosyl). These are natural compounds formed by the combination of QUE with sugar molecules and represent the main form of QUE in human blood. Among them, QUE glucoside exhibits improved water-solubility, increasing its bioavailability and stability. Its absorption rate ranges from 3% to 17%, making it a potent antioxidant.2 Isomers of QUE glycosides, also known as isoquercitrin, possess better stability and water-solubility, making them more readily absorbed and utilized in the body.26,27
Methylation
Methylated QUE can be isolated from various plants, including Sorbaria sorbifolia and Jatropha curcas,28,29 through commonly employed separation techniques such as organic solvent extraction, chromatographic separation, and spectroscopic identification.
Methoxy groups are introduced onto the hydroxyl groups of QUE, resulting in the formation of methylated QUE derivatives. Previous research has shown that strong methylation occurs in the liver for 90–95% of QUE derivatives, which enhances their solubility and stability. In vitro experiments have demonstrated that methylated QUE, such as 3,4′,7-o-trimethylquercetin (34’7TMQ), can inhibit the migration and invasion of ovarian cancer cells without affecting cell proliferation.30
Sulfation
Sulfated QUE can be isolated from various plants, including Echinacea purpurea and Echinacea angustifolia,31–33 using conventional techniques such as organic solvent extraction, chromatographic separation, and spectroscopic identification.
Compounds formed by esterification of QUE molecules with sulfate groups (–SO4) are known as sulfated QUE derivatives. Sulfated QUE exhibits better water-solubility and facilitates the preparation of water-soluble drugs. Compared with pure QUE, it is easily absorbed and utilized by the human body.
Glucuronidation
QUE glucuronides can be isolated from plants such as Uncarina through conventional techniques, including organic solvent extraction, chromatographic separation, and spectroscopic identification. Hydroxyl groups on QUE molecules undergo chemical reactions with glucuronic acid molecules, resulting in the formation of QUE glucuronides. This esterification process involves the binding of QUE to glucuronic acid, leading to the formation of QUE glucuronide. Glucuronidation increases the solubility and water-solubility of QUE, facilitating its excretion and metabolism34 (Figure 2).
![]() |
Figure 2 Common Structural Modifications of QUE. |
Sulfonation and glucuronidation have been shown to reduce the ability of QUE to induce cell cycle arrest in tumor cells to a certain extent.14 Previous studies have revealed that the metabolites Q3S and Q3G, similar to the parent compound QUE, exhibit significant antiproliferative and cytotoxic effects on MCF-7 breast cancer cells, with a dose-dependent relationship and potency in the order of QUE > Q3S > Q3G.35 Therefore, it can be concluded that sulfonation and glucuronidation of QUE can attenuate its inherent anticancer effects.
QUE -Cyclodextrin Inclusion Complexes
To address the issue of low bioavailability of QUE, researchers have employed various strategies in recent years to enhance its solubility. For example, using cyclodextrins as carriers to form inclusion complexes has been shown to significantly improve the water solubility of QUE. Studies indicate that the inclusion complex formed between quercetin and β-cyclodextrin can increase its solubility up to 20 times the original level.36 Additionally, nanotechnology, such as nano-emulsions and nanoparticles, has also been widely applied to enhance the solubility and bioavailability of QUE.37,38
Properties and Classification of Cyclodextrins
Cyclodextrins are a class of cyclic oligosaccharides formed by glucose units linked by α-1,4-glycosidic bonds. Due to their unique molecular structure and properties, they have been widely used in drug delivery, the food industry, and biomedicine. Based on the number of glucose units in the cyclic structure, cyclodextrins can be classified into α-cyclodextrin, β-cyclodextrin, and γ-cyclodextrin. These different types of cyclodextrins exhibit significant differences in physicochemical properties, inclusion capacity, and biocompatibility, allowing them to play their respective advantages in various application scenarios.
α-Cyclodextrin
α-Cyclodextrin (α-CD) consists of six glucose units, with a molecular weight of approximately 972 daltons. The exterior of α-cyclodextrin is hydrophilic, while the interior is hydrophobic, allowing it to encapsulate small molecular compounds such as drug molecules and fragrances. Research shows that α-CD exhibits good biocompatibility and biodegradability in drug delivery systems, effectively enhancing the solubility and stability of drugs. For example, the inclusion complex formed between α-CD and certain drugs can significantly improve the bioavailability of the drugs, thereby enhancing their therapeutic effects.39 Additionally, α-CD has important application value in the food industry, as it can improve the taste of food and extend shelf life, while also serving as a carrier for nutritional components to help enhance the nutritional value of food. Studies have found that α-CD can effectively encapsulate fat-soluble vitamins, thereby increasing their solubility and bioavailability in aqueous phases.
β-CD
β-Cyclodextrin (β-CD) consists of seven glucose units, with a molecular weight of approximately 1135 daltons. β-CD is the most widely used type of cyclodextrin, known for its strong inclusion capacity, allowing it to encapsulate various drug molecules and bioactive components. Due to its larger cavity, β-CD can effectively encapsulate compounds with larger molecular weights, enhancing their solubility and stability. Studies indicate that the inclusion complex formed between β-CD and drugs can significantly improve the solubility and bioavailability of the drugs, thereby enhancing their pharmacological effects.40 In the field of biomedicine, β-CD is widely used in the preparation of drug carriers and nano drug delivery systems. For instance, the complex formed between β-CD and anticancer drugs can improve the distribution and targeting of the drugs in the body, thereby enhancing the antitumor effects.36 Additionally, β-CD is also used to improve the sensory characteristics of food and extend shelf life, showing good application prospects.41
γ- CD
γ-Cyclodextrin (γ-CD) consists of eight glucose units, with a molecular weight of approximately 1290 daltons. The larger cavity of γ-CD allows it to encapsulate larger molecular compounds, thus holding great potential for applications in drug delivery and biomedicine. Research has shown that γ-CD can effectively encapsulate various bioactive components, improving their solubility and stability, thereby enhancing their biological activity. In the food industry, γ-CD is used as a food additive to improve the taste and flavor of food while extending shelf life.42 Its good biocompatibility and low toxicity make γ-CD promising for applications in both food and pharmaceuticals.43 Furthermore, γ-CD also shows good application potential in the preparation of functional foods and health products, effectively enhancing the bioavailability and stability of nutritional components.44
Formation of QUE Inclusion Complexes with Different Types of Cyclodextrins
QUE is a natural flavonoid widely found in plants, gaining significant attention due to its good biological activity. However, the low solubility of QUE in water limits its effectiveness in drug development and clinical applications. Cyclodextrins, as a class of carbohydrate compounds with unique molecular structures, can form inclusion complexes, significantly enhancing the solubility and bioavailability of QUE. The following sections will explore the interactions between QUE and different types of cyclodextrins and the formation of their inclusion complexes.
Interaction with α-CD
α-CD is a cyclic oligosaccharide composed of six glucose units, with a smaller cavity suitable for encapsulating small molecules. Studies have shown that the inclusion complex formed between quercetin and α-CD is stable, primarily relying on hydrogen bonding and hydrophobic interactions. The interaction between the phenyl ring of quercetin and the cavity of α-CD allows quercetin to effectively embed into the structure of α-CD, thereby improving its solubility and stability.39 In one study, through nuclear magnetic resonance (NMR) and molecular dynamics simulations, researchers found that the binding energy between quercetin and α-CD was high, indicating good inclusion capacity. This inclusion not only improved the water solubility of quercetin but also enhanced its bioavailability, thereby increasing its efficacy in the body.40 Additionally, the QUE-α-CD inclusion complex demonstrated better antioxidant activity and cell protection in in vitro experiments, indicating its potential application value in drug delivery systems.
Interaction with β-CD
β-Cyclodextrin (β-CD) consists of seven glucose units and has a relatively larger cavity suitable for encapsulating larger molecules. The inclusion reaction between quercetin and β-CD is achieved through both physical adsorption and chemical bonding. Studies indicate that the inclusion complex formed between quercetin and β-CD significantly improves both solubility and bioavailability, especially showing superiority in drug delivery and targeted therapy.45 In one study, the QUE-β-CD inclusion complex was characterized using high-performance liquid chromatography (HPLC) and Fourier-transform infrared spectroscopy (FT-IR), with results showing that the formation of the complex significantly increased the solubility of quercetin and exhibited good stability under different pH conditions.41 Furthermore, the quercetin-β-CD inclusion complex demonstrated enhanced anticancer activity in cell experiments, suggesting its potential application in cancer treatment.
Interaction with γ-CD
γ-CD consists of eight glucose units, with a larger cavity suitable for encapsulating more complex molecules. The inclusion reaction between quercetin and γ-CD also exhibits good inclusion capacity. Research shows that the inclusion complex formed between quercetin and γ-CD has higher solubility and bioavailability in water, with significantly enhanced antioxidant activity.46 In one study, the formation of the quercetin-γ-CD inclusion complex was confirmed through X-ray diffraction (XRD) and thermogravimetric analysis (TGA), with results indicating that the thermal stability and structural integrity of the complex were significantly improved.42 Additionally, the quercetin-γ-CD inclusion complex demonstrated excellent cytotoxicity and antitumor effects in in vitro experiments, indicating its potential application prospects in cancer treatment.43
Pharmacokinetics of QUE-Cyclodextrin Inclusion Complexes
QUE-cyclodextrin inclusion complexes also exhibit advantages in pharmacokinetics. Compared to free QUE, the inclusion complexes have a faster absorption rate in the body and higher peak blood concentrations, indicating a significant improvement in their bioavailability. This phenomenon may be related to the structural characteristics of cyclodextrins, which allow for better penetration of cell membranes, facilitating the absorption of QUE.47
It is noteworthy that cyclodextrin inclusion complexes can extend the half-life of QUE in the body, thereby enhancing its therapeutic effects. An increasing number of clinical trials are beginning to focus on the use of QUE -cyclodextrin inclusion complexes. For example, clinical studies targeting breast cancer patients have shown that QUE -cyclodextrin inclusion complexes can significantly improve patients’ quality of life and reduce side effects caused by chemotherapy.48
However, there are still some controversies and differing viewpoints. For instance, some studies indicate that the stability and biocompatibility of the inclusion complexes may vary under different environmental conditions, posing challenges to the reliability of their clinical applications. Therefore, balancing the viewpoints from different studies, especially regarding the actual application effects and mechanisms of the inclusion complexes, has become an important task for future research.
Nanoparticle Structures of QUE and Their Therapeutic Applications
The combination of QUE with nanomaterials forms QUE–nanoparticle composites, which can enhance the solubility and stability of QUE. Additionally, functionalized nano-carriers can achieve controlled release and targeted delivery of QUE, increasing its drug concentration in tumor tissues and reducing toxic side effects on normal tissues.49
Metal–QUE Complexes
QUE has the ability to bind with metal ions, particularly transition metal ions such as iron and copper. These complexes may facilitate the formation of ROS through involvement in oxidation–reduction reactions. Previous research has shown that QUE-conjugated gold nanoparticles (AuNPs-Qu-5), when applied to MCF-7 and MDA-MB-231 cells, exhibit a reduction in cancer cell EMT, migration, invasion, and angiogenesis capabilities.50 Furthermore, they strongly inhibit the PI3K/AKT pathway, which may be associated with decreased EGFR activity.51 Additionally, AuNPs-Qu-5 promote the repair and regeneration of mammary gland-like epithelial structures.52
Nanomaterials formed by the combination of metal oxide zinc oxide (ZnO) with QUE, known as ZnO@Quercetin, possess a larger surface area and stronger electrochemical properties, which are key features for the generation of ROS. In vitro experiments have confirmed that ZnO@Quercetin effectively induces apoptosis in human ovarian cancer cells by generating ROS and permeating mitochondrial membranes.53
QUE-Loaded Nanoparticles
QUE’s solubility and stability can be enhanced through encapsulation in nanoparticles. Nanoparticles facilitate the passive accumulation of drugs at tumor sites, thereby enhancing drug penetration and retention through the enhanced permeability and retention (EPR) effect.54–56 Additionally, the larger surface area of nanoparticles enables high drug payloads and selective control of drug concentration and distribution within the tumor.57,58 Furthermore, their surface properties can be chemically modified to achieve active drug targeting.58
Nanoparticle-based drug delivery systems offer improved safety, cost-effectiveness, fewer side effects, and the ability to provide stronger activity with lower doses.59 Examples of such nanoparticles include polyethylene glycol nanoparticles, polymer nanoparticles, zinc oxide nanoparticles, and poly (lactic-co-glycolic acid) (PLGA) nanoparticles. It has been shown that QUE encapsulated in polymer nanoparticles exhibits enhanced stability, activity, and higher bioavailability.60,61 Previous research has confirmed that nanoparticle-based drug delivery systems effectively increase cellular uptake of drugs, thereby enhancing therapeutic effects and reducing toxicity.62,63
QUE Liposomes
Liposomes are composed of phospholipids and cholesterol, forming a bilayer membrane structure that can encapsulate QUE for targeted delivery and release, thereby increasing its solubility and bioavailability. QUE embedded in solid lipid nanoparticles increases the stability of the complex in the bloodstream and possesses high drug-loading capacity.64 In vitro experiments have demonstrated that polyethylene glycol liposomes loaded with QUE can induce apoptosis and cell cycle arrest in ovarian cancer cells sensitive or resistant to platinum compounds, showing significant inhibitory effects compared with free QUE.65
QUE Nano-Micelles
Nano-micelles are self-assembled structures that can encapsulate QUE, thereby enhancing its solubility and stability. They can also achieve targeted drug delivery by modulating their surface properties and incorporating targeting ligands.15 In vivo experiments have shown that HA-QU polymer micelles can downregulate the expression of p-gp in liver cancer cells, thereby reducing multidrug resistance. Additionally, they can target PTX-loaded micelles to liver cancer cells and enhance their antitumor effects.66
QUE nanocarriers can be synthesized using various preparation methods and have the potential to improve drug performance and enhance drug effects (Table 1). However, further research, evaluation, and clinical trials are required before the clinical application of these nanocarriers to ensure their safety, efficacy, and stability. Furthermore, nanocarrier technology is still in a stage of continuous development and exploration, and there may be emerging novel nanocarriers for QUE in the future.
![]() |
Table 1 The Comparative of Physicochemical Properties and Applications of Various QUE Nanomaterials |
Overview of Articles on QUE in Cancer Therapy
Using bibliometric analysis,67 we summarized QUE in cancer therapy literature from the Web of Science (WoS) Core Collection over the last decade. The search strategy was as follows: TS= (Quercetin OR Quercetin Nano-Derivatives) AND TS= (cancer OR tumor OR malignancy OR carcinoma OR neoplasm). A total of 4066 articles on QUE and Nano-Derivatives in cancers were retrieved on November 23, 2024 (excluding reviews and irrelevant article).
The number of published articles has increased year by year, with annual growth rate of 33.64% (Figure 3A). World collaborations between the USA and China were the most frequent, followed by those between Saudi Arabia and Egypt (Figure 3B). Studies from the United Kingdom had the highest number of citations, followed by those from USA (Figure 3C). China contributed the most articles and single-country publications (SCP; Figure 3D), and exhibited the highest multiple-country publications (MCP; Figure 3D).
The global increase in QUE-related articles indicates its crucial biological significance. Reviewing the 10 most cited articles (Table 2), we found that QUE has a broad impact on various aspects of tumor biology, including apoptosis and autophagy,68–74 tumor microenvironment,75 and tumor metastasis and migration.52,76 Among the retrieved articles, the five words that appeared most frequently in Keywords Plus were “quercetin”, “apoptosis”, “cancer”, “expression”, and “flavonoids” (Figure 4A). The thematic map in Figure 4B shows that QUE (1849 occurrences), apoptosis (1083 occurrences), cancer (1006 occurrences), flavonoids (931 occurrences) and expression (917 occurrences) were the most popular research topics. We analyzed the main effects and interactions of various factors, and observed that these thematic words were concentrated in studies on lung and breast cancer, anti-apoptosis, and research at the cellular level in vitro. We will discuss the research progress of QUE in cancer from these aspects (Figure 4C and D).
![]() |
Table 2 Top 10 Articles with the Highest Citations of QUE and Nano-Derivatives Articles |
The Mechanisms of Action of QUE in Cancer Therapy
Apoptosis and Autophagy
Oxidative Stress
QUE induces production of reactive oxygen species (ROS) in tumor cells through various mechanisms. In general, the antioxidative activity of QUE is thought to be substantially due to its catechol moiety.77 It activates NADPH oxidase (NOX), leading to ROS generation. QUE-3′-sulfate and QUE-3-glucuronide inhibits antioxidases such as superoxide dismutase (SOD), thereby increasing ROS accumulation and promoting tumor cell death.14 Furthermore, QUE glucuronides disrupts the mitochondrial respiratory chain, leading to increased ROS production.78 QUE Nanoparticles also acts as a radical scavenger, reducing ROS accumulation and preserving normal mitochondrial function.79,80
Mitochondrial-Mediated Signaling Pathway
The mitochondrial-mediated pathway is one of the primary mechanisms through which QUE induces apoptosis in tumor cells.30,81,82 3′-Methoxy QUE induces apoptosis in tumor cells through the upregulation of proapoptotic proteins (Bax and Bad) and downregulation of antiapoptotic proteins (Bcl-2 and Bcl-xL).83,84 This leads to increased mitochondrial membrane permeability, release of cytochromec, and activation of caspase family proteins, initiating cell death.85 Apoptosis induced by QUE occurs through both intrinsic mitochondrial pathways and caspase-dependent pathways.86 QUE also activates proapoptotic molecules such as caspase-3 and caspase-9, and downregulates antiapoptotic molecules.87–89 It induces DNA damage in mitochondrial genome, triggering ROS production and enhancing oxidative stress.90,91 Moreover, QUE inhibits signaling pathways related to mitochondria, suppressing tumor-cell proliferation and invasion. QUE also disrupts protein folding and maintenance functions, inducing apoptosis in tumor cells.92,93 It effectively binds to pyruvate dehydrogenase kinase 3 (PDK3), inhibiting its kinase activity and eradicating tumor cells.94 At high doses, QUE can induce apoptosis in various cancer cells via death domain pathways.95
MAPK Signaling Pathway
The mitogen-activated protein kinase (MAPK) pathway is a well-known extracellular signal–regulated pathway. QUE inhibits the MAPK pathway, reducing cell proliferation and inducing apoptosis.96,97 This pathway plays a role in promoting cell proliferation through the phosphorylation of mTORC1, ULK1, and autophagy-related proteins. Inhibition of the MAPK pathway by QUE-loaded nanoparticles leads to reduced cell proliferation and transcriptional activity, ultimately inducing apoptosis.98
PI3K/AKT Signaling Pathway
QUE inhibits the PI3K/AKT pathway, promoting apoptosis and reducing cancer-cell proliferation and invasion, such as Pentahydroxyflavone dihydrate-quercetin.99–102 It also acts on the TLR4–MyD88–PI3K complex, suppressing downstream signaling pathways and inhibiting the expression of inflammatory factors and apoptosis inhibitory proteins.103 By directly binding to tubulin, QUE causes cell cycle arrest.104 It also promotes tumor-cell death and increases chemo-sensitivity through the RAGE/PI3K/AKT/mTOR signaling axis.100
The Regulatory Effect on the Immune System
QUE regulates immune cells, promoting extracellular signal–regulated kinase 2 (Erk2) and mitogen-activated protein (MAP) kinases in PBMCs and T lymphocytes.2 It blocks the tyrosine phosphorylation of JAK2, TYK2, STAT3, and STAT4 induced by IL-12, reducing T-cell proliferation and Th1 differentiation.105 QUE enhances the immune system’s ability to kill tumor cells by regulating cell signaling pathways and protein expression, such as PD-L1 and interferon-γ. It also synergizes with recombinant human tumor necrosis factor–related apoptosis-inducing ligand (rhTRAIL), enhancing its proapoptotic action.106 QUE glycosides influencing the immune system and inflammatory responses by acting on leukocytes and targeting intracellular kinases and phosphatases.107 Figure 5
![]() |
Figure 5 QUE regulates tumor apoptosis and autophagy. |
Tumor Metastasis and Migration
Blocking the Cell Cycle Progression
G1/S Phase
QUE inhibits G1-phase cyclin-dependent kinases (CDKs), specifically CDK4 and CDK6, leading to G1/S cell-cycle arrest. It also downregulates cyclin D1/CDK4 and E/CDK2 and upregulates p21, inducing G1 cell-cycle arrest. Moreover, QUE induces DNA damage, inhibiting DNA synthesis and arresting cells in the S phase.14,108
G2/M Phase
QUE glucuronides interferes with M-phase CDKs (CDK1 and Cyclin B1), upregulates kinase inhibitors (p21, p27, and p53), and prevents cells from entering mitosis.108,109 It also decreases the levels of survivin protein, causing growth arrest in the G2/M phase.110–112 Significant growth inhibition of G2/M-phase tumor cells is observed at a concentration of 7 µg/mL, with an IC50 determined for cancer cells.1,113 QUE also blocks the progression of the G2/M phase in prostate cancer cells.84
Inhibition of Extracellular Matrix Degradation
The overexpression of proteolytic enzymes such as matrix metalloproteinases (MMPs) and urokinase-type plasminogen activator (uPA) are associated with tumor tissue remodeling and metastasis. Dual docetaxel/QUE-loaded nanoparticles inhibit the production and release of MMPs in tumor cells, such as MMP-2 and MMP-9.98 By inhibiting these proteases, QUE reduces degradation of the surrounding matrix, thereby weakening migration and invasion abilities of tumor cells.114
Regulation of Epithelial–Mesenchymal Transition (EMT)
EMT promotes tumor progression by enhancing the invasive capabilities of epithelial-derived tumors.115 It involves a reduction in epithelial markers (E-cadherin and ZO-1) and an increase in mesenchymal markers (α-SMA and vimentin).116 Transforming growth factor-β1 (TGF-β1) upregulates mesenchymal marker transcription factors and downregulates E-cadherin transcription.117 Additionally, EMT can contribute to chemo-resistance, reducing sensitivity to epidermal growth factor receptor (EGFR) inhibitors.118,119
QUE regulates the expression of transcription factors, such as the Snail superfamily and Twist, to inhibit tumor-cell migration and invasion.120 It also inhibits the increase of p-Smad2, associated with methotrexate-induced EMT, and has antiproliferative effects in various cancer types.114,121 In prostate cancer, QUE downregulates TGF-β–induced vimentin and N-cadherin, promotes E-cadherin expression, and regulates Wnt signaling to target EMT.84 In lung cancer, QUE inhibits metastasis through modulation of the Akt/MAPK/β-catenin signaling pathway and inhibits β-catenin nuclear translocation.122 In nasopharyngeal carcinoma, QUE activates the Hippo pathway by suppressing Yes-associated protein (YAP) expression, thereby suppressing EMT.123 In pancreatic cancer, QUE reduces TGF-β1 levels and regulates SHH and TGF-β/Smad signaling to inhibit EMT.124–126 In glioblastoma, Schiff base QUE derivatives inhibits the migration and invasion of glioma cells by targeting the GSK3-β/β-catenin/ZEB1 signaling pathway, associated with mesenchymal transition.127 Receptor-gamma (PPAR-γ) activation by quantum dots can counteract EMT partially, and QUE regulates the TGF-β1–induced EMT pathway by partially activating PPAR-γ.117
However, it should be noted that the combination of QUE with the epidermal growth factor receptor (EGFR) inhibitor erlotinib may counteract the antitumor effect of erlotinib.128
Inhibition of Angiogenesis
Angiogenesis plays a crucial role in tumor growth and metastasis. Tumor cells produce various growth factors that promote angiogenesis and metastasis, including vascular endothelial growth factor (VEGF), EGFR, fibroblast growth factor receptor (FGFR) and chemokines.114 QUE inhibits angiogenesis by reducing the production of angiogenic factors such as VEGF, EGFR, FGFR, and chemokines. It inhibits VEGF expression and NF-κB signaling pathway activity in nasopharyngeal carcinoma, exerting antitumor effects.129 Figure 6
![]() |
Figure 6 QUE inhibits tumor metastasis and migration. |
The Efficacy and Safety of QUE in vivo Studies
QUE exerts anti-tumor effects through mechanisms such as antioxidant activity, anti-inflammatory properties, and inhibition of tumor cell proliferation. However, as a polyphenolic compound, QUE is prone to exhibiting non-specific effects. Consequently, in vitro experiments or computer simulations often result in false positive outcomes, rendering the research findings meaningless. We have conducted a statistical and analytical review of in vivo and clinical studies (Table 3). Among these studies, the pharmacokinetics of QUE in animals demonstrated that different administration methods have an impact on the characteristics of QUE metabolites. Gastric tube administration was found to be more effective in enhancing the bioavailability of QUE, whereas free feeding may lead to high binding and methylation of QUE, thereby reducing its antioxidant activity. Furthermore, the metabolic process of QUE is likely to involve multiple enzymes, including glucuronosyltransferase and sulfotransferase, and the activities of these enzymes may vary across different tissues.77 In human studies, it was shown that QUE concentrations significantly increased within 30 minutes of ingestion, indicating significant absorption in the human duodenum and exhibiting two peaks of absorption. However, it is excreted in large quantities within 24 hours, indicating that quercetin has a rapid clearance rate in the blood, a short half-life, and is quickly metabolized in the liver, circulating in the form of methylated, glucuronide, and sulfate metabolites. Therefore, when using quercetin, special attention should be paid to liver and gastrointestinal toxicity. The absorption of QUE was not affected by gender or contraceptives, and the clinical study dosage of QUE was close to the dosage in drug formulations, within the range of 2.6mg-38.3mg daily dietary intake, demonstrating good safety.130
![]() |
Table 3 In Vivo and Clinical Studies of QUE |
The potential for in vivo toxicity has always been a great concern in the development of nanomedicines. To study any potential changes in organ morphology of tumor-bearing mice, we have systematically gathered and analyzed the histopathological characteristics of the heart, liver, spleen, lung, and kidney following treatment across all pertinent articles. These results provide evidence that the QUE nanomedicines in vivo cancer treatment induced no significant side effects in the treated mice. The low in vivo toxicity of QUE nanomedicines further paves the way for their potential clinical application.
Discussion
The rising incidence of cancer worldwide underscores a pressing need for effective therapeutic strategies. Cancer remains one of the leading causes of morbidity and mortality, with complex pathophysiological mechanisms that necessitate innovative approaches for treatment. Among various therapeutic modalities, chemotherapy and radiotherapy have been cornerstones in cancer management; however, they are often associated with significant adverse effects and resistance mechanisms that limit their efficacy. As a result, there is a growing interest in adjunctive therapies that can enhance the effectiveness of these traditional treatments while minimizing their harmful side effects, particularly in solid tumors such as breast and lung cancer, which are prevalent and challenging to treat.
Recent studies have highlighted quercetin, a natural flavonoid, as a promising candidate for enhancing cancer therapy. Quercetin has demonstrated multifaceted mechanisms of action, including modulation of various signaling pathways that can sensitize cancer cells to chemotherapeutic agents and radiotherapy.16,138,139 Furthermore, it exhibits protective properties for normal tissues, thereby reducing the toxicity associated with conventional treatments. This review encapsulates the latest advancements in understanding the mechanisms underlying quercetin’s anticancer effects, its structural optimization through nano-derivatives, and its potential clinical applications as an adjunct therapy in cancer treatment. In recent years, the structural modifications and nanoparticle formulations of QUE have garnered significant attention in the realm of cancer therapy due to their potential to enhance the bioavailability and therapeutic efficacy of this flavonoid. Structural modifications, such as glycosylation, methylation, sulfation, and glucuronidation, play critical roles in altering the pharmacokinetics and pharmacodynamics of QUE. For instance, QUE glycosides, such as quercetin-3-glucoside, exhibit improved solubility and stability, which translates to enhanced bioavailability. Studies suggest that glycosylation increases the absorption of QUE in the intestines, with quercetin-3-glucoside being absorbed more effectively than its aglycone counterpart, thereby promoting its therapeutic efficacy in vivo. Moreover, methylated derivatives of QUE have been shown to inhibit cancer cell migration and invasion, indicating that these modifications can confer enhanced anticancer properties. However, the metabolic processes involved in the conjugation of QUE may reduce its inherent anticancer effects, as seen with sulfated and glucuronidated forms. Therefore, understanding the implications of these structural modifications is essential for optimizing QUE’s therapeutic potential in clinical settings.
Nanocarriers have emerged as a transformative approach in improving the delivery and efficacy of QUE in cancer therapy. By encapsulating QUE within various nanoparticle structures, such as metal-QUE complexes, polymer nanoparticles, liposomes, and nano-micelles, researchers have successfully enhanced the solubility, stability, and targeted delivery of QUE to tumor sites. For instance, the combination of QUE with metal nanoparticles, like Zn, has been shown to potentiate its anticancer effects while concurrently reducing the viability of cancer cells. Additionally, polymeric nanoparticles facilitate the passive accumulation of QUE at tumor sites through the enhanced permeability and retention (EPR) effect, thus maximizing therapeutic outcomes while minimizing side effects. The encapsulation of QUE in liposomes has also demonstrated promising results in inducing apoptosis in various cancer cell lines, indicating the potential of this delivery system in overcoming the limitations associated with conventional QUE formulations. Collectively, these advancements underscore the necessity for continued research and development of QUE-based nanocarrier systems, as they hold great promise in addressing the challenges of cancer therapy and enhancing the clinical applicability of QUE.
Despite the promising potential of QUE in cancer therapy, additional research and clinical trials are needed to validate its efficacy and safety in clinical practice. There are various challenges that need to be addressed, such as the stability and targeted delivery of QUE, as well as the evaluation of its long-term effects and safety profile. In addition to enhancing the efficacy of cancer treatment, QUE exhibits protective effects on normal tissues.140,141 Chemotherapy drugs and radiotherapy can induce cardiotoxicity, pulmonary fibrosis, hepatotoxicity, nephrotoxicity, and bone marrow suppression. QUE has been shown to ameliorate these toxicities through various mechanisms. For example, QUE attenuates chemotherapy-induced cardiotoxicity by inhibiting NF-κB and NLRP3 inflammasome activation, and it improves cardiac energy metabolism through the modulation of the AMPK signaling pathway. Furthermore, QUE can interfere with DNA repair mechanisms, leading to increased sensitivity of tumor cells to radiotherapy.142 Therefore, QUE nanomedicine assisted cancer radiotherapy and chemotherapy is also the focus of our next research. Continued research and optimization of nanocarrier systems for QUE delivery will contribute to the development of clinically applicable formulations.
Conclusions
QUE and its nano-derivatives represent a promising avenue for enhancing cancer therapy through their multifaceted mechanisms of action. The growing body of literature reflects an increasing recognition of QUE’s potential to synergize with conventional treatments while mitigating their toxicity. However, to translate these findings into clinical practice, comprehensive studies are essential to assess the safety, efficacy, and optimal formulations of QUE-based therapies. At present, clinical studies have shown that the maximum dose for the human body is 93.5mg, which is in line with the daily dietary intake range of 2.6–3.83mg and demonstrates good safety Future research directions should prioritize larger, well-controlled clinical trials, alongside investigations into the metabolic pathways and biological activities of QUE nanomedicines under diverse physiological contexts. By addressing these critical gaps, we can better harness the therapeutic benefits of QUE nanomedicines and improve patient outcomes in cancer treatment.
Acknowledgments
We thank all the authors and for their contributions to this manuscript.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This work was the Natural Science Foundation of Shandong Province (No. ZR2024QH141), the Shandong Provincial Key Laboratory of Traditional Chinese Medicine [No. (2022)4].
Disclosure
The authors report no conflicts of interest in this work.
References
1. Maugeri A, Calderaro A, Patanè GT, et al. Targets involved in the anti-cancer activity of quercetin in breast, colorectal and liver neoplasms. IJMS. 2023;24(3):2952. doi:10.3390/ijms24032952
2. Li Y, Yao J, Han C, et al. Quercetin, inflammation and immunity. Nutrients. 2016;8(3):167. doi:10.3390/nu8030167
3. Al Saber M, Biswas P, Dey D, et al. A comprehensive review of recent advancements in cancer immunotherapy and generation of CAR T cell by CRISPR-Cas9. Processes. 2021;10(1):16. doi:10.3390/pr10010016
4. Biswas P, Dey D, Biswas PK, et al. A comprehensive analysis and anti-cancer activities of quercetin in ROS-mediated cancer and cancer stem cells. IJMS. 2022;23(19):11746. doi:10.3390/ijms231911746
5. Baral SK, Biswas P, Kaium M, et al. A comprehensive discussion in vaginal cancer based on mechanisms, treatments, risk factors and prevention. Front Oncol. 2022;12:883805. doi:10.3389/fonc.2022.883805
6. Alharbi HM, Alaghaz ANMA, Al Hujran TA, Eldin ZE, Elbeltagi S. A novel zingerone-loaded zinc MOF coated by niosome nanocomposites to enhance antimicrobial properties and apoptosis in breast cancer cells. Mater Today Commun. 2024;41:110245. doi:10.1016/j.mtcomm.2024.110245
7. Elbeltagi S, Saeedi AM, Eldin ZE, et al. Biosynthesis, characterization, magnetic hyperthermia, and in vitro toxicity evaluation of quercetin-loaded magnetoliposome lipid bilayer hybrid system on MCF-7 breast cancer. Biochim Biophys Acta. 2024;1868(3):130543. doi:10.1016/j.bbagen.2023.130543
8. Alneghery LM, Al-Zharani M, Nasr FA, et al. Fabrication and optimization of naringin-loaded MOF-5 encapsulated by liponiosomes as smart drug delivery, cytotoxicity, and apoptotic on breast cancer cells. Drug Dev Ind Pharm. 2024;1–14. doi:10.1080/03639045.2024.2388786
9. Elbeltagi S, Abdel Shakor AB, M. Alharbi H. Synergistic effects of quercetin-loaded CoFe 2 O 4 @Liposomes regulate DNA damage and apoptosis in MCF-7 cancer cells: based on biophysical magnetic hyperthermia. Drug Dev Ind Pharm. 2024;50(6):561–575. doi:10.1080/03639045.2024.2363231
10. Atala E, Fuentes J, Wehrhahn MJ, Speisky H. Quercetin and related flavonoids conserve their antioxidant properties despite undergoing chemical or enzymatic oxidation. Food Chem. 2017;234:479–485. doi:10.1016/j.foodchem.2017.05.023
11. Nile SH, Nile AS, Keum YS, Sharma K. Utilization of quercetin and quercetin glycosides from onion (Allium cepa L.) solid waste as an antioxidant, urease and xanthine oxidase inhibitors. Food Chem. 2017;235:119–126. doi:10.1016/j.foodchem.2017.05.043
12. Shala AL, Arduino I, Salihu MB, Denora N. Quercetin and its nano-formulations for brain tumor therapy—current developments and future perspectives for paediatric studies. Pharmaceutics. 2023;15(3):963. doi:10.3390/pharmaceutics15030963
13. Institute of Botany, Chinese Academy of Sciences. Plant photo bank of china (PPBC). https://ppbc.iplant.cn.
14. Wu Q, Needs PW, Lu Y, Kroon PA, Ren D, Yang X. Different antitumor effects of quercetin, quercetin-3′-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food Funct. 2018;9(3):1736–1746. doi:10.1039/C7FO01964E
15. Pillai SA, Bharatiya B, Casas M, et al. A multitechnique approach on adsorption, self-assembly and quercetin solubilization by Tetronics® micelles in aqueous solutions modulated by glycine. Colloids Surf, B. 2016;148:411–421. doi:10.1016/j.colsurfb.2016.09.014
16. Shen P, Lin W, Deng X, et al. Potential implications of quercetin in autoimmune diseases. Front Immunol. 2021;12:689044. doi:10.3389/fimmu.2021.689044
17. De Boer VCJ, Dihal AA, Der Woude H V, et al. Tissue distribution of quercetin in rats and pigs. J Nutr. 2005;135(7):1718–1725. doi:10.1093/jn/135.7.1718
18. Zhu X, Ding G, Ren S, Xi J, Liu K. The bioavailability, absorption, metabolism, and regulation of glucolipid metabolism disorders by quercetin and its important glycosides: a review. Food Chem. 2024;458:140262. doi:10.1016/j.foodchem.2024.140262
19. Pei T, Yan M, Huang Y, Wei Y, Martin C, Zhao Q. Specific flavonoids and their biosynthetic pathway in Scutellaria baicalensis. Front Plant Sci. 2022;13:866282. doi:10.3389/fpls.2022.866282
20. Ma XQ, Shi Q, Duan JA, Dong TTX, Tsim KWK. Chemical analysis of radix astragali (Huangqi) in China: a comparison with its adulterants and seasonal variations. J Agric Food Chem. 2002;50(17):4861–4866. doi:10.1021/jf0202279
21. Fu J, Wang Z, Huang L, et al. Review of the botanical characteristics, phytochemistry, and pharmacology of
22. Liu E, Zhou T, Li G, et al. Characterization and identification of iridoid glucosides, flavonoids and anthraquinones in Hedyotis diffusa by high‐performance liquid chromatography/electrospray ionization tandem mass spectrometry. J Separation Sci. 2012;35(2):263–272. doi:10.1002/jssc.201100780
23. Dai Y, Zhang H, Wang X, et al. Efficient strategies for preparative separation of iridoid glycosides and flavonoid glycosides from Hedyotis diffusa. J Separation Sci. 2023;46(10):2300029. doi:10.1002/jssc.202300029
24. Prasain JK, Reppert A, Jones K, Lila MA. Identification of isoflavone glycosides in Pueraria lobata cultures by tandem mass spectrometry. Phytochem Anal. 2006.
25. Xuan T, Liu Y, Liu R, et al. Advances in extraction, purification, and analysis techniques of the main components of kudzu root: a comprehensive review. Molecules. 2023;28(18):6577. doi:10.3390/molecules28186577
26. Manach C, Mazur A, Scalbert A. Polyphenols and prevention of cardiovascular diseases: current opinion in lipidology. Curr Opin Lipidol. 2005;16(1):77–84. doi:10.1097/00041433-200502000-00013
27. Harwood M, Danielewska-Nikiel B, Borzelleca JF, Flamm GW, Williams GM, Lines TC. 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(11):2179–2205. doi:10.1016/j.fct.2007.05.015
28. Zhumanova K, Lee G, Baiseitova A, et al. Inhibitory mechanism of O-methylated quercetins, highly potent β-secretase inhibitors isolated from Caragana balchaschensis (Kom.) Pojark. J Ethnopharmacol. 2021;272:113935. doi:10.1016/j.jep.2021.113935
29. Docampo-Palacios ML, Alvarez-Hernández A, Adiji O, et al. Glucuronidation of methylated quercetin derivatives: chemical and biochemical approaches. J Agric Food Chem. 2020;68(50):14790–14807. doi:10.1021/acs.jafc.0c04500
30. Shafabakhsh R, Asemi Z. Quercetin: a natural compound for ovarian cancer treatment. J Ovarian Res. 2019;12(1):55. doi:10.1186/s13048-019-0530-4
31. Valentová K, Káňová K, Di Meo F, et al. Chemoenzymatic preparation and biophysical properties of sulfated quercetin metabolites. IJMS. 2017;18(11):2231. doi:10.3390/ijms18112231
32. Sato R, Nishidono Y, Tanaka K. Comprehensive analysis of sulfated flavonoids in eclipta prostrata for quality evaluation. Molecules. 2024;29(20):4888. doi:10.3390/molecules29204888
33. Straßmann S, Passon M, Schieber A. Chemical hemisynthesis of sulfated cyanidin-3-o-glucoside and cyanidin metabolites. Molecules. 2021;26(8):2146. doi:10.3390/molecules26082146
34. Ader P. Bioavailability and metabolism of the flavonol quercetin in the pig. Free Radic Biol Med. 2000;28(7):1056–1067. doi:10.1016/S0891-5849(00)00195-7
35. Gong C, Yang Z, Zhang L, Wang Y, Gong W, Liu Y. Quercetin suppresses DNA double-strand break repair and enhances the radiosensitivity of human ovarian cancer cells via p53-dependent endoplasmic reticulum stress pathway. OTT. 2017;Volume 11:17–27. doi:10.2147/OTT.S147316
36. Alishahi M, Xiao R, Kreismanis M, et al. Antibacterial, anti-inflammatory, and antioxidant cotton-based wound dressing coated with chitosan/cyclodextrin–quercetin inclusion complex nanofibers. ACS Appl Bio Mater. 2024;7(8):5662–5678. doi:10.1021/acsabm.4c00751
37. Wu N, Zhang Y, Ren J, Zeng A, Liu J. Preparation of quercetin–nicotinamide cocrystals and their evaluation under in vivo and in vitro conditions. RSC Adv. 2020;10(37):21852–21859. doi:10.1039/D0RA03324C
38. Sun Y, Zhao J, Lu Y, et al. In Silico prediction of quercetin analogs for targeting Death-Associated Protein Kinase 1 (DAPK1) against alzheimer’s disease. CN. 2024;22(14):2353–2367. doi:10.2174/1570159X22666240515090434
39. Papakyriakopoulou P, Manta K, Kostantini C, et al. Nasal powders of quercetin-β-cyclodextrin derivatives complexes with mannitol/lecithin microparticles for nose-to-brain delivery: in vitro and ex vivo evaluation. Int J Pharm. 2021;607:121016. doi:10.1016/j.ijpharm.2021.121016
40. Sousa Filho LF, Barbosa Santos MM, Menezes PDP, Lima BDS, Souza Araújo AAD, De Oliveira ED. A novel quercetin/β-cyclodextrin transdermal gel, combined or not with therapeutic ultrasound, reduces oxidative stress after skeletal muscle injury. RSC Adv. 2021;11(45):27837–27844. doi:10.1039/D1RA04708F
41. Wangsawangrung N, Choipang C, Chaiarwut S, et al. Quercetin/Hydroxypropyl-β-cyclodextrin inclusion complex-loaded hydrogels for accelerated wound healing. Gels. 2022;8(9):573. doi:10.3390/gels8090573
42. Qiu C, Zhang Z, Li X, et al. Co-encapsulation of curcumin and quercetin with zein/HP-β-CD conjugates to enhance environmental resistance and antioxidant activity. Npj Sci Food. 2023;7(1):29. doi:10.1038/s41538-023-00186-2
43. Joshi H, Gupta DS, Kaur G, et al. Nanoformulations of quercetin for controlled delivery: a review of preclinical anticancer studies. Naunyn-Schmiedeberg’s Arch Pharmacol. 2023;396(12):3443–3458. doi:10.1007/s00210-023-02625-z
44. Zhang Y, Gong S, Liu L, et al. Cyclodextrin-coordinated liposome-in-gel for transcutaneous quercetin delivery for psoriasis treatment. ACS Appl Mater Interfaces. 2023;15(34):40228–40240. doi:10.1021/acsami.3c07582
45. Vakali V, Papadourakis M, Georgiou N, et al. Comparative interaction studies of quercetin with 2-Hydroxyl-propyl-β-cyclodextrin and 2,6-Methylated-β-cyclodextrin. Molecules. 2022;27(17):5490. doi:10.3390/molecules27175490
46. Wang Z, Zou W, Liu L, Wang M, Li F, Shen W. Characterization and bacteriostatic effects of β-cyclodextrin/quercetin inclusion compound nanofilms prepared by electrospinning. Food Chem. 2021;338:127980. doi:10.1016/j.foodchem.2020.127980
47. Adami BS, Diz FM, Oliveira Gonçalves GP, et al. Morphological and mechanical changes induced by quercetin in human T24 bladder cancer cells. Micron. 2021;151:103152. doi:10.1016/j.micron.2021.103152
48. Cid-Samamed A, Rakmai J, Mejuto JC, Simal-Gandara J, Astray G. Cyclodextrins inclusion complex: preparation methods, analytical techniques and food industry applications. Food Chem. 2022;384:132467. doi:10.1016/j.foodchem.2022.132467
49. Abbaszadeh S, Rashidipour M, Khosravi P, et al. Biocompatibility, cytotoxicity, antimicrobial and epigenetic effects of novel chitosan-based quercetin nanohydrogel in human cancer cells. IJN. 2020;Volume 15:5963–5975. doi:10.2147/IJN.S263013
50. Balakrishnan S, Mukherjee S, Das S, et al. Gold nanoparticles–conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt–mediated pathway in breast cancer cell lines (MCF‐7 and MDA‐MB‐231). Cell Biochem Funct. 2017;35(4):217–231. doi:10.1002/cbf.3266
51. Boehm JS, Zhao JJ, Yao J, et al. Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell. 2007;129(6):1065–1079. doi:10.1016/j.cell.2007.03.052
52. Balakrishnan S, Bhat FA, Raja Singh P, et al. Gold nanoparticle-conjugated quercetin inhibits epithelial-mesenchymal transition, angiogenesis and invasiveness via EGFR/VEGFR-2-mediated pathway in breast cancer. Cell Prolif. 2016;49(6):678–697. doi:10.1111/cpr.12296
53. Ramalingam V, Muthukumar Sathya P, Srivalli T, Mohan H. Synthesis of quercetin functionalized wurtzite type zinc oxide nanoparticles and their potential to regulate intrinsic apoptosis signaling pathway in human metastatic ovarian cancer. Life Sci. 2022;309:121022. doi:10.1016/j.lfs.2022.121022
54. Cao LB, Zeng S, Zhao W. Highly stable PEGylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles for the effective delivery of docetaxel in prostate cancers. Nanoscale Res Lett. 2016;11(1):305. doi:10.1186/s11671-016-1509-3
55. El-Gogary RI, Rubio N, Wang JTW, 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(2):1384–1401. doi:10.1021/nn405155b
56. Yin J, Hou Y, Song X, Wang P, Li Y. Cholate-modified polymer-lipid hybrid nanoparticles for oral delivery of quercetin to potentiate the antileukemic effect. IJN. 2019;Volume 14:4045–4057. doi:10.2147/IJN.S210057
57. Shitole AA, Sharma N, Giram P, et al. LHRH-conjugated, PEGylated, poly-lactide-co-glycolide nanocapsules for targeted delivery of combinational chemotherapeutic drugs Docetaxel and Quercetin for prostate cancer. Mater Sci Eng C. 2020;114:111035. doi:10.1016/j.msec.2020.111035
58. Li M, Tang Z, Zhang Y, et al. LHRH-peptide conjugated dextran nanoparticles for targeted delivery of cisplatin to breast cancer. J Mater Chem B. 2014;2(22):3490. doi:10.1039/c4tb00077c
59. Khan T, Gurav P. PhytoNanotechnology: enhancing delivery of plant based anti-cancer drugs. Front Pharmacol. 2018;8:1002. doi:10.3389/fphar.2017.01002
60. Roy S, Rhim JW. Fabrication of chitosan-based functional nanocomposite films: effect of quercetin-loaded chitosan nanoparticles. Food Hydrocoll. 2021;121:107065. doi:10.1016/j.foodhyd.2021.107065
61. Zou Y, Qian Y, Rong X, Cao K, McClements DJ, Hu K. Encapsulation of quercetin in biopolymer-coated zein nanoparticles: formation, stability, antioxidant capacity, and bioaccessibility. Food Hydrocoll. 2021;120:106980. doi:10.1016/j.foodhyd.2021.106980
62. Ramalingam V, Varunkumar K, Ravikumar V, Rajaram R. Production and structure elucidation of anticancer potential surfactin from marine actinomycete Micromonospora marina. Process Biochem. 2019;78:169–177. doi:10.1016/j.procbio.2019.01.002
63. Miller EM, Samec TM, Alexander-Bryant AA. Nanoparticle delivery systems to combat drug resistance in ovarian cancer. Nanomed Nanotechnol Biol Med. 2021;31:102309. doi:10.1016/j.nano.2020.102309
64. Zakaria N, Khalil SR, Awad A, Khairy GM. Quercetin reverses altered energy metabolism in the heart of rats receiving adriamycin chemotherapy. Cardiovasc Toxicol. 2018;18(2):109–119. doi:10.1007/s12012-017-9420-4
65. Long Q, Xie Y, Huang Y, 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(6):965–975. doi:10.1166/jbn.2013.1596
66. Xu C, Ding Y, Ni J, Yin L, Zhou J, Yao J. Tumor-targeted docetaxel-loaded hyaluronic acid-quercetin polymeric micelles with p-gp inhibitory property for hepatic cancer therapy. RSC Adv. 2016;6(33):27542–27556. doi:10.1039/C6RA00460A
67. Aria M, Cuccurullo C. bibliometrix: an R-tool for comprehensive science mapping analysis. J Informetrics. 2017;11(4):959–975. doi:10.1016/j.joi.2017.08.007
68. Srivastava S, Somasagara RR, Hegde M, et al. Quercetin, a natural flavonoid interacts with DNA, arrests cell cycle and causes tumor regression by activating mitochondrial pathway of apoptosis. Sci Rep. 2016;6(1):24049. doi:10.1038/srep24049
69. Hashemzaei M, Far AD, Yari A, et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol Rep. 2017;38(2):819–828. doi:10.3892/or.2017.5766
70. Granato M, Rizzello C, Gilardini Montani MS, et al. Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J Nutr Biochem. 2017;41:124–136. doi:10.1016/j.jnutbio.2016.12.011
71. Hazafa A, Rehman KU, Jahan N, Jabeen Z. The role of polyphenol (Flavonoids) compounds in the treatment of cancer cells. Nutr Cancer. 2020;72(3):386–397. doi:10.1080/01635581.2019.1637006
72. 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. 2014;113:15–24. doi:10.1016/j.colsurfb.2013.08.032
73. Saw CLL, Guo Y, Yang AY, et al. The berry constituents quercetin, kaempferol, and pterostilbene synergistically attenuate reactive oxygen species: involvement of the Nrf2-ARE signaling pathway. Food Chem Toxicol. 2014;72:303–311. doi:10.1016/j.fct.2014.07.038
74. Zhu Y, Tchkonia T, Fuhrmann‐Stroissnigg H, et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl‐2 family of anti‐apoptotic factors. Aging Cell. 2016;15(3):428–435. doi:10.1111/acel.12445
75. Hu K, Miao L, Goodwin TJ, Li J, Liu Q, Huang L. Quercetin remodels the tumor microenvironment to improve the permeation, retention, and antitumor effects of nanoparticles. ACS Nano. 2017;11(5):4916–4925. doi:10.1021/acsnano.7b01522
76. Jia L, Huang S, Yin X, Zan Y, Guo Y, Han L. Quercetin suppresses the mobility of breast cancer by suppressing glycolysis through Akt-mTOR pathway mediated autophagy induction. Life Sci. 2018;208:123–130. doi:10.1016/j.lfs.2018.07.027
77. Kawai Y, Saito S, Nishikawa T, Ishisaka A, Murota K, Terao J. Different profiles of quercetin metabolites in rat plasma: comparison of two administration methods. Biosci Biotechnol, Biochem. 2009;73(3):517–523. doi:10.1271/bbb.80516
78. Day AJ, Bao Y, Morgan MRA, Williamson G. Conjugation position of quercetin glucuronides and effect on biological activity. Free Radic Biol Med. 2000;29(12):1234–1243. doi:10.1016/S0891-5849(00)00416-0
79. Li Y, Xiao D, Li S, Chen Z, Liu S, Li J. Silver@quercetin nanoparticles with aggregation-induced emission for bioimaging in vitro and in vivo. IJMS. 2022;23(13):7413. doi:10.3390/ijms23137413
80. Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. CIA. 2018;Volume 13:757–772. doi:10.2147/CIA.S158513
81. Tan J, Wang B, Zhu L. Regulation of survivin and Bcl-2 in HepG2 cell apoptosis induced by quercetin. C&b. 2009;6(7):1101–1110. doi:10.1002/cbdv.200800141
82. Zhang Q, Zhao XH, Wang ZJ. Flavones and flavonols exert cytotoxic effects on a human oesophageal adenocarcinoma cell line (OE33) by causing G2/M arrest and inducing apoptosis. Food Chem Toxicol. 2008;46(6):2042–2053. doi:10.1016/j.fct.2008.01.049
83. Ahmed Youness R, Amr Assal R, Mohamed Ezzat S, Zakaria Gad M, Abdel Motaal A. A methoxylated quercetin glycoside harnesses HCC tumor progression in a TP53/miR-15/miR-16 dependent manner. Natural Product Research. 2020;34(10):1475–1480. doi:10.1080/14786419.2018.1509326
84. Baruah MM, Khandwekar AP, Sharma N. Quercetin modulates Wnt signaling components in prostate cancer cell line by inhibiting cell viability, migration, and metastases. Tumor Biol. 2016;37(10):14025–14034. doi:10.1007/s13277-016-5277-6
85. Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res. 2000;301(1):133–142. doi:10.1007/s004410000188
86. Liu Y, Gong W, Yang ZY, et al. Quercetin induces protective autophagy and apoptosis through ER stress via the p-STAT3/Bcl-2 axis in ovarian cancer. Apoptosis. 2017;22(4):544–557. doi:10.1007/s10495-016-1334-2
87. Lv XF, Wen RQ, Liu K, et al. Role and molecular mechanism of traditional Chinese medicine in preventing cardiotoxicity associated with chemoradiotherapy. Front Cardiovasc Med. 2022;9:1047700. doi:10.3389/fcvm.2022.1047700
88. Zhang Q, Zhao XH, Wang ZJ. Cytotoxicity of flavones and flavonols to a human esophageal squamous cell carcinoma cell line (KYSE-510) by induction of G2/M arrest and apoptosis. Toxicol in vitro. 2009;23(5):797–807. doi:10.1016/j.tiv.2009.04.007
89. Roos WP, Kaina B. DNA damage-induced cell death by apoptosis. Trends Mol Med. 2006;12(9):440–450. doi:10.1016/j.molmed.2006.07.007
90. Granado-Serrano AB, Martiín MA, Bravo L, Goya L, Ramos S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-Kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J Nutr. 2006;136(11):2715–2721. doi:10.1093/jn/136.11.2715
91. D’Archivio M, Filesi C, Varì R, Scazzocchio B, Masella R. Bioavailability of the polyphenols: status and controversies. IJMS. 2010;11(4):1321–1342. doi:10.3390/ijms11041321
92. Aalinkeel R, Bindukumar B, Reynolds JL, et al. The dietary bioflavonoid, quercetin, selectively induces apoptosis of prostate cancer cells by down-regulating the expression of heat shock protein 90. Prostate. 2008;68(16):1773–1789. doi:10.1002/pros.20845
93. Wang RE, Kao JLF, Hilliard CA, et al. Inhibition of heat shock induction of heat shock protein 70 and enhancement of heat shock protein 27 phosphorylation by quercetin derivatives. J Med Chem. 2009;52(7):1912–1921. doi:10.1021/jm801445c
94. Dahiya R, Mohammad T, Roy S, et al. Investigation of inhibitory potential of quercetin to the pyruvate dehydrogenase kinase 3: towards implications in anticancer therapy. Int J Biol Macromol. 2019;136:1076–1085. doi:10.1016/j.ijbiomac.2019.06.158
95. Siegelin MD, Reuss DE, Habel A, Rami A, Von Deimling A. Quercetin promotes degradation of survivin and thereby enhances death-receptor–mediated apoptosis in glioma cells. Neuro-Oncology. 2009;11(2):122–131. doi:10.1215/15228517-2008-085
96. Zhao YY, Chen LH, Huang L, et al. Cardiovascular protective effects of GLP-1: a focus on the MAPK signaling pathway. Biochem Cell Biol. 2022;100(1):9–16. doi:10.1139/bcb-2021-0365
97. Yue J, López JM. Understanding MAPK Signaling Pathways in Apoptosis. IJMS. 2020;21(7):2346. doi:10.3390/ijms21072346
98. Li J, Zhang J, Wang Y, et al. Synergistic inhibition of migration and invasion of breast cancer cells by dual docetaxel/quercetin-loaded nanoparticles via Akt/MMP-9 pathway. Int J Pharm. 2017;523(1):300–309. doi:10.1016/j.ijpharm.2017.03.040
99. Bureau G, Longpré F, Martinoli MG. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J Neurosci Res. 2008;86(2):403–410. doi:10.1002/jnr.21503
100. Lan CY, Chen SY, Kuo CW, Lu CC, Yen GC. Quercetin facilitates cell death and chemosensitivity through RAGE/PI3K/AKT/mTOR axis in human pancreatic cancer cells. J Food Drug Anal. 2019;27(4):887–896. doi:10.1016/j.jfda.2019.07.001
101. Mamani-Matsuda M, Kauss T, Al-Kharrat A, et al. Therapeutic and preventive properties of quercetin in experimental arthritis correlate with decreased macrophage inflammatory mediators. Biochem Pharmacol. 2006;72(10):1304–1310. doi:10.1016/j.bcp.2006.08.001
102. Chen WJ, Tsai JH, Hsu LS, Lin CL, Hong HM, Pan MH. Quercetin blocks the aggressive phenotype of triple negative breast cancer by inhibiting IGF1/IGF1R-mediated EMT program. J Food Drug Anal. 2021;29(1):98–112. doi:10.38212/2224-6614.3090
103. Endale M, Park SC, Kim S, et al. Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and IKK/NF-κB-induced inflammatory mediators production in RAW 264.7 cells. Immunobiology. 2013;218(12):1452–1467. doi:10.1016/j.imbio.2013.04.019
104. Glaviano A, Kandimalla R, Mendiola M. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. 2023;22(1):13. doi:10.1186/s12943-022-01699-2
105. Muthian G, Bright JJ. Quercetin, a flavonoid phytoestrogen, ameliorates experimental allergic encephalomyelitis by blocking IL-12 signaling through JAK-STAT pathway in T lymphocyte. J Clin Immunol. 2004;24(5):542–552. doi:10.1023/B:JOCI.0000040925.55682.a5
106. Manouchehri JM, Turner KA, Kalafatis M. TRAIL-induced apoptosis in TRAIL-resistant breast carcinoma through quercetin cotreatment. Breast Cancer. 2018;12:117822341774985.
107. Chirumbolo S. The role of quercetin, flavonols and flavones in modulating inflammatory cell function. IADT. 2010;9(4):263–285. doi:10.2174/187152810793358741
108. Jeong JH, An JY, Kwon YT, Rhee JG, Lee YJ. Effects of low dose quercetin: cancer cell-specific inhibition of cell cycle progression. J Cell Biochem. 2009;106(1):73–82. doi:10.1002/jcb.21977
109. Yang JH, Hsia TC, Kuo HM, et al. Inhibition of lung cancer cell growth by quercetin glucuronides via G2/M arrest and induction of apoptosis. Drug Metab Dispos. 2006;34(2):296–304. doi:10.1124/dmd.105.005280
110. Ren MX, Deng XH, Ai F, Yuan GY, Song HY. Effect of quercetin on the proliferation of the human ovarian cancer cell line SKOV-3 in vitro. Exp Ther Med. 2015;10(2):579–583. doi:10.3892/etm.2015.2536
111. Kuo PC, Liu HF, Chao JI. Survivin and p53 modulate quercetin-induced cell growth inhibition and apoptosis in human lung carcinoma cells. J Biol Chem. 2004;279(53):55875–55885. doi:10.1074/jbc.M407985200
112. Ong C, Tran E, Nguyen T, et al. Quercetin-induced growth inhibition and cell death in nasopharyngeal carcinoma cells are associated with increase in bad and hypophosphorylated retinoblastoma expressions. Oncol Rep. 2004;11(3):727–733.
113. Xu Z, Jia Y, Liu J, et al. Naringenin and quercetin exert contradictory cytoprotective and cytotoxic effects on tamoxifen-induced apoptosis in HepG2 cells. Nutrients. 2022;14(24):5394. doi:10.3390/nu14245394
114. Elumalai P, Ezhilarasan D, Raghunandhakumar S. Quercetin inhibits the epithelial to mesenchymal transition through suppressing Akt mediated nuclear translocation of β-catenin in lung cancer cell line. Nutr Cancer. 2022;74(5):1894–1906. doi:10.1080/01635581.2021.1957487
115. Moustakas A, Heldin P. TGFβ and matrix-regulated epithelial to mesenchymal transition. Biochim Biophys Acta. 2014;1840(8):2621–2634. doi:10.1016/j.bbagen.2014.02.004
116. Takano M, Yamamoto C, Yamaguchi K, Kawami M, Yumoto R. Analysis of TGF-β1- and drug-induced epithelial–mesenchymal transition in cultured alveolar epithelial cell line RLE/Abca3. Drug Metab Pharmacokinet. 2015;30(1):111–118. doi:10.1016/j.dmpk.2014.10.007
117. Berkmen YM, Lande A. Chest roentgenography as a window to the diagnosis of Takayasu’s arteritis. Am J Roentgenol Radium Ther Nucl Med. 1975;125(4):842–846. doi:10.2214/ajr.125.4.842
118. Zhu X, Chen L, Liu L, Niu X. EMT-mediated acquired EGFR-TKI resistance in NSCLC: mechanisms and strategies. Front Oncol. 2019;9:1044. doi:10.3389/fonc.2019.01044
119. Nurwidya F, Takahashi F, Murakami A, Takahashi K. Epithelial mesenchymal transition in drug resistance and metastasis of lung cancer. Cancer Res Treat. 2012;44(3):151–156. doi:10.4143/crt.2012.44.3.151
120. Kawami M, Harabayashi R, Miyamoto M, Harada R, Yumoto R, Takano M. Methotrexate-induced epithelial–mesenchymal transition in the alveolar epithelial cell line A549. Lung. 2016;194(6):923–930. doi:10.1007/s00408-016-9935-7
121. Sharma N, Raut PW, Baruah MM, Sharma A. Combination of quercetin and 2-methoxyestradiol inhibits epithelial–mesenchymal transition in PC-3 cell line via Wnt signaling pathway. Future Science OA. 2021;7(9):FSO747. doi:10.2144/fsoa-2021-0028
122. Ju L, Shan L, Yin B, Song Y. δ‐Catenin regulates proliferation and apoptosis in renal cell carcinoma via promoting β‐catenin nuclear localization and activating its downstream target genes. Cancer Med. 2020;9(6):2201–2212. doi:10.1002/cam4.2857
123. Li T, Li Y. Quercetin acts as a novel anti-cancer drug to suppress cancer aggressiveness and cisplatin-resistance in nasopharyngeal carcinoma (NPC) through regulating the yes-associated protein/Hippo signaling pathway. Immunobiology. 2023;228(2):152324. doi:10.1016/j.imbio.2022.152324
124. Guo Y, Tong Y, Zhu H. Quercetin suppresses pancreatic ductal adenocarcinoma progression via inhibition of SHH and TGF-β/Smad signaling pathways. Cell Biol Toxicol. 2021;37(3):479–496. doi:10.1007/s10565-020-09562-0
125. Saini F, Argent RH, Grabowska AM. Sonic hedgehog ligand: a role in formation of a mesenchymal niche in human pancreatic ductal adenocarcinoma. Cells. 2019;8(5):424. doi:10.3390/cells8050424
126. Wang Q, Wang F, Li X, Ma Z, Jiang D. Quercetin inhibits the amphiregulin/
127. Ballav S, Ranjan A, Basu S. Partial activation of PPAR‐ γ by synthesized quercetin derivatives modulates TGF‐ β 1‐induced EMT in lung cancer cells. Advanced Biology. 2023;7(10):2300037. doi:10.1002/adbi.202300037
128. Chan CY, Hong SC, Chang CM, Chen YH, Liao PC, Huang CY. Oral squamous cell carcinoma cells with acquired resistance to erlotinib are sensitive to anti-cancer effect of quercetin via pyruvate kinase M2 (PKM2). Cells. 2023;12(1):179. doi:10.3390/cells12010179
129. Huang DY, Dai ZR, Li WM, Wang RG, Yang SM. Inhibition of EGF expression and NF-κB activity by treatment with quercetin leads to suppression of angiogenesis in nasopharyngeal carcinoma. Saudi J Biol Sci. 2018;25(4):826–831. doi:10.1016/j.sjbs.2016.11.011
130. Erlund I, Kosonen T, Alfthan G, et al. Pharmacokinetics of quercetin from quercetin aglycone and rutin in healthy volunteers. Eur J Clin Pharmacol. 2000;56(8):545–553. doi:10.1007/s002280000197
131. Zamani M, Aghajanzadeh M, Sharafi A, Danafar H. In vivo study of miktoarm star copolymers as a promising nanocarrier to transfer hydrophobic chemotherapeutic agents to breast cancer tumor. J Drug Delivery Sci Technol. 2022;74:103500. doi:10.1016/j.jddst.2022.103500
132. Sadhukhan P, Kundu M, Chatterjee S, et al. Targeted delivery of quercetin via pH-responsive zinc oxide nanoparticles for breast cancer therapy. Mater Sci Eng C. 2019;100:129–140. doi:10.1016/j.msec.2019.02.096
133. Jain AK, Thanki K, Jain S. Novel self-nanoemulsifying formulation of quercetin: implications of pro-oxidant activity on the anticancer efficacy. Nanomed Nanotechnol Biol Med. 2014;10(5):e959–69. doi:10.1016/j.nano.2013.12.010
134. Zhao J, Liu J, Wei T, et al. Quercetin-loaded nanomicelles to circumvent human castration-resistant prostate cancer in vitro and in vivo. Nanoscale. 2016;8(9):5126–5138. doi:10.1039/C5NR08966B
135. Xu G, Li B, Wang T, et al. Enhancing the anti-ovarian cancer activity of quercetin using a self-assembling micelle and thermosensitive hydrogel drug delivery system. RSC Adv. 2018;8(38):21229–21242. doi:10.1039/C8RA03274B
136. Yadav N, Tripathi AK, Parveen A, Parveen S, Banerjee M. PLGA-quercetin nano-formulation inhibits cancer progression via mitochondrial dependent caspase-3,7 and independent FoxO1 activation with concomitant PI3K/AKT suppression. Pharmaceutics. 2022;14(7):1326. doi:10.3390/pharmaceutics14071326
137. Xu Z, Dang Y, Chen X. Quercetin 7-rhamnoside from Sorbaria sorbifolia exerts anti-hepatocellular carcinoma effect via DHRS13/apoptotic pathway. Phytomedicine. 2024;135:156031. doi:10.1016/j.phymed.2024.156031
138. Lou M, Zhang L-N, Ji P-G. Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/Caspase-3 signaling pathway in human neuroglioma cells: in vitro and in vivo. Biomed Pharmacother. 2016;84:1–9. doi:10.1016/j.biopha.2016.08.055
139. Islam MT, Tuday E, Allen S, et al. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell. 2023;22(2):e13767. doi:10.1111/acel.13767
140. Li L, Zhang M, Liu T, et al. Quercetin-ferrum nanoparticles enhance photothermal therapy by modulating the tumor immunosuppressive microenvironment. Acta Biomater. 2022;154:454–466. doi:10.1016/j.actbio.2022.10.008
141. Li F, Liu J, Tang S, et al. Quercetin regulates inflammation, oxidative stress, apoptosis, and mitochondrial structure and function in H9C2 cells by promoting PVT1 expression. Acta Histochem. 2021;123(8):151819. doi:10.1016/j.acthis.2021.151819
142. Zhou B, Yang Y, Pang X, Shi J, Jiang T, Zheng X. Quercetin inhibits DNA damage responses to induce apoptosis via SIRT5/PI3K/AKT pathway in non-small cell lung cancer. Biomed Pharmacother. 2023;165:115071. doi:10.1016/j.biopha.2023.115071
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.