Abstract
Cancer has remained a major global health challenge, with around 20 million new cases and 9.7 million fatalities recorded each year. Even though there has been recent progress in therapies such as radiotherapy, chemotherapy, immunotherapy, and gene therapy, cancer remains a major treatment challenge due to late diagnosis and difficulties in therapeutic effectiveness. Flavonoids, a substantial category of naturally occurring polyphenols, have received considerable interest in recent years for their potential involvement in cancer management and prevention, especially concerning breast cancer. These bioactive compounds, abundant in vegetables, fruits, and herbs, exhibit various therapeutic actions, including antioxidant, anti-inflammatory, and antimutagenic effects. The advanced therapeutic potential of flavonoids, when combined with FDA-approved medicines, offers synergistic effects and enhanced clinical results. Additionally, flavonoid-loaded nano-formulations, involving co-delivery systems, are being explored to increase solubility, stability, and bioavailability, enabling targeted delivery to cancer cells while reducing off-target adverse effects. This review examines the role of flavonoids in the prevention and management of breast cancer, focusing on their dietary sources, metabolism, and pharmacokinetic properties. Furthermore, we explore novel strategies, such as combination therapies with FDA-approved drugs and the application of flavonoid-based nanoformulations, which have the potential to enhance therapeutic outcomes. The clinical application of these strategies has the potential to improve breast cancer treatment and create new opportunities for the advancement of flavonoid-based therapies.
1. Introduction
Cancer is a serious disease that still lacks an ultimate cure. According to the World Health Organization (WHO), there are 20 million new cancer cases and 9.7 million people dying annually. Cancer has a significant death rate attributable to diagnostic delays and ineffective treatment modalities [1]. The present cancer statistics of 2024 have shown that cancer cases have remained steady in women and there has been a 1.2% drop in cancer cases in males as compared to the prior statistics, but the risk continues to exist [2]. The decline in cancer case numbers is attributed to advancements in several therapeutic approaches, including radiation therapy, chemotherapy, neoadjuvant therapy, immunotherapy, and gene therapy [3].
Flavonoids (taken from the Latin term flavus, indicating yellow, owing to their shade in nature) are a wide category of polyphenols with low molecular weight, usually found in almost all fruits and vegetables [4]. Natural flavonoids represent a family of over 15,000 chemicals that contribute to the colorful pigments of fruits, plants, vegetables, and medicinal herbs, attracting pollinators to support seed and spore germination and facilitating their growth and development. Flavonoids protect plants from diverse biotic and abiotic challenges, functioning as unique UV filters, signaling molecules, and antibacterial agents [5]. In dietary intake, the physicochemical characteristics of flavonoids determine their metabolic process, i.e., their digestion, absorption, and biotransformation [6]. In cancer cells, flavonoids exert multiple biological effects, affecting proliferation through cell cycle arrest, induction of apoptosis and necrosis and displaying antioxidant, anti-inflammatory, anti-mutagenic, and anti-neoplastic activities. Notably, flavonoids impact cell proliferation via a wide variety of processes, and some of them could interact with numerous cellular components such as intracellular proteins, growth regulators, metal enzymes, transcription genes, and genetic material [7,8]. Flavonoids, when used in combination therapy, enhance the effectiveness of anticancer agents by targeting multiple pathways and mitigating drug resistance. They may potentially function synergistically with conventional therapies, improving therapeutic outcomes while minimizing adverse effects. Furthermore, flavonoids help regulate the tumor microenvironment, facilitating comprehensive cancer treatment [9].
Flavonoids contain strong anticancer effects, but their clinical use poses significant limitations. They demonstrate low water solubility, minimal absorption, and quick metabolism. These pharmacokinetic limitations diminish their absorption and limit their therapeutic efficacy in cancer therapy [7]. To overcome these challenges, nanotechnologies have emerged as a viable strategy in cancer therapy. Due to their nanoscale dimensions and extensive surface area, nanoparticles exhibit effective therapeutic drug transport and enhanced infiltration into tumor vasculature that prevents tumor proliferation [10]. Preliminary clinical studies suggest that nanoparticle-mediated drug delivery enhances treatment efficacy and mitigates adverse effects associated with conventional small-molecule chemotherapy. Furthermore, the use of nanoparticles has not been linked to any novel detrimental consequences [11]. Instead, nano-formulations offer several benefits, including biocompatibility, prolonged drug release, protection of medicines from degradation in biological fluids, biodegradability, and targeted delivery to specific areas via simple administration techniques [12]. Research indicates that plant-derived chemicals, whether used alone or in combination with conventional chemotherapy, can be effectively co-loaded onto nanocarriers to enhance their anti-cancer efficacy [13,14]. In recent years, flavonoid-based nanotechnology has garnered considerable interest as a potential approach to combat cancer [15,16].
Several experimental studies highlight the significant potential of flavonoids in managing breast cancer. This review examines their function in breast cancer prevention and treatment, focusing on dietary sources, pharmacokinetic challenges, including bioavailability and metabolism, and their novel applications for combination therapies and nanoformulation delivery systems. Previous studies have mainly emphasised the anticancer potential of flavonoids or the function of nano-formulations as drug delivery methods. In comparison, this review offers a concentrated discussion on flavonoid-based combination therapy and their nano-formulations in breast cancer. It gives a comparative viewpoint that has not been discussed in existing literature.
2. Flavonoids as Natural Therapeutics: Mechanisms and Applications
2.1. Structural Insights into the Chemistry of Flavonoids
Flavonoids are an important class of polyphenolic substances of plant-generated secondary metabolites that contribute to the physiological properties of plant-based products [17]. Flavonoids constitute the primary substances found in foods derived from plants, including vegetables, fruits, nuts, legumes, and grains, while tea and wine serve as extensive sources of consumable flavonoids. Green vegetables, onions, berries, cherries, apples, soybeans, and citrus fruits are considered rich sources of dietary flavonoids [18]. Flavonoids are structurally made up of a C6-C3-C6 flavan backbone, which is made up of 15 carbon atoms organized in two phenyl rings (A and B) joined by a heterocyclic pyran ring (C). Flavonoids are divided into subgroups, including flavones, flavanones, flavonols, flavanols, flavanonols, isoflavones, and anthocyanidins, according to their ring structure and level of saturation [19]. While differences within a subtype result from different substitutions on rings A and B, these subtypes vary in the oxidation state and alteration pattern of ring C [20]. In dietary sources, flavonoids are mostly found as methylated derivatives, glycones, or glycosides. For example, benzene, which may exist in a dihydro form, is fused to an α-pyrone molecule with a six-membered heterocyclic ring in flavonols and flavanones. While the benzenoid group is located at the second carbon in flavonoids, it moves to the third position in isoflavonoids. The presence of a double bond at the C2-C3 location and a hydroxyl group at the third position in flavonols is a crucial structural difference between them and flavanones [21] (Figure 1).
2.2. Nature’s Reservoir: Sources of Flavonoids
Flavonoids are commonly present in tomatoes, mulberries, Amazon grapes, apples, and citrus fruits [22,23,24,25,26]. These flavonoid-rich fruits and vegetables possess a beneficial impact against breast cancer and exhibit chemopreventive potential. From the consumption of foods, flavonoids bind with proteins, showing the possibility of utilizing synthetic analogs as anti-cancer treatments [27]. Tomatoes are an excellent source of flavonoids and other phytochemicals, including phenolic compounds, carotenoids, vitamins, and tomatins (glycoalkaloids), which are beneficial for preventing malignancies and treating chronic degenerative conditions [28,29]. Several flavonoids in tomatoes exhibit anti-proliferative capabilities by promoting apoptosis in various types of cancer cells [30].
Additionally, quercetin-3-β-D-glucoside, isolated from tomatoes, is a powerful, stable, and non-toxic molecule with anti-proliferative effects on cancer cells [31,32]. Among them, quercetin and related flavonoids are particularly effective in preventing the development of breast cancer in MCF7 cell lines, resulting in low cytotoxicity [33]. Furthermore, polyphenols and flavonoids obtained from tomatoes interact with the regulation of multiple transcription factors. Their actions also alter the phenylpropanoid biosynthesis pathway, as phenylalanine is transformed into trans-cinnamic acid by phenylalanine ammonia-lyase (PAL). It is further accelerated by cinnamic acid 4-hydroxylase (C4H) to yield p-coumaric acid. This mechanism is related to the strategic accumulation of polyphenols and the use of phenylalanine under the shikimate pathway [34,35].
Mulberries, a flavonoid-rich fruit, have been extensively utilized in indigenous practices for their antioxidant effects [36]. Mulberries, a food rich in flavonoids, have been historically used in folk medicine due to their antioxidant properties. Polyphenolic substances found in mulberries, including isoquercitrin, chlorogenic acid, quercetin, astragalin, and kaempferol, showed efficacy in mitigating inflammation associated with obesity and type 2 diabetes [37]. Mulberries have anti-cancer properties against breast cancer cells by inducing apoptosis via pathways associated with PI3K, tumor protein p53, c-Jun N-terminal kinase, and nuclear factor-kappaB (NF-κB) [38,39]. Furthermore, chlorogenic acid, derived from mulberry leaves, is shown to inhibit steatohepatitis by alleviating oxidative stress [40]. It enhances the viability of HepG2 cells by modulating the Nrf-2 signaling system. It decreases inflammation by lowering the production of pro-inflammatory markers such as TNF-α, interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS), and NF-κB [41]. Amazon grapes, which contain phenolic compounds, are reported to enhance the activity of several sulfur-oxidizing enzymes, including glutathione and superoxide dismutase. Such compounds also interact with the production and growth of catalase, antioxidant enzymes, and detoxification [42,43,44]. Amazon grapes are a valuable source of polyphenolic compounds, which exhibit significant disease-preventing effects and have been found beneficial for human health. These compounds give protection to protein and DNA, prevent poor iron activity, and inhibit enzymes.
Furthermore, Amazon grapes have been found to suppress the in vitro activities of tyrosinase, acetylcholinesterase (AChE), and α-amylase, indicating their potential as a preventative therapy for several disorders [45]. The cancer prevention advantages of Amazon grapes are attributed to flavonoids, as well as constituents such as quercetin, catechins, kaempferol, epicatechins, resveratrol, and anthocyanins, which contribute to their chemo-preventive and anti-proliferative activities [46]. Apples are an important source of bioactive components, such as flavonoids, terpenoids, phenols, and carotenoids, which are commonly recognized for their ability to reduce cancer risk [47,48]. These fruits, rich in flavonoids, have been scientifically proven to possess anti-inflammatory, antioxidant, and anticancer properties (Figure 2). The polyphenolic compounds in apples, which include epicatechin, chlorogenic acid, coumaric acid, caffeic acid, quercetin, phlorizin, quercetin-3-glucoside, and phloretin, are significantly beneficial in minimizing the risk of diabetes, cardiovascular diseases, and cancer due to their phytochemical properties [49]. Additionally, quercetin, a flavonoid found in apples, exhibits significant antioxidant properties attributed to its flavonoid, polyphenol, and phytochemical components [50]. The concentration of these active phytochemicals is influenced by harvest timing, with potential degradation during storage [51].
Nobiletin, important for alterations in flavonoid concentration and antioxidant solubility, is reduced as citrus fruits mature and develop. This reduction has been observed in several biologically active compounds present in citrus fruits [52]. Polyphenols are present in various parts of citrus fruits, such as the peels, skin, pulp membrane, seeds, and juice. Such flavonoids produced from citrus have anti-fungal, anti-viral, and anti-bacterial properties [53]. Increased concentrations of flavonoids are mainly found in various varieties of grapefruit and orange, whereas fruits such as mandarin orange, sweet orange, and lemon possess comparatively lower concentrations. In several Asian nations, citrus fruits are frequently used for their antioxidant properties, typically due to the hesperidin that occurs in mandarins, oranges, and grapefruits [53,54].
Furthermore, the flavonoid glycosides present in citrus plants have been identified for their ability to mitigate free radical activity by interacting with hydrogen peroxide, hence protecting against cellular damage. These flavonoids are essential for capillary protection, anti-cancer activity, the mitigation of leg edema, and the alleviation of symptoms associated with hemophilia [55,56]. Citrus fruits, especially lemon, pomelo, mandarin, lime, and grapefruit, are highly abundant in flavonoids, mostly found in their peels. The flavonoids found in citrus peels have potent anti-cancer actions, owing to their free radical scavenging capabilities [57]. The therapeutic efficacy of specific flavonoids present in fruits depends on their mechanism of action and bioavailability, highlighting the need for further pharmacological research and epidemiological studies to elucidate their benefits in cancer therapy [58].
2.3. Overcoming the Challenges: Bioavailability and Metabolism
A significant difficulty with flavonoids is their limited bioavailability, which is essential for assuring their efficacy. The approach of bioavailability involves many phases, including liberation, absorption, distribution, metabolism, and elimination (LADME). Several factors, including molecular mass, chemical nature, esterification, glycosylation, and botanical source, influence the uptake of dietary flavonoids [59]. Flavonoids are primarily absorbed due to their solubility and permeability. Moreover, due to their significantly lower molecular weight and lipophilicity, flavonoid aglycones may readily cross the intestinal epithelium. However, flavonoid glycosides have exhibited reduced permeability, likely due to their increased hydrophilicity and higher molecular mass. Hydrophilic aglycone flavonoid glucosides are carried into the epithelium of the small intestine through the Na+-dependent glucose cotransporter. They undergo hydrolysis by cytosolic β-glucosidase or lactase-phlorizin hydrolase, a glucosidase located in the brush border membrane of the small intestine. Aglycones are absorbed through the small intestine following hydrolysis, while complex glycosides are transported to the colon for hydrolysis by bacterial enzymes, releasing flavonoid aglycones [60]. Several studies have evaluated the absorption kinetics of naringenin and its glycosides in rats. It has been shown that the absorption kinetics of naringin and naringenin were identical, but naringin showed a delay in absorption in the intestine, which causes decreased bioavailability [61].
Furthermore, after administering naringin, no glucoside was observed in the cecum, indicating successful absorption and bioavailability related to its glycosidic form [62]. Besides, research on the quercetin bioavailability and its glycosidic form upon oral administration to rats has suggested that the aglycone part possesses lower bioavailability (2.0%) in comparison to the quercetin-3-O-maltoside and quercetin-3-O-glucoside, which produced 30% and 12% bioavailability, respectively [63]. Furthermore, several flavonoid glycosides, such as puerarin 7-O-isomaltoside and puerarin 7-O-glucoside, have been reported to have increased plasma concentrations and prolonged blood residence times compared to the aglycone upon intravenous dosing [64]. A pharmacokinetic study involving a single dose was performed on 10 healthy participants under 50 years old to evaluate the 24-h absorption and elimination of phenolic acids, flavonoids, and proanthocyanidins with a cranberry juice formulation comprising 54% liquid. The juice predominantly contained anthocyanins, specifically 3-arabinoside, 3-galactoside, cyanidin-3-arabinoside, and peonidin-3-galactoside, along with flavonols like quercetin and hyperoside. The average concentration of phenolic compounds, comprising flavonols, phenolic acids, and flavanols, observed in plasma reached 34.2 g/mL at 8–10 h, with some concentration of quercetin, protocatechuic acid, and vanillic acid [65]. The delivery and local circulation of nutritional flavonoids are affected by the binding ability to plasma proteins, including hemoglobin [66]. Methylated flavonoids have a protein affinity that is 2 to 16 times superior to that of non-methylated flavonoids, principally attributable to enhanced hydrophobic contact with human plasma albumin and ovalbumen [67]. During absorption, flavonoids are considered xenobiotics in the body and are rapidly eliminated from the circulation by hepatic mechanisms, including methylation, sulfation, and glucuronidation.
Furthermore, they may be metabolized into lighter phenolic molecules [68]. Such metabolic processes seem to be as effective as P450-associated oxidation and potentially contribute to the metabolism of certain dietary components [69]. The colon is essential for the biological absorption and metabolic processes of dietary phenolic and polyphenolic substances, releasing distinct catabolites that affect the gut microbiome [70]. A significant quantity of flavonoid derivatives in urine signifies extensive metabolism of such chemicals from the colonic microflora [71]. Yang et al. (2022) indicated that the urinary recovery percentages of the glucoside or rhamnoglucoside forms of naringenin were smaller (31% and 14%, respectively) compared with the aglycone form (14%), claiming that more than 70% of the flavonoid is potentially available to produce specific physiological effects [62].
3. Epidemiological Studies: Role of Flavonoids in Breast Cancer Prevention
One of the case–control studies carried out by Ingram et al. suggested a correlation between phytoestrogen intake and breast cancer outcome, showing a notable decrease in breast cancer risk among women with higher consumption of phytoestrogens, specifically the lignan enterolactone and isoflavonic phytoestrogen equol. The consumption of phytoestrogens was estimated using urine excretion examination in 144 specimens. The results indicated that increased consumption of these substances may have a promising role in preventing breast cancer [72]. An examination of 250 urine specimens of Chinese women in Shanghai showed that increased excretion values for total lignans and isoflavonoids were linked to a lower chance of breast cancer. The modified risk ratio for women exhibiting higher excretion levels of both phytoestrogen types, compared to those with reduced excretion, was 0.28 (95% CI: 0.15–0.50). This substantiated the claim that elevated phytoestrogen consumption may have a preventive effect against breast cancer [73]. Another case–control study conducted in New York provides further evidence for this association by analyzing flavonoid consumption among 1434 breast cancer patients and 1440 controls. Participants completed a meal frequency questionnaire to assess their dietary habits over the past year. The findings indicated a significantly decreased risk of cancer in subjects consuming higher levels of flavonoids, including lignans, flavones, flavonols, and flavan-3-ols, especially in postmenopausal women [74]. Similar case–control research done by Tu et al. identified a strong and inverse correlation between flavone consumption and breast cancer development in a group of 820 breast cancer patients and 1548 controls. This research indicated a 13% decrease in breast cancer incidence per standard deviation (0.5 mg/day) with a rise in flavone consumption. Inverse relationships were also identified for anthocyanidins, flavan-3-ols, and flavonols [75].
In U.S.-based cohort research comprising 56,630 postmenopausal women, a meta-analysis by Ying Wang et al. evaluated the association between seven subtypes of dietary flavonoids and the possibility of estrogen receptor (ER)-related invasive postmenopausal breast cancer. During the period from inception till August 2021, a total of 2116 incidences of invasive breast cancer were identified. The research showed a moderate inverse correlation between flavone consumption and the total probability of developing breast cancer and a connection between flavan-3-ol administration and ER-negative rates of breast cancer [76]. Multiple meta-analyses have also shown the connection between soy isoflavone consumption and lower breast cancer incidence [77,78,79]. Additionally, research by Hui et al., which analyzed 9513 incidents and 181,906 controls across six prospective cohort studies and six case–control analyses, found that consumption of flavonols and flavones, rather than other flavonoid subtypes, was associated with a decreased breast cancer risk among postmenopausal women [80]. Furthermore, another meta-analysis examining five cohort experiments comprising 11,206 patients revealed that soy food intake may be associated with lower recurrence and mortality rates, specifically in individuals with ER+/PR+, ER-negative, and postmenopausal breast tumors [81].
4. Unlocking the Chemotherapeutic Potential of Flavonoids in Breast Cancer Treatment
Flavonoids and polyphenolic substances are considered promising agents in the therapeutic arena of breast cancer treatment based on their potential to regulate various physiological processes involved in cancer development [82,83]. These naturally occurring compounds in teas, vegetables, fruits, and herbs provide a wide range of biological actions, including antioxidant, anti-inflammatory, and anticancer effects [84,85] (Table 1). Quercetin, an interesting flavonoid, is commonly found in apples, onions, and leafy green plants. It has been thoroughly investigated for its anticancer potential [86,87,88]. In this context, quercetin has been depicted to suppress cell growth in breast cancer cell lines, such as MCF-7 and MDA-MB-231, by activating multiple signaling pathways. Studies have demonstrated that quercetin promotes apoptosis by modulating the Bcl2/Bax ratio and inhibiting the PI3K/AKT pathway [89,90,91]. In triple-negative breast cancer (TNBC) cells, quercetin reduces the expression of fatty acid synthase and β-catenin, suggesting its ability to inhibit metastasis by modulating epithelial–mesenchymal transition (EMT) signals [92,93].
Additionally, quercetin synergizes with chemotherapy drugs, including tamoxifen and doxorubicin, improving their potency [94,95]. Luteolin is another flavonoid exhibiting significant anti-cancer properties. It has been identified for suppressing tumor proliferation and triggering apoptosis in breast cancer cells [96,97]. Research indicates that luteolin, when combined with lapatinib, enhances the susceptibility of HER2-positive breast cancer cells to therapy, thereby increasing the effectiveness of lapatinib [98]. Furthermore, luteolin inhibits angiogenesis by hindering VEGF, thereby enhancing its efficacy as an anticancer agent [99]. Likewise, puerarin, extracted from the kudzu plant, has gained interest due to its phytoestrogenic properties [100,101]. It increases apoptotic levels in MCF-7 cell lines and has been shown to reduce cell adhesion and movement. Although its clinical use is limited by inadequate solubility, new technologies, especially nanotechnology, have helped enhance its bioavailability, thereby offering an alternative to conventional treatments [102].
Additionally, apigenin is found in various vegetables and fruits and has demonstrated the ability to disrupt cell cycle development in breast cancer cells. The mechanism comprises the modulation of critical signaling pathways, the inhibition of IL-6, and the prevention of tumorigenesis [103,104]. Apigenin has also been demonstrated to augment the efficacy and protective properties of doxorubicin against chemotherapy-induced toxicity [105,106]. A flavanone, specifically isoliquiritigenin, extracted from liquorice, has the potential to induce apoptosis and inhibit proliferation in breast cancer cells [107]. It has also been demonstrated to inhibit the PI3K/AKT pathway, resulting in decreased levels of cyclins and inhibition of cancer cell growth [108]. Structural variations of isoliquiritigenin have yielded derivatives with enhanced bioactivity, underscoring its potential as a therapeutic agent [109].
Furthermore, curcumin, extracted from turmeric, is regarded as one of the most thoroughly researched plant-derived chemicals for the management of breast cancer. It demonstrates a wide array of anticancer effects, encompassing the suppression of cell growth, cancer metastases, and angiogenesis [110,111]. Curcumin also enhances the effectiveness of chemotherapy, including metformin, by promoting apoptosis and reducing tumor proliferation [112]. Considering its intriguing effects, the limited bioavailability of curcumin has stimulated research into various methods of delivery and structural modifications to enhance its therapeutic efficacy [113,114].
Table 1.
Therapeutic effects of flavonoids in breast cancer treatment.
Flavonoid | In Vitro/In Vivo | Breast Cancer Cell Lines | Effects | References |
---|---|---|---|---|
Hesperidin; Apigenin; Quercetin (Propolis) | In vitro | MCF-7 | Accumulation in G0/G1 phase, cell cycle, proliferation, apoptosis | [115,116] |
Quercetin | In vitro | MCF-7Ca/TAM-R | Increase in ERα and inhibition of HER2 | [117] |
Nude/MCF-7 | MCF-7 | Inhibition of von Willebrand Factor (vWF); suppression of calcineurin activity; tumor microvessel density modulation; decrease in VEGF/VEGFR2 signaling and NFAT activation. | [118] | |
Hesperidin | In vitro | MCF-7 | Increase apoptosis via G0/G1 phase arrest, caspase-3 and caspase-9 upregulation, increase BAX activation, and inhibit BCL-2 expression. | [119] |
Luteolin | In vitro | MCF-7 | Increase apoptosis via G0/G1 phase arrest, caspase-8 and caspase-9 upregulation and inhibit BCL-2, pAKT, pIGF-1R, Erα expression. | [119] |
Nude/T47D | T47D | VEGF secretion and mRNA expression, tumor microvessel density, tumor-specific VEGF expression, and BAX levels | [120,121] | |
Nobiletin | In vitro | MCF-7 | Increase in CYP1 enzyme activity, elevation of CYP1A1 protein expression, upregulation of CYP1B1 mRNA levels, and G1 cell cycle arrest. | [122] |
Eupatorin | In vitro | MCF-7 | Increased apoptosis in G2/M phase; enhanced activity of BAX, caspase-9, and caspase-8; modulation of RAF-1 and inhibition of VEGFA, BCL2L11, CHK1, CHK2, HIF1A, and AKT | [123] |
Xanthohumol | In vitro | MCF-7 | Enhance apoptosis during G1 phase arrest while reducing the levels of pAKT (S473); pMAPK (T202/Y204); and phosphorylated ERα at multiple sites (S104/S106, S118, S167, S305, Y537). | [124] |
Silibinin | In vitro | MCF-7, T47D | Enhanced BAX expression; mitochondrial cytochrome c release; nuclear translocation of AIF; induction of autophagy; activation of caspase-8, and reduced BCL-2 expression; along with modulation of ERα and ERβ activity. | [125,126] |
Kaempferol | Nude/MCF-7 | MCF-7 | Increase cleaved PARP; BAX and downregulation of BCL-2, pAKT, pMEK1/2, pERK1/2 and pIRS-1. | [127,128] |
Chalcone; Licochalcone A | In vitro | MCF-7 | Induce plasma membrane damage; BAX upregulation; cleaved PARP, and CIDEA; downregulate G2/M and S cell cycle phases, cyclin-D1, and BCL-2. | [129] |
LW-214 (flavone) | Nude/MCF-7 | MCF-7 | Enhance BAX expression; cleaved PARP, caspase-9, ROS generation; mitochondrial cytochrome c release; nuclear translocation of AIF, pJNK, and pASK1 levels; reducing BCL-2 and TRX-1 expression. | [130] |
NSC 686288 (flavone) | In vitro | MCF-7 | Increase cleaved PARP, caspase-9, and ROS levels while decreasing AhR signaling along with the expression of CYP1A1 and CYP1B1. | [131] |
2′-Nitroflavone | In vitro | MCF-7 | Cytotoxicity | [132] |
Pentamethoxylated-flavone | In vitro | MCF-7 | Alters the expression of the BCL-2 protein and promotes cell death. | [133] |
Puerarin | In vitro | MCF-7, LPS | Decrease NF-κB p65, MMP-9; MMP-2; CCR7; CXCR4; VCAM-1; ICAM-1; TNFα; IL-6; pNF-κB p65; pIκBα; pERK1/2 Downregulate NF-κB MMP-9, p65, CCR7, MMP-2, VCAM-1, TNFα, CXCR4, ICAM-1, pIκBα, p65, IL-6, pNF-κB and pERK1/2. | [134] |
Calycosin | In vitro | MCF-7, T47D | Decrease FOXP3; MMP-9; VEGF, MMP-9; | [135] |
Orientin | In vitro | MCF-7, TPA | Reduce IL-8 levels; PKCα membrane translocation; pERK activation, and nuclear translocation of c-JUN, c-FOS, and STAT3. | [136] |
Corylin | In vitro | MCF-7 | Increase miR-34c and decrease LINC00963 mRNAMMP-9; cytotoxicity | [137] |
Hinokiflavone | In vitro | MCF-7 | Decrease MMP-9; cytotoxicity | [138] |
3,6-Dihydroxy flavone | In vitro | MCF-7 | Upregulation of E-cadherin with downregulation of SNAIL, TWIST, SLUG, N-cadherin, NOTCH1, and NICD. | [139] |
LFG-500 (Flavone) | MMTV-PyMT transgenic mice | MCF-7 | Upregulation ZO-1; E-cadherin; pYAP; pMST1/2; pLATS1/2 and reduction in N-cadherin; vimentin; SLUG; SNAIL; YAP; ILK | [140] |
Hispidulin | In vitro | MCF-7 | Upregulation of E-cadherin and downregulation of occludin; pSMAD2/3 | [141] |
Calycosin | In vitro | Nude/T47D | Upregulation of E-cadherin and downregulation of N-cadherin, vimentin, CD147, MMP-2, MMP-9, and BATF. | [142] |
2′-Hydroxy flavanone | Nude/MCF-7 | MCF-7 | Upregulation of E-cadherin and downregulation of vimentin, along with modulation of RLIP76 and ERα expression. | [143,144] |
Kaempferol | In vitro | MCF-7 | Upregulation of E-cadherin and downregulation of N-cadherin, SNAIL, SLUG, cathepsin D, MMP-9, and MMP-2. | [127,128] |
Wogonoside | Nude/MCF-7 | MCF-7 | Downregulation of VEGF expression; inhibition of VEGF promoter activity; suppression of endothelial cell (EC) migration; reduction in EC invasion, and impairment of tubulogenesis. | [145] |
Jaceidin | Swiss albino /Ehrlich Ascites Carcinoma cells | MCF-7 | serum VEGF | [146] |
There are numerous mechanisms of action of flavonoids that demonstrate their therapeutic value in breast cancer therapy. Particularly, their capacity to control the cell cycle and promote apoptosis generates interesting credentials. Some of the compounds, including luteolin, quercetin, and hesperidin, promote G0/G1 phase arrest and downregulate anti-apoptotic proteins, such as BCL-2, while upregulating pro-apoptotic markers, including BAX, caspase-3, and caspase-9 [115,116,119,123,125,126]. These outcomes show their ability to promote programmed cell death and inhibit the growth of cancer cells. Flavonoids also display potent anti-angiogenic and anti-metastatic effects. Kaempferol and wogonoside, for this reason, inhibit VEGF expression, which in turn decreases endothelial cell migration and tumor microvessel density [127,128,145]. Hispidulin and kaempferol are two flavonoids that prevent the epithelial–mesenchymal transition (EMT) by reducing the levels of N-cadherin, SNAIL, and SLUG and increasing E-cadherin [141,147]. Their ability to inhibit both angiogenesis and metastasis demonstrates their diverse function in preventing the growth of tumours.
Furthermore, certain flavonoids, including quercetin and nobiletin, target specific pathways, such as tamoxifen resistance or HER2 suppression, to overcome treatment resistance in breast cancer [122]. Others, such as LW-214 and NSC 686288, enhance apoptosis without harming healthy tissues by specifically generating reactive oxygen species (ROS) in cancer cells [130,131]. These results highlight how flavonoids can be utilized to address various cancer hallmarks, making them beneficial substances for potential future treatment approaches.
Moreover, polyphenols, a diverse category of plant compounds characterized by numerous phenolic hydroxyl groups, are recognized for their unique chemical and biological properties. Polyphenols, including tannins, flavonoids, and phenolic acids, are prevalent in plant-based foods and offer various health benefits. They are thought to significantly contribute to the alleviation of oxidative stress, which is directly associated with cancer development [148,149,150,151]. Epigallocatechin gallate (EGCG), a prominent polyphenol in green tea, demonstrates significant anticancer efficacy. EGCG has been shown to have the ability to regulate oxidative damage, inflammatory reactions, and the growth of cancer cells [152]. It inhibits the development of cell cycles by blocking cells in the G1/S or G2/M phases, thereby successfully decreasing in vivo tumor proliferation [153]. EGCG exhibits a notable anti-angiogenic effect by inhibiting the induction of NF-κB and HIF-1α, while concurrently reducing VEGF expression, thereby inhibiting the generation of new blood vessels essential for tumour proliferation [154,155]. Another non-flavonoid polyphenol is resveratrol, which can be obtained from peanuts, red wine, and grapes. It is well-known for its capacity to prevent the growth of cancer cells and its anti-inflammatory properties [156,157]. Resveratrol enables breast cancer cells to undergo apoptosis and increases the efficacy of chemotherapeutic drugs. It also enhances the effectiveness of traditional treatments by targeting several key points implicated in cancer signaling pathways [158,159,160]. Another class of flavonoid compounds, called anthocyanins, which give colors to various fruits and vegetables, also exhibit strong anti-cancer effects. According to study findings, anthocyanins, like cyanidin-3-glucoside, may prevent the growth of breast cancer cells by causing apoptosis and inhibiting cyclin-dependent kinases [161,162,163]. They improve antioxidant capacity and alter important signaling pathways, like the PI3K/AKT pathway [164,165,166] (Figure 3). A flavonoid present in many fruits and vegetables, kaempferol has proven anti-cancer properties. Kaempferol, through processes like cell cycle arrest and ER suppression, prevents cell division and triggers apoptosis [147,148,167,168]. Interestingly, the establishment of nanosuspensions has improved the therapeutic efficacy of kaempferol by addressing its poor bioavailability [169,170]. Some of the compounds and their derivatives have undergone evaluation in clinical trials for the management of breast cancer (Table 2).
Clinical research exploring flavonoids and their combinations with anticancer agents indicates their therapeutic efficacy in breast cancer management. For example, genistein has been studied for its potential in preventing breast carcinoma and as an adjunct to gemcitabine in metastatic breast cancer [171]. AFP464, a derivative of aminoflavone, has been explored in estrogen receptor-positive breast cancer and solid tumours that exhibit resistance to conventional therapy [172]. ME-344, a synthetic flavonoid-like compound, has anti-angiogenic characteristics and modifies mitochondrial metabolism in HER2-negative breast cancer [173]. Extracts from plants, such as watercress, have been evaluated for their effects on chemotherapy results and the mitigation of DNA damage [174]. S-equol, an ERβ agonist, is being studied for its effectiveness in triple-negative breast cancer, highlighting the increasing focus on flavonoid-based approaches for targeted breast cancer treatment [175].
Recent trends in intellectual property emphasise the translational potential of flavonoid-based combination treatments for breast cancer. A number of patents have been published and granted, focused on the combination of flavonoids with kinase inhibitors, chemotherapeutic drugs, CDK inhibitors, and radiotherapy. These patents highlight various dosage forms, such as oral, intravenous, and topical preparations, to improve therapeutic potential and reduce toxicity. These results highlight the increasing acknowledgement of flavonoids as significant adjuvants to recent cancer treatment and provide perspectives on potential clinical and commercial uses (Table 3).
5. Synergizing Flavonoids with Synthetic Drugs in Combination Therapy
Natural products originating from the basic chemical constituents of plant material comprise quinonoids, alkaloids, flavonoids, essential oils, polysaccharides, coumarins, terpenoids, and saponins. Multiple studies have demonstrated that plant-derived natural products possess multiple physiological and therapeutic effects, including anti-inflammatory, neuroprotective, antioxidant, antiviral, and cardioprotective effects [176,177,178]. These natural substances are obtained from medicinal herbs globally and are very commonly studied as potential therapeutic interventions. Multiple in vitro and in vivo tests, including some clinical trials, have shown the chemopreventive and anticarcinogenic functions of natural substances against multiple cancers [179,180,181,182]. Intracellular mechanisms linked to tumour formation, such as tumour growth, including cell proliferation, DNA repair, cell differentiation, carcinogen metabolism, apoptosis, angiogenesis, and progression, are possibly linked to the pathways generating the anti-cancer actions [47,183]. Scientific studies on herbal substances against various cancer variants, including lung, breast, and prostate cancer, continue to be explored. As compared to the above-mentioned three kinds of cancers, significantly, a lot more studies have been conducted on breast cancer concerning herbal compounds. Several natural substances exhibit anti-cancer properties for breast cancer cell lines, offering another possibility of treatment for this specific type of cancer. Natural remedies used in combination with chemotherapy medicines may exhibit synergistic or additive effects [184,185]. The statistical concept of therapeutic synergies can be analysed using the combination index (CI), a concept of Chou–Talalay, which proposes whether a combination has an additive effect (CI = 1), synergistic effect (CI < 1), or antagonistic effect (CI > 1) [186]. Moreover, the harmful impact decreased and the drug resistance improved in the case of combination treatments.
Several in vitro and in vivo tests have examined the combined benefits of flavonoids and phenolic compounds along with different anti-cancer drugs, offering novel insights into their potential use in cancer treatment. The anti-cancer properties of various drugs, including doxorubicin, paclitaxel, 5-fluorouracil, and tamoxifen, are enhanced by flavonoids such as luteolin, quercetin, genistein, curcumin, and naringenin. These combinations target numerous molecular pathways, including angiogenesis, cell cycle control, apoptosis, and metastasis inhibition, that are implicated in the growth of cancer. In particular, luteolin and tamoxifen together inhibit Ras expression, which causes tamoxifen-resistant breast cancer cells to undergo apoptosis [187]. By increasing pro-apoptotic proteins like Bax and reducing anti-apoptotic proteins like Bcl-2, quercetin has also been reported in MDA-MB-231 and MCF-7 cell lines to increase the cytotoxicity of docetaxel and 5-fluorouracil, thus enhancing their therapeutic properties [188,189].
Furthermore, research has shown that genistein, an isoflavonoid, increases the cytotoxicity of treatments like tamoxifen and cisplatin and overcomes drug resistance in breast cancer models [190,191]. Another commonly used natural phenol, curcumin, was recently studied for the possible increase in the potency of drugs, including paclitaxel and doxorubicin. Curcumin has been proven in various studies to stimulate apoptosis, reduce the AKT/mTOR pathway, and initiate cell cycle arrest, thus reducing cancer cell proliferation [192,193,194,195]. Interestingly, in vivo studies show that curcumin may also synergize with chemotherapy treatments to decrease tumour development, improve the overall therapeutic outcome, and reduce the adverse effects of chemotherapeutic medications [192,196,197]. Likewise, natural substances like naringenin and naringin have demonstrated a remarkable potential to overcome chemoresistance and boost the anticancer properties of doxorubicin and 5-fluorouracil. Naringenin has been demonstrated to boost caspase activity, enhance the Bax/Bcl-2 ratio, and elevate cytokine production in several breast cancer models, hence stimulating therapeutic efficacy when combined with chemotherapeutic drugs [198,199]. These results emphasize the significance of flavonoids and phenolic substances in combination treatments for breast cancer. Their capacity to alter critical cancer-related processes, such as apoptosis, oxidative stress, and drug resistance processes, provides a viable technique for increasing the efficiency of conventional chemotherapy. The increasing number of findings shows that further clinical trials on these combination approaches may provide a new route for more potent, customized cancer therapies (Table 4).
The combined use of flavonoids with synthetic anti-cancer medicines indicates great potential in increasing treatment results via several mechanisms. Luteolin synergizes with tamoxifen to reduce Ras expression and initiate apoptosis in tamoxifen-resistant breast cancer cells [187]. Additionally, its combination with doxorubicin and paclitaxel highlights decreased cell survival and downregulated BCL-2 anti-apoptotic marker [200,201]. Quercetin, another important flavonoid, boosts the efficiency of docetaxel and letrozole by raising pro-apoptotic markers, including BAX and p53, while inhibiting proteins associated with cancer cell viability and migration, including STAT3, AKT, and MMPs [188,202]. Co-administration of curcumin with medications including docetaxel, sorafenib, and carboplatin underlines its potential in altering apoptotic pathways, ROS-induced DNA damage, and the tumor microenvironment, notably in triple-negative breast cancer (TNBC). Its anti-inflammatory and anti-metastatic capabilities, obtained by lowering COX-2 pathways and vimentin phases, further underscore its promise in combinatorial therapy [192,193,197]. Certain flavonoids, including naringenin and hesperidin, boost the efficiency of medications like doxorubicin and tamoxifen, as indicated by elevated pro-apoptotic gene expression and decreased tumor growth indicators in both in vitro and in vivo investigations [214,225]. Besides, the novel chemicals like xanthohumol and garcinol exhibit potential benefits in overcoming drug resistance and decreasing metastasis. In particular, xanthohumol efficiently reverses doxorubicin resistance by decreasing STAT3 and EGFR signaling, whereas garcinol reduces paclitaxel-induced pre-metastatic signaling via NF-κB/Twist1 pathways [211,213]. Furthermore, proanthocyanidins and mangiferin improve the action of 5-fluorouracil and doxorubicin, respectively, by boosting ROS generation, triggering cell cycle arrest, and reducing drug resistance proteins such as P-gp [219]. This study emphasizes the potential of flavonoid-based combinations to not only enhance treatment effectiveness but also reduce drug resistance in breast cancer, providing a strong case for additional clinical trials.
6. Advancement in Flavonoid Nano-Formulations and Codelivery Strategies in Breast Cancer Prevention
The application of flavonoids in nanoparticle-derived drug delivery systems (DDSs) facilitates in situ targeting of cancer tissues, notably increasing drug concentration at the site of cancer while minimizing systemic toxicity, avoiding vascular injury, and lowering the extent of drug administration [230]. The anti-cancer efficacy of flavonoid-based nanotechnology is linked to various mechanisms, such as the stimulation of caspase enzymes, the introduction of cellular arrest, the decrease in cancer vascularization, the prevention of tumour cell proliferation and dissemination, the introduction of mitochondrial dysfunction, and apoptosis [231] (Figure 4). Different nanotechnologies, including phytosomes, liposomes, solid-lipid nanoparticles, nanocapsules, nanoemulsions, polymeric nanoparticles, lipid-based carriers, and metal-based nanoparticles, have been investigated for encapsulating polyphenols to improve their therapeutic efficacy in cancer therapy [232] (Table 5).
Polymeric nanoformulations, notably quercetin-loaded vehicles containing hyaluronic acid (230–480 nm) [233], chitosan (<200 nm) [234], and MPEG-PLA (155 ± 3 nm) [235], have demonstrated effectiveness in tumour suppression through the passive increased permeability and retention (EPR) effect and selective targeting via CD44 receptors. Moreover, mesoporous silica-based quercetin nanoparticles (<200 nm) regulate AKT and Bax signaling pathways to trigger apoptosis and cell cycle arrest [236], whereas EGCG-PEG nanoparticles (140–182 nm) augment tumor suppression by elevating p21, PTEN, and Bax expression while downregulating oncogenic markers such as p-AKT and Cyclin D1 [237]. Fisetin-PLA nanoparticles (225 ± 4 nm) have been documented to enhance bioavailability and exhibit anticancer properties [238]. Lipid–polymer hybrid nanoparticles, exemplified by quercetin–mycophenolic acid formulations (135–175 nm), enhance pharmacokinetics by diminishing first-pass metabolism and improving synergistic pharmacodynamics [239]. On the other hand, solid lipid nanoparticles containing EGCG with Bombesin (164 ± 2 nm) exhibit apoptotic actions by limiting nutrition access to cancer cells, thereby inhibiting migration and angiogenesis [240]. Moreover, metallic nanoparticles, such as gold nanoparticles functionalized with quercetin (5 nm) and AgFeO2 (19 nm), have produced anti-metastatic characteristics by obstructing epithelial–mesenchymal transition (EMT) and enhancing photodynamic treatment under ultraviolet light [241]. Hesperidin-encapsulated gold nanoparticles (40 nm) have also shown potential in regulating inflammatory cytokine release [243], while silver nanoparticles containing apigenin (94 nm) promote caspase-3-mediated apoptosis, thus increasing cytotoxicity against tumour cells [244]. Notably, utilising polymeric, lipidic, and metallic nanocarrier approaches can provide improved drug transport, tumour-targeted accumulation, and activation of apoptosis, presenting them as promising candidates for subsequent clinical application.
Contextually, phytosomes and liposomes are widely used in the market owing to their efficacy in medication delivery. Here, phytosomes, created by the electrostatic relationship between phospholipids and phytochemicals, have improved bioavailability of natural compounds. The Phytosome® technology, created by an Italian pharmaceutical firm, has shown efficacy in enhancing oral absorption of poorly absorbed compounds [245]. When compared to their unencapsulated counterparts, lipid-based nanoformulations containing flavonoids, including myricetin, EGCG, and quercetin, have stronger anti-cancer action. Moreover, when compared to non-conjugated compositions, EGCG-loaded nanoparticles conjugated with the gastrin-releasing peptide receptor (GRPR)-specific peptide bombesin demonstrated enhanced therapeutic potency and anti-migration ability in in vitro MDA-MB-231 and B16F10 cancer cell lines and in vivo female C57/BL6 mice. [240]. Intriguingly, metallic nanoparticles containing flavonoids, such as EGCG, quercetin, hesperidin, and apigenin, have also received interest owing to their improved therapeutic properties. For example, hesperidin-loaded gold nanoparticles (Hsp-AuNPs) were demonstrated to promote cytotoxicity in MDA-MB-231 cells in vitro, whereas toxicity studies in BALB/c mice indicated no obvious harm or histological alterations at dosages of 20 mg/kg and 200 mg/kg [243]. Additionally, new research on naringenin coupled with cyclophosphamide in breast cancer cells has emphasized their anti-proliferative properties, especially using the activation of STAT-3 and JAK-2 pathways. This combined therapy exhibits the capacity to prevent the uncontrolled proliferation of cancer cells, highlighting its potential as an effective therapeutic agent for breast cancer chemotherapy [246]. Additionally, nanoemulsions have been suggested as a viable delivery strategy to boost the absorption and solubility of naringenin [247]. Nano-naringenin significantly blocks both PI3K and MAPK pathways, and conserves ER-alpha in the cell membrane, therefore lowering proliferation in tamoxifen-resistant breast cancer cells (Tam-R) [248]. Furthermore, silibinin-loaded nanomaterials (SLNs) have exhibited targeted cancer development, revealing higher inhibitory properties on MDA-MB-231 cells than with pure silibinin, mainly caused by the inhibition of Snail and MMP [249,250,251]. Genistein, another flavonoid with breast cancer-preventive characteristics, has been demonstrated to stop MCF-7 cell development in the G2/M phase while lowering proliferation in the S phase [252]. A unique genistein-loaded PEGylated silica hybrid nanoparticle has also been produced utilizing a basic aqueous dispersion process, which indicated increased breast cancer therapy effectiveness, as validated by infrared examination exhibiting packed encapsulation of genistein [253,254]. The mechanisms of action and different nanotechnologies of flavonoids and their co-delivery in breast cancer therapy are explained extensively in Table 6.
The use of flavonoid-based nano-formulations in breast cancer treatment is an effective approach for enhancing drug solubility, stability, and targeted delivery. Liposome-based systems have been extensively used, with quercetin co-encapsulated with medicines including vincristine, doxorubicin, and Adriamycin exhibiting synergistic effects, decreased tumor progression, and controlled release [255,256,257]. PEGylated liposomes and polymeric nanoparticles, particularly those that integrate quercetin with tamoxifen, successfully attenuate drug resistance by regulating pathways including P-glycoprotein and Nrf2, resulting in increased oxidative stress and apoptosis [272].
Furthermore, nano-formulations containing silibinin, EGCG, curcumin, and chrysin exhibit several therapeutic actions. This includes inducing apoptosis, inhibiting angiogenesis, and suppressing tumor-promoting pathways like PI3K/AKT/mTOR [260,266,269,281]. Likewise, lipid-based systems, especially lipid nanoparticles and nano-emulsions, enhance flavonoid stability and cellular absorption while decreasing toxicity. Additionally, polymeric micelles and gold nanoparticles have favorable effects on cancer-specific targets, such as DNA–topoisomerase complexes and apoptotic genes [284,285]. Additionally, combinations such as baicalein–paclitaxel or rapamycin–curcumin exhibit substantial tumor suppression and prolonged drug release, highlighting the significance of dual-loaded systems [271,278]. Hence, all these research reports focus on important issues in the treatment of breast cancer by highlighting the critical role that flavonoid-based nano-carriers provide to achieve improved therapeutic efficacy, decreased multidrug resistance, and targeted cancer cell death.
Various methods of production are used for the development of flavonoid-based nano-formulations [286]. Nanoprecipitation is a prevalent method that depends on the fast combination of a polymer or lipid solution with a non-solvent, which results in the natural production of nanoparticles with a homogeneous size distribution [287]. Thin-film hydration is a widely used technique in which lipids are dissolved in organic solvents, evaporated to form a thin film and then hydrated with an aqueous phase to produce liposomes or analogous carriers [288,289]. Solvent evaporation requires emulsifying a drug–polymer combination in an aqueous phase and then removing the solvent under decreased pressure to produce stable nanoparticles [290]. Sonication-assisted methods are often used to decrease particle size and enhance uniformity. They are typically utilised as a subsequent step to refine liposomes or other vesicular systems [291,292]. Recent investigations into microfluidic techniques have focused on attaining rigorous control over particle sizes and consistency, where green synthesis techniques prioritise the use of biocompatible solvents and natural stabilisers to improve safety and sustainability [293]. Optimization of these nano-formulations is necessary to obtain optimal therapeutic effects. Key factors involve the lipid-to-drug or polymer-to-drug ratio, which affects encapsulation efficiency and drug release [294,295]. Surfactant concentration and type influence particle stability and prevent aggregation. Process parameters, including hydration temperature, stirring velocity, and sonication duration, are critical in regulating particle size and polydispersity index [296,297]. Systematic optimising of these factors promotes drug loading, stability, and bioavailability, thus enhancing the efficacy of flavonoid-based nano-formulations in breast cancer therapy [298,299].
7. Safety, Toxicity, and Regulatory Aspects of Flavonoid-Based Nanoformulations
Flavonoid-based nanoformulations provide a potential technique to boost the bioavailability and therapeutic effectiveness of flavonoids in cancer therapy. However, their inclusion into nanocarriers raises significant safety and toxicity problems that need thorough consideration [300,301]. While flavonoids themselves are typically harmless, encapsulation in nanoparticles may change their pharmacokinetics and interactions with biological systems [302]. Nanotechnology-based carriers like lipid-based nanoparticles and polymeric micelles may increase stability and allow controlled release, thus decreasing systemic toxicity. However, particle size, surface charge, and composition might alter cellular absorption and distribution, which can lead to harmful effects in non-target tissues [303,304].
Additionally, nanoparticles may accumulate in organs, notably the liver, spleen, and kidneys. This is a potential concern of organ-specific damage, such as hepatotoxicity and nephrotoxicity [305,306,307]. Oxidative stress and inflammation in various organs have been identified in select investigations, stressing the necessity for long-term in vivo study. Interaction with the immune system is another important factor. Nanomaterials can induce immunological activation and pro-inflammatory responses, which may result in hypersensitivity or chronic inflammation [308,309]. Surface modifications, such as PEGylation, may decrease immunogenicity, but their long-term safety is yet to be completely understood [310,311].
The regulatory framework for flavonoid-based nanoformulations is challenging and varied between areas, reflecting the particular issues faced by nanomedicines. The US Food and Drug Administration offers recommendations for drug products incorporating nanomaterials, highlighting the necessity for rigorous assessment of physicochemical attributes, pharmacokinetics, and possible toxicity. Risk assessment and mitigation measures are needed to assure the safety, quality, and effectiveness of these formulations [312,313]. Additionally, the European Medicines Agency (EMA) has produced recommendations that concentrate on the quality, safety, and effectiveness of nanomedicines. The EMA underlines the necessity of analysing nanomaterial characteristics, biodistribution, and pharmacokinetics, supporting preclinical and clinical investigation of immunogenicity and organ-specific toxicity [314,315,316]. At the worldwide level, organizations such as the International Council for Harmonisation are attempting to unify regulatory standards. Efforts are ongoing to standardize procedures for nanomaterial characterisation and safety evaluation, facilitating worldwide research, approval, and clinical translation of nanomedicines, including flavonoid-based formulations [317].
8. Conclusion and Future Perspectives: Advancing Flavonoid Research in Cancer Therapy
Flavonoids, with their various pharmacological effects, have been identified as potential medicines in the prevention and treatment of breast cancer. Their capacity to cause apoptosis, suppress cell growth, and affect several biological pathways makes them viable options in the production of innovative anti-cancer medicines. However, despite their enormous medicinal potential, the clinical application of flavonoids has been limited by difficulties such as limited bioavailability, poor solubility, and quick metabolism. The development of nanotechnology has created new opportunities for addressing these limitations, since nanoparticle-based drug delivery systems have exhibited increased stability, solubility, and specific therapeutic administration of flavonoids. While multiple scientific studies have shown the anti-cancer potential of flavonoid-based treatments, additional clinical studies are required to assess their safety, efficacy, and appropriate dose in individuals. The combined administration of flavonoids with conventional chemotherapeutic drugs also has considerable promise, but understanding the synergistic advantages and reducing possible toxicities remains a crucial topic for future studies.
Current studies on flavonoid-based breast cancer therapy should advance beyond single-agent trials to extensively investigate combination approaches with existing chemotherapeutics, hormonal drugs, and targeted therapies. These strategies may overcome drug resistance, improve therapeutic effectiveness, and reduce toxicity. However, careful optimization of dosage, sequencing, and delivery methods is required to attain clinically significant results. Nanotechnology will remain an essential facilitator in this domain. Development in co-delivery systems, stimuli-responsive nanocarriers, and tailored nanomedicine techniques may greatly improve the pharmacokinetic limitations of flavonoids and allow precise targeting of tumor sites. Simultaneous synthesis of novel flavonoid combinations with enhanced stability and bioavailability is expected to provide new opportunities for therapeutic applications.
However, effective clinical translation will necessitate a deeper attention to safety and toxicity. Long-term in vivo studies are required to address problems such as organ-specific accumulation, immunogenicity, and chronic toxicity. Employing sophisticated analytical methods for nanoparticle characterization and continuous evaluation in biological systems will be vital to bridge the gap between laboratory effectiveness and patient safety. Furthermore, the regulatory framework must adapt to scientific advancement. Collaborative efforts with authorities such as the FDA, EMA, and ICH are vital for creating harmonized criteria for nanomedicine approval, including consistent techniques for characterization, toxicity assessment, and pharmacokinetic evaluation. Only by using the convergence of pharmacological innovation, nanotechnology, toxicology, and regulatory science can flavonoid-based nanoformulations proceed from promising laboratory results to safe and effective clinical therapy for breast cancer.
Author Contributions
Methodology, P.U.; validation, P.U. and A.A.; visualization, R.R.; supervision, R.R.; software, P.U.; data curation, P.U.; writing the original draft, P.U.; writing—reviewing and editing, A.A. and R.R. All authors have read and agreed to the published version of the manuscript.
Funding
This review received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
The raw data supporting the conclusions of this article will be made available by the authors on request.
Acknowledgments
The authors are thankful to UPES University for providing all the necessary facilities to write this communication.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Global Cancer Burden Growing, Amidst Mounting Need for Services. Available online: https://www.who.int/news/item/01-02-2024-global-cancer-burden-growing--amidst-mounting-need-for-services (accessed on 22 October 2024).
- Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer Statistics, 2024. CA A Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef]
- Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy. In Nano-Enabled Medical Applications; Jenny Stanford Publishing: Singapore, 2020; ISBN 978-0-429-39903-9. [Google Scholar]
- Kumar, S.; Pandey, A.K. Chemistry and Biological Activities of Flavonoids: An Overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
- Liu, W.; Feng, Y.; Yu, S.; Fan, Z.; Li, X.; Li, J.; Yin, H. The Flavonoid Biosynthesis Network in Plants. Int. J. Mol. Sci. 2021, 22, 12824. [Google Scholar] [CrossRef]
- Jiang, W.; Hu, M. Mutual Interactions between Flavonoids and Enzymatic and Transporter Elements Responsible for Flavonoid Disposition via Phase II Metabolic Pathways. RSC Adv. 2012, 2, 7948–7963. [Google Scholar] [CrossRef]
- Park, E.-J.; Pezzuto, J.M. Flavonoids in Cancer Prevention. Available online: http://www.eurekaselect.com (accessed on 10 November 2024).
- Hazafa, A.; Rehman, K.-U.-; Jahan, N.; Jabeen, Z. The Role of Polyphenol (Flavonoids) Compounds in the Treatment of Cancer Cells. Nutr. Cancer 2020, 72, 386–397. [Google Scholar] [CrossRef] [PubMed]
- Hussain, Y.; Luqman, S.; Meena, A. Research Progress in Flavonoids as Potential Anticancer Drug Including Synergy with Other Approaches. Curr. Top. Med. Chem. 2020, 20, 1791–1809. [Google Scholar] [CrossRef] [PubMed]
- Acharya, S.; Sahoo, S.K. PLGA Nanoparticles Containing Various Anticancer Agents and Tumour Delivery by EPR Effect. Adv. Drug Deliv. Rev. 2011, 63, 170–183. [Google Scholar] [CrossRef]
- Davis, M.E.; Chen, Z.; Shin, D.M. Nanoparticle therapeutics: An emerging treatment modality for cancer. Nat. Rev. Drug Discov. 2008, 7, 771–782. [Google Scholar] [CrossRef] [PubMed]
- Bhatia, S. Nanoparticles Types, Classification, Characterization, Fabrication Methods and Drug Delivery Applications. In Natural Polymer Drug Delivery Systems: Nanoparticles, Plants, and Algae; Bhatia, S., Ed.; Springer International Publishing: Cham, Switzerland, 2016; pp. 33–93. ISBN 978-3-319-41129-3. [Google Scholar]
- Mohapatra, P.; Singh, P.; Singh, D.; Sahoo, S.; Sahoo, S.K. Phytochemical based nanomedicine: A panacea for cancer treatment, present status and future prospective. OpenNano 2022, 7, 100055. [Google Scholar] [CrossRef]
- Mohapatra, P.; Singh, P.; Sahoo, S.K. Phytonanomedicine: A novel avenue to treat recurrent cancer by targeting cancer stem cells. Drug Discov. Today 2020, 25, 1307–1321. [Google Scholar] [CrossRef]
- Sharma, T.; Singh, D.; Mahapatra, A.; Mohapatra, P.; Sahoo, S.; Sahoo, S.K. Advancements in clinical translation of flavonoid nanoparticles for cancer treatment. OpenNano 2022, 8, 100074. [Google Scholar] [CrossRef]
- Morsy, H.M.; Zaky, M.Y.; Yassin, N.Y.S.; Khalifa, A.Y.Z. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int. J. Pharm. 2025, 670, 125135. [Google Scholar] [CrossRef]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
- Pallauf, K.; Duckstein, N.; Rimbach, G. A literature review of flavonoids and lifespan in model organisms. Proc. Nutr. Soc. 2017, 76, 145–162. [Google Scholar] [CrossRef] [PubMed]
- Hegde, M.; Lakshman, K.; Lakshman, K.; Lakshman, K. Role of Polyphenols and Flavonoids as Anti-Cancer Drug Candidates: A Review. Pharmacogn. Res. 2023, 15, 206–216. [Google Scholar] [CrossRef]
- Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef] [PubMed]
- Jucá, M.M.; Cysne Filho, F.M.; de Almeida, J.C.; Mesquita, D.D.; Barriga, J.R.; Dias, K.C.; Barbosa, T.M.; Vasconcelos, L.C.; Leal, L.K.; Ribeiro, J.E.; et al. Flavonoids: Biological activities and therapeutic potential. Nat. Product Res. 2020, 34, 692–705. [Google Scholar] [CrossRef]
- Azad, A.K.; Dayoob, M.; Zohera, F.T. Anticancer Activity of Flavonoids: Past, Present, and Future. In Advances in Medical Diagnosis, Treatment, and Care; Roy, A., Ed.; IGI Global: Hershey, PA, USA, 2024; pp. 1–21. ISBN 9798369316467. [Google Scholar]
- Tovar-Pérez, E.G.; Aguilera-Aguirre, S.; López-García, U.; Valdez-Morales, M.; Ibarra-Zurita, A.K.; Ortiz-Basurto, R.I.; Chacón-López, A. Effect of ultrasound treatment on the quality and contents of polyphenols, lycopene and rutin in tomato fruits. Czech J. Food Sci. 2020, 38, 20–27. [Google Scholar] [CrossRef]
- Wang, R.-S.; Dong, P.-H.; Shuai, X.-X.; Chen, M.-S. Evaluation of Different Black Mulberry Fruits (Morus nigra L.) Based on Phenolic Compounds and Antioxidant Activity. Foods 2022, 11, 1252. [Google Scholar] [CrossRef]
- Lopes-Lutz, D.; Dettmann, J.; Nimalaratne, C.; Schieber, A. Characterization and Quantification of Polyphenols in Amazon Grape (Pourouma Cecropiifolia Martius). Molecules 2010, 15, 8543–8552. [Google Scholar] [CrossRef]
- Shafi, W.; Mansoor, S.; Jan, S.; Singh, D.B.; Kazi, M.; Raish, M.; Alwadei, M.; Mir, J.I.; Ahmad, P. Variability in Catechin and Rutin Contents and Their Antioxidant Potential in Diverse Apple Genotypes. Molecules 2019, 24, 943. [Google Scholar] [CrossRef] [PubMed]
- Batra, P.; Sharma, A.K. Anti-cancer potential of flavonoids: Recent trends and future perspectives. 3 Biotech 2013, 3, 439–459. [Google Scholar] [CrossRef] [PubMed]
- Patel, K.; Patel, D.K. Chapter 26—The Beneficial Role of Rutin, A Naturally Occurring Flavonoid in Health Promotion and Disease Prevention: A Systematic Review and Update. In Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases, 2nd ed.; Watson, R.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 457–479. ISBN 978-0-12-813820-5. [Google Scholar]
- Chaudhary, P.; Sharma, A.; Singh, B.; Nagpal, A.K. Bioactivities of phytochemicals present in tomato. J. Food Sci. Technol. 2018, 55, 2833–2849. [Google Scholar] [CrossRef] [PubMed]
- Martí, R.; Roselló, S.; Cebolla-Cornejo, J. Tomato as a Source of Carotenoids and Polyphenols Targeted to Cancer Prevention. Cancers 2016, 8, 58. [Google Scholar] [CrossRef]
- Mazzucato, A.; Willems, D.; Bernini, R.; Picarella, M.E.; Santangelo, E.; Ruiu, F.; Tilesi, F.; Soressi, G.P. Novel phenotypes related to the breeding of purple-fruited tomatoes and effect of peel extracts on human cancer cell proliferation. Plant Physiol. Biochem. 2013, 72, 125–133. [Google Scholar] [CrossRef]
- De Araújo, M.E.M.B.; Moreira Franco, Y.E.; Alberto, T.G.; Sobreiro, M.A.; Conrado, M.A.; Priolli, D.G.; Frankland Sawaya, A.C.H.; Ruiz, A.L.T.G.; de Carvalho, J.E.; de Oliveira Carvalho, P. Enzymatic de-glycosylation of rutin improves its antioxidant and antiproliferative activities. Food Chem. 2013, 141, 266–273. [Google Scholar] [CrossRef]
- You, H.J.; Ahn, H.J.; Ji, G.E. Transformation of Rutin to Antiproliferative Quercetin-3-glucoside by Aspergillus niger. J. Agric. Food Chem. 2010, 58, 10886–10892. [Google Scholar] [CrossRef]
- Yang, J.; Liu, R.H. Synergistic Effect of Apple Extracts and Quercetin 3-β-d-Glucoside Combination on Antiproliferative Activity in MCF-7 Human Breast Cancer Cells in Vitro. J. Agric. Food Chem. 2009, 57, 8581–8586. [Google Scholar] [CrossRef]
- Khatri, P.; Chen, L.; Rajcan, I.; Dhaubhadel, S. Functional characterization of Cinnamate 4-hydroxylase gene family in soybean (Glycine max). PLoS ONE 2023, 18, e0285698. [Google Scholar] [CrossRef]
- Davies, K.M. Genetic modification of plant metabolism for human health benefits. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2007, 622, 122–137. [Google Scholar] [CrossRef]
- Ai, J.; Bao, B.; Battino, M.; Giampieri, F.; Chen, C.; You, L.; Cespedes-Acuña, C.L.; Ognyanov, M.; Tian, L.; Bai, W. Recent advances on bioactive polysaccharides from mulberry. Food Funct. 2021, 12, 5219–5235. [Google Scholar] [CrossRef]
- Peng, C.-H.; Lin, H.-T.; Chung, D.-J.; Huang, C.-N.; Wang, C.-J. Mulberry Leaf Extracts prevent obesity-induced NAFLD with regulating adipocytokines, inflammation and oxidative stress. J. Food Drug Anal. 2018, 26, 778–787. [Google Scholar] [CrossRef]
- Demir, S.; Turan, I.; Aliyazicioglu, Y.; Kilinc, K.; Yaman, S.O.; Ayazoglu Demir, E.; Arslan, A.; Mentese, A.; Deger, O. Morus Rubra Extract Induces Cell Cycle Arrest and Apoptosis in Human Colon Cancer Cells Through Endoplasmic Reticulum Stress and Telomerase. Nutr. Cancer 2017, 69, 74–83. [Google Scholar] [CrossRef]
- Shang, A.; Luo, M.; Gan, R.-Y.; Xu, X.-Y.; Xia, Y.; Guo, H.; Liu, Y.; Li, H.-B. Effects of Microwave-Assisted Extraction Conditions on Antioxidant Capacity of Sweet Tea (Lithocarpus polystachyus Rehd.). Antioxidants 2020, 9, 678. [Google Scholar] [CrossRef]
- Wu, T.-Y.; Liang, J.; Ai, J.-Y.; Cui, J.-L.; Huang, W.-D.; You, Y.-L.; Zhan, J.-C. Mulberry Ethanol Extract and Rutin Protect Alcohol-Damaged GES-1 Cells by Inhibiting the MAPK Pathway. Molecules 2022, 27, 4266. [Google Scholar] [CrossRef] [PubMed]
- Wu, T.; Yin, J.; Zhang, G.; Long, H.; Zheng, X. Mulberry and cherry anthocyanin consumption prevents oxidative stress and inflammation in diet-induced obese mice. Mol. Nutr. Food Res. 2016, 60, 687–694. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.-K.; Zhang, X.-H.; Seo, J.-S. Suppression of oxidative stress by grape seed supplementation in rats. Nutr. Res. Pract. 2012, 6, 3–8. [Google Scholar] [CrossRef] [PubMed]
- Cetin, A.; Kaynar, L.; Koçyiğit, I.; Hacioğlu, S.K.; Saraymen, R.; Oztürk, A.; Orhan, O.; Sağdiç, O. The effect of grape seed extract on radiation-induced oxidative stress in the rat liver. Turk. J. Gastroenterol. 2008, 19, 92–98. [Google Scholar]
- Finkel, T.; Holbrook, N.J. Oxidants, oxidative stress and the biology of ageing. Nature 2000, 408, 239–247. [Google Scholar] [CrossRef]
- Habib, H.M.; El-Fakharany, E.M.; Kheadr, E.; Ibrahim, W.H. Grape seed proanthocyanidin extract inhibits DNA and protein damage and labile iron, enzyme, and cancer cell activities. Sci. Rep. 2022, 12, 12393. [Google Scholar] [CrossRef]
- Singh, C.K.; Siddiqui, I.A.; El-Abd, S.; Mukhtar, H.; Ahmad, N. Combination chemoprevention with grape antioxidants. Mol. Nutr. Food Res. 2016, 60, 1406–1415. [Google Scholar] [CrossRef] [PubMed]
- Alberti, A.; Zielinski, A.A.F.; Zardo, D.M.; Demiate, I.M.; Nogueira, A.; Mafra, L.I. Optimisation of the extraction of phenolic compounds from apples using response surface methodology. Food Chem. 2014, 149, 151–158. [Google Scholar] [CrossRef] [PubMed]
- Perussello, C.A.; Zhang, Z.; Marzocchella, A.; Tiwari, B.K. Valorization of Apple Pomace by Extraction of Valuable Compounds. Compr. Rev. Food Sci. Food Saf. 2017, 16, 776–796. [Google Scholar] [CrossRef] [PubMed]
- Nile, A.; Nile, S.H.; Shin, J.; Park, G.; Oh, J.-W. Quercetin-3-Glucoside Extracted from Apple Pomace Induces Cell Cycle Arrest and Apoptosis by Increasing Intracellular ROS Levels. Int. J. Mol. Sci. 2021, 22, 10749. [Google Scholar] [CrossRef]
- Pandey, J.; Bastola, T.; Tripathi, J.; Tripathi, M.; Rokaya, R.K.; Dhakal, B.; DC, R.; Bhandari, R.; Poudel, A. Estimation of Total Quercetin and Rutin Content in Malus domestica of Nepalese Origin by HPLC Method and Determination of Their Antioxidative Activity. J. Food Qual. 2020, 2020, 8853426. [Google Scholar] [CrossRef]
- Zhu, C.; Zhou, X.; Long, C.; Du, Y.; Li, J.; Yue, J.; Pan, S. Variations of Flavonoid Composition and Antioxidant Properties among Different Cultivars, Fruit Tissues and Developmental Stages of Citrus Fruits. Chem. Biodivers. 2020, 17, e1900690. [Google Scholar] [CrossRef]
- Addi, M.; Elbouzidi, A.; Abid, M.; Tungmunnithum, D.; Elamrani, A.; Hano, C. An Overview of Bioactive Flavonoids from Citrus Fruits. Appl. Sci. 2022, 12, 29. [Google Scholar] [CrossRef]
- Bento-Silva, A.; Koistinen, V.M.; Mena, P.; Bronze, M.R.; Hanhineva, K.; Sahlstrøm, S.; Kitrytė, V.; Moco, S.; Aura, A.-M. Factors affecting intake, metabolism and health benefits of phenolic acids: Do we understand individual variability? Eur. J. Nutr. 2020, 59, 1275–1293. [Google Scholar] [CrossRef]
- Yao, Q.; Lin, M.-T.; Zhu, Y.-D.; Xu, H.-L.; Zhao, Y.-Z. Recent Trends in Potential Therapeutic Applications of the Dietary Flavonoid Didymin. Molecules 2018, 23, 2547. [Google Scholar] [CrossRef]
- Moustafa, A.M.Y.; Ahmed, S.H.; Nabil, Z.I.; Hussein, A.A.; Omran, M.A. Extraction and phytochemical investigation of Calotropis procera: Effect of plant extracts on the activity of diverse muscles. Pharm. Biol. 2010, 48, 1080–1190. [Google Scholar] [CrossRef]
- Benavente-García, O.; Castillo, J. Update on Uses and Properties of Citrus Flavonoids: New Findings in Anticancer, Cardiovascular, and Anti-inflammatory Activity. J. Agric. Food Chem. 2008, 56, 6185–6205. [Google Scholar] [CrossRef] [PubMed]
- Safari, F.; Hassanpour, H.; Alijanpour, A. Evaluation of hackberry (Celtis australis L.) fruits as sources of bioactive compounds. Sci. Rep. 2023, 13, 12233. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Cao, H.; Huang, Q.; Xiao, J.; Teng, H. Absorption, metabolism and bioavailability of flavonoids: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 7730–7742. [Google Scholar] [CrossRef] [PubMed]
- Patil, V.M.; Masand, N. Chapter 12—Anticancer Potential of Flavonoids: Chemistry, Biological Activities, and Future Perspectives. In Studies in Natural Products Chemistry; Atta-ur-Rahman, Ed.; Elsevier: Amsterdam, The Netherlands, 2018; Volume 59, pp. 401–430. [Google Scholar]
- Stabrauskiene, J.; Kopustinskiene, D.M.; Lazauskas, R.; Bernatoniene, J. Naringin and Naringenin: Their Mechanisms of Action and the Potential Anticancer Activities. Biomedicines 2022, 10, 1686. [Google Scholar] [CrossRef]
- Yang, Y.; Trevethan, M.; Wang, S.; Zhao, L. Beneficial effects of citrus flavanones naringin and naringenin and their food sources on lipid metabolism: An update on bioavailability, pharmacokinetics, and mechanisms. J. Nutr. Biochem. 2022, 104, 108967. [Google Scholar] [CrossRef]
- Tanaka, S.; Shinoki, A.; Hara, H. Melibiose, a Nondigestible Disaccharide, Promotes Absorption of Quercetin Glycosides in Rat Small Intestine. J. Agric. Food Chem. 2016, 64, 9335–9341. [Google Scholar] [CrossRef]
- Khodzhaieva, R.S.; Gladkov, E.S.; Kyrychenko, A.; Roshal, A.D. Progress and Achievements in Glycosylation of Flavonoids. Front. Chem. 2021, 9, 637994. [Google Scholar] [CrossRef]
- McKay, D.L.; Chen, C.-Y.O.; Zampariello, C.A.; Blumberg, J.B. Flavonoids and phenolic acids from cranberry juice are bioavailable and bioactive in healthy older adults. Food Chem. 2015, 168, 233–240. [Google Scholar] [CrossRef]
- Deng, R.; Liu, Y.; Wu, X.; Zhao, N.; Deng, J.; Pan, T.; Cao, L.; Zhan, F.; Qiao, X. Probing the interaction of hesperidin showing antiproliferative activity in colorectal cancer cells and human hemoglobin. Int. J. Biol. Macromol. 2024, 281, 136078. [Google Scholar] [CrossRef]
- Qadir Nanakali, N.M.; Maleki Dana, P.; Sadoughi, F.; Asemi, Z.; Sharifi, M.; Asemi, R.; Yousefi, B. The role of dietary polyphenols in alternating DNA methylation in cancer. Crit. Rev. Food Sci. Nutr. 2023, 63, 12256–12269. [Google Scholar] [CrossRef]
- Yuan, D.; Guo, Y.; Pu, F.; Yang, C.; Xiao, X.; Du, H.; He, J.; Lu, S. Opportunities and challenges in enhancing the bioavailability and bioactivity of dietary flavonoids: A novel delivery system perspective. Food Chem. 2024, 430, 137115. [Google Scholar] [CrossRef]
- Lin, Y.; Wang, Y.; Ye, Z.; Gao, N.; Xu, X.; Weng, Q.; Xu, R.; Ye, L. Effects of Dietary Flavonoids on the Metabolism of Vortioxetine andits Potential Mechanism. CMC 2024, 31, 3624–3630. [Google Scholar] [CrossRef]
- Lian, X.; Shi, M.; Liang, Y.; Lin, Q.; Zhang, L. The Effects of Unconventional Feed Fermentation on Intestinal Oxidative Stress in Animals. Antioxidants 2024, 13, 305. [Google Scholar] [CrossRef] [PubMed]
- Aatif, M. Current Understanding of Polyphenols to Enhance Bioavailability for Better Therapies. Biomedicines 2023, 11, 2078. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Gu, K.; Zhou, F. Dietary Flavonoid Intake and Cancer Mortality: A Population-Based Cohort Study. Nutrients 2023, 15, 976. [Google Scholar] [CrossRef]
- Li, M.; Cai, Q.; Gao, Y.-T.; Franke, A.A.; Zhang, X.; Zhao, Y.; Wen, W.; Lan, Q.; Rothman, N.; Shyr, Y.; et al. Phytoestrogens and lung cancer risk: A nested case-control study in never-smoking Chinese women. Am. J. Clin. Nutr. 2022, 115, 643–651. [Google Scholar] [CrossRef]
- Liu, F.; Xu, J.; Wang, X.; Peng, Y.; Wang, P.; Si, C.; Gong, J.; Zhou, H.; Zhang, M.; Chen, L.; et al. Dietary flavonoid intake and risk of hormone-related cancers: A population-based prospective cohort study. Phytomedicine 2024, 133, 155950. [Google Scholar] [CrossRef] [PubMed]
- Tu, K.; Liu, K.; Wang, Y.; Jiang, Y.; Zhang, C. Association of Dietary Intake of Zinc and Selenium with Breast Cancer Risk: A Case-Control Study in Chinese Women. Nutrients 2023, 15, 3253. [Google Scholar] [CrossRef]
- Yang, J.; Shen, H.; Mi, M.; Qin, Y. Isoflavone Consumption and Risk of Breast Cancer: An Updated Systematic Review with Meta-Analysis of Observational Studies. Nutrients 2023, 15, 2402. [Google Scholar] [CrossRef]
- Song, S.; Cheun, J.-H.; Moon, H.-G.; Noh, D.-Y.; Jung, S.-Y.; Lee, E.S.; Kim, Z.; Youn, H.J.; Cho, J.; Yoo, Y.B.; et al. Dietary Isoflavone Intake and Breast Cancer Prognosis: A Prospective Analysis and Meta-Analysis. Nutr. Cancer 2024, 76, 42–54. [Google Scholar] [CrossRef]
- Naghshi, S.; Tutunchi, H.; Yousefi, M.; Naeini, F.; Mobarak, S.; Asadi, M.; Sadeghi, O. Soy isoflavone intake and risk of cardiovascular disease in adults: A systematic review and dose-response meta-analysis of prospective cohort studies. Crit. Rev. Food Sci. Nutr. 2024, 64, 6087–6101. [Google Scholar] [CrossRef]
- Boutas, I.; Kontogeorgi, A.; Dimitrakakis, C.; Kalantaridou, S.N. Soy Isoflavones and Breast Cancer Risk: A Meta-analysis. In Vivo 2022, 36, 556–562. [Google Scholar] [CrossRef]
- Feng, X.-L.; Ho, S.C.; Mo, X.-F.; Lin, F.-Y.; Zhang, N.-Q.; Luo, H.; Zhang, X.; Zhang, C.-X. Association between flavonoids, flavonoid subclasses intake and breast cancer risk: A case-control study in China. Eur. J. Cancer Prev. 2020, 29, 493. [Google Scholar] [CrossRef] [PubMed]
- Ho, S.C.; Yeo, W.; Goggins, W.; Kwok, C.; Cheng, A.; Chong, M.; Lee, R.; Cheung, K.L. Pre-diagnosis and early post-diagnosis dietary soy isoflavone intake and survival outcomes: A prospective cohort study of early stage breast cancer survivors. Cancer Treat. Res. Commun. 2021, 27, 100350. [Google Scholar] [CrossRef] [PubMed]
- Seth, R.; Kushwaha, S.; Luqman, S.; Meena, A. Flavonoids as Prospective Aromatase Inhibitors in Breast Cancer Prevention/Therapy. Curr. Mol. Pharmacol. 2021, 14, 1112–1124. [Google Scholar] [CrossRef] [PubMed]
- Chagas, M.D.S.S.; Behrens, M.D.; Moragas-Tellis, C.J.; Penedo, G.X.M.; Silva, A.R.; Gonçalves-de-Albuquerque, C.F. Flavonols and Flavones as Potential anti-Inflammatory, Antioxidant, and Antibacterial Compounds. Oxidative Med. Cell. Longev. 2022, 2022, 9966750. [Google Scholar] [CrossRef]
- Boniface, P.K.; Elizabeth, F.I. Flavones as a Privileged Scaffold in Drug Discovery: Current Developments. Curr. Org. Synth. 2019, 16, 968–1001. [Google Scholar] [CrossRef]
- Nakai, S.; Fujita, M.; Kamei, Y. Health Promotion Effects of Soy Isoflavones. J. Nutr. Sci. Vitaminol. 2020, 66, 502–507. [Google Scholar] [CrossRef]
- Fan, M.; Zhang, G.; Hu, X.; Xu, X.; Gong, D. Quercetin as a tyrosinase inhibitor: Inhibitory activity, conformational change and mechanism. Food Res. Int. 2017, 100, 226–233. [Google Scholar] [CrossRef]
- Wang, Y.; Tao, B.; Wan, Y.; Sun, Y.; Wang, L.; Sun, J.; Li, C. Drug delivery based pharmacological enhancement and current insights of quercetin with therapeutic potential against oral diseases. Biomed. Pharmacother. 2020, 128, 110372. [Google Scholar] [CrossRef]
- Lotfi, N.; Yousefi, Z.; Golabi, M.; Khalilian, P.; Ghezelbash, B.; Montazeri, M.; Shams, M.H.; Baghbadorani, P.Z.; Eskandari, N. The potential anti-cancer effects of quercetin on blood, prostate and lung cancers: An update. Front. Immunol. 2023, 14, 1077531. [Google Scholar] [CrossRef]
- Srinivasan, A.; Thangavel, C.; Liu, Y.; Shoyele, S.; Den, R.B.; Selvakumar, P.; Lakshmikuttyamma, A. Quercetin regulates β-catenin signaling and reduces the migration of triple negative breast cancer. Mol. Carcinog. 2016, 55, 743–756. [Google Scholar] [CrossRef]
- Mawalizadeh, F.; Mohammadzadeh, G.; Khedri, A.; Rashidi, M. Quercetin potentiates the chemosensitivity of MCF-7 breast cancer cells to 5-fluorouracil. Mol. Biol. Rep. 2021, 48, 7733–7742. [Google Scholar] [CrossRef]
- Widyananda, M.H.; Pratama, S.K.; Ansori, A.N.M.; Antonius, Y.; Kharisma, V.D.; Murtadlo, A.A.A.; Jakhmola, V.; Rebezov, M.; Khayrullin, M.; Derkho, M.; et al. Quercetin as an anticancer candidate for glioblastoma multiforme by targeting AKT1, MMP9, ABCB1, and VEGFA: An in silico study. Karbala Int. J. Mod. Sci. 2023, 9, 10. [Google Scholar] [CrossRef]
- Zhang, J.; Teng, F.; Wu, T.; Li, S.; Li, K. Quercetin inhibits chronic stress-mediated progression of triple-negative breast cancer by blocking β2-AR/ERK1/2 pathway. Biomed. Pharmacother. 2024, 177, 116985. [Google Scholar] [CrossRef]
- Chen, W.-J.; Tsai, J.-H.; Hsu, L.-S.; Lin, C.-L.; Hong, H.-M.; Pan, M.-H. Quercetin blocks the aggressive phenotype of triple-negative breast cancer by inhibiting IGF1/IGF1R-mediated EMT program. J. Food Drug Anal. 2021, 29, 98. [Google Scholar] [CrossRef] [PubMed]
- Ramezani, F.; Samadi, N.; Mostafavi-Pour, Z. Sequential Therapy of Breast Cancer Cell Lines with Vitamin C and Quercetin Improves the Efficacy of Chemotherapeutic Drugs. Nutr. Cancer 2017, 69, 881–891. [Google Scholar] [CrossRef] [PubMed]
- Xu, Z.; Zhao, D.; Zheng, X.; Huang, B.; Xia, X.; Pan, X. Quercetin exerts bidirectional regulation effects on the efficacy of tamoxifen in estrogen receptor-positive breast cancer therapy: An in vitro study. Environ. Toxicol. 2020, 35, 1179–1193. [Google Scholar] [CrossRef] [PubMed]
- Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
- Arampatzis, A.S.; Pampori, A.; Droutsa, E.; Laskari, M.; Karakostas, P.; Tsalikis, L.; Barmpalexis, P.; Dordas, C.; Assimopoulou, A.N. Occurrence of Luteolin in the Greek Flora, Isolation of Luteolin and Its Action for the Treatment of Periodontal Diseases. Molecules 2023, 28, 7720. [Google Scholar] [CrossRef]
- Zhang, L.; Liu, Q.; Huang, L.; Yang, F.; Liu, A.; Zhang, J. Combination of lapatinib and luteolin enhances the therapeutic efficacy of lapatinib on human breast cancer through the FOXO3a/NQO1 pathway. Biochem. Biophys. Res. Commun. 2020, 531, 364–371. [Google Scholar] [CrossRef]
- Sato, Y.; Sasaki, N.; Saito, M.; Endo, N.; Kugawa, F.; Ueno, A. Luteolin Attenuates Doxorubicin-Induced Cytotoxicity to MCF-7 Human Breast Cancer Cells. Biol. Pharm. Bull. 2015, 38, 703–709. [Google Scholar] [CrossRef]
- Qin, W.; Guo, J.; Gou, W.; Wu, S.; Guo, N.; Zhao, Y.; Hou, W. Molecular mechanisms of isoflavone puerarin against cardiovascular diseases: What we know and where we go. Chin. Herbal. Med. 2022, 14, 234–243. [Google Scholar] [CrossRef] [PubMed]
- Yan, J.; Honglei, Y.; Yun, W.; Sheng, D.; Yun, H.; Anhua, Z.; Na, F.; Min, L.; Dandan, S.; Jing, W.; et al. Puerarin ameliorates myocardial remodeling of spontaneously hypertensive rats through inhibiting TRPC6-CaN-NFATc3 pathway. Eur. J. Pharmacol. 2022, 933, 175254. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Zhou, Z.; Zhang, Z.; Zhao, C.; Li, J.; Jiang, J.; Huang, B.; Qin, Y. Puerarin inhibits EMT induced by oxaliplatin via targeting carbonic anhydrase XII. Front. Pharmacol. 2022, 13, 969422. [Google Scholar] [CrossRef] [PubMed]
- Imran, M.; Aslam Gondal, T.; Atif, M.; Shahbaz, M.; Batool Qaisarani, T.; Hanif Mughal, M.; Salehi, B.; Martorell, M.; Sharifi-Rad, J. Apigenin as an anticancer agent. Phytother. Res. 2020, 34, 1812–1828. [Google Scholar] [CrossRef]
- Dou, X.; Zhou, Z.; Ren, R.; Xu, M. Apigenin, flavonoid component isolated from Gentiana veitchiorum flower suppresses the oxidative stress through LDLR-LCAT signaling pathway. Biomed. Pharmacother. 2020, 128, 110298. [Google Scholar] [CrossRef]
- Maashi, M.S.; Al-Mualm, M.; Al-Awsi, G.R.L.; Opulencia, M.J.C.; Al-Gazally, M.E.; Abdullaev, B.; Abdelbasset, W.K.; Ansari, M.J.; Jalil, A.T.; Alsaikhan, F.; et al. Apigenin alleviates resistance to doxorubicin in breast cancer cells by acting on the JAK/STAT signaling pathway. Mol. Biol. Rep. 2022, 49, 8777–8784. [Google Scholar] [CrossRef]
- Wu, H.-T.; Lin, J.; Liu, Y.-E.; Chen, H.-F.; Hsu, K.-W.; Lin, S.-H.; Peng, K.-Y.; Lin, K.-J.; Hsieh, C.-C.; Chen, D.-R. Luteolin suppresses androgen receptor-positive triple-negative breast cancer cell proliferation and metastasis by epigenetic regulation of MMP9 expression via the AKT/mTOR signaling pathway. Phytomedicine 2021, 81, 153437. [Google Scholar] [CrossRef]
- Lin, P.-H.; Chiang, Y.-F.; Shieh, T.-M.; Chen, H.-Y.; Shih, C.-K.; Wang, T.-H.; Wang, K.-L.; Huang, T.-C.; Hong, Y.-H.; Li, S.-C.; et al. Dietary Compound Isoliquiritigenin, an Antioxidant from Licorice, Suppresses Triple-Negative Breast Tumor Growth via Apoptotic Death Program Activation in Cell and Xenograft Animal Models. Antioxidants 2020, 9, 228. [Google Scholar] [CrossRef]
- Zheng, H.; Li, Y.; Wang, Y.; Zhao, H.; Zhang, J.; Chai, H.; Tang, T.; Yue, J.; Guo, A.M.; Yang, J. Downregulation of COX-2 and CYP 4A signaling by isoliquiritigenin inhibits human breast cancer metastasis through preventing anoikis resistance, migration and invasion. Toxicol. Appl. Pharmacol. 2014, 280, 10–20. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-P.; Zhang, Z.-Y.; Li, Y.-P.; Li, D.; Huang, S.-L.; Gu, L.-Q.; Xu, J.; Huang, Z.-S. Syntheses and evaluation of novel isoliquiritigenin derivatives as potential dual inhibitors for amyloid-beta aggregation and 5-lipoxygenase. Eur. J. Med. Chem. 2013, 66, 22–31. [Google Scholar] [CrossRef] [PubMed]
- Bimonte, S.; Barbieri, A.; Palma, G.; Rea, D.; Luciano, A.; D’Aiuto, M.; Arra, C.; Izzo, F. Dissecting the Role of Curcumin in Tumour Growth and Angiogenesis in Mouse Model of Human Breast Cancer. BioMed Res. Int. 2015, 2015, 878134. [Google Scholar] [CrossRef] [PubMed]
- Guneydas, G.; Topcul, M.R. Antiproliferative Effects of Curcumin Different Types of Breast Cancer. Asian Pac. J. Cancer Prev. 2022, 23, 911–917. [Google Scholar] [CrossRef]
- Falah, R.R.; Talib, W.H.; Shbailat, S.J. Combination of metformin and curcumin targets breast cancer in mice by angiogenesis inhibition, immune system modulation and induction of p53 independent apoptosis. Ther. Adv. Med. Oncol. 2017, 9, 235–252. [Google Scholar] [CrossRef]
- Liu, Z.; Smart, J.D.; Pannala, A.S. Recent developments in formulation design for improving oral bioavailability of curcumin: A review. J. Drug Deliv. Sci. Technol. 2020, 60, 102082. [Google Scholar] [CrossRef]
- Tabanelli, R.; Brogi, S.; Calderone, V. Improving Curcumin Bioavailability: Current Strategies and Future Perspectives. Pharmaceutics 2021, 13, 1715. [Google Scholar] [CrossRef]
- KabaÅ‚a-Dzik, A.; Rzepecka-Stojko, A.; Kubina, R.; Iriti, M.; Wojtyczka, R.D.; Buszman, E.; Stojko, J. Flavonoids, bioactive components of propolis, exhibit cytotoxic activity and induce cell cycle arrest and apoptosis in human breast cancer cells MDA-MB-231 and MCF-7—A comparative study. Cell Mol. Biol. 2018, 64, 1–10. [Google Scholar] [CrossRef]
- Wang, H.; Tao, L.; Qi, K.; Zhang, H.; Feng, D.; Wei, W.; Kong, H.; Chen, T.; Lin, Q. Quercetin reverses tamoxifen resistance in breast cancer cells. J. BUON 2015, 20, 707–713. [Google Scholar]
- Zhao, X.; Wang, Q.; Yang, S.; Chen, C.; Li, X.; Liu, J.; Zou, Z.; Cai, D. Quercetin inhibits angiogenesis by targeting calcineurin in the xenograft model of human breast cancer. Eur. J. Pharmacol. 2016, 781, 60–68. [Google Scholar] [CrossRef]
- Lei, H.; Luo, J.; Tong, L.; Peng, L.; Qi, Y.; Jia, Z.; Wei, Q. Quercetin binds to calcineurin at a similar region to cyclosporin A and tacrolimus. Food Chem. 2011, 127, 1169–1174. [Google Scholar] [CrossRef]
- Magura, J.; Moodley, R.; Mackraj, I. The effect of hesperidin and luteolin isolated from Eriocephalus africanus on apoptosis, cell cycle and miRNA expression in MCF-7. J. Biomol. Struct. Dyn. 2022, 40, 1791–1800. [Google Scholar] [CrossRef] [PubMed]
- Cook, M.T.; Liang, Y.; Besch-Williford, C.; Goyette, S.; Mafuvadze, B.; Hyder, S.M. Luteolin inhibits progestin-dependent angiogenesis, stem cell-like characteristics, and growth of human breast cancer xenografts. SpringerPlus 2015, 4, 444. [Google Scholar] [CrossRef] [PubMed]
- Cook, M.T. Mechanism of metastasis suppression by luteolin in breast cancer. Breast Cancer Targets Ther. 2018, 10, 89–100. [Google Scholar] [CrossRef] [PubMed]
- Surichan, S.; Androutsopoulos, V.P.; Sifakis, S.; Koutala, E.; Tsatsakis, A.; Arroo, R.R.J.; Boarder, M.R. Bioactivation of the citrus flavonoid nobiletin by CYP1 enzymes in MCF7 breast adenocarcinoma cells. Food Chem. Toxicol. 2012, 50, 3320–3328. [Google Scholar] [CrossRef]
- Razak, N.A.; Abu, N.; Ho, W.Y.; Zamberi, N.R.; Tan, S.W.; Alitheen, N.B.; Long, K.; Yeap, S.K. Cytotoxicity of eupatorin in MCF-7 and MDA-MB-231 human breast cancer cells via cell cycle arrest, anti-angiogenesis and induction of apoptosis. Sci. Rep. 2019, 9, 1514. [Google Scholar] [CrossRef]
- Yoshimaru, T.; Komatsu, M.; Tashiro, E.; Imoto, M.; Osada, H.; Miyoshi, Y.; Honda, J.; Sasa, M.; Katagiri, T. Xanthohumol suppresses oestrogen-signalling in breast cancer through the inhibition of BIG3-PHB2 interactions. Sci. Rep. 2014, 4, 7355. [Google Scholar] [CrossRef]
- Tiwari, P.; Kumar, A.; Balakrishnan, S.; Kushwaha, H.S.; Mishra, K.P. Silibinin-Induced Apoptosis in MCF7 and T47D Human Breast Carcinoma Cells Involves Caspase-8 Activation and Mitochondrial Pathway. Cancer Investig. 2011, 29, 12–20. [Google Scholar] [CrossRef]
- Liu, W.; Ji, Y.; Sun, Y.; Si, L.; Fu, J.; Hayashi, T.; Onodera, S.; Ikejima, T. Estrogen receptors participate in silibinin-caused nuclear translocation of apoptosis-inducing factor in human breast cancer MCF-7 cells. Arch. Biochem. Biophys. 2020, 689, 108458. [Google Scholar] [CrossRef]
- Yi, X.; Zuo, J.; Tan, C.; Xian, S.; Luo, C.; Chen, S.; Yu, L.; Luo, Y. Kaempferol, a flavonoid compound from gynura medica induced apoptosis and growth inhibition in MCF-7 breast cancer cell. Afr. J. Tradit. Complement. Altern. Med. 2016, 13, 210–215. [Google Scholar] [CrossRef]
- Kim, S.-H.; Hwang, K.-A.; Choi, K.-C. Treatment with kaempferol suppresses breast cancer cell growth caused by estrogen and triclosan in cellular and xenograft breast cancer models. J. Nutr. Biochem. 2016, 28, 70–82. [Google Scholar] [CrossRef]
- Bortolotto, L.F.B.; Barbosa, F.R.; Silva, G.; Bitencourt, T.A.; Beleboni, R.O.; Baek, S.J.; Marins, M.; Fachin, A.L. Cytotoxicity of trans-chalcone and licochalcone A against breast cancer cells is due to apoptosis induction and cell cycle arrest. Biomed. Pharmacother. 2017, 85, 425–433. [Google Scholar] [CrossRef]
- Pan, D.; Li, W.; Miao, H.; Yao, J.; Li, Z.; Wei, L.; Zhao, L.; Guo, Q. LW-214, a newly synthesized flavonoid, induces intrinsic apoptosis pathway by down-regulating Trx-1 in MCF-7 human breast cells. Biochem. Pharmacol. 2014, 87, 598–610. [Google Scholar] [CrossRef]
- Loaiza-Pérez, A.I.; Kenney, S.; Boswell, J.; Hollingshead, M.; Alley, M.C.; Hose, C.; Ciolino, H.P.; Yeh, G.C.; Trepel, J.B.; Vistica, D.T.; et al. Aryl hydrocarbon receptor activation of an antitumor aminoflavone: Basis of selective toxicity for MCF-7 breast tumor cells. Mol. Cancer Ther. 2004, 3, 715–725. [Google Scholar] [CrossRef]
- Cárdenas, M.; Marder, M.; Blank, V.C.; Roguin, L.P. Antitumor activity of some natural flavonoids and synthetic derivatives on various human and murine cancer cell lines. Bioorganic Med. Chem. 2006, 14, 2966–2971. [Google Scholar] [CrossRef] [PubMed]
- Anaya-Eugenio, G.; Carcache, P.; Ninh, T.; Ren, Y.; Soejarto, D.; Kinghorn, A. A pentamethoxylated flavone from Glycosmis ovoidea promotes apoptosis through the intrinsic pathway and inhibits migration of MCF-7 breast cancer cells. Phytother. Res. 2020, 35, 1634–1645. [Google Scholar] [CrossRef]
- Liu, X.; Zhao, W.; Wang, W.; Lin, S.; Yang, L. Puerarin suppresses LPS-induced breast cancer cell migration, invasion and adhesion by blockage NF-κB and Erk pathway. Biomed. Pharmacother. 2017, 92, 429–436. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wang, Y.; Feng, C.; Wu, G.; Ye, Y.; Tian, J. Calycosin Inhibits the Migration and Invasion of Human Breast Cancer Cells by Down-Regulation of Foxp3 Expression. Cell. Physiol. Biochem. 2017, 44, 1775–1784. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.-J.; Pham, T.-H.; Bak, Y.; Ryu, H.-W.; Oh, S.-R.; Yoon, D.-Y. Orientin inhibits invasion by suppressing MMP-9 and IL-8 expression via the PKCα/ERK/AP-1/STAT3-mediated signaling pathways in TPA-treated MCF-7 breast cancer cells. Phytomedicine 2018, 50, 35–42. [Google Scholar] [CrossRef]
- Liu, S.; Wang, L.; Zhang, R. Corylin suppresses metastasis of breast cancer cells by modulating miR-34c/LINC00963 target. Libyan J. Med. 2021, 16, 1883224. [Google Scholar] [CrossRef]
- Kalva, S.; Azhagiya Singam, E.R.; Rajapandian, V.; Saleena, L.M.; Subramanian, V. Discovery of potent inhibitor for matrix metalloproteinase-9 by pharmacophore based modeling and dynamics simulation studies. J. Mol. Graph. Model. 2014, 49, 25–37. [Google Scholar] [CrossRef]
- Chen, J.; Chang, H.; Peng, X.; Gu, Y.; Yi, L.; Zhang, Q.-Y.; Zhu, J.; Mi, M. 3,6-dihydroxyflavone suppresses the epithelial-mesenchymal transition in breast cancer cells by inhibiting the Notch signaling pathway. Sci. Rep. 2016, 6, 28858. [Google Scholar] [CrossRef]
- Li, C.; Li, J.; Gong, S.; Huang, M.; Li, R.; Xiong, G.; Wang, F.; Zou, Q.; Qi, Q.; Yin, X. Targeting the ILK/YAP axis by LFG-500 blocks epithelial–mesenchymal transition and metastasis. Acta Pharmacol. Sin. 2021, 42, 1847–1859. [Google Scholar] [CrossRef]
- Kim, H.; Lee, J. Hispidulin modulates epithelial-mesenchymal transition in breast cancer cells. Oncol. Lett. 2020, 21, 155. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Lin, M.; Wang, J.; Yang, F.; Yang, P.; Liu, Y.; Chen, Z.; Zheng, Y. Calycosin Inhibits Breast Cancer Cell Migration and Invasion by suppressing EMT via BATF/TGF-β1. Aging 2021, 13, 16009. [Google Scholar] [CrossRef] [PubMed]
- Singhal, J.; Nagaprashantha, L.; Chikara, S.; Awasthi, S.; Horne, D.; Singhal, S. 2’-Hydroxyflavanone: A novel strategy for targeting breast cancer. Oncotarget 2017, 8, 75025–75037. [Google Scholar] [CrossRef]
- Singhal, J.; Chikara, S.; Horne, D.; Salgia, R.; Awasthi, S.; Singhal, S.S. 2′-Hydroxyflavanone inhibits in vitro and in vivo growth of breast cancer cells by targeting RLIP76. Mol. Carcinog. 2018, 57, 1751–1762. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Zhao, K.; Hu, Y.; Zhou, Y.; Luo, X.; Li, X.; Wei, L.; Li, Z.; You, Q.; Guo, Q.; et al. Wogonoside inhibits angiogenesis in breast cancer via suppressing Wnt/β-catenin pathway. Mol. Carcinog. 2016, 55, 1598–1612. [Google Scholar] [CrossRef]
- Elhady, S.S.; Eltamany, E.E.; Shaaban, A.E.; Bagalagel, A.A.; Muhammad, Y.A.; El-Sayed, N.M.; Ayyad, S.N.; Ahmed, A.A.M.; Elgawish, M.S.; Ahmed, S.A. Jaceidin Flavonoid Isolated from Chiliadenus montanus Attenuates Tumor Progression in Mice via VEGF Inhibition: In Vivo and In Silico Studies. Plants 2020, 9, 1031. [Google Scholar] [CrossRef]
- Wang, R.; Deng, Z.; Zhu, Z.; Wang, J.; Yang, X.; Xu, M.; Wang, X.; Tang, Q.; Zhou, Q.; Wan, X.; et al. Kaempferol promotes non-small cell lung cancer cell autophagy via restricting Met pathway. Phytomedicine 2023, 121, 155090. [Google Scholar] [CrossRef]
- Farghadani, R.; Naidu, R. The anticancer mechanism of action of selected polyphenols in triple-negative breast cancer (TNBC). Biomed. Pharmacother. 2023, 165, 115170. [Google Scholar] [CrossRef]
- Zabaleta, M.E.; Forbes-Hernández, T.Y.; Simal-Gandara, J.; Quiles, J.L.; Cianciosi, D.; Bullon, B.; Giampieri, F.; Battino, M. Effect of polyphenols on HER2-positive breast cancer and related miRNAs: Epigenomic regulation. Food Res. Int. 2020, 137, 109623. [Google Scholar] [CrossRef]
- Gu, H.-F.; Mao, X.-Y.; Du, M. Prevention of breast cancer by dietary polyphenols—Role of cancer stem cells. Crit. Rev. Food Sci. Nutr. 2020, 60, 810–825. [Google Scholar] [CrossRef] [PubMed]
- Bender, O.; Atalay, A. Chapter 28—Polyphenol Chlorogenic Acid, Antioxidant Profile, and Breast Cancer. In Cancer, 2nd ed.; Preedy, V.R., Patel, V.B., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 311–321. ISBN 978-0-12-819547-5. [Google Scholar]
- Wei, R.; Mao, L.; Xu, P.; Zheng, X.; Hackman, R.M.; Mackenzie, G.G.; Wang, Y. Suppressing glucose metabolism with epigallocatechin-3-gallate (EGCG) reduces breast cancer cell growth in preclinical models. Food Funct. 2018, 9, 5682–5696. [Google Scholar] [CrossRef] [PubMed]
- Zan, L.; Chen, Q.; Zhang, L.; Li, X. Epigallocatechin gallate (EGCG) suppresses growth and tumorigenicity in breast cancer cells by downregulation of miR-25. Bioengineered 2019, 10, 374–382. [Google Scholar] [CrossRef] [PubMed]
- Sharifi-Rad, M.; Pezzani, R.; Redaelli, M.; Zorzan, M.; Imran, M.; Ahmed Khalil, A.; Salehi, B.; Sharopov, F.; Cho, W.C.; Sharifi-Rad, J. Preclinical Activities of Epigallocatechin Gallate in Signaling Pathways in Cancer. Molecules 2020, 25, 467. [Google Scholar] [CrossRef]
- Huang, Y.-J.; Wang, K.-L.; Chen, H.-Y.; Chiang, Y.-F.; Hsia, S.-M. Protective Effects of Epigallocatechin Gallate (EGCG) on Endometrial, Breast, and Ovarian Cancers. Biomolecules 2020, 10, 1481. [Google Scholar] [CrossRef]
- Háková, M.; Havlíková, L.C.; Švec, F.; Solich, P.; Erben, J.; Chvojka, J.; Šatínský, D. Novel nanofibrous sorbents for the extraction and determination of resveratrol in wine. Talanta 2020, 206, 120181. [Google Scholar] [CrossRef]
- Khatoon, E.; Banik, K.; Harsha, C.; Sailo, B.L.; Thakur, K.K.; Khwairakpam, A.D.; Vikkurthi, R.; Devi, T.B.; Gupta, S.C.; Kunnumakkara, A.B. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin. Cancer Biol. 2022, 80, 306–339. [Google Scholar] [CrossRef]
- Venkatadri, R.; Muni, T.; Iyer, A.K.V.; Yakisich, J.S.; Azad, N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell Death Dis. 2016, 7, e2104. [Google Scholar] [CrossRef]
- Li, Y.; Liu, J.; Liu, X.; Xing, K.; Wang, Y.; Li, F.; Yao, L. Resveratrol-induced cell inhibition of growth and apoptosis in MCF7 human breast cancer cells are associated with modulation of phosphorylated akt and caspase-9. Appl. Biochem. Biotechnol. 2006, 135, 181–192. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Chen, L.; Zhu, F.; Han, X.; Sun, L.; Chen, K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins 2019, 11, 731. [Google Scholar] [CrossRef] [PubMed]
- Safdar, M.A.; Aslam, R.M.N.; Shakeel, A.; Shiza; Waqar, M.; Jmail, A.; Mehmood, M.H.; Gul, H. Cyanidin as potential anticancer agent targeting various proliferative pathways. Chem. Biol. Drug Des. 2023, 101, 438–452. [Google Scholar] [CrossRef] [PubMed]
- Giampieri, F.; Gasparrini, M.; Forbes-Hernandez, T.Y.; Mazzoni, L.; Capocasa, F.; Sabbadini, S.; Alvarez-Suarez, J.M.; Afrin, S.; Rosati, C.; Pandolfini, T.; et al. Overexpression of the Anthocyanidin Synthase Gene in Strawberry Enhances Antioxidant Capacity and Cytotoxic Effects on Human Hepatic Cancer Cells. J. Agric. Food Chem. 2018, 66, 581–592. [Google Scholar] [CrossRef]
- Li, W.; Peng, C.; Zhaojie, L.; Wei, W. Chemopreventive and therapeutic properties of anthocyanins in breast cancer: A comprehensive review. Nutr. Res. 2022, 107, 48–64. [Google Scholar] [CrossRef]
- Paramanantham, A.; Kim, M.J.; Jung, E.J.; Kim, H.J.; Chang, S.-H.; Jung, J.-M.; Hong, S.C.; Shin, S.C.; Kim, G.S.; Lee, W.S. Anthocyanins Isolated from Vitis coignetiae Pulliat Enhances Cisplatin Sensitivity in MCF-7 Human Breast Cancer Cells through Inhibition of Akt and NF-κB Activation. Molecules 2020, 25, 3623. [Google Scholar] [CrossRef]
- Li, Z.-L.; Mi, J.; Lu, L.; Luo, Q.; Liu, X.; Yan, Y.-M.; Jin, B.; Cao, Y.-L.; Zeng, X.-X.; Ran, L.-W. The main anthocyanin monomer of Lycium ruthenicum Murray induces apoptosis through the ROS/PTEN/PI3K/Akt/caspase 3 signaling pathway in prostate cancer DU-145 cells. Food Funct. 2021, 12, 1818–1828. [Google Scholar] [CrossRef]
- Rabelo, A.C.S.; Guerreiro, C.D.A.; Shinzato, V.I.; Ong, T.P.; Noratto, G. Anthocyanins Reduce Cell Invasion and Migration through Akt/mTOR Downregulation and Apoptosis Activation in Triple-Negative Breast Cancer Cells: A Systematic Review and Meta-Analysis. Cancers 2023, 15, 2300. [Google Scholar] [CrossRef]
- Zhu, L.; Xue, L. Kaempferol Suppresses Proliferation and Induces Cell Cycle Arrest, Apoptosis, and DNA Damage in Breast Cancer Cells. Oncol. Res. 2019, 27, 629–634. [Google Scholar] [CrossRef]
- Xiao, X.; Hu, Q.; Deng, X.; Shi, K.; Zhang, W.; Jiang, Y.; Ma, X.; Zeng, J.; Wang, X. Old wine in new bottles: Kaempferol is a promising agent for treating the trilogy of liver diseases. Pharmacol. Res. 2022, 175, 106005. [Google Scholar] [CrossRef]
- He, W.; Zhang, J.; Ju, J.; Wu, Y.; Zhang, Y.; Zhan, L.; Li, C.; Wang, Y. Preparation, characterization, and evaluation of the antitumor effect of kaempferol nanosuspensions. Drug Deliv. Transl. Res. 2023, 13, 2885–2902. [Google Scholar] [CrossRef]
- Wang, X.; Yang, Y.; An, Y.; Fang, G. The mechanism of anticancer action and potential clinical use of kaempferol in the treatment of breast cancer. Biomed. Pharmacother. 2019, 117, 109086. [Google Scholar] [CrossRef] [PubMed]
- Modi, S.; Currie, V.E.; Seidman, A.D.; Bach, A.M.; Panageas, K.S.; Theodoulou, M.; Moasser, M.M.; D’Andrea, G.M.; Lake, D.E.; Choi, J.; et al. A phase II trial of gemcitabine in patients with metastatic breast cancer previously treated with an anthracycline and taxane. Clin. Breast Cancer 2005, 6, 55–60. [Google Scholar] [CrossRef] [PubMed]
- Bundred, N. Preclinical and clinical experience with fulvestrant (Faslodex) in postmenopausal women with hormone receptor-positive advanced breast cancer. Cancer Investig. 2005, 23, 173–181. [Google Scholar] [CrossRef] [PubMed]
- Quintela-Fandino, M.; Morales, S.; Cortés-Salgado, A.; Manso, L.; Apala, J.V.; Muñoz, M.; Gasol Cudos, A.; Salla Fortuny, J.; Gion, M.; Lopez-Alonso, A.; et al. Randomized Phase 0/I Trial of the Mitochondrial Inhibitor ME-344 or Placebo Added to Bevacizumab in Early HER2-Negative Breast Cancer. Clin. Cancer Res. 2020, 26, 35–45. [Google Scholar] [CrossRef]
- Ravasco, P. Does Watercress Intake Have an Impact on Cancer Patients Outcomes: A Randomized Longitudinal Trial. 2015. Available online: https://clinicaltrials.gov/study/NCT02468882 (accessed on 28 September 2025).
- Lathrop, K.; Kaklamani, V.; Brenner, A.; Li, R.; Nazarullah, A.; Hackman, S.; Thomas, C.; Rodriguez, M.; Elledge, R. Novel estrogen receptor beta agonist S-equol decreases tumor proliferation in patients with triple negative breast cancer (TNBC). J. Clin. Oncol. 2020, 38, 560. [Google Scholar] [CrossRef]
- Vladu, A.F.; Ficai, D.; Ene, A.G.; Ficai, A. Combination Therapy Using Polyphenols: An Efficient Way to Improve Antitumoral Activity and Reduce Resistance. Int. J. Mol. Sci. 2022, 23, 10244. [Google Scholar] [CrossRef]
- Patra, S.; Pradhan, B.; Nayak, R.; Behera, C.; Das, S.; Patra, S.K.; Efferth, T.; Jena, M.; Bhutia, S.K. Dietary polyphenols in chemoprevention and synergistic effect in cancer: Clinical evidences and molecular mechanisms of action. Phytomedicine 2021, 90, 153554. [Google Scholar] [CrossRef]
- Jakobušić Brala, C.; Karković Marković, A.; Kugić, A.; Torić, J.; Barbarić, M. Combination Chemotherapy with Selected Polyphenols in Preclinical and Clinical Studies—An Update Overview. Molecules 2023, 28, 3746. [Google Scholar] [CrossRef]
- Lotha, R.; Sivasubramanian, A. Flavonoids nutraceuticals in prevention and treatment of cancer: A review. Asian J. Pharm. Clin. Res. 2018, 11, 42–47. [Google Scholar] [CrossRef]
- Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [PubMed]
- Koolaji, N.; Shammugasamy, B.; Schindeler, A.; Dong, Q.; Dehghani, F.; Valtchev, P. Citrus Peel Flavonoids as Potential Cancer Prevention Agents. Curr. Dev. Nutr. 2020, 4, nzaa025. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.U.; Dagur, H.S.; Khan, M.; Malik, N.; Alam, M.; Mushtaque, M. Therapeutic role of flavonoids and flavones in cancer prevention: Current trends and future perspectives. Eur. J. Med. Chem. Rep. 2021, 3, 100010. [Google Scholar] [CrossRef]
- Rejhová, A.; Opattová, A.; Čumová, A.; Slíva, D.; Vodička, P. Natural compounds and combination therapy in colorectal cancer treatment. Eur. J. Med. Chem. 2018, 144, 582–594. [Google Scholar] [CrossRef] [PubMed]
- Cheng, Z.; Zhang, Z.; Han, Y.; Wang, J.; Wang, Y.; Chen, X.; Shao, Y.; Cheng, Y.; Zhou, W.; Lu, X.; et al. A review on anti-cancer effect of green tea catechins. J. Funct. Foods 2020, 74, 104172. [Google Scholar] [CrossRef]
- Li, X.-X.; Liu, C.; Dong, S.-L.; Ou, C.-S.; Lu, J.-L.; Ye, J.-H.; Liang, Y.-R.; Zheng, X.-Q. Anticarcinogenic potentials of tea catechins. Front. Nutr. 2022, 9, 1060783. [Google Scholar] [CrossRef]
- Chou, T.-C. Drug Combination Studies and Their Synergy Quantification Using the Chou-Talalay Method. Cancer Res. 2010, 70, 440–446. [Google Scholar] [CrossRef]
- Wu, H.-T.; Liu, Y.-E.; Hsu, K.-W.; Wang, Y.-F.; Chan, Y.-C.; Chen, Y.; Chen, D.-R. MLL3 Induced by Luteolin Causes Apoptosis in Tamoxifen-Resistant Breast Cancer Cells through H3K4 Monomethylation and Suppression of the PI3K/AKT/mTOR Pathway. Am. J. Chin. Med. 2020, 48, 1221–1241. [Google Scholar] [CrossRef]
- Safi, A.; Heidarian, E.; Ahmadi, R. Quercetin Synergistically Enhances the Anticancer Efficacy of Docetaxel through Induction of Apoptosis and Modulation of PI3K/AKT, MAPK/ERK, and JAK/STAT3 Signaling Pathways in MDA-MB-231 Breast Cancer Cell Line. Int. J. Mol. Cell Med. 2021, 10, 11. [Google Scholar] [CrossRef]
- Roshanazadeh, M.; Rezaei, H.B.; Rashidi, M. Quercetin synergistically potentiates the anti-metastatic effect of 5-fluorouracil on the MDA-MB-231 breast cancer cell line. Iran. J. Basic Med. Sci. 2021, 24, 928. [Google Scholar] [CrossRef]
- Xing, Y.; Shen, W.; Sun, C.; Wang, R.; Fan, B.; Liang, G. Genistein-Chitosan Derivative Nanoparticles for Targeting and Enhancing the Anti-Breast Cancer Effect of Tamoxifen In Vitro. J. Pharm. Sci. 2024, 113, 2575–2583. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.; Yu, X.; Min, J.; Chen, X.; Liu, R.; Cui, X.; Cheng, J.; Xie, M.; Diel, P.; Hu, X. Genistein interferes with antitumor effects of cisplatin in an ovariectomized breast cancer xenograft tumor model. Toxicol. Lett. 2022, 355, 106–115. [Google Scholar] [CrossRef] [PubMed]
- Dian, C.; Qian, Z.; Ran, M.; Yan, X.; Dian, L. Co-Delivery of Docetaxel and Curcumin Functionalized Mixed Micelles for the Treatment of Drug-Resistant Breast Cancer by Oral Administration. Int. J. Nanomed. 2024, 19, 8603–8620. [Google Scholar] [CrossRef] [PubMed]
- Effat, H.; El Houseini, M.E.; Abohashem, R.S. The Combined Impact of Curcumin: Piperine and Sorafenib on microRNAs and Different Pathways in Breast Cancer Cells. Ind. J. Clin. Biochem. 2024, 40, 32–45. [Google Scholar] [CrossRef]
- Sarkar, E.; Kotiya, A.; Khan, A.; Bhuyan, R.; Raza, S.T.; Misra, A.; Mahdi, A.A. The combination of Curcumin and Doxorubicin on targeting PI3K/AKT/mTOR signaling pathway: An in vitro and molecular docking study for inhibiting the survival of MDA-MB-231. Silico Pharmacol. 2024, 12, 58. [Google Scholar] [CrossRef]
- Sarkar, E.; Khan, A.; Ahmad, R.; Misra, A.; Dua, K.; Mahdi, A.A.; Raza, T. Synergism of Curcumin and Doxorubicin Proves an Effective Anticancer Therapeutics Against Breast Cancer: An in vitro Study. Res. Sq. 2024, 16, e75047. [Google Scholar]
- Deswal, B.; Bagchi, U.; Kapoor, S. Curcumin Suppresses M2 Macrophage-derived Paclitaxel Chemoresistance throughInhibition of PI3K-AKT/STAT3 Signaling. Anti-Cancer Agents Med. Chem. Anti-Cancer Agents 2024, 24, 146–156. [Google Scholar] [CrossRef]
- Wang, G.; Duan, P.; Wei, Z.; Liu, F. Curcumin sensitizes carboplatin treatment in triple negative breast cancer through reactive oxygen species induced DNA repair pathway. Mol. Biol. Rep. 2022, 49, 3259–3270. [Google Scholar] [CrossRef]
- Noori, S.; Rezaei Tavirani, M.; Deravi, N.; Mahboobi Rabbani, M.I.; Zarghi, A. Naringenin Enhances the Anti-Cancer Effect of Cyclophosphamide against MDA-MB-231 Breast Cancer Cells Via Targeting the STAT3 Signaling Pathway. Iran. J. Pharm. Res. IJPR 2020, 19, 122. [Google Scholar] [CrossRef]
- Pateliya, B.; Burade, V.; Goswami, S. Combining naringenin and metformin with doxorubicin enhances anticancer activity against triple-negative breast cancer in vitro and in vivo. Eur. J. Pharmacol. 2021, 891, 173725. [Google Scholar] [CrossRef]
- Alshamrani, S.; Kumar, A.; Aldughaim, M.S.; Alghamdi, K.M.; Hussain, M.D.; Alanazi, F.K.; Kazi, M. Development of Polymeric Micelles for Combined Delivery of Luteolin and Doxorubicin for Cancer Therapy. J. Cancer 2024, 15, 4717–4730. [Google Scholar] [CrossRef]
- Tamanna, S.; Perumal, E.; Rajanathadurai, J. Enhanced Apoptotic Effects in MDA-MB-231 Triple-Negative Breast Cancer Cells Through a Synergistic Action of Luteolin and Paclitaxel. Cureus 2024, 16, e65159. [Google Scholar] [CrossRef] [PubMed]
- Çilesiz, Y.; Çevik, Ö. Anticancer effect of the letrozole-quercetin combination mediated by FOXOs and estrogen receptors in breast cancer cells. J. Res. Pharm. 2021, 25, 479–489. [Google Scholar] [CrossRef]
- Almohammad Aljabr, B.; Zihlif, M.; Abu-Dahab, R.; Zalloum, H. Effect of quercetin on doxorubicin cytotoxicity in sensitive and resistant human MCF7 breast cancer cell lines. Biomed. Rep. 2024, 20, 58. [Google Scholar] [CrossRef] [PubMed]
- Roshanazadeh, M.; Babaahmadi Rezaei, H.; Rashidi, M. Quercetin Enhances the Suppressive Effects of Doxorubicin on the Migration of MDA-MB-231 Breast Cancer Cell Line. Int. J. Cancer Manag. 2021, 14, e119049. [Google Scholar] [CrossRef]
- Sharma, S.; Gupta, P.; Kawish, S.M.; Ahmad, S.; Iqbal, Z.; Vohora, D.; Kohli, K. Novel Chitosan-Coated Liposomes Coloaded with Exemestane and Genistein for an Effective Breast Cancer Therapy. ACS Omega 2024, 9, 9735–9752. [Google Scholar] [CrossRef]
- Synergistic Anti-Cancer Effects of ERB-041 and Genistein Through Estrogen Receptor Suppression-Mediated PI3K/AKT Pathway Downregulation in Canine Mammary Gland Tumor Cells. Available online: https://www.mdpi.com/1422-0067/25/5/2466 (accessed on 1 December 2024).
- Kaushik, S.; Shyam, H.; Sharma, R.; Balapure, A. Genistein synergizes centchroman action in human breast cancer cells. Indian J. Pharmacol. 2016, 48, 637. [Google Scholar] [CrossRef]
- Farhoudi Sefidan Jadid, M.; Jahangirzadehd, G.; Behroozi, J. Anti-proliferation effects of Apatinib in combination with Curcumin in breast cancer cells. Horm. Mol. Biol. Clin. Investig. 2023, 44, 27–32. [Google Scholar] [CrossRef]
- Alqahtani, A.M.; Chidambaram, K.; Pino-Figueroa, A.; Chandrasekaran, B.; Dhanaraj, P.; Venkatesan, K. Curcumin-Celecoxib: A synergistic and rationale combination chemotherapy for breast cancer. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 1916–1927. [Google Scholar]
- Ziasarabi, P.; Sahebkar, A.; Ghasemi, F. Evaluation of the Effects of Nanomicellar Curcumin, Berberine, and Their Combination with 5-Fluorouracil on Breast Cancer Cells. In Natural Products and Human Diseases; Sahebkar, A., Sathyapalan, T., Eds.; Advances in Experimental Medicine and Biology; Springer International Publishing: Cham, Switzerland, 2021; Volume 1328, pp. 21–35. ISBN 978-3-030-73233-2. [Google Scholar]
- Liu, M.; Yin, H.; Qian, X.; Dong, J.; Qian, Z.; Miao, J. Xanthohumol, a Prenylated Chalcone from Hops, Inhibits the Viability and Stemness of Doxorubicin-Resistant MCF-7/ADR Cells. Molecules 2016, 22, 36. [Google Scholar] [CrossRef]
- Kang, Y.; Park, M.-A.; Heo, S.-W.; Park, S.-Y.; Kang, K.W.; Park, P.-H.; Kim, J.-A. The radio-sensitizing effect of xanthohumol is mediated by STAT3 and EGFR suppression in doxorubicin-resistant MCF-7 human breast cancer cells. Biochim. Biophys. Acta (BBA) General Subj. 2013, 1830, 2638–2648. [Google Scholar] [CrossRef]
- Tu, S.-H.; Chiou, Y.-S.; Kalyanam, N.; Ho, C.-T.; Chen, L.-C.; Pan, M.-H. Garcinol sensitizes breast cancer cells to Taxol through the suppression of caspase-3/iPLA2 and NF-κB/Twist1 signaling pathways in a mouse 4T1 breast tumor model. Food Funct. 2017, 8, 1067–1079. [Google Scholar] [CrossRef]
- Pateliya, B.; Burade, V.; Goswami, S. Enhanced antitumor activity of doxorubicin by naringenin and metformin in breast carcinoma: An experimental study. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 1949–1961. [Google Scholar] [CrossRef] [PubMed]
- Junedi, S.; Hermawan, A.; Fitriasari, A.; Setiawati, A.; Susidarti, R.A.; Meiyanto, E. The Doxorubicin-Induced G2/M Arrest in Breast Cancer Cells Modulated by Natural Compounds Naringenin and Hesperidin. Indones. J. Cancer Chemoprev. 2021, 12, 83–89. [Google Scholar] [CrossRef]
- Soltannezhad, S.; Jouni, F.J.; Osgoei, L.T. The Combined Therapeutic Effect of Capecitabine and Naringin on HER2+ (SK-BR-3) and HER2- (MCF-7) Human Breast Cancer Cells Lines. Res. Sq. 2023. [Google Scholar] [CrossRef]
- Effat, H.; Abosharaf, H.A.; Radwan, A.M. Combined effects of naringin and doxorubicin on the JAK/STAT signaling pathway reduce the development and spread of breast cancer cells. Sci. Rep. 2024, 14, 2824. [Google Scholar] [CrossRef]
- Muthusamy, T.; Yadav, L.R.; Ramalingam, S.; Ramachandran, I.; Nagarajan, P. Synergistic Effect of 5-Fluorouracil Combined with Naringin in MDA-MB-231 Human Breast Cancer Cells. Int. J. Oncol. 2020, 3, 104–118. [Google Scholar]
- Louisa, M.; Mirawati, T.; Suyatna, F. in vitro Modulation of P-glycoprotein, MRP-1 and BCRP Expression by Mangiferin in Doxorubicin-Treated MCF-7 Cells. Asian Pac. J. Cancer Prev. 2014, 15, 1639–1642. [Google Scholar] [CrossRef]
- Wang, T.K.; Xu, S.; Fan, Y.; Wu, J.; Wang, Z.; Chen, Y.; Zhang, Y. The Synergistic Effect of Proanthocyanidin and HDAC Inhibitor Inhibit Breast Cancer Cell Growth and Promote Apoptosis. Int. J. Mol. Sci. 2023, 24, 10476. [Google Scholar] [CrossRef]
- Chen, X.-X.; Leung, G.P.-H.; Zhang, Z.-J.; Xiao, J.-B.; Lao, L.-X.; Feng, F.; Mak, J.C.-W.; Wang, Y.; Sze, S.C.-W.; Zhang, K.Y.-B. Proanthocyanidins from Uncaria rhynchophylla induced apoptosis in MDA-MB-231 breast cancer cells while enhancing cytotoxic effects of 5-fluorouracil. Food Chem. Toxicol. 2017, 107, 248–260. [Google Scholar] [CrossRef]
- Korga-Plewko, A.; Michalczyk, M.; Adamczuk, G.; Humeniuk, E.; Ostrowska-Lesko, M.; Jozefczyk, A.; Iwan, M.; Wojcik, M.; Dudka, J. Apigenin and Hesperidin Downregulate DNA Repair Genes in MCF-7 Breast Cancer Cells and Augment Doxorubicin Toxicity. Molecules 2020, 25, 4421. [Google Scholar] [CrossRef]
- Nimal, S.; Kumbhar, N.; Saruchi; Rathore, S.; Naik, N.; Paymal, S.; Gacche, R.N. Apigenin and its combination with Vorinostat induces apoptotic-mediated cell death in TNBC by modulating the epigenetic and apoptotic regulators and related miRNAs. Sci. Rep. 2024, 14, 9540. [Google Scholar] [CrossRef] [PubMed]
- Soltanian, S.; Riahirad, H.; Pabarja, A.; Karimzadeh, M.R.; Saeidi, K. Kaempferol and docetaxel diminish side population and down-regulate some cancer stem cell markers in breast cancer cell line MCF-7. Biocell 2017, 41, 33. [Google Scholar] [CrossRef]
- Kusharyanti, I.; Larasati, L.; Susidarti, R.A.; Meiyanto, E. Hesperidin Increase Cytotoxic Activity of Doxorubicin on Hela Cell Line Through Cell Cycle Modulation and Apoptotis Induction. Indones. J. Cancer Chemoprev. 2011, 2, 267. [Google Scholar] [CrossRef]
- Nurhayati, I.P.; Khumaira, A.; Ilmawati, G.P.N.; Meiyanto, E.; Hermawan, A. Cytotoxic and Antimetastatic Activity of Hesperetin and Doxorubicin Combination Toward Her2 Expressing Breast Cancer Cells. Asian Pac. J. Cancer Prev. 2020, 21, 1259–1267. [Google Scholar] [CrossRef]
- Febriansah, R.; Dyaningtyas, D.P.P.; Sarmoko; Nurulita, N.A.; Meiyanto, E.; Nugroho, A.E. Hesperidin as a preventive resistance agent in MCF–7 breast cancer cells line resistance to doxorubicin. Asian Pac. J. Trop. Biomed. 2014, 4, 228–233. [Google Scholar] [CrossRef]
- Amalina, N.D.; Salsabila, I.A.; Zulfin, U.M.; Jenie, R.I.; Meiyanto, E. In vitro synergistic effect of hesperidin and doxorubicin downregulates epithelial-mesenchymal transition in highly metastatic breast cancer cells. J. Egypt. Natl. Cancer Inst. 2023, 35, 6. [Google Scholar] [CrossRef]
- Khamis, A.A.; Ali, E.M.M.; Salim, E.I.; El-Moneim, M.A.A. Synergistic effects of bee venom, hesperidin, and piperine with tamoxifen on apoptotic and angiogenesis biomarker molecules against xerographic MCF-7 injected rats. Sci. Rep. 2024, 14, 1510. [Google Scholar] [CrossRef]
- Khan, H.; Ullah, H.; Martorell, M.; Valdes, S.E.; Belwal, T.; Tejada, S.; Sureda, A.; Kamal, M.A. Flavonoids nanoparticles in cancer: Treatment, prevention and clinical prospects. Semin. Cancer Biol. 2021, 69, 200–211. [Google Scholar] [CrossRef]
- Davatgaran-Taghipour, Y.; Masoomzadeh, S.; Farzaei, M.H.; Bahramsoltani, R.; Karimi-Soureh, Z.; Rahimi, R.; Abdollahi, M. Polyphenol nanoformulations for cancer therapy: Experimental evidence and clinical perspective. Int. J. Nanomed. 2017, 12, 2689–2702. [Google Scholar] [CrossRef]
- Yang, B.; Dong, Y.; Wang, F.; Zhang, Y. Nanoformulations to Enhance the Bioavailability and Physiological Functions of Polyphenols. Molecules 2020, 25, 4613. [Google Scholar] [CrossRef]
- Xiong, Q.; Wang, Y.; Wan, J.; Yuan, P.; Chen, H.; Zhang, L. Facile preparation of hyaluronic acid-based quercetin nanoformulation for targeted tumor therapy. Int. J. Biol. Macromol. 2020, 147, 937–945. [Google Scholar] [CrossRef]
- Baksi, R.; Singh, D.P.; Borse, S.P.; Rana, R.; Sharma, V.; Nivsarkar, M. In vitro and in vivo anticancer efficacy potential of Quercetin loaded polymeric nanoparticles. Biomed. Pharmacother. 2018, 106, 1513–1526. [Google Scholar] [CrossRef]
- Sharma, G.; Park, J.; Sharma, A.R.; Jung, J.-S.; Kim, H.; Chakraborty, C.; Song, D.-K.; Lee, S.-S.; Nam, J.-S. Methoxy Poly(ethylene glycol)-Poly(lactide) Nanoparticles Encapsulating Quercetin Act as an Effective Anticancer Agent by Inducing Apoptosis in Breast Cancer. Pharm. Res. 2015, 32, 723–735. [Google Scholar] [CrossRef] [PubMed]
- 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) General Subj. 2016, 1860, 2065–2075. [Google Scholar] [CrossRef] [PubMed]
- Zeng, L.; Yan, J.; Luo, L.; Ma, M.; Zhu, H. Preparation and characterization of (−)-Epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and their inhibitory effects on Human breast cancer MCF-7 cells. Sci. Rep. 2017, 7, 45521. [Google Scholar] [CrossRef]
- Feng, C.; Yuan, X.; Chu, K.; Zhang, H.; Ji, W.; Rui, M. Preparation and optimization of poly (lactic acid) nanoparticles loaded with fisetin to improve anti-cancer therapy. Int. J. Biol. Macromol. 2019, 125, 700–710. [Google Scholar] [CrossRef]
- Patel, G.; Thakur, N.S.; Kushwah, V.; Patil, M.D.; Nile, S.H.; Jain, S.; Kai, G.; Banerjee, U.C. Mycophenolate co-administration with quercetin via lipid-polymer hybrid nanoparticles for enhanced breast cancer management. Nanomed. Nanotechnol. Biol. Med. 2020, 24, 102147. [Google Scholar] [CrossRef]
- Radhakrishnan, R.; Pooja, D.; Kulhari, H.; Gudem, S.; Ravuri, H.G.; Bhargava, S.; Ramakrishna, S. Bombesin conjugated solid lipid nanoparticles for improved delivery of epigallocatechin gallate for breast cancer treatment. Chem. Phys. Lipids 2019, 224, 104770. [Google Scholar] [CrossRef]
- Naderi, E.; Aghajanzadeh, M.; Zamani, M.; Hashiri, A.; Sharafi, A.; Kamalianfar, A.; Naseri, M.; Danafar, H. Improving the anti-cancer activity of quercetin-loaded AgFeO2 through UV irradiation: Synthesis, characterization, and in vivo and in vitro biocompatibility study. J. Drug Deliv. Sci. Technol. 2020, 57, 101645. [Google Scholar] [CrossRef]
- Balakrishnan, S.; Bhat, F.A.; Raja Singh, P.; Mukherjee, S.; Elumalai, P.; Das, S.; Patra, C.R.; Arunakaran, J. Gold nanoparticle–conjugated quercetin inhibits epithelial–mesenchymal transition, angiogenesis and invasiveness via EGFR/VEGFR -2-mediated pathway in breast cancer. Cell Prolif. 2016, 49, 678–697. [Google Scholar] [CrossRef]
- Sulaiman, G.M.; Waheeb, H.M.; Jabir, M.S.; Khazaal, S.H.; Dewir, Y.H.; Naidoo, Y. Hesperidin Loaded on Gold Nanoparticles as a Drug Delivery System for a Successful Biocompatible, Anti-Cancer, Anti-Inflammatory and Phagocytosis Inducer Model. Sci. Rep. 2020, 10, 9362. [Google Scholar] [CrossRef] [PubMed]
- Zarei, M.; Karimi, E.; Oskoueian, E.; Es-Haghi, A.; Yazdi, M.E.T. Comparative Study on the Biological Effects of Sodium Citrate-Based and Apigenin-Based Synthesized Silver Nanoparticles. Nutr. Cancer 2021, 73, 1511–1519. [Google Scholar] [CrossRef] [PubMed]
- Alexander, A.; Ajazuddin; Patel, R.J.; Saraf, S.; Saraf, S. Recent expansion of pharmaceutical nanotechnologies and targeting strategies in the field of phytopharmaceuticals for the delivery of herbal extracts and bioactives. J. Control. Release 2016, 241, 110–124. [Google Scholar] [CrossRef] [PubMed]
- Dutta, D.; Chakraborty, A.; Mukherjee, B.; Gupta, S. Aptamer-Conjugated Apigenin Nanoparticles to Target Colorectal Carcinoma: A Promising Safe Alternative of Colorectal Cancer Chemotherapy. ACS Appl. Bio Mater. 2018, 1, 1538–1556. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Das, J.; Samadder, A.; Paul, A.; Khuda-Bukhsh, A.R. Strategic formulation of apigenin-loaded PLGA nanoparticles for intracellular trafficking, DNA targeting and improved therapeutic effects in skin melanoma in vitro. Toxicol. Lett. 2013, 223, 124–138. [Google Scholar] [CrossRef]
- Das, S.; Das, J.; Samadder, A.; Paul, A.; Khuda-Bukhsh, A.R. Efficacy of PLGA-loaded apigenin nanoparticles in Benzo[a]pyrene and ultraviolet-B induced skin cancer of mice: Mitochondria mediated apoptotic signalling cascades. Food Chem. Toxicol. 2013, 62, 670–680. [Google Scholar] [CrossRef]
- Pourgholi, A.; Dadashpour, M.; Mousapour, A.; Firouzi Amandi, A.; Zarghami, N. Anticancer potential of silibinin loaded polymeric nanoparticles against breast cancer cells: Insight into the apoptotic genes targets. Asian Pac. J. Cancer Prev. 2021, 22, 2587–2596. [Google Scholar] [CrossRef]
- 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. Int. J. Nanomed. 2019, 14, 4045–4057. [Google Scholar] [CrossRef]
- Zhao, J.; Liu, J.; Wei, T.; Ma, X.; Cheng, Q.; Huo, S.; Zhang, C.; Zhang, Y.; Duan, X.; Liang, X.-J. Quercetin-loaded nanomicelles to circumvent human castration-resistant prostate cancer in vitro and in vivo. Nanoscale 2016, 8, 5126–5138. [Google Scholar] [CrossRef]
- Date, A.A.; Nagarsenker, M.S.; Patere, S.; Dhawan, V.; Gude, R.P.; Hassan, P.A.; Aswal, V.; Steiniger, F.; Thamm, J.; Fahr, A. Lecithin-Based Novel Cationic Nanocarriers (Leciplex) II: Improving Therapeutic Efficacy of Quercetin on Oral Administration. Mol. Pharm. 2011, 8, 716–726. [Google Scholar] [CrossRef]
- Dora, C.L.; Costa Silva, L.F.; Mazzarino, L.; Siqueira, J.M.; Fernandes, D.; Pacheco, L.K.; Maioral, M.F.; Santos-Silva, M.C.; Muccillo Baisch, A.L.; Assreuy, J.; et al. Oral Delivery of a High Quercetin Payload Nanosized Emulsion: In Vitro and In Vivo Activity Against B16-F10 Melanoma. J. Nanosci. Nanotechnol. 2016, 16, 1275–1281. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Shi, M.; Ma, B.; Niu, R.; Zhang, H.; Kun, L. Antitumor activity and safety evaluation of nanaparticle-based delivery of quercetin through intravenous administration in mice. Mater. Sci. Eng. C 2017, 77, 803–810. [Google Scholar] [CrossRef] [PubMed]
- Wong, M.-Y.; Chiu, G.N.C. Simultaneous liposomal delivery of quercetin and vincristine for enhanced estrogen-receptor-negative breast cancer treatment. Anti-Cancer Drugs 2010, 21, 401. [Google Scholar] [CrossRef] [PubMed]
- Wong, M.-Y.; Chiu, G.N.C. Liposome formulation of co-encapsulated vincristine and quercetin enhanced antitumor activity in a trastuzumab-insensitive breast tumor xenograft model. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 834–840. [Google Scholar] [CrossRef]
- Yu, J.; Chen, H.; Jiang, L.; Wang, J.; Dai, J.; Wang, J. Codelivery of Adriamycin and P-gp Inhibitor Quercetin Using PEGylated Liposomes to Overcome Cancer Drug Resistance. J. Pharm. Sci. 2019, 108, 1788–1799. [Google Scholar] [CrossRef]
- Minaei, A.; Sabzichi, M.; Ramezani, F.; Hamishehkar, H.; Samadi, N. Co-delivery with nano-quercetin enhances doxorubicin-mediated cytotoxicity against MCF-7 cells. Mol. Biol. Rep. 2016, 43, 99–105. [Google Scholar] [CrossRef]
- Narayanan, S.; Mony, U.; Vijaykumar, D.K.; Koyakutty, M.; Paul-Prasanth, B.; Menon, D. Sequential release of epigallocatechin gallate and paclitaxel from PLGA-casein core/shell nanoparticles sensitizes drug-resistant breast cancer cells. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1399–1406. [Google Scholar] [CrossRef]
- Xu, P.; Yin, Q.; Shen, J.; Chen, L.; Yu, H.; Zhang, Z.; Li, Y. Synergistic inhibition of breast cancer metastasis by silibinin-loaded lipid nanoparticles containing TPGS. Int. J. Pharm. 2013, 454, 21–30. [Google Scholar] [CrossRef]
- Ghosh, P.; Bag, S.; Roy, A.S.; Subramani, E.; Chaudhury, K.; Dasgupta, S. Solubility enhancement of morin and epicatechin through encapsulation in an albumin based nanoparticulate system and their anticancer activity against the MDA-MB-468 breast cancer cell line. RSC Adv. 2016, 6, 101415–101429. [Google Scholar] [CrossRef]
- Rameshthangam, P.; Chitra, J.P. Synergistic anticancer effect of green synthesized nickel nanoparticles and quercetin extracted from Ocimum sanctum leaf extract. J. Mater. Sci. Technol. 2018, 34, 508–522. [Google Scholar] [CrossRef]
- Sabra, S.A.; Elzoghby, A.O.; Sheweita, S.A.; Haroun, M.; Helmy, M.W.; Eldemellawy, M.A.; Xia, Y.; Goodale, D.; Allan, A.L.; Rohani, S. Self-assembled amphiphilic zein-lactoferrin micelles for tumor targeted co-delivery of rapamycin and wogonin to breast cancer. Eur. J. Pharm. Biopharm. 2018, 128, 156–169. [Google Scholar] [CrossRef] [PubMed]
- Jiang, M.; He, K.; Qiu, T.; Sun, J.; Liu, Q.; Zhang, X.; Zheng, H. Tumor-targeted delivery of silibinin and IPI-549 synergistically inhibit breast cancer by remodeling the microenvironment. Int. J. Pharm. 2020, 581, 119239. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Xie, X.; Hou, X.; Shen, J.; Shi, J.; Chen, H.; He, Y.; Wang, Z.; Feng, N. Functional oral nanoparticles for delivering silibinin and cryptotanshinone against breast cancer lung metastasis. J. Nanobiotechnol. 2020, 18, 83. [Google Scholar] [CrossRef] [PubMed]
- Davaran, S.; Fazeli, H.; Ghamkhari, A.; Rahimi, F.; Molavi, O.; Anzabi, M.; Salehi, R. Synthesis and characterization of novel P(HEMA-LA-MADQUAT) micelles for co-delivery of methotrexate and Chrysin in combination cancer chemotherapy. J. Biomater. Sci. Polym. Ed. 2018, 29, 1265–1286. [Google Scholar] [CrossRef]
- Rasouli, S.; Montazeri, M.; Mashayekhi, S.; Sadeghi-Soureh, S.; Dadashpour, M.; Mousazadeh, H.; Nobakht, A.; Zarghami, N.; Pilehvar-Soltanahmadi, Y. Synergistic anticancer effects of electrospun nanofiber-mediated codelivery of Curcumin and Chrysin: Possible application in prevention of breast cancer local recurrence. J. Drug Deliv. Sci. Technol. 2020, 55, 101402. [Google Scholar] [CrossRef]
- Dahiya, S.; Rani, R.; Dhingra, D.; Kumar, S.; Dilbaghi, N. Conjugation of epigallocatechin gallate and piperine into a zein nanocarrier: Implication on antioxidant and anticancer potential. Adv. Nat. Sci Nanosci. Nanotechnol. 2018, 9, 035011. [Google Scholar] [CrossRef]
- Chavva, S.R.; Deshmukh, S.K.; Kanchanapally, R.; Tyagi, N.; Coym, J.W.; Singh, A.P.; Singh, S. Epigallocatechin Gallate-Gold Nanoparticles Exhibit Superior Antitumor Activity Compared to Conventional Gold Nanoparticles: Potential Synergistic Interactions. Nanomaterials 2019, 9, 396. [Google Scholar] [CrossRef]
- Karahaliloğlu, Z.; Kilicay, E.; Alpaslan, P.; Hazer, B.; Baki Denkbas, E. Enhanced antitumor activity of epigallocatechin gallate–conjugated dual-drug-loaded polystyrene–polysoyaoil–diethanol amine nanoparticles for breast cancer therapy. J. Bioact. Compat. Polym. 2018, 33, 38–62. [Google Scholar] [CrossRef]
- Meng, L.; Xia, X.; Yang, Y.; Ye, J.; Dong, W.; Ma, P.; Jin, Y.; Liu, Y. Co-encapsulation of paclitaxel and baicalein in nanoemulsions to overcome multidrug resistance via oxidative stress augmentation and P-glycoprotein inhibition. Int. J. Pharm. 2016, 513, 8–16. [Google Scholar] [CrossRef]
- Jain, A.K.; Thanki, K.; Jain, S. Co-encapsulation of Tamoxifen and Quercetin in Polymeric Nanoparticles: Implications on Oral Bioavailability, Antitumor Efficacy, and Drug-Induced Toxicity. Mol. Pharm. 2013, 10, 3459–3474. [Google Scholar] [CrossRef]
- Sandhu, P.S.; Kumar, R.; Beg, S.; Jain, S.; Kushwah, V.; Katare, O.P.; Singh, B. Natural lipids enriched self-nano-emulsifying systems for effective co-delivery of tamoxifen and naringenin: Systematic approach for improved breast cancer therapeutics. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1703–1713. [Google Scholar] [CrossRef]
- Chung, J.E.; Tan, S.; Gao, S.J.; Yongvongsoontorn, N.; Kim, S.H.; Lee, J.H.; Choi, H.S.; Yano, H.; Zhuo, L.; Kurisawa, M.; et al. Self-assembled micellar nanocomplexes comprising green tea catechin derivatives and protein drugs for cancer therapy. Nat. Nanotechnol. 2014, 9, 907–912. [Google Scholar] [CrossRef]
- Lv, L.; Liu, C.; Chen, C.; Yu, X.; Chen, G.; Shi, Y.; Qin, F.; Ou, J.; Qiu, K.; Li, G. Quercetin and doxorubicin co-encapsulated biotin receptor-targeting nanoparticles for minimizing drug resistance in breast cancer. Oncotarget 2016, 7, 32184–32199. [Google Scholar] [CrossRef]
- Murugan, C.; Rayappan, K.; Thangam, R.; Bhanumathi, R.; Shanthi, K.; Vivek, R.; Thirumurugan, R.; Bhattacharyya, A.; Sivasubramanian, S.; Gunasekaran, P.; et al. Combinatorial nanocarrier based drug delivery approach for amalgamation of anti-tumor agents in breast cancer cells: An improved nanomedicine strategy. Sci. Rep. 2016, 6, 34053. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Du, Q.; Wan, J.; Yu, D.-G.; Tan, F.; Yang, X. Improved synergistic anticancer action of quercetin and tamoxifen citrate supported by an electrospun complex nanostructure. Mater. Des. 2024, 238, 112657. [Google Scholar] [CrossRef]
- Alamri, A.H.; Debnath, S.; Alqahtani, T.; Alqahtani, A.; Alshehri, S.A.; Ghosh, A. Enhancing plant-derived smart nano inhibitor in targeting mammalian target of rapamycin (mTOR) in breast cancer using Curcuma longa-derived compound curcumin. Environ. Sci. Pollut. Res. 2023, 31, 46462–46469. [Google Scholar] [CrossRef] [PubMed]
- Rajaei, M.; Pourmadadi, M.; Rashedi, H.; Yazdian, F.; Navaei-Nigjeh, M.; Rahdar, A.; Ferreira, L.F.R. Improving Curcumin Constraints with pH-Responsive Chitosan Based Graphene Oxide/Montmorillonite Nanohybrid Modified Agarose in Breast Cancer Therapy. BioNanoScience 2024, 14, 1613–1626. [Google Scholar] [CrossRef]
- Nurjis, F.; Sarwar, U.; Ali, J.S.; Fayyaz, M. Doxorubicin and Curcumin-Loaded Nanomicelles Targeting Multidrug Resistance in Cancer. BioNanoScience 2024, 14, 2159–2169. [Google Scholar] [CrossRef]
- Wang, J.; Han, X.; Bai, D. Reversal of drug resistance in breast cancer cells by photodynamic action mediated by curcumin/Ko143/PLGA nanoparticles. AIP Adv. 2024, 14, 045341. [Google Scholar] [CrossRef]
- Lin, X.; Wang, Q.; Du, S.; Guan, Y.; Qiu, J.; Chen, X.; Yuan, D.; Chen, T. Nanoparticles for co-delivery of paclitaxel and curcumin to overcome chemoresistance against breast cancer. J. Drug Deliv. Sci. Technol. 2023, 79, 104050. [Google Scholar] [CrossRef]
- Saharkhiz, S.; Zarepour, A.; Nasri, N.; Cordani, M.; Zarrabi, A. A comparison study between doxorubicin and curcumin co-administration and co-loading in a smart niosomal formulation for MCF-7 breast cancer therapy. Eur. J. Pharm. Sci. 2023, 191, 106600. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, R.; Kasabri, V.; Sunoqrot, S.; Shalabi, D.; Alkhateeb, R.; Alhiari, Y. Preparation and Characterization of Rutin-Encapsulated Polymeric Micelles and Studies of Synergism with Bioactive Benzoic Acids and Triazolofluoroquinolones as Anticancer Nanomedicines. Asian Pac. J. Cancer Prev. 2023, 24, 977. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Liu, J. Facile fabrication of combination delivery of kaempferol and sorafenib with PEGylated gold nanoparticles delivery to breast cancer: Investigation of cytotoxicity and apoptosis induction. J. Macromol. Sci. 2023, 60, 628–639. [Google Scholar] [CrossRef]
- Pingale, P.; Kendre, P.; Pardeshi, K.; Rajput, A. An emerging era in manufacturing of drug delivery systems: Nanofabrication techniques. Heliyon 2023, 9, e14247. [Google Scholar] [CrossRef]
- Bilati, U.; Allémann, E.; Doelker, E. Development of a nanoprecipitation method intended for the entrapment of hydrophilic drugs into nanoparticles. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2005, 24, 67–75. [Google Scholar] [CrossRef]
- Javani, R.; Hashemi, F.S.; Ghanbarzadeh, B.; Hamishehkar, H. Quercetin-loaded niosomal nanoparticles prepared by the thin-layer hydration method: Formulation development, colloidal stability, and structural properties. LWT 2021, 141, 110865. [Google Scholar] [CrossRef]
- Halevas, E.G.; Avgoulas, D.I.; Katsipis, G.; Pantazaki, A.A. Flavonoid-liposomes formulations: Physico-chemical characteristics, biological activities and therapeutic applications. Eur. J. Med. Chem. Rep. 2022, 5, 100059. [Google Scholar] [CrossRef]
- Hoa, L.T.M.; Chi, N.T.; Nguyen, L.H.; Chien, D.M. Preparation and characterisation of nanoparticles containing ketoprofen and acrylic polymers prepared by emulsion solvent evaporation method. J. Exp. Nanosci. 2012, 7, 189–197. [Google Scholar] [CrossRef]
- Zhang, X.; Kang, H.; Zhang, L.; Yang, W. Sonication-Assisted Electrochemical Synthesis of Silver Nanoparticles. Langmuir 2024, 40, 22455–22461. [Google Scholar] [CrossRef]
- Nguyen, T.D.; Hoang, Y.H.; Thai, N.T.-T.; Trinh, G.T.-N. Synthesis of copper nanoparticles by a sonication-mediated method using Malpighia glabra fruit extract and their applications. RSC Adv. 2024, 14, 34119–34134. [Google Scholar] [CrossRef]
- Gimondi, S.; Ferreira, H.; Reis, R.L.; Neves, N.M. Microfluidic Devices: A Tool for Nanoparticle Synthesis and Performance Evaluation. ACS Nano 2023, 17, 14205–14228. [Google Scholar] [CrossRef] [PubMed]
- Viegas, C.; Seck, F.; Fonte, P. An insight on lipid nanoparticles for therapeutic proteins delivery. J. Drug Deliv. Sci. Technol. 2022, 77, 103839. [Google Scholar] [CrossRef]
- Ewii, U.E.; Attama, A.A.; Olorunsola, E.O.; Onugwu, A.L.; Nwakpa, F.U.; Anyiam, C.; Chijioke, C.; Ogbulie, T. Nanoparticles for drug delivery: Insight into in vitro and in vivo drug release from nanomedicines. Nano TransMed 2025, 4, 100083. [Google Scholar] [CrossRef]
- Anwar Ul Alam, M.; Kassu, A.; Kassama, L. Effect of sonication time and surfactant concentration on improving the bio-accessibility of lycopene synthesized in poly-lactic co-glycolic acid nanoparticles. arXiv 2023, arXiv:2301.10850. [Google Scholar]
- Sarheed, O.; Dibi, M.; Ramesh, K.V.R.N.S. Studies on the Effect of Oil and Surfactant on the Formation of Alginate-Based O/W Lidocaine Nanocarriers Using Nanoemulsion Template. Pharmaceutics 2020, 12, 1223. [Google Scholar] [CrossRef]
- Izadiyan, Z.; Webster, T.J.; Kia, P.; Kalantari, K.; Misran, M.; Rasouli, E.; Maghareh Esfahan, Z.; Shameli, K. Nanoemulsions Based Therapeutic Strategies: Enhancing Targeted Drug Delivery against Breast Cancer Cells. Int. J. Nanomed. 2025, 20, 6133–6162. [Google Scholar] [CrossRef]
- Kumar, G.; Virmani, T.; Chhabra, V.; Virmani, R.; Pathak, K.; Akhtar, M.S.; Hussain Asim, M.; Arshad, S.; Siddique, F.; Fonte, P. Transforming cancer treatment: The potential of nanonutraceuticals. Int. J. Pharm. 2024, 667, 124919. [Google Scholar] [CrossRef]
- Talib, W.H.; Abuawad, A.; Thiab, S.; Alshweiat, A.; Mahmod, A.I. Flavonoid-based nanomedicines to target tumor microenvironment. OpenNano 2022, 8, 100081. [Google Scholar] [CrossRef]
- Abdi Syahputra, R.; Dalimunthe, A.; Utari, Z.D.; Halim, P.; Sukarno, M.A.; Zainalabidin, S.; Salim, E.; Gunawan, M.; Nurkolis, F.; Park, M.N.; et al. Nanotechnology and flavonoids: Current research and future perspectives on cardiovascular health. J. Funct. Foods 2024, 120, 106355. [Google Scholar] [CrossRef]
- Stevens Barrón, J.C.; Chapa González, C.; Álvarez Parrilla, E.; De la Rosa, L.A. Nanoparticle-Mediated Delivery of Flavonoids: Impact on Proinflammatory Cytokine Production: A Systematic Review. Biomolecules 2023, 13, 1158. [Google Scholar] [CrossRef]
- Lu, H.; Zhang, S.; Wang, J.; Chen, Q. A Review on Polymer and Lipid-Based Nanocarriers and Its Application to Nano-Pharmaceutical and Food-Based Systems. Front. Nutr. 2021, 8, 783831. [Google Scholar] [CrossRef]
- Mall, J.; Naseem, N.; Haider, M.F.; Rahman, M.A.; Khan, S.; Siddiqui, S.N. Nanostructured lipid carriers as a drug delivery system: A comprehensive review with therapeutic applications. Intell. Pharm. 2025, 3, 243–255. [Google Scholar] [CrossRef]
- Hashim, M.; Mujahid, H.; Hassan, S.; Bukhari, S.; Anjum, I.; Hano, C.; Abbasi, B.H.; Anjum, S. Implication of Nanoparticles to Combat Chronic Liver and Kidney Diseases: Progress and Perspectives. Biomolecules 2022, 12, 1337. [Google Scholar] [CrossRef] [PubMed]
- Xuan, L.; Ju, Z.; Skonieczna, M.; Zhou, P.; Huang, R. Nanoparticles-induced potential toxicity on human health: Applications, toxicity mechanisms, and evaluation models. MedComm 2023, 4, e327. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; Kumar, A.; Bhardwaj, S.; Sikarwar, M.; Sriwastaw, S.; Sharma, G.; Gupta, M. Nanotoxicity unveiled: Evaluating exposure risks and assessing the impact of nanoparticles on human health. J. Trace Elem. Miner. 2025, 13, 100252. [Google Scholar] [CrossRef]
- Vignesh, A.; Menaka, C.; Amal, T.C.; Selvakumar, S.; Vasanth, K. Exploring the dual impact of nanoparticles on human well-being: A comprehensive review of risks and benefits. Next Nanotechnol. 2025, 8, 100223. [Google Scholar] [CrossRef]
- Aljabali, A.A.; Obeid, M.A.; Bashatwah, R.M.; Serrano-Aroca, Á.; Mishra, V.; Mishra, Y.; El-Tanani, M.; Hromić-Jahjefendić, A.; Kapoor, D.N.; Goyal, R.; et al. Nanomaterials and Their Impact on the Immune System. Int. J. Mol. Sci. 2023, 24, 2008. [Google Scholar] [CrossRef]
- Gefen, T.; Vaya, J.; Khatib, S.; Harkevich, N.; Artoul, F.; Heller, E.D.; Pitcovski, J.; Aizenshtein, E. The impact of PEGylation on protein immunogenicity. Int. Immunopharmacol. 2013, 15, 254–259. [Google Scholar] [CrossRef]
- Simberg, D.; Barenholz, Y.; Roffler, S.R.; Landfester, K.; Kabanov, A.V.; Moghimi, S.M. PEGylation technology: Addressing concerns, moving forward. Drug Deliv. 2025, 32, 2494775. [Google Scholar] [CrossRef]
- Office of the Commissioner of the FDA’s Approach to Regulation of Nanotechnology Products. 2019. Available online: https://www.fda.gov/science-research/nanotechnology-programs-fda/fdas-approach-regulation-nanotechnology-products (accessed on 1 January 2020).
- Drug Products, Including Biological Products That Contain Nanomaterials—Guidance for Industry. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/drug-products-including-biological-products-contain-nanomaterials-guidance-industry (accessed on 24 September 2025).
- European Medicines Agency (EMA). Joint MHLW-EMA Reflection Paper on the Development of Block Copolymer Micelle Medicinal Products. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/joint-mhlwema-reflection-paper-development-block-copolymer-micelle-medicinal-products_en.pdf (accessed on 28 September 2025).
- European Medicines Agency (EMA). Data Requirements for Intravenous Liposomal Products Developed with Reference to an Innovator Liposomal Product—Scientific Guideline. 2013. Available online: https://www.ema.europa.eu/en/data-requirements-intravenous-liposomal-products-developed-reference-innovator-liposomal-product-scientific-guideline (accessed on 24 September 2025).
- Committee for Medicinal Products for Human Use. Reflection Paper on Surface Coatings: General Issues for Consideration Regarding Parenteral Administration of Coated Nanomedicine Products; European Medicines Agency, Committee for Medicinal Products for Human Use: London, UK, 2013. [Google Scholar]
- International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use Guidelines. 2024. Available online: https://www.gov.uk/guidance/international-council-for-harmonisation-of-technical-requirements-for-pharmaceuticals-for-human-use-guidelines (accessed on 24 September 2025).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).