
Flavonoids as modulators of metabolic reprogramming in renal cell carcinoma (Review)
- Authors:
- Published online on: October 15, 2024 https://doi.org/10.3892/or.2024.8826
- Article Number: 167
-
Copyright: © Shahzad et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Renal cell carcinoma (RCC), also known as renal adenocarcinoma, is a type of kidney cancer that arises from the renal tubule cells, representing 2–3% of all adult cancers (1). Each year, physicians identify an estimated 210,000 new instances of kidney cancer worldwide, making it the seventh most prevalent cancer in men and the ninth most common cancer in women (2). The United States, northern Europe, Canada, Australia, and New Zealand have the highest rates of kidney cancer according to the National Cancer Institute. Thailand, the Philippines, and China have the lowest incidences (3). Clear cell RCC (ccRCC) accounts for 70–80% of RCC cases, while the other subtypes are primarily composed of chromophobe tumors (3–5%) and papillary tumors (10–15%) (4). Furthermore, ~4% of RCCs are hereditary and 96% are sporadic (5). Cancers are characterized by disordered metabolism (6). Tumor growth is facilitated by certain metabolic processes that are necessary for cell transformation or other related biological processes (7). Cancer cells can modify their metabolic systems in various ways to meet their needs for energy and biosynthesis. The first strategy involves increasing the intake and utilization of nutrients and substrates, mainly glucose since the glycolytic rate of some types of cancer cells is ~30 times higher than that of normal cells (8). The second approach is to use metabolic pathways that are advantageous to biosynthesis when breaking down nutrients (9). The Warburg effect, which refers to the observation that cancer cells tend to use anaerobic glycolysis to utilize glucose even in aerobic environments, is one of the most important examples (10). The third strategy involves the aberrant activation of biosynthetic pathways. Tumor cells actively engage in pathways related to fatty acid production and desaturation to meet the increased demands for their cell membranes and signaling molecules (11,12). Reprogrammed metabolic processes are crucial for the proliferation of cancer cells and are also becoming recognized as a critical factor in determining the outcome of the individual cells (13). Kidney cancer or renal cancer is a prime example of metabolic reprogramming among cancer types due to its distinct reliance on altered metabolic pathways to support tumor growth and adaption (14,15). Researchers have linked numerous altered, inactive, or hyperactivated genes in RCC to the control of several metabolic processes, including glutamine metabolism, glycolysis, and the tricarboxylic acid (TCA) cycle (16,17), ATP synthesis, and the regulation of pathways crucial for the balance of redox and hypoxia reactions. Kidney cancer can also be characterized by its metabolic alterations, as the disease involves marked changes in metabolic pathways that support tumor growth and survival (17,18).
Therapeutic approaches for renal cancer are often hindered by the resistance to multiple drugs exhibited by tumor cells, which is a major cause of chemotherapy failure (19). This resistance arises from metabolic and cellular physiological responses triggered by the tumor, including the evasion of drug-induced apoptosis, activation of detoxification pathways, reduction in drug uptake and activation of DNA repair mechanisms (20). In this context, the utilization of flavonoids in a clinical trial has been instrumental in suppressing resistance mechanisms and inducing reprogramming of cancer cells, and thus, is of utmost importance in the search for novel genotoxic therapeutic approaches against tumors (21). These natural products have facilitated the development of more effective strategic combinations with fewer side effects for the treatment of renal cancer and have enhanced the understanding of metabolic reprogramming, cancer cell defense and resistance mechanisms (22). The alteration of gene expression patterns in ccRCC cells is linked to genetic and epigenetic events (23). Flavonoids have gained prominence in anticancer pharmaceutical studies since they inhibit glycolysis and oxidative phosphorylation (OXPHOS), and modulate key enzymes such as hexokinase (HK) and pyruvate kinase (24,25). A summary of the metabolic reprogramming altered by flavonoids in ccRCC is shown in Fig. 1. Flavonoids possess potent epigenetic properties that regulate DNA methylation, histone modification, and microRNAs in the context of cancer therapy. Specifically, flavonoids exhibit the ability to modulate crucial metabolic pathways by targeting tumor suppressor genes and pivotal catalytic enzymes in cancer cells (24,26). Their efficacy has been demonstrated when combined with chemotherapy drugs or other natural compounds, thus driving extensive research efforts and the development of novel therapeutic strategies for cancer treatment (27).
Anticancer drugs targeting metabolic reprogramming
The field of cancer biology has observed a gradual increase in knowledge and comprehension of the numerous targets of metabolic reprogramming as advancements in research occur. These advancements have facilitated the development of customized pharmacological therapies for various components of metabolism (28). Medicines targeting pertinent metabolic pathways may inhibit the growth and proliferation of RCC cells (29). Scientists categorize drugs according to their target metabolic pathways, which include glucose metabolism, glycolysis, tricarboxylic acid cycle, oxidative phosphorylation, pentose phosphate pathway, lactate metabolism, lipid metabolism, and amino acid metabolism. Table SI summarizes these metabolic pathways, detailing the drugs targeting each pathway, the models in which they have been tested, and their current phase of research. While the majority of anticancer medications are still in the preclinical stage, a few have already exhibited significant promise in cancer treatment and have advanced to phase IV studies or clinical trials (30). For example, curcumin derived from Curcuma longa has been investigated for its potential to target metabolic reprogramming in cancer cells, including modulation of glycolysis and oxidative stress pathways (31). Resveratrol has shown potential in targeting metabolic pathways such as glucose metabolism and mitochondrial function in cancer cells (32). Regarding treatment options for recurrent colorectal cancer, tumor drug-induced cell drug resistance is considered to be the primary factor for the ineffectiveness of chemotherapy. This occurs because the drug-induced apoptosis decreases drug absorption and activates DNA repair mechanisms (33). Natural molecules have several advantages over synthetic medications or chemicals, including reasonable safety, minimal side effects, and multistep targeting (34–36). Numerous natural substances have been employed as preventative and therapeutic measures against various illnesses, including cancer (37–39). Some molecules from food and beverages, traditional Chinese medicines, and medicinal plants have an impact on the initiation, growth and metastasis of human cancers (40). For example, curcumin has been shown to inhibit cancer cell proliferation, induce apoptosis and reduce metastasis in various cancer types through multiple mechanisms, including modulation of signaling pathways and reduction of inflammation (41). Berberine has shown potential in inhibiting cancer cell growth, and modulating multiple signaling pathways involved in cancer progression (42). Epigallocatechin-3-gallate (EGCG) has demonstrated anticancer effects by inducing apoptosis and blocking angiogenesis (43). Although numerous secondary metabolites, such as flavonoids from medicinal plants, have been utilized for a long time, it is still unclear which molecular mechanism underlies their tumor-suppressive actions and which anticancer properties they have (44).
Role of flavonoids in RCC metabolic reprogramming
There are >10,000 known subtypes of bioflavonoids, making them a diverse class of natural compounds. The most prevalent phenolic compounds in the human diet are flavonoids, mostly in cereals. Dried food, nuts, seeds, fruits, vegetables, cereal-like foods, green tea and wine are everyday dietary items that are frequently consumed and can be sources of bioactive compounds, including flavonoids. These compounds have been studied for their potential health benefits, including their anticancer properties. These everyday dietary items are readily available and can be incorporated into a regular diet. Their consumption may contribute to overall health and potentially offer protective effects against cancer through their bioactive compounds (24). Depending on the degree of unsaturation, oxidation of the C ring, and chemical structure, flavonoids can be further classified into six subgroups. These subcategories include isoflavones or chalcones, anthocyanins, flavones, flavonols and flavanones or catechin. Each flavonoid is composed of two benzene rings joined by a heterocyclic pyran ring (2-phenyl-1,4-benzopyran) and has 15 carbons in its chemical structure (C6-C3-C6) (Fig. 2) (45,46). Flavonoids exhibit numerous biological properties, including antiviral (47), antifungal (48), antibacterial (49), antioxidant (50), anti-inflammatory (51), antidiabetic (52), antimutagenic (53), anti-obesity (54), cardioprotective (55) and anticancer (24) activities. Moderate levels of reactive oxygen species (ROS), generated through mitochondrial activity, function as redox signaling molecules that regulate growth, differentiation, and cell proliferation pathways. However, excessive ROS can be detrimental to cancer cells, leading to cell death. As a result, tumor cells develop adaptive detoxification mechanisms to counteract high levels of ROS (56). The elevation of ROS can induce apoptosis in cancer cells, and therapeutic strategies that modulate ROS levels have exhibited efficacy as anticancer drugs (57). Flavonoids exhibit dual activity: Antioxidant effects in non-tumor cells and pro-oxidant effects in cancer cells. In non-tumor cells, flavonoids exert antioxidant effects, reducing oxidative stress by scavenging ROS. Conversely, in cancer cells, flavonoids induce oxidative stress, increasing ROS levels and thereby inhibiting cell proliferation signaling and metabolic reprogramming of cancer cells, suppressing pro-inflammatory cytokines, and promoting apoptosis, necrosis, and autophagy (58). The ability of flavonoids to scavenge ROS is attributed to the presence of a number of phenolic hydroxyl groups in their molecular structure, which facilitate electron exchange and stable compound formation through substitution reactions with free radicals. Therefore, flavonoids with a higher number of hydroxyl groups exhibit greater antioxidant and pro-oxidant capacities (59,60). A study has demonstrated that ovarian cancer cells treated with flavonoids, such as apigenin, luteolin and myricetin, exhibit a dose-dependent increase in intracellular ROS levels compared with untreated control cells. This ROS elevation triggers activation of the intrinsic apoptotic pathway, leading to cell cycle arrest and inhibition of invasion (61). Similarly, quercetin has been reported to induce cancer cell death by positively modulating ROS levels (62).
ccRCC undergoes metabolic reprogramming to sustain excessive cell proliferation, directly influencing the maintenance and aggressiveness of neoplastic cells (63). For instance, glutathione (GSH) metabolism has been extensively investigated in tumor progression and as a targeted therapeutic strategy for cancer (64,65). The upregulation of GSH levels is closely associated with cellular detoxification mechanisms, providing certain types of cancer, including breast cancer, in which elevated GSH levels have been linked to resistance against chemotherapy drugs such as doxorubicin and cisplatin, and non-small cell lung cancer, in which high levels of GSH contribute to resistance against various chemotherapeutic agents such as cisplatin and paclitaxel, with an advantage by eliminating and detoxifying specific chemotherapeutic agents, thus conferring therapeutic resistance (66,67). Additionally, elevated levels of GSH contribute to tumor development and increase the likelihood of metastasis. Depletion of GSH levels can induce various types of cell death, including apoptosis, necroptosis, ferroptosis, and autophagy (68). This serves as the foundation for studies investigating the suppression of GSH levels as a chemo-sensitization approach in cancer therapies, rendering tumor cells more susceptible to the cytotoxic and cytoprotective effects of antineoplastic agents (69,70). In this context, tangeretin has been shown to prevent GSH depletion in cells exposed to tert-butyl hydroperoxide (71). The anticancer properties of flavonoids have been extensively studied, revealing that they mediate antitumor effects through multiple mechanisms. These include promoting autophagy and apoptosis, inhibiting tumor invasion, growth and angiogenesis, as well as modulating ROS levels in tumor cells (24). Additionally, flavonoids can inhibit carcinogens and regulate pro-inflammatory pathways, further contributing to their potential as therapeutic agents (51).
Flavonoids, a group of naturally occurring polyphenolic compounds, exhibit anticancer properties by modulating various key processes involved in carcinogenesis (72). These processes include apoptosis, proliferation, angiogenesis and metastatic progression, which are often driven by dysregulation of tumor suppressor genes and activation of oncogenes (73). Quercetin and genistein have been demonstrated to induce apoptosis in cancer cells through the activation of intrinsic and extrinsic apoptotic pathways. They enhance the expression of pro-apoptotic proteins (Bax and p53) and inhibit anti-apoptotic proteins (Bcl-2) (74). EGCG and lutein inhibit cyclin-dependent kinases (CDKs) and upregulate CDK inhibitors, including p21 and p27, leading to cycle arrest (75,76). Apigenin and kaempferol have been reported to inhibit angiogenesis by downregulating VEGF and its receptors, thereby reducing the supply of nutrients to tumors (77,78). Naringenin and hesperidin have been demonstrated to suppress epithelial-mesenchymal transition markers, including vimentin and N-cadherin, and reduce the activity of MMPs, limiting cancer cell invasion and migration (79). A study has demonstrated that flavonoids exert anticancer effects by regulating cascades that influence the metabolic reprogramming of various pathways, including lipid metabolism, amino acid metabolism and ketogenesis, in both in vitro and in vivo experiments (80), and they represent a viable strategy to inhibit key stages in the development of cancer (81). Table SII summarizes the findings of research studies investigating the influence of flavonoids on critical metabolic pathway components across various cancer types, with distinctions made where the studies focus specifically on RCC.
Glucose metabolism in RCC
The fundamental and significant source of energy in the biological system is glucose. In RCC cells, glucose deprivation causes oxidative stress and cellular cytotoxicity (82). Several mechanisms, including glucose absorption, glycolysis, glycogenolysis, gluconeogenesis, lactate reabsorption, and lactate excretion, maintain glucose homeostasis (83) (Fig. 3).
Transmembrane glucose transport
Renal carcinoma cells need more glucose than normal cells to function as an energy source and as a resource for the production of various chemicals (84). Several cancer cell types exhibit upregulated expression levels of glucose transporters (GLUTs), particularly GLUT1, to maintain steady glucose uptake. Therefore, targeting GLUTs using various natural substances, such as flavonoids, is an optimal strategy for RCC treatment. Catechins from green tea exhibit inhibitory action against GLUT1 in RCC (85). Other polyphenols, such as epicatechin gallate (ECG) and EGCG, inhibit GLUT1 by directly binding to the transporter (86), which can change how it recognizes its substrates competitively or non-competitively. The connections of the extracellular side of the transporter with ECG and EGCG can competitively prevent glucose from binding to GLUT1 (87). Studies have demonstrated that quercetin exerts non-competitive inhibition by binding to GLUT1. Kinetic analysis has also demonstrated that flavonoids can decrease the Michaelis constant and maximum velocity values of GLUT1 (87,88). Genistein, an iso-flavonoid compound, exerts a preventive effect against prostate and breast cancer by blocking GLUT1 activity and acting as a competitive inhibitor (89). Due to its potent activities, including antioxidant, chemopreventive and anticancer activities, resveratrol might directly inhibit GLUT1 by non-competitively binding to its internal domains, decreasing uptake of glucose in human leukemia cells (90). Flavonoids, natural substances in plants with anticancer and antioxidant properties, might affect GLUTs in RCC (91,92). Flavonoids may affect how RCC cells utilize glucose, interfering with their energy metabolism and preventing the formation of tumors (24). More research is required to fully comprehend the mechanism and therapeutic implications of flavonoids in the metabolic reprogramming of RCC. Research on the possible therapeutic effect of flavonoids in targeting glucose metabolism in renal cancer is ongoing.
Glycolysis
In normal cells, glycolysis converts the majority of glucose to pyruvate. The mitochondria undergo this process to participate in the TCA cycle (83). Through OXPHOS, pyruvate causes the synthesis of ATP. Renal cancer cells use the enzyme lactate dehydrogenase (LDH)A to ferment lactic acid to convert pyruvate into lactate (93). This mechanism produces less energy compared with OXPHOS (94). To compensate for this lower energy yield, renal cancer cells require a higher rate of glucose consumption. A complex web of processes, including glucose absorption, glycolysis, glycogenolysis, gluconeogenesis, glucose reabsorption, and glucose excretion, manages glucose homeostasis, which is influenced by kidney function (83). ccRCC cells exhibit increased glycolysis, suppressed pyruvate dehydrogenase (PDH) flux, and decreased TCA cycle activity compared with other tumor cells. A substantial difference is observed when comparing the rate of glycolysis in ccRCC cells with that in adjacent kidney cells (94). Renal cancer cells have been found to exhibit the conventional Warburg effect, which is characterized by increased cellular expression of all glycolysis-related enzymes (93). Fructose-1,6-bisphosphate, the rate-limiting enzyme, is also recognized as a tumor suppressor in RCC tumors (94). Various biological effects, including antioxidative, antiangiogenic and general antitumor effects, have been attributed to certain phytochemicals (24,47–49). Flavonoids target the modulation of certain glycolysis-related enzyme activities (24); therefore, flavonoids offer a promising therapeutic strategy for cancer-related studies. A common dietary flavonoid, apigenin, inhibits multiple biochemical pathways involved in the formation of tumors and has anticancer properties. Apigenin inhibits glycolysis by regulating pyruvate kinase M2 (PKM2) activity in HCT116 colon cancer cells (95). Inhibition by apigenin can result in the maintenance of a low PKM2/pyruvate kinase 1 ratio (96). Proanthocyanidin B2 influences PKM2 activity in hepatocellular carcinoma (97). The first irreversible stage in glycolysis is the phosphorylation of hexoses, carried out by the enzyme HK (98). Since cancer cells mostly exhibit upregulated expression levels of HK, HK may be a promising molecular target for flavonoid-based treatment (99). Xanthohumol is a flavonoid that affects colon cancer by inhibiting HK2, a crucial enzyme involved in glycolysis (98). In hepatocellular carcinoma, quercetin inhibits Akt/mTOR signaling and decreases the activity of HK2 (99). Both in vitro and in vivo, the synthetic flavonoids Gl-v9 and 10v reduce HK2 expression (100,101). Morin, a flavonoid, may prevent LDH from acting enzymatically in RCC (102). In addition, to efficiently inhibiting the growth and proliferation of several cancer cell lines, quercetin also reduces the levels of enzymes linked to glycolysis, such as LDH and LDHA (103). The flavonoid EGCG decreases the activity of LDH, LDHA and phosphofructokinase in in vitro analysis (86,104,105). Quercetin also downregulates the enzymatic activity of aldolase, GAPDH and α-enolase (106). Overall, researchers are currently studying how flavonoids affect glycolysis in the metabolic reprogramming of RCC. Although the preclinical results are encouraging, more thorough research, including clinical trials, is required to determine whether flavonoids can be used to treat RCC by modulating glycolytic pathways.
TCA cycle
Nephrological disorders, such as type 2 diabetes, chronic kidney disease, and kidney damage, can disrupt the TCA cycle by impairing mitochondrial function and reducing levels of key TCA cycle intermediates leading to altered energy metabolism (107,108). The enzyme PDH restores the metabolic flux to the TCA cycle in renal cancer cells, which is often disrupted by downregulation of pathways such as glycolysis, lipid metabolism, and amino acid metabolism (93). These enzymes catalyze the biochemical reactions that generate end products, which either enter the TCA cycle directly or are converted into intermediates that feed into the cycle (94). PDH catalyzes the conversion of pyruvate into acetyl-CoA (109,110). Different flavonoids, such as betulinic acid, can inhibit the activity of PDH in cancer cells, as demonstrated in an in vitro study (111). Typically, RCC cells divert glucose for aerobic glycolysis breakdown from the TCA cycle. Thus, glutamine and fatty acids are needed by kidney cancer cells to power the TCA cycle (111,112). Further study is required to fully establish the direct actions of flavonoids against the TCA cycle in the metabolic reprogramming of RCC. Future research is needed to fully understand how flavonoids affect RCC metabolism, particularly given the complex interactions suggested by their potential influence on mitochondrial function, redox balance, and enzyme activity.
OXPHOS
Normal kidney cells filter blood and reabsorb nutrients, which are processes that are heavily dependent on ATP. These cells have high OXPHOS activity, which is supported by the electron transport chain (ETC) (94). In renal cancer cells, decreasing the activity of the TCA cycle also results in diminished ETC activity (94). A study has measured the activity of OXPHOS and the ETC in RCC tissues (93). From the less aggressive to the most aggressive type of RCC, there is an increase in mitochondrial damage. Marked downregulation of OXPHOS complexes, which impairs the overall OXPHOS process, has been observed. This impairment is linked to dysfunction in the ETC, which is responsible for generating the proton gradient required for ATP synthesis (113). Different phytochemicals present in various medicinal plants control the activity of different complexes. Curcumin is a flavonoid that suppresses the activity of ATP synthase (113–116). A study examined the role of metabolic reprogramming in breast cancer, specifically focusing on the targeting of the ATP synthase complex (117). The flavonoid arctigenin also inhibits mitochondrial complexes II and IV, selectively killing only OXPHPOS-dependent pancreatic cancer cells (116). Some flavonoids, including luteolin, myricetin, fisetin, rhammetin, and baicalein, directly inhibit complex I activity by lowering H2O2 production in rat heart mitochondria (118). Other flavonoids, such as hispidulin and eupafolin, inhibit complex III by lowering H2O2 production (118). Flavonoids may influence mitochondrial processes that could modify cellular energy metabolism, thereby potentially affecting OXPHOS in the metabolic reprogramming of RCC. The therapeutic implications of flavonoids in targeting OXPHOS in RCC require additional investigation, including clinical studies.
Kidney cancer cells, particularly ccRCC cells, exhibit distinct metabolic reprogramming that supports their rapid proliferation and survival under hypoxic conditions. This reprogramming often involves a shift from OXPHOS to glycolysis, even in the presence of oxygen (91). Hypoxia-inducible factor 1 (HIF-1) serves a central role in this metabolic adaptation. HIF-1 is a transcription factor that is stabilized and activated under hypoxic conditions, leading to the upregulation of genes involved in glycolysis, angiogenesis, and cell survival (91,119). Quercetin, resveratrol, and EGCG have been shown to decrease HIF-1α protein levels by promoting its degradation and inhibiting its synthesis (91,120). This effect occurs through the inhibition of the PI3K/Akt/mTOR signaling pathway, which serves a crucial role in the translation and stabilization of HIF-1α (121). Kaempferol influences mitochondrial function and OXPHOS, providing an additional layer of metabolic regulation by activating the AMP-activated protein kinase pathway. This shifts the metabolic balance towards OXPHOS, reducing the reliance on glycolysis and inhibiting cancer cell proliferation (73). Luteolin promotes mitochondrial apoptosis by increasing the production of ROS and disrupting the mitochondrial membrane potential. This leads to the activation of caspase-dependent apoptotic pathways, thereby reducing the survival of cancer cells (122).
Pentose phosphate pathway (PPP)
Glycolysis is diverted from glucose 6-phosphate (G-6-P) to fructose 6-phosphate by the PPP. In the context of cellular metabolism, this process provides crucial components for nucleotide synthesis. Specifically, it generates five-carbon sugars (ribose and deoxyribose) and reduces equivalents in the form of NADPH. The five-carbon sugars are essential for the backbone structure of nucleotides, which are the building blocks of DNA and RNA. NADPH is a key reducing agent that supplies the necessary electrons for various biochemical reactions, including those involved in the synthesis of nucleotides. This synthesis is vital for cell proliferation and repair, making these components critical for maintaining cellular function and integrity (123). The PPP in renal cancer cells acts as a defense against high levels of oxidative stress. This pathway influences kidney diseases by altering key metabolic processes involved in disease progression. For example, in diabetic kidney disease, the pathway can affect the accumulation of advanced glycation end-products and oxidative stress, which are critical in the development of diabetic nephropathy. By modulating these factors, the pathway may help mitigate inflammation, fibrosis, and oxidative damage associated with kidney injury. In cases of kidney damage, the pathway modulation of oxidative stress and cellular repair mechanisms can impact the extent of tissue damage and repair processes, potentially slowing disease progression and improving renal function (12,93). According to a previous study, RCCs may rewire their metabolism to control glucose flow into the PPP (124). G-6-P enters the PPP pathway, which produces precursors for lipids, nucleotides, and NADPH. These molecules provide cells with the energy and substrates required for the creation of macromolecules, which promotes the growth of tumors (125). Two steps in the PPP are often reprogrammed in cancer. First, the oxidative phase, an irreversible step that involves the enzyme G6PD, which catalyzes the conversion of G6P to 6-phosphoglucose lactone. The step is considered rate-limiting and is crucial for generating NADPH, which is essential for counteracting oxidative stress. Second, the non-oxidative phase, the subsequent step involving the enzyme ribulose-5-phosphate epimerase, which converts ribulose-5-phosphate into xylulose-5-phosphate. This step is important for the generation of nucleotides and amino acids, which are often upregulated in cancer cells to support rapid cell proliferation (126).
Renal cancer cells express glucose-6-phosphate dehydrogenase (G6PD), the first and rate-limiting enzyme of the PPP, at high levels (127). Similar to G6PD, other oxidative branch NADPH-generating enzymes, such as 6-phosphogluconate dehydrogenase (6PGD), regulate PPP flux in renal cancer cells (93). Different phytochemicals, including caffeic acid, ellagic acid and physcion, directly downregulate the activity of G6PD and 6PGD in different types of cancer, including lung cancer, leukemia, and breast cancer (124,125,128). Limited research has been performed on the precise impact of flavonoids on the PPP in the metabolic reprogramming of RCC. Flavonoids may indirectly impact the PPP. The PPP is essential to sustain redox equilibrium and supply the building blocks for nucleotide synthesis (129,130). Although further research is required to understand the precise mechanism and clinical implications, the aforementioned information suggests that flavonoids may impact the course of RCC by modulating the PPP.
Fatty acid metabolism in RCC
One of the most prominent metabolic abnormalities found in cancer cells is defective lipid metabolism, which serves a major role in the development and metastasis of cancer cells (131). Dysregulated lipid de novo synthesis was observed in tumor cells in the 1950s, and this was found to be a critical metabolic state for cancer cells (132). Obesity is frequently linked to RCC, and patients with ccRCC have been found to exhibit greater levels of cholesterol ester accumulation in their kidneys (93). An outline of the metabolic pathways for fatty acids in ccRCC and how flavonoids alter them is shown in Fig. 4. Metabolomics research has revealed that ccRCC cells exhibit higher levels of fatty acyl-carnitines and carnitine than control cells (16). These differences were shown to be closely linked to the clinical features of patients with kidney cancer. Furthermore, RCC cells have a markedly compromised β-oxidation pathway, which may cause a greater accumulation of fatty acyl-carnitines (133). Since the formation of lipid droplets in the cytoplasm produces the characteristic clear cell phenotype, ccRCC is considered to be characterized by these droplets (134). Additionally, the accumulation of lipid droplets around the endoplasmic reticulum (ER) supports the ER integrity in ccRCC cells (135). It has been demonstrated that, compared with normal kidney cells, ccRCC cells exhibit downregulation of fatty acid oxidation enzymes, such as acetyl CoA carboxylase (ACC), fatty acid synthase (FAS), and stearoyl CoA desaturase (SCD) (136). NADPH oxidation is catalyzed by the β-ketoacyl reductase (KR) and enoyl reductase domains of the multifunctional enzyme FAS (93). EGCG can inhibit the enzymatic activity of FAS in vitro by competing with NADPH to bind the KR domain (135,137). Emodin flavonoids can also inhibit FAS activity in breast, liver, prostate, leukemia, and colon cancer (133). SCD1 is the enzyme that is responsible for lipid storage, which is highly expressed in ccRCC and serves an important role in growth and proliferation (138). Flavonoids, such as betulinic acid and platyphylloside, directly inhibit SCD1 in colon cancer (139). Flavonoids reduce the activity of SCD1, which subsequently inhibits RCC growth. FAS is also associated with RCC tumor growth aggressiveness and poor patient survival (140). The activity of FAS can be directly affected by flavonoids such as kaempferol, luteolin, morin, platyphylloside, quercetin, and resveratrol (131,133,135,137,141,142). HIFs are essential for the proliferation of RCC cells (91). Flavonoids such as oroxylin A, resveratrol, methylalpinumisoflavone, and EGCG can also regulate the activity of HIFs, which has been primarily observed in patients with breast cancer (133,143,144). Malonyl CoA is produced by ACC, a rate-limiting enzyme for fatty acid synthesis (145). Soraphen A is a polyketide that inhibits the activity of ACC1, which is upregulated in ccRCC (139). Additionally, quercetin reduces the production of triacylglycerol and fatty acids in rat hepatocytes and inhibits ACC without having any detectable influence on FAS (146). One of the distinctive features of kidney cancer is the reprogramming of the glycerophospholipids and arachidonic acid metabolism (147). The primary constituents of cell membranes are glycerophospholipids, which are also the sources of triacylglycerol, lysophosphatidic acid (LPA), and phosphatidic acid, which are the building blocks of lipid storage. Autotaxin, the enzyme responsible for producing LPA, is highly expressed in the endothelial cells surrounding the tumor, and its activity can be inhibited by several polyphenols (148). Arachidonic acid, a crucial compound in RCC derived from membrane phospholipids, is synthesized through pathways involving inflammatory enzymes such as cyclooxygenase (COX)-1, COX-2, and lipoxygenases (149). Different flavonoids have a potential effect on these inflammatory enzymes and have exhibited inhibitory effects on them. Quercetin, EGCG, and resveratrol have been reported to inhibit the activity of COX-1 and COX-2 in enzymatic assays (82,135,150). Compared with normal kidney cells, RCC cells express more of these enzymes (82). Numerous investigations have demonstrated that elevated COX-2 levels are linked to tumor size, stage, and grade in RCC. These findings imply that COX-2 may be a target in ccRCC (151,152). Sterol regulatory element binding protein-1 (SREBP-1) is a key regulator of lipid metabolism. Specifically, flavonoids may inhibit SREBP-1 activity, thereby reducing the expression of genes involved in lipogenesis and contributing to the reprogramming of metabolic pathways in ccRCC (153). Recognized antioxidants and anti-inflammatory flavonoids may affect important facets of RCC fatty acid metabolism. According to some research, flavonoids may alter lipid metabolism-related enzymes and pathways, which may have an impact on lipid synthesis, storage, and utilization in RCC cells. Targeting fatty acid metabolism with flavonoids offers a promising approach to understanding and creating treatment plans for RCC; however, further research is required to fully evaluate their effectiveness.
Amino acid metabolism in RCC
In the context of cancer cell metabolic reprogramming, amino acid metabolism is increasingly recognized for its critical role. Amino acids serve not only as substrates for protein synthesis but also act as essential mediators of redox homeostasis and support various biosynthetic pathways that are upregulated in kidney cells (154) (Fig. 5). Their significance is underscored by the rapid proliferation of renal cancer cells, which depend heavily on amino acids as both metabolites and metabolic regulators to promote growth (94). This underscores the potential of targeting amino acid metabolic pathways as a strategy to manage and inhibit cancer cell proliferation effectively in different ways mentioned below (155).
Glutamine metabolism
Glutamine is one of the primary nutrients that cancer cells use to preserve their biomass and bioenergetics. Furthermore, it serves as a component in the synthesis of lipids and proteins. In the renal cortex, glutamine is employed to keep the pH of the urinary system stable (156). Compared with that in normal kidney tissues, the use of glutamine in ccRCC is increased, and the GSH/GSH disulfide balance is strictly regulated (156). Increased levels of glutamine are linked to elevated levels of free fatty acids in RCC (93,156). Furthermore, in rapidly proliferating renal cancer cells, one of the predominant metabolic pathways is the reductive carboxylation of glutamine (156). In transgenic mouse models of human RCC, there was an increase in the amounts of glutamate and α-ketoglutarate, alongside upregulation of glutaminase (GLS), which serves a crucial role in glutamine metabolism and supports the metabolic needs of rapidly proliferating cancer cells (156). Based on assessment of the literature, it has been established that HIF expression serves a crucial role in triggering the reductive carboxylation of α-ketoglutarate in RCC cells. Additionally, the reversal of isocitrate dehydrogenase flux to the reductive carboxylation of glutamine to citrate has been predicted (93,156). GSH peroxidase 1 (GPX1) expression is upregulated in ccRCC cells. The activity of GLS and GPX1 can be controlled by the flavonoids present in different medicinal plants (93). Curcumin is a flavonoid that downregulates the activity of GLS in RCC (93). GLS is an important enzyme that converts glutamine into glutamate in the metabolic reprogramming of RCC, which can be directly inhibited by betulinic acid (150,157).
Serine and glycine metabolism
Serine can be obtained through extracellular absorption, with a portion of it derived from glucose metabolism (155). Serine and glycine are two amino acids that are connected during biosynthesis, and function as vital precursors for the formation of proteins, lipids and nucleic acid building blocks, all of which are essential for the growth of cancer (155). Under the catalysis of serine hydroxy methyltransferases, serine, whether synthesized de novo from 3-phosphoglycerate or imported from external sources, can be further converted into glycine (158). Threonine dehydrogenase and glycine C-acetyltransferase may also convert threonine into glycine (159). The creation of macromolecules, including lipids, proteins and nucleic acid, requires methyl groups for one-carbon metabolism, which glycine subsequently supplies (160). Serine is also involved in DNA methylation, an important step for the metabolic reprogramming of cancerous cells (161). Various synthetic drugs are currently in clinical trials targeting different metabolic pathways to reprogram renal cancer cells (159). However, synthetic drugs often come with side effects, which makes natural alternatives an appealing option (40). Plant-derived secondary metabolites, such as flavonoids, including quercetin, apigenin, morin and resveratrol, have shown efficacy in reprogramming cancer cells with potentially fewer side effects (162).
Arginine metabolism
Numerous solid tumor cells quickly succumb to growth media without arginine, which is an important amino acid (163). Additionally, it participates in several crucial cellular metabolic processes, including the urea cycle, the manufacture of nitric oxide, proline and glutamate, as well as nucleotide biosynthesis (16). Arginine can also be depleted by catalytically converting arginine to citrulline using the pegylated version of arginine deaminase (163). In addition to catalyzing the synthesis of argininosuccinate from citrulline and aspartic acid, argininosuccinate synthetase (ASS) is an enzyme that limits the pace at which arginine may be synthesized entirely from the beginning (163). Renal and parenchymal cells, which can recycle citrulline back to arginine, are examples of cells with normal expression levels of ASS1 (16). However, cells lacking ASS1, such as ccRCC cells, are unable to convert citrulline into arginine (16). Other enzymes, such as argininosuccinate lyase, catalyze the conversion of argininosuccinate to arginine and fumaric acid, which in turn connects arginine metabolism to the TCA cycle-generated energy metabolism of glucose (163). According to the literature, certain human malignancies, such as hepatocellular carcinoma and malignant melanoma, lack ASS, making them vulnerable to arginine deprivation treatment since they are unable to synthesize arginine (164). The activity of these enzymes may be influenced by the flavonoids present in different medicinal plants (165). During oncogenesis, cells often become dependent on external supplies of arginine due to the loss or absence of the enzyme ASS1. While ASS1 is normally expressed in proximal tubule cells, it is absent or not significantly expressed in ccRCC (16,163–165). In arginine metabolism, the activity of arginase and nitric oxide synthase serves vital roles in cancer cell proliferation and immune response (165). Researchers have investigated flavonoids such as quercetin and genistein for their potential effects on nitric oxide production, which is intricately linked to arginine metabolism (123,166). The specific impact of flavonoids on RCC and arginine metabolism requires further research.
Tryptophan
There are three main downstream pathways of tryptophan. The serotonin, indole acetate and kynurenine pathways are significant in understanding its metabolism (15,94). Indoleamine 2,3-deoxygenase (IDO) catabolizes the majority of tryptophan. Through the kynurenine pathways, it serves as a rate-limiting enzyme in this reaction. Tryptophan levels in ccRCC are lower compared with those in normal kidney cells, indicating higher consumption (167). ccRCC tissues exhibit elevated amounts of quinolinate and kynurenine (168). On the other hand, there is a decrease in the amounts of enzymes that support the kynurenine pathways, such as aldehyde dehydrogenase 2, monoamine oxidase and DOPA decarboxylase, in RCC, which is considered to be related to the serotonin and indole acetate pathways (169). These findings suggest the reduction of tryptophan/kynurenine through IDO enrichment in RCC (170). The approval of PD-1 inhibitors such as nivolumab as second-line treatments for RCC represents an advancement in immunotherapy. These inhibitors work by blocking the PD-1 pathways, which normally help to dampen the immune response and prevent autoimmunity. By inhibiting PD-1, these drugs enhance the activity of cytotoxic T cells against cancer cells, leading to increased tumor cell death. In addition to PD-1 inhibitors, adjuvant therapies that stimulate cytotoxic T-cell activity are employed to further boost the immune response (171). Tryptophan catabolism by IDO and other enzymes can suppress T-cell activity and contribute to immune evasion by tumor (172). These therapies help to optimize the effectiveness of PD-1 inhibitors by promoting a more robust and sustained immune attack on RCC cells (173). The activity of enzymatic pathways can be influenced by flavonoids such as morin, quercetin, EGCG, resveratrol and betulinic acid because tryptophan metabolism is involved in various cellular processes, including immune modulation and the production of metabolites, such as serotonin and kynurenine (174).
Toxicity associated with flavonoids
While it is commonly considered that using plant secondary metabolites as an alternative medication is safe or does not cause side effects (175), literature has also demonstrated that extended exposure to large dosages of some flavonoids can be potentially harmful (176). One flavonoid with anticancer properties is resveratrol. A study conducted in vivo suggested that consuming it at a higher dosage (3,000 mg/kg body weight) may be harmful to the kidneys (177). Turmeric, a commonly used traditional medicine in the Indian subcontinent, contains a complex mixture of chemical compounds. Among these, there are numerous substances that can be categorized based on their biological effects. Specifically, turmeric has been associated with 136 mutagenic, 153 carcinogenic, and 64 hepatotoxic compounds. Among the active ingredients with anticancer potential is curcumin, which exhibits dose-dependent hepatotoxicity (178). Overdosing and continuous usage of curcumin in rats produces ROS and proinflammatory cytokines. It also gradually reduces antioxidants, such as superoxide dismutase and glutathione S-transferase, which leads to liver damage (179). Genistein neutralizes the protective effect of letrozole, an aromatase inhibitor, against estrogen-dependent breast cancer (180). Although EGCG has numerous health advantages, research has demonstrated that a high dosage of the compound in mice can cause hepatotoxicity, which has been linked to the suppression of antioxidant enzymes (181). Both patients and doctors should be aware of the potential side effects of herbal medicines, as well as how they may interact with other prescription medications. Dietary supplements should be used cautiously, considering both the positive and negative consequences. Before being approved for use in the treatment of any disease, herbal drugs must undergo quality and pharmacological evaluations for toxicity.
Dietary bioactive compounds as chemopreventive agents
Considering the global increase in cancer cases, mortality, and treatment limits, cancer prevention must continue to be a focus for improved cancer management (182). Lifestyle changes can postpone the beginning of cancer, with nutrition serving a notable role (183). Additionally, consuming a variety of plant-based foods may help prevent cancer and aid in its treatment (182). Numerous plant-based bioactive compounds, including EGCG from green tea, lycopene from tomatoes, apigenin from parsley, curcumin from turmeric, resveratrol from grapes, genistein from soybeans, and gingerol from ginger, have anti-cancer properties and can be used as an easily accessible and affordable cancer prevention strategy (183,184). The American Cancer Society advises individuals, including patients with cancer, to include a variety of plant-based foods in their diet, as this can contribute to overall health and may lower the risk of developing various types of cancer. These include garlic, oranges, green tea, cereals, beans, soy-based food, peas, and other fruits and vegetables (185). The Food and Agriculture Organization and the World Health Organization jointly organized conferences in Japan in 2004 and 2021 titled ‘Fruits and Vegetables for Health’, which advocated for consuming 400 g of fruits and vegetables daily to lower the risk of various illnesses, including cancer (186).
Combination therapy with bioactive compounds against cancer
Numerous bioactive compounds such as resveratrol, quercetin and curcumin may have distal mechanisms of action to produce anticancer effects (187). Their combined utilization suggests promising treatment techniques. Numerous investigations have demonstrated that the combinations of chemicals, such as co-treatments with emodin and curcumin, might have a synergistic impact in preventing the proliferation and invasion of breast cancer cells (188–190). Additionally, the antitumor activities of paclitaxel against lung cancer are synergistically improved by emodin in vivo and in vitro (191). Cerulenin and emodin are inhibitors of FAS and have cumulative effects on FAS inhibition in colon cancer cells (133). As a result, combining two or more chemicals can result in stronger anticancer effects. However, this depends on the specific genes or metabolic pathways of each molecule and has to be validated by experiments.
Summary
The metabolic reprogramming observed in RCC exhibits heterogeneity, demonstrating varying metabolic preferences and patterns among different cancer types. Unlike molecular inhibitors and traditional chemotherapeutics, which mainly target a single metabolic pathway, flavonoids exhibit a unique mechanism of action by impacting several metabolic pathways to achieve their antitumor effects. The molecular mechanisms by which flavonoids control the reprogrammed metabolic pathways in RCC are described in the present review, with particular attention paid to how they target necessary metabolic rate-limiting enzymes such as HK2, FAS, LDHA and SREBP-1. Most flavonoids affect the metabolic pathways of glucose and fats, but some also affect the metabolic pathways of amino acids in RCC. For example, genistein inhibits both the intake of glucose and the process of glycolysis.
By interfering with metabolic pathways, some flavonoids not only directly lower RCC cell viability but also improve the antitumor effectiveness of traditional chemotherapeutics. For instance, shikonin activates the mitochondria to cause intracellular oxidative stress and inhibits the glycolytic process by lowering PKM2 activity. In contrast to traditional cancer treatments, such as radiation, chemotherapy, and surgery, which focus solely on the illness, the use of flavonoids is intended to enhance the defenses of the body against cancer by promoting the mobilization and regulation of all physiological systems. This holistic approach underscores the potential of flavonoids in directly impacting tumor metabolism but also synergistically enhancing the effectiveness of existing cancer treatments.
Flavonoid therapeutic approaches mainly target enhancing blood flow, supporting overall health, and strengthening the defenses of the body against disease. These approaches aim to provide systemic detoxification, reduce inflammation, and alleviate pain (192). For example, EGCG has the potential to improve the quality of life for patients with cancer by both preventing the growth of cancer and relieving the neuropathic pain caused by paclitaxel (193). Flavonoids exhibit marked advantages in impeding the growth and proliferation of cancer cells in vivo by modulating metabolic pathways. Several studies have validated the inhibitory effects of natural constituents in animal models (194,195); however, it is imperative to note that the quality of some studies requires further systematic investigation. Additionally, it is important to dispel the misconception that flavonoids are inherently gentler or less toxic than synthetic chemical drugs. While flavonoids are natural compounds with potential therapeutic benefits, they can still have adverse effects and interactions, and their safety profile must be carefully evaluated in clinical contexts, just as with synthetic drugs. Certain flavonoids exhibit hepatotoxicity or nephrotoxicity, potentially leading to irreversible impairments in patients (196).
According to the traditional theory of compatibility, toxic herbal medicines, including some flavonoids, could be combined with other appropriate conventional Chinese medicines to mitigate potential toxicity risks (178). Challenges persist in standardizing the production and quality control for flavonoids. Furthermore, evaluating the efficacy of flavonoids in interfering with reprogrammed cell metabolism in RCC necessitates randomized controlled clinical trials. Addressing these aspects is essential to advance the understanding of the therapeutic potential of flavonoids, and ensure their safe and effective application in the context of RCC treatment.
Flavonoids represent a promising class of compounds for the treatment of RCC due to their ability to target multiple metabolic pathways with lower toxicity compared with conventional chemotherapeutics (197). Future research should focus on exploring the synergistic effects of flavonoids with other therapeutic agents. Understanding the long-term outcomes of flavonoid-based interactions with specific metabolic targets is crucial for evaluating their efficacy and safety in cancer treatment. Comprehensive studies are needed to assess how these interactions affect metabolic pathways over extended periods and determine their potential impact on overall patient health and treatment outcomes. These studies could pave the way for the development of more effective and personalized treatment strategies for RCC.
Conclusion
Flavonoids hold significant promise as anticancer agents due to their ability to modulate various pathways involved in cancer metabolism. Their natural origin and multifunctional properties make them appealing alternatives to synthetic drugs. Flavonoids exhibit lower toxicity and fewer side effects compared with conventional chemotherapy agents, enhancing patient compliance and quality of life (197). This makes them preferable to other anticancer drugs targeting metabolic reprogramming. Furthermore, the ability of flavonoids to selectively inhibit cancer cell proliferation, induce apoptosis, and prevent metastasis underscores their potential as more effective anticancer agents compared with other phytochemicals and synthetic drugs (197).
Because of the extensive reprogramming of metabolic pathways, extracellular stress and the immune system, renal cancer is frequently recognized as a metabolic disorder. By improving tumor imaging and identifying novel therapeutic targets, all of these programmed metabolic pathways may be used to create a more successful RCC treatment. By controlling the metabolic reprogramming of cancer cells, flavonoids have effective inhibitory effects on tumor cells and may also improve the sensitization of cancer cells to chemotherapy treatments. Due to poly-pharmacological actions, flavonoids may be able to reduce cancer pain and enhance the quality of life of patients with cancer. Flavonoids counteract metabolic reprogramming in RCC through diverse mechanisms, including inhibition of key metabolic pathways, modulation of glucose transporters and reduction of oxidative stress. By targeting multiple signaling pathways and metabolic processes, flavonoids may disrupt the survival advantage conferred by the altered metabolism of cancer cells.
Additionally, their low toxicity profile and accessibility from natural sources make them attractive drug candidates for further clinical investigation. Integrating flavonoid-based interventions into comprehensive therapeutic strategies may offer novel avenues to improve the prognosis and treatment outcomes of individuals with RCC. Continued research in this field is essential to advance the understanding of the molecular mechanisms involved and ultimately translate these findings into effective clinical applications.
Supplementary Material
Supporting Data
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant nos. 82460510, 82203565, 82103388, and 31960145), Yunnan Province Applied Research Funds (grant nos. 202201AY070001-011, 202201AY070001-043 and 202201AS070077), and Science and Technology Innovation Team of Tumor Metabolism Research, Kunming Medical University (grant no. CXTD202102).
Availability of data and materials
Not applicable.
Authors' contributions
QZ and ZY conceived the study, supervised the research and contributed to the critical revision of the manuscript. AS conceived the study, collected data, wrote the original draft, and reviewed and edited the manuscript. WL, BS and YZ provided resources, contributed to drafting the manuscript, prepared the tables and figures, and reviewed and edited the manuscript. XL, YS, JX and KC provided resources, contributed to the critical revision of the manuscript, and reviewed and edited the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
RCC |
renal cell carcinoma |
ccRCC |
clear cell renal cell carcinoma |
GLUT |
glucose transporter |
ECG |
epicatechin gallate |
EGCG |
epigallocatechin-3-gallate |
TCA |
tricarboxylic acid |
LDH |
lactate dehydrogenase |
LDHA |
LDH A |
PKM2 |
pyruvate kinase M2 |
HK |
hexokinase |
PDH |
pyruvate dehydrogenase |
ETC |
electron transport chain |
PPP |
pentose phosphate pathway |
6PGD |
6-phosphogluconate dehydrogenase |
ACC |
acetyl CoA carboxylase |
FAS |
fatty acid synthase |
SCD |
stearoyl CoA desaturase |
LPA |
lysophosphatidic acid |
COX |
cyclooxygenase |
GLS |
glutaminase |
GPX1 |
glutathione peroxidase 1 |
References
Rini BI, Campbell SC and Escudier B: Renal cell carcinoma. Lancet. 373:1119–1132. 2009. View Article : Google Scholar : PubMed/NCBI | |
Ferlay J, Bray F, Pisani P and Parkin D: Globocan 2002: Cancer incidence, mortality and prevalence worldwide. IARC Cancerbase. 52004. | |
Parkin DM and Bray F: International patterns of cancer incidence and mortality. Cancer Epidemiol Prevention. 101–138. 2006. View Article : Google Scholar | |
Mohammadian M, Pakzad R, Towhidi F, Makhsosi BR, Ahmadi A and Salehiniya H: Incidence and mortality of kidney cancer and its relationship with HDI (Human Development Index) in the world in 2012. Clujul Med. 90:2862017.PubMed/NCBI | |
Lobo J, Ohashi R, Amin MB, Berney DM, Compérat EM, Cree IA, Gill AJ, Hartmann A, Menon S, Netto GJ, et al: Who 2022 landscape of papillary and chromophobe renal cell carcinoma. Histopathology. 81:426–438. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hoerner CR, Miao SY, Hsieh JJ and Fan AC: Targeting metabolic pathways in kidney cancer: Rationale and therapeutic opportunities. Cancer J. 26:407–418. 2020. View Article : Google Scholar : PubMed/NCBI | |
Barron CC, Bilan PJ, Tsakiridis T and Tsiani E: Facilitative glucose transporters: implications for cancer detection, prognosis and treatment. Metabolism. 65:124–139. 2016. View Article : Google Scholar : PubMed/NCBI | |
Furuta E, Okuda H, Kobayashi A and Watabe K: Metabolic genes in cancer: Their roles in tumor progression and clinical implications. Biochim Biophys Acta. 1805:141–152. 2010.PubMed/NCBI | |
Menendez JA and Lupu R: Fatty acid synthase (FASN) as a therapeutic target in breast cancer. Expert Opin Ther Targets. 21:1001–1016. 2017. View Article : Google Scholar : PubMed/NCBI | |
Vander Heiden MG, Cantley LC and Thompson CB: Understanding the warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI | |
DeBerardinis RJ, Lum JJ, Hatzivassiliou G and Thompson CB: The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7:11–20. 2008. View Article : Google Scholar : PubMed/NCBI | |
Cai L and Tu BP: Driving the cell cycle through metabolism. Annu Rev Cell Dev Biol. 28:59–87. 2012. View Article : Google Scholar : PubMed/NCBI | |
Rathmell WK, Rathmell JC and Linehan WM: Metabolic pathways in kidney cancer: Current therapies and future directions. J Clin Oncol. JCO2018792309. 2018.doi: 10.1200/JCO.2018.79.2309 (Epub ahead of print). View Article : Google Scholar | |
Weiss RH: Metabolomics and metabolic reprogramming in kidney cancer. Semin Nephrol. 38:175–182. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wettersten HI: Reprogramming of metabolism in kidney cancer. Semin Nephrol. 40:2–13. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wettersten HI, Aboud OA, Lara PN Jr and Weiss RH: Metabolic reprogramming in clear cell renal cell carcinoma. Nat Rev Nephrol. 13:410–419. 2017. View Article : Google Scholar : PubMed/NCBI | |
Linehan WM, Srinivasan R and Schmidt LS: The genetic basis of kidney cancer: A metabolic disease. Nat Rev Urol. 7:277–285. 2010. View Article : Google Scholar : PubMed/NCBI | |
De Marinis F, Rinaldi M, Ardizzoni A, Bruzzi P, Pennucci MC, Portalone L, D'Aprile M, Ripanti P, Romano F, Belli M, et al: The role of vindesine and lonidamine in the treatment of elderly patients with advanced non-small cell lung cancer: A phase III randomized FONICAP trial. Italian Lung Cancer Task Force. Tumori. 85:177–182. 1999. View Article : Google Scholar : PubMed/NCBI | |
Acharya N and Singh KP: Recent advances in the molecular basis of chemotherapy resistance and potential application of epigenetic therapeutics in chemorefractory renal cell carcinoma. WIREs Mech Dis. 14:e15752022. View Article : Google Scholar : PubMed/NCBI | |
Hussain SA, Sulaiman AA, Balch C, Chauhan H, Alhadidi QM and Tiwari AK: Natural polyphenols in cancer chemoresistance. Nutr Cancer. 68:879–891. 2016. View Article : Google Scholar : PubMed/NCBI | |
de Luna FCF, Ferreira WAS, Casseb SMM and de Oliveira EHC: Anticancer potential of flavonoids: An overview with an emphasis on tangeretin. Pharmaceuticals (Basel). 16:12292023. View Article : Google Scholar : PubMed/NCBI | |
Kumar A and Jaitak V: Natural products as multidrug resistance modulators in cancer. Eur J Med Chem. 176:268–291. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chrun ES, Modolo F and Daniel FI: Histone modifications: A review about the presence of this epigenetic phenomenon in carcinogenesis. Pathol Res Pract. 213:1329–1339. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kopustinskiene DM, Jakstas V, Savickas A and Bernatoniene J: Flavonoids as anticancer agents. Nutrients. 12:4572020. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar : PubMed/NCBI | |
Slika H, Mansour H, Wehbe N, Nasser SA, Iratni R, Nasrallah G, Shaito A, Ghaddar T, Kobeissy F and Eid AH: Therapeutic potential of flavonoids in cancer: ROS-mediated mechanisms. Biomed Pharmacother. 146:1124422022. View Article : Google Scholar : PubMed/NCBI | |
Garcia-Oliveira P, Otero P, Pereira AG, Chamorro F, Carpena M, Echave J, Fraga-Corral M, Simal-Gandara J and Prieto MA: Status and challenges of Plant-anticancer compounds in cancer treatment. Pharmaceuticals (Basel). 14:1572021. View Article : Google Scholar : PubMed/NCBI | |
Liu Y: Inhibitors of basal glucose transport and their anticancer activities and mechanism. Ohio University; 2012 | |
Chan DA, Sutphin PD, Nguyen P, Turcotte S, Lai EW, Banh A, Reynolds GE, Chi JT, Wu J, Solow-Cordero DE, et al: Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci Transl Med. 3:94ra702011. View Article : Google Scholar : PubMed/NCBI | |
Kodama M and Nakayama KI: A second warburg-like effect in cancer metabolism: The metabolic shift of glutamine-derived nitrogen: A shift in glutamine-derived nitrogen metabolism from glutaminolysis to de novo nucleotide biosynthesis contributes to malignant evolution of cancer. Bioessays. 42:20001692020. View Article : Google Scholar | |
Sharma RA, Gescher AJ and Steward WP: Curcumin: The story so far. Eur J Cancer. 41:1955–1968. 2005. View Article : Google Scholar : PubMed/NCBI | |
Pavan AR, Silva GD, Jornada DH, Chiba DE, Fernandes GF, Man Chin C and Dos Santos JL: Unraveling the anticancer effect of curcumin and resveratrol. Nutrients. 8:6282016. View Article : Google Scholar : PubMed/NCBI | |
Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J and Ma X: Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (2020). 4:e2182023. View Article : Google Scholar : PubMed/NCBI | |
Alamgir A and Alamgir A: Drugs: Their natural, synthetic, and biosynthetic sources. Therapeutic Use of Medicinal Plants and Their Extracts: Volume 1 = Pharmacognosy. 105–123. 2017. View Article : Google Scholar | |
Yuan H, Ma Q, Ye L and Piao G: The traditional medicine and modern medicine from natural products. Molecules. 21:5592016. View Article : Google Scholar : PubMed/NCBI | |
Atanasov AG, Zotchev SB, Dirsch VM; International Natural Product Sciences Taskforce, ; Supuran CT: Natural products in drug discovery: Advances and opportunities. Nat Rev Drug Discov. 20:200–216. 2021. View Article : Google Scholar : PubMed/NCBI | |
Fabiani R: Antitumoral properties of natural products. Molecules. 25:6502020. View Article : Google Scholar : PubMed/NCBI | |
Lichota A and Gwozdzinski K: Anticancer activity of natural compounds from plant and marine environment. Int J Mol Sci. 19:35332018. View Article : Google Scholar : PubMed/NCBI | |
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W and Zheng Q: Natural products as anticancer agents: Current status and future perspectives. Molecules. 27:83672022. View Article : Google Scholar : PubMed/NCBI | |
Karimi A, Majlesi M and Rafieian-Kopaei M: Herbal versus synthetic drugs; beliefs and facts. J Nephropharmacol. 4:27–30. 2015.PubMed/NCBI | |
Anand P, Kunnumakkara AB, Newman RA and Aggarwal BB: Bioavailability of curcumin: Problems and promises. Mol Pharm. 4:807–818. 2007. View Article : Google Scholar : PubMed/NCBI | |
Jiang X, Jiang Z, Jiang M and Sun Y: Berberine as a potential agent for the treatment of colorectal cancer. Front Med (Lausanne). 9:8869962022. View Article : Google Scholar : PubMed/NCBI | |
Yang CS and Wang X: Green tea and cancer prevention. Nutr Cancer. 62:931–937. 2010. View Article : Google Scholar : PubMed/NCBI | |
Bosetti C, Rossi M, McLaughlin JK, Negri E, Talamini R, Lagiou P, Montella M, Ramazzotti V, Franceschi S and LaVecchia C: Flavonoids and the risk of renal cell carcinoma. Cancer Epidemiol Biomarkers Prev. 16:98–101. 2007. View Article : Google Scholar : PubMed/NCBI | |
Silva A, Silva V, Igrejas G, Aires A, Falco V, Valentão P and Poeta P: Phenolic compounds classification and their distribution in winemaking by-products. Eur Food Res Technol. 249:207–239. 2023. View Article : Google Scholar | |
Razi SM and Rashidinejad A: Bioactive compounds: Chemistry, structure, and functionality. In: Spray drying encapsulation of bioactive materials; CRC Press: pp. 1–46. 2021 | |
Badshah SL, Faisal S, Muhammad A, Poulson BG, Emwas AH and Jaremko M: Antiviral activities of flavonoids. Biomed Pharmacother. 140:1115962021. View Article : Google Scholar : PubMed/NCBI | |
Al Aboody MS and Mickymaray S: Anti-fungal efficacy and mechanisms of flavonoids. Antibiotics (Basel). 9:452020. View Article : Google Scholar | |
Xie Y, Yang W, Tang F, Chen X and Ren L: Antibacterial activities of flavonoids: Structure-activity relationship and mechanism. Curr Med Chem. 22:132–149. 2015. View Article : Google Scholar : PubMed/NCBI | |
Heim KE, Tagliaferro AR and Bobilya DJ: Flavonoid antioxidants: Chemistry, metabolism and structure-activity relationships. J Nutr Biochem. 13:572–584. 2002. View Article : Google Scholar : PubMed/NCBI | |
Rathee P, Chaudhary H, Rathee S, Rathee D, Kumar V and Kohli K: Mechanism of action of flavonoids as anti-inflammatory agents: A review. Inflamm Allergy Drug Targets. 8:229–235. 2009. View Article : Google Scholar : PubMed/NCBI | |
Al-Ishaq RK, Abotaleb M, Kubatka P, Kajo K and Büsselberg D: Flavonoids and their Anti-diabetic effects: Cellular mechanisms and effects to improve blood sugar levels. Biomolecules. 9:4302019. View Article : Google Scholar : PubMed/NCBI | |
Snijman PW, Swanevelder S, Joubert E, Green IR and Gelderblom WC: The antimutagenic activity of the major flavonoids of rooibos (Aspalathus linearis): Some dose-response effects on mutagen activation-flavonoid interactions. Mutat Res. 631:111–123. 2007. View Article : Google Scholar : PubMed/NCBI | |
Oliveira AKS, de Oliveira E Silva AM, Pereira RO, Santos AS, Barbosa Junior EV, Bezerra MT, Barreto RSS, Quintans-Junior LJ and Quintans JSS: Anti-obesity properties and mechanism of action of flavonoids: A review. Crit Rev Food Sci Nutr. 62:7827–7848. 2022. View Article : Google Scholar : PubMed/NCBI | |
Luo Y, Shang P and Li D: Luteolin: A flavonoid that has multiple Cardio-protective effects and its molecular mechanisms. Front Pharmacol. 8:6922017. View Article : Google Scholar : PubMed/NCBI | |
Trachootham D, Alexandre J and Huang P: Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat Rev Drug Discov. 8:579–591. 2009. View Article : Google Scholar : PubMed/NCBI | |
Gorrini C, Harris IS and Mak TW: Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov. 12:931–947. 2013. View Article : Google Scholar : PubMed/NCBI | |
Huang MZ and Li JY: Physiological regulation of reactive oxygen species in organisms based on their physicochemical properties. Acta Physiol (Oxf). 228:e133512020. View Article : Google Scholar : PubMed/NCBI | |
Cao G, Sofic E and Prior RL: Antioxidant and prooxidant behavior of flavonoids: Structure-activity relationships. Free Radic Biol Med. 22:749–760. 1997. View Article : Google Scholar : PubMed/NCBI | |
Havsteen BH: The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 96:67–202. 2002. View Article : Google Scholar : PubMed/NCBI | |
Tavsan Z and Kayali HA: Flavonoids showed anticancer effects on the ovarian cancer cells: Involvement of reactive oxygen species, apoptosis, cell cycle and invasion. Biomed Pharmacother. 116:1090042019. View Article : Google Scholar : PubMed/NCBI | |
Biswas P, Dey D, Biswas PK, Rahaman TI, Saha S, Parvez A, Khan DA, Lily NJ, Saha K, Sohel M, et al: A comprehensive analysis and Anti-cancer activities of quercetin in ROS-mediated cancer and cancer stem cells. Int J Mol Sci. 23:117462022. View Article : Google Scholar : PubMed/NCBI | |
Reyes-Farias M and Carrasco-Pozo C: The anti-cancer effect of quercetin: Molecular implications in cancer metabolism. Int J Mol Sci. 20:31772019. View Article : Google Scholar : PubMed/NCBI | |
Bansal A and Simon MC: Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 217:22912018. View Article : Google Scholar : PubMed/NCBI | |
Desideri E, Ciccarone F and Ciriolo MR: Targeting glutathione metabolism: Partner in crime in anticancer therapy. Nutrients. 11:19262019. View Article : Google Scholar : PubMed/NCBI | |
Bansal A and Simon MC: Glutathione metabolism in cancer progression and treatment resistance. J Cell Biol. 217:2291–2298. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yoo D, Jung E, Noh J, Hyun H, Seon S, Hong S, Kim D and Lee D: Glutathione-depleting Pro-oxidant as a selective anticancer therapeutic agent. ACS Omega. 4:10070–10077. 2019. View Article : Google Scholar : PubMed/NCBI | |
Traverso N, Ricciarelli R, Nitti M, Marengo B, Furfaro AL, Pronzato MA, Marinari UM and Domenicotti C: Role of glutathione in cancer progression and chemoresistance. Oxid Med Cell Longev. 2013:9729132013. View Article : Google Scholar : PubMed/NCBI | |
Wu JH and Batist G: Glutathione and glutathione analogues; therapeutic potentials. Biochim Biophys Acta. 1830:3350–3353. 2013. View Article : Google Scholar : PubMed/NCBI | |
Sobhakumari A: Dual role of oxidative stress in head and neck cancer chemotherapy: Cytotoxicity and pro-survival autophagy. The University of Iowa; 2013, View Article : Google Scholar | |
Liang F, Fang Y, Cao W, Zhang Z, Pan S and Xu X: Attenuation of tert-Butyl Hydroperoxide (t-BHP)-induced oxidative damage in HepG2 cells by tangeretin: Relevance of the Nrf2-ARE and MAPK signaling pathways. J Agric Food Chem. 66:6317–6325. 2018. View Article : Google Scholar : PubMed/NCBI | |
Fatima N, Baqri SSR, Bhattacharya A, Koney NK-K, Husain K, Abbas A and Ansari RA: Role of flavonoids as epigenetic modulators in cancer prevention and therapy. Front Genet. 12:7587332021. View Article : Google Scholar : PubMed/NCBI | |
Ponte LGS, Pavan ICB, Mancini MCS, da Silva LGS, Morelli AP, Severino MB, Bezerra RMN and Simabuco FM: The hallmarks of flavonoids in cancer. Molecules. 26:20292021. View Article : Google Scholar : PubMed/NCBI | |
Seo HS, Ku JM, Choi HS, Choi YK, Woo JK, Kim M, Kim I, Na CH, Hur H, Jang BH, et al: Quercetin induces Caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol Rep. 36:31–42. 2016. View Article : Google Scholar : PubMed/NCBI | |
Akhtar MF, Saleem A, Rasul A, Baig MM, Bin-Jumah M and Daim MM: Anticancer natural medicines: An overview of cell signaling and other targets of anticancer phytochemicals. Eur J Pharmacol. 888:1734882020. View Article : Google Scholar : PubMed/NCBI | |
Hung PF, Wu BT, Chen HC, Chen YH, Chen CL, Wu MH, Liu HC, Lee MJ and Kao YH: Antimitogenic effect of green tea (−)-epigallocatechin gallate on 3T3-L1 preadipocytes depends on the ERK and Cdk2 pathways. Am J Physiol Cell Physiol. 288:C1094–C108. 2005. View Article : Google Scholar : PubMed/NCBI | |
Han DW, Lee MH, Kim HH, Hyon SH and Park JC: Epigallocatechin-3-gallate regulates cell growth, cell cycle and phosphorylated nuclear factor-κB in human dermal fibroblasts. Acta Pharmacol Sin. 32:637–646. 2011. View Article : Google Scholar : PubMed/NCBI | |
Shih LJ, Hsu PC, Chuu CP, Shui HA, Yeh CC, Chen YC and Kao YH: Epigallocatechin-3-gallate synergistically enhanced arecoline-induced cytotoxicity by redirecting cycle arrest to apoptosis. Curr Issues Mol Biol. 46:1516–1529. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Hu Z, Horta CA and Yang J: Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol. 88:46–66. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mendes LF, Gaspar VM, Conde TA, Mano JF and Duarte IF: Flavonoid-mediated immunomodulation of human macrophages involves key metabolites and metabolic pathways. Sci Rep. 9:149062019. View Article : Google Scholar : PubMed/NCBI | |
Usuwanthim K, Wisitpongpun P and Luetragoon T: Molecular identification of phytochemical for anticancer treatment. Anticancer Agents Med Chem. 20:651–666. 2020. View Article : Google Scholar : PubMed/NCBI | |
Hay N: Reprogramming glucose metabolism in cancer: Can it be exploited for cancer therapy? Nat Rev Cancer. 16:635–649. 2016. View Article : Google Scholar : PubMed/NCBI | |
Triplitt CL: Understanding the kidneys' role in blood glucose regulation. Am J Manag Care. 18 (1 Suppl):S11–S16. 2012.PubMed/NCBI | |
Boroughs LK and DeBerardinis RJ: Metabolic pathways promoting cancer cell survival and growth. Nat Cell Biol. 17:351–359. 2015. View Article : Google Scholar : PubMed/NCBI | |
Morani F, Phadngam S, Follo C, Titone R, Aimaretti G, Galetto A, Alabiso O and Isidoro C: PTEN regulates plasma membrane expression of glucose transporter 1 and glucose uptake in thyroid cancer cells. J Mol Endocrinol. 53:247–258. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wei R, Mao L, Xu P, Zheng X, Hackman RM, Mackenzie GG and Wang Y: Suppressing glucose metabolism with epigallocatechin-3-gallate (EGCG) reduces breast cancer cell growth in preclinical models. Food Funct. 9:5682–5696. 2018. View Article : Google Scholar : PubMed/NCBI | |
Moreira L, Araújo I, Costa T, Correia-Branco A, Faria A, Martel F and Keating E: Quercetin and epigallocatechin gallate inhibit glucose uptake and metabolism by breast cancer cells by an estrogen receptor-independent mechanism. Exp Cell Res. 319:1784–1795. 2013. View Article : Google Scholar : PubMed/NCBI | |
Prpa E: A mechanistic investigation into the acute effects of apple polyphenols on carbohydrate digestion and absorption. King's College London; 2021 | |
Pérez A, Ojeda P, Ojeda L, Salas Mn, Rivas CI, Vera JC and Reyes AM: Hexose transporter GLUT1 harbors several distinct regulatory binding sites for flavones and tyrphostins. Biochemistry. 50:8834–8845. 2011. View Article : Google Scholar : PubMed/NCBI | |
Patra S, Pradhan B, Nayak R, Behera C, Rout L, Jena M, Efferth T and Bhutia SK: Chemotherapeutic efficacy of curcumin and resveratrol against cancer: Chemoprevention, chemoprotection, drug synergism and clinical pharmacokinetics. Semin Cancer Biol. 73:310–320. 2021. View Article : Google Scholar : PubMed/NCBI | |
Samec M, Liskova A, Koklesova L, Mersakova S, Strnadel J, Kajo K, Pec M, Zhai K, Smejkal K, Mirzaei S, et al: Flavonoids targeting HIF-1: Implications on cancer metabolism. Cancers. 13:1302021. View Article : Google Scholar : PubMed/NCBI | |
Zambrano A, Molt M, Uribe E and Salas M: Glut 1 in cancer cells and the inhibitory action of resveratrol as a potential therapeutic strategy. Int J Mol Sci. 20:33742019. View Article : Google Scholar : PubMed/NCBI | |
Hakimi AA, Reznik E, Lee CH, Creighton CJ, Brannon AR, Luna A, Aksoy BA, Liu EM, Shen R, Lee W, et al: An integrated metabolic atlas of clear cell renal cell carcinoma. Cancer Cell. 29:104–116. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chakraborty S, Balan M, Sabarwal A, Choueiri TK and Pal S: Metabolic reprogramming in renal cancer: Events of a metabolic disease. Biochim Biophys Acta Rev Cancer. 1876:1885592021. View Article : Google Scholar : PubMed/NCBI | |
Shan S, Shi J, Yang P, Jia B, Wu H, Zhang X and Li Z: Apigenin restrains colon cancer cell proliferation via targeted blocking of pyruvate kinase M2-dependent glycolysis. J Agric Food Chem. 65:8136–8144. 2017. View Article : Google Scholar : PubMed/NCBI | |
Monteiro F and Shetty SS: Natural antioxidants as inhibitors of pyruvate kinase M2 in warburg phenotypes. J Herbal Med. 42:1007502023. View Article : Google Scholar | |
Feng J, Wu L, Ji J, Chen K, Yu Q, Zhang J, Chen J, Mao Y, Wang F, Dai W, et al: PKM2 is the target of proanthocyanidin B2 during the inhibition of hepatocellular carcinoma. J Exp Clin Cancer Res. 38:2042019. View Article : Google Scholar : PubMed/NCBI | |
Liu W, Li W, Liu H and Yu X: Xanthohumol inhibits colorectal cancer cells via downregulation of hexokinases II-mediated glycolysis. Int J Biol Sci. 15:24972019. View Article : Google Scholar : PubMed/NCBI | |
Wu H, Pan L, Gao C, Xu H, Li Y, Zhang L, Ma L, Meng L, Sun X and Qin H: Quercetin inhibits the proliferation of Glycolysis-Addicted HCC cells by reducing hexokinase 2 and Akt-mTOR pathway. Molecules. 24:19932019. View Article : Google Scholar : PubMed/NCBI | |
Deng X, Liu R, Li J, Li Z, Liu J, Xiong R, Lei X, Zheng X, Xie Z and Tang G: Design, synthesis, and preliminary biological evaluation of 3′,4′,5′-trimethoxy flavonoid salicylate derivatives as potential anti-tumor agents. N J Chemistry. 43:1874–1884. 2019. View Article : Google Scholar | |
Guo Y, Wei L, Zhou Y, Lu N, Tang X, Li Z and Wang X: Flavonoid Gl-V9 induces apoptosis and inhibits glycolysis of breast cancer via disrupting GSK-3β-modulated mitochondrial binding of HKII. Free Radic Biol Med. 146:119–129. 2020. View Article : Google Scholar : PubMed/NCBI | |
Mazlaghaninia M, Atri MS and Seyedalipour B: Scopoletin and morin inhibit lactate dehydrogenase enzyme activity, which is critical for cancer metabolism. Hormozgan Med J. 23:e882692019. View Article : Google Scholar | |
Jia L, Huang S, Yin X, Zan Y, Guo Y and Han L: Quercetin suppresses the mobility of breast cancer by suppressing glycolysis through akt-mtor pathway mediated autophagy induction. Life Sci. 208:123–130. 2018. View Article : Google Scholar : PubMed/NCBI | |
Bader A, Tuccinardi T, Granchi C, Martinelli A, Macchia M, Minutolo F, De Tommasi N and Braca A: Phenylpropanoids and flavonoids from phlomis kurdica as inhibitors of human lactate dehydrogenase. Phytochemistry. 116:262–268. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li S, Wu L, Feng J, Li J, Liu T, Zhang R, Xu S, Cheng K, Zhou Y, Zhou S, et al: In vitro and in vivo study of epigallocatechin-3-gallate-induced apoptosis in aerobic glycolytic hepatocellular carcinoma cells involving inhibition of phosphofructokinase activity. Sci Rep. 6:284792016. View Article : Google Scholar : PubMed/NCBI | |
Dihal AA, van der Woude H, Hendriksen PJ, Charif H, Dekker LJ, IJsselstijn L, de Boer VC, Alink GM, Burgers PC, Rietjens IM, et al: Transcriptome and proteome profiling of colon mucosa from quercetin fed F344 rats point to tumor preventive mechanisms, increased mitochondrial fatty acid degradation and decreased glycolysis. Proteomics. 8:45–61. 2008. View Article : Google Scholar : PubMed/NCBI | |
Jiménez-Uribe AP, Hernández-Cruz EY, Ramírez-Magaña KJ and Pedraza-Chaverri J: Involvement of tricarboxylic acid cycle metabolites in kidney diseases. Biomolecules. 11:12592021. View Article : Google Scholar : PubMed/NCBI | |
Liu JJ, Liu S, Gurung RL, Ching J, Kovalik J-P, Tan TY and Lim SC: Urine tricarboxylic acid cycle metabolites predict progressive chronic kidney disease in type 2 diabetes. J Clin Endocrinol Metab. 103:4357–4364. 2018. View Article : Google Scholar : PubMed/NCBI | |
Saunier E, Benelli C and Bortoli S: The pyruvate dehydrogenase complex in cancer: An old metabolic gatekeeper regulated by new pathways and pharmacological agents. Int J Cancer. 138:809–817. 2016. View Article : Google Scholar : PubMed/NCBI | |
Blouin JM, Penot G, Collinet M, Nacfer M, Forest C, Laurent-Puig P, Coumoul X, Barouki R, Benelli C and Bortoli S: Butyrate elicits a metabolic switch in human colon cancer cells by targeting the pyruvate dehydrogenase complex. Int J Cancer. 128:2591–2601. 2011. View Article : Google Scholar : PubMed/NCBI | |
Coricovac D, Dehelean CA, Pinzaru I, Mioc A, Aburel OM, Macasoi I, Draghici GA, Petean C, Soica C, Boruga M, et al: Assessment of betulinic acid cytotoxicity and mitochondrial metabolism impairment in a human melanoma cell line. Int J Mol Sci. 22:48702021. View Article : Google Scholar : PubMed/NCBI | |
Peeters TH, Lenting K, Breukels V, van Lith SA, van den Heuvel CN, Molenaar R, van Rooij A, Wevers R, Span PN, Heerschap A and Leenders WPJ: Isocitrate dehydrogenase 1-mutated cancers are sensitive to the green tea polyphenol epigallocatechin-3-gallate. Cancer Metab. 7:42019. View Article : Google Scholar : PubMed/NCBI | |
Bianchi G, Ravera S, Traverso C, Amaro A, Piaggio F, Emionite L, Bachetti T, Pfeffer U and Raffaghello L: Curcumin induces a fatal energetic impairment in tumor cells in vitro and in vivo by inhibiting ATP-synthase activity. Carcinogenesis. 39:1141–1150. 2018. View Article : Google Scholar : PubMed/NCBI | |
Patel K, Singh GK and Patel DK: A review on pharmacological and analytical aspects of naringenin. Chi J Integr Med. 24:551–560. 2018. View Article : Google Scholar : PubMed/NCBI | |
Abotaleb M, Samuel SM, Varghese E, Varghese S, Kubatka P, Liskova A and Büsselberg D: Flavonoids in cancer and apoptosis. Cancers (Basel). 11:282018. View Article : Google Scholar : PubMed/NCBI | |
Brecht K, Riebel V, Couttet P, Paech F, Wolf A, Chibout SD, Pognan F, Krähenbühl S and Uteng M: Mechanistic insights into selective killing of oxphos-dependent cancer cells by arctigenin. Toxicol In Vitro. 40:55–65. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wang T, Ma F and Qian HL: Defueling the cancer: ATP synthase as an emerging target in cancer therapy. Mol Ther Oncolytics. 23:82–95. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kicinska A and Jarmuszkiewicz W: Flavonoids and mitochondria: Activation of cytoprotective pathways? Molecules. 25:30602020. View Article : Google Scholar : PubMed/NCBI | |
Zheng KY, Choi RC, Cheung AW, Guo AJ, Bi CW, Zhu KY, Fu Q, Du Y, Zhang WL, Zhan JY, et al: Flavonoids from radix astragali induce the expression of erythropoietin in cultured cells: A signaling mediated via the accumulation of hypoxia-inducible factor-1α. J Agric Food Chem. 59:1697–1704. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wang R, Zhou S and Li S: Cancer therapeutic agents targeting hypoxia-inducible factor-1. Curr Med Chem. 18:3168–3189. 2011. View Article : Google Scholar : PubMed/NCBI | |
Roy M and Datta A: Cancer genetics and therapeutics: Focus on phytochemicals. Springer Nature; 2019, View Article : Google Scholar | |
Kittiratphatthana N, Kukongviriyapan V, Prawan A and Senggunprai L: Luteolin induces cholangiocarcinoma cell apoptosis through the mitochondrial-dependent pathway mediated by reactive oxygen species. J Pharmacy Pharmacol. 68:1184–1192. 2016. View Article : Google Scholar : PubMed/NCBI | |
Nenkov M, Ma Y, Gaßler N and Chen Y: Metabolic reprogramming of colorectal cancer cells and the microenvironment: Implication for therapy. Int J Mol Sci. 22:62622021. View Article : Google Scholar : PubMed/NCBI | |
Adem S, Comakli V, Kuzu M and Demirdag R: Investigation of the effects of some phenolic compounds on the activities of glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase from human erythrocytes. J Biochem Mol Toxicol. 28:510–514. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gomez LS, Zancan P, Marcondes MC, Ramos-Santos L, Meyer-Fernandes JR, Sola-Penna M and Da Silva D: Resveratrol decreases breast cancer cell viability and glucose metabolism by inhibiting 6-phosphofructo-1-kinase. Biochimie. 95:1336–1343. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jiang P, Du W and Wu M: Regulation of the pentose phosphate pathway in cancer. Protein Cell. 5:592–602. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Zhang Z, Zhu Y and Qin S: Glucose-6-phosphate dehydrogenase: A biomarker and potential therapeutic target for cancer. Anticancer Agents Med Chem. 14:280–289. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lin R, Elf S, Shan C, Kang HB, Ji Q, Zhou L, Hitosugi T, Zhang L, Zhang S, Seo JH, et al: 6-phosphogluconate dehydrogenase links oxidative PPP, lipogenesis and tumour growth by inhibiting LKB1-AMPK signalling. Nat Cell Biol. 17:1484–1496. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kerimi A and Williamson G: Differential impact of flavonoids on redox modulation, bioenergetics, and cell signaling in normal and tumor cells: A comprehensive review. Antioxid Redox Signal. 29:1633–1659. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mazzio EA, Close F and Soliman KF: The biochemical and cellular basis for nutraceutical strategies to attenuate neurodegeneration in parkinson's disease. Int J Mol Sci. 12:506–569. 2011. View Article : Google Scholar : PubMed/NCBI | |
Li P, Tian W and Ma X: Alpha-mangostin inhibits intracellular fatty acid synthase and induces apoptosis in breast cancer cells. Mol Cancer. 13:1382014. View Article : Google Scholar : PubMed/NCBI | |
Sciacovelli M, Gaude E, Hilvo M and Frezza C: The metabolic alterations of cancer cells. Methods Enzymol. 542:1–23. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lee KH, Lee MS, Cha EY, Sul JY, Lee JS, Kim JS, Park JB and Kim JY: Inhibitory effect of emodin on fatty acid synthase, colon cancer proliferation and apoptosis. Mol Med Rep. 15:2163–2173. 2017. View Article : Google Scholar : PubMed/NCBI | |
Sainero-Alcolado L, Garde-Lapido E, Snaebjörnsson MT, Schoch S, Stevens I, Ruiz-Pérez MV, Dyrager C, Pelechano V, Axelson H, Schulze A and Arsenian-Henriksson M: Targeting myc induces lipid droplet accumulation by upregulation of hilpda in clear cell renal cell carcinoma. Proc Natl Acad Sci USA. 121:e23104791212024. View Article : Google Scholar : PubMed/NCBI | |
Wang X and Tian W: Green tea epigallocatechin gallate: A natural inhibitor of Fatty-acid synthase. Biochem Biophys Res Commun. 288:1200–1206. 2001. View Article : Google Scholar : PubMed/NCBI | |
Tan SK, Hougen HY, Merchan JR, Gonzalgo ML and Welford SM: Fatty acid metabolism reprogramming in ccRCC: Mechanisms and potential targets. Nat Rev Urol. 20:48–60. 2023. View Article : Google Scholar : PubMed/NCBI | |
Huang CH, Tsai SJ, Wang YJ, Pan MH, Kao JY and Way TD: EGCG inhibits protein synthesis, lipogenesis, and cell cycle progression through activation of AMPK in p53 positive and negative human hepatoma cells. Mol Nutr Food Res. 53:1156–1165. 2009. View Article : Google Scholar : PubMed/NCBI | |
Qi X, Li Q, Che X, Wang Q and Wu G: The uniqueness of clear cell renal cell carcinoma: Summary of the process and abnormality of glucose metabolism and lipid metabolism in ccRCC. Front Oncol. 11:7277782021. View Article : Google Scholar : PubMed/NCBI | |
Potze L, Di Franco S, Grandela C, Pras-Raves ML, Picavet DI, van Veen HA, van Lenthe H, Mullauer FB, van der Wel NN, Luyf A, et al: Betulinic acid induces a novel cell death pathway that depends on cardiolipin modification. Oncogene. 35:427–437. 2016. View Article : Google Scholar : PubMed/NCBI | |
Horiguchi A, Asano T, Asano T, Ito K, Sumitomo M and Hayakawa M: Fatty acid synthase over expression is an indicator of tumor aggressiveness and poor prognosis in renal cell carcinoma. J Urol. 180:1137–1140. 2008. View Article : Google Scholar : PubMed/NCBI | |
Funabashi H, Kawaguchi A, Tomoda H, Omura S, Okuda S and Iwasaki S: Binding site of cerulenin in fatty acid synthetase. J Biochem. 105:751–755. 1989. View Article : Google Scholar : PubMed/NCBI | |
Li BH and Tian WX: Inhibitory effects of flavonoids on animal fatty acid synthase. J Biochem. 135:85–91. 2004. View Article : Google Scholar : PubMed/NCBI | |
Jung KH, Lee JH, Quach CHT, Paik JY, Oh H, Park JW, Lee EJ, Moon SH and Lee KH: Resveratrol suppresses cancer cell glucose uptake by targeting reactive oxygen species-mediated Hypoxia-inducible factor-1α activation. J Nucl Med. 54:2161–2167. 2013. View Article : Google Scholar : PubMed/NCBI | |
Chen F, Zhuang M, Zhong C, Peng J, Wang X, Li J, Chen Z and Huang Y: Baicalein reverses hypoxia-induced 5-FU resistance in gastric cancer AGS cells through suppression of glycolysis and the PTEN/Akt/HIF-1α signaling pathway. Oncol Rep. 33:457–463. 2015. View Article : Google Scholar : PubMed/NCBI | |
Liu D, Xiao Y, Evans BS and Zhang F: Negative feedback regulation of fatty acid production based on a Malonyl-coA Sensor-actuator. ACS Synth Biol. 4:132–140. 2015. View Article : Google Scholar : PubMed/NCBI | |
Vahlensieck H, Pridzun L, Reichenbach H and Hinnen A: Identification of the yeast ACC1 gene product (acetyl-CoA carboxylase) as the target of the polyketide fungicide soraphen A. Curr Genet. 25:95–100. 1994. View Article : Google Scholar : PubMed/NCBI | |
Kandori S, Kojima T, Matsuoka T, Yoshino T, Sugiyama A, Nakamura E, Shimazui T, Funakoshi Y, Kanaho Y and Nishiyama H: Phospholipase D2 promotes disease progression of renal cell carcinoma through the induction of angiogenin. Cancer Sci. 109:1865–1875. 2018. View Article : Google Scholar : PubMed/NCBI | |
Daurkin I, Eruslanov E, Stoffs T, Perrin GQ, Algood C, Gilbert SM, Rosser CJ, Su LM, Vieweg J and Kusmartsev S: Tumor-associated macrophages mediate immunosuppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Res. 71:6400–6409. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wu J, Zhang Y, Frilot N, Kim JI, Kim WJ and Daaka Y: Prostaglandin E2 regulates renal cell carcinoma invasion through the EP4 Receptor-Rap GTPase signal transduction pathway. J Biol Chem. 286:33954–33962. 2011. View Article : Google Scholar : PubMed/NCBI | |
Fan WH, Wang FC, Jin Z, Zhu L and Zhang JX: Curcumin synergizes with cisplatin to inhibit colon cancer through targeting the MicroRNA-137-Glutaminase axis. Curr Med Sci. 42:108–117. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hassanein M, Hoeksema MD, Shiota M, Qian J, Harris BK, Chen H, Clark JE, Alborn WE, Eisenberg R and Massion PP: SLC1A5 mediates glutamine transport required for lung cancer cell growth and survival. Clin Cancer Res. 19:560–570. 2013. View Article : Google Scholar : PubMed/NCBI | |
Tuna B, Yorukoglu K, Gurel D, Mungan U and Kirkali Z: Significance of COX-2 expression in human renal cell carcinoma. Urology. 64:1116–1120. 2004. View Article : Google Scholar : PubMed/NCBI | |
Galleano M, Calabro V, Prince PD, Litterio MC, Piotrkowski B, Vazquez-Prieto MA, Miatello RM, Oteiza PI and Fraga CG: Flavonoids and metabolic syndrome. Ann N Y Acad Sci. 1259:87–94. 2012. View Article : Google Scholar : PubMed/NCBI | |
Brusselmans K, Vrolix R, Verhoeven G and Swinnen JV: Induction of cancer cell apoptosis by flavonoids is associated with their ability to inhibit fatty acid synthase activity. J Biol Chem. 280:5636–5645. 2005. View Article : Google Scholar : PubMed/NCBI | |
Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E and Vousden KH: Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature. 493:542–546. 2013. View Article : Google Scholar : PubMed/NCBI | |
Metallo CM, Gameiro PA, Bell EL, Mattaini KR, Yang J, Hiller K, Jewell CM, Johnson ZR, Irvine DJ, Guarente L, et al: Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia. Nature. 481:380–384. 2012. View Article : Google Scholar | |
Wang G, Wang YZ, Yu Y, Yin PH and Xu K: The antitumor activity of betulinic Acid-loaded nanoliposomes against colorectal cancer in vitro and in vivo via glycolytic and glutaminolytic pathways. J Biomed Nanotechnol. 16:235–251. 2020. View Article : Google Scholar : PubMed/NCBI | |
Newman AC, Labuschagne CF, Vousden KH and Maddocks OD: Use of 13C315N1-Serine or 13C515N1-Methionine for studying methylation dynamics in cancer cell metabolism and epigenetics. Methods Mol Biol. 1928:55–67. 2019. View Article : Google Scholar : PubMed/NCBI | |
Pan S, Fan M, Liu Z, Li X and Wang H: Serine, glycine and One-carbon metabolism in cancer (Review). Int J Oncol. 58:158–170. 2021. View Article : Google Scholar : PubMed/NCBI | |
Papalazarou V, Newman AC, Huerta-Uribe A, Legrave NM, Falcone M, Zhang T, McGarry L, Athineos D, Shanks E, Blyth K, et al: Phenotypic profiling of solute carriers characterizes serine transport in cancer. Nat Metab. 5:2148–2168. 2023. View Article : Google Scholar : PubMed/NCBI | |
Amelio I, Cutruzzolá F, Antonov A, Agostini M and Melino G: Serine and glycine metabolism in cancer. Trends Biochem Sci. 39:191–198. 2014. View Article : Google Scholar : PubMed/NCBI | |
Samec M, Mazurakova A, Lucansky V, Koklesova L, Pecova R, Pec M, Golubnitschaja O, Al-Ishaq RK, Caprnda M, Gaspar L, et al: Flavonoids attenuate cancer metabolism by modulating lipid metabolism, amino acids, ketone bodies and redox state mediated by Nrf2. Eur J Pharmacol. 949:1756552023. View Article : Google Scholar : PubMed/NCBI | |
Rabinovich S, Adler L, Yizhak K, Sarver A, Silberman A, Agron S, Stettner N, Sun Q, Brandis A, Helbling D, et al: Diversion of aspartate in ASS1-deficient tumours fosters de novo pyrimidine synthesis. Nature. 527:379–383. 2015. View Article : Google Scholar : PubMed/NCBI | |
Bowles TL, Kim R, Galante J, Parsons CM, Virudachalam S, Kung HJ and Bold RJ: Pancreatic cancer cell lines deficient in argininosuccinate synthetase are sensitive to arginine deprivation by arginine deiminase. Int J Cancer. 123:1950–1955. 2008. View Article : Google Scholar : PubMed/NCBI | |
Ensor CM, Holtsberg FW, Bomalaski JS and Clark MA: Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 62:5443–5450. 2002.PubMed/NCBI | |
Pournourmohammadi S, Grimaldi M, Stridh MH, Lavallard V, Waagepetersen HS, Wollheim CB and Maechler P: Epigallocatechin-3-gallate (EGCG) activates AMPK through the inhibition of glutamate dehydrogenase in muscle and pancreatic ß-cells: A potential beneficial effect in the pre-diabetic state? Int J Biochem Cell Biol. 88:220–225. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wettersten HI, Hakimi AA, Morin D, Bianchi C, Johnstone ME, Donohoe DR, Trott JF, Aboud OA, Stirdivant S, Neri B, et al: Grade-dependent metabolic reprogramming in kidney cancer revealed by combined proteomics and metabolomics analysis. Cancer Res. 75:2541–2552. 2015. View Article : Google Scholar : PubMed/NCBI | |
Hornigold N, Dunn KR, Craven RA, Zougman A, Trainor S, Shreeve R, Brown J, Sewell H, Shires M, Knowles M, et al: Dysregulation at multiple points of the kynurenine pathway is a ubiquitous feature of renal cancer: Implications for tumour immune evasion. Br J Cancer. 123:137–147. 2020. View Article : Google Scholar : PubMed/NCBI | |
Riesenberg R, Weiler C, Spring O, Eder M, Buchner A, Popp T, Castro M, Kammerer R, Takikawa O, Hatz RA, et al: Expression of indoleamine 2,3-dioxygenase in tumor endothelial cells correlates with Long-term survival of patients with renal cell carcinoma. Clin Cancer Res. 13:6993–7002. 2007. View Article : Google Scholar : PubMed/NCBI | |
Trott JF, Kim J, Aboud OA, Wettersten H, Stewart B, Berryhill G, Uzal F, Hovey RC, Chen CH, Anderson K, et al: Inhibiting tryptophan metabolism enhances interferon therapy in kidney cancer. Oncotarget. 7:66540–66557. 2016. View Article : Google Scholar : PubMed/NCBI | |
Beckermann KE, Johnson DB and Sosman JA: PD-1/PD-l1 blockade in renal cell cancer. Expert Rev Clin Immunol. 13:77–84. 2017. View Article : Google Scholar : PubMed/NCBI | |
Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A and Mellor AL: Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med. 189:1363–1372. 1999. View Article : Google Scholar : PubMed/NCBI | |
Jochems C, Fantini M, Fernando RI, Kwilas AR, Donahue RN, Lepone LM, Grenga I, Kim YS, Brechbiel MW, Gulley JL, et al: The IDO1 selective inhibitor epacadostat enhances dendritic cell immunogenicity and lytic ability of tumor Antigen-specific T cells. Oncotarget. 7:37762–37772. 2016. View Article : Google Scholar : PubMed/NCBI | |
Naoi M, Wu Y, Shamoto-Nagai M and Maruyama W: Mitochondria in neuroprotection by phytochemicals: Bioactive polyphenols modulate mitochondrial apoptosis system, function and structure. Int J Mol Sci. 20:24512019. View Article : Google Scholar : PubMed/NCBI | |
Abdel-Aziz SM, Aeron A and Kahil TA: Health benefits and possible risks of herbal medicine. Microbes Food Health. 97–116. 2016. View Article : Google Scholar | |
Ojo O, Ajuwape A, Otesile E, Owoade A, Oyekunle M and Adetosoye A: Potentially zoonotic shiga Toxin-producing escherichia coli serogroups in the faeces and meat of Food-producing animals in ibadan, Nigeria. Int J Food Microbiol. 142:214–221. 2010. View Article : Google Scholar : PubMed/NCBI | |
Crowell JA, Korytko PJ, Morrissey RL, Booth TD and Levine BS: Resveratrol-associated renal toxicity. Toxicol Sci. 82:614–619. 2004. View Article : Google Scholar : PubMed/NCBI | |
Balaji S and Chempakam B: Toxicity prediction of compounds from turmeric (Curcuma longa L). Food Chem Toxicol. 48:2951–2959. 2010. View Article : Google Scholar : PubMed/NCBI | |
Qiu P, Man S, Li J, Liu J, Zhang L, Yu P and Gao W: Overdose intake of curcumin initiates the unbalanced state of bodies. J Agric Food Chem. 64:2765–2771. 2016. View Article : Google Scholar : PubMed/NCBI | |
van Duursen MB, Nijmeijer S, De Morree E, de Jong PC and van den Berg M: Genistein induces breast Cancer-associated aromatase and stimulates Estrogen-dependent tumor cell growth in in vitro breast cancer model. Toxicology. 289:67–73. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Wang Y, Wan X, Yang CS and Zhang J: Green tea polyphenol (−)-epigallocatechin-3-gallate triggered hepatotoxicity in mice: Responses of major antioxidant enzymes and the Nrf2 rescue pathway. Toxicol Appl Pharmacol. 283:65–74. 2015. View Article : Google Scholar : PubMed/NCBI | |
McCullough ML and Giovannucci EL: Diet and cancer prevention. Oncogene. 23:6349–6364. 2004. View Article : Google Scholar : PubMed/NCBI | |
Gonzalez CA: Nutrition and cancer: The current epidemiological evidence. Br J Nutr. 96 (Suppl 1):S42–S45. 2006. View Article : Google Scholar : PubMed/NCBI | |
Surh YJ: Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 3:768–780. 2003. View Article : Google Scholar : PubMed/NCBI | |
Shree TJ, Poompavai S, Begum S, Gowrisree V, Hemalatha S, Sieni E and Sundararajan R: Cancer-fighting phytochemicals: Another look. J Nanomed Biother Discov. 9:1622019. | |
Tohill BC and Joint F: Dietary intake of fruit and vegetables and management of body weight (electronic resource). World Health Organization; 2005 | |
Chen H and Liu RH: Potential mechanisms of action of dietary phytochemicals for cancer prevention by targeting cellular signaling transduction pathways. J Agric Food Chem. 66:3260–3276. 2018. View Article : Google Scholar : PubMed/NCBI | |
Guo J, Li W, Shi H, Xie X, Li L, Tang H, Wu M, Kong Y, Yang L, Gao J, et al: Synergistic effects of curcumin with emodin against the proliferation and invasion of breast cancer cells through upregulation of mir-34a. Mol Cell Biochem. 382:103–111. 2013. View Article : Google Scholar : PubMed/NCBI | |
Islam MR, Rahman MM, Dhar PS, Nowrin FT, Sultana N, Akter M, Rauf A, Khalil AA, Gianoncelli A and Ribaudo G: The role of natural and Semi-synthetic compounds in ovarian cancer: Updates on mechanisms of action, current trends and perspectives. Molecules. 28:20702023. View Article : Google Scholar : PubMed/NCBI | |
Hemaiswarya S, Prabhakar PK and Doble M: Synergistic herb interactions with anticancer drugs. In: Herb-drug combinations: A new complementary therapeutic strategy; Springer: pp. 145–173. 2022 | |
Chen S, Zhang Z and Zhang J: Emodin enhances antitumor effect of paclitaxel on human non-small-cell lung cancer cells in vitro and in vivo. Drug Des Devel Ther. 1145–1153. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bai L, Li X, He L, Zheng Y, Lu H and Li J, Zhong L, Tong R, Jiang Z, Shi J and Li J: Antidiabetic potential of flavonoids from traditional chinese medicine: A review. Am J Chin Med. 47:933–957. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wu BY, Liu CT, Su YL, Chen SY, Chen YH and Tsai MY: A review of complementary therapies with medicinal plants for Chemotherapy-induced peripheral neuropathy. Complement Ther Med. 42:226–232. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang ZJ: Therapeutic effects of herbal extracts and constituents in animal models of psychiatric disorders. Life Sci. 75:1659–1699. 2004. View Article : Google Scholar : PubMed/NCBI | |
Mahmoud NN, Carothers AM, Grunberger D, Bilinski RT, Churchill MR, Martucci C, Newmark HL and Bertagnolli MM: Plant phenolics decrease intestinal tumors in an animal model of familial adenomatous polyposis. Carcinogenesis. 21:921–927. 2000. View Article : Google Scholar : PubMed/NCBI | |
Galati G and O'brien PJ: Potential toxicity of flavonoids and other dietary phenolics: Significance for their chemopreventive and anticancer properties. Free Radic Biol Med. 37:287–303. 2004. View Article : Google Scholar : PubMed/NCBI | |
Siddiqi A, Rani M, Bansal P and Rizvi MMA: Renal cell carcinoma management: A step to Nano-chemoprevention. Life Sci. 308:1209222022. View Article : Google Scholar : PubMed/NCBI |