International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
In the past decades, the application of certain new research methods and technologies have led to the proposal of new treatments for cancer, mainly neoadjuvant chemotherapy and targeted immunotherapy. Whilst they have often shown improved efficacy in the initial clinical application, they are associated with drug resistance and side effects (1). Therefore, seeking complementary and alternative treatments from traditional herbal medicine has become one new approach in the treatment of cancer. For example, astragalin has attracted widespread attention due to its wide availability, affordable cost, potent anticancer activity and high safety profile. Moreover, astragalin and its derivatives are considered to have definite cytotoxicity (2).
Astragalin, also known as kaempferol-3-O-glucoside (Fig. 1), is a flavonoid compound found in a number of medicinal plants such as Astragalus, Apocynum venetum leaves, Rosaceae, safflower, cassia seed and edible plants such as green tea, legumes, Allium ursinum and Allium victorialis (3). It has been reported to have anti-inflammatory (4), anti-glycemic, anti-adipogenic (5,6), antioxidant (7) and neuroprotective effects (8,9). Modern pharmacological studies have also reported that astragalin and its derivatives have several biological effects on the cardiovascular system (10,11), the endocrine system (12) and cancer (13,14). Furthermore, it has been reported to be associated with the inhibition of proliferation, migration and invasion of tumor cells, induction of apoptosis and inhibition of angiogenesis (3,15).
Regarding the anticancer effects of astragalin, previous studies have reported effects of astragalin on specific cancers through in vitro and ex vivo experiments, or network pharmacological analyses (16–18). However, unlike previous literature reports on this topic, the present paper summarizes the anticancer effects of the natural compound Astragalin in several types of cancers, systematically outlining the specific mechanisms of the cancer-inhibiting effects of Astragalin, and suggesting future research directions for Astragalin. The present paper presents a comprehensive and systematic review of the mechanism of action of astragalin and its derivatives in several cancers, with an in-depth exploration to elucidate the potential and possibilities of astragalin for anticancer use. Table I summarizes the mechanism of action of astragalin on several cancers and Fig. 2 presents the specific types of anticancer mechanisms of astragalin.
The PubMed (https://pubmed.ncbi.nlm.nih.gov/.), Web of Science (https://www.webofscience.com/) and China Knowledge (https://www.cnki.net/) databases were used for data mining using the following search terms: [‘astragalin’ in the Mesh database (title/abstract) or all its synonyms (kaempferol-3-O-glucoside glycoside)] and [‘Neoplasms’ or all its synonyms (cancer) in the Mesh database (title/abstract)]. Articles published between the creation of the database and April 1, 2025, were imported into Zetero 6.0 (https://www.zotero.org/blog/zotero-6/). A total of 89 papers were obtained after removing duplicates.
The inclusion criteria were as follows: i) In vivo and in vitro experiments with astragalin in the treatment of different types of cancer; ii) preclinical trials of astragalin in different types of cancer; iii) clinical trials of astragalin combined with other drugs; and iv) clinical trials of astragalin for different types of cancer. Articles such as meta-analyses, reviews, abstracts, letters and case reports were not included in the present review. The exclusion criteria were as follows: i) Repeated literature and research; ii) irrelevant articles, defined as articles that did not examine the anticancer mechanism of astragalin; and iii) investigation of the anticancer mechanism of a complex that does not contain astragalin in its composition.
Lung cancer poses a serious health and economic burden to the global population, with the number of new lung cancer cases expected to reach ~3.8 million by 2050 (19). Issues with current lung cancer treatment include high morbidity and mortality rates, late diagnosis, limited therapeutic options, restrictions on targeted therapies, economic and resource constraints, tumor heterogeneity and drug resistance (20), nutritional and quality of life issues (21), and research and clinical translational challenges (22). Therefore, the option of developing novel therapeutic agents with high safety margins may improve this situation.
Among the existing herbal medicine developments, astragalin is expected to be a safe and reliable novel ingredient due to its potent potency to inhibit the activity of lung cancer cells, impede cell migration, proliferation, invasion and induce apoptosis. Astragalin belongs to the class of lotus leaf flavonoids (LLF), and in vitro studies (23,24) have reported that high concentrations (500 µg/ml) of LLF can notably induce apoptosis in human lung cancer A549 cells (25). LLF increases the content of reactive oxygen species and the level of the oxidative end-product malondialdehyde, and at the same time, reduces superoxide dismutase activity. It upregulates several apoptotic factors including caspase-3 and caspase-9, and inhibits the expression of related factors [such as B-cell lymphoma-2 (Bcl-2) and nuclear factor erythroid 2-related factor 2], thereby achieving an anti-lung cancer effect (25). Astragalin acts on the JAK/STAT pathway to regulate the expression of apoptotic factors and related genes in a dose-dependent manner, causing damage to cellular DNA and augmenting reactive oxygen species to promote apoptosis and inhibit migration and invasion (26). Moreover, in vivo studies (27,28) have reported that astragalin may induce cell death via the aspartase, ERK-1/2, AKT and NF-κB signaling pathways, a process that may be associated with an increased Bax:Bcl-2 ratio (29). Network pharmacological studies (30,31) have also reported that astragalin treatment of lung adenocarcinoma may exert its unique inhibitory effects on cell proliferation, migration and apoptosis through the microRNA (miRNA/miR)-140-3p/3-phosphoinositide dependent protein kinase 1 axis (32). Finally, astragalin has been reported to act on the lung cancer A549 cell line with potent cytotoxicity, activating effector caspase-3 and inducing apoptosis (33).
In 2022, ovarian cancer was associated with ~3 million deaths worldwide (34). Whilst current standard treatment regimens have improved patient survival to a certain extent, overall survival remains low due to issues of drug resistance and relapse (35). For example, whilst offering hope for patients with breast cancer gene mutations, poly (ADP-ribose) polymerase inhibitors have limited efficacy, prolonging survival by only a few months, and are associated with resistance (36,37). This has led to the need for the research and development of new treatment modalities and novel drugs. In this process, the active components of traditional Chinese herbs have received attention.
Astragalin has been reported to inhibit cell proliferation and migration and induce apoptosis in ovarian cancer cells by regulating signaling pathways, affecting the expression of specific cytokines and inhibiting the glycolytic pathway. This effect is associated with the upregulation of prolyl hydroxylase domain-containing protein 2 (PHD2) expression and inhibition of hypoxia-inducible factor 1α (HIF-1α) (38,39). PHD2 can act as an oxygen sensor and improve perfusion and oxidization of tumor cells (40), and participates in the programming of macrophage glycolysis (41). HIF-1α is stabilized by binding to hypoxia-responsive element in the anaerobic state, activating key enzymes such as pyruvate dehydrogenase kinase 1 (PDK1) and pyruvate kinase type M2, and inhibiting the tricarboxylic acid cycle, thus shifting glucose metabolism from oxidative phosphorylation to anaerobic glycolysis (42–44). This metabolic reprogramming enhances cell survival. Astragalin inhibits the phosphorylation of related proteins, whereas 3-methyladenine or bafilomycin A1, which act as autophagy inhibitors, can reverse this process. Astragalin can also regulate cellular autophagy by targeting the PI3K/AKT/mTOR signaling pathway (45), a classic signaling pathway in ovarian cancer tumorigenesis, proliferation and progression (46). Thus, astragalin may exert its unique biological effects on ovarian cancer cells.
Cervical cancer ranks second among malignant neoplastic diseases in women and has a high mortality rate (47). Due to the high resistance to platinum-based chemical drugs, this disease is now often treated with radiotherapy (48); however, this can cause damage to the mucosa of the vagina and urinary tract, which can affect patient quality of life (49). Therefore, it is necessary to explore new therapeutic methods with low toxicity and high safety.
It is conceivable that if astragalin is applied in conjunction with radiotherapy drugs, it may be able to exert neuroprotective effects locally, reduce the discomfort of vaginal and urinary tract mucosa, and improve the quality of life of patients. Astragalin inhibits cell proliferation and accelerates the process of apoptosis in cervical cancer cells by regulating signaling pathways and cellular oxygenation (50). Network pharmacological analysis has reported that, in HeLa cells, astragalin can exert regulatory effects on signaling proteins such as EGFR, STAT3 and cyclin D1, affecting the ErbB and forkhead box protein O signaling pathways. This induces an increase in reactive oxygen species and notably inhibits the proliferation and promoting apoptosis in a manner that is linearly associated with concentration (51). Moreover, astragalin, has been reported to exhibit clear anti-cervical cancer activity (52).
Gastric cancer is a great challenge to global public health with the fifth highest global incidence rate, the third highest mortality rate and a high rate of metastasis (53). The molecular and phenotypic heterogeneity of gastric cancer is particularly high (54), and >70% of patients with gastric cancer are diagnosed at an intermediate-to-advanced stage, missing the optimal time for treatment, with limited efficacy of chemotherapy, targeted therapies and immunotherapy, and issues of drug resistance and side effects (55,56).
Astragalin has been reported to act as a gastroprotective component (57), demonstrating satisfactory results in inhibiting gastric cancer cell activity and suppressing cell proliferation (58,59). Astragalin may be used in the treatment of undifferentiated gastric cancer as it can inhibit the EGFR/PDK/AKT signaling pathway, arrest the cell cycle at pre-G1 stage, induce apoptosis by upregulating the transcription of BAX and BAD genes whilst downregulating PDK1 and BCL-2, and reduce the expression of PI3K and AKT proteins (59). Astragalin inhibits the PI3K/AKT signaling pathway and induces an increase in the level of apoptosis-related proteins, thereby effectively suppressing the viability of gastric cancer cells and inhibiting the escape of cancer cells (17). The inhibitory effect of astragalin on mucin 1 mRNA was reported to be the strongest among the related proteins, reducing the expression of ppGalNAcT2 mRNA and fucosyltransferase 4 mRNA, and controlling tumor growth and proliferation (58). Similar to in lung cancer A549 cells, astragalin can also act on the gastric cancer AGS cell line through the same mechanism of action (33). Persistent Helicobacter pylori infection is also associated with the development of gastric cancer (60), and astragalin has been reported to bind to potential protein targets and exhibit unique antibacterial activity, preventing gastric cancer by eradicating persistent infection by this bacterium (61).
In 2022, the global number of colorectal cancer deaths was ~900,000 (34,62). Conventional treatments have limited success in advanced or metastatic colorectal cancer, and chemotherapeutic agents such as 5-fluorouracil, oxaliplatin and irinotecan are widely used but have notable side effects (63). Advanced technologies and new therapeutic ideas [including targeted drugs, immunotherapy and nanotechnology (64,65)] are not yet available for clinical treatment (66). However, herbs may exert anticancer effects by affecting the intestinal flora and improving the intestinal microenvironment (67). For example, astragalin may weaken the activity of colorectal cancer cells, inhibit their proliferation and induce apoptosis through several mechanisms (16,68).
In vitro and in vivo studies have reported that astragalin targets the NF-κB pathway in a dose-dependent manner to reduce the number of colorectal cancer cells, inhibit their growth, induce apoptosis and block their escape (16,27). Its molecular mechanism includes the following: Increasing the level of pro-apoptotic protein factors caspase 6, 7 and 8, whilst inhibiting the expression of Bcl-2; and inhibiting the concentration of CDK2, CDK4 and cell cycle protein D1, increasing the concentration of P21 and P27, so as to cause stalling of the cell growth cycle and promote the cell death (16). Previous research reported that with increasing time and astragalin concentration, the value-added rate and IC50 value of HCT116 cells decreased (16). Moreover, the ability of astragalin to disrupt cell membranes is proportional to its concentration (68). Molecular docking experiments have also reported that astragalin has a high affinity for amino acid targets in the PI3K/AKT/mTOR pathway and is able to retard the growth of cancer foci (69). Novel biological silver nanoparticles combined with astragalin can precisely target carbonic anhydrase IX enzyme and promote anticancer activity (14). Furthermore, the antiproliferative activity of astragalin may be associated with the inhibition of ERK1/2 phosphorylation (70).
In 2022, there were ~2 million women with breast cancer worldwide (34). Moreover, the incidence of male breast cancer is increasing every year by an average of 1.1% per year, which has been associated with unregulated levels of estrogen receptors (71). Chemotherapy, radiotherapy and endocrine therapy, although effective, are often accompanied by serious side effects, such as nausea, vomiting and bone marrow suppression (72–74). In addition, issues such as lymphoedema and chronic arm complications after breast cancer treatment also seriously affect patient quality of life (75).
Astragalin inhibits the activity of the drug metabolizing enzyme cytochrome P450 family 1 subfamily B member 1 and, in vivo, it inhibits estradiolation to produce the carcinogenic metabolite 4-hydroxy-estradiol (76). Furthermore, astragalin inhibits breast cancer cell activity and proliferation and angiogenesis (18,77). Astragalin can act on AKT, zinc finger E-box binding homeobox 1, VEGF and matrix metallopeptidase (MMP)9 targets to inhibit cell motility and exert anti-angiogenic effects (18). It downregulates the protein expression levels of glucose transporter protein (GLUT)-1, lactate dehydrogenase A and hexokinase 2, activates AMP-activated protein kinase (AMPK) and inhibits mTOR activity, reduces glucose uptake, inhibits glycolysis, and inhibits cell proliferation through the AMPK/mTOR pathway (77). Activation of the estrogen receptor is associated with the development of >50% of invasive breast cancers (78), and astragalin, which exerts therapeutic effects comparable with those of hormones with a high margin of safety, may offer new hope for clinical application (79). Moreover, astragalin may mainly interact with human estrogen receptor α receptors to exert anticancer effects (80), and it can also bind to human epidermal growth factor receptor-2 (HER2) and become a potential inhibitor of HER2 (81). Astragalin may also be an effective therapeutic agent for metastasized advanced breast cancer, and this biological effect may be achieved by reducing the expression of TNF-α, which impairs the expression of MMP-9 (82,83).
The incidence and mortality of renal cell carcinoma is increasing annually (84). By contrast, conventional therapies including surgery, ablation and radiation face notable challenges, including more extensive surgery, nephrotoxicity and suboptimal therapeutic range (85). Patients undergoing kidney transplant have a markedly increased risk of new cancers due to the use of potent immunosuppressants (86). Furthermore, dysregulated miRNA expression is associated with cancer cell emergence, growth and cellular drug resistance (87). As a renoprotective drug (88), lower doses of astragalin have been reported to upregulate miRNAs in renal cancer cells to impede tumor cell growth, affect the cell cycle and promote apoptosis in renal cancer cells (89). Previous research reported that 20–50 µM astragalin was cytotoxic to all renal cancer cells, especially the A498 cell line, and the therapeutic index value of astragalin treatment for renal cancer was high. It upregulated the RNA levels of tumor suppressor genes, causing cancer cells to stagnate at the G2/M stage. It also upregulated the expression of apoptotic proteins and inhibited the colony formation of renal cancer cells, which was positively associated with the dosage of astragalin (89).
Melanoma is an aggressive metastatic cancer, with a 5-year survival rate of only 4.6% (90). Global melanoma incidence and mortality in 2040 will be >150% of what it was in 2020 (91). In recent years, immunotherapy and targeted therapies have achieved a certain degree of efficacy as the first-line recommended drugs (92); however, there is a substantial risk of toxicity and drug resistance (93).
Astragalin is cytotoxic and capable of inducing apoptosis. Mitogen- and stress-activated protein kinase 1 (MSK1) is an enzyme involved in several cancer processes, and in melanoma, high levels of MSK1 are often accompanied by high expression of CREB, and increased activity of the MSK1-CREB pathway promotes cell proliferation and metastasis (94). Astragalin directly binds to and inhibits p38 MAPK activity, antagonizing MSK1 phosphorylation and high levels of γ-H2AX in a time- and dose-dependent manner, thereby reducing precancerous skin lesions (95). Astragalin has also been reported to exert toxic effects on melanocytic tumor A375P and SK-MEL-2 cells, increasing the levels of positive cells and sub-G1 populations, activating cleaved poly(ADP-ribose) polymerase, inhibiting SRY-box transcription factor 10 signaling, acting on cell cycle-related proteins and promoting apoptosis (96). Moreover, astragalin has been reported to upregulate the expression of superoxide dismutase and exhibit skin protection effects (97).
Prostate cancer ranks second among malignancies in men (34), and it has a high degree of homogeneous lesion heterogeneity, with common point mutations in <30% of the same surgically resected specimen (98). Androgen deprivation therapy is widely used in metastatic hormone-sensitive prostate cancer and improves the quality of life of patients with advanced disease; however, it also affects their cardiovascular health (99). With disease progression, it can further develop into metastatic castration resistant prostate cancer (mCRPC). Docetaxel can prolong the overall survival of patients with mCRPC (100); however, adverse effects, such as neutropenia and neurotoxicity, can occur (101,102).
Astragalin has been reported to have notable biological effects for the treatment of prostate cancer by inhibiting cell proliferation, migration and invasion, and inducing apoptosis (103). Astragaloside, the most abundant component of Cyperus exaltatus var. iwasakii (CE) secondary metabolites, modulates transcription factors and inhibits the expression of MMP-9, thereby inhibiting invasion, migration and proliferation of prostate cancer cells. In vivo studies have also reported that in an allogeneic model, CE inhibits tumor loading (104). Moreover, Jurinea mesopotamica Hand.-Mazz. contains high levels of astragalin, which can destroy cell organelles, promote the generation of reactive oxygen species, increase apoptotic gene expression and transcription, regulate energy metabolism, inhibit cell proliferation and induce cell death (103).
Liver cancer is a common primary cancer and a high-risk site for cancer metastasis (105). Emerging Yttrium 90 microsphere therapy has been reported to have notable efficacy in patients with advanced, non-surgical liver cancer (106); however, this method requires strict pre-operative screening, is relatively expensive and requires a high level of expertise to perform the surgery (107). By contrast, it is advantageous to explore the development of Traditional Chinese Medicine herbal active ingredients. For example, astragalin has been reported to inhibit the activity of hepatocellular carcinoma cells, suppressing their proliferation and inducing apoptosis through several mechanisms (108–110).
In vitro and in vivo experiments have reported that astragalin can promote miR-125b expression, inhibit hexokinase 2 and shrink allogeneic tumors. It can also promote the oxidative phosphorylation of hepatocellular carcinoma cells, reduce glycolysis, generate reactive oxygen species and antagonize apoptotic proteins to serve an anti-cell proliferation role (109,111). Previous research reported that astragalin reduced Bcl-2 levels, promoted Bax expression, cleaved caspase 3, 8 and 9, and exerted anti-hepatocellular carcinoma activity by regulating apoptosis pathway (110). Astragalin has also been reported to inhibit the activity of HepG2 cells (112), as well as serve as a potential complementary treatment for insulin resistance in patients with hepatocellular carcinoma. Its mechanism of action may include activation of insulin receptor substrate 1, PI3K and AKT, and the regulation of glycolipid metabolism and oxidative stress in cancer cells to a certain extent (113).
With 58,903 deaths, China had the highest number of leukemia deaths in the world in 2021 (114). Personalized treatment approaches for leukemia have been developed; however, there are issues of treatment resistance, long-term side effects and quality of life (115). Several studies have reported that Chinese herbal medicine has satisfactory therapeutic effects on myelosuppression (116). For example, astragalin has been reported to inhibit growth and induce apoptosis in leukemia cells (117).
Certain researchers have isolated drug components with anticancer activity and clarified that astragalin is its main antileukemia component (118). The flavonoid derivative astragalin heptaacetate (AHA) can induce cell death through non-specific caspases. AHA has been reported to concentrate and cleave cell chromatin under aerobic conditions, block the cell cycle in the G0-G1 phase and affect the mitochondrial membrane potential. It also has been reported to regulate cytokine expression, activate MAPK, exhibit notable cytotoxicity and induce apoptosis at the organelle level (117).
The development of NB may be associated with the neural spine during embryonic development (119). The disease is highly malignant (120), with >50% of patients presenting with high-risk phenotypes (121) and a 5-year survival rate of <50% (122). The immune microenvironment of NB is complex, with low immunogenicity of tumor-associated antigens and immunosuppressive factors (such as TGF-β and IL-10), hindering the effectiveness of immunotherapy (123). Moreover, the high clinical behavioral heterogeneity of NB makes it more difficult to develop standardized treatment protocols (124,125).
Previous research has reported that mutations associated with the RAS/MAPK pathway are present in the majority of recurrent NBs (126,127). Astragalin exhibits biological effects on this pathway, inhibiting cell activity and inducing apoptosis (128). Furthermore, astragalin pretreatment reverses the cytoprotective effect, which is associated with the inhibition of oxidative stress and phosphorylation of MAPK, as shown by a reduction in the levels of extracellular protein-regulated protein kinases, p38 and c-Jun N-terminal kinase, exerting their cytotoxic effects and inducing apoptosis in NB SK-N-SH cells (128).
Oral cavity cancer accounts for ~90% of oral malignancies (129). Risk factors for the disease include smoking, alcohol consumption and betel nut consumption (130), and human papillomavirus has also been reported to have an association (131). The disadvantages of traditional treatments include systemic damage, limited efficacy and drug resistance (132). Emerging therapies, such as transoral laser surgery and intensity modulated radiation therapy show potential; however, the popularization and application of these therapies still face technical and economic challenges (133,134). By contrast, herbal medicines have advantages in improving survival, enhancing immune function and reducing the toxicity of chemotherapy (135–137).
Research has demonstrated that the Chinese herbal formulation, FFBZL, composed of Scutellaria barbata D. Don, Astragalus membranaceus and Ligusticum chuanxiong Hort, may be used in the treatment of OSCC; therefore, its molecular mechanisms warrant in-depth exploration. As a crucial component, Astragalus membranaceus exhibits marked antitumor activity in laryngeal squamous cell carcinoma through its extract, total flavonoids of Astragalus, suggesting the potential of its active ingredients (138). Astragalin, one of the key active components of Astragalus membranaceus, currently lacks direct data demonstrating its inhibitory effect on OSCC; however, network pharmacology combined with bioinformatics analysis has revealed that six key ingredients of FFBZL (including components from Astragalus membranaceus) may interact with 820 potential target genes. These targets intersect with OSCC-related target genes, generating 151 common targets (139).
DLBCL is a common aggressive non-Hodgkin's lymphoma that accounts for 30–40% of all non-Hodgkin's lymphomas (140). The R-CHOP regimen (rituximab + cyclophosphamide + adriamycin + vincristine + prednisone) is the first-line standard of care for DLBCL. However, certain patients still relapse after treatment or transition to refractory cases (141).
Astragalin has been reported to induce apoptosis in DLBCL cells by modulating cytokines (142). A previous study reported that astragalin inhibits DLBCL OCI-LY8 cell viability and disrupts the nucleus to promote the emergence of apoptotic vesicles by affecting the JAK/STAT signaling pathway, upregulating the levels of p53, caspase-3 and Bax, and inhibiting Bcl-2 expression (142).
Astragalin has notable anticancer effects in several types of cancer. It also scavenges free radicals in the body (143). Furthermore, overexpression of GLUT5 has been reported to lead to marked changes in glucose metabolism and enhance glycolysis, which is a prominent manifestation of cancer cells. This protein is expected to be used as a biomarker for the diagnosis of cancer and as a therapeutic target (144). A derivative of astragalin, astragalin-6-glucoside, has been reported to inhibit GLUT5 (145). Certain studies suggest that it is associated with glycolysis; however, the exact mechanism of action is not clear (146).
Based on a review of the mechanisms of astragalin and its derivatives against several malignant tumors, the anticancer mechanisms of astragalin are summarized as follows: i) Regulation of signaling pathways: Astragalin can regulate the PI3K/AKT, MAPKs, JAK/STAT and NF-κB signaling pathways, miRNA expression and the Notch1 pathway (27,147), which inhibits tumor cell growth and survival (148); ii) induction of apoptosis: Astragalin can cause loss of caspase activity, mutation of p53 gene and imbalance in the regulation of Bcl2 protein, leading to reduced activity and programmed death of cancer cells (149). It also affects mitochondrial function, reduces mitochondrial membrane potential and increases the Bax/Bcl-2 ratio (150). Notably, a study reported that astragalin ameliorated testicular germ cell apoptosis in rats with varicocele by reducing the expression of caspase 3 and the Bax/Bcl-2 ratio (151). Moreover, astragalin reduced intracellular oxidative stress, activated caspase 3, 7 and 9, and promoted programmed cell death in cancer cells, whilst protecting normal cells from oxidative damage, suggesting that it has a dual effect on the human body (7); iii) inhibition of cell proliferation and migration: Astragalin is closely associated with several angiogenesis-related factors (such as prostaglandin-endoperoxide synthase 2, kinase insert domain receptor and MMP9), revealing potential biological effects with angiogenesis and suggesting new ideas for the treatment of tumors in terms of reducing the number of blood vessels (152). Astragalin downregulates the expression of specific protein factors, regulates miRNAs and induces cell cycle arrest (26); and iv) enhancement of drug sensitivity: In a previous study, astragalin was administered with carboplatin and cisplatin over a range of concentrations, and the results demonstrated that astragalin was associated with an improved combined effect in OVCAR-8 and SKOV-3 cells compared with carboplatin or cisplatin alone (39). This indicates that astragalin could enhance the accumulation of chemotherapeutic drugs in tumor cells and reverse resistance to chemotherapeutic drugs.
Astragalin has a wide range of advantages in cancer treatment, especially in the process of cooperating with chemotherapeutic drugs to reduce toxicity and increase efficiency.
For different cancer types, astragalin has varied anticancer mechanisms of action, but there are also cross-overlapping mechanisms of effect. In general, they include reducing the growth activity of tumor cells, inhibiting tumor cell angiogenesis, inducing apoptosis, blocking the cell growth cycle, and reprogramming the metabolic process of tumor cells, affecting the proliferation, migration and invasion of tumor cells (58). Moreover, several studies have reported that astragalin does not cause harm to normal human cells (8,153).
Astragalin has shown promising results in different types of cancer. For example, radiotherapy may lead to neurotoxicity (154), cancer-caused fatigue (155) and gastrointestinal impairment, and other therapeutic adverse effects (156,157). However, previous research has suggested that if astragalin is used in combination with chemotherapeutic drugs, the neuroprotective, anti-inflammatory, antioxidant (158,159) and antibacterial effects of astragalin could assist the action of the chemotherapeutic drugs (160). Moreover, research has reported that astragalin is involved in targeting cellular resistance factors (89): It can enhance the accumulation of chemotherapeutic drugs in cells and increase the sensitivity of cells to the drugs, thus reversing cellular resistance (39). However, at present, there is a lack of direct research data on the application of astragalin and chemotherapeutic agents in other cancers. Based on the collection of existing literature, we hypothesize that astragalin can serve a direct inhibitory and blocking role in the growth and metabolism of tumor cells, hindering cell growth, proliferation, invasion and migration, and inducing apoptosis through several mechanisms. At the same time, we hypothesize that it can serve a unique toxicity-reducing and synergistic effect in the standardized radiotherapy process. This gives it the potential to be used as a complementary or alternative treatment to first-line chemotherapeutic agents.
Absorption, distribution, metabolism, excretion and toxicity model predictions have reported that astragalin is not susceptible to acute hazards or systemic toxicity and, meeting drug safety requirements, may reduce accumulation toxicity to normal tissues through exocytosis mechanisms (153). In A549 lung cancer cells, astragalin had an IC50 of 150 µM (after 72 h treatment) and inhibited 85% of lung cancer cells; however, to the best of our knowledge, at this dose, there is no evidence of toxicity to normal lung bronchial epithelial cells and cell survival was markedly higher compared with that of cancer cells (26). Moreover, the IC50 was 20–50 µM in A498 renal carcinoma cells and ≤110 µM in normal renal cells, indicating that the toxicity to normal cells was notably lower than that to cancer cells (89). Additionally, the IC50 of astragalin-containing extract of Citrus × aurantium was 23.26 µg/ml in colon cancer HCT-116 cells; however, the toxicity in normal lung fibroblasts (WI-38) was low, with no marked IC50 value reported. This suggests that astragalin may have a selective effect on cancer and normal cells (161). Moreover, the aforementioned findings indicate that astragalin has a wide therapeutic window and exhibits notable anticancer activity with a high safety profile for normal cells.
In summary, astragalin has potent antitumor biological effects with little or no toxic effects on normal tissues; however, this needs to be supported by further evidence from numerous future clinical trials.
Previous research reported that the absolute oral bioavailability of astragalin in rats was <5% (162). This is mainly due to its molecular structure (higher molecular weight and worse water solubility) (163) and its easy conversion by metabolic enzymes [such as uridine diphosphate-glucuronosyltransferases (UGTs)] in the liver and intestine, which is characterized by rapid absorption and clearance (8). However, improved processing or structural optimization may enhance gastric absorption and delay metabolism, thereby increasing bioavailability (164,165). UGT enzymes have been reported to be key metabolizing enzymes of astragalin in vivo. They include CtUGT3 (166) and AtUGT78D2 (167). Moreover, astragalin is distributed in several organs, with the highest concentration in the gastrointestinal tract. It can also cross the blood-brain barrier (168) and can be detected in the liver, lungs and kidneys (169). However, although there are studies that investigated the pharmacokinetics of astragalin (8,170,171), there is a lack of clinical pharmacokinetic validation, and the pharmacological properties of the metabolites, lower bioavailability and stability enhancement need to be further optimized.
Most of the literature included in the present review involved in vivo or in vitro experiments, which largely fail to provide a comprehensive and systematic picture of the drug effects of astragalin within the complex human environment when used in combination or alone. The lack of clinical data, drug metabolism and pharmacokinetics hinders the development of standardized therapeutic protocols for astragalin. Therefore, before the approval of the marketing of this Chinese medicine monomer and its use in large quantities in patients with cancer, several studies are needed: i) Further elucidation of the antitumor mechanism of astragalin through bioinformatics analyses and well-established cyberpharmacological studies; ii) several human studies on the antitumor effect of astragalin and determination of the standardized course of treatment and dosage; iii) exploration of the pharmacological and toxicological effects of astragalin and the possible adverse reactions; and iv) improvement of the description of the pharmacokinetic profile of astragalin. This will aid in the systematic investigation of the different targets of astragalin in several cancer types and promote the discovery and development of new drugs (Fig. 3). At the same time, treatment protocols that maximize anticancer effects should be assessed for different types of cancer to enhance efficacy and reduce toxic side effects. By analyzing the outcomes of treating different types of patients, the differences in the effects of astragalin on different types of cancers can be explored, and the treatment plan can be refined.
Therefore, future in-depth studies on astragalin should focus on the advancement of data on the real drug environment in the clinic, as well as data associated with drug metabolism pathways and pharmacokinetics in a standardized manner, with a view to providing an objective basis for the development of new drugs and the update of therapeutic regimens.
In the research of malignant tumors, the development and utilization of herbal medicines has become a major topic. The results of the present review indicate that astragalin, with its wide and natural sources, strong anticancer activity, high safety value and low cost, has potential to be an alternative drug with considerable efficacy in the prevention and treatment of malignant tumors; however, further research is required. Although astragalin has demonstrated promising antitumor effects, its pharmacokinetic profile is incompletely defined, and its low oral bioavailability limits its potential clinical application. Future studies should investigate the bioavailability of astragalin through systematic in vitro and animal experiments, and the optimization of formulation processes for novel drug delivery systems. With further research, astragalin has the potential to become an alternative drug for cancer treatment in the future.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant no. 82274566).
Not applicable.
YFZ retrieved the data and participated in the conceptualization and design of the article as well as the data analysis; YQF wrote the original manuscript and made several revisions with the assistance and editing of CL, DND and FYL. FJH was involved in the design of the manuscript and the interpretation of the data. Data authentication is not applicable. All authors contributed to the article and read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
AHA |
astragalin heptaacetate |
|
Bcl-2 |
B-cell lymphoma-2 |
|
CE |
Cyperus exaltatus var. iwasakii |
|
DLBCL |
diffuse large B-cell lymphoma |
|
GLUT5 |
glucose transporter protein 5 |
|
HER2 |
human epidermal growth factor receptor-2 |
|
HIF-1α |
hypoxia inducible factor-1α |
|
LLF |
lotus leaf flavonoid |
|
mCRPC |
metastatic castration resistant prostate cancer |
|
MMP |
matrix metalloproteinase |
|
MSK1 |
mitogen- and stress-activated protein kinase 1 |
|
NB |
neuroblastoma |
|
Nussinov R, Tsai CJ and Jang H: Anticancer drug resistance: An update and perspective. Drug Resist Updat. 59:1007962021. View Article : Google Scholar : PubMed/NCBI | |
|
Zilla MK, Nayak D, Amin H, Nalli Y, Rah B, Chakraborty S, Kitchlu S, Goswami A and Ali A: 4′-Demethyl-deoxypodophyllotoxin glucoside isolated from Podophyllum hexandrum exhibits potential anticancer activities by altering Chk-2 signaling pathway in MCF-7 breast cancer cells. Chem Biol Interact. 224:100–107. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Martău GA, Bernadette-Emőke T, Odocheanu R, Soporan DA, Bochiș M, Simon E and Vodnar DC: Vaccinium species (Ericaceae): Phytochemistry and biological properties of medicinal plants. Molecules. 28:15332023. View Article : Google Scholar : PubMed/NCBI | |
|
Ruan J, Shi Z, Cao X, Dang Z, Zhang Q, Zhang W, Wu L, Zhang Y and Wang T: Research progress on anti-inflammatory effects and related mechanisms of astragalin. Int J Mol Sci. 25:44762024. View Article : Google Scholar : PubMed/NCBI | |
|
Barge S, Deka B, Kashyap B, Bharadwaj S, Kandimalla R, Ghosh A, Dutta PP, Samanta SK, Manna P, Borah JC and Talukdar NC: Astragalin mediates the pharmacological effects of Lysimachia candida Lindl on adipogenesis via downregulating PPARG and FKBP51 signaling cascade. Phytother Res. 35:6990–7003. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mladenova SG, Vasileva LV, Savova MS, Marchev AS, Tews D, Wabitsch M, Ferrante C, Orlando G and Georgiev MI: Anti-Adipogenic effect of alchemilla monticola is mediated Via PI3K/AKT signaling inhibition in human adipocytes. Front Pharmacol. 12:7075072021. View Article : Google Scholar : PubMed/NCBI | |
|
Ceramella J, Loizzo MR, Iacopetta D, Bonesi M, Sicari V, Pellicanò TM, Saturnino C, Malzert-Fréon A, Tundis R, Sinicropi MS, et al: Anchusa azurea Mill. (Boraginaceae) aerial parts methanol extract interfering with cytoskeleton organization induces programmed cancer cells death. Food Funct. 10:4280–4290. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Zhong K, Qin S, Jing Y, Liu S, Li D and Peng C: Astragalin: A food-origin flavonoid with therapeutic effect for multiple diseases. Front Pharmacol. 14:12659602023. View Article : Google Scholar : PubMed/NCBI | |
|
Tao Y, Bao J, Zhu F, Pan M, Liu Q and Wang P: Ethnopharmacology of Rubus idaeus Linnaeus: A critical review on ethnobotany, processing methods, phytochemicals, pharmacology and quality control. J Ethnopharmacol. 302:1158702023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao ZW, Zhang M, Wang G, Zou J, Gao JH, Zhou L, Wan XJ, Zhang DW, Yu XH and Tang CK: Astragalin retards atherosclerosis by promoting cholesterol efflux and inhibiting the inflammatory response via upregulating ABCA1 and ABCG1 expression in macrophages. J Cardiovasc Pharmacol. 77:217–227. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rajani V, Umadevi S and Raju CN: A review on exploring the phytochemical and pharmacological significance of Indigofera astragalina. Pharmacogn Mag. 20:363–371. 2024. View Article : Google Scholar | |
|
Wang N, Singh D and Wu Q: Astragalin attenuates diabetic cataracts via inhibiting aldose reductase activity in rats. Int J Ophthalmol. 16:1186–1195. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bangar SP, Singh A, Chaudhary V, Sharma N and Lorenzo JM: Beetroot as a novel ingredient for its versatile food applications. Crit Rev Food Sci Nutr. 63:8403–8427. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mohammed AE, Alghamdi SS, Shami A, Suliman RS, Aabed K, Alotaibi MO and Rahman I: In silico prediction of malvaviscus arboreus metabolites and green synthesis of silver nanoparticles-opportunities for safer anti-bacterial and anti-cancer precision medicine. Int J Nanomedicine. 18:2141–2162. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen SC, Xiang XY, Han WC, Chen MY and Xie BC: Research progress on pharmacological properties and mechanism of astragalin. Chin J Tradit Chin Med. 40:118–123+287. 2022.(In Chinese). | |
|
Yang M, Li WY, Xie J, Wang ZL, Wen YL, Zhao CC, Tao L, Li LF, Tian Y and Sheng J: Astragalin inhibits the proliferation and migration of human colon cancer HCT116 cells by regulating the NF-κB signaling pathway. Front Pharmacol. 12:6392562021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Lv J, Li X and Lin Q: The flavonoid Astragalin shows anti-tumor activity and inhibits PI3K/AKT signaling in gastric cancer. Chem Biol Drug Des. 98:779–786. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tian S, Wei Y, Hu H and Zhao H: Mixed computational-experimental study to reveal the anti-metastasis and anti-angiogenesis effects of Astragalin in human breast cancer. Comput Biol Med. 150:1061312022. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma R: Mapping of global, regional and national incidence, mortality and mortality-to-incidence ratio of lung cancer in 2020 and 2050. Int J Clin Oncol. 27:665–675. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Rossi R, De Angelis ML, Xhelili E, Sette G, Eramo A, De Maria R, Incani UC, Francescangeli F and Zeuner A: Lung cancer organoids: The rough path to personalized medicine. Cancers (Basel). 14:37032022. View Article : Google Scholar : PubMed/NCBI | |
|
Prete M, Ballarin G, Porciello G, Arianna A, Luongo A, Belli V, Scalfi L and Celentano E: Bioelectrical impedance analysis-derived phase angle (PhA) in lung cancer patients: A systematic review. BMC Cancer. 24:6082024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu S, Wei W, Wang J and Chen T: Theranostic applications of selenium nanomedicines against lung cancer. J Nanobiotechnology. 21:962023. View Article : Google Scholar : PubMed/NCBI | |
|
Xi Chen and Jin Qi: Flavonoids and Alkaloids in Lotus Leaves. Chin J Exp Tradit Med Formulae. 211–214. 2015. | |
|
Wang D, Yu J, Cheng L, Hu X, Xiang M, Zhou Y, Tao B, Liu C and Xia Y: Research Progress on Main Constituents and Biological Activities of Lotus Leaves. Agricultural Technology & Equipment. 71–72. 742021.(In Chinese). | |
|
Jia XB, Zhang Q, Xu L, Yao WJ and Wei L: Lotus leaf flavonoids induce apoptosis of human lung cancer A549 cells through the ROS/p38 MAPK pathway. Biol Res. 54:72021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu G, Yu B, Wang R, Jiang J, Wen F and Shi X: Astragalin flavonoid inhibits proliferation in human lung carcinoma cells mediated via induction of caspase-dependent intrinsic pathway, ROS production, cell migration and invasion inhibition and targeting JAK/STAT signalling pathway. Cell Mol Biol (Noisy-le-grand). 67:44–49. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao D, Bai JC and Zhang B: Research progress of signal pathways related to the anti-tumor effect of Astragalin. J Pract Oncol. 38:62–66. 2024. | |
|
Liang Y and Chen W: Effect of Astragalin on proliferation, migration, apoptosis and oxidative stress of lung cancer A549 cells. Chin J Clin Pharmacol. 39:999–1003. 2023.(In Chinese). | |
|
Chen M, Cai F, Zha D, Wang X, Zhang W, He Y, Huang Q, Zhuang H and Hua ZC: Astragalin-induced cell death is caspase-dependent and enhances the susceptibility of lung cancer cells to tumor necrosis factor by inhibiting the NF-кB pathway. Oncotarget. 8:26941–26958. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yan C, Pang X, Fan W, Dai W, Hu X, Mei Q and Zeng C: Mechanism of leaves of Microcos paniculata improving of dyspepsia based on network pharmacology and molecular docking. Drugs & Clinic. 38:1335–1343. 2023.(In Chinese). | |
|
Li M, Zhao Z, Xuan J, Li Z and Ma T: Advances in Studies on Chemical Constituents and Pharmacological Effects of Lotus Leaves. Journal of Liaoning University of TCM. 22:135–138. 2020.(In Chinese). | |
|
Bai J, Chen Y, Zhao G and Gui R: In vitro and vivo experiments revealing astragalin inhibited lung adenocarcinoma development via LINC00582/miR-140-3P/PDPK1. J Biochem Mol Toxicol. 38:e700422024. View Article : Google Scholar : PubMed/NCBI | |
|
Ribeiro V, Ferreres F, Macedo T, Gil-Izquierdo Á, Oliveira AP, Gomes NGM, Araújo L, Pereira DM, Andrade PB and Valentão P: Activation of caspase-3 in gastric adenocarcinoma AGS cells by Xylopia aethiopica (Dunal) A. Rich. fruit and characterization of its phenolic fingerprint by HPLC-DAD-ESI(Ion Trap)-MSn and UPLC-ESI-QTOF-MS2. Food Res Int. 141:1101212021. View Article : Google Scholar : PubMed/NCBI | |
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Webb PM and Jordan SJ: Global epidemiology of epithelial ovarian cancer. Nat Rev Clin Oncol. 21:389–400. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang C, Xia BR, Zhang ZC, Zhang YJ, Lou G and Jin WL: Immunotherapy for ovarian cancer: Adjuvant, combination, and neoadjuvant. Front Immunol. 11:5778692020. View Article : Google Scholar : PubMed/NCBI | |
|
Gu L, Du N, Jin Q, Li S, Xie L, Mo J, Shen Z, Mao D, Ji J, et al: Magnitude of benefit of the addition of poly ADP-ribose polymerase (PARP) inhibitors to therapy for malignant tumor: A meta-analysis. Crit Rev Oncol Hematol. 147:1028882020. View Article : Google Scholar : PubMed/NCBI | |
|
Song L and Fu Q: Study of the effect of astragalin on proliferation of ovarian cancer cells by inhibiting the glycolytic pathway induced Via HIF-1α. Pract Oncol J. 32:503–509. 2018. | |
|
Ling Song and Qiong Fu: Astragalin inhibits hypoxia-induced proliferation and invasion of ovarian cancer cells by up-regulating PHD2. J Pract Oncol. 33:407–414. 2019. | |
|
Mazzone M, Dettori D, de Oliveira RL, Loges S, Schmidt T, Jonckx B, Tian YM, Lanahan AA, Pollard P, de Almodovar CR, et al: Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell. 136:839–851. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Guentsch A, Beneke A, Swain L, Farhat K, Nagarajan S, Wielockx B, Raithatha K, Dudek J, Rehling P and Zieseniss A: PHD2 is a regulator for glycolytic reprogramming in macrophages. Mol Cell Biol. 37:e00236–e00216. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mehla K and Singh PK: MUC1: A novel metabolic master regulator. Biochim Biophys Acta. 1845:126–135. 2014.PubMed/NCBI | |
|
Katayama Y, Uchino J, Chihara Y, Tamiya N, Kaneko Y, Yamada T and Takayama K: Tumor neovascularization and developments in therapeutics. Cancers (Basel). 11:3162019. View Article : Google Scholar : PubMed/NCBI | |
|
Li RL, He LY, Zhang Q, Liu J, Lu F, Duan HX, Fan LH, Peng W, Huang YL and Wu CJ: HIF-1α is a potential molecular target for herbal medicine to treat diseases. Drug Des Devel Ther. 14:4915–4949. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang CZ, Wang SH, Zhang RH, Lin JH, Tian YH, Yang YQ, Liu J and Ma YX: Neuroprotective effect of astragalin via activating PI3K/Akt-mTOR-mediated autophagy on APP/PS1 mice. Cell Death Discov. 9:152023. View Article : Google Scholar : PubMed/NCBI | |
|
Ediriweera MK, Tennekoon KH and Samarakoon SR: Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin Cancer Biol. 59:147–160. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Shu-ping Qu, Chun-jun Liu and Ju-hua Liang: Correlation between serum miR-410,miR-875 expression with HPV infection in cervical cancer patients. J Path Biol. 19:1404–1407. 2024. | |
|
Abu-Rustum NR, Yashar CM, Arend R, Barber E, Bradley K, Brooks R, Campos SM, Chino J, Chon HS, Crispens MA, et al: NCCN Guidelines® insights: Cervical cancer, version 1.2024. J Natl Compr Canc Netw. 21:1224–1233. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Raj S, Prasad RR and Ranjan A: Incidence of vaginal toxicities following definitive chemoradiation in intact cervical cancer: A meta-analysis. J Contemp Brachytherapy. 16:241–256. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zeng W and Chen L: Astragalin inhibits the proliferation of high-risk HPV-positive cervical epithelial cells and attenuates malignant cervical lesions. Cytotechnology. 77:802025. View Article : Google Scholar : PubMed/NCBI | |
|
He M, Yasin K, Yu S, Li J and Xia L: Total flavonoids in Artemisia absinthium L. and evaluation of its anticancer activity. Int J Mol Sci. 24:163482023. View Article : Google Scholar : PubMed/NCBI | |
|
Zerrouki S, Mezhoud S, Yilmaz MA, Yaglioglu AS, Bakir D, Demirtas I and Mekkiou R: LC/MS-MS analyses and in vitro anticancer activity of Tourneuxia variifolia extracts. Nat Prod Res. 36:4506–4510. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Zhou N, Lan H, Yang F, Dong B and Zhang X: The ferroptosis molecular subtype reveals characteristics of the tumor microenvironment, immunotherapeutic response, and prognosis in gastric cancer. Int J Mol Sci. 23:97672022. View Article : Google Scholar : PubMed/NCBI | |
|
Mamun TI, Younus S and Rahman MH: Gastric cancer-Epidemiology, modifiable and non-modifiable risk factors, challenges and opportunities: An updated review. Cancer Treat Res Commun. 41:1008452024.PubMed/NCBI | |
|
Wang J, Du L and Chen X: Adenosine signaling: Optimal target for gastric cancer immunotherapy. Front Immunol. 13:10278382022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Huang T, Wang L, Wang Y, Liu Y, Bai J, Wen X, Li Y, Long K and Zhang H: Traditional Chinese medicine in the treatment of chronic atrophic gastritis, precancerous lesions and gastric cancer. J Ethnopharmacol. 337:1188122025. View Article : Google Scholar : PubMed/NCBI | |
|
Radziejewska I, Supruniuk K, Tomczyk M, Izdebska W, Borzym-Kluczyk M, Bielawska A, Bielawski K and Galicka A: p-Coumaric acid, Kaempferol, Astragalin and Tiliroside influence the expression of glycoforms in AGS gastric cancer cells. Int J Mol Sci. 23:86022022. View Article : Google Scholar : PubMed/NCBI | |
|
Chu ZH, He SY, Wang Y, Zhu RT, Gu YL and Chen JY: Effects of Astragalin on apoptosis of undifferentiated gastric cancer cells. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 38:520–524. 2022.(In Chinese). PubMed/NCBI | |
|
Sharafutdinov I, Tegtmeyer N, Linz B, Rohde M, Vieth M, Tay AC, Lamichhane B, Tuan VP, Fauzia KA, Sticht H, et al: A single-nucleotide polymorphism in Helicobacter pylori promotes gastric cancer development. Cell Host Microbe. 31:1345–1358.e6. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Tan A, Scortecci KC, De Medeiros NM, Kukula-Koch W, Butler TJ, Smith SM and Boylan F: Plukenetia volubilis leaves as source of anti-Helicobacter pylori agents. Front Pharmacol. 15:14614472024. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy CC and Zaki TA: Changing epidemiology of colorectal cancer-birth cohort effects and emerging risk factors. Nat Rev Gastroenterol Hepatol. 21:25–34. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ebnudesita FR, Dienanta SB, Jannah AR and I'tishom R: Potency of soursop leaf extract and curcumin with magnetic and mucus-penetrating nanoparticle as colorectal cancer alternative therapY. J Vocational Health Studies. 5:186–191. 2022. View Article : Google Scholar | |
|
Abedizadeh R, Majidi F, Khorasani HR, Abedi H and Sabour D: Colorectal cancer: A comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev. 43:729–753. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Chen C, Wang P and Song T: Mechanisms of cellular effects directly induced by magnetic nanoparticles under magnetic fields. J Nanomaterials. 2017:1–13. 2017. View Article : Google Scholar | |
|
Eng C, Yoshino T, Ruíz-García E, Mostafa N, Cann CG, O'Brian B, Benny A, Perez RO and Cremolini C: Colorectal cancer. Lancet. 404:294–310. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zou Y, Wang S, Zhang H, Gu Y, Chen H, Huang Z, Yang F, Li W, Chen C, Men L, et al: The triangular relationship between traditional Chinese medicines, intestinal flora, and colorectal cancer. Med Res Rev. 44:539–567. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Augustynowicz D, Lemieszek MK, Strawa JW, Wiater A and Tomczyk M: Anticancer potential of acetone extracts from selected Potentilla species against human colorectal cancer cells. Front Pharmacol. 13:10273152022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhai Y, Sun J, Sun C, Zhao H, Li X, Yao J, Su J, Xu X, Xu X, Hu J, et al: Total flavonoids from the dried root of Tetrastigma hemsleyanum Diels et Gilg inhibit colorectal cancer growth through PI3K/AKT/mTOR signaling pathway. Phytother Res. 36:4263–4277. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Tragulpakseerojn J, Yamaguchi N, Pamonsinlapatham P, Wetwitayaklung P, Yoneyama T, Ishikawa N, Ishibashi M and Apirakaramwong A: Anti-proliferative effect of Moringa oleifera Lam (Moringaceae) leaf extract on human colon cancer HCT116 cell line. Trop J Pharm Res. 16:371–378. 2017. View Article : Google Scholar | |
|
Chidambaram A, Prabhakaran R, Sivasamy S, Kanagasabai T, Thekkumalai M, Singh A, Tyagi MS and Dhandayuthapani S: Male breast cancer: Current scenario and future perspectives. Technol Cancer Res Treat. 23:153303382412618362024. View Article : Google Scholar : PubMed/NCBI | |
|
Palmer BV, Walsh GA, McKinna JA and Greening WP: Adjuvant chemotherapy for breast cancer: Side effects and quality of life. BMJ. 281:1594–1597. 1980. View Article : Google Scholar : PubMed/NCBI | |
|
Di Nardo P, Lisanti C, Garutti M, Buriolla S, Alberti M, Mazzeo R and Puglisi F: Chemotherapy in patients with early breast cancer: Clinical overview and management of long-term side effects. Expert Opin Drug Saf. 21:1341–1355. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kim SD, Kwag EB, Yang MX and Yoo HS: Efficacy and safety of ginger on the side effects of chemotherapy in breast cancer patients: systematic review and meta-analysis. Int J Mol Sci. 23:112672022. View Article : Google Scholar : PubMed/NCBI | |
|
Peeters NJMC, Kaplan ZLR, Clarijs ME, Mureau MAM, Verhoef C, van Dalen T, Husson O and Koppert LB: Health-related quality of life (HRQoL) after different axillary treatments in women with breast cancer: A 1-year longitudinal cohort study. Qual Life Res. 33:467–479. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Meng X, Zhang A, Wang X and Sun H: A kaempferol-3-O-β-D-glucoside, intervention effect of astragalin on estradiol metabolism. Steroids. 149:1084132019. View Article : Google Scholar : PubMed/NCBI | |
|
Zeb A, Khan W, Ul Islam W, Khan F, Khan A, Khan H, Khalid A, Rehman NU, Ullah A and Al-Harrasi A: Exploring the anticancer potential of astragalin in triple negative breast cancer cells by attenuating glycolytic pathway through AMPK/mTOR. Curr Med Chem. Jul 25–2024.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI | |
|
Nunnery SE and Mayer IA: Targeting the PI3K/AKT/mTOR pathway in hormone-positive breast cancer. Drugs. 80:1685–1697. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Song X, Shen L, Contreras JM, Liu Z, Ma K, Ma B, Liu X and Wang DO: New potential selective estrogen receptor modulators in traditional Chinese medicine for treating menopausal syndrome. Phytother Res. 38:4736–4756. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huq AM, Stanslas J, Nizhum N, Uddin MN, Maulidiani M, Roney M, Abas F and Jamal JA: Estrogenic post-menopausal anti-osteoporotic mechanism of Achyranthes aspera L.: Phytochemicals and network pharmacology approaches. Heliyon. 10:e387922024. View Article : Google Scholar : PubMed/NCBI | |
|
Bellai A, Deka S, Tag H, Bhattacharya K and Hui PK: Integrated pharmacognostic and computational analysis of Hydrocotyle javanica thunb. Phytochemicals as a potential HER2 tyrosine kinase inhibitor in breast cancer. Pept Sci. 119:e243722024. View Article : Google Scholar | |
|
Ahn S, Jung H, Jung Y, Lee J, Shin SY, Lim Y and Lee Y: Identification of the active components inhibiting the expression of matrix metallopeptidase-9 by TNFα in ethyl acetate extract of Euphorbia humifusa Willd. J Appl Biol Chem. 62:367–374. 2019. View Article : Google Scholar | |
|
Shin SY, Kim CG, Jung YJ, Jung Y, Jung H, Im J, Lim Y and Lee YH: Euphorbia humifusa Willd exerts inhibition of breast cancer cell invasion and metastasis through inhibition of TNFα-induced MMP-9 expression. BMC Complement Altern Med. 16:4132016. View Article : Google Scholar : PubMed/NCBI | |
|
Cirillo L, Innocenti S and Becherucci F: Global epidemiology of kidney cancer. Nephrol Dial Transplant. 39:920–928. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Young M, Jackson-Spence F, Beltran L, Day E, Suarez C, Bex A, Powles T and Szabados B: Renal cell carcinoma. Lancet. 404:476–491. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Imamura R, Tanaka R, Taniguchi A, Nakazawa S, Kato T, Yamanaka K, Namba-Hamano T, Kakuta Y, Abe T, Tsutahara K, et al: Everolimus reduces cancer incidence and improves patient and graft survival rates after kidney transplantation: A multi-center study. J Clin Med. 11:2492022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H: microRNA-Based cancer diagnosis and therapy. Int J Mol Sci. 25:2302024. View Article : Google Scholar | |
|
Zhang W, Chan C, Zhang K, Qin H, Yu BY, Xue Z, Zheng X and Tian J: Discovering a new drug against acute kidney injury by using a tailored photoacoustic imaging probe. Adv Mater. 36:e23113972024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu L, Zhu L, Chen J, Cui T and Liao W: Astragalin induced selective kidney cancer cell death and these effects are mediated via mitochondrial mediated cell apoptosis, cell cycle arrest, and modulation of key tumor-suppressive miRNAs. J BUON. 24:1245–1251. 2019.PubMed/NCBI | |
|
Wu N, Li J and Tao J: Hot spots in diagnosis of malignant melanoma. J Diagn Concepts Pract. 22:215–219. 2023.(In Chinese). | |
|
Arnold M, Singh D, Laversanne M, Vignat J, Vaccarella S, Meheus F, Cust AE, de Vries E, Whiteman DC and Bray F: Global burden of cutaneous melanoma in 2020 and projections to 2040. JAMA Dermatol. 158:495–503. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Swetter SM, Johnson D, Albertini MR, Barker CA, Bateni S, Baumgartner J, Bhatia S, Bichakjian C, Boland G, Chandra S, et al: NCCN Guidelines® insights: Melanoma: Cutaneous, version 2.2024: Featured updates to the NCCN guidelines. J Natl Compr Canc Netw. 22:290–298. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ernst M and Giubellino A: The current state of treatment and future directions in cutaneous malignant melanoma. Biomedicines. 10:8222022. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Liu X, Wang W, Li C and Li X: MSK1 promotes cell proliferation and metastasis in uveal melanoma by phosphorylating CREB. Arch Med Sci. 16:1176–1188. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li N, Zhang K, Mu X, Tian Q, Liu W, Gao T, Ma X and Zhang J: Astragalin attenuates UVB radiation-induced actinic keratosis formation. Anticancer Agents Med Chem. 18:1001–1008. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
You OH, Shin EA, Lee H, Kim JH, Sim DY, Kim JH, Kim Y, Khil JH, Baek NI and Kim SH: Apoptotic effect of astragalin in melanoma skin cancers via activation of caspases and inhibition of sry-related HMg-Box gene 10. Phytother Res. 31:1614–1620. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Bian Y, Cheng Y, Sun R and Li G: Effect of lemon peel flavonoids on UVB-induced skin damage in mice. RSC Adv. 10:31470–31478. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Løvf M, Zhao S, Axcrona U, Johannessen B, Bakken AC, Carm KT, Hoff AM, Myklebost O, Meza-Zepeda LA, Lie AK, et al: multifocal primary prostate cancer exhibits high degree of genomic heterogeneity. Eur Urol. 75:498–505. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lafontaine ML and Kokorovic A: Cardiometabolic side effects of androgen deprivation therapy in prostate cancer. Curr Opin Support Palliat Care. 16:216–222. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Moses KA, Sprenkle PC, Bahler C, Box G, Carlsson SV, Catalona WJ, Dahl DM, Dall'Era M, Davis JW, Drake BF, et al: NCCN Guidelines® insights: Prostate cancer early detection, version 1.2023. J Natl Compr Canc Netw. 21:236–246. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Beer TM, Hotte SJ, Saad F, Alekseev B, Matveev V, Fléchon A, Gravis G, Joly F, Chi KN, Malik Z, et al: Custirsen (OGX-011) combined with cabazitaxel and prednisone versus cabazitaxel and prednisone alone in patients with metastatic castration-resistant prostate cancer previously treated with docetaxel (AFFINITY): A randomised, open-label, international, phase 3 trial. Lancet Oncol. 18:1532–1542. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shahab MRF and Shahab N: Recent advances and future directions in the management of metastatic castration-resistant prostate cancer asing androgen receptor signaling inhibitors and poly-adipose polymerase inhibitors: A literature review. International J Multidisciplinary Research. 6:22024. | |
|
Koyuncu I, Temiz E, Seker F, Balos MM, Akkafa F, Yuksekdag O, Yılmaz MA and Zengin G: A mixed-apoptotic effect of Jurinea mesopotamica extract on prostate cancer cells: A promising source for natural chemotherapeutics. Chem Biodivers. 21:e2023017472024. View Article : Google Scholar : PubMed/NCBI | |
|
Kim H, Hwang B, Cho S, Kim WJ, Myung SC, Choi YH, Kim WJ, Lee S and Moon SK: The ethanol extract of Cyperus exaltatus var. iwasakii exhibits cell cycle dysregulation, ERK1/2/p38 MAPK/AKT phosphorylation, and reduced MMP-9-mediated metastatic capacity in prostate cancer models in vitro and in vivo. Phytomedicine. 114:1547942023. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Ramadori P, Pfister D, Seehawer M, Zender L and Heikenwalder M: The immunological and metabolic landscape in primary and metastatic liver cancer. Nat Rev Cancer. 21:541–557. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chiang CL, Chiu KWH, Chan KSK, Lee FAS, Li JCB, Wan CWS, Dai WC, Lam TC, Chen W, Wong NSM, et al: Sequential transarterial chemoembolisation and stereotactic body radiotherapy followed by immunotherapy as conversion therapy for patients with locally advanced, unresectable hepatocellular carcinoma (START-FIT): A single-arm, phase 2 trial. Lancet Gastroenterol Hepatol. 8:169–178. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pennington B, Akehurst R, Wasan H, Sangro B, Kennedy AS, Sennfält K and Bester L: Cost-effectiveness of selective internal radiation therapy using yttrium-90 resin microspheres in treating patients with inoperable colorectal liver metastases in the UK. J Med Econ. 18:797–804. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Chen F, Gu L, Jiang Y, Cai Z, Zhao Y, Chen L, Zhu Z and Liu X: Discovery and validation of COX2 as a target of flavonoids in Apocyni Veneti Folium: Implications for the treatment of liver injury. J Ethnopharmacol. 326:1179192024. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Hao J, Zhang L, Cheng Z, Deng X and Shu G: Astragalin reduces hexokinase 2 through increasing miR-125b to inhibit the proliferation of hepatocellular carcinoma cells in vitro and in vivo. J Agric Food Chem. 65:5961–5972. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hong Z, Lu Y, Ran C, Tang P, Huang J, Yang Y, Duan X and Wu H: The bioactive ingredients in Actinidia chinensis Planch. Inhibit liver cancer by inducing apoptosis. J Ethnopharmacol. 281:1145532021. View Article : Google Scholar : PubMed/NCBI | |
|
Pirvu L, Stefaniu A, Neagu G, Albu B and Pintilie L: In vitro cytotoxic and Antiproliferative Activity of Cydonia oblonga flower petals, leaf and fruit pellet ethanolic extracts. Docking simulation of the active flavonoids on anti-apoptotic protein Bcl-2. Open Chemistry. 16:591–604. 2018. View Article : Google Scholar | |
|
Kaidash OA, Kostikova VA, Udut EV, Shaykin VV and Kashapov DR: Extracts of Spiraea hypericifolia L. and Spiraea crenata L.: The phenolic profile and biological activities. Plants. 11:27282022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou K, Xiao S, Cao S, Zhao C, Zhang M and Fu Y: Improvement of glucolipid metabolism and oxidative stress via modulating PI3K/Akt pathway in insulin resistance HepG2 cells by chickpea flavonoids. Food Chem X. 23:1016302024. View Article : Google Scholar : PubMed/NCBI | |
|
Murray CJL: The global burden of disease study at 30 years. Nat Med. 28:2019–2026. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Nadiminti KVG, Sahasrabudhe KD and Liu H: Menin inhibitors for the treatment of acute myeloid leukemia: Challenges and opportunities ahead. J Hematol Oncol. 17:1132024. View Article : Google Scholar : PubMed/NCBI | |
|
Lam CS, Peng LW, Yang LS, Chou HWJ, Li CK, Zuo Z, Koon HK and Cheung YT: Examining patterns of traditional Chinese medicine use in pediatric oncology: A systematic review, meta-analysis and data-mining study. J Integr Med. 20:402–415. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Burmistrova O, Quintana J, Díaz JG and Estévez F: Astragalin heptaacetate-induced cell death in human leukemia cells is dependent on caspases and activates the MAPK pathway. Cancer Lett. 309:71–77. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lee KH, Tagahara K, Suzuki H, Wu RY, Haruna M, Hall IH, Huang HC, Ito K, Iida T and Lai JS: Antitumor agents. 49 tricin, kaempferol-3-O-beta-D-glucopyranoside and (+)-nortrachelogenin, antileukemic principles from Wikstroemia indica. J Nat Prod. 44:530–535. 1981. View Article : Google Scholar : PubMed/NCBI | |
|
Delloye-Bourgeois C and Castellani V: Hijacking of embryonic programs by neural crest-derived neuroblastoma: From physiological migration to metastatic dissemination. Front Mol Neurosci. 12:522019. View Article : Google Scholar : PubMed/NCBI | |
|
Gatta G, Botta L, Rossi S, Aareleid T, Bielska-Lasota M, Clavel J, Dimitrova N, Jakab Z, Kaatsch P, Lacour B, et al: Childhood cancer survival in Europe 1999–2007: Results of EUROCARE-5-a population-based study. Lancet Oncol. 15:35–47. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Kholodenko IV, Kalinovsky DV, Doronin II, Deyev SM and Kholodenko RV: Neuroblastoma origin and therapeutic targets for immunotherapy. J Immunol Res. 2018:73942682018. View Article : Google Scholar : PubMed/NCBI | |
|
Berlanga P, Cañete A and Castel V: Advances in emerging drugs for the treatment of neuroblastoma. Expert Opin Emerg Drugs. 22:63–75. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shekarian T and Valsesia-Wittmann S: Considerations for the development of innovative therapies against aggressive neuroblastoma: Immunotherapy and Twist1 targeting. Neuroblastoma-Current State and Recent Updates. Gowda C: InTech; 2017, View Article : Google Scholar | |
|
Luksch R, Castellani MR, Collini P, De Bernardi B, Conte M, Gambini C, Gandola L, Garaventa A, Biasoni D, Podda M, et al: Neuroblastoma (Peripheral neuroblastic tumours). Crit Rev Oncol Hematol. 107:163–181. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lodrini M, Sprüssel A, Astrahantseff K, Tiburtius D, Konschak R, Lode HN, Fischer M, Keilholz U, Eggert A and Deubzer HE: Using droplet digital PCR to analyze MYCN and ALK copy number in plasma from patients with neuroblastoma. Oncotarget. 8:85234–85251. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Johnsen JI, Dyberg C, Fransson S and Wickström M: Molecular mechanisms and therapeutic targets in neuroblastoma. Pharmacol Res. 131:164–176. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zafar A, Wang W, Liu G, Wang X, Xian W, McKeon F, Foster J, Zhou J and Zhang R: Molecular targeting therapies for neuroblastoma: Progress and challenges. Med Res Rev. 41:961–1021. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chung MJ, Lee S, Park YI, Lee J and Kwon KH: Neuroprotective effects of phytosterols and flavonoids from Cirsium setidens and Aster scaber in human brain neuroblastoma SK-N-SH cells. Life Sci. 148:173–182. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Chamoli A, Gosavi AS, Shirwadkar UP, Wangdale KV, Behera SK, Kurrey NK, Kalia K and Mandoli A: Overview of oral cavity squamous cell carcinoma: Risk factors, mechanisms, and diagnostics. Oral Oncol. 121:1054512021. View Article : Google Scholar : PubMed/NCBI | |
|
Tan Y, Wang Z, Xu M, Li B, Huang Z, Qin S, Nice EC, Tang J and Huang C: Oral squamous cell carcinomas: State of the field and emerging directions. Int J Oral Sci. 15:442023. View Article : Google Scholar : PubMed/NCBI | |
|
Katirachi SK, Grønlund MP, Jakobsen KK, Grønhøj C and von Buchwald C: The prevalence of HPV in oral cavity squamous cell carcinoma. Viruses. 15:4512023. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Zhang Y, Xu M and Yang D: Current trends of targeted therapy for oral squamous cell carcinoma. J Cancer Res Clin Oncol. 148:2169–2186. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Arboleda LPA, de Carvalho GB, Santos-Silva AR, Fernandes GA, Vartanian JG, Conway DI, Virani S, Brennan P, Kowalski LP and Curado MP: Squamous cell carcinoma of the oral cavity, oropharynx, and larynx: A scoping review of treatment guidelines worldwide. Cancers (Basel). 15:44052023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma YX, Li XJ, Jin Q, Ji L and Qiao Q: Application of neoadjuvant immunotherapy in locally advanced squamous cell carcinoma of head and neck. Chin J Cancer Prev Treat. 31:968–974. 2024. | |
|
Wang X, Li J, Chen R, Li T and Chen M: Active ingredients from Chinese medicine for combination cancer therapy. Int J Biol Sci. 19:3499–3525. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Qiu H, Li C, Cai P and Qi F: The positive role of traditional Chinese medicine as an adjunctive therapy for cancer. Biosci Trends. 15:283–298. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Miao K, Liu W, Xu J, Qian Z and Zhang Q: Harnessing the power of traditional Chinese medicine monomers and compound prescriptions to boost cancer immunotherapy. Front Immunol. 14:12772432023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao S, Xiao S, Wang W, Dong X, Liu X, Wang Q, Jiang Y and Wu W: Network pharmacology and transcriptomics reveal the mechanisms of FFBZL in the treatment of oral squamous cell carcinoma. Front Pharmacol. 15:14055962024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Cai J, Ding S and Bao S: Network Pharmacology Was Used to Predict the Active Components and Prospective Targets of Paeoniae Radix Alba for Treatment in Endometriosis. Reprod Sci. 30:1103–1117. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu WL, Ji MW, Wang YY, Song LN, Jiang Y and Shao YK: A case report of diffuse large B-cell lymphoma with brachial plexus nerve injury as the first symptom. J Apoplexy Nerv Dis. 37:842–844. 2020. | |
|
Qi RL, Qiu MH, Zhang T, Zhao XZ, He GP, Mei HW and Wang HQ: Progress in treatment of recurrent/refractory diffuse large B-cell lymphoma. Chin Clin Oncol. 27:758–763. 2022. | |
|
Wang Y, Zhu ML, He SY, Chen H and Chen JY: Astragalin induces apoptosis of DLBCL cell line OCI-LY8. Chin J Pathophysiol. 37:885–890. 2021.(In Chinese). | |
|
Lu Y, Wu N, Fang Y, Shaheen N and Wei Y: An automatic on-line 2,2-diphenyl-1-picrylhydrazyl-high performance liquid chromatography method for high-throughput screening of antioxidants from natural products. J Chromatogr A. 1521:100–109. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hadzi-Petrushev N, Stojchevski R, Jakimovska A, Stamenkovska M, Josifovska S, Stamatoski A, Sazdova I, Sopi R, Kamkin A, Gagov H, et al: GLUT5-overexpression-related tumorigenic implications. Mol Med. 30:1142024. View Article : Google Scholar : PubMed/NCBI | |
|
Thompson AM, Iancu CV, Nguyen TTH, Kim D and Choe J: Inhibition of human GLUT1 and GLUT5 by plant carbohydrate products; insights into transport specificity. Sci Rep. 5:128042015. View Article : Google Scholar : PubMed/NCBI | |
|
Ingram DK and Roth GS: Glycolytic inhibition: An effective strategy for developing calorie restriction mimetics. Geroscience. 43:1159–1169. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Harikrishnan H, Jantan I, Alagan A and Haque MA: Modulation of cell signaling pathways by Phyllanthus amarus and its major constituents: Potential role in the prevention and treatment of inflammation and cancer. Inflammopharmacology. 28:1–18. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Devanaboyina M, Kaur J, Whiteley E, Lin L, Einloth K, Morand S, Stanbery L, Hamouda D and Nemunaitis J: NF-κB signaling in tumor pathways focusing on breast and ovarian cancer. Oncol Rev. 16:105682022. View Article : Google Scholar : PubMed/NCBI | |
|
Adebayo IA, Balogun WG and Arsad H: Moringa oleifera: An apoptosis inducer in cancer cells. Trop J Pharm Res. 16:2289–2296. 2017. View Article : Google Scholar | |
|
Yang F, Li AM, Zhang YW, Ma CM and Zhang HR: Study on the effect of astragalin on apoptosis of human colon cancer SW480 cells. Guo Ji Yi Yao Wei Sheng Dao Bao She. 26:3013–3017. 2020.(In Chinese). | |
|
Karna KK, Choi BR, You JH, Shin YS, Cui WS, Lee SW, Kim JH, Kim CY, Kim HK and Park JK: The ameliorative effect of monotropein, astragalin, and spiraeoside on oxidative stress, endoplasmic reticulum stress, and mitochondrial signaling pathway in varicocelized rats. BMC Complement Altern Med. 19:3332019. View Article : Google Scholar : PubMed/NCBI | |
|
Yun W, Dan W, Liu J, Guo X, Li M and He Q: Investigation of the mechanism of traditional Chinese medicines in angiogenesis through network pharmacology and data mining. Evid Based Complement Alternat Med. 2021:55399702021. View Article : Google Scholar : PubMed/NCBI | |
|
Riaz A, Rasul A, Hussain G, Zahoor MK, Jabeen F, Subhani Z, Younis T, Ali M, Sarfraz I and Selamoglu Z: Astragalin: A bioactive phytochemical with potential therapeutic activities. Adv Phar Sc. 2018:1–15. 2018. | |
|
Lee JH, Saxena A and Giaccone G: Advancements in small cell lung cancer. Semin Cancer Biol. 93:123–128. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou SB and Chen GZ: Research overview and reflection on the treatment of cancer-related fatigue in lung cancer by traditional Chinese medicine. Acta Chin Med Pharm. 52:1–4. 2024.(In Chinese). | |
|
Adelstein DJ, Moon J, Hanna E, Giri PGS, Mills GM, Wolf GT and Urba SG: Docetaxel, cisplatin, and fluorouracil induction chemotherapy followed by accelerated fractionation/concomitant boost radiation and concurrent cisplatin in patients with advanced squamous cell head and neck cancer: A Southwest oncology group phase II trial (S0216). Head Neck. 32:221–228. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Andreyev HJN, Davidson SE, Gillespie C, Allum WH and Swarbrick E; British Society of Gastroenterology; Association of Colo-Proctology of Great Britain Ireland; Association of Upper Gastrointestinal Surgeons; Faculty of Clinical Oncology Section of the Royal College of Radiologists, : Practice guidance on the management of acute and chronic gastrointestinal problems arising as a result of treatment for cancer. Gut. 61:179–192. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Bower JE: The role of neuro-immune interactions in cancer-related fatigue: Biobehavioral risk factors and mechanisms. Cancer. 125:353–364. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
de Raaf PJ, Sleijfer S, Lamers CHJ, Jager A, Gratama JW and van der Rijt CCD: Inflammation and fatigue dimensions in advanced cancer patients and cancer survivors: an explorative study. Cancer. 118:6005–6011. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wen YL, Li WY, Zhang N, Su RZ, Xia JR, Li LF and Tian Y: The inhibitory effect of astragalin on escherichia coli and its mechanism. J Yunnan Agricultural University. 37:471–477. 2022. | |
|
Oraibi AI, Dawood AH, Trabelsi G, Mahamat OB, Chekir-Ghedira L and Kilani-Jaziri S: Antioxidant activity and selective cytotoxicity in HCT-116 and WI-38 cell lines of LC-MS/MS profiled extract from Capparis spinosa L. Front Chem. 13:15401742025. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Ma B, Zhang Q, Wu X, Gu G, Li J, Sun J, Tang B, Zhu J, Qia H and Ying H: Comparative pharmacokinetics with single substances and Semen Cuscutae extract after oral administration and intravenous administration Semen Cuscutae extract and single hyperoside and astragalin to rats. Anal Methods. 6:72502014. View Article : Google Scholar : PubMed/NCBI | |
|
Adamu UM, Renggasamy R, Stanslas J, Razis AF, Fauzi F, Mulyani SW and Ramasamy R: In-silico prediction analysis of polyphenolic contents of ethanolic extract of moringa oleifera leaves. Malays J Med Health Sci. 19:9–15. 2023. | |
|
Liu J, Zou S, Liu W, Li J, Wang H, Hao J, He J, Gao X, Liu E and Chang Y: An Established HPLC-MS/MS method for evaluation of the influence of salt processing on pharmacokinetics of six compounds in cuscutae semen. Molecules. 24:25022019. View Article : Google Scholar : PubMed/NCBI | |
|
Choung WJ, Hwang SH, Ko DS, Kim SB, Kim SH, Jeon SH, Choi HD, Lim SS and Shim JH: Enzymatic synthesis of a novel Kaempferol-3-O-β-d-glucopyranosyl-(1→4)-O-α-d-glucopyranoside using cyclodextrin glucanotransferase and its inhibitory effects on aldose reductase, inflammation, and oxidative stress. J Agric Food Chem. 65:2760–2767. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ren C, Xi Z, Xian B, Chen C, Huang X, Jiang H, Chen J, Peng C and Pei J: Identification and characterization of CtUGT3 as the key player of astragalin biosynthesis in Carthamus Tinctorius L. J Agric Food Chem. 71:16221–16232. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Pei J, Dong P, Wu T, Zhao L, Fang X, Cao F, Tang F and Yue Y: Metabolic engineering of Escherichia Coli for astragalin biosynthesis. J Agric Food Chem. 64:7966–7972. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Lei X, Xu H, Liu G, Wang Y, Sun H, Geng F and Zhang N: Tissue distribution of total flavonoids extracts of drynariae rhizoma in young and old rats by UPLC-MS/MS determination. J Anal Methods Chem. 2022:1–30. 2022. View Article : Google Scholar | |
|
Li N, Lv T, Pan J, Liu C, Sun J, Lan Y, Wang A, Li Y, Wang Y and Lu Y: Comparative tissue distribution of 6 major polyphenolic compounds in normal and myocardial ischemia model rats after oral administration of the Polygonum orientale L. Extract. Nat Prod Commun. 15:1934578X209294472020. | |
|
He B, Bai X, Tan Y, Xie W, Feng Y and Yang GY: Glycosyltransferases: Mining, engineering and applications in biosynthesis of glycosylated plant natural products. Synth Syst Biotechnol. 7:602–620. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Han YM, Koh J, Kim JH, Lee J, Im JP and Kim JS: Astragalin inhibits nuclear Factor-κB signaling in human colonic epithelial cells and attenuates experimental colitis in mice. Falk Symp. 15:100–108. 2021. |