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 recent years, the incidence of new global cancer cases has been consistently increasing. As indicated in the World Health Organization report, malignant neoplasms are among the predominant causes of mortality worldwide, and it is estimated that the global population of patients with cancer will reach 28.4 million by 2040 (1). The onset and progression of malignant tumors are closely associated with the tumor microenvironment, which comprises tumor cells, immune cells, fibroblasts, the extracellular matrix and a variety of cytokines that enhance the proliferation, migration and immune evasion capabilities of tumor cells (2). As research into the tumor microenvironment has increased, tumor immunotherapy has emerged as a novel and promising therapeutic modality for the management of cancer. This type of therapy uniquely activates the immune microcirculation, thereby eliciting a targeted immune response against tumor cells and facilitating their destruction. Traditional Chinese Medicine (TCM) has been used in the treatment of tumors, with an emphasis on individualized patient care (3). Throughout the course of tumor progression, TCM has demonstrated efficacy in bolstering immunity, thereby restoring and maintaining normal immune functions, preventing the invasion of exogenous pathogens and mitigating the detrimental effects of tumor cells. These principles are consistent with the modern medical concept of tumor immunotherapy. Astragalus polysaccharide (APS) can modulate various immune cells, including macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), T lymphocytes and natural killer (NK) cells, thereby altering their functional activities and enhancing immune responsiveness. Consequently, the tumor immune microenvironment and the cytotoxic capability of immune cells against tumor cells are improved (4). In the present study, research on the antitumor immune properties of APS has been comprehensively reviewed, and its regulatory effects on immune cells and the underlying antitumor mechanisms are discussed, thereby providing a theoretical foundation for the development of novel Astragalus-based antitumor pharmacological agents.
Huangqi (Jianqi) is the dried root of the leguminous plant Astragalus mongholicus or Astragalus membranaceus (5). As a common traditional Chinese herbal medicine, Astragalus has a history of more than 2,000 years of use in China, is used extensively in numerous countries and has been included in the pharmacopeias of the United States, Japan, and South Korea (6). The constituents of Astragalus encompass a variety of compounds such as APS, triterpenoid saponins, alkaloids, flavonoids and different mineral elements (7–10). APS, which serves as the principal active constituent within Astragalus, is characterized by its copious availability, affordability, and minimal toxic or adverse effects (11). The categorization of polysaccharides is performed in accordance with the structural classification principles employed for proteins and DNA. Investigating the constituent monosaccharides, relative molecular mass and sequential arrangement of monosaccharide residues is necessary to elucidate the structure of APS (12). APS is classified as an acidic complex polysaccharide, predominantly comprising glucose (Glc), galactose (Gal), arabinose (Ara), rhamnose (Rha) and mannose (Man). Infrared spectroscopic analysis has revealed that APS possesses the typical infrared absorption characteristics of sugar, along with distinct signals that are attributed to the carboxyl group, indicating the presence of glucuronic acid. The signals corresponding to sugar rings indicate that APS constitutes a pyranose ring structure with α and β configurations (13). Glucose is a monosaccharide based on a glucopyranose structure, including α-D-Glu, β-D-Glu and α, β-D-Glu, with β-D-Glu being the most extensively investigated monosaccharide (14).
Given the presence of numerous branched chains and a number of hydroxyl groups, APS exhibits a high degree of structural diversity and complexity (Table I). APS MAPS-5 is composed of an α-D-(1–4) Glc backbone, where, on average, approximately two out of every 15 sugar residues at the C-6 position are substituted with terminal glucose residues, with an approximate relative molecular weight of 1.32×104 (15). Furthermore, three distinct polysaccharides, namely APS I, II and III, have been isolated from the aqueous extract of Astragalus roots sourced from Inner Mongolia. APS I is a heteropolysaccharide composed of D-Glu, D-Gal, and L-Ara in a molar ratio of 1.75:1.63:1, with an average relative molecular mass of approximately 3.63×104. APS II and III are predominantly composed of D-Glu, with average relative molecular weights of ~1.23×104 and 3.46×104, respectively. Their structure primarily comprises α-(1→6)-D-Glc condensation linkages, with a minor fraction of α-(1→6)-D-Glc condensation bonds (16). Through complete acid hydrolysis, four types of APS have been differentiated, including two glucans (AG-1 and AG-2) and two heteropolysaccharides (AH-1 and AH-2) (17). AG-1 is a water-soluble glucan, whereas AG-2 is water insoluble and is characterized as an α-(1→4) glucan. AH-1 is an acidic, water-soluble polysaccharide; after hydrolysis, AH-1 contains hexuronic acids (galacturonic and glucuronic acids), glucose, rhamnose and arabinose, as identified through paper chromatography, with a molar ratio of 1:0.04:0.02:0.01. AH-2 is also water soluble, and the presence of glucose and arabinose can be confirmed via paper and gas chromatography post-hydrolysis, with a molar ratio of 1:0.15 (17).
Tang et al (18) extracted and isolated polysaccharides from Astragalus, and characterized their physicochemical properties and structure. This previous study revealed that the composition of monosaccharides in APS-I was mannose (Man), rhamnose (Rha), Glc uronic acid (Glc UA), Gal UA, Glc and Gal, with a substance amount ratio of 29.12:1.89:4.00:1.35:1:81.97, whereas the composition of monosaccharides in APS-II was Man, Rha, Glc UA, Gal UA, Glc, Gal and xylose, with a substance amount ratio of 50.46:1.16:1:2.27:2.66:15.72:7.86. The relative molecular mass of APS-I was ~1.06×104, and the relative molecular mass of APS-II was ~2.47×106. Cao et al (19) extracted from the experiment that the total polysaccharide content of APS was 74.6%, the protein content was 0.42%, and the relative molecular weight distribution was 300–2×106. In addition, its monosaccharide composition was Rha, Gal UA, Glc, Gal and Ara, with a substance amount ratio of 0.43:0.23:19.36:0.69:1. Furthermore, a previous study characterized the monosaccharide group of two polysaccharides obtained from the isolation and purification of Astragalus membranaceus from Mongolia using ion chromatography. The results showed that APS I was composed of Gal and Glc, with a substance amount ratio of 1:49.76, whereas APS II mainly consisted of Rha, Gal and Glc, with a substance amount ratio of 1:2.99:16.26 (20). Although a number of technical methods have been used to analyze APS, the specific molecular structure of APS remains difficult to elucidate, which results in certain obstacles in the study of the molecular mechanism underlying APS bioactivity.
As academic research on tumor immunotherapy continues, the successful development of immune checkpoint inhibitors that target PD-1, CTLA-4 and PD-L1 has led to the gradual emergence of immunotherapy as a potential method to treat tumors (21). Body immunity is divided into active and passive immunity. Tumor cells evade elimination by the immune system, which not only promotes the state of immunosuppression and the production of regulatory immune cells, but also recruits a large number of protumor cells to establish a tumor microenvironment (22). In the tumor microenvironment, tumor cells inhibit the antigen presentation function of DCs; promote the polarization of tumor-associated macrophages from M1 type to M2 type; release the immune-suppressive factors TGF-β, VEGF and IL-10; deliver the abnormally expressed tumor-associated antigens to T cells; induce T-cell apoptosis; and inhibit the function of cytotoxic T lymphocytes, which together affect the immune system and result in tumor immune escape and the acceleration of tumor progression (23–26). In determining the mechanism underlying tumor immune escape, current immunotherapeutic drugs, including drugs that block negative regulatory signals, immunostimulatory drugs, tumor vaccines and exogenous recombinant cytokines, have been used (27), among which immune checkpoint inhibitors, such as anti CTLA-4 and anti-PD-1/PD-L1 antibodies, have shown good therapeutic effects in clinical tumor therapy. A number of active ingredients of TCM have shown effects on regulating immune cells and improving the tumor microenvironment, indicating their therapeutic potential in synergistic radiotherapy and chemotherapy against tumors (28).
APS, as a main active ingredient of Astragalus, has important pharmacological effects, including immune-regulatory and antitumor properties, which can regulate the microenvironment of numerous solid tumors, improve the state of immune suppression and inhibit tumor growth. APS inhibits the M2-type polarization of tumor-associated macrophages, increases cytotoxic T lymphocyte infiltration, and promotes the maturation of DCs and the function of NK cells (29). In addition, APS not only reduces the release of the immunosuppressive factors IL-10, TGF-β and VEGF in the tumor microenvironment, but also increases the levels of the immune activating factors TNF-α, IL-2 and IL-12 (30). In clinical trials for patients with cancer, APS has been shown to relieve pain, nausea and vomiting symptoms, enhance immune function, reduce the toxic side effects of chemotherapeutic drugs and improve quality of life (31) (Table II; Figs. 1 and 2).
Macrophages develop from bone marrow precursor cells and serve an important role in the immune system. They are present in almost all tissues of the body, and they directly phagocytose and kill foreign pathogens. In addition, they can activate lymphocytes or other immune cells in the body to indirectly respond to pathogens, and protect tissues and organs from foreign pathogens (32). Macrophages are classified into M1 and M2 types. M1 macrophages, as classically activated macrophages, produce nitric oxide (NO), reactive oxygen species (ROS), inducible NO synthase and a large number of pro-inflammatory cytokines, such as IL-1β, interferon (IFN)-γ and TNF-α, which participate in the positive immune response. M1 macrophages also have a role in phagocytosis and tumor cell killing (33).
By contrast, M2 macrophages, as alternatively activated macrophages, produce a large number of anti-inflammatory cytokines, such as IL-4, IL-10 and IL-13, which suppress the immune response to a certain extent. Tumor cells selectively recruit bone marrow-derived macrophages to differentiate into M2 macrophages, that is, tumor-associated macrophages (34), which promote tumor cell proliferation, neovascularization, invasion and metastasis (35). Adjustment of M1/M2 macrophage polarization is key to improve the effectiveness of antitumor therapy. In an experimental study in mice with non-small cell lung cancer (NSCLC), APS modulates the Notch signaling pathway, promotes the production of IL-6, TNF-α and iNOS cytokines, activates M1 macrophages, inhibits macrophage M2-type polarization, and enhances phagocytosis and tumor cell killing (36). Bioinformatics analysis indicates that APS may remodel the tumor microenvironment (TME) and influence cell-cell interactions, specifically through modulation of macrophage M2 polarization and CD8+ T cell exhaustion, thereby overcoming DDP resistance (37). Li et al (38) reported that APS can increase the proportion of M1 macrophages in hepatocellular carcinoma cell-derived xenograft tumors, reduce macrophage M2-type polarization and inhibit the proliferation of hepatocellular carcinoma tumor cells. Furthermore, APS improves the energy metabolism of mouse macrophages in vivo, and enhances the phagocytic activity of RAW 264.7 monocyte macrophages and the immune response in mice (39,40). Mouse macrophages activated by APS are triggered through the Toll-like receptor (TLR)4-mediated signaling pathway; this activation event results in the upregulation of the levels of phosphorylated (p)-p38, p-ERK1/2 and p-JNK. Concurrently, it induces the degradation of IκB-α and the subsequent translocation of NF-κB. Eventually, these molecular events culminate in the production of TNF-α, IL-6 and NO, thereby exerting an inhibitory effect on the proliferation of mouse tumor cells (41,42). Li et al (43) demonstrated that APS enhances the proliferation and phagocytic functions of mouse macrophages, increases the peripheral blood levels of IL-2, TNF-α and IFN-γ, and improves the ability of macrophages to phagocytose tumor cells and inhibit the proliferation of tumor cells. Therefore, APS may enhance macrophage phagocytosis and tumor cell killing, and inhibit tumor cell proliferation and metastasis by increasing M1 macrophage activity, regulating M1/M2-type macrophage polarization and altering the levels of immune cytokines.
DCs are derived from bone marrow CD34+ hematopoietic stem cells and are a specialized class of antigen-presenting cells, which are divided into immature DCs (imDCs), mature DCs (mDCs) and regulatory DCs, in accordance with the different stages of cell differentiation (44). ImDCs capture tumor antigens and are stimulated to differentiate into mature mDCs. The mDCs migrate to draining lymph nodes via lymphatic vessels, where they present the antigens to T cells. They secrete various co-stimulatory molecules such as CD80, CD86, CD40 and CD70, which further activate effector T cells, thereby inducing and maintaining anti-tumor immunity (45,46). Enhancing the function of DCs and promoting T-cell activation in patients with cancer are an effective means of antitumor immunotherapy. APS promotes DC maturation, enhances DC antigen-presenting ability, improves the accuracy of tumor cell killing and inhibits tumor cell proliferation (47). When DCs are induced to mature further, the expression of the DC surface co-radical molecules CD80 and CD86 are enhanced. DCs present antigens to T lymphocytes, T lymphocytes increase IL-12 and IFN-γ secretion, improve antitumor immunity and inhibit the proliferation of tumor cells (48).
APS induces the DC cell surface molecule CD80, CD86 and IL-12 expression through the TLR2/4 receptor-mediated MyD88 signaling pathway, increases T lymphocyte activity, releases more IL-2 and enhances antitumor ability (49,50). APS nanoparticles have been shown to modulate the TLR-mediated signaling pathway to activate DCs, to increase the expression levels of co-stimulatory molecules on the surface of DCs and enhance their tumor cell antigen-presenting ability. Subsequently, DCs migrate rapidly to lymph nodes, activate the initial T lymphocytes at lymph nodes and inhibit secondary lymph node tumor growth (51). Furthermore, after APS treatment, DCs have been shown to promote the proliferation of CD4+ and CD8+ T cells in mice with breast cancer, and a DC-based antitumor vaccine can increase the expression levels of CD40, CD80 and CD86 molecules on the surface of DCs, and inhibit the growth and metastasis of breast cancer cells (52). APS induces morphological changes, the expression of activation markers and the upregulation of inflammatory cytokines in human blood monocyte-derived DCs. In addition, APS promotes the activation of blood DC antigen (BDCA)1 and BDCA3 peripheral blood dendritic cells (pBDCs), which results in the proliferative activation of T cells (53). Therefore, APS may induce the activation of DCs, increased the expression levels of co-stimulatory factors on the surface of DCs, and accelerate the maturation of DCs and antigen presentation. Subsequently, this may enhance the proliferation and activation of CD4+ and CD8+ T cells, and thus the activation of immune function under the stimulation of cytokines, thereby effectively inhibiting the proliferation and metastasis of tumor cells.
MDSCs are a group of heterogeneous cells derived from the bone marrow; the number of MDSCs in the peripheral blood and spleen is small, accounting for only 4% of cells (54). Under pathological conditions, these immature myeloid-derived heterogeneous cells stop differentiating and become immunosuppressive MDSCs (55,56). MDSCs promote tumor cell survival, angiogenesis, and invasion and metastasis to healthy tissues.
APS inhibit MDSC activity, promote MDSC differentiation and reduce MDSC population. The underlying mechanisms represent a current research hotspot in immunotherapy (51). APS suppress MDSC generation and reduce the MDSC population in the spleens of tumor-bearing mice. Concurrently, APS downregulates immunosuppressive cytokines (IL-10, TGF-β) while upregulating pro-inflammatory cytokines (IFN-γ, TNF-α), thereby enhancing overall immune function (57). Ding et al (58) revealed that APS exert a marked regulatory effect on gut microbiota. APS effectively remodels the gut microbiota environment, and elevates the levels of glutamate and creatine metabolites. Notably, glutamate and creatine serve crucial roles in controlling tumor growth. Simultaneously, APS contributes to the reduction in the number of MDSCs. Furthermore, in mice with melanoma, APS has been shown to curtail the immunosuppressive activity of MDSCs, as manifested by decreased expression levels of Arg-1, IL-10 and TGF-β; this series of changes bolsters the cytotoxic capacity of CD8+ T cells against tumor cells. Consequently, this previous study demonstrated that the tumor inhibition rate in mice with melanoma was significantly increased, highlighting the potential of APS in antitumor regulation. APS has been reported to effectively reduce the level of myeloid-derived suppressor cells in the peripheral circulation of lung cancer, continuously improve immune function, and increase the overall clinical treatment efficacy (59). As a common malignant tumor, lung cancer has shown an increasing trend in morbidity and mortality in recent years (60). Multi-targeted, low-toxicity Chinese medicines intervene in the ‘pre-metastatic niche’, a potentially effective means of preventing and treating tumor metastasis. APS has been reported to inhibit the formation of the lung pre-metastatic niche and suppress the recruitment of lung MDSCs. Mechanistically, the expression of S1PR1, STAT3 and p-STAT3 in the S1PR1/STAT3 signaling pathway have been shown to be inhibited by APS, which interferes with the S1PR1/STAT3 signaling pathway and inhibits the accumulation of MDSCs in the pre-metastatic niche to achieve antitumor effects (61). Therefore, APS promote the rapid differentiation of MDSCs, reduce the number and activity of MDSCs, influence the release of immune cytokines, enhance immune functions, and inhibit the proliferation and metastasis of tumor cells.
T lymphocytes develop from bone marrow pluripotent stem cells, differentiate and mature under the induction of thymic hormone, and become immunologically active T cells, playing antitumor specific immunity. T lymphocytes are classified into CD4+ T and CD8+ T cells in accordance with their surface molecular types. CD4+ and CD8+ are the two functionally different subpopulations of T lymphocytes. CD4+ T cells are also known as helper T cells, which play a role in assisting the induction of body immunity. Th cells are differentiated into functional subpopulations such as Th1 cells, Th2 cells, and Th17 cells. The main functional subpopulation of CD8+ T cells is cytotoxic T lymphocytes. Th1 cells secrete IL-2 and TNF-α and stimulate the development of cytotoxic T lymphocytes. TNF-α, which stimulates the activation of macrophages and NK cells, also secretes IFN-γ to kill tumor cells. Th2 cells secrete IL-4, IL-6, and IL-10, which assist B lymphocytes to produce specific antibodies and participate in humoral immunity. Under normal circumstances, the number of Th1 and Th2 cells remains relatively balanced. However, in patients with tumor, a transformation from Th1 cells to Th2 cells occurs, which suppresses the body's cellular immunity. APS enhances the expression of Th1 cytokines such as IL-2 and IFN-γ in the spleen tissues of mice with lung cancer through the TLR4/MyD88/NF-κB signaling pathway while reducing the expression of Th2 cytokines, including IL-4 and IL-10. It also regulates the ratio of Th1/Th2 cells, reverses the transformation from Th1 to Th2, improves the immunity of mice with lung cancer, and inhibits the growth of tumor cells (62). APS not only improves the immune organ index in mice with lung cancer, but also increases the ratio of CD4+/CD8+ T cells in mouse serum and enhances cellular immune activity (63,64). APS protects immune organs, regulates the ratio of CD4+/CD8+ T lymphocyte subpopulations, and inhibits the growth of mouse tumors in vivo; APS also activates antitumor immune cells, promotes the tumor microenvironment of anaerobic metabolism, and effectively induced apoptosis in tumor cells (65). APS increases the number of allogeneic T lymphocytes and the expression level of IL-12 and IFN-γ. In addition, cytotoxic T lymphocytes activated by sensitized DCs kill tumor cells (66). APS reverses Th1-to-Th2 conversion, maintains Th1/Th2 cell balance, regulates CD4+/CD8+ T cell ratio and T cell-related cytokine expression, enhances cellular immunity, and inhibits tumor growth.
Tregs are a group of T lymphocytes that can regulate the immune response and maintain immune tolerance. Tumor immunotherapy reduces the number of Tregs, inhibits their cellular activity, maintains immune function of the body and achieves the killing effects against tumor cells. DCs are a key target for Tregs to inhibit the tumor immune response (67), and Tregs interact with tumor-associated CD11c+ DCs to reduce the expression of DC co-stimulatory ligands and inhibit T-cell proliferation. Conversely, the clearance of Tregs leads to the restoration of CD11c+ DC immunogenicity, co-stimulatory ligand expression and CD8+ T-cell activation, which in turn inhibits tumor growth (68).
APS has been reported to reduce the number of Tregs, decrease the expression levels of splenic TGF-β and IL-10, and inhibit the proliferation and metastasis of malignant melanoma tumor cells in mice (69). APS also repairs cytokine imbalance, inhibits forkhead box protein P3 expression, and suppresses Treg activity and function (70). Through the CXCR4/CXCL12 signaling pathway, APS has been shown to inhibit the recruitment of Tregs by stromal cell-derived factor-1, and to attenuate the proliferation and migration of Tregs. Furthermore, APS activates the TLR4-mediated signaling pathway, thereby inhibiting the expression of TGF-β, decreasing the number of Tregs and sustaining the killing effect of immune cells on tumor cells (71). APS significantly enhanced the PBMC proliferation, reduced Treg frequency, decreased anti-inflammatory cytokines including IL-10, TGF-β, and VEGF-A, and increased the pro-inflammatory cytokine IL-6 (72). Therefore, APS may inhibit Treg activity by reducing the number of Tregs and their cytokine secretion, thus restoring and maintaining the normal immune function of T lymphocytes, achieving the killing effect of T cells on tumor cells, and inhibiting tumor growth and metastasis.
NK cells are derived from bone marrow lymphoid stem cells, which are important members of the lymphocyte family, and they are not controlled by high-resolution antigen specificity. NK cells can rapidly recognize and remove heterogeneous cells, and the degree of attenuation in vivo is closely related to the malignant progression of tumors (73). NK cells secrete cytokines, regulate adaptive immunity and kill tumor cells, and they have killing and inhibitory effects on hepatocellular carcinoma, NSCLC, breast cancer and ovarian cancer cells (74–77). APS has been reported to inhibit the proliferation of leukemia cells by activating MHC class I polypeptide-related sequence A molecules on the surface of leukemia cells, and enhances the sensitivity of HL-60 cells to NK cell killing activity and inhibit leukemia cell growth (78). Low-molecular-weight APS can protect the immune organs of mice with hepatocellular carcinoma and stimulate NK cells to kill tumor cells. In addition, APS promotes the proliferation of splenic lymphocytes, increases the activity of NK cells and enhances the immune response of rats with cancer (79,80).
Tumor cell proliferation primarily aggravates cancer progression, and pharmacological intervention in tumor cell proliferation serves an important role in tumor therapy. F-box and WD-40 structural domain protein 7 (FBXW7) is a classical tumor suppressor, the translation of which is inhibited by microRNA (miR)-27a in ovarian cancer cells. APS inhibits the proliferation of ovarian cancer cells through the miR-27a/FBXW7 signaling pathway, revealing the therapeutic potential of APS for ovarian cancer (81). Lung cancer is the second most common and the deadliest type of cancer worldwide. Clinically, non-small cell lung cancer (NSCLC) is the most common pathological type of lung cancer (82). APS can inhibit the proliferation and migration of NSCLC cells, and its mechanism of action is related to the regulation of the miR-195-5p signaling pathway; in addition, APS inhibits the lung cancer cell cycle and improves the morphology of lung cancer cells (83,84). APS inhibit TGF-β1-induced epithelial-mesenchymal transition (EMT) in A549/DDP cell xenografts of lung adenocarcinoma, which is associated with inhibition of PI3K/Akt pathway protein activation (85). In a study exploring the synergistic inhibitory effect of APS and cisplatin on the growth of human carcinoma nasopharyngeal cell (CNE), APS was shown to exert an inhibitory effect on the proliferation of nasopharyngeal carcinoma cells in a time- and dose-dependent manner, and the combined application of APS and cisplatin could inhibit the migration and invasion of CNE-1 cells, which was more effective than the use of either drug alone with regard to the antitumor effect of either drug (86). In addition, the study demonstrated that APS at varying concentrations inhibited the proliferation of human colorectal cancer SW620 cells, inducing SW620 cell cycle arrest predominantly at the G2/M phase (87). In addition, APS can markedly inhibit the proliferation and invasion of prostate cancer (PCa) cells in a time- and dose-dependent manner. Under APS treatment, the cellular triacylglycerol and cholesterol levels of patients were revealed to be markedly reduced, and APS can regulate the miR-138-5p/SIRT1/SREBP1 pathway in PCa to inhibit lipid metabolism and tumorigenesis (88). Furthermore, APS enhances the expression of miR-133a in MG63 osteosarcoma cells, inhibits the JNK signaling pathway, the proliferation, migration and invasion of MG63 cells, and induces the apoptosis of MG63 cells (89).
Apoptosis is a type of programmed cell death regulated by various genes, which serves an important role in tumorigenesis and development (90). The occurrence of malignant tumors is due to the uncontrolled and excessive proliferation of tumor cells. In recent years, promoting the apoptosis of tumor cells has become a focus of research for the treatment of tumors. There are two main apoptotic pathways: i) The extrinsic pathway, which is mediated by the activation of caspases through the cell membrane death receptor; and ii) the intrinsic pathway, which is mediated by mitochondria in the cytoplasm that release apoptotic factors and activate caspases. These activated caspases degrade the key proteins in the cell, causing apoptotic cell death. The genes and proteins that have been studied include caspase-3, caspase-8, caspase-9, the Bcl-2 family and cyclooxygenase-2 (91). In the mitochondrion-mediated intrinsic apoptotic pathway, proteins belonging to the Bcl-2 family alter mitochondrial membrane permeability, regulate cytochrome c release and modulate the apoptotic process (92). Bax and Bcl-2 belong to the Bcl-2 superfamily, which are key genes in the regulation of apoptosis, and they are in a balanced state in normal organisms. Notably, apoptosis can be induced by a decrease in the expression levels of the apoptosis inhibitory protein Bcl-2, or an increase in the expression levels of the pro-apoptotic protein Bax (93).
APS markedly promotes the anti-proliferative and apoptotic effects of cisplatin on nasopharyngeal carcinoma cells, by regulating the expression levels of Bax/Bcl-2, caspase-3, and caspase-9 (94). APS also markedly decreases gastric cancer (GC) cell viability, and accelerates GC cell apoptosis in a time- and dose-dependent manner; these effects are associated with an increase in p-AMPK. APS-treated MGC-803 cells have been shown to exhibit the typical morphological features of apoptosis, alongside cell cycle arrest at the S phase (95). A novel cold-water-soluble polysaccharide (APS4) was isolated from Astragalus membranaceus, induce mitochondria-dependent apoptosis, which is associated with intracellular ROS accumulation, mitochondrial membrane potential imbalance, increased pro-apoptotic/anti-apoptotic Bax/Bcl-2 ratio and cytochrome c release (96).
The Notch signaling has an important role in the transformation and proliferation of human malignant tumors, and it is readily activated in human non-small cell lung carcinoma, whereas the inhibitors of Notch signaling can attenuate cell proliferation and induce apoptosis in lung cancer cell lines. APS has been shown to inhibit the expression of Notch1 and Notch3 in NSCLC cell lines to upregulate the pro-apoptotic Bax and caspase 8, promoting apoptosis in NSCLC cell lines (97-101). APS may also inhibit tumor cells of A549, MC38 and B16, induce apoptosis and prevent cell transformation from the G1 phase to the S phase of the cell cycle, and is associated with the expression of molecules related to immunogenic cell death and the regulation of immune cells (102). APS has also been reported to decrease the number of HepG2 liver cancer cells in S-phase, and cells were arrested at the G0-G1 and G2-M phases, block the cellular proliferation cycle and induce apoptosis in cancer cells (103,104). Wang (105) investigated the role of ERK1/2 in the promotion of apoptosis in HepG2 cells by APS and found that caspase-3 activity is increased, ERK1/2 expression is decreased and APS inhibits the ERK1/2 signaling pathway to promote apoptosis in tumor cells.
Protein O-GlcNAc modification is a unique post-translational modification, and various nuclear and cytoplasmic proteins can be modified by the O-GlcNAcylation of free hydroxyl groups of selected serine and threonine residues (106). The cell modification cycle is mediated by O-GlcNAc transferase, which transfers N-acetylglucosamine to the protein substrate, and the O-GlcNAcase enzyme, which removes this modification from the protein (107–109). O-GlcNAcylation affects a wide range of protein functions, including transcription, subcellular localization, protein-protein interactions and protein stability (110). In numerous types of cancer, O-GlcNAc modification is associated with endoplasmic reticulum (ER) stress, and reduced O-GlcNAcylation leads to the activation of the ER stress response in a variety of cancer cells (111–113). APS promotes doxorubicin (Dox)-induced apoptosis and ER stress response in a dose-dependent manner. In addition, APS decreases O-GlcNAc transferase mRNA levels and protein stability. and increases O-GlcNAcase expression level. APS thus reduces O-GlcNAcylation and promotes Dox-induced apoptosis in hepatocellular carcinoma cells, leading to the exacerbation of ER stress and the activation of apoptotic pathways (114).
Invasion and metastasis are crucial biological characteristics of malignant tumors, as well as important factors contributing to tumor recurrence following surgery. Tumor cells disseminate to new organs and tissues through the blood and lymphatic systems, subsequently proliferating. Notably, tumor vasculature serves as a conduit for metastasizing tumor cells; therefore, inhibiting tumor cell angiogenesis can impede the progressive deterioration of tumors and aid in the treatment of neoplastic diseases. Vascular endothelial growth factor (VEGF) is widely recognized as a pivotal mediator of tumor angiogenesis (115). APS effectively inhibits tumor cell metastasis, with APS efficiently suppressing Lewis lung cancer growth and metastasis in mice while enhancing immune organ function. Moreover, it suppresses the protein expression levels of VEGF and EGFR in tumor tissues, exhibiting a concentration-dependent relationship (116). The NF-κB signaling pathway has a critical role in lung cancer prevention and treatment; APS significantly downregulates NF-κB transcriptional activity, thereby inhibiting the proliferation and metastasis of A549 and NCI-H358 NSCLC cells (117,118). Furthermore, the angiogenesis of human colorectal cancer tumors is markedly inhibited by APS, which is closely associated with the reduction in VEGF expression within tumor tissues (119). APS can inhibit the proliferation and invasion of HS-746T cells, suppress the expression of miR-25 mRNA, and promote the expression of FBXW7 mRNA. It can also reverse the increase in miR-25 and the suppression of FBXW7 caused by transfection with miR-25 mimic. APS can inhibit the proliferation and invasion of gastric cancer HS-746T cells through the miR-25/FBXW7 signaling pathway (120). miR-133, which serves as a tumor suppressor, inhibits proliferation, migration in tumor cells and promotes their apoptosis. APS has been shown to upregulate the expression levels of miR-133a, promote the inactivation of the JNK signal transduction pathway, and inhibit the proliferation, metastasis and invasion of MG63 osteosarcoma cells while inducing apoptosis (89,121). EMT serves a crucial role in tumor migration and invasion. APS can serve as a therapeutic agent for breast cancer by inhibiting the Wnt/β-catenin signaling pathway to reduce proliferation and the EMT-mediated migration and invasion of breast cancer cells (122,123). Moesin (MSN) serves as a potential biomarker associated with tumor invasion and metastasis while maintaining PD-L1 stability. In hepatocellular carcinoma, MSN is targeted by miR-133a-3p, which negatively regulates its expression; notably, APS attenuates PD-L1-mediated immunosuppression by modulating the miR-133a-3p/MSN pathway (124).
Under physiological conditions, free radicals enhance immune function; by contrast, under pathological conditions, free radicals cause damage to normal tissues and cells, leading to diseases (125). Scavenging free radicals is another important tool for tumor prevention and treatment. The activities of free radical-scavenging enzymes, such as catalase and superoxide dismutase (SOD), are markedly decreased in patients with tumors, whereas the content of lipid peroxides and lipid peroxidation products such as malondialdehyde (MDA) are elevated (126). In addition, the anti-oxidative stress function is decreased. APS have been reported to reduce free radical production, increase SOD activity, decrease MDA content, and scavenge ROS and reactive nitrogen species generated by intracellular oxidative stress (127).
In patients with cancer who are receiving chemotherapy, most treatment failures or deaths are attributed to tumor MDR (128). Modern pharmaceutical research focuses on drugs that can reverse or escape MDR, and the key to reversing tumor drug resistance lies in blocking the MDR pathway, reducing drug efflux and improving the chemosensitivity of tumor cells (129). APS markedly increases tumor response to chemotherapeutic drugs and reverses the MDR effect, while reducing the toxicity response induced by chemotherapeutic drugs (130). APS can also inhibit TGF-β1 overexpression-mediated EMT, and inhibit the proliferation of cisplatin-resistant A549 cell xenograft tumors (131). In vitro experiments confirmed that APS induces apoptosis in GC cells and enhances the pro-apoptotic effect of Adriamycin on GC cells, and that APS serves as a chemotherapeutic sensitizer to a certain extent (95). In addition, in the HL-60/A cell line with high drug resistance, APS induces apoptosis, activates the caspase cascade reaction, decreases the expression levels of MDR-associated protein and increases the concentration of intracellular antitumor drugs (132). Furthermore, APS increases the sensitivity of tumor cells to chemotherapeutic drugs, such as apatinib, escapes MDR and strengthens the antitumor effect (133). In the experimental study using H22 hepatoma cell xenograft mouse models, the expression levels of IL-6 and TNF-α were increased, whereas those of IL-10, P-glycoprotein and MDR1 were decreased in the APS treatment group. In addition, APS was shown to enhance the antitumor effect of Adriamycin (134). APS can also serve as a chemosensitive enhancer by activating the JNK signaling pathway and enhancing the sensitivity of SKOV3 ovarian cancer cells to cisplatin (135). Furthermore, APS has been shown to enhance the sensitivity of HeLa cervical cancer cells to cisplatin chemotherapy by upregulating the autophagy-related protein beclin1, promoting the conversion of LC3 I to LC3 II, and downregulating the marker protein p62 to enhance the autophagic activity of HeLa cells (133). APS promotes the apoptosis of gefitinib-resistant cells, and reduces cell proliferation and migration. Moreover, APS inhibits the PD-L1/SREBP-1/EMT signaling pathway, increases the expression levels of E-cadherin, reduces the expression levels of N-cadherin and vimentin, and reverses the acquired resistance of lung cancer cells to gefitinib (136).
Astragalus membranaceus has a medicinal history of >2,000 years in China. As an active ingredient with the highest content in Astragalus membranaceus, APS has shown great application potential in regulating the tumor microenvironment and improving immunity. Numerous previous experimental studies have confirmed that APS can regulate the activation of immune cells and the release of cytokines in the tumor microenvironment, as well as inhibit tumor immune escape. The combination of APS and chemotherapeutic drugs may alleviate the adverse reactions of patients after chemotherapy and improve their quality of life, which provides great reference for the use of APS as an adjuvant in clinical antitumor medication. APS also has certain effects on the treatment of other diseases, such as diabetes and inflammatory diseases (137). Therefore, the therapeutic use of APS for the treatment of other diseases requires further exploration.
The structure of APS is complex; thus, its bioavailability can be further improved through chemical means, such as structural modification, so that it can serve a better targeted role in cancer. Although APS has been proven to have good application potential in antitumor immunity, it still faces a number of problems: i) The structure of APS is complex, and it contains a large number of chemical components. Its extraction, separation, purification and structural analysis are currently the biggest challenges, which require further in-depth exploration. ii) Tumor immune escape is a complex process in the tumor microenvironment. Tumor cells can not only directly affect the function of immune cells, but also indirectly inhibit immunity by regulating immune checkpoints and cytokines. Although APS can improve the function of immune cells in the tumor microenvironment, the specific mechanism by which APS regulates immune cells remains unclear. iii) At present, most of the studies on the antitumor effect of APS immunotherapy are limited to cell experiments or in vivo experiments using mice, and evidence from clinical-related trials is lacking. Therefore, more abundant clinical trials are needed for verification. In conclusion, the research on the mechanism of action of APS in regulating immune cells, improving the tumor immune microenvironment and resisting tumors, as well as further research on the mechanism by which APS serves a regulatory role in the immunotherapy of malignant tumors, is of great importance to the research and development of clinical antitumor drugs. This research may improve the treatment of tumor-related diseases by combining traditional Chinese and Western medicine.
Not applicable.
The present study was supported by the National Natural Science Foundation of China (grant no. 82173996).
Not applicable.
ZQ, ZW and YW conceived and designed the study. ZQ wrote the manuscript. YW and ZW revised the manuscript. WC and XX conducted literature organization and proofreading Data authentication is not applicable. All authors read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI | |
|
Liu Y, Zhao Y, Song H, Li Y, Liu Z, Ye Z, Zhao J, Wu Y, Tang J and Yao M: Metabolic reprogramming in tumor immune microenvironment: Impact on immune cell function and therapeutic implications. Cancer Lett. 597:2170762024. View Article : Google Scholar : PubMed/NCBI | |
|
Xie D, Wang N, Chen Z, Liu Z, Zhou Z, Zeng J, Zhong Y and Wang Z: Medication Rules and Core Formula Analysis of Famous National Traditional. Chinese Medicine Masters for Cancer Treatment Based on Data Mining World Chin Med. 15:2726–2734. 2020.(In Chinese). | |
|
He Z, Liu X, Qin S, Yang Q, Na J, Xue Z and Zhong L: Anticancer mechanism of Astragalus polysaccharide and its application in cancer immunotherapy. Pharmaceuticals (Basel). 17:6362024. View Article : Google Scholar : PubMed/NCBI | |
|
National Pharmacopoeia Commission: Pharmacopoeia of the People's Republic of China (Part 1). Pharmaceutical Science and Technology Press. (Beijing, China). 3632020.(In Chinese). | |
|
Su HF, Shaker S, Kuang Y, Zhang M, Ye M and Qiao X: Phytochemistry and cardiovascular protective effects of Huang-Qi (Astragali Radix). Med Res Rev. 41:1999–2038. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tong X: The Pharmacology Study of Active Constituents from Astragalus. Lishizhen Medicine and Materia Medica Research. 22:1246–1249. 2011.(In Chinese). | |
|
Xiao M, Chen H, Shi Z and Rui W: Rapid and reliable method for analysis of raw and honey-processed Astragalus by UPLC/ESI-Q-TOF-MS using HSS T3 columns. Anal Methods. 6:8045–8054. 2014. View Article : Google Scholar | |
|
Liao J, Huang J, Liu W, Liao S, Chen H and Rui W: Effects of total flavonoids and astragalosides of honey-processed Astragalus on NO secretion levels. J Guangdong Pharm Univ. 33:162–166. 2017.(In Chinese). | |
|
Huang J, Liao J, Liu W, Liao S, Feng Y, Chen H and Rui W: Establishment of determination method for three main active ingredients of honey-processed Astragalus in rat urine based on UPLC/Q-TOF-MS. Lishizhen Med Mater Med Res. 28:2564–2567. 2017.(In Chinese). | |
|
Zhong RZ, Yu M, Liu HW, Sun HX, Cao Y and Zhou DW: Effects of dietary Astragalus polysaccharide and Astragalus membranaceus root supplementation on growth performance, rumen fermentation, immune responses, and antioxidant status of lambs. Animal Feed Sci Technol. 174:60–67. 2012. View Article : Google Scholar | |
|
Zeng P, Li J, Chen Y and Zhang L: The structures and biological functions of polysaccharides from traditional Chinese herbs. Prog Mol Biol Transl Sci. 163:423–444. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Fan X, Li K, Yang Y, Li H, Li X and Qin X: Research Progress on structural characterization of astragalus polysaccharides. J Shanxi Coll Tradit Chin Med. 23:260–267. 2022.(In Chinese). | |
|
Synytsya A and Novák M: Structural diversity of fungal glucans. Carbohydr Polym. 92:792–809. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Lin MG, Yang YF and Xu HY: Chemical structure of Astragalus polysaccharide MAPS-5 and its in vitro lymphocyte proliferative activity. Chin Tradit Herbal Drugs. 40:1865–1868. 2009.(In Chinese). | |
|
Fang SD, Chen Y, Xu XY and Ye C: Studies on the Active Principles of Astragalus Mongholicus Bunge I. Isolation, Characterization and Biological Effect of its Polysaccharaides. Chin J Org Chem. 2:26–31. 1982.(In Chinese). | |
|
Huang Q, Lu G, Li Y, Guo J and Wang R: Studies on the polysaccharides of “Huang Qi” (Astragalus Mongholicus Bunge.). Acta Pharm Sin. 17:200–206. 1982.(In Chinese). | |
|
Tang Y, Zhang Y, Wang Y, Zhao H and Shen Y: Isolation and Structural Feature Analysis of Astragalus Polysaccharides. Lishizhen Med Mater Med Res. 25:1097–1100. 2014.(In Chinese). | |
|
Cao Y, Li K, Qin X, Jiao S, Du Y, Li S and Li X: Quality evaluation of different areas of Astragali Radix based on carbohydrate specific chromatograms and immune cell activities. Acta Pharmaceutica Sinica. 54:1277–1287. 2019.(In Chinese). | |
|
Liu W, Yu X, Xu J, et al: Isolation, structural characterization and prebiotic activity of polysaccharides from Astragalus membranaceus. Food Ferment Ind. 46:50–56. 2020.(In Chinese). | |
|
Bergman PJ: Cancer immunotherapies. Vet Clin North Am Small Anim Pract. 49:881–902. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Rui R, Zhou L and He S: Cancer immunotherapies: Advances and bottlenecks. Front Immunol. 14:12124762023. View Article : Google Scholar : PubMed/NCBI | |
|
Helmy KY, Patel SA, Nahas GR and Rameshwar P: Cancer immunotherapy: Accomplishments to date and future promise. Ther Deliv. 4:1307–1320. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH and Diaz LA Jr: Immunotherapy in colorectal cancer: Rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 16:361–375. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lei X, Lei Y, Li JK, Du WX, Li RG, Yang J, Li J, Li F and Tan HB: Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 470:126–133. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Katz JB, Muller AJ and Prendergast GC: Indoleamine 2,3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol Rev. 222:206–221. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Velcheti V and Schalper K: Basic overview of current immunotherapy approaches in cancer. Am Soc Clin Oncol Educ Book. 35:298–308. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Li M, Bi L, Hu X and Wang Y: Traditional Chinese medicine in regulating tumor microenvironment. Onco Targets Ther. 17:313–325. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kong F, Chen T, Li X and Jia Y: The current application and future prospects of Astragalus polysaccharide combined with cancer immunotherapy: A review. Front Pharmacol. 12:7376742021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou L, Liu Z, Wang Z, Yu S, Long T, Zhou X and Bao Y: Astragalus polysaccharides exerts immunomodulatory effects via TLR4-mediated MyD88-dependent signaling pathway in vitro and in vivo. Sci Rep. 7:448222017. View Article : Google Scholar : PubMed/NCBI | |
|
Huang WC, Kuo KT, Bamodu OA, Lin YK, Wang CH, Lee KY, Wang LS, Yeh CT and Tsai JT: Astragalus polysaccharide (PG2) ameliorates cancer symptom clusters, as well as improves quality of life in patients with metastatic disease, through modulation of the inflammatory cascade. Cancers (Basel). 11:10542019. View Article : Google Scholar : PubMed/NCBI | |
|
Bonnardel J and Guilliams M: Developmental control of macrophage function. Curr Opin Immunol. 50:64–74. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Yang Y, Xiong L, Jiang P, Wang J and Li C: Metabolism, metabolites, and macrophages in cancer. J Hematol Oncol. 16:802023. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A, Marchesi F, Malesci A, Laghi L and Allavena P: Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 14:399–416. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mantovani A and Allavena P: The interaction of anticancer therapies with tumor-associated macrophages. J Exp Med. 212:435–445. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wei W, Li ZP, Bian ZX and Han QB: Astragalus polysaccharide RAP induces macrophage phenotype polarization to M1 via the notch signaling pathway. Molecules. 24:20162019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen R, Li Y, Zuo L, Xiong H, Sun R, Song X and Liu H: Astragalus polysaccharides inhibits tumor proliferation and enhances cisplatin sensitivity in bladder cancer by regulating the PI3K/AKT/FoxO1 axis. Int J Biological Macromolecules. 311:1437392025. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Pan XY, Ma M, Zhao J, Zhao F and Lv YP: Astragalus polysacharin inhibits hepatocellular carcinoma-like phenotypes in a murine HCC model through repression of M2 polarization of tumour-associated macrophages. Pharm Biol. 59:1531–1537. 2021. View Article : Google Scholar | |
|
Li SY, Li K, Qin XM, Li XR and Du YG: Study on active constituents of Astragalus polysaccharides for injection to regulate metabolism of macrophages based on LC-MS metabolomics. Chin Tradit Herb Drugs. 51:1575–1585. 2020.(In Chinese). | |
|
Feng S, Ding H, Liu L, Peng C, Huang Y, Zhong F, Li W, Meng T, Li J, Wang X, et al: Astragalus polysaccharide enhances the immune function of RAW264.7 macrophages via the NF-κB p65/MAPK signaling pathway. Exp Ther Med. 21:202021. View Article : Google Scholar : PubMed/NCBI | |
|
Shao BM, Xu W, Dai H, Tu P, Li Z and Gao XM: A study on the immune receptors for polysaccharides from the roots of Astragalus membranaceus, a Chinese medicinal herb. Biochem Biophys Res Commun. 320:1103–1111. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Wei W, Xiao HT, Bao WR, Ma DL, Leung CH, Han XQ, Ko CH, Lau CB, Wong CK, Fung KP, et al: TLR-4 may mediate signaling pathways of Astragalus polysaccharide RAP induced cytokine expression of RAW264.7 cells. J Ethnopharmacol. 179:243–252. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Li W, Hu X, Wang S, Jiao Z, Sun T, Liu T and Song K: Characterization and anti-tumor bioactivity of Astragalus polysaccharides by immunomodulation. Int J Biol Macromol. 145:985–997. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin W, Liu T, Wang B and Bi H: The role of ocular dendritic cells in uveitis. Immunol Lett. 209:4–10. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Villar J and Segura E: Decoding the heterogeneity of human dendritic cell subsets. Trends Immunol. 41:1062–1071. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chudnovskiy A, Pasqual G and Victora GD: Studying interactions between Dendritic Cells and T cells in vivo. Curr Opin Immunol. 58:24–30. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bamodu OA, Kuo KT, Wang CH, Huang WC, Wu ATH, Tsai JT, Lee KY, Yeh CT and Wang LS: Astragalus polysaccharides (PG2) enhances the M1 polarization of macrophages, functional maturation of dendritic cells, and T cell-mediated anticancer immune responses inpatients with lung cancer. Nutrients. 11:22642019. View Article : Google Scholar : PubMed/NCBI | |
|
Jing XN, Qiu B, Wang JF, Wu YG, Wu JB and Chen DD: In vitro anti-tumor effect of human dendritic cells vaccine induced by Astragalus polysacharin: An experimental study. Zhongguo Zhong Xi Yi Jie He Za Zhi. 34:1103–1107. 2014.(In Chinese). PubMed/NCBI | |
|
Gu Y, Pu X, Yin N and Wan C: The study on mechanism of the anti-tumor effect of Fufang Huangqi Oral Solution combined chemotherapy. J Tianjin Univ Tradit Chin Med. 41:85–89. 2022.(In Chinese). | |
|
Bian Y, Zhang J, Wang L, Wang X, Zhang K, Wang X, Meng J and Li J: Progress of research on the effect of Astragalus polysaccharide on the morphology and function of dendritic cells. Prog Vet Med. 37:86–89. 2016.(In Chinese). | |
|
Pang G, Chen C, Liu Y, Jiang T, Yu H, Wu Y, Wang Y, Wang FJ, Liu Z and Zhang LW: Bioactive polysaccharide nanoparticles improve radiation-induced abscopal effect through manipulation of dendritic cells. ACS Appl Mater Interfaces. 11:42661–42670. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chang WT, Lai TH, Chyan YJ, Yin SY, Chen YH, Wei WC and Yang NS: Specific medicinal plant polysaccharides effectively enhance the potency of a DC-based vaccine against mouse mammary tumor metastasis. PLoS One. 10:e01223742015. View Article : Google Scholar : PubMed/NCBI | |
|
An EK, Zhang W, Kwak M, Lee PC and Jin JO: Polysaccharides from Astragalus membranaceus elicit T cell immunity by activation of human peripheral blood dendritic cells. Int J Biol Macromol. 223:370–377. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J and Umansky V: Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol. 21:147–164. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Xia L, Zhao S, Wei Q and Xu X: Study on changes of myeloid-derived suppressor cells in tumor tissues of tumor-bearing mice and its significance. Curr Immunol. 34:232–236. 2014.(In Chinese). | |
|
Hao WB, Xiang FF, Liu QL, Yue HK and Kang XD: Research adances of myeloid inhibitory cells in tumor microenvironment. Immunol J. 33:729–732. 2017.(In Chinese). | |
|
Chai W, He X, Zhu J, Lv C, Zhao H, Lv A and Yv C: Immunoregulatory effects of astragalus polysaccharides on myeloid derived suppressor cells in B16-F10 tumor bearing mice. Chin J Basic Med Tradit Chin Med. 18:63–65. 2012.(In Chinese). | |
|
Ding G, Gong Q, Ma J, Liu X, Wang Y and Cheng X: Immunosuppressive activity is attenuated by Astragalus polysaccharides through remodeling the gut microenvironment in melanoma mice. Cancer Sci. 112:4050–4063. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mei Q, Zhang W, Ran R and Wang J: Effect of astragalus polysaccharide on peripheral circulation MDSC in lung cancer and its clinical effect. Modern Doctor. 55:10–13. 2017.(In Chinese). | |
|
Li Y, Yan B and He S: Advances and challenges in the treatment of lung cancer. Biomed Pharmacother. 169:1158912023. View Article : Google Scholar : PubMed/NCBI | |
|
Shen M, Wang YJ, Liu ZH, Chen YW, Liang QK, Li Y and Ming HX: Inhibitory effect of Astragalus polysaccharide on premetastatic niche of lung cancer through the S1PR1-STAT3 signaling pathway. Evid Based Complement Alternat Med. 2023:40107972023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Yuan J, Guo M, Zhang Y, Lu J and Cao W: Effect of astragalus polysaccharides based on TLR4/MyD88/NF-κB signaling pathway on immune function of lung cancer mice and its regulatory effect on Th1/ Th2. Chin J Immunol. 37:676–682. 2021.(In Chinese). | |
|
Li K, Li S, Wang D, Li X, Wu X, Liu X, Du G, Li X, Qin X and Du Y: Extraction, characterization, antitumor and immunological activities of hemicellulose polysaccharide from Astragalus radix herb residue. Molecules. 24:36442019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Liu Z, Long T, Zhou L and Bao Y: Immunomodulatory effects of herbal formula of Astragalus polysaccharide (APS) and polysaccharopeptide (PSP) in mice with lung cancer. Int J Biol Macromol. 106:596–601. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yu J, Dong XD, Jiao JS, Ji HY and Liu AJ: Antitumor and immunoregulatory activities of a novel polysaccharide from Astragalus membranaceus on S180 tumor-bearing mice. Int J Biol Macromol. 189:930–938. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Deng M, Chen Z, Dou X, Shi Y and Wo X: The Anti - leukemia Effect Mediated by DCs Derived from the Human Cord Blood Monocyte Induced by the Astragalus Polysaccharides (APS) in Vitro. Chin Arch Tradit Chin Med. 27:2360–2365. 2009.(In Chinese). | |
|
Jang JE, Hajdu CH, Liot C, Miller G, Dustin ML and Bar-Sagi D: Crosstalk between regulatory T cells and tumor-associated dendritic cells negates anti-tumor immunity in pancreatic cancer. Cell Rep. 20:558–571. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Mentucci FM, Ferrara MG, Ercole A, Rumie Vittar NB and Lamberti MJ: Interplay between cancer-associated fibroblasts and dendritic cells: implications for tumor immunity. Front Immunol. 16:15153902025. View Article : Google Scholar : PubMed/NCBI | |
|
Sun S, He X, Chai W, Lv C, Wang P, Lv A and Yu C: Effects of Astragalus Polysaccharide on Treg Cells in Tumor-bearing Mice. Chin J Exp Tradit Med Formulae. 19:176–178. 2013.(In Chinese). | |
|
Li Q, Bao JM, Li XL, Zhang T and Shen XH: Inhibiting effect of Astragalus polysaccharides on the functions of CD4+CD25 highTreg cells in the tumor microenvironment of human hepatocellular carcinoma. Chin Med J (Engl). 125:786–793. 2012.PubMed/NCBI | |
|
Du X, Chen X, Zhao B, Lv Y, Zhang H, Liu H, Chen Z, Chen Y and Zeng X: Astragalus polysaccharides enhance the humoral and cellular immune responses of hepatitis B surface antigen vaccination through inhibiting the expression of transforming growth factor β and the frequency of regulatory T cells. FEMS Immunol Med Microbiol. 63:228–235. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Shokati E, Safari E and Motalebnezhad M: Astragalus Polysaccharide Mediates Immunomodulatory Effects on Crosstalk between Human Peripheral Blood Mononuclear Cells and Ovarian Cancer Cell Line. Iran J Allergy Asthma Immunol. 22:172–182. 2023.PubMed/NCBI | |
|
Guillerey C and Smyth MJ: NK cells and cancer immunoediting. Curr Top Microbiol Immunol. 395:115–145. 2016.PubMed/NCBI | |
|
Zhou J, Peng H and Tian ZG: Research progress on negative regulation of adaptive immune response by NK cells. Chin J Immunol. 32:769–776. 2016.(In Chinese). | |
|
Wang W, Xi J, Zhao Y, He L, Zhou J, Fan Z, Zhang B, Wang H, Zeng Q, et al: Specific cytotoxicity of natural killer cells against hepatocellular carcinoma and potential mechanism. Mil Med Sci. 41:775–783. 2017.(In Chinese). | |
|
Yang Y, Li H, Guo S, Li X, Yin J, Zhao P, Gao L and Wu S: Killing effect of NK cells on non-small cell lung cancer cells NCI-H292 and the effect on their β-catenin protein expression. Shandong Med J. 57:33–35. 2017.(In Chinese). | |
|
Liu C, Zhang Y and Lu H: Efficient amplification of NK cells and their anti-tumor cytotoxic activity in vitro. Chin J Immunol. 33:1186–1190+1196. 2017.(In Chinese). | |
|
Zeng PY, Deng LL, Yue LL and Zhang L: Effect of Astragalus Polysaccharide on Sensitivity of Leukemic Cell Line HL- 60 to NK Cell Cytotoxicity and Its Mechanism. J Exp Hematol. 20:880–883. 2012.(In Chinese). | |
|
Li H: Effects of Astragalus Polysaccharide on Quality of Life and Cellular Immune Function in Elderly Patients with Advanced Pancreatic Cancer. Chinese Journal of Traditional Chinese Medicine and Pharmacy. 17:243–244. 2010.(In Chinese). | |
|
Li R, Chen WC, Wang WP, Tian WY and Zhang XG: Extraction, characterization of Astragalus polysaccharides and its immune modulating activities in rats with gastric cancer. Carbohydr Polym. 78:738–742. 2009. View Article : Google Scholar | |
|
Guo Y, Zhang Z, Wang Z, Liu G, Liu Y and Wang H: Astragalus polysaccharides inhibit ovarian cancer cell growth via microRNA-27a/FBXW7 signaling pathway. Biosci Rep. 40:BSR201933962020. View Article : Google Scholar : PubMed/NCBI | |
|
Miao D, Zhao J, Han Y, Zhou J, Li X, Zhang T, Li W and Xia Y: Management of locally advanced non-small cell lung cancer: State of the art and future directions. Cancer Commun (Lond). 44:23–46. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Tao X, Zhang X and Feng F: Astragalus polysaccharide suppresses cell proliferation and invasion by up-regulation of miR-195-5p in non-small cell lung cancer. Biol Pharm Bull. 45:553–560. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang YM, Liu YQ, Liu D, Zhang L, Qin J, Zhang Z, Su Y, Yan C, Luo YL, Li J, et al: The effects of Astragalus polysaccharide on bone marrow-derived mesenchymal stem cell proliferation and morphology induced by A549 lung cancer cells. Med Sci Monit. 25:4110–4121. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang CH, Yu D, Gao Y, Jing H, Wang Y and Liu C: Effect of Astragalus Polysaccharide on EMT of A549/DDP Lung Adenocarcinoma Xenograft: An Exploration Based on PI3K/Akt Signaling Pathway. Chin J Exp Tradit Med Formulae. 27:51–58. 2021.(In Chinese). | |
|
Yang YL, Lin ZW, He PT, Nie H, Yao QY and Zhang SY: Inhibitory effect of Astragalus polysaccharide combined with cisplatin on cell cycle and migration of nasopharyngeal carcinoma cell lines. Biol Pharm Bull. 44:926–931. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Hong T, Luo J, Cao X, Liu W, Gao Y, Cui W and Wang F: Effect of Astragali Radix Polysaccharides on Proliferation and Apoptosis of Human Colon Cancer Cell Line SW620. Chin J Exp Tradit Med Formulae. 23:97–101. 2017.(In Chinese). | |
|
Guo S, Ma B, Jiang X, Li X and Jia Y: Astragalus polysaccharides inhibits tumorigenesis and lipid metabolism through miR-138-5p/SIRT1/SREBP1 pathway in prostate cancer. Front Pharmacol. 11:5982020. View Article : Google Scholar : PubMed/NCBI | |
|
Chu Y, Fang Y, Chi J, Li J, Zhang D, Zou Y and Wang Z: Astragalus polysaccharides decrease proliferation, migration, and invasion but increase apoptosis of human osteosarcoma cells by up-regulation of microRNA-133a. Braz J Med Biol Res. 51:e76652018. View Article : Google Scholar : PubMed/NCBI | |
|
Mocan L, Matea C, Tabaran FA, Mosteanu O, Pop T, Mocan T and Iancu C: Photothermal treatment of liver cancer with albumin-conjugated gold nanoparticles initiates Golgi Apparatus-ER dysfunction and caspase-3 apoptotic pathway activation by selective targeting of Gp60 receptor. Int J Nanomedicine. 10:5435–5445. 2015.PubMed/NCBI | |
|
Moyer A, Tanaka K and Cheng EH: Apoptosis in cancer biology and therapy. Annu Rev Pathol. 20:303–328. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Ouyang L, Shi Z, Zhao S, Wang FT, Zhou TT, Liu B and Bao JK: Programmed cell death pathways in cancer: A review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 45:487–498. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Z, Meng M and Ni H: Chemosensitizing effect of Astragalus polysaccharides on nasopharyngeal carcinoma cells by inducing apoptosis and modulating expression of Bax/Bcl-2 ratio and caspases. Med Sci Monit. 23:462–469. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Song J, Chen Y, He D, Tan W, Lv F, Liang B, Xia T and Li J: Astragalus polysaccharide promotes adriamycin-induced apoptosis in gastric cancer cells. Cancer Manag Res. 12:2405–2414. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yu J, Ji H, Dong X, Feng Y and Liu A: Apoptosis of human gastric carcinoma MGC-803 cells induced by a novel Astragalus membranaceus polysaccharide via intrinsic mitochondrial pathways. Int J Biol Macromol. 126:811–819. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ohnuki H, Jiang K, Wang D, Salvucci O, Kwak H, Sánchez-Martín D, Maric D and Tosato G: Tumor-infiltrating myeloid cells activate Dll4/Notch/TGF-β signaling to drive malignant progression. Cancer Res. 74:2038–2049. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Zhang S, Lu Y, Zhang Y, Wang E and Cui Z: The roles of Notch3 on the cell proliferation and apoptosis induced by CHIR99021 in NSCLC cell lines: A functional link between Wnt and Notch signaling pathways. PLoS One. 8:e846592013. View Article : Google Scholar : PubMed/NCBI | |
|
Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, Carbone DP and Dang TP: Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 65:3555–3561. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Konishi J, Kawaguchi KS, Vo H, Haruki N, Gonzalez A, Carbone DP and Dang TP: Gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation in human lung cancers. Cancer Res. 67:8051–8057. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang JX, Han YP, Bai C and Li Q: Notch1/3 and p53/p21 are a potential therapeutic target for APS-induced apoptosis in non-small cell lung carcinoma cell lines. Int J Clin Exp Med. 8:12539–12547. 2015.PubMed/NCBI | |
|
Sha X, Xu X, Liao S, Chen H and Rui W: Evidence of immunogenic cancer cell death induced by honey-processed Astragalus polysaccharides in vitro and in vivo. Exp Cell Res. 410:1129482022. View Article : Google Scholar : PubMed/NCBI | |
|
Feng T and Zhang BG: Inhibitory effect of Astragalus polysaccharide on hepatocellular carcinoma HepG2 cells and its mechanism. Prac Geriatr. 6:486–488. 4952010.(In Chinese). | |
|
Jiao J, Yu J, Ji H and Liu A: Synthesis of macromolecular Astragalus polysaccharide-nano selenium complex and the inhibitory effects on HepG2 cells. Int J Biol Macromol. 211:481–489. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang HY: The Role of ERK1 /2 in Astragalus Polysaccharide-induced Apoptosis in HepG2 Cell Line. Chin J Exp Tradit Med Formulae. 7:235–238. 2012.(In Chinese). | |
|
Hart GW, Housley MP and Slawson C: Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature. 446:1017–1022. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Dong DL and Hart GW: Purification and characterization of an O-GlcNAc selective N-acetyl-beta-D-glucosaminidase from rat spleen cytosol. J Biol Chem. 269:19321–19330. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Kreppel LK, Blomberg MA and Hart GW: Dynamic glycosylation of nuclear and cytosolic proteins. Cloning and characterization of a unique O-GlcNAc transferase with multiple tetratricopeptide repeats. J Biol Chem. 272:9308–9315. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Haltiwanger RS, Holt GD and Hart GW: Enzymatic addition of O-GlcNAc to nuclear and cytoplasmic proteins. Identification of a uridine diphospho-N-acetylglucosamine: Peptide beta-N-acetylglucosaminyltransferase. J Biol Chem. 265:2563–2568. 1990. View Article : Google Scholar : PubMed/NCBI | |
|
Hart GW, Slawson C, Ramirez-Correa G and Lagerlof O: Cross talk between O-GlcNAcylation and phosphorylation: Roles in signaling, transcription, and chronic disease. Annu Rev Biochem. 80:825–858. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lee SJ, Lee DE, Choi SY and Kwon OS: OSMI-1 enhances TRAIL-induced apoptosis through ER stress and NF-κB signaling in colon cancer cells. Int J Mol Sci. 22:110732021. View Article : Google Scholar : PubMed/NCBI | |
|
Lee SJ and Kwon OS: O-GlcNAc transferase inhibitor synergistically enhances doxorubicin-induced apoptosis in HepG2 cells. Cancers (Basel). 12:31542020. View Article : Google Scholar : PubMed/NCBI | |
|
Ferrer CM, Lynch TP, Sodi VL, Falcone JN, Schwab LP, Peacock DL, Vocadlo DJ, Seagroves TN and Reginato MJ: O-GlcNAcylation regulates cancer metabolism and survival stress signaling via regulation of the HIF-1 pathway. Mol Cell. 54:820–831. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Duan F, Pan Z, Liu X, Lu W, Liang C, Fang Z, Peng P and Jia D: Astragalus polysaccharide promotes doxorubicin-induced apoptosis by reducing O-GlcNAcylation in hepatocellular carcinoma. Cells. 12:8662023. View Article : Google Scholar : PubMed/NCBI | |
|
Kemse NG, Kale A and Joshi SR: Supplementation of maternal omega-3 fatty acids to pregnancy induced hypertension Wistar rats improves IL-10 and VEGF levels. PLEFA. 43:276–279. 2015. | |
|
Zhao L, Zhong Y, Liang J, Gao H and Tang N: Effect of Astragalus polysaccharide on the expression of VEGF and EGFR in mice with lewis transplantable lung cancer. J Coll Physicians Surg Pak. 29:392–394. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen W, Li Z, Bai L and Lin Y: NF-kappaB in lung cancer, a carcinogenesis mediator and a prevention and therapy target. Front Biosci (Landmark Ed). 16:1172–1185. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wu CY, Ke Y, Zeng YF, Zhang YW and Yu HJ: Anticancer activity of Astragalus polysaccharide in human non-small cell lung cancer cells. Cancer Cell Int. 17:1152017. View Article : Google Scholar : PubMed/NCBI | |
|
Auyeung KK, Law PC and Ko JK: Novel anti-angiogenic effects of formononetin in human colon cancer cells and tumor xenograft. Oncol Rep. 28:2188–2194. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Zhao W, Geng L, Jiang J, Yang N, Gu L, Wang L and Wang Y: Astragalus Polysaccharides Inhibiting Gastric Cancer Cell Proliferation and Invasion via miR-25/FBXW7 Signaling Pathway. New Chinese Medicine. 57:150–155. 2025.(In Chinese). | |
|
Ji F, Zhang H, Wang Y, Li M, Xu W, Kang Y, Wang Z, Wang Z, Cheng P, Tong D, et al: MicroRNA-133a, downregulated in osteosarcoma, suppresses proliferation and promotes apoptosis by targeting Bcl-xL and Mcl-1. Bone. 56:220–226. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Medici D, Shore EM, Lounev VY, Kaplan FS, Kalluri R and Olsen BR: Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med. 16:1400–1406. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Yang S, Sun S, Xu W, Yu B, Wang G and Wang H: Astragalus polysaccharide inhibits breast cancer cell migration and invasion by regulating epithelial-mesenchymal transition via the Wnt/β-catenin signaling pathway. Mol Med Rep. 21:1819–1832. 2020.PubMed/NCBI | |
|
He L, Xu K, Niu L and Lin L: Astragalus polysaccharide (APS) attenuated PD-L1-mediated immunosuppression via the miR-133a-3p/MSN axis in HCC. Pharm Biol. 60:1710–1720. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang N, Jia X, Zhang Z, Sun E and Yan H: Advance in studies on anti-cancer activity and mechanism of flavonoids. China J Chin Mater Med. 40:373–381. 2015.(In Chinese). | |
|
Glorieux C, Liu S, Trachootham D and Huang P: Targeting ROS in cancer: Rationale and strategies. Nat Rev Drug Discov. 23:583–606. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Pu X, Fan W, Yu S, Li Y, Ma X, Liu L, Ren J and Zhang W: Polysaccharides from angelica and Astragalus exert hepatoprotective effects against carbon-tetrachloride-induced intoxication in mice. Can J Physiol Pharmacol. 93:39–43. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Bukowski K, Kciuk M and Kontek R: Mechanisms of multidrug resistance in cancer chemotherapy. IntJ Mol Sci. 21:32332020. View Article : Google Scholar | |
|
Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, Zhang DM and Chen ZS: Autophagy and multidrug resistance in cancer. Chin J Cancer. 36:522017. View Article : Google Scholar : PubMed/NCBI | |
|
Cao Y, Ruan Y, Shen T, Huang X, Li M, Yu W, Zhu Y, Man Y, Wang S and Li J: Astragalus polysaccharide suppresses doxorubicin-induced cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways. Oxid Med Cell Longev. 2014:6742192014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Wang CH, Yu D, Gao Y, Jing H, Tang Y and Liu C: Astragalus Polysaccharides Improve Cisplatin Resistance by Inhibiting EMT of Lung Adenocarcinoma A549/DDP Cells Transplanted into Nude Mice. Chin J Exp Tradit Med Formulae. 28:79–85. 2022.(In Chinese). | |
|
Peng H, Zhang X and Li F: Mechanisms of Astragalus Polysaccharids Synergized with Doxorubicin against Drug Resistance in HL-60/A Cells. J Exp Hemat. 25:743–748. 2017.(In Chinese). | |
|
Zhai QL, Hu XD, Xiao J and Yu DQ: Astragalus polysaccharide may increase sensitivity of cervical cancer HeLa cells to cisplatin by regulating cell autophagy. Zhongguo Zhong Yao Za Zhi. 43:805–812. 2018.(In Chinese). PubMed/NCBI | |
|
Tian Q: The study on the adjunct anticancer mechanism of astragalus polysaccharide in vivo and in vitro. PhD Thesis. Central South University. 26–49. 2013.(In Chinese). | |
|
Li C, Hong L, Liu C, Min J, Hu M and Guo W: Astragalus polysaccharides increase the sensitivity of SKOV3 cells to cisplatin. Arch Gynecol Obstet. 297:381–386. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wei J, Li Y, Xu B and Yu J: Astragalus polysaccharides reverse gefitinib resistance by inhibiting mesenchymal transformation in lung adenocarcinoma cells. Am J Transl Res. 12:1640–1657. 2020.PubMed/NCBI | |
|
Zhang Y, Chen Z, Chen L, Dong Q, Yang DH, Zhang Q, Zeng J, Wang Y, Liu X, Cui Y, et al: Astragali radix (Huangqi): A time-honored nourishing herbal medicine. Chin Med. 19:1192024. View Article : Google Scholar : PubMed/NCBI |