Next Article in Journal
Using In Silico Molecular Docking to Explain Differences in Receptor Binding Behavior of HHC and THCV Isomers: Revealing New Binding Modes
Previous Article in Journal
Evaluating the Role of Neddylation Modifications in Kidney Renal Clear Cell Carcinoma: An Integrated Approach Using Bioinformatics, MLN4924 Dosing Experiments, and RNA Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer Mechanism of Astragalus Polysaccharide and Its Application in Cancer Immunotherapy

1
State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
2
School of Pharmacy, Guangxi Medical University, Nanning 530021, China
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(5), 636; https://doi.org/10.3390/ph17050636
Submission received: 2 April 2024 / Revised: 6 May 2024 / Accepted: 8 May 2024 / Published: 15 May 2024
(This article belongs to the Section Biopharmaceuticals)

Abstract

:
Astragalus polysaccharide (APS) derived from A. membranaceus plays a crucial role in traditional Chinese medicine. These polysaccharides have shown antitumor effects and are considered safe. Thus, they have become increasingly important in cancer immunotherapy. APS can limit the spread of cancer by influencing immune cells, promoting cell death, triggering cancer cell autophagy, and impacting the tumor microenvironment. When used in combination with other therapies, APS can enhance treatment outcomes and reduce toxicity and side effects. APS combined with immune checkpoint inhibitors, relay cellular immunotherapy, and cancer vaccines have broadened the application of cancer immunotherapy and enhanced treatment effectiveness. By summarizing the research on APS in cancer immunotherapy over the past two decades, this review elaborates on the anticancer mechanism of APS and its use in cancer immunotherapy and clinical trials. Considering the multiple roles of APS, this review emphasizes the importance of using APS as an adjunct to cancer immunotherapy and compares other polysaccharides with APS. This discussion provides insights into the specific mechanism of action of APS, reveals the molecular targets of APS for developing effective clinical strategies, and highlights the wide application of APS in clinical cancer therapy in the future.

1. Introduction

Cancer is a significant global health issue owing to its high incidence and mortality rates, imposing a considerable burden on public health systems. As reported in the 2023 data of the World Health Organization, cancer stands as the primary or secondary contributor to death in individuals under 70 years of age in 183 countries [1]. Typical methods of cancer treatment include chemotherapy, radiation therapy, and surgery. Although surgery and radiation play essential roles in treating early-stage cancers, they often fail to meet the needs of patients with more advanced cancer stages. The main reasons for this lack of effectiveness are tumor recurrence and spread. Despite being tailored to a specific type, stage, and location of cancer, various restrictions exist [2]. Surgical resection combined with systemic chemotherapy is the cornerstone of localized cancer therapy. The cytotoxic effects of chemotherapy, the primary component of combination treatment, not only affect cancerous cells but also harm healthy cells, leading to drug resistance and adverse reactions [3,4,5,6]. Extended chemotherapy regimens may result in drug resistance, causing cancer cells to become tolerant to chemotherapeutic drugs, necessitating higher doses for therapeutic effectiveness, a situation that may be exacerbated by increased toxicity [6]. Given these complexities, the discovery of complementary therapies that enhance the efficacy of traditional treatment while reducing adverse effects is crucial. Recent studies have explored the anticancer and anti-inflammatory qualities of conventional plant extracts [7,8]. These extracts have shown potential to enhance the effectiveness of anticancer vaccines and, when used in conjunction with standard chemotherapy and radiation therapy, can improve treatment outcomes and minimize harmful effects through various mechanisms [9,10,11,12].
Astragalus membranaceus, commonly known as Huangqi (Chinese name) or Radix Astragali, is a perennial herb of the legume family that is widely distributed throughout temperate regions worldwide. The dried root of Radix Astragali has a long history in China and has been widely used in the treatment of many diseases since the Ming Dynasty [13]. The dried roots of Astragalus membranaceus contain complex bioactive constituents, including polysaccharides, flavonoids, Astragalus methyl glycosides, amino acids, trace elements, and more than 100 compounds [14]. Recent studies revealed the pharmacological functions of these components, particularly their polysaccharide fractions [15]. Astragalus polysaccharide (APS) has been identified as one of the main active ingredients. It consists of nine monosaccharides, including glucose, galactose, arabinose, and rhamnose, and may contain glucuronic acid and galacturonic acid [16]. Due to its water solubility and low-toxicity characteristics, APS has demonstrated significant biological activities in immune activation and antioxidant, antitumor, antidiabetic, anti-inflammatory, and anti-aging properties; thus, it has great potential for drug discovery in the pharmaceutical and nutraceutical fields [17,18,19,20,21,22,23]. Research indicates that APS is safe and effective for cancer treatment because it enhances the body’s immune response, mitigates the side effects of chemotherapy, and activates immune defenses [24,25].
This review focuses on the potential adverse effects of APS in cancer immunotherapy and its interactions with various immunotherapeutic methods, summarizing the findings of studies involving different cancer cell lines, animal models, and human trials. We highlight the ability of APS to promote the growth and activation of immune cells, as well as its role in inducing apoptosis in cancer cells. Additionally, we examine the supportive role of APS in immunotherapy and its synergistic outcomes when combined with immune checkpoint inhibitors (ICIs), overdose immunotherapy, and cancer vaccines to suggest novel approaches and directions for future cancer treatment strategies.

2. Anticancer Mechanism of Astragalus Polysaccharide

APS has recently attracted considerable attention in cancer therapy, showing great potential for fighting cancer through its unique immune-activating and immune-modulating functions. APS functions through several mechanisms, including the stimulation of immune cells, cell cycle disruption, the inhibition of signal transduction, the activation of cellular autophagy, the inhibition of lipid metabolism, and improvements in the ability of immune cells to adapt to the tumor microenvironment (TME). Together, these mechanisms make APS a promising adjuvant for cancer immunotherapy.

2.1. Activation and Regulation of Immune Cells

The influence of APS on immune cell activity in the TME is summarized in Figure 1 and Table 1.
Dendritic cells (DCs) present antigens, trigger immune responses and modulate immunity [44]. These cells transition from an immature state with low major histocompatibility class (MHC) II expression to maturity as they process antigens and move to lymphoid tissues, thereby elevating MHC II levels. Mature DCs display processed antigens in the lymph nodes, present endogenous antigens via MHC I molecules, and process antigens through various pathways, leading to T-cell activation [45,46]. Studies indicate that APS injections induce a significant elevation of MHC-I and MHC-II co-stimulatory molecules in splenic DC subgroups in C57BL/6 mice [26]. APS enhanced DC maturation by stimulating the secretion of interleukin-12 (IL-12) [27] and amplifying the surface expression of CD40, CD80, and HLA-DR in these cells [47]. APS intensified the immune response by enhancing the pDC pathway [28]. APS-loaded nanoparticles have shown the potential for radiotherapy to activate antigen presenting cells (APCs) and reverse TME [29].
When APCs take up and process tumor-specific antigens before migrating to lymphoid organs, they collectively produce both activating and inhibitory signals that trigger the activation of T cells, resulting in the formation of effector T cells targeting tumor-specific antigens. This is a defense mechanism against cancer cell evasion of the immune system [48,49]. APS facilitates T-cell activation [50,51]. In a study by Bamodu et al., APS was shown to boost the transformation of splenic DCs into CD11c (high) and CD45RB (low) DCs, facilitate the shift from a Th2 to Th1 response, and augment the Th1/Th2 ratio, thereby enhancing the immune function of T lymphocytes [30]. Another study by Hwang et al. illustrated that the intranasal administration of APS can activate DCs and further stimulate natural killer (NK) cells and T cells within the lymph nodes [31]. In vivo experiments have shown that APS can enhance immunity in cyclophosphamide-induced immunosuppressed mice by promoting the proliferation of T lymphocytes and B lymphocytes and inducing the production of immunoglobulins (IgA, IgG, and IgM) and a variety of cytokines, including IL-6, IL-2, interferon-gamma (IFN-γ), complement 3, complement 4, and tumor necrosis factor-alpha (TNF-α) [52].
Helper T cells (Th cells) play a crucial role as mediators of the immune response, and are predominantly categorized into Th1 and Th2 cells, which connect and activate other immune cells within the peripheral effector memory T (TEM) cell compartment [53]. The combination of APS and polysaccharide peptides led to expansion of the Th cell subset, a reduction in the proportion of suppressor T cells/cytotoxic T cells, and an increase in the CD4+/CD8+ ratio. Through the regulation of immune responses and reinforcement of antitumor effects, these interventions influence the levels of Th1, Th17, and Th2 cytokines, augmenting IL-2, tumor TNF-α, IFN-γ, and IL-17A, while diminishing IL-10 levels [32].
Studies have been carried out using nanoparticles as carriers to improve immunotherapy through APS activation of T cells. These specialized nanoparticles target lymph nodes, assist in escaping lysosomes, activate distant DCs, and control T-cell differentiation through cross-presentation [33]. They also trigger specific IgG expression, eliciting a Th1-polarized immune response [34] that effectively combats cancer cell growth, spread, and infiltration [35], ultimately disrupting the immunosuppressive microenvironment and establishing an enduring and strong tumor antigen-specific immunity.
Macrophages possess anti-infective, antitumor, and immunomodulatory properties [54]. Acting as a potent regulator, APS primarily operates through the NOTCH signaling pathway and is closely linked to factors such as IL-6, TNF-α, inducible nitric oxide synthase, and CXCL10 [36]. APS enhanced mucosal bacterial defense in vivo and in vitro by activating Toll-like receptor 4 (TLR-4) and nuclear factor κB (NF-κB)/Rel complexes, strengthening innate immunity during infections. This activation led to macrophage activation and the release of nitric oxide (NO) and TNF-α, which aid in inhibiting cancer cell proliferation [37]. An aqueous extract from Astragalus demonstrated protection against methotrexate-induced inhibition of RAW264.7 macrophage proliferation and induced the expression of pro-inflammatory factors associated with RAW264.7 cells [38]. APS shifted the pro-inflammatory and anti-inflammatory signaling balance in favor of an anticancer phenotype by proportionally increasing the M1/M2 macrophage ratio [30]. Evaluation of the anticancer effects of APS in a tissue-engineered tumor model showed that APS-stimulated RAW246.7 macrophages and their supernatants prompted apoptosis and reduced carcinoid tumor size [39].
APS activates macrophages to stimulate B cells, thereby enhancing humoral immune responses [40]. In a study on C4H/HeJ mouse splenocytes, APS effectively induced B-cell proliferation through a pathway that was not TLR3-dependent. This stimulatory effect was primarily mediated by immunoglobulins on the cell surface, offering new insights into the specific mechanisms by which APS regulates B cells within the immune system [18].
As the first line of antitumor immunity, NK cells are vital to the innate immune system. APS regulates the activation and quantity of NK cells, boosting the growth-inhibitory activity of ICIs and immune cell cytotoxicity against tumors. This effect was demonstrated in a mouse model of melanoma. The APS-treated group showed a significant increase in the number of CD3-NK1.1+ NK cells within the mesenteric lymph nodes. Additionally, there was a rise in the expression of activation markers by NK cells, such as IFN-γ, granzyme B, and perforin [31,55]. Moreover, APS synergized with tanshinone to enhance the effectiveness of the chemotherapeutic agent carboplatin in inhibiting melanoma cell proliferation. This was accomplished by enhancing the body’s immune response, increasing the proportion of CD4+ and CD8+ cells in the spleen, and boosting the activity of NK cells and cytotoxic T cells [41].
In the H22 mouse hepatocellular carcinoma (HCC) model, the effect of APS was significant. It triggered apoptosis and reduced further damage to tumor cells while enhancing the levels of cytokines like TNF-α, IL-2, and immune cells, including macrophages, lymphocytes, and NK cells [42]. MHC I chain-associated molecule A (MICA) and B (MICB) are the primary ligands for the activation receptor NKG2D on NK cells. The binding of MICA/B and NKG2D led to NK cell activation by increasing the levels of phosphorylated extracellular signal-regulated kinases. APS also upregulated the expression of MICA and MICB on the surface of H22 tumor cells, stimulating NK cell activation with the release of IFN-γ, granzyme-b, and perforin to promote cancer cell clearance [43].
Traditional cancer treatment methods, such as specific chemotherapy medications, can induce a stress response in malignant cells. The combination of therapeutic approaches, including APS, enhances the expression of NK cell-stimulating proteins in tumor cells, thereby increasing their susceptibility to NK cell-triggered breakdown.

2.2. Anti-Proliferative and Apoptosis-Inducing Effects on Cancer Cells

The anti-proliferation and apoptosis-inducing effects of APS on cancer cells are summarized in Figure 2 and Table 2.

2.2.1. APS Interferes with the Cell Cycle

APS can effectively inhibit cancer cell proliferation by blocking the S, G2/M, and G0/G1 phases of the cell cycle.
APS effectively hindered the growth of cancer cells by blocking their progression through the S phase of the cell cycle. A novel variant of APS, APS4, induced apoptosis in poorly differentiated sarcoma S180 cells and human gastric cancer MGC-803 cells by inhibiting the S phase, and its efficacy increased in a dose-dependent manner [56,57]. Furthermore, nanoparticles carrying APS (APS-SeNPs) significantly increased the number of HepG2 cells arrested in the S phase, resulting in apoptosis via the disruption of mitochondrial function [58].
Apoptosis is triggered through three main pathways: the intrinsic mitochondrial, intrinsic endoplasmic reticulum, and extrinsic death receptor pathways [57]. APS affected the cell cycle and induced apoptosis in cancer cells via the mitochondrial apoptotic pathway [85,86]. Despite exhibiting minimal cytotoxic effects in breast cancer 4T1 cells, APS effectively stimulated macrophage activation [59,61]. Activated macrophages enhanced mitochondrial membrane permeability, increased the Bax/Bcl-2 ratio, and facilitated the release of cytochrome C, thereby amplifying the activities of caspases 3 and 9, which initiate apoptosis. This process also arrested the cell cycle in the G2 phase, effectively inhibiting 4T1 cell growth [59]. Additionally, APS inhibited the proliferation of SW620 colon cancer cells by dominating G2/M phase arrest. APS also protected RAW 264.7 cells from the damaging effects of paclitaxel by altering the drug’s blockage effect in the G2/M phase and impacting the rate of apoptosis [60].
In studies of the G0/G1 phase, APS induced SCG-1 cells to arrest in the G0 phase and MCF-7 cells to arrest in the G1 phase, thus inhibiting the proliferation of cancer cells [61,62]. A study on APS combined with chemotherapeutic anticancer drugs found that the combination of APS and cisplatin intensified the inhibition of the G0/G1- and S-phase cell cycle of the pharyngeal carcinoma cells CNE-1. This combination also increased the expression of p53 [63]. The p53 protein, which is essential for cell cycle checkpoints, especially DNA repair and replication, plays a significant role in various malignant tumors [87].

2.2.2. APS Inhibits Signal Transduction Pathways

APS affects various signaling pathways such as PI3K/Akt, ERK/MAPK, and NF-κB [64,88].
Abnormal activation of the PI3K/Akt pathway in cancer cells enhanced cell survival by phosphorylating downstream target proteins and inhibiting apoptosis [89,90]. When combined with apatinib, APS significantly decreased phosphorylated Akt and MMP-9 expression in AGS human gastric cancer cells, strengthening the antitumor effects of apatinib by inducing autophagy and apoptosis [64]. When paired with Palmarosa sempervirens or radix trichosanthis, APS induces apoptosis in cancer cells, such as B16F10 and A375 melanoma cells, by suppressing PI3K, Akt, and Bcl-2 mRNA expression levels [65,66].
Improper activation of the ERK/MAPK pathway in various cancers is correlated with cell proliferation owing to ERK overphosphorylation [91]. APS enhanced the anticancer properties of apatinib by inhibiting the phosphorylation of RAS, a critical effector of the ERK/MAPK pathway [92]. APS activated the MAPK pathway in mouse macrophages, involving ERK, p38, and JNK, and regulated cell functions such as growth, differentiation, and cell death [67]. In addition, APS enhanced the pro-apoptotic effects of adriamycin in gastric cancer cells by increasing the expression of cysteinyl asparaginase-3, activating the MAPK signaling pathway, inducing DNA fragmentation, and increasing the expression of tumor suppressor genes (SEMA21F, P21WAF1/CIP1, and FBXW7). These findings laid the foundation for further research and development of APS as a chemotherapeutic sensitizer against anticancer drugs [68].
Additionally, APS opposed the activation of NF-κB and decreased the expression of anti-apoptotic genes such as Bcl-2, Bcl-xL, and IAP family members, enhancing cancer cell sensitivity to chemotherapy [69,70]. It effectively reduced mRNA and protein levels of p65 and p50, suppressed the NF-κB pathway’s transcriptional activity, decreased Bcl-xL protein abundance, and altered Cyclin D1 expression in non-small-cell lung cancer (NSCLC) cells. These actions inhibited A549 and NCI-H358 cell proliferation in laboratory experiments and slowed A549 xenograft growth in animal models [69]. In Fang et al.’s research, APS was found to regulate the TLR4/NF-κB pathway by activating the TLR4-TIRAP/Mal-MyD88 pathway and suppressing NF-κB p65 activity by lowering TLR-4, Myd88, Cyclin D, and Bcl-2 protein levels and preventing p65 nuclear translocation in PANC-1 cells. This led to apoptosis induction in PANC-1 cells and inhibited pancreatic cancer progression [70]. Moreover, APS reversed MAPK/NF-κB pathway phosphorylation in bone marrow mesenchymal stromal cells (BMSCs) co-cultured with lung cancer A549 cells, attenuated A549-induced tumorigenesis, and reduced RAS/ERK signaling in the BMSCs by decreasing RAS and ERK protein levels, thereby restraining BMSC proliferation and abnormal morphological changes [71].
The interaction of APS and Salvia extract influenced the TGF-β/MAPK/Smad pathway by modulating microRNA levels, predominantly through the upregulation of miR-145 and the downregulation of miR-21. This process consequently impacted the TGF-β/MAPK/Smad pathway, converting pSmad3L to pSmad3C and increasing pSmad3C protein expression, thereby demonstrating anti-HCC properties [72]. In addition to the classical TGF-β-Smad pathway, TGF-β can also regulate Smad signaling via non-Smad pathways [93]. ERK, JNK, and p38-MAPK can phosphorylate R-Smad (including Smad1/2/3/5/8) and Co-Smad-Smad4 mainly by targeting their linker regions [94]. APS can inhibit hepatocarcinogenesis by modulating TGF-β/Smad signaling through Smad3 phosphorylation mechanisms [73].

2.2.3. APS Induces Apoptosis in Cancer Cells

APS can effectively induce apoptosis in cancer cells by regulating Bcl-2 protein, microRNA, p53 protein, and the Fas receptor [95].
Two typical proteins of the Bcl-2 family, Bcl-2 and Bax, play key roles in cysteine-dependent apoptosis. Bcl-2 is an anti-apoptotic protein located in the nucleus and mitochondrial membrane, and Bax is a pro-apoptotic protein located in the cytoplasm. Bcl-2 inhibits Bax-induced apoptosis, so the decrease in the level of Bcl-2/Bax is considered a pro-apoptotic manifestation [96]. APS induced apoptosis in H22 HCC cells and MDA-MB-231 breast cancer cells by upregulating Bax and downregulating Bcl-2 [74,75]. Similarly, APS can accelerate apoptosis in HepG2 HCC cells by decreasing Bcl-2, β-catenin, c-Myc, and Cyclin D1, which may be related to the Wnt/β-catenin signaling pathway [76]. In both nasopharyngeal and ovarian cancer models, APS was combined with cisplatin to reduce the levels of Bcl-2 and elevate Bax, caspase-3, and caspase-9 levels. APS significantly reduced the Bcl-2/Bax ratio due to the activation of chemotherapeutic agent chemosensitivity in combination compared to treatment with cisplatin alone [77,78].
During tumor development, the expression of various miRNAs changes, leading to corresponding changes in the expression of downstream target genes. miRNAs regulate tumor proliferation, autonomous growth signaling, apoptosis, and other processes that affect tumor progression [97]. MiR-27a is a well-known oncogene in a variety of tumor types [98,99,100]. APS reduced the expression of miR-27a, leading to upregulation of the tumor suppressor gene FBXW7, which ultimately inhibited proliferation and induced apoptosis in OV-90 and SKOV-3 cells [79]. MiR-133a is an oncogene in various cancer types [101,102,103]. APS reduced the expression of MiR-237a by upregulating miR-133a, inactivating the JNK pathway, and exerting anticancer effects in human osteosarcoma MG63 cells [80]. Bioassay analysis showed that the upregulation of miR-195-5p was positively correlated with the survival rate of patients with lung cancer and that APS could inhibit the proliferation and migration of NSCLC cells by increasing the expression level of miR-195-5p [81].
As an important oncogene, the normal function of the p53 protein is an effective barrier to cancer [104]. One of the biological functions of p53 is its ability to induce apoptotic cell suicide, which is central to its role as a tumor suppressor [105]. APS significantly upregulated p53, p21, and p16 in the large-cell lung cancer cell line H460, inhibited the expression of Notch1 and Notch3, and ultimately inhibited proliferation and promoted apoptosis [82].
Fas is a transmembrane receptor that induces apoptosis by crosslinking with agonistic antibodies or Fas ligands (FasL). In addition to its pro-apoptotic function, Fas can also activate many non-apoptotic signaling pathways, and activation of these pathways can lead to increased tumorigenicity and metastasis. Thus, the Fas/FasL system plays an important role in tumor development [106]. APS induced apoptosis in CD133+/CD44+ co-positive colon cancer stem cells in a concentration-dependent manner via the Fas death receptor pathway [83].
Furthermore, APS can trigger ER stress in tumor cells by decreasing the expression of O-GlcNAc transferase (OGT) and increasing the expression of O-GlcNAcase (OGA). Through this mechanism, the APS-upregulated CHOP pathway led to doxorubicin-induced apoptosis in cancer cells [84].

2.3. Other Antitumor Effects of APS

2.3.1. APS Activates Cellular Autophagy

Although various tumor types display diverse genetic and phenotypic characteristics, they often exhibit shared metabolic changes, including alterations in autophagy [107,108]. The suppression of autophagy leads to a significant decrease in tumor cell viability. Combining autophagy inhibitors with anticancer medications can diminish chemotherapy resistance in cancer cells and effectively suppress cancer progression [109,110].
APS can regulate autophagic activity in cancer cells. APS influenced proteins such as beclin-1 and LC3 by activating the PI3K/Akt/mTOR pathway, promoting the formation and maturation of autophagosomes, and accelerating the removal of damaged components from cancer cells. APS also affected the intracellular energy balance and protein synthesis, enhancing the role of autophagy in inhibiting cancer cell growth and inducing apoptosis [64,111,112]. Cellular autophagy can be induced by apatinib or APS alone. However, the combination of the two significantly enhanced the antitumor effect in gastric cancer AGS cells by suppressing the PI3K/Akt/mTOR pathway. Additionally, the combination of apatinib, APS, and the autophagy inhibitor 3-methyladenine effectively inhibited cell growth and increased the rate of apoptosis [64]. At the cellular autophagy level, APS increased autophagic activity in cervical cancer HeLa cells and improved chemosensitivity to cisplatin by upregulating beclin-1, promoting the conversion of LC3I to LC3II, and downregulating p62 [113]. APS reduced xanthine oxidase-induced autophagy in small-cell lung cancer A549 cells by downregulating LC3B and beclin-1 and increasing mTOR expression [111]. APS nanoparticles exhibited cytotoxicity against breast cancer MCF-7 cells by inhibiting autophagy, inducing oxidative stress, generating excess reactive oxygen species, causing mitochondrial damage, and activating apoptotic pathways [112].

2.3.2. APS Inhibits Lipid Metabolism

Lipid metabolism plays an important role in tumorigenesis, invasion, and metastasis [114,115]. Sterol regulatory element binding proteins (SREBPs) are key transcription factors that regulate the expression of lipid synthesis, uptake genes, and play a central role in lipid metabolism under both physiological and pathological conditions; high upregulation of SREBPs in various cancers promotes tumor growth [116].
Abnormal lipid metabolism is accompanied by overexpression of related enzymes, aberrant transcription of non-coding RNAs, and activation of oncogenic signaling pathways [117,118,119]. It has been shown that overexpression of silent information regulator-1 (SIRT-1) inhibited SREBP-1 expression and nuclear translocation by suppressing lipid metabolism through activation of AMPK phosphorylation [120]. APS significantly reduces triglyceride and cholesterol levels in prostate cancer cells. By promoting miR-138-5p targeting to its 3’UTR region, APS inhibited the expression of SIRT-1 and SREBP-1, promoted signaling in the miR-138-5p/ SIRT-1/SREBP-1 pathway in PCa and DU145 cells, and suppressed tumorigenesis and lipid metabolism [121].

2.3.3. APS Affects the Tumour Microenvironment and Regulates Cytokine and Hormone Levels

The TME is crucial for tumor progression, invasion, and dissemination because of its intricate composition [122,123]. Recent studies have emphasized the significant impact of APS on modifying the TME and influencing tumor cell proliferation, apoptosis, and motility [16,24,56,61,122,124,125,126,127,128,129].
APS interacts with the TME through multiple pathways, including the modulation of immune cell functions. For example, when combined with 5-fluorouracil (5-FU) for colon cancer treatment, APS decreased tumor size by reducing indoleamine-2,3-dioxygenase (IDO1) expression in the TME and enhancing CD8+ T-lymphocyte infiltration [124]. In the S180 mouse model, APS altered anaerobic metabolism within the TME by affecting CD3+, CD4+, CD8+ T cells, and CD19+ B cells, leading to tumor cell death [56]. Furthermore, APS co-cultured with HeLa cervical cancer cells boosted peripheral blood mononuclear cell proliferation and adjusted the immune response by reducing suppressive cytokine levels like IL-10 and TGF-β, thereby enhancing antitumor efficacy [125]. In a mouse model of lung cancer, polysaccharides isolated by Li et al. demonstrated antitumor effects in mice with 4-methyl-4-nitro-8-nitrosoguanidine-induced gastric cancer by promoting the proliferation of splenic lymphocytes, enhancing the activity of NK cells, and increasing the levels of IgA, IgG, and IgM, as well as the expression of CD1+ and CD2+/CD1+ in the blood [130].
In addition to immune cells, APS also influences cytokines and chemokines in the TME. It regulated cytokine expression, including nitric oxide and TNF-α, restraining MCF-7 breast cancer cell proliferation by inducing G1-phase cell cycle arrest and apoptosis [61]. In an H22 liver carcinoma model, APS effectively suppressed tumor growth, improved overall health in tumor-bearing mice, and enhanced macrophage phagocytosis. These effects were associated with increased cytokine production and inhibition of IL-10, MDR1 mRNA, and P-glycoprotein by APS [16,24]. In the microenvironment of human HCC, APS blocked chemokine matrix-derived factor-1 and its receptor by inhibiting the activation of the chemokine receptor 4 CXCR4/CXCL12 signaling pathway, restoring cytokine imbalance (elevated IFN-γ expression and reduced IL-4 and IL-10 expression), and inhibiting the immunosuppressive T lymphocyte CD4+CD25+ Treg cell growth, proliferation, and migration [131].
The effect of APS on cytokines within the TME influences cancer cell migration and invasion. In research on lung cancer, APS was found to decrease levels of VEGF and EGFR while enhancing the activity of immune organs, thus inhibiting the progression and spread of tumors [52,126,127]. At the same time, APS was shown to decrease levels of macrophage migration inhibitory factor, reduce the presence of epithelial–mesenchymal transition markers, and gradually diminish the migratory and invasive capabilities of cancer cells in a dose-dependent manner [128,129]. In colon and breast cancer microenvironments, APS inhibited epithelial–mesenchymal transition and metastasis in mice through the WnT5/β-catenin signaling pathway [132,133], inhibited IDO1 expression, and increased intratumoral CD8+ T-cell infiltration [124]. APS enhanced the chemosensitivity of HCC cell lines and inhibited cell migration and invasion in HCC by inhibiting MDR1 and P-glycoprotein efflux pump functions [134].
The effect of APS on the TME involves various pathways that influence cytokines and hormones [55]. In addition to altering the function and activity of immune cells, APS significantly reduces levels of IL-10 and TGF-β while increasing levels of TNF-α, IL-1α, IL-2, IL-6, and IL-12. This regulation affects growth factors like VEGF and EGFR, rejuvenating an immune-suppressed TME.

3. APS-Assisted Cancer Immunotherapy

The assisted effects of APS on cancer immunotherapy are summarized in Figure 3.

3.1. APS Boosts the Effectiveness of Immune Checkpoint Inhibitors in Therapy

ICIs and natural polysaccharide-based substances have shown promise for overcoming the immunosuppressive environment of tumors and enhancing the efficacy of cancer immunotherapy, particularly against solid tumors. APS has demonstrated potential as a therapeutic agent in cancer treatment by influencing PD-1/PD-L1 signaling and related pathways [55,66,135,136,137]. The specific mechanism was demonstrated by the fact that S12, a single-chain variable fragment isolated from APS, bound well to PD-1 and inhibited PD-L1-based T-cell depletion [138]. For example, a traditional Chinese medicine blend known as Bu Zhong Yi Qi Tang (BYD), which contains APS as a key ingredient, effectively reduced PD-L1 expression in gastric cancer tissues and targeted PD-1 and PD-L1 within tumors through the PI3K/Akt/mTOR pathway, resulting in significant suppression [136]. In addition, by inhibiting the Akt/mTOR/p70S6K pathway in tumor-bearing mice, APS downregulated the expression of PD-L1 on the surface of breast cancer 4T1 cells and colorectal cancer CT26 cells, and blocked PD-L1-induced T-cell exhaustion [137]. APS can also decrease PD-L1 expression in cisplatin-resistant melanoma cells, thus increasing their sensitivity to chemotherapy by acting on the PD-L1/PI3K/Akt pathway [66]. In a small retrospective study involving patients with lung cancer treated with ICI therapy, the addition of APS enhanced the antitumor effects of immunotherapy by reducing inflammation, lowering the prognostic marker NLR, and changing the relative proportion of circulating lymphocytes [139].
In combination with anticancer drugs, there was no substantial difference between the effects of ixabepilone and APS on tumor suppression in combination with PD-1 inhibitors, suggesting that APS prevented tumorigenesis or progression in the presence of increased T-cell activation, enhancing the synergistic effect with anti-PD-L1 [138].
APS, derived from the traditional Chinese herb Astragalus membranaceus, enhances anticancer immune responses by modulating the PI3K/Akt/mTOR and Akt/mTOR/p70S6K pathways, which are crucial for cell survival, proliferation, and metabolism. Suppressing these pathways diminishes cancer cell survival and PD-L1 expression on their surfaces. ICIs, which block PD-L1, aid the immune system in recognizing and eliminating cancer cells. Thus, APS not only acts independently but also potentially augments established cancer treatments by boosting the body’s natural immune response to tumors.

3.2. APS Enhances Relay Cellular Immunotherapy

Cellular immunotherapy, also known as permissive immune cell therapy, is an important antitumor therapy. This therapy is carried out by isolating autologous tumor-infiltrating lymphocytes (TILs) or peripheral blood lymphocytes (including lymphokine-activated killer (LAK) cells, CD3-activated killer (CD3-AK) cells, cytokine-induced killer (CIK) cells, and DC-CIK cells), which are activated or genetically modified in vitro, to amplify immune cells with antitumor activity (chimeric antigen receptor T (CAR-T) cells, T-cell receptor-engineered T (TCR-T) cells, and chimeric antigen receptor natural killer (CAR-NK) cells), equip them with the capability of tumor antigen-targeting recognition, and then infuse them back into the tumor patient’s body, thereby amplifying the cellular immune function in the patient’s body and improving the antitumor effect [140,141].
With regard to the effects on peripheral lymphocytes, when APS was combined with CIK cells, the antitumor response of DC-CIK cells was stronger than that of normal DC-CIK cells [142,143,144,145,146,147]. APS can promote the expression of specific markers on the surface of DC on the one hand and significantly enhance the DC-induced T-cell proliferative response; on the other hand, it can enhance the effect of CIK on CD3+ and CD4+ T lymphocytes and exert an antitumor effect. For instance, a study conducted by Zhang and Wang demonstrated that DC-CIK cells induced by APS not only exhibited increased proliferation, but also displayed enhanced cytotoxicity against A549 lung adenocarcinoma and Eca-109 esophageal cancer cells [142,143]. Another study by Zhang revealed that APS used in conjunction with CIK cell therapy effectively controlled tumor growth in the intermediate and advanced stages of NSCLC. This combination led to a significant increase in CD3+ and CD4+ T cells in the peripheral blood, improved the overall physical well-being of patients, and alleviated symptoms associated with qi deficiency syndrome, while maintaining a better safety profile [144]. In addition, the cytotoxicity of CIK cells combined with APS was higher than that of CIK cells alone in HeLa and SKOV3 cells. In vivo results showed that CIK combined with APS significantly inhibited the growth of HeLa-implanted tumors, with an inhibition rate of 80.6% [148].
Stilbene Silver San Liang San (SLS), a Chinese herbal formula consisting of Astragalus, honeysuckle, Angelica sinensis, Glycyrrhiza glabra, and centipede, is used as an adjuvant treatment for B-cell lymphomas with CD19 CAR-T cells. At the cellular level, SLS dose-dependently promoted the killing effect of unmodified T cells and anti-CD19 CAR-T cells on Raji cell lines, while at the same time reducing T-cell and CAR-T cell exhaustion to a certain extent, promoting the proliferation of anti-CD19 CAR-T cells, decreasing the levels of IL-6, IL-10, and TNF-α, and increasing the levels of granzyme B. SLS in in vivo studies effectively increased the anti-B-cell lymphoma function of anti-CD19 CART cells, prolonged the survival of mice, and decreased the levels of IL-6, GM-CSF, and IL-17. The specific mechanism of action of SLS is mediated by the IL-17 signaling pathway and the CD8+ T-cell apoptosis pathway, which regulates CAR-T cells [149]. In addition, a project to explore the role and mechanism of APS in promoting CAR-T cell therapy for HCC based on T-cell subpopulation differentiation is underway [150].

3.3. Utilizing APS as an Adjunct to Enhance the Effectiveness of Cancer Vaccines in Therapy

Cancer-preventive and therapeutic vaccines are key components of cancer-specific active immunotherapy. These vaccines stimulate a patient’s immune system against tumor-specific antigens [151]. However, many vaccines, especially those using peptides and nucleic acids, have a limited ability to activate the immune system. Therefore, their combination with potent adjuvants is crucial to achieve the desired therapeutic results.
Studies have shown that APS can be used as an enhancer of DC-based cancer immunotherapy vaccination [55]. In an animal model of breast cancer, researchers evaluated the adjuvant effects of two polysaccharides extracted from the roots of Astragalus and Radix et Rhizoma Ginseng on DC cancer vaccine-treated mouse 4T1 cells and found that dextran extracted from APS could be an effective alternative to bacterial lipopolysaccharide (LPS) as an adjuvant for the formulation of DC-based cancer immunotherapy vaccines. This combination not only significantly increased the number of CD40, CD80, and CD86 markers in DCs, but also upregulated the secretion of IL-6, TNF-α, and IL-1β. In an animal model of anal sarcoma, the researchers used APS instead of TNF-α to induce maturation of bone marrow-derived DCs, which were then sensitized with the S180 tumor antigen, thereby obtaining a DC tumor vaccine. After immunotherapy in mice, antitumor cytokines such as TNF-α and IL-12 were produced to achieve anticancer effects [152]. It is also possible that the mechanism of this anticancer effect is related to the increase in thymic and splenic indices in mice, which promotes the transfer of the Th1/Th2 imbalance to cellular immunity, in which Th1 cells predominate [153]. In addition, a complex of Astragalus with an aluminum hydroxide adjuvant may enhance anti-breast-cancer activity by inducing an immune response to the basic fibroblast growth factor (bFGF) tumor vaccine in the TME, causing it to produce potent bFGF-specific cytotoxic T-lymphocyte activity [154]. In contrast to typical vaccine adjuvants, APS does not lead to a “cytokine storm” caused by the overexpression of vaccine adjuvants [28].
Although APS and similar herbal therapies have demonstrated encouraging outcomes in both preclinical and clinical trials, achieving complete cancer remission without side effects remains a significant challenge. Ongoing research into APS and other traditional herbal medicines is a vital path in devising more holistic and less detrimental cancer treatment methods. By blending traditional medicine with contemporary medical practices, we can offer a more thorough and patient-centered approach to cancer care.

4. Clinical Trials on the Antitumor Effects of APS

Although no antineoplastic drugs have been able to achieve complete remission of cancer without adverse effects, traditional Chinese herbs have been used for thousands of years in cancer treatment in China, Japan, and other East Asian countries, alleviating the symptoms in patients with cancer, and also mitigating the adverse effects and complications caused by chemotherapy and radiotherapy. These findings further strengthen the complementary role of herbs in modern cancer treatment and provide more options and possibilities for comprehensive treatment [155].
Among these herbs, Astragalus has attracted attention for its potential efficacy in cancer treatment. Several randomized clinical trials have demonstrated that Astragalus-based interventions can reduce cancer symptoms, improve the quality of life and immune function, increase plasma nerve growth factor levels, and slow the progression of chemotherapy-induced peripheral neuropathy [78,156]. Currently, Astragalus extracts or preparations [157] have been used in clinical trials for their antitumor effects on digestive, respiratory, epithelial tissue, and hematological malignancies (Table 3) [158,159,160].

4.1. Malignant Tumors of the Digestive System

According to the fifth edition of the Classification of Tumors of the Digestive System published by the World Health Organization (WHO) in 2019, malignant tumors of the digestive system include esophageal cancer, gastric cancer, cancer of ampullar carcinoma, gobelet cell adenocarcinoma, colorectal cancer, HCC, and pancreatic cancer [182]. APS has been used in clinical antitumor trials for gastric, colorectal, liver, and pancreatic cancers.
Gastric cancer (GC) is the fourth most prevalent cancer globally [183]. Phytochemicals protect against GC through various mechanisms, including the inhibition of cell growth, the induction of cell death and autophagy, the prevention of cell invasion and migration, hindering angiogenesis, combating Helicobacter pylori infection, and impacting the TME [92,184]. Polysaccharides, especially those derived from fungi, algae, tea, APS, Dendrobium, and other edible plants and herbs, are recognized for their anticancer properties against GC [62]. The anti-GC effects of APS are manifested not only in the enhancement of immune activity and the direct inhibition of tumor cell proliferation but also in the alleviation of adverse reactions to anticancer drugs [57,68,170,185,186,187,188,189].
FOLFOX is a globally recognized first-line chemotherapeutic regimen for GC. However, 5-FU and oxaliplatin (L-OHP) may cause adverse reactions such as peripheral neurotoxicity, total hematopenia, and severe gastrointestinal responses [188,189]. Clinical trials have demonstrated that APS injections, when combined with FOLFOX, can reduce gastrointestinal reactions and leukopenia, offering new possibilities for patients with GC to enhance clinical outcomes and physical well-being while mitigating adverse effects [170]. Additionally, clinical studies have investigated the enhancement of advanced GC sensitivity to ICIs by modulating intestinal flora and exploring the metabolomics-based effects of Aiqi Shi Cao Tang (APS) in modulating the gastric environment for low-grade gastric intraepithelial neoplasia treatment [169,190].
Colorectal cancer (CRC) is the third and second most common cancer among men and women, respectively. The reasons for the increase in its occurrence are not fully understood, although genetics, lifestyle, obesity, and environmental factors may play roles [191]. Various studies have demonstrated that herbal medicines containing Astragalus in combination with chemotherapy increased the tumor remission rate, reduced chemotherapy-related side effects such as neutropenia, anemia, nausea, vomiting, and neurotoxicity, and enhanced the quality of life of patients with CRC compared to chemotherapy alone [175,176]. Two additional studies on APS combined with FOLFOX4 demonstrated similar effectiveness [177,178]. Shenqi Fuzheng Injection is a Chinese herbal formula consisting of Astragalus and Codonopsis. This injection was approved by the Chinese Food and Drug Administration in the 1990s [192]. Clinical studies have been conducted on the use of Shenqi Fuzheng Injection in combination with chemotherapy or radiotherapy for the treatment of lung, breast, and colorectal cancers, and some studies have suggested that it may play an important role in the treatment of advanced cancers by improving tumor response and reducing the toxicity of chemotherapy [193,194,195]. A meta-analysis showed that Shenqi Fuzheng Injection combined with chemotherapy regimens (FOLFOX regimen, XELOX regimen) in the treatment of CRC could improve the efficacy of chemotherapy and the quality of survival, enhance cellular immune function, and reduce adverse events such as leukopenia, thrombocytopenia, and gastrointestinal toxicity [194]. Another meta-analysis showed that the efficacy of ginseng Astragalus tonic injection added to conventional chemotherapy for the treatment of lung cancer, breast cancer, and gastrointestinal tract tumors was better than that of conventional chemotherapy alone, and the objective tumor responses were higher than those in the conventional chemotherapy group. Compared with the conventional chemotherapy group, the combination treatment with Shenqi Fuzheng Injection increased NK, CD3+, and CD4+ levels and the CD4+/CD8+ ratio [193]. AC591, a standardized extract of Astragalus and Gui Zhi Wu Tang, is a herbal formulation that improves numbness and pain in the limbs. AC591 can prevent oxaliplatin-induced neuropathy without reducing its antitumor activity against CRC. It is a promising adjunctive drug for the clinical relief of sensory symptoms [196]. An open clinical study of Chinese herbal medicine (Astragalus/tansy) in combination with a novel tumor-lysing vaccine for the treatment of recurrent or metastatic colorectal cancer is underway [174].
Despite the predominant use of Western medical interventions for HCC, the overall outlook for patients is frequently bleak, highlighting the critical need for preventive strategies against HCC [197]. A comprehensive analysis indicated that administering APS in conjunction with transcatheter hepatic artery chemoembolization for HCC therapy surpassed the performance of other Chinese herbal injections, enhancing clinical efficacy and treatment outcomes [198]. A recent clinical investigation was conducted on the combined use of Astragalus/tansychia and a novel lysosomal virus/fusion cell vaccine to treat advanced HCC and lung cancer [167].
The pathogenesis of pancreatic cancer remains unclear. The onset is inconspicuous, with symptoms often evident only in the middle or late stages. Although surgical removal is the ideal approach for patients with intermediate and advanced pancreatic cancer, chemotherapy in conjunction with traditional Chinese medicine can alleviate clinical symptoms, improve the TME, eliminate tumor recurrence factors, and reduce focal metastasis, thereby enhancing efficiency and minimizing toxicity [199].
During the investigation into APS and Shenqi Fuzheng Injection (composed mainly of Radix et Rhizome Ginseng and Radix Astragali) for treating gastrointestinal diseases, APS not only slowed pancreatic tumor growth but also reduced tumor-associated fibroblasts’ ability to secrete βig-H3, thereby promoting the expansion of CD8+ T cells and inhibiting the transition of macrophages to the M2 type, and consequently enhancing the pancreatic tumor’s sensitivity to gemcitabine and other therapies. An observational study found that combining APS with chemotherapy significantly prolonged the survival of patients with metastatic pancreatic cancer compared with chemotherapy alone [173]. Therefore, a randomized double-blind trial is planned to compare the efficacy of APS combined with gemcitabine versus gemcitabine alone as a neoadjuvant treatment for pancreatic cancer [172].
Esophageal cancer is a common tumor of the digestive tract. Because patients with esophageal cancer may have obvious cellular immunosuppression after surgery, some relevant postoperative studies have been conducted. In a postoperative study of 60 patients with thoracic segment esophageal cancer, postoperative intravenous Shenqi Fuzheng Injection significantly improved the cellular immune function after modern two-field resection compared to the group without immunosupportive therapy [200]. Another study showed that four weeks of perioperative treatment with Astragalus-enhancing flour in 37 patients with esophageal cancer resulted in a more pronounced number of mesenchymal mast cells, focal lymphocyte aggregates, increased mesenchymal microvascular damage, and improved immune function [201]. A randomized open clinical trial is currently underway to evaluate APS in conjunction with concurrent chemoradiotherapy for the treatment of locally advanced esophageal cancer [180].

4.2. Malignant Tumors of the Respiratory System

Malignant tumors of the respiratory system include lung, nasal, and laryngeal cancers. APS is being used in clinical antitumor trials related to NSCLC and nasopharyngeal cancer.
NSCLC accounts for the majority of lung cancer cases, with a high rate of recurrence and limited efficacy of current treatments [202]. Astragalus-containing herbs are effective in preventing radiotherapy pneumonia as an adjunctive therapy during conservative radiotherapy [203]. Zhou et al. observed decreased cell growth and increased apoptosis in the H460 NSCLC cell line following administration of APS [204]. In addition, Gan et al. explored the potential of APS for clinical use in lung cancer. Combining platinum-based chemotherapy with Astragalus-based herbal treatments has shown enhanced efficacy in the treatment of advanced NSCLC [205]. By injecting APS in combination with the chemotherapeutic agents vincristine and cisplatin in patients with advanced NSCLC, it was found that APS significantly improved immune function and reduced the toxicity and side effects of chemotherapy and radiotherapy, leading to a better quality of life compared to chemotherapy alone. In light of these benefits, researchers have conducted clinical trials using APS to alleviate cancer-related fatigue induced by chemotherapy in patients with lung cancer, as well as for maintenance therapy after initial or subsequent chemotherapy and targeted therapy [162]. Studies have shown that Shenqi Fuzheng Injection can improve the clinical efficacy and reduce the radiotoxicity in patients with NSCLC [195]. Ten-perfect tonic soup, composed of Astragalus, ginseng, angelica, and other herbs, is commonly used for outpatient chemotherapy in patients with lung cancer. A quality-of-life questionnaire on anticancer drug therapy and another randomized, double-blind, placebo-controlled trial conducted on patients with NSCLC showed significant improvements in both physical symptoms and anorexia after treatment with decapod tonic soup [206,207]. Decapentaplegic tonic soup may also increase the regulatory activity of T cells by decreasing the Foxp3+ Treg population in patients with advanced pancreatic cancer, leading to immune enhancement with various combination therapies [208]. Aidi injection, a commonly used adjuvant chemotherapeutic agent in China, consists of plant extracts of Astragalus, Echinococcus sp., and ginseng, with antitumor, immunomodulatory, and attenuating acute or subacute chemotherapy toxicity. Aidi injection combined with radiotherapy can reduce myelosuppression, radiation pneumonitis, and radiation esophagitis after radiotherapy, and significantly improve the clinical efficacy and quality of life of patients with lung cancer [209,210].
Nasopharyngeal cancer is highly aggressive [211]. Although platinum-based chemotherapy is an effective treatment, the associated adverse effects limit its use [212]. A case–control study linked the use of APS to a decreased incidence of nasopharyngeal cancer [213].

4.3. Other Malignant Tumors

Malignancies of epithelial origin include squamous cell carcinoma, adenocarcinoma, colorectal carcinoma, and breast carcinoma. APS has been tested in clinical antitumor trials related to malignancies of epithelial tissue origin and hematological malignancies.
Metastasis accounts for 90 percent of breast-cancer-related deaths. Treatment options for metastatic breast cancer are limited, emphasizing the urgent need for innovative therapeutic strategies to address this critical issue [214]. Investigations have elucidated the mechanism of action of APS against breast cancer by examining the essential target proteins and mRNA expression. Seventy-three downregulated and seven upregulated genes were identified, with EGFR and ANXA1 identified as the pivotal genes associated with breast cancer progression. Treatment with APS results in a significant decrease in EGFR levels and a marked increase in ANXA1 levels in breast cancer cells [215]. Furthermore, Wang et al. successfully developed a novel nanoparticle (quercetin-3’3-dithiobispropionic acid-Astragaloside-folic acid) that not only hindered drug release into the environment but also facilitated delivery into cancer cells, effectively inhibiting estrogen receptor α-positive breast tumors [216]. Studies have shown that APS treatment can reduce chemotherapy-induced toxicity, leading to research on stage II/III breast cancer patients receiving APS as adjuvant chemotherapy [168], underscoring the importance of ongoing clinical trials on APS in breast cancer care.
Concurrent chemoradiotherapy (CCRT) is a crucial treatment for patients with advanced head and neck squamous cell carcinoma (HNSCC) [217]. A phase II double-blind, randomized trial compared CCRT with APS injection to CCRT with placebo for advanced pharynx or larynx squamous cell carcinoma, and found that the CCRT and APS group had improved safety, enhanced quality of life, and reduced adverse events commonly associated with intense anticancer therapies in patients with HNSCC undergoing CCRT [218].
The enhanced antitumor effects of Astragalus injection in combination with chemotherapy may improve the short-term prognosis and clinical regression of acute lymphoblastic leukemia (ALL) in children [219]. Remission induction chemotherapy can reduce myelosuppression induced by chemotherapeutic agents, increase neutrophil counts, and reduce the incidence and duration of infections in children with ALL [220].

5. Comparison of Anticancer Effects of Astragalus Polysaccharide and Other Polysaccharides

In recent years, polysaccharides isolated from natural sources such as plants, animals, fungi, and seaweeds have received increasing attention owing to their various pharmacological activities, including antitumor, immunomodulatory, antioxidant, and anti-inflammatory activities [221,222,223,224]. Pectin polysaccharides from plants, β-glucans from mushrooms, and sulfated polysaccharides from seaweeds have been shown to possess antioxidant and immunomodulatory activities [225,226,227]. Astragalus, mushrooms, Ganoderma lucidum, and fucoidan polysaccharides are used in domestic and international markets.

5.1. Astragalus Polysaccharide and β-Glucan

β-glucan is found in the cell walls of mushrooms, yeast, and grains such as oats and barley [228]. Both β-glucan and Astragalus polysaccharide are known for their effects on modulating immune function. However, β-glucan generally activates white blood cells such as macrophages and neutrophils, whereas Astragalus polysaccharide tends to enhance the body’s production of various immune cells [229]. In terms of applications, β-glucans are commonly used in immune-enhancing supplements and food additives, while Astragalus polysaccharide is often used in herbal formulations to enhance vitality and treat diseases such as colds or flu [230,231]. Among the β-glucans, shiitake polysaccharide is a typical T-cell activator that enhances the immune function of immune cells such as T cells, macrophages, and natural killer cells, and has antitumor effects [232]. Polysaccharide peptides extracted from medicinal mushrooms induced Molt4 leukemia cells to enter the S phase, eventually leading to apoptosis [233,234]. Ganoderma lucidum polysaccharides reduced ERK expression and inhibited the proliferation of MT-1 human breast malignant carcinoma cells tumor cells through the ERK signaling pathway [235]. Lycium barbarum polysaccharides (LBPs) enhanced macrophages in a dose-dependent manner [236]. They exhibited antitumor activity against many types of cancer cells, and inhibited tumor growth in nude mice by inducing apoptosis and cell cycle blockade [237,238]. Ke et al. reported that LBP inhibited the growth of the human bladder cancer cell line BIU87 and induced apoptosis in BIU87 cells [239].

5.2. Astragalus Polysaccharide and Pectin Polysaccharide

Pectin is a polysaccharide derived from citrus fruits and apples that can form gels and is more structurally complex with more branching than APS [240,241]. Pectin is primarily used to improve digestive health and reduce cholesterol [242]. In addition, APS has antioxidant properties that help neutralize free radicals and protect cells from oxidative damage [243]. Pectin mainly produces reactive oxygen species to induce apoptosis, which is the opposite mechanism of action of APS [244]. Pectin is widely used as a gelling agent in jams and preserves by the food industry, whereas APS is mainly found in dietary supplements and traditional medicine [23,245].
In addition to their benefits as soluble dietary fibers, modified pectins, particularly modified citrus pectins, have beneficial effects on the development and spread of malignant tumors, including colon and breast cancer [245]. Pectin-derived chemicals slow cell development and promote apoptosis [246]. The combination of the multiplant compounds BreastDefend or ProstaCaid with modified citrus pectin reduced the invasive potential of highly metastatic human breast cancer MDA-MB-231 cells and may prevent the development of breast cancer in mice by reducing angiogenesis [247]. Furthermore, in addition to activating macrophages, citrus pectin inhibited activator protein 1 and NF-κB signaling and activated LPS/TLR-4 signaling [248]. Citrus and apple pectin inhibited the growth of MDA-MB-231, MCF-7, and T47D breast cancer cells by inhibiting the S phase and G1 phase or G2/M phase of the cell cycle. They stimulated the intrinsic mitochondrial apoptotic pathway by generating reactive oxygen species and inhibiting matrix metalloproteinases in breast cancer cells, thereby inducing cell death through DNA damage [249].

5.3. Astragalus Polysaccharide and Xanthanate Gum

Xanthan gum is a naturally occurring polysaccharide obtained from Xanthomonas campestris with a β-d-glucose linkage in its main chain, similar to cellulose [250]. Unlike xanthan polysaccharides, which have therapeutic properties, xanthan gum is mainly used in foods as a thickening and stabilizing agent and has few direct health benefits [251]. However, some studies have confirmed that xanthan gum has antitumor effects, in addition to acting as an adjuvant to modulate immunity [252]. In vitro cultured macrophages were induced to produce IL-12 and TNF-α in an MyD88-dependent manner by xanthan gum, being recognized by TLR4. In in vivo experiments, oral administration of xanthan gum significantly delayed tumor growth and prolonged survival in mice with melanoma and enhanced NK cell activity and CD8+ T-cell tumor specificity [253].

5.4. Astragalus Polysaccharide and Marine Complex Polysaccharide Substances

In the search for novel polysaccharides with antitumor properties, marine resources have attracted considerable attention because of their unique biological activities [254,255,256]. Fucoidans are biologically active sulfated polysaccharides found in brown algae. They have great structural diversity and a wide range of bioactivities and have been shown to be useful in antitumor therapy, cardiovascular disease risk reduction, and the treatment of gastric ulcers, which differ from the therapeutic effects of APS for a large part of the population [257]. Studies have shown that fucoidan exerts anticancer effects by mediating different signaling pathways to regulate apoptosis, inhibit tumor metastasis, and enhance the toxic effects of chemical drugs [258,259,260,261]. The anticancer properties of fucoidans are believed to be determined by their chemical structures and are closely related to their molecular weights; the larger the molecular weight and the higher the water solubility of polysaccharides, the stronger their antitumor activity [260]. As an adjuvant, fucoidan enhances the efficacy of ICIs in the treatment of melanoma and metastatic lung cancer [260,262,263]. In a clinical study, the combination of fucoidan and tamoxifen showed favorable efficacy for the treatment of breast cancer, and no adverse effects were detected [264]. This suggests that fucoidan could be used as an effective adjuvant in combination with other hormonal therapeutic agents. In addition, sulfated polysaccharides purified from marine microalgae showed cytotoxicity against the colorectal cancer HTC-116, breast cancer MCF-7, and human leukemia HL-60 cell lines [265].

5.5. Production of Astragalus Polysaccharide

Although APS has shown promise in laboratory and clinical trials, research on its specific mechanisms and long-term effects is relatively limited. APS is less well studied than longer-established and more widely used polysaccharides such as β-glucan and pectin. In terms of accessibility and assessment of production costs, APS is primarily derived from Astragalus root, which may limit the global availability and scale of production. Fucoidan may also be affected by the quality and safety of brown algae due to environmental factors such as marine pollution. The cost is high compared to that of more readily available sources, such as citrus fruits and marine complex polysaccharides. While APS is generally considered safe, there may be safety concerns and side effects when used at high doses or over long periods of time. For example, when associated with certain drugs for autoimmune diseases, immune activation may result. APS requires more careful regulation than food-grade polysaccharides such as gum and xanthan gum. Thus, while APS offers significant advantages in terms of immune enhancement and anti-inflammatory and antioxidant properties, it may have disadvantages in terms of the depth of research, cost-effectiveness, and safety. When selecting a suitable polysaccharide supplement, consumers should consider these factors and make decisions based on their health status and needs.

6. Conclusions and Outlook

In the current landscape of cancer therapy, APS has been recognized for its immunomodulatory potential as a beneficial bioactive substance with various promising applications. APS is considered a suitable adjuvant drug with no significant safety issues [266]; however, numerous studies have highlighted its role in enhancing patient quality of life by reducing cancer-related fatigue and symptoms and modulating inflammatory responses [23,52].
This review comprehensively evaluates the anticancer mechanisms of APS, focusing on its potential use in cancer immunotherapy. The results underscore the ability of APS to activate and modulate immune cells, disrupt the cell cycle, induce cancer cell apoptosis, trigger cellular autophagy, and regulate signal transduction pathways, especially showing potential in enhancing the sensitivity of antitumor therapy, attenuating the treatment-related side effects, and reversing drug resistance. The advantages and disadvantages of APS in comparison with other polysaccharides were also considered.
Nonetheless, several key issues must be addressed to realize the widespread clinical application of APS. First, the bioavailability of APS is low, mainly due to the complex structure of its macromolecules, making its absorption in the gastrointestinal tract inefficient [267]. In addition, owing to differences in plant sources and extraction methods, there may be variability in the APS composition, which affects the consistency and reproducibility of the clinical results [268]. Therefore, it is crucial to develop standardized extraction and purification processes to ensure product quality. Second, although APS is generally considered safe, the issue of dose optimization in terms of potential interaction with other drugs and side effects in specific populations requires further research [269].
Future studies should focus on gaining a deeper understanding of the specific mechanism of action and pharmacokinetic properties of APS and revealing the molecular targets of APS through high-throughput screening, genomic studies, and advanced imaging techniques to develop effective clinical strategies. Meanwhile, exploring innovative delivery methods, such as extended-release gel systems for injection, may provide solutions to improve its absorption and sustained release in vivo and reduce the need for frequent dosing in long-term therapy [270]. Through these efforts, APS is expected to play a greater role in modern healthcare systems, especially in the field of cancer therapy.

Author Contributions

L.Z. and Z.X. designed this study. Z.H. and X.L. wrote the manuscript and constructed the figures and tables. S.Q., Q.Y. and J.N. provided detailed guidance and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Scientific and Technological Innovation Major Base of Guangxi (No. 82350106).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Stoop, T.F.; Theijse, R.T.; Seelen, L.W.F.; Groot Koerkamp, B.; Van Eijck, C.H.J.; Wolfgang, C.L.; Van Tienhoven, G.; Van Santvoort, H.C.; Molenaar, I.Q.; Wilmink, J.W.; et al. Preoperative Chemotherapy, Radiotherapy and Surgical Decision-Making in Patients with Borderline Resectable and Locally Advanced Pancreatic Cancer. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 101–124. [Google Scholar] [CrossRef] [PubMed]
  3. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef] [PubMed]
  4. Engle, K.; Kumar, G. Cancer Multidrug-Resistance Reversal by ABCB1 Inhibition: A Recent Update. Eur. J. Med. Chem. 2022, 239, 114542. [Google Scholar] [CrossRef] [PubMed]
  5. Abdelbaky, S.B.; Ibrahim, M.T.; Samy, H.; Mohamed, M.; Mohamed, H.; Mustafa, M.; Abdelaziz, M.M.; Forrest, M.L.; Khalil, I.A. Cancer Immunotherapy from Biology to Nanomedicine. J. Control. Release 2021, 336, 410–432. [Google Scholar] [CrossRef]
  6. Nygren, P.; SBU-group. Swedish Council on Technology Assessment in Health Care What Is Cancer Chemotherapy? Acta Oncol. 2001, 40, 166–174. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, D.; Kim, S.W.; Charchoghlyan, H.; Jeong, H.; Han, G.D. Combinatorial Herbal Extracts Alleviate Alcohol-Induced Hepatic Disorders. Plant Foods Hum. Nutr. 2023, 78, 432–438. [Google Scholar] [CrossRef] [PubMed]
  8. Pu, R.; Zhang, L.; Zhou, X.; Yin, W.; Zhang, X.; Liu, H.; Duan, X.; Yu, C.; Luo, X.; Zhong, Q.; et al. Strategy of Using Herbal Extract Preparations for Stage IV Cancer in Palliative Care: An Enhanced and Maintenance Therapy. J. Clin. Oncol. 2016, 34, 89. [Google Scholar] [CrossRef]
  9. Nai, J.; Zhang, C.; Shao, H.; Li, B.; Li, H.; Gao, L.; Dai, M.; Zhu, L.; Sheng, H. Extraction, Structure, Pharmacological Activities and Drug Carrier Applications of Angelica Sinensis Polysaccharide. Int. J. Biol. Macromol. 2021, 183, 2337–2353. [Google Scholar] [CrossRef]
  10. Wang, Y.; Zhang, Q.; Chen, Y.; Liang, C.L.; Liu, H.; Qiu, F.; Dai, Z. Antitumor Effects of Immunity-Enhancing Traditional Chinese Medicine. Biomed. Pharmacother. 2020, 121, 109570. [Google Scholar] [CrossRef]
  11. Tian, X.; Liang, T.; Liu, Y.; Ding, G.; Zhang, F.; Ma, Z. Extraction, Structural Characterization, and Biological Functions of Lycium barbarum Polysaccharides: A Review. Biomolecules 2019, 9, 389. [Google Scholar] [CrossRef] [PubMed]
  12. Sun, K.; Wu, L.; Wang, S.; Deng, W. Antitumor Effects of Chinese Herbal Medicine Compounds and Their Nano-Formulations on Regulating the Immune System Microenvironment. Front. Oncol. 2022, 12, 949332. [Google Scholar] [CrossRef] [PubMed]
  13. Jin, M.; Zhao, K.; Huang, Q.; Shang, P. Structural Features and Biological Activities of the Polysaccharides from Astragalus membranaceus. Int. J. Biol. Macromol. 2014, 64, 257–266. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, M.; Zhang, Z.F.; Ding, Y.; Wang, J.B.; Li, Y. Astragalus Polysaccharide Improves Palmitate-Induced Insulin Resistance by Inhibiting PTP1B and NF-κB in C2C12 Myotubes. Molecules 2012, 17, 7083–7092. [Google Scholar] [CrossRef] [PubMed]
  15. Fan, Y.; Hu, Y.; Wang, D.; Liu, J.; Zhang, J.; Zhao, X.; Liu, X.; Liu, C.; Yuan, J.; Ruan, S. Effects of Astragalus Polysaccharide Liposome on Lymphocyte Proliferation In Vitro and Adjuvanticity In Vivo. Carbohydr. Polym. 2012, 88, 68–74. [Google Scholar] [CrossRef]
  16. Tian, Q.-E.; Li, H.-D.; Yan, M.; Cai, H.-L.; Tan, Q.-Y.; Zhang, W.-Y. Astragalus Polysaccharides Can Regulate Cytokine and P-Glycoprotein Expression in H22 Tumor-Bearing Mice. World J. Gastroenterol. 2012, 18, 7079–7086. [Google Scholar] [CrossRef] [PubMed]
  17. Gong, H.; Li, W.; Sun, J.; Jia, L.; Guan, Q.; Guo, Y.; Wang, Y. A Review on Plant Polysaccharide Based on Drug Delivery System for Construction and Application, with Emphasis on Traditional Chinese Medicine Polysaccharide. Int. J. Biol. Macromol. 2022, 211, 711–728. [Google Scholar] [CrossRef] [PubMed]
  18. Xu, Q.; Cheng, W.; Wei, J.; Ou, Y.; Xiao, X.; Jia, Y. Synergist for Antitumor Therapy: Astragalus Polysaccharides Acting on Immune Microenvironment. Discov. Oncol. 2023, 14, 179. [Google Scholar] [CrossRef] [PubMed]
  19. Yang, J.; Qin, L.; Huang, J.; Li, Y.; Xu, S.; Wang, H.; Zhu, S.; Wang, J.; Zhu, B.; Li, F.; et al. Astragalus Polysaccharide Attenuates LPS-Related Inflammatory Osteolysis by Suppressing Osteoclastogenesis by Reducing the MAPK Signalling Pathway. J. Cell. Mol. Med. 2021, 25, 6800–6814. [Google Scholar] [CrossRef]
  20. Long, H.; Lin, H.; Zheng, P.; Hou, L.; Zhang, M.; Lin, S.; Yin, K.; Zhao, G. WTAP Mediates the Anti-Inflammatory Effect of Astragalus mongholicus Polysaccharide on THP-1 Macrophages. Front. Pharmacol. 2022, 13, 1023878. [Google Scholar] [CrossRef]
  21. Yan, X.; Miao, J.; Zhang, B.; Liu, H.; Ma, H.; Sun, Y.; Liu, P.; Zhang, X.; Wang, R.; Kan, J.; et al. Study on Semi-Bionic Extraction of Astragalus Polysaccharide and Its Anti-Aging Activity in Vivo. Front. Nutr. 2023, 10, 1201919. [Google Scholar] [CrossRef] [PubMed]
  22. Dong, M.; Li, J.; Yang, D.; Li, M.; Wei, J. Biosynthesis and Pharmacological Activities of Flavonoids, Triterpene Saponins and Polysaccharides Derived from Astragalus membranaceus. Molecules 2023, 28, 5018. [Google Scholar] [CrossRef] [PubMed]
  23. Zheng, Y.; Ren, W.; Zhang, L.; Zhang, Y.; Liu, D.; Liu, Y. A Review of the Pharmacological Action of Astragalus Polysaccharide. Front. Pharmacol. 2020, 11, 349. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, B.; Xiao, B.; Sun, T. Antitumor and Immunomodulatory Activity of Astragalus membranaceus Polysaccharides in H22 Tumor-Bearing Mice. Int. J. Biol. Macromol. 2013, 62, 287–290. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, L.; Bai, S.P.; Zhao, L.; Wang, X.H. Astragalus Polysaccharide Injection Integrated with Vinorelbine and Cisplatin for Patients with Advanced Non-Small Cell Lung Cancer: Effects on Quality of Life and Survival. Med. Oncol. 2012, 29, 1656–1662. [Google Scholar] [CrossRef] [PubMed]
  26. Lim, S.M.; Park, H.B.; Jin, J.O. Polysaccharide from Astragalus membranaceus Promotes the Activation of Human Peripheral Blood and Mouse Spleen Dendritic Cells. Chin. J. Nat. Med. 2021, 19, 56–62. [Google Scholar] [CrossRef] [PubMed]
  27. Zhu, N.; Lv, X.; Wang, Y.; Li, J.; Liu, Y.; Lu, W.; Yang, L.; Zhao, J.; Wang, F.; Zhang, L.W. Comparison of Immunoregulatory Effects of Polysaccharides from Three Natural Herbs and Cellular Uptake in Dendritic Cells. Int. J. Biol. Macromol. 2016, 93, 940–951. [Google Scholar] [CrossRef]
  28. Chang, W.T.; Lai, T.H.; Chyan, Y.J.; Yin, S.Y.; Chen, Y.H.; Wei, W.C.; Yang, N.S. Specific Medicinal Plant Polysaccharides Effectively Enhance the Potency of a DC-Based Vaccine against Mouse Mammary Tumor Metastasis. PLoS ONE 2015, 10, e0122374. [Google Scholar] [CrossRef]
  29. Pang, G.; Chen, C.; Liu, Y.; Jiang, T.; Yu, H.; Wu, Y.; Wang, Y.; Wang, F.J.; Liu, Z.; Zhang, L.W. Bioactive Polysaccharide Nanoparticles Improve Radiation-Induced Abscopal Effect through Manipulation of Dendritic Cells. ACS Appl. Mater. Interfaces 2019, 11, 42661–42670. [Google Scholar] [CrossRef]
  30. Bamodu, O.A.; Kuo, K.T.; Wang, C.H.; Huang, W.C.; Wu, A.T.H.; Tsai, J.T.; Lee, K.Y.; Yeh, C.T.; Wang, L.S. Astragalus Polysaccharides (PG2) Enhances the M1 Polarization of Macrophages, Functional Maturation of Dendritic Cells, and T Cell-Mediated Anticancer Immune Responses in Patients with Lung Cancer. Nutrients 2019, 11, 2264. [Google Scholar] [CrossRef]
  31. Hwang, J.; Zhang, W.; Dhananjay, Y.; An, E.K.; Kwak, M.; You, S.; Lee, P.C.W.; Jin, J.O. Astragalus membranaceus Polysaccharides Potentiate the Growth-Inhibitory Activity of Immune Checkpoint Inhibitors against Pulmonary Metastatic Melanoma in Mice. Int. J. Biol. Macromol. 2021, 182, 1292–1300. [Google Scholar] [CrossRef] [PubMed]
  32. Zhou, X.; Liu, Z.; Long, T.; Zhou, L.; Bao, Y. Immunomodulatory Effects of Herbal Formula of Astragalus Polysaccharide (APS) and Polysaccharopeptide (PSP) in Mice with Lung Cancer. Int. J. Biol. Macromol. 2018, 106, 596–601. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, Z.; Wang, D.; Qi, Y.; Liu, J.; Zhou, T.; Rao, W.; Hu, K. Autologous-Cancer-Cryoablation-Mediated Nanovaccine Augments Systematic Immunotherapy. Mater. Horiz. 2023, 10, 1661–1677. [Google Scholar] [CrossRef]
  34. Xu, S.; Wu, Z.; Cai, G.; Zhang, Y.; Peng, S.; Jiao, L.; Liu, Z.; Yang, Y.; Wang, D. Astragalus Polysaccharides Combined with Simvastatin as an Immunostimulant Enhances the Immune Adjuvanticity of Oil-in-Water Emulsion and Immune Responses in Mice. Vaccine 2023, 41, 1684–1693. [Google Scholar] [CrossRef] [PubMed]
  35. Meng, Y.; Zhang, Y.; Jia, N.; Qiao, H.; Zhu, M.; Meng, Q.; Lu, Q.; Zu, Y. Synthesis and Evaluation of a Novel Water-Soluble High Se-Enriched Astragalus Polysaccharide Nanoparticles. Int. J. Biol. Macromol. 2018, 118, 1438–1448. [Google Scholar] [CrossRef] [PubMed]
  36. Wei, W.; Li, Z.P.; Bian, Z.X.; Han, Q.B. Astragalus Polysaccharide RAP Induces Macrophage Phenotype Polarization to M1 via the Notch Signaling Pathway. Molecules 2019, 24, 2016. [Google Scholar] [CrossRef] [PubMed]
  37. Wei, W.; Xiao, H.T.; Bao, W.R.; Ma, D.L.; Leung, C.H.; Han, X.Q.; Ko, C.H.; Lau, C.B.S.; Wong, C.K.; Fung, K.P.; et al. TLR-4 May Mediate Signaling Pathways of Astragalus Polysaccharide RAP Induced Cytokine Expression of RAW264.7 Cells. J. Ethnopharmacol. 2016, 179, 243–252. [Google Scholar] [CrossRef]
  38. Lee, Y.S.; Han, O.K.; Park, C.W.; Yang, C.H.; Jeon, T.W.; Yoo, W.K.; Kim, S.H.; Kim, H.J. Pro-Inflammatory Cytokine Gene Expression and Nitric Oxide Regulation of Aqueous Extracted Astragali Radix in RAW 264.7 Macrophage Cells. J. Ethnopharmacol. 2005, 100, 289–294. [Google Scholar] [CrossRef]
  39. Li, W.; Hu, X.; Wang, S.; Wang, H.; Parungao, R.; Wang, Y.; Liu, T.; Song, K. Detection and Evaluation of Anti-Cancer Efficiency of Astragalus Polysaccharide via a Tissue Engineered Tumor Model. Macromol. Biosci. 2018, 18, 1800223. [Google Scholar] [CrossRef]
  40. Shao, B.M.; Xu, W.; Dai, H.; Tu, P.; Li, Z.; Gao, X.M. A Study on the Immune Receptors for Polysaccharides from the Roots of Astragalus membranaceus, a Chinese Medicinal Herb. Biochem. Biophys. Res. Commun. 2004, 320, 1103–1111. [Google Scholar] [CrossRef]
  41. Wu, J.; Xu, H.; Zhang, L.; Zhang, X. Radix Astragali and Tanshinone Help Carboplatin Inhibit B16 Tumor Cell Growth. Technol. Cancer Res. Treat. 2016, 15, 583–588. [Google Scholar] [CrossRef]
  42. Yu, J.; Ji, H.; Liu, A. Alcohol-Soluble Polysaccharide from Astragalus membranaceus: Preparation, Characteristics and Antitumor Activity. Int. J. Biol. Macromol. 2018, 118, 2057–2064. [Google Scholar] [CrossRef] [PubMed]
  43. Mu, J.Y.; Li, Y.D.; Zhao, X.K.; Li, J.; Yang, A.J. Astragalus Polysaccharide Restores Activation of NK Cells in Radiation Therapy of Tumors. Int. J. Clin. Exp. Med. 2019, 12, 8609–8621. [Google Scholar]
  44. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic Cells in Cancer Immunology and Immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef] [PubMed]
  45. Banchereau, J.; Briere, F.; Caux, C.; Davoust, J.; Lebecque, S.; Liu, Y.J.; Pulendran, B.; Palucka, K. Immunobiology of Dendritic Cells. Annu. Rev. Immunol. 2000, 18, 767–811. [Google Scholar] [CrossRef] [PubMed]
  46. Joffre, O.P.; Segura, E.; Savina, A.; Amigorena, S. Cross-Presentation by Dendritic Cells. Nat. Rev. Immunol. 2012, 12, 557–569. [Google Scholar] [CrossRef] [PubMed]
  47. Cao, Y.; Chen, Z.; Sun, L.; Lin, Y.; Yang, Y.; Cui, X.; Wang, C. Herb Polysaccharide-Based Drug Delivery System: Fabrication, Properties, and Applications for Immunotherapy. Pharmaceutics 2022, 14, 1703. [Google Scholar] [CrossRef] [PubMed]
  48. Sanmamed, M.F.; Chen, L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell 2019, 176, 677. [Google Scholar] [CrossRef] [PubMed]
  49. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer Immunoediting and Resistance to T Cell-Based Immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  50. Raskov, H.; Orhan, A.; Christensen, J.P.; Gögenur, I. Cytotoxic CD8+ T Cells in Cancer and Cancer Immunotherapy. Br. J. Cancer 2021, 124, 359–367. [Google Scholar] [CrossRef]
  51. Banstola, A.; Jeong, J.H.; Yook, S. Immunoadjuvants for Cancer Immunotherapy: A Review of Recent Developments. Acta Biomater. 2020, 114, 16–30. [Google Scholar] [CrossRef] [PubMed]
  52. Li, C.X.; Liu, Y.; Zhang, Y.Z.; Li, J.C.; Lai, J. Astragalus Polysaccharide: A Review of Its Immunomodulatory Effect. Arch. Pharm. Res. 2022, 45, 367–389. [Google Scholar] [CrossRef] [PubMed]
  53. Crotty, S. Follicular Helper CD4+ T Cells (TFH). Annu. Rev. Immunol. 2011, 29, 621–663. [Google Scholar] [CrossRef] [PubMed]
  54. Sheu, K.M.; Hoffmann, A. Functional Hallmarks of Healthy Macrophage Responses: Their Regulatory Basis and Disease Relevance. Annu. Rev. Immunol. 2022, 40, 295–321. [Google Scholar] [CrossRef] [PubMed]
  55. Kong, F.; Chen, T.; Li, X.; Jia, Y. The Current Application and Future Prospects of Astragalus Polysaccharide Combined With Cancer Immunotherapy: A Review. Front. Pharmacol. 2021, 12, 737674. [Google Scholar] [CrossRef] [PubMed]
  56. Yu, J.; Dong, X.; Jiao, J.; Ji, H.; Liu, A. Antitumor and Immunoregulatory Activities of a Novel Polysaccharide from Astragalus membranaceus on S180 Tumor-Bearing Mice. Int. J. Biol. Macromol. 2021, 189, 930–938. [Google Scholar] [CrossRef] [PubMed]
  57. Yu, J.; Ji, H.; Dong, X.; Feng, Y.; 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. 2019, 126, 811–819. [Google Scholar] [CrossRef] [PubMed]
  58. Jiao, J.; Yu, J.; Ji, H.; Liu, A. Synthesis of Macromolecular Astragalus Polysaccharide-Nano Selenium Complex and the Inhibitory Effects on HepG2 Cells. Int. J. Biol. Macromol. 2022, 211, 481–489. [Google Scholar] [CrossRef] [PubMed]
  59. Li, W.; Hu, X.; Wang, S.; Jiao, Z.; Sun, T.; Liu, T.; Song, K. Characterization and Anti-Tumor Bioactivity of Astragalus Polysaccharides by Immunomodulation. Int. J. Biol. Macromol. 2020, 145, 985–997. [Google Scholar] [CrossRef]
  60. Bao, W.R.; Li, Z.P.; Zhang, Q.W.; Li, L.F.; Liu, H.B.; Ma, D.L.; Leung, C.H.; Lu, A.P.; Bian, Z.X.; Han, Q.B. Astragalus Polysaccharide RAP Selectively Attenuates Paclitaxel-Induced Cytotoxicity Toward RAW 264.7 Cells by Reversing Cell Cycle Arrest and Apoptosis. Front. Pharmacol. 2019, 9, 1580. [Google Scholar] [CrossRef]
  61. Li, W.; Song, K.; Wang, S.; Zhang, C.; Zhuang, M.; Wang, Y.; Liu, T. Anti-Tumor Potential of Astragalus Polysaccharides on Breast Cancer Cell Line Mediated by Macrophage Activation. Mater. Sci. Eng. C 2019, 98, 685–695. [Google Scholar] [CrossRef]
  62. Chen, L.; He, C.; Zhou, M.; Long, J.; Li, L. Research Progress on the Mechanisms of Polysaccharides against Gastric Cancer. Molecules 2022, 27, 5828. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, Y.L.; Lin, Z.W.; He, P.T.; Nie, H.; Yao, Q.Y.; Zhang, S.Y. Inhibitory Effect of Astragalus Polysaccharide Combined with Cisplatin on Cell Cycle and Migration of Nasopharyngeal Carcinoma Cell Lines. Biol. Pharm. Bull. 2021, 44, 926–931. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, J.; Yu, J.; Wang, J.; Zhang, C.; Shang, K.; Yao, X.; Cao, B. Astragalus Polysaccharide Enhanced Antitumor Effects of Apatinib in Gastric Cancer AGS Cells by Inhibiting AKT Signalling Pathway. Biomed. Pharmacother. 2018, 100, 176–183. [Google Scholar] [CrossRef]
  65. Wang, F.; Bai, J.; Li, F.; Liu, J.; Wang, Y.; Li, N.; Wang, Y.; Xu, J.; Liu, W.; Xu, L.; et al. Investigation of the Mechanism of the Anti-Cancer Effects of Astragalus Propinquus Schischkin and Pinellia Pedatisecta Schott (A&P) on Melanoma via Network Pharmacology and Experimental Verification. Front. Pharmacol. 2022, 13, 895738. [Google Scholar] [CrossRef]
  66. Gong, Q.; Yu, H.; Ding, G.; Ma, J.; Wang, Y.; Cheng, X. Suppression of Stemness and Enhancement of Chemosensibility in the Resistant Melanoma Were Induced by Astragalus Polysaccharide through PD-L1 Downregulation. Eur. J. Pharmacol. 2022, 916, 174726. [Google Scholar] [CrossRef]
  67. 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. 2021, 21, 20. [Google Scholar] [CrossRef]
  68. Song, J.; Peng, L. IDDF2019-ABS-0248 Astragalus Polysaccharide Promotes Adriamycin-Induced Apoptosis in Gastric Cancer Cells. Gut 2019, 68, A25–A26. [Google Scholar] [CrossRef]
  69. Wu, C.Y.; Ke, Y.; Zeng, Y.F.; Zhang, Y.W.; Yu, H.J. Anticancer Activity of Astragalus Polysaccharide in Human Non-Small Cell Lung Cancer Cells. Cancer Cell Int. 2017, 17, 115. [Google Scholar] [CrossRef]
  70. Fang, J.; Wu, Z.; Wang, J.; Feng, J.; Ying, L. Astragalus Polysaccharides Inhibit Pancreatic Cancer Progression by Downregulation of TLR4/NF-κB Signaling Pathway. J. Nanomater. 2022, 2022, 5351611. [Google Scholar] [CrossRef]
  71. Zhang, Y.M.; Liu, Y.Q.; Liu, D.; Zhang, L.; Qin, J.; Zhang, Z.; Su, Y.; Yan, C.; Luo, Y.L.; 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. 2019, 25, 4110–4121. [Google Scholar] [CrossRef]
  72. Wu, C.; Chen, W.; Fang, M.; Boye, A.; Tao, X.; Xu, Y.; Hou, S.; Yang, Y. Compound Astragalus and Salvia Miltiorrhiza Extract Inhibits Hepatocellular Carcinoma Progression via miR-145/miR-21 Mediated Smad3 Phosphorylation. J. Ethnopharmacol. 2019, 231, 98–112. [Google Scholar] [CrossRef]
  73. Liu, X.; Yang, Y.; Zhang, X.; Xu, S.; He, S.; Huang, W.; Roberts, M.S. Compound Astragalus and Salvia miltiorrhiza Extract Inhibits Cell Invasion by Modulating Transforming Growth Factor-β/Smad in HepG2 Cell. J. Gastroenterol. Hepatol. 2010, 25, 420–426. [Google Scholar] [CrossRef] [PubMed]
  74. Lai, X.; Xia, W.; Wei, J.; Ding, X. Therapeutic Effect of Astragalus Polysaccharides on Hepatocellular Carcinoma H22-Bearing Mice. Dose-Response 2017, 15, 155932581668518. [Google Scholar] [CrossRef] [PubMed]
  75. Xie, R.D.; Sun, S.B.; He, J.X.; Dong, J.N.; Long, F. Effect of astragalus polysaccharide on growth and tumor-related apoptosis protein of human breast cancer Mda-Mb-231 transplanted tumor in nude mice. Chin. J. Exp. Tradit. Med. Form. 2019, 25, 27–43. [Google Scholar] [CrossRef]
  76. Lv, J.; Zhu, P.F.; Liu, Y.M.; Zeng, Q.L.; Yu, Z.J. Astragalus polysaccharides downregulates apoptosis in Hepg2 cell through Wnt/B-catenin signaling pathway. Chin. Tradit. Herbal Drug. 2018, 49, 5155–5160. [Google Scholar] [CrossRef]
  77. Li, C.; Hong, L.; Liu, C.; Min, J.; Hu, M.; Guo, W. Astragalus Polysaccharides Increase the Sensitivity of SKOV3 Cells to Cisplatin. Arch. Gynecol. Obstet. 2018, 297, 381–386. [Google Scholar] [CrossRef]
  78. Zhou, Z.; Meng, M.; 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. 2017, 23, 462–469. [Google Scholar] [CrossRef] [PubMed]
  79. Guo, Y.; Zhang, Z.; Wang, Z.; Liu, G.; Liu, Y.; Wang, H. Astragalus Polysaccharides Inhibit Ovarian Cancer Cell Growth via microRNA-27a/FBXW7 Signaling Pathway. Biosci. Rep. 2020, 40, BSR20193396. [Google Scholar] [CrossRef]
  80. Chu, Y.; Fang, Y.; Chi, J.; Li, J.; Zhang, D.; Zou, Y.; 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. 2018, 51, e7665. [Google Scholar] [CrossRef]
  81. Tao, X.; Zhang, X.; 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. 2022, 45, 553–560. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, J.X.; Han, Y.P.; Bai, C.; 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. 2015, 8, 12539–12547. [Google Scholar] [PubMed]
  83. Li, C.J.; Shen, G.H. Mechanism of astragalus polysaccharides promote colon cancer stem cell apoptosis byinducing autophagy. Chin. J. Tradit. Chin. Med. Pharm. 2022, 37, 2274–2279. [Google Scholar]
  84. Li, M.; Duan, F.; Pan, Z.; Liu, X.; Lu, W.; Liang, C.; Fang, Z.; Peng, P.; Jia, D. Astragalus Polysaccharide Promotes Doxorubicin-Induced Apoptosis by Reducing O-GlcNAcylation in Hepatocellular Carcinoma. Cells 2023, 12, 866. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, X.; Liu, F.; Zhao, S.; Guo, B.; Ling, P.; Han, G.; Cui, Z. Purification of an Acidic Polysaccharide from Suaeda Salsa Plant and Its Anti-Tumor Activity by Activating Mitochondrial Pathway in MCF-7 Cells. Carbohydr. Polym. 2019, 215, 99–107. [Google Scholar] [CrossRef] [PubMed]
  86. Jose, G.M.; Raghavankutty, M.; Kurup, G.M. Sulfated Polysaccharides from Padina Tetrastromatica Induce Apoptosis in HeLa Cells through ROS Triggered Mitochondrial Pathway. Process Biochem. 2018, 68, 197–204. [Google Scholar] [CrossRef]
  87. Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The Multiple Mechanisms That Regulate P53 Activity and Cell Fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef]
  88. Zhang, Q.; Gao, L.; Huang, S.; Liang, Y.; Hu, J.; Zhang, Y.; Wei, S.; Hu, X. Cocktail of Astragalus membranaceus and Radix Trichosanthis Suppresses Melanoma Tumor Growth and Cell Migration Through Regulation of Akt-Related Signaling Pathway. Front. Pharmacol. 2022, 13, 880215. [Google Scholar] [CrossRef]
  89. Backman, L.J.; Danielson, P. Akt-mediated Anti-apoptotic Effects of Substance P in Anti-Fas-induced Apoptosis of Human Tenocytes. J. Cell. Mol. Med. 2013, 17, 723–733. [Google Scholar] [CrossRef]
  90. Li, S.; Sun, Y.; Huang, J.; Wang, B.; Gong, Y.; Fang, Y.; Liu, Y.; Wang, S.; Guo, Y.; Wang, H.; et al. Anti-Tumor Effects and Mechanisms of Astragalus membranaceus (AM) and Its Specific Immunopotentiation: Status and Prospect. J. Ethnopharmacol. 2020, 258, 112797. [Google Scholar] [CrossRef]
  91. García-Gómez, R.; Bustelo, X.R.; Crespo, P. Protein–Protein Interactions: Emerging Oncotargets in the RAS-ERK Pathway. Trends Cancer 2018, 4, 616–633. [Google Scholar] [CrossRef] [PubMed]
  92. Liao, X.; Bu, Y.; Jia, Q. Traditional Chinese Medicine as Supportive Care for the Management of Liver Cancer: Past, Present, and Future. Genes Dis. 2020, 7, 370–379. [Google Scholar] [CrossRef]
  93. Derynck, R.; Zhang, Y.E. Smad-Dependent and Smad-Independent Pathways in TGF-β Family Signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
  94. Iwayama, H.; Sakamoto, T.; Nawa, A.; Ueda, N. Crosstalk between Smad and Mitogen-Activated Protein Kinases for the Regulation of Apoptosis in Cyclosporine A-Induced Renal Tubular Injury. Nephron Extra 2011, 1, 178–189. [Google Scholar] [CrossRef] [PubMed]
  95. Yang, Q.; Meng, D.; Zhang, Q.; Wang, J. Advances in Research on the Anti-Tumor Mechanism of Astragalus Polysaccharides. Front. Oncol. 2024, 14, 1334915. [Google Scholar] [CrossRef] [PubMed]
  96. Akyol, Z.; Çoker-Gürkan, A.; Arisan, E.D.; Obakan-Yerlikaya, P.; Palavan-Ünsal, N. DENSpm Overcame Bcl-2 Mediated Resistance against Paclitaxel Treatment in MCF-7 Breast Cancer Cells via Activating Polyamine Catabolic Machinery. Biomed. Pharmacother. 2016, 84, 2029–2041. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, Y.S.; Dutta, A. MicroRNAs in Cancer. Annu. Rev. Pathol. Mech. Dis. 2009, 4, 199–227. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, Z.; Ji, G.; Wu, Q.; Feng, S.; Zhao, Y.; Cao, Z.; Tao, C. Integrated Microarray Meta-Analysis Identifies miRNA-27a as an Oncogene in Ovarian Cancer by Inhibiting FOXO1. Life Sci. 2018, 210, 263–270. [Google Scholar] [CrossRef] [PubMed]
  99. Ding, L.; Zhang, S.; Xu, M.; Zhang, R.; Sui, P.; Yang, Q. MicroRNA-27a Contributes to the Malignant Behavior of Gastric Cancer Cells by Directly Targeting PH Domain and Leucine-Rich Repeat Protein Phosphatase 2. J. Exp. Clin. Cancer Res. 2017, 36, 45. [Google Scholar] [CrossRef] [PubMed]
  100. Peng, H.; Wang, X.; Zhang, P.; Sun, T.; Ren, X.; Xia, Z. miR-27a Promotes Cell Proliferation and Metastasis in Renal Cell Carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 2259–2266. [Google Scholar]
  101. Cheng, Y.; Wang, Y.; Cheng, Y.; Yang, Q.; Zhang, L.; Li, Z.; Cheng, J. FOXD3-Induced miR-133a Blocks Progression and Metastasis of Colorectal Cancer through Regulating UBA2. J. Cancer 2021, 12, 6145–6154. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, X.; Zhu, L.; Lin, X.; Huang, Y.; Lin, Z. MiR-133a-3p Inhibits the Malignant Progression of Oesophageal Cancer by Targeting CDCA8. J. Biochem. 2022, 170, 689–698. [Google Scholar] [CrossRef] [PubMed]
  103. Sakr, M.; Takino, T.; Sabit, H.; Nakada, M.; Li, Z.; Sato, H. miR-150-5p and miR-133a Suppress Glioma Cell Proliferation and Migration through Targeting Membrane-Type-1 Matrix Metalloproteinase. Gene 2016, 587, 155–162. [Google Scholar] [CrossRef] [PubMed]
  104. Whibley, C.; Pharoah, P.D.P.; Hollstein, M. P53 Polymorphisms: Cancer Implications. Nat. Rev. Cancer 2009, 9, 95–107. [Google Scholar] [CrossRef]
  105. Gottlieb, T.M.; Oren, M. P53 and Apoptosis. Semin. Cancer Biol. 1998, 8, 359–368. [Google Scholar] [CrossRef] [PubMed]
  106. O’Brien, D.I.; Nally, K.; Kelly, R.G.; O’Connor, T.M.; Shanahan, F.; O’Connell, J. Targeting the Fas/Fas Ligand Pathway in Cancer. Expert Opin. Ther. Targets 2005, 9, 1031–1044. [Google Scholar] [CrossRef]
  107. Liang, X.H.; Jackson, S.; Seaman, M.; Brown, K.; Kempkes, B.; Hibshoosh, H.; Levine, B. Induction of Autophagy and Inhibition of Tumorigenesis by Beclin 1. Nature 1999, 402, 672–676. [Google Scholar] [CrossRef]
  108. Mathew, R.; Karantza-Wadsworth, V.; White, E. Role of Autophagy in Cancer. Nat. Rev. Cancer 2007, 7, 961–967. [Google Scholar] [CrossRef]
  109. Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The Role of Autophagy in Cancer: Therapeutic Implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef]
  110. Cordani, M.; Somoza, Á. Targeting Autophagy Using Metallic Nanoparticles: A Promising Strategy for Cancer Treatment. Cell. Mol. Life Sci. 2019, 76, 1215–1242. [Google Scholar] [CrossRef]
  111. Wang, X.; Li, Y.; Liu, D.; Wang, Y.; Ming, H. Astragalus polysaccharide inhibits autophagy and regulates expression of autophagy-related proteins in lung cancer A549 cells induced by xanthine oxidase. Chin. J. Cell. Mol. Immunol. 2019, 35, 619–624. [Google Scholar]
  112. Duan, Z.; Liang, M.; Yang, C.; Yan, C.; Wang, L.; Song, J.; Han, L.; Fan, Y.; Li, W.; Liang, T.; et al. Selenium Nanoparticles Coupling with Astragalus Polysaccharides Exert Their Cytotoxicities in MCF-7 Cells by Inhibiting Autophagy and Promoting Apoptosis. J. Trace Elem. Med. Biol. 2022, 73, 127006. [Google Scholar] [CrossRef] [PubMed]
  113. Zhai, Q.L.; Hu, X.D.; Xiao, J.; Yu, D.Q. Astragalus polysaccharide may increase sensitivity of cervical cancer HeLa cells to cisplatin by regulating cell autophagy. J. Chin. Mater. Medica 2018, 43, 805–812. [Google Scholar] [CrossRef] [PubMed]
  114. Röhrig, F.; Schulze, A. The Multifaceted Roles of Fatty Acid Synthesis in Cancer. Nat. Rev. Cancer 2016, 16, 732–749. [Google Scholar] [CrossRef] [PubMed]
  115. Li, D.; Li, Y. The Interaction between Ferroptosis and Lipid Metabolism in Cancer. Signal Transduct. Target. Ther. 2020, 5, 108. [Google Scholar] [CrossRef]
  116. Cheng, C.; Geng, F.; Cheng, X.; Guo, D. Lipid Metabolism Reprogramming and Its Potential Targets in Cancer. Cancer Commun. 2018, 38, 27. [Google Scholar] [CrossRef]
  117. Pope, E.D.; Kimbrough, E.O.; Vemireddy, L.P.; Surapaneni, P.K.; Copland, J.A.; Mody, K. Aberrant Lipid Metabolism as a Therapeutic Target in Liver Cancer. Expert Opin. Ther. Targets 2019, 23, 473–483. [Google Scholar] [CrossRef]
  118. Yi, M.; Li, J.; Chen, S.; Cai, J.; Ban, Y.; Peng, Q.; Zhou, Y.; Zeng, Z.; Peng, S.; Li, X.; et al. Emerging Role of Lipid Metabolism Alterations in Cancer Stem Cells. J. Exp. Clin. Cancer Res. 2018, 37, 118. [Google Scholar] [CrossRef]
  119. Cheng, L.; Zhu, Y.; Han, H.; Zhang, Q.; Cui, K.; Shen, H.; Zhang, J.; Yan, J.; Prochownik, E.; Li, Y. MicroRNA-148a Deficiency Promotes Hepatic Lipid Metabolism and Hepatocarcinogenesis in Mice. Cell Death Dis. 2017, 8, e2916. [Google Scholar] [CrossRef]
  120. Pham, D.V.; Park, P.H. Adiponectin Triggers Breast Cancer Cell Death via Fatty Acid Metabolic Reprogramming. J. Exp. Clin. Cancer Res. 2022, 41, 9. [Google Scholar] [CrossRef]
  121. Guo, S.; Ma, B.; Jiang, X.; Li, X.; Jia, Y. Astragalus Polysaccharides Inhibits Tumorigenesis and Lipid Metabolism Through miR-138-5p/SIRT1/SREBP1 Pathway in Prostate Cancer. Front. Pharmacol. 2020, 11, 598. [Google Scholar] [CrossRef]
  122. Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.-C.; Kroemer, G.; Zitvogel, L. Targeting the Tumor Microenvironment: Removing Obstruction to Anticancer Immune Responses and Immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef] [PubMed]
  123. Xiao, Y.; Yu, D. Tumor Microenvironment as a Therapeutic Target in Cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, Y.N.; Gui, M.B.; Qu, L.P.; Zou, M.; Gao, F. Astragalus polysaccharide inhibits IDO1 expression in colon tumor microenvironment to increase intratumoral CD8+ T cell infiltration. China J. Chin. Mater. Medica 2023, 48, 4722–4730. [Google Scholar] [CrossRef] [PubMed]
  125. Elham, S.; Mohammad-Reza, S.; Kobra, E.; Samaneh, K.; Mona, A.; Morteza, M.; Elahe, S. Immunomodulatory Effects of Astragalus Polysaccharide on Human Peripheral Blood Mononuclear Cells Co-Cultured with Cervical Cancer Cell Line. J. Tradit. Chin. Med. 2021, 41, 684–694. [Google Scholar] [CrossRef] [PubMed]
  126. Zhao, L.; Zhong, Y.; Liang, J.; Gao, H.; 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. 2019, 29, 392–394. [Google Scholar] [CrossRef]
  127. Lee, Y.C.; Huang, H.T.; Chang, C.D.; Chen, C.T.; Lin, T.Y.; Yang, T.W.; Chang, F.L.; Lu, M.K.; Chiou, C.T.; Chen, W.C.; et al. Isolation of Anti-VEGF Monoclonal Antibodies with Neutralizing Effects from an Astragalus-Induced Immune Antibody Library. Int. Immunopharmacol. 2020, 88, 107007. [Google Scholar] [CrossRef] [PubMed]
  128. Brock, S.E.; Rendon, B.E.; Yaddanapudi, K.; Mitchell, R.A. Negative Regulation of AMP-Activated Protein Kinase (AMPK) Activity by Macrophage Migration Inhibitory Factor (MIF) Family Members in Non-Small Cell Lung Carcinomas. J. Biol. Chem. 2012, 287, 37917–37925. [Google Scholar] [CrossRef] [PubMed]
  129. Liao, C.H.; Yong, C.Y.; Lai, G.M.; Chow, J.M.; Cheng, C.F.; Fang, C.L.; Lin, P.C.; Chang, C.L.; Zheng, Y.M.; Chuang, S.E.; et al. Astragalus Polysaccharide (PG2) Suppresses Macrophage Migration Inhibitory Factor and Aggressiveness of Lung Adenocarcinoma Cells. Am. J. Chin. Med. 2020, 48, 1491–1509. [Google Scholar] [CrossRef]
  130. Li, R.; Chen, W.; Wang, W.; Tian, W.; Zhang, X. Extraction, Characterization of Astragalus Polysaccharides and Its Immune Modulating Activities in Rats with Gastric Cancer. Carbohydr. Polym. 2009, 78, 738–742. [Google Scholar] [CrossRef]
  131. Li, Q.; Bao, J.; Li, X.; Zhang, T.; Shen, X. Inhibiting Effect of Astragalus Polysaccharides on the Functions of CD4+CD25 highTreg Cells in the Tumor Microenvironment of Human Hepatocellular Carcinoma. Chin. Med. J. 2012, 125, 786–793. [Google Scholar]
  132. Bian, Y.; Zhang, Y.; Zhou, Y.; Li, G.H.; Feng, X.S. Progress in the Pretreatment and Analysis of Flavonoids: An Update since 2013. Sep. Purif. Rev. 2022, 51, 11–37. [Google Scholar] [CrossRef]
  133. Yang, S.; Sun, S.; Xu, W.; Yu, B.; Wang, G.; 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. 2020, 21, 1819–1832. [Google Scholar] [CrossRef]
  134. Tian, Q.E.; De Li, H.; Yan, M.; Cai, H.L.; Tan, Q.Y.; Zhang, W.Y. Effects of Astragalus Polysaccharides on P-Glycoprotein Efflux Pump Function and Protein Expression in H22 Hepatoma Cells in Vitro. BMC Complement. Altern. Med. 2012, 12, 94. [Google Scholar] [CrossRef]
  135. Wang, Y.; Zhang, X.; Wang, Y.; Zhao, W.; Li, H.; Zhang, L.; Li, X.; Zhang, T.; Zhang, H.; Huang, H.; et al. Application of Immune Checkpoint Targets in the Anti-Tumor Novel Drugs and Traditional Chinese Medicine Development. Acta Pharm. Sin. B 2021, 11, 2957–2972. [Google Scholar] [CrossRef]
  136. Di Ciaula, A.; Stella, A.; Bonfrate, L.; Wang, D.Q.H.; Portincasa, P. Gut Microbiota between Environment and Genetic Background in Familial Mediterranean Fever (FMF). Genes 2020, 11, 1041. [Google Scholar] [CrossRef] [PubMed]
  137. Chang, H.L.; Kuo, Y.H.; Wu, L.H.; Chang, C.M.; Cheng, K.J.; Tyan, Y.C.; Lee, C.H. The Extracts of Astragalus membranaceus Overcome Tumor Immune Tolerance by Inhibition of Tumor Programmed Cell Death Protein Ligand-1 Expression. Int. J. Med. Sci. 2020, 17, 939–945. [Google Scholar] [CrossRef]
  138. Chang, F.L.; Tsai, K.C.; Lin, T.Y.; Yang, T.W.; Lo, Y.N.; Chen, W.C.; Chang, J.H.; Lu, M.K.; Chiou, C.T.; Chen, P.-H.; et al. Astragalus membranaceus-Derived Anti-Programmed Death-1 Monoclonal Antibodies with Immunomodulatory Therapeutic Effects against Tumors. BioMed Res. Int. 2020, 2020, 3415471. [Google Scholar] [CrossRef]
  139. Tsao, S.M.; Wu, T.C.; Chen, J.; Chang, F.; Tsao, T. Astragalus Polysaccharide Injection (PG2) Normalizes the Neutrophil-to-Lymphocyte Ratio in Patients with Advanced Lung Cancer Receiving Immunotherapy. Integr. Cancer Ther. 2021, 20, 153473542199525. [Google Scholar] [CrossRef]
  140. Zhang, Y.; Zhang, Z. The History and Advances in Cancer Immunotherapy: Understanding the Characteristics of Tumor-Infiltrating Immune Cells and Their Therapeutic Implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef]
  141. Rosenberg, S.A.; Restifo, N.P. Adoptive Cell Transfer as Personalized Immunotherapy for Human Cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef]
  142. Zhang, S.; Mou, X.Y.; Wang, H.M.; Jiang, S.J. Astragalus polysaccharide-induced enhancement of CIK cell killing in dendritic cells. Chin. J. Immunol. 2009, 140–142. [Google Scholar]
  143. Wang, J.T.; Han, Q.Q.; Wang, B.Y.; Yan, X.J.; Li, W.; Zhang, M.H.; Zhou, H.R.; Zhu, Y.Q. Effects of astragalus polysaccharide-induced DC combined with CIK on human oesophageal cancer Eca-109 cells. J. Tra. Chin. Med. 2016, 31, 478–481. [Google Scholar] [CrossRef]
  144. Zhang, Y.; Jia, Y.J.; Li, X.J.; Wang, L.; Du, M.N.; Zhang, X.X. Clinical observation of injectable astragalus polysaccharide combined with CIK cells in the treatment of middle and advanced qi deficiency type non-small cell lung cancer. Chin. Herb. Med. 2018, 1647–1651. [Google Scholar]
  145. Xu, Y.X.; Liu, X.J.; Qu, F. Combined treatment of ovarian cancer with herbal polysaccharides and overt immunisation. Chin. J. Exp. For. 2011, 17, 231–234. [Google Scholar] [CrossRef]
  146. Huo, H.Q.; Shan, B.E. Study on the Killing Effect of CIK Combined with Astragalus Polysaccharide on Hela Cells In Vitro and Ex Vivo; Hebei Medical University: Shijiazhuang, China, 2016. [Google Scholar]
  147. Chen, H.L.; Zhang, L.S. Astragalus Polysaccharide-Induced Killing Effect of DC Combined with CIK on Leukaemic K562 Cells; Lanzhou University: Lanzhou, China, 2013. [Google Scholar]
  148. Gong, Y.Z.; Gu, X.L. Effect of Four Kinds of Polysaccharides on Cytokine-Induced Anti-Tumor Activity of Killer Cells and Dendritic Cells; Shandong University: Jinan, China, 2016. [Google Scholar]
  149. Dong, S.; Wang, P.; Zhang, L.; Zhang, X.; Li, X.; Wang, J.; Cui, X.; Lan, T.; Gao, C.; Shi, Y.; et al. The Qi Yin San Liang San Decoction Enhances Anti-CD19 CAR-T Cell Function in the Treatment of B-Cell Lymphomas. J. Ethnopharmacol. 2024, 319, 117109. [Google Scholar] [CrossRef] [PubMed]
  150. To Investigate the Effect and Mechanism of Astragalus Polysaccharide (APS) on CAR T Therapy for HCC Based on T Cell Subsets Differentiation—National Natural Science Foundation Abstract Inquiry—MedSci.cn. Available online: https://www.medsci.cn/sci/nsfc_show.do?id=5275104433e32 (accessed on 20 April 2024).
  151. Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer Vaccines as Promising Immuno-Therapeutics: Platforms and Current Progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef] [PubMed]
  152. Qiu, B.; Jing, X.N.; Wu, J.B.; Chen, D.D.; Liu, X.Q. Antitumor effect of dendritic cell vaccine induced by Astragalus polysaccharide on S180 tumor-bearing mice. J. Nanjing Univ. Chin. Med. 2015, 31, 44–47. [Google Scholar]
  153. Qiu, X.N.; Qiu, B.; Zhan, W.X.; Wu, J.B. Effect of dendritic cell vaccine induced by Astragalus polysaccharide on Th1/Th2 cytokines in S180 tumor-bearing mice. Tianjin Pharm. 2014, 42, 1080–1083. [Google Scholar]
  154. Yang, L.; Guo, C.; Zhang, X.P.; Ming, H.X.; Chen, Y.W. Effect of astragalus saponin-alumina hydroxide adjuvant complex on anti-breast cancer activity of bFGF tumor vaccine. J. Hexi Coll. 2018, 34, 26–31. [Google Scholar]
  155. Zhang, X.; Qiu, H.; Li, C.; Cai, P.; Qi, F. The Positive Role of Traditional Chinese Medicine as an Adjunctive Therapy for Cancer. Biosci. Trends 2021, 15, 283–298. [Google Scholar] [CrossRef] [PubMed]
  156. Deng, B.; Jia, L.; Cheng, Z. Radix Astragali-Based Chinese Herbal Medicine for Oxaliplatin-Induced Peripheral Neuropathy: A Systematic Review and Meta-Analysis. Evid.-Based Complement. Altern. Med. 2016, 2016, 2421876. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, Z.; Qi, F.; Cui, Y.; Zhao, L.; Sun, X.; Tang, W.; Cai, P. An Update on Chinese Herbal Medicines as Adjuvant Treatment of Anticancer Therapeutics. Biosci. Trends 2018, 12, 220–239. [Google Scholar] [CrossRef] [PubMed]
  158. Sellami, M.; Slimeni, O.; Pokrywka, A.; Kuvačić, G.; D Hayes, L.; Milic, M.; Padulo, J. Herbal Medicine for Sports: A Review. J. Int. Soc. Sports Nutr. 2018, 15, 14. [Google Scholar] [CrossRef] [PubMed]
  159. Samuel, A.O.; Huang, B.T.; Chen, Y.; Guo, F.X.; Yang, D.D.; Jin, J.Q. Antioxidant and Antibacterial Insights into the Leaves, Leaf Tea and Medicinal Roots from Astragalus membranaceus (Fisch.) Bge. Sci. Rep. 2021, 11, 19625. [Google Scholar] [CrossRef] [PubMed]
  160. Wu, X.; Li, X.; Wang, W.; Shan, Y.; Wang, C.; Zhu, M.; La, Q.; Zhong, Y.; Xu, Y.; Nan, P.; et al. Integrated Metabolomics and Transcriptomics Study of Traditional Herb Astragalus membranaceus Bge. Var. Mongolicus (Bge.) Hsiao Reveals Global Metabolic Profile and Novel Phytochemical Ingredients. BMC Genom. 2020, 21, 697. [Google Scholar] [CrossRef] [PubMed]
  161. Chang Gung Memorial Hospital. Efficacy Study of Chinese Medicine on Modulating Immune Alterations in Advanced Stage, Non-Small Cell Lung Cancer Patients Receiving 1st Line Doublet Chemotherapy and 2nd Line Target Therapy; ClinicalTrials.gov: Bethesda, MD, USA, 2013.
  162. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2300068199 (accessed on 19 February 2024).
  163. Yan, L. Clinical study of Astragalus polysaccharide combined with Iodine-125 in treatment of advanced lung cancer. Chin. J. Endem. Dis. Control 2014, 210. [Google Scholar]
  164. Qin, Z.Q.; Lu, L.Q.; Yuan, G.R.; Wu, G.Q.; Xue, Y. Clinical observation of Astragalus polysaccharide for injection combined with GP regimen in the treatment of advanced non-small cell lung cancer. J. Chin. Med. 2009, 73, 664–666. [Google Scholar] [CrossRef]
  165. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ITMCTR2100004716 (accessed on 24 February 2024).
  166. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2000040911 (accessed on 24 February 2024).
  167. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ITMCTR2000003215 (accessed on 24 February 2024).
  168. PhytoHealth Corporation. PG2 Treatment for Reduction of Chemotherapy-Induced Toxicity and Encouraging Compliance with Chemotherapy Among Stage II/III Breast Cancer Patients Receiving Adjuvant Chemotherapy; ClinicalTrials.gov: Bethesda, MD, USA, 2023.
  169. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2300076131 (accessed on 24 February 2024).
  170. Zhang, D.; Zheng, J.; Ni, M.; Wu, J.; Wang, K.; Duan, X.; Zhang, X.; Zhang, B. Comparative Efficacy and Safety of Chinese Herbal Injections Combined with the FOLFOX Regimen for Treating Gastric Cancer in China: A Network Meta-Analysis. Oncotarget 2017, 8, 68873–68889. [Google Scholar] [CrossRef]
  171. China Clinical Trial Registry. A Randomized Controlled Clinical Study of Astragalus membranaceus to Enhance Susceptibility to Immune Checkpoint Inhibitors in Advanced Gastric Cancer by Regulating Intestinal Flora. Available online: https://www.chictr.org.cn/showproj.html?proj=189330 (accessed on 19 February 2024).
  172. Zhang, X. Comparison of Neoadjuvant Treatment for Pancreatic Cancer: Astragalus Combined with Gemcitabine Versus Gemcitabine Alone—A Single-Center, Randomized, Double-Blind Study; ClinicalTrials.gov: Bethesda, MD, USA, 2024.
  173. Kim, E.H.; Yoon, J.H.; Yoon, S.S.; Lee, J.Y.; Yoon, S.W. Efficacy of Chemotherapy Integrated with Traditional Korean Medicine in Patients with Metastatic Pancreatic Cancer: A Single-Center Retrospective Study. Integr. Cancer Ther. 2020, 19, 1534735420983457. [Google Scholar] [CrossRef]
  174. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2000037982 (accessed on 18 February 2024).
  175. Lin, S.; An, X.; Guo, Y.; Gu, J.; Xie, T.; Wu, Q.; Sui, X. Meta-Analysis of Astragalus-Containing Traditional Chinese Medicine Combined With Chemotherapy for Colorectal Cancer: Efficacy and Safety to Tumor Response. Front. Oncol. 2019, 9, 749. [Google Scholar] [CrossRef] [PubMed]
  176. Chen, Y.; Cheng, C.S.; Tan, H.Y.; Tam, C.W.; Wang, N.; Feng, Y. Efficacy of Herbal Medicines Intervention for Colorectal Cancer Patients With Chemotherapy-Induced Gastrointestinal Toxicity—A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 629132. [Google Scholar] [CrossRef]
  177. Chen, M.; May, B.H.; Zhou, I.W.; Xue, C.C.L.; Zhang, A.L. FOLFOX 4 Combined with Herbal Medicine for Advanced Colorectal Cancer: A Systematic Review. Phytother. Res. 2014, 28, 976–991. [Google Scholar] [CrossRef] [PubMed]
  178. Han, J.; Lai, H.; Li, W.; Liao, H.; Xiao, C.; Li, X.; You, F.; Guo, J. Efficacy and Safety of Traditional Plant-Based Medicines for Preventing Chronic Oxaliplatin-Induced Peripheral Neurotoxicity in Patients with Colorectal Cancer: A Systematic Review and Meta-Analysis with Core Herb Contribution. J. Ethnopharmacol. 2024, 326, 117735. [Google Scholar] [CrossRef] [PubMed]
  179. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=NCT01720563 (accessed on 24 February 2024).
  180. PhytoHealth Corporation. PG2 Concurrent with Chemoradiation for Locally Advanced Esophageal Cancer; ClinicalTrials.gov: Bethesda, MD, USA, 2023.
  181. PhytoHealth Corporation. PG2 Treatment for Improving Fatigue Among Advanced Cancer Patients under Standard Palliative Care; ClinicalTrials.gov: Bethesda, MD, USA, 2023.
  182. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.M.; Carneiro, F.; Cree, I.A.; WHO Classification of Tumours Editorial Board. The 2019 WHO Classification of Tumours of the Digestive System. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [PubMed]
  183. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric Cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef] [PubMed]
  184. Xu, W.; Li, B.; Xu, M.; Yang, T.; Hao, X. Traditional Chinese Medicine for Precancerous Lesions of Gastric Cancer: A Review. Biomed. Pharmacother. 2022, 146, 112542. [Google Scholar] [CrossRef] [PubMed]
  185. Liang, Z.; Xu, Y.; Zhang, Y.; Zhang, X.; Song, J.; Jin, J.; Qian, H. Anticancer Applications of Phytochemicals in Gastric Cancer: Effects and Molecular Mechanism. Front. Pharmacol. 2023, 13, 1078090. [Google Scholar] [CrossRef]
  186. Wang, X.; Chen, J.; Yang, F.; Ali, F.; Mao, Y.; Hu, A.; Xu, T.; Yang, Y.; Wang, F.; Zhou, G.; et al. Two Kinds of Traditional Chinese Medicine Prescriptions Reduce Thymic Inflammation Levels and Improve Humoral Immunity of Finishing Pigs. Front. Vet. Sci. 2022, 9, 929112. [Google Scholar] [CrossRef]
  187. Ionkova, I.; Shkondrov, A.; Zarev, Y.; Kozuharova, E.; Krasteva, I. Anticancer Secondary Metabolites: From Ethnopharmacology and Identification in Native Complexes to Biotechnological Studies in Species of Genus Astragalus L. and Gloriosa L. Curr. Issues Mol. Biol. 2022, 44, 3884–3904. [Google Scholar] [CrossRef]
  188. Sakurai, M.; Egashira, N.; Kawashiri, T.; Yano, T.; Ikesue, H.; Oishi, R. Oxaliplatin-Induced Neuropathy in the Rat: Involvement of Oxalate in Cold Hyperalgesia but Not Mechanical Allodynia. Pain 2009, 147, 165–174. [Google Scholar] [CrossRef] [PubMed]
  189. Henricks, L.M.; Lunenburg, C.A.T.C.; De Man, F.M.; Meulendijks, D.; Frederix, G.W.J.; Kienhuis, E.; Creemers, G.J.; Baars, A.; Dezentjé, V.O.; Imholz, A.L.T.; et al. DPYD Genotype-Guided Dose Individualisation of Fluoropyrimidine Therapy in Patients with Cancer: A Prospective Safety Analysis. Lancet Oncol. 2018, 19, 1459–1467. [Google Scholar] [CrossRef]
  190. China Clinical Trial Registry. Clinical Study on the Treatment of Low Grade Gastric Intraepithelial Neoplasia by Regulating the Gastric Environment with Eqi Shicao Tang Based on Metabolomics Exploration. Available online: https://www.chictr.org.cn/showproj.html?proj=207037 (accessed on 19 February 2024).
  191. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal Cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef] [PubMed]
  192. Qi, F.; Li, A.; Inagaki, Y.; Gao, J.; Li, J.; Kokudo, N.; Li, X.; Tang, W. Chinese Herbal Medicines as Adjuvant Treatment during Chemo- or Radio-Therapy for Cancer. Biosci. Trends 2010, 4, 297–307. [Google Scholar] [PubMed]
  193. Yang, Y.; Ting, W.; Xiao, L.; Shufei, F.; Wangxiao, T.; Xiaoying, W.; Xiumei, G.; Boli, Z. Immunoregulation of Shenqi Fuzheng Injection Combined with Chemotherapy in Cancer Patients: A Systematic Review and Meta-Analysis. Evid. Based Complement. Altern. Med. 2017, 2017, 5121538. [Google Scholar] [CrossRef] [PubMed]
  194. Xu, R.; Lin, L.; Li, Y.; Li, Y. ShenQi FuZheng Injection Combined with Chemotherapy in the Treatment of Colorectal Cancer: A Meta-Analysis. PLoS ONE 2017, 12, e0185254. [Google Scholar] [CrossRef]
  195. Jiang, H.; Zhang, H.; Hu, X.; Ma, J. A Meta-Analysis of Shenqi Fuzheng Combined with Radiation in the Treatment of Nonsmall Cell Lung Cancer. J. Cancer Res. Ther. 2015, 11, C101–C103. [Google Scholar] [CrossRef] [PubMed]
  196. Cheng, X.; Huo, J.; Wang, D.; Cai, X.; Sun, X.; Lu, W.; Yang, Y.; Hu, C.; Wang, X.; Cao, P. Herbal Medicine AC591 Prevents Oxaliplatin-Induced Peripheral Neuropathy in Animal Model and Cancer Patients. Front. Pharmacol. 2017, 8, 344. [Google Scholar] [CrossRef] [PubMed]
  197. Li, X.; Ramadori, P.; Pfister, D.; Seehawer, M.; Zender, L.; Heikenwalder, M. The Immunological and Metabolic Landscape in Primary and Metastatic Liver Cancer. Nat. Rev. Cancer 2021, 21, 541–557. [Google Scholar] [CrossRef]
  198. Zhang, D.; Wang, K.; Zheng, J.; Wu, J.; Duan, X.; Ni, M.; Liu, S.; Zhang, B.; Zhao, Y. Comparative Efficacy and Safety of Chinese Herbal Injections Combined with Transcatheter Hepatic Arterial Chemoembolization in Treatment of Liver Cancer: A Bayesian Network Meta-Analysis. J. Tradit. Chin. Med. 2020, 40, 167–187. [Google Scholar]
  199. Li, Y.; Wang, W.P. Chinese Medicine Treatment of Pancreatic Cancer Analysis and Mechanism Analysis; Liaoning University of Traditional Chinese Medicine: Shenyang, China, 2023. [Google Scholar]
  200. Wang, J.Y.; Ma, G.W.; Dai, S.Q.; Rong, T.H.; Wang, X.; Lin, P.; Ye, W.F.; Zhang, L.J.; Li, X.D.; Zhang, X.; et al. Effect of Cellular Immune Supportive Treatment on Immunity of Esophageal Carcinoma Patients after Modern Two-Field Lymph Node Dissection. Chin. J. Cancer. 2007, 26, 778–781. [Google Scholar]
  201. Gao, X.P.; Zhao, W.X.; Zhang, Z.L. Effect of Huangqi Zengmian Powder on Interstitial Response in Patients with Esophageal Cancer at Peri-Operational Period. Chin. J. Integr. Tradit. West. Med. 2001, 21, 171–173. [Google Scholar]
  202. Brody, H. Lung Cancer. Nature 2020, 587, S7. [Google Scholar] [CrossRef]
  203. He, H.; Zhou, X.; Wang, Q.; Zhao, Y. Does the Couse of Astragalus-Containing Chinese Herbal Prescriptions and Radiotherapy Benefit to Non-Small-Cell Lung Cancer Treatment: A Meta-Analysis of Randomized Trials. Evid.-Based Complement. Altern. Med. 2013, 2013, 426207. [Google Scholar] [CrossRef]
  204. Zhou, Y.; Hong, T.; Tong, L.; Liu, W.; Yang, X.; Luo, J.; Wang, F.; Li, J.; Yan, L. Astragalus Polysaccharide Combined with 10-Hydroxycamptothecin Inhibits Metastasis in Non-Small Cell Lung Carcinoma Cell Lines via the MAP4K3/mTOR Signaling Pathway. Int. J. Mol. Med. 2018, 42, 3093–3104. [Google Scholar] [CrossRef]
  205. Gan, Q.; Wang, J.; Hu, J.; Lou, G.; Xiong, H.; Peng, C.; Huang, Q. Modulation of Apoptosis by Plant Polysaccharides for Exerting Anti-Cancer Effects: A Review. Front. Pharmacol. 2020, 11, 792. [Google Scholar] [CrossRef]
  206. Cheon, C.; Yoo, J.E.; Yoo, H.S.; Cho, C.K.; Kang, S.; Kim, M.; Jang, B.H.; Shin, Y.C.; Ko, S.G. Efficacy and Safety of Sipjeondaebo-Tang for Anorexia in Patients with Cancer: A Pilot, Randomized, Double-Blind, Placebo-Controlled Trial. Evid.-Based Complement. Altern. Med. 2017, 2017, 8780325. [Google Scholar] [CrossRef]
  207. Ishiura, Y.; Shiba, Y.; Terasaki, Y.; Hayase, H.; Hamada, M.; Izawa, K.; Sugimoto, A.; Hirokami, K.; Segawa, M.; Kasahara, K.; et al. Effect of Japanese Traditional Medicine, TJ-48, on the Quality of Life of Patients with Non-Small Cell Lung Cancer Receiving Outpatient Chemotherapy. Gan Kagaku Ryoho 2016, 43, 331–334. [Google Scholar]
  208. Ikemoto, T.; Shimada, M.; Iwahashi, S.; Saito, Y.; Kanamoto, M.; Mori, H.; Morine, Y.; Imura, S.; Utsunomiya, T. Changes of Immunological Parameters with Administration of Japanese Kampo Medicine (Juzen-Taihoto/TJ-48) in Patients with Advanced Pancreatic Cancer. Int. J. Clin. Oncol. 2014, 19, 81–86. [Google Scholar] [CrossRef] [PubMed]
  209. Xiao, Z.; Liang, R.; Wang, C.Q.; Xu, S.; Li, N.; He, Y.; Tang, F.; Chen, L.; Ma, H. Can Aidi Injection Alleviate the Toxicity and Improve the Clinical Efficacy of Radiotherapy in Lung Cancer?: A Meta-Analysis of 16 Randomized Controlled Trials Following the PRISMA Guidelines. Medicine 2016, 95, e4517. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, J.; Li, G.; Yu, L.; Mo, T.; Wu, Q.; Zhou, Z. Aidi Injection plus Platinum-Based Chemotherapy for Stage IIIB/IV Non-Small Cell Lung Cancer: A Meta-Analysis of 42 RCTs Following the PRISMA Guidelines. J. Ethnopharmacol. 2018, 221, 137–150. [Google Scholar] [CrossRef] [PubMed]
  211. Chen, Y.P.; Chan, A.T.C.; Le, Q.T.; Blanchard, P.; Sun, Y.; Ma, J. Nasopharyngeal Carcinoma. Lancet 2019, 394, 64–80. [Google Scholar] [CrossRef] [PubMed]
  212. Miao, J.; Wang, L.; Tan, S.H.; Li, J.G.; Yi, J.; Ong, E.H.W.; Tan, L.L.Y.; Zhang, Y.; Gong, X.; Chen, Q.; et al. Adjuvant Capecitabine Following Concurrent Chemoradiotherapy in Locoregionally Advanced Nasopharyngeal Carcinoma: A Randomized Clinical Trial. JAMA Oncol. 2022, 8, 1776–1785. [Google Scholar] [CrossRef] [PubMed]
  213. Lin, C.; Cao, S.M.; Chang, E.T.; Liu, Z.; Cai, Y.; Zhang, Z.; Chen, G.; Huang, Q.H.; Xie, S.H.; Zhang, Y.; et al. Chinese Nonmedicinal Herbal Diet and Risk of Nasopharyngeal Carcinoma: A Population-Based Case-Control Study. Cancer 2019, 125, 4462–4470. [Google Scholar] [CrossRef] [PubMed]
  214. Harbeck, N.; Gnant, M. Breast Cancer. Lancet 2017, 389, 1134–1150. [Google Scholar] [CrossRef]
  215. Li, W.; Hu, X.; Li, Y.; Song, K. Cytotoxicity and Growth-Inhibiting Activity of Astragalus Polysaccharides against Breast Cancer via the Regulation of EGFR and ANXA1. J. Nat. Med. 2021, 75, 854–870. [Google Scholar] [CrossRef]
  216. Wang, B.; Guo, C.; Liu, Y.; Han, G.; Li, Y.; Zhang, Y.; Xu, H.; Chen, D. Novel Nano-Pomegranates Based on Astragalus Polysaccharides for Targeting ERα-Positive Breast Cancer and Multidrug Resistance. Drug Deliv. 2020, 27, 607–621. [Google Scholar] [CrossRef]
  217. Ghi, M.G.; Paccagnella, A.; Ferrari, D.; Foa, P.; Alterio, D.; Codecà, C.; Nolè, F.; Verri, E.; Orecchia, R.; Morelli, F.; et al. Induction TPF Followed by Concomitant Treatment versus Concomitant Treatment Alone in Locally Advanced Head and Neck Cancer. A Phase II-III Trial. Ann. Oncol. 2017, 28, 2206–2212. [Google Scholar] [CrossRef]
  218. Hsieh, C.H.; Lin, C.Y.; Hsu, C.L.; Fan, K.H.; Huang, S.F.; Liao, C.T.; Lee, L.Y.; Ng, S.K.; Yen, T.C.; Chang, J.T.C.; et al. Incorporation of Astragalus Polysaccharides Injection during Concurrent Chemoradiotherapy in Advanced Pharyngeal or Laryngeal Squamous Cell Carcinoma: Preliminary Experience of a Phase II Double-Blind, Randomized Trial. J. Cancer Res. Clin. Oncol. 2020, 146, 33–41. [Google Scholar] [CrossRef]
  219. Yan, P.H.; Yan, M.; Wang, X.M.; Wang, S.H. Effect of Huangqi Injection on Short-Term Prognosis in Children with Acute Lymphoblastic Leukemia. Chin. J. Contemp. Pediatr. 2014, 16, 141–146. [Google Scholar]
  220. Zhang, X.J.; Yan, M.; Liu, Y.; Wang, X.M.; Nuriding, H. Effects of Huangqi Injection on Infection Factors in Children with Acute Lymphoblastic Leukemia. Chin. J. Contemp. Pediatr. 2014, 16, 147–151. [Google Scholar]
  221. Li, L.J.; Li, M.Y.; Li, Y.T.; Feng, J.J.; Hao, F.Q.; Zhang, L. Adjuvant Activity of Sargassum Pallidum Polysaccharides against Combined Newcastle Disease, Infectious Bronchitis and Avian Influenza Inactivated Vaccines. Mar. Drugs 2012, 10, 2648–2660. [Google Scholar] [CrossRef] [PubMed]
  222. Cho, C.W.; Han, C.; Rhee, Y.K.; Lee, Y.C.; Shin, K.S.; Shin, J.S.; Lee, K.T.; Hong, H.D. Cheonggukjang Polysaccharides Enhance Immune Activities and Prevent Cyclophosphamide-Induced Immunosuppression. Int. J. Biol. Macromol. 2015, 72, 519–525. [Google Scholar] [CrossRef]
  223. Li, N.; Li, L.; Fang, J.C.; Wong, J.H.; Ng, T.B.; Jiang, Y.; Wang, C.R.; Zhang, N.Y.; Wen, T.Y.; Qu, L.Y.; et al. Isolation and Identification of a Novel Polysaccharide-Peptide Complex with Antioxidant, Anti-Proliferative and Hypoglycaemic Activities from the Abalone Mushroom. Biosci. Rep. 2012, 32, 221–228. [Google Scholar] [CrossRef] [PubMed]
  224. Xie, J.H.; Xie, M.Y.; Nie, S.P.; Shen, M.Y.; Wang, Y.X.; Li, C. Isolation, Chemical Composition and Antioxidant Activities of a Water-Soluble Polysaccharide from Cyclocarya paliurus (Batal.) Iljinskaja. Food Chem. 2010, 119, 1626–1632. [Google Scholar] [CrossRef]
  225. Cai, D.; He, F.; Wu, S.; Wang, Z.; Bian, Y.; Wen, C.; Ding, K. Functional Structural Domain Synthesis of Anti-Pancreatic Carcinoma Pectin-like Polysaccharide RN1. Carbohydr. Polym. 2024, 327, 121668. [Google Scholar] [CrossRef] [PubMed]
  226. Di, T.; Chen, G.; Sun, Y.; Ou, S.; Zeng, X.; Ye, H. Antioxidant and Immunostimulating Activities in Vitro of Sulfated Polysaccharides Isolated from Gracilaria Rubra. J. Funct. Foods 2017, 28, 64–75. [Google Scholar] [CrossRef]
  227. Liu, Y.; Zhao, J.; Zhao, Y.; Zong, S.; Tian, Y.; Chen, S.; Li, M.; Liu, H.; Zhang, Q.; Jing, X.; et al. Therapeutic Effects of Lentinan on Inflammatory Bowel Disease and Colitis-Associated Cancer. J. Cell. Mol. Med. 2019, 23, 750–760. [Google Scholar] [CrossRef]
  228. Wang, H.; Liu, Y.; Qi, Z.; Wang, S.; Liu, S.; Li, X.; Wang, H.; Xia, X. An Overview on Natural Polysaccharides with Antioxidant Properties. Curr. Med. Chem. 2013, 20, 2899–2913. [Google Scholar] [CrossRef]
  229. Chen, N.; Johnson, M.M.; Collier, M.A.; Gallovic, M.D.; Bachelder, E.M.; Ainslie, K.M. Tunable Degradation of Acetalated Dextran Microparticles Enables Controlled Vaccine Adjuvant and Antigen Delivery to Modulate Adaptive Immune Responses. J. Control. Release 2018, 273, 147–159. [Google Scholar] [CrossRef]
  230. Seweryn, E.; Ziała, A.; Gamian, A. Health-Promoting of Polysaccharides Extracted from Ganoderma lucidum. Nutrients 2021, 13, 2725. [Google Scholar] [CrossRef] [PubMed]
  231. Łysakowska, P.; Sobota, A.; Wirkijowska, A. Medicinal Mushrooms: Their Bioactive Components, Nutritional Value and Application in Functional Food Production—A Review. Molecules 2023, 28, 5393. [Google Scholar] [CrossRef] [PubMed]
  232. Zhao, Y.; Wang, J.; Wu, Z.; Yang, J.; Li, W.; Shen, L. Extraction, Purification and Anti-Proliferative Activities of Polysaccharides from Lentinus Edodes. Int. J. Biol. Macromol. 2016, 93, 136–144. [Google Scholar] [CrossRef] [PubMed]
  233. Lee, C.L.; Yang, X.; Wan, J.M.F. The Culture Duration Affects the Immunomodulatory and Anticancer Effect of Polysaccharopeptide Derived from Coriolus Versicolor. Enzym. Microb. Technol. 2006, 38, 14–21. [Google Scholar] [CrossRef]
  234. Dou, H.; Chang, Y.; Zhang, L. Chapter Fifteen—Coriolus Versicolor Polysaccharopeptide as an Immunotherapeutic in China. In Progress in Molecular Biology and Translational Science; Zhang, L., Ed.; Academic Press: Cambridge, MA, USA, 2019; Volume 163, pp. 361–381. ISBN 1877-1173. [Google Scholar]
  235. Wang, Y.Y.; Khoo, K.H.; Chen, S.T.; Lin, C.C.; Wong, C.H.; Lin, C.H. Studies on the Immuno-Modulating and Antitumor Activities of Ganoderma lucidum (Reishi) Polysaccharides: Functional and Proteomic Analyses of a Fucose-Containing Glycoprotein Fraction Responsible for the Activities. Bioorg. Med. Chem. 2002, 10, 1057–1062. [Google Scholar] [CrossRef] [PubMed]
  236. Zhao, L.; Dong, Y.; Chen, G.; Hu, Q. Extraction, Purification, Characterization and Antitumor Activity of Polysaccharides from Ganoderma lucidum. Carbohydr. Polym. 2010, 80, 783–789. [Google Scholar] [CrossRef]
  237. Zhou, Z.Q.; Xiao, J.; Fan, H.X.; Yu, Y.; He, R.R.; Feng, X.L.; Kurihara, H.; So, K.F.; Yao, X.S.; Gao, H. Polyphenols from Wolfberry and Their Bioactivities. Food Chem. 2017, 214, 644–654. [Google Scholar] [CrossRef] [PubMed]
  238. Xie, J.H.; Tang, W.; Jin, M.L.; Li, J.E.; Xie, M.Y. Recent Advances in Bioactive Polysaccharides from Lycium barbarum L., Zizyphus jujuba Mill, Plantago spp., and Morus spp.: Structures and Functionalities. Food Hydrocoll. 2016, 60, 148–160. [Google Scholar] [CrossRef]
  239. Ke, M.; Zhang, X.J.; Han, Z.H.; Yu, H.Y.; Lin, Y.; Zhang, W.G.; Sun, F.H.; Wang, T.J. Extraction, Purification of Lycium barbarum Polysaccharides and Bioactivity of Purified Fraction. Carbohydr. Polym. 2011, 86, 136–141. [Google Scholar] [CrossRef]
  240. Zhu, Z.; Liu, R.; Si, C.; Zhou, F.; Wang, Y.; Ding, L.; Jing, C.; Liu, A.; Zhang, Y. Structural Analysis and Anti-Tumor Activity Comparison of Polysaccharides from Astragalus. Carbohydr. Polym. 2011, 85, 895–902. [Google Scholar] [CrossRef]
  241. Morris, V.J.; Belshaw, N.J.; Waldron, K.W.; Maxwell, E.G. The Bioactivity of Modified Pectin Fragments. Bioact. Carbohydr. Diet. Fibre 2013, 1, 21–37. [Google Scholar] [CrossRef]
  242. Brouns, F.; Theuwissen, E.; Adam, A.; Bell, M.; Berger, A.; Mensink, R.P. Cholesterol-Lowering Properties of Different Pectin Types in Mildly Hyper-Cholesterolemic Men and Women. Eur. J. Clin. Nutr. 2012, 66, 591–599. [Google Scholar] [CrossRef] [PubMed]
  243. Zhang, Q.; Liu, J.; Duan, H.; Li, R.; Peng, W.; Wu, C. Activation of Nrf2/HO-1 Signaling: An Important Molecular Mechanism of Herbal Medicine in the Treatment of Atherosclerosis via the Protection of Vascular Endothelial Cells from Oxidative Stress. J. Adv. Res. 2021, 34, 43–63. [Google Scholar] [CrossRef] [PubMed]
  244. Wang, T.; Tao, Y.; Lai, C.; Huang, C.; Ling, Z.; Yong, Q. Influence of Glycosyl Composition on the Immunological Activity of Pectin and Pectin-Derived Oligosaccharide. Int. J. Biol. Macromol. 2022, 222, 671–679. [Google Scholar] [CrossRef] [PubMed]
  245. Emran, T.B.; Islam, F.; Mitra, S.; Paul, S.; Nath, N.; Khan, Z.; Das, R.; Chandran, D.; Sharma, R.; Lima, C.M.G.; et al. Pectin: A Bioactive Food Polysaccharide with Cancer Preventive Potential. Molecules 2022, 27, 7405. [Google Scholar] [CrossRef] [PubMed]
  246. Mukhtar, E.; Adhami, V.M.; Khan, N.; Mukhtar, H. Apoptosis and Autophagy Induction as Mechanism of Cancer Prevention by Naturally Occurring Dietary Agents. Curr. Drug Targets 2012, 13, 1831–1841. [Google Scholar] [CrossRef] [PubMed]
  247. Rahman, J.; Tareq, A.M.; Hossain, M.M.; Sakib, S.A.; Islam, M.N.; Ali, M.H.; Uddin, A.B.M.N.; Hoque, M.; Nasrin, M.S.; Emran, T.B.; et al. Biological Evaluation, DFT Calculations and Molecular Docking Studies on the Antidepressant and Cytotoxicity Activities of Cycas Pectinata Buch.-Ham. Compounds. Pharmaceuticals 2020, 13, 232. [Google Scholar] [CrossRef]
  248. Jiang, J.; Eliaz, I.; Sliva, D. Synergistic and Additive Effects of Modified Citrus Pectin with Two Polybotanical Compounds, in the Suppression of Invasive Behavior of Human Breast and Prostate Cancer Cells. Integr. Cancer Ther. 2013, 12, 145–152. [Google Scholar] [CrossRef] [PubMed]
  249. Ahmed, S.; Rakib, A.; Islam, M.A.; Khanam, B.H.; Faiz, F.B.; Paul, A.; Chy, M.N.U.; Bhuiya, N.M.M.A.; Uddin, M.M.N.; Ullah, S.M.A.; et al. In Vivo and in Vitro Pharmacological Activities of Tacca integrifolia Rhizome and Investigation of Possible Lead Compounds against Breast Cancer through in Silico Approaches. Clin. Phytosci. 2019, 5, 36. [Google Scholar] [CrossRef]
  250. Nsengiyumva, E.M.; Alexandridis, P. Xanthan Gum in Aqueous Solutions: Fundamentals and Applications. Int. J. Biol. Macromol. 2022, 216, 583–604. [Google Scholar] [CrossRef]
  251. Cortes, H.; Caballero-Florán, I.H.; Mendoza-Muñoz, N.; Escutia-Guadarrama, L.; Figueroa-González, G.; Reyes-Hernández, O.D.; González-Del Carmen, M.; Varela-Cardoso, M.; González-Torres, M.; Florán, B.; et al. Xanthan Gum in Drug Release. Cell. Mol. Biol. 2020, 66, 199–207. [Google Scholar] [CrossRef]
  252. Pei, Z.; Lou, Z.; Zhang, B.; Wang, H.; Li, Y. Development of a Compound Oral Liquid Containing Herbal Extracts and Its Effect on Immunity and Gastric Mucosa. J. Food Sci. 2021, 86, 2684–2699. [Google Scholar] [CrossRef]
  253. Takeuchi, A.; Kamiryou, Y.; Yamada, H.; Eto, M.; Shibata, K.; Haruna, K.; Naito, S.; Yoshikai, Y. Oral Administration of Xanthan Gum Enhances Antitumor Activity through Toll-like Receptor 4. Int. Immunopharmacol. 2009, 9, 1562–1567. [Google Scholar] [CrossRef] [PubMed]
  254. Shao, P.; Pei, Y.; Fang, Z.; Sun, P. Effects of Partial Desulfation on Antioxidant and Inhibition of DLD Cancer Cell of Ulva Fasciata Polysaccharide. Int. J. Biol. Macromol. 2014, 65, 307–313. [Google Scholar] [CrossRef]
  255. Shao, P.; Chen, X.; Sun, P. Chemical Characterization, Antioxidant and Antitumor Activity of Sulfated Polysaccharide from Sargassum Horneri. Carbohydr. Polym. 2014, 105, 260–269. [Google Scholar] [CrossRef]
  256. Li, A.N.; Li, S.; Zhang, Y.J.; Xu, X.R.; Chen, Y.M.; Li, H.B. Resources and Biological Activities of Natural Polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef] [PubMed]
  257. Zvyagintseva, T.N.; Usoltseva, R.V.; Shevchenko, N.M.; Surits, V.V.; Imbs, T.I.; Malyarenko, O.S.; Besednova, N.N.; Ivanushko, L.A.; Ermakova, S.P. Structural Diversity of Fucoidans and Their Radioprotective Effect. Carbohydr. Polym. 2021, 273, 118551. [Google Scholar] [CrossRef] [PubMed]
  258. Saliba, J.; Manseur, C.; Groult, H.; Akil, H.; Tannoury, M.; Troutaud, D.; Maugard, T.; Feuillard, J.; Arnaudin, I.; Jayat-Vignoles, C. Anti-Proliferative and Pro-Apoptotic vLMW Fucoidan Formulas Decrease PD-L1 Surface Expression in EBV Latency III and DLBCL Tumoral B-Cells by Decreasing Actin Network. Mar. Drugs 2023, 21, 132. [Google Scholar] [CrossRef]
  259. El-Sheekh, M.M.; Ward, F.; Deyab, M.A.; Al-Zahrani, M.; Touliabah, H.E. Chemical Composition, Antioxidant, and Antitumor Activity of Fucoidan from the Brown Alga Dictyota Dichotoma. Molecules 2023, 28, 7175. [Google Scholar] [CrossRef]
  260. Yang, J.; Yang, X.; Pan, W.; Wang, M.; Lu, Y.; Zhang, J.; Fang, Z.; Zhang, X.; Ji, Y.; Bei, J.-X.; et al. Fucoidan-Supplemented Diet Potentiates Immune Checkpoint Blockage by Enhancing Antitumor Immunity. Front. Cell Dev. Biol. 2021, 9, 733246. [Google Scholar] [CrossRef]
  261. Kiselevskiy, M.V.; Anisimova, N.Y.; Ustyuzhanina, N.E.; Vinnitskiy, D.Z.; Tokatly, A.I.; Reshetnikova, V.V.; Chikileva, I.O.; Shubina, I.Z.; Kirgizov, K.I.; Nifantiev, N.E. Perspectives for the Use of Fucoidans in Clinical Oncology. Int. J. Mol. Sci. 2022, 23, 11821. [Google Scholar] [CrossRef]
  262. Zhang, W.; Hwang, J.; Yadav, D.; An, E.K.; Kwak, M.; Lee, P.C.; Jin, J.O. Enhancement of Immune Checkpoint Inhibitor-Mediated Anti-Cancer Immunity by Intranasal Treatment of Ecklonia Cava Fucoidan against Metastatic Lung Cancer. Int. J. Mol. Sci. 2021, 22, 9125. [Google Scholar] [CrossRef]
  263. Park, H.B.; Hwang, J.; Lim, S.-M.; Zhang, W.; Jin, J.O. Dendritic Cell-Mediated Cancer Immunotherapy with Ecklonia Cava Fucoidan. Int. J. Biol. Macromol. 2020, 159, 941–947. [Google Scholar] [CrossRef]
  264. Tocaciu, S.; Oliver, L.J.; Lowenthal, R.M.; Peterson, G.M.; Patel, R.; Shastri, M.; McGuinness, G.; Olesen, I.; Fitton, J.H. The Effect of Undaria Pinnatifida Fucoidan on the Pharmacokinetics of Letrozole and Tamoxifen in Patients with Breast Cancer. Integr. Cancer Ther. 2018, 17, 99–105. [Google Scholar] [CrossRef]
  265. Figueroa, F.A.; Abdala-Díaz, R.T.; Pérez, C.; Casas-Arrojo, V.; Nesic, A.; Tapia, C.; Durán, C.; Valdes, O.; Parra, C.; Bravo-Arrepol, G.; et al. Sulfated Polysaccharide Extracted from the Green Algae Codium Bernabei: Physicochemical Characterization and Antioxidant, Anticoagulant and Antitumor Activity. Mar. Drugs 2022, 20, 458. [Google Scholar] [CrossRef]
  266. Xie, J.H.; Jin, M.L.; Morris, G.A.; Zha, X.Q.; Chen, H.Q.; Yi, Y.; Li, J.E.; Wang, Z.J.; Gao, J.; Nie, S.P.; et al. Advances on Bioactive Polysaccharides from Medicinal Plants. Crit. Rev. Food Sci. Nutr. 2016, 56 (Suppl. S1), S60–S84. [Google Scholar] [CrossRef]
  267. Chen, G.; Jiang, N.; Zheng, J.; Hu, H.; Yang, H.; Lin, A.; Hu, B.; Liu, H. Structural Characterization and Anti-Inflammatory Activity of Polysaccharides from Astragalus membranaceus. Int. J. Biol. Macromol. 2023, 241, 124386. [Google Scholar] [CrossRef]
  268. Tang, Z.; Huang, G. Extraction, Structure, and Activity of Polysaccharide from Radix Astragali. Biomed. Pharmacother. Biomed. Pharmacother. 2022, 150, 113015. [Google Scholar] [CrossRef]
  269. Berezutsky, M.A.; Matvienko, U.A.; Karetnikova, A.Y.; Durnova, N.A. Anti-Cancer Activity of Astragalus membranaceus—A Review. Int. J. Pharm. Res. 2021, 13, 09752366. [Google Scholar] [CrossRef]
  270. Yu, Z.; Guo, F.; Guo, Y.; Zhang, Z.; Wu, F.; Luo, X. Optimization and Evaluation of Astragalus Polysaccharide Injectable Thermoresponsive In-Situ Gels. PLoS ONE 2017, 12, e0173949. [Google Scholar] [CrossRef]
Figure 1. The impact of APS on immune cell activity in the tumor microenvironment. APS promotes the expression of CD40, CD80, and HLA-DR on dendritic cells, facilitating their maturation process. These mature dendritic cells subsequently stimulate the production of IL-12, direct antigens towards lymphocytes, shift Th2 cell populations towards Th1, and reinforce the immune response of T cells and NK cells. This leads to an increase in IL-2, TNF-α, IFN-γ, and IL-17A by T cells, accompanied by a reduction in IL-10. NK cells exhibit elevated levels of IFN-γ, granzyme B, and perforin; APS also enhances the proportion of M1/M2 macrophages and promotes the secretion of NO, CXCL10, IL-6, and TNF-α, thereby enhancing the humoral immunity of B cells. The solid line is the effect that one cell can have on another cell, and the dashed line is the effect that the cell has on immunity. Down and up directions means more or less related factors.
Figure 1. The impact of APS on immune cell activity in the tumor microenvironment. APS promotes the expression of CD40, CD80, and HLA-DR on dendritic cells, facilitating their maturation process. These mature dendritic cells subsequently stimulate the production of IL-12, direct antigens towards lymphocytes, shift Th2 cell populations towards Th1, and reinforce the immune response of T cells and NK cells. This leads to an increase in IL-2, TNF-α, IFN-γ, and IL-17A by T cells, accompanied by a reduction in IL-10. NK cells exhibit elevated levels of IFN-γ, granzyme B, and perforin; APS also enhances the proportion of M1/M2 macrophages and promotes the secretion of NO, CXCL10, IL-6, and TNF-α, thereby enhancing the humoral immunity of B cells. The solid line is the effect that one cell can have on another cell, and the dashed line is the effect that the cell has on immunity. Down and up directions means more or less related factors.
Pharmaceuticals 17 00636 g001
Figure 2. Diagram of the mechanism by which APS induces apoptosis in cancer cells by interfering with the cell cycle, inhibiting signal transduction pathways, and inducing apoptosis. This involves the induction of S, G2/M, and G0/G1 cell cycle arrest via the Wnt-β-catenin pathway. It is involved in the inhibition of PI3K/Akt, ERK/MAPK, and NF-κB pathways in tumor therapy and induces apoptosis in cancer cells by inhibiting the expression of Bcl-2 proteins and miRNAs and upregulating the expression of p53 proteins. Yellow represents the factor on the receptor, which is also the initiating factor of the pathway; Green represents factors on the conventional signaling pathway; Orange represents factors involved in both apoptosis and autophagy; Pink like Bax represents both apoptosis and cell cycle involvement; Earthy yellow represents cell cycle related factors; Blue represents cytokines involved in apoptosis alone. The red up arrow indicates that Astragalus polysaccharide leads to the increase of related factors, and the blue down arrow indicates that Astragalus polysaccharide leads to the decrease of related factors.
Figure 2. Diagram of the mechanism by which APS induces apoptosis in cancer cells by interfering with the cell cycle, inhibiting signal transduction pathways, and inducing apoptosis. This involves the induction of S, G2/M, and G0/G1 cell cycle arrest via the Wnt-β-catenin pathway. It is involved in the inhibition of PI3K/Akt, ERK/MAPK, and NF-κB pathways in tumor therapy and induces apoptosis in cancer cells by inhibiting the expression of Bcl-2 proteins and miRNAs and upregulating the expression of p53 proteins. Yellow represents the factor on the receptor, which is also the initiating factor of the pathway; Green represents factors on the conventional signaling pathway; Orange represents factors involved in both apoptosis and autophagy; Pink like Bax represents both apoptosis and cell cycle involvement; Earthy yellow represents cell cycle related factors; Blue represents cytokines involved in apoptosis alone. The red up arrow indicates that Astragalus polysaccharide leads to the increase of related factors, and the blue down arrow indicates that Astragalus polysaccharide leads to the decrease of related factors.
Pharmaceuticals 17 00636 g002
Figure 3. Effects of APS on ICIs, CAR-T therapy, and DC cancer vaccines within the TME. This involves the enhancement of ICIs through the PI3K/Akt/mTOR pathway. APS-induced activation of DCs improves the effectiveness of adoptive immunotherapy with cytokine-induced killer (CIK) cells and APS components, particularly abundant in dextran, which act as potent adjuvants for DC-based cancer immunotherapy vaccines. The blue arrow belongs to the APS-stimulated adoptive cell immunotherapy arrow, the purple arrow belongs to the APS-stimulated immune checkpoint inhibitor arrow, and the pink arrow belongs to the APS-stimulated DC cancer vaccine arrow. The black up arrow indicates an increase in a certain factor during APS stimulation, and the black down arrow indicates a decrease in a certain factor during APS stimulation.
Figure 3. Effects of APS on ICIs, CAR-T therapy, and DC cancer vaccines within the TME. This involves the enhancement of ICIs through the PI3K/Akt/mTOR pathway. APS-induced activation of DCs improves the effectiveness of adoptive immunotherapy with cytokine-induced killer (CIK) cells and APS components, particularly abundant in dextran, which act as potent adjuvants for DC-based cancer immunotherapy vaccines. The blue arrow belongs to the APS-stimulated adoptive cell immunotherapy arrow, the purple arrow belongs to the APS-stimulated immune checkpoint inhibitor arrow, and the pink arrow belongs to the APS-stimulated DC cancer vaccine arrow. The black up arrow indicates an increase in a certain factor during APS stimulation, and the black down arrow indicates a decrease in a certain factor during APS stimulation.
Pharmaceuticals 17 00636 g003
Table 1. Effects of APS on immune cells (DC, B, T, macrophage, NK).
Table 1. Effects of APS on immune cells (DC, B, T, macrophage, NK).
Model SourceDosageEffect of APS on Immune CellsReference
C57BL/6 mice200 μg/mL, 24 hbone marrow-derived dendritic cells(BMDCs) ↑
Splenic DCs in mice 6 h ↑18 h ↓
Human peripheral blood DCs (PBDCs) 6 h ↑
[26]
C57BL/6J mice45 μg/mL, 24 hBMDC ↑[27]
BALB/c mice200 μg/mL, 24 hCD8+ T ↑
CD4+ T ↑
pDC ↑
[28]
BALB/c mice50 μg/mL, 24 hBMDC ↑
(CD4+ T/Treg cell)/(CD8+ T/Treg cell) ↑
CD4+ T ↑
[29]
C57BL/6 mice16 mg/mL, 48 hM1/M2 ↑
CD11c(high) CD45RB(low) DCs ↑
Th1/Th2 ↑
[30]
C57BL/6 mice50 mg/kg, 18 hCD11c+ DC ↑
CD3-NK1.1+
CD8+ T ↑
[31]
Kunming mice
C57 mice
100 mL/kg, 3 daysCD4+ T/CD8+ T ↑[32]
C57BL/6J mice0.2 mg/mLTreg ↓
BMDC ↑
CD3+CD8+ T ↑
MHC-I+CD11c+ cells ↑
[33]
Swiss-Hauschka (ICR) mice5 mg/mL, 14 daysTh1/Th2 ↑
CD4+ T/CD8+ T ↑
[34]
Spleen lymphocyte0.156 mg/mLTh1/Th2 ↑
CD4+ T/CD8+ T ↑
[35]
BALB/c mice300 μg/mLMacrophage (M2 → M1) ↑[36]
Mouse peritoneal macrophages100 ng/mL, 24 hRaw 264.7 ↑[37]
Murine macrophage RAW 264.7 cells2000 μg/mL, 24 hRaw 264.7 ↑ (IL-1α, IL-1β, IL-6 ↑, NO ↓)[38]
The fresh lung tissues of adult pigs500 μg/mL or 1000 μg/mL, 24 hRaw 264.7 ↑ (TNF-α, NO ↑)[39]
BALB/c mice or C3H/HeJ mice100 μg/mL, 6 hRaw 264.7 ↑, B ↑ (IL-1β, TNF-α ↑)[40]
ICR mice10 g/kgCD8+ T ↑
CD4+ T ↑
NK ↑
CTL ↑
[41]
BALB/c mice100, 200, 300 mg/kg, 30 daysRaw 264.7 ↑
NK ↑
B ↑
T ↑
[42]
BALB/c mice100 mg/kgNK ↑[43]
Arrows in down and up directions means more or less related factors: ↑ increased, ↑ decreased.
Table 2. Effect of APS on cell cycle, signal transduction pathways, and apoptosis.
Table 2. Effect of APS on cell cycle, signal transduction pathways, and apoptosis.
Model SourceDosageEffect of APS Cell Cycle, Signal Transduction Pathways, and ApoptosisReference
Kunming mice150, 300 mg/kg, 15 daysG1 phase retention ↓
S phase retention ↑
[56]
MGC-803 cell200, 400, 800 μg/mL, 24 hG0/G1 phase retention ↓
S phase retention ↑
G2/M phase retention ↓
[57]
HepG2 cell200, 400, 800 μg/mL, 72 hS phase retention ↑[58]
4T1 cell50, 100, 200, 500, 1000 μg/mL, 72 hG2/M phase retention ↑[59]
4T1 cell50 μg/mL, 24 hG1 phase retention ↓
G2/M phase retention ↑
[60]
MCF-7 cell500, 1000 μg/mL, 24 hG1 phase retention ↑[61]
SCG-1 cells25, 50, 100 mg/L, 12 hG0 phase retention ↑[62]
CNE-1 cell200 μg/mL, 24 hG0/G1 phase retention ↑
S phase retention ↑
[63]
AGS cell200 μg/mL, 24 hPI3K/Akt pathway ↓[64]
B16F10 cell1–5 mg/mL, 24 hPI3K/Akt pathway ↓[65]
C57BL/6 mice200 mg/kg, 24 hPI3K/Akt pathway ↓[66]
RAW264.7 cell0–100 μg/mL, 24 hNF-κB/MAPK pathway ↑
P65 protein ↑
G2/M phase retention ↑
[67]
GC-SGC-7901 cell
GCSGC-7901/ADR cell
GES-1 cell
100–400 μg/mL, 24 hp-AMPK level ↑[68]
A549 cell
NCI-H358 cell
20, 40 mg/mLNF-κB pathway ↓[69]
PANC-1 cell0, 1, 5, 10, 15, 20 mg/mLNF-κB P65 ↓
TLR4/NF-κB pathway ↓
[70]
A549 cell
BMSC cell
50 μg/mLRAS, ERK, NF-κB p65 protein ↓
TP53, caspase-3 protein ↑
[71]
HepG2 cell
Diethylnitrosamine-induced HCC in rats
60, 120, 240 mg/kg (rats)
20, 40, 80 µg/mL (HepG2 cell)
TGF-β/MAPK/Smad pathway ↑
miR-145 ↑, miR-21 ↓
pSmad3L→pSmad3C ↑
[72]
HepG2 cell40 mg/mLTGF-β/Smad pathway ↑
pSmad3L→pSmad3C ↑
[73]
H22 cell
Kunming mice
100, 200 mg/kg, 16 daysBax ↑
Bcl-2 ↓
[74]
MDA-MB-231 cell
BALB/C mice
200, 400 mg/kg, 21 daysBax, Caspase7, Caspase9 ↑
Bcl-2 ↓
[75]
HepG2 cell100, 200 mg/L, 48 hBcl-2 ↓
Caspase3 ↑
β-catenin, c-myc, Cyclin D1 mRNA ↓
[76]
SKOV3 cell800 µg/mL, 24 hBcl-2 ↓
Bax, Caspase3 ↑
[77]
BALB/C mice
CNE-1 cell
CNE-2 cell
SUNE-1 cell
40 µg/mL, 48 hBcl-2 ↓
Bax, Caspase3, Caspase9 ↑
[78]
OV-90 cell
SKOV-3 cell
1 mg/mL, 24 hmiR-27a ↑
Caspase3 ↑
[79]
OS MG63 cell10 mg/mL, 24 hmiR-133a ↑
S phase retention ↑
CyclinD1 ↓ p21 ↑
Bcl-2 ↓
Bax, Caspase3, Caspase9 ↑
[80]
A549 cell
NCIH1299 cell
0, 5, 10, 20 µg/mL,miR-195-5p ↑[81]
H460 cell0–30 mg/mL, 24/48 hP53, P21, P16 ↑
Notch1,Notch3 ↑
Bcl-2 ↓
Bax, Caspase8 ↑
[82]
CD133+/CD44+ cell0, 12.5, 25, 50 mg/mL, 48 hFas
Caspase 3, Caspase 9, Fas, Bax ↑
Class III PI3K, Beclin 1 ↑
Bcl-2, XIAP ↓
LC3-I ↓LC3-II ↑
[83]
Hep3B cell
BALB/c mice
10 mg/LCHOP ↑
Bcl-2 ↓
Bax, Bim, Caspase3 ↑
OGT ↓OGA ↑
O-GlcNAc ↓
[84]
Arrows in down and up directions means more or less related factors: ↑ increased, ↑ decreased.
Table 3. Clinical investigations of APS in cancer.
Table 3. Clinical investigations of APS in cancer.
NumberPharmaceutical IngredientDiseaseStatusPhase
NCT01802021 [161]Astragalus-based FormulaNon-small-cell
lung cancer
RecruitingII/III
ChiCTR2300068199 [162]Astragalus polysaccharides, Paclitaxel; CarboplatinNon-small-cell
lung cancer
Not Recruiting
[163]Astragalus polysaccharides, Iodide 125Lung cancerNot RecruitingII/III
[25]Astragalus polysaccharides, Vinorelbine; CisplatinNon-small-cell
lung cancer
Not RecruitingII/III
[164]Astragalus polysaccharides, Gemcitabine; CisplatinNon-small-cell
lung cancer
Not RecruitingII/III
ITMCTR2100004716 [165]Astragalus polysaccharides, ApatinibExtensive-stage small-cell
lung cancer
Not Recruiting
ChiCTR2000040911 [166]Astragalus polysaccharides, Carrelizumab; ApatinibLung cancerRecruiting
ITMCTR2000003215 [167]Astragalus polysaccharides, TansychiaHepatocellular carcinoma;
Lung cancer
Recruiting
NCT03314805 [168]Astragalus polysaccharidesBreast cancerNot recruitingII
ChiCTR2300076131 [169]Chinese herbal decoction (contains Astragalus polysaccharides)Low-grade gastric intraepithelial neoplasiaRecruiting
[170]Astragalus polysaccharides, FOLFOXGastric cancerNot recruiting
ChiCTR2300068896 [171]Astragalus polysaccharidesGastric cancerNot recruitingIV
NCT06234072 [172]Astragalus polysaccharides, GemcitabinePancreatic cancerNot recruitingII
[173]Traditional Korean Medicine (contains Astragalus polysaccharides), CTXMetastatic
pancreatic cancer
Not recruiting
ChiCTR2000037982 [174]Astragalus,
Panax-notoginseng, Oncolytic vaccine
Colorectal cancerRecruiting
[175,176]Astragalus polysaccharidesColorectal cancerNot recruiting
[177]FOLFOX 4, Ginseng,
Astragalus polysaccharides,
Atractylodes rhizome,
Poria cocos, Coix seed,
Sophora flavescens
Advanced
colorectal cancer
Not recruiting
[178]Astragalus polysaccharides,
Oxaliplatin, Poria cocos,
Atractylodes macrocephala Koidz, Pilosula
Colorectal cancerNot recruiting
NCT01720563 [179]APS, Cisplatin, Leucovorin,
Tegafur plus uracil
Advanced pharyngeal/laryngeal squamous
cell carcinoma
Not recruitingII
NCT03611712 [180]Astragalus polysaccharidesLocally advanced esophageal cancerNot recruitingII
NCT01720550 [181]Astragalus polysaccharidesAdvanced cancerNot recruitingIV
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

He, Z.; Liu, X.; Qin, S.; Yang, Q.; Na, J.; Xue, Z.; Zhong, L. Anticancer Mechanism of Astragalus Polysaccharide and Its Application in Cancer Immunotherapy. Pharmaceuticals 2024, 17, 636. https://doi.org/10.3390/ph17050636

AMA Style

He Z, Liu X, Qin S, Yang Q, Na J, Xue Z, Zhong L. Anticancer Mechanism of Astragalus Polysaccharide and Its Application in Cancer Immunotherapy. Pharmaceuticals. 2024; 17(5):636. https://doi.org/10.3390/ph17050636

Chicago/Turabian Style

He, Ziqing, Xiyu Liu, Simin Qin, Qun Yang, Jintong Na, Zhigang Xue, and Liping Zhong. 2024. "Anticancer Mechanism of Astragalus Polysaccharide and Its Application in Cancer Immunotherapy" Pharmaceuticals 17, no. 5: 636. https://doi.org/10.3390/ph17050636

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop