Volume: 3 | Issue: 3 | View as PDF | Cite
  • OPEN ACCESS

Ginsenosides as Multi-target Agents against Hepatocellular Carcinoma: Mechanisms and Therapeutic Prospects

  • Chang-Hai Luan* 
 Author information
Nature Cell and Science   2025;3(3):e00016

doi: 10.61474/ncs.2025.00016

Abstract

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with limited therapeutic options and poor prognosis. Ginsenosides, the bioactive constituents of Panax ginseng, have emerged as promising anti-HCC agents due to their multi-targeted mechanisms. This review comprehensively summarizes the anti-HCC effects of ginsenosides, including induction of apoptosis; inhibition of invasion and metastasis; inhibition of proliferation; restricting glucose consumption; reversal of multidrug resistance; and enhancement of host immunity. Key findings highlight the role of ginsenosides such as Rg3, Rh2, and compound K in regulating critical signaling pathways (e.g., PI3K/AKT, MAPK, and AMPK), oxidative stress, and immune responses. Additionally, ginsenosides exhibit synergistic effects when combined with conventional therapies, improving efficacy and overcoming drug resistance. Despite these advances, further research is needed to elucidate the precise molecular targets and optimize clinical applications. This review underscores the potential of ginsenosides as adjunctive or alternative therapies for HCC, providing a foundation for future translational studies.

Keywords

Ginsenoside, Hepatocellular carcinoma, Apoptosis, Multidrug resistance, Tumor microenvironment, Clinical translation

Introduction

Primary liver cancer is one of the most common malignant tumors worldwide, with approximately 865,269 new cases and 757,948 deaths reported globally in 2022, making it the third leading cause of cancer-related mortality.1–4 Hepatocellular carcinoma (HCC) represents the predominant pathological type, accounting for 75–85% of primary liver cancer cases.5,6 Major risk factors include chronic hepatitis B/C virus infection, cirrhosis, and metabolic diseases.7 Due to its asymptomatic early stages, most patients are diagnosed at advanced stages.7 Current therapeutic approaches include surgical resection, liver transplantation, transarterial chemoembolization (TACE), radiofrequency ablation, and targeted therapy, yet challenges such as tumor recurrence, drug resistance, and adverse effects persist.8 Although liver transplantation remains a curative option, its application is limited by donor shortage and post-transplant recurrence.9 The overall prognosis of HCC remains poor, with a five-year survival rate of only ∼10%,10 and palliative care is often administered for advanced-stage patients.11

Traditional Chinese medicine (TCM) serves as an important component of comprehensive treatment, demonstrating the potential to prolong patient survival,12 improve postoperative survival rates, and enhance quality of life.13,14 For instance, herbal formulations such as Fuzheng Jiedu Xiaoji formulation have shown clinically confirmed efficacy.15 Integrated Chinese-Western therapy (e.g., TCM combined with radiofrequency ablation) has been reported to improve short-term therapeutic outcomes and quality of life.16 Network pharmacology studies reveal that TCM exerts multi-target inhibitory effects on HCC progression, including modulation of key signaling pathways.17 Additionally, TCM demonstrates potential in delaying disease progression and mitigating adverse effects associated with radiotherapy and chemotherapy.18

Recent studies have revealed that ginsenosides, the primary bioactive constituents of Panax ginseng, exhibit multi-faceted potential in cancer therapy. In 1983, Japanese researchers first isolated ginsenoside Rg3 (GRg3) from Korean red ginseng. As a tetracyclic triterpenoid dammarane-type rare ginsenoside, GRg3 is present in low quantities in fresh ginseng, American ginseng, and Panax notoginseng roots/rhizomes, but accumulates at higher levels in aged ginseng, ginseng stems/leaves, processed red ginseng, and black ginseng.19 Structurally classified as a protopanaxadiol-type saponin, GRg3 has a molecular weight of 784 Da. It can be derived from the thermal or enzymatic degradation of natural ginsenosides Rb1, Rb2, and Rd.19 The spatial configuration at the C20 position generates two enantiomers: 20(S)-Rg3 and 20(R)-Rg3.20 The tertiary structural differences between these isomers result in superior anti-tumor activity of 20(S)-Rg3 compared to its R-form. Specifically, 20(S)-Rg3 demonstrates significantly stronger inhibition of HepG2 cell proliferation and apoptosis induction.21 The alkene chain in the aglycone moiety of 20(S)-Rg3 forms tight hydrophobic packing near C20, enhancing hydrogen bonding with Ca2+/Na+ ion channels and ligand-gated ion channels, thereby strengthening interactions with receptor domains.22

Current evidence indicates that various ginsenoside subtypes exert anti-tumor effects through multiple mechanisms, including induction of apoptosis, suppression of proliferation and metastasis, modulation of the tumor microenvironment, and reversal of chemotherapy resistance.

This review aims to systematically summarize the multi-targeted anti-HCC mechanisms of ginsenosides, discuss their synergistic potential with conventional therapies, and highlight future directions for clinical translation (Fig. 1).

Ginsenosides and their anti-hepatocellular carcinoma mechanisms.
Fig. 1  Ginsenosides and their anti-hepatocellular carcinoma mechanisms.

Inhibition of HCC cell proliferation

The pathogenesis of HCC is closely associated with an imbalance between cell proliferation and apoptosis, where excessive proliferation over apoptosis leads to malignant tumor growth. Ginsenoside compound K suppresses HepG2 cell proliferation by inducing ferroptosis.23,24 Its acetylated derivative, compound K-3, further enhances the antitumor effect by arresting the cell cycle at the G2/M phase and inducing apoptosis.24

Additionally, ginsenoside Rk1 exerts anti-proliferative effects in HCC models by inducing autophagy-dependent apoptosis.25 Rg3 effectively inhibits the growth of human HCC cell lines through dual mechanisms: suppressing cancer cell proliferation and promoting apoptosis. This pro-apoptotic effect may occur via the intrinsic mitochondrial-mediated caspase-dependent pathway.26

Notably, Rg3 inhibits HCC cell proliferation and induces apoptosis in a concentration- and time-dependent manner in vitro. Studies demonstrate that Rg3, either alone or in combination with cyclophosphamide, suppresses tumor growth in vivo and prolongs survival in mouse models. This effect is mediated through the intrinsic apoptotic pathway, involving altered expression of B-cell lymphoma 2 (Bcl-2) family proteins.27

Inhibition of HCC cell invasion and metastasis

Tumor invasion and metastasis constitute a complex, multi-step cascade process regulated by distinct genetic mechanisms. Both ginsenosides Rh1 and Rb1 demonstrate concentration- and time-dependent inhibition of HepG2 cell invasion and metastasis.28,29 Rd-treated HepG2 cells exhibit significantly suppressed metastatic potential, primarily through inhibition of matrix metalloproteinase (MMP) activation and modulation of the MAPK signaling pathway involved in tumor cell migration.30

Ginsenoside Rh2 effectively attenuates the migratory capacity of HepG2 cells by activating glycogen synthase kinase-3 beta, degrading β-catenin, and downregulating metastasis-related proteins such as MMP3, thereby reducing tumor invasion and migration.31 Furthermore, compound K, a metabolite of ginsenosides, has also demonstrated anti-invasive and anti-metastatic properties, which may be attributed to the translocation of nuclear factor-κB p65 and the reduction of MMP-2 and MMP-9.32 Additionally, in the diethylnitrosamine-induced HCC mouse model, NpRg3 (Fe@Fe3O4 conjugated with GRg3) eliminates HCC lung metastasis, possibly by remodeling gut microbiota and metabolism.33

Induction of apoptosis in HCC cells

One of the key factors contributing to the rapid growth of HCC is the reduced apoptosis of tumor cells. Consequently, artificial induction of apoptosis in HCC cells has become a research focus in anticancer therapy. Ginsenoside Rh2 has been demonstrated to effectively induce apoptosis in human SK-HEP-1 HCC cells. The pro-apoptotic mechanisms of ginsenosides against HCC primarily involve the following molecular pathways:

Ginsenosides can regulate apoptosis-related proteins. For instance, GRg3 and G protein-coupled receptor kinase 1 (GRk1) promote apoptosis in HCC cells by modulating the expression of apoptosis-associated proteins. Studies indicate that GRg3 and GRk1 may induce apoptosis through downregulation of the anti-apoptotic protein Bcl-2 and upregulation of pro-apoptotic proteins such as caspase-3.34,35 Additionally, research demonstrates that ginsenoside compound K can influence the expression of other apoptosis-related proteins, including caspase-8, Fas, and FasL.36

Rh2 induces rapid and significant translocation of pro-apoptotic proteins Bax and Bak, subsequently triggering mitochondrial cytochrome C release and caspase activation.37 Experimental studies revealed that small interfering RNA-based caspase-8 gene inactivation effectively delays Rh2-induced caspase-9 activation and apoptosis. These findings suggest that Rh2 employs multiple pro-apoptotic pathways to execute cancer cell death.37 Investigations on the effects of Korean red ginseng and its major components Rg3 and Rh2 on apoptosis in human Hep3B HCC cells showed decreased cell viability and mitochondrial membrane potential, along with increased lactate dehydrogenase release, confirming the significant cytotoxic and pro-apoptotic effects of Korean red ginseng, Rg3, and Rh2 on Hep3B cells.38

Ginsenosides can inhibit the PI3K/AKT signaling pathway. For example, GRk3 enhances apoptosis in HCC cells by suppressing the PI3K/AKT pathway. As this pathway serves as a crucial regulator of cell survival and proliferation, its inhibition by GRk3 leads to enhanced pro-apoptotic effects.39

Ginsenosides can modulate tumor metabolism. Specifically, ginsenoside Rk1 inhibits HCC proliferation by regulating glutamine metabolism. This mechanism involves suppression of the ERK/c-Myc signaling pathway, downregulation of glutaminase glutaminase 1 expression, reduction in glutathione production, and concurrent increase in reactive oxygen species accumulation, thereby promoting apoptosis both in vitro and in vivo.35

In summary, ginsenosides induce apoptosis in HCC cells through multiple pathways, including the regulation of apoptosis-related proteins (e.g., Bcl-2, caspases), inhibition of the PI3K/AKT pathway, modulation of tumor metabolism, and oxidative stress. These mechanisms collectively contribute to their anti-HCC efficacy.

Restricting glucose consumption

Due to impaired or lost glucagon inactivation in the liver of HCC patients, hepatogenous diabetes often develops, making blood glucose reduction an essential component of HCC treatment. Ginsenoside Rh4 inhibits inflammation-related HCC progression by targeting the HDAC4/IL-6/STAT3 signaling pathway. This mechanism involves downregulation of key glycolytic enzymes glucose transporter 1 and lactate dehydrogenase A, leading to reduced glucose consumption and lactic acid production, thereby suppressing glycolysis and HCC growth.40 Similarly, GRg3, in combination with sorafenib, inhibits HCC progression by suppressing HK2-mediated glycolysis, leading to reduced glucose consumption and lactate production.41

Reversal of multidrug resistance in HCC cells

Multidrug resistance refers to a phenomenon where tumor cells exposed to a particular chemotherapeutic agent develop resistance not only to that specific drug but also to other structurally unrelated compounds with different mechanisms of action. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent due to its selective cytotoxicity against tumor cells. However, prolonged use often leads to drug resistance.

Studies have demonstrated that standardized red ginseng extract (RGE) and 20(S)-Rg3 can sensitize HCC cell lines to TRAIL-induced cell death.42 Experimental evidence confirms that RGE-associated upregulation of the transcription factor CCAAT/enhancer-binding protein homologous protein plays a crucial role in sensitizing TRAIL-mediated apoptosis. Furthermore, Rg3 has been shown to upregulate the expression of CCAAT/enhancer-binding protein homologous protein-mediated death receptor-5 at the transcriptional level, thereby enhancing TRAIL-induced apoptosis in various HCC cell lines. These findings suggest that both RGE and Rg3 may serve as effective chemosensitizers when combined with TRAIL therapy to reverse multidrug resistance in HCC cells.43,44

Enhancement of host immune function

The immune function of the organism is closely associated with tumor development and progression. Immunosuppression or compromised immune function leads to increased tumor incidence, while progressive tumor growth conversely exacerbates immune suppression, forming a reciprocal causal relationship. Consequently, immune enhancement has become an indispensable component of antitumor therapy. GRg3 has been demonstrated to inhibit the growth of H22 hepatoma cell xenografts in vivo, at least partially through a stereospecific mechanism that enhances host cellular immunity (Table 1).23-41,43,45-47,48

Table 1

Structure and anti-hepatocellular carcinoma mechanism of ginsenosides

NameStructureMechanismReferences
Ginsenoside Rg3
1. Inhibits proliferation; 2. Induces apoptosis; 3. Inhibits invasion and metastasis; 4. Suppresses glycolysis; 5. Enhances chemotherapy sensitivity; 6. Enhances host immunity; 7. Reversal of multidrug resistance; 8. Anti-angiogenesis26,27,33,34,41,43,45,47,48
Ginsenoside Rh2
1. Inhibits proliferation; 2. Induces apoptosis; 3. Inhibits migration31,37,38
Ginsenoside compound K
1. Induces ferroptosis; 2. Inhibits proliferation; 3. Suppresses metastasis; 4. Induces apoptosis23,24,32,36
Ginsenoside Rk1
1. Inhibits proliferation; 2. Induces apoptosis25,35
Ginsenoside Rh1
1. Inhibits invasion and metastasis29
Ginsenoside Rd
1. Inhibits invasion and metastasis30
Ginsenoside Rb1
1. Inhibits invasion and metastasis;28
Ginsenoside Rk3
1. Induces apoptosis; 2. Induces autophagy39
Ginsenoside Rh4
1. Suppresses glycolysis40

Synergistic effects in combination with other therapeutic approaches

Currently, monotherapy demonstrates limited efficacy in HCC treatment, making the combination of different therapeutic strategies to suppress tumor growth through multiple pathways a critical research direction.

Rg3 has been shown to inhibit autophagy and enhance the sensitivity of HCC cell lines to doxorubicin in vitro.47 Rh2, when combined with betulinic acid, synergistically induces apoptosis in human HCC HepG2 cells, suggesting that this combination could serve as a novel strategy to improve the efficacy of anticancer therapies.49 In a murine H22 HCC model, Shen-Ling-Bai-Zhu Powder—containing ginsenosides Rb1 and Rg1—potentiated the antitumor effect of cyclophosphamide. The combination synergistically suppressed tumor growth by inhibiting the Akt/NF-κB pathway, downregulating anti-apoptotic proteins (Bcl-2, Bcl-xL, Survivin), and enhancing pro-apoptotic caspase-3/9 activation. This demonstrates ginsenosides’ role in a multi-component formula to promote apoptosis and improve chemotherapy efficacy in HCC.50

In addition, an optimized QHF formula containing GRg3, cinobufotalin, panax notoginseng saponins, and lentinan was developed using a uniform design. In HCC mice, the QHF formula demonstrated superior antitumor efficacy (55.91% tumor inhibition, 38.13% prolonged survival) compared to any single agent. When combined with cisplatin (DDP), QHF synergistically enhanced tumor growth inhibition (82.54%) and survival (87.02%), while significantly alleviating DDP-induced toxicities such as leukopenia and immune organ atrophy. This highlights GRg3’s role within a multi-component formula to potentiate chemotherapy and reduce adverse effects in HCC treatment (Table 2).34,41–43,47,49–51

Table 2

Summary of ginsenoside-based combination therapies for HCC

Combination therapy regimenTarget (cell line/animal model)Synergistic effectsReferences
Ginsenoside Rg3 + CyclophosphamideH22 HCC mouse modelInhibits tumor growth and prolongs survival in mice34
Ginsenoside Rg3 + SorafenibHCC cell lines / HCC mouse modelsInhibits HCC progression and reduces glucose consumption as well as lactate production41
Ginsenoside Rg3 + DoxorubicinHCC cell lines (in vitro)Enhances the sensitivity of HCC cells to doxorubicin47
Ginsenoside Rg3 (in QHF formula) + Cisplatin (DDP)HCC mouse modelsImproved tumor inhibition rate (82.54%), prolonged survival (87.02%), and alleviated DDP-induced toxicities (e.g., leukopenia, immune organ atrophy)51
Ginsenoside Rh2 + Betulinic AcidHepG2 HCC cell line (in vitro)Synergistically induces apoptosis in HCC cells49
Ginsenosides Rb1/Rg1 (in Shen-Ling-Bai-Zhu Powder) + CyclophosphamideH22 HCC mouse modelInhibits tumor growth and promotes apoptosis of HCC cells50
Standardized red ginseng extract (RGE) / 20(S)-Rg3 + TRAILHCC cell lines (in vitro)Reverses TRAIL resistance and enhances TRAIL-induced apoptosis in HCC cells42,43

Key challenges in clinical translation

The compelling preclinical data summarized in this review firmly establish ginsenosides as promising anti-HCC agents. Their multi-targeted actions—inducing apoptosis, inhibiting metastasis, reversing multidrug resistance, and modulating immunity—align perfectly with the complex pathophysiology of HCC. This polypharmacological profile is particularly advantageous for treating a heterogeneous and adaptive cancer like HCC.

Building upon the promising preclinical evidence, the clinical efficacy of ginsenosides, particularly Rg3, has been preliminarily validated in a prospective, randomized controlled trial. A study by Zhou et al.52 demonstrated that in patients with advanced HCC (BCLC stage C), the combination of TACE and GRg3 significantly prolonged median overall survival compared to TACE alone (13.2 months vs. 10.1 months; HR = 0.63, P = 0.002), alongside an improved disease control rate and a reduction in several TACE-related adverse events. This landmark study provides a crucial clinical foundation for the application of ginsenosides in HCC management.

Despite these encouraging clinical findings, several significant challenges remain for the broader clinical translation of ginsenosides.

A significant hurdle for clinical translation is the inherently poor oral bioavailability of many ginsenosides.53 This is due to their large molecular size, low permeability, and extensive metabolism by gut microbiota. Efforts to overcome this include the development of novel drug delivery systems, such as nanoparticles, liposomes, and micelles, which can enhance solubility, protect against degradation, and promote targeted delivery to tumor tissue.

While some ginsenoside preparations (e.g., Yangzheng Xiaoji Capsule) are approved as adjuvants for cancer therapy in China, large-scale, randomized, placebo-controlled clinical trials specifically focused on HCC outcomes are still limited. Most existing human data come from retrospective studies or small-scale trials, which, while encouraging, are not sufficient to establish ginsenosides as standard care.

Future directions and strategies in translational research

Given their established role as chemosensitizers and radiosensitizers, a pragmatic clinical pathway is to develop ginsenosides as adjuvants to existing therapies. Combining ginsenosides with sorafenib or lenvatinib may enhance efficacy and overcome resistance. Furthermore, their integration with immune checkpoint inhibitors holds significant promise. Ginsenosides, through potent anti-angiogenic effects via vascular endothelial growth factor downregulation,48 can help normalize the immunosuppressive tumor microenvironment, thereby potentially augmenting T-cell infiltration and response to immunotherapy.54

Furthermore, a comprehensive safety profile and toxicity assessment of ginsenosides must be established as a priority for clinical translation. Although most preclinical studies have reported a favorable safety profile with no significant toxicity, contradictory reports of potential hepatotoxicity underscore the necessity for rigorous, systematic evaluation.55 Future investigations should therefore focus on elucidating the long-term safety, potential drug-drug interactions, and dose-limiting toxicities of ginsenosides, particularly in combination regimens. Defining the therapeutic window will be crucial for optimizing dosing schedules and mitigating adverse effects, ensuring that the promising efficacy of ginsenoside-based therapies can be safely realized in a broader patient population.

Moreover, overcoming the poor oral bioavailability of many ginsenosides is critical for their clinical success. Future research should prioritize pharmaceutical optimization strategies, including structural modification of the ginsenoside backbone to enhance metabolic stability and permeability, the development of advanced nanoformulations (e.g., liposomes or polymeric nanoparticles) for improved solubility and targeted delivery, and the exploration of prodrug approaches to facilitate intestinal absorption. Success in these areas will be pivotal for achieving therapeutic systemic concentrations and maximizing the clinical potential of ginsenosides.

Conclusions

Extensive experimental studies have demonstrated that various ginsenoside monomers can exert anti-HCC effects through multiple pathways. However, current research on the mechanisms of individual ginsenosides in HCC treatment remains insufficiently comprehensive, with their primary signaling pathways and molecular targets not yet fully elucidated. Clinically, while GRg3 combined with TACE shows promise in advanced HCC, key challenges persist: most ginsenosides have poor oral bioavailability, and large-scale placebo-controlled RCTs for HCC are scarce. Future research directions should include developing targeted delivery systems (e.g., nanoparticles, liposomes) and combining ginsenosides with sorafenib or immune checkpoint inhibitors to realize their preclinical potential.

By systematically addressing these translational challenges, ginsenosides are poised to transition from promising preclinical agents to integral components of personalized, multi-targeted HCC treatment regimens in the clinic.

Declarations

Acknowledgement

The schematic diagrams in this article were created using templates from BioRender.com. The publication rights have been secured through a subscription license (Created in BioRender. Luan C. (2025) https://BioRender.com/wk50rtk).

Funding

No funding was received for conducting this study.

Conflict of interest

The author declares that he has no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Authors’ contributions

CHL is the sole author of the manuscript.

References

  1. Brown ZJ, Tsilimigras DI, Ruff SM, Mohseni A, Kamel IR, Cloyd JM, et al. Management of Hepatocellular Carcinoma: A Review. JAMA Surg 2023;158(4):410-420 View Article PubMed/NCBI
  2. Kim DB, Lee DK, Cheon C, Ribeiro RIMA, Kim B. Natural Products for Liver Cancer Treatment: From Traditional Medicine to Modern Drug Discovery. Nutrients 2022;14(20):4252 View Article PubMed/NCBI
  3. Wang D, Xiao M, Wan ZM, Lin X, Li QY, Zheng SS. Surgical treatment for recurrent hepatocellular carcinoma: Current status and challenges. World J Gastrointest Surg 2023;15(4):544-552 View Article PubMed/NCBI
  4. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74(3):229-263 View Article PubMed/NCBI
  5. Yang Y, Xiong L, Li M, Jiang P, Wang J, Li C. Advances in radiotherapy and immunity in hepatocellular carcinoma. J Transl Med 2023;21(1):526 View Article PubMed/NCBI
  6. Li HY, Jing YM, Shen X, Tang MY, Shen HH, Li XW, et al. Protein tyrosine phosphatase non-receptor II: A possible biomarker of poor prognosis and mediator of immune evasion in hepatocellular carcinoma. World J Gastrointest Oncol 2024;16(9):3913-3931 View Article PubMed/NCBI
  7. Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol 2022;77(6):1598-1606 View Article PubMed/NCBI
  8. Llovet JM, Pinyol R, Yarchoan M, Singal AG, Marron TU, Schwartz M, et al. Adjuvant and neoadjuvant immunotherapies in hepatocellular carcinoma. Nat Rev Clin Oncol 2024;21(4):294-311 View Article PubMed/NCBI
  9. Lindemann J, Yu J, Doyle MMB. New horizons in liver transplantation for hepatocellular carcinoma. S Afr J Surg 2024;62(2):8-12 PubMed/NCBI
  10. Feng F, Zhao Y. Hepatocellular Carcinoma: Prevention, Diagnosis, and Treatment. Med Princ Pract 2024;33(5):414-423 View Article PubMed/NCBI
  11. Vogel A, Saborowski A. Current strategies for the treatment of intermediate and advanced hepatocellular carcinoma. Cancer Treat Rev 2020;82:101946 View Article PubMed/NCBI
  12. Cheng S, Zhao H, Meng Y, Guo Y, Yao M, Xu X, et al. Impact of treatment-duration-ratio of traditional Chinese medicine on survival period of primary liver cancer -A real-world study. Heliyon 2022;8(12):e12358 View Article PubMed/NCBI
  13. Zhao R, Wang L, Liu Y, Shao M, Yang W, Fu Y, et al. The influence of adjunctive traditional Chinese medicine therapy on survival in primary liver cancer: a real-world study based on electronic medical records. Front Pharmacol 2023;14:1231933 View Article PubMed/NCBI
  14. Zheng S, Gu Y, Qi W, Wang W, Li X, Zao X, et al. Traditional Chinese Medicine for Liver Cancer Treatment: Network Pharmacology Research. Curr Top Med Chem 2025;25(26):3027-3041 View Article PubMed/NCBI
  15. Yang X, Feng Y, Liu Y, Ye X, Ji X, Sun L, et al. Fuzheng Jiedu Xiaoji formulation inhibits hepatocellular carcinoma progression in patients by targeting the AKT/CyclinD1/p21/p27 pathway. Phytomedicine 2021;87:153575 View Article PubMed/NCBI
  16. Kong Y, Zhu X, Zhang X, Li Z, Yin Y, Wang J, et al. Traditional Chinese medicine combined with radiofrequency ablation improves primary liver cancer outcomes: A systematic review with meta-analysis. Heliyon 2023;9(8):e18591 View Article PubMed/NCBI
  17. Zheng S, Xue T, Xue C, Li S, Zao X, Li X, et al. Regulatory mechanisms of signaling pathways in liver cancer treatment with traditional Chinese medicine. J Ethnopharmacol 2025;342:119386 View Article PubMed/NCBI
  18. Wang Z, Zhang Y, Zhang Q, Ao Q, Luo C, Wang B, et al. On the Core Prescriptions and Their Mechanisms of Traditional Chinese Medicine in Hepatitis B, Liver Cirrhosis, and Liver Cancer Treatment. J Oncol 2022;2022:5300523 View Article PubMed/NCBI
  19. Kim WY, Kim JM, Han SB, Lee SK, Kim ND, Park MK, et al. Steaming of ginseng at high temperature enhances biological activity. J Nat Prod 2000;63(12):1702-1704 View Article PubMed/NCBI
  20. Kwok HH, Guo GL, Lau JK, Cheng YK, Wang JR, Jiang ZH, et al. Stereoisomers ginsenosides-20(S)-Rg3 and -20(R)-Rg3 differentially induce angiogenesis through peroxisome proliferator-activated receptor-gamma. Biochem Pharmacol 2012;83(7):893-902 View Article PubMed/NCBI
  21. Cheong JH, Kim H, Hong MJ, Yang MH, Kim JW, Yoo H, et al. Stereoisomer-specific anticancer activities of ginsenoside Rg3 and Rh2 in HepG2 cells: disparity in cytotoxicity and autophagy-inducing effects due to 20(S)-epimers. Biol Pharm Bull 2015;38(1):102-108 PubMed/NCBI
  22. Jeong SM, Lee JH, Kim JH, Lee BH, Yoon IS, Lee JH, et al. Stereospecificity of ginsenoside Rg3 action on ion channels. Mol Cells 2004;18(3):383-389 PubMed/NCBI
  23. Chen J, Wang Z, Fu J, Cai Y, Cheng H, Cui X, et al. Ginsenoside compound K induces ferroptosis via the FOXO pathway in liver cancer cells. BMC Complement Med Ther 2024;24(1):174 View Article PubMed/NCBI
  24. Zhang J, Tong Y, Lu X, Dong F, Ma X, Yin S, et al. A derivant of ginsenoside CK and its inhibitory effect on hepatocellular carcinoma. Life Sci 2022;304:120698 View Article PubMed/NCBI
  25. Wu H, Qu L, Bai X, Zhu C, Liu Y, Duan Z, et al. Ginsenoside Rk1 induces autophagy-dependent apoptosis in hepatocellular carcinoma by AMPK/mTOR signaling pathway. Food Chem Toxicol 2024;186:114587 View Article PubMed/NCBI
  26. Wang W, Li K, Xiao W. The pharmacological role of Ginsenoside Rg3 in liver diseases: A review on molecular mechanisms. J Ginseng Res 2024;48(2):129-139 View Article PubMed/NCBI
  27. Zhou N, Mao F, Cheng S. Mechanism Research and Application for Ginsenosides in the Treatment of Hepatocellular Carcinoma. Biomed Res Int 2023;2023:7214037 View Article PubMed/NCBI
  28. Sun WT, Yang CLH, Or TCT, Luo D, Li JCB. Ginsenoside Rb1 from Panax notoginseng Suppressed TNF-α-Induced Matrix Metalloproteinase-9 via the Suppression of Double-Strand RNA-Dependent Protein Kinase (PKR)/NF-κB Pathway. Molecules 2022;27(22):8050 View Article PubMed/NCBI
  29. Yoon JH, Choi YJ, Lee SG. Ginsenoside Rh1 suppresses matrix metalloproteinase-1 expression through inhibition of activator protein-1 and mitogen-activated protein kinase signaling pathway in human hepatocellular carcinoma cells. Eur J Pharmacol 2012;679(1-3):24-33 View Article PubMed/NCBI
  30. Yoon JH, Choi YJ, Cha SW, Lee SG. Anti-metastatic effects of ginsenoside Rd via inactivation of MAPK signaling and induction of focal adhesion formation. Phytomedicine 2012;19(3-4):284-292 View Article PubMed/NCBI
  31. Shi Q, Shi X, Zuo G, Xiong W, Li H, Guo P, et al. Anticancer effect of 20(S)-ginsenoside Rh2 on HepG2 liver carcinoma cells: Activating GSK-3β and degrading β-catenin. Oncol Rep 2016;36(4):2059-2070 View Article PubMed/NCBI
  32. Ming Y, Chen Z, Chen L, Lin D, Tong Q, Zheng Z, et al. Ginsenoside compound K attenuates metastatic growth of hepatocellular carcinoma, which is associated with the translocation of nuclear factor-κB p65 and reduction of matrix metalloproteinase-2/9. Planta Med 2011;77(5):428-433 View Article PubMed/NCBI
  33. Ren Z, Chen X, Hong L, Zhao X, Cui G, Li A, et al. Nanoparticle Conjugation of Ginsenoside Rg3 Inhibits Hepatocellular Carcinoma Development and Metastasis. Small 2020;16(2):e1905233 View Article PubMed/NCBI
  34. Jiang JW, Chen XM, Chen XH, Zheng SS. Ginsenoside Rg3 inhibit hepatocellular carcinoma growth via intrinsic apoptotic pathway. World J Gastroenterol 2011;17(31):3605-3613 View Article PubMed/NCBI
  35. Lu H, Yin H, Qu L, Ma X, Fu R, Fan D. Ginsenoside Rk1 regulates glutamine metabolism in hepatocellular carcinoma through inhibition of the ERK/c-Myc pathway. Food Funct 2022;13(7):3793-3811 View Article PubMed/NCBI
  36. Zheng ZZ, Ming YL, Chen LH, Zheng GH, Liu SS, Chen QX. Compound K-induced apoptosis of human hepatocellular carcinoma MHCC97-H cells in vitro. Oncol Rep 2014;32(1):325-331 View Article PubMed/NCBI
  37. Guo XX, Guo Q, Li Y, Lee SK, Wei XN, Jin YH. Ginsenoside Rh2 induces human hepatoma cell apoptosisvia bax/bak triggered cytochrome C release and caspase-9/caspase-8 activation. Int J Mol Sci 2012;13(12):15523-15535 View Article PubMed/NCBI
  38. Park HM, Kim SJ, Kim JS, Kang HS. Reactive oxygen species mediated ginsenoside Rg3- and Rh2-induced apoptosis in hepatoma cells through mitochondrial signaling pathways. Food Chem Toxicol 2012;50(8):2736-2741 View Article PubMed/NCBI
  39. Qu L, Liu Y, Deng J, Ma X, Fan D. Ginsenoside Rk3 is a novel PI3K/AKT-targeting therapeutics agent that regulates autophagy and apoptosis in hepatocellular carcinoma. J Pharm Anal 2023;13(5):463-482 View Article PubMed/NCBI
  40. Jiang R, Luo S, Zhang M, Wang W, Zhuo S, Wu Y, et al. Ginsenoside Rh4 inhibits inflammation-related hepatocellular carcinoma progression by targeting HDAC4/IL-6/STAT3 signaling. Mol Genet Genomics 2023;298(6):1479-1492 View Article PubMed/NCBI
  41. Wei Q, Ren Y, Zheng X, Yang S, Lu T, Ji H, et al. Ginsenoside Rg3 and sorafenib combination therapy relieves the hepatocellular carcinomaprogression through regulating the HK2-mediated glycolysis and PI3K/Akt signaling pathway. Bioengineered 2022;13(5):13919-13928 View Article PubMed/NCBI
  42. Lee YS, Lee DG, Lee JY, Kim TR, Hong SS, Kwon SW, et al. A formulated red ginseng extract upregulates CHOP and increases TRAIL-mediated cytotoxicity in human hepatocellular carcinoma cells. Int J Oncol 2013;43(2):591-599 View Article PubMed/NCBI
  43. Lee JY, Jung KH, Morgan MJ, Kang YR, Lee HS, Koo GB, et al. Sensitization of TRAIL-induced cell death by 20(S)-ginsenoside Rg3 via CHOP-mediated DR5 upregulation in human hepatocellular carcinoma cells. Mol Cancer Ther 2013;12(3):274-285 View Article PubMed/NCBI
  44. Fayyaz S, Yaylim I, Turan S, Kanwal S, Farooqi AA. Hepatocellular carcinoma: targeting of oncogenic signaling networks in TRAIL resistant cancer cells. Mol Biol Rep 2014;41(10):6909-6917 View Article PubMed/NCBI
  45. Wu R, Ru Q, Chen L, Ma B, Li C. Stereospecificity of ginsenoside Rg3 in the promotion of cellular immunity in hepatoma H22-bearing mice. J Food Sci 2014;79(7):H1430-H1435 View Article PubMed/NCBI
  46. Wu L, Bai L, Dai W, Wu Y, Xi P, Zhang J, et al. Ginsenoside Rg3: A Review of its Anticancer Mechanisms and Potential Therapeutic Applications. Curr Top Med Chem 2024;24(10):869-884 View Article PubMed/NCBI
  47. Kim DG, Jung KH, Lee DG, Yoon JH, Choi KS, Kwon SW, et al. 20(S)-Ginsenoside Rg3 is a novel inhibitor of autophagy and sensitizes hepatocellular carcinoma to doxorubicin. Oncotarget 2014;5(12):4438-4451 View Article PubMed/NCBI
  48. Nakhjavani M, Smith E, Townsend AR, Price TJ, Hardingham JE. Anti-Angiogenic Properties of Ginsenoside Rg3. Molecules 2020;25(21):4905 View Article PubMed/NCBI
  49. Li Q, Li Y, Wang X, Fang X, He K, Guo X, et al. Co-treatment with ginsenoside Rh2 and betulinic acid synergistically induces apoptosis in human cancer cells in association with enhanced capsase-8 activation, bax translocation, and cytochrome c release. Mol Carcinog 2011;50(10):760-769 View Article PubMed/NCBI
  50. Xi S, Peng Y, Minuk GY, Shi M, Fu B, Yang J, et al. The combination effects of Shen-Ling-Bai-Zhu on promoting apoptosis of transplanted H22 hepatocellular carcinoma in mice receiving chemotherapy. J Ethnopharmacol 2016;190:1-12 View Article PubMed/NCBI
  51. Chen T, Li D, Fu YL, Hu W. Screening of QHF formula for effective ingredients from Chinese herbs and its anti-hepatic cell cancer effect in combination with chemotherapy. Chin Med J (Engl) 2008;121(4):363-368 PubMed/NCBI
  52. Zhou B, Yan Z, Liu R, Shi P, Qian S, Qu X, et al. Prospective Study of Transcatheter Arterial Chemoembolization (TACE) with Ginsenoside Rg3 versus TACE Alone for the Treatment of Patients with Advanced Hepatocellular Carcinoma. Radiology 2016;280(2):630-639 View Article PubMed/NCBI
  53. Pan W, Xue B, Yang C, Miao L, Zhou L, Chen Q, et al. Biopharmaceutical characters and bioavailability improving strategies of ginsenosides. Fitoterapia 2018;129:272-282 View Article PubMed/NCBI
  54. Llovet JM, Castet F, Heikenwalder M, Maini MK, Mazzaferro V, Pinato DJ, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 2022;19(3):151-172 View Article PubMed/NCBI
  55. Gao YL, Liu ZF, Li CM, Shen JY, Yin HX, Li GS. Subchronic toxicity studies with ginsenoside compound K delivered to dogs via intravenous administration. Food Chem Toxicol 2011;49(8):1857-1862 View Article PubMed/NCBI
  • Nature Cell and Science
  • 2958-695X

Ginsenosides as Multi-target Agents against Hepatocellular Carcinoma: Mechanisms and Therapeutic Prospects

Chang-Hai Luan
  • Reset Zoom
  • Download TIFF