Anticancer effects and potential mechanisms of ginsenoside Rh2 in various cancer types (Review)

  • Authors:
    • Haibo Zhang
    • Song Park
    • Hai Huang
    • Eungyung Kim
    • Junkoo Yi
    • Seong-Kyoon Choi
    • Zaeyoung Ryoo
    • Myoungok Kim
  • View Affiliations

  • Published online on: February 19, 2021     https://doi.org/10.3892/or.2021.7984
  • Article Number: 33
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ginsenoside Rh2 (G‑Rh2) is a natural bioactive product derived from Panax ginseng Meyer (P. ginseng). G‑Rh2 exhibits anticancer activity in various human cancer cell lines both in vitro and in vivo by modulating several signaling pathways, such as those of PDZ‑binding kinase/T‑LAK cell‑originated protein kinase, phosphatidylinositol 3‑kinase, protein kinase B, mammalian target of rapamycin, epidermal growth factor receptor, p53, and reactive oxygen species. Moreover, G‑Rh2 could effectively reverse drug resistance and enhance therapeutic effects in cancer therapy. This review summarizes the chemical properties, in vitro and in vivo anticancer activity, and underlying molecular mechanisms of G‑Rh2 to facilitate cancer chemoprevention studies.

Introduction

Cancer is the second leading cause of death globally, responsible for ~9.6 million deaths in 2018 (1). The main forms of cancer treatments are surgery, radiotherapy, chemotherapy and hormone therapy. Adjuvant chemotherapy following surgery has been proven to decrease recurrence and improve patient survival time (2,3). Natural products have been major sources for the active ingredients in numerous medicines. Compared with synthetic products, natural products generally have fewer toxic effects and are less expensive. Ginseng, derived from the rhizome and root of Panax ginseng Meyer (P. ginseng), is a popular herbal medicine that has been used in Asian countries (e.g., China, Japan and Korea) for thousands of years. It is well known for its disease-preventing and therapeutic effects.

Ginsenosides are the main active chemical constituents of P. ginseng. To date, >100 types of ginsenosides have been isolated and identified from P. ginseng (4). Among them, ginsenoside Rh2 (G-Rh2) exhibits various biological activities, such as improvement in learning and memory (5), promotion of immunity (6), and antioxidant (7), anti-inflammatory (811), antihyperglycemic (12) and antitumor (1321) effects. Increasing evidence shows that G-Rh2 exerts antitumor effects in a variety of cancer models, including human lung (2226), liver (27,28), gastric (29), colorectal (17,18,30,31), breast (3234), prostate (13,14,3537) and pancreatic (20) cancer, leukemia (3841) and ovarian cancer (4245). The functional mechanisms of G-Rh2 mainly include inducing apoptosis, arresting the cell cycle, inhibiting proliferation, angiogenesis and metastasis, and regulating the tumor microenvironment to promote immunity. Additionally, combining G-Rh2 with chemotherapy drugs can reverse drug resistance and enhance drug sensitivity in various cancer types. These topics will be described later in the review. Therefore, this review provides a systematic summary of the anticancer effects of G-Rh2 in order to facilitate further studies involving G-Rh2.

Chemical properties of G-Rh2

The ginsenosides are classified as 20(S)-protopanaxadiol compounds (including ginsenosides Rb1, Rb2, Rc, Rh2 and Rg3, among others) or 20(S)-protopanaxatriol compounds (including ginsenosides Re, Rg1 and Rg2, among others). G-Rh2 has been found to inhibit the growth of various human cancer cell types, such as lung cancer cells (2225), liver cancer cells (27,28) and colorectal cancer cells (17,30,31). The chemical formula for G-Rh2 is C36H62O8, and the molecular weight is 622.87; it is a white crystal with strong biological activity. The antitumor activity of ginsenosides is related to factors such as the type of ginsenoside, the substituents, and the number and configuration of sugars. Due to the different spatial structures at the C20 position, there are two stereoisomeric forms of G-Rh2: 20(S)-G-Rh2 and 20(R)-G-Rh2 (Fig. 1). In comparison with 20(R)-G-Rh2, 20(S)-G-Rh2 shows more potent anticancer activity among different cancer cells (35,46). One study reported that the half maximal inhibitory concentration values of 20(S)-G-Rh2 and 20(R)-G-Rh2 in A549 cells were 45.7 and 53.6 µM, respectively (46). Another study compared the effect of 20(S)-G-Rh2 and 20(R)-G-Rh2 on LNCaP, PC3 and DU145 cells. The results showed that 25 µM 20(S)-G-Rh2 inhibited LNCaP proliferation by 70%, PC3 cell proliferation by 40% and DU145 cell proliferation by 20%, while 25 µM 20(R)-G-Rh2 did not affect the proliferation of these cells (35). In addition, cytotoxic potency is generally in the descending order of protopanaxadiol, 20(S)-G-Rh2 and then 20(R)-G-Rh2, indicating structure-related activities in which the compound with less polar chemical structures possesses higher cytotoxic activity towards cancer cells (47). As much of the literature does not indicate whether 20(S)-G-Rh2 or 20(R)-G-Rh2 is used, the term G-Rh2 in this review includes both of these configurations.

Anticancer effects and mechanisms of G-Rh2 in in vitro studies

Previous studies have demonstrated that G-Rh2 exerts significant anticancer activities through multiple molecular mechanisms. The mechanisms are mainly related to cell cycle arrest, apoptosis, proliferation, invasion, metastasis, angiogenesis, autophagy and immunity, which are summarized in Table I and Fig. 2. Although the anticancer activity of G-Rh2 has been widely investigated, the exact molecular mechanisms remain unclear. Based on existing research, the possible mechanisms of action of G-Rh2 are described in this review.

Table I.

Anticancer effects and mechanisms of ginsenoside-Rh2 in in vitro studies.

Table I.

Anticancer effects and mechanisms of ginsenoside-Rh2 in in vitro studies.

Cancer typeCell typeCellular effectsMechanisms(Refs.)
Lung cancerH1299, A549, Lu-99, EBC-1, H460, CH27Proliferation, apoptosis, cell cycle arrestp21↓, CDK6↓, cyclin D1↓, cyclin E↓, ATF4↑, CHOP↑, caspase-4↑, mir-491↑, pRb2↑, DR4↑(22,2426)
Prostate cancerPC3, DU145, LNCaPProliferation, apoptosis, invasionp-STAT3↓, cyclin D1↓, cyclinB1↓, MMP2↓, MMP9↓, CDKN1A↑, PPAR-δ↑, miR-4295↓, pSmad2↑, p27↑(14,3537)
Colorectal cancerHCT116, SW480, LoVo, SW620, HCT-8Proliferation, apoptosis, invasion, cell cycle arrestp-ERK↓, p-p90RSK↓, Bcl2↓, Bcl-xl↓, P-pg↓, cyclin D1↓, CDK2↓, p-Rb↓, Bcl-2↓, N-cadherin↓, vimentin↓, MMP9↓, Smad4↑, caspase-3↑, p-AMPK↑, NF-κB↑, ROS↑, Bad↑, Bax↑, cleaved caspase-3↑, p-IκB-α↑, E-cadherin↑(1719,30,31,78,80)
Breast cancerMCF7, MDA-MB-231Proliferation, apoptosis, cell cycle arrestCASP1↓, INSL5↓, OR52A1↓, CDK2↓, CDK4↓, CDK6↓, cyclin A↓, cyclin D1↓, cyclin E↓, CLINT1↑, ST3GAL4↑, C1orf198↑, p21↑(3234)
Gastric cancerSGC-7901Proliferation, apoptosis, cell cycle arrestBcl2↓, Bax↑(29)
Liver cancerHepG2, Huh7, SMMC-7721, Hep3BProliferation, apoptosis, migrationMCL1↓, Nrf2↓, NF-κB↓, annexin A2↓, EZH2↓, H3K27me3↓, EGFR↓, cyclin D1↓, Bcl2↓, MMP3↓, p-AMPK↑, p-p38↑, p-JNK↑, p-ERK↑(19,27,28,84,85)
Cervical cancerHeLaProliferation, apoptosisp-Akt↓, p-GSK-3β↓, N-cadherin↓, vimentin↓, ZEB1↓, Snail-1↓, E-cadherin↑, Fas↑, TNFR1↑, cleaved caspase-8↑, cleaved caspase-9↑, cleaved PARP↑, TNF-α↑, p53↑(15,56)
Endometrial cancerHEC1A, IshikawaProliferation, apoptosis, invasion, migrationvimentin↓, TGF-β↓, Snail↓, cleaved PARP↑, cleaved caspase-3↑, E-cadherin↑(65)
Skin squamous cell carcinomaA431Proliferation, autophagyβ-catenin↓, Beclin-1↑, Atg7↑, LC3-I↑, LC3-II↑(76)
Pancreatic cancerBxpc-3Proliferation, apoptosis, migration, cell cycle arrestcaspase-9↑, Bcl-2↓, survivin↓, cyclin D1↓, MMP-2↓, MMP-9↓, Bax↑, cleaved caspase-3↑(20)
Ovarian cancerHRA, KK, KF, KFr, SKOV3, SKOv3ip, HeyProliferation, apoptosis, cell cycle arrestBcl-2↓, cleaved PARP↑, cleaved caspase-3↑(42,44,45)
LeukemiaU937, K562, Jurkat, HL-60, Kasumi-1, KG1-αProliferation, apoptosis, autophagy, cell cycle arrestβ-catenin↓, TCF4↓, cyclin D1↓, NF-κB↓, Bcl-2↓, LC3-I↓, CDK4↓, HDAC1↓, HDAC2↓, HDAC6↓, p-ERK↓, caspase-3↑, PARP↓, LC3B↑, p62↓, cleaved caspase-3↑, cleaved caspase-9↑, cytochrome c↑, Bax↑, Beclin-1 ↑, LC3-II↑, p16↑, p21↑, p-p38↑(3841,57,61,74)
GliomaA172, U87MG, U251Proliferation, cell cycle arrest, apoptosisp-Akt↓, Akt↓, p-EGFR↓, p-mTOR↓, CD31↓, MMP13↓, CDK4↓, cyclin E↓, cyclin D↓, CDK2↓, p27↑(21,93,95)
NeuroblastomaSK-N-BE(2)Apoptosiscaspase-1↑, caspase-3↑, Bax↑(91)
RetinoblastomaY79, RBL-13Proliferation, apoptosis, autophagyp-PI3K↓, p-Akt↓, p-mTOR↓, p62↓, Bcl2↓, cyclin D1↓, Beclin-1↑, ATG7↑, p53↑, Bax↑, cleaved caspase-3↑, cleaved caspase-9↑(77)
Oral cancerYD10B, Ca9-22, KBProliferation, apoptosis, cell cycle arrest, migration, invasioncyclin D1↓, cyclin E1↓, vimentin↓, N-cadherin↓, MMP-2↓, VEGF↓, p-Src↓, p-B-Raf↓, p-ERK1/2↓, cleaved PARP↑, cleaved caspase-3↑, p53↑, E-cadherin↑(62,67)

[i] ATF4, activating transcription factor 4; CHOP, CCAAT/enhancer-binding protein homologous protein; CDK, cyclin-dependent kinase; DR4, death receptor 4; PPARδ, peroxisome proliferator-activated receptor δ; STAT3, signal transducer and activator of transcription 3; CDKN1A, cyclin-dependent kinase inhibitor 1A; MMP, matrix metallopeptidase; ERK, extracellular regulated protein kinases; NF-κB, nuclear factor-kB; ROS, reactive oxygen species; Bad, BCL2-associated agonist of cell death; Bax, BCL2-associated X protein; P-gp, permeability glycoprotein; AMPK, AMP-activated protein kinase; CASP1, caspase-1; INSL5, insulin-like peptide 5; OR52A1, olfactory receptor family 52 subfamily A member 1; CLINT1, clathrin interactor 1; ST3GAL4, ST3 β-galactoside α-2,3-sialyltransferase 4; C1orf198, chromosome 1 open reading frame 198; Nrf2, nuclear factor erythroid 2-related factor 2; JNK, c-Jun N-terminal kinase; ERK, extracellular-signal-regulated kinase; EZH2, enhancer of zeste homolog 2; EGFR, epidermal growth factor receptor; Akt, protein kinase B; GSK-3β, glycogen synthase kinase 3β; ZEB1, zinc finger E-box-binding homeobox 1; TNFR1, tumor necrosis factor receptor 1; PARP, poly (ADP-ribose) polymerase; Atg7, autophagy-related 7; LC3, microtubule-associated protein light chain 3; TCF-4, transcription factor 4; HDAC, histone deacetylases; mTOR, mammalian target of rapamycin; CD31, cluster of differentiation 31; PI3K, phosphoinositide 3-kinase; VEGF, vascular endothelial growth factor.

Induction of cell cycle arrest

The cell cycle is a controlled process involved in the growth, differentiation and proliferation of eukaryotic cells (48). Cells that undergo cell cycle arrest lose their ability to replicate and divide. Cyclin-dependent kinase (CDK) inhibitors are crucial for controlling the cell cycle and cell proliferation (49). The CDK inhibitor p21 plays a key role in the G1 phase cell cycle checkpoint (50). G-Rh2 was reported to induce cell cycle G1 phase arrest in MCF-7 cells by increasing p21 levels and decreasing CDK2 and cyclin E-dependent kinase activities (32). In human glioma A172 cells, G-Rh2 induced cell cycle G1 phase arrest by downregulating CDK4 and cyclin E (21). In human lung cancer A549 cells, G-Rh2 induced cell cycle G1 phase arrest by significantly reducing the expression of CDK4 and cyclin D1 (51). Furthermore, G-Rh2 induced cell cycle G1 phase arrest in HL-60 and U937 cells by downregulating the expression of CDK4, CDK6, cyclin D1, cyclin D2, cyclin D3 and cyclin E. G-Rh2-mediated G1 phase arrest and differentiation are closely linked to the regulation of TGF-β production in human leukemia cells (52).

Induction of apoptosis

Apoptosis (programmed cell death) plays an important role in animal development and adult life by eliminating damaged cells (53). The induction of apoptosis in cancer cells can inhibit tumor growth (54). Treatment of colorectal cancer cells with G-Rh2 has been proven to activate the p53 pathway, increasing the expression of the proapoptotic regulator Bax and decreasing the expression of the antiapoptotic regulator Bcl-2 (30). The extrinsic pathway and the intrinsic pathway are two core apoptotic pathways in mammalian cells (55). As shown in Table I, G-Rh2 can induce apoptosis via these two pathways. It was previously shown that G-Rh2 induced apoptosis by downregulating Bcl-2 and survivin and upregulating Bax, cleaved caspase-3 and cleaved caspase-9 in human pancreatic cancer Bxpc-3 cells (20). G-Rh2-triggered intrinsic apoptosis was related to the induced translocation of cytosolic Bak and Bax to the mitochondria, cytochrome c release and caspase-9 activation in HeLa and SW480 cells (56). G-Rh2 may induce apoptosis of Kasumi-1 and U937 leukemia cells via microRNA-21-modulated suppression of Bcl-2 (57). In addition, cotreatment with G-Rh2 and betulinic acid could induce apoptosis of HeLa, A549 and HepG2 cancer cells by enhancing caspase-8 expression, cytochrome c release and Bax translocation (58).

Inhibition of proliferation

Abnormal regulation of cell proliferation is the key cause of cancer development and progression (59). Both activation of oncogenes and the inactivation of tumor suppressor genes can promote the proliferation of cancer cells. Protein kinase B (Akt) is one of the well-known proto-oncogenes, and activated Akt promotes cancer cell proliferation and survival (60). G-Rh2 has been shown to inhibit HeLa and A172 cell proliferation by suppressing the Akt pathway (15,21). Another study revealed that G-Rh2 inhibited the proliferation of HCT116 colon cancer cells by targeting PDZ-binding kinase/T-LAK cell-originated protein kinase and downregulating the expression of p-ERK1/2 and p-histone H3 (17). G-Rh2 inhibits the proliferation of K562 and KG1-α cells by suppressing the expression and activity of HDAC1, HDAC2, and HDCA6, increasing histone H3 acetylation and regulating MAPK/JNK signaling pathways (61). Additionally, G-Rh2 could inhibit cancer cell proliferation by suppressing endoplasmic reticulum (ER) stress (22), by inhibiting the Src/ERK signaling pathway (62) and by targeting microRNA-128 (63).

Inhibition of invasion and metastasis

Metastasis is the main cause of cancer treatment failure and recurrence. During metastasis, the cells become highly plastic or adaptive, similar to stem cells. This change is called epithelial-mesenchymal transition (EMT) (64). G-Rh2 could effectively inhibit tumor metastasis by suppressing EMT. G-Rh2 has been found to inhibit migration and invasion by increasing E-cadherin and suppressing vimentin expression in endometrial cancer cells (65) and oral cancer cells (62). In addition, matrix metalloproteinases (MMPs) play an important role in tumor metastasis, where they can degrade the extracellular matrix and basement membrane (66). G-Rh2 was previously found to effectively inhibit Bxpc-3 cell migration and invasion by downregulating MMP-2 and MMP-9 (20). Moreover, G-Rh2 was reported to significantly reduce the protein levels of VEGF, MMP-2 and MMP-9 in co-cultured lung cancer cells (23) and oral cancer cells (67). Therefore, G-Rh2 may play an inhibitory role in the process of cancer metastasis.

Anti-angiogenesis

Angiogenesis plays an important role in tumor growth and metastasis by providing the necessary nutrients and oxygen (68). TGF-β is a potent angiogenesis inducer in vivo (69). Anti-angiogenic treatment for tumors is considered a promising therapeutic strategy (70). G-Rh2 was reported to inhibit prostate cancer growth by impeding angiogenesis via decreasing the expression of CD31, VEGF, platelet-derived growth factor and CNNM1 in cancer cells (13). Furthermore, G-Rh2 could affect tumor angiogenesis by downregulating JAM expression in tumors (71).

Induction of autophagy

Autophagy is a catabolic process that degrades cytoplasmic constituents and organelles in lysosomes (72). There is increasing evidence that autophagy signaling is closely related to oncogenic signaling. Autophagy may be one of the effective ways to prevent the formation and progression of tumors (72). Selective targeting of autophagy for cancer treatment has attracted considerable attention (73). The autophagy-related genes ATG5, ATG7, LC3B and beclin-1 were upregulated after treatment with G-Rh2 in U937 and K562 cells (74). The formation of autophagosomes involves the conversion from cytosolic LC3-I to the autophagosome-associating form of LC3-II (75). Treatment of U937 and K562 cells with G-Rh2 was found to induce the conversion from LC3-I to LC3-II and downregulate the protein level of p62 (74). G-Rh2 treatment increased autophagy through upregulating autophagy-related proteins Beclin, Atg7 and the ratio of LC3-II to LC3-I in A431 cells (76). Another study reported that G-Rh2 could promote cell autophagy in human retinoblastoma cell lines Y79 and RBL-13 by inactivating the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway (77).

Reverse drug resistance

Drug resistance is the leading cause of failure of cancer chemotherapy. Several studies have suggested that G-Rh2 has a role in reversing drug resistance and improving treatment efficacy. G-Rh2 has been found to reverse oxaliplatin resistance in colon cancer cells through decreasing the expression of P-glycoprotein (78), and to reverse drug resistance in Adriamycin-resistant human breast cancer MCF-7 cells (79). Another study reported that G-Rh2 effectively reversed 5-flurouracil resistance in colorectal cancer cells by regulating MDR1, MRP1, LRP and GST gene expression (80). In addition, G-Rh2 could enhance the antitumor effects of SMI-4a by increasing autophagy in melanoma cells (81) and could enhance the anticancer effects of cisplatin and prolong the survival time of nude mice (43). These reports suggest that G-Rh2 has an adjunctive effect in cancer chemotherapy and can delay the occurrence of drug resistance.

Promotion of immunity

It is well known that certain cancer treatments can temporarily weaken the immune system, and that enhancing the immune response could play an important role in cancer treatment. Ginseng has a long history of improving the immunity of patients in Asia. A growing number of studies indicate that G-Rh2 can improve immunity. One study reported that G-Rh2 enhanced the antitumor immunological response by triggering CD4+ and CD8a+ T-lymphocyte infiltration in B16-F10 melanoma cells derived from xenograft tumor tissues, as well as enhancing the cytotoxicity in spleen lymphocytes (82). Another study found that G-Rh2 could downregulate CASP1, INSL5 and OR52A1, and upregulate CLINT1, ST3GAL4 and C1orf198 expression in MCF-7 cells, indicating that G-Rh2 induces epigenetic methylation changes in genes involved in the immune response and tumorigenesis, thereby contributing to enhanced immunogenicity and inhibiting the growth of cancer cells (33). G-Rh2 suppressed T-cell acute lymphoblastic leukemia (T-ALL) by blocking the PI3K/Akt/mTOR signaling pathway and enhanced immunity in the spleen by downregulating IL-4, IL-6, IL-10, CD3 and CD45, and upregulating IL-2 and INF-γ, and increased the number of natural killer cells (83). These studies indicate that G-Rh2 has the ability to enhance the immune response, which may play a role in the prevention and treatment of cancer.

Anticancer effects and mechanisms of G-Rh2 in in vivo studies

G-Rh2 exhibits anticancer effects in a number of animal models (17,22,36,43,61,83,84). Table II summarizes the anticancer effects of G-Rh2 in in vivo studies. In colon cancer xenograft mouse models, 10 and 50 mg/kg G-Rh2 three times a week via intraperitoneal injection significantly suppressed HCT116 ×enograft tumor growth, and further research indicated that G-Rh2 could downregulate p-ERK1/2 and p-H3 expression by inhibiting TOPK activity in vivo (17). In lung cancer H1299 cell xenograft mouse models, G-Rh2 significantly inhibited lung cancer cell growth by inducing reactive oxygen species (ROS)-mediated ER stress in vivo (22). In leukemia K562 cell xenograft mouse models, treatment with G-Rh2 (20 mg/kg once a day for 3 weeks) significantly inhibited tumor growth in vivo (61). G-Rh2 was found to suppress HepG2 cell xenograft tumor growth (84,85), and the anticancer mechanism of G-Rh2 in HepG2 cells was related to downregulating β-catenin through the activation of glycogen synthase kinase-3β (85). In addition, G-Rh2 has poor oral absorption and low bioavailability. In vivo metabolism and pharmacokinetic studies indicated that oxygenation and deglycosylation were the major metabolic pathways of G-Rh2 (86); the deglycosylation of G-Rh2 led to formation of protopanaxadiol and the oxygenation of G-Rh2, and protopanaxadiol produced two monooxygenated metabolites (86). Another study reported that (24R)-pseudo-ginsenoside HQ and (24S)-pseudo-ginsenoside HQ are the main metabolites of 20(S)-G-Rh2 in vivo, with both of them showing antitumor activity through caspase and VEGF signaling pathways in H22-tumor bearing mice (87). After oral dosing, G-Rh2 was found to be distributed mainly to the liver and gastrointestinal tissues in rats. The bioavailability of G-Rh2 is ~5% in rats and 16% in dogs (88). Thus, modification of the chemical structure of G-Rh2 to increase bioavailability and enhance its pharmacological activity is also one of the current research directions.

Table II.

Anticancer effects and mechanisms of ginsenoside-Rh2 in in vivo studies.

Table II.

Anticancer effects and mechanisms of ginsenoside-Rh2 in in vivo studies.

Animal modelDoseRouteEffects(Refs.)
Lung cancer xenograft mouse model (H1299 cells)20 mg/kg/day IntraperitoneallyReduced tumor size and tumor weight; increased ATF4, CHOP, and caspase-4 levels(22)
Prostate cancer xenograft mouse model (PC3 cells)1 mg/kg, 2 times a week for 4 weeksTail-vein injectionInhibited tumor growth(36)
Colon cancer xenograft mouse model (HCT116 cells)10,50 mg/kg; 3 times/week for 29 days IntraperitoneallyReduced tumor size and tumor weight; decreased p-ERK and p-H3 levels(17)
Ovarian cancer xenograft mouse model (HRA cells)0.4 to 1.6 mg/kg/day, 21 or 28 daysGavageReduced tumor size and prolong survival time(42)
Leukemia xenograft mouse model (K562)20 mg/kg/day for 3 weeksGavageReduced tumor size and tumor weight; decreased HDAC1, HDAC2, and HDAC6 levels(61)
Liver cancer xenograft mouse model (HepG2)5 mg/kg for 5 weeks IntraperitoneallyReduced tumor size; decreased EZH2 and H3K27me3 levels(84)
Liver cancer xenograft mouse model (HepG2)20 mg/kg/day for 20 daysGavageDecreased tumor weight by downregulating β-catenin via activation of GSK-3β(85)
Liver cancer xenograft mouse model (SMMC-7721)1 mg/kg, 2 times a week for 1 monthTail-vein injectionDecreased tumor volume and weight by targeting EGFR through upregulating miR-491(28)
T-cell acute lymphoblastic leukemia (Jurkat cells)40 mg/kg/day for 3 weeksGavageAlleviated spleen infiltration by blocking the PI3K/Akt/mTOR signaling pathway, and enhanced immunity(83)

[i] ATF4, activating transcription factor 4; CHOP, CCAAT/enhancer-binding protein homologous protein; ERK, extracellular-signal-regulated kinase; p-H3, p-histone H3; HDAC, histone deacetylases; EZH2, enhancer of zeste homolog 2; GSK-3β, glycogen synthase kinase 3β; EGFR, epidermal growth factor receptor; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; mTOR, mammalian target of rapamycin.

Signaling pathway of G-Rh2 in cancer

According to current studies, the anticancer signaling pathway of G-Rh2 remains unclear. It was reported that G-Rh2 suppresses growth of oral squamous cell carcinoma cells by decreasing ROS, MMP-2 and VEGF (67). G-Rh2 inhibits glioma cell growth by targeting microRNA-128 (63) and inhibiting epidermal growth factor receptor (89). G-Rh2 induces leukemia cell differentiation and cell cycle arrest by upregulating TGF-β expression (52) and inducing apoptosis by inducing the release of mitochondrial cytochrome c and the activation of caspase-3 and −9 (90). In neuroblastoma cells, G-Rh2 induces apoptosis via activation of caspase-1 and −3 and upregulation of Bax (91). G-Rh2 suppresses cancer cell migration and invasion by downregulating the expression levels of MMP3 (92) and MMP13 (93), and by regulating CDKN2A-2B gene cluster transcription (84). G-Rh2 exerts anticancer activity in T-cell acute lymphoblastic leukemia cells (94), glioblastoma multiforme cells (95) and osteosarcoma cells (96) by suppressing the PI3K/Akt/mTOR signaling pathway. In addition, G-Rh2-induced DNA damage and autophagy in vestibular schwannoma is dependent on LAMP2 transcriptional suppression (97), and it improves the cisplatin effect in lung adenocarcinoma A549 cells by repressing superoxide generation and PD-L1 expression (98). Based on these reports, we hypothesize that PI3K/Akt/mTOR could be an important signaling pathway for G-Rh2 to exert its activity, which provides some context for further research on G-Rh2. The potential signaling pathways of G-Rh2 in cancer are demonstrated in Fig. 3.

Conclusions

As one of the main active components of ginseng, G-Rh2 has a wide range of pharmacological effects and plays a therapeutic role in numerous diseases. A number of studies have demonstrated that G-Rh2 exerts excellent anticancer activity in vitro and in vivo. G-Rh2 exerts its anticancer activity by inducing apoptosis, autophagy, cell cycle arrest and immunity, as well as by inhibiting proliferation, invasion, metastasis and angiogenesis. In addition, G-Rh2 in combination with specific anticancer drugs can overcome drug resistance and enhance the immune response. In summary, G-Rh2 exerts anticancer effects in vitro and in vivo, and is a promising agent for cancer prevention and treatment.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

HBZ, SP, HH, EK and JY collected the literature and designed the study. HBZ drafted the manuscript. SKC, ZYR and MK revised the manuscript. ZYR and MK confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare hat they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Shahab D, Gabriel E, Attwood K, Ma WW, Francescutti V, Nurkin S and Boland PM: Adjuvant chemotherapy is associated with improved overall survival in locally advanced rectal cancer after achievement of a pathologic complete response to chemoradiation. Clin Colorectal Cancer. 16:300–307. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Fujita K, Taneishi K, Inamoto T, Ishizuya Y, Takada S, Tsujihata M, Tanigawa G, Minato N, Nakazawa S, Takada T, et al: Adjuvant chemotherapy improves survival of patients with high-risk upper urinary tract urothelial carcinoma: A propensity score-matched analysis. BMC Urol. 17:1102017. View Article : Google Scholar : PubMed/NCBI

4 

Lu JM, Yao Q and Chen C: Ginseng compounds: An update on their molecular mechanisms and medical applications. Curr Vasc Pharmacol. 7:293–302. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Hou J, Xue J, Lee M, Liu L, Zhang D, Sun M, Zheng Y and Sung C: Ginsenoside Rh2 improves learning and memory in mice. J Med Food. 16:772–776. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Qian Y, Huang R, Li S, Xie R, Qian B, Zhang Z, Li L, Wang B, Tian C, Yang J, et al: Ginsenoside Rh2 reverses cyclophosphamide-induced immune deficiency by regulating fatty acid metabolism. J Leukoc Biol. 106:1089–1100. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Choi WY, Lim HW and Lim CJ: Anti-inflammatory, antioxidative and matrix metalloproteinase inhibitory properties of 20(R)-ginsenoside Rh2 in cultured macrophages and keratinocytes. J Pharm Pharmacol. 65:310–316. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Zhang XP, Li KR, Yu Q, Yao MD, Ge HM, Li XM, Jiang Q, Yao J and Cao C: Ginsenoside Rh2 inhibits vascular endothelial growth factor-induced corneal neovascularization. FASEB J. 32:3782–3791. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Zhou J, Gao Y, Yi X and Ding Y: Ginsenoside Rh2 suppresses neovascularization in xenograft psoriasis model. Cell Physiol Biochem. 36:980–987. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Choi K, Kim M, Ryu J and Choi C: Ginsenosides compound K and Rh(2) inhibit tumor necrosis factor-alpha-induced activation of the NF-kappaB and JNK pathways in human astroglial cells. Neurosci Lett. 421:37–41. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Bae EA, Kim EJ, Park JS, Kim HS, Ryu JH and Kim DH: Ginsenosides Rg3 and Rh2 inhibit the activation of AP-1 and protein kinase A pathway in lipopolysaccharide/interferon-gamma-stimulated BV-2 microglial cells. Planta Med. 72:627–633. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Wang Y, Wang H, Liu Y, Li C, Qi P and Bao J: Antihyperglycemic effect of ginsenoside Rh2 by inducing islet β-cell regeneration in mice. Horm Metab Res. 44:33–40. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Huang YQ, Huang HX, Han ZD, Li W, Mai ZP and Yuan RQ: Ginsenoside Rh2 Inhibits Angiogenesis in Prostate Cancer by Targeting CNNM1. J Nanosci Nanotechnol. 19:1942–1950. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Tong-Lin Wu T, Tong YC, Chen IH, Niu HS, Li Y and Cheng JT: Induction of apoptosis in prostate cancer by ginsenoside Rh2. Oncotarget. 9:11109–11118. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Shi X, Yang J and Wei G: Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through the Akt/GSK3β signaling pathway in human cervical cancer cells. Mol Med Rep. 17:4811–4816. 2018.PubMed/NCBI

16 

Wang YS, Lin Y, Li H, Li Y, Song Z and Jin YH: The identification of molecular target of (20S) ginsenoside Rh2 for its anti-cancer activity. Sci Rep. 7:124082017. View Article : Google Scholar : PubMed/NCBI

17 

Yang J, Yuan D, Xing T, Su H, Zhang S, Wen J, Bai Q and Dang D: Ginsenoside Rh2 inhibiting HCT116 colon cancer cell proliferation through blocking PDZ-binding kinase/T-LAK cell-originated protein kinase. J Ginseng Res. 40:400–408. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Han S, Jeong AJ, Yang H, Bin Kang K, Lee H, Yi EH, Kim BH, Cho CH, Chung JW, Sung SH and Ye SK: Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through targeting IL-6-induced JAK2/STAT3 pathway in human colorectal cancer cells. J Ethnopharmacol. 194:83–90. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Kim MJ, Yun H, Kim DH, Kang I, Choe W, Kim SS and Ha J: AMP-activated protein kinase determines apoptotic sensitivity of cancer cells to ginsenoside-Rh2. J Ginseng Res. 38:16–21. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Tang XP, Tang GD, Fang CY, Liang ZH and Zhang LY: Effects of ginsenoside Rh2 on growth and migration of pancreatic cancer cells. World J Gastroenterol. 19:1582–1592. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Li KF, Kang CM, Yin XF, Li HX, Chen ZY, Li Y, Zhang Q and Qiu YR: Ginsenoside Rh2 inhibits human A172 glioma cell proliferation and induces cell cycle arrest status via modulating Akt signaling pathway. Mol Med Rep. 17:3062–3068. 2018.PubMed/NCBI

22 

Ge G, Yan Y and Cai H: Ginsenoside Rh2 inhibited proliferation by inducing ROS mediated ER stress dependent apoptosis in lung cancer cells. Biol Pharma Bull. 40:2117–2124. 2017. View Article : Google Scholar

23 

Li H, Huang N, Zhu W, Wu J, Yang X, Teng W, Tian J, Fang Z, Luo Y, Chen M and Li Y: Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2. BMC Cancer. 18:5792018. View Article : Google Scholar : PubMed/NCBI

24 

An IS, An S, Kwon KJ, Kim YJ and Bae S: Ginsenoside Rh2 mediates changes in the microRNA expression profile of human non-small cell lung cancer A549 cells. Oncol Rep. 29:523–528. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Chen Y, Zhang Y, Song W, Zhang Y, Dong X and Tan M: Ginsenoside Rh2 inhibits migration of lung cancer cells under hypoxia via mir-491. Anticancer Agents Med Chem. 19:1633–1641. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Cheng CC, Yang SM, Huang CY, Chen JC, Chang WM and Hsu SL: Molecular mechanisms of ginsenoside Rh2-mediated G1 growth arrest and apoptosis in human lung adenocarcinoma A549 cells. Cancer Chemother Pharmacol. 55:531–540. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Chen W, Chu S, Li H and Qiu Y: MicroRNA-146a-5p enhances ginsenoside Rh2-induced anti-proliferation and the apoptosis of the human liver cancer cell line HepG2. Oncol Lett. 16:5367–5374. 2018.PubMed/NCBI

28 

Chen W and Qiu Y: Ginsenoside Rh2 Targets EGFR by up-regulation of miR-491 to enhance anti-tumor activity in hepatitis B virus-related hepatocellular carcinoma. Cell Biochem Biophys. 72:325–331. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Qian J, Li J, Jia JG, Jin X, Yu DJ, Guo CX, Xie B and Qian LY: Ginsenoside-Rh2 inhibits proliferation and induces apoptosis of human gastric cancer SGC-7901 side population cells. Asian Pac J Cancer Prev. 17:1817–1821. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Li B, Zhao J, Wang CZ, Searle J, He TC, Yuan CS and Du W: Ginsenoside Rh2 induces apoptosis and paraptosis-like cell death in colorectal cancer cells through activation of p53. Cancer Lett. 301:185–192. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Zhu C, Liu F, Qian W, Zhang T and Li F: Combined effect of sodium selenite and Ginsenoside Rh2 on hct116 human colorectal carcinoma cells. Arch Iran Med. 19:23–29. 2016.PubMed/NCBI

32 

Oh M, Choi YH, Choi S, Chung H, Kim K, Kim SI, Kim DK and Kim ND: Anti-proliferating effects of ginsenoside Rh2 on MCF-7 human breast cancer cells. Int J Oncol. 14:869–875. 1999.PubMed/NCBI

33 

Lee H, Lee S, Jeong D and Kim SJ: Ginsenoside Rh2 epigenetically regulates cell-mediated immune pathway to inhibit proliferation of MCF-7 breast cancer cells. J Ginseng Res. 42:455–462. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Choi S, Kim TW and Singh SV: Ginsenoside Rh2-mediated G1 phase cell cycle arrest in human breast cancer cells is caused by p15 Ink4B and p27 Kip1-dependent inhibition of cyclin-dependent kinases. Pharm Res. 26:2280–2288. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Liu J, Shimizu K, Yu H, Zhang C, Jin F and Kondo R: Stereospecificity of hydroxyl group at C-20 in antiproliferative action of ginsenoside Rh2 on prostate cancer cells. Fitoterapia. 81:902–905. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Zhang Q, Hong B, Wu S and Niu T: Inhibition of prostatic cancer growth by ginsenoside Rh2. Tumour Biol. 36:2377–2381. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Gao Q and Zheng J: Ginsenoside Rh2 inhibits prostate cancer cell growth through suppression of microRNA-4295 that activates CDKN1A. Cell Prolif. 51:e124382018. View Article : Google Scholar : PubMed/NCBI

38 

Xia T, Wang YN, Zhou CX, Wu LM, Liu Y, Zeng QH, Zhang XL, Yao JH, Wang M and Fang JP: Ginsenoside Rh2 and Rg3 inhibit cell proliferation and induce apoptosis by increasing mitochondrial reactive oxygen species in human leukemia Jurkat cells. Mol Med Rep. 15:3591–3598. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Xia T, Wang J, Wang Y, Wang Y, Cai J, Wang M, Chen Q, Song J, Yu Z, Huang W and Fang J: Inhibition of autophagy potentiates anticancer property of 20(S)-ginsenoside Rh2 by promoting mitochondria-dependent apoptosis in human acute lymphoblastic leukaemia cells. Oncotarget. 7:27336–27349. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Huang J, Peng K, Wang L, Wen B, Zhou L, Luo T, Su M, Li J and Luo Z: Ginsenoside Rh2 inhibits proliferation and induces apoptosis in human leukemia cells via TNF-alpha signaling pathway. Acta Biochim Biophys Sin (Shanghai). 48:750–755. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Chen Y, Liu ZH, Xia J, Li XP, Li KQ, Xiong W, Li J and Chen DL: 20(S)-ginsenoside Rh2 inhibits the proliferation and induces the apoptosis of KG-1a cells through the Wnt/β-catenin signaling pathway. Oncol Rep. 36:137–146. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Nakata H, Kikuchi Y, Tode T, Hirata J, Kita T, Ishii K, Kudoh K, Nagata I and Shinomiya N: Inhibitory effects of ginsenoside Rh2 on tumor growth in nude mice bearing human ovarian cancer cells. Jpn J Cancer Res. 89:733–740. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Tode T, Kikuchi Y, Kita T, Hirata J, Imaizumi E and Nagata I: Inhibitory effects by oral administration of ginsenoside Rh2 on the growth of human ovarian cancer cells in nude mice. J Cancer Res Clin Oncol. 120:24–26. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Li N, Lin Z, Chen W, Zheng Y, Ming Y, Zheng Z, Huang W, Chen L, Xiao J and Lin H: Corilagin from longan seed: Identification, quantification, and synergistic cytotoxicity on SKOv3ip and hey cells with ginsenoside Rh2 and 5-fluorouracil. Food Chem Toxicol. 119:133–140. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Kim JH and Choi JS: Effect of ginsenoside Rh-2 via activation of caspase-3 and Bcl-2-insensitive pathway in ovarian cancer cells. Physiol Res. 65:1031–1037. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Zhang C, Yu H and Hou J: Effects of 20 (S)-ginsenoside Rh2 and 20 (R)-ginsenoside Rh2 on proliferation and apoptosis of human lung adenocarcinoma A549 cells. Zhongguo Zhong Yao Za Zhi. 36:1670–1674. 2011.(In Chinese). PubMed/NCBI

47 

Dong H, Bai LP, Wong VK, Zhou H, Wang JR, Liu Y, Jiang ZH and Liu L: The in vitro structure-related anti-cancer activity of ginsenosides and their derivatives. Molecules. 16:10619–10630. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Harashima H, Dissmeyer N and Schnittger A: Cell cycle control across the eukaryotic kingdom. Trends Cell Biol. 23:345–356. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Roskoski R Jr: Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs. Pharmacol Res. 139:471–488. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Abbas T and Dutta A: p21 in cancer: Intricate networks and multiple activities. Nat Rev Cancer. 9:400–414. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Liu X, Sun Y, Yue L, Li S, Qi X, Zhao H, Yang Y, Zhang C and Yu H: JNK pathway and relative transcriptional factor were involved in ginsenoside Rh2-mediated G1 growth arrest and apoptosis in human lung adenocarcinoma A549 cells. Genet Mol Res. 15:2016. View Article : Google Scholar

52 

Chung KS, Cho SH, Shin JS, Kim DH, Choi JH, Choi SY, Rhee YK, Hong HD and Lee KT: Ginsenoside Rh2 induces cell cycle arrest and differentiation in human leukemia cells by upregulating TGF-β expression. Carcinogenesis. 34:331–340. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Fuchs Y and Steller H: Programmed cell death in animal development and disease. Cell. 147:742–758. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Plati J, Bucur O and Khosravi-Far R: Dysregulation of apoptotic signaling in cancer: Molecular mechanisms and therapeutic opportunities. J Cell Biochem. 104:1124–1149. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Guo XX, Li Y, Sun C, Jiang D, Lin YJ, Jin FX, Lee SK and Jin YH: p53-dependent Fas expression is critical for Ginsenoside Rh2 triggered caspase-8 activation in HeLa cells. Protein Cell. 5:224–234. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Wang X and Wang Y: Ginsenoside Rh2 mitigates pediatric leukemia through suppression of Bcl-2 in leukemia cells. Cell Physiol Biochem. 37:641–650. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Li Q, Li Y, Wang X, Fang X, He K, Guo X, Zhan Z, Sun C and Jin YH: 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. 50:760–769. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Evan GI and Vousden KH: Proliferation, cell cycle and apoptosis in cancer. Nature. 411:342–348. 2001. View Article : Google Scholar : PubMed/NCBI

60 

Manning BD and Cantley LC: AKT/PKB signaling: Navigating downstream. Cell. 129:1261–1274. 2007. View Article : Google Scholar : PubMed/NCBI

61 

Liu ZH, Li J, Xia J, Jiang R, Zuo GW, Li XP, Chen Y, Xiong W and Chen DL: Ginsenoside 20(s)-Rh2 as potent natural histone deacetylase inhibitors suppressing the growth of human leukemia cells. Chem Biol Interact. 242:227–234. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Zhang H, Yi J, Kim E, Choo Y, Hai H, Kim K, Kim EK, Ryoo Z and Kim M: 20(S)-Ginsenoside Rh2 suppresses oral cancer cell growth by inhibiting the Src-Raf-ERK signaling pathway. Anticancer Res. 41:227–235. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Wu N, Wu GC, Hu R, Li M and Feng H: Ginsenoside Rh2 inhibits glioma cell proliferation by targeting microRNA-128. Acta Pharmacol Sin. 32:345–353. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Ishay-Ronen D, Diepenbruck M, Kalathur RKR, Sugiyama N, Tiede S, Ivanek R, Bantug G, Morini MF, Wang J, Hess C and Christofori G: Gain fat-lose metastasis: Converting invasive breast cancer cells into adipocytes inhibits cancer metastasis. Cancer Cell. 35:17–32.e6. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Kim JH, Kim M, Yun SM, Lee S, No JH, Suh DH, Kim K and Kim YB: Ginsenoside Rh2 induces apoptosis and inhibits epithelial-mesenchymal transition in HEC1A and Ishikawa endometrial cancer cells. Biomed Pharmacother. 96:871–876. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Deryugina EI and Quigley JP: Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 25:9–34. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Zhang BP, Li B, Cheng JY, Cao R, Gao ST, Huang CJ, Li RP, Ning J, Liu B and Li ZG: Anti-cancer Effect of 20(S)-Ginsenoside-Rh2 on oral squamous cell carcinoma cells via the decrease in ROS and downregulation of MMP-2 and VEGF. Biomed Environ Sci. 33:713–717. 2020.PubMed/NCBI

68 

Riabov V, Gudima A, Wang N, Mickley A, Orekhov A and Kzhyshkowska J: Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front Physiol. 5:752014. View Article : Google Scholar : PubMed/NCBI

69 

Goumans MJ, Liu Z and ten Dijke P: TGF-beta signaling in vascular biology and dysfunction. Cell Res. 19:116–127. 2009. View Article : Google Scholar : PubMed/NCBI

70 

Fidler IJ and Ellis LM: The implications of angiogenesis for the biology and therapy of cancer metastasis. Cell. 79:185–188. 1994. View Article : Google Scholar : PubMed/NCBI

71 

Wang Q, Wu MQ, Zhao LH, Yang HK and Lv XH: Effect of ginsenoside Rh2 on transplanted-tumor and expression of JAM in mice. Zhongguo Zhong Yao Za Zhi. 33:2116–2119. 2008.(In Chinese). PubMed/NCBI

72 

Mizushima N, Levine B, Cuervo AM and Klionsky DJ: Autophagy fights disease through cellular self-digestion. Nature. 451:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Ryan KM: p53 and autophagy in cancer: Guardian of the genome meets guardian of the proteome. Eur J Cancer. 47:44–50. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Zhuang J, Yin J, Xu C, Mu Y and Lv S: 20(S)-Ginsenoside Rh2 Induce the Apoptosis and Autophagy in U937 and K562 Cells. Nutrients. 10:3282018. View Article : Google Scholar

75 

Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, Adachi H, Adams CM, Adams PD, Adeli K, et al: Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 12:1–222. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Liu S, Chen M, Li P, Wu Y, Chang C, Qiu Y, Cao L, Liu Z and Jia C: Ginsenoside rh2 inhibits cancer stem-like cells in skin squamous cell carcinoma. Cell Physiol Biochem. 36:499–508. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Li M, Zhang D, Cheng J, Liang J and Yu F: Ginsenoside Rh2 inhibits proliferation but promotes apoptosis and autophagy by down-regulating microRNA-638 in human retinoblastoma cells. Exp Mol Pathol. 108:17–23. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Ma J, Gao G, Lu H, Fang D, Li L, Wei G, Chen A, Yang Y, Zhang H and Huo J: Reversal effect of ginsenoside Rh2 on oxaliplatin-resistant colon cancer cells and its mechanism. Exp Ther Med. 18:630–636. 2019.PubMed/NCBI

79 

Zhou B, Xiao X, Xu L, Zhu L, Tan L, Tang H, Zhang Y, Xie Q and Yao S: A dynamic study on reversal of multidrug resistance by ginsenoside Rh(2) in adriamycin-resistant human breast cancer MCF-7 cells. Talanta. 88:345–351. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Liu GW, Liu YH, Jiang GS and Ren WD: The reversal effect of Ginsenoside Rh2 on drug resistance in human colorectal carcinoma cells and its mechanism. Hum Cell. 31:189–198. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Lv DL, Chen L, Ding W, Zhang W, Wang HL, Wang S and Liu WB: Ginsenoside G-Rh2 synergizes with SMI-4a in anti-melanoma activity through autophagic cell death. Chin Med. 13:112018. View Article : Google Scholar : PubMed/NCBI

82 

Wang M, Yan SJ, Zhang HT, Li N, Liu T, Zhang YL, Li XX, Ma Q, Qiu XC, Fan QY and Ma BA: Ginsenoside Rh2 enhances the antitumor immunological response of a melanoma mice model. Oncol Lett. 13:681–685. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Xia T, Zhang B, Li Y, Fang B, Zhu XX, Xu BC, Zhang J, Wang M and Fang JP: New insight into 20(S)-ginsenoside Rh2 against T-cell acute lymphoblastic leukemia associated with the gut microbiota and the immune system. Eur J Med Chem. 203:1125392020. View Article : Google Scholar : PubMed/NCBI

84 

Li Q, Li B, Dong C, Wang Y and Li Q: 20(S)-Ginsenoside Rh2 suppresses proliferation and migration of hepatocellular carcinoma cells by targeting EZH2 to regulate CDKN2A-2B gene cluster transcription. Eur J Pharmacol. 815:173–180. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Shi Q, Shi X, Zuo G, Xiong W, Li H, Guo P, Wang F, Chen Y, Li J and Chen DL: Anticancer effect of 20(S)-ginsenoside Rh2 on HepG2 liver carcinoma cells: Activating GSK-3β and degrading β-catenin. Oncol Rep. 36:2059–2070. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Qian T, Cai Z, Wong RN and Jiang ZH: Liquid chromatography/mass spectrometric analysis of rat samples for in vivo metabolism and pharmacokinetic studies of ginsenoside Rh2. Rapid Commun Mass Spectrom. 19:3549–3554. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Qi Z, Chen L, Li Z, Shao Z, Qi Y, Gao K, Liu S, Sun Y, Li P and Liu J: Immunomodulatory effects of (24R)-Pseudo-Ginsenoside HQ and (24S)-Pseudo-Ginsenoside HQ on cyclophosphamide-induced immunosuppression and their anti-tumor effects study. Int J Mol Sci. 20:8362019. View Article : Google Scholar

88 

Gu Y, Wang GJ, Sun JG, Jia YW, Wang W, Xu MJ, Lv T, Zheng YT and Sai Y: Pharmacokinetic characterization of ginsenoside Rh2, an anticancer nutrient from ginseng, in rats and dogs. Food Chem Toxicol. 47:2257–2268. 2009. View Article : Google Scholar : PubMed/NCBI

89 

Li S, Gao Y, Ma W, Guo W, Zhou G, Cheng T and Liu Y: EGFR signaling-dependent inhibition of glioblastoma growth by ginsenoside Rh2. Tumour Biol. 35:5593–5598. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Xia T, Wang JC, Xu W, Xu LH, Lao CH, Ye QX and Fang JP: 20S-Ginsenoside Rh2 induces apoptosis in human Leukaemia Reh cells through mitochondrial signaling pathways. Biol Pharm Bull. 37:248–254. 2014. View Article : Google Scholar : PubMed/NCBI

91 

Kim YS and Jin SH: Ginsenoside Rh2 induces apoptosis via activation of caspase-1 and −3 and up-regulation of Bax in human neuroblastoma. Arch Pharm Res. 27:834–839. 2004. View Article : Google Scholar : PubMed/NCBI

92 

Shi Q, Li J, Feng Z, Zhao L, Luo L, You Z, Li D, Xia J, Zuo G and Chen D: Effect of ginsenoside Rh2 on the migratory ability of HepG2 liver carcinoma cells: Recruiting histone deacetylase and inhibiting activator protein 1 transcription factors. Mol Med Rep. 10:1779–1785. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Guan N, Huo X, Zhang Z, Zhang S, Luo J and Guo W: Ginsenoside Rh2 inhibits metastasis of glioblastoma multiforme through Akt-regulated MMP13. Tumour Biol. 36:6789–6795. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Xia T, Zhang J, Zhou C, Li Y, Duan W, Zhang B, Wang M and Fang J: 20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway. J Ginseng Res. 44:725–737. 2020. View Article : Google Scholar : PubMed/NCBI

95 

Li S, Guo W, Gao Y and Liu Y: Ginsenoside Rh2 inhibits growth of glioblastoma multiforme through mTor. Tumour Biol. 36:2607–2612. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Li C, Gao H, Feng X, Bi C, Zhang J and Yin J: Ginsenoside Rh2 impedes proliferation and migration and induces apoptosis by regulating NF-κB, MAPK, and PI3K/Akt/mTOR signaling pathways in osteosarcoma cells. J Biochem Mol Toxicol. 34:e225972020. View Article : Google Scholar : PubMed/NCBI

97 

Yang D, Li X and Zhang X: Ginsenoside Rh2 induces DNA damage and autophagy in vestibular schwannoma is dependent of LAMP2 transcriptional suppression. Biochem Biophys Res Commun. 522:300–307. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Chen Y, Zhang Y, Song W, Zhang Y, Dong X and Tan M: Ginsenoside Rh2 improves the cisplatin anti-tumor effect in lung adenocarcinoma A549 cells via superoxide and PD-L1. Anticancer Agents Med Chem. 20:495–503. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 45 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang H, Park S, Huang H, Kim E, Yi J, Choi S, Ryoo Z and Kim M: Anticancer effects and potential mechanisms of ginsenoside Rh2 in various cancer types (Review). Oncol Rep 45: 33, 2021
APA
Zhang, H., Park, S., Huang, H., Kim, E., Yi, J., Choi, S. ... Kim, M. (2021). Anticancer effects and potential mechanisms of ginsenoside Rh2 in various cancer types (Review). Oncology Reports, 45, 33. https://doi.org/10.3892/or.2021.7984
MLA
Zhang, H., Park, S., Huang, H., Kim, E., Yi, J., Choi, S., Ryoo, Z., Kim, M."Anticancer effects and potential mechanisms of ginsenoside Rh2 in various cancer types (Review)". Oncology Reports 45.4 (2021): 33.
Chicago
Zhang, H., Park, S., Huang, H., Kim, E., Yi, J., Choi, S., Ryoo, Z., Kim, M."Anticancer effects and potential mechanisms of ginsenoside Rh2 in various cancer types (Review)". Oncology Reports 45, no. 4 (2021): 33. https://doi.org/10.3892/or.2021.7984