Skip to main content

Table 4 DDSs for ginsenoside studied in preclinical cancer models

From: Ginsenosides emerging as both bifunctional drugs and nanocarriers for enhanced antitumor therapies

DDS

Carrier

Bioactive compound

Target form/molecular

Cancer model

Loading efficiency

Encapsulation efficiency

Main results

References

Polymeric NPs

PEG-COOH

Rh2/Rh1

Passive tumor targeting

A549 cells

NA

NA

In vitro: PEG-Rh1 conjugate showed stronger anticancer activity in human non-small cell lung cancer cell line

[87]

NA

Rh2/Re

NA

MCF-7 cells

38%/32%

NA

In vitro: GS-Rh2 showed significant cytotoxicity to MCF-7 cancer cells

[21]

DA-OCMC

CK

NA

HepG2 cells

10.65% ± 1.49%

42.65% ± 1.24%

In vitro: CK-NPs showed dose-dependent inhibitory effects on HepG2 cells with IC50 values of 23.33 and 16.58 μg/mL

[105]

mPEG-b-P (Glu-co-Phe)

20 (S)-Rg3

Passive tumor targeting

Colorectal cancer/ mice

8.90%

82.40%

In vitro: Drug-loaded NPs possessed longer circulation time in blood

In vivo: Proliferation of tumors can be significantly inhibited by Rg3-NPs through reducing expression of proliferating cell nuclear antigen and inducing apoptosis by increasing expression of caspase-3 in subcutaneous colon cancer model in mice

[88]

ANG-Rg3-NP

Rg3

NA

C6 glioma cells

27.2% ± 1.4%

80.6% ± 3.0%

In vitro: ANG-Rg3-NPs inhibited the proliferation of C6 glioma cells in a concentration-dependent manner. Angioprep-2 functionalized NPs were easier to cross the BBB and accelerate uptake of NPs by cells

[89]

CS/HA /HPC

20 (R)-Rg3

NA

A549 cells

15.87% ± 0.09%

100.8% ± 6.1%

In vitro: Proliferation of A549 cells can be inhibited effectively by microparticles

[90]

Liposomes

ePC

Rg3

NA

A549 cells/ HepG2 cells/mice

NA

82.47% ± 0.74%

In vitro: Cytotoxicity of A549 and HepG-2 cells could be enhanced by L-Rg3

In vivo: Cmax and AUC of L-Rg3 were 1.19 × and 1.52 × higher than those of Rg3. Growth rate of BALB/c nude mice inoculated with A549 tumor cells was significantly inhibited by L-Rg3. Besides, Tumor growth can be inhibited by liposome by reducing MVD and enhancing angiogenesis inhibition

[97]

DSPE-PEG2000

Rg3

NA

NA

7.44% ± 0.08%

85.24% ± 1.02%

In vitro: Rg3-PEGylated liposomes showed sustained release

[96]

DSPE-PEG2000

Rh2

NA

A549 cells/mice

15.3%

88.2%

In vitro: IC50 values of A549 cells treated with DLT indicated that tumor growth could be inhibited by DLT

In vivo: DLT showed stronger antiproliferation and apoptosis effects on xenografted tumors. DDS was safer than cisplatin in treatment of tumors

[98]

mPEG-PLA

Rh2

Passive tumor targeting

HepG2/mice

NA

94.93% ± 4.18%

In vivo: Fluorescence intensity at tumor site decreased gradually after injection of PLP for 8 h and lasted for 24 h. Rh2-PLP was superior to Rh2-LP and Rh2-CLP in anti-tumor effect

[100]

EYPC/Rh2 /Rg3/Rg5

PTX/Rh2/Rg3 /Rg5

Active targeting: Rh2/Rg3/Rg5

BGC-823 cells/mice

Rh2: 5.6% ± 0.3%

Rg3: 7.3% ± 0.4%

Rg5: 4% ± 0.1%

Rh2: 91.3% ± 2.1%

Rg3: 95.5% ± 3.3%

Rg5: 82.8% ± 1.6%

In vitro: Ginsenoside liposome can be accumulated in tumor for recognizing GLUT carrier on tumor cell membrane

In vivo: Ginsenosides showed significant synergistic effects with PTX for antitumor activity

[26]

EYPC/Rh2

PTX/Rh2

Active targeting: Rh2

4T1 cells /mice

5.6%

91.3%

In vitro: PTX-Rh2-liposome showed ~ 80% cell apoptosis to 4T1 cells

In vivo: PTX-Rh2-liposome reduced tumor growth to certain extent comparable with lipisu

[25]

EPC/Rg3

PTX/Rg3

Active targeting: Rg3

C6 murine glioma cells/mice

9.80% ± 0.13%

94.15% ± 1.34%

In vitro: Rg3-liposome promoted C6 glioma cell’s uptake efficiency and tumor penetration simultaneously

In vivo: PTX-Rg3-liposome showed antiproliferation effects. Immune microenvironment in glioma was activated, with promoting T cell immune response

[28]

Lecithin/Rg5

PTX/Rg5

Active targeting: Rg5

HGC-27 /MCF-7 /A549 cells

NA

97.20%

In vivo: G-PTX achieved curative effects through targeting GLUT receptor on tumor surface

In vivo: Broad-spectrum targeting ability of G-PTX was confirmed with HGC-27, A549, and MCF-7 subcutaneous tumor models, through clathrin and caveolae-dependent pathways for endocytosis

[27]

DSPE-PEG2000-tLyp-1

CK

Active targeting: tLyp-1 peptide

A549 cells/mice

14.80%

83.40%

In vitro: tLyp-1 liposomes induced mitochondrial apoptosis of A549 tumor cells against tumor

In vivo: tLyp-1 liposomes showed stronger antitumor effect and fewer side-effects on normal tissues than drug combinations

[101]

Micelles

pNP-PEG-pNP

Rh2

NA

A549 cells

NA

85.23% ± 4.38%

In vitro: CG-M showed stronger cell uptake ability, apoptosis induction ability and antiproliferation activity of A549 cells

[113]

Solutol HS15/TPGS

Rh2

NA

A549 cells

7.68% ± 1.34%

95.27% ± 1.26%

In vitro: Rh2-M synthesized with Solutol HS15 and TPGS were capable of enhancing solubility and antitumor effects of Rh2

In vivo: Rh2-M displayed a higher tumor inhibition rate in tumor-bearing nude mice

[112]

TPGS/PEG-PCL

CK

Passive tumor targeting

A549 and PC-9 cells/ mice

11.19% ± 0.87%

94.60% ± 1.45%

In vitro: Growth of A549 and PC-9 cells could be inhibited by CK-M by blocking G1 phase. Bax and Bcl-2 were regulated to promote tumor cell apoptosis and inhibit tumor cell invasion, metastasis, and outflow

In vivo: CK-M micelles showed higher tumor inhibition and longer maintenance time of micelles in tumor tissue

[106]

PC/DP

CK

Passive tumor targeting

A549 cells/ mice

11.76% ± 1.32%

NA

In vitro: Micelles exerted proapoptotic effects and antitumor efficacy against human lung carcinoma A549 cells

In vivo: Micelles exhibited higher tumor inhibition than free CK through increased permeability and retention effects

[107]

DA-OCMC /A54 peptide

CK

Active targeting: peptide A54

HepG2/Huh-7 cells

3.18% ± 1.49%

76.56%

In vitro: Cytotoxicity of APD-CK micelles to HepG2 and Huh-7 cells was significantly higher than that of free CK. APD-CK micelles could promote protein expression of caspase-3, caspase-9, and poly (ADP-ribose) polymerase

[108]

AP/TPGS

CK

Passive tumor targeting

A549 cells/mice

13.26% ± 1.89%

91.34% ± 5.24%

In vitro: Mixed micelles induced cell apoptosis and inhibited cell migration by inducing cell cycle arrest in the G0/G1 phase of A549 cells

In vivo: A549 lung cancer xenografts in mice showed that mixed micelles were an efficient tumor-targeting DDS with obvious antitumor effects

[109]

MEs

PLA

20 (R)-Rg3

NA

NA

0.2853

0.78

In vitro: Ginsenoside Rg3 PLA microspheres exhibited controlled release of drugs

[104]

PLGA

ac-Rb1

NA

NA

NA

0.96

In vitro: Controlled release of ac-Rb1 followed the Fickian diffusion

[102]

Etoposide, coix seed oil

Rh2

NA

A549 cells/mice

NA

0.9

In vitro: Cytotoxicity and apoptosis induced by ECG-MEs were significantly enhanced in A549 cells

In vivo: Oral ECG-MEs could enter blood circulation through intestinal barrier, then prolonged blood circulation time and accumulated in tumor site. Mechanism of antitumor effect was related to small-scale mediated tumor penetration depth and increased serum Th1 cytokine concentration

[122]

Protein-based nanocarriers

BSA

Rg5

Active targeting: FA

A549 cells/mice

12.64% ± 4.02%

73.59% ± 5.50%

In vitro: EPR effect and receptor-mediated targeting led to MCF-7 cell apoptosis

In vivo: FA-modified targeted NPs efficiently accumulated Rg5 within 8 h at tumor site in MCF-7 xenograft mouse model, showing strong tumor aggregation capacity

[118]

BSA

Rh2

NA

A549/HT29 cells

0.36 mg of Rh2/mg of BSA-Rh2 NPs

NA

In vitro: BSA-CK NPs had stronger inhibitory effects on lung cancer A549, HepG2 hepatoma, and HT29 colon cancer cell lines

[115]

GNPs

DCY51T-AuCKNps

CK

NA

A549/HT29 cells

11.03%

NA

In vitro: DCY51T-AuCKNp showed enhanced cell apoptosis in A549, HT29, and AGS cells, suggesting that DCY51T-AuCKNp was an effective photothermal agent with synergistic chemotherapy effects

[110]

Carbon nanomaterials

CDs

Re

NA

MCF-7/HepG2/A375

NA

NA

In vitro: Small-sized Re-CDs were beneficial to cellular uptake, which had strong fluorescence imaging properties. Re-CDs could inhibit tumor cell proliferation through ROS-mediated pathway

[117]

CNTs

Rb1/Rg1

NA

MCF-7/PANC-1 cells

NA

NA

In vitro: Induction effect on MCF-7 and PANC-1 cell death pathway of ginsenoside CNT was stronger than pure ginsenoside

[103]

GO

Rh2

NA

OVCAR3/MDA-MB/A375 cells

NA

NA

In vitro: Rh2, amino acid Lys and Arg modified GO showed higher antitumor activity and lowest toxicity to coagulation system and heart tissue

[116]

MSNPs

MSNPs

CK/Rh2

NA

A549/HepG2/HT-29 cells

NA

NA

In vitro: MSNPs enhanced efficacy of CK and Rh2, exerting anticancer effects on HepG2, A549 and HT-29 colon cancer cells

[111]

  1. DDS drug delivery system, NPs nanoparticles, MEs microemulsions, GNPs gold nanoparticles, MSNPs mesoporous silica nanoparticles, CK compound K, GO graphene oxide, CDs carbon dots, pNP-PEG-pNP bis (4-nitrophenylcarbonate) polyethylene glycol, TPGS/PEG-PCL d-alpha Tocopheryl polyethylene glycol 1000 succinate/Poly (ethylene glycol)-poly (ε-caprolactone), PC/DP phosphatidylcholine/1,2-distearoyl-sn-glycero-3-phosphoethanolamine polyethylene glycol 2000, DA-OCMC deoxycholic acid-O carboxymethyl chitosan, A54 peptide liver cancer-specific binding peptide A54, AP/TPGS ascorbyl palmitate/d-α-tocopheryl polyethylene glycol 1000 succinate monoester, mPEG-b-P (Glu-co-Phe) poly (ethylene glycol)-block-poly (L-glutamic acid-co-l-phenylalanine), ANG-Rg3-NP angioep-2 polypeptide-Rg3, CS chitosan, HA hyaluronic acid, HPC hydroxypropyl cellulose, ePC yolk phosphatidylcholine, DSPE-PEG2000 1,2-distearoyl-sn-glycero-3-phosphoethanolamine poly (ethylene glycol) 2000, EYPC egg yolk lecithin, mPEG-PLA methoxy poly (ethylene glycol)-poly (lactide), PTX paclitaxel, tLyp-1 peptide CGNKRTR, PLA polylactide, PLGA poly (dl-lactide-co-glycolide), ac-Rb1 6″-O-Acetylginsenoside Rb1, BSA bovine serum albumin, FA folic acid, CDs carbon dots, CNTs carbon nanotubes, GO graphene oxide, DCY51T lactobacillus kimchicus, MSNPs mesoporous silica nanoparticles, NA not applicable, BBB blood–brain barrier, AUC area under the curve