Planta Med 2024; 90(05): 353-367
DOI: 10.1055/a-2258-6663
Biological and Pharmacological Activity
Reviews

Unveiling Gambogenic Acid as a Promising Antitumor Compound: A Review

Li Mi
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Zhichao Xing
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Yujie Zhang
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Ting He
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Anping Su
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Tao Wei
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Zhihui Li
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
,
Wenshuang Wu
Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
› Author Affiliations
Supported by: 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University ZYJC21033
Supported by: the project of West China Hospital 19HXFH010
Supported by: Foundation of Sichuan University 2018SCUH0067
 

Abstract

Gambogenic acid is a derivative of gambogic acid, a polyprenylated xanthone isolated from Garcinia hanburyi. Compared with the more widely studied gambogic acid, gambogenic acid has demonstrated advantages such as a more potent antitumor effect and less systemic toxicity than gambogic acid according to early investigations. Therefore, the present review summarizes the effectiveness and mechanisms of gambogenic acid in different cancers and highlights the mechanisms of action. In addition, drug delivery systems to improve the bioavailability of gambogenic acid and its pharmacokinetic profile are included. Gambogenic acid has been applied to treat a wide range of cancers, such as lung, liver, colorectal, breast, gastric, bladder, and prostate cancers. Gambogenic acid exerts its antitumor effects as a novel class of enhancer of zeste homolog 2 inhibitors. It prevents cancer cell proliferation by inducing apoptosis, ferroptosis, and necroptosis and controlling the cell cycle as well as autophagy. Gambogenic acid also hinders tumor cell invasion and metastasis by downregulating metastasis-related proteins. Moreover, gambogenic acid increases the sensitivity of cancer cells to chemotherapy and has shown effects on multidrug resistance in malignancy. This review adds insights for the prevention and treatment of cancers using gambogenic acid.


#

Introduction

Cancer continues to be the primary cause of mortality worldwide [1], with staggering numbers reported in the “Global Cancer Statistics 2020′′ report; approximately 19.3 million new cancer cases and nearly 10.0 million cancer-related deaths were recorded in 2020 alone [2]. Furthermore, it is projected that the global cancer burden will increase to 28.4 million cases by 2040 [2]. These statistics highlight the urgent need for comprehensive efforts in cancer prevention, diagnosis, and treatment to alleviate the global burden of this devastating disease. Over the years, several treatment modalities, such as surgery, radiotherapy, chemotherapy, and immunotherapy, have been developed for cancer. However, these approaches often come with side effects and have limitations in tackling tumor recurrence and metastasis. In recent decades, there has been a notable increase in the use of natural products with diverse bioactivities to combat different types of cancer. Natural products are recognized as potentially safer alternatives to traditional drug therapy, given the lower propensity for adverse effects [3], [4].

Garcinia hanburyi Hook. f., a plant belonging to the Guttiferae family, is a small tree distributed throughout India, Cambodia, Thailand, and the southern part of China [5]. Its resin, named gamboge, has been historically utilized in traditional folk medicine to address various conditions. Its applications include the treatment of chronic dermatitis, hemorrhoids, bedsores, and tapeworm infections [6]. In recent years, this resin has garnered heightened attention due to its broad spectrum of biological and pharmacological properties. These properties encompass cytotoxic [7], [8], [9], antiproliferative [10], [11], [12], antitumor [13], [14], antiangiogenesis [15], anti-HIV-1 [16], [17], antibacterial [18], and anti-inflammatory effects [19]. Gamboge contains a diverse array of bioactive components, primarily caged xanthones. To date, over 40 different caged xanthones have been identified and explored for their potential medicinal applications [20] ([Fig. 1]). Gambogic acid (GA; C38H44O8) has been claimed to be the major constituent of gamboge [21]. It has shown immense potential as a candidate broad-spectrum anticancer drug [22], [23], [24], [25]. In 2014, GA was approved by the Chinese Food and Drug Administration for a phase II clinical trial in lung cancer and other solid tumor therapies [26], but this clinical trial has been terminated.

Zoom Image
Fig. 1 Structure diagrams of some caged xanthones with antitumor activity. The chemical structures of compounds were derived from PubChem (https://pubchem.ncbi.nlm.nih.gov/).

Asano et al. [27] isolated a polyprenylated xanthone from gamboge with a structure similar to GA, referred to as gambogenic acid (GNA; C38H46O8). GNA shares structural similarity with GA but differs in the presence of a geranyl group and a hydroxyl group instead of the ether ring found in GA. Many studies have reported that GNA has multiple biological functions, such as anti-inflammation [28], [29], anti-fibrosis [30], and antiangiogenesis [31]. Meanwhile, GNA has demonstrated a notable antitumor effect and serves as an inhibitor of enhancer of zeste homolog 2 (EZH2), employed in a study of diverse cancer types [32]. Compared with the more widely studied GA, GNA has shown several advantages in terms of its antitumor effect and systemic toxicity, as indicated by early investigations [27], [33], [34], [35], [36], [37]. Nevertheless, to date, there has been no comprehensive review of the antitumor effect of GNA and its underlying mechanisms, as well as the drug delivery of GNA for cancer therapeutics. Therefore, this paper aims to systematically review the antitumor effects and mechanisms, pharmacokinetics, and nanotechnology-mediated delivery of GNA, with the goal of providing reference material for its application in antitumor research and promoting its further development and utilization ([Fig. 2]). By searching PubMed, Web of Science, and ScienceDirect databases, covering the period from 1996 to 2023, this paper has retrieved a total of 104 articles using the main terms “gambogenic acid”, “Garcinia hanburyi”, “Guttiferae”, “gamboge”, “cancer”, “pharmacokinetic”, and their combinations. We evaluated both experimental papers and reviews that were deemed relevant and identified 55 articles that were considered to be useful and appropriate for analysis.

Zoom Image
Fig. 2 A schematic representation of the antitumor activity, pharmacokinetics, nanotechnology-mediated delivery of GNA.

#

Therapeutic Activity of Gambogenic Acid in Cancer

In vitro studies

Numerous preclinical investigations have shown that GNA has an impact on a variety of human malignancies, including lung [38], breast [39], colorectal [36], cervical [40], gastric [41], bladder [42], prostate [43], epithelial cancers [32], hepatic carcinoma (HCC) [44], melanoma [45], multiple myeloma [33], nasopharyngeal carcinoma [46], glioblastomas [47], osteosarcoma [48], and leukemia [49] ([Table 1]). GNA suppressed the cell growth of cancer cell lines but also showed low toxicity toward normal cells [36], [50].

Table 1 Antitumor effect of GNA in vitro.

Cancer

Cell type

Concentration range (µM)

IC50 values (µM)/duration

Antitumor effect

Refs.

Breast cancer

MCF-7, MCF-7/ADR

0.06 – 31.73

Apoptosis

[54]

MDA-MB-231

0.32 – 4.76

1.24/72 h

Apoptosis

[39]

4 T1

0.78 – 310.00

7.57/24 h

Cytotoxicity

[55]

Lung cancer

H446

0.6 2– 2.40

1.40/48 h

Apoptosis

[37]

H1688

1.2 0– 3.20

2.40/48 h

H1975, H460

0.50 – 10.00

~ 2.50/48 h

Autophagy

[38]

HCC827

1.33/72 h

Proliferation inhibition

[52]

HCC827/ER

1.51/72 h

H1650

0.91/72 h

A549

1.50 – 12.00

Autophagy

[74]

A549

0.32 – 39.68

Cytotoxicity

[40]

A549

0.32 – 32.00

Apoptosis

[67]

A549

0.31 – 10.00

2.50/24 h

[35]

A549

2.83/24 h

Apoptosis, necroptosis

[51]

A549

8.07/24 h

Apoptosis

[41]

A549

7.00/48 h

Autophagy

[38]

A549

0.50 – 8.00

2.00/72 h

Apoptosis

[68]

A549/Cis

0.50 – 6.00

Apoptosis

[64]

Hepatic carcinoma

HepG2

0.75 – 12.00

3.23/24 h, 2.62/48 h, 2.14/72 h

Apoptosis

[44]

HepG2

0.50 – 4.00

Proliferation inhibition

[50]

HepG2

0.32 – 32.00

10.27/24 h

Apoptosis

[95]

HepG2

0.32 – 16.00

3.40/48 h

Proliferation inhibition

[53]

HepG2

7.94 – 39.68

11.16/24 h

Cytotoxicity

[93]

HepG2

0.51 – 16.43

15.70/24 h

Cytotoxicity

[92]

HepG2

10.71/24 h

Apoptosis

[41]

HepG2

0.78– 15.50

7.43/24 h

Cytotoxicity

[94]

HepG2

0.50 – 16.00

4.40/48 h

Cytotoxicity

[91]

HepG2

Apoptosis

[66]

HepG2

1.59 – 50.79

7.94 – 9.52/48 h

Apoptosis

[96]

HepG2

1.00 – 8.00

3.90/48 h

Proliferation inhibition

[77]

HepG2/ADR

0.32 – 16.00

7.19/48 h

Proliferation inhibition

[53]

Hepa 1 – 6

0.16 – 5.08

1.27 – 1.59/48 h

Apoptosis

[96]

BEL-7402/ADR

1.00 – 32.00

Apoptosis, autophagy

[70]

Bel-7402

0.54 – 4.00

Proliferation inhibition

[50]

SMMC-7721

1.00 – 16.00

10.68/24 h

Apoptosis

[65]

Melanoma

A375

1.00 – 16.00

2.88/24 h

Ferroptosis, autophagy

[73]

A2058

1.00 – 16.00

1.26/24 h

B16

0.50 – 8.00

2.09/24 h

B16F10

0.5 0– 8.00

1.00/24 h

A375, A2058

1.00 – 16.00

Ferroptosis, metastasis inhibition

[45]

B16, B16F10

0.10 – 4.00

Metastasis inhibition

[81]

OCM-1

0.75 – 6.00

Proliferation and metastasis inhibition

[34]

Multiple myeloma

MM.1S

1.75/24 h, 0.90/48 h, 0.84/72 h

Apoptosis

[33]

U266

0.1 0– 1.60

Apoptosis, proliferation inhibition

[78]

Colorectal cancer

HCT116

0.50 – 3.00

1.88/24 h, 1.48/48 h

Apoptosis, proliferation inhibition

[36]

SW620

0.50 – 3.00

2.83/24 h, 1.81/48 h

DLD-1

0.50 – 3.00

1.97/24 h, 1.55/48 h

HCT116

0.50 – 5.00

Apoptosis

[57]

HCT116, HT29

0.5 0– 5.00

Ferroptosis, proliferation inhibition

[77]

Bladder cancer

BIU-87, T24, J82

0.75 – 3.00

Apoptosis, metastasis inhibition

[42]

Prostate cancer

PC3, DU145

2.00 – 12.00

Apoptosis, autophagy

[43]

Nasopharyngeal carcinoma

CNE-1

0.25 – 8.00

1.87/72 h

Apoptosis

[46]

CNE-2Z

0.25 – 8.00

2.25/24 h, 1.33/48 h

Apoptosis

[72]

Glioblastoma multiforme

U251

0.75 – 6.00

Apoptosis, metastasis inhibition

[47]

Epithelial cancer

HN-6

Proliferation inhibition

[32]

Gastric cancer

SGC-7901

16.15/24 h

Apoptosis

[41]

Cervical cancer

HeLa

0.1 0– 1.59

Cytotoxicity

[40]

HeLa

1.50 – 12.00

Autophagy

[74]

Leukemia

K562

3.83/44 h

Cytotoxicity

[49]

K562/ADR

4.78/44 h

Osteosarcoma

143B

0.25 – 8.00

1.18/24 h

Apoptosis, ferroptosis, metastasis inhibition

[48]

HOS

0.25 – 8.00

1.10/24 h

Moreover, several recent studies have indicated that GNA exhibits significant synergistic effects when combined with different therapeutic agents, including 5-fluorouracil (5-FU) [51], erlotinib [52], and bortezomib (BTZ) [33], against various types of cancer cells. GNA could also overcome chemotherapeutic medication resistance in HCC [53], non-small cell lung cancer (NSCLC) [52], and breast cancer [54].


#

In vivo studies

Various studies have confirmed that GNA has antitumor potential in vivo. GNA can counteract tumor growth in vivo in various tumor mouse models, such as lung cancer [37], breast cancer [55], and multiple myeloma [33] ([Table 2]). These experiments mostly utilize xenograft models, among which the patient-derived tumor xenograft (PDX) model has emerged as the most trustworthy in vivo human cancer model due to its ability to preserve the features of the original patient tumor, such as gene expression profiles and treatment responses [56]. GNA increased the antitumor activity of erlotinib in a fibroblast growth factor receptor (FGFR)-expressing PDX xenograft model [52]. In addition, GNA significantly suppressed tumor growth and progression in the APCmin/+ mouse model [36] and the azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model [57]. GNA exerts antitumor effects with low toxicity in animal models. No apoptotic cell death was observed in tissues [37]. Between the vehicle- and GNA-treated groups, there were no appreciable variations in body weight or blood biochemical markers [alanine aminotransferase (ALT) and aspartate aminotransferase AST)] [36].

Table 2 Antitumor effect of GNA in vivo.

Cancer

Animal model type

Dose (mg/kg)

Route

Duration

Antitumor effect

Molecular mechanism

Refs.

AOM, azoxymethane; APC, adenomatous polyposis coli; BiP, immunoglobulin heavy chain-binding protein; CHOP, C/EBP-homologous protein; DSS, dextran sulfate sodium; FGFR, fibroblast growth factor receptor; GPX4, glutathione peroxidase 4; IRE1, inositol-requiring enzyme 1; JNK, c-Jun N-terminal kinase; Nrf2, nuclear factor E2-related factor 2; PARP, poly(ADP-ribose) polymerase; PCNA, proliferating cell nuclear antigen; PDX, patient-derived xenograft; PTEN, phosphatase and tensin homolog

Breast cancer

4 T1 xenograft

24.0

i. g.

23 days

Tumor growth and weight↓

[55]

MDA-MB-231 xenograft

4.0, 8.0, 12.0

16 days

Tumor growth and weight ↓

[39]

Lung cancer

NCI-H446 xenograft

4.0, 12.0

i. v.

14 days

Tumor growth and weight ↓

Apoptosis↑

[37]

A549 xenograft

7.5, 30.0

i. g.

14 days

Tumor growth and weight ↓

[85]

A549 xenograft

1.5

i. p.

14 days

Tumor growth and weight ↓

A549 xenograft

16.0

i. v.

Tumor growth and weight ↓

LC3-II/I↑, p62↑

[74]

HCC827/ER xenograft

10.0

i. p.

27 days

Tumor growth and weight ↓

FGFR signaling pathway↓

[52]

PDX model

7.5

i. p.

22 days

A549 xenograft

8.0, 16.0, 32.0

i. p.

2 weeks

Tumor growth ↓

Apoptosis ↑

[35]

Melanoma

B16F10 xenograft

2.0, 4.0, 8.0

i. p.

18 days

Tumor growth and weight ↓

[73]

B16F10 xenograft

2.0, 4.0, 8.0

i. p.

22 days

Tumor growth and weight ↓, pulmonary metastasis ↓

EMT↓

[81]

Multiple myeloma

MM.1S xenograft

2.0

i. v.

2 weeks

Tumor growth and weight ↓

Apoptosis ↑

[33]

U266 xenograft

5.0

i. v.

14 days

Tumor growth and weight ↓

microRNA-21/PTEN↓

[78]

Colorectal cancer

HCT116 xenograft

1.0, 2.0

i. p.

21 days

Tumor growth ↓

IRE1α/JNK ↑, Noxa-induced apoptosis ↑

[36]

HCT116 xenograft

4.0

i. p.

21 days

Tumor growth and weight ↓

[77]

APCmin/+ model

1.0, 2.0

i. p.

2 months

Number and size of intestinal polyps ↓

[36]

AOM/DSS mouse model

Colon length ↑

BiP ↑, CHOP ↑

[57]

Prostate cancer

PC3 xenograft

4.0

i. p.

25 days

Tumor growth and weight ↓

PCNA ↓, PARP ↑, LC3 ↑, p-IRE1 ↑, p-JNK↑, Nrf2↑

[43]

Hepatic carcinoma

Hepa1-6 xenograft

4.0

i. v.

12 days

Tumor growth and weight ↓

[96]

Osteosarcoma

143B xenograft

30.0, 60.0

i. g.

3 weeks

Tumor growth ↓

GPX4↓, caspase-3↑

[48]


#

The antitumor mechanism of gambogenic acid in cancer

GNA blocks cancer development in vitro and in vivo by inhibiting the proliferation of tumor cells, activating cell cycle arrest, and inducing tumor cell death through different processes (apoptosis, autophagy, necrosis and ferroptosis). GNA also hinders tumor cell invasion and metastasis. Furthermore, GNA increases the sensitivity of cancer cells to chemotherapy and has shown effects on drug resistance in malignancy. We summarized the effectiveness and mechanisms of GNA in different cancers ([Table 3] and [Fig. 3]).

Table 3 Molecular mechanisms underlying antitumor activities of GNA.

Effect

Cancer type

Molecular mechanism

Refs.

AMPK, AMP-activated protein kinase; Bad, Bcl-2-associated death; Bax, Bcl-2 associated X; Bcl-2, B-cell lymphoma 2; CDKs, cyclin-dependent kinases; cIAP2, cellular inhibitor of apoptosis 2; CIP2A, cancerous inhibitor of protein phosphatase 2A; COX, cyclooxygenase; EMT, epithelial-to-mesenchymal transition; ERK1/2, extracellular signal-regulated kinase 1/2; FGFR, fibroblast growth factor receptor; FOXA2, forkhead box protein A2; GADD45A, growth arrest and DNA damage-inducible α; GPX4, glutathione peroxidase 4; GSK3β, glycogen synthase kinase-3beta; IRE1, inositol-requiring enzyme 1; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MEG3, maternally expressed gene 3; mTOR, mammalian target of rapamycin; NEAT1, nuclear-enriched abundant transcript 1; NF-κB, nuclear transcription factor-kappa B; PARP, poly(ADP-ribose) polymerase; PCNA, proliferating cell nuclear antigen; P-gp, P-glycoprotein; PI3K, the phosphatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog; RIP1, receptor-interacting protein 1; ROS, reactive oxygen species; STMN1, stathmin 1; VSOR · Cl, volume-sensitive outwardly rectifying chloride; XIAP, X-linked inhibitor of apoptosis

G0/G1 phase arrest

Lung cancer

cyclin D1, D3↓, CDK 4, 6↓, COX-2↓, GSK3β↓, p21↑, p27↑, GADD45A↑, p53↑

[35], [38], [64]

Glioblastoma multiforme

cyclin D1, E↓, CDK 2, 4↓, p21↑, p27↑, GSK3β↓, Akt↓

[47]

Choroidal melanoma

cyclin D1, E↓, CDK 2↓

[34]

Nasopharyngeal carcinoma

[46]

G2/M phase arrest

Prostate cancer

cyclin A2, B1↓, p27↑

[43]

Multiple myeloma

[33]

S phase arrest

Lung cancer

p53↑

[37]

Prostate cancer

cyclin A2, B1↓, p27↑

[43]

Apoptosis

Lung cancer

caspase-3, -7, -8, -9↑, PARP↑, cytochrome c↑, Bax↑, Bcl-2↓, p53↑, p38↓, MAPK↓

[35], [37], [41], [51], [64], [67], [68]

Hepatic carcinoma

caspase-3, -9↑, Bax↑, Bcl-2↓, ROS↑, p-p38↑, p-ERK1/2↓

[41], [44], [65], [66], [70]

Breast cancer

caspase-3, -8, -9↑, Fas↑, Bax↑, Bcl-2↓

[39], [54]

Multiple myeloma

caspase-3↑, PARP↑, p53↑, Bax↑, Bcl-2↓

[33], [78]

Colorectal cancer

caspase-3, -8, -9↑, PARP↑, ROS/IRE1α/JNK↑, Aurora A ↓

[36], [57]

Nasopharyngeal carcinoma

caspase-9↑, cytochrome c↑, Ca2+↑, Bax↑, Bad↑, Bcl-2↓, Akt↓, VSOR · Cl channels↑

[46], [72]

Glioblastoma multiforme

caspase-3↑, Akt↓

[47]

Gastric cancer

caspase-3, -9↑, Bax↑, Bcl-2↓

[41]

Osteosarcoma

caspase-3, -9↑, Bax↑, Bcl-2↓, p53↑

[48]

Prostate cancer

PARP↑, Bax↑, Bcl-2↓, JNK/c-JUN↑, ROS↑

[43]

Bladder cancer

cIAP2↓, XIAP↓, Survivin↓

[42]

Ferroptosis

Melanoma

SLC7A11/GPX4↓, lncRNA NEAT1↓

[45], [73]

Osteosarcoma

SLC7A11/GPX4↓

[48]

Colorectal cancer

Target the miR-1291/FOXA2 and AMPKα/SLC7A11/GPX4 axis

[77]

Necroptosis

Lung cancer

RIP1↑

[51]

Autophagy

Lung cancer

LC3-II/I↑, p62↑, Beclin1↑, Akt/mTOR↓, GSK3β

[38], [74]

Hepatic carcinoma

LC3-II/I↑, p62↑, Beclin1↑

[70]

Melanoma

AMPK/mTOR↓, lncRNA NEAT1↓

[73]

Prostate cancer

JNK/c-JUN↑, p62↑

[43]

Proliferation inhibition

Hepatic carcinoma

CIP2A↓, STMN1↓

[50], [77]

Multiple myeloma

microRNA-21/PTEN↓

[78]

Colorectal cancer

c-Myc↓, PCNA↓

[36]

Choroidal melanoma

PI3K/Akt↓

[34]

Metastasis inhibition

Melanoma

EMT↓

[34], [45]

Melanoma

EMT↓, lncRNA MEG3 ↑

[81]

Bladder cancer

NF-κB↓

[42]

Glioblastoma multiforme

[47]

Osteosarcoma

[48]

Synergistic effect

Lung cancer (Erlotinib)

FGFR signaling pathway↓

[52]

Lung cancer (5-Fluorouracil)

ROS-mitochondria pathway↑

[51]

Multiple myeloma (Bortezomib)

apoptosis↑

[33]

Reversal drug resistance

Lung cancer (Erlotinib)

FGFR signaling pathway↓

[52]

Lung cancer (Cisplatin)

apoptosis↑

[64]

Liver cancer (Adriamycin)

P-gp↓, MAPK↓, NF-κB↓

[53]

Breast cancer (Adriamycin)

PTEN/PI3K/Akt↓

[54]

Hepatic carcinoma (Adriamycin)

basal autophagy↓

[70]

Zoom Image
Fig. 3 Mechanisms underlying the antitumor effect of GNA. GNA serves as an inhibitor of EZH2, effectively suppressing tumor cell proliferation through regulating various proteins such as PTEN, CIP2A, and STMN1. Through the activation of p21/p27, GNA modulates downstream cell cycle regulatory proteins like CDK2 and cyclins A2/B1/E, leading to cell cycle arrest. Another mechanism involves GNA inducing cell cycle arrest by inhibiting Akt, thereby impacting CDK4/6 and cyclin D1/D3 through downstream GSK3β. GNAʼs inhibition of Akt also triggers autophagy in tumor cells via the AMPK-mTOR pathway and facilitates mitochondrial apoptosis by modulating the expression of Bcl-2 family proteins. In some tumor cells, GNA also prompts apoptosis through ER stress. Furthermore, GNA activates caspase-8 through Fas, initiating cell apoptosis. In alternative antitumor mechanisms, GNA induces cell ferroptosis via the p53/SLC7A11/GPX4 signaling pathway and induces cell necrosis by activating RIP1, as well as hinders tumor cell migration through the NF-κB-mediated EMT pathway.

#

Gambogenic acid target-directed enhancer of zeste homolog 2

EZH2 serves as the enzymatic component within polycomb repressive complex 2 (PRC2), which is responsible for methylating lysine 27 on histone H3. This methylation event ultimately aids in the process of transcriptional silencing, where gene expression is suppressed [58]. Dysregulation of EZH2 causes alterations in gene expression and functions, thereby promoting cancer development. Numerous studies have revealed that both overexpression and mutation of EZH2 have been detected in a wide array of human cancers, spanning from breast cancer, prostate cancer, endometrial cancer, melanoma, bladder cancer, colon cancer, liver cancer, and lung cancer to lymphoma [59]. Elevated levels of EZH2 have been associated with increased tumor cell proliferation, migration, and angiogenesis within tumor tissue. These effects contribute to further deterioration of the tumor tissue, leading to poor prognosis and shorter survival times for patients [60]. Therefore, EZH2 is regarded as a critical oncogene and a potential drug target for various human malignant tumors. Research on antitumor drugs directed at this target has garnered significant attention. From 2012 onwards, numerous EZH2 inhibitors have progressed into clinical trials, reflecting the growing interest in developing therapeutics targeting EZH2 [61], [62].

Wang et al. [32] documented that GNA and its derivatives serve as a novel category of EZH2 inhibitors, functioning through direct and covalent binding with EZH2. This process effectively disrupts the PRC2 complex, thus inhibiting its methyltransferase activity. Additionally, GNA demonstrates superior antitumor effects by not only inhibiting EZH2 enzymatic activity but also facilitating its ubiquitination-mediated degradation, thereby fully suppressing its oncogenic functions.


#

Gambogenic acid induces cell cycle arrest in cancer cells

The cell cycle plays a crucial role in regulating cell growth, proliferation, and survival. When prompted by extracellular signals, cyclin D is one of the primary proteins to be expressed, which then binds to cyclin-dependent kinases (CDKs) to activate the downstream cascade reaction [63]. GNA markedly arrested the cell cycle at the G0/G1 phase in lung cancer [38], nasopharyngeal carcinoma [46], choroidal melanoma [34], and glioblastoma multiforme cells [47]. Yu et al. [38] showed that GNA is a cyclin D1 inhibitor and induces G1 arrest via glycogen synthase kinase-3beta (GSK3β)-dependent cyclin D1 degradation in the lung cancer cell lines H1975 and H460. GNA arrested the cell cycle at the G1 phase in the cisplatin-resistant lung cancer cell line A549/Cis through the downregulation of cyclin D1, cyclin D3, CDK4, and CDK6 and the upregulation of p21, p53, and growth arrest and DNA damage-inducible α (GADD45A) [64]. Meanwhile, GNA arrested A549 cells in the G0/G1 phase and downregulated the expression of cyclin D1 and cyclooxygenase (COX)-2 at the mRNA level [35]. In glioblastoma multiforme, GNA efficiently arrested U251 cells at the G0/G1 phase by specifically repressing the expression of cyclin D1, cyclin E, CDK2, and CDK4 and increasing the expression of p21 and p27 [47].

In addition, GNA could induce G2/M phase arrest in myeloma MM.1S cells [33]. GNA induced G2/M phase arrest in prostate cancer cells through the downregulation of cyclin A2 and cyclin B1 expression levels and the enhancement of p27 expression [43]. GNA also inhibited cell cycle progression in lung cancer and prostate cancer cell lines by inducing arrest at the S phase [37], [43].


#

Gambogenic acid induces apoptosis in cancer cells

Apoptosis was confirmed to be a crucial pathway in the chemotherapeutic approach to antitumor therapy. Multiple studies have indicated that GNA can hinder proliferation by inducing apoptosis in breast cancer [54], lung cancer [37], colorectal cancer (CRC) [36], bladder cancer [42], prostate cancer [43], gastric cancer [41], HCC [41], [65], [66], multiple myeloma [33], nasopharyngeal carcinoma [46], and glioblastoma multiforme [47]. Among them, most reports are on GNA-induced apoptosis in lung cancer cells. GNA facilitated the apoptosis of lung cancer cells in a dose- and time-dependent manner, a process associated with the modulation of proteins involved in apoptosis pathways in A549 [35], [41], [51], [67], [68], H446, and H1688 cells [37]. Caspase-3, -7, -8, and -9, Bcl-2 associated X (Bax), and cytochrome c were upregulated, while the B-cell lymphoma 2 (Bcl-2) protein was downregulated. The hallmark of apoptosis, poly(ADP-ribose) polymerase (PARP), which is associated with DNA repair, and p53 proteins were significantly increased by GNA treatment in lung cancer cell lines [37]. GNA was also able to significantly increase the activation of caspase-3 and caspase-7 in addition to the cleavage of PARP, in the cisplatin-resistant NSCLC cell line A549/Cis [64].

Apoptosis encompasses three pathways: (1) the death receptor or extrinsic pathway, (2) the mitochondrial or intrinsic pathway, and (3) the endoplasmic reticulum pathway. The mitochondrial apoptotic pathway serves as a significant cellular death pathway, involving death receptors initiating apoptosis from the cell surface, Bcl-2 family members acting as the guardians of the mitochondrial pathway, and caspases-3, -7, and − 9 as the executor enzymes [69]. Many studies have reported that GNA induces mitochondria-dependent apoptosis in various cancers, such as HCC [41], [70], osteosarcoma [48], multiple myeloma [33], nasopharyngeal carcinoma [46], breast cancer [54], gastric cancer [41], CRC [36], prostate cancer [43], and lung cancer [41], [51]. The p38 mitogen-activated protein kinase (MAPK) cascade inhibits apoptosis. GNA induced mitochondria-dependent apoptosis via the extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 MAPK pathways in human hepatoma HepG2 cells and lung cancer A549 cells [44], [68]. Yan et al. [46] stated that GNA induces apoptosis through mitochondrial oxidative stress, and its molecular mechanisms are linked to the generation of ROS, intracellular calcium overload, and inactivation of the Akt signaling pathway in nasopharyngeal carcinoma CNE-1 cells. Noxa, a proapoptotic member of the Bcl-2 protein family, induces Bax-mediated mitochondrial dysfunction through indirect inhibition of other Bcl-2 family members. GNA induced Noxa-mediated apoptosis by inducing reactive oxygen species (ROS) generation and c-Jun N-terminal kinase (JNK) activation in CRC both in vitro and in vivo [36]. Zhou et al. [39] confirmed that GNA could effectively suppress breast cancer MDA-MB-231 cell growth by mediating apoptosis not only through the mitochondrial pathway but also through the Fas/FasL death receptor pathway in vitro and in vivo.

ROS overproduction induces the accumulation of misfolded proteins in the endoplasmic reticulum (ER), subsequently leading to ER stress [71]. GNA increased the expression of the ER stress-associated proteins p-IRE1, immunoglobulin heavy chain-binding protein (BiP), and C/EBP-homologous protein (CHOP) in prostate cancer cells [43]. GNA triggers ER stress-mediated apoptosis through the ROS/inositol-requiring enzyme 1 (IRE1α)/c-Jun JNK signaling pathway in CRC [36] and triggers apoptosis via BiP, activating transcription factor 4 (ATF4), and CHOP in nasopharyngeal carcinoma [72]. Liu et al. [57] showed that GNA inhibited CRC proliferation by activating ER stress in vitro and in vivo and triggered ER stress by regulating Aurora A. GNA can activate volume-sensitive outwardly rectifying chloride (VSOR · Cl) channels, leading to ER stress, inducing apoptosis, and inhibiting proliferation in CNE-2Z cells [72].

Moreover, GNA treatment significantly inhibited the expression of antiapoptotic proteins, including cellular inhibitor of apoptosis 2 (cIAP2), X-linked inhibitor of apoptosis (XIAP), and survivin, in bladder cancer cells [42]. The proapoptotic effect of GNA on U251 glioblastoma cells was shown to be mediated through inactivation of the Akt pathway [47]. Furthermore, GNA-mediated inactivation of EZH2 led to elevated expression of the proapoptotic protein Bim, which is a well-characterized EZH2 downstream transcriptional target [32].


#

Gambogenic acid induces autophagy in cancers

Autophagy, a self-degradation mechanism, plays a critical role in maintaining cellular homeostasis during stress and exhibits complex, dual roles in both tumorigenesis and cancer development. On the one hand, GNA induces “autophagic cell death”, which is also known as type II programmed cell death, in lung cancer [38], prostate cancer [43], and melanoma [73]. GNA induces autophagy in lung cancer cells, possibly due to activation of GSK3β and inactivation of the Akt/mammalian target of rapamycin (mTOR) signaling pathway [38]. GNA autophagy in melanoma cells could be induced by inhibiting the activation of AMP-activated protein kinase (AMPK) by long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1), indirectly inhibiting the phosphorylation of downstream mTOR proteins [73]. GNA induces apoptosis and autophagy through ROS-mediated ER stress via the JNK signaling pathway in prostate cancer cells [43].

On the other hand, the levels of autophagy in tumor cells can influence cellular resistance. Inhibition of autophagy can reverse drug resistance in cancer cells, potentially increasing the effectiveness of chemotherapy. GNA inhibits protective autophagy in BEL-7402/ADR HCC cells [70]. Furthermore, GNA has the ability to impede the fusion of autophagosomes with lysosomes by inhibiting lysosomal acidification. This dysfunctional autophagy contributes to the pro-death role in GNA-mediated lung cancer cell death [74].


#

Gambogenic acid induces necroptosis and ferroptosis in cancers

Necroptosis, a form of programmed cell death, differs from apoptosis in that it does not engage key apoptosis regulators, such as Bcl-2 family members. In this pathway, receptor interacting protein 1 (RIP1) is a specific target associated with necroptosis [75]. The combination of GNA and 5-FU induced cell death in A549 cells by activating caspase-independent necroptosis [51].

Ferroptosis, a mode of regulated necrosis that is dependent on iron, has emerged as a novel form of cell death. The three core features of ferroptosis include sufficient polyunsaturated fatty acid phospholipid oxidation, the concentration of active iron, and the loss of lipid peroxidation [76]. GNA induced ferroptosis in osteosarcoma cells [48] and transforming growth factor beta (TGF-β)1-stimulated melanoma cells via the p53/SLC7A11/glutathione peroxidase 4 (GPX4) signaling pathway [45]. Furthermore, GNA downregulated lncRNA NEAT1, which can weaken the direct binding of SLC7A11, indirectly leading to inhibition of GPX4 activity and subsequent ferroptosis in melanoma cells [73]. In addition, GNA inhibited proliferation and ferroptosis by targeting the microRNA-1291 (miR-1291)/FOXA2 and AMPKα/SLC7A11/GPX4 axis in CRC [77].


#

Other pathways by which gambogenic acid inhibits the proliferation of cancer cells

By using a proteomics approach, Wang et al. [78] identified that stathmin 1 (STMN1) might be a major molecular target by which GNA inhibits HCC cell proliferation. Meanwhile, GNA was identified as a cancerous inhibitor of protein phosphatase 2A (CIP2A) inhibitor that interferes with the ubiquitination and destabilization of CIP2A and showed potent antiproliferative activity and enhanced the effect of chemotherapeutic agents against HCC by suppressing the CIP2A-Akt pathway [50]. Li et al. [34] reported that GNA may inhibit choroidal melanoma cell growth via inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway.

In addition, microRNA-21 (miR-21) has been found to be overexpressed in multiple myeloma patients and associated with the occurrence and development of multiple myeloma. GNA inhibited tumor proliferation in hypoxic multiple myeloma cells by regulating the miR-21/phosphatase and tensin homolog (PTEN) pathway [79].


#

Gambogenic acid inhibits the metastasis of cancer cells

Extensive invasion and migration into surrounding tissue are hallmark characteristics of cancer, making these lesions resistant to definitive surgical treatment. The wound healing assay showed that the addition of nondeath-inducing concentrations of GNA significantly reduced the migration ability of U251 glioblastoma cells [47]. Transwell invasion assays showed that GNA decreased the invasive ability of osteosarcoma 143B and HOS cells [48]. GNA could remarkably impede the migration and invasion abilities of bladder cancer cells by inhibiting the nuclear transcription factor-kappa B (NF-κB) signal transduction pathway [42].

Epithelial-to-mesenchymal transition (EMT) denotes the process through which polar epithelial cells transform into stromal cells with migratory capacity, and it represents a crucial mechanism associated with the invasive and migratory capabilities of tumor cells [80]. GNA inhibits invasion, metastasis, and EMT in OCM-1 choroidal melanoma cells and TGF-β1-treated A375 melanoma cells [34], [45]. GNA could also improve EMT by upregulating the expression of lncRNA MEG3, thereby inhibiting melanoma metastasis in vitro and in vivo [81].


#

Gambogenic acid shows synergistic effects in cancers

Combination therapy is a pharmaceutical regimen involving the use of multiple drugs to treat a disease, aiming to achieve higher response rates than a single treatment. 5-FU is a widely used chemotherapeutic drug for various cancer treatments. GNA combined with 5-FU induced cell death caused by apoptotic and necroptotic mechanisms via the ROS-mitochondrial pathway in A549 cells [51]. Erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, was approved by the FDA as a first-line treatment for metastatic NSCLC patients with EGFR mutations. GNA enhances the antitumor activity of erlotinib through the FGFR signaling pathway [52]. BTZ is one of the most widely used agents in the current therapy for multiple myeloma. BTZ and GNA combination treatment resulted in a strong synergistic action against the MM.1S cell line in vitro and in vivo [33].


#

Gambogenic acid reverses the effect on drug resistance in cancers

Drug resistance is a major obstacle in chemotherapy. GNA has been revealed to have a potent inhibitory effect on cell growth in the cisplatin-resistant NSCLC cell line A549/Cis by blocking the cell cycle and inducing apoptosis [64]. In addition, GNA treatment increased the chemosensitivity of breast cancer cells to adriamycin (ADR) by suppressing the PTEN/PI3K/Akt pathway, which led to apoptosis in MCF-7/ADR-resistant cells [54]. GNA may enhance ADR sensitivity and promote ADR-induced apoptosis by inhibiting basal autophagy levels in BEL-7402/ADR HCC cells [70]. Studies have shown that GNA can reverse the multidrug resistance of HepG2/ADR cells by inhibiting the expression of P-glycoprotein (P-gp), which may result from the inhibition of the NF-κB and MAPK pathways [53]. GNA efficiently overcomes erlotinib resistance in NSCLC in vitro and in vivo by inhibiting the FGFR signaling pathway [52].


#

Pharmacokinetic study of gambogenic acid

It is well known that the bioavailability of drugs can vary with different administration methods, and GNA is not an exception. The area under the plasma concentration-time (AUC) values after oral administration are very low compared with those after intravenous injection administration of GNA [82], [83]. GNA and GA were found to have poor absorption, and their pharmacokinetic parameters were similar, suggesting that minor changes in the positions of the substituent groups of the alkyl side chain do not significantly impact the in vivo distribution of the compounds [82]. Moreover, the maximum plasma concentration (Cmax) of GNA increased significantly after processing, which showed a processing influence on the absorption of GNA [84].

Regarding tissue distribution after oral administration, GNA was distributed widely and rapidly in rats and was mainly distributed in the stomach, small intestine, and liver and less distributed in spleen tissues [85]. The cytochrome P450 (CYP450) superfamily is usually considered the most important phase I drug-metabolizing enzyme system. GNA increased the activity of liver CYP1A2, CYP2E1, CYP2C, and CYP3A [86], [87]. Therefore, when GNA is administered with other drugs, potential drug-drug interactions mediated by CYP1A2, CYP2E1, CYP2C, and CYP3A induction should be taken into consideration.


#

Drug delivery system of gambogenic acid in cancers

GNA can inhibit the initiation and progression of many cancers. However, its poor aqueous solubility, short elimination half-life, low bioavailability, and excessive irritation to blood vessels by intravenous administration could reduce its antitumor activities. Therefore, various strategies have been developed to improve GNA bioavailability and enhance its antitumor activities ([Table 4]). These techniques mainly include diverse novel drug delivery systems such as solid lipid nanoparticles [88], long circulation lipid nanoparticles [89], PEGylated liposomes [90], PEGylated niosomes [91], and zein nanoparticles [92].

Table 4 Drug delivery systems of GNA.

Delivery systems

Labels

Cancer type

Cancer cell

IC50 values

Refs.

Nanoparticles

GNA-PDA-FA SA NPs

Breast cancer

4 T1

2.58 µM

[55]

GNA-Zein-PDA NPs

Hepatic carcinoma

HepG2

1.59 µg/mL

[92]

GNA-Zein-NPs

[90]

FA-GNA-MNPs

Cervical cancer

HeLa

[40]

GNA-SLNs

[86]

CRNP-GNA

Hepatic carcinoma

HepG2, Hepa 1 – 6

[96]

Nanostructured lipid carrier

GNA-PEG-NLC

[87]

Nanosuspensions

GNA-NS

Hepatic carcinoma

HepG2

2.22 µM

[91]

Cubs

GNA-Cubs

Hepatic carcinoma

SMMC-7721

4.25 µM

[65]

Nonionic surfactant vesicles

FA-GNA-NISVs

Lung cancer

A549

[67]

PEG-GNA-NISVs

[89]

Micelles

GNA-PAA-b-PCL micelles

Hepatic carcinoma

HepG2

5.04 µg/mL

[93]

GNA-mPEGPCL/mPEG-PLA mixed micelles

Hepatic carcinoma

HepG2

[66]

GNA-PLC micelles

Hepatic carcinoma

HepG2

4.53 µM

[94]

Liposomes

GNA-PEG-LPs

Hepatic carcinoma

HepG2

5.16 µM

[41]

GNA-PEG-LPs

Gastric cancer

SGC-7901

7.66 µM

[41]

GNA-PEG-LPs

Lung cancer

A549

4.83 µM

[41]

GNA-PEI/siRNA-liposome

Hepatic carcinoma

HepG2

3.28 µg/mL

[95]

PEG-GNA-L

[88]

Many researchers have developed GNA-loaded drug delivery systems for the treatment of liver cancer. Luo et al. [65] prepared GNA-loaded cubosomes (GNA-Cubs), which showed higher in vitro cytotoxicity and higher cellular uptake against HCC SMMC-7721 cells. Yuan et al. [93] developed GNA nanosuspensions (GNA-NS) with PVPK30 and PEG2000 as stabilizers. Compared to GNA solution, GNA-NS exerted a much slower in vitro dissolution rate and enhanced cytotoxicity in HCC HepG2 cells. Tang et al. [41] prepared GNA-loaded PEGylated liposomes (GNA-PEG-LPs), which showed enhanced cytotoxicity against lung cancer (A549), gastric cancer (SGC-7901), and HCC (HepG2) cells and induced apoptosis via the mitochondrial pathway in vitro and in vivo. Cheng et al. [92] illustrated that GNA-loaded zein nanoparticles (GNA-Zein-NPs) showed hepatic targeting properties. Zha et al. [94] further proposed polydopamine (PDA)-coated GNA-loaded zein nanoparticles (GNA-Zein-PDA NPs), which had higher inhibitory activity on HepG2 cells. Compared with the above nanocarriers, polymeric micelles have great potential for the solubilization of poorly water-soluble drugs. Liu et al. [95] reported GNA-loaded polymeric micelles based on pH-responsive copolymers, which delivered the highest drug loading efficiency (DLE) and drug loading capacity (DLC) value, enhancing both the cytotoxicity and cellular uptake against HepG2 cells. Lin et al. [66] demonstrated that GNA-loaded mixed polymeric micelles (GNA-MMs) featured a small size and high entrapment efficiency and maintained the cytotoxicity of GNA on HepG2 cells. Wang et al. [96] presented GNA-phospholipid complex (GNA-PLC) micelles. GNA has a higher cytotoxic effect on HepG2 cells after forming GNA-PLC micelles.

Preliminary studies have indicated that liver cancer cells express high levels of vascular endothelial growth factor (VEGF), a key component that fosters the proliferation of vascular endothelial cells. Yu et al. [97] developed a lipopolyplex delivery system composed of anionic liposomes and polyethylenimine complexes to codeliver GNA and VEGF siRNA to HepG2 cells. VEGF-siRNA could mediate VEGF silencing through a lipopolyplex delivery system. The combination of the two drugs increased cell sensitivity and further promoted cell apoptosis. Du et al. [98] demonstrated that GNA was an anti-vascular agent with dual pathways of antiangiogenesis and vascular disruption and reported charge-reversible nanoparticles of GNA for self-augmented accumulation and antitumor efficacy by inducing a positive feedback loop between enhanced tumor vascular permeability and improved accumulation of GNA in tumors.

In addition, Wang et al. [55] encapsulated and stabilized GNA in polydopamine nanoparticles (PDA-NPs), which further modified the surface of the particles with folic acid (FA) and finally coated it with sodium alginate (SA) to form GNA@PDA-FA SA NPs, where FA was used as an active targeting ligand due to its high specific binding with folate receptors, making it easily taken up by 4 T1 cells, while SA was used to prevent the modified nanoparticles from being degraded by the gastric fluid when taken orally. Compared with GNA, GNA@PDA-FA SA NPs exhibited a higher anti-breast cancer therapeutic effect in vivo and in vitro. Huang et al. [40] synthesized GNA-loaded FA-armed magnetic and superparamagnetic nanoparticles (MNPs), which exhibited substantial inhibitory effects in folate receptor-expressing HeLa cancer cells. Lin et al. [67] developed FA-modified nonionic surfactant vesicles (NISVs, niosomes) as carrier systems for the targeted delivery of GNA. The FA-GNA-NISVs prolonged the residence time of GNA in blood circulation, increased accumulation in the lung, and enhanced cytotoxicity against A549 cells by inducing apoptosis.


#
#

Conclusion and Further Directions

Bioactive compounds derived from natural sources are acknowledged for their potential in preventing various human diseases, including cancer. One such compound, GNA, a derivative of GA extracted from G. hanburyi, has demonstrated promising antitumor activity in lung, liver, colorectal, breast, gastric, bladder, and prostate cancers. Research has highlighted its ability to induce apoptosis, necroptosis, ferroptosis, and autophagy as well as cell cycle arrest and inhibition of metastasis across multiple cancer cell lines. GNA has also shown promise in increasing the sensitivity of tumor cells to chemotherapy and in reversing chemotherapy resistance.

However, it should be noted that Pesonen et al. [99] cautioned against the potential negative impact of GNA on cancer therapies due to its ability to induce a strong heat shock response. Furthermore, the information regarding the effects and potential applications of GNA mentioned earlier is primarily based on animal and in vitro experiments, while clinical study is lacking. Therefore, to promote its development for acceptable clinical application, future research should consider focusing primarily on the following aspects: (1) carrying out in-depth research on its antitumor molecular targets and mechanism, (2) performing more in vivo experiments to clarify its safety, (3) expanding the clinical use of GNA through chemical structure modification and combination with other drugs, and (4) conducting more investigations on the drug delivery system of GNA to improve its oral bioavailability. Overall, this study provides a comprehensive review of the antitumor effects and mechanisms, pharmacokinetic properties, and nanotechnology-mediated delivery of GNA and thus serves as a valuable resource for researchers exploring the potential of GNA as a promising candidate for treating various types of cancers.


#

Contributorsʼ Statement

Data collection and analysis: L. Mi, Z. C. Xing, and Y. J. Zhang, T. He; design of the study: L. Mi, Z. H. Li, and W. S. Wu; drafting the manuscript: L. Mi, Z. C. Xing, Y. J. Zhang, T. He, A. P. Su, and T. Wei; critical revision of the manuscript: L. Mi, Z. H. Li, and W. S. Wu.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

This work was financially supported by the Foundation of Sichuan University (2018SCUH0067), the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University (No. ZYJC21033), and the project of West China Hospital (No. 19HXFH010).

  • References

  • 1 GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2018; 392: 1736-1788
  • 2 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209-249
  • 3 Buyel JF. Plants as sources of natural and recombinant anticancer agents. Biotechnol Adv 2018; 36: 506-520
  • 4 Liu Y, Yang S, Wang K, Lu J, Bao X, Wang R, Qiu Y, Wang T, Yu H. Cellular senescence and cancer: Focusing on traditional Chinese medicine and natural products. Cell Prolif 2020; 53: e12894
  • 5 Anantachoke N, Tuchinda P, Kuhakarn C, Pohmakotr M, Reutrakul V. Prenylated caged xanthones: chemistry and biology. Pharm Biol 2012; 50: 78-91
  • 6 Han QB, Xu HX. Caged Garcinia xanthones: Development since 1937. Curr Med Chem 2009; 16: 3775-3796
  • 7 Deng YX, Pan SL, Zhao SY, Wu MQ, Sun ZQ, Chen XH, Shao ZY. Cytotoxic alkoxylated xanthones from the resin of Garcinia hanburyi . Fitoterapia 2012; 83: 1548-1552
  • 8 Deng YX, Guo T, Shao ZY, Xie H, Pan SL. Three new xanthones from the resin of Garcinia hanburyi . Planta Med 2013; 79: 792-796
  • 9 Tao SJ, Guan SH, Wang W, Lu ZQ, Chen GT, Sha N, Yue QX, Liu X, Guo DA. Cytotoxic polyprenylated xanthones from the resin of Garcinia hanburyi . J Nat Prod 2009; 72: 117-124
  • 10 Hahnvajanawong C, Boonyanugomol W, Nasomyon T, Loilome W, Namwat N, Anantachoke N, Tassaneeyakul W, Sripa B, Namwat W, Reutrakul V. Apoptotic activity of caged xanthones from Garcinia hanburyi in cholangiocarcinoma cell lines. World J Gastroenterol 2010; 16: 2235-2243
  • 11 Vichitsakul K, Laowichuwakonnukul K, Soontornworajit B, Poomipark N, Itharat A, Rotkrua P. Anti-proliferation and induction of mitochondria-mediated apoptosis by Garcinia hanburyi resin in colorectal cancer cells. Heliyon 2023; 9: e16411
  • 12 Wang LL, Li ZL, Song DD, Sun L, Pei YH, Jing YK, Hua HM. Two novel triterpenoids with antiproliferative and apoptotic activities in human leukemia cells isolated from the resin of Garcinia hanburyi . Planta Med 2018; 74: 1735-1740
  • 13 Boueroy P, Hahnvajanawong C, Boonmars T, Saensa-Ard S, Anantachoke N, Vaeteewoottacharn K, Reutrakul V. Antitumor effect of forbesione isolated from Garcinia hanburyi on cholangiocarcinoma in vitro and in vivo . Oncol Lett 2016; 12: 4685-4698
  • 14 Wang W, Li Y, Chen Y, Chen H, Zhu P, Xu M, Wang H, Wu M, Yang Z, Hoffman RM, Gu Y. Ethanolic extract of traditional Chinese medicine (TCM) gamboge inhibits colon cancer via the Wnt/β-catenin signaling pathway in an orthotopic mouse model. Anticancer Res 2018; 38: 1917-1925
  • 15 Yang J, He S, Li S, Zhang R, Peng A, Chen L. In vitro and in vivo antiangiogenic activity of caged polyprenylated xanthones isolated from Garcinia hanburyi Hook. f. Molecules 2013; 18: 15305-15313
  • 16 Reutrakul V, Anantachoke N, Pohmakotr M, Jaipetch T, Sophasan S, Yoosook C, Kasisit J, Napaswat C, Santisuk T, Tuchinda P. Cytotoxic and anti-HIV-1 caged xanthones from the resin and fruits of Garcinia hanburyi . Planta Med 2007; 73: 33-40
  • 17 Reutrakul V, Anantachoke N, Pohmakotr M, Jaipetch T, Yoosook C, Kasisit J, Napaswa C, Panthong A, Santisuk T, Prabpai S, Kongsaeree P, Tuchinda P. Anti-HIV-1 and anti-inflammatory lupanes from the leaves, twigs, and resin of Garcinia hanburyi . Planta Med 2009; 76: 368-371
  • 18 Sukpondma Y, Rukachaisirikul V, Phongpaichit S. Antibacterial caged-tetraprenylated xanthones from the fruits of Garcinia hanburyi . Chem Pharm Bull (Tokyo) 2005; 53: 850-852
  • 19 Panthong A, Norkaew P, Kanjanapothi D, Taesotikul T, Anantachoke N, Reutrakul V. Anti-inflammatory, analgesic and antipyretic activities of the extract of gamboge from Garcinia hanburyi Hook. f. J Ethnopharmacol 2007; 111: 335-340
  • 20 Jia B, Li S, Hu X. Recent research on bioactive xanthones from natural medicine: Garcinia hanburyi . AAPS PharmSciTech 2015; 16: 742-758
  • 21 Song JZ, Yip YK, Han QB, Oiao CF, Xu HX. Rapid determination of polyprenylated xanthones in gamboge resin of Garcinia hanburyi by HPLC. J Sep Sci 2007; 30: 304-309
  • 22 Hatami E, Jaggi M, Chauhan SC, Yallapu MM. Gambogic acid: A shining natural compound to nanomedicine for cancer therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874: 188381
  • 23 Liu Y, Chen Y, Lin L, Li H. Gambogic acid as a candidate for cancer therapy: A review. Int J Nanomedicine 2020; 15: 10385-10399
  • 24 Banik K, Harsha C, Bordoloi D, Sailo BL, Sethi G, Leong HC, Arfuso F, Mishra S, Wang L, Kumar AP, Kunnumakkara AB. Therapeutic potential of gambogic acid, a caged xanthone, to target cancer. Cancer Lett 2018; 416: 75-86
  • 25 Yang LJ, Chen Y. New targets for the antitumor activity of gambogic acid in hematologic malignancies. Acta Pharmacol Sin 2013; 34: 191-198
  • 26 Chi Y, Zhan XK, Yu H, Xie GR, Wang ZZ, Xiao W, Wang YG, Xiong FX, Hu JF, Yang L, Cui CX, Wang JW. An open-labeled, randomized, multicenter phase IIa study of gambogic acid injection for advanced malignant tumors. Chin Med J 2013; 26: 1642-1646
  • 27 Asano J, Chiba K, Tada M, Yoshii T. Cytotoxic xanthones from Garcinia hanburyi . Phytochemistry 1996; 41: 815-820
  • 28 Ding Z, Li Y, Tang Z, Song X, Jing F, Wu H, Lu B. Role of gambogenic acid in regulating PI3K/Akt/NF-κB signaling pathways in rat model of acute hepatotoxicity. Biosci Biotechnol Biochem 2021; 85: 520-527
  • 29 Yu X, Zhao Q, Zhang H, Fan C, Zhang X, Xie Q, Xu C, Liu Y, Wu X, Han Q, Zhang H. Gambogenic acid inhibits LPS-simulated inflammatory response by suppressing NF-κB and MAPK in macrophages. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai). Acta Biochim Biophys Sin 2016; 48: 454-461
  • 30 Tao S, Yang L, Wu C, Hu Y, Guo F, Ren Q, Ma L, Fu P. Gambogenic acid alleviates kidney fibrosis via epigenetic inhibition of EZH2 to regulate Smad7-dependent mechanism. Phytomedicine 2022; 106: 154390
  • 31 Zeng J, Huang TY, Wang ZZ, Gong YF, Liu XC, Zhang XM, Huang XY. Scar-reducing effects of gambogenic acid on skin wounds in rabbit ears. Int Immunopharmacol 2021; 90: 107200
  • 32 Wang X, Cao W, Zhang J, Yan M, Xu Q, Wu X, Wan L, Zhang Z, Zhang C, Qin X, Xiao M, Ye D, Liu Y, Han Z, Wang S, Mao L, Wei W, Chen W. A covalently bound inhibitor triggers EZH2 degradation through CHIP-mediated ubiquitination. EMBO J 2017; 36: 1243-1260
  • 33 Chen R, Zhang H, Liu P, Wu X, Chen B. Gambogenic acid synergistically potentiates bortezomib-induced apoptosis in multiple myeloma. J Cancer 2017; 8: 839-851
  • 34 Li F, Wang Y, Yan Y. Gambogenic acid induces cell growth inhibition, cell cycle arrest and metastasis inhibition in choroidal melanoma in a dose-dependent manner. Exp Ther Med 2017; 13: 2456-2462
  • 35 Li Q, Cheng H, Zhu G, Yang L, Zhou A, Wang X, Fang N, Xia L, Su J, Wang M, Peng D, Xu Q. Gambogenic acid inhibits proliferation of A549 cells through apoptosis-inducing and cell cycle arresting. Biol Pharm Bull 2010; 33: 415-420
  • 36 Zhao Q, Zhong J, Bi Y, Liu Y, Liu Y, Guo J, Pan L, Tan Y, Yu X. Gambogenic acid induces Noxa-mediated apoptosis in colorectal cancer through ROS-dependent activation of IRE1α/JNK. Phytomedicine 2020; 78: 153306
  • 37 Huang T, Zhang H, Wang X, Xu L, Jia J, Zhu X. Gambogenic acid inhibits the proliferation of small-cell lung cancer cells by arresting the cell cycle and inducing apoptosis. Oncol Rep 2019; 41: 1700-1706
  • 38 Yu XJ, Han QB, Wen ZS, Ma L, Gao J, Zhou GB. Gambogenic acid induces G1 arrest via GSK3β-dependent cyclin D1 degradation and triggers autophagy in lung cancer cells. Cancer Lett 2012; 322: 185-194
  • 39 Zhou J, Luo YH, Wang JR, Lu BB, Wang KM, Tian Y. Gambogenic acid induction of apoptosis in a breast cancer cell line. Asian Pac J Cancer Prev 2013; 14: 7601-7605
  • 40 Huang P, Yang LL, Wang CY, Chen JP, Wang SS, Wang SJ, Chen YJ, Wang DL, Huang HP. Preparation and characterization of folate targeting magnetic nanomedicine loaded with gambogenic acid. J Nanosci Nanotechnol 2015; 15: 4774-4783
  • 41 Tang X, Sun J, Ge T, Zhang K, Gui Q, Zhang S, Chen W. PEGylated liposomes as delivery systems for gambogenic acid: Characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces 2018; 172: 26-36
  • 42 Zhou S, Zhao N, Wang J. Gambogenic acid suppresses bladder cancer cells growth and metastasis by regulating NF-κB signaling. Chem Biol Drug Des 2020; 96: 1272-1279
  • 43 Wu J, Wang D, Zhou J, Li J, Xie R, Li Y, Huang J, Liu B, Qiu J. Gambogenic acid induces apoptosis and autophagy through ROS-mediated endoplasmic reticulum stress via JNK pathway in prostate cancer cells. Phytother Res 2023; 37: 310-328
  • 44 Yan F, Wang M, Li J, Cheng H, Su J, Wang X, Wu H, Xia L, Li X, Chang HC, Li Q. Gambogenic acid induced mitochondrial-dependent apoptosis and referred to phospho-Erk1/2 and phospho-p 38 MAPK in human hepatoma HepG2 cells. Environ Toxicol Pharmacol 2012; 33: 181-190
  • 45 Wang M, Li S, Wang Y, Cheng H, Su J, Li Q. Gambogenic acid induces ferroptosis in melanoma cells undergoing epithelial-to-mesenchymal transition. Toxicol Appl Pharmacol 2020; 401: 115110
  • 46 Yan F, Wang M, Chen H, Su J, Wang X, Wang F, Xia L, Li Q. Gambogenic acid mediated apoptosis through the mitochondrial oxidative stress and inactivation of Akt signaling pathway in human nasopharyngeal carcinoma CNE-1 cells. Eur J Pharmacol 2011; 652: 23-32
  • 47 Chen HB, Zhou LZ, Mei L, Shi XJ, Wang XS, Li QL, Huang L. Gambogenic acid-induced time- and dose-dependent growth inhibition and apoptosis involving Akt pathway inactivation in U251 glioblastoma cells. J Nat Med 2012; 66: 62-69
  • 48 Liu Z, Wang X, Li J, Yang X, Huang J, Ji C, Li X, Li L, Zhou J, Hu Y. Gambogenic acid induces cell death in human osteosarcoma through altering iron metabolism, disturbing the redox balance, and activating the P53 signaling pathway. Chem Biol Interact 2023; 382: 110602
  • 49 Han QB, Wang YL, Yang L, Tso TF, Qiao CF, Song JZ, Xu LJ, Chen SL, Yang DJ, Xu HX. Cytotoxic polyprenylated xanthones from the resin of Garcinia hanburyi . Chem Pharm Bull (Tokyo) 2006; 54: 265-267
  • 50 Yu XJ, Zhao Q, Wang XB, Zhang JX, Wang XB. Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents. Oncol Rep 2016; 36: 3611-3618
  • 51 Su J, Cheng H, Zhang D, Wang M, Xie C, Hu Y, Chang HW, Li Q. Synergistic effects of 5-fluorouracil and gambogenic acid on A549 cells: Activation of cell death caused by apoptotic and necroptotic mechanisms via the ROS-mitochondria pathway. Biol Pharm Bull 2014; 37: 1259-1268
  • 52 Xu L, Meng X, Xu N, Fu W, Tan H, Zhang L, Zhou Q, Qian J, Tu S, Li X, Lao Y, Xu H. Gambogenic acid inhibits fibroblast growth factor receptor signaling pathway in erlotinib-resistant non-small-cell lung cancer and suppresses patient-derived xenograft growth. Cell Death Dis 2018; 9: 262
  • 53 Xu Q, Guo J, Chen W. Gambogenic acid reverses P-glycoprotein mediated multidrug resistance in HepG2/ADR cells and its underlying mechanism. Biochem Biophys Res Commun 2019; 508: 882-888
  • 54 He Y, Ding J, Lin Y, Li J, Shi Y, Wang J, Zhu Y, Wang K, Hu X. Gambogenic acid alters chemosensitivity of breast cancer cells to adriamycin. BMC Complement Altern Med 2015; 15: 181
  • 55 Wang B, Yuan T, Zha L, Liu Y, Chen W, Zhang C, Bao Y, Dong Q. Oral delivery of gambogenic acid by functional polydopamine nanoparticles for targeted tumor therapy. Mol Pharm 2021; 18: 1470-1479
  • 56 Okada S, Vaeteewoottacharn K, Kariya R. Application of highly immunocompromised mice for the establishment of Patient-Derived Xenograft (PDX) models. Cells 2019; 8: 889
  • 57 Liu C, Xu J, Guo C, Chen X, Qian C, Zhang X, Zhou P, Yang Y. Gambogenic acid induces endoplasmic reticulum stress in colorectal cancer via the aurora a pathway. Front Cell Dev Biol 2021; 9: 736350
  • 58 Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med 2016; 22: 128-134
  • 59 Huang J, Gou H, Yao J, Yi K, Jin Z, Matsuoka M, Zhao T. The noncanonical role of EZH2 in cancer. Cancer Sci 2021; 112: 1376-1382
  • 60 Zeng J, Zhang J, Sun Y, Wang J, Ren C, Banerjee S, Ouyang L, Wang Y. Targeting EZH2 for cancer therapy: From current progress to novel strategies. Eur J Med Chem 2022; 238: 114419
  • 61 Eich ML, Athar M, Ferguson JE, Varambally S. EZH2-targeted therapies in cancer: Hype or a reality. Cancer Res 2020; 80: 5449-5458
  • 62 Liu Y, Yang Q. The roles of EZH2 in cancer and its inhibitors. Med Oncol 2023; 40: 167
  • 63 Montalto FI, De Amicis F. Cyclin D1 in cancer: A molecular connection for cell cycle control, adhesion and invasion in tumor and stroma. Cells 2020; 9: 2648
  • 64 Shen D, Wang Y, Niu H, Liu C. Gambogenic acid exerts anticancer effects in cisplatin-resistant non-small cell lung cancer cells. Mol Med Rep 2020; 21: 1267-1275
  • 65 Luo Q, Lin T, Zhang CY, Zhu T, Wang L, Ji Z, Jia B, Ge T, Peng D, Chen W. A novel glyceryl monoolein-bearing cubosomes for gambogenic acid: Preparation, cytotoxicity and intracellular uptake. Int J Pharm 2015; 49: 30-39
  • 66 Lin TY, Zhu TT, Xun Y, Tao YS, Yang YQ, Xie JL, Zhang XM, Chen SX, Ding BJ, Chen WD. A novel drug delivery system of mixed micelles based on poly(ethylene glycol)-poly(lactide) and poly(ethylene glycol)-poly(ε-caprolactone) for gambogenic acid. Kaohsiung J Med Sci 2019; 35: 757-764
  • 67 Lin TY, Chang JL, Xun Y, Zhao Y, Peng W, Yang W, Ding BJ, Chen WD. Folic acid-modified nonionic surfactant vesicles for gambogenic acid targeting: Preparation, characterization, and in vitro and in vivo evaluation. Kaohsiung J Med Sci 2020; 36: 344-353
  • 68 Cheng H, Su JJ, Peng JY, Wang M, Wang XC, Yan FG, Wang XS, Li QL. Gambogenic acid inhibits proliferation of A549 cells through apoptosis inducing through up-regulation of the p 38 MAPK cascade. J Asian Nat Prod Res 2011; 13: 993-1002
  • 69 Kashyap D, Garg VK, Goel N. Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv Protein Chem Struct Biol 2021; 125: 73-120
  • 70 Wang M, Zhan F, Cheng H, Li Q. Gambogenic acid inhibits basal autophagy of drug-resistant hepatoma cells and improves its sensitivity to adriamycin. Biol Pharm Bull 2022; 45: 63-70
  • 71 Zeeshan HM, Lee GH, Kim HR, Chae HJ. Endoplasmic reticulum stress and associated ROS. Int J Mol Sci 2016; 17: 327
  • 72 Su J, Xu T, Jiang G, Hou M, Liang M, Cheng H, Li Q. Gambogenic acid triggers apoptosis in human nasopharyngeal carcinoma CNE-2Z cells by activating volume-sensitive outwardly rectifying chloride channel. Fitoterapia 2019; 133: 150-158
  • 73 Wang M, Cheng H, Wu H, Liu C, Li S, Li B, Su J, Luo S, Li Q. Gambogenic acid antagonizes the expression and effects of long non-coding RNA NEAT1 and triggers autophagy and ferroptosis in melanoma. Biomed Pharmacother 2022; 154: 113636
  • 74 Mei W, Dong C, Hui C, Bin L, Fenggen Y, Jingjing S, Cheng P, Meiling S, Yawen H, Xiaoshan W, Guanghui W, Zhiwu C, Qinglin L. Gambogenic acid kills lung cancer cells through aberrant autophagy. PLoS One 2014; 9: e83604
  • 75 Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, Cheng H, Jin K, Ni Q, Yu X, Liu C. The role of necroptosis in cancer biology and therapy. Mol Cancer 2019; 18: 100
  • 76 Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W, Wang J. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med 2019; 23: 4900-4912
  • 77 Ma X, Xu M, Zhang X, Wang X, Su K, Xu Z, Wang X, Yang Y. Gambogenic acid inhibits proliferation and ferroptosis by targeting the miR‐1291/FOXA2 and AMPKα/SLC7A11/GPX4 axis in colorectal cancer. Cell Biol Int 2023; 47: 1813-1824
  • 78 Wang X, Chen Y, Han QB, Chan CY, Wang H, Liu Z, Cheng CH, Yew DT, Lin MCM, He ML, Xu HX, Sung JYY, Kung HF. Proteomic identification of molecular targets of gambogic acid: Role of stathmin in hepatocellular carcinoma. Proteomics 2009; 9: 242-253
  • 79 Liu P, Wu X, Dai L, Ge Z, Gao C, Zhang H, Wang F, Zhang X, Chen B. Gambogenic acid exerts antitumor activity in hypoxic multiple myeloma cells by regulation of miR-21. J Cancer 2017; 8: 3278-3286
  • 80 Giannelli G, Koudelkova P, Dituri F, Mikulits W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol 2016; 65: 798-808
  • 81 Wang M, Tu Y, Liu C, Cheng H, Zhang M, Li Q. Gambogenic acid inhibits invasion and metastasis of melanoma through regulation of lncRNA MEG3. Biol Pharm Bull 2023; 46: 1385-1393
  • 82 Hua X, Liang C, Dong L, Qu X, Zhao T. Simultaneous determination and pharmacokinetic study of gambogic acid and gambogenic acid in rat plasma after oral administration of Garcinia hanburyi extracts by LC-MS/MS. Biomed Chromatogr 2015; 29: 545-551
  • 83 Chen JP, Wang DL, Yang LL, Wang CY, Wang SS. Ultra-high-performance liquid chromatography tandem mass spectrometry method for the determination of gambogenic acid in dog plasma and its application to a pharmacokinetic study. Biomed Chromatogr 2014; 28: 1854-1859
  • 84 Pan LY, Wang YS, Liu XH, Wang N, Xu W, Xiu YF. Pharmacokinetic comparison of five xanthones in rat plasma after oral administration of crude and processed Garcinia hanburyi extracts. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1126 – 1127: 121737
  • 85 Pan L, Xu M, Wang N, Jia Y, Xiu Y. Determination and tissue distribution comparisons of five xanthones after orally administering crude and processed gamboge. Biomed Chromatogr 2023; 37: e5516
  • 86 Sun J, Tang X, Xu Q, Ge T, Peng D, Chen W. Effect of gambogenic acid on cytochrome P450 1A2, 2B1 and 2E1, and constitutive androstane receptor in rats. Eur J Drug Metab Pharmacokinet 2018; 43: 655-664
  • 87 Sun J, Pang M, Tang X, Xu Q, Peng D, Chen W. Antitumor effect of gambogenic acid and its effect on CYP2C and CYP3A after oral administration. Chem Pharm Bull (Tokyo) 2023; 71: 334-341
  • 88 Huang X, Chen YJ, Peng DY, Li QL, Wang XS, Wang DL, Chen WD. Solid lipid nanoparticles as delivery systems for gambogenic acid. Colloids Surf B Biointerfaces 2013; 102: 391-397
  • 89 Lin T, Huang X, Wang Y, Zhu T, Luo Q, Wang X, Zhou K, Cheng H, Peng D, Chen W. Long circulation nanostructured lipid carriers for gambogenic acid: Formulation design, characterization, and pharmacokinetic. Xenobiotica 2017; 47: 793-799
  • 90 Wang F, Ye X, Wu Y, Wang H, Sheng C, Peng D, Chen W. Time interval of two injections and first-dose dependent of accelerated blood clearance phenomenon induced by PEGylated liposomal gambogenic acid: The contribution of PEG-specific IgM. J Pharm Sci 2019; 108: 641-651
  • 91 Lin T, Fang Q, Peng D, Huang X, Zhu T, Luo Q, Zhou K, Chen W. PEGylated non-ionic surfactant vesicles as drug delivery systems for gambogenic acid. Drug Deliv 2013; 20: 277-284
  • 92 Cheng W, Wang B, Zhang C, Dong Q, Qian J, Zha L, Chen W, Hong L. Preparation and preliminary pharmacokinetics study of GNA-loaded zein nanoparticles. J Pharm Pharmacol 2019; 71: 1626-1634
  • 93 Yuan H, Li X, Zhang C, Pan W, Liang Y, Chen Y, Chen W, Liu L, Wang X. Nanosuspensions as delivery system for gambogenic acid: Characterization and in vitro/in vivo evaluation. Drug Deliv 2016; 23: 2772-2779
  • 94 Zha L, Qian J, Wang B, Liu H, Zhang C, Dong Q, Chen W, Hong L. In vitro/in vivo evaluation of pH-sensitive gambogenic acid loaded zein nanoparticles with polydopamine coating. Int J Pharm 2020; 587: 119665
  • 95 Liu H, Chen H, Cao F, Peng D, Chen W, Zhang C. Amphiphilic block copolymer poly (Acrylic Acid)-B-polycaprolactone as a novel pH-sensitive nanocarrier for anticancer drugs delivery: In-vitro and in-vivo evaluation. Polymers (Basel) 2019; 11: 820
  • 96 Wang B, Cheng W, Zhang C, Bao Y, Zha L, Qian J, Hong L, Chen W. Self-assembled micelles based on gambogenic acid-phospholipid complex for sustained-release drug delivery. J Microencapsul 2019; 36: 566-575
  • 97 Yu Q, Zhang B, Zhou Y, Ge Q, Jiali C, Chen Y, Zhang K, Peng D, Chen W. Co-delivery of gambogenic acid and VEGF-siRNA with anionic liposome and polyethylenimine complexes to HepG2 cells. J Liposome Res 2019; 29: 322-331
  • 98 Du M, Geng T, Yu R, Song G, Cheng H, Cao Y, He W, Haleem A, Li Q, Hu R, Chen S. Smart anti-vascular nanoagent induces positive feedback loop for self-augmented tumor accumulation. J Control Release 2023; 356: 595-609
  • 99 Pesonen L, Svartsjö S, Bäck V, de Thonel A, Mezger V, Sabéran-Djoneidi D, Roos-Mattjus P. Gambogic acid and gambogenic acid induce a thiol-dependent heat shock response and disrupt the interaction between HSP90 and HSF1 or HSF2. Cell Stress Chaperones 2021; 26: 819-833

Correspondence

Dr. Zhihui Li
Division of Thyroid Surgery
Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease
Frontiers Science Center for Disease-Related Molecular Network
West China Hospital
Sichuan University
No. 2222 Xinchuan Road
610000 Chengdu
China   
Phone: + 8 60 28 85 42 24 67   
Fax: + 8 60 28 85 42 24 67   

 

Prof. Wenshuang Wu
Division of Thyroid Surgery
Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease
Frontiers Science Center for Disease-Related Molecular Network
West China Hospital
Sichuan University
No. 2222 Xinchuan Road
610000 Chengdu
China   
Phone: + 8 60 28 85 42 24 67   
Fax: + 8 60 28 85 42 24 67   

Publication History

Received: 24 November 2023

Accepted after revision: 31 January 2024

Accepted Manuscript online:
31 January 2024

Article published online:
29 February 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: A systematic analysis for the global burden of disease study 2017. Lancet 2018; 392: 1736-1788
  • 2 Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209-249
  • 3 Buyel JF. Plants as sources of natural and recombinant anticancer agents. Biotechnol Adv 2018; 36: 506-520
  • 4 Liu Y, Yang S, Wang K, Lu J, Bao X, Wang R, Qiu Y, Wang T, Yu H. Cellular senescence and cancer: Focusing on traditional Chinese medicine and natural products. Cell Prolif 2020; 53: e12894
  • 5 Anantachoke N, Tuchinda P, Kuhakarn C, Pohmakotr M, Reutrakul V. Prenylated caged xanthones: chemistry and biology. Pharm Biol 2012; 50: 78-91
  • 6 Han QB, Xu HX. Caged Garcinia xanthones: Development since 1937. Curr Med Chem 2009; 16: 3775-3796
  • 7 Deng YX, Pan SL, Zhao SY, Wu MQ, Sun ZQ, Chen XH, Shao ZY. Cytotoxic alkoxylated xanthones from the resin of Garcinia hanburyi . Fitoterapia 2012; 83: 1548-1552
  • 8 Deng YX, Guo T, Shao ZY, Xie H, Pan SL. Three new xanthones from the resin of Garcinia hanburyi . Planta Med 2013; 79: 792-796
  • 9 Tao SJ, Guan SH, Wang W, Lu ZQ, Chen GT, Sha N, Yue QX, Liu X, Guo DA. Cytotoxic polyprenylated xanthones from the resin of Garcinia hanburyi . J Nat Prod 2009; 72: 117-124
  • 10 Hahnvajanawong C, Boonyanugomol W, Nasomyon T, Loilome W, Namwat N, Anantachoke N, Tassaneeyakul W, Sripa B, Namwat W, Reutrakul V. Apoptotic activity of caged xanthones from Garcinia hanburyi in cholangiocarcinoma cell lines. World J Gastroenterol 2010; 16: 2235-2243
  • 11 Vichitsakul K, Laowichuwakonnukul K, Soontornworajit B, Poomipark N, Itharat A, Rotkrua P. Anti-proliferation and induction of mitochondria-mediated apoptosis by Garcinia hanburyi resin in colorectal cancer cells. Heliyon 2023; 9: e16411
  • 12 Wang LL, Li ZL, Song DD, Sun L, Pei YH, Jing YK, Hua HM. Two novel triterpenoids with antiproliferative and apoptotic activities in human leukemia cells isolated from the resin of Garcinia hanburyi . Planta Med 2018; 74: 1735-1740
  • 13 Boueroy P, Hahnvajanawong C, Boonmars T, Saensa-Ard S, Anantachoke N, Vaeteewoottacharn K, Reutrakul V. Antitumor effect of forbesione isolated from Garcinia hanburyi on cholangiocarcinoma in vitro and in vivo . Oncol Lett 2016; 12: 4685-4698
  • 14 Wang W, Li Y, Chen Y, Chen H, Zhu P, Xu M, Wang H, Wu M, Yang Z, Hoffman RM, Gu Y. Ethanolic extract of traditional Chinese medicine (TCM) gamboge inhibits colon cancer via the Wnt/β-catenin signaling pathway in an orthotopic mouse model. Anticancer Res 2018; 38: 1917-1925
  • 15 Yang J, He S, Li S, Zhang R, Peng A, Chen L. In vitro and in vivo antiangiogenic activity of caged polyprenylated xanthones isolated from Garcinia hanburyi Hook. f. Molecules 2013; 18: 15305-15313
  • 16 Reutrakul V, Anantachoke N, Pohmakotr M, Jaipetch T, Sophasan S, Yoosook C, Kasisit J, Napaswat C, Santisuk T, Tuchinda P. Cytotoxic and anti-HIV-1 caged xanthones from the resin and fruits of Garcinia hanburyi . Planta Med 2007; 73: 33-40
  • 17 Reutrakul V, Anantachoke N, Pohmakotr M, Jaipetch T, Yoosook C, Kasisit J, Napaswa C, Panthong A, Santisuk T, Prabpai S, Kongsaeree P, Tuchinda P. Anti-HIV-1 and anti-inflammatory lupanes from the leaves, twigs, and resin of Garcinia hanburyi . Planta Med 2009; 76: 368-371
  • 18 Sukpondma Y, Rukachaisirikul V, Phongpaichit S. Antibacterial caged-tetraprenylated xanthones from the fruits of Garcinia hanburyi . Chem Pharm Bull (Tokyo) 2005; 53: 850-852
  • 19 Panthong A, Norkaew P, Kanjanapothi D, Taesotikul T, Anantachoke N, Reutrakul V. Anti-inflammatory, analgesic and antipyretic activities of the extract of gamboge from Garcinia hanburyi Hook. f. J Ethnopharmacol 2007; 111: 335-340
  • 20 Jia B, Li S, Hu X. Recent research on bioactive xanthones from natural medicine: Garcinia hanburyi . AAPS PharmSciTech 2015; 16: 742-758
  • 21 Song JZ, Yip YK, Han QB, Oiao CF, Xu HX. Rapid determination of polyprenylated xanthones in gamboge resin of Garcinia hanburyi by HPLC. J Sep Sci 2007; 30: 304-309
  • 22 Hatami E, Jaggi M, Chauhan SC, Yallapu MM. Gambogic acid: A shining natural compound to nanomedicine for cancer therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874: 188381
  • 23 Liu Y, Chen Y, Lin L, Li H. Gambogic acid as a candidate for cancer therapy: A review. Int J Nanomedicine 2020; 15: 10385-10399
  • 24 Banik K, Harsha C, Bordoloi D, Sailo BL, Sethi G, Leong HC, Arfuso F, Mishra S, Wang L, Kumar AP, Kunnumakkara AB. Therapeutic potential of gambogic acid, a caged xanthone, to target cancer. Cancer Lett 2018; 416: 75-86
  • 25 Yang LJ, Chen Y. New targets for the antitumor activity of gambogic acid in hematologic malignancies. Acta Pharmacol Sin 2013; 34: 191-198
  • 26 Chi Y, Zhan XK, Yu H, Xie GR, Wang ZZ, Xiao W, Wang YG, Xiong FX, Hu JF, Yang L, Cui CX, Wang JW. An open-labeled, randomized, multicenter phase IIa study of gambogic acid injection for advanced malignant tumors. Chin Med J 2013; 26: 1642-1646
  • 27 Asano J, Chiba K, Tada M, Yoshii T. Cytotoxic xanthones from Garcinia hanburyi . Phytochemistry 1996; 41: 815-820
  • 28 Ding Z, Li Y, Tang Z, Song X, Jing F, Wu H, Lu B. Role of gambogenic acid in regulating PI3K/Akt/NF-κB signaling pathways in rat model of acute hepatotoxicity. Biosci Biotechnol Biochem 2021; 85: 520-527
  • 29 Yu X, Zhao Q, Zhang H, Fan C, Zhang X, Xie Q, Xu C, Liu Y, Wu X, Han Q, Zhang H. Gambogenic acid inhibits LPS-simulated inflammatory response by suppressing NF-κB and MAPK in macrophages. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai). Acta Biochim Biophys Sin 2016; 48: 454-461
  • 30 Tao S, Yang L, Wu C, Hu Y, Guo F, Ren Q, Ma L, Fu P. Gambogenic acid alleviates kidney fibrosis via epigenetic inhibition of EZH2 to regulate Smad7-dependent mechanism. Phytomedicine 2022; 106: 154390
  • 31 Zeng J, Huang TY, Wang ZZ, Gong YF, Liu XC, Zhang XM, Huang XY. Scar-reducing effects of gambogenic acid on skin wounds in rabbit ears. Int Immunopharmacol 2021; 90: 107200
  • 32 Wang X, Cao W, Zhang J, Yan M, Xu Q, Wu X, Wan L, Zhang Z, Zhang C, Qin X, Xiao M, Ye D, Liu Y, Han Z, Wang S, Mao L, Wei W, Chen W. A covalently bound inhibitor triggers EZH2 degradation through CHIP-mediated ubiquitination. EMBO J 2017; 36: 1243-1260
  • 33 Chen R, Zhang H, Liu P, Wu X, Chen B. Gambogenic acid synergistically potentiates bortezomib-induced apoptosis in multiple myeloma. J Cancer 2017; 8: 839-851
  • 34 Li F, Wang Y, Yan Y. Gambogenic acid induces cell growth inhibition, cell cycle arrest and metastasis inhibition in choroidal melanoma in a dose-dependent manner. Exp Ther Med 2017; 13: 2456-2462
  • 35 Li Q, Cheng H, Zhu G, Yang L, Zhou A, Wang X, Fang N, Xia L, Su J, Wang M, Peng D, Xu Q. Gambogenic acid inhibits proliferation of A549 cells through apoptosis-inducing and cell cycle arresting. Biol Pharm Bull 2010; 33: 415-420
  • 36 Zhao Q, Zhong J, Bi Y, Liu Y, Liu Y, Guo J, Pan L, Tan Y, Yu X. Gambogenic acid induces Noxa-mediated apoptosis in colorectal cancer through ROS-dependent activation of IRE1α/JNK. Phytomedicine 2020; 78: 153306
  • 37 Huang T, Zhang H, Wang X, Xu L, Jia J, Zhu X. Gambogenic acid inhibits the proliferation of small-cell lung cancer cells by arresting the cell cycle and inducing apoptosis. Oncol Rep 2019; 41: 1700-1706
  • 38 Yu XJ, Han QB, Wen ZS, Ma L, Gao J, Zhou GB. Gambogenic acid induces G1 arrest via GSK3β-dependent cyclin D1 degradation and triggers autophagy in lung cancer cells. Cancer Lett 2012; 322: 185-194
  • 39 Zhou J, Luo YH, Wang JR, Lu BB, Wang KM, Tian Y. Gambogenic acid induction of apoptosis in a breast cancer cell line. Asian Pac J Cancer Prev 2013; 14: 7601-7605
  • 40 Huang P, Yang LL, Wang CY, Chen JP, Wang SS, Wang SJ, Chen YJ, Wang DL, Huang HP. Preparation and characterization of folate targeting magnetic nanomedicine loaded with gambogenic acid. J Nanosci Nanotechnol 2015; 15: 4774-4783
  • 41 Tang X, Sun J, Ge T, Zhang K, Gui Q, Zhang S, Chen W. PEGylated liposomes as delivery systems for gambogenic acid: Characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces 2018; 172: 26-36
  • 42 Zhou S, Zhao N, Wang J. Gambogenic acid suppresses bladder cancer cells growth and metastasis by regulating NF-κB signaling. Chem Biol Drug Des 2020; 96: 1272-1279
  • 43 Wu J, Wang D, Zhou J, Li J, Xie R, Li Y, Huang J, Liu B, Qiu J. Gambogenic acid induces apoptosis and autophagy through ROS-mediated endoplasmic reticulum stress via JNK pathway in prostate cancer cells. Phytother Res 2023; 37: 310-328
  • 44 Yan F, Wang M, Li J, Cheng H, Su J, Wang X, Wu H, Xia L, Li X, Chang HC, Li Q. Gambogenic acid induced mitochondrial-dependent apoptosis and referred to phospho-Erk1/2 and phospho-p 38 MAPK in human hepatoma HepG2 cells. Environ Toxicol Pharmacol 2012; 33: 181-190
  • 45 Wang M, Li S, Wang Y, Cheng H, Su J, Li Q. Gambogenic acid induces ferroptosis in melanoma cells undergoing epithelial-to-mesenchymal transition. Toxicol Appl Pharmacol 2020; 401: 115110
  • 46 Yan F, Wang M, Chen H, Su J, Wang X, Wang F, Xia L, Li Q. Gambogenic acid mediated apoptosis through the mitochondrial oxidative stress and inactivation of Akt signaling pathway in human nasopharyngeal carcinoma CNE-1 cells. Eur J Pharmacol 2011; 652: 23-32
  • 47 Chen HB, Zhou LZ, Mei L, Shi XJ, Wang XS, Li QL, Huang L. Gambogenic acid-induced time- and dose-dependent growth inhibition and apoptosis involving Akt pathway inactivation in U251 glioblastoma cells. J Nat Med 2012; 66: 62-69
  • 48 Liu Z, Wang X, Li J, Yang X, Huang J, Ji C, Li X, Li L, Zhou J, Hu Y. Gambogenic acid induces cell death in human osteosarcoma through altering iron metabolism, disturbing the redox balance, and activating the P53 signaling pathway. Chem Biol Interact 2023; 382: 110602
  • 49 Han QB, Wang YL, Yang L, Tso TF, Qiao CF, Song JZ, Xu LJ, Chen SL, Yang DJ, Xu HX. Cytotoxic polyprenylated xanthones from the resin of Garcinia hanburyi . Chem Pharm Bull (Tokyo) 2006; 54: 265-267
  • 50 Yu XJ, Zhao Q, Wang XB, Zhang JX, Wang XB. Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents. Oncol Rep 2016; 36: 3611-3618
  • 51 Su J, Cheng H, Zhang D, Wang M, Xie C, Hu Y, Chang HW, Li Q. Synergistic effects of 5-fluorouracil and gambogenic acid on A549 cells: Activation of cell death caused by apoptotic and necroptotic mechanisms via the ROS-mitochondria pathway. Biol Pharm Bull 2014; 37: 1259-1268
  • 52 Xu L, Meng X, Xu N, Fu W, Tan H, Zhang L, Zhou Q, Qian J, Tu S, Li X, Lao Y, Xu H. Gambogenic acid inhibits fibroblast growth factor receptor signaling pathway in erlotinib-resistant non-small-cell lung cancer and suppresses patient-derived xenograft growth. Cell Death Dis 2018; 9: 262
  • 53 Xu Q, Guo J, Chen W. Gambogenic acid reverses P-glycoprotein mediated multidrug resistance in HepG2/ADR cells and its underlying mechanism. Biochem Biophys Res Commun 2019; 508: 882-888
  • 54 He Y, Ding J, Lin Y, Li J, Shi Y, Wang J, Zhu Y, Wang K, Hu X. Gambogenic acid alters chemosensitivity of breast cancer cells to adriamycin. BMC Complement Altern Med 2015; 15: 181
  • 55 Wang B, Yuan T, Zha L, Liu Y, Chen W, Zhang C, Bao Y, Dong Q. Oral delivery of gambogenic acid by functional polydopamine nanoparticles for targeted tumor therapy. Mol Pharm 2021; 18: 1470-1479
  • 56 Okada S, Vaeteewoottacharn K, Kariya R. Application of highly immunocompromised mice for the establishment of Patient-Derived Xenograft (PDX) models. Cells 2019; 8: 889
  • 57 Liu C, Xu J, Guo C, Chen X, Qian C, Zhang X, Zhou P, Yang Y. Gambogenic acid induces endoplasmic reticulum stress in colorectal cancer via the aurora a pathway. Front Cell Dev Biol 2021; 9: 736350
  • 58 Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med 2016; 22: 128-134
  • 59 Huang J, Gou H, Yao J, Yi K, Jin Z, Matsuoka M, Zhao T. The noncanonical role of EZH2 in cancer. Cancer Sci 2021; 112: 1376-1382
  • 60 Zeng J, Zhang J, Sun Y, Wang J, Ren C, Banerjee S, Ouyang L, Wang Y. Targeting EZH2 for cancer therapy: From current progress to novel strategies. Eur J Med Chem 2022; 238: 114419
  • 61 Eich ML, Athar M, Ferguson JE, Varambally S. EZH2-targeted therapies in cancer: Hype or a reality. Cancer Res 2020; 80: 5449-5458
  • 62 Liu Y, Yang Q. The roles of EZH2 in cancer and its inhibitors. Med Oncol 2023; 40: 167
  • 63 Montalto FI, De Amicis F. Cyclin D1 in cancer: A molecular connection for cell cycle control, adhesion and invasion in tumor and stroma. Cells 2020; 9: 2648
  • 64 Shen D, Wang Y, Niu H, Liu C. Gambogenic acid exerts anticancer effects in cisplatin-resistant non-small cell lung cancer cells. Mol Med Rep 2020; 21: 1267-1275
  • 65 Luo Q, Lin T, Zhang CY, Zhu T, Wang L, Ji Z, Jia B, Ge T, Peng D, Chen W. A novel glyceryl monoolein-bearing cubosomes for gambogenic acid: Preparation, cytotoxicity and intracellular uptake. Int J Pharm 2015; 49: 30-39
  • 66 Lin TY, Zhu TT, Xun Y, Tao YS, Yang YQ, Xie JL, Zhang XM, Chen SX, Ding BJ, Chen WD. A novel drug delivery system of mixed micelles based on poly(ethylene glycol)-poly(lactide) and poly(ethylene glycol)-poly(ε-caprolactone) for gambogenic acid. Kaohsiung J Med Sci 2019; 35: 757-764
  • 67 Lin TY, Chang JL, Xun Y, Zhao Y, Peng W, Yang W, Ding BJ, Chen WD. Folic acid-modified nonionic surfactant vesicles for gambogenic acid targeting: Preparation, characterization, and in vitro and in vivo evaluation. Kaohsiung J Med Sci 2020; 36: 344-353
  • 68 Cheng H, Su JJ, Peng JY, Wang M, Wang XC, Yan FG, Wang XS, Li QL. Gambogenic acid inhibits proliferation of A549 cells through apoptosis inducing through up-regulation of the p 38 MAPK cascade. J Asian Nat Prod Res 2011; 13: 993-1002
  • 69 Kashyap D, Garg VK, Goel N. Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv Protein Chem Struct Biol 2021; 125: 73-120
  • 70 Wang M, Zhan F, Cheng H, Li Q. Gambogenic acid inhibits basal autophagy of drug-resistant hepatoma cells and improves its sensitivity to adriamycin. Biol Pharm Bull 2022; 45: 63-70
  • 71 Zeeshan HM, Lee GH, Kim HR, Chae HJ. Endoplasmic reticulum stress and associated ROS. Int J Mol Sci 2016; 17: 327
  • 72 Su J, Xu T, Jiang G, Hou M, Liang M, Cheng H, Li Q. Gambogenic acid triggers apoptosis in human nasopharyngeal carcinoma CNE-2Z cells by activating volume-sensitive outwardly rectifying chloride channel. Fitoterapia 2019; 133: 150-158
  • 73 Wang M, Cheng H, Wu H, Liu C, Li S, Li B, Su J, Luo S, Li Q. Gambogenic acid antagonizes the expression and effects of long non-coding RNA NEAT1 and triggers autophagy and ferroptosis in melanoma. Biomed Pharmacother 2022; 154: 113636
  • 74 Mei W, Dong C, Hui C, Bin L, Fenggen Y, Jingjing S, Cheng P, Meiling S, Yawen H, Xiaoshan W, Guanghui W, Zhiwu C, Qinglin L. Gambogenic acid kills lung cancer cells through aberrant autophagy. PLoS One 2014; 9: e83604
  • 75 Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, Cheng H, Jin K, Ni Q, Yu X, Liu C. The role of necroptosis in cancer biology and therapy. Mol Cancer 2019; 18: 100
  • 76 Xu T, Ding W, Ji X, Ao X, Liu Y, Yu W, Wang J. Molecular mechanisms of ferroptosis and its role in cancer therapy. J Cell Mol Med 2019; 23: 4900-4912
  • 77 Ma X, Xu M, Zhang X, Wang X, Su K, Xu Z, Wang X, Yang Y. Gambogenic acid inhibits proliferation and ferroptosis by targeting the miR‐1291/FOXA2 and AMPKα/SLC7A11/GPX4 axis in colorectal cancer. Cell Biol Int 2023; 47: 1813-1824
  • 78 Wang X, Chen Y, Han QB, Chan CY, Wang H, Liu Z, Cheng CH, Yew DT, Lin MCM, He ML, Xu HX, Sung JYY, Kung HF. Proteomic identification of molecular targets of gambogic acid: Role of stathmin in hepatocellular carcinoma. Proteomics 2009; 9: 242-253
  • 79 Liu P, Wu X, Dai L, Ge Z, Gao C, Zhang H, Wang F, Zhang X, Chen B. Gambogenic acid exerts antitumor activity in hypoxic multiple myeloma cells by regulation of miR-21. J Cancer 2017; 8: 3278-3286
  • 80 Giannelli G, Koudelkova P, Dituri F, Mikulits W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol 2016; 65: 798-808
  • 81 Wang M, Tu Y, Liu C, Cheng H, Zhang M, Li Q. Gambogenic acid inhibits invasion and metastasis of melanoma through regulation of lncRNA MEG3. Biol Pharm Bull 2023; 46: 1385-1393
  • 82 Hua X, Liang C, Dong L, Qu X, Zhao T. Simultaneous determination and pharmacokinetic study of gambogic acid and gambogenic acid in rat plasma after oral administration of Garcinia hanburyi extracts by LC-MS/MS. Biomed Chromatogr 2015; 29: 545-551
  • 83 Chen JP, Wang DL, Yang LL, Wang CY, Wang SS. Ultra-high-performance liquid chromatography tandem mass spectrometry method for the determination of gambogenic acid in dog plasma and its application to a pharmacokinetic study. Biomed Chromatogr 2014; 28: 1854-1859
  • 84 Pan LY, Wang YS, Liu XH, Wang N, Xu W, Xiu YF. Pharmacokinetic comparison of five xanthones in rat plasma after oral administration of crude and processed Garcinia hanburyi extracts. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1126 – 1127: 121737
  • 85 Pan L, Xu M, Wang N, Jia Y, Xiu Y. Determination and tissue distribution comparisons of five xanthones after orally administering crude and processed gamboge. Biomed Chromatogr 2023; 37: e5516
  • 86 Sun J, Tang X, Xu Q, Ge T, Peng D, Chen W. Effect of gambogenic acid on cytochrome P450 1A2, 2B1 and 2E1, and constitutive androstane receptor in rats. Eur J Drug Metab Pharmacokinet 2018; 43: 655-664
  • 87 Sun J, Pang M, Tang X, Xu Q, Peng D, Chen W. Antitumor effect of gambogenic acid and its effect on CYP2C and CYP3A after oral administration. Chem Pharm Bull (Tokyo) 2023; 71: 334-341
  • 88 Huang X, Chen YJ, Peng DY, Li QL, Wang XS, Wang DL, Chen WD. Solid lipid nanoparticles as delivery systems for gambogenic acid. Colloids Surf B Biointerfaces 2013; 102: 391-397
  • 89 Lin T, Huang X, Wang Y, Zhu T, Luo Q, Wang X, Zhou K, Cheng H, Peng D, Chen W. Long circulation nanostructured lipid carriers for gambogenic acid: Formulation design, characterization, and pharmacokinetic. Xenobiotica 2017; 47: 793-799
  • 90 Wang F, Ye X, Wu Y, Wang H, Sheng C, Peng D, Chen W. Time interval of two injections and first-dose dependent of accelerated blood clearance phenomenon induced by PEGylated liposomal gambogenic acid: The contribution of PEG-specific IgM. J Pharm Sci 2019; 108: 641-651
  • 91 Lin T, Fang Q, Peng D, Huang X, Zhu T, Luo Q, Zhou K, Chen W. PEGylated non-ionic surfactant vesicles as drug delivery systems for gambogenic acid. Drug Deliv 2013; 20: 277-284
  • 92 Cheng W, Wang B, Zhang C, Dong Q, Qian J, Zha L, Chen W, Hong L. Preparation and preliminary pharmacokinetics study of GNA-loaded zein nanoparticles. J Pharm Pharmacol 2019; 71: 1626-1634
  • 93 Yuan H, Li X, Zhang C, Pan W, Liang Y, Chen Y, Chen W, Liu L, Wang X. Nanosuspensions as delivery system for gambogenic acid: Characterization and in vitro/in vivo evaluation. Drug Deliv 2016; 23: 2772-2779
  • 94 Zha L, Qian J, Wang B, Liu H, Zhang C, Dong Q, Chen W, Hong L. In vitro/in vivo evaluation of pH-sensitive gambogenic acid loaded zein nanoparticles with polydopamine coating. Int J Pharm 2020; 587: 119665
  • 95 Liu H, Chen H, Cao F, Peng D, Chen W, Zhang C. Amphiphilic block copolymer poly (Acrylic Acid)-B-polycaprolactone as a novel pH-sensitive nanocarrier for anticancer drugs delivery: In-vitro and in-vivo evaluation. Polymers (Basel) 2019; 11: 820
  • 96 Wang B, Cheng W, Zhang C, Bao Y, Zha L, Qian J, Hong L, Chen W. Self-assembled micelles based on gambogenic acid-phospholipid complex for sustained-release drug delivery. J Microencapsul 2019; 36: 566-575
  • 97 Yu Q, Zhang B, Zhou Y, Ge Q, Jiali C, Chen Y, Zhang K, Peng D, Chen W. Co-delivery of gambogenic acid and VEGF-siRNA with anionic liposome and polyethylenimine complexes to HepG2 cells. J Liposome Res 2019; 29: 322-331
  • 98 Du M, Geng T, Yu R, Song G, Cheng H, Cao Y, He W, Haleem A, Li Q, Hu R, Chen S. Smart anti-vascular nanoagent induces positive feedback loop for self-augmented tumor accumulation. J Control Release 2023; 356: 595-609
  • 99 Pesonen L, Svartsjö S, Bäck V, de Thonel A, Mezger V, Sabéran-Djoneidi D, Roos-Mattjus P. Gambogic acid and gambogenic acid induce a thiol-dependent heat shock response and disrupt the interaction between HSP90 and HSF1 or HSF2. Cell Stress Chaperones 2021; 26: 819-833

Zoom Image
Fig. 1 Structure diagrams of some caged xanthones with antitumor activity. The chemical structures of compounds were derived from PubChem (https://pubchem.ncbi.nlm.nih.gov/).
Zoom Image
Fig. 2 A schematic representation of the antitumor activity, pharmacokinetics, nanotechnology-mediated delivery of GNA.
Zoom Image
Fig. 3 Mechanisms underlying the antitumor effect of GNA. GNA serves as an inhibitor of EZH2, effectively suppressing tumor cell proliferation through regulating various proteins such as PTEN, CIP2A, and STMN1. Through the activation of p21/p27, GNA modulates downstream cell cycle regulatory proteins like CDK2 and cyclins A2/B1/E, leading to cell cycle arrest. Another mechanism involves GNA inducing cell cycle arrest by inhibiting Akt, thereby impacting CDK4/6 and cyclin D1/D3 through downstream GSK3β. GNAʼs inhibition of Akt also triggers autophagy in tumor cells via the AMPK-mTOR pathway and facilitates mitochondrial apoptosis by modulating the expression of Bcl-2 family proteins. In some tumor cells, GNA also prompts apoptosis through ER stress. Furthermore, GNA activates caspase-8 through Fas, initiating cell apoptosis. In alternative antitumor mechanisms, GNA induces cell ferroptosis via the p53/SLC7A11/GPX4 signaling pathway and induces cell necrosis by activating RIP1, as well as hinders tumor cell migration through the NF-κB-mediated EMT pathway.