Biological effects of corosolic acid as an anti‑inflammatory, anti‑metabolic syndrome and anti‑neoplasic natural compound (Review)

  • Authors:
    • Jinwei Zhao
    • Hong Zhou
    • Yanan An
    • Keshu Shen
    • Lu Yu
  • View Affiliations

  • Published online on: December 2, 2020     https://doi.org/10.3892/ol.2020.12345
  • Article Number: 84
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Accumulating evidence has indicated that corosolic acid exerts anti‑diabetic, anti‑obesity, anti‑inflammatory, anti‑hyperlipidemic and anti‑viral effects. More importantly, corosolic acid has recently attracted much attention due to its anticancer properties and innocuous effects on normal cells. Furthermore, the increasing proportion of obese and/or diabetic populations has led to an epidemic of non‑alcoholic fatty liver disease (NAFLD), which frequently progresses to hepatocellular carcinoma (HCC). Evidence has indicated that NAFLD is closely associated with the development of HCC and comprises a high risk factor. The present review summarizes the anticancer effects of corosolic acid in vitro and in vivo, and its related molecular mechanisms. It also describes the inhibitory effects of corosolic acid on the progression of NAFLD and its associated molecular mechanisms, providing guidance for future research on corosolic acid in NAFLD‑related HCC prevention and treatment. To the best of our knowledge, a review of corosolic acid as an anticancer agent has not yet been reported. Due to its multitargeted activity in cancer cells, corosolic acid exerts anticancer effects when administered alone, and acts synergistically when administered with chemotherapeutic drugs, even in drug‑resistant cells. In addition, as a novel tool to treat metabolic syndromes, corosolic acid uses the same mechanism in its action against cancer as that used in the progression of NAFLD‑related HCC. Therefore, corosolic acid has been suggested as an agent for the prevention and treatment of NAFLD‑related HCC.

Introduction

Cancer is one of the most common causes of mortality worldwide. However, it is not only a serious threat to public health, but also a global socioeconomic burden (1). An estimated 2,814,000 cases of cancer-related death and 4,292,000 new cancer cases occurred in China in 2015 (2). Based on GLOBOCAN (a global cancer statistics database), in 2018 the number of cases of cancer-related death was 9.6 million, and the number of new cancer cases was 18.1 million worldwide (3). However, data also indicate a decline in the number of new cases, which may be associated with lifestyle changes or reduced exposure to high-risk environmental factors in developed countries (4). Accumulating evidence also suggests that the proteins encoded by a variety of aberrantly-expressed regulatory genes promote tumorigenesis; these include anti-apoptotic proteins, transcription factors, growth factors and their respective receptors (57). Tumorigenesis is a multistep process characterized by numerous abnormalities, rather than a single mutation, during cancer initiation, promotion and progression; therefore, a single target agent is unlikely to inhibit cancer growth (8,9). Currently, the primary treatment strategies against tumors include the following: Surgery, chemoradiotherapy, immunotherapy, molecular targeted therapy and Traditional Chinese Medicine. Although chemotherapy has been proven to improve survival in patients with cancer, drug resistance and severe adverse side effects, such as damage to liver function, bone marrow suppression and neurotoxicity, are major obstacles that cause treatment failure (10,11). There is therefore an urgent need to develop novel and more effective drugs with fewer side effects for various types of cancer.

Due to their selective molecular targets, novel bioactive components from plant sources have emerged as new and reliable therapeutic elements for treating various types of human cancer (12,13). Indeed, over the past half century, numerous plant derivatives and secondary metabolites have been used in clinical practice for the treatment of cancer (14,15). For example, pentacyclic triterpenes constitute a group of promising anticancer drugs that comprise the lupane, oleanane and ursane groups (16,17). Since Pisha et al (18) first reported in 1995 that betulinic acid (19), a plant secondary metabolite, is a highly promising anticancer drug, experimental studies have largely focused on the cytotoxic effects of betulinic acid and other types of triterpenes, particularly their apoptosis-inducing mechanisms, initially in melanoma cell lines in vitro and in vivo (2022). The cytotoxic effects of betulinic acid were subsequently confirmed in other cell lines, such as those derived from breast (23), colon and lung cancer (24), as well as neuroblastoma (25). In the last decade, triterpenes were also found to have additional effects on cancer through several modes of action, such as induction of apoptosis and enhancement of endoplasmic reticulum (ER) stress (2325).

Corosolic acid, also known as 2α-hydroxyursolic acid, has a molecular formula of C30H48O4, and a molecular weight of 472.70 g/mol (Fig. 1). As a prevalent pentacyclic triterpenoid and the principal component of Banaba leaves, corosolic acid has received a great deal of attention due to its anti-diabetic properties (26). Corosolic acid is known as a ‘phyto-insulin’ or ‘botanical insulin’ (27). It is the principal component of Lagerstroemia speciosa leaves (also called Banaba), a tropical plant found in the Philippines, Vietnam, Malaysia and Southern China (28,29). Table I lists the plant species able to biosynthesize corosolic acid (2850). Corosolic acid has also been isolated from European and South American plants.

Table I.

Corosolic acid biosynthesizing/accumulating plant species.

Table I.

Corosolic acid biosynthesizing/accumulating plant species.

First author/s, yearPlant species(Refs.)
Ulbricht et al, 2007Banaba(28)
Park and Lee, 2011Banaba(29)
Kim et al, 2011Vaccinium macrocarpon (cranberry)(30)
Aguirre et al, 2006Ugni molinae(31)
Hou et al, 2009Eriobotrya japonica(32)
Hu et al, 2006Eriobotrya japonica(33)
LV et al, 2008Eriobotrya japonica(34)
Lu et al, 2009Eriobotrya japonica(35)
Rollinger et al, 2010Eriobotrya japonica(36)
Banno et al, 2004Perilla frutescens(37)
Kim et al, 2005Campsis grandiflora(38)
Na et al, 2006Symplocos paniculata(39)
Thuong et al, 2006Weigela subsessilis(40)
Lee and Thuong, 2010Weigela subsessilis(41)
Yang et al, 2006Glechoma longituba(42)
Shen et al, 2006Potentilla chinensis(43)
Kang et al, 2008Rubus bioflorus(44)
Liu et al, 2007Phlomis umbrosa(45)
Li et al, 2017Rosa laevigata Michx(46)
Huang et al, 2014Rubus stans(47)
Huang et al, 2016Rosa cymosa Tratt(47)
Ku et al, 2015Actinidia chinensis(48)
Cheng et al, 2017Actinidia valvata Dunn.Radixa(49)
Manayi et al, 2013L. Salicaria(50)

a Dunn.Radix means root.

Experimental studies have indicated that corosolic acid plays a pivotal anticancer role in several tumorigenic processes in vitro and in vivo, including cellular proliferation, apoptosis, angiogenesis, lymphangiogenesis, metastasis and tumor immunity, and it exerts a synergistic effect when administered with other anticancer agents (Fig. 2) (5153). In addition, corosolic acid has the ability to modulate multiple cancer-related signaling pathways and processes, such as the nuclear factor kappa-B (NF-κB), phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) and Wnt/β-catenin pathways, apoptosis, nuclear factor erythroid 2-related factor 2 (Nrf2) and several other components associated with cellular proliferation or mortality (Table II) (49,51,54,55). However, more research is required to determine its potential in human clinical trials. The most recent registry data from Surveillance, Epidemiology and End Results shows that the morbidity of liver and intrahepatic bile duct cancers have risen on average 3.0% each year between 2004 and 2013 in the United States (56). In particular, hepatocellular carcinoma (HCC) is an aggressive cancer with a poor prognosis. Chronic liver diseases, such as hepatitis B and C virus infections, alcoholic liver disease, non-alcoholic fatty liver disease (NAFLD) and cirrhosis are the most common underlying causes of HCC (41). NAFLD in particular, has been recognized as one of the leading etiologies for the development of HCC (57,58). NAFLD encompasses a spectrum of chronic liver diseases, ranging from simple steatosis to liver injury, which are closely associated with metabolic syndrome (MS) and are characterized by conditions such as obesity, diabetes and dyslipidemia (5961). The understanding of the pathogenesis of NAFLD-related HCC is limited, and several possible mechanisms of NAFLD-related HCC have been described, including obesity-induced inflammation (6264), insulin resistance (IR) (6568), oxidative stress (69,70) and adaptive immune responses (71,72).

Table II.

Summary of the effects of corosolic acid in different types of cancers in vitro.

Table II.

Summary of the effects of corosolic acid in different types of cancers in vitro.

First author/s, yearCancer typeCell typesMolecular mechanismEffectsDrug synergism(Refs.)
Xu et al, 2017 and Ku et al, 2015Liver cancerHuh7βTrCP-dependent Ubiquitination of YAP (↑ YAP, ↓ βTrCP) VEGFR2/Src/FAK pathway (↓ VEGFR2, Src, FAK)↑ Apoptosis ↓ Migration activity, cell motilityActino-mycin DNA(48,80)
Woo et al, 2018 SK-Hep1, Huh7↑ Lipid peroxidation↑ Non-apoptotic cell deathNA(90)
Lee et al, 2010Gastric cancerNCI-N87HER2/neu oncogene↑ Cell cycle arrest and apoptosis pathway (↓ HER2, Akt, Erk; ↑ P27kip1, cyclin D1)Adria-mycin, 5-FU(81)
Cheng et al, 2017 BGC823NF-κB pathway (↓ P65, Fas, FasL, Bcl-2, Smac; ↑ IκBα, Bax, survivin)↑ Cell cycle arrest and apoptosisNA(49)
Lee et al, 2010 SNU-601AMPK-mTOR pathway (↑ AMPK; ↓ mTOR)↑ Cell cycle arrest and apoptosisNA(82)
Lee et al, 2015 SNU-620mTOR signaling pathway (↓ mTOR); caspase-dependent pathway (↑ caspase-8,-9 and −3)↑ Cell cycle arrest and apoptosis5-FU(106)
Yamaguchi et al, 2006 SNU-620/5-FUR (↓ Bcl-2, TS, mTOR/4EBP1, PARP; ↑ AMPK, Bim, caspase-3, poly-ADP-ribose)AMPK-mTOR pathway 5-FU(109)
Sung et al, 2014Colon cancerHCT116caspase-dependent pathway (↑ caspase-8,-9 and −3, Bax, Fas, FasL; ↓ Bcl-2, survivin)↑ Apoptotic cell deathNA(83)
Yoo et al, 2015 CT-26FAK pathway (↓ angiopoetin-1, FAK, ERK1/2); caspase-dependent pathway (↑ caspase-8,-9 and −3)↓ Tumor proliferation; ↑ cell cycle arrest and apoptosisNA(53)
Kim et al, 2014 APC-mutated HCT15Wnt/β-catenin pathway (↓ β-catenin)↓ Tumor proliferationNA(51)
Nho et al, 2013Lung cancerA549 Mitochondrial/caspase-dependent pathway (↑ caspase −7,-8,-9 and −3, ROS; ↓ Bcl-2, survivin, Bid)↑ Apoptotic cell deathNA(99)
Woo et al, 2018Kidney cancerCaki, ACHN, A498↑ Lipid peroxidation↑ Non-apoptotic cell deathNA(90)
Woo et al, 2018Breast cancerMDA-M, B231↑ Lipid peroxidation↑ Non-apoptotic cell deathNA(90)
Ma et al, 2018Prostate cancerPC-3, DU145ER stress; IRE-1/ASK1/JNK signaling pathway and PERK/eIF2α/ATF4/CHOP signaling pathway (↑ IRE-1, PERK, CHOP, TRIB3; ↓ AKT)↑ ER stress-dependent apoptosisNA(89)
Yang et al, 2017 TRAMP-C1Nrf2/HO-1 pathway (↑ H3KK27ac; ↓ DNMTs, HDACs, H3k27me3)↑ Epigenetic alterationsNA(54)
Xu et al, 2009Cervical cancerHeLamitochondrial pathway and caspases activation (↑ Bax/Bcl-2 ratio, caspase-8,-9 and −3)↑ Cell cycle arrest and apoptosisNA(91)
Yong et al, 2016 CaSkiPI3K/Akt signaling pathway (↓ PI3K/Akt)↑ Cell cycle arrest and apoptosisNA(55)
Fujiwara et al, 2013Ovarian CancerSKOV3, RMG-1 and ES-2 SKOV3STAT3 pathway (↓ STAT3) and ↓ M2 macrophage polarization↓ Chemoresistance; ↓ Tumorigenic macrophagesPTX, CDDP and DOX(95)
Fujiwara et al, 2011GlioblastomaU373, T98GJAK-STAT3, NF-κB pathway (↑ T lymphocytes infiltration; ↓ MDSC, COX2 mRNA, CCL-2 mRNA, M2 polarization)↑Antitumor immunityNA(96)
Cai et al, 2011OsteosarcomaMG-63 Mitochondria-mediated apoptosis pathway (↑ caspase-3/9)↑ Mitochondria-mediated apoptosisAdriamycin, cisplatin(97)
Wang et al, 2018RetinoblastomaY-79MELK-FoxM1 signaling (↓ MELK, FoxM1)↑ Cell cycle arrest and apoptosis; ↑ cytotoxicityNA(101)

[i] NA, not applicable; HER2, human epidermal growth factor receptor 2; AMPK, adenosine monophosphate; mTOR, activated protein kinase-mammalian target of rapamycin; CCL-2, chemokine (C-C motif) ligand 2; Fas, apoptosis antigen 1; VEGFR, vascular growth factor receptor; Src, steroid receptor coactivator; FAK, focal adhesion kinase; cdc42, cell division cycle42; Smac, second mitochondria derived activator of caspase; Bax, B-cell lymphoma-2 associated X; NF-κB, nuclear factor kappa-B; IκBα, inhibitor of NF-κBα; ER, endoplasmic reticulum; IRE-1, inositol-requiring ER-to-nucleus signal kinase 1; ASK1, apoptosis signal regulating kinase 1; JNK, Jun N-terminal kinase; PERK, protein kinase RNA-like ER kinase; eIF2α, eukaryotic initiation factor 2α; ATF4, activating transcription factor 4; CHOP, C/EBP-homologous protein; p27Kip1, cyclin-dependent kinase inhibitor 1B; MELK, maternal embryonic leucine-zipper kinase; FoxM1, forkhead box M1; Nrf2, nuclear factor erythroid 2-related factor 2; HO-1, heme oxygenase-1; STAT3, signal transducer and activator of transcription 3; MDSCs, myeloid-derived suppressor cells; COX-2, cyclooxygenase-2; Akt, protein kinase B; ERK, extracellular signal-regulated protein kinase; YAP, Yes-associated protein; FasL, TNF ligand superfamily member 6; P65, NF-кB subunit; 5-Fu, 5-Fluorouracil; TS, thymidine synthase; Bim, Bcl-2 interacting mediator of cell death; PARP, poly ADP-ribose polymerase; Bid, BH3 interacting domain death agonist; ROS, reactive oxygen species; H3KK27ac, lysine H3K27 acetylation; DNMTs, DNA methyltransferases; HDACs, histone deacetylases; H3K27me3, trimethylation of lysine 27 on histone 3; PTX, paclitaxel; CDDP, Cisplatin; DOX, doxorubicin; ↑, indicates upregulation; ↓, indicates downregulation.

Accumulating experimental evidence has suggested that corosolic acid possesses a variety of biological properties, exerting anti-diabetic, anti-obesity, anti-hyperlipidemic, anti-viral, anti-inflammatory and anticancer effects (26,73,74). Therefore, the present review describes the anticancer effects and related molecular mechanisms of corosolic acid, highlighting its ability to inhibit NAFLD progression, and providing guidelines for future research on its use as an agent in NAFLD-related HCC prevention and treatment.

Corosolic acid exerts anticancer effects in vitro

Effects and mechanisms of corosolic acid in neoplasic cell lines from the digestive system

Cancer cell migration is a critical process in tumor development and metastasis (75,76), and is closely associated with vascular growth factor receptor (VEGFR) signaling (57,58); thus, the inhibition of VEGFR, and VEGFR2 in particular, is considered an important treatment approach for HCC and prevent HCC metastasis (7779). Ku et al (48) showed that the half-maximal inhibitory concentration (IC50) for corosolic acid was 2.5 µM for migratory ability, and 50 µM for cytotoxicity on the HCC Huh7 cell line. In addition, corosolic acid treatment resulted in a decrease in Huh7 cell migration in a dose-dependent manner, and corosolic acid at a dose of 2.5 µM induced low cytotoxicity for 24 h (IC50 cytotoxicity/IC50 migration=20), compared to the untreated control (48). The authors further demonstrated that the cytotoxic effects observed with corosolic acid might be associated with the markedly suppression of the VEGFR2/steroid receptor coactivator/focal adhesion kinase (FAK)/cell division cycle42 (cdc42) signaling pathway and the inhibition of the kinase activity of VEGFR2. On the other hand, Xu et al (80) reported that corosolic acid had reduced efficacy in treating liver cancer, since it accelerated the degradation of the transcription factors of Yes-associated protein (YAP) by enhancing large tumor suppressor gene 1-induced phosphorylation and β-transductin repeat containing protein (βTrCP)-dependent ubiquitination. However, Xu et al (80) also demonstrated that corosolic acid-induced apoptosis of liver cancer cells was enhanced by combined treatment with actinomycin D, which resulted in elevated YAP protein levels and decreased βTrCP protein activity. This study suggests that the effectiveness of liver cancer treatment with corosolic acid (at a final concentration of 10 µM) might be improved by its combined administration with 5 µg/ml actinomycin D for 24 h (80).

In gastric cancer cells, corosolic acid has been shown to effectively inhibit the progression of carcinogenesis through multiple mechanisms, including targeting of the adenosine monophosphate-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR) signaling pathway, the inhibition of the NF-κB pathway, the downregulation of EGFR2/neu oncogene, the promotion of the anticancer activities of 5-fluorouracil (5-FU) via mTOR inhibition, and the reduction of 5-FU chemoresistance through the activation of the AMPK pathway (49,81,82). In human gastric cancer NCI-N87 cells, corosolic acid has been shown to inhibit the expression of human epidermal growth factor receptor 2 (HER2) and AMPK-mTOR signal phosphorylated proteins, such as Akt and extracellular signal-regulated protein kinase (ERK), which are involved in signaling pathways downstream of HER2, with the inhibitory effect of corosolic acid being both dose- and time-dependent (25 µM for 12, 24 and 48 h, and 50 µM for 24 h) (81). Furthermore, corosolic acid has been found to induce G0/G1 arrest, which was associated with the induction of cyclin-dependent kinase inhibitor 1B and the downregulation of cyclin D1 (81). In addition, Lee et al (81) found that corosolic acid could effectively inhibit cell proliferation in both a dose- and time-dependent manner (1, 5, 10 and 50 µM for 24 h, and 25 µM for 3, 6, 12, 24 and 48 h). Furthermore, corosolic acid has been shown to induce cell cycle arrest and apoptosis through the downregulation of the HER2/neu oncogene, suggesting that it may play a role in patients with HER2-amplified gastric cancers (81). Moreover, at an IC50 value of 16.9±2.9 µM, corosolic acid has been shown to inhibit the proliferation of human gastric cancer SNU-601 cells via AMPK-mTOR signaling (82). Another study has reported that corosolic acid treatment at a concentration of 10, 20, 40 and 80 mg/ml for 72 h induces apoptosis in human gastric cancer BGC823 cells in a dose-dependent manner (49). This effect is achieved by inhibiting the NF-κB (p65 subunit) pathway, by decreasing the mRNA and protein expression of p65, apoptosis antigen 1 (Fas), second mitochondria derived activator of caspase, and B-cell lymphoma-2 (Bcl-2), whilst increasing that of Bcl-2 associated X (Bax), inhibitor of NF-κB (IκB) α and survivin (49). In addition, the experimental data of Sung et al (83) provides insights into the molecular mechanisms through which corosolic acid induces the apoptosis of colorectal cancer cells. Corosolic acid, at an IC50 value of 24 µM for 24 h, inhibits the viability of colorectal cancer HCT116 cells by inducing apoptotic cell death in a dose-dependent manner, through a molecular mechanism associated with the upregulation of the proapoptotic proteins Bax, Fas and Fas ligand (FasL), and the downregulation of the anti-apoptotic proteins Bcl-2 and survivin. Of note, corosolic acid was proven to be an ideal antagonist of the Wnt/β-catenin pathway (51). Corosolic acid decreased the level of intracellular β-catenin and suppressed the proliferation of colon cancer HCT-15 and DLD-1 cells with an APC mutation in a dose-dependent manner (20, 40 and 60 µM for 8 h), which was achieved by promoting N-terminal phosphorylation and degrading the proteasomes of β-catenin (Table II) (51).

Effects and mechanisms of corosolic acid on tumor cells from the urogenital system

Accumulating evidence has suggested that activated Nrf2 plays a critical role in the proliferation and survival of tumor cells, making its inhibition a promising therapeutic strategy for cancer treatment (8487). A previous report on several Nrf2 inhibitors showed that these are promising therapeutic agents (88). Of note, corosolic acid at a concentration of 0.25–32 µM for 3 or 5 days inhibited the proliferation of TRAMP-C1 cells, a type of anchorage-independent human prostate cancer (PCa) cell line with increased levels of mRNA and protein expression of Nrf2, heme oxygenase-1 (HO-1) and nicotinamide adenine dinucleotide phosphate quinone oxidoreductase 1; however, corosolic acid did not exert the same inhibitory effect in Nrf2-knockout TRAMP-C1 cells (54). These findings indicate that the significant cytotoxic effect of corosolic acid might be associated with its ability to restore the expression of Nrf2 via epigenetic modification (54). In addition, in the PCa, PC-3 and DU145 cell lines, (ER) stress was activated by 0, 5, 10 and 15 µM corosolic acid after 24 and 48 h, through two proapoptotic signaling pathways: The inositol-requiring ER-to-nucleus signal kinase 1/apoptosis signal regulating kinase 1/Jun N-terminal kinase (JNK) pathway and the protein kinase RNA-like ER kinase/eukaryotic initiation factor 2 α/activating transcription factor 4/C/EBP-homologous protein signaling pathway, which induced apoptosis and suppressed cell proliferation (89). However, Woo et al (90) found that the corosolic acid-induced death of human renal carcinoma Caki cells (at 10 µM for 24 h) was inhibited by the use of α-tocopherol (a hydrophobic anti-oxidant that prevents free radical damage), but was not inhibited by benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (an apoptosis inhibitor), necrostatin-1 (a necroptosis inhibitor), ferrostatin-1 or deferoxamine (ferroptosis inhibitors) (90). Futhermore, corosolic acid induces lipid oxidation, and α-tocopherol markedly prevents corosolic acid-induced lipid peroxidation and cell death. Anti-chemotherapeutic effects of α-tocopherol are dependent on inhibition of lipid oxidation rather than inhibition of ROS production (90). It was therefore speculated that corosolic acid induced the non-apoptotic cell death associated with lipid peroxidation in cancer cells (90). Furthermore, in renal carcinoma ACHN and A498 cells, treatment with 10 µM corosolic acid for 24 h induced non-apoptotic cell death (90). Xu et al (91) reported that treating human cervix adenocarcinoma HeLa cells with 40 µM corosolic acid for 24 h could induce cell cycle arrest at the S phase, and promote apoptosis by activating caspases-8, −9 and −3 and disrupting mitochondrial membrane potential (91). In another report on CaSki human cervical cancer cells, the results indicated that 10, 50 and 100 µM corosolic acid treatment for 12, 24 and 48 h effectively inhibited proliferation in a dose- and time-dependent manner (55). In addition, the results revealed that the cytotoxic effects of corosolic acid inhibited tumor cell proliferation by inducing apoptosis and cell cycle arrest, and suppressing the PI3K/Akt signaling pathway (55). It has also been demonstrated that in epithelial ovarian cancer (92), glioma and lymphoma (93,94) cells, the activation of signal transducer and activator of transcription 3 (STAT3) was induced by co-culturing the cells with M2, but not M1 macrophages. However, Fujiwara et al (95) demonstrated that corosolic acid, at a minimum of 30 µM for 48 h, suppressed STAT3 activation in co-culture experiments with epithelial ovarian cancer ES-2 cells treated with bromodeoxyuridine (used to abrogate macrophage differentiation into the M2 phenotype), and that STAT3 inhibition was associated with the prevention of M2 macrophage polarization. In addition, the epithelial ovarian cancer cell line SKOV3 treated with 20 µM corosolic acid for 24 h, showed no effect on the viability of these cells, suggesting that corosolic acid have no anticancer properties at this concentration. By contrast, 20 µM corosolic acid enhanced the inhibitory effect of paclitaxel (PTX; 10 µM) on the proliferation of the epithelial ovarian cancer cell lines SKOV3, RMG-1 and ES-2. These results demonstrated that corosolic acid enhances the anticancer activity of anticancer drugs such as PTX in epithelial ovarian carcinoma cells (95). Notably, the combination of 20 µM corosolic acid and 10 µM paclitaxel for 24 h also inhibited STAT3 activity in the epithelial ovarian cancer cells, but corosolic acid alone or PTX alone had lesser effects on the STAT3 activity (95). These data suggested that corosolic acid enhanced cancer cell chemosensitivity and effectively inhibited cancer cell proliferation, which was also found to be associated with the prevention of M2 polarization via the suppression of STAT3 activation (95). These findings were similar to those showing that corosolic acid (30 µM for 1 h) suppressed the M2 macrophage polarization and proliferation of U373 and T98G glioblastoma cells in parallel with inhibiting both STAT3 and NF-κB activation (Table II) (96).

Effects and mechanisms of corosolic acid in neoplasic cell lines from osteosarcoma and lung metastasis

The response of osteosarcoma MG-63 cells to corosolic acid treatment has been previously reported (97,98). The results shared by both studies indicate that the viability of osteosarcoma MG-63 cells was significantly inhibited by corosolic acid (35 µM for 12 h, and 20, 30 and 40 µM for 24 h), and that corosolic acid induced apoptosis through the activation of caspases-3 and −9 to cause mitochondrial dysfunction (97,98). Moreover, using human osteosarcoma Saos2 and HSOS-1 cell lines and the murine osteosarcoma LM8 cell line, Horlad et al (52) reported that treatment with 30 µM corosolic acid for 24 h inhibited lung metastasis by inhibiting STAT3 activation, increasing the number of infiltrating lymphocytes in the tumor tissues and abrogating the immunosuppressive effect of myeloid-derived suppressor cells (MDSCs) through the decreased expression of cyclooxygenase-2 (COX-2) and chemokine (C-C motif) ligand 2 (CCL2) mRNA in these MDSCs (52) (Table II).

Effects and mechanisms of corosolic acid in the lung cancer A549 cell line

Corosolic acid (10–40 µM for 6–48 h) had a significant inhibitory effect on A549 cells, a human lung adenocarcinoma cell line, in a concentration- and time-dependent manner (99). Exposure to corosolic acid induced cell cycle arrest at the sub-G1 stage and caused apoptotic death in A549 cells (99). In addition, corosolic acid also activated caspases-3/-7, −8 and −9 and poly (ADP-ribose) polymerase, and increased the levels of reactive oxygen species (ROS). Corosolic acid-induced apoptosis was inhibited by exposure to the ROS scavenger N-acetylcysteine (99). These results indicate that corosolic acid induced apoptosis through mitochondria-mediated and caspase-dependent processes in a ROS-dependent manner (99). In addition, under CoCl2-stimulated hypoxic conditions, corosolic acid (IC50 of 12.5 µg/ml for 48 h) induced marked cytotoxicity in cancerous cells, and its action was associated with the suppressed expression of hypoxia-inducible factor-1 α and β and its downstream target genes (Table II) (100).

Effects and mechanisms of corosolic acid in the retinoblastoma Y79 cell line

The response of human retinoblastoma Y-79 cells to corosolic acid was investigated (101). The results showed that corosolic acid (10 µM for 24 h) could induce cell cycle arrest and apoptosis through its cytotoxic activity (IC50 of 4.15 µM for 24 h or 3.37 µM for 48 h) in a dose-dependent manner (101). The results also showed that the transcriptional activity of forkhead box M1 (FoxM1) was self-induced or driven by maternal embryonic leucine-zipper kinase (MELK), and that corosolic acid inhibited the expression levels of MELK and FoxM1 (101). The report established a promising therapeutic target of human retinoblastoma via MELK-FoxM1 signaling (Table II) (101).

Corosolic acid exerts anticancer effects in vivo

Banno et al (37) published the first study on the cancer-preventing and anti-inflammatory activities of corosolic acid in vivo. Corosolic acid exhibited a marked anti-inflammatory effect, with an IC50 of 0.09–0.3 mg per ear on 12-O-tetradecanoylphorbol-13-acetate-induced inflammation (1 µg/ear) in mice; however, corosolic acid with an IC50 of 0.09–0.3 mg per ear did not exhibit an anticancer activity in a mouse tumor model. In vivo experiments in a murine sarcoma model showed that subcutaneous tumor development and lung metastasis was significantly suppressed by orally administered corosolic acid (17.5 mg/kg, 2 times/week for 21 days) (102). Corosolic acid was indicated as a potential new anticancer agent, as it targets macrophage polarization (102). In a murine osteosarcoma model, it was shown that orally administered corosolic acid (17.5 mg/kg/day for 7 days) significantly suppressed subcutaneous tumor development and pulmonary metastasis (52). It was also indicated that corosolic acid has a potential anticancer effect through targeting macrophage polarization and the immunosuppressive activity of MDSCs (52) Corosolic acid (20 µM) also displayed synergistic effects with anticancer agents, such as adriamycin (10 µM) and cisplatin (10 µM) after 24 h (52). In a mouse model of colon carcinoma, 5 and 25 mg/kg/day corosolic acid, administered via a peritumoral injection for 12 days inhibited allograft colon tumor growth. The results found that corosolic acid reduced the final tumor volume and the blood and lymphatic vessel densities of tumors, indicating that it suppresses in vivo angiogenesis and lymphangiogenesis (53). This was the first report of the anti-angiogenic and anti-lymphangiogenic effects of corosolic acid (53). Ma et al (89) established a xenograft tumor model of castration-resistant prostate cancer, and 10 and 20 mg/kg corosolic acid every 2 days for 14 days, administered via an intraperitoneal injection, was found to reduce tumor growth. Ku et al (48) reported that 5 mg/kg/day corosolic acid for 21 days effectively inhibited HCC Huh7 tumor growth in a male NOD/SCID mice model, and combined treatment of corosolic acid with sorafenib showed a synergistic inhibitory effect on tumor growth (corosolic acid 2.5 mg/kg/day with sorafenib 10 or 20 mg/kg/day) compared with corosolic acid alone, for 21 days in a mouse model (Table III).

Table III.

Summary of the effects and mechanisms of corosolic acid in different types of cancer in vivo.

Table III.

Summary of the effects and mechanisms of corosolic acid in different types of cancer in vivo.

First author, yearCancer model typeCorosolic acid mechanism of actionCorosolic acid dose; administrationEffects(Refs.)
Horlad et al, 2013Murine sarcoma xenograft model↓ STAT3 activation, ↑ CD4+ and CD8+ lymphocytes, ↓ the suppressive effect of MDSC17.5 mg/kg/day; oralImpaired subcutaneous tumor development and lung metastasis(52)
Ku et al, 2015Mouse HCC Huh7 ×enograft modelVEGFR2/Src/FAK pathway (↓ VEGFR2, Src, FAK, ↓ phosphorylation of VEGFR2 and FAK)5 mg/kg/day; intraperitoneal injection85% reduction in tumor mass compared to the control group(48)
Yoo et al, 2015Mouse xenograft colon CT-26 modelAnti-angiogenic and anti-lymphangiogenic effects5 or 25 mg/kg/day; peritumor rejectionReduced the final tumor volume and blood and lymphatic vessel density of tumors(53)
Ma et al, 2018Mouse xenograft PC-3 modelER stress, IRE-1/ASK1/JNK signaling pathway, and PERK/eIF2α/ ATF4/CHOP signaling pathway (↑ IRE-1, PERK, CHOP, TRIB3; ↓ AKT)10 or 20 mg/kg/2 days; intraperitoneal injectionReduced the final tumor volume in a dose-dependent manner(89)
Fujiwara et al, 2014Mouse LM8 sarcoma modelInhibits macrophage polarization to M2 phenotype by suppressing STAT3 activation17.5 mg/kg, 2 times/week; oralReduced the final tumor volume(102)

[i] HCC, hepatocellular carcinoma; MDSCs, myeloid-derived suppressor cells; Src, steroid receptor coactivator; FAK, focal adhesion kinase; cdc42, cell division cycle42; ER, endoplasmic reticulum; STAT3, signal transducer and activator of transcription 3; VEGFR2, VEGFR, vascular growth factor receptor 2; IRE-1, inositol-requiring ER-to-nucleus signal kinase 1; ASK1, apoptosis signal regulating kinase 1; JNK, Jun N-terminal kinase; PERK, protein kinase RNA-like ER kinase; eIF2α, eukaryotic initiation factor 2α; ATF4, activating transcription factor 4; CHOP, C/EBP-homologous protein; TRIB3, tribbles pseudo-kinase 3; ↑, indicates upregulation; ↓, indicates downregulation.

Corosolic acid exerts synergistic anticancer activity with chemotherapeutic drugs

Accumulating experimental evidence has highlighted the pivotal role of STAT3 activation in the resistance to chemotherapy and radiotherapy in the thyroid cancer-derived CD133+ cells (103) and human epithelial ovarian cancer cells (104). It is thought that inhibiting STAT3 might be effective for treating patients with malignant tumors (103105). A report by Fujiwara et al (95) suggested that 20 µM corosolic acid, as a selective STAT3 inhibitor, is able to increase sensitivity to chemotherapeutic agents, including paclitaxel (10 µM), cisplatin (10 µM) and doxorubicin (10 µM), in epithelial ovarian cancer SKOV3, RMG-1 and ES-2 cell lines for 24 h. In addition, the results of a study by Lee et al (81) showed that 25 µM corosolic acid enhances the inhibitory effect on human gastric cancer NCI-N87 cell proliferation when combined with adriamycin (0.01 to 2 mg/ml) and 5-FU (0.1 to 50 mg/ml), but not with docetaxel (0.01 to 2 mg/ml) or paclitaxel (0.01 to 6 mg/ml). Lee et al (106) indicated that corosolic acid (50 µM) enhances the anticancer activity of 5-FU (20 µg) after 24 h in human gastric carcinoma SNU-620 cells in an mTOR inhibition-dependent manner. In addition, a report by Fujiwara et al (102) showed that corosolic acid (20 µM) also displayed synergistic effects with anticancer agents, such as adriamycin (10 µM) and cisplatin (10 µM) 24 h. Furthermore, in a study by Park et al (107), a 5-FU-resistant gastric cancer cell line (SNU-620/5-FUR) was established, which had a marked reduced AMPK phosphorylation when compared with the parental cell line, SNU-620. Cell treatment with 25 µM corosolic acid for 24 h was found to enhance the chemosensitivity of 5-FU-resistant gastric cancer cells, and the reduction of AMPK phosphorylation by compound c (AMPK inhibitor) was revealed to be associated with increased 5-FU-resistant cell viability (107). Corosolic acid treatment significantly reduced cell viability while compound c reversed corosolic acid-induced cell growth inhibition (107). The corosolic acid-induced AMPK activation was markedly increased by additional 5-FU treatment, while compound c reversed AMPK phosphorylation (107). These results imply that corosolic acid can activate AMPK and sensitize gastric cancer to 5-FU (150 µM; Table II).

Corosolic acid exerts anti-inflammatory and anti-MS effects

Nelson et al (108) first reported that corosolic acid (2 µmol twice-weekly over a 2-week period) may be an effective anti-inflammatory agent. Yamaguchi et al (109) further explored corosolic acid isolated from Banaba leaves and found that it prevented oxidative stress and reduced the inflammation caused by MS. In SHR-cp rats with characteristics that included obesity, hyperglycemia, hyperlipidemia, hypertension, hyperinsulinemia, oxidative stress and inflammation, a diet rich in 0.072% corosolic acid for 14 weeks ameliorated hypertension, regulated hyperlipidemia, prevented oxidative stress and ameliorated inflammation (109). A report by Chen et al (110) suggested that 6 µM corosolic acid treatment for 30 min was able to inhibit monocyte chemoattractant protein-1 expression, and that 2 µg/kg/day corosolic acid for 10 days ameliorated atherosclerosis by regulating the nuclear factor-κB signaling pathway in apolipoprotein E-deficient mice. Furthermore, Kim et al (111) reported that exposure of lipopolysaccharide (LPS)-pretreated bone marrow-derived monocytes to corosolic acid downregulated the NF-κB target genes pyrin domain-containing protein 3 (NLRP3) and interleukin-1 (IL-1), which was similar to the effects observed for LPS-pretreated bone marrow-derived monocytes with an inhibitor of IL-1 receptor-associated kinase (IRAK; a signaling molecule upstream of LPS-induced activated toll-like receptor 4) or with LPS and Bay11-7082 (an IκB) (111). Treatment with Bay11-7082 (an inhibitor of IκB-α), had no effect on corosolic acid-mediated inhibition of IRAK-1 activation, indicating that corosolic acid-mediated attenuation of IRAK-1 phosphorylation was independent of NF-κB signaling (111). These data indicate that corosolic acid plays a vital inhibitory role in acute inflammation by regulating IRAK-1 phosphorylation in an NF-κB-independent manner (111). In addition, a report by Yang et al (27) revealed that 10 mg/kg/day corosolic acid for 8 weeks improved insulin sensitivity and glucose intolerance, and attenuated hyperlipidemia in C57BL/6 mice. In addition, corosolic acid suppressed the phosphorylation of inhibitor of nuclear factor kappa-B kinase (IKKβ) and downregulated the expression of proinflammatory cytokine genes, which in turn alleviated adipose tissue inflammation (27). Corosolic acid also enhanced the phosphorylation of serine (Ser)/threonine on insulin receptor substrate-1 (IRS-1) and its downstream effector Akt, and enhanced insulin signal transduction (27). Finally, in AMPKα-knockdown adipocytes, the inhibitory effects of corosolic acid on IRS-1 and IKKβ Ser phosphorylation were abolished, indicating that corosolic acid ameliorated IR and inhibited inflammation through the activation of AMPK in a liver kinase B1-dependent manner (27) (Fig. 3).

Proposed mechanisms underlying the inhibition of NAFLD-related HCC progression by corosolic acid

The characteristics of NAFLD include obesity, IR, hypertension and dyslipidemia, which are also the most common characteristics observed in livers affected by MS (112). Furthermore, the development of NAFLD-related HCC is increasingly recognized, since patients with NAFLD are at high risk of developing HCC (112). NAFLD-associated HCC has been estimated to account for 10–12% of HCC cases in Western populations and 1–6% of HCC cases in Asian populations from 42 sites in 14 countries from 2005 to 2012 (58). Based on multiple studies, accumulated evidence has suggested that type 2 diabetes mellitus (T2DM) and obesity are independent risk factors for the development of HCC in patients with NAFLD.

Animal and human studies and in vitro systems have indicated that corosolic acid has multiple properties, including anti-diabetic, anti-obesity, anti-inflammatory, anti-hyperlipidemic and anti-viral activity (26,73,74). On the other hand, as aforementioned, corosolic acid has shown an ability to modulate multiple cancer-related signaling pathways, including the adenosine mitogen-activated protein kinase (AMPK), NF-κB, PI3K/Akt/mTOR, Wnt/β-catenin, FAK, ERK1/2, STAT3 in MDSCs, Nrf2/HO-1 and numerous other signaling pathways associated with cell proliferation and cell death, among other cellular processes in multiple types of malignant tumors (as observed in preclinical in vitro and in vivo experiments) (48,49,51,52,54,55,8183,9196,107). Due to its anticancer and anti-immunity activities, corosolic acid has attracted growing attention. A schematic plot of the proposed mechanisms of the corosolic acid-induced inhibition of NAFLD-related HCC progression is presented in Fig. 5. The release of an increased number of proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), is promoted by obesity and adipose tissue expansion (62). NF-κB, JNK, mTOR and extracellular signal-related kinases, such as those associated with pro-oncogenic pathways, are stimulated by TNF-α (63). It is highly likely that hepatocytes with previously acquired oncogenic mutations will continue the malignant transformation that is induced by the chronic activation of the IL-6/STAT3 axis (64). As aforementioned, as an agent with anticancer and anti-inflammatory activity, corosolic acid plays vital roles in the inhibition of proinflammatory cytokine and mTOR expression, and the downregulation of ERK, while as a STAT3 and NF-κB inhibitor, it can enhance anticancer activity (53,95,111). NAFLD promotes systemic and hepatic IR with the resultant hyperinsulinemia-activated proinflammatory cytokines and lipotoxic activity in obesity and T2DM (112). A previous report showed that the production of IRS-1 and insulin-like growth factor-1 (IGF-1) was increased by IR and hyperinsulinemia (65). IGF-1 promotes cell proliferation, inhibits apoptosis and stimulates cell growth (65). Furthermore, IGF-1 contributes to the upregulated expression of the proto-oncogenes c-fos and c-Jun in vitro, and the downregulation of AMPK, which is associated with the development of HCC (66). JNK, another important intracellular marker, is closely linked to obesity, IR, NAFLD and HCC (67). It has also been indicated that JNK-induced phosphorylation and activation of IRS-1 are responsible for obesity-induced IR (67). A report by Chang et al (68) showed that JNK signaling might play a pivotal role in hepatocarcinogenesis, where an increased JNK1 activation was detected by immunostaining in 17/31 HCC samples relative to their paired adjacent normal tissues. In addition, recent studies have revealed the potential role of the adaptive immune system in the development of NAFLD-related HCC (71,72). A report by Ma et al (71) revealed that hepatocytes exhibit increased linoleic acid secretion and mitochondria-derived ROS, both of which led to enhanced carcinogenesis. The same report also found that CD4+ T lymphocytes have greater mitochondrial mass than CD8+ T lymphocytes and generate higher levels of mitochondrially derived ROS. The disruption of mitochondrial function by free fatty acids such as palmitic acid accumulated in NAFLD, caused more oxidative damage and in turn promoted selective depletion of CD4+ T lymphocytes. In addition, blockade of ROS reversed NAFLD-induced hepatic CD4+ T lymphocyte decrease and delayed NAFLD-promoted HCC in mouse models of NAFLD-associated HCC. Wolf et al (72) developed a mouse model recapitulating key features of human metabolic syndrome, non-alcoholic steatohepatitis, and HCC by feeding mice a choline-deficient high-fat diet, and found that carcinogenesis was induced by the metabolic activation of CD8+ T lymphocytes and natural killer cells in the liver. Corosolic acid exerted anticancer immunity by inhibiting STAT3 and NF-κB activation; this immunity was associated with MDSC depletion, decreased levels of COX-2 and CCL2 expression, and an increased number of infiltrating CD8+ T lymphocytes. Furthermore, corosolic acid can also downregulate the activation of P38 and JNK via the inhibition of ROS (Fig. 4) (99,109).

Figure 4.

Molecular mechanisms of NAFLD-HCC. (A) TNF-α/NF-κB, JNK, mTOR axis. Pro-oncogenic pathways, such as NF-κB, JNK, mTOR are stimulated by TNF-α; (B) Chronic activation of the IL-6/STAT3 axis; Hepatocytes with previously acquired oncogenic mutations will continue malignant transformation that is induced by the chronic activation of the IL-6/STAT3 axis. (C) IR/IGF-1, IRS-1/c-fos, c- Jun, JNK, AMPK axis; IGF-1 is increased by IR, IGF-1 contributes to the upregulated expression of the proto-oncogenes c-fos and c-Jun, and the downregulation of AMPK, which is associated with the development of HCC. JNK-induced phosphorylation and downregulation of IRS-1 are responsible for obesity-induced IR, and JNK signaling plays a pivotal role in hepatocarcinogenesis. (D) ROS and adaptive immune response. Mitochondria-derived ROS promotes selective depletion of CD4+ T lymphocytes; carcinogenesis is induced by the metabolic activation of CD8+ T lymphocytes and NK cells in the liver. NAFLD, non-alcoholic fatty liver disease; HCC, hepatocellular carcinoma; IR, insulin resistance; IRS-1, insulin receptor substrate-1; IL-6, interleukin-6; NF-κB, nuclear factor kappa-B; TNF-α, tumor necrosis factor-α; JNK, Jun N-terminal kinase; mTOR, adenosine monophosphate-activated protein kinase; STAT3, signal transducer and activator of transcription 3; IGF-1, insulin-like growth factor-1; AMPK, adenosine monophosphate-activated protein kinase; ROS, reactive oxygen species; NK, natural killer; ↑, indicates upregulation; ↓, indicates downregulation.

Conclusions and future perspectives

The present review summarizes current advancements in our understanding of the anticancer activity and mechanisms of corosolic acid in vitro and in vivo. Due to the ability of corosolic acid to target multiple components of cancer cells, it acts not only as an anticancer agent but also as a synergistic adjuvant when administered alongside chemotherapeutic drugs, even in drug-resistant cells. In addition, parts of the same corosolic acid mechanism that ameliorates MS also induces anticancer activity and suppresses the progression of NAFLD-related HCC. Therefore, corosolic acid, a potential tool in MS treatment, is being considered as a possible agent in NAFLD-related HCC prevention and treatment (Figs. 3 and 5).

Acknowledgements

Not applicable.

Funding

This work was supported by the Fund for Science & Technology Development of Jilin Province (grant nos. 20200201544JC, 20160101060JC and 20150101108JC), the National Key R&D Program of China (grant nos. 2017YFD0502200 and 2016YFD0501302), and the Project of the Education Department of Jilin Province (grant no. 2016444).

Availability of data and materials

Not applicable.

Authors' contributions

JZ, HZ, YA, KS and LY participated in the design and interpretation of the studies, the revision of the manuscript. JZ, HZ, YA and KS wrote the review. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Chen WQ, Li H, Sun KX, Zheng RS, Zhang SW, Zeng HM, Zou XN, Gu XY and He J: Report of cancer incidence and mortality in China, 2014. Zhonghua Zhong Liu Za Zhi. 40:5–13. 2018.(In Chinese). PubMed/NCBI

3 

Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 144:1941–1953. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Islami F, Goding Sauer A, Miller KD, Siegel RL, Fedewa SA, Jacobs EJ, McCullough ML, Patel AV, Ma J, Soerjomataram I, et al: Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States. CA Cancer J Clin. 68:31–54. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Khan M, Maryam A, Qazi JI and Ma T: Targeting apoptosis and multiple signaling pathways with icariside II in cancer cells. Int J Biol Sci. 11:1100–1112. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Millimouno FM, Dong J, Yang L, Li J and Li X: Targeting apoptosis pathways in cancer and perspectives with natural compounds from mother nature. Cancer Prev Res (Phila). 7:1081–1107. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Khan M, Maryam A, Zhang H, Mehmood T and Ma T: Killing cancer with platycodin D through multiple mechanisms. J Cell Mol Med. 20:389–402. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Faivre S, Djelloul S and Raymond E: New paradigms in anticancer therapy: Targeting multiple signaling pathways with kinase inhibitors. Semin Oncol. 33:407–420. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Shu L, Cheung KL, Khor TO, Chen C and Kong AN: Phytochemicals: Cancer chemoprevention and suppression of tumor onset and metastasis. Cancer Metastasis Rev. 29:483–502. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Ye B, Li J, Li Z, Yang J, Niu T and Wang S: Anti-tumor activity and relative mechanism of ethanolic extract of Marsdenia tenacissima (Asclepiadaceae) against human hematologic neoplasm in vitro and in vivo. J Ethnopharmacol. 153:258–267. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Yan Y, Xu Z, Dai S, Qian L, Sun L and Gong Z: Targeting autophagy to sensitive glioma to temozolomide treatment. J Exp Clin Cancer Res. 35:232016. View Article : Google Scholar : PubMed/NCBI

12 

Rengarajan T, Nandakumar N, Rajendran P, Haribabu L, Nishigaki I and Balasubramanian MP: D-pinitol promotes apoptosis in MCF-7 cells via induction of p53 and Bax and inhibition of Bcl-2 and NF-κB. Asian Pac J Cancer Prev. 15:1757–1762. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Montaser R and Luesch H: Marine natural products: A new wave of drugs? Future Med Chem. 3:1475–1489. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Efferth T: From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy. Semin Cancer Biol. 46:65–83. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Cheng CS, Chen J, Tan HY, Wang N, Chen Z and Feng Y: Scutellaria baicalensis and cancer treatment: Recent progress and perspectives in biomedical and clinical studies. Am J Chin Med. 46:25–54. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Shanmugam MK, Dai X, Kumar AP, Tan BK, Sethi G and Bishayee A: Oleanolic acid and its synthetic derivatives for the prevention and therapy of cancer: Preclinical and clinical evidence. Cancer Lett. 346:206–216. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Salvador JAR, Leal AS, Valdeira AS, Gonçalves BMF, Alho DPS, Figueiredo SAC, Silvestre SM and Mendes VIS: Oleanane-, ursane-, and quinone methide friedelane-type triterpenoid derivatives: Recent advances in cancer treatment. Eur J Med Chem. 142:95–130. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Pisha E, Chai H, Lee IS, Chagwedera TE, Farnsworth NR, Cordell GA, Beecher CW, Fong HH, Kinghorn AD, Brown DM, et al: Discovery of betulinic acid as a selective inhibitor of human melanoma that functions by induction of apoptosis. Nat Med. 1:1046–1051. 1995. View Article : Google Scholar : PubMed/NCBI

19 

Knowles J and Gromo G: A guide to drug discovery: Target selection in drug discovery. Nat Rev Drug Discov. 2:63–69. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Strüh CM, Jäger S, Schempp CM, Scheffler A and Martin SF: A novel triterpene extract from mistletoe induces rapid apoptosis in murine B16.F10 melanoma cells. Phytother Res. 26:1507–1512. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhao Y, He J, Li J, Peng X, Wang X, Dong Z, Zhao E, Liu Y, Wu Z and Cui H: Demethylzeylasteral inhibits cell proliferation and induces apoptosis through suppressing MCL1 in melanoma cells. Cell Death Dis. 8:e31332017. View Article : Google Scholar : PubMed/NCBI

22 

Liu W, Liu X, Pan Z, Wang D, Li M, Chen X, Zhou L, Xu M, Li D and Zheng Q: Ailanthone induces cell cycle arrest and apoptosis in melanoma B16 and A375 cells. Biomolecules. 9:2752019. View Article : Google Scholar

23 

Tiwari R, Puthli A, Balakrishnan S, Sapra BK and Mishra KP: Betulinic acid-induced cytotoxicity in human breast tumor cell lines MCF-7 and T47D and its modification by tocopherol. Cancer Invest. 32:402–408. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Mullauer FB, van Bloois L, Daalhuisen JB, Ten Brink MS, Storm G, Medema JP, Schiffelers RM and Kessler JH: Betulinic acid delivered in liposomes reduces growth of human lung and colon cancers in mice without causing systemic toxicity. Anticancer Drugs. 22:223–233. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Fulda S, Jeremias I, Steiner HH, Pietsch T and Debatin KM: Betulinic acid: A new cytotoxic agent against malignant brain-tumor cells. Int J Cancer. 82:435–441. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Sivakumar G, Vail DR, Nair V, Medina-Bolivar F and Lay JO Jr: Plant-based corosolic acid: Future anti-diabetic drug? Biotechnol J. 4:1704–1711. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Yang J, Leng J, Li JJ, Tang JF, Li Y, Liu BL and Wen XD: Corosolic acid inhibits adipose tissue inflammation and ameliorates insulin resistance via AMPK activation in high-fat fed mice. Phytomedicine. 23:181–190. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Ulbricht C, Dam C, Milkin T, Seamon E, Weissner W and Woods J: Banaba (Lagerstroemia speciosa L.): An evidence-based systematic review by the natural standard research collaboration. J Herb Pharmacother. 7:99–113. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Park C and Lee JS: Banaba: The natural remedy as antidiabetic drug. Biomed Res. 22:127–131. 2011.

30 

Kim E, Sy-Cordero A, Graf TN, Brantley SJ, Paine MF and Oberlies NH: Isolation and identification of intestinal CYP3A inhibitors from cranberry (Vaccinium macrocarpon) using human intestinal microsomes. Planta Med. 77:265–270. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Aguirre MC, Delporte C, Backhouse N, Erazo S, Letelier ME, Cassels BK, Silva X, Alegría S and Negrete R: Topical anti-inflammatory activity of 2alpha-hydroxy pentacyclic triterpene acids from the leaves of Ugni molinae. Bioorg Med Chem. 14:5673–5677. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Hou W, Li Y, Zhang Q, Wei X, Peng A, Chen L and Wei Y: Triterpene acids isolated from Lagerstroemia speciosa leaves as alpha-glucosidase inhibitors. Phytother Res. 23:614–618. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Hu C, Chen L, Xin Y and Cai Q: Determination of corosolic acid in Eriobotrya japonica leaves by reversed-phase high performance liquid chromatography. Se Pu. 24:492–494. 2006.(In Chinese). PubMed/NCBI

34 

Lv H, Chen J, Li WL and Zhang HQ: Studies on the triterpenes from loquat leaf (Eriobotrya japonica). Zhong Yao Cai. 31:1351–1354. 2008.(In Chinese). PubMed/NCBI

35 

Lu H, Xi C, Chen J and Li W: Determination of triterpenoid acids in leaves of Eriobotrya japonica collected at in different seasons. Zhongguo Zhong Yao Za Zhi. 34:2353–2355. 2009.(In Chinese). PubMed/NCBI

36 

Rollinger JM, Kratschmar DV, Schuster D, Pfisterer PH, Gumy C, Aubry EM, Brandstötter S, Stuppner H, Wolber G and Odermatt A: 11beta-Hydroxysteroid dehydrogenase 1 inhibiting constituents from Eriobotrya japonica revealed by bioactivity-guided isolation and computational approaches. Bioorg Med Chem. 18:1507–1515. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Banno N, Akihisa T, Tokuda H, Yasukawa K, Higashihara H, Ukiya M, Watanabe K, Kimura Y, Hasegawa J and Nishino H: Triterpene acids from the leaves of Perilla frutescens and their anti-inflammatory and antitumor-promoting effects. Biosci Biotechnol Biochem. 68:85–90. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Kim DH, Han KM, Chung IS, Kim DK, Kim SH, Kwon BM, Jeong TS, Park MH, Ahn EM and Baek NI: Triterpenoids from the flower of Campsis grandiflora K. Schum. as human acyl-CoA: Cholesterol acyltransferase inhibitors. Arch Pharm Res. 28:550–556. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Na M, Yang S, He L, Oh H, Kim BS, Oh WK, Kim BY and Ahn JS: Inhibition of protein tyrosine phosphatase 1B by ursane-type triterpenes isolated from Symplocos paniculata. Planta Med. 72:261–263. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Thuong PT, Min BS, Jin W, Na M, Lee J, Seong R, Lee YM, Song K, Seong Y, Lee HK, et al: Anti-complementary activity of ursane-type triterpenoids from Weigela subsessilis. Biol Pharm Bull. 29:830–833. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Lee MS and Thuong PT: Stimulation of glucose uptake by triterpenoids from Weigela subsessilis. Phytother Res. 24:49–53. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Yang NY, Duan JA, Li P and Qian SH: Chemical Constituents of Glechoma longituba. Yao Xue Xue Bao. 41:431–434. 2006.PubMed/NCBI

43 

Shen Y, Wang QH, Lin HW, Shu W, Zhou JB and Li ZY: Study on chemical constituents of Potentilla chinensis Ser. Zhong Yao Cai. 29:237–239. 2006.(In Chinese). PubMed/NCBI

44 

Kang SH, Shi YQ and Yang CX: Triterpenoids and steroids of root of Rubus biflorus. Zhong Yao Cai. 31:1669–1671. 2008.(In Chinese). PubMed/NCBI

45 

Liu P, Teng J, Zhang YW, Takaishi Y and Duan HQ: Chemical constituents from rhizome of Phlomis umbrosa. Yao Xue Xue Bao. 42:401–404. 2007.(In Chinese). PubMed/NCBI

46 

Li YL, Dai HN, Ma GX, Zhang W, Wu TY, Wang YQ, Zou JM, Zhong XQ, Zhou YL, Yuan JQ, et al: A new triterpenic acid from the roots of Rosa laevigata. Yao Xue Xue Bao. 52:425–429. 2017.(In Chinese). PubMed/NCBI

47 

Huang XY, Ma GX, Zhong XQ, Zhou YL, Dai HN, Wu HF, Zhu YD, Yang JS, Yuan JQ and Xu XD: Triterpene constituents from Rosa cymosa Tratt. Zhongguo Zhong Yao Za Zhi. 39:4637–4641. 2014.(In Chinese). PubMed/NCBI

48 

Ku CY, Wang YR, Lin HY, Lu SC and Lin JY: Corosolic acid inhibits hepatocellular carcinoma cell migration by targeting the VEGFR2/Src/FAK pathway. PLoS One. 10:e01267252015. View Article : Google Scholar : PubMed/NCBI

49 

Cheng QL, Li HL, Li YC, Liu ZW, Guo XH and Cheng YJ: CRA(Crosolic Acid) isolated from Actinidia valvata Dunn. Radix induces apoptosis of human gastric cancer cell line BGC823 in vitro via down-regulation of the NF-κB pathway. Food Chem Toxicol. 105:475–485. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Manayi A, Saeidnia S, Ostad SN, Hadjiakhoondi A, Ardekani MR, Vazirian M, Akhtar Y and Khanavi M: Chemical constituents and cytotoxic effect of the main compounds of Lythrum salicaria L. Z Naturforsch C J Biosci. 68:367–375. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Kim JH, Kim YH, Song GY, Kim DE, Jeong YJ, Liu KH, Chung YH and Oh S: Ursolic acid and its natural derivative corosolic acid suppress the proliferation of APC-mutated colon cancer cells through promotion of β-catenin degradation. Food Chem Toxicol. 67:87–95. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Horlad H, Fujiwara Y, Takemura K, Ohnishi K, Ikeda T, Tsukamoto H, Mizuta H, Nishimura Y, Takeya M and Komohara Y: Corosolic acid impairs tumor development and lung metastasis by inhibiting the immunosuppressive activity of myeloid-derived suppressor cells. Mol Nutr Food Res. 57:1046–1054. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Yoo KH, Park JH, Lee DY, Hwang-Bo J, Baek NI and Chung IS: Corosolic acid exhibits anti-angiogenic and anti-lymphangiogenic effects on in vitro endothelial cells and on an in vivo CT-26 colon carcinoma animal model. Phytother Res. 29:714–723. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Yang J, Wu R, Li W, Gao L, Yang Y, Li P and Kong AN: The triterpenoid corosolic acid blocks transformation and epigenetically reactivates Nrf2 in TRAMP-C1 prostate cells. Mol Carcinog. 57:512–521. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Yong QX, Jian HZ and Xing SY: Corosolic acid induces potent anti-cancer effects in CaSki cervical cancer cells through the induction of apoptosis, cell cycle arrest and PI3K/Akt signalling pathway. Bangladesh J Pharmacol. 11:453–459. 2016. View Article : Google Scholar

56 

National Cancer Institute (NIH), . Surveillance, Epidemiology, and End Results (SEER) Program. NIH. 2016.simplehttps://seer.cancer.gov/

57 

Baffy G: Hepatocellular carcinoma in non-alcoholic fatty liver disease: Epidemiology, pathogenesis, and prevention. J Clin Transl Hepatol. 1:131–137. 2013.PubMed/NCBI

58 

Wong SW, Ting YW and Chan WK: Epidemiology of non-alcoholic fatty liver disease-related hepatocellular carcinoma and its implications. JGH Open. 2:235–241. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Rinella ME: Nonalcoholic fatty liver disease: A systematic review. JAMA. 313:2263–2273. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Starley BQ, Calcagno CJ and Harrison SA: Nonalcoholic fatty liver disease and hepatocellular carcinoma: A weighty connection. Hepatology. 51:1820–1832. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Welzel TM, Graubard BI, Zeuzem S, El-Serag HB, Davila JA and Mcglynn KA: Metabolic syndrome increases the risk of primary liver cancer in the United States: A study in the SEER-medicare database. Hepatology. 54:463–471. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Hotamisligil GS: Inflammation and metabolic disorders. Nature. 444:860–867. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Stickel F and Hellerbrand C: Non-alcoholic fatty liver disease as a risk factor for hepatocellular carcinoma: Mechanisms and implications. Gut. 59:1303–1307. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, Osterreicher CH, Takahashi H and Karin M: Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 140:197–208. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Ish-Shalom D, Christoffersen CT, Vorwerk P, Sacerdoti-Sierra N, Shymko RM, Naor D and De Meyts P: Mitogenic properties of insulin and insulin analogues mediated by the insulin receptor. Diabetologia. 40 (Suppl 2):S25–S31. 1997. View Article : Google Scholar : PubMed/NCBI

66 

Buzzelli G, Dattolo P, Pinzani M, Brocchi A, Romano S and Gentilini P: Circulating growth hormone and insulin-like growth factor-I in nonalcoholic liver cirrhosis with or without superimposed hepatocarcinoma: Evidence of an altered circadian rhythm. Am J Gastroenterol. 88:1744–1748. 1993.PubMed/NCBI

67 

Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal KT, Maeda K, Karin M and Hotamisligil GS: A central role for JNK in obesity and insulin resistance. Nature. 420:333–336. 2002. View Article : Google Scholar : PubMed/NCBI

68 

Chang Q, Zhang Y, Beezhold KJ, Bhatia D, Zhao H, Chen J, Castranova V, Shi X and Chen F: Sustained JNK1 activation is associated with altered histone H3 methylations in human liver cancer. J Hepatol. 50:323–233. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Yang S, Zhu H, Li Y, Lin H, Gabrielson K, Trush MA and Diehl AM: Mitochondrial adaptations to obesity-related oxidant stress. Arch Biochem Biophys. 378:259–268. 2000. View Article : Google Scholar : PubMed/NCBI

70 

Ikura Y, Mita E and Nakamori S: Hepatocellular carcinomas can develop in simple fatty livers in the setting of oxidative stress. Pathology. 43:167–168. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Ma C, Kesarwala AH, Eggert T, Medina-Echeverz J, Kleiner DE, Jin P, Stroncek DF, Terabe M, Kapoor V, ElGindi M, et al: NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature. 531:253–257. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Wolf MJ, Adili A, Piotrowitz K, Abdullah Z, Boege Y, Stemmer K, Ringelhan M, Simonavicius N, Egger M, Wohlleber D, et al: Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell. 26:549–564. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Stohs SJ, Miller H and Kaats GR: A review of the efficacy and safety of banaba (Lagerstroemia speciosa L.) and corosolic acid. Phytother Res. 26:317–324. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Sharma H, Kumar P, Deshmukh RR, Bishayee A and Kumar S: Pentacyclic triterpenes: New tools to fight metabolic syndrome. Phytomedicine. 50:166–177. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Fife CM, McCarroll JA and Kavallaris M: Movers and shakers: Cell cytoskeleton in cancer metastasis. Br J Pharmacol. 171:5507–5523. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Chiang AC and Massagué J: Molecular basis of metastasis. N Engl J Med. 359:2814–2823. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Zhang L, Wang JN, Tang JM, Kong X, Yang JY, Zheng F, Guo LY, Huang YZ, Zhang L, Tian L, et al: VEGF is essential for the growth and migration of human hepatocellular carcinoma cells. Mol Biol Rep. 39:5085–5093. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Lang SA, Brecht I, Moser C, Obed A, Batt D, Schlitt HJ, Geissler EK and Stoeltzing O: Dual inhibition of Raf and VEGFR2 reduces growth and vascularization of hepatocellular carcinoma in an experimental model. Langenbecks Arch Surg. 393:333–341. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Zhang C, Wu X, Zhang M, Zhu L, Zhao R, Xu D, Lin Z, Liang C, Chen T, Chen L, et al: Small molecule R1498 as a well-tolerated and orally active kinase inhibitor for hepatocellular carcinoma and gastric cancer treatment via targeting angiogenesis and mitosis pathways. PLoS One. 8:e652642013. View Article : Google Scholar : PubMed/NCBI

80 

Xu Y, Zhao Y, Xu Y, Guan Y, Zhang X, Chen Y, Wu Q, Zhu G, Chen Y, Sun F, et al: Blocking inhibition to YAP by ActinomycinD enhances anti-tumor efficacy of Corosolic acid in treating liver cancer. Cell Signal. 29:209–217. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Lee MS, Cha EY, Thuong PT, Kim JY, Ahn MS and Sul JY: Down-regulation of human epidermal growth factor receptor 2/neu oncogene by corosolic acid induces cell cycle arrest and apoptosis in NCI-N87 human gastric cancer cells. Biol Pharm Bull. 33:931–937. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Lee MS, Lee CM, Cha EY, Thuong PT, Bae K, Song IS, Noh SM and Sul JY: Activation of AMP-activated protein kinase on human gastric cancer cells by apoptosis induced by corosolic acid isolated from Weigela subsessilis. Phytother Res. 24:1857–1861. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Sung B, Kang YJ, Kim DH, Hwang SY, Lee Y, Kim M, Yoon JH, Kim CM, Chung HY and Kim ND: Corosolic acid induces apoptotic cell death in HCT116 human colon cancer cells through a caspase-dependent pathway. Int J Mol Med. 33:943–949. 2014. View Article : Google Scholar : PubMed/NCBI

84 

Ohta T, Iijima K, Miyamoto M, Nakahara I, Tanaka H, Ohtsuji M, Suzuki T, Kobayashi A, Yokota J, Sakiyama T, et al: Loss of Keap1 function activates Nrf2 and provides advantages for lung cancer cell growth. Cancer Res. 68:1303–1309. 2008. View Article : Google Scholar : PubMed/NCBI

85 

DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES, et al: Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 475:106–109. 2011. View Article : Google Scholar : PubMed/NCBI

86 

Mitsuishi Y, Taguchi K, Kawatani Y, Shibata T, Nukiwa T, Aburatani H, Yamamoto M and Motohashi H: Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell. 22:66–79. 2012. View Article : Google Scholar : PubMed/NCBI

87 

Jia Y, Wang H, Wang Q, Ding H, Wu H and Pan H: Silencing Nrf2 impairs glioma cell proliferation via AMPK-activated mTOR inhibition. Biochem Biophys Res Commun. 469:665–671. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Zhu J, Wang H, Chen F, Fu J, Xu Y, Hou Y, Kou HH, Zhai C, Nelson MB, Zhang Q, et al: An overview of chemical inhibitors of the Nrf2-ARE signaling pathway and their potential applications in cancer therapy. Free Radic Biol Med. 99:544–556. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Ma B, Zhang H, Wang Y, Zhao A, Zhu Z, Bao X, Sun Y, Li L and Zhang Q: Corosolic acid, a natural triterpenoid, induces ER stress-dependent apoptosis in human castration resistant prostate cancer cells via activation of IRE-1/JNK, PERK/CHOP and TRIB3. J Exp Clin Cancer Res. 37:2102018. View Article : Google Scholar : PubMed/NCBI

90 

Woo SM, Seo SU, Min KJ, Im SS, Nam JO, Chang JS, Kim S, Park JW and Kwon TK: Corosolic acid induces non-apoptotic cell death through generation of lipid reactive oxygen species production in human renal carcinoma caki cells. Int J Mol Sci. 19:13092018. View Article : Google Scholar

91 

Xu Y, Ge R, Du J, Xin H, Yi T, Sheng J, Wang Y and Ling C: Corosolic acid induces apoptosis through mitochondrial pathway and caspase activation in human cervix adenocarcinoma HeLa cells. Cancer Lett. 284:229–237. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Takaishi K, Komohara Y, Tashiro H, Ohtake H, Nakagawa T, Katabuchi H and Takeya M: Involvement of M2-polarized macrophages in the ascites from advanced epithelial ovarian carcinoma in tumor progression via Stat3 activation. Cancer Sci. 101:2128–2136. 2010. View Article : Google Scholar : PubMed/NCBI

93 

Komohara Y, Horlad H, Ohnishi K, Ohta K, Makino K, Hondo H, Yamanaka R, Kajiwara K, Saito T, Kuratsu J and Takeya M: M2 macrophage/microglial cells induce activation of Stat3 in primary central nervous system lymphoma. J Clin Exp Hematop. 51:93–99. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Komohara Y, Horlad H, Ohnishi K, Fujiwara Y, Bai B, Nakagawa T, Suzu S, Nakamura H, Kuratsu J and Takeya M: Importance of direct macrophage-tumor cell interaction on progression of human glioma. Cancer Sci. 103:2165–2172. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Fujiwara Y, Takaishi K, Nakao J, Ikeda T, Katabuchi H, Takeya M and Komohara Y: Corosolic acid enhances the antitumor effects of chemotherapy on epithelial ovarian cancer by inhibiting signal transducer and activator of transcription 3 signaling. Oncol Lett. 6:1619–1623. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Fujiwara Y, Komohara Y, Ikeda T and Takeya M: Corosolic acid inhibits glioblastoma cell proliferation by suppressing the activation of signal transducer and activator of transcription-3 and nuclear factor-kappa B in tumor cells and tumor-associated macrophages. Cancer Sci. 102:206–211. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Cai X, Zhang H, Tong D, Tan Z, Han D, Ji F and Hu W: Corosolic acid triggers mitochondria and caspase-dependent apoptotic cell death in osteosarcoma MG-63 cells. Phytother Res. 25:1354–1361. 2011. View Article : Google Scholar : PubMed/NCBI

98 

Jia Y, Yuan H, Shan S, Xu G, Yu J, Zhao C and Mou X: Corosolic acid inhibits the proliferation of osteosarcoma cells by inducing apoptosis. Oncol Lett. 12:4187–4194. 2016. View Article : Google Scholar : PubMed/NCBI

99 

Nho KJ, Chun JM and Kim HK: Corosolic acid induces apoptotic cell death in human lung adenocarcinoma A549 cells in vitro. Food Chem Toxicol. 56:8–17. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Bahadori MB, Vandghanooni S, Dinparast L, Eskandani M, Ayatollahi SA, Ata A and Nazemiyeh H: Triterpenoid corosolic acid attenuates HIF-1 stabilization upon cobalt (II) chloride-induced hypoxia in A549 human lung epithelial cancer cells. Fitoterapia. 134:493–500. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Wang K, Zhu X, Yao Y, Yang M, Zhou F and Zhu L: Corosolic acid induces cell cycle arrest and cell apoptosis in human retinoblastoma Y-79 cells via disruption of MELK-FoxM1 signaling. Oncol Rep. 39:2777–2786. 2018.PubMed/NCBI

102 

Fujiwara Y, Takeya M and Komohara Y: A novel strategy for inducing the antitumor effects of triterpenoid compounds: Blocking the protumoral functions of tumor-associated macrophages via STAT3 inhibition. Biomed Res Int. 2014:3485392014. View Article : Google Scholar : PubMed/NCBI

103 

Tseng LM, Huang PI, Chen YR, Chen YC, Chou YC, Chen YW, Chang YL, Hsu HS, Lan YT, Chen KH, et al: Targeting signal transducer and activator of transcription 3 pathway by cucurbitacin I diminishes self-renewing and radiochemoresistant abilities in thyroid cancer-derived CD133+ cells. J Pharmacol Exp Ther. 341:410–423. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Zhang X, Liu P, Zhang B, Wang A and Yang M: Role of STAT3 decoy oligodeoxynucleotides on cell invasion and chemosensitivity in human epithelial ovarian cancer cells. Cancer Genet Cytogenet. 197:46–53. 2010. View Article : Google Scholar : PubMed/NCBI

105 

Page BD, Ball DP and Gunning PT: Signal transducer and activator of transcription 3 inhibitors: A patent review. Expert Opin Ther Pat. 21:65–83. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Lee HS, Park JB, Lee MS, Cha EY, Kim JY and Sul JY: Corosolic acid enhances 5-fluorouracil-induced apoptosis against SNU-620 human gastric carcinoma cells by inhibition of mammalian target of rapamycin. Mol Med Rep. 12:4782–4788. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Park JB, Lee JS, Lee MS, Cha EY, Kim S and Sul JY: Corosolic acid reduces 5-FU chemoresistance in human gastric cancer cells by activating AMPK. Mol Med Rep. 18:2880–2888. 2018.PubMed/NCBI

108 

Nelson AT, Camelio AM, Claussen KR, Cho J, Tremmel L, Digiovanni J and Siegel D: Synthesis of oxygenated oleanolic and ursolic acid derivatives with anti-inflammatory properties. Bioorg Med Chem Lett. 25:4342–4346. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Yamaguchi Y, Yamada K, Yoshikawa N, Nakamura K, Haginaka J and Kunitomo M: Corosolic acid prevents oxidative stress, inflammation and hypertension in SHR/NDmcr-cp rats, a model of metabolic syndrome. Life Sci. 79:2474–2479. 2006. View Article : Google Scholar : PubMed/NCBI

110 

Chen H, Yang J, Zhang Q, Chen LH and Wang Q: Corosolic acid ameliorates atherosclerosis in apolipoprotein E-deficient mice by regulating the nuclear factor-κB signaling pathway and inhibiting monocyte chemoattractant protein-1 expression. Circ J. 76:995–1003. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Kim SJ, Cha JY, Kang HS, Lee JH, Ji YL, Park JH, Bae JH, Song DK and Im SS: Corosolic acid ameliorates acute inflammation through inhibition of IRAK-1 phosphorylation in macrophages. BMB Rep. 49:276–281. 2016. View Article : Google Scholar : PubMed/NCBI

112 

Festa A, D'Agostino R Jr, Howard G, Mykkänen L, Tracy RP and Haffner SM: Chronic subclinical inflammation as part of the insulin resistance syndrome: The insulin resistance atherosclerosis study (IRAS). Circulation. 102:42–47. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao J, Zhou H, An Y, Shen K and Yu L: Biological effects of corosolic acid as an anti‑inflammatory, anti‑metabolic syndrome and anti‑neoplasic natural compound (Review). Oncol Lett 21: 84, 2021
APA
Zhao, J., Zhou, H., An, Y., Shen, K., & Yu, L. (2021). Biological effects of corosolic acid as an anti‑inflammatory, anti‑metabolic syndrome and anti‑neoplasic natural compound (Review). Oncology Letters, 21, 84. https://doi.org/10.3892/ol.2020.12345
MLA
Zhao, J., Zhou, H., An, Y., Shen, K., Yu, L."Biological effects of corosolic acid as an anti‑inflammatory, anti‑metabolic syndrome and anti‑neoplasic natural compound (Review)". Oncology Letters 21.2 (2021): 84.
Chicago
Zhao, J., Zhou, H., An, Y., Shen, K., Yu, L."Biological effects of corosolic acid as an anti‑inflammatory, anti‑metabolic syndrome and anti‑neoplasic natural compound (Review)". Oncology Letters 21, no. 2 (2021): 84. https://doi.org/10.3892/ol.2020.12345