Next Article in Journal
Celebrating Todd Marder: 65th Birthday and His Contributions to Inorganic Chemistry
Next Article in Special Issue
Recent Developments in Small-Molecule Ligands of Medicinal Relevance for Harnessing the Anticancer Potential of G-Quadruplexes
Previous Article in Journal
Mechanical and Morphological Properties of Bio-Phenolic/Epoxy Polymer Blends
Previous Article in Special Issue
Identification of Phytochemical-Based β-Catenin Nuclear Localization Inhibitor in NSCLC: Differential Targeting Population from Member of Isothiocyanates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Insight into the Anti-Angiogenic and Anti-Metastatic Effects of Oridonin: Current Knowledge and Future Potential

by
Nurul Akmaryanti Abdullah
1,*,
Nur Fariesha Md Hashim
1,*,
Aula Ammar
2 and
Noraina Muhamad Zakuan
1
1
Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
2
Wolfson Wohl Translational Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow City G61 1BD, UK
*
Authors to whom correspondence should be addressed.
Submission received: 3 December 2020 / Revised: 5 January 2021 / Accepted: 6 January 2021 / Published: 3 February 2021
(This article belongs to the Special Issue Recent Advances in Anticancer Drugs II)

Abstract

:
Cancer is one of the leading causes of death worldwide, with a mortality rate of more than 9 million deaths reported in 2018. Conventional anti-cancer therapy can greatly improve survival however treatment resistance is still a major problem especially in metastatic disease. Targeted anti-cancer therapy is increasingly used with conventional therapy to improve patients’ outcomes in advanced and metastatic tumors. However, due to the complexity of cancer biology and metastasis, it is urgent to develop new agents and evaluate the anti-cancer efficacy of available treatments. Many phytochemicals from medicinal plants have been reported to possess anti-cancer properties. One such compound is known as oridonin, a bioactive component of Rabdosia rubescens. Several studies have demonstrated that oridonin inhibits angiogenesis in various types of cancer, including breast, pancreatic, lung, colon and skin cancer. Oridonin’s anti-cancer effects are mediated through the modulation of several signaling pathways which include upregulation of oncogenes and pro-angiogenic growth factors. Furthermore, oridonin also inhibits cell migration, invasion and metastasis via suppressing epithelial-to-mesenchymal transition and blocking downstream signaling targets in the cancer metastasis process. This review summarizes the recent applications of oridonin as an anti-angiogenic and anti-metastatic drug both in vitro and in vivo, and its potential mechanisms of action.

1. Introduction

Cancer metastasis is considered the main cause of cancer mortality [1]. Even though metastatic cancer can be treated, this disease is generally considered incurable with a low survival rate [1,2]. Tumor metastasis is a dynamic multistep event that involves changes in various biochemical, genetic and epigenetic factors in the primary tumor that contributes to the invasion–metastasis cascade. This series of metastatic cascade begins with the invasion of the primary tumor cells into the surrounding tissues which is governed by the epithelial-to-mesenchymal transition (EMT) followed by the tumor cells entering the circulatory system and extravasating through the vascular walls into the parenchyma of distant tissues to form secondary cancers [3]. Tumor vascularization and angiogenesis are required for the dissemination and establishment of cancer metastasis [4]. According to Weinberg and Hanahan, inducing angiogenesis and the ability to invade surrounding tissues and metastasize are hallmarks of tumor malignancy [4].
New agents that present anti-cancer effects are largely tested for their ability to cause tumor shrinkage which focuses mainly on inhibiting cell division and proliferation. However, in advanced stages, cancer cells begin to invade the extracellular matrix (ECM), induce the EMT process, and create secondary tumors. The current cancer treatment strategies such as chemotherapy are often associated with adverse side effects and greatly affects the quality of life. Moreover, treatment resistance is also often observed when cancer is presented at an advanced stage [5]. It is therefore important to vigorously identify new agents with low susceptibility to drug resistance and to explicitly block key molecules or their downstream signaling targets in the cancer metastasis process.
Natural products derived from Chinese medicinal plants have recently attracted a growing interest in the treatment of cancer, especially those with low side effects such as shikonin, berberine, and curcumin [6]. In China, Rabdosia rubescens is used as an alternative medicine to treat sore throat, gingivitis, and rheumatoid arthritis [7,8]. Oridonin (C20H28O6) (Figure 1) is an active diterpenoid component from Rabdosia rubescens first identified in 1966 by Furida and colleagues [9]. Oridonin possesses many therapeutic potentials such as neuroprotective [10], anti-inflammatory [11] and antibacterial [12] effects. In recent years, the anti-cancer properties of oridonin were studied in a wide range of tumors including breast [13], colon [14], pancreatic [15], lung [16], gastric [17], prostate [18] and skin [19] cancer. This review discusses the different molecular pathways involved in angiogenesis, cell invasion, and metastasis that can be targeted by oridonin.

2. The Process of Angiogenesis as a Target of Oridonin

Tumor angiogenesis is a highly regulated process through which new blood vessels form in the tumor environment to supply oxygen and support tumor growth [20]. In this process, extensive interplays between endothelial cells, angiogenic growth factors, and ECM is required [20,21]. In tumor angiogenesis, various pro-angiogenic signals such as vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and transforming growth factor (TGF) are released to induce the formation of blood vessels and to support tumor proliferation [20,22]. Such a process is also referred to as the ‘angiogenic switch’ and can occur at different stages of tumor progression as a result of genetic or microenvironmental changes [23]. During the switch, tumors in dormancy re-enter the cell cycle and become actively growing malignant cells [23,24]. One of the angiogenic models includes new vessels sprouting from pre-existing ones in the tumor microenvironment [25]. In this process, endothelial cells emerge towards an angiogenic stimulus, and endothelial cells involved are classified into tip and stalk cells [25]. The tip cells are responsible for ECM degradation while the stalk cells facilitate vascular lumen formation [26,27]. During sprouting angiogenesis, VEGF and Notch signaling are activated to guide the vascular patterning by directing the tip cell migration and stalk cell proliferation. VEGF can induce jagged ligands which then increases Notch expression in cancer endothelial cells to promote Notch-dependent angiogenesis [28]. VEGF also activates other signaling cascades such as phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinase. After the establishment of new blood vessels, platelet-derived growth factor β (PDGF) is secreted to assist the maturation process of blood vessels [25]. In addition to sprouting angiogenesis, cancer cells use vascular mimicry to acquire blood supply [29]. Highly invasive tumors can differentiate into endothelial cells and induced tube-like structures. This allows tumors to have a secondary circulatory system that is not dependent on angiogenesis [29].
Based on the understanding of the diverse pathways involved in angiogenesis, strategies to inhibit the formation of new blood vessels in tumors can be applied at different stages such as targeting the proangiogenic factors or disrupt the active dividing endothelial cells. Over the years, many anti-angiogenic inhibitors have been developed such as bevacizumab [30] and sunitinib which target VEGF pathways [31]. VEGF is one of the most extensively studied angiogenic factors and a key mediator in tumor angiogenesis. The VEGF family has at least seven isoforms (i.e., VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, placental growth factor, and VEGF-F) that bind to the tyrosine kinase receptor known as vascular endothelial growth factor receptor (VEGFR) [32]. High expression of VEGF has been reported in many cancers such as breast [33], prostate [34], and ovarian [35] cancer.
Targeting angiogenesis has so far shown limited success, which may be attributed to the heterogeneity of blood vessels in the tumor environment and tumor hypoxia [20]. The newly formed blood vessel in tumor is usually abnormal in structure with immature and leaky blood vessels. Unlike normal blood vessels, the tumor vessels are narrower in diameter, have diverse vessel density, and high permeability [36]. Moreover, tumor blood vessels are not efficient in delivering oxygen and removal of waste products which can lead to an aggressive tumor microenvironment [20]. As a result of the abnormality of tumor blood vessels, the efficacy of anti-cancer treatment may be decreased due to the inaccessibility of the drug to the tumor area and increased drug resistance [25]. The development of new anti-angiogenic agents with high efficacy and fewer side effects is needed to overcome resistance to the existing agents and improve cancer therapy.
In recent years, oridonin has been shown to be a promising anti-angiogenic agent. Dong and coworkers reported that oridonin inhibits angiogenesis by blocking VEGF-induced micro-vessels sprouting. In human umbilical vein endothelial cells (HUVEC) cells, oridonin treatment reduced more than 90% tubular formation [37]. In another study, Jiang and colleagues reported that VEGF-induced migration was reduced following oridonin treatment. The tubular formation was also decreased by 70% in the oridonin treated group when compared to the control group. Also, oridonin treatment has resulted in irregular and disorganized tube formation and the depolymerization of F-actin [38]. Vascular assay in zebrafish embryos showed that oridonin reduced the diameter of the complete intersegment vessels when compared to the control group [39]. Taken together, these suggest that oridonin may interfere with capillary network formation and actin organization.
The key targets of oridonin to suppress angiogenesis include VEGF, Notch, and PI3K signaling pathways. Oridonin has been shown to inhibit the expression of the VEGF family such as VEGF-A, VEGFR-2, and VEGFR-3 [39,40,41]. In endothelial cells, when VEGF binds to its receptor, the VEGF/PI3K signaling pathway is activated to induce vessel formation [42]. Oridonin also inhibited the VEGF-induced Notch activation by reducing the expression of key ligand and downstream genes including Jagged-1 and -2, Notch 1, Hes-1, HESR-1, and DII-1 [37]. The inhibition of VEGF expression by oridonin ultimately leads to the suppression of angiogenesis due to the inactivation of its downstream targets.
The combination treatment of chemotherapy agents with anti-angiogenic inhibitors has been shown to improve treatment efficacy in ovarian cancer. For example, the administration of bevacizumab with selected chemotherapy agents such as pegylated liposomal doxorubicin and paclitaxel improved progression-free survival in platinum-resistant ovarian cancer [43]. Li and colleagues explored the potential of an anti-angiogenesis effect of oridonin in combination with doxorubicin [44]. Treatment with doxorubicin alone did not inhibit cell migration and invasion of HUVECS cells. However, oridonin treatment as a single agent resulted in reduced VEGF-induced endothelial cell migration and tube formation. Interestingly, the combination of both compounds synergistically impedes cell migration and invasion of HUVECS cells. Based on the molecular docking study, a combination of the two compounds showed a high affinity towards the ATP-binding domain of VEGFR-2 kinase which suggests that the interaction may inhibit the activation of VEGFR-2 [44]. Figure 2 summarizes the effect of oridonin on angiogenesis.

3. Oridonin in EMT

EMT is a complex biological process in which epithelial transitions into a mesenchymal phenotype and it is one of the important events in driving tumor progression and metastasis [45]. In EMT, epithelial cells lose their epithelial characteristics and gain motile mesenchymal properties such as loss of cell-cell adhesion, augmented motility, and invasiveness [46]. The shift from one state to another is controlled by a range of growth factors and signaling pathways [47]. The strong integration between these growth factors such as TGF-β forms a robust network promoting the growth of cancer cells and that makes EMT a possible target for cancer metastases. There are various ways to target the EMT process in cancer which include (i) blocking the activation signal of EMT markers such as E-cadherin, N-cadherin vimentin, fibronectin, matrix metalloproteinases (MMPs), and TGF-β [48], (ii) reversed the EMT process, and (iii) suppressing the growth EMT-like cells. E-cadherin and N-cadherin share a similar structure and are involved in cell-cell adhesion [46]. On the other hand, vimentin and fibronectin play a role in maintaining cell shape and cell adhesion, respectively [49,50]. The reduced expression of E-cadherin along with the concomitant increased expression of specific mesenchymal markers such as zinc finger E-box binding homeobox 1 (ZEB1), N-cadherin, and vimentin are considered hallmarks of EMT [46,51]. Growing evidence showed that oridonin exhibits anti-metastatic effects by altering the EMT pathway. To date, the effects of oridonin on the EMT pathways have been reported in many cancers such as pancreatic, breast, melanoma, and lung cancer. Previous studies suggest that oridonin increased E-cadherin expression while decreased the expression of ZEB1, N-cadherin, fibronectin, vimentin, snail, and slug [52,53,54]. The MMPs play a pivotal role in cancer cell invasiveness and metastasis by facilitating ECM degradation [55]. As an invasion promoter, MMPs can facilitate EMT through invasion and metastasis behaviors (i.e., regulation of actin cytoskeleton, increase motility, and proliferation) [56]. Various studies demonstrated that oridonin could inhibit the expression of MMP-2 and MMP-9 in various cancers such as breast [13], acute myeloid leukemia [57] bone [58], and ovarian [59] cancer. Oridonin also decreased MMP-12 expression which is an important mediator to degrade ECM in lung cancer [60]. The effects of oridonin on EMT makers and regulators are summarized in Table 1.

4. Cancer Invasion and Metastasis

The invasion-metastatic pathway is a multi-step process that starts with the invasion of the cancer cells into the surrounding tissues (Figure 3). In this process, the release of MMPs, hyaluronidase, and metalloproteinase (ADAM) are required to assist ECM degradation and remodeling [63]. Following ECM degradation, tumor cells then migrate into the blood and lymphatic vessels crossing the endothelial cell barrier and this step is known as intravasation. Cancer cells escape into the circulation either as single circulating tumor cells (CTC) or as clustered CTC. To promote cancer cells transmigration into the circulatory system and protect them from external insults, various growth factors and cytokines are released including tumor necrosis factor 1-α (TNF1-α), TGF-β, VEGF, and EGF [3]. The release of EGF and TGF-β enhances the intravasation process by allowing the cancer cells to cross blood vessels barriers [64]. To establish a secondary tumor at a distant tissue site, cancer cells need to extravasate from the vascular system. The releases of integrins, insulin-like growth factor-1 (IGF-1), VEGF, MMP, PDGF, and help to facilitate cancer cell extravasation [3] (Figure 3). Integrins support the anchorage-independent survival of CTC while VEGF and IGF1 induce vascular permeability to allow them to penetrate endothelial cells and migrate into tissue parenchyma [65]. Metastatic colonization begins once cancer cells survive the stressful processes of intravasation and extravasation. In this stage, cancer cells can either proliferate continuously or enter dormancy [66]. The tumor microenvironment at the metastatic site plays a key role in determining whether cancer cells can survive. Factors such as favorable conditions in ECM, effective vascular system, ability to escape immune system surveillance, and resistance to anoikis will determine whether cancer cells enter dormancy or proliferate into macrometastases [66].
Limited progress has been made in metastatic cancer therapy due to multiple factors such as tumor growth in different organs, the complexity of metastatic cascade, and increased resistance to cytotoxic agents [45]. Furthermore, the survival of patients with metastatic cancer remains poor [2]. Several strategies have been identified to target metastatic cascades. These include targeting the early steps in the metastatic processes, prevent tumor dissemination into the circulatory system, and killing dormant cancer cells [65]. Over the years, various inhibitors targeting key molecules in cancer metastasis have been developed such as MMP, VEGF, and endothelial growth factor receptor (EGFR) inhibitors [65,67]. However, many of these agents failed to show efficacy and safety in clinical trials [68,69]. Identification of an agent that can inhibit a target integral to multiple stages of metastasis can significantly enhance metastasis inhibition and increase patient survival. Oridonin has been reported to exhibit an anti-metastatic effect in cancer. Oridonin inhibits cancer cells invasion and metastasis through targeting various transcription factors and their signaling pathways such as TGF-β1 [70], EGFR [60], mammalian target of rapamycin (mTOR) [71], long non-coding RNAs (lncRNAs) [53] as well as increasing the activity of tumor suppressor genes (i.e., p53 and protein phosphatase 2A (PP2A)) [72]. The effects of oridonin on these targets are discussed below.

4.1. Oridonin Inhibits TGF-β/Smad Pathway

In human cancers, dysregulation of TGF-β is common in many human cancers such as prostate and bone cancer [73,74]. In the early stage of cancer development, TGF-β acts as a tumor suppressor, but as cancer progresses TGF-β enhances cancer cell invasiveness and increases metastases [75]. Smad is activated by phosphorylation via TGF-β1, translocates to the nucleus where it forms a complex that works as a transcription factor and triggers a cascade of gene expression involved in cell migration such as integrins and MMPs [76,77]. In osteosarcoma, 24 h treatment with low concentrations of oridonin (0.5–2 µM) decreased the migration and invasion capabilities of 143B and MG-63 bone cancer cells by significantly inhibiting the phosphorylation of TGF-β and preventing the Smad dimer from translocating into the nucleus [54]. Bu and coworkers also demonstrated that oridonin blocks the activation of the TGF-β1/Smad signaling pathway in colon cancer. In this study, pre-treatment with 8 µg/mL of oridonin for 48 h downregulated the phosphorylation of TGF-β1 downstream effectors like Smad proteins (Smad2, Smad3, and Smad4) and subsequently deactivated plasminogen activator inhibitor type 1 (PAI-1), a molecule regulated by Smad pathway [70].
Oridonin was shown to inhibit the cellular migration and invasion and decrease the number of cells adhesiveness to fibronectin in B16-F10 (mouse) and A375 melanoma cancer (human) cells via downregulation of TGF-β1 expression and inhibition of the PI3K/Akt/GSK-3β signaling pathway [78]. In a recent study, treatment with oridonin (4–16 µM) for 24 h inhibited regulatory T cell (Treg) differentiation in 4T1 murine breast cancer cells via downregulation of the protein levels of TGF-β receptor, Smad2, and Smad3. In vivo study also demonstrated that oridonin at doses of 2.5, 5 and 10 mg/kg reduced Treg phosphorylation thereby suppressing breast cancer growth and progression [79]. Such data suggest that oridonin exhibits an anti-metastatic effect by inhibits TGF-β/Smad pathway.

4.2. Oridonin Blocks the Activation of EGF/EGFR/ERK Signaling Pathways

High expression of EGFR was observed in many tumors such as in lung, breast, and ovarian cancer [80,81]. The aberration in EGFR expression and downstream signaling influenced tumor progression and maintenance of the malignant phenotype [82]. Phosphorylation of EGFR activates the extracellular-signal-regulated kinase (ERK) pathway that is involved in various pathological processes including angiogenesis, migration, and invasion. In lung cancer, treatment with oridonin (0–10 µM) for 24 h suppressed cell migration, invasion, and adhesion of H1975 human non-small cell lung cancer (NSCLC) cell line through suppression of phosphorylation of EGFR and its downstream signaling pathway via ERK [60]. Protein phosphatase 2A (PP2A) is a tumor suppressor gene that inactivates the ERK pathway [83]. Xiao and colleagues demonstrated that oridonin (0–10 µM) increased PP2A activity and inactivated the ERK/Akt pathway which resulted in inhibition of cell migration and invasion of H1975 NSCLC cells after 24 h treatment. The inhibition of the ERK/Akt pathway also promoted apoptosis in these cells [60]. Previous studies have reported that in transformed cells and cancer cell lines, inhibition of PP2A promotes cell motility [84,85]. The activation of the ERK pathway is also mediated by focal adhesion kinase (FAK) [86]. The activation of FAK can lead to increased motility of cancer cells. Wang and colleagues showed that treatment with oridonin (1.25–5 µM) for 24 h inhibits MDA-MB-231 breast cancer cell motility and migration activities via suppression of FAK and integrin β1 expressions [13]. In H1688 NSCLC cell line, oridonin at a concentration of 10 µM markedly inhibited cell migration via downregulating the expression of metalloproteinases and phosphorylated FAK (p-FAK) [61]. The inhibition of EGF/EGFR signaling pathway by oridonin results in the suppression of ERK and FAK leading to a decrease in cell motility, migration, and invasion capacities of cancer cells.

4.3. Oridonin Inhibits the Phosphorylation of mTOR Signaling Pathway

Growing evidence supports the role of mTOR is in cancer cell invasion and metastasis especially by regulating the organization of actin cytoskeleton [87,88]. The anti-invasive and anti-metastatic effects of oridonin via inhibiting the mTOR pathways have been studied in ovarian cancer. The treatment with oridonin at 2.5 to 10 µM for 24 h decreased cell migration and invasion of SKOV3 ovarian cancer cells by blocking the phosphorylation of the mTOR signaling pathway [71,89]. The inhibition of mTOR is also accompanied by the upregulation of forkhead box P3 (FOXP3) following oridonin treatment [89]. FOXP3 plays a role in regulating the function of regulatory T-cell [90]. Moreover, FOXP3 was also reported to play role in cancer metastasis [91]. Previous studies also showed that the inhibition of mTOR by oridonin induced cell apoptosis [92,93]. Such findings suggest that the inhibition of mTOR signaling pathways and the involvement of FOXP3 are essential for the anti-tumorous effect of oridonin.

4.4. Oridonin Downregulates the lncRNA AFAP1-AS1 Expression

The long non-coding RNAs (lncRNAs) are involved in various biological processes including cell differentiation, proliferation, growth, and apoptosis [94]. RNA actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) is a recently identified cancer-associated lncRNA originating from the antisense DNA strand of the AFAP1 coding gene locus. The aberrant expression of lncRNAs is frequently reported in cancer [95]. In pancreatic cancer, overexpression of lncRNA AFAP1-AS1 is associated with low survival and disease progression [96]. As a result, lncRNA AFAP1-AS1 has become a possible target in the treatment of pancreatic cancer. A recent study has shown that treatment with oridonin (95 µM) and/or knockdown of lncRNAs AFAP1-AS1 for 24 h inhibited cell invasion capacity of PANC-1 and BxPC-3 pancreatic cancer cell lines as assessed by transwell migration assay. In lncRNA AFAP1-AS1 knockdown cells, oridonin treatment resulted in fewer cells penetrating through the membranes in transwell assays compared to siAFAP1-AS1 alone. The author suggested that oridonin maintained AFAP1-AS1 inhibition which further decreased the metastasis activity of PANC-1 and BxPC-3 cells [53]. This indicates that oridonin increases its anti-metastatic effect against pancreatic cancer by continuously inhibiting AFAP1-AS1.

4.5. Oridonin Increases the Expression of p53

Any mutations or loss in the p53 tumor suppressor gene can result in uncontrolled cell division, avoidance of apoptosis, and changes in cell migration and polarity [97]. Studies have shown that loss of p53 function is associated with an increase in cell motility and hence facilitates cancer development and metastases [97,98]. The p53 level and activity are mainly mediated by ubiquitin E3 ligase Mdm2, which binds directly to p53 and facilitates p53 ubiquitination and proteasomal degradation [99]. Mdm2 is known as the principal negative regulator of p53 [99]. Treatment with oridonin (10–80 µM) for 24 h was shown to suppress the migration activity of SNU-16 gastric cancer cells via apoptosis, increase the expression of p53, and downregulate the expression of Mdm2. The inhibitory effect of oridonin was reversed in a stable knockdown of p53 by siRNA in SNU-16 cells validating the above observation [72]. A previous study reported that p53 mutations occur at a late stage in cancer progression pathway in 56% of gastric carcinoma cases [100]. The mutational spectrum of p53 in gastric cancer is wide however, the most common sites occur at codon 175, 213, 245, 248, 273, and 282 [101]. An increase in p53 level and activity following oridonin treatment (20–40 µM) was also observed in other cancer such as neuroblastoma [102] and esophageal [103] cancer. Taken together, these data propose that oridonin exert its anti-cancer activity by enhancing p53 protein expression in cancer cells and promotes apoptosis.

5. Oridonin in Hypoxia

Hypoxia is the main feature of solid tumors and it arises as a result of uncontrolled proliferation of cancer cells which limit the availability of oxygen supply and, often associated with poor overall survival [104,105]. The unorganized vascular networks at the tumor site make a significant contribution to a reduced level of oxygen in the solid tumor [104]. In metastatic cancer, hypoxia serves as one of the main drivers for cancer to develop a more aggressive and resistant phenotype as well as enhanced survival in a nutrient-deprived environment [106,107]. This is because hypoxia increases the expression of many angiogenic inflammatory markers and growth factors in cancer cells particularly VEGF-A [108]. Besides, hypoxia may induce EMT by promoting the transcription of EMT markers such as Snail, ZEB1, and TWIST [107]. The activation of hypoxia-induced EMT markers promotes cancer cell motility, migration, and invasion, resulting in tumor progression and metastases [109]. Hypoxia-inducible factor 1 (HIF-1α) is a major transcriptional regulator in hypoxia which can directly or indirectly regulate EMT markers [109]. For instance, HIF-1α may activate N-cadherin and vimentin to mediate EMT by promoting the loss of cell-cell adhesion which subsequently results in more migratory and invasive cancer cells. HIF-1α has been considered a therapeutic target for the treatment of cancer metastasis by inhibiting HIF-1 and its downstream molecules using bioreductive drugs and gene therapy [110]. Bioreductive drugs are inactive agents that undergo biotransformation to generate highly reactive electrophiles through enzymatic reduction catalyzed by endogenous oxidoreductases. The activation of such agents occurs in hypoxic regions where oxygen levels are low [111]. Examples of these agents include tirapazamine, mitomycin C and E09. These agents, however, showed limited success in clinical trials due to toxicity and rapid clearance [112,113]. Therefore, it is important to identify new agents that can target hypoxia without inducing severe toxicity. Oridonin has been shown to inhibit hypoxia-induced migration and EMT via targeting HIF-1α. In human gallbladder cancer cells (GBC-SD), treatment with 5 µM of oridonin for 24 h significantly inhibited EMT and reversed hypoxia-induced migration via downregulation of HIF-1α/MMP-9 signaling pathways. Similarly, in tumor xenograft tissue, treatment with 5 µM of oridonin suppressed the protein expression of HIF-1α and MMP-9 [114]. In MDA-MB-231 and 4T1 breast cancer cells, treatment with increasing concentration of oridonin (0–16 µM) for 24 h inhibited angiogenesis and cell migration via downregulation of HIF-1α protein expression [40]. Taken together, these findings suggest that, through downregulation of HIF-1α protein expression, oridonin is able to suppress cell migration, angiogenesis and EMT.

6. Conclusions and Future Perspectives

Disturbance of metastases processes carries a great amount of clinical significance for patients with or at risk of developing metastatic cancer. Herbal medicines have long been an essential source for the discovery and development of new drugs against human diseases. The use of natural products in the treatment of cancer has gained the attention of the research community due to low cost and few side effects. In this review, we have summarized many potential therapeutic advantages of oridonin particularly in targeting angiogenesis and metastasis. The anti-angiogenic effect of oridonin is observed in its ability to target VEGF and suppress the formation of blood vessels. Moreover, when combined with other chemotherapy drugs such as doxorubicin, the effectiveness of treatment is improved. The ability of oridonin to interrupt several metastasis pathways holds preclinical promise for its development as a potential anti-metastatic agent in clinical settings. Oridonin has been shown to inhibit cell migration and invasion by targeting several metastatic signaling pathways. However, as tumor metastasis is a complex disease, more studies on other metastatic pathways should be considered. Over the years, researchers also have developed novel oridonin analogs such as HAO472 [115], CYD-6-17 [116], 1-O- and 14-O-derivative compounds [117]. The development of these novel analogs is aimed to improve oridonin’s water solubility and therapeutic efficacy. Many of these agents, however, have not been tested for their anti-angiogenic and anti-metastatic effects. Additional research is warranted to explore the potential therapeutic benefits of oridonin and its derivatives in the management of metastatic cancer especially in animal models and clinical trials.

Author Contributions

N.A.A.—conceptualization, data curation, writing, revised, and edited the manuscript; N.F.M.H., N.M.Z., and A.A.—provide intellectual inputs, revised and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EMTepithelial-to-mesenchymal transition
ECMextracellular matrix
VEGFvascular endothelial growth factor
EGFepidermal growth factor
TGFtransforming growth factor
VEGFRvascular endothelial growth factor receptor
MMPmatrix metalloproteinases
ZEB1zinc finger E-box binding homeobox 1
ADAMmetalloproteinase
CTCcirculating tumor cells
TNF1-αtumor necrosis factor 1-α
PDGFplatelet-derived growth factor
IGF-1insulin-like growth factor-1
lncRNAslong non-coding RNAs
EGFRendothelial growth factor receptor
mTORmammalian target of rapamycin
NSCLChuman non-small cell lung cancer
ERKextracellular-signal-regulated kinase
PP2Aprotein phosphatase 2A
FAKfocal adhesion kinase
FOXP3forkhead box P3
HIF-1αhypoxia-inducible factor 1

References

  1. Dillekås, H.; Rogers, M.S.; Straume, O. Are 90% of deaths from cancer caused by metastases? Cancer Med. 2019, 8, 5574–5576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. American Cancer Society. Cancer Facts and Figures; American Cancer Society (ACS): Atlanta, GA, USA, 2020. [Google Scholar]
  3. Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging biological principles of metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rivera, E.; Gomez, H. Chemotherapy resistance in metastatic breast cancer: The evolving role of ixabepilone. In Proceedings of the Breast Cancer Research; Springer: Berlin/Heidelberg, Germany, 2010; Volume 12, p. S2. [Google Scholar]
  6. Luo, H.; Vong, C.T.; Chen, H.; Gao, Y.; Lyu, P.; Qiu, L.; Zhao, M.; Liu, Q.; Cheng, Z.; Zou, J.; et al. Naturally occurring anti-cancer compounds: Shining from Chinese herbal medicine. Chin. Med. 2019, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Chen, S.; Liu, J.; Zhang, H. Efficacy of rabdosia rubescens in the treatment of gingivitis. J. Huazhong Univ. Sci. Technol. Med. Sci. 2009, 29, 659. [Google Scholar] [CrossRef] [PubMed]
  8. Peng, M.; Liu, B.; Mao, M. Study on the Application of Chinese Patent Drug and Chinese Formula of Rabdosia Rubescens. IOP Conf. Ser. Mater. Sci. Eng. 2018, 301, 12060. [Google Scholar] [CrossRef]
  9. Fujita, E.; Fujita, T.; Shibuya, M. Diterpenoid constituents of Isodon trichocarpus and Isodon japonicus (terpenoids IV). Tetrahedron Lett. 1966, 7, 3153–3162. [Google Scholar] [CrossRef]
  10. Wang, S.; Yu, L.; Yang, H.; Li, C.; Hui, Z.; Xu, Y.; Zhu, X. Oridonin attenuates synaptic loss and cognitive deficits in an Aβ1–42-induced mouse model of Alzheimer’s disease. PLoS ONE 2016, 11, e0151397. [Google Scholar] [CrossRef] [PubMed]
  11. He, H.; Jiang, H.; Chen, Y.; Ye, J.; Wang, A.; Wang, C.; Liu, Q.; Liang, G.; Deng, X.; Jiang, W. Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat. Commun. 2018, 9, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Yuan, Z.; Ouyang, P.; Gu, K.; Rehman, T.; Zhang, T.; Yin, Z.; Fu, H.; Lin, J.; He, C.; Shu, G. The antibacterial mechanism of oridonin against methicillin-resistant Staphylococcus aureus (MRSA). Pharm. Biol. 2019, 57, 710–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wang, S.; Zhong, Z.; Wan, J.; Tan, W.; Wu, G.; Chen, M.; Wang, Y. Oridonin induces apoptosis, inhibits migration and invasion on highly-metastatic human breast cancer cells. Am. J. Chin. Med. 2013, 41, 177–196. [Google Scholar] [CrossRef] [PubMed]
  14. Yao, Z.; Xie, F.; Li, M.; Liang, Z.; Xu, W.; Yang, J.; Liu, C.; Li, H.; Zhou, H.; Qu, L.-H. Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death Dis. 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, B.; Shen, C.; Li, Y.; Zhang, T.; Huang, H.; Ren, J.; Hu, Z.; Xu, J.; Xu, B. Oridonin overcomes the gemcitabine resistant PANC-1/Gem cells by regulating GST pi and LRP/1 ERK/JNK signalling. OncoTargets Ther. 2019, 12, 5751–5765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Xu, L.; Bi, Y.; Xu, Y.; Zhang, Z.; Xu, W.; Zhang, S.; Chen, J. Oridonin inhibits the migration and epithelial-to-mesenchymal transition of small cell lung cancer cells by suppressing FAK-ERK1/2 signalling pathway. J. Cell. Mol. Med. 2020, 24, 4480–4493. [Google Scholar] [CrossRef] [PubMed]
  17. Ren, D.-L.; Ghoorun, R.A.; Wu, X.-H.; Chen, H.-L.; Zhou, Q.; Wu, X.-B. Oridonin induces apoptosis in HGC-27 cells by activating the JNK signaling pathway. Oncol. Lett. 2020, 19, 255–260. [Google Scholar] [CrossRef]
  18. Lu, J.; Chen, X.; Qu, S.; Yao, B.; Xu, Y.; Wu, J.; Jin, Y.; Ma, C. Oridonin induces G2/M cell cycle arrest and apoptosis via the PI3K/Akt signaling pathway in hormone-independent prostate cancer cells. Oncol. Lett. 2017, 13, 2838–2846. [Google Scholar] [CrossRef] [Green Version]
  19. Ren, K.-K.; Wang, H.-Z.; Xie, L.-P.; Chen, D.-W.; Liu, X.; Sun, J.; Nie, Y.-C.; Zhang, R.-Q. The effects of oridonin on cell growth, cell cycle, cell migration and differentiation in melanoma cells. J. Ethnopharmacol. 2006, 103, 176–180. [Google Scholar] [CrossRef]
  20. Nagy, J.A.; Dvorak, H.F. Heterogeneity of the tumor vasculature: The need for new tumor blood vessel type-specific targets. Clin. Exp. Metastasis 2012, 29, 657–662. [Google Scholar] [CrossRef] [Green Version]
  21. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [Green Version]
  22. Nasir, A. Angiogenic signaling pathways and anti-angiogenic therapies in human cancer. In Predictive Biomarkers in Oncology; Springer: Cham, Switzerland, 2019; pp. 243–262. [Google Scholar]
  23. Bergers, G.; Benjamin, L.E. Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 2003, 3, 401–410. [Google Scholar] [CrossRef]
  24. Almog, N.; Ma, L.; Raychowdhury, R.; Schwager, C.; Erber, R.; Short, S.; Hlatky, L.; Vajkoczy, P.; Huber, P.E.; Folkman, J. Transcriptional switch of dormant tumors to fast-growing angiogenic phenotype. Cancer Res. 2009, 69, 836–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zuazo-Gaztelu, I.; Casanovas, O. Unraveling the role of angiogenesis in cancer ecosystems. Front. Oncol. 2018, 8, 248. [Google Scholar] [CrossRef] [PubMed]
  26. Isogai, S.; Lawson, N.D.; Torrealday, S.; Horiguchi, M.; Weinstein, B.M. Angiogenic network formation in the developing vertebrate trunk. Development 2003, 130, 5281–5290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zecchin, A.; Kalucka, J.; Dubois, C.; Carmeliet, P. How endothelial cells adapt their metabolism to form vessels in tumors. Front. Immunol. 2017, 8, 1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Shawber, C.J.; Kitajewski, J. Notch function in the vasculature: Insights from zebrafish, mouse and man. Bioessays 2004, 26, 225–234. [Google Scholar] [CrossRef] [PubMed]
  29. Qiao, L.; Liang, N.; Zhang, J.; Xie, J.; Liu, F.; Xu, D.; Yu, X.; Tian, Y. Advanced research on vasculogenic mimicry in cancer. J. Cell. Mol. Med. 2015, 19, 315–326. [Google Scholar] [CrossRef] [PubMed]
  30. Tewari, K.S.; Sill, M.W.; Long, H.J., III; Penson, R.T.; Huang, H.; Ramondetta, L.M.; Landrum, L.M.; Oaknin, A.; Reid, T.J.; Leitao, M.M. Improved survival with bevacizumab in advanced cervical cancer. N. Engl. J. Med. 2014, 370, 734–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Finke, J.; Ko, J.; Rini, B.; Rayman, P.; Ireland, J.; Cohen, P. MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy. Int. Immunopharmacol. 2011, 11, 856–861. [Google Scholar] [CrossRef] [Green Version]
  32. Shibuya, M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: A crucial target for anti-and pro-angiogenic therapies. Genes Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef]
  33. Sa‑Nguanraksa, D.; Chuangsuwanich, T.; Pongpruttipan, T. High vascular endothelial growth factor gene expression predicts poor outcome in patients with non-luminal A breast cancer. Mol. Clin. Oncol. 2015, 3, 1103–1108. [Google Scholar] [CrossRef] [Green Version]
  34. George, D.J.; Halabi, S.; Shepard, T.F.; Vogelzang, N.J.; Hayes, D.F.; Small, E.J.; Kantoff, P.W. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin. Cancer Res. 2001, 7, 1932–1936. [Google Scholar] [PubMed]
  35. Horikawa, N.; Abiko, K.; Matsumura, N.; Hamanishi, J.; Baba, T.; Yamaguchi, K.; Yoshioka, Y.; Koshiyama, M.; Konishi, I. Expression of vascular endothelial growth factor in ovarian cancer inhibits tumor immunity through the accumulation of myeloid-derived suppressor cells. Clin. Cancer Res. 2017, 23, 587–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zanotelli, M.R.; Reinhart-King, C.A. Mechanical forces in tumor angiogenesis. In Biomechanics in Oncology; Springer: New York, NY, USA, 2018; pp. 91–112. [Google Scholar]
  37. Dong, Y.; Zhang, T.; Li, J.; Deng, H.; Song, Y.; Zhai, D.; Peng, Y.; Lu, X.; Liu, M.; Zhao, Y.; et al. Oridonin inhibits tumor growth and metastasis through anti-angiogenesis by blocking the notch signaling. PLoS ONE 2014, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Jiang, J.-H.; Pi, J.; Cai, J.-Y. Oridonin exhibits anti-angiogenic activity in human umbilical vein endothelial cells by inhibiting VEGF-induced VEGFR-2 signaling pathway. Pathol. Res. Pract. 2020, 216, 153031. [Google Scholar] [CrossRef] [PubMed]
  39. Tian, L.; Xie, K.; Sheng, D.; Wan, X.; Zhu, G. Antiangiogenic effects of oridonin. BMC Complement. Altern. Med. 2017, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Li, C.; Wang, Q.; Shen, S.; Wei, X.; Li, G. Oridonin inhibits vegf-a-associated angiogenesis and epithelial-mesenchymal transition of breast cancer in vitro and in vivo. Oncol. Lett. 2018, 16, 2289–2298. [Google Scholar] [CrossRef] [Green Version]
  41. Xia, S.; Zhang, X.; Li, C.; Guan, H. Oridonin inhibits breast cancer growth and metastasis through blocking the Notch signaling. Saudi Pharm. J. 2017, 25, 638–643. [Google Scholar] [CrossRef]
  42. Karar, J.; Maity, A. PI3K/AKT/mTOR pathway in angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef] [Green Version]
  43. Pujade-Lauraine, E.; Hilpert, F.; Weber, B.; Reuss, A.; Poveda, A.; Kristensen, G.; Sorio, R.; Vergote, I.; Witteveen, P.; Bamias, A. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. Obstet. Gynecol. Surv. 2014, 69, 402–404. [Google Scholar] [CrossRef]
  44. Li, J.; Wu, Y.; Wang, D.; Zou, L.; Fu, C.; Zhang, J.; Leung, G.P.-H. Oridonin synergistically enhances the anti-tumor efficacy of doxorubicin against aggressive breast cancer via pro-apoptotic and anti-angiogenic effects. Pharmacol. Res. 2019, 146. [Google Scholar] [CrossRef]
  45. Steeg, P.S. Targeting metastasis. Nat. Rev. Cancer 2016, 16, 201. [Google Scholar] [CrossRef] [PubMed]
  46. Thiery, J.P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef] [PubMed]
  47. De Craene, B.; Berx, G. Regulatory networks defining EMT during cancer initiation and progression. Nat. Rev. Cancer 2013, 13, 97–110. [Google Scholar] [CrossRef] [PubMed]
  48. Wu, S.; Du, Y.; Beckford, J.; Alachkar, H. Upregulation of the EMT marker vimentin is associated with poor clinical outcome in acute myeloid leukemia. J. Transl. Med. 2018, 16, 1–9. [Google Scholar] [CrossRef] [Green Version]
  49. Mendez, M.G.; Kojima, S.; Goldman, R.D. Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J. 2010, 24, 1838–1851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Hsiao, C.-T.; Cheng, H.-W.; Huang, C.-M.; Li, H.-R.; Ou, M.-H.; Huang, J.-R.; Khoo, K.-H.; Yu, H.W.; Chen, Y.-Q.; Wang, Y.-K. Fibronectin in cell adhesion and migration via N-glycosylation. Oncotarget 2017, 8, 70653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Santamaria, P.G.; Moreno-Bueno, G.; Portillo, F.; Cano, A. EMT: Present and future in clinical oncology. Mol. Oncol. 2017, 11, 718–738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Gui, Z.; Luo, F.; Yang, Y.; Shen, C.; Li, S.; Xu, J. Oridonin inhibition and miR-200b-3p/ZEB1 axis in human pancreatic cancer. Int. J. Oncol. 2017, 50, 111–120. [Google Scholar] [CrossRef] [Green Version]
  53. Lou, S.; Xu, J.; Wang, B.; Li, S.; Ren, J.; Hu, Z.; Xu, B.; Luo, F. Downregulation of lncRNA AFAP1-AS1 by oridonin inhibits the epithelial-to-mesenchymal transition and proliferation of pancreatic cancer cells. Acta Biochim. Biophys. Sin. 2018, 51, 814–825. [Google Scholar] [CrossRef]
  54. Sun, Y.; Jiang, X.; Lu, Y.; Zhu, J.; Yu, L.; Ma, B.; Zhang, Q. Oridonin prevents epithelial-mesenchymal transition and TGF-β1-induced epithelial-mesenchymal transition by inhibiting TGF-β1/Smad2/3 in osteosarcoma. Chem. Biol. Interact. 2018, 296, 57–64. [Google Scholar] [CrossRef]
  55. Wieczorek, E.; Jablonska, E.; Wasowicz, W.; Reszka, E. Matrix metalloproteinases and genetic mouse models in cancer research: A mini-review. Tumor Biol. 2015, 36, 163–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Smith, B.N.; Bhowmick, N.A. Role of EMT in metastasis and therapy resistance. J. Clin. Med. 2016, 5, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ma, S.; Tan, W.; Du, B.; Liu, W.; Li, W.; Che, D.; Zhang, G. Oridonin effectively reverses cisplatin drug resistance in human ovarian cancer cells via induction of cell apoptosis and inhibition of matrix metalloproteinase expression. Mol. Med. Rep. 2016, 13, 3342–3348. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, J.; Yan, S.; Tao, Z.; Wang, C.; Huang, M.; Zhang, X.; Du, Y. Oridonin inhibits the proliferation, migration and invasion of human osteosarcoma cells via suppression of matrix metalloproteinase expression and STAT3 signalling pathway. J. BU ON 2019, 24, 1175–1180. [Google Scholar]
  59. Zhang, Y.; Wang, L.; Zi, Y.; Zhang, L.; Guo, Y.; Huang, Y. Oridonin effectively reverses the drug resistance of cisplatin involving induction of cell apoptosis and inhibition of MMP expression in human acute myeloid leukemia cells. Saudi J. Biol. Sci. 2017, 24, 678–686. [Google Scholar] [CrossRef] [PubMed]
  60. Xiao, X.; He, Z.; Cao, W.; Cai, F.; Zhang, L.; Huang, Q.; Fan, C.; Duan, C.; Wang, X.; Wang, J.; et al. Oridonin inhibits gefitinib-resistant lung cancer cells by suppressing EGFR/ERK/MMP-12 and CIP2A/Akt signaling pathways. Int. J. Oncol. 2016, 48, 2608–2618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Liu, Q.-Q.; Chen, K.; Ye, Q.; Jiang, X.-H.; Sun, Y.-W. Oridonin inhibits pancreatic cancer cell migration and epithelial-mesenchymal transition by suppressing Wnt/β-catenin signaling pathway. Cancer Cell Int. 2016, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Zhang, Y.; Zhang, X.; Ye, M.; Jing, P.; Xiong, J.; Han, Z.; Kong, J.; Li, M.; Lai, X.; Chang, N.; et al. FBW7 loss promotes epithelial-to-mesenchymal transition in non-small cell lung cancer through the stabilization of Snail protein. Cancer Lett. 2018, 419, 75–83. [Google Scholar] [CrossRef] [PubMed]
  63. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 1–19. [Google Scholar]
  64. Chiang, S.P.H.; Cabrera, R.M.; Segall, J.E. Tumor cell intravasation. Am. J. Physiol. Physiol. 2016, 311, C1–C14. [Google Scholar] [CrossRef] [Green Version]
  65. Fontebasso, Y.; Dubinett, S.M. Drug development for metastasis prevention. Crit. Rev. Oncog. 2015, 20, 449–473. [Google Scholar] [CrossRef] [PubMed]
  66. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 1–17. [Google Scholar] [CrossRef] [PubMed]
  67. Valastyan, S.; Weinberg, R.A. Tumor metastasis: Molecular insights and evolving paradigms. Cell 2011, 147, 275–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Vandenbroucke, R.E.; Libert, C. Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat. Rev. Drug Discov. 2014, 13, 904–927. [Google Scholar] [CrossRef]
  69. Teixeira, A.F.; Dijke, P.T.; Zhu, H.-J. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front. Cell Dev. Biol. 2020, 8, 605. [Google Scholar] [CrossRef]
  70. Bu, H.-Q.; Shen, F.; Cui, J. The inhibitory effect of oridonin on colon cancer was mediated by deactivation of TGF-β1/smads-pai-1 signaling pathway in vitro and vivo. OncoTargets Ther. 2019, 12, 7467–7476. [Google Scholar] [CrossRef] [Green Version]
  71. Xia, R.; Chen, S.-X.; Qin, Q.; Chen, Y.; Zhang, W.-W.; Zhu, R.R.; Deng, A.-M. Oridonin suppresses proliferation of human ovarian cancer cells via blockage of mTOR signaling. Asian Pac. J. Cancer Prev. 2016, 17, 667–671. [Google Scholar] [CrossRef] [Green Version]
  72. Bi, E.; Liu, D.; Li, Y.; Mao, X.; Wang, A.; Wang, J. Oridonin induces growth inhibition and apoptosis in human gastric carcinoma cells by enhancement of p53 expression and function. Braz. J. Med. Biol. Res. 2018, 51. [Google Scholar] [CrossRef] [Green Version]
  73. Xu, S.; Yang, S.; Sun, G.; Huang, W.; Zhang, Y. Transforming growth factor-beta polymorphisms and serum level in the development of osteosarcoma. DNA Cell Biol. 2014, 33, 802–806. [Google Scholar] [CrossRef]
  74. Ahel, J.; Hudorović, N.; Vičić-Hudorović, V.; Nikles, H. Tgf-Beta in the Natural History of Prostate Cancer. Acta Clin. Croat. 2019, 58, 128–138. [Google Scholar] [CrossRef] [Green Version]
  75. Yang, L. TGFβ and cancer metastasis: An inflammation link. Cancer Metastasis Rev. 2010, 29, 263–271. [Google Scholar] [CrossRef] [PubMed]
  76. Margadant, C.; Sonnenberg, A. Integrin–TGF-β crosstalk in fibrosis, cancer and wound healing. EMBO Rep. 2010, 11, 97–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Sarwar, M.D.; Xia, Y.-X.; Liang, Z.-M.; Tsang, S.W.; Zhang, H.-J. Mechanistic pathways and molecular targets of plant-derived anticancer ent-kaurane diterpenes. Biomolecules 2020, 10, 144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Li, C.-Y.; Wang, Q.; Shen, S.; Wei, X.-L.; Li, G.-X. Oridonin inhibits migration, invasion, adhesion and TGF-ß1-induced epithelial-mesenchymal transition of melanoma cells by inhibiting the activity of PI3K/AKT/GSK-3ß signaling pathway. Oncol. Lett. 2018, 15, 1362–1372. [Google Scholar] [CrossRef] [PubMed]
  79. Guo, J.; Chen, T.; Ma, Z.; Qiao, C.; Yuan, F.; Guo, X.; Liu, J.; Shen, Y.; Yu, L.; Xiang, A. Oridonin inhibits 4T1 tumor growth by suppressing Treg differentiation via TGF-β receptor. Int. Immunopharmacol. 2020, 88, 106831. [Google Scholar] [CrossRef] [PubMed]
  80. Park, H.S.; Jang, M.H.; Kim, E.J.; Kim, H.J.; Lee, H.J.; Kim, Y.J.; Kim, J.H.; Kang, E.; Kim, S.-W.; Kim, I.A. High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. Mod. Pathol. 2014, 27, 1212–1222. [Google Scholar] [CrossRef] [Green Version]
  81. Wang, K.; Li, D.; Sun, L. High levels of EGFR expression in tumor stroma are associated with aggressive clinical features in epithelial ovarian cancer. OncoTargets Ther. 2016, 9, 377. [Google Scholar] [CrossRef] [Green Version]
  82. Hsieh, C.-Y.; Tsai, P.-C.; Tseng, C.-H.; Chen, Y.; Chang, L.-S.; Lin, S.-R. Inhibition of EGF/EGFR activation with naphtho [1, 2-b] furan-4, 5-dione blocks migration and invasion of MDA-MB-231 cells. Toxicol. In Vitro 2013, 27, 1–10. [Google Scholar] [CrossRef]
  83. Guo, Y.; Pan, W.; Liu, S.; Shen, Z.; Xu, Y.; Hu, L. ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef] [Green Version]
  84. Jackson, J.L.; Young, M.R.I. Protein phosphatase-2A regulates protein tyrosine phosphatase activity in Lewis lung carcinoma tumor variants. Clin. Exp. Metastasis 2003, 20, 357–364. [Google Scholar] [CrossRef]
  85. Benefield, J.; Meisinger, J.; Petruzzelli, G.J.; Young, M.R. Endothelial cell response to human head and neck squamous cell carcinomas involves downregulation of protein phosphatases-1/2A, cytoskeletal depolymerization and increased motility. Invasion Metastasis 1997, 17, 210–220. [Google Scholar] [PubMed]
  86. Zhang, J.; Hochwald, S.N. The role of FAK in tumor metabolism and therapy. Pharmacol. Ther. 2014, 142, 154–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Schmidt, A.; Kunz, J.; Hall, M.N. TOR2 is required for organization of the actin cytoskeleton in yeast. Proc. Natl. Acad. Sci. USA 1996, 93, 13780–13785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Dada, S.; Demartines, N.; Dormond, O. mTORC2 regulates PGE2-mediated endothelial cell survival and migration. Biochem. Biophys. Res. Commun. 2008, 372, 875–879. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, Y.; Zhu, Z. Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway. Arch. Med. Sci. 2019, 15, 1017–1027. [Google Scholar] [CrossRef]
  90. Lu, L.; Barbi, J.; Pan, F. The regulation of immune tolerance by FOXP3. Nat. Rev. Immunol. 2017, 17, 703. [Google Scholar] [CrossRef]
  91. Zhang, C.; Xu, Y.; Hao, Q.; Wang, S.; Li, H.; Li, J.; Gao, Y.; Li, M.; Li, W.; Xue, X. FOXP 3 suppresses breast cancer metastasis through downregulation of CD 44. Int. J. Cancer 2015, 137, 1279–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yang, H.; Gao, Y.; Fan, X.; Liu, X.; Peng, L.; Ci, X. Oridonin sensitizes cisplatin-induced apoptosis via AMPK/Akt/mTOR-dependent autophagosome accumulation in A549 cells. Front. Oncol. 2019, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bu, H.; Liu, D.; Zhang, G.; Chen, L.; Song, Z. AMPK/mTOR/ULK1 Axis-Mediated Pathway Participates in Apoptosis and Autophagy Induction by Oridonin in Colon Cancer DLD-1 Cells. OncoTargets Ther. 2020, 13, 8533. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, Q.; Wan, Q.; Zhang, L.; Li, Y.; Zhang, P.; Li, D.; Feng, C.; Yi, F.; Zhang, L.; Ding, X. Analysis of LncRNA expression in cell differentiation. RNA Biol. 2018, 15, 413–422. [Google Scholar] [CrossRef]
  95. Gibb, E.A.; Brown, C.J.; Lam, W.L. The functional role of long non-coding RNA in human carcinomas. Mol. Cancer 2011, 10, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Nalbantoglu, S.; Abu-Asab, M.; Tan, M.; Zhang, X.; Cai, L.; Amri, H. Study of clinical survival and gene expression in a sample of pancreatic ductal adenocarcinoma by parsimony phylogenetic analysis. OMICS A J. Integr. Biol. 2016, 20, 442–447. [Google Scholar] [CrossRef] [PubMed]
  97. Muller, P.A.J.; Vousden, K.H.; Norman, J.C. p53 and its mutants in tumor cell migration and invasion. J. Cell Biol. 2011, 192, 209–218. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, S.-P.; Wang, W.-L.; Chang, Y.-L.; Wu, C.-T.; Chao, Y.-C.; Kao, S.-H.; Yuan, A.; Lin, C.-W.; Yang, S.-C.; Chan, W.-K. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat. Cell Biol. 2009, 11, 694–704. [Google Scholar] [CrossRef] [PubMed]
  99. Shi, D.; Gu, W. Dual roles of MDM2 in the regulation of p53: Ubiquitination dependent and ubiquitination independent mechanisms of MDM2 repression of p53 activity. Genes Cancer 2012, 3, 240–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Busuttil, R.A.; Zapparoli, G.V.; Haupt, S.; Fennell, C.; Wong, S.Q.; Pang, J.-M.B.; Takeno, E.A.; Mitchell, C.; Di Costanzo, N.; Fox, S. Role of p53 in the progression of gastric cancer. Oncotarget 2014, 5, 12016. [Google Scholar] [CrossRef] [Green Version]
  101. Fenoglio-Preiser, C.M.; Wang, J.; Stemmermann, G.N.; Noffsinger, A. TP53 and gastric carcinoma: A review. Hum. Mutat. 2003, 21, 258–270. [Google Scholar] [CrossRef] [PubMed]
  102. Zhu, H.-Q.; Zhang, C.; Guo, Z.-Y.; Yang, J.-M.; Guo, J.-H.; Chen, C.; Yao, Q.-H.; Liu, F.; Zhang, Q.-W.; Gao, F.-H. Oridonin induces Mdm2-p60 to promote p53-mediated apoptosis and cell cycle arrest in neuroblastoma. Cancer Med. 2019, 8, 5313–5326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Jiang, J.-H.; Pi, J.; Jin, H.; Cai, J.-Y. Oridonin-induced mitochondria-dependent apoptosis in esophageal cancer cells by inhibiting PI3K/AKT/mTOR and Ras/Raf pathways. J. Cell. Biochem. 2019, 120, 3736–3746. [Google Scholar] [CrossRef] [PubMed]
  104. Vaupel, P.; Mayer, A.; Höckel, M. Tumor hypoxia and malignant progression. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2004; Volume 381, pp. 335–354. ISBN 0076-6879. [Google Scholar]
  105. Jögi, A.; Ehinger, A.; Hartman, L.; Alkner, S. Expression of HIF-1α is related to a poor prognosis and tamoxifen resistance in contralateral breast cancer. PLoS ONE 2019, 14, e0226150. [Google Scholar] [CrossRef] [Green Version]
  106. Yang, M.-H.; Wu, K.-J. TWIST activation by hypoxia inducible factor-1 (HIF-1): Implications in metastasis and development. Cell Cycle 2008, 7, 2090–2096. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, W.; Shi, X.; Peng, Y.; Wu, M.; Zhang, P.; Xie, R.; Wu, Y.; Yan, Q.; Liu, S.; Wang, J. HIF-1α promotes epithelial-mesenchymal transition and metastasis through direct regulation of ZEB1 in colorectal cancer. PLoS ONE 2015, 10, e0129603. [Google Scholar] [CrossRef] [PubMed]
  108. Krock, B.L.; Skuli, N.; Simon, M.C. Hypoxia-induced angiogenesis: Good and evil. Genes Cancer 2011, 2, 1117–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Tsai, Y.-P.; Wu, K.-J. Hypoxia-regulated target genes implicated in tumor metastasis. J. Biomed. Sci. 2012, 19, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [Green Version]
  111. Guise, C.P.; Mowday, A.M.; Ashoorzadeh, A.; Yuan, R.; Lin, W.-H.; Wu, D.-H.; Smaill, J.B.; Patterson, A.V.; Ding, K. Bioreductive prodrugs as cancer therapeutics: Targeting tumor hypoxia. Chin. J. Cancer 2014, 33, 80. [Google Scholar] [CrossRef] [PubMed]
  112. Marcu, L.; Olver, I. Tirapazamine: From bench to clinical trials. Curr. Clin. Pharmacol. 2006, 1, 71–79. [Google Scholar] [CrossRef] [PubMed]
  113. Phillips, R.M.; Hendriks, H.R.; Peters, G.J.; on Behalf of the EORTC-Pharmacology; Molecular Mechanism Group. EO9 (Apaziquone): From the clinic to the laboratory and back again. Br. J. Pharmacol. 2013, 168, 11–18. [Google Scholar] [CrossRef] [Green Version]
  114. Chen, K.; Ye, J.; Qi, L.; Liao, Y.; Li, R.; Song, S.; Zhou, C.; Feng, R.; Zhai, W. Oridonin inhibits hypoxia-induced epithelial-mesenchymal transition and cell migration by the hypoxia-inducible factor-1α/matrix metallopeptidase-9 signal pathway in gallbladder cancer. Anti Cancer Drugs 2019, 30, 925–932. [Google Scholar] [CrossRef]
  115. Ding, Y.; Ding, C.; Ye, N.; Liu, Z.; Wold, E.A.; Chen, H.; Wild, C.; Shen, Q.; Zhou, J. Discovery and development of natural product oridonin-inspired anticancer agents. Eur. J. Med. Chem. 2016, 122, 102–117. [Google Scholar] [CrossRef] [Green Version]
  116. Zhou, J.; Yun, E.-J.; Chen, W.; Ding, Y.; Wu, K.; Wang, B.; Ding, C.; Hernandez, E.; Santoyo, J.; Pong, R.-C.; et al. Targeting 3-phosphoinositide-dependent protein kinase 1 associated with drug-resistant renal cell carcinoma using new oridonin analogs. Cell Death Dis. 2017, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Xu, J.; Yang, J.; Ran, Q.; Wang, L.; Liu, J.; Wang, Z.; Wu, X.; Hua, W.; Yuan, S.; Zhang, L.; et al. Synthesis and biological evaluation of novel 1-O- and 14-O-derivatives of oridonin as potential anticancer drug candidates. Bioorg. Med. Chem. Lett. 2008, 18, 4741–4744. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The chemical structure of oridonin.
Figure 1. The chemical structure of oridonin.
Molecules 26 00775 g001
Figure 2. The effects of oridonin on angiogenesis. Oridonin inhibits angiogenesis by blocking the activation of VEGF and its downstream signaling pathways such as Src, PI3K, ERK 1/2, and Notch in endothelial cells. Abbreviations: vascular endothelial growth factor (VEGF); vascular endothelial growth factor receptor (VEGFR); focal adhesion kinase (FAK); phosphatidylinositol 3-kinase (PI3K), mammalian target of rapamycin (mTOR); extracellular signal-regulated protein kinase (ERK).
Figure 2. The effects of oridonin on angiogenesis. Oridonin inhibits angiogenesis by blocking the activation of VEGF and its downstream signaling pathways such as Src, PI3K, ERK 1/2, and Notch in endothelial cells. Abbreviations: vascular endothelial growth factor (VEGF); vascular endothelial growth factor receptor (VEGFR); focal adhesion kinase (FAK); phosphatidylinositol 3-kinase (PI3K), mammalian target of rapamycin (mTOR); extracellular signal-regulated protein kinase (ERK).
Molecules 26 00775 g002
Figure 3. Overview of the metastasis process from primary tumor cells intravasate into the blood circulation followed by extravasation, supported by various growth factors, and settlement of the cancer cells at a distinct tissue site. Abbreviations: tumor necrosis factor 1α (TNF1-α); transforming growth factor- β (TGF-β),vascular endothelial growth factor (VEGF); epidermal growth factor (EGF); matrix metalloproteinase (MMP); platelet-derived growth factor (PDGF); insulin-like growth factor-1 (IGF-1).
Figure 3. Overview of the metastasis process from primary tumor cells intravasate into the blood circulation followed by extravasation, supported by various growth factors, and settlement of the cancer cells at a distinct tissue site. Abbreviations: tumor necrosis factor 1α (TNF1-α); transforming growth factor- β (TGF-β),vascular endothelial growth factor (VEGF); epidermal growth factor (EGF); matrix metalloproteinase (MMP); platelet-derived growth factor (PDGF); insulin-like growth factor-1 (IGF-1).
Molecules 26 00775 g003
Table 1. Effects of EMT markers modulated by oridonin.
Table 1. Effects of EMT markers modulated by oridonin.
EMT MarkerCancer TypeCell LineConcentration and Treatment TimeEffectReferences
ZEB1PancreaticBxPC-387.8–95 µM (24 h)Downregulation of ZEB1 protein expression[52,53]
PANC-155.8 μM–95 µM (24 h)
E-cadherinPancreaticBxPC-395 µM (24 h)Enhances of E-cadherin protein expression[53]
PANC-195 µM (24 h)
SW199015 µM (24 h)Enhances of E-cadherin protein expression[61]
BreastMDA-MB-2314–16 µM (24 h)Upregulation of E-cadherin mRNA and protein expression[40]
4T1
BoneMG-630.8–2 µM (24 h)Upregulation of E-cadherin mRNA and protein expression[54]
143B
LungH16885–10 µM (24 h)Upregulation of E-cadherin mRNA and protein expression[16]
N-cadherinPancreaticBxPC-387.8–95 µM (24 h)Downregulation of N-cadherin protein expression[52,53]
PANC-155.8 μM–95 µM (24 h)
BreastMDA-MB-2314–16 µM (24 h)Downregulation of N-cadherin mRNA and protein expression[40]
4T1
BoneMG-630.8–2 µM (24 h)Downregulation N-cadherin mRNA and protein expression[54]
143B
FibronectinPancreaticBxPC-387.8 µM (24 h)Downregulation of fibronectin protein expression[52]
PANC-155.8 μM (24 h)
VimentinBreastMDA-MB-2314–16 µM (24 h)Downregulation of vimentin mRNA and protein expression[40]
4T1
PancreaticSW199015 µM (24 h)Downregulation of vimentin mRNA levels[61]
BoneMG-630.8–2 µM (24 h)Downregulation of vimentin mRNA and protein expression[54]
143B
LungH16885–10 µM (24 h)Downregulation of vimentin mRNA and protein expression[16]
SnailBreastMDA-MB-2314–16 µM (24 h)Downregulation of snail mRNA and protein expression[40]
4T1
PancreaticSW199015 µM (24 h)Decreases snail mRNA levels[61]
BxPC-395 µM (24 h)Downregulation of snail protein expression[53]
PANC-1
BoneMG-630.8–2 µM (24 h)Downregulation of snail mRNA and protein expression[54]
143B
LungH16885–10 µM (24 h)Downregulation of snail mRNA and protein expression[16]
SkinA37520 µM (12 h)Downregulation of snail protein expression[62]
MDA-MB-435S
SlugPancreaticSW199015 µM (24 h)Decreases slug mRNA levels[61]
BxPC-395 µM (24 h)Downregulation of slug protein expression[53]
PANC-1
BoneMG-630.8–2 µM (24 h)Downregulation of slug mRNA and protein expression[54]
143B
LungH16885–10 µM (24 h)Downregulation of slug mRNA and protein expression[16]
MMP-2BreastMDA-MB-2311–5 µM (24 h)Downregulation of MMP-2 protein expression[13]
AMLMV4-11/DDP10–80 µM (48 h)Downregulation of MMP-2 protein expression[59]
BoneU2OS15–60 µM (48 h)Downregulation of MMP-2 protein expression[58]
OvarianA2780/DDP10–80 µM (48 h)Downregulation of MMP-2 protein expression[57]
MMP-3BoneU2OS15–60 µM (48 h)Downregulation of MMP-3 protein expression[58]
MMP-9BreastMDA-MB-2311–5 µM (24 h)Downregulation of MMP-9 protein expression[13]
AMLMV4-11/DDP10–80 µM (48 h)Downregulation of MMP-9 protein expression[59]
BoneU2OS15–60 µM (48 h)Downregulation of MMP-9 protein expression[58]
OvarianA2780/DDP10–80 µM (48 h)Downregulation of MMP-9 protein expression[57]
MMP-12LungH197510–20 µM (24 h)Downregulation of MMP-12 protein expression[60]
Abbreviations: epithelial-to-mesenchymal transition (EMT); zinc finger E-box binding homeobox 1 (ZEB1), matrix metalloproteinases (MMP); acute myeloid leukemia (AML); messenger RNA (mRNA).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abdullah, N.A.; Md Hashim, N.F.; Ammar, A.; Muhamad Zakuan, N. An Insight into the Anti-Angiogenic and Anti-Metastatic Effects of Oridonin: Current Knowledge and Future Potential. Molecules 2021, 26, 775. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26040775

AMA Style

Abdullah NA, Md Hashim NF, Ammar A, Muhamad Zakuan N. An Insight into the Anti-Angiogenic and Anti-Metastatic Effects of Oridonin: Current Knowledge and Future Potential. Molecules. 2021; 26(4):775. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26040775

Chicago/Turabian Style

Abdullah, Nurul Akmaryanti, Nur Fariesha Md Hashim, Aula Ammar, and Noraina Muhamad Zakuan. 2021. "An Insight into the Anti-Angiogenic and Anti-Metastatic Effects of Oridonin: Current Knowledge and Future Potential" Molecules 26, no. 4: 775. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26040775

Article Metrics

Back to TopTop