Next Article in Journal
Exogenous Melatonin Mitigates Acid Rain Stress to Tomato Plants through Modulation of Leaf Ultrastructure, Photosynthesis and Antioxidant Potential
Previous Article in Journal
Melatonin Alleviates High Temperature-Induced Pollen Abortion in Solanum lycopersicum
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Cancer Activities of Diterpenoids Derived from Euphorbia fischeriana Steud

1
Graduate School of Heilongjiang University of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China
2
Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
3
College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
*
Author to whom correspondence should be addressed.
Molecules 2018, 23(2), 387; https://doi.org/10.3390/molecules23020387
Submission received: 19 December 2017 / Revised: 8 February 2018 / Accepted: 9 February 2018 / Published: 11 February 2018

Abstract

:
Euphorbia fischeriana Steud is an essential oriental folk medicine used for healing cancer, edema and tuberculosis. Recently, its anticancer activitity has attracted more attention. A volume of research has indicated that diterpenoids are the major anticancer active constituents from this medicinal herb. In this review, we aimed to provide a summary of the promising anticancer diterpenoids from this plant; many diterpenoids mentioned in this article are newly discovered diterpenoids. According to the carbon skeleton and substituents, they can be classified into eight subtypes: ent-abietane, daphnane, tigliane, ingenane, ent-atisane, ent-rosane, ent-kaurane, and lathyrane. Futhermore, their key anticancer mechanisms and protein targets of these compounds will be discussed. These natural diterpenoids could provide a reservoir for drug discovery.

1. Introduction

Cancer is a dreaded disease that ranks as the second-leading cause of death worldwide [1]. Systematic research proves that abnormal cell proliferation and metastasis are the two main characteristics of cancer development. Treatments for cancer include surgery, radiotherapy, and chemotherapy [2,3]. However, complete inhibition of tumor recurrence and metastasis is difficult to achieve using these methods [2,3]. Furthermore, the resistance of tumor cells to chemotherapeutic drugs is a significant problem, and urges the discovery of novel adjuvant therapies [1,4]. Traditional Chinese Medicine contains a lot of anticancer active substances and can provide considerable drug leads and candidates [5,6]. For this reason, researchers are always looking for inspiration from natural products.
Euphorbia fischeriana Steud, a perennial herbaceous plant of the family Euphorbiaceae, is mainly distributed in northern China [7,8]. Modern medical reseach has shown that the extracts and pure compounds of E. fischeriana exhibit a variety of pharmacological properties, incluing antitumor, antimicrobial, antiviral, immune enhancing, sedative and analgesic activities [7]. Among them, the research related to anticancer activity has attracted additional attention recently. Extracts of E. fischeriana have been proven to be effective against several types of cancer, including malignant melanoma, lewis lung carcinoma and ascitic hepatoma, in mice [7,9,10].
Chemical investigations of E. fischeriana have revealed the presence of diterpenoids, triterpenes, steroids and aromatic tannins [7]. Diterpenoids are the major components of E. fischeriana. They present variable skeletons with high oxidative functional moieties or acylated substituents [11,12,13]. These chemical and structural characteristics have attracted scientists interested in drug discovery research. Therefore, in recent years, study on the biological activities of diterpenoids from E. fischeriana has become a research focus. The body of research has noted that many of isolated diterpenoid compounds from this medicinal herb have cytotoxicities against a range of cancer cell types, and great strides have been taken in unraveling the mechanisms behind these effects. Diterpenoids are believed to be the major anticancer constituents of E. fischeriana.
In this paper, we aimed to provide a summary of the promising anticancer diterpenoids from this plant; many diterpenoids mentioned in this article are newly discovered diterpenoids. Futhermore, their key anticancer mechanisms and the protein targets of these compounds will be reported. These natural diterpenoids could provide a reservoir for drug discovery.

2. Chemical Structure of Diterpenoids

E. fischeriana produces a diversity of diterpenoids; researchers have found that approximately 24 diterpenoids have anticancer activities in E. fischeriana. Concerning the carbon skeleton and substituents at specific positions, these diterpenoids have been classified into eight subtypes; namely, ent-abietane (1, 2, 3, 4, 15, 16, 17), tigliane (5, 7, 8), daphnane (6), ingenane (9, 10, 11, 18), ent-atisane (12, 13, 19, 20) ent-rosane (21, 22, 23), ent-kaurane (14), and lathyrane (24).
Figure 1 and Figure 2 show the chemical structures of the diterpenoids derived from E. fischeriana with anticancer activities.

3. Anticancer Activities of Diterpenoids

In in vitro study, 14 diterpenoids (114) isolated from E. fischeriana have been found to inhibit the proliferation of several cancer cells with promising IC50 values. Their names, subtypes, cell toxicities and corresponding references are compiled in Table 1. 13 diterpenoids (1, 10, 11, 1524) showed inhibitory activities on the formation of mammospheres in human breast cancer MCF-7 cells [14,15]. This research result indicates the potential of these bioactive diterpenoids for further investigation of the action targeting cancer stem cells [14]. Their names, subtypes, and corresponding references are compiled in Table 2.
Several in vivo research studies have provided evidence supporting the anti-cancer activities of compound 1 and compound 8. Here, we summarize detailed information about these studies in Table 3. The in vivo antitumor effects of other compounds need to be tested in future experiments.

4. The Anti-Cancer Mechanism of Diterpenoids

4.1. Induction of Apoptosis

Apoptosis is an ordered and orchestrated cellular process that occurs in physiological and pathological conditions [33,34,35]. The process involves ordered morphological and biochemical events, including nuclear chromatin condensation, cell shrinkage, membrane blebbing, chromosomal DNA breakage and modulation of precise signaling circuitry [36,37]. Evading apoptosis and resisting cell death is one of the ten hallmarks of cancer [38]. Therefore, apoptosis is becoming a focus for oncology research. A hopeful field of anticancer strategies is applying medicine to start the tumor cell apotosis process [39,40].
Compound 1, an ent-abietane type diterpenoid extracted from plants of the E. fischeriana, has been reported to exhibit promising anticancer activity by activating apoptosis in solid and liquid tumors, including human Leukemic, breast cancer and mouse melanoma [17,18,21,22,23]. At the molecular level, Jolkinolide B was found to inhibit JAK2/STAT3 pathway in human Leukemic HL-60 and THP-1 cells [21]. Compound 1 treatment led to downregulation of JAK2/STAT3 and bcl-2 and upregulation of Bax and cytosolic cytochrome c, thus triggering caspase-3, -8 and -9 activation-mediated apoptotic induction. On the other hand, compound 1 can interfere with PI3K/Akt pathways, leading to cancer cell apoptosis in MDA-MB-231 cells and Human Leukemic U937 cells [18,19]. In addition, a novel mechanistic finding showed that Jolkinolide B induced apoptosis in mouse melanoma B16F10 cells by altering glycolysis [23]. In the course of study, compound 1 was found to downregulate the mRNA expression of glucose transporter genes (Glut1, Glut3 and Glut4) and glycolysis-related kinase genes (Hk2 and Ldha), increase ROS level, and decrease the potential of the mitochondrial membrane, subsequently inducing tumor cells apoptosis in B16F10 cells [23]. Recent research has demonstrated that aerobic glycolysis is the main metabolic way by which most tumor cells produce ATP for growth and proliferation [23,41,42]. Therefore, inhibition of the glycolytic pathway may be a promising approach to inhibit cancer cell proliferation and induce cell apoptosis in tumor cells [23,43]. Compound 3 has also been shown to induce apoptosis in human cancer cells. The anticancer mechanism operates through inactivation of the JAK family kinases—JAK1, JAK2, and TYK2—by covalent cross-linking of the JAKs and blocking JAK/STAT3 signaling [25]. It is a promising anticancer drug candidate as a potent STAT3 signaling inhibitor [25]. Compound 4 has been reported as a novel type of NF-kB pathway inhibitor. It can keep IKK in its phosphorylated form irreversibly, and this effect leads to compound 4 effectively inhibiting tumor necrosis factor-A–induced NF-KB activation and inducing apoptosis of tumor cells [26]. It is another novel type of anticancer drug candidate [26]. All three diterpenoids belong to the abietane type. Compound 8, a tigliane type diterpenoid extracted from this plant, has been found to induce apoptosis in BGC823 cells via caspase-3/caspase-9-dependent pathway [29]. In short, these diterpenoids modulate several signaling pathways, which results in apoptosis of tumor cells. The various mechanisms of Compounds 1, 3, 4, 8 involved in inducing apoptosis are summarized in Table 4.

4.2. Cell Cycle Arrest

Cell cycle deregulation is one of the important features of tumor cells. The abnormal expression and activity of cyclins, cyclin-dependent kinases and tumor suppressor proteins may directly affect cell cycle progression and tumorigenesis [44,45,46]. Some of the diterpenoids possess the ability to induce cell cycle arrest (Table 5). For instance, compound 1 can block cell cycles at G1 in human myeloid Leukemic cell K562 [20]. Likewise, compound 1 can cause cell cycle arrest of prostate cancer in the G1 phase [24]. Compound 8 can restrain cell cycle arrest at the G2-M checkpoint in gastric cancer through downregulation of cdc2/cyclin B, cyclin A and p-chk1 protein expression [29]. The effects of other diterpenoids from E. fischeriana on the blockage of cell cycles are an ongoing topic of research.

4.3. Inhibition of Metastasis

Tumors possess the ability to transfer throughout the body and grow, a process known as metastasis, which is the leading cause of death from cancer. It is difficult to prevent of metastasis at present, since this process involves multiple stages, such as passing through the extracellular matrix, interaction with host lymphoid cells, and adhesion to basement membranes to form metastases. Fortunately many plant-derived compounds have been discovered to effectively suppress metastasis of tumor cells [47,48,49]. Compound 1 was found to inhibit the attachment of MDA-MB-231 cells to fibronectin, with these effects being mediated by the integrin/FAK and ERK pathways [50]. Compound 8 has been demonstrated to inhibit VEGF-induced angiogenesis by targeting the VEGFR-2 signaling pathway [51]. Treatment with compound 8 caused decreased expression levels of TIMP-1, TIMP-2, VEGF, bFGF, MMP-2, VEGFR-2 and VEGFR-3 in HUVEC [51]. These proteins are closely associated with the metastasis of cancer. Additionally, in MCF-7 cells, compound 8 can diminish the expression of VEGF and HIF-1α by blocking the PI3K/Akt/mTOR signaling pathway [28]. Animal studies have also been conducted to explore the potential in vivo therapeutic efficacy of compound 8. In a MCF-7 xenografted mouse model, it was able to significantly suppress tumor growth and angiogenesis by inhibiting the VEGFR-2 signaling pathway [51].

5. Conclusions and Future Perspectives

In this review, we aimed to highlight 24 cytotoxic diterpenoids from E. fischeriana, which have been the subject of relatively little research, and yet have been shown to be effective agianst numerous cancer types. Emerging anticancer active diterpenoids can be divided into 8 types—ent-abietane, daphnane, tigliane, ingenane, ent-atisane, ent-rosane, ent-kaurane, and lathyrane—according to their carbon skeleton and substituents. The antineoplastic mechanisms of these diterpenoids generally include modulation of apoptosis, arresting the cell cycle at various checkpoints, and inhibiting tumor cell metastasis by interfering with multiple signaling pathways. These chemical, structural and molecular approaches represent the basis for more advanced reseach on these anticancer active diterpenoids.
The chemical structure characteristics may affect the anticancer activities of a compound, such as the kind and position of substituents and the linker-chain length [52,53,54]. Hence, structural modifications on the eight subtypes of cytotoxic diterpenoids will become a research topic. This article can serve as a reference for researchers studying their variable relationships between skeleton structure and anticancer activities in order to design novel, highly effective, low-toxicity diterpenoids.
Cancer stem cells (CSCs) are characterized by self-renewal, marked proliferation, and multilineage differentiation [54,55,56,57]. They have been proved to be the vital factor of malignant tumor recurrence and metastasis [54,55,56,57]. The theory of CSCs offers a new target and orientation for tumor therapy. The stem-targeted efficacy and mechanism of Compounds 1, 10, 11, 1524 will be verified in future study.
Recent findings: Nitric Oxide plays an important role in the occurrence and development of tumors [58,59]. In conditions where NO is at lower concentrations (<500 nM), it aids in angiogenesis [58,60]. On the contrary, higher levels of NO (<500 nM) tend to be cytotoxic to cancer cells [58,60]. Therefore, NO has become a new target in tumor treatment. Compounds 1, 3, 23, 24 exhibited promising inhibitory effects on NO production in LPS-induced RAW 264.7 macrophages [61,62]. However, the influence of these diterpenoids on NO production in various cancer cells is still unknown. Investigating the effect of diterpenoids on NO production in cancer cells will help to elucidate the anticancer mechanisms of these compounds.
At present, plant-derived diterpenoids against cancer find it difficult to avoid weak selectivity and toxicity [63,64,65]. A future challenge would be to explore new diterpenoids with high selectivity on the basis of chemical structure diversity. For this reason, these diterpenoids are being continuously tested on normal cells and tissues to evaluate their specificity.
Many diterpenoids mentioned in this article are new diterpenoids isolated from E. fischeriana. Therefore, various problems need to be solved. Their cytotoxic activities against a variety of cancer cell lines and their potential molecular mechanisms need to be studied further. More research is essential to exploring how these prodigious molecules interact with the cellular components through molecular chemistry and molecular docking analysis. These would lay the foundation for designing new anticancer drugs with high efficiency and low toxicity.

Acknowledgments

This work was supported by a grant from the National Natural Science Foundation of China (No. 81573660) and the Intramural Research Program of the Qiqihar Medical University (No. QY2016Z-01).

Author Contributions

Baiyu Jian conceived and wrote the manuscript; Hao Zhang and Cuicui Han collected the literature; Jicheng Liu revised the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Habli, Z.; Toumieh, G.; Fatfat, M.; Rahal, O.N.; Gali-Muhtasib, H. Emerging Cytotoxic Alkaloids in the Battle against Cancer: Overview of Molecular Mechanisms. Molecules 2017, 22, 250. [Google Scholar] [CrossRef] [PubMed]
  2. Zhao, Y.; Yang, A.; Tu, P.; Hu, Z. Anti-Tumor Effects of the American Cockroach, Periplaneta americana. Chin. Med. 2017, 12, 26. [Google Scholar] [CrossRef] [PubMed]
  3. He, G.; Liu, J.; Wang, J.; Xie, S. The Anti-Tumor Mechanism Research Status of Traditional Chinese Medicine. J. Sichuan Tradit. Chin. Med. 2008, 26, 47–49. [Google Scholar]
  4. Morin, P.J. Drug Resistance and the Microenvironment: Nature and Nurture. Drug Resist. Updates 2003, 6, 169–172. [Google Scholar] [CrossRef]
  5. Carter, G.T. Natural Products and Pharma 2011: Strategic Changes Spur New Opportunities. Nat. Prod. Rep. 2011, 28, 1783–1789. [Google Scholar] [CrossRef] [PubMed]
  6. Pezzuto, J.M. Plant-Derived Anticancer Agents. Biochem. Pharmacol. 1997, 53, 121–133. [Google Scholar] [CrossRef]
  7. Sun, Y.X.; Liu, J.C. Chemical Constituents and Biological Activities of Euphorbia fischeriana Steud. Chem. Biodivers. 2011, 8, 1205–1214. [Google Scholar] [CrossRef] [PubMed]
  8. Barrero, R.A.; Chapman, B.; Yang, Y.; Moolhuijzen, P.; Keeble-Gagnere, G.; Zhang, N.; Tang, Q.; Bellgard, M.I.; Qiu, D. De Novo Assembly of Euphorbia fischeriana Root Transcriptome Identifies Prostratin Pathway Related Genes. BMC Genom. 2011, 12, 600. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, L.; Duan, H.; Wang, Y.; Liu, K.; Jiang, P.; Qu, Z.; Yagasaki, K.; Zhang, G. Inhibitory Effects of Lang-Du Extract on the in Vitro and in Vivo Growth of Melanoma Cells and Its Molecular Mechanisms of Action. Cytotechnology 2010, 62, 357–366. [Google Scholar] [CrossRef] [PubMed]
  10. Dong, M.H.; Zhang, Q.; Wang, Y.Y.; Zhou, B.S.; Sun, Y.F.; Fu, Q. Euphorbia fischeriana Steud Inhibits Malignant Melanoma via Modulation of the Phosphoinositide-3-Kinase/Akt Signaling Pathway. Exp. Ther. Med. 2016, 11, 1475–1480. [Google Scholar] [CrossRef] [PubMed]
  11. Shi, Q.W.; Su, X.H.; Kiyota, H. Chemical and Pharmacological Research of the Plants in Genus Euphorbia. Chem. Rev. 2008, 108, 4295–4327. [Google Scholar] [CrossRef] [PubMed]
  12. Vasas, A.; Hohmann, J. Euphorbia Diterpenes: Isolation, Structure, Biological Activity, and Synthesis (2008–2012). Chem. Rev. 2014, 114, 8579–8612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wang, H.B.; Wang, X.Y.; Liu, L.P.; Qin, G.W.; Kang, T.G. Tigliane Diterpenoids from the Euphorbiaceae and Thymelaeaceae Families. Chem. Rev. 2015, 115, 2975–3011. [Google Scholar] [CrossRef] [PubMed]
  14. Kuang, X.; Li, W.; Kanno, Y.; Yamashita, N.; Kikkawa, S.; Azumaya, I.; Nemoto, K.; Asada, Y.; Koike, K. Euphorins A-H: Bioactive Diterpenoids from Euphorbia fischeriana. J. Nat. Med. 2016, 70, 412–422. [Google Scholar] [CrossRef] [PubMed]
  15. Kuang, X.; Li, W.; Kanno, Y.; Yamashita, N.; Nemoto, K.; Asada, Y.; Koike, K. Ent-Atisane Diterpenoids from Euphorbia fischeriana Inhibit Mammosphere Formation in MCF-7 Cells. J. Nat. Med. 2016, 70, 120–126. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, M.; Li, J.; Qing, W.; Wang, Q.H.; Yang, B.Y.; Kuang, H.X. Antitumor Activity of Abietane Type Diterpene from Euphorbia fischeriana Steud. China Pharm. 2015, 8, 1261–1262. [Google Scholar]
  17. Xu, H.Y.; Chen, Z.W.; Hou, J.C.; Du, F.X.; Liu, J.C. Jolkinolide B Induces Apoptosis in MCF-7 Cells through Inhibition of the PI3k/Akt/Mtor Signaling Pathway. Oncol. Rep. 2013, 29, 212–218. [Google Scholar] [CrossRef] [PubMed]
  18. Lin, Y.; Cui, H.; Xu, H.; Yue, L.; Xu, H.; Jiang, L.; Liu, J. Jolkinolide B Induces Apoptosis in MDA-MB-231 Cells through Inhibition of the PI3k/Akt Signaling Pathway. Oncol. Rep. 2012, 27, 1976–1980. [Google Scholar] [PubMed]
  19. Wang, J.H.; Zhou, Y.J.; Bai, X.; He, P. Jolkinolide B from Euphorbia fischeriana Steud Induces Apoptosis in Human Leukemic U937 Cells through PI3k/Akt and Xiap Pathways. Mol. Cells 2011, 32, 451–457. [Google Scholar] [CrossRef] [PubMed]
  20. Luo, H.; Wang, A. Induction of Apoptosis in K562 Cells by Jolkinolide B. Can. J. Physiol. Pharmacol. 2006, 84, 959–965. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, J.H.; Zhang, K.; Niu, H.Y.; Shu, L.H.; Yue, D.M.; Li, D.; He, P. Jolkinolide B from Euphorbia fischeriana Steud Induces in Human Leukemic Cells Apoptosis Via Jak2/Stat3 Pathways. Int. J. Clin. Pharmacol. Ther. 2013, 51, 170–178. [Google Scholar] [CrossRef] [PubMed]
  22. Shen, L.; Zhang, S.Q.; Liu, L.; Sun, Y.; Wu, Y.X.; Xie, L.P.; Liu, J.C. Jolkinolide A and Jolkinolide B Inhibit Proliferation of A549 Cells and Activity of Human Umbilical Vein Endothelial Cells. Med. Sci. Monit. 2017, 23, 223–237. [Google Scholar] [CrossRef] [PubMed]
  23. Gao, C.; Yan, X.; Wang, B.; Yu, L.; Han, J.; Li, D.; Zheng, Q. Jolkinolide B Induces Apoptosis and Inhibits Tumor Growth in Mouse Melanoma B16f10 Cells by Altering Glycolysis. Sci. Rep. 2016, 6, 36114. [Google Scholar] [CrossRef] [PubMed]
  24. Liu, W.K.; Ho, J.C.; Qin, G.; Che, C.T. Jolkinolide B Induces Neuroendocrine Differentiation of Human Prostate Lncap Cancer Cell Line. Biochem. Pharmacol. 2002, 63, 951–957. [Google Scholar] [CrossRef]
  25. Wang, Y.; Ma, X.; Yan, S.; Shen, S.; Zhu, H.; Gu, Y.; Wang, H.; Qin, G.; Yu, Q. 17-Hydroxy-Jolkinolide B Inhibits Signal Transducers and Activators of Transcription 3 Signaling by Covalently Cross-Linking Janus Kinases and Induces Apoptosis of Human Cancer Cells. Cancer Res. 2009, 69, 7302–7310. [Google Scholar] [CrossRef] [PubMed]
  26. Yan, S.S.; Li, Y.; Wang, Y.; Shen, S.S.; Gu, Y.; Wang, H.B.; Qin, G.W.; Yu, Q. 17-Acetoxyjolkinolide B Irreversibly Inhibits Ikappab Kinase and Induces Apoptosis of Tumor Cells. Mol. Cancer Ther. 2008, 7, 1523–1532. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, M.; Wei, Q.; Wang, Q.H.; Xia, Y.G.; Yang, B.Y.; Kuang, H.X. Antitumor Activity of Tigliane Type Diterpene from Euphorbia fischeriana Steud. Acta Chin. Med. Pharmacol. 2013, 4, 11–14. [Google Scholar]
  28. Yang, Y.; Cong, H.; Han, C.; Yue, L.; Dong, H.; Liu, J. 12-Deoxyphorbol 13-Palmitate Inhibits the Expression of VEGF and HIF-1α in MCF-7 Cells by Blocking the PI3k/Akt/Mtor Signaling Pathway. Oncol. Rep. 2015, 34, 1755–1760. [Google Scholar] [CrossRef] [PubMed]
  29. Xu, H.Y.; Chen, Z.W.; Li, H.; Zhou, L.; Liu, F.; Lv, Y.Y.; Liu, J.C. 12-Deoxyphorbol 13-Palmitate Mediated Cell Growth Inhibition, G2-M Cell Cycle Arrest and Apoptosis in BGC823 Cells. Eur. J. Pharmacol. 2013, 700, 13–22. [Google Scholar] [CrossRef] [PubMed]
  30. Shi, Q.; Sun, Y.W.; Meng, D. Phytochemical and Cytotoxic Studies on the Roots of Euphorbia fischeriana. Bioorg. Med. Chem. Lett. 2017, 27, 266–270. [Google Scholar] [CrossRef] [PubMed]
  31. Wang, H.B.; Chen, W.; Zhang, Y.Y.; Wang, X.Y.; Liu, L.P.; Tong, L.J.; Chen, Y. Four New Diterpenoids from the Roots of Euphorbia fischeriana. Fitoterapia 2013, 91, 211–216. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, M.; Wang, Q.; Wei, Q.; Li, J.; Guo, C.; Yang, B.; Kuang, H. Two New ent-Atisanes from the Root of Euphorbia fischeriana Steud. Nat. Prod. Res. 2016, 30, 144–149. [Google Scholar] [CrossRef] [PubMed]
  33. Wong, R.S. Apoptosis in Cancer: From Pathogenesis to Treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef] [PubMed]
  34. Bie, B.; Sun, J.; Guo, Y.; Li, J.; Jiang, W.; Yang, J.; Huang, C.; Li, Z. Baicalein: A Review of Its Anti-Cancer Effects and Mechanisms in Hepatocellular Carcinoma. Biomed. Pharmacother. 2017, 93, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  35. Elmore, S. Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  36. Saraste, A.; Pulkki, K. Morphologic and Biochemical Hallmarks of Apoptosis. Cardiovasc. Res. 2000, 45, 528–537. [Google Scholar] [CrossRef]
  37. Tsuruo, T. Molecular Cancer Therapeutics: Recent Progress and Targets in Drug Resistance. Intern. Med. 2003, 42, 237–243. [Google Scholar] [CrossRef] [PubMed]
  38. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  39. Kasibhatla, S.; Tseng, B. Why Target Apoptosis in Cancer Treatment? Mol. Cancer Ther. 2003, 2, 573–580. [Google Scholar] [PubMed]
  40. Millimouno, F.M.; Dong, J.; Yang, L.; Li, J.; Li, X. Targeting Apoptosis Pathways in Cancer and Perspectives with Natural Compounds from Mother Nature. Cancer Prev. Res. 2014, 7, 1081–1107. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, T.A.; Zhang, X.D.; Guo, X.Y.; Xian, S.L.; Lu, Y.F. 3-Bromopyruvate and Sodium Citrate Target Glycolysis, Suppress Survivin, and Induce Mitochondrial-Mediated Apoptosis in Gastric Cancer Cells and Inhibit Gastric Orthotopic Transplantation Tumor Growth. Oncol. Rep. 2016, 35, 1287–1296. [Google Scholar] [CrossRef] [PubMed]
  42. Kwiatkowski, T.J.; Bosco, D.A., Jr.; Leclerc, A.L.; Tamrazian, E.; Vanderburg, C.R.; Russ, C.; Davis, A.; Gilchrist, J.; Kasarskis, E.J.; Munsat, T.; et al. Mutations in the Fus/Tls Gene on Chromosome 16 Cause Familial Amyotrophic Lateral Sclerosis. Science 2009, 323, 1205–1208. [Google Scholar] [CrossRef] [PubMed]
  43. Yoshida, G.J. Metabolic Reprogramming: The Emerging Concept and Associated Therapeutic Strategies. J. Exp. Clin. Cancer Res. 2015, 34, 111. [Google Scholar] [CrossRef] [PubMed]
  44. Besson, A.; Dowdy, S.F.; Roberts, J.M. Cdk Inhibitors: Cell Cycle Regulators and Beyond. Dev. Cell 2008, 14, 159–169. [Google Scholar] [CrossRef] [PubMed]
  45. Giacinti, C.; Giordano, A. Rb and Cell Cycle Progression. Oncogene 2006, 25, 5220–5227. [Google Scholar] [CrossRef] [PubMed]
  46. Foster, D.A.; Yellen, P.; Xu, L.; Saqcena, M. Regulation of G1 Cell Cycle Progression: Distinguishing the Restriction Point from a Nutrient-Sensing Cell Growth Checkpoint(S). Genes Cancer 2010, 1, 1124–1131. [Google Scholar] [CrossRef] [PubMed]
  47. Fidler, I.J.; Kripke, M.L. The Challenge of Targeting Metastasis. Cancer Metastasis Rev. 2015, 34, 635–641. [Google Scholar] [CrossRef] [PubMed]
  48. Pulley, J.M.; Jerome, R.N.; Ogletree, M.L.; Bernard, G.R.; Lavieri, R.R.; Zaleski, N.M.; Hong, C.C.; Shirey-Rice, J.K.; Arteaga, C.L.; Mayer, I.A.; et al. Motivation for Launching a Cancer Metastasis Inhibition (CMI) Program. Target. Oncol. 2017. [Google Scholar] [CrossRef] [PubMed]
  49. Ricciuti, B.; Foglietta, J.; Chiari, R.; Sahebkar, A.; Banach, M.; Bianconi, V.; Pirro, M. Emerging Enzymatic Targets Controlling Angiogenesis in Cancer: Preclinical Evidence and Potential Clinical Applications. Med. Oncol. 2017, 35, 4. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, C.; Cui, H.; Yang, H.; Du, X.; Yue, L.; Liu, J.; Lin, Y.U. Anti-Metastatic Effect of Jolkinolide B and the Mechanism of Activity in Breast Cancer Mda-Mb-231 Cells. Oncol. Lett. 2015, 10, 1117–1122. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, H.Y.; Pan, Y.M.; Chen, Z.W.; Lin, Y.; Wang, L.H.; Chen, Y.H.; Jie, T.T.; Lu, Y.Y.; Liu, J.C. 12-Deoxyphorbol 13-Palmitate Inhibit Vegf-Induced Angiogenesis Via Suppression of Vegfr-2-Signaling Pathway. J. Ethnopharmacol. 2013, 146, 724–733. [Google Scholar] [CrossRef] [PubMed]
  52. Ren, M.Y.; Yu, Q.T.; Shi, C.Y.; Luo, J.B. Anticancer Activities of C18-, C19-, C20-, and Bis-Diterpenoid Alkaloids Derived from Genus Aconitum. Molecules 2017, 22, 267. [Google Scholar] [CrossRef] [PubMed]
  53. Akhtar, J.; Khan, A.A.; Ali, Z.; Haider, R.; Yar, M.S. Structure-Activity Relationship (Sar) Study and Design Strategies of Nitrogen-Containing Heterocyclic Moieties for Their Anticancer Activities. Eur. J. Med. Chem. 2017, 125, 143–189. [Google Scholar] [CrossRef] [PubMed]
  54. Akhtar, M.J.; Yar, M.S.; Khan, A.A.; Ali, Z.; Haider, M.R. Recent Advances in the Synthesis and Anticancer Activity of Some Molecules Other Than Nitrogen Containing Heterocyclic Moeities. Mini Rev. Med. Chem. 2017, 17, 1602–1632. [Google Scholar] [CrossRef] [PubMed]
  55. Clevers, H. The Cancer Stem Cell: Premises, Promises and Challenges. Nat. Med. 2011, 17, 313–319. [Google Scholar] [CrossRef] [PubMed]
  56. Greaves, M. Cancer Stem Cells Renew Their Impact. Nat. Med. 2011, 17, 1046–1048. [Google Scholar] [CrossRef] [PubMed]
  57. Pattabiraman, D.R.; Weinberg, R.A. Tackling the Cancer Stem Cells—What Challenges Do They Pose? Nat. Rev. Drug Discov. 2014, 13, 497–512. [Google Scholar] [CrossRef] [PubMed]
  58. Vahora, H.; Khan, M.A.; Alalami, U.; Hussain, A. The Potential Role of Nitric Oxide in Halting Cancer Progression through Chemoprevention. J. Cancer Prev. 2016, 21, 1–12. [Google Scholar] [CrossRef] [PubMed]
  59. Reynolds, M.M.; Witzeling, S.D.; Damodaran, V.B.; Medeiros, T.N.; Knodle, R.D.; Edwards, M.A.; Lookian, P.P.; Brown, M.A. Applications for Nitric Oxide in Halting Proliferation of Tumor Cells. Biochem. Biophys. Res. Commun. 2013, 431, 647–651. [Google Scholar] [CrossRef] [PubMed]
  60. Lee, S.Y.; Rim, Y.; McPherson, D.D.; Huang, S.L.; Kim, H. A Novel Liposomal Nanomedicine for Nitric Oxide Delivery and Breast Cancer Treatment. Biomed. Mater. Eng. 2014, 24, 61–67. [Google Scholar] [PubMed]
  61. Lee, J.W.; Lee, C.; Jin, Q.; Jang, H.; Lee, D.; Lee, H.J.; Shin, J.W.; Han, S.B.; Hong, J.T.; Kim, Y.; et al. Diterpenoids from the Roots of Euphorbia fischeriana with Inhibitory Effects on Nitric Oxide Production. J. Nat. Prod. 2016, 79, 126–131. [Google Scholar] [CrossRef] [PubMed]
  62. Uto, T.; Qin, G.W.; Morinaga, O.; Shoyama, Y. 17-Hydroxy-Jolkinolide B, a Diterpenoid from Euphorbia fischeriana, Inhibits Inflammatory Mediators but Activates Heme Oxygenase-1 Expression in Lipopolysaccharide-Stimulated Murine Macrophages. Int. Immunopharmacol. 2012, 12, 101–109. [Google Scholar] [CrossRef] [PubMed]
  63. Bonavida, B.; Ng, C.P.; Jazirehi, A.; Schiller, G.; Mizutani, Y. Selectivity of Trail-Mediated Apoptosis of Cancer Cells and Synergy with Drugs: The Trail to Non-Toxic Cancer Therapeutics (Review). Int. J. Oncol. 1999, 15, 793–802. [Google Scholar] [CrossRef] [PubMed]
  64. Chabner, B.A.; Roberts, T.G., Jr. Timeline: Chemotherapy and the War on Cancer. Nat. Rev. Cancer 2005, 5, 65–72. [Google Scholar] [CrossRef] [PubMed]
  65. Bishayee, A.; Sethi, G. Bioactive Natural Products in Cancer Prevention and Therapy: Progress and Promise. Semin. Cancer Biol. 2016, 40–41, 1–3. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are not available or not from the authors.
Figure 1. The chemical structures of ent-abietane-, tigliane- and daphnane-type diterpenoids derived from E. fischeriana.
Figure 1. The chemical structures of ent-abietane-, tigliane- and daphnane-type diterpenoids derived from E. fischeriana.
Molecules 23 00387 g001
Figure 2. The chemical structures of ingenane-, ent-atisane-, ent-rosane-, ent-kaurane- and lathyrane-type diterpenoids derived from E. fischeriana.
Figure 2. The chemical structures of ingenane-, ent-atisane-, ent-rosane-, ent-kaurane- and lathyrane-type diterpenoids derived from E. fischeriana.
Molecules 23 00387 g002
Table 1. Emerging cytotoxic diterpenoids in E. fischeriana in vitro.
Table 1. Emerging cytotoxic diterpenoids in E. fischeriana in vitro.
No.CompoundSubtypeType of CancerCell Lines ((IC50)Ref.
1jolkinolide Bent-abietaneliverHepG-2 (24.43 μM/48 h)[16]
breastMCF-7 (22.76 μM/48 h)[16,17]
breastMDA-MB-231[18]
gastricSGC-7901 (31.32 μM/48 h)[16]
gastricBGC-823 (32.45 μM/48 h)[16]
gastricMGC-803 (34.7 μM/48 h)[16]
cervicalHela (23.12 μM/48 h)[16]
human leukemicU937[19]
human leukemicK562 (12.1 μg/mL/24 h)[20]
human leukemicK562 (11.3 μg/mL/48 h)[20]
human leukemicK562 (10.7 μg/mL/72 h)[20]
human leukemicTHP-1[21]
human leukemicHL-60[21]
lungA549 (28.24 μM)[22]
melanomaB16F10[23]
prostateLNCaP (40 μM/48 h)[24]
prostateDU145 (145 μM/48 h)[24]
prostatePC3 (244 μM/48 h)[24]
2jolkinolide Aent-abietaneliverHepG-2 (80.12 μM/48 h)[16]
breastMCF-7 (56.34 μM/48 h)[16]
lungA549[22]
gastricSGC-7901 (>100 μM/48 h)[16]
gastricBGC-823 (>100 μM/48 h)[16]
gastricMGC-803 (>100 μM/48 h)[16]
cervicalHela (>100 μM/48 h)[16]
317-hydroxyjolkinolide Bent-abietaneliverHepG-2 (42.13 μM/48 h)[16]
breastMCF-7 (25.33 μM/48 h)[16]
gastricSGC-7901 (44.34 μM/48 h)[16]
gastricBGC-823 (48.12 μM/48 h)[16]
gastricMGC-803 (43.89 μM/48 h)[16]
cervicalHela (35.11 μM/48 h)[16]
lungH460[16]
ovarySkov3[25]
coloColo205[25]
breastMDA-MB-453[25]
breastMDA-MB-231[25]
breastMDA_MB-468[25]
cervixHela[25]
liverHepG2[25]
bloodJurkat[25]
bloodU937[25]
bloodTHP-1[25]
417-acetoxyjolkinolide Bent-abietanebloodU937 (0.74 μM)[26]
bloodJurkat (1.06 μM)[26]
colonColo205 (2.34 μM)[26]
gastricHGC (3.64 μM))[26]
breastMCF-7 (8.74 μM))[26]
5prostratintiglianeliverHepG-2 (11.77 μM/48 h)[27]
breastMCF-7 (17.4 μM/48 h)[27]
gastricSGC-7901 (25.4 μM)[27]
6langduin AdaphnaneliverHepG-2 (35 μM/48 h)[27]
breastMCF-7 (19.4 μM/48 h)[27]
gastricSGC-7901 (21.3μM/48 h)[27]
713-O-acetylphorboltiglianeliverHepG-2 (32.3 μM/48 h)[27]
breastMCF-7 (18.1 μM/48 h)[27]
gastricSGC-7901 (24.91 μM/48 h)[27]
812-deoxyphorbol 13-palmitatetiglianebreastMCF-7[28]
gastricBGC823[29]
liverHep-3B (12.01 μM)[30]
lungA549 (9.38 μM)[30]
9ingenol-6,7-epoxy-3-tetradecanoateingenanelungA549 (3.35 μg/mL/72 h)[31]
liverBEL7402 (13.05 μg/mL/72 h)[31]
colonHCT116 (14.62 μg/mL/72 h)[31]
breastMDA-MB-231 (14.42 μg/mL/72 h)[31]
10ingenol-3-myristinateingenanelungA549 (2.85 μg/mL/72 h)[31]
liverBEL7402 (15.72 μg/mL/72 h)[31]
colonHCT116 (16.05 μg/mL/72 h)[31]
breastMDA-MB-231 (18.91μg/mL/72 h)[31]
11ingenol 3-palmitateingenanelungA549 (2.88 μg/mL/72 h)[31]
liverBEL7402 (25.87 μg/mL/72 h)[31]
colonHCT116 (14.38 μg/mL/72 h)[31]
breastMDA-MB-231 (22 μg/mL/72 h)[31]
12ent-1β,3β,16β, 17-tetrahydroxyatisaneent-atisanebreastMCF-7 (23.21 μM)[32]
13ent-1β,3α,16β, 17-tetrahydroxyatisaneent-atisanebreastMCF-7 (15.42 μM)[32]
14ent-kaurane-3-oxo-16β, 17-acetonideent-kauraneliverHep-3B (8.15 μM)[30]
Table 2. Diterpenoids from E. fischeriana inhibiting mammosphere formation in MCF-7 cells.
Table 2. Diterpenoids from E. fischeriana inhibiting mammosphere formation in MCF-7 cells.
No.Bioactive IngredientSubtypeRef.
1jolkinolide bent-abietane[14,15]
10ingenol-3-myristinateingenane
11ingenol-3-palmitateingenane
15euphorin Eent-abietane
16euphorin Hent-abietane
17yuexiandajisu Eent-abietane
18ingenol-20-myristinateingenane
19ent-3β-hydroxyatis-16-ene-2,14-dioneent-atisane
2019-O-β-d-glucopyranosyl-ent-atis-16-ene-3,14-dioneent-atisane
21euphorin Cent-rosane
22ebractenoid Cent-rosane
23ebractenoid Fent-rosane
24jolkinol Alathyrane
Table 3. Summary of the anticancer activities of diterpenoids in vivo.
Table 3. Summary of the anticancer activities of diterpenoids in vivo.
Animal ModelsDrug DoseConclusionsRef.
MCF-7 cells xenograft in nude micefour groups: the negative control group, the jolkinolide B group (40 mg/kg), the 5-Fu group (5 mg/kg), and the jolkinolide B+5-Fu group for 28 daystumor volume and weight in the 5-Fu, the 5-Fu + jolkinolide B and the jolkinolide B group were greatly reduced, while tumors in the control group reached 1207 mm. However, no significant difference was observed between the JB and the JB+5-Fu group[17]
B16F10 cells xenograft in C57BL/6 mice10, 20 and 40 mg/kg of jolkinolide B by intragastric administration for 7 daysThe tumor growth inhibition rates were 17.3%, 34.6% and 54.4% in JB-treated groups (10, 20 and 40 mg/Kg)[23]
BGC823 cells in Female Balb/c nude mice12-deoxyphorbol 13-palmitate (40 mg/kg) was administered intraperitoneally every three days for two monthstumor growth was significantly suppressed in the 40 mg/mL group compared to the control group[29]
Table 4. Mechanisms of diterpenoids in inducing apoptosis.
Table 4. Mechanisms of diterpenoids in inducing apoptosis.
No.Bioactive IngredientType of CancerCell LinesMechanisms of ActionRef.
1jolkinolide BbreastMDA-MB-231suppression of the PI3K/Akt signaling pathway[18]
human leukemicU937suppression of PI3K/Akt and XIAP pathways.
cIAP1/2 ↓, Survivin ↓ XIAP ↓ expression
Smac ↑ expression
activation of caspase-3 and -9.
[19]
HL-60
THP-1
suppression of the JAK2/STAT3 signaling pathway
↓ JAK2/STAT3 and bcl-2 expression
↑ Bax and cytosolic cytochrome c
triggering of caspase-3, -8 and-9 activation
[21]
mouse melanomaB16F10inhibition of glycolysis
↓ mRNA expression of glucose transporter genes (Glut1, Glut3 and Glut4) and glycolysis-related kinase gene(Hk2 and Ldha)
↑ ROS level
decreased the potential of mitochondrial membrane
[23]
317-hydroxyjolkinolide BLiver
Breast
breast
HepG2
MDA-MB-231
MDA_MB-468
inhibit STAT3 activation by direct inhibition of JAK kinase activity through covalent crosslinking of the JAKs[25]
417-acetoxyjolkinolide BLiver
Cervical
lung
HepG2
Hela
A549
a inhibitor of IKK
inhibit tumor NF-KB activation
[26]
812-deoxyphorbol 13-palmitategastricBGC823activation of caspase-3 and -9.[29]
Table 5. Effects of diterpenoids on cell cycles.
Table 5. Effects of diterpenoids on cell cycles.
NoBioactive IngredientType of CancerCell LinesEffects of Diterpenoids on Cell CycleRef.
1jolkinolide Bhuman leukemicK562Cell cycle arrest at G1[20]
prostateLNCapCell cycle arrest at G1[24]
812-deoxyphorbol 13-palmitategastricBGC823cell cycle arrest at G2-M checkpoint
↓ cdc2/cyclin B, cyclin A and p-chk1 protein expression
[29]

Share and Cite

MDPI and ACS Style

Jian, B.; Zhang, H.; Han, C.; Liu, J. Anti-Cancer Activities of Diterpenoids Derived from Euphorbia fischeriana Steud. Molecules 2018, 23, 387. https://doi.org/10.3390/molecules23020387

AMA Style

Jian B, Zhang H, Han C, Liu J. Anti-Cancer Activities of Diterpenoids Derived from Euphorbia fischeriana Steud. Molecules. 2018; 23(2):387. https://doi.org/10.3390/molecules23020387

Chicago/Turabian Style

Jian, Baiyu, Hao Zhang, Cuicui Han, and Jicheng Liu. 2018. "Anti-Cancer Activities of Diterpenoids Derived from Euphorbia fischeriana Steud" Molecules 23, no. 2: 387. https://doi.org/10.3390/molecules23020387

Article Metrics

Back to TopTop