Next Article in Journal
Ginkgolic Acid C 17:1, Derived from Ginkgo biloba Leaves, Suppresses Constitutive and Inducible STAT3 Activation through Induction of PTEN and SHP-1 Tyrosine Phosphatase
Next Article in Special Issue
Ent-Abietanoids Isolated from Isodon serra
Previous Article in Journal
Porous Hydrogen-Bonded Organic Frameworks
Previous Article in Special Issue
Penduliflaworosin, a Diterpenoid from Croton crassifolius, Exerts Anti-Angiogenic Effect via VEGF Receptor-2 Signaling Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer Activities of C18-, C19-, C20-, and Bis-Diterpenoid Alkaloids Derived from Genus Aconitum

1
School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
2
Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, China
*
Author to whom correspondence should be addressed.
Molecules 2017, 22(2), 267; https://doi.org/10.3390/molecules22020267
Submission received: 27 December 2016 / Revised: 5 February 2017 / Accepted: 6 February 2017 / Published: 13 February 2017
(This article belongs to the Special Issue Diterpene and Its Significance in Natural Medicine)

Abstract

:
Cancer is one of the most common lethal diseases, and natural products have been extensively studied as anticancer agents considering their availability, low toxicity, and economic affordability. Plants belonging to the genus Aconitum have been widely used medically in many Asian countries since ancient times. These plants have been proven effective for treating several types of cancer, such as lung, stomach, and liver cancers. The main effective components of Aconitum plants are diterpenoid alkaloids—which are divided into C18-, C19-, C20-, and bis-diterpenoid alkaloids—are reportedly some of the most promising, naturally abundant compounds for treating cancer. This review focuses on the progress of diterpenoid alkaloids with different structures derived from Aconitum plants and some of their derivatives with potential anticancer activities. We hope that this work can serve as a reference for further developing Aconitum diterpenoid alkaloids as anticancer agents.

1. Introduction

Cancer is one of the most common lethal diseases, with approximately 14 million new cases of cancer diagnosed and 8 million cancer-related deaths in 2012. This disease affects all populations in all regions according to the World Health Organization [1]. The five most common incident sites of cancers are the lung, breast, colorectum, prostate, and stomach, constituting half of incident sites worldwide [1]. In recent years, natural products, including materials originating from plants, animals and their derivatives, have been extensively studied as anticancer agents considering their availability, low toxicity, and economic affordability. Over 60% of anticancer drugs are natural products that have shown potential anticancer activities [2], such as anti-proliferation [3], anti-angiogenesis [4], reversal of multidrug resistance (MDR) [5], and antimetastasis [6] effects.
The genus Aconitum belongs to the family Ranunculaceae, which comprises about 400 species distributed in the temperate regions of the northern hemisphere, with half of them distributed in China [7]. Since ancient times, about 40 of these species have been widely used to treat apoplexy hemiplegia [8], asthma [9], and rheumatoid arthritis [10] in China, Japan, and other Asian countries; some examples are A. carmichaeli Debx., A. kusnezoffii Rchb., A. sinomontanum Nakai, and A. leucostomum Vorosch. [11]. Modern pharmacological studies have demonstrated that medicinal Aconitum plants can exert anti-inflammatory, analgesic [12,13], anti-arrhythmia [14], antioxidant [15], antibacterial [16], and anticancer effects [7,17]. In anticancer therapy, Aconitum plants have been proven effective for several types of cancer, such as lung, stomach, and liver cancers [18,19,20].
Aconitum plants chemically comprise alkaloids, flavonoids, steroids, and glycosides, and the main efficacy components as well as the toxic components are diterpenoid alkaloids [21,22,23], which are reportedly some of the most promising, naturally abundant compounds for treating cancer [24]. Diterpenoid alkaloids have been studied since the 1940s, and based on structural differences such as the number of carbon atoms on the mother nucleus, diterpenoid alkaloids are generally divided into four categories: C18-, C19-, C20-, and bis-diterpenoid alkaloids [25,26,27,28,29]. This review focuses on the progress of diterpenoid alkaloids with different structures derived from Aconitum plants and some of their derivatives (e.g., lappaconitine, aconitine, songorine, pseudokobusine, and 11-veratroylpseudokobusine) with potential anticancer activities. We also summarize some of their antitumor mechanisms. We hope this work can serve as a reference for further developing Aconitum diterpenoid alkaloids as anticancer agents.

2. Chemical Structure of Diterpenoid Alkaloids

Nearly a thousand natural diterpenoid alkaloids have been reported to date, and a large part of them originate from Aconitum plants [30], and C19-diterpenoid alkaloids are the most reported among them [27]. C19-diterpenoid alkaloids evolve from C20-diterpenoid alkaloids and degenerate into C18-diterpenoid alkaloids by losing the 18th carbon atom [25,31].
Based on the presence of oxygen-containing functional groups at the C-7 position, C18-diterpenoid alkaloids, which constitute a small group within the diterpenoid alkaloids, are classified aslappaconine or ranaconine types [28]. Concerning the carbon skeleton and substituents at specific positions, the C19-diterpenoid alkaloids may be initially divided into aconitine, lycoctonine, pyro, lactonepe, 7,17-seco, and rearranged types [27]. Compared with C18- and C19-diterpenoid alkaloids, the skeletal types of the C20-diterpenoid alkaloids are extremely complex, which may be divided into four classes, including 19 types [29]. The majority of C20-diterpenoid alkaloids have an exocyclic double bond structure and are generally divided into atisine, denudatine, hetidine, hetisine, napelline, and anopterine types nowadays [32].
Figure 1 and Figure 2 show the chemical structures of C18-, C19-, bis-diterpenoid alkaloids and C20-diterpenoid alkaloids with anticancer activities derived from genus Aconitum, respectively.

3. Anticancer Activities of Diterpenoid Alkaloids

3.1. C18-Diterpenoid Alkaloids

Lappaconitine (1), a typical C18-diterpenoid alkaloid extracted for the first time in China from A. sinomontanum Nakai, is commonly used as postoperative analgesia and relief for clinical cancer pain as a non-addictive analgesic [33,34]. Lappaconitine exerts an analgesic effect by inhibiting the voltage-dependent sodium channels, increasing norepinephrine release in the synaptic cleft, and inhibiting the release of substance P [35]. Lappaconitine reportedly inhibits the proliferation of the human non-small cell lung cancer cells A549 dose dependently [36]. With increased lappaconitine concentration, the proportion of A549 cells increased gradually in G1 + G0 phase and decreased in S and G2+M phases, and the apoptosis rate increased with the down-regulated expression of Cyclin E1. Lappaconitine can also inhibit the expression of VEGF-A, and the combination of lappaconitine and oxaliplatin can arrest the cells in G1/G0 phase and inhibit the expression of Cyclin E1 [37].
As the derivative of lappaconitine (1), lappaconite hydrobromide can reportedly exert an efficient antitumor effect in mice by the National Institutes of Health (NIH) mice. In particular, the inhibition rates ranged within 11.20%–53.08%for liver tumor growth and within 29.81%–53.96% for S180 tumor growth [38].

3.2. C19-Diterpenoid Alkaloids

Lycaconitine (2) is a C19-diterpenoid alkaloid isolated from the roots of Aconitum pseudo-laeve var. erectum through bioassay-guided fractionation and repeated column chromatography. Although lycaconitine (2) does not present cytotoxicity to KB cells, it has potent inhibitory effects on pgp-MDR upon testing on the multidrug resistant human fibrocarcinoma KB V20C (resistant to 20 nM vincristine) [39].
In the 1980s, preliminary experimental studies on the antitumor effect of aconitine (3) were performed by multiple medical institutions. They demonstrated that 200 μg/mL aconitine inhibited the proliferation of gastric cancer cells by inhibiting its mitosis, and that the inhibitory rate of hepatocellular carcinoma in mice was 47.77%–57.38% [18]. Aconitine also reportedly has anticancer activity to the mice inoculated with gastric cancer cells and S180 cells, as well as the ability to inhibit the spontaneous metastasis of Lewis lung cancer cells [40]. Moreover, aconitine (150–400 μg/mL) can significantly inhibit the proliferation of Hepal-6 hepatoma cells in vitro, with the inhibitory rate of Hepal-6 cells in C75BL/6 male mice ranging within 26.12%–65.43% at concentrations of 0.15 and 0.375 mg/kg [41].
MDR is a key factor that hinders cancer treatment. The anticancer effect of aconitine (3) has been evaluated in drug-resistant human oral squamous cell carcinoma (KBv200), which shows that aconitine has a small inhibitory effect on the growth of KBv200 (IC50 = 224.91 μg/mL). However, aconitine can increase the sensitivity of vincristine to kill cells, and the IC50 values of vincristine in KBv200 are 0.2715 and 0.9185 μg/mL when combined with 12.5 and 6.25 μg/mL aconitine, respectively [42]. Thus, aconitine is considered to have no significant cytotoxic effect and can even reverse the MDR of cancer cells. Through immunohistochemistry and gene chip technology, follow-up studies has shown that aconitine can downregulate the expression of Protein Pgp and change the expression of Mdr1 gene by affecting apoptosis-related genes and the mitogen-activated protein kinase (MAPK) signal transduction system, thereby ultimately reversing the drug resistance [43,44].
(1α,6α,8α,14α,16α)-20-Ethyl-8,14-dihydroxy-1,6,16-trimethoxy-4-(methoxymethyl)-aconitane (4) was isolated from the roots of Aconitum taipaicum Hand.–Mazz, and cytotoxicity assays indicate that compound 4 exhibits stronger growth inhibitory than adriamycin against leukaemia cells HL-60 and K-562 [45]. In the same year, compound 4 has been found to inhibit the proliferation and invasion of HepG2 (liver hepatocellular carcinoma) cells and arrest cells in G0/G1 phase to promote cell apoptosis, the mechanism involves the upregulation of Bax and Caspase-3 expression and the downregulation of Bcl-2 (B-cell lymphoma-2) and CCND1 expression [46].
Found in A. carmichaeli Debx., five compounds including oxonitine (5), deoxyaconitine (6), hypaconitine (7), mesaconitine (8), and crassicauline A (9) show obvious cytotoxic activities against various cancers, such as leucocythemia, breast cancer, and liver cancer. Compared with two other diterpenoid alkaloids without cytotoxic activities, compounds 57 and 9 have two ester groups in the structure, which may have an effect on the cytotoxicity of the compounds [47]. 8-O-Azeloyl-14-benzoylaconine (10) is also a new C19-diterpenoid alkaloid with two ester groups in the structure found in the roots of A. karacolicum Rapcs. It shows good antiproliferative activities with an IC50 of about 10–20 µM against HCT-15 (colon cancer cell), A549 (lung cancer cell line), and MCF-7 (breast cancer cell line) cells [48].
Cammaconine (11) was isolated from the ethanol extract of Aconitum vaginatum Pritz. and identified by spectroscopic analysis. It has greater inhibitory effect on AGS (gastric cancer cell), HepG2, and A549 cells compared with 5-Fluorouracil [49]. Two C19-diterpenoid alkaloids, neoline (12) and 14-O-acetylneoline (13) were further isolated and identified from an enriched alkaloid fraction of Aconitum flavum Hand.–Mazz; they have been proven to possess growth-inhibition effects on human gastric carcinoma SGC-7901, hepatic carcinoma HepG2, and lung cancer A549 cells [50].

3.3. C20-Diterpenoid Alkaloids

Together with cammaconine (11), anatisine-type C20-diterpenoid alkaloid named atisinium chloride (14) was isolated from A. vaginatum Pritz. and found to inhibit the growth of various cancers [49]. In addition, songorine (15), 12-epi-napelline (16), and 12-epi-dehydronapelline (17) derived from Aconitum flavum Hand.–Mazz. inhibited the growth of SGC-7901 (gastric carcinoma), HepG2, and A549 cells such as neoline (12) [50].
In 2007, 13 natural diterpenoid alkaloids were isolated and purified from Aconitum yesoense var. macroyesoense and Aconitum japonicum and 22 derivatives were subsequently prepared from the parent alkaloids. The veatchine-type C20-diterpenoid alkaloid named 12-acetylluciculine (18) and the six derivatives designed from pseudokobusine (19), including 6,11-dibenzoylpseudokobusine (20), 11-veratroylpseudokobusine (21), 11-cinnamoylpseudokobusine (22), 11-(m-trifluoromethylbenzoyl)pseudokobusine (23), 11-anisoylpseudokobusine (24), and 11-p-nitrobenzoylpseudokobusine (25) are proven to inhibit the growth of human malignant A172 cells [51]. The hydroxyl groups at C-6 and C-15 of pseudokobusine are considered to be essential to the inhibitory effect, and the esterification of the hydroxyl group at C-11 may enhance such activity. In 2009, Koji Wada detected the anticancer activities of the same above mentioned diterpenoid alkaloids with four different cancer cells. They demonstrated that all six derivatives (2025) have strong inhibitory activity against A172, A549, HeLa (cervical cancer cell line), and Raji (lymphoma cell line) cells (except compound 21 to HeLa cells) [52]. Compounds 23 and 24, which show significant suppressive effects against Raji cells, have the same structure except for the group in the C-11 position. Compound 23 inhibits the phosphorylation of extracellular signal-regulated kinasein Raji cells but does not affect the growth of human CD34+ hematopoietic stem/progenitor cells, which can be significantly inhibited by compound 24 [53].
Ten new acylated alkaloid derivatives were prepared from the natural diterpenoid alkaloids of A. yesoense var. macroyesoense and A. japonicum; they are 11,15-dianisoylpseudokobusine (26), 11,15-di-p-nitrobenzoylpseudokobusin (27), 11-(p-trifluoromethylbenzoyl)kobusine (28), 11-(m-trifluoromethylbenzoyl)kobusine (29), 11,15-di-p-nitrobenzoylkobusine (30), 11-p-nitrobenzoylpseudokobusine (31), 11-cinnamoylpseudokobusine (32), 6,11-dianisoylpseudokobusine (33), 11-veratroylpseudokobusine (34), and 11-anisoylpseudokobusine (35). They inhibited the growth of A549 cells through G1 arrest, and their IC50 values ranged within 1.72–5.44 µM. Their cytotoxic effects can be enhanced by replacing an acyl group at both C-11 and C-15 positions [54].
In 2015, the antiproliferative effects of 108 diterpenoid alkaloids were tested by the same research team above against four cancer cells, namely, lung, prostate, nasopharyngeal, and vincristine-resistant nasopharyngeal (KB-VIN) cancer cell lines. The alkaloids that show substantial suppressive effects in 11 newly synthesized C20-diterpenoid alkaloid derivatives [55]: 11,15-dibenzoylkobusine (36), 11,15-dianisoylkobusine (37), 11,15-di-(4-nitrobenzoyl)kobusine (38), 11,15-di-(4-fluorobenzoyl)kobusine (39), 11,15-di-(3-trifluoromethylcinnamoyl)kobusine (40), 11,15-dibenzoylpseudokobusine (41), 11-(4-nitrobenzoyl)pseudokobusine (42), 11,15-di-(3-nitrobenzoyl)pseudokobusine (43), 11-(3-trifluoromethylbenzoyl)pseudokobusine (44), 11-cinnamoylpseudokobusine (45), and 11-tritylpseudokobusine (46). All of them were hetisine-type C20-diterpenoid alkaloids with two different substitution patterns of C-11 and C-11, 15, and the GI50s of them were summarized in Table 1.

3.4. Bis-Diterpenoid Alkaloids

Three bis-[O-(14-benzoylaconine-8-yl)]esters [56], including new semisynthetic alkaloids with diverse alkyl chains on the heterocyclic moiety, including bis-[O-(14-benzoylaconine-8-yl)]-pimelate (47), bis-[O-(14-benzoylaconine-8-yl)]-suberate (48), and bis-[O-(14-benzoylaconine-8-yl)]-azelate (49), built from the 8-O-azeloyl-14-benzoylaconine (11) skeleton, present remarkable cytotoxic activity in vitro against lung cancer A-549, colon cancer HCT-15, and breast cancer MCF-7 cells; their IC50s were <28 µM. The anticancer activities in vivo of bis-[O-(14-benzoylaconine-8-yl)]-suberate (48) was subsequently tested in immunodeficient mice transplanted with human tumors MCF-7 and HCT-15 cells because of its significant cytotoxicity in vitro. Its antitumor activity is obviously shown at a dose below the maximum tolerated dose. The impact of the alkyl-linker length of the designed bis-diterpenoid alkaloids on cytotoxicity is clearly elucidated in the study and can serve as a reference for designing novel antiproliferative agents [57].

4. Discussion and Conclusions

Diterpenoid alkaloids isolated and designed from Aconitum plants have shown effective anticancer properties in various cancer cell lines. Such properties include inhibiting cell growth, inducing apoptosis, interfering with the cell cycle, and altering MDR. The in vitro anticancer activities (IC50 values) of diterpenoid alkaloids derived from Aconitum and their derivatives are presented in Table 1. Some of them also exert noteworthy anticancer effects in animal models.
Most of natural diterpenoid alkaloids with anticancer effect in Aconitum are C19-diterpenoid alkaloids, although derivatives of C20-diterpenoid alkaloids also have notable anticancer potential. Many diterpenoid alkaloids tend to exhibit improved activity after simple structural modification [58], and many structures may affect the activity of a compound, such as the kind and position of substituents and the linker-chain length [59].
Diterpenoid alkaloids from Aconitum have great potential use as new drugs for treating cancer. This review can serve as a useful reference for researchers in their search for highly effective, low-toxicity diterpenoid alkaloids through structure modification and structure–activity analysis. We also provide a theoretical basis for safety medication in clinical settings and further development of new anticancer drugs.

Acknowledgments

This work was supported by a grant from the National Natural Science Foundation of China (No. 81030066).

Author Contributions

M.-Y.R. conceived and wrote the manuscript; Q.-T.Y. and C.-Y.S. collected the literature; X.-M.T., L.-L.J. edited the information of chemical components; J.-B.L. revised the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stewart, B.W.; Wild, C.P. World Cancer Report 2014; International Agency for Research on Cancer: Lyon, France, 2014. [Google Scholar]
  2. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the last 25 years. J. Nat. Prod. 2007, 70, 461–477. [Google Scholar] [CrossRef] [PubMed]
  3. Simoben, C.V.; Ibezim, A.; Ntie-Kang, F.; Nwodo, J.N.; Lifongo, L.L. Exploring cancer therapeutics with natural products from African medicinal plants, part I: Xanthones, quinones, steroids, coumarins, phenolics and other classes of compounds. Anti-Cancer Agents Med. Chem. 2015, 15, 1092–1111. [Google Scholar] [CrossRef]
  4. Wang, Z.; Dabrosin, C.; Yin, X.; Fuster, M.M.; Arreola, A.; Rathmell, W.K.; Generali, D.; Nagaraju, G.P.; El-Rayes, B.; Ribatti, D.; et al. Broad targeting of angiogenesis for cancer prevention and therapy. Semin. Cancer Biol. 2015, 35, S224–S243. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, M.; Guan, X.; Chi, Y.; Robinson, N.; Liu, J.P. Chinese herbal medicine as adjuvant treatment to chemotherapy for multidrug-resistant tuberculosis (MDR-TB): A systematic review of randomised clinical trials. Tuberculosis (Edinb.) 2015, 95, 364–372. [Google Scholar] [CrossRef] [PubMed]
  6. Nakamura, K.; Shinozuka, K.; Yoshikawa, N. Anticancer and antimetastatic effects of cordycepin, an active component of Cordyceps sinensis. J. Pharmacol. Sci. 2015, 127, 53–56. [Google Scholar] [CrossRef] [PubMed]
  7. Zhou, G.H.; Tang, L.Y.; Zhou, X.D.; Wang, T.; Kou, Z.Z.; Wang, Z.J. A review on phytochemistry and pharmacological activities of the processed lateral root of Aconitum carmichaelii Debeaux. J. Ethnopharmacol. 2015, 60, 173–193. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, X.Z.; Zhu, P.L. Experience for treating stroke with Aconitum from professor WANG Xin-zhi. Clin. J. Chin. Med. 2012, 4, 96–97. [Google Scholar]
  9. Guo, W.L.; Shi, S.F. Professor Suofang Shi using high–dose Aconite to treat asthma with cold symptoms. Jilin J. Tradit. Chin. Med. 2011, 31, 112–114. [Google Scholar]
  10. Gu, Y.R.; Tong, B.L. 30 cases of the treatment of rheumatoid arthritis with high–dose Aconite. J. Anhui Univ. Chin. Med. 1996, 15, 25. [Google Scholar]
  11. Li, Q.; Guo, L.N.; Zheng, J.; Ma, S.C. Reaserch progress of medicinal genus Aconitum. Chin. J. Pharm. Anal. 2016, 36, 1129–1145. [Google Scholar]
  12. Shao, F.; Li, S.L.; Liu, R.H.; Huang, H.L.; Ren, G.; Yao, Y.X.; Hao, X.C. Analgesic and anti-inflammatory effects of different processed products of Aconiti lateralis radix praeparata. Lishizhen. Med. Mater. Med. Res. 2011, 22, 2329–2330. [Google Scholar]
  13. Li, L.J.; Zhang, F.L.; Wu, R.Z.; Lin, Q.; Liu, P.R. A comparative study on the functions of anti-inflammatory and analgesic of monkshood and its small pieces processed by a new method. Yunnan J. Tradit. Chin. Med. Mater. Med. 2004, 25, 34–35. [Google Scholar]
  14. Tong, Y.; Li, N.; Wu, X.Q. Effect of Fuzi on cAMP-PKA signal transduction pathways in rat of chronic arrhythmia. Pharmacol. Clin. Chin. Mater. Med. 2013, 29, 90–92. [Google Scholar]
  15. Gao, T.T.; Ma, S.; Song, J.Y.; Bi, H.T.; Tao, Y.D. Antioxidant and immunological activities of water-soluble polysaccharides from Aconitum kusnezoffii Reichb. Int. J. Biol. Macromol. 2011, 49, 580–586. [Google Scholar] [CrossRef] [PubMed]
  16. Shi, Y.B.; Liu, L.; Shao, W.; Wei, T.; Lin, G.M. Microcalorimetry studies of the antimicrobial actions of Aconitum alkaloids. J. Zhejiang Univ. Sci. B 2015, 16, 690–695. [Google Scholar] [CrossRef] [PubMed]
  17. Fan, Y.P.; Jiang, Y.D.; Liu, J.J.; Kang, Y.X.; Li, R.Q.; Wang, J.Y. The anti-tumor activity and mechanism of alkaloids from Aconitum szechenyianum Gay. Bioorg. Med. Chem. Lett. 2016, 26, 380–387. [Google Scholar] [CrossRef] [PubMed]
  18. Huang, Y.R. A review on anticancer activity of Aconitum. J. Fujian Univ. Tradit. Chin. Med. 1991, 22, 54–56. [Google Scholar]
  19. Bai, X.Y. Toxicity and anti-tumor effect of Aconitum carmichaelii. J. Pract. Tradit. Chin. Med. 2005, 21, 125. [Google Scholar]
  20. Xu, Q.P.; Liu, J.H.; Liu, B.R. Progress in study on antitumor activity of C19-, C20-diterpenoid alkaloids. Progr. Pharm. Sci. 2016, 40, 3–10. [Google Scholar]
  21. Nyirimigabo, E.; Xu, Y.Y.; Li, Y.B.; Wang, Y.M.; Agyemang, K.; Zhang, Y.J. A review on phytochemistry, pharmacology and toxicology studies of Aconitum. J. Pharm. Pharmacol. 2015, 67, 1–19. [Google Scholar] [CrossRef] [PubMed]
  22. Xu, T.F.; Liu, S.; Meng, L.L.; Pi, Z.F.; Song, F.R.; Liu, Z.Q. Bioactive heterocyclic alkaloids with diterpene structure isolated from traditional Chinese medicines. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2016, 1026, 56–66. [Google Scholar] [CrossRef] [PubMed]
  23. Ai, C.; Zhu, Y.Y.; Zhao, C.Q. Recent advances on chemical constituents, pharmacological study and the endophytes of the genus Aconitum. Nat. Prod. Res. Dev. 2012, 24, 248–259. [Google Scholar]
  24. Zhang, Y.; Xiang, C. Research progress of antitumor drugs. Jiangxi Med. J. 2004, 39, 445–448. [Google Scholar]
  25. An, J.X.; Liu, F.; Liu, F.; Zeng, G.Y.; Zhou, Y.J. Recent research progress on diterpenoid alkaloids from genus Aconitum and their analgesic activity. Cent. South Pharm. 2016, 14, 521–522. [Google Scholar]
  26. Wang, F.P.; Liang, X.T. Chemistry of the diterpenoid alkaloids. In The Alkaloids: Chemistry and Pharmacology; Cordell, G.A., Ed.; Academic Press: London, UK, 1992; Volume 42, pp. 151–247. [Google Scholar]
  27. Wang, F.P.; Chen, Q.H. C19-diterpenoid alkaloids. In The Alkaloids: Chemistry and Biology; Cordell, G.A., Ed.; Academic Press: London, UK, 2010; Volume 69, pp. 1–609. [Google Scholar]
  28. Wang, F.P.; Chen, Q.H.; Liang, X.-T. C18-diterpenoid alkaloids. In The Alkaloids: Chemistry and Biology; Cordell, G.A., Ed.; Academic Press: London, UK, 2009; Volume 67, pp. 1–78. [Google Scholar]
  29. Wang, F.P.; Liang, X.T. C20-diterpenoid alkaloids. In The Alkaloids: Chemistry and Biology; Cordell, G.A., Ed.; Academic Press: London, UK, 2002; Volume 59, pp. 1–280. [Google Scholar]
  30. Xue, J.; Yang, C.H.; Liu, J.H.; Liang, J.Y.; Tang, Q.F.; Zhang, S.J. Recent advance of diterpenoid alkaloids in genus Aconitum. Strait Pharm. J. 2009, 21, 1–10. [Google Scholar]
  31. Cai, C.Q.; Yang, C.H.; Liang, J.Y.; Liu, J.H. Advance in studies on structure-activity relationships of diterpenoid alkaloids in genus Aconitum. Strait Pharm. J. 2013, 25, 1–4. [Google Scholar]
  32. Wang, F.P.; Chen, Q.H.; Liu, X.Y. Diterpenoid alkaloids. Nat. Prod. Rep. 2010, 27, 529–570. [Google Scholar] [CrossRef] [PubMed]
  33. Gong, Q.A.; Li, M. Effect of lappaconitine on postoperative pain and serum complement 3 and 4 levels of cancer patients undergoing pectum surgery. Chin. J. Integr. Tradit. West. Med. 2015, 35, 668–672. [Google Scholar]
  34. Su, M.Y. Study of Lappaconitine with Ropivacaine for Postoperative Analgesia for Cancer of Stomach. Master Thesis, Zhengzhou University, Zhengzhou, China, 28 April 2005. [Google Scholar]
  35. Lin, C.C.; Chen, W.N.; Chen, C.J.; Lin, Y.W.; Zimmer, A.; Chen, C.C. An antinociceptive role for substance P in acid-induced chronic muscle pain. Proc. Natl. Acad. Sci. USA 2012, 109, E76–E83. [Google Scholar] [CrossRef] [PubMed]
  36. Sheng, L.R.; Xu, M.; Xu, L.Q.; Xiong, F. Cytotoxic effect of lappaconitine on non-small cell lung cancer in vitro and its molecular mechanism. J. Chin. Med. Mater. 2014, 37, 840–843. [Google Scholar]
  37. Sheng, L.R. The Effect of Lappaconitine and Its Synergistic Effect with Docetaxol and Oxaliplatinon Lung Cancer. Master Thesis, Jinan University, Guangzhou, China, 4 May 2010. [Google Scholar]
  38. Lin, L.; Xiao, L.Y.; Lin, P.Y.; Zhang, D.; Chen, Q.W. Experimental study on the anti-tumor effect of lappaconitine hydrobromide. TCM Res. 2005, 18, 16–18. [Google Scholar]
  39. Kim, D.K.; Kwon, H.Y.; Lee, K.R.; Rhee, D.K.; Zee, O.P. Isolation of a multidrug resistance inhibitor from Aconitum pseudo-laeve var. erectum. Arch. Pharm. Res. 1998, 21, 344–347. [Google Scholar] [CrossRef] [PubMed]
  40. Tang, X.M.; Sun, G.Z. Study on anti-tumor and antimetastasis effects and clinical treatment of cancer of aconitine. Beijing J. Tradit. Chin. Med. 1986, 8, 27–28. [Google Scholar]
  41. Qian, Z. The Effect and Preliminary Mechanism Study on Monkshood Polysaccharide Combined with Aconitine to the Hepatocellualar Carcinoma Cell. Master Thesis, Nanjing University of Chinese Medicine, Nanjing, China, June 2015. [Google Scholar]
  42. Liu, X.Q.; Chen, X.Y.; Wang, Y.Z.; Yuan, S.J.; Tang, Y. Study on reversing multi-drug tolerance of KBV200 cell by aconitine. Chin. J. Basic Med. Tradit. Chin. Med. 2004, 10, 55–57. [Google Scholar]
  43. Hou, L.; Liu, X.Y.; Chen, X.Y.; Zhang, K.T.; Wang, Y.Z.; Wang, X.M. Using gene chip technology to investigate the mechanism of aconitine's reversing the drug resistan. Chin. J. Inf. Tradit. Chin. Med. 2005, 12, 34–36. [Google Scholar]
  44. Tian, S.D.; Liu, X.Q.; Wang, X.M.; Tang, Y.; Chen, X.Y. Immunohistochemistry study of aconitine on the expression of Pgp protein in KBV200 cell. Chin. Arch. Tradit. Chin. Med. 2006, 24, 55–56. [Google Scholar]
  45. Guo, Z.J.; Xu, Y.; Zhang, H.; Li, M.Y.; Xi, K. New alkaloids from Aconitum taipaicum and their cytotoxic activities. Nat. Prod. Res. 2014, 28, 164–168. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, H.; Guo, Z.J.; Han, L.; You, X.Y.; Xu, Y. The antitumor effect and mechanism of taipeinine A, a new C19-diterpenoid alkaloid from Aconitum taipeicum, on the HepG2 human hepatocellular carcinoma cell line. J. BUON 2014, 19, 705–712. [Google Scholar] [PubMed]
  47. Gao, F.; Li, Y.Y.; Wang, D.; Huang, X.; Liu, Q. Diterpenoid alkaloids from the Chinese traditional herbal “Fuzi” and their cytotoxic activity. Molecules 2012, 17, 5187–5194. [Google Scholar] [CrossRef] [PubMed]
  48. Chodoeva, A.; Bosc, J.J.; Guillon, J.; Decendit, A.; Petraud, M.; Absalon, C.; Vitry, C.; Jarry, C.; Robert, J. 8-O-Azeloyl-14-benzoylaconine: A new alkaloid from the roots of Aconitum karacolicum Rapcs and its antiproliferative activities. Bioorg. Med. Chem. 2005, 13, 6493–6501. [Google Scholar] [CrossRef] [PubMed]
  49. Zhu, T. Studies on the Antitumor Constituents of Aconitum vaginayum. Master Thesis, Huazhong University of Science and Technology, Wuhan, China, 30 May 2008. [Google Scholar]
  50. Hao, W.J. Study on the Chemical Constituents of the Alkaloids from the Root of Aconitum flavum Hand.—Mazz and Its Anti-Tumor Activities. Master Thesis, Ningxia Medical University, Yinchuan, China, April 2014. [Google Scholar]
  51. Wada, K.; Hazawa, M.; Takahashi, K.; Mori, T.; Kawahara, N.; Kashiwakura, I. Inhibitory effects of diterpenoid alkaloids on the growth of A172 human malignant cells. J. Nat. Prod. 2007, 70, 1854–1858. [Google Scholar] [CrossRef] [PubMed]
  52. Hazawa, M.; Wada, K.; Takahashi, K.; Mori, T.; Kawahara, N.; Kashiwakura, I. Suppressive effects of novel derivatives prepared from Aconitum alkaloids on tumor growth. Investig. New Drugs 2009, 27, 111–119. [Google Scholar] [CrossRef] [PubMed]
  53. Hazawa, M.; Takahashi, K.; Wada, K.; Mori, T.; Kawahara, N.; Kashiwakura, I. Structure-activity relationships between the Aconitum C20-diterpenoid alkaloid derivatives and the growth suppressive activities of Non-Hodgkin’s lymphoma Raji cells and human hematopoietic stem/progenitor cells. Investig. New Drugs 2011, 29, 1–8. [Google Scholar] [CrossRef] [PubMed]
  54. Wada, K.; Hazawa, M.; Takahashi, K.; Mori, T.; Kawahara, N.; Kashiwakura, I. Structure-activity relationships and the cytotoxic effects of novel diterpenoid alkaloid derivatives against A549 human lung carcinoma cells. J. Nat. Med. 2011, 65, 43–49. [Google Scholar] [CrossRef] [PubMed]
  55. Wada, K.; Ohkoshi, E.; Zhao, Y.; Goto, M.; Morris-Natschke, S.L.; Lee, K.H. Evaluation of Aconitum diterpenoid alkaloids as antiproliferative agents. Bioorg. Med. Chem. Lett. 2015, 25, 1525–1531. [Google Scholar] [CrossRef] [PubMed]
  56. Chodoeva, A.; Bosc, J.J.; Guillon, J.; Costet, P.; Decendit, A.; Mérillon, J.M.; Léger, J.M.; Jarry, C.; Robert, J. Hemisynthesis and antiproliferative properties of mono-[O-(14-benzoylaconine-8-yl)]esters and bis-[O-(14-benzoylaconine-8-yl)]esters. Eur. J. Med. Chem. 2012, 54, 343–351. [Google Scholar] [CrossRef] [PubMed]
  57. Chodoeva, A.; Bosc, J.J.; Lartigue, L.; Guillon, J.; Auzanneau, C.; Costet, P.; Zurdinov, A.; Jarry, C.; Robert, J. Antitumor activity of semisynthetic derivatives of Aconitum alkaloids. Investig. New Drugs 2014, 32, 60–67. [Google Scholar] [CrossRef] [PubMed]
  58. Akhtar, J.; Khan, A.A.; Ali, Z.; Haider, R.; Shahar, Y.M. 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]
  59. Traboulsi, T.; El-Ezzy, M.; Gleason, J.L.; Mader, S. Antiestrogens: Structure-activity relationships and use in breast cancer treatment. J. Mol. Endocrinol. 2017, 58, R15–R31. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of the C18-, C19-, and bis-diterpenoid alkaloids with anticancer activities derived from the genus Aconitum.
Figure 1. Chemical structures of the C18-, C19-, and bis-diterpenoid alkaloids with anticancer activities derived from the genus Aconitum.
Molecules 22 00267 g001
Figure 2. Chemical structures of the C20-diterpenoid alkaloids with anticancer activities derived from the genus Aconitum.
Figure 2. Chemical structures of the C20-diterpenoid alkaloids with anticancer activities derived from the genus Aconitum.
Molecules 22 00267 g002
Table 1. The anti-proliferative activities of the diterpenoid alkaloids derived from the genus Aconitum.
Table 1. The anti-proliferative activities of the diterpenoid alkaloids derived from the genus Aconitum.
No.CompoundsCancer TypesCell LinesIC50Reference
C18-diterpenoid alkaloids
1LappaconitineLung cancerA5496.71 × 103 µM/48 h[39]
C19-diterpenoid alkaloids
2LycaconitineFibroblast carcinomaKB V20C110.65 µM/72 h[39]
3AconitineOral squamous cell carcinomaKBv200348.29 µM/72 h[42]
Hepatoma carcinomaHepal-6590.03 µM/48 h[41]
Hepatoma carcinomaHePG20.85 × 10−2 µM/72 h[47]
Colon cancerHCT88.12 × 10−2 µM/72 h
Breast cancerMCF72.45 × 10−2 µM/72 h
4(1α,6α,8α,14α,16α)-20-ethyl-8,14-dihydroxy-1,6,16-trimethoxy-4-(methoxymethyl)-aconitaneLeukemiaHL-600.44 µM/24 h[46]
LeukemiaK-5621.55 µM/24 h
5OxonitineColon cancerHCT829.48 × 10−2 µM/72 h[47]
Breast cancerMCF73.13 × 10−2 µM/72 h
Hepatoma carcinomaHePG28.61 × 10−2 µM/72 h
6DeoxyaconitineColon cancerHCT85.14 × 10−2 µM/72 h[47]
Breast cancerMCF710.35 × 10−2µM/72 h
Hepatoma carcinomaHePG29.21 × 10−2 µM/72 h
7HypaconitineColon cancerHCT812.05 × 10−2 µM/72 h[47]
Breast cancerMCF76.46 × 10−2 µM/72 h
Hepatoma carcinomaHePG20.92 × 10−2 µM/72 h
8MesaconitineColon cancerHCT813.16 × 10−2 µM/72 h[47]
Breast cancerMCF74.57 × 10−2 µM/72 h
Hepatoma carcinomaHePG21.45 × 10−2 µM/72 h
9Crassicauline AColon cancerHCT816.45 × 10−2 µM/72 h[47]
Breast cancerMCF712.86 × 10−2 µM/72 h
Hepatoma carcinomaHePG22.36 × 10−2 µM/72 h
108-O-Azeloyl-14-benzoylaconineColon cancerHCT-1516.8 µM/24h[48]
Lung cancerA54919.4 µM/24 h
Breast cancerMCF-710.3 µM/24 h
11CammaconineGastric carcinomaAGS0.32 µM/48 h[49]
Hepatoma carcinomaHepG234.55 µM/48 h
Lung cancerA5490.32 µM/48 h
12NeolineGastric carcinomaSGC-790137.55 µM/48 h[50]
Hepatoma carcinomaHepG228.36 µM/48 h
Lung cancerA54934.74 µM/48 h
1314-O-acetylneolineGastric carcinomaSGC-790116.97 µM/48 h[50]
Hepatoma carcinomaHepG233.76 µM/48 h
Lung cancerA54918.75 µM/48 h
C20-diterpenoid alkaloids
14Atisinium chlorideGastric carcinomaAGS0.44 µM/48 h[49]
Hepatoma carcinomaHepG266.69 µM/48 h
Lung cancerA5492.29 µM/48 h
15SongorineGastric carcinomaSGC-790146.55 µM/48 h[50]
Hepatoma carcinomaHepG287.72 µM/48 h
Lung cancerA54961.90 µM/48 h
1612-epi-napellineGastric carcinomaSGC-790164.79 µM/48 h[50]
Hepatoma carcinomaHepG296.99 µM/48 h
Lung cancerA54965.91 µM/48 h
1712-epi-dehydronapellineGastric carcinomaSGC-790165.00 µM/48 h[50]
Hepatoma carcinomaHepG246.63 µM/48 h
Lung cancerA54976.50 µM/48 h
1812-acetylluciculineMalignant gliomaA17213.95 µM/24 h[51]
19PseudokobusineMalignant gliomaA172>15.18 µM/24 h[51]
206,11-dibenzoylpseudokobusineMalignant gliomaA1722.42 µM/24 h[51]
2111-veratroylpseudokobusineMalignant gliomaA1722.52 µM/24 h[51]
Lung cancerA5493.5 µM/24 h[52]
2211-cinnamoylpseudokobusineMalignant gliomaA1721.94 µM/24 h[51]
Lung cancerA5495.1 µM/24 h[52]
2311-(m-trifluoromethylbenzoyl)pseudokobusineMalignant gliomaA172Not shown[51]
Lung cancerA5494.4 µM/24 h[52]
Lung cancerA5494.67 µM/24 h[54]
LymphomaRaji4.39 µM/96 h[53]
2411-anisoylpseudokobusineMalignant gliomaA1722.80 µM/24 h[51]
Lung cancerA5491.7 µM/24 h[52]
LymphomaRaji5.18 µM/96 h[53]
2511-p-nitrobenzoylpseudokobusineMalignant gliomaA1723.13 µM/24 h[51]
Lung cancerA5493.5 µM/24 h[52]
2611,15-dianisoylpseudokobusineLung cancerA5491.72 µM/24 h[54]
2711,15-di-p-nitrobenzoylpseudokobusinLung cancerA5492.66 µM/24 h[54]
2811-(p-trifluoromethylbenzoyl)kobusineLung cancerA5495.44 µM/24 h[54]
2911-(m-trifluoromethylbenzoyl)kobusineLung cancerA5493.75 µM/24 h[54]
3011,15-di-p-nitrobenzoylkobusineLung cancerA5495.08 µM/24 h[54]
3111-p-nitrobenzoylpseudokobusineLung cancerA5494.24 µM/24 h[54]
3211-cinnamoylpseudokobusineLung cancerA5493.02 µM/24 h[54]
336,11-dianisoylpseudokobusineLung cancerA5493.68 µM/24 h[54]
3411-veratroylpseudokobusineLung cancerA5494.07 µM/24 h[54]
3511-anisoylpseudokobusineLung cancerA5492.20 µM/24 h[54]
3611,15-dibenzoylkobusineLung cancerA549GI50 = 8.4 µM/72 h[55]
Prostate cancerDU145GI50 = 9.3 µM/72 h
Epidermoid carcinomaKBGI50 = 6.0 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 7.5 µM/72 h
3711,15-dianisoylkobusineLung cancerA549GI50 = 6.7 µM/72 h[55]
Prostate cancerDU145GI50 = 7.1 µM/72 h
Epidermoid carcinomaKBGI50 = 5.3 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 5.2 µM/72 h
3811,15-di-(4-nitrobenzoyl)kobusineLung cancerA549GI50 = 6.9 µM/72 h[55]
Prostate cancerDU145GI50 = 7.0 µM/72 h
Epidermoid carcinomaKBGI50 = 5.3 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 5.5 µM/72 h
3911,15-di-(4-fluorobenzoyl)kobusineLung cancerA549GI50 = 8.1 µM/72 h[55]
Prostate cancerDU145GI50 = 6.8 µM/72 h
Epidermoid carcinomaKBGI50 = 5.2 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 7.1 µM/72 h
4011,15-di-(3-trifluoromethylcinnamoyl)kobusineLung cancerA549GI50 = 5.5 µM/72 h[55]
Prostate cancerDU145GI50 = 6.2 µM/72 h
Epidermoid carcinomaKBGI50 = 4.1 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 3.1 µM/72 h
4111,15-dibenzoylpseudokobusineLung cancerA549GI50 = 8.8 µM/72 h[55]
Prostate cancerDU145GI50 = 7.6 µM/72 h
Epidermoid carcinomaKBGI50 = 5.2 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 6.3 µM/72 h
4211-(4-nitrobenzoyl)pseudokobusineLung cancerA549GI50 = 5.8 µM/72 h[55]
Prostate cancerDU145GI50 = 7.2 µM/72 h
Epidermoid carcinomaKBGI50 = 6.4 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 6.4 µM/72 h
4311,15-di-(3-nitrobenzoyl)pseudokobusineLung cancerA549GI50 = 5.0 µM/72 h[55]
Prostate cancerDU145GI50 = 5.2 µM/72 h
Epidermoid carcinomaKBGI50 = 5.6 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 5.6 µM/72 h
4411-(3-trifluoromethylbenzoyl)pseudokobusineLung cancerA549GI50 = 6.8 µM/72 h[55]
Prostate cancerDU145GI50 = 7.7 µM/72 h
Epidermoid carcinomaKBGI50 = 8.9 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 6.2 µM/72 h
4511-cinnamoylpseudokobusineLung cancerA549GI50 = 8.4 µM/72 h[55]
Prostate cancerDU145GI50 = 6.5 µM/72 h
Epidermoid carcinomaKBGI50 = 7.0 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 6.4 µM/72 h
4611-tritylpseudokobusineLung cancerA549GI50 = 6.4 µM/72 h[55]
Prostate cancerDU145GI50 = 6.0 µM/72 h
Epidermoid carcinomaKBGI50 = 6.6 µM/72 h
Epidermoid carcinomaKB-VINGI50 = 5.3 µM/72 h
Bis-diterpenoid alkaloids
47Bis-[O-(14-benzoylaconine-8-yl)]-pimelateLung cancerA5499.50 µM/72 h[56]
Breast cancerMCF-77.56 µM/72 h
Colon cancerHCT-154.64 µM/72 h
48Bis-[O-(14-benzoylaconine-8-yl)]-suberateLung cancerA5497.53 µM/72 h[56]
Breast cancerMCF-76.90 µM/72 h
Colon cancerHCT-154.01 µM/72 h
49Bis-[O-(14-benzoylaconine-8-yl)]-azelateLung cancerA54919.5 µM/72 h[56]
Breast cancerMCF-716.9 µM/72 h
Colon cancerHCT-1528.0 µM/72 h

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/molecules22020267

AMA Style

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(2):267. https://doi.org/10.3390/molecules22020267

Chicago/Turabian Style

Ren, Meng-Yue, Qing-Tian Yu, Chun-Yu Shi, and Jia-Bo Luo. 2017. "Anticancer Activities of C18-, C19-, C20-, and Bis-Diterpenoid Alkaloids Derived from Genus Aconitum" Molecules 22, no. 2: 267. https://doi.org/10.3390/molecules22020267

Article Metrics

Back to TopTop