Category Archives: type

Biochanin A

Cancer: Breast

Action: Multi-drug resistance, anti-inflammatory, chemo-preventive

Biochanin is a derivative found in fruits, vegetables, plant-derived beverages, and herbal dietary supplements. It is isolated from a range of plants, including red clover, soy, alfalfa sprouts, and garbanzo beans (Trifolium pratense (L.), Glycine max [(L.) Merr.], Medicago sativa (L.), Cicer arietinum (L.))

MDR; Breast Cancer

Multi-drug resistance (MDR) is one of the most significant obstacles in cancer chemotherapy. One of the mechanisms involved in the development of MDR is the over-expression of P-glycoprotein (P-gp). It is widely known that natural compounds found in vegetables, fruits, plant-derived beverages and herbal dietary supplements not only have anti-cancer properties, but may also modulate P-gp activity. To further elucidate this, the effect of biochanin on P-gp function in human breast cancer cell lines, MCF-7 (sensitive) and MCF-7/ADR (resistant) was therefore examined.

The IC50 value of DNM in the resistant cells was about 22 times higher than that in the sensitive cells, indicating an over-expression of P-gp in the resistant cells, MCF-7/ADR. Biochanin was found to significantly decrease the IC50 value of DNM. Biochanin also showed a significant increase in [3H]-DNM accumulation, increasing by 454.3±19.5% in the resistant cells. Moreover, biochanin significantly decreased DNM efflux from MCF-7/ADR cells compared with the control. These results suggest that biochanin may reverse MDR by inhibiting the P-gp function (Chung et al., 2005).

Chemo-preventive

Biochanin A (BCA), a major isoflavone in red clover and many other legumes, has been reported to display estrogenic as well as cancer chemo-preventive properties. Ingested BCA is known to display low bioavailability due to poor solubility, extensive metabolism and rapid clearance. Esters of bioactive isoflavones are known to increase metabolic stability and bioavailability following local rather than systemic administration (Fokialakis et al., 2012).

Anti-inflammatory

Biochanin inhibits NF-κB activation not only by blocking the upstream IKK, but also PTK that phosphorylate tyrosine residues of IκBα. The double-edged sword effect of inhibition of NF-κB via inhibition of both serine/threonine kinase and PTK by biochanin might show useful therapeutic value against activities of cells that lead to tumorigenesis and inflammation (Manna et al., 2012).

References

Chung SY, Sung MK, Kim NH, et al. (2005). Inhibition of P-glycoprotein by natural products in human breast cancer cells. Archives of Pharmacal Research, 28(7):823-828. doi: 10.1007/BF02977349


Fokialakis N, Alexi X, Aligiannis N, et al. (2012). Ester and carbamate ester derivatives of Biochanin A: synthesis and in vitro evaluation of estrogenic and anti-proliferative activities. Bioorg Med Chem, 20(9):2962-70. doi: 10.1016/j.bmc.2012.03.012.


Manna SK. (2012). Double-edged sword effect of biochanin to inhibit nuclear factor kappaB: suppression of serine/threonine and tyrosine kinases. Biochem Pharmacol, 83(10):1383-92. doi: 10.1016/j.bcp.2012.02.011.

Shikonin

Cancer: Sarcoma-180, lung, melanoma, leukemia

Action: Anti-inflammatory, inhibits angiogenesis, MDR

Shiunko is a Kampo herbal ointment often used for the treatment of burns in Japan. It is mainly isolated from the root of Lithospermum erythrorhizon (Siebold & Zuccarini), which had been used for treating tumors and inflammation in China since the 5th century. The naphthoquinone pigment shikonin is the most important pharmacologically active substance in the dried root of Lithospermum erythrorhizon. In traditional Chinese medicine root extracts of Lithospermum erythrorhizon have been used to treat macular eruption, measles, sore throat, carbuncles, and burns (Chen et al., 2002). The anti-tumor effect of shikonin was first evidenced by its activity against murine sarcoma-180 (Sankawa et al., 1977).

Melanoma

It has been reported that shikonin, the main chemical ingredient of L. erythrorhizon is a novel inhibitor of angiogenesis. Angiogenesis is critical for tumor growth and inflammation. It inhibited tumor necrosis factor-alpha-induced and B16 melanoma-induced angiogenesis in mice and normal developmental angiogenesis in the yolk-sac membranes of chick embryos. Shikonin also inhibited proliferation and migration of endothelial cells in culture and network formation by endothelial cells on Matrigel in vitro. The dose-responsive study suggests that the mechanism of this inhibitory effect on angiogenesis involves the prevention of network formation by endothelial cells via blocking integrin alpha v beta 3 expression (Hisa et al., 1998).

Anti-inflammatory

Shikonin also reported to exert anti-inflammatory and anti-cancer effects both in vitro and in vivo. It has been found that proteasome was a molecular target of shikonin in tumor cells, but whether shikonin targets macrophage proteasome needs to be investigated. Consistently, shikonin accumulated IκB-α, an inhibitor of NF-κB, and ubiquitinated proteins in rat primary macrophage cultures, demonstrating that the proteasome is a target of shikonin under inflammatory conditions.

Shikonin also induced macrophage cell apoptosis and cell death. These results demonstrate for the first time that proteasome inhibition by shikonin contributes to its anti-inflammatory effect. The novel finding about macrophage proteasome as a target of shikonin suggests that this medicinal compound has great potential to be developed into an anti-inflammatory agent (Lu et al., 2011).

Leukemia, MDR

Shikonin has a strong cytotoxic effect on a wide variety of cancer cell lines, especially different types of leukemia and several known MDR cell lines. Microarray-based gene expression analysis of U937 leukemia cells suggested that the cytotoxicity of shikonin is based on the disruption of normal mitochondrial function, overproduction of ROS, inhibition of cytoskeleton formation, and finally induction of cell-cycle arrest and apoptosis. These effects were validated using in vitro cell culture experiments exploiting the specific natural fluorescence of shikonin and thereby identifying the possible primary cellular mechanism of shikonin's cytotoxicity (Wiench et al., 2012).

Lung Cancer

To better understand the anti-metastatic role of shikonin in lung cancer, the effect of shikonin on lung cancer cell proliferation was investigated, as well as its adhesion to extracellular matrices (ECM), migration and invasion in non-small-cell lung cancer A549 cells. Taken together, findings provide new evidence that shikonin suppresses lung cancer invasion and metastasis by inhibiting integrin β1 expression and the ERK1/2 signaling pathway. Integrin β1 facilitates cancer cell adhesion, migration and metastasis by activating intracellular signaling pathways including the ERK and PI3K signaling pathways, and it is in this way that shikonin exerts its anti-cancer activity (Wang et al., 2013).

MDR

Numerous previous studies have proven that shikonin and its analogs not only are highly tumoricidal but also can bypass drug-transporter and apoptotic defect mediated drug resistance. Cancer drug resistance is a major obstacle for the success of chemotherapy. Since most clinical anti-cancer drugs could induce drug resistance, it is desired to develop candidate drugs that are highly efficacious but incompetent to induce drug resistance. Shikonin was investigated for its ability as an inducer of cancer drug resistance. Different cell lines (K562, MCF-7, and a MDR cell line K562/Adr), after repeatedly treated with shikonin for 18 months, were assayed for drug resistance and gene expression profiling. After an 18-month treatment, cells only developed a mere 2-fold resistance to shikonin and a marginal resistance to cisplatin and paclitaxel, without cross-resistance to shikonin analogs and other anti-cancer agents. These merits make shikonin and its analogs potential candidates for cancer therapy with the advantages of avoiding induction of drug resistance and bypassing existing drug resistance (Wu et al., 2013).

References

Chen X, Yang L, Oppenheim JJ, Howard OMZ. (2002). Cellular pharmacology studies of shikonin derivatives. Phytotherapy Research, 16(3):199–209.


Hisa T, Kimura Y, Takada K, Suzuki F, Takigawa M. (1998). Shikonin, an ingredient of Lithospermum erythrorhizon, inhibits angiogenesis in vivo and in vitro. Anti-cancer Res, 18(2A):783-90.


Lu L, Qin A, Huang H, et al. (2011). Shikonin extracted from medicinal Chinese herbs exerts anti-inflammatory effect via proteasome inhibition. Eur J Pharmacol. 658(2–3):242–247.


Sankawa U, Ebizuka Y, Miyazaki T, et al. (1977). Anti-tumor activity of shikonin and its derivatives. Chemical and Pharmaceutical Bulletin, 25(9):2392–2395.


Wang H, Wu C, Wan S, et al. (2013). Shikonin attenuates lung cancer cell adhesion to extracellular matrix and metastasis by inhibiting integrin β 1 expression and the ERK1/2 signaling pathway. Toxicology, 308:104-12. doi: 10.1016/j.tox.2013.03.015. Epub 2013 Apr 4.


Wiench B, Eichhorn T, Malte Paulsen M, Efferth T. (2012). Shikonin Directly Targets Mitochondria and Causes Mitochondrial Dysfunction in Cancer Cells. Evidence-Based Complementary and Alternative Medicine, 2012:726025. doi:10.1155/2012/726025


Wu H, Xie J, Pan Q, et al. (2013). Anti-cancer agent shikonin is an incompetent inducer of cancer drug resistance. PLoS One, 8(1):e52706. doi: 10.1371/journal.pone.0052706.

Costunolide and Dehydrocostus Lactone

Cancers:
Breast, cervical., lung, ovarian, bladder, leukemia, prostate, gastric

Action: Anti-inflammatory, pro-oxidative, MDR, lymphangiogenesis inhibitor, anti-metastasis, mediates apoptosis, anti-metastatic

Components of Saussurea lappa Clarke, Vladimiria souliei (Franchet) Lingelsheim (Compositae)

Breast cancer; Anti-metastatic

It was found that costunolide inhibited the growth and telomerase activity of MCF-7 and MDA-MB-231 cells in a concentration- and time-dependent manner. The expression of hTERT mRNA was also inhibited but hTR mRNA was not. In addition, the bindings of transcription factors in hTERT promoters were significantly decreased in both cells by the treatment of costunolide. These results suggest that costunolide inhibited the growth of both MCF-7 and MDA-MB-231 cells and this effect was mediated at least in part by a significant reduction in telomerase activity (Choi et al., 2005).

Breast Cancer

Costunolide has been demonstrated to suppress tumor growth and metastases of MDA-MB-231 highly metastatic human breast cancer cells via inhibiting TNF-α induced NF-kB activation. Costunolide also inhibited MDA-MB-231 tumor growth and metastases without affecting body weights in the in vivo mouse orthotopic tumor growth assays.

In addition, costunolide inhibited in vitro TNF-α induced invasion and migration of MDA-MB-231 cells. Costunolide further suppressed TNF-α induced NF-kB signaling activation, resulting in a reduced expression of MMP-9, a well-known NF-kB-dependent gene to mediate breast cancer cell growth and metastases. Taken together, these results suggest that SLC and its derivative costunolide suppress breast cancer growth and metastases by inhibiting TNF-α induced NF-k B activation, suggesting that costunolide as well as SLC may be promising anti-cancer drugs, especially for metastatic breast cancer (Choi et al., 2013).

Several Chinese herbs, namely, Herba Taraxaci Mongolici (Pu Gong Ying), Radix Glycyrrhizae Uralensis (Gan Cao), Radix Bupleuri (Chai Hu), Radix Aucklandiae Lappae/ Radix Aucklandiae Lappae (Mu Xiang), Fructus Trichosanthis (Gua Lou) and Rhizoma Dioscoreae Bulbiferae (Huang Yao Zi) are frequently used in complex traditional Chinese medicine formulas for breast hyperplasia and breast tumor therapy.

The pharmacological effects of these Chinese herbs are all described as 'clearing heat-toxin and resolving masses' in traditional use. A bioactivity-oriented screening platform, which was based on a human breast cancer MCF-7 cellular model was developed to rapidly screen the 6 Chinese herbs. Two potential anti-breast cancer compounds, which were costunolide (Cos) and dehydrocostus lactone (Dehy), were identified in Radix Aucklandiae Lappae.

Combination of the two compounds showed a synergism on inhibiting the proliferation of MCF-7 cells in vitro, which exhibits a potential application prospect for breast cancer therapy. This bioactivity-oriented screening strategy is rapid, economical., reliable and specific for screening potential anti-breast cancer compounds in traditional Chinese medicines (Peng et al., 2013).

Dehydrocostuslactone (DHE) suppresses the expression of cyclin D, cyclin A, cyclin-dependent kinase 2, and cdc25A and increases the amount of p53 and p21, resulting in G(0)/G(1)-S phase arrest in MCF-7 cells. In contrast, DHE caused S-G(2)/M arrest by increasing p21 expression and chk1 activation and inhibiting cyclin A, cyclin B, cdc25A, and cdc25C expression in MDA-MB-231 cells. Reduction of SOCS-1 and SOCS-3 expression by small interfering RNA inhibits DHE-mediated signal transducer and activator of transcription-3 inhibition, p21 up-regulation, and cyclin-dependent kinase 2 blockade, supporting the hypothesis that DHE inhibits cell-cycle progression and cell death through SOCS-1 and SOCS-3.

Significantly, animal studies have revealed a 50% reduction in tumor volume after a 45-day treatment period. Taken together, this study provides new insights into the molecular mechanism of the DHE action that may contribute to the chemoprevention of breast cancer (Kuo et al., 2009).

ER- Breast Cancer

Costunolide induced apoptosis through the extrinsic pathway, including the activation of Fas, caspase-8, caspase-3, and degradation of PARP. However, it did not have the same effect on the intrinsic pathway as revealed by analysis of mitochondrial membrane potential (Δψ m) with JC-1 dye and expression of Bcl2 and Bax proteins level.

Furthermore, costunolide induced cell-cycle arrest in the G2/M phase via decrease in Cdc2, cyclin B1 and increase in p21WAF1 expression, independent of p53 pathway in p53-mutant MDA-MB-231 cells, and increases Cdc2-p21WAF1 binding/

Through this study it was confirmed that costunolide induces G2/M cell-cycle arrest and apoptotic cell death via extrinsic pathway in MDA-MB-231 cells, suggesting that it could be a promising anti-cancer drug especially for ER negative breast cancer (Choi et al., 2012).

Bladder Cancer

Costunolide, a member of sesquiterpene lactone family, possesses potent anti-cancer properties. The effects of costunolide were investigated on the cell viability and apoptosis in human bladder cancer T24 cells. Treatment of T24 cells with costunolide resulted in a dose-dependent inhibition of cell viability and induction of apoptosis, which was associated with the generation of ROS and disruption of mitochondrial membrane potential (Δψm).

These effects were significantly blocked when the cells were pre-treated with N-acetyl- cysteine (NAC), a specific ROS inhibitor. Exposure of T24 cells to costunolide was also associated with increased expression of Bax, down-regulation of Bcl-2, and of   survivin and significant activation of caspase-3, and its downstream target PARP. These findings provide the rationale for further in vivo and clinical investigation of costunolide against human bladder cancer (Rasul et al., 2013).

Sarcomas; MDR

Human soft tissue sarcomas represent a rare group of malignant tumors that frequently exhibit chemotherapeutic resistance and increased metastatic potential following unsuccessful treatment.

The effects on cell proliferation, cell-cycle distribution, apoptosis induction, and ABC transporter expression were analyzed. Cells treated with costunolide showed no changes in cell-cycle, little in caspase 3/7 activity, and low levels of cleaved caspase-3 after 24 and 48 hours. Dehydrocostus lactone caused a significant reduction of cells in the G1 phase and an increase of cells in the S and G2/M phase. Moreover, it led to enhanced caspase 3/7 activity, cleaved caspase-3, and cleaved PARP indicating apoptosis induction.

These data demonstrate that dehydrocostus lactone affects cell viability, cell-cycle distribution and ABC transporter expression in soft tissue sarcoma cell lines. Furthermore, it led to caspase 3/7 activity as well as caspase-3 and PARP cleavage, which are indicators of apoptosis. Therefore, this compound may be a promising lead candidate for the development of therapeutic agents against drug-resistant tumors (Kretschmer et al., 2013).

Leukemia, Lung Cancer

Costunolide, an active compound isolated from the stem bark of Magnolia sieboldii, has been found to induce apoptosis via reactive oxygen species (ROS) and Bcl-2-dependent mitochondrial permeability transition in human leukemia cells. Mitogen-activated protein kinases (MAPKs) were investigated for their involvement in the costunolide-induced apoptosis in human promonocytic leukemia U937 cells.

Treatment with costunolide resulted in the significant activation of c-Jun N-terminal kinase (JNK), but not of extracellular-signal-related kinase (ERK1/2) or p38. In vitro kinase assays showed that JNK activity was low in untreated cells but increased dramatically after 30 minutes of costunolide treatment. U937 cells co-treated with costunolide and sorbitol, a JNK activator, exhibited higher levels of cell death. In addition, inhibition of the JNK pathway using a dominant-negative mutation of c-jun and JNK inhibitor SP600125, significantly prevented costunolide-induced apoptosis.

Furthermore, pre-treatment with the anti-oxidant NAC (N-acetyl-L-cysteine) blocked the costunolide-stimulated activation of JNK while the overexpression of Bcl-2 failed to reverse JNK activation. These results indicate that costunolide-induced JNK activation acts downstream of ROS but upstream of Bcl-2, and suggest that ROS-mediated JNK activation plays a key role in costunolide-induced apoptosis. Moreover, the administration of costunolide (intraperitoneally once a day for 7 days) significantly suppressed tumor growth and increased survival in 3LL Lewis lung carcinoma-bearing model (Choi et al., 2009).

Prostate Cancer

Several pharmacological and biochemical assays were used to characterize the apoptotic-signaling pathways of costunolide in prostate cancer cells. Costunolide showed effective anti-proliferative activity against hormone dependent (LNCaP) and independent (PC-3 and DU-145) prostate cancer cells (ATCC¨) by sulforhodamine B assay, clonogenic test and flow cytometric analysis of carboxyfluorescein succinimidyl ester labeling. In PC-3 cells data showed that costunolide induced a rapid overload of nuclear Ca(2+), DNA damage response and ATR phosphorylation.

This indicated the crucial role of intracellular Ca(2+) mobilization and thiol depletion but not of reactive oxygen species production in apoptotic signaling. Data suggest that costunolide induces the depletion of intracellular thiols and overload of nuclear Ca(2+) that cause DNA damage and p21 up-regulation. The association of p21 with the cyclin dependent kinase 2/cyclin E complex blocks cyclin dependent kinase 2 activity and inhibits Rb phosphorylation, leading to G1 arrest of the cell-cycle and subsequent apoptotic cell death in human prostate cancer cells (Hsu et al., 2011).

Gastric Cancer, Prostate Cancer

Radix Aucklandiae Lappae/Saussurea lappa has been used in Chinese traditional medicine for the treatment of abdominal pain, tenesmus, nausea, and cancer; previous studies have shown that S. lappa also induces G(2) growth arrest and apoptosis in gastric cancer cells. The effects of hexane extracts of S. lappa (HESLs) on the migration of DU145 and TRAMP-C2 prostate cancer cells were investigated.

The active compound, dehydrocostus lactone (DHCL), in fraction 7 dose-dependently inhibited the basal and EGF-induced migration of prostate cancer cells. HESL and DHCL reduced matrix metalloproteinase (MMP)-9 and tissue inhibitor of metalloproteinase (TIMP)-1 secretion but increased TIMP-2 levels in both the absence and presence of EGF. These results demonstrate that the inhibition of MMP-9 secretion and the stimulation of TIMP-2 secretion contribute to reduced migration of DU145 cells treated with HESL and DHCL.

This indicates that HESL containing its active principle, DHCL, has potential as an anti-metastatic agent for the treatment of prostate cancer (Kim et al., 2012).

Anti-metastatic

Lymphangiogenesis inhibitors from crude drugs used in Japan and Korea were investigated for their impact on metastasis. The three crude drugs Saussureae Radix, Psoraleae Semen and Aurantti Fructus Immaturus significantly inhibited the proliferation of temperature-sensitive rat lymphatic endothelial (TR-LE) cells in vitro.

Among isolated compounds, several compounds; costunolide, dehydrocostus lactone, psoracorylifol D, bavachinin, bakuchiol, showed an inhibitory effect on the proliferation and the capillary-like tube formation of TR-LE cells. In addition, all compounds showed selective inhibition of the proliferation of TR-LE cells compared to Hela and Lewis lung carcinoma (LLC) cells.

These compounds might offer clinical benefits as lymphangiogenesis inhibitors and may be good candidates for novel anti-cancer and anti-metastatic agents (Jeong et al., 2013).

Ovarian Cancer, MDR

The apoptosis-inducing effect of costunolide, a natural sesquiterpene lactone, was studied in platinum-resistant human ovarian cancer cells relative to cisplatin.

The MTT assay for cell viability, PI staining for cell-cycle profiling, and annexin V assay for apoptosis analysis were performed. Costunolide induced apoptosis of platinum-resistant cells in a time and dose-dependent manner and suppressed tumor growth in the SKOV3 (PT)-bearing mouse model. In addition, costunolide triggered the activation of caspase-3, caspase-8, and caspase-9. Pre-treatment with caspase inhibitors neutralized the pro-apoptotic activity of costunolide. We further demonstrated that costunolide induced a significant increase in intracellular reactive oxygen species (ROS). Moreover, costunolide synergized with cisplatin to induce cell death in platinum-resistant ovarian cancer cells.

Data suggests that costunolide, alone or in combination with cisplatin, may be of therapeutic potential in platinum-resistant ovarian cancers (Yang, Kim, Lee, & Choi, 2011).

Anti-inflammatory, Anti-oxidant, Mediates Apoptosis

Cheon et al. (2013) found that costunolide significantly inhibited RANKL-induced BMM differentiation into osteoclasts in a dose-dependent manner without causing cytotoxicity. Costunolide did not regulate the early signaling pathways of RANKL, including the mitogen-activated protein kinase and NF-κB pathways.

However, costunolide suppressed nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) expression via inhibition of c-Fos transcriptional activity without affecting RANKL-induced c-Fos expression. The inhibitory effects of costunolide were rescued by overexpression of constitutively active (CA)-NFATc1. Taken together, these results suggest that costunolide inhibited RANKL-induced osteoclast differentiation by suppressing RANKL-mediated c-Fos transcriptional activity.

References

Cheon YH, Song MJ, Kim JY, Kwak SC, Park JH, Lee CH, Kim JJ, Kim JY, Choi MK, Oh J, Kim YC, Yoon KH., Kwak HB, Lee MS. (2013). Costunolide inhibits osteoclast differentiation by suppressing c-Fos transcriptional activity. Phytotherapy, July, (6). doi: 10.1002/ptr.5034.

Choi SH, Im E, Kang HK, et al. (2005). Inhibitory effects of costunolide on the telomerase activity in human breast carcinoma cells. Cancer Lett, 227(2):153-62.


Choi JH, Lee KT. (2009). Costunolide-induced apoptosis in human leukemia cells: involvement of c-jun N-terminal kinase activation. Biol Pharm Bull, 32(10):1803-8.


Choi YK, Seo HS, Choi HS, et al. (2012). Induction of Fas-mediated extrinsic apoptosis, p21WAF1-related G2/M cell-cycle arrest and ROS generation by costunolide in estrogen receptor-negative breast cancer cells, MDA-MB-231. Mol Cell Biochem, 363(1-2):119-28. doi: 10.1007/s11010-011-1164-z.


Choi YK, Cho S-G, Woo S-M, et al. (2013). Saussurea lappa Clarke-Derived Costunolide Prevents TNF α-Induced Breast Cancer Cell Migration and Invasion by Inhibiting NF-κ B Activity. Evidence-Based Complementary and Alternative Medicine. doi:10.1155/2013/936257.


Hsu JL, Pan SL, Ho YF, Het al. (2011). Costunolide induces apoptosis through nuclear calcium2+ overload and DNA damage response in human prostate cancer. The Journal of Urology, 185(5):1967-74. doi: 10.1016/j.juro.2010.12.091.


Jeong D, Watari K, Shirouzu T, et al. (2013). Studies on lymphangiogenesis inhibitors from Korean and Japanese crude drugs. Biol Pharm Bull, 36(1):152-7.


Kim EJ, Hong JE, Lim SS, et al. (2012). The hexane extract of Saussurea lappa and its active principle, dehydrocostus lactone, inhibit prostate cancer cell migration. J Med Food, 15(1):24-32. doi: 10.1089/jmf.2011.1735.


Kretschmer N, Rinner B, Stuendl N, et al. (2012). Effect of costunolide and dehydrocostus lactone on cell-cycle, apoptosis, and ABC transporter expression in human soft tissue sarcoma cells. Planta Med, 78(16):1749-56. doi: 10.1055/s-0032-1315385.


Kuo PL, Ni WC, Tsai EM, Hsu YL. (2009). Dehydrocostuslactone disrupts signal transducers and activators of transcription 3 through up-regulation of suppressor of cytokine signaling in breast cancer cells. Mol Cancer Ther, 8(5):1328-39. doi: 10.1158/1535-7163.MCT-08-0914.


Peng ZX, Wang Y, Gu X, Wen YY, Yan C. (2013). A platform for fast screening potential anti-breast cancer compounds in traditional Chinese medicines. Biomed Chromatogr. doi: 10.1002/bmc.2990.


Rasul A, Bao R, Malhi M, et al. (2013). Induction of apoptosis by costunolide in bladder cancer cells is mediated through ROS generation and mitochondrial dysfunction. Molecules, 18(2):1418-33. doi: 10.3390/molecules18021418.


Yang YI, Kim JH, Lee KT, & Choi JH. (2011). Costunolide induces apoptosis in platinum-resistant human ovarian cancer cells by generating reactive oxygen species. Gynecologic Oncology, 123(3), 588-96. doi: 10.1016/j.ygyno.2011.08.031.

Isorhamnetin

Cancer:
Lung, colon, acute myeloid leukemia, T lymphoma, Ehrlich carcinoma, gastric, esophageal squamous cell, chronic myelogenous leukemia

Action: Dox-induced cardiotoxicity, anti-oxidant

Isorhamnetin, the anti-tumor component of Hippophae rhamnoides Linn, is also a member of the ßavonoid class of compounds. Its chemical name is 3,5,7-trihydroxy-2-(4-hydroxy-3-methoxyphenyl) chromen-4-one and its molecular formula is C16H12O7.

Lung Cancer

Isorhamnetin shows good inhibitory effects on human lung adenocarcinoma A549 cells, human colon cancer HT-29 cells, human chronic myeloid leukemia K562 cells, human acute myeloid leukemia HL-60 cells, mouse T lymphoma YAC-1 cells and mouse Ehrlich carcinoma. In terms of its mechanism of action, it seems that isorhamnetin simultaneously reduces the expression of Bcl-2 and increases the expression of Bax, which activates caspase-9 and its downstream factor caspase-3, thus resulting in cell death (Zhu et al. 2005).

Colorectal Cancer

It was demonstrated that isorhamnetin prevents colorectal tumorigenesis. Dietary isorhamnetin decreased mortality, tumor number, and tumor burden by 62%, 35%, and 59%, respectively. Magnetic resonance imaging, histopathology, and immunohistochemical analysis revealed that dietary isorhamnetin resolved the DSS-induced inflammatory response faster than control diet.

These observations suggest the chemo-protective effects of isorhamnetin in colon cancer are linked to its anti-inflammatory activities and its inhibition of oncogenic Src activity and consequential loss of nuclear β-catenin, activities that are dependent on CSK expression (Saud et al., 2013).

Gastric Cancer

The potential effects of isorhamnetin (IH), a 3'-O-methylated metabolite of quercetin, were investigated on the peroxisome proliferator-activated receptor γ (PPAR-γ) signaling cascade using proteomics technology platform, gastric cancer (GC) cell lines, and xenograft mice model.

It was observed that IH exerted a strong anti-proliferative effect and increased cytotoxicity in combination with chemotherapeutic drugs. IH also inhibited the migratory/invasive properties of gastric cancer cells, which could be reversed in the presence of PPAR-γ inhibitor.

Using molecular docking analysis, Ramachandran et al. (2013) demonstratd that IH formed interactions with seven polar residues and six nonpolar residues within the ligand-binding pocket of PPAR-γ that are reported to be critical for its activity and could competitively bind to PPAR-γ. IH significantly increased the expression of PPAR-γ in tumor tissues obtained from xenograft model of GC. Overall, these findings clearly indicate that anti-tumor effects of IH may be mediated through modulation of the PPAR-γ activation pathway in GC.

Cardiac-protective; Doxorubicin

Isorhamnetin is a natural anti-oxidant with obvious cardiac-protective effect. Its action against doxorubicin-induced cardotoxicity and underlying mechanisms were investigated. Doxorubicin (Dox) is an anthracycline antibiotic for cancer therapy with limited usage due to cardiotoxicity. The aim of this study is to investigate the possible protective effect of isorhamnetin against Dox-induced cardiotoxicity and its underlying mechanisms. In an in vivo investigation, rats were intraperitoneally (i.p.) administered with Dox to duplicate the model of Dox-induced chronic cardiotoxicity.

Daily pre-treatment with isorhamnetin (5 mg/kg, i.p.) for 7 days was found to reduce Dox-induced myocardial damage significantly, including the decline of cardiac index, decrease in the release of serum cardiac enzymes, and amelioration of heart vacuolation. In vitro studies on H9c2 cardiomyocytes, isorhamnetin was effective to reduce Dox-induced cell toxicity. Isorhamnetin also potentiated the anti-cancer activity of Dox in MCF-7, HepG2 and Hep2 cells. These findings indicated that isorhamnetin can be used as an adjuvant therapy for the long-term clinical use of Dox (Sun et al., 2013).

Chronic Myelogenous Leukemia

The isorhamnetin 3-o-robinobioside and its original extract, ethyl acetate extract, from Nitraria retusa leaves, were evaluated for their ability to induce anti-oxidant and anti-genotoxic effects in human chronic myelogenous leukemia cell line. They were shown to have a great anti-oxidant and anti-genotoxic potential on human chronic myelogenous leukemia cell line K562 (Boubaker et al., 2012).

Esophageal Cancer

The flavonol aglycone isorhamnetin shows anti-proliferative activity in a variety of cancer cells and it inhibits the proliferation of human esophageal squamous carcinoma Eca-109 cells in vitro (Shi et al., 2012).

Cancer:
Actions: Overcomes MDR; P-glycoproteins, breast cancer resistance proteins (BCRP), efflux transporters

Flavonoid isorhamnetin occurs in various plants and herbs, and demonstrates various biological effects in humans. This work will clarify the isorhamnetin absorption mechanism using the Caco-2 monolayer cell model. The isorhamnetin transport characteristics at different concentrations, pHs, temperatures, tight junctions and potential transporters were systemically investigated.

Isorhamnetin was poorly absorbed by both passive diffusion and active transport mechanisms. Both trans- and paracellular pathways were involved during isorhamnetin transport. Active transport under an ATP-dependent transport mechanism was mediated by the organic anion transporting peptide (OATP); isorhamnetin’s permeability from the apical to the basolateral side significantly decreased after estrone-3-sulfate was added (p<0.01).

Efflux transporters, P-glycoproteins (P-gp), breast cancer resistance proteins (BCRP) and multidrug resistance proteins (MRPs) participated in the isorhamnetin transport process. Among them, the MRPs (especially MRP2) were the main efflux transporters for isorhamnetin; transport from the apical to the basolateral side increased 10.8-fold after adding an MRP inhibitor (MK571).

References

Boubaker J, Ben Sghaier M, Skandrani I, et al. (2012). Isorhamnetin 3-O-robinobioside from Nitraria retusa leaves enhance anti-oxidant and anti-genotoxic activity in human chronic myelogenous leukemia cell line K562. BMC Complement Altern Med, 12:135. doi: 10.1186/1472-6882-12-135.


Ramachandran L, Manu KA, Shanmugam MK, et al. (2013). Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. J Biol Chem, 288(26):18777. doi: 10.1074/jbc.A112.388702.


Saud SM, Young MR, Jones-Hall YL, et al. (2013). Chemo-preventive activity of plant flavonoid isorhamnetin in colorectal cancer is mediated by oncogenic Src and β -catenin. Cancer Res, 73:5473.


Shi C, Fan LY, Cai Z, Liu YY, Yang CL. (2012). Cellular stress response in Eca-109 cells inhibits apoptosis during early exposure to isorhamnetin. Neoplasma, 59(4):361-9. doi: 10.4149/neo_2012_047.


Sun J, Sun G, Meng X, et al. (2013). Isorhamnetin protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS One, 8(5):e64526. doi: 10.1371/journal.pone.0064526.


Zhu L, Wang ZR, Zhou LM, et al. (2005). Effects and mechanisms of isorhamnetin on lung carcinoma. Space Med Med Eng (Chin), 18:381-383.


Duan J, Xie Y, Luo H, Li G, Wu T, Zhang T. (2014) Transport characteristics of isorhamnetin across intestinal Caco-2 cell monolayers and the effects of transporters on it. Food Chem Toxicol. 2014 Apr;66:313-20. doi: 10.1016/j.fct.2014.02.003.

Daidzein & S-(-)-equol

Cancer: Breast, prostate

Action: Enhances tamoxifen, radio-sensitizer

Daidzein & Genistein Concentrations; Breast Cancer

A systematic search by Mário L de Lemos (2001) through primary English-language literature on MEDLINE (1966–January 2001), EMBASE (1982–January 2001) and Current Contents (1998–January 2001) found that genistein and daidzein at low concentrations were found to stimulate breast tumor growth in in vitro and in vivo animal studies, and antagonize the anti-tumor effect of tamoxifen in vitro. However, at high concentrations, genistein inhibited tumor growth and enhanced the effect of tamoxifen in vitro.

At concentrations below 10 µmol/L, phytoestrogens can stimulate breast tumor growth and antagonize the anti-tumor effects of tamoxifen, particularly in an environment of low endogenous estrogen. In contrast, phytoestrogens inhibit breast tumor growth and enhance the anti-tumor effects of tamoxifen at concentrations above 10 µmol/L (de Lemos, 2002; Akaza et al., 2004).

Breast cancer

Daidzein or its major metabolite equol at a dose molar equivalent to tamoxifen [1.0 mg(2.7 µmol)/kg or 10 mg (27 µmol)/kg/day] was treated orally to rats bearing 7,12-dimethylbenz(a)anthracene(DMBA)-induced mammary tumors or ovariectomized athymic nude mice implanted with human MCF-7 breast cancer xenograft and an estrogen pellet. The growth of tumors was monitored for several weeks after the treatment. The cell-cycle and apoptotic stages in mammary tumors collected from rats were analyzed by flow cytometry. Immunohistochemistry analysis was also used to determine the expression of caspase-3.

Oral treatment with daidzein or equol at a human equivalent dose suppressed the growth of both DMBA-induced mammary tumors and human MCF-7 breast cancer xenografts in rodents, the inhibitory activity being superior to that of genistein or tamoxifen. Strong apoptosis induced by daidzein or equol contributes to the anti-tumor potential.

Daidzein and its metabolite equol showed the potential of inhibiting the growth of mammary tumors in rodents. Daidzein or equol could be used as a core structure to design new drugs for breast cancer therapy. Our results indicate that consumption of daidzein may protect against breast cancer (Liu et al., 2012).

Equol Production

S-(-)-equol is a metabolite of the soy isoflavone daidzein and is produced by intestinal bacteria in some, but not in all, humans after soy consumption (Setchell & Clerici, 2010). The ability of S-equol to play a role in the treatment of estrogen or androgen-mediated diseases or disorders was first proposed in 1984 (Setchell et al., 1984). The ability to do so depends on two things: soy and bacteria. First, the soy must contain the soy isoflavone daidzein and the amount may influence equol production. Second, the human must have certain strains of bacteria living within the intestine. Twenty-one different strains of intestinal bacteria cultured from humans have the ability to transform daidzein into S-equol or a related intermediate compound (Setchell & Clerici, 2010).

Several studies indicate that only 25 to 30 percent of the adult population of Western countries produces S-equol after eating soy foods containing isoflavones, (Rowland et al., 2000; Atkinson et al., 2005) significantly lower than the reported 50–60% frequency of equol-producers in adults from Japan, Korea, or China (Song et al., 2006).

Equol Production; Prostate Cancer

Akaza et al. (2004) recently conducted a case-controlled study on those who are able to degrade daidzein, a soybean isoflavone, to equol and those without this ability. The incidence of prostate cancer is known to be lower in residents in Japan. On the other hand, American residents in the United States have a markedly higher incidence of prostate cancer.

The number of subjects was 295 in Japan (133 patients and 162 controls), 122 in Korea (61 patients and 61 controls) and 45 in the United States (24 patients and 21 controls). The percentage of equol producers among patients and controls was 29% and 46% in Japan (P = 0.004) and 30% and 59% in Korea (P = 0.001), respectively. The active isoflavone level was markedly lower and the percentage of equol producers was also lower (17% for patients and 14% for controls) for Americans as compared to the Japanese and Koreans.

These results suggest that the ability to produce equol, or equol itself, is closely related to the lower incidence of prostate cancer. The results also suggest that a diet based on soybean isoflavones will be useful in preventing prostate cancer.

Prostate cancer

The age-adjusted incidence rate of prostate cancer (PCa) has been reported to be lower among Asians than Western populations. A traditional Japanese meal., high in soybean products or isoflavones, may be associated with a decreased risk of PCa. Equol, which is converted from daidzein by human intestinal flora, is biologically more active than any other isoflavone aglycone.

Five out of 6 articles showed significant association of isoflavones with a decreased risk of PCa, and two of them consistently showed that equol-producers carry a significantly reduced risk of PCa. Furthermore, 5 human intestinal bacteria that can convert daidzein into equol were identified in the last 5 years. If equol can reduce risk of PCa, a possible strategy for reducing the risk of PCa may be to increase the proportion of equol-producers by changing the intestinal flora to carry an equol-producing bacterium, with dietary alteration or probiotic technology (Sugiyama et al., 2013).

Equol Enhances Tamoxifen

Charalambous, Pitta, & Constantinou (2013) found that equol (>50  µM) and 4-hydroxy-tamoxifen (4-OHT; >100 nM) significantly reduced the breast cancer MCF-7 cell viability. Furthermore, the combination of equol (100 µM) and 4-OHT (10 µM) induced apoptosis more effectively than each compound alone. Subsequent treatment of MCF-7 cells with the pan-caspase inhibitor Z-VAD-FMK inhibited equol- and 4-OHT-mediated apoptosis, which was accompanied by PARP and α-fodrin cleavage, indicating that apoptosis is mainly caspase-mediated.

Equol may be used therapeutically in combination treatments and clinical studies to enhance tamoxifen's effect by providing additional protection against estrogen-responsive breast cancers.

Radio-sensitizer

Sensitivity of cells to equol, radiation and a combination of both was determined by colonogenic assays. Induction of apoptosis by equol, radiation and the combination of both was also determined by acridine orange/ethidium bromide double staining fluorescence microscopy. DNA strand breaks were assessed by Comet assay.

MTT assay showed that equol (0.1-350 µM) inhibited MDA-MB-231 and T47D cell growth in a time- and dose-dependent manner. Treatment of cells with equol for 72 hours (MDA-MB-231) and 24 hours (T47D) was found to inhibit cell growth with IC50 values of 252 µM and 228 µM, respectively. Furthermore, pre-treatment of cells with 50 µM equol for 72 hours (MDA-MB-231) and 24 hours (T47D) sensitized the cells to irradiation. Equol was also found to enhance radiation-induced apoptosis. Comet assay results showed that the radio-sensitizing effect of equol was accompanied by increased radiation-induced DNA damage.

These results suggest for the first time that equol can be considered as a radio-sensitizing agent and its effects may be due to increasing cell death following irradiation, increasing the remaining radiation-induced DNA damage and thus reducing the surviving fraction of irradiated cells (Taghizadeh et al., 2013).

References

Akaza H, Miyanaga N, Takashima N, Naito S, et al. (2004). Comparisons of percent equol producers between prostate cancer patients and controls: case-controlled studies of isoflavones in Japanese, Korean and American residents. Japanese Journal of Clinical Oncology, 34(2): 86–9.


Atkinson, C., Frankenfeld, C.L., Lampe, J.W. (2005). Gut bacterial metabolism of the soy isoflavone daidzein: exploring the relevance to human health. Experimental Biology and Medicine (Maywood, N.J.), 230(3):155–70.


Charalambous C, Pitta CA, Constantinou AI. (2013). Equol enhances tamoxifen's anti-tumor activity by induction of caspase-mediated apoptosis in MCF-7 breast cancer cells. BMC Cancer, 13:238. doi: 10.1186/1471-2407-13-238.


de Lemos ML. (2001). Effects of soy phytoestrogens genistein and daidzein on breast cancer growth. Ann Pharmacother, 35(9):1118-21.


de Lemos ML. (2002). Safety Issues of Soy Phytoestrogens in Breast Cancer Patients. JCO, 20(13):3040-3042.


Liu X, Suzuki N, Santosh Laxmi YR, Okamoto Y, Shibutani S. (2012). Anti-breast cancer potential of daidzein in rodents. Life Sci, 91(11-12):415-9.


Rowland IR, Wiseman H, Sanders TA, Adlercreutz H, Bowey EA. (2000). Interindividual variation in metabolism of soy isoflavones and lignans: influence of habitual diet on equol production by the gut microflora. Nutrition and Cancer, 36(1):27–32. doi:10.1207/S15327914NC3601_5.


Setchell KD, Borriello SP, Hulme P, Kirk DN, Axelson M. (1984). Nonsteroidal estrogens of dietary origin: possible roles in hormone-dependent disease. The American Journal of Clinical Nutrition, 40(3):569–78.


Setchell KD, Clerici C. (2010). Equol: history, chemistry, and formation. The Journal of Nutrition, 140 (7): 1355S–62S. doi:10.3945/jn.109.119776.


Song KB, Atkinson C, Frankenfeld CL, Jokela T, et al. (2006). Prevalence of daidzein-metabolizing phenotypes differs between Caucasian and Korean American women and girls. The Journal of Nutrition, 136(5):1347–51.


Sugiyama Y, Masumori N, Fukuta F, et al. (2013). Influence of isoflavone intake and equol-producing intestinal flora on prostate cancer risk. Asian Pac J Cancer Prev, 14(1):1-4.


Taghizadeh B, Ghavami L, Nikoofar A, Goliaei B. (2013). Equol as a potent radiosensitizer in estrogen receptor-positive and -negative human breast cancer cell lines. Breast Cancer.

Tanshinone II A & Tanshinone A (See also Cryptotanshinone)

Cancer:
Leukemia, prostate, breast, gastric, colorectal, nasopharyngeal carcinoma

Action: Chemo-sensitizer, cytostatic, cancer stem cells, anti-cancer, autophagic cell death, cell-cycle arrest

Anti-cancer

Tanshinone IIA and cryptotanshinone could induce CYP3A4 activity (Qiu et al., 2103).

Tanshinone II-A (Tan IIA) is the most abundant diterpene quinone isolated from Danshen (Salvia miltiorrhiza), which has been used in treating cardiovascular diseases for more than 2,000 years in China. Interest in its versatile protective effects in cardiovascular, metabolic, neurodegenerative diseases, and cancers has been growing over the last decade.

Tan IIA is a multi-target drug, whose molecular targets include transcription factors, scavenger receptors, ion channels, kinases, pro- and anti-apoptotic proteins, growth factors, inflammatory mediators, microRNA, and others. More recently, enhanced or synergistic effects can be observed when Tan IIA is used in combination therapy with cardio-protective and anti-cancer drugs (Xu & Liu, 2013).

Leukemia

The in vitro anti-proliferation and apoptosis-inducing effects of Tanshinone IIA on leukemia THP-1 cell lines and its mechanisms of action were investigated. MTT assay was used to detect the cell growth-inhibitory rate; cell apoptotic rate and the mitochondrial membrane potential (Deltapsim) were investigated by flow cytometry (FCM); apoptotic morphology was observed by Hoechst 33258 staining and DNA fragmentation analysis.

It was therefore concluded that Tanshinone IIA has significant growth inhibition effects on THP-1 cells by induction of apoptosis, and that Tanshinone IIA-induced apoptosis on THP-1 cells is mainly related to the disruption of Deltapsim and activation of caspase-3 as well as down-regulation of anti-apoptotic protein Bcl-2, survivin and up-regulation of pro-apoptotic protein Bax. The results indicate that Tanshinone IIA may serve as a potential anti-leukemia agent (Liu et al., 2009).

Prostate Cancer

Chiu et al. (2013) explored the mechanisms of cell death induced by Tan-IIA treatment in prostate cancer cells in vitro and in vivo. Results showed that Tan-IIA caused prostate cancer cell death in a dose-dependent manner, and cell-cycle arrest at G0/G1 phase was noted, in LNCaP cells. The G0/G1 phase arrest correlated with increased levels of CDK inhibitors (p16, p21 and p27) and decrease of the checkpoint proteins. Tan-IIA also induced ER stress in prostate cancer cells: activation and nuclear translocation of GADD153/CCAAT/enhancer-binding protein-homologous protein (CHOP) were identified, and increased expression of the downstream molecules GRP78/BiP, inositol-requiring protein-1α and GADD153/CHOP were evidenced. Blockage of GADD153/CHOP expression by siRNA reduced Tan-IIA-induced cell death in LNCaP cells.

Gastric Cancer

Tan IIA can reverse the malignant phenotype of SGC7901 gastric cancer cells, indicating that it may be a promising therapeutic agent.

Tan IIA (1, 5, 10 µg/ml) exerted powerful inhibitory effects on cell proliferation (P < 0.05, and P < 0.01), and this effect was time- and dose-dependent. FCM results showed that Tan IIA induced apoptosis of SGC7901 cells, reduced the number of cells in S phase and increased those in G0/G1 phase. Tan IIA also significantly increased the sensitivity of SGC7901 gastric cancer cells to ADR and Fu. Moreover, wound-healing and transwell assays showed that Tan IIA markedly decreased migratory and invasive abilities of SGC7901 cells (Xu et al., 2013).

Cell-cycle Arrest

MTT and SRB assays were applied to measure the effects of tanshinone A on cell viability. Cell-cycle distribution and apoptosis were assessed via flow cytometry using PI staining and the Annexin V/PI double staining method respectively. Changes to mitochondrial membrane potential was also detected by flow cytometry. The spectrophotometric method was utilized to detect changes of caspase-3 activity. Western blotting assay was used to evaluate the expression of Bcl-2, Bax and c-Myc proteins.

Results indicated that Tan-IIA displayed significant inhibitory effect on the growth of K562 cells in a dose- and time- dependent manner, and displayed only minimal damage to hepatic LO2 cells.

Tan-IIA could arrest K562 cells in the G0/G1 phase and induce apoptosis, decrease mitochondrial transmembrane potential, and the expressions of Bcl-2 and c-Myc proteins, increase the expression of Bax protein and activity of caspase-3. Accordingly, it was presumed that the induction of apoptosis may be through the endogenous pathway. Subsequently, tanshinone A could be a promising candidate in the development of a novel anti-tumor agent (Zhen et al., 2011).

Prostate Cancer, Chemo-sensitizer

Treatment with a combination of Chinese herbs and cytotoxic chemotherapies has shown a higher survival rate in clinical trials.

Tan-IIA displayed synergistic anti-tumor effects on human prostate cancer PC3 cells and LNCaP cells, when combined with cisplatin in vitro. Anti-proliferative effects were detected via MTT assay. Cell-cycle distribution and apoptosis were detected by flow cytometer. Protein expression was detected by Western blotting. The intracellular concentration of cisplatin was detected by high performance liquid chromatography (HPLC).

Results demonstrated that tanshinone II A significantly enhanced the anti-proliferative effects of cisplatin on human prostate cancer PC3 cells and LNCaP cells with an increase in the intracellular concentration of cisplatin. These effects were correlated with cell-cycle arrest at the S phase and induction of cell apoptosis. Apoptosis could potentially be achieved through the death receptor and mitochondrial pathways, decreased expression of Bcl-2.

Collectively, results indicated that the combination of tanshinone II A and cisplatin had a better treatment effect, in vitro, not only on androgen-dependent LNCaP cells but also on androgen-independent PC3 cells (Hou, Xu, Hu, & Xie, 2013).

Autophagic Cell Death, CSCs

Tan IIA significantly increased the expression of microtubule-associated protein light chain 3 (LC3) II as a hallmark of autophagy in Western blotting and immunofluorescence staining. Tan IIA augmented the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and attenuated the phosphorylation of mammalian target of rapamycin (mTOR) and p70 S6K in a dose-dependent manner.Tan IIA dramatically activated the extracellular signal regulated kinase (ERK) signaling pathway including Raf, ERK and p90 RSK in a dose-dependent and time-dependent manner. Consistently, ERK inhibitor PD184352 suppressed LC3-II activation induced by Tan IIA, whereas PD184352 and PD98059 did not affect poly (ADP-ribose) polymerase cleavage and sub-G1 accumulation induced by Tan IIA in KBM-5 leukemia cells.

Tan IIA induces autophagic cell death via activation of AMPK and ERK and inhibition of mTOR and p70 S6K in KBM-5 cells as a potent natural compound for leukemia treatment (Yun et al., 2013).

Cancer stem cells (CSCs) are maintained by inflammatory cytokines and signaling pathways. Tanshinone IIA (Tan-IIA) possesses anti-cancer and anti-inflammatory activities. The purpose of this study is to confirm the growth inhibition effect of Tan-IIA on human breast CSCs growth in vitro and in vivo and to explore the possible mechanism of its activity. After Tan-IIA treatment, cell proliferation and mammosphere formation of CSCs were decreased significantly; the expression levels of IL-6, STAT3, phospho-STAT3 (Tyr705), NF-κBp65 in nucleus and cyclin D1 proteins were decreased significantly; the tumor growth and mean tumor weight were reduced significantly.

Tan-IIA has the potential to target and kill CSCs, and can inhibit human breast CSCs growth both in vitro and in vivo through attenuation of IL-6/STAT3/NF-kB signaling pathways (Lin et al., 2013).

Colorectal Cancer

Tan II-A can effectively inhibit tumor growth and angiogenesis of human colorectal cancer via inhibiting the expression level of COX-2 and VEGF. Angiogenesis plays a significant role in colorectal cancer (CRC) and cyclooxygenase-2 (COX-2) appears to be involved with multiple aspects of CRC angiogenesis (Zhou et al., 2012). The results showed that Tan IIA inhibited the proliferation of inflammation-related colon cancer cells HCT116 and HT-29 by decreasing the production of inflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), which are generated by macrophage RAW264.7 cell line.

Treatment with TanshinoneIIA prevented increased PU.1, a transcriptional activator of miR-155, and hence increased miR-155, whereas aspirin could not. These findings support that the interruption of signal conduction between activated macrophages and colon cancer cells could be considered as a new therapeutic strategy and miR-155 could be a potential target for the prevention of inflammation-related cancer (Tu et al., 2012).

Breast Cancer

The proliferation rate of T47D and MDA-MB-231 cells influenced by 1×10-6 mol·L-1 and 1×10-7 mol·L-1 Tanshinone IIA was analyzed by MTT assay. Estrogen receptor antagonist ICI182, 780 was employed as a tool. Level of ERα and ERβ mRNA in T47D cells was quantified by Real-time RT-PCR assay. Expression of ERα and ERβ protein was measured by flow cytometry. The proliferation rates of T47D cells treated with Tanshinone IIA decreased significantly. Such effects could be partly blocked by ICI182, 780.

Meanwhile, the proliferation rates of MDA-MB-231 cells treated with Tanshinone IIA decreased much more dramatically. Real-time RT-PCR and flow cytometry results showed that Tanshinone IIA could induce elevation of ERα and ERβ, especially ERα mRNA, and protein expression level in T47D cells. Tanshinone IIA shows inhibitory effects on proliferation of breast cancer cell lines (Zhao et al., 2010).

The role of cell adhesion molecules in the process of inflammation has been studied extensively, and these molecules are critical components of carcinogenesis and cancer metastasis. This study investigated the effect of tanshinone I on cancer growth, invasion and angiogenesis on human breast cancer cells MDA-MB-231, both in vitro and in vivo. Tanshinone I dose-dependently inhibited ICAM-1 and VCAM-1 expressions in human umbilical vein endothelial cells (HUVECs) that were stimulated with TNF-α for 6 h.

Additionally, reduction of tumor mass volume and decrease of metastasis incidents by tanshinone I were observed in vivo. In conclusion, this study provides a potential mechanism for the anti-cancer effect of tanshinone I on breast cancer cells, suggesting that tanshinone I may serve as an effective drug for the treatment of breast cancer (Nizamutdinova et al., 2008).

Nasopharyngeal Carcinoma

To investigate anti-cancer effect and potential mechanism of tanshinone II(A) (Tan II(A)) on human nasopharyngeal carcinoma cell line CNE cells, the anti-proliferative effect of Tan II(A) on CNE cells was evaluated by morphological examination, cell growth curves, colonial assay and MTT assay. Tan II(A) could inhibit CNE cell proliferation in dose- and time-dependent manner. After treatment with Tan II(A), intracellular Ca2+ concentration of CNE cells was increased, mitochondria membrane potential of the cells was decreased, relative mRNA level of Bad and MT-1A was up-regulated. Tan II(A) had an anti-cancer effect on CNE cells through apoptosis via a calcineurin-dependent pathway and MT-1A down-regulation, and may be the next generation of chemotherapy (Dai et al., 2011).

References

Chiu SC, Huang SY, Chen SP, et al. (2013). Tanshinone IIA inhibits human prostate cancer cells growth by induction of endoplasmic reticulum stress in vitro and in vivo. Prostate Cancer Prostatic Dis. doi: 10.1038/pcan.2013.38.


Dai Z, Huang D, Shi J, Yu L, Wu Q, Xu Q. (2011). Apoptosis inducing effect of tanshinone II(A) on human nasopharyngeal carcinoma CNE cells. Zhongguo Zhong Yao Za Zhi, 36(15):2129-33.


Hou LL, Xu QJ, Hu GQ, Xie SQ. (2013). Synergistic anti-tumor effects of tanshinone II A in combination with cisplatin via apoptosis in the prostate cancer cells. Acta Pharmaceutica Sinica, 48(5), 675-679.


Lin C, Wang L, Wang H, et al. (2013). Tanshinone IIA inhibits breast cancer stem cells growth in vitro and in vivo through attenuation of IL-6/STAT3/NF-kB signaling pathways. J Cell Biochem, 114(9):2061-70. doi: 10.1002/jcb.24553.


Liu JJ, Zhang Y, Lin DJ, Xiao RZ. (2009). Tanshinone IIA inhibits leukemia THP-1 cell growth by induction of apoptosis. Oncol Rep, 21(4):1075-81.


Nizamutdinova IT, Lee GW, Lee JS, et al. (2008). Tanshinone I suppresses growth and invasion of human breast cancer cells, MDA-MB-231, through regulation of adhesion molecules. Carcinogenesis, 29(10):1885-1892. doi:10.1093/carcin/bgn151


Qiu F, Jiang J, Ma Ym, et al. (2013). Opposite Effects of Single-Dose and Multidose Administration of the Ethanol Extract of Danshen on CYP3A in Healthy Volunteers. Evidence-Based Complementary and Alternative Medicine, 2013(2013) http://dx.doi.org/10.1155/2013/730734


Tu J, Xing Y, Guo Y, et al. (2012). TanshinoneIIA ameliorates inflammatory microenvironment of colon cancer cells via repression of microRNA-155. Int Immunopharmacol, 14(4):353-61. doi: 10.1016/j.intimp.2012.08.015.


Xu M, Cao FL, Li NY, et al. (2013). Tanshinone IIA reverses the malignant phenotype of SGC7901 gastric cancer cells. Asian Pac J Cancer Prev, 14(1):173-7.


Xu S, Liu P. (2013). Tanshinone II-A: new perspectives for old remedies. Expert Opin Ther Pat, 23(2):149-53. doi: 10.1517/13543776.2013.743995.


Yun SM, Jung JH, Jeong SJ, et al. (2013). Tanshinone IIA Induces Autophagic Cell Death via Activation of AMPK and ERK and Inhibition of mTOR and p70 S6K in KBM-5 Leukemia Cells. Phytother Res. doi: 10.1002/ptr.5015.


Zhen X, Cen J, Li YM, Yan F, Guan T, Tang, XZ. (2011). Cytotoxic effect and apoptotic mechanism of tanshinone A, a novel tanshinone derivative, on human erythroleukemic K562 cells. European Journal of Pharmacology, 667(1-3), 129-135. doi: 10.1016/j.ejphar.2011.06.004.


Zhao PW, Niu JZ, Wang JF, Hao QX, Yu J, et al. (2010). Research on the inhibitory effect of Tanshinone IIA on breast cancer cell proliferation. Zhong Guo Yao Li Xue Tong Bao, 26(7):903-906.


Zhou LH, Hu Q, Sui H, et al. (2012). Tanshinone II–a inhibits angiogenesis through down regulation of COX-2 in human colorectal cancer. Asian Pac J Cancer Prev, 13(9):4453-8.

Luteolin

Cancer: Colorectal., ovarian, pancreatic

Action: Anti-inflammatory, immunomodulatory, radio-sensitizer, chemo-sensitizer

Luteolin is a flavonoid found in many plants and foods, including Terminalia chebula (Retz.), Prunella vulgaris (L.) and Perilla frutescens [(L.) Britton].

Luteolin is contained in Ocimum sanctum L . or Ocimum tenuiflorum L , commonly known as Holy Basil in English or Tulsi in various Indian languages, which is an important medicinal plant in the various traditional and folk systems of medicine in Southeast Asia. Scientific studies have shown it to possess anti-inflammatory, analgesic, anti-pyretic, anti-diabetic, hepato-protective, hypolipidemic, anti-stress, and immunomodulatory activities. It has been found to prevent chemical-induced skin, liver, oral., and lung cancers and mediates these effects by increasing the anti-oxidant activity, altering the gene expressions, inducing apoptosis, and inhibiting angiogenesis and metastasis.

Colon Cancer

Luteolin inhibited cyclin-dependent kinase (CDK)4 and CDK2 activity, resulting in G1 arrest with a concomitant decrease of phosphorylation of retinoblastoma protein. Activities of CDK4 and CDK2 decreased within 2 hours after luteolin treatment, with a 38% decrease in CDK2 activity (P < 0.05) observed in cells treated with 40 µmol/l luteolin. Luteolin also promoted G2/M arrest at 24 hours post-treatment by down-regulating cyclin B1 expression and inhibiting cell division cycle (CDC)2 activity. Luteolin promoted apoptosis with increased activation of caspases 3, 7, and 9 and enhanced poly(ADP-ribose) polymerase cleavage and decreased expression of p21CIP1/WAF1, survivin, Mcl-1, Bcl-xL, and Mdm-2. Lim et al. (2007) demonstrated that luteolin promotes both cell-cycle arrest and apoptosis in the HT-29 colon cancer cell line, providing insight about the mechanisms underlying its anti-tumorigenic activities.

Radio-protective

The aqueous extract of Perilla frutescens has been shown to protect mice against γ-radiation-induced sickness and mortality and to selectively protect the normal tissues against the tumoricidal effects of radiation. The chemo-preventive and radio-protective properties of Perilla emphasize aspects that warrant future research to establish its activity and utility in cancer prevention and treatment (Baliga et al., 2013).

Anti-inflammatory

Pre-treatment of RAW 264.7 macrophages with luteolin, luteolin-7-glucoside, quercetin, and the isoflavonoid genistein inhibited both the LPS-stimulated TNF-α and interleukin-6 release, whereas eriodictyol and hesperetin only inhibited TNF-α release. From the compounds tested, luteolin and quercetin were the most potent in inhibiting cytokine production with an IC50 of less than 1 and 5 µM for TNF-α release, respectively. Moreover, luteolin inhibited LPS-induced phosphorylation of Akt. Treatment of macrophages with LPS resulted in increased IκB-α phosphorylation and reduced the levels of IκB-α. Pre-treatment of cells with luteolin abolished the effects of LPS on IκB-α.

Xagorari et al. (2001) concluded that luteolin inhibits protein tyrosine phosphorylation, nuclear factor-κB-mediated gene expression and pro-inflammatory cytokine production in murine macrophages.

Anti-inflammatory; Neuroinflammation

Pre-treatment of primary murine microglia and BV-2 microglial cells with luteolin inhibited LPS-stimulated IL-6 production at both the mRNA and protein levels. Whereas luteolin had no effect on the LPS-induced increase in NF-κB DNA binding activity, it markedly reduced AP-1 transcription factor binding activity. Consistent with this finding, luteolin did not inhibit LPS-induced degradation of IκB-α but inhibited JNK phosphorylation.

Luteolin consumption reduced LPS-induced IL-6 in plasma 4 hours after injection. Furthermore, luteolin decreased the induction of IL-6 mRNA by LPS in the hippocampus but not in the cortex or cerebellum. Taken together, these data suggest luteolin inhibits LPS-induced IL-6 production in the brain by inhibiting the JNK signaling pathway and activation of AP-1 in microglia. Thus, luteolin may be useful for mitigating neuroinflammation (Jang et al., 2008).

Immunostimulatory and Anti-inflammatory

Luteolin (Lut) possesses significant anti-inflammatory activity in well-established models of acute and chronic inflammation, such as xylene-induced ear edema in mice (ED50= 107 mg/ kg), carrageenin-induced swellingof the ankle, acetic acid-induced pleurisy and croton oil-induced gaseous pouch granuloma in rats. Lut had a marked inhibitory effect on the inflammatory exudation, but did not affect the number of leucocytes. Its combined immunostimulatory and anti-inflammatory activity, and inhibitory effect upon immediate hypersensitive response, provide the pharmacologic bases for the beneficial effects of Lut in the treatment of chronic bronchitis (Chen et al., 1986).

Anti-inflammatory

Luteolin dose-dependently inhibited the expression and production of those inflammatory genes and mediators in macrophages stimulated with lipopolysaccharide (LPS). Semi-quantitative reverse-transcription polymerase chain reaction (RT-PCR) assay further confirmed the suppression of LPS-induced TNF- α, IL-6, iNOS and COX-2 gene expression by luteolin at a transcriptional level. Luteolin also reduced the DNA binding activity of nuclear factor-kappa B (NF-κB) in LPS-activated macrophages.

In addition, luteolin significantly inhibited the LPS-induced DNA binding activity of activating protein-1 (AP-1). It was also found that luteolin attenuated the LPS-mediated protein kinase B (Akt) and IKK phosphorylation, as well as reactive oxygen species (ROS) production. In sum, these data suggest that, by blocking NF-κB and AP-1 activation, luteolin acts to suppress the LPS-elicited inflammatory events in mouse alveolar macrophages, and this effect was mediated, at least in part, by inhibiting the generation of reactive oxygen species. These observations suggest a possible therapeutic application of this agent for treating inflammatory disorders in the lung (Chen et al., 2007).

Pancreatic Cancer; Chemo-enhancing

Simultaneous treatment or pre-treatment (0, 6, 24 and 42h) of flavonoids and chemotherapeutic drugs and various concentrations (0-50µM) were assessed using the MTS cell proliferation assay. Pre-treatment for 24 hours with 13µM of either Apigenin or Luteolin, followed by Gem for 36 h was optimal to inhibit cell proliferation.

Pre-treatment of cells with 11-19µM of either flavonoid for 24 hours resulted in 59%–73% growth inhibition when followed by Gem (10µM, 36 hours). Lut (15µM, 24 hours) pre-treatment followed by Gem (10µM, 36h), significantly decreased protein expression of nuclear GSK-3β and NF-κB p65 and increased pro-apoptotic cytosolic cytochrome c. Pre-treatment of human pancreatic cancer cells BxPC-3 with low concentrations of Lut effectively aid in the anti-proliferative activity of chemotherapeutic drugs (Johnson et al., 2013).

Ovarian Cancer

Recent studies further indicate that luteolin potently inhibits VEGF production and suppresses ovarian cancer cell metastasis in vitro. Lastly, oridonin and wogonin were suggested to suppress ovarian CSCs as is reflected by down-regulation of the surface marker EpCAM.

Unlike NSAIDS (non-steroid anti-inflammatory drugs), well-documented clinical data for phyto-active compounds are lacking. In order to evaluate objectively the potential benefit of these compounds in the treatment of ovarian cancer, strategically designed, large scale studies are warranted (Chen et al., 2012).

Chemo-sensitizer

The sensitization effect of luteolin on cisplatin-induced apoptosis is p53 dependent, as such effect is only found in p53 wild-type cancer cells but not in p53 mutant cancer cells. Moreover, knockdown of p53 by small interfering RNA made p53 wild-type cancer cells resistant to luteolin and cisplatin. The critical role of c-Jun NH(2)-terminal kinase (JNK) was identified in regulation of p53 protein stability: luteolin activates JNK, and JNK then stabilizes p53 via phosphorylation, leading to reduced ubiquitination and proteasomal degradation.

An in vivo nude mice xenograft model confirmed that luteolin enhanced the cancer therapeutic activity of cisplatin via p53 stabilization and accumulation. In summary, data from this study reveal a novel molecular mechanism involved in the anti-cancer effects of luteolin and support its potential clinical application as a chemo-sensitizer in cancer therapy (Shi et al., 2007).

Breast Cancer; Chemo-sensitzer

Luteolin is a flavonoid that has been identified in many plant tissues and exhibits chemo-preventive or chemo-sensitizing properties against human breast cancer. However, the oncogenic molecules in human breast cancer cells that are inhibited by luteolin treatment have not been identified.

Relatively high levels of cyclin E2 (CCNE2) protein expression were detected in tamoxifen-resistant (TAM-R) MCF-7 cells. These results showed that the level of CCNE2 protein expression was specifically inhibited in luteolin-treated (5µM) TAM-R cells, either in the presence or absence of 4-OH-TAM (100nM). Combined treatment with 4-OH-TAM and luteolin synergistically sensitized the TAM-R cells to 4-OH-TAM. The results of this study suggest that luteolin can be used as a chemo-sensitizer to target the expression level of CCNE2 and that it could be a novel strategy to overcome TAM resistance in breast cancer patients (Tu et al., 2013).

References

Baliga MS, Jimmy R, Thilakchand KR, et al. (2013). Ocimum sanctum L (Holy Basil or Tulsi) and its phytochemicals in the prevention and treatment of cancer. Nutr Cancer, 65(1):26-35. doi: 10.1080/01635581.2013.785010.

Chen CY, Peng WH, Tsai KD and Hsu SL. (2007). Luteolin suppresses inflammation-associated gene expression by blocking NF- κ B and AP-1 activation pathway in mouse alveolar macrophages. Life Sciences, 81(23-24):1602-1614. doi:10.1016/j.lfs.2007.09.028

Chen MZ, Jin WZ, Dai LM, Xu SY. (1986). Effect of luteolin on inflammation and immune function. Chinese Journal of Pharmacology and Toxicology, 1986-01.

Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.

Jang S, Kelley KW, Johnson RW. (2008). Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. PNAS, 105(21):7534-7539

Johnson JL, Gonzalez de Mejia E. (2013). Interactions between dietary flavonoids apigenin or luteolin and chemotherapeutic drugs to potentiate anti-proliferative effect on human pancreatic cancer cells, in vitro. Food Chem Toxicol, S0278-6915(13)00491-2. doi: 10.1016/j.fct.2013.07.036.

Lim DY, Jeong Y, Tyner Al., Park JHY. (2007). Induction of cell-cycle arrest and apoptosis in HT-29 human colon cancer cells by the dietary compound luteolin. Am J Physiol Gastrointest Liver Physiol, 292: G66-G75. doi:10.1152/ajpgi.00248.2006.

Shi R, Huang Q, Zhu X, et al. (2007). Luteolin sensitizes the anti-cancer effect of cisplatin via c-Jun NH2-terminal kinase-mediated p53 phosphorylation and stabilization. Molecular Cancer Therapeutics, 6(4):1338-1347. doi: 10.1158/1535-7163.MCT-06-0638.

Tu SH, Ho CT, Liu MF, et al. (2013). Luteolin sensitizes drug-resistant human breast cancer cells to tamoxifen via the inhibition of cyclin E2 expression. Food Chem, 141(2):1553-61. doi: 10.1016/j.foodchem.2013.04.077.

Xagorari A, Papapetropoulos A, Mauromatis A, et al. (2001). Luteolin inhibits an endotoxin-stimulated phosphorylation cascade and pro-inflammatory cytokine production in macrophages. JPET, 296(1):181-187.

Kaempferol

Cancer:
Pancreatic, osteosarcoma, metastasis, ovarian cancer

Action: Anti-oxidation, anti-inflammatory, cell-cycle arrest, blood sugar regulation, estrogen receptor modulator, metastasis

Kaempferol is a flavonol compound present in various plants and Chinese medicinal herbs, including Allium cepa (L.).

Blood Sugar Regulation

It has been found that kaempferol, a flavonol compound present in various Chinese medicinal herbs, has cyto-protective effects on cultured clonal beta-cells and pancreatic human islets. Kaempferol treatment dose-dependently promoted viability, inhibited cellular apoptosis, and reduced caspase-3 activity in beta-cells and human islets exposed to chronic high glucose, with 10 µM kaempferol exerting the maximum effect. In addition, kaempferol treatment improved the expression of anti-apoptotic proteins Akt and Bcl-2, that was significantly reduced in beta-cells and human islets chronically exposed to hyperglycemia.

Furthermore, exposure of beta-cells and human islets to kaempferol restored high glucose-attenuated intracellular cAMP and ATP production. Inhibition of protein kinase A or Akt activation ablated the anti-apoptotic effect of kaempferol. These cytoprotective effects of kaempferol were associated with improved insulin secretory function and synthesis in beta-cells and human islets.

These findings provide evidence that kaempferol may be a naturally occurring anti-diabetic compound through protecting pancreatic beta-cell survival and function in a hostile environment that would otherwise lead to type 2 diabetes (Zhang et al., 2011).

Ovarian Cancer

Recent studies indicate that apigenin, genistein, kaempferol, luteolin, and quercetin potently inhibit VEGF production and suppress ovarian cancer cell metastasis in vitro. Unlike NSAIDS (non-steroid anti-inflammatory drugs), well-documented clinical data for phyto-active compounds are lacking. In order to evaluate objectively the potential benefit of these compounds in the treatment of ovarian cancer, strategically designed, large scale studies are warranted (Chen et al., 2012).

Estrogen Receptor Modulator

Kaempferol is a dietary flavonoid that can function as a selective estrogen receptor modulator (SERM). Estrogen-related receptors alpha and gamma (ERR α and ERRγ) are orphan nuclear receptors that play important roles in mitochondrial biogenesis and cancer development. Wang, Gao, & Wang (2013) have shown that kaempferol can functionally antagonize the activities of ERRs based on both response element reporter systems and target gene analysis. Kaempferol modulation of mitochondrial function and suppression of cancer cell growth has been confirmed. These findings suggest that kaempferol may exert its anti-cancer activities through antagonizing ERRs activity.

Osteosarcoma; Metastasis

Kaempferol displayed inhibitory effects on the invasion and adhesion of U-2 osteosarcoma (OS) cells in a concentration-dependent manner and it also inhibited the migration of U-2 OS cells in a concentration-dependent manner. Kaempferol treatment reduced the enzymatic activities and protein levels of matrix metalloproteinase (MMP)-2, MMP-9 and urokinase plasminogen activator (uPA) and furthermore, kaempferol was able to reduce the protein phosphorylation of ERK, p38 and JNK. Results suggest a potential role for kaempferol in the therapy of tumor metastasis of OS (Chen et al., 2013).

Cell-cycle Arrest

Kaempferol decreased cell viability as determined by MTT assays and induced a G2/M phase cell-cycle arrest in a concentration-dependent manner. CDK1/cyclin B expression and the AMPK and AKT signaling pathways contributed to kaempferol-induced G2/M cell-cycle arrest and autophagic cell death in SK-HEP-1 human hepatic cancer cells (Huang et al., 2013).

References

Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.


Chen HJ, Lin CM, Lee CY, et al. (2013). Kaempferol suppresses cell metastasis via inhibition of the ERK-p38-JNK and AP-1 signaling pathways in U-2 OS human osteosarcoma cells. Oncol Rep, 30(2):925-32. doi: 10.3892/or.2013.2490.


Huang WW, Tsai SC, Peng SF, et al. (2013). Kaempferol induces autophagy through AMPK and AKT signaling molecules and causes G2/M arrest via down-regulation of CDK1/cyclin B in SK-HEP-1 human hepatic cancer cells. Int J Oncol, 42(6):2069-77. doi: 10.3892/ijo.2013.1909.


Wang H, Gao M, Wang J. (2013). Kaempferol inhibits cancer cell growth by antagonizing estrogen-related receptor α and γ activities. Cell Biol Int. doi: 10.1002/cbin.10152.


Zhang Y, Liu D. (2011). Flavonol kaempferol improves chronic hyperglycemia-impaired pancreatic beta-cell viability and insulin secretory function. Eur J Pharmacol, 670(1):325-32. doi: 10.1016/j.ejphar.2011.08.011.

Evodiamine

Cancer: Pancreatic, gastric, breast; ER+, ER-, lung

Action: Inhibits NF- κB, inhibits metastasis, increases intracellular ROS, apoptosis, cell-cycle arrest, anti-cancer, MDR

Evodiamine, a naturally occurring indole alkaloid, is one of the main bioactive ingredients of Evodia rutaecarpa [(Juss.) Benth.] (alkaloidal component of the extract). With respect to the pharmacological actions of evodiamine, more attention has been paid to beneficial effects in insults involving cancer, obesity, nociception, inflammation, cardiovascular diseases, Alzheimer's disease, infectious diseases and thermo-regulative effects. Evodiamine has evolved a superior ability to bind various proteins (Yu et al., 2013). Evodiamine exhibits anti-proliferative, anti-metastatic, and apoptotic activities.

Anti-cancer, MDR

Evodiamine possesses anti-anxiety, anti-obesity, anti-nociceptive, anti-inflammatory, anti-allergic, and anti-cancer effects. As well, it has thermoregulation, protection of myocardial ischemia-reperfusion injury and vessel-relaxing activities (Kobayashi, 2003; Shin et al., 2007; Ko et al., 2007; Ji, 2011). Evodiamine exhibits anti-cancer activities both in vitro and in vivo by inducing cell-cycle arrest or apoptosis, and inhibiting angiogenesis, invasion, and metastasis in a variety of cancer cell lines (Ogasawara et al., 2001; Ogasawara et al., 2002; Fei et al., 2003; Shyu et al., 2006). It presents anti-cancer potentials at micromolar concentrations and even at the nanomolar level in some cell lines in vitro (Lee et al., 2006; Wang, Li, & Wang, 2010). Evodiamine also stimulates autophagy, which serves as a survival function (Yang et al., 2008). Compared with other compounds, evodiamine is less toxic to normal human cells, such as human peripheral blood mononuclear cells (Fei et al., 2003; Zhang et al., 2004). It also inhibits the proliferation of adriamycin-resistant human breast cancer NCI/ADR-RES cells both in vitro and in Balb-c/nude mice (Liao et al., 2005).

Lung Cancer, Cell-cycle Arrest

Evodiamine (10  mg/kg) administrated orally twice daily significantly inhibits   tumor growth (Liao et al., 2005). Moreover, treatment with 10 mg/kg evodiamine from the 6th day after tumor inoculation into mice reduces lung metastasis and does not affect the body weight of mice during the experimental period (Ogasawara et al., 2001).

Cell-cycle Arrest

Evodiamine inhibits TopI enzyme, forms the DNA covalent complex with a similar concentration to that of irinotecan, and induces DNA damage (Chan et al., 2009; Tsai et al., 2010; Dong et al., 2010). However, TopI may not be the main target of this compound. Cancer cells treated with evodiamine exhibit G 2 / M phase arrest (Kan et al., 2004; Huang et al., 2004; Liao et al., 2005) rather than S phase arrest, which is not consistent with the mechanism of classic TopI inhibitors, such as irinotecan. Therefore, other targets aside from TopI may also be important for realizing the anti-cancer potentials of evodiamine. This statement is supported by the fact that evodiamine has effects on tubulin polymerization (Huang et al., 2004).

Increases Intracellular ROS, Apoptosis

Exposure to evodiamine rapidly increases intracellular ROS followed by an onset of mitochondrial depolarization (Yang et al., 2007). The generation of ROS and nitric oxide acts in synergy and triggers mitochondria-dependent apoptosis (Yang et al., 2008). Evodiamine also induces caspase-dependent and caspase-independent apoptosis, down-regulates Bcl-2 expression, and up-regulates Bax expression in some cancer cells (Zhang et al., 2003; Lee et al., 2006). The phosphatidylinositol 3-kinase/Akt/caspase and Fas ligand (Fas-L)/NF-κB signaling pathways might account for evodiamine-induced cell death. Moreover, these signals could be increased by the ubiquitin-proteasome pathway (Wang, Li, & Wang, 2010).

Inhibits Metastasis

Evodiamine has a marked inhibitory activity on tumor cell migration in vitro. When evodiamine at 10 mg/kg was administered into mice from the 6th day after tumor inoculation, the number of tumor nodules in lungs was decreased by 48% as compared to control. The inhibition rate was equivalent to that produced by cisplatin. Results suggest that evodiamine may be regarded as a promising agent in tumor metastasis therapy (Ogasawara et al., 2005).

Inhibits NF-κB

Evodiamine inhibited tumor necrosis factor (TNF)-induced Akt activation and its association with IKK. This down-regulation potentiated the apoptosis induced by cytokines and chemotherapeutic agents and suppressed TNF-induced invasive activity. Overall, these results indicate that evodiamine inhibits both constitutive and induced NF-κB activation and NF-κB-regulated gene expression (Takada et al., 2005).

Breast Cancer

Endocrine sensitivity, assessed by the expression of estrogen receptor (ER), has long been the predict factor to guide therapeutic decisions. Tamoxifen has been the most successful hormonal treatment in endocrine-sensitive breast cancer. However, in estrogen-insensitive cancer tamoxifen showed less effectiveness than in estrogen-sensitive cancer. It is interesting to develop new drugs against both hormone-sensitive and insensitive tumor. In this present study Wang et al. (2013) examined anti-cancer effects of evodiamine extracted from the Chinese herb, Evodiae fructus, in estrogen-dependent and -independent human breast cancer cells, MCF-7 and MDA-MB-231 cells, respectively.

Breast Cancer; ER+, ER-

The expression of ER α and β in protein and mRNA levels was down-regulated by evodiamine according to data from immunoblotting and RT-PCR analysis. Overall, results indicate that evodiamine mediates degradation of ER and induces caspase-dependent pathway leading to inhibition of proliferation of breast cancer cell lines. It suggests that evodiamine may in part mediate through ER-inhibitory pathway to inhibit breast cancer cell proliferation.

Evodiamine (10 mg/kg) significantly reduced tumor growth and pulmonary metastasis. In vitro, evodiamine inhibited cell migration and invasion abilities through down-regulation of MMP-9, urokinase-type plasminogen activator (uPA) and uPAR expression. Evodiamine-induced G0/G1 arrest and apoptosis were associated with a decrease in Bcl-2, cyclin D1 and cyclin-dependent kinase 6 (CDK6) expression and an increase in Bax and p27Kip1 expression (Du et al., 201).

Gastric Cancer

A study by Rasul et al. (2012) was conducted to investigate the synchronized role of autophagy and apoptosis in evodiamine-induced cytotoxic activity on SGC-7901 human gastric adenocarcinoma cells and further to elucidate the underlying molecular mechanisms. Evodiamine significantly inhibited the proliferation of SGC-7901 cells and induced G2/M phase cell-cycle arrest.

Evodiamine-induced autophagy is partially involved in the death of SGC-7901 cells which was confirmed by using the autophagy inhibitor 3-methyladenine (3-MA). Evodiamine has therapeutic potential against cancers.

Pancreatic Cancer

In vitro application of the combination therapy triggered significantly higher frequency of pancreatic cancer cells apoptosis, inhibited the activities of PI3K, Akt, PKA, mTOR and PTEN, and decreased the activation of NF-κB and expression of NF- κB-regulated products. Evodiamine can augment the therapeutic effect of gemcitabine in pancreatic cancer through direct or indirect negative regulation of the PI3K/Akt pathway (Wei et al., 2012).

References

Chan ALF, Chang WS, Chen LM et al. (2009). Evodiamine stabilizes topoisomerase I-DNA cleavable complex to inhibit topoisomerase I activity. Molecules, (14):4:1342–1352.


Dong G, Sheng C, Wang CS, et al. (2010). Selection of evodiamine as a novel topoisomerase i inhibitor by structure-based virtual screening and hit optimization of evodiamine derivatives as anti-tumor agents. Journal of Medicinal Chemistry, 53(21):7521–7531.


Du J, Wang XF, Zhou QM, et al. (2013). Evodiamine induces apoptosis and inhibits metastasis in MDA “American Typewriter”; “American Typewriter”;‑ MB-231 human breast cancer cells in vitro and in vivo. Oncol Rep, 30(2):685-94. doi: 10.3892/or.2013.2498.


Fei XF, Wang BX, T. Li TJ et al. (2003). Evodiamine, a constituent of Evodiae Fructus, induces anti-proliferating effects in tumor cells. Cancer Science, 94(1):92–98.


Huang YC, Guh JH, Teng CM. (2004). Induction of mitotic arrest and apoptosis by evodiamine in human leukemic T-lymphocytes. Life Sciences, 75(1):35–49.


Ji YB. (2011). Active Ingredients of Traditional Chinese Medicine: Pharmacology and Application. People's Medical Publishing House Co., LTD. Connecticut USA


Kan SF, Huang WJ, Lin LC, Wang PS. (2004). Inhibitory effects of evodiamine on the growth of human prostate cancer cell line LNCaP. International Journal of Cancer, 110(5):641–651.


Ko HC, Wang YH, Liou KT et al. (2007). Anti-inflammatory effects and mechanisms of the ethanol extract of Evodia rutaecarpa and its bioactive components on neutrophils and microglial cells. European Journal of Pharmacology, 555(2-3):211–217.


Kobayashi Y. (2003). The nociceptive and anti-nociceptive effects of evodiamine from fruits of Evodia rutaecarpa in mice. Planta Medica, 69(5):425–428.


Lee TJ, Kim EJ, Kim S et al. (2006). Caspase-dependent and caspase-independent apoptosis induced by evodiamine in human leukemic U937 cells. Molecular Cancer Therapeutics, 5(9):2398–2407.


Liao CH, Pan SL, Guh JH et al. (2005). Anti-tumor mechanism of evodiamine, a constituent from Chinese herb Evodiae fructus, in human multiple-drug resistant breast cancer NCI/ADR-RES cells in vitro and in vivo. Carcinogenesis, 26(5):968–975.


Ogasawara M, Matsubara T, Suzuki H. (2001). Inhibitory effects of evodiamine on in vitro invasion and experimental lung metastasis of murine colon cancer cells. Biological and Pharmaceutical Bulletin, 24(8):917–920.


Ogasawara M, Matsunaga T, Takahashi S, Saiki I, Suzuki H. (2002). Anti-invasive and metastatic activities of evodiamine. Biological and Pharmaceutical Bulletin, 25(11):1491–1493.


Rasul A, Yu B, Zhong L, et al. (2012). Cytotoxic effect of evodiamine in SGC-7901 human gastric adenocarcinoma cells via simultaneous induction of apoptosis and autophagy. Oncol Rep, 27(5):1481-7. doi: 10.3892/or.2012.1694


Shin YW, Bae EA, Cai XF, Lee JJ, and Kim DH. (2007). In vitro and in vivo antiallergic effect of the fructus of Evodia rutaecarpa and its constituents, Biological and Pharmaceutical Bulletin, 30(1):197–199, 2007.


Shyu KG, Lin S, Lee CC et al. (2006). Evodiamine inhibits in vitro angiogenesis: implication for anti-tumorgenicity. Life Sciences, 78(19):2234–2243.


Takada Y, Kobayashi Y, Aggarwal BB. (2005). Evodiamine Abolishes Constitutive and Inducible NF- κB Activation by Inhibiting IκBα Kinase Activation, Thereby Suppressing NF-κ B-regulated Antiapoptotic and Metastatic Gene Expression, Up-regulating Apoptosis, and Inhibiting Invasion. The Journal of Biological Chemistry, 280:17203-17212. doi: 10.1074/jbc.M500077200.


Tsai HP, Lin LW, Lai ZY et al. (2010). Immobilizing topoisomerase I on a surface plasmon resonance biosensor chip to screen for inhibitors. Journal of Biomedical Science, 17(1):49.


Wang C, Li S, Wang MW. (2010). Evodiamine-induced human melanoma A375-S2 cell death was mediated by PI3K/Akt/caspase and Fas-L/NF- κ B signaling pathways and augmented by ubiquitin-proteasome inhibition. Toxicology in Vitro, 24(3):898–904.


Wang KL, Hsia SM, Yeh JY, et al. (2013). Anti-Proliferative Effects of Evodiamine on Human Breast Cancer Cells. PLoS One, 8(6):e67297.


Wei WT, Chen H, Wang ZH, et al. (2012). Enhanced anti-tumor efficacy of gemcitabine by evodiamine on pancreatic cancer via regulating PI3K/Akt pathway. Int J Biol Sci, 8(1):1-14.


Yu H, Jin H, Gong W, Wang Z, Liang H. (2013). Pharmacological actions of multi-target-directed evodiamine. Molecules, 18(2):1826-43. doi: 10.3390/molecules18021826.


Yang J, Wu LJ, Tashino SI, et al. (2007). Critical roles of reactive oxygen species in mitochondrial permeability transition in mediating evodiamine-induced human melanoma A375-S2 cell apoptosis. Free Radical Research, 41(10):1099–1108.


Zhang Y, Wu LJ, Tashiro SI, Onodera S, Ikejima T. (2003). Intracellular regulation of evodiamine-induced A375-S2 cell death. Biological and Pharmaceutical Bulletin, 26(11):1543–1547.


Zhang Y, Zhang QH, Wu LJ, et al. (2004). Atypical apoptosis in L929 cells induced by evodiamine isolated from Evodia rutaecarpa. Journal of Asian Natural Products Research, 6(1):19–27.

Apigenin

Cancer:
Breast, gastrointestinal., prostate, ovarian, pancreatic

Action: Anti-proliferative effect, induces apoptosis, chemo-sensitizer

Apigenin (4′,5,7-trihydroxyflavone, 5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a flavonoid found in many fruits, vegetables, and herbs, the most abundant sources being the leafy herb parsley and dried flowers of chamomile. Present in dietary sources as a glycoside, it is cleaved in the gastrointestinal lumen to be absorbed and distributed as apigenin itself. For this reason, the epithelium of the gastrointestinal tract is exposed to higher concentrations of apigenin than tissues at other locations. This would also be true for epithelial cancers of the gastrointestinal tract. There is evidence that the actions of apigenin might hinder the ability of gastrointestinal cancers to progress and spread.

Induces Apoptosis, Anti-metastatic

Apigenin has been shown to inhibit cell growth, sensitize cancer cells to elimination by apoptosis, and hinder the development of blood vessels to serve the growing tumor. It also has actions that alter the relationship of the cancer cells with their microenvironment. Apigenin is able to reduce cancer cell glucose uptake, inhibit remodeling of the extracellular matrix, inhibit cell adhesion molecules that participate in cancer progression, and oppose chemokine signaling pathways that direct the course of metastasis into other locations. As such, apigenin may provide some additional benefit beyond existing drugs in slowing the emergence of metastatic disease (Lefort, 2013).

Chemo-sensitizer, Induces Apoptosis

Choi & Kim (2009) investigated the effects of combined treatment with 5-fluorouracil and apigenin on proliferation and apoptosis, as well as the underlying mechanism, in human breast cancer MDA-MB-453 cells. The MDA-MB-453 cells, which have been shown to overexpress ErbB2, were resistant to 5-fluorouracil; 5-fluorouracil exhibited a small dose-dependent anti-proliferative effect, with an IC50 of 90 microM. Interestingly, combined treatment with apigenin significantly decreased the resistance. Cellular proliferation was significantly inhibited in cells exposed to 5-fluorouracil at its IC50 and apigenin (5, 10, 50 and 100 microM), compared with proliferation in cells exposed to 5-fluorouracil alone.

This inhibition in turn led to apoptosis, as evidenced by an increased number of apoptotic cells and the activation of caspase-3. Moreover, compared with 5-fluorouracil alone, 5-fluorouracil in combination with apigenin at concentrations >10 microM exerted a pro-apoptotic effect via the inhibition of Akt expression.

Taken together, results suggest that 5-fluorouracil acts synergistically with apigenin inhibiting cell growth and inducing apoptosis via the down-regulation of ErbB2 expression and Akt signaling (Choi, 2009).

Breast Cancer, Prostate Cancer

Two flavonoids, genistein and apigenin, have been implicated as chemo-preventive agents against prostate and breast cancers; however, the mechanisms behind their respective cancer-protective effects may vary significantly. It was thought that the anti-proliferative action of these flavonoids on prostate (DU-145) and breast (MDA-MB-231) cancer cells expressing only estrogen receptor (ER) β is mediated by this ER subtype. It was found that both genistein and apigenin, although not 17β-estradiol, exhibited anti-proliferative effects and pro-apoptotic activities through caspase-3 activation in these two cell lines. In yeast transcription assays, both flavonoids displayed high specificity toward ERβ transactivation, particularly at lower concentrations.

However, in mammalian assay, apigenin was found to be more ERβ-selective than genistein, which has equal potency in inducing transactivation through ERα and ERβ. Small interfering RNA-mediated down-regulation of ERβ abrogated the anti-proliferative effect of apigenin in both cancer cells but did not reverse that of genistein. These results unveil that the anti-cancer action of apigenin is mediated, in part, by ERβ. The differential use of ERα and ERβ signaling for transaction between genistein and apigenin demonstrates the complexity of phytoestrogen action in the context of their anti-cancer properties (Mak, 2006).

Ovarian Cancer

Id1 (inhibitor of differentiation or DNA binding protein 1) contributes to tumorigenesis by stimulating cell proliferation, inhibiting cell differentiation and facilitating tumor neoangiogenesis. Elevated Id1 is found in ovarian cancers and its level correlates with the malignant potential of ovarian tumors. Therefore, Id1 is a potential target for ovarian cancer treatment. It has been demonstrated that apigenin inhibits proliferation and tumorigenesis of human ovarian cancer A2780 cells through Id1. Apigenin has been found to suppress the expression of Id1 through activating transcription factor 3 (ATF3). These results may elucidate a new mechanism underlying the inhibitory effects of apigenin on cancer cells (Li, 2009).

Pancreatic Cancer

Simultaneous treatment or pre-treatment (0, 6, 24 and 42 hours) of apigenin and chemotherapeutic drugs and various concentrations (0-50µM) were assessed using the MTS cell proliferation assay. Simultaneous treatment with apigenin (0,13, 25 or 50µM) and chemotherapeutic drugs 5-fluorouracil (5-FU, 50µM) or gemcitabine (Gem, 10µM) for 60 hours resulted in less-than-additive effect (p<0.05). Pre-treatment for 24 hours with 13µM of apigenin, followed by Gem for 36 hours was optimal to inhibit cell proliferation.

Pre-treatment of cells with 11-19µM of apigenin for 24 hours resulted in 59-73% growth inhibition when followed by Gem (10µM, 36h). Pre-treatment of human pancreatic cancer cells BxPC-3 with low concentrations of apigenin hence effectively aids in the anti-proliferative activity of chemotherapeutic drugs (Johnson, 2013).

Induces Apoptosis, Inhibits Angiogenesis and Metastasis.

Preclinical studies have also shown that Ocimum sanctum L. and some of the phytochemicals it contains (including apigenin) prevents chemical-induced skin, liver, oral., and lung cancers. These effects are thought to be mediated by increasing the anti-oxidant activity, altering gene expression, inducing apoptosis, and inhibiting angiogenesis and metastasis. The aqueous extract of Ocimum sanctum L. has been shown to protect mice against γ-radiation-induced sickness and mortality and to selectively protect the normal tissues against the tumoricidal effects of radiation. In particular, important phytochemicals like apigenin have also been shown to prevent radiation-induced DNA damage. This warrants its future research to establish its activity and utility in cancer prevention and treatment (Baliga, 2013).

Lung Cancer

Apigenin has been found to induce apoptosis and cell death in lung epithelium cancer (A549) cells with an IC50 value of 93.7 ± 3.7 µM for 48 hours treatment. Target identification investigations using A549 cells and in cell-free systems demonstrate that apigenin depolymerized microtubules and inhibited reassembly of cold depolymerized microtubules of A549 cells. Again apigenin inhibited polymerization of purified tubulin with an IC50 value of 79.8 ± 2.4 µM. Interestingly, apigenin also showed synergistic anti-cancer effects with another natural anti-tubulin agent, curcumin. Apigenin and curcumin synergistically induce cell death and apoptosis and also block cell-cycle progression at G2/M phase of A549 cells.

Understanding the mechanism of the synergistic effect of apigenin and curcumin could help to develop anti-cancer combination drugs from cheap and readily available nutraceuticals (Choudhury, 2013).

Induces Apoptosis

It has been shown that the dietary flavonoid apigenin binds and inhibits adenine nucleotide translocase-2 (ANT2), resulting in enhancement of Apo2L/TRAIL-induced apoptosis by up-regulation of DR5, making it a potential cancer therapeutic agent. Apigenin has been found to enhance Apo2L/TRAIL-induced apoptosis in cancer cells by inducing DR5 expression through binding ANT2. Similarly to apigenin, knockdown of ANT2 enhanced Apo2L/TRAIL-induced apoptosis by up-regulating DR5 expression at the post-transcriptional level.

Moreover, silencing of ANT2 attenuated the enhancement of Apo2L/TRAIL-induced apoptosis by apigenin. These results suggest that apigenin Up-regulates DR5 and enhances Apo2L/TRAIL-induced apoptosis by binding and inhibiting ANT2. ANT2 inhibitors like apigenin may hence contribute to Apo2L/TRAIL therapy (Oishi, 2013).

Colorectal Cancer

Apigenin has anti-proliferation, anti-invasion and anti-migration effects in three kinds of colorectal adenocarcinoma cell lines, namely SW480, DLD-1 and LS174T. Proteomic analysis with SW480 indicated that apigenin up-regulated the expression of transgelin (TAGLN) in mitochondria to exert its anti-tumor growth and anti-metastasis effects. Apigenin decreased the expression of MMP-9 in a dose-dependent manner. Transfection of three truncated forms of TAGLN and wild type has identified TAGLN as a repressor of MMP-9 expression.

This research provides direct evidence that apigenin inhibits tumor growth and metastasis both in vitro and in vivo. Apigenin up-regulates TAGLN and down-regulates MMP-9 expression through decreasing phosphorylation of Akt at Ser473 and in particular Thr308 to prevent cancer cell proliferation and migration (Chunhua, 2013).

References

Baliga MS, Jimmy R, Thilakchand KR, et al. (2013). Ocimum Sanctum L (Holy Basil or Tulsi) and Its Phytochemicals in the Prevention and Treatment of Cancer. Nutr Cancer, 65(1):26-35. doi: 10.1080/01635581.2013.785010.

 

 

Choi EJ, Kim GH. (2009). 5-Fluorouracil combined with apigenin enhances anti-cancer activity through induction of apoptosis in human breast cancer MDA-MB-453 cells. Oncol Rep, 22(6):1533-7.

 

Choudhury D, Ganguli A, Dastidar DG, et al. (2013). Apigenin shows synergistic anti-cancer activity with curcumin by binding at different sites of tubulin. Biochimie, 95(6):1297-309. doi: 10.1016/j.biochi.2013.02.010.

 

Chunhua L, Donglan L, Xiuqiong F, et al. (2013). Apigenin up-regulates transgelin and inhibits invasion and migration of colorectal cancer through decreased phosphorylation of AKT. J Nutr Biochem. doi: 10.1016/j.jnutbio.2013.03.006.

 

Johnson JL, Gonzalez de Mejia E. (2013). Interactions between dietary flavonoids apigenin or luteolin and chemotherapeutic drugs to potentiate anti-proliferative effect on human pancreatic cancer cells, in vitro. Food Chem Toxicol, 20:83-91. doi: 10.1016/j.fct.2013.07.036.

 


Lefort ƒC, Blay J. (2013). Apigenin and its impact on gastrointestinal cancers. Mol Nutr Food Res, 57(1):126-44. doi: 10.1002/mnfr.201200424.

 

Li ZD, Hu XW, Wang YT & Fang J. (2009). Apigenin inhibits proliferation of ovarian cancer A2780 cells through Id1. FEBS Letters, 583(12):1999-2003 doi:10.1016/j.febslet.2009.05.013.

 

Mak P, Leung YK, Tang WY, Harwood C & Ho SM. (2006). Apigenin suppresses cancer cell growth through ERβ. Neoplasia, 8(11):896–904.

 

Oishi M, Iizumi Y, Taniguchi T, et al. (2013). Apigenin Sensitizes Prostate Cancer Cells to Apo2L/TRAIL by Targeting Adenine Nucleotide Translocase-2. PLoS One, 8(2):e55922. doi: 10.1371/journal.pone.0055922.

Icariin

Cancer: Breast, gastric, Leydig cell, gall bladder

Action: Potentiates chemotherapy, restores T cell function, MDR, induces apoptosis

Estrogen Agonist

Icariin is a pure extract of the traditional Chinese medicine Herba epimedii. It is a flavonoid found in several species of the genus Epimedium (L.).

The estrogenic activities of icariin (ICA) and its derivatives were investigated, and their structure-estrogenic activity relationship determined. Icaritin (ICT) and desmethylicaritin (DICT) were derived from ICA. The estrogenic activities of ICA, ICT and DICT were examined by cell proliferation and progestogen receptor mRNA expression of estrogen-receptor-positive MCF-7 cells.

These studies indicated that ICT and DICT both markedly enhanced the proliferation of MCF-7 cells; as compared to estradiol (100%); their relative proliferative effects (RPE) were 90% and 94%, respectively. Those phenomena were not observed with ICA. Results demonstrate that ICT and DICT (nonconjugated forms) possess estrogen-like activity; however, ICA appears to have no estrogenicity in the MCF-7 cell line model in vitro (Ye et al., 2005).

Gastric Cancer

In an in vitro study, the inhibitory effect and underlying molecular mechanism of icariin was investigated on the invasive and migration properties of human gastric cancer cell line BGC-823. At 50% growth-inhibiting concentration, icariin significantly suppressed tumor cells migration and invasion, which were traceable to down-regulation of Rac1 and VASP.

Together with icariin, the selected siRNA targeting Rac1 or VASP reinforced these inhibitory effects. Moreover, transfection with Rac1 plasmids pcDNA3-EGFP-Rac1-Q61L led to the enhancement in expression level of both Rac1 and VASP.

These results indicate that icariin exerts negative effects on tumor cell invasion and migration via the Rac1-dependent VASP pathway and may be a potential anti-cancer drug (Wang et al., 2010).

Gallbladder Cancer; Gemcitabine

Icariin, by suppressing NF-κB activity, exerts anti-tumor activity, and potentiates the anti-tumor activity of gemcitabine in gallbladder cancer. Combined administration of gemcitabine and icariin may offer a better therapeutic option for patients with gallbladder cancer. Icariin (40-160 µg/mL) dose-dependently suppressed cell proliferation and induced apoptosis in both GBC-SD and SGC-996 cells, with SGC-996 cells being less sensitive to the drug. Icariin (40 µg/mL) significantly enhanced the anti-tumor activity of gemcitabine (0.5 µmol/L) in both GBC-SD and SGC-996 cells (Zhang et al., 2013).

Restores T cell function

Tumor-induced myeloid-derived suppressor cells (MDSCs) are a critical barrier to effective immunotherapy of cancer. We identified that Docetaxel and a natural compound, Icariin, can target MDSCs with preferential apoptosis of M2 cells and polarization of the surviving cells towards M1 cells. Such strategic targeting of MDSCs restored T cell function accompanied by tumor retardation in vivo (Djeu & Wei, 2012).

Leydig Cell (Testicle)

Findings suggest a novel anti-cancer effect of icariin in Leydig cell tumor, derived from interstitial cells (rare neoplasm) through activation of the mitochondrial pathway and down-regulation of the expression of piwil4 (Wang et al., 2011).

Induces Apoptosis

Icariin triggered the mitochondrial/caspase apoptotic pathway indicated by enhanced Bax-to-Bcl-2 ratio, loss of mitochondrial membrane potential., cytochrome c release, and caspase cascade. Moreover, icariin induced a sustained activation of the phosphorylation of c-Jun N-terminal kinase (JNK) but not p38 and ERK1/2, and SP600125 (an inhibitor of JNK) almost reversed icariin-induced apoptosis in SMMC-7721 cells. In addition, icariin provoked the generation of reactive oxygen species (ROS) in SMMC-7721 cells, while the anti-oxidant N-acetyl cysteine almost completely blocked icariin-induced JNK activation and apoptosis. Taken together, these findings suggest that icariin induces apoptosis through a ROS/JNK-dependent mitochondrial pathway (Li et al., 2010).

References

Djeu J, Wei S. (2012). Chemoimmunomodulation of MDSCs as a novel strategy for cancer therapy. Oncoimmunology, 1(1):121-122.


Li S, Dong P, Wang J, et al. (2010). Icariin, a natural flavonol glycoside, induces apoptosis in human hepatoma SMMC-7721 cells via a ROS/JNK-dependent mitochondrial pathway. Cancer Lett, 298(2):222-30. doi: 10.1016/j.canlet.2010.07.009.


Wang Y, Dong H, Zhu M, et al. (2010). Icariin exterts negative effects on human gastric cancer cell invasion and migration by vasodilator-stimulated phosphoprotein via Rac1 pathway. Eur J Pharmacol, 635(1-3):40-8. doi: 10.1016/j.ejphar.2010.03.017.


Wang Q, Hao J, Pu J, et al. (2011). Icariin induces apoptosis in mouse MLTC-10 Leydig tumor cells through activation of the mitochondrial pathway and down-regulation of the expression of piwil4. Int J Oncol, 39(4):973-80. doi: 10.3892/ijo.2011.1086.


Ye HY, Lou YJ. (2005). Estrogenic effects of two derivatives of icariin on human breast cancer MCF-7 cells. Phytomedicine, 12(10):735-41.


Zhang DC, Liu JL, Ding YB, Xia JG, Chen GY. (2013). Icariin potentiates the anti-tumor activity of gemcitabine in gallbladder cancer by suppressing NF-κ B. Acta Pharmacol Sin, 34(2):301-8. doi: 10.1038/aps.2012.162.

Isoflavones

Cancer: Prostate, breast, endometrial

Action: Anti-estrogenic effects, radio-protective effect, pneumonitis, cachexia-inhibiting

Prostate Cancer, Breast Cancer

Isoflavones have been investigated in detail for their role in the prevention and therapy of prostate cancer. This is primarily because of the overwhelming data connecting high dietary isoflavone intake with reduced risk of developing prostate cancer. A number of investigations have evaluated the mechanism(s) of anti-cancer action of isoflavones such as genistein, daidzein, biochanin A, equol, etc., in various prostate cancer models, both in vitro and in vivo.

Nuclear receptors are considered to be a central goal for maximizing treatment opportunities in breast cancer. Among natural ligands for estrogen receptors (ER and ERβ), which are members of the nuclear receptors super-family, are found isoflavones. These natural compounds have a similar structure to the main female hormone 17β-estradiol. A rich source of isoflavones is soy and its products. Three isoflavones of the aglycone form (genistein, daidzein, glycitein) are predominantly found in soybean and red clover. Other important isoflavones are biochanin A and formononetin (Bialešová et al., 2013).

Breast Cancer

Soy isoflavones do not function as an estrogen, but rather exhibit anti-estrogenic properties. However, their metabolism differs between humans and animals and therefore the outcomes of animal studies may not be applicable to humans. The majority of breast cancer cases are hormone-receptor-positive; therefore, soy isoflavones should be considered a potential anti-cancer therapeutic agent (Douglas et al., 2013).

Anti-cancer Effects

Use of soy isoflavone mixture has been advocated as an alternative, wherein daidzein can negate harmful effects of genistein. Recent research indicates the novel role of genistein and other isoflavones in the potentiation of radiation therapy, epigenetic regulation of key tumor suppressors and oncogenes, and the modulation of miRNAs, epithelial-to-mesenchymal transition, and cancer stem cells, which has renewed the interest of cancer researchers in this class of anti-cancer compounds (Ahmad et al. 2013).

Radiation-induced Pneumonitis, Radiation-induced Side-effects

Radiation-induced pneumonitis and fibrosis have restricted radiotherapy for lung cancer. In a preclinical lung tumor model, soy isoflavones showed the potential to enhance radiation damage in tumor nodules and simultaneously protect normal lung from radiation injury. Soy isoflavones given pre- and post-radiation protected the lungs against adverse effects of radiation including skin injury, hair loss, increased breathing rates, inflammation, pneumonitis and fibrosis, providing evidence for a radio-protective effect of soy (Hillman et al., 2013 a).

Radio-sensitizer

Combined soy and radiation caused a significantly stronger inhibition of tumor progression compared to each modality alone in contrast to large invasive tumor nodules seen in control mice. At the same time, soy reduced radiation injury in lung tissue by decreasing pneumonitis, fibrosis and protecting alveolar septa, bronchioles and vessels (Hillman et al., 2013 b).

Endometrial Cancer

Because of their anti-oxidant and anti-mutagenic properties, flavonoids may reduce cancer risk. Some flavonoids have anti-estrogenic effects that can inhibit the growth and proliferation of endometrial cancer cells. The intake of flavanols, flavanones, flavonols, anthocyanidins, flavones, isoflavones, and proanthocyanidins was measured and high consumption of selected proanthocyanidins may reduce endometrial cancer risk (Rossi et al., 2013).

Breast Cancer Protection

The evidence to date from observational epidemiologic studies, suggests that soy food intake, in the amount consumed in Asian populations (about 10 to 20 mg isoflavones per day), may be associated with a reduction of risk of breast cancer development as well as mortality and recurrence among women with breast cancer. The large number of clinical intervention studies on soy that have investigated intermediate biomarkers of breast cancer risk, including circulating estrogen levels, mammographic density, and breast tissue changes (cell proliferation), have not shown clear beneficial or deleterious effects (Wu et al., 2013).

Cachexia-Inhibiting

Isoflavones possess anti-proliferative effects of cachexia-inducing cells (MKN45cl85 and 85As2mLuc) cancer cell lines. Isoflavone treatment on the models induced tumor cytostasis, attenuation of cachexia, and prolonged survival whereas discontinuation of the treatment resulted in progressive tumor growth and weight loss (Yanagihara et al., 2013).

Methylation Effects

There is an inverse correlation between estrogenic marker complement (C)3 and genistein, which suggests an anti-estrogenic effect. Isoflavones induced dose-specific changes in RARβ2 and CCND2 gene methylation, which correlated with genistein levels. Research by Qin & Zhu (2009) provides novel insights into estrogenic and methylation effects of dietary isoflavones.

References

Ahmad A, Biersack B, Li Y, et al. (2013). Perspectives on the Role of Isoflavones in Prostate Cancer. AAPS J, 15(4):991-1000.


Bialešová L, Brtko J, Lenko V, Macejov‡ D. (2013). Nuclear receptors – target molecules for isoflavones in cancer chemoprevention. Gen Physiol Biophys.


Douglas CC, Johnson SA, Arjmandi BH. (2013). Soy and its isoflavones: the truth behind the science in breast cancer. Anti-cancer Agents Med Chem, 13(8):1178-87.


Hillman GG, Singh-Gupta V, Lonardo F, et al [a]. (2013). Radioprotection of Lung Tissue by Soy Isoflavones. J Thorac Oncol.


Hillman GG, Singh-Gupta V, Hoogstra DJ, et al [b]. (2013). Differential effect of soy isoflavones in enhancing high intensity radiotherapy and protecting lung tissue in a preclinical model of lung carcinoma. Radiother Oncol. doi: 10.1016/j.radonc.2013.08.015.


Rossi M, Edefonti V, Parpinel M, et al. (2013). Proanthocyanidins and other flavonoids in relation to endometrial cancer risk: a case-control study in Italy. Br J Cancer, 109(7):1914-1920. doi: 10.1038/bjc.2013.447.


Wu AH, Lee E, Vigen C. (2013). Soy isoflavones and breast cancer. Am Soc Clin Oncol Educ Book, 2013:102-6. doi: E10.1200/EdBook_AM.2013.33.102.


Yanagihara K, Takigahira M, Mihara K, et al. (2013). Inhibitory effects of isoflavones on tumor growth and cachexia in newly established cachectic mouse models carrying human stomach cancers. Nutr Cancer, 65(4):578-89. doi: 10.1080/01635581.2013.776089.

Andrographolide

Cancer: Leukemia, colorectal, lung

Action: Immunomodulatory,anti-inflammatory,anti-metastatic

Andrographolide (Andro), a diterpenoid lactone isolated from a traditional herbal medicine Andrographis paniculata [(Burm. f.) Wall. Ex Nees], is known to possess multiple pharmacological activities. Andrographolide has been shown to exhibit antioxidative, anti-cancer, anti-inflammatory, anti-diabetes, and anti-aging properties (Trivedi et al., 2007; Chao et al., 2010).

Immunomodulatory Activity

The immunomodulatory activity of HN-02, an extract containing a mixture of andrographolides, was evaluated at 1.0, 1.5, and 2.5 mg/kg on different in vivo and in vitro experimental models. It was also found that HN-02 treatment stimulated phagocytosis in mice. A significant increase in total WBC count and relative weight of spleen and thymus was observed in mice during 30 days of treatment with HN-02.

The present experimental findings demonstrate that HN-02 has the ability to enhance immune function, possibly through modulation of immune responses altered during antigen interaction, and to reverse the immunosuppression induced by CYP (Naik, 2009).

The ethanol extract and purified diterpene andrographolides of Andrographis paniculata (Acanthaceae) induced significant stimulation of antibody and delayed type hypersensitivity (DTH) response to sheep red blood cells (SRBC) in mice. The plant preparations also stimulated non-specific immune response of the animals measured in terms of macrophage migration index (MMI) phagocytosis of Escherichia coli and proliferation of splenic lymphocytes. The stimulation of both antigen specific and non-specific immune response was, however, of lower order with andrographolide than with the ethanol extract, suggesting that substance(s) other than andrographolide present in the extract may also be contributing towards immunostimulation (Puri, 1993)

Anti-inflammatory and Leukemic Therapies

Andrographolide has been shown to attenuate MMP-9 expression, with its main mechanism likely involving the NF-κB signal pathway. These results provide new opportunities for the development of new anti-inflammatory and leukemic therapies. This activity was shown in a study in which andrographolide (1–50µM) exhibited concentration-dependent inhibition of MMP-9 activation, induced by either tumor necrosis factor-α (TNF-α), or lipopolysaccharide (LPS), in THP-1cells.

Anti-inflammatory

Lee et al (2012) found that andrographolide could significantly inhibit the degradation of inhibitor-κB-α (IκB-α) induced by TNF-α. They used electrophoretic mobility shift assay and reporter gene detection to show that andrographolide also markedly inhibited NF-signaling, anti-translocation and anti-activation. These results provide new opportunities for the development of new anti-inflammatory and leukemic therapies.

Lung Cancer Metastasis

Andrographolide is known to have the potential to be developed as a chemotherapeutic agent, in particular in the treatment of lung cancer. In order to understand the anti-cancer properties of andrographolide, its effect on migration and invasion in human lung cancer A549 cells was examined. The results of the wound-healing assay and the in vitro transwell assay revealed that andrographolide inhibited dose-dependently the migration and invasion of A549 cells under non-cytotoxic concentrations.

These results indicated that andrographolide exerted an inhibitory effect on the activity and the mRNA and protein levels of MMP-7, but not MMP-2 or MMP-9. The andrographolide-inhibited MMP-7 expression or activity appeared to occur via activator protein-1 (AP-1) because its DNA binding activity was suppressed by andrographolide. Additionally, the transfection of Akt over-expression vector (Akt1 cDNA) to A549 cells could result in an increase expression of MMP-7 concomitantly with a marked induction on cell invasion. These findings suggested that the inhibition on MMP-7 expression by andrographolide may be through suppression on PI3K/Akt/AP-1 signaling pathway, which in turn leads to the reduced invasiveness of the cancer cells (Lee, 2010).

Colorectal Cancer

Andrographolide has also been shown to have potent anti-cancer activity against human colorectal carcinoma Lovo cells by inhibiting cell-cycle progression. To further investigate the mechanism for the anti-cancer properties of andrographolide, it was used to examine the effect on migration and invasion of Lovo cells. The results of wound-healing assay and in vitro transwell assay revealed that andrographolide inhibited dose-dependently the migration and invasion of Lovo cells under non-cytotoxic concentrations.

The down-regulation of MMP-7 appeared to be via the inactivation of activator protein-1 (AP-1) since the treatment with andrographolide suppressed the nuclear protein level of AP-1, which was accompanied by a decrease in DNA-binding level of the factor. Taken together, these results indicate that andrographolide reduces the MMP-7-mediated cellular events in Lovo cells, and provide a new mechanism for its anti-cancer activity (Shi, 2009)

Anti-inflammatory, Induces Apoptosis

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an important member of the tumor necrosis factor subfamily with great potential in cancer therapy; additionally andrographolide is known to possess potent anti-inflammatory and anti-cancer activities which may be attributed to its action on TRAIL. It has been shown that pre-treatment with andrographolide significantly enhances TRAIL-induced apoptosis in various human cancer cell lines, including those TRAIL-resistant cells.

Pre-treatment with an anti-oxidant (N-acetylcysteine) or a c-Jun NH(2)-terminal kinase inhibitor (SP600125) effectively prevented andrographolide-induced p53 activation and DR4 up-regulation and eventually blocked the andrographolide-induced sensitization on TRAIL-induced apoptosis. Taken together, these results present a novel anti-cancer effect of andrographolide and support its potential application in cancer therapy to overcome TRAIL resistance (Zhou, 2008).

References

Chao HP, Kuo CD, Chiu JH, Fu SL. (2010). Andrographolide exhibits anti-invasive activity against colon cancer cells via inhibition of MMP2 activity. Planta Medica, 76(16):1827–1833. doi: 10.1055/s-0030-1250039.


Lee WR, Chung CL, Hsiao CJ, et al. (2012). Suppression of matrix metalloproteinase-9 expression by andrographolide in human monocytic THP-1 cells via inhibition of NF- κB activation. Phytomedicine, 19(3):270-277. doi: 10.1016/j.phymed.2011.11.012


Lee YC, Lin HH, Hsu CH, et al. (2010). Inhibitory effects of andrographolide on migration and invasion in human non-small-cell lung cancer A549 cells via down-regulation of PI3K/Akt signaling pathway. Eur J Pharmacol, 632(1-3):23-32. doi: 10.1016/j.ejphar.2010.01.009.


Naik SR, Hule A. (2009). Evaluation of Immunomodulatory Activity of an Extract of Andrographolides from Andographis paniculata. Planta Med, 75(8):785-91. doi: 10.1055/s-0029-1185398.


Puri A, Saxena R, Saxena RP, et al. (1993). Immunostimulant agents from Andrographis paniculata. J Nat Prod, 56(7):995-9.


Shi MD, Lin HH, Chiang TA, et al. (2009). Andrographolide could inhibit human colorectal carcinoma Lovo cells migration and invasion via down-regulation of MMP-7 expression. Chem Biol Interact, 180(3):344-52. doi: 10.1016/j.cbi.2009.04.011.


Trivedi NP, Rawal UM, Patel BP. (2007). Hepato-protective effect of andrographolide against hexachlorocyclohexane- induced oxidative injury. Integrative Cancer Therapies, 6(3):271–280. doi: 10.1177/1534735407305985.


Zhou J, Lu GD, Ong CS, Ong CN, Shen HM. (2008). Andrographolide sensitizes cancer cells to TRAIL-induced apoptosis via p53-mediated death receptor 4 up-regulation. Mol Cancer Ther, 7(7):2170-80. doi: 10.1158/1535-7163.MCT-08-0071.

Ellagic Acid

Cancer:
Pancreatic, prostate, ovarian, breast, bladder, lymphoma, oral., melanoma

Action: Anti-cancer, induces apoptosis, promoted ROS and Ca2+ productions

Ellagic acid (EA) is a polyphenol compound widely found in fruits such as berries, walnuts, pecans, pomegranate, cranberries, and longan. It is well known to have a free radical scavenging activity and has been approved in Japan as an 'existing food additive' for anti-oxidative purposes (HHLW, 1996). In vitro evidence revealed that 100µM EA represented little toxic effect on human normal cells (Losso et al., 2004; Larrosa et al., 2006). A subchronic toxicity study further demonstrated that orally feeding EA (9.4, 19.1, 39.1g/kg b.w., resp.) could not induce mortality or treatment-related clinical signs throughout the experimental period on F344 rats (Tasaki et al., 2008), indicating the low toxicity of EA to mammalians. Furthermore, EA exhibits potent anti-cancer and anti-carcinogenesis activities towards breast, colorectal., oral., prostate (Losso et al., 2004; Larrosa et al., 2006; Malik et al., 2011), pancreatic (Edderkaoui et al., 2008), bladder (Li et al., 2005), neuroblastoma (Fjaeraa et al., 2009), melanoma (Kim et al., 2009), and lymphoma cells (Mishra et al., 2011).

Pancreatic Cancer

Edderkaoui et al. (2008) show that ellagic acid, a polyphenolic compound in fruits and berries, at concentrations 10 to 50 mmol/L stimulates apoptosis in human pancreatic adenocarcinoma cells. Ellagic acid stimulates the mitochondrial pathway of apoptosis associated with mitochondrial depolarization, cytochrome C release, and the downstream caspase activation. Ellagic acid does not directly affect mitochondria. Ellagic acid dose-dependently decreased NF-kappa B binding activity.

Furthermore, inhibition of NF-kappa B activity using IkB wild type plasmid prevented the effect of ellagic acid on apoptosis.

Pancreatic Cancer (PANC-1) cells were injected subcutaneously into Balb c nude mice, and tumor-bearing mice were treated with ellagic acid (EA). Treatment of PANC-1 xenografted mice with EA resulted in significant inhibition in tumor growth which was associated with suppression of cell proliferation and caspase-3 activation, and induction of PARP cleavage. EA also reversed epithelial to mesenchymal transition by up-regulating E-cadherin and inhibiting the expression of Snail, MMP-2 and MMP-9.

These data suggest that EA can inhibit pancreatic cancer growth, angiogenesis and metastasis by suppressing Akt, Shh and Notch pathways. In view of the fact that EA could effectively inhibit human pancreatic cancer growth by suppressing Akt, Shh and Notch pathways, our findings suggest that the use of EA would be beneficial for the management of pancreatic cancer (Zhao et al., 2013).

Ovarian Cancer

Ovarian carcinoma ES-2 and PA-1 cells were treated with EA (10~100  µ M) and assessed for viability, cell-cycle, apoptosis, anoikis, autophagy, and chemosensitivity to doxorubicin and their molecular mechanisms. EA inhibited cell proliferation in a dose- and time-dependent manner by arresting both cell lines at the G1 phase of the cell-cycle, which were from elevating p53 and Cip1/p21 and decreasing cyclin D1 and E levels. EA also induced caspase-3-mediated apoptosis by increasing the Bax :  Bcl-2 ratio and restored anoikis in both cell lines.

The enhancement of apoptosis and/or inhibition of autophagy in these cells by EA assisted the chemotherapy efficacy. The results indicated that EA is a potential novel chemoprevention and treatment assistant agent for human ovarian carcinoma Chung et al., 2013).

Prostate Cancer; AR+

In the present study, Pitchakarn et al. (2013) investigated anti-invasive effects of ellagic acid (EA) in androgen-independent human (PC-3) and rat (PLS10) prostate cancer cell lines in vitro. The results indicated that non-toxic concentrations of EA significantly inhibited the motility and invasion of cells examined in migration and invasion assays. They found that EA significantly reduced proteolytic activity of collagenase/gelatinase secreted from the PLS-10 cell line. Collagenase IV activity was also concentration-dependently inhibited by EA. These results demonstrated that EA has an ability to inhibit invasive potential of prostate cancer cells through action on protease activity.

Breast Cancer

The role of estrogen (E2) in regulation of cell proliferation and breast carcinogenesis is well-known. Recent reports have associated several miRNAs with estrogen receptors in breast cancers. Investigation of the regulatory role of miRNAs is critical for understanding the effect of E2 in human breast cancer, as well as developing strategies for cancer chemoprevention.

In this study Munagala et al. (2013) used the well-established ACI rat model that develops mammary tumors upon E2 exposure and identified a 'signature' of 33 significantly modulated miRNAs during the process of mammary tumorigenesis. Several of these miRNAs were altered as early as 3 weeks after initial E2 treatment and their modulation persisted throughout the mammary carcinogenesis process, suggesting that these molecular changes are early events. This is the first systematic study examining the changes in miRNA expression associated with E2 treatment in ACI rats as early as 3weeks until tumor time point. The effect of a chemo-preventive agent, ellagic acid in reversing miRNAs modulated during E2-mediated mammary tumorigenesis is also established. These observations provide mechanistic insights into the new molecular events behind the chemo-preventive action of ellagic acid and treatment of breast cancer.

Bladder Cancer

To investigate the effects of ellagic acid on the growth inhibition of TSGH8301 human bladder cancer cells in vitro, cells were incubated with various doses of ellagic acid for different time periods. Results indicated that ellagic acid induced morphological changes, decreased the percentage of viable cells through the induction of G0/G1 phase arrest and apoptosis, and also showed that ellagic acid promoted ROS and Ca2+ productions and decreased the level of ΔΨm and promoted activities of caspase-9 and -3.

On the basis of these observations, Ho et al (2013) suggest that ellagic acid induced cytotoxic effects for causing a decrease in the percentage of viable cells via G0/G1 phase arrest and induction of apoptosis in TSGH8301 cells.

Lymphoma

Protein Kinase C (PKC) isozymes are key components involved in cell proliferation and their over activation leads to abnormal tumor growth. PKC follows signaling pathway by activation of downstream gene NF-kB and early transcription factor c-Myc. Over activation of NF-kB and c-Myc gene are also linked with unregulated proliferation of cancer cells.

Therefore any agent which can inhibit the activation of Protein kinase C, NF-kB and c-Myc may be useful in reducing cancer progression. The role of ellagic acid was tested in regulation of tumor suppressor gene Transforming growth factor-β1 (TGF-β1). DL mice were treated with three different doses (40, 60 and 80 mg/kg body weight) of ellagic acid. Ascites cells of mice were used for the experiments. Ellagic acid administration to DL mice decreased oxidative stress by reducing lipid peroxidation.

The anti-carcinogenic action of ellagic acid was also confirmed by up-regulation of TGF-β1 and down-regulation of c-Myc. Lymphoma prevention by ellagic acid is further supported by decrease in cell proliferation, cell viability, ascites fluid accumulation and increase in life span of DL mice. All these findings suggest that ellagic acid prevents the cancer progression by down- regulation of PKC signaling pathway leading to cell proliferation (Mishra et al., 2013).

References

Chung YC, Lu LC, Tsai MH, et al. (2013). The inhibitory effect of ellagic Acid on cell growth of ovarian carcinoma cells. Evid Based Complement Alternat Med, 2013(2013):306705. doi: 10.1155/2013/306705.


Edderkaoui M, Odinokova I, Ohno I, et al. (2008). Ellagic acid induces apoptosis through inhibition of nuclear factor κ B in pancreatic cancer cells. World Journal of Gastroenterology, 14(23):3672–3680.


Fjaeraa C, NŒnberg E. (2009). Effect of ellagic acid on proliferation, cell adhesion and apoptosis in SH-SY5Y human neuroblastoma cells. Biomedicine and Pharmacotherapy, 63(4):254–261.


HHLW (Ministry of Health, Labor and Welfare of Japan). (1996). List of Existing Food Additives, Notification No. 120 of the Ministry of Health and Welfare.


Ho CC, Huang AC, Yu CS, Lien JC, et al. (2013). Ellagic acid induces apoptosis in tsgh8301 human bladder cancer cells through the endoplasmic reticulum stress- and mitochondria-dependent signaling pathways. Environ Toxicol. doi: 10.1002/tox.21857.


Kim S, Liu Y, Gaber MW, Bumgardner JD, Haggard WO, Yang Y. (2009). Development of chitosan-ellagic acid films as a local drug delivery system to induce apoptotic death of human melanoma cells. Journal of Biomedical Materials Research, 90(1):145–155.


Larrosa M, Tomás-Barberán FA, Espín JC. (2006). The dietary hydrolysable tannin punicalagin releases ellagic acid that induces apoptosis in human colon adenocarcinoma Caco-2 cells by using the mitochondrial pathway. Journal of Nutritional Biochemistry, 17(9):611–625.


Li TM, Chen GW, Su CC, et al. (2005). Ellagic acid induced p53/p21 expression, G1 arrest and apoptosis in human bladder cancer T24 cells. Anti-cancer Research, 25(2 A):971–979.


Losso JN, Bansode RR, Trappey A, II, Bawadi HA, Truax R. (2004). In vitro anti-proliferative activities of ellagic acid. Journal of Nutritional Biochemistry, 15(11):672–678.


Mishra S, Vinayak M. (2013). Ellagic acid checks lymphoma promotion via regulation of PKC signaling pathway. Mol Biol Rep, 40(2):1417-28. doi: 10.1007/s11033-012-2185-8.


Malik A, Afaq S, Shahid M, Akhtar K, Assiri A. (2011). Influence of ellagic acid on prostate cancer cell proliferation: a caspase-dependent pathway. Asian Pacific Journal of Tropical Medicine, 4(7):550–555.


Mishra S, Vinayak M. (2011). Anti-carcinogenic action of ellagic acid mediated via modulation of oxidative stress regulated genes in Dalton lymphoma bearing mice. Leukemia and Lymphoma, 52(11):2155–2161.


Munagala R, Aqil F, Vadhanam MV, Gupta RC. (2013). MicroRNA 'signature' during estrogen-mediated mammary carcinogenesis and its reversal by ellagic acid intervention. Cancer Lett, S0304-3835(13)00462-X. doi: 10.1016/j.canlet.2013.06.012.


Pitchakarn P, Chewonarin T, Ogawa K, et al. (2013). Ellagic Acid inhibits migration and invasion by prostate cancer cell lines. Asian Pac J Cancer Prev, 14(5):2859-63.


Tasaki M, Umemura T, Maeda M, et al. (2008). Safety assessment of ellagic acid, a food additive, in a subchronic toxicity study using F344 rats. Food and Chemical Toxicology, 46(3):1119–1124.


Zhao M, Tang SN, Marsh JL, et al. (2013). Ellagic acid inhibits human pancreatic cancer growth in Balb c nude mice. Cancer Letters, 337(2):210–217

Genistein (See also Daidzien)

Cancer:
Breast, kidney, prostate, renal., liver, endometrial., ovarian

Action: Anti-angiogenesis, cell-cycle arrest, cancer stem cells, VEGF, radiotherapy, sex hormone-binding globulin (SHBG), insulin-like growth factor-1 (IGF-1)

Genistein is a natural isoflavone phytoestrogen present in a number of plants, including soy, fava, and kudzu (Glycine max [(L.) Merr.], Vicia faba (L.), Pueraria lobata [(Willd.) Ohwi]).

Phytoestrogens

Phytoestrogens have been investigated at the epidemiological., clinical and molecular levels to determine their potential health benefits. The two major groups of phytoestrogens, isoflavones and lignans, are abundant in soy products and flax respectively, but are also present in a variety of other foods. It is thought that these estrogen-like compounds may protect against chronic diseases, such as hormone-dependent cancers, cardiovascular disease and osteoporosis (Stark & Madar, 2002).

S-Equol Production and Isoflavone Metabolism

S-Equol and Breast Cancer

Differences in ability to metabolize daidzein to equol might help explain inconsistent findings about isoflavones and breast cancer. Tseng et al. (2013) examined equol-producing status in relation to breast density, a marker of breast cancer risk, and evaluated whether an association of isoflavone intake with breast density differs by equol-producing status in a sample of Chinese immigrant women. In their sample, 30% were classified as equol producers. In adjusted linear regression models, equol producers had significantly lower mean dense tissue area (32.8 vs. 37.7 cm(2), P = 0.03) and lower mean percent breast density (32% vs. 35%, P = 0.03) than nonproducers. Significant inverse associations of isoflavone intake with dense area and percent density were apparent, but only in equol producers (interaction P = 0.05 for both).

Although these findings warrant confirmation in a larger sample, they offer a possible explanation for the inconsistent findings about soy intake and breast density and possibly breast cancer risk as well. The findings further suggest the importance of identifying factors that influence equol-producing status and exploring appropriate targeting of interventions.

S-Equol and Dietary Factors

S-(-)equol, an intestinally derived metabolite of the soy isoflavone daidzein, is proposed to enhance the efficacy of soy diets. Setchell et al. (2013) performed a comprehensive dietary analysis of 143 macro- and micro-nutrients in 159 healthy adults to determine whether the intake of specific nutrients favors equol production. Three-day diet records were collected and analyzed using Nutrition Data System for Research software and S-(-)equol was measured in urine by mass spectrometry.

Equol producers accounted for 29.6% of participants. No significant differences were observed for total protein, carbohydrate, fat, saturated fat, or fiber intakes between equol producers and nonproducers. However, principal component analysis revealed differences in several nutrients, including higher intakes of polyunsaturated fatty acids (P = 0.039), maltose (P = 0.02), and vitamins A (P = 0.01) and E (P = 0.035) and a lower intake of total cholesterol (P = 0.010) in equol producers.

Subtle differences in some nutrients may influence the ability to produce equol.

S-Equol and Dietary Factors; Fats

The soy isoflavones, daidzein and genistein, and the lignans, matairesinol and secoisolariciresinol, are phytoestrogens metabolized extensively by the intestinal microflora. Considerable important evidence is already available that shows extensive interindividual variation in isoflavone metabolism. There was a 16-fold variation in total isoflavonoid excretion in urine after the high-isoflavone treatment period. The variation in urinary equol excretion was greatest (664-fold), and subjects fell into two groups: poor equol excretors and good equol excretors (36%). A significant negative correlation was found between the proportion of energy from fat in the habitual diet and urinary equol excretion (r = -0.55; p = 0.012). Good equol excretors consumed less fat as percentage of energy than poor excretors (26 +/- 2.3% compared with 35 +/- 1.6%, p < 0.01) and more carbohydrate as percentage of energy than poor excretors (55 +/- 2.9% compared with 47 +/- 1.7%, p < 0.05).

It is suggested that the dietary fat intake decreases the capacity of gut microbial flora to synthesize equol (Rowland et al., 2000).

Isoflavones and Fermented Soy Foods

Serum concentrations of total isoflavones after 1–4 hours were significantly higher in the aglycone-rich fermented soybeans (Fsoy) group than in the glucoside-rich non-fermented soybeans (Soy) group. The Fsoy group showed significantly higher maximum concentration (Cmax: 2.79 ± 0.13 vs 1.74 ± 0.13 µmol L(-1) ) and area under the curve (AUC(0-24 h) : 23.78 ± 2.41 vs 19.95 ± 2.03 µmol day L(-1) ) and lower maximum concentration time (Tmax: 1.00 ± 0.00 vs 5.00 ± 0.67 h) compared with the Soy group. The cumulative urinary excretion of total isoflavones after 2 hours was significantly higher in the Fsoy group than in the Soy group. Individual isoflavones (daidzein, genistein and glycitein) showed similar trends to total isoflavones. Equol (a metabolite from daidzein) did not differ between the two groups.

The results of this study demonstrated that the isoflavones of aglycone-rich Fsoy were absorbed faster and in greater amounts than those of glucoside-rich Soy in postmenopausal Japanese women (Okabe et al., 2011).

Phytoestrogens and Breast Cancer; ER+/ER-, ER α /ER β

Dietary-derived Anti-angiogenic Compounds

Consumption of a plant-based diet can prevent the development and progression of chronic diseases that are associated with extensive neovascularization; however, little is known about the mechanisms. To determine whether prevention might be associated with dietary-derived angiogenesis inhibitors, the urine of healthy human subjects consuming a plant-based diet was fractionated and the fractions examined for their ability to inhibit the proliferation of vascular endothelial cells.

The isoflavonoid genistein was the most potent, and inhibited endothelial cell proliferation and in vitro angiogenesis at concentrations giving half-maximal inhibition of 5 and 150 microM, respectively. Genistein concentrations in urine of subjects consuming a plant-based diet are in the micromolar range, while those of subjects consuming a traditional Western diet are lower by a factor of > 30. The high excretion of genistein in urine of vegetarians and in addition to these results suggest that genistein may contribute to the preventive effect of a plant-based diet on chronic diseases, including solid tumors, by inhibiting neovascularization.

Thus, genistein may represent a member of a new class of dietary-derived anti-angiogenic compounds (Fotsis et al., 1993).

ERβ as a Down-regulator of ER+ Breast Cancer

The estrogen receptor (ER) isoform known as ERβ has become the focus of intense investigation as a potential drug target. The existence of clear-cut differences in ERβ and ERα expression suggests that tissues could be differentially targeted with ligands selective for either isoform (Couse et al., 1997; Enmark et al., 1997). In particular, the fact that ER β is widely expressed but not the primary estrogen receptor in, for example, the uterus (where estrogenic effects are mediated via ERα) (Harris, Katzenellenbogen, & Katzenellenbogen, 2002) opens up the possibility of targeting other tissues while avoiding certain classical estrogenic effects.

A major advance toward understanding how some phytoestrogens achieve modest ERβ selectivity was the X-ray structure determination of the ERβ ligand binding domain (LBD) complexed with genistein (GEN) (Pike et al., 1999), a 40-fold ERβ-selective ligand (Harris et al., 2002). This study clearly showed that there are only two residue substitutions in close proximity to GEN: ERα Leu384 is replaced by ER β Met336, and ERα Met421 is replaced by ER β Ile373.

ERbeta works as counter partner of ERalpha through inhibition of the transactivating function of ERalpha by heterodimerization, distinct regulation on several specific promoters by ERalpha or ERbeta, and ERbeta-specific regulated genes which are probably related to its anti-proliferative properties. Epidemiological studies of hormone replacement therapy and isoflavone (genistein) consumption indicate the possible contribution of ERbeta-specific signaling in breast cancer prevention. A selective estrogen receptor modulator, which works as an antagonist of ERalpha and an agonist of ERbeta, may be a promising chemo-preventive treatment (Saji, Hirose, & Toi, 2005).

Genistein and Apoptosis

The association between consumption of genistein containing soybean products and lower risk of breast cancer suggests a cancer chemo-preventive role for genistein. Consistent with this suggestion, exposing cultured human breast cancer cells to genistein inhibits cell proliferation, although this is not completely understood. To better understand how genistein works, the ability of genistein to induce apoptosis was compared in phenotypically dissimilar MCF-7 and MDA-MB-231 human breast cancer cells that express the wild-type and mutant p53 gene, respectively.

After 6 days of incubation with 50 microM genistein, MCF-7, but not MDA-MB-231 cells, showed morphological signs of apoptosis. Marginal proteolytic cleavage of poly-(ADP-ribose)-polymerase and significant DNA fragmentation were also detected in MCF-7 cells.

In elucidating these findings, it was determined that after 2 days of incubation with genistein, MCF-7, but not MDA-MB-231 cells, had significantly higher levels of p53. Accordingly, the expression of certain proteins modulated by p53 was also studied. Levels of p21 increased in both of the genistein-treated cell lines, suggesting that p21 gene expression was activated but in a p53-independent manner; whereas no significant changes in levels of the pro-apoptotic protein, Bax, were found. In MCF-7 cells, levels of the anti-apoptotic protein, Bcl-2, decreased slightly at 18–24 hours but then increased considerably after 48 hours. Hence, the Bax:Bcl-2 ratio initially increased but later decreased.

Data suggests that at the concentration tested, MCF-7 cells, in contrast to MDA-MB-231 cells, were sensitive to the induction of apoptosis by genistein. However, the roles of Bax and Bcl-2 are unclear (Xu & Loo, 2001).

Genistein Derivatives and Breast Cancer Inhibition

Genistein binds to estrogen receptors and stimulates growth at concentrations that would be achieved by a high soy diet, but inhibits growth at high experimental concentrations.

The estrogen receptor (ER) is a major target for the treatment of breast cancer cells. Genistein, a soy isoflavone, possesses a structure similar to estrogen and can both mimic and antagonize estrogen effects although at high concentrations it inhibits breast cancer cell proliferation. Hence, to enhance the anti-cancer activity of Genistein at lower concentrations, seven structurally modified derivatives of Genistein based on the structural requirements for an optimal anti-cancer effect were synthesised. Among those seven, three derivatives showed high anti-proliferative activity with IC(50) levels in the range of 1-2.5 µM, i.e., at much lower concentrations range than Genistein itself, in three ER-positive breast cancer cell lines (MCF-7, 21PT and T47D) studied. In our analysis, we noticed that at IC(50) concentrations, the MA-6, MA-8 and MA-19 Genistein derivatives induced apoptosis, inhibited ER-α messenger RNA expression and increased the ratio of ER-β to ER-α levels in a manner comparable to that of the parent compound Genistein.

Of note, these three modified Genistein derivatives exerted their effects at concentrations 10–15 times lower than the parent compound, decreasing the likelihood of significant ER- α pathway activation, which has been a concern for Genistein. Hence these compounds might play a useful role in breast cancer chemoprevention (Marik et al., 2011).

Genistein and ER α

To determine the effects of low-dose, long-term genistein exposure MCF-7 breast cancer cells were cultured in 10nM genistein for 10-12 weeks and investigated whether or not this long-term genistein treatment (LTGT) altered the expression of estrogen receptor alpha (ERalpha) and the activity of the PI3-K/Akt signaling pathway. This is known to be pivotal in the signaling of mitogens such as oestradiol (E(2)), insulin-like growth factor-1 (IGF-1) and epidermal growth factor (EGF). LTGT significantly reduced the growth promoting effects of E(2) and increased the dose-dependent growth-inhibitory effect of the PI3-K inhibitor, LY 294002, compared to untreated control MCF-7 cells.

This was associated with a significant decreased protein expression of total Akt and phosphorylated Akt but not ERalpha. Rapamycin, an inhibitor of one of the downstream targets of Akt, mammalian target of rapamycin (mTOR), also dose-dependently inhibited growth but the response to this drug was similar in LTGT and control MCF-7 cells. The protein expression of liver receptor homologue-1 (LRH1), an orphan nuclear receptor implicated in tumorigenesis was not affected by LTGT.

These results show that LTGT results in a down-regulation of the PI3-K/Akt signaling pathway and may be a mechanism through which genistein could offer protection against breast cancer (Anastasius et al., 2009).

Genistein and ER+/ER-

Genistein was found to cause a dose-dependent growth inhibition of the two hormone-sensitive cell lines T47D and ZR75.1 and of the two hormone-independent cell lines MDAMB-231 and BT20. Flow cytometric analysis of cells treated for 4 days with 15 and 30 M genistein showed a dose-dependent accumulation in the G2M phase of the cell-cycle. At the highest tested concentration, there was a 7-fold increase in the percentage of cells in G2M (63%) with respect to the control (9%) in the case of T47D cells and a 2.4-fold increase in the case of BT20. An intermediate 4-fold accumulation was observed in the case of MDAMB-231 and ZR75.1. The G2M arrest was coupled with a parallel depletion of the G0/G1 phase.

To understand the mechanism of action underlying the block in G2M induced by genistein, Cappelletti et al. (2000) investigated the expression and the activity of cyclins and of cyclin-dependent kinases specifically involved in the G2M transition. As expected, p34cdc-2 expression, monitored by Western blotting, was unaffected by genistein treatment in all cell lines. With the exception of the T47D cell line, we revealed an increase in the tyrosine phosphorylated form of p34, suggesting an inactivation of the p34cdc-2 catalytic activity consequent to treatment of cells with genistein. In fact, immunoprecipitates from genistein-treated MDAMB-231 and BT20 cells displayed a 4-fold decrease in kinase activity evaluated using the histone H1 as substrate.

Conversely, no variation in kinase activity was observed between treated and untreated ZR75.1 cells despite the increase in p34 phosphorylation. In cells treated with 30 M genistein, cyclin B1 (p62) increased 2.8-,8-and 103-fold, respectively, in BT20, MDAMB-231, and ZR75.1 cells, suggesting an accumulation of the p62, which is instead rapidly degraded in cycling cells. No effects were observed on cyclin expression in T47D cells.

We therefore conclude that genistein causes a G2M arrest in breast cancer cell lines, but that such growth arrest is not necessarily coupled with deregulation of the p34cdc-2/cyclin B1 complex only in all of the studied cell lines.

Genistein and ER+/ER-; MDR

Genistein is a potent inhibitor of the growth of the human breast carcinoma cell lines, MDA-468 (estrogen receptor negative), and MCF-7 and MCF-7-D-40 (estrogen receptor positive) (IC50 values from 6.5 to 12.0 µg/ml). The presence of the estrogen receptor is not required for the isoflavones to inhibit tumor cell growth (MDA-468 vs MCF-7 cells). In addition, the effects of genistein and biochanin A are not attenuated by over expression of the multi-drug resistance gene product (MCF-7-D40 vs MCF-7 cells (Peterson et al., 1991).

Studies have shown that genistein exerts multiple suppressive effects on both estrogen receptor positive (ER+) as well as estrogen receptor negative (ER-) human breast carcinoma lines suggesting that the mechanisms of these effects may be independent of ER pathways.

In the present study however Shao et al. (2000) provide evidence that in the ER+ MCF-7, T47D and 549 lines but not in the ER-MDA-MB-231 and MDA-MB-468 lines both presumed 'ER-dependent' and 'ER-independent' actions of genistein are mediated through ER pathways. Genistein's anti-proliferative effects are estrogen dependent in these ER+ lines, being more pronounced in estrogen-containing media and in the presence of exogenous 17-beta estradiol. Genistein also inhibits the expression of ER-downstream genes including pS2 and TGF-beta in these ER+ lines and this inhibition is also dependent on the presence of estrogen. Genistein inhibits estrogen-induced protein tyrosine kinase (PTK) activity. Genistein is only a weak transcriptional activator and actually decreases ERE-CAT levels induced by 17-beta estradiol in the ER+ lines.

Genistein also decreases steady state ER mRNA only in the presence of estrogen in the ER+ lines thereby manifesting another suppression of and through the ER pathway. Their observations resurrect the hypothesis that genistein functions as a 'good estrogen' in ER+ breast carcinomas. Since chemo-preventive effects of genistein would be targeted to normal ER-positive ductal-lobular cells of the breast, this 'good estrogen' action of genistein is most relevant to our understanding of chemoprevention.

Genistein and Concentration

The anti-proliferative activity of the isoflavones daidzein and genistein were investigated in three breast cancer cell lines with different patterns of estrogen receptor (ER) and c erbB 2 protein expression (ERα positive MCF 7 cells, c erbB 2 positive SK BR 3 cells and ERα/c erbB 2 positive ZR 75 1). After treatment at various concentrations (1 200 µM for 72 hours), the effect of daidzein and genistein on the proliferation of different cell types varied; these effects were found to be associated with ERα and c erbB 2 expression. Daidzein and genistein exhibited biphasic effects (stimulatory or inhibitory) on proliferation and ERα expression in MCF 7 cells. Although 1 µM daidzein significantly stimulated cell growth, ERα expression was unaffected. However, genistein showed marked increases in proliferation and ERα expression after exposure to <10 µM genistein.

Notably, the inhibition of cell proliferation by 200 µM genistein was greater compared to that by daidzein at the same concentration. Daidzein and genistein significantly inhibited proliferation of SK BR 3 and ZR 75 1 cells in a dose-dependent manner. In addition, ERα and c erbB 2 expression was reduced by daidzein and genistein in both SK BR 3 and ZR 75 1 cells in a dose-dependent manner. However, the effect of genistein was greater compared to that of daidzein.

In conclusion, the isoflavones daidzein and genistein showed anti breast cancer activity, which was associated with expression of the ERα and c erbB 2 receptors (Choi et al., 2013).

ER- α / ER β Receptors

Isoflavones are phytoestrogens that have been linked to both beneficial as well as adverse effects in relation to cell proliferation and cancer risks. The mechanisms that could be involved in this dualistic mode of action were investigated. One mechanism relates to the different ultimate cellular effects of activation of estrogen receptor (ER) α, promoting cell proliferation, and of ERβ, promoting apoptosis, with the major soy isoflavones genistein and daidzein activating especially ERβ.

A second mode of action includes the role of epigenetics, including effects of isoflavones on DNA methylation, histone modification and miRNA expression patterns. The overview presented reveals that we are only at the start of unraveling the complex underlying mode of action for effects of isoflavones, both beneficial or adverse, on cell proliferation and cancer risks. It is evident that whatever model system will be applied, its relevance to human tissues with respect to ERα and ERβ levels, co-repressor and co-activator characteristics as well as its relevance to human exposure regimens, needs to be considered and defined (Rietjens et al., 2013).

Genistein and ER+/ER-, ER- α / ER β Receptors

A novel mechanism of adipokine, adiponectin (APN) -mediated signaling that influences mammary epithelial cell proliferation, differentiation, and apoptosis to modify breast cancer risk has been identified. It was demonstrated that early dietary exposure to soy protein isolate induced mammary tissue APN production without corresponding effects on systemic APN levels. In estrogen receptor (ER)-negative MCF-10A cells, recombinant APN promoted lobuloalveolar differentiation by inhibiting oncogenic signal transducer and activator of transcription 3 activity.

In ER-positive HC11 cells, recombinant APN increased ERβ expression, inhibited cell proliferation, and induced apoptosis. Using the estrogen-responsive 4X-estrogen response element promoter-reporter construct to assess ER transactivation and small interfering RNA targeting of ERα and ERβ, Rahal et al. (2011) show that APN synergized with the soy phytoestrogen genistein to promote ERβ signaling in the presence of estrogen (17β-estradiol) and ERβ-specific agonist 2,3-bis(4-hydroxyphenyl)-propionitrile and to oppose ERα signaling in the presence of the ERα-specific agonist 4,4',4'-(4-propyl-(1H)-pyrazole-1,3,5-triyl)trisphenol.

The enhancement of ERβ signaling with APN + genistein co-treatments was associated with induction of apoptosis, increased expression of pro-apoptotic/prodifferentiation genes (Bad, p53, and Pten), and decreased anti-apoptotic (Bcl2 and survivin) transcript levels. These results suggest that mammary-derived APN can influence adjacent epithelial function by ER-dependent and ER-independent mechanisms that are consistent with reduction of breast cancer risk and suggest local APN induction by dietary factors as a targeted approach for promotion of breast health.

Genistein and Non-breast Cancer

Genistein Concentrations; Endometrial Cancer

The influence of two phytoestrogens (Genistein and Daidzein) on estrogen-related receptor-α in endometrial cancer cell line Ishikawa was investigated on the proliferation of the cells in this cell line. Ishikawa cells were incubated with different concentrations of Genistein and Daidzein (40, 20, 10, 5 µmol/L) for 24 hours or 48 hours, followed by Real-Time PCR for analyzing the expression of ERR-α mRNA in the cell line. MTT assay was then performed to evaluate the proliferation of Ishikawa cells.

The expression level of ERR-α mRNA in Ishikawa cells was higher than that of the control group after being dealt for 24 hours or 48 hours with Genistein, and the concentration 20 µmol/L was most effective. Nevertheless, this up-regulation was blocked when the cells were treated with 40 µmol/L Genistein. Lower concentration (5, 10 µmol/L) Genistein had depressant effect on proliferation of the cells, while higher concentrations (20, 40 µmol/L) had stimulant effect. After being treated with different concentrations of Daidzein, the expression of ERR- α mRNA in all experimental groups was significantly higher than that in the control group. In the 24 hour group, the concentration 40 µmol/L had most obvious effect; but in the 48 hour group, the concentration 20 µmol/L had most obvious effect, and this up-regulation was blocked when the concentration was elevated to 40 µmol/L.

Noticeably, all concentrations of Daidzein had depressant effect on the proliferation of Ishikawa cells in both 24 hour and 48 hour groups. In the 24 hour group, lower concentrations were more effective, but in the 48 hour group, concentration showed no significant effect. In lower concentrations, both Genistein and Daidzein have up-regulation effect on the expression of ERR-α, and block the proliferation of Ishikawa cells; but in higher concentrations, the up-regulation effect on ERR-α mRNA expression by these two phytoestrogens is not obvious. Genistein stimulates the proliferation of lshikawa cells in higher concentrations, while Daidzein suppresses the proliferation, especially in lower concentrations (Xin et al., 2009).

Genistein and VEGF; Ovarian Cancer

Genistein represses NF-kappaB (NF-κB), a pro-inflammatory transcription factor, and inhibits pro-inflammatory cytokines such as TNF-α and IL-6 in epithelial ovarian cancer. Additionally, it has been shown to stabilize p53 protein, sensitize TRAIL (TNF receptor apoptosis-inducing ligand) induce apoptosis, and prevent or delay chemotherapy-resistance. Recent studies further indicate that genistein potently inhibits VEGF production and suppresses ovarian cancer cell metastasis in vitro.

Based on widely published in vitro and mouse-model data, some anti-inflammatory phytochemicals appear to exhibit activity in modulating the tumor microenvironment. Specifically, apiegenin, baicalein, curcumin, EGCG, genistein, luteolin, oridonin, quercetin, and wogonin repress NF-kappaB (NF-κB, a pro-inflammatory transcription factor) and inhibit pro-inflammatory cytokines such as TNF-α and IL-6. Recent studies further indicate that apigenin, genistein, kaempferol, luteolin, and quercetin potently inhibit VEGF production and suppress ovarian cancer cell metastasis in vitro. Lastly, oridonin and wogonin were suggested to suppress ovarian CSCs as is reflected by down-regulation of the surface marker EpCAM (Chen, Michael, & Butler-Manuel, 2012).

Renal Cell Carcinoma, Prostate Cancer; Radiotherapy

The KCI-18 RCC cell line was generated from a patient with papillary renal cell carcinoma. Tumor cells metastasize from the primary renal tumor to the lungs, liver and mesentery mimicking the progression of RCC in humans. Treatment of established kidney tumors with genistein demonstrated a tendency to stimulate the growth of the primary kidney tumor and increase the incidence of metastasis to the mesentery lining the bowel. In contrast, when given in conjunction with kidney tumor irradiation, genistein significantly inhibited the growth and progression of established kidney tumors. These findings confirm the potentiation of radiotherapy by genistein in the orthotopic RCC model as previously shown in orthotopic models of prostate cancer. These studies in both RCC and prostate tumor models demonstrate that the combination of genistein with primary tumor irradiation is a more effective and safer therapeutic approach as the tumor growth and progression are inhibited both in the primary and metastatic sites (Gilda et al., 2007).

Cell-cycle Arrest

Genistein treatment increased Wee1 levels and decreased phospho-Wee1 (Ser 642). Moreover, genistein substantially decreased the Ser473 and Thr308 phosphorylation of Akt and up-regulated PTEN expression. Down-regulation of PTEN by siRNA in genistein-treated cells increased phospho-Wee1 (Ser642), whereas it decreased phospho-Cdc2 (Tyr15), resulting in decreased G2/M cell-cycle-arrest. Therefore, induction of G2/M cell-cycle arrest by genistein involved up-regulation of PTEN (Liu et al., 2013).

Cancer Stem Cells (CSCs)

Cancer stem cells (CSCs) are cells that exist within a tumor with a capacity for self-renewal and an ability to differentiate, giving rise to heterogeneous populations of cancer cells. These cells are increasingly being implicated in resistance to conventional therapeutics and have also been implicated in tumor recurrence. Several cellular signaling pathways including Notch, Wnt, phosphoinositide-3-kinase-Akt-mammalian target of rapamycin pathways, and known markers such as CD44, CD133, CD166, ALDH, etc. have been associated with CSCs.

Here, we have reviewed our current understanding of self-renewal pathways and factors that help in the survival of CSCs with special emphasis on those that have been documented to be modulated by well characterized natural agents such as curcumin, sulforaphane, resveratrol, genistein, and epigallocatechin gallate (Dandawate et al., 2013).

Genistein and Sex Hormone-binding Globulin (SHBG)

Studies have indicated a correlation between a high level of urinary lignans and isoflavonoid phytoestrogens, particularly genistein, and a low incidence of hormone-dependent cancers, such as breast and prostate cancer. Previously it has been observed that a vegetarian diet is associated with high plasma levels of sex hormone-binding globulin (SHBG), reducing clearance of sex hormones and probably risk of breast and prostate cancer. In the present study we investigated the in vitro effect of genistein on the production of SHBG by human hepatocarcinoma (Hep-G2) cells in culture and its effect on cell proliferation.

It has additionally been found that genistein not only significantly increases the SHBG production by Hep-G2 cells, but also suppresses the proliferation of those cancer cells already at a stage when SHBG production continues to be high. It is hence concluded that, in addition to the lignan enterolactone, the most abundant urinary isoflavonoid genistein stimulates SHBG production and inhibits Hep-G2 cancer cell proliferation (Mousavi et al., 1993).

Insulin-like Growth Factor-1 (IGF-1); Prostate Cancer

Elevated levels of insulin-like growth factor-1 (IGF-1) are associated with an increased risk of several different cancers, including prostate cancer. Inhibition of IGF-1 and the downstream signaling pathways mediated by the activation of the IGF-1 receptor (IGF-1R) may be involved in inhibiting prostate carcinogenesis. Genistein treatment caused a significant inhibition of IGF-1-stimulated cell growth. Flow cytometry analysis revealed that genistein significantly decreased the number of IGF-1-stimulated cells in the G0/G1 phase of the cell-cycle. In IGF-1-treated cells, genistein effectively inhibited the phosphorylation of IGF-1R and the phosphorylation of its downstream targets, such as Src, Akt, and glycogen synthase kinase-3β (GSk-3β). IGF-1 treatment decreased the levels of E-cadherin but increased the levels of β-catenin and cyclin D1.

However, genistein treatment greatly attenuated IGF-1-induced β-catenin signaling that correlated with increasing the levels of E-cadherin and decreasing cyclin D1 levels in PC-3 cells. In addition, genistein inhibited T-cell factor/lymphoid enhancer factor (TCF/LEF)-dependent transcriptional activity. These results showed that genistein effectively inhibited cell growth in IGF-1-stimulated PC-3 cells, possibly by inhibiting downstream of IGF-1R activation (Lee et al., 2012).

Sex Hormone-binding Globulin (SHBG); Hepatoma

Sex hormone-binding globulin (SHBG) is the main transport binding protein for sex steroid hormones in plasma and regulates their accessibility to target cells. Plasma SHBG is secreted by the liver under the control of hormones and nutritional factors. In the human hepatoma cell line (HepG2), thyroid and estrogenic hormones, and a variety of drugs including the anti-estrogen tamoxifen, the phytoestrogen, genistein and mitotane (Op'DDD) increase SHBG production and SHBG gene promoter activity. In contrast, monosaccharides (glucose or fructose) effectively decrease SHBG expression by inducing lipogenesis, which reduces hepatic HNF-4alpha levels, a transcription factor that plays a critical role in controlling the SHBG promoter. Interestingly, diminishing hepatic lipogenesis and free fatty acid liver biosynthesis also appear to be associated with the positive effects of thyroid hormones and PPARgamma antagonists on SHBG expression.

This mechanism provides a biological explanation for why SHBG is a sensitive biomarker of insulin resistance and the metabolic syndrome, and why low plasma SHBG levels are a risk factor for developing hyperglycemia and type 2 diabetes, especially in women (Pugeat et al., 2009).

Cancer: Pancreatic

Pancreatic cancer remains the fourth most common cause of cancer related death in the United States. Therefore, novel strategies for the prevention and treatment are urgently needed. Genistein is a prominent isoflavonoid found in soy products and has been proposed to be responsible for lowering the rate of pancreatic cancer in Asians. However, the molecular mechanism(s) by which genistein elicits its effects on pancreatic cancer cells has not been fully elucidated.

Wang et al., (2006) have previously shown that genistein induces apoptosis and inhibits the activation of nuclear factor kappaB (NF-kappaB) pathway. Moreover, Notch signaling is known to play a critical role in maintaining the balance between cell proliferation, differentiation and apoptosis, and thereby may contribute to the development of pancreatic cancer. Hence, in our study, they investigated whether there is any cross talk between Notch and NF-kappaB during genistein-induced apoptosis in BxPC-3 pancreatic cancer cells. They found that genistein inhibits cell growth and induces apoptotic processes in BxPC-3 pancreatic cancer cells.

This was partly due to inhibition of Notch-1 activity. BxPC-3 cells transfected with Notch-1 cDNA showed induction of NF-kappaB activity, and this was inhibited by genistein treatment. From these results, we conclude that the inhibition of Notch-1 and NF-kappaB activity and their cross talk provides a novel mechanism by which genistein inhibits cell growth and induces apoptotic processes in pancreatic cancer cells.

References

Anastasius N, Boston S, Lacey M, Storing N, Whitehead SA. (2009). Evidence that low-dose, long-term genistein treatment inhibits oestradiol-stimulated growth in MCF-7 cells by down-regulation of the PI3-kinase/Akt signaling pathway. J Steroid Biochem Mol Biol, 116(1-2):50-55.


Cappelletti V, Fioravanti L, Miodini P, Di Fronzo G J. (2000). Genistein blocks breast cancer cells in the G2M phase of the cell-cycle. Cell. Biochem, 79(4):594-600. doi: 10.1002/1097-4644(20001215)79:4<594::AID-JCB80>3.0.CO;2-4.


Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.


Choi EJ, Kim GH. (2013). Anti-proliferative activity of daidzein and genistein may be related to ERα /c-erbB-2 expression in human breast cancer cells. Mol Med Rep, 7(3):781-4. doi: 10.3892/mmr.2013.1283.


Couse JF, Lindzey J, Grandien K, Gustafsson JA, Korach KS. (1997). Tissue distribution and quantitative analysis of estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) messenger ribonucleic acid in the wild-type and ERalpha-knockout mouse. Endocrinology, 138(1997):4613–4621


Dandawate P, Padhye S, Ahmad A, Sarkar FH. (2013). Novel strategies targeting cancer stem cells through phytochemicals and their analogs. Drug Deliv Transl Res, 3(2):165-182.


Enmark E, Peltohuikko M, Grandien K, et al. (1997). Human estrogen receptor beta-gene structure, chromosomal localization, and expression pattern. J. Clin. Endocrinol. Metab, 82(1997):4258–4265.


Fotsis T, Pepper M, Adlercreutz H, et al. (1993). Genistein, a dietary-derived inhibitor of in vitro angiogenesis. Proc Natl Acad Sci, 90(7):2690-4.


Harris HA, Albert LM, Leathurby Y, et al. (2002). Evaluation of an estrogen receptor- β agonist in animal models of human disease. Endocrinology, 144(2003):4241–4249


Harris HA, Katzenellenbogen JA, Katzenellenbogen BS. (2002). Characterization of the biological roles of the estrogen receptors, ER alpha and ER beta, in estrogen target tissues in vivo through the use of an ER alpha-selective ligand. Endocrinology, 143(2002):4172–4177.


Hillman GG, Wang Y, Che M, et al. (2007). Progression of renal cell carcinoma is inhibited by genistein and radiation in an orthotopic model. BMC Cancer, 7:4. doi:10.1186/1471-2407-7-4.


Lee J, Ju J, Park S, et al. (2012). Inhibition of IGF-1 Signaling by Genistein: Modulation of E-Cadherin Expression and Down-regulation of β -Catenin Signaling in Hormone Refractory PC-3 Prostate Cancer Cells. Nutrition and Cancer, 64(1). doi:10.1080/01635581.2012.630161


Liu YL, Zhang GQ, Yang Y, et al. (2013). Genistein Induces G2/M Arrest in Gastric Cancer Cells by Increasing the Tumor Suppressor PTEN Expression. Nutr Cancer.


Marik R, Allu M, Anchoori R, et al. (2011). Potent genistein derivatives as inhibitors of estrogen receptor alpha-positive breast cancer. Cancer Biol Ther, 11(10):883-92.


Mousavi Y, Adlercreutz H. (1993). Genistein is an effective stimulator of sex hormone-binding globulin production in hepatocarcinoma human liver cancer cells and suppresses proliferation of these cells in culture. Steroids, 58(7):301-4.


Okabe Y, Shimazu T, Tanimoto H. (2011). Higher bioavailability of isoflavones after a single ingestion of aglycone-rich fermented soybeans compared with glucoside-rich non-fermented soybeans in Japanese postmenopausal women. J Sci Food Agric, 91(4):658-63. doi: 10.1002/jsfa.4228.


Peterson G, Barnes S. (1991). Genistein inhibition of the growth of human breast cancer cells: independence from estrogen receptors and the multi-drug resistance gene. Biochemical and Biophysical Research Communications, 179(1):661-667. doi:10.1016/0006-291X(91)91423-A.


Pike ACW, Brzozowski AM, Hubbard RE, et al. (1999). Structure of the ligand-binding domain of oestrogen receptor beta in the presence of a partial agonist and a full antagonist. EMBO J, 18(1999): 4608–4618


Pugeat M, Nader N, Hogeveen K, et al. (2010). Sex hormone-binding globulin gene expression in the liver: Drugs and the metabolic syndrome. Mol Cell Endocrinol, 316(1):53-9. doi: 10.1016/j.mce.2009.09.020.


Rahal OM, Simmen RC. (2011). Paracrine-Acting Adiponectin Promotes Mammary Epithelial Differentiation and Synergizes with Genistein to Enhance Transcriptional Response to Estrogen Receptor β Signaling. Endocrinology, 152(9):3409-21. doi: 10.1210/en.2011-1085.


Rietjens IM, Sotoca AM, Vervoort J, Louisse J. (2013). Mechanisms underlying the dualistic mode of action of major soy isoflavones in relation to cell proliferation and cancer risks. Mol Nutr Food Res, 57(1):100-13. doi: 10.1002/mnfr.201200439.


Rowland IR, Wiseman H, Sanders TA, Adlercreutz H, Bowey EA. (2000). Interindividual variation in metabolism of soy isoflavones and lignans: influence of habitual diet on equol production by the gut microflora. Nutr Cancer, 36(1):27-32.


Saji S, Hirose M, Toi M. (2005). Clinical significance of estrogen receptor beta in breast cancer. Cancer Chemother Pharmacol, 56(1):21-6.


Setchell KD, Brown NM, Summer S, et al. (2013). Dietary Factors Influence Production of the Soy Isoflavone Metabolite S-(-)Equol in Healthy Adults. J Nutr.


Shao ZM, Shen ZZ, Fontana JA, Barsky SH. (2000). Genistein's ER-dependent and independent actions are mediated through ER pathways in ER-positive breast carcinoma cell lines. Anti-cancer Res, 20(4):2409-16.


Stark A, Madar Z. (2002). Phytoestrogens: a review of recent findings. J Pediatr Endocrinol Metab, 15(5):561-72.


Tseng M, Byrne C, Kurzer MS, Fang CY. (2013). Equol-producing status, isoflavone intake, and breast density in a sample of u.s. Chinese women. Cancer Epidemiol Biomarkers Prev, 22(11):1975-83. doi: 10.1158/1055-9965.EPI-13-0593.


Xin Z, Siji L, Yan D, Weijuan X, Jie S, Qianyu W. (2009). Influence of Genistein and Daidzein on estrogen-related receptor- α in an Endometrial Carcinoma Cell Line. Tong Ji Da Xue Xue Bao (Yi Xue Ban), 30(4): 12-17.


Xu J, Loo G. (2001). Different effects of genistein on molecular markers related to apoptosis in two phenotypically dissimilar breast cancer cell lines. Journal of Cellular Biochemistry, 82(1), 78-88.

Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. (2006) Inhibition of nuclear factor kappab activity by genistein is mediated via Notch-1 signaling pathway in pancreatic cancer cells. Int J Cancer. 2006 Apr 15;118(8):1930-6.

Mangiferin

Cancer: Breast, gliomas, colon

Action: Chemo-preventive agent, chelating agent

Mangiferin, a glucosylxanthone isolated from Anemarrhena asphodeloides, is an efficient iron chelator, therefore preventing the generation of hydroxyl radical in Fenton-type reactions. Numerous published in vitro and in vivo pharmacological studies have demonstrated many other activities of mangiferin: analgesic, anti-diabetic, anti-sclerotic, anti-microbial and anti-viral., cardio-, hepato-, and neuro-protective, anti-inflammatory, anti-allergic, MAO-inhibiting and memory-improving, as well as radio-protective against X-ray, gamma, and UV radiation. Several studies also indicated its ability to inhibit cancerogenesis and cancer cell growth by apoptosis induction in vitro and in vivo (Matkowski et al., 2013).

Colon Cancer; Pre-neoplastic Lesions

Recent studies have shown that mangiferin has potential as an anti-oxidant, and as an anti-viral agent. The effects of mangiferin in rat colon carcinogenesis induced by the chemical carcinogen, azoxymethane (AOM), were studied. Two experiments were carried out: a short-term   assay to investigate the effects of mangiferin on the development of pre-neoplastic lesions by AOM, aberrant crypt foci (ACF); and a long-term assay for the influence of mangiferin on tumorigenesis induced by AOM. In the short-term assay, 0.1% mangiferin in a diet significantly inhibited the ACF development in rats treated with AOM, compared to rats treated with AOM alone (64.6±22.0 vs. 108.3±43.0).

In the long-term assay, the group treated with 0.1% mangiferin in initiation phase of the experimental protocol had significantly lower incidence and multiplicity of intestinal neoplasms induced by AOM (47.3% and 41.8% reductions of the group treated with AOM alone for incidence and multiplicity, respectively). In addition, the cell proliferation in colonic mucosa was reduced in rats treated with mangiferin (65–85% reductions of the group treated with AOM alone). These results suggest that mangiferin has potential as a naturally-occurring chemo-preventive agent (Yoshimi et al., 2001).

Mangiferin may be used as an effective chemo-preventive agent against breast cancer. Mangiferin, which is a naturally occurring glucosylxanthone, has exhibited promising anti-cancer activities. In this study, the anti-cancer activity of mangiferin was evaluated in breast cancer cell line-based in vitro and in vivo models. Li et al. (2013) showed that mangiferin treatment resulted in decreased cell viability and suppression of metastatic potential in breast cancer cells.

Gliomas

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases which play a key role in invasion, migration, and angiogenesis of astrogliomas and other malignant tumors. Thus, controlling MMPs has been considered an important therapeutic strategy for prevention and/or treatment of gliomas.

Mangiferin inhibits the binding of NF- κB and AP-1 to the MMP-9 promoter and suppresses the PMA-induced phosphorylation of Akt and MAP kinases, which are upstream signaling molecules in MMP-9 expression. Thus, the specific inhibition of MMP-9 by mangiferin may provide a valuable pharmacological tool for treatment of gliomas (Jung et al., 2012,).

References

Jung JS, Jung K, Kim DH, Kim HS. (2012). Selective inhibition of MMP-9 gene expression by mangiferin in PMA-stimulated human astroglioma cells: involvement of PI3K/Akt and MAPK signaling pathways. Pharmacol Res, 66(1):95-103. doi: 10.1016/j.phrs.2012.02.013.


Li H, Huang J, Yang B, et al. (2013). Mangiferin exerts anti-tumor activity in breast cancer cells by regulating matrix metalloproteinases, epithelial to mesenchymal transition, and β -catenin signaling pathway. Toxicol Appl Pharmacol, 272(1):180-90. doi: 10.1016/j.taap.2013.05.011.


Matkowski A, Ku ś P, G-ralska E, Wo ź niak D. (2013). Mangiferin – a bioactive xanthonoid, not only from mango and not just anti-oxidant. Mini Rev Med Chem, 13(3):439-55.


Yoshimi N, Matsunaga K, Katayama M, et al. (2001). The inhibitory effects of mangiferin, a naturally occurring glucosylxanthone, in bowel carcinogenesis of male F344 rats. Cancer Letters, 163(2):163-70. doi:10.1016/S0304-3835(00)00678-9

Artesunate, oral (See also Injectables)

Cancer:
Non-resectable tumors, Retinoblastoma, colon, esophageal., retinoblastoma, ovarian, lung, glioblastoma, MDR, gastric

Action: Anti-cancer

Artesunate is a semisynthetic derivative of the herbal anti-malaria drug artemisinin, which is the active agent from Artemisia annua L. used in traditional Chinese medicine.

Anti-cancer; Canine

The anti-malarial drug artesunate has shown anti-cancer activity in vitro and in preliminary animal experiments, but experience in patients with cancer is very limited. Preclinical studies in dogs indicated morbidity at high dosage levels. The effects of artesunate have been examined in canine cancer cell lines and in canine cancer patients. A safety/efficacy field study with artesunate was conducted in 23 dogs with non-resectable tumors.

Artesunate was administered for 7–385 days at a dosage of 651-1178 (median 922) mg/m(2). No neurological or cardiac toxicity was observed and seven dogs exhibited no adverse effects at all. Fever and haematological/gastrointestinal toxicity, mostly transient, occurred in 16 dogs. Plasma artesunate and DHA levels fell below the limit of detection within 8–12 hours after artesunate administration, while levels after two hours were close to 1 µM. Artesunate produced a long-lasting complete remission in one case of cancer and short-term stabilization of another 7 cases. This study suggests artesunate may be an effective anti-cancer agent in humans (Rutteman, 2013).

Lung Cancer

The exact molecular mechanism by which artesunate induces apoptosis in human lung adenocarcinoma (ASTC-a-1 and A549) cell lines has been examined, and it was found that artesunate induces apoptosis via a Bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells. Artesunate treatment was found to induce ROS-mediated apoptosis in a concentration- and time-dependent fashion accompanying the loss of mitochondrial potential and subsequent release of Smac and AIF indicative of intrinsic apoptosis pathway. Furthermore, although ART treatment did not induce a significant down-regulation of voltage-dependent anion channel 2 (VDAC2) expression and up-regulation of Bim expression, silencing VDAC2 potently enhanced the artesunate-induced Bak activation and apoptosis which were significantly prevented by silencing Bim.

Collectively, our data firstly demonstrate that artesunate induces Bak-mediated caspase-independent intrinsic apoptosis in which Bim and VDAC2 as well as AIF play important roles in both ASTC-a-1 and A549 cell lines, indicating a potential therapeutic effect of artesunate for lung cancer (Zhou, 2012).

Glioblastoma

Trials that include artesunate in cancer therapy are ongoing due to its action as a powerful inducer of oxidative DNA damage, giving rise to formamidopyrimidine DNA glycosylase-sensitive sites and the formation of 8-oxoguanine and 1,N6-ethenoadenine. Oxidative DNA damage was induced in LN-229 human glioblastoma cells dose-dependently and was paralleled by cell death executed by apoptosis and necrosis, which could be attenuated by radical scavengers such as N-acetyl cysteine.

These data indicate that both homologous recombination and nonhomologous end joining are involved in the repair of artesunate-induced DNA double-strand break (DSB). Artesunate provoked a DNA damage response (DDR) with phosphorylation of ATM, ATR, Chk1, and Chk2.

Overall, these data revealed that artesunate induces oxidative DNA lesions and DSB that continuously increase during the treatment period and accumulate until they trigger discoidin domain receptors (DDR) and finally tumor cell death (Berdelle, 2011).

Esophageal Cancer, MDR

The Eca109/ABCG2 cell line was established by transfecting the ABCG2 gene into Eca109 cells. The Eca109/ABCG2 esophageal cancer cells with ABCG2 gene overexpression were resistant to adriamycin (ADM), daunorubicin (DNR) and mitoxantrone (MIT), which indicated that ABCG2 may be associated with drug resistance in esophageal cancer.

Artesunate (ART) exerted profound anti-cancer activity. The mechanism for the reversal of multi-drug resistance by Art in esophageal carcinoma was analyzed using cellular experiments (Liu, Zuo, & Guo, 2013).

Artesunate was found to stop the growth of esophageal cancer cells transplated subcutaneous tumors in nude mice in the G1 stage. It is hence thought that the role of Artesunate against esophageal carcinoma maybe relate to cell-cycle blockage. Artesunate was also found to increase the expression of SMAD3 and TGF-β1, and reduce the expression of CDC25A and CDC25B which may also play a role in its anti-cancer activity.

Retinoblastoma

Zhao et al. (2013) found that the cytotoxic action of artesunate (ART) is specific for Retinoblastoma (RB) cells in a dose-dependent manner, with low toxicity in normal retina cells. ART is more effective in RB than carboplatin with a markedly strong cytotoxic effect on carboplatin-resistant RB cells. RB had higher CD71 levels at the membrane compared to normal retinal cells. ART is a promising drug exhibiting high selective cytotoxicity even against multi-drug-resistant RB cells.

Gastric Cancer

Artesunate has concentration-dependent inhibitory activities against gastric cancer in vitro and in vivo by promoting cell oncosis through an impact of calcium, vascular endothelial growth factor, and calpain-2 expression (Zhou et al., 2013).

Ovarian Cancer

Advanced-stage ovarian cancer (OVCA) has a unifocal origin in the pelvis. Molecular pathways associated with extrapelvic OVCA spread are also associated with metastasis from other human cancers and with overall patient survival. Such pathways represent appealing therapeutic targets for patients with metastatic disease. Artesunate-induced TGF-WNT pathway inhibition impaired OVCA cell migration (Marchion et al., 2013).

Colon Cancer

After colon cancer SW620 cells were treated with different doses of Artemisunate, anchorage independence was studied in soft agar colony formation. Invasiveness was assessed by Boyden chamber, and the protein level of intercellular adhesion molecule-1 (ICAM-1) was detected by Western blot assay. Artemisunate significantly inhibited both the invasiveness and anchorage independence in a dose-dependent manner. The protein level of ICAM-1 was down-regulated as relative to the control group.

Artemisunate could potentially inhibit invasion of the colon carcinoma cell line SW620 by down-regulating ICAM-1 expression (Fan, Zhang, Yao, & Li, 2008).

References

Berdelle N, Nikolova T, Quiros S, Efferth T, Kaina B. (2011). Artesunate Induces Oxidative DNA Damage, Sustained DNA Double-Strand Breaks, and the ATM/ATR Damage Response in Cancer Cells. Mol Cancer Ther, 10(12):2224-33. doi: 10.1158/1535-7163.MCT-11-0534.


Fan, Y, Zhang, YL, Yao, GT, & Li, YK. (2008). Inhibition of Artemisunate on the invasion of human colon cancer line SW620. Lishizzhen Medicine and Materia Medica Research, 19(7), 1740-1741.


Liu, L, Zuo, LF, Guo, JW. (2013). Reversal of Multi-drug resistance by the anti-malaria drug artesunate in the esophageal cancer Eca109/ABCG2 cell line. Oncol Lett, 6(5): 1475–1481. doi: 10.3892/ol.2013.1545i


Marchion DC, Xiong Y, Chon HS, et al. (2013). Gene expression data reveal common pathways that characterize the unifocal nature of ovarian cancer. Am J Obstet Gynecol, S0002-9378(13)00827-2. doi: 10.1016/j.ajog.2013.08.004.


Rutteman GR, Erich SA, Mol JA, et al. (2013). Safety and Efficacy Field Study of Artesunate for Dogs with Non-resectable Tumors. Anti-cancer Res, 33(5):1819-27.


Zhao F, Wang H, Kunda P, et al. (2013). Artesunate exerts specific cytotoxicity in retinoblastoma cells via CD71. Oncol Rep. doi: 10.3892/or.2013.2574.


Zhou C, Pan W, Wang XP, Chen TS. (2012). Artesunate induces apoptosis via a Bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells. J Cell Physiol, 227(12):3778-86. doi: 10.1002/jcp.24086.


Zhou X, Sun WJ, Wang WM, et al. (2013). Artesunate inhibits the growth of gastric cancer cells through the mechanism of promoting oncosis both in vitro and in vivo. Anti-cancer Drugs, 24(9):920-7. doi: 10.1097/CAD.0b013e328364a109.

Green Tea 95% Polyphenols

Cancer: Lung, colorectal., head and neck

Action: Hemoprevention, chemo-preventive effects

Green tea polyphenols include compounds found in fruits and vegetables, particularly green tea Camellia sinensis [(L.) Kuntze].

Chemo-preventive Effects

Polyphenolic compounds in fruits and vegetables have been associated with lower risk of some diseases, including cancer. Recent research has shown that the polyphenolic anti-oxidants in green tea possess cancer chemo-preventive effects (Ahmad et al., 1999).

Green tea polyphenols were found to increase the activities of glutathione peroxidase, catalase, and quinone reductase in small bowel, liver, and lungs, and glutathione S-transferase in small bowel and liver. These can be implicated in relation to the cancer chemo-preventive effects of green tea polyphenols against the induction of tumors in various target organs (Khan et al., 1992).

Head and Neck Cancers

Additionally, green tea polyphenol-induced production of H2O2 may mediate apoptosis and this may also contribute to the growth-inhibitory activities in cancer cells in vitro (Yang et al., 1998).

A synergistic inhibition between green tea (-)-epigallocatechin-3-gallate and EGFR tyrosine kinase inhibitor in head and neck tumor growth was shown (Zhang et al., 2008).

References

Ahmad N, Mukhtar H. (1999). Green Tea Polyphenols and Cancer: Biologic Mechanisms and Practical Implications. Nutrition Reviews, 57(3), 78-83.


Khan SG, Katiyar SK, Agarwal R, Mukhtar H. (1992). Enhancement of Anti-oxidant and Phase II Enzymes by Oral Feeding of Green Tea Polyphenols in Drinking Water to SKH-1 Hairless Mice: Possible Role in Cancer Chemoprevention. Cancer Research, 52(14), 4050-4052.


Yang GY, Liao J, Kim K, Yurkow EJ, Yang CS. (1998). Inhibition of growth and induction of apoptosis in human cancer cell lines by tea polyphenols. Carcinogenesis, 19(4), 611-616.


Zhang X, Zhang H, Tighiouart M, Lee JE, et al. (2008). Synergistic inhibition of head and neck tumor growth by green tea (-)-epigallocatechin-3-gallate and EGFR tyrosine kinase inhibitor. Int. J. Cancer, 123(5):1005–1014

Parthenolide

Cancer:
Myeloma, ovarian adenocarcinoma, breast, acute myelogenous leukemia, epithelial-to-mesenchymal transition (EMT)

Action: Anti-cancer, anti-inflammatory, inhibits NF-κB, promotes apoptosis

Inhibits NF-κB & Promotes Apoptosis

Parthenolide, a sesquiterpene lactone derived from the leaves of feverfew (Tanacetum parthenium (L.)), is considered a main bioactive component of this herb. Feverfew has been used orally or as an infusion for the treatment of migraine, arthritis, fever, and stomachache. Besides its anti-inflammatory and anti-migraine properties, parthenolide also shows anti-cancer activities in a variety of cell lines. It contains an alpha-methylene-gamma-lactone ring and an epoxide moiety which are able to interact with nucleophilic sites of biologically important molecules.

Parthenolide modulates multiple targets, thereby contributing to its various in vitro and in vivo effects. Inhibition of NF-kappaB activity, constitutive in many types of cancers via either interaction with IKK or more directly with the p65 subunit of NF-kappaB, is considered one of the main mechanisms of its action. In addition, inhibition of STAT and MAP kinase activities and the induction of sustained JNK activity as well as p53 activity via influencing MDM2 and HDAC1 levels lead to an increased susceptibility of cancer cells to chemo- and radio- therapy. At the epigenetic level, parthenolide reduces HDAC1 level and, by inhibiting DNMT2 activity, induces global hypomethylation of DNA, which can restore the expressions of some suppressor genes.

Moreover, this compound reduces the cellular level of GSH in cancer cells, followed by ROS accumulation and apoptosis. A unique property of parthenolide is its ability to induce cell death mainly in cancer cells, while sparing healthy ones and it also protects normal cells from UVB and oxidative stress. More remarkably, it seems to have the potential to target some cancer stem cells. Its wide array of biological activity and low toxicity make parthenolide a very promising drug with multi-pharmacological potential, largely dependent on the cellular context (Koprowska et al., 2010).

Multiple Myeloma

It has been shown that parthenolide is a potent anti-MM-CSC agent. Multiple myeloma (MM) is an incurable plasma cell malignancy where nearly all patients succumb to a relapse. The current preclinical models of MM target the plasma cells, constituting the bulk of the tumor, leaving the cancer stem cells to trigger a relapse. It demonstrated preferential toxicity toward MM-CSCs over non-tumorigenic MM cells. Addition of the bone marrow stromal compartment abrogated andrographolide activity while having no effect on parthenolide cytoxicity. It hence has anti-CSC activity in myeloma, suggesting that it has the potential to improve the survival of patients with MM by eliminating the relapse-causing MM-CSCs (Gunn et al., 2011).

Acute Myelogenous Leukemia

Parthenolide (PTL), a naturally occurring small molecule, induces robust apoptosis in primary human acute myelogenous leukemia (AML) cells and blast crisis CML (bcCML) cells while sparing normal hematopoietic cells. Furthermore, analysis of progenitor cells using in vitro colony assays, as well as stem cells using the non-obese diabetic/severe combined immunodeficient (NOD/SCID) xenograft model, show that PTL also preferentially targets AML progenitor and stem cell populations.

Notably, in comparison to the standard chemotherapy drug cytosine arabinoside (Ara-C), PTL is much more specific to leukemia cells. The molecular mechanism of PTL-mediated apoptosis is strongly associated with inhibition of nuclear factor κB (NF-κB), pro-apoptotic activation of p53, and increased reactive oxygen species (ROS) (Guzman, et al., 2005).

PTL is known to be a potent inhibitor of NF-κB (Bork et al., 1997). The mechanism of NF-κB down-regulation appears to occur via inhibition of the IKK complex. Guzman et al. (2005) observed strong inhibition of NF-κB in primary AML cells and speculate that this activity contributes to the efficacy of PTL. However, previous genetic studies using a dominant-negative repressor of NF-κB activity have shown that inhibition of NF-κB alone is not sufficient to mediate the robust cell death observed with PTL. Rather, blockade of the NF-κB pathway appears to sensitize primary AML cells to death and induces a relatively slow spontaneous apoptosis (~50% cell death in 36 h) (Guzman et al., 2002). Similarly, studies by Romano et al. (2000) showed that treatment of primary AML blasts with NF-κB decoy oligonucleotides was not sufficient to induce a strong apoptotic response.

Consequently, PTL must be affecting other pathways relevant to AML-specific survival. One such pathway appears to be mediated by the activity of p53. PTL induced rapid up-regulation of p53 protein with concomitant phosphorylation on serine (Woynarowski & Konopa, 1981).

Ovarian Carcinoma

Results suggest that parthenolide may induce apoptotic cell death in ovarian carcinoma cell lines by activating the mitochondrial pathway and the caspase-8- and Bid-dependent pathways. The apoptotic effect of parthenolide appears to be mediated by the formation of reactive oxygen species and the depletion of GSH. Parthenolide might be beneficial in the treatment of epithelial ovarian adenocarcinoma and combination therapy (Kwak et al., 2013).

Epithelial-to-Mesenchymal Transition (EMT)

Detyrosinated tubulin, a post-translational modification of α-tubulin and a hallmark of stable microtubules, has gained recent attention given its association with tumor progression, invasiveness, and chemoresistance. Also, epithelial-to-mesenchymal transition (EMT) promotes tubulin detyrosination through tubulin tyrosine ligase (TTL) suppression. Given the induction of EMT associated with inflammation and cancer progression, Whipple et al. (2013) tested anti-inflammatory nuclear factor-kappaB (NF-κB) inhibitors on a panel of human breast carcinoma cells to examine their effects on detyrosinated tubulin to identify more specific tubulin-directed anti-cancer treatments.

Breast Cancer

Sesquiterpene lactones, parthenolide and costunolide, selectively decrease detyrosinated tubulin independent of their inhibition of NF-κB. Live-cell scoring of suspended cells treated with parthenolide and costunolide show reduction in the frequency of microtentacles and inhibition of reattachment. Selective targeting of detyrosinated tubulin with parthenolide and costunolide can reduce McTN frequency and inhibit tumor cell reattachment. These actions are independent of their effects on NF-κB inhibition, presenting a novel anti-cancer property and therapeutic opportunity to selectively target a stable subset of microtubules in circulating tumor cells to reduce metastatic potential with less toxicity in breast cancer patients (Whipple et al., 2013).

References

Bork PM, Schmitz ML, Kuhnt M, Escher C, Heinrich M. (1997). Sesquiterpene lactone containing Mexican Indian medicinal plants and pure sesquiterpene lactones as potent inhibitors of transcription factor NF-kappaB. FEBS Lett, 402:85-90.


Gunn EJ, Williams JT, Huynh DT, et al. (2011). The natural products parthenolide and andrographolide exhibit anti-cancer stem cell activity in multiple myeloma. Leuk Lymphoma, 52(6):1085-97.


Guzman ML, Rossi RM, Karnischky L, et al. (2005). The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cell. Blood, 105(11): 4163–4169. doi: 10.1182/blood-2004-10-4135.


Guzman ML, Swiderski CF, Howard DS, et al. (2002). Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci U S A, 99:16220-16225.


Koprowska K, Czyz M. (2010). Molecular mechanisms of parthenolide's action: Old drug with a new face. Postepy Hig Med Dosw, 64:100-14.


Kwak SW, Park ES, Lee CS. (2013). Parthenolide induces apoptosis by activating the mitochondrial and death receptor pathways and inhibits FAK-mediated cell invasion. Mol Cell Biochem.


Romano MF, Lamberti A, Bisogni R, et al. (2000). Enhancement of cytosine arabinoside-induced apoptosis in human myeloblastic leukemia cells by NF-kappa B/Rel-specific decoy oligodeoxynucleotides. Gene Ther, 7:1234-1237.


Whipple RA, Vitolo MI, Boggs AE, et al. (2013). Parthenolide and costunolide reduce microtentacles and tumor cell attachment by selectively targeting detyrosinated tubulin independent from NF- κ B inhibition. Breast Cancer Res,15(5):R83.


Woynarowski JM, Konopa J. (1981). Inhibition of DNA biosynthesis in HeLa cells by cytotoxic and anti-tumor sesquiterpene lactones. Mol Pharmacol,19:97-102.

Oridonin

Cancer: Prostate, acute promyelocytic leukemia, breast, non-small-cell lung (NSCL), Ehrlich ascites, P388 lymphocytic leukemia, colorectal., ovarian, esphageal

Action: Induces apoptosis

Oridonin is a tetracycline diterpenoid isolated from the plant Rabdosia rubescens (RR) [(Hemsl.). Hara (Lamiaceae)] (dong ling cao) is a Chinese medicinal herb used widely in provinces including Henan. The aerial parts of RR and other species of the same genus has been reported to have the functions of clearing “heat” and “toxicity”, nourishing “yin”, removing “blood stasis”, and relieving swelling. RR has been used to treat stomach-ache, sore throat and cough.

Gastric Cancer, Esophageal Cancer, Liver Cancer, Prostate Cancer

RR and its extracts have been shown to be able to suppress disease progress, reduce tumor burden, alleviate syndrome and prolong survival in patients with gastric carcinoma, esophageal., liver and prostate cancers (Tang & Eisenbrand, 1992). Interestingly, other Isodon plants including Isodon japonicus Hara (IJ) and I. trichocarpus (IT) are also applied as home remedies for similar disorders in Japan and Korea.

Induces Apoptosis

These reports suggest that Isodon plants should have at least one essential anti-tumor component. In the 1970s, a bitter tetracycline diterpenoid compound, oridonin, was isolated from RR, IJ, and IT separately, and was shown to be a potent apoptosis inducer in a variety of cancer cells (Fujita et al., 1970; Fujita et al., 1976; Henan Medical Institute, 1978; Fujita et al., 1988).

Anti-cancer

There is currently research being undertaken regarding the relationship between the chemical structure/modifications and the molecular mechanisms underlying its anti-cancer activity, such as suppression of tumor proliferation and induction of tumor cell death, and the cell signal transduction in anti-cancer activity of oridonin (Zhang et al., 2010).

Prostate Cancer, Breast Cancer, NSCLC, Leukemia, Glioblastoma

Oridonin has been found to effectively inhibit the proliferation of a wide variety of cancer cells including those from prostate (LNCaP, DU145, PC3), breast (MCF-7, MDA-MB231), non-small-cell lung (NSCL) (NCI-H520, NCI-H460, NCI-H1299) cancers, acute promyelocytic leukemia (NB4), and glioblastoma multiforme (U118, U138).

Oridonin induced apoptosis and G0/G1 cell-cycle arrest in LNCaP prostate cancer cells. In addition, expression of p21waf1 was induced in a p53-dependent manner. Taken together, oridonin inhibited the proliferation of cancer cells via apoptosis and cell-cycle arrest with p53 playing a central role in several cancer types which express the wild-type p53 gene. Oridonin may be a novel, adjunctive therapy for a large variety of malignancies (Ikezoe et al., 2003).

Breast Cancer; Anti-metastatic

According to the flow cytometric analysis, oridonin suppressed MCF-7 cell growth by cell-cycle arrest at the G2/M phase and caused accumulation of MDA-MB-231 cells in the Sub-G1 phase. The induced apoptotic effect of oridonin was further confirmed by a morphologic characteristics assay and TUNEL assay. Meanwhile, oridonin significantly suppressed MDA-MB-231 cell migration and invasion, decreased MMP-2/MMP-9 activation and inhibited the expression of Integrin β1 and FAK. In conclusion, oridonin inhibited growth and induced apoptosis in breast cancer cells, which might be related to DNA damage and activation of intrinsic or extrinsic apoptotic pathways. Moreover, oridonin also inhibited tumor invasion and metastasis in vitro possibly via decreasing the expression of MMPs and regulating the Integrin β1/FAK pathway in MDA-MB-231 cells (Wang et al., 2013).

Gastric Cancer

The inhibitory effect of oridonin on gastric cancer HGC-27 cells was detected using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. After treated with oridonin (0, 1.25, 2.5, 5 and 10 µg/mL), HGC-27 cells were collected for anexin V-phycoerythrin and 7-amino-actinomycin D double staining and tested by flow cytometric analysis, and oridonin- induced apoptosis in HGC-27 cells was detected.

Oridonin significantly inhibited the proliferation of HGC-27 cells in a dose- and time-dependent manner. The inhibition rates of HGC-27 treated with four different concentrations of oridonin for 24 h (1.25, 2.5, 5 and 10 µg/mL) were 1.78% ± 0.36%, 4.96% ± 1.59%, 10.35% ± 2.76% and 41.6% ± 4.29%, respectively, which showed a significant difference (P < 0.05. Cells treated with oridonin showed typical apoptotic features with acridine orange/ethidium bromide staining. After treatment with oridonin, the cells became round, shrank, and developed small buds around the nuclear membrane while forming apoptotic bodies. However, the change in the release of LDH caused by necrosis was insignificant, suggesting that the major cause of oridonin-induced HGC-27 cell death was apoptosis. Flow cytometric analysis also revealed that oridonin induced significant apoptosis compared with the controls (P < 0.05).

Apoptosis of HGC-27 induced by oridonin may be associated with differential expression of Apaf-1, caspase-3 and cytochrome c, which are highly dependent upon the mitochondrial pathway (Sun et al., 2012).

Ehrlich Ascites, Leukemia

Oridonin has been found to also increase lifespan of mice bearing Ehrlich ascites or P388 lymphocytic leukemia. Oridonin triggered apoptosis in more than 50% of t(8;21) leukemic cells in vitro at concentration of 2 M or higher accompanied by degradation of AE oncoprotein, and showed significant anti-leukemia efficacies with low adverse effects in vivo. These data suggest possible beneficial effects for patients with t(8;21) acute myeloid leukemia (AML) (Zhou et al., 2007).

Prostate Cancer, Breast Cancer, Ovarian Cancer

Oridonin exhibited anti-proliferative activity toward all cancer cell lines tested, with an IC50 estimated by the MTT cell viability assay ranging from 5.8+/-2.3 to 11.72+/-4.8 microM. The increased incidence of apoptosis, identified by characteristic changes in cell morphology, was seen in tumor lines treated with oridonin. Notably, at concentrations that induced apoptosis among tumor cells, oridonin failed to induce apoptosis in cultures of normal human fibroblasts. Oridonin up-regulated p53 and Bax and down-regulated Bcl-2 expression in a dose-dependent manner and its absorption spectrum was measured in the presence and absence of double stranded (ds) DNA. Oridonin inhibits cancer cell growth in a cell-cycle specific manner and shifts the balance between pro- and anti-apoptotic proteins in favor of apoptosis. The present data suggest that further studies are warranted to assess the potential of oridonin in cancer prevention and/or treatment (Chen et al., 2005).

Ovarian Cancer Stem Cells; Chemotherapy Resistance

Oridonin was suggested to suppress ovarian CSCs as is reflected by down-regulation of the surface marker EpCAM. Unlike NSAIDS (non-steroid anti-inflammatory drugs), well documented clinical data for phyto-active compounds are lacking. In order to evaluate objectively the potential benefit of these types of compounds in the treatment of ovarian cancer, strategically designed, large scale studies are warranted (Chen et al., 2012).

Colorectal Cancer

Oridonin induced potent growth inhibition, cell-cycle arrest, apoptosis, senescence and colony-forming inhibition in three colorectal cancer cell lines in a dose-dependent manner in vitro. Daily i.p. injection of oridonin (6.25, 12.5 or 25 mg/kg) for 28 days significantly inhibited the growth of SW1116 s.c. xenografts in BABL/C nude mice.

Oridonin possesses potent in vitro and in vivo anti-colorectal cancer activities that correlated with induction of histone hyperacetylation and regulation of pathways critical for maintaining growth inhibition and cell-cycle arrest. Therefore, oridonin may represent a novel therapeutic option in colorectal cancer treatment as it has been shown to induce apoptosis and senescence of colon cancer cells in vitro and in vivo (Gao et al., 2010).

Colon Cancer; Apoptosis

Oridonin increased intracellular hydrogen peroxide levels and reduced the glutathione content in a dose-dependent manner. N-acetylcysteine, a reactive oxygen species scavenger, not only blocked the oridonin-induced increase in hydrogen peroxide and glutathione depletion, but also blocked apoptosis and senescence induced by oridonin.

Moreover, exogenous catalase could inhibit the increase in hydrogen peroxide and apoptosis induced by oridonin, but not the glutathione depletion and senescence. Furthermore, thioredoxin reductase (TrxR) activity was reduced by oridonin in vitro and in cells, which may cause the increase in hydrogen peroxide. In conclusion, the increase in hydrogen peroxide and glutathione depletion account for oridonin-induced apoptosis and senescence in colorectal cancer cells, and TrxR inhibition is involved in this process.

Given the importance of TrxR as a novel cancer target in colon cancer, oridonin would be a promising clinical candidate (Gao et al., 2012).

Prostate Cancer; Apoptosis

Oridonin (ORI) could inhibit the proliferation and induce apoptosis in various cancer cell lines. After ORI treatment, the proliferations of human prostate cancer (HPC) cell lines PC-3 and LNCaP were inhibited in a concentration and time-dependent manner. ORI induced cell-cycle arrest at the G2/M phase. Autophagy occurred before the onset of apoptosis and protected cancer cells in ORI-treated HPC cells. P21 was involved in ORI-induced autophagy and apoptosis (Li et al., 2012).

References

Chen S, Gao J, Halicka HD, et al. (2005). The cytostatic and cytotoxic effects of oridonin (Rubescenin), a diterpenoid from Rabdosia rubescens, on tumor cells of different lineage. Int J Oncol, 26(3):579-88.


Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.


Fujita E, Fujita T, Katayama H, Shibuya M. (1970). Terpenoids. Part XV. Structure and absolute configuration of oridonin isolated from Isodon japonicus trichocarpus. J Chem Soc (Chem Comm), 21:1674–1681


Fujita E, Nagao Y, Node M, et al. (1976). Anti-tumor activity of the Isodon diterpenoids: structural requirements for the activity. Experientia, 32:203–206.


Fujita T, Takeda Y, Sun HD, et al. (1988). Cytotoxic and anti-tumor activities of Rabdosia diterpenoids. Planta Med, 54:414–417.


Henan Medical Institute, Henan Medical College, Yunnan Institute of Botany. (1978). Oridonin–a new anti-tumor subject. Chin Science Bull, 23:53–56.


Ikezoe T, Chen SS, Tong XJ, et al. (2003). Oridonin induces growth inhibition and apoptosis of a variety of human cancer cells. Int J Oncol, 23(4):1187-93.


Gao FH, Hu XH, Li W, Liu H, et al. (2010). Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc. BMC Cancer, 10:610. doi: 10.1186/1471-2407-10-610.


Gao FH, Liu F, Wei W, et al. (2012). Oridonin induces apoptosis and senescence by increasing hydrogen peroxide and glutathione depletion in colorectal cancer cells. Int J Mol Med, 29(4):649-55. doi: 10.3892/ijmm.2012.895.


Li X, Li X, Wang J, Ye Z, Li JC. (2012) Oridonin up-regulates expression of P21 and induces autophagy and apoptosis in human prostate cancer cells. Int J Biol Sci. 2012;8(6):901-12. doi: 10.7150/ijbs.4554.


Sun KW, Ma YY, Guan TP, et al. (2012). Oridonin induces apoptosis in gastric cancer through Apaf-1, cytochrome c and caspase-3 signaling pathway. World J Gastroenterol, 18(48):7166-74. doi: 10.3748/wjg.v18.i48.7166.


Tang W, Eisenbrand G. (1992). Chinese drugs of plant origin: chemistry, pharmacology, and use in traditional and modern medicine. Berlin: Springer-Verlag, 817–847.


Wang S, Zhong Z, Wan J, et al. (2013). Oridonin induces apoptosis, inhibits migration and invasion on highly-metastatic human breast cancer cells. Am J Chin Med, 41(1):177-96. doi: 10.1142/S0192415X13500134.


Zhang Wj, Huang Ql, Hua Z-C. (2010). Oridonin: A promising anti-cancer drug from China. Frontiers in Biology, 5(6):540-545.


Zhou G-B, Kang H, Wang L, et al. (2007). Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent anti-tumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood, 109(8):3441-3450.