Category Archives: Ovarian cancer

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.

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.

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.

Honokiol (See also Injectables)

Cancer:
Lung, breast, prostate, leukemia, colorectal., esophageal., ovarian, myeloma, pancreatic, stomach, uterine

Action: Anti-angiogenic, chemo-sensitizer, multi-drug resistance reversal., anti-inflammatory, anxiolytic, anti-depressant, inhibits VEGF, anti-metastatic, synergistic effects with other cancer treatments

Honokiol is a phenolic compound purified from plants of the Magnolia genus, including Magnolia officinalis (Rehder & Wilson) and Magnolia grandiflora (L.), that exhibits anti-cancer effects in experimental models with various types of cancer cells, including esophageal., ovarian, breast, and lung cancer, as well as myeloma and leukemia. It is speculated that this compound causes cancer cell death in part through targeting mitochondria (Munroe et al., 2007; Chen et al., 2009; Fried & Arbiser, 2009).

Inhibits Angiogenesis, MDR, Anti-inflammatory, Inhibits VEGF

Honokiol is one of two dominant biphenolic compounds isolated from Magnolia spp. bark, and is the most widely researched active constituent of the bark. In vivo studies suggest that honokiol's greatest value is in its multiple anti-cancer actions. In vitro research suggests honokiol has potential to enhance current anti-cancer regimens by inhibiting angiogenesis, promoting apoptosis, providing direct cytotoxic activity, down-regulating cancer cell signaling pathways, regulating genetic expression, enhancing the effects of specific chemotherapeutic agents, radio-sensitizing cancer cells to radiation therapy, and inhibiting multi-drug resistance.

Honokiol also shows potential in preventive health by reducing inflammation and oxidative stress, providing neurological protection, and regulating glucose; in mental illness by its effects against anxiety and depression; and in helping regulate stress response signaling. Its anti-microbial effects demonstrate potential for partnering with anti-viral/antibiotic therapy, and treating secondary infections.

Honokiol may occupy a distinct therapeutic niche because of its unique characteristics: the ability to cross the blood brain barrier (BBB) and blood cerebrospinal fluid barrier (BCSFB), high systemic bioavailability, and its actions on a multiplicity of signaling pathways and genomic activity. There is a need for research on honokiol to progress to human studies and on into clinical use.

The preclinical research on honokiol's broad-ranging capabilities shows its potential as a therapeutic compound for numerous solid and hematological cancers, including its effectiveness in combating multi-drug resistance (MDR) and its synergy with other anti-cancer therapies. Research thus far shows no toxicity or serious adverse effects in animal models.

Honokiol has also been shown to inhibit spread of cancer cells through the lymph system by inhibiting one of the primary pathways involved in growth stimulation related to vascular endothelial growth factor (VEGF) (Wen et al., 2009).

Inhibits Angiogenesis, Gastric Cancer

A 2012 in vivo study in PLoS One showed that honokiol, by inhibiting angiogenic pathways such as STAT-3, dampened peritoneal dissemination of gastric cancer in mice (5 mg/kg delivered intraperitoneally) (Liu et al., 2012).    

Induces Apoptosis; Leukemia

Honokiol induces cell apoptosis in several cell lines, such as leukemia cell lines HL-60, colon cancer cell lines RKO, lung cancer cell lines A549 and CH27 (Hirano et al., 1994; Wang et al., 2004; Hibasami et al., 1998; Konoshima et al., 1991;Yang et al., 2002; Kong et al., 2005). It also has remarkable in vivo anti-tumor activities in tumor mouse models (Bai et al., 2003). Honokiol has demonstrated potent anti-angiogenic and anti-tumor properties against aggressive angiosarcoma by blocking of VEGF-induced VEGF receptor 2 autophosphorylation (Konoshima et al., 1991; Yang et al., 2002).

MDR

Honokiol has also been found to down-regulate the expression of P-glycoprotein at mRNA and protein levels in MCF-7/ADR, a human breast MDR cancer cell line. The down-regulation of P-glycoprotein is accompanied with a partial recovery of the intracellular drug accumulation (Xu et al., 2006).

Prostate Cancer

In addition, it has been shown that prostate cancer cells that failed to respond to hormone withdrawal responded to honokiol-induced apoptosis. It was found to significantly induce death in cells surrounding primary and metastatic prostate cancers, the prostate stromal fibroblasts, marrow stromal cells, and bone marrow-associated endothelial cells. Honokiol is hence a promising nontoxic agent that could be used as an adjuvant with low-dose docetaxel for the treatment of hormone-refractory prostate cancer and its distant bone metastases (Shigemura et al., 2007).

Anti-metastatic

Honokiol inhibited the activity of MMP-9, which may be responsible, in part, for the inhibition of tumor cell invasiveness (Nagase et al., 2001).

Breast Cancer

The development of more targeted and low toxic drugs from traditional Chinese medicines for breast cancer are needed due to most of the anti-breast cancer drugs often being limited because of drug resistance and serious adverse reactions. Results have shown that honokiol inhibited the rate of breast cancer MDA-MB-231 cell growth (Nagalingam et al., 2012).

Synergistic Effects with Other Cancer Treatments

One of the most promising benefits of honokiol is its ability to synergize with other cancer treatments. Clinical trials are desperately needed to validate the potential synergy that has been demonstrated in vitro and in vivo.

Chemotherapy

• A 2013 in vitro study published in the International Journal of Oncology showed that honokiol synergized chemotherapy drugs in Multi-drug-resistant breast cancer (Tian et al., 2013). A 2011 in vitro study published in PLoS One found that honokiol enhanced the apoptotic effects of the anti-cancer drug gemcitabine against pancreatic cancer (Arora et al., 2011).

• In vivo research published in Oncology Letters in 2011 found honokiol enhanced the action of cisplatin against colon cancer (Cheng et al., 2011).

• A 2010 in vitro study from the Journal of Biological Regulators and Homeostatic Agents showed that honokiol resensitized cancer cells to doxorubicin in Multi-drug-resistant uterine cancer (Angelini et al., 2010).

• A 2010 in vitro study published in Toxicology Mechanisms and Methods showed honokiol performed synergistically with the drug imatinib against human leukemia cells (Wang et al., 2010).

• 2008 in vivo research published in the International Journal of Gynecological Cancer showed honokiol to potentiate the activity of cisplatin in murine models of ovarian cancer (Liu et al., 2008).

• 2005 in vitro research published in Blood showed honokiol enhanced the cytotoxicity induced by fludarabine, cladribine, and chlorambucil, indicating it is a potent inducer of apoptosis in B-CLL cells (Battle et al., 2005).

Radiation treatment

• 2012 in vitro research published in Molecular Cancer Therapeutics showed that honokiol was able to sensitize cancer cells to radiation treatments (Ponnurangam et al., 2012).

• A 2011 in vitro study published in American Journal of Physiology Gastrointestinal and Liver Physiology showed honokiol sensitized treatment-resistant colon cancer cells to radiation therapy (He et al., 2011).

Inhibition of multi-drug resistance

Honokiol has been shown to interact with genes that are involved with mechanisms of drug efflux, thus reversing MDR in experimental models. The exact mechanisms of action in this regard are thought to be related to effects of blocking of NF-kB activity, but other mechanisms may also be involved (Xu et al., 2006).

References

Angelini A, Di Ilio C, Castellani ML, Conti P, Cuccurullo F. (2010). Modulation of Multi-drug resistance p-glycoprotein activity by flavonoids and honokiol in human doxorubicin-resistant sarcoma cells (MES-SA/DX-5): Implications for natural sedatives as chemosensitizing agents in cancer therapy. Journal of Biological Regulators & Homeostatic Agents, 24(2). 197-205.


Arora S, Bhardwaj A, Srivastava SK, et al. (2011). Honokiol arrests Cell-cycle, induces apoptosis, and potentiates the cytotoxic effect of gemcitabine in human pancreatic cancer cells. PLoS One, 6(6), e21573. doi: 10.1371/journal.pone.0021573.


Bai X, Cerimele F, Ushio-Fukai M, et al. (2003). Honokiol, a small molecular weight natural product, inhibits angiogenesis in vitro and tumor growth in vivo. J Biol Chem, 278: 35501–7.


Battle TE, Arbiser J, Frank DA. (2005). The natural product honokiol induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia (B-CLL) cells. Blood, 106(2), 690-697.


Chen G, Izzo J, Demizu Y, et al. (2009). Different redox states in malignant and nonmalignant esophageal epithelial cells and differential cytotoxic responses to bile acid and honokiol. Antioxid. Redox Signal., 11(5):1083–1095


Cheng N, Xia T, Han Y, et al. (2001). Synergistic anti-tumor effects of liposomal honokiol combined with cisplatin in colon cancer models. Oncology Letters, 2(5), 957-962.


Eliaz I. (2013). Honokiol research review: A promising extract with multiple applications. Natural Medicine Journal., 5(7).


Fried LE, Arbiser JL. (2009). Honokiol, a multifunctional anti-angiogenic and anti-tumor agent. Antioxid. Redox Signal., 1(5):1139–1148. doi: 10.1089/ARS.2009.2440.


He Z, Subramaniam D, Ramalingam S, et al. (2011). Honokiol radiosensitizes colorectal cancer cells: enhanced activity in cells with mismatch repair defects. American Journal of Physiology: Gastrointest and Liver Physiology, 301(5):G929-937.


Hibasami H, Achiwa Y, Katsuzaki H, et al. (1998). Honokiol induces apoptosis in human lymphoid leukemia Molt 4B cells. Int J Mol Med, 2:671–3.


Hirano T, Gotoh M, Oka K. (1994). Natural flavonoids and lignans are potent cytostatic agents against human leukemic HL-60 cells. Life Sci, 55:1061–9.


Hou X, Yuan X, Zhang B, Wang S, Chen Q. (2013). Screening active anti-breast cancer compounds from Cortex Magnolia officinalis by 2D LC-MS. J Sep Sci, 36(4):706-12. doi: 10.1002/jssc.201200896.


Kong ZL, Tzeng SC, Liu YC. (2005). Cytotoxic neolignans: an SAR study. Bioorg Med Chem Lett, 15: 163–6.


Konoshima T, Kozuka M, Tokuda H, et al. (1991). Studies on inhibitors of skin tumor promotion. IX. Neolignans from Magnolia officinalis. J Nat Prod, 54: 816–22.


Liu Y, Chen L, He X, et al. (2010). Enhancement of therapeutic effectiveness by combining liposomal honokiol with cisplatin in ovarian carcinoma. International Journal of Gynecological Cancer, 18(4), 652-659.


Liu SH, Wang KB, Lan KH, et al. (2012). Calpain/SHP-1 interaction by honokiol dampening peritoneal dissemination of gastric cancer in nu/nu mice. PLoS One, 7(8):e43711.


Munroe ME, Arbiser JL, Bishop GA. (2007). Honokiol, a natural plant product, inhibits inflammatory signals and alleviates inflammatory arthritis. J. Immunol., 179(2):753–763


Nagalingam A, Arbiser JL, Bonner MY, Saxena NK, Sharma D. (2012). Honokiol activates AMP-activated protein kinase in breast cancer cells via an LKB1-dependent pathway and inhibits breast carcinogenesis. Breast Cancer Research, 14:R35 doi:10.1186/bcr3128


Nagase H, Ikeda K, Sakai Y. (2001). Inhibitory Effect of Magnolol and Honokiol from Magnolia obovata on Human Fibrosarcoma HT-1080 Invasiveness in vitro. Planta Med, 67(8): 705-708. DOI: 10.1055/s-2001-18345


Ponnurangam S, Mammen JM, Ramalingam S, et al. (2012). Honokiol in combination with radiation targets notch signaling to inhibit colon cancer stem cells. Molecular Cancer Therapeutics, 11(4), 963-972. doi: 10.1371/journal.pone.0043711.


Shigemura K, Arbiser JL, Sun SY, et al. (2007). Honokiol, a natural plant product, inhibits the bone metastatic growth of human prostate cancer cells. Cancer, 109(7), 1279-1289.


Tian W, Deng Y, Li L, et al. (2013). Honokiol synergizes chemotherapy drugs in Multi-drug-resistant breast cancer cells via enhanced apoptosis and additional programmed necrotic death. International Journal of Oncology, 42(2), 721-732. doi: 10.3892/ijo.2012.1739.


Wang Y, Yang Z, Zhao X. (2010). Honokiol induces parapoptosis and apoptosis and exhibits schedule-dependent synergy in combination with imatinib in human leukemia cells. Toxicology Mechanisms and Methods, 20(5), 234-241. doi: 10.3109/15376511003758831.


Wang T, Chen F, Chen Z, et al. (2004). Honokiol induces apoptosis through p53-independent pathway in human colorectal cell line RKO. World J Gastroenterol, 10: 2205–8.


Wen J, Fu AF, Chen LJ, et al. (2009). Liposomal honokiol inhibits VEGF-D-induced lymphangiogenesis and metastasis in xenograft tumor model. International Journal of Cancer, 124(11), 2709-2718. doi: 10.1002/ijc.24244.


Xu D, Lu Q, Hu X. (2006). Down-regulation of P-glycoprotein expression in MDR breast cancer cell MCF-7/ADR by honokiol. Cancer Letters, 243(2), 274-280.


Yang SE, Hsieh MT, Tsai TH, Hsu SL. (2002). Down-modulation of Bcl-XL, release of cytochrome c and sequential activation of caspases during honokiol-induced apoptosis in human squamous lung cancer CH27 cells. Biochemical Pharmacology, 63(9), 1641-1651.

Source

Eliaz I. (2013). Honokiol research review: A promising extract with multiple applications. Natural Medicine Journal., 5(7). Retrieved from http://www.naturalmedicinejournal.com/article_content.asp?edition=1.

Curcumin

Cancer: Colorectal., prostate, pancreatic

Action: MDR, chemo-preventive activity, anti-inflammatory, attenuation of immune suppression

Chemo-preventive Activity

Curcumin is a naturally occurring, dietary polyphenolic phytochemical that is under preclinical trial evaluation for cancer-preventive drug development. It is derived from the rhizome of Curcuma longa L. and has both anti-oxidant and anti-inflammatory properties; it inhibits chemically-induced carcinogenesis in the skin, forestomach, and colon when it is administered during initiation and/or postinitiation stages. Chemo-preventive activity of curcumin is observed when it is administered prior to, during, and after carcinogen treatment as well as when it is given only during the promotion/progression phase (starting late in premalignant stage) of colon carcinogenesis (Kawamori et al., 1999)

Anti-inflammatory

With respect to inflammation, in vitro, it inhibits the activation of free radical-activated transcription factors, such as nuclear factor κB (NFκB) and AP-1, and reduces the production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNFα), interleukin-1β (IL-1β), and interleukin-8 (Chan et al., 1998)

Prostate Cancer

In addition, NF-kappaB and AP-1 may play a role in the survival of prostate cancer cells, and curcumin may abrogate their survival mechanisms (Mukhopadhyay et al., 2001).

Pancreatic Cancer

In patients suffering from pancreatic cancer, orally-administered curcumin was found to be well-tolerated and despite limited absorption, had a reasonable impact on biological activity in some patients. This was attributed to its potent nuclear factor-kappaB (NF-kappaB) and tumor-inhibitory properties, against advanced pancreatic cancer (Dhillon et al., 2008)

MDR

Curcumin, the major component in Curcuma longa (Jianghuang), inhibited the transport activity of all three major ABC transporters, i.e. Pgp, MRP1 and ABCG2 (Ganta et al., 2009).

Curcumin reversed MDR of doxorubicin or daunorubicin in K562/DOX cell line and decreased Pgp expression in a time-dependent manner (Chang et al., 2006). Curcumin enhanced the sensitivity to vincristine by the inhibition of Pgp in SGC7901/VCR cell line (Tang et al., 2005). Moreover, curcumin was useful in reversing MDR associated with a decrease in bcl-2 and survivin expression but an increase in caspase-3 expression in COC1/DDP cell line (Ying et al., 2007).

The cytotoxicity of vincristine and paclitaxel were also partially restored by curcumin in resistant KBV20C cell line. Curcumin derivatives reversed MDR by inhibiting Pgp efflux (Um et al., 2008). A chlorine substituent at the meta-or para-position on benzamide improved MDR reversal [72]. Bisdemethoxycurcumin modified from curcumin resulted in greater inhibition of Pgp expression (Limtrakul et al., 2004).

Attenuation of Immune Suppression

Curcumin (a chalcone) exhibited toxicity to human neural stem cells (hNSCs). Although oridonin (a diterpene) showed a null toxicity toward hNSCs, it repressed the enzymatic function only marginally in contrast to its potent cytotoxicity in various cancer cell lines. While the mode of action of the enzyme-polyphenol complex awaits to be investigated, the sensitivity of enzyme inhibition was compared to the anti-proliferative activities toward three cancer cell lines.

The IC50s obtained from both sets of the experiments indicate that they are in the vicinity of micromolar concentration with the enzyme inhibition slightly more active.

These results suggest that attenuation of immune suppression via inhibition of IDO-1 enzyme activity may be one of the important mechanisms of polyphenols in chemoprevention or combinatorial cancer therapy (Chen et al., 2012).

Cancer Stem Cells

In cancers that appear to follow the stem cell model, pathways such as Wnt, Notch and Hedgehog may be targeted with natural compounds such as curcumin or drugs to reduce the risk of initiation of new tumors. Disease progression of established tumors could also potentially be inhibited by targeting the tumorigenic stem cells alone, rather than aiming to reduce overall tumor size.

Cancer treatments could be evaluated by assessing stem cell markers before and after treatment. Targeted stem cell specific treatment of cancers may not result in 'complete' or 'partial' responses radiologically, as stem cell targeting may not reduce the tumor bulk, but eliminate further tumorigenic potential. These changes are discussed using breast, pancreatic, and lung cancer as examples (Reddy et al., 2011).

Multiple Cancer Effects; Cell-signaling

Curcumin has been shown to interfere with multiple cell signaling pathways, including cell-cycle (cyclin D1 and cyclin E), apoptosis (activation of caspases and down-regulation of anti-apoptotic gene products), proliferation (HER-2, EGFR, and AP-1), survival (PI3K/AKT pathway), invasion (MMP-9 and adhesion molecules), angiogenesis (VEGF), metastasis (CXCR-4) and inflammation (NF- κB, TNF, IL-6, IL-1, COX-2, and 5-LOX).

The activity of curcumin reported against leukemia and lymphoma, gastrointestinal cancers, genitourinary cancers, breast cancer, ovarian cancer, head and neck squamous cell carcinoma, lung cancer, melanoma, neurological cancers, and sarcoma reflects its ability to affect multiple targets (Anand et al., 2008).

Anti-inflammatory; Cell-signaling

Curcumin, a liposoluble polyphenolic pigment isolated from the rhizomes of Curcuma longa L. (Zingiberaceae), is another potential candidate for new anti-cancer drug development. Curcumin has been reported to influence many cell-signaling pathways involved in tumor initiation and proliferation. Curcumin inhibits COX-2 activity, cyclin D1 and MMPs overexpresion, NF-kB, STAT and TNF-alpha signaling pathways and regulates the expression of p53 tumor suppressing gene.

Curcumin is well-tolerated but has a reduced systemic bioavailability. Polycurcumins (PCurc 8) and curcumin encapsulated in biodegradable polymeric nanoparticles showed higher bioavailability than curcumin together with a significant tumor growth inhibition in both in vitro and in vivo studies (Cretu et al., 2012). Curcumin also sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 (DR5) (Jung et al., 2005).

Curcumin and bioavailability

Curcumin, a major constituent of the spice turmeric, suppresses expression of the enzyme cyclooxygenase 2 (Cox-2) and has cancer chemo-preventive properties in rodents. It possesses poor systemic availability. Marczylo et al. (2007) explored whether formulation with phosphatidylcholine increases the oral bioavailability or affects the metabolite profile of curcumin. Their results suggest that curcumin formulated with phosphatidylcholine furnishes higher systemic levels of parent agent than unformulated curcumin.

Curcuminoids are poorly water-soluble compounds and to overcome some of the drawbacks of curcuminoids, Aditya et al. (2012) explored the potential of liposomes for the intravenous delivery of curcuminoids. The curcuminoids-loaded liposomes were formulated from phosphatidylcholine (soy PC). Curcumin/curcuminoids were encapsulated in phosphatidylcholine vesicles with high yields. Vesicles in the size range around 200 nm were selected for stability and cell experiments. Liposomal curcumin were found to be twofold to sixfold more potent than corresponding curcuminoids. Moreover, the mixture of curcuminoids was found to be more potent than pure curcumin in regard to the anti-oxidant and anti-inflammatory activities (Basnet et al., 2012). Results suggest that the curcumin-phosphatidylcholine complex improves the survival rate by increasing the anti-oxidant activity (Inokuma et al., 2012). Recent clinical trials on the effectiveness of phosphatidylcholine formulated curcumin in treating eye diseases have also shown promising results, making curcumin a potent therapeutic drug candidate for inflammatory and degenerative retinal and eye diseases (Wang et al., 2012). Data demonstrate that treatment with curcumin dissolved in sesame oil or phosphatidylcholine curcumin improves the peripheral neuropathy of R98C mice by alleviating endoplasmic reticulum stress, by reducing the activation of unfolded protein response (Patzk- et al., 2012).

References

Aditya NP, Chimote G, Gunalan K, et al. (2012). Curcuminoids-loaded liposomes in combination with arteether protects against Plasmodium berghei infection in mice. Exp Parasitol, 131(3):292-9. doi: 10.1016/j.exppara.2012.04.010.


Anand P, Sundaram C, Jhurani S, Kunnumakkara AB, Aggarwal BB. (2008). Curcumin and cancer: An 'old-age' disease with an 'age-old' solution. Cancer Letters, 267(1):133–164. doi: 10.1016/j.canlet.2008.03.025.


Basnet P, Hussain H, Tho I, Skalko-Basnet N. (2012). Liposomal delivery system enhances anti-inflammatory properties of curcumin. J Pharm Sci, 101(2):598-609. doi: 10.1002/jps.22785.


Chan MY, Huang HI, Fenton MR, Fong D. (1998). In Vivo Inhibition of Nitric Oxide Synthase Gene Expression by Curcumin, a Cancer-preventive Natural Product with Anti-Inflammatory Properties. Biochemical Pharmacology, 55(12), 1955-1962.


Chang HY, Pan KL, Ma FC, et al. (2006). The study on reversing mechanism of Multi-drug resistance of K562/DOX cell line by curcumin and erythromycin. Chin J Hem, 27(4):254-258.


Chen SS, Corteling R, Stevanato L, Sinden J. (2012). Polyphenols Inhibit Indoleamine 3,5-Dioxygenase-1 Enzymatic Activity — A Role of Immunomodulation in Chemoprevention. Discovery Medicine.


Cre ţ u E, Trifan A, Vasincu A, Miron A. (2012). Plant-derived anti-cancer agents – curcumin in cancer prevention and treatment. Rev Med Chir Soc Med Nat Iasi, 116(4):1223-9.


Dhillon N, Aggarwal BB, Newman RA, et al. (2008). Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res,14(14):4491-9. doi: 10.1158/1078-0432.CCR-08-0024.


Ganta S, Amiji M. (2009). Coadministration of paclitaxel and curcumin in nanoemulsion formulations To overcome Multi-drug resistance in tumor cells. Mol Pharm, 6(3):928-939. doi: 10.1021/mp800240j.


Inokuma T, Yamanouchi K, Tomonaga T, et al. (2012). Curcumin improves the survival rate after a massive hepatectomy in rats. Hepatogastroenterology, 59(119):2243-7. doi: 10.5754/hge10650.


Jung EM, Lim JH, Lee TJ, et al. (2005). Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated up-regulation of death receptor 5 (DR5). Carcinogenesis, 26(11):1905-1913.


Kawamori T, Lubet R, Steele V E, et al. (1999). Chemo-preventive Effect of Curcumin, a Naturally Occurring Anti-Inflammatory Agent, during the Promotion/Progression Stages of Colon Cancer. Cancer Research, 59(3), 597-601.


Limtrakul P, Anuchapreeda S, Buddhasukh D. (2004). Modulation of human Multi-drug resistance MDR-1 gene by natural curcuminoids. BMC Cancer, 4:13.


Marczylo TH, Verschoyle RD, Cooke DN, et al. (2007). Comparison of systemic availability of curcumin with that of curcumin formulated with phosphatidylcholine. Cancer Chemother Pharmacol, 60(2):171-7.


Mukhopadhyay A, Bueso-Ramos C, Chatterjee D, Pantazis P, & Aggarwal., B. B. (2001). Curcumin downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene, 20(52), 7597-7609.


Patzk- A, Bai Y, Saporta MA, et al. (2012). Curcumin derivatives promote Schwann cell differentiation and improve neuropathy in R98C CMT1B mice. Brain, 135(Pt 12):3551-66. doi: 10.1093/brain/aws299.


Reddy RM, Kakarala M, Wicha MS. (2011). Clinical trial design for testing the stem cell model for the prevention and treatment of cancer. Cancers (Basel), 3(2):2696-708. doi: 10.3390/cancers3022696.


Tang XQ, Bi H, Feng JQ, Cao JG. (2005). Effect of curcumin on Multi-drug resistance in resistant human gastric carcinoma cell line SGC7901/VCR. Acta Pharmacol Sin, 26(8):1009-1016.


Um Y, Cho S, Woo HB, et al. (2008). Synthesis of curcumin mimics with Multi-drug resistance reversal activities. Bioorg Med Chem,16(7):3608-3615.


Wang LL, Sun Y, Huang K, Zheng L. (2012). Curcumin, a potential therapeutic candidate for retinal diseases. Mol Nutr Food Res, 57(9):1557-68. doi: 10.1002/mnfr.201200718.


Ying HC, Zhang SL, Lv J. (2007). Drug-resistant reversing effect of curcumin on COC1/DDP and its mechanism. J Mod Oncol, 15(5):604-607.

Oplopanax horridus

Cancer: Ovarian

Action: Chemotherapy sensitising, anti-proliferation, apoptosis inducing

To search for more effective treatment of ovarian cancer, Tai et al., (2010) investigated the in vitro anti-proliferation activities of Oplopanax horridus (Devil’s club/OH) root bark extracts, an important medicinal plant of North America, on cisplatin sensitive and resistant human ovarian cancer cell lines. Their data showed that water, 70% ethanol, 100% ethanol, and ethyl acetate extracts of OH inhibited the proliferation of human ovarian cancer cell lines A2780, A2780CP70, OVCAR3, and OVCAR10 in vitro. The respective 50% inhibition (IC(50)) was estimated at 1/256, 1/74, 1/69, 1/53; 1/4156, 1/1847, 1/1029, 1/4530; 1/25,753, 1/3310, 1/3462, 1/5049; and 1/29,916, 1/2912 1/3828, and 1/4232 dilutions. Some combinations of non-cytotoxic dilutions (<IC(50)) of 70% ethanol OH extract with cisplatin and paclitaxel enhanced its anti-proliferation IC(50) on A2780 and A2780CP70 cells. Cell cycle analysis demonstrated that the effect of OH extract on cell cycle was dependent on the concentration tested, blocking cells in the S and G2/M phases. At low concentrations it induced cell death by apoptosis, while at high concentrations, it kills cells by necrosis. Their data showed that OH extracts exhibited significant anti-proliferation effect against both cisplatin sensitive and resistant human ovarian cell lines. Further research might result in discovery of agent(s) that can potentially be useful as an adjunct therapy for ovarian cancer cells. It is one of the few North American medicinal herbs that have been tested for anti-ovarian cancer activities.

Reference

Tai J, Cheung S, Chan E, Hasman D. (2010) Inhibition of human ovarian cancer cell lines by devil’s club Oplopanax horridus. J Ethnopharmacol. 2010 Feb 3;127(2):478-85. doi: 10.1016/j.jep.2009.10.010.