Category Archives: SHH

Zerumbone

Cancer:
Colorectal, renal carcinoma, glioblastoma, ovarian and cervical

Action: CSCs, anti-inflammatory

Zerumbone is isolated from Zingiber zerumbet [(L.) Roscoe ex Sm.].

Colorectal Cancer

Numerous agents from 'mother nature' (also called nutraceuticals) that have potential to both prevent and treat CRC have been identified. The most significant discoveries relate to compounds such as cardamonin, celastrol, curcumin, deguelin, diosgenin, thymoquinone, tocotrienol, ursolic acid, and zerumbone. Unlike pharmaceutical drugs, these agents modulate multiple targets, including transcription factors, growth factors, tumor cell survival factors, inflammatory pathways, and invasion and angiogenesis linked closely to CRC. We describe the potential of these dietary agents to suppress the growth of human CRC cells in culture and to inhibit tumor growth in animal models (Aggarwal et al., 2013).

Cancer Stem Cells (CSCs)

Cancer stem cells (CSCs) are a major cause of cancer treatment failure, relapse, and drug resistance and are known to be responsible for cancer cell invasion and metastasis. The Sonic hedgehog (Shh) signaling pathway is crucial to embryonic development. Intriguingly, the aberrant activation of the Shh pathway plays a critical role in developing CSCs and leads to angiogenesis, migration, invasion, and metastasis. Natural compounds and chemical structure modified derivatives from complementary and alternative medicine have received increasing attention as cancer chemo-preventives, and their anti-tumor effects have been demonstrated both in vitro and in vivo.

Compounds cyclopamine, curcumin, epigallocatechin-3-gallate, genistein, resveratrol, zerumbone, norcantharidin, and arsenic trioxide, with a focus on Shh signaling blockade, were reviewed by Huang et al. (2013) and given that Shh signaling antagonism has been clinically proven as an effective strategy against CSCs, this review may be exploitable for the development of novel anti-cancer agents from complementary and alternative medicine.

Renal Carcinoma

Sun et al. (2013) reported that zerumbone, a monosesquiterpine, shows anti-cancer effects on human RCC cells via induction of apoptosis in vitro. Human renal clear cell carcinoma 786-0 and 769-P cell lines were used as the model system. Exposure of RCC cells to zerumbone resulted in cell viability inhibition, accompanied by DNA fragmentation and increased apoptotic index. Mechanically, treatment of RCC cells with zerumbone activated caspase-3 and caspase-9 finally led to cleavage of PARR.

Taken together, our studies provided the first evidence that zerumbone imparted strong inhibitory and apoptotic effects on human RCC cells. The zerumbone-induced apoptosis might be related to the activation of the caspase cascade and deregulation of the Gli-1/Bcl-2 pathway. Our results suggest that zerumbone merit further investigation as an apoptosis inducer as well as a novel RCC chemotherapeutic agent in the clinical setting.

Glioblastoma

Zerumbone (10~50 µM) induced death of human glioblastoma multiforme (GBM8401) cells in a dose-dependent manner. Flow cytometry studies showed that zerumbone increased the percentage of apoptotic GBM cells. Zerumbone also caused caspase-3 activation and poly (ADP-ribose) polymerase (PARP) production. N-benzyloxycarbonyl -Val-Ala-Asp- fluoromethylketone (zVAD-fmk), a broad-spectrum caspase inhibitor, hindered zerumbone-induced cell death. Moreover, transfection of GBM8401 cells with WT IKKα reduced the zerumbone-induced decrease in Akt and FOXO1 phosphorylation. However, transfection with WT Akt decreased FOXO1, but not IKKα, phosphorylation.

The results suggest that inactivation of IKKα, followed by Akt and FOXO1 phosphorylation and caspase-3 activation, contributes to zerumbone-induced GBM cell apoptosis (Weng et al., 2012).

Ovarian and Cervical Cancer

A study by Abdelwahab et al., (2012) was designed to investigate the role of IL-6 and IL6 receptors in the cytotoxic effects of zerumbone in ovarian and cervical cancer cell lines (Caov-3 and HeLa, respectively). Exposure of both cancer cells to zerumbone or cisplatin demonstrated growth inhibition in a dose-dependent manner as determined by the MTT reduction assay. The studies conducted seem to suggest that zerumbone induces cell death by stimulating apoptosis better than cisplatin, based on the significantly higher percentage of apoptotic cells in zerumbone's treated cancer cells as compared to cisplatin. In addition, zerumbone and cisplatin arrest cancer cells at G2/M phase as analyzed by flow cytometry. These results indicated that zerumbone significantly decreased the levels of IL-6 secreted by both cancer cells.

This study concludes that the compound, zerumbone, inhibits cancer cell growth through the induction of apoptosis, arrests cell-cycle at G2/M phase and inhibits the secretion levels of IL-6 in both cancer cells.

References

Abdelwahab SI, Abdul AB, Zain ZN, Hadi AH. (2012). Zerumbone inhibits interleukin-6 and induces apoptosis and cell-cycle arrest in ovarian and cervical cancer cells. Int Immunopharmacol,12(4):594-602. doi: 10.1016/j.intimp.2012.01.014.


Aggarwal B, Prasad S, Sung B, Krishnan S, Guha S. (2013). Prevention and Treatment of Colorectal Cancer by Natural Agents From Mother Nature. Curr Colorectal Cancer Rep, 9(1):37-56.


Huang YC, Chao KS, Liao HF, Chen YJ. (2013). Targeting sonic hedgehog signaling by compounds and derivatives from natural products. Evid Based Complement Alternat Med, 2013:748587. doi: 10.1155/2013/748587.


Sun Y, Sheng Q, Cheng Y, et al. (2013). Zerumbone induces apoptosis in human renal cell carcinoma via Gli-1/Bcl-2 pathway. Pharmazie, 68(2):141-5.


Weng HY, Hsu MJ, Wang CC, et al. (2012). Zerumbone suppresses IKK α , Akt, and FOXO1 activation, resulting in apoptosis of GBM 8401 cells. J Biomed Sci, 19:86. doi: 10.1186/1423-0127-19-86.

Hedyotis Diffusa Extract

Cancer: Colon

Action: CYP3A4 induction, inhibits angiogenesis

Hedyotis diffusa is a herb native to East Asia, particularly China, Japan, and Nepal.

Inhibition of tumor angiogenesis has become an attractive target of anti-cancer chemotherapy. However, drug resistance and cytotoxicity against non-tumor-associated endothelial cells limit the long-term use and the therapeutic effectiveness of angiogenesis inhibitors, thus increasing the necessity for the development of multi-target agents with minimal side effects. Hedyotis Diffusa Willd (EEHDW) has long been used as an important component in several TCM formulas to treat various types of cancer.

Inhibits Angiogenesis

The angiogenic effects of the ethanol extract of EEHDW were investigated, in order to find a molecular mechanism for its anti-cancer activity. It was found that EEHDW inhibited angiogenesis in vivo in chick embryo chorioallantoic membrane (CAM). In addition, EEHDW dose- and time-dependently inhibited the proliferation of human umbilical vein endothelial cells (HUVEC) by blocking the cell-cycle G1 to S progression.

Moreover, EEHDW inhibited the migration and tube formation of HUVECs. Furthermore, EEHDW treatment down-regulated the mRNA and protein expression levels of VEGF-A in HT-29 human colon carcinoma cells and HUVECs. These findings suggest that inhibiting tumor angiogenesis is one of the mechanisms by which EEHDW is involved in cancer therapy (Lin et al., 2011).

Colorectal Cancer

Hedyotis diffusa Willd has been used as a major component in several Chinese medicine formulas for the clinical treatment of colorectal cancer (CRC). The ethanol extract of Hedyotis diffusa Willd (EEHDW) reduced tumor volume and tumor weight, and suppressed STAT3 phosphorylation in tumor tissues, which in turn resulted in the promotion of cancer cell apoptosis and inhibition of proliferation. Moreover, EEHDW treatment altered the expression pattern of several important target genes of the STAT3 signaling pathway, i.e., decreased expression of Cyclin D1, CDK4 and Bcl-2 as well as up-regulated p21 and Bax (Cai et al., 2012).

EEHDW reduced HT-29 cell viability and survival in a dose- and time-dependent manner. Lin et al. (2012) observed that EEHDW treatment blocked the cell-cycle, preventing G1 to S progression, and reduced mRNA expression of pro-proliferative PCNA, Cyclin D1 and CDK4, but increased that of anti-proliferative p21 (Lin et al., 2012).

Recently, Lin et al. (2013) reported that HDW could inhibit colorectal cancer growth in vivo and in vitro via suppression of the STAT3 pathway. EEHDW could significantly reduce intratumoral microvessel density (MVD), indicating its activity of anti-tumor angiogenesis in vivo. EEHDW suppressed the activation of SHH signaling in CRC xenograft tumors since it significantly decreased the expression of key mediators of SHH pathway. EEHDW treatment inhibited the expression of the critical SHH signaling target gene VEGF-A as well as its specific receptor VEGFR2 (Lin et al., 2013).

CYP3A4 Induction

Patients are warned against the concomitant use of Oldenlandia diffusa and Rehmannia glutinosa, which could result in induction of CYP3A4, leading to a reduced efficacy of drugs that are CYP3A4 substrates and have a narrow therapeutic window (Lau et al., 2013).

References

Cai Q, Lin J, Wei L, Zhang L, et al. (2012). Hedyotis diffusa Willd Inhibits Colorectal Cancer Growth in Vivo via Inhibition of STAT3 Signaling Pathway. Int J Mol Sci, 13(5):6117-28. doi: 10.3390/ijms13056117.


Lau C, Mooiman KD, Maas-Bakker RF, et al. (2013). Effect of Chinese herbs on CYP3A4 activity and expression in vitro. J Ethnopharmacol, 149(2):543-9. doi: 10.1016/j.jep.2013.07.014.


Lin J, Wei L, Xu W, et al. (2011). Effect of Hedyotis Diffusa Willd extract on tumor angiogenesis. Mol Med Report, 4(6):1283-8. doi: 10.3892/mmr.2011.577.


Lin M, Lin J, Wei L, et al. (2012). Hedyotis diffusa Willd extract inhibits HT-29 cell proliferation via cell-cycle arrest. Exp Ther Med, 4(2):307-310.


Lin J, Wei L, Shen A, et al. (2013). Hedyotis diffusa Willd extract suppresses Sonic hedgehog signaling leading to the inhibition of colorectal cancer angiogenesis. Int J Oncol, 42(2):651-6. doi: 10.3892/ijo.2012.1753.

Dietary Flavones

Cancer:
Prostate, colorectal., breast, pancreatic, bladder, ovarian, leukemia, liver, glioma, osteosarcoma, melanoma

Action: Anti-inflammatory, TAM resistance, cancer stem cells, down-regulate COX-2, apoptosis, cell-cycle arrest, anti-angiogenic, chemo-sensitzer, adramycin (ADM) resistance

Sulforaphane, Phenethyl isothiocyanate (PEITC), quercetin, epicatechin, catechin, Luteolin, apigenin

Anti-inflammatory

The anti-inflammatory activities of celery extracts, some rich in flavone aglycones and others rich in flavone glycosides, were tested on the inflammatory mediators tumor necrosis factor α (TNF-α) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in lipopolysaccharide-stimulated macrophages. Pure flavone aglycones and aglycone-rich extracts effectively reduced TNF-α production and inhibited the transcriptional activity of NF-κB, while glycoside-rich extracts showed no significant effects.

Celery diets with different glycoside or aglycone contents were formulated and absorption was evaluated in mice fed with 5% or 10% celery diets. Relative absorption in vivo was significantly higher in mice fed with aglycone-rich diets as determined by HPLC-MS/MS (where MS/MS is tandem mass spectrometry). These results demonstrate that deglycosylation increases absorption of dietary flavones in vivo and modulates inflammation by reducing TNF-α and NF-κB, suggesting the potential use of functional foods rich in flavones for the treatment and prevention of inflammatory diseases (Hostetler et al., 2012).

Colorectal Cancer

Association between the 6 main classes of flavonoids and the risk of colorectal cancer was examined using data from a national prospective case-control study in Scotland, including 1,456 incident cases and 1,456 population-based controls matched on age, sex, and residence area.

Dietary, including flavonoid, data were obtained from a validated, self-administered food frequency questionnaire. Risk of colorectal cancer was estimated using conditional logistic regression models in the whole sample and stratified by sex, smoking status, and cancer site and adjusted for established and putative risk factors.

The significant dose-dependent reductions in colorectal cancer risk that were associated with increased consumption of the flavonols quercetin, catechin, and epicatechin, remained robust after controlling for overall fruit and vegetable consumption or for other flavonoid intake. The risk reductions were greater among nonsmokers, but no interaction beyond a multiplicative effect was present.

This was the first of several a priori hypotheses to be tested in this large study and showed strong and linear inverse associations of flavonoids with colorectal cancer risk (Theodoratou et al., 2007).

Anti-angiogenic, Prostate Cancer

Luteolin is a common dietary flavonoid found in fruits and vegetables. The anti-angiogenic activity of luteolin was examined using in vitro, ex vivo, and in vivo models. Angiogenesis, the formation of new blood vessels from pre-existing vascular beds, is essential for tumor growth, invasion, and metastasis; hence, examination of this mechanism of tumor growth is essential to understanding new chemo-preventive targets. In vitro studies using rat aortic ring assay showed that luteolin at non-toxic concentrations significantly inhibited microvessel sprouting and proliferation, migration, invasion and tube formation of endothelial cells, which are key events in the process of angiogenesis. Luteolin also inhibited ex vivo angiogenesis as revealed by chicken egg chorioallantoic membrane assay (CAM) and matrigel plug assay.

Pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, and TNF-α level were significantly reduced by the treatment of luteolin in PC-3 cells. Luteolin (10 mg/kg/d) significantly reduced the volume and the weight of solid tumors in prostate xenograft mouse model, indicating that luteolin inhibited tumorigenesis by targeting angiogenesis. Moreover, luteolin reduced cell viability and induced apoptosis in prostate cancer cells, which were correlated with the down-regulation of AKT, ERK, mTOR, P70S6K, MMP-2, and MMP-9 expressions.

Taken together, these findings demonstrate that luteolin inhibits human prostate tumor growth by suppressing vascular endothelial growth factor receptor 2-mediated angiogenesis (Pratheeshkumar et al., 2012).

Pancreatic Cancer; Chemo-sensitizer

The potential of dietary flavonoids apigenin (Api) and luteolin (Lut) were assessed in their ability to enhance the anti-proliferative effects of chemotherapeutic drugs on BxPC-3 human pancreatic cancer cells; additionally, the molecular mechanism of the action was probed.

Simultaneous treatment with either flavonoid (0,13, 25 or 50µM) and chemotherapeutic drugs 5-fluorouracil (5-FU, 50µM) or gemcitabine (Gem, 10µM) for 60 hours resulted in less-than-additive effect (p<0.05). Pre-treatment for 24 hours with 13µM of either Api or Lut, followed by Gem for 36 hours was optimal to inhibit cell proliferation. Pre-treatment of cells with 11-19µM of either flavonoid for 24 hours resulted in 59-73% growth inhibition when followed by Gem (10µM, 36h). Lut (15µM, 24h) pre-treatment followed by Gem (10µM, 36h), significantly decreased protein expression of nuclear GSK-3β and NF-κB p65 and increased pro-apoptotic cytosolic cytochrome c. Pre-treatment of human pancreatic cancer cells BxPC-3 with low concentrations of Api or Lut hence effectively aid in the anti-proliferative activity of chemotherapeutic drugs (Johnson et al., 2013).

Breast Cancer; Chemo-sensitizer, Tamoxifen

The oncogenic molecules in human breast cancer cells are inhibited by luteolin treatment and it was found that the level of cyclin E2 (CCNE2) mRNA was higher in tumor cells than in normal paired tissue samples as assessed using real-time reverse-transcriptase polymerase chain reaction (RT-PCR) analysis (n=257).

Combined treatment with 4-OH-TAM and luteolin synergistically sensitized the TAM-R cells to 4-OH-TAM. These results suggest that luteolin can be used as a chemo-sensitizer to target the expression level of CCNE2 and that it could be a novel strategy to overcome TAM resistance in breast cancer patients (Tu et al., 2013).

Breast Cancer

Consumers of higher levels of Brassica vegetables, particularly those of the genus Brassica (broccoli, Brussels sprouts and cabbage), reduce their susceptibility to cancer at a variety of organ sites. Brassica vegetables contain high concentrations of glucosinolates that can be hydrolyzed by the plant enzyme, myrosinase, or intestinal microflora to isothiocyanates, potent inducers of cytoprotective enzymes and inhibitors of carcinogenesis. Oral administration of either the isothiocyanate, sulforaphane, or its glucosinolate precursor, glucoraphanin, inhibits mammary carcinogenesis in rats treated with 7,12-dimethylbenz[a]anthracene. To determine whether sulforaphane exerts a direct chemo-preventive action on animal and human mammary tissue, the pharmacokinetics and pharmacodynamics of a single 150 µmol oral dose of sulforaphane were evaluated in the rat mammary gland.

Sulforaphane metabolites were detected at concentrations known to alter gene expression in cell culture. Elevated cytoprotective NAD(P)H:quinone oxidoreductase (NQO1) and heme oxygenase-1 (HO-1) gene transcripts were measured using quantitative real-time polymerase chain reaction. An observed 3-fold increase in NQO1 enzymatic activity, as well as 4-fold elevated immunostaining of HO-1 in rat mammary epithelium, provide strong evidence of a pronounced pharmacodynamic action of sulforaphane. In a subsequent pilot study, eight healthy women undergoing reduction mammoplasty were given a single dose of a broccoli sprout preparation containing 200 µmol of sulforaphane. Following oral dosing, sulforaphane metabolites were readily measurable in human breast tissue enriched for epithelial cells. These findings provide a strong rationale for evaluating the protective effects of a broccoli sprout preparation in clinical trials of women at risk for breast cancer (Cornblatt et al., 2007).

In a proof of principle clinical study, the presence of disseminated tumor cells (DTCs) was demonstrated in human breast tissue after a single dose of a broccoli sprout preparation containing 200 µmol of sulforaphane. Together, these studies demonstrate that sulforaphane distributes to the breast epithelial cells in vivo and exerts a pharmacodynamic action in these target cells consistent with its mechanism of chemo-protective efficacy.

Such efficacy, coupled with earlier randomized clinical trials revealing the safety of repeated doses of broccoli sprout preparations , supports further evaluation of broccoli sprouts in the chemoprevention of breast and other cancers (Cornblatt et al., 2007).

CSCs

Recent research into the effects of sulforaphane on cancer stem cells (CSCs) has drawn a great deal of interest. CSCs are suggested to be responsible for initiating and maintaining cancer, and to contribute to recurrence and drug resistance. A number of studies have indicated that sulforaphane may target CSCs in different types of cancer through modulation of NF- κB, SHH, epithelial-mesenchymal transition and Wnt/β-catenin pathways. Combination therapy with sulforaphane and chemotherapy in preclinical settings has shown promising results (Li et al., 2013).

Anti-inflammatory

Sulforaphane has been found to down-regulate COX-2 expression in human bladder transitional cancer T24 cells at both transcriptional- and translational levels. Cyclooxygenase-2 (COX-2) overexpression has been associated with the grade, prognosis and recurrence of transitional cell carcinoma (TCC) of the bladder. Sulforaphane (5-20 microM) induced nuclear translocation of NF-kappaB and reduced its binding to the COX-2 promoter, a key mechanism for suppressing COX-2 expression by sulforaphane. Moreover, sulforaphane increased expression of p38 and phosphorylated-p38 protein. Taken together, these data suggest that p38 is essential in sulforaphane-mediated COX-2 suppression and provide new insights into the molecular mechanisms of sulforaphane in the chemoprevention of bladder cancer (Shan et al., 2009).

Bladder Cancer

An aqueous extract of broccoli sprouts potently inhibits the growth of human bladder carcinoma cells in culture and this inhibition is almost exclusively due to the isothiocyanates. Isothiocyanates are present in broccoli sprouts as their glucosinolate precursors and blocking their conversion to isothiocyanates abolishes the anti-proliferative activity of the extract.

Moreover, the potency of isothiocyanates in the extract in inhibiting cancer cell growth was almost identical to that of synthetic sulforaphane, as judged by their IC50 values (6.6 versus 6.8 micromol/L), suggesting that other isothiocyanates in the extract may be biologically similar to sulforaphane and that nonisothiocyanate substances in the extract may not interfere with the anti-proliferative activity of the isothiocyanates. These data show that broccoli sprout isothiocyanate extract is a highly promising substance for cancer prevention/treatment and that its anti-proliferative activity is exclusively derived from isothiocyanates (Tang et al., 2006).

Ovarian Cancer

Sulforaphane is an extract from the mustard family recognized for its anti-oxidation abilities, phase 2 enzyme induction, and anti-tumor activity. The cell-cycle arrest in G2/M by sulforaphane and the expression of cyclin B1, Cdc2, and the cyclin B1/CDC2 complex in PA-1 cells using Western blotting and co-IP Western blotting. The anti-cancer effects of dietary isothiocyanate sulforaphane on ovarian cancer were investigated using cancer cells line PA-1.

Sulforaphane -treated cells accumulated in metaphase by CDC2 down-regulation and dissociation of the cyclin B1/CDC2 complex.

These findings suggest that, in addition to the known effects on cancer prevention, sulforaphane may also provide anti-tumor activity in established ovarian cancer (Chang et al., 2013).

Leukemia Stem Cells

Isolated leukemia stem cells (LSCs) showed high expression of Oct4, CD133, β-catenin, and Sox2 and imatinib (IM) resistance. Differentially, CD34(+)/CD38(-) LSCs demonstrated higher BCR-ABL and β-catenin expression and IM resistance than CD34(+)/CD38(+) counterparts. IM and sulforaphane (SFN) combined treatment sensitized CD34(+)/CD38(-) LSCs and induced apoptosis, shown by increased caspase 3, PARP, and Bax while decreased Bcl-2 expression. Mechanistically, imatinib (IM) and sulforaphane (SFN) combined treatment resensitized LSCs by inducing intracellular reactive oxygen species (ROS). Importantly, β-catenin-silenced LSCs exhibited reduced glutathione S-transferase pi 1 (GSTP1) expression and intracellular GSH level, which led to increased sensitivity toward IM and sulforaphane.

It was hence demonstrated that IM and sulforaphane combined treatment effectively eliminated CD34(+)/CD38(-) LSCs. Since SFN has been shown to be well tolerated in both animals and human, this regimen could be considered for clinical trials (Lin et al., 2012).

DCIS Stem Cells

A miR-140/ALDH1/SOX9 axis has been found to be critical to basal cancer stem cell self-renewal and tumor formation in vivo, suggesting that the miR-140 pathway may be a promising target for preventive strategies in patients with basal-like Ductal Carcinoma in Situ (DCIS). The dietary compound sulforaphane has been found to decrease Transcription factor SOX-9 and Acetaldehyde dehydrogenases (ALDH1), and thereby reduced tumor growth in vivo (Li et al., 2013).

Glioma, Prostate Cancer, Colon Cancer, Breast Cancer, Liver Cancer

Phenethyl isothiocyanate (PEITC), a natural dietary isothiocyanate, inhibits angiogenesis. The effects of PEITC were examined under hypoxic conditions on the intracellular level of the hypoxia inducible factor (HIF-1α) and extracellular level of the vascular endothelial growth factor (VEGF) in a variety of human cancer cell lines. Gupta et al., (2013) observed that PEITC suppressed the HIF-1α accumulation during hypoxia in human glioma U87, human prostate cancer DU145, colon cancer HCT116, liver cancer HepG2, and breast cancer SkBr3 cells. PEITC treatment also significantly reduced the hypoxia-induced secretion of VEGF.

Suppression of HIF-1α accumulation during treatment with PEITC in hypoxia was related to PI3K and MAPK pathways.

Taken together, these results suggest that PEITC inhibits the HIF-1α expression through inhibiting the PI3K and MAPK signaling pathway and provide a new insight into a potential mechanism of the anti-cancer properties of PEITC.

Breast Cancer Metastasis

Breast tumor metastasis is a leading cause of cancer-related deaths worldwide. Breast tumor cells frequently metastasize to brain and initiate severe therapeutic complications. The chances of brain metastasis are further elevated in patients with HER2 overexpression. The MDA-MB-231-BR (BR-brain seeking) breast tumor cells stably transfected with luciferase were injected into the left ventricle of mouse heart and the migration of cells to brain was monitored using a non-invasive IVIS bio-luminescent imaging system.

Results demonstrate that the growth of metastatic brain tumors in PEITC treated mice was about 50% less than that of control. According to Kaplan Meir's curve, median survival of tumor-bearing mice treated with PEITC was prolonged by 20.5%. Furthermore, as compared to controls, we observed reduced HER2, EGFR and VEGF expression in the brain sections of PEITC treated mice. These results demonstrate the anti-metastatic effects of PEITC in vivo in a novel breast tumor metastasis model and provides the rationale for further clinical investigation (Gupta et al., 2013).

Osteosarcoma, Melanoma

Phenethyl isothiocyanate (PEITC) has been found to induce apoptosis in human osteosarcoma U-2 OS cells. The following end points were determined in regard to human malignant melanoma cancer A375.S2 cells: cell morphological changes, cell-cycle arrest, DNA damage and fragmentation assays and morphological assessment of nuclear change, reactive oxygen species (ROS) and Ca2+ generations, mitochondrial membrane potential disruption, and nitric oxide and 10-N-nonyl acridine orange productions, expression and activation of caspase-3 and -9, B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax), Bcl-2, poly (adenosine diphosphate-ribose) polymerase, and cytochrome c release, apoptosis-inducing factor and endonuclease G. PEITC

It was therefore concluded that PEITC-triggered apoptotic death in A375.S2 cells occurs through ROS-mediated mitochondria-dependent pathways (Huang et al., 2013).

Prostate Cancer

The glucosinolate-derived phenethyl isothiocyanate (PEITC) has recently been demonstrated to reduce the risk of prostate cancer (PCa) and inhibit PCa cell growth. It has been shown that p300/CBP-associated factor (PCAF), a co-regulator for the androgen receptor (AR), is upregulated in PCa cells through suppression of the mir-17 gene. Using AR-responsive LNCaP cells, the inhibitory effects of PEITC were observed on the dihydrotestosterone-stimulated AR transcriptional activity and cell growth of PCa cells.

Expression of PCAF was upregulated in PCa cells through suppression of miR-17. PEITC treatment significantly decreased PCAF expression and promoted transcription of miR-17 in LNCaP cells. Functional inhibition of miR-17 attenuated the suppression of PCAF in cells treated by PEITC. Results indicate that PEITC inhibits AR-regulated transcriptional activity and cell growth of PCa cells through miR-17-mediated suppression of PCAF, suggesting a new mechanism by which PEITC modulates PCa cell growth (Yu et al., 2013).

Bladder Cancer; Adramycin (ADM) Resistance

The role of PEITC on ADM resistance reversal of human bladder carcinoma T24/ADM cells has been examined, including an increased drug sensitivity to ADM, cell apoptosis rates, intracellular accumulation of Rhodamine-123 (Rh-123), an increased expression of DNA topoisomerase II (Topo-II), and a decreased expression of multi-drug resistance gene (MDR1), multi-drug resistance-associated protein (MRP1), bcl-2 and glutathione s transferase π (GST-π). The results indicated that PEITC might be used as a potential therapeutic strategy to ADM resistance through blocking Akt and activating MAPK pathway in human bladder carcinoma (Tang et al., 2013).

Breast Cancer; Chemo-enhancing

The synergistic effect between paclitaxel (taxol) and phenethyl isothiocyanate (PEITC) on the inhibition of breast cancer cells has been examined. Two drug-resistant breast cancer cell lines, MCF7 and MDA-MB-231, were treated with PEITC and taxol. Cell growth, cell-cycle, and apoptosis were examined.

The combination of PEITC and taxol significantly decreased the IC50 of PEITC and taxol over each agent alone. The combination also increased apoptosis by more than 2-fold over each single agent in both cell lines. A significant increase of cells in the G2/M phases was detected. Taken together, these results indicated that the combination of PEITC and taxol exhibits a synergistic effect on growth inhibition in breast cancer cells. This combination deserves further study in vivo (Liu et al., 2013).

References

Chang CC, Hung CM, Yang YR, Lee MJ, Hsu YC. (2013). Sulforaphane induced cell-cycle arrest in the G2/M phase via the blockade of cyclin B1/CDC2 in human ovarian cancer cells. J Ovarian Res, 6(1):41. doi: 10.1186/1757-2215-6-41


Cornblatt BS, Ye LX, Dinkova-Kostova AT, et al. (2007). Preclinical and clinical evaluation of sulforaphane for chemoprevention in the breast. Carcinogenesis, 28(7):1485-1490. doi: 10.1093/carcin/bgm049


Gupta B, Chiang L, Chae K, Lee DH. (2013). Phenethyl isothiocyanate inhibits hypoxia-induced accumulation of HIF-1 α and VEGF expression in human glioma cells. Food Chem, 141(3):1841-6. doi: 10.1016/j.foodchem.2013.05.006.


Gupta P, Adkins C, Lockman P, Srivastava SK. (2013). Metastasis of Breast Tumor Cells to Brain Is Suppressed by Phenethyl Isothiocyanate in a Novel In Vivo Metastasis Model. PLoS One, 8(6):e67278. doi:10.1371/journal.pone.0067278


Hostetler G, Riedl K, Cardenas H, et al. (2012). Flavone deglycosylation increases their anti-inflammatory activity and absorption. Molecular Nutrition & Food Research, 56(4):558-569. doi: 10.1002/mnfr.201100596


Huang SH, Hsu MH, Hsu SC, et al. (2013). Phenethyl isothiocyanate triggers apoptosis in human malignant melanoma A375.S2 cells through reactive oxygen species and the mitochondria-dependent pathways. Hum Exp Toxicol. doi: 10.1177/0960327113491508


Johnson JL, Gonzalez de Mejia E. (2013). Interactions between dietary flavonoids apigenin or luteolin and chemotherapeutic drugs to potentiate anti-proliferative effect on human pancreatic cancer cells, in vitro. Food Chem Toxicol, 60:83-91. doi: 10.1016/j.fct.2013.07.036.


Li Q, Yao Y, Eades G, Liu Z, Zhang Y, Zhou Q. (2013). Down-regulation of miR-140 promotes cancer stem cell formation in basal-like early stage breast cancer. Oncogene. doi: 10.1038/onc.2013.226.


Li Y, Zhang T. (2013). Targeting cancer stem cells with sulforaphane, a dietary component from broccoli and broccoli sprouts. Future Oncol, 9(8):1097-103. doi: 10.2217/fon.13.108.


Lin LC, Yeh CT, Kuo CC, et al. (2012). Sulforaphane potentiates the efficacy of imatinib against chronic leukemia cancer stem cells through enhanced abrogation of Wnt/ β-catenin function. J Agric Food Chem, 60(28):7031-9. doi: 10.1021/jf301981n.


Liu K, Cang S, Ma Y, Chiao JW. (2013). Synergistic effect of paclitaxel and epigenetic agent phenethyl isothiocyanate on growth inhibition, cell-cycle arrest and apoptosis in breast cancer cells. Cancer Cell Int, 13(1):10. doi: 10.1186/1475-2867-13-10.


Pratheeshkumar P, Son YO, Budhraja A, et al. (2012). Luteolin inhibits human prostate tumor growth by suppressing vascular endothelial growth factor receptor 2-mediated angiogenesis. PLoS One, 7(12):52279. doi: 10.1371/journal.pone.0052279.


Tang K, Lin Y, Li LM. (2013). The role of phenethyl isothiocyanate on bladder cancer ADM resistance reversal and its molecular mechanism. Anat Rec (Hoboken), 296(6):899-906. doi: 10.1002/ar.22677.


Tang L, Zhang Y, Jobson HE, et al. (2006). Potent activation of mitochondria-mediated apoptosis and arrest in S and M phases of cancer cells by a broccoli sprout extract. Mol Cancer Ther, 5(4):935-44. doi: 10.1158/1535-7163.MCT-05-0476


Theodoratou E, Kyle J, Cetnarskyj R, et al. (2007). Dietary flavonoids and the risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev,16(4):684-93.


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


Shan Y, Wu K, Wang W, et al. (2009). Sulforaphane down-regulates COX-2 expression by activating p38 and inhibiting NF-kappaB-DNA-binding activity in human bladder T24 cells. Int J Oncol, 34(4):1129-34.


Yu C, Gong AY, Chen D, et al. (2013). Phenethyl isothiocyanate inhibits androgen receptor-regulated transcriptional activity in prostate cancer cells through suppressing PCAF. Mol Nutr Food Res. doi: 10.1002/mnfr.201200810.

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