Category Archives: MDR

Kanglaite injection (KLT)

Cancer: Lung, stomach, liver, kidney, breast, nasopharynx, esophagus, pancreas, colon-rectum, ovarian, prostate, lymphoma, leukemia

Action: Anti-tumoral, immunomodular, chemotherapy support, radiation support

Ingredients: yi yi ren (Coix Lacryma-jobi seed oil, CLSO).

Indications: primary NSCLC and primary liver cancer, which are not suitable for surgery, of qi and yin deficiency, lingering “Dampness due to Spleen deficiency types”. It has synergic effect when combined with radiotherapy or chemotherapy. It has certain anti-cachexia and analgesic effects for middle or late-stage tumor patients.

Dosage and usage:

Slow intravenous drip: 200 ml, once daily, 21 days as a course of treatment with 3-5 days interval.

When combined with radiotherapy or chemotherapy, the dosage can be reduced according to the practical conditions. (Drug Information Reference in Chinese, 2000. See end).

Invented by the famous pharmacological professor, Prof. Li Dapeng, Kanglaite Injection (KLT) has been listed by the Chinese government as a “State Basic Drug”, a “State Basic Medical Insurance Drug” and a “State Key New Drug”.

Based on pre-clinical studies at John Hopkins University, USA, tumor-inhibitive rate of KLT on transplanted breast carcinoma induced by cell strain MDA-MB-231 was over 50%. KLT could inhibit the expression of COX2 of the strain in vitro and act as an inhibitor of fatty acid synthase.

The broad ranged basic studies in China also revealed KLT different mechanisms such as inducing cancer cell apoptosis, inhibiting angiogenesis, reversing MDR and regulating gene expression of Fas/Apo-1 and Bcl-2.

Both Chinese and overseas clinical experiences have shown that KLT has proven effect in the treatment of cancers mainly at the sites of lung, breast, liver, nasopharynx, esophagus, stomach, pancreas, kidney, colon-rectum, ovary and prostate. This agent is also applied in the treatment of malignant lymphoma and acute leukemia. KLT has brought great benefits to over 500,000 cancer patients in more than 2,000 big or medium hospitals in China since 1997.

The year 1995 witnessed KLT patent certificates granted from China and the USA. In August 1997 the phase III clinical study was successfully completed and the injection was officially launched in China after final approval from the Ministry of Public Health.

Doctors in America carried out a phase 1 study of Kanglaite in 2003. They gave it to 16 people who had different types of cancer including lung, prostate and oesophageal cancers. The results showed people did not have many side-effects but the effect on their cancer varied. Some people showed no response, and their cancers continued to grow. But in others, the cancer stopped growing for a few months.

Standard treatment course for KLT is 200 ml (2 bottles) per day via intravenous drip x 42 days (84 bottles). There is a break for 4-5 days after 21 days. Clinical experiences in China and Russia suggest 2 treatment courses for those with late stage advanced and metastatic tumors for better therapeutic effect and evident prolongation of life (Conti, n.d.).

A consecutive cohort of 60 patients was divided into two groups, the experimental group receiving Kanglaite” Injection combined with chemotherapy and the control group receiving chemotherapy alone. After more than two courses of treatment, efficacy, quality of life and side-effects were evaluated. The response rate and KPS score of the experimental group were significantly improved as compared with those of the control group(P<0.05). In addition, gastrointestinal reactions and bone marrow suppression were significantly lower than in the control group(P<0.05). Kanglaite” Injection enhanced efficacy and reduced the side-effects of chemotherapy, improving quality of life of gastric cancer patients (Zhan et al., 2012).

Lung Cancer

C57BL/6 mice with Lewis lung carcinoma were divided into four groups: the control group (C), cisplatin group (1 mg/kg, DDP), low KLT group (6.25 ml/kg body weight [L]), and high KLT group (12.5 ml/kg body weight [H]). T cell proliferation was determined by the MTT assay. Nuclear factor-kappa B (NF-κB), inhibitor kappa B alpha

(IκBα), IκB kinase (IKK) and epidermal growth factor receptor (EGFR) levels were measured by western blotting. An enzyme-linked immunosorbent assay was used to analyze the expression of interleukin-2 (IL-2).

Intraperitoneal KLT significantly inhibited the growth of Lewis lung carcinoma, and the spleen index was significantly higher in the L and H groups than in the C group. KLT stimulated T cell proliferation in a dose-dependent manner. Treatment with KLT at either 6.25 or 12.5 ml/kg decreased the level of NF-κB in the nucleus in a dose-dependent manner, and KLT markedly decreased the expression of IκBα, IKK and EGFR in the cytoplasm of tumor cells and overall. IL-2 was significantly increased in the supernatant of splenocytes in the H group.

These results demonstrate that KLT has pronounced anti-tumor and immunostimulatory activities in C57BL/6 mice with Lewis lung carcinoma. These may affect the regulation of NF-κB/IκB expression, in addition to cytokines such as IL-2 and EGFR. Further work needs to investigate the relevant signaling pathway effects, but our findings suggest that KLT may be a promising anti-tumor drug for clinical use (Pan et al., 2012).

Skin Keratinocytes

Ultraviolet (UV) radiation plays an important role in the pathogenesis of skin photoaging. Depending on the wavelength of UV, the epidermis is affected primarily by UVB. One major characteristic of photoaging is the dehydration of the skin. Membrane-inserted water channels (aquaporins) are involved in this process. In this study we demonstrated that UVB radiation induced aquaporin-3 (AQP3) down-regulation in cultured human skin keratinocytes. Kanglaite is a mixture consisting of extractions of Coix Seed, which is an effective anti-neoplastic agent and can inhibit the activities of protein kinase C and NF-κB. We demonstrated that Kanglaite inhibited UVB-induced AQP3 down-regulation of cultured human skin keratinocytes. Our findings provide a potential new agent for anti-photoaging (Shan et al., 2012).

Hepatocellular Carcinoma

KLT produced an obvious time and dose-dependent inhibitory effect on HepG2 cells, and marked apoptosis was detected by FCM. The protein of Fas increased by 11.01%, 18.71%, 28.71% and 37.15%; the protein of FasL increased by 1.49%, 1.91%, 3.27% and 3.38% in comparison with the control (P<0.05). Real-time fluorescent quantitative RT-PCR showed that treating HepG2 cells with KLT caused the up-regulation of Fas and FasL mRNA. KLT inhibits HepG2 growth by inducing apoptosis, which may be mediated through activation of the Fas/FasL pathway (Lu et al., 2009).

Glomerular Nephritis

MTT, telomere repeat amplification protocol (TRAP), ELISA, PAGE and silver-stain were applied to detect the growth rate and telomerase activity of mesengial cell (MC) after stimulation of Kang Lai Te (KLT) and IL-1. The growth rate of MC was enhanced by IL-1 stimulation, which was accompanied with a reduction of the activity of telomerase. Adversely, the growth rate of MC was reduced by KLT, which was accompanied with an enhancement of activity of telomerase. Moreover, the growth rate of MC and the activity of telomerase were both inhibited by the combinative use of IL-1 and KLT without any influence from the sequence of their administration. KLT could inhibit proliferation and telomerase activity of MC with or without pre-stimulation with IL-1. KLT might be useful to prevent and treat glomerular nephritis related to MC proliferation (Hu et al., 2005).

Lung Metastasis

To screen the differential expression genes of Kanglaite in anti-tumor metastasis mRNA was extracted and purified from the lung of the mouse with LA795 lung metastasis, and hybridized respectively on 4 096-gene chip. cDNA microarray was scanned for the fluorescent signals and analyzing difference expression. Twenty-seven differential expressed genes were obtained.

Among these genes, 25 were up-regulated and 2 were down-regulated. Twelve of them were Mus musculus cDNA clone. Six genes related with genesis, development and metastasis of tumor. cDNA microarray for analysis of gene expression patterns is a powerful method to identify differential expressed genes. In this study, 6 genes are thought to be associated genes of Kanglaite in anti-tumor metastasis (Wu et al., 2003).

Lung Cancer; Chemo Side Effects

Sixteen reports were included in the meta-analysis. The quality of 16 studies was low. Pooling data of 5 studies indicated that the effect of Kanglaite+NP (Vinorelbine+Cisplatin) was better than NP with RR 1.46, 95% Confidence Interval 1.13 to 1.91. Pooling data of 3 studies of MVP (Mitomycin+Vindsine+ Cisplatin) plus Kanglaite indicated that the effect was better with RR 1.84, 95%CI 1.22 to 2.76. Pooling data of 2 studies showed that the effect of GP (Gemcitabine+Cisplatin) plus Kanglaite was better than GP with RR 1.63, 95%CI 1.09 to 2.43.

Fourteen studies revealed that Kanglaite may reduce the side-effects induced by regular treatment. Ten studies showed regular treatment plus Kanglaite can stabilize/improve quality of life (Zhu et al., 2009).

Apoptosis

Some studies show Kanglaite could inhibit some anti-apoptotic genes and activate some pro-apoptotic genes. Its injection solution is one of the new anti-cancer medicines that can significantly inhibit various kinds of tumor cells, so it has become the core of research into how to further explore KLT injection to promote tumor cell apoptosis by impacting on related genes (Lu et al., 2008).

References

Conti, M. (n.d.). Anti-cancer Chinese herbal kanglaite. Cancer Evolution. Retrieved from: http://www.cancerevolution.info/cancer-therapies/alternative-therapies/83-anticancer-chinese-herbal-kanglaite.html.


Hu, Y,H., Liang, W.K. Gong, Z.F. Xu,Q.L. Zou. (2005). The effect of kanglaite injection (KLT) on the proliferation and telomerase activity of rat mesangial cells. Zhongguo Zhong Yao Za Zhi, 30(6):450-453.


Lu, Y., Li, C.S., Dong, Q. (2008) Chinese herb related molecules of cancer-cell-apoptosis: a mini-review of progress between Kanglaite injection and related genes. J Exp Clin Cancer Res, 27:31. doi: 10.1186/1756-9966-27-31.


Lu, Y., L.Q. Wu, Q. Dong,C.S. Li. (2009). Experimental study on the effect of Kang-Lai-Te induced apoptosis of human hepatoma carcinoma cell HepG2. Hepatobiliary Pancreat Dis Int, 8(3):267-272.


Pan, P.,Y. Wu,Z.Y. Guo,R. et al. (2012). Anti-tumor activity and immunomodulatory effects of the intraperitoneal administration of Kanglaite in vivo in Lewis lung carcinoma. J Ethnopharmacol, 143(2):680-685.


Shan, S.J., Xiao T., Chen J., et al. (2012). Kanglaite attenuates UVB-induced down-regulation of aquaporin-3 in cultured human skin keratinocytes. Int J Mol Med, 29(4):625-629.


Wu, Y., Yang Y., Wu D. (2003). Study on the gene expression patterns of Kanglaite in anti-lung metastasis of LA795 mouse. Zhongguo Fei Ai Za Zhi, 6(6):473-476.


Zhan, Y.P., Huang X.E., Cao J. (2012). Clinical safety and efficacy of Kanglaite(R) (Coix Seed Oil) injection combined with chemotherapy in treating patients with gastric cancer. Asian Pac J Cancer Prev, 13(10):5319-5321.


Zhu, L.Z. Yang, S. Wang, Y. Tang. (2009). Kanglaite for Treating Advanced Non-small-cell Lung Cancer: A Systematic Review. Zhongguo Fei Ai Za Zhi, 12(3):208-215.

Decursin

Cancer: Prostate, breast, fibrosarcoma, sarcoma

Action: MDR, inflammation, anti-cancer, angiogenesis

Decursin is isolated from Angelica gigas (Nakai).

Angelica gigas NAKAI is used to treat dysmenorrhea, amenorrhea, menopause, abdominal pain, injuries, migraine, and arthritis. The physicochemical and toxicological characterization of compounds in A. gigas NAKAI, decursin, decursinol angelate, diketone decursin, ether decursin, epoxide decursin and oxim decursin, have been extensively studied (Mahat et al., 2012).

Sarcoma; Anti-cancer

The in vivo anti-tumor activities of decursinol angelate (1) and decursin (2) isolated from the roots of Angelica gigas were investigated. These two compounds, when administered consecutively for 9 days at 50 and 100 mg/kg i.p. in mice, caused a significant increase in the life span and a significant decrease in the tumor weight and volume of mice inoculated with Sarcoma-180 tumor cells. These results suggest that decursinol angelate (1) and decursin (2) from A. gigas have anti-tumor activities (Lee et al., 2003).

Fibrosarcoma

Decursin and related coumarin compounds in herbal extracts have a number of biological activities against inflammation, angiogenesis and cancer. The human fibrosarcoma cell line, HT1080, was treated with TNFα (tumor necrosis factor α) in the presence or absence of CSL-32. Treatment of HT1080 cells with a derivative of decursin (CSL-32) inhibited their proliferation, without affecting cell viability, and TNF α-induced expression of pro-inflammatory mediators, such as MMP-9 (matrix metalloproteinase-9) and IL-8 (interleukin-8) (Lee et al., 2012).

Prostate Cancer

Androgen and androgen receptor (AR) signaling are crucial for the genesis of prostate cancer (PCa), which can often develop into androgen-ligand-independent diseases that are lethal to patients. As current chemotherapy is largely ineffective for PCa and has serious toxic side-effects, a collaborative effort has been initiated to identify and develop novel, safe and naturally occurring agents that target AR signaling from Oriental medicinal herbs for the chemoprevention and treatment of PCa. The discovery of decursin from an Oriental formula containing Korean Angelica gigas Nakai (Dang Gui) root as a novel anti-androgen/AR agent has been highlighted and the mechanisms to account for the specific anti-AR actions have been identified: rapid block of AR nuclear translocation, inhibition of binding of 5-dihydrotestesterone to AR, and increased proteasomal degradation of AR protein. Structure-activity analyzes reveal a critical requirement of the side-chain on decursin or its structural isomer decursinol angelate for anti-AR, cell-cycle arrest and pro-apoptotic activities.

This work demonstrates the feasibility of using activity-guided fractionation in cell culture assays combined with mechanistic studies to identify novel anti-androgen/AR agents from complex herbal mixtures (Lu et al., 2007).

MDR

Combination cancer therapy is one of the attractive approaches to overcome drug resistance of cancer cells. In the present study, Jang et al (2013) investigated the synergistic effect of decursin from Angelica gigas and doxorubicin on the induction of apoptosis in three human multiple myeloma cells. The combined treatment reduced mitochondrial membrane potential., suppressed the phosphorylation of JAK2, STAT3, and Src, activated SHP-2, and attenuated the expression of cyclind-D1 and survivin in U266 cells.

Overall, the combination treatment of decursin and doxorubicin can enhance apoptotic activity via mTOR and/or STAT3 signaling pathway in multiple myeloma cells.

Breast Cancer

Decursin significantly reduced protein expression and enzymatic activity of Pin1 in MDA-MB-231 cells. Kim et al (2013) found that decursin treatment enhanced the p53 expression level and failed to down-regulate Pin1 in the cells transfected with p53 siRNA, indicating the importance of p53 in the decursin-mediated Pin1 inhibition in MDA-MB-231 cells. Decursin stimulated association between peptidyl-prolyl cis/trans isomerase Pin1 to p53. Moreover, decursin facilitated p53 transcription in MDA-MB-231 cells. Overall, the study suggests the potential of decursin as an attractive cancer therapeutic agent for breast cancer by targeting Pin1.

References

Jang J, Jeong SJ, Kwon HY, Jung JH, et al. (2013). Decursin and Doxorubicin Are in Synergy for the Induction of Apoptosis via STAT3 and/or mTOR Pathways in Human Multiple Myeloma Cells. Evid Based Complement Alternat Med. 2013:506324. doi: 10.1155/2013/506324.

Kim JH, Jung JH, Kim SH, Jeong SJ. (2013). Decursin Exerts Anti-cancer Activity in MDA-MB-231 Breast Cancer Cells Via Inhibition of the Pin1 Activity and Enhancement of the Pin1/p53 Association.Phytother Res. doi: 10.1002/ptr.4986.

Lee S, Lee YS, Jung SH, et al. (2003). Anti-tumor activities of decursinol angelate and decursin from Angelica gigas. Arch Pharm Res, 26(9):727-30.

Lee SH, Lee JH, Kim EJ, et al. (2012). A novel derivative of decursin, CSL-32, blocks migration and production of inflammatory mediators and modulates PI3K and NF- κB activities in HT1080 cells. Cell Biol Int, 36(7):683-8. doi: 10.1042/CBI20110257.

Lu JX, Kim SH, Jiang C, Lee JJ, Guo JM. (2007). Oriental herbs as a source of novel anti-androgen and prostate cancer chemo-preventive agents. Acta Pharmacologica Sinica, 28, 1365–1372. doi:10.1111/j.1745-7254.2007.00683.x

Mahat B, Chae JW, Baek IH, et al. (2012). Physicochemical characterization and toxicity of decursin and their derivatives from Angelica gigas. Biol Pharm Bull, 35(7):1084-90.

Artesunate

Cancer: Colon, esophageal., pancreatic, ovarian, multiple myeloma and diffuse large B-cell lymphoma, osteosarcoma, lung, breast, skin, leukemia/lymphoma

Action: Anti-metastatic, MDR, radio-sensitizer

Pulmonary Adenocarcinomas

Artesunate exerts anti-proliferative effects in pulmonary adenocarcinomas. It mediates these anti-neoplastic effects by virtue of activating Bak (Zhou et al., 2012). At the same time, it down-regulates epidermal growth factor receptor expression. This results in augmented non-caspase dependent apoptosis in the adenocarcinoma cells. Artesunate mediated apoptosis is time as well as dose-dependent. Interestingly, AIF and Bim play significant roles in this Bak-dependent accentuated apoptosis (Ma et al., 2011). Adenosine triphosphate (ATP)-binding cassette subfamily G member 2 (ABCG2) expression is also attenuated while transcription of matrix metallopeptidase 7 (MMP-7) is also down-regulated (Zhao et al., 2011). In addition, arsenuate enhances the radio-sensitization of lung carcinoma cells. It mediates this effect by down-regulating cyclin B1 expression, resulting in augmented G2/M phase arrest (Rasheed et al., 2010).

Breast Cancer

Similarly, artesunate exhibits anti-neoplastic effects in breast carcinomas. Artesunate administration is typically accompanied by attenuated turnover as well as accentuated peri-nuclear localization of autophagosomes in the breast carcinoma cells. Mitochondrial outer membrane permeability is typically augmented. As a result, artesunate augments programmed cellular decline in breast carcinoma cells (Hamacher-Brady et al., 2011).

Skin Cancer

Artesunate also exerts anti-neoplastic effects in skin malignancies. It mediates these effects by up-regulating p21. At the same time it down-regulates cyclin D1 (Jiang et al., 2012).

Colon Cancer

Artemisunate significantly inhibited both the invasiveness and anchorage independence of colon cancer SW620 cells in a dose-dependent manner. The protein level of intercellular adhesion molecule 1 (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).

Multi-drug resistance; Colon Cancer

A profound cytotoxic action of the antimalarial., artesunate (ART), was identified against 55 cancer cell lines of the U.S. National Cancer Institute (NCI). The 50% inhibition concentrations (IC50 values) for ART correlated significantly to the cell doubling times (P = 0.00132) and the portion of cells in the G0/G1 (P = 0.02244) or S cell-cycle phases (P = 0.03567).

Efferth et al., (2003) selected mRNA expression data of 465 genes obtained by microarray hybridization from the NCI data-base. These genes belong to different biological categories (drug resistance genes, DNA damage response and repair genes, oncogenes and tumor suppressor genes, apoptosis-regulating genes, proliferation-associated genes, and cytokines and cytokine-associated genes). The constitutive expression of 54 of 465 (=12%) genes correlated significantly to the IC50 values for ART. Hierarchical cluster analysis of these 12 genes allowed the differentiation of clusters with ART-sensitive or ART-resistant cell lines (P = 0.00017).

Multi-drug-resistant cells differentially expressing the MDR1, MRP1, or BCRP genes were not cross-resistant to ART. ART acts via p53-dependent and- independent pathways in isogenic p53+/+ p21WAF1/CIP1+/+, p53-/- p21WAF1/CIP1+/+, and p53+/+ p21WAF1/CIP1-/- colon carcinoma cells.

Multi-drug resistance; Esophageal Cancer

The present study aimed to investigate the correlation between ABCG2 expression and the MDR of esophageal cancer and to estimate the therapeutic benefit of down-regulating ABCG2 expression and reversing chemoresistance in esophageal cells using artesunate (ART).

ART is a noteworthy antimalarial agent, particularly in severe and drug-resistant cancer cases, as ART is able to reverse drug resistance. 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, but still remains largely unknown (Liu, Zuo, & Guo, 2013).

Pancreatic Cancer

The combination of triptolide and artesunate could inhibit pancreatic cancer cell line growth, and induce apoptosis, accompanied by expression of HSP 20 and HSP 27, indicating important roles in the synergic effects. Moreover, tumor growth was decreased with triptolide and artesunate synergy. Results indicated that triptolide and artesunate in combination at low concentrations can exert synergistic anti-tumor effects in pancreatic cancer cells with potential clinical applications (Liu & Cui, 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.

Pelvic and extrapelvic OVCA implants demonstrated similar patterns of signaling pathway expression and identical p53 mutations.

However, Marchion et al. (2013) identified 3 molecular pathways/cellular processes that were differentially expressed between pelvic and extrapelvic OVCA samples and between primary/early-stage and metastatic/advanced or recurrent ovarian, oral., and prostate cancers. Furthermore, their expression was associated with overall survival from ovarian cancer (P = .006), colon cancer (1 pathway at P = .005), and leukemia (P = .05). Artesunate-induced TGF-WNT pathway inhibition impaired OVCA cell migration.

Multiple Myeloma, B-cell Lymphoma

Findings indicate that artesunate is a potential drug for treatment of multiple myeloma and diffuse large B-cell lymphoma (DLBCL) at doses of the same order as currently in use for treatment of malaria without serious adverse effects. Artesunate treatment efficiently inhibited cell growth and induced apoptosis in cell lines. Apoptosis was induced concomitantly with down-regulation of MYC and anti-apoptotic Bcl-2 family proteins, as well as with cleavage of caspase-3. The IC50 values of artesunate in cell lines varied between 0.3 and 16.6 µm. Furthermore, some primary myeloma cells were also sensitive to artesunate at doses around 10 µm. Concentrations of this order are pharmacologically relevant as they can be obtained in plasma after intravenous administration of artesunate for malaria treatment (Holien et al., 2013).

Osteosarcoma, Leukemia/Lymphoma

Artesunate inhibits growth and induces apoptosis in human osteosarcoma HOS cell line in vitro and in vivo (Xu et al. 2011). ART alone or combined with chemotherapy drugs could inhibit the proliferation of B/T lymphocytic tumor cell lines as well ALL primary cells in vitro, probably through the mechanism of apoptosis, which suggest that ART is likely to be a potential drug in the treatment of leukemia/lymphoma (Zeng et al., 2009).

References

Efferth, T., Sauerbrey, A., Olbrich, A., et al. (2003) Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol, 64(2):382-94.


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


Hamacher-Brady, A., Stein, H.A., Turschner, S., et al. (2011). Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J Biol Chem. 2011;286(8):6587–6601. doi: 10.1074/jbc.M110.210047.


Holien, T., Olsen, O.E., Misund, K., et al. (2013). Lymphoma and myeloma cells are highly sensitive to growth arrest and apoptosis induced by artesunate. Eur J Haematol, 91(4):339-46. doi: 10.1111/ejh.12176.


Jiang, Z., Chai, J., Chuang, H.H., et al. (2012). Artesunate induces G0/G1 cell-cycle arrest and iron-mediated mitochondrial apoptosis in A431 human epidermoid carcinoma cells. Anti-cancer Drugs, 23(6):606–613. doi: 10.1097/CAD.0b013e328350e8ac.


Liu, L., Zuo, L.F., Guo, J.W. (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.


Liu, Y. & Cui, Y.F. (2013). Synergism of cytotoxicity effects of triptolide and artesunate combination treatment in pancreatic cancer cell lines. Asian Pac J Cancer Prev, 14(9):5243-8.


Ma, H., Yaom Q., Zhang, A.M., et al. (2011). The effects of artesunate on the expression of EGFR and ABCG2 in A549 human lung cancer cells and a xenograft model. Molecules, 16(12):10556–10569. doi: 10.3390/molecules161210556.


Marchion, D.C., Xiong, Y., Chon, H.S., 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.


Rasheed, S.A., Efferth, T., Asangani, I.A., Allgayer, H. (2010). First evidence that the antimalarial drug artesunate inhibits invasion and in vivo metastasis in lung cancer by targeting essential extracellular proteases. Int J Cancer, 127(6):1475–1485. doi: 10.1002/ijc.25315.


Xu, Q., Li, Z.X., Peng, H.Q., et al. (2011). Artesunate inhibits growth and induces apoptosis in human osteosarcoma HOS cell line in vitro and in vivo. J Zhejiang Univ-Sci B (Biomed & Biotechnol), 12(4):247–255. doi: 10.1631/jzus.B1000373.


Zhao, Y., Jiang, W., Li, B., et al. (2011). Artesunate enhances radiosensitivity of human non-small-cell lung cancer A549 cells via increasing no production to induce cell-cycle arrest at G2/M phase. Int Immunopharmacol, 11(12):2039–2046. doi: 10.1016/j.intimp.2011.08.017.


Zeng, Y., Ni, X., Meng, W.T., Wen, Q., Jia, Y.Q. (2009). Inhibitive effect of artesunate on human lymphoblastic leukemia/lymphoma cells. Sichuan Da Xue Xue Bao Yi Xue Ban, 40(6):1038-43.


Zhou, C., Pan, W., Wang, X.P., Chen, T.S. (2012). Artesunate induces apoptosis via a bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells. J Cell Physiol, 227(12):3778–3786. doi: 10.1002/jcp.24086.

Multi-drug resistance

Multi-drug resistance in cancer chemotherapy refers to the ability of cancer cells to survive from treatment of a wide range of drugs (Meszaros et al., 2009).

In addition to the MDR induced by drugs in early exposure, the MDR cancer cells may subsequently develop cross-resistance to several unexposed and structurally unrelated chemotherapeutic agents (Biedler et al., 1970).

How to tackle the MDR cells in chemotherapy is a pressing issue in cancer treatments. Verapamil was the first known Pgp inhibitor to increase the intracellular concentration of anti-cancer agents in MDR cells by binding to Pgp and inhibiting the Pgp-mediated efflux (Twentyman, 1992). It was believed that anti-cancer drug resistance could be reversed by drug efflux inhibition. Researchers developed and tested a range of Pgp inhibitors to improve the pharmacological effects of chemotherapy in cancer patients (Tsuruo et al., 1981; Stewart et al., 2000; Toppmeyer et al., 2002).

Mechanisms of MDR include decreased uptake of drugs, alterations in cellular pathways and increased active efflux of drugs (Gottesman, 2002; La Porta, 2007; Watson, 1991).

Overexpression of ATP-binding cassette (ABC) transporters is one of the most common mechanisms. Overexpression of the three major ABC transporters, i.e. P-glycoprotein (Pgp), multi-drug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP/ABCG2), is frequently observed in cancer cell lines selected with chemotherapeutic drugs (Szakacs et al., 2006) and critical to clinical drug resistance (Leonard, 2003).

Fractions from 17 clinically used anti-tumor traditional Chinese medicinal herbs were tested for their potential to restore the sensitivity of MCF-7/ADR and A549/Taxol cells to a known anti-neoplastic agent. Five herbs, Curcuma wenyujin, Chrysanthemum indicum, Salvia chinensis, Ligusticum chuanxiong Hort. and Cassia tora L., could sensitize these resistant cancer cells at a non-toxic concentration (10  µg mL–1), and markedly increased doxorubicin accumulation in MCF-7/ADR cells, which necessitates further investigations into the active ingredients of these herbs and their underlying mechanisms (Yang et al., 2011).

Natural sources are a fertile ground to find novel drugs with activity against MDR cancer cells. In some countries, especially China, traditional herbal medicines are often used together with mainstream chemotherapeutic agents. The clinically used traditional Chinese herbs for the treatment of tumor can be classified into four categories based on the theory of Traditional Chinese Medicine (TCM): drugs (CH group) for 'Clearing away Heat and Toxins', drugs (PB group) for 'Promoting Blood Flow to Remove Stasis', drugs for 'Invigoration' and toxic drugs. Drugs for 'Invigoration' have indirect anti-neoplastic action by enhancing an organism's immunity and have been used clinically to minimise radiotherapy- and chemotherapy-induced toxicity (Fu & Chen, 2008; Chai, To, Lin, 2010).

Some of the recent findings on the circumvention of ABC transporters-mediated MDR by various ingredients and extracts of CM and their formulae, based on whether the MDR reversal involved Pgp alteration, are reviewed below.

Saponins

Ginsenosides are the major active components from Panax ginseng (Renshen). Ginsenosides are mainly triterpenoid dammarane derivatives. Several ginsenosides, namely Rg1, Rg3, Re, Rc and Rd inhibited drug efflux (Kim et al., 2003). A combination of purified saponins containing Rb1, Rb2, Rc, Rd, Re and Rg1 reversed MDR whereas individual ginsenosides did not produce any effect (Park et al., 2006). Ginsenosides reversed MDR of several chemotherapeutic drugs such as homoharringtonine, cytarabine, doxorubicin and etoposide in K562/VCR and in a dose-dependent manner in K562/DOX (Gao et al., 2004).

Pgp expression decreased but bcl-2 expression remained the same (Wang, 2003). Rb1 reversed MDR of harringtonolide and vincristine in K562/HHT and HL60/VCR cell lines respectively (Shi et al. , 2005).

Panax notoginseng (Sanqi) total saponins reversed MDR of doxorubicin in MCF-7/DOX and K562/VCR cell lines. The mechanism may be related to the decrease of Pgp expression (Si & Tien, 2005; Liu, Liu, & Fang, 2008).

Rg3, one of the active ginsenosides from Panax ginseng, restored the sensitivity of resistant KBV20 cell line to various anti-cancer drugs, including vincristine, doxorubicin, etoposide and colchicine in a time-and dose-dependent manner. This ginsenoside competitively inhibited the binding of substrate drugs to Pgp and its binding affinity to Pgp was remarkably higher than that of verapamil. In contrast to the dose-dependent effects in vitro, Rg3 increased animal life span in an in vivo MDR model in a dose-independent manner (Kim et al., 2003).

Flavonoids

Quercetin is one of the most widely distributed flavonoids in natural products including Chinese medicinal herbs such as Sophora japonica (Huai). Quercetin inhibited the binding of heat shock factor at the MDR1 promoter, thereby decreasing MDR1 transcription and reducing Pgp expression (Kim et al., 1998). Quercetin also inhibited the overexpression of Pgp mediated by arsenite (Kioka et al., 1992). In HL-60/DOX and K562/DOX cell lines, quercetin enhanced the anti-cancer sensitivity to daunorubicin and decreased Pgp expression (Cai et al., 2004; Cai et al., 2005). MDR reversal effect of quercetin was probably mediated by its action on mitochondrial membrane potential and the induction of apoptosis. Furthermore, quercetin derivatives rather than quercetin itself reversed MDR (Kothan et al., 2004). Quercetin increased the sensitivity of Pgp-overexpressing KBV1 cell line towards vinblastine and paclitaxel in a dose-dependent manner. Among many active flavonoids, quercetin was less potent than kaempferol but more effective than genistein and daidzein in reversing MDR. Genistein and daidzein had no effect on Pgp expression (Limtrakul, Khantamat, & Pintha, 2005).

Although quercetin may be a potential MDR reversing agent, lethal drug-drug interaction between quercetin and digoxin has been reported. Quercetin (40 mg/kg) elevated the peak blood concentration of digoxin and caused sudden death of tested animals (Wang et al., 2004).

Paeonol is a weak calcium channel blocker isolated from the root of Paeonia suffruticosa (Mudan). In K562/DOX cell line, paeonol showed positive MDR reversal effect towards doxorubicin, daunorubicin, vincristine and vinblastine without modulating Pgp expression [100]. In parental K562 cells, paeonol induced apoptosis in a time-and dose-dependent manner (Sun et al., 2004).

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 & Amiji, 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 (Um et al., 2008). 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 (Um et al., 2008). Bisdemethoxycurcumin modified from curcumin resulted in greater inhibition of Pgp expression (Limtrakul, Anuchapreeda, & Buddhasukh, 2004). Tetrahydrocurcumin, the major metabolite of curcumin, inhibited all three major ABC transporters (Limtrakul et al., 2007). Curcumin induced atypical and caspase-independent cell death in MDR cells (Piwocka, Bielak-Mijewska, & Sikora, 2002). In leukaemic cells collected from 78 childhood leukaemia patients, curcumin reduced Pgp expression (Anuchapreeda et al., 2006). A specialized nanoemulsion of curcumin is better than conventional solution form drugs in enhancing the efficiency of drug delivery into the cells, down-regulating Pgp expression, inhibiting the NFκB pathway and promoting apoptotic response (Choi et al., 2008).

Other Compounds

Schizandrins, the active constituents of Schisandra chinensis (Wuweizi), were investigated for their MDR reversal effects. Schizandrin A was the most potent in reversing MDR by enhancing apoptosis and down-regulating Pgp and total protein kinase C expression. The crude extract of Schisandra chinensis reversed the resistance against vincristine in vivo (Huang et al., 2008). Deoxyschizandrin and γ-schizandrin, among the nine dibenzo[a,c]cyclooctadiene lignans examined, enhanced intracellular drug concentration and induced cell-cycle arrest at the G2/M phase when combined with sub-toxic dosages of doxorubicin (Slaninová et al., 2009). Gomisin A, on the other hand, altered Pgp-substrate interaction by binding to Pgp simultaneously with substrates (Wan et al., 2006).

Formulae – injections (See Injectables)

'Shengmai Injection', consisting of Panax ginseng and Ophiopogon japonicus (Maidong), down-regulated Pgp expression in peripheral blood lymphocyte membrane. When used together with oxaliplatin, 5-fluorouracil or folinic acid, the injection prolonged the survival rate of colon cancer patients (Cao et al., 2005). The injection also enhanced the efficacy of tamoxifen and nifedipine in combination therapy (Lin et al., 2002).

'KLT Injection' consisting of the extract of Coix lacryma-jobi (Yiyi) enhanced the anti-cancer activities of paclitaxel and docetaxel and reversed MDR in a dose-dependent manner (Dong, Zheng, & Lu, 2002).

Formulae – powders

'Shenghe Powder', consisting of Panax ginseng, Scorophularia ningpoensis (Xuanshen) and Atractylodes macrocephala (Baizhu), increased the intracellular concentration of vincristine in resistant SGC-7901/VCR cell line, possibly due to the induction of apoptosis and down-regulation of Pgp and bcl-2 expression (Wang et al., 2007).

'Modified Sanwubai Powder', consisting of herbs such as Croton tiglium (Badou), Platycodon grandiflorum (Jiegeng) and Fritillaria thunbergii, induced apoptosis in SGC-7901 cell line and down-regulated the gene expressions of p53, bcl-2, rasP21CD44 and Pgp (Xu et al., 2005).

Formulae – others

Three herbal extracts used to treat diseases other than cancer, namely Ams-11, Fw-13 and Tul-17, greatly enhanced the efficacy of vincristine both in vitro and in vivo and reversed MDR in a dose-dependent manner. Tul-17 inhibited Pgp expression (Qu et al., 2006).

Oil emulsion from Brucea javanica (Yadanzi) reversed MDR when used together with other chemotherapeutic drugs such as vincristine, doxorubicin, cisplatin, mitomycin C, 5-fluorouracil or etoposide, probably due to down-regulation of Pgp expression or inhibition of TOPO II or both (Yu, Wu, Zhang, 2001).

'Sangeng Mixture Decoction', consisting of Reynoutria japonica (Huzhang), Actinidia arguta (Mihouligen) and Geum aleppicum (Shuiyangmeigen), reversed MDR of doxorubicin via down-regulation of Pgp expression (Feng et al., 2003).

FFTLG, a formula containing Actinidia arguta, reversed MDR in K562/DOX cell line by increasing the intracellular doxorubicin concentration (Guo, Xie, Feng, 2002).

R1, consisting of Ligusticum chuanxiong, Curcuma longa and Millettia dielsiana (Jixueteng), enhanced the anti-cancer activities of doxorubicin in MCF-7/DOX via down-regulation of Pgp expression (Chen et al., 2003; Lin, 2007).

Formulae

'Ganli Injection', consisting of matrine and tetramethylpyazine hydrochloride, reversed MDR by increasing the sensitivity of 5-fluorouracil and the intracellular concentration of doxorubicin in BEL-7402/5-FU cell line (Gu et al., 2007).

'Bushen Huayu Jiedu Formula', consisting of Cinnamomum cassia (Rougui), Psoralea corylifolia (Buguzhi) and Rheum palmatum, was tested in A549/DDP cell line and S180 tumor-bearing mice. In vitro, the formula significantly increased the intracellular concentration of cisplatin at high doses and inhibited the activity of calcium channel and LRP-56 expression at both high and low doses. In vivo, the formula improved the serum concentration, reduced the inflow and the release of Ca2+ and inhibited the LRP gene expression (Cao et al., 2004; Cao et al., 2008).

Four CM formulae, namely Glycyrrhiza glabra (GLYC), Hedyotis diffusa (OLEN), a formula consisting of 15 herbs including Cistanche deserticola (Roucongrong), Rabdosia rubescens (Donglingcao) and Zanthoxylum nitidum (Liangmianzhen) (SPES), and a formula consisting of eight herbs including Serenoa repens (Juyezhong), Scutellaria baicalensis (Huangqin), Panax ginseng and Glycyrrhiza glabra (PC-SPES) were cytotoxic to cancer cell lines in a dose-dependent manner. SPES, PC-SPES, OLEN decreased the bcl-2 gene expression and were pro-apoptotic, while GLYC was pro-necrotic without altering the over-expression of bcl-2 in MDR cells. Furthermore, OLEN, SPES and PC-SPES exhibited similar pharmacological effects to etoposide and vincristine (Sadava et al., 2002).

References

Anuchapreeda S, Thanarattanakorn P, Sittipreechacharn S, et al. (2006). Inhibitory effect of curcumin on MDR1 gene expression in patient leukemic cells. Arch Pharm Res, 29(10):866-873

Biedler JL, Riehm H. (1970). Cellular resistance to actinomycin D in Chinese hamster cells in vitro: cross-resistance, radioautographic, and cytogenetic studies. Cancer Res, 30:1174-1184.

Cai X, Chen FY, Han JY, et al. (2004). Restorative effect of quercetin on subcellular distribution of daunorubicin in Multi-drug-resistant leukemia cell lines K562/ADM and HL-60/ADM. Chin J Cancer, 23(12):1611-1615.

Cai X, Chen FY, Han JY, et al. (2005). Reversal of Multi-drug resistance of HL-60 adriamycin resistant leukemia cell line by quercetin and its mechanisms. Chin J Oncol, 27(6):326-329.

Cao CM, Ding XD, Wang XH, Liu P. (2005). Clinical study of shengmai injection in its reversing MDR effect in late phase colon carcinoma patients. Shandong J Tradit Chin Med, 24(9):529-532.

Cao Y, Zhang D, Zheng GJ, Yang Y, Zhang J. (2004). Study on drug resistance reversion and mechanism of bushen huayu jiedu formula in lung cancer cells of drug resistance. Shandong J Trad Chin Med, 23(2):100-104.

Cao Y, Xia Q, Meng H, Zhong A. (2008). Pharmacological effects of serum containing chinese medicine bushen huayu jiedu compound recipe in lung cancer drug-resistance cells. Chin J Integr Med, 14(1):46-50.

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.

Choi BH, Kim CG, Lim Y, Shin SY, Lee YH. (2008). Curcumin down-regulates the Multi-drug resistance mdr1b gene by inhibiting the PI3K/Akt/NF kappa B pathway. Cancer Lett, 259(1):111-118.

Chen XY, Liu JT. (2003). Study on tumor cells' Multi-drug resistance and its reversion by Chinese herbs. J Chin Integr Med, 1(3):221-225.

Dong QH, Zheng S, Lu QH. (2002). Study on the effect of Kanglaite injection on MDR human leukemia cell lines. J Pract Oncol, 17(1):24-26.

Feng ZQ, Guo Y, Zhu NX, et al. (2003). The experiment of SANGENG mixture decoction on reversing Multi-drug resistance. Bull Chin Cancer, 12(6):370-371.

Fu YL and Chen T. (2008). Research of traditional Chinese medicine in the treatment of leukemia: current status. Journal of Chinese Integrative Medicine, 6: 867–872.

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.

Gao RL, Lin XJ, Qian XD, Chen XH, Niu YP. (2004). Effect of ginsenosides and panaxatriol extracted from ginseng on inhibition of proliferation, inducing apoptosis and cytotoxic drug sensitivity in leukemic cells. J Chin Med Res, 4(2):97-99.

Gottesman MM. (2002). Mechanisms of cancer drug resistance. Annu Rev Med, 53:615-617.

Gu W, Zhai XF, Zhang YN, Ling CQ. (2007). In vitro study of Ganli injection on reversing acquired Multi-drug resistance of human hepatocellular carcinoma. Chin Trad Herbal Drugs, 38(6):871-874.

Guo Y, Xie CS, Feng ZQ. (2002). The study of effects on accumulation and efflux of intracellular adrimycine with FFTLG for Multi-drug-resistant cell lines K562/ADR and K562/VCR in vitro. Chin J Mod Appl Pharm, 19(4):268-272.

Huang M, Jin J, Sun H, Liu GT. (2008). Reversal of P-glycoprotein-mediated Multi-drug resistance of cancer cells by five schizandrins isolated from the Chinese herb Fructus Schizandrae. Cancer Chemother Pharmacol, 62(6):1015-1026.

Kim SH, Yeo GS, Lim YS, et al. (1998). Suppression of Multi-drug resistance via inhibition of heat shock factor by quercetin in MDR cells. Exp Mol Med, 30(2):87-92.

Kim SW, Kwon HY, Chi DW, (2003). Reversal of P-glycoprotein-mediated Multi-drug resistance by ginsenoside Rg3. Biochem Pharmacol, 65(1):75-82.

Kioka N, Hosokawa N, Komano T, et al. (1992). Quercetin, a bioflavonoid, inhibits the increase of human Multi-drug resistance gene (MDR1) expression caused by arsenite. FEBS Lett, 301(3):307-309.

Kothan S, Dechsupa S, Leger G, et al. (2004). Spontaneous mitochondrial membrane potential change during apoptotic induction by quercetin in K562 and K562/adr cells. Can J Physiol Pharm, 82(12):1084-1090.

La Porta CAM. (2007). Drug resistance in melanoma: new perspectives. Curr Med Chem, 14(4):387-91.

Leonard GD, Fojo T, Bates SE. (2003). The role of ABC transporters in clinical practice. Oncologist, 8(5):411-424.

Li Y, Wang ZZ, Yu TF (2005). In vitro study on the reversal of Multi-drug resistance (MDR) in HL60/VCR cell line with ginsenoside – Rb1. J Radioimmunol, 18(5):362-365.

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

Limtrakul P, Chearwae W, Shukla S, Phisalphong C, Ambudkar SV. (2007). Modulation of function of three ABC drug transporters, P-glycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2) and Multi-drug resistance protein 1 (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol Cell Biochem, 296(1-2):85-95.

Limtrakul P, Khantamat O, Pintha K. (2005). Inhibition of P-glycoprotein function and expression by kaempferol and quercetin. J Chemother, 17(1):86-95.

Lin SY, Qin ZQ, Xu YF, Wu LC, Liu LM. (2002). Significance of combination shenmal injection, tamoxifen and nifedipine in treatment stage IV non-small-cell lung cancer. Chin J Cancer Prev Treat, 9(4):454-455.

Lin GF. (2007). Current situation in studying the reversing effect in Multi-drug-resistant in cancer cells by TCM. China Pharm, 16(10):63-64.

Liu LL, Liu YE, Fang GT. (2008). Reversal effect of Panax notoginseng saponins on Multi-drug resistance breast cancer cell MCF/ADM. Lishizhen Med Materia Med Res, 19(4):954-956.

Meszaros A, Balogh G. (2009). Multiple Drug Resistance. Gazelle Distribution, Lancaster, UK.

Park JD, Kim DS, Kwon HY, et al. (1996). Effects of ginseng saponin on modulation of Multi-drug resistance. Arch Pharm Res, 19(3):213-218.

Wang L. (2003). Reversing drug resistance of human erythroleukemia cell line K562/DOX on DOX by total saponings Panax ginseng. J Chongqing Med Univ, 28(4):424-427, 435.

Piwocka K, Bielak-Mijewska A, Sikora E. (2002). Curcumin induces caspase-3-independent apoptosis in human Multi-drug-resistant cells. Ann N Y Acad Sci, 973:250-254.

Qu Y, Liu SQ, Zhang XB, et al. (2006). Reversal of P-glycoprotein mediated Multi-drug resistance by traditional Chinese medicines. Nat Prod Res Dev, 18(6):932-936.

Sadava D, Ahn J, Zhan M, et al. (2002). Effects of four Chinese herbal extracts on drug-sensitive and Multi-drug-resistant small-cell lung carcinoma cells. Cancer Chemother Pharmacol, 49(4):261-266.

Shi XK, Zhang YJ, Zhao CJ. (1999). The reverse effects of ginseng saponin Rb1 on Multi-drug resistance of human leukemic cell line K562/HHT in vitro. Acta Acad Med Militaris Tertiae, 21(11):825-827.

Si YQ, Tien TD (2005). The Multi-drug-resistant reversing effect of total Panax notoginseng saponins studied in K562/VCR cell line. Chin J Tradit Med Sci Technol, 12(5):292-294.

Slaninová I, Brezinová L, Koubíková L, Slanina J. (2009). Dibenzocyclooctadiene lignans overcome drug resistance in lung cancer cells–study of structure-activity relationship. Toxicol In Vitro, 23(6):1047-1054.

Stewart A, Steiner J, Mellows G, et al. (2000). Phase I trial of XR9576 in healthy volunteers demonstrates modulation of Pglycoprotein in CD56+ lymphocytes after oral and intravenous administration. Clin Cancer Res, 6(11):4186-4191.

Sun GP, Wang H, Shen YX, et al. (2004). Study on effects of paeonol in inhibiting growth of K562 and inducing its apoptosis. Chin Pharmacol Bull, 20(5):550-552.

Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. (2006). Targeting Multi-drug resistance in cancer. Nat Rev Drug Discov, 5(3):219-234.

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.

Toppmeyer D, Seidman AD, Pollak M, et al. (2002). Safety and efficacy of the Multi-drug resistance inhibitor Incel (biricodar; VX-710) in combination with paclitaxel for advanced breast cancer refractory to paclitaxel. Clin Cancer Res, 8(3):670-678.

Tsuruo T, Iida H, Tsukagoshi S, Sakurai Y. (1981). Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res, 41(5):1967-1972.

Twentyman PR. (1992). Cyclosporins as drug resistance modifiers. Biochem Pharmacol, 43(1):109-117.

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.

Wan CK, Zhu GY, Shen XL, et al. (2006). Gomisin A alters substrate interaction and reverses P-glycoprotein-mediated Multi-drug resistance in HepG2-DR cells. Biochem Pharmacol, 72(7):824-837.

Wang Y, Chao PL, Hsiu SL, Wen K, Hou Y. (2004). Lethal quercetin-digoxin interaction in pigs. Life Sci, 74(10):1191-1197.

Wang J, Xia Y, Wang H, Hou Z. (2007). Chinese herbs of Shenghe powder reverse Multi-drug resistance of gastric carcinoma SGC-7901. Integr Cancer Ther, 6(4):400-404

Watson JV. (1991). Introduction to Flow Cytometry Cambridge. Cambridge University Press, Cambridge, UK.

Xu L, Wang MY, Xu DQ, Zhou CX. (2005). Experimental study on modified 'Sanwubai Powder' in affecting multi-drug resistance gene expression of tumor. Shanghai J Tradit Chin Med, 39(8):59-60.

Yang L, Wei DD, Chen Z, et al. (2011). Reversal effects of traditional Chinese herbs on Multi-drug resistance in cancer cells. Natural Product Research (Formerly Natural Product Letters), 25(19):1885-1889. doi: 10.1080/14786419.2010.541395

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.

Yu LF, Wu YL, Zhang YP. (2001). Reversal of drug resistance in the vincristine-resistant human gastric cancer cell lines MKN28/VCR by emulsion of seminal oil of Brucea Javanica. World Chin J Digestol, 9(4):376-378.

Quercetin

Cancer: Leukemia, prostate

Action: MDR, apoptosis-inducing

Quercetin is a plant-derived flavonol found in many fruits, vegetables, leaves and grains. It is also found in red wine.

MDR/ Apoptotic-inducing

Natural products from plants such as flavonoids are potential drugs to overcome multi-drug resistance (MDR) in cancer treatments. Quercetin exhibits cytotoxicity against erythroleukemic cells: IC50 are 11.0 +/- 2.0 micromol/L and 5.0 +/- 0.4 micromol/L for K562 and K562/adr, respectively. Quercetin induces cell death via apoptosis in both K562 and K562/adr cells and does not inhibit Pgp-mediated efflux of 99mTc-MIBI. Quercetin (10 micromol/L, 3 h) and etoposide (100 micromol/L, 24 hours) induce similar levels of apoptosis in K562 and K562/adr cells.

Quercetin induces an increase followed by a decrease in inner mitochondrial membrane potential   |DeltaPsim| value depending on its concentration. A decrease in the |DeltaPsim| value is associated with an increase in the percentage of early apoptotic cells.

It is clearly shown that quercetin results in a spontaneous DeltaPsim change during apoptotic induction. Therefore, quercetin is potentially an apoptotic-inducing agent, which reacts at the mitochondrial level (Kothan et al., 2004).

MDR

Expression of the MDR1 gene, which encodes P-glycoprotein, is increased under some stress conditions. It has been reported that quercetin, a bioflavonoid, inhibits the expression of heat-shock proteins. The effects of quercetin have been identified on the MDR1 gene expression in the human hepatocarcinoma cells line, HepG2. The increase of P-glycoprotein synthesis and MDR1 mRNA accumulation caused by exposure to arsenite were inhibited by quercetin. Although many drugs that prevent the P-glycoprotein function have been reported, this is the first report to describe the inhibition of MDR1 expression by a reagent (Kioka et al., 1992).

Leukemia

Leukemia cells were treated with quercetin, after which apoptosis, Mcl-1 expression, and Bax activation and translocation were evaluated. Quercetin-induced apoptosis was accompanied by Mcl-1 down-regulation and Bax conformational change and mitochondrial translocation that triggered cytochrome c release. In vivo administration of quercetin attenuated tumor growth in U937 xenografts. The TUNEL-positive apoptotic cells in tumor sections increased in quercetin-treated mice as compared with controls.

These data suggest that quercetin may be useful for the treatment of leukemia by preferentially inducing apoptosis in leukemia versus normal hematopoietic cells through a process involving Mcl-1 down-regulation, which, in turn, potentiates Bax activation and mitochondrial translocation, culminating in apoptosis (Cheng et al., 2010).

Prostate Cancer

The anti-angiogenic activity of quercetin was probed using ex vivo, in vivo and in vitro models. Angiogenesis is a crucial step in the growth and metastasis of cancers, since it enables the growing tumor to receive oxygen and nutrients. Quercetin (20 mg/kg/d) significantly reduced the volume and the weight of solid tumors in prostate xenograft mouse model, indicating that quercetin inhibited tumorigenesis by targeting angiogenesis.

Furthermore, quercetin reduced the cell viability and induced apoptosis in prostate cancer cells, which were correlated with the down-regulation of AKT, mTOR and P70S6K expressions. Collectively, these results suggest that quercetin inhibits tumor growth and angiogenesis by targeting VEGF-R2 regulated AKT/mTOR/P70S6K signaling pathway, and could be used as a potential drug candidate for cancer therapy (Pratheeshkumar et al., 2012).

References

Cheng SP, Gao N, Zhang Z, et al. (2010). Quercetin Induces Tumor-Selective Apoptosis through Down-regulation of Mcl-1 and Activation of Bax. Clin Cancer Res, 16(23):5679-91. doi: 10.1158/1078-0432.CCR-10-1565


Kioka N, Hosokawa N, Komano T, Hirayoshi K, Nagate K, Ueda K. (1992). Quercetin, a bioflavonoid, inhibits the increase of human Multi-drug resistance gene (< i> MDR1</i>) expression caused by arsenite. FEBS Lett, 301(3):307-9.


Kothan S, Dechsupa S, Leger G, et al. (2004). Spontaneous mitochondrial membrane potential change during apoptotic induction by quercetin in K562 and K562/adr cells. Can J Physiol Pharmacol, 82(12):1084-90.


Pratheeshkumar P, Budhraja A, et al. (2012). Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR- 2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS One, 7(10):e47516. doi: 10.1371/journal.pone.0047516.

Mollugin

Cancer: Breast, ovarian

Action: Multi-drug resistance, anti-inflammatory, blocks neurotoxicity

Mollugin originally isolated from Rubia cordifolia (L.) is a pharmacological compound for its anti-inflammation, anti-cancer, and anti-viral activity. Mollugin-caused inhibition of phenacetin O-deethylation was concentration-dependent in hierarchical linear models (HLMs), but not time-dependent. In addition, the Lineweaver-Burk plot indicated a typical competitive inhibition. Inhibitory effects of mollugin on human recombinant cDNA-expressed CYP1A1 and 1A2 were comparable. Taken together, the results suggested that mollugin might cause herb-drug interaction through selective inhibition of CYP1A2 in humans receiving herbal medications, including R. cordifolia (Kim et al., 2013).

MDR, Anti-inflammatory

Mollugin treatment significantly inhibited MDR1 expression by blocking MDR1 transcription. P-glycoprotein (P-gp), an important efflux transporter, is encoded by the MDR1 class of genes and is a central element of the multi-drug resistance (MDR) phenomenon in cancer cells. The suppression of MDR1 promoter activity and protein expression was mediated through mollugin-induced activation of AMP-activated protein kinase (AMPK). Furthermore, mollugin inhibited MDR1 expression through the suppression of NF-κB and cAMP-response element binding protein (CREB) activation. These results suggest that mollugin treatment enhanced suppression of P-gp expression by inhibiting the NF-κB signaling pathway and COX-2 expression, as well as attenuating cAMP-response element (CRE) transcriptional activity through AMPK activation (Tran et al., 2013).

Breast Cancer; Ovarian Cancer

Mollugin exhibited potent inhibitory effects on cancer cell proliferation, especially in HER2-overexpressing SK-BR-3 human breast cancer cells and SK-OV-3 human ovarian cancer cells in a dose- and time-dependent manner without affecting immortalized normal mammary epithelial cell line MCF-10A. Mollugin treatment caused a dose-dependent inhibition of HER2 gene expression at the transcriptional level, potentially in part through suppression of NF-κB activation. The combination of mollugin with a MEK1/2 inhibitor may be required in order to achieve optimal efficacy in HER2-overexpressing cancers.

These findings suggest that mollugin is a novel modulator of the HER2 pathway in HER2-overexpressing cancer cells with a potential role in the treatment and prevention of human breast and ovarian cancer with HER2 overexpression (Do et al., 2013).

Blocks Neurotoxicity, Anti-inflammatory

Mollugin also has effects as a neuro-protective agent in glutamate-induced neurotoxicity in the mouse hippocampal HT22 cell line and as an anti-inflammatory agent in lipopolysaccharide-induced microglial activation in BV2 cells. Mollugin showed potent neuro-protective effects against glutamate-induced neuro-toxicity and reactive oxygen species generation in mouse hippocampal HT22 cells.

In addition, the anti-inflammatory effects of mollugin were demonstrated by the suppression of pro-inflammatory mediators, including pro-inflammatory enzymes (inducible nitric oxide synthase and cyclooxygenase-2) and cytokines (tumor necrosis factor-α and interleukin-6). Furthermore, mollugin also activated the p38 mitogen-activated protein kinase (MAPK) pathway both in HT22 and BV2 cells. These results suggest that mollugin may be a promising candidate for the treatment of neurodegenerative diseases related to neuroinflammation (Jeong et al., 2011).

References

Do MT, Hwang YP, Kim HG, et al. (2013). Mollugin inhibits proliferation and induces apoptosis by suppressing fatty acid synthase in HER2-overexpressing cancer cells. Journal of Cellular Physiology, 228(5):1087–1097. doi: 10.1002/jcp.24258.


Jeong GS, Lee DS, Kim DC, et al. (2011). Neuroprotective and anti-inflammatory effects of mollugin via up-regulation of heme oxygenase-1 in mouse hippocampal and microglial cells. Eur J Pharmacol, 654(3):226-34. doi: 10.1016/j.ejphar.2010.12.027.


Kim H, Choi HK, Jeong TC, et al. (2013). Selective inhibitory effects of mollugin on CYP1A2 in human liver microsomes. Food Chem Toxicol, 51:33-7. doi: 10.1016/j.fct.2012.09.013.


Tran TP, Kim HG, Choi JH, et al. (2013). Reversal of P-glycoprotein-mediated Multi-drug resistance is induced by mollugin in MCF-7/adriamycin cells. Phytomedicine. doi:10.1016/j.phymed.2013.01.014.

Emodin (See also Aloe-Emodin)

Cancer:
Breast, colon, liver, chemotherapy, myeloma, oral., pancreatic, hepatocellular carcinoma, lung, leukemia

Action: MDR-1, cell-cycle arrest

Emodin is an active natural anthraquinone derivative component of a traditional Chinese and Japanese medicine isolated from the root and rhizomes of Rheum palmatum L., Senna obtusifolia [(L.) H.S.Irwin & Barneby], Fallopia japonica [Houtt. (Ronse Decr.)], Kalimeris indica (L.) Sch.Bip., Ventilago madraspatana (Gaertn.), Rumex nepalensis (Spreng.), Fallopia multiflora [(Thunb.) Haraldson], Cassia occidentalis [(L.) Link], Senna siamea [(Lam.) Irwin et Barneby] and Acalypha australis (L.).

Aloe-emodin is an active natural anthraquinone derivative, and is found in the roots and rhizomes of numerous Chinese medicinal herbs (including Rheum palmatum L) and exhibits anti-cancer effects on many types of human cancer cell lines.

Administration of rhubarb (Emodin) can effectively reverse severe acute pancreatitis (SAP) by regulating the levels of IL-15 and IL-18 (Yu & Yang, 2013).

Pancreatic Cancer

Emodin is a tyrosine kinase inhibitor that has an inhibitory effect on mammalian cell-cycle modulation in specific oncogene-overexpressing cells. Recently, there has been great progress in the preclinical study of the anti-cancer mechanisms of emodin. A recent study revealed that emodin has therapeutic effects on pancreatic cancer through various anti-tumor mechanisms. Notably, the therapeutic efficacy of emodin in combination with chemotherapy was found to be higher than the comparable single chemotherapeutic regime, and the combination therapy also exhibited fewer side-effects (Wei et al., 2013).

Hepatocellular Carcinoma, Pancreatic, Breast, Colorectal and Lung Cancers, and Leukemia

Emodin is found as an active ingredient in different Chinese herbs including Rheum palmatum and Polygonam multiflorum, and has diuretic, vasorelaxant, anti-bacterial., anti-viral., anti-ulcerogenic, anti-inflammatory, and anti-cancer effects. The anti-inflammatory effects of emodin have been exhibited in various in vitro as well as in vivo models of inflammation including pancreatitis, arthritis, asthma, atherosclerosis and glomerulonephritis. As an anti-cancer agent, emodin has been shown to suppress the growth of various tumor cell lines including hepatocellular carcinoma, pancreatic, breast, colorectal., leukemia, and lung cancers. Emodin is a pleiotropic molecule capable of interacting with several major molecular targets including NF-κB, casein kinase II, HER2/neu, HIF-1α, AKT/mTOR, STAT3, CXCR4, topoisomerase II, p53, p21, and androgen receptors which are involved in inflammation and cancer (Shrimali et al., 2013).

Hepatocellular Carcinoma

It has been found that emodin induces apoptotic responses in the human hepatocellular carcinoma cell lines (HCC) Mahlavu, PLC/PRF/5 and HepG2. The addition of emodin to these three cell lines led to inhibition of growth in a time-and dose-dependent manner. Emodin generated reactive oxygen species (ROS) in these cells which brought about a reduction of the intracellular mitochondrial transmembrane potential (ΔΨ m), followed by the activation of caspase–9 and caspase–3, leading to DNA fragmentation and apoptosis.

Preincubation of hepatoma cell lines with the hydrogen peroxide-scavenging enzyme, catalase (CAT) and cyclosporin A (CsA), partially inhibited apoptosis. These results demonstrate that enhancement of generation of ROS, DeltaPsim disruption and caspase activation may be involved in the apoptotic pathway induced by emodin (Jing et al., 2002).

Colon Cancer

In in vitro study, emodin induced cell morphological changes, decreased the percentage of viability, induced G2/M phase arrest and increased ROS and Ca(2+) productions as well as loss of mitochondrial membrane potential (ΔΨ(m)) in LS1034 cells. Emodin-triggered apoptosis was also confirmed by DAPI staining and these effects are concentration-dependent.

In in vivo study, emodin effectively suppressed tumor growth in tumor nude mice xenografts bearing LS1034. Overall, the potent in vitro and in vivo anti-tumor activities of emodin suggest that it might be developed for treatment of colon cancer in the future (Ma et al., 2012).

Myeloid Leukemia

It has been shown that emodin significantly induces cytotoxicity in the human myeloma cells through the elimination of myeloid cell leukemia 1 (Mcl-1). Emodin inhibited interleukin-6–induced activation of Janus-activated kinase 2 (JAK2) and phosphorylation of signal transducer and activator of transcription 3 (STAT3), followed by the decreased expression of Mcl-1. Activation of caspase-3 and caspase-9 was triggered by emodin, but the expression of other anti-apoptotic Bcl-2 family members, except Mcl-1, did not change in the presence of emodin. To clarify the importance of Mcl-1 in emodin-induced apoptosis, the Mcl-1 expression vector was introduced into the human myeloma cells by electroporation. Induction of apoptosis by emodin was almost abrogated in Mcl-1–overexpressing myeloma cells as the same level as in parental cells, which were not treated with emodin. Emodin therefore inhibits interleukin-6–induced JAK2/STAT3 pathway selectively and induces apoptosis in myeloma cells via down-regulation of Mcl-1, which is a good target for treating myeloma. Taken together, these results show emodin as a new potent anti-cancer agent for the treatment of multiple myeloma patients (Muto et al., 2007).

Breast Cancer; Block HER-2

The mechanism by which emodin prevents breast cancer is unknown; however the product of the HER-2/neu proto-oncogene, HER2 has been proposed to be involved. The product of the HER-2/neu proto-oncogene, HER2, is the second member of the human epidermal growth factor receptor (HER) family of tyrosine kinase receptors and has been suggested to be a ligand orphan receptor. Amplification of the HER2 gene and overexpression of the HER2 protein induces cell transformation and has been demonstrated in 10% to 40% of human breast cancer. HER2 overexpression has been suggested to associate with tumor aggressiveness, prognosis and responsiveness to hormonal and cytotoxic agents in breast cancer patients. These findings indicate that HER2 is an appropriate target for tumor-specific therapies.

A number of approaches have been investigated: (1) a humanized monoclonal antibody against HER2, rhuMAbHER2 (trastuzumab), which is already approved for clinical use in the treatment of patients with metastatic breast cancer; (2) tyrosine kinase inhibitors, such as emodin, which block HER2 phosphorylation and its intracellullar signaling; (3) active immunotherapy, such as vaccination; and (4) heat shock protein (Hsp) 90-associated signal inhibitors, such as radicicol derivatives, which induce degradation of tyrosine kinase receptors, such as HER2 (Kurebayashi, 2001).

MDR

The effects of emodin on the nucleoside transport and multi-drug resistance in cancer cells has also been investigated. Nucleoside transport inhibition was determined by thymidine incorporation assay. The cytotoxicity to cancer cells was determined by MTT assay. The pump efflux activity and the expression of P glycoprotein were examined by flow cytometric assay. Emodin was active in the inhibition of nucleoside transport, with an IC 50 value of 9 9 µmol·L -1. Emodin markedly enhanced the cytotoxicity of 5 FU, MMC and MTX against human hepatoma BEL 7402 cells and partly reversed the multi-drug resistance in human breast cancer MCF 7/Adr cells.

Emodin inhibited P-gp pump efflux activity and reduced the expression of P gp in MCF 7/Adr cells. These findings provide a biological basis for the application of emodin as a biochemical modulator to potentiate the effects of anti-tumor drugs and reverse the multi-drug resistance in cancer cells (Jiang et al., 2009).

Cell-cycle Arrest

Large quantities of emodin were isolated from the roots of Rheum emodi and a library of novel emodin derivatives 2–15 were prepared to evaluate their anti-proliferative activities against HepG2, MDA-MB-231 and NIH/3T3 cells lines. The derivatives 3 and 12 strongly inhibited the proliferation of HepG2 and MDA-MB-231 cancer cell line with an IC50 of 5.6, 13.03 and 10.44, 5.027, respectively, which is comparable to marketed drug epirubicin (III). The compounds 3 and 12 were also capable of inducing cell-cycle arrest and caspase dependent apoptosis in HepG2 cell lines and exhibit DNA intercalating activity. These emodin derivatives hold promise for developing safer alternatives to the marketed epirubicin (Narender et al., 2013).

Cell-cycle Arrest; MDR1 & AZT

3'-azido-3'-deoxythymidine (AZT) and emodin altered the cell-cycle distribution and led to an accumulation of cells in S phase. Meanwhile, the expression of MDR1 mRNA/p-gp protein was markedly decreased. These results show a synergistic growth-inhibitory effect of AZT and emodin in K562/ADM cells, which is achieved through S phase arrest. MDR1 might ultimately be responsible for these phenomena (Chen et al., 2013).

References

Chen P, Liu Y, Sun Y, et al. (2013). AZT and emodin exhibit synergistic growth-inhibitory effects on K562/ADM cells by inducing S phase cell-cycle arrest and suppressing MDR1 mRNA/p-gp protein expression. Pharm Biol.


Garg AK, Buchholz TA, Aggarwal BB. (2005). Chemo-sensitization and Radiosensitization of Tumors by Plant Polyphenols. Antioxid Redox Signal., 7(11-12):1630-47.


Jiang XF & Zhen YS. (1999). Reversal of Multi-drug resistance by emodin in cancer cells. Acta Pharmaceutica Sinica, 1999-03.


Jing X, Ueki N, Cheng J, Imanishi H, Hada T. (2002). Induction of apoptosis in hepatocellular carcinoma cell lines by emodin. Cancer Science, 93(8):874–882.


Kurebayashi J. (2001). Biological and clinical significance of HER2 overexpression in breast cancer. Breast Cancer, 8(1):45-51


Ma YS, Weng SW, Lin MW, et al. (2012). Anti-tumor effects of emodin on LS1034 human colon cancer cells in vitro and in vivo: Roles of apoptotic cell death and LS1034 tumor xenografts model. Food Chem Toxicol, 50(5): 1271–1278. doi: 10.1016/j.fct.2012.01.033.


Muto A, Hori M, Sasaki Y, et al. (2007). Emodin has a cytotoxic activity against human multiple myeloma as a Janus-activated kinase 2 inhibitor. Mol Cancer Ther. doi: 10.1158/1535-7163.MCT-06-0605.


Narender T, Sukanya P, Sharma K, et al. (2013). Preparation of novel anti-proliferative emodin derivatives and studies on their cell-cycle arrest, caspase dependent apoptosis and DNA binding interaction. Phytomedicine, 20(10):890-896.


Shrimali D, Shanmugam MK, Kumar AP, et al. (2013). Targeted abrogation of diverse signal transduction cascades by emodin for the treatment of inflammatory disorders and cancer. Cancer Lett:S0304-3835(13)00598-3. doi: 10.1016/j.canlet.2013.08.023.


Wei WT, Lin SZ, Liu DL, Wang ZH. (2013). The distinct mechanisms of the anti-tumor activity of emodin in different types of cancer (Review). Oncol Rep. doi: 10.3892/or.2013.2741.


Yu XW, Yang RZ. (2013). Effects of crude rhubarb on serum IL-15 and IL-18 levels in patients with severe acute pancreatitis. An Hui Yi Xue, 34(3): 285-287.

Cnidiadin

Cancer: none noted

Action: MDR

Cnidiadin is an iaolate of Cnidium monnieri (L.) Cuss. (Umbelliferae)

Preliminary studies show some of the compounds in cnidium may have anti-histamine, anti-itch, anti-fungal., and anti-bacterial effects, along with having an influence on the pituitary-adrenocortex axis. Additional studies on rodents indicate that cnidium may improve bone strength (coumarins from fruits of cnidium inhibit formation and differentiation of multinucleated osteoclasts of rats).

In rabbits, a compound found in cnidium called osthol or osthole was found to help relax the corpus cavernosa of the penis, which would potentially help with blood flow and erections. Cnidium may also have anti-cancer properties.

Active Compounds:

Osthole, Imperatorin, Bornyl isovalerate, Cnidimine, Xanthotoxin, Xanthotoxol, Isopimpinelline, Bergapten, Cnidiadin, Cniforin A, Dihydrooroselol, Columbianadin, berapten, cnidiadin, sopimpinellin, imperation, thymine, hypoxanthine (http://www.mdidea.com/products/herbextract/cnidium/data01.html)

MDR

Overexpression of P-glycoprotein (Pgp) encoded by the MDR1 gene is one of the major obstacles to successful cancer chemotherapy. Cnidiadin, a furanocoumarin present in traditional Chinese medications and in a spice commonly used in Greek food, was hence evaluated for its ability to inhibit Pgp transport activity and its potential to reverse MDR1 multi-drug resistance. The dose-dependent increase in [3H]-VBL uptake (IC50 26.5 microM) induced by cnidiadin in the dose range 1–100 microM correlated with inhibition of Pgp photolabeling. At 10 microM cnidiadin inhibited photolabeling by 59% and sensitized both MDCK-MDR1 and KB/VCR cells to vinca alkaloids.

Cnidiadin is a cytotoxic agent capable in vitro of competitively inhibiting the binding and efflux of drug by Pgp and of enhancing the cell toxicity of vinca alkaloids in two cell lines (MDCK-MDR1 and mutant human carcinoma KB/VCR) overexpressing Pgp. This suggests that diet or traditional preparation containing cnidiadin may contribute to the reversal of MDR1 multi-drug resistance and may affect the bioavailability of Pgp substrates orally administered. However, due to its cell toxicity, clinical interest in cnidiadin as a chemo-sensitizer appears to be limited (Barthomeuf et al., 2005).

Reference

Barthomeuf C, Grassi J, Demeule M, et al. (2005). Inhibition of P-glycoprotein transport function and reversion of MDR1 Multi-drug resistance by cnidiadin. Cancer Chemother Pharmacol, 56(2):173-81.

Chelerythrine, Chelidonine and Sanguinarine

Cancer:
Leukemia, oral squamous cell carcinoma, melanoma

Action: Cytotoxic, MDR, apoptosis-triggering, inhibits proliferation

Sanguinarine, chelerythrine and chelidonine are isoquinoline alkaloids derived from the greater celandine. They possess a broad spectrum of pharmacological activities. It has been shown that their anti-tumor activity is mediated via different mechanisms, which can be promising targets for anti-cancer therapy. This study focuses on the differential effects of these alkaloids upon cell viability, DNA damage, and nucleus integrity in mouse primary spleen and lymphocytic leukemic cells, L1210.

Data suggests that cytotoxic and DNA-damaging effects of chelerythrine and sanguinarine are more selective against mouse leukemic cells and primary mouse spleen cells, whereas chelidonine blocks proliferation of L1210 cells. The action of chelidonine on normal and tumor cells requires further investigation (Kaminsky, Lin, Filyak, & Stoika, 2008).

MDR

Cancer cells often develop multi-drug resistance (MDR) which is a multidimensional problem involving several mechanisms and targets. This study demonstrates that chelidonine, an alkaloid extract from Chelidonium majus, which contains protoberberine and benzo[c]phenanthridine alkaloids, has the ability to overcome MDR of different cancer cell lines through interaction with ABC-transporters, CYP3A4 and GST, by induction of apoptosis, and cytotoxic effects.

Chelidonine and the alkaloid extract inhibited P-gp/MDR1 activity in a concentration-dependent manner in Caco-2 and CEM/ADR5000 and reversed their doxorubicin resistance. In addition, chelidonine and the alkaloid extract inhibited the activity of the drug, modifying enzymes CYP3A4 and GST in a dose-dependent manner. The expression analysis identified a common set of regulated genes related to apoptosis, cell-cycle, and drug metabolism.

Results suggest that chelidonine is a promising compound for overcoming MDR and enhancing cytotoxicity of chemotherapeutics, especially against leukemia cells. Its efficacy needs to be confirmed in animal models (El-Readi, Eid, Ashour, Tahrani & Wink, 2013).

Induces Apoptosis, Leukemia

Sanguinarine, chelerythrine and chelidonine possess prominent apoptotic effects towards cancer cells. This study found that sanguinarine and chelerythrine induced apoptosis in human CEM T-leukemia cells, accompanied by an early increase in cytosolic cytochrome C that precedes caspases-8, -9 and -3 processing. Effects of sanguinarine and chelerythrine on mitochondria were confirmed by clear changes in morphology (3h), howerver chelidonine did not affect mitochondrial integrity. Sanguinarine and chelerythrine also caused marked DNA damage in cells after 1h, but a more significant increase in impaired cells occurred after 6h. Chelidonine induced intensive DNA damage in 15–20% cells after 24h.

Results demonstrated that rapid cytochrome C release in CEM T-leukemia cells exposed to sanguinarine or chelerythrine was not accompanied by changes in Bax, Bcl-2 and Bcl-X((L/S)) proteins in the mitochondrial fraction, and preceded activation of the initiator caspase-8 (Kaminskyy, Kulachkovskyy, & Stoika, 2008).

Induces Apoptosis

Chelerythrine, formerly identified as a protein kinase C inhibitor, has also been shown to inhibit the anti-apoptotic Bcl-2 family proteins. Chelerythrine initiates the rapid mitochondrial apoptotic death of H9c2 cardiomyoblastoma cells in a manner that is likely independent of the generation of ROS from mitochondria (Funakoshi et al., 2011).

Oral Cancer, Inhibits cell proliferation

The effects of benzo[c] phenanthridine alkaloids (QBA), known mainly as sanguinarine and chelerythrine, on the inhibition of some kinds of cancer cell proliferation have been established. Sanguinarine is a potential inhibitor of tumorigenesis which suggests that it may be valuable in the development of new anti-cancer drugs for the treatment of oral squamous cell carcinoma (OSCC) (Tsukamoto et al., 2011).

Apoptotic Effects; Melanoma

Mixtures of isoquinoline alkaloids containing protopine, chelidonine, sanguinarine, allocryptopine, and stylopine were applied to murine fibroblast NIH/3T3, mouse melanoma B16F10, and human breast cancer MCF7 cell cultures for 20 and 40 min, and the content of alkaloids in the cell media was measured by capillary electrophoresis (CE). CE separation of isoquinoline alkaloids was performed in 30 mM phosphate buffer (pH 2.5). As these alkaloids have native fluorescence, they were directly detected using the commercially available UV light-emitting diode without fluorescent derivatization. The results showed a differential ability of celandine alkaloids to penetrate into the normal and cancer cell interior, which was inversely proportional to their cytotoxic activity.

While the most effective transport of celandine alkaloids from the cell medium to the cell interior was observed for normal murine fibroblast NIH/3T3 cells (about 55% of total content), cytotoxicity tests demonstrated selective and profound apoptotic effects of a five-alkaloid combination in the mouse melanoma B16F10 cell line (Kulp & Bragina, 2013).

Leukemia

The methanol extract isolated from the greater celandine Chelidonium majus L. (CME) has a strong anti-oxidant potential and exerted the anti-proliferative activity via apoptosis on leukemia cells. CME, due to the presence of the isoquinoline alkaloids and the flavonoid components may play an important role in both cancer chemoprevention through its anti-oxidant activity and modern cancer chemotherapy as a cytotoxic and apoptosis-inducing agent (Nadova et al., 2008).

Apoptosis-inducing Activity

Apoptogenic and DNA-damaging effects of chelidonine (CHE) and sanguinarine (SAN), two structurally related benzophenanthridine alkaloids isolated from Chelidonium majus L. (Papaveraceae), were compared. Both alkaloids induced apoptosis in human acute T-lymphoblastic leukaemia MT-4 cells. Apoptosis induction by CHE and SAN in these cells was accompanied by caspase-9 and -3 activation and an increase in the pro-apoptotic Bax protein. An elevation in the percentage of MT-4 cells possessing caspase-3 in active form after their treatment with CHE or SAN was in parallel to a corresponding increase in the fraction of apoptotic cells. CHE, in contrast to SAN, does not interact directly with DNA.

This fact is in line with DNA-damaging effects of the alkaloids detected in the COMET assay. Nevertheless, apoptosis-inducing activity of CHE even slightly exceeded that of SAN (Philchenkov et al., 2008).

Chelidonium majus L. alkaloids chelidonine, sanguinarine, chelerythrine, protopine and allocryptopine were identified as major components of Ukrain. Apart from sanguinarine and chelerythrine, chelidonine turned out to be a potent inducer of apoptosis, triggering cell death at concentrations of 0.001 mM, while protopine and allocryptopine were less effective. Similar to Ukrain, apoptosis signaling of chelidonine involved Bcl-2 controlled mitochondrial alterations and caspase-activation (Habermehl et al., 2006).

References

El-Readi MZ, Eid S, Ashour ML, Tahrani A, & Wink M. (2013). Modulation of Multi-drug resistance in cancer cells by chelidonine and Chelidonium majus alkaloids. Phytomedicine, 20(3-4), 282-94. doi: 10.1016/j.phymed.2012.11.005.


Funakoshi T, Aki T, Nakayama H, et al. (2011). Reactive oxygen species-independent rapid initiation of mitochondrial apoptotic pathway by chelerythrine. Toxicol In Vitro, 25(8):1581-7. doi: 10.1016/j.tiv.2011.05.028.


Habermehl D, Kammerer B, Handrick R, et al. (2006). Pro-apoptotic activity of Ukrain is based on Chelidonium majus L. alkaloids and mediated via a mitochondrial death pathway. BMC Cancer, 6:14.


Kaminskyy V, Lin KW, Filyak Y, & Stoika R. (2008). Differential effect of sanguinarine, chelerythrine and chelidonine on DNA damage and cell viability in primary mouse spleen cells and mouse leukemic cells. Cell Biology International., 32(2), 271-277.


Kaminskyy V, Kulachkovskyy O,Stoika R. (2008). A decisive role of mitochondria in defining rate and intensity of apoptosis induction by different alkaloids. Toxicology Letters, 177(3), 168-81. doi: 10.1016/j.toxlet.2008.01.009.


Kulp M, Bragina O. (2013). Capillary electrophoretic study of the synergistic biological effects of alkaloids from Chelidonium majus L. in normal and cancer cells. Analytical and Bioanalytical Chemistry, 405(10), 3391-7. doi: 10.1007/s00216-013-6755-y.


Nadova S, Miadokova E, Alfoldiova L, et al. (2008). Potential anti-oxidant activity, cytotoxic and apoptosis-inducing effects of Chelidonium majus L. extract on leukemia cells. Neuro Endocrinol Lett, 29(5):649-52.


Philchenkov A., Kaminskyy V., Zavelevich M., Stoika R. (2008). Apoptogenic activity of two benzophenanthridine alkaloids from Chelidonium majus L. does not correlate with their DNA-damaging effects. Toxicology In Vitro, 22(2), 287-95.


Tsukamoto H, Kondo S, Mukudai Y, et al., (2011). Evaluation of anti-cancer activities of benzo[c]phenanthridine alkaloid sanguinarine in oral squamous cell carcinoma cell line. Anti-cancer Res, 31(9):2841-6.


Zhe C, Li-Juan W, Ming Hui W, et al. (2011). Mechanism governing reversal of Multi-drug resistance in human breast carcinoma cells by chelerythrine. Zhongguo Yi Xue Ke Xue Yuan Xue Bao, 33(1):45-50. doi: 10.3881/j.issn.1000-503X.2011.01.010.

Biochanin A

Cancer: Breast

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

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

MDR; Breast Cancer

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

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

Chemo-preventive

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

Anti-inflammatory

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

References

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


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


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

Shikonin

Cancer: Sarcoma-180, lung, melanoma, leukemia

Action: Anti-inflammatory, inhibits angiogenesis, MDR

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

Melanoma

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

Anti-inflammatory

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

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

Leukemia, MDR

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

Lung Cancer

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

MDR

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

References

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


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


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


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


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


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


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

Coniferyl Ferulate

Cancer: Lung

Action: Multi-drug resistance reversal

MDR

Glutathione S-transferase (GST) is a key enzyme in the development of multi-drug resistance (MDR) in tumors. Inhibition of the expression, or activity, of GST has emerged as a promising therapeutic strategy for the reversal of MDR.

Coniferyl ferulate (CF), isolated from the root of Radix Angelica sinensis (RAS), showed strong inhibition of human placental GST. Using the high-throughput screening model, CF's 50% inhibition concentration (IC50) was 0.3   µM, which was greater than the established GSTP1-1 inhibitor, ethacrynic acid (EA).

Moreover, CF showed strong apoptotic activity and could markedly decrease the overexpression of P-gp. The results demonstrated that CF could inhibit GST activity in a concentration-dependent manner and showed a potential MDR reversal effect for anti-tumor adjuvant therapy.

CF demonstrates strong GST inhibitory activity and may serve as a prospective MDR reversal agent in the treatment of cancer. In addition, CF could potentially be used as a promising chemo-sensitizer capable of indirectly regulating P-gp expression via modulation of GST activity, and with fewer adverse effects. Further investigation of CF in anti-tumor adjuvant therapy is warranted (Chen et al., 2013).

Ferulic acid (FA) is widely considered as a biologically active component in Angelica sinensis, and used as one of the marker compounds for the quality control of Angelica sinensis. However, in A. sinensis, FA mainly exists as its ester, coniferyl ferulate (CF). The potency of CF is much higher than that of FA, and the IC50 values for AA, ADP and THR were 7.1 ± 0.3, 276.4 ± 53.4 and 77.5 ± 23.1 µg/ml, respectively (Yu et al., 2009).

References

Chen C, Wu C, Lu X, Yan, Z, Gao, H, Li, S (2013). Coniferyl ferulate, a strong inhibitor of glutathione S-transferase isolated from radix Angelicae sinensis, reverses Multi-drug resistance and downregulates P-glycoprotein. Evidence-Based Complementary and Alternative Medicine, 2013(2013), ID639083. http://dx.doi.org/10.1155/2013/639083.


Yu Y, Lin BQ, Yu L, et al. (2009). Inhibitory Effects of Two Ferulates from Angelica Sinensis on Platelet Aggregation and Oxytocin-induced Uterine Contraction. The Open Bioactive Compounds Journal., 2009, 2, 43-46.

Costunolide and Dehydrocostus Lactone

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

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

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

Breast cancer; Anti-metastatic

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

Breast Cancer

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

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

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

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

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

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

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

ER- Breast Cancer

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

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

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

Bladder Cancer

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

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

Sarcomas; MDR

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

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

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

Leukemia, Lung Cancer

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

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

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

Prostate Cancer

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

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

Gastric Cancer, Prostate Cancer

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

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

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

Anti-metastatic

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

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

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

Ovarian Cancer, MDR

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

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

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

Anti-inflammatory, Anti-oxidant, Mediates Apoptosis

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

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

References

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

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


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


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


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


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


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


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


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


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


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


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


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

Evodiamine

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

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

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

Anti-cancer, MDR

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

Lung Cancer, Cell-cycle Arrest

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

Cell-cycle Arrest

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

Increases Intracellular ROS, Apoptosis

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

Inhibits Metastasis

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

Inhibits NF-κB

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

Breast Cancer

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

Breast Cancer; ER+, ER-

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

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

Gastric Cancer

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

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

Pancreatic Cancer

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

References

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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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

Icariin

Cancer: Breast, gastric, Leydig cell, gall bladder

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

Estrogen Agonist

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

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

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

Gastric Cancer

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

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

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

Gallbladder Cancer; Gemcitabine

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

Restores T cell function

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

Leydig Cell (Testicle)

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

Induces Apoptosis

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

References

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


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


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


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


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


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

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.

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.

Formononetin

Cancer: Prostate, colorectal., breast, cervical

Action: Cell-cycle arrest, MDR, growth-inhibitory

Estrogenic or Anti-estrogenic

Formononetin is one of the main active components of red clover plants, and considered as a phytoestrogen. Its pharmacological effects in vivo may be either estrogenic or anti-estrogenic, mainly depending upon the estrogen levels (Chen & Sun., 2012).

Cell-cycle Arrest, Prostate Cancer

Formononetin has been demonstrated to cause cell-cycle arrest at the G0/G1 phase by inactivating insulin-like growth factor 1(IGF1)/IGF1R-phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway in MCF-7 cells. The molecular mechanisms involved in the effect of formononetin on prostate cancer cells were hence investigated. These results suggest that higher concentrations of formononetin inhibit the proliferation of prostate cancer cells (LNCaP and PC-3), while the most striking effect was observed in LNCaP cells.

From these results, it was concluded that the induced apoptosis effect of formononetin on human prostate cancer cells was related to ERK1/2 MAPK-Bax pathway. Considering that red clover plants were widely used clinically, these results provided the foundation for future development of different concentrations of formononetin for treatment of prostate cancer (Ye et al., 2012).

Colon Cancer

Formononetin is a novel herbal isoflavonoid isolated from Astragalus membranaceus, a medicinal plant that possesses anti-tumorigenic properties. It has been demonstrated that formononetin initiates growth-inhibitory and pro-apoptotic activities in human colon cancer cells. The potential of formononetin in controlling angiogenesis and tumor cell invasiveness has further been examined in human colon cancer cells and tumor xenografts. The results showed that formononetin downregulated the expression of the key pro-angiogenic factors, including vascular endothelial growth factor (VEGF) and matrix metalloproteinases. The tumor size and the number of proliferating cells were reduced in the tumor tissues obtained from the formononetin-treated group.

The serum VEGF level was also reduced in the drug-treated animals when compared to the controls. These findings suggest that formononetin inhibits angiogenesis and tumor cell invasion, and thus support its use in the treatment of advanced and metastatic colon cancers (Auyeung et al., 2012).

Cervical Cancer

Formononetin may potentiate the cytotoxicity of epirubicin in HeLa cells through the ROS-mediated MRP inhibition and concurrent activation of the mitochondrial and death receptor pathways of apoptosis. Hence, the circumvention of pump and non-pump resistance using formononetin and epirubicin may pave the way for a powerful chemotherapeutic regimen for treating human cervical cancer (Lo et al., 2013).

Breast Cancer

Recent studies by Chen & Sun (2012) suggest that formononetin inactivated IGF1/IGF1R-PI3K/Akt pathways and decreased cyclin D1 mRNA and protein expression in human breast cancer cells in vitro and in vivo. In their present study, they further investigated the molecular mechanisms involved in the induced apoptosis effect of formononetin on breast cancer cells and formononetin inhibited the proliferation of ER-positive MCF-7 cells and T47D cells. The induced apoptosis effect of formononetin on human breast cancer cells was related to Ras-p38MAPK pathway.

Formononetin causes cell-cycle arrest at the G0/G1 phase by inactivating IGF1/IGF1R-PI3K/Akt pathways and decreasing cyclin D1 mRNA and protein expression, indicating the use of formononetin in the prevention of breast cancer carcinogenesis (Chen et al., 2011).

References

Auyeung KK, Law PC, Ko JK. (2012). Novel anti-angiogenic effects of formononetin in human colon cancer cells and tumor xenograft. Oncol Rep, 28(6):2188-94. doi: 10.3892/or.2012.2056.


Chen J, Zeng J, Xin M, Huang W, Chen X. (2011). Formononetin induces cell-cycle arrest of human breast cancer cells via IGF1/PI3K/Akt pathways in vitro and in vivo. Horm Metab Res, 43(10):681-6. doi: 10.1055/s-0031-1286306.


Chen J, Sun L. (2012). Formononetin-induced apoptosis by activation of Ras/p38 mitogen-activated protein kinase in estrogen receptor-positive human breast cancer cells. Horm Metab Res, 44(13):943-8. doi: 10.1055/s-0032-1321818.


Lo YL, Wang W. (2013). Formononetin potentiates epirubicin-induced apoptosis via ROS production in HeLa cells in vitro. Chem Biol Interact, 205(3):188-97. doi: 10.1016/j.cbi.2013.07.003.


Ye Y, Hou R, Chen J, et al. (2012). Formononetin-induced apoptosis of human prostate cancer cells through ERK1/2 mitogen-activated protein kinase inactivation. Horm Metab Res, 44(4):263-7. doi: 10.1055/s-0032-1301922.

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.