Category Archives: Anti-angiogenesis

Siphonaxanthin

Cancer: none noted

Action: Anti-angiogenesis

Siphonaxanthin is the active anti-angiogenic constituent of the green algae (Codium fragile [(Suringar) Hariot]).

Siphonaxanthin significantly suppressed HUVEC proliferation (p<0.05) at the concentration of 2.5µM (50% as compared with control) and above, while the effect on chemotaxis was not significant. Siphonaxanthin exhibited strong inhibitory effect on HUVEC tube formation. It suppressed the formation of tube length by 44% at the concentration of 10µM, while no tube formation was observed at 25µM, suggesting that it could be due to the suppression of angiogenic mediators.

The ex vivo angiogenesis assay exhibited reduced microvessel outgrowth in a dose-dependent manner and the reduction was significant at more than 2.5µM. These results imply a new insight into the novel function of siphonaxanthin in preventing angiogenesis related diseases (Ganesan et al., 2010).

Reference

Ganesan P, Matsubara K, Ohkubo T, et al. (2010). Anti-angiogenic effect of siphonaxanthin from green alga, Codium fragile. Phytomedicine, 17(14):1140-1144.

RG3 (See also Ginsenosides)

Cancer: Glioblastoma, prostate, breast, colon

Action: Anti-angiogenesis, MDR, enhances chemotherapy, MDR, enhanced paclitaxel absorption, anti-metastatic

RG3 is a ginsenoside isolated from red ginseng (Panax ginseng (L.)), after being peeled, heated, and dried.

Angiosuppressive Activity

Aberrant angiogenesis is an essential step for the progression of solid tumors. Thus anti-angiogenic therapy is one of the most promising approaches to control tumor growth.

Rg3 was found to inhibit the proliferation of human umbilical vein endothelial cells (HUVEC) with an IC50 of 10 nM in Trypan blue exclusion assay.

Rg3 (1-10(3) nM) also dose-dependently suppressed the capillary tube formation of HUVEC on the Matrigel in the presence or absence of 20 ng/ml vascular endothelial growth factor (VEGF). The Matrix metalloproteinases (MMPs), such as MMP-2 and MMP-9, which play an important role in the degradation of basement membrane in angiogenesis and tumor metastasis present in the culture supernatant of Rg3-treated aortic ring culture were found to decrease in their gelatinolytic activities. Taken together, these data underpin the anti-tumor properties of Rg3 through its angiosuppressive activity (Yue et al., 2006).

Glioblastoma

Rg3 has been reported to exert anti-cancer activities through inhibition of angiogenesis and cell proliferation. The mechanisms of apoptosis by ginsenoside Rg3 were related with the MEK signaling pathway and reactive oxygen species. Our data suggest that ginsenoside Rg3 is a novel agent for the chemotherapy of glioblastoma multiforme (GBM) (Choi et al., 2013).

Sin, Kim, & Kim (2012) report that chronic treatment with Rg3 in a sub-lethal concentration induced senescence-like growth arrest in human glioma cells. Rg3-induced senescence was partially rescued when the p53/p21 pathway was inactivated. Data indicate that Rg3 induces senescence-like growth arrest in human glioma cancer through the Akt and p53/p21-dependent signaling pathways.

MDR/Enhanced Paclitaxel Absorption

The penetration of paclitaxel through the Caco-2 monolayer from the apical side to the basal side was facilitated by 20(s)-ginsenoside Rg3 in a concentration-dependent manner. Rg3 also inhibited P-glycoprotein (P-gp), and the maximum inhibition was achieved at 80 µM (p < 0.05). The relative bioavailability (RB)% of paclitaxel with 20(s)-ginsenoside Rg3 was 3.4-fold (10 mg/kg) higher than that of the control. Paclitaxel (20 mg/kg) co-administered with 20(s)-ginsenoside Rg3 (10 mg/kg) exhibited an effective anti-tumor activity with the relative tumor growth rate (T/C) values of 39.36% (p <0.05).

The results showed that 20(s)-ginsenoside Rg3 enhanced the oral bioavailability of paclitaxel in rats and improved the anti-tumor activity in nude mice, indicating that oral co-administration of paclitaxel with 20(s)-ginsenoside Rg3 could provide an effective strategy in addition to the established i.v. route (Yang et al., 2012).

Prostate Cancer

The anti-proliferation effect of Rg3 on prostate cancer cells has been well reported. Rg3 treatment triggered the activation of p38 MAPK; and SB202190, a specific inhibitor of p38 MAPK, antagonized the Rg3-induced regulation of AQP1 and cell migration, suggesting a crucial role for p38 in the regulation process. Rg3 effectively suppresses migration of PC-3M cells by down-regulating AQP1 expression through p38 MAPK pathway and some transcription factors acting on the AQP1 promoter (Pan et al., 2012).

Enhances Chemotherapy

The clinical use of cisplatin (cis-diamminedichloroplatinum II) has been limited by the frequent emergence of cisplatin-resistant cell populations and numerous other adverse effects. Therefore, new agents are required to improve the therapy and health of cancer patients. Oral administration of ginsenoside Rg3 significantly inhibited tumor growth and promoted the anti-neoplastic efficacy of cisplatin in mice inoculated with CT-26 colon cancer cells. In addition, Rg3 administration remarkably inhibited cisplatin-induced nephrotoxicity, hepatotoxicity and oxidative stress.

Rg3 promotes the efficacy of cisplatin by inhibiting HO-1 and NQO-1 expression in cancer cells and protects the kidney and liver against tissue damage by preventing cisplatin-induced intracellular ROS generation (Lee et al., 2012).

Colon Cancer

Rg3-induced apoptosis in HT-29 cells is mediated via the AMPK signaling pathway, and that 20(S)-Rg3 is capable of inducing apoptosis in colon cancer. Rg3-treated cells displayed several apoptotic features, including DNA fragmentation, proteolytic cleavage of poly (ADP-ribose) polymerase (PARP) and morphological changes. 20(S)-Rg3 down-regulated the expression of anti-apoptotic protein B-cell CLL/lymphoma 2 (Bcl2), up-regulated the expression of pro-apoptotic protein of p53 and Bcl-2-associated X protein (Bax), and caused the release of mitochondrial cytochrome c, PARP, caspase-9 and caspase-3 (Yuan et al., 2010).

Anti-metastatic

Studies have linked Rg3 with anti-metastasis of cancer in vivo and in vitro and the CXC receptor 4 (CXCR4) is a vital molecule in migration and homing of cancer to the docking regions. At a dosage without obvious cytotoxicity, Rg3 treatment elicits a weak CXCR4 stain color, decreases the number of migrated cells in CXCL12-elicited chemotaxis and reduces the width of the scar in wound healing and Rg3 is a new CXCR4 inhibitor (Chen et al., 2011).

References

Chen XP, Qian LL, Jiang H, Chen JH. (2011). Ginsenoside Rg3 inhibits CXCR4 expression and related migrations in a breast cancer cell line. Int J Clin Oncol, 16(5):519-23. doi: 10.1007/s10147-011-0222-6.


Choi YJ, Lee HJ, Kang DW, et al. (2013). Ginsenoside Rg3 induces apoptosis in the U87MG human glioblastoma cell line through the MEK signaling pathway and reactive oxygen species. Oncol Rep. doi: 10.3892/or.2013.2555.


Lee CK, Park KK, Chung AS, Chung WY. (2012). Ginsenoside Rg3 enhances the chemosensitivity of tumors to cisplatin by reducing the basal level of nuclear factor erythroid 2-related factor 2-mediated heme oxygenase-1/NAD(P)H quinone oxidoreductase-1 and prevents normal tissue damage by scavenging cisplatin-induced intracellular reactive oxygen species. Food Chem Toxicol, 50(7):2565-74. doi: 10.1016/j.fct.2012.01.005.


Pan XY, Guo H, Han J, et al. (2012). Ginsenoside Rg3 attenuates cell migration via inhibition of aquaporin 1 expression in PC-3M prostate cancer cells. Eur J Pharmacol, 683(1-3):27-34. doi: 10.1016/j.ejphar.2012.02.040.


Sin S, Kim SY, Kim SS. (2012). Chronic treatment with ginsenoside Rg3 induces Akt-dependent senescence in human glioma cells. Int J Oncol., 41(5):1669-74. doi: 10.3892/ijo.2012.1604.


Yang LQ, Wang B, Gan H, et al. (2012). Enhanced oral bioavailability and anti-tumor effect of paclitaxel by 20(s)-ginsenoside Rg3 in vivo. Biopharm Drug Dispos., 33(8):425-36. doi: 10.1002/bdd.1806.


Yuan HD, Quan HY, Zhang Y, et al. (2010). 20(S)-Ginsenoside Rg3-induced apoptosis in HT-29 colon cancer cells is associated with AMPK signaling pathway. Mol Med Rep., 3(5):825-31. doi: 10.3892/mmr.2010.328.


Yue PY, Wong DY, Wu PK, et al. (2006). The angiosuppressive effects of 20 (R)-ginsenoside Rg3. Biochem Pharmacol, 72(4):437-45.

Oxymatrine (Ku Shen)

Cancer:
Sarcoma, pancreatic, breast, liver, lung, oral, colorectal, stomach, gastric, adenoid cystic carcinoma

Action: Anti-angiogenesis, anti-inflammatory, anti-proliferative, chemo-sensitizer, chemotherapy support, cytostatic, radiation support, immunotolerance, induces apoptosis, decreases side-effects of Intensity Modulated Radiation Therapy (IMRT), Transcatheter Hepatic Arterial Chemoembolization (TACE)

Anti-cancer

Oxymatrine, isolated from the dried roots of Sophora flavescens (Aiton), has a long history of use in traditional Chinese medicine to treat inflammatory diseases and cancer. Kushen alkaloids (KS-As) and kushen flavonoids (KS-Fs) are well-characterized components in kushen. KS-As containing oxymatrine, matrine, and total alkaloids have been developed in China as anti-cancer drugs. More potent anti-tumor activities were identified in KS-Fs than in KS-As in vitro and in vivo (Sun et al., 2012).

Angiogenesis

Oxymatrine has been found to inhibit angiogenesis when administered by injection. The tumor-inhibitory rate and the vascular density were tested in animal tumor model with experimental treatment. The expression of VEGF and bFGF were measured by immunistological methods. When high doses were used, the tumor-inhibitory rate of oxymatrine was 31.36%, and the vascular density of S180 sarcoma was lower than that in the control group, and the expression of VEGF and bFGF was down-regulated. Oxymatrine hence has an inhibitory effect on S180 sarcoma and strong inhibitory effects on angiogenesis. Its mechanism may be associated with the down-regulating of VEGF and bFGF expression (Kong et al., 2003).

Immunotolerance

Matrine, a small molecule derived from the root of Sophora flavescens AIT, was demonstrated to be effective in inducing T cell anergy in human Jurkat cells. Induction of immunotolerance has become a new strategy for treating autoimmune conditions in recent decades. However, so far there is no ideal therapeutics available for clinical use. Medicinal herbs are a promising potential source of immunotolerance inducers. Bioactive compounds derived from medicinal plants were screened for inducing T cell anergy in comparison with the effect of well-known T cell anergy inducer, ionomycin.

The results showed that passage of the cells, and concentration and stimulation time of ionomycin on the cells, could influence the ability of T cell anergy induction. The cells exposed to matrine showed markedly decreased mRNA expression of interleukin-2, an indicator of T cell anergy, when the cells were stimulated by antigens, anti-OKT3 plus anti-CD28. Mechanistic study showed that ionomycin and matrine could up-regulate the anergy-associated gene expressions of CD98 and Jumonji and activate nuclear factor of activated T-cells (NFAT) nuclear translocation in absence of cooperation of AP-1 in Jurkat cells. Pre-incubation with matrine or ionomycin could also shorten extracellular signal-regulated kinase (ERK) and suppress c-Jun NH(2)-terminal kinase (JNK) expression on the anergic Jurkat cells when the cells were stimulated with anti-OKT-3 plus anti-CD28 antibodies. Thus, matrine is a strong candidate for further investigation as a T cell immunotolerance inducer (Li et al., 2010).

Induces Apoptosis

The cytotoxic effects of oxymatrine on MNNG/HOS cells were examined by MTT and bromodeoxyuridine (BrdU) incorporation assays. The percentage of apoptotic cells and the level of mitochondrial membrane potential ( Δψ m) were assayed by flow cytometry. The levels of apoptosis-related proteins were measured by Western blot analysis or enzyme assay Kit.

Results showed that treatment with oxymatrine resulted in a significant inhibition of cell proliferation and DNA synthesis in a dose-dependent manner, which has been attributed to apoptosis. Oxymatrine considerably inhibited the expression of Bcl-2 whilst increasing that of Bax.

Oxymatrine significantly suppressed tumor growth in female BALB/C nude mice bearing MNNG/HOS xenograft tumors. In addition, no evidence of drug-related toxicity was identified in the treated animals by comparing the body weight increase and mortality (Zhang et al., 2013).

Pancreatic Cancer

Cell viability assay showed that treatment of PANC-1 pancreatic cancer cells with oxymatrine resulted in cell growth inhibition in a dose- and time-dependent manner. Oxymatrine decreased the expression of angiogenesis-associated factors, including nuclear factor κB (NF-κB) and vascular endothelial growth factor (VEGF). Finally, the anti-proliferative and anti-angiogenic effects of oxymatrine on human pancreatic cancer were further confirmed in pancreatic cancer xenograft tumors in nude mice (Chen et al., 2013).

Induces Apoptosis in Pancreatic Cancer

Oxymatrine inhibited cell viability and induced apoptosis of PANC-1 cells in a time- and dose-dependent manner. This was accompanied by down-regulated expression of Livin and Survivin genes while the Bax/Bcl-2 ratio was up-regulated. Furthermore, oxymatrine treatment led to the release of cytochrome c and activation of caspase-3 proteins. Oxymatrine can induce apoptotic cell death of human pancreatic cancer, which might be attributed to the regulation of Bcl-2 and IAP families, release of mitochondrial cytochrome c, and activation of caspase-3 (Ling et al., 2011).

Decreases Side-effects of Intensity Modulated Radiation Therapy (IMRT)

The levels of sIL-2R and IL-8 in peripheral blood cells of patients with rectal cancer were measured after treatment with the compound matrine, in combination with radiation. Eighty-four patients diagnosed with rectal carcinoma were randomly divided into two groups: therapeutic group and control group.

The patients in the therapeutic group were treated with compound matrine and intensity- modulated radiation therapy (IMRT) (30 Gy/10 f/2 W), while the patients in control group were treated with IMRT. The clinical effects and the levels of IL-8 and sIL-2R tested by ELISA pre-radiation and post-radiation were compared. In addition, 42 healthy people were singled out from the physical examination center in the People's Hospital of Yichun city, which were considered as healthy controls.

The clinical effect and survival rate in the therapeutic group was significantly higher (47.6%) than those in the control group (21.4%). All patients were divided by improvement, stability, and progression of disease in accordance with Karnofsky Performance Scale (KPS). According to the KPS, 16 patients had improvement, 17 stabilized and 9 had disease progress, in the therapeutic group. However, the control group had 12 improvements, 14 stabilized, and 16 progress.

The quality of life in the therapeutic group was higher than tthat in the control group, by rank sum test. SIL-2R and IL-8 examination found that serum levels of sIL-2R and IL-8 were higher in rectal cancer patients before treatments than those in the healthy groups, by student test.

However, sIL-2R and IL-8 serum levels were found significantly lower in the 84 rectal cancer patients after radiotherapy. The level of sIL-2R and IL-8 in the therapeutic group was lower on the first and 14th day, post-radiation, when compared to the control group. However, there was no significant difference on the first day and 14th day, between both experimental groups post- therapy, according to the student test. Side-effects of hepatotoxicity (11.9%) and radiation proctitis (9.52%) were fewer in the therapeutic group.

Compound matrine can decrease the side-effects of IMRT, significantly inhibit sIL-2R and IL-8 in peripheral blood from radiation, and can improve survival quality in patients with rectal cancer (Yin et al., 2013).

Gastric Cancer

The clinical effect of matrine injection, combined with S-1 and cisplatin (SP), in the treatment of advanced gastric cancer was investigated. Seventy-six cases of advanced gastric cancer were randomly divided into either an experimental group or control group. Patients in the two groups were treated with matrine injection combined with SP regimen, or SP regimen alone, respectively.

The effectiveness rate of the experimental group and control group was 57.5% and 52.8% respectively. Therapeutic effect of the two groups of patients did not differ significantly. Occurrence rate of symptom indexes in the treatment group were lower than those of control group, with exception of nausea and vomiting, in which there was no significant difference.

The treatment of advanced gastric cancer with matrine injection, combined with the SP regimen, can significantly improve levels of white blood cells and hemoglobin, liver function, incidence of diarrhea and constipation, and neurotoxicity, to improve the quality of life in patients with advanced gastric cancer (Xia, 2013).

Adenoid Cystic Carcinoma

The effects of compound radix Sophorae flavescentis injection on proliferation, apoptosis and Caspase-3 expression in human adenoid cystic carcinoma ACC-2 cells was investigated.

Compound radix Sophorae flavescentis injection could inhibit the proliferation of ACC-2 cells in vitro, and the dosage effect relationship was significant (P < 0.01). IC50 of ACC-2 was 0.84 g/ml. Flow cytometry indicated that radix Sophorae flavescentis injection could arrest ACC-2 cells at the G0/G1 phase, with a gradual decrease of presence in the G2/M period and S phase. With an increase in dosage, ACC-2 cell apoptosis rate increased significantly (P < 0.05 or P < 0.01).

Radix Sophorae flavescentis injection could enhance ACC-2 cells Caspase-3 protein expression (P < 0.05 or P < 0.01), in a dose-dependent manner. It also could effectively restrain human adenoid cystic carcinoma ACC-2 cells Caspases-3 protein expression, and induce apoptosis, inhibiting tumor cell proliferation (Shi & Hu, 2012).

Breast Cancer Post-operative Chemotherapy

A retrospective analysis of oncological data of 70 post-operative patients with breast cancer from January 2008 to August 2011 was performed. According to the treatment method, the patients were divided into a therapy group (n=35) or control group (n=35). Patients in the control group were treated with the taxotere, adriamycin and cyclophosphamide regimen (TAC). The therapy group was treated with a combination of TAC and sophora root injection. Improved quality of life and incidence of adverse events, before and after treatment, for 2 cycles (21 days to a cycle) were compared.

The objective remission rate of therapy group compared with that of control group was not statistically significant (P > 0.05), while the difference of the disease control rate in two groups was statistically significant (P < 0.05). The improvement rate of total quality of life in the therapy group was higher than that of the control group (P < 0.05). The drop of white blood cells and platelets, gastrointestinal reaction, elevated SGPT, and the incidence of hair loss in the therapy group were lower than those of the control group (P < 0.05).

Sophora root injection combined with chemotherapy in treatment of breast cancer can enhance the effect of chemotherapy, reduce toxicity and side-effects, and improve quality of life (An, An & Wu, 2012).

Lung Cancer Pleural Effusions

The therapeutic efficiency of fufangkushen injection, IL-2, α-IFN on lung cancer accompanied with malignancy pleural effusions, was observed.

One hundred and fifty patients with lung cancer, accompanied with pleural effusions, were randomly divided into treatment and control groups. The treatment group was divided into three groups: injected fufangkushen plus IL-2, fufangkushen plus α-tFN, and IL-2 plus α-IFN, respectively. The control group was divided into three groups and injected fufangkushen, IL-2 and α-IFN, respectively. Therapeutic efficiency and adverse reactions were observed after four weeks.

The effective rate of fufangkushen, IL-2, and α-IFN in a combination was significantly superior to single pharmacotherapy. The effective rate of fufangkushen plus ct-IFN was highest. In adverse reactions, the incidence of fever, chest pains, and the reaction of gastrointestinal tract in the treatment group were significantly less than in the matched group.

The effect of fufangkushen, IL-2, and α-IFN, in a combination, on lung cancer with pleural effusions was significantly better than single pharmacotherapy. Moreover, the effect of fufangknshen plus IL-2 or α-IFN had the greatest effect (Hu & Mei, 2012).

Colorectal Cancer Immunologic Function

The effects of compound Kushen (Radix sophorae flavescentis) injection on the immunologic function of patients after colorectal cancer resection, were studied.

Eighty patients after colorectal cancer resection were randomly divided into two groups: 40 patients in the control group were treated with routine chemotherapy including 5-fluorouridine(5-FU), calcium folinate(CF) and oxaliplatin, and 40 patients in the experimental group were treated with the same chemotherapy regime combined with 20 mL·d-1 compound Kushen injection, for 10 days during chemotherapy.

In the control group the numbers of CD3+,CD4+T cells, NK cells and CD4+/CD8+ ratio significantly declined relative to prior to chemotherapy (P < 0.05), while CD8+T lymphocyte number increased significantly. In the experimental group, there were no significant differences between the numbers of CD3+,CD4+,CD8+T cells, NK cells, and CD4+/CD8+ ratio, before and after chemotherapy (P > 0.05).

After chemotherapy, the numbers of CD3+,CD4+T cells, NK cells and CD4+/CD8+ ratio were higher in the experimental group than in the control group (P0.05), while the number of CD8+T lymphocyte was similar between two groups. Compound Kushen injection can improve the immunologic function of patients receiving chemotherapy after colorectal cancer resection (Chen, Yu, Yuan, & Yuan, 2009).

Stage III and IV non-small-cell lung cancer (NSCLC)

A total of 286 patients with advanced NSCLC were enrolled for study. The patients were treated with either compound Kushen injection in combination with NP (NVB + CBP) chemotherapy (vinorelbine and carboplatin, n = 144), or with NP (NVB + CBP) chemotherapy alone (n = 142). The chemotherapy was performed for 4 cycles of 3 weeks, and the therapeutic efficacy was evaluated every 2 weeks. The following indicators were observed: levels of Hb, WBC, PLT and T cell subpopulations in blood, serum IgG level, short-term efficacy, adverse effects and quality of life.

The gastrointestinal reactions and the myelosuppression in the combination chemotherapy group were alleviated when compared with the chemotherapy alone group, showing a significant difference. (P < 0.05). CD (8)(+) cells were markedly declined in the combination chemotherapy group, and the CD (4)(+)/CD (8)(+) ratio showed an elevation trend in the chemotherapy alone group.

The Karnofsky Performance Scale (KPS) scores and serum IgM and IgG levels were higher in the combination chemotherapy group than those in the chemotherapy alone group (P < 0.01 and P < 0.05). The serum lgA levels were not significantly different in the two groups.

The compound Kushen injection plus NP chemotherapy regimen showed better therapeutic effect, reduced adverse effects of chemotherapy and improved the quality of life in patients with stage III and IV NSCLC (Fan et al., 2010).

Lung Adenocarcinoma

Suppression effects of different concentrations of matrine injection and matrine injection combined with anti-tumor drugs on lung cancer cells were measured by methyl thiazolyl tetrazolium (MTT) colorimetric assay.

Different concentrations of matrine injection could inhibit the growth of SPCA/I human lung adenocarcinoma cells. There was a positive correlation between the inhibition rate and the drug concentration. Different concentrations of matrine injection combined with anti-tumor drugs had a higher growth inhibition rate than anti-tumor drugs alone.

Matrine injection has direct growth suppression effect on SPCA/I human lung adenocarcinoma cells and SS+ injection combined with anti-tumor drugs shows a significant synergistic effect on tumor cells (Zhu, Jiang, Lu, Guo, & Gan, 2008).

Transcatheter Hepatic Arterial Chemoembolization (TACE)

The effect of composite Kushen injection combined with transcatheter hepatic arterial chemoembolization (TACE) on unresectable primary liver cancer, was studied.

Fifty-seven patients with unresectable primary liver cancer were randomly divided into two groups. The treatment group with 27 cases was treated by TACE combined with composite Kushen injection, and the control group with 30 cases was treated by TACE alone. The clinical curative effects were observed after treatment in both groups.

One-, 2-, and 3-year survival rates of the treatment group were 67%, 48%, and 37% respectively, and those of control group were 53%, 37%, and 20% respectively. There were significant differences between both groups (P < 0.05).

Combined TACE with composite Kushen injection can increase the efficacy of patients with unresectable primary liver cancer (Wang & Cheng, 2009).

References

An AJ, An GW, Wu YC. (2012). Observation of compound recipe light yellow Sophora root injection combined with chemotherapy in treatment of 35 postoperative patients with breast cancer. Medical & Pharmaceutical Journal of Chinese People's Liberation Army, 24(10), 43-46. doi: 10.3969/j.issn.2095-140X.2012.10.016.


Chen G, Yu B, Yuan SJ, Yuan Q. (2009). Effects of compound Kushen injection on the immunologic function of patients after colorectal cancer resection. Evaluation and Analysis of Drug-Use in Hospitals of China, 2009(9), R735.3. doi: cnki:sun:yypf.0.2009-09-025.


Chen H, Zhang J, Luo J, et al. (2013) Anti-angiogenic effects of oxymatrine on pancreatic cancer by inhibition of the NF- κ B-mediated VEGF signaling pathway. Oncol Rep, 30(2):589-95. doi: 10.3892/or.2013.2529.


Fan CX, Lin CL, Liang L, et al. (2010). Enhancing effect of compound Kushen injection in combination with chemotherapy for patients with advanced non-small-cell lung cancer. Chinese Journal of Oncology, 32(4), 294-297.


Hu DJ, Mei, XD. (2012). Observing therapeutic efficiency of fufangkushen injection, IL-2, α -IFN on lung cancer accompanied with malignancy pleural effusions. Journal of Clinical Pulmonology, 17(10), 1844-1845.


Kong QZ, Huang DS, Huang T, et al. (2003). Experimental study on inhibiting angiogenesis in mice S180 by injections of three traditional Chinese herbs. Chinese Journal of Hospital Pharmacy, 2003-11. doi: CNKI:SUN:ZGYZ.0.2003-11-002


Li T, Wong VK, Yi XQ, et al. (2010). Matrine induces cell anergy in human Jurkat T cells through modulation of mitogen-activated protein kinases and nuclear factor of activated T-cells signaling with concomitant up-regulation of anergy-associated genes expression. Biol Pharm Bull, 33(1):40-6.


Ling Q, Xu X, Wei X, et al. (2011). Oxymatrine induces human pancreatic cancer PANC-1 cells apoptosis via regulating expression of Bcl-2 and IAP families, and releasing of cytochrome c. J Exp Clin Cancer Res, 30:66. doi: 10.1186/1756-9966-30-66.


Shi B, Xu H. (2012). Effects of compound radix Sophorae flavescentis injection on proliferation, apoptosis and caspase-3 expression in adenoid cystic carcinoma ACC-2 cells. Chinese Pharmacological Bulletin, 5(10), 721-724.


Sun M, Cao H, Sun L, et al. (2012). Anti-tumor activities of kushen: literature review. Evid Based Complement Alternat Med, 2012;2012:373219. doi: 10.1155/2012/373219.


Wang HM, Cheng XM. (2009). Composite Ku Shen injection combined with hepatic artery embolism on unresectable primary liver cancer. Modern Journal of Integrated Traditional Chinese and Western Medicine, 18(2), 1334–1335.


Xia G. (2013). Clinical observation of compound matrine injection combined with SP regimen in advanced gastric cancer. Journal of Liaoning Medical University, 2013(1), 37-38.


Yin WH, Sheng JW, Xia HM, et al. (2013). Study on the effect of compound matrine on the level of sIL-2R and IL-8 in peripheral blood cells of patients with rectal cancer to radiation. Global Traditional Chinese Medicine, 2013(2), 100-104.


Zhang Y, Sun S, Chen J, et al. (2013). Oxymatrine induces mitochondria dependent apoptosis in human osteosarcoma MNNG/HOS cells through inhibition of PI3K/Akt pathway. Tumor Biol.


Zhu MY, Jiang ZH, Lu YW, Guo Y, Gan JJ. (2008). Matrine and anti-tumor drugs in inhibiting the growth of human lung cancer cell line. Journal of Chinese Integrative Medicine, 6(2), 163-165. doi: 10.3736/jcim20080211.

Betulin and Betulinic acid

Cancer:
Neuroblastoma, medulloblastoma, glioblastoma, colon, lung, oesophageal, leukemia, melanoma, pancreatic, prostate, breast, head & neck, myeloma, nasopharyngeal, cervical, ovarian, esophageal squamous carcinoma

Action: Anti-angiogenic effects, induces apoptosis, anti-oxidant, cytotoxic and immunomodifying activities

Betulin is a naturally occurring pentacyclic triterpene found in many plant species including, among others, in Betula platyphylla (white birch tree), Betula X caerulea [Blanch. (pro sp.)], Betula cordifolia (Regel), Betula papyrifera (Marsh.), Betula populifolia (Marsh.) and Dillenia indica L . It has anti-retroviral., anti-malarial., and anti-inflammatory properties, as well as a more recently discovered potential as an anti-cancer agent, by inhibition of topoisomerase (Chowdhury et al., 2002).

Betulin is found in the bark of several species of plants, principally the white birch (Betula pubescens ) (Tan et al., 2003) from which it gets its name, but also the ber tree (Ziziphus mauritiana ), selfheal (Prunella vulgaris ), the tropical carnivorous plants Triphyophyllum peltatum and Ancistrocladus heyneanus, Diospyros leucomelas , a member of the persimmon family, Tetracera boiviniana , the jambul (Syzygium formosanum ) (Zuco et al., 2002), flowering quince (Chaenomeles sinensis ) (Gao et al., 2003), rosemary (Abe et al., 2002) and Pulsatilla chinensis (Ji et al., 2002).

Anti-cancer, Induces Apoptosis

The in vitro characterization of the anti-cancer activity of betulin in a range of human tumor cell lines (neuroblastoma, rhabdomyosarcoma-medulloblastoma, glioma, thyroid, breast, lung and colon carcinoma, leukaemia and multiple myeloma), and in primary tumor cultures isolated from patients (ovarian carcinoma, cervical carcinoma and glioblastoma multiforme) was carried out to probe its anti-cancer effect. The remarkable anti-proliferative effect of betulin in all tested tumor cell cultures was demonstrated. Furthermore, betulin altered tumor cell morphology, decreased their motility and induced apoptotic cell death. These findings demonstrate the anti-cancer potential of betulin and suggest that it may be applied as an adjunctive measure in cancer treatment (Rzeski, 2009).

Lung Cancer

Betulin has also shown anti-cancer activity on human lung cancer A549 cells by inducing apoptosis and changes in protein expression profiles. Differentially expressed proteins explained the cytotoxicity of betulin against human lung cancer A549 cells, and the proteomic approach was thus shown to be a potential tool for understanding the pharmacological activities of pharmacophores (Pyo, 2009).

Esophageal Squamous Carcinoma

The anti-tumor activity of betulin was investigated in EC109 cells. With the increasing doses of betulin, the inhibition rate of EC109 cell growth was increased, and their morphological characteristics were changed significantly. The inhibition rate showed dose-dependent relation.

Leukemia

Betulin hence showed potent inhibiting effects on EC109 cells growth in vitro (Cai, 2006).

A major compound of the methanolic extract of Dillenia indica L. fruits, betulinic acid, showed significant anti-leukaemic activity in human leukaemic cell lines U937, HL60 and K562 (Kumar, 2009).

Betulinic acid effectively induces apoptosis in neuroectodermal and epithelial tumor cells and exerts little toxicity in animal trials. It has been shown that betulinic acid induced marked apoptosis in 65% of primary pediatric acute leukemia cells and all leukemia cell lines tested. When compared for in vitro efficiency with conventionally used cytotoxic drugs, betulinic acid was more potent than nine out of 10 standard therapeutics and especially efficient in tumor relapse. In isolated mitochondria, betulinic acid induced release of both cytochrome c and Smac. Taken together, these results indicated that betulinic acid potently induces apoptosis in leukemia cells and should be further evaluated as a future drug to treat leukemia (Ehrhardt, 2009).

Multiple Myeloma

The effect of betulinic acid on the induction apoptosis of human multiple myeloma RPMI-8226 cell line was investigated. The results showed that within a certain concentration range (0, 5, 10, 15, 20 microg/ml), IC50 of betulinic acid to RPMI-8226 at 24 hours was 10.156+/-0.659 microg/ml, while the IC50 at 48 hours was 5.434+/-0.212 microg/ml, and its inhibiting effect on proliferation of RPMI-8226 showed both a time-and dose-dependent manner.

It is therefore concluded that betulinic acid can induce apoptosis of RPMI-8226 within a certain range of concentration in a time- and dose-dependent manner. This phenomenon may be related to the transcriptional level increase of caspase 3 gene and decrease of bcl-xl. Betulinic acid also affects G1/S in cell-cycle which arrests cells at phase G0/G1 (Cheng, 2009).

Anti-angiogenic Effects, Colorectal Cancer

Betulinic acid isolated from Syzygium campanulatum Korth (Myrtaceae) was found to have anti-angiogenic effects on rat aortic rings, matrigel tube formation, cell proliferation and migration, and expression of vascular endothelial growth factor (VEGF). The anti-tumor effect was studied using a subcutaneous tumor model of HCT 116 colorectal carcinoma cells established in nude mice. Anti-angiogenesis studies showed potent inhibition of microvessels outgrowth in rat aortic rings, and studies on normal and cancer cells did not show any significant cytotoxic effect.

In vivo anti-angiogenic study showed inhibition of new blood vessels in chicken embryo chorioallantoic membrane (CAM), and in vivo anti-tumor study showed significant inhibition of tumor growth due to reduction of intratumor blood vessels and induction of cell death. Collectively, these results indicate betulinic acid as an anti-angiogenic and anti-tumor candidate (Aisha, 2013).

Nasopharyngeal Carcinoma Melanoma, Leukemia, Lung, Colon, Breast,Prostate, Ovarian Cancer

Betulinic acid is an effective and potential anti-cancer chemical derived from plants. Betulinic acid can kill a broad range of tumor cell lines, but has no effect on untransformed cells. The chemical also kills melanoma, leukemia, lung, colon, breast, prostate and ovarian cancer cells via induction of apoptosis, which depends on caspase activation. However, no reports are yet available about the effects of betulinic acid on nasopharyngeal carcinoma (NPC), a widely spread malignancy in the world, especially in East Asia.

In a study, Liu & Luo (2012) showed that betulinic acid can effectively kill CNE2 cells, a cell line derived from NPC. Betulinic acid-induced CNE2 apoptosis was characterized by typical apoptosis hallmarks: caspase activation, DNA fragmentation, and cytochrome c release.

These observations suggest that betulinic acid may serve as a potent and effective anti-cancer agent in NPC treatment. Further exploration of the mechanism of action of betulinic acid could yield novel breakthroughs in anti-cancer drug discovery.

Cervical Carcinoma

Betulinic acid has shown anti-tumor activity in some cell lines in previous studies. Its anti-tumor effect and possible mechanisms were investigated in cervical carcinoma U14 tumor-bearing mice. The results showed that betulinic acid (100 mg/kg and 200 mg/kg) effectively suppressed tumor growth in vivo. Compared with the control group, betulinic acid significantly improved the levels of IL-2 and TNF-alpha in tumor-bearing mice and increased the number of CD4+ lymphocytes subsets, as well as the ratio of CD4+/CD8+ at a dose of 200 mg/kg.

Furthermore, treatment with betulinic acid induced cell apoptosis in a dose-dependent manner in tumor-bearing mice, and inhibited the expression of Bcl-2 and Ki-67 protein while upregulating the expression of caspase-8 protein. The mechanisms by which BetA exerted anti-tumor effects might involve the induction of tumor cell apoptosis. This process is also related to improvement in the body's immune response (Wang, 2012).

Anti-oxidant, Cytotoxic and Immunomodifying Activities

Betulinic acid exerted cytotoxic activity through dose-dependent impairment of viability and mitochondrial activity of rat insulinoma m5F (RINm5F) cells. Decrease of RINm5F viability was mediated by nitric oxide (NO)-induced apoptosis. Betulinic acid also potentiated NO and TNF-α release from macrophages therefore enhancing their cytocidal action. The rosemary extract developed more pronounced anti-oxidant, cytotoxic and immunomodifying activities, probably due to the presence of betulinic acid (Kontogianni, 2013).

Pancreatic Cancer

Lamin B1 is a novel therapeutic target of Betulinic Acid in pancreatic cancer. The role and regulation of lamin B1 (LMNB1) expression in human pancreatic cancer pathogenesis and betulinic acid-based therapy was investigated. Lamin proteins are thought to be involved in nuclear stability, chromatin structure and gene expression. Elevation of circulating LMNB1 marker in plasma could detect early stages of HCC patients, with 76% sensitivity and 82% specificity. Lamin B1 is a clinically useful biomarker for early stages of HCC in tumor tissues and plasma (Sun, 2010).

It was found that lamin B1 was significantly down-regulated by BA treatment in pancreatic cancer in both in vitro culture and xenograft models. Overexpression of lamin B1 was pronounced in human pancreatic cancer and increased lamin B1 expression was directly associated with low grade differentiation, increased incidence of distant metastasis and poor prognosis of pancreatic cancer patients.

Furthermore, knockdown of lamin B1 significantly attenuated the proliferation, invasion and tumorigenicity of pancreatic cancer cells. Lamin B1 hence plays an important role in pancreatic cancer pathogenesis and is a novel therapeutic target of betulinic acid treatment (Li, 2013).

Multiple Myeloma, Prostate Cancer

The inhibition of the ubiquitin-proteasome system (UPS) of protein degradation is a valid anti-cancer strategy and has led to the approval of bortezomib for the treatment of multiple myeloma. However, the alternative approach of enhancing the degradation of oncoproteins that are frequently overexpressed in cancers is less developed. Betulinic acid (BA) is a plant-derived small molecule that can increase apoptosis specifically in cancer but not in normal cells, making it an attractive anti-cancer agent.

Results in prostate cancer suggest that BA inhibits multiple deubiquitinases (DUBs), which results in the accumulation of poly-ubiquitinated proteins, decreased levels of oncoproteins, and increased apoptotic cell death. In the TRAMP transgenic mouse model of prostate cancer, treatment with BA (10 mg/kg) inhibited primary tumors, increased apoptosis, decreased angiogenesis and proliferation, and lowered androgen receptor and cyclin D1 protein.

BA treatment also inhibited DUB activity and increased ubiquitinated proteins in TRAMP prostate cancer but had no effect on apoptosis or ubiquitination in normal mouse tissues. Overall, this data suggests that BA-mediated inhibition of DUBs and induction of apoptotic cell death specifically in prostate cancer but not in normal cells and tissues may provide an effective non-toxic and clinically selective agent for chemotherapy (Reiner, 2013).

Melanoma

Betulinic acid was recently described as a melanoma-specific inducer of apoptosis, and it was investigated for its comparable efficacy against metastatic tumors and those in which metastatic ability and 92-kD gelatinase activity had been decreased by introduction of a normal chromosome 6. Human metastatic C8161 melanoma cells showed greater DNA fragmentation and growth arrest and earlier loss of viability in response to betulinic acid than their non-metastatic C8161/neo 6.3 counterpart.

These effects involved induction of p53 without activation of p21WAF1 and were synergized by bromodeoxyuridine in metastatic Mel Juso, with no comparable responses in non-metastatic Mel Juso/neo 6 cells. These data suggest that betulinic acid exerts its inhibitory effect partly by increasing p53 without a comparable effect on p21WAF1 (Rieber, 1998).

As a result of bioassay–guided fractionation, betulinic acid has been identified as a melanoma-specific cytotoxic agent. In follow-up studies conducted with athymic mice carrying human melanomas, tumor growth was completely inhibited without toxicity. As judged by a variety of cellular responses, anti-tumor activity was mediated by the induction of apoptosis. Betulinic acid is inexpensive and available in abundant supply from common natural sources, notably the bark of white birch trees. The compound is currently undergoing preclinical development for the treatment or prevention of malignant melanoma (Pisha, 1995).

Betulinic acid strongly and consistently suppressed the growth and colony-forming ability of all human melanoma cell lines investigated. In combination with ionizing radiation the effect of betulinic acid on growth inhibition was additive in colony-forming assays.

Betulinic acid also induced apoptosis in human melanoma cells as demonstrated by Annexin V binding and by the emergence of cells with apoptotic morphology. The growth-inhibitory action of betulinic acid was more pronounced in human melanoma cell lines than in normal human melanocytes.

The properties of betulinic acid make it an interesting candidate, not only as a single agent but also in combination with radiotherapy. It is therefore concluded that the strictly additive mode of growth inhibition in combination with irradiation suggests that the two treatment modalities may function by inducing different cell death pathways or by affecting different target cell populations (Selzer, 2000).

Betulinic acid has been demonstrated to induce programmed cell death with melanoma and certain neuroectodermal tumor cells. It has been demonstrated currently that the treatment of cultured UISO-Mel-1 (human melanoma cells) with betulinic acid leads to the activation of p38 and stress activated protein kinase/c-Jun NH2-terminal kinase (a widely accepted pro-apoptotic mitogen-activated protein kinases (MAPKs)) with no change in the phosphorylation of extracellular signal-regulated kinases (anti-apoptotic MAPK). Moreover, these results support a link between the MAPKs and reactive oxygen species (ROS).

These data provide additional insight in regard to the mechanism by which betulinic acid induces programmed cell death in cultured human melanoma cells, and it likely that similar responses contribute to the anti-tumor effect mediated with human melanoma carried in athymic mice (Tan, 2003).

Glioma

Betulinic acid triggers apoptosis in five human glioma cell lines. Betulinic acid-induced apoptosis requires new protein, but not RNA, synthesis, is independent of p53, and results in p21 protein accumulation in the absence of a cell-cycle arrest. Betulinic acid-induced apoptosis involves the activation of caspases that cleave poly(ADP ribose)polymerase.

Betulinic acid induces the formation of reactive oxygen species that are essential for BA-triggered cell death. The generation of reactive oxygen species is blocked by BCL-2 and requires new protein synthesis but is unaffected by caspase inhibitors, suggesting that betulinic acid toxicity sequentially involves new protein synthesis, formation of reactive oxygen species, and activation of crm-A-insensitive caspases (Wolfgang, 1999).

Head and Neck Carcinoma

In two head and neck squamous carcinoma (HNSCC) cell lines betulinic acid induced apoptosis, which was characterized by a dose-dependent reduction in cell numbers, emergence of apoptotic cells, and an increase in caspase activity. Western blot analysis of the expression of various Bcl-2 family members in betulinic acid–treated cells showed, surprisingly, a suppression of the expression of the pro-apoptotic protein Bax but no changes in Mcl-1 or Bcl-2 expression.

These data clearly demonstrate for the first time that betulinic acid has apoptotic activity against HNSCC cells (Thurnher et al., 2003).

References

Abe F, Yamauchi T, Nagao T, et al. (2002). Ursolic acid as a trypanocidal constituent in rosemary. Biological & Pharmaceutical Bulletin, 25(11):1485–7. doi:10.1248/bpb.25.1485. PMID 12419966.


Aisha AF, Ismail Z, Abu-Salah KM, et al. (2013). Syzygium campanulatum korth methanolic extract inhibits angiogenesis and tumor growth in nude mice. BMC Complement Altern Med,13:168. doi: 10.1186/1472-6882-13-168.


Cai WJ, Ma YQ, Qi YM et al. (2006). Ai bian ji bian tu bian can kao wen xian ge shi    Carcinogenesis,Teratogenesis & Mutagenesis,18(1):16-8.


Cheng YQ, Chen Y, Wu QL, Fang J, Yang LJ. (2009). Zhongguo Shi Yan Xue Ye Xue Za Zhi, 17(5):1224-9.


Chowdhury AR, Mandal S, Mittra B, et al. (2002). Betulinic acid, a potent inhibitor of eukaryotic topoisomerase I: identification of the inhibitory step, the major functional group responsible and development of more potent derivatives. Medical Science Monitor, 8(7): BR254–65. PMID 12118187.


Ehrhardt H, Fulda S, FŸhrer M, Debatin KM & Jeremias I. (2004). Betulinic acid-induced apoptosis in leukemia cells. Leukemia, 18:1406–1412. doi:10.1038/sj.leu.2403406


Gao H, Wu L, Kuroyanagi M, et al. (2003). Anti-tumor-promoting constituents from Chaenomeles sinensis KOEHNE and their activities in JB6 mouse epidermal cells. Chemical & Pharmaceutical Bulletin, 51(11):1318–21. doi:10.1248/cpb.51.1318. PMID 14600382.


Ji ZN, Ye WC, Liu GG, Hsiao WL. (2002). 23-Hydroxybetulinic acid-mediated apoptosis is accompanied by decreases in bcl-2 expression and telomerase activity in HL-60 Cells. Life Sciences, 72(1):1–9. doi:10.1016/S0024-3205(02)02176-8. PMID 12409140.


Kontogianni VG, Tomic G, Nikolic I, et al. (2013). Phytochemical profile of Rosmarinus officinalis and Salvia officinalis extracts and correlation to their anti-oxidant and anti-proliferative activity. Food Chem,136(1):120-9. doi: 10.1016/j.foodchem.2012.07.091.


Kumar D, Mallick S, Vedasiromoni JR, Pal BC. (2010). Anti-leukemic activity of Dillenia indica L. fruit extract and quantification of betulinic acid by HPLC. Phytomedicine, 17(6):431-5.


Li L, Du Y, Kong X, et al. (2013). Lamin B1 Is a Novel Therapeutic Target of Betulinic Acid in Pancreatic Cancer. Clin Cancer Res, Epub July 9. doi: 10.1158/1078-0432.CCR-12-3630


Liu Y, Luo W. (2012). Betulinic acid induces Bax/Bak-independent cytochrome c release in human nasopharyngeal carcinoma cells. Molecules and cells, 33(5):517-524. doi: 10.1007/s10059-012-0022-5


Pisha E, Chai H, Lee I-S, et al. (1995). Discovery of betulinic acid as a selective inhibitor of human melanoma that functions by induction of apoptosis. Nature Medicine, 1:1046 – 1051. doi: 10.1038/nm1095-1046


Pyo JS, Roh SH, Kim DK, et al. (2009). Anti-Cancer Effect of Betulin on a Human Lung Cancer Cell Line: A Pharmacoproteomic Approach Using 2 D SDS PAGE Coupled with Nano-HPLC Tandem Mass Spectrometry. Planta Med, 75(2): 127-131. doi: 10.1055/s-0028-1088366


Reiner T, Parrondo R, de Las Pozas A, Palenzuela D, Perez-Stable C. (2013). Betulinic Acid Selectively Increases Protein Degradation and Enhances Prostate Cancer-Specific Apoptosis: Possible Role for Inhibition of Deubiquitinase Activity. PLoS One, 8(2):e56234. doi: 10.1371/journal.pone.0056234.


Rieber M & Strasberg-Rieber M. (1998). Induction of p53 without increase in p21WAF1 in betulinic acid-mediated cell death is preferential for human metastatic melanoma. DNA Cell Biol, 17(5):399–406. doi:10.1089/dna.1998.17.399.


Rzeski W, Stepulak A, Szymanski M, et al. (2009). Betulin Elicits Anti-Cancer Effects in Tumor Primary Cultures and Cell Lines In Vitro. Basic and Clinical Pharmacology and Toxicology, 105(6):425–432. doi: 10.1111/j.1742-7843.2009.00471.x


Selzer E, Pimentel E, Wacheck V, et al. (2000). Effects of Betulinic Acid Alone and in Combination with Irradiation in Human Melanoma Cells. Journal of Investigative Dermatology, 114:935–940; doi:10.1046/j.1523-1747.2000.00972.x


Sun S, Xu MZ, Poon RT, Day PJ, Luk JM. (2010). Circulating Lamin B1 (LMNB1) biomarker detects early stages of liver cancer in patients. J Proteome Res, 9(1):70-8. doi: 10.1021/pr9002118.


Tan YM, Yu R, Pezzuto JM. (2003). Betulinic Acid-induced Programmed Cell Death in Human Melanoma Cells Involves Mitogen-activated Protein Kinase Activation. Clin Cancer Res, 9:2866.


Thurnher D, Turhani D, Pelzmann M, et al. (2003). Betulinic acid: A new cytotoxic compound against malignant head and neck cancer cells. Head & Neck. 25(9):732–740. doi: 10.1002/hed.10231


Wang P, Li Q, Li K, Zhang X, et al. (2012). Betulinic acid exerts immunoregulation and anti-tumor effect on cervical carcinoma (U14) tumor-bearing mice. Pharmazie, 67(8):733-9.


Wick W, Grimmel C, Wagenknecht B, Dichgans J, Weller M. (1999). Betulinic Acid-Induced Apoptosis in Glioma Cells: A Sequential Requirement for New Protein Synthesis, Formation of Reactive Oxygen Species, and Caspase Processing. JPET, 289(3):1306-1312.


Zuco V, Supino R, Righetti SC, et al. (2002). Selective cytotoxicity of betulinic acid on tumor cell lines, but not on normal cells. Cancer Letters, 175(1): 17–25. doi:10.1016/S0304-3835(01)00718-2. PMID 11734332.

Oxymatrine or Compound Matrine (Ku Shen)

Cancer: Sarcoma, pancreatic, breast, liver, lung, oral., rectal., stomach, leukemia, adenoid cystic carcinoma

Action: Anti-inflammatory, anti-proliferative, chemo-sensitizer, chemotherapy support, cytostatic, radiation support, anti-angiogenesis

Ingredients: ku shen (Sophora flavescens), bai tu ling (Heterosmilax chinensis).

TCM functions: Clearing Heat, inducing diuresis, cooling Blood, removing Toxin, dispersing lumps and relieving pain (Drug Information Reference in Chinese: See end, 2000-12).

Indications: Pain and bleeding caused by cancer.

Dosage and usage:

Intramuscular injection: 2-4 ml each time, twice daily; intravenous drip: 12 ml mixed in 200 ml NaCl injection, once daily. The total amount of 200 ml administration makes up a course of treatment. 2-3 consecutive courses can be applied.

Anti-cancer

Oxymatrine, isolated from the dried roots of Sophora flavescens (Aiton), has a long history of use in traditional Chinese medicine to treat inflammatory diseases and cancer. Kushen alkaloids (KS-As) and kushen flavonoids (KS-Fs) are well-characterized components in kushen. KS-As containing oxymatrine, matrine, and total alkaloids have been developed in China as anti-cancer drugs. More potent anti-tumor activities were identified in KS-Fs than in KS-As in vitro and in vivo (Sun et al., 2012). The four major alkaloids in compound Ku Shen injection are matrine, sophoridine, oxymatrine and oxysophocarpine (Qi, Zhang, & Zhang, 2013).

Sarcoma

When a high dose was used, the tumor-inhibitory rate of oxymatrine was 31.36%, and the vascular density of S180 sarcoma was lower than that in the control group and the expression of VEGF and bFGF was down-regulated. Oxymatrine hence has an inhibitory effect on S180 sarcoma and strong inhibitory effects on angiogenesis. Its mechanism may be associated with the down-regulating of VEGF and bFGF expression (Kong et al., 2003).

T Cell Leukemia

Matrine, a small molecule derived from the root of Sophora flavescens AIT was demonstrated to be effective in inducing T cell anergy in human T cell leukemia Jurkat cells.

The results showed that passage of the cells, and concentration and stimulation time of ionomycin on the cells could influence the ability of T cell anergy induction.

The cells exposed to matrine showed markedly decreased mRNA expression of interleukin-2, an indicator of T cell anergy. Pre-incubation with matrine or ionomycin could also shorten extracellular signal-regulated kinase (ERK) and suppress c-Jun NH(2)-terminal kinase (JNK) expression on the anergic Jurkat cells when the cells were stimulated with anti-OKT-3 plus anti-CD28 antibodies. Thus, matrine is a strong candidate for further investigation as a T cell immunotolerance inducer (Li et al., 2010).

Osteosarcoma

Results showed that treatment with oxymatrine resulted in a significant inhibition of cell proliferation and DNA synthesis in a dose-dependent manner, which has been attributed to apoptosis. Oxymatrine considerably inhibited the expression of Bcl-2 whilst increasing that of Bax.

Oxymatrine significantly suppressed tumor growth in female BALB/C nude mice bearing osteosarcoma MNNG/HOS xenograft tumors. In addition, no evidence of drug-related toxicity was identified in the treated animals by comparing the body weight increase and mortality (Zhang et al., 2013).

Pancreatic Cancer

Oxymatrine decreased the expression of angiogenesis-associated factors, including nuclear factor κB (NF-κB) and vascular endothelial growth factor (VEGF). Finally, the anti-proliferative and anti-angiogenic effects of oxymatrine on human pancreatic cancer were further confirmed in pancreatic cancer xenograft tumors in nude mice (Chen et al., 2013).

Furthermore, oxymatrine treatment led to the release of cytochrome c and activation of caspase-3 proteins. Oxymatrine can induce apoptotic cell death of human pancreatic cancer, which might be attributed to the regulation of Bcl-2 and IAP families, release of mitochondrial cytochrome c and activation of caspase-3 (Ling et al., 2011).

Rectal Carcinoma

Eighty-four patients diagnosed with rectal carcinoma at the People”s Hospital of Yichun city in Jiangxi province from September 2006 to September 2011, were randomly divided into two groups: therapeutic group and control group. The patients in the therapeutic group were treated with compound matrine and intensity modulated radiation therapy (IMRT) (30 Gy/10 f/2 W), while the patients in control group were treated with IMRT.

The clinical effect and survival rate in the therapeutic group were significantly higher (47.6%) than those in the control group (21.4%). All patients were divided by improvement, stability, and progression of disease in accordance with Karnofsky Performance Scale (KPS). According to the KPS, 16 patients had improvement, 17 stabilized and 9 had disease progress in the therapeutic group.

However, the control group had 12 improvements, 14 stabilized, and 16 disease progress. Quality of life in the therapeutic group was higher than that in the control group by rank sum test. The level of sIL-2R and IL-8 in the therapeutic group was lower on the first and 14th day, post radiation, when compared to the control group. However, there was no significant difference on the first day and 14th day, between both experimental groups post therapy, according to the student test. Compound matrine can decrease the side-effects of IMRT, significantly inhibit sIL-2R and IL-8 in peripheral blood from radiation, and can improve survival quality in patients with rectal cancer (Yin et al., 2013).

Gastric Cancer

Seventy-six cases of advanced gastric cancer were collected from June 2010 to November 2011, and randomly divided into either an experimental group or control group. Patients in the two groups were treated with matrine injection combined with SP regimen, or SP regimen alone, respectively. The effectiveness rate of the experimental group and control group was 57.5% and 52.8% respectively.

The treatment of advanced gastric cancer with matrine injection, combined with the SP regimen, can significantly improve levels of white blood cells and hemoglobin, liver function, incidence of diarrhea and constipation, and neurotoxicity, to improve the quality of life in patients with advanced gastric cancer (Xia, 2013).

Adenoid Cystic Carcinoma

Adenoid cystic carcinoma (ACC-2) cells were cultured in vitro. MTT assay was used to measure the cell proliferative effect. Compound radix Sophorae flavescentis injection could inhibit the proliferation of ACC-2 cells in vitro, and the dosage effect relationship was significant (P < 0.01). Radix Sophorae flavescentis injection could enhance ACC-2 cells Caspase-3 protein expression (P < 0.05 or P < 0.01), in a dose-dependent manner. It also could effectively restrain human adenoid cystic carcinoma ACC-2 cells Caspases-3 protein expression, and induce apoptosis, inhibiting tumor cell proliferation (Shi & Hu, 2012).

Breast Cancer; Chemotherapy

A retrospective analysis of oncological data of 70 postoperative patients with breast cancer from January 2008 to August 2011 was performed. According to the treatment method, the patients were divided into a therapy group (n=35) or control group (n=35). Patients in the control group were treated with the taxotere, adriamycin and cyclophosphamide regimen (TAC). The therapy group was treated with a combination of TAC and sophora root injection. Improved quality of life and incidence of adverse events, before and after treatment, for 2 cycles (21 days for a cycle) were compared.

The improvement rate of total quality of life in the therapy group was higher than that of the control group (P < 0.05). The drop of white blood cells and platelets, gastrointestinal reaction, elevated SGPT, and the incidence of hair loss in the therapy group were lower than those of the control group (P < 0.05).

Sophora root injection combined with chemotherapy in treatment of breast cancer can enhance the effect of chemotherapy, reduce toxicity and side-effects, and improve quality of life (An, An, & Wu, 2012).

Lung cancer; Pleural Effusion

The therapeutic efficiency of Fufang Kushen Injection Liquid (FFKSIL), IL-2, α-IFN on lung cancer accompanied with malignancy pleural effusions, was observed.

One hundred and fifty patients with lung cancer, accompanied with pleural effusions, were randomly divided into treatment and control groups. The treatment group was divided into three groups: injected FFKSIL plus IL-2, FFKSIL plus α-tFN, and IL-2 plus α>-IFN, respectively. The control group was divided into three groups and injected FFKSIL, IL-2 and α>-IFN, respectively. The effective rate of FFKSIL, IL-2, and α-IFN in a combination was significantly superior to single pharmacotherapy. The effective rate of fufangkushen plus ct-IFN was highest. The effect of FFKSIL, IL-2, and α-IFN, in a combination, on lung cancer with pleural effusions was significantly better than single pharmacotherapy. Moreover, the effect of FFKSIL plus IL-2 or α-IFN had the greatest effect (Hu & Mei, 2012).

Gastric Cancer

Administration of FFKSIL significantly enhanced serum IgA, IgG, IgM, IL-2, IL-4 and IL-10 levels, decreased serum IL-6 and TNF-αlevels, lowered the levels of lipid peroxides and enhanced GSH levels and activities of GSH-dependent enzymes. Our results suggest that FFKSIL blocks experimental gastric carcinogenesis by protecting against carcinogen-induced oxidative damage and improving immunity activity (Zhou et al., 2012).

Colorectal Cancer; Chemotherapy

Eighty patients after colorectal cancer resection were randomly divided into two groups: 40 patients in the control group were treated with routine chemotherapy including 5-fluorouridine(5-FU), calcium folinate(CF) and oxaliplatin, and 40 patients in the experimental group were treated with the same chemotherapy regime combined with 20 mLád-1 compound Kushen injection, for 10d during chemotherapy. In the control group the numbers of CD3+,CD4+T cells,NK cells and CD4+/CD8+ ratio significantly declined relative to prior to chemotherapy (P < 0.05), while CD8+T lymphocyte number increased significantly. In the experimental group, there were no significant differences between the numbers of CD3+,CD4+,CD8+T cells ,NK cells, and CD4+/CD8+ ratio, before and after chemotherapy (P > 0.05).

Compound Kushen injection can improve the immunologic function of patients receiving chemotherapy after colorectal cancer resection (Chen, Yu, Yuan, & Yuan, 2009).

NSCLC; Chemotherapy

A total of 286 patients with advanced NSCLC were enrolled for study. The patients were treated with either compound Kushen injection in combination with NP (NVB + CBP) chemotherapy (vinorelbine and carboplatin, n = 144), or with NP (NVB + CBP) chemotherapy alone (n = 142). The following indicators were observed: levels of Hb, WBC, PLT and T cell subpopulations in blood, serum IgG level, short-term  efficacy, adverse effects and quality of life.

The gastrointestinal reactions and the myelosuppression in the combination chemotherapy group were alleviated when compared with the chemotherapy alone group, showing a significant difference (P < 0.05). CD (8)(+) cells were markedly declined in the combination chemotherapy group, and the CD (4)(+)/CD (8)(+) ratio showed an elevation trend in the chemotherapy alone group. The Karnofsky Performance Scale (KPS) scores and serum IgM and IgG levels were higher in the combination chemotherapy group than those in the chemotherapy alone group (P < 0.01 and P < 0.05).

The compound Kushen injection plus NP chemotherapy regimen showed better therapeutic effect, reduced adverse effects of chemotherapy and improved the quality of life in patients with stage III and IV NSCLC (Fan et al., 2010).

Lung Adenocarcinoma

Different concentrations of matrine injection could inhibit the growth of SPCA/I human lung adenocarcinoma cells. There was a positive correlation between the inhibition rate and the drug concentration. Different concentrations of matrine injection combined with anti-tumor drugs had a higher growth inhibition rate than anti-tumor drugs alone. Matrine injection has direct growth suppression effect on SPCA/I human lung adenocarcinoma cells and SS+ injection combined with anti-tumor drugs shows a significant synergistic effect on tumor cells (Zhu, Jiang, Lu, Guo, & Gan, 2008).

Liver Cancer

Fifty-seven patients with unresectable primary liver cancer were randomly divided into 2 groups. The treatment group with 27 cases was treated by TACE combined with composite Kushen injection, and the control group with 30 cases was treated by TACE alone. One, two, and three year survival rates of the treatment group were 67%, 48%, and 37% respectively, and those of control group were 53%, 37%, and 20% respectively. There were significant differences between both groups (P < 0.05).

Combined TACE with composite Kushen injection can increase the efficacy of patients with unresectable primary liver cancer (Wang & Cheng, 2009).

Chemotherapy

Ten RCTs were included in a meta-analysis, whose results suggest that compared with chemotherapy alone, the combination had a statistically significant benefit in healing efficacy and improving quality of life. As well,  the combination also had a statistically significant benefit in myelosuppression, white blood cell, hematoblast, liver function and in reducing the gastroenteric reaction, decreasing the of CD3, CD4, CD4/CD8, and NK cells (Huang et al., 2011).

Colorectal Cancer, NSCLC, Breast Cancer; Chemotherapy

Fufang kushen Injection might improve the efficacies of chemotherapy in patients with colorectal cancer, NSCLC and breast cancer.

The results of a meta-analysis of 33 studies of randomized controlled trials with a total of 2,897 patients demonstrated that the short-term efficacies in patients with colorectal cancer, NSCLC, and breast cancer receiving Fufangkushen Injection plus chemotherapy were significantly better than for those receiving chemotherapy alone. However the results for patients with gastric cancer on combined chemotherapy were not significantly different from those for patients on chemotherapy alone (Fang, Lin, & Fan, 2011).

References

An, A.J., An, G.W., & Wu, Y.C. (2012). Observation of compound recipe light yellow Sophora root injection combined with chemotherapy in treatment of 35 postoperative patients with breast cancer. Medical & Pharmaceutical Journal of Chinese People”s Liberation Army, 24(10), 43-46. doi: 10.3969/j.issn.2095-140X.2012.10.016.


Chen, G., Yu, B., Yuan, S.J., & Yuan, Q. (2009). Effects of compound Kushen injection on the immunologic function of patients after colorectal cancer resection. Evaluation and Analysis of Drug-Use in Hospitals of China, 2009(9), R735.3. doi: cnki:sun:yypf.0.2009-09-025.


Chen H, Zhang J, Luo J, et al. (2013). Anti-angiogenic effects of oxymatrine on pancreatic cancer by inhibition of the NF-κB-mediated VEGF signaling pathway. Oncol Rep, 30(2):589-95. doi: 10.3892/or.2013.2529.


Fan, C.X., Lin, C.L., Liang, L., Zhao, Y.Y., Liu, J., Cui, J., Yang, Q.M., Wang, Y.L., & Zhang, A.R. (2010). Enhancing effect of compound Kushen injection in combination with chemotherapy for patients with advanced non-small-cell lung cancer. Chinese Journal of Oncology, 32(4), 294-297.


Fang, L., Lin, N.M., Fan, Y. (2011). Short-term  efficacies of Fufangkushen Injection plus chemotherapy in patients with solid tumors: a meta-analysis of randomized trials. Zhonghua Yi Xue Za Zhi, 91(35):2476-81.


Hu, D.J., & Mei, X.D. (2012). Observing therapeutic efficiency of fufangkushen injection, IL-2, α-IFN on lung cancer accompanied with malignancy pleural effusions. Journal of Clinical Pulmonology, 17(10), 1844-1845.


Huang S, Fan W, Liu P, Tian J. (2011). Meta-analysis of compound matrine injection combined with cisplatin chemotherapy for advanced gastric cancer. Zhongguo Zhong Yao Za Zhi, 36(22):3198-202.


Kong, Q-Z., Huang, D-S., Huang, T. et al. (2003). Experimental study on inhibiting angiogenesis in mice S180 by injections of three traditional Chinese herbs. Chinese Journal of Hospital Pharmacy, 2003-11. doi: CNKI:SUN:ZGYZ.0.2003-11-002


Li T, Wong VK, Yi XQ, et al. (2010). Matrine induces cell anergy in human Jurkat T cells through modulation of mitogen-activated protein kinases and nuclear factor of activated T-cells signaling with concomitant up-regulation of anergy-associated genes expression. Biol Pharm Bull, 33(1):40-6.


Ling Q, Xu X, Wei X, et al. (2011). Oxymatrine induces human pancreatic cancer PANC-1 cells apoptosis via regulating expression of Bcl-2 and IAP families, and releasing of cytochrome c. J Exp Clin Cancer Res, 30:66. doi: 10.1186/1756-9966-30-66.


Qi, L., Zhang, J., Zhang, Z. (2013). Determination of four alkaloids in Compound Kushen Injection by high performance liquid chromatography with ionic liquid as mobile phase additive. Chinese Journal of Chromatography, 31(3): 249-253. doi: 10.3724/SP.J.1123.2012.10039.


Shi, B., & Xu, H. (2012). Effects of compound radix Sophorae flavescentis injection on proliferation, apoptosis and caspase-3 expression in adenoid cystic carcinoma ACC-2 cells. Chinese Pharmacological Bulletin, 5(10), 721-724.


Sun M, Cao H, Sun L, et al. (2012). Anti-tumor activities of kushen: literature review. Evid Based Complement Alternat Med, 2012:373219. doi: 10.1155/2012/373219.


Wang, H.M., & Cheng, X.M. (2009). Composite Ku Shen injection combined with hepatic artery embolism on unresectable primary liver cancer. Modern Journal of Integrated Traditional Chinese and Western Medicine, 18(2), 1334–1335.


Xia, G. (2013). Clinical observation of compound matrine injection combined with SP regimen in advanced gastric cancer. Journal of Liaoning Medical University, 2013(1), 37-38.


Yin, W.H., Sheng, J.W., Xia, H.M., Chen, J., Wu, Y.W., & Fan, H.Z. (2013). Study on the effect of compound matrine on the level of sIL-2R and IL-8 in peripheral blood cells of patients with rectal cancer to radiation. Global Traditional Chinese Medicine, 2013(2), 100-104.


Zhang Y, Sun S, Chen J, et al. (2013). Oxymatrine induces mitochondria dependent apoptosis in human osteosarcoma MNNG/HOS cells through inhibition of PI3K/Akt pathway. Tumor Biol.


Zhou, S-K., Zhang, R-L., Xu, Y-F., Bi, T-N. (2012) Anti-oxidant and Immunity Activities of Fufang Kushen Injection Liquid. Molecules 2012, 17(6), 6481-6490; doi:10.3390/molecules17066481


Zhu, M.Y., Jiang, Z.H., Lu, Y.W., Guo, Y., & Gan, J.J. (2008). Matrine and anti-tumor drugs in inhibiting the growth of human lung cancer cell line. Journal of Chinese Integrative Medicine, 6(2), 163-165. doi: 10.3736/jcim20080211.

Silibinin

Cancer:
Lung, leukemia, colorectal, thyroid, breast, bladder

Action: Anti-angiogenesis, EMT, cell-cycle arrest

Cell-cycle Arrest, Colon Cancer

Silibinin, an active constituent of milk thistle (Silybum marianum [(L.) Gaertn.]), has been reported to inhibit proliferation and induce cell-cycle arrest of human colon cancer cells, Fet, Geo, and HCT116 (Hogan et al., 2007). Silibinin Up-regulates the expression of cyclin-dependent kinase inhibitors and induces cell-cycle arrest and apoptosis in human colon carcinoma HT-29 cells (Agarwal et al., 2003). Also in HT-29 cells, treatment with beta-escin, a principal component of horse chestnut, tinduces growth arrest at the G1-S phase together with an induction of Cip1/p21 and an associated reduction in the phosphorylation of retinoblastoma protein (Patlolla et al., 2006).

Lung Cancer

Silibinin also has anti-angiogenic effects on lung adenocarcinomas in vitro, as it strongly decreased both tumor number and tumor size (an anti-tumor effect that correlates with reduced anti-angiogenic activity) (Tyagi et al., 2009). Further, silibinin inhibits mouse lung tumorigenesis in vivo, in part by targeting tumor microenvironment. Tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) can be pro- or anti-tumorigenic, but in lung cancer cell lines they induce pro-inflammatory enzymes cyclooxygenase 2 (COX2) and inducible nitric oxide synthase (iNOS). Accordingly, the mechanism of silibinin action was examined on TNF-α + IFN-γ (hereafter referred as cytokine mixture) elicited signaling in tumor-derived mouse lung epithelial LM2 cells.

Both signal transducers and activators of the transcription (STAT)3 (tyr705 and ser727) and STAT1 (tyr701) were activated within 15 min of cytokine mixture exposure, while STAT1 (ser727) activated after 3 h. Cytokine mixture also activated Erk1/2 and caused an increase in both COX2 and iNOS levels. Pre-treatment of cells with a MEK, NF-κB, and/or epidermal growth factor receptor (EGFR) inhibitor inhibited cytokine mixture-induced activation of Erk1/2, NF-κB, or EGFR, respectively, and strongly decreased phosphorylation of STAT3 and STAT1 and expression of COX2 and iNOS.

Together, the results show that STAT3 and STAT1 could be valuable chemo-preventive and therapeutic targets within the lung tumor microenvironment in addition to being targets within the tumor itself, and that silibinin inhibit their activation as a plausible mechanism of its efficacy against lung cancer (Tyagi et al., 2011).

Leukemia

Silibinin also affects cellular differentiation in the human promyelocytic leukemia HL-60 cell culture system. Treatment of HL-60 cells with silibinin inhibited cellular proliferation and induced cellular differentiation in a dose-dependent manner.

Silibinin enhanced protein kinase C (PKC) activity and increased protein levels of both PKCα and PKCβ in 1,25-(OH)2D3-treated HL-60 cells. PKC and extracellular signal-regulated kinase (ERK) inhibitors significantly inhibited HL-60 cell differentiation induced by silibinin alone or in combination with 1,25-(OH)2D3, indicating that PKC and ERK may be involved in silibinin-induced HL-60 cell differentiation (Kang et al., 2001).

Thyroid Cancer, Breast Cancer

Silibinin inhibits TPA-induced cell migration and MMP-9 expression in thyroid and breast cancer cells. Matrix metalloproteinases (MMPs) play an important role in cancer metastasis, cell migration and invasion. The effects of silibinin were investigated on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cell migration and MMP-9 expression in thyroid and breast cancer cells. These results revealed that the levels of MMP-9 mRNA and protein expression were significantly increased by TPA but not MMP-2 in TPC-1 and MCF7 cells.

TPA-induced phosphorylation of MEK and ERK was also inhibited by silibinin. Taken together, these results suggest that silibinin suppresses TPA-induced cell migration and MMP-9 expression through the MEK/ERK-dependent pathway in thyroid and breast cancer cells (Oh et al., 2013).

Bladder Cancer

Silibinin induced apoptosis and inhibited proliferation of bladder cancer cells and metastasis. In the present study, Wu et al. (2013) utilized a novel highly metastatic T24-L cell model, and found that silibinin treatment not only resulted in the suppression of cell migration and invasion in vitro, but also decreased bladder cancer lung metastasis and prolonged animal survival in vivo. Inactivation of β-catenin/ZEB1 signaling by silibinin leads to dual-block of EMT and stemness.

Lung Cancer, EMT

Silibinin formulation might facilitate the design of clinical trials to test the administration of silibinin meglumine-containing injections, granules, or beverages in combination with EGFR TKIs in patients with EGFR-mutated NSCLC. Silibinin meglumine notably decreased the overall volumes of NSCLC tumors as efficiently as did the EGFR tyrosine kinase inhibitor (TKI) gefitinib. Concurrent treatment with silibinin meglumine impeded the regrowth of gefitinib-unresponsive tumors, resulting in drastic tumor growth prevention.

Because the epithelial-to-mesenchymal transition (EMT) is required by a multiplicity of mechanisms of resistance to EGFR TKIs, we evaluated the ability of silibinin meglumine to impede the EMT in vitro and in vivo. Silibinin-meglumine efficiently prevented the loss of markers associated with a polarized epithelial phenotype as well as the de novo synthesis of proteins associated with the mesenchymal morphology of transitioning cells (Cuf` et al., 2013).

Breast cancer

Myeloid-derived suppressor cells (MDSC)s increase in blood and accumulate in the tumor microenvironment of tumor-bearing animals, contributing to immune suppression in cancer. Silibinin, a natural flavonoid from the seeds of milk thistle, has been developed as an anti-inflammatory agent and supportive care agent to reduce the toxicity of cancer chemotherapy. The goals of this study were to evaluate the effect of silibinin on MDSCs in tumor-bearing mice and antitumor activity of silibinin in a mouse model of breast cancer. 4T1 luciferase-transfected mammary carcinoma cells were injected into in the mammary fat pad female BALB/c mice, and female CB17-Prkdc Scid/J mice. Silibinin treatment started on day 4 or day 14 after tumor inoculation continued every other day.

Tumor growth was monitored by bioluminescent imaging (BLI) measuring total photon flux. Flow cytometry measured total leukocytes, CD11b+ Gr-1+ MDSC, and T cells in the blood and tumors of tumor-bearing mice. The effects of silibinin on 4T1 cell viability in vitro were measured by BLI. Treatment with silibinin increased overall survival in mice harboring tumors derived from the 4T1-luciferase breast cancer cell line, and reduced tumor volumes and numbers of CD11b+Gr-1+ MDSCs in the blood and tumor, and increased the content of T cells in the tumor microenvironment.

Silibinin failed to inhibit tumor growth in immunocompromised severe combined immunodeficiency mice, supporting the hypothesis that anticancer effect of silibinin is immune-mediated. The antitumor activity of silibinin requires an intact host immune system and is associated with decreased accumulation of blood and tumor-associated MDSCs.

References

 

Agarwal C, Singh RP, Dhanalakshmi S, et al. (2003). Silibinin Up-regulates the expression of cyclin-dependent kinase inhibitors and causes cell-cycle arrest and apoptosis in human colon carcinoma HT-29 cells. Oncogene, 22:8271–8282.

 

Cufí S, Bonavia R, Vazquez-Martin A, Corominas-Faja B, et al. (2013). Silibinin meglumine, a water-soluble form of milk thistle silymarin, is an orally active anti-cancer agent that impedes the epithelial-to-mesenchymal transition (EMT) in EGFR-mutant non-small-cell lung carcinoma cells. Food Chem Toxicol, 60:360-8. doi: 10.1016/j.fct.2013.07.063.

Hogan FS, Krishnegowda NK, Mikhailova M, Kahlenberg MS. (2007). Flavonoid, silibinin, inhibits proliferation and promotes cell-cycle arrest of human colon cancer. J Surg Res, 143:58–65.

Kang SN, Lee MH, Kim KM, Cho D, Kim TS. (2001). Induction of human promyelocytic leukemia HL-60 cell differentiation into monocytes by silibinin: involvement of protein kinase C. Biochemical Pharmacology, 61(12):1487–1495

Oh SJ, Jung SP, Han J, et al. (2013). Silibinin inhibits TPA-induced cell migration and MMP-9 expression in thyroid and breast cancer cells. Oncol Rep, 29(4):1343-8. doi: 10.3892/or.2013.2252.

Patlolla JM, Raju J, Swamy MV, Rao CV. (2006). Beta-escin inhibits colonic aberrant crypt foci formation in rats and regulates the Cell-cycle growth by inducing p21(waf1/cip1) in colon cancer cells. Mol Cancer Ther, 5:1459–1466.

Tyagi A, Singh RP, Ramasamy K, et al. (2009). Growth Inhibition and Regression of Lung Tumors by Silibinin: Modulation of Angiogenesis by Macrophage-Associated Cytokines and Nuclear Factor-κ B and Signal Transducers and Activators of Transcription 3. Cancer Prev Res, 2(1):74-83

Tyagi A, Agarwal C, Dwyer-Nield LD, et al. (2011). Silibinin modulates TNF‐α and IFN ‐γ mediated signaling to regulate COX2 and iNOS expression in tumorigenic mouse lung epithelial LM2 cells. Molecular Carcinogenesis. doi: 10.1002/mc.20851.

Wu K, Ning Z, Zeng J, et al. (2013). Silibinin inhibits β -catenin/ZEB1 signaling and suppresses bladder cancer metastasis via dual-blocking epithelial-mesenchymal transition and stemness. Cell Signal, 25(12):2625-2633. doi: 10.1016/j.cellsig.2013.08.028.

Forghani P, Khorramizadeh MR & Waller EK. (2014) Silibinin inhibits accumulation of myeloid-derived suppressor cells and tumor growth of murine breast cancer. Cancer Medicine. Volume 3, Issue 2, pages 215–224, April 2014 DOI: 10.1002/cam4.186

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.

Torilin

Cancer: none noted

Action: Anti-angiogenesis

Torilin is a sesquiterpene compound purified from fruits of Torilis japonica (Umbelliferae).

Torilin decreased both neovascularization and basic fibroblast growth factor-induced vessel formation. Torilin also reduced the proliferation and tube formation of human umbilical vein endothelial cells. In addition, the concentrated conditioned media obtained from torilin-treated HepG2 human hepatoblastoma cells blocked the angiogenic activation of torilin-untreated concentrated conditioned media, indicating that torilin may have an inhibitory effect on tumor-induced angiogenesis.

Torilin significantly down-regulated the expression of hypoxia-inducible vascular endothelial growth factor and insulin-like growth factor-II. Taken together, our data suggest that torilin may be a strong angiogenic inhibitor with the ability to decrease tube formation of vascular endothelial cells and to reduce expression of angiogenic factors of tumor cells.

Reference

Kim MS, Lee YM, Moon EJ, et al. (2000). Anti-angiogenic activity of torilin, a sesquiterpene compound isolated from Torilis japonica. Int J Cancer, 87(2):269-75.