Category Archives: Chapter 7 Isolates and Cancer Research

Paeoniflorin

Cancer: Hepatocellular carcinoma, colorectal, liver

Action: Radio-protective, ameliorated myelosuppression, MDR

Radio-protective

The radio-protective effect of paeoniflorin (PF), a main bioactive component in the traditional Chinese herb peony, on irradiated thymocytes and the possible mechanisms of protection have been investigated. Ionizing radiation can induce DNA damage and cell death by generating reactive oxygen species (ROS).

It was found 60Co γ-ray irradiation increased cell death and DNA fragmentation in a dose-dependent manner while increasing intracellular ROS. Pre-treatment of thymocytes with PF (50–200 µg/ml) reversed this tendency and attenuated irradiation-induced ROS generation. Hydroxyl-scavenging action of PF in vitro was detected through electron spin resonance assay. Several anti-apoptotic characteristics of PF, including the ability to diminish cytosolic Ca2+ concentration, inhibit caspase-3 activation, and up-regulate Bcl-2 and down-regulate Bax in 4 Gy-irradiated thymocytes, were determined.

Extracellular regulated kinase (ERK), c-Jun NH2-terminal kinase (JNK), and p38 kinase, were activated by 4 Gy irradiation, with their activation partly blocked by pre-treatment of cells with PF. The presence of ERK inhibitor PD98059, JNK inhibitor SP600125 and p38 inhibitor SB203580 decreased cell death in 4 Gy-irradiated thymocytes. These results suggest PF protects thymocytes against irradiation-induced cell damage by scavenging ROS and attenuating the activation of the mitogen-activated protein kinases (Li et al., 2007).

Liver Cancer

Prostaglandin E2 (PGE2) has been shown to play an important role in tumor development and progression. PGE2 mediates its biological activity by binding any one of four prostanoid receptors (EP1 through EP4). Paeoniflorin, a monoterpene glycoside, significantly inhibited the proliferation of HepG2 and SMMC-7721 cells stimulated by butaprost at multiple time points (24, 48, and 72 hours). Paeoniflorin induced apoptosis in HepG2 and SMMC-7721 cells, which was quantified by annexin-V and propidium iodide staining. Our results indicate that the expression of the EP2 receptor and Bcl-2 was significantly increased, whereas that of Bax and cleaved caspase-3 was decreased in HepG2 and SMMC-7721 cells.

Paeoniflorin, which may be a promising agent in the treatment of liver cancer, induced apoptosis in hepatocellular carcinoma cells by down-regulating EP2 expression and also increased the Bax-to-Bcl-2 ratio, thus up-regulating the activation of caspase-3 (Hu et al., 2013).

Colorectal Cancer

Results showed that positive cells of Proliferating Cell Nuclear Antigen (PCNA) in paeoniflorin (PF) and docetaxel-treated group was decreased to 30% and 15% respectively, compared with control group of tumors. But apoptosis cells in docetaxel treated groups studied by TUNEL is increased to 40 ± 1.2% and 30 ± 1.5% respectively, compared with 24 ± 2.3% in negative control. Furthermore, the efficiency of tumor-bearing mice treated by PF was superior to docetaxel in vivo. Overall, PF may be an effective chemo-preventive agent against colorectal cancer HT29 (Wang et al., 2012).

Ameliorates Myelosuppression

The administration of paeoniflorin and albiflorin (CPA) extracted from Paeonia radix, significantly ameliorated myelosuppression in all cases. For the X-ray irradiated mice and the chemotherapy treated mice and rabbits, high dosages of CPA resulted in the recovery of, respectively, 94.4%, 95.3% and 97.7% of hemoglobin content; 67.7%, 92.0% and 94.3% of platelet numbers; 26.8%, 137.1% and 107.3% of white blood cell counts; as well as a reversal in the reduction of peripheral differential white blood cell counts.

There was also a recovery of 50.9%, 146.1% and 92.3%, respectively, in the animals' relative spleen weight. Additionally, a recovery of 35.7% and 87.2% respectively in the number of bone marrow nucleated cells was observed in the radio- and chemo -therapy-treated mice. Bone marrow white blood cell counts also resumed to normal levels (Xu et al., 2011).

MDR

Studies have shown that NF-κB activation may play an essential role in the development of chemotherapy resistance in carcinoma cells. Paeonißorin, a principal bioactive component of the root of Paeonia lactißora, has been reported to exhibit various pharmacological effects. In the present study, Fanh et al. (2012) reported for the first time that paeoniflorin at non-toxic concentrations may effectively modulate multi-drug resistance (MDR) of the human gastric cancer cell line SGC7901/vincristine (VCR) via the inhibition of NF-κB activation and, at least partly, by subsequently down-regulating its target genes MDR1, BCL-XL and BCL-2.

References

Fang S, Zhu W, Zhang Y, Shu Y, Liu P. (2012). Paeoniflorin modulates Multi-drug resistance of a human gastric cancer cell line via the inhibition of NF- κB activation. Mol Med Rep, 5(2):351-6. doi: 10.3892/mmr.2011.652.


Hu S, Sun W, Wei W, et al. (2013). Involvement of the prostaglandin E receptor EP2 in paeoniflorin-induced human hepatoma cell apoptosis. Anti-cancer Drugs, 24(2):140-9. doi: 10.1097/CAD.0b013e32835a4dac.


Li CR, Zhou Z, Zhu D, et al. (2007). Protective effect of paeoniflorin on irradiation-induced cell damage involved in modulation of reactive oxygen species and the mitogen-activated protein kinases. The International Journal of Biochemistry & Cell Biology, 39(2):426–438


Wang H, Zhou H, Wang CX, et al. (2012). Paeoniflorin inhibits growth of human colorectal carcinoma HT 29 cells in vitro and in vivo. Food Chem Toxicol, 50(5):1560-7. doi: 10.1016/j.fct.2012.01.035.


Xu W, Zhou L, Ma X, et al. (2011). Therapeutic effects of combination of paeoniflorin and albiflorin from Paeonia radix on radiation and chemotherapy-induced myelosuppression in mice and rabbits. Asian Pac J Cancer Prev, 12(8):2031-7.

Paenol

Cancer: Gastric

Action: Attenuates nephrotoxicity, anti-inflammatory, anti-oxidant, inhibits TNF- α , induces apoptosis, COX-2 down-regulation

Inhibits TNF- α

Moutan Cortex, the root bark of Paeonia suffruticosa Andrews, has been used extensively as a traditional medicine for treatment of various diseases such as atherosclerosis, infection, and inflammation. Previous studies have revealed that the extracts of Moutan Cortex can inhibit nitric oxide and TNF- α in activated mouse peritoneal macrophages (Chung et al., 2007).

A variety of compounds including paeonoside, paeonolide, apiopaeonoside, paeoniflorin, oxypaeoniflorin, benzoyloxypaeoniflorin, benzoylpaeoniflorin, paeonol, and sugars have been identified in Moutan Cortex (Chen et al., 2006).

Attenuates Nephrotoxicity

Paeonol, a major compound of Moutan Cortex, has been found to attenuate cisplatin-induced nephrotoxicity in mice. Cisplatin is an effective chemotherapeutic agent that is used for the treatment of a variety of cancers; however, its nephrotoxicity limits the use of this drug.

Balb/c mice (6 to 8  w of age, weighing 20 to 25  g) were administered with Moutan Cortex (300  mg/kg) or paeonol (20 mg/kg) once a day. At day 4, mice received cisplatin (30, 20, or 10   mg/kg) intraperitoneally.

The paeonol-treated group showed marked attenuation of serum creatine and blood urea nitrogen levels as well as reduced levels of pro-inflammatory cytokines and nitric oxide when compared to the control group. In addition, the paeonol-treated group showed prolonged survival and marked attenuation of renal tissue injury. Taken together, these results demonstrated that paeonol can prevent the renal toxic effects of cisplatin (Lee et al., 2013).

Paeonol, a major phenolic component of Moutan Cortex, has various biological activities such as anti-aggregatory, anti-oxidant, anxiolytic-like, and anti-inflammatory functions (Ishiguro et al., 2006). In this study, paeonol treatment significantly reduced the elevated levels of serum creatinine and BUN. In addition, the role of pro-inflammatory cytokines in cisplatin-induced acute renal failure has been well documented (Faubel et al., 2007; Ramesh & Reeves, 2002), and elevation of the pro-inflammatory cytokines TNF-α and IL-1β as well as that of IL-6 has been demonstrated in humans with acute renal failure (Simmons et al., 2004).

Apoptosis-inducing & Gastric Cancer

Paeonol has significantly growth-inhibitory and apoptosis-inducing effects in gastric cancer cells both in vitro and in vivo. In vitro, paeonol caused dose-dependent inhibition on cell proliferation and induced apoptosis. Cell cycle analysis revealed a decreased proportion of cells in G0/G1 phase, with arrest at S. Paeonol treatment in gastric cancer cell line MFC and SGC-790 cells significantly reduced the expression of Bcl-2 and increased the expression of Bax in a concentration-related manner. Administration of paeonol to MFC tumor-bearing mice significantly lowered the tumor growth and caused tumor regression (Li et al., 2010).

COX-2 Down-regulation

One of the apoptotic mechanisms of paeonol is down-regulation of COX-2. p27 is up-regulated simultaneously and plays an important part in controlling cell proliferation and is a crucial factor in the Fas/FasL apoptosis pathway. Cell proliferation was inhibited by different concentrations of paeonol. By immunocytochemical staining, Ye et al. (2009) found that HT-29 cells treated with paeonol (0.024-1.504 mmol/L) reflected reduced expression of COX-2 and increased expression of p27 in a dose-dependent manner. RT-PCR showed that paeonol down-regulated COX-2 and up-regulated p27 in a dose- and time-dependent manner in HT-29 cells.

References

Chen G, Zhang L, Zhu Y. (2006). Determination of glycosides and sugars in moutan cortex by capillary electrophoresis with electrochemical detection. Journal of Pharmaceutical and Biomedical Analysis, 41(1):129–134.


Chung HS, M. Kang, C. Cho et al. (2007). Inhibition of nitric oxide and tumor necrosis factor-alpha by moutan cortex in activated mouse peritoneal macrophages. Biological and Pharmaceutical Bulletin, 30(5):912–916.


Faubel F, Lewis EC, Reznikov L et al. (2007). Cisplatin-induced acute renal failure is associated with an increase in the cytokines interleukin (IL)-1 β , IL-18, IL-6, and neutrophil infiltration in the kidney. Journal of Pharmacology and Experimental Therapeutics, 322(1):8–15.


Ishiguro K, Ando T, Maeda O et al. (2006). Paeonol attenuates TNBS-induced colitis by inhibiting NF- κ B and STAT1 transactivation. Toxicology and Applied Pharmacology, 217(1):35–42.


Lee HJ, Lee GY, Kim Hs, Bae Hs. (2013). Paeonol, a Major Compound of Moutan Cortex, Attenuates Cisplatin-Induced Nephrotoxicity in Mice. Evidence-Based Complementary and Alternative Medicine, 2013(2013), http://dx.doi.org/10.1155/2013/310989


Li N, Fan LL, Sun GP, et al. (2010). Paeonol inhibits tumor growth in gastric cancer in vitro and in vivo. World J Gastroenterol., 16(35):4483-90.


Ramesh G, Reeves wb. (2002). TNF- α mediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity. Journal of Clinical Investigation, 110(6):835–842.


Simmons EM, Himmelfarb j, Sezer MT et al. (2004). Plasma cytokine levels predict mortality in patients with acute renal failure. Kidney International, 65(4):1357–1365.


Ye JM, Deng T, Zhang JB. (2009) Influence of paeonol on expression of COX-2 and p27 in HT-29 cells. World J Gastroenterol, 15(35):4410-4.

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.

Oleanolic Acid (OA)

Cancer:
Pancreatic, hepatocellular carcinoma, prostate, lung, gastric, breast

Action: Radio-sensitizer, pro-apoptotic with 5-FU

Oleanolic acid (OA), a pentacyclic triterpenoid isolated from several plants, including Rosa woodsii (Lindl.), Prosopis glandulosa (Torr.), Phoradendron juniperinum (Engelm. ex A. Gray), Syzygium claviflorum (Roxburgh), Hyptis capitata (Jacq.) and Ternstromia gymnanthera (L.) exhibits potential anti-tumor activity against many tumor cell lines. Mistletoe contains water-insoluble triterpenoids, mainly oleanolic acid, that have anti-tumorigenic effects (StrŸh et al., 2013).

Pancreatic Cancer

Results of a study by Wei et al. (2012) showed that the proliferation of Panc-28 cells was inhibited by OA in a concentration-dependent manner, with an IC50 (The half maximal inhibitory concentration) value of 46.35 µg ml−1. The study also showed that OA could induce remarkable apoptosis and revealed that OA could induce Reactive Oxygen Species (ROS) generation, mitochondrial depolarization, release of cytochrome C, lysosomal membrane permeabilization and leakage of cathepin B. Further study confirmed that ROS scavenger vitamin C could reverse the apoptosis induced by OA in Panc-28 cells.

These results provide evidence that OA arrests the cell-cycle and induces apoptosis, possibly via ROS-mediated mitochondrial and a lysosomal pathway in Panc-28 cell.

The effects of the combination of OA and 5-fluorouracil (5-FU) on Panc-28 human pancreatic cells showed that combined use synergistically potentiated cell death effects on these cells, and that the pro-apoptotic effects were also increased. The expression of apoptosis related proteins was also affected in cells treated with the combination of OA and 5-FU, including activation of caspases-3 and the expression of Bcl-2/Bax, survivin and NF-κB (Wei et al., 2012).

Radio-sensitizer

The combined treatment of radiation with OA significantly decreased the clonogenic growth of tumor cells and enhanced the numbers of intracellular MN compared to irradiation alone. Furthermore, it was found that the synthesis of cellular GSH was inhibited concomitantly with the down-regulation of γ-GCS activity. Therefore, the utilization of OA as a radio-sensitizing agent for irradiation-inducing cell death offers a potential therapeutic approach to treat cancer (Wang et al., 2013).

Prostate Cancer, Lung Cancer, Gastric Cancer, Breast Cancer

Twelve derivatives of oleanolic acid (OA) have been synthesized and evaluated for their inhibitory activities against the growth of prostate PC3, breast MCF-7, lung A549, and gastric BGC-823 cancer cells by MTT assays. Within these series of derivatives, compound 17 exhibited the most potent cytotoxicity against PC3 cell line (IC50=0.39 µM) and compound 28 displayed the best activity against A549 cell line (IC50=0.22 µM). SAR analysis indicates that H-donor substitution at C-3 position of oleanolic acid may be advantageous for improvement of cytotoxicity against PC3, A549 and MCF-7 cell lines (Hao et al., 2013).

Hepatocellular Carcinoma

OA induced G2/M cell-cycle arrest through p21-mediated down-regulation of cyclin B1/cdc2. Cyclooxygenase-2 (COX-2) and p53 were involved in OA-exerted effect, and extracellular signal-regulated kinase-p53 signaling played a central role in OA-activated cascades responsible for apoptosis and cell-cycle arrest. OA demonstrated significant anti-tumor activities in hepatocellular carcinoma (HCC) in vivo and in vitro models. These data provide new insights into the mechanisms underlying the anti-tumor effect of OA (Wang et al., 2013).

References

Hao J, Liu J, Wen X, Sun H. (2013). Synthesis and cytotoxicity evaluation of oleanolic acid derivatives. Bioorg Med Chem Lett, 23(7):2074-7. doi: 10.1016/j.bmcl.2013.01.129.


StrŸh CM, JŠger S, Kersten A, et al. (2013). Triterpenoids amplify anti-tumoral effects of mistletoe extracts on murine B16.f10 melanoma in vivo. PLoS One, 8(4):e62168. doi: 10.1371/journal.pone.0062168.


Wang J, Yu M, Xiao L, et al. (2013). Radio-sensitizing effect of oleanolic acid on tumor cells through the inhibition of GSH synthesis in vitro. Oncol Rep, 30(2):917-24. doi: 10.3892/or.2013.2510.


Wang X, Bai H, Zhang X, et al. (2013). Inhibitory effect of oleanolic acid on hepatocellular carcinoma via ERK-p53-mediated cell-cycle arrest and mitochondrial-dependent apoptosis. Carcinogenesis, 34(6):1323-30. doi: 10.1093/carcin/bgt058.


Wei JT, Liu M, Liuz, et al. (2012). Oleanolic acid arrests cell-cycle and induces apoptosis via ROS-mediated mitochondrial depolarization and lysosomal membrane permeabilization in human pancreatic cancer cells. Journal of Applied Toxicology, 33(8):756–765. doi: 10.1002/jat.2725


Wei J, Liu H, Liu M, et al. (2012). Oleanolic acid potentiates the anti-tumor activity of 5-fluorouracil in pancreatic cancer cells. Oncol Rep, 28(4):1339-45. doi: 10.3892/or.2012.1921.

Oleandrin

Cancer: Prostate, glioma, melanoma

Action: Radio-sensitizer

Anvirzel is an extract of Nerium oleander (L.) currently undergoing, as Anvirzelª Phase I clinical evaluation as a potential treatment for cancer. Two of the active components of Anvirzel are the cardiac glycosides, oleandrin and oleandrigenin.

Prostate Cancer

In continuing research on the anti-tumor activity of this novel plant extract, the relative abilities of oleandrin and oleandrigenin to inhibit FGF-2 export from two human prostate cancer cell lines, DU145 and PC3, were examined. An ELISA assay was utilized to determine the FGF-2 concentration in the cell culture medium before and after exposure to cardiac glycosides or the parent extract material Anvirzel.

Studies also were conducted with Anvirzel (a hot water extract of Nerium oleander, known as Anvirzelª) and ouabain (found in the ripe seeds of African plants Strophanthus gratus). Oleandrin (0.1 ng/mL) produced a 45.7% inhibition of FGF-2 release from PC3 cells and a 49.9% inhibition from DU145 cells. Non-cytotoxic concentrations (100 ng/mL) of Anvirzel produced a 51.9% and 30.8% inhibition of FGF-2 release, respectively, in the two cell lines. These results demonstrate that Anvirzel, like oleandrin, inhibited FGF-2 export in vitro from PC3 and DU145 prostate cancer cells in a concentration- and time-dependent fashion and may, therefore, contribute to the anti-tumor activity of this novel treatment for cancer (Smith et al., 2001).

Radio-sensitizers; Prostate Cancer

In the present study Nasu et al. (2002) explored the relative radio-sensitization potential of oleandrin, a cardiac glycoside contained within the plant extract known as Anvirzelª. The data show that oleandrin produces an enhancement of sensitivity of PC-3 human prostate cells to radiation; at a cell survival of 0.1, the enhancement factor was 1.32. The magnitude of radio-sensitization depended on duration of exposure of cells to drug prior to radiation treatment.

While a radio-sensitizing effect of oleandrin was evident with only 1 hour of cell exposure to drug, the effect greatly increased with 24 hours of oleandrin pre-treatment.

Activation was greatest when cells were exposed simultaneously to oleandrin and radiation. Inhibition of caspase-3 activation with Z-DEVD-FMK abrogated the oleandrin-induced enhancement of radiation response suggesting that both oleandrin and radiation share a caspase-3 dependent mechanism of apoptosis in the PC-3 cell line.

Glioma, Melanoma

Twelve human tumor cell lines were chosen to examine determinants of human tumor cell sensitivity to cardiac glycosides. In vitro cell culture models of human glioma HF U251 and U251 cells as well as human parental and modified melanoma BRO cells were also included in these studies. Cardiac glycosides such as oleandrin, ouabain and bufalin increased expression of Na+, K+ -ATPase alpha 1 and therefore total Na+, K+ -ATPase activity, which is associated with increased cellular levels of glutathione. Additionally, an increased colony-forming ability was noted in cells with high levels of Na+, K+ -ATPase alpha 1 expression, suggesting that Na+, K+ -ATPase alpha 1 isoform may be actively involved in tumor growth and cell survival (Lin, Ho, & Newman, 2010)

References

Lin Y, Ho DH, Newman RA. (2010). Human tumor cell sensitivity to oleandrin is dependent on relative expression of Na+, K+ -ATPase subunitst. J Exp Ther Oncol, 8(4):271-86.


Nasu S, Milas L, Kawabe S, Raju U, Newman R. (2002). Enhancement of radiotherapy by oleandrin is a caspase-3 dependent process. Cancer Letters, 185(2):145–151. doi:10.1016/S0304-3835(02)00263-X


Smith JA, Madden T, Vijjeswarapu M, Newman RA. (2001). Inhibition of export of fibroblast growth factor-2 (FGF-2) from the prostate cancer cell lines PC3 and DU145 by anvirzel and its cardiac glycoside component, oleandrin. Biochemical Pharmacology, 62(4):469-472. doi:10.1016/S0006-2952(01)00690-6.

Norcantharidin (NCTD)

Cancer: Colorectal., CSCs, breast

Action: Anti-metastatic, MDR

Norcantharidin is a metastatic inhibitor derived from cantharidin, which is found in many species of blister beetles, including Mylabris phalerata (Pall.) and Lytta vesicatoria (Linnaeus).

Norcantharidin (NCTD) is a small-molecule metastatic inhibitor without renal toxicity derived from a renal toxic compound cantharidin, which is found in blister beetles (Mylabris phalerata Pall.), commonly used in traditional Chinese medicine.

Colorectal Cancer; Anti-metastatic

The aim of this study was to clarify the transcriptional regulation of MMP-9 gene by NCTD in colorectal cancer CT-26 cells. NCTD not only down-regulated MMP-9 mRNA and protein expression, but also inhibited gelatinase activity in a concentration- and time-dependent manner. Evidence by electrophoretic mobility shift assay demonstrated that NCTD inhibited the DNA-binding activity of Sp1. In addition, the increase effect of NF-kappaB-luciferase activity by NCTD may include the up-expression of nuclear STAT1 and result in competitive suppression of NF-kappaB-binding activity in MMP-9 promoter. In conclusion, the metastasis inhibitor NCTD down-regulates MMP-9 expression by inhibiting Sp1 transcriptional activity in colorectal cancer CT26 cells (Chen et al., 2009).

MDR; Cancer Stem Cells

Hsieh et al. (2013) investigated the modulation of self-renewal pathways and MDR in CSCs by NCTD. They suggest that using NCTD to develop more effective strategies for cancer treatment to reduce resistance and recurrence.

Breast Cancer

Cantharidin and norcantharidin induced apoptosis and repressed MCF-7 cell growth, adhesion and migration. They repressed MCF-7 cell adhesion to platelets through down-regulation of α2 integrin, an adhesion molecule present on the surface of cancer cells. The repression of α2 integrin expression was found to be executed through the protein kinase C pathway, the activation of which could have been due to PP2A inhibition (Shou et al. 2013).

References

Chen YJ, Chang WM, Liu YW, et al. (2009). A small-molecule metastasis inhibitor, norcantharidin, downregulates matrix metalloproteinase-9 expression by inhibiting Sp1 transcriptional activity in colorectal cancer cells. Chem Biol Interact., 181(3):440-6.


Hsieh CH, Chao KS, Liao HF, Chen YJ. (2013). Norcantharidin, Derivative of Cantharidin, for Cancer Stem Cells. Evid Based Complement Alternat Med, 2013;2013:838651.


Shou LM, Zhang QY, Li W, et al. (2013). Cantharidin and norcantharidin inhibit the ability of MCF-7 cells to adhere to platelets via protein kinase C pathway-dependent down-regulation of α 2 integrin. Oncol Rep. doi: 10.3892/or.2013.2601.

Nomilin

Cancer: Melanoma, breast cancer

Action: Anti-angiogenic

Nomilin is a triterpenoid present in common edible citrus fruits (Citrus grandis [(L.) Osb.], Citrus unshiu [(Swingle) Marcow.] and Citrus reticulata (Blanco)) with putative anti-cancer properties.

Melanoma

Nomilin possess anti-metastatic action, inducing metastasis in C57BL/6 mice through the lateral tail vein using highly metastatic B16F-10 melanoma cells. Administration of nomilin inhibited tumor nodule formation in the lungs (68%) and markedly increased the survival rate of the metastatic tumor–bearing animals. Nomilin showed an inhibition of tumor cell invasion and activation of matrix metalloproteinases. Treatment with nomilin induced apoptotic response.

Nomilin treatment also exhibited a down-regulated Bcl-2 and cyclin-D1 expression and up-regulated p53, Bax, caspase-9, caspase-3, p21, and p27 gene expression in B16F-10 cells. Pro-inflammatory cytokine production and gene expression were found to be down-regulated in nomilin-treated cells. The study also reveals that nomilin could inhibit the activation and nuclear translocation of anti-apoptotic transcription factors such as nuclear factor (NF)-κB, CREB, and ATF-2 in B16F-10 cells (Pratheeshkumar et al., 2011).

Breast Cancer; ER+

A panel of 9 purified limonoids, including limonin, nomilin, obacunone, limonexic acid (LNA), isolimonexic acid (ILNA), nomilinic acid glucoside (NAG), deacetyl nomilinic acid glucoside (DNAG), limonin glucoside (LG) and obacunone glucoside (OG) as well as 4 modified compounds such as limonin methoxime (LM), limonin oxime (LO), defuran limonin (DL), and defuran nomilin (DN), were screened for their cytotoxicity on estrogen receptor (ER)-positive (MCF-7) or ER-negative (MDA-MB-231) human breast cancer cells. Findings indicated that the citrus limonoids may have potential for the prevention of estrogen-responsive breast cancer (MCF-7) via caspase-7 dependent pathways (Lin et al., 2013).

Blocks Angoigenesis

Nomilin significantly inhibited tumor-directed capillary formation. Serum pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and GM-CSF and also serum NO levels were significantly reduced by the treatment of nomilin. Administration of nomilin significantly reduced the serum level of VEGF, a pro-angiogenic factor and increased the anti-angiogenic factors IL-2 and TIMP-1. Nomilin significantly retarded endothelial cell proliferation, migration, invasion and tube formation. These data clearly demonstrate the anti-angiogenic potential of nomilin by down-regulating the activation of MMPs, production of VEGF, NO and pro-inflammatory cytokines as well as up-regulating IL-2 and TIMP (Pratheeshkumar et al., 2011).

References

Kim J, Jayaprakasha GK, Patil BS. (2013). Limonoids and their anti-proliferative and anti-aromatase properties in human breast cancer cells. Food Funct, 4(2):258-65. doi: 10.1039/c2fo30209h.


Pratheeshkumar P, Raphael TJ & Kuttan G. (2011). Nomilin Inhibits Metastasis via Induction of Apoptosis and Regulates the Activation of Transcription Factors and the Cytokine Profile in B16F-10 Cells. Integr Cancer Ther. doi: 10.1177/1534735411403307


Pratheeshkumar P, Kuttan G. (2011). Nomilin inhibits tumor-specific angiogenesis by down-regulating VEGF, NO and pro-inflammatory cytokine profile and also by inhibiting the activation of MMP-2 and MMP-9. Eur J Pharmacol, 668(3):450-8. doi: 10.1016/j.ejphar.2011.07.029.

Nelumbo Extract (NLE):Neferine

Cancer: Liver, osteosarcoma, breast, melanoma

Action: Anti-angiogenic, cytostatic

Neferine is a major bis-benzylisoquinoline alkaloid derived from the green seed embryos of the Indian lotus (Nelumbo nucifera (Gaertn.)).

Identification of natural products that have anti-tumor activity is invaluable to the chemo-prevention and therapy of cancer. The embryos of lotus (Nelumbo nucifera) seeds are consumed in beverage in some parts of the world for their presumed health-benefiting effects. Neferine is a major alkaloid component in lotus embryos.

Hepatitis

Experimental results suggest that neferine exhibited cytotoxicity against HCC Hep3B cells, but not against HCC Sk-Hep1 and THLE-3, a normal human liver cell line. Results demonstrated neferine induced ER stress and apoptosis, acting through multiple signaling cascades by the activation of Bim, Bid, Bax, Bak, Puma, caspases-3, -6, -7, -8 and PARP, and the protein expression levels of Bip, calnexin, PDI, calpain-2 and caspase-12 were also upregulated dramatically by neferine treatment.

These observations reveal that the therapeutic potential of neferine in treating HCC Hep3B cells, containing copies of hepatitis B virus (HBV) genomes (Yoon et al., 2013).

Osteosarcoma

It was found that neferine possessed a potent growth-inhibitory effect on human osteosarcoma cells, but not on non-neoplastic human osteoblast cells. The inhibitory effect of neferine on human osteosarcoma cells was largely attributed to cell-cycle arrest at G1. The up-regulation of p21 by neferine was due to an increase in the half-life of p21 protein. Zhang et al. (2012) showed that neferine treatment led to an increased phosphorylation of p21 at Ser130 that was dependent on p38. Their results for the first time showed a direct anti-tumor effect of neferine, suggesting that consumption of neferine may have cancer-preventive and cancer-therapeutic benefit.

Breast Cancer

Qualitative analysis showed that NLE contained several compounds, including polyphenols. The polyphenols identified in NLE consisted primarily of gallic acid, rutin, and quercetin. Cell cycle analysis revealed that breast cancer MCF-7 cells treated with NLE were arrested at the G0/G1 phase. In an in vivo analysis, treatment with NLE (0.5 and 1%) effectively reduced tumor volume and tumor weight in mice inoculated with MCF-7 cells compared to the control samples.

These results confirmed that cell-cycle arrest was sufficient to elicit tumor regression following NLE treatment (Yang et al., 2011).

Melanoma

Methanolic extracts from the flower buds and leaves of sacred lotus (Nelumbo nucifera) were found to show inhibitory effects on melanogenesis in theophylline-stimulated murine B16 melanoma 4A5 cells. 3-30 µM nuciferine and N-methylasimilobine inhibited the expression of tyrosinase mRNA, 3-30 µM N-methylasimilobine inhibited the expression of TRP-1 mRNA, and 10-30 µM nuciferine inhibited the expression of TRP-2 mRNA (Nakamura et al., 2013).

References

Nakamura S, Nakashima S, Tanabe G, et al. (2013). Alkaloid constituents from flower buds and leaves of sacred lotus (Nelumbo nucifera, Nymphaeaceae) with melanogenesis inhibitory activity in B16 melanoma cells. Bioorg Med Chem, 21(3):779-87. doi: 10.1016/j.bmc.2012.11.038.


Yang MY, Chang YC, Chan KC et al. (2011). Flavonoid-enriched extracts from Nelumbo nucifera leaves inhibits proliferation of breast cancer in vitro and in vivo. European Journal of Integrative Medicine, 3(3):153-163. doi:10.1016/j.eujim.2011.08.008


Yoon JS, Kim HM, Yadunandam AK, et al. (2013). Neferine isolated from Nelumbo nucifera enhances anti-cancer activities in Hep3B cells: Molecular mechanisms of cell-cycle arrest, ER stress induced apoptosis and anti-angiogenic response. Phytomedicine, 20(11):1013–1022. doi:10.1016/j.phymed.2013.03.024.


Zhang XY, Liu ZJ, Xu B, et al. (2012). Neferine, an alkaloid ingredient in lotus seed embryo, inhibits proliferation of human osteosarcoma cells by promoting p38 MAPK-mediated p21 stabilization. European Journal of Pharmacology, 677(1–3):47–54.

Naringin

Cancer: TNBCa, melanoma, breast, colon, cervical

Action: Anti-inflammatory, anti-carcinogenic

Citrus plants are known to possess beneficial biological activities for human health. The total phenolics and flavonoids from a methanolic extract contained high total phenolics and flavonoids compared to ethanolic and boiling water extracts of Citrus aurantium. The anti-inflammatory result of methanolic extract showed appreciable reduction in nitric oxide production of stimulated RAW 264.7 cells at the presence of plant extract.

Breast Cancer, Colon Cancer

The anti-cancer activity of the methanolic extract of Citrus aurantium was investigated in vitro against human cancer cell lines; breast cancer MCF-7; MDA-MB-231 cell lines, human colon adenocarcinoma HT-29 cell line and Chang cell as a normal human hepatocyte. The obtained result demonstrated the moderate to appreciable activities against all cell lines tested and the compounds present in the extracts are non-toxic which make them suitable as potential therapeutics (Karimi et al., 2012).

Triple Negative (ER-/PR-/HER2-)

Breast Cancer (TNBCa)

Camargo et al. (2012) demonstrated that naringin inhibited cell proliferation, and promoted cell apoptosis and G1 cycle arrest, accompanied by increased p21 and decreased survivin. Meanwhile, β-catenin signaling pathway was found to be suppressed by naringin.

Levels of the pro-inflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) are raised in patients with TNBCa. Inhibition of tumor growth, survival increase and the reduction of TNF-α and IL-6 levels in rats bearing W256 treated with naringin strongly suggest that this compound has potential as an anti-carcinogenic drug.

Results indicate that naringin could inhibit growth potential of Triple-negative (ER-/PR-/HER2-) breast cancer (TNBC) by modulating -catenin pathway, which suggests naringin might be used as a potential supplement for the prevention and treatment of breast cancer (Li et al., 2013).

Cervical Cancer

Fruit-based cancer prevention entities, such as flavonoids and their derivatives, have demonstrated a marked ability to inhibit preclinical models of epithelial cancer cell growth and tumor formation. Ramesh & Alshatwi (2013) looked at the role of naringin-mediated chemo-prevention in relation to cervical carcinogenesis. The results suggest that the induction of apoptosis by naringin is through both death-receptor and mitochondrial pathways. Taken together, our results suggest that naringin might be an effective agent to treat human cervical cancer.

Melanoma

A study by Huang, Yang, Chiou (2011) investigated the molecular events of melanogenesis induced by naringenin in murine B16-F10 melanoma cells. Melanin content, tyrosinase activity and Western blot analysis were performed to elucidate the possible underlying mechanisms. Exposure of melanoma cells to naringenin resulted in morphological changes accompanied by the induction of melanocyte differentiation-related markers, such as melanin synthesis, tyrosinase activity, and the expression of tyrosinase and microphthalmia-associated transcription factor (MITF). They concluded that naringenin induced melanogenesis through the Wnt-β-catenin-signaling pathway.

References

Camargo CA, Gomes-Marcondes MC, Wutzki NC, Aoyama H. (2013). Naringin inhibits tumor growth and reduces interleukin-6 and tumor necrosis factor α levels in rats with Walker 256 carcinosarcoma. Anti-cancer Res, 32(1):129-33.


Huang YC, Yang CH, Chiou YL. (2011). Citrus flavanone naringenin enhances melanogenesis through the activation of Wnt/ β -catenin signaling in mouse melanoma cells. Phytomedicine. 18(14):1244-9. doi: 10.1016/j.phymed.2011.06.028.


Karimi E, Oskoueian E, Hendra R, Oskoueian A, Jaafar HZ. (2012). Phenolic compounds characterization and biological activities of Citrus aurantium bloom. Molecules, 17(2):1203-18. doi: 10.3390/molecules17021203.


Li HZ, Yang B, Huang J, et al. (2013). Naringin inhibits growth potential of human triple-negative breast cancer cells by targeting -catenin signaling pathway. Toxicology Letters, 220(2013):219-228


Ramesh E, Alshatwi AA. (2013). Naringin induces death receptor and mitochondria-mediated apoptosis in human cervical cancer (SiHa) cells. Food Chem Toxicol. 51:97-105. doi: 10.1016/j.fct.2012.07.033.

Moscatilin

Cancers:
Colon, lung, placenta, stomach, breast metastasis

Action: Anti-angiogenic, anti-metastatic, anti-tubulin, cytostatic, cytotoxic, cell-cycle arrest, anti-inflammatory

Stomach Cancer, Lung Cancer, Placental

The efficacy of using moscatilin, a natural anti-platelet agent extracted from the stems of Dendrobrium loddigesii, as an anti-cancer agent was studied. Results demonstrated that moscatilin exerts potent cytotoxic effect against cancer cell lines derived from different tissue origins, including those from the placenta, stomach, and lung, but not those from the liver. In addition, the mechanism of action of moscatilin may be related to its ability to induce a G2 phase arrest in responsive cells.

However, unlike some G2 arresting agents, moscatilin has no detectable inhibitory effect on cyclin B–cdc-2 kinase activity. Thus, the precise nature of its cytotoxic mechanism remains to be determined.

Results suggest that moscatilin is potentially efficacious for chemo-prevention and/or chemotherapy against some types of cancer (Ho & Chen, 2003).

Colorectal Cancer

The growth inhibition of moscatilin was screened on several human cancer cell lines. The effect of moscatilin on tubulin was detected in vitro. Following moscatilin treatment on colorectal HCT-116 cells, c-Jun NH(2)-terminal protein kinase (JNK) and caspase activation was studied by Western blot analysis, and DNA damage was done by Comet assay. Moscatilin induced a time-dependent arrest of the cell-cycle at G2/M, with an increase of cells at sub-G1. Moscatilin inhibited tubulin polymerization, suggesting that it might bind to tubulins. A parallel experiment showed that SP600125 significantly inhibits Taxol and vincristine induced HCT-116 cell apoptosis. This suggests that the JNK activation may be a common mechanism for tubulin-binding agents.

Collectively, results suggest that moscatilin induces apoptosis of colorectal HCT-116 cells via tubulin depolymerization and DNA damage leading to the activation of JNK and mitochondria-involved intrinsic apoptosis pathway (Chen et al., 2008).

Anti-inflammatory

Results showed that moscatilin (10-100 microM) had a significant inhibition in a concentration-dependent manner on pro-inflammatory enzymes (COX-2 and iNOS) expression and macrophage activation under LPS (100 ng/mL) treatment.

Hypoxia-inducible factor 1 (HIF-1) alpha was reported to initiate inflammation under cytokine stimulation or hypoxic conditions. Moscatilin had significant inhibition on HIF-1 expression via down-regulation of HIF-1 mRNA without affecting cell viability, translation machinery, or proteasome-mediated degradation of HIF-1. Collective data demonstrarted that moscatilin inhibited both COX-2 and iNOS expressions after LPS treatment in RAW264.7. Furthermore, moscatilin's inhibitory effect appears to be dependent on the repression of HIF-1alpha accumulation and NF-kappaB activation (Liu et al., 2010).

Lung Cancer; Angiogenesis

Moscatilin significantly inhibited growth of lung cancer cell line A549 (NSCLC) and suppressed growth factor-induced neovascularization. In addition, VEGF- and bFGF-induced cell proliferation, migration, and tube formation of HUVECs was markedly inhibited by moscatilin. Western blotting analysis of cell signaling molecules indicated that moscatilin inhibited ERK1/2, Akt, and eNOS signaling pathways in HUVECs.

Results suggest that inhibition of angiogenesis by moscatilin may be a major mechanism in cancer therapy (Tsai et al., 2010).

Lung Cancer

Investigation demonstrated that non-toxic concentrations of moscatilin were able to inhibit human non-small-cell lung cancer H23 cell migration and invasion. The inhibitory effect of moscatilin was associated with an attenuation of endogenous reactive oxygen species (ROS), in which hydroxyl radical was identified as a dominant species in the suppression of filopodia formation.

Results indicate a novel molecular basis of moscalitin inhibiting lung cancer cell motility and invasion. Moscalitin may have promising anti-metastatic potential as an agent for lung cancer therapy (Kowitdamrong, Chanvorachote, Sritularak & Pongrakhananon, 2013).

Breast Cancer; Metastasis

Moscatilin, derived from the orchid Dendrobrium loddigesii, has shown anti-cancer activity. The mechanism by which moscatilin suppresses the migration and metastasis of human breast cancer MDA-MB-231 cells in vitro and in vivo was evaluated.

Moscatilin was found to significantly inhibit breast cancer MDA-MB-231 cell migration by using scratch assays and Boyden chambers.

In an MDA-MB-231 metastatic animal model, moscatilin (100 mg/kg) significantly suppressed breast cancer metastasis to the lungs and reduced the number of metastatic lung nodules and lung weight without causing any toxicity.

Results indicated that moscatilin inhibited MDA-MB-231 cell migration via Akt- and Twist-dependent pathways, consistent with moscatilin's anti-metastatic activity in vivo. Therefore, moscatilin may be an effective compound for the prevention of human breast cancer metastasis (Pai et al., 2013).

References

Chen TH, Pan SL, Guh JH, et al. (2008). Moscatilin induces apoptosis in human colorectal cancer cells: a crucial role of c-Jun NH2-terminal protein kinase activation caused by tubulin depolymerization and DNA damage. Clinical Cancer Research, 14(13), 4250-4258. doi: 10.1158/1078-0432.CCR-07-4578.


Ho CK, Chen CC. (2003). Moscatilin from the orchid Dendrobrium loddigesii is a potential anti-cancer agent. Cancer Investigation, 21(5), 729-736.


Kowitdamrong A, Chanvorachote P, Sritularak B, Pongrakhananon V. (2013). Moscatilin inhibits lung cancer cell motility and invasion via suppression of endogenous reactive oxygen species. BioMed Research International., 2013, 765894. doi: 10.1155/2013/765894.


Liu YN, Pan SL, Peng CY, et al. (2010). Moscatilin repressed lipopolysaccharide-induced HIF-1alpha accumulation and NF-kappaB activation in murine RAW264.7 cells. Shock, 33(1), 70-5. doi: 10.1097/SHK.0b013e3181a7ff4a.


Pai HC, Chang LH, Peng CY, et al. (2013). Moscatilin inhibits migration and metastasis of human breast cancer MDA-MB-231 cells through inhibition of Akt and Twist signaling pathway.

Journal of Molecular Medicine (Berlin), 91(3), 347-56. doi: 10.1007/s00109-012-0945-5.

Tsai AC, Pan SL, Liao CH, et al. (2010). Moscatilin, a bibenzyl derivative from the India orchid Dendrobrium loddigesii, suppresses tumor angiogenesis and growth in vitro and in vivo. Cancer Letters, 292(2), 163-70. doi: 10.1016/j.canlet.2009.11.020.

Methyl Myristate, Methyl Palmitate, Methyl Stearate

Cancer: Leukemia

Action: Cytotoxic

Leukemia

Chemical investigation of the methanolic extract of the ascidian Didemnum psammatodes has led to the identification of 14 known compounds including three methyl esters: methyl myristate, methyl palmitate and methyl stearate.

The cytotoxic activity of these compounds was evaluated against a human leukemia cell line panel using the MTT assay. The mixture of the three methyl esters was the most active group of compounds, showing anti-proliferative and cytotoxic effects. Further studies on their mode of action suggest that these activities are connected with inhibition of DNA synthesis and induction of both necrosis and apoptosis (Takeara et al., 2008).

Reference

Takeara R, Jimenez PC, Wilke DV, et al. (2008). Antileukemic effects of Didemnum psammatodes (Tunicata: Ascidiacea) constituents. Comparative Biochemistry and Physiology: Molecular & Integrative Physiology, 151(3), 363-9.

Matricaria chamomilla/Matricaria recutita

Cancer: Colorectal., ovarian, testicular, bladder, lung

Action: Neuropathy, anti-inflammatory

Colorectal Cancer; Ovarian Cancer; Testicular Cancer; Bladder Cancer; Lung Cancer; Chemotherapy

Studies have shown that cisplatin could have painful effects on human and animal models. Matricaria chamomilla (MC) has analgesic and anti-inflammatory effects, and may hence be an effective treatment for ciplatin-induced peripheral neuropathy as a replacement for morphine. Experiments were performed on 60 NMRI male mice weighed 25 g to 30 g, which have been divided into 6 groups. The first group received normal saline; the second group received MC hydroalcoholic extract; the third group received cisplatin; the fourth group received MC hydroalcoholic extract and cisplatin, 96 hours before formalin test; the fifth group received morphine and the sixth group received cisplatin and morphine.

Results showed that formalin induced significant (P < 0.05) pain response (the first phase: 0–5 min and the second phase: 15–40 min after injection). Administration of MC extract before formalin injection showed significant (P < 0.05) decrease of pain responses in the first and second phase. Administration of cisplatin produced significant (P < 0.05) increase in pain response in both phases of formalin test. Injection of MC extract and cisplatin together have shown that MC is able to decrease the second phase of cisplatin-induced pain significantly (P < 0.05).

In comparison morphine has analgesic effects in the first phase and MC extract has anti- inflammatory effects in the second phase of formalin test significantly (P < 0.05). MC and cisplatin have analgesic and painful neuropathic respective effects, and MC hydroalcoholic extract is able to decrease cisplatin-induced pain and inflammation better than morphine (Abad et al., 2011).

Anti-inflammatory

Flavonoid-7-glycosides, major constituents of chamomile flowers, may be responsible for the anti-inflammatory action, which is due to the inhibition of neutrophil elastase and gastric metalloproteinase-9 activity and secretion; the inhibition occurring in a concentration dependent manner (Bulgari et al., 2012).

The anti-cancer properties of aqueous and methanolic extracts of Matricaria chamomilla against various human cancer cell lines were investigated. Chamomile exposure resulted in differential apoptosis in cancer cells but not in normal cells at similar doses. HPLC analysis of chamomile extract confirmed apigenin 7-O-glucoside as the major constituent of chamomile; some minor glycoside components were also observed. Apigenin glucosides inhibited cancer cell growth but to a lesser extent than the parent aglycone, apigenin (Srivastava & Gupta, 2007).

References

Abad NA, Nouri MHK, Gharjanie A, Tavakoli F. (2011). Effect of Matricaria chamomilla Hydroalcoholic Extract on Cisplatin-induced Neuropathy in Mice. Chinese Journal of Natural Medicines, 9(2):126–131


Bulgari M, Sangiovanni E, Colombo E, et al. (2012). Inhibition of neutrophil elastase and metalloprotease-9 of human adenocarcinoma gastric cells by chamomile (Matricaria recutita L.) infusion. Phytother Res, 26(12):1817-22. doi: 10.1002/ptr.4657.


Srivastava JK, Gupta S. (2007). Anti-proliferative and apoptotic effects of chamomile extract in various human cancer cells. J Agric Food Chem, 55(23):9470-8.

Magnolol

Cancer:
Bladder, breast, colon, prostate, glioblastoma, ovarian, leukemia, lung

Action: Anti-inflammatory, apoptosis, inhibits angiogenesis, anti-metastatic

Magnolol (Mag), an active constituent isolated from the Chinese herb hou po (Magnolia officinalis (Rehder & Wilson)) has long been used to suppress inflammatory processes. It has anti-cancer activity in colon, hepatoma, and leukemia cell lines.

Anti-inflammatory

Magnolol (Mag) suppressed IL-6-induced promoter activity of cyclin D1 and monocyte chemotactic protein (MCP)-1 for which STAT3 activation plays a role. Pre-treatment of ECs with Mag dose-dependently inhibited IL-6-induced Tyr705 and Ser727 phosphorylation in STAT3 without affecting the phosphorylation of JAK1, JAK2, and ERK1/2. Mag pre-treatment of these ECs dose-dependently suppressed IL-6-induced promoter activity of intracellular cell adhesion molecule (ICAM)-1 that contains functional IL-6 response elements (IREs).

In conclusion, our results indicate that Mag inhibits IL-6-induced STAT3 activation and subsequently results in the suppression of downstream target gene expression in ECs. These results provide a therapeutic basis for the development of Mag as an anti-inflammatory agent for vascular disorders including atherosclerosis (Chen et al., 2006).

Bladder Cancer; Inhibits Angiogenesis

In the present study, Chen et al. (2013) demonstrated that magnolol significantly inhibited angiogenesis in vitro and in vivo, evidenced by the attenuation of hypoxia and vascular endothelial growth factor (VEGF)-induced tube formation of human umbilical vascular endothelial cells, vasculature generation in chicken chorioallantoic membrane, and Matrigel plug.

In hypoxic human bladder cancer cells (T24), treatment with magnolol inhibited hypoxia-stimulated H2O2 formation, HIF-1α induction including mRNA, protein expression, and transcriptional activity as well as VEGF secretion. Interestingly, magnolol also acts as a VEGFR2 antagonist, and subsequently attenuates the downstream AKT/mTOR/p70S6K/4E-BP-1 kinase activation both in hypoxic T24 cells and tumor tissues. As expected, administration of magnolol greatly attenuated tumor growth, angiogenesis and the protein expression of HIF-1α, VEGF, CD31, a marker of endothelial cells, and carbonic anhydrase IX, an endogenous marker for hypoxia, in the T24 xenograft mouse model.

Collectively, these findings strongly indicate that the anti-angiogenic activity of magnolol is, at least in part, mediated by suppressing HIF-1α/VEGF-dependent pathways, and suggest that magnolol may be a potential drug for human bladder cancer therapy.

Colon Cancer; Induces Apoptosis

Emerging evidence has suggested that activation of AMP-activated protein kinase (AMPK), a potential cancer therapeutic target, is involved in apoptosis in colon cancer cells. However, the effects of magnolol on human colon cancer through activation of AMPK remain unexplored.

Magnolol displayed several apoptotic features, including propidium iodide labeling, DNA fragmentation, and caspase-3 and poly(ADP-ribose) polymerase cleavages. Park et al. (2012) showed that magnolol induced the phosphorylation of AMPK in dose- and time-dependent manners.

Magnolol down-regulated expression of the anti-apoptotic protein Bcl2, up-regulated expression of pro-apoptotic protein p53 and Bax, and caused the release of mitochondrial cytochrome c. Magnolol-induced p53 and Bcl2 expression was abolished in the presence of compound C. Magnolol inhibited migration and invasion of HCT-116 cells through AMPK activation. These findings demonstrate that AMPK mediates the anti-cancer effects of magnolol through apoptosis in HCT-116 cells.

Ovarian Cancer

Treatment of HER-2 overexpressing ovarian cancer cells with magnolol down-regulated the HER-2 downstream PI3K/Akt signaling pathway, and suppressed the expression of downstream target genes, vascular endothelial growth factor (VEGF), matrix metalloproteinase 2 (MMP2) and cyclin D1. Consistently, magnolol-mediated inhibition of MMP2 activity could be prevented by co-treatment with epidermal growth factor. Migration assays revealed that magnolol treatment markedly reduced the motility of HER-2 overexpressing ovarian cancer cells. These findings suggest that magnolol may act against HER-2 and its downstream PI3K/Akt/mTOR-signaling network, thus resulting in suppression of HER-2mediated transformation and metastatic potential in HER-2 overexpressing ovarian cancers. These results provide a novel mechanism to explain the anti-cancer effect of magnolol (Chuang et al., 2011).

Lung Cancer

Magnolol has been found to inhibit cell growth, increase lactate dehydrogenase release, and modulate cell cycle in human lung carcinoma A549 cells. Magnolol induced the activation of caspase-3 and cleavage of Poly-(ADP)-ribose polymerase, and decreased the expression level of nuclear factor-κB/Rel A in the nucleus. In addition, magnolol inhibited basic fibroblast growth factor-induced proliferation and capillary tube formation of human umbilical vein endothelial cells. These data indicate that magnolol is a potential candidate for the treatment of human lung carcinoma (Seo et al., 2011).

Prostate Cancer; Anti-metastatic

Matrix metalloproteinases (MMPs) are enzymes involved in various steps of metastasis development. The objective of this study was to study the effects of magnolol on cancer invasion and metastasis using PC-3 human prostate carcinoma cells. Magnolol inhibited cell growth in a dose-dependent manner. In an invasion assay conducted in Transwell chambers, magnolol showed 33 and 98% inhibition of cancer cell at 10 microM and 20 microM concentrations, respectively, compared to the control. The protein and mRNA levels of both MMP-2 and MMP-9 were down-regulated by magnolol treatment in a dose-dependent manner.

These results demonstrate the anti-metastatic properties of magnolol in inhibiting the adhesion, invasion, and migration of PC-3 human prostate cancer cells (Hwang et al., 2010).

Glioblastoma Cancer

Magnolol has been found to concentration-dependently (0-40 microM) decrease the cell number in a cultured human glioblastoma cancer cell line (U373) and arrest the cells at the G0/G1 phase of the cell-cycle.

Pre-treatment of U373 with p21/Cip1 specific antisense oligodeoxynucleotide prevented the magnolol-induced increase of p21/Cip1 protein levels and the decrease of DNA synthesis. Magnolol at a concentration of 100 microM induced DNA fragmentation in U373. These findings suggest the potential applications of magnolol in the treatment of human brain cancers (Chen et al. 2011).

Inhibits Angiogenesis

Magnolol inhibited VEGF-induced Ras activation and subsequently suppressed extracellular signal-regulated kinase (ERK), phosphatidylinositol-3-kinase (PI3K)/Akt and p38, but not Src and focal adhesion kinase (FAK). Interestingly, the knockdown of Ras by short interfering RNA produced inhibitory effects that were similar to the effects of magnolol on VEGF-induced angiogenic signaling events, such as ERK and Akt/eNOS activation, and resulted in the inhibition of proliferation, migration, and vessel sprouting in HUVECs.

In combination, these results demonstrate that magnolol is an inhibitor of angiogenesis and suggest that this compound could be a potential candidate in the treatment of angiogenesis-related diseases (Kim et al., 2013).

References

Chen LC, Liu YC, Liang YC, Ho YS, Lee WS. (2009). Magnolol inhibits human glioblastoma cell proliferation through up-regulation of p21/Cip1. J Agric Food Chem, 57(16):7331-7. doi: 10.1021/jf901477g.


Chen MC, Lee CF, Huang WH, Chou TC. (2013). Magnolol suppresses hypoxia-induced angiogenesis via inhibition of HIF-1 α /VEGF signaling pathway in human bladder cancer cells. Biochem Pharmacol, 85(9):1278-87. doi: 10.1016/j.bcp.2013.02.009.


Chen SC, Chang YL, Wang DL, Cheng JJ. (2006). Herbal remedy magnolol suppresses IL-6-induced STAT3 activation and gene expression in endothelial cells. Br J Pharmacol, 148(2): 226–232. doi: 10.1038/sj.bjp.0706647


Chuang TC, Hsu SC, Cheng YT, et al. (2011). Magnolol down-regulates HER2 gene expression, leading to inhibition of HER2-mediated metastatic potential in ovarian cancer cells. Cancer Lett, 311(1):11-9. doi: 10.1016/j.canlet.2011.06.007.


Hwang ES, Park KK. (2010). Magnolol suppresses metastasis via inhibition of invasion, migration, and matrix metalloproteinase-2/-9 activities in PC-3 human prostate carcinoma cells. Biosci Biotechnol Biochem, 74(5):961-7.


Kim KM, Kim NS, Kim J, et al. (2013). Magnolol Suppresses Vascular Endothelial Growth Factor-Induced Angiogenesis by Inhibiting Ras-Dependent Mitogen-Activated Protein Kinase and Phosphatidylinositol 3-Kinase/Akt Signaling Pathways. Nutr Cancer.


Park JB, Lee MS, Cha EY, et al. (2012). Magnolol-induced apoptosis in HCT-116 colon cancer cells is associated with the AMP-activated protein kinase signaling pathway. Biol Pharm Bull, 35(9):1614-20.


Seo JU, Kim MH, Kim HM, Jeong HJ. (2011). Anti-cancer potential of magnolol for lung cancer treatment. Arch Pharm Res, 34(4):625-33. doi: 10.1007/s12272-011-0413-8.

Lunasin

Cancer: Colon, breast, liver metastasis

Action: Induces apoptosis, MDR

Lunasin is a peptide found in soy, barley, wheat, and rye, including Glycine max [(L.) Merr.], Hordeum vulgare L., Triticum (L.) genus and Secale cereale L.

Colon Cancer; Metastasis

Lunasin bound with α(5)β(1) integrin and internalized into the nucleus of KM12L4 human colon cancer cells. Lunasin (10µM) inhibited the activation of focal adhesion kinase (FAK) by 28%, 39% and 60% in RKO, HCT-116 and KM12L4 human colon cancer cells, respectively. Lunasin caused an increase in the expression of the inhibitor of kappa B alpha (IκB-α), a decrease in nuclear p50 NF-κB and a reduction in the migration of cancer cells. Lunasin (4mg/kg bw) inhibited metastasis and potentiated the effect of oxaliplatin by reducing the expression of proliferating cell nuclear antigen.

Liver metastatic nodules were reduced from 28 (PBS) to 14 (lunasin, P=0.047) while combination of lunasin and oxaliplatin to 5 (P=0.004). The tumor burden was reduced from 0.13 (PBS) to 0.10 (lunasin, P=0.039) to 0.04 (lunasin+oxaliplatin, P<0.0001). Moreover, lunasin potentiated the effect of oxaliplatin in modifying expression of proteins involved in apoptosis and metastasis including Bax, Bcl-2, IKK-α and p-p65. Lunasin inhibited metastasis of human colon cancer cells by direct binding with α(5)β(1) integrin suppressing FAK/ERK/NF-κB signaling, and potentiated the effect of oxaliplatin in preventing the outgrowth of metastasis (Dia et al., 2011).

Induces Apoptosis

Galvez et al. (2001) demonstrated previously that transfection of mammalian cells with the lunasin gene arrests mitosis, leading to cell death. Here they show that exogenous application of the lunasin peptide inhibits chemical carcinogen-induced transformation of murine fibroblast cells to cancerous foci. The results suggest a mechanism whereby lunasin selectively induces apoptosis, mostly in cells undergoing transformation, by preventing histone acetylation. In support of this, lunasin selectively induces apoptosis in E1A-transfected cells but not in nontransformed cells. Finally, in the SENCAR mouse skin cancer model, dermal application of lunasin (250 microg/week) reduces skin tumor incidence by approximately 70%, decreases tumor yield/mouse, and delays the appearance of tumors by 2 weeks relative to the positive control. These results point to the role of lunasin as a new chemo-preventive agent that functions possibly via a chromatin modification mechanism.

Breast Cancer

Combinations of two or more chemo-preventive agents are currently being used to achieve greater inhibitory effects on breast cancer cells. This study reveals that both aspirin and lunasin inhibit, in a dose-dependent manner, human estrogen-independent breast cancer MDA-MB-231 cell proliferation.

These compounds arrest the cell-cycle in the S- and G1-phases, respectively, acting synergistically to induce apoptosis. The cell growth-inhibitory effect of a lunasin/aspirin combination is achieved, at least partially, by modulating the expression of genes encoding G1 and S-phase regulatory proteins. Lunasin/aspirin therapy exerts its potent pro-apoptotic effect, at least partially achieved through modulating the extrinsic-apoptosis dependent pathway.

Therefore, our results suggest that a combination of these two compounds is a promising strategy to prevent/treat breast cancer (Hsieh et al., 2010).

Colon Cancer; MDR

Various human colon cancer cell lines which underwent metastasis were evaluated in vitro using cell flow cytometry and fluorescence microscopy. Lunasin cytotoxicity to different colon cancer cells correlated with the expression of α5b1 integrin was investigated, being most potent to KM12L4 cells (IC50 = 13 µM). Lunasin arrested cell-cycle at G2/M phase with concomitant increase in the expression of cyclin-dependent kinase inhibitors p21 and p27. Lunasin (5–25 µM) activated the apoptotic mitochondrial pathway as evidenced by changes in the expressions of Bcl-2, Bax, nuclear clusterin, cytochrome c and caspase-3 in KM12L4 and KM12L4-OxR.

Lunasin increased the activity of initiator caspase-9 leading to the activation of caspase-3 and also modified the expression of human extracellular matrix and adhesion genes, down-regulating integrin α5, SELE, MMP10, integrin β2 and COL6A1 by 5.01-, 6.53-, 7.71-, 8.19- and 10.10-fold, respectively, while up-regulating COL12A1 by 11.61-fold. Lunasin can be used in cases where resistance to chemotherapy developed (Dia et al., 2011).

References

Dia VP, Gonzalez de Mejia E. (2011). Lunasin potentiates the effect of oxaliplatin preventing outgrowth of colon cancer metastasis, binds to α5β1 integrin and suppresses FAK/ERK/NF-κ B signaling, Cancer Lett, 313(2):167-80.


Dia VP, Gonzalez de Mejia E. (2011). Lunasin induces apoptosis and modifies the expression of genes associated with extracellular matrix and cell adhesion in human metastatic colon cancer cells. Mol Nutr Food Res, 55(4):623-34. doi: 10.1002/mnfr.201000419.


Galvez AF, Chen N, Macasieb J, de Lumen BO. (2001). Chemo-preventive property of a soybean peptide (lunasin) that binds to deacetylated histones and inhibits acetylation. Cancer Res, 61(20):7473-8.


Hsieh CC, Hern‡ndez-Ledesma B, de Lumen BO. (2010). Lunasin, a novel seed peptide, sensitizes human breast cancer MDA-MB-231 cells to aspirin-arrested cell-cycle and induced apoptosis. Chem Biol Interact, 186(2):127-34. doi: 10.1016/j.cbi.2010.04.027.

Koetjapic acid

Cancer: none noted

Action: Anti-angiogenic

Koetjapic acid is isolated from Sandoricum koetjape (Merr.).

Angiogenesis, the formation of new blood vessels, has become an important target in cancer therapy. Angiogenesis plays an important role in tumor growth and metastasis. The solvent extract of this plant species was shown previously to have strong anti-angiogenic activity; however the active ingredient(s) that conferred the biological activity, and the mode of action, were not established. Given the high concentration of koetjapic acid (KA) in S. koetjape, an attempt has been made in this study to investigate the anti-angiogenic properties of KA.

Treatment with 10-50 mug/ml KA resulted in dose-dependent inhibition of new blood vessel growth in ex vivo rat aortic ring assay. KA was found to be non-cytotoxic against HUVECs with IC50 40.97 +/- 0.37 mug/ml. KA inhibited major angiogenesis process steps, endothelial cell migration and differentiation as well as VEGF expression. The non-cytotoxic compound, KA, may be a potent anti-angiogenic agent and its activity may be attributed to inhibition of endothelial cells migration and differentiation as well VEGF suppression (Nassar et al., 2011).

References

Nassar ZD, Aisha AFAA, Ahamed MBK, et al. (2011). Anti-angiogenic properties of Koetjapic acid, a natural triterpene isolated from Sandoricum koetjaoe Merr. Cancer Cell International., 11:12. doi:10.1186/1475-2867-11-12.

Jasminoside

Cancer: Melanoma

Action: Anti-oxidation, anti-inflammatory

Melanoma

A new iridoid glycoside, 10-O-(4'-O-methylsuccinoyl) geniposide (7), and two new pyronane glycosides, jasminosides Q and R (13 and 14, resp.), along with nine known iridoid glycosides, 1-6 and 8-10, and two known pyronane glycosides, 11 and 12, were isolated from a MeOH extract of Gardeniae Fructus, the dried ripe fruit of Gardenia jasminoides (Rubiaceae).

The structures of new compounds were elucidated on the basis of extensive spectroscopic analyzes and comparison with literature.

Upon evaluation of compounds 1-14 on the melanogenesis in B16 melanoma cells induced with α-melanocyte-stimulating hormone (α-MSH), three compounds, i.e., 6-O-p-coumaroylgeniposide (3), 7, and 6'-O-sinapoyljasminoside (12), exhibited inhibitory effects with 21.6-41.0 and 37.5-47.7% reduction of melanin content at 30 and 50 µM, respectively, with almost no toxicity to the cells (83.7-106.1% of cell viability at 50 µM) (Akisha et al., 2012).

Anti-oxidation, Anti-inflammatory

The phytochemistry of Gardenia jasminoides J. Ellis (GJE) and its main constituents crocins and iridoid glycosides was investigated. Numerous studies have confirmed that crocins and iridoid glycosides have effects of anti-oxidation, anti-inflammatory, anti-atherosclerosis, anti-ischemic brain injuries, anti-platelet aggregation, anti-hyperglycemia, and anti-hyperlipidemia. It has been shown that GJE extract markedly prolonged bleeding time and inhibited platelet aggregation and thrombosis. It also has significant proliferation effect on both endothelial cells and endothelial progenitor cells (Liu et al., 2013).

References

Akihisa T, Watanabe K, Yamamoto A, et al. (2012). Melanogenesis inhibitory activity of monoterpene glycosides from Gardeniae Fructus. Chemistry & Biodiversity, 9(8), 1490-9. doi: 10.1002/cbdv.201200030.


Liu H, Chen YF, Li F, Zhang HY. (2013). Fructus Gardenia (Gardenia jasminoides J. Ellis) phytochemistry, pharmacology of cardiovascular, and safety with the perspective of new drugs development. J Asian Nat Prod Res, 15(1):94-110. doi: 10.1080/10286020.2012.723203.

Indirubin

Cancer:
Chronic myelogenous leukemia, lung, breast, head and neck, prostate, acute myeloid leukemia, prostate

Action: Aryl hydrocarbon Receptor (AhR) regulator, inhibits angiogenesis

Indirubin is the active component of many plants from the Isatis (L.) genus, including Isatis tinctoria (L.).

Indirubin is the active ingredient of Danggui Longhui Wan, a mixture of plants that is used in traditional Chinese medicine to treat chronic diseases. Indirubin and its analogues are potent inhibitors of cyclin-dependent kinases (CDKs). The crystal structure of CDK2 in complex with indirubin derivatives shows that indirubin interacts with the kinase's ATP-binding site through van der Waals interactions and three hydrogen bonds. Indirubin-3'-monoxime inhibits the proliferation of a large range of cells, mainly through arresting the cells in the G2/M phase of the cell-cycle. These results have implications for therapeutic optimization of indigoids (Hoessel et al., 1999).

Formula; Huang Lian (Rhizoma Coptidis Recens), Huang Qin (Radix Scutellariae Baicalensis), Huang Bai (Cortex Phellodendri), Zhi Zi (Fructus Gardeniae Jasminoidis), Dang Gui (Radix Angelicae Sinensis), Lu Hui (Herba Aloes), Long Dan Cao (Radix Gentianae Longdancao), Da Huang (Radix et Rhizoma Rhei), Mu Xiang (Radix Aucklandiae Lappae), Qing Dai (Indigo Pulverata Levis), She Xiang (Secretio Moschus)

Leukemia

Indirubin, a 3, 2' bisindole isomer of indigo was originally identified as the active principle of a traditional Chinese preparation and has been proven to exhibit anti-leukemic effectiveness in chronic myelocytic leukemia. Indirubin was detected to represent a novel lead structure with potent inhibitory potential towards cyclin-dependent kinases (CDKs) resulting from high affinity binding into the enzymes ATP binding site. This seminal finding triggered research to improve the pharmacological activities of the parent molecule within comprehensive structure-activity studies. Molecular modifications made novel anti-cancer compounds accessible with strongly improved CDK inhibitory potential and with broad-spectrum anti-tumor activity.

This novel family of compounds holds strong promise for clinical anti-cancer activity and might be useful also in several important non-cancer indications, including Alzheimer's disease or diabetes (Eisenbrand et al., 2004).

Aryl Hydrocarbon Receptor (AhR) Regulator; Breast Cancer

The aryl hydrocarbon receptor (AhR), when activated by exogenous ligands such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), regulates expression of several phase I and phase II enzymes and is also involved in the regulation of cell proliferation. One putative endogenous ligand is indirubin, which was recently identified in human urine and bovine serum. We determined the effect of indirubin in MCF-7 breast cancer cells on induction of the activities of cytochromes P450 (CYP) 1A1 and 1B1. With 4 hours exposure, the effects of indirubin and TCDD at 10nM on CYP activity were comparable, but the effects of indirubin, unlike those of TCDD, were transitory. Indirubin-induced ethoxyresorufin-O-deethylase activity was maximal by 6–9 hours post-exposure and had disappeared by 24 hours, whereas TCDD-induced activities remained elevated for at least 72 hours.

Thus, if indirubin is an endogenous AhR ligand, then AhR-mediated signaling by indirubin is likely to be transient and tightly controlled by the ability of indirubin to induce CYP1A1 and CYP1B1, and hence its own metabolism (Spink et al., 2003).

Chronic Myelogenous Leukemia (CML)

Indirubin is the major active anti-tumor component of a traditional Chinese herbal medicine used for treatment of chronic myelogenous leukemia (CML). In a study investigating its mechanism of action, indirubin derivatives (IRDs) were found to potently inhibit Signal Transducer and Activator of Transcription 5 (Stat5) protein in CML cells.

Compound E804, which is the most potent in this series of IRDs, blocked Stat5 signaling in human K562 CML cells, imatinib-resistant human KCL-22 CML cells expressing the T315I mutant Bcr-Abl (KCL-22M), and CD34-positive primary CML cells from patients.

In sum, these findings identify IRDs as potent inhibitors of the SFK/Stat5 signaling pathway downstream of Bcr-Abl, leading to apoptosis of K562, KCL-22M and primary CML cells. IRDs represent a promising structural class for development of new therapeutics for wild type or T315I mutant Bcr-Abl-positive CML patients (Nam et al., 2012).

Lung Cancer

A novel indirubin derivative, 5'-nitro-indirubinoxime (5'-NIO), exhibits a strong anti-cancer activity against human cancer cells. Here, the 5'-NIO-mediated G1 cell-cycle arrest in lung cancer cells was associated with a decrease in protein levels of polo-like kinase 1 (Plk1) and peptidyl-prolyl cis/trans isomerase Pin1. These findings suggest that 5'-NIO have potential anti-cancer efficacy through the inhibition of Plk1 or/and Pin1 expression (Yoon et al., 2012).

The control lung tissue showed a normal architecture with clear alveolar spaces. Interestingly, the indirubin-3-monoxime treated groups showed reduced adenocarcinoma with appearance of alveolar spaces. Transmission Electron Microscopic (TEM) studies of lung sections of [B(α)P]-induced lung cancer mice showed the presence of phaemorphic cells with dense granules and increased mitochondria.

The lung sections of mice treated with indirubin-3-monoxime showed the presence of shrunken, fragmented, and condensed nuclei implying apoptosis. The effects were dose-dependent and prominent in 10 mg/kg/5 d/week groups, suggesting the therapeutic role of indirubin analogue against this deadly human malignancy. These results indicate that indirubin-3-monoxime brings anti-tumor effect against [B(α)P]-induced lung cancer by its apoptotic action in A/J mice (Ravichandran et al., 2010).

Head and Neck Cancer

The effects of 5'-nitro-indirubinoxime (5'-NIO), an indirubin derivative, on metastasis of head and neck cancer cells were investigated and the underlying molecular mechanisms involved in this process explored.

After treatment of head and neck cancer cells with 5'-NIO, cell metastatic behaviors such as colony formation, invasion, and migration were inhibited in a concentration-dependent manner. 5'-NIO inhibited the beta1 Integrin/FAK/Akt pathway which can then facilitate invasion and/or migration of cancer cells through the extracellular matrix (ECM). Moreover, treatment of head and neck cancer cell with Integrin β1 siRNA or FAK inhibitor effectively inhibited the invasion and migration, suggesting their regulatory role in invasiveness and migration of head and neck cancer cells. It was concluded that 5'-NIO inhibits the metastatic ability of head and neck cancer cells by blocking the Integrin β1/FAK/Akt pathway (Kim et al., 2011).

Prostate Cancer; Inhibits Angiogenesis

Indirubin, the active component of a traditional Chinese herbal medicine, Banlangen, has been shown to exhibit anti-tumor and anti-inflammation effects; however, its role in tumor angiogenesis, the key step involved in tumor growth and metastasis, and the involved molecular mechanism is unknown.

To address this shortfall in the existing research, it was identified that indirubin inhibited prostate tumor growth through inhibiting tumor angiogenesis. It was found that indirubin inhibited angiogenesis in vivo. The inhibition activity of indirubin in endothelial cell migration, tube formation and cell survival in vitro has also been shown. Furthermore, indirubin suppressed vascular endothelial growth factor receptor 2-mediated Janus kinase (JAK)/STAT3 signaling pathway. This study provided the first evidence for anti-tumor angiogenesis activity of indirubin and the related molecular mechanism.

These investigations suggest that indirubin is a potential drug candidate for angiogenesis-related diseases (Zhang et al., 2011).

Acute Myeloid Leukemia

Indirubin derivatives were identified as potent FLT3 tyrosine kinase inhibitors with anti-proliferative activity at acute myeloid leukemic cell lines, RS4;11 and MV4;11 which express FLT3-WT and FLT3-ITD mutation, respectively. Among several 5 and 5'-substituted indirubin derivatives, 5-fluoro analog, 13 exhibited potent inhibitory activity at FLT3 (IC(50)=15 nM) with more than 100-fold selectivity versus 6 other kinases and potent anti-proliferative effect for MV4;11 cells (IC(50)=72 nM) with 30-fold selectivity versus RS4;11 cells.

Cell cycle analysis indicated that compound 13 induced cell-cycle arrest at G(0)/G(1) phase in MV4;11 cells (Choi et al., 2010).

References

Choi SJ, Moon MJ, Lee SD, et al. (2010). Indirubin derivatives as potent FLT3 inhibitors with anti-proliferative activity of acute myeloid leukemic cells. Bioorg Med Chem Lett, 20(6):2033-7.


Eisenbrand G, Hippe F, Jakobs S, Muehlbeyer S. (2004). Molecular mechanisms of indirubin and its derivatives: novel anti-cancer molecules with their origin in traditional Chinese phytomedicine. J Cancer Res Clin Oncol, 130(11):627-35


Hoessel R, Leclerc S, Endicott JA, et al. (1999). Indirubin, the active constituent of a Chinese antileukaemia medicine, inhibits cyclin-dependent kinases. Nat Cell Biol, 1(1):60-7.


Kim SA, Kwon SM, Kim JA, et al. (2011). 5'-Nitro-indirubinoxime, an indirubin derivative, suppresses metastatic ability of human head and neck cancer cells through the inhibition of Integrin β 1/FAK/Akt signaling. Cancer Lett, 306(2):197-204.


Nam S, Scuto A, Yang F, et al. (2012). Indirubin derivatives induce apoptosis of chronic myelogenous leukemia cells involving inhibition of Stat5 signaling. Mol Oncol, 6(3):276-83.


Ravichandran K, Pal A, Ravichandran R. (2010). Effect of indirubin-3-monoxime against lung cancer as evaluated by histological and transmission electron microscopic studies. Microsc Res Tech, 73(11):1053-8.


Spink BC, Hussain MM, Katz BH, Eisele L, Spink DC. (2003). Transient induction of cytochromes P450 1A1 and 1B1 in MCF-7 human breast cancer cells by indirubin. Biochem Pharmacol, 66(12):2313-21.


Yoon HE, Kim SA, Choi HS, et al. (2012). Inhibition of Plk1 and Pin1 by 5'-nitro-indirubinoxime suppresses human lung cancer cells. Cancer Lett, 316(1):97-104.


Zhang X, Song Y, Wu Y, et al. (2011). Indirubin inhibits tumor growth by anti-tumor angiogenesis via blocking VEGFR2-mediated JAK/STAT3 signaling in endothelial cell. Int J Cancer, 129(10):2502-11. doi: 10.1002/ijc.25909.

Icaritin

Cancer:
Endometrial., chronic myeloid leukemia, prostate, breast

Action: Radio-sensitizer, cell-cycle arrest, ER modulator

Icaritin is a compound in several species of the genus Epimedium (L.).

Cell-cycle Arrest

Icariin and icaritin with prenyl group have been demonstrated to have selective estrogen receptor modulating activities. Icaritin-induced growth inhibition was associated with G(1) arrest (P<0.05), and G(2)-M arrest depending upon doses. Consistent with G(1) arrest, icaritin increased protein expressions of pRb, p27(Kip1) and p16(Ink4a), while showing decrease in phosphorylated pRb, Cyclin D1 and CDK4.

Comparatively, icariin has much lower effects on PC-3 cells and showed only weak G(1) arrest, suggesting a possible structure-activity relationship. These findings suggested a novel anti-cancer efficacy of icaritin mediated selectively via induction of cell-cycle arrest but not associated with estrogen receptors in PC-3 cells (Huang et al., 2007).

Estrogen Receptor (ER) Modulator; Endometrial Cancer

Icaritin has selective estrogen receptor (ER) modulating activities, and posseses anti-tumor activity. The effect of icaritin on cell growth of human endometrial cancer Hec1A cells was investigated and it was found that icaritin potently inhibited proliferation of Hec1A cells. Icaritin also induced cell apoptosis accompanied by activation of caspases. Icaritin treatment also induced expression of pro-apoptotic protein Bax with a concomitant decrease of Bcl-2 expression.

These results demonstrate that icaritin induced sustained ERK 1/2 activation and inhibited growth of endometrial cancer Hec1A cells, and provided a rationale for preclinical and clinical evaluation of icaritin for endometrial cancer therapy (Tong et al., 2011).

Breast cancer

In research carried out to probe breast cancer cell growth mechanisms, icaritin has been found to strongly inhibit the growth of breast cancer MDA-MB-453 and MCF7 cells. At concentrations of 2–3 µM, icaritin induced cell-cycle arrest at the G2/M phase accompanied by a down-regulation of the expression levels of the G2/M regulatory proteins such as cyclinB, cdc2 and cdc25C.

Icaritin at concentrations of 4–5 µM, however, induced apoptotic cell death. In addition, icaritin also induced a sustained phosphorylation of extracellular signal-regulated kinase (ERK) in these breast cancer cells.

Icaritin more potently inhibited growth of the breast cancer stem/progenitor cells compared to anti-estrogen tamoxifen. These results indicate that icaritin is a potent growth inhibitor for breast cancer cells and provides a rationale for preclinical and clinical evaluations of icaritin for breast cancer therapy (Guo et al., 2011).

Radio-sensitizer

The combination of Icaritin at 3 µM or 6 µM with 6 or 8 Gy of ionizing radiation (IR) in the clonogenic assay yielded an ER (enhancement ratio) of 1.18 or 1.28, CI (combination index) of 0.38 or 0.19 and DRI (dose reducing index) of 2.51 or 5.07, respectively. These findings strongly suggest that Icaritin exerted a synergistic killing effect with radiation on the tumor cells. It suppressed angiogenesis in chick embryo chorioallantoic membrane (CAM) assay. These results, taken together, indicate Icaritin is a new radio-sensitizer and can enhance anti-cancer effect of IR or other therapies (Hong et al., 2013).

Chronic Myeloid Leukemia (CML)

The mechanism of anti-leukemia for Icaritin is involved in the regulation of Bcr/Abl downstream signaling. Icaritin may be useful for an alternative therapeutic choice of Imatinib-resistant forms of CML. Icaritin potently inhibited proliferation of K562 cells (IC50 was 8 µM) and primary CML cells (IC50 was 13.4 µM for CML-CP and 18 µM for CML-BC), induced CML cells apoptosis, and promoted the erythroid differentiation of K562 cells in a time-dependent manner. Furthermore, Icaritin was able to suppress the growth of primary CD34+ leukemia cells (CML) and Imatinib-resistant cells, and to induce apoptosis (Zhu et al., 2011).

References

Guo YM, Zhang XT, Meng J, Wang ZY. (2011). An anti-cancer agent icaritin induces sustained activation of the extracellular signal-regulated kinase (ERK) pathway and inhibits growth of breast cancer cells. European Journal of Pharmacology, 658(2–3):114–122. doi:10.1016/j.ejphar.2011.02.005.


Hong J, Zhang Z, Lv W, et al. (2013). Icaritin Synergistically Enhances the Radiosensitivity of 4T1 Breast Cancer Cells. PLoS One, 8(8):e71347. doi: 10.1371/journal.pone.0071347.


Huang X, Zhu D, Lou Y. (2007). A novel anti-cancer agent, icaritin, induced cell growth inhibition, G1 arrest and mitochondrial transmembrane potential drop in human prostate carcinoma PC-3 cells. Eur J Pharmacol, 564(1-3):26-36.


Tong JS, Zhang QH, Huang X, et al. (2011). Icaritin Causes Sustained ERK1/2 Activation and Induces Apoptosis in Human Endometrial Cancer Cells. PLoS ONE, 6(3): e16781. doi:10.1371/journal.pone.0016781.


Zhu JF, Li ZJ, Zhang GS, et al. (2011). Icaritin shows potent anti-leukemia activity on chronic myeloid leukemia in vitro and in vivo by regulating MAPK/ERK/JNK and JAK2/STAT3 /AKT signalings. PLoS One, 6(8):e23720. doi: 10.1371/journal.pone.0023720.

I3C

Cancer: Prostate

Action: Inhibits telomerase activity, anti-cancer

Indole-3-carbinol (I3C) is a phytochemical with anti-carcinogenic properties. Telomerase activity is key in carcinogenesis. The effect of I3C on telomerase was investigated in human prostate cancer cell lines LNCaP and PC3. Cells were treated with I3C at 100 and 250 µM with and without 10-50 µM diethylstilbestrol (DES). Telomerase activity was performed using TRAPaze Telomerase Detection Kit, and hTERT gene expression by real time quantitative RT-PCR. I3C (250 µM) inhibited telomerase activity and mRNA expression of hTERT in LNCaP and PC3 cells. I3C at 250 µM combined with any concentration of DES was cytotoxic to LNCaP. Telomerase activity in PC3 cells with 250 µM of I3C and 25 or 50 µM of DES was significantly reduced or inhibited, respectively.

I3C combined with DES reduced PC3 viability and eliminated LNCaP cells. I3C significantly inhibited telomerase activity and hTERT mRNA expression in LNCaP and PC3 cells. Combination of I3C and DES enhanced the inhibitory effect on telomerase activity, gene expression, and cell viability. These results implied that I3C and DES combined might help in prostate cancer treatment (Adler et al., 2011).

Reference

Adler S, Rashid G, Klein A. (2011). Indole-3-carbinol inhibits telomerase activity and gene expression in prostate cancer cell lines. Anti-cancer Res, 31(11):3733-7.

Homoharringtonine/Omacetaxine

Cancer:
Leukemia, AML, CML, myelodysplastic syndrome (MDS)

Action: Induces apoptosis, anti-tumor activity

Homoharringtonine (also known as Omacetaxine mepesuccinate) is isolated from Cephalotaxus harringtonia (K.Koch).

Homoharringtonine/omacetaxine is a unique agent with a long history of research development. It has been recently approved by the Food and Drug Administration for the treatment of chronic myeloid leukemia after failure of 2 or more tyrosine kinase inhibitors. Research with this agent has spanned over 40 years (Kantarjian, O'Brien, & Cortes, 2013).

Leukemia

Homoharringtonine (HHT), first isolated from the Chinese evergreen Cephalotaxus harringtonia, has been demonstrated to have a broad anti-tumor activity in rodents and anti-leukemic effects in humans. It was found that HHT was metabolized to an acid product [HHT acid; 2'hydroxy2' (acetic acid) 6'hydroxy6'methylheptanoyl cephalotaxine] when incubated with either human plasma or mouse plasma in vitro. The HHT concentration inhibiting 50% of the growth of human leukemic HL60 cells was 20 ng/ml, while for HHT acid it was 14,500 ng/ml, indicating that the acid form was more than 700 times less cytotoxic than HHT. The lethal dose of HHT affecting 50%(LD50) of mice was 6.7 mg/kg, but HHT acid produced no apparent toxic effects at doses up to 280 mg/kg (Ni et al., 2003).

Acute Myeloid Leukemia (AML)

The response to remission induction in elderly patients with acute myeloid leukemia (AML) remains poor. Patients were treated with the HA regimen consisting of homoharringtonine (2 mg/m2/day for 7 days) and cytarabine (Ara-C, 100 mg/m2/day for 7 days). The overall response rate was 56.5% with complete remission (CR) rate of 39.1% and partial remission of 17.4%.

There was no early death in this cohort of patients. The estimated median overall survival (OS) time of all patients was (12.0 ± 3.0) months. The estimated OS time of the CR patients was 15 months. The estimated one-year OS rate of all patients treated with HA protocol was (49.3 ± 13.5) %. The estimated one-year OS rate of the CR patients was (62.5 ± 17.1) % (Wang et al., 2009).

Leukemia; Telomerase

The effect of HHT on the telomerase activity and apoptosis of human leukemia HL-60 cells was investigated. Telomerase activity of HL-60 cells was examined by the telomeric repeat amplification protocol (TRAP)–an enzyme-linked immunosorbent assay (ELISA). Apoptosis was analyzed by morphological observation, DNA agarose gel electrophoresis, flow cytometry (FCM), and TdT-mediated dUTP-biotin nick end labeling (TUNEL).

After treatment with HHT at 5-500 microg/l for 48 hours, the level of telomerase activity in HL-60 cells decreased in a dose-and time-dependent manner. Simultaneously, HL-60 cells underwent apoptosis. In conclusion, these data suggest that HHT can inhibit the telomerase content of HL- 60 cells effectively and induce apoptosis (Xie et al., 2006).

Chronic Myeloid Leukemia (CML)

Evidence confirmed HHT as an apoptosis inducer in tumor cell lines and fresh cells from cancer patients. The CR rate reported with HHT-based regimen in acute nonlymphocytic leukemia showed no statistical differences from that with DNR-based regimen, although the case number was limited.

Although with anti-growth activity in vitro and laudable achievement in acute and chronic myeloid leukemia treatment, the drug shows no beneficial effect in lymphocytic leukemia and solid tumors. The underlying mechanism for the discrepancy of efficacy remains unknown, and is a subject for further research (Luo et al., 2004).

Myelodysplastic Syndrome (MDS)

Homoharringtonine might have clinical activity in some patients with myelodysplastic syndrome (MDS) (Daver et al., 2013).

References

Daver N, Vega-Ruiz A, Kantarjian HM, et al. (2013). A phase II open-label study of the intravenous administration of homoharringtonine in the treatment of myelodysplastic syndrome. Eur J Cancer Care, 22(5):605-11. doi: 10.1111/ecc.12065.


Kantarjian HM, O'Brien S, Cortes J. (2013). Homoharringtonine/Omacetaxine mepesuccinate: the long and winding road to food and drug administration approval. Clin Lymphoma Myeloma Leuk, 13(5):530-3. doi: 10.1016/j.clml.2013.03.017.


Luo CY, Tang JY, Wang YP. (2004). Homoharringtonine: a new treatment option for myeloid leukemia. Hematology, 9(4):259-70.


Ni D, Ho DH, Vijjeswarapu M, et al. (2003). Metabolism of homoharringtonine, a cytotoxic component of the evergreen plant Cephalotaxus harringtonia. Journal of Experimental Therapeutics and Oncology, 3(1):47.


Wang J, LŸ S, Yang J, et al. (2009). A homoharringtonine-based induction regimen for the treatment of elderly patients with acute myeloid leukemia: a single center experience from China. Journal of Hematology & Oncology, 2:32. doi:10.1186/1756-8722-2-32


Xie WZ, Lin MF, Huang H, Cai Z. (2006). Homoharringtonine-induced apoptosis of human leukemia HL-60 cells is associated with down-regulation of telomerase. Am J Chin Med, 34(2):233-44.