Category Archives: Chapter 8 Injectables and Cancer Research

Shen qi fu zheng (Vital-qi Fortify)

Cancers: Stomach, lung

Action: Anti-metastatic, chemotherapy support

Ingredients: dang shen (Codonopsis pilosula), huang qi (Astragalus membranaceus).

TCM functions: Invigorating qi and supporting Vital-qi,

Indications: Adjuvant treatment for late-stage lung cancer not suitable for radiotherapy or chemotherapy, of qi deficiency type. It can be combined with chemotherapy for the adjuvant treatment of lung cancer and stomach cancer of qi deficiency type.

Dosage and usage:

Combination with radiotherapy or chemotherapy: Be administrated 3 days before chemotherapy, then be used synchronously with chemotherapy. 1 bottle daily for intravenous drip. The course is the same as that of radiotherapy or chemotherapy.

The clinical efficacy of Shenqifuzheng injection, combined with Bozhi glycopeptide, in treating aged lung cancer patients was observed.

NSCLC; Chemotherapy

In China, Shenqi Fuzheng, a newly developed injection concocted from Chinese medicinal herbs has been reported that may increase efficacy and reduce toxicity when combined with platinum-based chemotherapy, but little is known about it outside of China.

Twenty nine studies were included in this review based on our selection criteria. Of them, ten studies were of high quality and the rest were of low quality, according to the modified Jadad scale. The meta-analysis showed there was a statistically significant higher tumor response when the SFI plus platinum-based chemotherapy treatment group was compared with the platinum-based chemotherapy control group (Dong et al., 2010).

NSCLC

Sixty patients with advanced non-small-cell lung cancer were randomly divided into a treatment group or control group. The treatment group was treated by Shenqi Fuzheng injection combined with chemotherapy, and the control group with chemotherapy alone.

The effect of Shenqi Fuzheng injection combined with chemotherapy vs the chemotherapy alone showed no significant difference. However, in reducing toxicity and side-effects of chemotherapy, such as gastrointestinal reaction and leukopenia, it was superior to the chemotherapy alone (P < 0. 05).

The effect of Shenqi Fuzheng injection combined with chemotherapy in the treatment of advanced non-small-cell lung cancer can improve the quality of life of patients, reduce the toxic side-effects of chemotherapeutic agents, and enhance immunity (Qiao, 2012).

Lung Cancer

In the treatment group; symptoms of fatigue, anorexia, and nausea and vomiting were lower versus the control group. Also, the occurrence of leukopenia and thrombocytopenia in the treatment group was lower than that of the control group.

Shenqi Fuzheng injection plus chemotherapy for advanced lung cancer can reduce drug toxicity, improve the patient”s fatigue, loss of appetite, gastrointestinal symptoms and improve the quality of life of patients (Jiang, 2012).

Lung Cancer; Chemotherapy

Seventy four lung cancer patients diagnosed on their initial visit were chosen, and randomly divided into 3 groups: chemotherapy alone group (control group) of 21 cases, Shenqifuzheng injection combined with chemotherapy (single drug treatment) group of 25 cases, and Shenqifuzheng injection combined with Bozhi glycopeptide and chemotherapy (combined treatment) group of 28 cases.

The difference in short-term  effect, Karnofsky score, and clinical symptoms among the three groups had statistical significance (P < 0.05). The CD4, CDs, CD4/CD8, NK in peripheral blood lymphocytes, and percentage of natural killer cells were decreased significantly after chemotherapy in the control group; while the same indexes in the treatment group with Shenqifuzheng injection and Bozhi glycopeptide and chemotherapy improved significantly (P < 0.05).

Shenqifuzheng injection combined with Bozhi glycopeptide and chemotherapy can relieve adverse reactions in treating aged lung cancer patients. The combined treatment could also enhance the quality of life, immune function, and reduce complications, and thus is worthy of application in clinical settings (Wang, Zhou, Chang & Shao, 2013).

Gastric Cancer

Rats were injected with different dosages of Shenqi Fuzheng injection (0.04 g/m1, 0.08 g/ml and O. 16 g/ml) for 1 week. Matrigel TM test was performed to detect invasion ability of cells, and RT-PCR was used to check Tn-C mRNA expression.

In treated groups, invasiveness of cells was most significant (the inhibit ratio was 34.7% in 0.16 g/ml group). The expressions of Tenascin-C were lower than those of the control. There was significant difference between the high concentration group and the control.

The expressions of Tenascin-C are down-regulated when gastric cancer MGC-803 cells were treated with Shenqi Fuzheng injection at high concentration, therefore the invasiveness of gastric tumors may be reduced (Ma et al., 2013).

Fifty-three patients with advanced gastric cancer were randomly divided into a treatment group (26 cases) or a control group (27 cases). The treatment group received S-1/cisplatin (SP) chemotherapy plus Shenqi-Fuzheng injection, while the control group was treated with SP chemotherapy only. After 2 cycles of chemotherapy, the efficacy, cellular immune function index, and adverse reactions were observed.

Shenqi-Fuzheng injection plus SP chemotherapy can improve cellular immune function, tolerance to chemotherapy, and reduce toxicity in patients with advanced gastric cancer (Yin & Jiang, 2013).

References

Dong, J., Su, S-Y., Wang, M-Y., Zhan, Z. (2010). Shenqi fuzheng, an injection concocted from chinese medicinal herbs, combined with platinum-based chemotherapy for advanced non-small-cell lung cancer: a systematic review. J Exp Clin Cancer Res, 29(1):137. doi:  10.1186/1756-9966-29-137


Jiang, H. (2012). Clinical observation of the Shenqi Fuzheng injection on the response to chemotherapy in advanced lung cancer patients. Zhong Yi Lin Chuang Yan Jiu, 4(14), 11-12.


Ma, J.W., Song, Y.C., Zhang, Y., Jia, Y., Dang, C.X., & Hou, J. (2013). Effects of Shenqi Fuzheng injection on the invasiveness of cells and expression of Tenascin-C in human gastric cancer MGC- 803 cells. Journal of Modern Oncology, 21(2), 263-266.


Qiao, S. (2012). Effect of Shenqi Fuzheng injection combined with chemotherapy in treatment of advanced non small cell lung cancer clinical observation. Chinese Journal of Practical Medicine, 7(34), 25-26.


Wang, D.H., Zhou, L.H., Chang, C., & Shao, N. (2013). Observation of clinical effects of Shenqifuzheng injection combined with Bozhi glycopeptide and chemotherapy in treating aged lung cancer patients. Medical Review, 19(4), 733-735.


Yin, L.L., & Jiang, C.Y. (2013). Observation on the influence of Shenqi-Fuzheng injection on T-lymphocyte subsets, NKcell and the leukocyte of the patients with advanced gastric cancer. International Journal of Traditional Chinese Medicine, 35(1), 22-24.

Qingkailing

Cancer: Leukemia, sarcoma

Action: Antibiotic, anti-apoptotic, anti-inflammatory, neuro-protective, pro-apoptotic, immunomodulating, MMPs regulation

Anti-inflammatory and Immunomodulating

Qingkailing and Shuanghuanglian (SHHL) are two commonly used Chinese herbal preparations with reported anti-inflammatory activity. The effects of these two preparations on the capacity of staphylococcal toxic shock syndrome toxin 1 (TSST-1), to stimulate the production of cytokines (IL-1β, IL-6, TNF-α, IFN-γ) and chemokines (MIP-1α, MIP-1β and MCP-1) by peripheral blood mononuclear cell (PBMC), was tested. Their effect on LPS-stimulated NF-κB transcriptional activity in a THP-1 cell line, and on human monocyte chemotactic response to chemoattractants, was also evaluated.

The results suggested that the pharmacological basis for the anti-inflammatory effects of Qingkailing and SHHL is the result of suppression of NF-κB regulated gene transcription, leading to suppressed production of pro-inflammatory cytokines and chemokines. Interference with leukocyte chemotaxis also contributes to the anti-inflammatory and immunomodulating effects of these medicinals. Identification of the responsible components in these two herbal preparations may yield compounds suitable for structural modification into potent novel drugs (Chen et al., 2002).

Leukemia

The MTT assay, cell morphology, DNA gel electrophoresis, and flow-cytometry were utilized to study the apoptotic effect of Qingkailing, and its active compounds, on the human acute promyelocytic leukemia (HL-60) cell line.

Qingkailing and its active compounds, Baicalin and hyodeoxycholic acid, exhibited strong cytotoxicity in inhibiting HL-60 cells, while Bezoar cholic acid showed a weaker effect. Apoptosis could be induced after being treated for 6 h by the former two compounds, displaying a typical apoptosis peak under flow-cytometry, but could not be induced by the latter.

Qingkailing could induce apoptosis in leukemia cells in vitro, which could serve as a mechanism of Qingkailing in the treatment of acute promyelocytic leukemia (Chen, Dong, & Zhang, 2001).

Qingkailing injection could prevent the decrease of MMP induced by injury of hypoxia-hypoglycemia-reoxygenation, stabilize MMP, inhibit cell apoptosis, and protect hippocampal neurons (Tsing, 2006).

Matrix Metalloproteinases (MMPs) Regulation

Matrix metalloproteinases (MMPs) play vital roles in many pathological conditions, including cancer, cardiovascular disease, arthritis and inflammation. Modulating MMP activity may therefore be a useful therapeutic approach in treating these diseases. Qingkailing is a popular Chinese anti-inflammatory formulation used to treat symptoms such as rheumatoid arthritis, acute hypertensive cerebral hemorrhage, hepatitis and upper respiratory tract infection.

One of the components of Qingkailing, Fructus gardeniae, strongly inhibits MMP activity. The IC50 values for the primary herbal extract and water extract against MMP-16 were 32 and 27 µg/ml, respectively. In addition, the herbal extracts influenced HT1080 human fibrosarcoma cell growth and morphology.

These data may provide molecular mechanisms for the therapeutic effects of Qingkailing and herbal medicinal Fructus gardenia (Yang et al., 2008).

Sources

Chen X, Howard OM, Yang X, Wang L, Oppenheim JJ, Krakauer T. (2002). Effects of Shuanghuanglian and Qingkailing, two multi-components of traditional Chinese medicinal preparations, on human leukocyte function. Life Sciences, 70(24), 2897-2913.


Chen ZT, Dong Q, Zhang L. (2001). Study on the effect of Qingkailing injection and its active principle in inducing cell apoptosis in human acute promyelocytic leukemia. Chinese Journal of Integrated Traditional and Western Medicine, 21(11), 840-842.


Tsing H. (2006). Influences of Qingkailing Injection on neuron apoptosis and mitochondrial membrane potential. Journal of Beijing University of Traditional Chinese Medicine, 2006(2), R285.5.


Yang JG, Shen YH, Hong Y, Jin FH, Zhao SH, Wang MC, Shi XJ,   Fang XX. (2008). Stir-baked Fructus gardeniae (L.) extracts inhibit matrix metalloproteinases and alter cell morphology. Journal of Ethnopharmacology, 117(2), 285-289.

Pseudostellaria heterophylla

A mitogenic fraction (PH-I) separated from Pseudostellaria heterophylla exhibits both immunomodulatory and anti-tumor activities. It was found that fraction PH-I C from P. heterophylla could markedly suppress the growth of EAT cells in vivo. Mechanistic studies have shown that i.p. injection of PH-I C into mice could enhance the phagocytic activity of thioglycollate-elicited peritoneal macrophages.

Moreover, PH-I C showed a potent activating effect on the cytotoxic activity of natural killer (NK) cells and alloreactive cytotoxic T cells (Tc) as well as increased the MurIL-2-induced lymphokine activated killer cell (LAK) activity in vitro. In addition, PH-I C could increase the number of tumor infiltrating lymphocytes (TILs) in the tumor site of WEHI-164-bearing mice. Finally, i.v. injection of PH-I C significantly elevated the levels of IFN-gamma and IL-4 in sera of EAT-bearing mice (Wong et al., 1994).

References

Wong CK, Leung KN, Fung KP, Choy YM. (1994). The immunostimulating activities of anti-tumor polysaccharides from Pseudostellaria heterophylla. Immunopharmacology, 28(1):47-54.

Polyphyllin D

Paris polyphylla is a traditional Chinese medicinal herb that has been used in treating cancer for thousands of years. Polyphyllin D is the steroidal saponin of P. polyphylla.

Breast Cancer

Polyphyllin D elicits apoptosis through mitochondria dysfunction. In vivo study demonstrated that daily administration of Polyphyllin D (2.73 mg/kg body weight) through intravenous injection for ten days in nude mice bearing MCF-7 cells effectively reduced tumor growth for 50% in terms of tumor weight and size, given no significant toxicity in heart and liver to the host. All these findings provide novel insights that Polyphyllin D could serve as a candidate in breast cancer treatment (Lee et al., 2005).

Reference

Lee MS, Yuet-Wa JC, Kong SK, et al. (2005). Effects of Polyphyllin D, a steroidal saponin in Paris polyphylla, in growth inhibition of human breast cancer cells and in xenograft. Cancer Biol Ther, 4(11):1248-54.

Oxymatrine or Compound Matrine (Ku Shen)

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

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

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

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

Indications: Pain and bleeding caused by cancer.

Dosage and usage:

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

Anti-cancer

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

Sarcoma

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

T Cell Leukemia

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

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

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

Osteosarcoma

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

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

Pancreatic Cancer

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

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

Rectal Carcinoma

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

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

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

Gastric Cancer

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

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

Adenoid Cystic Carcinoma

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

Breast Cancer; Chemotherapy

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

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

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

Lung cancer; Pleural Effusion

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

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

Gastric Cancer

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

Colorectal Cancer; Chemotherapy

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

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

NSCLC; Chemotherapy

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

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

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

Lung Adenocarcinoma

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

Liver Cancer

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

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

Chemotherapy

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

Colorectal Cancer, NSCLC, Breast Cancer; Chemotherapy

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

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

References

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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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


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

Oridonin

Cancer: Prostate

Action: Growth arrest, autophagy

To investigate the mechanism of oridonin (ORI)-induced autophagy in prostate cancer PC-3 cells, PC-3 cells cultured in vitro were treated with ORI, and the inhibitory ratio of ORI on PC-3 cells was assayed by 3-4,5- dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide. After ORI treatment, the proliferation of PC-3 cells was inhibited significantly in a concentration and time-dependent manner. SEM examination revealed cellular shrinkage and disappearance of surface microvilli in ORI-treated cells. Under TEM examination, the nuclei exhibited chromatin condensation and the appearance of a large number of autophagosomes with double-membrane structure in cytoplasm. AO staining showed the existence of AVOs. The expression of LC3 and the mRNA level of beclin 1 was increased by ORI. Furthermore, autophagy inhibitor 3-methyladenine reversed the increase of beclin 1 mRNA. The growth of PC-3 cells was inhibited, and autophagy was induced by ORI, indicating ORI may have a potential antitumor effect.

Source
Ye LH, Li WJ, Jiang XQ, et al. Study on the autophagy of prostate cancer PC-3 cells induced by oridonin. Anat Rec (Hoboken). 2012 Mar;295(3):417-22. doi: 10.1002/ar.21528.

 

Cancer: Multiple myeloma

Action: Inhibits proliferation and induces apoptosis

This study was purposed to investigate the antitumor effect of oridonin on human multiple myeloma cell line U266 The results showed that the oridonin obviously inhibited the growth of U266 cell in dose-and time-dependent manners. As for morphological changes, characteristic apoptotic cells presented in U266 cells treated with 10 µmol/L oridonin for 24 hours. The apoptotic rate of U266 cells increased in dose and time dependent manners; after treatment of U266 cells with oridonin the mRNA levels of FGFR3, BCL2, CCND1 and MYC as well as the their protein levels decreased. Occasionally, the oridonin up-regulated the protein levels of P53 in the same manner. It is concluded that the oridonin can exert its anti-tumor effect by inhibiting proliferation and inducing apoptosis of U266 cell in dose dependent and time dependent manners, that maybe give the clues about new program of target therapy for multiple myeloma.

Source:

Duan HQ, Li MY, Gao L, et al. Mechanism concerning antitumor effect of oridonin on multiple myeloma cell line U266. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2014 Apr;22(2):364-9. doi: 10.7534/j.issn.1009-2137.2014.02.018.

Cancer: Multiple myeloma

Action: Induces apoptosis and autophagy

Exposure to oridonin (1-64 μmol/L) inhibited the proliferation of RPMI8266 cells in a concentration-dependent manner with an IC(50) value of 6.74 μmol/L. Exposure to oridonin (7 μmol/L) simultaneously induced caspase 3-mediated apoptosis and Beclin 1-dependent autophagy of RPMI8266 cells. Both the apoptosis and autophagy were time-dependent, and apoptosis was the main effector pathway of cell death. Exposure to oridonin (7 μmol/L) increased intracellular ROS and reduced SIRT1 nuclear protein in a time-dependent manner.

Oridonin simultaneously induces apoptosis and autophagy of human multiple myeloma RPMI8266 cells via regulation of intracellular ROS generation and SIRT1 nuclear protein. The cytotoxicity of oridonin is mainly mediated through the apoptotic pathway, whereas the autophagy protects the cells from apoptosis.

Source

Zeng R, Chen Y, Zhao S, Cui GH.Autophagy counteracts apoptosis in human multiple myeloma cells exposed to oridonin in vitro via regulating intracellular ROS and SIRT1. Acta Pharmacol Sin. 2012 Jan;33(1):91-100. doi: 10.1038/aps.2011.143.

Cancer: Prostate, acute promyelocytic leukemia, breast, non-small-cell lung (NSCL), Ehrlich ascites, P388 lymphocytic leukemia, colorectal., ovarian, esphageal

Action: Chemoresistance, Ara-C, VP-16 

Cancer cell arises in part through the acquisition of apoptotic resistance. Leukemia cells resistant to chemotherapy-induced apoptosis have been found to be sensitive to oridonin, a natural agent with potent anticancer activity. Weng et al., (2014) compared the response of human leukemia cells with oridonin and the antileukemia drugs Ara-C and VP-16. Compared with HL60 cells, K562 and K562/ADR cells displayed resistance to apoptosis stimulated by Ara-C and VP-16 but sensitivity to oridonin. Mechanistic investigations revealed that oridonin upregulated BIM-S by diminishing the expression of miR-17 and miR-20a, leading to mitochondria-dependent apoptosis. In contrast, neither Ara-C nor VP-16 could reduce miR-17 and miR-20a expression or could trigger BIM-S–mediated apoptosis.

Notably, silencing miR-17 or miR-20a expression by treatment with microRNA (miRNA; miR) inhibitors or oridonin restored sensitivity of K562 cells to VP-16. Synergistic effects of oridonin and VP-16 were documented in cultured cells as well as mouse tumor xenograft assays. Inhibiting miR-17 or miR-20a also augmented the proapoptotic activity of oridonin. Taken together, our results identify a miRNA-dependent mechanism underlying the anticancer effect of oridonin and provide a rationale for its combination with chemotherapy drugs in addressing chemoresistant leukemia cells.

Reference

Weng Hy, Huang Hl, Dong B, et al. Inhibition of miR-17 and miR-20a by Oridonin Triggers Apoptosis and Reverses Chemoresistance by Derepressing BIM-S. Cancer Res; 74(16); 1–11. doi: 10.1158/0008-5472.CAN-13-1748

Action: Induces apoptosis

Oridonin is a tetracycline diterpenoid isolated from the plant Rabdosia rubescens (RR) [(Hemsl.). Hara (Lamiaceae)] (dong ling cao) is a Chinese medicinal herb used widely in provinces including Henan. The aerial parts of RR and other species of the same genus has been reported to have the functions of clearing “heat” and “toxicity”, nourishing “yin”, removing “blood stasis”, and relieving swelling. RR has been used to treat stomach-ache, sore throat and cough.

Gastric Cancer, Esophageal Cancer, Liver Cancer, Prostate Cancer

RR and its extracts have been shown to be able to suppress disease progress, reduce tumor burden, alleviate syndrome and prolong survival in patients with gastric carcinoma, esophageal., liver and prostate cancers (Tang & Eisenbrand, 1992). Interestingly, other Isodon plants including Isodon japonicus Hara (IJ) and I. trichocarpus (IT) are also applied as home remedies for similar disorders in Japan and Korea.

Induces Apoptosis

These reports suggest that Isodon plants should have at least one essential anti-tumor component. In the 1970s, a bitter tetracycline diterpenoid compound, oridonin, was isolated from RR, IJ, and IT separately, and was shown to be a potent apoptosis inducer in a variety of cancer cells (Fujita et al., 1970; Fujita et al., 1976; Henan Medical Institute, 1978; Fujita et al., 1988).

Anti-cancer

There is currently research being undertaken regarding the relationship between the chemical structure/modifications and the molecular mechanisms underlying its anti-cancer activity, such as suppression of tumor proliferation and induction of tumor cell death, and the cell signal transduction in anti-cancer activity of oridonin (Zhang et al., 2010).

Prostate Cancer, Breast Cancer, NSCLC, Leukemia, Glioblastoma

Oridonin has been found to effectively inhibit the proliferation of a wide variety of cancer cells including those from prostate (LNCaP, DU145, PC3), breast (MCF-7, MDA-MB231), non-small-cell lung (NSCL) (NCI-H520, NCI-H460, NCI-H1299) cancers, acute promyelocytic leukemia (NB4), and glioblastoma multiforme (U118, U138).

Oridonin induced apoptosis and G0/G1 cell-cycle arrest in LNCaP prostate cancer cells. In addition, expression of p21waf1 was induced in a p53-dependent manner. Taken together, oridonin inhibited the proliferation of cancer cells via apoptosis and cell-cycle arrest with p53 playing a central role in several cancer types which express the wild-type p53 gene. Oridonin may be a novel, adjunctive therapy for a large variety of malignancies (Ikezoe et al., 2003).

Breast Cancer; Anti-metastatic

According to the flow cytometric analysis, oridonin suppressed MCF-7 cell growth by cell-cycle arrest at the G2/M phase and caused accumulation of MDA-MB-231 cells in the Sub-G1 phase. The induced apoptotic effect of oridonin was further confirmed by a morphologic characteristics assay and TUNEL assay. Meanwhile, oridonin significantly suppressed MDA-MB-231 cell migration and invasion, decreased MMP-2/MMP-9 activation and inhibited the expression of Integrin β1 and FAK. In conclusion, oridonin inhibited growth and induced apoptosis in breast cancer cells, which might be related to DNA damage and activation of intrinsic or extrinsic apoptotic pathways. Moreover, oridonin also inhibited tumor invasion and metastasis in vitro possibly via decreasing the expression of MMPs and regulating the Integrin β1/FAK pathway in MDA-MB-231 cells (Wang et al., 2013).

Gastric Cancer

The inhibitory effect of oridonin on gastric cancer HGC-27 cells was detected using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. After treated with oridonin (0, 1.25, 2.5, 5 and 10 µg/mL), HGC-27 cells were collected for anexin V-phycoerythrin and 7-amino-actinomycin D double staining and tested by flow cytometric analysis, and oridonin- induced apoptosis in HGC-27 cells was detected.

Oridonin significantly inhibited the proliferation of HGC-27 cells in a dose- and time-dependent manner. The inhibition rates of HGC-27 treated with four different concentrations of oridonin for 24 h (1.25, 2.5, 5 and 10 µg/mL) were 1.78% ± 0.36%, 4.96% ± 1.59%, 10.35% ± 2.76% and 41.6% ± 4.29%, respectively, which showed a significant difference (P < 0.05. Cells treated with oridonin showed typical apoptotic features with acridine orange/ethidium bromide staining. After treatment with oridonin, the cells became round, shrank, and developed small buds around the nuclear membrane while forming apoptotic bodies. However, the change in the release of LDH caused by necrosis was insignificant, suggesting that the major cause of oridonin-induced HGC-27 cell death was apoptosis. Flow cytometric analysis also revealed that oridonin induced significant apoptosis compared with the controls (P < 0.05).

Apoptosis of HGC-27 induced by oridonin may be associated with differential expression of Apaf-1, caspase-3 and cytochrome c, which are highly dependent upon the mitochondrial pathway (Sun et al., 2012).

Ehrlich Ascites, Leukemia

Oridonin has been found to also increase lifespan of mice bearing Ehrlich ascites or P388 lymphocytic leukemia. Oridonin triggered apoptosis in more than 50% of t(8;21) leukemic cells in vitro at concentration of 2 M or higher accompanied by degradation of AE oncoprotein, and showed significant anti-leukemia efficacies with low adverse effects in vivo. These data suggest possible beneficial effects for patients with t(8;21) acute myeloid leukemia (AML) (Zhou et al., 2007).

Prostate Cancer, Breast Cancer, Ovarian Cancer

Oridonin exhibited anti-proliferative activity toward all cancer cell lines tested, with an IC50 estimated by the MTT cell viability assay ranging from 5.8+/-2.3 to 11.72+/-4.8 microM. The increased incidence of apoptosis, identified by characteristic changes in cell morphology, was seen in tumor lines treated with oridonin. Notably, at concentrations that induced apoptosis among tumor cells, oridonin failed to induce apoptosis in cultures of normal human fibroblasts. Oridonin up-regulated p53 and Bax and down-regulated Bcl-2 expression in a dose-dependent manner and its absorption spectrum was measured in the presence and absence of double stranded (ds) DNA. Oridonin inhibits cancer cell growth in a cell-cycle specific manner and shifts the balance between pro- and anti-apoptotic proteins in favor of apoptosis. The present data suggest that further studies are warranted to assess the potential of oridonin in cancer prevention and/or treatment (Chen et al., 2005).

Ovarian Cancer Stem Cells; Chemotherapy Resistance

Oridonin was suggested to suppress ovarian CSCs as is reflected by down-regulation of the surface marker EpCAM. Unlike NSAIDS (non-steroid anti-inflammatory drugs), well documented clinical data for phyto-active compounds are lacking. In order to evaluate objectively the potential benefit of these types of compounds in the treatment of ovarian cancer, strategically designed, large scale studies are warranted (Chen et al., 2012).

Colorectal Cancer

Oridonin induced potent growth inhibition, cell-cycle arrest, apoptosis, senescence and colony-forming inhibition in three colorectal cancer cell lines in a dose-dependent manner in vitro. Daily i.p. injection of oridonin (6.25, 12.5 or 25 mg/kg) for 28 days significantly inhibited the growth of SW1116 s.c. xenografts in BABL/C nude mice.

Oridonin possesses potent in vitro and in vivo anti-colorectal cancer activities that correlated with induction of histone hyperacetylation and regulation of pathways critical for maintaining growth inhibition and cell-cycle arrest. Therefore, oridonin may represent a novel therapeutic option in colorectal cancer treatment as it has been shown to induce apoptosis and senescence of colon cancer cells in vitro and in vivo (Gao et al., 2010).

Colon Cancer; Apoptosis

Oridonin increased intracellular hydrogen peroxide levels and reduced the glutathione content in a dose-dependent manner. N-acetylcysteine, a reactive oxygen species scavenger, not only blocked the oridonin-induced increase in hydrogen peroxide and glutathione depletion, but also blocked apoptosis and senescence induced by oridonin.

Moreover, exogenous catalase could inhibit the increase in hydrogen peroxide and apoptosis induced by oridonin, but not the glutathione depletion and senescence. Furthermore, thioredoxin reductase (TrxR) activity was reduced by oridonin in vitro and in cells, which may cause the increase in hydrogen peroxide. In conclusion, the increase in hydrogen peroxide and glutathione depletion account for oridonin-induced apoptosis and senescence in colorectal cancer cells, and TrxR inhibition is involved in this process.

Given the importance of TrxR as a novel cancer target in colon cancer, oridonin would be a promising clinical candidate (Gao et al., 2012).

Prostate Cancer; Apoptosis

Oridonin (ORI) could inhibit the proliferation and induce apoptosis in various cancer cell lines. After ORI treatment, the proliferations of human prostate cancer (HPC) cell lines PC-3 and LNCaP were inhibited in a concentration and time-dependent manner. ORI induced cell-cycle arrest at the G2/M phase. Autophagy occurred before the onset of apoptosis and protected cancer cells in ORI-treated HPC cells. P21 was involved in ORI-induced autophagy and apoptosis (Li et al., 2012).

References

Chen S, Gao J, Halicka HD, et al. (2005). The cytostatic and cytotoxic effects of oridonin (Rubescenin), a diterpenoid from Rabdosia rubescens, on tumor cells of different lineage. Int J Oncol, 26(3):579-88.

 

Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.

 

Fujita E, Fujita T, Katayama H, Shibuya M. (1970). Terpenoids. Part XV. Structure and absolute configuration of oridonin isolated from Isodon japonicus trichocarpus. J Chem Soc (Chem Comm), 21:1674–1681

 

Fujita E, Nagao Y, Node M, et al. (1976). Anti-tumor activity of the Isodon diterpenoids: structural requirements for the activity. Experientia, 32:203–206.

 

Fujita T, Takeda Y, Sun HD, et al. (1988). Cytotoxic and anti-tumor activities of Rabdosia diterpenoids. Planta Med, 54:414–417.

 

Henan Medical Institute, Henan Medical College, Yunnan Institute of Botany. (1978). Oridonin–a new anti-tumor subject. Chin Science Bull, 23:53–56.

 

Ikezoe T, Chen SS, Tong XJ, et al. (2003). Oridonin induces growth inhibition and apoptosis of a variety of human cancer cells. Int J Oncol, 23(4):1187-93.

 

Gao FH, Hu XH, Li W, Liu H, et al. (2010). Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc. BMC Cancer, 10:610. doi: 10.1186/1471-2407-10-610.

 

Gao FH, Liu F, Wei W, et al. (2012). Oridonin induces apoptosis and senescence by increasing hydrogen peroxide and glutathione depletion in colorectal cancer cells. Int J Mol Med, 29(4):649-55. doi: 10.3892/ijmm.2012.895.

 

Li X, Li X, Wang J, Ye Z, Li JC. (2012) Oridonin up-regulates expression of P21 and induces autophagy and apoptosis in human prostate cancer cells. Int J Biol Sci. 2012;8(6):901-12. doi: 10.7150/ijbs.4554.

 

Sun KW, Ma YY, Guan TP, et al. (2012). Oridonin induces apoptosis in gastric cancer through Apaf-1, cytochrome c and caspase-3 signaling pathway. World J Gastroenterol, 18(48):7166-74. doi: 10.3748/wjg.v18.i48.7166.

 

Tang W, Eisenbrand G. (1992). Chinese drugs of plant origin: chemistry, pharmacology, and use in traditional and modern medicine. Berlin: Springer-Verlag, 817–847.

 

Wang S, Zhong Z, Wan J, et al. (2013). Oridonin induces apoptosis, inhibits migration and invasion on highly-metastatic human breast cancer cells. Am J Chin Med, 41(1):177-96. doi: 10.1142/S0192415X13500134.

 

Zhang Wj, Huang Ql, Hua Z-C. (2010). Oridonin: A promising anti-cancer drug from China. Frontiers in Biology, 5(6):540-545.

 

Zhou G-B, Kang H, Wang L, et al. (2007). Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent anti-tumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood, 109(8):3441-3450.

Luteolin

Cancer: Colorectal., pancreatic, ovarian, breast

Action: Anti-inflammatory, radio-protective, TAM chemo-sensitizer

Luteolin is a flavonoid found in many plants and foods, including Terminalia chebula (Retz.), Prunella vulgaris (L.) and Perilla frutescens [(L.) Britton].

Luteolin is contained in Ocimum sanctum L. or Ocimum tenuiflorum L, commonly known as Holy Basil in English or Tulsi in various Indian languages; it is an important medicinal plant in the various traditional and folk systems of medicine in Southeast Asia. Scientific studies have shown it to possess anti-inflammatory, anti-analgesic, anti-pyretic, anti-diabetic, hepato-protective, hypolipidemic, anti-stress, and immunomodulatory activities. It has been found to prevent chemical-induced skin, liver, oral., and lung cancers and mediates these effects by increasing the anti-oxidant activity, altering the gene expressions, inducing apoptosis, and inhibiting angiogenesis and metastasis.

Radio-protective

The aqueous extract of Tulsi has been shown to protect mice against γ-radiation-induced sickness and mortality and to selectively protect the normal tissues against the tumoricidal effects of radiation. The chemo-preventive and radio-protective properties of Tulsi emphasize aspects that warrant future research to establish its activity and utility in cancer prevention and treatment (Baliga et al., 2013).

Anti-inflammatory

Pre-treatment of RAW 264.7 with luteolin, luteolin-7-glucoside, quercetin, and the isoflavonoid genistein inhibited both the LPS-stimulated TNF-αand interleukin-6 release, whereas eriodictyol and hesperetin only inhibited TNF-αrelease. From the compounds tested luteolin and quercetin were the most potent in inhibiting cytokine production with an IC50 of less than 1 and 5 µM for TNF-αrelease, respectively. Pre-treatment of the cells with luteolin attenuated LPS-induced tyrosine phosphorylation of many discrete proteins. Luteolin inhibited LPS-induced phosphorylation of Akt. Treatment of macrophages with LPS resulted in increased IκB-αphosphorylation and reduced the levels of IκB-α. It was concluded that luteolin inhibits protein tyrosine phosphorylation, nuclear factor-κB-mediated gene expression and pro-inflammatory cytokine production in murine macrophages (Xagorari et al., 2001).

Luteolin (Lut) possesses significant anti-inflammatory activity in well established models of acute and chronic inflammation, such as xylene-induced ear edema in mice (ED50= 107 mg/ kg), carrageenin-induced swellingof the ankle, acetic acid-induced pleurisy and croton oil-induced gaseous pouch granuloma in rats. Its combined immunostimulatory and anti-inflammatory activity, and inhibitory effect upon immediate hypersensitive response provide the pharmacologic bases for the beneficial effects of Lut in the treatment of chronic bronchitis (Chen et al., 1986).

Anti-inflammatory; Lung

Luteolin dose-dependently inhibited the expression and production of nitric oxide (NO) and prostaglandin E2 (PGE2), as well as the expression of inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6). Luteolin also reduced the DNA binding activity of nuclear factor-kappa B (NF-κB) in LPS-activated macrophages. Moreover, luteolin blocked the degradation of IκB-α and nuclear translocation of NF-κB p65 subunit.

In sum, these data suggest that, by blocking NF-κ>B and AP-1 activation, luteolin acts to suppress the LPS-elicited inflammatory events in mouse alveolar macrophages, and this effect was mediated, at least in part, by inhibiting the generation of reactive oxygen species. These observations suggest a possible therapeutic application of this agent for treating inflammatory disorders in the lung (Chen et al., 2007).

Anti-inflammatory; Neuroinflammation

Pre-treatment of primary murine microglia and BV-2 microglial cells with luteolin inhibited LPS-stimulated IL-6 production at both the mRNA and protein levels. Whereas luteolin had no effect on the LPS-induced increase in NF-κB DNA binding activity, it markedly reduced AP-1 transcription factor binding activity. To determine whether luteolin might have similar effects in vivo, mice were provided drinking water supplemented with luteolin for 21 days and then they were injected i.p. with LPS. Luteolin consumption reduced LPS-induced IL-6 in plasma 4 hours after injection. Taken together, these data suggest luteolin inhibits LPS-induced IL-6 production in the brain by inhibiting the JNK signaling pathway and activation of AP-1 in microglia. Thus, luteolin may be useful for mitigating neuroinflammation (Jang et al., 2008).

Colon Cancer

Activities of CDK4 and CDK2 decreased within 2 hours after luteolin treatment, with a 38% decrease in CDK2 activity (P < 0.05) observed in cells treated with 40 µmol/l luteolin. Luteolin inhibited CDK2 activity in a cell-free system, suggesting that it directly inhibits CDK2.

tLuteolin promoted G2/M arrest at 24 hours post-treatment  by down-regulating cyclin B1 expression and inhibiting cell division cycle (CDC)2 activity. Luteolin promoted apoptosis with increased activation of caspases 3, 7, and 9 and enhanced poly(ADP-ribose) polymerase cleavage and decreased expression of p21CIP1/WAF1, survivin, Mcl-1, Bcl-xL, and Mdm-2. Decreased expression of these key antiapoptotic proteins could contribute to the increase in p53-independent apoptosis that was observed in HT-29 cells. Lim et al., (2007) demonstrated that luteolin promotes both cell-cycle arrest and apoptosis in the HT-29 colon cancer cell line, providing insight about the mechanisms underlying its anti-tumorigenic activities.

Pancreatic Cancer; Chemotherapy

Simultaneous treatment or pre-treatment (0, 6, 24 and 42 hours) of flavonoids and chemotherapeutic drugs and various concentrations (0-50µM) were assessed using the MTS cell proliferation assay. Simultaneous treatment with either flavonoid (0,13, 25 or 50µM) and chemotherapeutic drugs 5-fluorouracil (5-FU, 50µM) or gemcitabine (Gem, 10µM) for 60h resulted in less-than-additive effect (p<0.05). Pre-treatment for 24 hours with 13µM of either Api or Lut, followed by Gem for 36 hours was optimal to inhibit cell proliferation.

Pre-treatment of cells with 11-19µM of either flavonoid for 24 hours resulted in 59-73% growth inhibition when followed by Gem (10µM, 36h). Lut (15µM, 24h) Pre-treatment followed by Gem (10µM, 36h), significantly decreased protein expression of nuclear GSK-3βand NF-κB p65 and increased pro-apoptotic cytosolic cytochrome c. Pre-treatment of human pancreatic cancer cells BxPC-3 with low concentrations of Lut effectively aid in the anti-proliferative activity of chemotherapeutic drugs (Johnson et al., 2013).

Ovarian Cancer

Luteolin has been found to repress NF-kappaB (NF-κ>B, a pro-inflammatory transcription factor) and inhibit pro-inflammatory cytokines such as TNF-αand IL-6. Additionally, it has been shown to stabilize p53 protein, sensitize TRAIL (TNF receptor apoptosis-inducing ligand) induced apoptosis, and prevent or delay chemotherapy-resistance.

Recent studies further indicate that luteolin potently inhibits VEGF production and suppresses ovarian cancer cell metastasis in vitro. Lastly, oridonin and wogonin were suggested to suppress ovarian CSCs as is reflected by down-regulation of the surface marker EpCAM. Unlike NSAIDS (non-steroid anti-inflammatory drugs), well documented clinical data for phyto-active compounds are lacking. In order to evaluate objectively the potential benefit of these compounds in the treatment of ovarian cancer, strategically designed, large scale studies are warranted (Chen et al., 2012).

Chemo-sensitizer

The sensitization effect of luteolin on cisplatin-induced apoptosis is p53 dependent, as such effect is only found in p53 wild-type cancer cells but not in p53 mutant cancer cells. Moreover, knockdown of p53 by small interfering RNA made p53 wild-type cancer cells resistant to luteolin and cisplatin. Second, Shi et al., (2007) observed a significant increase of p53 protein level in luteolin-treated cancer cells without increase of p53 mRNA level, indicating the possible effect of luteolin on p53 posttranscriptional regulation.

In summary, data from this study reveal a novel molecular mechanism involved in the anti-cancer effect of luteolin and support its potential clinical application as a chemo-sensitizer in cancer therapy.

Breast Cancer; TAM Chemo-sensitizer

This study found that the level of cyclin E2 (CCNE2) mRNA was higher in tumor cells (4.89-fold, (∗)P=0.005) than in normal paired tissue samples as assessed using real-time reverse-transcriptase polymerase chain reaction (RT-PCR) analysis (n=257). Further, relatively high levels of CCNE2 protein expression were detected in tamoxifen-resistant (TAM-R) MCF-7 cells.

These results showed that the level of CCNE2 protein expression was specifically inhibited in luteolin-treated (5µM) TAM-R cells, either in the presence or absence of 4-OH-TAM (100nM). Combined treatment with 4-OH-TAM and luteolin synergistically sensitized the TAM-R cells to 4-OH-TAM. The results of this study suggest that luteolin can be used as a chemo-sensitizer to target the expression level of CCNE2 and that it could be a novel strategy to overcome TAM resistance in breast cancer patients (Tu et al., 2013).

References

Baliga MS, Jimmy R, Thilakchand KR, et al. (2013). Ocimum sanctum L (Holy Basil or Tulsi) and its phytochemicals in the prevention and treatment of cancer. Nutr Cancer, 65(1):26-35. doi: 10.1080/01635581.2013.785010.


Chen CY, Peng WH, Tsai KD and Hsu SL. (2007). Luteolin suppresses inflammation-associated gene expression by blocking NF-κB and AP-1 activation pathway in mouse alveolar macrophages. Life Sciences, 81(23-24):1602-1614. doi:10.1016/j.lfs.2007.09.028


Chen MZ, Jin WZ, Dai LM, Xu SY. (1986). Effect of luteolin on inflammation and immune function. Chinese Journal of Pharmacology and Toxicology, 1986-01.


Chen SS, Michael A, Butler-Manuel SA. (2012). Advances in the treatment of ovarian cancer: a potential role of anti-inflammatory phytochemicals. Discov Med, 13(68):7-17.


Jang S, Kelley KW, Johnson RW. (2008). Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. PNAS, 105(21):7534-7539


Johnson JL, Gonzalez de Mejia E. (2013). Interactions between dietary flavonoids apigenin or luteolin and chemotherapeutic drugs to potentiate anti-proliferative effect on human pancreatic cancer cells, in vitro. Food Chem Toxicol, S0278-6915(13)00491-2. doi: 10.1016/j.fct.2013.07.036.


Lim DY, Jeong Y, Tyner Al., Park JHY. (2007). Induction of cell-cycle arrest and apoptosis in HT-29 human colon cancer cells by the dietary compound luteolin. Am J Physiol Gastrointest Liver Physiol, 292: G66-G75. doi:10.1152/ajpgi.00248.2006.


Shi R, Huang Q, Zhu X, et al. (2007). Luteolin sensitizes the anti-cancer effect of cisplatin via c-Jun NH2-terminal kinase-mediated p53 phosphorylation and stabilization. Molecular Cancer Therapeutics, 6(4):1338-1347. doi: 10.1158/1535-7163.MCT-06-0638.


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


Xagorari A, Papapetropoulos A, Mauromatis A, et al. (2001). Luteolin inhibits an endotoxin-stimulated phosphorylation cascade and pro-inflammatory cytokine production in macrophages. JPET, 296(1):181-187.

Kanglaite injection (KLT)

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

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

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

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

Dosage and usage:

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

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

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

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

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

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

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

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

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

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

Lung Cancer

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

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

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

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

Skin Keratinocytes

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

Hepatocellular Carcinoma

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

Glomerular Nephritis

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

Lung Metastasis

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

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

Lung Cancer; Chemo Side Effects

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

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

Apoptosis

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

References

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


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


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


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


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


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


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


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


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

Kangai Injection

Cancers: Cervical., lung, non-Hodgkin”s lymphoma, stomach

Action: Anti-proliferative, chemotherapy support, immunomodulary, radio-sensitizer

Non-Hodgkin’s Lymphoma

The influence of Kangai injection on blood serum vascular endothelial growth factor of non-Hodgkin”s lymphoma patients, and its synergistic effect, attenuation and improvement of quality of life was evaluated.

Eighty-five non-Hodgkin”s lymphoma patients were randomized into a treatment group or control group. The patients in the treatment group were treated by Kangai injection and cyclophosphamide / doxorubicin / vincristine / prednisolone (CHOP) combined chemotherapy, while those in the control group were treated by CHOP chemotherapy only.

The concentration of vascular endothelial growth factor in blood serum of the patients of the treatment group decreased after therapy (P < 0.05), acute curative effect gradually increased, quality of life was raised significantly (P < 0.05), and adverse reactions of the combined chemotherapy decreased markedly (P < 0.05).

Kangai injection, with CHOP chemotherapy, has a synergistic effect. It can attenuate progression of non-Hodgkin”s lymphoma, and improve quality of life. Additionally, it can decrease the concentration of serum vascular endothelial growth (Tang, 2006).

Stomach Cancer; Chemotherapy

Eighty patients with advanced stomach cancer were randomly divided into treatment group (chemotherapy+ GAMA injection) and control group (chemotherapy only). Observation was conducted on cellular immunization, short-termeffect, quality of life improvement, and toxic side-effects in both groups.

In the treatment group, both NK cellular activity and CD4/CD8 ratios were higher after the treatment (P < 0.01). CD3 and CD4 were both increased (P < 0.05). In the control group, the NK cellular activity, CD3, CD4, CD4/CD8 ratio were all lower after the treatment (P < 0.05). The short-term  efficacy rate was 45% in the treatment group and 40% in the control group. The difference was not significant. The treatment group was apparently lower than the control group in leukopenia, nausea and/or vomiting, and peripheral nerve toxicity (P < 0.05). Compared with the control group, less fatigue, better appetite, and Karnofsky score increases were observed in the treatment group (P < 0.01). The treatment group was also more effective in relieving pain and promoting weight gain than the control group (P < 0.05).

Treating advanced stomach cancer, with the combination of Kangai injection and chemotherapy, may decrease the adverse effects of chemotherapy on patients′cellular immune functions and other side effects, and thereby, improve the quality of life of patients (Wu & Yang, 2007).

NSCLC; Chemotherapy

Seventy eight patients with stage IIIB/IV NSCLC were randomly divided into two groups: treatment group (n=40) received GAMA injection and chemotherapy, and control group (n=38) only received chemotherapy.

The short-termeffect, Karnofsky scores of life quality, and the incidence of pancytopenia in treatment group were superior to those in the control group (72.5% vs 47.4%, P<0.05; 87.5% vs 55.3%, P < 0.01; P < 0.01).

Kangai injection can improve the short-term effect, quality of life, and pancytopenia prevalence in patients with intermediate and advanced-stage NSCLC (Wen, Xie, Xie & Feng, 2006).

Radiotherapy side-effects

One hundred ten cases of patients with malignant tumors wasrandomly divided into the treatment group or the control group. The treatment group was given Kangai injection for 40 days after radiotherapy, while the control group was treated by radiotherapy only.

Tumor growth in the treatment group and the control group were 66.7% and 43.4%, respectively. Karnofsky score improvements were 52.6% and 32.1%, respectively. The incidence of leukopenia was 22.8% and 42.5%, respectively. All differences were significant (P < 0.05). There was no significant difference in levels of lymphocytres between the treatment group before and after therapy (P > 0.05). However, there was significant difference in the control group before and after therapy (P< 0.05).

Kangai injection can improve the curative effect and alleviate the side-effects of radiotherapy on treating malignant tumors (Cao et al., 2005).

Leukemia

Kangai injection combination of fludarabine (Flud), cytosine arabinoside (Ara-C), and granulocyte colony-stimulating factor (G-CSF) (FLAG) in refractory/relapsed acute leukemia (AL) patients was investigated. The remission rate of treatment and total effective rate treatment group were 57.1% (16/28) and 71.4% (21/28), the control group were 52.3% (11/21) and 61.9% (13/21); there were no significant differences in the two groups. Duration of neutrophils less than 0.5 x 10(9)/L in treatment group was (14 +/- 6) day, control group was (23 +/- 3) day, Duration of platelet less than 25 x 10(9)/L in treatment group was (17 +/- 6) day, control group was (31 +/- 2) day, treatment group of III-IV degree of infection was 6.9% (1/28) and control group was 23.8% (5/21) between the two groups were significantly different (P < 0.05). treatment group of III- IV degree of gastrointestinal; toxicity was 10.7% (3/28) and control group was 28. 5% (6/ 21).

Kangai injection plus FLAG regimen could increase the remission rate, shorten the period of bone marrow suppression, significantly reduced the incidence and degree of infection, play an important role in attenuated efficiency (Wan et al., 2011).

References

Cao, H. (2005). Treating 57 cases of malignant tumor by Kangai injection and radiotherapy. Zhejiang Journal of Integrated Traditional Chinese and Western Medicine, 2005(12), R730.5. doi: cnki:sun:zjzh.0.2005-12-005.


Tang, Q. (2006). Influence of Kangai injection on blood serum vascular endothelial growth factor of non-Hodgkin lymphoma patient. Journal of Leukemia & Lymphoma, 15(1).


Wan, Q., Xi, A., Zhang, C., Liu X.(2011) Clinical study of kangai injection plus FLAG regimen for refractory/relapsed acute leukemia. Zhongguo Zhong Yao Za Zhi, 36(22):3207-9.


Wen, J.Y., Xie, Z., Xie, J.R., & Feng, L.P. (2006). Kangai injection mixed with chemotherapy in intermediate and advanced-stage non-small-cell lung cancer. Journal of Guandong Medical College, 24(1), 1005-4057.


Wu, L., & Yang, Y. (2007). A clinical study of treating advanced gastric cancer with the combination of Kangai injection and chemotherapy. Proceeding of Clinical Medicine, 18(7), 1671-8631.

Isorhamnetin

Cancer:
Lung, colon, acute myeloid leukemia, T lymphoma, Ehrlich carcinoma, gastric, esophageal squamous cell, chronic myelogenous leukemia

Action: Dox-induced cardiotoxicity, anti-oxidant

Isorhamnetin, the anti-tumor component of Hippophae rhamnoides Linn, is also a member of the ßavonoid class of compounds. Its chemical name is 3,5,7-trihydroxy-2-(4-hydroxy-3-methoxyphenyl) chromen-4-one and its molecular formula is C16H12O7.

Lung Cancer

Isorhamnetin shows good inhibitory effects on human lung adenocarcinoma A549 cells, human colon cancer HT-29 cells, human chronic myeloid leukemia K562 cells, human acute myeloid leukemia HL-60 cells, mouse T lymphoma YAC-1 cells and mouse Ehrlich carcinoma. In terms of its mechanism of action, it seems that isorhamnetin simultaneously reduces the expression of Bcl-2 and increases the expression of Bax, which activates caspase-9 and its downstream factor caspase-3, thus resulting in cell death (Zhu et al. 2005).

Colorectal Cancer

It was demonstrated that isorhamnetin prevents colorectal tumorigenesis. Dietary isorhamnetin decreased mortality, tumor number, and tumor burden by 62%, 35%, and 59%, respectively. Magnetic resonance imaging, histopathology, and immunohistochemical analysis revealed that dietary isorhamnetin resolved the DSS-induced inflammatory response faster than control diet.

These observations suggest the chemo-protective effects of isorhamnetin in colon cancer are linked to its anti-inflammatory activities and its inhibition of oncogenic Src activity and consequential loss of nuclear β-catenin, activities that are dependent on CSK expression (Saud et al., 2013).

Gastric Cancer

The potential effects of isorhamnetin (IH), a 3'-O-methylated metabolite of quercetin, were investigated on the peroxisome proliferator-activated receptor γ (PPAR-γ) signaling cascade using proteomics technology platform, gastric cancer (GC) cell lines, and xenograft mice model.

It was observed that IH exerted a strong anti-proliferative effect and increased cytotoxicity in combination with chemotherapeutic drugs. IH also inhibited the migratory/invasive properties of gastric cancer cells, which could be reversed in the presence of PPAR-γ inhibitor.

Using molecular docking analysis, Ramachandran et al. (2013) demonstratd that IH formed interactions with seven polar residues and six nonpolar residues within the ligand-binding pocket of PPAR-γ that are reported to be critical for its activity and could competitively bind to PPAR-γ. IH significantly increased the expression of PPAR-γ in tumor tissues obtained from xenograft model of GC. Overall, these findings clearly indicate that anti-tumor effects of IH may be mediated through modulation of the PPAR-γ activation pathway in GC.

Cardiac-protective; Doxorubicin

Isorhamnetin is a natural anti-oxidant with obvious cardiac-protective effect. Its action against doxorubicin-induced cardotoxicity and underlying mechanisms were investigated. Doxorubicin (Dox) is an anthracycline antibiotic for cancer therapy with limited usage due to cardiotoxicity. The aim of this study is to investigate the possible protective effect of isorhamnetin against Dox-induced cardiotoxicity and its underlying mechanisms. In an in vivo investigation, rats were intraperitoneally (i.p.) administered with Dox to duplicate the model of Dox-induced chronic cardiotoxicity.

Daily pre-treatment with isorhamnetin (5 mg/kg, i.p.) for 7 days was found to reduce Dox-induced myocardial damage significantly, including the decline of cardiac index, decrease in the release of serum cardiac enzymes, and amelioration of heart vacuolation. In vitro studies on H9c2 cardiomyocytes, isorhamnetin was effective to reduce Dox-induced cell toxicity. Isorhamnetin also potentiated the anti-cancer activity of Dox in MCF-7, HepG2 and Hep2 cells. These findings indicated that isorhamnetin can be used as an adjuvant therapy for the long-term clinical use of Dox (Sun et al., 2013).

Chronic Myelogenous Leukemia

The isorhamnetin 3-o-robinobioside and its original extract, ethyl acetate extract, from Nitraria retusa leaves, were evaluated for their ability to induce anti-oxidant and anti-genotoxic effects in human chronic myelogenous leukemia cell line. They were shown to have a great anti-oxidant and anti-genotoxic potential on human chronic myelogenous leukemia cell line K562 (Boubaker et al., 2012).

Esophageal Cancer

The flavonol aglycone isorhamnetin shows anti-proliferative activity in a variety of cancer cells and it inhibits the proliferation of human esophageal squamous carcinoma Eca-109 cells in vitro (Shi et al., 2012).

References

Boubaker J, Ben Sghaier M, Skandrani I, et al. (2012). Isorhamnetin 3-O-robinobioside from Nitraria retusa leaves enhance anti-oxidant and anti-genotoxic activity in human chronic myelogenous leukemia cell line K562. BMC Complement Altern Med, 12:135. doi: 10.1186/1472-6882-12-135.


Ramachandran L, Manu KA, Shanmugam MK, et al. (2013). Isorhamnetin inhibits proliferation and invasion and induces apoptosis through the modulation of peroxisome proliferator-activated receptor γ activation pathway in gastric cancer. J Biol Chem, 288(26):18777. doi: 10.1074/jbc.A112.388702.


Saud SM, Young MR, Jones-Hall YL, et al. (2013). Chemo-preventive activity of plant flavonoid isorhamnetin in colorectal cancer is mediated by oncogenic Src and β -catenin. Cancer Res, 73:5473.


Shi C, Fan LY, Cai Z, Liu YY, Yang CL. (2012). Cellular stress response in Eca-109 cells inhibits apoptosis during early exposure to isorhamnetin. Neoplasma, 59(4):361-9. doi: 10.4149/neo_2012_047.


Sun J, Sun G, Meng X, et al. (2013). Isorhamnetin protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS One, 8(5):e64526. doi: 10.1371/journal.pone.0064526.


Zhu L, Wang ZR, Zhou LM, et al. (2005). Effects and mechanisms of isorhamnetin on lung carcinoma. Space Med Med Eng (Chin), 18:381-383.

Honokiol

Cancer: Sarcoma

Honokiol, isolated from the Chinese traditional herb magnolia, is a poorly water-soluble component and has been found to have anti-tumor properties.

Sarcoma

In the current study, honokiol submicron lipid emulsions (HK-SLEs) were prepared by high-pressure homogenization technology. After HK-SLEs were physically characterized, their pharmacokinetics, tissue distribution and anti-tumor activity after intravenous (i.v.) administration to tumor-burdened mice were examined, using honokiol solution (HK-SOL) as the control.

Both honokiol submicron lipid emulsions (HK-SLEs) and honokiol solution (HK-SOL) tended to accumulate in brain tissue. In vivo study showed that HK-SLEs treatment caused significant inhibition of mouse sarcoma S180 tumor growth compared to HK-SOL. These results suggest that HK-SLEs might be an effective parenteral carrier for honokiol delivery in cancer treatment (Zheng et al., 2013).

Reference

Zheng J, Tang Y, Sun M, et al. (2013). Characterization, pharmacokinetics, tissue distribution and anti-tumor activity of honokiol submicron lipid emulsions in tumor-burdened mice. Pharmazie, 68(1):41-6.

Cinobufacini Injection

Cancer: Liver, lung

Action: Chemo-sensitizer, chemotherapy support, cytostatic

Ingredients: chan su (Dried toad skin/Bufo bufo gargarizans)

TCM functions: Removing Toxin, reducing swelling, relieving pain.

Indications: Anti-tumor, immune enhancing and anti-viral effects, and can be used in middle and late-stage tumors, chronic hepatitis B.

Dosage and usage:

Intramuscular injection: 2-4 ml once, twice daily, 2-3 months as a course of treatment.

Cervical Cancer; Radiotherapy

Sixty patients with early cervical cancer were randomly divided into two groups. Twenty eight cases in treatment group were treated by intensity modulated radiation therapy combined with Brucea javanica oil emulsion injection. Thirty two cases in control group were treated only by intensity modulated radiation therapy. There was no significant difference between the two groups on the short-term  effect and lesion local control rate (P > 0.05). The 3-year overall survival rate in the treatment group was higher than that in control group (P<0.05). There was significant difference between the two groups on radiation proctitis (P<0.05).

Intensity modulated radiation therapy combined with Brucea javanica oil emulsion injection can improve efficacy and reduce adverse reactions in early cervical cancer, worthy of clinical application. 10-20 ml mixed with 500 ml of 5% glucose for slow intravenous drip. Four weeks as a course of treatment, and 1-2 days interval after each week”s treatment.

Cinobufacini Injection (CI) showed better tumor inhibition effects on tumor-bearing rats of with a “heat syndrome” constitution, indicating CI was of a “cold property”. It may potentially be used in tumor-bearing rats of a “heat syndrome” constitution (Wang et al., 2011).

Induces Apoptosis

Chan Su is a traditional Chinese medicine prepared from the dried white secretion of the auricular and skin glands of toads, and has been used as an oriental drug for the treatment of a number of diseases, including cancer. In lung carcinoma A549 cells, treatment with the skin of Venenum Bufonis (SVB) resulted in the inhibition of cell growth and viability, and the induction of apoptosis.

SBV treatment induced the proteolytic activation of caspases and the concomitant degradation of poly(ADP-ribose)-polymerase and beta-catenin protein. Cleavage of Bid and a down-regulation of the inhibitor of apoptosis family proteins were also observed in SBV-treated A549 cells. Data from this study indicates that SVB induces the apoptosis of A549 cells through a signaling cascade of death receptor-mediated extrinsic and mitochondria-mediated intrinsic caspase pathways (Yun et al., 2009).

Blocks Metastasis

The effect of Cinobufacini injection on proliferation, heterogeneous adhesion, and invasiveness of human hepatoma HepG-2 cells co-cultured with human lymphatic endothelial cells (HLEC) was studied.

A co-culture system of human hepatoma HepG-2 cells and HLEC was established by means of Transwell chamber. Cell proliferation was analyzed by Trypan blue stain assay. MTT assay was used to observe the heterogeneous adhesion capacity of HepG-2 cells co-cultured with HLEC. Transwell invasion chamber was used to observe the invasiveness capacity of HepG-2 cells co-cultured with HLEC.

Cinobufacini Injection significantly inhibits proliferation, heterogeneous adhesion and invasiveness of hepG-2 cells co-cultured with HLEC in dose-dependent ways (all P0.05). Cinobufacini injection can inhibit the capability of proliferation, invasiveness and heterogeneous adhesion of HepG-2 cells, which might contribute to the inhibiting mechanisms of Cinobufacini injection on tumor metastasis (Fu, Gao, Tian, Chen, & Cui, 2013).

Inhibits Human Lymphatic Endothelial Cells (HLEC)

The effect of Cinobufacini injection on proliferation, migration and tubulin formation of human lymphatic endothelial cells (HLEC) was investigated.

Cell growth curve was used to observe the effect of Cinobufacini injection on the proliferation of HLEC; migration assay was used to observe the effect of Cinobufacini injection on the migration of HLEC; Matrigel assay was used to observe the effect of Cinobufacini injection on the tubulin formation of HLEC; Western blot was used to analyze the expression of VEGFR-3 and HGF in HLEC.

As the dosage of Cinobufacini injection increased (0.105, 0.21 and 0.42 µg/mL), so did the inhibition of HLCE. Cinobufacini injection demonstrated significant inhibition of HLEC proliferation (P < 0.05), migration (P < 0.05) and tubulin formation, in a dose-dependent manner (P < 0.05). Cinobufacini injection significantly decreased the expression of VEGFR-3 and HGF in HLEC, in a dose-dependent manner (P < 0.05).

Cinobufacini injection significantly inhibits HLEC proliferation, migration, and tubulin formation. The down-regulation of VEGFR-3 and HGF may contribute to the inhibitory effect of Cinobufacini injection on HLEC (Gao, Chen, Xiu, Fu, & Cui, 2013).

NSCLC; Chemotherapy

The efficacy and safety of Cinobufacini injection, combined with chemotherapy, as a treatment for advanced non-small-cell lung cancer (NSCLC) was investigated. Based on existing clinical information, a search of databases, such as Medline (1966-2011), Cochrane Library (2011, Issue 11), CNKI (1978-2011), VIP (1989-2011), Wanfang Data (1988-2011), CBMdisc (1978-2011) was done.

A total of seven RCTs of 498 patients were included. Meta-analysis results show that the experimental group and control group have significant differences in the response rate [RR=1.29, 95% CI (1.07, 1.56)], Karnofsky score [RR=1.86, 95% CI (1.14, 3.05)], weight change [RR=1.56, 95% CI (1.20, 2.03)], gastrointestinal side-effects [RR=0.72, 95% CI (0.53, 0.99)], neutropenia [RR=0.70, 95%CI(0.54, 0.91)], thrombocytopenia [RR=0.53, 95% CI (0.38, 0.75)], and renal function [RR=0.37, 95% CI (0.17, 0.79).

Cinobufacini, combined with chemotherapy, is suitable for advanced NSCLC by improving the response rate, increasing Karnofsky score, gaining weight and reducing major side-effects (Tu, Yin, & He, 2012).

Liver Cancer

The clinical effect of Cinobufacini injection, combined with transcatheter arterial chemoembolization (TACE), on treating primary liver cancer was investigated.

Seventy-eight patients with moderate and advanced primary liver cancer were randomly divided. The treatment group (n=38) was treated by Cinobufacini injection combined with TACE, and the control group (n=40), was treated by TACE only.

Quality of life of patients in the treatment group was significantly higher than that in control group. The 12 months survival rate of the treatment group was significantly higher than that of control group. There was no statistical difference in the rate of effectiveness between the two groups. Laboratory tests, after three cycles, in the treatment group were better than that of the control group, and the difference between the two groups was statistically significant.

Cinobufacini injection, combined with TACE, can decrease TACE induced liver damage, prolong survival time, and improve body immunity (Ke, Lu, & Li, 2011).

Hepatoma

Cinobufacini injection significantly inhibited HepG-2 cells proliferation in a dose and time-dependent manner. FCM analysis showed Cinobufacini injection induced cell-cycle arrest at the S phase. RT-PCR assay showed Cinobufacini injection down-regulated Cyclin A, and CDK2 expression at mRNA levels. Quantitative colorimetric assay showed Cinobufacini injection deceased Cyclin A/CDK2 activity in HepG-2 cells.

Cinobufacini injection can inhibit human hepatoma HepG-2 cells growth, induce cell apoptosis and induce cell-cycle arrest at the S phase. Its mechanism might be partly related to the down-regulation of Cyclin A, CDK2 mRNA expression, and inhibition of Cyclin A/CDK2 activity (Sun, Lu, Liang, & Cui, 2011).

Cell-cycle Arrest

Studies in China by Sun et al., (2011), Ke et al., (2011) and Tu et al., (2012) demonstrated that Cinobufacini Injection induced cell-cycle arrest, and could be used in the treatment of primary liver cancer, as well as in conjunction with chemotherapy in the treatment of non-small-cell lung cancer.

Caution

Resibufogenin (RBG), one of the major components in chan su, significantly affected all parameters of transmembrane action potential., induced delayed response after depolarization, and triggered arrhythmias in sheep and canine Purkinje fibers. Chan su toxicity carries a high mortality rate in the United States and this study focused upon the cardiac electrophysiological and electro-toxicity effects of RBG (Xie et al., 2000).

References

Fu, H.Y., Gao, S., Tian, L.L., Chen, X.Y., & Cui, X.N. (2013). Effect of Cinobufacini injection on proliferation and invasiveness of human hepatoma HepG-2 cells co-cultured with human lymphatic endothelial cells. The Chinese Journal of Clinical Pharmacology, 29(3), 199-201.


Gao, S., Chen, X.Y., Fu, H.Y., & Cui, X.Z. (2013). The effect of Cinobufacini injection on proliferation and tube-like structure formation of human lymphatic endothelial cells. China Oncology, 23(1), 36-41.


Ke, J, Lu, K., & Li, Y. (2011). Clinical observation of patients with primary liver cancer treated by Cinobufagin Injection combined with transcatheter arterial chemoembolization. Chinese Journal of Clinical Hepatology.


Sun, Y., Lu, X.X., Liang, X.M., & Cui, X.N. (2011). Impact of Cinobufacini injection on proliferation and cell-cycle of human hepatoma HepG-2 cells. The Chinese-German Journal of Clinical Oncology, 10(6), 321-324.


Tu, C., Yin, J., & He, J. Meta-analysis of Cinobufacini injection plus chemotherapy in the treatment of non-small-cell lung cancer. Anti-tumor Pharmacy, 2(1), 67-72.


Wang, S.S., Zhai, X.F., Li, B. (2011) Effect of cinobufacini injection on the tumor growth of tumor-bearing rats of different constitutions. Zhongguo Zhong Xi Yi Jie He Za Zhi, 31(8):1101-3.


Xie, J-T., Wang, Hs., Attele A.S., Yuan, C-S. (2000). Effects of Resibufogenin from Toad Venom on Isolated Purkinje Fibers. American Journal of Chinese Medicine, 28(2):187-196.


Yun, H.R., Yoo, H.S., Shin, D.Y., et al. (2009). Apoptosis induction of human lung carcinoma cells by Chan Su (Venenum Bufonis) through activation of caspases. J Acupunct Meridian Stud, 2(3):210-7. doi: 10.1016/S2005-2901(09)60057-1.

Carnosol

Cancer: Breast, prostate, skin, colon, leukemia, stomach

Action: Anti-inflammatrory, anti-angiogenic

Carnosol is found in certain Mediterranean meats, fruits, vegetables, and olive oil. In particular, it is sourced from rosemary (Rosmarinus officinalis (L.)) and desert sage (Salvia pachyphylla (Epling ex Munz)).

Prostate Cancer, Breast Cancer, Skin Cancer, Colon Cancer, Leukemia

One agent, carnosol, has been evaluated for anti-cancer property in prostate, breast, skin, leukemia, and colon cancer with promising results. These studies have provided evidence that carnosol targets multiple deregulated pathways associated with inflammation and cancer that include nuclear factor kappa B (NFκB), apoptotic related proteins, phosphatidylinositol-3-kinase (PI3 K)/Akt, androgen and estrogen receptors, as well as molecular targets. In addition, carnosol appears to be well tolerated in that it has a selective toxicity towards cancer cells versus non-tumorigenic cells and is well tolerated when administered to animals.

This mini-review reports on the pre-clinical studies that have been performed to date with carnosol describing mechanistic, efficacy, and safety/tolerability studies as a cancer chemoprevention and anti-cancer agent (Johnson, 2011).

Literature evidence from animal and cell culture studies demonstrates the anti-cancer potential of rosemary extract, carnosol, carnosic acid, ursolic acid, and rosmarinic acid to suppress the development of tumors in several organs including the colon, breast, liver, stomach, as well as melanoma and leukemia cells (Ngo et al., 2011).

Anti-inflammatory

Treatment with retinoic acid (RA) or carnosol, two structurally unrelated compounds with anti-cancer properties, inhibited phorbol ester (PMA)-mediated induction of activator protein-1 (AP-1) activity and cyclooxygenase-2 (COX-2) expression in human mammary epithelial cells. Treatment with carnosol but not RA blocked increased binding of AP-1 to the COX-2 promoter. Carnosol but not RA inhibited the activation of PKC, ERK1/2, p38, and c-Jun NH2-terminal kinase mitogen-activated protein kinase. Overexpressing c-Jun but not CBP/p300 reversed the suppressive effect of carnosol on PMA-mediated stimulation of COX-2 promoter activity.

Carnosol inhibited the induction of COX-2 by blocking PKC signaling and thereby the binding of AP-1 to the CRE of the COX-2 promoter. Taken together, these results show that small molecules can block the activation of COX-2 transcription by distinct mechanisms (Subbaramaiah, 2002).

Breast Cancer

Two rosemary components, carnosol and ursolic acid, appear to be partly responsible for the anti-tumorigenic activity of rosemary. Supplementation of diets for 2 weeks with rosemary extract (0.5% by wt) but not carnosol (1.0%) or ursolic acid (0.5%) resulted in a significant decrease in the in vivo formation of rat mammary DMBA-DNA adducts, compared to controls. When injected intraperitoneally (i.p.) for 5 days at 200 mg/kg body wt, rosemary and carnosol, but not ursolic acid, significantly inhibited mammary adduct formation by 44% and 40%, respectively, compared to controls. Injection of this dose of rosemary and carnosol was associated with a significant 74% and 65% decrease, respectively, in the number of DMBA-induced mammary adenocarcinomas per rat, compared to controls. Ursolic acid injection had no effect on mammary tumorigenesis.

Therefore, carnosol is one rosemary constituent that can prevent DMBA-induced DNA damage and tumor formation in the rat mammary gland, and, thus, has potential for use as a breast cancer chemopreventative agent (Singletary et al., 1996).

Anti-angiogenic

The anti-angiogenic activity of carnosol and carnosic acid could contribute to the chemo-preventive, anti-tumoral and anti-metastatic activities of rosemary extracts and suggests that there is potential in the treatment of other angiogenesis-related malignancies (L-pez-JimŽnez et al., 2013).

References:

Johnson JJ. (2011). Carnosol: A promising anti-cancer and anti-inflammatory agent. Cancer Letters, 305(1):1-7. doi:10.1016/j.canlet.2011.02.005.


L-pez-JimŽnez A, Garc'a-Caballero M, Medina Mç, Quesada AR. (2013). Anti-angiogenic properties of carnosol and carnosic acid, two major dietary compounds from rosemary. Eur J Nutr, 52(1):85-95. doi: 10.1007/s00394-011-0289-x.


Ngo SN, Williams DB, Head RJ. (2011). Rosemary and cancer prevention: preclinical perspectives. Crit Rev Food Sci Nutr, 51(10):946-54. doi: 10.1080/10408398.2010.490883.


Singletary K, MacDonald C & Wallig M. (1996). Inhibition by rosemary and carnosol of 7,12-dimethylbenz[a]anthracene (DMBA)-induced rat mammary tumorigenesis and in vivo DMBA-DNA adduct formation. Cancer Letters, 104(1):43-8. doi: 10.1016/0304-3835(96)04227-9


Subbaramaiah K, Cole PA, Dannenberg AJ. (2002). Retinoids and Carnosol Suppress Cyclooxygenase-2 Transcription by CREB-binding Protein/p300-dependent and -independent Mechanisms. Cancer Res, 62:2522

Camptothecin (CPT) & 10-hydroxycamptothecin (HCPT)

Cancer: Breast, colon

Action: Cytostatic

Breast Cancer

Recently, natural product DNA topoisomerase I inhibitors 10-hydroxycamptothecin (HCPT) and camptothecin (CPT) have been shown to have therapeutic effects in both in vitro and in vivo models of human breast cancer. After evaluation, the apoptotic pathways were characterized in vitro and in vivo in the human breast cancer cell lines MCF-7 and MDA-MB-468.

The elevation of p53 protein levels in MCF-7 cells treated with CPT was significantly inhibited by preincubation with DNA breaks inhibitor aphidicolin, while the elevation of p21WAF1/CIP1 protein levels was not inhibited. The elevation of p21WAF1/CIP1 in MDA-MB-468 cells treated with CPT was not inhibited by aphidicolin.

Results suggest that treatment with HCPT and CPT results in increased levels of p21WAF1/CIP1 protein and mRNA, and that they induce apoptosis in human breast cancer cells through both p53-dependent and -independent pathways. Findings may be significant in further understanding the mechanisms of actions of camptothecins in the treatment of human cancers (Liu & Zhang, 1998).

Colon Cancer

10-HCPT significantly repressed the proliferation of Colo 205 cells at a relatively low concentration (5-20 nM). Flow cytometry analysis and western blot and apoptosis assays demonstrated that low-dose 10-HCPT arrested Colo 205 cells in the G2 phase of the cell-cycle and triggered apoptosis through a caspase-3-dependent pathway.

Moreover, following oral administration at doses of 2.5-7.5 mg/kg/2 days, significant suppression of tumor growth by 10-HCPT was observed in mouse xenografts. No acute toxicity was observed after an oral challenge of 10-HCPT in BALB/c-nude mice every 2 days.

Results suggest that a relatively low dose of 10-HCPT (p.o.) is able to inhibit the growth of colon cancer, facilitating the development of a new protocol of human trials with this anti-cancer drug (Ping et al., 2006).

References

Liu, W., & Zhang, R. (1998). Up-regulation of p21WAF1/CIP1 in human breast cancer cell lines MCF-7 and MDA-MB-468 undergoing apoptosis induced by natural product anti-cancer drugs 10-hydroxycamptothecin and camptothecin through p53-dependent and independent pathways. International Journal of Oncology, 12(4), 793-804.


Ping, Y.H., Lee, H.C., Lee, J.Y., et al. (2006). Anti-cancer effects of low-dose 10-hydroxycamptothecin in human colon cancer. Oncology Reports, 15(5), 1273-9.

Astragalus (huang qi)

Cancer: Non-small-cell lung cancer, breast, colon, stomach

NSCLC; Chemotherapy

Guo et al. (2012) reported that treatment with Astragalus polysaccharide (APS) injections integrated with vinorelbine and cisplatin significantly improved quality of life in patients with advanced non-small-cell lung cancer over vinorelbine and cisplatin alone.

NSCLC

Astragalus injection (AI) combined with chemotherapy can significantly improve the QOF in NSCLC patients of advanced stage. The effective rate in the treated group was 40.0% and in the control group was 36.7%, the mean remission rate in the treated and control group was 5.4 months and 3.3 months, the median survival period 11 months and 7 months, and the 1-year survival rate 46.75% and 30.0%, respectively; the differences of these indexes between the two groups were all significant (P < 0.05). Moreover, the clinical improving rate and QOF elevation rate in the treated group was 80.4% and 43.3%, as compared with those in the control group (50.0% and 23.3% respectively); the difference was also significant (P < 0.01) (Zou & Liu, 2003).

Breast Cancer

In physiological dose E2, Astragalus mongholicus injection inhibited MCF-7 breast cancer cells proliferation at all concentration groups. As time lasting, Astragalus mongholicus injection showed better inhibitory effect than TAM (P<0.05). Among 2 x 10(-1) g/mL-2 x 10(-4) g/mL concentration, Astragalus mongholicus injection significantly increased the proliferative percent of G0/G1 and S-phase cell, decreased percent of G2-M phase cell (P<0.05) at 24 hours. After cocultured 72 hours, Astragalus mongholicus injection increased the rate of apoptosis to 16.7% at 2 x 10(-1) g/mL concentration (Zhou, Liu, & Tan, 2009).

Acute Exacerbations, Respiratory Failure in Chronic Obstructive Pulmonary Disease

A total of 112 patients with acute chronic obstructive pulmonary disease (AECOPD)were randomly divided into the treatment group (56 cases) and control group (56 cases). The treatment group received a 40 mL astragalus injection, with 5% glucose, 250 mL intravenous drip once a day at the start of conventional therapy. The control group received conventional therapy only. The therapeutic course of both groups was 14 days, and clinical therapeutic effects were observed. Serum levels of TNF-α>, IL-8, IL-2, lung function and blood gas analysis index of both groups were measured before and after treatment. The treatment group”s effectiveness rate was 94.64%, compared to the control group”s 67.86%, which was statistically significant (P<0.05).

Astragalus injection may significantly decrease the serum levels of TNF-α and IL-8, and increase the level of IL-2. It may improve the lung function and the curative effect in the patients with AECOPD (Xiong, Guo, & Xiong, 2013).

Residual Renal Function

The effect of astragalus injection on hemodialysis patient”s RRF (residual renal function, RRF) was observed.

Sixty hemodialysis patients with a RRF of more than 2ml/min were randomly divided into either an astragalus injection treatment group or a control group treated with normal saline. One hour prior to hemodialysis completion, the treatment group was administered an astragalus injection of 30ml, while the control group was given 30 ml of normal saline. Follow up after 6 months compared data of daily urine output and RRF.

Astragalus injection can potentially delay the rate of daily urine output reduction and protect RRF to some extent (Qi et al., 2013).

Stomach Cancer, Colon Cancer; Oxaliplatin-induced Neurotoxicity

40 patients with stomach or colon cancer were enrolled in the study. Patients comprised of 23 men and 17 women, from the ages of 32-75 years (mean age 60 years), and were randomly divided into two groups: the test group and the control group (20 cases in each group). All patients were treated with one cycle of an Oxaliplatin-containing chemotherapy regimen, entailing: oxaliplatin 130 mg/m2 on day 1, fluorouracil 0.5 g on days 1-5, and calcium foliate 0.2 g on days 1-5. In the test group 30 ml of Huangqi injection was added to the regimen on days 1-7. The manifestation of peripheral neurotoxic reactions were observed and nerve growth factor levels were measured.

In the control group, 2 patients had grade 0 toxicity, 10 had grade 1 toxicity, 6 had grade 2 toxicity, and 2 had grade 3 toxicity. In the test group, 14 patients had grade 0 toxicity and 6 had grade 1 toxicity. The incidence rate of neurotoxicity in the test and control groups was 30% and 90%, respectively. In the test and control groups, the nerve growth factor levels were (167 ± 10) ng/ml and (204 ± 19) ng/ml before chemotherapy, as well as (152 ± 8) ng/ml and (133 ± 12) ng/ml 2 days after chemotherapy, respectively. In the control group, the nerve growth factor levels were markedly decreased 2 days after chemotherapy compared to before chemotherapy. The difference between the two groups was statistically significant (P < 0.01).

Huangqi injection has some degree of efficacy in the prevention and treatment of Oxaliplatin-induced neurotoxicity (Cui, Li, Tan, & Li, 2009).

Myelosuppression

Astragalus membranaceus injection (AMI), administered at (500 mg/kg) improved the hematopoietic microenvironment by enhancing the BMSC survival and proliferation of colony-forming unit-fibroblast (CFU-F) formation, production of IL-6 as well as Granulocyte-macrophage colony-stimulating factor (GM-CSF) by BMSC and bcl-2 protein and mRNA expression in BMSC, which promoted myelopoiesis. The data may provide a mechanistic basis for applying this ancient Chinese herb to promote hematopoiesis as an efficacious adjuvant therapy against myelosuppression induced by anti-cancer therapy (Zhu & Zhu, 2007).

References

Cui, H.J., Li, O.J., Ying, H.Y., & Li, Y. (2009). Clinical observation of efficacy of huangqi injection in the prevention and treatment of neurotoxicity induced by oxaliplatin-containing chemotherapy regimen. Adverse Drug Reactions Journal., 11(4), 1671-8585.


Guo, L., Bai, S.P., Zhao, L., Wang, X.H. (2012). Astragalus polysaccharide injection integrated with vinorelbine and cisplatin for patients with advanced non-small-cell lung cancer: effects on quality of life and survival. Med Oncol. http://dx.doi.org/10.1007/s12032-011-0068-9.


Qi, Y.H., Qu, X.L., Tang, Y.H., Dai, Q., Zhang, S.B., & Yao, C.Y. (2013). The impact of Astragalus injection on residual renal function in hemodialysis patients. New Medicine, 2013(2), 105-107.


Xiong, S., Guo, Y., & Xiong, X. (2013). Influence of astragalus injection on serum cytokines and lung function in acute exacerbation of chronic obstructive pulmonary disease. China Modern Doctor, 51(9), 43-45.


Zhou, R.F., Liu, P.X., Tan, M. (2009). Effect of Astragalus mongholicus injection on proliferation and apoptosis of hormone sensitive (MCF-7) breast cancer cell lines with physiological dose E2. Zhong Yao Cai, 32(5):744-7.


Zou, Y.H., Liu, X.M. (2003). Effect of astragalus injection combined with chemotherapy on quality of life in patients with advanced non-small-cell lung cancer. Zhongguo Zhong Xi Yi Jie He Za Zhi, 23(10):733–735.


Zhu XL, Zhu BD. (2007). Mechanisms by which Astragalus membranaceus injection regulates hematopoiesis in myelosuppressed mice. Phytother Res, 21(7):663-7.

Artesunate

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

Action: Anti-metastatic, MDR, radio-sensitizer

Pulmonary Adenocarcinomas

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

Breast Cancer

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

Skin Cancer

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

Colon Cancer

Artemisunate significantly inhibited both the invasiveness and anchorage independence of colon cancer SW620 cells in a dose-dependent manner. The protein level of intercellular adhesion molecule 1 (ICAM-1) was down-regulated as relative to the control group.

Artemisunate could potentially inhibit invasion of the colon carcinoma cell line SW620 by down-regulating ICAM-1 expression (Fan, Zhang, Yao & Li, 2008).

Multi-drug resistance; Colon Cancer

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

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

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

Multi-drug resistance; Esophageal Cancer

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

ART is a noteworthy antimalarial agent, particularly in severe and drug-resistant cancer cases, as ART is able to reverse drug resistance. ART exerted profound anti-cancer activity. The mechanism for the reversal of multi-drug resistance by ART in esophageal carcinoma was analyzed using cellular experiments, but still remains largely unknown (Liu, Zuo, & Guo, 2013).

Pancreatic Cancer

The combination of triptolide and artesunate could inhibit pancreatic cancer cell line growth, and induce apoptosis, accompanied by expression of HSP 20 and HSP 27, indicating important roles in the synergic effects. Moreover, tumor growth was decreased with triptolide and artesunate synergy. Results indicated that triptolide and artesunate in combination at low concentrations can exert synergistic anti-tumor effects in pancreatic cancer cells with potential clinical applications (Liu & Cui, 2013).

Ovarian Cancer

Advanced-stage ovarian cancer (OVCA) has a unifocal origin in the pelvis. Molecular pathways associated with extrapelvic OVCA spread are also associated with metastasis from other human cancers and with overall patient survival. Such pathways represent appealing therapeutic targets for patients with metastatic disease.

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

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

Multiple Myeloma, B-cell Lymphoma

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

Osteosarcoma, Leukemia/Lymphoma

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

References

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


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


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


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


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


Liu, L., Zuo, L.F., Guo, J.W. (2013). Reversal of Multi-drug resistance by the anti-malaria drug artesunate in the esophageal cancer Eca109/ABCG2 cell line. Oncol Lett, 6(5):1475-1481.


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


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


Marchion, D.C., Xiong, Y., Chon, H.S., et al. (2013). Gene expression data reveal common pathways that characterize the unifocal nature of ovarian cancer. Am J Obstet Gynecol, S0002-9378(13)00827-2. doi: 10.1016/j.ajog.2013.08.004.


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


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


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


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


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

Ai Di Injection (ADI)

Cancers: Breast, colorectal., glioma, lung

Action: Chemo-sensitizer, cytostatic, radio-sensitizer

 

Ingredients: Mylabris phalerata (ban mao), Panax ginseng (ren shen), Astragalus membranaceus (huang qi).

TCM functions: Clearing Heat, removing Toxin, resolving stagnant Blood, dissolving lumps.

Indications: Primary liver cancer, lung cancer, colorectal cancer, malignant lymphoma, and gynecological malignancies.

Dosage and usage:

For adults: 50-100ml, mixed with 400-500ml of 0.9% NaCl injection or 5-10% glucose injection for intravenous drip, once daily.

When combined with radiotherapy or chemotherapy, the course of treatment is synchronized to radiotherapy or chemotherapy.

Application before or after the surgery: 10 days as a course of treatment.

Intervention treatment: 10 days as a course of treatment.

Single application: 15 days as a cycle, with 3 days interval., 2 cycles as a course of treatment.

 

Cachexia patients in advanced stage: 30 consecutive days as a course of treatment (Drug Information Reference in Chinese: See end).

 

Glioma; Radio-sensitization

The inhibition ratio was determined by MTT assay, the change in the cell-cycle was analyzed by flow cytometry and the expression of cyclin B1 and Wee1 was detected by Western blot analysis. The reproductive activity of the group treated with irradiation (IR) and Aidi injection was suppressed significantly, and the cloning efficiency and divisional index also declined. Aidi injection (15 µg/ml) induced G2/M phase arrest in the cell line after 48 h.

 

Aidi injection (ADI) is effective in radio-sensitization. The possible mechanisms involved may be associated with G2/M phase cell arrest, the down-regulation of cyclin B1 and up-regulation of Wee1 expression, which influences cell size by inhibiting the entry into mitosis, through inhibiting Cyclin-dependent kinase 1 (Xu, Song, Qin, Wang, & Zhou, 2012).

 

Breast Cancer

ADI significantly inhibited the proliferation of MCF-7 cells in a dose-dependent manner. The IC50 of ADI was 55.71 mg/mL after treatment for 48 h. The 60 mg/mL ADI was used as the therapeutic drug concentration. Microarray analysis identified 45 miRNAs that were up-regulated and 55 miRNAs that were down-regulated in response to ADI treatment. Many ADI-induced miRNAs were related to breast cancers. The 12 potential target genes of mir-126 were predicted by both TargetScan and PicTar software.

 

The miRNA may serve as therapeutic targets for ADI, and its modulation of expression is an important mechanism of ADI inhibition of breast cancer cell growth (Zhang, Zhou, Lu, Du, & Su, 2011).

 

Colorectal Cancer; FOLFOX4

A consecutive cohort of 100 patients was divided into two groups. The experimental group was treated with a combination of Aidi injection and FOLFOX4, while the control group was only administered FOLFOX4. After a minimum of two courses of treatment, efficacy, quality of life, and side-effects were evaluated.

 

The response rate of the experimental group was not significantly different compared to the control group (P > 0.05). However, there were significant differences in clinical benefit response and KPS score. In addition, adverse gastrointestinal reactions and the incidence of leukopenia were lower than that of the control group (P < 0.05).

Aidi injection, combined with FOLFOX4, is associated with reduced toxicity of chemotherapy, enhanced clinical benefit response, and improved quality of life in patients with advanced colorectal cancer (Xu, Huang, Li, Li, & Tang, 2011).

 

NSCLC

Ninety-eight cases of advanced NSCLC were randomly divided into two groups: a trial group and control group. In the trial group Navelbine/Cisplatin (NP) plus Ai Di Injection (ADI) (60-80 ml) was administered intravenously, via dissolution in 400 ml of normal saline, per day for 8-10 days. In the control group, only NP chemotherapy was administered at the dosages of: Navelbine (25 mg/m², d1, 8) and Cisplastin (40 mg/m², d1-3). Each patient received at least two cycles of treatment.

 

The effective rate in the trial group and the control group was 53.1% and 44.9% respectively, without significant difference between the two groups (P > 0.05). However, the rate of progression, adverse reactions in the bone marrow, digestive tract, and immune function in the trial group were all lower than those in the control group (P < 0.05). In addition, improvement in Karnofsky score in the trial group was higher than that in the control group (P < 0.05).

 

A chemotherapy regiment of NP, combined with ADI, shows benefit in the treatment of advanced NSCLC. AI could minimize the adverse reactions of chemotherapy, and improve the quality of life in patients with NSCLC (Wang et al., 2004).

 

NSCLC; Meta-analysis

PubMed (1980-2008), Cochrane Central Register of Controlled Trials (The Cochrane Library, Issue 3, 2008), EMBASE (1984-2008), CancerLit (1996-2003), CBMdisc (1980-2008), CNKI database (1980-2008), Wanfang database (1980-2008), and Chongqing VIP database (1980-2008) were searched. Relevant Chinese periodicals were manually searched as well. All randomized controlled trials comparing Aidi Injection with other treatment methods of NSCLC were included. Two reviewers selected studies, assessed the quality of studies, and extracted the data independently.

 

Fourteen randomized controlled trials were included in the meta-analysis, but unfortunately, the quality of reports of the 14 included studies were poor. Aidi Injection combined with cobalt-60, or navelbine and platinol (NP), showed statistically significant differences in improving the response rate, compared to the use of cobalt-60 alone (P = 0.0002) or NP alone (P = 0.04). However, Aidi Injection combined with etoposide and platinol (EP), taxinol and platinol (TP) or gamma knife showed no significant differences when compared with single use of EP (P=0.60), TP (P=0.16) or gamma knife (P=0.34), respectively. The RR and 95% CI of EP, TP, and gamma knife were 1.17 [0.65, 2.09], 1.27 [0.91, 1.78] and 1.08 [0.92, 1.26] respectively.

 

Six studies indicated that Aidi Injection, combined with NP or gamma knife, could improve quality of life. Six studies showed that Aidi Injection, combined with NP or TP, could improve the bone marrow’s hematopoietic function. The results of the meta-analysis indicate that Aidi Injection may have adjuvant therapeutic effects in the treatment of NSCLC patients. However, sample sizes are small, study quality is poor, and the existence of publication bias had been found. The effects of Aidi Injection need to be confirmed by large multicenter randomized controlled trials (Ma, Duan, Feng, She, Chen & Zhang, 2009).

 

NSCLC; Neo-adjuvant Chemotherapy

Sixty patients, with stage IIIA non-small-cell lung cancer (NSCLC), underwent two courses of bronchial arterial infusion (BAI) chemotherapy, before tumor incision. They were assigned to either the treatment or control group, using a random number table. Thirty patients were allocated to each. An ADI of 100 mL, added into 500 mL of 5% glucose, was given to the patients in the treatment group via intravenous drip. Treatment was once a day, beginning 3 days prior and throughout each of two 14-day courses of chemotherapy.

 

Levels of T-lymphocyte subsets, natural killer cell activity, and interleukin-2 in peripheral blood were measured before and after the treatment. The effective rate in the treatment group was higher than that in the control group (70.0% vs. 56.7%, P < 0.05).

 

Moreover, bone marrow suppression and liver function damage (P < 0.05) was less in the treatment group relative to the control. Cellular immune function was suppressed in NSCLC patients, but was ameliorated after treatment, showing a significant difference when compared to the control group (P < 0.05).

 

ADI could potentially act as an ideal auxiliary drug for patients with stage IIIA NSCLC, receiving BAI neo-adjuvant chemotherapy, before surgical operation. It could enhance the effectiveness of chemotherapy, ameliorate adverse reactions, and elevate patient’s cellular immune function (Sun, Pei, Yin, Wu & Yang, 2010).

 

References

Ma, W.H., Duan, K.N., Feng, M., She, B., Chen, Y., & Zhang, R.M. (2009). Aidi Injection as an adjunct therapy for non-small-cell lung cancer: a systematic review. Journal of Chinese Integrative Medicine, 7(4), 315-324.

Sun, X.F., Pei, Y.T., Yin, Q.W., Wu, M.S., & Yang, G.T. (2010). Application of Aidi injection in the bronchial artery infused neo-adjuvant chemotherapy for stage III A non-small-cell lung cancer before surgical operation. Chinese Journal of Integrative Medicine, 16(6), 537-541.

Wang, D., Chen, Y., Ren, J., Cai, Y., M. Liu, M., & Zhan, Q. (2004). A randomized clinical study on efficacy of Aidi injection combined with chemotherapy in the treatment of advanced non-small-cell lung cancer. Journal of Chinese Integrative Medicine, 7(3), 247-249.

Xu, H.X., Huang, X.E., Li, Y., Li, C.G., & Tang, J.H. (2011). A clinical study on safety and efficacy of Aidi injection combined with chemotherapy. Asian Pacific Journal of Cancer Prevention, 12(9), 2233-2236.

Xu, X.T., Song, Y., Qin, S., Wang, L.L., & Zhou, J.Y. (2012). Radio-sensitization of SHG44 glioma cells by Aidi injection in vitro. Molecular Medicine Reports, 5(6), 1415-1418.

Zhang, H., Zhou, Q.M., Lu, L.L., Du, J., & Su, S.B. (2011). Aidi injection alters the expression profiles of microRNAs in human breast cancer cells. Journal of Traditional Chinese Medicine, 31(1), 10-16.

Acteoside

Cancer: Melanoma; metastasis

Ohno et al. (2009) suggests that acteoside injection, isolated from Plantago lanceolata showed suppressive effect on lung metastasis of B16 melanoma cells. Male C57BL/6 mice were injected intravenously with 2 x 10(5) of B16 melanoma cells, while acetoside at a dose of 50 mg/kg was administered intraperitoneally every other day from 13 d before B16 melanoma cell injection until all mice had succumbed to the metastatic tumor burden in the lung.

Administration of acteoside prolonged survival time significantly and the average survival time was 63.3 +/- 3.4d compared with 52.1 +/- 2.5d in control mice.

Reference

Ohno, T., Inoue, M., Ogihara, Y., Saracoglu, I. (2012) Anti-metastatic activity of acteoside, a phenylethanoid glycoside. Biological & Pharmaceutical Bulletin, 25(5):666-8. doi: 10.1248/bpb.25.666

β-Elemene

Cancer: Lung, malignant ascites, glioblastoma, gastric

Action: Anti-tumoral., chemotherapy support

Ingredients: Mixed liquid of β-, γ-, δ-elemene.

Indications: Increases the therapeutic effect and lowers the toxic and side-effects of radiotherapy and chemotherapy when in combination with routine regiments of radiotherapy or chemotherapy for lung cancer, liver cancer, esophageal cancer, nasopharyngeal cancer, brain tumors, metastatic bone cancer and other malignancies. It can also be used for intervention, intracavitary chemotherapy and pleural effusion or ascites caused by cancer.

Dosage and usage:

Intravenous injection: 0.4-0.6 g, once daily, 2-3 weeks as a course of treatment.

Pleural injection: 300 ml + 10 ml of 2% procaine. The treatment can be repeated once after 5-7 days if the pleural effusion does not reduce.

Abdominal injection: 500 ml + 10 ml of 2% procaine, 1-2 times every week for 2 consecutive weeks.

Topical administration: 25-50 mg, once daily, 5-10 times as a course of treatment.

Arterial infusion: 300-400 mg once.

Elemene Injection is made from mixed liquid of β-, γ-, δ-elemene. It can increase the therapeutic effect and lower the toxicity and side-effects of radiotherapy and chemotherapy when combined with routine regiments of radiotherapy or chemotherapy for lung cancer, liver cancer, esophageal cancer, nasopharyngeal cancer, brain tumors, metastatic bone cancer and other malignancies. It can also be used for intervention, intraperitoneal chemotherapy, and pleural effusion or ascites caused by cancer (Drug Information Reference in Chinese: See end. 2000-12).

NSCLC; Chemotherapy

Randomized controlled trials (RCTs) of elemene injection combined with cisplatin chemotherapeuties in treating small cell lung cancer (NSCLC) were collected by Xu et al., (2013). Their meta-analysis results suggested that compared with cisplatin chemotherapy alone, the combination of elemene injection and cisplatin chemotherapeutics showed a higher clinical benefit rate (OR = 2. 03, 95% CI:1.43-2. 88, P <0. 000 1) and a better quality of life (OR = 3.23, 95% CI:2. 20-4. 74, P <0. 000 01). As well, the combination could also reduce leucopenia (OR =0. 50, 95% CI:0. 33-0. 76, P <0. 001), and thrombocytopenia (OR =0. 38, 95% CI:0. 16-0. 85, P <0. 02), increase CD4 (MD = 3.32, 95% C1:2. 94-3.70, P <0. 000 01), and CD4/CD8 (MD = 0. 36, 95% CI:0. 28-0. 44, P < 0. 000 01), and relieve gastrointestinal reactions such as nausea and vomiting (OR = 0. 37, 95% CI: 0. 19-0. 71, P = 0. 003).

The analysis indicates that elemene can enhance the chemotherapeutic effect on NSCLC, improve the quality of life, and reduce adverse effect of platinum-contained chemotherapeutics, thereby being worth promoting in clinic.

Lung Cancer

Randomized controlled clinical trials related to the use of β>-elemene injection, as an adjunctive treatment for lung cancer, were retrieved from the Chinese Biomedical (CBMweb), Chinese Medical Current Content (CMCC), China National Knowledge Infrastructure (CNKI), ChinaInfo, Cochrane Central Register of Controlled Trials; MEDLINE, EMBASE, OVID and TCMLARS databases.

A total of 21 source documents (1,467 patients) matched pre-specified criteria for determining the effectiveness and safety of β>-elemene injection as an adjunctive treatment for lung cancer. Five studies involving 285 NSCLC patients reported a higher 24-month survival rate (39.09%) with the adjunctive treatment than with chemotherapy alone (26.17%; RR, 1.51; 95% CI, 1.03 to 2.21). Four studies involving 445 patients reported that the increased probability for improved performance status for patients treated with elemene-based combinations was higher than that of patients treated with chemotherapy alone (RR, 1.82; 95% CI, 1.45 to 2.29).

The results from a subgroup analysis on 12 studies involving 974 NSCLC patients and 9 studies involving 593 patients with both SCLC and NSCLC showed that the tumor control rate for NSCLC improved more in the elemene-based combinations treatment group (78.70%) than in the chemotherapy alone control group (71.31%; RR, 1.06; 95% CI, 1.00 to 1.12). The tumor response rate for NSCLC also improved more among patients treated with elemene based combinations (50.71%) than among patients treated with chemotherapy alone (38.04%; RR, 1.34; 95% CI, 1.17 to 1.54). The effectiveness of chemotherapy for the treatment of lung cancer may improve when combined with β-elemene injection as an adjunctive treatment. The combined treatment can result in an improved quality of life and prolonged survival (Wang et al., 2012).

Malignant Ascites

The effective combination therapy for malignant ascites, the therapeutic value of the combination of Endostar, a modified recombinant human endostatin, and β-elemene, an active component of a traditional Chinese herb, in an H22 mouse malignant ascites model was investigated by Jiang et al. (2012). The results of this study revealed that the combination therapy had significant synergistic effects on the inhibition of ascites formation and a deceased number of tumor cells and protein levels in ascites compared with the results of treatment with a single agent. A decreased peritoneal microvascular permeability and reduction in VEGF, MMP-2 and hypoxia inducible factor 1α(HIF1α) was noted in the combination group, when compared with single agent treatment.

These studies found that in the ascitic tumor cells, the protein levels of VEGF and MMP-2, as well as levels of VEGF mRNA, were significantly inhibited by the combination therapy. The potentiating effects of the combination of Endostar with β-elemene suggest that this novel therapy may yield an effective therapy for the treatment of malignant ascites.

Glioblastoma

Anti-proliferation of glioblastoma cells induced by beta-elemene was dependent on p38 MAPK activation. Treatment of glioblastoma cell lines with beta-elemene, led to phosphorylation of p38 MAPK, cell-cycle arrest in G0/G1 phase and inhibition of proliferation of these cells. Inhibition of p38 MAPK reversed beta-elemene-mediated anti-proliferation effect. Furthermore, the growth of glioblastoma cell-transplanted tumors in nude mice was inhibited by intraperitoneal injection of beta-elemene (Yao et al., 2008).

Breast Cancer; Chemotherapy

Beta-elemene had synergistic effect with Paclitaxel, and its possible mechanism might be correlated with down-regulating the cell-cycle protein cyclin-B1 expression and up-regulating the P27(kip1) expression. Beta-elemene (20 and 40 microg/mL respectively) and Paclitaxel (0.016 and 0.008 microg/mL respectively) synergistically inhibited cell proliferation of MB-468 breast cancer cells, with Q value > 1.15. Beta-elemene alone (52.59 microg/mL) apparently decreased the expression of cyclin-B1 protein. The expression of cyclin-B1 protein in the combined group was also lower than that in the PI group (1.698 microg/mL). The expression of P27(kip1) was up-regulated when compared with that in the betaI group or the PI group (Cai et al., 2013).

Gastric Cancer

TCM therapy applied in the 34 patients assigned in the TCM group (group I) included intravenous injection of Cinobufotalin, beta-elemene, or orally taking of anti-cancer Chinese herbs. The same TCM was also applied in the 36 patients of the combined treatment group (group II), but in combined use of FOLFOX chemotherapeutic protocol.

The median survival period in group II was 31 months, while it was 30 months in group I; the 1-, 2-, 3-year survival rates in group II were 88.89%, 84.38% and 59.26%, and those in the group I were 82.35%, 71.43% and 65.00%, respectively with insignificant difference between the two groups (chi2 = 0.298, P > 0.05); QOF in group I was significantly superior to that in group II (P < 0.05), and the adverse reaction occurrence was significantly less in group I than that in group II.

Chinese medicine treatment can improve the QOF and prolong the survival period of patients with progressive gastric cancer with few side-effects (Liu et al., 2008).

References

Jiang, Z.Y., Qin, S.K., Yin, X.J., Chen, Y.L., Zhu, L. (2012). Synergistic effects of Endostar combined with β-elemene on malignant ascites in a mouse model. Exp Ther Med, 4(2):277-284.

Liu X, Hua BJ. (2008). Effect of traditional Chinese medicine on quality of life and survival period in patients with progressive gastric cancer. Zhongguo Zhong Xi Yi Jie He Za Zhi, 28(2):105-7.

Wang, B., Peng, X.X., Sun, R., Li, J., Zhan, X.R., Wu, L.J., Wang, S.L., & Xie, T. (2012). Systematic review of β-elemene injection as adjunctive treatment for lung cancer. Chinese Journal of Integrative Medicine, 18(11), 8313-823.

Xu, X.W., Yuan, Z.Z., Hu, W.H., Wang, X.K. (2013). Meta-analysis on elemene injection combined with cisplatin chemotherapeutics in treatment of non-small-cell lung cancer. Zhongguo Zhong Yao Za Zhi, 38(9):1430-7.

Yao, Y.Q., Ding, X., Jia, Y.C, et al. (2008). Anti-tumor effect of beta-elemene in glioblastoma cells depends on p38 MAPK activation. Cancer Lett, 264(1):127-34. doi: 10.1016/j.canlet.2008.01.049.