Category Archives: type

Compound Zhebei Granules (CZG) 复方浙贝颗粒

Bulbus Thunberg Fritillaria (zhe bei mu)

RadixStephania tetrandra (han fang ji)

Rhizoma Chuanxiong (chuan xiong)

Compound Zhebei Granules (CZG) can increase the clinical remission rate for refractory acute leukemia during chemotherapy. Using a randomised, double-blind and multi-central concurrent control clinical research project, the patients conformed with the diagnostic criteria, according to the drug randomised method, were divided into a CZG group and a control group. The patients of the two groups respectively took the observation drug or a placebo 3 days before chemotherapy, and the therapeutic effects were evaluated after one course of chemotherapy.

The clinical complete remission (CR) rate was 42.3% in the CZG group with a total effective rate of 73.2%, and it was 25.8% in the control group with a total effective rate of 53.0%.

Source

Li DY, Huang S, Chen XY. Clinical observation of Compound Zhebei Granule in improving the survival time of refractory acute leukaemia patients.  Journal of Traditional Chinese Medicine. Volume 29, Issue 3, September 2009, Pages 190-194 doi:10.1016/S0254-6272(09)60063-7

Results demonstrated by Chen Xy, et al (2013) prove that the CZG in combination with chemotherapy can significantly improve chemotherapy remission rate after one cycle of treatment, showing good prospects for clinical application. In this multicenter double-blind, placebo-controlled clinical trial, they randomly assigned 238 patients who meet the diagnostic criteria of refractory acute leukaemia to receive chemotherapy combined with CZBG or chemotherapy plus placebo. 

There was statistically significant difference between the two arms according to Z/Cmh test (P<0.05). In the Per Protocol Set (PPS), the CR, CR + PR rates were 33.67%, 52.04% respectively in chemotherapy plus CZG arm and 24.24%, 37.37% in control arm. 

Source

Chen Xy, Hou L, Yang Sl, et al. Clinical study on Compound Zhe Bei Granules (CZG) combined with chemotherapy to improve the clinical efficacy of refractory acute leukaemia. Cancer Research. DOI: 10.1158/1538-7445.AM2013-4661 Published 15 April 2013 

 

Compared with the single treatment of doxorubicin group the groups the doxorubicin and CZG with dosage classified by three types(high, middle, low) decreased IOD of P-glycoprotein (PGP) lung resistance protein (LRP) and multidrug resistance associated protein (MRP) in K562/ A02 tumour xenografts with statistical significance (p0.05).

There no LRP expression in K562/A02 tumour xenografts in five groups. The combination of CZG and doxorubicin can decrease the expression of P-gpMRP in K562/A02 multidrug resistance tumour xenografts.

A drug-combination of Compound Zhe Bei Granule (CZG) and doxorubicin effects on the expression of Multidrug Resistance Associated Proteins in K562/A02 cell line multidrug resistance tumor xenografts in mice.

Source

Zheng Z, Wang X, Li Z-P, Zeng J-Q. 首届浙赣两省肿瘤研究交流会论文汇编 2012

 

CZG combining chemotherapy could reduce the percentages of CD34+ CD123+ and CD33+ CD123+ LSC, which might improve the clinical efficacy of refractory or relapsed acute myeloid leukemia (AML).

Seventy-eight patients with AML received bone marrow aspiration and the percentages of CD34+ CD123+ and CD33+ CD123+ cells were tested using flow cytometry method. A total of 24 refractory or relapsed AML patients were enrolled and treated with one cycle of standard chemotherapy combined with CZG.

Compared with refractory or relapsed AML patients, patients achieved remission had a significant lower percentage of CD34+ CD123+ cells (P<0.01) and CD33+ CD123+ cells (P<0.01), indicating that controlling the leukemia stem cell (LSC) percentage may be important for patients with AML to achieve sustainable remission.

Source

Wang J, Lai Z-l, Chen Y-y, et al. Effect of Compound Zhebei Granule (复方浙贝颗粒) combined with chemotherapy on surface markers of leukemia stem cell in patients with acute myeloid leukemia. Chinese Journal of Integrative Medicine. June 2016, Volume 22, Issue 6, pp 438-444

MDR

Ganoderma lucidum (Reishi)

Cancer: Ovarian
Action: Suppresses VEGF expression
Human ovarian cancer cells HO 8910 (HOCC) and human primary ovarian cells (HPOC) were treated with G. lucidum. Effects of G. lucidum treatment on cell proliferation were studied by MTT assay. The expression of vascular endothelial growth factor (VEGF) and connexin 43 (Cx43) were measured by immunohistochemistry and real time polymerase chain reaction. To study the molecular mechanism of CX43 mediated anti-tumor activity, small interference RNA (siRNA) was used to knockdown Cx43 expression in HOCC.
G. lucidum treatment resulted in reduced proliferation of HOCC. Inhibition of proliferation was accompanied by a decrease in VEGF expression and increase in Cx43 expression in the cancer cells. The extent of immune-reactivity of Cx43 or VEGF in cancer cells were correlated with the concentrations of G. lucidum used for treatment.
G. lucidum inhibits ovarian cancer by down-regulating the expression of VEGF and up-regulating the downstream Cx43 expression. G. lucidum may be a promising therapeutic agent for the treatment of ovarian cancer.

Source
Dai S, Liu J, Sun X, Wang N. Ganoderma lucidum inhibits proliferation of human ovarian cancer cells by suppressing VEGF expression and up-regulating the expression of connexin 43. BMC Complement Altern Med. 2014 Nov 5;14:434. doi: 10.1186/1472-6882-14-434.

Avocatin B

Cancer: Acute myeloid leukemia (AML)
Action: Mitochondrial localization
Lee et al., (2015) identified avocatin B, a lipid derived from avocado fruit, as a novel compound with cytotoxic activity in AML. Avocatin B reduced human primary AML cell viability without effect on normal peripheral blood stem cells. Functional stem cell assays demonstrated selectivity toward AML progenitor and stem cells without effects on normal hematopoietic stem cells.
Mechanistic investigations indicated that cytotoxicity relied on mitochondrial localization, as cells lacking functional mitochondria or CPT1, the enzyme that facilitates mitochondria lipid transport, were insensitive to avocatin B. Furthermore, avocatin B inhibited fatty acid oxidation and decreased NADPH levels, resulting in ROS-dependent leukemia cell death characterized by the release of mitochondrial proteins, apoptosis-inducing factor, and cytochrome c.
This study reveals a novel strategy for selective leukemia cell eradication based on a specific difference in mitochondrial function.

Source
Lee EA, Angka L, Rota S-G et al. Targeting Mitochondria with Avocatin B Induces Selective Leukemia Cell Death. Cancer Res June 15, 2015 75; 2478 doi: 10.1158/0008-5472.CAN-14-2676

Matrine

Cancer: Acute myeloid leukemia (AML)
Action: Induces apoptosis, reduces ratio of Bcl-2/Bax
Zhang et al., (2012) investigated Matrine’s anticancer effects and underlying mechanisms on human AML cells in vitro and in vivo. The results showed that matrine inhibited cell viability and induced cell apoptosis in AML cell lines as well as primary AML cells from patients with AML in a dose-and time-dependent manner.
Matrine induced apoptosis by collapsing the mitochondrial membrane potential, inducing cytochrome c release from mitochondria, reducing the ratio of Bcl-2/Bax, increasing activation of caspase-3, and decreasing the levels of p-Akt and p-ERK1/2. The apoptotic effects of matrine on AML cells were partially blocked by a caspase-3 inhibitor Z-DEVD-FMK and a PI3K/Akt activator IGF-1, respectively.
Matrine potently inhibited in vivo tumor growth following subcutaneous inoculation of HL-60 cells in SCID mice. These findings indicate that matrine can inhibit cell proliferation and induce apoptosis of AML cells and may be a novel effective candidate as chemotherapeutic agent against AML

Source
Zhang S, Zhang Y, Zhuang Y, et al. Matrine induces apoptosis in human acute myeloid leukemia cells via the mitochondrial pathway and Akt inactivation. PLoS One. 2012;7(10):e46853. doi: 10.1371/journal.pone.0046853.

 

Cancer: Osteosarcoma
Action: Activation of caspase-3,-8, and-9, upregulates Bax and Fas/FasL, downregulates of Bcl-2
Four human osteosarcoma cell lines: MG-63, U-2OS, Saos-2, and MNNG/HOS were treated by matrine and subjected to MTT assay, annexin V-FITC/PI double staining, and TUNEL assay. The activation of caspases and the expression of pro-apoptotic and anti-apoptotic factors were examined by qRT-PCR and Western blot.
Liang et al., (2012) found that matrine inhibited the proliferation and induced apoptosis of the four osteosarcoma cell lines in vitro and induced the activation of caspase-3,-8, and-9 in a dose-dependent manner. Furthermore, the pro-apoptotic factors Bax and Fas/FasL were upregulated, and the anti-apoptotic Bcl-2 was downregulated. More importantly our in vivo, studies showed that administration of matrine decreased tumor growth in a dose-dependent manner. Immunohistochemistry analysis demonstrated the downregulation of Bcl-2 and upregulation of Bax and Fas/FasL in MNNG/HOS tumor tissues following matrine treatment, consistent with the in vitro results.
Results demonstrate that matrine inhibits the proliferation and induces apoptosis of human osteosarcoma cells in vitro and in vivo. The induction of apoptosis appears to occur through the upregulation of Fas/FasL and Bax, downregulation of Bcl-2, and activation of caspase-3, -8, and -9, which then trigger major apoptotic cascades.

Source
Liang CZ, Zhang JK, Shi Z, Liu B, Shen CQ, Tao HM. Matrine induces caspase-dependent apoptosis in human osteosarcoma cells in vitro and in vivo through the upregulation of Bax and Fas/FasL and downregulation of Bcl-2. Cancer Chemother Pharmacol. 2012 Feb;69(2):317-31. doi: 10.1007/s00280-011-1699-4.

Cinnamaldehyde

Cancer: Leukemia, melanoma, colorectal

Action: Apoptosis, AP-1 transcriptional activity

Cinnamaldehyde is an active compound isolated from the stem bark of Cinnamomum cassia, a traditional oriental medicinal herb, which has been shown to inhibit tumor cell proliferation. In this study, Ka et al., (2003) investigated the effects of cinnamaldehyde on the cytotoxicity, induction of apoptosis and the putative pathways of its actions in human promyelocytic leukemia cells. Using apoptosis analysis, measurement of reactive oxygen species (ROS), and assessment of mitochondrial membrane potentials (Δψm), they show that cinnamaldehyde is a potent inducer of apoptosis and that it transduces the apoptotic signal via ROS generation, thereby inducing mitochondrial permeability transition (MPT) and cytochrome c release to the cytosol. Taken together, the data indicate that cinnamaldehyde induces the ROS-mediated mitochondrial permeability transition and resultant cytochrome c release. This is the first report on the mechanism of the anticancer effect of cinnamaldehyde.

Source
Ka H, Park H-J, Jung H-J, et al. Cinnamaldehyde induces apoptosis by ROS-mediated mitochondrial permeability transition in human promyelocytic leukemia HL-60 cells. Cancer Letters. Volume 196, Issue 2, 10 July 2003, Pages 143–152. doi:10.1016/S0304-3835(03)00238-6

To investigate the anti-tumor activities of several cinnamaldehyde derivatives, we compared the inhibitory effect of cinnamaldehyde derivatives on cell growth and AP-1 transcriptional activity in SW620 human colon cancer cells since AP-1 is a transcriptional factor implicated to control cancer cell growth. In further studies on the mechanism, Lee et al., (2007) found that consistent with the inhibitory effect on cell growth, 2′-hydroxycinnamaldehyde (HCA) dose-dependently (0 – 20 μg/ml) inhibited DNA binding activity of AP-1 accompanied with down regulation of c-Jun and c-Fos expressions. HCA also induced apoptotic cell death as well as expression of the apoptosis-regulating gene caspase-3, but inhibited the anti-apoptosis regulating gene bcl-2 in a dose-dependent manner. These results suggested that HCA has the most potent inhibitory effect against human colon cancer cell growth, and AP-1 may be an important target of HCA.

Source
Lee CW, Lee SH, Lee JW, et al. 2-Hydroxycinnamaldehyde Inhibits SW620 Colon Cancer Cell Growth Through AP-1 Inactivation. Journal of Pharmacological Sciences. Vol. 104 (2007) No. 1 P 19-28. http://doi.org/10.1254/jphs.FP0061204

Berberine

Cancer: Nasopharyngeal carcinoma
Action: Anti-inflammatory, inhibits STAT3
Growth inhibitory effects of berberine on multiple types of human cancer cells have been reported. Berberine inhibits invasion, induces cell cycle arrest and apoptosis in human cancer cells. The anti-inflammatory property of berberine, involving inhibition of Signal Transducer and Activator of Transcription 3 (STAT3) activation, has also been documented.
Berberine effectively inhibited the tumorigenicity and growth of an EBV-positive nasopharyngeal carcinoma (NPC) cell line (C666-1).
In vitro, berberine inhibited both constitutive and IL-6-induced STAT3 activation in NPC cells. Inhibition of STAT3 activation by berberine induced growth inhibition and apoptotic response in NPC cells. Tumor-associated fibroblasts were found to secret IL-6 and the conditioned medium harvested from the fibroblasts also induced STAT3 activation in NPC cells. Inhibition of tumorigenic growth of NPC cells in vivo was also correlated with effective inhibition of STAT3 activation.

Source
Sang CM, Cheung YC, Lui Vw-Y et al. Berberine suppresses tumorigenicity and growth of nasopharyngeal carcinoma cells by inhibiting STAT3 activation induced by tumor associated fibroblasts. BMC Cancer 2013, 13:619 doi:10.1186/1471-2407-13-619

Rheum officinale (emodin)

Cancer: lung, breast

Action: Growth inhibition, apoptosis

Human lung adenocarcinoma A549 and human breast cancer MCF-7 cell lines were treated with different concentrations of Rheum officinale (da huang) water extract at different time intervals. Growth inhibition was detected by MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] and colony formation assays; apoptosis was detected by cell morphologic analysis, DNA fragmentation analysis and COMET assay.
Da Huang water extract was found to have significant growth inhibitory effects on both A549 and MCF-7 cell lines with IC(50) values 620+/-12.7 and 515+/-10.1 microg/ml, respectively. Growth inhibitory effects were dose- and time-dependent. A significant decrease in cell number, DNA fragmentation and single DNA strand breakages were observed in the Da Huang water extract treated A549 and MCF-7 cells.
This suggests that the water extract of Da Huang exerts potential anticancer activity through growth inhibition and apoptosis on MCF-7 and A549 cells lines.

Source
Li WY, Chan SW, Guo DJ, Chung MK, Leung TY, Yu PH. Water extract of Rheum officinale Baill. induces apoptosis in human lung adenocarcinoma A549 and human breast cancer MCF-7 cell lines. J Ethnopharmacol. 2009 Jul 15;124(2):251-6. doi: 10.1016/j.jep.2009.04.030.

Epigallocatechin Gallate (EGCG)

Epigallocatechin Gallate (EGCG)
Curcumin
Cancer: Follicular lymphoma

Action: Regulates NF-κB, c-Myc, cyclooxygenase-2, induces apoptosis

NF-κB, c-Myc, cyclooxygenase-2, apoptosis

Treatment of patients with the combination of curcumin and EGCG, significantly lower cytoplasmic APE1 and the levels of the transcription factor were lower than those predicted from the effects of the CHOP agents (cyclophosphamide, doxorubicin, vincristine, and prednisone) alone, especially with a blunting of the remarkable increases in NF- κB activation induced by CHOP.

Cancer: Leukemia

Action: Inhibits NF-kB nuclearization and stimulation of matrix metalloproteinase-9 (MMP-9),

EGCG can inhibit proliferation and reduce the invasive potential of HTLV-1- positive leukemia cells. It apparently exerted its effects by suppressing Tax expression, manifested by inhibiting the activation of NF-kB pathway and induction of MMP-9 transcription in HTLV-1 positive cells.

Cancer: Lymphoma

Action: Decreases malignant cell proliferation

Co-treatment with EGCG and trichostatin A (TSA) decreased p16(INK4a) gene methylation, which coincided with increased p16(INK4a) mRNA and protein expression. Thus, EGCG and TSA synergistically reactivate p16(INK4a) gene expression in part through reducing promoter methylation, which may decrease human malignant lymphoma CA46 cell proliferation.

Cancer: Promyelocytic leukemia and non-Hodgkin’s lymphoma

Action: Suppresses cell growth

EGCG suppressed the cell growth of HL60 myeloid leukemia cells and Raji lymphoid leukemic cells independent of 67 LR expression. Moreover, there was no discernible change in the levels of intracellular reactive oxygen species, characteristics of apoptosis such as phosphatidylserine translocation and activated caspase-3.

Source
Bassiouny AR, Atteya MA, El-Rashidy FH, Neenaa HM. Curcumin and EGCG Suppress Apurinic/Apyrimidinic Endonuclease 1 and Induce Complete Remission in B-cell Non-Hodgkin’s lymphoma Patients. Functional Foods in Health and Disease 2011, 1(12):525-544

Harakeh S, Diab-Assaf M, Azar R, Hassan HM, et al. Epigallocatechin-3-gallate inhibits tax-dependent activation of nuclear factor kappa B and of matrix metalloproteinase 9 in human T-cell lymphotropic virus-1 positive leukemia cells. Asian Pac J Cancer Prev. 2014;15(3):1219-25.

Wu DS, Shen JZ, Yu AF, et al. Epigallocatechin-3-gallate and trichostatin A synergistically inhibit human lymphoma cell proliferation through epigenetic modification of p16INK4a. Oncol Rep. 2013 Dec;30(6):2969-75. doi: 10.3892/or.2013.2734.

Hazawa M, Takahashi K, Sugata S, Kashiwakura I. (-)-Epigallocatechin-3-O-gallate induces nonapoptotic cell death in leukemia cells independent of the 67 kDa laminin receptor. J Nat Prod. 2011 Apr 25;74(4):695-700. doi: 10.1021/np1007729.

Anti-Bladder-Tumor Effect of Baicalein from Scutellaria baicalensis Georgi and Its Application In Vivo

Cancer: Bladder

Action: reduces cell growth and inhibits cell invasion

Evidence-Based Complementary and Alternative Medicine
Volume 2013 (2013), Article ID 579751, 12 pages
http://dx.doi.org/10.1155/2013/579751

Research Article

Jin-Yi Wu,1 Kun-Wei Tsai,2 Yi-Zhen Li,1 Yi-Sheng Chang,1 Yi-Chien Lai,1 Yu-Han Laio,1 Jiann-Der Wu,3 and Yi-Wen Liu1
1Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, No. 300 Syuefu Road, Chiayi 600, Taiwan
2Department of Internal Medicine, Buddhist Tzuchi Dalin General Hospital, Dalin Town, Chiayi 622, Taiwan
3Department of Pathology, Chiayi Christian Hospital, Chiayi 600, Taiwan

Received 16 November 2012; Revised 22 January 2013; Accepted 5 February 2013

Academic Editor: Jen-Hwey Chiu

Copyright © 2013 Jin-Yi Wu et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Some phytochemicals with the characteristics of cytotoxicity and/or antimetastasis have generated intense interest among the anticancer studies. In this study, a natural flavonoid baicalein was evaluated in bladder cancer in vitro and in vivo. Baicalein inhibits 5637 cell proliferation. It arrests cells in G1 phase at 100 μM and in S phase below 75 μM. The protein expression of cyclin B1 and cyclin D1 is reduced by baicalein. Baicalein-induced p-ERK plays a minor role in cyclin B1 reduction. Baicalein-inhibited p65NF-κB results in reduction of cell growth. Baicalein-induced pGSK(ser9) has a little effect in increasing cyclin B1/D1 expression instead. The translation inhibitor cycloheximide blocks baicalein-reduced cyclin B1, suggesting that the reduction is caused by protein synthesis inhibition. On the other hand, neither cycloheximide nor proteasome inhibitor MG132 completely blocks baicalein-reduced cyclin D1, suggesting that baicalein reduces cyclin D1 through protein synthesis inhibition and proteasomal degradation activation. In addition, baicalein also inhibits cell invasion by inhibiting MMP-2 and MMP-9 mRNA expression and activity. In mouse orthotopic bladder tumor model, baicalein slightly reduces tumor size but with some hepatic toxicity. In summary, these results demonstrate the anti-bladder-tumor properties of the natural compound baicalein which shows a slight anti-bladder-tumor effect in vivo.

1. Introduction

Bladder cancer is the seventh most common type of cancer in worldwide man [1] and fourth in man of United States [2]. More than 90% of bladder cancers are transitional cell carcinoma (TCC), and approximately 80% of TCC belong to noninvasive papillary carcinoma that is a low-grade intraurothelial neoplasia with high recurrence. The other 20% of TCCs initiated from carcinoma in situ are at a high risk of processing to muscle invasive disease with a substantial risk for the development of distant metastasis [3, 4]. More than 10% of the low-grade papillary tumors eventually progress to high-grade muscle invasive bladder tumors. Most of the deaths from bladder cancer patients are due to invasive cancer metastasis [5], which has been a leading problem in the cancer therapy field. Multiple drugs chemotherapy has been applied for the therapy of metastatic bladder cancer; however, the adverse effect and resistance usually limit its clinical result. Therefore, some phytochemicals with the characteristics of cytotoxicity and/or antimetastasis have generated intense interest among the anticancer studies.

Baicalein, one of four major flavonoids existed in the root of Scutellaria baicalensis Georgi, has excellent antioxidant and anti-inflammatory activities [6, 7]. In traditional Chinese herb medicine, the root of Scutellaria baicalensis Georgi was usually gathered before Tomb-Sweeping Day and decocted for the purpose of “cleansing heart” and “removing toxins,” for example, cough with yellow sputum, jaundice, swelling and pain of eye, and so on. Wogonin, another one of the major flavonoids in the root of Scutellaria baicalensis Georgi, has been reported to reduce inflammatory cyclooxygenase-2 expression by c-Jun inhibition [8]. In addition to the anti-inflammatory effect of wogonin, baicalein has been reported to apply in cancer therapy by its cytotoxicity [9–11] and its anti-metastasis activity [12–14] recently. In human pancreatic cancer cells, 15~50 μM baicalein induces apoptotic cell death through downregulation of an antiapoptotic protein Mcl-1 [11]. In human bladder cancer cells, 60~80 μM baicalein retards cell growth by inhibiting CDC2 kinase activity [9]. Sixty μM baicalein also induces bladder cancer cells death, but baicalein-induced p-Akt and -H2AX expression plays a protective role against cell death [10]. Moreover, 10~50 μM baicalein inhibits cell migration and invasion through inhibiting MMP-2/9 activity in human hepatoma cells [12] and human breast cancer cells [13]. In human skin carcinoma, 40 μM baicalein inhibits cell invasion through inhibiting an anchor protein Ezrin expression [14]. Recently, baicalein is proven to be genotoxic without producing chromosomal alterations and mutagenesis which results in the severe side effect in cancer chemotherapy [15]. According to the above data, baicalein is a candidate worth development in anticancer therapy.

In this study, the anticancer effect of baicalein was analyzed in bladder cancer cells in vitro and in an orthotopic bladder tumor model in vivo. In vitro, the correlation of baicalein-induced change in Akt, ERK, p38, and p65NF-κB pathways and cell viability was analyzed. In vivo, the antitumor effect and renal and hepatic toxicities were evaluated.

2. Materials and Methods

2.1. Cell Culture and Drug Preparation

Human bladder papillary transitional cell carcinoma 5637 cells were obtained from the Bioresource Collection and Research Center (Hsinchu, Taiwan). Mouse bladder carcinoma MB49 cells were kindly provided by Dr. Timonthy L. Ratliff (Purdue Cancer Center, West Lafayette, IN, USA). 5637 and MB49 cells were maintained in RPMI 1640 medium supplied with 10% fetal bovine serum (FBS), 1% penicillin, and 1% streptomycin. Cells were incubated in a CO2 incubator at 37°C, with 5%  CO2 and 95% filtered air. Baicalein was isolated from the root of Scutellaria baicalensis Georgi, identified [16] and dissolved in DMSO. For culture cell assay, baicalein was added in culture medium containing 0.1% DMSO. For mouse assay, baicalein was intraperitoneally injected in mice containing 10% DMSO and 90% propylene glycol (0.8 mg/100 μL/mouse).

2.2. Reagents and Antibodies

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), propidium iodide (PI), ribonuclease A (RNase A), propylene glycol, MG-132, and crystal violet were purchased from Sigma (St. Louis, MO, USA). Anti-phospho-AKT(thr308), anti-phospho-AKT(ser473), and anti-cyclin D1 antibodies were purchased from Santa Cruz (Santa Cruz, CA, USA). Anti-α-tubulin, anti-β-actin, and anti-phospho-GSK-3β(ser9) antibodies were purchased from GeneTex (Taichung, Taiwan). Anti-cyclin B1 was purchased from Epitomics (Burlingame, CA, USA). Anti-Bub3 was purchased from BD Biosciences (San Jose, CA, USA). Anti-p65NF-κB, anti-phospho-ERK(thr202/tyr204), and anti-phospho-p38(thr180/tyr182) were purchased from Cell Signaling Technology (Danvers, MA, USA). The Millicell Hanging Cell Culture Inserts of Transwell system was purchased from Millipore (Billerica, MA, USA). Peroxidase-conjugated secondary antibodies were purchased from Jackson ImmunoResearch (West Grove, PA, USA).

2.3. Cell Viability Assay

Cell number was determined by colorimetric MTT assay. 5637 cells were cultured in 24-well plates at a density of 5 × 104 cells/well. After 24 h, cells were incubated with various concentrations of baicalein or 0.1% DMSO for another 24~72 h. Then MTT was added into medium for 2 h, the medium was discarded, and DMSO was added to dissolve the formazan product. Each well was measured by light absorbance at 490 nm. The result was expressed as a percentage, relative to 0.1% DMSO-treated control group.

2.4. Cell Cycle Analysis

Around 2 × 106 5637 cells were seeded in 100 mm dishes. After 24 h incubation for attachment, baicalein or DMSO was added. After baicalein treatment for 24 h and 48 h, cells were trypsinised, centrifuged, and fixed with ice-cold 75% ethanol overnight at 4°C. After removing the ethanol, cells were stained with a DNA staining solution (containing 1 mg/mL PI and 10 mg/mL RNase A dissolved in PBS) for 30 min at room temperature. The DNA content of the stained cells was measured using a FACScan flow cytometer. The cell doublets were removed by gating the left area of FL2-W/FL2-A plot for analysis. Cell cycle data from flow cytometry was analysed using ModFit LT software.

2.5. Cell Migration Assay

5637 cells were seeded in 6-well plates. After cells had reached confluence, a wound was made by a 200 μL plastic tip in each well. The wells were then washed twice with PBS to remove cell debris and then incubated with culture medium with DMSO (control) or baicalein. After 24 h incubation, each well was photographed by a phase contrast microscopy. The empty area was calculated by computer, and the cell mobility was calculated by (scratch area − empty area of baicalein treatment) × 100%/(scratch area − empty area of control). Measurements were performed in triplicate and presented as mean ± SE from three independent experiments.

2.6. Cell Invasion Assay

The invasion assay was analyzed using a Matrigel (BD Biosciences)-coated Transwell system (Millipore). The upper chamber of the transwell was coated with 25 μg Matrigel. 5637 cells (1 × 105) in serum-free RPMI-1640 media were seeded onto Matrigel-coated Transwell. The upper and lower chamber media were added baicalein or 0.1% DMSO. In the lower chambers, 10% FBS was added as a chemoattractant. After a 24 h incubation time, the cells that remained on the upper surface of the filter membrane were removed, and the cells on the opposite surface of the filter membrane were stained with 4% paraformaldehyde for 30 s and photographed under microscopy at 200x magnification. The number of migrated cells was counted in five randomly chosen microscope fields.

2.7. RT-PCR

Total RNA was isolated from cells. Reverse transcription (RT) was performed on 2 μg of total RNA by 1.5 μM random hexamer and RevertAid reverse transcriptase (Fermentas); then 1/20 volume of reaction mixture was used for PCR with MMP-2 specific primers (5′CTTCCAAGTCTGGAGCGATGT3′, 5′TACCGTCAAAGGGGTATCCAT3′), MMP-9 specific primers (5′AAGATGCTGCTGTTCAGCGGG3′, 5′GTCCTCTGGGCACTGCAGGAT3′), and GAPDH specific primers (5′CGGATTTGGTCGTATTGG3′, 5′AGATGGTGATGGGATTTC3′). The PCR products were analyzed by 1% agarose gel.

2.8. Gelatin-Zymography Assay

The enzymatic activities of MMP-2 and MMP-9 were determined by gelatin-zymography. 3 × 106 cells were seeded in 10 cm dish for 24 h and then maintained in serum-free medium with various concentrations of baicalein. The conditioned medium was collected 24 h after drug treatment, concentrated by using an Amicon Ultracel YM-10 filter. Twenty micrograms of protein obtained from the concentrated medium was mixed with nonreducing sample buffer and subjected to electrophoresis (8% SDS-PAGE copolymerize with 0.1% gelatin as substrate). The gel was washed twice (15 min/time) with 2.5% Triton X-100 and incubated at 37°C for 16~20 h in 50 mM Tris-HCl (pH 7.8), 10 mM CaCl2, and 0.01% NaN3. The gel was stained with 0.15% Coomassie brilliant blue R-250 and destained in 50% methanol and 10% acetic acid until the gelatinolytic activities were detected as clear bands against a blue background.

2.9. Mouse Orthotopic Bladder Tumor Model

The female C57BL/6 mice aged five to six weeks were provided by the National Laboratory Animal Center (Taipei, Taiwan) and maintained at our animal care facility for one week prior to use. The implantation of murine bladder cancer cells MB49 into C57BL/6 mice was carried out similarly as previous report [17, 18].After MB49 inoculation (day 1), mice were randomly assigned to two groups (10 mice/group). One group was intraperitoneally treated with vehicle (10% DMSO and 90% propylene glycol), and the other group received 0.8 mg/mouse baicalein intraperitoneally for 9 times. At the 21th day, the mice were sacrificed and the bladder volumes were measured before formalin fixation. After cutting into 4 μm sections, the slides of each mouse were confirmed under a microscope in histology by hematoxylin and eosin staining. The experiment was approved by the Institutional Animal Care and Use Committee of National Chiayi University.

2.10. Statistical Analysis

The values shown are mean ± SEM. Data are statistically evaluated by one-way ANOVA of SigmaPlot 11.0 and shown significantly different in , , and .

3. Results

3.1. Cytotoxicity and Proliferation Inhibition of Baicalein in 5637 Bladder Cancer Cells

Cytotoxicity of baicalein was analyzed by MTT assay. The result shows that baicalein dose-dependently inhibits cell viability after 24 h treatment (Figure 1(a)). Below 50 μM, baicalein did not induce cell death because there were no floating cells after treatment. When the concentration reached 100 μM, baicalein causes 33% cell number down with dead floating cells in the culture medium. To distinguish the fact of cell death and proliferation inhibition, the direct cell count analysis was applied after baicalein treatment. The result of Figure 1(b) suggests that under 50 μM, baicalein does not reduce total cell number after treatment for 72 h. Only for the concentration higher than 75 μM, baicalein induces cell death dose-dependently. These data suggest that baicalein induces growth inhibition at a dose lower than 50 μM and causes cell death at a dose higher than 75 μM in 5637 cells.

fig1
Figure 1: Effect of baicalein on cell growth. (a) Cytotoxicity of baicalein in 5637 bladder cancer cells. 5637 cells were initially seeded at 1 × 105 cells per well in 24-well plates and then treated with various concentrations of baicalein or vehicle (0.1% DMSO) for 24 h. The cell viability was measured by MTT assay. Measurement is performed from three independent experiments ( compared with vehicle). (b) Baicalein dose-dependently inhibits cell growth of 5637 cells. Cells were initially seeded at 1 × 105 cells (day-1) per well in 24-well plates and then treated with various concentrations of baicalein or vehicle (0.1% DMSO) for 24~72 h. The cell number was counted by trypan blue dye exclusion assay. The dotted line indicates the cell number on day 0.
3.2. Baicalein Induces Cell Cycle Arrest and Decreases Cyclin B1/D1 Expression of 5637 Bladder Cancer Cells

The cell cycle distribution changed by baicalein was analyzed by flow cytometric assay. Baicalein arrests cells in S phase after 24 h treatment at the concentration under 75 μM and in G1 phase at 100 μM. After treatment for 48 h, 100 μM baicalein continued to arrest cells in G1 without sub-G1 formation (Figure 2(a)). It suggests that baicalein, less than 50 μM, caused S phase arrest without significant cytotoxicity. One hundred μM baicalein arrested cells in G1 phase and induced cytotoxicity. One of G1/S transition promotion factors, cyclin D1, was dose-dependently decreased by baicalein (Figure 2(b)). It may contribute the reason to 100 μM baicalein-induced G1 arrest. Baicalein also decreased cyclin B1 expression dose-dependently (Figure 2(b)). Because cyclin B1 is an essential factor for entering G2/M phase, baicalein-decreased cyclin B1 may lead to S phase arrest. Baicalein immediately decreases cyclin D1 expression after treatment for 2 h (Figure 2(b)). It suggests that 100 μM baicalein effectively and quickly inhibits cell cycle progression at G1 phase.

579751.fig.002afig2
Figure 2: Effect of baicalein on cell cycle phase distribution in 5637 cells. (a) Baicalein induces cell cycle arrest. Cells were treated with vehicle or baicalein for 24 and 48 h, then were collected for cell cycle analysis (, , compared with vehicle). (b) Effect of baicalein on cyclin B1/D1 expression. Left, cells were treated with baicalein for 24 h. Right, cells were treated with 100 μM baicalein for 2, 6, 12, and 24 h.
3.3. Effect of Baicalein on the Regulation of Upstream Signal Factors

The intracellular signal factors p-GSK3β(ser9), p-AKT(thr308), p-AKT(ser473), p-ERK, and p-p38 were analyzed after baicalein treatment. The result of Figure 3(a) indicates that baicalein increases the phosphorylation of GSK3β(ser9), ERK(thr202/tyr204), and p38(thr180/tyr182). Though baicalein decreased the phosphorylation of AKT(thr308) and did not change p-AKT(ser473), the downstream GSK3β(ser9) was still phosphorylated at 24 h treatment. The time course of these baicalein-induced changes was also analyzed. As shown in Figure 3(b), baicalein inhibited p-AKT(thr308) phosphorylation from 2 h to 24 h; it suggests that baicalein inhibits Akt activity. GSK3β(ser9) was phosphorylated by baicalein from 2 h to 24 h, suggesting that baicalein also inhibits GSK3β activity. Both ERK and p38 pathways were early activated from 2 h to 24 h after baicalein treatment, ERK especially. The effect of baicalein on p65NF-κB was also analyzed. Without extracellular stimulation, most of p65NF-κB was found in cytoplasmic fraction in 5637 cells (Figure 3(c)). Baicalein dose-dependently inhibited the nuclear protein expression of p65NF-κB (Figure 3(c)). In summary, baicalein inhibits AKT and GSK3β activities, activates ERK and p38 pathways, and inhibits p65NF-κB-driven signals.

579751.fig.003afig3
Figure 3: Influence of baicalein on the signal pathways. (a) Effect of baicalein on the phosphorylation of GSK3β, AKT, ERK, and p38. 5637 cells were treated with 0.1% DMSO or baicalein for 24 h. (b) Time-course of baicalein-changed signal protein phosphorylation. 5637 cells treated with 100 μM baicalein or 0.1% DMSO for 2, 6, 12, and 24 h. The total cell lysates were extracted for western blot analysis. β-Actin was used as a loading control. (c) Effect of baicalein on the nuclear p65NF-κB expression. 5637 cells were treated with 0.1% DMSO or baicalein for 24 h. The cytoplasmic and nuclear extracts were prepared for Western blot analysis. α-Tubulin and Bub3 are the loading control of cytoplasmic and nuclear fraction, respectively.
3.4. Effect of Various Signal Protein Inhibitors on the Baicalein-Changed Cyclin B1/D1 Expression and Cell Viability

In order to understand the correlation between upstream signals and cyclin B1/D1 reduction, some specific inhibitors were used. Lithium chloride (LiCl) induces GSK3β(ser9) phosphorylation and inhibits GSK3βactivity [19, 20]. Baicalein or LiCl increased p-GSK3β(ser9), but only baicalein decreased cyclin B1/D1 expression (Figure 4(a)). It suggests that baicalein-inhibited cyclin B1/D1 expression is not mediated by GSK3βinhibition. On the contrary, LiCl dose-dependently increased cyclin B1/D1 expression, it suggests that baicalein-inhibited GSK3β pathway causes cyclin B1/D1 increase instead. LY294002, the inhibitor of PI3K-Akt pathway, inhibited the phosphorylation of AKT(ser473) but increased the phosphorylation of GSK3β(ser9) (Figure 4(b)). However, unlike baicalein, LY294002 did not reduce cyclin B1/D1 expression (Figure 4(b)). U0126, the inhibitors of MEK-ERK, slightly reversed baicalein-decreased cyclin B1 but not cyclin D1 (Figure 4(c)). The p38 kinase inhibitor SB203580 did not reverse baicalein-decreased cyclin B1/D1 expression (Figure 4(d)). Ro106-9920, an inhibitor of p65NF-κB, did not decrease cyclin B1/D1 expression (Figure 4(e)). These data indicate that baicalein-inhibited cyclin B1 is slightly mediated by ERK activation. The relationship of cell viability and baicalein-induced change in p-GSK3β(ser9), p-ERK, p-p38, and p65NF-κB was also analyzed. Using MTT assay (Figure 4(f)), MEK-ERK inhibitor U0126 and p38 kinase inhibitor SB203580 did not affect baicalein-reduced cell viability; the PI3K inhibitor LY294002 deteriorated baicalein-reduced cell viability; the p65NF-κB inhibitor Ro106-9920 reduced cell viability directly. It suggests that baicalein-induced p38, ERK, and GSK3β(ser9) phosphorylation does not play essential roles in cell growth inhibition. Only the baicalein-inhibited p65NF-κB activity leads to reduction of cell viability. In order to find out baicalein-reduced cyclin B1/D1 caused by de novo protein synthesis inhibition or proteasomal degradation stimulation, the translation inhibitor cycloheximide and the proteasome inhibitor MG132 were used for this study. After cyclohexamide treatment, baicalein did not reduce cyclin B1 anymore (Figure 4(g)). But baicalein still reduced cyclin D1 expression in the presence of cycloheximide or MG132 (Figure 4(g)). It suggests both de novo protein synthesis inhibition and proteasomal degradation stimulation are involved in baicalein-reduced cyclin D1 expression, and cyclin B1 decrease is only caused by de novo protein synthesis inhibition.

579751.fig.004afig4
Figure 4: Effect of various inhibitors on baicalein-reduced cyclin B1/D1 expression. (a–e) Effect of LiCl (a), LY294002 (b), U0126 (c), SB203580 (d), and Ro106-9920 (e) on baicalein-reduced cyclin B1/D1 expression. (f) Effect of various inhibitors on baicalein-inhibited cell viability. All above inhibitors were pretreated for 1 h and baicalein treatment for 24~72 h. The concentration of each chemicals: baicalein is 100 μM and others are 10 μM. (g) Effect of cycloheximide or MG132 on baicalein-reduced cyclin B1/D1 expression. Cycloheximide was pretreated for 30 min and baicalein treatment for 1 h in cyclin D1 detection and baicalein treatment for 6 h in cyclin B1 detection. MG132 was pretreated for 1 h and baicalein treatment for 6 h. The extracted cell lysates or nuclear proteins were analyzed by western blot.
3.5. Baicalein Blocks Migration and Invasion of 5637 Bladder Cancer Cells

Using scratch assay, baicalein dose-dependently inhibited cell migration (Figure 5(a)). At 100 μM, baicalein shows 60% inhibition in cell migration, which is more effective than the inhibition in cell viability (33% inhibition at 100 μM, Figure 1(a)). By matrigel-coated invasion assay, baicalein also shows a significant inhibition dose-dependently (Figure 5(b)). On the other hand, baicalein reduced MMP2 and MMP9 mRNA expression (Figure 5(c)) and enzymatic activity (Figure 5(d)) in 5637 cells. It suggests that the baicalein-inhibited MMP2/9 activity may contribute its anti-migration and anti-invasion activity.

579751.fig.005afig5
Figure 5: Anti-migration and anti-invasion activities of baicalein in 5637 cells. (a) Baicalein inhibits 5637 cell migration. Bottom chart is the percentage of migrated cells as control is 100%. (b) Baicalein inhibits 5637 cell invasion. Bottom chart is the percentage of invaded cells as control is 100%. (c) Effect of baicalein on the mRNA expression of MMP-2 and MMP-9. Bottom charts are the quantitative results from three independent experiments. (d) Effect of baicalein on the activities of MMP-2 and MMP-9. The conditioned medium was collected 24 h after drug treatment. Twenty micrograms of protein obtained from the concentrated medium was analyzed by gelatin-zymography assay. (, , compared with vehicle).
3.6. Baicalein Slightly Inhibits Tumor Growth with Some Hepatotoxicity in a Mouse Orthotopic Bladder Tumor Model In Vivo

Based on the antigrowth and antimetastasis activity of baicalein in cell assay, the in vivo antitumor assay was analyzed. After bladder cell implantation on day 1, baicalein treatment started on day 8. The treatment did not show toxicity in appearance and body weight (Figure 6(a)). Baicalein did not significantly reduce bladder size, but the mean bladder volume was still reduced in baicalein-treated mice (from 49.5 mm3 to 35.9 mm3 in Figure 6(b)). The blood biochemical analysis shows no significant change in serum BUN and creatinine between control and baicalein treatment groups, a little increase in GPT value but without statistical significance, and a significant increase in serum GOT (Table 1). It suggests that baicalein treatment causes some hepatic toxicity in mice.

table1tab1
Table 1: Effect of vehicle and baicalein on the plasma biochemical parameters of mice at termination of treatment. Values are mean ± SE.
579751.fig.006afig6
Figure 6: The antitumor effect of baicalein in vivo. (a) Mouse body weight and drug schedule in the mouse orthotopic bladder tumor model. After MB49 cell implantation, baicalein or vehicle was applied by intraperitoneal injection. Mouse body weight was recorded every day. At the 21th day, the mice were sacrificed. (b) Effect of baicalein in reducing bladder tumor size. The bladder volume of each mouse was measured. There are 9 mice survived at the 21th day in vehicle group and 7 in baicalein group.
4. Discussion

This study provides some new information about baicalein used in the anticancer therapy. In cell study, baicalein decreases cyclin B1 protein expression through inhibiting de novo protein synthesis and inhibits cyclin D1 by inhibiting protein synthesis and promoting proteasomal degradation. Baicalein-inhibited cyclin B1 is partially mediated by ERK activation. Among the signal transduction molecules AKT, GSK3β, ERK, p38, and p65NF-κB, p65NF-κB inhibition plays the most important role in baicalein-reduced cell viability. In mouse orthotopic bladder tumor model, baicalein has a little inhibition effect on orthotopic bladder tumor growth but with some hepatic toxicity.

Baicalein produces different cytotoxicity in different cell lines. For example, it causes cell cycle arrest at G1 phase in breast cancer [21] and oral squamous cell carcinoma [22], at S phase in lung nonsmall carcinoma cell [23] and at G2/M phase in BFTC905 bladder cancer cells [9]. The differences may be caused by different doses and different cells used. In 5637 bladder cancer cells, baicalein arrests cells at S phase under 75 μM and at G1 phase at 100 μM without apoptotic cells (Figure 2(a)). In pancreatic carcinoma PaCa cells [11], bladder cancer BFTC905 cells [9], and colorectal carcinoma HCT116 cells [10], baicalein induces cell apoptotic death at the dose between 5 to 60 μM. Baicalein also has a wide range on cytotoxicity of different cell lines, the IC50 is under 20 μM in gastric cancer cells AGS and MKN-28 [24], prostate carcinoma LNCaP [25], and JCA-1 [26], between 20 to 50 μM in leukemia HL-60 [27], bladder cancer BFTC905 [9], hepatic cancer Hep G2 [28], and myeloma cell U266 [29], and more than 100 μM in 5637 bladder cancer (Figure 1(a)), oral squamous carcinoma HSC-3 [22], leukemia THP-1, and osteogenic cancer cell HOS [30]. Although the detail mechanisms about the wide-range cytotoxicity are still unclear, this property may provide a specific and lower hazard anticancer effect for the higher sensitive tumors.

PI3K, the upstream signal of AKT, has been reported to be inhibited by baicalein [31]. In our study, baicalein inhibits pAKT(thr308) phosphorylation and has no influence on pAKT(ser473) (Figure 2(b)). The pAKT inhibition phenomenon also has been reported in prostate cancer cell DU145 [32] and oral squamous carcinoma HSC-3 [22]. On the other hand, the pAKT(ser473) activation has been reported in bladder cancer BFTC905 [9, 10]. Because the pAKT(thr308) phosphorylation site is the direct target site for PI3K-PDK1 [33], it will be downregulated after PI3K inhibition by baicalein (Figure 4(b)). The ser473 site of AKT is phosphorylated by rictor-mTOR [34], not PI3K; therefore, it may be the reason for the no effect of baicalein on the phosphorylation of pAKT(ser473). Even though the pAKT(thr308) is decreased by baicalein, pGSK3β(ser9), one AKT downstream [35], is still phosphorylated by baicalein (Figure 3(b)). Because the phosphorylation of pGSK3β(ser9) is achieved by numerous kinases, not only AKT [35], baicalein-induced pGSK3β(ser9) may be caused by the influence of other kinase(s).

NF-κB, an important inflammatory transcription factor, is inhibited by baicalein in 5637 cells (Figure 3(c)). Baicalein-inhibited p65NF-κB activation has also been reported in human mast cells [36], mouse macrophages [37, 38], human myeloma cells [29], and brain microglia [39, 40]. According to the important role of p65NF-κB in tumor progression and metastasis [41, 42], the inhibition of baicalein on nuclear NF-κB is a critical function in its anti-inflammation and anticancer application. In human hepatoma cells, baicalein shows anti-migration property with NF-κB inhibition [12]. There is one report indicates that GSK3β inhibition results in inhibiting NF-κB activity [43]; therefore, the mechanism of baicalein-inhibited p65NF-κB activity may be partially mediated by baicalein-inhibited GSK3β. In Figure 4(f), among the 4 signal inhibitors, the cell viability decreases at most by NF-κB inhibitor Ro106-9920, which indicates that NF-κB is a critical factor for proliferation of 5637 cells.

Baicalein inhibits the protein expression of cyclin B1 [9, 23, 44] and cyclin D1 [22, 32, 44], has also been reported by some studies, but the mechanism is still unclear. In this study, we first suggest that baicalein decreases cyclin B1 expression through inhibiting de novo protein synthesis but not promoting proteasomal degradation and decreases cyclin D1 by both ways (Figure 4(g)). On the other hand, cyclin B1 reduction is partially mediated by ERK activation (Figure 4(c)). Luteolin, a natural flavonoid with structure similar to baicalein, decreases cyclin D1 expression by increasing proteasomal degradation [45]. Though the structures are similar between baicalein and luteolin, the mechanisms for cyclin D1 reduction are different. Luteolin enhances proteasomal degradation via decreasing GSK3β(ser9) phosphorylation, but baicalein increases GSK3β(ser9) phosphorylation (Figures 3(a) and 3(b)). Therefore, there is (are) other pathway(s) for inducing cyclin D1 degradation by baicalein.

In addition to anti-proliferation, baicalein also inhibits cancer cell metastasis. Either in scratch assay (Figure 5(a)) or in Matrigel-coated transwell assay (Figure 5(b)), they point out the anti-migration and anti-invasion property of baicalein. In this study, we confirm this inhibition, like others [12, 13], mediated by inhibiting MMP-2/9 activities (Figure 5(d)). The correlated signal pathways need to be further investigated. In the orthotopic bladder tumor model, baicalein shows a little effect on inhibiting bladder tumor growth (Figure 6). One report indicates that baicalein significantly reduces tumor volume in a nude mice model [12]. Comparing these two animal models, we use higher dose and lower frequency of baicalein, which may result in the lower efficiency. But it still notices that baicalein induces hepatic toxicity with GOT value increase. In order to avoid hepatic toxicity, it is better to use baicalein locally, for example, by intravesical application for bladder tumor therapy.

5. Conclusions

In this study, baicalein decreases cyclin D1 protein expression through inhibiting de novo protein synthesis and promoting proteasomal degradation and decreases cyclin B1 by inhibiting de novo protein synthesis. Baicalein-inhibited cyclin B1 expression is slightly mediated by ERK activation. The mechanism of baicalein in anti-proliferation and anti-metastasis is concluded in Figure 7. Among the signal transduction molecules AKT, GSK3β, ERK, p38, and p65NF-κB, p65NF-κB inhibition plays the most important role in baicalein-reduced cell viability. In mouse orthotopic bladder tumor model, baicalein has a little effect on orthotopic bladder tumor growth inhibition but with some hepatic toxicity.

579751.fig.007fig7
Figure 7: The pathway scheme of baicalein on the cell proliferation inhibition and cell metastasis inhibition in human bladder cancer cell 5637. The anti-proliferation and anti-metastasis activities of baicalein may contribute the anti-bladder-tumor effect in vivo.
Abbreviations

FBS: Fetal bovine serum
MMP-2: Matrix metalloproteinase-2
MMP-9: Matrix metalloproteinase-9
MTT: 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide
NF-κB: Nuclear factor-kappaB
PI3K: Phosphatidylinositol 3-kinase.
Conflict of Interests

The authors declare that there is no conflict of interests.

Authors’ Contribution

J.-Y. Wu and K.-W. Tsai contributed equally to this work.

Acknowledgments

This work is supported by Grants from the National Science Council NSC101-2320-B-415-002-MY3 of the Republic of China and Buddhist Tzuchi Dalin General Hospital, Dalin Town, Chiayi, Taiwan.

References

1 A. Jemal, F. Bray, M. M. Center, J. Ferlay, E. Ward, and D. Forman, “Global cancer statistics,” CA Cancer Journal for Clinicians, vol. 61, no. 2, pp. 69–90, 2011. View at Publisher · View at Google Scholar · View at Scopus
2 R. Siegel, D. Naishadham, and A. Jemal, “Cancer statistics 2012,” A Cancer Journal for Clinicians, vol. 62, no. 1, pp. 10–29, 2012.
3 D. J. McConkey, S. Lee, W. Choi et al., “Molecular genetics of bladder cancer: emerging mechanisms of tumor initiation and progression,” Urologic Oncology, vol. 28, no. 4, pp. 429–440, 2010. View at Publisher· View at Google Scholar · View at Scopus
4 X. R. Wu, “Urothelial tumorigenesis: a tale of divergent pathways,” Nature Reviews Cancer, vol. 5, no. 9, pp. 713–725, 2005. View at Publisher · View at Google Scholar · View at Scopus
5 L. C. Pagliaro and P. Sharma, “Review of metastatic bladder cancer,” Minerva Urologica e Nefrologica, vol. 58, no. 1, pp. 53–71, 2006. View at Scopus
6 C. C. Lin and D. E. Shieh, “The anti-inflammatory activity of Scutellaria rivularis extracts and its active components, baicalin, baicalein and wogonin,” American Journal of Chinese Medicine, vol. 24, no. 1, pp. 31–36, 1996. View at Scopus
7 Z. H. Shao, T. L. Vanden Hoek, Y. Qin et al., “Baicalein attenuates oxidant stress in cardiomyocytes,” American Journal of Physiology, vol. 282, no. 3, pp. H999–H1006, 2002. View at Scopus
8 L. G. Chen, L. Y. Hung, K. W. Tsai et al., “Wogonin, a bioactive flavonoid in herbal tea, inhibits inflammatory cyclooxygenase-2 gene expression in human lung epithelial cancer cells,” Molecular Nutrition and Food Research, vol. 52, no. 11, pp. 1349–1357, 2008. View at Publisher · View at Google Scholar · View at Scopus
9 J. I. Chao, W. C. Su, and H. F. Liu, “Baicalein induces cancer cell death and proliferation retardation by the inhibition of CDC2 kinase and survivin associated with opposite role of p38 mitogen-activated protein kinase and AKT,” Molecular Cancer Therapeutics, vol. 6, no. 11, pp. 3039–3048, 2007. View at Publisher · View at Google Scholar · View at Scopus
10 R. H. Jiang, W. C. Su, H. F. Liu, H. S. Huang, and J. I. Chao, “Opposite expression of securin and γ-H2AX regulates baicalein-induced cancer cell death,” Journal of Cellular Biochemistry, vol. 111, no. 2, pp. 274–283, 2010. View at Publisher · View at Google Scholar · View at Scopus
11 H. Takahashi, M. C. Chen, H. Pham et al., “Baicalein, a component of Scutellaria baicalensis, induces apoptosis by Mcl-1 down-regulation in human pancreatic cancer cells,” Biochimica et Biophysica Acta, vol. 1813, no. 8, pp. 1465–1474, 2011. View at Publisher · View at Google Scholar · View at Scopus
12 Y. W. Chiu, T. H. Lin, W. S. Huang, et al., “Baicalein inhibits the migration and invasive properties of human hepatoma cells,” Toxicology and Applied Pharmacology, vol. 255, no. 3, pp. 316–326, 2011.
13 L. Wang, Y. Ling, Y. Chen et al., “Flavonoid baicalein suppresses adhesion, migration and invasion of MDA-MB-231 human breast cancer cells,” Cancer Letters, vol. 297, no. 1, pp. 42–48, 2010. View at Publisher · View at Google Scholar · View at Scopus
14 B. Wu, J. Li, D. Huang, et al., “Baicalein mediates inhibition of migration and invasiveness of skin carcinoma through Ezrin in A431 cells,” BMC Cancer, vol. 11, article 527, 2011.
15 J. T. Fox, S. Sakamuru, R. Huang, et al., “High-throughput genotoxicity assay identifies antioxidants as inducers of DNA damage response and cell death,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 14, pp. 5423–5428, 2012.
16 J. Y. Wu, K. T. Chung, Y. W. Liu et al., “Synthesis and biological evaluation of novel C(6) modified baicalein derivatives as antioxidative agents,” Journal of Agricultural and Food Chemistry, vol. 56, no. 8, pp. 2838–2845, 2008. View at Publisher · View at Google Scholar · View at Scopus
17 C. H. Shen, J. J. Shee, J. Y. Wu, Y. W. Lin, J. D. Wu, and Y. W. Liu, “Combretastatin A-4 inhibits cell growth and metastasis in bladder cancer cells and retards tumour growth in a murine orthotopic bladder tumour model,” British Journal of Pharmacology, vol. 160, no. 8, pp. 2008–2027, 2010. View at Publisher · View at Google Scholar · View at Scopus
18 S. Y. Wu, Y. R. Lee, C. C. Huang, et al., “Curcumin-induced heme oxygenase-1 expression plays a negative role for its anti-cancer effect in bladder cancers,” Food and Chemical Toxicology, vol. 50, no. 10, pp. 3530–3536, 2012.
19 P. S. Klein and D. A. Melton, “A molecular mechanism for the effect of lithium on development,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 16, pp. 8455–8459, 1996. View at Publisher · View at Google Scholar · View at Scopus
20 F. Zhan, C. J. Phiel, L. Spece, N. Gurvich, and P. S. Klein, “Inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3) in response to lithium: evidence for autoregulation of GSK-3,” Journal of Biological Chemistry, vol. 278, no. 35, pp. 33067–33077, 2003. View at Publisher · View at Google Scholar· View at Scopus
21 Q. M. Zhou, S. Wang, H. Zhang et al., “The combination of baicalin and baicalein enhances apoptosis via the ERK/p38 MAPK pathway in human breast cancer cells,” Acta Pharmacologica Sinica, vol. 30, no. 12, pp. 1648–1658, 2009. View at Publisher · View at Google Scholar · View at Scopus
22 Y. H. Cheng, L. A. Li, P. Lin, et al., “Baicalein induces G1 arrest in oral cancer cells by enhancing the degradation of cyclin D1 and activating AhR to decrease Rb phosphorylation,” Toxicology and Applied Pharmacology, vol. 263, no. 3, pp. 360–367, 2012.
23 H. W. C. Leung, W. H. Yang, M. Y. Lai, C. J. Lin, and H. Z. Lee, “Inhibition of 12-lipoxygenase during baicalein-induced human lung nonsmall carcinoma H460 cell apoptosis,” Food and Chemical Toxicology, vol. 45, no. 3, pp. 403–411, 2007. View at Publisher · View at Google Scholar · View at Scopus
24 B. C. Y. Wong, W. P. Wang, C. H. Cho et al., “12-Lipoxygenase inhibition induced apoptosis in human gastric cancer cells,” Carcinogenesis, vol. 22, no. 9, pp. 1349–1354, 2001. View at Scopus
25 M. Bonham, J. Posakony, I. Coleman, B. Montgomery, J. Simon, and P. S. Nelson, “Characterization of chemical constituents in Scutellaria baicalensis with antiandrogenic and growth-inhibitory activities toward prostate carcinoma,” Clinical Cancer Research, vol. 11, no. 10, pp. 3905–3914, 2005. View at Publisher · View at Google Scholar · View at Scopus
26 S. Chen, Q. Ruan, E. Bedner et al., “Effects of the flavonoid baicalin and its metabolite baicalein on androgen receptor expression, cell cycle progression and apoptosis of prostate cancer cell lines,” Cell Proliferation, vol. 34, no. 5, pp. 293–304, 2001. View at Publisher · View at Google Scholar · View at Scopus
27 M. K. Roy, K. Nakahara, V. Na Thalang et al., “Baicalein, a flavonoid extracted from a methanolic extract of Oroxylum indicum inhibits proliferation of a cancer cell line in vitro via induction of apoptosis,” Pharmazie, vol. 62, no. 2, pp. 149–153, 2007. View at Publisher · View at Google Scholar · View at Scopus
28 W. H. Chang, C. H. Chen, and F. J. Lu, “Different effects of baicalein, baicalin and wogonin on mitochondrial function, glutathione content and cell cycle progression in human hepatoma cell lines,” Planta Medica, vol. 68, no. 2, pp. 128–132, 2002. View at Publisher · View at Google Scholar · View at Scopus
29 Z. Ma, K. I. Otsuyama, S. Liu et al., “Baicalein, a component of Scutellaria radix from Huang-Lian-Jie-Du-Tang (HLJDT), leads to suppression of proliferation and induction of apoptosis in human myeloma cells,” Blood, vol. 105, no. 8, pp. 3312–3318, 2005. View at Publisher · View at Google Scholar · View at Scopus
30 M. Himeji, T. Ohtsuki, H. Fukazawa et al., “Difference of growth-inhibitory effect of Scutellaria baicalensis-producing flavonoid wogonin among human cancer cells and normal diploid cell,” Cancer Letters, vol. 245, no. 1-2, pp. 269–274, 2007. View at Publisher · View at Google Scholar · View at Scopus
31 D. Kong, Y. Zhang, T. Yamori, H. Duan, and M. Jin, “Inhibitory activity of flavonoids against class I phosphatidylinositol 3-kinase isoforms,” Molecules, vol. 16, no. 6, pp. 5159–5167, 2011. View at Publisher· View at Google Scholar · View at Scopus
32 G. P. Pidgeon, M. Kandouz, A. Meram, and K. V. Honn, “Mechanisms controlling cell cycle arrest and induction of apoptosis after 12-lipoxygenase inhibition in prostate cancer cells,” Cancer Research, vol. 62, no. 9, pp. 2721–2727, 2002. View at Scopus
33 L. Stephens, K. Anderson, D. Stokoe et al., “Prohtein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B,” Science, vol. 279, no. 5351, pp. 710–714, 1998. View at Publisher · View at Google Scholar · View at Scopus
34 D. D. Sarbassov, D. A. Guertin, S. M. Ali, and D. M. Sabatini, “Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex,” Science, vol. 307, no. 5712, pp. 1098–1101, 2005. View at Publisher · View at Google Scholar · View at Scopus
35 R. S. Jope, C. J. Yuskaitis, and E. Beurel, “Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics,” Neurochemical Research, vol. 32, no. 4-5, pp. 577–595, 2007. View at Publisher · View at Google Scholar · View at Scopus
36 C. J. Hsieh, K. Hall, T. Ha, C. Li, G. Krishnaswamy, and D. S. Chi, “Baicalein inhibits IL-1β- and TNF-α-induced inflammatory cytokine production from human mast cells via regulation of the NF-κB pathway,” Clinical and Molecular Allergy, vol. 5, article 5, 2007. View at Publisher · View at Google Scholar · View at Scopus
37 B. Y. Kang, S. W. Chung, S. H. Kim, D. Cho, and T. S. Kim, “Involvement of nuclear factor-κB in the inhibition of interleukin-12 production from mouse macrophages by baicalein, a flavonoid in Scutellaria baicalensis,” Planta Medica, vol. 69, no. 8, pp. 687–691, 2003. View at Publisher · View at Google Scholar · View at Scopus
38 M. H. Kim, S. Y. Ryu, M. A. Bae, J. S. Choi, Y. K. Min, and S. H. Kim, “Baicalein inhibits osteoclast differentiation and induces mature osteoclast apoptosis,” Food and Chemical Toxicology, vol. 46, no. 11, pp. 3375–3382, 2008. View at Publisher · View at Google Scholar · View at Scopus
39 C. J. Chen, S. L. Raung, S. L. Liao, and S. Y. Chen, “Inhibition of inducible nitric oxide synthase expression by baicalein in endotoxin/cytokine-stimulated microglia,” Biochemical Pharmacology, vol. 67, no. 5, pp. 957–965, 2004. View at Publisher · View at Google Scholar · View at Scopus
40 K. Suk, H. Lee, S. S. Kang, G. J. Cho, and W. S. Choi, “Flavonoid baicalein attenuates activation-induced cell death of brain microglia,” Journal of Pharmacology and Experimental Therapeutics, vol. 305, no. 2, pp. 638–645, 2003. View at Publisher · View at Google Scholar · View at Scopus
41 J. I. Inoue, J. Gohda, T. Akiyama, and K. Semba, “NF-κB activation in development and progression of cancer,” Cancer Science, vol. 98, no. 3, pp. 268–274, 2007. View at Publisher · View at Google Scholar · View at Scopus
42 G. Sethi, B. Sung, and B. B. Aggarwal, “Nuclear factor-κB activation: from bench to bedside,” Experimental Biology and Medicine, vol. 233, no. 1, pp. 21–31, 2008. View at Publisher · View at Google Scholar · View at Scopus
43 J. Deng, W. Xia, S. A. Miller, Y. Wen, H. Y. Wang, and M. C. Hung, “Crossregulation of NF-κB by the APC/GSK-3β/β-catenin pathway,” Molecular Carcinogenesis, vol. 39, no. 3, pp. 139–146, 2004. View at Publisher · View at Google Scholar · View at Scopus
44 H. Z. Lee, H. W. C. Leung, M. Y. Lai, and C. H. Wu, “Baicalein induced cell cycle arrest and apoptosis in human lung squamous carcinoma CH27 cells,” Anticancer Research, vol. 25, no. 2, pp. 959–964, 2005.View at Scopus
45 C. S. Ong, J. Zhou, C. N. Ong, and H. M. Shen, “Luteolin induces G1 arrest in human nasopharyngeal carcinoma cells via the Akt-GSK-3β-cyclin D1 pathway,” Cancer Letters, vol. 298, no. 2, pp. 167–175, 2010. View at Publisher · View at Google Scholar · View at Scopus

δ-Elemene (delta Elemene)

Cancer: Lung
Action: Induces apoptosis, inhibits NF-kappa B
δ-elemene significantly induced apoptosis of NCI-H292, as shown by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, DNA fragmentation measurement, Annexin V (AnV) binding of externalized phosphatidylserine and the mitochondrial probe JC-1 using flow cytometry.

Treatment of NCI-H292 with δ-elemene increased both p38 mitogen-activated protein kinase (MAPK) and inducible nitric oxide synthese (iNOS) levels, suggesting these two molecules maybe relate to the apoptotic effect of δ-elemene. The cells with Bcl-2 or Bcl-xL over-expression showed an elevation of nuclear factor kappa B (NF-kappa B) activity, accompanying a significant reduction of δ-elemene-induced apoptosis.

Furthermore, inhibition of NF-kappa B by IkBαSR, which is a powerful inhibitor of NF-kappa B, restored the ability of δ-elemene to induce apoptosis in the cells transfected with Bcl-2. These data strongly indicated that the apoptotic effect of δ-elemene on NCI-H292 was closely associated with the activity of NF-kappa B, which was up-regulated by Bcl-2 and Bcl-xL.
In conclusion, δ-elemene induced apoptosis in NCI-H292 cells. The apoptotic effect of δ-elemene could be significantly offset by over-expression of either Bcl-2 or Bcl-xL. Bcl-2 and Bcl-xL were able to increase the activity of NF-kappa B, which was a known anti-apoptotic molecule in human lung cancer cells.

Source
Xie CY, Yang W, Ying J, et al. B-cell lymphoma-2 over-expression protects δ-elemene-induced apoptosis in human lung carcinoma mucoepidermoid cells via a nuclear factor kappa B-related pathway. Biol Pharm Bull. 2011;34(8):1279-86.

Oridonin phosphate

Cancer: Breast

Action: Induces apoptosis, up-regulates Bax and down-reguates Bcl-2, induces autophagy

Oridonin phosphate is one of the derivatives of oridonin. The results by Li et al., (2015) demonstrated that oridonin phosphate inhibited the proliferation of MDA-MB-436 and MDA-MB-231 cells in a dose- and time-dependent manner. Cell apoptosis rate was detected in oridonin phosphate-treated breast cancer cells and the data demonstrated that oridonin phosphate induced cell apoptosis of breast cancer cells in time- and dose-dependent manner.

The results showed that the expression level of Bax was up-regulated and the expression level of Bcl-2 was down-regulated. Meanwhile, the level of cleaved caspase-9 was significantly increased when the cells were treated with 40 μM of oridonin phosphate for 48 h, although the expression level of pro-caspase-9 was not obviously changed. All of the data revealed that mitochondrial apoptosis pathway may be involved in the cell apoptosis induced by oridonin phosphate in breast cancer cells.

Additionally, they further explored the relationship between apoptosis and autophagy specifically induced by oridonin phosphate in breast cancer cells. Taken together, the compound of oridonin phosphate simultaneously induced cell apoptosis and autophagy in breast cancer cells.

Inhibition oridonin phosphate-induced cell autophagy suppressed the progression of cell apoptosis, which revealed that oridonin phosphate-induced autophagy participated in up-regulation of apoptosis in human breast cancer cells.

Source
Li Y, Wang Y, Wang S, Gao Y, Zhang X, Lu C. Oridonin phosphate-induced autophagy effectively enhances cell apoptosis of human breast cancer cells. Med Oncol. 2015 Jan;32(1):365. doi: 10.1007/s12032-014-0365-1.

Berberine

Cancer: Multiple Myeloma

Action: Down-regulates miR-21 levels through IL6/STAT3

Berberine is known to modulate microRNA (miRNA) levels, although the mechanism for this action is unknown. Luo et al. previously demonstrate that the expression of 87 miRNAs is differentially affected by berberine in multiple myeloma cells. Among 49 miRNAs that are down-regulated, nine act as oncomirs, including miR-21. Integrative analysis showed that 28 of the down-regulated miRNAs participate in tumor protein p53 (TP53) signaling and other cancer pathways. miR-21 is involved in all these pathways, and is one of the most important oncomirs to be affected by berberine in multiple myeloma cells.

They confirmed that berberine down-regulated miRNA-21 expression and significantly up-regulated the expression of programmed cell death 4 (PDCD4), a predicted miR-21 target. Depletion of PDCD4 by short interfering RNA could rescue berberine-induced cytotoxicity in multiple

Results suggest that berberine suppresses multiple myeloma cell growth, at least in part, by down-regulating miR-21 levels possibly through IL6/STAT3. This led to increased PDCD4 expression, which is likely to result in suppression of the p53 signaling pathway.

Source

Luo X, Gu J, Zhu R, et al. Integrative analysis of differential miRNA and functional study of miR-21 by seed-targeting inhibition in multiple myeloma cells in response to berberine. BMC Syst Biol. 2014 Jul 7;8:82. doi: 10.1186/1752-0509-8-82

 

 

Cancer: Multiple Myeloma

Action: Triggers hypomethylation

Berberine reduces the proliferation and induces apoptosis in the multiple myeloma cell line, U266.Qing et al., (2014) explored the detailed mechanism by analysing the gene expression profiles in U266 treated with or without berberine. DNMT1 andDNMT3B, encoding for a highly conserved member of the DNA methyltransferases, decreased significantly. Results show that berberine can repress the expression of DNMT1 and DNMT3B, which triggers hypomethylation of TP53 by changing the DNA methylation level and the alteration of p53 dependent signal pathway in human multiple melanoma cell U266.

Source

Qing Y, Hu H, Liu Y, Feng T, Meng W, Jiang L, Sun Y, Yao Y. Berberine induces apoptosis in human multiple myeloma cell line U266 through hypomethylation of p53 promoter. Cell Biol Int. 2014 May;38(5):563-70.

Emodin

Cancer: Multiple Myeloma

Action: Inhibits interleukin-6, activates of caspase-3 and caspase-9

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

Source

Muto A, Hori M, Sasaki Y, et al. Emodin has a cytotoxic activity against human multiple myeloma as a Janus-activated kinase 2 inhibitor. Mol Cancer Ther. 2007 Mar;6(3):987-94.

Cancer: Multiple myeloma

Action: Induces apoptosis

Although novel drugs have contributed immensely to improving outcomes of patients with multiple myeloma (MM), many patients develop drug resistance and ultimately succumb to MM. Papanikolaou et al., (2014) show that artesunate, an anti-malarial drug, reliably induces cell death in vitro in naïve as well as drug-resistant MM cells at concentrations shown to be safe in humans. Artesunate induced apoptosis predominantly through the non-caspase mediated pathway by primarily targeting mitochondria and causing outer mitochondrial membrane permeabilization that led to cytosolic and subsequent nuclear translocation of mitochondrial proteins apoptosis inducing factor (AIF) and endonuclease G (EndoG). These effects were present before apoptosis was evident and were related to intracellular levels of bivalent iron (Fe+2). Artesunate’s unique mechanism probably was at least partially responsible for, its ability to act synergistically with multiple anti-myeloma agents. Their findings suggest that artesunate acts through iron to affect the mitochondria and induce low ROS and non-caspase-mediated apoptosis. Its potency, toxicity profile, and synergism with other drugs make it an intriguing new candidate for MM treatment.

Source

Papanikolaou X, Johnson S, Garg T et al. Artesunate overcomes drug resistance in multiple myeloma by inducing mitochondrial stress and non-caspase apoptosis. Oncotarget. 2014 Jun 30;5(12):4118-28.

(DCIS) Ductal Cell Carcinoma in Situ

(DCIS) Ductal Cell Carcinoma in Situ

Action: inhibits and reverses precancerous lesions of breast cancer
inhibits ki67 and mRNA expression

Yanghe Huayan Decoction (YHD) Cornu Cervi Degelatinatium (lu jiao shuang) 12 g, prepared rhizome of rehmannia (shu di) 9 g, Cortex Cinnamomi (rou gui) 6 g, Semen Sinapis (bai jie zi) 3 g, Radix Curcumae (yu jin) 12 g, Psuedobulbus Cremastrae (shan ci gu) 15 g, Bulbus Frittillariae Thunbergii (zhe bei mu) 9 g, licorice root (gan cao) 6 g at the daily dose of 7.2 g/kg.

YHD could partially inhibit and reverse canceration of DCIS. It also could inhibit ki67 protein and mRNA expression. Its effect was similar to tamoxifen.

It is suitable for prevention and treatment of precancerous lesions of breast cancer (Li et al., 2014).

References

 

Li JW, Liu XF, Chen HZ, Chen HH, Shi GX, Wang SJ. [Effect of yanghe huayan decoction on precancerosis of breast cancer, protein and mRNA expression of ki67: an experimental research]. Zhongguo Zhong Xi Yi Jie He Za Zhi. 2014 Aug;34(8):970-5. [Article in Chinese]

Panax Ginseng and Salvia miltiorrhiza

Action: Chemo-sensitizing

An increasing number of cancer patients are using herbs in combination with conventional chemotherapeutic treatment. It is therefore important to study the potential consequences of the interactions between herbs and anticancer drugs. The effects of extracts from Panax ginseng (PGS) and Salvia miltiorrhiza Bunge (SMB) on the pharmacokinetics of 5-fluorouracil (5-FU) were performed in vivo and detected by high performance liquid chromatography (HPLC), while, an ATP assay was used to study the pharmacodynamic interactions in vitro. The results of the pharmacokinetic experiments showed a significant increase in the elimination half-life (t1/2(k e )) of 5-FU in the PGS-pretreated group and in the area under the curve (AUC) in the SMB-pretreated group compared with the control group.

However, after SMB pretreatment, weight loss was observed in rats. The results of pharmacodynamic experiments showed that neither PGS nor SMB, when used alone, directly inhibited cancer cell growth at 0.1-100 μg/ml. Moreover, PGS had a synergistic cytotoxic effect with 5-FU on human gastric cancer cells but not on normal gastric cells. The results imply that when combined with 5-FU, PGS may be a better candidate for further study. This study might provide insights for the selection of herbal-chemotherapy agent interactions (Gu et al., 2013).

Reference

Gu C, Qiao J, Zhu M, et al. (2013) Preliminary evaluation of the interactions of Panax ginseng and Salvia miltiorrhiza Bunge with 5-fluorouracil on pharmacokinetics in rats and pharmacodynamics in human cells. Am J Chin Med. 2013;41(2):443-58. doi: 10.1142/S0192415X13500328.

Scutellaria barbatae herba

Cancer: Breast, ovarian

Action: Apoptotic, bcl2

Herba Scutellaria barbatae, was cytotoxic to 100% (11 of 11) of actively proliferating ovarian lines tested and 50% (2 of 4) of actively proliferating breast cell lines tested. Confluent cultures were resistant to killing by herb, whereas subconfluent cultures were sensitive. Resistant proliferating cell lines expressed higher levels of bcl2. Transfection of the most sensitive ovarian cancer cell line (A2780) with bcl2 resulted in a noticeable protection against apoptosis, but there was no protection in other transfected lines.

These results will be useful in guiding future studies of herbal aqueous extracts, as well as providing information for clinicians when patients are concurrently using these herbs along with conventional cancer therapies.

Source:

Powell CB, Fung P, Jackson J, Dall’Era J, Lewkowicz D, Cohen I, Smith-McCune K. Aqueous extract of herba Scutellaria barbatae, a chinese herb used for ovarian cancer, induces apoptosis of ovarian cancer cell lines. Gynecol Oncol. 2003 Nov;91(2):332-40.

Cancer: Tongue

Berberine

Cancer: Tongue

Action: Down-regulates u-PA, MMP-2 and -9 expressions

There is increasing evidence that urokinase-type plasminogen activator (u-PA) and matrix metalloproteinases (MMPs) play an important role in cancer metastasis and angiogenesis. Inhibition of u-PA and MMPs could suppress migration and invasion of cancer cells. Berberine, one of the main constituents of the plant Rhizoma coptidis, is a type of isoquinoline alkaloid, reported to have anti-cancer effects in different human cancer cell lines.

Here, we report that berberine inhibited migration and invasion of human SCC-4 tongue squamous carcinoma cells. This action was mediated by the p-JNK, p-ERK, p-p38, IkappaK and NF-kappaB signaling pathways resulting in inhibition of MMP-2 and -9 in human SCC-4 tongue squamous carcinoma cells. Analysis also showed that berberine inhibited the levels of urokinase-plasminogen activator (u-PA). These results suggest that berberine down-regulates u-PA, MMP-2 and -9 expressions in SCC-4 cells through the FAK, IKK and NF-kappaB mediated pathways and a novel function of berberine is to inhibit the invasive capacity of malignant cells.

Reference

Ho YT, Yang JS, Li TC, Lin JJ, et al. Berberine suppresses in vitro migration and invasion of human SCC-4 tongue squamous cancer cells through the inhibitions of FAK, IKK, NF-kappaB, u-PA and MMP-2 and -9. Cancer Lett. 2009 Jul 8;279(2):155-62. doi: 10.1016/j.canlet.2009.01.033.

Gypenosides

Cancer: Oral

Action: Downregulates NF-κB, COX-2, extracellular signal-regulated kinase 1/2 (ERK1/ 2), matrix metalloproteinase-9, -2 (MMP-9, -2), sevenless homolog (SOS), Ras, urokinase-type plasminogen activator (uPA), focal adhesion kinase (FAK) and RAC-alpha serine/threonine-protein kinase (Akt)

Gypenosides (Gyp), found in Gynostemma pentaphyllum Makino, has been used as a folk medicine in the Chinese population for centuries and is known to have diverse pharmacologic effects, including anti-proliferative and anti-cancer actions. However, the effects of Gyp on prevention from invasion and migration of oral cancer cells are still unsatisfactory.

The purpose of this study was to investigate effects of Gyp treatment on migration and invasion of SAS human oral cancer cells. SAS cells were cultured in the presence of 90 and 180 μg/mL Gyp for 24 and 48 hours. Gyp induced cytotoxic effects and inhibited SAS cells migration and invasion in dose- and time-dependent response.

Gyp decreased the abundance of several proteins, including nuclear factor-kappa B (NF-κB), cyclooxygenase-2 (COX-2), extracellular signal-regulated kinase 1/2 (ERK1/ 2), matrix metalloproteinase-9, -2 (MMP-9, -2), sevenless homolog (SOS), Ras, urokinase-type plasminogen activator (uPA), focal adhesion kinase (FAK) and RAC-alpha serine/threonine-protein kinase (Akt), in a time-dependent manner. The inhibition of NF-κB and MMP-2, -7 and -9 signaling may be one of the mechanisms that is present in Gyp-inhibited cancer cell invasion and migration.

Reference

Lu KW, Chen JC, Lai TY, Yang JS, et al. Gypenosides inhibits migration and invasion of human oral cancer SAS cells through the inhibition of matrix metalloproteinase-2 -9 and urokinase-plasminogen by ERK1/2 and NF-kappa B signaling pathways. Hum Exp Toxicol. 2011 May;30(5):406-15. doi: 10.1177/0960327110372405

Emodin

Cancer: Tongue

Action: Downregulates MMP-2, u-PA

Emodin, aloe-emodin and rhein are major compounds in rhubarb (Rheum palmatum L.), used in Chinese herbal medicine, and found to have antitumor properties including cell cycle arrest and apoptosis in many human cancer cells. Our previous studies also showed that emodin, aloe-emodin and rhein induced apoptosis in human tongue cancer SCC-4 cells. In the present study, we investigated whether or not emodin, aloe-emodin and rhein inhibited migration and invasion of SCC-4 cells.

Herein, we demonstrate that emodin, aloe-emodin and rhein inhibit the protein levels and activities of matrix metalloproteinase-2 (MMP-2) but did not affect gene expression of MMP-2, however, they inhibited the gene expression of MMP-9 and all also inhibited the migration and invasion of human tongue cancer SCC-4 cells.

MMP-9 (gelatinase-B) plays an important role and is the most associated with tumor migration, invasion and metastasis in various human cancers. Results from zymography and Western blotting showed that emodin, aloe-emodin and rhein treatment decrease the levels of MMP-2, urokinase plasminogen activator (u-PA) in a concentration-dependent manner. The order of inhibition of associated protein levels and gene expression of migration and invasion in SCC-4 cells are emodin >aloe-emodin >rhein.

In conclusion, these findings suggest that molecular targeting of MMP-9 mRNA expression by emodin, aloe-emodin and rhein might be a useful strategy for chemo-prevention and/or chemo-therapeutics of tongue cancers.

Reference:

Chen YY, Chiang SY, Lin JG, et al. Emodin, aloe-emodin and rhein inhibit migration and invasion in human tongue cancer SCC-4 cells through the inhibition of gene expression of matrix metalloproteinase-9. Int J Oncol. 2010 May;36(5):1113-20.

Phytoestrogens/Flavonoids

Cancer: Breast

Action: MDR

Breast cancer resistance protein (BCRP), also called ABCG2, confers resistance to anticancer agents such as 7-ethyl-10-hydroxycamptothecin (SN-38), mitoxantrone, and topotecan. We found previously that sulfated estrogens are physiologic substrates of BCRP. Flavonoids with weak estrogenic activities are called phytoestrogens. In this study, we show that phytoestrogens/flavonoids, such as genistein, naringenin, acacetin, and kaempferol, potentiated the cytotoxicity of SN-38 and mitoxantrone in BCRP-transduced K562 (K562/BCRP) cells. Some glycosylated flavonoids, such as naringenin-7-glucoside, also effectively inhibited BCRP. These flavonoids showed marginal effect on the drug sensitivity of K562 cells. Genistein and naringenin reversed neither P-glycoprotein-mediated vincristine resistance nor multidrug resistance-related protein 1-mediated VP-16 resistance.

Genistein and naringenin increased cellular accumulation of topotecan in K562/BCRP cells. K562/BCRP cells also accumulated less [3H]genistein than K562 cells. [3H]genistein transport in the basal-to-apical direction was greater in BCRP-transduced LLC-PK1 (LLC/BCRP) cells, which express exogenous BCRP in the apical membrane, than in parental cells. Fumitremorgin C abolished the increased transport of [3H]genistein in LLC/BCRP cells compared with parental cells. TLC analysis revealed that genistein was transported in its native form but not in its metabolized form.

These results suggest that genistein is among the natural substrates of BCRP and competitively inhibits BCRP-mediated drug efflux. The results have two important clinical implications: (a) flavonoids and glycosylated flavonoids may be useful in overcoming BCRP-mediated drug resistance in tumor cells; and (b) coadministration of flavonoids with BCRP-substrate antitumor agents may alter the pharmacokinetics and consequently increase the toxicity of specific antitumor agents in cancer patients.

Reference

Imai Y, Tsukahara S, Asada S, Sugimoto Y. Phytoestrogens/Flavonoids Reverse Breast Cancer Resistance Protein/ABCG2-Mediated Multidrug Resistance Cancer Res June 15, 2004 64; 4346 doi: 10.1158/0008-5472.CAN-04-0078

Carnosic Acid

Cancer: Colorectal

Action: suppresses carcinogenesis

A study by Kim et al., (2014) determined the efficacy of carnosic acid (CA) for suppressing colon carcinogenesis associated with excess adiposity. The proliferation of HT-29 cells co-cultured with 3T3L1 cells was significantly higher than proliferation of control cells (p<0.05). CA treatment suppressed the growth of co-cultured HT-29 cells through cell cycle arrest and enhanced apoptosis by inhibiting leptin receptor (Ob-R) signaling including Akt and ERK phosphorylation. CA supplementation in vivo decreased the number of colon tumors and reduced circulating concentrations of leptin, adiponectin, insulin, and IGF-1. Colonic expression of Ob-R, insulin receptor (IR), p-Akt, p-ERK, Bcl-x,L and cyclinD1 protein was also suppressed in animals fed CA.

CA appears to alleviate adipocity-related acceleration of colon tumor formation.

Carnosic acid is a natural benzenediol abietane diterpene found in rosemary (Rosmarinus officinalis) and common sage (Salvia officinalis). Dried leaves of rosemary or sage contain 1.5 to 2.5% carnosic acid.

Source

Kim Y-J, Kim J-S, Seo Y-R, et al. Carnosic acid suppresses colon tumor formation in association with anti-adipogenic activity. Molecular Nutrition & Food Research. 2014 DOI: 10.1002/mnfr.201400293