The present invention relates to a pharmaceutical composition composed of flavonoid compound. In particular, the present invention relates to a pharmaceutical composition composed of a novel flavonoid, protoapigenone, for treating cancer such as gynecological cancers and prostate cancer, etc. and relates to the research on the signaling pathways of cancer cells induced by the pharmaceutical composition.
Flavonoid or bioflavonoid, a polyphenolic compound, can inhibit human cancer cell growth. Flavonoid has a structure of phenylbenzopyrone (C6-C3-C6), mainly including flavone, flavanol, isoflavone, flavonol, flavanone and flavanonol (Ren et al., 2003). The above-mentioned flavonoids can be found in some dietary plants, some medical plants and herbal remedies (Kajimoto et al., 2002; Samuelsen, 2000). Some flavonoids such as apigenin, genistein and catechin are proved to own the growth inhibition on ovarian cancer, breast cancer, colon cancer and leukemia (Birt et al., 2001; Brusselmans et al., 2005; Fang et al., 2007; Friedman et al., 2007; Gossner et al., 2007; Hamblin, 2006; Seo et al., 2006; Spinella et al., 2006). The bioactivity of flavonoid includes apoptosis induction, cell cycle arrest, growth inhibition, and angiogenesis inhibition, antioxidation and the combination thereof (Ren et al., 2003). These bioactivities is accomplished by regulating the signaling transduction pathways, such as nuclear factor-κB (NFκB), activator protein-1 or mitogen-activated protein kinases (MAPKs) (Fresco et al., 2006; Kong et al., 2001; Sarkar and Li, 2004). This renders that falvonoid might be the effective anticancer agent.
Protoapigenone, a novel flavonoid, has a structure as formula I and is extracted from the whole plant Thelypteris torresiana (Gaud), a native fern in Taiwan. The preparation method of protoapigenone was disclosed in Taiwan Patent Application No. 094139201. In addition, Lin et al. (2005) has proved that protoapigenone has cytotoxicity on human liver cancer cell lines Hep G2 and Hep 3B, human breast adenocarcinoma cell line MCF-7, human lung adenocarcinoma epithelial cell line A549 and human breast cancer cell line MDA-MB-231. However, the literature does not disclose whether protoapigenone can inhibit growth of other cancer cell lines and the signaling pathways thereof. Simultaneously, one skilled in the art also cannot infer that protoapigenone also can inhibit the growth of other cancer cell lines and has other bioactivities from the applications of protoapigenone on the abovementioned cancers.
Gynecological cancer is a cancer originated from female reproductive organs, such as cervix, fallopian tubes, ovaries, uterus, vagina and vulva. Half of eighty thousand new diagnosed cases in the United States are uterus cancer. The risk to suffer cancer increases along with ages, and genetic mutation or family inheritance also increases the risk. Paclitaxel (Taxol®) and paraplatin (Carboplatin®) have good early therapeutic effect in the later-stage ovarian cancer which has a remission rate of 60% to 80%. However, the recurrence is relatively high. Finally, up to 70% patients die.
Prostate cancer is another common carcinoma and is ranked in the third one of the female cancer-related death causes in the United States (Jemal et al., 2006). Surgery and radiotherapy are the major curative methods for low-to-moderately differentiated prostate cancer (Kish et al., 2001). Once the cancer cells spreads beyond the pelvis, there is no effective cure for the prostate cancer, and the treatment relies heavily on the chemotherapy to control the cancer growth. However, the clinically used chemotherapeutic agents remain highly toxic to normal tissues (Moss and Petrylak, 2006).
Therefore, to resolve the drug problems of gynecological cancers and prostate cancer, and the difficulty of newly synthetic drugs by chemosynthesis being overcome by the natural drugs extracted from the plant, flavonoid becomes a new choice for the new drug development.
It is therefore attempted by the applicant to deal with the above situation encountered in the prior art.
In order to the gynecological cancer and the prostate cancer treatments, the pharmaceutical composition composed of a flavonoid compound is extracted from the fern, and is performed in vitro and in vivo experiments on the gynecological cancer cells, the prostate cancer cells and the urinary bladder cancer cells to prove the cytotoxicity of flavonoid on the cancer cells and the signaling transduction pathway thereon. The clinical application value of the flavonoid compound is promoted.
In accordance with the first aspect of the present invention, a pharmaceutical composition for treating a cancer is provided. The pharmaceutical composition includes a flavonoid represented by formula I:
The cancer is one selected from a group consisting of a gynecological cancer, a prostate cancer, a bladder cancer and a liver cancer.
Preferably, the gynecological cancer is one selected from a group consisting of an ovarian cancer, a breast cancer and a cervical cancer. The ovarian cancer is demonstrated by one of an MDAH-2774 cell line and an cell line, the breast cancer is demonstrated by one of an MDA-MB-468 cell line, an MDA-C33A cell line and a T47D cell line, and the cervical cancer is demonstrated by a HeLa cell line.
Preferably, the prostate cancer is demonstrated by an LNCap cell line, the bladder cancer is demonstrated by one of an RT4 cell line and a T24 cell line, and the liver cancer is demonstrated by a Hep 3B cell line.
Preferably, the flavonoid is an extract of a fern which is Thelypteris torresiana (Gaud).
Preferably, the flavonoid arrests a cell at one of an S phase and a G2/M phase by regulating an expression of at least one cell cycle protein of the cell.
Preferably, the at least one cell cycle protein is one selected from a group consisting of a phosphated cyclin B1 (p-cyclin B1) protein, a cyclin B1 protein, a phosphated cyclin-dependent kinase 2 (p-Cdk2) protein, a Cdk2 protein and a cell division cycle 25 homolog C (Cdc25C) protein.
Preferably, the at lease one cell cycle protein is one selected from a group consisting of a caspase-3 protein, a poly(ADP)ribose polymerase (PARP) protein, a B-cell leukemia/lymphoma x long (Bcl-xL) protein and a B-cell leukemia/lymphoma 2 (Bcl-2) protein so as to induces the cell to go into an apoptosis.
Preferably, the flavonoid regulates one of a p38 mitogen-activated protein kinase (MAPK) protein, a c-Jun NH2-terminal kinase (JNK) ½ protein and a combination thereof so as to induce the cell to go into an apoptosis.
Preferably, the pharmaceutical composition further includes cis-diamminedichloridoplatinum having a structure of Formula II:
and synergistically functioning with the flavonoid to treat the cancer.
Preferably, the pharmaceutical composition further includes a pharmaceutically acceptable carrier.
Preferably, the flavonoid inhibits a cancer cell of a mammal which includes a human being and a rodent. The rodent includes a mouse and a rat.
In accordance with the second aspect of the present invention, a pharmaceutical composition for treating a cancer is provided. The pharmaceutical composition includes a flavonoid having a first structure of Formula I:
and a cis-diamminedichloridoplatinum having a second structure of Formula II:
Preferably, the pharmaceutical composition inhibits a growth of a cancer cell.
The above objectives and advantages of the present invention will become more readily apparent to those ordinarily skilled in the art after reviewing the following detailed descriptions and accompanying drawings, in which:
The present invention will now be described more specifically with reference to the following Embodiments. It is to be noted that the following descriptions of preferred Embodiments of this invention are presented herein for purpose of illustration and description only; it is not intended to be exhaustive or to be limited to the precise form disclosed.
I. Experimental Materials:
Protoapigenone was isolated from the whole plant, Thelypteris torresiana (Gaud) (Lin et al., 2005), and was dissolved in dimethyl sulfoxide (DMSO) and added to the cultured cells at a 1 to 1000.
The gynecological cancer cell lines used in the present invention were listed as follows. (1) Human ovarian cancer cell lines MDAH-2774 and SKOV3, human breast cancer cell lines MDA-MB-468, MDA-MB-231 and T47D, human cervical cancer cell lines HeLa and C33A and the immortalized non-cancer human breast epithelial cell line MCF-10A were purchased from American Type Culture Collection (ATCC), Manassas, Va. These cell lines were incubated in DMEM-F12 medium supplemented with 10% of fetal bovine serum and penicillin/streptomycin/amphotericin B. (2) Immortalized human ovarian surface epithelial (HOSE) cell lines, HOSE 6-3 and HOSE 11-12, were provided by Professor George S. W. Tsao (University of Hong Kong, Poffulam, China). These cell lines were incubated in 1:1 mixture of MCDB 105 and M199 medium (Sigma, St. Louis, Mo.) supplemented with 10% of fetal calf serum and penicillin/streptomycin/amphotericin B.
Human prostate cancer cell line LNCap used in the present invention was purchased from ATCC, Manassas, Va. and was cultured in RPMI 1640 medium supplemented with 10% FBS and penicillin/streptomycin/amphotericin B. LNCap cells were treated with 2.5, 5 and 10 μM protoapigenone respectively and were analyzed. In some experiments, LNCap cells were treated with p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 or c-Jun NH2-terminal kinase (JNK) ½ inhibitor SP600125 for one hour before protoapigenone treatment.
II. Experimental Methods:
1. XTT Cell Proliferation and Clonogenic Assay:
Cells were seeded at 7×103 per well in 96-well culture plates before treatment with different concentrations of protoapigenone for 12, 24 and/or 48 hours. Next, the cytotoxicity of protoapigenone was determined using XTT cell proliferation assay (Sigma, St. Louis, Mo.). The absorbance of wavelength at 490 nm (OD490) and 650 nm (OD650) were determined respectively by enzyme-linked immunosorbent assay (ELISA) reader, and 50% of inhibitory concentration (IC50) was calculated by the value of OD490-OD650. In addition, in order to evaluate the long-term effect, the cells were treated with different concentration of protoapigenone for 3 hours, then were incubated with fresh medium for 14 days so as to form colonies. Finally, the cells were stained with crystal violet and were observed under the microscope.
2. Cell Survival Assay:
SKOV3 cells at various cell cycle phases were incubated in the 6-cm Petri dishes and treated with protoapigenone at different dosages (0, 5 and 10 μM/ml). Subsequently, the cells were harvested and mixed with trypan blue at a ratio of 1:1. The stained apoptotic cells were observed under the microscope.
3. Cell Cycle and Sub-G1 Analysis:
The ovarian cancer cells and the prostate cancer cells respectively were treated with protoapigenone at different concentrations (0, 2.5, 5 and 10 μM) for 24 hours and 6 hours (or 12 hours). These cells were harvested by trypsin and fixed with 70% ethanol for 1 hour, washed with phosphate buffered saline (PBS) twice, and resuspended in propidium iodide (PI)/RNase A solution for 30 minutes. In accordance with the mechanism of DNA stained with propidium iodide, the cell cycle and sub-G1 distribution were detected by the FACScan flow cytometry (Becton Dickinson, San Jose, Calif.), and data analysis was done with CellQuest software (BD Bioscience).
4. Annexin V Apoptosis Assay:
The theory of this assay is to detect early apoptotic cells during the apoptotic progression. The ovarian cancer cells and the prostate cancer cells respectively were incubated with protoapigenone at 10 μM for 3 hours on chamber slides, washed with cold PBS twice and stained with a binding buffer containing annexin V-fluorescein isothiocyanate (FITC) (1 mg/ml; Strong Biotech, Taipei, Taiwan) and 4,6-diamidino-2-phenylinodole (Sigma-Aldrich) at 25° C. for 15 minutes. The stained cells were washed with PBS twice and observed under the fluorescent microscope.
5. Terminal Deoxynucleotidyl Transferase dUTP Nick-End Labeling (TUNEL) Assay:
TUNEL assay is performed to detect whether cellular DNA is fragmented. The prostate cancer cells were treated with protoapigenone at various concentrations (2.5, 5 and 10 μM) for 12 or 24 hours, then were stained with TUNEL using the DeadEnd Colorimetric TUNEL system (Promega, Madison, Wis.). In brief, the prostate cancer cells were fixed with 4% paraformaldehyde for 30 minutes after protoapigenone treatment. Next, the fixed cells were incubated with digoxigenin-conjugated dUTP and nucleotide mixture in a recombinant terminal deoxynucleotide transferase-catalyzed reaction in the humidified atmosphere at 37° C. Subsequently, they were immersed in the stop buffer for 15 minutes at 37° C. The Cells were washed with PBS, then were incubated in DAB solutions for 15 minutes in dark. The stained cells were observed under the microscope. The percentage of TUNEL-positive cells were counted in 1000 cells, and the apoptosis index in the later period was calculated.
6. Immunoblotting Analysis:
The cells treated with protoapigenone at various concentrations (0, 2.5, 5 and 10 μM) for 24 hours were washed twice and lysed with EBC buffer (50 mM Tris (pH 7.6), 120 mM NaCl, 0.5% Nonidet P-40, 1 mM β-mercaptoethanol, 50 mM NaF, and 1 mM Na3VO4). The concentration of protein supernatant was further determined using the Bio-Rad protein assay kit (Bio-Rad Laboratories, Hercules, Calif.). Whole cell lysates were separated on polyacrylamide sodium dodecyl sulfate (SDS) gels and the proteins were transferred onto the nitrocellulose membrane. Proteins were detected sequentially by the specific primary antibodies, followed by peroxidase-conjugated secondary antibodies, and visualized by the Enhanced chemiluminescence detection system (Amersham, N.J.).
7. Nude Mice Model:
MDAH-2774 human ovarian cancer cells of 2×106 (or LNCap prostate cancer cells of 1×106) in 0.1 ml PBS were injected subcutaneously at the right flank of 6-week-old female nude mice (Foxnlun/Foxulun). When tumors became visible (approximately 3×3 mm in size), the mice were randomly grouped and treated intraperitoneally with protoapigenone or vehicle every other day. In the xenograft experiment of MDAH-2774 human ovarian cancer cells, the control group, the low dose group and the high dose group were given PBS, 0.069 μM/g body weight of protoapigenone in PBS (a dose equals to one-tenth of the IC50 for MDAH-2774 cells), and 0.69 μM/g body weight of protoapigenone in PBS (a dose equals to the IC50 for MDAH-2774 cells) individually. In the xenograft experiment of LNCap prostate cancer cells, the control group, the low dose group and the high dose group were given PBS, 0.37 μM/g body weight of protoapigenone in PBS (a dose equals to one-tenth of the IC50 for LNCap cells), and 3.7 μM/g body weight of protoapigenone in PBS (a dose equals to the IC50 for LNCap cells) individually. Tumor size and body weight were measured twice a week using calipers, and their tumor volumes were calculated according to a standard formula: width2×length/2. After treatment with protoapigenone for 5 weeks, the mice xenografted with LNCap cells were sacrificed by deep anesthesia, and the blood sampling by cardiac aspiration was done immediately after anesthesia. Complete blood count of the nude mice blood was determined by Sysmex XE-2100 (TOA Medical Electronics, Kobe, Japan), and the plasma blood urea nitrogen (BUN), creatine (Cr), asparatate aminotransferase (AST), and alanine aminotransferase (ALT) levels were determined by Beckman LX20 (Beckman-Coulter, U.S.A.)
8. Small Interfering RNA (siRNA) Knockdown:
The theory of this assay is to attenuate the p38 MAPK and JNK½ expressions of LNCap cells using specific siRNA (Santa Cruz Biotechnology, Inc.) and transfecting with Lipofectamine 2000 (Invitrogen). LNCap cells were treated with protoapigenone after incubating for 48 hours, and the treated LNCap cells were harvested to analyze.
9. Immunohistochemical Analysis:
After the tissue specimen were fixed with 10% formaldehyde or 4% tripolyaldehyde (Yeh et al., 2006), the tissue samples were dissected, dehydrated and coated with wax. The thickness of specimen was 3 μm or 4 μm, and the specimen were immunostained with hematoxylin-eosin (H & E) or primary antibody, then were labeled with Universal LAB+kit/horseradish peroxidase (HRP) (Dako Denmark A/S, Glostrup, Denmark), or were counterstained with hematoxylin. The cells were observed under the microscope.
III. Experimental Results:
1. Inhibition of Protoapigenone on Ovarian Cancer Cells:
Please refer to Table 1, which is the cytotoxicity of protoapigenone on various cancer cell lines. In Table 1, protoapigenone has the highest cytotoxicity on the ovarian cancer cell lines MDAH-2774 and SKOV3; however, protoapigenone has the lower cytotoxicity on the immortalized human ovarian epithelial cells HOSE 6-3 and HOSE 11-12. The identical results can be corresponded to the breast cancer cells MDA-MB-468 and T47D and the immortalized human breast epithelial cells MCF-10A.
aMeans ± SD of three independent experiments
In addition, please refer to
Please refer to
Please refer to
A quantitative evaluation of apoptosis is sought using an annexin V-FITC dye to detect the translocation of phosphatidylserine from cytoplasmic leaflet of the plasma membrane to the cell surface. Please refer to
Please refer to
In the past clinical research, cisplatin (also named as cisplatinum or cis-diamminedichloridoplatinum (II), CDDP, as Formula II) is a common, platinum-based anti-cancer or chemotherapeutic agent for treating cancers, such as sarcoma, small cell lung cancer, ovarian cancer, lymphoma and germ cell tumor, etc. Cisplatin forms platinum complex bounded with or interconnected with DNA in the cells, and finally triggers apoptosis program or automatic apoptosis. Therefore, a pharmaceutical composition including protoapigenone and cisplatin is evaluated whether the pharmaceutical composition has synergistic cytotoxicity on cells. Please refer to
Please refer to
Please refer to
The MDAH-2774 tumor xenografted from the control and the protoapigenone-treatment mice are harvested, and apoptosis is accessed by immunoblotting and immunohistochemical analysis. Please refer to
2. Inhibition of Prostate Cancer Cells by Protoapigenone:
Please refer to
Please refer to
Please refer to
The anticancer activity of several flavonoids, including (−)-epigallocatechin gallate (a catechin, abbreviated as EGCG) and apigenin, are associated with the modulation of MAPK pathway (Kim et al., 2005; Van Dross et al., 2003). Thus, whether the MAPK pathway-related proteins of LNCap cells after the protoapigenone treatment are involved is further determined. Please refer to
To evaluate the role of p38 MAPK and JNK½ activation in the protoapigenone-induced cell death, LNCap cells are pretreated for 1 hour with the pharmacological inhibitors of p38 MAPK (SB203580) and JNK½ (SP600125) respectively (
Whether the activation of p38 MAPK and JNK½ is involved in the protoapigenone-induced apoptosis and cell cycle arrest is further investigated. Please refer to
To further investigate the role of p38 MAPK and JNK½ in the protoapigenone-induced cell death, the siRNA specific for p38 MAPK and JNK½ are applied to knockdown the expression of p38 MAPk and JNK½, respectively (
To determine whether protoapigenone suppresses the prostate cancer cell growth in vivo, LNCap cells are injected into the right flank of the rude mice, and the inhibition of tumor growth by protoapigenone is analyzed. Please refer to
From the above-mentioned results, it is known that the activation of p38 MAPK and JNK½ signaling pathways, the increased levels of the cleaved PARP and caspase-3 and apoptosis induction in LNCap prostate cancer cells can be proceeded by protoapigenone treatment. The activities of p38 MAPK and JNK½ respectively can be inhibited by its inhibitor SB203580 and SP600125 and by the specific siRNA so as to decrease apoptosis, and the activities of the cleaved caspase-3 is proved by the present invention. Therefore, the important role of p38 MAPK and JNK½ on the protoapigenone-induced apoptosis is proved in the present invention.
In the present invention, the activated p38 MAPK and JNK½ induce the cell cycle to be arrested in G1/S or G2/M phase, which is achieved by increasing the inactivated p-Cdc25C (Ser216) and by decreasing the levels of p-cyclin B1 (Ser147) and Cdk2.
3. Inhibition of Other Cancer Cells by Protoapigenone:
In addition to ovarian cancer and prostate cancer, bladder cancer, liver cancer and cervical cancer are also the research targets of the present invention to investigate whether the growth of these cancer cells can be inhibited by protoapigenone. First of all, the IC50 values of protoapigenone on human urinary bladder transitional cell papilloma RT4 (ATCC number: HTB-2), T24 (ATCC number: HTB-4) and human hepatocellular carcinoma Hep 3B (ATCC number: HB-8064) respectively are 5.105±3.25 μM, 5.52±0.598 μM and 1.86=0.656 μM. Please refer to
Please refer to
In conclusion, apoptosis of the ovarian cancer cells, the prostate cancer cells, the urinary bladder cancer cells and the cervical cancer cells can be induced effectively by the protoapigenone treatment, and the signaling pathways thereof are proved. No significant hepatotoxicity, nephrotoxicity and hematological toxicity of protoapigenone on the nude mice renders that protoapigenone can be the chemotherapeutic agent on human and other mammals. Further, protoapigenone of the present invention and cisplatin can be combined as the pharmaceutical composition to achieve the effective effect of cancer therapy. Protoapigenone of the present invention can be manufactured as the dosage form with the pharmaceutically acceptable carrier.
While the invention has been described in terms of what is presently considered to be the most practical and preferred Embodiments, it is to be understood that the invention needs not be limited to the disclosed Embodiments. On the contrary, it is intended to cover various modifications and similar arrangements included within the spirit and scope of the appended claims, which are to be accorded with the broadest interpretation so as to encompass all such modifications and similar structures.
aGot means glutamic-oxaloacetic transaminase
bGpt means glutamic-pyruvic transaminase
cHCT means hematocrit
dMCV means mean corpuscular volume
aGot means glutamic-oxaloacetic transaminase
bGpt means glutamic-pyruvic transaminase
cMCV means mean corpuscular volume
Number | Date | Country | Kind |
---|---|---|---|
097134287 | Sep 2008 | TW | national |