Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Phytomedicine ; 116: 154860, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37201366

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is upregulated in prostate cancer (PCa). However, suppression of EGFR did not improve the patient outcome, possibly due to the activation of PI3K/Akt signaling in PCa. Compounds able to suppress both PI3K/Akt and EGFR signaling may be effective for treating advanced PCa. PURPOSE: We examined if caffeic acid phenethyl ester (CAPE) simultaneously suppresses the EGFR and Akt signaling, migration and tumor growth in PCa cells. METHODS: Wound healing assay, transwell migration assay and xenograft mice model were used to determine the effects of CAPE on migration and proliferation of PCa cells. Western blot, immunoprecipitation, and immunohistochemistry staining were performed to determine the effects of CAPE on EGFR and Akt signaling. RESULTS: CAPE treatment decreased the gene expression of HRAS, RAF1, AKT2, GSK3A, and EGF and the protein expression of phospho-EGFR (Y845, Y1069, Y1148, Y1173), phospho-FAK, Akt, and ERK1/2 in PCa cells. CAPE treatment inhibited the EGF-induced migration of PCa cells. Combined treatment of CAPE with EGFR inhibitor gefitinib showed additive inhibition on migration and proliferation of PCa cells. Injection of CAPE (15 mg/kg/3 days) for 14 days suppressed the tumor growth of prostate xenografts in nude mice as well as suppressed the levels of Ki67, phospho-EGFR Y845, MMP-9, phospho-Akt S473, phospho-Akt T308, Ras, and Raf-1 in prostate xenografts. CONCLUSIONS: Our study suggested that CAPE can simultaneously suppress the EGFR and Akt signaling in PCa cells and is a potential therapeutic agent for advanced PCa.


Subject(s)
Phenylethyl Alcohol , Prostatic Neoplasms , Male , Humans , Animals , Mice , Proto-Oncogene Proteins c-akt/metabolism , Prostate/pathology , Phosphatidylinositol 3-Kinases/metabolism , Mice, Nude , Epidermal Growth Factor , Prostatic Neoplasms/pathology , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , ErbB Receptors , Phenylethyl Alcohol/pharmacology , Cell Line, Tumor , Cell Proliferation
2.
PLoS One ; 17(7): e0270803, 2022.
Article in English | MEDLINE | ID: mdl-35776912

ABSTRACT

Enzalutamide, a nonsteroidal antiandrogen, significantly prolonged the survival of patients with metastatic castration-resistant prostate cancer (CRPC). However, patients receiving enzalutamide frequently develop drug resistance. Rooibos (Aspalathus linearis) is a shrub-like leguminous fynbos plant endemic to the Cedarberg Mountains area in South Africa. We evaluated the possibility of using a pharmaceutical-grade green rooibos extract (GRT, containing 12.78% aspalathin) to suppress the proliferation and survival of enzalutamide-resistant prostate cancer (PCa) cells. Treatment with GRT dose-dependently suppressed the proliferation, survival, and colony formation of enzalutamide-resistant C4-2 MDV3100r cells and PC-3 cells. Non-cancerous human cells were more resistant to GRT treatment. GRT suppressed the expression of proteins involved in phosphoinositide 3-kinase (PI3K)-Akt signaling, androgen receptor (AR), phospho-AR (Ser81), cyclin-dependent kinase 1 (Cdk1), c-Myc and Bcl-2 but increased the expression of apoptotic proteins. Overexpression of c-Myc antagonized the suppressive effects of GRT, while knockdown of c-Myc increased the sensitivity of PCa cells to GRT treatment. Expression level of c-Myc correlated to resistance of PCa cells to GRT treatment. Additionally, immunofluorescence microscopy demonstrated that GRT reduced the abundance of AR proteins both in nucleus and cytoplasm. Treatment with cycloheximide revealed that GRT reduced the stability of AR. GRT suppressed protein expression of AR and AR's downstream target prostate specific antigen (PSA) in C4-2 MDV3100r cells. Interestingly, we observed that AR proteins accumulate in nucleus and PSA expression is activated in the AR-positive enzalutamide-resistant PCa cells even in the absence of androgen. Our results suggested that GRT treatment suppressed the cell proliferation and survival of enzalutamide-resistant PCa cells via inhibition of c-Myc, induction of apoptosis, as well as the suppression of expression, signaling and stability of AR. GRT is a potential adjuvant therapeutic agent for enzalutamide-resistant PCa.


Subject(s)
Aspalathus , Prostatic Neoplasms, Castration-Resistant , Aspalathus/metabolism , Benzamides , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Male , Nitriles , Phenylthiohydantoin , Phosphatidylinositol 3-Kinases , Prostate-Specific Antigen/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
4.
J Biomed Sci ; 29(1): 16, 2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35197069

ABSTRACT

BACKGROUND: Docetaxel has been approved by USFDA as a first-line treatment for castration-resistant prostate cancer (CRPC) patients. Patients receiving androgen deprivation therapy along with docetaxel result in superior survival, lower serum prostate specific antigen (PSA) level, and better quality of life. However, a significant proportion of these patients ultimately develop resistance to docetaxel within months. Caffeic acid phenethyl ester (CAPE), one of the main bioactive components extracted from the propolis, has been reported to be effective for repressing the tumor growth, the migration and invasion of prostate cancer (PCa) cells, as well as the downstream signaling and stability of androgen receptor (AR). We hence determined if combination treatment of docetaxel with CAPE can suppress the proliferation and the survival of docetaxel-resistant PCa cells. METHODS: We established docetaxel-resistant PC/DX25 and DU/DX50 CRPC cell lines from PC-3 and DU-145 human PCa cells, respectively. Proliferation assay, MTT assay, flow cytometry with Annexin V staining, Comet Assay, and nude mice xenograft model were applied to determine the effects of combination treatment on cell proliferation and survival of the docetaxel-resistant PCa cells. Micro-Western Array (MWA) and qRT-PCR were used to investigate the molecular mechanism lying underneath. RESULTS: Combination treatment effectively suppressed the proliferation, survival and tumor growth of docetaxel-resistant PCa cells both in vitro and in nude mice. Comet assay and flow cytometry indicated that combination treatment induced apoptosis in docetaxel-resistant PCa cells. MWA and Western blotting assay revealed that combination treatment suppressed protein expression of Bcl-2, AKT2, c-Myc, apoptosis and caspase activation inhibitor (AVEN), pyruvate kinase M2 (PKM2) but increased protein expression of Bax, caspase 3, cytochrome c, glucose-6-phosphate dehydrogenase (G6PD) and acylglycerol kinase (AGK). Overexpression of Bcl-2 in the docetaxel-resistant PCa cells enhanced cell proliferation of docetaxel-resistant PCa cells under combination treatment. Analysis with qRT-PCR suggested that combination treatment decreased cholesterol biosynthesis genes DHCR24 (24-dehydrocholesterol reductase) and LSS (lanosterol synthase) but increased genes involved in glycolysis and TCA cycle. CONCLUSIONS: Combination treatment of docetaxel with CAPE effectively suppressed the proliferation and survival of docetaxel-resistant PCa cells via inhibition of Bcl-2 and c-Myc as well as induction of metabolism interference. Combination treatment can be beneficial for patients with docetaxel-resistant PCa.


Subject(s)
Prostatic Neoplasms , Androgen Antagonists/pharmacology , Animals , Apoptosis , Caffeic Acids , Cell Line, Tumor , Cell Proliferation , Docetaxel/pharmacology , Humans , Male , Mice , Mice, Nude , Phenylethyl Alcohol/analogs & derivatives , Quality of Life
5.
Ann Palliat Med ; 10(10): 10506-10514, 2021 10.
Article in English | MEDLINE | ID: mdl-34763497

ABSTRACT

BACKGROUND: Research has shown that traditional Chinese medicine (TCM) can achieve good results in the treatment of angina pectoris. In this study, we aimed to explore the therapeutic effect of TCM in the treatment of angina pectoris of coronary heart disease (CHD) through a literature search and meta-analysis. METHODS: The PubMed, Embase, CBM (China Biology Medicine) Web of Science databases were searched for studies on the treatment of angina pectoris of CHD with TCM. Inclusion and exclusion criteria were applied, and high-quality articles published from 2010.1 to 2021.8 were selected. The RevMan 5.3.5 software was used to evaluate the therapeutic effect indicators of TCM. RESULTS: Nine studies involving 824 patients were included in the meta-analysis, and the overall risk of literature bias was low. The results of meta-analysis showed that compared with conventional Western medicine, TCM + conventional Western medicine had a better efficacy indicators of angina pectoris using the fixed-effects model [odd rate (OR) =3.20, 95% confidence interval (CI): (2.09, 4.90), Z=5.35, P<0.00001]. The frequency of angina pectoris was measured by random-effects model, and the statistical results were [standard mean difference (SMD) =-1.85, 95% CI: (-2.29, -1.41), Z=8.22, P<0.00001]. The adverse events was measured by fixed-effects model, and the statistical results were [OR =0.48, 95% CI: (0.21, 1.08), Z=1.78, P=0.08]. DISCUSSION: The application of TCM in the treatment of angina pectoris of CHD can improve the therapeutic effect, reduce the frequency of angina pectoris, shorten the attack time, reduce serum total cholesterol, and improve the quality of life after treatment, but it has no obvious reducing effect on blood lipids.


Subject(s)
Coronary Disease , Drugs, Chinese Herbal , Angina Pectoris , Drugs, Chinese Herbal/therapeutic use , Humans , Medicine, Chinese Traditional , Quality of Life
6.
Ann Palliat Med ; 10(6): 6867-6872, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34237984

ABSTRACT

BACKGROUND: Diabetic nephropathy (DN) affects about 40% of diabetes mellitus (DM) patients and is the leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD) globally, especially in advanced countries. We aimed to explore the risk factors affecting the prognosis of DN, and to establish a prognostic evaluation line map. METHODS: We analyzed 471 cases of DN from December 2011 to April 2020, and extracted the basic clinical factors, including gender, age, and history of diabetes. Analysis included that of associations between DN and hypertension, creatinine (CR), body mass index (BMI), and fundus lesions. Statistical analysis was performed using R software and the related R package. The above clinical factors were analyzed by both single- and multiple-factor Cox regression. The participants were divided into two groups, including a high risk and a low risk group. A Kaplan-Meier curve was drawn for survival analysis of the high and low risk groups, and the log-rank method was used for statistical testing. A receiver operating characteristic (ROC) curve was drawn with the area under the curve (AUC) calculated to evaluate the predictive effectiveness of the line map. RESULTS: This study initially included 471 patients; however, 33 patients (7.0%) were lost to follow-up due to inaccessibility. A total of 93 cases (21.2%) died during the follow-up. The 3-year and 5-year renal survival rates were 74.5% and 22.6%, respectively. Single factor Cox analysis showed that the course of diabetes, fundus lesions, BMI, and grade of hypertension were risk factors for renal survival, and had adverse effects on prognosis (P<0.05). Multivariate Cox regression analysis showed that BMI and grade of hypertension were independent risk factors for survival of DKD, and had adverse effects on prognosis (P<0.05). Survival analysis showed that low risk group participants had significantly better survival rates than high risk group participants (P<0.05). The AUC was 0.742, which meant that the line map could accurately predict the survival rate of DN patients. CONCLUSIONS: The influence of risk factors on prognosis can be accurately evaluated by line diagram which can provide a basis for clinical decision making.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Kidney Failure, Chronic , Renal Insufficiency, Chronic , Humans , Kidney , Prognosis , Risk Factors
7.
Cell Death Dis ; 11(5): 376, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415173

ABSTRACT

Bones are the most common metastatic sites for prostate cancer (PCa). Receptor tyrosine kinase-like orphan receptor 2 (ROR2), a noncanonical Wnt receptor, plays crucial roles in skeletal morphogenesis, osteoblast differentiation, and bone formation. The role of ROR2 in PCa metastasis is unclear. We analyzed online datasets from Oncomine as well as using IHC staining on tissue array to determine the relationship between ROR2 expression level and disease outcome of PCa. To investigate how ROR2 regulates migration and invasion of PCa cells, we performed transwell assay and orthotopic xenograft model in nude mice. We then applied the Micro-Western Array (MWA), a high-throughput western blotting platform to analyze the downstream signaling pathways being regulated by ROR2. Compared with nonmalignant PZ-HPV-7 and RWPE-1 cells, PCa cell lines express lower level of ROR2 protein. Constitutive expression of ROR2 in PC-3, DU-145, or C4-2B PCa cells significantly suppressed the cell migration, invasion, and epithelial-mesenchymal transition (EMT) proteins. MWA, western blotting, and microRNA analysis showed that elevation of ROR2 suppressed the expression of miR-199a-5p, which in turn increased the expression of PIAS3. The upregulation of PIAS3 then decreased AKT2 and the phosphorylation of AKT, resulting in the inhibition of migration and invasion of PCa cells both in vitro and in orthotopic xenograft mice model. IHC staining of tissue array and Oncomine datasets analysis indicated that the gene and protein level of ROR2 is much lower in metastatic prostate tumors as compared with primary tumors or adjacent normal prostate tissues. Low level of ROR2 correlated to poor survival and high recurrent frequency in PCa patients. In conclusion, we discovered that ROR2 suppresses PCa metastasis via regulation of PIAS3-PI3K-AKT2 signaling axis.


Subject(s)
Neoplasm Metastasis/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Molecular Chaperones , Protein Inhibitors of Activated STAT , Proto-Oncogene Proteins c-akt/metabolism
8.
Cancers (Basel) ; 11(11)2019 Nov 13.
Article in English | MEDLINE | ID: mdl-31766290

ABSTRACT

Our three-dimensional organotypic culture revealed that human histone demethylase (KDM) 4C, a histone lysine demethylase, hindered the acini morphogenesis of RWPE-1 prostate cells, suggesting its potential oncogenic role. Knockdown (KD) of KDM4C suppressed cell proliferation, soft agar colony formation, and androgen receptor (AR) transcriptional activity in PCa cells as well as reduced tumor growth of human PCa cells in zebrafish xenotransplantation assay. Micro-Western array (MWA) analysis indicated that KD of KDM4C protein decreased the phosphorylation of AKT, c-Myc, AR, mTOR, PDK1, phospho-PDK1 S241, KDM8, and proteins involved in cell cycle regulators, while it increased the expression of PTEN. Fluorescent microscopy revealed that KDM4C co-localized with AR and c-Myc in the nuclei of PCa cells. Overexpression of either AKT or c-Myc rescued the suppressive effect of KDM4C KD on PCa cell proliferation. Echoing the above findings, the mRNA and protein expression of KDM4C was higher in human prostate tumor tissues as compared to adjacent normal prostate tissues, and higher KDM4C protein expression in prostate tumors correlated to higher protein expression level of AKT and c-Myc. In conclusion, KDM4C promotes the proliferation of PCa cells via activation of c-Myc and AKT.

9.
Phytomedicine ; 64: 153068, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31419729

ABSTRACT

BACKGROUND: Androgen ablation therapy is the primary treatment for metastatic prostate cancer (PCa). However, the majority of PCa patients receiving the androgen deprivation therapy develop recurrent castration-resistant prostate cancer (CRPC) within two years. Chemotherapies show little effect on prolonging survival of CRPC patients and new treatments are needed. Previous studies reported that the extracts from rooibos (Aspalathus linearis) exhibit chemopreventive properties in some cancer models, including skin, liver and oesophagus cancers in animals. We therefore investigate if extracts from rooibos can suppress the proliferation of CRPC cells. PURPOSE: We investigated whether an aspalathin-rich green rooibos extract (GRT™; 12.78 g aspalathin/100 g extract) demonstrates anti-cancer activity against CRPC cells. METHODS: High performance liquid chromatography (HPLC) was used to profile the major flavonoids in GRT. Hoechst-dye proliferation assay, 3,4,5-dimethylthiazol-2-yl)-2-5-diphenyltetrazolium bromide (MTT) viability assay and flow cytometry assay were used to explore the effects of GRT on the proliferation and cell cycle progression of CRPC cells. Comet assay was used to survey whether GRT induces apoptosis in CRPC cells. LNCaP 104-R1 xenograft nude mice model was used to determine the inhibitory effect of GRT on CRPC tumors in vivo. Micro-Western Array (MWA) and Western blot analysis were carried out to unravel the underlying molecular mechanism. RESULTS: GRT contained aspalathin as the most abundant flavonoid. GRT suppressed the proliferation and survival of LNCaP 104-R1, LNCaP FGC and PC-3 PCa cells. Flow cytometry analysis showed that GRT decreased the population of PCa cells in S phase but increased the cell population in G2/M phase. Comet assay confirmed that GRT induced apoptosis in LNCaP 104-R1 cells. Gavage of 400 mg/kg GRT suppressed LNCaP 104-R1 xenografts in castrated nude mice. MWA and Western blot analysis indicated that GRT treatment suppressed Akt1, phospho-Akt Ser473, Cdc2, Bcl-2, TRAF4 and Aven, but increased activated Caspase 3, cytochrome c, and p27Kip1. Overexpression of Akt rescued the suppressive effects of GRT on CRPC cells. Co-treatment of GRT with Bcl-2 inhibitor ABT-737, PI3K inhibitor LY294002 and Akt inhibitor GSK 690693 exhibited additive inhibitory effect on proliferation of CRPC cells. CONCLUSIONS: GRT suppresses the proliferation of CRPC cells via inhibition of Akt signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Aspalathus/chemistry , Chalcones/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
10.
Oncogene ; 38(1): 17-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30072740

ABSTRACT

During the evolution into castration or therapy resistance, prostate cancer cells reprogram the androgen responses to cope with the diminishing level of androgens, and undergo metabolic adaption to the nutritionally deprived and hypoxia conditions. AR (androgen receptor) and PKM2 (pyruvate kinase M2) have key roles in these processes. We report in this study, KDM8/JMJD5, a histone lysine demethylase/dioxygnase, exhibits a novel property as a dual coactivator of AR and PKM2 and as such, it is a potent inducer of castration and therapy resistance. Previously, we showed that KDM8 is involved in the regulation of cell cycle and tumor metabolism in breast cancer cells. Its role in prostate cancer has not been explored. Here, we show that KDM8's oncogenic properties in prostate cancer come from its direct interaction (1) with AR to affect androgen response and (2) with PKM2 to regulate tumor metabolism. The interaction with AR leads to the elevated expression of androgen response genes in androgen-deprived conditions. They include ANCCA/ATAD2 and EZH2, which are directly targeted by KDM8 and involved in sustaining the survival of the cells under hormone-deprived conditions. Notably, in enzalutamide-resistant cells, the expressions of both KDM8 and EZH2 are further elevated, so are neuroendocrine markers. Consequently, EZH2 inhibitors or KDM8 knockdown both resensitize the cells toward enzalutamide. In the cytosol, KDM8 associates with PKM2, the gatekeeper of pyruvate flux and translocates PKM2 into the nucleus, where the KDM8/PKM2 complex serves as a coactivator of HIF-1α to upregulate glycolytic genes. Using shRNA knockdown, we validate KDM8's functions as a regulator for both androgen-responsive and metabolic genes. KDM8 thus presents itself as an ideal therapeutic target for metabolic adaptation and castration-resistance of prostate cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Histone Demethylases/physiology , Membrane Proteins/metabolism , Neoplasm Proteins/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Thyroid Hormones/metabolism , ATPases Associated with Diverse Cellular Activities/physiology , Active Transport, Cell Nucleus , Adenocarcinoma/pathology , Animals , Benzamides , Cell Line, Tumor , DNA-Binding Proteins/physiology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Knockdown Techniques , Glycolysis/genetics , Heterografts , Histone Demethylases/biosynthesis , Histone Demethylases/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Interaction Mapping , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Thyroid Hormone-Binding Proteins
11.
Cancer Sci ; 109(11): 3564-3574, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30142696

ABSTRACT

Androgen receptor (AR), an androgen-activated transcription factor, belongs to the nuclear receptor superfamily. AR plays an important role in the development and progression of prostate cancer (PCa). However, the role of AR in PCa metastasis is not fully understood. To investigate the role of AR in PCa metastasis, we examined AR expression level in primary and metastatic PCa by analyzing gene array data of 378 primary prostate tumors and 120 metastatic prostate tumors from Oncomine, as well as carrying out immunohistochemical (IHC) staining of 56 prostate cancer samples. Expression of mRNA and protein of AR as well as its target gene prostate-specific antigen (PSA) was much higher in metastatic prostate tumors than in primary prostate tumors. Knockdown of AR with siRNA or treating with anti-androgen Casodex reduced migration and invasion ability of C4-2B PCa cells. Knockdown of AR increased protein expression of E-cadherin and AR coregulator KAT5 but reduced expression of epithelial-mesenchymal transition (EMT) marker proteins Slug, Snail, MMP-2, vimentin, and ß-catenin. Knockdown of KAT5 increased migration of C4-2B cells, whereas overexpression of KAT5 suppressed cell migration. KAT5 knockdown rescues the suppressive effect of AR knockdown on migration of C4-2B cells. Gene expression level of AR and KAT5 showed a negative correlation. PCa patients with higher AR expression or lower KAT5 expression correlated with shorter recurrence-free survival. Our study suggested that elevation of AR expression and AR signaling in prostate tumors promotes PCa metastasis by induction of EMT and reduction of KAT5.


Subject(s)
Lysine Acetyltransferase 5/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Up-Regulation , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Lysine Acetyltransferase 5/metabolism , Male , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Prostatic Neoplasms/metabolism , Survival Analysis
12.
Int J Mol Sci ; 16(5): 10748-66, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25984601

ABSTRACT

Head and neck cancers, which affect 650,000 people and cause 350,000 deaths per year, is the sixth leading cancer by cancer incidence and eighth by cancer-related death worldwide. Oral cancer is the most common type of head and neck cancer. More than 90% of oral cancers are oral and oropharyngeal squamous cell carcinoma (OSCC). The overall five-year survival rate of OSCC patients is approximately 63%, which is due to the low response rate to current therapeutic drugs. In this review we discuss the possibility of using caffeic acid phenethyl ester (CAPE) as an alternative treatment for oral cancer. CAPE is a strong antioxidant extracted from honeybee hive propolis. Recent studies indicate that CAPE treatment can effectively suppress the proliferation, survival, and metastasis of oral cancer cells. CAPE treatment inhibits Akt signaling, cell cycle regulatory proteins, NF-κB function, as well as activity of matrix metalloproteinase (MMPs), epidermal growth factor receptor (EGFR), and Cyclooxygenase-2 (COX-2). Therefore, CAPE treatment induces cell cycle arrest and apoptosis in oral cancer cells. According to the evidence that aberrations in the EGFR/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling, NF-κB function, COX-2 activity, and MMPs activity are frequently found in oral cancers, and that the phosphorylation of Akt, EGFR, and COX-2 correlates to oral cancer patient survival and clinical progression, we believe that CAPE treatment will be useful for treatment of advanced oral cancer patients.


Subject(s)
Caffeic Acids/therapeutic use , Mouth Neoplasms/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Animals , Humans , Phenylethyl Alcohol/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...