Open Access

Inhibition of phosphoinositide 3‑kinase/Akt pathway decreases hypoxia inducible factor‑1α expression and increases therapeutic efficacy of paclitaxel in human hypoxic gastric cancer cells

  • Authors:
    • Jing Zhang
    • Hua Guo
    • Jin‑Shui Zhu
    • Yu‑Chen Yang
    • Wei‑Xiong Chen
    • Ni‑Wei Chen
  • View Affiliations

  • Published online on: March 11, 2014     https://doi.org/10.3892/ol.2014.1963
  • Pages: 1401-1408
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The phosphatidylinositol‑3‑kinase (PI3K)̸Akt signaling pathway plays an important role in cell proliferation, transformation, apoptosis, tumor growth and angiogenesis. Paclitaxel is commonly used to treat multiple human malignancies; however, the underlying mechanisms of paclitaxel in gastric cancer (GC) have not been fully investigated. In the present study, specimens from 45 GC and 36 chronic gastritis patients were collected, and the correlations of PI3K, phosphorylated‑Akt (p‑Akt) and hypoxia‑inducible factor‑1α (HIF‑1α) expression with the clinicopathological characteristics of GC were analyzed by immunohistochemistry. The human SGC‑7901 GC cells under hypoxic conditions were pretreated with the PI3K inhibitor, LY294002 (40 µM), and paclitaxel (0.1 µM). The expression levels of PI3K, p‑Akt and HIF‑1α were detected by quantitative polymerase chain reaction and western blotting. Cell proliferative activity and apoptosis were evaluated by the Cell Counting Kit‑8 assay and flow cytometry. As a result, the rates of positive expression of PI3K, p‑Akt and HIF‑1α were significantly higher in GC compared with chronic gastritis patients (each P<0.01), and were positively associated with the tumor‑node‑metastasis (TNM) staging, lymph node metastases, lymphatic infiltration and vascular infiltration (each P<0.01), but inversely correlated with tumor differentiation (P<0.01) in patients with GC. Under hypoxic conditions, the combined inhibition of the PI3K̸Akt pathway with paclitaxel markedly reduced the proliferative activity and induced cell apoptosis in GC cells compared with the single treatment of PI3K inhibitor or paclitaxel (each P<0.01), and was accompanied by a decreased expression of HIF‑1α. Overall, our findings indicate that the increased expression of the PI3K̸Akt̸HIF‑1α pathway was closely correlated with tumor differentiation, TNM staging, lymph node metastases and lymphatic and vascular infiltration. The inhibition of the PI3K̸Akt pathway enhanced the therapeutic efficacy of paclitaxel in GC cells under hypoxic conditions, suggesting that the PI3K̸Akt̸HIF‑1α pathway may act as an important therapeutic target for paclitaxel treatment of GC.

Introduction

Gastric cancer (GC) is one of the most common types of malignancies worldwide, with an estimated 934,000 cases reported globally in 2002, and is the second most common cause of cancer-related mortality (1). GC is also a genetic disease developing from a multistep process. Single or multiple mutations in genes associated with growth control, invasion and metastasis, form the molecular genetic basis of malignant transformation and tumor progression (2). Chemotherapy, to a certain extent, plays a critical role in the treatment of malignant tumors. However, the identification of predictive biomarkers of resistance or sensitivity to chemotherapy remains a fundamental challenge in the selection of patients most likely to benefit from it.

The phosphatidylinositol-3-kinase (PI3K)/Akt pathway has been shown to be activated in a variety of cancer types. Studies have shown that the increased expression of PI3K or phosphoinositide 3-Akt (p-Akt) contributes to gallbladder carcinogenesis (3) or predicts the survival of advanced endometrial carcinoma (4). Activation of the PI3K/Akt pathway is required for the apoptotic evasion (5) and is significantly associated with increasing tumor grade, decreased apoptosis and clinical outcome in human gliomas (6). It supports the development of metastatic cancer and promotes the aggressive behavior of soft tissue sarcoma (7,8), indicating the PI3K/Akt pathway as an important biomarker for the prognosis of cancer patients.

Hypoxia-inducible factor-1α (HIF-1α) plays an essential role in the adaptive response of cells to hypoxia and is associated with aggressive tumor behavior. HIF-1α is highly expressed in small-cell lung cancer and aids in predicting the overall survival of patients (9), as well as in selecting patients most likely to benefit from HIF-1α-targeted therapies (10). HIF-1α is also overexpressed in mantle cell lymphoma, where it enhances the aggressive potential and, therefore, this observation may result in more efficient target therapies (11). Notably, hypoxia induces a biphasic effect on HIF-1α stabilization with accumulation in early hypoxia, depending on activation of the PI3K/Akt pathway (12). In hypoxic tumor cells, reactive oxygen species increase HIF-1α transcription via the PI3K/Akt pathway (13), and silencing of HIF-1α suppresses tumorigenicity of renal cell carcinoma through the regulation of the PI3K/Akt pathway (14). Activation of PI3K/Akt signaling promotes the progression of hepatocarcinogenesis, while its blockade controls angiogenesis and tumor growth by regulating the expression of HIF-1α (15,16).

Notably, the expression of PI3K/Akt is increased in non-small cell lung cancer treated with adjuvant chemotherapy and serves as a novel independent prognostic biomarker (17). Deregulation of the PI3K/Akt pathway is associated with resistance to the chemotherapeutic agent and confers drug resistance to treatment with paclitaxel in breast cancer (18,19). Although certain studies have demonstrated the enhanced effectiveness of targeting tumor cells with combinations of chemotherapeutic agents and signal transduction inhibitors (20), the enhancing effects of blockade of the PI3K/Akt pathway on paclitaxel in hypoxic GC cells remains unclear. In the present study, the correlations of PI3K, p-Akt and HIF-1α expression with the clinicopathological characteristics of patients with GC were analyzed. Hypoxic GC SGC-7901 cells were pretreated with LY294002 and/or paclitaxel to investigate the enhancing effects of the PI3K inhibitor on paclitaxel through cell proliferation activity and apoptosis.

Materials and methods

Materials

The human GC SGC7901 cell line was donated from the Shanghai Tumor Research Institute (no. 01842; Shanghai, China). The primers of PI3K and HIF-1α were synthesized by the Shanghai Biological Engineering Technology Co., Ltd. (Shanghai, China). The PI3K rabbit-anti-human polyclonal antibody (sc-1637) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA); p-Akt rabbit-anti-human polyclonal antibody (D9E) and β-actin were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA); and HIF-1α rabbit-anti-human polyclonal antibody (BA0912) was obtained form Wuhan Boster Biological Engineering Co., Ltd (Wuhan, China). The GC tissue samples treated with paclitaxel were obtained from severe combined immune deficiency (SCID) mice orthotopically implanted with human GC cells from the Gastrointestinal Disease Laboratory of Shanghai Sixth People’s Hospital (Shanghai, China).

Drugs and reagents

Paclitaxel was donated by Professor G-Y Fan from the Kunming Botany Institute (Yunnan, China); the Cell Counting Kit-8 (CCK-8) was purchased from Tongren Chemical Institute (Japan); Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were purchased from Thermo Fisher Scientific Inc. (Waltham, MA, USA); TRIzol® reagent and LY294002 were obtained from Invitrogen Life Technologies (Carlsbad, CA, USA); M-MLV Reverse Transcriptase was purchased from Promega Corporation (Madison, WI, USA); SYBR Green master mix was purchased from Takara Bio, Inc., (Otsu, Japan); the Annexin V-fluorescein isothiocyanate cell apoptosis detection kit was obtained from KeyGen Biotech., Co., Ltd. (ab14085; Nanjing, China); and the Enhanced Chemiluminescence (ECL)-PLUS™ western blotting reagents were purchased from GE Healthcare (Piscataway, NJ, USA).

Clinical samples and data

A total of 45 patients with GC and 36 patients with chronic gastritis were enrolled in this study at the General Surgery and Digestive Endoscopy Room from December, 2007 to June, 2008. The pathological staging was determined according to the American Joint Committee on Cancer tumor-node-metastasis (TNM) staging system. The use of the tissue samples and clinical data was approved by the Medical Ethics Committee of Shanghai Jiao Tong University (Shanghai, China).

Immunohistochemical analysis

The protein expression of PI3K, p-Akt and HIF-1α were analyzed by immunohistochemical staining. The anti-PI3K, p-Akt and HIF-1α antibodies were used at 1:100 dilutions. Endogenous peroxidase was inhibited by incubation with freshly prepared 3% hydrogen peroxide with 0.1% sodium azide. Non-specific staining was blocked with 0.5% casein and 5% normal serum. The tissues were incubated with biotinylated antibodies and horseradish peroxidase (Cell Signaling Technology, Inc.). Staining was developed with diaminobenzidine substrate and sections were counterstained with hematoxylin and eosin. Normal serum or phosphate-buffered saline (PBS) replaced the antibodies in negative controls. The images were analyzed with Image-Pro Plus 4.5 System (Media Cybernetics, Inc., Rockville, MD, USA). The total optical density value and area of intracellular fluorescence for each section was measured.

Cell culture and pretreatment

The SGC7901 cells were cultured in DMEM supplemented with 10% heat-inactivated FBS, 100 U/ml penicillin and 100 μg/ml streptomycin. The cells were stored in a humidified atmosphere of 5% CO2 at 37°C for 30 min. Under normal oxygen, the cells were incubated in 20% O2 and 5% CO2 with saturated humidity at 37°C for 30 min. The hypoxic cells were incubated in an AnaeroPack™ containing 20% CO2 and <1% O2 at 37°C for 30 min. The cells pretreated under hypoxic conditions were further treated with the PI3K inhibitor, LY294002, (40 Mm) for 30 min, followed by paclitaxel (0.1 Mm).

Quantitative polymerase chain reaction (qPCR)

To quantitatively determine the mRNA expression levels of PI3K and HIF-1α in GC SGC-7901 cells, qPCR was used. Total RNA of each clone was extracted with TRIzol reagent according to the manufacturer’s instructions. Reverse-transcription using M-MLV, and cDNA amplification using SYBR Green master mix kit, were performed according to the manufacturer’s protocol. The genes were amplified using specific oligonucleotide primers and a human β-actin gene was used as an endogenous control. The PCR primer sequences were as follows: Forward, 5′-AAGAAGCAAGCAGCTGAG-3′ and reverse, 5′-CTACAGAGCAGGCATAG-3′ for PI3K; forward, 5′-TCAAAGTCGGACAGCCTC-3′ and reverse, 5′-CCAGCAGTCTACATGC-3′ for HIF-1α; forward, 5′-CTTCGAGCAAGAGATGGC-3′ and reverse, 5′-CTCCTTCTGCATCCTGTC-3′ for β-actin. Data were analyzed using the comparative Ct method (2−ΔΔCt). Three separate experiments were performed for each clone.

Western blot analysis

The hypoxic SGC-7901 cells treated with LY294002 and/or paclitaxel were harvested and extracted using lysis buffer [Tris-HCl, sodium-dodecyl sulfate (SDS), mercaptoethanol and glycerol] purchased from Santa Cruz Biotechnology, Inc. Cell extracts were heated to boiling point for 5 min in loading buffer and equal amounts of cell extracts were separated on 15% SDS-PAGE gels. Separated protein bands were transferred into polyvinylidene fluoride membranes, which were blocked in 5% skimmed milk powder. The primary antibodies against PI3K, p-Akt and HIF-1α were diluted according to the manufacturer’s instructions and incubated overnight at 4°C. Horseradish peroxidase-linked secondary biotinylated antibodies were added at a dilution ratio of 1:1,000 and incubated at room temperature for 2 h. The membranes were washed with PBS three times and the immunoreactive bands were visualized using the ECL-PLUS/Kit according to the manufacturer’s instructions. The relative protein levels in different cell lines were normalized to the GAPDH concentration. Three separate experiments were performed for each clone. Finally, the immune complexes were developed using an ECL detection kit according to the manufacturer’s instructions (ECL GST western blotting detection kit, Pierce Biotechnology, Inc., Waltham, MA, USA) and the GelGDoc2000 imaging system (Bio-Rad Laboratories GmbH, Munich, Germany) was employed to analyze the bands, and the protein levels by the relative optical density.

CCK-8 assay

Cell proliferation following treatment with LY294002 and/or paclitaxel was measured by the CCK-8 assay. The GC SGC-7901 cells were seeded at a density of 2×104 cells/100 μl/well in 96-well plates and left to attach overnight. The medium was then removed and 200 μl of FBS was added followed by LY294002 and/or paclitaxel. The cells were incubated under these conditions for 24, 48, 72, 96, 120 and 148 h at 37°C in a humidified atmosphere of 5% CO2. After the designated time, CCK-8 was added to each well containing 200 μl of the culture medium and the oligopeptide mixture, and further incubated for 4 h at 37°C. The amount of formazan dye was measured at 450 nm using the Multi-Mode Microplate Reader (Nanchang Biotek Medical Device Co., Ltd., Nanchang, China). All the experiments were performed in triplicate and repeated three times.

Cell apoptosis analysis

Cell apoptosis detection was performed using the BD Accuri™ C6 Flow cytometer (BD Biosciences, San Jose, CA, USA). The exposure of PBS on the extracellular side of the cell membrane was quantified by propidium iodide (PI) staining (Invitrogen Life Technologies). The SGC-7901 cells were placed in six-well plates and, after 24 h of incubation, the cells were treated with LY294002 and/or paclitaxel for 24 h and then harvested. Following centrifugation, cell pellets were washed twice with cold PBS. The cells were then incubated with 5 μl of PI at room temperature for 15 min in the dark. Following incubation, 400 μl of 1X binding buffer was added to each tube. The cells were immediately analyzed by flow cytometry.

Statistical analysis

Data are expressed as the means ± standard deviation where applicable. Statistically significant differences in each assay were determined by SPSS software, version 20.0 (SPSS, Inc., Chicago, IL, USA). Differences in each group were tested for significance using Student’s t-test or one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference.

Results

Correlations of PI3K, p-Akt and HIF-1α expression with the clinicopathological characteristics

The GC tissue sections were analyzed by immunohistochemistry (IHC) and Image-Pro Plus 4.5 software (Media Cybernetics, Inc.). As shown in Fig. 1 and Table I, the positive expression of PI3K, p-Akt and HIF-1α was predominantly localized in the cytoplasm of the GC tissue cells, but was not identified in the chronic gastritis cells. The expression intensities of PI3K, p-Akt and HIF-1α were significantly increased in GC tissues compared with chronic gastritis tissues (each P<0.01).

Table I

Expression of PI3K, p-AKT and HIF-1α in gastric cancer (Gray values).

Table I

Expression of PI3K, p-AKT and HIF-1α in gastric cancer (Gray values).

MarkersGroupCasesGray valueP-value
PI3KGastric cancer45202.4±10.7<0.01
Chronic gastritis36220.3±4.3
p-AKTGastric cancer45223.6±11.7<0.01
Chronic gastritis36234.0±4.6
HIF-1αGastric cancer45200.3±6.9<0.01
Chronic gastritis36218.7±4.4

[i] PI3K, phosphatidylinositol-3-kinase; p-AKT, phosphorylated-Akt; HIF-1α, hypoxia-inducible factor-1α.

The correlations of PI3K, p-Akt and HIF-1α expression and various clinical and pathological characteristics were analyzed. As summarized in Table II, no significant correlation was found between PI3K, p-Akt and HIF-1α expression and gender, age, tumor size and peripheral nerve infiltration in patients with GC (each P>0.05), while their expression was significantly correlated with TNM staging, lymph node metastases, lymphatic infiltration and vascular infiltration (each P<0.01), but inversely correlated with tumor differentiation (P<0.01).

Table II

Correlation of PI3K, p-AKT and HIF-1α expression with the clinicopathological characteristics of patients with GC.

Table II

Correlation of PI3K, p-AKT and HIF-1α expression with the clinicopathological characteristics of patients with GC.

VariablesCasesPI3KP-valuep-AKTP-valueHIF-1αP-value
Age, years
 ≤6823201.0±9.20.052223.7±10.70.841200.7±6.80.335
 >6822203.9±10.3223.4±11.7199.8±7.0
Gender
 Male32202.9±10.90.279223.9±11.00.526200.7±6.60.142
 Female13201.2±10.4223.0±11.7199.2±7.4
Tumor size, cm
 ≤530202.4±8.40.926224.2±10.80.256200.8±5.40.184
 >515202.9±8.4200.3±6.9199.3±9.1
Degree of differentiation
 Well/moderately15206.3±10.9<0.01227.6±10.6<0.01202.1±6.30.005
 Poorly30200.4±10.2221.6±10.9199.4±7.0
TNM staging
 I+II20207.7±8.8<0.01228.6±9.5<0.01204.3±5.6<0.01
 III+IV25198.1±10.3219.6±10.8197.1±6.1
Lymph node metastases
 No19208.7±8.4<0.01228.7±9.6<0.01204.3±5.7<0.01
 Yes26197.8±10.2219.8±10.7197.3±6.1
Lymphatic vessel infiltration
 −11207.6±7.0<0.01230.7±8.4<0.01205.7±5.3<0.01
 +34200.7±11.2221.3±11.0198.5±6.4
Vascular infiltration
 −29205.0±9.9<0.01226.6±10.6<0.01201.6±7.2<0.01
 +16197.6±10.4218.1±10.0197.9±5.5
Perineural infiltration
 −6203.6±6.60.324225.7±10.40.265202.5±4.80.053
 +39202.2±11.3223.2±11.3199.9±7.1

[i] PI3K, phosphatidylinositol-3-kinase; p-AKT, phosphorylated-Akt; HIF-1α, hypoxia-inducible factor-1α; TNM, tumor-node-metastasis.

Effects of LY294002 and/or paclitaxel on the expression of PI3K, p-Akt and HIF-1α

qPCR and western blot analysis were performed to detect the effects of LY294002 and paclitaxel on the expression of PI3K, p-Akt and HIF-1α in the GC SGC-7901 cells. LY294002 combined with paclitaxel markedly inhibited the mRNA (Fig. 2A and B) expression levels of PI3K and HIF-1α (it was not necessary to detect the expression of p-Akt as it is downstream of PI3K) and protein (Fig. 3A and B) expression levels of PI3K, p-Akt and HIF-1α in GC SGC-7901 cells compared with the single treatment of LY294002 or paclitaxel (each P<0.01). LY294002 or paclitaxel decreased the expression of PI3K, p-Akt and HIF-1α at the transcriptional and translational levels compared with the hypoxic group (each P<0.01).

Effects of LY294002 and/or paclitaxel on cell proliferation

To determine whether LY294002 and/or paclitaxel affect the proliferative activity of hypoxic GC cells, the CCK-8 assay was performed to detect cell viability. As summarized in Table III, LY294002 combined with paclitaxel significantly decreased the cell viability in SGC-7901 cells compared with the single treatment of LY294002 or paclitaxel (each P<0.01). In addition, LY294002 or paclitaxel reduced cell viability compared with the hypoxic group (each P<0.01).

Table III

Proliferative activity of gastric cancer cells (optical density values).

Table III

Proliferative activity of gastric cancer cells (optical density values).

Day

Groups123456
Normoxia0.45±0.010.75±0.021.25±0.061.60±0.041.86±0.042.12±0.10
Normoxia+paclitaxel0.43±0.020.60±0.03a0.87±0.0a1.22±0.04a1.54±0.05a1.74±0.07a
Hypoxia0.46±0.030.68±0.04a1.05±0.05a1.40±0.07a1.65±0.02a1.85±0.04a
Hypoxia+paclitaxel0.44±0.060.59±0.04a,b0.83±0.05a,b1.11±0.05a,b1.38±0.07a,b1.60±0.10a,b
Hypoxia+LY2940020.45±0.040.64±0.03ac0.85±0.02ac1.10±0.10a,b1.33±0.12ac1.61±0.11ac
Hypoxia+paclitaxel+LY2940020.42±0.070.53±0.03ac0.67±0.03ac0.87±0.03ac1.11±0.12ac1.29±0.04ac

a P<0.01, vs. normoxia;

b P<0.01, vs. hypoxia;

c P<0.01, vs. hypoxia+paclitaxel.

Effects of LY294002 and/or paclitaxel on cell apoptosis

Cell apoptosis was measured by flow cytometry using PI staining. Following treatment with LY294002 and/or paclitaxel for 24 h, LY294002 combined with paclitaxel significantly increased the percentage of early cell apoptosis compared with the single treatment of LY294002 or paclitaxel (each P<0.01) (Fig. 4A and B). LY294002 or paclitaxel increased the percentage of early cell apoptosis compared with the hypoxic group (both P<0.01).

Effects of paclitaxel on the expression of PI3K, p-Akt and HIF-1α in vivo

SCID mice orthotopically implanted with human GC tissues were treated with paclitaxel (intraperitoneal administration of 5 mg/kg) and the effects of paclitaxel on the expression of PI3K, p-Akt and HIF-1α were assessed by IHC. Paclitaxel decreased the expression of PI3K, p-Akt and HIF-1α (Fig. 5) compared with the GC and normal control groups (each P<0.01).

Discussion

The PI3K/Akt pathway is one of the most important signaling networks in cancer. Emerging evidence indicates that activation of this pathway plays a significant role in the development and progression of certain malignancies. Zhang et al reported that the PI3K/Akt pathway is expressed in 71.7 (43/60) and 68.3% (41/60) of colon cancer and is closely associated with serous coat infiltration and lymphatic metastasis (21), serving as an independent prognostic marker for patients with colorectal cancer (CRC) (22). HIF-1α is a transcription factor recognized to control the delivery of oxygen and nutrients through the induction of angiogenesis under hypoxic conditions. Overexpression of HIF-1α is significantly correlated with histology, depth of invasion and poor prognosis for patients with GC, and may be utilized for tumor-specific molecular target-based therapy (23). In the present study, the positive expression of PI3K, p-Akt and HIF-1α were significantly increased in GC tissues compared with chronic gastritis, and were associated with TNM staging, lymph node metastases and lymphatic and vascular infiltration, but inversely correlated with tumor differentiation. This suggested that PI3K, p-Akt and HIF-1α are potential therapeutic targets for GC.

Inhibiting the molecules involved in the PI3K/Akt or HIF-1α signal transduction pathway is a possible strategy for the treatment of cancer. The PI3K inhibitor and conventional chemotherapy provide an effective approach to inhibiting tumor growth in ovarian cancer (24). Paclitaxel is extensively used in chemotherapy for various cancers. PI3K is involved in low susceptibility of CRC to paclitaxel and PI3K-targeting agents may enable a new paclitaxel-based chemotherapy for CRC (25). Furthermore, the Akt inhibitor increases the therapeutic efficacy of paclitaxel for patients with ovarian cancer (26). HIF-1α affects the sensitivity of paclitaxel in lung cancer cells and targeted inhibition of HIF-1α may overcome the drug resistance of paclitaxel (27). However, an individual study has reported that pharmacological PI3K/Akt inhibition antagonizes the efficacy of chemotherapeutic agents primarily effective in the S or G2 phase of the cell cycle (28). Our current study indicated that combining the PI3K inhibitor, LY294002, with paclitaxel significantly decreased cell viability and increased cell apoptosis in hypoxic GC SGC-7901 cells compared with the single treatment of LY294002 or paclitaxel. Moreover, treatment with LY294002 or paclitaxel reduced cell viability and induced apoptosis compared with the hypoxic GC cells, suggesting that PI3K/Akt-targeted therapy with paclitaxel is more efficacious for treating GC with activated PI3K/Akt signaling.

Notably, the PI3K/Akt pathway regulates HIFα activity (29), and HIF-1α-dependent transcription is blocked by negative Akt or PI3K and by the wild-type phosphatase and tensin homolog (30). Direct evidence has also revealed that activation of the PI3K/Akt/HIF-1α pathway plays a critical role in mediating hypoxia-induced drug resistance resulting in an unfavorable treatment outcome of hepatocellular carcinoma (31). However, Manohar et al (32) reported that the HIF-1α inhibitor modulates the PI3K/Akt pathway and contributes to antitumor activity. In the present study, LY294002 combined with paclitaxel markedly inhibited the expression of PI3K, p-Akt and HIF-1α in GC SGC-7901 cells compared with the single treatment of LY294002 or paclitaxel, and LY294002 or paclitaxel decreased the expression of PI3K and p-Akt in hypoxic conditions, suggesting that targeting PI3K/Akt signaling in tumor cells may inhibit HIF-1α expression and increase the therapeutic efficacy of paclitaxel.

In conclusion, our findings suggest that the increased expression of the PI3K/Akt or HIF-1α pathway is closely correlated with tumor differentiation, TNM staging, lymph node metastases, and lymphatic and vascular infiltration. In addition, inhibition of the PI3K/Akt pathway enhanced the therapeutic efficacy of paclitaxel in GC cells under hypoxic conditions, suggesting that the PI3K/Akt or HIF-1α pathway may serve as an important therapeutic target for the paclitaxel treatment of cancer.

Acknowledgements

This study was supported by the National Nature Science Foundation of China (grant nos. 81272752 and 81302093).

References

1 

Jemal A, Bray F, Center MM, et al: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

2 

Tajima Y, Yamazaki K, Makino R, et al: Gastric and intestinal phenotypic marker expression in early differentiated-type tumors of the stomach: clinicopathologic significance and genetic background. Clin Cancer Res. 12:6469–6479. 2006. View Article : Google Scholar

3 

Li Q and Yang Z: Expression of phospho-ERK1/2 and PI3-K in benign and malignant gallbladder lesions and its clinical and pathological correlations. J Exp Clin Cancer Res. 28:652009. View Article : Google Scholar : PubMed/NCBI

4 

Uegaki K, Kanamori Y, Kigawa J, et al: PTEN-positive and phosphorylated-Akt-negative expression is a predictor of survival for patients with advanced endometrial carcinoma. Oncol Rep. 14:389–392. 2005.

5 

Sun ZJ, Chen G, Hu X, et al: Activation of PI3K/Akt/IKK-alpha/NF-kappaB signaling pathway is required for the apoptosis-evasion in human salivary adenoid cystic carcinoma: its inhibition by quercetin. Apoptosis. 15:850–863. 2010. View Article : Google Scholar

6 

Chakravarti A, Zhai G, Suzuki Y, et al: The prognostic significance of phosphatidylinositol 3-kinase pathway activation in human gliomas. J Clin Oncol. 22:1926–1933. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Xue G, Restuccia DF, Lan Q, et al: Akt/PKB-mediated phosphorylation of Twist1 promotes tumor metastasis via mediating cross-talk between PI3K/Akt and TGF-β signaling axes. Cancer Discov. 2:248–259. 2012.PubMed/NCBI

8 

Valkov A, Kilvaer TK, Sorbye SW, et al: The prognostic impact of Akt isoforms, PI3K and PTEN related to female steroid hormone receptors in soft tissue sarcomas. J Transl Med. 9:2002011. View Article : Google Scholar : PubMed/NCBI

9 

Lee GW, Go SI, Cho YJ, et al: Hypoxia-inducible factor-1α and excision repair cross-complementing 1 in patients with small cell lung cancer who received front-line platinum-based chemotherapy: a retrospective study. J Thorac Oncol. 7:528–534. 2012.

10 

Park S, Ha SY, Cho HY, et al: Prognostic implications of hypoxia-inducible factor-1α in epidermal growth factor receptor-negative non-small cell lung cancer. Lung Cancer. 72:100–107. 2011.

11 

Argyriou P, Papageorgiou SG, Panteleon V, et al: Hypoxia-inducible factors in mantle cell lymphoma: implication for an activated mTORC1→HIF-1α pathway. Ann Hematol. 90:315–322. 2011.PubMed/NCBI

12 

Mottet D, Dumont V, Deccache Y, et al: Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J Biol Chem. 278:31277–31285. 2003. View Article : Google Scholar

13 

Koshikawa N, Hayashi J, Nakagawara A and Takenaga K: Reactive oxygen species-generating mitochondrial DNA mutation up-regulates hypoxia-inducible factor-1alpha gene transcription via phosphatidylinositol 3-kinase-Akt/protein kinase C/histone deacetylase pathway. J Biol Chem. 284:33185–33194. 2009. View Article : Google Scholar

14 

Xu K, Ding Q, Fang Z, et al: Silencing of HIF-1alpha suppresses tumorigenicity of renal cell carcinoma through induction of apoptosis. Cancer Gene Ther. 17:212–222. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Tanaka H, Yamamoto M, Hashimoto N, et al: Hypoxia-independent overexpression of hypoxia-inducible factor 1alpha as an early change in mouse hepatocarcinogenesis. Cancer Res. 66:11263–11270. 2006. View Article : Google Scholar

16 

Fang J, Ding M, Yang L, et al: PI3K/PTEN/AKT signaling regulates prostate tumor angiogenesis. Cell Signal. 19:2487–2497. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Shi Y, Chen L, Li J, et al: Prognostic and predictive values of pERK1/2 and pAkt-1 expression in non-small cell lung cancer patients treated with adjuvant chemotherapy. Tumour Biol. 32:381–390. 2011. View Article : Google Scholar

18 

McCubrey JA, Steelman LS, Abrams SL, et al: Roles of the RAF/MEK/ERK and PI3K/PTEN/AKT pathways in malignant transformation and drug resistance. Adv Enzyme Regul. 46:249–279. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Liang K, Lu Y, Li X, et al: Differential roles of phosphoinositide-dependent protein kinase-1 and akt1 expression and phosphorylation in breast cancer cell resistance to Paclitaxel, Doxorubicin, and gemcitabine. Mol Pharmacol. 70:1045–1052. 2006. View Article : Google Scholar

20 

Abrams SL, Steelman LS, Shelton JG, et al: Enhancing therapeutic efficacy by targeting non-oncogene addicted cells with combinations of signal transduction inhibitors and chemotherapy. Cell Cycle. 9:1839–1846. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Zhang Y, Liu X, Zhang J, et al: The expression and clinical significance of PI3K, pAkt and VEGF in colon cancer. Oncol Lett. 4:763–766. 2012.PubMed/NCBI

22 

Kim JG, Chae YS, Sohn SK, et al: Clinical significance of genetic variations in the PI3K/PTEN/AKT/mTOR pathway in Korean patients with colorectal cancer. Oncology. 79:278–282. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Isobe T, Aoyagi K, Koufuji K, et al: Clinicopathological significance of hypoxia-inducible factor-1 alpha (HIF-1α) expression in gastric cancer. Int J Clin Oncol. 18:293–304. 2013.

24 

Hu L, Hofmann J, Lu Y, et al: Inhibition of phosphatidylinositol 3′-kinase increases efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Cancer Res. 62:1087–1092. 2002.

25 

Xu R, Nakano K, Iwasaki H, et al: Dual blockade of phosphatidylinositol 3′-kinase and mitogen-activated protein kinase pathways overcomes paclitaxel-resistance in colorectal cancer. Cancer Lett. 306:151–160. 2011.

26 

Kim SH, Juhnn YS and Song YS: Akt involvement in paclitaxel chemoresistance of human ovarian cancer cells. Ann N Y Acad Sci. 1095:82–89. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Zeng L, Kizaka-Kondoh S, Itasaka S, et al: Hypoxia inducible factor-1 influences sensitivity to paclitaxel of human lung cancer cell lines under normoxic conditions. Cancer Sci. 98:1394–1401. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Fekete M, Santiskulvong C, Eng C and Dorigo O: Effect of PI3K/Akt pathway inhibition-mediated G1 arrest on chemosensitization in ovarian cancer cells. Anticancer Res. 32:445–452. 2012.PubMed/NCBI

29 

Shafee N, Kaluz S, Ru N and Stanbridge EJ: PI3K/Akt activity has variable cell-specific effects on expression of HIF target genes, CA9 and VEGF, in human cancer cell lines. Cancer Lett. 282:109–115. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Zhong H, Chiles K, Feldser D, et al: Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 60:1541–1545. 2000.

31 

Jiao M and Nan KJ: Activation of PI3 kinase/Akt/HIF-1α pathway contributes to hypoxia-induced epithelial-mesenchymal transition and chemoresistance in hepatocellular carcinoma. Int J Oncol. 40:461–468. 2012.

32 

Manohar SM, Padgaonkar AA, Jalota-Badhwar A, et al: A novel inhibitor of hypoxia-inducible factor-1α P3155 also modulates PI3K pathway and inhibits growth of prostate cancer cells. BMC Cancer. 11:3382011.

Related Articles

Journal Cover

May-2014
Volume 7 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang J, Guo H, Zhu JS, Yang YC, Chen WX and Chen NW: Inhibition of phosphoinositide 3‑kinase/Akt pathway decreases hypoxia inducible factor‑1α expression and increases therapeutic efficacy of paclitaxel in human hypoxic gastric cancer cells. Oncol Lett 7: 1401-1408, 2014
APA
Zhang, J., Guo, H., Zhu, J., Yang, Y., Chen, W., & Chen, N. (2014). Inhibition of phosphoinositide 3‑kinase/Akt pathway decreases hypoxia inducible factor‑1α expression and increases therapeutic efficacy of paclitaxel in human hypoxic gastric cancer cells. Oncology Letters, 7, 1401-1408. https://doi.org/10.3892/ol.2014.1963
MLA
Zhang, J., Guo, H., Zhu, J., Yang, Y., Chen, W., Chen, N."Inhibition of phosphoinositide 3‑kinase/Akt pathway decreases hypoxia inducible factor‑1α expression and increases therapeutic efficacy of paclitaxel in human hypoxic gastric cancer cells". Oncology Letters 7.5 (2014): 1401-1408.
Chicago
Zhang, J., Guo, H., Zhu, J., Yang, Y., Chen, W., Chen, N."Inhibition of phosphoinositide 3‑kinase/Akt pathway decreases hypoxia inducible factor‑1α expression and increases therapeutic efficacy of paclitaxel in human hypoxic gastric cancer cells". Oncology Letters 7, no. 5 (2014): 1401-1408. https://doi.org/10.3892/ol.2014.1963