Immunohistochemical expression of mTOR negatively correlates with PTEN expression in gastric carcinoma

  • Authors:
    • Min Li
    • Huawen Sun
    • Lujun Song
    • Xiaodong Gao
    • Wenju Chang
    • Xinyu Qin
  • View Affiliations

  • Published online on: September 21, 2012     https://doi.org/10.3892/ol.2012.930
  • Pages: 1213-1218
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The phosphoinositide‑3 kinase (PI3K)‑AKT-mammalian target of rapamycin (mTOR) pathway is a cellular pathway involved in cell growth, tumorigenesis and cell invasion which is frequently activated in various types of cancer. The downstream effector of the pathway is mTOR which is important in cellular growth and homeostasis and aberrant activation of mTOR has been reported in several types of cancer. The tumor suppressor gene phosphatase and tensin homolog (PTEN) is essential in this pathway for inhibiting tumor invasion and metastasis. However, the involvement of mTOR and PTEN in the progression of human gastric cancer remains to be identified. Immunohistochemical staining was performed to detect the expression of mTOR and PTEN in paraffin‑embedded gastric tissue sections obtained from 33 patients with gastric cancer and 30 normal controls. The expressed mTOR was mainly distributed in the cytoplasm, while PTEN was mainly localized to the nucleus. By considering negative mTOR expression with positive PTEN expression as one group and negative PTEN expression with positive mTOR expression as the other, significant statistical differences were observed in various categories, including histological types and metastatic and clinical pathology stages, between the 2 groups (P<0.01 or 0.05). The results indicated that the expression levels of mTOR and PTEN were negatively correlated in the PI3K‑AKT‑mTOR signaling pathway. Combined detection of mTOR and PTEN expression may be used to evaluate the degree of malignancy in gastric cancer and may be a useful marker for the early diagnosis of gastric cancer.

Introduction

Despite a declining incidence rate in the United States and a number of other Western countries, gastric cancer continues to be a worldwide health problem with more than 600,000 cases reported annually, far higher than pancreatic cancer with 125,000 cases (1). Gastric cancer is the most common gastrointestinal malignancy in East Asia, Eastern Europe and parts of Central and South America and is the second leading cause of cancer-related mortality (2). Despite improvements in surgery, radiotherapy and cytotoxic chemotherapy, survival rates for advanced gastric cancer are poor. Five years after multimodal treatment, less than 40% of Western patients with stage II or III disease are likely to be alive. At metastatic stage IV disease, the mean survival is only 10 months (3).

The phosphoinositide-3 kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) pathway is an important cellular pathway involved in cell growth, tumorigenesis, cell invasion and drug response (4,5). PI3K is a heterodimer of 85- and 110-kDa subunits and has a tyrosine kinase activity, the activation of which stimulates the production of phosphatidylinositol 3,4,5-triphosphate resulting in activation of the kinases PDK1 and Akt. Akt is a kinase that phosphorylates a variety of target molecules to mediate signals, including mTOR, while mTOR phosphorylates and activates p70 S6 kinase (S6K)-1 and also inhibits eukaryotic translation initiation factor 4E-binding protein (4E-BP), resulting in enhanced protein synthesis and cell proliferation (68). This pathway is frequently activated in numerous types of cancer and uncontrolled PI3K-AKT-mTOR signaling may also result in a poor clinical outcome in lung, cervical, ovarian and esophageal cancers (4,6).

mTOR was identified in 1994 by several groups of investigators as the kinase targeted by rapamycin linked to the cellular protein FKBP12 (FK506-binding protein) (9). It was therefore also named FKBP-RAP-associated protein (FRAP), RAP FKBP12 target (RAFT) 1 and RAP target (RAPT) 1. mTOR is a 289-kDa, ubiquitously expressed, evolutionarily conserved serine/threonine protein kinase (9) which is important in cellular protein synthesis and energy balance, affecting numerous aspects of cell growth and proliferation, including differentiation, cell-cycle progression, angiogenesis, protein degradation and apoptosis (10). mTOR is also instrumental in protein translation initiation (the rate-limiting step of protein synthesis) by enabling the recruitment of ribosomes to mRNA by eukaryotic initiation factor. Consequently, mTOR activates its downstream mediator ribosomal S6K and is responsible for the progression of the cell from G0/G1 to S phase (11). Consistent with its essential role in cell growth, aberrant activity of the mTOR pathway is frequently observed in a number of types of cancer (12).

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is critical in cell growth, migration and death. It is mutated or deleted at a high frequency in various human cancer tissues to promote tumorigenesis (13). The PI3K-AKT-mTOR pathway is one of the most upregulated pathways in neoplastic cells through mechanisms such as PTEN loss of function or PI3K activating mutations (14). PTEN antagonizes PI3K pathways by dephosphorylating phosphatidylinositol 3,4,5-triphosphate to convert it back to phosphatidylinositol 4,5-biphosphate. Thus, PTEN is considered to be a negative regulator of the PI3K-AKT-mTOR pathway (15).

The aim of the present study was to explore the involvement of the PI3K-AKT-mTOR signaling pathway in the progression of human gastric cancer. The expression levels of mTOR and PTEN in human gastric cancer tissues were determined using immunohistochemical study using biopsies from 33 patients and correlations with pathological parameters and prognoses were evaluated.

Materials and methods

Patients and tumor samples

Paraffin-embedded sections were obtained from patients with gastric cancer who had undergone surgery at Renmin Hospital of Wuhan University (Wuhan, China) between 2005 and 2008. Tissues obtained from 30 cases of chronic superficial gastritis diagnosed by gastroscopic biopsy were used as control samples for the immunohistochemical staining. General informed consent with regard to the use of the patients’ tissue specimens and clinical information was obtained from all patients. None of the patients recruited in this study had undergone chemotherapy or radiotherapy prior to surgery. The patients’ clinicopathological data are summarized in Table I. The histological diagnosis was determined using hematoxylin and eosin staining according to the WHO criteria (16). Pathological staging was performed according to the American Joint Committee on Cancer (AJCC) Cancer Staging Manual revised in 2010 (17). This study was approved by the Institutional Ethics Board of the Renmin Hospital of Wuhan University.

Table I.

Clinicopathological parameters of the patients in the study.

Table I.

Clinicopathological parameters of the patients in the study.

FactorsValues
Age, years, mean (range)51.6 ±11.6 (24–74)
Gender, n (%)
  Male15 (45.5)
  Female18 (54.5)
Invasive depth, n (%)
  Early stage6 (18.2)
  Advanced stage27 (81.8)
Differentiation, n (%)
  Well and moderate15 (45.5)
  Poor18 (54.5)
Lymph node metastasis, n (%)
  Positive16 (48.5)
  Negative17 (51.5)
Pathological stage, n (%)
  I+II18 (54.5)
  III+IV15 (45.5)
Immunohistochemical staining

For malin-fixed, paraffin-embedded tissue blocks obtained from human tissue were cut into 4-μm thick sections and mounted on adhesive-coated glass slides. mTOR was detected with a rabbit monoclonal anti-mTOR antibody (Cell Signaling Technology, Inc., Danvers, MA, USA) and PTEN was observed with a mouse monoclonal anti-PTEN antibody (Maxim Biotechnology Development Co. Ltd, Fuzhou, China). Sections were dewaxed in xylene and rehydrated using graded ethanol and were then incubated in 3% solution of hydrogen peroxide in methanol for 10 min to inactivate endogenous peroxidase. This was followed by an antigen retrieval step. The slides were placed in 0.01 mol/l citrate buffer solution (pH 6.0) and heated (92–100°C) for 10 min in a microwave oven. Following a wash with phosphate-buffered saline (PBS; pH 7.2) buffer, the sections were covered with 2% normal serum for 20 min at room temperature to eliminate non-specific binding of the antibody and were then incubated overnight at 4°C with the primary antibodies diluted in PBS. After washing with PBS, the secondary biotinylated antibody was added for a 20 min incubation at 37°C. Slides were then rinsed with PBS and treated with streptavidin-peroxidase solution for 10 min. Tissue sections were washed once in PBS buffer and covered with 3,3′-diaminobenzidine solution for 10 min. Finally, the specimens were counterstained with hematoxylin. Normal gastric sections served as positive controls while negative control slides were incubated with the antibody diluents instead of the primary antibody.

Evaluation of slides

Immunohistochemical staining was evaluated by 3 independent experienced pathologists who were blinded to the clinicopathological parameters and clinical outcomes of the patients. In cases of disagreement between the observers slides were re-evaluated until a consensus was achieved. The sections were examined at x200 magnification using light microscopy. The immunostaining was considered to be positive when the neoplastic cells exhibited specific immunoreactivity in the cytoplasm for mTOR or in the nucleus for PTEN. The immunostaining results were assessed semiquantitatively. For each sample the positive rate was calculated according to the percentage of positive cells of all counted cells from 5 randomly selected representative fields. Additionally, the expression was classified according to the percentage of stained tumor cells as low expression (−, <10% positive carcinoma cells), intermediate expression (+, ≥10% and <50% positive carcinoma cells) and high expression (++, ≥50% positive carcinoma cells) (18).

Statistical analysis

Statistical analysis was performed using the PASW 18.0 software program for Windows. The results for the correlation between mTOR and PTEN were evaluated using the Chi-squared test. P<0.05 was considered to indicate statistically significant differences.

Results

Differential expression of mTOR and PTEN in human gastric cancer

The expression levels and cellular distribution of mTOR and PTEN in the 33 specimens of human gastric cancer and 30 normal gastric tissues were examined by immunohistochemical staining. mTOR was distributed mainly in the cytoplasm. Staining was weaker in low-grade tumors. Positive mTOR expression was observed in tumor cells in 51.5% (17/33) of the gastric cancer patients. By contrast, little or no expression of mTOR was observed in normal gastric tissues (Fig. 1). PTEN was distributed mainly in the nuclei. Staining was stronger in low-grade tumors. Positive PTEN expression was observed in tumor cells in 54.5% (18/33) of the gastric cancer patients (Fig. 2).

The expression of the mTOR and PTEN antigens was assessed by immunohistochemical staining in sections obtained from 33 gastric cancer patients with various histological diagnoses and pathological staging according to the AJCC handbook (Table II). In early and advanced cases, the respective positive expression rates were 16.7% (1/6) and 59.3% (16/27) for mTOR and 100.0% (6/6) and 44.4% (12/27) for PTEN. In well- and moderately differentiated tissues and in poorly differentiated specimens, the respective positive expression rates were 26.7% (4/15) and 72.2% (13/18) for mTOR and 73.3% (11/15) and 38.9% (7/18) for PTEN. In patients with or without lymph node metastasis, the respective positive expression rates were 81.3% (13/16) and 23.5% (4/17) for mTOR and 18.8% (3/16) and 88.2% (15/17) for PTEN. In stage I+II and stage III+IV, the respective positive expression rates were 16.7% (3/18) and 93.3% (14/15) for mTOR and 94.4% (17/18) and 6.7% (1/15) for PTEN.

Table II.

Correlations between mTOR and PTEN expression and clinicopathological characteristics in gastric carcinoma cases.

Table II.

Correlations between mTOR and PTEN expression and clinicopathological characteristics in gastric carcinoma cases.

mTOR expression
PTEN expression
FactorsPositive (%)Negative (%)P-valuePositive (%)Negative (%)P-value
Gender0.8490.407
  Male8 (53.3)7 (46.7)7 (46.7)8 (53.3)
  Female9 (50.0)9 (50.0)11 (61.1)7 (38.9)
Age (years)0.2210.009
  <5410 (62.5)6 (37.5)5 (31.3)11 (68.8)
  ≥547 (41.2)10 (58.8)13 (76.5)4 (23.5)
Invasive depth0.0850.021
  Early stage1 (16.7)5 (83.3)6 (100.0)0 (0)
  Advanced stage16 (59.3)11 (40.7)12 (44.4)15 (55.6)
Differentiation0.0090.048
  Well and moderate4 (26.7)11 (73.3)11 (73.3)4 (26.7)
  Poor13 (72.2)5 (27.8)7 (38.9)11 (61.1)
Lymph node metastasis0.0010.000
  Positive13 (81.3)3 (18.8)3 (18.8)13 (81.3)
  Negative4 (23.5)13 (76.5)15 (88.2)2 (11.8)
Pathological stage0.0000.000
  I+II3 (16.7)15 (83.3)17 (94.4)1 (5.6)
  III+IV14 (93.3)1 (6.7)1 (6.7)14 (93.3)

[i] mTOR, mammalian target of rapamycin; PTEN, phosphate and tensin homolog.

The expression of mTOR had a significant positive correlation with differentiation, lymph node metastasis and clinical staging (P<0.01), but was independent of gender, age and invasive depth (P>0.05). The expression of PTEN was negatively correlated with invasive depth and differentiation (P<0.05) and significantly negatively correlated with age, lymph node metastasis and clinical pathological staging (P<0.01), but was not associated with gender (P>0.05).

Correlation between mTOR and PTEN in human gastric cancer

When the expression of mTOR was analyzed with regard to PTEN expression, the staining pattern was divided into 4 groups: mTOR/PTEN, mTOR+/PTEN+, mTOR+/PTEN and mTOR/ PTEN+. A comparison of the mTOR/PTEN+ and mTOR+/PTEN groups (Table III) revealed that that in early gastric cancer, the size of the former group was larger than the that of latter and that the differences between the 2 groups were statistically significant with regard to invasive depth (P=0.041). In well- and moderately differentiated gastric cancer, the mTOR/PTEN+ group was larger than the mTOR+/PTEN group and the difference between the 2 groups was statistically significant with regard to differentiation (P=0.012). In patients without lymph node metastasis, the mTOR/PTEN+ group was larger than the mTOR+/PTEN group and the difference between the 2 groups was statistically significant with regard to lymph node metastasis (P=0.000). In stage I+II, the mTOR/PTEN+ group was larger than the mTOR+/PTEN group and the difference between two groups was statistically significant with regard to pathological stage (P=0.000).

Table III.

Co-expression of mTOR and PTEN and clinicopathological characteristics.

Table III.

Co-expression of mTOR and PTEN and clinicopathological characteristics.

Factors mTOR/PTEN+ mTOR+/PTENP-value
Invasive depth
  Early stage500.041
  Advanced stage913
Differentiation
  Well and moderate1030.012
  Poor410
Lymph node metastasis
  Positive2120.000
  Negative121
Pathological stage
  I+II1400.000
  III+IV013

[i] mTOR, mammalian target of rapamycin; PTEN, phosphate and tensin homolog.

Discussion

Previous studies have suggested that the PI3K-AKT-mTOR pathway is frequently activated in various types of cancer and that this pathway is considered to be important for cancer cell survival, proliferation, angiogenesis and resistance to chemotherapy (1921). However, the activated molecule of the PI3K-AKT-mTOR pathway in gastric cancer has not yet been studied. In the present study, 33 cases of gastric cancer were investigated and statistical analyses were performed concerning the correlation between the clinicopathological parameters in gastric cancer and the immunohistochemical expression levels of mTOR and PTEN. The results indicated that mTOR and PTEN were negatively correlated in the pathogenesis of gastric cancer. The overexpression of mTOR and low expression of PTEN proteins were strongly correlated with the pathological staging. These results suggested that mTOR and PTEN may be clinically useful prognostic markers and may provide additional information for the histological diagnosis and pathological staging of gastric cancer.

Possible correlations between mTOR expression in gastric cancer and pathological parameters were investigated. The present study demonstrated that mTOR was activated in human gastric cancer and was significantly correlated with invasive depth, differentiation and lymph node metastasis, suggesting that the high expression of mTOR contributes to the progression and metastasis of gastric cancer. Positive mTOR expression was detected in tumor cells in 51.5% (17/33) of gastric cancer patients, while little or no expression was observed in normal gastric tissues. Furthermore, 93.3% (14/15) of gastric cancer patients had positive expression of mTOR at stage III+IV suggesting that the hyperactivation of mTOR kinase was a late event in the development of gastric cancer. Similarly, the positive expression rate of mTOR was high for those patients with lymph node metastasis (81.3%, 13/16). mTOR immunoreactivity intensity data revealed that the high expression levels of mTOR significantly increased with the tumor progression.

It has been reported that mTOR is a powerful oncoprotein overexpressed in numerous types of cancer, including hepatocellular carcinoma (22), lung cancer (23), esophageal squamous cell carcinoma (24) and breast cancer (25,26). Furthermore, patients with breast cancer and mTOR overexpression had a risk of recurrence 3 times greater than that of patients without mTOR overexpression (25,27). The mTOR inhibitor everolimus has demonstrated promising clinical efficacy in a phase III randomized and double-blind trial in patients with metastatic renal cell cancer (28). The exact mechanisms for the overexpression of mTOR in carcinoma remain unclear. As a highly conserved, ubiquitously expressed signaling molecule, mTOR is activated downstream of multiple distinct growth factor receptors and is crucial for mediating cell proliferation and survival (29). Several mTOR inhibitors, including rapamycin/sirolimus (Wyeth) and derivatives, such as RAD001/everolimus (Novartis), CCI-779/temsirolimus (Wyeth) and AP23573 (Ariad), are being developed as anti-cancer agents against various types of malignancies (30).

By contrast, PTEN was shown by immunohistochemistry to be expressed in normal gastric tissues and almost all the early gastric cancer cases. The underexpression of PTEN was significantly correlated with invasive depth, differentiation and lymph node metastasis. Approximately one-half of the gastric cancer patients exhibited a loss of PTEN expression. Additionally, the loss of PTEN expression was correlated with invasive depth and differentiation (P<0.05) and closely correlated with lymph node metastasis and clinical pathological staging (P<0.01). PTEN immunoreactivity intensity data were also analyzed and the results revealed that the high expression levels of PTEN significantly decreased with the tumor progression.

PTEN, which is located at human chromosome 10q23, has been identified as a tumor suppressor gene and an important negative regulator of the PI3K-AKT-mTOR signaling pathway that promotes cell proliferation and inhibits apoptosis (31). Inactivation of PTEN by mutation, deletion and promoter hypermethylation has been demonstrated in a range of cancer types, including lung, breast, prostate and esophageal carcinomas (3135). Langlois et al observed that PTEN controls the cellular polarity, establishment of cell-cell junctions, paracellular permeability, migration and tumorigenic potential of human colorectal cancer cells (36). Varied analysis of colorectal carcinomas suggested that the patients without PTEN expression had shorter survival times than the patients with PTEN expression (P=0.003) (37). Abnormal expression of PTEN may predict the metastasis and prognosis of gastric cancer (21,38,39).

We concluded that mTOR facilitated the development of gastric cancer while PTEN, a tumor suppressor gene, was able to inhibit tumor invasion and metastasis. mTOR and PTEN co-regulate the progression of tumors and participate in proliferation, invasion and metastasis in gastric cancer. Bakarakos et al discovered that the loss of PTEN and activation of mTOR was closely correlated with breast cancer (40). In vitro studies suggested that PTEN is capable of inhibiting cell proliferation and promoting apoptosis via inhibition of the activity of the PI3K-Akt-mTOR pathway (41). The combined deletion of PTEN and Lkb1 in the mouse bladder significantly activated the mTOR pathway and increased bladder epithelial cell proliferation and tumorigenesis (42). When we compared the mTOR-/PTEN+ and mTOR+/PTEN groups, the differences between them were statistically significant with regard to invasive depth, histological type, lymph node metastasis and pathological stage. Consequently, collaborative detection of mTOR and PTEN expression may be more useful in the diagnosis of gastric cancer.

In summary, upregulated expression of mTOR and downregulated expression of PTEN were involved in carcinogenesis and progression of gastric cancer. A negative correlation between mTOR and PTEN expression implied that their modified expression may be important in the pathogenesis, invasion and metastasis of carcinoma tissue. Combined detection of mTOR and PTEN expression may be used to evaluate the degree of malignancy in gastric cancer, which may be a useful marker for the early diagnosis of gastric cancer. Further studies with more patients, including follow-up and different molecular biomarkers in addition to these two molecules, would aid the clarification of the disease pathogenesis and identification of potential therapeutic approaches.

Acknowledgements

The author Min Li gratefully acknowledges the assistance of his elder sister Li Li for her critical reading of the manuscript before its submission. We also gratefully acknowledge the assistance of Xinyu Qin, Huawen Sun and Lujun Song in the preparation of this study.

References

1. 

K Washington7th edition of the AJCC cancer staging manual: stomachAnn Surg Oncol1730773079201010.1245/s10434-010-1362-z20882416

2. 

S LiangL HeX ZhaoMicroRNA let-7f inhibits tumor invasion and metastasis by targeting myh9 in human gastric cancerPloS One6e18409201110.1371/journal.pone.001840921533124

3. 

DH RoukosTargeting gastric cancer with trastuzumab: new clinical practice and innovative developments to overcome resistanceAnn Surg Oncol171417201010.1245/s10434-009-0766-019841980

4. 

N BalNE KocerME ErtorerET CanpolatF KayaselcukMaspin, E-selectin, and P-selectin expressions in papillary thyroid carcinomas and their correlation with prognostic parametersPathol Res Pract204743750200810.1016/j.prp.2008.04.01618597952

5. 

X ChenJ LiaoY LuX DuanW SunActivation of the PI3K Akt pathway mediates bone morphogenetic protein 2-induced invasion of pancreatic cancer cells Panc-1Pathol Oncol Res17257261201110.1007/s12253-010-9307-120848249

6. 

Z XuY ZhangJ JiangEpidermal growth factor induces HCCR expression via PI3K/Akt/mTOR signaling in PANC-1 pancreatic cancer cellsBMC Cancer10161201010.1186/1471-2407-10-16120423485

7. 

AP BhattPM BhendeSH SinD RoyDP DittmerB DamaniaDual inhibition of PI3K and mTOR inhibits autocrine and paracrine proliferative loops in PI3K/Akt/mTOR-addicted lymphomasBlood11544554463201010.1182/blood-2009-10-25108220299510

8. 

M SajiMD RingelThe PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumorsMol Cell Endocrinol3212028201010.1016/j.mce.2009.10.01619897009

9. 

S VignotS FaivreD AguirreE RaymondmTOR-targeted therapy of cancer with rapamycin derivativesAnn Oncol16525537200510.1093/annonc/mdi11315728109

10. 

D MahalingamK SankhalaA MitaFJ GilesMM MitaTargeting the mTOR pathway using deforolimus in cancer therapyFuture Oncol5291303200910.2217/fon.09.919374536

11. 

CB ChingDE HanselExpanding therapeutic targets in bladder cancer: the PI3K/Akt/mTOR pathwayLab Invest9014061414201010.1038/labinvest.2010.13320661228

12. 

JS CarewKR KellyST NawrockiMechanisms of mTOR inhibitor resistance in cancer therapyTarget Oncol61727201110.1007/s11523-011-0167-821547705

13. 

A YoshimiS GoyamaN Watanabe-OkochiEvi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteinsBlood11736173628201110.1182/blood-2009-12-26160221289308

14. 

M MireutaA DarnelM PollakIGFBP-2 expression in MCF-7 cells is regulated by the PI3K/AKT/mTOR pathway through Sp1-induced increase in transcriptionGrowth Factors28243255201010.3109/0897719100374547220370577

15. 

C LiuJL WuK XuNeuroprotection by baicalein in ischemic brain injury involves PTEN/AKT pathwayJ Neurochem11215001512201010.1111/j.1471-4159.2009.06561.x20050973

16. 

SR HamiltonLA AaltonenPathology and Genetics of Tumors of the Digestive SystemWHO PressGeneva2000

17. 

SB EdgeDR ByrdCC ComptonAG FritzF GreeneA TrottiAJCC cancer staging manual7th editionSpringerNew York2010

18. 

X HeQ WeiX ZhangImmunohistochemical expression of CXCR4 in thyroid carcinomas and thyroid benign lesionsPathol Res Pract206712715201010.1016/j.prp.2010.05.00320646838

19. 

HY NiuJH WangH LiP HeRapamycin potentiates cytotoxicity by docetaxel possibly through downregulation of Survivin in lung cancer cellsJ Exp Clin Canc Res3028201110.1186/1756-9966-30-2821392382

20. 

M ChenJ GuGL DelclosGenetic variations of the PI3KAKT-mTOR pathway and clinical outcome in muscle invasive and metastatic bladder cancer patientsCarcinogenesis3113871391201010.1093/carcin/bgq11020530239

21. 

M LiL SongX QinGlycan changes: cancer metastasis and anti-cancer vaccinesJ Biosci35665673201010.1007/s12038-010-0073-821289447

22. 

F SahinR KannangaiO AdegbolaJZ WangG SuM TorbensonmTOR and P70 S6 kinase expression in primary liver neoplasmsClin Cancer Res1084218425200410.1158/1078-0432.CCR-04-094115623621

23. 

K SchmidZ Bago-HorvathW BergerDual inhibition of EGFR and mTOR pathways in small cell lung cancerBr J Cancer103622628201010.1038/sj.bjc.660576120683448

24. 

K HirashimaY BabaM WatanabePhosphorylated mTOR expression is associated with poor prognosis for patients with esophageal squamous cell carcinomaAnn Surg Oncol1724862493201010.1245/s10434-010-1040-120339946

25. 

G YuJ WangY ChenOverexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of Chinese patients with gastric cancerClin Cancer Res1518211829200910.1158/1078-0432.CCR-08-213819223493

26. 

J AnH JeongY LeeSU WooJH SeoA KimPhosphorylated Akt and phosphorylated mTOR expression in breast invasive carcinomas: analysis of 530 casesJ Breast Cancer13337348201010.4048/jbc.2010.13.4.337

27. 

S BoseS ChandranJM MirochaN BoseThe Akt pathway in human breast cancer: a tissue-array-based analysisMod Pathol19238245200610.1038/modpathol.3800525

28. 

RJ MotzerB EscudierS OudardEfficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trialLancet372449456200810.1016/S0140-6736(08)61039-918653228

29. 

M MarinovA ZiogasOE PardoAKT/mTOR pathway activation and BCL-2 family proteins modulate the sensitivity of human small cell lung cancer cells to RAD001Clin Cancer Res1512771287200910.1158/1078-0432.CCR-08-216619228731

30. 

KH TamZF YangCK LauCT LamRW PangRT PoonInhibition of mTOR enhances chemosensitivity in hepatocellular carcinomaCancer Lett273201209200910.1016/j.canlet.2008.08.01818824293

31. 

KS JangYS SongSH JangClinicopathological significance of nuclear PTEN expression in colorectal adenocarcinomaHistopathology56229239201010.1111/j.1365-2559.2009.03468.x20102402

32. 

T AndjelkovicJ BankovicJ StojsicCoalterations of p53 and PTEN tumor suppressor genes in non-small cell lung carcinoma patientsTransl Res1571928201110.1016/j.trsl.2010.09.00421146147

33. 

AM Gonzalez-AnguloJ Ferrer-LozanoK Stemke-HalePI3K pathway mutations and PTEN levels in primary and metastatic breast cancerMol Cancer Ther1010931101201110.1158/1535-7163.MCT-10-108921490305

34. 

DJ MulhollandLM TranY LiCell autonomous role of PTEN in regulating castration-resistant prostate cancer growthCancer Cell19792804201110.1016/j.ccr.2011.05.00621620777

35. 

G HouZ LuM LiuH LiuL XueMutational analysis of the PTEN gene and its effects in esophageal squamous cell carcinomaDig Dis Sci5613151322201110.1007/s10620-010-1474-021116717

36. 

MJ LangloisS BergeronG BernatchezThe PTEN phosphatase controls intestinal epithelial cell polarity and barrier function: role in colorectal cancer progressionPloS One5e15742201010.1371/journal.pone.001574221203412

37. 

XH LiHC ZhengH TakahashiS MasudaXH YangY TakanoPTEN expression and mutation in colorectal carcinomasOncol Rep22757764200919724853

38. 

CY GuoXF XuJY WuSF LiuPCR-SSCP-DNA sequencing method in detecting PTEN gene mutation and its significance in human gastric cancerWorld J Gastroenterol1438043811200810.3748/wjg.14.380418609703

39. 

M LiL SongX GaoW ChangX QinToll-like receptor 4 on islet β cells senses expression changes in high-mobility group box 1 and contributes to the initiation of type 1 diabetesExp Mol Med442602672012

40. 

P BakarakosI TheohariA NomikosImmunohistochemical study of PTEN and phosphorylated mTOR proteins in familial and sporadic invasive breast carcinomasHistopathology56876882201010.1111/j.1365-2559.2010.03570.x20636791

41. 

ZY ChengXL GuoXY YangPTEN and rapamycin inhibiting the growth of K562 cells through regulating mTOR signaling pathwayJ Exp Clin Cancer Res2787200810.1186/1756-9966-27-8719115995

42. 

BY ShorningD GriffithsAR ClarkeLkb1 and Pten synergise to suppress mTOR-mediated tumorigenesis and epithelial-mesenchymal transition in the mouse bladderPloS One6e16209201110.1371/journal.pone.001620921283818

Related Articles

Journal Cover

December 2012
Volume 4 Issue 6

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
Li M, Sun H, Song L, Gao X, Chang W and Qin X: Immunohistochemical expression of mTOR negatively correlates with PTEN expression in gastric carcinoma. Oncol Lett 4: 1213-1218, 2012
APA
Li, M., Sun, H., Song, L., Gao, X., Chang, W., & Qin, X. (2012). Immunohistochemical expression of mTOR negatively correlates with PTEN expression in gastric carcinoma. Oncology Letters, 4, 1213-1218. https://doi.org/10.3892/ol.2012.930
MLA
Li, M., Sun, H., Song, L., Gao, X., Chang, W., Qin, X."Immunohistochemical expression of mTOR negatively correlates with PTEN expression in gastric carcinoma". Oncology Letters 4.6 (2012): 1213-1218.
Chicago
Li, M., Sun, H., Song, L., Gao, X., Chang, W., Qin, X."Immunohistochemical expression of mTOR negatively correlates with PTEN expression in gastric carcinoma". Oncology Letters 4, no. 6 (2012): 1213-1218. https://doi.org/10.3892/ol.2012.930