Significance of GSTP1 for predicting the prognosis and chemotherapeutic efficacy in esophageal squamous cell carcinoma

  • Authors:
    • Yusuke Yamamoto
    • Hirotaka Konishi
    • Daisuke Ichikawa
    • Tomohiro Arita
    • Katsutoshi Shoda
    • Shuhei Komatsu
    • Atsushi Shiozaki
    • Hisashi Ikoma
    • Hitoshi Fujiwara
    • Kazuma Okamoto
    • Toshiya Ochiai
    • Jun Inoue
    • Johji Inazawa
    • Eigo Otsuji
  • View Affiliations

  • Published online on: July 9, 2013     https://doi.org/10.3892/or.2013.2606
  • Pages: 1687-1694
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glutathione S-transferases (GSTs) have been reported to be activated in several types of cancers, including esophageal squamous cell carcinoma (ESCC). The aim of the present study was to investigate whether GSTP1 protein expression is a useful predictor of the clinical outcome or drug resistance in ESCC. Immunohistochemistry was conducted with 75 ESCC resected specimens using a monoclonal antibody against GSTP1. The patients were divided into two groups according to the degree of GSTP1 staining, and the relationship between the GSTP1 level and the clinicopathological features was examined. Seventy-five patients were divided into low (grade 1, n=36) and high (grade 2, n=39) GSTP1 expression groups. The overall survival was significantly worse in the grade 2 patients than in the grade 1 patients (5‑year survival rate, 78.5 vs. 51.2%; p=0.027). Cox proportional hazard analysis revealed that macroscopic type 3 or 4 disease (p=0.001), lymph node metastasis (p=0.010), and high GSTP1 expression (p=0.029) were independent predictors of a poor prognosis. With regard to the subgroup analysis among the 31 patients undergoing adjuvant chemotherapy, the grade 2 patients had a worse prognosis than did the grade 1 patients (5‑year survival rate, 45.0 vs. 81.8%; p=0.081). This tendency was not observed in the subgroup without adjuvant chemotherapy (5‑year survival rate, 51.7 vs. 59.9%; p=0.979). In conclusion, the GSTP1 expression is a good predictor of prognosis, and it may be closely related to the chemotherapeutic efficacy of 5-FU plus cisplatin in ESCC patients.

Introduction

Esophageal carcinoma is the sixth most common cause of cancer-related death among adult malignant tumors, and it is considered to possess a relatively high malignant potential (15). Squamous cell carcinoma is the predominant cellular type of esophageal cancer worldwide, although the incidence of esophageal adenocarcinoma has been increasing in Western countries (6). More recent advances in surgical techniques, chemotherapy and radiotherapy have resulted in an improvement in the prognosis of esophageal squamous cell carcinoma (ESCC) (7); however, the long-term outcomes remain unacceptable. It is important to detect more useful predictors of the prognosis and efficacy of chemotherapy for the further improvement of the prognosis of ESCC patients.

Many genetic and epigenetic alterations have been reported in ESCC. In the present study, we focused on the expression of glutathione S-transferase (GST) P1 protein, which is the main GST expressed in the human esophagus. The GST family consists of a group of important phase II drug-metabolizing enzymes that is instrumental in providing cell protection or detoxification of toxic substances and anticancer drugs through conjugation with glutathione (GSH) (8). GSTP1 is widely expressed in normal tissues, while the enzyme activity has been shown to be highly overexpressed in several types of cancers, for example, blood, head and neck, lung, colorectal, esophagus and breast cancers (913). The high expression level of GSTP1 has been reported to be regulated by inflammation, various chemical carcinogens, the expression of Nrf2 and other factors. Thus, overexpression of GSTP1 may influence the clinical outcome or the resistance to anticancer drugs. However, the available information on the association between the degree of GSTP1 expression and the prognosis of ESCC is conflicting (14).

In the present study, we simply evaluated the immunohistochemical expression of GSTP1 in resected ESCC specimens and investigated whether GSTP1 expression is associated with the clinical outcome and the response to adjuvant chemotherapy.

Patients and methods

Study population

We selected 75 patients who underwent macroscopically curative right-transthoracic esophagectomy with extensive lymph node dissection for ESCC between January 2000 and December 2009 at the Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan. Of the 75 patients, 14 (19%) patients underwent three-field lymph node dissection (cervical, mediastinal and abdominal nodes), while 61 (81%) patients underwent two-field lymph node dissection (mediastinal and abdominal nodes) under pre- or intra-operative diagnosis (15). Thirty-one of the 75 patients (41%) received post-operative adjuvant therapy, high-dose FP or low-dose FP plus oral fluoropyrimidine [5-FU (150–200 mg/body/day) or UFT (300–400 mg/body/day)] (16) and 4 patients were treated with pre-operative chemotherapy, high-dose FP [5-FU (800 mg/m2/day, days 1–5) plus cisplatin (80 mg/m2/day, day 1)], followed by planned esophagectomy. We excluded the patients who underwent neoadjuvant chemo-radiation therapy. Staging was based principally on the International Union Against Cancer (UICC)/TNM Classification of Malignant Tumors, 7th edition (17). The macroscopic type was classified as follows: type 0, macroscopic early cancer; type 1, polypoid tumors; type 2, ulcerated carcinomas with sharply demarcated and raised margins; type 3, ulcerated carcinomas without definite limits, infiltration into the surrounding wall; type 4, diffusely infiltrating carcinomas in which ulceration is usually not a marked feature.

Postsurgical management

Following discharge from the hospital, the patients were regularly monitored in the outpatient clinic at intervals of 3 months for the first 2 years and at intervals of 6 months thereafter. Multi-slice computed tomography of the neck, chest and upper abdomen was performed every 6 months. Since 2007, positron emission tomography with computed tomography has been used in screening for recurrence (18,19).

All recurrent tumors were evaluated using imaging studies. Of the 32 patients with recurrent tumors, 18 received intensive treatment: 10 received chemoradiotherapy, 1 received lymphadenectomy, 8 received chemotherapy; and 14 received the best supportive care.

Cell lines and western blotting

Each cell line was purchased from the Cell Resource Center of Tohoku University, Riken Bio Resource Center, or the American Type Culture Collection. Whole-cell lysates were prepared in SDS sample buffer, resolved by SDS-PAGE and transferred to PVDF membranes. After blocking with TBS containing 0.05% Tween-20 and 5% non-fat dry milk for 1 h, each membrane was incubated with an antibody overnight. The primary antibodies and dilutions were anti-GSTP1 (1/500, HPA019779; Sigma-Aldrich) and anti-GAPDH (1/4,000, sc-25778; Santa Cruz Biotechnology, Inc.) antibodies. The membrane was washed and exposed to horseradish peroxidase-conjugated secondary antibody (at 1/5,000, #7074S; Cell Signaling Technology). The bound antibodies were visualized with an HRP staining solution or with an ECL Western Detection kit on an Image Quant LAS 500 (GE Healthcare) according to the manufacturer’s instructions (Cell Signaling Technology).

Immunohistochemistry

Tumor samples were fixed with 10% formaldehyde in PBS, embedded in paraffin, and sectioned into 5-μm slices. After deparaffinization by xylene and rehydration with ethanol, antigen retrieval was performed by boiling in Dako REAL Red Target Retrieval Solution (pH 6.0) at 95°C for 30 min and the sections were treated with 0.3% hydrogen peroxide in methanol at room temperature for 20 min to inactivate the endogenous peroxidase. After treatment with Block Ace (Vectastain Elite ABC universal kit; Vector Laboratories, Inc., Burlingame, CA, USA) for 30 min at room temperature, the sections were incubated at 4°C overnight with a primary antibody against GSTP1 (1:500 dilution). The avidin-biotin-peroxidase complex system (Vectastain Elite ABC universal kit) was used for secondary antibody detection and color development with diaminobenzidine tetrahydrochloride at room temperature for 30 min. The slides were lightly counterstained with hematoxylin.

The patients were divided into two groups, grade 1 and 2, according to the degree of GSTP1 expression. Tissues of grade 2 were defined as those with even staining of at least 90% of the cancer area (Fig. 1B–a and –b) and tissues of grade 1 were defined as areas with spotty staining corresponding to <90% of the cancer area (Fig. 1B–c and –d).

Inter-observer reproducibility for identifying the immunohistological characteristics

The reproducibility of the grading classification was tested by asking another independent observer (K.H.) to blindly review all of the examples. This observer was not provided with any clinical information regarding the outcomes of the patients. The reproducibility was tested by obtaining the κ-scores according to the widely used statistical chart that grades the strength of agreement to six categories [poor (κ-score, <0.00), slight (0.00–0.20), fair (0.21–0.40), moderate (0.41–0.60), substantial (0.61–0.80) and almost perfect (0.81–1.00)] (20).

Statistical analysis

We performed univariate analyses of the 15 clinical and pathologic factors that were potentially associated with overall survival. Survival was calculated using the Kaplan Meier method and compared between groups using the log-rank test. A multivariate analysis using the Cox hazards model was performed to identify independent predictors of survival.

The relationship of GSTP1 expression and 10 pathological factors was compared using the Chi-squared test with Yates correction. All significant factors determined by the univariate analysis were entered into a multivariate regression analysis to identify independent factors. These tests were one-tailed and a p<0.05 was considered to be statistically significant. All statistical analyses were performed using the SPSS for Windows 11.5 software program (SPSS, Chicago, IL, USA).

Results

GSTP1 protein expression in the ESCC cell lines

We performed a western blot analysis with an antibody against GSTP1 in 6 ESCC cell lines (TE2, TE5, TE9, TE13, KYSE70 and KYSE170) and a normal fibroblast cell line, WI-38. The GSTP1 protein expression level was upregulated in all of the ESCC cell lines compared with the level in the WI-38 cells (Fig. 1A).

Overall survival and clinicopathological features according to GSTP1 grade

Thirty-six patients had tumors that showed a low GSTP1 protein expression level defined as grade 1 (Fig. 1B-a and -b), whereas 39 had tumors that showed a high GSTP1 protein expression level defined as grade 2 (Fig. 1B-c and -d). The κ-value between the classifications of the two reviewers was 0.893 (almost perfect). The overall 5-year survival rate of all patients was 63.5% and Fig. 2A showed the results of a survival analysis according to the GSTP1 grade. The 5-year survival rate was 78.5% for grade 1 patients and 51.2% for grade 2 patients. There were significant differences in the survival rates between both groups (p=0.027).

Table I summarizes the results of the univariate and multivariate analyses of the prognostic factors associated with overall survival. Macroscopic type 3 or 4 disease (p=0.001), lymph node metastasis (p=0.010) and grade 2 GSTP1 expression (p=0.029) were independently associated with a poor prognosis.

Table I

Results of the univariate and multivariate analyses of the prognostic factors associated with overall survival.

Table I

Results of the univariate and multivariate analyses of the prognostic factors associated with overall survival.

Univariate analysisMultivariate analysis


n5-year OS (%)p-valueHR (95% CI)p-value
Gender0.715
 Male6462.7
 Female1168.6
Age, years0.094
 <655056.8
 ≥652577.4
Curability0.013
 Curative resection5470.0
 Non-curative resection2145.4
Main tumor location0.048
 Lower thoracic esophagus2648.9
 Middle or upper thoracic esophagus4971.9
Neoadjuvant chemotherapy0.241
 Absent7165.4
 Present440.0
Macroscopic type0.0010.001
 Type 0–26470.21
 Type 3 and 41127.34.200 (1.751–10.071)
Predominant differentiation0.358
 Well or moderately differentiated5269.3
 Poorly differentiated or others2351.7
Depth of tumor invasion (pT)0.094
 pT1 or pT24468.4
 pT3 or pT43155.7
Tumor size (mm)0.022
 <403974.5
 ≥403651.4
Lymphatic invasion0.007
 Absent3477.9
 Present4151.8
Venous invasion0.019
 Absent4276.3
 Present3347.5
Infiltrative growth pattern0.181
 Expansive pattern1678.6
 Intermediate or infiltrative pattern5859.2
Intramural metastasis0.336
 Absent7164.2
 Present450.0
Lymph node metastasis0.0060.010
 Absent3479.61
 Present4150.03.396 (1.335–8.637)
GSTP10.0290.029
 Grade 13678.51
 Grade 23951.22.704 (1.107–6.604)

[i] OS, overall survival; HR, hazard ratio; CI, confidence interval; GSTP1, glutathione S-transferase P1.

The 10 clinicopathological factors related to the GSTP1 grade in tumor specimens were analyzed (Table II, the Chi-squared test and the logistic regression model). A tumor size >40 mm (p=0.007) and venous invasion (p=0.011) were identified as independent factors associated with grade 2 GSTP1 expression.

Table II

Results of the univariate and multivariate analyses of the 10 clinicopathological factors as related to the GSTP1 expression in the tumor specimens.

Table II

Results of the univariate and multivariate analyses of the 10 clinicopathological factors as related to the GSTP1 expression in the tumor specimens.

nGrade 1 (n=36)Grade 2 (n=39)Univariate analysisMultivariate analysis


p-valueRR (95% CI)p-value
Gender0.855
 Male643133
 Female1156
Age, years0.327
 <65502228
 ≥65251411
Main tumor location0.091
 Lower thoracic esophagus26917
 Middle or upper thoracic esophagus492722
Macroscopic type0.403
 Type 0–2643232
 Type 3 and 41147
Tumor size (mm)0.0150.007
 <403924151
 ≥403612244.193 (1.478–11.897)
Lymphatic invasion0.030
 Absent342113
 Present411526
Venous invasion0.0240.011
 Absent4225171
 Present3311223.918 (1.367–11.230)
Infiltrative growth pattern0.211
 Expansive pattern16106
 Intermediate or infiltrative pattern582632
Intramural metastasis0.344
 Absent713536
 Present413
Lymph node metastasis0.088
 Absent342014
 Present411625

[i] GSTP1, glutathione S-transferase P1; RR, relative risk; CI, confidence interval.

Relationship between the GSTP1 expression level and the efficacy of adjuvant chemotherapy

With regard to the subgroup analysis among the 44 patients who did not undergo adjuvant chemotherapy, there were no significant differences in survival between the grade 1 and grade 2 groups (5-year rate, 51.7 vs. 59.9%, p=0.979) (Fig. 2B). On the other hand, among the 31 patients undergoing adjuvant chemotherapy, the grade 1 group had a better prognosis than the grade 2 group (5-year rate, 81.8 vs. 45.0%). There were, however, no significant differences between the groups (p=0.081) (Fig. 2C).

Furthermore, a subgroup analysis of progression-free survival among the pStage II, III, or IV patients according to the GSTP1 grade was performed. In the grade 1 group, the patients who underwent adjuvant chemotherapy were shown to have a better prognosis than those who did not undergo adjuvant chemotherapy (p=0.067) (Fig. 3A), whereas no difference was observed in the grade 2 group (p=0.874) (Fig. 3B).

Discussion

GST expression has been widely detected in various organs and tumors, such as prostate cancer, colorectal cancer, breast cancer, gastric cancer and esophageal cancer (21,22). GST enzymes catalyze the conjugation of toxic and carcinogenic electrophilic molecules to GSH and their activities constitute an important cellular protection mechanism for many types of damage, such as those that involve the activation of phase II detoxification enzymes (11,23). In mammals, 7 classes of GSTs have been identified on the basis of amino acid sequence similarities, substrate specificity and immunological cross reactivity, such as GSTA, GSTM and GSTP (23). In the normal esophageal epithelium, several GST variants are expressed and GSTP1 is the main one (23).

For many types of cancers, including ESCC, the role of the GST protein has been discussed in the context of the variants of GST and the polymorphisms of each variant. In ESCC, many authors have reported several genotypes of GSTP1, such as lle 105 Val and Ala 114 Val (13,2224). However, these differences among the several variants or genotypes are conflicting between reports. In the present study, we simply assessed the relationship between GSTP1 protein expression and various clinicopathological parameters in ESCC (Fig. 1; Table II) and we found that high levels of GSTP1 expression, which were significantly associated with severe venous invasion and larger tumor size, led to poor prognosis (Fig. 2A; Table I). Moreover, the reproducibility of our classification of GSTP1 expression was found to be ‘almost perfect’ (κ-value, 0.893) when all slides were assessed by another independent observer. These findings indicate that our classification of GSTP1 expression is a simple and reproducible predictor of the prognosis of ESCC.

There are two important reasons that we focused on GSTP1 expression in ESCC. The first is that GSTP1 has two tumor-protective roles involving the deactivation of anticancer reagents and the inhibition of signaling pathways that lead to apoptosis through the inhibition of the c-Jun N-terminal kinase (JNK). The deactivation of anticancer reagents by GST expression has been reported in many types of cancers, including ESCC, and it has been considered one of the main reasons for the acquisition of resistance to chemotherapeutic agents (25,26). Furthermore, several GSTs have been shown to be associated with members of the mitogen-activated protein kinase (MAPK) pathway, such as JNK and p38, which is involved in cell survival and cell death signaling (23,27,28). Based on these two major roles, we aimed to ascertain whether GSTP1 also affects cancer prognosis in ESCC.

The second reason is that GSTP1 expression is partially regulated by the Nrf2 gene, which is a transcriptional factor that is frequently upregulated in lung cancer, esophageal cancer and several other squamous cell carcinomas (29,30). In ESCC cell lines and primary samples, mutations in Nrf2 are frequently reported (31). Therefore, we considered whether the high expression of GSTP1 may be partially affected by the activation of Nrf2.

In our study, the majority of patients treated by adjuvant chemotherapy with a low GSTP1 protein expression level showed better prognosis than the patients with a high GSTP1 expression level. This tendency was not shown in the patients with high GSTP1 protein expression level (Fig. 3). These results may indicate the role of GSTP1 for the detoxification of anticancer drugs in ESCC.

Since the publication of the results of the JCOG 9907 study, neoadjuvant chemotherapy followed by radical esophagectomy has been accepted as the standard therapeutic approach for resectable cStage II/III esophageal cancer in Japan (32). However, the clinical response rate of preoperative chemotherapy is only 38%, which is not sufficient. Therefore, it seems to be quite important to develop predictive factors for the response to chemotherapy for patients who are unlikely to derive benefits from such therapy.

Retrospective analysis in a relatively small case series was a limitation of the present study, and a conflicting result regarding the utility of GSTP1 expression has also been reported (14). Therefore, our results need to be confirmed through additional studies of a large number of patients, and the clinical significance of GSTP1 in the prediction of the response to chemotherapy must be established in ESCC. If the relationship between the GSTP1 expression level and efficacy of chemotherapy is clinically confirmed, then the expression level may also have the potential to be used as a preoperative marker for the effect of neoadjuvant chemotherapy.

In conclusion, we showed that high GSTP1 protein expression in ESCC tissue is a sensitive marker of poor prognosis, and the resistance to adjuvant chemotherapy may be influenced by high GSTP1 expression. These findings indicate that classification of GSTP1 expression may be used as a simple, accurate, and reproducible predictor of prognosis and drug resistance for ESCC patients.

References

1 

Kamangar F, Dores GM and Anderson WF: Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol. 24:2137–2150. 2006. View Article : Google Scholar : PubMed/NCBI

2 

De Vita F, Di Martino N, Orditura M, Cosenza A, Galizia G, Del Genio A and Catalano G: Preoperative chemoradiotherapy for squamous cell carcinoma and adenocarcinoma of the esophagus: a phase II study. Chest. 122:1302–1308. 2002.PubMed/NCBI

3 

Hofstetter W, Swisher SG, Correa AM, Hess K, Putnam JB Jr, Ajani JA, Dolormente M, Francisco R, Komaki RR, Lara A, Martin F, Rice DC, Sarabia AJ, Smythe WR, Vaporciyan AA, Walsh GL and Roth JA: Treatment outcomes of resected esophageal cancer. Ann Surg. 236:376–384. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Medical Research Council Oesophageal Cancer Working Group. Surgical resection with or without preoperative chemotherapy in oesophageal cancer: a randomised controlled trial. Lancet. 359:1727–1733. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Cooper JS, Guo MD, Herskovic A, Macdonald JS, Martenson JA Jr, Al-Sarraf M, Byhardt R, Russell AH, Beitler JJ, Spencer S, Asbell SO, Graham MV and Leichman LL: Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85–01). Radiation Therapy Oncology Group. JAMA. 281:1623–1627. 1999.PubMed/NCBI

6 

McKinney A, Sharp L, Macfarlane GJ and Muir CS: Oesophageal and gastric cancer in Scotland 1960–90. Br J Cancer. 71:411–415. 1995.

7 

Ando N, Ozawa S, Kitagawa Y, Shinozawa Y and Kitajima M: Improvement in the results of surgical treatment of advanced squamous esophageal carcinoma during 15 consecutive years. Ann Surg. 232:225–232. 2000.PubMed/NCBI

8 

Coles BF and Kadlubar FF: Detoxification of electrophilic compounds by glutathione S-transferase catalysis: determinants of individual response to chemical carcinogens and chemotherapeutic drugs? Biofactors. 17:115–130. 2003. View Article : Google Scholar

9 

Matthias C, Jahnke V, Fryer AA and Strange RC: Influence of glutathione s-transferase and cytochrome p450 polymorphisms on prognosis of head and neck cancer. Laryngorhinootologie. 81:406–412. 2002.(In German).

10 

Anderer G, Schrappe M, Brechlin AM, Lauten M, Muti P, Welte K and Stanulla M: Polymorphisms within glutathione S-transferase genes and initial response to glucocorticoids in childhood acute lymphoblastic leukaemia. Pharmacogenetics. 10:715–726. 2000. View Article : Google Scholar

11 

Stoehlmacher J, Park DJ, Zhang W, Groshen S, Tsao-Wei DD, Yu MC and Lenz HJ: Association between glutathione S-transferase P1, T1, and M1 genetic polymorphism and survival of patients with metastatic colorectal cancer. J Natl Cancer Inst. 94:936–942. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Sweeney C, Ambrosone CB, Joseph L, Stone A, Hutchins LF, Kadlubar FF and Coles BF: Association between a glutathione S-transferase A1 promoter polymorphism and survival after breast cancer treatment. Int J Cancer. 103:810–814. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Lee JM, Wu MT, Lee YC, Yang SY, Chen JS, Hsu HH, Huang PM, Kuo SW, Lee CJ and Chen CJ: Association of GSTP1 polymorphism and survival for esophageal cancer. Clin Cancer Res. 11:4749–4753. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Wang Z, He W, Yang G, Wang J, Wang Z, Nesland JM, Holm R and Suo Z: Decreased expression of GST pi is correlated with a poor prognosis in human esophageal squamous carcinoma. BMC Cancer. 10:3522010. View Article : Google Scholar : PubMed/NCBI

15 

Ueda Y, Shiozaki A, Itoi H, Okamoto K, Fujiwara H, Ichikawa D, Kikuchi S, Fuji N, Itoh T, Ochiai T, Komatsu S and Yamagishi H: Intraoperative pathological investigation of recurrent nerve nodal metastasis can guide the decision whether to perform cervical lymph node dissection in thoracic esophageal cancer. Oncol Rep. 16:1061–1066. 2006.

16 

Shiozaki A, Yamagishi H, Itoi H, Fujiwara H, Kikuchi S, Okamoto K, Ichikawa D, Fuji N, Ochiai T, Sonoyama T and Ueda Y: Long-term administration of low-dose cisplatin plus 5-fluorouracil prolongs the postoperative survival of patients with esophageal cancer. Oncol Rep. 13:667–672. 2005.PubMed/NCBI

17 

Sobin LH, Gospodarowicz MK and Wittekind C: International Union Against Cancer. TNM Classification of Malignant Tumours. 7th edition. Hoboken, NJ: Wiley-Blackwell; pp. 73–77. 2010

18 

Kato H, Miyazaki T, Nakajima M, Fukuchi M, Manda R and Kuwano H: Value of positron emission tomography in the diagnosis of recurrent oesophageal carcinoma. Br J Surg. 91:1004–1009. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Roedl JB, Harisinghani MG, Colen RR, Fischman AJ, Blake MA, Mathisen DJ and Mueller PR: Assessment of treatment response and recurrence in esophageal carcinoma based on tumor length and standardized uptake value on positron emission tomography-computed tomography. Ann Thorac Surg. 86:1131–1138. 2008. View Article : Google Scholar

20 

Fleiss JL: Measuring nominal scale agreement among many rates. Psychol Bull. 76:378–382. 1971. View Article : Google Scholar

21 

Nakajima T, Wang RS, Nimura Y, Pin YM, He M, Vainio H, Murayama N, Aoyama T and Iida F: Expression of cytochrome P450s and glutathione S-transferases in human esophagus with squamous-cell carcinomas. Carcinogenesis. 17:1477–1481. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Laborde E: Glutathione transferases as mediators of signaling pathways involved in cell proliferation and cell death. Cell Death Differ. 17:1373–1380. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Rossini A, Rapozo DC, Soares Lima SC, Guimarães DP, Ferreira MA, Teixeira R, Kruel CD, Barros SG, Andreollo NA, Acatauassú R, Matos HJ, Albano RM and Pinto LF: Polymorphisms of GSTP1 and GSTT1, but not of CYP2A6, CYP2E1 or GSTM1, modify the risk for esophageal cancer in a western population. Carcinogenesis. 28:2537–2542. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Li D, Dandara C and Parker MI: The 341C/T polymorphism in the GSTP1 gene is associated with increased risk of oesophageal cancer. BMC Genet. 11:472010. View Article : Google Scholar : PubMed/NCBI

25 

Peklak-Scott C, Smitherman PK, Townsend AJ and Morrow CS: Role of glutathione S-transferase P1-1 in the cellular detoxification of cisplatin. Mol Cancer Ther. 7:3247–3255. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Miyake T, Nakayama T, Naoi Y, Yamamoto N, Otani Y, Kim SJ, Shimazu K, Shimomura A, Maruyama N, Tamaki Y and Noguchi S: GSTP1 expression predicts poor pathological complete response to neoadjuvant chemotherapy in ER-negative breast cancer. Cancer Sci. 103:913–920. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Adler V, Yin Z, Fuchs SY, Benezra M, Rosario L, Tew KD, Pincus MR, Sardana M, Henderson CJ, Wolf CR, Davis RJ and Ronai Z: Regulation of JNK signaling by GSTp. EMBO J. 18:1321–1334. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Sau A, Filomeni G, Pezzola S, D’Aguanno S, Tregno FP, Urbani A, Serra M, Pasello M, Picci P, Federici G and Caccuri AM: Targeting GSTP1-1 induces JNK activation and leads to apoptosis in cisplatin-sensitive and -resistant human osteosarcoma cell lines. Mol Biosyst. 8:994–1006. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Hu Y, Ju Y, Lin D, Wang Z, Huang Y, Zhang S, Wu C and Jiao S: Mutation of the Nrf2 gene in non-small cell lung cancer. Mol Biol Rep. 39:4743–4747. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kim YR, Oh JE, Kim MS, Kang MR, Park SW, Han JY, Eom HS, Yoo NJ and Lee SH: Oncogenic NRF2 mutations in squamous cell carcinomas of oesophagus and skin. J Pathol. 220:446–451. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Shibata T, Kokubu A, Saito S, Narisawa-Saito M, Sasaki H, Aoyagi K, Yoshimatsu Y, Tachimori Y, Kushima R, Kiyono T and Yamamoto M: NRF2 mutation confers malignant potential and resistance to chemoradiation therapy in advanced esophageal squamous cancer. Neoplasia. 13:864–873. 2011.PubMed/NCBI

32 

Ando N, Kato H, Igaki H, Shinoda M, Ozawa S, Shimizu H, Nakamura T, Yabusaki H, Aoyama N, Kurita A, Ikeda K, Kanda T, Tsujinaka T, Nakamura K and Fukuda H: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol. 19:68–74. 2012. View Article : Google Scholar

Related Articles

Journal Cover

October 2013
Volume 30 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamamoto Y, Konishi H, Ichikawa D, Arita T, Shoda K, Komatsu S, Shiozaki A, Ikoma H, Fujiwara H, Okamoto K, Okamoto K, et al: Significance of GSTP1 for predicting the prognosis and chemotherapeutic efficacy in esophageal squamous cell carcinoma. Oncol Rep 30: 1687-1694, 2013
APA
Yamamoto, Y., Konishi, H., Ichikawa, D., Arita, T., Shoda, K., Komatsu, S. ... Otsuji, E. (2013). Significance of GSTP1 for predicting the prognosis and chemotherapeutic efficacy in esophageal squamous cell carcinoma. Oncology Reports, 30, 1687-1694. https://doi.org/10.3892/or.2013.2606
MLA
Yamamoto, Y., Konishi, H., Ichikawa, D., Arita, T., Shoda, K., Komatsu, S., Shiozaki, A., Ikoma, H., Fujiwara, H., Okamoto, K., Ochiai, T., Inoue, J., Inazawa, J., Otsuji, E."Significance of GSTP1 for predicting the prognosis and chemotherapeutic efficacy in esophageal squamous cell carcinoma". Oncology Reports 30.4 (2013): 1687-1694.
Chicago
Yamamoto, Y., Konishi, H., Ichikawa, D., Arita, T., Shoda, K., Komatsu, S., Shiozaki, A., Ikoma, H., Fujiwara, H., Okamoto, K., Ochiai, T., Inoue, J., Inazawa, J., Otsuji, E."Significance of GSTP1 for predicting the prognosis and chemotherapeutic efficacy in esophageal squamous cell carcinoma". Oncology Reports 30, no. 4 (2013): 1687-1694. https://doi.org/10.3892/or.2013.2606