Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Predictive Biomarkers and Personalized Medicine

Prognostic and Predictive Role of JWA and XRCC1 Expressions in Gastric Cancer

Shouyu Wang, Xuming Wu, Yansu Chen, Jianbing Zhang, Jingjing Ding, Yan Zhou, Song He, Yongfei Tan, Fulin Qiang, Jin Bai, Jinyan Zeng, Zhenghua Gong, Aiping Li, Gang Li, Oluf Dimitri Røe and Jianwei Zhou
Shouyu Wang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xuming Wu
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yansu Chen
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jianbing Zhang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jingjing Ding
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yan Zhou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Song He
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yongfei Tan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Fulin Qiang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jin Bai
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jinyan Zeng
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhenghua Gong
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Aiping Li
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gang Li
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Oluf Dimitri Røe
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jianwei Zhou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-11-2863 Published May 2012
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: To investigate the expression pattern and significance of DNA repair genes JWA and X-ray repair cross complement group 1 (XRCC1) in gastric cancer.

Experimental Design: Expressions of JWA and XRCC1 were assessed by immunohistochemistry in a training cohort and they went into a second testing cohort and finally to a validating cohort. Prognostic and predictive role of JWA and XRCC1 expression status in cases treated with surgery alone or combined with adjuvant chemotherapy was evaluated, respectively.

Results: JWA and XRCC1 protein levels were significantly downregulated in gastric cancer lesions compared with adjacent noncancerous tissues. Low tumoral JWA or XRCC1 expression significantly correlated with shorter overall survival (OS), as well as with clinicopathologic characteristics in patients without adjuvant treatment. Multivariate regression analysis showed that low JWA and XRCC1 expressions, separately and together, were independent negative markers of OS. Adjuvant fluorouracil-leucovorin-oxaliplatin (FLO) significantly improved OS compared with surgery alone (log-rank test, P = 0.01). However, this effect was evident only in the JWA or XRCC1 low expression group (HR = 0.44; 95% CI: 0.26–0.73; P = 0.002, and HR = 0.44, 95% CI: 0.26–0.75; P = 0.002, respectively); Adjuvant fluorouracil-leucovorin-platinol (FLP) did not improve OS, except in the patients with low JWA and XRCC1 expressions (P = 0.010 for JWA and 0.024 for XRCC1, respectively).

Conclusions: JWA and XRCC1 protein expressions in tumor are novel candidate prognostic markers and predictive factors for benefit from adjuvant platinum-based chemotherapy (FLO or FLP) in resectable human gastric carcinoma. Clin Cancer Res; 18(10); 2987–96. ©2012 AACR.

Translational Relevance

This is the first report that has examined expression of the JWA and X-ray repair cross complement group 1 (XRCC1) and their prognostic and predictive significance in human gastric carcinoma. The expressions of JWA and XRCC1 were reduced in gastric cancer tissues and significantly correlated with shorter overall survival, as well as with advanced clinicopathologic features in patients. Moreover, resectable gastric cancer patients with low JWA and XRCC1 expressions could have a survival benefit from adjuvant platinum-based chemotherapy (FLO or FLP). Our findings indicate that JWA and XRCC1 may be candidate prognostic and predictive biomarkers and potentially interesting for the personalized chemotherapy of gastric cancer patients.

Introduction

Gastric cancer is the fourth most common cancer and the second leading cause of cancer-related death worldwide (1). Despite the improved prognosis of patients with gastric cancer resulting from earlier diagnosis, radical surgery, and the development of adjuvant therapy, the 5-year survival rate across all stages is only about 20%. Chemotherapy both in resectable and advanced disease has only limited efficacy (2, 3). New molecular markers pivotal to tumor biology, to improve prognosis and prediction of the adjuvant treatment regimens are urgently needed.

DNA repair systems have been increasingly implicated both in carcinogenesis and treatment resistance (4–6). Reactive oxygen species–induced oxidative base lesions are important to carcinogenesis (7). Base excision repair (BER) pathway is the primary mechanism for repair of these lesions (8), and abnormal expression of the molecular targets in the BER pathway has also been associated with carcinogenesis (9). X-ray repair cross complement group 1 (XRCC1) protein, acts as a scaffold in the process of BER. It recognizes DNA breaks, binds the DNA and recruits other components of the repair machinery (10–12). Molecular epidemiologic studies indicate that single-nucleotide polymorphisms of XRCC1 were associated with the risk of various cancers including gastric cancer as well as being predictive for chemotherapy outcome (13–15). Few studies, however, have investigated XRCC1 expression in human tumors. Low XRCC1 expression was reported in pancreatic cancer versus adjacent tissue as well as in predicting the outcome after bladder cancer radiotherapy (16, 17).

We recently showed that JWA, also called ADP ribosylation–like factor 6 interacting protein 5 (ARL6ip5), may serve as a novel regulator of XRCC1 in the BER protein complex to facilitate repair of DNA damage (18). In addition, we showed that JWA is a novel microtubule-associated protein, which regulates cancer cell migration via mitogen-activated protein kinase cascades (19) and inhibits cell adhesion, invasion, and the metastasis of melanoma cells by suppressing integrin-αVβ3 signaling (20). Our group also showed that polymorphisms in the JWA gene are associated with increased susceptibility to gastric cancer in a Chinese population (21).

In this context, we aimed to address this paucity of translational information and identify the expression patterns of JWA and XRCC1 in 3 large independent cohorts of gastric cancer patients and to examine the possible prognostic and predictive role of these markers.

Materials and Methods

Patients and specimens

Three independent retrospective patient cohorts were studied. The training cohort and testing cohort were recruited in Nantong Cancer Hospital, Nantong City, in the North of Jiangsu Province, China. The validation cohort was recruited in Yixing People's Hospital, Yixing City, in the South of Jiangsu Province, China. The tissues were obtained from the respective pathology departments. Inclusion criteria were gastric carcinoma treated with radical gastrectomy with or without adjuvant chemotherapy. Exclusion criteria were previous gastric cancer or active nongastric cancer, preoperative chemotherapy or radiotherapy.

The training cohort included 103 patients who underwent radical gastrectomy at Nantong Cancer Hospital from May 1, 1990 to June 1, 1995. A tissue microarray (TMA) was constructed, including the gastric cancer samples and matched noncancerous gastric mucosa more than 5 cm from the tumoral margins. Two more independent tumor TMAs were constructed to validate training cohort data, all patients operated before 2006 to evaluate at least 5-year survival. The testing cohort consisted of all 640 surgical cases from the Nantong Cancer Hospital from December 1, 2000 to April 1, 2005 and the validation cohort included all 1,022 surgical cases in Yixing People's Hospital from January 1, 1999 to December 31, 2006. These patients were treated with surgery only or with postoperative adjuvant chemotherapy (for details, see the Supplementary Methods and Supplementary Fig. S1). As shown in Supplementary Table S1, the distributions of demographic characteristics and the selected clinicopathologic variables of patients between the 2 districts (Nantong and Yixing) were significantly different (all of P < 0.05). However, the distributions of these variables of patients between the training cohort and testing cohort in Nantong district were mostly matched, except depth of invasion and distant metastasis (P = 0.005 and 0.003, respectively; data not shown). For the resectable gastric cancer patients with chemotherapy, the distributions of demographic characteristics and the selected clinicopathologic variables of patients between fluorouracil-leucovorin-oxaliplatin (FLO) and fluorouracil-leucovorin-platinol (FLP) groups were similar (all of P > 0.05), except histologic type (P = 0.003; Supplementary Table S2). In addition, 11 pathologically confirmed gastric cancer and respective noncancerous fresh-frozen gastric mucosa tissues from recent patients from Nantong Cancer Hospital were obtained for Western blot analysis after signed informed consent. Institutional approval was obtained from the Review Board of the respective institutions before this study.

Overall survival (OS) was the primary endpoint of this analysis. Survival time was calculated from the date of surgery to the date of death or to the last follow-up. Date of death was obtained from patient records or patients' families through follow-up telephone calls. Date of death for each case was double verified by local civil affairs department and public security department. Detailed clinicopathologic information was obtained. The Lauren criteria were used to classify the tumors into intestinal or diffuse types (22) and staged according to the tumor-node-metastasis (TNM) guidelines (23).

TMA construction and immunohistochemistry

The gastric cancer TMAs were created by contract service at the National Engineering Center for Biochip, Shanghai, China. Duplicate 1.0-mm diameter cores of tissue from each sample were punched from paraffin tumor block and corresponding nontumoral tissues in the training cohort or from cores of primary tumor biopsies in the validation cohorts. As a tissue control, the biopsies of normal gastric epithelium tissues were inserted in the 4 angles and the center of each slide.

A standard protocol was used for the immunostaining of the TMAs (for details, see Supplementary Methods). The polyclonal rabbit anti-JWA antibody (1:200 dilution; Research Genetics Inc.) and monoclonal mouse anti-XRCC1 antibody (1:300 dilution; Abcam), were used as described previously (18). The omission of the primary antibody served as negative control. The staining scores of the tissue controls in each microarray slide were preevaluated as a quality control of the immunostaining.

Assessment of immunohistochemistry

At first, staining of JWA and XRCC1 in the tissue was scored independently by 2 pathologists blinded to the clinical data, by applying a semiquantitative immunoreactivity score (IRS) in the training cohort, as reported elsewhere (24). Category A documented the intensity of immunostaining as 0–3 (0, negative; 1, weak; 2, moderate; 3, strong; Supplementary Fig. S2). Category B documented the percentage of immunoreactive cells as 1 (0%–25%), 2 (26%–50%), 3 (51%–75%), and 4 (76%–100%). Multiplication of category A and B resulted in an IRS ranging from 0 to 12 for each tumor or nontumor. The concordance for IRS staining score of JWA and XRCC1 between the 2 pathologists was 73 (91%) and 71 (89%) in 80 tumors of the training set, respectively; the few discrepancies were resolved by consensus using a multihead microscope. The variability in JWA and XRCC1 staining was 3 (4%) and 2 (3%) in the duplicate cores of 80 tumors, respectively. These cases were stained by whole-slide immunohistochemistry and further scored.

The optimum cutoff value of IRS is obtained by receiver-operator characteristic (ROC) analysis, the area under the curve (AUC) at different cutoff values of JWA or XRCC1 IRS for 1, 3, and 5 years of OS time was calculated. The optimal value of cutoff points of the JWA or XRCC1 IRS in Nantong district cohort (combined training cohort and testing cohort) was 4 or 3 due to the predictive value of this cutoff point for death was the best (Supplementary Fig. S3). Under these conditions, samples with IRS 0–4 and IRS 5–12 or IRS 0–3 and IRS 4–12 were classified as low and high expression of JWA or XRCC1 in tumors, respectively. After establishing the immunohistochemical assessment criteria in the Nantong district cohort, the expressions of JWA and XRCC1 in the Yixing district cohort (validation cohort) were scored by the same pathologists with the same procedure.

Western blotting

Total 11 pairs of recently collected fresh tissues were ground in liquid nitrogen and washed 3 times with phosphate-buffered saline. Tissue extracts were made with a detergent lysis buffer [50 mmol/L Tris (pH 7.4); 150 mmol/L NaCl; 1% NP-40; 0.5% sodium deoxycholate; 0.1% SDS; and the protease inhibitor, 1 mmol/L phenylmethanesulfonyl fluoride). Protein (60 μg) was run on a 12.5% or 7.5% PAGE and transferred to a nitrocellulose membrane (Hybond-ECL; Amersham Pharmacia Biotech). The membrane was blocked with Tris-buffered saline containing 0.1% Tween 20 and 5% nonfat milk (w/v; TBSTM) for 2 hours at room temperature and then incubated overnight at 4°C with primary antibody diluted in TBSTM, which included the polyclonal rabbit anti-JWA antibody (1:1,000 dilution, Research Genetics Inc.), monoclonal mouse anti-XRCC1 antibody (1:1,000 dilution; Abcam), and monoclonal mouse anti–β-actin antibody (1:2,000 dilution; Boster Biotechnology). Immunoreactive bands were detected with a Phototope-horseradish peroxide Western blot detection kit (Cell Signaling Technology Inc.). The intensity of the JWA and XRCC1 protein bands were analyzed by densitometry, after normalization to the corresponding β-actin level.

Statistical analysis

The associations between JWA or XRCC1 expression and clinicopathologic parameters were evaluated by Fisher exact test. The significance of correlations of JWA or XRCC1 staining in primary tumors and their corresponding nontumors were assessed by the Wilcoxon test (grouped) and by Spearman rank-order correlation (raw scores). The correlation of the expressions of JWA and XRCC1 was established by Spearman rank-order correlation (raw scores) and Fisher exact test (grouped). Probability of differences in OS as a function of time was ascertained by use of the Kaplan–Meier method, with a log-rank test probe for significance. Univariate or multivariate Cox regression analysis was conducted to estimate the crude HRs, adjusted HRs and their 95% CIs, with adjustment for potential confounders. Based on the sample size in the testing (374 cases) and validation cohorts (385 cases), 0.51 difference HR (upper limit of CI in the training cohort) between JWA low and high expression levels with both power of 1.0, and 0.69 difference HR between XRCC1 low and high expression levels with both power of 0.95 could be determined. We analyzed the predictive value of the parameters using time-dependent ROC curve analysis for censored data and calculated AUC of the ROC curves (25). We evaluated the performances of different scores by plotting (t, AUC [t]) for different values of follow-up time (t). All the statistical analyses were conducted by Statistical Analysis System software (version 9.1.3; SAS Institute), STATA statistical software (version 10.1; StataCorp), and R software (version 2.10.1; The R Foundation for Statistical Computing). P < 0.05 was deemed statistically significant.

Results

Reduced JWA and XRCC1 expressions in gastric cancer versus noncancer tissues

Eleven pairs of human gastric cancer samples, including primary gastric cancer tissues and matched normal gastric mucosa were selected to test JWA and XRCC1 protein expressions by Western blot. Decreased expressions of JWA and XRCC1 occurred in all of gastric tumors compared with the paired normal gastric mucosa (Fig. 1A). Immunohistochemical staining of the gastric TMA was used to further investigate JWA and XRCC1 expressions in 80 gastric cancer patients in the training cohort. Because some samples were lost during antigen retrieval or with no relevant cells present in the core, JWA and XRCC1 expressions were examined in 79 and 77 gastric cancer patients having both of gastric cancerous tissues and matched normal gastric mucosa. It was shown that JWA staining was mainly localized in the cytoplasm, whereas XRCC1 was exclusively expressed in the nuclei (Fig. 1B). The distribution of the differences of IRS for JWA and XRCC1 expressions in nontumors and matched tumors was shown in Fig. 1C. Moreover, JWA and XRCC1 expressions were significantly decreased in 69 of 79 (87.3%) and 59 of 77 (76.6%) of gastric cancers compared with the matched normal gastric tissues (both P < 0.001, Wilcoxon test; Fig. 1C).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Correlation of JWA and XRCC1 expressions in primary tumors and corresponding nontumors in gastric cancer patients. A, JWA and XRCC1 protein levels in 11 cancer tissues and paired noncancerous normal tissues of gastric cancer patients were analyzed by Western blotting. The level of each protein was normalized against β-actin, and the protein levels in cancer tissues indicated as a ratio to paired noncancerous normal tissues. N, noncancerous normal tissue; T, tumor tissue. B and D, immunohistochemical staining for JWA and XRCC1 in TMA, respectively. C, gastric cancerous tissues; N, paired noncancerous gastric tissues. Top, original magnification: ×40; bottom, magnification: ×200. C and E, the distribution of the difference of JWA and XRCC1 staining (Δ IRS = IRSN-IRST), respectively. P values were calculated with the Wilcoxon test.

In all 3 independent cohorts of patients treated only with surgery, the expression of JWA correlated significantly with XRCC1 expression in the cancerous tissues (P < 0.001 for all correlations; Table 1).

View this table:
  • View inline
  • View popup
Table 1.

Relationship between expression levels of JWA and clinicopathologic features of the individuals in 3 cohorts of gastric cancers treated with surgery alone

Association of JWA and XRCC1 expressions with clinicopathologic characteristics in patients treated only with surgery

Protein expressions of JWA and XRCC1 in the cancerous tissues of all 3 cohorts were significantly associated with clinicopathologic features, such as lymph node metastasis (N-category), and TNM stage (Table 1 and Supplementary Table S3). Significantly more low JWA expression was seen in diffuse type in all 3 cohorts, but low XRCC1 was significant in the 2 larger cohorts. However, there was no correlation between JWA or XRCC1 protein expression in noncancerous tissues and clinicopathologic features in the training cohort (Supplementary Table S4).

Correlation of JWA, XRCC1 expression, and OS in patients treated only with surgery

In the training cohort, 80 primary tumor samples eligible for analysis showed a statistically significant positive correlation between JWA or XRCC1 expression and overall 5-year survival using Kaplan–Meier survival curves (P < 0.001 for both). These findings were confirmed in 2 independent and larger (n = 374 and 385, respectively) cohorts of gastric cancer patients with minimum 5-year follow-up (Fig. 2A–C). The patients were then stratified into 3 distinct groups depending on staining for JWA and XRCC1: both high, 1 high (JWA or XRCC1 high), and both low. It was shown that patients with both high had a better outcome of survival than in the 2 other groups (P < 0.001, log-rank test; Fig. 2). Other significant negative predictors for survival by univariate analysis in the 3 independent cohorts were lymph node metastasis (N-category; P < 0.01 for all) and clinical TNM stage (P < 0.001 for all; data not shown). Nontumoral JWA or XRCC1 expression was not correlated with OS (Supplementary Fig. S4A and B).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Kaplan–Meier curves depicting OS according to expression patterns of JWA, XRCC1, and combined with JWA/XRCC1 expression in training cohort (A), testing cohort (B), and validation cohort (C). P values were calculated with the log-rank test.

The multivariate Cox regression analysis indicated that high JWA and XRCC1 expressions were independent positive prognostic factors separately or together for gastric cancer in all 3 cohorts (P < 0.05 for all, Table 2).

View this table:
  • View inline
  • View popup
Table 2.

Multivariate Cox regression analysis of JWA, XRCC1, or JWA/XRCC1 expression and clinicopathologic variables predicting survival in 3 cohorts of gastric cancers treated with surgery alone

To further evaluate the prognostic efficacy of JWA and XRCC1 expressions, we conducted a time-dependent ROC analysis for the censored data, which indicated that the combination of the clinical risk score (TNM stage, histologic type, and tumor diameter) and JWA or XRCC1 or JWA plus XRCC1 contributed much more than either one alone in both of training and testing cohorts (Fig. 3). For example, in the testing cohort, the AUC at year 5 was 0.715 (95% CI: 0.662–0.769) for clinical risk score, whereas it was significantly increased to 0.912 (95% CI: 0.881–0.942) when combination of the clinical risk score with JWA plus XRCC1 risk score. However, this effect was not significant in the validation cohort due to the relatively higher AUC (about 0.8) of clinical predictors (Supplementary Fig. S5).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Time-dependent ROC analyses for the clinical risk score (TNM stage, histologic type, and tumor diameter), the combined JWA, XRCC1, or JWA plus XRCC1 and clinical risk score in the training cohort (A) and testing cohort (B).

Correlation between JWA or XRCC1 expressions and OS in patients with adjuvant chemotherapy

In the testing and validation cohorts, OS was analyzed between the patients who received adjuvant chemotherapy versus those who did not. Data showed no difference in OS between the surgery only group and any regimen of postoperative adjuvant chemotherapy (data not shown) except in the group receiving FLO (n = 93, log-rank test, P = 0.01; Fig. 4). A multivariate Cox regression analysis including 6 variables (age, gender, TNM stage, histologic types, tumor diameter, and chemotherapy treatment) was conducted to indicate the benefit of chemotherapy on OS. There was a statistically significant benefit only of FLO chemotherapy over surgery alone (HR = 0.50, 95% CI: 0.34–0.73, data not shown). Conspicuously, this effect was only found in low JWA or XRCC1 expression patients in which adjuvant FLO significantly increased OS as compared with surgery alone (for JWA, HR = 0.44, 95% CI: 0.26–0.73, Supplementary Table S4; log-rank test, P = 0.002; Fig. 4; for XRCC1, HR = 0.44, 95% CI: 0.26–0.75, Supplementary Table S4; log-rank test, P = 0.02; Fig. 4). Moreover, patients with high JWA or XRCC1 expression in tumors had no additional survival benefit from adjuvant chemotherapy (Supplementary Table S4 and Fig. 4).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Kaplan–Meier curves depicting OS according to JWA (A) or XRCC1 (B) expression patterns in validation cohort patients treated with or without FLO. P values were calculated with the log-rank test. S, surgery alone.

We also analyzed the significance of another platinum-based chemotherapy, FLP regimen (n = 78) in resectable gastric cancer. The results did not show a significant survival difference (log-rank test, P = 0.364; Supplementary Fig. S6), whereas low JWA or XRCC1 expression patients receiving FLP regimen showed a trend of longer survival compared with those with surgery only (Supplementary Fig. S6). In contrast, high JWA or XRCC1 expression patients with FLP regimen had significantly shorter survival compared with those with surgery only (JWA, P < 0.001; XRCC1, P = 0.003; Supplementary Fig. S6). Further multivariate analysis indicated that lower risk for mortality was observed in those with FLP chemotherapy compared with surgery only (HR = 0.55, 95% CI: 0.35–0.86 for JWA; HR = 0.58, 95% CI: 0.36–0.93 for XRCC1; Supplementary Table S6).

Discussion

One of the most challenging problems in oncology is that we know that a large percentage of the cancer patients are treated unnecessary, but we do not know how to select them. Even in patients with similar clinical or pathologic features, their survival outcomes vary. Discovery of prognostic and predictive biomarkers may enable personalized cancer therapies. In this study, we recorded and independently confirmed that low expression of JWA and XRCC1 were significantly associated with unfavorable clinicopathologic parameters and decreased overall patient survival. Moreover, patients with low JWA or XRCC1 expression in tumors had significant survival benefit from adjuvant first-line platinum-based-chemotherapy (FLO or FLP).

In this study, we show significant underexpression of both JWA and XRCC1 proteins in gastric cancer cells versus paired normal tissue, indicating a potentially important role of these genes in gastric carcinogenesis. These data were consistent with previous studies, which reported that normal nevi and normal melanocytes had high JWA expression and low expression in dysplastic nevi and malignant melanoma tissues and cell lines (20). Moreover, XRCC1 expression was downregulated in pancreatic cancer versus normal cells (16). To study the possible tumor suppressor effect of JWA, we recently constructed conditional JWA knockout mice. However, spontaneous tumors of these mice were not observed (Wang and colleagues; unpublished data), indicating that environment–gene interactions in tumorigenesis should be considered. The exact mechanisms of XRCC1 in tumorigenesis are formidable to study in animal models due to the embryonic lethality in XRCC1 knockout mice (26).

An interesting question that arises in this study is, how can low expression of the same protein be both a negative prognostic factor and a positive predictive factor? These Janus-like properties in cancer biology and treatment resistance has been documented for the nucleotide excision repair (NER) protein ERCC1 in which a high ERCC1 expression in resectable lung cancer was a positive prognostic factor, but a negative predictive factor for platinum treatment (27–29). In vitro and animal studies indicated that the loss of JWA decreased cell differentiation (30) and increased cell migration and metastasis (19, 20). Similarly, in our study, low JWA expression in gastric tumors correlated with unfavorable TNM stage and diffuse type, thus to a more malignant, aggressive phenotype with a negative prognosis when untreated. Conversely, the positive predictive effect of low JWA and XRCC1 on survival in both platinum treated patients was highly significant. This may point to a role of both proteins in chemoresistance, probably related to platinum. Dysregulation of several DNA repair mechanisms is an important modulator of platinum effect (31). The role of JWA in these DNA repair mechanisms remains to be elucidated, but lowering of JWA was shown to increase susceptibility to DNA-damaging agents (18). Moreover, JWA was shown to play a role in transcriptional and translational regulation of XRCC1 levels (18). XRCC1 is a critical component of the BER pathway, and downregulation of BER sensitized cancer to cisplatin or oxaliplatin (32, 33). XRCC1 is also involved in NER (34), homologous recombination, and nonhomologous end joining (35, 36), which are involved in platinum resistance (31). However, the direct roles of JWA and XRCC1 in platinum treatment need to be further provided.

In interpreting our results, several issues need to be considered and clarified. First, a timely question about this type of study is how reliable is the interobserver concordance on scoring the immunohistochemical expression. IRS was a reliable tool in our hands and reported to be robust for other antibodies, even with basic training only (37). Moreover, we applied ROC analysis to exclude the subjective division of IRS. Second, the use of only duplicate 1.0-mm diameter cores of tissue from each sample when preparing TMA might lead to a limitation of reprehensive samples due to tumor heterogeneity. However, 3 relatively large independent TMA were used to minimize the impact of tumor heterogeneity and determine a reliable value of our studies. Third, it must be noted that the patients received more survival benefit from FLO than from FLP, which may be partially due to that FLO reduces toxicity as compared with FLP (38) and oxaliplatin has recently been shown to induce immunogenic cell death in vivo (39), while cisplatin fails to do so (40). As the study is retrospective in nature, and the number of patients receiving first-line oxaliplatin or cisplatin-based adjuvant chemotherapy is relatively small, large efforts were done to obtain correct clinical and survival data (see Materials and Methods). The database built upon this information is thus as complete as possible. Nevertheless, despite highly significant results in such a large patient material, these markers should be validated in different ethnic population and prospective studies are warranted before using these markers in the clinic.

Taken together, we found that 2 BER proteins, JWA and XRCC1 were higher expressed in noncancerous gastric mucosa than in gastric cancer tissues. We report for the first time that JWA and XRCC1 are potential prognostic biomarkers and predictors for adjuvant chemotherapy with platinum-based regimen (FLO or FLP) in resectable gastric cancer patients. The simplicity of immunostaining and assessment by IRS makes these proteins as interesting candidates for personalizing gastric cancer treatment.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Authors' Contributions

Conception and design: S. Wang, X. Wu, J. Ding, Y. Tan, G. Li, F. Qiang, J. Zhou

Development of methodology: S. Wang, J. Ding, A. Li, J. Zhou

Acquisition of data: X. Wu, J. Ding, Y. Zhou, Y. Tan, J. Zhang, S. He, F. Qiang, J. Zhou

Analysis and interpretation of data: S. Wang, Y. Chen, J. Ding, O. Dimitri

Writing, review, and/or revision of the manuscript: S. Wang, G. Li, O. Dimitri, J. Zhou

Administrative, technical, or material support: S. Wang, J. Bai, J. Zeng, Z. Gong, A. Li, J. Zhou

Study supervision: S. Wang, J. Zhou

Grant Support

This work was supported in part by the project funded by the Priority Academic Program Development (PAPD) of Jiangsu Higher Education Institutions, the National Natural Science Foundation of China (30930080 and 81001231), the Foundation of Cancer Center of Nanjing Medical University (08ZLKF08), and the Postdoctoral Science Foundation of China (20100481165).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Footnotes

  • Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

  • Received November 8, 2011.
  • Revision received February 1, 2012.
  • Accepted February 18, 2012.
  • ©2012 American Association for Cancer Research.

References

  1. 1.↵
    1. Parkin DM,
    2. Bray F,
    3. Ferlay J,
    4. Pisani P
    . Global cancer statistics, 2002. CA Cancer J Clin 2005;55:74–108.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Paoletti X,
    2. Oba K,
    3. Burzykowski T,
    4. Michiels S,
    5. Ohashi Y,
    6. Pignon JP,
    7. et al.
    Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA 2010;303:1729–37.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Van Cutsem E,
    2. Moiseyenko VM,
    3. Tjulandin S,
    4. Majlis A,
    5. Constenla M,
    6. Boni C,
    7. et al.
    Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. J Clin Oncol 2006;24:4991–7.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Caldecott KW
    . Mammalian single-strand break repair: mechanisms and links with chromatin. DNA Repair (Amst) 2007;6:443–53.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. de Boer J,
    2. Hoeijmakers JH
    . Nucleotide excision repair and human syndromes. Carcinogenesis 2000;21:453–60.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Helleday T,
    2. Lo J,
    3. van Gent DC,
    4. Engelward BP
    . DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amst) 2007;6:923–35.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Klaunig JE,
    2. Kamendulis LM
    . The role of oxidative stress in carcinogenesis. Annu Rev Pharmacol Toxicol 2004;44:239–67.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Wilson DM 3rd.,
    2. Bohr VA
    . The mechanics of base excision repair, and its relationship to aging and disease. DNA Repair (Amst) 2007;6:544–59.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Caldecott KW
    . Single-strand break repair and genetic disease. Nat Rev Genet 2008;9:619–31.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Kubota Y,
    2. Nash RA,
    3. Klungland A,
    4. Schar P,
    5. Barnes DE,
    6. Lindahl T
    . Reconstitution of DNA base excision-repair with purified human proteins: interaction between DNA polymerase beta and the XRCC1 protein. Embo J 1996;15:6662–70.
    OpenUrlPubMed
  11. 11.↵
    1. Taylor RM,
    2. Wickstead B,
    3. Cronin S,
    4. Caldecott KW
    . Role of a BRCT domain in the interaction of DNA ligase III-alpha with the DNA repair protein XRCC1. Curr Biol 1998;8:877–80.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. El-Khamisy SF,
    2. Masutani M,
    3. Suzuki H,
    4. Caldecott KW
    . A requirement for PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucleic Acids Res 2003;31:5526–33.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Li WQ,
    2. Zhang L,
    3. Ma JL,
    4. Zhang Y,
    5. Li JY,
    6. Pan KF,
    7. et al.
    Association between genetic polymorphisms of DNA base excision repair genes and evolution of precancerous gastric lesions in a Chinese population. Carcinogenesis 2009;30:500–5.
    OpenUrlAbstract/FREE Full Text
  14. 14.↵
    1. Goekkurt E,
    2. Al-Batran SE,
    3. Hartmann JT,
    4. Mogck U,
    5. Schuch G,
    6. Kramer M,
    7. et al.
    Pharmacogenetic analyses of a phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil and leucovorin plus either oxaliplatin or cisplatin: a study of the arbeitsgemeinschaft internistische onkologie. J Clin Oncol 2009;27:2863–73.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    1. Wu X,
    2. Gu J,
    3. Wu TT,
    4. Swisher SG,
    5. Liao Z,
    6. Correa AM,
    7. et al.
    Genetic variations in radiation and chemotherapy drug action pathways predict clinical outcomes in esophageal cancer. J Clin Oncol 2006;24:3789–98.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Crnogorac-Jurcevic T,
    2. Efthimiou E,
    3. Nielsen T,
    4. Loader J,
    5. Terris B,
    6. Stamp G,
    7. et al.
    Expression profiling of microdissected pancreatic adenocarcinomas. Oncogene 2002;21:4587–94.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Sak SC,
    2. Harnden P,
    3. Johnston CF,
    4. Paul AB,
    5. Kiltie AE
    . APE1 and XRCC1 protein expression levels predict cancer-specific survival following radical radiotherapy in bladder cancer. Clin Cancer Res 2005;11:6205–11.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Wang S,
    2. Gong Z,
    3. Chen R,
    4. Liu Y,
    5. Li A,
    6. Li G,
    7. et al.
    JWA regulates XRCC1 and functions as a novel base excision repair protein in oxidative-stress-induced DNA single-strand breaks. Nucleic Acids Res 2009;37:1936–50.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Chen H,
    2. Bai J,
    3. Ye J,
    4. Liu Z,
    5. Chen R,
    6. Mao W,
    7. et al.
    JWA as a functional molecule to regulate cancer cells migration via MAPK cascades and F-actin cytoskeleton. Cell Signal 2007;19:1315–27.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Bai J,
    2. Zhang J,
    3. Wu J,
    4. Shen L,
    5. Zeng J,
    6. Ding J,
    7. et al.
    JWA regulates melanoma metastasis by integrin alphaVbeta3 signaling. Oncogene 2010;29:1227–37.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Tang WY,
    2. Wang L,
    3. Li C,
    4. Hu ZB,
    5. Chen R,
    6. Zhu YJ,
    7. et al.
    Identification and functional characterization of JWA polymorphisms and their association with risk of gastric cancer and esophageal squamous cell carcinoma in a Chinese population. J Toxicol Environ Health A 2007;70:885–94.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Sakamoto H,
    2. Yoshimura K,
    3. Saeki N,
    4. Katai H,
    5. Shimoda T,
    6. Matsuno Y,
    7. et al.
    Genetic variation in PSCA is associated with susceptibility to diffuse-type gastric cancer. Nat Genet 2008;40:730–40.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Japanese Gastric Cancer A
    . Japanese classification of gastric carcinoma - 2nd English Edition. Gastric Cancer 1998;1:10–24.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Weichert W,
    2. Roske A,
    3. Gekeler V,
    4. Beckers T,
    5. Ebert MP,
    6. Pross M,
    7. et al.
    Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol 2008;9:139–48.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Heagerty PJ,
    2. Lumley T,
    3. Pepe MS
    . Time-dependent ROC curves for censored survival data and a diagnostic marker. Biometrics 2000;56:337–44.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Tebbs RS,
    2. Thompson LH,
    3. Cleaver JE
    . Rescue of Xrcc1 knockout mouse embryo lethality by transgene-complementation. DNA Repair (Amst) 2003;2:1405–17.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Zheng Z,
    2. Chen T,
    3. Li X,
    4. Haura E,
    5. Sharma A,
    6. Bepler G
    . DNA synthesis and repair genes RRM1 and ERCC1 in lung cancer. N Engl J Med 2007;356:800–8.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Gazdar AF
    . DNA repair and survival in lung cancer–the two faces of Janus. N Engl J Med 2007;356:771–3.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Olaussen KA,
    2. Dunant A,
    3. Fouret P,
    4. Brambilla E,
    5. Andre F,
    6. Haddad V,
    7. et al.
    DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med 2006;355:983–91.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Huang S,
    2. Shen Q,
    3. Mao WG,
    4. Li AP,
    5. Ye J,
    6. Liu QZ,
    7. et al.
    JWA, a novel signaling molecule, involved in the induction of differentiation of human myeloid leukemia cells. Biochem Biophys Res Commun 2006;341:440–50.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Rabik CA,
    2. Dolan ME
    . Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 2007;33:9–23.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Yang J,
    2. Parsons J,
    3. Nicolay NH,
    4. Caporali S,
    5. Harrington CF,
    6. Singh R,
    7. et al.
    Cells deficient in the base excision repair protein, DNA polymerase beta, are hypersensitive to oxaliplatin chemotherapy. Oncogene 2010;29:463–8.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Zhang R,
    2. Niu Y,
    3. Zhou Y
    . Increase the cisplatin cytotoxicity and cisplatin-induced DNA damage in HepG2 cells by XRCC1 abrogation related mechanisms. Toxicol Lett 2010;192:108–14.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Moser J,
    2. Kool H,
    3. Giakzidis I,
    4. Caldecott K,
    5. Mullenders LH,
    6. Fousteri MI
    . Sealing of chromosomal DNA nicks during nucleotide excision repair requires XRCC1 and DNA ligase III alpha in a cell-cycle-specific manner. Mol Cell 2007;27:311–23.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Taylor RM,
    2. Moore DJ,
    3. Whitehouse J,
    4. Johnson P,
    5. Caldecott KW
    . A cell cycle-specific requirement for the XRCC1 BRCT II domain during mammalian DNA strand break repair. Mol Cell Biol 2000;20:735–40.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    1. Levy N,
    2. Martz A,
    3. Bresson A,
    4. Spenlehauer C,
    5. de Murcia G,
    6. Menissier-de Murcia J
    . XRCC1 is phosphorylated by DNA-dependent protein kinase in response to DNA damage. Nucleic Acids Res 2006;34:32–41.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Camp RL,
    2. Neumeister V,
    3. Rimm DL
    . A decade of tissue microarrays: progress in the discovery and validation of cancer biomarkers. J Clin Oncol 2008;26:5630–7.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Al-Batran SE,
    2. Hartmann JT,
    3. Probst S,
    4. Schmalenberg H,
    5. Hollerbach S,
    6. Hofheinz R,
    7. et al.
    Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol 2008;26:1435–42.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    1. Tesniere A,
    2. Schlemmer F,
    3. Boige V,
    4. Kepp O,
    5. Martins I,
    6. Ghiringhelli F,
    7. et al.
    Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene 2010;29:482–91.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Obeid M,
    2. Tesniere A,
    3. Ghiringhelli F,
    4. Fimia GM,
    5. Apetoh L,
    6. Perfettini JL,
    7. et al.
    Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007;13:54–61.
    OpenUrlCrossRefPubMed
View Abstract
Previous
Back to top
Clinical Cancer Research: 18 (10)
May 2012
Volume 18, Issue 10
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Prognostic and Predictive Role of JWA and XRCC1 Expressions in Gastric Cancer
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Prognostic and Predictive Role of JWA and XRCC1 Expressions in Gastric Cancer
Shouyu Wang, Xuming Wu, Yansu Chen, Jianbing Zhang, Jingjing Ding, Yan Zhou, Song He, Yongfei Tan, Fulin Qiang, Jin Bai, Jinyan Zeng, Zhenghua Gong, Aiping Li, Gang Li, Oluf Dimitri Røe and Jianwei Zhou
Clin Cancer Res May 15 2012 (18) (10) 2987-2996; DOI: 10.1158/1078-0432.CCR-11-2863

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Prognostic and Predictive Role of JWA and XRCC1 Expressions in Gastric Cancer
Shouyu Wang, Xuming Wu, Yansu Chen, Jianbing Zhang, Jingjing Ding, Yan Zhou, Song He, Yongfei Tan, Fulin Qiang, Jin Bai, Jinyan Zeng, Zhenghua Gong, Aiping Li, Gang Li, Oluf Dimitri Røe and Jianwei Zhou
Clin Cancer Res May 15 2012 (18) (10) 2987-2996; DOI: 10.1158/1078-0432.CCR-11-2863
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Grant Support
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Aromatase in Lung Adenocarcinomas with EGFR Mutations
  • Somatic Mutations and Clinical Outcome in Melanoma Samples
  • FGFR1 Expression Predicts FGFR1-Dependent Lung Cancer
Show more Predictive Biomarkers and Personalized Medicine
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement