Copine 5 expression predicts prognosis following curative resection of esophageal squamous cell carcinoma

  • Authors:
    • Shinichi Umeda
    • Mitsuro Kanda
    • Masahiko Koike
    • Haruyoshi Tanaka
    • Takashi Miwa
    • Chie Tanaka
    • Daisuke Kobayashi
    • Masaya Suenaga
    • Masamichi Hayashi
    • Suguru Yamada
    • Goro Nakayama
    • Yasuhiro Kodera
  • View Affiliations

  • Published online on: September 27, 2018     https://doi.org/10.3892/or.2018.6742
  • Pages: 3772-3780
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Patients with esophageal squamous cell carcinoma (ESCC) have a poor prognosis. Identification of biomarkers to accurately predict the risk of recurrence and survival following curative esophageal resection is required to improve patient outcomes. The copine 5 (CPNE5) gene encodes a calcium‑dependent lipid‑binding intracellular protein. Copine proteins interact with diverse target proteins that are components of pathways that aberrantly regulate the phenotypes of malignant cells. However, limited information is available on the role of CPNE5 in cancer. The present study investigated whether CPNE5 may serve as a predictive marker of the prognosis of patients with ESCC following curative resection. CPNE5 mRNA expression levels and the methylation status of the CPNE5 promotor region were measured in 11 ESCC cell lines. CPNE5 mRNA expression levels in 106 pairs of surgically resected specimens were measured, and their associations with clinicopathological characteristics were analyzed. The CPNE5 mRNA expression levels in 9 ESCC cell lines were decreased compared with those of the non-tumorigenic esophageal mucosa cell line Het‑1A. Bisulfite sequencing detected the methylation of the CPNE5 promotor region in all cell lines tested, including Het‑1A. Furthermore, analysis of tissues revealed that CPNE5 mRNA expression was significantly lower in ESCC cells compared with cognate non-cancerous adjacent mucosal cells. Kaplan‑Meier analysis revealed that patients with low CPNE5 expression experienced significantly shorter overall survival. Multivariable analysis identified low CPNE5 expression to be an independent prognostic factor of OS. Analysis of recurrence patterns revealed that significantly more patients with local recurrence expressed lower levels of CPNE5 mRNA. These findings indicated that CPNE5 expression in ESCC tissues may serve as an informative biomarker for predicting ESCC recurrence, particularly in patients with local recurrence, and may help to ensure that patients receive optimal treatment and follow‑up.

Introduction

Esophageal squamous cell carcinoma (ESCC) is associated with a considerable decline in quality of life, in addition to a poor prognosis (1,2). A primary goal of efforts to improve the management of the disease and patient outcomes is establishing methods to accurately predict the risk of recurrence, in addition to survival, following curative esophageal resection (3). Such information is urgently required to provide appropriate individualized perioperative follow-up and treatment (4,5). Furthermore, a better understanding of the molecular mechanisms of disease progression is essential, and identification of molecules that contribute to the pathogenesis of ESCC may lead to the development of novel biomarkers that facilitate precise risk stratification and monitoring of recurrence following esophagectomy (6,7).

The copine 5 (CPNE5) gene located on human chromosome 6p21.2 (8) belongs to the copine gene family, encoding calcium-dependent lipid-binding proteins comprising two N-terminal C2 domains (C2Ds) and a C-terminal A domain (9). CPNE5 localizes to the cytosol and is expressed at high levels in the brain, lymph nodes, testes and heart (10). CPNE5 is expressed by differentiated neurons during neural development, suggesting that CPNE5 function is important for the function of the central nervous system (11). Furthermore, CPNE5 expression is associated with alcohol dependence and obesity in Caucasians (12). CPNE5 expression may be associated with the progression of ESCC, since copine proteins interact with diverse target proteins, including dual specificity mitogen-activated protein kinase kinase 1 (13), protein phosphatase 5 (14), and CDC42-regulated kinase (15), which are components of intracellular signaling pathways that influence the malignant phenotype (16). However, the role of CPNE5 in cancer is unknown.

The present study assessed whether CPNE5 may serve as a predictive marker of ESCC outcomes following curative resection. To answer this question, the expression of CPNE5 mRNA and the methylation of the CPNE5 promoter was measured in ESCC cell lines and in surgically-resected ESCC tissues.

Materials and methods

Ethics approval and consent to participate

The present study rigidly adhered to the ethical guidelines of the World Medical Association Declaration of Helsinki Ethical Principles for Medical Research Involving Human Subjects. Written informed consent for the use of clinical samples and data was obtained from all patients, as required by the Institutional Review Board of Nagoya University (Nagoya, Japan; approval no. 2014-0044).

Sample collection

ESCC cell lines (TE1, TT and TTn) and a non-tumorigenic epithelial cell line (Het-1A) were obtained from the American Type Culture Collection (Manassas, VA, USA). NUEC2 and WSSC cell lines were established at Nagoya University (17). KYSE510, KYSE590, KYSE890, KYSE1170, KYSE1260 and KYSE1440 cells were purchased from the Japanese Collection of Research Bioresources Cell Bank (Osaka, Japan) (18). Cells were stored at −80°C in a cell preservative (Cell Banker; LSI Medience Corporation, Tokyo, Japan) and cultured in RPMI-1640 medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc., Waltham, MA, USA) in an atmosphere containing 5% CO2 at 37°C. A total of 106 primary ESCC tissues and adjacent normal tissues were acquired from patients who underwent radical esophageal resection at Nagoya University Hospital between October 2001 and January 2016 (19). The tumors were determined to be radically resected when pathologically diagnosed as stage I to III. All tissue samples were histologically diagnosed as ESCC, immediately frozen following resection and stored at −80°C. Specimens were histologically classified using the 7th edition of the UICC staging system for esophageal cancer (20). Patients who received neoadjuvant chemotherapy were excluded. Postoperative follow-up included physical examination, measurement of serum tumor markers every 3 months, and enhanced computed tomography of the chest and abdominal cavity every 6 months. Adjuvant chemotherapy was administered to selected patients according to their condition and at the discretion of the physician.

Analysis of CPNE5 mRNA expression levels

The expression levels of CPNE5 mRNA were measured using a reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay. Total RNA isolated using RNeasy Mini Kit (Qiagen GmbH, Hilden, Germany) from cell lines and 106 pairs of surgically-resected primary ESCCs and adjacent normal tissues served as template for cDNA synthesis. Reverse transcription was performed as follows: 10.5 µl 1 µg/µl RNA, 4 µl of 5X first strand buffer (Thermo Fisher Scientific, Inc., Waltham, MA, USA), 2 µl 100 mM dithiothreitol (Thermo Fisher Scientific, Inc.), 1 µl 10 mM dNTP mix (Promega Corporation, Madison, WI, USA), 1 µl random primer (Roche Diagnostics, Basel, Switzerland), 1 µl 200 U/µl Moloney murine leukemia virus reverse transcriptase (Thermo Fisher Scientific, Inc.) and 0.5 µl RNase inhibitor (Roche Diagnostics) were mixed and incubated for 60 min at 37°C. GAPDH mRNA expression levels (TaqMan; GAPDH control reagents; Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA) were quantified, and the data were used to normalize the expression levels. RT-qPCR was performed using the SYBR Green PCR Core Reagents kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) as follows: One cycle at 95°C for 10 min; 40 cycles at 95°C for 5 sec and 60°C for 60 sec without a final extension step. The samples were tested in triplicate, and samples without a template were included in each PCR plate as a negative control (21). Real-time SYBR Green fluorescence was detected using an ABI StepOnePlus Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) (22) and the 2−ΔΔCq method was used for PCR quantification (23). The expression level of each sample is expressed as the value of the CPNE5 amplicon divided by that of GAPDH. The sequences of the specific primers are listed in Table I.

Table I.

Primers and annealing temperatures.

Table I.

Primers and annealing temperatures.

GeneExperimentTypeSequence (5′-3′)Product size (bp)Annealing temperature
CPNE5RT-qPCRForward CATGTTTTCCAAGTCCGACC10660°C
Reverse ATTGAGCGTGTTGTCGATGA
Bisulfite sequencingForward GGTAGGAGTTTTTAGATTTGGAGGT17256°C
Reverse ATTTCCCAATAACCCAAATAAAATC
GAPDHRT-qPCRForward GAAGGTGAAGGTCGGAGTC22660°C
Probe CAAGCTTCCCGTTCTCAGCC
Reverse GAAGATGGTGATGGGATTTC

[i] CPNE5, copine 5; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; bp, base pairs.

Western blot analysis

The protein was extracted from each cell line using radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.) and protein concentration was determined using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA). For SDS-PAGE, 20 µg protein was added to a NuPAGE 4–12% Bis-Tris Gel (Thermo Fisher Scientific, Inc.) and electrophoresed for 35 min a 200 V. A polyvinylidene difluoride membrane was used for blotting and the membrane was blocked with 5% skim milk (Wako Pure Chemical Industries, Ltd., Osaka, Japan) for 60 min at room temperature. The CPNE5 protein expression levels in ESCC cell lines were evaluated with a rabbit anti-CPNE5 polyclonal antibody (1:100 dilution and overnight incubation at 4°C; cat. no. HPA031369; Atlas Antibodies AB, Bromma, Sweden) as a primary antibody and anti-rabbit IgG HRP-linked antibody (1:1,000 dilution and 60 min incubation at room temperature; cat. no. 7074S; Cell Signaling Technology, Inc., Danvers, MA, USA) as a secondary antibody. As an internal control, β-actin protein expression was detected with a mouse anti-β-actin polyclonal antibody (1:10,000 dilution and incubated for 60 min at room temperature; cat. no. ab6276; Abcam, Cambridge, UK) as a primary antibody and anti-mouse IgG HRP-linked antibody (1:1,000 dilution and 60 min incubation at room temperature; cat. no. 7076S; Cell Signaling Technology, Inc.) as a secondary antibody. Enhanced Chemiluminescence Western Blot Analysis System (GE Healthcare, Chicago, IL, USA) was used for visualization of the secondary antibody. An ESCC cell line with relatively high CPNE5 mRNA expression (KYSE590) and a low-expression ESCC cell line (TT) were evaluated.

Bisulfite nucleotide sequencing

Genomic DNA was isolated from the cell lines using a QIAamp DNA Mini kit (Qiagen GmbH) and treated with bisulfite (2 cycles at 95°C for 5 min and 60°C for 10 min). Bisulfite-modified DNA from ESCC cell lines and a non-cancerous esophageal mucosa cell line (Het-1A) were amplified as follows: One cycle at 94°C for 2 min; and 50 cycles at 94°C for 15 sec, 56°C for 15 sec and 72°C for 30 sec, using specific primers (Table I). Sequencing was performed by Eurofins Genomics Tokyo Co., Ltd. (Tokyo, Japan), using a Big Dye Terminator v3.1 Cycle Sequencing kit (Thermo Fisher Scientific, Inc.) and a 3730× l DNA Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.) (24).

Immunohistochemistry

Immunohistochemical staining was performed to determine the difference in CPNE5 protein expression between cancerous tissue and non-cancerous tissues in 45 representative clinical cases. Sections were incubated for 16 h at 4°C with a rabbit polyclonal antibody raised against CPNE5 (cat. no. HPA031369; Atlas Antibodies AB) diluted 1:100 in Antibody Diluent (Dako; Agilent Technologies, Inc., Santa Clara, CA, USA). Sections were incubated with secondary antibody (SignalStain® Boost IHC Detection Reagent labelled with HRP; Cell Signaling Technology, Inc.) for 30 min at room temperature. Antigen antibody complexes were visualized by exposure with liquid 3,3′-diaminobenzidine (Nichirei, Tokyo, Japan) for 2 min. A total of two independent observers evaluated the specimens using an optical microscope with ×400 magnification as follows: Cancerous tissue >non-cancerous tissue; equivalent; or cancerous tissue <non-cancerous tissue.

Statistical analysis

Quantitative data are presented as the mean ± standard deviation. Patients were divided into low and high CPNE5 groups according to the median levels of CPNE5 mRNA expression in the cancerous tissues. The differences between CPNE5 mRNA expression values in the two groups (differentiated cell lines vs. undifferentiated cell lines, or cancerous tissues vs. non-cancerous tissues) were compared using the Mann-Whitney test. The χ2 test was used to analyze the association between CPNE5 mRNA expression levels and clinicopathological characteristics. Overall survival (OS) and disease-free survival (DFS) rates were calculated using the Kaplan-Meier method. The Cox proportional hazards model was used to compare survival rates, and multivariable regression analysis was used to identify prognostic factors. Statistical analysis was performed using JMP 10 software (SAS Institute Inc., Cary, NC, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Expression levels of CPNE5 and promoter methylation in ESCC cell lines

CPNE5 mRNA expression levels differed among the 11 ESCC cell lines (Fig. 1A), and were lower compared with those of Het-1A cells, except for KYSE590 and KYSE1440. There were no significant differences in CPNE5 mRNA expression levels between differentiated (0.00197±0.00172) and undifferentiated (0.00161±0.00133) cell lines (P=0.897). ESCC cell lines established from metastatic sites, including TT and TTn, and those established from lymph node metastases, including KYSE1170 and KYSE1260, expressed low levels of CPNE5 mRNA. Western blot analysis using an anti-CPNE5 antibody illustrated high CPNE5 protein expression in an ESCC cell line with high CPNE5 mRNA expression (KYSE590) and low CPNE5 protein expression in a low-expression ESCC cell line (TT) (Fig. 1B). Bisulfite sequencing analysis of CPNE5 revealed that the CpG sites in the CPNE5 DNA promotor region in all ESCC cell lines and Het-1A were completely methylated. The bisulfite sequencing results for Het-1A, KYSE590 and TT cells are presented as representative cell lines expressing high and low levels of CPNE5 mRNA, respectively (Fig. 1C).

Characteristics of patients with ESCC

The median age of the 106 patients was 65 years (range, 44–84 years), and the female: Male ratio was 20:86. According to the UICC staging system (7th edition), 24, 29 and 53 patients were diagnosed with disease at pathological stages I, II and III, respectively. Adjuvant chemotherapy was administered to 36 patients (34%). The median duration of follow-up was 34.1 months, during which 44 patients (42%) experienced recurrence and 39 patients (37%) succumbed to the disease.

CPNE5 mRNA expression levels in clinical samples

The mean normalized CPNE5 mRNA expression level was significantly lower in ESCC tissues (0.0107±0.0138) compared with the corresponding non-cancerous adjacent mucosal tissues (0.00829±0.0123; P=0.003; Fig. 2A).

Association between levels of CPNE5 mRNA and the clinicopathological characteristics of patients who underwent resection

There was no significant association between the low and high CPNE5 expression groups with their clinicopathological characteristics (sex, tumor size and depth, lymphatic involvement, vascular invasion and pathological stage). By contrast, the percentage of patients who received adjuvant chemotherapy was significantly higher in the low CPNE5 group (Table II).

Table II.

Association between the expression level of CPNE5 mRNA and clinicopathological parameters in 106 patients with resected esophageal cancer.

Table II.

Association between the expression level of CPNE5 mRNA and clinicopathological parameters in 106 patients with resected esophageal cancer.

Clinicopathological parametersLow CPNE5 in ESCC tissue, no. of patientsHigh CPNE5 in ESCC tissue, no. of patientsP-value
Age, years 0.437
  <652328
  ≥653025
Sex 1.000
  Male4343
  Female1010
Smoking history 0.487
  Yes3943
  No1410
Double cancer 0.698
  Present710
  Absent4643
Tumor location 0.111
  Ce01
  Ut35
  Mt2427
  Lt2515
  Ae15
Tumor multiplicity 1.000
  Present78
  Absent4645
Tumor size, mm 0.151
  <503139
  ≥502214
CEA, ng/ml 0.761
  ≤54846
  >557
SCC, IU/ml 0.531
  ≤1.53633
  >1.51519
pT 0.116
  T1 or 21827
  T33526
Lymph node metastasis 0.843
  Present3133
  Absent2220
Differentiation 0.267
  Differentiated4345
  Undifferentiated105
Lymphatic involvement 0.822
  Present3941
  Absent1412
Vascular invasion 1.000
  Present2122
  Absent3231
Intraepithelial progress 0.057
  Present1822
  Absent2310
Pathological UICC stage 0.466
  I1014
  II1712
  III2627
Postoperative adjuvant chemotherapy 0.007
  Present2511
  Absent2842

[i] CPNE5, copine 5; Ce, cervical esophagus; Ut, Upper thoracic esophagus; Mt, Middle thoracic esophagus; Lt, lower thoracic esophagus, Ae, abdominal esophagus; CEA, carcinoembryonic antigen; SCC, squamous cell carcinoma-related antigen; UICC, Union for International Cancer Control; pT, tumor depth.

Ability of CPNE5 mRNA expression levels to predict prognosis

Patients in the low CPNE5 expression group experienced a significantly shorter OS time compared with those in the high CPNE5 expression group (the 5-year OS rates were 55.6 and 77.3% for the low and high expression groups, respectively; P=0.016; Fig. 2B). Though the difference was not statistically significant, there was a similar trend observed in DFS (P=0.052; Fig. 2C). Univariate analysis revealed that tumor depth, lymph node metastasis, undifferentiated tumor phenotype, lymphatic involvement, postoperative chemotherapy and low CPNE5 expression were significantly associated with lower survival rates. Multivariable analysis identified low CPNE5 expression and lymphatic involvement as independent prognostic factors for OS (hazard ratio, 2.55; 95% confidence interval, 1.21–5.68; P=0.014; and hazard ratio, 6.28; 95% confidence interval, 1.70–40.6; P=0.004, respectively; Table III).

Table III.

Prognostic factors for overall survival of 106 patients.

Table III.

Prognostic factors for overall survival of 106 patients.

UnivariateMultivariable


Hazard ratio95% CIP-valueHazard ratio95% CIP-value
Age, ≥65 years1.450.75–2.790.259
Sex, male2.190.94–6.430.072
Smoking0.980.49–2.200.963
Double cancer1.290.52–2.760.559
Tumor multiplicity0.700.21–1.740.470
Tumor size, ≥50 mm1.280.66–2.410.462
CEA, >5 ng/ml1.340.50–2.970.528
SCC, >1.5 IU/ml1.110.55–2.140.769
Tumor depth, pT32.751.38–5.960.0031.480.72–3.270.293
Lymph node metastasis2.361.19–5.090.0131.530.64–3.920.349
Tumor differentiation, undifferentiated2.751.22–5.620.0022.100.92–4.370.075
Lymphatic involvement8.612.63–53.0<0.0016.281.70–40.60.004a
Vascular invasion1.390.73–2.610.304
Intraepithelial progress0.640.32–1.270.204
Postoperative adjuvant chemotherapy2.081.11–3.930.0230.920.42–2.040.837
Low CPNE5 expression2.251.17–4.620.0152.551.21–5.680.014a

a Statistically significant in multivariable analysis. CI, confidence interval; CEA, carcinoembryonic antigen; SCC, squamous cell carcinoma-related antigen; UICC, Union for International Cancer Control; CPNE5, copine 5.

Analysis of patients with recurrence

Differences between recurrence patterns were predicted, since the Kaplan-Meier analysis revealed fold-differences between OS and DFS. Among patients with recurrence, a significant number were members of the low CPNE5 group (P=0.018). Among patients with local recurrence, significantly more were members of the low CPNE5 group (P=0.027; Fig. 2D). A similar tendency was observed in patients with lymph node recurrence (P=0.250; Fig. 2D). By contrast, the rates of distant metastasis to tissues including the lung/pleura and liver were not significantly different between groups (Fig. 2D).

CPNE5 protein expression levels in clinical samples

The expression patterns of CPNE5 protein were evaluated using immunohistochemical staining. Among the 45 clinical samples, CPNE5 protein expression was suppressed in the cancerous tissue in 14 (31%) samples, equally expressed in cancerous and non-cancerous tissue in 19 (42%) samples and overexpressed in cancerous tissue in 12 (27%) samples. Representative examples of suppression in cancerous tissue, and an example of equivalent expression, are presented (Fig. 3).

Discussion

The results of the present study provided evidence to support the predictive value of CPNE5 expression levels in ESCC tissues following curative resection. Specifically, the CPNE5 mRNA expression levels of the majority ESCC cell lines were lower compared with those of a non-tumorigenic esophageal cell line. Bisulfite sequencing analysis was performed to reveal the mechanism of downregulation of CPNE5 transcription, as the promoter region harbors CpG islands (25,26). Methylation of the CPNE5 promoter region was detected in all ESCC cell lines and in a non-tumorigenic esophageal cell line, indicating that promoter hypermethylation did not contribute to the regulation of CPNE5 transcription. Acetylation of histones (27), copy-number alterations (28), microRNAs (27) and genomic mutations (29) may therefore contribute to the downregulation of CPNE5 transcription.

The expression levels of CPNE5 mRNA in ESCC tissues were lower compared with those of adjacent non-cancerous tissue, revealing an association between CPNE5 expression and the pathology of ESCC. CPNE5 mRNA expression levels were not significantly associated with clinicopathological characteristics known to be associated with an unfavorable prognosis of ESCC [including tumor multiplicity, tumor size, tumor markers, lymphatic involvement, vascular invasion, and UICC stage (30)], although low levels of CPNE5 mRNA were associated with shorter OS. Furthermore, multivariable analysis identified low levels of CPNE5 mRNA as an independent risk factor of shorter OS. Therefore, CPNE5 may serve as an effective marker for predicting prognosis compared with the tumor-node-metastasis classification of esophageal cancer.

CPNE5 mRNA expression levels were lower in patients who received adjuvant chemotherapy, although the administration of chemotherapy was at the physician's discretion. The association between adjuvant chemotherapy and CPNE5 expression may be explained by the association, albeit not statistically significant, of CPNE5 mRNA expression levels with tumor size and depth, in addition to intraepithelial progression, and physicians may therefore decide to administer adjuvant chemotherapy according to their interpretations of the totality of pathological findings. However, it was noted that patients in the low CPNE5 expression group had poor prognoses despite adjuvant chemotherapy (31), suggesting an association between CPNE5 expression and resistance to chemotherapy.

OS and DFS rates following curative resection of ESCC were lower in the low CPNE5 group compared with the high CPNE5 group. The difference in OS was statistically significant, although that for DFS was not. The Kaplan-Meier analysis demonstrated that the primary curves for DFS of the two groups overlapped. By contrast, the primary curves of OS were separated, and this difference may reflect the statistical difference. These results demonstrated that the low CPNE5 group experienced shorter survival following recurrence. Accordingly, two hypotheses were developed to explain the data as follows: i) The clinicopathological findings indicate resistance to chemotherapy; thus, patients in the low CPNE5 group may succumb following recurrence, as they did not benefit from chemotherapy; and ii) the differences in recurrence patterns may explain survival patterns; it was expected that recurrence in the low CPNE5 group may arise in a site that is difficult to treat, for example the lung or liver, which is associated with poor prognosis following recurrence (32). The analysis of recurrence patterns, which was conducted to evaluate these hypotheses, revealed that significantly more patients in the low CPNE5 group experienced more frequent recurrence compared with those in the high CPNE5 group. In contrast to expectations, the difference in the numbers of patients in the low and high CPNE5 groups was not significant for patients with lung and hepatic recurrences, which are associated with poor prognosis following recurrence.

However, it was noted that the differences in recurrence rates between the low and high CPNE5 groups was explained by local and lymphatic recurrences, indicating that CPNE5 expression may be associated with the local growth of esophageal cancer. To translate these findings into clinical practice, patients with low CPNE5 expression ought to undergo surgery with rigorous lymph node dissection (33) and adequate surgical margins (34). Frequent follow-up, including esophagoscopy (35) and computed tomography (36), may enhance the detection of local recurrence.

There are certain limitations to the present study. First, an association between CPNE5 expression and resistance to chemotherapy was identified. Unfortunately, it was not possible to obtain details of patients who received chemotherapy following recurrence. The analysis of biopsied or micro-dissected resected tissues from patients who have received neoadjuvant chemotherapy may help to investigate this further. Second, this was a retrospective study of a small number patients treated at a single center. External validation using large cohorts from multiple institutions is required to validate the present findings. Third, the function of CPNE5 in esophageal cancer and its mechanism of regulation remain unexplained. Functional analysis of CPNE5-knockdown cell lines, protein expression and animal tumor xenograft models are required to overcome these limitations.

In conclusion, the present data suggested that CPNE5 expression in ESCC tissues may represent a promising biomarker for predicting ESCC recurrence, particularly for patients with local recurrence, and may help ensure that patients receive optimal treatment and follow-up.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

SU, MKa, TM and HT performed the experiments and the data analysis. SU, MKa, TM, HT, CT, DK, MKo, MS, MH, SY, GN and YK collected the cases and the clinical data. SU and MKa conceived and designed the study and prepared the initial manuscript. YK supervised the project. All authors contributed to the final manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study conformed to the ethical guidelines of the World Medical Association Declaration of Helsinki, Ethical Principles for Medical Research Involving Human Subjects, and was approved by the Institutional Review Board of Nagoya University (Nagoya, Japan). Written informed consent for the use of clinical samples and data, as required by the institutional review board, was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lagergren J, Smyth E, Cunningham D and Lagergren P: Oesophageal cancer. Lancet. 390:2383–2396. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, Bonaventure A, Valkov M, Johnson CJ, Estève J, et al: Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): Analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 391:1023–1075. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Tanaka H, Kanda M, Koike M, Iwata N, Shimizu D, Ezaka K, Sueoka S, Tanaka Y, Takami H, Hashimoto R, et al: Adherens junctions associated protein 1 serves as a predictor of recurrence of squamous cell carcinoma of the esophagus. Int J Oncol. 47:1811–1818. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Xi M, Yang Y, Zhang L, Yang H, Merrell KW, Hallemeier CL, Shen RK, Haddock MG, Hofstetter WL, Maru DM, et al: Multi-institutional analysis of recurrence and survival after neoadjuvant chemoradiotherapy of esophageal cancer: Impact of histology on recurrence patterns and outcomes. Ann Surg. 2018. View Article : Google Scholar

5 

Chen HS, Hsu PK, Liu CC and Wu SC: Upfront surgery and pathological stage-based adjuvant chemoradiation strategy in locally advanced esophageal squamous cell carcinoma. Sci Rep. 8:21802018. View Article : Google Scholar : PubMed/NCBI

6 

Hibino S, Kanda M, Oya H, Takami H, Shimizu D, Nomoto S, Hishida M, Niwa Y, Koike M, Yamada S, et al: Reduced expression of DENND2D through promoter hypermethylation is an adverse prognostic factor in squamous cell carcinoma of the esophagus. Oncol Rep. 31:693–700. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Kunzmann AT, McMenamin ÚC, Spence AD, Gray RT, Murray LJ, Turkington RC and Coleman HG: Blood biomarkers for early diagnosis of oesophageal cancer: A systematic review. Eur J Gastroenterol Hepatol. 30:263–273. 2018.PubMed/NCBI

8 

Tripodis N, Mason R, Humphray SJ, Davies AF, Herberg JA, Trowsdale J, Nizetic D, Senger G and Ragoussis J: Physical map of human 6p21.2-6p21.3: Region flanking the centromeric end of the major histocompatibility complex. Genome Res. 8:631–643. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Creutz CE, Tomsig JL, Snyder SL, Gautier MC, Skouri F, Beisson J and Cohen J: The copines, a novel class of C2 domain-containing, calcium-dependent, phospholipid-binding proteins conserved from Paramecium to humans. J Biol Chem. 273:1393–1402. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, et al: Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 13:397–406. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Ding X, Jin Y, Wu Y, Wu Y, Wu H, Xiong L, Song X, Liu S, Fan W and Fan M: Localization and cellular distribution of CPNE5 in embryonic mouse brain. Brain Res. 1224:20–28. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Wang KS, Zuo L, Pan Y, Xie C and Luo X: Genetic variants in the CPNE5 gene are associated with alcohol dependence and obesity in Caucasian populations. J Psychiatr Res. 71:1–7. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Kidger AM, Sipthorp J and Cook SJ: ERK1/2 inhibitors: New weapons to inhibit the RAS-regulated RAF-MEK1/2-ERK1/2 pathway. Pharmacol Ther. 187:45–60. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Hsieh FS, Hung MH, Wang CY, Chen YL, Hsiao YJ, Tsai MH, Li JR, Chen LJ, Shih CT, Chao TI, et al: Inhibition of protein phosphatase 5 suppresses non-small cell lung cancer through AMP-activated kinase activation. Lung Cancer. 112:81–89. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Schlessinger K, McManus EJ and Hall A: Cdc42 and noncanonical Wnt signal transduction pathways cooperate to promote cell polarity. J Cell Biol. 178:355–361. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Tomsig JL, Snyder SL and Creutz CE: Identification of targets for calcium signaling through the copine family of proteins. Characterization of a coiled-coil copine-binding motif. J Biol Chem. 278:10048–10054. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Tsunoo H, Komura S, Ohishi N, Yajima H, Akiyama S, Kasai Y, Ito K, Nakao A and Yagi K: Effect of transfection with human interferon-beta gene entrapped in cationic multilamellar liposomes in combination with 5-fluorouracil on the growth of human esophageal cancer cells in vitro. Anticancer Res. 22:1537–1543. 2002.PubMed/NCBI

18 

Shimada Y, Imamura M, Wagata T, Yamaguchi N and Tobe T: Characterization of 21 newly established esophageal cancer cell lines. Cancer. 69:277–284. 1992. View Article : Google Scholar : PubMed/NCBI

19 

Miwa T, Kanda M, Koike M, Iwata N, Tanaka H, Umeda S, Tanaka C, Kobayashi D, Hayashi M, Yamada S, et al: Identification of NCCRP1 as an epigenetically regulated tumor suppressor and biomarker for malignant phenotypes of squamous cell carcinoma of the esophagus. Oncol Lett. 14:4822–4828. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Sobin LH, Gospodarowicz MK and Wittekind Ch: International Union Against CancerTNM Classification of Malignant Tumours. 7th edition. Wiley-Blackwell; New York, NY: pp. 66–72. 2009

21 

Kanda M, Shimizu D, Sueoka S, Nomoto S, Oya H, Takami H, Ezaka K, Hashimoto R, Tanaka Y, Kobayashi D, et al: Prognostic relevance of SAMSN1 expression in gastric cancer. Oncol Lett. 12:4708–4716. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Shimizu D, Kanda M, Tanaka H, Kobayashi D, Tanaka C, Hayashi M, Iwata N, Niwa Y, Takami H, Yamada S, et al: GPR155 serves as a predictive biomarker for hematogenous metastasis in patients with gastric cancer. Sci Rep. 7:420892017. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Kanda M, Tanaka C, Kobayashi D, Tanaka H, Shimizu D, Shibata M, Takami H, Hayashi M, Iwata N, Niwa Y, et al: Epigenetic suppression of the immunoregulator MZB1 is associated with the malignant phenotype of gastric cancer. Int J Cancer. 139:2290–2298. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Beck S, Rhee C, Song J, Lee BK, LeBlanc L, Cannon L and Kim J: Implications of CpG islands on chromosomal architectures and modes of global gene regulation. Nucleic Acids Res. 46:4362–4391. 2018. View Article : Google Scholar

26 

Pu W, Wang C, Chen S, Zhao D, Zhou Y, Ma Y, Wang Y, Li C, Huang Z, Jin L, et al: Targeted bisulfite sequencing identified a panel of DNA methylation-based biomarkers for esophageal squamous cell carcinoma (ESCC). Clin Epigenetics. 9:1292017. View Article : Google Scholar : PubMed/NCBI

27 

Kailasam A, Mittal SK and Agrawal DK: Epigenetics in the pathogenesis of esophageal adenocarcinoma. Clin Transl Sci. 8:394–402. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Dong G, Mao Q, Yu D, Zhang Y, Qiu M, Dong G, Chen Q, Xia W, Wang J, Xu L, et al: Integrative analysis of copy number and transcriptional expression profiles in esophageal cancer to identify a novel driver gene for therapy. Sci Rep. 7:420602017. View Article : Google Scholar : PubMed/NCBI

29 

Chen XX, Zhong Q, Liu Y, Yan SM, Chen ZH, Jin SZ, Xia TL, Li RY, Zhou AJ, Su Z, et al: Genomic comparison of esophageal squamous cell carcinoma and its precursor lesions by multi-region whole-exome sequencing. Nat Commun. 8:5242017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang D, Zheng Y, Wang Z, Huang Q, Cao X, Wang F and Liu S: Comparison of the 7th and proposed 8th editions of the AJCC/UICC TNM staging system for esophageal squamous cell carcinoma underwent radical surgery. Eur J Surg Oncol. 43:1949–1955. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Zhang SS, Yang H, Xie X, Luo KJ, Wen J, Bella AE, Hu Y, Yang F and Fu JH: Adjuvant chemotherapy versus surgery alone for esophageal squamous cell carcinoma: A meta-analysis of randomized controlled trials and nonrandomized studies. Dis Esophagus. 27:574–584. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Wu SG, Zhang WW, He ZY, Sun JY, Chen YX and Guo L: Sites of metastasis and overall survival in esophageal cancer: A population-based study. Cancer Manag Res. 9:781–788. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Guo JC, Lin CC, Huang TC, Huang PM, Kuo HY, Chang CH, Wang CC, Cheng JC, Yeh KH, Hsu CH, et al: Number of resected lymph nodes and survival of patients with locally advanced esophageal squamous cell carcinoma receiving preoperative chemoradiotherapy. Anticancer Res. 38:1569–1577. 2018.PubMed/NCBI

34 

Tam PC, Siu KF, Cheung HC, Ma L and Wong J: Local recurrences after subtotal esophagectomy for squamous cell carcinoma. Ann Surg. 205:189–194. 1987. View Article : Google Scholar : PubMed/NCBI

35 

Catalano MF, Sivak MV Jr, Rice TW and Van Dam J: Postoperative screening for anastomotic recurrence of esophageal carcinoma by endoscopic ultrasonography. Gastrointest Endosc. 42:540–544. 1995. View Article : Google Scholar : PubMed/NCBI

36 

Kim TJ, Lee KH, Kim YH, Sung SW, Jheon S, Cho SK and Lee KW: Postoperative imaging of esophageal cancer: What chest radiologists need to know. Radiographics. 27:409–429. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 Issue 6

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
Umeda S, Kanda M, Koike M, Tanaka H, Miwa T, Tanaka C, Kobayashi D, Suenaga M, Hayashi M, Yamada S, Yamada S, et al: Copine 5 expression predicts prognosis following curative resection of esophageal squamous cell carcinoma. Oncol Rep 40: 3772-3780, 2018
APA
Umeda, S., Kanda, M., Koike, M., Tanaka, H., Miwa, T., Tanaka, C. ... Kodera, Y. (2018). Copine 5 expression predicts prognosis following curative resection of esophageal squamous cell carcinoma. Oncology Reports, 40, 3772-3780. https://doi.org/10.3892/or.2018.6742
MLA
Umeda, S., Kanda, M., Koike, M., Tanaka, H., Miwa, T., Tanaka, C., Kobayashi, D., Suenaga, M., Hayashi, M., Yamada, S., Nakayama, G., Kodera, Y."Copine 5 expression predicts prognosis following curative resection of esophageal squamous cell carcinoma". Oncology Reports 40.6 (2018): 3772-3780.
Chicago
Umeda, S., Kanda, M., Koike, M., Tanaka, H., Miwa, T., Tanaka, C., Kobayashi, D., Suenaga, M., Hayashi, M., Yamada, S., Nakayama, G., Kodera, Y."Copine 5 expression predicts prognosis following curative resection of esophageal squamous cell carcinoma". Oncology Reports 40, no. 6 (2018): 3772-3780. https://doi.org/10.3892/or.2018.6742