Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy

  • Authors:
    • Susumu Saigusa
    • Koji Tanaka
    • Masaki Ohi
    • Yuji Toiyama
    • Hiromi Yasuda
    • Takahito Kitajima
    • Yoshinaga Okugawa
    • Yasuhiro Inoue
    • Yasuhiko Mohri
    • Masato Kusunoki
  • View Affiliations

  • Published online on: September 26, 2014     https://doi.org/10.3892/mco.2014.431
  • Pages: 151-156
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recent epidemiological studies demonstrated that the incidence of esophageal squamous cell carcinoma (ESCC) is on the increase. Although neoadjuvant chemoradiotherapy (CRT) followed by surgery may improve long‑term survival and reduce local recurrence in patients with esophageal cancer, the overall cure rate of esophageal cancer is low. Fas/Fas ligand (FasL) signaling initiates the cell death pathway. The roles of FasL in tumor growth, progression and resistance to treatment have been demonstrated in several malignancies. The aim of this preliminary study was to evaluate Fas/FasL expression in ESCC with neoadjuvant CRT. A total of 20 patients who received neoadjuvant CRT (30‑40 Gy; 5‑fluorouracil plus cisplatin followed by surgery) were enrolled. We evaluated the expression of Fas, FasL and Ki67 (a proliferative marker) using immunohistochemistry and analyzed the correlations between their expression and clinical outcomes. Additionally, we investigated the association of Fas/FasL expression with peritumoral immune CD8‑positive and Foxp3‑positive cells. High FasL expression was significantly correlated with disease recurrence (P=0.0134). Patients with high FasL expression exhibited poorer recurrence‑free and overall survival (P=0.0102 and 0.0385, respectively). Patients with low Fas and high FasL exhibited significantly poorer recurrence‑free survival (P=0.0035). Although statistical significance was not reached, Fas expression appeared to be inversely correlated with Foxp3‑positive cells and FasL expression appeared to be inversely correlated with CD8‑positive cells. In conclusion, FasL expression was associated with tumor relapse and poor prognosis in patients with ESCC following CRT. Pharmacological control of Fas/FasL signaling may improve therapeutic efficacy and outcome in ESCC patients receiving preoperative CRT.

Introduction

Esophageal squamous cell carcinoma (ESCC) is a highly malignant tumor and its prognosis is generally poor. Recent epidemiological studies demonstrated that the incidence of ESCC is on the increase (1). Neoadjuvant chemoradiotherapy (CRT) followed by surgery may improve long-term survival and reduce local recurrence in patients with esophageal cancer. However, the overall cure rate of esophageal cancer remains <20% (24).

Fas and Fas ligand (FasL) are transmembrane proteins that belong to the tumor necrosis factor family and their signaling pathway is a key regulator of apoptotic cell death, i.e., Fas binding of FasL induces an apoptotic cascade. Furthermore, the Fas/FasL pathway regulates the tumor microenvironment, including the host immune system and extracellular matrix (512). Several authors have reported that Fas/FasL expression is correlated with tumor progression and poor prognosis in esophageal cancer (6,13,14). By contrast, Takikita et al (15) reported that the Fas/FasL apoptotic pathway, including Fas-associated death domain protein and caspases 8 and 10 were not prognostic factors in ESCC.

In the present study, we evaluated the expression of Fas, FasL and Ki67 (a proliferation marker) in ESCC following neoadjuvant CRT and analyzed the correlation of their expression with clinical outcome. Additionally, the association of Fas/FasL expression with peritumoral immune cells was investigated.

Materials and methods

Patients and specimens

A total of 20 patients who had received neoadjuvant CRT followed by surgery were enrolled in this study. All the formalin-fixed, paraffin-embedded (FFPE) specimens of the patients were available for evaluation. The study protocol was approved by the Ethics Review Board of Mie University Hospital and all the included patients provided written informed consent for their tissues to be used in this study.

5-Fluorouracil (5-FU) and cisplatin (CDDP)-based CRT

All the patients received systemic 5-FU and CDDP chemotherapy with concurrent radiotherapy. The regimen included 4 cycles of 5-FU (600 mg/m2 administered intravenously over 24 h), plus tegafur and uracil (400 mg/kg of body weight administered orally for 5 days) and CDDP (4 mg/day administered intravenously for 5 days) with concurrent 40 Gy radiation followed by surgery. Preoperative radiotherapy was delivered to the primary tumor as well as the peritumoral area at a dose of 40 Gy in 20 fractions within 4 weeks. The time interval between neoadjuvant CRT and surgery was 2–3 weeks.

Clinical response and histopathological analysis of tumor regression following CRT

The clinical response following preoperative CRT was evaluated by barium esophagography, endoscopy and computed tomography. The results were graded as complete response (CR), partial response (PR), no change (NC) or progressive disease. The pathological response to CRT was evaluated using the Mandard tumor regression grade (TRG) (16). The tumors were classified according to the Mandard system into 5 grades as follows: i) TRG1, CR with absence of residual cancer and fibrosis extending through the wall; ii) TRG2, presence of residual tumor cells scattered through the fibrotic area; iii) TRG3, increased numbers of residual cancer cells, with predominant fibrosis; iv) TRG4, residual cancer outgrowing the fibrotic area; v) TRG5, absence of regressive changes. We categorized patients in categories TRG1 and 2 as responders and those in categories TRG3-5 as non-responders.

Immunohistochemistry (IHC)

The FFPE specimens were cut into 2- to 3-µm sections. Following deparaffinization and dehydration, the sections were placed in a 10 mmol/l sodium citrate buffer (pH 6.0) and autoclaved at 121°C for 10 min for antigen retrieval. The sections were incubated in 3% hydrogen peroxide for 10 min, blocked and incubated with a primary antibody overnight at 4°C. Monoclonal mouse anti-human Fas antibody (B-10, catalog no. sc-8009; Santa Cruz Biotechnology, Santa Cruz, CA, USA; dilution 1:100), polyclonal rabbit anti-human FasL antibody (C-20, catalog no. sc-957; Santa Cruz Biotechnology; dilution 1:100) and monoclonal mouse anti-human Ki67 antibody (MIB-1, code M7240; Dako Cytomation, Glostrup, Denmark; dilution 1:100) were the primary antibodies used in a labeled streptavidin-biotin system (EnVision™ + Dual Link System-HRP; Dako Cytomation). Antibody binding was visualized using 3,3′-diaminobenzidine (Dako Cytomation). All the sections were counterstained with hematoxylin prior to being dehydrated and mounted. At least 2 sections per specimen were stained to confirm reproducibility. Negative controls were prepared simultaneously with pre-immune immunoglobulin.

IHC evaluation

The sections were observed under a light microscope (BX50, Olympus, Tokyo, Japan). We calculated IHC scores by multiplying the percentage of positive epithelial cells (0–100%) by the staining intensity, as previously described (17). The staining intensities were scored as follows: 0, negative; 1, weak; and 2, strong for Fas and Ki67; and 0, negative; 1, weak; 2, moderate; and 3, strong for FasL. The IHC scores ranged between 0 and 200 for Fas and Ki67; and between 0 and 300 for FasL. Each sample was scored by two investigators (K.T. and Y.O.) who were blinded to the clinicopathological information regarding the origin of the samples.

Enumeration of peritumoral CD8- and Foxp3-positive cells

We previously recorded the numbers of peritumoral CD8- and Foxp3-positive cells using light microscopy and the median count was calculated for each sample. The median values for the number of CD8- and Foxp3-positive cells were 70 (range, 18–250) at a magnification x200 and 13 (range, 1–57) at a magnification x100 (18). We analyzed the correlation of Fas/FasL expression with peritumoral CD8- and Foxp3-positive cells.

Statistical analysis

Statistical analysis was performed using StatView v5.0 software (SAS Institute Inc., Cary, NC, USA). Significant differences were analyzed using the Chi-square test. The Pearson's correlation test was used to determine statistical correlations. Recurrence-free and overall survival probabilities were calculated from the date of surgery to the date of recurrence or death, using the Kaplan-Meier product limit method; intergroup differences were determined using the log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

Patient characteristics

The patient characteristics are summarized in Table I. The median age of the patients was 68 years (range, 52–77 years) and the male:female ratio was 9:1. The median follow-up period was 16 months (range, 4–91 months). Of the 20 tumors, 12 were located in the lower, 7 in the middle and 1 in the upper part of the esophagus. The mean size of the tumors was 40 mm (range, 20–82 mm). The post-CRT pathological T stages were as follows: pT1 (n=1), pT2 (n=9), pT3 (n=9) and pT4 (n=1). A total of 10 patients (50%) presented with lymph node metastases. The median number of total dissected lymph nodes was 14 (range, 0–39) and the median value of lymph node density was 5.45% (range, 0–55.6%). A total of 17 tumors (85%) exhibited well or moderately differentiated squamous cell carcinoma histology. R0 resection was performed in 80% of the cases. The clinical response was classified as follows: NC, 9 patients; PR, 10 patients; and CR, 1 patient. The Mandard TRG grades were as follows: TRG1, no patients; TRG2, 9 patients; TRG3, 7 patients; TRG4, 4 patients; and TRG5, no patients (non-responders, n=11; responders, n=9).

Table I

Patient characteristics (n=20).

Table I

Patient characteristics (n=20).

CharacteristicsValues (%)
Age, years
  Mediam  69
Range52–77
Gender
  Male18 (90)
  Female   2(10)
Location
  Upper  1 (5)
  Middle  7 (35)
  Lower12 (60)
ypT
  T1/210 (50)
  T3/410 (50)
ypN
  Absent10 (50)
  Present10 (50)
Postoperative stage
  I  1 (5)
  II  9 (45)
  III  7 (35)
  IV  3 (15)
Lymphatic invasion
  Absent  4 (20)
  Present16 (80)
Vascular invasion
  Absent  12 (60)
  Present  8 (40)
Histological differentiation
  High/moderate  17 (85)
  Poor  3 (15)
R0 resection
  Yes  16 (80)
  No  4 (20)
Mandard TRG
  5  0 (0)
  4  4 (20)
  3  7 (35)
  2  9 (45)
  1  0 (0)
Recurrence(R0, n=16)
  Absent10 (62.5)
  Present  6 (37.5)

[i] TRG, tumor regression grade.

IHC findings for Fas, FasL and Ki67 in ESCC following neoadjuvant CRT

Fig. 1 shows the IHC findings for Fas, FasL and Ki67 in ESCC following neoadjuvant CRT. Fas and FasL were expressed in the cytoplasm and nuclei of cancer cells. Additionally, FasL expression was diffusely detected in stromal cells. Ki67 expression was detected in cancer cell nuclei. There was no correlation between the IHC findings for Fas/FasL and Ki67. The median values of the IHC scores for Fas, FasL and Ki67 were 80 (range, 1–200), 190 (range, 30–300) and 10 (range, 1–180), respectively. We classified those cases with values above the median IHC score as the high-expression group and the remainder as the low-expression group.

Correlations of Fas, FasL and Ki67 expression with clinicopathological variables

High FasL expression was significantly correlated with disease recurrence in patients treated with curative intent (P=0.0134). However, there were no correlations between Fas/FasL expression and other clinicopathological characteristics (Table II). High Ki67 expression was significantly correlated with lymph node metastasis and lymphatic invasion (P=0.007 and 0.025, respectively) (data not shown).

Table II

Correlations of Fas, Fas ligand (FasL) and Ki67 expression with clinicopathological variables.

Table II

Correlations of Fas, Fas ligand (FasL) and Ki67 expression with clinicopathological variables.

VariablesFas-lowFas-highP-valueaFasL-lowFasL-highP-valuea
Median age, years
  ≤69640.371460.371
  >694664
Gender
  Male8100.136990.999
  Female2011
Location
 Upper100.270100.565
 Middle2534
 Lower7566
Size, mm
  <40680.329770.999
  ≥404233
ypT
  T0/1/2730.074460.371
  T3/43764
Lymph node metastasis
  Absent370.0746640.371
  Present7346
Postoperative stage
  0/I/II550.999640.371
  III/IV5546
Lymphatic invasion
  Absent130.264310.264
  Present9779
Vascular invasion
  Absent660.999660.999
  Present4444
Histological differentiation
  High/moderate980.531980.531
  Poor1212
R0 resection
  Yes970.264970.264
  No1313
Pathological response
  Non-responders650.653560.653
  Responders4554
Recurrence (R0, n=16)0.09070.0134
  Absent4682
  Present5115

a Chisquare test. Bold print denotes statistical significance.

Survival analysis based on Fas and FasL expression

The recurrence-free survival and overall survival according to Fas and FasL expression using the Kaplan-Meier product limit method are shown in Fig. 2. High expression of FasL was found to be significantly associated with poor recurrence-free and overall survival (P=0.0102 and 0.0385, respectively). Patients with low Fas and high FasL expression exhibited a poorer recurrence-free survival (P=0.0035).

Correlation of Fas/FasL expression with peritumoral CD8- and Foxp3-positive cells

Although the differences did not achieve statistical significance, Fas expression was inversely correlated with peritumoral Foxp3-positive cells, whereas FasL expression appeared to be inversely correlated with peritumoral CD8-positive cells and directly correlated with Foxp3-positive cells (Fig. 3).

Discussion

In esophageal cancer, upregulation of FasL and downregulation of Fas have been reported (19). In the present study, the median value of the FasL IHC score was significantly higher compared to that of the Fas IHC score in ESCC following preoperative CRT (P=0.006). Gratas et al (19) demonstrated that FasL expression was found in over half of the examined cancer cells, whereas Fas expression was rarely observed in tumor cells in ESCC. We observed that Fas expression was present in all the cases (median percentage of Fas-positive cancer cells, 80%; range, 1–100%). To explain this difference, we surmised that Fas expression may have been upregulated by radiation based on a previous study reporting that Fas expression in ESCC was increased by irradiation in vitro in a dose-dependent manner (20). In the present study, we did not compare the expression of Fas pre- and post-CRT.

We observed that a high FasL expression was correlated with tumor relapse. Moreover, patients with high FasL expression exhibited poorer recurrence-free and overall survival among ESCC patients treated with preoperative CRT. Shibakita et al (6) reported that FasL expression did not affect survival, whereas Fas expression was an independent favorable prognostic factor for patient survival in ESCC. In our data, patients with low Fas expression tended to have poor recurrence-free survival compared to those with high Fas expression.

One possible mechanism explaining the involvement of Fas/FasL expression in the prognosis of ESCC is evasion of the host immune response. Tumor-derived FasL counteracts the host immune system by eliminating Fas-sensitive cytotoxic T cells, such as CD8-positive cells (6,19). In the present study, FasL expression was diffusely observed in stromal cells in ESCC following CRT. Although significant differences were not observed, Fas appeared to be negatively correlated with peritumoral Foxp3-positive cells (regulatory T cells) and FasL appeared to be positively correlated with peritumoral CD8-positive cells. Rigberg et al (20) hypothesized that Fas/FasL proteins may provide certain tumors with an immune privilege and increase their resistance to radiotherapy. Moreover, several authors have reported that Fas/FasL signaling plays a role in chemoresistance to doxorubicin and oxaliplatin and immune responses by interaction with matrix metalloproteinase-7 (1012). Although there were no correlations between Fas/FasL expression and TRG in this study, pharmacological control of Fas/FasL signaling may result in improved therapeutic efficacy and outcome in ESCC patients who receive preoperative CRT.

Rigberg et al (20) demonstrated that neither anti-Fas monoclonal antibody nor transduction of FasL directly inhibited tumor cell growth in an in vitro study. Thus, the main role of Fas/FasL signaling in tumor progression is likely the regulation of the tumor microenvironment. Sun et al (21) reported that polymorphisms of Fas/FasL genes appeared to be associated with an increased risk of developing ESCC. Taken together, the Fas/FasL pathway may be considered to be a candidate therapeutic target in ESCC, as the Fas/FasL signaling pathway plays an important role in tumorigenesis and tumor progression (22,23).

In conclusion, high FasL expression was associated with poor prognosis and the evaluation of FasL expression may provide clinically useful prognostic information for ESCC patients receiving neoadjuvant CRT. Moreover, the control of Fas/FasL signaling may lead to improved therapeutic efficacy and outcome in ESCC patients following neoadjuvant CRT. However, the data in this study should be interpreted with caution. The major limitations were the limited patient sample and the retrospective nature of the study. A larger study population is required to validate our conclusions.

References

1 

Stahl M, Stuschke M, Lehmann N, et al: Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J Clin Oncol. 23:2310–2317. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Lv J, Cao XF, Zhu B, Ji L, Tao L and Wang DD: Effect of neoadjuvant chemoradiotherapy on prognosis and surgery for esophageal carcinoma. World J Gastroenterol. 15:4962–4968. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Sjoquist KM, Burmeister BH, Smithers BM, et al: Survival after neoadjuvant chemotherapy or chemoradiotherapy for resectable oesophageal carcinoma: an updated meta-analysis. Lancet Oncol. 12:681–692. 2011. View Article : Google Scholar : PubMed/NCBI

4 

van Hagen P, Hulshof MC, van Lanschot JJ, et al: Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 366:2074–2084. 2012.

5 

Igney FH and Krammer PH: Tumor counterattack: fact or fiction? Cancer Immunol Immunother. 54:1127–1136. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Shibakita M, Tachibana M, Dhar DK, et al: Prognostic significance of Fas and Fas ligand expressions in human esophageal cancer. Clin Cancer Res. 5:2464–2469. 1999.PubMed/NCBI

7 

Green DR and Ferguson TA: The role of Fas ligand in immune privilege. Nat Rev Mol Cell Biol. 2:917–924. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Abrahams VM, Kamsteeg M and Mor G: The Fas/Fas ligand system and cancer: immune privilege and apoptosis. Mol Biotechnol. 25:19–30. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Scholz M and Cinatl J: Fas/FasL interaction: a novel immune therapy approach with immobilized biologicals. Med Res Rev. 25:331–342. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Mitsiades N, Yu WH, Poulaki V, Tsokos M and Stamenkovic I: Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity. Cancer Res. 61:577–581. 2001.PubMed/NCBI

11 

Wang WS, Chen PM, Wang HS, Liang WY and Su Y: Matrix metalloproteinase-7 increases resistance to Fas-mediated apoptosis and is a poor prognostic factor of patients with colorectal carcinoma. Carcinogenesis. 27:1113–1120. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Almendro V, Ametller E, Garcia-Recio S, et al: The role of MMP7 and its cross-talk with the FAS/FASL system during the acquisition of chemoresistance to oxaliplatin. PLoS One. 4:e47282009. View Article : Google Scholar : PubMed/NCBI

13 

Younes M, Schwartz MR, Ertan A, Finnie D and Younes A: Fas ligand expression in esophageal carcinomas and their lymph node metastases. Cancer. 88:524–528. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Kase S, Osaki M, Adachi H, Kaibara N and Ito H: Expression of Fas and Fas ligand in esophageal tissue mucosa and carcinomas. Int J Oncol. 20:291–297. 2002.PubMed/NCBI

15 

Takikita M, Hu N, Shou JZ, et al: Biomarkers of apoptosis and survival in esophageal squamous cell carcinoma. BMC Cancer. 9(310)2009. View Article : Google Scholar

16 

Mandard AM, Dalibard F, Mandard JC, et al: Pathologic assessment of tumor regression after preoperative chemoradiotherapy of esophageal carcinoma. Clinicopathologic correlations. Cancer. 73:2680–2686. 1994. View Article : Google Scholar

17 

Wong SC, Lo SF, Lee KC, Yam JW, Chan JK and Wendy Hsiao WL: Expression of frizzled-related protein and Wnt-signalling molecules in invasive human breast tumours. J Pathol. 196:145–153. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Saigusa S, Tanaka K, Ohi M, et al: Clinical significance of peritumoral mast cells in esophageal squamous cell carcinoma with neoadjuvant chemoradiotherapy. Esophagus. 10:12–19. 2013. View Article : Google Scholar

19 

Gratas C, Tohma Y, Barnas C, Taniere P, Hainaut P and Ohgaki H: Up-regulation of Fas (APO-1/CD95) ligand and down-regulation of Fas expression in human esophageal cancer. Cancer Res. 58:2057–2062. 1998.PubMed/NCBI

20 

Rigberg DA, Centeno J, Kim FS, et al: Irradiation-induced up-regulation of Fas in esophageal squamous cell carcinoma is not accompanied by Fas ligand-mediated apoptosis. J Surg Oncol. 71:91–96. 1999. View Article : Google Scholar

21 

Sun T, Miao X, Zhang X, Tan W, Xiong P and Lin D: Polymorphisms of death pathway genes FAS and FASL in esophageal squamous-cell carcinoma. J Natl Cancer Inst. 96:1030–1036. 2004. View Article : Google Scholar : PubMed/NCBI

22 

O'Brien DI, Nally K, Kelly RG, O'Connor TM, Shanahan F and O'Connell J: Targeting the Fas/Fas ligand pathway in cancer. Expert Opin Ther Targets. 9:1031–1044. 2005.

23 

Villa-Morales M and Fernandez-Piqueras J: Targeting the Fas/FasL signaling pathway in cancer therapy. Expert Opin Ther Targets. 16:85–101. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-February 2015
Volume 3 Issue 1

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Saigusa S, Tanaka K, Ohi M, Toiyama Y, Yasuda H, Kitajima T, Okugawa Y, Inoue Y, Mohri Y, Kusunoki M, Kusunoki M, et al: Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy. Mol Clin Oncol 3: 151-156, 2015
APA
Saigusa, S., Tanaka, K., Ohi, M., Toiyama, Y., Yasuda, H., Kitajima, T. ... Kusunoki, M. (2015). Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy. Molecular and Clinical Oncology, 3, 151-156. https://doi.org/10.3892/mco.2014.431
MLA
Saigusa, S., Tanaka, K., Ohi, M., Toiyama, Y., Yasuda, H., Kitajima, T., Okugawa, Y., Inoue, Y., Mohri, Y., Kusunoki, M."Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy". Molecular and Clinical Oncology 3.1 (2015): 151-156.
Chicago
Saigusa, S., Tanaka, K., Ohi, M., Toiyama, Y., Yasuda, H., Kitajima, T., Okugawa, Y., Inoue, Y., Mohri, Y., Kusunoki, M."Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy". Molecular and Clinical Oncology 3, no. 1 (2015): 151-156. https://doi.org/10.3892/mco.2014.431