Next Article in Journal
Osteoarthritis Severely Decreases the Elasticity and Hardness of Knee Joint Cartilage: A Nanoindentation Study
Previous Article in Journal
Associations of Anti-Aquaporin 5 Autoantibodies with Serologic and Histopathological Features of Sjögren’s Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy

1
Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
2
Affiliation 2; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung 83102, Taiwan
3
Department of Thoracic and Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
4
Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
5
Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
6
Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2019, 8(11), 1864; https://doi.org/10.3390/jcm8111864
Submission received: 10 October 2019 / Revised: 26 October 2019 / Accepted: 31 October 2019 / Published: 3 November 2019
(This article belongs to the Section Oncology)

Abstract

:
Background: Programmed cell death-ligand 1 (PD-L1) is present in a subgroup of cancer patients who may be favorable targets for immune checkpoint inhibitor therapies. However, the significance of the PD-L1 expression in esophageal squamous cell carcinoma (ESCC) patients receiving neoadjuvant chemoradiotherapy remains unclear. Methods: By means of PD-L1 immunohistochemistry 22C3 pharmDx assay, we evaluate the PD-L1 expression and its association with clinical outcome in 107 ESCC patients receiving neoadjuvant chemoradiotherapy. Results: Patients with positive PD-L1 expression have significantly lower pathological complete response rates (13% versus 32%; p = 0.036) than those with negative PD-L1 expression. Univariate survival analysis found that positive PD-L1 expression were correlated with poor overall survival (p = 0.004) and inferior disease-free survival (p < 0.001). In a multivariate analysis, positive PD-L1 expression was independently associated with the absence of a pathologically complete response (p = 0.044, hazard ratio: 3.542), worse overall survival (p = 0.006, hazard ratio: 2.017), and inferior disease-free survival (p < 0.001, hazard ratio: 2.516). Conclusions: For patients with ESCC receiving neoadjuvant chemoradiotherapy, positive PD-L1 expression independently predicts the poor chemoradiotherapy response and worse treatment outcome. Thus, our data suggests that PD-L1 may be an influential biomarker for prognostic classification and for immune checkpoint inhibitor therapies in ESCC patients receiving neoadjuvant chemoradiotherapy.

1. Introduction

In 2018, esophageal cancer was the sixth most common cause of cancer-related death worldwide [1]. In Asia, squamous cell carcinoma is the most prevalent histology of esophageal cancer. At diagnosis, patients with esophageal squamous cell carcinoma (ESCC) usually present with the advanced disease. The standard treatment modality for ESCC is esophagectomy. However, previous research has shown that the five-year overall survival rate of patients with advanced ESCC after receiving esophagectomy alone is only 20–30% [2,3,4,5]. Neoadjuvant chemoradiotherapy has been proposed for patients with advanced ESCC to reduce the primary tumor size and dispose of the micrometastases. Recent randomized trials and meta-analysis have revealed that neoadjuvant chemoradiotherapy followed by esophagectomy has a significant survival benefit compared to esophagectomy alone [6,7,8,9,10]. In particular, patients who achieved a pathological complete response following neoadjuvant chemoradiotherapy had improved survival odds than those who did not [11,12]. However, following neoadjuvant chemoradiotherapy, esophagectomy specimens show that only 20–40% patients can achieve pathological complete response [7,13,14], indicating that a large portion of patients do not respond to chemoradiotherapy. Furthermore, patients receiving neoadjuvant chemoradiotherapy followed by esophagectomy had higher morbidity and mortality rates postoperatively than those receiving esophagectomy alone [15]. Therefore, a biomarker is helpful to forecast the chemoradiotherapy response and its presence may shed light on novel target development.
The immune checkpoint proteins (programmed as cell death 1 (PD-1) receptor) and its ligand (programmed cell death ligand 1 (PD-L1)) are involved in the immune escape of cancer cells [16]. PD-L1 expressed on cancer cells binds to PD-1 receptor on T cells, which leads to T cell inactivation and exhaustion. This hampers cytokine production and causes T cell apoptosis. Cumulatively, these effects contribute to the growth of cancer cells [17,18]. Several studies have reported that PD-L1 overexpression predicts anticancer therapy resistances and poor treatment outcomes [19,20,21]. In addition, the use of PD-1/PD-L1 axis inhibitors in recent clinical trials had the meaningful activity and overall survival benefit in several types of cancers, including esophageal cancer [22,23,24,25]. Studies have also demonstrated that PD-L1 protein expression on the surface of cancer cells is associated with enhanced responses to PD-1/PD-L1 axis inhibitors [23,26]. However, the significance of PD-L1 expression in patients with ESCC after receiving neoadjuvant chemoradiotherapy followed by esophagectomy has not been investigated. Although biopsy specimens before treatment are often very small and show significant difference, they are the only tumor tissue samples for predicting clinical outcome in ESCC patients that have received neoadjuvant chemoradiotherapy. Therefore, we performed PD-L1 immunohistochemistry on pre-treatment biopsy specimens obtained from patients with advanced ESCC receiving neoadjuvant chemoradiotherapy followed by esophagectomy and then correlated the immunohistochemical results with treatment outcomes.

2. Materials and Methods

2.1. Patient and Tumor Materials

Between 1999 and 2013, ESCC patients that underwent neoadjuvant chemoradiotherapy followed by esophagectomy at Chang Gung Memorial Hospital and the Kaohsiung medical center were retrospectively analyzed. Patients without biopsy specimens before neoadjuvant chemoradiothrapy for immunohistochemistry were not allowed. The institutional review board of the Chang Gung Memorial Hospital approved the present study. The seventh American Joint Committee on Cancer (AJCC) staging system was used for clinical staging. The clinical staging was determined according to image examinations including a computed tomography (CT) scan of the abdomen and chest, endoscopic ultrasound (EUS), and/or positron emission tomography (PET) scan. The protocol of neoadjuvant chemoradiotherapy followed by esophagectomy was previously described [14,27]. Overall survival (OS) was computed from diagnosis date until the death date or the last follow-up. Disease-free survival (DFS) was calculated from the esophagectomy date until death due to any cause without recurrence evidence or date of recurrence. Ultimately, 107 ESCC patients who received neoadjuvant chemoradiotherapy followed by esophagectomy were enrolled for further analysis.

2.2. Immunohistochemistry (IHC)

The specimens were fixed in buffered formalin and embedded in paraffin. The IHC was carried out using standard reagents and techniques on a Dako Autostainer Link 48 platform (Agilent Technologies, Santa Clara, CA, USA) and EnVision FLEX visualization system (Agilent Technologies, citySanta Clara, CA, countryUSA). An automated IHC staining protocol of the PD-L1 IHC 22C3 pharmDx assay (Dako, Carpinteria, CA, USA) was verified with positive and negative controls used per manufacturer instructions. Briefly, in the PT Link (Dako PT100), deparaffinization, rehydration, and target antigen retrieval were performed through a three in one process. Then, specimens were incubated with the monocloncal mouse control IgG antibody (negative control) or anti-human PD-L1 monoclonal mouse antibody (clone 22C3), then with an anti-mouse linker antibody specific to the host species of the primary antibody, and then with a ready-to-use visualization reagent consisting of goat secondary antibody molecules and horseradish peroxidase molecules coupled with dextran. The enzymatic conversion of the subsequently added 3,3′-diaminobenzidine tetrahydrochloride (DAB) chromogen with color modification using a DAB enhancer resulted in precipitation of a visible reaction product at the site of antigen. The sections were then counterstained with hematoxylin and interpreted by a pathologist using a light microscope.
To determine the expression of PD-L1 protein, total viable tumor cells were evaluated and only tumors containing at least 100 viable tumor cells were scored. The positive staining was assessed in the context of non-specific background with 0 specific staining and <1+ intensity in the negative control reagent slide. The positivity of PD-L1 was represented as the complete circumferential or partial cell membrane staining of viable cancer cells. Tumor proportion score (TPS) was defined as the percentage of positive tumor cells over total tumor cells in the denominator. Tumor-associated immune cells or tumor cells with cytoplasmic staining were excluded from the scoring. Positive PD-L1 expression was defined as TPS > 1%.

2.3. Statistical Analysis

Statistical analysis was performed using the SPSS 17 software package (manufactureIBM Corp., cityArmonk, NY, countryUSA). We used the chi-square test to compare the categorical data between the two groups. Logistic regression was used for the multivariate analysis of the pathological complete response. For univariate survival analysis in these patients, the Kaplan–Meier method was performed to plot the figures of OS and DFS. The difference between the two groups was evaluated by the log rank test. For multivariate survival analysis, the cox proportional hazards regression model was used. For every analysis, two-sided tests of significance were performed and the p value < 0.05 was considered as significant.

3. Results

3.1. Patient Clinicopathological Characteristics

Table 1 shows the clinicopathological characteristics of these 107 patients. The median age of was 52 years (range: 37–77 years). The stage was revealed to be AJCC seventh stage II in 21 (20%) patients and AJCC seventh stage III in 86 (80%) patients. Meanwhile, the T classifications were T2 in 11 (10%) patients, T3 in 46 (43%) patients, and T4 in 50 (47%) patients. Additionally, 22 (21%) patients had N0 status, 36 (34%) patients had N1 status, 35 (33%) patients had N2 status, and 14 (13%) patients had N3 status. The locations of the primary tumor were as follows: upper esophagus in 20 (19%) patients, the middle esophagus in 43 (40%) patients, and the lower esophagus in 44 (41%) patients. In terms of a histologic grade, 22 (21%) patients were diagnosed with a grade 1 lesion, 58 (54%) patients were diagnosed with a grade 2 lesion, and 27 (25%) patients were diagnosed with a grade 3 lesion. Among these 107 patients receiving neoadjuvant chemoradiotherapy followed by esophagectomy, 28 (26%) patients achieved pathological complete response. The three-year OS and DFS rates of these 107 patients were 37% and 32%, respectively.
As shown in Table 1, 75 (70%) and 32 (30%) patients were negative and positive for PD-L1 expression, respectively (Figure 1). There were no significant correlations between PD-L1 expression with histologic grade, primary tumor location, age, clinical T classification, clinical N classification, and clinical AJCC seventh staging (Table 2)

3.2. Associations between Pathological Complete Respoznse with Clinicopathological Characteristics

Table 3 revealed the correlation between pathological complete responses with clinicopathological characteristics. We observed that positive PD-L1 expression (p = 0.036) and clinical T classification, T4 (p = 0.025) were significantly associated with the absence of pathological complete response. The multivariate analysis demonstrated that positive PD-L1 expression (p = 0.044, hazard ratio: 3.542, 95% confidence interval: 1.033–12.152) and clinical T classification, T4 (p = 0.047, hazard ratio: 3.225, 95% confidence interval: 1.016–10.231), were independently associated with the absence of a pathologically complete response.

3.3. Associations between Patient Survival with Clinicopathological Characteristics

Table 4 shows the association between OS and DFS with clinicopathological characteristics and PD-L1 expression. We found that positive PD-L1 expression (p = 0.004; Figure 2A), clinical T classification, T4 (p = 0.015), and clinical N classification, N2/3 (p = 0.025) were correlated with inferior OS significantly at univariate level. Besides, univariate analysis also revealed that positive PD-L1 expression (p < 0.001; Figure 2B), clinical T classification, T4 (p = 0.006), and clinical N classification, N2/3 (p = 0.044) were significantly correlated with poor DFS. In a multivariate analysis, positive PD-L1 expression (p = 0.006, hazard ratio: 2.017, 95% confidence interval: 1.223–3.326) and clinical T classification, T4 (p = 0.022, hazard ratio: 1.910, 95% confidence interval: 1.097–3.325), were independently poor prognosticators for inferior OS. Meanwhile, positive PD-L1 expression (p < 0.001, hazard ratio: 2.516, 95% confidence interval: 1.537–4.120) and clinical T classification, T4 (p = 0.03, hazard ratio: 1.857, 95% confidence interval: 1.060–3.252), were independently associated with worse DFS. The three-year OS rates were 47% in patients with negative PD-L1 expression, and 16% in patients with positive PD-L1 expression. The three-year DFS rates were 43% in patients with negative PD-L1 expression, and 6% in patients with positive PD-L1 expression.

4. Discussion

Previous studies [26,28] have described that positive PD-L1 expression is correlated with worse prognosis in numerous human cancers and also suggested that such expression could serve as a biomarker which predicts the response to anti-PD-1/PD-L1 therapies. However, the significance of PD-L1 expression in patients with ESCC receiving neoadjuvant chemoradiotherapy followed by esophagectomy remain largely undefined. Therefore, we conducted the current study in order to determine the significance of PD-L1 expression in ESCC patients who underwent neoadjuvant chemoradiotherapy followed by esophagectomy.
In the present study, positive PD-L1 expression was noted in 32 (30%) of 107 patients with ESCC. Hatogai et al. [29] reported that positive PD-L1 expression was noted in 67 (23.4%) of 286 patients with ESCC receiving curative surgical resection. Meanwhile, a recent meta-analysis [30] showed that positive PD-L1 expression was observed in 559 (41.4%) of 1350 patients with ESCC. The discrepancy between these studies may have resulted from differences in the PD-L1 antibodies and IHC assessment methods used.
In our study, we did not observe a significant correlation between histologic grades with pathologically complete responses, despite a previous study by Tamaoki et al. [31] that reported a single-minded 2 (SIM2) increased chemoradiotherapy sensitivity through tumor differentiation in ESCC. However, we found that positive PD-L1 expression was significantly associated with the absence of pathological complete response. We observed that pathologically complete responses after neoadjuvant chemoradiotherapy was noted in 24 (32%) of the 75 patients with negative PD-L1 expression. However, 4 (13%) of the 32 patients with positive PD-L1 expressions achieved a pathologically complete response. For patients with locally advanced ESCC, multimodality treatment including definitive chemoradiotherapy or neoadjuvant chemoradiotherapy followed by esophagectomy has been commonly used. The ability to distinguish responders from non-responders could provide more suitable multimodality treatment options.
Our study revealed that a positive PD-L1 expression is an independent prognosticator in patients with advanced ESCC treated with preoperative chemoradiotherapy. Kudo et al. [25] showed that the use of the anti-PD-1 antibody, nivolumab, in 65 chemotherapy-refractory ESCC patients showed promising activity, with a 17% objective response rate and a manageable safety profile. Tanaka et al. [32] first reported that many interferon-gamma-inducible genes including PD-L1 and cytotoxic T-lymphocyte markers such as perforin (PRF1) and granzyme B (GZMB) were activated by chemoradiotherapy in ESCC. Lim et al. [33] acquired 19 paired ESCC tumor tissues before and after preoperative chemoradiotherapy and found that PD-L1 expression in ESCC cells increased after preoperative chemoradiotherapy. The recent phase III PACIFIC study [34] reported that after chemoradiotherapy, the administration of the anti-PD-L1 antibody, durvalumab, can ameliorate progression-free survival in non-small-cell lung cancer patients with unresectable stage III disease. Taken together, these previous studies and our findings highlight the potential for a combination of anti-PD-1/PD-L1 therapy and chemoradiotherapy in patients with advanced ESCC.
The present study has limitations. The patient number in our study is relatively small. Our analysis was retrospective. Furthermore, we did not evaluate PD-L1 expression in immune cells because some of the biopsy specimens were too small to have enough cells evaluated. Besides, biopsy specimens are too small to show significant difference. For clinical use, the cut-off value should be determined by extensive IHC using multiple sections in the multi-institutional cohorts in the future.

5. Conclusions

In conclusion, our study showed that positive PD-L1 expression independently predicts poor response to chemoradiotherapy and worse survival of patients with ESCC receiving neoadjuvant chemoradiotherapy followed by esophagectomy. Therefore, PD-L1 might be a potential target in advanced ESCC patients receiving chemoradiotherapy.

Author Contributions

Conceptualization: S.-H.L.; Methodology: W.-T.H., Y.-C.L., L.-H.T., and S.-H.L.; Software: Y.-M.W., Y.-H.C., C.-M.L., and W.-C.L.; Validation: W.-T.H., H.-I.L., and S.-H.L.; Formal Analysis: H.-I.L., W.-C.L., and S.-H.L.; Investigation: S.-H.L.; Resources: W.-T.H., H.-I.L., Y.-M.W., Y.-H.C., C.-M.L., and S.-H.L.; Data Curation: W.-T.H., H.-I.L., Y.-M.W., Y.-H.C., C.-M.L., W.-C.L., Y.-C.L., L.-H.T., and S.-H.L.; Writing—Original Draft Preparation: W.-T.H. and S.-H.L.; Writing—Review and Editing: S.-H.L.; Supervision: S.-H.L.; Funding Acquisition: S.-H.L.; All authors read and approved the final manuscript.

Funding

This work was supported in part by grants from the National Science Council, Taiwan (MOST 106-2314-B-182A-159-MY3 and MOST 107-2314-B-182A-156-MY3), and Chang Gung Memorial Hospital (CMRPG8J0401, CMRPG8G0892, CMRPG8I0201, and CMRPG8J1061).

Acknowledgments

We would also like to thank Chang Gung Medical Foundation Kaohsiung Chang Gung Memorial Hospital Biobank and Tissue Bank Core Laboratory (CLRPG8F1701 and CLRPG8F1602) for excellent technical support.

Conflicts of Interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this article.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  2. Li, S.H.; Chen, C.H.; Lu, H.I.; Huang, W.T.; Tien, W.Y.; Lan, Y.C.; Lee, C.C.; Chen, Y.H.; Huang, H.Y.; Chang, A.Y.; et al. Phosphorylated p70S6K expression is an independent prognosticator for patients with esophageal squamous cell carcinoma. Surgery 2015, 157, 570–580. [Google Scholar] [CrossRef] [PubMed]
  3. Li, S.H.; Lu, H.I.; Chang, A.Y.; Huang, W.T.; Lin, W.C.; Lee, C.C.; Tien, W.Y.; Lan, Y.C.; Tsai, H.T.; Chen, C.H. Angiotensin II type I receptor (AT1R) is an independent prognosticator of esophageal squamous cell carcinoma and promotes cells proliferation via mTOR activation. Oncotarget 2016, 7, 67150–67165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sherman, C.A.; Turrisi, A.T.; Wallace, M.B.; Reed, C.E. Locally advanced esophageal cancer. Curr. Treat. Options Oncol. 2002, 3, 475–485. [Google Scholar] [CrossRef] [PubMed]
  5. Van der Werf, L.R.; Dikken, J.L.; van Berge Henegouwen, M.I.; Lemmens, V.; Nieuwenhuijzen, G.A.P.; Wijnhoven, B.P.L.; Dutch Upper GI Cancer Audit Group. A Population-based Study on Lymph Node Retrieval in Patients with Esophageal Cancer: Results from the Dutch Upper Gastrointestinal Cancer Audit. Ann. Surg. Oncol. 2018, 25, 1211–1220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lu, H.I.; Li, S.H.; Huang, W.T.; Rau, K.M.; Fang, F.M.; Wang, Y.M.; Lin, W.C.; Tien, W.Y. A comparative study of isolated and metachronous oesophageal squamous cell carcinoma with antecedent upper aerodigestive tract cancer. Eur. J. Cardiothorac. Surg. 2013, 44, 860–865. [Google Scholar] [CrossRef] [Green Version]
  7. Bedenne, L.; Michel, P.; Bouche, O.; Milan, C.; Mariette, C.; Conroy, T.; Pezet, D.; Roullet, B.; Seitz, J.F.; Herr, J.P.; et al. Chemoradiation followed by surgery compared with chemoradiation alone in squamous cancer of the esophagus: FFCD 9102. J. Clin. Oncol. 2007, 25, 1160–1168. [Google Scholar] [CrossRef]
  8. Toxopeus, E.; van der Schaaf, M.; van Lanschot, J.; Lagergren, J.; Lagergren, P.; van der Gaast, A.; Wijnhoven, B. Outcome of Patients Treated Within and Outside a Randomized Clinical Trial on Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer: Extrapolation of a Randomized Clinical Trial (CROSS). Ann. Surg. Oncol. 2018, 25, 2441–2448. [Google Scholar] [CrossRef]
  9. Nygaard, K.; Hagen, S.; Hansen, H.S.; Hatlevoll, R.; Hultborn, R.; Jakobsen, A.; Mantyla, M.; Modig, H.; Munck-Wikland, E.; Rosengren, B.; et al. Pre-operative radiotherapy prolongs survival in operable esophageal carcinoma: A randomized, multicenter study of pre-operative radiotherapy and chemotherapy. The second Scandinavian trial in esophageal cancer. World J. Surg. 1992, 16, 1104–1109. [Google Scholar] [CrossRef]
  10. Gebski, V.; Burmeister, B.; Smithers, B.M.; Foo, K.; Zalcberg, J.; Simes, J. Survival benefits from neoadjuvant chemoradiotherapy or chemotherapy in oesophageal carcinoma: A meta-analysis. Lancet Oncol. 2007, 8, 226–234. [Google Scholar] [CrossRef]
  11. Donington, J.S.; Miller, D.L.; Allen, M.S.; Deschamps, C.; Nichols, F.C., III; Pairolero, P.C. Tumor response to induction chemoradiation: Influence on survival after esophagectomy. Eur. J. Cardiothorac. Surg. 2003, 24, 631–636. [Google Scholar] [CrossRef]
  12. Jones, D.R.; Detterbeck, F.C.; Egan, T.M.; Parker, L.A., Jr.; Bernard, S.A.; Tepper, J.E. Induction chemoradiotherapy followed by esophagectomy in patients with carcinoma of the esophagus. Ann. Thorac. Surg. 1997, 64, 185–191. [Google Scholar] [CrossRef]
  13. Stahl, M.; Stuschke, M.; Lehmann, N.; Meyer, H.J.; Walz, M.K.; Seeber, S.; Klump, B.; Budach, W.; Teichmann, R.; Schmitt, M.; et al. Chemoradiation with and without surgery in patients with locally advanced squamous cell carcinoma of the esophagus. J. Clin. Oncol. 2005, 23, 2310–2317. [Google Scholar] [CrossRef] [PubMed]
  14. Li, S.H.; Huang, E.Y.; Lu, H.I.; Huang, W.T.; Yen, C.C.; Huang, W.C.; Chen, C.H. Phosphorylated mammalian target of rapamycin expression is associated with the response to chemoradiotherapy in patients with esophageal squamous cell carcinoma. J. Thorac. Cardiovasc. Surg. 2012, 144, 1352–1359. [Google Scholar] [CrossRef] [PubMed]
  15. Steyerberg, E.W.; Neville, B.A.; Koppert, L.B.; Lemmens, V.E.; Tilanus, H.W.; Coebergh, J.W.; Weeks, J.C.; Earle, C.C. Surgical mortality in patients with esophageal cancer: Development and validation of a simple risk score. J. Clin. Oncol. 2006, 24, 4277–4284. [Google Scholar] [CrossRef]
  16. Butte, M.J.; Keir, M.E.; Phamduy, T.B.; Sharpe, A.H.; Freeman, G.J. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 2007, 27, 111–122. [Google Scholar] [CrossRef]
  17. Dong, H.; Zhu, G.; Tamada, K.; Chen, L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 1999, 5, 1365–1369. [Google Scholar] [CrossRef]
  18. Ceeraz, S.; Nowak, E.C.; Noelle, R.J. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol. 2013, 34, 556–563. [Google Scholar] [CrossRef] [Green Version]
  19. Ritprajak, P.; Azuma, M. Intrinsic and extrinsic control of expression of the immunoregulatory molecule PD-L1 in epithelial cells and squamous cell carcinoma. Oral Oncol. 2015, 51, 221–228. [Google Scholar] [CrossRef] [Green Version]
  20. Pilones, K.A.; Vanpouille-Box, C.; Demaria, S. Combination of radiotherapy and immune checkpoint inhibitors. Semin. Radiat. Oncol. 2015, 25, 28–33. [Google Scholar] [CrossRef]
  21. Afreen, S.; Dermime, S. The immunoinhibitory B7-H1 molecule as a potential target in cancer: Killing many birds with one stone. Hematol. Oncol. Stem Cell Ther. 2014, 7, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  23. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
  24. Kojima, T.; Doi, T. Immunotherapy for Esophageal Squamous Cell Carcinoma. Curr. Oncol. Rep. 2017, 19, 33. [Google Scholar] [CrossRef]
  25. Kudo, T.; Hamamoto, Y.; Kato, K.; Ura, T.; Kojima, T.; Tsushima, T.; Hironaka, S.; Hara, H.; Satoh, T.; Iwasa, S.; et al. Nivolumab treatment for oesophageal squamous-cell carcinoma: An open-label, multicentre, phase 2 trial. Lancet Oncol. 2017, 18, 631–639. [Google Scholar] [CrossRef]
  26. Sznol, M.; Chen, L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin. Cancer Res. 2013, 19, 1021–1034. [Google Scholar] [CrossRef]
  27. Chiu, T.J.; Lu, H.I.; Chen, C.H.; Huang, W.T.; Wang, Y.M.; Lin, W.C.; Li, S.H. Osteopontin Expression Is Associated with the Poor Prognosis in Patients with Locally Advanced Esophageal Squamous Cell Carcinoma Receiving Preoperative Chemoradiotherapy. BioMed. Res. Int. 2018, 2018, 9098215. [Google Scholar] [CrossRef]
  28. Wang, Q.; Liu, F.; Liu, L. Prognostic significance of PD-L1 in solid tumor: An updated meta-analysis. Medicine 2017, 96, 6369. [Google Scholar] [CrossRef]
  29. Hatogai, K.; Fujii, S.; Kojima, T.; Daiko, H.; Nomura, S.; Doi, T.; Kitano, S.; Ohtsu, A.; Takiguchi, Y.; Yoshino, T.; et al. Large-scale comprehensive immunohistochemical biomarker analyses in esophageal squamous cell carcinoma. J. Cancer Res. Clin. Oncol. 2017, 143, 2351–2361. [Google Scholar] [CrossRef]
  30. Qu, H.X.; Zhao, L.P.; Zhan, S.H.; Geng, C.X.; Xu, L.; Xin, Y.N.; Jiang, X.J. Clinicopathological and prognostic significance of programmed cell death ligand 1 (PD-L1) expression in patients with esophageal squamous cell carcinoma: A meta-analysis. J. Thorac. Dis. 2016, 8, 3197–3204. [Google Scholar] [CrossRef]
  31. Tamaoki, M.; Komatsuzaki, R.; Komatsu, M.; Minashi, K.; Aoyagi, K.; Nishimura, T.; Chiwaki, F.; Hiroki, T.; Daiko, H.; Morishita, K.; et al. Multiple roles of single-minded 2 in esophageal squamous cell carcinoma and its clinical implications. Cancer Sci. 2018, 109, 1121–1134. [Google Scholar] [CrossRef] [PubMed]
  32. Tanaka, Y.; Aoyagi, K.; Minashi, K.; Komatsuzaki, R.; Komatsu, M.; Chiwaki, F.; Tamaoki, M.; Nishimura, T.; Takahashi, N.; Oda, I.; et al. Discovery of a Good Responder Subtype of Esophageal Squamous Cell Carcinoma with Cytotoxic T-Lymphocyte Signatures Activated by Chemoradiotherapy. PLoS ONE 2015, 10, e0143804. [Google Scholar] [CrossRef] [PubMed]
  33. Lim, S.H.; Hong, M.; Ahn, S.; Choi, Y.L.; Kim, K.M.; Oh, D.; Ahn, Y.C.; Jung, S.H.; Ahn, M.J.; Park, K.; et al. Changes in tumour expression of programmed death-ligand 1 after neoadjuvant concurrent chemoradiotherapy in patients with squamous oesophageal cancer. Eur. J. Cancer 2016, 52, 1–9. [Google Scholar] [CrossRef] [PubMed]
  34. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
Figure 1. Representative photographs of PD-L1 immunostaining in esophageal squamous cell carcinoma. The positive staining was assessed against the negative control staining (NC) (original magnification 200x). PD-L1, programmed death-ligand 1.
Figure 1. Representative photographs of PD-L1 immunostaining in esophageal squamous cell carcinoma. The positive staining was assessed against the negative control staining (NC) (original magnification 200x). PD-L1, programmed death-ligand 1.
Jcm 08 01864 g001
Figure 2. (A) Overall survival according to PD-L1 expression. (B) Disease-free survival according to PD-L1 expression. PD-L1, programmed death-ligand 1.
Figure 2. (A) Overall survival according to PD-L1 expression. (B) Disease-free survival according to PD-L1 expression. PD-L1, programmed death-ligand 1.
Jcm 08 01864 g002
Table 1. Clinicopathologic features of 107 patients with locally advanced esophageal squamous cell carcinoma who received preoperative chemoradiotherapy.
Table 1. Clinicopathologic features of 107 patients with locally advanced esophageal squamous cell carcinoma who received preoperative chemoradiotherapy.
ParametersNo. of Cases (Percentage)
Age (years) (mean: 53.6, median: 52, range 37–77)
<5040 (37%)
50 ≤ Age < 6036 (34%)
60 ≤ Age< 7027 (25%)
70 ≤ Age4 (4%)
Clinical seventh American Joint Committee on Cancer (AJCC) stage
II21 (20%)
III86 (80%)
Clinical T classification
T211 (10%)
T346 (43%)
T450 (47%)
Clinical N classification
N022 (21%)
N136 (34%)
N235 (33%)
N314 (13%)
Histologic grade (Tumor differentiation)
Grade 1 (Well differentiated)22 (21%)
Grade 2 (Moderately differentiated)58 (54%)
Grade 3 (poorly differentiated, undifferentiated)27 (25%)
Primary tumor location
Upper20 (19%)
Middle43 (40%)
Lower44 (41%)
PD-L1 expression
Negative75 (70%)
Positive32 (30%)
pCR
Absent79 (74%)
Present28 (26%)
PD-L1, programmed death-ligand 1; pCR, pathological complete response.
Table 2. Associations between PD-L1 expression and clinicopathologic parameters.
Table 2. Associations between PD-L1 expression and clinicopathologic parameters.
Parameters PD-L1 Expression
NegativePositivep value
Age≤52 years old40150.54
>52 years old3517
Clinical seventh AJCC stageII1560.88
III6026
Clinical T classificationT2/339180.69
T43614
Clinical N classificationN01840.18
N1/2/35728
Clinical N classificationN0/140180.78
N2/33514
Histologic gradeGrade 1/256240.97
Grade 3198
Histologic gradeGrade 11840.18
Grade 2/35728
Primary tumor locationUpper/Middle44190.95
Lower3113
PD-L1, programmed death-ligand 1. x2 test was used for statistical analysis.
Table 3. Associations between a pathologically complete response and clinicopathologic parameters.
Table 3. Associations between a pathologically complete response and clinicopathologic parameters.
Parameters Pathological Complete Response
PresentAbsentp value
Age≤52 years old11440.14
>52 years old1735
Clinical seventh AJCC stageII8130.17
III2066
Clinical T classificationT2/320370.025*
T4842
Clinical N classificationN09130.078
N1/2/31966
Clinical N classificationN0/117410.42
N2/31138
Histologic gradeGrade 1/219610.87
Grade 3621
Histologic gradeGrade 18140.22
Grade 2/32065
Primary tumor locationUpper/Middle12510.21
Lower1331
PD-L1 expressionNegative24510.036*
Positive428
PD-L1, programmed death-ligand 1. * Statistically significant. x2 test or Fisher’s exact test was used for statistical analysis.
Table 4. Results of univariate log-rank analysis of prognostic factors for overall survival and disease-free survival in 107 patients with locally advanced esophageal squamous cell carcinoma who received preoperative chemoradiotherapy.
Table 4. Results of univariate log-rank analysis of prognostic factors for overall survival and disease-free survival in 107 patients with locally advanced esophageal squamous cell carcinoma who received preoperative chemoradiotherapy.
FactorsNo. of PatientsOverall Survival (OS)Disease-Free Survival (DFS)
3-Year OS Rate (%)p Value3-Year DFS Rate (%)p Value
Age
≤52 years old5535%0.5129%0.86
>52 years old5240% 35%
Clinical seventh AJCC stage
II2152%0.1452%0.084
III8634% 27%
Clinical T classification
T2/35749%0.015*42%0.006*
T45024% 20%
Clinical N classification
N02255%0.1055%0.046*
N1/2/38533% 26%
Clinical N classification
N0/15847%0.025*38%0.044*
N2/34927% 25%
Histologic grade
Grade 1/28039%0.1833%0.82
Grade 32733% 30%
Histologic grade
Grade 12250%0.1750%0.24
Grade 2/38534% 27%
Primary tumor location
Upper/Middle6338%0.8930%0.75
Lower4436% 34%
PD-L1 expression
Negative7547%0.004*43%<0.001*
Positive3216% 6%
PD-L1, programmed death-ligand 1. *Statistically significant.

Share and Cite

MDPI and ACS Style

Huang, W.-T.; Lu, H.-I.; Wang, Y.-M.; Chen, Y.-H.; Lo, C.-M.; Lin, W.-C.; Lan, Y.-C.; Tseng, L.-H.; Li, S.-H. Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy. J. Clin. Med. 2019, 8, 1864. https://doi.org/10.3390/jcm8111864

AMA Style

Huang W-T, Lu H-I, Wang Y-M, Chen Y-H, Lo C-M, Lin W-C, Lan Y-C, Tseng L-H, Li S-H. Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy. Journal of Clinical Medicine. 2019; 8(11):1864. https://doi.org/10.3390/jcm8111864

Chicago/Turabian Style

Huang, Wan-Ting, Hung-I Lu, Yu-Ming Wang, Yen-Hao Chen, Chien-Ming Lo, Wei-Che Lin, Ya-Chun Lan, Ling-Huei Tseng, and Shau-Hsuan Li. 2019. "Positive Programmed Cell Death-Ligand 1 Expression Predicts Poor Treatment Outcomes in Esophageal Squamous Cell Carcinoma Patients Receiving Neoadjuvant Chemoradiotherapy" Journal of Clinical Medicine 8, no. 11: 1864. https://doi.org/10.3390/jcm8111864

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop