Open Access

CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer

  • Authors:
    • Qing Li
    • Dachuan Zhang
    • Wenting He
    • Tongbing Chen
    • Zhantao Yan
    • Xie Gao
    • Lujun Chen
    • Xiao Zheng
    • Bin Xu
    • Binfeng Lu
    • Jingting Jiang
  • View Affiliations

  • Published online on: July 8, 2020     https://doi.org/10.3892/ol.2020.11828
  • Pages: 2655-2664
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The presence of tumor infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLSs) in tumor tissues are of great prognostic significance in several types of human cancer. The present study investigated the density of TILs and TLSs in gastric cancer (GC) tissues and their association with pathological parameters. Moreover, the clinical significance of follicular CD8+ cytotoxic T cells present within the germinal centers of the tumor‑associated TLSs was investigated. Immunohistochemistry and H&E staining were used to examine the infiltration and distribution patterns of TILs, TLSs and germinal center (gc) CD8+ TILs in tumor tissues obtained from 63 patients with GC. The number of TILs, TLSs, combination of TILs and TLSs (TILs‑TLSs) and gcCD8+ TILs were used to define tumoral immune parameters, and the prognostic value of these parameters was assessed. The analysis revealed that patients with GC with increased levels of TILs, TLSs, or gcCD8+ TILs exhibited improved overall survival. In addition, gcCD8+ TILs levels were significantly associated with patient age, histological grade and pTN stage. Increased levels of TILs‑TLSs were positively associated with nerve invasion, tumor thrombus, nodal metastasis and histological grade. Multivariate Cox regression analysis revealed that TILs‑TLSs and gcCD8+ TILs were independent prognostic factors. The data obtained in the present study demonstrated that high levels of tumoral immune parameters are important independent prognostic predictors for human GC. The results also suggested a possible role of gcCD8+ TILs in tumor immune surveillance.

Introduction

Previous studies have, demonstrate that immune cell infiltration in human gastrointestinal cancers is associated with cancer progression and is a favorable prognostic predictor (15). Both cellular composition and organization of tumor infiltrating lymphocytes (TILs) are crucial for inhibiting cancer progression and are implicated in the success of cancer immunotherapy (1,2,6). Tertiary lymphoid structure (TLSs) are an important source of TILs, characterized by ectopic aggregated lymphocytes with high endothelial venules and have a similar function to secondary lymphoid organs (SLOs). The lymphocytes in TLSs have easy access to tumor antigens as TLSs are not encapsulated and embedded within the tumor microenvironment (5). The numbers of both TILs and TLSs within solid tumor tissues can be used for the assessment of tumor immune surveillance and are important prognostic factors for cancer (711).

Although TLSs are thought to be associated with anti-tumor immune responses (1215), the functional role of their cellular components remains unclear. TLSs are divided into B- and T-zones, which consist of follicular dendritic cells (FDCs) and fibroblastic reticular cells (FRCs), respectively (16). TLSs can be further defined as primary and secondary TLSs, based on the absence or presence of a germinal center in B-cell follicles. Following stimulation by tumor antigens, B cells differentiate and form the germinal center, which is the site of B cell proliferation, class switching and somatic hyper mutation (1719). CD4+ CXCR5+ T follicular helper (Tfh) cells are a subtype of CD4+ helper T cells, principally located in germinal centers, and play critical roles in recruiting, activating and regulating the germinal center (2022). Previous studies suggested that Tfh cells mediate follicular and germinal center formation in TLSs (23,24). In recent studies, CXCR5+ CD8+ T cells were found within the lymphoid follicle, primarily within the germinal center, where they controlled viral replication during chronic HIV and lymphocytic choriomeningitis virus (LMCV) infection (25,26). The CD20+ B cells and CD8+ T cells co-localize in the tumor nest (20) and TLSs (21). This distribution pattern of follicular CD8+ T cells might be involved in antitumor immune responses.

The present study investigated follicular CD8+ T cells in the tumor tissues of patients with gastric cancer (GC). In addition, germinal center CD8+ (gcCD8+) TILs were quantified. The relationship between tumoral immune parameters such as TILs, TLSs, TILs-TLSs and gcCD8+ TILs and clinical pathological parameters was determined.

Materials and methods

Patients and tissue samples

The present study retrospectively analyzed data of patients admitted to the Department of Pathology, Third Affiliated Hospital of Soochow University between 2006 to 2008. The patients were enrolled according to the following criteria: i) Pathologically-confirmed diagnosis of primary GC (adenocarcinoma); ii) did not receive pre-operative chemotherapy or radiotherapy; iii) presence of adequate paraffin-embedded fixed tissue blocks; iv) at least one slide contained the invasive margin of the tumor; and v) availability of complete medical records and follow-up information. A total of 63 patients with GC were included in this present study. Tumor clinical and pathological staging system was based on the Eighth Edition of the Union for International Cancer Control/American Joint Committee on Cancer. Three slides contain cancer tissue were available for each patient, and a total of 189 slides were reviewed in the present study. Patient survival data were available until the end of November 2011. Patient clinical data are presented in Table I. The present study was approved by the Ethics Committee of Soochow University, and complied with the Declaration of Helsinki. Informed consent to use the tissue sample for scientific research was obtained from all patients.

Table I.

Details of parameters in the study cohort.

Table I.

Details of parameters in the study cohort.

Total study cohort

Parametersn%
Sex
  Male4977.8
  Female1422.2
Age, years
  ≤50812.7
  >505587.3
Tumor size
  ≤5 cm3961.9
  >5 cm2438.1
Nerve invasion
  Yes2539.7
  No3860.3
Tumor thrombus
  Yes2234.9
  No4165.1
Nodal metastasis
  Yes3657.1
  No2742.9
Histological grade
  I–II3047.6
  III3352.4
pTN stage
  I2336.5
  II1320.6
  III2742.9
TILs
  High4469.8
  Low1930.2
TLSs
  High4368.3
  Low2031.7
TILs and TLSs
  High3250.8
  Low3149.2
gcCD8+ TILsa
  High2341.8
  Low3258.2

a gcCD8+ TILs were obtained in 55 patients. TILs, tumor infiltrating lymphocytes; TLSs, tertiary lymphoid structures.

Pathomorphological evaluation of TILs and TLSs

Cancer tissue was fixed in 10% (v/v) formalin and embedded in paraffin until use. The H&E stained slides of the resected GC tissues were reviewed and scored independently by two pathologists who were blinded to the clinical data and prognosis of the patients. The pathologists were trained in the pathomorphological evaluation of TILs and TLSs, and any problematic cases were discussed with them during subsequent scoring. The TILs and TLSs in the center of the tumor (CT) and the invasive margin (IM) were examined (11,27,28). The TILs scoring system incorporated two aspects: i) The number of infiltrating lymphocytes; score 0, no infiltration; score 1, mild infiltration; score 2, moderate infiltration; score 3, extensive infiltration (Fig. 1); and ii) the percentage of the tumor area containing TILs in the CT or IM. The CT-TILs and IM-TILs location scores were defined as the number of infiltrating lymphocytes multiplied by the percentage, and then, the final score was computed by summation of the CT- and IM-TILs scores. TLSs were also evaluated in the CT and IM by measuring two factors: The number of TLSs (aggregates of lymphocytes with and without germinal centers were counted) and the percentage, similar to the TILs, in the CT or IM (29,30). The CT-TLSs and IM-TLSs location scores were defined as the numbers multiplied by the percentage, and then, the final score of TLSs was computed by summation of the CT- and IM-TLS scores.

Immunohistochemistry and evaluation of the gcCD8+ T cells

Formalin-fixed and paraffin-embedded (FFPE) tissues were cut into 4 µm thick consecutive sections deparaffinized in xylene and rehydrated in graded ethanol solutions. Monoclonal mouse antibody against human CD8 (MAB-0021, ready to use, Maixin Biotechnology Limited Corporation) was used to stain the T lymphocytes. The CD8 antigen was retrieved by boiling the slides in citrate buffer (10 mmol/l; pH 6.0) under high pressure for 2 min. Then, the sections were immersed in 3% hydrogen peroxide for 10 min to block endogenous peroxidase activity, rinsed three times in PBS, and then incubated with primary antibodies at 4°C overnight. The negative control was performed with PBS. The sections were incubated with horseradish peroxidase-labeled goat anti-mouse secondary antibody (ready to use, Maixin Biotechnology Limited Corporation). Diaminobenzene was used as the chromogen and hematoxylin as the nuclear counterstain. The scoring system for gcCD8+ TILs was assessed in every slide. The number of germinal centers and gcCD8+ TILs in each germinal center were counted. The average values were subsequently obtained.

Statistical analysis

Statistical analyses were performed using SPSS (version 24.0; IBM Corp.) and GraphPad Prism software (version 6; GraphPad Software, Inc.). Data were analyzed using the χ2 test, Kaplan-Meier method and Cox regression analysis as appropriate. All tests were two-sided and P<0.05 was considered to indicate a statistically significant difference.

Results

Relationship between tumoral immune parameters and clinical pathological parameters

The TILs, TLSs, and gcCD8+ TILs were defined as tumoral immune parameters in the present study (Tables I and II). In order to make the scoring system more facilitative to statistical analysis, these variables were converted into binary variables. TILs were sub-grouped by the TIL final score: TILhi 44 cases, score >0.45 [area under the receiver operating characteristic curve (AUC)=0.35; sensitivity, 82.1%; specificity, 50.0%], and TILlow 19 cases, score ≤0.45 (Table I; Fig. 1A-C). TLSs were sub-grouped by the TLS final score: TLShi 43 cases, score >0.42 (AUC=0.39; sensitivity, 79.5%; specificity, 50.0%), and TLSlow 20 cases, score ≤0.42 (Table I; Fig. 1D-F). Considering the combination of TILs and TLSs, the samples were divided into two groups. The TIL-TLShi group (32 cases) consisted of samples that scored as both TILhi and TLShi. The TIL-TLSlow group (31 cases) consisted of samples that scored as TILhi-TLSlow, TILlow-TLShi and TILlow-TLSlow (Table I). Based on the density of gcCD8+ TILs, 55 samples (specimens from eight cases were excluded due to absence of germinal centers) were divided into two groups: gcCD8hi TIL, 23 cases, score >6.93 (AUC=0.19; sensitivity, 59.5%; specificity, 94.4%; Fig. 2A-F), and gcCD8low TIL, 32 cases, score ≤6.93 (Table I; Fig. 2G-L).

Table II.

Association between clinicopathological parameters and tumoral immune parameters.

Table II.

Association between clinicopathological parameters and tumoral immune parameters.

gcCD8+ TILs TILs TLSs TILs-TLSs




ParametersLowHighχ2P-valueLowHighχ2P-valueLowHighχ2P-valueLowHighχ2P-value
Sex
  Male23190.8540.35517322.1530.14215340.1310.71826231.3110.252
  Female94 212 59 59
Age (years)
  ≤50604.8410.028441.7130.191350.1400.708530.6480.421
  >502623 1540 1738 2629
Tumor size
  ≤5 cm18171.8040.1797327.2460.00712270.0450.83216232.7410.098
  >5 cm146 1212 816 159
Nerve invasion
  Yes1280.0430.836141113.140 <0.00110151.3030.2541878.6160.003
  No2015 533 1028 1325
Tumor thrombus
  Yes1343.3830.06610123.7550.05310122.9320.0871667.4830.006
  No1919 932 1031 1526
Nodal metastasis
  Yes2092.9320.08716208.1390.00412240.0980.75522144.7630.029
  No1214 324 819 918
Histological grade
  I–II11166.6310.0105254.9500.0268220.6820.40910205.7730.016
  III217 1419 1221 2112
pTN stage
  I9136.0530.0482218.0720.0186170.6420.7257165.3990.067
  II65 58 58 76
  III175 1215 918 1710

[i] Values in bold indicate P<0.05. TILs, tumor infiltrating lymphocytes; TLSs, tertiary lymphoid structures.

The TILs were significantly associated with the tumor size, nerve invasion, lymph nodal metastasis, histological grade and pTN stage (P=0.007, 0.001, 0.004, 0.026 and 0.018, respectively; Table II). The data showed that the TLSs were not significantly associated with clinical pathological parameters. The combination of TILs-TLSs was significantly associated with nerve invasion, tumor thrombus, lymph nodal metastasis and histological grade (P=0.003, 0.006, 0.029 and 0.016, respectively; Table II). The data also showed the gcCD8+ TILs was significantly associated with patient age, histological grade and pTN stage (P=0.028, 0.010 and 0.048, respectively; Table II). The higher levels of gcCD8+ TILs were associated with lower histological grade and lower pTN stage. These data suggested that the gcCD8+ TILs might be involved in antitumor immunity in GC.

Association between tumoral immune parameters and patient prognosis

Kaplan-Meier survival analysis showed that the patients with TILhi, TLShi, TIL-TLShi or gcCD8hi TIL had favorable prognosis than those with lower levels (P=0.0008, 0.0108, 0.0005 and 0.0003, respectively; Fig. 3A-D). The prognostic value of tumoral immune parameters in patients with advanced GC (pTN III stage) was investigated. The patients in the TIL-TLShi and gcCD8hi TIL groups had an improved overall survival (OS) compared with patients in the TIL-TLSlow and gcCD8low TIL groups (P=0.012 and 0.002; Table III; Fig. 3G and H).

Table III.

Kaplan-Meier analysis of tumoral immune parameters.

Table III.

Kaplan-Meier analysis of tumoral immune parameters.

A, Tumoral immune parameters in all cases

ParametersCases, nStatus of deathMean OS, monthsLog-Rank χ2P-value
TILs
  High441249.2511.1980.001
  Low191224.21
TLSs
  High431248.146.5010.011
  Low201233.40
TILs-TLSs
  High32653.0312.148 <0.001
  Low311833.42
gcCD8+ TILs
  High23157.8313.322 <0.001
  Low321736.16

B, Tumoral immune parameters in pTN III stage cases

ParametersCases, nStatus of deathMean OS, monthsLog-Rank χ2P-value

TILs
  High121030.333.5270.06
  Low151018.58
TLSs
  High181119.113.0890.079
  Low9918.89
TILs-TLSs
  High10535.506.3100.012
  Low171519.00
gcCD8+ TILs
  High509.3340.002
  Low171518.65

[i] Values in bold indicate P<0.05. TILs, tumor infiltrating lymphocytes; TLSs, tertiary lymphoid structures.

In the univariate Cox regression analysis (analysisa), smaller tumor size, lower histological grade, absence of tumor thrombus or lymph nodal metastasis, lower pTN stage and higher levels of tumoral immune parameters were favorable prognostic factors for OS (Table IV). Considering the possible interference among TILs, TLSs and TILs-TLSs, we did a cohort of multivariate Cox regression analysis. Multivariate Cox regression analysisb (based on the clinicopathological feature, TILs, TLSs and gcCD8+ TILs) revealed that the gcCD8+ TILs were the only independent prognostic factor for OS (HR=0.087; 95% CI: 0.011–0.692; P=0.021; Table IV). Multivariate Cox regression analysisc (based on the clinicopathological feature, TILs-TLSs and gcCD8+ TILs) demonstrated that TILs-TLSs and gcCD8+ TILs were independent prognostic factors (HR=0.247, 95% CI: 0.069–0.882, P=0.031; HR=0.067, 95% CI: 0.008–0.561, P=0.013, respectively). Moreover, the multivariate Cox regression analysisd was designed to determine the association between tumoral immune parameters (TILs, TLSs and gcCD8+ TILs). The result showed TILs and gcCD8+ TILs could be used as independent prognostic factors (HR=0.322, 95% CI: 0.124–0.835, P=0.020; HR=0.059, 95% CI: 0.008–0.451, P=0.006, respectively). The tumoral immune parameters exhibited an HR <1 in the Cox regression analysis, suggesting that may protect against tumor progression.

Table IV.

Univariate and multivariate Cox regression analyses of clinicopathological parameters and tumoral immune parameters.

Table IV.

Univariate and multivariate Cox regression analyses of clinicopathological parameters and tumoral immune parameters.

A, Univariate analysis

Variables Unfavorable/favorableHR95% CIP-value
SexMale/Female0.8630.343–2.1760.755
Age, years≤50/>500.7470.255–2.1870.594
Tumor size, cm≤5/>53.6431.587–8.3630.002
Histological gradeI–II/III4.6241.722–12.4210.002
Neural InvasionNo/Yes2.1680.969–4.8470.060
Tumor thrombusNo/Yes6.6192.716–16.1340.001
Lymphatic metastasisNo/Yes11.5282.702–49.1840.001
pTN stageI–II/III18.7322.525–138.9760.004
TILsHigh/Low0.2790.125–0.6230.002
TLSsHigh/Low0.3700.166–0.8250.015
gcCD8+ TILsHigh/Low0.0620.008–0.4700.007
TILs-TLSsHigh/Low0.3760.222–0.6360.000

B, Multivariate Cox regression analysis
Variables Unfavorable/favorableHR95% CIP-value

Tumor size, cm≤5/>50.8960.198–4.0460.886
Histological grade   I–II/III1.0090.277–3.6710.990
Tumor thrombusNo/Yes2.2270.482–10.7480.299
Lymphatic metastasisNo/Yes1.2510.135–11.5600.844
pTN stageI–II/III5.5940.336–93.2370.230
TILsHigh/Low0.5030.169–1.4920.215
TLSsHigh/Low0.6090.203–1.8310.377
gcCD8+ TILsHigh/Low0.0870.011–0.6920.021

C, Multivariate Cox regression analysis

Variables Unfavorable/favorableHR95% CIP-value

Tumor size, cm≤5/>51.2080.273–5.3550.803
Histological gradeI–II/III0.8060.208–3.1160.754
Tumor thrombusNo/Yes1.7420.381–7.9700.474
Lymphatic metastasisNo/Yes0.9800.103–9.2960.986
pTN stageI–II/III7.1590.419–122.4100.174
gcCD8+ TILsHigh/Low0.0670.008–0.5610.013
TILs-TLSsHigh/Low0.2470.069–0.8820.031

D, Multivariate Cox regression analysis

Variables Unfavorable/favorableHR95% CIP-value

TILsHigh/Low0.3220.124–0.8350.020
TLSsHigh/Low0.3960.148–1.0560.064
gcCD8+ TILsHigh/Low0.0590.008–0.4510.006

[i] Values in bold indicate P<0.05. TILs, tumor infiltrating lymphocytes; TLSs, tertiary lymphoid structures; HR, hazard ratio.

Discussion

The present study demostrated that patients with GC with higher levels of TILs, or TLSs, or gcCD8+ TILs had an improved OS. Multivariate Cox regression analysis revealed that TILs-TLSs and gcCD8+ TILs were independent prognostic factors. In addition, gcCD8+ TILs were significantly associated with patient age, histological grade and pTN stage. Higher levels of TILs-TLSs was positively associated with the nerve invasion, tumor thrombus, lymph nodal metastasis and histological grade. The data obtained in the present study suggested that high levels of tumoral immune parameters were associated with improved prognosis in patients with GC.

Cancer progression is a multi-step process that involves genetic, epigenetic, as well as histopathological change (3,3133). Infiltrating immune cells play an important role in preventing or promoting cancer progression (34). Previous studies have demonstrated that high levels of TILs may inhibit the progression of breast and colorectal cancer (8,31,35). The present study revealed that a high level of TILs was associated with improved prognosis in patient with GC. Univariate and multivariate Cox regression analyses revealed that TILs could be used as an independent favorable prognostic factor.

Previous studies demonstrated that TLSs are required for the development of the T- and B-cell mediated immune response against human cancer and various other diseases (25,26,31,36,37). TLSs have been previously described in several types of cancer, such as lung cancer and breast cancer, and are associated with patient prognosis (10,17,29). TLSs are organization by heterotopic lymphoid tissues, and exhibit similar organization and functionality to SLOs. The basic components of TLSs include the T-zone, comprising of T cells and FRCs, and the B-zone, comprising of B cells and FDCs. TLSs are involved in establishing a systemic memory response to protect patients against circulating metastatic cancer cells, therefor inhibiting tumor recurrence for several years (8,31). The present study revealed that the high levels of TLSs were associated with improved outcome in patients with GC, and could therefor be used as an independent prognostic factor. The combination of TILs and TLSs is superior to TILs or TLSs alone for predicting survival. In advanced cancer, the high levels of TILs-TLSs were associated with improved OS. Based on the results obtained in the present study, TLSs are an important niche in which tumor antigen specific T and B cells are generated.

Despite the association of gcCD8+ T cells with improved prognosis, the role of gcCD8+ T cells in antitumor immune responses and germinal center B cell responses remains unknow. Previous studies investigating chronic infection with LMCV and HIV revealed that there are two subsets of CD8+ effector T cells, namely CXCR5+ PD-1+ CD8+ T and CXCR5 PD-1+ CD8+ T cells (26,38). The CXCR5+ subset exhibits stem cell-like properties and is localized in the B cell follicle and germinal center during chronic infection. The CXCR5+ subset undergo self-renewal and can differentiate into CXCR5 CD8+ T cells. The de novo converted CXCR5 subset exhibits increased cytotoxicity and removes virus infected cells. Hornquist et al (39) studied the role of CD8+ T cells in the regulation of gut mucosal immune responses and showed that CD8+ T cells inhibited local B cell responses. By contrast, B cells and plasma cells have been shown to impede T cell-mediated antitumor immune responses (4042). Based on the aforementioned findings, gcCD8+ T cells are likely to promote cell-mediate antitumor immune responses and inhibit humoral immunity. A diagrammatic representation based on two hypotheses is presented in Fig. S1. Based on the results obtained from studies investigating chronic HIV or LMCV infection (43,44), it is hypothesized that PD1+ CD8+ TILs may be further divided into CXCR5+ and CXCR5 subsets, which are regulated by the inhibitor of DNA binding 2 (ID2)/transcription factor E2-α axis. The CXCR5+ subset located in the B cell follicle and the germinal center. The follicular CXCR5+ CD8+ T cells subset can undergo proliferation and differentiate into the CXCR5 subset following ID2 upregulation. 2. According to the studies by Hornquist et al (39) and Li et al (44), germinal center CXCR5+ CD8+ T cells can exert a suppressive effect on germinal B cell responses and inhibit the generation of plasma cells. Some mechanisms in these hypotheses were demonstrated. Elimination of immunosuppressive B cells expressing IgA, IL-10 and PD-L1 allows CD8+ cytotoxic T cells eradication of oxaliplatin-resistance prostate cancer (40). The expression of CXCR5 on CD8+ T cells was necessary for T cells infiltrating into B-cells follicular (43). The present study revealed that the phenotype of TILs, TLSs and gcCD8+ TILs exhibited significant heterogeneity in patients with GC. TILs, TLSs and gcCD8+ TILs may be had the potential function associated with GC immunotherapy. Further investigation is required to validate the hypotheses proposed in the present study.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No. 31701111), Key R&D Project of Science and Technology Department of Jiangsu Province (BE2015633). Changzhou Health and Family Planning Commission Youth Talent Science and Technology Project (QN201709). This work was also supported in part by Roswell Park Cancer Institute/University of Pittsburgh Cancer Institute Ovarian Cancer Specialized Programs of Research Excellence Grants (P50CA159981).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

QL, DZ and WH designed the study and wrote the manuscript. DZ, TC, ZY, XG and LC performed all of the experiments. XZ and BX performed the statistical analysis. BL and JJ conceived and designed the study, guided the experiments and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Soochow University, and complied with the Declaration of Helsinki. Informed consent to use the tissue sample for scientific research was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TILs

tumor infiltrating lymphocytes

TLSs

tertiary lymphoid structures

GC

gastric cancer

Tfc

follicular CD8+ cytotoxic T cells

gcCD8+ TILs

germinal center CD8+ TILs

SLO

secondary lymphoid organs

TME

tumor microenvironment

FDC

follicular dendritic cell

FRC

fibroblastic reticular cell

CT

center of the tumor

IM

invasive margin

pTN

pathological tumor and lymph node

AUC

area under the receiver operating charatcteristic curve

References

1 

Chen LJ, Sun J, Wu HY, Zhou SM, Tan Y, Tan M, Shan BE, Lu BF and Zhang XG: B7-H4 expression associates with cancer progression and predicts patient's survival in human esophageal squamous cell carcinoma. Cancer Immunol Immunother. 60:1047–1055. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Chen LJ, Zheng X, Shen YP, Zhu YB, Li Q, Chen J, Xia R, Zhou SM, Wu CP, Zhang XG, et al: Higher numbers of T-bet(+) intratumoral lymphoid cells correlate with better survival in gastric cancer. Cancer Immunol Immunother. 62:553–561. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Xu Y, Chen L, Xu B, Xiong Y, Yang M, Rui X, Shi L, Wu C, Jiang J and Lu B: Higher numbers of T-Bet+ tumor-infiltrating lymphocytes associate with better survival in human epithelial ovarian cancer. Cell Physiol Biochem. 41:475–483. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Zhu Y, Ju S, Chen E, Dai S, Li C, Morel P, Liu L, Zhang X and Lu B: T-bet and eomesodermin are required for T cell-mediated antitumor immune responses. J Immunol. 185:3174–3183. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Pimenta EM and Barnes BJ: Role of tertiary lymphoid structures (TLS) in anti-tumor immunity: Potential tumor-induced cytokines/chemokines that regulate TLS formation in epithelial-derived cancers. Cancers (Basel). 6:969–997. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Singer M, Wang C, Cong L, Marjanovic ND, Kowalczyk MS, Zhang H, Nyman J, Sakuishi K, Kurtulus S, Gennert D, et al: A distinct gene module for dysfunction uncoupled from activation in tumor-infiltrating T cells. Cell. 166:1500–1511.e9. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Dieu-Nosjean MC, Antoine M, Danel C, Heudes D, Wislez M, Poulot V, Rabbe N, Laurans L, Tartour E, de Chaisemartin L, et al: Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol. 26:4410–4417. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, Tosolini M, Camus M, Berger A, Wind P, et al: Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 313:1960–1964. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D, et al: Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 353:2654–2666. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Goc J, Fridman WH, Sautès-Fridman C and Dieu-Nosjean MC: Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology. 2:e268362013. View Article : Google Scholar : PubMed/NCBI

11 

Zhang D, He W, Wu C, Tan Y, He Y, Xu B, Chen L, Li Q and Jiang J: Scoring system for tumor-infiltrating lymphocytes and its prognostic value for gastric cancer. Front Immunol. 10:712019. View Article : Google Scholar : PubMed/NCBI

12 

Martinet L, Filleron T, Le Guellec S, Rochaix P, Garrido I and Girard JP: High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin beta-producing dendritic cells in human breast cancer. J Immunol. 191:2001–2008. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Cipponi A, Mercier M, Seremet T, Baurain JF, Théate I, van den Oord J, Stas M, Boon T, Coulie PG and van Baren N: Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res. 72:3997–4007. 2012. View Article : Google Scholar : PubMed/NCBI

14 

de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, Cremer I, Fridman WH, Sautès-Fridman C and Dieu-Nosjean MC: Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res. 71:6391–6399. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Bergomas F, Grizzi F, Doni A, Pesce S, Laghi L, Allavena P, Mantovani A and Marchesi F: Tertiary intratumor lymphoid tissue in colo-rectal cancer. Cancers (Basel). 4:1–10. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Joshi NS, Akama-Garren EH, Lu Y, Lee DY, Chang GP, Li A, DuPage M, Tammela T, Kerper NR, Farago AF, et al: Regulatory T cells in tumor-Associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity. 43:579–590. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Goc J, Germain C, Vo-Bourgais TK, Lupo A, Klein C, Knockaert S, de Chaisemartin L, Ouakrim H, Becht E, Alifano M, et al: Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 74:705–715. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Gu-Trantien C, Loi S, Garaud S, Equeter C, Libin M, de Wind A, Ravoet M, Le Buanec H, Sibille C, Manfouo-Foutsop G, et al: CD4+ follicular helper T cell infiltration predicts breast cancer survival. J Clin Invest. 123:2873–2892. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Gottlin EB, Bentley RC, Campa MJ, Pisetsky DS, Herndon JE II and Patz EF Jr: The association of intratumoral germinal centers with early-stage non-small cell lung cancer. J Thorac Oncol. 6:1687–1690. 2011. View Article : Google Scholar : PubMed/NCBI

20 

McHeyzer-Williams LJ and McHeyzer-Williams MG: Antigen-specific memory B cell development. Annu Rev Immunol. 23:487–513. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Bos R and Sherman LA: CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes. Cancer Res. 70:8368–8377. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Bos R, Marquardt KL, Cheung J and Sherman LA: Functional differences between low- and high-affinity CD8(+) T cells in the tumor environment. Oncoimmunology. 1:1239–1247. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Kroenke MA, Eto D, Locci M, Cho M, Davidson T, Haddad EK and Crotty S: Bcl6 and Maf cooperate to instruct human follicular helper CD4 T cell differentiation. J Immunol. 188:3734–3744. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Wang C, Hillsamer P and Kim CH: Phenotype, effector function, and tissue localization of PD-1-expressing human follicular helper T cell subsets. BMC Immunol. 12:532011. View Article : Google Scholar : PubMed/NCBI

25 

Petrovas C, Ferrando-Martinez S, Gerner MY, Casazza JP, Pegu A, Deleage C, Cooper A, Hataye J, Andrews S, Ambrozak D, et al: Follicular CD8 T cells accumulate in HIV infection and can kill infected cells in vitro via bispecific antibodies. Science translational medicine. 9:eaag22852017. View Article : Google Scholar : PubMed/NCBI

26 

He R, Hou S, Liu C, Zhang A, Bai Q, Han M, Yang Y, Wei G, Shen T, Yang X, et al: Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature. 537:412–428. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Galon J, Mlecnik B, Bindea G, Angell HK, Berger A, Lagorce C, Lugli A, Zlobec I, Hartmann A, Bifulco C, et al: Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 232:199–209. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Hennequin A, Derangère V, Boidot R, Apetoh L, Vincent J, Orry D, Fraisse J, Causeret S, Martin F, Arnould L, et al: Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. Oncoimmunology. 5:e10545982016. View Article : Google Scholar : PubMed/NCBI

29 

Lee HJ, Park IA, Song IH, Shin SJ, Kim JY, Yu JH and Gong G: Tertiary lymphoid structures: Prognostic significance and relationship with tumour-infiltrating lymphocytes in triple-negative breast cancer. J Clin Pathol. 69:422–430. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Carney JA: Gastric mucosal lymphoid follicles: Histology, distribution, frequency, and etiologic features. Am J Surg Pathol. 34:1019–1024. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Dieu-Nosjean MC, Giraldo NA, Kaplon H, Germain C, Fridman WH and Sautes-Fridman C: Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev. 271:260–275. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Geng Y, Shao Y, He W, Hu W, Xu Y, Chen J, Wu C and Jiang J: Prognostic role of tumor-infiltrating lymphocytes in lung cancer: A meta-analysis. Cell Physiol Biochem. 37:1560–1571. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Li J, Chen L, Xiong Y, Zheng X, Xie Q, Zhou Q, Shi L, Wu C, Jiang J and Wang H: Knockdown of PD-L1 in human gastric cancer cells inhibits tumor progression and improves the cytotoxic sensitivity to CIK therapy. Cell Physiol Biochem. 41:907–920. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Zou W and Chen L: Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 8:467–477. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Lee HJ, Kim JY, Park IA, Song IH, Yu JH, Ahn JH and Gong G: Prognostic significance of tumor-infiltrating lymphocytes and the tertiary lymphoid structures in HER2-positive breast cancer treated with adjuvant trastuzumab. Am J Clin Pathol. 144:278–288. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Gu-Trantien C, Migliori E, Buisseret L, de Wind A, Brohée S, Garaud S, Noël G, Dang Chi VL, Lodewyckx JN, Naveaux C, et al: CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight. 2:e914872017. View Article : Google Scholar

37 

Schweiger T, Berghoff AS, Glogner C, Glueck O, Rajky O, Traxler D, Birner P, Preusser M, Klepetko W and Hoetzenecker K: Tumor-infiltrating lymphocyte subsets and tertiary lymphoid structures in pulmonary metastases from colorectal cancer. Clin Exp Metastasis. 33:727–739. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, et al: Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature. 537:417–421. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Hornquist E, Grdic D, Mak T and Lycke N: CD8-deficient mice exhibit augmented mucosal immune responses and intact adjuvant effects to cholera toxin. Immunology. 87:220–229. 1996. View Article : Google Scholar : PubMed/NCBI

40 

Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D, Willimsky G, Ammirante M, Strasner A, Hansel DE, et al: Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature. 521:94–98. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Shalapour S, Lin XJ, Bastian IN, Brain J, Burt AD, Aksenov AA, Vrbanac AF, Li W, Perkins A, Matsutani T, et al: Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity. Nature. 551:340–345. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Lampropoulou V, Hoehlig K, Roch T, Neves P, Calderón Gómez E, Sweenie CH, Hao Y, Freitas AA, Steinhoff U, Anderton SM and Fillatreau S: TLR-activated B cells suppress T cell-mediated autoimmunity. J Immunol. 180:4763–4773. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Ayala VI, Deleage C, Trivett MT, Jain S, Coren LV, Breed MW, Kramer JA, Thomas JA, Estes JD, Lifson JD and Ott DE: CXCR5-dependent entry of CD8 T cells into rhesus macaque B-Cell follicles achieved through T-cell engineering. J Virol. 91:2017. View Article : Google Scholar : PubMed/NCBI

44 

Li S, Folkvord JM, Rakasz EG, Abdelaal HM, Wagstaff RK, Kovacs KJ, Kim HO, Sawahata R, MaWhinney S, Masopust D, et al: Simian immunodeficiency virus-producing cells in follicles are partially suppressed by CD8+ cells in vivo. J Virol. 90:11168–11180. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Q, Zhang D, He W, Chen T, Yan Z, Gao X, Chen L, Zheng X, Xu B, Lu B, Lu B, et al: CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer. Oncol Lett 20: 2655-2664, 2020
APA
Li, Q., Zhang, D., He, W., Chen, T., Yan, Z., Gao, X. ... Jiang, J. (2020). CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer. Oncology Letters, 20, 2655-2664. https://doi.org/10.3892/ol.2020.11828
MLA
Li, Q., Zhang, D., He, W., Chen, T., Yan, Z., Gao, X., Chen, L., Zheng, X., Xu, B., Lu, B., Jiang, J."CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer". Oncology Letters 20.3 (2020): 2655-2664.
Chicago
Li, Q., Zhang, D., He, W., Chen, T., Yan, Z., Gao, X., Chen, L., Zheng, X., Xu, B., Lu, B., Jiang, J."CD8+ T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer". Oncology Letters 20, no. 3 (2020): 2655-2664. https://doi.org/10.3892/ol.2020.11828