Association between Tim‑3 and Gal‑9 expression and gastric cancer prognosis

  • Authors:
    • Yangyang Wang
    • Enhao Zhao
    • Zizhen Zhang
    • Gang Zhao
    • Hui Cao
  • View Affiliations

  • Published online on: August 6, 2018     https://doi.org/10.3892/or.2018.6627
  • Pages: 2115-2126
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The T‑cell immunoglobulin and mucin domain‑containing protein 3 (Tim‑3)/galectin 9 (Gal‑9) pathway, which serves a pivotal role in immune regulation, is similar to the programmed death (PD)‑1/PD‑ligand 1 pathway. Recent evidence has suggested that Tim‑3 is differentially regulated in a variety of tumors and is a potential therapeutic target. The aim of the present study was to evaluate Tim‑3 and Gal‑9 expression and cluster of differentiation (CD)3+, CD8+ and forkhead box (FOX)p3+ T cell tumor‑infiltration in gastric cancer, as well as their impact on prognosis. Tissue samples from 587 patients with gastric cancer were used to create a tissue microarray (TMA). The immune markers Tim‑3, Gal‑9, CD3, CD8 and FOXp3 were immunostained in the TMA, and correlations with clinicopathological findings and prognosis were analyzed. Several Gene Expression Omnibus gastric cancer databases and the K‑M plotter website were used to analyze the association between the expression of Tim‑3, Gal‑9 and CD8A RNA and patient survival. The results demonstrated that Tim‑3 was mainly expressed in immune cells, with minimal expression in gastric cancer cells. Its ligand, Gal‑9, was significantly overexpressed in tumor cells. Tim‑3 and Gal‑9 expression and Foxp3+ T cell density were negatively associated with the patient overall survival (OS) rate. The density of CD8+ T cells was positively associated with the patient OS rate. Tim‑3 expression and CD8+ T cell density were revealed to be independent prognostic factors for patients with gastric cancer.

Introduction

Gastric cancer (GC) is the fifth most common type of malignancy and the third leading cause of cancer-associated mortality worldwide (1). Half of all cases worldwide occur in East Asia (1). The prognosis of GC is generally poor, particularly in patients with advanced stages of the disease (1). At present, the routine treatment for advanced GC involves surgical resection and perioperative chemotherapy (2). Immunotherapeutic drugs, particularly immune checkpoint inhibitors, including anti-programmed death (PD)1 (nivolumab and pembrolizumab) and anti-cytotoxin T-lymphocyte-associated protein 4 (CTLA4) (ipilimumab) drugs, were recently approved by the Food and Drug Administration to treat a number of solid tumor types. Although there have been successful cases of immune checkpoint inhibitor use in these tumor types, certain patients have been revealed to be resistant to therapies targeting CTLA-4 and PD-1 (3). The T-cell immunoglobulin and mucin domain-containing protein 3 (Tim-3)/galectin 9 (Gal-9) pathway, which is yet to be entirely characterized, may be a potential site of immune checkpoint blockade in immunotherapy, which could be used to overcome drug resistance (3).

Tim-3 was first identified as a molecule expressed in interferon (IFN)-γ-producing cluster of differentiation (CD)4+ T helper 1 (Th1) and CD8+ T cytotoxic 1 (Tc1) cells (4). It has been reported that Tim-3 dysregulation in CD4+ and CD8+ T cells is associated with tumorigenesis (5). When Tim-3 interacts with its ligand, Gal-9, the Th1 response is blocked, resulting in the death of IFN-induced Th1 cells. In a previous study, it was demonstrated that Tim-3 reduces the antigen-specific T-cell response and downregulates antitumor immunity in vivo by inhibiting the Th1 response (6).

Galectins are characterized by their β-galactoside-binding affinity, and have an evolutionarily conserved carbohydrate recognition domain (CRD) (7). Galectins are classified according to their CRD and are subdivided into three groups: Prototype galectins (galectin-1, −2, −7, −10, −13 and −14), chimera-type galectins (galectin-3) (each has a single CRD) and tandem-repeat-type galectins (galectin-4, −8, −9 and −12) (each has two CRDs joined by a flexible peptide linker). Gal-9 is a tandem-repeat-type galectin that is known to serve key roles in eosinophil chemoattraction and activation (8).

Forkhead box (FOX)p3, a member of the forkhead family, is specifically expressed in Treg cells, including CD4+ CD25high Treg cells and CD8+ CD25high Treg cells (9). Treg cells are important factors in the development of immune suppression and tolerance (10). A previous study confirmed that high FOXp3 expression in Tregs allows tumor cells to escape immune surveillance, thereby promoting the proliferation and development of tumor cells (11).

To assess the association between the Tim-3/Gal-9 pathway and GC prognosis, Gal-9 and Tim-3 expression was measured in 587 patients with gastric cancer using Gene Expression Omnibus (GEO) databases and the K-M plotter website. The association between CD3+, CD8+ and FOXp3+ T cell infiltration and GC prognosis was also analyzed in the present study.

Materials and methods

Patients and tissue microarray (TMA) construction

The primary objective of the present study was to evaluate the association between immune characteristics, clinicopathological features and GC prognosis. This was a retrospective analysis of 587 patients with primary GC who underwent gastrectomy at the Department of Gastrointestinal Surgery, Ren Ji Hospital, (School of Medicine, Shanghai Jiao Tong University, Shanghai, China) between January 2006 and December 2011. The final follow-up date was December 31, 2015 for all cases examined. The mean age of the patients was 61.6 years (range, 22–89 years), including 401 males and 186 females. A total of 251 cancer-associated mortalities occurred. All patients received standard treatments, including D2 radical resection [excluding 6 cases with Tumor-Node-Metastasis (TNM) (12) stage IV] and first-line adjuvant chemotherapy (for patients with advanced GC) according to the National Comprehensive Cancer Network guidelines (https://www.nccn.org/). Lauren type was divided into intestinal, diffuse and mixed as previously described (13). The location of the lesion was classified into top, middle and bottom through linking the corresponding trisection points of the lesser gastric curvature and the greater gastric curvature. No patients had received neoadjuvant chemotherapy or human epidermal growth factor receptor 2 targeted therapy. A number of patients with advanced GC did not complete the standard chemotherapy regimen for personal reasons or an inability to tolerate side effects. There was no difference in the number of patients not completing the standard chemotherapy regimen between the Tim-3 high and low expression groups. The following exclusion criteria were used: i) Recurrent GC following radical surgery; ii) receipt of previous neoadjuvant chemotherapy or radiotherapy; iii) the presence of other malignant tumors; and iv) evidence of autoimmune or immunodeficiency diseases.

Formalin-fixed paraffin-embedded (FFPE) tissue blocks were collected from the Pathology Department of Ren Ji Hospital. TNM staging was performed based on the American Joint Committee on Cancer (7th Edition) staging system (12). For each case, the diagnosis was confirmed by two senior pathologists through a review of H&E-stained slides. Representative FFPE blocks were selected for construction of the TMA using a tissue arrayer of 5-µm thickness.

The present study was approved by the Ethics Committee of Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine. Written informed consent was obtained from all enrolled patients prior to their inclusion in the study.

TMA staining, fluorescence-activated cell sorting (FACS) and evaluation

Immunohistochemistry (IHC) was performed on 5-µm thick TMA sections with antibodies specific to Tim-3 (dilution, 1:200; cat. no. 45208; Cell Signaling Technology, Inc., Danvers, MA, USA), Gal-9 (dilution, 1:250; cat. no. ab69630; Abcam, Cambridge, UK), CD3 (dilution, 1:200; cat. no. GB11014; Wuhan Goodbio Technology Co., Ltd., Wuhan, China), CD8 (dilution, 1:100; cat. no. GB11068; Wuhan Goodbio Technology Co., Ltd.) and FOXp3 (dilution, 1:200; cat. no. 98377; Cell Signaling Technology, Inc.). In brief, tissue sections were deparaffinized, rehydrated in a graded ethanol series, and incubated with citrate antigen retrieval solution for 15 min at 95°C and 3% hydrogen peroxide for 30 min at 37°C. Additionally, 1X PBS was used as washing reagent. Tissue sections were blocked with 10% BSA (Sangon Biotech Co., Ltd., Shanghai, China) at room temperature for 1 h. Tissues were subsequently incubated with primary antibodies at 4°C overnight, followed by incubation with a horseradish peroxidase-conjugated rabbit secondary antibody (dilution, 1:20,000; cat. no. 31460; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at room temperature for 1 h. Positive staining was visualized using 3,3′-diaminobenzinidene substrate liquid (Gene Tech Biotechnology Co., Ltd., Shanghai, China) and counterstained with hematoxylin at room temperature for 10 sec. All sections were observed and images were captured using an optical microscope (magnification, ×200 and ×400; Zeiss GmbH, Jena, Germany).

In the subsequent analysis, immune cells in vessels, lymph nodes, lymphatics, necrotic tissue or necrosis-adjacent areas were excluded. Tim-3 was mainly expressed in immune cells with low expression in tumor cells, whereas Gal-9 was more highly expressed in tumor cells. Tumor cell staining was used to evaluate Gal-9 expression. Based on the staining intensity and area, patients were divided into high and low expression groups. Staining intensity was defined as follows: Weak staining, 1; medium staining, 2; and strong staining, 3. The median staining intensity value obtained was multiplied by the percentage of the tissue area that was stained. Nikon DR-Si2 cell counting and imaging software (version 4.30.01; Nikon DR-Si2; Nikon Corporation, Tokyo, Japan) was used for this analysis. A total of 4 randomly selected fields of view (magnification, ×200; 0.34 mm2) were used to analyze each case, and the average number of Tim-3+, CD3+, CD8+ and FOXp3+ cells was counted. According to the median number of stained immune cells (Tim-3) or T cells (CD3 and CD8), patients were divided into low and high infiltration groups as previously described (14). To evaluate Foxp3+ T cells, positive staining was defined as >5 stained cells/high-power field (HPF) and negative staining was defined as ≤5 cells/HPF. The results were verified by two senior pathologists who were blinded to the clinicopathological data of the patients.

Detection of Tim-3 expression in CD3+ T cells by FACS staining

Surgical resection of tumor tissues from one male patient with GC (age, 62 years; TNM stage III) was performed in the present study. A total of 1 g dissociated tissue was treated with collagenase IV (cat. no. C5138; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 90 min at 37°C on a rotating shaker. The homogenates were filtered through membranes of 48 µm (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) to remove cell mass and fiber agglomerates. Tumor tissue-derived single cells were obtained by centrifugation (400 × g at room temperature for 10 min) and suspended in 1X PBS. Subsequently, the cells were incubated with a mouse anti-human Tim-3 antibody (PE-CF594; cat. no. 565561; dilution, 1:200; BD Biosciences, Franklin Lakes, NJ, USA) and a fluorescein isothiocyanate-conjugated CD3 antibody (cat. no. ab 210316; dilution, 1:200, Abcam) for 30 min at 4°C in a dark environment. Following washing in 1X PBS, Tim-3 expression was detected in CD3+ T cells using a flow cytometer (BD Biosciences). CD3 was considered to be a molecular marker of T cells in the present study. FlowJo version 10 software was used for analysis (FlowJo LLC, Ashland, OR, USA).

Cell culture and reagents

The human gastric cancer AGS, BGC-823, HGC-27, MGC-803, MKN-45 and SGC-7901 cell lines were all maintained in the Shanghai Cancer Institute, Ren Ji Hospital. All cells were cultured in RPMI-1640 medium (Beijing Solarbio Science & Technology Co., Ltd.) or F-12 Medium (Gibco; Thermo Fisher Scientific, Inc.) according to ATCC protocols and supplemented with 10% (v/v) fetal bovine serum (FBS) and 1% antibiotics (100 µg/ml streptomycin and 100 U/ml penicillin) at 37°C in a humidified incubator at 5% CO2. The cell medium was replaced every 2–3 days, and 1X PBS was used for cell washing prior to the medium being replaced. Cells were collected in the logarithmic growth phase for protein and RNA extraction.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

The thermocycling conditions were as follows: 60°C for 34 sec and 95°C for 15 sec for 40 cycles. Total RNA was extracted from AGS, BGC-823, HGC-27, MGC-803, MKN-45 and SGC-7901 cells using TRIzol reagent (Thermo Fisher Scientific, Inc.) and reverse transcribed using a PrimeScript RT-PCR kit (Takara Bio., Inc., Otsu, Japan), according to the manufacturer's protocols. RT-qPCR was performed with SYBR Premix Ex Taq (Takara Bio., Inc.) using a 7500 Real-time PCR system (Thermo Fisher Scientific, Inc.). The primer sequences used in the present study were as follows: Tim-3 forward, 5′-CTGCTGCTACTACTTACAAGGTC-3′ [melting temperature (Tm)=60.1°C] and reverse, 5′-GCAGGGCAGATAGGCATTCT-3′ (Tm=61.8°C); Gal-9 forward, 5′-TCTGGGACTATTCAAGGAGGTC-3′ (Tm=60.3°C) and reverse, 5′-CCACTGGAGCTGAGAACGG-3′ (Tm=62.0°C); and β-actin forward, 5′-CATGTACGTTGCTATCCAGGC-3′ (Tm=60.8°C) and reverse, 5′-CTCCTTAATGTCACGCACGAT-3′ (Tm=60.2°C). The 2−∆ΔCq method (15) was used to quantify relative Tim-3 and Gal-9 expression, which was normalized to β-actin.

Western blotting

Total protein was extracted using a total protein extraction buffer (Beyotime Institute of Biotechnology, Haimen, China) and the protein concentration was measured using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc.). Proteins (30 µg per lane) were separated using 10% SDS-PAGE and transferred onto a nitrocellulose membrane. Following blocking with 1% BSA at room temperature for 1 h, the membrane was probed with Tim-3 (dilution, 1:1,000; cat. no. 45208; Cell Signaling Technology, Inc.), Gal-9 (dilution, 1:1,000; cat. no. ab69630; Abcam) or β-actin (dilution, 1:1,000; cat. no. 20536-1-AP; ProteinTech Group, Inc., Chicago, IL, USA) primary antibodies overnight at 4°C, and a rabbit immunoglobulin G (H+L) secondary antibody (dilution, 1:10,000; cat. no. 31460; Thermo Fisher Scientific, Inc.) at room temperature for 1 h. Proteins were visualized using the Molecular Imager ChemiDoc XRS+ System (Bio-Rad Laboratories, CA, USA).

Analysis using K-M plotter

To analyze the association between immune marker expression and patient prognosis, a number of GEO databases (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi) regarding Tim-3, Gal-9 and CD8A expression levels in GC and the K-M plotter website (http://kmplot.com/analysis/) (16) were consulted. The following GEO datasets were used: gse14210 (17), gse15459 (18), gse22377 (19), gse29272 (20), gse51105 (21) and gse62254 (22).

Statistical analysis

Associations between Tim-3 expression and clinicopathological factors were analyzed using the χ2 test or Fisher's exact test. Survival analysis was performed using the Kaplan-Meier method and the log-rank test. Univariate and multivariate analyses were conducted using the Cox proportional hazards model to identify prognostic factors. All statistical tests were two-sided and P<0.05 was considered to indicate a statistically significant difference. Statistical analyses were performed using SPSS 13.0 statistical package software (SPSS, Inc., Chicago, IL, USA) or GraphPad Prism (GraphPad Software Inc., La Jolla, CA, USA).

Results

Clinicopathological features of patients with GC

To study the association between Tim-3 expression and patient clinicopathological features, patient age, sex, tumor location, tumor diameter, Lauren type, perineuronal invasion, blood-vessel invasion and TNM stage were analyzed. Follow-up information was available for all 587 patients. The follow-up time ranged between 1 and 117 months following surgery, with a median follow-up time of 48 months.

Association between patient prognosis and immune characteristics

For immune characteristic analysis, immunohistochemical staining was performed on the TMA for immune checkpoint Tim-3 and Gal-9, as well as T-cell markers, CD3 (tumor infiltrating lymphocytes, TILs), CD8 (cytotoxic T lymphocytes, CTLs) and FOXp3 (T regulatory cells, Tregs). Images of Tim-3 and Gal-9 expression are shown in Fig. 1A and B. The majority of the GC tissues exhibited cytoplasmic and extracellular Gal-9 staining. However, few gastric cancer cells exhibited Tim-3 staining, which is strongly expressed in infiltrating immune cells. Considerable infiltration of the GC parenchyma by CD3+ and CD8+ T cells was evident in the patients with a favorable prognosis The number of FOXp3+ T cells that infiltrated the tumor parenchyma was less than that of the other types of T cells (Fig. 1C-E). FACS was performed in order to detect Tim-3 expression in CD3+ T cells. The results showed that 16.8% of CD3+ T cells exhibited notable expression of Tim-3 protein. Therefore, it was demonstrated that Tim-3 is notably expressed in the tumor-infiltrating lymphocytes of patients with GC (Fig. 1F).

The present study analyzed the association between Tim-3 expression in immune cells in patients with GC and patient prognosis. It was revealed that the expression of Tim-3 in immune cells was significantly associated with patient survival. Increased Tim-3+ immune cell infiltration into the tumor was associated with a lower OS rate (P=0.001; Fig. 2A). Gal-9 expression in GC was also compared with patient prognosis. Patients exhibiting high Gal-9 expression had a relatively poor prognosis and a poorer OS rate (P=0.0028; Fig. 2B). As Gal-9 is a ligand of Tim-3, combined analysis involving the expression of the two factors was performed. Patients with low Tim-3 and Gal-9 expression had a significantly greater OS rate than other patients (P<0.001; Fig. 2C).

TIL, CTL and Treg infiltration into the tumor parenchyma was analyzed by measuring the number of CD3+, CD8+ and FOXp3+ T cells. CD3+ T-cell density in tumor tissues was not associated with OS rate (P=0.0776; Fig. 3A), while CD8+ T cell density in tumor tissue was positively associated with OS rate (P=0.0395; Fig. 3B). However, a high density of FOXp3+ T cells in tumor tissue was associated with a poorer OS rate (P=0.0164; Fig. 3C).

Association between patient clinicopathological parameters and immune characteristics

It was demonstrated that age and tumor diameter were significantly different in the Tim-3 high and low expression groups. Tumor location and blood vessel invasion were significantly different in the Gal-9 high and low expression groups. The Tim-3 or Gal-9 expression status was positively associated with N stage and TNM stage (Tables I and II). However, in terms of the association between TIL, CTL and Tregs density, and patient clinicopathological parameters, only tumor diameter was significantly different between the CTL high- and low-density groups (Tables IIIV).

Table I.

Association between tumor Tim-3 expression and clinicopathological parameters in patients with gastric cancer.

Table I.

Association between tumor Tim-3 expression and clinicopathological parameters in patients with gastric cancer.

Tim-3 expression

Clinicopathological featureLow (n=294)High (n=293)P-value
Age, years
  ≤65 (n=371)1981730.0370a
  >65 (n=216)96120
Sex
  Male (n=401)1992020.7439
  Female (n=186)9591
Tumor location
  Top (n=118)63550.7212
  Middle (n=202)99103
  Bottom (n=267)132135
Diameter, cm
  ≤5 (n=375)2021730.0148a
  >5 (n=212)92120
Pathological Lauren type
  Intestinal (n=194)106880.1715
  Diffuse (n=326)152174
  Mixed (n=67)3631
Perineuronal invasion
  Negative (n=518)2632550.3617
  Positive (n=69)3138
Blood vessel invasion
  Negative (n=493)2532400.1711
  Positive (n=94)4153
pT stage
  T1 (n=94)56380.0795
  T2 (n=80)4436
  T3 (n=151)6685
  T4 (n=262)128134
pN stage
  N0 (n=234)1331010.0478a
  N1 (n=105)5055
  N2 (n=114)5460
  N3 (n=134)5777
TNM stage
  I (n=137)81560.0221a
  II (n=165)8580
  III (n=279)127152
  IV (n=6)15

a P<0.05. Tim-3, T-cell immunoglobulin and mucin domain-containing protein 3; TNM, Tumor-Node-Metastasis.

Table II.

Association between tumor Gal-9 expression and clinicopathological parameters in patients with gastric cancer.

Table II.

Association between tumor Gal-9 expression and clinicopathological parameters in patients with gastric cancer.

Gal-9 expression

Clinicopathological featureLow (n=294)High (n=293)P-value
Age, years
  ≤65 (n=371)1941770.1613
  >65 (n=216)100116
Sex
  Male (n=401)2021990.8327
  Female (n=186)9294
Tumor location
  Top (n=118)72460.0291a
  Middle (n=202)95107
  Bottom (n=267)127140
Diameter, cm
  ≤5 (n=375)1991760.0547
  >5 (n=212)95117
Pathological Lauren type
  Intestinal (n=194)106880.0841
  Diffuse (n=326)150176
  Mixed (n=67)3829
Perineuronal invasion
  Negative (n=518)2632550.3617
  Positive (n=69)3138
Blood vessel invasion
  Negative (n=493)2562370.0410a
  Positive (n=94)3856
pT stage
  T1 (n=94)49450.3397
  T2 (n=80)4139
  T3 (n=151)6685
  T4 (n=262)138124
pN stage
  N0 (n=234)1341000.0060b
  N1 (n=105)5550
  N2 (n=114)5361
  N3 (n=134)5282
TNM stage
  I (n=137)75620.0292a
  II (n=165)9471
  III (n=279)123156
  IV (n=6)24

a P<0.05

b P<0.01. Gal-9, galectin 9; TNM, Tumor-Node-Metastasis.

Table III.

Association between tumor CD3 expression and clinicopathological parameters in patients with gastric cancer.

Table III.

Association between tumor CD3 expression and clinicopathological parameters in patients with gastric cancer.

CD3 expression

Clinicopathological featureLow (n=294)High (n=293)P-value
Age, years
  ≤65 (n=371)1831880.6298
  >65 (n=216)111105
Sex
  Male (n=401)2031980.7017
  Female (n=186)9195
Tumor location
  Top (n=118)61570.8201
  Middle (n=202)10399
  Bottom (n=267)130137
Diameter, cm
  ≤5 (n=375)1811940.2412
  >5 (n=212)11399
Pathological Lauren type
  Intestinal (n=194)106880.3006
  Diffuse (n=326)156170
  Mixed (n=67)3235
Perineuronal invasion
  Negative (n=518)2562620.3778
  Positive (n=69)3831
Blood-vessel invasion
  Negative (n=493)2452480.6656
  Positive (n=94)4945
pT stage
  T1 (n=94)41530.1804
  T2 (n=80)3743
  T3 (n=151)7279
  T4 (n=262)144118
pN stage
  N0 (n=234)1161180.3688
  N1 (n=105)4857
  N2 (n=114)6549
  N3 (n=134)6569
TNM stage
  I (n=137)63740.6838
  II (n=165)8283
  III (n=279)146133
  IV (n=6)33

[i] CD, cluster of differentiation; TNM, Tumor-Node-Metastasis.

Table V.

Association between tumor FOXp3 expression and clinicopathological parameters in patients with gastric cancer.

Table V.

Association between tumor FOXp3 expression and clinicopathological parameters in patients with gastric cancer.

FOXp3 expression

Clinicopathological featureNegative (n=481)Positive (n=106)P-value
Age, years
  ≤65 (n=371)309620.2664
  >65 (n=216)17244
Sex
  Male (n=401)332690.4313
  Female (n=186)14937
Tumor location
  Top (n=118)95230.7776
  Middle (n=202)16438
  Bottom (n=267)22245
Diameter, cm
  ≤5 (n=375)304710.4634
  >5 (n=212)17735
Pathological Lauren type
  Intestinal (n=194)165290.0785
  Diffuse (n=326)25769
  Mixed (n=67)598
Perineuronal invasion
  Negative (n=518)427910.3974
  Positive (n=69)5415
Blood-vessel invasion
  Negative (n=493)408850.2389
  Positive (n=94)7321
pT stage
  T1 (n=94)84100.1746
  T2 (n=80)6713
  T3 (n=151)12229
  T4 (n=262)20854
pN stage
  N0 (n=234)197370.7098
  N1 (n=105)8520
  N2 (n=114)9222
  N3 (n=134)10727
TNM stage
  I (n=137)120170.1364
  II (n=165)13332
  III (n=279)22257
  IV (n=6)60

[i] FOXp3, forkhead box p3; TNM, Tumor-Node-Metastasis.

Association between biomarker expression levels

CD3+ T cell density in tumor tissue was correlated with CD8+ T cell density (r=0.6281, P<0.0001, Fig. 3D). However, there was no correlation between Tim-3+ immune cell density and CD3+ or CD8+ T cell density (r=0.2405, P<0.0001 and r=0.1550, P=0.0002, respectively; Fig. 3E and F).

K-M plotter analysis

Tim-3, Gal-9 and CD8A mRNA expression was compared with patient survival using the following GEO databases on the K-M plotter website: gse14210, gse15459, gse22377, gse29272, gse51105 and gse62254. High Tim-3 expression in patients with GC was associated with relatively short survival times (Fig. 4A and B). High Gal-9 expression in patients with GC was also associated with relatively short survival times (Fig. 4C). High CD8A expression was associated with relatively long survival times (Fig. 4D).

Expression of Tim-3 and Gal-9 in GC cell lines

To detect Tim-3 and Gal-9 expression in tumor cell lines, western blotting and RT-qPCR were performed using AGS, BGC-823, HGC-27, MGC-803, MKN-45 and SGC-7901 cells. GC cells exhibited low expression levels of Tim-3 but relatively high Gal-9 expression (Fig. 4E-G).

CD8+ T cell density and Tim-3+ immune cell infiltration are independent prognostic factors in GC

Table VI shows the univariate and multivariate analysis of clinicopathological factors. The univariate analysis revealed that the following factors were significantly associated with patient postoperative survival: CD8+ T cell density (P=0.025), FOXp3+ T cell density (P=0.017), Tim-3+ immune cell density (P<0.0001), Gal-9 expression (P=0.002), age (P=0.009), tumor diameter (P<0.0001), Lauren type (P=0.001), perineuronal invasion (P<0.0001), blood vessel invasion (P<0.0001) and TNM stage (P<0.0001). Multivariate regression analysis indicated that CD8+ T cell density (P=0.031), Tim-3+ immune cell density (P=0.012), tumor diameter (P=0.001), blood vessel invasion (P=0.003) and TNM stage (P<0.001) were independent prognostic factors in GC.

Table VI.

Univariate and multivariate analysis of prognostic parameters for the overall survival of patients with gastric cancer.

Table VI.

Univariate and multivariate analysis of prognostic parameters for the overall survival of patients with gastric cancer.

Univariate analysisMultivariate analysis


Prognostic parameterHR95% CIP-valueHR95% CIP-value
Age1.3971.087–1.7970.009b1.0840.826–1.4240.559
Sex1.1540.889–1.4990.281
Tumor location
  Top1Reference
  Middle0.8240.594–1.1440.248
  Bottom0.8580.608–1.2110.383
Diameter0.3580.279–0.459 <0.001b0.6230.474–0.8180.001b
Lauren type (intestinal/mixed vs. diffuse)1.6131.218–2.1370.001b1.1200.837–1.4990.445
Perineuronal invasion0.4410.318–0.612 <0.001b0.7850.557–1.1050.165
Blood vessel invasion0.3700.277–0.493 <0.001b0.6200.455–0.8460.003b
TNM stage0.2130.160–0.283 <0.001b0.2950.217–0.402 <0.001b
CD30.7810.609–1.0010.051
CD80.7530.587–0.9660.025a0.7550.585–0.9750.031a
FOXp31.4391.067–1.9410.017a1.1760.887–1.5590.260
Tim-31.7101.327–2.203 <0.001b1.3951.078–1.8070.012a
Gal-91.4891.160–1.9120.002b1.2050.933–1.5560.153

a P<0.05

b P<0.01. HR, hazard ratio; CI, confidence interval; TNM, Tumor-Node-Metastasis; CD, cluster of differentiation; FOXp3, forkhead box p3; Tim-3, T-cell immunoglobulin and mucin domain-containing protein 3; Gal-9, galectin 9.

Discussion

At present, the treatment options available for GC are limited, particularly for patients with advanced stages of disease (23). In China, early diagnosis remains problematic and therefore the majority of patients have advanced GC at the time of diagnosis (23). This is likely because gastroendoscopy is not performed as regularly in China as in other developed countries (23). The current conventional treatments used for GC include surgery and perioperative chemotherapy (2). Immunotherapies, particularly immune checkpoint inhibitors, have great potential as effective treatments for GC in the future and have been used successfully to treat other solid tumors.

Tim-3 is selectively expressed in IFN-γ-producing CD4+ Th1 and CD8+ Tc1 cells (4). Previous studies have demonstrated that Tim-3 is required to induce immune tolerance, as Tim-3-deficient mice and mice treated with a Tim-3 Ig protein had defects in the induction of antigen-specific tolerance (24,25). Tim-3, as an immune checkpoint receptor, limits the duration and magnitude of the Th1 and Tc1 T-cell response. In the present study, Tim-3-positive immune cells were demonstrated to infiltrate GC tissue. GC cells rarely express Tim-3 protein but Tim-3 expression in tumor cells has been previously reported (26); however, the majority of previous studies have indicated that Tim-3+ cells are immune cells, including T cells, macrophages and dendritic cells (2729). In the present study, a significant association between Tim-3+-cell tumor infiltration, and tumor diameter and TNM stage was observed. Furthermore, the survival of patients exhibiting high Tim-3+-cell tumor infiltration was significantly lower than that in those exhibiting low infiltration. Univariate and multivariate analyses also revealed that Tim-3+ infiltrating immune cells in tumors were associated with a poor prognosis. The mechanism behind this association is not yet fully understood. However, it is possible that the Tim-3/Gal-9 pathway downregulates T-cell responses by mediating apoptosis. A recent study revealed a negative regulatory effect by Tim-3 expression on CD4+ and CD8+ T cell viability (30). Furthermore, the Tim-3/Gal-9 pathway contributes toward the suppressive tumor microenvironment via Treg activation upon TCR activation (31). Expression of the Tim-3 surface protein by CD8+ T cells provides immune tolerance upon encounter of cancer cells highly expressing Gal-9.

Gal-9, a galectin protein, regulates the survival, proliferation and cytokine synthesis of effector helper and cytotoxic T cells (32). Gal-9 has recently become a major molecule of interest due to the identification of its negative influence on the adaptive immune response. In the present study, Gal-9 was demonstrated to be highly expressed in GC and subcellularly localized to the extracellular area and cytoplasm of tumor cells. A significant association between Gal-9 expression, blood vessel invasion and TNM stage in GC was observed. Additionally, high expression of Gal-9 was associated with poor patient survival. Gal-9 and other galectins have been indicated to be prognostic markers in other types of cancer (3335). Tim-3/Gal-9 binding interactions inhibit Th17 polarization, driving the proliferation of FOXp3+ Tregs (26). Tim-3/Gal-9 binding can also induce apoptosis or necrosis in pro-inflammatory T cell subsets (37). While Gal-9 and other galectins induce pro-apoptotic features in pro-inflammatory T cell subsets, they function through Bcl-2 blocking Gal-9-induced apoptosis (34). In vitro studies using lung cancer-specific T-cell lines revealed that apoptosis is induced in Tim-3+ CD8+ T-cell clones following interaction with Gal-9, which can be inhibited by the addition of anti-Gal-9 or anti-Tim-3 antibodies. The release of soluble Gal-9 can therefore negatively regulate T-cell function or induce apoptosis via Tim-3 (35). Another study demonstrated that Gal-9 induces Tim-3+ Th1, Th17 and Tc1 T cell apoptosis in hyperimmune conditions (36). However, the function of Gal-9 in breast cancer, cervical carcinoma and malignant melanoma has also been reported, which is in contrast to the results of the present study (3840). This may be due to individual variation in the immune state, including differing Tim-3 protein expression and cytokine levels. Univariate analysis demonstrated that Gal-9 overexpression in tumor cells is associated with a poor prognosis in GC. However, multivariate analysis did not reveal any statistical significance, suggesting that Gal-9 functions as a tumorigenesis promoter depending on the immune state of the patient. High expression of Tim-3 and Gal-9 was associated with a poor survival in patients with GC.

Regarding CD3+ and CD8+ T cell infiltration in tumors and survival in patients with GC, previous studies have reported an association between high CD3+ and CD8+ T cell density and a favorable prognosis (41,42). The same results were observed in the present study; the density of CD8+ T cells in GC tumors was associated with tumor size. Univariate and multivariate analyses also revealed that high CD8+ T cell infiltration was associated with a good prognosis. CTLs are also associated with a good prognosis. In adaptive immunity, CTLs directly kill tumor cells in the tumor microenvironment. Adaptive immunity, which is mediated by T cells, has been suggested to serve a major role in antitumor immunity (42).

Forkhead box protein 3 (FOXp3), a member of the forkhead transcription factor family, is considered to be a distinctive molecular marker of regulatory T cells (43). It has been suggested that Tregs can suppress the majority of immune cells, including CD4+ and CD8+ T cells, B cells, natural killer cells and NK T cells. Tregs also inhibit the proliferation of effector T cells, reduce cytokine secretion, promote B-cell anergy, impede antibody production, inhibit the expression of co-stimulatory and antigen-presenting molecules, and reduce the ability to stimulate T cell responses (44). A high frequency of Tregs is generally considered to be a marker of poor prognosis in various types of cancer. Tregs may mediate the suppression of antitumor immunity, which promotes tumor growth. The present study revealed a high level of Tregs infiltrating the tumor tissue, which was associated with a poor survival in patients with GC. A previous study demonstrated that enhanced FOXp3+ T cells expression was associated with a low OS rate and a poor prognosis (45). A high density of Tregs in tumors and peripheral blood is therefore generally considered to be a marker of a poor prognosis in cancer.

The present study has a number of limitations. The IHC of the TMA may not completely represent the immune marker expression. Previous studies have used large areas of tissue for immunostaining (46). Tim-3 is mainly expressed in T cells but is also expressed in other immune cells. Future studies should aim to define the detailed function of Tim-3 and the immune cells in which it is expressed in GC.

While two previous studies have investigated the association between Tim-3 expression on T cells and NK cells, and patient clinicopathological features (47,48). However, the present study compared its ligand Gal-9 expression and T-cell infiltration in tumor tissues with patient clinicopathological features and survival. The large patient sample size made the conclusions of the present study more robust. The association between Tim-3 expression and Gal-9 expression was also discussed in the present study. Tim-3 is primarily expressed by infiltrating T cells, while Gal-9 is expressed by GC cells. A large number of TILs and CTLs were revealed to infiltrate GC, and CTL infiltration was associated with OS. Univariate and multivariate analyses revealed that Tim-3 expression and CD8+ T cell density in tumors are associated with GC prognosis and can act as independent prognostic factors. However, the mechanism by which Tim-3 functions in GC and its interaction with Gal-9 remain unclear and require investigation in the future. Finally, the Tim-3/Gal-9 pathway may be a valuable immunotherapy target for the treatment of GC and other types of solid tumor.

Acknowledgements

The authors would like to thank members of the Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University (Shanghai, China) for providing assistance.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81272743).

Availability of data and materials

The datasets generated and analyzed during the study are available in Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/pubmed) and Kaplan Meier-Plotter website (http://kmplot.com/analysis/index.php?p=service&cancer=gastric). Other datasets used in the study are available from the corresponding author upon reasonable request.

Authors' contributions

HC and GZ conceived and designed the study. YW, EZ and ZZ performed the experiments. YW wrote the study. HC, GZ, EZ and ZZ reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine (Shanghai, China). Written informed consent was obtained from all enrolled patients prior to their inclusion in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–386. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Japanese Gastric Cancer Association: Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric cancer. 20:1–19. 2017. View Article : Google Scholar

3 

Anderson AC: Tim-3: An emerging target in the cancer immunotherapy landscape. Cancer Immunol Res. 2:393–398. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, Manning S, Greenfield EA, Coyle AJ, Sobel RA, et al: Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 415:536–541. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Shin DS and Ribas A: The evolution of checkpoint blockade as a cancer therapy: What's here, what's next? Curr Opin Immunol. 33:23–35. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Geng H, Zhang GM, Li D, Zhang H, Yuan Y, Zhu HG, Xiao H, Han LF and Feng ZH: Soluble form of T cell Ig mucin 3 is an inhibitory molecule in T cell-mediated immune response. J Immunol. 176:1411–1420. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Yang J, Zhu L, Cai Y, Suo J and Jin J: Role of downregulation of galectin-9 in the tumorigenesis of gastric cancer. Int J Oncol. 45:1313–1320. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Takano J, Morishita A, Fujihara S, Iwama H, Kokado F, Fujikawa K, Fujita K, Chiyo T, Tadokoro T, Sakamoto T, et al: Galectin-9 suppresses the proliferation of gastric cancer cells in vitro. Oncol Rep. 35:851–860. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG and Rudensky AY: Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 22:329–341. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Sakaguchi S, Yamaguchi T, Nomura T and Ono M: Regulatory T cells and immune tolerance. Cell. 133:775–787. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Sun X, Feng Z, Wang Y, Qu Y and Gai Y: Expression of Foxp3 and its prognostic significance in colorectal cancer. Int J Immunopathol Pharmacol. 30:201–206. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Washington K: 7th edition of the AJCC cancer staging manual: Stomach. Ann Surg Oncol. 17:3077–3079. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Van Cutsem E, Sagaert X, Topal B, Haustermans K and Prenen H: Gastric cancer. Lancet. 388:2654–2664. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Nosho K, Baba Y, Tanaka N, Shima K, Hayashi M, Meyerhardt JA, Giovannucci E, Dranoff G, Fuchs CS and Ogino S: Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: Cohort study and literature review. J Pathol. 222:350–366. 2010. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Szasz AM, Lanczky A, Nagy Á, Förster S, Hark K, Green JE, Boussioutas A, Busuttil R, Szabó A and Győrffy B: Cross-validation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget. 7:49322–49333. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Kim HK, Choi IJ, Kim CG, Kim HS, Oshima A, Michalowski A and Green JE: A gene expression signature of acquired chemoresistance to cisplatin and fluorouracil combination chemotherapy in gastric cancer patients. PLoS One. 6:e166942011. View Article : Google Scholar : PubMed/NCBI

18 

Ooi CH, Ivanova T, Wu J, Lee M, Tan IB, Tao J, Ward L, Koo JH, Gopalakrishnan V, Zhu Y, et al: Oncogenic pathway combinations predict clinical prognosis in gastric cancer. PLoS Genetics. 5:e10006762009. View Article : Google Scholar : PubMed/NCBI

19 

Forster S, Gretschel S, Jons T, Yashiro M and Kemmner W: THBS4, a novel stromal molecule of diffuse-type gastric adenocarcinomas, identified by transcriptome-wide expression profiling. Mod Pathol. 24:1390–1403. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Wang G, Hu N, Yang HH, Wang L, Su H, Wang C, Clifford R, Dawsey EM, Li JM, Ding T, et al: Comparison of global gene expression of gastric cardia and noncardia cancers from a high-risk population in china. PLoS One. 8:e638262013. View Article : Google Scholar : PubMed/NCBI

21 

Busuttil RA, George J, Tothill RW, Ioculano K, Kowalczyk A, Mitchell C, Lade S, Tan P, Haviv I and Boussioutas A: A signature predicting poor prognosis in gastric and ovarian cancer represents a coordinated macrophage and stromal response. Clin Cancer Res. 20:2761–2772. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Cristescu R, Lee J, Nebozhyn M, Kim KM, Ting JC, Wong SS, Liu J, Yue YG, Wang J, Yu K, et al: Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 21:449–456. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Shen L, Shan YS, Hu HM, Price TJ, Sirohi B, Yeh KH, Yang YH, Sano T, Yang HK, Zhang X, et al: Management of gastric cancer in Asia: Resource-stratified guidelines. Lancet Oncol. 14:e535–e547. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Sanchez-Fueyo A, Tian J, Picarella D, Domenig C, Zheng XX, Sabatos CA, Manlongat N, Bender O, Kamradt T, Kuchroo VK, et al: Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol. 4:1093–1101. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Sabatos CA, Chakravarti S, Cha E, Schubart A, Sánchez-Fueyo A, Zheng XX, Coyle AJ, Strom TB, Freeman GJ and Kuchroo VK: Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol. 4:1102–1110. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Jiang J, Jin MS, Kong F, Cao D, Ma HX, Jia Z, Wang YP, Suo J and Cao X: Decreased galectin-9 and increased Tim-3 expression are related to poor prognosis in gastric cancer. PLoS One. 8:e817992013. View Article : Google Scholar : PubMed/NCBI

27 

Xu Y, Zhang H, Huang Y, Rui X and Zheng F: Role of TIM-3 in ovarian cancer. Clin Transl Oncol. 19:1079–1083. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Ngiow SF, Teng MW and Smyth MJ: Prospects for TIM3-targeted antitumor immunotherapy. Cancer Res. 71:6567–6571. 2011. View Article : Google Scholar : PubMed/NCBI

29 

da Silva IP, Gallois A, Jimenez-Baranda S, Khan S, Anderson AC, Kuchroo VK, Osman I and Bhardwaj N: Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res. 2:410–422. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Anderson AC: Tim-3, a negative regulator of anti-tumor immunity. Curr Opin Immunol. 24:213–216. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, Zhou T, Li Y, Guo X, Xiao H, et al: T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol. 190:2068–2079. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Liu FT and Rabinovich GA: Galectins as modulators of tumour progression. Nat Rev Cancer. 5:29–41. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Szoke T, Kayser K, Baumhakel JD, Trojan I, Furak J, Tiszlavicz L, Horvath A, Szluha K, Gabius HJ and Andre S: Prognostic significance of endogenous adhesion/growth-regulatory lectins in lung cancer. Oncology. 69:167–174. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Cedeno-Laurent F and Dimitroff CJ: Galectins and their ligands: Negative regulators of anti-tumor immunity. Glycoconj J. 29:619–625. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Ohue Y, Kurose K, Nozawa R, Isobe M, Nishio Y, Tanaka T, Doki Y, Hori T, Fukuoka J, Oka M and Nakayama E: Survival of lung adenocarcinoma patients predicted from expression of PD-L1, Galectin-9, and XAGE1 (GAGED2a) on tumor cells and tumor-infiltrating T cells. Cancer Immunol Res. 4:1049–1060. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Seki M, Oomizu S, Sakata KM, Sakata A, Arikawa T, Watanabe K, Ito K, Takeshita K, Niki T, Saita N, et al: Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol. 127:78–88. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, Zheng XX, Strom TB and Kuchroo VK: The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 6:1245–1252. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Yamauchi A, Kontani K, Kihara M, Nishi N, Yokomise H and Hirashima M: Galectin-9, a novel prognostic factor with antimetastatic potential in breast cancer. Breast J. 12 5 Suppl 2:S196–S200. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Liang M, Ueno M, Oomizu S, Arikawa T, Shinonaga R, Zhang S, Yamauchi A and Hirashima M: Galectin-9 expression links to malignant potential of cervical squamous cell carcinoma. J Cancer Res Clin Oncol. 134:899–907. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Kageshita T, Kashio Y, Yamauchi A, Seki M, Abedin MJ, Nishi N, Shoji H, Nakamura T, Ono T and Hirashima M: Possible role of galectin-9 in cell aggregation and apoptosis of human melanoma cell lines and its clinical significance. Int J Cancer. 99:809–816. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Kawazoe A, Kuwata T, Kuboki Y, Shitara K, Nagatsuma AK, Aizawa M, Yoshino T, Doi T, Ohtsu A and Ochiai A: Clinicopathological features of programmed death ligand 1 expression with tumor-infiltrating lymphocyte, mismatch repair, and Epstein-Barr virus status in a large cohort of gastric cancer patients. Gastric Cancer. 20:407–415. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Lee HE, Chae SW, Lee YJ, Kim MA, Lee HS, Lee BL and Kim WH: Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. Br J Cancer. 99:1704–1711. 2008. View Article : Google Scholar : PubMed/NCBI

43 

de Reuver PR, Mehta S, Gill P, Andrici J, D'Urso L, Clarkson A, Mittal A, Hugh TJ, Samra JS and Gill AJ: Immunoregulatory forkhead box protein p3-positive lymphocytes are associated with overall survival in patients with pancreatic neuroendocrine tumors. J Am Coll Surg. 222:281–287. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Frydrychowicz M, Boruczkowski M, Kolecka-Bednarczyk A and Dworacki G: The dual role of Treg in cancer. Scand J Immunol. 86:436–443. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Hou J, Yu Z, Xiang R, Li C and Wang L, Chen S, Li Q, Chen M and Wang L: Correlation between infiltration of FOXP3+ regulatory T cells and expression of B7-H1 in the tumor tissues of gastric cancer. Exp Mol Pathol. 96:284–291. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Cimino-Mathews A, Thompson E, Taube JM, Ye X, Lu Y, Meeker A, Xu H, Sharma R, Lecksell K, Cornish TC, et al: PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum Pathol. 47:52–63. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Wang Z, Zhu J, Gu H, Yuan Y, Zhang B, Zhu D, Zhou J, Zhu Y and Chen W: The clinical significance of abnormal Tim-3 expression on NK cells from patients with gastric cancer. Immunol Invest. 44:578–589. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Cheng G, Li M, Wu J, Ji M, Fang C, Shi H, Zhu D, Chen L, Zhao J, Shi L, et al: Expression of Tim-3 in gastric cancer tissue and its relationship with prognosis. Int J Clin Exp Pathol. 8:9452–9457. 2015.PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 40 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Zhao E, Zhang Z, Zhao G and Cao H: Association between Tim‑3 and Gal‑9 expression and gastric cancer prognosis. Oncol Rep 40: 2115-2126, 2018
APA
Wang, Y., Zhao, E., Zhang, Z., Zhao, G., & Cao, H. (2018). Association between Tim‑3 and Gal‑9 expression and gastric cancer prognosis. Oncology Reports, 40, 2115-2126. https://doi.org/10.3892/or.2018.6627
MLA
Wang, Y., Zhao, E., Zhang, Z., Zhao, G., Cao, H."Association between Tim‑3 and Gal‑9 expression and gastric cancer prognosis". Oncology Reports 40.4 (2018): 2115-2126.
Chicago
Wang, Y., Zhao, E., Zhang, Z., Zhao, G., Cao, H."Association between Tim‑3 and Gal‑9 expression and gastric cancer prognosis". Oncology Reports 40, no. 4 (2018): 2115-2126. https://doi.org/10.3892/or.2018.6627