Skip to main content

Prognostic significance of natural killer cell-associated markers in gastric cancer: quantitative analysis using multiplex immunohistochemistry

Abstract

Background

Natural killer (NK) cells mediate the anti-tumoral immune response as an important component of innate immunity. The aim of this study was to investigate the prognostic significance and functional implication of NK cell-associated surface receptors in gastric cancer (GC) by using multiplex immunohistochemistry (mIHC).

Methods

We performed an mIHC on tissue microarray slides, including 55 GC tissue samples. A total of 11 antibodies including CD57, NKG2A, CD16, HLA-E, CD3, CD20, CD45, CD68, CK, SMA, and ki-67 were used. CD45 + CD3-CD57 + cells were considered as CD57 + NK cells.

Results

Among CD45 + immune cells, the proportion of CD57 + NK cell was the lowest (3.8%), whereas that of CD57 + and CD57- T cells (65.5%) was the highest, followed by macrophages (25.4%), and B cells (5.3%). CD57 + NK cells constituted 20% of CD45 + CD57 + immune cells while the remaining 80% were CD57 + T cells. The expression of HLA-E in tumor cells correlated with that in tumoral T cells, B cells, and macrophages, but not CD57 + NK cells. The higher density of tumoral CD57 + NK cells and tumoral CD57 + NKG2A + NK cells was associated with inferior survival.

Conclusions

Although the number of CD57 + NK cells was lower than that of other immune cells, CD57 + NK cells and CD57 + NKG2A + NK cells were significantly associated with poor outcomes, suggesting that NK cell subsets play a critical role in GC progression. NK cells and their inhibitory receptor, NKG2A, may be potential targets in GC.

Background

Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide [1]. The incidence rate varies geographically, which is high in East Asia (including Korea and China), Eastern Europe and South America and relatively lower in North America [2]. Although the survival rates of GC patients have increased with proper screening systems, standardized surgical protocols, and development of chemotherapy regimens, the general outcome of GC patients, especially those in advanced stages, remains poor. Therefore, numerous efforts have been made to find more effective treatments in GC.

The importance of the tumor immune microenvironment (TME) has been well-established in the last two decades [3]. Studies show that cancer cells undergo a series of sequential phases called the “elimination phase”, “equilibrium phase”, and “immune escape phase” [4]. In each phase, various immune cell populations and stromal cells in the TME and tumor cells play an important role by interacting with each other, leading to tumor suppression or progression [4]. Characterizing the interactions between immune cells and tumor cells mediated by diverse inhibiting or activating cell surface receptors and their ligands, especially in regard to their spatial relationship, is critical to understand the TME.

A recently developed multiplex immunohistochemistry (mIHC) technique, enables the comprehensive visualization of various cell populations with their spatial information, by analyzing high-throughput image data with computer software program, and is therefore a suitable tool for investigating the TME [5]. In contrast to mass cytometry (CyTOF) [6], and single-cell RNA sequencing [7], which are also reliable methods to elucidate the TME, mIHC is more practical and convenient to implement as it employs formalin fixed paraffin embedded block.

Among the various immune cells in the TME, cytotoxic T lymphocytes (CTLs) are one of the most important components which directly eliminate tumor cells [8]. Many researchers have demonstrated that a higher number of CD8 + CTLs is associated with better prognosis in various cancers, including colorectal cancer [9], and gastric cancer [10]. It is also well-established that upregulation of two critical inhibitory receptors of CTL, namely PD-1 and CTLA-4, leads to CTL exhaustion and thereby tumor progression [11]. Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis or CTLA-4 can restore the anti-tumoral function of CTLs and impede tumor progression [12]. These drugs have been a breakthrough in many cancers, and the Food and Drug Administration (FDA) approved pembrolizumab (a PD-1 inhibitor) in locally advanced or metastatic gastric cancer with progression after 2 or more prior systemic chemotherapy [13]. Despite the safety and durable responses of these immunotherapeutic agents, only a subset of patients benefit from therapy [14], encouraging many researchers to explore new targets or alternative drug regimens by combining ICIs and other therapeutic agents.

Macrophages are also known as key modulators of tumor microenvironment. Macrophages can be differentiated into two extreme phenotype called M1 and M2 in response to the different stimuli [15]. M1 macrophages phagocytose and lyse tumor cells, and promote other immune cells by presenting tumor antigens [16], while M2 macrophages suppress anti-tumor immune response and contribute to tumor progression by promoting angiogenesis [17, 18]. The prognostic significance of macrophages in GC varies among studies which implemented conventional IHC method to detect surface expression of CD68 [19,20,21,22]. Compared to T cells and macropahges, the data regarding prognostic role of B cells in cancer are limited to date. In GC, some researchers demonstrated positive prognostic significance [23, 24] while others reported no association with prognosis [19].

Natural killer (NK) cells, that eliminate infected cells or tumor cells as a component of innate immunity [25], along with their various cell surface ligands, have recently emerged as a potential therapeutic target in solid tumors in the way of exploring novel targets in immunotherapy. Among different NK cell-associated surface markers, CD57 is expressed in mature NK cells that are less proliferative but more actively cytotoxic [26]. Besides these surface markers, NK cells also express various activating or inhibitory receptors, such as CD16 and NKG2A. CD16 is one of the most potent activating receptors of NK cells, which recognize tumor antigens and kill the tumor cells [27]. In contrast, NKG2A is an inhibitory receptor that recognizes HLA-E molecules on the tumor cells and lead to immune tolerance [28]. The prognostic significance of NK cells has been robustly studied by many researchers. Although higher NK cell density has been described as an indicator of good prognosis in multiple cancers, including colorectal cancer [29], and GC [30], contradictory results have been reported in breast cancer [31], and glioblastoma [32]. This discrepancy might result from the use of different antibodies such as CD56, CD57, or NKp46, which are used to detect the NK cell population. Moreover, all previous studies used conventional IHC methods, which made it difficult to analyze the subpopulation of NK cells expression specific receptors.

In the present study, we aimed to evaluate the prognostic significance of various immune cells or subset of immune cells with special regard to the NK cell-associated surface markers and receptors. We investigated the proportion of CD57 + NK cells (CD45 + CD3- CD20- CD57 + CD16 ± /NKG2A ±), T cells (CD45 + CD3 + CD20- CD57 ±), B cells (CD45 + CD3- CD20 +), and macrophages (CD45 + CD3- CD20- CD57- CD68 +) in GC by comprehensive visualization and analysis using mIHC.

Methods

Case selection

A total of 55 cases of consecutive stage II-III GC surgically resected between 2006 and 2008 at Seoul National University Bundang Hospital (SNUBH) were recruited for the study. The FFPE tissue samples were reviewed, and representative tissue areas were dissected for the construct a tissue microarray (TMA). Clinicopathological information was retrieved from electronic medical records.

Reagents and resources

All reagents, staining devices and software for the image analyses used in this study are listed in Additional file 1: Table S1. The antibodies used in this study and the staining order are also listed in Additional file 2: Table S2.

mIHC and computer-assisted image analyses

All implementations and analysis of mIHC were performed on the SuperBioChips (SuperBioChips Laboratories) as previously described [5]. After obtaining the digitally scanned images, the results of mIHC were analyzed, using publicly available software list in Additional file 2: Table S2. To quantify the amount of tumor-infiltrating immune cells, the cut-off values for each antibody were designated by either manual inspection of images or by analyzing the distribution of staining intensity of cells. Immune cells in the CK-positive tumor cell area were considered tumoral immune cells, whereas immune cells that are not in the CK-positive tumor cell area were considered stromal immune cells. Cell type of immune cells was determined based on lineage-specific antibody expression (Table 1). After quantification, the numbers of each cell type from different GC cases were compared and correlations with clinicopathological characteristics were investigated.

Table 1 Determination of immune cell types

Conventional IHC and EBV in situ hybridization (ISH)

For molecular classification of 55 GC samples, conventional immunohistochemistry (IHC) for E-cadherin (clone 36, mouse monoclonal; BD Biosciences, San Jose, CA, USA) and p53 (DO7, mouse monoclonal; Dako;Agilent Technologies, Santa Clara, CA) was performed on 3-μm-thick TMA slides using an automated immunostainer (BenchMark XT; Ventana Medical Systems) according to the manufacturer’s protocol. EBV ISH was performed using the INFORM EBV-encoded RNA probe (Ventana Medical Systems). The results of conventional IHC and EBV ISH were interpreted by two pathologists (J.K. and H.S.L.) in blinded manner. Discrepant cases were discussed to reach a consensus. For E-cadherin, strong membranous staining in tumor cells was defined as positive expression, whereas complete loss of membranous staining or aberrant cytoplasmic staining was interpreted as altered expression. For p53, strong nuclear staining in more than 10% of the tumor cells was defined as p53 positive. Cases were considered p53 negative when less than 10% of tumor cells were positive, and samples showing weak, variably scattered, or patchy positive tumor cells were determined as negative [33].

Microsatellite instability (MSI) test

The MSI test was performed according to the revised Bethesda guidelines [34]. Polymerase chain reaction (PCR) amplification of the extracted DNA from tumor and normal cells was performed and the PCR products were analyzed using a DNA autosequencer (ABI 3731 Genetic Analyzer, Applied Biosystems, Foster City, CA). Allele profiles of five markers (BAT-26, BAT-25, D5S346, D17S250, and S2S123) in tumor cells were compared to those of matched normal cells. Tumors with additional alleles in two or more markers were classified as MSI-high (MSI-H), tumors with novel bands in one marker were defined as MSI-low (MSI-L), and those with identical bands in all five markers were classified as microsatellite stable (MSS).

Molecular classification of GCs

We classified 55 GCs into 5 subtypes according to conventional IHC, EBV ISH, and MSI status as previously described [35]. A total of 4, 7, 21, 4, and 19 cases were classified as EBV + , MSI-H, EBV-MSS epithelial-mesenchymal transition (EMT)-like, EBV- MSS non-EMT-like p53 + , and EBV- MSS non-EMT-like p53- type, respectively.

Statistical analyses

All analyses were performed using R 4.0.3, RStudio 1.3.1093, and SPSS 25.0 (SPSS, Inc., Chicago, IL, USA). For mIHC, the t-test was used to identify statistical significance between immune cells, molecular classification, and clinical information. Spearman's rank correlation was used to analyze the correlation between immune cell density and human leukocyte antigen (HLA)-E expression in tumor cells. Linear regression analysis was performed using the ‘lm() function of the “stats” package. For univariate survival analysis of immune cell context, the ‘coxph()’ function of the package “survival” (Terry M Therneau (2021), R package version 3.2–10; https://cran.r-project.org/web/packages/survival/index.html) was used. The ‘surv_cutpoint()’ function of the package “survminer” (Alboukadel Kassambara (2021), R package version 0.4.9; https://cran.r-project.org/web/packages/survminer/index.html) was used to identify the ideal cut-off for determining the high and low levels for each immune cells [36]. A probability value of less than 0.05 was considered statistically significant.

Results

Patient characteristics

The clinicopathological characteristics are summarized in Table 2. There were 31 (56.4%) male and 24 (43.6%) female patients with a median age of 55 years (range, 30–70 years). Histologic diagnoses showed tubular adenocarcinoma in 50 (90.9%), mucinous adenocarcinoma in 4 (7.3%), and poorly cohesive carcinoma (signet ring cell carcinoma) in 1 (1.8%) of the cases. A total of 14 (25.5%) cases were stage II and 41 (74.5%) cases were stage III. Median progression-free survival (PFS) and overall survival (OS) were 65.4 months (range, 4.0–102.9 months) and 85.4 months (range, 13.3–107.9 months), respectively.

Table 2 Clinicopathological characteristics of 55 patients

Immune cell composition in gastric carcinoma focusing on NK cells and NK cell receptors

We performed chromogenic mIHC in 55 stage II-III GCs to investigate the immune context focused on NK cell-associated surface markers in GC. The densities of CD57 + T cells (yellow), CD57- T cells (green) and CD57 + NK cells (red) were determined in each case (Fig. 1A and Additional file 3: Figure S1). Among CD45 + immune cells, the proportion of CD57 + NK cells was the lowest (3.8%), while the proportion of T cells (65.5%) was the highest, followed by macrophages (25.4%) and B cells (5.3%) (Fig. 1B). The median densities of T cells, B cells, CD57 + NK cells and macrophages were 1281.1/mm2, 38.7/mm2, 44.6/mm2, and 469.1/mm2, respectively (Additional file 4: Table S3). The proportions of T cells, B cells, macrophages, and CD57 + NK cells varied in each case (Fig. 1C). Among CD45 + CD57 + immune cells, 80.0 ± 2.1% also expressed CD3, leaving only 20.0 ± 2.1% CD57 + NK cells (Fig. 1D). The proportion of CD57 + T cells and CD57 + NK cells among CD45 + CD57 + immune cells also varied in each case (Fig. 1E).

Fig. 1
figure 1

Immune cell composition in gastric cancer with an emphasis on NK cell-associated markers. CD57- T cells, CD57 + T cells, CD57 + NK cell, non-T/NK immune cells are presented as green, yellow, red and blue color (A). Tumor and stromal area were marked dark green and black color, respectively. The proportion of T cells, B cells, macrophages, and CD57 + NK cells is as depicted (B). The proportion of each immune cells in each case is illustrated (C). Among CD45 + CD57 + immune cells, CD57 + T cells and CD57 + NK cells constituted 80% and 20%, respectively (D). The proportion of CD57 + T cells and CD57 + NK cell among CD45 + CD57 + immune cells in each case is illustrated (E). A total of 55.1% and 27.9% of T cells showed CD16 and NKG2A expression (F). A total of 59.3% and 14.8% of CD57 + NK cells expressed CD16 and NKG2A (G)

Regarding the NK cell-associated activating and inhibitory receptors CD16 and NKG2A, 55.1 ± 2.34% of T cells were expressing CD16, while 27.9 ± 3.5% showed NKG2A expression (Fig. 1F). Similarly, 59.3 ± 2.9% of CD57 + NK cells were positive for CD16, and 14.8 ± 2.6% were positive for NKG2A (Fig. 1G). Of note, CD57 + NK cells constituted 2.4 ± 0.3% and 3.8 ± 0.8% of CD16 and NKG2A expressing immune cells (Additional file 5: Figure S2A and S2B). T cells constituted majority of CD16 and NKG2A expressing immune cells, followed by macrophages, CD57 + NK cells and B cells (Additional file 5: Figure S2A and S2B). The detailed numbers of immune cells are listed in Additional file 4: Table S3.

Correlation with molecular classification and other clinicopathological characteristics

When compared with EBV- GCs, EBV + GCs harbored a significantly higher number of T cells (p = 0.012) (Fig. 2A). There was no significant difference in the number of other immune cells between the two groups (Additional file 6: Figure S3A – S3C). The number of T cells, CD57 + NK cells and macrophages was higher in MSI-H GCs than in MSS/MSI-L GCs although the differences were not statistically significant (all p > 0.05) (Additional file 6: Figure S3D – S3F). When immune cells were segregated based on location, the density of stromal CD57 + immune cells and CD57 + T cells was significantly lower in EBV- MSS EMT-like GCs than in other 4 subtypes (Fig. 2B and 2C). And the number of tumoral T cells and tumoral CD57 + T cells was higher in EBV- MSS non-EMT-like p53- GCs than in EBV- MSS non-EMT-like p53 + counterpart (Fig. 2D and 2E).

Fig. 2
figure 2

Correlation with molecular classification and other clinicopathological characteristics. T cell density was higher in EBV + gastric cancer than in EBV- gastric cancer (A). CD57 + immune cell (B) and CD57 + T cell (C) density was lower in EBV- MSS/EMT-like gastric cancer than in other molecular subtypes. T cell density (D) and CD57 + T cell density (E) was higher in EMV- MSS/non-EMT-like p53- subtype than in EBV- MSS/non-EMT-like p53 + subtype. CD45 + CD57 + immune cell (F), CD57 + NK cell (G), and CD57 + NKG2A + NK cell (H) density did not differ between stage 3 and stage 2 gastric cancer

The density of CD57 + immune cells, CD57 + NK cells and CD57 + NKG2A + NK cells did not correlate with other clinicopathological characteristics, including stages (Fig. 2F–H), lymphovascular invasion and degree of tumor differentiation.

Correlation among immune cell densities and HLA-E expression in tumor cells

The density of T cells correlated with that of B cells, and macrophages (all p < 0.05). Notably, the density of CD57 + NK cells showed correlation with the density of CD57 + lymphocytes and CD57 + T cells (all p < 0.05) (Fig. 3A) and not with T cells, B cells, and macrophages (all p > 0.05).

Fig. 3
figure 3

A correlation between immune cells are illustrated in heatmap (*: P < 0.05 by Spearman's Rank-Order Correlation) (A). HLA-E + tumor cells correlated with T cells (B), B cells (C), macrophages (D), and CD57 + NK cells (E)

We also analyzed whether HLA-E expression in tumor cells displayed any correlation with immune cell density. The median number of tumor cells with HLA-E expression per mm2 was 1085.3 (range 10.7 – 4602.1). The expression of HLA-E correlated with the densities of intratumoral T cells, B cells, and macrophages (all p < 0.05), whereas it did not correlate with CD57 + NK cell density or CD57 + NKG2A + NK cell density (all p > 0.05) (Fig. 3B–E).

Survival analysis

Univariate Cox regression analysis was performed to find out which immune cells or subset of immune cells are associated with prognosis. For each immune cells, the significance of the density of tumoral and stromal immune cells was investigated. In addition, the significance of the density of total (“tumoral and stromal”) immune cells was also analyzed. Higher density of “tumoral and stromal” and stromal T cells and macrophages was associated with longer OS (all p < 0.05) (Fig. 4A). High tumoral CD57 + NK cells, CD16 + CD57 + NK cells, and NGS2A + CD57 + NK cells was associated with shorter OS (all p < 0.05) (Fig. 4A). Likewise, high “tumoral and stromal”, tumoral, and stromal T cells and macrophages was associated with longer PFS (all p < 0.05) (Fig. 4B). Tumoral CD57 + T cells was associated with longer PFS (p < 0.05). And higher density of tumoral CD57 + NK cells, and NKG2A + CD57 + NK cells was associated with superior PFS (all p < 0.05) (Fig. 4B). High density of B cells showed a tendency of longer OS and PFS, but the differences were not statistically significant (Fig. 4A, B).

Fig. 4
figure 4

A univariate survival analysis representing hazard ratio of each immune cell type or a subset of immune cells. The proportion of each immune cell types or a subset of immune cells in all immune cells were represented with the size of circle. Tumoral CD57 + NK cells were associated with inferior overall survival (A) and progressive-free survival (B)

Discussion

In the current study, we uncovered the TME of GCs by using mIHC, with a special reference to NK cells and their surface receptors. Comprehensive analysis of TME revealed that high density of T cells and macrophages are associated with better prognosis, similar with previous studies. Although NK cells constitute a minor component of tumor-infiltrating immune cells, the density of tumoral CD57 + NK cells and CD57 + NKG2A + NK cells was significantly associated with inferior OS and PFS in our cohort. This suggests that CD57 + NK cells in GC are not actively cytotoxic, which might be related to the expression of NKG2A or possibly other inhibitory receptors.

ICIs targeting PD-1/PD-L1 [37] and CTLA-4 [38] has proved to be a breakthrough in cancer treatment in the last decade. However, only a minority of patients benefit from these agents, thereby necessitating patient selection and new target discovery [39]. NK cells and their surface receptors have been proposed as novel immunotherapeutic targets [40]. Although the prognostic significance of NK cells in various cancers has been studied, the results are discrepant and NK cell receptors CD16 or NKG2A have not been thoroughly investigated. Moreover, most previous studies used conventional IHC, which made detailed subgrouping of immune cells according to their receptor expression impossible [29,30,31,32, 41, 42]. Therefore, we aimed to thoroughly investigate the expression of activating or inhibitory receptors in NK cells and their prognostic significance in GC.

NK cells are key to innate immunity and can kill cancer cells as well as infected cells faster than T cells [27]. NK cells are characterized by the expression of CD56 and lack of CD3 expression and can be subdivided into less mature CD56bright and more mature CD56dim subsets [43]. CD57, which is induced in the CD56dimCD16 + subset, is a marker of terminal differentiation [44]. CD57 + NK cells are less proliferative but more actively cytotoxic. Among various surface receptors, CD16 is one of the most potent activating receptors of NK cells, which recognize tumor antigens via immunoglobulin G and lead to antibody-dependent cell-mediated cytotoxicity [27]. In contrast, NKG2A is an inhibitory receptor that recognizes HLA-E molecules and prevents NK cell activation [28].

By using mIHC, including anti-CD57, anti-CD16, anti-NKG2A antibodies, and other immune cell markers, we observed that CD57 + NK cells constituted only 3.8% of all immune cells in GC. Of interest, 80% of CD45 + CD57 + immune cells were T cells, leaving only 20% as CD57 + NK cells. In addition, CD57 + NK cells are a minor component of CD16 + or NKG2A + immune cells. Our results confirmed that the number of CD57 + NK cells is far lower than that of T cells in GC [45]. It is also well known that CD57 [46] and NKG2A [47] are expressed CD8 + T cells as well as NK cells. Chen et al. showed that CD8 + T cells predominate NKG2A + lymphocytes in lung cancer [48].

In univariate Cox regression analysis, a higher density of T cells and macrophages was associated with longer OS and PFS. These results are similar to previous analyses, which demonstrated a high number of T cells as an indicator of favorable prognosis in GC patients [49,50,51]. Interestingly, “tumoral and stromal”, and “stromal” CD57 + T cells were associated with inferior OS, although it was not statistically significant, while tumoral CD57 + T cells were associated with longer PFS. Although the majority of CD57 + T cells are CD8 + cytotoxic T cells, the cytotoxic activity of CD8 + CD57 + T cells depend on the expression of CD27 and CD28. CD8 + CD57 + T cells in peripheral blood which do not express CD27 and CD28 have enhanced cytotoxic potential, while CD27 and CD28 expressing CD8 + CD57 + T cells in tumor have impaired cytotoxic activity [52]. Although we did not investigate the expression of CD27 and CD28 in tumoral or stromal CD57 + T cells, differential expression of CD27 and CD28 in tumoral and stromal CD57 + T cells might be one possible explanation for our results. Regarding macrophages, the prognostic significance varies greatly among studies in which some researchers reported them as good prognostic factor [19, 20] while others demonstrated contradicting results [21, 22] by using single anti-CD68 antibody. When macrophages were subdivided into M1 and M2 subtypes, M1 macrophages were generally associated with good prognosis [53, 54] whereas M2 subtypes were enriched in cases showing poor outcomes [55,56,57,58]. In the present study, subtyping of macrophages is limited since we did not perform specific antibodies to detect macrophage polarization. Nevertheless, our study confirmed that macrophages are an important component of the TME, constituting 25.4% of all GC-associated immune cells and affect clinical outcomes.

The density of B cells was generally associated with better survival although statistical significance was not reached in the present study. Similar with the current study, some researchers also demonstrated that B cells are associated with favorable prognosis in GC [23, 24], although others reported no specific association between B cells and prognosis [19]. B cells not only produce antibodies but also act as antigen presenting cells when properly activated [59]. These activated B cells can contribute to anti-tumor immune response by inducing both CD4 + and CD8 + T cells response [60, 61]. However, the prognostic role of B cells in GC is controversial to date and requires further validation.

Although the proportion of CD57 + NK cells was relatively low, higher tumoral CD57 + NK cells and tumoral CD57 + NKG2A + NK cell density was significantly associated with shorter OS and PFS. The prognostic implication of NK cells remains controversial across various types of cancer; some researchers have reported they were associated with good prognosis in lung cancer [62], and colorectal cancer [29], but others have demonstrated the opposite results in breast cancer [31], and glioblastoma [32]. In GC, a high density of NK cells was generally associated with improved clinical outcomes [30, 41, 63, 64]. A few studies have investigated the expression of tumor cell receptors which interact with NK cell surface receptors. Mimura et al. [42] reported that elevated expression of tumor cell MHC Class I chain-related A (MICA), MHC Class I chain-related B (MICB), and several UL-16-binding proteins (ULBPs), which are receptors for the NK cell-activating receptor, NKG2D, was associated with improved survival outcomes. On the other hand, Ishigami et al. [65] demonstrated that high expression of tumor cell HLA-E, which interacts with NKG2A, was associated with inferior outcomes. Our study revealed that not only CD57 + NKG2A + NK cells but also CD57 + NK cells in total were indicative of poor outcomes. Previous studies used single conventional IHC such as CD57, which renders it impossible to differentiate between CD57 + NK cells and CD57 + T cells may explain the discrepancy between the current study and the previous studies. As mentioned above, majority of CD57 + lymphocytes are CD8 + T cells, which might have affected the prognostic impact of CD57 marker, because higher CD8 + T cell infiltration is usually associated with better prognosis. By applying mIHC, we demonstrated that infiltration of CD57 + NK cells is, in fact, associated with inferior outcomes. Our results suggest that the cytotoxic function of CD57 + NK cells might be impaired in GC. Impaired NK cell function could be due to the expression of inhibitory receptors other than NKG2A, such as, killer cell immunoglobulin-like receptors (KIR), lymphocyte activation gene-3 (LAG-3), PD-1, or CTLA-4, which we did not include in the present study. In addition, researchers have demonstrated that transforming growth factor-beta (TGF-β) [66] or prostaglandin E2 (PGE2) [45] secreted by tumor-associated macrophages and tumor cells can inhibit NK cell cytotoxicity and proliferation in GC. Therefore, therapies that can restore the cytotoxic activity of NK cell, including the recently developed monoclonal NKG2A antibody, KIR antagonist, and inhibiting TGF-β signaling are expected to improve survival in GC patients.

Regarding molecular classification, EBV + GCs showed a higher density of T cells, which is in line with previous studies [35, 67]. The higher number of T cells in EBV + GCs is attributed to the relatively favorable clinical course in patients with EBV + GCs [68, 69]. Although the density of stromal and intratumoral CD57 + T cells differed according to the molecular subtypes, interpreting these results in this study is limited due to small sized sample. The significance of these differences with reference to clinical outcomes should be further validated in a large cohort.

Lastly, we demonstrated that HLA-E expression correlated with intratumoral T cells, B cells, and macrophages, but not CD57 + NK cells. Upon ligation with peptide-loaded HLA-E molecules on tumor cells, NKG2A induces the downregulation of NK cell function, thereby leading to immune escape. Ironically, HLA-E expression on tumor cells is induced by interferon-gamma (IFN-γ), which is produced by active immune cells [70]. A positive correlation between HLA-E expression in tumor cells and T cells [71] and NKG2A + immune cells [72] was reported, as well as a negative correlation with CD57 + immune cells [73]. Although our study failed to identify a direct correlation between HLA-E expression with CD57 + NK cells or NKG2A + cells, correlation with other immune cells suggests that HLA-E could be induced by active anti-tumor immune response possibly to bypass NKG2A + T and CD57 + NK cells.

Conclusions

Our study has a limitation because this was a retrospective study and was composed of small sized cohort. However, we observed that intratumoral CD57 + NK cells and CD57 + NKG2A + NK cells were associated with poor outcomes. This suggests that NK cells play an important role in GC progression, although their density was the lowest among other immune cells. Therefore, targeting NK cell inhibitory receptors, such as NKG2A, may lead to successful GC treatment strategies.

Availability of data and materials

The authors declare that the data supporting the fndings of this study are available within the paper (and its supplementary information files). The additional datasets generated and/or analyzed during the current study are available from the corresponding authors upon reasonable request.

Abbreviations

CTL:

Cytotoxic T lymphocyte

CyTOF:

Mass cytometry

EMT:

Epithelial-mesenchymal transition

FDA:

Food and drug administration

GC:

Gastric cancer

ICI:

Immune checkpoint inhibitor

IFN-γ:

Interferone-gamma

IHC:

Immunohistochemistry

ISH:

In situ hybridization

KIR:

Killer cell immunoglobulin-like receptors

LAG-3:

Lymphocyte activation gene-3

MICA:

MHC class I chain-related A

MICB:

MHC class I chain-related B

mIHC:

Multiplex immunohistochemistry

MSI:

Microsatellite instability

MSI-H:

MSI-high

MSI-L:

MSI-low

MSS:

Microsatellite stable

NK:

Natural killer

OS:

Overall survival

PCR:

Polymerase chain reaction

PFS:

Progression-free survival

PGE2:

Prostaglandin E2

TGF-β:

Transforming growth factor-beta

TMA:

Tissue microarray

TME:

Tumor immune microenvironment

ULBP:

UL-16-binding protein

References

  1. Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, Dandona L, Fleming T, Forouzanfar MH, Hancock J, Hay RJ, Hunter-Merrill R, Huynh C, Hosgood HD, Johnson CO, Jonas JB, Khubchandani J, Kumar GA, Kutz M, Lan Q, Larson HJ, Liang X, Lim SS, Lopez AD, MacIntyre MF, Marczak L, Marquez N, Mokdad AH, Pinho C, Pourmalek F, Salomon JA, Sanabria JR, Sandar L, Sartorius B, Schwartz SM, Shackelford KA, Shibuya K, Stanaway J, Steiner C, Sun J, Takahashi K, Vollset SE, Vos T, Wagner JA, Wang H, Westerman R, Zeeb H, Zoeckler L, Abd-Allah F, Ahmed MB, Alabed S, Alam NK, Aldhahri SF, Alem G, Alemayohu MA, Ali R, Al-Raddadi R, Amare A, Amoako Y, Artaman A, Asayesh H, Atnafu N, Awasthi A, Saleem HB, Barac A, Bedi N, Bensenor I, Berhane A, Bernabe E, Betsu B, Binagwaho A, Boneya D, Campos-Nonato I, Castaneda-Orjuela C, Catala-Lopez F, Chiang P, Chibueze C, Chitheer A, Choi JY, Cowie B, Damtew S, das Neves J, Dey S, Dharmaratne S, Dhillon P, Ding E, Driscoll T, Ekwueme D, Endries AY, Farvid M, Farzadfar F, Fernandes J, Fischer F, TT GH, Gebru A, Gopalani S, Hailu A, Horino M, Horita N, Husseini A, Huybrechts I, Inoue M, Islami F, Jakovljevic M, James S, Javanbakht M, Jee SH, Kasaeian A, Kedir MS, Khader YS, Khang YH, Kim D, Leigh J, Linn S, Lunevicius R, El Razek HMA, Malekzadeh R, Malta DC, Marcenes W, Markos D, Melaku YA, Meles KG, Mendoza W, Mengiste DT, Meretoja TJ, Miller TR, Mohammad KA, Mohammadi A, Mohammed S, Moradi-Lakeh M, Nagel G, Nand D, Le Nguyen Q, Nolte S, Ogbo FA, Oladimeji KE, Oren E, Pa M, Park EK, Pereira DM, Plass D, Qorbani M, Radfar A, Rafay A, Rahman M, Rana SM, Soreide K, Satpathy M, Sawhney M, Sepanlou SG, Shaikh MA, She J, Shiue I, Shore HR, Shrime MG, So S, Soneji S, Stathopoulou V, Stroumpoulis K, Sufiyan MB, Sykes BL, Tabares-Seisdedos R, Tadese F, Tedla BA, Tessema GA, Thakur JS, Tran BX, Ukwaja KN, Uzochukwu BSC, Vlassov VV, Weiderpass E, Wubshet Terefe M, Yebyo HG, Yimam HH, Yonemoto N, Younis MZ, Yu C, Zaidi Z, Zaki MES, Zenebe ZM, Murray CJL, Naghavi M, Global Burden of Disease Cancer C. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017;3(4):524–48.

  2. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.

    Article  CAS  PubMed  Google Scholar 

  3. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137–48.

    Article  CAS  PubMed  Google Scholar 

  5. Koh J, Kwak Y, Kim J, Kim WH. High-throughput multiplex immunohistochemical imaging of the tumor and its microenvironment. Cancer Res Treat. 2020;52(1):98–108.

    Article  CAS  PubMed  Google Scholar 

  6. Giesen C, Wang HA, Schapiro D, Zivanovic N, Jacobs A, Hattendorf B, Schuffler PJ, Grolimund D, Buhmann JM, Brandt S, Varga Z, Wild PJ, Gunther D, Bodenmiller B. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat Methods. 2014;11(4):417–22.

    Article  CAS  PubMed  Google Scholar 

  7. Chen H, Ye F, Guo G. Revolutionizing immunology with single-cell RNA sequencing. Cell Mol Immunol. 2019;16(3):242–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Farhood B, Najafi M, Mortezaee K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: a review. J Cell Physiol. 2019;234(6):8509–21.

    Article  CAS  PubMed  Google Scholar 

  9. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pages C, Tosolini M, Camus M, Berger A, Wind P, Zinzindohoue F, Bruneval P, Cugnenc PH, Trajanoski Z, Fridman WH, Pages F. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313(5795):1960–4.

    Article  CAS  PubMed  Google Scholar 

  10. Koh J, Ock CY, Kim JW, Nam SK, Kwak Y, Yun S, Ahn SH, Park DJ, Kim HH, Kim WH, Lee HS. Clinicopathologic implications of immune classification by PD-L1 expression and CD8-positive tumor-infiltrating lymphocytes in stage II and III gastric cancer patients. Oncotarget. 2017;8(16):26356–67.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and ANTI-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 2018;8:86.

    Article  PubMed  PubMed Central  Google Scholar 

  13. FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication: U.S. Food and Drug Administration; 2017 [updated 2017 May 30]. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-first-tissuesite-agnostic-indication.

  14. Fashoyin-Aje L, Donoghue M, Chen H, He K, Veeraraghavan J, Goldberg KB, Keegan P, McKee AE, Pazdur R. FDA approval summary: pembrolizumab for recurrent locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma expressing PD-L1. Oncologist. 2019;24(1):103–9.

    Article  CAS  PubMed  Google Scholar 

  15. Enderlin Vaz da Silva Z, Lehr HA, Velin D. In vitro and in vivo repair activities of undifferentiated and classically and alternatively activated macrophages. Pathobiology. 2014;81(2):86–93.

    Article  PubMed  Google Scholar 

  16. Su Z, Zhang P, Yu Y, Lu H, Liu Y, Ni P, Su X, Wang D, Liu Y, Wang J, Shen H, Xu W, Xu H. HMGB1 facilitated macrophage reprogramming towards a proinflammatory M1-like phenotype in experimental autoimmune myocarditis development. Sci Rep. 2016;6:21884.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhou D, Yang K, Chen L, Zhang W, Xu Z, Zuo J, Jiang H, Luan J. Promising landscape for regulating macrophage polarization: epigenetic viewpoint. Oncotarget. 2017;8(34):57693–706.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Spiller KL, Anfang RR, Spiller KJ, Ng J, Nakazawa KR, Daulton JW, Vunjak-Novakovic G. The role of macrophage phenotype in vascularization of tissue engineering scaffolds. Biomaterials. 2014;35(15):4477–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Haas M, Dimmler A, Hohenberger W, Grabenbauer GG, Niedobitek G, Distel LV. Stromal regulatory T-cells are associated with a favourable prognosis in gastric cancer of the cardia. BMC Gastroenterol. 2009;9:65.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Ohno S, Inagawa H, Dhar DK, Fujii T, Ueda S, Tachibana M, Suzuki N, Inoue M, Soma G, Nagasue N. The degree of macrophage infiltration into the cancer cell nest is a significant predictor of survival in gastric cancer patients. Anticancer Res. 2003;23(6D):5015–22.

    PubMed  Google Scholar 

  21. Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Okumura H, Matsumoto M, Miyazono F, Hokita S, Aikou T. Tumor-associated macrophage (TAM) infiltration in gastric cancer. Anticancer Res. 2003;23(5A):4079–83.

    CAS  PubMed  Google Scholar 

  22. Osinsky S, Bubnovskaya L, Ganusevich I, Kovelskaya A, Gumenyuk L, Olijnichenko G, Merentsev S. Hypoxia, tumour-associated macrophages, microvessel density, VEGF and matrix metalloproteinases in human gastric cancer: interaction and impact on survival. Clin Transl Oncol. 2011;13(2):133–8.

    Article  CAS  PubMed  Google Scholar 

  23. Sakimura C, Tanaka H, Okuno T, Hiramatsu S, Muguruma K, Hirakawa K, Wanibuchi H, Ohira M. B cells in tertiary lymphoid structures are associated with favorable prognosis in gastric cancer. J Surg Res. 2017;215:74–82.

    Article  CAS  PubMed  Google Scholar 

  24. Hennequin A, Derangere V, Boidot R, Apetoh L, Vincent J, Orry D, Fraisse J, Causeret S, Martin F, Arnould L, Beltjens F, Ghiringhelli F, Ladoire S. Tumor infiltration by Tbet+ effector T cells and CD20+ B cells is associated with survival in gastric cancer patients. Oncoimmunology. 2016;5(2):e1054598.

    Article  PubMed  Google Scholar 

  25. Lopez-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK cells. Cancer Cell. 2017;32(2):135–54.

    Article  CAS  PubMed  Google Scholar 

  26. Lanier LL, Le AM, Phillips JH, Warner NL, Babcock GF. Subpopulations of human natural killer cells defined by expression of the Leu-7 (HNK-1) and Leu-11 (NK-15) antigens. J Immunol. 1983;131(4):1789–96.

    Article  CAS  PubMed  Google Scholar 

  27. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. 2008;9(5):503–10.

    Article  CAS  PubMed  Google Scholar 

  28. Braud VM, Allan DS, O’Callaghan CA, Soderstrom K, D’Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, Lanier LL, McMichael AJ. HLA-E binds to natural killer cell receptors CD94/NKG2A B and C. Nature. 1998;391(6669):795–9.

    Article  CAS  PubMed  Google Scholar 

  29. Alderdice M, Dunne PD, Cole AJ, O’Reilly PG, McArt DG, Bingham V, Fuchs MA, McQuaid S, Loughrey MB, Murray GI, Samuel LM, Lawler M, Wilson RH, Salto-Tellez M, Coyle VM. Natural killer-like signature observed post therapy in locally advanced rectal cancer is a determinant of pathological response and improved survival. Mod Pathol. 2017;30(9):1287–98.

    Article  CAS  PubMed  Google Scholar 

  30. Ishigami S, Natsugoe S, Tokuda K, Nakajo A, Xiangming C, Iwashige H, Aridome K, Hokita S, Aikou T. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Lett. 2000;159(1):103–8.

    Article  CAS  PubMed  Google Scholar 

  31. Rathore AS, Goel MM, Makker A, Kumar S, Srivastava AN. Is the tumor infiltrating natural killer cell (NK-TILs) count in infiltrating ductal carcinoma of breast prognostically significant? Asian Pac J Cancer Prev. 2014;15(8):3757–61.

    Article  PubMed  Google Scholar 

  32. Wu S, Yang W, Zhang H, Ren Y, Fang Z, Yuan C, Yao Z. The prognostic landscape of tumor-infiltrating immune cells and immune checkpoints in glioblastoma. Technol Cancer Res Treat. 2019;18:1533033819869949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lee HS, Lee HK, Kim HS, Yang HK, Kim WH. Tumour suppressor gene expression correlates with gastric cancer prognosis. J Pathol. 2003;200(1):39–46.

    Article  CAS  PubMed  Google Scholar 

  34. Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, Fishel R, Lindor NM, Burgart LJ, Hamelin R, Hamilton SR, Hiatt RA, Jass J, Lindblom A, Lynch HT, Peltomaki P, Ramsey SD, Rodriguez-Bigas MA, Vasen HF, Hawk ET, Barrett JC, Freedman AN, Srivastava S. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96(4):261–8.

    Article  CAS  PubMed  Google Scholar 

  35. Koh J, Lee KW, Nam SK, Seo AN, Kim JW, Kim JW, Park DJ, Kim HH, Kim WH, Lee HS. Development and validation of an easy-to-implement, practical algorithm for the identification of molecular subtypes of gastric cancer: prognostic and therapeutic implications. Oncologist. 2019;24(12):e1321–30.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bester AC, Lee JD, Chavez A, Lee YR, Nachmani D, Vora S, Victor J, Sauvageau M, Monteleone E, Rinn JL, Provero P, Church GM, Clohessy JG, Pandolfi PP. An integrated genome-wide CRISPRa approach to functionalize lncRNAs in drug resistance. Cell 2018; 173(3): 649–64 e20.

  37. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, Carcereny E, Ahn MJ, Felip E, Lee JS, Hellmann MD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Emancipator K, Gandhi L, Investigators K-. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–28.

    Article  PubMed  Google Scholar 

  38. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim N, Kim HS. Targeting checkpoint receptors and molecules for therapeutic modulation of natural killer cells. Front Immunol. 2018;9:2041.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Kijima Y, Ishigami S, Hokita S, Koriyama C, Akiba S, Eizuru Y, Aikou T. The comparison of the prognosis between Epstein-Barr virus (EBV)-positive gastric carcinomas and EBV-negative ones. Cancer Lett. 2003;200(1):33–40.

    Article  CAS  PubMed  Google Scholar 

  42. Mimura K, Kamiya T, Shiraishi K, Kua LF, Shabbir A, So J, Yong WP, Suzuki Y, Yoshimoto Y, Nakano T, Fujii H, Campana D, Kono K. Therapeutic potential of highly cytotoxic natural killer cells for gastric cancer. Int J Cancer. 2014;135(6):1390–8.

    Article  CAS  PubMed  Google Scholar 

  43. Stabile H, Fionda C, Gismondi A, Santoni A. Role of distinct natural killer cell subsets in anticancer response. Front Immunol. 2017;8:293.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lopez-Verges S, Milush JM, Pandey S, York VA, Arakawa-Hoyt J, Pircher H, Norris PJ, Nixon DF, Lanier LL. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010;116(19):3865–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li T, Zhang Q, Jiang Y, Yu J, Hu Y, Mou T, Chen G, Li G. Gastric cancer cells inhibit natural killer cell proliferation and induce apoptosis via prostaglandin E2. Oncoimmunology. 2016;5(2):e1069936.

    Article  PubMed  Google Scholar 

  46. Focosi D, Bestagno M, Burrone O, Petrini M. CD57+ T lymphocytes and functional immune deficiency. J Leukoc Biol. 2010;87(1):107–16.

    Article  CAS  PubMed  Google Scholar 

  47. Cho JH, Kim HO, Webster K, Palendira M, Hahm B, Kim KS, King C, Tangye SG, Sprent J. Calcineurin-dependent negative regulation of CD94/NKG2A expression on naive CD8+ T cells. Blood. 2011;118(1):116–28.

    Article  CAS  PubMed  Google Scholar 

  48. Chen Y, Xin Z, Huang L, Zhao L, Wang S, Cheng J, Wu P, Chai Y. CD8(+) T cells form the predominant subset of NKG2A(+) cells in human lung cancer. Front Immunol. 2019;10:3002.

    Article  CAS  PubMed  Google Scholar 

  49. Arigami T, Uenosono Y, Ishigami S, Matsushita D, Hirahara T, Yanagita S, Okumura H, Uchikado Y, Nakajo A, Kijima Y, Natsugoe S. Decreased density of CD3+ tumor-infiltrating lymphocytes during gastric cancer progression. J Gastroenterol Hepatol. 2014;29(7):1435–41.

    Article  CAS  PubMed  Google Scholar 

  50. Kim KJ, Lee KS, Cho HJ, Kim YH, Yang HK, Kim WH, Kang GH. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Hum Pathol. 2014;45(2):285–93.

    Article  CAS  PubMed  Google Scholar 

  51. Chiaravalli AM, Feltri M, Bertolini V, Bagnoli E, Furlan D, Cerutti R, Novario R, Capella C. Intratumour T cells, their activation status and survival in gastric carcinomas characterised for microsatellite instability and Epstein-Barr virus infection. Virchows Arch. 2006;448(3):344–53.

    Article  PubMed  Google Scholar 

  52. Huang B, Liu R, Wang P, Yuan Z, Yang J, Xiong H, Zhang N, Huang Q, Fu X, Sun W, Li L. CD8(+)CD57(+) T cells exhibit distinct features in human non-small cell lung cancer. J Immunother Cancer. 2020;8(1):e000639.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Wang B, Xu D, Yu X, Ding T, Rao H, Zhan Y, Zheng L, Li L. Association of intra-tumoral infiltrating macrophages and regulatory T cells is an independent prognostic factor in gastric cancer after radical resection. Ann Surg Oncol. 2011;18(9):2585–93.

    Article  PubMed  Google Scholar 

  54. Zhang H, Wang X, Shen Z, Xu J, Qin J, Sun Y. Infiltration of diametrically polarized macrophages predicts overall survival of patients with gastric cancer after surgical resection. Gastric Cancer. 2015;18(4):740–50.

    Article  CAS  PubMed  Google Scholar 

  55. Kawahara A, Hattori S, Akiba J, Nakashima K, Taira T, Watari K, Hosoi F, Uba M, Basaki Y, Koufuji K, Shirouzu K, Akiyama S, Kuwano M, Kage M, Ono M. Infiltration of thymidine phosphorylase-positive macrophages is closely associated with tumor angiogenesis and survival in intestinal type gastric cancer. Oncol Rep. 2010;24(2):405–15.

    Article  CAS  PubMed  Google Scholar 

  56. Kim KJ, Wen XY, Yang HK, Kim WH, Kang GH. Prognostic implication of M2 macrophages are determined by the proportional balance of tumor associated macrophages and tumor infiltrating lymphocytes in microsatellite-unstable gastric carcinoma. PLoS ONE. 2015;10(12):e0144192.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Lin CN, Wang CJ, Chao YJ, Lai MD, Shan YS. The significance of the co-existence of osteopontin and tumor-associated macrophages in gastric cancer progression. BMC Cancer. 2015;15:128.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Park JY, Sung JY, Lee J, Park YK, Kim YW, Kim GY, Won KY, Lim SJ. Polarized CD163+ tumor-associated macrophages are associated with increased angiogenesis and CXCL12 expression in gastric cancer. Clin Res Hepatol Gastroenterol. 2016;40(3):357–65.

    Article  CAS  PubMed  Google Scholar 

  59. Van Belle K, Herman J, Boon L, Waer M, Sprangers B, Louat T. Comparative in vitro immune stimulation analysis of primary human B cells and B cell lines. J Immunol Res. 2016;2016:5281823.

    PubMed  PubMed Central  Google Scholar 

  60. Zheng J, Liu Y, Qin G, Chan PL, Mao H, Lam KT, Lewis DB, Lau YL, Tu W. Efficient induction and expansion of human alloantigen-specific CD8 regulatory T cells from naive precursors by CD40-activated B cells. J Immunol. 2009;183(6):3742–50.

    Article  CAS  PubMed  Google Scholar 

  61. von Bergwelt-Baildon M, Schultze JL, Maecker B, Menezes I, Nadler LM. Correspondence re R. Lapointe et al., CD40-stimulated B lymphocytes pulsed with tumor antigens are effective antigen-presenting cells that can generate specific T cells. Cancer Res 2003;63:2836–43. Cancer Res. 2004;64(11):4055–6; author reply 6–7.

  62. Platonova S, Cherfils-Vicini J, Damotte D, Crozet L, Vieillard V, Validire P, Andre P, Dieu-Nosjean MC, Alifano M, Regnard JF, Fridman WH, Sautes-Fridman C, Cremer I. Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. 2011;71(16):5412–22.

    Article  CAS  PubMed  Google Scholar 

  63. Amoueian S, Attaranzadeh A, Montazer M. Intratumoral CD68-, CD117-, CD56-, and CD1a-positive immune cells and the survival of Iranian patients with non-metastatic intestinal-type gastric carcinoma. Pathol Res Pract. 2015;211(4):326–31.

    Article  CAS  PubMed  Google Scholar 

  64. Svensson MC, Warfvinge CF, Fristedt R, Hedner C, Borg D, Eberhard J, Micke P, Nodin B, Leandersson K, Jirstrom K. The integrative clinical impact of tumor-infiltrating T lymphocytes and NK cells in relation to B lymphocyte and plasma cell density in esophageal and gastric adenocarcinoma. Oncotarget. 2017;8(42):72108–26.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Ishigami S, Arigami T, Okumura H, Uchikado Y, Kita Y, Kurahara H, Maemura K, Kijima Y, Ishihara Y, Sasaki K, Uenosono Y, Natsugoe S. Human leukocyte antigen (HLA)-E and HLA-F expression in gastric cancer. Anticancer Res. 2015;35(4):2279–85.

    CAS  PubMed  Google Scholar 

  66. Peng LS, Zhang JY, Teng YS, Zhao YL, Wang TT, Mao FY, Lv YP, Cheng P, Li WH, Chen N, Duan M, Chen W, Guo G, Zou QM, Zhuang Y. Tumor-associated monocytes/macrophages impair NK-cell function via TGFβ1 in human gastric cancer. Cancer Immunol Res. 2017;5(3):248–56.

    Article  CAS  PubMed  Google Scholar 

  67. Mansuri N, Birkman E-M, Heuser VD, Lintunen M, Ålgars A, Sundström J, Ristamäki R, Lehtinen L, Carpén O. Association of tumor-infiltrating T lymphocytes with intestinal-type gastric cancer molecular subtypes and outcome. Virchows Arch. 2020;4:707–17.

    Google Scholar 

  68. Song HJ, Srivastava A, Lee J, Kim YS, Kim KM, Ki Kang W, Kim M, Kim S, Park CK, Kim S. Host inflammatory response predicts survival of patients with Epstein-Barr virus-associated gastric carcinoma. Gastroenterology. 2010;139(1):84-92.e2.

    Article  PubMed  Google Scholar 

  69. Van Beek J, Zur Hausen A, Klein Kranenbarg E, van de Velde CJ, Middeldorp JM, van den Brule AJ, Meijer CJ, Bloemena E. EBV-positive gastric adenocarcinomas: a distinct clinicopathologic entity with a low frequency of lymph node involvement. J Clin Oncol. 2004;22(4):664–70.

    Article  PubMed  Google Scholar 

  70. Gustafson KS, Ginder GD. Interferon-gamma induction of the human leukocyte antigen-E gene is mediated through binding of a complex containing STAT1alpha to a distinct interferon-gamma-responsive element. J Biol Chem. 1996;271(33):20035–46.

    Article  CAS  PubMed  Google Scholar 

  71. Gooden M, Lampen M, Jordanova ES, Leffers N, Trimbos JB, van der Burg SH, Nijman H, van Hall T. HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8+ T lymphocytes. Proc Natl Acad Sci U S A. 2011;108(26):10656–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Benevolo M, Mottolese M, Tremante E, Rollo F, Diodoro MG, Ercolani C, Sperduti I, Lo Monaco E, Cosimelli M, Giacomini P. High expression of HLA-E in colorectal carcinoma is associated with a favorable prognosis. J Transl Med. 2011;9:184.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Levy EM, Bianchini M, Von Euw EM, Barrio MM, Bravo AI, Furman D, Domenichini E, Macagno C, Pinsky V, Zucchini C, Valvassori L, Mordoh J. Human leukocyte antigen-E protein is overexpressed in primary human colorectal cancer. Int J Oncol. 2008;32(3):633–41.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Seoul National University Bundang Hospital Research Fund (Grant Number: 02-2019-002).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, HN, YP, KL, HL; methodology, SM, JK, YK; formal analysis, YP, SN, JK, YK; investigation, SA, DP, HK; data curation, YP, SN; original draft preparation, HN, YP; review and editing, SA, DP, HK, KL, HL; supervision, KL, HL; funding acquisition, KL, HL. All the authors critically revised the manuscript and gave final approval and agreed to be accountable for all aspects of the work, to ensure its integrity and accuracy. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Kyu Sang Lee or Hye Seung Lee.

Ethics declarations

Ethics approval and consent to participate

This article does not contain any studies with human or animal subjects performed by any of the authors. This study was approved by the Institutional Review Board (IRB) of Seoul National University Bundang Hospital (SNUBH IRB B-1606/349–308). The requirement for infromed consent was waived.

Consent for publication

None.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1

. List of reagents and software used in the present study

Additional file 2: Table S2

. The panel information and order of sequential IHC antibody.

Additional file 3: Figure S1

. Representative figures of immune and tumor cells expressing CD3 (A), CD57 (B) and cytokeratin (C) (40×). Cells expressing each molecular marker were visualized by assigning colors, CD3 (D), CD57 (E), cytokeratin (F).

Additional file 4: Table S3

. Immune cell density in spatial context.

Additional file 5: Figure S2

. The proportion of each immune cells. T cells, macrophages, CD57+ NK cells, and B cells among CD16+ immune cells (A), and NKG2A+ immune cells (B).

Additional file 6: Figure S3

. Comparison of immune cells according to EBV and microsatellite status. The number of B cells (A), macrophages (B), and CD57+ NK cells (C) did not differ significantly between EBV-negative gastric cancer and EBV-positive gastric cancer. Similarly, the number of T cells (D), B cells (E), macrophages (F), and CD57+ NK cells (G) did not differ significantly between microsatellite unstable gastric cancer and microsatellite stable gastric cancer.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Na, H.Y., Park, Y., Nam, S.K. et al. Prognostic significance of natural killer cell-associated markers in gastric cancer: quantitative analysis using multiplex immunohistochemistry. J Transl Med 19, 529 (2021). https://doi.org/10.1186/s12967-021-03203-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-021-03203-8

Keywords