H. pylori infection was associated with better survival in Chinese gastric cancer patients
In order to test the infection of H. pylori on the survival of gastric cancer, a total of 488 patients were enrolled in the study and divided into two groups according to the H. pylori infection status (Supplementary Table 1). Among these patients, 372 men and 116 women participated with median age of 63 years, in which 224 were negative for H. pylori (hereafter “H. pylori -”) whereas the rest of the participants(264 patients)were H. pylori positive (hereafter “H. pylori +”). The H. pylori positive rate was 54.10%, which was consistent with the overall H. pylori infection rate in Chinese population with 55.8% (21). In addition, there were more smokers and drinkers in the H. pylori + group, while the patients with stage IV was relative lower than the H. pylori - group. For the chemotherapy after surgical operation, 67.41% patients in the H. pylori - group accepted the chemotherapy, higher than those in the H. pylori + group (58.33%).
As shown in Table 1, the median survival time was 36.33 months, and 190 patients (38.93%) died of gastric cancer in our cohort. The median survival time was 142.3 months in patients positive for H. pylori, compared with 82.1 months in patients negative for H. pylori (Figure 1A, HR=0.64, 95% CI=0.48-0.85, P=2.35×10-3). In addition, we found that age, gender, clinical stage, and radiotherapy were significantly associated with the survival of our subjects with gastric cancer (Table 1, Figure1B and Supplementary Figure 1). In multivariate analyses, we revealed that positive H. pylori status was significantly associated with better prognosis of gastric cancer, with adjustments of age, gender, clinical stage and radiotherapy status. Additionally, age and gender were also prognostic factors for overall survival (Table 1). When adjusted for all variables in our study, only clinical stage and H. pylori status were independent prognostic factor for survival, in which patients with positive H. pylori status had a significantly longer survival time compared to patients with negative H. pylori status (Table 1, HR=0.74, 95% CI=0.55-0.99, P= 0.045).
Table 1
Univariate and multivariate analysis of predictive factors for overall survival in 488 patients with gastric cancer
Variables
|
Patients
|
Deaths
|
MST (mo)
|
HR (95% CI)
|
P
|
Adjusted HR (95% CI) a
|
Adjusted Pa
|
Adjusted HR (95% CI) b
|
Adjusted Pb
|
N=488
|
N=190
|
Age
|
|
|
|
|
|
|
|
|
|
< 63
|
244
|
83
|
133.3
|
1.00
|
|
1.00
|
|
1.00
|
|
≥ 63
|
244
|
107
|
84.3
|
1.44 (1.08-1.92)
|
0.012
|
1.39 (1.04-1.86)
|
0.025
|
1.32 (0.98-1.78)
|
0.070
|
Gender
|
|
|
|
|
|
|
|
|
|
Male
|
372
|
157
|
85.2
|
1.00
|
|
1.00
|
|
1.00
|
|
Female
|
116
|
33
|
NA
|
0.64 (0.44-0.93)
|
0.018
|
0.68 (0.46-0.99)
|
0.044
|
0.64 (0.43-0.95)
|
0.026
|
Smoker
|
|
|
|
|
|
|
|
|
|
No
|
324
|
123
|
101.3
|
1.00
|
|
1.00
|
|
1.00
|
|
Yes
|
164
|
67
|
83.4
|
1.01 (0.75-1.36)
|
0.946
|
0.95 (0.70-1.31)
|
0.771
|
1.02 (0.68-1.54)
|
0.921
|
Drinker
|
|
|
|
|
|
|
|
|
|
No
|
351
|
132
|
101.3
|
1.00
|
|
1.00
|
|
1.00
|
|
Yes
|
137
|
58
|
83.4
|
1.02 (0.75-1.39)
|
0.905
|
0.89 (0.64-1.22)
|
0.458
|
0.88 (0.58-1.33)
|
0.536
|
Clinical stage
|
|
|
|
|
|
|
|
|
|
AJCC I
|
102
|
10
|
NA
|
1.00
|
|
1.00
|
|
1.00
|
|
AJCC II
|
71
|
21
|
142.3
|
2.69 (1.26-5.73)
|
0.010
|
2.52 (1.18-5.37)
|
0.017
|
2.61 (1.22-5.59)
|
0.013
|
AJCC III
|
137
|
63
|
83.4
|
5.19 (2.66-10.11)
|
1.37×10-6
|
4.72 (2.41-9.24)
|
6.00×10-6
|
4.92 (2.50-9.66)
|
3.77×10-6
|
AJCC IV
|
178
|
96
|
30.2
|
13.77 (7.07-26.82)
|
1.27×10-14
|
11.94 (6.09-23.40)
|
1.27×10-14
|
12.56 (6.39-24.70)
|
2.25×10-13
|
P trend
|
|
|
|
2.37 (1.98-2.84)
|
< 2.0×10-16
|
2.27 (1.89-2.72)
|
< 2.0×10-16
|
2.29 (1.91-2.76)
|
< 2.0×10-16
|
Chemotherapy
|
|
|
|
|
|
|
|
|
No
|
183
|
65
|
NA
|
1.00
|
|
1.00
|
|
1.00
|
|
Yes
|
305
|
125
|
88.0
|
1.06 (0.78-1.43)
|
0.713
|
0.81 (0.58-1.12)
|
0.200
|
0.81 (0.58-1.13)
|
0.212
|
Radiotherapy
|
|
|
|
|
|
|
|
|
No
|
429
|
158
|
133.3
|
1.00
|
|
1.00
|
|
1.00
|
|
Yes
|
59
|
32
|
63.7
|
1.73 (1.18-2.53)
|
5.12×10-3
|
1.32 (0.90-1.95)
|
0.158
|
1.41 (0.94-2.12)
|
0.096
|
H. pylori
|
|
|
|
|
|
|
|
|
|
Negative
|
224
|
100
|
82.1
|
1.00
|
|
1.00
|
|
1.00
|
|
Positive
|
264
|
90
|
142.3
|
0.64 (0.48-0.85)
|
2.35×10-3
|
0.74 (0.56-1.00)
|
0.049
|
0.74 (0.55-0.99)
|
0.045
|
aAdjusted for age, gender, clinical stage, radiotheraphy status and H. pylori infection status.
bAdjusted for age, gender, smoking, drinking, clinical stage, chemotheraphy status, radiotheraphy status and H. pylori infection status.
H. pylori, Helicobacter pylori
|
Especially, for patients with early and intermediate stage gastric cancer (ie, AJCC I, II and III), we found a significant difference in overall survival between those who were positive for H. pylori and those who were negative (P=0.024); however, we found no such association for patients with advanced cancer (ie, AJCC IV, P=0.979, Figure 1C). Findings were much the same on stratification of patients by other classifications for early and intermediate versus advanced disease, like patients who were positive for H. pylori status had significantly higher survival than did those who were negative only for those with tumor depth without invasion of visceral peritoneum or adjacent structures (ie, T1, T2 and T3, P=0.028, Supplementary Figure 2A) and for those with nodal involvement less than 2 (ie, N0 and N1, P=1.3×10-3, Supplementary Figure 2B). Hence, we identified H. pylori as an independent, beneficial prognostic factor, the effect of which was most pronounced in patients with early-stage cancer.
H. pylori affect gastric cancer mainly targeting CD8+ T cells within tumor microenvironment
It has been suggested that tumor-specific immune responses are upregulated in patients with gastric cancer who are positive for H. pylori (14). In order to evaluate the possible immune mechanism of H. pylori on gastric cancer microenvironment, we first explored the content of tumor-infiltrating lymphocytes (TILs) and found that CD3+ T cells were the dominant TIL population both in H. pylori - and H. pylori + group (Supplementary Figure 3A). As CD8+ T cells play a pivotal role in clearing intracellular pathogens and tumors (22), we next examined the frequency of naive and memory subsets of CD3+CD8+ T cells based on CD45RA and CCR7 expression. We found that the effector memory T cell (TEM, CD45RA–CCR7–) followed by central memory T cell (TCM, CD45RA–CCR7+) were the most prevalent subsets. In addition, the frequency of TEM in H. pylori + gastric cancer was much more compared with H. pylori - gastric cancer (Supplementary Figure 3B), which indicated that H. pylori + gastric cancer have a stronger immediate effector function than H. pylori - gastric cancer.
Single-cell transcriptomic profiling of the T cells in gastric cancer tumors among different H. pylori infection status
To shed light on the complexity of tumor-infiltrating T cells in gastric cancer in an unbiased manner, we further conducted 3’ droplet-based scRNA-seq (BD RhapsodyTM) on 18,717 flow-sorted CD3+CD45+ T cells freshly isolated from 3 H. pylori - and 3 H. pylori + gastric cancer patients (Figure 2A and Supplementary Table 2). T cell clusters were visualized using t-distributed stochastic neighbor embedding (t-SNE) after preprocessing, normalization and batch correction (Supplementary Figure 4). Overall, we identified ten unique clusters based on their gene expression profiles, including six distinct CD8+ T cell clusters and four distinct CD4+ T cell clusters (Supplementary Figure 5A-C). Each of the ten clusters harbored differentially expressed genes (DEGs) representing distinct cell types or subtypes (Supplementary Table 3 and Supplementary Figure 5D).
To address the intrinsic T cell heterogeneity, we applied unsupervised re-clustering based on t-SNE and identified ten CD4+ and twelve CD8+ clusters (Figure 2B, Supplementary Figure 6). We further surveyed the expression and distribution of canonical T cell markers among these clusters, respectively (Figure 2C, Supplementary Figure 7). Among the ten CD4+ T cell clusters (Supplementary Table 4), we found that CD4-C2-FOXP3, CD4-C3-TNFRSF4, CD4-C8-COL5A3 and CD4-C9-IFIT1 clusters represented regulatory T cells (Tregs) with high expression levels of FOXP3, IKZF2 and IL2RA, as well as coinhibitory molecules TIGIT and CTLA4 (Figure 2C, Supplementary Figure 7A). Cells in CD4-C5-CXCL13 and CD4-C6-TOX2 showed high expression levels of PDCD1 and CXCL13 (Figure 2C, Supplementary Figure 7A), thus representing follicular T helper cells involved in the formation of ectopic lymphoid-like structures in inflammatory sites (23). Two clusters of CD4+ T cells (CD4-C1-CCR7 and CD4-C7-FBLN7) were characterized by a gene signature including CCR7, LEF1, TCF7 and SELL (Figure 2C, Supplementary Figure 7A), which are typical features of naive T cells. Of interest, T cells from CD4-C0-CCL5 and CD4-C4-SLC4A10 showed high expression levels of CD69, which has recently been reported to be elevated in activating MAIT cells of patients with COVID-19 (24). Given that the MAIT cells can display effector functions involved in the defense against infectious pathogens (25). We found that the proportion of these two clusters were relatively increased in H. pylori + (Supplementary Figure 7B), which may be activated by H. pylori.
When focusing on the different CD8A+ clusters (Supplementary Table 5), we noted that CD8-C7-KLF2 characterized by a gene signature including CCR7, LEF1, TCF7 and SELL, which are typical features of naive T cells (Figure 2C, Supplementary Figure 7C). Among identified CD8+ TEM, which characterized by low expression of CCR7, CD8-C0-CXCL13, CD8-C5-XCL2, CD8-C8-HSPA1B and CD8-C10-IFIT1 T cells with an activated cellular state (CD8+ TEM activated-state), characterized by the expression of effector molecules such as IFNG, CCL4 and CCL5 (Figure 2C, Supplementary Figure 7C); and CD8-C6-FGFBP2 T cells, with features of natural killer (NK) cells, expressing genes such as NKG7, FGFBP2, and FCGR3A, which we refer to as ‘CD8+ TEM NK-like’ (Figure 2C, Supplementary Figure 7C). Interestingly, CD8-C0-CXCL13 and CD8-C10-IFIT1 also exhibited variable expression of exhaustion markers, like LAG3, HAVCR2 and PDCD1 (Figure 2C, Supplementary Figure 7C), indicating an activation-coupled exhaustion program putatively caused by both H. pylori infection and tumor cells. Besides, cytotoxic CD8+ T cells (CD8+ TCYTOTOXIC) from CD8-C1-ZNF683 and CD8-C9-KIR2DL3 showed high expression levels of cytotoxicity-related genes such as GNLY, GZMB, PRF1 and tissue residency gene ZNF683 (HOBIT) (Figure 2C, Supplementary Figure 7C). We further noted that CD8-C4-AREG cluster had expression of molecules suggestive of a tissue-resident memory T cells (TRM) with high expression of ITGAE (CD103), ITGA1 and CD69, while showing low expression of SELL, S1PR1 and KLF2 (Figure 2C, Supplementary Figure 7C), akin to TRM cells described in humans and mice (26). CD8-C11-TRDC was assigned to T-gd cells, expressing TRDC, TRGC1, and genes associated with cytotoxicity, including GNLY (27), while CD8-C2-KLRB1 cluster was characterized by high expression of KLRB1, which are known hallmarks of mucosal-associated invariant T cells (Figure 2C, Supplementary Figure 7C) (28).
Unlike CD4+ T cells (Supplementary Figure 7B), the clusters in CD8+ T cells appeared to exhibit distinct distributions. Especially, CD8-C0-CXCL13, contained mostly cells of H. pylori +, while CD8-C4-AREG and CD8-C7-KLF2 were almost exclusively populated with cells from H. pylori - (Supplementary Figure 7D). We further analyzed the developmental fate of these differentially distributed cells using the Monocle 2 algorithm to establish a pseudotemporal ordering reflective of cell lineage. Since cluster CD8-C7-KLF2 was naive T cells, two major developmental trajectories were observed (Figure 2D), in which the TRM-like and TEM activated-state cells were located at opposite ends of the pseudotime path, supporting the distinct gene expression profiles of these cells. In addition, we also calculated a cytotoxicity score and exhaustion score for each cell based on the expression of canonical markers. Along the trajectory, most CD8+ T cells exhibited gradually increasing cytotoxic activity, which was accompanied by gradually increasing exhaustion (Figure 2E). Especially, the score of cytotoxic activity was downregulated in trajectory 1 toward cluster CD8-C4-AREG cells, while was increased in trajectory 2 of both CD8-C0-CXCL13 and CD8-C10-IFIT1 T cells (Figure 2F), which retained the ability for active cell division in the immune microenvironment. Hence, we show using single-cell analysis that the CD8+ population is heterogeneous with distinct subsets.
H. pylori infection promote intratumoral immune activation with enhanced interaction between CD8+ T cells and epithelium
We first focusing on CD8-C0-CXCL13 cluster, we noted that except for the high expression of CXCL13, it also specifically expressed genes like MYO7A, TOX and PHLDA1 (Figure 3A). We then compared the single cell data between CD8-C0-CXCL13 group and the other groups to determine the differential expression genes, 192 up-regulated and 118 down-regulated genes were detected in CD8-C0-CXCL13 T cells (p.adj ≤ 0.01 and |log2FoldChange| ≥ 0.25) (Supplementary Figure 8A and Supplementary Table 6). Gene ontology (GO) functional enrichment analysis revealed that these upregulated genes in CD8-C0-CXCL13 T cells were enriched for signaling pathways such as T cell activation, regulation of lymphocyte activation and regulation of T cell activation (Supplementary Figure 8B). There are also other enriched gene sets that are crucial for anti-pathogen infection such as Th1 and Th2 cell differentiation, Th17 cell differentiation as well as antigen processing and presentation by KEGG analysis (Supplementary Figure 8C), which was potentially associated with H. pylori infection.
To further decipher the molecular characteristics difference of CD8-C0-CXCL13 T cells resulted from H. pylori, gene set enrichment analysis (GSEA) was conducted and we revealed that T cells from H. pylori + were associated with cell activation involved in immune response and regulation of response to cytokine stimulus, indicating a potential protection role against local H. pylori infections (Figure 3B). In addition, we investigated the expression level of genes between H. pylori infection status. Interestingly, we found that cytotoxicity-associated genes, such as IFNG and GZMB were upregulated in the patients with H. pylori +, while the expression of exhaustion marker PDCD1 was downregulated (Figure 3C). As expected, a higher level of IFNG and GZMB were associated with a better prognosis in gastric cancer (Supplementary Figure 8D, 8E).
Early studies established the concept that gastric epithelial cells from H. pylori infected patients contain increased TNF receptors against invading infection with subsequent activation of an adaptive immune response. Using data from single cell sequencing of stomach tissues (GSE134520), we estimated TNF-dependent T cell functions in CD8+ T cells and epithelium. We found prominent TNF-TNFRSF1A and TNF-DAG1 interaction in stomach, which was enhanced with H. pylori infection (Figure 3D), suggesting that the molecular interaction is crucial in creating an immune activation tumor microenvironment with the response to H. pylori infection. Notably, TNFRSF1A, not DAG1, was positively correlated with the CD8+ T cells signature as well as TEM signature, while negatively correlated with the naïve T cells in the TCGA STAD cohort using the TIMER2.0 webserver (Supplementary Figure 8F), which indicated that TNFRSF1A may play an important role to induce antitumor immunity against gastric cancer. More important, using bulk RNA sequencing data from normal gastric mucosa tissues, we found that the expression of TNFRSF1A was significantly increased in samples with H. pylori infection, while DAG1 was not changed (Figure 3E). Together, our results predicted that H. pylori infection potentially be able to recruit T cells into the tumor by TNFRSF1A-TNF interaction and then enhanced the immune activity.
As our data suggest that the CD8-C0-CXCL13 T cell subset is a highly prevalent effector population in the microenvironment of gastric cancer, we predicted that the single-cell-derived gene signature from the CD8-C0-CXCL13 cluster (Supplementary Table 5) would provide important prognostic information. Using available gene expression data, we found that the CD8-C0-CXCL13 signature was significantly associated with improved overall survival (Figure 4A). We further performed multicolor immunofluorescence staining on stroma and tumor sections from gastric cancer patients. Among CD8+ T cells, CXCL13 and CD103 were both activated by H. pylori infection in stroma and tumor tissues (Figure 4B), supporting the presence of these activated cells in gastric cancer.
TRM cells marked by PTGER2 worsen prognosis in H. pylori negative gastric cancer
It has been reported that there exists virus- or other pathogen-specific (bystander) CD8+ TRM-like cells in tumor and can be re-activated to induce antitumor immunity (29). However, we found 151 down-regulated genes in CD8-C4-AREG T cells (p.adj ≤ 0.01 and |log2FoldChange| ≥ 0.25) (Supplementary Figure 9A and Supplementary Table 7), enriched for signaling pathways such as T cell activation, response to IFN-γ and antigen processing and presentation (Supplementary Figure 9B), which indicated an immunosuppression state in CD8-C4-AREG TRM cells. In addition, though we found that the gene expression profiles of these T cells were similar between H. pylori - and H. pylori + (Figure 5A), GSEA analysis revealed that T cells from H. pylori + were associated with inflammatory response and cytokine mediated signaling pathway (Figure 5B, Supplementary Figure 9C), while response to steroid hormone pathway was enriched in T cells from H. pylori – (Supplementary Figure 9C), in which AREG and PTGER2 were both increased (Figure 5C). Similarly, further immunohistochemistry staining also confirmed that AREG and PTGER2 were both inactivated by H. pylori infection in stroma and tumor tissues (Figure 5D). Using prognostic data from TCGA and GSE15459, we show that the PTGER2 can discriminate between patients with high CD8 expression, in which high PTGER2 expression was significantly associated with worse prognosis, but not in patients with low CD8 expression (Figure 5E and Supplementary Figure 9D). These data suggest that TRM cells with high expression of PTGER2 may be the key therapeutic target to improve the clinical outcomes of gastric cancer.