In this study, to understand whether T-cell exhaustion could take place in DLBCL, RNA-sequencing data associated with the features of T-cell exhaustion, including effector molecules and inhibitory receptors, were mined by means of the GEPIA and TIMER 2.0 databases. Compared with normal controls, TNFA, IFNG and GZMA, all three classical effector molecules of cytotoxic CD8 + T cells were found to be dramatically reduced in DLBCL. Meanwhile, the inhibitory receptors we obtained, including TIGIT, TIM3, PD-L1 and PD-1, whose expression was significantly elevated in DLBCL. Moreover, some important inflammatory cytokines were reported to be capable of promoting T-cell exhaustion, for instance, IL-10 and IL-6, whose expression was markedly reduced in DLBCL relative to the normal control. Notably, CD8A and FOXP3, reflecting the infiltrated status of CD8 and Treg cells, were expectedly downregulated in DLBCL in comparison with the normal control. In addition, DKK3 and WNT2, two key members of the WNT signaling pathway involved in T-cell exclusion in the tumor microenvironment, were found to be profoundly upregulated in DLBCL. Aside from these key molecules described above, both PTEN and STAT3, the two important molecules we have been focusing on and interested in, were shown to be lower in DLBCL, but without being statistically significant, in comparison with the normal control. Taken together, all these data we gleaned explicitly and strongly indicate that T-cell exhaustion took place in DLBCL.
The original observation describing the phenomenon of exhausted T cells came from the human immunodeficiency virus (HIV) infection study [12], which led to a proliferation of literature concerning T-cell exhaustion since the publication of this observation. Subsequently, this phenomenon was rapidly and extensively reproduced in both human and animal models infected with human hepatitis B virus (HBV) and hepatitis C virus (HCV)[7], other than HIV infection. Notably, observations regarding T-cell exhaustion were made from virus infection[13] to tumors [14] wherein T-cell dysfunction or exhaustion also existed. A great deal of studies revealed that T-cell exhaustion took place in tumors, including hematologic malignancies [3, 15, 16] as well as solid tumors [17, 18], albeit mechanistically less well-known. To understand the nuts and bolts of the big picture regarding T-cell exhaustion, we refer the readers to previous seminal reviews [1, 19] written on molecular and cellular insights into T-cell exhaustion, and here, we mainly focus our discussion on more recent findings with respect to CD8 + T-cell exhaustion [20, 21] that occurred in DLBCL. Marked by the continuous loss of effectors (such as PRF1, GZMB, and TNFA) and the expression of multiple inhibitory receptors (e.g., PD-1, LAG3, and TIGIT) [21, 22], CD8 + T-cell exhaustion was tentatively deemed to be occurring. In our study, taking advantage of available data regarding DLBCL from the GEPIA and TIMER databases, we showed canonical effector molecules of cytotoxic CD8 + T cells, including CD8A, TNFA, IFNG and GZMA, whose mRNA levels were strikingly upregulated in DLBCL tissues in comparison with normal controls. Interestingly, immune inhibitory receptors, including TIGIT[3], PD-1[3], PD-L1, and TIM3[5], were also remarkably elevated in DLBCL, along with the upregulation of inflammatory cytokines, including IL-2 [23], IL-6 [24]and IL-10[25], which have been reported to actively participate in CD8 T + cell exhaustion.
Considering these findings, we hypothesized that T-cell exhaustion could take place in the tumor microenvironment of DLBCL. Despite the hypothesis we proposed here lacking experimental evidence supporting it, Chen B et al [15] detected, using immunostaining, the expression of immune checkpoint molecules, including PD-1, PD-L1, TIM-3 and LAG-3, demonstrating that the four checkpoints were all markedly higher in DLBCL than in normal controls, which was totally in support of our bioinformatic data. However, we did not find any significant difference in survival, regardless of DFS or OS, regarding the expression of PD-1, PD-L1 and TIM-3. Similar conclusions can also be found in other published literature [26, 27].
Prognositcally, among all biomarkers we were interested in in this study, only TIGIT, whose mRNA expression level was shown to be pronouncedly associated with survival of DLBCL, in terms of disease-free survival (DFS). For the remainder, a nonsignificant association can be observed with survival. It would be somewhat discouraging to exhibit that in overall survival (OS) analysis, no significant association can be found between mRNA expression of biomarkers we were concerned with and overall survival of DLBCL. Inconsistent with our data, Nakayama S et al [28] evaluated the expression of TNFA in 62 cases of DLBCL tissues using immunohistochemistry and concluded that TNFA expression was a significant predictor for OS and DFS in DLBCL. Similar to TNFA, there was also some controversy regarding the prognostic significance of PD-L1 expression, with some [29, 30] supporting that PD-L1 expression can be significantly predictive of OS or DFS, while others [27, 31, 32] showed a nonsignificant association of PD-L1 with survival in DLBCL. In addition to PD-L1, the same can be applied to TIM3 in DLBCL, with some [15] supporting while others [26] negating the prognostic significance of TIM3 expression in DLBCL. Regarding PTEN mRNA expression, we did not find a significant difference in survival in DLBCL. The same was true for Foxp3, WNT2, DKK3, STAT3 and inflammatory cytokines, including IL-10 and IL-6.
Notably, deletion of PTEN has been revealed to be linked with immune suppression [33] and T-cell exclusion [34] in the tumor microenvironment. Suggested by these reports, we therefore explored the correlation of PTEN expression with the infiltration of tumor-infiltrating lymphocytes we were most concerned with, showing that there was a significantly positive correlation between PTEN expression and the infiltration of M1 macrophages, CD8 + T cells and neutrophils in DLBCL. For Treg cells, a significantly negative correlation can be observed between PTEN expression level and infiltration of Treg cells, meaning that the lower the expression of PTEN was, the less infiltration of CD8 + T cells will be; conversely, the more Treg cells will be infiltrating the tumor microenvironment of DLBCL. Fundamentally consistent with these findings, there were profoundly positive correlations between PTEN expression and CD8A, IFNG, PD-L1, PD-L2, IL-10 and IL-6 in DLBCL. On the other hand, a question naturally aroused here is whether PTEN loss is the sole cause of the immune suppression of CD8 + T cells in tumors. Combining these findings with the related literature seems to suggest that it is not necessarily the only reason.
Apart from PTEN deficiency, an abnormal Wnt/β-catenin signaling pathway in tumor cells has been repeatedly demonstrated to be responsible for T-cell exclusion or dysfunction in various types of tumors [35–38]. Given these findings, special attention was given to WNT2 and DKK3, which are inhibitor molecules of the Wnt/β-catenin pathway, in DLBCL, and WNT2 was significantly higher, along with DKK3, in DLBCL than in normal controls. The data we gleaned here explicitly indicate that the Wnt/β-catenin pathway was partly activated [39], which might play a crucial role in preventing CD8 + T cells from infiltrating the tumor microenvironment of DLBCL. In addition, the signal transducer and activator (STAT) signaling pathway [11] is another pivotal pathway involved in the pathogenesis of DLBCL. STAT3 mRNA expression was lower in DLBCL than in normal controls, but the difference was not statistically significant. Moreover, no significant difference in survival was observed between high and low mRNA expression of STAT3 in DLBCL. Our data were different from the study by Fei Y et al [40], who reported that soluble PD-L1 and STAT3 derived from plasma can predict the prognosis in DLBCL. In light of this controversy, in studies that follow, more work needs to be done to determine the prognostic significance of STAT3, no matter what level it is on, including transcriptional, translational and posttranslational modification.