Abstract Tumor-infiltrating T cells are essential players in tumor immunotherapy. Great progress has been achieved in the investigation of T cell heterogeneity. However, little is well known about the shared characteristics of tumor-infiltrating T cells across cancers. In this study, we conduct a pan-cancer analysis of 349,799 T cells across 15 cancers. The results show that the same T cell types had similar expression patterns regulated by specific transcription factor (TF) regulons across cancers. Multiple T cell type transition paths were consistent in cancers. We found that TF regulons associated with CD8^+ T cells transitioned to terminally differentiated effector memory (Temra) or exhausted (Tex) states were associated with patient clinical classification. We also observed universal activated cell-cell interaction pathways of tumor-infiltrating T cells in all cancers, some of which specifically mediated crosstalk in certain cell types. Moreover, consistent characteristics of TCRs in the aspect of variable and joining region genes were found across cancers. Overall, our study reveals common features of tumor-infiltrating T cells in different cancers and suggests future avenues for rational, targeted immunotherapies. Keywords: MT: Bioinformatics, tumor-infiltrating T cells, pan-cancer, single-cell sequencing, TF regulon, state transition, cell-cell communication, T cell receptor Graphical abstract graphic file with name fx1.jpg [47]Open in a new tab __________________________________________________________________ Jiang and colleagues integrated extensive single-cell data from 15 cancers as well as bulk gene expression, systematically delineated the immune ecosystem of tumor-infiltrating T cells and reveals the landscape of shared characteristics of tumor-infiltrating T cells across cancers. Introduction Cancer development and progression are associated with alterations of the tumor microenvironment, which is complex and continuously evolving.[48]^1 Tumor-infiltrating T cells are important components of the tumor microenvironment,[49]^2 and they display a wide range of (dysfunctional) functional states that are shaped by a variety of inhibitory signals emerging in the tumor microenvironment.[50]^3 CD8^+ T cells recognize and kill cancer cells directly after recognizing antigens, and adjust their developmental lineage and status in response to various cues. CD4^+ T cells coordinate a variety of immune responses to enhance or suppress immunity against cancer cells.[51]^4 The rise of single-cell technology enables the study of T cell state transition in the tumor microenvironment at single-cell resolution. Existing studies of tumor-infiltrating T cells cover a wide range of cancers. T cells show a continuous trajectory of activation and differentiation in breast cancer (BRCA).[52]^5 In melanoma, a continuous transition of CD8^+ T cells from an effector state to a dysfunctional state has been found.[53]^6 In addition to exhausted CD8^+ T cells, pre-exhausted cells were also identified in lung cancer patients.[54]^7 The study of bladder cancer (BLCA) patients identified several tumor-specific CD4^+ T cells, including regulatory T cells (Tregs) and cytotoxic CD4^+ T cells.[55]^8 Meanwhile, the presence of cytotoxic CD4^+ T cells was also observed in melanoma patients.[56]^9 Most of these previous studies have focused on the heterogeneity of different T cell types within cancer. Elucidating the common functional status and differentiation process of T cells in different cancers is of great significance for understanding cancer immunity. A wide range of transcription factor (TF) regulation has been observed in cancers.[57]^10 For example, hypoxia induces the expression of HIF-1α and subsequently stimulates the expression of CXCL12.[58]^10 Continuously activated STAT3 and STAT5 increase the proliferation, survival, and invasion of tumor cells, and inhibit antitumor immunity.[59]^11 Moreover, The complex regulatory network of TFs is a key factor controlling the development and differentiation of T cells.[60]^12 For instance, TCF1 is essential for the early differentiation of T follicular helper (Tfh) cells, and the lack of TCF1 impairs the generation of Tfh cells.[61]^13 FOXP3 is a TF of Tregs that can inhibit immune response and play a critical role in maintaining immune tolerance.[62]^14 However, the development of T cells is dynamic and involves a variety of cellular states, and current studies are insufficient to elucidate the regulation of the complex immune microenvironment in tumors. Further studies are still needed to investigate the key TF regulators in pan-cancer. Here, we conducted a systematically comprehensive analysis of T cells in 15 cancers. We identified T cells with different functional states, as well as active TF regulons for each cell state in pan-cancer. T cell state transitions were accompanied by changes in TF regulon expression. Furthermore, on the basis of cell-cell interaction analysis, we identified common activated interaction pathways in cancers. In addition, we characterized clonal expansion, preference variable (V) and joining (J) segment usage, and CDR3 characteristics of T cell receptor (TCR) in CD4^+ and CD8^+ T cells across cancers. Overall, our study of common features in tumor-infiltrating T cells across cancers may provide new insights into tumor immunity. Results Sub-phenotyping of T cells across 15 cancers We collected 15 cancer single-cell RNA sequencing (scRNA-seq) datasets on T cells ([63]Figure 1A; [64]Table S1). A standard single-cell analysis process was performed to identify T cells in esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC) datasets because their cell type labels were not available ([65]Figure S1). After quality-control filtering, a total of 349,799 T cells were obtained from tumor, adjacent normal tissue, and peripheral blood of 101 patients, in which 35.5%–87.5% cells had TCR data ([66]Figure 1A). To further understand the characteristics of tumor-infiltrating T cells, we performed unsupervised clustering analysis of CD4^+ and CD8^+ T cells, respectively. A total of 19 CD4^+ and 17 CD8^+ T cell clusters were identified ([67]Figures 1B, [68]S2A, and S2B). These clusters exhibited distinct tissue distributions ([69]Figures 1C and [70]S2C). Naive clusters were dominant in PBMCs, while Trm were mainly distributed in normal tissue. CD4_CX3CR1^+ Temra was mainly distributed in adjacent normal tissue and PBMCs and CD8_CX3CR1^+ Temra was mainly distributed in PBMCs. CD8_CXCL13^+ Tex and Tregs were tumor enriched. In addition, we identified cell clusters with high expression of proliferative genes in both CD4^+ and CD8^+ T cells. Cell-cycle score showed that these clusters were in G2M or S phase, and these clusters also showed high expression of exhausted or Treg-related marker genes ([71]Figures 1D, [72]S2A, and S2B). Figure 1. [73]Figure 1 [74]Open in a new tab T cell clusters across cancers (A) The data overview of 15 cancers. (B) UMAP visualization of CD8^+ and CD4^+ T cell clusters. (C) The R(O/E) values of each cluster in tumor, normal, and PBMCs. The chi-square test was used to calculate the R(O/E) value. R(O/E) > 1 (above the dashed line) indicates enrichment. (D) Cell cycle score of T cell clusters. We observed similar distribution of typical T cell types in different cancers, including naive, memory, and exhausted T cells. ([75]Figure S3). However, there are some cell types that were heterogeneous across cancers. For example, MAIT cells were enriched in hepatocellular carcinoma (HCC). Studies have shown that MAIT cells can represent up to 45% of liver lymphocytes.[76]^15 Consistent with a previous study, CD8_CD160^+ T cells were intraepithelial lymphocytes, which were mainly distributed in colorectal cancer (CRC).[77]^16 Intraepithelial lymphocytes are an important contributors to intestinal immune homeostasis.[78]^17 Shared T cell state-transition paths in 15 cancers Because T cells can transition between different functional states, we employed Monocle3 to infer cell trajectory. The results showed a variety of state-transition paths between T cell clusters. For CD4^+ T cells, we found that CD4_CCL5^+ Tm and CD4_CXCR6^+ Th17 appeared to be a hub for the development of naive cells into other cell types ([79]Figure 2A). When developmental trajectories were inferred individually for each cancer, this phenomenon was found to be retained in B cell lymphoma (BCL), BLCA, esophageal cancer (ESCA), renal carcinoma (RC), thyroid carcinoma (THCA), and uterine corpus endometrial carcinoma (UCEC) ([80]Figure S4A). However, in ESCC and multiple myeloma (MM), only CD4_CXCR6^+ Th17 was shown as a hub, while in BRCA, CRC, HCC, NPC, non-small cell lung cancer (NSCLC), ovarian cancer (OV), and pancreatic cancer (PACA), CD4_CCL5^+ Tm was shown as a hub ([81]FigureS4A). CD4_CCL5^+ Tm and CD4_CXCR6^+ Th17 significantly share marker genes with downstream cell clusters ([82]Figure 2B). Enrichment analysis showed that the marker genes of CD4_CCL5^+ Tm and CD4_CXCR6^+ Th17 were simultaneously enriched in cell adhesion, differentiation, and migration ([83]Figure S4B). CD4_CX3CR1^+ Temra was transitioned from CD4_GZMK^+ Tem, and TCR similarity analysis showed that these two groups shared TCR clonotypes ([84]Figure S4C). We observed several clusters with high expression of Treg marker genes, among which CD4_TNFRSE9^+ Treg and CD4_TNFRSF9^− Treg were enriched in tumor and PBMCs, respectively ([85]Figures 1C and [86]S2C). In addition, high TCR similarity was observed among these clusters, especially between CD4_TNFRSE9^+ Treg and CD4_HSPA1A^+ Treg ([87]Figure S4C). Trajectory analysis revealed that CD4_ISG^+ Treg, CD4_NMB^+ Treg, and CD4_HSPA1A^+ Treg appeared as transitional states in different cancers and then developed into CD4_TNFRSE9^+ Treg ([88]Figures 2A and [89]S4A). The development path between CD4_STMN1^+ Treg and CD4_TNFRSE9^+ Treg was observed in ESCA, OV, RC, and PACA ([90]Figure S4A). Figure 2. [91]Figure 2 [92]Open in a new tab CD8_TXNIP^+ T cells migrated from PBMCs to tumor tissue and developed into Temra or Tex cells (A) UMAP showing the development trajectories of T cell clusters in all cancers inferred by Monocle3. (B) The overlapped marker genes of CD4_CXCR6^+ Th17, CD4_CCL5^+ Tm, CD4_Th1-like, CD4_ANXA1^+ Tm, and CD4_GZMK^+ Tem. Significance p value was calculated by hypergeometric test. (C) Shared marker genes between CD8_TXNIP^+ T cells and other T cells. Significance was calculated by hypergeometric test. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗∗p < 0.0001. (D) Migration potentials from PBMCs to tumor of CD8_TXNIP^+ T cells quantified by STARTRAC-pMigr. (E) Developmental transition of CD8_TXNIP^+ T cells with other T cells quantified by STARTRAC-pTran. (F) The similarity of GO terms enriched by CD8_TXNIP^+ T cell marker genes and annotated with word clouds. In CD8^+ T cells, we observed multiple exhausted paths, of which the path from CD8_Tn to CD8_IL7R^+ Tm to CD8_ZNF683^+ Trm to Tex in CRC, ESCA, MM, OV, PACA, THCA, and UCEC has been found previously ([93]Figures 2A and [94]S4D).[95]^18 In addition, the path from CD8_Tn to CD8_IL7R^+ Tm to CD8_CD69^+ Trm to CD8_ISG^+ T to Tex was found in CRC, ESCA, NPC, and THCA. Remarkably, we observed a branched structure in all cancers, whether trajectory inference was made collectively or individually for each cancer, which from CD8_TXNIP^+ T to Temra or Tex, with CD8_KLRG1^+ T and CD8_GZMK^+ T located in between ([96]Figures 2A and [97]S4D). High TCR similarity was also observed among these clusters ([98]Figure S4E). Moreover, we found that the marker genes of CD8_TXNIP^+ T were significantly shared with that of CD8_ZNF683^+ Trm, including ZNF683, CXCR6, ITGAE, and KLRC1 ([99]Figure 2C). CD8_TXNIP^+ T cells were enriched in all tissues, especially in PBMCs, but the clonality was highest in the tumor ([100]Figures S2C and S4F). STARTRAC-migr analysis revealed a high degree of TCR sharing of CD8_TXNIP^+ T cells between PBMCs and tumor, especially in MM ([101]Figure 2D).[102]^16 At the same time, STARTRAC-tran analysis indicated that CD8_TXNIP^+ T cells were highly associated with Trm and CD8_RPL36A^+ T cells ([103]Figure 2E). Enrichment analysis showed that CD8_TXNIP^+ T cells involved immune cell activation and proliferation, including T cell activation, lymphocyte differentiation, and regulation of T cell proliferation ([104]Figure 2F). Clinically relevant state-transition TF regulons across cancers TFs have been long recognized to play a central role in the maintenance of cell identity.[105]^19 We applied SCENIC to identify key regulons. SCENIC can simultaneously reconstruct gene regulatory networks and identify cell states from scRNA-seq data.[106]^20 Significantly active regulons were identified in each cancer ([107]Tables S2 and [108]S3). By calculating regulon specificity score (RSS), we identified cell-type-specific regulons. For tumor-infiltrating T cells, most regulons were cancer-type specific, indicating that T cells undergo specific transcriptional regulation in specific contexts ([109]Figure S5A). However, we also observed universal cell-type-specific regulons of tumor-infiltrating T cells across cancers. We screened each cell type for regulons that were present in at least five cancers and obtained 67 and 58 regulons in CD4^+ and CD8^+ T cell clusters, respectively. These regulons were significantly shared between CD4^+ and CD8^+ T cells ([110]Figures 3A, [111]S5B, and [112]S6A). The regulons were related to the function of the corresponding cell type. For example, naive specific regulons TCF7 and LEF1 were essential for early T cell development.[113]^21 Regulons in CD69^+ Trm included multiple zinc finger TFs (EGR1/2/3/4, KLF5/10). EGR1, EGR2, and EGR3 have been shown to regulate T cell activation, antigen-induced T cell proliferation, and effector differentiation.[114]^22^,[115]^23 TBX21 was identified as Temra-specific regulon in both CD4^+ and CD8^+ T cells ([116]Figure 3B). RUNX3, CD4_CX3CR1+ Temra-specific regulon, was a TF that regulated the cytotoxic phenotype in CD4^+ cytotoxic T cells.[117]^24 Another CD8_CX3CR1+ Temra-specific regulon, KLF2, was shown to inhibit tumor cell growth and migration in HCC,[118]^25 NSCLC,[119]^26 and ccRCC.[120]^27 ETV1, a Tex-specific regulon, was associated with immune invasion and poor prognosis in CRC.[121]^28 At the same time, we also calculated the correlation between the regulon activity score (RAS) of specific regulons and the TCR clonality in each cluster ([122]Figures 3B, [123]S5B, and [124]S6A). The activity of these regulons was correlated with TCR clonality in different cancers. For example, TBX21 was positively correlated with TCR clonality in five cancers. Figure 3. [125]Figure 3 [126]Open in a new tab Shared state-transition TF regulons across cancers (A) Venn diagrams showing the overlap cluster-specific regulons of CD4 and CD8. (B) The statistics of cluster-specific regulons (left) and the cluster-specific regulons in at least five cancers (right). The colors around the dot represent the relationship between RSS and TCR clonality. (C) The differential regulons between CD8_TXNIP^+ T, CD8_CX3CR1^+ Temra, and CD8_CXCL13^+ Tex. (D) Differential expression of regulons (C) in tumor compared with normal or PBMCs. (E) The expression level of regulons (C) in KIRC subtypes. (F) The Kaplan-Meier plot of KIRC patient-based classifications generated from consensus clustering. The survival difference among groups was calculated by log rank test. (G) Pathologic stage distribution of KIRC patients in each subtype. (H) The volcano plot of differentially expressed genes between pre-therapy and post-therapy patients in melanoma ([127]GSE115821). Differential expression analysis was performed using wilcox.test. (I) The volcano plot of differentially expressed genes between responder and non-responder patients in melanoma (PRJEB23709). Differential expression analysis was performed using wilcox.test. In view of the above-mentioned trajectories, we analyzed the differential regulons between CD8_TXNIP^+ T, Temra, and Tex in tumor tissue. Finally, in five or more cancers, we identified 14 regulons that were upregulated in Temra and downregulated in Tex (Temra-activated TFs) or downregulated in Temra and upregulated in Tex (Tex-activated TFs) compared with CD8_TXNIP^+ T, which we termed state-transition TFs ([128]Figure 3C). The expression of state-transition TFs was significantly different between tumor and other tissues ([129]Figure 3D). Remarkably, ETV1 was upregulated in tumor-infiltrating T cells of all cancers and its target genes shared by cancers were also upregulated ([130]Figure S6B). We further investigated the association between the expression of state-transition TFs and patient survival using TCGA cancer data, and the result showed that they significantly affected overall survival of patients with different cancers ([131]Figure S6C). Next, we performed cancer subtyping based on the expression of state-transition TFs in TCGA. In 13 cancers, the results showed that they divided tumor samples into different subtypes with significantly different survival probability ([132]Figure S7A). We observed that the C2 subtype in KIRC with high expression levels of Temra-activated TFs had a better prognosis ([133]Figures 3E and 3F). There were no significant differences in gender and age between the two classes, but the pathologic stage of most samples in C2 was low stage ([134]Figures 3G, [135]S7B, and S7C). Next, we explored the changes of these TFs after immune checkpoint blocking therapy by using cancer immunotherapy data that were downloaded from the TIGER database.[136]^29 The result showed that the expression of state-transition TFs was significantly upregulated after treatment in melanoma, except for IRF4 ([137]Figure 3H). Moreover, in another melanoma immunotherapy dataset, state-transition TFs were found to be highly expressed in responders, except for ETV1 and EZH2, which were highly expressed in non-responders ([138]Figure 3I). Overall, the T cell state-transition TF regulons we identified were helpful in tumor classification and could be used as independent prognostic factors and immunotherapy targets. Universal activated T cell interaction pathways in cancers Cancer development relies on a complicate network of cell-cell interactions.[139]^30 We inferred communications for all T cell clusters by CellChat.[140]^31 There were more interactions in tumor compared with other tissues, with the highest number in CRC and the fewest in BLCA ([141]Figure S8A). By calculating incoming and outgoing interaction strength, we found that there were significant differences in the interaction strength of cell clusters between different tissues ([142]Figure S8B). For example, in tumor and normal tissues, the interaction strength of CD4_TNFRSF9^+ Treg, CD8_ZNF683^+ Trm, and CD8_CXCL13^+ Tex was increased, while it was decreased in naive and Temra. Cell-cell interactions involve a variety of signaling pathways. Next we mapped the signaling pathways inferred from the different tissues of each cancer onto a two-dimensional manifold and clustered into different groups ([143]Figure S9A). Tumor-specific groups were observed in BCL, OV, and PACA, and there were normal-specific groups in PACA and OV. Groups dominated by different tissues have also been observed in other cancers. By computing the Euclidean distance between any pairs of shared signaling pathways in different tissues, we observed a large distance for CCL, CXCL, and FASLG pathways and a lesser distance for the IFN-II pathway ([144]Figure 4A). Furthermore, according to the information flow of each signaling pathway, we found that the changes of some pathways were different in cancers, including TNF, PARs, CLEC, ITGB2, and IL-16. In contrast, we also found 11 pathways (e.g., CCL, CXCL, and CD70) that were turned on or increased in the tumor of all cancers compared with other tissues, except BLCA, due to fewer interaction pathways found. The contribution of ligand-receptor (LR) pairs in these pathways was consistent in all cancers ([145]Figure 4B), and the expression of these LR genes was upregulated in tumor-infiltrating T cells ([146]Figure S9B). There was no cell type bias in the incoming and outgoing patterns of CADM, CCL, CXCL, CD70, FASLG, GALECTIN, and LIGHT pathways. But an interaction pattern preference for CD4^+ T cells or CD8^+ T cells was observed in VCAM, BLTA, and LT pathways ([147]Figure S9C). Figure 4. [148]Figure 4 [149]Open in a new tab Universal activated T cell interaction pathways in 15 cancers (A) The Euclidean distances of overlapping signaling pathways between different tissues in the shared two-dimensional manifold. Tumor vs. normal (left), tumor vs. PBMCs (right). (B) Circos plot of signaling pathways that significantly differ in overall information flow between different tissues (left) and the relative contribution of ligand-receptor (right). (C) The incoming signaling patterns of CD8_TXNIP^+ T cells in all cancers. (D) The outgoing signaling patterns of the GALECTIN pathway. (E) The expression of LGALS9 in each T cell cluster. (F) PPI network of ligand and receptor in the GALECTIN pathway; TFs in ISG^+ T cells and differential regulons between CD8_TXNIP^+ T, CD8_Temra, and CD8_Tex. Next, we found that GALECTIN was the incoming signaling pathway for the CD8_TXNIP^+ T cells in all cancers except OV ([150]Figure 4C). The outgoing cells of GALECTIN were mainly from STMN1^+ and ISG^+ T cell clusters ([151]Figure 4D). The contribution LR pair of the GALECTIN pathway was LGALS9-HAVCR2/CD45/CD44 ([152]Figure 4B). LGALS9 was specifically expressed in ISG^+ and STMN^+ T cells, especially in ISG^+ T cells ([153]Figure 4E). Protein-protein interaction (PPI) network analysis of LR pairs in GALECTIN, ISG^+ T cell cluster-specific regulons, and previously identified differential regulons between CD8_TXNIP^+ T, CD8_Temra, and CD8_Tex revealed that they interact with each other ([154]Figure 4F). This seems to imply a regulatory relationship between ISG^+ T cells and CD8_TXNIP^+ T cells. In addition, we observed that the outgoing signaling of the LT pathway was only found in Tregs ([155]Figure 5A). Its ligand LTA was the target gene of Treg-specific regulons REL and NFKB2, and the downstream TFs of its paired receptor were TNF signaling pathway-related genes ([156]Figure 5B). At the same time, we observed that TNF signaling pathway-related genes were simultaneously upregulated in tumor tissue ([157]Figure 5C). The activity of the TNF signaling pathway was different in each T cell type ([158]Figure 5D). These results suggested that Tregs activate the TNF signaling pathway in receptor cells through LTA-TNFRSF1B and then influence the fate of other cells ([159]Figure 5E). Figure 5. [160]Figure 5 [161]Open in a new tab Tregs activated the TNF pathway in other cells through the LT pathway (A) The outgoing signaling patterns of the LT pathway. (B) The Sankey plot of ligand-receptor in the LT pathway and TFs upstream of the ligand and downstream of the receptor. (C) Differentially expressed TNF signaling pathway-related genes in tumor compared with normal or PBMCs. (D) The TNF signaling pathway activity in each CD4 (top) and CD8 (bottom) clusters. (E) Schematic illustration of the LTA-TNFRSF1B interaction activating the TNF signaling pathway. The gene expression changes are consistent with (C). TCR landscape of T cells in pan-cancer TCR is a key factor mediating T cell immunity, and TCR analysis is helpful to understand T cell immunity. First, we analyzed the baseline repertoire of CD4^+ and CD8^+ T cells. The TCR diversity and clonality of T cells in tumor tissue were significantly higher than that in normal tissue and PBMCs ([162]Figure 6A). TCR clustering by GLPH2[163]^32 obtained 26,922 and 14,565 groups in CD4^+ and CD8^+ T cells, of which 22.1% and 47.6% were cancer type specific ([164]Tables S4 and [165]S5). T cells with the same V, J, and CDR3 sequences were defined as the same clonotype. The percentage of clonal TCRs of CD8^+ T cells was significantly higher than that of CD4^+ T cells ([166]Figure 6B). The clonal TCRs in CD8^+ T cells ranged from 7.6% of BCL to 20.1% of RC, and from 0.5% of NPC to 7.1% of RC in CD4^+ T cells. The clone size of individual clonotypes was proportional to the overall number of clonal TCRs in cancer ([167]Figure 6C). The maximum TCR clone size can reach to more than 2,000 in THCA, which was 200 times the maximum TCR clone size in NPC ([168]Table S6). In addition, there were significant differences in the proportion of V and J gene usage between CD4^+ and CD8^+ T cells ([169]Figure S10A). For example, TRAV1-2 was the most frequently used Vα gene segment in CD8^+ T cells, while TRAV9-2 was the most frequently used Vα gene segment in CD4^+ T cells. Next, we compared V and J segments across different tissues and found significant differences in the usage of V and J segments between tumor and other tissues, but only a few segments were consistent across cancers ([170]Figure S10B). Figure 6. [171]Figure 6 [172]Open in a new tab The consistent features of V and J segments and CDR3s across cancers (A) The TCR diversity and clonality of CD4^+ and CD8^+ T cells. The significant p values were calculated by Wilcoxon rank-sum test. (B) Comparison of clonal TCRs between CD4^+ and CD8^+ T cells. The significant p values were calculated by Wilcoxon rank-sum test. (C) The statistics of clonal TCRs in CD8^+ T cells and CD4^+ T cells. (D) The TCR clonality of each cluster in tumor, normal, and PBMCs. The significant p values were calculated by Wilcoxon rank-sum test. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001,∗∗∗∗p < 0.0001. (E) The fold change of clonality between CD8_CX3CR1^+ Temra and CD8_CXCL13^+ Tex in each cancer. (F) The line plot represents the trend of TCR clonality between T cell clusters on CD8_TXNIP^+ T cell development trajectories. (G) The proportion of amino acid types in CDR3s between T cell clusters on CD8_TXNIP^+ T cell development trajectories. The significant p values were calculated by Wilcoxon rank-sum test. Next, by analyzing the TCR characteristics of T cell clusters, we found that there was little clonal expansion of naive T cells, and the highly expanded TCRs were mainly in Temra and Tex cells ([173]Figure 6D). In addition, we compared the expansion of CD8_CX3CR1^+ Temra and CD8_CXCL13^+ Tex in tumor, and found that CD8_CX3CR1^+ Temra was highly expanded in BRCA, MM, THCA, and RC, while CD8_CXCL13^+ Tex was highly expanded in UCEC, PACA, ESCA, and ESCC ([174]Figure 6E). Since trajectory analysis showed that CD8_TXNIP^+ T cells transition to Temra or Tex, we investigate the changes of TCR characteristics among these clusters. The result showed that the clonality of TCR increased gradually along the developmental trajectory ([175]Figure 6F). In addition, we found that the amino acid types in the CDR3 region were different between the two development paths ([176]Figure 6G). For example, on the Tex path the proportion of polar amino acids in CDR3β significantly increased, but on the Temra path the proportion of non-polar amino acids was significantly increased. Discussion The last decade has witnessed tremendous progress in cancer immunotherapy, such as immune checkpoint blockade (ICB), chimeric antigen receptor T cell therapy, and TCR-T.[177]^33^,[178]^34^,[179]^35 Despite the successful application of immunotherapy across a broad range of cancers, only a minority of patients benefit from it. T cells mediate anti-tumor immune responses as targets of ICB therapy.[180]^36 Each T cell is endowed with a clonally restricted surface TCR, which is a key mediator in peptide-MHC recognition and cell activation.[181]^37^,[182]^38 The heterogeneity of T cells has made great progress. However, the similar characteristics of T cells in different cancers remain to be characterized. Therefore, a clear understanding of the co-altered biology of T cells across cancers is needed to fully exploit the therapeutic capacity of immunotherapy. In this study, we performed a systematic analysis of T cells from 15 cancer datasets. We identified T cell types in a variety of functional states that were enriched in different tissues. In particular, we observed multiple Treg subsets in CD4^+ T cells, with CD4_TNFRSF9^+ Treg representing the activated state. Tregs inhibit aberrant immune response against autoantigens and also suppress anti-tumor immune response.[183]^39 Trajectory analysis revealed the transition of these Treg clusters to the activated state across cancers. Among them, the transition from CD4_TNFRSF9^− Treg and CD4_ISG^+ Treg to the activated state has been observed in pan-cancer.[184]^18 We also observed the transition from CD4_HSPA1A^+ Treg to the activated state in multiple cancers. Overexpression of HSPA1A can inhibit the antitumor effect of histone deacetylase inhibitors.[185]^40 These results suggest a potential contribution of interferon-stimulated genes and heat shock protein genes in Treg activation, which may promote tumor immune evasion. T cell exhaustion is a state of T cell dysfunction. It prevents optimal control of malignant cells.[186]^41 In addition to previously reported T cell exhausted paths,[187]^18 we also found the state transition from CD8_TXNIP^+ T to CD8_KLRG^+ T and CD8_GZMK^+ T, and finally to CD8_Temra and CD8_Tex in all cancers. This trajectory persisted even in HCC with the smallest number of cells. Moreover, there were 14 TF regulons that significantly changed their activity in CD8_CX3CR1^+ Temra and CD8_CXCL13^+ Tex compared with CD8_TXNIP^+ T. Also, clinically relevant cancer subtypes were identified in TCGA cancers based on the expression of these TFs. Among them, ETV1 has been shown to be a drug therapeutic target for gastrointestinal stromal tumor.[188]^42 While the expression of TBX21 in tumor CD8^+ T cells was induced by immune checkpoint blocking.[189]^43 In addition, CD8_TXNIP^+ T cells were enriched in all tissues and the STARTRAC analysis suggested its ability to migrate from the periphery to the tumor and then differentiate into effector cells. These results implied that CD8_TXNIP^+ T cells may be a target for tumor immunotherapy. Analysis of cell-cell communication patterns can reveal coordinated responses between different cell types. We separately analyzed cell-cell interactions of all cell types in different tissues and revealed the rewired communications in T cells. We found that 11 pathways play important roles in mediating the tumor-infiltrating T cell communications in all cancers. These included chemokine (CCL, CXCL) and cell adhesion (CADM, VCAM) pathways associated with T cell trafficking. CXCR6 is essential for antitumor effect of CD8^+ T cells,[190]^44 and the CXCL13/CXCR5 axis plays an important role in tumor immunity.[191]^45 We observed that LT was a Treg-specific outgoing signaling pathway. Its LR pair and downstream genes were involved in the TNF signaling pathway. TNF pathway-related genes were significantly overexpressed in tumor-infiltrating T cells, and TNF pathway activity was highest in CD69^+ Trm. Studies have shown that certain types of Tregs can affect Trm function.[192]^46^,[193]^47 These results suggested that Tregs may regulate other cells by activating the TNF signaling pathway. Further studies of the TNF pathway are still needed to explore its effect on tumor immunity. In addition, the GALECTIN pathway mediated the interaction between ISG^+ T cells and CD8_TXNIP^+ T cells via LR pairs for LGALS9-HAVCR2/CD45/CD44. LGALS9 is highly expressed by induced Tregs.[194]^48 LGALS9 interacting with HAVCR2 can regulate T cell exhaustion and apoptosis,[195]^49 while LGALS9 interacting with CD44 can promote T cell activity.[196]^48^,[197]^50 This may be the potential mechanism that triggers CD8_TXNIP^+ T cells to differentiate into different cell states. Limiting the interaction between LGALS9 and HAVCR2 may prevent T cell exhaustion. Then we systematically characterized the TCR of all cancers. Tumor-infiltrating T cells had both higher TCR diversity and higher TCR clonality. CD8^+ T cells had a significantly higher percentage of clonal expansion than CD4^+ T cells, which was found in all cancers. Studies have shown that CD8^+ T cells have larger clones than CD4^+ T cells, and the size of CD4 clones was more tightly regulated.[198]^51 Further analysis revealed differences in TCR characteristics between CD8_CX3CR1^+ Temra and CD8_CXCL13^+ Tex cells. In summary, our study systematically delineates the immune ecosystem of tumor-infiltrating T cells and reveals the landscape of shared characteristics of tumor-infiltrating T cells across cancers, which will provide new insights into the underlying mechanisms of tumor immunity. Materials and methods Single-cell data collection We downloaded 15 cancer scRNA-seq datasets from the Gene Expression Omnibus (GEO) ([199]https://www.ncbi.nlm.nih.gov/geo/), which were generated from different platforms, including 10x and Smart-Seq2. Each dataset contains single-cell transcriptome count matrix and paired TCR data. A total of 521,472 cells involved 103 patients and 3 different tissue types, including tumor, adjacent normal tissue, and peripheral blood ([200]Table S1). We extracted T cells for subsequent analysis. Low-quality cells (<200 genes/cell and >10% mitochondrial genes) were excluded. Finally, a total of 349,799 T cells were obtained from 101 patients. Data integration, unsupervised clustering, and cell type annotation To subcluster T cells from all cancer data, we used Seurat (version 3.2.3) to create the gene expression matrix object. All functions were run with default parameters, unless otherwise specified. Genes shared by all datasets were retained. Then data normalization, variable feature identification, data scaling, and principal-component analysis were performed. To integrate T cells from different platforms and tissues, we used the harmony algorithm.[201]^52 Next, a KNN graph was constructed using the FindNeighbors function in Seurat. Finally, cells were clustered using the FindClusters function in Seurat. The resolution parameter was set to 1 and 1.5 for CD4^+ T cells and CD8^+ T cells, respectively. Uniform Manifold Approximation and Projection (UMAP) was used to visualize T cells in a two-dimensional space. Cluster biomarkers were identified using the FindAllMarkers procedure in Seurat, which identified differentially expressed genes for each cluster using a Wilcoxon rank-sum test. The significant differentially expressed gene was determined by the following thresholds: (1) the gene was expressed in at least 10% of the cells in one group, (2) a Bonferroni-adjusted p value lower 0.05, (3) the average log-scale fold-change between two groups greater than 0.25. We annotated the clusters as different major cell types based on the top ranked differentially expressed genes in each cluster. Trajectory analysis Monocle3 (version 1.0.0) was utilized to investigate transcriptional and functional trajectories of T cell clusters.[202]^53 Specifically, principal components were calculated from the expression matrix for each cancer. The cells were divided into large and well-separated groups using Louvain/Leiden community detection by Cluster_Cells function, and within each group a principal graph was fitted using the function Learn_Graph. The principal graph was shown as a trajectory on the UMAP, then the function order_cells was used to calculate the pseudotime. SCENIC analysis TF activity was analyzed using pySCENIC per cancer with raw count matrices as input.[203]^20 First, the grnboost2 function was used to infer gene co-expression modules and then identify the regulons enriched for the motifs of the TF. Finally, the aucell function was used to calculate the RAS. The RSS was calculated by calcRSS function in R. Subtype classification of TCGA cancer samples We classified samples into optimal clusters by referencing the expression of TF regulons using the R package “ConsensusClusterPlus.” The parameters were “maxK = 3, reps = 1000, clusterAlg = pam, distance = spearman.” Survival analysis R package TCGAbiolinks was used to obtain the gene expression of 33 cancer types. We also downloaded the clinical data by UCSC Xena ([204]https://xenabrowser.net/datapages/).[205]^54 We used univariate Cox regression analysis to evaluate the association between survival time and the expression level of each gene. Genes with hazard ratio (HR) > 1 and p < 0.5 were identified as risk factors in cancer, while genes with HR < 1 and p < 0.05 were identified as protective factors in cancer. The Kaplan-Meier method was used to estimate the overall survival time between different groups, and differences in survival time were analyzed using the log rank test (p < 0.05). Pathway enrichment analysis We performed pathway enrichment analysis for target genes of cell-type-specific TF by clusterProfiler (version 4.4.4).[206]^55 The parameters were “pvalueCutoff = 0.05, pAdjustMethod = ‘BH,’ minGSSize = 10.” Next, all the GO terms were clustered based on the implifyEnrichment R package.[207]^56 The pathway activity of TNF signaling pathway was calculated by gene set variation analysis (version 1.38.1).[208]^57 Cell-cell communication analysis We used CellChat (version 1.4.0) for cell-cell communication analysis based on known LR pairs.[209]^31 Specifically, the combined CD4 and CD8 Seurat object was subjected as input for CellChat. Then the functions computeCommunProb and computeCommunProbPathway were used to infer interactions and communication probability. The computeNetSimilarityPairwise function was used to calculate the similarity between pathways, and pathways were clustered into different groups based on functional similarities. In addition, we calculated the difference of the same pathway between different tissues, using the function rankSimilarity. The downstream TFs list of receptors were downloaded from CellCall ([210]https://github.com/ShellyCoder/cellcall).[211]^58 PPI analysis The PPI network was constructed by STRING database ([212]https://cn.string-db.org/).[213]^59 TCR diversity and clonality The clonal diversity was calculated using Shannon’s entropy: [MATH: Shannonsentropy=i =1NP(i)lnP(i) :MATH] (Equation 1) where N is the number of all clonotypes in each cancer, P(i) represents the frequency of the ith clonotype. The higher Shannon’s entropy, the more diverse the distribution of CDR3 clones. Clonality is defined based on diversity: [MATH: Clonality=1i=1NP(i)lnP(i)lnM< /mrow> :MATH] (Equation 2) where M is the number of unique TCR in each cancer. TCR similarity analysis The similarity of V-J pairs between different tissues and the similarity of TCRs between different cell clusters were calculated using Jaccard similarity coefficient: [MATH: J(A,B)=ABAB :MATH] (Equation 3) For TCR similarity, we calculated the median similarity across all cancers as the final similarity. Then Cytoscape (V.3.7.2) was used for network visualization.[214]^60 TCR clustering analysis GLIPH2 ([215]http://50.255.35.37:8080/) was used for TCR clustering.[216]^32 The input information includes CDR3b, Vb, Jb, CDR3a, clone size, and sample information of each TCR. We clustered the TCRs of all cancers together. Statistics analysis Statistical analyses were performed using R. Fisher’s exact test was used to assess differences in usage of V-J pairs (p < 0.05). The Pearson correlation coefficient was used to measure the relation of regulon activity score and TCR clonality (p < 0.05). The Wilcoxon rank-sum test was used for differentially expressed genes between tumor and other tissues (adjust p <0.05). Acknowledgments