Graphical abstract graphic file with name fx1.jpg [48]Open in a new tab Highlights * • TIL infusion product is heterogeneous and comprises CD4, CD8, and (DN) T cells * • CD3^+CD8^−CD4^−(DN) TILs restrict CD8^+ TIL expansion, correlating with worse TIL-ACT * • CD8^+ TIL expansion is restricted by (DN) TILs via Fas-FasL, TGF-β, and IL-10 signaling * • Nine TME gene signatures and 14 intracellular T cell genes hold prognostic value of TIL-ACT __________________________________________________________________ Liu et al. establish a transcriptomic and translational approach across outcomes and characteristics of tumor-infiltrating T cell (TIL) products and baseline tumor microenvironment from high-risk nasopharyngeal carcinoma patients receiving TIL-based immunotherapy. They identify the role of CD3^+CD4^−CD8^− double-negative TILs and other biomarkers in the prediction of TIL immunotherapy outcome. Introduction Adoptive cell therapy (ACT) has become a well-established treatment option for cancer patients.[49]^1 ACT using ex vivo expanded tumor-infiltrating lymphocytes (TILs), which has advantages that derive from native genetically unmodified cells and comprises polyclonal cells capable of targeting multiple tumor antigens, is becoming more and more widespread in the treatment of solid tumors.[50]^2 Despite the exciting clinical achievement of autologous TIL-ACT in solid tumors, such as melanoma[51]^3 and cervical,[52]^4 colorectal,[53]^5 and breast cancer,[54]^6 in recent years, only a minority of patients attain long-term benefits with TIL-ACT. Identifying patients who are most likely to benefit from TIL-ACT remains a key challenge. The clinical success of immunotherapy including ACT and immune checkpoint blockade (ICB) is influenced by the features of the tumor immune context, such as the presence of activated or exhausted T cells; infiltration with immune regulatory myeloid cells or other categories of immune cells; and gene expression programs that may influence the recruitment, expansion, and activity of T cells.[55]^7 In a high-dimensional analysis of ACT products in patients with metastatic melanoma, TIL-ACT responders were reported to retain a pool of stem-like neoantigen-specific TILs that are absent in ACT non-responders.[56]^8 However, the clinically validated biomarkers associated with disease progression in TIL-ACT are still unclear due to the heterogeneity of tumor microenvironments (TMEs). Nasopharyngeal carcinoma (NPC) is strongly associated with Epstein-Barr virus (EBV) infection, and the highest rates occur in South China, Southeastern Asia, and North Africa.[57]^9 A large number of lymphocytes home into NPC tissues for the novel viral antigens presented by NPC cells, including EBNA1, LMP1, and LMP2. Thus, NPC represents a good model for cancer immunotherapy.[58]^10 We previously identified the safety and objective clinical response, along with increased EBV-specific immunity, in vivo after autologous TIL infusion in a phase 1 study in locoregionally advanced NPC patients.[59]^11 However, the patients who received autologous TIL treatment following concurrent chemoradiotherapy (CCRT) did not obtain a significant survival benefit in a phase 2 clinical trial in advanced NPC patients with plasma EBV (pEBV) load ≥4,000 copies (n = 156, [60]NCT02421640).[61]^12 To address the issue of who will likely benefit from TIL-ACT among these EBV DNA-selected high-risk advanced NPC patients, we conducted a retrospective analysis of a cohort from this phase 2 clinical study ([62]NCT02421640) to screen the variants and biomarkers that predict clinical outcome after treatment with TIL infusion products. We also investigated the underlying molecular mechanisms that influence clinical outcomes. Results T cell diversity and outcome relevance of TIL infusion products To evaluate the associations of TME features and TIL infusion product compositions with the clinical outcomes of TIL-ACT, as well as the underlying molecular mechanisms influencing TIL-ACT outcomes, we utilized tumor tissue biospecimens and TIL infusion products procured from NPC patients who received adjuvant autologous TIL therapy after CCRT in a phase 2 clinical trial study ([63]NCT02421640; [64]Table S1). The methodology is outlined in [65]Figure 1A. Sixty-two tumor samples from patients treated with TIL plus CCRT (n = 22) or CCRT alone (n = 40) were analyzed by bulk RNA sequencing (RNA-seq), and 47 TIL infusion products from patients treated with TIL plus CCRT were analyzed by flow cytometry. In addition, 26 of 47 TIL infusion products were evaluated by single-cell (sc)RNA-seq. After quality filtering, we obtained 65,978 high-quality T cells and identified 14 T cell clusters with distinct G1 phase, S phase, and G2M phase ([66]Figures 1B, [67]S1A, and S1B). As expected, CD3 expression was distributed across all the 14 clusters ([68]Figure S1C). Based on the expression of canonical marker genes, 11 clusters were assigned to CD3^+CD8^+ T cells (89.8% of total cells), including C0_activated_CD8_T_cells, C1_cytotoxic_CD8_T_cells, C2_proliferating_CD8_T_cells-PCNA, C3_proliferating_CD8_T_cells-LTB, C4_CD8_MHC_II_T_cells, C5_proliferating_CD8_T_cells-TNFRSF8, C6_proliferating_CD8_T_cells-TOP2A, C9_effector_memory_CD8_T_cells, C10_proliferating_CD8_T_cells-LGALS1, C12_tissue_resident_memory_CD8_T_cells, and C13_stem_like_CD8_T_cells, and the CD3^+CD4^+ T cells (6.3% of total cells) were separated into two clusters, including C7_naive_CD4_T_cells and C11_Th2_CD4_T_cells ([69]Figures 1C and [70]S1D). In addition, the CD3^+CD4^−CD8^− double-negative (DN) T cells (3.9% of total cells) were a separate cluster: C8_DN_T_cells, referred to as (DN) TILs ([71]Figure 1D). Similar percentages of the CD3^+CD8^+, CD3^+CD4^+, and CD3^+CD4^−CD8^− T cell subsets were validated by flow cytometry in 47 TIL samples ([72]Figure 1E). The (DN) TIL frequency in these 14 T cell clusters was significantly lower in patients with non-progression than in those with progression ([73]Figures 1F and 1G). A lower frequency of (DN) TILs correlated with favorable progression-free survival (PFS) and overall survival (OS) in patients undergoing TIL-ACT ([74]Figures 1H, [75]S1E, and S1F). Figure 1. [76]Figure 1 [77]Open in a new tab Study design and single-cell transcriptomic landscape of TIL infusion products (A) Graphical overview of the experimental design and bioinformatics workflow. In our clinical trial cohort, bulk RNA sequencing was performed on tumor tissue from NPC patients before CCRT+TIL (n = 22) or CCRT alone (n = 40), and TIL infusion products were isolated from pretreatment tumor tissue from these patients, expanded ex vivo, and subsequently prepared for single-cell (sc)RNA sequencing (n = 26) and flow cytometry (n = 47). (B) Uniform manifold approximation projection (UMAP) plot of 65,978 quality-controlled T cells colored by 14 T cell subsets with the distinct G1 phase, S phase, and G2M phase. The cell-cycle phase of 14 T cell subsets was labeled in the UMAP plot and determined by the cell-cycle phase scores calculated by the Seurat package. (C) Heatmap showing the expression of the top 50 differentially expressed genes among cell subsets obtained by cell type annotation utilizing the expression of canonical marker genes in 14 cell subgroups. Information on the 14 cell subsets is displayed on the right. (D) UMAP plots showing the distribution of indicative T cell subsets between the non-progression group and progression group. (E) Dot plot showing the composition of CD3^+CD4^−CD8^−, CD3^+CD4^+, and CD3^+CD8^+ T cells in NPC TIL infusion products according to flow cytometry (n = 47). Scatter dot plots show individual values and mean ± SEM. (F) Bar plot comparing the frequency of 14 cell subgroups in the TIL infusion products from NPC patients in the non-progression (n = 15) and progression groups (n = 11). The data are presented as mean ± SEM. The Wilcoxon rank-sum test was used to determine the significance. ∗p < 0.05; ns, not significant; adjusted for multiple comparisons using the Benjamini-Hochberg procedure. (G) Boxplot comparing the frequency of (DN) TILs in the non-progression (n = 28) and progression groups (n = 19) according to flow cytometry. Median and interquartile were shown in the boxplots. Wilcoxon rank-sum test was used to determine the significance. (H) Kaplan-Meier survival curves for progression-free survival (PFS) and overall survival (OS) in NPC patients stratified according to the abundance (high abundance vs. low abundance) of the C8_DN_ T_cells cluster inferred from (sc)RNA sequencing data (n = 26). p values were determined based on the two-sided log rank test. Optimal cutoff values were defined using the “survminer” R package. See also [78]Figure S1 and [79]Table S1. Transcriptome characterization, origin, and composition of (DN) TILs in NPC The frequency of peripheral CD3^+CD8^−CD4^− (DN) T cells from NPC patients was notably increased relative to healthy donors ([80]Figures S2A and S2B). The transcriptome profile showed that (DN) TILs expressed high levels of genes encoding innate lymphocyte markers, such as FCGR3A, NCAM1, and NKG7; molecules characteristic of regulatory T cells (Tregs), including IKZF2, TNFRSF18, IL2RA, STAT5A, and ICOS, but not FOXP3 and CTLA4; suppressive and cytotoxic cytokines, such as interleukin (IL)-10, TGFB3, IFNG, and tumor necrosis factor (TNF); and activated (DN) T cell markers, such as CD69, XCL1, CD160, LTA, and CD44, compared with CD4^+ and CD8^+ TILs ([81]Figure 2A). In addition, Treg differentiation-related signaling pathways, such as transforming growth factor β (TGF-β) and IL-10, and the Treg gene signature were up-regulated in the (DN) TIL cluster ([82]Figures 2B and 2C). Flow cytometric analysis validated that the (DN) TILs from NPC patients exhibited high levels of IL-10, TGF-β, and IKZF2, while showing an absence of FOXP3 and CTLA4 expression compared with CD4^+ and CD8^+ TILs ([83]Figures 2D and [84]S2C). Furthermore, we confirmed that the trajectory from peripheral to tumor-infiltrating (DN) T cells contained two distinct branches, including Cell Fate #1 (CF1) and Cell Fate #2 (CF2), in treatment-naive NPC patients (GEO: [85]GSE162025 ; n = 10, [86]Figure 2E). The CF1 (DN) T cells were mainly found in tumor tissues and exhibited increased expression of genes encoding characteristic molecules of activated Tregs, such as CD69, IKZF2, TNFRSF18, and IL-10, along with an increase in the Treg gene signature compared to CF2 (DN) T cells, which were mainly located in peripheral blood and peripheral (DN) T cells ([87]Figure S3). We observed a close resemblance between the characteristics of the CF1 cluster and the (DN) TIL gene signature in the baseline TMEs ([88]Figure 2F). Accordingly, we observed that the expression of IL-10, TGF-β, and IKZF2 was increased in (DN) TILs compared to peripheral (DN) T cells from NPC patients or healthy donors, but not the expression of FOXP3 and CTLA4 ([89]Figures 2G and [90]S2D). These data suggest that (DN) TILs exhibit an activated IL-10^+TGF-β^+IKZF2^+FOXP3^−CTLA4^−Treg-like phenotype in the pretreatment TME. Figure 2. [91]Figure 2 [92]Open in a new tab Transcriptional characterization and developmental trajectory of CD3^+CD8^−CD4^− (DN) TILs (A) Heatmap showing the expression levels of genes encoding NK cell markers, Treg-related molecules, immune checkpoint proteins, cytokines, and (DN) T cell-related markers among CD8^+, CD4^+, and (DN) TIL clusters. (B) Gene set enrichment analysis (GSEA) showing significant upregulation of TGF-β (left) and IL-10 (right) signaling pathways in (DN) TILs compared with other TIL subsets. (C) Violin plot showing the expression of a regulatory T cell signature across (DN) TIL, CD4^+ TIL, and CD8^+ TIL clusters. A dashed line indicates the median of the signature score of the (DN) TIL cluster. Median and interquartile were shown in the boxplots. Non-parametric Kruskal-Wallis test was used to determine the significance. (D) Representative overlap histogram for the expression of IL-10, TGF-β, IKZF2, FOXP3, and CTLA4 among (DN) TILs, CD4^+ TILs, and CD8^+ TILs determined by flow cytometry. (E) Developmental trajectory of 16,978 (DN) T cells from tumor tissues and peripheral blood of naive treated NPC patients (n = 10) inferred by Monocle 2 and CytoTRACE algorithm. Solid and dotted lines denote distinct cell fates based on expression profiles, with colors indicating the origin of (DN) T cells, pseudo-time, and CytoTRACE score (GEO: [93]GSE162025 ). (F) Dot plot showing the correlation between (DN) TIL signature scores of TMEs in this study and each state of (DN) T cells (GEO: [94]GSE162025 ; PB, Cell Fate_1, and Cell Fate_2). The sizes and colors of the circles represent the strength of the relationship, assessed using Spearman’s correlation test. PB, peripheral blood. (G) Representative overlap histogram for the expression of IL-10, TGF-β, IKZF2, FOXP3, and CTLA4 in the (DN) T cells from TIL infusion products, peripheral blood of NPC patients, or healthy donors determined by flow cytometry. See also [95]Figures S2 and [96]S3. Subsequently, the 2,609 (DN) TILs (C8 cluster) from 26 TIL infusion products could be clustered into six subsets ([97]Figure 3A). NCAM1 encoding CD56 and FCGR3A encoding CD16 were expressed in two of the six subsets ([98]Figure 3B). Some previous studies had found that (DN) T cells could originate from the thymic T cell DN3 stage, double-positive (DP) stage, or single-positive (SP) stage.[99]^13^,[100]^14 However, we found that the expression of genes encoding T cell receptor (TCR) alpha-beta (αβ) and TCR gamma-delta (γδ) were distributed in all six (DN) TIL subsets, with the TCRαβ^+ T cells comprising 76% of the total (DN) TILs, suggesting that these (DN) TILs may mainly originate from T cells after the DP stage ([101]Figures S4A and S4B). To further investigate the origin of (DN) TILs, we analyzed the scRNA-seq and TCR sequencing (TCR-seq) datasets of three paired TIL infusion products and peripheral blood samples from NPC patients treated with CCRT plus TIL-ACT. We found that TCRαβ-positive cells were distributed within the CD3^+ T cell population, including CD4^+, CD8^+, and (DN) T cells, in both the TIL infusion products and peripheral blood ([102]Figures S4C and S4D). Further analysis revealed that 60.2% of TCRαβ-positive (DN) TILs shared common TCRs (10 shared clones) with 57.5% of CD8^+ TILs, and 63.4% of TCRαβ-positive (DN) TILs shared common TCRs (12 shared clones) with 49.5% of CD4^+ TILs. Among these shared TCR clones, 8 were shared by CD4^+, CD8^+, and (DN) TILs, whereas 33.4% of TCRαβ-positive (DN) TILs exhibited unique TCR clones ([103]Figure S4E). In contrast, only 31.3% of TCRαβ-positive (DN) T cells shared common TCRs (31 shared clones) with 16.6% of CD8^+ T cells, and 1.6% shared common TCRs (2 shared clones) with 0.3% of CD4^+ T cells. Among these shared TCR clones, only 1 was shared by CD4^+, CD8^+, and (DN) T cells together, whereas 68.2% of TCRαβ-positive (DN) T cells exhibited unique TCR clones ([104]Figure S4F). We also observed that 20.4% of TCRαβ-positive (DN) TILs shared common TCRs (2 shared clones) with T cells from peripheral blood ([105]Figure S4G). These data suggest that most TCRαβ-positive (DN) TILs originate from CD4^+ and CD8^+ T cells (DP or SP stage T cells) in the TME, with only a small fraction recruiting peripheral (DN) T cells. Further comparing the transcriptional features of the CD56^− and CD56^+ (DN) TIL clusters, we found that innate lymphocyte markers, such as NCAM1, and several genes encoding Treg-like characteristics, such as IKZF2, TNFRSF18, TGFB3, and PRF1, were highly expressed in CD56^+ (DN) TILs. Most genes encoding activated suppressive Treg-like characteristics, such as IL2RA, ICOS, TIGITIL10, IFNG, CD69, XCL1, and CD44, were highly expressed in CD56^− (DN) TILs ([106]Figures 3C–3E). IKZF2 is a key factor in maintaining the suppressive function and stability of Tregs and is highly expressed in (DN) T cells,[107]^15^,[108]^16 both CD56^− (DN) TILs and CD56^+ (DN) TILs, and the activated DN Treg signature was the highest in CD56^− IKZF2^+ (DN) TILs compared with other (DN) TIL subsets ([109]Figures 3B and 3F). A higher frequency of CD56^− IKZF2^+ (DN) TILs was significantly associated with poor PFS and OS in NPC patients treated with CCRT plus TIL ([110]Figure 3G). Figure 3. [111]Figure 3 [112]Open in a new tab Transcriptomic characterization and association of CD56^− (DN) TILs with clinical outcome (A) UMAP plot depicting 2,609 quality-controlled (DN) TILs colored by six cell subsets. (B) UMAP plot of (DN) TILs, with cells colored based on the relative normalized expression of NCAM1 (encoding CD56), FCGR3A (encoding CD16), and IKZF2. (C) Volcano plot showcasing differentially expressed genes (DEGs) between CD56^− (DN) TILs and CD56^+ (DN) TILs. Red dots and blue dots indicate up-regulated genes (n = 770) and down-regulated genes (n = 316) in CD56^− (DN) TILs, respectively. Selected genes were labeled. (D) Heatmap showing the expression of genes encoding TCR proteins, NK cell markers, immune checkpoint proteins, cytokines, and (DN) T cell-related markers among CD56^− and CD56^+ (DN) TIL clusters. (E) GSEA plots showing that Treg signaling pathways were significantly up-regulated in CD56^− (DN) TILs compared with CD56^+ (DN) TILs. (F) Violin plot showing the expression of an activated DN Treg signature across four (DN) TIL clusters (CD56^−IKZF2^+, CD56^−IKZF2^-, CD56^+IKZF2^-, and CD56^+IKZF2^+ (DN) TILs). A dashed line indicates the median of the signature score of the CD56^−IKZF2^+ (DN) TIL. Median and interquartile were shown in the boxplots. Kruskal-Wallis test was used to determine the significance. (G) Kaplan-Meier survival curves for PFS (top) and OS (bottom) in NPC patients stratified according to the abundance (high abundance vs. low abundance) of CD56^−IKZF2^+ (DN) TILs inferred from (sc)RNA sequencing data (n = 26). p values were determined based on the two-sided log rank test. See also [113]Figure S4. Relationship of CD8^+ TIL subsets with (DN) TILs and TIL-ACT outcomes Cell-cell communication analysis revealed that (DN) TILs interact with CD4^+ and CD8^+ TIL subsets through the TGF-β, IL-10, and Fas/Fas ligand (FasL) signaling pathways ([114]Figure 4A). We further determined that the (DN) TILs from NPC patients could suppress the proliferation of naive CD4^+ and CD8^+ T cells, and the suppression of (DN) TILs was largely diminished by the administration of anti-IL-10 and anti-TGF-β antibodies, as well as Fas-FasL inhibitor in vitro. Notably, the (DN) TILs exhibited suppressive ability toward naive CD4^+ and CD8^+ T cells comparable to that of conventional CD4^+ induced (i)Tregs ([115]Figures 4B, 4C, [116]S5A, and S5B). However, a significant negative association was found between the frequency of (DN) TILs and C2_proliferating_CD8_T_cells-PCNA in the Spearman correlation analysis ([117]Figure 4D). Importantly, the frequency of (DN) TILs was significantly negatively associated with the total CD8^+ TIL density in the TIL infusion products ([118]Figure 4E), suggesting that a high frequency of (DN) TILs will limit the expansion of CD8^+ TILs in TIL infusion products. Moreover, we demonstrated that the (DN) TILs from NPC patients could suppress the proliferation of autologous ex vivo expanded CD8^+ TILs ([119]Figure 4F). In addition, the TIL infusion products exhibited cytotoxicity against human leukocyte antigen (HLA)-matched EBV^+ TW03 cells in a dose-dependent manner, but not the HLA-mismatched cytokine-induced killer (CIK) cells. However, the cytotoxicity of TIL infusion products containing a higher percentage of (DN) TILs was lower than that of TIL infusion products containing a lower percentage of (DN) TILs ([120]Figures S5C and S5D). Notably, the depletion of (DN) T cells from these TIL infusion products significantly enhanced their cytotoxicity against EBV^+ TW03 cells ([121]Figure S5E). These data suggest that (DN) TILs restrict CD8^+ TIL expansion and anti-tumor activity. Figure 4. [122]Figure 4 [123]Open in a new tab Associations of CD8^+ TIL subsets with (DN) TILs and clinical outcome (A) Dot color represents the communication probability of the specific ligand-receptor pairs, including TGF-β, IL-10, and Fas-FasL signaling between the indicated sender cluster and receiver clusters. (B) Representative histograms and statistical graph showing carboxyfluorescein succinimidyl ester (CFSE) dilution (left) and proliferation inhibition rates (right) of (DN) TILs with CD4^+ and CD8^+ naive T cells at a ratio of 4:1, 2:1, 1:1, and 1:2. The data are presented as mean ± SD, and three biological replicates were included. (C) Representative histograms and statistical graph showing CFSE dilution (left) and proliferation inhibition rates (right) of (DN) TILs with CD4^+ and CD8^+ naive T cells at a 1:1 ratio with neutralizing antibodies against TGF-β and IL-10, as well as the presence or absence of a Fas-FasL signaling antagonist. The data are presented as mean ± SD, and three biological replicates were included. p values were evaluated by t test (two-sided). ∗∗p < 0.01; ∗∗∗p < 0.001. (D) Bubble plot showing the correlation between the frequency of (DN) TILs and other TIL subsets in TIL infusion products (n = 26). p values were determined by Spearman correlation analysis. ∗p < 0.05. (E) Dot plot showing the correlation between the frequency of (DN) TILs and CD8^+ TILs in TIL infusion products (n = 26). p values were determined by Spearman correlation analysis. (F) Representative histograms and statistical graph showing CFSE dilution (left) and proliferation inhibition rates (right) of (DN) TILs with autologous expanded CD8^+ TILs at ratios of 4:1, 2:1, and 1:1. Conventional CD4^+ (i)Tregs were included as a control. The data are presented as mean ± SD, and three biological replicates were included. (G–K) Kaplan-Meier survival curves of PFS and OS in NPC patients stratified according to the abundance (high vs. low) of total CD8^+ TILs (G), C2_proliferation_CD8_T_cells-PCNA (H), C4_MHC_II_CD8_T_cells (I), C6_proliferation_CD8_T_cells-TOP2A (J), and C12_tissue_resident_memory_CD8_T_cells (K) inferred from (sc)RNA sequencing data (n = 26). p values were determined based on the two-sided log rank test. See also [124]Figures S5 and [125]S6. In CD8^+ TIL clusters, the frequencies of C2_proliferating_CD8_T_cells-PCNA, C4_MHC_II_CD8_T_cells, C6_proliferating_CD8-T_cells-TOP2A, and total CD8^+ TILs correlated with improved PFS and OS, and the frequencies of C3_proliferating_CD8_T_cells-LTB and C5_prolifearting_CD8_T_cells-TNFRSF8 were related to better PFS. In contrast, the frequencies of C1_cytotoxic_CD8_T_cells, C10_proliferating_CD8_T_cells-LGALS1, and C12_Tissue_resident_CD8_T_cells were linked to worse prognosis, and the frequencies of C0_activated_CD8_T_cells, C9_effector_memory_CD8_T_cells, and C13_stem_like_CD8_T_cells were not associated with prognosis in NPC patients treated with CCRT plus TILs ([126]Figures 4G–4K and [127]S6A). Notably, the CD8^+ TIL subsets (C1, C10, and C12) related to poor prognosis had lower expression of proliferation-related genes, such as TNFRSF8 (encoding CD30) and PCNA, as well as proliferation gene signatures, but had higher expression of exhaustion-related genes, such as TIGIT, as well as dysfunction gene signatures, compared to the CD8^+ TIL clusters (C2, C3, C4, C5, and C6) being related to favorable prognosis, indicating that these CD8^+ TIL clusters related to worse prognosis were the exhausted TILs ([128]Figures S6B and S6C). Furthermore, we demonstrated that the cytotoxicity of CD8^+ TILs against TW03 cells was significantly reduced following the administration of a neutralizing antibody targeting CD137, a marker of activated cytotoxic T cells[129]^17 and CD30 in vitro. CD137 was highly expressed in the C4_MHC_II_CD8_T_cells and C5_Proliferating_CD8_T_cells_TNFRSF8 clusters ([130]Figures S6D and S6E). These data suggest that distinct CD8^+ TIL subsets exhibit different impacts on TIL-ACT outcome. Associations of baseline TME transcriptome features with TIL-ACT outcomes and the composition of TIL infusion products Gene expression profiling analysis using gene signatures representing both immune cell and tumor characteristics[131]^18^,[132]^19^,[133]^20 ([134]Table S2) was performed in 62 pretreatment NPC tissues from 22 patients treated with CCRT plus TIL-ACT and 40 patients treated with CCRT alone ([135]Figure 5A). Cox regression analysis showed that the expression of natural killer (NK) cell, major histocompatibility complex (MHC) class II, and mismatch repair gene signatures was associated with PFS in the TIL+CCRT arm but not in the CCRT-alone arm ([136]Figure 5B). Kaplan-Meier survival analyses demonstrated that the patients with high levels of NK cell, MHC class II, CD8^+ cytotoxic T cell, CD39^−CD69^− stem-like T cell, NeoTCR8, B cell receptor (BCR), LAMP3^+ dendritic cell (DC), and interferon receptor signatures had longer PFS after CCRT plus TIL-ACT but not CCRT alone, whereas the patients with a high mismatch repair signature, which is indicative of malignant cells,[137]^21 had shorter PFS for TIL-ACT plus CCRT but not for CCRT alone ([138]Figures 5C and [139]S7). Figure 5. [140]Figure 5 [141]Open in a new tab Relationship between transcriptome features in the baseline tumor microenvironment and clinical outcome and TIL subset composition (A) Heatmap showing the expression of the indicative gene signatures of the baseline TME in 62 NPC patients. (B) Bubble plots showing the Cox regression analysis of gene expression signatures in patients treated with CCRT+TIL-ACT or CCRT alone. The p values and hazard ratios were estimated from a stratified Cox model with the low-gene-signature-score group as the reference group. (C) Kaplan-Meier survival curves for PFS in NPC patients treated with CCRT+TIL-ACT (n = 22) stratified according to the indicative gene signature expression (high score vs. low score) of NK cell, MHC class II, CD8^+ cytotoxic T cell, CD39^−CD69^− stem-like T cell, NeoTCR8, BCR, LAMP3^+ DC, interferon receptor, and mismatch repair signatures. (D) Heatmap showing the correlation between gene signature scores and the frequency of 14 TIL subsets in TIL infusion products, with significance assessed by the Spearman correlation. ∗p < 0.05, ∗∗p < 0.01. See also [142]Figure S7 and [143]Table S2. Notably, the TCR signaling, T cell, macrophage, cytotoxic activity, and CD8^+ cytotoxic T cell signatures were positively associated with the frequency of the C6 subset, which related to better TIL-ACT outcomes, whereas the LAMP3^+ DC and co-activation molecules signatures negatively correlated with the C10 cluster, which related to poor TIL-ACT outcome. Notably, the frequency of (DN) TILs, which correlated with poor TIL-ACT outcome, was positively associated with the tumor-associated macrophage (TAM) signature but negatively correlated with the LAMP3^+ DC and interferon receptor signatures ([144]Figure 5D). These data suggest that the transcriptome profile of the baseline TME influences the clinical outcomes of TIL-ACT and the composition of TIL subsets in TIL infusion products. Identification of 14 intracellular T cell genes with prognostic value for TIL-ACT To identify predictive molecular biomarkers of clinical efficacy, as well as the potential therapeutic optimization targets for TIL-ACT in NPC patients, we identified a total of 414 differentially expressed genes (DEGs), comprising 205 up-regulated genes and 209 down-regulated genes, in TIL infusion products from non-progression versus progression patients ([145]Figure S8A). Among the 205 up-regulated DEGs, 13 genes, namely HHLA2, SPOCK1, EPDR1, RHOU, HYAL3, CD86, HLA-DOA, TBXAS1, CLU, SCD, CCDC34, FAM111B, and CYB561, were significantly associated with favorable PFS and OS in the CCRT+TIL-ACT arm, as well as the 13-gene signature ([146]Figures 6A, [147]S8B, and S8C). We determined that these 13 genes are over-represented in the pathways related to T cell activation, T cell co-stimulation, and leukocyte cell-cell adhesion ([148]Figure 6B). Among the TIL subsets, these 13 prognostic genes were highly expressed in CD8^+ TIL clusters, including C2, C3, C5, C6, C10, and C4 ([149]Figure 6C). Interestingly, among 209 down-regulated DEGs in the non-progression group, only TNFRSF18, encoding checkpoint protein, which was highly expressed in the (DN) TIL cluster and enhanced proliferative suppression mediated by the (DN) TIL, was associated with poor OS in patients treated with CCRT plus TIL-ACT ([150]Figures 6D, 6E, [151]S8D, and S8E). These data indicate that these 14 prognostic-related intracellular T cell genes may serve as potential targets to remold the anti-tumor immunity of TIL infusion products and optimize TIL-ACT. Figure 6. [152]Figure 6 [153]Open in a new tab Prognostic value analysis of 14 differentially expressed genes in TIL infusion products from non-progression and progression groups (A) Euler diagrams represent the overlapping genes enriched in the non-progression group and associated with PFS and OS. Thirteen overlapped genes were associated with non-progression, PFS, and OS. The right diagram represents 13 overlapped genes significantly associated with favorable PFS and OS. The colors of dots represent the influence of the gene on prognosis: red indicates a protective factor and green indicates a risk factor. (B) Pathway enrichment analysis of the 13 overlapped genes. The top 8 significant pathways are shown. (C) Dot plot showing the expression of 13 overlapped genes for each T cell subset in NPC TIL infusion products. For each gene, dot colors represent the average expression in each cell type (scaled and log-normalized), whereas size reflects the percentage of cells with detectable expression in each TIL subset. (D) Dot plot showing the expression of TNFRSF18 for each T cell subset in NPC TIL infusion products. (E) Kaplan-Meier survival curves for PFS (top) and OS (bottom) in NPC patients stratified according to the expression of TNFRSF18 based on a median split inferred from (sc)RNA sequencing data (n = 26). p values were determined based on the two-sided log rank test. See also [154]Figure S8. Discussion In 2024, the US Food and Drug Administration approved the usage of the first TIL drug, lifileucel, in advanced melanoma, supporting the potential for more widespread application of TIL therapy in the future.[155]^22 However, only a minority of patients obtain a survival benefit from TIL-ACT in NPC. Thus, it is crucial to know how to choose advanced NPC patients who are likely to obtain a clinical benefit from TIL-ACT. In this study, using single-cell and bulk RNA sequencing and flow cytometric analysis, we identified that the frequencies of CD3^+CD8^−CD4^− (DN) TILs and CD8^+ T cell clusters in TIL infusion products are associated with TIL-ACT outcome. Mechanically, the (DN) TILs exhibited a DN Treg phenotype and restricted CD8^+ TIL expansion through Fas-FasL, TGF-β, and IL-10 signaling. In addition, the transcriptome of the pretreatment TME could serve as a predictor of TIL-ACT outcome and was associated with the composition of TIL infusion products in these NPC patients. We identified that the CD8^+ TILs comprised 11 of 14 T cell clusters (89.8% of total cells) in 65,978 T cells from 26 TIL infusion products, suggesting that the CD8^+ T cell subset is the predominant constituent of TIL infusion products. However, only the frequency of the (DN) TIL cluster was significantly different in non-progression versus progression patients in the CCRT+TIL-ACT arm. Patients receiving TIL-ACT with a low frequency of (DN) TILs or high CD8^+ TILs in TIL infusion products had favorable PFS and OS and a longer restricted mean survival time (RMST) for PFS and OS than patients receiving CCRT alone ([156]Figure S1F), suggesting that the (DN) TILs hinder the clinical efficiency of TIL therapy, but the CD8^+ TILs benefit the clinical efficiency of TIL therapy. Some researchers have reported the existence of a CD3^+CD8^−CD4^− (DN) T cell subset in ex vivo-expanded TIL infusion products in clinical trials for autologous TIL-ACT in other solid cancers, but the association between the (DN) TIL proportion and clinical response of TIL-ACT, as well as the function of these (DN) TILs, was not mentioned in these studies.[157]^23^,[158]^24 CD3^+CD8^−CD4^− (DN) T cells comprise one of the least studied T lymphocyte subgroups and express TCRαβ or TCRγδ but lack CD4 and CD8 coreceptors.[159]^13 In previous reports, (DN) T cells exhibited dual functions under disease conditions; some researchers think that the peripheral (DN) T cells function as cytotoxic cells and use autologous ex vivo-expanded peripheral (DN) T cells to treat cancer patients.[160]^25^,[161]^26 Conversely, some researchers have found that peripheral (DN) T cells with a Treg-like phenotype can suppress T cell proliferation and send the death signal to CD4^+ T cells and CD8^+ T cells with the same TCR specificity, preventing autoimmune disease.[162]^27^,[163]^28 Here, we found an increased proportion of (DN) T cells in peripheral blood from NPC patients compared to healthy donors ([164]Figures S2A and S2B). However, peripheral (DN) T cells did not exhibit a Treg-like phenotype ([165]Figures S2C, S2D, and [166]S3) and were not associated with the outcomes of NPC patients treated with either CCRT plus TIL-ACT or CCRT alone ([167]Figure S9). The transcriptome profile of the C8_DN_TIL cluster displayed an activated Treg-like phenotype: up-regulated genes encoding immune activation and characteristic Treg-like molecules, such as CD69, IKZF2, TNFRSF18, XCL1, FASLG, and IL-10, as well as Treg differentiation-related signaling pathways, such as TGF-β and IL-10 signaling. The (DN) TILs had an IL-10^+TGF-β^+IKZF2^+FOXP3^−CTLA4^− DN Treg phenotype, which is consistent with other reports of the peripheral (DN) T cell phenotype in autoimmune disease.[168]^29^,[169]^30^,[170]^31 Importantly, we further identified that the peripheral (DN) T cells could differentiate into activated Treg-like cells in tumor tissues from pretreated NPC patients (GEO: [171]GSE162025 , [172]Figure S3). Accordingly, the tumor-infiltrated (DN) T cells have been identified in the pretreatment TMEs of several solid tumors, but the function of (DN) T cells in tumor tissue, including pro-tumor or anti-tumor effects, is dependent on the specific type of tumor.[173]^32^,[174]^33 In the present study, we further demonstrated that the (DN) TIL cluster comprised six clusters and expressed both of TCRαβ (76% of total cells) and TCRγδ. In addition, most TCRαβ-positive (DN) TILs shared TCR clones with CD8^+ and CD4^+ TILs, but a small subset of TCRαβ-positive (DN) TILs had unique TCR clones or shared clones with peripheral T cells ([175]Figure S4). These results suggest that most of the (DN) TILs were induced from CD8^+ and CD4^+ T cells in the TMEs of NPC patients. CD56 was highly expressed in two of these (DN) TIL subsets, and most of the activated Treg-related genes were highly expressed in CD56^− (DN) TILs, but IKZF2 was highly expressed in both CD56^− and CD56^+ (DN) TILs. The CD56^−IKZF2^+ (DN) TILs had predominantly activated suppressive Treg characteristics in (DN) TILs and were significantly associated with poor TIL-ACT outcome. The cell-cell interaction analysis and in vitro data demonstrated that the suppressive effect of (DN) TILs on allogeneic naive CD4^+ and CD8^+ T cells, as well as autologous CD8^+ effector T cells, was mediated through the Fas-FasL, IL-10, and TGF-β pathways. In addition, the (DN) TILs not only restricted CD8^+ TIL number but also suppressed the cytotoxicity of CD8^+ TILs against NPC cells ([176]Figures S5C–S5E). Most researchers think that the high proportion of CD8^+ TILs in the infusion product correlates with improved clinical response of TIL-ACT, but the CD8^+ TILs represent a heterogeneous cell population comprising tumor-specific and bystander T cells in varying differentiation status, including stem-like, effector, exhaustion, and senescence status; among them, the CD8^+CD39^−CD69^− stem-like neoantigen-specific TILs have been shown to correlate with improved clinical response.[177]^8^,[178]^34^,[179]^35^,[180]^36 Here, we observed that the frequency of total CD8^+ TILs and two proliferating CD8 TIL clusters (C2 and C6) and C4_MHC_II_CD8_T_cells with high proliferation ability and low dysfunction scores were associated with better survival, whereas the C1_cytotoxic_CD8_T_cells, C10_proliferating_CD8_T_cells-LGALS1, and C12_tissue_resident_memory_CD8_T_cells clusters with low proliferation ability and high dysfunction scores were associated with worse prognosis in NPC patients treated with CCRT plus TIL-ACT. In addition, three CD8^+ TIL clusters (C0_activated_CD8_T_cells, C9_effector_memory_CD8_T_cells, and C13_stem_like_CD8_T_cells) were not associated with the clinical outcomes of NPC patients treated with CCRT plus TIL-ACT ([181]Figure S6A). Taken together, these findings suggest that TILs with a stem-like phenotype are effective mediators of tumor control in NPC. The TME is a complex, heterogeneous cellular environment composed of various cell types and plays a significant role in clinical outcomes and the response to therapy.[182]^37 Recently, several (sc)RNA sequencing studies of pretreatment NPC tissues revealed that multiplex factors, such as CD70^+ malignant cells, LAMP3^+ DCs, and Tregs, alter the T cell fate, resulting in NPC immune escape.[183]^38^,[184]^39^,[185]^40^,[186]^41 In this study, we found that nine gene signatures in the pretreatment TME are associated with outcome in patients treated with CCRT plus TIL-ACT but not CCRT alone ([187]Figure S7). Among these signatures, the high levels of NK cell, MHC class II, CD8^+ cytotoxic T cell, CD39^−CD69^− stem-like T cell, NeoTCR8, BCR, LAMP3^+ DC, and interferon receptor signatures showed a survival benefit for TIL-ACT but not CCRT alone, whereas the mismatch repair signature, a malignant cell signature,[188]^21 was associated with worse survival after TIL-ACT but not CCRT alone. Associations were found between gene signature expression in the baseline TME and the composition of TIL infusion products. For example, TCR signaling, T cell, macrophage, cytotoxic activity, and CD8^+ cytotoxic T cell signatures were positively associated with the frequency of the C6_proliferacting_CD8_T_cells-TOP2A subset, which was related to better survival. In contrast, the LAMP3^+ DC and co-activation molecules signatures negatively correlated with the frequency of the C10_proliferating_CD8_T_cells-LGALS1 subset, which was related to poor survival. Notably, the frequency of the C8_DN_T_cells subset was positively associated with the TAM signature but negatively correlated with the LAMP3^+ DC and interferon receptor signatures. We further demonstrated that the LAMP3^+ DC cluster could interact with the (DN) TIL cluster through the binding of LGALS9 to its ligands CD44, CD45, or HAVCR2, a pathway associated with T cell exhaustion and death,[189]^42 as observed in the pretreatment TME of NPC patients (GEO: [190]GSE162025 ) ([191]Figure S10A), indicating that the LAMP3^+ DC cluster may promote exhaustion or apoptosis of (DN) TILs. In addition, the (sc)TCR-seq showed a substantial clonal overlap between CD4^+ and CD8^+ T cells and (DN) T cells ([192]Figure S4), and we found that EBV^+ NPC cells could induce the expansion of (DN) T cells in vitro ([193]Figures S10B and S10C). These findings suggest that multiple factors in the TME, including suppressive conditions, tumor cells, and EBV antigens, may promote the expansion of (DN) TILs in NPC. The tumor antigen-specific conventional CD4^+ Tregs induced in TME and the antigen-specific suppression have been identified in cancers,[194]^43^,[195]^44 and Oliveira et al. has identified that HLA class II tumor-associated antigens could indirectly and directly induce the engagement of immunosuppressive CD4^+ Tregs in melanoma.[196]^45 However, the underlying mechanisms driving (DN) TIL expansion in NPC require further investigation. Recently, neoantigen load and the transcriptional and epigenetic states in the TME were reported to correlate with treatment outcomes in a cohort of patients with metastatic melanoma receiving TIL therapy.[197]^46^,[198]^47 Our study showed that the expression of 14 intracellular T cell genes in TIL infusion products was linked to TIL-ACT outcome through overlap analysis. Among them, 13 up-regulated genes in TIL infusion products from patients with non-progression, including RHOU, HYAL3, HHLA2, CD86, SPOCK1, HLA-DOA, EPDR1, TBXAS1, CLU, SCD, CCDC34, FAM111B, and CYB561, predicted improved PFS and OS after TIL-ACT, and combining these 13 genes into a signature (13-gene signature), the RMST of PFS and OS in the high-gene-signature group receiving CCRT plus TIL-ACT was 67.5 and 75.10 months, respectively, exceeding the durations of 61.17 and 70.66 months (RMST of PFS and OS) in patients receiving CCRT alone ([199]Figure S8B-S8C). We further found that the 13 genes were significantly enriched in high activation and proliferation TIL subsets, such as C4_MHC_II_CD8_T_cells, C5_proliferating_CD8_T_cells-TNFRSF8, and C2_proliferating_CD8_T_cells-PCNA, which were linked to better survival after TIL-ACT. In contrast, TNFRSF18 encoding checkpoint protein[200]^48 was down-regulated in the non-progression group and highly expressed in the (DN) TIL cluster and was related to worse OS after TIL-ACT ([201]Figure S8D). Notably, these genes may function as potential targets to alter T cell bioactivity and enhance the clinical efficiency of TIL-ACT. In recent years, PD-1 antibody immunotherapy plus chemoradiotherapy has shown outstanding clinical achievement in advanced NPC patients in clinical trials,[202]^49 and some biomarkers, such as high stromal MHC class II positive cell density and tumor PD-L1 expression, are linked to clinical benefits of PD-1 antibody treatment.[203]^50 However, pEBV-DNA >10,000 copies/mL is linked to poor outcome of anti-PD-1 treatment,[204]^51 and patients with pEBV-DNA ≥4,000 copies/mL fail to achieve a survival benefit from sintilimab plus CCRT compared with CCRT alone.[205]^52 The pEBV-DNA load is a biomarker of recurrence and poorer prognosis in NPC patients.[206]^53 In our previous phase 2 clinical trial, the median baseline pEBV-DNA copy number was 28,300 copies/mL, indicating that most patients may not obtain benefit from immunotherapy, such as anti-PD-1 ICB treatment and TIL-ACT.[207]^12^,[208]^52 Viral infection has been reported to induce the expansion of (DN) T cells in the host,[209]^54 but the association and mechanism between heavy EBV DNA load and increased (DN) TILs in NPC patients need to be studied in the near future. If we selected the patients according to the frequency of CD8^+ TILs or (DN) TILs, the transcription level of prognostic genes (e.g., HHLA2) in the TIL infusion products, or gene signatures (e.g., MHC class II) in the baseline TME, we could observe longer survival in the CCRT+TIL-ACT arm compared with the CCRT-alone arm ([210]Figures S1F and [211]S8). In summary, we found that advanced NPC patients with a low (DN) TIL subset and high activating and proliferating CD8^+ TIL frequencies will obtain a survival benefit from TIL-ACT. In the functional analysis, we first determined that the (DN) TIL cluster functions as an activated Treg-like subset with a CD3^+CD4^−CD8^−IL-10^+TGF-β^+IKZF2^+FOXP3^−CTLA4^- T cell phenotype. The (DN) TILs hinder the clinical efficiency of TIL-ACT by suppressing CD8^+ TIL expansion and cytotoxic activity. In addition, the TME transcriptome status decides the composition of TIL infusion products and outcome of TIL-ACT. Taken together, our findings may aid in identifying suitable patients with advanced NPC for TIL-ACT and contribute to the development of strategies to enhance the clinical efficacy of TIL-ACT in the near future. Limitations of the study This is a retrospective study based on a clinical trial in high-risk NPC patients at a single center. The association between the frequency of (DN) TILs in TIL infusion products and clinical benefit of TIL-ACT needs to be confirmed in a prospective multicenter clinical trial including several types of solid tumors. The functional validation of these (DN) TILs is limited due to the difficulty of in vitro culture and scarcity of patient samples in this clinical trial. The regulatory mechanism of (DN) TIL expansion in the TME and its relationship with EBV antigen gene expression in NPC cells was not explored in this study. Nevertheless, this identification of (DN) TIL number in TIL infusion products as a negative biomarker of the clinical efficacy of TIL-ACT in high-risk NPC patients informs the future developmental directions of TIL-based immunotherapy strategies that can be personalized for maximal clinical benefit. Resource availability Lead contact Requests for further information and reagents may be directed to and will be fulfilled by the lead contact, Jiang Li (lijiang2@mail.sysu.edu.cn). Materials availability This study did not generate new unique reagents. Data and code availability * • The generated bulk RNA-seq, scRNA-seq, and scTCR-seq data in this study have been deposited to Genome Sequence Archive (GSA) in BIG Data Center, Beijing Institute of Genomics (BIG), with the following accession number GSA-human: [212]HRA008590. Other publicly available data can be downloaded from the Gene Expression Omnibus (GEO) database at accession number GEO: [213]GSE162025. All raw data have been deposited in the Research Data Deposit (RDD) public platform at accession number RDD: RDDB2025729586 ([214]www.researchdata.org.cn). * • This paper does not report original code. All custom code used for the analyses was written with existing software as detailed in the [215]STAR Methods section and is available upon request. * • Any additional information required to reanalyze the data reported in this work paper is available from the [216]lead contact upon request. Acknowledgments