Re-evaluating the melanoma TIL compartment and its unexpected spectrum of exhausted and functional T cells ========================================================================================================== * Cheryl M. Cameron * Brian Richardson * Jackelyn B. Golden * Yee Peng Phoon * Banumathi Tamilselvan * Lukas Pfannenstiel * Samjhana Thapaliya * Gustavo Roversi * Xing-Huang Gao * Leah L. Zagore * Mark J. Cameron * Brian R. Gastman ## Abstract Significant heterogeneity exists within the tumor infiltrating CD8 T cell population, and exhausted T cells harbor a subpopulation that may be replicating and retain signatures of activation, with potential functional consequences in tumor progression. Dysfunctional immunity in the tumor microenvironment is associated with poor cancer outcomes, making exploration of these exhausted but activated (Tex/act) subpopulations critical to the improvement of therapeutic approaches. To investigate mechanisms associated with Tex/act cells, we sorted and performed transcriptional profiling of CD8+ tumor infiltrating lymphocytes (TIL) coexpressing the exhaustion markers PD-1 and TIM-3, from large volume melanoma tumors. We additionally performed immunologic phenotyping and functional validation, including at the single cell level, to identify potential mechanisms that underlie their dysfunctional phenotype. We identified novel dysregulated pathways in CD8+PD-1+TIM-3+ cells that have not been well studied in TIL; these include bile acid and peroxisome pathway-related metabolism, and mammalian target of rapamycin (mTOR) signaling pathways, which are highly correlated with immune checkpoint receptor expression. Through bioinformatic integration of immunophenotypic data and network analysis, we propose unexpected targets for therapies to rescue the immune response to tumors in melanoma. ## Introduction Immune checkpoint inhibition (ICI) has revolutionized the treatment of many forms of cancer, especially UV induced skin cancers like melanoma 1–6. The success of ICI has also escalated the need for a comprehensive mechanistic understanding of the role cellular ICI targets play in oncogenesis and response to treatment. Furthermore, this successful clinical outcome has prompted a search for additional checkpoints and negative regulators of T cell function that can lead to new treatment options for the substantial subset of patients that are refractory 1–3, 5–7. Although many immune cells in the tumor microenvironment (TME) express immune checkpoints like programmed cell death-1 (PD-1), it is the exhausted tumor infiltrating CD8+ T cell that was shown to be the key target of reinvigoration by ICI 8–13. Systems biology has revolutionized the comprehensive objective assessment of the state of T cells in myriad disease models. Transcriptional analysis via RNASeq has transformed the field of tumor immunology and facilitated the identification of new subpopulations within what was previously thought to be a homogeneous set of immune populations 14, 15. The vast majority of literature in melanoma, however, relies on small biopsies, so it is likely that the data may not represent the entire spectrum of alterations occurring in the immune system in the TME 16, 17. An integrated systems biology approach may yield key transcriptomic biological maps of the TME (and/or identify gaps) to provide insight into the delicate balance of regulation within the tumor and immune cells to pinpoint therapeutically targetable states within the spectrum of function. We have a unique biobank of fresh, large volume tumors from surgical resections allowing us to sort down to very specific individual populations to investigate subset specific transcriptional regulation for discovery. We recently showed that the so-called “exhausted” CD8+PD-1+TIM-3+ TIL functionally inhibit healthy autologous T cell proliferation which can be inhibited by use of ICI therapies 18, 19. This finding contrasts with reports that these exhausted cells are dysfunctional, 20, 21. Interestingly, in the last few years several transcriptomic evaluations of CD8+ TIL identified exhausted T cell populations with high levels of activation 22 and even significant populations that are proliferating 23, however little has been done to investigate these TIL even though they express the same targets of modern ICI. Given that we study actively suppressive CD8 T cells that express checkpoints, we hypothesized such cells are a subset amongst others generally considered to be exhausted T cells. We further hypothesized that investigation of CD8+PD-1+TIM-3+ T cells that have pathways indicating activation (Tex/act) will yield key transcriptomic information that can be exploited to identify new targets for immunotherapy. We strategically sorted and transcriptomically profiled CD8+PD-1+TIM-3+ TIL from melanoma and from cutaneous squamous cell carcinoma (SCC) tumors to answer these questions. We undertook what appears to be the largest unbiased transcriptomic evaluation (in terms of number of patients) of this key cellular ICI target within the TME and identified numerous differentially regulated biochemical pathways. Of note, these Tex/act TIL are metabolically active and demonstrate profound upregulation of pathways including bile acid metabolism, mammalian target of rapamycin (mTOR) signaling, and peroxisome-related pathways. Given the fact that there is still a large group of melanoma and other cancer patients that do not derive benefit from ICI or other therapies, our findings may have broad application in helping reach the goal for therapeutic success in all patients. ## Results ### Transcriptomic profiling of dysfunctional CD8+ TIL CD8+ TIL in melanoma and SCC share important common features, including the coordinate expression of immune checkpoint receptors and the development of resistance to immunotherapy 18, 24, 25, yet the mechanisms underlying any TIL dysfunction in these tumors remain elusive. To address this, we performed transcriptomic profiling on flow-sorted CD8+PD-1+TIM-3+ from melanoma and SCC (total CD8+/P+/T+ TIL) and compared them to peripheral CD8+ T cells (pCD8) from healthy donors. A total of 3,957 genes were differentially expressed (p≤0.05) in total CD8+/P+/T+ TIL vs. pCD8+ T cells. (Fig. 1A). Gene Set Variation Analysis (GSVA) beginning with gene ontology (GO) molecular function annotation identified the corresponding top upregulated pathways including chemokine activity, and three related to metabolism including carbohydrate kinase activity, oxidoreductase activity II, and aspartic type peptidase activity (Fig. 1A). The top downregulated pathways included C2H2 zinc finger domain binding, p53 binding, rRNA binding, ARF guanyl nucleotide exchange factory activity, and androgen receptor binding (Fig. 1A). Clear differential expression of genes in total CD8+/P+/T+ (SCC and MEL) vs. pCD8 indicates upregulation of immune checkpoint receptors including TIM-3 (HAVCR2), PD-1 (PDCD1), LAG3, and TIGIT (Fig. 1B). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F1) Figure 1: Melanoma and SCC TIL display a distinct transcriptional profile from CD8+ T cells in the periphery. **(A)** Heatmap of 3,957 differentially expressed genes (DEG) (nominal p<0.05) in PD-1+ TIM-3+ TIL from melanoma (MEL TIL CD8+/P+/T+) and SCC (SCC TIL CD8+/P+/T+) vs. pCD8 cells. Gene expression is normalized by z-score with green indicating higher relative levels of expression and purple indicating relatively lower levels of gene expression. GO Molecular Function analysis (via GSVA) of DGE in melanoma and SCC CD8+/P+/T+ TIL (total CD8+/P+/T+ TIL) vs. pCD8 is shown to the right of the heatmap. Asterisks indicate truncated pathway names; full pathways are as follows: Oxidoreductase activity acting on the CH-NH group of donors (oxidoreductase activity I), oxidoreductase activity acting on the CH-NH2 group of donors oxygen as acceptor (oxidoreductase activity II), and hydrolase activity acting on carbon nitrogen but not peptide bonds in cyclic amidines (hydrolase activity). **(B)** Volcano plot of the top 50 up- or downregulated genes in total CD8+/P+/T+ TIL vs. pCD8s. Upregulated genes are shown in red while downregulated genes are shown in blue, significant genes of interest to this study are annotated in black. The red dashed line indicates the significance cutoff (nominal p < 0.05). **(C)** Volcano plot of the Hallmark pathway enrichment in total CD8+/P+/T+ TIL vs. pCD8s. Significant pathways of interest are annotated; the red dashed line indicates the significance cutoff (nominal p < 0.05). **(D)** Enrichment map of significantly differentially enriched Hallmark pathways (nominal p <0.05) in total CD8+/P+/T+ TIL vs pCD8s. Node size indicates negative (-)log10(pvalue), node color with associated scale denotes log2 fold change values for enrichment scores while edge weight represents the Jaccard coefficient between significant sets of genes in each pathway. **(E)** Regression of PD-1 (GEOMEAN) surface expression on CD8+ T cells on sample pathway enrichment scores from the Hallmark database. Pathway enrichment scores are normalized by z-score with green indicating higher relative levels of enrichment and purple indicating relatively lower levels of enrichment. Moreover, additional pathway enrichment analysis via GSEA using the Hallmark database (MSigDB) identified significant induction of multiple metabolic pathways in total CD8+/P+/T+ TIL including glycolysis, xenobiotic metabolism, fatty acid metabolism, mTORC1 signaling/PI3K AKT MTOR signaling, adipogenesis, and bile acid metabolism, corroborating our GSVA data (Fig. 1C). We performed analysis of pathway interactivity (via shared DEGs by Jaccard similarity coefficient) to provide a broader overview of how the enriched pathways interact, and as expected demonstrated a close correlation between mTORC1 signaling, PI3K AKT MTOR signaling, and glycolysis, with a less direct connection to bile acid metabolism (via mTORC1 signaling) (Fig. 1D). Glycolysis has been previously well described in TIL in melanoma and SCC; therefore, we were more intrigued with the significant differential enrichment of unexpected metabolic pathways, mTOR and bile acid metabolism in both cancers, which have not yet been proposed as key regulatory pathways in TIL function 26. Linear regression analysis of PD-1 (surface expression determined by flow cytometry) with gene expression revealed a strong positive correlation between PD-1 protein expression and upregulation of the bile acid metabolism, peroxisome, and glycolysis pathways in CD8+ TIL and PBMC (Fig. 1E). Moreover, core biological pathways that have been previously shown to be associated with TGF-β attenuation of the tumor response to immune checkpoint blockade also were significantly differentially regulated within the dysfunctional CD8+PD-1+TIM-3+ melanoma and SCC TIL (Supp. Fig. 1A). ### Identification of unique metabolic-associated dysfunctional CD8+ T cell clusters by UMAP analysis To integrate the expression of immune checkpoint receptors (ICRs), T cell phenotype, and activation markers with our transcriptional profiles, as well as to determine key pathways associated with TIL dysfunction, we carried out flow cytometric analysis of a randomly selected subset of MEL and SSC samples from tumors and peripheral blood (Fig. 2). We measured the expression of immune checkpoint receptors (PD-1, TIM-3, TIGIT, BTLA, LAG3, CD38) which have all been shown to regulate T cell function 27, with the hypothesis that coordinate expression of multiple immune checkpoint molecules will dramatically reduce T cell function. ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F2) Figure 2: UMAP analysis and linear regression with immune checkpoint cell surface markers **(A)** UMAP dimensional reduction analysis of CD8+ TIL compared to CD8+ PBMC reveals 17 unique clusters of coordinate cell surface protein expression. **(B)** Two-way hierarchically clustered heatmap of the MFI (geometric mean fluorescence intensity) of surface expression of CD38, TIGIT, PD-1, TIM-3, LAG3, BTLA, CD3, CD8, CD45RA, CD27, and CCR7 within the clusters in the UMAP analysis. Clusters significant for all TIL vs. pCD8 are indicated by red asterisks (2, 3, 5, 6, 8, 9, 15, 16), for MEL TIL vs. SCC TIL by blue asterisks (1, 5), for MEL TIL vs. pCD8 by orange asterisks (9, 15, 16), and SCC TIL vs. pCD8 by green asterisks (5, 9, 16). Heatmap is z-score transformed with upregulated MFI in red and downregulated MFI in blue. **(C)** Individual counts per selected cluster are shown in the box and whisker plots (clusters 6, 15, and 16 are notably enriched in all TIL compared to pCD8 cells; p<0.05; Mann-Whitney test). Linear regression modeling of each cluster with the RNAseq data reveals pathways enriched that are unique to each cluster. We employed uniform manifold approximation and projection (UMAP), a nonlinear dimensionality-reduction machine learning approach, to analyze our multiparametric flow data and objectively determine the heterogeneity of CD8+ T cells based on surface expression of BTLA, CD3, CD8, CD27, CD38, CD45RA, CCR7, LAG3, PD-1, TIGIT, and TIM-3. UMAP analysis revealed 17 unique clusters, 9 of which show significantly different frequencies in CD8+ TIL compared to PBMC (Fig. 2A). A two-way hierarchically clustered heatmap of the MFI (geometric mean fluorescence intensity) based on surface expression showed the distinct surface signature profiles of the 17 clusters identified by UMAP analysis (Fig. 2B). Notably, the frequencies of lymphocytes co-expressing multiple immune checkpoints (CD38, TIGIT, LAG3, PD-1, TIM-3), the negative regulators of T cell function, were enriched in CD8+ TIL compared to periphery, especially in clusters 6, 15 and 16. Of note, cluster 6 represents what is likely to be the most highly dysfunctional CD8 population, characterized by relative upregulation of CD38, TIGIT, PD-1, LAG3 and TIM-3, and is significantly enriched in the CD8+ TIL population compared to the peripheral CD8+ T cells (Fig. 2B and 2C). Similar to cluster 6, clusters 10, 15 and 16 have similar ICR expression patterns with increased coordinate immune checkpoint expression levels (Fig. 2B). Interestingly, cluster 16 is significantly enriched in CD8+ TIL of both melanoma and SCC with high expression of PD-1 and TIM-3 (Fig. 2B and 2C). Likewise, cluster 15 is also enriched in CD8+ TIL, but only in melanoma, with moderate immune checkpoint expression (Fig. 2B and 2C). Our findings indicate that clusters 6, 10, 15 and 16, with upregulation of ICRs, are likely to represent progressively more exhausted CD8+ T cell clusters (Fig. 2B). We then performed linear regression modeling of gene expression with frequencies of cells from each flow UMAP cluster in each donor to identify transcriptomic pathway enrichment that correlates significantly with frequencies of CD8+ T cell clusters identified by UMAP. Interestingly, higher frequencies of cells in cluster 6 were strongly correlated with positive enrichment of the bile acid metabolism and peroxisome pathways (Fig. 2C). Likewise, cluster 16 frequency was also positively correlated with enrichment of the peroxisome and glycolysis pathways (Fig. 2C). In summary, our data demonstrate significant correlation between upregulation of immune checkpoint-expressing dysfunctional CD8+ TIL with activation of metabolic function, particularly bile acid metabolism, peroxidase and glycolysis pathways at the transcriptomic level. Targeting this metabolic-associated dysfunctional CD8+ Tex/act cell population could reinvigorate exhausted CD8+ T cells, thus enhancing immunotherapy responses in cancer patients. ### Upregulation of proliferation and metabolic associated genes in dysfunctional CD8+ TIL in melanoma To determine whether these metabolic pathways were specifically dysregulated in melanoma, we performed differential gene expression analysis of the melanoma CD8+ TIL versus healthy peripheral CD8+ T cells resulting in a distinct gene expression signature as shown by the heatmap in Figure 3A. A total of 4,280 genes were differentially expressed in MEL CD8+ TIL versus peripheral CD8+ T cells, while 2,057 of those genes were commonly differentially regulated in MEL and SCC TIL (p≤0.05, Supp. Fig. 1B). The most statistically upregulated pathways using GO molecular function annotation (via GSVA) were oxidoreductase, structural constituent of muscle, and carbohydrate binding pathways (Fig. 3A). The top downregulated pathways include C2H2 zinc finger domain binding, N-acetyltransferase activity, rRNA binding, peptide N-acetyltransferase activity, and transcription coactivator activity. The top 50 differentially regulated genes between MEL TIL and pCD8 by statistical significance are shown in Figure 3B and the ICRs PD-1 (PDCD1), TIM-3 (HAVCR2), CTLA4 and LAG3 were among these. Also significantly upregulated were FABP5 (epidermal fatty acid binding protein) which can act as an intracellular receptor that binds to free fatty acids (FFA) in the cytosol and Ki-67 (MKI67), indicating enhanced proliferation of the CD8+ TIL. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F3) Figure 3: PD-1+TIM-3+ TIL from melanoma display a distinct transcriptional profile from CD8+ T cells in the periphery **(A)** Heatmap of 4,280 differentially expressed genes (DEG) (nominal p<0.05) in TIL from melanoma vs. peripheral CD8 cells. Gene expression is normalized by z-score with green indicating higher relative levels of expression and purple indicating relatively lower levels of gene expression. Genes of interest are annotated. GO Molecular Function analysis (via GSVA) of DGE in melanoma TIL vs. pCD8 is shown to the right of the heatmap. **(B)** Top 50 differentially expressed genes from the MEL TIL vs. pCD8 contrast by p-value (nominal p < 8.93e-9). **(C)** Heatmap of significant (nominal p<0.05) Hallmark pathways associated with pCD8 versus MEL TIL. **(D)** Enrichment map of significantly differentially enriched Hallmark pathways (nominal p <0.05) in TIL from melanoma vs pCD8s. Node size indicates -log10(pvalue), node color with the associated scale denotes log2 fold change values for enrichment scores while edge weight represents the Jaccard coefficient between the sets of significant genes in each pathway. **(E)** Western blot analysis of control pCD8+ T cells (n=3) and melanoma TIL (n=3). Protein targets are shown on the right y-axis; β-actin was used as a loading control. **(F)** Volcano plot of 1072 differentially expressed genes (nominal p<0.05) in patients who have undergone immunotherapy (anti-PD-1). Upregulated genes are shown in red while downregulated genes are shown in blue; the top 10 up- and downregulated genes are annotated (red dashed line indicates p-value cutoff (nominal p<0.05)). ### Enrichment of metabolism pathways in CD8+ TIL in melanoma To further evaluate the biology underlying differentially expressed genes found in CD8+ TIL and PBMC in melanoma, we performed pathway enrichment analysis of key canonical pathways. Notably, the peroxisome and bile acid metabolism pathways were coordinately regulated in melanoma TIL compared to pCD8 (Fig. 3C). Although a small number of reports have recently emerged showing the potential immunomodulatory effects of bile acids on the immune response, to the best of our knowledge ours is the first report of bile acid metabolism pathway dysregulation in tumor infiltrating lymphocytes in cancer 28–30. Not only are *peroxisome proliferator-activated receptor* (PPAR) pathways associated with FABP5, which was upregulated in dysfunctional CD8+ TIL, but they also regulate bile acid synthesis 31–33. Our data showed a strong positive enrichment of bile acid metabolism pathways in dysfunctional CD8+ T cells from melanoma tumors. One of the key regulatory enzymes of the bile acid metabolism pathway is CYP27, a cytochrome P450 enzyme encoded by the CYP27A1 gene 34. CYP27 metabolizes cholesterol into 27-hydroxycholesterol (27HC) through a hydroxylation reaction. Current evidence indicates 27HC can exert modulatory effects on the immune system and act as a selective estrogen receptor modulator (SERM) 35, 36. Strikingly, 27HC stimulates the proliferation of mouse melanoma cells by activating estrogen receptor alpha (ERα) and triggering the AKT and MAPK pathways 37. Compelling research into potential therapeutic strategies targeting bile acid metabolism can be found in breast cancer research 38, 39. For example, pharmacologic inhibition of CYP27A1 improved the efficacy of anti-PD-1 treatment and decreased metastatic breast cancer growth in mice 40. Additional studies have shown that CYP21A expression in human breast cancer tumors is correlated with disease progression 38. Notably, CYP27A1 was significantly upregulated in the CD8+ T cells of melanoma TIL as compared to PBMC (Fig. 3A). Upregulation of additional genes from the bile acid and peroxisome signaling pathways was also evident in TIL gene and pathway expression analysis, including SOD1, ACSL5, FADS2, CAT, DHCR24, SLC27A2, and IDH2 in melanoma TIL compared to pCD8s (Supp. Fig. 2). As shown in Figure 3C, pathway enrichment analysis showed that the mTORC1 signaling pathway was significantly and positively enriched in CD8+ melanoma TIL as compared to pCD8s. The glycolysis pathway was highly integrated with many other pathways, particularly the mTORC1 and peroxisome pathways, which interact with bile acid metabolism (Fig. 3D). Compared to pCD8 T cells of control individuals, the glycolysis pathway was the most significantly upregulated pathway within the pathway interaction network analysis of the Hallmark canonical pathway database in CD8+ TIL of melanoma patients (Fig 3D). The differential regulation of multiple metabolic pathways, including bile acid metabolism, mTORC1, and glycolysis (Fig. 3D) likely reflects the fact that the CD8+ TIL cells are hosted in a hypoxic, acidic, and nutrient-depleted TME where the cellular metabolism has been reprogrammed as an adaptive mechanism to facilitate their proliferation and survival in melanoma. Our western blot analysis successfully revealed increased expression of CYP27A1, which was upregulated in the bile acid metabolic pathway, and three additional glycolytic genes (PKM2, PHGDH and PCK2) in melanoma CD8+ TIL compared to PBMCs (Fig. 3E). ### Identification of immunotherapy signatures A subset of our cohort of melanoma patients were treated with anti-PD-1 immunotherapy. There were 1,072 differentially expressed genes (p≤0.05) in patients receiving therapy (n=5) as compared to those that did not receive treatment (n=8) as summarized in the volcano plot in Figure 3F. CXCR6 was upregulated in the melanoma anti-PD-1 immunotherapy signature, indicating a potential shift towards a proinflammatory tumor microenvironment which could lead to subsequent metastasis. CTTNBP2NL (CTTNBP2 N-terminal-like protein) was also upregulated and is known to be associated with STRIPAK complexes which have been broadly linked to metabolism, immune regulation, and cancer tumorigenesis 41. On the other hand, CD160 and HRH2 (histamine receptor H2) were downregulated in the anti-PD-1 therapy signature. ### mTOR Pathway Validation The mTORC1 signaling pathway was highly significantly enriched in tumors as compared to peripheral blood (Figs. 1C and 3C). Using intracellular flow cytometry, we went further to discover that two of the key targets of mTORC1 complex activation, S6 ribosomal protein (target of S6 kinase which is a direct target of mTOR phosphorylation) and 4EBP1 (a direct target of mTOR phosphorylation), are significantly more phosphorylated in the MEL TIL compared to peripheral CD8 T cells, validating our findings at the phosphoprotein level (Fig. 4A). Furthermore, treatment with rapamycin (sirolimus), which specifically blocks MTORC1 signaling during TCR (anti-CD3/CD28) stimulation of melanoma TIL, resulted in consistent downregulation of PD-1 expression, particularly in the CD8+CD45RA- memory T cells and CD8+CD45RA-CCR7-CD27- effector memory T cells in the tumors (Fig. 4B). ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F4) Figure 4: Validation of mTOR pathway (A) Intracellular flow cytometry of two phosphorylated targets, S6 ribosomal protein and 4EBP1, demonstrates significantly higher in the CD8+ TIL of melanoma compared to peripheral CD8+ T cells. *p=0.01 and **p=0.04 by Welch’s t-test **(B)** PD-1 expression is downregulated with rapamycin (25nM) treatment during CD3/CD28 stimulation (72hr) in melanoma TIL. CD8+CD45RA-CCR7-CD27- effector memory T cells are shown on the left (*p=0.05, paired t-test) and CD8+CD45RA- memory T cell trends are shown on the right (n.s.). **(C)** scRNAseq of CD8+ TIL in melanoma visualized with UMAP dimensional reduction analysis demonstrates 9 unique clusters. **(D)** Pathway analysis of Cluster 8 at single cell resolution demonstrates 18 significantly enriched pathways (nominal p<0.05) with positive enrichment of mTOR-related pathways such as mTORC1 and PI3K-AKT-MTOR (shown in orange text). **(E)** Violin plots of selected MTOR-related gene expression (PHGDH, E2F1, PSMA4, BUB1, CDK2) in the scRNAseq clusters. Moreover, to validate differential enrichment of the mTOR pathway, we analyzed the single cell transcriptomes of CD8+ TIL from a new cohort of metastatic melanoma patients collected prior to anti-PD-1 immunotherapy. UMAP dimensional reduction of the single cell RNA sequencing (scRNAseq) dataset revealed a total of 9 clusters (Fig. 4C). Cluster 8 demonstrated positive enrichment of the mTORC1 and PI3K/AKT/MTOR signaling pathways at the single cell resolution, validating our bulkseq findings (Fig. 4D). Violin plots of MTOR-related genes unique to cluster 8 (PHGDH, E2F1, PSMA4, BUB1, CDK2) are shown in Figure 4E, all of which were also differentially regulated in our bulk RNAseq signature of dysfunctional CD8+ TIL. We also confirmed PHGDH expression at the protein level (Fig. 3E). ## Discussion Our study identified several novel gene signatures that distinguish CD8+ melanoma TIL from peripheral blood. More importantly, our findings reveal potential metabolic pathways that can be targeted to reinvigorate the dysfunctional immune system observed in melanoma and SCC-derived CD8+PD-1+TIM-3+ TIL. Through comprehensive transcriptomic analysis of immune checkpoint enriched CD8+ TIL and PBMC, we show that several metabolic signaling pathways, such as bile acid and peroxisome, along with mTOR pathways, are enriched in dysfunctional CD8+ Tex/act cells. The observation of dysregulated bile acid metabolism in melanoma and SCC TIL is especially interesting given the considerable amount of data highlighting the relationship between antitumor immune response and the microbiome 42–44. The liver produces primary bile acids, which are then modified by gut microbes into a wide range of compounds with roles in gut metabolism, cell signaling, and microbe composition. Perturbations in the microbial ecosystem have been linked to immune evasion, carcinogenesis, and chronic inflammation and can affect the efficacy of cancer immunotherapies, including ICIs 45. While most studies have focused on the gut microbiome and its connection to immune surveillance dysregulation; the interplay between cancer cells, immune cells, and microbiota is also relevant in the tumor microenvironment 46, 47. Notably, it was observed that numerous tumor types, including melanoma, have distinct microbial signatures. The abundance of specific intratumor bacteria in melanoma has been linked to CD8+ T cell infiltration and patient survival 48. Additionally, patient immunotherapy response has been linked to metabolic functions encoded by a unique pattern of intratumor bacteria 49. An intriguing possibility is that dysregulation of the bile acid metabolism pathway in CD8+ TILs is interconnected with tumor microbiome dysbiosis in the TME and may influence the anticancer immune response. It is plausible that manipulating the tumor-associated microbiome may also influence tumor immunity and patient response to immune therapy, as has been demonstrated in the gut microbiome 42, 50, 51. Further investigation is needed to explore the interdependence of the tumor microbiome, tumor metabolic reprogramming, and CD8+ T cell dysfunction. Upregulation of bile acid-related genes in tumor infiltrating CD8+ T cells correlates with T cell dysfunction activity and may also be related to the differential regulation of metabolic-related pathways such as mTORC1. Connections between bile acid and mTOR pathway activation have been reported. Yamada et al. showed that secondary bile acids activate the mTOR pathway via FXR signaling 52. The bile acid activation of mTOR signaling may be caused by upregulation of DGKH (diacylglyerol kinase), an enzyme that converts diacylglycerol to phosphatidic acid which directly activates mTOR 53. DGKH was significantly upregulated in tumor infiltrating CD8+ T cells in melanoma and SCC in our study. Our flow cytometric UMAP analysis (Fig. 2) revealed clusters representative of different T cell populations. Dimension reduction of our multiparametric flow cytometry data from peripheral CD8 T cells and CD8+ TILs clearly demonstrated that the frequency of cells belonging to cluster 6, with the highest coordinate normalized expression of all immune checkpoint receptors (ICR: CD38, TIGIT, PD-1, TIM-3, and LAG3) as well as CD8 and CD27, was significantly correlated with enrichment of genes from the bile acid metabolism, peroxisome, and fatty acid metabolism pathways in a linear regression model (Fig. 2C). Peroxisome, fatty acid metabolism and glycolysis pathways were also positively correlated with the frequency of cells in cluster 16. We were intrigued to discover that the frequency of cells in clusters that expressed the highest levels of coordinate immune checkpoint receptor expression were significantly positively correlated with enrichment of key metabolic pathways. We propose that TIL clusters 15, 16 and 6 are enriched for CD8+ Tex/act activity and may identify progressive levels of immune exhaustion/dysfunction in the CD8 compartment, with progressive coordinate expression of ICR to maximal levels in cluster 6, whose frequency has significant correlation with enrichment of the bile acid metabolism pathway. Our results are consistent with the hypothesis that transcriptional reprogramming is likely to play a role in driving the changes of cellular metabolism in dysfunctional CD8+ TIL in melanoma. TOX, a transcription factor that drives CD8 T cells toward an exhausted state, is upregulated in SCC and melanoma TIL when compared to pCD8 (Fig. 1B and 3A) 54, indicating that targeting TOX along with PD-1 could off-set T cell exhaustion. Two key metabolic genes, PKM2 and PHGDH, are upregulated in melanoma TIL compared to healthy pCD8 (3A, and 3E). PKM2 (pyruvate kinase M1/2) expression, which controls glycolysis, has been associated with tumor progression and poor survival in a small pilot study of metastatic melanoma patients 55. In addition, PHGDH (D-3-phosphoglycerate dehydrogenase) is overexpressed in multiple cancers, correlates with tumor growth, and has been shown to be increased in 40% of melanoma samples 56, 57. In a subset of our patients that received anti-PD-1 immunotherapy (Fig. 3F), we observed upregulation of CXCR6 and CTTNBP2NL and downregulation of CD160 and HRH2. CXCR6, C-X-C chemokine receptor type 6, is known to be preferentially expressed on CD8+PD-1high exhausted T cells in hepatocellular carcinoma (HCC) and is hypothesized to play a role in recruiting CD8+ T cells into the tumor microenvironment 58. Overexpression of CTTNBP2NL and subsequent STRIPAK complexes could fuel the exhausted CD8+ T cells, thus suppressing an immunotherapy effect and promoting tumor progression. These overexpressed refractory gene profiles open new avenues for novel immunotherapy targets. CD160, a degranulation marker, was downregulated in our immune failure signature. Its expression has been demonstrated to be downregulated with repeated antigen stimulation while PD-1 and LAG3 remained high and TIM-3 increased further in T cells from a murine breast cancer model 59. Furthermore, in CD8+ T cells from individuals with HIV, CD160 and PD-1 co-expression represented a subset of exhausted T cells both functionally and transcriptionally 60. Therefore, downregulation of CD160 may indicate dysfunctional TIL that are either no longer degranulating or lack continued TCR engagement. Finally, downregulation of HRH2 (histamine receptor H2) could be indicative of tumor metastasis in the context of immune failure as a balance of histamine with its receptor is necessary to either stimulate or suppress the growth of melanoma 61. scRNASeq and UMAP analysis indicated that cluster 8 correlates with increased mTOR-related signaling, and we identified five genes unique to this cluster (Figure 4C-E). PHGDH, validated in our western blot analysis and scRNAseq, is involved in diverting glycolytic fluxes and is highly expressed in tumors. One study demonstrated it to be amplified at a 40% frequency in melanoma samples 57 as reviewed in 56. CDK2, E2F1, and BUB1 (Mitotic Checkpoint Serine/Threonine-Protein Kinase BUB1) are involved in cell cycle progression and targeting them could prove to be a viable therapeutic strategy. Inhibition of CDK2 in melanoma cell lines was shown to overcome their resistance to BRAF and Hsp90 inhibitors 62 while BUB1 has been identified as a novel target downstream of SIRT1 in melanoma 63. E2F1 is overexpressed in melanoma and its inhibition initiates cell cycle arrest/apoptosis as well as increasing the sensitivity of the melanoma cells to BRAF inhibitors 64. Finally, PSMA4 was upregulated in OT1 CD8+ T cells following transient and continuous Ag-independent stimulation 65. Our highly focused transcriptomic profiling of dysfunctional CD8+PD-1+TIM-3+ TIL in human melanoma and SCC has identified several seemingly disparate and independent metabolic pathways that are differentially regulated in CD8+ Tex/act cells in the tumor microenvironment. Singer et al. applied a similar systems immunology approach to analyzing TIL from WT versus MT-/- pMEL mice implanted with B16 murine melanoma cells 22. While their study was performed in mice, Singer et al. identified a gene expression signature distinguishing CD8+ T cell dysfunction from T cell activation that could also be distinguished (via meta-analysis) amongst a previous scRNAseq study of TIL from human metastatic melanoma tumors 66. We recently employed a biased single cell transcriptomic approach to link higher levels of oxidative phosphorylation in tumor- and peripheral blood–derived total CD8+ T cells with ICI resistance in melanoma patients 67. Our current systems biology analysis of human melanoma was conducted in an unbiased fashion to fully interrogate dysfunctional CD8+PD-1+TIM-3+ TIL (sorted) transcriptomic profiles, allowing for identification of novel and unexpected signaling pathways that may reveal enhancement targets in more recent immune checkpoint blockade therapies and be more capable of discerning responders from nonresponders in broad cohorts. Taken together, our findings indicate that mTOR, bile acid metabolism, and peroxisome metabolic pathways are coordinately dysregulated and highly upregulated in the CD8+ TIL of melanoma and SCC patients, impacting the capacity of the host immune system to suppress and eradicate tumor cells. The discovery of novel metabolic targets in Tex/act cells could also reinvigorate resistance in immune checkpoint therapy failure, thus improving clinical efficacy. Our results provide a new platform and public resource for datamining and developing novel treatment modalities to further improve current immunotherapy advancement. ## Methods ### Patient selection and demographics All human tissue was obtained at the Cleveland Clinic under a protocol approved by Cleveland Clinic’s institutional review board, and written informed consent was obtained from each patient. Peripheral blood lymphocytes (PBLs) and tumor specimens were obtained from patients with cutaneous melanoma (MEL, n=23), squamous cell carcinoma (SCC, n=8), and control individuals without skin disease (n=8) as previously described 18. MEL patients were 60.9% male with a mean age of 58 (±15.8); 3 patients presented with a primary tumor while 20 patients had metastatic disease. Fourteen of the MEL patients received some type of immunotherapy (IO), out of these patients, 8 received anti-PD-1/PD-L1 directed IO (either pembrolizumab or nivolumab). SCC patients were 85.7% male with a mean age of 73 (±7.9); 3 patients presented with a primary tumor while 5 patients had metastatic disease. Age and sex are unknown for one SCC patient. Tissue for scRNAseq libraries was obtained from patients with cutaneous melanoma (n=8). The patient cohort was 37.5% male with a mean age of 60 (±12.7). 2 patients presented with a primary tumor while 6 patients had metastatic disease. ### Isolation of TILs and PBLs After surgical resection, tumor specimens were washed with antibiotic-containing media and minced with crossed scalpels under sterile conditions. Tissue was dissociated via enzymatic digestion using 1,500 U/ml collagenase IV (Gibco/Life Technologies), 1,000 U/ml hyaluronidase (Sigma), and 0.05 mU/ml DNase IV (Gibco) in RPMI for 1 h at 37°C followed by mechanical agitation. Centrifugation over Ficoll-Hypaque gradient was used to separate debris from the single cell suspension. Finally, cells were cryopreserved in 10% DMSO + bovine serum. Similarly, peripheral blood mononuclear cells were purified from buffy coats by centrifugation over a Ficoll-Hypaque gradient followed by cryopreservation. ### Flow cytometric analysis Flow cytometric analysis of immune checkpoint receptor and activation marker expression and T cell memory subset distribution was carried out as follows: for memory subset phenotyping and assessment of negative regulator and ligand expression, a cocktail of the following monoclonal antibodies was used: anti-CD3 Alexa700 (BD), anti-CD4 Q.605 (Invitrogen), anti-CD8 PerCP (Biolegend), anti-CD45RA BV650 (Biolegend), anti-CD27 APC-eFluor 780 (eBioscience), anti-CCR7 PE-CF594 (BD), anti-CD14 V500 (BD), anti-CD19 BV510 (Biolegend), Live/Dead Amcyan (Invitrogen), anti-BTLA (BD), anti-TIM3 BV421 (Biolegend), anti-PD-1 PE-Cy7 (Biolegend), anti-CTLA-4 APC (BD), anti-TIGIT PE (eBioscience), anti-LAG3 FITC (Novus). After washing, cells were resuspended in staining buffer and sorted on an ARIA-SORP. The following antibodies were used for intracellular flow cytometry to assess phosphorylation of mTOR targets: anti-S6 (S235/S236) V450 (BD), anti-p4E-BP1 (T36/46) PE (BD). For the rapamycin assay, cells were stimulated for 3 days with CD3/CD28 beads and then treated with 25nM rapamycin overnight. Cells were stained and analyzed for PD-1 surface expression. The following antibodies were used to identify memory T cells and effector memory T cells: anti-CD8 BV711 (BD), anti-CD45RA BV650 (Biolegend), anti-CCR7 FITC (R&D), anti-CD27 APC eFluor 780 (eBioscience). Data were analyzed using FlowJo software (TreeStar) and our custom pipeline for dimensionality reduction. ### Western blot Isolated CD8 cells (50,000) from TIL were lysed in 50 uL RIPA buffer (150mM NaCl, 1mM EDTA, 0.5% Triton X-100, 0.5% deoxycholic acid, 0.5% SDS, and 100mM Tris (pH 7.5) containing protease and phosphatase inhibitors. An equal sample volume (100ul) was used for reducing gel electrophoresis (4-12%) followed by immunodetection with antibodies toward CYP27A1 (Abcam), Actin (Abcam), PHGDH (Cell Signaling), PKM2 (Cell Signaling), and PCK2 (Cell Signaling). Three biological replicates were performed. ### Bulk RNA-seq and bioinformatic analysis RNA was purified from CD8 T cells using RNeasy Micro Kits (Qiagen), followed by low input RNASeq library generation using Takara SMART-Seq v4 Ultra Low/Nextera XT with Nextera Index v2 Set A. Paired-end sequencing reactions were run on an Illumina NextSeq 550 High Output platform (25M total reads per sample). Raw demultiplexed fastq paired-end read files were trimmed of adapters and filtered using the program skewer to remove reads with an average phred quality score of less than 30 or trimmed to a length of less than 36 68. Trimmed reads were then aligned using the HISAT2 aligner to the Homo sapiens NCBI reference genome assembly version GRCh38 and sorted using SAMtools 69, 70. Aligned reads were counted and assigned to gene meta-features using the program featureCounts as part of the Subread package 71. These count files were imported into the R programming language and were assessed for quality control, normalized and analyzed, utilizing the limma-trend method 72 for differential gene expression testing as well as regression modelling and GSVA 73. Linear regression modelling was performed using the limma framework. RNA-seq data supporting this study have been deposited in the Gene Expression Omnibus (GEO) public database with the accession number pending. ### scRNAseq Library Preparation and Data Processing All cells were resuspended in DPBS with 0.04% BSA and immediately processed for scRNAseq as follows. Cell count and viability were determined using trypan blue on a Countess FL II, and approximately 12,000 cells were loaded for capture onto the Chromium System using the v2 single cell reagent kit according to the manufacturer’s protocol (10X Genomics). Following capture and lysis, cDNA was synthesized and amplified (12 cycles) as per manufacturer’s protocol (10X Genomics). The amplified cDNA from each channel of the Chromium System was used to construct an Illumina sequencing library and was sequenced on an Illumina HiSeq 2500 with 150-cycle sequencing (asymmetric reads per 10X Genomics). Illumina basecall files (*.bcl) were converted to FASTQs using CellRanger v3.0, which uses bcl2fastq v2.17.1.14. FASTQ files were then aligned to GRCh38 human reference genome and transcriptome using the CellRanger v3.0 software pipeline with default parameters as reported previously 74; this demultiplexes the samples and generates a gene versus cell expression matrix based on the barcodes and assigns UMIs that enables determination of the individual cell from which the RNA molecule originated. Overall, an estimated total of 71,238 cells were analyzed. ### scRNASseq Bioinformatic Analysis Bioinformatic analysis of cells based on whole transcriptomes was performed by the R package Seurat (version 3.0) 75. Dimensionality of gene-barcode matrices was first reduced to 18 principal components using principal components analysis (PCA). PCA-reduced data were further reduced to 2-dimensional space using the UMAP method and visualized. Graph-based clustering of cells was conducted in the PCA space; a sparse nearest-neighbor graph of the cells was built first and Louvain modularity optimization was then applied. The number of nearest neighbors was logarithmic in accordance with the number of cells. In the last step, repeated cycles of hierarchical clustering and merging of cluster pairs that had no significant differential expression were performed, until no more cluster pairs could merge. Differential gene expression analyses of each cluster were conducted by Wilcoxon rank sum test. The log2 fold-change of a certain gene’s expression (UMIs) in one cluster vs. all other clusters, and the corresponding adjusted p-values, were calculated for each cluster with pathway enrichment scores generated using the Gene Set Enrichment Analysis (GSEA) method 76. ### Statistics Unless otherwise indicated, the Student’s t-test was used with p≤0.05 chosen as the level of significance and the analysis was performed with Graphpad Prism 8. ## Data Availability RNA-seq data supporting this study have been deposited in the Gene Expression Omnibus (GEO) public database with accession number pending. ## Competing interests B. Gastman is a consultant/advisory board member for Merck. No potential conflicts of interest or competing interests were disclosed by the other authors. ![Supplemental Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F5.medium.gif) [Supplemental Figure 1:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F5) Supplemental Figure 1: Characterization of dysfunctional CD8+ T cells in total TIL and PBMC **(A)** Venn analysis identified genes that are common and unique in total (melanoma and SCC) CD8+PD-1+TIM-3+ TIL (nominal p≤0.05). 2,057 genes were commonly differentially regulated in both total CD8+/P+/T+ TIL and peripheral CD8+ T cells, while 2,223 and 789 were unique to melanoma and SCC CD8+/P+/T+ TIL, respectively, in comparison to healthy peripheral CD8+ T cells. **(B)** Core biological pathways associated with TGF-β attenuation of the tumor response to immune checkpoint blockade were significantly differentially regulated within the dysfunctional CD8+/P+/T+ melanoma and SCC TIL as shown by the two way heatmap. Pathways and genes (noted on the right y-axis) include those involved in FGFR3 signaling (TP63), CD8 T cell effector function (GZMA, GZMB, PRF1 and CXCL10), immune checkpoint receptors and their ligands (PDCD1LG2, CTLA4, PD-1, LAG3, TIM-3, and TIGIT), cell cycle (MKI67, CCNE1, BUB1, BUB1B, CCNB2, CDC25C, CDK2, MCM4, and MCM6), DNA damage response (BRCA2, ERCC2, FANCA, and RAD51c), and TGF-β response signature genes (ACTA2 and COL4A1). HLA-B and TGFB1 genes were uniquely downregulated in the CD8+ TIL as compared to PBMC. All p≤0.05. ![Supplemental Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/04/03/2023.04.02.23288048/F6.medium.gif) [Supplemental Figure 2:](http://medrxiv.org/content/early/2023/04/03/2023.04.02.23288048/F6) Supplemental Figure 2: Hallmark pathway enrichment in melanoma TIL. Top 20 differentially enriched pathways between melanoma TILs and pCD8s with associated top 50 shared differentially expressed genes. Pathways and genes were selected based on a nominal p≤0.05 and the number of genes in common between pathways. Heatmap values show the logFC values for each gene between melanoma TILs and pCD8s while their column annotations denote p values. Row annotations denote the logFC values of pathway enrichment scores between the two groups. ## Acknowledgments We thank the Genomics Core at the Lerner Research Institute of Cleveland Clinic and the Genomics and Applied Functional Genomics Cores at Case Western Reserve University for their technical and analytical support. This research is supported by Cleveland Clinic and Case Western Reserve University internal funding. ## Abbreviations Tex/act : exhausted but activated T cell subpopulation TIL : tumor infiltrating lymphocyte mTOR : mammalian target of rapamycin ICR : immune checkpoint receptor TME : tumor microenvironment PD-1 : programmed cell death-1 SCC : cutaneous squamous cell carcinoma pCD8 : peripheral CD8 T cells MEL : melanoma IO : immunotherapy RIPA : radioimmunoprecipitation assay buffer GEO : Gene Expression Omnibus PCA : principal components analysis UMAP : uniform manifold approximation and projection MFI : mean fluorescence intensity HCC : hepatocellular carcinoma; GSVA : Gene Set Variation Analysis DEG : differentially expressed genes PBMC : peripheral blood mononuclear cells scRNAseq : single cell RNA sequencing Tim-3 : T cell immunoglobulin and mucin protein 3 * Received April 2, 2023. * Revision received April 2, 2023. * Accepted April 3, 2023. * © 2023, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## References 1. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366, 2443–2454 (2012). [https://doi.org:10.1056/NEJMoa1200690](https://doi.org:10.1056/NEJMoa1200690) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1200690&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22658127&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000305747000004&link_type=ISI) 2. Garon, E. B. et al. Five-Year Overall Survival for Patients With Advanced Non‒Small-Cell Lung Cancer Treated With Pembrolizumab: Results From the Phase I KEYNOTE-001 Study. J Clin Oncol 37, 2518–2527 (2019). [https://doi.org:10.1200/jco.19.00934](https://doi.org:10.1200/jco.19.00934) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1200/JCO.19.00934&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 3. Rizvi, N. A., et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015). [https://doi.org:10.1126/science.aaa1348](https://doi.org:10.1126/science.aaa1348) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNDgvNjIzMC8xMjQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. Zheng, C. et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell 169, 1342–1356 e1316 (2017). [https://doi.org:10.1016/j.cell.2017.05.035](https://doi.org:10.1016/j.cell.2017.05.035) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2017.05.035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28622514&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 5. Hamid, O. et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369, 134–144 (2013). [https://doi.org:10.1056/NEJMoa1305133](https://doi.org:10.1056/NEJMoa1305133) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1305133&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23724846&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000321567300009&link_type=ISI) 6. Krieg, C. et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med 24, 144–153 (2018). [https://doi.org:10.1038/nm.4466](https://doi.org:10.1038/nm.4466) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.4466&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 7. Hui, R. et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann Oncol 28, 874–881 (2017). [https://doi.org:10.1093/annonc/mdx008](https://doi.org:10.1093/annonc/mdx008) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdx008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 8. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014). [https://doi.org:10.1038/nature13954](https://doi.org:10.1038/nature13954) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13954&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25428505&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000346247600054&link_type=ISI) 9. Wei, S. C. et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 170, 1120–1133 e1117 (2017). [https://doi.org:10.1016/j.cell.2017.07.024](https://doi.org:10.1016/j.cell.2017.07.024) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2017.07.024&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28803728&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 10. Massi, D. et al. The density and spatial tissue distribution of CD8(+) and CD163(+) immune cells predict response and outcome in melanoma patients receiving MAPK inhibitors. J Immunother Cancer 7, 308 (2019). [https://doi.org:10.1186/s40425-019-0797-4](https://doi.org:10.1186/s40425-019-0797-4) 11. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 24, 541–550 (2018). [https://doi.org:10.1038/s41591-018-0014-x](https://doi.org:10.1038/s41591-018-0014-x) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-018-0014-x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 12. Chuah, S. & Chew, V. High-dimensional immune-profiling in cancer: implications for immunotherapy. J Immunother Cancer 8 (2020). [https://doi.org:10.1136/jitc-2019-000363](https://doi.org:10.1136/jitc-2019-000363) 13. Gide, T. N. et al. Close proximity of immune and tumor cells underlies response to anti-PD-1 based therapies in metastatic melanoma patients. Oncoimmunology 9, 1659093 (2020). [https://doi.org:10.1080/2162402x.2019.1659093](https://doi.org:10.1080/2162402x.2019.1659093) 14. Sade-Feldman, M. et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 175, 998–1013.e1020 (2018). [https://doi.org:10.1016/j.cell.2018.10.038](https://doi.org:10.1016/j.cell.2018.10.038) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2018.10.038&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30388456&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 15. van der Leun, A. M., Thommen, D. S. & Schumacher, T. N. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat Rev Cancer (2020). [https://doi.org:10.1038/s41568-019-0235-4](https://doi.org:10.1038/s41568-019-0235-4) 16. Rojahn, T. B. et al. Single-cell transcriptomics combined with interstitial fluid proteomics defines cell type-specific immune regulation in atopic dermatitis. J Allergy Clin Immunol 146, 1056–1069 (2020). [https://doi.org:10.1016/j.jaci.2020.03.041](https://doi.org:10.1016/j.jaci.2020.03.041) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2020.03.041&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32344053&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 17. Wen, T. et al. Single-cell RNA sequencing identifies inflammatory tissue T cells in eosinophilic esophagitis. J Clin Invest 129, 2014–2028 (2019). [https://doi.org:10.1172/jci125917](https://doi.org:10.1172/jci125917) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 18. Pfannenstiel, L. W. et al. Immune-Checkpoint Blockade Opposes CD8(+) T-cell Suppression in Human and Murine Cancer. Cancer Immunol Res 7, 510–525 (2019). [https://doi.org:10.1158/2326-6066.CIR-18-0054](https://doi.org:10.1158/2326-6066.CIR-18-0054) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjc6IjcvMy81MTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. Maybruck, B. T., Pfannenstiel, L. W., Diaz-Montero, M. & Gastman, B. R. Tumor-derived exosomes induce CD8(+) T cell suppressors. J Immunother Cancer 5, 65 (2017). [https://doi.org:10.1186/s40425-017-0269-7](https://doi.org:10.1186/s40425-017-0269-7) 20. Ahmadzadeh, M. et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 (2009). [https://doi.org:10.1182/blood-2008-12-195792](https://doi.org:10.1182/blood-2008-12-195792) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImJsb29kam91cm5hbCI7czo1OiJyZXNpZCI7czoxMDoiMTE0LzgvMTUzNyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA0LzAzLzIwMjMuMDQuMDIuMjMyODgwNDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 21. Saeidi, A. et al. T-Cell Exhaustion in Chronic Infections: Reversing the State of Exhaustion and Reinvigorating Optimal Protective Immune Responses. Front Immunol 9, 2569 (2018). [https://doi.org:10.3389/fimmu.2018.02569](https://doi.org:10.3389/fimmu.2018.02569) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2018.02569&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30473697&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 22. Singer, M. et al. A Distinct Gene Module for Dysfunction Uncoupled from Activation in Tumor-Infiltrating T Cells. Cell 166, 1500–1511 e1509 (2016). [https://doi.org:10.1016/j.cell.2016.08.052](https://doi.org:10.1016/j.cell.2016.08.052) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2016.08.052&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27610572&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 23. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018). [https://doi.org:10.1038/s41586-018-0694-x](https://doi.org:10.1038/s41586-018-0694-x) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 24. Jie, H. B. et al. Increased PD-1(+) and TIM-3(+) TILs during Cetuximab Therapy Inversely Correlate with Response in Head and Neck Cancer Patients. Cancer Immunol Res 5, 408–416 (2017). [https://doi.org:10.1158/2326-6066.CIR-16-0333](https://doi.org:10.1158/2326-6066.CIR-16-0333) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiY2FuaW1tIjtzOjU6InJlc2lkIjtzOjc6IjUvNS80MDgiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 25. Shayan, G. et al. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology 6, e1261779 (2017). [https://doi.org:10.1080/2162402X.2016.1261779](https://doi.org:10.1080/2162402X.2016.1261779) 26. Koch, A. et al. Characterization of glycolysis-related gene expression in malignant melanoma. Pathol Res Pract 216, 152752 (2020). [https://doi.org:10.1016/j.prp.2019.152752](https://doi.org:10.1016/j.prp.2019.152752) 27. Van der Sluis, R. M. et al. Combination Immune Checkpoint Blockade to Reverse HIV Latency. J Immunol 204, 1242–1254 (2020). [https://doi.org:10.4049/jimmunol.1901191](https://doi.org:10.4049/jimmunol.1901191) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjIwNC81LzEyNDIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 28. Li, T. & Chiang, J. Y. Bile acid signaling in metabolic disease and drug therapy. Pharmacol Rev 66, 948–983 (2014). [https://doi.org:10.1124/pr.113.008201](https://doi.org:10.1124/pr.113.008201) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoicGhhcm1yZXYiO3M6NToicmVzaWQiO3M6ODoiNjYvNC85NDgiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 29. Fiorucci, S., Biagioli, M., Zampella, A. & Distrutti, E. Bile Acids Activated Receptors Regulate Innate Immunity. Front Immunol 9, 1853 (2018). [https://doi.org:10.3389/fimmu.2018.01853](https://doi.org:10.3389/fimmu.2018.01853) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2018.01853&link_type=DOI) 30. Biagioli, M. et al. The Bile Acid Receptor GPBAR1 Regulates the M1/M2 Phenotype of Intestinal Macrophages and Activation of GPBAR1 Rescues Mice from Murine Colitis. J Immunol 199, 718–733 (2017). [https://doi.org:10.4049/jimmunol.1700183](https://doi.org:10.4049/jimmunol.1700183) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6OToiMTk5LzIvNzE4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDQvMDMvMjAyMy4wNC4wMi4yMzI4ODA0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 31. Hunt, M. C. et al. The peroxisome proliferator-activated receptor alpha (PPARalpha) regulates bile acid biosynthesis. J Biol Chem 275, 28947–28953 (2000). [https://doi.org:10.1074/jbc.M002782200](https://doi.org:10.1074/jbc.M002782200) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyNzUvMzcvMjg5NDciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 32. Li, F., Patterson, A. D., Krausz, K. W., Tanaka, N. & Gonzalez, F. J. Metabolomics reveals an essential role for peroxisome proliferator-activated receptor alpha in bile acid homeostasis. J Lipid Res 53, 1625–1635 (2012). [https://doi.org:10.1194/jlr.M027433](https://doi.org:10.1194/jlr.M027433) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamxyIjtzOjU6InJlc2lkIjtzOjk6IjUzLzgvMTYyNSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA0LzAzLzIwMjMuMDQuMDIuMjMyODgwNDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 33. Savkur, R. S. et al. Ligand-dependent coactivation of the human bile acid receptor FXR by the peroxisome proliferator-activated receptor gamma coactivator-1alpha. J Pharmacol Exp Ther 312, 170–178 (2005). [https://doi.org:10.1124/jpet.104.072124](https://doi.org:10.1124/jpet.104.072124) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoianBldCI7czo1OiJyZXNpZCI7czo5OiIzMTIvMS8xNzAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 34. Chiang, J. Y. Regulation of bile acid synthesis. Front Biosci 3, d176–193 (1998). [https://doi.org:10.2741/a273](https://doi.org:10.2741/a273) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9450986&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 35. Shahoei, S. H. & Nelson, E. R. Nuclear receptors, cholesterol homeostasis and the immune system. J Steroid Biochem Mol Biol 191, 105364 (2019). [https://doi.org:10.1016/j.jsbmb.2019.04.013](https://doi.org:10.1016/j.jsbmb.2019.04.013) 36. DuSell, C. D., Umetani, M., Shaul, P. W., Mangelsdorf, D. J. & McDonnell, D. P. 27-hydroxycholesterol is an endogenous selective estrogen receptor modulator. Mol Endocrinol 22, 65–77 (2008). [https://doi.org:10.1210/me.2007-0383](https://doi.org:10.1210/me.2007-0383) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/me.2007-0383&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17872378&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251994200007&link_type=ISI) 37. Tian, W. et al. Hepatocyte-generated 27-hydroxycholesterol promotes the growth of melanoma by activation of estrogen receptor alpha. J Cell Biochem 119, 2929–2938 (2018). [https://doi.org:10.1002/jcb.26498](https://doi.org:10.1002/jcb.26498) 38. Nelson, E. R. et al. 27-Hydroxycholesterol links hypercholesterolemia and breast cancer pathophysiology. Science 342, 1094–1098 (2013). [https://doi.org:10.1126/science.1241908](https://doi.org:10.1126/science.1241908) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNDIvNjE2Mi8xMDk0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDQvMDMvMjAyMy4wNC4wMi4yMzI4ODA0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 39. Abdalkareem Jasim, S., et al. The emerging role of 27-hydroxycholesterol in cancer development and progression: An update. Int Immunopharmacol 110, 109074 (2022). [https://doi.org:10.1016/j.intimp.2022.109074](https://doi.org:10.1016/j.intimp.2022.109074) 40. Ma, L. et al. 27-Hydroxycholesterol acts on myeloid immune cells to induce T cell dysfunction, promoting breast cancer progression. Cancer Lett 493, 266–283 (2020). [https://doi.org:10.1016/j.canlet.2020.08.020](https://doi.org:10.1016/j.canlet.2020.08.020) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.canlet.2020.08.020&link_type=DOI) 41. Shi, Z., Jiao, S. & Zhou, Z. STRIPAK complexes in cell signaling and cancer. Oncogene 35, 4549–4557 (2016). [https://doi.org:10.1038/onc.2016.9](https://doi.org:10.1038/onc.2016.9) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/onc.2016.9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26876214&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 42. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018). [https://doi.org:10.1126/science.aan4236](https://doi.org:10.1126/science.aan4236) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjExOiIzNTkvNjM3MS85NyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA0LzAzLzIwMjMuMDQuMDIuMjMyODgwNDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 43. Lu, Y. et al. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J Hematol Oncol 15, 47 (2022). [https://doi.org:10.1186/s13045-022-01273-9](https://doi.org:10.1186/s13045-022-01273-9) 44. Fessler, J., Matson, V. & Gajewski, T. F. Exploring the emerging role of the microbiome in cancer immunotherapy. J Immunother Cancer 7, 108 (2019). [https://doi.org:10.1186/s40425-019-0574-4](https://doi.org:10.1186/s40425-019-0574-4) 45. Bhatt, A. P., Redinbo, M. R. & Bultman, S. J. The role of the microbiome in cancer development and therapy. CA Cancer J Clin 67, 326–344 (2017). [https://doi.org:10.3322/caac.21398](https://doi.org:10.3322/caac.21398) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3322/caac.21398&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 46. Chen, Y., Wu, F. H., Wu, P. Q., Xing, H. Y. & Ma, T. The Role of The Tumor Microbiome in Tumor Development and Its Treatment. Front Immunol 13, 935846 (2022). [https://doi.org:10.3389/fimmu.2022.935846](https://doi.org:10.3389/fimmu.2022.935846) 47. Boesch, M. et al. Compartmentalization of the host microbiome: how tumor microbiota shapes checkpoint immunotherapy outcome and offers therapeutic prospects. J Immunother Cancer 10 (2022). [https://doi.org:10.1136/jitc-2022-005401](https://doi.org:10.1136/jitc-2022-005401) 48. Zhu, G. et al. Intratumour microbiome associated with the infiltration of cytotoxic CD8+ T cells and patient survival in cutaneous melanoma. Eur J Cancer 151, 25–34 (2021). [https://doi.org:10.1016/j.ejca.2021.03.053](https://doi.org:10.1016/j.ejca.2021.03.053) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 49. Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020). [https://doi.org:10.1126/science.aay9189](https://doi.org:10.1126/science.aay9189) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNjgvNjQ5NC85NzMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 50. Helmink, B. A., Khan, M. A. W., Hermann, A., Gopalakrishnan, V. & Wargo, J. A. The microbiome, cancer, and cancer therapy. Nat Med 25, 377–388 (2019). [https://doi.org:10.1038/s41591-019-0377-7](https://doi.org:10.1038/s41591-019-0377-7) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-019-0377-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30842679&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 51. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018). [https://doi.org:10.1126/science.aan3706](https://doi.org:10.1126/science.aan3706) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjExOiIzNTkvNjM3MS85MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA0LzAzLzIwMjMuMDQuMDIuMjMyODgwNDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 52. Yamada, S. et al. Bile acid metabolism regulated by the gut microbiota promotes non-alcoholic steatohepatitis-associated hepatocellular carcinoma in mice. Oncotarget 9, 9925–9939 (2018). [https://doi.org:10.18632/oncotarget.24066](https://doi.org:10.18632/oncotarget.24066) 53. Cai, K. & Sewer, M. B. Diacylglycerol kinase theta couples farnesoid X receptor-dependent bile acid signalling to Akt activation and glucose homoeostasis in hepatocytes. Biochem J 454, 267–274 (2013). [https://doi.org:10.1042/BJ20130609](https://doi.org:10.1042/BJ20130609) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6InBwYmlvY2hlbWoiO3M6NToicmVzaWQiO3M6OToiNDU0LzIvMjY3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDQvMDMvMjAyMy4wNC4wMi4yMzI4ODA0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 54. Mann, T. H. & Kaech, S. M. Tick-TOX, it’s time for T cell exhaustion. Nat Immunol 20, 1092–1094 (2019). [https://doi.org:10.1038/s41590-019-0478-y](https://doi.org:10.1038/s41590-019-0478-y) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41590-019-0478-y&link_type=DOI) 55. Welinder, C. et al. Correlation of histopathologic characteristics to protein expression and function in malignant melanoma. PLoS One 12, e0176167 (2017). [https://doi.org:10.1371/journal.pone.0176167](https://doi.org:10.1371/journal.pone.0176167) 56. Zhao, X., Fu, J., Du, J. & Xu, W. The Role of D-3-Phosphoglycerate Dehydrogenase in Cancer. Int J Biol Sci 16, 1495–1506 (2020). [https://doi.org:10.7150/ijbs.41051](https://doi.org:10.7150/ijbs.41051) 57. Locasale, J. W. et al. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat Genet 43, 869–874 (2011). [https://doi.org:10.1038/ng.890](https://doi.org:10.1038/ng.890) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.890&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21804546&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 58. Ma, J. et al. PD1(Hi) CD8(+) T cells correlate with exhausted signature and poor clinical outcome in hepatocellular carcinoma. J Immunother Cancer 7, 331 (2019). [https://doi.org:10.1186/s40425-019-0814-7](https://doi.org:10.1186/s40425-019-0814-7) 59. Boldajipour, B., Nelson, A. & Krummel, M. F. Tumor-infiltrating lymphocytes are dynamically desensitized to antigen but are maintained by homeostatic cytokine. JCI Insight 1, e89289 (2016). [https://doi.org:10.1172/jci.insight.89289](https://doi.org:10.1172/jci.insight.89289) 60. Peretz, Y. et al. CD160 and PD-1 co-expression on HIV-specific CD8 T cells defines a subset with advanced dysfunction. PLoS Pathog 8, e1002840 (2012). [https://doi.org:10.1371/journal.ppat.1002840](https://doi.org:10.1371/journal.ppat.1002840) 61. Falus, A. et al. Paracrine and autocrine interactions in melanoma: histamine is a relevant player in local regulation. Trends Immunol 22, 648–652 (2001). [https://doi.org:10.1016/s1471-4906(01)02050-6](https://doi.org:10.1016/s1471-4906(01)02050-6) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1471-4906(01)02050-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11738976&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 62. Azimi, A. et al. Targeting CDK2 overcomes melanoma resistance against BRAF and Hsp90 inhibitors. Mol Syst Biol 14, e7858 (2018). [https://doi.org:10.15252/msb.20177858](https://doi.org:10.15252/msb.20177858) 63. Singh, C. K. et al. Novel downstream molecular targets of SIRT1 in melanoma: a quantitative proteomics approach. Oncotarget 5, 1987–1999 (2014). [https://doi.org:10.18632/oncotarget.1898](https://doi.org:10.18632/oncotarget.1898) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.18632/oncotarget.1898&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24743044&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 64. Rouaud, F. et al. E2F1 inhibition mediates cell death of metastatic melanoma. Cell Death Dis 9, 527 (2018). [https://doi.org:10.1038/s41419-018-0566-1](https://doi.org:10.1038/s41419-018-0566-1) 65. Rabenstein, H. et al. Differential kinetics of antigen dependency of CD4+ and CD8+ T cells. J Immunol 192, 3507–3517 (2014). [https://doi.org:10.4049/jimmunol.1302725](https://doi.org:10.4049/jimmunol.1302725) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiamltbXVub2wiO3M6NToicmVzaWQiO3M6MTA6IjE5Mi84LzM1MDciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 66. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016). [https://doi.org:10.1126/science.aad0501](https://doi.org:10.1126/science.aad0501) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNTIvNjI4Mi8xODkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wNC8wMy8yMDIzLjA0LjAyLjIzMjg4MDQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 67. Li, C. et al. A high OXPHOS CD8 T cell subset is predictive of immunotherapy resistance in melanoma patients. J Exp Med 219 (2022). [https://doi.org:10.1084/jem.20202084](https://doi.org:10.1084/jem.20202084) 68. Jiang, H., Lei, R., Ding, S. W. & Zhu, S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics 15, 182 (2014). [https://doi.org:10.1186/1471-2105-15-182](https://doi.org:10.1186/1471-2105-15-182) 69. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol 37, 907–915 (2019). [https://doi.org:10.1038/s41587-019-0201-4](https://doi.org:10.1038/s41587-019-0201-4) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41587-019-0201-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31375807&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 70. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009). [https://doi.org:10.1093/bioinformatics/btp352](https://doi.org:10.1093/bioinformatics/btp352) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btp352&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19505943&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000268808600014&link_type=ISI) 71. Hu, Y. et al. Bile acids regulate nuclear receptor (Nur77) expression and intracellular location to control proliferation and apoptosis. Mol Cancer Res 13, 281–292 (2015). [https://doi.org:10.1158/1541-7786.MCR-14-0230](https://doi.org:10.1158/1541-7786.MCR-14-0230) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibW9sY2FucmVzIjtzOjU6InJlc2lkIjtzOjg6IjEzLzIvMjgxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDQvMDMvMjAyMy4wNC4wMi4yMzI4ODA0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 72. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43, e47 (2015). [https://doi.org:10.1093/nar/gkv007](https://doi.org:10.1093/nar/gkv007) 73. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013). [https://doi.org:10.1186/1471-2105-14-7](https://doi.org:10.1186/1471-2105-14-7) 74. Li, H. et al. Dysfunctional CD8 T Cells Form a Proliferative, Dynamically Regulated Compartment within Human Melanoma. Cell 176, 775–789 e718 (2019). [https://doi.org:10.1016/j.cell.2018.11.043](https://doi.org:10.1016/j.cell.2018.11.043) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 75. Stuart, T. et al. Comprehensive Integration of Single-Cell Data. Cell 177, 1888–1902 e1821 (2019). [https://doi.org:10.1016/j.cell.2019.05.031](https://doi.org:10.1016/j.cell.2019.05.031) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2019.05.031&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F04%2F03%2F2023.04.02.23288048.atom) 76. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545–15550 (2005). [https://doi.org:10.1073/pnas.0506580102](https://doi.org:10.1073/pnas.0506580102) [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTAyLzQzLzE1NTQ1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDQvMDMvMjAyMy4wNC4wMi4yMzI4ODA0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=)