High expression of NAT10 correlates with poor survival and low immune cell infiltration in cancer
NAT10, the only writer for ac4C modification on mRNAs, has been reported to have many important functions, such as affecting stem cells differentiation, promoting glycolysis addiction. Additionally, the potential role of NAT10 in pro-tumor effect has been uncovered, but the anti-tumor immunity of cancer-intrinsic NAT10 has not been reported.
To determine the role of NAT10, we utilized GEPIA database to explore the influence of NAT10 in lung adenocarcinoma (LUAD) [16]. The expression of NAT10 was increased in TCGA-LUAD cohort with the stage increase of the disease (Fig. 1A), which indicated that NAT10 was related with the progression of cancer. Moreover, we download the raw data and survival information from TCGA-LUAD cohort to obtain a Kaplan-Meier survival curve. The results showed that higher NAT10 expression was correlated significantly with shorter overall survival (Fig. 1B). Concurrently, we collected 38 pathology slides of lung cancer patients and performed immuno-histochemical staining to obtain immuno-histochemistry stain score for survival analysis. The results was in accordance with TCGA database, as shown by higher NAT10 expression with lower survival (Fig. 1C). Moreover, ROC curve was utilized to validate the ability of the prognostic efficiency of NAT10, which indicated that the NAT10 has a good predictive value in lung cancer (Fig. 1D). Altogether, these results suggest that NAT10 is a cancer-promoting gene, which may be an important risk factor for the development of lung cancer.
Given our findings above, we further explored the oncogenic effect of NAT10 in vivo. Remodelin, a well-established inhibitor for NAT10 [17], was used for the further study. The immunocompetent (C57BL/6N) mice were used to establish syngeneic tumor models. Mice were injected subcutaneously with murine lung cancer cells (TC1) or murine fibrosarcoma cells (MCA205). The tumor masses on the flank of the mice indicated the successful establishment of the tumor after 14 days. Importantly, the tumors in mice exposed to low-dose Remodelin were significantly reduced in weight and size compared to the saline group (Fig. 1E, 1F). On the other hand, Immunodeficient (Nude/Nude) mice injected subcutaneously with TC1 or MCA205 cells were established. Our results showed that there was no difference in tumor size and weight whether exposed to saline or low-dose Remodelin in the nude mice (Fig. 1G, 1H). Additionally, we further explored the effect of NAT10 on cancer cell in vitro. Colony formation assay was conducted to detect the role of NAT10 in the cell proliferation. The results showed that Remodelin significantly suppressed the proliferation of cancer cells (Fig. S1A, S1B). The above results indicated that the effect of NAT10 on tumor growth was partly related to host immunity.
Analysis of immune cell infiltration was performed using the CIBERSORT algorithm between high and low NAT10 expression groups in TCGA-LUAD cohort [18]. The results showed that LUAD patients with lower expression of NAT10 appeared to have higher proportions of immune cells, include CD8+ T cells and DCs (Fig. 1I). The expression analysis of NAT10 within individual patients was conducted based on the scRNAseq data (GSE148071) [19], whereby the summation of NAT10 expression levels across all cells within each patient was followed by division by the total number of sequenced cells within the same patient. The results also indicated a positive relationship between NAT10 expression and malignant cells percentage while a negative correlation with the proportion of T cells or DCs (Fig. S1C). Furthermore, our immunohistochemical staining results in lung cancer samples (n = 37) showed the same negative association between NAT10 and CD8+ T cells (Fig. 1J). Collectively, these results suggest that NAT10 is a proto-oncogene and maybe affect tumor growth in an immune-dependent manner.
NAT10 deficiency suppresses tumor growth via eliciting immunological protection
Given the observed phenomenon, it is imperative to further investigate the mechanisms underlying NAT10 in anti-tumor immunity. CRISPR/Cas9 technology utilizing Nat10-specific sgRNA (sgNAT10) pairs was employed to knockout NAT10 in TC1 and MCA205 cells (Figs. 2A, S2A). Wild-type (WT) cells transfected with an empty vector served as controls. To evaluate whether NAT10 inhibition within cancer cells triggers immune responses, we established syngeneic tumor models in immunocompetent (C57BL/6N) mice and immunodeficient (Nude/Nude) mice transplanted with either WT or sgNAT10 cancer cells. All C57BL/6N mice bearing with WT TC1 or MCA205 cells had developed substantial tumor masses post-transplantation, whereas the tumor masses disappeared quickly in those bearing with sgNAT10 TC1 or MCA205 cells (Fig. 2A, 2B), indicating that NAT10 deficiency might impede subcutaneous tumor growth in immunocompetent mice. Subsequently, we established transplant tumor models in immunocompetent Nude mice. The results demonstrated that Nude mice in both WT and sgNAT10 groups developed apparently substantial tumors though with a significant difference (Fig. 2C, 2D), further suggesting that NAT10 deficiency may suppress tumors in an immune-dependent manner. As the mice receiving transplants of NAT10-deficient TC1 and MCA205 cells exhibited slightly smaller tumors compared to those with WT cells in Nude/Nude mice, we speculated that NAT10 might have additional suppressive effects on tumor cell proliferation besides of eliciting adaptive immune responses. Consequently, we performed colony formation assays and CCK8 assays. Our findings demonstrated that NAT10 deficiency significantly impeded colony formation (Figs. S2B, S2C) and proliferation (Figs. S2D, S2E) in both TC1 and MCA205 cells.
Moreover, we further investigated whether NAT10 could influence the survival via the host immune system. C57BL/6N mice bearing NAT10-deficient TC1 or MCA205 cell transplants exhibited significantly prolonged survival compared to mice with WT cell transplants; however, they succumbed to the tumor within 60 days. In contrast, all C57BL/6N mice receiving NAT10-deficient cancer cell transplants survived until day 60 (Fig. 2E, 2F). In the xenograft model using Nude/Nude mice, no significant difference of survival between WT and sgNAT10 groups was found and all mice died within 40 days (Fig. 2G, 2H). These differing outcomes between immunocompetent and immunodeficient mice further support the notion that NAT10 inhibition may enhance mouse survival by activating immunity.
Considering the possible nonspecific effects of the CRISPR/Cas9 system, we restored NAT10 expression in the NAT10-deficient TC1 cells. We first constructed a plasmid VP64-NAT10-GFP, in which the base sequence corresponding to the NAT10 sgRNA position in the VP64-NAT10-GFP plasmid was modified, with the encoded amino acids unchanged to avoid cleavage by the CRISPR CAS9 enzyme. Our findings demonstrated that NAT10 restoration enabled NAT10-deficient TC1 cells to successfully develop tumors (Fig. S2F), providing clear evidence of the pro-oncogene effect of NAT10.
For the assessment of possible involved immune memory, we subcutaneously immunized C57BL/6N mice with either sgNAT10 cancer cells or freeze-thawed WT cancer cells on the left side, followed by re-challenge with comparable numbers of live WT cancer cells on the right side after 2 weeks (Fig. 2I, left panel). Intriguingly, each mouse immunized with sgNAT10 cancer cells completely inhibited WT tumor growth on the right side, resulting in tumor-free mice, whereas those immunized with freeze-thawed WT cancer cells showed a significantly weaker effect (Fig. 2I). These results suggested that NAT10 deficiency elicited immunological protection.
NAT10 deficiency triggers immune responses of CD8 + T cells in vivo
Based on our previous findings suggesting that NAT10 deficiency may impede tumor growth by activating anti-immune mechanisms, we conducted transcriptomic RNA-sequencing (RNA-seq) analysis to comprehensively investigate whether NAT10 has impacts on immune-response signaling in vivo. Tumor tissues inoculated with WT or sgNAT10 cancer cells were harvested on day 8, and total RNA was extracted for RNA sequencing. Gene Set Enrichment Analysis (GSEA) revealed upregulation of "hallmark" signatures including "Interferon-gamma (IFN-γ) response", "Interferon-alpha (IFN-α) response", and "Inflammatory response" in sgNAT10 TC1 tumor tissues [20]. Heatmaps depicting differentially regulated genes from the GSEA analysis in WT and sgNAT10 TC1 tumor tissues showed increased expression of numerous cytokines and chemokines, such as C-X-C motif chemokine ligands 9, 10 and 11 (CXCL9, 10, 11) (Fig. 3A), which contribute to robust anti-tumor immunity. CXCL9/10/11 are responsible for recruiting and activating T cells via binding with CXCR3 [21]. Moreover, our results found that genes associated with the antigen presentation machinery (APM) and CD8+ Teff were upregulated in sgNAT10 tumor tissues (Fig. 3B), while cell cycle-related genes linked to proliferation were downregulated (Fig. 3C). Collectively, these findings indicate that NAT10 deficiency plays a crucial role in anti-tumor immunity through regulating immunological response factors especially genes associated with CD8+ Teff cells.
To further clarify the detailed immune cells involved in NAT10 deficiency-induced anti-tumor immunity, we applied multicolor immunofluorescence experiments. Our results demonstrated a notable elevation in CD8+ T cells and DCs in sgNAT10 group compared to the WT group, while no noticeable difference was initially observed in Tregs (Fig. 3D). Considering that CD8+ T cells play a crucial role in anti-tumor immunity, we conducted antibody-based depletion of CD8+ T cells prior to in vivo transplantation of sgNAT10 TC1 cancer cells. The results showed that depletion of CD8+ T cells markedly impeded NAT10-deficient-induced tumor regression, suggesting deletion of NAT10 primarily exerts anti-tumor immune effects via CD8+ T cells (Fig. 3E).
Given that CD8 antibodies reversed the protective effect of NAT10 deficiency, we further investigated CD8+ T cell infiltration and functionality. Our results of immunofluorescence assay revealed increased tumor-infiltrating CD8+ T cells in NAT10-deficient tumor tissues (Fig. S3A). Moreover, flow cytometry results quantified higher CD8+ T cell frequencies in NAT10-deficient tumor tissues, consistent with immunofluorescence results (Fig. S3B). For the functional assessment, we further demonstrated the elevated IFN-γ and Granzyme B (GZMB) levels in tumor-infiltrating CD8+ T cells in the sgNAT10 tumor group (Fig. 3F, 3G). Importantly, inguinal lymph nodes, critical for anti-tumor immunity, were also used in our study to investigate the CD8+ T cells. Our results also showed the upregulation of IFN-γ+CD8+ T cells in the NAT10-deficient group (Fig. S3C, S3D). Additionally, gene expression analysis by real-time PCR method confirmed the upregulation of CD8a, IFN-γ, Granzyme A (GZMA), GZMB, CXCL9, and CXCL10 in sgNAT10 group (Fig. 3H). IFN-γ secretion was increased in the sgNAT10 group, as observed in IFN-γ ELISpot assay (Fig. 3I). Moreover, T-cell proliferation assay indicated enhanced proliferation of both CD4+ and CD8+ T cells in NAT10-deficient cancer cells (Fig. 3J, 3K and S3E). These findings collectively suggest adaptive immune responses, particularly CD8+ T cell-mediated antitumor immunity, have been activated induced by NAT10 deficiency in vivo.
NAT10 deficiency induces IFN-I responses in cancer cells
The above results showing an enhanced IFN response and an increased infiltration of tumor-infiltrating lymphocytes (TIL) in the NAT10 deficient tumor microenvironment suggest a possible link between IFN-mediated tumor cell chemokine expression and increased TIL infiltration, which may be responsible for the enhanced antitumor immune responses. To test this hypothesis, RNA-seq was performed with total mRNA extracted from WT and sgNAT10 TC1 or MCA205 cells. GSEA analysis showed that these pathways were mainly involved in the “IFN-I” signaling pathway (Fig. S4A, S4B). Compared to WT cancer cells, NAT10 deletion induced the expression of genes related to IFN-I response (Fig. 4A, 4B). By RT-qPCR analysis, we further confirmed the increased expression of some of these genes in sgNAT10 cancer cells, including the type I IFN gene Ifnb1 itself, the transcription factor Stat1, the antiviral gene Mx2, the pattern recognition receptor genes Tlr3 and Ddx58, the antigen presentation related gene Tap1, as well as the chemokine-encoding genes Ccl5 and Ccl7 (Fig. 4C, 4D).
NAT10 deficiency could induce IFN-I responses in cancer cells, which can play key roles in the activation of cellular components of the immune response, such as dendritic cells and T cells. To verify that IFN-I responses underlined the outcomes, sgNAT10 cancer cells were transplanted into type I IFN receptor KO (Ifnar1 KO) mice. The results showed that both WT and sgNAT10 cancer cells developed apparently substantial tumors in Ifnar1 KO mice, suggesting the effects favoring anti-tumor immune responses triggered by NAT10 deficiency were significantly abolished on an Ifnar1 KO background (Fig. 4E, 4F). These data indicated that NAT10 deficiency in cancer cells may drive IFN-I responses to promote protective anti-tumor CD8+ T cell immunity.
NAT10 increases MYC expression through regulating mRNA acetylation
Next, we explore the mechanism by which NAT10 deletion induces interferon production. To identify whether the acetyltransferase NAT10 directly mediated antitumor immune response, acRIP-seq analysis was performed. The sequential analysis of ac4C peaks showed that typical GAGGAGA motifs were highly enriched within ac4C sites of mRNA (Fig. 5A). Further analytic results showed that the ac4C peaks predominantly occurred within coding sequences (CDS) and 3’untranslated regions (3’UTR) (Fig. 5B, 5C). As reported, the acetyltransferase NAT10 can confer enhanced mRNA stability, and ac4C peaks within wobble sites can stimulate translation efficiency [22]. We therefore investigated potential targets using a combination of acRIP-seq and Label-free quantitative proteomics. We identified 7 candidate genes (Phf2, Myc, Wwc2, Kmt2a, Gigyf1, Timeless, and Nufip2) that showed concomitant decreased mRNA acetylation and reduced protein levels in sgNAT10 cancer cells (Fig. 5D, S5A).
Among the 7 candidate genes, MYC has been reported to be related to both cell proliferation and antitumor immunity [23]. We then performed Western blot, and our results showed that NAT10 deficiency resulted in decreased protein expression of MYC in cancer cells (Fig. 5E). To identify the key ac4C sites that regulate mRNA stability, we further analyzed the acetylation peaks of MYC mRNA. AcRIP-seq data showed that the ac4C peaks were distributed in the CDS and 3’/5’UTR region of MYC mRNA (Fig. 5F). Interestingly, the 3’UTR region of MYC mRNA contains a nucleic acid sequence consistent with the typical GAGGAGA motifs (Fig. 5A), suggesting that this ac4C site may be more dynamic in regulating MYC mRNA stability. Subsequently, we constructed 3’UTR reporters containing wild type or mutant MYC 3’UTR after the firefly luciferase reporter gene (Fig. 5G). The dual-luciferase assay showed significantly attenuated fluorescence activity in the mut-3’UTR groups compared to WT-3’UTR groups, mirroring reduced mRNA stability due to the loss of acetylated position (Fig. 5H). Moreover, the acRIP-PCR results confirmed that NAT10 may bind to the 3'UTR of MYC (Fig. 5I). Furthermore, our results also showed that the half-life of MYC mRNA was ≈ 16 hours for WT cells and significantly decreased in sgNAT10 cells, meaning reduced ac4C enrichment was accompanied by increased decay of MYC mRNA (Fig. 5J). Overall, NAT10 promoted MYC mRNA stability and translation efficiency via ac4C modification, and the ac4C peak within the 3’UTR region was responsible for mRNA stability.
Considering the important expression-regulating role of NAT10 on MYC, we investigate whether NAT10 modulates anti-tumor immunity via MYC. Firstly, CRISPR/Cas9 technology utilizing MYC-specific sgRNA (sgMYC) pairs was employed to knockout MYC in TC1 (Fig. S5B). To evaluate whether intrinsic MYC deficiency inhibits tumor growth by triggering an immune response, we established syngeneic tumor models in C57BL/6N mice transplanted with either WT or sgMYC cancer cells. The results showed that sgMYC TC-1 tumors exhibited a significant reduction in tumor growth as compared with their WT parental cells (Fig. S5C). At day 10 after subcutaneous transplantation, a considerably higher percentage of CD8+ T cells were observed in sgMYC tumors than in WT tumors (Fig. S5D), indicating that adaptive immunity might be involved in MYC-deficient induced tumor reduction. The MYC protein restored in sgNAT10 TC1 cells was significantly abolished NAT10-deficient induced tumor regression (Fig. S5E, S5F). The elevated IFN-γ secretion induced by NAT10 deficiency in vivo were also significantly abolished in Myc-overexpressed sgNAT10 cells (Fig. S5G). These data suggest that NAT10 might modulate anti-tumor immunity via regulating MYC expression.
NAT10 depletion induces dsRNA-mediated RIG-I-dependent IFN-I signaling via Myc/CDK2/DNMT1 pathway
As shown above that NAT10 enhanced mRNA stability and translation efficiency of MYC, we then aimed to elucidate IFN-I signaling induced by NAT10 inhibition. Considering the ability of NAT10 to promote cell proliferation, we reanalyzed the RNA-seq data and revealed several differentially-expressed genes associated with proliferation, in which CDK2, a member of the cyclin-dependent kinases family [24], was the most significantly down-regulated in NAT10 deficient cells (Fig. 6A, S6A). It has been reported that MYC could directly regulate CDK2 expression [25], and our western blot results also showed that MYC deletion significantly inhibited the expression of CDK2 in cancer cells (Fig. S6B). Moreover, our results showed that knocking out CDK2 (sgCDK2) led to the inhibition of tumor growth (Fig. S6D), consistent with the effect of siNAT10. Next, we explored the effects of CDK2 on antitumor immune response. The results showed that CDK2 deficiency in cancer cell elevated CD8+ T cells infiltration and IFN-γ expression, which is consistent with the phenomenon caused by NAT10 deletion (Fig. S6E, S6F). These findings suggest that NAT10 deficiency might enhance anti-tumor immunity via Myc-mediated regulation of CDK2 expression.
How does CDK2 deletion induce IFN-I responses? CDK2-deficient cells have been proven to inhibit the activity of DNMT, and loss of its activity can induce IFN-I responses by increasing production of dsRNA [26]. We then reanalyzed our RNA-seq data and found that DNMT1 has the highest expression in cancer cells (Fig. 6B, S6C). Western blot analysis also showed that the protein levels of CDK2 and DNMT1 were significantly reduced in NAT10-deficient cells compared to WT cells (Fig. 6C). Furthermore, DNMT1 expression was restored by overexpressing the CDK2 in NAT10 deficient cells (Fig. S6G). More importantly, overexpression of CDK2 in sgNAT10 cells could promote the development of tumors (Fig. S6G). Correlation analysis between NAT10 and several downstream genes performed with the GEPIA website [34] revealed statistically positive correlations between NAT10 and MYC, CDK2, DNMT1 (Fig. S6H). These data suggest the critical role of NAT10 in maintaining the expression of CDK2 and DNMT1 through MYC.
Increased IFN-I response in cancer cells has been shown to occur in response to DNA demethylation caused by 5-azacytidine, which inhibits the activity of DNMT1 [27]. DNMT1 inhibition could trigger IFN-I response by inducing dsRNA. In our study, quantification of dsRNA performed by immunofluorescence using the dsRNA-specific J2 antibody showed a significantly higher abundance of dsRNA within sgNAT10 and sgCDK2 cells than those within WT cells. Restored MYC and CDK2 significantly abolished NAT10 deficiency-induced dsRNA production (Fig. 6D, 6E). It has been reported that dsRNA could be sensed by RIG-I and MDA-5, which triggers IFN-I response [28]. Therefore, our next objective was to investigate whether NAT10 deletion-induced dsRNA production predominantly activates IFN via the RIG-I or MDA-5 signaling pathway. GSEA enrichment analyzed by RNA-seq data showed that “RIG-I like receptor signaling pathway” were upregulated in sgNAT10 cancer cells compared to WT cells (Fig. S6I, S6J). Therefore, we silenced RIG-I in sgNAT10 TC1 cells and assessed the functionality of the IFN-I signaling pathway. Our results showed that deletion of RIG-I partially negated the elevated expressions of IFN stimulated genes (ISGs) induced by NAT10 deletion (Fig. 6F) [29]. Together, our results demonstrate that NAT10 modulates the IFN-I signaling pathway via RIG-I-mediated dsRNA sensing.
Inhibition of NAT10 with Remodelin enhances response to ICIs therapy
The above data indicate that NAT10 deletion enhances intratumoral IFN-I production and T cell infiltration, two biomarkers associated with sensitivity to ICIs therapy. Previous studies also reported that activating the IFN-I pathway and enhancing T cell infiltration could promote the therapeutic effect of ICIs therapy [30]. Therefore, we next investigated cooperation between NAT10 inhibitor and PD-1 treatment using syngeneic tumor models. Mice were gavaged with Remodelin once a day for 7 consecutive days. On day 7, we treated mice with isotype control (vehicle), or anti-PD-1 mAb (10 mg/kg, intraperitoneally (ip), twice a week for 2 weeks), and a humane endpoint was reached in a vehicle group mouse on day 29 (Fig. 7A). The results showed that either PD-1mAb or Remodelin effectively inhibited tumor growth compared to control group. Importantly, the tumor size of the combined treatment was much smaller than either of the other two groups (Fig. 7B, 7C), suggesting combining inhibition of NAT10 and PD-1 synergistically suppresses cancer growth in vivo.
Furthermore, we investigated the immunological changes and our results present with a significantly increased number of tumor-infiltrating CD8+ T cells after single treatment of Remodelin or anti-PD-1 mAb compared to control, while the combination group has the much higher CD8+ T cells infiltration (Fig. 7D, 7E). Moreover, a remarkable increase in the number of IFN-γ-positive active CD8+ T cells was seen in the Remodelin single treatment group; the effects were significantly enhanced by the combination treatment (Fig. 7F, 7G). Importantly, the combination treatment secreted more IFN-γ in the tumor microenvironment (Fig. 7H, 7I). Considering the clinical setting, we further detected and analyzed the relationship of NAT10 and PD-L1 in lung cancer samples. Our results demonstrate a positive correlation between the expression levels of PD-L1 and NAT10 (Fig. S7A). Overall, these data suggest that inhibition of NAT10 enhances the efficacy of PD-1 blockade therapy in suppressing tumor growth.
Intratumoral delivery of siNAT10-lipid nanoparticles (LNPs) for cancer immunotherapy
Considering the limited absorption of Remodelin that could mitigate its therapeutic effect on tumors, we developed two commonly-used delivery systems, SM102 and PEI/PC7A nanoparticles, to enhance inhibitory efficiency of NAT10 expression both in vivo and in vitro. SM102, a cationic amino lipid approved for mRNA delivery in the Moderna COVID-19 vaccine, also functions as an ionizable component in LNPs for RNAi-based therapeutics [31]. Additionally, PEI/PC7A nanoparticle, composed of polyethyleneimine (PEI) and a pH-responsive PC7A polymer, is developed for efficient siRNA transfection [32]. The particle size of siNAT10 was determined using dynamic light scattering (DLS, Malvern) and confirmed to be approximately 160 nm (Fig. 8A, S8A). Confocal laser scanning microscopy (CLSM) analysis demonstrated the overlap of fluorescence signals representing lysosomes (red fluorescence) with siRNAs (green fluorescence) within 4 hours. Moreover, a significant amount of green fluorescence was observed outside the lysosomes, indicating the escape of siRNA from the lysosomes (Fig. 8B, S8B). Successful release of siRNA from endosomes and lysosomes indicated the formation of the RNA-induced silencing complex in the cytosol.
RT-qPCR analysis was applied to assess the inhibitory efficiency of nanoparticles on NAT10 expression. The results demonstrated a significant reduction in NAT10 mRNA expression levels with both SM102 and PEI/PC7A/siNAT10 nanoparticles compared to only siNAT10 transfection (Fig. 8C, S8C). Furthermore, our in vivo experiments revealed that PEI/PC7A/siNAT10 has more effective inhibition on tumor growth than SM102 (Fig. S8D, S8E). Consequently, we employed PEI/PC7A/siNAT10 nanoparticles to evaluate its tumor inhibitory effect for the following study.
Moreover, western blotting results further confirmed significant NAT10 protein expression suppression by PEI/PC7A/siNAT10 nanoparticles (Fig. 8D). And intratumoral delivery of PEI/PC7A/siNAT10 nanoparticles treatment significantly reduced TC1 tumor growth in C57/BL6N mice, showing much more superior efficacy compared to Remodelin (Fig. 8E). Subsequently, we combined PEI/PC7A/siNAT10 nanoparticles with ICIs therapy to enhance the effect of inhibiting tumors. Our results demonstrated the effective tumor growth inhibition with both PD-1mAb and PEI/PC7A/siNAT10 nanoparticles, with the combined treatment resulting in much smaller tumor sizes compared to other groups (Fig. 8F). Furthermore, we observed a significant increase in tumor-infiltrating CD8+ T cells following PEI/PC7A/siNAT10 nanoparticle treatment or combination therapy, with the combination group exhibiting substantially higher CD8+ T cell levels (Fig. 8G). Additionally, a notable rise in IFN-γ-positive active CD8+ T cells was observed in the PEI/PC7A/siNAT10 nanoparticle single treatment group, with significantly enhanced effects noted in the combination therapy group (Fig. 8H). Overall, these findings suggest that NAT10 suppression by nanoparticles enhances the therapeutic effects of ICIs in controlling tumor growth.