Graphical abstract graphic file with name fx1.jpg [57]Open in a new tab Highlights * • Immunocyte-tropic LNP-delivered circRNAs form in vivo panCAR (CAR-T/NK/Mφ) cells * • In vivo panCAR effectively inhibits tumor growth and reshapes the TME in mice * • CircRNA vaccines synergistically boost the anti-tumor efficacy of in vivo panCAR * • Depleting NK cells or macrophages impairs the efficacy of in vivo panCAR-VAC __________________________________________________________________ Wang et al. establish circRNA-based in vivo panCAR (CAR-T/NK/Mφ) immunotherapy, effectively inhibiting tumor growth and reprogramming the tumor microenvironment in mice. In combination with vaccination, in vivo panCAR demonstrates a synergistic enhancement of anti-tumor immunity across various mouse models, thereby establishing a framework for the synergistic in vivo panCAR-VAC immunotherapy. Introduction Chimeric antigen receptor (CAR) T cell therapy is an important method of cancer immunotherapy that has successfully treated hematologic malignancies, particularly B lymphomas.[58]^1 However, CAR-T cell therapy still faces several challenges, including high costs, a time-consuming manufacturing process, the necessity of lymphodepletion, and side effects such as cytokine storm and on-target, off-tumor killing.[59]^2^,[60]^3^,[61]^4^,[62]^5 It is important to solve these issues and improve the effectiveness of adoptive CAR-T cell therapy. To address the limitations of conventional adoptive CAR-T cell therapy, shorten preparation and processing times, and decrease treatment costs,[63]^6 researchers recently reported that in vivo delivery of CAR-encoding mRNAs to T cells or macrophages could generate functional CAR-T or CAR-macrophage (CAR-M) to exert tumor-killing function.[64]^7^,[65]^8^,[66]^9^,[67]^10^,[68]^11 This innovative approach has demonstrated efficacy in treating hematologic malignancies and myocarditis in murine models, providing an off-the-shelf immunotherapy strategy with significant potential for therapeutic applications.[69]^7^,[70]^8^,[71]^9^,[72]^10^,[73]^11 Compared to DNA transposase-mediated[74]^7 or lentivirus-based[75]^12^,[76]^13 delivery, mRNA technology provides a significant safety advantage without integrating into the genome. Similar to mRNAs, circular RNAs (circRNAs) are also not integrated into the genome. However, unlike the linear conformation of mRNA, circRNAs are covalently closed circular RNA molecules that are more stable than linear mRNAs in mammalian cells.[77]^14^,[78]^15^,[79]^16 Therefore, circRNA-based in vivo CAR might enable higher and more durable expression of CAR proteins on the cell membrane of functional immune cells than mRNAs, holding the potential to generate more effective killing effects. Recent studies have also shown that the combination therapy of adoptive CAR-T therapy and vaccination could enhance anti-tumor immunity, which might occur through further in vivo activation of reinfused CAR-T cells via the interaction between antigen-presenting cells expressing transmembrane tumor antigens and the reinfused CAR-T cells expressing CAR molecules against the corresponding antigens.[80]^17^,[81]^18^,[82]^19^,[83]^20 However, while previous studies have explored combining adoptive CAR-T cell therapy with vaccines, the potential effects of in vivo CAR immunotherapy on cancer vaccines still remain unclear. Moreover, the anti-tumor activity of vaccination-elicited antibodies, especially the antibody-dependent cellular cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP) effects, still remains to be clarified, while the therapeutic antibody drugs were widely used in treating cancers, such as anti-programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) antibodies and anti-human epidermal growth factor receptor 2 (HER2) antibodies.[84]^21^,[85]^22 Therefore, it is tempting to explore whether circRNA vaccines could synergistically boost the anti-tumor immunity of circRNA-based in vivo CAR. Given the potential superiority of circRNAs in generating CAR molecules and producing transmembrane tumor antigens in vivo, we attempted to develop the combination immunotherapy between circRNA-based in vivo CAR and cancer vaccines, aiming to synergistically enhance the anti-tumor activity of in vivo CAR via simultaneously mobilizing both the adaptive and innate immune responses. Results CircRNA^CAR efficiently expressed CAR proteins in both human primary T cells and macrophages We employed the group I intron self-splicing strategy to produce circRNAs that encode anti-HER2 CAR transmembrane proteins, designated as circRNA^CAR ([86]Figure 1A; [87]Table S1).[88]^16^,[89]^23 We also generated control circRNA, termed circRNA^Ctrl, which was an empty vector comprising circRNA circularization elements without the protein-encoding sequence. To assess the translation efficacy of circRNA^CAR, we transfected the prepared circRNA^CAR into HEK293T cells. Western blot revealed clear CAR protein expression ([90]Figure 1B), and flow cytometry analysis further confirmed the expression of CAR proteins ([91]Figure 1C). Prolonged CAR protein expression is crucial for in vivo CAR-mediated functionality. To compare circRNA^CAR and its linear counterpart mRNA^CAR, we also generated 1-methylpseudouridine-modified mRNA (1mΨ-mRNA) and unmodified mRNA through in vitro transcription. The flow cytometry result showed that circRNA^CAR exhibited significantly higher and more durable CAR expression ([92]Figure 1D). The elevated levels and prolonged presence of CAR proteins produced by circRNA^CAR suggest the potential superiority of circRNA-based in vivo CAR immunotherapy. Figure 1. [93]Figure 1 [94]Open in a new tab CircRNA^CAR efficiently expressed CAR proteins and mediated remarkable tumor killing (A) Schematic representation of circRNA^Anti-HER2-CAR circularization via group I intron autocatalysis. (B and C) Detecting the expression of CAR proteins in HEK293T cells after circRNA^Anti-HER2-CAR transfection via western blot (B) and flow cytometry (C). (D) Comparative analysis of CAR expression levels from circRNA^Anti-HER2-CAR, 1mΨ-mRNA^Anti-HER2-CAR, and unmodified mRNA^Anti-HER2-CAR in HEK293T cells. (E–G) Optimization of circRNA^Anti-HER2-CAR encoding CAR in Jurkat (E), THP-1 (F), and J774A.1 (G). (H) Detection of CAR expression in primary T cells using flow cytometry. (I–K) Cytotoxic effects of primary T cells transfected with circRNA^Anti-HER2-CAR on SK-OV-3 (I), B16F10-HER2 (J), and 4T1-HER2 (K) tumor cells. (L–N) Cytotoxic effects of Jurkat cells transfected with circRNA^Anti-HER2-CAR on SK-OV-3 (L), B16F10-HER2 (M), and 4T1-HER2 (N) tumor cells. (O–Q) Cytotoxic effects of THP-1 cells transfected with circRNA^Anti-HER2-CAR on SK-OV-3 (O), B16F10-HER2 (P), and 4T1-HER2 (Q) tumor cells. In (C)–(Q), data were presented as mean ± SEM (n = 3). An unpaired two-sided Student’s t test was performed for comparison; ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See also [95]Figure S1; [96]Table S1. However, while circRNA^CAR expressed high-level CAR proteins in HEK293T cells ([97]Figure 1D), it produced CAR proteins at much lower levels in both T cells and macrophages ([98]Figures S1A and S1B). Therefore, we further optimized the protein-encoding sequence of circRNA^CAR based on the degeneracy of codons in mammalian cells, aiming to increase its expression in immune cells. We screened out several circRNAs that enable higher expression of CAR proteins in both T cells and macrophages ([99]Figures 1E–1G and [100]S1C). Among these optimized circRNAs, the circRNA^CAR-opt-3 exhibits the most efficient expression of CAR proteins in Jurkat cells, THP-1 cells, J774A.1 cells, and human primary T cells ([101]Figures 1E–1H). Therefore, we used circRNA^CAR-opt-3 to encode anti-HER2-CAR proteins for the following research in this study. CircRNA^CAR mediated effective tumor killing in both human primary T cells and macrophages Next, we investigated whether T cells or macrophages transfected with circRNA^CAR exhibited cytotoxic effects on the targeted tumor cells. The human primary T cells, Jurkat cells, THP-1 cells, or J774A.1 were transfected with circRNA^Anti-HER2-CAR and then co-cultured with various targeted tumor cells, including SK-OV-3 cells, B16F10-HER2 cells, 4T1-HER2 cells, MC38-HER2 cells, and CT26-HER2 cells, respectively. The results showed that the human primary T cells transfected with circRNA^CAR could significantly kill those tumor cells in an effector:target (E:T) ratio-dependent manner ([102]Figures 1I–1K, [103]S1D, and S1E). Similarly, circRNA^CAR also mediated efficient tumor killing in immortalized Jurkat cells, THP-1 cells, and J774A.1 cells in an E:T ratio-dependent manner ([104]Figures 1L–1Q and [105]S1F–S1J). Macrophages exhibited efficient tumor phagocytosis and pro-inflammatory polarization induced by circRNA^CAR As a unique mechanism of macrophage, the immune defense mechanism of macrophages plays a pivotal role in phagocytosis, a process crucial for combating various threats, including tumors.[106]^24 Previous studies have illuminated the role of adenovirally transduced CAR-macrophages in mediating tumor phagocytosis.[107]^24 To explore the potential of circRNA^CAR in orchestrating tumor phagocytosis by macrophages, the THP-1 or J774A.1 cells were transfected with circRNA^CAR and then co-cultured with various targeted tumor cells, respectively. The results unveiled that circRNA^CAR mediated significant phagocytosis of diverse cancer cells in macrophage in an E:T ratio-dependent manner ([108]Figures 2A–2C and [109]S1K–S1O). Figure 2. [110]Figure 2 [111]Open in a new tab Macrophages exhibited efficient tumor phagocytosis and pro-inflammatory polarization induced by circRNA^CAR (A and B) Phagocytosis of THP-1 cells transfected with circRNA^Anti-HER2-CAR against SK-OV-3 (A) and MC38-HER2 (B) cells. (C) Phagocytosis of J774A.1 cells transfected with circRNA^Anti-HER2-CAR against CT26-HER2 cells. (D and E) Effects of circRNA^Anti-HER2-CAR on iNOS (D) and CD206 (E) expression in J774A.1. (F and G) Effects of circRNA^Anti-HER2-CAR on iNOS (F) and CD206 (G) expression in THP-1 cells. (H) Volcano plot illustrating differentially expressed genes in THP-1 cells. (I) Heatmap depicting gene expression patterns in THP-1 cells (n = 2). (J) Bubble chart of relevant biological processes via Gene Ontology (GO) analysis (n = 2). Bubble size represents the number of genes. In (A)–(G), data were presented as mean ± SEM (n = 3). An unpaired two-sided Student’s t test was conducted for comparison; ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See also [112]Figure S1. In the tumor microenvironment (TME), macrophages could be polarized to two opposite states, pro-inflammatory state (M1) or anti-inflammatory state (M2).[113]^25 Subsequently, we tested whether circRNA^CAR could affect the polarization of macrophages. Flow cytometry analysis results showed that circRNA^CAR-transfected J774A.1 cells demonstrated elevated levels of inducible nitric oxide synthase (iNOS) ([114]Figure 2D). Notably, there was only a very slight increase in CD206 ([115]Figure 2E). Similarly, circRNA^CAR-transfected THP-1 cells also exhibited the pro-inflammatory M1 phenotype but not the anti-inflammatory M2 phenotype ([116]Figures 2F and 2G). To further verify the impact of circRNA^CAR on the polarization of macrophages, we collected these transfected THP-1 cells for transcriptome-wide RNA sequencing (RNA-seq) analysis. The RNA-seq results showed that circRNA^CAR group exhibited upregulation of a series of genes related pro-inflammatory function, phagocytic capability, metabolism, and other immune responses of macrophages in comparison with circRNA^Ctrl or untreated group ([117]Figures 2H, 2I, [118]S1P, and S1Q). The pathway enrichment analysis showed that circRNA^CAR exhibited enhanced intracellular signal transduction and several enriched inflammatory-associated pathways such as tumor necrosis factor (TNF) production, interferon-gamma (IFN-γ) production, nuclear factor κB signaling, and mitogen-activated protein kinase signaling ([119]Figures 2J and [120]S1R). Collectively, these results suggested that circRNA^CAR held the potential to induce a pro-inflammatory M1 polarization in macrophages, which provided potential insights into the immunomodulatory capabilities of circRNA^CAR. Immunocyte-tropic lipid nanoparticles efficiently delivered circRNAs into immune cells in mice It is crucial to deliver circRNA^CAR into T cells and macrophages in vivo. The spleen is an important immune organ for the targeted delivery of circRNA^CAR using lipid nanoparticles (LNPs).[121]^26^,[122]^27^,[123]^28^,[124]^29^,[125]^30 Therefore, we synthesized various cationic lipids for LNPs by altering the head and tail groups. We screened out an LNP composition that contains four key components: the specific cationic lipids, cholesterol, 1,2-distearoyl-sn-glycero-3-phosphorylcholine (DSPC), and 1,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol-2000 (DMG-PEG2000), which efficiently targeted the spleen ([126]Figure 3A). Then, the circRNA^Luciferase was encapsulated within this LNP. The average size of LNP-circRNA^Luciferase formulation was approximately 110 nm, and the zeta potential was approximately 8 mV ([127]Figures 3B and 3C). To investigate the in vivo targeting effects, the LNP-circRNA^Luciferase was intravenously injected into BALB/c mice. The in vivo and ex vivo luciferase imaging results showed that LNP-circRNA^Luciferase injected intravenously was mainly concentrated in the spleen, indicating the immunocyte-tropic feature of this LNP ([128]Figures 3D and 3E). In addition, the TME contained substantial tumor-associated macrophages, T cells, and dendritic cells.[129]^31 The LNP-circRNA^Luciferase was subsequently injected into the tumors of subcutaneous tumor-bearing mice. In vivo luciferase imaging results showed that luciferase was mainly concentrated within the local tumor region ([130]Figure S2A). Figure 3. [131]Figure 3 [132]Open in a new tab Screening for immunocyte-tropic LNPs that efficiently delivered circRNAs into immune cells in mice (A) Schematic representation of the LNP-circRNA complex. (B and C) The size distribution (B) and zeta potential (C) of the LNP-circRNA^Luciferase complex. (D and E) Bioluminescence imaging in vivo (D) or ex vivo (E) of BALB/c mice intravenously injected with PBS or LNP-circRNA^Luciferase. (F and G) Bioluminescence imaging in vivo (F) or ex vivo (G) of BALB/c mice intravenously injected with PBS or SORT-circRNA^Luciferase. (H) Evaluation of targeting efficiency of SORT-circRNA^Cre in the spleen of reporter mice. (I) Detection of anti-HER2-CAR expression at different time points in various immune cells of mouse spleen. In (H) and (I), data were presented as mean ± SEM, an unpaired two-sided Student’s t test was performed for comparison; ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See also [133]Figure S2. Besides, we also tested the selective organ targeting (SORT) approach, a previously reported method where LNPs are engineered with complementary lipids for targeting extrahepatic tissues.[134]^26 This technique employs negatively charged SORT-LNPs for targeted spleen delivery, and Imaging results revealed a predominant luciferase expression in the spleen via intravenous injection of SORT-circRNA^Luciferase, indicating the immunocyte-tropic specificity of negatively charged SORT-LNPs ([135]Figures 3F and 3G). We also tested the intratumoral injection of SORT-circRNA^Luciferase in tumor-bearing BALB/C mice, the imaging results elucidated a localized high concentration in the tumor region ([136]Figure S2B), which was consistent with the above LNP we initially prepared ([137]Figure S2A). Next, we further utilized flow cytometry analysis to detect the cell-targeting specificity of SORT-circRNA using reporter mice, harboring a CAG-loxp-zsGreen-stop-loxp-tdTomato-PolyA cassette integrated into the mouse genome. PBS or SORT-circRNA^Cre was intravenously administered into the reporter mice, and then the spleens and lymph nodes were collected for flow cytometry analysis. The results revealed that SORT-circRNA^Cre enables effective delivery of circRNA in T cells, natural killer (NK) cells, macrophages, and dendritic cells ([138]Figures 3H and [139]S2C). Moreover, to further assess the expression of circRNA-encoded anti-HER2-CAR proteins at different time points in immune cells of mice, BALB/c mice were intravenously injected with PBS, SORT-circRNA^Ctrl, or SORT-circRNA^CAR, respectively. The flow cytometry analysis results demonstrated that SORT-circRNA^CAR effectively expressed anti-HER2-CAR proteins in T cells, NK cells, and macrophages in the spleens and lymph nodes, forming in vivo panCAR (CAR-T, CAR-NK, and CAR-Macrophage) cells ([140]Figures 3I and [141]S2D). A single dose of circRNA-based in vivo CAR could produce in vivo panCAR lasting more than 72 h in the spleens and lymph nodes ([142]Figures 3I and [143]S2D). These results indicated that the circRNA-based in vivo panCAR could simultaneously utilize both adaptive and innate immune responses to exert targeted anti-tumor immunity. CircRNA^CAR efficiently inhibited tumor growth and improved survival time in mice To test whether circRNA^CAR can inhibit tumor growth in vivo, we firstly used immune-competent BALB/c mice with CT26-HER2 colorectal carcinomas ([144]Figure 4A). When the subcutaneous CT26-HER2 tumor volume reached approximately 50–80 mm^3, the tumor-bearing mice were randomly grouped and intravenously administered three times with PBS, 15 μg of SORT-circRNA^Ctrl, or 15 μg of SORT-circRNA^CAR ([145]Figure 4A). The result showed that SORT-circRNA^CAR significantly inhibited tumor growth, in comparison with PBS or SORT-circRNA^Ctrl ([146]Figure 4B). Figure 4. [147]Figure 4 [148]Open in a new tab CircRNA^CAR efficiently inhibited tumor growth and improved survival time in mice (A) Schematic illustration of intravenous PBS, LNP-circRNA^Ctrl, or SORT-circRNA^Anti-HER2-CAR treatment in the CT26-HER2 tumor model. (B) Tumor growth curves for CT26-HER2 tumor-bearing mice treated as indicated in (A). (C) Schematic illustration of intratumoral PBS, LNP-circRNA^Ctrl, or LNP-circRNA^Anti-HER2-CAR treatment in the CT26-HER2 tumor model. (D and E) Tumor growth curves (D) and survival curves (E) of CT26-HER2 tumor-bearing mice treated as indicated in (C). (F) In vivo bioluminescence imaging of CT26-HER2 tumor-bearing mice treated as indicated in (C). (G) The quantified signal intensity of bioluminescence imaging in (F). (H) Schematic illustration of intratumoral PBS, LNP-circRNA^Ctrl, or LNP-circRNA^Anti-HER2-CAR treatment in the 4T1-HER2 tumor model. (I) Tumor growth curves of 4T1-HER2 tumor-bearing mice treated as indicated in (H). (J) Tumor growth curves of individual 4T1-HER2 tumor-bearing mice treated as indicated in (H). (K) Schematic illustration of intratumoral PBS, LNP-circRNA^Ctrl, or LNP-circRNA^CAR treatment in the MC38-HER2 tumor model. (L and M) Tumor growth curves (L) and survival curves (M) of MC38-HER2 tumor-bearing mice treated as indicated in (K). In (B), (D), (I), and (L), data were represented as the mean ± SEM, the tumor growth curves were calculated by two-way ANOVA analysis (n = 6). In (E) and (M), data were represented as the mean ± SEM, the survival curves were calculated by Kaplan-Meier simple survival analysis (n = 6). In (G), data were represented as the mean ± SEM, an unpaired two-sided Student’s t test was conducted for comparison. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [149]Figure S2. Apart from the intravenous administration, we also tested the intratumoral administration in mice with subcutaneous CT26-HER2 tumors ([150]Figure 4C). Similarly, when the tumor volume reached approximately 50–80 mm^3, the mice were randomly grouped and intratumorally injected four times with PBS, LNP-circRNA^Ctrl, or LNP-circRNA^CAR ([151]Figure 4C). We found that LNP-circRNA^CAR could also significantly reduce tumor burden and prolong survival time, in comparison with PBS or LNP-circRNA^Ctrl ([152]Figures 4D–4G and [153]S2E). Similarly, in 4T1 breast carcinoma-bearing mouse models ([154]Figure 4H), the intratumoral administration of LNP-circRNA^CAR significantly inhibited tumor growth and even achieved tumor regression in comparison with PBS or LNP-circRNA^Ctrl ([155]Figures 4I and 4J). As for the subcutaneous MC38-HER2 tumors ([156]Figure 4K), the intratumoral administration of LNP-circRNA^CAR also significantly inhibited tumor growth and increased the survival time of tumor-bearing mice in comparison with PBS or LNP-circRNA^Ctrl ([157]Figures 4L, 4M, and [158]S2F). Collectively, these results demonstrated that the circRNA-based in vivo CAR exerted potent efficacy in treating tumors. CircRNA^CAR reshaped the TME to a pro-inflammatory state in mice Solid tumors are often refractory partly due to the immunosuppressive TME.[159]^32 Therefore, we further investigated the effects of circRNA^CAR on the TME. At the end of experiments via intravenous administration of PBS, circRNA^Ctrl, or circRNA^CAR, the mouse spleens and tumor tissues were collected for detecting the infiltration of immune cells via flow cytometry analysis ([160]Figures S3A–S3C). The results showed that circRNA^CAR exhibited a significant increase in the proportion of CD8^+ T cells, CD8^+ effector memory T cells (Tem), CD8^+ central memory T cells (Tcm), and CD4^+ T cells but a significant decrease in the proportion of regulatory T (Treg) cells in the spleen, compared to the PBS or circRNA^Ctrl ([161]Figures 5A and [162]S3D). As for tumor tissues, we also found that circRNA^CAR exhibited increased proportions of tumor-infiltrating CD45^+ cells, CD8^+ T cells, and CD4^+ T cells but a decreased proportion of tumor-infiltrating Treg cells, compared to the PBS or circRNA^Ctrl ([163]Figures 5B and [164]S3E). The flow cytometry results also showed that circRNA^CAR exhibited a significantly increased proportion of pro-inflammatory M1-polarized macrophages (major histocompatibility complex [MHC] II^+ macrophages) in the TME, in comparison with PBS or circRNA^Ctrl ([165]Figures 5B and [166]S3E). Figure 5. [167]Figure 5 [168]Open in a new tab In vivo panCAR reshaped the tumor microenvironment to a pro-inflammatory state (A) Changes in the proportion of immune cells of spleen via flow cytometry. CD8^+ T cells, CD8^+ Tem cells, CD8^+ Tcm cells, or CD4^+ T cells were gated from CD45^+ cell population, and Treg cells were gated from CD4^+ T cell population. (B) Flow cytometric analysis of changes in the proportion of infiltrating immune cells in tumors. CD8^+ T cells, CD4^+ T cells, or MHC II^+ macrophages were gated from CD45^+ cell population, and Treg cells were gated from CD4^+ T cell population. (C and D) H&E staining (C) or IHC staining (D) of tumor tissue sections obtained from CT26-HER2 tumor-bearing mice after PBS, SORT-LNP-circRNA^Ctrl, or SORT-LNP-circRNA^Anti-HER2-CAR treatment. The integrated density of IHC staining was quantified using ImageJ. (E) Heatmap of gene expression patterns of immune cells extracted from tumor tissues (n = 2). (F) Bubble chart of relevant biological processes through GO analysis (n = 2). The size of the bubbles represented the number of genes. (G) Gene set enrichment analysis (GSEA) showing enriched pathways in immune cells extracted from tumor tissues (n = 2). In (A), (B), and (D), data were represented as the mean ± SEM; an unpaired two-sided Student’s t test was conducted for comparison; ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. Each symbol represents an individual mouse. See also [169]Figures S3 and [170]S4. Besides, we also harvested tumor tissue specimens from treated mice for hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) staining. H&E staining showed that circRNA^CAR exhibited a significant increase in tumor cell death with enlargement of tumor cell gaps, pale cytoplasm, cell disintegration, and loss of structure, while PBS or circRNA^Ctrl exhibited diffuse growth and dense arrangement in both the CT26-HER2 and MC38-HER2 colorectal carcinoma-bearing mice ([171]Figures 5C, [172]S4A, and S4B). IHC staining demonstrated increased infiltration of both CD8^+ T cells and CD86^+ cells in the circRNA^CAR group, compared to the PBS or circRNA^Ctrl group in both MC38-HER2 and CT26-HER2 tumor-bearing mice ([173]Figures 5D, [174]S4C, and S4D). To further explore the impact of circRNA^CAR on the TME, we utilized Percoll to isolate immune cells from tumor tissues via density gradient centrifugation and performed RNA-seq analysis. The differential gene expression analysis showed that circRNA^CAR exhibited upregulation of genes related to pro-inflammatory cytokines, chemokines, and chemokine receptors, in comparison with circRNA^Ctrl or PBS, suggesting the infiltration of immune cells in the TME ([175]Figures 5E and [176]S4E). The pathway enrichment analysis showed that circRNA^CAR exhibited several enriched pathways related to T cell proliferation, dendritic cell migration, phagocytosis, and pro-inflammatory responses such as IFN-γ signaling, TNF alpha (TNF-α) signaling, and interleukin (IL)-6-JAK-STAT3 signaling, in comparison with PBS or circRNA^Ctrl ([177]Figures 5F, 5G, [178]S4F, and S4G). We found that multiple pathways related to B cell immunity, such as B cell activation, B cell proliferation, immunoglobulin-mediated immune response, immunoglobulin production, and complement activation, were significantly enriched in the circRNA^CAR group ([179]Figures 5F and [180]S4F). This finding suggested that the antibodies might play an important role in the anti-tumor immunity of in vivo panCAR immunotherapy, warranting further investigation. Collectively, these results demonstrated that circRNA^CAR could markedly reshape TME to a pro-inflammatory state in tumor mouse models, which tended to sensitize immunotherapy. CircRNA vaccine synergistically boosted the anti-tumor activity of in vivo panCAR Recent reports proposed that vaccines might further enhance the therapeutic efficacy of CAR-T adoptive cell therapy in mouse tumor models.[181]^17^,[182]^18^,[183]^19^,[184]^20 Notably, the targeted proteins of the CAR molecules align with the antigens of the cancer vaccine,[185]^19^,[186]^20 which were designed to induce antigen spreading through the reactivation of infused CAR-T cells by the corresponding antigens of cancer vaccine.[187]^17 The aforementioned RNA-seq results suggested that the anti-tumor effects of circRNA^CAR therapy might be related to B cell responses ([188]Figures 5F and [189]S4F). Additionally, antibodies could promote phagocytosis or killing effects of macrophages or NK cells through ADCC and ADCP.[190]^21^,[191]^22 Therefore, we aimed to investigate whether circRNA vaccines could synergistically boost the anti-tumor immunity of the circRNA-based in vivo panCAR, in the manner of ADCC or ADCP. As the aforementioned circRNA^CAR encoded anti-HER2 CAR molecules that target human HER2 proteins, we developed a circRNA vaccine (circRNA^VAC) that encoded a human HER2 fusion protein, in which the endocytosis prevention motif (EPM) and ESCRT- and ALIX-binding region (EABR) domain were fused at the C-terminal instead of the original intracellular domain of human HER2 protein, aiming to help elicit higher level of HER2-specific antibodies ([192]Figure 6A; [193]Table S1).[194]^33 Therefore, we tested the combination immunotherapy between the circRNA-based in vivo CAR and circRNA-based cancer vaccines (in vivo panCAR-VAC). In HEK293T cells, circRNA^VAC exhibited efficient expression of HER2-EPM-EABR fusion protein ([195]Figure 6B), concomitant with the efficient secretion of vesicles in the supernatant ([196]Figure 6C). Next, we tested whether circRNA^VAC could induce HER2-specific antibodies. The BALB/c mice were immunized with LNP-circRNA^VAC through intramuscular injection twice at a 3-week interval, and the sera of immunized mice were collected at 1 week after the boost. The ELISA result showed that LNP-circRNA^VAC could elicit HER2-specific IgG antibodies, and the geometric mean titer was ∼4.9 × 10^6 ([197]Figure 6D). Figure 6. [198]Figure 6 [199]Open in a new tab CircRNA vaccine synergistically boosted the anti-tumor activity of in vivo panCAR (A) Schematic illustration of circRNA^VAC design. EPM, the endocytosis prevention motif; EABR, the ESCRT- and ALIX-binding region domain. (B) Flow cytometric analysis detecting the translation of circRNA^HER2 in HEK293T cells (n = 3). (C) Electron microscopy showing the vesicles secreted by HEK293T cells transfected with circRNA^HER2-EPM-EABR. (D) Measurement of the endpoint titer of HER2-specific IgG with ELISA. (E) Schematic illustration of 4T1-HER2 tumor-bearing mice receiving PBS (intravenously), circRNA^CAR (intravenously), circRNA^VAC (intramuscularly), or circRNA^CAR (intravenously) plus circRNA^VAC (intramuscularly) combined therapy (n = 5). (F) Tumor growth curves of overall mice treated as indicated in (E). (G) Tumor growth curves of individual mouse treated as indicated in (E). (H) Schematic illustration of B16F10-HER2 tumor-bearing mice receiving PBS (intravenously), circRNA^CAR (intravenously), circRNA^VAC (intramuscularly), or circRNA^CAR (intravenously) plus circRNA^VAC (intramuscularly) combined therapy (n = 5). (I) Tumor growth curves of overall mice treated as indicated in (H). (J) Tumor growth curves of individual mouse treated as indicated in (H). (K) Survival curves of B16F10-HER2 tumor-bearing mice treated as indicated in (H). In (B), data were represented as the mean ± SEM; an unpaired two-sided Student’s t test was performed for comparison. In (D), data were represented as the geometric mean ± geometric SD; an unpaired two-sided Student’s t test was performed for comparison. In (F) and (I), data were represented as the mean ± SEM, and the tumor growth curves were calculated by two-way ANOVA analysis. In (K), the survival curves were calculated by Kaplan-Meier simple survival analysis. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See also [200]Figures S5−S7; [201]Table S1. To test the effects of in vivo panCAR-VAC therapy, the BALB/c mice with subcutaneous 4T1-HER2 tumors were injected with PBS (intravenously), circRNA^CAR (intravenously), circRNA^VAC (intramuscularly), or circRNA^CAR (intravenously) plus circRNA^VAC (intramuscularly) at 6, 10, and 18 days post tumor engraftment ([202]Figure 6E). The results showed that the combination of circRNA^CAR and circRNA^VAC showed improved tumor inhibition, in comparison with PBS, individual circRNA^CAR, or circRNA^VAC ([203]Figures 6F and 6G). To further confirm the enhanced effect of in vivo panCAR-VAC on tumor inhibition in mice models, we used additional tumor models, including B16F10-HER2 and MC38-HER2 tumor models ([204]Figures 6H and [205]S5A). The results showed that both circRNA^CAR and circRNA^VAC individually inhibited tumor growth compared to the PBS group, and the combination of circRNA^CAR and circRNA^VAC showed improved tumor inhibition and significantly enhanced survival rate ([206]Figures 6I–6K, [207]S5B, and S5C). In addition to delivering circRNA^CAR via SORT-LNP, we also assessed the impact of delivering circRNA^CAR using the LNP we screened out ([208]Figures 3A–3D) on tumor growth. As before, CT26-HER2 tumors were subcutaneously implanted in BALB/c mice, and when the tumor volume reached 50–80 mm^3, the mice were randomly divided and injected with PBS, circRNA^CAR, circRNA^VAC, or the combination group of circRNA^CAR and circRNA^VAC ([209]Figure S5D). The results showed that compared to PBS, the combination group had a significant effect on inhibiting tumor growth and improving survival rates in mice ([210]Figures S5E–S5G). Additionally, we also investigated the efficacy of intratumoral administration, as an alternative to the intravenous administration, for the delivery of circRNA^CAR ([211]Figure S5H). Impressively, the combination of intratumorally delivered circRNA^CAR and circRNA^VAC vaccine (intramuscularly) markedly impeded tumor growth, resulting in a noteworthy enhancement in the survival rate of tumor-bearing mice ([212]Figures S5I–S5K). Collectively, these results demonstrated that the circRNA vaccines could synergistically boost the anti-tumor immunity of circRNA-based in vivo panCAR administered either intratumorally or intravenously, which holds the potential for an upgraded off-the-shelf in vivo panCAR-VAC immunotherapy. To further investigate the effects of in vivo panCAR-VAC immunotherapy on the TME, we euthanized MC38-HER2 tumor-bearing mice at 1 week after the final injection and collected the tumor tissues for analysis. The flow cytometry results showed that the in vivo panCAR-VAC-treated group significantly enhanced immune cell infiltration in the TME ([213]Figure S6A). Specifically, the panCAR-VAC group exhibited a significantly increased proportion of pro-inflammatory M1-polarized macrophages and a reduced proportion of anti-inflammatory M2-polarized macrophages compared to PBS, individual circRNA^CAR, or individual circRNA^VAC ([214]Figures S6B–S6D), while the proportions of exhausted T cells, exhausted NK cells, regulatory T cells, and myeloid-derived suppressor cells were reduced in the in vivo panCAR-VAC-treated group ([215]Figures S6E–S6J). H&E staining revealed that tumor tissues in the PBS group were tightly structured with deeper nuclear staining, while the individual circRNA^CAR or circRNA^VAC groups disrupted the tumor cell structure, and the in vivo panCAR-VAC combination group exhibited a significant increase in tumor cell death, characterized by enlarged tumor cell gaps and cell disintegration ([216]Figure S6K). IHC staining demonstrated that in vivo panCAR-VAC combination therapy promoted the infiltration of CD8^+ cells, CD11b^+ cells, and CD86^+ cells while reducing the infiltration of immunosuppressive Foxp3^+ cells ([217]Figures S6L and S6M). Collectively, these results demonstrated that in vivo panCAR-VAC combination therapy could markedly reprogram the TME to a pro-inflammatory state. To assess the safety of in vivo panCAR and panCAR-VAC therapies in mice, the BALB/c mice were treated with PBS, circRNA^CAR, circRNA^VAC, or the combination of circRNA^CAR and circRNA^VAC for short-term and long-term observations. The ELISA results indicated that neither the individual nor the combined groups exhibited increased levels of cytokines, including TNF-α, IL-1β, and IL-6, as well as alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase in both the short-term and long-term observations ([218]Figures S7A–S7C). Additionally, H&E staining revealed that in vivo panCAR and in vivo panCAR-VAC therapies resulted in no significant organ damage or major abnormalities in the important organs (heart, liver, spleen, lung, and kidney) of treated mice ([219]Figure S7D). CircRNA^CAR boosted the level of circRNA vaccine-elicited antibodies enhancing the anti-tumor activity via antibody-mediated cellular cytotoxicity Inspired by the finding of potential antibody-mediated anti-tumor immunity of in vivo panCAR therapy ([220]Figures 5F and [221]S4F), we further collected the antibodies of serum from mice treated with PBS, circRNA^CAR, circRNA^VAC, or circRNA^CAR and circRNA^VAC combined therapy. The ELISA results showed that the circRNA^CAR and circRNA^VAC combined group elicited higher endpoint titer of total IgG, IgG1, IgG2A, IgG2B, and IgG2C subtype than individual circRNA^VAC, circRNA^CAR, or PBS, while the endpoint titer of IgG3 and unmatured IgM showed no obvious difference ([222]Figures 7A and [223]S7E–S7G), indicating that the in vivo panCAR therapy might boost the level of circRNA vaccine-elicited functional antibodies. Figure 7. [224]Figure 7 [225]Open in a new tab In vivo panCAR enhanced the anti-tumor activity via antibody-mediated cellular cytotoxicity (A) Measurement of HER2-specific IgG, IgG1, IgG2A, IgG2B, or IgG2C-binding antibodies with ELISA (n = 5 or 6). (B and C) HER2-specific antibodies in (A) mediated cellular cytotoxicity against SK-OV-3 (B) and MC38-HER2 (C) tumor cells in J774A.1. (D and E) HER2-specific antibodies in (A) mediated cellular cytotoxicity against SK-OV-3 (D) and MC38-HER2 (E) tumor cells in RAW 264.7. (F) Tumor growth curves of overall CT26 tumor-bearing mice treated with PBS, in vivo panCAR-VAC, or in vivo panCAR-VAC plus anti-NK1.1 antibodies to deplete NK cells (n = 5). (G) Tumor growth curves of individual mouse treated as indicated in (F). (H) Survival curves of mice treated as indicated in (F). (I) Tumor growth curves of overall CT26 tumor-bearing mice treated with PBS, in vivo panCAR-VAC, or in vivo panCAR-VAC plus anti-CSF1R antibody to deplete macrophages (n = 5). (J) Tumor growth curves of individual mouse treated as indicated in (I). (K) Survival curves of mice treated as indicated in (I). (L) The potential mechanism diagram of synergistic in vivo panCAR-VAC immunotherapy. In (A), data are shown as the mean ± SEM; In (B)–(E), data were represented as the mean ± SEM; an unpaired two-sided Student’s t test was performed for comparison. In (F) and (I), tumor growth curves were calculated by two-way ANOVA analysis (n = 5). In (H) and (K), survival curves were calculated by Kaplan-Meier simple survival analysis (n = 5). ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See also [226]Figure S7. Next, we further explored whether the HER2-specific antibodies could exercise anti-tumor function, such as ADCC or ADCP activity. The in vitro tumor-killing results showed that the HER2-specific antibodies elicited by circRNA^CAR and circRNA^VAC combined therapy mediated significantly higher killing effects in both J774A.1 and RAW 264.7 macrophages, in comparison with PBS, circRNA^CAR, or circRNA^VAC ([227]Figures 7B–7E). Next, to further verify the function of NK cells or macrophages in in vivo panCAR-VAC-mediated tumor inhibition, we employed an anti-NK1.1 antibody or anti-CSF1R antibody to deplete NK cells or macrophages in tumor-bearing mice treated with in vivo panCAR-VAC, respectively. The results demonstrated that depleting NK cells or macrophages significantly reduced the efficacy of in vivo panCAR-VAC-mediated tumor inhibition and the survival of mice ([228]Figures 7F–7K), indicating that both NK cells and macrophages played crucial roles in the anti-tumor immunity mediated by in vivo panCAR-VAC. Traditional strategies of cancer vaccines have been mainly focusing on the cellular immunity but not the humoral immunity, whereas in this study, we found that B cell-mediated immune responses played an important role in tumor inhibition. On one hand, circRNA vaccine-elicited tumor-specific antibodies might boost the in vivo circRNA^CAR-mediated tumor killing via ADCC and ADCP effects, beyond the basic CAR-mediated phagocytosis or killing effects in the TME ([229]Figure 7L). On the other hand, in return, the elevated exposure of tumor antigens might further boost the humoral immunity to produce potent tumor-specific antibodies ([230]Figure 7L). This potential positive feedback loop between antibodies and antigens might partly explain the synergically enhanced anti-tumor immunity of in vivo panCAR-VAC immunotherapy. In summary, we provided the proof-of-concept study, which established the combined immunotherapy of circRNA-based in vivo panCAR and cancer vaccine, termed in vivo panCAR-VAC, achieving synergically enhanced anti-tumor immunity via simultaneously mobilizing both the adaptive and innate immune responses. Nevertheless, the potential of antibody-mediated anti-tumor immunity elicited by cancer vaccines still awaits further investigation. Discussion CircRNA, a covalently closed circular RNA molecule, is highly stable and has been engineered for RNA editing, RNA vaccines against infectious diseases, cancer vaccines, and gene therapy.[231]^34^,[232]^35^,[233]^36^,[234]^37^,[235]^38^,[236]^39 Recent research reported the circRNA-based T cell receptor-T adoptive cell therapy for treating cytomegalovirus infection.[237]^40 Unlike DNA vector platforms, such as DNA transposase,[238]^7 adeno-associated virus vectors,[239]^41^,[240]^42 and lentivirus-based vectors,[241]^12^,[242]^13 the expression of circRNA^CAR was transient and reversible without integrating into the genomic DNA. In this study, we generated circRNA^CAR, which encoded anti-HER2 CAR proteins for in vivo CAR immunotherapy to treat tumors in mice. We demonstrated that circRNA^CAR exhibited higher and more durable expression of CAR proteins compared with mRNA^CAR, indicating the potential superiority of circRNA-based in vivo CAR immunotherapy ([243]Figure 1D). For in vivo study, using the immunocyte-tropic LNPs ([244]Figure 3), circRNA^CAR was delivered into tumor-bearing mice and effectively suppressed tumor growth and markedly improved the survival rate in multiple mouse models ([245]Figure 4). Moreover, we also found that circRNA^CAR could reshape the TME to a highly pro-inflammatory state ([246]Figures 5, [247]S3, and [248]S4). Therefore, this circRNA-based in vivo CAR therapy held the potential to sensitize the anti-tumor immunity via combination with other cancer therapies, such as the targeted therapy or immune checkpoint blockade therapies (e.g., anti-PD-1/PD-L1 antibodies). In this study, we observed that circRNA^Ctrl also exhibited partial anti-tumor effects ([249]Figure 4), which might result from the innate immune responses of in vitro transcription-produced circRNAs[250]^16^,[251]^43^,[252]^44^,[253]^45 or the adjuvant activity of LNP.[254]^46^,[255]^47^,[256]^48^,[257]^49 In comparison with the adoptive CAR-T cell therapy, this off-the-shelf in vivo circRNA^CAR therapy potentially had multiple advantages such as (1) lower cost, (2) off-the-shelf therapy, (3) avoiding the risk of genomic integration, (4) no need for lymphodepletion, and (5) repeatable dosing. It was worth mentioning that in this study, circRNA^CAR was delivered to a variety of immune cells in the spleen and lymph nodes, including T cells, macrophages, and NK cells, through immunocyte-tropic LNPs, forming panCAR (CAR-T, CAR-M, and CAR-NK) immune effector cells ([258]Figure 3), thereby exerting efficient targeted killing function. Compared with CAR-T cell therapy[259]^50^,[260]^51 or T cell engager therapy,[261]^52^,[262]^53 in vivo panCAR therapy simultaneously mobilized adaptive (T cell) and innate (NK cells and macrophages) immunity to exert targeted anti-tumor function and would have higher universality in disease treatment applications, especially for people with T cell immunodeficiency or T cell immune dysfunction. In addition, T cell exhaustion was the main difficulty in treating solid tumors.[263]^54 The in vivo panCAR strategy also mobilized innate immune cells such as NK cells and macrophages to exert anti-tumor immunity, which was an important supplement to adaptive immunity to eliminate tumors. Previous research reported that mRNA vaccines encoding tumor-associated antigens could help enhance the anti-tumor effects of adoptive CAR-T cell therapy.[264]^19^,[265]^20 Up to now, there are no reports about the combined immunotherapy between in vivo CAR therapy and RNA vaccines. More importantly, antibodies play an important role in macrophages or NK cell-mediated phagocytosis or killing via ADCC or ADCP effects.[266]^21^,[267]^22^,[268]^55^,[269]^56 Current studies on the combined therapy of CAR-T adoptive cell therapy and vaccines have only reported on the interactions between CAR-T cells and antigen-presenting cells,[270]^17 while the functions of vaccination-elicited antibodies still remain to be clarified. In this study, we formulated a circRNA-based cancer vaccine encoding human HER2 antigens with the fused intracellular EPM-EABR motif to enhance the level of HER2-specific antibodies.[271]^33 We further demonstrated that this circRNA vaccine could synergistically boost the anti-tumor immunity of circRNA-based in vivo panCAR ([272]Figures 6 and [273]S5). More importantly, vaccination-induced endogenous antibodies had a complete structure and could effectively mediate ADCC and ADCP effects, which were an important bridge connecting innate immune responses and adaptive immune responses. We also demonstrated that depleting NK cells or macrophages significantly reduced the effects of in vivo panCAR-VAC-mediated tumor inhibition and the survival rate of mice ([274]Figures 7F–7K). These results indicated that the anti-HER2 antibody promoted tumor killing, possibly via ADCC or ADCP, but likely did not interfere with CAR binding. Cancer immunotherapy relied on systemic immune responses. The in vivo panCAR-VAC strategy could effectively mobilize adaptive and innate immune cells to synergistically exert anti-tumor immunity. Mechanistically, it was possible that the immune cells expressing CAR molecules, such as T cells, macrophages, and NK cells, could specifically kill tumor cells, release inflammatory cytokines, reshape the immunosuppressive TME, and promote the infiltration of pro-inflammatory immune cells. Beyond the CAR-mediated tumor killing, the corresponding circRNA vaccine could concurrently elicit strong B cell responses and produce high-level tumor-specific antibodies to boost the in vivo circRNA^CAR-mediated tumor killing via ADCC and ADCP. On the other hand, the elevated tumor killing generated more exposure of tumor antigens to further boost humoral immunity producing potent tumor-specific antibodies in the germinal centers of tumor-draining lymph node (TDLN) or spleen, forming the positive feedback loop of antibody-antigen ([275]Figure 7L). This proof-of-concept study established a potent combined immunotherapy between in vivo panCAR and cancer vaccines, which achieved synergistically enhanced anti-tumor effects in multiple mouse models. Notably, this study found that the vaccination-elicited antibodies held an important role in anti-tumor immunity, providing a concept for the synergistic in vivo panCAR-VAC immunotherapy. Limitations of the study In this study, the intravenous administration of circRNA^Anti-HER2-CAR exhibited slightly inferior anti-tumor activity, in comparison with the intratumoral administration of circRNA^Anti-HER2-CAR, which may be related to the targeting efficiency of LNPs. In the future, using optimized LNPs with higher targeting efficiency, the intravenously delivered circRNA^Anti-HER2-CAR might achieve remarkable anti-tumor effects. Besides, this study also lacked detailed research about the mechanism of synergistically enhanced anti-tumor immunity of in vivo panCAR-VAC therapy, and the further analysis of the TME with single-cell RNA-seq method and immunological experiments with gene knockout mouse models might help further clarify the synergistic mechanism of in vivo panCAR-VAC immunotherapy. Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Liang Qu (quliang@fudan.edu.cn). Materials availability All unique reagents generated in this study are available from the [276]lead contact with a completed material transfer agreement. Data and code availability The data supporting the findings of this study are available from the [277]lead contact upon a reasonable request under a completed material transfer agreement. This study did not generate any unique code. Any additional information required to re-analyze the data reported in this study is available from the [278]lead contact upon request. The accession code of the transcriptome-wide RNA-seq data in this study was GEO: [279]GSE268105 of NCBI Gene Expression Omnibus database. Acknowledgments