Abstract The immunosuppressive tumor microenvironment (TME) critically undermines the efficacy of T cell-based tumor immunotherapy by impeding CD8^+ T cell infiltration and cytotoxic function, primarily through tumor-associated macrophages (TAMs) and immune checkpoint molecules such as programmed death ligand 1 (PD-L1). Here, we present a multifunctional nanoplatform, IN@OMV-PDL1nb, designed to simultaneously inhibit TAM-derived immunosuppressive metabolite itaconic acid (ITA) by targeting immune-responsive gene 1 (IRG1) and block PD-L1 within the TME. Engineered outer membrane vesicles (OMVs) serve as precision delivery vehicles for the IRG1 inhibitor IRG1-IN-1 (IN) and as carriers for PD-L1 nanobody release, activated by matrix metalloproteinase-2 (MMP-2). IN@OMV-PDL1nb effectively inhibits IRG1 expression in TAMs, thus reducing the accumulation of ITA, restoring chemokines (CXCL9 and CXCL10) secretion, and enhancing CD8^+ T cells infiltration within tumors. The released PD-L1 nanobody protects CD8^+ T cells, preserving their tumoricidal activity. In murine tumor models, IN@OMV-PDL1nb significantly inhibited tumor growth, increased survival, and enhanced antigen presentation and T cell recruitment. Additionally, IN@OMV-PDL1nb induced robust adaptive immunity, facilitating antigen-specific immune memory that prevented tumor recurrence and metastasis. This dual-targeting approach offers a promising strategy to overcome TME-driven immunosuppression in tumor immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03507-7. Keywords: Tumor immunotherapy, Tumor microenvironment, Immune checkpoint blockade, Tumor-associated macrophages, Outer membrane vesicles Graphical Abstract [62]graphic file with name 12951_2025_3507_Figa_HTML.jpg [63]Open in a new tab A multifunctional nanoplatform, IN@OMV-PDL1nb@CaP, is engineered through genetic modifications to express a matrix metalloproteinase-2 (MMP-2)-responsive peptide (PLGLAG) and the PD-L1 nanobody (PD-L1nb) on its surface, while IRG1-IN-1 is loaded into its internal cavity. Additionally, calcium phosphate (CaP) is biomineralized on the surface. This platform effectively alleviates immune suppression induced by tumor-associated macrophages (TAMs) and the immune checkpoint molecule PD-L1 within the tumor microenvironment (TME), offering a promising approach for tumor immunotherapy Introduction The recruitment, infiltration, and functional activity of T cells within tumor tissues are critical determinants of the success of anti-tumor immunotherapy [[64]1, [65]2]. However, the efficacy of T cell-mediated immune responses is severely constrained by the immunosuppressive tumor microenvironment (TME) [[66]3–[67]5]. Tumor-associated macrophages (TAMs), the most abundant immune cells within tumors, are key players in establishing this suppressive milieu [[68]6, [69]7]. Tumor cells induce high expression of IRG1 in TAMs via activation of the nuclear factor-κB (NF-κB) pathway. The downstream metabolite of IRG1, ITA, suppresses the production of macrophage-derived chemokines such as CXCL9 and CXCL10, reducing CD8^+ T cell infiltration into tumor tissues [[70]8, [71]9]. Furthermore, the infiltrating T cells often fail to execute their cytotoxic functions due to the high expression of immune checkpoints, such as PD-L1, within the TME. This leads to programmed exhaustion and functional impairment of T cells [[72]10–[73]12]. Thus, a strategy that combines IRG1 inhibition in TAMs with PD-L1 blockade to remodel the TME holds significant potential for improving anti-tumor immunotherapy. One major challenge in targeting IRG1 lies in the lack of cellular specificity of its inhibitors, making selective delivery to TAMs crucial for regulating their functions. OMVs, naturally secreted spherical nanoparticles from Gram-negative bacteria (20–250 nm in size), offer a promising solution [[74]13–[75]15]. OMVs possess unique biological properties, including their composition of pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS), flagellin, and peptidoglycan, which render them highly immunogenic and capable of activating multiple toll-like receptor (TLR) signaling pathways [[76]16–[77]18]. OMVs are readily recognized and phagocytosed by macrophages and, due to their internal cavity structure, serve as ideal carriers for delivering therapeutic agents specifically to TAMs [[78]19, [79]20]. This makes OMVs an ideal delivery vehicle for selectively targeting TAMs and modulating their immunosuppressive functions within the TME. In this study, we developed a multifunctional nanoplatform, IN@OMV-PDL1nb, to simultaneously target IRG1 in TAMs and block the PD-L1 in TME, thereby remodeling the TME and enhancing anti-tumor immunity. Using genetically engineered Escherichia coli (E. coli), OMVs were designed to express MMP-2-responsive peptide (PLGLAG) and PD-L1 nanobody (PDL1nb) on their surface (OMV-PDL1nb) [[80]21, [81]22]. Next, we used electroporation to package the IRG1 inhibitor IRG1-IN-1 into the internal cavity of OMV-PDL1nb to form IN@OMV-PDL1nb (Fig. [82]1A). In the tumor site, the abundant MMP-2 cleaves its substrate peptide between OMVs and PDL1nb, releasing PDL1nb to block PD-L1 and restore CD8^+ T cell cytotoxicity. Simultaneously, the activation of TAMs by IN@OMVs is accompanied by efficient phagocytosis, releasing IRG1-IN-1 within these cells. The inhibition of IRG1 reduces the production of ITA, restoring the secretion of chemokines such as CXCL9 and CXCL10, which facilitate the recruitment and infiltration of CD8^+ T cells into the tumor (Fig. [83]1B). Subsequently, neoantigens released by dead tumor cells were drained into lymph nodes, which induced stronger adaptive immunity and long-term immune memory that prevented tumor recurrence and metastasis in the mice (Fig. [84]1C). This dual-targeting strategy effectively overcomes immune suppression within the TME, promoting synergistic anti-tumor responses and offering a promising platform for advancing tumor immunotherapy. Fig. 1. [85]Fig. 1 [86]Open in a new tab Schematic illustration of construction and antitumor strategy of IN@OMV-PDL1nb. (A) The preparation of IN@OMV-PDL1nb involved engineering E. coli to express the MMP-2-responsive peptide (PLGLAG) and the PD-L1 nanobody (PDL1nb) on the surface protein ClyA, resulting in OMV-PDL1nb. Subsequently, the IRG1 inhibitor IRG1-IN-1 was loaded into OMV-PDL1nb via electroporation to form IN@OMV-PDL1nb. (B) In the tumor site, IN@OMV-PDL1nb facilitates the recruitment and infiltration of CD8^+ T cells by increasing the secretion of chemokines. It also blocks PD-L1 through the released PDL1nb, thereby restoring CD8^+ T cell cytotoxicity, which can then attack tumor cells effectively. (C) Neoantigens released from dead tumor cells are drained into the lymph nodes, which can induce stronger adaptive immunity and promote long-term immune memory in mice. MMP-2, matrix metalloproteinase-2. TLR4, toll-like receptor 4. IRG1, immune-responsive gene 1. ITA, itaconic acid Materials and methods Materials The following antibodies for flow cytometry were purchased from BioLegend Inc. (USA): anti-mouse CD3ε-PerCP/Cyanine5.5 (100327), anti-mouse CD8α-FITC (100706), anti-mouse CD45-PE/Cyanine7 (103114), anti-mouse CD11b-PE (101208), anti-mouse F480-FITC (123108), anti-mouse CD86-APC (105012), anti-mouse CD11b-PerCP/Cyanine5.5 (101227), anti-mouse Gr-1-APC (108411), anti-mouse IFN-γ-PE (505807), anti-mouse MHCI-OVA-PE (141603), anti-mouse CD44-PE (103007), anti-mouse CD62L-APC (108411), anti-mouse MHC-I-APC (116517), anti-mouse MHC-II-Brilliant Violet421 (107631), anti-mouse CD40-APC (124611), anti-mouse CD11c-FITC (117305), anti-mouse CD80-Brilliant Violet421 (104725), anti-mouse PD-1-PE (135205), anti-mouse LAG-3- APC (125209), anti-mouse TIM-3-APC (119705), anti-mouse Ly6G-FITC (127605). Isopropyl-β-d-thiogalactoside (I8070), kanamycin (K8020) and TRIzol (R1100) were purchased from Solarbio Biotechnology Co., Ltd. (China). Anti-FLAG antibody (M185-3L) was purchased from MBL (Japan). And the secondary antibody (7076 S) used in the western blot procedure was purchased from Cell Signaling Technology Inc. (USA). Enhanced chemiluminescence reagents (SQ202L-2) was purchased from Epizyme (USA). IRG1-IN-1 (HY-148335) was purchased from MedChemExpress (USA). αPD-L1 (BP0101) was purchased from Bio X cell (USA). Cy5.5-NHS ester (A8103) was purchased from APExBIO (USA). Phalloidin-iFluor 594 conjugate (ab176757) was purchased from Abcam (UK). Hoechst 33,342 (C1022) was purchased from Beyotime (China). Recombinant MMP-2 Protein (902-MP-010) was purchased from R&D Systems (USA). Murine macrophage colony-stimulating factor (M-CSF, 315-02-10), murine IL-4 (214-14-20), and murine IFN-γ (315-05-20) were purchased from Peprotech (USA). Mouse IFN-γ precoated enzyme-linked immunospot assay (ELISPOT) kit (2210005) was purchased from BioLegend Inc. (USA). Erythrocyte lysate (BL503B) was purchased from Biosharp (China). C[13]-labeled ITA (I931004) was purchased from Toronto Research Chemicals (Canada). ELISA kit of CXCL9 (70-EK2143) and CXCL10 (EK268) were purchased from Multisciences (China). ELISA kit of MMP-2 (EK0460) were purchased from Boster Biological Technology Co., Ltd. (China). PD-L1 protein and ELISA detection reagent kit of PD-L1 (SEKM-0259) were purchased from Solarbio Biotechnology Co., Ltd. (China). ELISA kit of TNF-α (1217202) and IL-6 (1210602) were purchased from Dakewe Biotechnology Co., Ltd. (China). The antibody used for immunofluorescence analysis of CD8^+ T cells infiltration within tumor tissues was the following: Anti-CD8α antibody (Abcam, USA, ab217344). The antibody used for immunohistochemical analysis of PD-L1expression within tumor tissues was the following: PD-L1 polyclonal antibody (Proteintech, USA, 17952-1-AP). OVA[257–264] peptide (SIINFEKL) was purchased from Qiangyao Biotechnology Co., Ltd. (China). Plasmid construction and OMV-PDL1nb Preparation The gene encoding ClyA-FLAG-PLGLAG-PDL1nb was cloned into the pET28a vector to construct the expression plasmid (GENEWIZ, China). Then, the plasmid was transformed into Escherichia coli (E. coli) BL21 (DE3) to form engineered bacteria. The engineered bacteria were inoculated into LB medium (50 µg mL^− 1 kanamycin was added) at 37 °C with shaking (180 rpm). When the OD[600] reached 0.6, IPTG (1 mM in LB medium) was added to induce protein expression at 16 °C for 16 h with shaking (180 rpm). The collected bacterial suspension was centrifuged at 5000 ×g for 15 min at 4 °C to remove bacteria, and the supernatant was then filtered through 0.45 μm and 0.22 μm filters (BIOFIL, China, FPE404030 and FPE204030). Next, the filtrate was ultracentrifuged at 150,000 ×g for 3 h at 4 °C to collect OMV-PDL1nb. The pellet was resuspended in phosphate buffered saline (PBS), filtered using a 0.22 μm filter and stored at -80 °C until use. OMV-PDL1nb was characterized using TEM (HITACHI, Japan, HT7700) and DLS (Malvern, UK, Zetasizer Nano ZS90). Western blot analysis OMVs were resuspended in IP lysis buffer and lysed at 4 °C for 30 min, followed by centrifugation at 13,000 × g for 15 min at 4 °C, and the supernatant was collected. The protein concentration of the supernatant was quantified using the BCA protein assay Kit (Thermo Scientific, USA, 23228 and 1859078). Protein samples were electrophoresed using sodium dodecyl sulfide-polyacrylamide gels and subsequently transferred to a polyvinylidene fluoride membrane. The membranes were blocked in 5% nonfat milk and then incubated with an anti-FLAG antibody overnight at 4 °C. The immunoreactive proteins were visualized using enhanced chemiluminescence reagents. Preparation of IN@OMVs, IN@OMV-PDL1nb and IN@OMV-PDL1nb@CaP IRG1-IN-1 was loaded into OMVs or OMV-PDL1nb by electroporation to prepare IN@OMVs and IN@OMV-PDL1nb, respectively. Briefly, 100 µg IRG1-IN-1 and 100 µg OMVs or 100 µg OMV-PDL1nb were separately mixed and suspended in electroporation cuvettes, and electroporation was performed using a MicroPulser Electroporator (Bio-Rad, USA) at 700 V and 50µF. Then, the free IRG1-IN-1 was removed by ultracentrifugation at 150,000 ×g for 3 h at 4 °C. The encapsulation efficiency of IRG1-IN-1 in IN@OMVs or IN@OMV-PDL1nb was determined by UV-vis spectrum. To prepare IN@OMV-PDL1nb@CaP, IN@OMV-PDL1nb (1 mg vesicle protein) was suspended in 1 mL of DMEM overnight at 4 °C, and then calcium chloride (CaCl[2], the final concentration:1 mmol/L) was added to the reaction, followed by incubation at 37 °C for 2 h. Next, the biomineralized OMV-PDL1nb was washed 2–3 times using ultrapure water and following centrifugated at 14,000 × g for 10 min at 4 °C to obtain IN@OMV-PDL1nb@CaP. IN@OMVs, IN@OMV-PDL1nb, and IN@OMV-PDL1nb@CaP were characterized using TEM and DLS. Animals and cell culture Female C57BL/6 mice (6–8-week-old) were purchased from SPF Biotechnology Co., Ltd. (Beijing, China). All animal studies were approved by the Ethics Committee of Tianjin Medical University Cancer Institute and Hospital. B16-F10, B16-OVA, Pan02, MC38 and MC38-OVA Cells were obtained from the National Center for Nanoscience and Technology. B16-F10, B16-OVA, Pan02, MC38 and MC38-OVA cells were cultured in DMEM supplemented with 10% fetal bovine serum, 100 U mL^− 1 penicillin-streptomycin. These cells were tested mycoplasma-negative. The cells were cultured at 37 °C in a humidified atmosphere incubator with 5% CO[2]. To prepare for tumor cell-conditioned medium, MC38, Pan02 or B16-F10 tumor cells were cultured for 24 h, then the supernatant was collected and filtered through 0.45 μm filters and stored at -20 °C until use. Analysis of the binding specificity and affinity between PD-L1nb and PD-L1, and the cleavage efficiency of substrate peptide PLGLAG on OMV-PDL1nb by MMP-2 Firstly, OMVs (12.5 µg vesicle protein per well) and OMV-PDL1nb (12.5 µg vesicle protein per well) were separately spread into 96 well plates and incubated overnight at 4 ℃ to prepare coated ELISA plates. Then, the plates were blocked by 5% nonfat milk. And the concentration of MMP-2 in mouse MC38-OVA tumors was detected using ELISA. Next, MMP-2 was added into the OMV-PDL1nb coated wells and incubated for 0 h, 0.5 h, 1 h, and 2 h at 37 ℃, respectively. Finally, the binding specificity and affinity between PD-L1nb and PD-L1, and the cleavage efficiency of substrate peptide PLGLAG in OMV-PDL1nb by MMP-2 were measured using ELISA as recommended by the manufacturer’s protocol. The released PD-L1 nanobody’s function in binding PD-L1 In brief, OMV-PDL1nb was mixed with MMP-2 for 0 h, 0.5 h, 1 h, and 2 h at 37 ℃, respectively. Then, the supernatant which containing the released PD-L1 nanobody on OMV-PDL1nb cleaved by MMP-2 was collected using ultracentrifugation at 150,000 × g for 3 h at 4 °C. Next, the released PD-L1 nanobody was separately spread into 96 well plates to prepare coated ELISA plates, followed by detection of the released nanobody’s function in binding PD-L1 as mentioned above. Cell binding assay in vitro To assess the binding of OMV-PDL1nb onto tumor cells, OMVs and OMV-PDL1nb were labeled using Cy5.5-NHS. The free Cy5.5-NHS was removed by ultracentrifugation at 150,000 ×g for 3 h at 4 °C. Then, Cy5.5-labeled vesicles (10 µg vesicle protein mL^− 1) were incubated with MC38 cells for 2 h at 4 °C. For blockade of PD-L1, MC38 cells were pre-incubated with anti-PD-L1 (10 µg mL^− 1 αPD-L1) for 12 h at 4 °C. In addition, to detect the expression of MMP-2 substrate peptide PLGLAG, MMP-2 (0.1 µg mL ^− 1) was added after the binding of OMV-PDL1nb onto MC38 cells. Next, the cells were washed three times with PBS, and then the binding state was detected by flow cytometry (BECKMAN, USA). To further visualize OMV-PDL1nb binding onto tumor cells, MC38 cells were treated with the formulations previously mentioned, followed by staining for cytoskeleton actin with phalloidin-iFluor 594 conjugate and labeling of cell nuclei with Hoechst 33,342. Finally, the binding state was detected using CLSM (Zeiss, LSM700, Germany). BMDMs isolation and culture Briefly, bone marrow cells in the femur and tibia of mice were harvested and then cultured in DMEM medium containing 10% fetal bovine serum, 100 U mL^− 1 penicillin-streptomycin, 0.05 mM β-ME and 20 ng mL^− 1 M-CSF. Half of the medium was replaced every 3 days until the bone marrow cells differentiated into M0-type BMDMs on day 7. To obtain M2-polarized BMDMs, M0-type BMDMs were treated with 40 ng mL^− 1 IL-4 for 48 h. Cellular uptake of IN@OMVs and IN@OMV-PDL1nb in vitro To assess cellular uptake, IN@OMVs and IN@OMV-PDL1nb were prepared and labeled using Cy5.5-NHS as previously mentioned. IN@OMVs (12.5 µg vesicle protein mL^− 1) or IN@OMV-PDL1nb (12.5 µg vesicle protein mL^− 1) were incubated with BMDMs at 37 °C for 4 h and then the cell nuclei of BMDMs were stained with Hoechst 33,342. Afterward, the cellular uptake was detected by CLSM. In addition, to detect the cellular time-uptake efficiency of IN@OMVs and IN@OMV-PDL1nb by macrophages, Cy5.5-NHS labeled IN@OMVs (12.5 µg vesicle protein mL^− 1) and IN@OMV-PDL1nb (12.5 µg vesicle protein mL^− 1) were incubated with BMDMs at 37 °C for 0 h, 0.5 h, 1 h, 2 h and 4 h at 37℃, respectively. Next, these BMDMs were stained with Hoechst 33,342. Finally, the cellular time-uptake efficiency was quantified using flow cytometry. Analysis of BMDMs polarization and antigen presentation capacity For BMDMs polarization assay, M0-type BMDMs or M2-polarized BMDMs were incubated with PBS, OMVs (12.5 µg vesicle protein mL^− 1) or IN@OMVs (12.5 µg vesicle protein mL^− 1) for 24 h, respectively. Then, the M1-related marker (CD86) in the CD11b^+ F4/80^+ macrophages was evaluated by flow cytometry. For BMDMs antigen presentation capacity assay, BMDMs were incubated with PBS, OMVs (12.5 µg vesicle protein mL^− 1) or IN@OMVs (12.5 µg vesicle protein mL^− 1) for 24 h, respectively. Then, the antigen presentation capacity-related markers (MHC-I, MHC-II, CD40) in the CD11b^+ F4/80^+ macrophages were detected by flow cytometry. Quantitative real-time PCR analysis BMDMs were incubated with TCM, OMVs (12.5 µg vesicle protein mL^− 1), TCM + IRG1-IN-1 (3.25 µg mL^− 1), TCM + OMVs (12.5 µg vesicle protein mL^− 1) or TCM + IN@OMVs (12.5 µg vesicle protein mL^− 1) for 6 h, respectively. Then, total RNA of cells was extracted using TRIzol reagent and reversely transcribed to synthesize the DNA, followed by qRT-PCR reactions to detect the IRG1, CXCL9 and CXCL10 mRNA expression, respectively. The qRT-PCR primer sequences were as follows: IRG1-F: AGTTTTCTGGCCTCGACCTG, IRG1-R: AGAGGGAGGGTGGAATCTCT; CXCL9-F: GAGCAGTGTGGAGTTCGAGG, CXCL9-R: TCCGGATCTAGGCAGGTTTG; CXCL10-F: AATGAGGGCCATAGGGAAGC, CXCL10-R: AGCCATCCACTGGGTAAAGG; Rn18S-F: CGCGGTTCTATTTTGTTGGT, Rn18S-R: AGTCGGCATCGTTTATGGTC. Intracellular ITA quantification by LC-MS/MS Briefly, BMDMs were incubated with TCM, TCM + IRG1-IN-1 (3.25 µg mL^− 1), TCM + OMVs (12.5 µg vesicle protein mL^− 1) or TCM + IN@OMVs (12.5 µg vesicle protein mL^− 1) for 12 h, respectively. Next, the cells were washed three times with PBS. After that, the cells were fixed by adding 1 ml of 80% (v/v) chilled (-80 °C) methanol containing C[13]-labeled ITA as an internal standard. The cell extracts were analyzed by ultrahigh performance liquid chromatography (Thermo scientific, UAS, vanquish UHPLC) coupled to an orbitrap Mass Spectrometer (Thermo scientific, UAS, Orbitrap Exploris 480). And the intracellular ITA quantification of macrophages within tumor sites was analyzed in the same way. Enzyme-linked immunosorbent assay (ELISA) BMDMs were incubated with TCM, TCM + OMVs (12.5 µg vesicle protein mL^− 1) or TCM + IN@OMVs (12.5 µg vesicle protein mL^− 1) for 24 h, and then the cell-free supernatants were obtained for the cytokines assessment by ELISA kit of CXCL9 and CXCL10 as recommended by the manufacturer′s protocol. T cell migration assay The migration ability of CD8^+ T cells was detected by Transwell assay. In brief, 1 × 10^6 BMDMs were incubated with TCM, TCM + OMVs (12.5 µg vesicle protein mL^− 1) or TCM + IN@OMVs (12.5 µg vesicle protein mL^− 1) for 24 h at the bottom chamber. At the same time, the CD8^+ T cells were isolated from the spleens of wild-type C57BL/6 mice (female, 6–8-week-old) by MojoSort™ Mouse CD8^+ T Cell Isolation Kit (BioLegend, USA, 480008). Then, the CD8^+ T (1 × 10^6) cells were placed in the top chamber. The number of migrated CD8^+ T cells from the top to the bottom chamber was counted after incubation for 12 h at 37 °C in a humidified atmosphere incubator with 5% CO[2]. RNA sequencing Firstly, 1 × 10^6 MC38 cells were injected subcutaneously into the right back of 6–8-week-old female C57BL/6 mice to generate mouse tumor models. On day 9, mice were randomly divided into three experimental groups: Control, OMVs (625 µg vesicle protein/kg body weight) and IN@OMVs (625 µg vesicle protein/kg body weight). OMVs, IN@OMVs were injected intratumorally, the control mice were injected intratumorally with an equal volume of solvent, and tumors were harvested from mice after treatment. Next, and the CD11b^+ F4/80^+ -labeled cells (TAMs) were sorted by FACS. Cellular RNA was isolated from TAMs using the TRIzol reagent and then the samples were immediately shipped frozen to BGI Genomics Co., Ltd. China for transcriptome sequencing. Evaluation of antitumor efficacy in vivo To establish mouse tumor models, 1 × 10^6 MC38-OVA cells were injected subcutaneously into the right back of 6–8-week-old female C57BL/6 mice. On day 9, mice were randomly divided into six experimental groups: Control, αPD-L1(10 mg/kg body weight), IRG1-IN-1 (162.5 µg /kg body weight), IN@OMVs (625 µg vesicle protein/kg body weight), OMV-PDL1nb (625 µg vesicle protein/kg body weight), IN@OMV-PDL1nb (625 µg vesicle protein/kg body weight). IRG1-IN-1 and OMV-based formulations were injected intratumorally, αPD-L1 was injected intraperitoneally in mice, and the control mice were injected intratumorally with an equal volume of solvent. The treatment was given every 3 days for a total of 3 rounds. Tumor size was measured during treatment using a vernier caliper, and the volume of the tumor was calculated as 1/2 × (length × width^2). When the tumor volume exceeded 2000 mm^3, mice were sacrificed and the data were used to generate survival curves. On day 16, a fraction of the mice was sacrificed for adaptive immunity analysis. On day 22, some mice were sacrificed and the tumors were harvested for further immune response analysis. For safety evaluation, the main organs (heart, liver, spleen, lung, and kidneys) of mice were harvested for the H&E staining, while the levels of ALT, AST, ALP, BUN, CREA, and LDH in serum were analyzed. The remaining animals continued to monitor tumor volume up to day 60, and the data were used to generate survival curves. Immune response analysis in vivo For adaptive immunity analysis, splenocytes were harvested and cultured in RPMI1640 medium containing 10% fetal bovine serum. The splenocytes were stimulated with OVA[257–264] peptide (10 µg mL^− 1) at 37 °C in a humidified atmosphere incubator with 5% CO[2] for 16 h, and the IFN-γ^+ cells in CD3^+CD8^+ T cells were detected by flow cytometry, while IFN-γ secretion was analyzed using the mouse IFN-γ precoated ELISPOT kit as recommended by the manufacturer′s protocol. Concurrently, the levels of cytokines (TNF-α, IFN-γ and IL-6) in tumor tissues and the serum of mice were detected to evaluate immune activation. On day 16, the tumor tissues and serum of some mice were harvested. Tumor tissues were homogenized, and the homogenates were centrifuged at 12,000 × g for 15 min at 4 °C to collect the supernatant. Then, the levels of TNF-α, IFN-γ and IL-6 in tumor tissues and the serum of mice were detected using ELISA as recommended by the manufacturer’s protocol. To investigate potential off-target inflammatory effects of IN@OMV-PDL1nb treatment, the proportions of CD3^+ T lymphocytes and neutrophils within spleens of mice were analyzed by flow cytometry. And the levels of lymphocytes and neutrophils in blood of mice were detected using automatic hematology analyzer. In addition, lymphocytes from the tumor-side draining lymph nodes were isolated and then prepared as single cell suspensions. The DCs were stained with anti-mouse CD11c and MHCI-OVA antibodies, followed by detection of antigen presentation in DCs using flow cytometry. On day 22, subcutaneous tumor tissues were prepared as single cell suspensions, and then tumor cells were stained using antibodies. Next, the proportions of CD8^+ T cells, M1-phenotype macrophages, MHC-II^+ macrophages and MDSCs in the tumor tissues were analyzed by flow cytometry. In addition, part of the tumor tissues was sectioned for detection of CD8^+ T infiltration and PD-L1 expression by immunofluorescence and immunohistochemistry, respectively. Evaluation of antitumor efficacy of IN@OMV-PDL1nb@CaP treatment The mouse tumor models were established as mentioned above. On day 5, mice were randomly divided into three experimental groups: Control, IN@OMV-PDL1nb (625 µg vesicle protein/kg body weight) and IN@OMV-PDL1nb@CaP (2500 µg vesicle protein/kg body weight). IN@OMV-PDL1nb was injected intratumorally, IN@OMV-PDL1nb@CaP was injected intravenously in mice, and the control mice were injected with an equal volume of solvent. The treatment was given on day 5 and day 9 for a total of 2 rounds. Tumor size was measured during treatment every two days. The intracellular levels of ITA in macrophages within tumors after treatment measured by LC-MS/MS on day 10. On day 15, the mice were sacrificed and the tumors were harvested to evaluate the antitumor efficacy of IN@OMV-PDL1nb@CaP treatment. Evaluation of immune memory Female C57BL/6 mice (6–8-week-old) were injected subcutaneously into the right back with 1 × 10^6 MC38-OVA cells. On day 7, mice were randomly divided into two experimental groups: Control and IN@OMV-PDL1nb (625 µg vesicle protein/kg body weight). Mice were injected intratumorally with IN@OMV-PDL1nb or the same solvent for 3 rounds. On day 18, tumors in these two groups were removed by surgery. On day 60, the mice were rechallenged by subcutaneously inoculating with 1 × 10^6 MC38-OVA cells or injecting intravenously with 2 × 10^5 B16-OVA cells. The subcutaneous tumor volume was monitored every four days. The mice were sacrificed for immune memory analysis and examination for lung metastasis nodules on day 80. For evaluation of memory T cells, the splenocytes were harvested from the mice and stained with anti-CD3ε, anti-CD8α, anti-CD44 and anti-CD62L antibodies followed by detection using flow cytometry. The proportion of IFN-γ^+ cells in CD3^+CD8^+ T cells within splenocytes was detected as previously mentioned. Statistical analysis Data were analyzed using GraphPad Prism software version 8 and presented as the mean ± standard deviation (SD) and represented at least three independent experiments. The P values were determined using a two-tailed unpaired t test or one-way ANOVA with a Tukey post-hoc test. The P values of survival analysis were calculated using log-rank (Mantel-Cox) test. And the values of P < 0.05 were considered statistically significant. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, n.s., no significance. Results Preparation and characterization of OMV-PDL1nb, in@omvs and IN@OMV-PDL1nb To obtain OMV-PDL1nb, the genes encoding ClyA (one of the most abundant proteins on the surface of OMVs), PLGLAG and PDL1nb were recombined in the expression plasmid pET28a. The resulting fusion protein was then expressed in E.coli BL21 (DE3) (Fig. [87]2A). Western blot analysis confirmed the expression of ClyA-PLGLAG-PDL1nb in OMV-PDL1nb following Isopropyl-β-d-thiogalactoside (IPTG) induction, as evidenced by the detection of the FLAG tag (Fig. [88]2B). Transmission electron microscopy (TEM) and dynamic light scattering (DLS) were employed to characterize the OMVs generated from the original E.coli and OMV-PDL1nb generated from the engineered E.coli. Both OMVs and OMV-PDL1nb exhibited regular spherical double-layer membrane structures, with average diameters of 20–30 nm (Fig. [89]2C). The zeta potential of OMVs was approximately − 20 mV, while that of OMV-PDL1nb measured around − 24 mV (Fig. [90]2D). We then performed an ELISA to evaluate the binding specificity and affinity of PD-L1nb (on OMV-PDL1nb) for PD-L1. Our results showed that PD-L1 can efficiently bind to OMV-PDL1nb, significantly more than to OMVs. Additionally, the substrate peptide PLGLAG on OMV-PDL1nb was rapidly cleaved by MMP-2, with a cleavage rate of approximately 74% at 0.5 h, and increasing to nearly complete cleavage (99%) at 2 h (Supplementary Fig. [91]1). Concurrently, we collected the PD-L1 nanobody released from OMV-PDL1nb cleaved by MMP-2 at different time points and assessed its functionality in binding PD-L1 using ELISA. The results demonstrate that the released PD-L1 nanobody retains full functionality and effectively binds to PD-L1 (Supplementary Fig. [92]2). Subsequently, an in vitro tumor cell binding assay was performed to evaluate the affinity between PD-L1nb (on OMV-PDL1nb) and PD-L1 (expressed on tumor cells). In this experiment, both OMVs and OMV-PDL1nb were labeled with Cy5.5. Utilizing flow cytometry and confocal laser scanning microscopy (CLSM), it was demonstrated that OMV-PDL1nb effectively adhered to the surface of MC38 cells. Furthermore, pre-incubation with anti-PD-L1 antibodies significantly inhibited the binding of OMV-PDL1nb to the MC38 cell’s surface, thereby confirming the specificity of the interaction between PD-L1nb (on OMV-PDL1nb) and PD-L1 (on tumor cells). Additionally, OMV-PDL1nb bound to tumor cells was effectively cleaved by MMP-2, further validating the expression of ClyA-PLGLAG-PDL1nb in OMV-PDL1nb (Fig. [93]2E-F). Fig. 2. [94]Fig. 2 [95]Open in a new tab Characterization of OMV-PDL1nb, IN@OMVs and IN@OMV-PDL1nb. (A) Schematic representation of the pET28a-ClyA-PLGLAG-PDL1nb expression plasmid construct. (B) Western blot analysis of ClyA-PLGLAG-PDL1nb expression in OMV-PDL1nb using an anti-FLAG antibody. Isopropyl-β-d-thiogalactoside (IPTG) was utilized as the inducer for protein expression. (C) TEM images and size distribution of OMVs and OMV-PDL1nb. Scale bar = 100 nm. d., diameter. (D) The zeta potentials of OMVs and OMV-PDL1nb measured by DLS (n = 3). (E-F) Flow cytometry E) and CLSM F) analysis of the affinity between PDL1nb (on OMV-PDL1nb) and PD-L1 (on MC38 cells). MC38 cells were treated with the indicated formulations for 2 h. In group III, MC38 cells were pre-incubated with anti-PDL1 for 12 h to block PD-L1. In group IV, MMP-2 was added after the binding of OMV-PDL1nb to tumor cells. Cell nuclei of MC38 were stained with Hoechst 33342 (blue). The cytoskeleton actin of MC38 cells were stained with phalloidin-iFluor 594 conjugate (green) and OMVs, OMV-PDL1nb were labeled with Cy5.5(red). Scale bar = 20 μm. (G) TEM images and size distribution of IN@OMVs and IN@OMV-PDL1nb. Scale bar = 100 nm. d., diameter. (H) The zeta potentials of IN@OMVs and IN@OMV-PDL1nb measured by DLS (n = 3). (I) IRG1-IN-1 encapsulation efficiency of IN@OMVs and IN@OMV-PDL1nb were determined using UV-visible spectrum (n = 3). (J-K) Cellular uptake of IN@OMVs and IN@OMV-PDL1nb in BMDMs were detected using CLSM. Cell nuclei of BMDMs were stained with Hoechst 33342 (blue), while IN@OMVs and IN@OMV-PDL1nb were labeled with Cy5.5 (red). Scale bar = 50 μm. The average Cy5.5 fluorescence intensity per macrophage of the ELISA experiment in the IN@OMVs group was used as the control. Data were processed by GraphPad Prism software version 8 and presented as the mean ± SD. The P value was calculated using a two-tailed unpaired t test. n.s., no significance Based on the structural characteristics of OMVs with internal cavity structure, we encapsulated IRG1-IN-1 into OMVs or OMV-PDL1nb to form IN@OMVs or IN@OMV-PDL1nb via electroporation, respectively. TEM observations and DLS analysis revealed that both IN@OMVs and IN@OMV-PDL1nb exhibited regular spherical double-layer membrane structures, with diameters of approximately 20–30 nm (Fig. [96]2G). The zeta potential of IN@OMVs was approximately − 23 mV, while that of IN@OMV-PDL1nb was around − 27 mV (Fig. [97]2H). Furthermore, UV-visible spectrum determined that the encapsulation efficiency of IRG1-IN-1 in both IN@OMVs and IN@OMV-PDL1nb was about 26% (Fig. [98]2I). Subsequently, the cellular uptake efficiency of IN@OMVs and IN@OMV-PDL1nb by bone marrow-derived macrophages (BMDMs) was assessed by CLSM, it was demonstrated that IN@OMVs and IN@OMV-PDL1nb could be effectively uptaken by BMDMs, and the uptake of IN@OMV-PDL1nb was higher, indicating that the PD-L1nb might enhance its macrophage targeting (Fig. [99]2J-K). Moreover, flow cytometry analysis revealed a time-dependent increase in cellular uptake of both IN@OMVs and IN@OMV-PDL1nb by macrophages, with uptake efficiencies of 19% and 21% at 0.5 h, respectively, and 84% and 88% at 4 h (Supplementary Fig. [100]3). It was evident that the rate of cellular uptake was significantly slower than the cleavage rate of PD-L1nb from OMV-PDL1nb, indicating that PD-L1nb on OMV-PDL1nb could be efficiently released before phagocytosis by macrophages. Evaluation of the role of IN@OMVs in vitro Firstly, we treated BMDMs with different types of tumors conditioned medium (TCM) and OMVs and found that both of these treatments could induce high IRG1 mRNA expression in BMDMs (Supplementary Fig. [101]4) [[102]23]. Next, we investigated the IRG1 mRNA expression and ITA accumulation in BMDMs treated by IRG1-IN-1 and IN@OMVs using qRT-PCR and liquid chromatography-tandem mass spectrometry (LC-MS/MS), respectively. It was demonstrated that MC38-TCM highly induced IRG1 expression and ITA accumulation in BMDMs, and IRG1-IN-1 treatment significantly reduced their expression levels. However, the combination of MC38-TCM and OMVs treatment further increased the expression levels of IRG1 and ITA. Encouragingly, treatment with IN@OMVs significantly reduced their expression levels, closed to the TCM + IRG1-IN-1 treatment group, indicating the advantages of OMV-based nanoplatforms for drug delivery (Fig. [103]3A-B). Fig. 3. [104]Fig. 3 [105]Open in a new tab The effect of IN@OMVs on macrophages in vitro. BMDMs were treated with the indicated formulations. TCM, MC38 cell-tumor conditioned medium. (A-B) IRG1 mRNA expression A) and ITA accumulation B) in BMDMs measured by qRT-PCR (n = 4) and LC-MS/MS (n = 3), respectively. (C-D) Transwell assay was used to analyze the number of CD8^+ T cells migration (n = 3). (E-H) CXCL9 E-F) and CXCL10 G-H) expression in BMDMs measured by qRT-PCR (E, G; n = 4) and ELISA (F, H; n = 3). (I-K) Analysis of antigen presentation of macrophages (n = 3). Flow cytometry evaluated the antigen presentation-related markers (MHC-I, MHC-II, CD40) in the CD11b^+F4/80^+ BMDMs. (L-M) Analysis of macrophages M1 polarization (n = 3). Flow cytometry evaluated the M1-related marker (CD86) in the CD11b^+F4/80^+ BMDMs. Data were processed by GraphPad Prism software version 8 and presented as the mean ± SD. The P values were determined using one-way ANOVA with a Tukey post-hoc test. **P < 0.01, ***P < 0.001, and ****P < 0.0001, n.s., no significance Subsequently, we examined the effect of macrophages treated with OMVs and IN@OMVs on the migration ability of CD8^+ T cells by transwell assay, which showed that OMVs treatment increased the number of CD8^+ T cells migrating, while macrophages treated with IN@OMVs further increased CD8^+ T cell migration (Fig. [106]3C-D). The results of quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assays (ELISA) showed that OMVs treatment increased macrophage-derived CXCL9 and CXCL10 mRNA expression and extracellular secretion. Moreover, the mRNA expression and extracellular secretion of CXCL9 and CXCL10 in macrophages were further increased after IN@OMVs treatment (Fig. [107]3E-H). These results indicate that IN@OMVs can efficiently inhibite IRG1 expression and ITA accumulation in macrophages, thereby increasing the expression of macrophage-derived CXCL9 and CXCL10 and enhancing the ability of macrophages to promote CD8^+ T cell migration. As an important antigen presenting cell, macrophages play a crucial role in antigen presentation [[108]24]. The antigen presentation capacity of macrophages was evaluated by flow cytometry, and the results showed that compared with the PBS (control) and OMVs-treated groups, IN@OMVs treatment significantly increased the antigen presentation capacity of macrophages, as indicated by higher expression of antigen presentation-related markers MHC-I, MHC-II and CD40 on the surface of macrophages (Fig. [109]3I-K, Supplementary Fig. [110]5). In addition, the results of flow cytometry demonstrated that IN@OMVs treatment could significantly promote the M0-type and M2-polarized macrophages to M1-phenotype macrophages (Fig. [111]3L-M). These results indicate that IN@OMVs treatment can reduce IRG1 mRNA expression and ITA accumulation levels in macrophages induced by tumor cells and OMVs, thereby reversing macrophage dysfunction caused by excessive ITA accumulation. Transcriptome profiling of TAMs after IN@OMVs treatment To further investigate the mechanisms by which IN@OMVs affect macrophages, we isolated TAMs from the tumors of control, OMVs and IN@OMVs treated mice using fluorescence-activated cell sorting (FACS) for transcriptome analysis (Fig. [112]4A). The results indicated that OMVs treatment led to an increased expression of IRG1 in TAMs compared with the control group, whereas IN@OMVs treatment effectively inhibited the expression of IRG1 in these cells. Additionally, OMVs treatment enhanced the expression of genes associated with chemotaxis, including Ccl2, Ccl7, Ccl8, Ccl12, Cxcl9, and Cxcl10. Notably, these genes were even more significantly upregulated following treatment with IN@OMVs. Simultaneously, IN@OMVs treatment markedly increased the expression of antigen presentation-related genes in TAMs, such as Cd40, Cd86, Icam1, H2-M2, H2-Aa, H2-Ab1, Tap1, and Tap2. Furthermore, the results showed that OMVs treatment reduced the expression of genes associated with M2 polarization, such as Cd163 and protein kinase AMP-activated catalytic subunit alpha 1 (Prkaa1), with IN@OMVs treatment leading to even more significant downregulation of these genes [[113]25]. IN@OMVs treatment markedly decreased the expression of Mmp8 and Mmp9, genes that promote tumor metastasis [[114]26], compared to other treatment groups. In addition, IN@OMVs treatment significantly reduced the expression of carcinoembryonic antigen-related cell-adhesion molecule 1 (ceacam1) in tumor-associated macrophages (TAMs), a molecule involved in angiogenesis and tumorigenesis (Fig. [115]4B) [[116]27, [117]28]. These findings corroborate the results of previous experiments in vitro. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses revealed that OMVs treatment enhanced the activation of antigen processing and presentation pathways, as well as the NF-κB pathway and response to interferon-β (IFN-β) in TAMs compared to the control group (Fig. [118]4C-E). In comparison with the control and OMVs treatment, the IN@OMVs treatment significantly enhanced the activation of pathways related to the response to IFN-β, antigen processing and presentation, and NF-kB in TAMs (Fig. [119]4F-H, Supplementary Fig. [120]6). Concurrently, GO pathway enrichment analysis revealed that OMVs treatment downregulated cell growth and cell adhesion pathways in TAMs compared to the control group (Supplementary Fig. [121]7A). Notably, IN@OMVs treatment further decreased the activation of pathways related to angiogenesis, cell-cell adhesion, and growth factor activity in TAMs (Supplementary Fig. [122]7B-C). Additionally, Gene Set Enrichment Analysis (GSEA) demonstrated downregulation of hypoxia and epithelial-mesenchymal transition (EMT) pathways after IN@OMVs treatment compared to OMVs treatment, while Reactome gene set analysis indicated suppression of IL-4, IL-13, and IL-10 signaling pathways (Supplementary Fig. [123]7D, and F-G). Similar results were observed in the comparison between IN@OMVs and the control group, with angiogenesis also being downregulated (Supplementary Fig. [124]7E, and H-I). These findings suggest that IN@OMVs treatment modulates the tumor microenvironment to inhibit tumor progression and immune escape. Previous studies have indicated that the activation of the NF-kB pathway in TAMs can lead to the expression of downstream genes such as Irg1, Cxcl9, and Cxcl10. ITA, the downstream metabolite of IRG1, has the capacity to inhibit the persistent activation of the NF-kB pathway, resulting in decreased expression of CXCL9 and CXCL10, which ultimately hinders T cell infiltration [[125]8, [126]9]. In this study, we demonstrated that OMVs can also activate the NF-kB pathway in TAMs, leading to elevated IRG1 expression and subsequent suppression of CXCL9 and CXCL10. IN@OMVs, leveraging its efficient drug delivery capabilities, maintained the activation of the NF-kB pathway while delivering IRG1-IN-1 into TAMs, thereby reducing ITA accumulation. This reduction eliminated the inhibition of sustained NF-kB pathway activation and the expression of CXCL9 and CXCL10 by ITA (Fig. [127]4I). Furthermore, IN@OMVs also facilitated the activation of pathways related to antigen processing and presentation in TAMs. Fig. 4. [128]Fig. 4 [129]Open in a new tab Transcriptome analysis of TAMs in mice after treatment. (A) Schematic illustration of the experiment schedule. (B) Gene expression heatmap in TAMs from the MC38 tumor-bearing mice (n = 3). (C-E) Transcriptomic changes in TAMs after OMVs treatment compared with the control group. C) The volcano plot showed the number of differentially expressed genes distribution. P value (-Log[10], y axis) and the fold change (Log[2], x axis). Significant changes (P value < 0.05,|Log[2] Fold change| > 0.3) were indicated in red (upregulation) and blue (downregulation). D) Gene ontology (GO) enrichment pathways of up-regulated genes. Ontology: Biological process (BP), Cell component (CC) and Molecular function (MF). E) Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways of up-regulated genes. (F-H) Transcriptomic changes in TAMs after IN@OMVs treatment compared with OMVs treatment group. F) The volcano plot showed the number of differentially expressed genes distribution. P value (-Log[10], y axis) and the fold change (Log[2], x axis). Significant changes (P value < 0.05,|Log[2] Fold change| > 0.3) were indicated in red (upregulation) and blue (downregulation). G) GO enrichment pathways of up-regulated genes. H) KEGG enrichment pathways of up-regulated genes. (I) Schematic illustration of IN@OMVs triggering TLR4-NF-κB pathway activation in macrophages Antitumor effects of and TME remodeling by IN@OMV-PDL1nb In order to evaluate the antitumor effects of IN@OMV-PDL1nb, we constructed a subcutaneous tumor-bearing mouse model using MC38-OVA cells (murine colon cancer cells expressing ovalbumin). The mice received three intratumoral injections of either αPD-L1, IRG1-IN-1, IN@OMVs, OMV-PDL1nb, or IN@OMV-PDL1nb, as illustrated in Fig. [130]5A. Initially, we assessed the adaptive immunity induced by the IN@OMV-PDL1nb treatment on day 16. Flow cytometry analysis revealed a higher proportion of MHCI-OVA^+ DCs in the tumor-draining lymph nodes of the IN@OMV-PDL1nb treatment group on the seventh day post-treatment, indicating that IN@OMV-PDL1nb effectively enhanced antigen presentation by DCs (Fig. [131]5B). Additionally, we observed a significant increase in both the volume and weight of the spleen in the IN@OMV, OMV-PDL1nb, and IN@OMV-PDL1nb treatment groups, which may be attributed to the systemic immune enhancement provided by OMVs (Supplementary Fig. [132]8A-B). Furthermore, analysis of IFN-γ secretion through flow cytometry and enzyme-linked immunospot (ELISPOT) assays demonstrated that IN@OMV-PDL1nb treatment significantly activated OVA-specific CD8^+ T cells within splenocytes (Fig. [133]5C, Supplementary Fig. [134]9A-B). These specific CD8^+ T cells can effectively recognize and kill MC38-OVA cells after they infiltrating into the tumor tissues. And cytokine levels (TNF-α, IFN-γ, and IL-6) were measured in tumor tissues and serum of mice following treatment. IN@OMV-PDL1nb treatment significantly enhanced the secretion of these cytokines compared to other treatments, suggesting its ability to promote immune activation (Supplementary Fig. [135]10). Simultaneously, we assessed immune cell populations in non-tumor tissues to evaluate potential off-target inflammatory effects of IN@OMV-PDL1nb treatment, and the results showed mild elevations in systemic immune cell levels, all remaining within the normal range, indicating no off-target inflammatory effects while activating the immune system (Supplementary Fig. [136]11). Additionally, the volume of the draining lymph nodes on the tumor side in mice significantly increased following IN@OMV-PDL1nb treatment, indicating an accumulation of immune cells in these lymph nodes (Supplementary Fig. [137]12). These results suggest that after IN@OMV-PDL1nb treatment, tumor cells are eliminated, releasing tumor antigens that are subsequently taken up and processed by DCs in the tumor-draining lymph nodes. These processed antigens are then presented to T cells, thereby inducing a robust tumor antigen-specific adaptive immune response. Furthermore, the LC-MS/MS assay demonstrated that IN@OMV-PDL1nb treatment significantly reduced ITA levels in macrophages within the tumors, which is critical for promoting the infiltration of CD8^+ T cells into the tumor tissues (Supplementary Fig. [138]13). At the endpoint of this experiment, we assessed the antitumor effects of the indicated treatment approaches. The results indicated that the IN@OMV-PDL1nb treatment was more effective in inhibiting tumor growth compared with αPD-L1, IRG1-IN-1, IN@OMVs, and OMV-PDL1nb treatments. Mice treated with IN@OMV-PDL1nb exhibited significantly smaller tumor volumes than those in the other groups (Fig. [139]5D-E, Supplementary Fig. [140]14). To investigate the role of IN@OMV-PDL1nb treatment in the TME, we assessed the infiltration of immune cells in the tumor tissue post-treatment using flow cytometry. The IN@OMV-PDL1nb treatment notably increased the proportion of infiltrating CD8^+ T cells in the tumor compared with other treatments (Fig. [141]5F). Concurrently, IN@OMV-PDL1nb treatment significantly reduced the proportion of exhausted CD8^+ T cells in the tumor microenvironment, enhancing their functional state (Supplementary Fig. [142]15), thereby reinforcing its robust anti-tumor efficacy. Additionally, this treatment significantly enhanced the proportion of M1-phenotype macrophages within the tumor tissues and elevated the expression of markers associated with macrophage antigen presentation (Fig. [143]5G-H). Further flow cytometry analysis revealed that IN@OMV-PDL1nb treatment reduced the proportion of infiltrating myeloid-derived suppressor cells (MDSCs) in the tumor tissues (Fig. [144]5I). And the survival time of mice treated with IN@OMV-PDL1nb was significantly prolonged (Fig. [145]5J). Furthermore, the assay of immunofluorescence revealed that IN@OMV-PDL1nb treatment markedly increased the proportion of infiltrating CD8^+ T cells in tumor tissues (Fig. [146]5K). Immunohistochemical analyses indicated that IN@OMV-PDL1nb treatment significantly reduced PD-L1 expression in tumor tissues (Fig. [147]5L). In addition, we evaluated the systemic toxicity associated with IN@OMV-PDL1nb treatment. The hematoxylin and eosin (H&E) staining images of the main organs in mice demonstrated that IN@OMV-PDL1nb treatment did not result in noticeable organ damage (Supplementary Fig. [148]16). Concurrently, the assessment of serum biochemical indices, including alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), blood urea nitrogen (BUN), creatinine (CREA), and lactate dehydrogenase (LDH), further confirmed the safety of the IN@OMV-PDL1nb treatment (Supplementary Fig. [149]17). In summary, these experimental results suggest that IN@OMV-PDL1nb treatment effectively induces adaptive immunity, thereby remodeling the TME and generating potent anti-tumor effects. Fig. 5. [150]Fig. 5 [151]Open in a new tab The antitumor effects of IN@OMV-PDL1nb in MC38-OVA subcutaneous tumor model. (A) Schematic illustration of the experiment schedule. (B) The proportion of MHCI-OVA^+ cells in CD11c^+ DCs in the tumor side draining lymph nodes on day 16 (n = 5). (C) The proportion of IFN-γ^+ cells in CD3^+CD8^+ T cells in spleen on day 16 (n = 5). (D) The images of tumors after treatment (n = 6). Scale bar = 1 cm. (E) Tumor weight (n = 6). (F-I) The proportions of CD8^+ T cells, M1-phenotype macrophages, MHC-II^+ macrophages and MDSCs within tumor tissues (n = 6). (J) Survival curves (n = 6). (K) Immunofluorescence (IF) staining of CD8^+ cells in tumors of mice. Scale bar = 20 μm. (L) Immunohistochemical (IHC) images showing the expression of PD-L1 in tumor tissue of mice. Scale bar = 50 μm. Data were processed by GraphPad Prism software version 8 and presented as the mean ± SD. The P values of survival analysis were calculated using log-rank (Mantel-Cox) test. And other P values were determined using one-way ANOVA with a Tukey post-hoc test. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001, n.s., no significance Long-term immune memory induced by IN@OMV-PDL1nb The challenge of tumor immunotherapy lies not only in whether the selected treatment regimen can achieve clinical cure or remission, but also in the effective control of long-term tumor recurrence and metastasis. Previous studies have reported that OMVs can induce long-term immune memory effects, thanks to their natural size advantages and efficient immune activation ability on DCs. OMVs can be efficiently drained into lymph nodes. In addition, the abundant PAMPs molecules on the surface of OMVs can effectively promote the rapid maturation of DCs in lymph nodes. Mature DCs can process, present tumor antigens, and activate T cells, thereby inducing strong adaptive immunity and long-term immune memory effects. The adaptive immune and immune memory effects induced by OMVs are significantly superior to conventional immunotherapy strategies [[152]29, [153]30]. Thus, we investigated the immune memory effects induced by IN@OMV-PDL1nb treatment (Fig. [154]6A). We constructed the subcutaneous MC38-OVA tumor-bearing mice model and treated the mice for three times. Compared with the control group, IN@OMV-PDL1nb treatment significantly inhibited the tumor growth (Fig. [155]6B). Following this treatment, we surgically excised subcutaneous tumors from the mice. Then we inoculated mice subcutaneously with MC38-OVA or intravenously with B16-OVA cells on day 60. The results indicated that in mice treated with IN@OMV-PDL1nb, the re-challenged subcutaneous tumors regressed almost completely (Fig. [156]6C). Moreover, the mice treated with IN@OMV-PDL1nb had significantly fewer metastasis nodules in the lungs compared with the control group (Fig. [157]6D-E). Additionally, flow cytometry analysis revealed that the proportions of effector memory T cells and central memory T cells in the splenocytes of IN@OMV-PDL1nb treated mice were significantly elevated compared with the control mice on day 80 (Fig. [158]6F-H). Compared with the control group, the proportion of IFN-γ^+ cells in CD3^+CD8^+ T cells within spleens of the IN@OMV-PDL1nb treated mice was significantly increased, suggesting that the immune system of the mice treated with IN@OMV-PDL1nb could still recognize and response to tumor antigens efficiently (Fig. [159]6I). These findings imply that IN@OMV-PDL1nb treatment can elicit antigen-specific immune memory, which contributes to a long-term tumor prevention effect against tumor recurrence and metastasis. Fig. 6. [160]Fig. 6 [161]Open in a new tab Evaluation of long-term immune memory induced by IN@OMV-PDL1nb. (A) Schematic illustration of the experiment schedule. (B) Tumor growth curves of the first subcutaneously implanted MC38-OVA cells in each mouse (n = 5). (C) Tumor growth curves of the re-challenge of subcutaneous inoculation with MC38-OVA cells in each mouse (n = 5). (D-E) The images of lung and numbers of metastatic nodules in each mouse re-challenged with intravenous injection of B16-OVA cells (n = 5). (F-H) The proportions of effector memory T cells and central memory T cells in spleen (n = 5). (I) The proportion of IFN-γ^+ cells in CD3^+CD8^+ T cells in splenocytes (n = 5). Data were processed by GraphPad Prism software version 8 and presented as the mean ± SD. The P value was calculated using a two-tailed unpaired t test. **P < 0.01, ****P < 0.0001 Evaluation of antitumor efficacy by IN@OMV-PDL1nb@CaP treatment in vivo In our study, we initially employed intratumoral injection of IN@OMV-PDL1nb, which demonstrated potent anti-tumor effects. Building upon this, following the methodology established in our previous work [[162]31], we encapsulated IN@OMV-PDL1nb in CaP to form IN@OMV-PDL1nb@CaP, which retained the spherical double-layer membrane structure with an average diameter of approximately 120 nm (Fig. [163]7A-C), and a zeta potential of around − 15 mV (Fig. [164]7D). To further evaluate its potential for systemic administration, we developed a subcutaneous tumor-bearing mouse model using MC38-OVA cells. The mice were administered two intratumoral injection of IN@OMV-PDL1nb or intravenous (i.v.) injections of IN@OMV-PDL1nb@CaP. The results showed that both formulations effectively inhibited tumor growth, with similar tumor volumes observed in the IN@OMV-PDL1nb@CaP and IN@OMV-PDL1nb treatment groups (Fig. [165]7E-H). Furthermore, LC-MS/MS analysis revealed that IN@OMV-PDL1nb@CaP treatment significantly reduced ITA levels in macrophages within the tumor tissues (Fig. [166]7I), confirming the therapeutic potential of this formulation. These results suggest that while intratumoral injection offers direct therapeutic benefits, encapsulation of IN@OMV-PDL1nb in CaP also allows for enhanced tumor targeting via intravenous administration, expanding the potential application of this dual-target nanoplatform. Fig. 7. [167]Fig. 7 [168]Open in a new tab The antitumor effects of IN@OMV-PDL1nb@CaP in MC38-OVA subcutaneous tumor model. (A) The Schematic illustration of the preparation of IN@OMV-PDL1nb@CaP. (B-C) TEM image and size distribution of OMV-PDL1nb@CaP. Scale bar = 100 nm. d., diameter. (D) The zeta potential of OMV-PDL1nb@CaP measured by DLS (n = 3). (E) Schematic illustration of IN@OMV-PDL1nb@CaP treatment. (F) Tumor growth curves in each mouse (n = 5). (G) The images of tumors after treatment (n = 5). (H) Tumor weight (n = 5). (I) The intracellular levels of ITA in macrophages within tumors after treatment measured by LC-MS/MS on day 10 (n = 3). Data were processed by GraphPad Prism software version 8 and presented as the mean ± SD. The P values were determined using one-way ANOVA with a Tukey post-hoc test. ***P < 0.001,****P < 0.0001, n.s., no significance Discussion Tumor immunotherapy, particularly through the use of PD-1/PD-L1 inhibitors, has demonstrated clinical efficacy in certain tumor types; however, the overall therapeutic effect of these inhibitors remains limited [[169]32–[170]34]. The low response rates associated with PD-1/PD-L1 inhibitors may be attributed to the immunosuppressive nature of the tumor microenvironment, which mainly includes inadequate T cell infiltration in tumor sites [[171]35, [172]36]. Consequently, activating immune remodeling within the tumor microenvironment, in conjunction with PD-1/PD-L1 inhibitor therapy, represents a promising strategy for enhancing tumor immunotherapy [[173]37–[174]39]. ITA, a byproduct of the Krebs cycle, accumulates within macrophages following the activation of the NF-κB pathway by tumor cells. ITA inhibits TET DNA dioxygenase, subsequently reducing the secretion of chemokines, which ultimately disrupts CD8^+ T cells infiltration into tumor sites [[175]8, [176]9, [177]40, [178]41]. Therefore, the targeted inhibition of ITA production within macrophages is crucial for remodeling the TME. OMVs contain abundant PAMPs and possess a strong capacity for immune activation, effectively engaging TLRs such as TLR2, TLR4, and TLR5, among other innate immune pathways [[179]42–[180]45]. Furthermore, OMVs are readily obtainable and amenable to genetic engineering modifications, which has garnered increasing interest in the field of tumor immunotherapy. OMVs can serve as vectors for tumor vaccines or immune activators to elicit anti-tumor responses [[181]46, [182]47]. In addition, their internal cavity structure and easily phagocytosis by macrophages, positioning them as excellent drug delivery vehicles. In our previous research, we modified OMVs to investigate their potential as therapeutic tumor vaccines and activators of tumor immunotherapy, yielding promising results [[183]29, [184]30, [185]48, [186]49]. As a significant component of OMVs, LPS can stimulate tumor immunity via the TLR4-NF-kB pathway [[187]50]. Notably, in this study, we observed that OMVs induced IRG1 overexpression and ITA accumulation within macrophages while activating the expression of chemokine-related genes. This finding suggests that LPS-mediated immune activation in OMVs may represent a double-edged sword; while LPS activates the TLR4-NF-kB pathway in macrophages to enhance immunity, it concurrently promotes the production of immunosuppressive factors, such as ITA, which accumulate in macrophages and potentially inhibit the efficacy of immune activation. This phenomenon may explain the limited effectiveness of OMVs monotherapy in tumor treatment. Currently, efforts to reduce the endotoxin levels of LPS in OMVs are primarily achieved through genetic modification of the parent strain, which introduces new challenges, particularly regarding the diminished capacity of LPS to activate immunity [[188]29, [189]51, [190]52]. Therefore, a strategic approach that preserves LPS-induced immune activation while mitigating the associated immunosuppression could enhance the anti-tumor efficacy of OMVs. Conclusions In conclusion, IN@OMV-PDL1nb nanoplatform effectively inhibits TAM-derived ITA through IRG1 suppression while concurrently blocking PD-L1 to restore CD8^+ T cell infiltration and cytotoxic functions. The platform leverages the unique biological properties of engineered OMVs for precise delivery of therapeutic agents to TAMs, ensuring targeted modulation of the TAMs in the TME. In preclinical models, IN@OMV-PDL1nb not only inhibited tumor growth and enhanced survival but also promoted robust antigen presentation, adaptive immune activation, and long-term immune memory. These findings underscore the potential of IN@OMV-PDL1nb to overcome critical barriers in T cell-based tumor immunotherapy, offering a promising strategy for improving tumor treatment outcomes. Electronic supplementary material Below is the link to the electronic supplementary material. [191]Supplementary Material 1^ (2.5MB, pdf) Acknowledgements