High PSMA expression and M2 macrophages infiltration in PCa
Because Magic is designed to specifically bind to PSMA, we first checked the expression of PSMA at the protein level based on the samples in the Human Protein Atlas (HPA) database (https://www.proteinatlas.org). PSMA was found to be exclusively expressed in prostatic glandular cells (Fig. 1a), and its expression level was positively correlated with the histological grade of PCa and, thus, tumor aggressiveness and therapeutic outcome (Fig. 1b and Supplementary Fig. 1), indicating that PSMA is not only a promising biomarker in the diagnosis and therapy of PCa, but also useful in determining the prognosis of patients with PCa.
Considering that M2 macrophages contribute to the pro-tumoral environment and are associated with adverse clinical outcomes in many types of cancers, we next investigated the involvement of M2 macrophages in PCa progression. CIBERSORT was applied to statistically estimate the relative proportions of immune cell subsets among different grades of PCa samples from The Cancer Genome Atlas (TCGA) dataset. As shown in Fig. 1c and Supplementary Fig. 2, the infiltration levels of macrophages were higher in tumor tissues than in the normal prostate tissues. Notably, M2 macrophages not only accounted for a large proportion of tumor-infiltrating immune cells, but also were significantly enriched in high-grade compared with low-grade PCa tissues, suggesting that they may play a key role in the progression of PCa. Immunofluorescence staining of tumor tissues from patients with PCa showed consistent results. Compared with adjacent normal tissues, the number of CD68+ macrophages, especially CD68+/CD206+ M2 macrophages, was markedly increased in tumors and positively correlated with advanced tumor stage, confirming the prognostic value of M2 macrophages in PCa (Fig. 1d and Supplementary Fig. 3,4). Notably, PSMA expression also increased in parallel with tumor progression, as shown by immunohistochemistry (IHC) testing performed on tumor tissues (Fig. 1e). Further analysis revealed a positive correlation between M2 macrophage infiltration and PSMA expression (Fig. 1f and Supplementary Fig. 5a), suggesting that it might be possible to combine PSMA-targeted therapy with immunoregulation of the tumor microenvironment to promote enhanced anti-PCa treatment based on PSMA recognition. In addition, M2 macrophages have been reported to participate in epithelial-mesenchymal transition (EMT) and tumor angiogenesis by secreting the tumor-promoting cytokines, transforming growth factor-β (TGF-β) and vascular endothelial growth factor C (VEGFC), resulting in enhanced tumor invasion and metastasis.45 Therefore, we analyzed the expression levels of TGF-β and VEGFC in PCa. The results shown in Fig. 1g clearly demonstrate that both TGF-β and VEGFC are positively correlated with M2 macrophage infiltration in PCa. Patients with high TGF-β or VEGFC expression appeared to have much poorer disease-free survival (DFS) than those with low TGF-β or VEGFC expression (Fig. 1h). Consistently, reduced E-cadherin (ECAD) expression (a hallmark of EMT) and Endoglin (ENG, CD105) overexpression (a marker of tumor angiogenesis) were significantly associated with shorter DFS in patients with PCa (Supplementary Fig. 5b). All these results indicate that M2 macrophages are probably involved in PCa progression via tumor-promoting cascades (i.e., M2 macrophage → cytokines → phenotypes → prognosis). Therefore, depletion or re-education of M2 macrophages may be a promising modality for immunomodulatory anti-PCa treatment.
Preparation and characterization of Magic
Inspired by the importance of PSMA in PCa treatment, and in light of the high affinity of gy-1 towards PSMA,37–39 we envisioned that a macrophage-based platform with enhanced PSMA-targeting ability might be obtained through the integration of gy-1 with macrophages. To confirm this, we genetically engineered Raw264.7 cells to stably TM-express gy-1 through lentiviral infection, resulting in Mgy−1. The lentiviral vector used and preparative procedures of Mgy−1 are schematically shown in Fig. 2a and Scheme 1a. Briefly, the extracellular segment of gy-1 acting as the PSMA-targeting moiety and cytoplasmic domain of the enhanced green fluorescent protein (eGFP) as a reporter were anchored onto the macrophage membrane via a rational intracellular protein synthesis process with the help of the CD8 hinge domain and CD8 TM region. High level (near 100%) expression of the gy-1-eGFP fusion protein can be easily detected by confocal laser scanning microscopy (CLSM), flow cytometry, and western blotting measurements, confirming that gy-1 was efficiently expressed on the macrophage membrane (Fig. 2b and Supplementary Fig. 6). Moreover, as depicted by the enzyme-linked immunosorbent assay (ELISA) results in Fig. 2c, the introduction of gy-1 significantly enhanced the binding affinity of Raw264.7 to PSMA, further demonstrating the successful fabrication of Mgy−1. This is a prerequisite for endowing macrophage membrane-based nanomedicines with improved PCa targetability.
Subsequently, multifunctional FeAuNPs were synthesized according to previous report43 and then wrapped with Mgy−1 membranes to yield Magic. The transmission electron microscopy (TEM) images shown in Fig. 2d (upper panel) clearly reveal that the FeAuNPs were highly uniform spheres with an average diameter of 101.30 ± 7.29 nm. The core-shell structure of the FeAuNPs was identified by elemental mapping analysis (Supplementary Fig. 7). In agreement with the changes in the diameter and zeta potential (Supplementary Fig. 8a,b), the scanning electron microscopy (SEM) images in Fig. 2d (bottom panel) clearly show that the FeAuNPs core was homogeneously coated with the Mgy−1 membrane, confirming the formation of Magic. Western blotting results indicate that the composition of the membrane proteins and their biological activities were well retained during Magic synthesis (Fig. 2e). Moreover, the hydrodynamic size of Magic remained nearly unchanged after lyophilization, indicating its excellent stability (Supplementary Fig. 8c).
Cellular targeting and internalization of Magic
Having successfully fabricated Magic, we next investigated whether Magic with TM-expressed gy-1 could inherit the prominent PSMA-targeting ability and tumor cell-specific internalization of gy-1 (Fig. 2f). Magic or Mgy−1-derived vesicles (MVsgy−1, Fig. 2d) were incubated with PC3PSMA+, PC3PSMA−, or LNCaP (PSMA+) cells for 30 min. The binding affinity to PSMA was analyzed using flow cytometry. The results show that both Magic and MVsgy−1 were able to specifically bind to PSMA-positive PCa cells (PC3PSMA+ and LNCaP) rather than PSMA-negative PC3PSMA− (Fig. 2g and Supplementary Fig. 9–11), probably because of the higher binding efficiency of Magic and MVsgy−1 to PSMA in tumor cells. Furthermore, Magic rapidly bound to PC3PSMA+ cells within 15 min and was internalized into the cells within 30 min (Supplementary Fig. 12). However, internalization was almost completely inhibited by pretreatment of PC3PSMA+ cells with a PSMA antibody (PSMAb) with the same binding site as gy-1, demonstrating the effect of TM-expressed gy-1 on enhanced tumor targeting and cellular uptake of Magic (Supplementary Fig. 13). For better comparison, the conventional macrophage membrane-coated FeAuNPs (M-NPs) were labeled with the lipophilic dye 3,3′-dioctadecyloxacarbocyanine perchlorate (DiO) to yield the MDiO-NPs. In contrast to the superior PSMA-targeting capability of Magic, MDiO-NPs failed to distinguish PC3PSMA+ cells from PC3PSMA− cells, suggesting that TM-expressed gy-1 contributed to enhanced PSMA recognition of Magic (Supplementary Fig. 14).
In vitro performance evaluation of Magic
After confirming the prominent targeting performance of Magic, we conducted multiple experiments to explore its other functions. To verify whether the Mgy−1 membrane coating could endow Magic with the capacity to avoid immune clearance, Rhodamine (Rho)-PEG-SH-modified Magic (Rho-Magic) and Rho-PEG-SH-modified FeAuNPs (Rho-NPs) were fabricated to compare their macrophage-mediated phagocytosis. The CLSM images in Fig. 2h,i shows that Rho-Magic escaped phagocytosis by macrophages more effectively than PEGylated Rho-NPs. Flow cytometry measurement further confirmed that 97.53% of Rho-NPs were phagocytized by Raw264.7 versus 11.87% for Rho-Magic (Supplementary Fig. 15), firmly demonstrating that the Mgy−1 membrane coating did confer Magic enhanced immune evasion capability, which would be helpful to prolong the retention time of Magic in blood circulation and further benefit its tumor targeting.
Furthermore, the immunomodulatory potential of Magic was investigated. To verify whether Magic could re-educate M2 macrophages by neutralizing tumor-promoting cytokines secreted by M2 macrophages, ELISA was used first to measure the concentrations of representative cytokines including TGF-β, interleukin (IL)-4, and IL-10 after pretreatment with Magic for 2 h. As shown in Fig. 2j, Magic pretreatment substantially decreased the amounts of TGF-β, IL-4, and IL-10, indicative of the cytokine-neutralization capability of Magic endowed by the Mgy−1 membrane coating. As a result, cytokine-induced M2 macrophage polarization was greatly inhibited, as illustrated in Fig. 2k,l. Collectively, our results suggest that Magic has great potential to effectively ameliorate the immunosuppressive tumor microenvironment, which is crucial for improving anti-tumor efficacy.
Encouraged by the modulatory effect of Magic on the immune microenvironment, we next investigated the possibility of using it as a drug carrier for chemotherapy. The Au-S bond-mediated drug loading capacity and release property of the FeAuNPs were first examined using fluorescein as a drug model. To this end, the FeAuNPs were modified with fluorescein-polyethylene glycol 2000-thiol (FITC-PEG-SH). As shown in Supplementary Fig. 16, FeAuNPs exhibited high drug loading capacity and glutathione (GSH)-responsive drug release within tumor cells, suggesting that FeAuNPs can act as promising carriers of chemotherapeutic drugs to maximize therapeutic effectiveness while minimizing adverse effects. Next, we evaluated the Au-mediated photothermal conversion capability of Magic under continuous laser irradiation (Supplementary Fig. 17). Magic displays the concentration- and power-dependent photothermal behavior and excellent photothermal stability, making it favorable for tumor PTT applications. Taken together, Magic could act as a potential therapeutic agent with multiple favorable features for tumor therapy. In addition, Magic exhibited good MRI and CT capabilities in vitro (Supplementary Fig. 18), which provided the opportunity to evaluate the targeting performance of Magic through multimodality imaging.
In vivo biodistribution of Magic after systematic injection
The above in vitro results encouraged us to assess the biodistribution of Magic in xenograft mice. Indocyanine green (ICG)-PEG-SH functionalized Magic (ICG-Magic) was first intravenously injected into normal BALB/c nude mice to evaluate the pharmacokinetics of Magic. ICG-PEG-SH-functionalized FeAuNPs (ICG-NPs) were used as a control. Subsequently, 20 µL of blood was collected from the mice at different time points, and the fluorescence intensity of ICG in the plasma was measured. As depicted in Supplementary Fig. 19a, circulatory half-life of ICG-Magic was significantly extended to 10 h, in sharp contrast to 3.3 h for the ICG-NPs, indicating that macrophage membrane coating did prevent Magic from being cleared from the blood. Furthermore, in vivo fluorescence images were acquired after the intravenous injection of ICG-Magic or ICG-NPs in normal mice. Distinct from the high accumulation of ICG-NPs in the liver and spleen owing to clearance by MPS, ICG-Magic effectively avoided MPS recognition and elimination (Supplementary Fig. 19b), which was confirmed by hematoxylin and eosin (H&E) staining (Supplementary Fig. 19c).
We further evaluated the targeting ability of Magic to PCa tumors in vivo using M-NPs functionalized with ICG (termed M-ICG-NPs) as a control. ICG-Magic and M-ICG-NPs were injected intravenously into PC3PSMA+ subcutaneous tumor-bearing mice (n = 3 for each group). As shown in Fig. 3a, the tumors could be clearly delineated with strong fluorescence even at 96 h post-injection of Magic, indicating its appreciable tumor specificity and blood circulation. In contrast, the signal was barely visible at the tumor site for the M-ICG-NPs. The excellent tumor recognition capability of Magic could also be confirmed by ex vivo imaging of the tumors and major organs collected from mice that were sacrificed 96 h post-injection (Fig. 3b and Supplementary Fig. 20). Quantitative analysis clearly revealed a 12-fold accumulation of Magic at the tumor site compared to that of M-ICG-NPs (Fig. 3c), which is consistent with the in vivo results. Collectively, Mgy-1 membrane ingeniously combines the advantages of the macrophage membrane and gy-1, endowing Magic with enhanced immune evasion and tumor targeting. In addition to the fluorescence imaging results, Fe3O4-based MRI and Au-based CT imaging consistently demonstrated that Magic is capable of imaging prostate tumors with remarkable specificity and selectivity both in vivo and ex vivo (Supplementary Fig. 21) owing to the presence of TM-expressed gy-1.
Active targeting ability of Magic to PCa metastasis
Having demonstrated the high targeting capability of Magic to primary prostate tumors, we further explored its feasibility for actively targeting PCa bone metastases, which are the leading cause of morbidity and mortality in patients with metastatic PCa (Fig. 3d and Supplementary Fig. 22a).46 ICG-Magic and M-ICG-NPs were intravenously injected into PC3PSMA+ metastatic tumor-bearing mice (n = 3 for each group). As shown in Fig. 3e, for the M-ICG-NPs group, the fluorescence intensity in the tumor decreased rapidly 1 h post-injection, suggesting low tumor accumulation of M-ICG-NPs. In contrast, the fluorescence at the tumor site after injection with ICG-Magic gradually intensified at 1 h and 12 h, indicative of efficient enrichment of ICG-Magic in tumor tissue, which could be attributed to higher PSMA-mediated tumor targeting of ICG-Magic (Supplementary Fig. 22b). Surprisingly, a micro-metastasis with a diameter of 3 mm in the groin next to the bone metastasis could also be effectively identified by Magic instead of M-ICG-NPs after excluding the interference from urine signal. Such striking metastasis targeting was confirmed by ex vivo imaging of the tibia with metastatic tumors and micro-metastatic lesions resected from mice sacrificed 12 h post-injection (Fig. 3f,g). Compared to the fluorescence signals of M-ICG-NPs, Magic could lead to a more than 7- and 9-fold increase in signals at the tibia metastatic and micro-metastatic foci, respectively (Supplementary Fig. 22c,d).
It is worth noting that the micro-metastases were further confirmed to be metastatic lymph nodes by H&E and PSMA-IHC staining (Fig. 3h). TM-eGFP-labeled Magic was specifically internalized into PC3PSMA+ cells rather than germinal centers of metastatic lymph nodes (Fig. 3i and Supplementary Fig. 22e), demonstrating that Magic with superior PSMA-binding affinity specifically promoted its internalization into PCaPSMA+ cells in vivo, which is a prerequisite for targeted tumor therapy.
In vitro evaluation of the anti-tumor effects of Magic-based nanomedicine
DM1 is a potent inhibitor of microtubule assembly with an anti-mitotic effect at sub-nanomole concentrations (median effective dose, ED50 = 10–5–10–4 µg mL–1), making it promising for tumor chemotherapy.47 Although DM1 has been widely explored in cancer treatment, the risk of systemic toxicity remains a significant concern.40,41 Thus, the possibility of using Magic as a drug carrier for the targeted delivery of DM1 was investigated. Thiol-containing DM1 can be incorporated into Magic through Au-S bonds (DM1-Magic), and the cytotoxicity of DM1-Magic towards PC3PSMA+ and PC3PSMA− cells was evaluated using the Cell Counting Kit-8 (CCK-8) assay. As shown in Fig. 4a and Supplementary Fig. 23a, DM1-Magic showed highly targeted cytotoxicity against PC3PSMA+ cells with a half reduction in cell viability (IC50) of 1.131 µg mL− 1 and significantly reduced non-specific cytotoxicity of free DM1, indicating that Magic could act as a potent carrier to enable PSMA-targeted DM1 delivery. As a result, tubulin was severely damaged and mitosis of PC3PSMA+ cells was inhibited, as depicted in the CLSM results (Fig. 4b-d). Additionally, Magic displayed notable photothermal ablation efficacy on PC3PSMA+ cells (Supplementary Fig. 23b). Collectively, Magic could improve the targeting capability of DM1 to ensure high anti-tumor activity while minimizing side effects. Meanwhile, the immunomodulatory effects and PTT capability of Magic synergistically sensitize chemotherapy, thus further enhancing the killing of cancer cells.
Anti-tumor effect of Magic-based nanomedicine in vivo
Having demonstrated the potential of Magic for targeted DM1 delivery and synergistic anti-PCa benefits in vitro, we further verified the therapeutic efficacy of Magic in luciferase-expressing PC3PSMA+ xenograft mice. Tumor-bearing BALB/c nude mice were randomly divided into five groups (n = 5 per group): (1) intravenous injection with phosphate-buffered saline (PBS) as a control, (2) intravenous injection with Magic without laser irradiation, (3) intravenous injection with Magic followed by 808 nm laser irradiation, (4) intravenous injection with DM1-Magic without irradiation, and (5) intravenous injection with DM1-Magic followed by 808 nm laser irradiation. Tumor growth was monitored using a Xenogen IVIS in vivo imaging system for the detection of bioluminescence or by measuring the average tumor size. In contrast to the continuous tumor growth in the PBS group, all other four groups showed significant inhibition of tumor growth (Fig. 4e-g, and Supplementary Fig. 24). It is noteworthy that the injection of Magic alone substantially suppressed tumor progression. Compared to other treated groups, the integration of DM1 with PTT exerted stronger anti-tumor efficacy during the entire treatment period, strongly demonstrating the competence of the Magic-based platform for highly specific treatment of PCa through its prominent PSMA-mediated tumor targeting. Moreover, no evident changes in mice body weight were observed, revealing the good biosafety of Magic-based formulations.
The excellent anti-PCa efficacy of Magic-based nanomedicine is further supported by Ki67 immunohistochemistry and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end (TUNEL) staining. It can be seen clearly that all treatment modalities, even with Magic only, strongly suppressed tumor cell proliferation and efficiently induced cell apoptosis (Fig. 4h,i and Supplementary Fig. 25). The synergistic treatment using DM1 and PTT showed dramatically improved anti-tumor efficacy, consistent with the in vivo results described above.
Anti-tumor immunomodulation of Magic in vivo
To gain deeper insight into the inhibitory mechanism of pure Magic in vivo, we further investigated whether it could exert anti-tumor effects by blocking M2 macrophage-related tumor-promoting cascades, considering that Magic neutralized tumor-promoting cytokines secreted by M2 macrophages in vitro (Fig. 5a). To verify our hypothesis, ELISA was first performed to detect representative cytokines secreted by M1 and M2 macrophages in tumor tissues collected from mice after treatment with Magic. As shown in Fig. 5b, untreated tumors exhibited greater amounts of the pro-tumoral cytokines IL-4, IL-10, and TGF-β, which are mainly produced by M2 macrophages, than tumors treated with Magic. Meanwhile, those of M1-related anti-tumor cytokines IL-1β , IL-6, and tumor necrosis factor-alpha (TNF-α) were elevated in Magic-treated tumors. These results indicate that Magic effectively reprogramed macrophages from the M2 phenotype to the M1 phenotype by neutralizing pro-tumoral cytokines to improve anti-tumor immunomodulation. Further immunofluorescence staining of macrophages in tumor tissues revealed that Magic-treated tumors displayed a clear reversal in the ratio of M1/M2, which is in agreement with the ELISA results described above (Fig. 5c,d).
As mentioned previously, M2 macrophages stimulate endothelial cell proliferation-based tumor angiogenesis and EMT-mediated tumor metastasis by secreting vascular endothelial growth factor C (VEGFC) and TGF-β. Inspired by the excellent capability of Magic to neutralize TGF-β and downregulate VEGFC (Supplementary Fig. 26a), we further investigated the potential of Magic to inhibit the EMT process and tumor angiogenesis. The expression levels of EMT-associated proteins (vimentin and ECAD) and markers of tumor angiogenesis (VEGF receptor 2, VEGFR2, and CD105) were evaluated by immunofluorescence staining. Compared to the PBS control, Magic significantly reversed EMT and inhibited tumor angiogenesis, indicating the strong potential of Magic to effectively prevent tumor progression (Fig. 5e,f and Supplementary Fig. 26b-d). Collectively, Magic not only specifically delivered therapeutic drugs to prostate tumors for improved anti-cancer treatment, but also exerted immunomodulatory effects to promote the polarization of M2 to M1, thus blocking M2 macrophage-mediated tumor-promoting cascades and tumor progression.
Anti-tumor effects of Magic-based nanomedicine in PCa bone metastasis mode
Active targeting-mediated anti-tumor effects were observed in PCa bone metastasis models (Fig. 6a,b and Supplementary Fig. 27). Consistently, synergistic treatment with DM1 chemotherapy, PTT, and immunomodulation of the tumor microenvironment resulted in optimal therapeutic outcomes, and the metastatic tumors were completely eliminated after 21-d treatment. Furthermore, tumor-induced tibial destruction was significantly improved after effective treatment of metastases, as illustrated by the Micro-CT results in Fig. 6c and Supplementary Fig. 28. The three synergistic effects not only recovered the mineral content of the tibia and the microstructure of bone trabeculae, but also enhanced the mechanical properties of the tumor-metastatic tibia by more than 50% (Fig. 6d and Supplementary Fig. 29). Finite element analysis further verified that Magic-mediated tumor treatment was able to reduce the risk of fracture compared to the PBS group (Fig. 6e-g). Therefore, Magic benefited from its impressive PSMA targetability and exhibited great potential for the successful management of primary prostate tumors, micro-metastases, and concomitant damages.
In addition, neurotoxicity, peripheral organ toxicity, and embryotoxicity of Magic have been comprehensively evaluated. As shown in Supplementary Fig. 30–32, no obvious physiological abnormalities or systemic toxicity were observed in any of the treated mice, demonstrating that Magic can be safely applied as an excellent platform for tumor therapy.