Preparation and characterization of Da-CD@CET@CQ NPs
Poly-β-CD was synthesized as previously described in the literature[39, 40]. The ER-targeted PS CET was obtained by alkylation as well as amidation reactions (Fig. S1). The properties of the Da-CD@CET@CQ NPs were characterized. Transmission electron microscopy (TEM) images revealed the specific morphology of the Da-CD and Da-CD@CET@CQ NPs, both of which exhibited uniform spherical structures with diameters of approximately 60 nm (Fig. 1A and B). The results of dynamic light scattering experiments further confirmed that the average hydrodynamic diameters of the Da-CD NPs and Da-CD@CET@CQ NPs were about 58 nm and 62 nm, and the polydispersity indices (PDIs) were approximately 0.23 and 0.22, respectively, as shown in Fig. 1C and D. The stability of materials has always been a prerequisite and key to their applicability and effectiveness, so we evaluated the stability of several nanomaterials. As shown in Fig. 1E, the solutions of Da-CD NPs, CD@CET@CQ NPs, Da-CD@Ce6@CQ NPs and Da-CD@CET@CQ NPs did not show obvious turbidity or precipitation after 7 days in deionized water, similar to the initial state ( Fig. S2), and the PDIs of several nanomaterials were very small and remained basically unchanged within 7 days (Fig. 1F). Moreover, the average hydrodynamic diameters of the Da-CD@CET@CQ NPs were stable and unchanged after 7 days of treatment in deionized water or RPMI 1640 medium (Fig. 1G and H), which suggests that these nanocomplex based on poly-β-CD are stable and homogeneous in the long term. The UV-vis absorption spectra of the Da-CD@CET@CQ NPs at 330 nm showed characteristic absorbance peaks for Da, as well as for CET and CQ at 670 nm and 260 nm (Fig. 1I), indicating that Da had been modified on the Da-CD@CET@CQ NPs surface by stronger host-guest interactions, and the successful loading of CET and CQ. In addition, the emission spectra of the Da-CD@CET@CQ NPs had nearly the same sharp fluorescence band centered at 680 nm as that of CET, further confirming the effective loading of CET (Fig. S3). The zeta potential results showed that the Da-CD NPs were positively charged, decreasing from 15.6 mV to 8.7 mV, further indicating the successful loading of CQ in the Da-CD@CET@CQ NPs (Fig. 1J). The drug loading capacities (DLCs) of CET and CQ were 6.32 ± 0.15% and 7.04 ± 0.19%, respectively, and the drug loading efficiencies (DLEs) were 79.10 ± 0.18% and 87.50% ± 0.23%, respectively, as calculated by using UV-vis spectrophotometry and based on the drug standard curve, which demonstrated that the Da-CD NPs had good loading capacities.
Poly-β-CD is in a stable state under physiological conditions, which ensures safety during drug delivery, but its structure is damaged, resulting in sparse nanoparticles, in acidic tumor regions according to previous studies[41]; in addition, under acidic conditions, the payload drug is easily protonated, which in turn reduces its noncovalent interaction with poly-β-CD, leading to a decrease in the degree of poly-β-CD crosslinking and disintegration[42–45]. To verify the release mechanism and effects of the Da-CD@CET@CQ NPs, the UV-vis spectrophotometry was used to determine the release kinetics of the drug under different acidic conditions. The release rates of CET and CQ after 24 h were only 3.2% and 5.2%, respectively (pH = 7.4), which effectively prevented the premature release of the drugs during transportation (Fig. 1K and L). Moreover, the release rate increased significantly when the pH was further reduced to 6.0 and 5.0, especially at pH = 5.0, the release rates of CET and CQ reached 89.1% and 91.5%, respectively. These results indicated that the lower pH of the tumor microenvironment could trigger the release of CET and CQ from the Da-CD@CET@CQ NPs, thereby increasing the biosafety and effectiveness of the drugs.
Intracellular uptake and endoplasmic reticulum localization of Da-CD@CET@CQ NPs
To verify the ability of the Da-CD@CET@CQ NPs to target TNBC cells, we used 4T1 cells with high expression of SRC proteins for in vitro cell uptake experiments. Confocal laser scanning microscopy (CLSM) and flow cytometry (FCM) were used to determine the fluorescence intensity of the Da-CD@CET@CQ NPs in the cells to assess their ability to target 4T1 cells. Based on the time-dependent of drug uptake and the data at different time points (Fig. S4), 12 h was chosen as the appropriate time point for uptake studies. The CLSM and FCM fluorescence data showed that the Da-CD@CET@CQ NPs had stronger intracellular fluorescence after uptake for 12 h, which was approximately 5-fold greater than the cellular fluorescence of the CD@CET@CQ NPs, suggesting that they exhibited more pronounced cellular uptake (Fig. 2A and B, Fig. S5). We hypothesize that this result may be due to the high expression of SRC in 4T1 cells and dasatinib-mediated targeting. To test this hypothesis, we preincubated 4T1 cells with different concentrations of free Da (0, 5, 10, and 20 nM) for 12 h before they were incubated with the Da-CD@CET@CQ NPs so that they could pre-bind to intracellular SRC, thereby reducing the amount of free intracellular SRC. The CLSM (Fig. S6) and FCM results (Fig. 2C and D) showed that with prebound SRC, the intracellular fluorescence intensity of the Da-CD@CET@CQ NPs decreased with increasing concentrations of free Da. The intracellular fluorescence intensity was significantly reduced by approximately 9-fold when the preincubation concentration was 20 nM, indicating that the Da-CD@CET@CQ NPs could indeed achieve greater accumulation in 4T1 cells through Da and SRC-mediated targeting. Subsequently, to further demonstrate the tumor specificity of the Da-CD@CET@CQ NPs for TNBC, we used a human embryonic kidney cell line (HEK293T) with SRC-negative expression as the control. The CLSM (Fig. S7) and FCM (Fig. 2E and F) results showed that the Da-CD@CET@CQ NPs had an approximately 5-fold greater fluorescence intensity in 4T1 cells than in HEK293T cells. Taken together, the cellular uptake results indicated that Da modification significantly increased the ability of Da-CD@CET@CQ NPs to specifically target TNBC cells, providing a potential strategy for targeted PDT of TNBC.
Based on the exciting tumor cell targeting results, we validated the ability of the Da-CD@CET@CQ NPs to further localize PS in ER. The PS modified with p-toluenesulfonamide were heavily enriched in ER and in turn directly and instantaneously damaged the structure of ER and increased ER stress, thereby increasing the efficiency of apoptosis. Therefore, the ER-Tracker Green probe was used to study the ER localization of free Ce6, CET and Da-CD@CET@CQ NPs in 4T1 cells. As shown in Fig. 2G, all the red fluorescence of free Ce6 had almost no overlap with the green fluorescence signal of the ER probe. Further qualitative analysis of the line-scanning spectra of the fluorescence intensities showed that Ce6 and the ER-Tracker Green had substantially separated signals (Fig. 2J-I). Qualitative analysis of the line-scanning spectra of the fluorescence intensities showed a partial overlap of CET and the ER-Tracker Green, which indicated that the free CET exhibited further ER localization after passive cellular uptake (Fig. 2H, J-II). Compared with that of free CET, the red fluorescence signal of the Da-CD@CET@CQ NPs group significantly overlapped with the green fluorescence signal of the ER probe, and the qualitative analysis of the line-scanning spectra of the fluorescence intensities showed a high degree of overlap (Fig. 2I, J-III). Thus, these data provide strong evidence that the Da-CD@CET@CQ NPs can significantly increase the ability of PS to effectively enter ER.
Intracellular ROS imaging and In vitro phototoxicity
Reactive oxygen species (ROS) with superior oxidative ability play a critical role in directly causing apoptosis and necrosis of tumor cells and play a key role in continuously increasing ER stress. The ROS indicator we selected was nonfluorescent 2',7'-dichlorodihydrofluorescein (DCFH-DA), which can be oxidized by ROS to green fluorescent DCF. Herein, we used CLSM to determine the fluorescence intensity of DCF. As shown in Fig. 3A and B, the green fluorescence intensity of the Da-CD NPs and CQ groups without PS involvement was negligible. Compared with those of the free single-targeting CET and CD@Ce6@CQ NPs groups, the green fluorescence intensities of the Da-CD@Ce6@CQ NPs, Da-CD@CET NPs and Da-CD@CET@CQ NPs groups were approximately 5 times greater. These results indicated that the nanocomplex group with surface-modified Da had a higher ROS generation efficiency, which was also consistent with the cell uptake results. In addition, our further study revealed that the intracellular ROS production increased with increasing Da-CD@CET@CQ NPs incubation concentration (Fig. S8). Thus, these results suggest that poly-β-CD surface modification of Da increases the cell-targeted uptake of Da-CD@CET@CQ NPs and further promotes ROS generation.
To determine whether the notable tumor cell targeting and ER localization ability of the Da-CD@CET@CQ NPs also resulted in superior cellular photodynamic activity, we evaluated the ability of the Da-CD@CET@CQ NPs to inhibit cellular activity via the MTT assay. First, as shown in Fig. 3C, both the poly-β-CD-based nanomaterials and PS showed no significant cytotoxicity against 4T1 cells in the absence of light, whereas free CQ and the nanomaterials encapsulating CQ (CD@CET@CQ NPs, Da-CD@Ce6@CQ NPs and Da-CD@CET@CQ NPs) exhibited weak cytotoxicity as the concentration increased but still maintained a cell survival rate above 85%. Moreover, as shown in Fig. 3D, the cellular phototoxicities of the Da-CD@Ce6@CQ NPs, Da-CD@CET NPs and Da-CD@CET@CQ NPs with cellular targeting were significantly greater than those of free CET and the CD@CET@CQ NPs with single-targeting ability after laser irradiation (λ = 670 nm, 4 mW·cm− 2), which might be due to the increased intracellular uptake mediated by Da. More importantly, at limited concentrations up to 25–50 µg/mL, the live cell inhibitory abilities of the Da-CD@CET@CQ NPs (15.2% and 7.3%) were both significantly greater than those of the Da-CD@Ce6@CQ NPs (37.9% and 21.6%), which also suggested that PS with both tumor cell and ER-targeting abilities exhibit increased photodynamic activity and proapoptotic abilities. In addition, we further evaluated its cytotoxicity under light exposure by live and dead assay. As shown in Fig. 3E and F, the Da-CD NPs and CQ did not significantly inhibit cell activity. The red-to-green ratio was significantly higher in the free CET and CD@CET@CQ NPs groups, and the ratio was close to 50%. Notably, the red fluorescence after Da-CD@CET@CQ NPs treatment was significantly stronger than that in the Da-CD@Ce6@CQ NPs and Da-CD@CET NPs groups, which further indicated that the Da-CD@CET@CQ NPs treatment induced more apoptotic cell death. Moreover, as shown in Fig. 3G and H, the cell death increased with increasing Da-CD@CET@CQ NPs incubation concentration, which was also consistent with the MTT results. In addition, apoptotic fluorescence staining was performed by using Annexin V-FITC and PI analysis of different drug groups after light irradiation. As shown in Fig. 3I and J, the proportions of the two regions of early apoptosis (Q2) and late apoptosis (Q3) in the Da-CD@CET@CQ NPs group (95.76%) were higher than those in the Da-CD@Ce6@CQ NP group (69.06%) and Da-CD@CET NPs group (81.36%), and late apoptosis was predominant (76.68%), which indicated that the Da-CD@CET@CQ NPs had a greater ability to induce apoptosis; this result was consistent with the above MTT assay and the live-dead cell staining results. Therefore, the Da-CD@CET@CQ NPs, which showed tumor cell targeting and further localization of PS to ER, could effectively improve the photodynamic antitumor effect.
Autophagy inhibition In Vitro and ICD effect induced by ER stress
Autophagy maintains cell homeostasis through digestion and degradation of damaged, denatured or senescent proteins and organelles, which significantly inhibits the ability of PDT to induce cell apoptosis. Therefore, inhibition of autophagy has become one of the key factor to improve the therapeutic effect of PDT. To evaluate the autophagic inhibitory effect of the Da-CD@CET@CQ NPs loaded with CQ, a series of studies were performed by Western blot assay and confocal microscopy. The presence of LC3 in autophagosomes and its conversion to the smaller molecular weight LC3-II are indicators of autophagic[46]. After 4T1 cells were co-incubated with Da-CD@CET NPs and irradiated by a laser, the LC3-Ⅱ protein in the cells was significantly increased compared with cells without irradiation (Fig. S9), indicating that PDT effect can induce obvious autophagy, which is consistent with previous literature reports[47–49]. As shown in Fig. 4A and B, compared with other groups, the LC3-II protein content in the groups treated with free CQ or the nanocomplex encapsulated with CQ was significantly higher, which indicated that free or loaded CQ molecules did not prevent the protein conversion of LC3-I to LC3-Ⅱ in cells. In addition, P62, a bridge linking LC3 and polyubiquitinated proteins, can be degraded by proteolytic enzymes in autophagic lysosomes during autophagy. In the case of reduced or defective autophagy, the P62 protein accumulates in the cytoplasm, which is used as the indicator of autophagic inhibition[50]. As shown in Fig. 4A and C, the abundant expression of the P62 protein was found in the presence of free CQ and CQ-encapsulated nanocomplex compared to control cells, indicating that autophagy was significantly inhibited. Moreover, LysoTracker Green staining was performed on intracellular acidic vesicles (lysosomes). The results showed that Da-CD@CET@CQ NPs exhibited more significant intracellular accumulation of autophagy vesicles (green) (Fig. 4D). These results further indicate that CQ molecules released by Da-CD@CET@CQ NPs had no affect on the transformation of LC3-I to LC3-Ⅱ, but can significantly inhibit lysosomal degradation of autophagosome, and thus promote the accumulation of more autophagy vesicles in cells. Thus, it can be observed that Da-CD@CET@CQ NPs had a more significant anti-tumor effect of PDT in vitro.
ER is a multifunctional organelle in the cell and the main reservoir of intracellular Ca2+. Excessive production of ROS in ER leads to dysregulation of intracellular Ca2+ homeostasis. To determine whether the Da-CD@CET@CQ NPs disrupt Ca2+ homeostasis after laser irradiation, we used the sensitive Ca2+ probe Fluo-4 AM. Free Fluo-4 AM has very weak fluorescence, however, when it was sheared by intracellular esterases to form Fluo-4, which can bind with Ca2+ to produce strong green fluorescence. CLSM revealed that the cells treated with Da-CD@CET NPs and Da-CD@CET@CQ NPs displayed stronger green fluorescence, with intensities 5-fold and 2-fold higher than those of the ER-targeted CET group and the Da-CD@Ce6@CQ NP group, respectively (Fig. 4E and Fig. S10). Thus, these results demonstrated that the nanomaterials with cellular and ER targeting capabilities have a more significant ability to disrupt intracellular Ca2+ homeostasis.
Dysregulation of intracellular Ca2+ homeostasis impedes protein synthesis and folding, which leads to the accumulation of unfolded or misfolded proteins in ER, inducing the ER stress. On the one hand, at the onset of ER stress, unfolded or misfolded proteins in ER lumen are highly bound to the molecular chaperone binding immunoglobulin (BIP), leading to its dissociation from ER stress sensors and promoting the activation of activating transcription factor 6 (ATF6), inositol-required enzyme 1 alpha (IRE1 alpha), and protein kinase R-like ER kinase (PERK), which further induce an unfolded protein response (UPR) to remove unfolded or misfolded proteins to maintain the homeostatic balance of the ER. On the other hand, excessive ROS continuously increase ER stress and induce apoptosis by upregulating the expression of proapoptotic C/EPB homologous protein (CHOP), which was mediated by the PERK/eIF2/ATF6/CHOP pathway[51–53]. To assess the extent of ER stress, we first used CLSM to analyze the expression of CHOP proteins. The CLSM revealed that the red fluorescent signal of CHOP were stronger in the Da-CD@CET NPs and Da-CD@CET@CQ NPs group, which had an approximately 2-fold greater value than that in the Da-CD@Ce6@CQ NPs group, suggesting increased CHOP expression by targeting PS to ER (Fig. 4F and Fig. S11). In summary, these experimental results fully confirmed that the Da-CD@CET@CQ NPs can produce a large amount of ROS in ER, resulting in redox balance disorders and Ca2+ homeostatic imbalances and activating proapoptotic pathways through continuous ER stress.
Recently, a growing number of studies have demonstrated that the continuous increase in ER stress generated by ROS can induce ICD[54–57]. Tumor cells induce ICD while producing a series of DAMPs, including CRT, which is exposed on the cell surface; HMGB1, which is secreted to the outside world; and ATP molecules, which are released by the cells[58–61]. Therefore, to determine whether ER stress triggered by the Da-CD@CET@CQ NPs can induce more intense ICD effects and promote DAMPs release, we conducted immunofluorescence and western blot analyses. First, we investigated the migration of HMGB1 from the nucleus to the cellular matrix. As shown in Fig. 4G and Fig. S12, compared with those in the other groups, the overlap between HMGB1 (red) and the nucleus (blue) almost disappeared (pink) in the Da-CD@CET NPs and Da-CD@CET@CQ NPs group, which suggested that the tumor cells and ER targeting nanocomplex can increased HMGB1 migration from the nucleus. Second, we assessed the exposure of CRT on the cell surface by CLSM fluorescence intensity. Compared with those of the other groups, the Da-CD@CET NPs and Da-CD@CET@CQ NPs groups with dual-targeting ability displayed almost uniform and stronger green fluorescence, which was 3.2 times and 2.0 times higher than that of the CD@CET@CQ NPs and Da-CD@Ce6@CQ NPs groups, respectively, indicating that the nanocomplex with tumor cells and ER targeting can trigger higher CRT expression after light treatment (Fig. 4H and I). We further evaluated ATP production in the cell culture medium. As shown in Fig. 4J, there were more ATP secretion in the cellular supernatant of the Da-CD@CET NPs and Da-CD@CET@CQ NPs groups, which was almost 3.0 times and 2.0 times greater than that of the CD@CET@CQ NP and Da-CD@Ce6@CQ NP groups, respectively. Finally, western blotting was used to further analyze the expression of ER stress related proteins. As shown in Fig. 4K and Fig. S13, the western blot results showed that the expression of ER stress marker, such as CHOP, PERK, ATF6, and p-IRE-1α, was significantly higher in the Da-CD@CET NPs and Da-CD@CET@CQ NPs group than that of other groups. All these results indicated that ROS continuously increased ER stress in the Da-CD@CET NPs and Da-CD@CET@CQ NPs groups, effectively induced ICD in tumor cells and further promoted antitumor effects.
In vivo tumor targeting
To further confirm the ability of the Da-CD@CET@CQ NPs to target TNBC tumors in vivo, we used small animal live imaging system to assess the distribution of the nanocomplex in mouse xenograft tumor model. CD@CET@CQ NPs and Da-CD@CET@CQ NPs were injected into BALB/c mice with tumors derived from 4T1 cells via the tail vein, and the fluorescence intensity of both nanocomplex at the tumor site was assessed. As shown in Fig. 5A and B, the fluorescence intensity at the tumor site of the mice in the Da-CD@CET@CQ NPs group was significantly increased at 2 h post-injection and was approximately 3.0 times higher than that in the CD@CET@CQ NPs group, which indicated that the Da-CD@CET@CQ NPs could be enriched at the tumor site faster and more abundantly. With prolonged injection time, the enrichment of both the CD@CET@CQ NPs and the Da-CD@CET@CQ NPs at the tumor site peaked at 12 h, and the fluorescence intensity at the tumor site in the Da-CD@CET@CQ NPs group was approximately 2.5 times higher than that in the CD@CET@CQ NPs group. The fluorescence intensity at the tumor site gradually decreased over time, indicating that this type of nanocomplex could be metabolized by the organism in a timely manner. In addition, to further assess the fluorescence intensity at the tumor site and major organs, the mice were sacrificed at 12 h after administration. As shown in Fig. 5C and D, the fluorescence intensity of the Da-CD@CET@CQ NP group at the tumor site was approximately 2.7 times higher than that of the CD@CET@CQ NP group, which was consistent with the in vivo fluorescence imaging data. Moreover, the concentration of Da-CD@CET@CQ NPs or CD@CET@CQ NPs at the tumor site was lower than that in the liver but higher than that in other major organs. Given the good biocompatibility and negligible cytotoxicity of the Da-CD@CET@CQ NPs and CD@CET@CQ NPs, the adverse effects of their accumulation in other organs in mice should be negligible. Meanwhile, fluorescence detection was performed on the tumor sections dissociated 12 h after administration, as shown in Fig. 5E and F, Da-CD@CET@CQ NPs group showed more significant fluorescence intensity at the edge site of tumor. Moreover, fluorescence intensity 3 times higher than that of CD@CET@CQ group was also observed in the tumor core site of Da-CD@CET@CQ NPs group, which further proved that Da-CD@CET@CQ NPs can achieve abundant and more uniform accumulation of photosensitizer CET at the tumor site (Fig. 5G and H). All these results demonstrate that Da-CD@CET@CQ NPs indeed exhibit more significant 4T1 tumor-targeting ability, which can be attributed to Da-mediated tumor specificity.
In vivo antitumor activity
To assess whether drug enrichment results in more significant antitumor effects, we conducted antitumor studies using BALB/c mice with a 4T1 tumor model. First, the mice were randomly divided into eight groups (n = 5 mice per group): control, Da-CD NPs, CQ, CET, CD@CET@CQ NPs, Da-CD@Ce6@CQ NPs, Da-CD@CET NPs and Da-CD@CET@CQ NPs. Based on the results of the drug targeting experiment, a laser was applied 12 h after injection (670 nm, 50 mW·cm− 2, 10 min), and then, the tumor volume and body weight of the mice were monitored every other day (Fig. 6A). As shown in Fig. 6B and C, in the control group, almost no significant inhibition of tumor growth was observed after the injection of Da-CD NPs or CQ. The free CET and single-targeting groups, including the CD@CET@CQ NP and Da-CD@Ce6@CQ NP groups, only showed partial inhibition of tumor growth. Compared with the Da-CD@CET NP group, the Da-CD@CET@CQ NP group exhibited a more significant tumor inhibitory effect, and the tumors almost completely stopped growing, which may be attribute to the autophagic inhibitory ability of the Da-CD@CET@CQ NPs group, resulting in an increase in the final PDT effect. After 15 days of treatment, the mice were dissected, and the tumor tissues were removed for weighing. As shown in Fig. 6D, the average tumor weight further confirmed that the Da-CD@CET@CQ NPs had a more significant antitumor effect. In addition, we further confirmed the antitumor effect of each group by H&E and TUNEL staining, and immunohistochemical analysis of the tumor tissues. As shown by H&E staining, more pronounced necrosis and increased vesicles, as well as nuclear shrinkage, were found in the Da-CD@CET@CQ NPs group. Moreover, a stronger green fluorescence signal was observed in the Da-CD@CET@CQ NPs group in the TUNEL experiment, which indicated that the Da-CD@CET@CQ NPs group had a greater number of apoptosis cells. Subsequently, the tumor stem cell marker CD34 and the cell proliferation marker Ki67 were selected for immunohistochemistry experiments to further assess vascular proliferation and tumor proliferation. The percentage of positive cells in Da-CD@CET@CQ NPs group was significantly lower than that in other groups, which indicated that Da-CD@CET@CQ NPs effectively inhibited vascular regeneration and cell proliferation (Fig. 6E). Finally, we also performed H&E staining of isolated lung tissues. Compared with those in the other groups with more metastatic nodules in the lung tissues, no obvious metastatic nodules in the lung tissues were observed in the Da-CD@CET@CQ NPs group, which indicated that this treatment effectively inhibited tumor metastasis (Fig. 6F and Fig. S14). The relative body weight of each group of mice remained almost unchanged throughout the experimental period (Fig. S15), and H&E staining analysis of isolated organs (heart, liver, spleen, lungs, and kidneys) revealed normal physiological morphology of tissues in each group (Fig. S16), indicating that all nanocomplexes have almost negligible systemic toxicity as well as side effects in vivo. These results strongly demonstrate that precise delivery of PS to ER of tumor cells and combined with autophagy inhibition has significant in vivo antitumor and metastasis inhibition effects.
In vivo antitumor immunity
We further explored the antitumor immune responses of the Da-CD@CET@CQ NPs by immunofluorescence staining. First, in order to evaluate ER stress and DAMPs release, we detected CHOP expression, CRT exposure and HMGB1 release (red fluorescence) in different tumor tissues by IF staining. The results showed that the red fluorescence intensity of CHOP in the Da-CD@CET NPs and Da-CD@CET@CQ NPs groups was significantly higher than that in the other groups due to the targeting of tumor cells and the presence of ER targeting, indicating the enhancement of ER stress (Fig. S17). In addition, the red fluorescence intensity of CRT and HMGB1 in the Da-CD@CET@CQ NPs group was significantly higher than that in the other groups, indicating that ER-PDT combined with autophagy inhibition could induce stronger ICD effects (Fig. S17). Second, we used a nuclear probe and four different antibodies (CD19-labeled B cells, CD8-labeled T cells, CD49B-labeled NK cells, and iNOS-labeled M1 macrophages) to determine the effect of the body's immune response in the different groups, and the expression levels of the different immunoantibodies were determined by fluorescence intensity. As shown in Fig. 7A, the fluorescence signals of different antibodies in the dual-targeting Da-CD@CET NPs and Da-CD@CET@CQ NPs groups exhibited stronger than other groups. Meanwhile, we measured the expression levels of different antibodies based on fluorescence intensity. As shown in Fig. 7B and C, the expression level of CD8 and CD19, compared with other groups, the Da-CD@CET NPs and Da-CD@CET@CQ NPs group show a significant increase. In addition, the expression level of CD49B and iNOS in the Da-CD@CET NPs and Da-CD@CET@CQ NPs group exhibit a obviously higher level compared to other groups (Fig. 7D and E). These results suggest that in situ ER-PDT increased B cell, T-cell and NK cell expression and promoted macrophage polarization to M1 macrophages. More importantly, four antibodies in the Da-CD@CET@CQ NPs group exhibited a higher expression level than the Da-CD@CET NPs group, possibly because autophagic inhibition further promoted the antitumor effect of PDT, which in turn induced a stronger immune response. Moreover, peripheral blood of mice in different treatment groups were collected on the 15th day after laser irradiation for analysis of cytokine levels. The results showed that the mean levels of TNF-α and IFN-γ in Da-CD@CET@CQ NPs treatment group were significantly higher than those in other groups (Fig. 7F and G), while the mean levels of IL-10 were lower than those in other groups (Fig. 7H). Thus, these results suggest that the PS with tumor cells and ER targeting ability, combined with autophagic inhibition can further increase the ability of PDT to activate the body's immune response, which in turn promotes the synergistic effect of tumor treatment.