Design of novel afterglow luminescent molecules
For typical organic afterglow process, the vinyl bonds (C=C) of afterglow substrate (MEHPPV, DO, SO and HBA, Supplementary Table 3, Supplementary Scheme 1) are oxidized by singlet oxygen (1O2) under light excitation to form the unstable intermediates (1,2-dioxetane) via cycloaddition reaction, followed by radiative relaxation to emit luminescence13, 24. However, the generation of 1O2 required the triplet energy of sensitizer to be greater than the energy barrier between 3O2 and 1O2 (22.5 kcal/mol), which may be a major obstacle that greatly limits the afterglow performance of those probes30. Given the generation of intermediates through electron transfer path is not limited by the triplet energy barrier between 3O2 and 1O230, herein we exploit the electron transfer mechanism to boost the production efficacy of intermediates and thereby afterglow intensity. We introduce the electron-rich anthracene into the afterglow molecule design to synthesize trianthracene derivatives (TA), which was substituted with alkoxy groups at ortho- and bay-positions to enhance the electro-rich properties. Its isomer dianthraphenanthrene (DAP) was also synthesized as a comparison (Fig. 1a; 2. Supplementary Molecule design, Synthesis and Characterization). Such afterglow luminescence mechanism of TA-NPs involves two complementary types of photooxidation process (i.e., Type I and Type II reaction) (Fig. 1a). Specifically, Type I reaction yields superoxide radicals (O2•−) and radical cations of TA (TA•+) through electron transfer, while Type II reaction produces 1O2 and TA molecules via energy transfer. We hypothesize that Type I pathway will greatly promote the reactivity of TA molecules with O2, resulting in the formation of sufficient endoperoxides (EPOs), because the generation of O2•− is not hampered by the triplet energy of sensitizer in Type II pathway. Subsequently, those EPOs underwent O-O bond cleavage into aromatic-carbonyl compounds, and spontaneously generated photons.
In order to construct biocompatible afterglow luminescent nanoparticles, we transformed TA or DAP molecules into water-soluble nanoparticles through one-step nanoprecipitation in the presence of dual polymers (poly (styrene-co-maleic anhydride) (PSMA) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine- N-[methoxy(poly(ethylene glycol))] (DSPE-PEG)) (Fig. 1a, Supplementary Table 4). In particular, transmission electron microscope (TEM) image further revealed that TA-based nanoparticles (TA-NPs) appeared as mono-disperse nanoparticles, with a mean hydrodynamic size of ~ 40 nm (Fig. 1b). The hydrodynamic diameters of the nanoparticles measured by dynamic light scattering (DLS) were about 60 nm (Fig. 1b, Supplementary Fig. 1). Figure 1c and 1d unveiled the characteristic absorption bands and the fluorescence emission spectra of TA-NPs and DAP-based nanoparticles (DAP-NPs). Subsequently, the afterglow luminescence of TA-NPs and DAP-NPs was recorded with the IVIS Lumina XR imaging system under bioluminescence (without excitation) modes to identify the capability of afterglow luminescence. Notably, TA-NPs showed stronger luminescence than DAP-NPs after cessation of white light irradiation (Fig. 1e, f). Besides, the afterglow spectra of TA-NPs were similar to its fluorescence spectra from 600-750 nm (Fig. 1g).
Mechanism of afterglow luminescence
To study the underlying mechanism governing afterglow luminescence through Type I pathway, we conducted the following research. Firstly, to determine the O2•- generation of TA, electron spin resonance (ESR) spectroscopy was initially carried out, using 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) as spin-trap agent for O2•-. As shown in Figure 2a, neither TA in dark nor light irradiation alone had any impact on the ESR signals. In contrast, a characteristic paramagnetic adduct was observed for TA under light irradiation and matched with that of referential KO2 (a well-known O2•-generator), indicating the production of O2•-. Besides, upon the addition of superoxide dismutase (SOD, O2•- scavenger), the ESR amplitude was dramatically reduced to background level, which was ascribed to that SOD can consume O2•-. Next, we characterized the formation of radical cations (TA•+) through UV-vis-NIR absorption spectra. TA•+ was formed after TA receiving irradiation, and the absorption spectra of TA and TA•+ in DCM were recorded (Fig. 2b). Compared with the neutral TA, TA•+ exhibited several typical absorption peaks at the near-infrared region (750 to 1350 nm) and greatly decreased absorption at λmax = 594 nm. Meanwhile, DAP could also produce the radical cations DAP•+ and O2•- under irradiation (Supplementary Fig. 2). Interestingly, TA showed lower first oxidation potential (-0.15 V) compared with the isomer DAP (-0.12 V), according to the cyclic voltammogram measurement, indicating that TA was easier to give electrons to form free radical cation (Fig. 2c, d). Next, SOD was added into TA-NPs solutions, to further validate that the enhanced afterglow signal was indeed related to the generated O2•-, as expected, the afterglow luminescence showed 26.8 % of decrease (Fig. 2e). Furthermore, we have measured 1O2 generation from TA-NPs via the fluorescence intensities of SOSG at 535 nm and found that a considerable amount of 1O2 was produced during irradiation of TA-NPs (Fig. 2f). Subsequently, the incubation of 1O2 (Na2MoO4 + H2O2) with TA-NPs resulted in higher luminescence intensity of TA-NPs, than that of PBS, H2O2 or Na2MoO4 alone (Fig. 2g), which confirmed that 1O2 was also responsible for afterglow luminescence via Type II pathway. Figure 2h showed that the afterglow of TA-NPs in O2-saturated solutions was 1.9 - fold that in N2-saturated solutions, highlighting the key role of O2 in afterglow luminescence.
To further understand the mechanism on EPOs processes, density functional theory (DFT) calculations were applied to predict the relative energies of EPOs intermediates briefly, and gave one of possible reaction paths (Fig. 2i, j). The reaction process of O2•- or 1O2 cycloaddition in anthracene units to anthracene-dioxetane intermediate was determined, based on the corresponding intermediate EPOs found, confirmed by MALDI-TOF MS measurement (Fig. 2i and Supplementary Fig. 3). When exposed to light and air, TA molecules undergo a serial of cycloaddition with O2•- or 1O2 through electron transfer (Type I pathway) or energy transfer (Type II pathway) reactions to give various EPOs intermediates (II-VI). Specifically, initial compound TA was first additively oxidized by one O2•- or 1O2 molecule, forming the corresponding intermediate EPOs (II or III). Since EPOs (II or III) contained surplus reaction sites in other anthracene units, additional O2•- or 1O2 molecule was added to generate the intermediate EPOs (IV, V or VI). Given that the energy of EPOs (II and IV) was slightly lower than that of EPOs (III and V) (Fig. 2j), it might be prone to generate EPOs (II and IV) during gradual cycloaddition reaction. After long-term light irradiation of TA, the typical UV-vis absorption bonds (500 - 600 nm) and typical fluorescent emission peak at 650 nm were significantly decreased, indicating the decomposition of chemical structures for TA molecules (Supplementary Fig. 4). Besides, the obvious carbonyl characteristic peak of oxidized TA fragments was detected in Fourier transform infrared spectroscopy (FTIR) at 1,735 cm-1, which confirmed the oxidation of TA into various aromatic-carbonyl compounds (Fig. 2k).
Based on the above results, we created a possible picture of underlying mechanism for TA-NPs afterglow luminescence. In the photooxidation, TA-NPs underwent two different photochemical reaction pathways to generate EPOs and thereby afterglow luminescence: electron transfer pathway (Type I) and energy transfer pathway (Type II)30, 31. During the Type I afterglow, TA molecules are firstly excited by light and subsequently transfer an electron to 3O2 to produce O2•- and TA•+. During the Type II afterglow, the triplet state of TA molecules excited by light can transfer energy to 3O2 to generate 1O2. Next, TA or TA•+ provide reaction sites to trap O2•- or 1O2, resulting in a large amount of EPOs. Because the generation of O2•- is no longer restricted by the triplet energy of sensitizer, the Type I pathway may greatly increase the reactivity of TA molecules with 3O2 for formation of a large amount of EPOs. Finally, intermediate EPOs can undergo O-O cleavage and thereby generate afterglow luminescence.
Compared with DAP-NPs, TA-NPs possess three complete anthracene units and showed higher afterglow luminescent intensity, indicating the introduction of electron-rich anthracene units was crucial for enhancing afterglow luminescence. Similarly, DAP-NPs could also produce O2•- and 1O2 under photooxidation (Supplementary Figs. 2 and 5). Notably, TA-NPs had a lower oxidation potential in contrast to DAP, and were more likely to give electrons to form TA•+ and O2•-, which may increase the path of Type I afterglow reaction (Fig. 2c). We also calculated the process of addition of 1O2 to DAP using DFT calculations, and found that the energy of EPOs (II-VI) from TA were lower than those corresponding EPOs from DAP, suggesting the larger possibility of forming EPOs for TA (Supplementary Fig. 6).
Optimization and performance study of afterglow luminescence
We subsequently investigated the effect of amphiphilic polymers on TA-NPs afterglow luminescence, and found that the dual polymers (PSMA + DSPE-PEG) co-coating resulted in higher afterglow luminescence, in contrast to other kinds of polymer coating groups (e.g., pluronic-F127 or PSMA only) (Supplementary Fig. 7). Therefore, the polymers (PSMA + DSPE-PEG) co-coated nanoparticles were used for the subsequent in vitro/vivo experiments.
Currently, the semiconducting polymer (MEHPPV) is the most commonly used organic afterglow material, reported in the previous articles13, 14, 19. We further compared the afterglow performance of TA-NPs with MEHPPV-based nanoparticles (MEHPPV-NPs) and other organic molecules-based nanoparticles such as anthracene, perylene, naphthalene, respectively (Fig. 3a, Supplementary Table 5). Then, we characterized those nanoparticles including UV-vis absorption spectra, fluorescence emission spectra and DLS sizes (Supplementary Figs. 8 - 11). We found that TA-NPs showed extremely higher afterglow intensity than that of MEHPPV-, anthracene-, perylene-, and naphthalene-based nanoparticles. Especially, the afterglow intensity of TA-NPs was ~ 500-fold that of MEHPPV-NPs (Fig. 3b, c, Supplementary Fig. 12).
Through the continuous acquisition of afterglow images, the long-lasting afterglow luminescence could persist for over 60 mins for TA-NPs after light cessation, with a prolonged half-life of ~ 16 min at biologically relevant conditions (pH = 7.4 at 37 °C; Fig. 3d), compared with half-life of ~ 6 min for MEHPPV-NPs. Moreover, as the power density of light irradiation increasing from 1.4 to 15 mW/cm2, TA-NPs showed gradually brighter afterglow luminescence (Fig. 3f, Supplementary Fig. 13). The afterglow intensities of TA-NPs were continuously enhanced with the irradiation time increasing, and reached the maximum at 8 s of irradiation (15 mW/cm2) (Fig. 3g, Supplementary Fig. 14). Furthermore, afterglow luminescence of TA-NPs became brighter as the concentration increasing, and showed a linear correlation between afterglow intensities and concentrations (0 - 40 μg/mL) (Fig. 3h, Supplementary Fig. 15). As a result, under various power density, irradiation time or concentration, TA-NPs showed extremely higher afterglow intensities than that of MEHPPV-NPs (Fig. 3f, g, h and Supplementary Figs. 13 - 15). Notably, we found TA-NPs could be effectively excited by the white light source with ultra-low power density (room light, 58 μW/cm2) and was able to exhibit bright afterglow (Fig. 3i). This ultra-bright afterglow luminescence of TA-NPs facilitated to shorten the signal acquisition time. Especially, when the acquisition time was reduced to 0.01s, TA-NPs could still produce strong afterglow luminescence. However, no obvious afterglow signals were detected from MEHPPV-NPs under the same conditions (Fig. 3i, Supplementary Fig. 16). Notably, the ultra-low power density (58 μW/cm2) that is far below the intensity of direct sunshine (0.13 W/cm2) can provide safe excitation condition and thereby avoid photo-induced tissue damage.
Moreover, the ultra-low power of excitation light leads to the negligible chemical decomposition of afterglow molecules. Thereby, the afterglow signal could be recharged by more than 15-cycle light irradiation (30 mW/cm2, 3s) or 30-cycle light irradiation (6.6 mW/cm2, 1s), with no obvious decay of maximum afterglow intensity (Fig. 3j, Supplementary Fig. 17). Such negligible afterglow photobleaching may be attributed to the efficient photo-afterglow conversion and ultra-low power density of excitation light, which enables to guarantee the accurate quantification of afterglow signals during hours-long and repeated molecular imaging.
Prior to afterglow luminescence imaging in vivo, we studied the tissue penetration depth of afterglow luminescence emitted from TA-NPs. Although afterglow and fluorescence signals were declined with the increase of tissues thickness (Fig. 3k, Supplementary Fig. 18), the afterglow luminescence exerted significantly higher signal-to-background ratio (SBR) and tissue penetration (up to 6.0 cm), compared with that of fluorescence, owing to the no autofluorescence from tissues during afterglow imaging. Besides, the tissue penetration and SBR of afterglow luminescence for TA-NPs was much higher than that for MEHPPV-NPs (Supplementary Fig. 19).
Two reasons may contribute to the significant improvement of afterglow intensity for TA-NPs, in contrast to MEHPPV-NPs. Firstly, for Type I afterglow, TA-NPs can generate O2•- through the electron transfer and produce a large amount of EPOs via Type I pathway, while there was no obvious generation of O2•- and a lack of Type I path for MEHPPV-NPs (Supplementary Fig. 20a). Secondly, for Type II afterglow, TA-NPs display the stronger ability to generate 1O2, compared to MEHPPV-NPs (Supplementary Fig. 20b and c), which could promote the subsequent generation of EPOs via Type II pathway. Thus, TA-NPs emitted an extremely high afterglow luminescence than MEHPPV-NPs, through both electron transfer path (Type I afterglow) and the enhanced energy transfer path (Type II afterglow) (Fig. 3e).
Afterglow imaging of tumors and plaque-bearing carotid
The features of TA-NPs, such as extremely high afterglow luminescence and prolonged luminescence lifetime, offer it noteworthy advantages in designing afterglow molecular imaging in vivo. Prior to afterglow luminescence imaging using TA-NPs in vivo, the stability and biocompatibility of TA-NPs were examined. The hydrodynamic size and luminescence intensity of TA-NPs remained highly stable after incubating those in different buffer solution (cell culture medium, PBS, water) for various time (Supplementary Figs. 7 and 21). Meanwhile, TA-NPs demonstrated no obvious adverse effect on cellular viability to CT-26 cells (mouse colon cancer cells) (Supplementary Fig. 22).
Inspired by the excellent afterglow performance of TA-NPs, we applied TA-NPs for afterglow luminescence imaging of subcutaneous CT-26 tumor-bearing mice after intratumoral injection, through IVIS Lumina XR imaging system (Fig. 4a). In dark condition, there was no afterglow signal at the tumor site at the beginning. Then, those mice were taken out of dark box and accepted the irradiation of smart mobile phone (1.0 mW/cm2) or electric torch (4.0 mW/cm2). The afterglow luminescence of mice was collected immediately after irradiation, and the tumor areas showed strong afterglow luminescence. Importantly, mice could be even excited by room light with ultra-low power density (58 μW/cm2) (Fig. 4a). Moreover, the afterglow luminescence of tumor could well be recharged by multiple irradiation (more than 3 cycles) with electric torch, mobile phones or room light irradiation (Fig. 4b, Supplementary Fig. 23), as well as showed no obvious attenuation in maximum intensity during 15-cycle light irradiation (30 mW/cm2) (Fig. 4c, Supplementary Fig. 23). The above data demonstrated TA-NPs in mice could be efficiently re-excited via low power density of portable light sources and displayed no notable afterglow photobleaching.
Highly efficient afterglow luminescence of TA-NPs can significantly reduce the required dose for imaging in vivo. Under white light at low-power density of 6.6 mW/cm2, TA-NPs (20×) showed afterglow signal as large as ~ 6×107 p/sec/cm2/sr, which is significantly higher than that of MEHPPV-NPs (20×). When the lower dose (1×) was used for TA-NPs, the signal intensity was still ~ 4 time that of MEHPPV-NPs (20×) (Fig. 4d, Supplementary Fig. 24). Afterward, we applied TA-NPs for afterglow luminescence imaging and fluorescence imaging of subcutaneous CT-26 tumors after intravenous injection. Afterglow and fluorescence images showed TA-NPs could effectively target tumor areas via enhanced permeability and retention (EPR) effect (Supplementary Fig. 25).
Tightly controlled behavior (mice in anesthetization) is more experimentally tractable, but it provides a limited view or information of the mice32. The ability to image a freely moving animal would represent a critical step towards understanding the relationship between the brain function and the established learning paradigms (fear conditioning, novel object recognition or social interactions), or the pathophysiology of conditions33-35. However, the real-time imaging of those unrestrained animals is greatly challenging. Mice movements would cause motion artifacts and distorted images, when the rapid movements of mice body were faster than the frame acquisition on a time scale, which is manageable in anesthetized animals but is exacerbated in awaken state36, 37. Thereby, the visualizing study of freely moving animals requires the imaging system with ultra-high signal intensity of probe and ultra-short acquisition time. Fortunately, the Type I and Type II afterglow of TA-NPs guarantee extremely large luminescence intensity and ultra-short acquisition time (0.01 s), thereby we tried to investigate the applicability of TA-NPs for ultra-fast afterglow imaging of naturally behaving animals (Fig. 4e). Amazingly, TA-NPs allowed the observation of afterglow luminescence in a freely moving mouse for more than 10 min, through subcutaneous injection (Fig. 4f, Supplementary Fig. 26). Especially, we were able to monitor the development of afterglow signal in subcutaneous CT-26 tumors in the freely moving mouse via intravenous injection of TA-NPs (Fig. 4g, Supplementary Fig. 28), while no afterglow signals were detected from MEHPPV-NPs -injected mice under same condition (acquisition time: 0.01 s; irradiation conditions: 30 mW/cm2) (Supplementary Figs. 27 and 29). By providing a moment-by-moment record of behavior, TA-NPs with high luminescence could create a serial of frameworks for relating natural behaviors to the afterglow signals, so as to assess sensitive behavioral changes in the freely moving animals.
Glioblastoma multiforme (GBM) is the most common and aggressive malignant tumor found in the central nervous system, with remarkably poor prognosis showing 5-year survival rate of 4 - 5 % in clinic38. Thereby, the precise imaging of GBM is beneficial to early diagnosis, and of great value in assessment of GBM aggressiveness, treatment response, prognosis and recurrence39-41. Owing to the non-invasion, high sensitivity and bright luminescence, we promoted TA-NPs for imaging orthotopic GBM-bearing mice (Fig. 4h). The orthotopic GBM was prepared via implantation of human glioma cells (U87) into nude mice by stereotactic implantation and confirmed via hematoxylineosin (H&E) staining of brain slice (Fig. 4k). After intravenous injection of TA-NPs, the orthotopic GBM-bearing mice showed the dynamically enhanced afterglow signals in head areas, while healthy mice displayed no obvious afterglow signals of mice head (Fig. 4i, j, Supplementary Fig. 30). Thus, TA-NPs could diffuse into the orthotopic glioma tumor via EPR effect through disrupted blood-brain barrier, and exerted high-sensitivity and accurate imaging of glioma tumor model.
The vascular system dysfunction due to atherosclerosis is one of leading causes for severe health problems around the world42, 43. The plaque buildup in the walls of carotid arteries subsequently leads to luminal stenosis, low blood supply to brain, and even acute stroke incidents with plaque rupture44. Next, TA-NPs were applied to image the plaque in carotid atherosclerosis-bearing mice through intravenous injection of TA-NPs (Fig. 4l). From afterglow and fluorescence images of mice (Fig. 4m, n, Supplementary Figs. 31 - 32), the atherosclerotic plaque-bearing carotid (right side) showed significantly higher signal intensity than the normal carotid (left side). Moreover, ex vivo afterglow and fluorescence images displayed the plaque-bearing carotid with stronger signal than normal carotid (Fig. 4o, Supplementary Fig. 33), which was consistent with the histological analysis of Oil red O staining and H&E staining (Supplementary Figs. 34 - 35). Those results suggested that TA-NPs were able to target atherosclerotic lesions and exert sensitive and non-invasive afterglow imaging of atherosclerotic plaque in living mice.
Afterglow imaging of immune responses after chemotherapy in vivo
Dynamic imaging of immune cell activity (cytotoxic T lymphocytes (T cell) activity) during immunotherapy could enable to directly monitor immune response of tumor toward immunotherapy45. Granzyme B is a serine-protease released by CD8+ T cells during the cellular immune response and represents one of the two dominant mechanisms by which T cells mediate cancer cell death46-50. To visualize granzyme B during immunotherapy, probe should possess the sufficient signal intensity, high imaging penetration depth, high SBR and no obvious afterglow photobleaching, which allows for accurate quantification over the repeated imaging. Encouraged by the excellent afterglow performance of TA-NPs, we further promoted TA-NPs for afterglow imaging cytotoxic T lymphocytes activity within tumors after chemotherapy or radiotherapy. To achieve this, we designed, synthesized and characterized a functional activity-based Granzyme B afterglow nanoprobe (TA-BHQ), where a Granzyme B-cleavable peptide sequence (Ile-Glu-Phe-Asp, IEFD) was conjugated with TA-NPs as luminophores and BHQ-3 as quenchers (Fig. 5a; Supplementary Methods: Preparation of TA-BHQ and Supplementary Fig. 36). These donor-acceptor pairs with excellent spectral overlap resulted in the maximum efficacy of quenching luminescence (“off” state) through afterglow resonance energy transfer (ARET), when spatially constrained (less than 10 nm) (Fig. 5b). In the presence of Granzyme B, the IEFD peptide between TA-NPs and BHQ-3 was cleaved and ARET was reduced, leading to the recovery of afterglow luminescence (“on” state) (Fig. 5a). As a result, the gradual increase in afterglow intensity was observed over concentrations of Granzyme B (Fig. 5c). Signal quantification from afterglow or fluorescent images showed a linear correlation between luminescence intensities and Granzyme B concentrations (Fig. 5d, Supplementary Fig. 37). Besides, the luminescence signals remained nearly unchanged for other enzymes and biomolecules, demonstrating the selective response of TA-NPs probes toward Granzyme B (Supplementary Fig. 38).
The capabilities of TA-BHQ probes for in vivo real-time afterglow luminescence imaging of Granzyme B were validated against subcutaneous CT-26 tumor-bearing mice model. To activate the immune response, three immunotherapeutic agents including NLG919, BMS-1 and BEC were administered into the living mice (Fig. 5e, Supplementary Fig. 39). These immunotherapeutic agents mainly acted in tumor microenvironment to facilitate the infiltration of CD8+ T cells into tumor. Using BEC-treated mice as an example, the afterglow and fluorescence signals of tumors were increased gradually after injection of TA-BHQ (Supplementary Fig. 40). At 6 h post-injection of TA-BHQ, strong afterglow signals were observed for the tumor of immunotherapeutic-treated mice, which was higher than that of PBS group. Moreover, multiple-time injection of BEC showed stronger afterglow signals than that of one-time injection, indicating the dose-dependent immune response (Fig. 5f, g). Although the fluorescence imaging of Granzyme B via TA-BHQ were consistent with that of those afterglow imaging, the SBR of afterglow was higher than that of fluorescence (Supplementary Figs. 41 - 42).
Next, immunofluorescence staining and flow cytometry analysis were performed for measuring CD8+ T cells infiltration and Granzyme B level within tumors after various treatments (Fig. 5h, Supplementary Figs. 43 - 45). Those tumor tissues excised from BEC-treated mice displayed higher infiltration of CD8+ T cells and higher expression of Granzyme B, as compared to other groups, which revealed a higher level of immunoactivation after BEC-treatment. As summarized in heat map (Fig. 5i), the ex vivo quantitative analyses of immunofluorescence staining were coincided with the signals of afterglow, which validated that the activated signals of TA-BHQ were well correlated with the immunoactivation.
Afterglow imaging of radiotherapy or/and immune checkpoint blockade therapy
Radiotherapy (RT), referring to the external-beam X-ray irradiation, has gained acceptance for treating over 50 % of cancer patients in clinic51, 52. Ionizing radiation can trigger immune system response through multiple ways, such as augmenting the expression of MHC-I in the affected cells, attenuating PD-1/ CTLA-4 in T lymphocytes, or inducing immunogenic cell death53, 54. Thereby, there is a growing consensus that the combination of immune checkpoint inhibitors (such as anti-PD-L1) with RT can increase immune response rates55, 56. Unfortunately, studies have also provided the intriguing outcomes of immune response to different doses and fractionation schedules of RT54.
Considering the lack of validated tools for predicting the immune response to RT or/and immune checkpoint blockade therapy, we favored to promote TA-BHQ for evaluating the immune response in mice receiving RT combined with anti-PD-L1, by monitoring Granzyme B level. The subcutaneous CT-26 tumor-bearing mice received RT or/and anti-PD-L1 treatment, followed by injection of TA-BHQ and afterglow luminescence imaging (Fig. 6a). Notably, mice treated with anti-PD-L1 + RT showed dynamic enhancement of afterglow signals from tumor sites post injection TA-BHQ (Fig. 6b, c, Supplementary Fig. 46). Specifically, compared with mice treated with PBS, those tumors treated with anti-PD-L1, RT or anti-PD-L1 + RT exhibited higher afterglow signals (Fig. 6d, e), which indicated TA-BHQ could monitor Granzyme B level during RT or/and immune checkpoint blockade therapy. Besides, those fluorescence imaging results were well matched with that of afterglow imaging (Supplementary Fig. 47).
Next, the expression level of Granzyme B and population of T cell in tumors were measured by immunofluorescence staining and flow cytometry analysis (Fig. 6f, g, h, Supplementary Fig. 48). Those results demonstrated that the combination treatment (anti-PD-L1 + RT) led to more infiltration of CD8+ T cells and higher level of Granzyme B in subcutaneous CT-26 tumor, compared with single treatment (RT or anti-PD-L1) or PBS treatment, which validated that the activation of TA-BHQ probe at tumor site was owing to the increased level of Granzyme B released from activated CD8+ T cells infiltrated in the tumors. Moreover, the heat map (Fig. 6i) displayed the afterglow signals were well correlated with both CD8+ T cells infiltration and Granzyme B levels within tumors after various treatments. Therefore, TA-BHQ was able to assess the real-time immunoactivation in living mice during chemotherapy, RT or/and immune checkpoint blockade therapy.