Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Dec 19;22(1):782.
doi: 10.1186/s12951-024-02999-z.

Zinc-based radioenhancers to activate tumor radioimmunotherapy by PD-L1 and cGAS-STING pathway

Affiliations

Zinc-based radioenhancers to activate tumor radioimmunotherapy by PD-L1 and cGAS-STING pathway

Mengjiao Xu et al. J Nanobiotechnology. .

Abstract

Radiotherapy and immunotherapy have already become the primary form of treatment for non-small-cell lung cancer (NSCLC), but are limited by high radiotherapy dose and low immune response rate. Herein, a multi-pronged strategy using a radio-immuno-enhancer (ZnO-Au@mSiO2) is developed by inducing tumor cells apoptosis and reprograming the immunosuppressive tumor microenvironment (TME). The radio-immuno-enhancer employed Au as a radiosensitizer, transition Zn ions as immune activators, which not only tremendously enhances the anti-proliferative activity of radiotherapy toward cancer cells, but also activates the immune response with multi-targets to let "exhausted" T cells "back to life" by triggering immunogenic cell death (ICD), immune checkpoint blockade (ICB) that target PD-1/PD-L1 and cGAS-STING under X-ray irradiation with a low dosage. The in vivo results demonstrate desirable antitumor and immunogenic effects of radio-immuno-enhancer-mediated immune activation by increasing the ratio of cytotoxic T cells (CTLs) and helper T cells. This work provides a feasible approach for future development of effective transition metal ion-activated radio-immunotherapeutic agents.

PubMed Disclaimer

Conflict of interest statement

Declarations. Ethics approval and consent to participate: All animal experiments were approved by the Animal Management and Ethics Committee of the Xiamen University. Consent for publication: Not applicable. Competing interests: The authors declare no competing interests.

Figures

Scheme 1
Scheme 1
Scheme illustration of the ZnO–Au@mSiO2 NPs for cancer immunotherapy. Mesoporous silica-encapsulated gold (Au@mSiO2) nanoparticles were used as carriers for Zn2+-dependent DNAzyme, which can act to degrade PD-L1, only after being activated by Zn2+. ZnO, as a double activator for DNAzyme and cGAS-STING, was used as a “caretaker” to block off the pore of the SiO2 for stability and safety (ZnO–Au@mSiO2). The nanoparticles exposed to X-ray could enhance oxidative stress and immunogenic cell death (ICD), which would cause tumor cell apoptosis and activate the tumor immune response; the released Zn2+ in acidic microenvironment activate the cGAS-STING signaling pathway for further amplifying the immune response, and activate DNAzyme for regulating PD-1/PD-L1 immunosuppression
Fig. 1
Fig. 1
Preparation and characterization of ZnO–Au@mSiO2. a Synthetic route of ZnO–Au@mSiO2 nanocomposites. b TEM imaging of ZnO–Au@mSiO2 nanoparticles. c STEM mapping analysis of ZnO–Au@mSiO2. d UV–vis absorption spectra of ZnO–Au@mSiO2. e The hydrodynamic diameter of structures. f Zeta potentials of structures. g Release of Zn2+ from ZnO–Au@mSiO2 under different conditions (pH 5.8, pH 6.5 and pH 7.4). h Schematic diagram of Zn2+ release leading to activation of DNAzyme to cleavage target mRNA. i Gel electrophoresis images showing DNAzyme efficiency for substrate cleavage
Fig. 2
Fig. 2
In vitro inhibition of tumor cell growth. a Cell uptake on LLC cells incubated with ZnO–Au@mSiO2 analyzed by Flow cytometry at different time. b Intracellular ·OH generation detected by DCFH-DA probe (scale bar, 20 μm). c Flow cytometry data for DCFH-DA probe treated with different therapeutic groups. d Detection of lipid peroxidation by BODIPY-C11 staining in LLC cells incubated with different groups. e Cell viabilities of LLC cells and LO2 cells measured by MTT assays, after incubating with different concentration of ZnO–Au@mSiO2 with or without X-ray. f Flow cytogram representing apoptosis assay based on Annexin V-FITC and propidium iodide staining of LLC cells after treatment with different therapeutic groups. Group 1: PBS, Group 2: PBS + X-ray, Group 3: Au@mSiO2, Group 4: Au@mSiO2 + X-ray, Group 5: ZnO–Au@mSiO2u, Group 6: ZnO–Au@mSiO2 + X-ray
Fig. 3
Fig. 3
In vitro effects of mitochondrial damage, DNA damage, and proliferation ability. a Schematic diagram of the cGAS-STING signaling pathway induced by Zn2+ and RT in tumor cells. b CLSM observation on the changes in the mitochondrial membrane potential of LLC cells after incubation with different treatment. The blue, red, and green colors indicate cell nucleus, and JC-1 aggregates and monomer, respectively (scale bar, 30 μm). c Flow cytometry analysis of mitochondrial membrane potentials using JC-1 after different treatments. d Active mitochondria were stained by mitochondrial probe Mito-Tracker Green after treatment with different groups and detected by FCM. e TEM images of mitochondrial morphological changes of LLC cells with different treatments (scale bar, local images: 1 μm, enlarged cell sections images: 200 nm). f The immunofluorescence images of γ-H2AX induced by DNA damage (scale bar, 10 μm). g The photographs of colony formation assay of LLC cells treated with PBS and different concentration of ZnO–Au@mSiO2 under various radiation doses (0, 2, 4, 6 and 8 Gy). h Colony formation rate after treatment with ZnO–Au@mSiO2. i Cell cycle analysis of LLC cells treated with PBS and ZnO–Au@mSiO2 with or without X-ray and j analysis of flow cytometry
Fig. 4
Fig. 4
In vitro gene editing, RT-induced ICD and the cGAS-STING signal pathway-related genetic changes. a The role of ZnO–Au@mSiO2 nanosystem after entering the cells. b Western blotting analysis for protein expression of cGAS-STING-associated proteins (IFN β, TBK1 and p-TBK1) in DC 2.4 cells. c qPCR assay measuring the STING mRNA in DC 2.4 cells (*p < 0.05). d Western blotting analysis for protein expression of PD-L1 in LLC cells. e qPCR assay measuring the PD-L1 mRNA in LLC cells, ZnO–Au@mSiO2 (–) indicates that DNAzyme is not loaded, ZnO–Au@mSiO2 ( +) indicates that DNAzyme is loaded (*p < 0.05, **p < 0.01, ****p < 0.0001). f Western blotting analysis for protein expression of ICD-associated proteins (HMGB1 and CRT) in LLC cells. g qPCR assay measuring the CRT mRNA in LLC cells (*p < 0.05 and ns means no significant difference). h qPCR assay measuring the HMGB1 mRN A in LLC cells (**p < 0.01, ****p < 0.0001)
Fig. 5
Fig. 5
In vivo biodistribution and anti-tumor efficacy. a Pharmacokinetics of ZnO–Au@mSiO2. b Fluorescence imaging of mice bearing LLC tumor at 0, 3, 7, 12, 24 h post-injection intravenously of ZnO–Au@mSiO2-Cy5.5 (n = 3). c The corresponding fluorescence signals of subcutaneous tumors at 0, 3, 7, 12, 24 h post-injection intravenously of ZnO–Au@mSiO2-Cy5.5. d Schematic illustration of the treatment process in mice bearing tumor. e The administration of the tumor volume changes in different treatment groups (n = 3) (*p < 0.05, ***p < 0.001). f The administration of mouse body weights in different treatment groups (n = 3). g Representative H&E staining images of tumor tissues collected from group 1, group 2 and group 3 (scale bar, 100 μm). h Immunofluorescence staining of PD-L1 in the tumor slices collected from group 1, group 2 and group 3 (scale bar, 100 μm). i Western blotting analysis for protein expression of PD-L1 and cGAS-STING-associated proteins (IFN β, TBK1 and p-TBK1) in tumor tissues. j The relative protein levels of survivin after different treatments
Fig. 6
Fig. 6
In vivo antitumor immune responses. a Representative flow cytometry plots and b corresponding quantification of DCs maturation in lymph node tissues (n = 3) (*p < 0.05, ****p < 0.0001). c Representative flow cytometry plots and d corresponding quantification of DCs maturation in spleen tissues (n = 3) (***p < 0.001, ****p < 0.0001). e Representative flow cytometry plots and f, g corresponding quantification of CTLs in spleen tissues (n = 3) (**p < 0.01, ***p < 0.001, ****p < 0.0001). h The expression levels of TNF-α and i Granzyme in serum analyzed by ELISA kit (*p < 0.05, **p < 0.01, ***p < 0.001)

References

    1. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet. 2021;398:535–54. - PubMed
    1. Passaro A, Brahmer J, Antonia S, Mok T, Peters S. Managing resistance to immune checkpoint inhibitors in lung cancer: treatment and novel strategies. J Clin Oncol. 2022;40:598–610. - PubMed
    1. Dasgupta Q, Jiang A, Wen AM, Mannix RJ, Man Y, Hall S, Javorsky E, Ingber DE. A human lung alveolus-on-a-chip model of acute radiation-induced lung injury. Nat Commun. 2023;14:6506–20. - PMC - PubMed
    1. Arroyo-Hernández M, Maldonado F, Lozano-Ruiz F, Muñoz-Montaño W, Nuñez-Baez M, Arrieta O. Radiation-induced lung injury: current evidence. BMC Pulm Med. 2021;21:9–21. - PMC - PubMed
    1. Käsmann L, Dietrich A, Staab-Weijnitz CA, Manapov F, Behr J, Rimner A, Jeremic B, Senan S, De Ruysscher D, Lauber K, Belka C. Radiation-induced lung toxicity-cellular and molecular mechanisms of pathogenesis, management, and literature review. Radiat Oncol. 2020;15:214–30. - PMC - PubMed

MeSH terms

LinkOut - more resources