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
. 2023 Jul 15;14(1):4223.
doi: 10.1038/s41467-023-39938-9.

mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy

Affiliations

mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy

Fengqiao Li et al. Nat Commun. .

Abstract

Synergistically improving T-cell responsiveness is promising for favorable therapeutic outcomes in immunologically cold tumors, yet current treatments often fail to induce a cascade of cancer-immunity cycle for effective antitumor immunity. Gasdermin-mediated pyroptosis is a newly discovered mechanism in cancer immunotherapy; however, cleavage in the N terminus is required to activate pyroptosis. Here, we report a single-agent mRNA nanomedicine-based strategy that utilizes mRNA lipid nanoparticles (LNPs) encoding only the N-terminus of gasdermin to trigger pyroptosis, eliciting robust antitumor immunity. In multiple female mouse models, we show that pyroptosis-triggering mRNA/LNPs turn cold tumors into hot ones and create a positive feedback loop to promote antitumor immunity. Additionally, mRNA/LNP-induced pyroptosis sensitizes tumors to anti-PD-1 immunotherapy, facilitating tumor growth inhibition. Antitumor activity extends beyond the treated lesions and suppresses the growth of distant tumors. We implement a strategy for inducing potent antitumor immunity, enhancing immunotherapy responses in immunologically cold tumors.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1. Schematic of antitumor immunity via GSDMBNT mRNA@LNP-mediated pyroptosis and characterization of GSDMBNT mRNA@LNPs.
a Intratumoral administration of mRNA lipid nanoparticles encoding only the N-terminal domain of GSDMB triggers pyroptosis, eliciting antitumor immunity and facilitating anti-PD-1-mediated immunotherapy in immunologically cold tumors. b TEM image of GSDMBNT mRNA@LNPs. Scale bar = 200 nm. c The particle size, PDI, and zeta potential of GSDMBNT mRNA@LNPs were analyzed by DLS. Data are representative of four independent experiments. d DLS measurement of GSDMBNT mRNA@LNPs under the conditions indicated, including pH 6.5 buffer, pH 7.4 buffer, 10% or 20% plasma. Data are presented as means ± SD (n = 3). e Cellular uptake of FITC-labeled LNPs (Luc mRNA@FITCLNPs) was monitored in HEK 293, HeLa, 4T1, and B16F10 cells at different time points. Scale bar = 20 μm. f LNP-mediated endosomal/lysosomal escape and cytoplasmic release of Luc Cy5mRNA in HEK 293, HeLa, 4T1, and B16F10 cells 4 h after incubation. DAPI (blue), Endo/lysosome (green), Luc Cy5mRNA@LNPs (red), scale bar = 20 μm. Data shown in e, f are representative of two independent experiments. Source data are provided as a Source Data file. Cartoon in panel a was created with BioRender.com.
Fig. 2
Fig. 2. Lipid nanoparticles deliver mRNA encoding the N-terminal domain of GSDMB into cells to induce pyroptosis.
a Cell morphologies of the treated HEK 293, HeLa, 4T1 and B16F10 cells were detected using a confocal microscope. Before imaging, cells were treated with annexin V-FITC and propidium iodide (PI) and incubated for 15 min. Scale bars = 20 μm. Data are representative of three independent experiments. b LDH release-based cell death assay in HEK 293, HeLa, 4T1, and B16F10 cells after treatment with naked GSDMBNT mRNA, LNPs, or GSDMBNT mRNA@LNPs, respectively. Data are presented as means ± SD (n = 3). Statistical significance was calculated via one-way ANOVA. c Flow-cytometry analysis of cells positive for propidium iodide and annexin V. Data are presented as means ± SD (n = 3). Statistical significance was calculated using a two-tailed Student’s t test. Untreated cells served as the control (Ctrl) in all experiments. Source data are provided as a Source Data file.
Fig. 3
Fig. 3. GSDMBNT mRNA@LNPs induce immunogenic pyroptosis and stimulate the maturation of bone marrow-derived dendritic cells (BMDCs).
a A confocal microscope was used to detect CRT expression in treated HEK 293, HeLa, 4T1, and B16F10 cells. Scale bar = 20 μm. Data are representative of three independent experiments. b, c Extracellular HMGB1 and ATP expression were analyzed by ELISA in HEK 293, HeLa, 4T1, and B16F10 cells after different treatments. Data in b are presented as means ± SD (n = 3). Data in c are presented as means ± SD (n = 4). Statistical significance was calculated using a two-tailed Student’s t test. d, e Immune stimulation of BMDCs by GSDMBNT mRNA@LNPs. B16F10 cells were pretreated with PBS (Ctrl), naked GSDMBNT mRNA, blank LNPs or GSDMBNT mRNA@LNPs, followed by coculture with BMDCs for 48 h. d Quantitative determination of proinflammatory cytokines IFN-γ, IL-1β and TNF-α using ELISA assay. Data are presented as means ± SD (n = 3). Statistical significance was calculated via one-way ANOVA. e Analysis of DC maturation biomarkers (MHC-II and CD86) using flow cytometry (n = 3). Source data are provided as a Source Data file.
Fig. 4
Fig. 4. Treatment with GSDMBNT mRNA@LNPs promotes tumor control in an anti-PD-1-resistant 4T1 breast cancer mouse model.
a 6, 24, and 72 h after a single dose of GSDMBNT mRNA@LNPs (40 μg mRNA), cytokine concentrations were measured in tumor tissue or serum by ELISA. Data are presented as means ± SD (n = 4 mice per group). Statistical significance was calculated using a two-tailed Student’s t test. b Experimental timeline for treatment of 4T1 orthotopic tumor-bearing mice. s.c., subcutaneous; it, intratumoral; ip, intraperitoneal. c Individual growth curves of tumor size for mice treated as indicated. d, e The average tumor growth curves and survival percentages for mice treated as indicated (n = 7 mice per group). Data shown in e are represented as means ± SD. P values were determined by two-tailed unpaired Student’s t test in c, d or by log-rank (Mantel-Cox) test in e. Source data are provided as a Source Data file. Panel b was created with BioRender.com.
Fig. 5
Fig. 5. Treatment with GSDMBNT mRNA@LNPs induces antitumor immunity in an anti-PD-1-resistant 4T1 breast cancer mouse model.
a Immunofluorescence staining of tumors for CD8+ T cell infiltration and CRT expression after indicated treatments. Scale bar = 50 μm. b Quantitative analysis of immunofluorescence staining in terms of CD8+ and CRT intensities. Data are presented as means ± SD (n = 5). Statistical significance was calculated using a two-tailed Student’s t test. c ELISA analysis of IL-1β, IL-18, and HMGB1 in tumor and serum samples from 4T1 tumor-bearing mice receiving the treatments indicated. Data are presented as means ± SD (n = 4 mice per group). Statistical significance was calculated using a two-tailed Student’s t test. d Blood samples were collected on the second day after the final injection for aminotransaminase analyses. Data are presented as means ± SD (n = 4 mice per group). Source data are provided as a Source Data file.
Fig. 6
Fig. 6. Treatment with GSDMBNT mRNA@LNPs enhances potent antitumor activity in an aggressive melanoma mouse model.
a Experimental timeline for treatment of B16F10 tumor-bearing mice. b, c In vivo bioluminescence images and quantification of luciferase signals in mice treated as indicated for monitoring tumor growth. Imaging was performed every 5 days from the initial treatment day (day 7 after tumor inoculation) until day 23. d The survival percentages for mice treated as indicated. n = 8 mice for PBS, aPD-1, or GSDMBNT mRNA@LNP treatment groups, n = 7 mice for LNP treatment group, and n = 10 mice for aPD-1 + GSDMBNT mRNA@LNP treatment group. Survival analysis was analyzed using the log-rank (Mantel-Cox) test. e ELISA analysis of HMGB1 in the supernatant of B16F10 tumors excised from mice treated as indicated. Data are presented as means ± SD (n = 4 mice per group). fh Representative images and quantitative analysis of immunofluorescence staining for CD8+ T cell infiltration and CRT expression in tumors after indicated treatments. Scale bar = 50 μm. Results are presented as means ± SD (n = 5). Statistical significance was calculated using a two-tailed Student’s t test. Source data are provided as a Source Data file. Panel a is created with BioRender.com.
Fig. 7
Fig. 7. Treatment with GSDMBNT mRNA@LNPs remodels the tumor microenvironment in an aggressive melanoma mouse model.
a The experimental timeline for the treatment of B16F10 tumor-bearing mice and cytokines and immune cell tests were conducted on day 18. b Cytokine concentrations were measured in tumor tissue or serum by ELISA. c Flow cytometry analysis results of the percentage of CD11c+ MHC-II+ DCs, CD3+ CD4+ T cells, CD3+ CD8+ T cells, CD3- NK1.1+ T cells, CD3+ NK1.1+ T cells, and monocytes isolated from lymph nodes or tumors. d Tumor weights of B16F10 tumor-bearing mice with different treatments. All results are presented as means ± SD (n = 4 mice per group). Statistical significance was calculated using a two-tailed Student’s t test. Source data are provided as a Source Data file. Panel a was created with BioRender.com.
Fig. 8
Fig. 8. Local combinational treatment of GSDMBNT mRNA@LNPs and aPD-1 controls tumor burden at distant sites.
C57BL/6 mice (n = 4 mice per group) were inoculated subcutaneously (s.c.) with 5 × 105 and 2.5 × 105 B16F10 cells in the left and right flanks, respectively. a Experimental timeline for treatment of B16F10 dual-tumor-bearing mice. b In vivo bioluminescence images of luciferase signals in mice treated as indicated for monitoring tumor growth. Imaging was performed every 5 days from the initial treatment day (day 7 after tumor inoculation) until day 23. c Individual curves of luciferase signals for mice treated as indicated. d, e Representative images and quantitative analysis of immunofluorescence staining for CD8+ T cell infiltration and CRT expression in tumors after indicated treatments. Scale bar = 50 μm. Results are presented as means ± SD (n = 5). Statistical significance was calculated using a two-tailed Student’s t test. Source data are provided as a Source Data file. Panel a is created with BioRender.com.

Similar articles

Cited by

References

    1. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019;18:175–196. doi: 10.1038/s41573-018-0006-z. - DOI - PMC - PubMed
    1. Tan S, Li D, Zhu X. Cancer immunotherapy: Pros, cons and beyond. Biomed. Pharmacother. 2020;124:109821. doi: 10.1016/j.biopha.2020.109821. - DOI - PubMed
    1. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 2018;15:325–340. doi: 10.1038/nrclinonc.2018.29. - DOI - PMC - PubMed
    1. Dine J, Gordon R, Shames Y, Kasler MK, Barton-Burke M. Immune checkpoint inhibitors: an innovation in immunotherapy for the treatment and management of patients with cancer. Asia-Pac. J. Oncol. Nurs. 2017;4:127–135. doi: 10.4103/apjon.apjon_4_17. - DOI - PMC - PubMed
    1. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–723. doi: 10.1016/j.cell.2017.01.017. - DOI - PMC - PubMed

Publication types