Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

An in vitro-transcribed circular RNA targets the mitochondrial inner membrane cardiolipin to ablate EIF4G2+/PTBP1+ pan-adenocarcinoma

Abstract

In vitro-transcribed (IVT) mRNA has arisen as a rapid method for the production of nucleic acid drugs. Here, we have constructed an oncolytic IVT mRNA that utilizes human rhinovirus type 2 (HRV2) internal ribosomal entry sites (IRESs) to selectively trigger translation in cancer cells with high expression of EIF4G2 and PTBP1. The oncolytic effect was provided by a long hGSDMDc .825 T>A/c.884 A>G-F1LCT mutant mRNA sequence with mitochondrial inner membrane cardiolipin targeting toxicity that triggers mitophagy. Utilizing the permuted intron-exon (PIE) splicing circularization strategy and lipid nanoparticle (LNP) encapsulation reduced immunogenicity of the mRNA and enabled delivery to eukaryotic cells in vivo. Engineered HRV2 IRESs-GSDMDp.D275E/E295G-F1LCT circRNA-LNPs (GSDMDENG circRNA) successfully inhibited EIF4G2+/PTBP1+ pan-adenocarcinoma xenografts growth. Importantly, in a spontaneous tumor model with abnormal EIF4G2 and PTBP1 caused by KRASG12D mutation, GSDMDENG circRNA significantly prevented the occurrence of pancreatic, lung and colon adenocarcinoma, improved the survival rate and induced persistent KRASG12D tumor antigen-specific cytotoxic T lymphocyte responses.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Construction of selective HRV2-IRES-driven GSDMDNT IVT mRNA.
Fig. 2: LNPs encapsulate HRV2 IRES-GSDMDNT IVT mRNA for eukaryotic cell delivery.
Fig. 3: The C-terminal F1L peptide translocates GSDMDNT into mitochondria.
Fig. 4: Acquired mitochondrial enzymatic hydrolytic activity via GSDMD base modification.
Fig. 5: PIE splicing to form circRNA.
Fig. 6: Safety and efficacy of GSDMDENG circRNA-LNPs.
Fig. 7: GSDMDENG circRNA-LNPs for the preventive treatment of pan-adenocarcinoma caused by KRAS mutation.

Similar content being viewed by others

Data availability

Source data for Figs. 17 and Extended Data Figs. 19 have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Sahin, U., Karikó, K. & Türeci, Ö. mRNA-based therapeutics–developing a new class of drugs. Nat. Rev. Drug Discov. 13, 759–780 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Pilkington, E. H. et al. From influenza to COVID-19: Lipid nanoparticle mRNA vaccines at the frontiers of infectious diseases. Acta Biomater. 131, 16–40 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078–1094 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. McKinlay, C. J. et al. Charge-altering releasable transporters (CARTs) for the delivery and release of mRNA in living animals. Proc. Natl Acad. Sci. USA 114, E448–E456 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kübler, H. et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/IIa study. J. Immunother. Cancer. 3, 26 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Sonenberg, N. & Hinnebusch, A. G. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell. 136, 731–745 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang, Y. & Wang, Z. IRES-mediated cap-independent translation, a path leading to hidden proteome. J. Mol. Cell. Biol. 11, 911–919 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pelletier, J. & Sonenberg, N. Internal initiation of translation of eukaryotic mRNA directed by a sequence derived from poliovirus RNA. Nature 334, 320–325 (1988).

    Article  CAS  PubMed  Google Scholar 

  9. de Breyne, S., Yu, Y., Unbehaun, A., Pestova, T. V. & Hellen, C. U. Direct functional interaction of initiation factor eIF4G with type 1 internal ribosomal entry sites. Proc. Natl Acad. Sci. USA 106, 9197–9202 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Henis-Korenblit, S. et al. The caspase-cleaved DAP5 protein supports internal ribosome entry site-mediated translation of death proteins. Proc. Natl Acad. Sci. USA 99, 5400–5405 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jain, R. et al. MicroRNAs enable mRNA therapeutics to selectively program cancer cells to self-destruct. Nucleic Acid Ther. 28, 285–296 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Van Hoecke, L. et al. Treatment with mRNA coding for the necroptosis mediator MLKL induces antitumor immunity directed against neo-epitopes. Nat. Commun. 9, 3417 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Broz, P., Pelegrín, P. & Shao, F. The gasdermins, a protein family executing cell death and inflammation. Nat. Rev. Immunol. 20, 143–157 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Heilig, R. et al. The Gasdermin-D pore acts as a conduit for IL-1β secretion in mice. Eur. J. Immunol. 48, 584–592 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Liu, X. et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535, 153–158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ding, J. et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535, 111–116 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Andreev, D. E. et al. Differential contribution of the m7G-cap to the 5’ end-dependent translation initiation of mammalian mRNAs. Nucleic Acids Res. 37, 6135–6147 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 486–541 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Matsumoto, K., Kusaka, J., Nishibori, A. & Hara, H. Lipid domains in bacterial membranes. Mol. Microbiol. 61, 1110–1117 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Rühl, S. et al. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science 362, 956–960 (2018).

    Article  PubMed  Google Scholar 

  22. Aits, S. et al. Sensitive detection of lysosomal membrane permeabilization by lysosomal galectin puncta assay. Autophagy. 11, 1408–1424 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Andrews, N. W., Almeida, P. E. & Corrotte, M. Damage control: cellular mechanisms of plasma membrane repair. Trends Cell Biol. 24, 734–742 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ketel, K. et al. A phosphoinositide conversion mechanism for exit from endosomes. Nature 529, 408–412 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Tatsuta, T., Scharwey, M. & Langer, T. Mitochondrial lipid trafficking. Trends Cell Biol. 24, 44–52 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Wasilenko, S. T., Stewart, T. L., Meyers, A. F. & Barry, M. Vaccinia virus encodes a previously uncharacterized mitochondrial-associated inhibitor of apoptosis. Proc. Natl Acad. Sci. USA 100, 14345–14350 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Weindel, C. G. et al. Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell. 185, 3214–3231 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Garland, K. M., Sheehy, T. L. & Wilson, J. T. Chemical and biomolecular strategies for STING pathway activation in cancer immunotherapy. Chem. Rev. 122, 5977–6039 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lazarou, M. et al. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524, 309–314 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Kunová, N. et al. Mitochondrial processing peptidases-structure, function and the role in human diseases. Int. J. Mol. Sci. 23, 1297 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Taylor, A. B. et al. Crystal structures of mitochondrial processing peptidase reveal the mode for specific cleavage of import signal sequences. Structure. 9, 615–625 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Kuang, S. et al. Structure insight of GSDMD reveals the basis of GSDMD autoinhibition in cell pyroptosis. Proc. Natl Acad. Sci. USA. 114, 10642–10647 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Anderson, B. R. et al. Nucleoside modifications in RNA limit activation of 2′-5′-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Res. 39, 9329–9338 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Enuka, Y. et al. Circular RNAs are long-lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res. 44, 1370–1383 (2016).

    Article  CAS  PubMed  Google Scholar 

  36. Petkovic, S. & Müller, S. RNA circularization strategies in vivo and in vitro. Nucleic Acids Res. 43, 2454–2465 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wesselhoeft, R. A., Kowalski, P. S. & Anderson, D. G. Engineering circular RNA for potent and stable translation in eukaryotic cells. Nat. Commun. 9, 2629 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Prota, G. et al. Enhanced immunogenicity of mitochondrial-localized proteins in cancer cells. Cancer Immunol. Res. 8, 685–697 (2020).

    Article  CAS  PubMed  Google Scholar 

  39. Kim, D., Xue, J. Y. & Lito, P. Targeting KRAS(G12C): From inhibitory mechanism to modulation of antitumor effects in patients. Cell. 183, 850–859 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li, S., Balmain, A. & Counter, C. M. A model for RAS mutation patterns in cancers: finding the sweet spot. Nat. Rev. Cancer 18, 767–777 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Yan, L. et al. Targeting glucose metabolism sensitizes pancreatic cancer to MEK inhibition. Cancer Res. 81, 4054–4065 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Brown, M. C. et al. Induction of viral, 7-methyl-guanosine cap-independent translation and oncolysis by mitogen-activated protein kinase-interacting kinase-mediated effects on the serine/arginine-rich protein kinase. J. Virol. 88, 13135–13148 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Brown, M. C., Dobrikov, M. I. & Gromeier, M. Mitogen-activated protein kinase-interacting kinase regulates mTOR/AKT signaling and controls the serine/arginine-rich protein kinase-responsive type 1 internal ribosome entry site-mediated translation and viral oncolysis. J. Virol. 88, 13149–13160 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell. 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pardi, N., Hogan, M. J., Porter, F. W. & Weissman, D. mRNA vaccines: a new era in vaccinology. Nat. Rev. Drug Discov. 17, 261–279 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Van Hoecke, L. et al. mRNA in cancer immunotherapy: beyond a source of antigen. Mol. Cancer. 20, 48 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ahmadpour, S. T., Mahéo, K., Servais, S., Brisson, L. & Dumas, J. F. Cardiolipin, the mitochondrial signature lipid: implication in cancer. Int. J. Mol. Sci. 21, 8031 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ziegler, P. K. et al. Mitophagy in intestinal epithelial cells triggers adaptive immunity during tumorigenesis. Cell. 174, 88–101.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Komar, A. A. & Hatzoglou, M. Cellular IRES-mediated translation: the war of ITAFs in pathophysiological states. Cell Cycle. 10, 229–240 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Martínez-Salas, E. et al. RNA-binding proteins impacting on internal initiation of translation. Int. J. Mol. Sci. 14, 21705–21726 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Komar, A. A. & Hatzoglou, M. Exploring internal ribosome entry sites as therapeutic targets. Front. Oncol. 5, 233 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Brown, M. C. et al. Cancer immunotherapy with recombinant poliovirus induces IFN-dominant activation of dendritic cells and tumor antigen-specific CTLs. Sci. Transl. Med. 9, eaan4220 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Dobrikova, E. Y. et al. Attenuation of neurovirulence, biodistribution, and shedding of a poliovirus:rhinovirus chimera after intrathalamic inoculation in Macaca fascicularis. J. Virol. 86, 2750–2759 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cobbold, L. C. et al. Identification of internal ribosome entry segment (IRES)-trans-acting factors for the Myc family of IRESs. Mol. Cell. Biol. 28, 40–49 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Li, S. et al. MiR-144-3p-mediated dysregulation of EIF4G2 contributes to the development of hepatocellular carcinoma through the ERK pathway. J. Exp. Clin. Cancer Res. 40, 53 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Georgilis, A. et al. PTBP1-mediated alternative splicing regulates the inflammatory secretome and the pro-tumorigenic effects of senescent cells. Cancer Cell. 34, 85–102.e9 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sawicka, K., Bushell, M., Spriggs, K. A. & Willis, A. E. Polypyrimidine-tract-binding protein: a multifunctional RNA-binding protein. Biochem. Soc. Trans. 36, 641–647 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Wood, K., Hensing, T., Malik, R. & Salgia, R. Prognostic and predictive value in KRAS in non-small-cell lung cancer: a review. JAMA Oncol. 2, 805–812 (2016).

    Article  PubMed  Google Scholar 

  59. Nagasaka, M. et al. KRAS inhibitors—yes, but what next? Direct targeting of KRAS- vaccines, adoptive T cell therapy and beyond. Cancer Treat. Rev. 101, 102309 (2021).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was supported by the National Natural Science Foundation of China (82072739, 81800082, 81172790 and 81671586), The Recruitment Program of Overseas High-Level Young Talents, ‘Innovative and Entrepreneurial Talent and Team’ (No. (2018) 2015) of Jiangsu Province; Beijing Xisike Clinical Oncology Research Foundation (YBMS2019-071 and Y-MSDZD2021-0169), the start-up funding of Southeast University (No. 4024002312), the First Affiliated Hospital; Excellent Scientific Research and Innovation Team in Natural Sciences of Anhui province (2022AH010073); the National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine), National Medical Research Council (CG21APR1005, MOH-001388-00), Singapore Ministry of Education (MOE-000387-00) and National Research Foundation (NRF-000352-00). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Z.F. and X.C. conceived and designed this study. Z.F., J.Z. and Q.L. performed the construction, breeding, reproduction and experiments of animal models. X.Z. assisted in the synthesis of LNPs and mRNA assembly, as well as electron microscope detection. L.C. and Z.X. performed data analysis, interpreted the results and maintained RNA quality control. Q.C. collected clinical samples. M.W. provided pathological support. Z.F., J.Z. and Q.L. performed the experiments and data collection. Z.F. wrote the original draft. X.J., H.X. and X.C. supervised the project and revised the manuscript, with all authors contributing to writing and providing feedback.

Corresponding authors

Correspondence to Xiaochun Jiang, Hongping Xia or Xiaoyuan Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Michael Mitchell, Dan Peer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Expression of EIF4G2 and PTBP1 in tumors.

(a and b) Dot plot and (c and d) Bar plot of the EIF4G2 and PTBP1 mRNA expression profile across all tumor samples and paired normal tissues (www.gepia.cancer-pku.cn.). (e and f) Overall survival of eIF4G2 and PTBP1 in DLBC, PAAD, COAD, LUAD and GBM. (g) Detection of eIF4E2, eIF4G2 and PTBP1 mRNA in five types of tumor cell lines by qPCR. Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by analysis of variance (ANOVA) followed by a post hoc test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 2 Expression of IRES-driven dicistronics in tumor cell lines.

The dicistronic IVT-mRNA constructs based on (a) EMCV-IRES, (b) PV_type1_Mahoney IRES and (c) PV_type3_Leon IRES were electrotransfected into tumor cell lines, and after 48 h, the fluorescence intensities of Renilla (480 nm) and Firefly (560 nm) luciferases were detected. Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by analysis of variance (ANOVA) followed by a post hoc test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 3 Expression of HRV2 IRES-GSDMDNT IVT mRNA-LNPs in tumor cell lines.

(a) Representative single-cell fluorescence microscopy images of HRV2 IRES-GSDMDNT IVT mRNA-LNPs transfected with DLBC, COAD, LUAD and GBM and corresponding noncancer/nonmalignant cell lines. Scale bar: 5 μm. (b) After HRV2 IRES-GSDMDNT IVT mRNA-LNPs were transfected into DLBC, COAD, LUAD and GBM and corresponding noncancer/nonmalignant cell lines, the fluorescence intensity of eGFP on the cell surface was detected by flow cytometry. (c and d) The dose‒response relationship of transfected LNPs encapsulating HRV2 IRES-GSDMDNT IVT mRNA in WT or CHMP4A−/− DLBC, COAD, LUAD and GBM cell lines. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 4 N/MTS-linked GSDMDNT IVT-mRNA targets mitochondria but is nonfunctional.

(a) Schematic diagram of the ligation of MTS peptide mRNA to the N terminus of GSDMDNT mRNA. (b) After encapsulating LNPs with N/MTS-GSDMDNT IVT-mRNA, they were transfected into AsPC-1 and hTRET-hPNE cells, and the expression of GSDMDNT in the isolated cytoplasm and mitochondria was detected. (c) Rb.sN/MTS-GSDMDNT was incubated with lipid strips, followed by detection of specific lipid binding with anti-GSDMDNT antibody. (d) Cleavage of PI(4,5)P2 and cardiolipin liposomes by Rb.sN/MTS-GSDMDNT was monitored by measuring released Tb3+. (e) Schematic diagram of the construction of His-tagged GSDMDNT truncated polypeptides of different lengths. (f) Coimmunoprecipitation of truncated His-tagged GSDMDNT with Flag-N-MTS polypeptide. (g) Coimmunoprecipitation of truncated His-tagged GSDMDNT with Flag-FL1CT polypeptide. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 5 GSDMDNT-F1LCT-LNPs target mitochondria without plasma membrane toxicity.

(a) Validation of polystyrene bead size standard resolution at the 1, 2 and 4 μm levels by FACS. Forward scatter (FSC-A) x-axis and side scatter (SSC-A) y-axis. (b) The effects of GSDMDNT-F1LCT-LNPs on the expression of p-DRP1S616 and p-DRP1S637 in AsPC-1 and hTRET-hPNE cells. (c) Representative fluorescence microscopy images of the expression distribution of CHMP4A and Tsg101 in hTRET-hPNE and AsPC-1 cells 24 h after transfection with GSDMDNT-F1LCT-LNPs. Scale bar: 10 μm. (d)The AsPC-1 cells were pretreated with the pRIP inhibitor Nec-1, MLKL siRNA, and mtROS scavenger Necrox-5 before induction of cell death by GSDMDNT-F1LCT-LNPs for 48 h. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 6 GSDMDp. E295G-F1LCT mRNA-LNP targets mitochondria without subcellular organelle membrane toxicity.

(a, b) Representative histograms of cell lysosomal and endoplasmic reticulum volumes detected by flow cytometry after GSDMDp. E295G-F1LCT mRNA-LNP-transfected hTRET-hPNE cells; the right panel shows the quantitative statistics. (c) Representative scatter plots of mitochondrial membrane potential measured by GSDMDp. E295G-F1LCT mRNA-LNPs after transfection of hTRET-hPNE and AsPC-1 cells by flow cytometry. The right panel shows the percentage quantification of cells with low mitochondrial membrane potential. (d) The ratio of MitoSox (cytoplasmic ROS) and MitoTracker Green (MTG, mitochondrial ROS) was measured to assess the cytoplasmic leakage of mtROS. (e) The cytosolic fraction of mtDNA (ND1) and control nuclear DNA (β-actin) were assayed by qPCR to assess mtDNA leakage. (f) Representative fluorescence microscopy images of the expression distribution of CHMP4A and Tsg101 in hTRET-hPNE and AsPC-1 cells. Scale bar: 10 μm. (g) The dose‒response relationship of transfected LNPs encapsulating HRV2 IRES-GSDMDp. E295G-F1LCT mRNA in WT or DLBC, COAD, LUAD and GBM cell lines. Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by analysis of variance (ANOVA) followed by a post hoc test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 7 m6A-modified GSDMDENGcircRNA-LNPs attenuates non-antiviral immunogenic in vitro.

(a–f) Detection of rigi, mda5, oas1 and ifnb1 mRNA in five types of human normal cell lines and Primary Dermal Fibroblast (HDFa) by qPCR. Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by analysis of variance (ANOVA) followed by a post hoc test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 8 GSDMDEngcircRNA-LNPs shrink tumors in xenogeneic tumor-bearing mice.

(a) After 42 days of 400 μg/kg GSDMDEngcircRNA-LNPs i.v., the levels of AST, ALT, BUN, creatinine and platelets in the blood of mice were detected. (b, c) Human SU-DH-5, SW480, HCC-827 and U118-MG cells were subcutaneously inoculated into NGS mice with a humanized immune system, and 400 μg/kg GSDMDEngcircRNA-LNPs were injected peritumorally. Tumor size was continuously monitored for the following 42 days, and tumor weights were recorded on day 42. Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by Student’s t test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 9 Tumor-specific mitochondrial toxicity of GSDMDENGcircRNA-LNPs in vivo.

(a) Flow cytometry detection of FSC-A of isolated mitochondria from AsPC-1 tumor-bearing mouse tissues relative to a representative histogram of standard beads 42 days after peritumoral injection of 400 μg/kg GSDMDEngcircRNA-LNPs. (b) qPCR detection of mtDNA leakage in tissues of AsPC-1 tumor-bearing mice peritumorally injected with 400 μg/kg GSDMDENGcircRNA-LNPs. (c) Representative scatter plot of mitochondrial membrane potential measured by flow cytometry in tissues of AsPC-1 tumor-bearing mice peritumorally injected with 400 μg/kg GSDMDENGcircRNA-LNPs. (d) STING−/− AsPC-1 and MLKL−/− AsPC-1 cells were subcutaneously inoculated into NGS mice with a humanized immune system, and on day 3, 400 μg/kg GSDMDENG circRNA-LNPs were injected peritumorally, and the tumor size was continuously monitored for the following 42 days (bottom). (e) Mice were injected with AsPC-1-mtOVA cells encoding mtOVA fused with mCherry. 400 μg/kg GSDMDENG circRNA-LNPs were injected peritumorally, and on day 14 mice were sacrificed and cells from DLNs were stained and mCherry+ CD11b + CD11c+of APCs were identified, and the expression of CD86/CD80/CD40 in mCherry+ CD11c + CD103 + CD11b- or mCherry- CD11c + CD103 + CD11b-. (f) Adoptive transfer of CFSE-labeled OVA-specific transgenes CD8+ (OT-I) and CD4+ (OT-II) into WT, STING−/− and MLKL−/− AsPC-1-mtOVA tumor-bearing mice, followed by 400 μg/kg GSDMDENG circRNA-LNPs were injected peritumorally, and the proliferation activity of DLNs OT-I (left) and OT-II (right) cells was detected by flow cytometry. (g) Analysis of OVA OT-I (top)- or OVA OT-II (bottom)-specific IFN-γ T-cell responses in the DLNs of AsPC-1-mtOVA tumor-bearing mice by IFN-γ ELISPOT. The quantification plot on the right depicts the number of puncta from a single mouse DLNs.Data are shown as the mean ± SD, *p < 0.05 and **p < 0.01 by analysis of variance (ANOVA) followed by a post hoc test. All cell experiment data were derived from 3 independent experiments.

Source data

Extended Data Fig. 10 Schematic diagram of GSDMDENG circRNA-LNPs therapy and prevention of adenocarcinoma.

GSDMDENG circRNA-LNPs exhibit dual properties of tumor-specific expression and conditional activation. The HRV2-IRES structure ensures translation initiation in KRAS mutation-driven EIF4G2/PTBP1-activated cancer cells, while engineered double mutations and C-terminal signal peptides enable their mitochondrial localization and activation. Cyclization strategies and lipid nanoparticle encapsulation enhance their expression and delivery efficiency in vivo. These comprehensive strategies hold great promise for using GSDMDENG circRNA-LNPs in the treatment and prevention of KRAS mutation-induced pan-adenocarcinoma.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig./Table 1

Statistical Source Data

Source Data Extended Data Fig./Table 2

Statistical Source Data

Source Data Extended Data Fig./Table 3

Statistical Source Data

Source Data Extended Data Fig./Table 4

Statistical Source Data

Source Data Extended Data Fig./Table 5

Statistical Source Data

Source Data Extended Data Fig./Table 6

Statistical Source Data

Source Data Extended Data Fig./Table 7

Statistical Source Data

Source Data Extended Data Fig./Table 8

Statistical Source Data

Source Data Extended Data Fig./Table 9

Statistical Source Data

Source Data Extended Data Fig./Table 10

Unprocessed western blots

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Feng, Z., Zhang, X., Zhou, J. et al. An in vitro-transcribed circular RNA targets the mitochondrial inner membrane cardiolipin to ablate EIF4G2+/PTBP1+ pan-adenocarcinoma. Nat Cancer 5, 30–46 (2024). https://doi.org/10.1038/s43018-023-00650-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00650-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer