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:

Self-assembled peptide–poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis

Abstract

Developing safe and efficient non-viral delivery systems remains a major challenge for in vivo applications of gene therapy, especially in cystic fibrosis. Unlike conventional cationic polymers or lipids, the emerging poloxamine-based copolymers display promising in vivo gene delivery capabilities. However, poloxamines are invalid for in vitro applications and their in vivo transfection efficiency is still low compared with viral vectors. Here, we show that peptides developed by modular design approaches can spontaneously form compact and monodisperse nanoparticles with poloxamines and nucleic acids via self-assembly. Both messenger RNA and plasmid DNA expression mediated by peptide-poloxamine nanoparticles are greatly boosted in vitro and in the lungs of cystic fibrosis mice with negligible toxicity. Peptide–poloxamine nanoparticles containing integrating vectors enable successful in vitro and in vivo long-term restoration of cystic fibrosis transmembrane conductance regulator deficiency with a safe integration profile. Our dataset provides a new framework for designing non-viral gene delivery systems qualified for in vivo genetic modifications.

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: Schematic of the multi-modular peptide-based gene transfection platform used in this study.
Fig. 2: Characterization and mechanism studies of the ternary complex and binary counterpart.
Fig. 3: In vitro evaluation of the ternary complex.
Fig. 4: In vitro long-term exogenous gene expression mediated by ternary complexes.
Fig. 5: In vivo evaluation of the ternary complex.
Fig. 6: In vivo long-term transgene expression mediated by ternary complexes.

Similar content being viewed by others

Data availability

All of the primary data that support the findings of this study are available from the corresponding author on request.

References

  1. Stoltz, D. A., Meyerholz, D. K. & Welsh, M. J. Origins of cystic fibrosis lung disease. N. Engl. J. Med. 372, 351–362 (2015).

    Google Scholar 

  2. Joseph, P. M. et al. Aerosol and lobar administration of a recombinant adenovirus to individuals with cystic fibrosis. I. Methods, safety, and clinical implications. Hum. Gene Ther. 12, 1369–1382 (2001).

    CAS  Google Scholar 

  3. Moss, R. B. et al. Repeated aerosolized AAV-CFTR for treatment of cystic fibrosis: a randomized placebo-controlled phase 2B trial. Hum. Gene Ther. 18, 726–732 (2007).

    CAS  Google Scholar 

  4. Konstan, M. W. et al. Compacted DNA nanoparticles administered to the nasal mucosa of cystic fibrosis subjects are safe and demonstrate partial to complete cystic fibrosis transmembrane regulator reconstitution. Hum. Gene Ther. 15, 1255–1269 (2004).

    CAS  Google Scholar 

  5. Elborn, J. S. Cystic fibrosis. Lancet 388, 2519–2531 (2016).

    CAS  Google Scholar 

  6. Yin, H. et al. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 15, 541–555 (2014).

    CAS  Google Scholar 

  7. Alton, E. W. F. W. et al. Repeated nebulisation of non-viral CFTR gene therapy in patients with cystic fibrosis: a randomised, double-blind, placebo-controlled, phase 2B trial. Lancet Respir. Med. 3, 684–691 (2015).

    CAS  Google Scholar 

  8. Cox, D. B. T., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121–131 (2015).

    CAS  Google Scholar 

  9. Richard-Fiardo, P. et al. Evaluation of tetrafunctional block copolymers as synthetic vectors for lung gene transfer. Biomaterials 45, 10–17 (2015).

    CAS  Google Scholar 

  10. Dahlman, J. E. et al. In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat. Nanotechnol. 9, 648–655 (2014).

    CAS  Google Scholar 

  11. Zuris, J. A. et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol. 33, 73–80 (2015).

    CAS  Google Scholar 

  12. Alvarez-Lorenzo, C., Rey-Rico, A., Sosnik, A., Taboada, P. & Concheiro, A. Poloxamine-based nanomaterials for drug delivery. Front. Biosci. (Elite Ed.) 2, 424–440 (2010).

    Google Scholar 

  13. Pitard, B. et al. Negatively charged self-assembling DNA/poloxamine nanospheres for in vivo gene transfer. Nucleic Acids Res. 32, e159 (2004).

    Google Scholar 

  14. Dowdy, S. F. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 35, 222–229 (2017).

    CAS  Google Scholar 

  15. Blanco, E., Shen, H. & Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33, 941–951 (2015).

    CAS  Google Scholar 

  16. Stewart, M. P. et al. Break and enter: in vitro and ex vivo strategies for intracellular delivery. Nature 538, 183–192 (2016).

    CAS  Google Scholar 

  17. Zeng, J., Wang, X. & Wang, S. Self-assembled ternary complexes of plasmid DNA, low molecular weight polyethylenimine and targeting peptide for nonviral gene delivery into neurons. Biomaterials 28, 1443–1451 (2007).

    CAS  Google Scholar 

  18. Tagalakis, A. D., He, L., Saraiva, L., Gustafsson, K. T. & Hart, S. L. Receptor-targeted liposome–peptide nanocomplexes for siRNA delivery. Biomaterials 32, 6302–6315 (2011).

    CAS  Google Scholar 

  19. Yin, H., Kauffman, K. J. & Anderson, D. G. Delivery technologies for genome editing. Nat. Rev. Drug Discov. 16, 387–399 (2017).

    CAS  Google Scholar 

  20. McNeer, N. A. et al. Nanoparticles that deliver triplex-forming peptide nucleic acid molecules correct F508del CFTR in airway epithelium. Nat. Commun. 6, 6952 (2015).

    CAS  Google Scholar 

  21. Kreda, S. M. et al. Characterization of wild-type and F508 cystic fibrosis transmembrane regulator in human respiratory epithelia. Mol. Biol. Cell 16, 2154–2167 (2005).

    CAS  Google Scholar 

  22. Writer, M. J. et al. Targeted gene delivery to human airway epithelial cells with synthetic vectors incorporating novel targeting peptides selected by phage display. J. Drug Target. 12, 185–193 (2004).

    CAS  Google Scholar 

  23. Hackett, P. B. Integrating DNA vectors for gene therapy. Mol. Ther. 15, 10–12 (2007).

    CAS  Google Scholar 

  24. Charizopoulou, N. et al. Spontaneous rescue from cystic fibrosis in a mouse model. BMC Genet. 7, 18 (2006).

    Google Scholar 

  25. Cheng, S. H. et al. Defective intracellular transport and processing of CFTR is the molecular basis of most cystic fibrosis. Cell 63, 827–834 (1990).

    CAS  Google Scholar 

  26. Dekkers, J. F. et al. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Nat. Med. 19, 939–945 (2013).

    CAS  Google Scholar 

  27. Alton, E. W. F. W. et al. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax 72, 137–147 (2017).

    Google Scholar 

  28. Nakai, H. et al. Large-scale molecular characterization of adeno-associated virus vector integration in mouse liver. J. Virol. 79, 3606–3614 (2005).

    CAS  Google Scholar 

  29. Tipanee, J., Chai, Y. C., VandenDriessche, T. & Chuah, M. K. Preclinical and clinical advances in transposon-based gene therapy. Biosci. Rep. 37, BSR20160614 (2017).

    CAS  Google Scholar 

  30. Kebriaei, P., Izsvák, Z., Narayanavari, S. A., Singh, H. & Ivics, Z. Gene therapy with the S leeping Beauty transposon system. Trends Genet. 33, 852–870 (2017).

    CAS  Google Scholar 

  31. Voigt, K. et al. Retargeting Sleeping Beauty transposon insertions by engineered zinc finger DNA-binding domains. Mol. Ther. 20, 1852–1862 (2012).

    CAS  Google Scholar 

  32. Kovač, A. & Ivics, Z. Specifically integrating vectors for targeted gene delivery: progress and prospects. Cell Gene Ther. Insights 3, 103–123 (2017).

    Google Scholar 

  33. Lee, C. M., Flynn, R., Hollywood, J. A., Scallan, M. F. & Harrison, P. T. Correction of the ΔF508 mutation in the cystic fibrosis transmembrane conductance regulator gene by zinc-finger nuclease homology-directed repair. Biores. Open Access 1, 99–108 (2012).

    CAS  Google Scholar 

  34. Schwank, G. et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653–658 (2013).

    CAS  Google Scholar 

  35. Suk, J. S. et al. The penetration of fresh undiluted sputum expectorated by cystic fibrosis patients by non-adhesive polymer nanoparticles. Biomaterials 30, 2591–2597 (2009).

    CAS  Google Scholar 

  36. Duncan, G. A., Jung, J., Hanes, J. & Suk, J. S. The mucus barrier to inhaled gene therapy. Mol. Ther. 24, 2043–2053 (2016).

    CAS  Google Scholar 

  37. Huckaby, J. T. & Lai, S. K. PEGylation for enhancing nanoparticle diffusion in mucus. Adv. Drug Deliv. Rev. 124, 125–139 (2018).

    CAS  Google Scholar 

  38. Kim, N., Duncan, G. A., Hanes, J. & Suk, J. S. Barriers to inhaled gene therapy of obstructive lung diseases: a review. J. Control. Release 240, 465–488 (2016).

    CAS  Google Scholar 

  39. Di Gioia, S. et al. Nanocomplexes for gene therapy of respiratory diseases: targeting and overcoming the mucus barrier. Pulm. Pharmacol. Ther. 34, 8–24 (2015).

    CAS  Google Scholar 

  40. Nafee, N. et al. Antibiotic-free nanotherapeutics: ultra-small, mucus-penetrating solid lipid nanoparticles enhance the pulmonary delivery and anti-virulence efficacy of novel quorum sensing inhibitors. J. Control. Release 192, 131–140 (2014).

    CAS  Google Scholar 

  41. Yang, M. et al. Biodegradable nanoparticles composed entirely of safe materials that rapidly penetrate human mucus. Angew. Chem. Int. Ed. Engl. 50, 2597–2600 (2011).

    CAS  Google Scholar 

  42. Suk, J. S. et al. Lung gene therapy with highly compacted DNA nanoparticles that overcome the mucus barrier. J. Control. Release 178, 8–17 (2014).

    CAS  Google Scholar 

  43. Mastorakos, P. et al. Highly compacted biodegradable DNA nanoparticles capable of overcoming the mucus barrier for inhaled lung gene therapy. Proc. Natl Acad. Sci. USA 112, 8720–8725 (2015).

    CAS  Google Scholar 

  44. Rogers, C. S. et al. Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science 321, 1837–1841 (2008).

    CAS  Google Scholar 

  45. Rosen, B. H. et al. Animal and model systems for studying cystic fibrosis. J. Cyst. Fibros. 17, S28–S34 (2018).

    CAS  Google Scholar 

  46. Cooney, A. L., Singh, B. K. & Sinn, P. L. Hybrid nonviral/viral vector systems for improved piggyBac DNA transposon in vivo delivery. Mol. Ther. 23, 667–674 (2015).

    CAS  Google Scholar 

  47. Bello-Roufaï, M., Lambert, O. & Pitard, B. Relationships between the physicochemical properties of an amphiphilic triblock copolymers/DNA complexes and their intramuscular transfection efficiency. Nucleic Acids Res. 35, 728–739 (2007).

    Google Scholar 

  48. Roques, C., Bouchemal, K., Ponchel, G., Fromes, Y. & Fattal, E. Parameters affecting organization and transfection efficiency of amphiphilic copolymers/DNA carriers. J. Control. Release 138, 71–77 (2009).

    CAS  Google Scholar 

  49. Johler, S. M., Rejman, J., Guan, S. & Rosenecker, J. Nebulisation of IVT mRNA complexes for intrapulmonary administration. PLoS ONE 10, e0137504 (2015).

    Google Scholar 

  50. Kormann, M. S. D. et al. Expression of therapeutic proteins after delivery of chemically modified mRNA in mice. Nat. Biotechnol. 29, 154–157 (2011).

    CAS  Google Scholar 

Download references

Acknowledgements

The authors are grateful to G. Vassaux (University of Nice Sophia Antipolis) and B. Pitard (University of Nantes) for introducing and providing poloxamine 704, and to S. Hyde (University of Oxford) for providing a CpG-depleted CFTR expression cassette. Special thanks go to S. L. Hart (University College London) for developing targeting elements showing high affinity to human airway epithelial cells. We also thank J. Geiger (Ethris) for providing SNIM-mRNA, M. K. Aneja (Ethris) for careful proofreading of the manuscript, Y. Liu, J. Rejman and J. Zhang (Ludwig Maximilian University of Munich) for technical support, and J. Tang (University of Queensland) for assistance with the Prism software. This work was supported by the Bundesministerium für Bildung und Forschung under project number 01GM1106A.

Author information

Authors and Affiliations

Authors

Contributions

S. Guan and J.R. conceived and developed the delivery system. C.R. and Z.I. generated the SB transposon system. A.M., G.H., S.H., W.G. and S. Guan designed and performed the animal experiments. S. Guan, L.Z. and A.S. characterized the nanoparticles. S. Guan, A.S. and S.M.J. performed the in vitro experiments. A.M., S.H., P.B., S. Glage, S.L., F.S. and C.M. performed the in vivo endpoint studies. Y.K. was responsible for the bioinformatics and integration sites analysis. B.T., C.R., Z.I. and J.R. designed and supervised the research. S. Guan and J.R. wrote the manuscript with comments from all authors.

Corresponding author

Correspondence to Joseph Rosenecker.

Ethics declarations

Competing interests

S. Guan and J.R. have filed patent applications on the subject matter related to this study. C.R. is a scientific co-founder and chief executive officer of the biotechnology company Ethris, which focuses on the development of mRNA therapies. G.H. is an employee of Ethris. Y.K. is an employee of Eurofins GATC Biotech. The other authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figure 1–28, Supplementary Tables 1,2, Supplementary Methods, Supplementary Results, Supplementary Notes, Supplementary References

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guan, S., Munder, A., Hedtfeld, S. et al. Self-assembled peptide–poloxamine nanoparticles enable in vitro and in vivo genome restoration for cystic fibrosis. Nat. Nanotechnol. 14, 287–297 (2019). https://doi.org/10.1038/s41565-018-0358-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-018-0358-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research