Abstract
Systemic delivery of messenger RNA (mRNA) for tissue-specific targeting using lipid nanoparticles (LNPs) holds great therapeutic potential. Nevertheless, how the structural characteristics of ionizable lipids (lipidoids) impact their capability to target cells and organs remains unclear. Here we engineered a class of siloxane-based ionizable lipids with varying structures and formulated siloxane-incorporated LNPs (SiLNPs) to control in vivo mRNA delivery to the liver, lung and spleen in mice. The siloxane moieties enhance cellular internalization of mRNA-LNPs and improve their endosomal escape capacity, augmenting their mRNA delivery efficacy. Using organ-specific SiLNPs to deliver gene editing machinery, we achieve robust gene knockout in the liver of wild-type mice and in the lungs of both transgenic GFP and Lewis lung carcinoma (LLC) tumour-bearing mice. Moreover, we showed effective recovery from viral infection-induced lung damage by delivering angiogenic factors with lung-targeted Si5-N14 LNPs. We envision that our SiLNPs will aid in the clinical translation of mRNA therapeutics for next-generation tissue-specific protein replacement therapies, regenerative medicine and gene editing.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
All relevant data supporting the findings of this study are available within the paper, Supplementary Information or Source Data file. Source data are provided with this paper.
References
Chaudhary, N., Weissman, D. & Whitehead, K. A. mRNA vaccines for infectious diseases: principles, delivery and clinical translation. Nat. Rev. Drug Discov. 20, 817 (2021).
Pardi, N., Hogan, M. J., Porter, F. W. & Weissman, D. mRNA vaccines—a new era in vaccinology. Nat. Rev. Drug Discov. 17, 261 (2018).
Sahin, U., Karikó, K. & Türeci, O. mRNA-based therapeutics—developing a new class of drugs. Nat. Rev. Drug Discov. 13, 759 (2014).
Mendes, B. B. et al. Nanodelivery of nucleic acids. Nat. Rev. Methods Prim. 2, 24 (2022).
Pastor, F. et al. An RNA toolbox for cancer immunotherapy. Nat. Rev. Drug Discov. 17, 751 (2018).
Miao, L., Zhang, Y. & Huang, L. mRNA vaccine for cancer immunotherapy. Mol. Cancer 20, 41 (2021).
Zhang, H., Zhang, Y. & Yin, H. Genome editing with mRNA encoding ZFN, TALEN, and Cas9. Mol. Ther. 27, 735 (2019).
Yin, H., Kauffman, K. J. & Anderson, D. G. Delivery technologies for genome editing. Nat. Rev. Drug Discov. 16, 387 (2017).
Akinc, A. et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat. Nanotechnol. 14, 1084 (2019).
Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078 (2021).
Hajj, K. A. & Whitehead, K. A. Tools for translation: non-viral materials for therapeutic mRNA delivery. Nat. Rev. Mater. 2, 17056 (2017).
Finn, J. D. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 22, 2227 (2018).
Gillmore, J. D. et al. CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N. Engl. J. Med. 385, 493 (2021).
Whitehead, K. A. et al. Degradable lipid nanoparticles with predictable in vivo siRNA delivery activity. Nat. Commun. 5, 4277 (2014).
Qiu, M., Li, Y., Bloomer, H. & Xu, Q. Developing biodegradable lipid nanoparticles for intracellular mRNA delivery and genome editing. Acc. Chem. Res. 54, 4001 (2021).
Hou, X. et al. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. Nat. Nanotechnol. 15, 41 (2020).
Zhao, X. et al. Imidazole-based synthetic lipidoids for in vivo mRNA delivery into primary T lymphocytes. Angew. Chem. Int. Ed. 59, 20083 (2020).
Zhou, K. et al. Modular degradable dendrimers enable small RNAs to extend survival in an aggressive liver cancer model. Proc. Natl Acad. Sci. USA 113, 520 (2016).
Miao, L. et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat. Biotechnol. 37, 1174 (2019).
Xue, L. et al. Rational design of bisphosphonate lipid-like materials for mRNA delivery to the bone microenvironment. J. Am. Chem. Soc. 144, 9926 (2022).
Li, W. et al. Biomimetic nanoparticles deliver mRNAs encoding costimulatory receptors and enhance T cell mediated cancer immunotherapy. Nat. Commun. 12, 7264 (2021).
Cheng, Q. et al. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing. Nat. Nanotechnol. 15, 313 (2020).
Kulkarni, J. A., Witzigmann, D., Chen, S., Cullis, P. R. & van der Meel, R. Lipid nanoparticle technology for clinical translation of siRNA therapeutics. Acc. Chem. Res. 52, 2435 (2019).
Liu, S. et al. Membrane-destabilizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing. Nat. Mater. 20, 701 (2021).
Dilliard, S. A., Cheng, Q. & Siegwart, D. J. On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc. Natl Acad. Sci. USA 118, e2109256118 (2021).
Qiu, M. et al. Lung-selective mRNA delivery of synthetic lipid nanoparticles for the treatment of pulmonary lymphangioleiomyomatosis. Proc. Natl Acad. Sci. USA 119, e2116271119 (2022).
Shahbazi, M. A., Herranz, B. & Santos, H. A. Nanostructured porous Si-based nanoparticles for targeted drug delivery. Biomatter 2, 296 (2012).
Tang, F., Li, L. & Chen, D. Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery. Adv. Mater. 24, 1504 (2012).
Frampton, M. B. et al. Exploring the utility of hybrid siloxane-phosphocholine (SiPC) liposomes as drug delivery vehicles. RSC Adv. 11, 13014 (2021).
Semple, S. C. et al. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 28, 172 (2010).
Zhu, Y. et al. Multi-step screening of DNA/lipid nanoparticles and co-delivery with siRNA to enhance and prolong gene expression. Nat. Commun. 13, 4282 (2022).
Hu, B. et al. Thermostable ionizable lipid-like nanoparticles (iLAND) for RNAi treatment of hyperlipidemia. Sci. Adv. 8, eabm1418 (2022).
Ni, X., Kelly, S. S., Xu, S. & Xian, M. The path to controlled delivery of reactive sulfur species. Acc. Chem. Res. 54, 3968 (2021).
Behzadi, S. et al. Cellular uptake of nanoparticles: journey inside the cell. Chem. Soc. Rev. 46, 4218 (2017).
Wei, Y. et al. A cationic lipid with advanced membrane fusion performance for pDNA and mRNA delivery. J. Mater. Chem. B 11, 2095 (2023).
Tokudome, Y. et al. Preparation and characterization of ceramide-based liposomes with high fusion activity and high membrane fluidity. Colloids Surf. B 73, 92 (2009).
Akinc, A. et al. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics. Nat. Biotechnol. 26, 561 (2008).
Paunovska, K. et al. A direct comparison of in vitro and in vivo nucleic acid delivery mediated by hundreds of nanoparticles reveals a weak correlation. Nano Lett. 18, 2148 (2018).
Nagy, A. Cre recombinase: the universal reagent for genome tailoring. Genesis 26, 99 (2000).
Hajj, K. A. et al. A potent branched-tail lipid nanoparticle enables multiplexed mRNA delivery and gene editing in vivo. Nano Lett. 20, 5167 (2020).
Singh, B., Fu, C. & Bhattacharya, J. Vascular expression of the αvβ3-integrin in lung and other organs. Am. J. Physiol. Lung Cell. Mol. Physiol. 278, L217 (2000).
Alton, E. et al. Toxicology study assessing efficacy and safety of repeated administration of lipid/DNA complexes to mouse lung. Gene Ther. 21, 89 (2014).
Ebos, J. & Kerbel, R. S. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat. Rev. Clin. Oncol. 8, 210 (2011).
Xue, L. et al. High-throughput barcoding of nanoparticles identifies cationic, degradable lipid-like materials for mRNA delivery to the lungs in female preclinical models. Nat. Commun. 15, 1884 (2024).
Zhao, G. et al. TGF-βR2 signaling coordinates pulmonary vascular repair after viral injury in mice and human tissue. Sci. Trans. Med. 16, eadg6229 (2024).
Jia, T. et al. FGF-2 promotes angiogenesis through a SRSF1/SRSF3/SRPK1-dependent axis that controls VEGFR1 splicing in endothelial cells. BMC Biol. 19, 173 (2021).
Cao, R. et al. Comparative evaluation of FGF-2-, VEGF-A-, and VEGF-C-induced angiogenesis, lymphangiogenesis, vascular fenestrations, and permeability. Circ. Res. 94, 664 (2004).
Dahlman, J. E. et al. In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat. Nanotechnol. 9, 648 (2014).
McDermott, M. R., Brook, M. A. & Bartzoka, V. Adjuvancy effect of different types of silicone gel. J. Biomed. Mater. Res. 46, 132 (1999).
Huang, X. et al. Genome editing abrogates angiogenesis in vivo. Nat. Commun. 8, 112 (2017).
Wei, T. et al. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. Commun. 11, 3232 (2020).
Momany, F. & Rone, R. Validation of the general purpose QUANTA ®3.2/CHARMm® force field. J. Comput. Chem. 13, 888 (1992).
Acknowledgements
M.J.M. acknowledges support from a US National Institutes of Health (NIH) Director’s New Innovator Award (DP2 TR002776), a Burroughs Wellcome Fund Career Award at the Scientific Interface (CASI), an American Cancer Society Research Scholar Grant (RSG-22-122-01-ET), a US National Science Foundation CAREER Award (CBET-2145491) and the National Institutes of Health (NICHD R01 HD115877). A.E.V. acknowledges support from NIH grants (R01HL153539) and the Margaret Q. Landenberger Foundation. S.J.S. and K.L.S. are supported by an NSF Graduate Research Fellowship (award 1845298). R.P. was supported by an NIH F30 fellowship (F30HL162465-01A1). Z.X. is supported by a CRI Irvington fellowship (grant number CRI4168) from the Cancer Research Institute. We thank The Wistar Institute, the Pathology Core, Nucleic Acids Technology Core, and Program for Comparative Medicine at the Gene Therapy Program for technical assistance and I. Muthuramu for analysis of deep sequencing data. Elements in Figs. 1, 4, 5 and 6 were created with BioRender.com.
Author information
Authors and Affiliations
Contributions
L.X. and M.J.M. conceived the concept. L.X. and X.X. designed and synthesized all the siloxane-incorporated ionizable lipids used in this study. L.X., N.G., G.Z., X.H., C.C.W., V.C., R.P., R.E.-M. and Y.S. performed the experiments. L.X., N.G., S.J.S., K.L.S., K.W. and M.J.M. wrote the paper. L.X., G.Z., N.G., X.H., S.J.S., X.X., Z.X., R.P., J.X., K.L.S., C.C.W., R.E.-M., V.C., I.-C.Y., J.X., J.C., Y.S., M.-G.A., K.W., L.W., D.J.P., D.W., A.E.V., J.M.W. and M.J.M. reviewed and commented on the paper.
Corresponding author
Ethics declarations
Competing interests
L.X. and M.J.M. are inventors on a patent filed by the Trustees of the University of Pennsylvania (International Patent Application No. PCT/US23/66564) describing the lipid nanoparticle technology in this study. J.M.W. is a paid advisor to and holds equity in iECURE, Passage Bio and the Center for Breakthrough Medicines (CBM). He also holds equity in the former G2 Bio asset companies and Ceva Santé Animale. He has sponsored research agreements with Alexion Pharmaceuticals, Amicus Therapeutics, CBM, Ceva Santé Animale, Elaaj Bio, FA212, Foundation for Angelman Syndrome Therapeutics, former G2 Bio asset companies, iECURE and Passage Bio, which are licensees of Penn Technology. J.M.W., L.W. and C.C.W are inventors on patents that have been licensed to various biopharmaceutical companies and for which they may receive payments. D.W. is named on patents that describe the use of nucleoside-modified mRNA as a platform to deliver therapeutic proteins and vaccines. M.J.M., D.W. and M.-G.A. are also named on patents describing the use of lipid nanoparticles and lipid compositions for nucleic acid delivery. The other authors declare no competing interests.
Peer review
Peer review information
Nature Nanotechnology thanks Yusuke Sato 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 Formulation parameters and characterization of SiLNPs.
a, SiLNPs formulation parameters. Siloxane-incorporated lipidoids, DOPE, cholesterol, and C14PEG2K with molar ratio of 35%, 16%, 46.5%, and 2.5% were used for SiLNPs formulation. b, Representative cryogenic transmission electron microscopy (cryo-TEM) image of SiLNP morphology. Scale bar: 100 nm. c, Hydrodynamic size distribution of representative SiLNP.
Extended Data Fig. 2 Blood chemistry evaluation of mice after administration of Si6-C14b LNP co-delivering Cas9 mRNA and TTR sgRNA.
(a) AST, (b) ALT, (c) BUN, and (d) Creatinine levels of blood samples obtained from mice treated with PBS and Si6-C14b LNP (RNA dose: 3 mg kg−1). Data are presented as mean ± s.e.m. (n = 3 mice for PBS treated groups; n =4 for LNP treated groups).
Supplementary information
Supplementary Information
Supplementary Text, Tables 1–9, Schemes 1–5 and Figs. 1–51.
Supplementary Data 1
Statistical source data for the supplementary figures.
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 Extended Data Fig./Table 1
Statistical source data.
Source Data Extended Data Fig./Table 2
Statistical source data.
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.
About this article
Cite this article
Xue, L., Zhao, G., Gong, N. et al. Combinatorial design of siloxane-incorporated lipid nanoparticles augments intracellular processing for tissue-specific mRNA therapeutic delivery. Nat. Nanotechnol. (2024). https://doi.org/10.1038/s41565-024-01747-6
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41565-024-01747-6