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.

  • Letter
  • Published:

Efficient delivery of RNAi prodrugs containing reversible charge-neutralizing phosphotriester backbone modifications

Subjects

Abstract

RNA interference (RNAi) has great potential to treat human disease1,2,3. However, in vivo delivery of short interfering RNAs (siRNAs), which are negatively charged double-stranded RNA macromolecules, remains a major hurdle4,5,6,7,8,9. Current siRNA delivery has begun to move away from large lipid and synthetic nanoparticles to more defined molecular conjugates9. Here we address this issue by synthesis of short interfering ribonucleic neutrals (siRNNs) whose phosphate backbone contains neutral phosphotriester groups, allowing for delivery into cells. Once inside cells, siRNNs are converted by cytoplasmic thioesterases into native, charged phosphodiester-backbone siRNAs, which induce robust RNAi responses. siRNNs have favorable drug-like properties, including high synthetic yields, serum stability and absence of innate immune responses. Unlike siRNAs, siRNNs avidly bind serum albumin to positively influence pharmacokinetic properties. Systemic delivery of siRNNs conjugated to a hepatocyte-specific targeting domain induced extended dose-dependent in vivo RNAi responses in mice. We believe that siRNNs represent a technology that will open new avenues for development of RNAi therapeutics.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Synthesis and biophysical properties of bioreversible phosphotriester siRNN oligonucleotides.
Figure 2: Biological activity of siRNNs.
Figure 3: In vivo delivery of GalNAc-siRNN conjugates.

Similar content being viewed by others

References

  1. Castanotto, D. & Rossi, J.J. The promises and pitfalls of RNA-interference-based therapeutics. Nature 457, 426–433 (2009).

    Article  CAS  Google Scholar 

  2. Pecot, C.V., Calin, G.A., Coleman, R.L., Lopez-Berestein, G. & Sood, A.K. RNA interference in the clinic: challenges and future directions. Nat. Rev. Cancer 11, 59–67 (2011).

    Article  CAS  Google Scholar 

  3. Davidson, B.L. & McCray, P.B. Jr. Current prospects for RNA interference-based therapies. Nat. Rev. Genet. 12, 329–340 (2011).

    Article  CAS  Google Scholar 

  4. Behlke, M.A. Chemical modification of siRNAs for in vivo use. Oligonucleotides 18, 305–320 (2008).

    Article  CAS  Google Scholar 

  5. Rettig, G.R. & Behlke, M.A. Progress toward in vivo use of siRNAs-II. Mol. Ther. 20, 483–512 (2012).

    Article  CAS  Google Scholar 

  6. Bramsen, J.B. & Kjems, J. Chemical modification of small interfering RNA. Methods Mol. Biol. 721, 77–103 (2011).

    Article  CAS  Google Scholar 

  7. Meade, B.R. & Dowdy, S.F. The road to therapeutic RNA interference (RNAi): tackling the 800 pound siRNA delivery gorilla. Discov. Med. 8, 253–256 (2009).

    PubMed  Google Scholar 

  8. Zhou, J., Shum, K.-T., Burnett, J.C. & Rossi, J.J. Nanoparticle-based delivery of RNAi therapeutics: progress and challenges. Pharmaceuticals 6, 85–107 (2013).

    Article  CAS  Google Scholar 

  9. Kanasty, R., Dorkin, J.R., Vegas, A. & Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 12, 967–977 (2013).

    Article  CAS  Google Scholar 

  10. Lipinski, C.A., Lombardo, F., Dominy, B.W. & Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 46, 3–26 (2001).

    Article  CAS  Google Scholar 

  11. Joshua-Tor, L. & Hannon, G.J. Ancestral roles of small RNAs: an Ago-centric perspective. Cold Spring Harb. Perspect. Biol. 3, a003772 (2011).

    Article  Google Scholar 

  12. Carthew, R.W. & Sontheimer, E.J. Origins and mechanisms of miRNAs and siRNAs. Cell 136, 642–655 (2009).

    Article  CAS  Google Scholar 

  13. Reese, C.B. Oligo- and poly-nucleotides: 50 years of chemical synthesis. Org. Biomol. Chem. 3, 3851–3868 (2005).

    Article  CAS  Google Scholar 

  14. Gates, K.S. An overview of chemical processes that damage the cellular DNA: spontaneous hydrolysis, alkylation, and reactions with radicals. Chem. Res. Toxicol. 22, 1747–1760 (2009).

    Article  CAS  Google Scholar 

  15. Grajkowski, A., Wilk, A., Chmielewski, M.K., Phillips, L.R. & Beaucage, S.L. The 2-(N-formyl-N-methyl) aminoethyl group as a potential phosphate/thiophosphate protecting group in solid-phase oligodeoxyribonucleotide synthesis. Org. Lett. 3, 1287–1290 (2001).

    Article  CAS  Google Scholar 

  16. Dellinger, D.J., Sheehan, D.M., Christensen, N., Lindberg, J.G. & Caruthers, M.H. Solid phase chemical synthesis of phosphonoacetate and thiophosphonoacetate oligodeoxynucleotides. J. Am. Chem. Soc. 125, 940–950 (2003).

    Article  CAS  Google Scholar 

  17. Tanabe, K., Ando, Y. & Nishimoto, S. Reversible modification of oligodeoxynucleotides: click reaction at phosphate group and alkali treatment. Tetrahedron Lett. 52, 7135–7137 (2011).

    Article  CAS  Google Scholar 

  18. Krishna, H. & Caruthers, M.H. Alkynyl phosphonate DNA: a versatile “click”able backbone for DNA-based biological applications. J. Am. Chem. Soc. 134, 11618–11631 (2012).

    Article  CAS  Google Scholar 

  19. Tosquellas, G. et al. The pro-oligonucleotide approach: solid phase synthesis and preliminary evaluation of model pro-dodecathymidylates. Nucleic Acids Res. 26, 2069–2074 (1998).

    Article  CAS  Google Scholar 

  20. Périgaud, C. et al. Rational design for cytosolic delivery of nucleoside monophosphates: “SATE” and “DTE” as enzyme-labile transient phosphate protecting groups. Bioorg. Med. Chem. Lett. 3, 2521–2526 (1993).

    Article  Google Scholar 

  21. Lefebvre, I. et al. Mononucleoside phosphotriester derivatives with S-acyl-2-thioethyl bioreversible phosphate-protecting groups: intracellular delivery of 3′-azido-2',3′-dideoxythymidine 5′-monophosphate. J. Med. Chem. 38, 3941–3950 (1995).

    Article  CAS  Google Scholar 

  22. Faraj, A. et al. Intracellular metabolism of beta-L-ddAMP-bis(tbutylSATE), a potent inhibitor of hepatitis B virus replication. Nucleosides Nucleotides 18, 987–988 (1999).

    Article  CAS  Google Scholar 

  23. Bologna, J.C., Morvan, F. & Imbach, J.L. The prooligonucleotide approach: synthesis of mixed phosphodiester and SATE phosphotriester prooligonucleotides using H-Phosphonate and phosphoramidite chemistries. Eur. J. Org. Chem. 9, 2353–2358 (1999).

    Article  Google Scholar 

  24. Guzaev, A.P., Balow, G. & Manoharan, M. Synthesis of chimerical oligonucleotides containing internucleosidic phosphodiester and S-pivaloyl mercaptoethyl phosphotriester linkages. Nucleosides Nucleotides 18, 1391–1392 (1999).

    Article  CAS  Google Scholar 

  25. Peyrottes, S. et al. SATE pronucleotide approaches: an overview. Mini Rev. Med. Chem. 4, 395–408 (2004).

    Article  CAS  Google Scholar 

  26. Breslow, R. & Xu, R. Recognition and catalysis in nucleic acid chemistry. Proc. Natl. Acad. Sci. USA 90, 1201–1207 (1993).

    Article  CAS  Google Scholar 

  27. Beaucage, S.L. Solid-phase synthesis of siRNA oligonucleotides. Curr. Opin. Drug Discov. Devel. 11, 203–216 (2008).

    CAS  PubMed  Google Scholar 

  28. Wadia, J.S., Stan, R.V. & Dowdy, S.F. Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat. Med. 10, 310–315 (2004).

    Article  CAS  Google Scholar 

  29. van den Berg, A. & Dowdy, S.F. Protein transduction domain delivery of therapeutic macromolecules. Curr. Opin. Biotechnol. 22, 888–893 (2011).

    Article  CAS  Google Scholar 

  30. Robbins, M., Judge, A. & MacLachlan, I. siRNA and innate immunity. Oligonucleotides 19, 89–102 (2009).

    Article  CAS  Google Scholar 

  31. Whitehead, K.A., Dahlman, J.E., Langer, R.S. & Anderson, D.G. Silencing or stimulation? siRNA delivery and the immune system. Annu. Rev. Chem. Biomol. Eng. 2, 77–96 (2011).

    Article  CAS  Google Scholar 

  32. Judge, A.D. et al. Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo. Mol. Ther. 13, 494–505 (2006).

    Article  CAS  Google Scholar 

  33. Petersen, S. Self-delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating RNA interference. USPTO 8,691,971 (2014).

  34. Sliedregt, L.J.A.M. et al. Design and synthesis of novel amphiphilic dendritic galactosides for selective targeting of liposomes to the hepatic asialoglycoprotein receptor. J. Med. Chem. 42, 609–618 (1999).

    Article  CAS  Google Scholar 

  35. Gao, S. et al. The effect of chemical modification and nanoparticle formulation on stability and biodistribution of siRNA in mice. Mol. Ther. 17, 1225–1233 (2009).

    Article  CAS  Google Scholar 

  36. Pon, R.T. & Yu, S. Hydroquinone-O,O'-diacetic acid ('Q-linker') as a replacement for succinyl and oxalyl linker arms in solid phase oligonucleotide synthesis. Nucleic Acids Res. 25, 3629–3635 (1997).

    Article  CAS  Google Scholar 

  37. Kuijpers, W.H., Huskens, J., Koole, L.H. & van Boeckel, C.A. Synthesis of well-defined phosphate-methylated DNA fragments: the application of potassium carbonate in methanol as deprotecting reagent. Nucleic Acids Res. 18, 5197–5205 (1990).

    Article  CAS  Google Scholar 

  38. Eguchi, A. et al. Efficient siRNA delivery into primary cells by a peptide transduction domain-dsRNA binding domain fusion protein. Nat. Biotechnol. 27, 567–571 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y. Tor (UCSD) for critical input, Y. Su (UCSD) for help with mass spectrometry, and Z. Rudolph and L.D.F. Vasconcelos for technical assistance. B.R.M., A.S.H. and A.D.S. were supported by a T32 Cancer Biology Training grant (NCI); A.S.H. was also supported by a Blasker Award from The San Diego Foundation; A.P. was supported by a C.I.R.M. Fellowship; C.P.-A. was supported by fellowships from Knut and Alice Wallenberg Foundation and Sweden-America Foundation; J.C.H. was supported by the CT2 training grant (N.C.I.); P.L. was supported by a Swedish Research Council grant; A.D.K. was supported by a IRACDA Training grant (N.I.H.). This work was supported by the W.M. Keck Foundation (S.F.D.), the Department of Defense (S.F.D.), SCOR grant from the Leukemia & Lymphoma Society (S.F.D.), the Pardee Foundation (S.F.D.), a grant from an anonymous donor (S.F.D.) and the Howard Hughes Medical Institute (S.F.D.).

Author information

Authors and Affiliations

Authors

Contributions

B.R.M. designed and performed experiments, analyzed and interpreted data, synthesized RNN oligonucleotides; K.G. designed and synthesized phosphotriesters, phosphoramidites, conjugating approaches, and analyzed and interpreted data; A.S.H. performed in vivo experiments, analyzed and interpreted data, and synthesized RNN oligonucleotides; C.P.-A. and A.D.S. performed experiments, analyzed and interpreted data, and synthesized RNN oligonucleotides; A.v.d.B., J.C.H., A.E., A.D.K. A.P., P.L. and M.K. performed experiments and analyzed data; C.F.D. synthesized RNN oligonucleotides; N.Y. and E.G. helped with data analysis; X.-S.C. synthesized peptides; S.F.D. conceived of siRNN concept, designed experiments, and analyzed and interpreted data. B.R.M., K.G. and S.F.D. wrote the manuscript, and all authors refined the manuscript.

Corresponding author

Correspondence to Steven F Dowdy.

Ethics declarations

Competing interests

The authors declare competing financial interests. S.F.D., B.R.M. and K.G. (UCSD) have filed patents on this work that were licensed by Solstice Biologics, Inc. (San Diego). S.F.D. and B.R.M. are cofounders of Solstice Biologics. S.F.D. is a board director of Solstice Biologics.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–24, Supplementary Table 1 and Supplementary Note (PDF 2376 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meade, B., Gogoi, K., Hamil, A. et al. Efficient delivery of RNAi prodrugs containing reversible charge-neutralizing phosphotriester backbone modifications. Nat Biotechnol 32, 1256–1261 (2014). https://doi.org/10.1038/nbt.3078

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3078

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing