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:

Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides

Abstract

Whereas stereochemical purity in drugs has become the standard for small molecules, stereoisomeric mixtures containing as many as a half million components persist in antisense oligonucleotide (ASO) therapeutics because it has been feasible neither to separate the individual stereoisomers, nor to synthesize stereochemically pure ASOs. Here we report the development of a scalable synthetic process that yields therapeutic ASOs having high stereochemical and chemical purity. Using this method, we synthesized rationally designed stereopure components of mipomersen, a drug comprising 524,288 stereoisomers. We demonstrate that phosphorothioate (PS) stereochemistry substantially affects the pharmacologic properties of ASOs. We report that Sp-configured PS linkages are stabilized relative to Rp, providing stereochemical protection from pharmacologic inactivation of the drug. Further, we elucidated a triplet stereochemical code in the stereopure ASOs, 3′-SpSpRp, that promotes target RNA cleavage by RNase H1 in vitro and provides a more durable response in mice than stereorandom ASOs.

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

Figure 1: The synthesis cycle of the SOSICS method.
Figure 2: The stereochemical preferences of RNase H1.
Figure 3: Stereochemistry affects ASO pharmacological properties in vitro.
Figure 4: An SSR code promotes RNase HC activity in vitro.
Figure 5: In vivo serum APOB and APOC3 protein kinetics in transgenic mice that were treated with mixtures or stereopure ASOs.

Similar content being viewed by others

References

  1. Crooke, S.T. Molecular mechanisms of action of antisense drugs. Biochim. Biophys. Acta 1489, 31–44 (1999).

    Article  CAS  Google Scholar 

  2. Deleavey, G.F. & Damha, M.J. Designing chemically modified oligonucleotides for targeted gene silencing. Chem. Biol. 19, 937–954 (2012).

    Article  CAS  Google Scholar 

  3. Swayze, E.E. et al. Antisense oligonucleotides containing locked nucleic acid improve potency but cause significant hepatotoxicity in animals. Nucleic Acids Res. 35, 687–700 (2007).

    Article  CAS  Google Scholar 

  4. Sharma, V.K., Sharma, R.K. & Singh, S.K. Antisense oligonucleotides: modifications and clinical trials. MedChemComm. 5, 1454–1471 (2014).

    Article  CAS  Google Scholar 

  5. Eckstein, F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther. 24, 374–387 (2014).

    Article  CAS  Google Scholar 

  6. Crooke, S.T. & Geary, R.S. Clinical pharmacological properties of mipomersen (Kynamro), a second generation antisense inhibitor of apolipoprotein B. Br. J. Clin. Pharmacol. 76, 269–276 (2013).

    Article  CAS  Google Scholar 

  7. Stec, W.J., Grajkowski, A., Koziolkiewicz, M. & Uznanski, B. Novel route to oligo(deoxyribonucleoside phosphorothioates). Stereocontrolled synthesis of P-chiral oligo(deoxyribonucleoside phosphorothioates). Nucleic Acids Res. 19, 5883–5888 (1991).

    Article  CAS  Google Scholar 

  8. Stec, W.J. et al. Deoxyribonucleoside 3′-O-(2-thio- and 2-oxo-“spiro”-4,4-pentamethylene-1,3,2-oxathiaphospholane)s: monomers for stereocontrolled synthesis of oligo(deoxyribonucleoside phosphorothioate)s and chimeric PS/PO Oligonucleotides. J. Am. Chem. Soc. 120, 7156–7167 (1998).

    Article  CAS  Google Scholar 

  9. Iyer, R.P., Yu, D., Ho, N.-H., Tan, W. & Agrawal, S. A novel nucleotide phosphoramidite synthon derived from1R, 2S-ephedrine. Tetrahedron Asymmetry 6, 1051–1054 (1995).

    Article  CAS  Google Scholar 

  10. Guo, M., Yu, D., Iyer, R.P. & Agrawal, S. Solid-phase stereoselective synthesis of 2′-O-methyl-oligoribonucleoside phosphorothioates using nucleoside bicyclic oxazaphospholidines. Bioorg. Med. Chem. Lett. 8, 2539–2544 (1998).

    Article  CAS  Google Scholar 

  11. Wilk, A., Grajkowski, A., Phillips, L.R. & Beaucage, S.L. Deoxyribonucleoside cyclic N-acylphosphoramidites as a new class of monomers for the stereocontrolled synthesis of oligothymidylyl- and oligodeoxycytidylyl- phosphorothioates. J. Am. Chem. Soc. 122, 2149–2156 (2000).

    Article  CAS  Google Scholar 

  12. Oka, N., Yamamoto, M., Sato, T. & Wada, T. Solid-phase synthesis of stereoregular oligodeoxyribonucleoside phosphorothioates using bicyclic oxazaphospholidine derivatives as monomer units. J. Am. Chem. Soc. 130, 16031–16037 (2008).

    Article  CAS  Google Scholar 

  13. Nukaga, Y., Yamada, K., Ogata, T., Oka, N. & Wada, T. Stereocontrolled solid-phase synthesis of phosphorothioate oligoribonucleotides using 2′-O-(2-cyanoethoxymethyl)-nucleoside 3′-O-oxazaphospholidine monomers. J. Org. Chem. 77, 7913–7922 (2012).

    Article  CAS  Google Scholar 

  14. Oka, N., Kondo, T., Fujiwara, S., Maizuru, Y. & Wada, T. Stereocontrolled synthesis of oligoribonucleoside phosphorothioates by an oxazaphospholidine approach. Org. Lett. 11, 967–970 (2009).

    Article  CAS  Google Scholar 

  15. Li, M. et al. Synthesis and cellular activity of stereochemically-pure 2′-O-(2-methoxyethyl)-phosphorothioate oligonucleotides. Chem. Commun. (Camb.) 53, 541–544 (2017).

    Article  CAS  Google Scholar 

  16. Iwamoto, N., Oka, N., Sato, T. & Wada, T. Stereocontrolled solid-phase synthesis of oligonucleoside H-phosphonates by an oxazaphospholidine approach. Angew. Chem. Int. Edn Engl. 48, 496–499 (2009).

    Article  CAS  Google Scholar 

  17. Brill, W.K.-D. Thioalkylation of nucleoside-H-phosphonates and its application to solid phase synthesis of oligonucleotides. Tetrahedr. Lett. 36, 703–706 (1995).

    Article  CAS  Google Scholar 

  18. Nielsen, J. & Caruthers, M.H. Directed Arbuzov-type reactions of 2-cyano-1,1-dimethylethyl deoxynucleoside phosphites. J. Am. Chem. Soc. 110, 6275–6276 (1988).

    Article  CAS  Google Scholar 

  19. Nowotny, M. et al. Structure of human RNase H1 complexed with an RNA/DNA hybrid: insight into HIV reverse transcription. Mol. Cell 28, 264–276 (2007).

    Article  CAS  Google Scholar 

  20. Boczkowska, M., Guga, P. & Stec, W.J. Stereodefined phosphorothioate analogues of DNA: relative thermodynamic stability of the model PS-DNA/DNA and PS-DNA/RNA complexes. Biochemistry 41, 12483–12487 (2002).

    Article  CAS  Google Scholar 

  21. Oka, N. & Wada, T. Stereocontrolled synthesis of oligonucleotide analogs containing chiral internucleotidic phosphorus atoms. Chem. Soc. Rev. 40, 5829–5843 (2011).

    Article  CAS  Google Scholar 

  22. Koziołkiewicz, M. et al. Stability of stereoregular oligo(nucleoside phosphorothioate)s in human plasma: diastereoselectivity of plasma 3′-exonuclease. Antisense Nucleic Acid Drug Dev. 7, 43–48 (1997).

    Article  Google Scholar 

  23. Lima, W.F. et al. Human RNase H1 discriminates between subtle variations in the structure of the heteroduplex substrate. Mol. Pharmacol. 71, 83–91 (2007).

    Article  CAS  Google Scholar 

  24. Koziolkiewicz, M., Krakowiak, A., Kwinkowski, M., Boczkowska, M. & Stec, W.J. Stereodifferentiation--the effect of P chirality of oligo(nucleoside phosphorothioates) on the activity of bacterial RNase H. Nucleic Acids Res. 23, 5000–5005 (1995).

    Article  CAS  Google Scholar 

  25. Yu, D. et al. Stereo-enriched phosphorothioate oligodeoxynucleotides: synthesis, biophysical and biological properties. Bioorg. Med. Chem. 8, 275–284 (2000).

    Article  CAS  Google Scholar 

  26. Watts, J.K. & Corey, D.R. Silencing disease genes in the laboratory and the clinic. J. Pathol. 226, 365–379 (2012).

    Article  CAS  Google Scholar 

  27. Linton, M.F. et al. Transgenic mice expressing high plasma concentrations of human apolipoprotein B100 and lipoprotein(a). J. Clin. Invest. 92, 3029–3037 (1993).

    Article  CAS  Google Scholar 

  28. Prakash, T.P. et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N-acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res. 42, 8796–8807 (2014).

    Article  CAS  Google Scholar 

  29. Reaven, G.M., Mondon, C.E., Chen, Y.D. & Breslow, J.L. Hypertriglyceridemic mice transgenic for the human apolipoprotein C-III gene are neither insulin resistant nor hyperinsulinemic. J. Lipid Res. 35, 820–824 (1994).

    CAS  PubMed  Google Scholar 

  30. Wan, W.B. et al. Synthesis, biophysical properties and biological activity of second generation antisense oligonucleotides containing chiral phosphorothioate linkages. Nucleic Acids Res. 42, 13456–13468 (2014).

    Article  CAS  Google Scholar 

  31. Stec, W.J. et al. Stereodependent inhibition of plasminogen activator inhibitor type 1 by phosphorothioate oligonucleotides: proof of sequence specificity in cell culture and in vivo rat experiments. Antisense Nucleic Acid Drug Dev. 7, 567–573 (1997).

    Article  CAS  Google Scholar 

  32. Krieg, A.M., Guga, P. & Stec, W. P-chirality-dependent immune activation by phosphorothioate CpG oligodeoxynucleotides. Oligonucleotides 13, 491–499 (2003).

    Article  CAS  Google Scholar 

  33. Bumcrot, D., Manoharan, M., Koteliansky, V. & Sah, D.W. RNAi therapeutics: a potential new class of pharmaceutical drugs. Nat. Chem. Biol. 2, 711–719 (2006).

    Article  CAS  Google Scholar 

  34. Jahns, H. et al. Stereochemical bias introduced during RNA synthesis modulates the activity of phosphorothioate siRNAs. Nat. Commun. 6, 6317 (2015).

    Article  CAS  Google Scholar 

  35. Hammond, S.M. & Wood, M.J. Genetic therapies for RNA mis-splicing diseases. Trends Genet. 27, 196–205 (2011).

    Article  CAS  Google Scholar 

  36. Østergaard, M.E. et al. Efficient synthesis and biological evaluation of 5′-GalNAc conjugated antisense oligonucleotides. Bioconjug. Chem. 26, 1451–1455 (2015).

    Article  Google Scholar 

  37. Aaronson, J.G. et al. Rapid HATU-mediated solution phase siRNA conjugation. Bioconjug. Chem. 22, 1723–1728 (2011).

    Article  CAS  Google Scholar 

  38. Koseoglu, M., Hur, A., Atay, A. & Cuhadar, S. Effects of hemolysis interferences on routine biochemistry parameters. Biochem. Med. (Zagreb) 21, 79–85 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Mathieu, D. Boulay, S. Divakaramenon, K. Bowman, V. Vathipadiekal, M. Melkonian, J.C. Dodart, H. Yang, Y. (Benny) Yin, F. Desai and Z. Zhong of Wave Life Sciences for helpful discussions and support for experiments. We thank T. Wada of Tokyo University of Science for general discussions regarding the original oxazaphospholidine method. We thank U. Shigdel of Harvard University (present affiliation, Warp Drive Bio) for purifying the human RNase HC protein. We thank W. Yang of National Institute of Diabetes and Digestive and Kidney Diseases for providing the human RNase HC clone. While at RA Capital Management, A. Donner (present affiliation, The Chemical Probes Portal) provided expert assistance on production of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the designing, planning and/or data collection of this project. N.I. designed and developed the SOSICS platform. N.I., D.C.D.B., I.Z., S.M.S. and G.L. carried out the synthesis and characterization of ASOs and reagents. N.I. and Meena conducted rat liver homogenate stability studies and rat serum stability studies. N.I., S.M., N.S. and Meena conducted RNase HC cleavage studies. N.S., L.H.A., M.F.-K. and J.J.Z. performed in vitro and in vivo biological analyses. N.I. and G.L.V. wrote the manuscript, and all authors refined the manuscript.

Corresponding author

Correspondence to Gregory L Verdine.

Ethics declarations

Competing interests

Wave Life Sciences filed patent applications on this work. G.L.V. is one of the co-founders of Wave Life Sciences and serves as the chairman of the board of directors. D.W.Y.S. was a consultant under contract with Wave Life Sciences. N.I., D.C.D.B., N.S., S.M., I.Z., Meena, S.M.S., G.L., L.H.A., M.F.K., J.J.Z. and C.V. are or were employed by Wave Life Science.

Integrated supplementary information

Supplementary Figure 1 Synthesis of 3’-O-oxazaphospholidine monomers. All oxazaphospholidine monomers were synthesized according to published protocols with minor modifications.

DMTr, 4,4’-dimethoxytrityl; Et3N, triethylamine; Ph, phenyl; THF, tetrahydrofuran

Supplementary Figure 2 Reversed-phase HPLC data for dimers (Sp)-8a-i and (Rp)-8a-i and purified ASOs mipomersen and WV-1-7.

Sp dimers (left column, top to bottom: (Sp)-8a [(Sp)-d(AsT)], (Sp)-8b [(Sp)-d(GsT)], (Sp)-8c [(Sp)-d(5mCsT)], (Sp)-8d [(Sp)-d(CsT)], (Sp)-8e [(Sp)-d(TsT)], (Sp)-8f [(Sp)-A(MOE)sdT], (Sp)-8g [(Sp)-G(MOE)sdT], (Sp)-8h [(Sp)-5mC(MOE)sdT], (Sp)-8i [(Sp)-T(MOE)sdT]) and Rp dimers (center column, top to bottom: (Rp)-8a [(Rp)-d(AsT)], (Rp)-8b [(Rp)-d(GsT)], (Rp)-8c [(Rp)-d(5mCsT)], (Rp)-8d [(Rp)-d(CsT)], (Rp)-8e [(Rp)-d(TsT)], (Rp)-8f [(Rp)-A(MOE)sdT], (Rp)-8g [(Rp)-G(MOE)sdT], (Rp)-8h [(Rp)-5mC(MOE)sdT], (Rp)-8i [(Rp)-T(MOE)sdT]) were eluted using different gradients (see Methods). Dimers were synthesized to demonstrate stereochemical control of synthesis. Absorbance at 254 nm is shown with respect to elution time (min) for each dimer. Purified ASOs (right column, top to bottom: Mipomersen, WV-1, WV-2, WV-3, WB-4, WV-5, WV-6, WV-7), showing purity on C18 column with elution time (x axis, min) and Absorbance at 254 nm (y axis).

Supplementary Figure 3 Diastereomixture and stereochemically pure ASOs show different behaviors on reversed-phase and ion-exchange HPLC.

(a) Ion-exchange HPLC absorbance profiles of mipomersen and WV-1-6. (b) Reverse-phase HPLC data of mousomersen (the mouse sequence of mipomersen, see Supplementary Table 3) and WV-8-11. (c) Ion-exchange HPLC data of mousomersen and WV-8-11. Elution time (x axis) and Absorbance (Abs.) at 254 nm (y axis) are shown. Light green curves show the gradients of percentage of buffer B.

Supplementary Figure 4 Determination of initial velocities (V0) for WV-5 and mipomersen in RNase HC cleavage experiment.

Slope of the best fit line (V0) for WV-5 was 6.90 ± 0.54 μM/min and mipomersen was 2.37 ± 0.20 μM/min under the conditions described in Methods.

Supplementary Figure 5 Dose-response data for mipomersen and ASOs, WV-1-6, in cultured HepB3 cells.

APOB100 mRNA levels were quanitified by RT-PCR and standardized to GAPDH. There is no correlation between potency (IC50) in these assays and efficacy in mice (Fig. 5). Error bars indicate s.d. (n=3), and statistical analysis was performed in Graphpad Prism 6.01.

Supplementary Figure 6 Mipomersen dose-finding study.

Mipomersen was dosed by intraperitoneal injection on days 1, 4, 8, 11, 15, 18, 22, 25, 29, 32, 36, and 39. Human APOB serum protein (shown as a percentage compared with PBS control) dose-dependently decreases in response to mipomersen at days 17 and 24 (n=5 mice per group). We opted for the 10 mg/kg dose in subsequent experiments.

Supplementary Figure 7 Comparative efficacy and duration of action study data.

(a) Human APOB100 serum protein levels (shown as percentage compared with PBS control) decreases in response to 10 mg/kg mipomersen or its components, and recovers in the wash out period (Dosing days: 1, 4, 8, 11, 15, 18, 22, 25, 29, 32; blood collection days 17, 24, 31, 38, 45, 52, 60, 66; n=5 mice per group, error bars represent s.d.). (b) The concentration of WV-5, WV-6 and mipomersen persisting in mouse liver (μg/g) on days 3, 24 and 45 (n=4, error bars represent s.d.) is shown. The concentrations of the ASOs detected at each time point were not statistically significantly different. (c) LC-MS quantification of ASOs in the liver. Calibration curves for mipomersen, WV-5 and WV-6 were generated using the protocol described with a concentration range of 5-500 μg/g. The oligonucleotides were detected using selected reaction monitoring for the following transitions involving loss of phosphate from intact oligonucleotide: m/z 896.5 94.9 for mipomersen, WV-5 and WV-6, and m/z 855.9 94.9 for the internal standard.

Supplementary Figure 8 Chemical structure of WV-12 (top) and WV-13 (bottom).

(a) ASOs are GalNAc-conjugated, 20mer PS oligonucleotides composed of unmodified 10-nucleotide DNA cores and 2′-MOE-modified ends. C is 5MeC in the 2′-MOE modified ends. Complementary RNA strand (human APOC3 mRNA): rArUrArArArGrCrUrGrGrArCrArArGrArArGrCrU. (b) Ion-exchange HPLC data for WV-12 (left) and WV-13 (right). Absorbance at 254 nm is shown with respect to elution time (min) for each oligo.

Supplementary Figure 9 WV-12 (black) and WV-13 (pink) persists in mouse liver on day 78 (n=5, error bars represent s.d.).

The concentrations of the ASOs (μg/g) detected were not statistically significantly different.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iwamoto, N., Butler, D., Svrzikapa, N. et al. Control of phosphorothioate stereochemistry substantially increases the efficacy of antisense oligonucleotides. Nat Biotechnol 35, 845–851 (2017). https://doi.org/10.1038/nbt.3948

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3948

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