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:

Targeted drug delivery through the traceless release of tertiary and heteroaryl amines from antibody–drug conjugates

This article has been updated

Abstract

The reversible attachment of a small-molecule drug to a carrier for targeted delivery can improve pharmacokinetics and the therapeutic index. Previous studies have reported the delivery of molecules that contain primary and secondary amines via an amide or carbamate bond; however, the ability to employ tertiary-amine-containing bioactive molecules has been elusive. Here we describe a bioreversible linkage based on a quaternary ammonium that can be used to connect a broad array of tertiary and heteroaryl amines to a carrier protein. Using a concise, protecting-group-free synthesis we demonstrate the chemoselective modification of 12 complex molecules that contain a range of reactive functional groups. We also show the utility of this connection with both protease-cleavable and reductively cleavable antibody–drug conjugates that were effective and stable in vitro and in vivo. Studies with a tertiary-amine-containing antibiotic show that the resulting antibody–antibiotic conjugate provided appropriate stability and release characteristics and led to an unexpected improvement in activity over the conjugates previously connected via a carbamate.

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: Linker cleavage leads to traceless drug release.
Figure 2: Structures of drugs that show potential linkage sites.
Figure 3: A chemoselective two-step synthesis of a library of quaternary-ammonium-linked anticancer and antibacterial natural products and small molecules.
Figure 4: In vivo stability and efficacy of an antitumour agent conjugated to an antibody.
Figure 5: Synthesis of a disulfide-containing quaternary-ammonium-linked drug conjugate and the mechanism of reductive activation and drug release.
Figure 6: A quaternary-ammonium-linked antibiotic improves efficacy.

Similar content being viewed by others

Change history

  • 03 November 2016

    In the version of this Article originally published, the structure of tubulysin was shown three times in Figure 5 and in each instance the five-membered ring was missing a nitrogen atom. This has been corrected in the online versions of this Article.

References

  1. Pérez-Herrero, E. & Fernández-Medarde, A. Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy. Eur. J. Pharm. Biopharm. 93, 52–79 (2015).

    Article  Google Scholar 

  2. Chari, R. V. J., Miller, M. L. & Widdison, W. C. Antibody–drug conjugates: an emerging concept in cancer therapy. Angew. Chem. Int. Ed. 53, 3796–3827 (2014).

    Article  CAS  Google Scholar 

  3. Kratz, F., Müller, I. A., Ryppa, C. & Warnecke, A. Prodrug strategies in anticancer chemotherapy. ChemMedChem 3, 20–53 (2008).

    Article  CAS  Google Scholar 

  4. Guillemard, V. & Uri Saragovi, H. Prodrug chemotherapeutics bypass P-glycoprotein resistance and kill tumors in vivo with high efficacy and target-dependent selectivity. Oncogene 23, 3613–3621 (2004).

    Article  CAS  Google Scholar 

  5. Dubikovskaya, E. A., Thorne, S. H., Pillow, T. H., Contag, C. H. & Wender, P. A. Overcoming multidrug resistance of small-molecule therapeutics through conjugation with releasable octaarginine transporters. Proc. Natl Acad. Sci. USA 105, 12128–12133 (2008).

    Article  CAS  Google Scholar 

  6. Kirtane, A. R., Kalscheuer, S. M. & Panyam, J. Exploiting nanotechnology to overcome tumor drug resistance: challenges and opportunities. Adv. Drug Deliv. Rev. 65, 1731–1747 (2013).

    Article  CAS  Google Scholar 

  7. Kratz, F., Abu Ajaj, K. & Warnecke, A. Anticancer carrier-linked prodrugs in clinical trials. Expert Opin. Invest. Drugs 16, 1037–1058 (2007).

    Article  CAS  Google Scholar 

  8. Van Bambeke, F., Barcia-Macay, M., Lemaire, S. & Tulkens, P. M. Cellular pharmacodynamics and pharmacokinetics of antibiotics: current views and perspectives. Curr. Opin. Drug Discov. Dev. 9, 218–230 (2006).

    CAS  Google Scholar 

  9. Lehar, S. M. et al. Novel antibody–antibiotic conjugate eliminates intracellular S. aureus. Nature 527, 323–328 (2015).

    Article  CAS  Google Scholar 

  10. Wang, R. E. et al. An immunosuppressive antibody–drug conjugate. J. Am. Chem. Soc. 137, 3229–3232 (2015).

    Article  CAS  Google Scholar 

  11. Dubowchik, G. M. & Firestone, R. A. Cathepsin B-sensitive dipeptide prodrugs. 1. A model study of structural requirements for efficient release of doxorubicin. Bioorg. Med. Chem. Lett. 8, 3341–3346 (1998).

    Article  CAS  Google Scholar 

  12. Carl, P. L., Chakravarty, P. K. & Katzenellenbogen, J. A. A novel connector linkage applicable in prodrug design. J. Med. Chem. 24, 479–480 (1981).

    Article  CAS  Google Scholar 

  13. Doronina, S. O. et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nat. Biotechnol. 21, 778–784 (2003).

    Article  CAS  Google Scholar 

  14. Gromek, S. M. & Balunas, M. J. Natural products as exquisitely potent cytotoxic payloads for antibody–drug conjugates. Curr. Top. Med. Chem. 14, 2822–2834 (2015).

    Article  Google Scholar 

  15. Adem, Y. T. et al. Auristatin antibody–drug conjugate physical instability and the role of drug payload. Bioconjugate Chem. 25, 656–664 (2014).

    Article  CAS  Google Scholar 

  16. Chennamsetty, N., Voynov, V., Kayser, V., Helk, B. & Trout, B. L. Design of therapeutic proteins with enhanced stability. Proc. Natl Acad. Sci. USA 106, 11937–11942 (2009).

    Article  CAS  Google Scholar 

  17. Zhao, R. Y. et al. Synthesis and evaluation of hydrophilic linkers for antibody–maytansinoid conjugates. J. Med. Chem. 54, 3606–3623 (2011).

    Article  CAS  Google Scholar 

  18. Jeffrey, S. C. et al. Design, synthesis, and in vitro evaluation of dipeptide-based antibody minor groove binder conjugates. J. Med. Chem. 48, 1344–1358 (2005).

    Article  CAS  Google Scholar 

  19. Xiao, J., Burn, A. & Tolbert, T. J. Increasing solubility of proteins and peptides by site-specific modification with betaine. Bioconjugate Chem. 19, 1113–1118 (2008).

    Article  CAS  Google Scholar 

  20. Stephanopoulos, N. & Francis, M. B. Choosing an effective protein bioconjugation strategy. Nat. Chem. Biol. 7, 876–884 (2011).

    Article  CAS  Google Scholar 

  21. Agarwal, P. & Bertozzi, C. R. Site-specific antibody–drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjugate Chem. 26, 176–192 (2015).

    Article  CAS  Google Scholar 

  22. Spicer, C. D. & Davis, B. G. Selective chemical protein modification. Nat. Commun. 5, 4740 (2014).

    Article  CAS  Google Scholar 

  23. Russell, G. A. & Danen, W. C. Electron-transfer processes. VIII. Coupling reactions of radicals with carbanions. J. Am. Chem. Soc. 90, 347–353 (1968).

    Article  CAS  Google Scholar 

  24. Stock, L. M. & Wasielewski, M. R. Electron paramagnetic resonance spectra of α-substituted nitrotoluene anion radicals. Influence of electron-withdrawing substituents on the coupling constants for β-hydrogen atoms. J. Am. Chem. Soc. 97, 5620–5622 (1975).

    Article  CAS  Google Scholar 

  25. Tercel, M., Wilson, W. R. & Denny, W. A. Nitrobenzyl mustard quaternary salts: a new class of hypoxia-selective cytotoxins showing very high in vitro selectivity. J. Med. Chem. 36, 2578–2579 (1993).

    Article  CAS  Google Scholar 

  26. Hansch, C., Leo, A. & Taft, R. W. A survey of Hammett substituent constants and resonance and field parameters. Chem. Rev. 91, 165–195 (1991).

    Article  CAS  Google Scholar 

  27. Mohamed, M. M. & Sloane, B. F. Cysteine cathepsins: multifunctional enzymes in cancer. Nat. Rev. Cancer 6, 764–775 (2006).

    Article  CAS  Google Scholar 

  28. Musil, D. et al. The refined 2.15 Å X-ray crystal structure of human liver cathepsin B: the structural basis for its specificity. EMBO J. 10, 2321–2330 (1991).

    Article  CAS  Google Scholar 

  29. Pettit, G. R. et al. The isolation and structure of a remarkable marine animal antineoplastic constituent: dolastatin 10. J. Am. Chem. Soc. 109, 6883–6885 (1987).

    Article  CAS  Google Scholar 

  30. Maderna, A. et al. Discovery of cytotoxic dolastatin 10 analogues with N-terminal modifications. J. Med. Chem. 57, 10527–10543 (2014).

    Article  CAS  Google Scholar 

  31. Steinmetz, H. et al. Isolation, crystal and solution structure determination, and biosynthesis of tubulysins—powerful inhibitors of tubulin polymerization from myxobacteria. Angew. Chem. Int. Ed. 43, 4888–4892 (2004).

    Article  CAS  Google Scholar 

  32. Barker, T. J., Duncan, K. K., Otrubova, K. & Boger, D. L. Potent vinblastine C20′ ureas displaying additionally improved activity against a vinblastine-resistant cancer cell line. ACS Med. Chem. Lett. 4, 985–988 (2013).

    Article  CAS  Google Scholar 

  33. Laguzza, B. C. et al. New antitumor monoclonal antibody–vinca conjugates LY203725 and related compounds: design, preparation, and representative in vivo activity. J. Med. Chem. 32, 548–555 (1989).

    Article  CAS  Google Scholar 

  34. Henry, G. D. De novo synthesis of substituted pyridines. Tetrahedron 60, 6043–6061 (2004).

    Article  CAS  Google Scholar 

  35. Wienecke, A. & Bacher, G. Indibulin, a novel microtubule inhibitor, discriminates between mature neuronal and nonneuronal tubulin. Cancer Res. 69, 171–177 (2009).

    Article  CAS  Google Scholar 

  36. Pauli, J. et al. Suitable labels for molecular imaging—influence of dye structure and hydrophilicity on the spectroscopic properties of IgG conjugates. Bioconjugate Chem. 22, 1298–1308 (2011).

    Article  CAS  Google Scholar 

  37. Flygare, J. A . et al. Peptidomimetic compounds and antibody–drug conjugates thereof. World Intellectual Property Organization patent 2015/095227 A2 (2015).

  38. Jones, L. R., et al. Releasable luciferin-transporter conjugates: tools for the real-time analysis of cellular uptake and release. J. Am. Chem. Soc. 128, 6526–6527 (2006).

    Article  CAS  Google Scholar 

  39. Yang, J., Chen, H., Vlahov, I. R., Cheng, J.-X. & Low, P. S. Evaluation of disulfide reduction during receptor-mediated endocytosis by using FRET imaging. Proc. Natl Acad. Sci. USA 103, 13872–13877 (2006).

    Article  CAS  Google Scholar 

  40. Vlahov, I. R. et al. An assembly concept for the consecutive introduction of unsymmetrical disulfide bonds: synthesis of a releasable multidrug conjugate of folic acid. J. Org. Chem. 72, 5968–5972 (2007).

    Article  CAS  Google Scholar 

  41. Pillow, T. et al. Decoupling stability and release in disulfide bonds with antibody-small molecule conjugates. Chem. Sci. http://dx.doi.org/10.1039/C6SC01831A (2016).

  42. Miles, L. W. C. & Owen, L. N. 149. Dithiols. part XII. The alkaline hydrolysis of acetylated hydroxy-thiols: a new reaction for the formation of cyclic sulphides. J. Chem. Soc. http://dx.doi.org/10.1039/JR9520000817 (1952).

  43. Satyam, A. Design and synthesis of releasable folate–drug conjugates using a novel heterobifunctional disulfide-containing linker. Bioorg. Med. Chem. Lett. 18, 3196–3199 (2008).

    Article  CAS  Google Scholar 

  44. Junutula, J. R. et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26, 925–932 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank F. Fan, Z. Xu, J. Wai, H. Raab, B. Lin, K. Xu and A. Deese for helping in the preparation and analysis of the materials used in the study, and S. Spencer and R. Rowntree for coordinating the studies.

Author information

Authors and Affiliations

Authors

Contributions

L.R.S., S.G.K., X.L., T.W., J.C. and T.H.P. designed and/or synthesized the linker–drug conjugates. S.M.L. designed, executed and analysed data for the in vitro antibiotic experiment. R.V. characterized cathepsin cleavage of linker–drug conjugates and ADCs. D.Z. characterized GSH reduction of the disulfide linker–drug conjugate. J.C. characterized the in vitro potency of cytotoxic ADCs. S.-F.Y. designed and analysed data for in vivo efficacy. C.N. characterized and analysed data for in vivo stability. J.G. characterized the potency of the free cytotoxic drugs. Y.L. confirmed the structure of linker–drug conjugates through NMR studies. A.F.-O. characterized the conjugate stability in whole blood. M.G. performed in vivo efficacy experiments. N.L.S. purified and characterized an antibiotic linker–drug conjugate. B.W. designed a peptidomimetic linker. G.D.L.P, K.X., K.R.K, S.M., J.A.F. and T.H.P. led groups on the project responsible for chemistry, biology and in vitro or in vivo characterization. T.H.P. conceived and initiated the project. T.H.P. wrote the manuscript with help and input from all of the authors.

Corresponding author

Correspondence to Thomas H. Pillow.

Ethics declarations

Competing interests

All authors are full time employees of Genentech or WuXi AppTec.

Supplementary information

Supplementary information

Supplementary information (PDF 6227 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Staben, L., Koenig, S., Lehar, S. et al. Targeted drug delivery through the traceless release of tertiary and heteroaryl amines from antibody–drug conjugates. Nature Chem 8, 1112–1119 (2016). https://doi.org/10.1038/nchem.2635

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchem.2635

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