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:

Computationally designed antibody–drug conjugates self-assembled via affinity ligands

An Author Correction to this article was published on 03 January 2020

This article has been updated

Abstract

Antibody–drug conjugates (ADCs) combine the high specificity of antibodies with cytotoxic payloads. However, the present strategies for the synthesis of ADCs either yield unstable or heterogeneous products or involve complex processes. Here, we report a computational approach that leverages molecular docking and molecular dynamics simulations to design ADCs that self-assemble through the non-covalent binding of the antibody to a payload that we designed to act as an affinity ligand for specific conserved amino acid residues in the antibody. This method does not require modifications to the antibody structure and yields homogenous ADCs that form in less than 8 min. We show that two conjugates, which consist of hydrophilic and hydrophobic payloads conjugated to two different antibodies, retain the structure and binding properties of the antibody and its biological specificity, are stable in plasma and improve anti-tumour efficacy in mice with non-small cell lung tumour xenografts. The relative simplicity of the approach may facilitate the production of ADCs for the targeted delivery of cytotoxic payloads.

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: Engineering a MAGNET ADC.
Fig. 2: Physicochemical characterization of a MAGNET ADC.
Fig. 3: MAGNET ADCs bind to the target and are stable in human plasma.
Fig. 4: The efficacy of MAGNET ADCs in vitro.
Fig. 5: MAGNET ADCs improve anti-tumour outcomes in vivo.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are available from the corresponding authors on reasonable request.

Change history

  • 03 January 2020

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Beck, A. et al. Strategies and challenges for the next generation of antibody drug conjugates. Nat. Rev. Drug Discov. 16, 315–337 (2017).

    CAS  PubMed  Google Scholar 

  2. Gordon, M. R. et al. Field guide to challenges and opportunities in antibody-drug conjugates for chemists. Bioconjug. Chem. 26, 2198–2215 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Axup, J. Y. et al. Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. Proc. Natl Acad. Sci. USA 109, 16101–16106 (2012).

    CAS  PubMed  Google Scholar 

  4. 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).

    CAS  PubMed  Google Scholar 

  5. Jeger, S. et al. Site-specific and stoichiometric modification of antibodies by bacterial transglutaminase. Angew. Chemie Int. Ed. 49, 9995–9997 (2010).

    CAS  Google Scholar 

  6. Badescu, G. et al. Bridging disulfides for stable and defined antibody drug conjugates. Bioconjug. Chem. 25, 1124–1136 (2014).

    CAS  PubMed  Google Scholar 

  7. Lyon, R. P. et al. Self-hydrolyzing maleimides improve the stability and pharmacological properties of antibody–drug conjugates. Nat. Biotechnol. 32, 1059–1062 (2014).

    CAS  PubMed  Google Scholar 

  8. Hui, J. Z. & Tsourkas, A. Optimization of photoactive protein Z for fast and efficient site-specific conjugation of native IgG. Bioconjug. Chem. 25, 1709–1719 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Jain, N., Smith, S. W., Ghone, S. & Tomczuk, B. Current ADC linker chemistry. Pharm. Res. 32, 3526–3540 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, R., Dowd, V., Stewart, D. J., Burton, S. J. & Lowe, C. R. Design, synthesis, and application of a protein A mimetic. Nat. Biotechnol. 16, 190–195 (1998).

    CAS  PubMed  Google Scholar 

  11. Arakawa, T., Tsumoto, K. & Ejima, D. Alternative downstream processes for production of antibodies and antibody fragments. Biochim. Biophys. Acta 1844, 2032–2040 (2014).

    CAS  PubMed  Google Scholar 

  12. Lyon, R. P. et al. Reducing hydrophobicity of homogeneous antibody–drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33, 733–735 (2015).

    CAS  PubMed  Google Scholar 

  13. Zhang, L. & Sun, Y. Effect of ligand chain length on hydrophobic charge induction chromatography revealed by molecular dynamics simulations. Front. Chem. Sci. Eng. 7, 456–463 (2013).

    CAS  Google Scholar 

  14. Ducry, L. & Stump, B. Antibody-drug conjugates: linking cytotoxic payloads to monoclonal antibodies. Bioconjug. Chem. 21, 5–13 (2010).

    CAS  PubMed  Google Scholar 

  15. Lund, L. N. et al. Novel peptide ligand with high binding capacity for antibody purification. J. Chromatogr. A 1225, 158–167 (2012).

    CAS  PubMed  Google Scholar 

  16. Kabsch, W. & Sander, C. Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical features. Biopolymers 22, 2577–2637 (1983).

    CAS  PubMed  Google Scholar 

  17. Lin, D., Tong, H., Wang, H. & Yao, S. Molecular insight into the ligand–IgG interactions for 4-mercaptoethyl-pyridine based hydrophobic charge-induction chromatography. J. Phys. Chem. B 116, 1393–1400 (2012).

    CAS  PubMed  Google Scholar 

  18. Bak, H. & Thomas, O. R. T. Evaluation of commercial chromatographic adsorbents for the direct capture of polyclonal rabbit antibodies from clarified antiserum. J. Chromatogr. B 848, 116–130 (2007).

    CAS  Google Scholar 

  19. Bronowska, A. in Thermodynamics—Interaction Studies—Solids, Liquids and Gases (InTech, 2011).

  20. Cheng, F., Li, M. M.-Y., Wang, H.-Q. H., Lin, D.-Q. & Qu, J. J.-P. Antibody–ligand interactions for hydrophobic charge-induction chromatography: a surface plasmon resonance study. Langmuir 31, 3422–3430 (2015).

    CAS  PubMed  Google Scholar 

  21. Hamblett, K. J. et al. Effects of drug loading on the antitumour activity of a monoclonal antibody drug conjugate. Clin. Cancer Res. 10, 7063–7070 (2004).

    CAS  PubMed  Google Scholar 

  22. Yuan, X.-M., Lin, D.-Q., Zhang, Q.-L., Gao, D. & Yao, S.-J. A microcalorimetric study of molecular interactions between immunoglobulin G and hydrophobic charge-induction ligand. J. Chromatogr. A 1443, 145–151 (2016).

    CAS  PubMed  Google Scholar 

  23. Storniolo, A. M., Allerheiligen, S. R. & Pearce, H. L. Preclinical, pharmacologic, and phase I studies of gemcitabine. Semin. Oncol. 24, S7-2–S7-7 (1997).

    Google Scholar 

  24. Senter, P. D. & Sievers, E. L. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat. Biotechnol. 30, 631–637 (2012).

    CAS  PubMed  Google Scholar 

  25. Strop., P. et al. Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading. Nat. Biotechnol. 33, 694–696 (2015).

    CAS  PubMed  Google Scholar 

  26. Kruljec, N. & Bratkovič, T. Alternative affinity ligands for immunoglobulins. Bioconjug. Chem. 28, 2009–2030 (2017).

    CAS  PubMed  Google Scholar 

  27. Yang, H. et al. Binding site on human immunoglobulin G for the affinity ligand HWRGWV. J. Mol. Recognit. 23, 271–282 (2009).

    Google Scholar 

  28. DeLano, W. L., Ultsch, M. H., de Vos, A. M. & Wells, J. A. Convergent solutions to binding at a protein-protein interface. Science 287, 1279–1283 (2000).

    CAS  PubMed  Google Scholar 

  29. Frisch, M. J. et al. Gaussian 09, Revision A. 02 https://gaussian.com/ (Gaussian Inc, 2009).

  30. Becke, A. D. Density‐functional thermochemistry III: the role of exact exchange. J. Chem. Phys. 98, 5648–5652 (1993).

    CAS  Google Scholar 

  31. Vosko, S. H., Wilk, L. & Nusair, M. Accurate spin-dependent electron liquid correlation energies for local spin density calculations: a critical analysis. Can. J. Phys. 58, 1200–1211 (1980).

    CAS  Google Scholar 

  32. Francl, M. M. et al. Self‐consistent molecular orbital methods XXIII: a polarization‐type basis set for second‐row elements. J. Chem. Phys. 77, 3654–3665 (1982).

    CAS  Google Scholar 

  33. Binning, R. C. & Curtiss, L. A. Compact contracted basis sets for third-row atoms: Ga-Kr. J. Comput. Chem. 11, 1206–1216 (1990).

    CAS  Google Scholar 

  34. Vanommeslaeghe, K. et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J. Comput. Chem. 31, 671–690 (2009).

    Google Scholar 

  35. Singh, U. C. & Kollman, P. A. An approach to computing electrostatic charges for molecules. J. Comput. Chem. 5, 129–145 (1984).

    CAS  Google Scholar 

  36. Hess, B., Kutzner, C., van der Spoel, D. & Lindahl, E. GROMACS 4: algorithms for highly efficient, load-balanced, and scalable molecular simulation. J. Chem. Theory Comput. 4, 435–447 (2008).

    CAS  PubMed  Google Scholar 

  37. Deisenhofer, J. Crystallographic refinement and atomic models of a human Fc fragment and its complex with fragment B of protein A from Staphylococcus aureus at 2.9- and 2.8-Å resolution. Biochemistry 20, 2361–2370 (1981).

    CAS  PubMed  Google Scholar 

  38. Donaldson, J. M. et al. Identification and grafting of a unique peptide-binding site in the Fab framework of monoclonal antibodies. Proc. Natl Acad. Sci. USA 110, 17456–17461 (2013).

    CAS  PubMed  Google Scholar 

  39. Li, S. et al. Structural basis for inhibition of the epidermal growth factor receptor by cetuximab. Cancer Cell 7, 301–311 (2005).

    CAS  PubMed  Google Scholar 

  40. Sanner, M. F. Python: a programming language for software integration and development. J. Mol. Graph. Model. 17, 57–61 (1999).

    CAS  PubMed  Google Scholar 

  41. Gasteiger, J. & Marsili, M. A new model for calculating atomic charges in molecules. Tetrahedron Lett. 19, 3181–3184 (1978).

    Google Scholar 

  42. Trott, O. & Olson, A. J. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455–461 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Price, D. J. & Brooks, C. L. A modified TIP3P water potential for simulation with Ewald summation. J. Chem. Phys. 121, 10096–10103 (2004).

    CAS  PubMed  Google Scholar 

  44. Huang, J. & MacKerell, A. D. CHARMM36 all-atom additive protein force field: Validation based on comparison to NMR data. J. Comput. Chem. 34, 2135–2145 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hess, B., Bekker, H., Berendsen, H. J. C. & Fraaije, J. G. E. M. LINCS: A linear constraint solver for molecular simulations. J. Comput. Chem. 18, 1463–1472 (1997).

    CAS  Google Scholar 

  46. Essmann, U. et al. A smooth particle mesh Ewald method. J. Chem. Phys. 103, 8577–8593 (1995).

    CAS  Google Scholar 

  47. Verlet, L. Computer ‘Experiments’ on Classical Fluids. I. Thermodynamical Properties of Lennard-Jones Molecules. Phys. Rev. 159, 98–103 (1967).

    CAS  Google Scholar 

  48. Parrinello, M. & Rahman, A. Polymorphic transitions in single crystals: A new molecular dynamics method. J. Appl. Phys. 52, 7182–7190 (1981).

    CAS  Google Scholar 

  49. Bussi, G., Donadio, D. & Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 126, 14101 (2007).

    Google Scholar 

  50. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    CAS  PubMed  Google Scholar 

  51. Torrie, G. M. & Valleau, J. P. Nonphysical sampling distributions in Monte Carlo free-energy estimation: umbrella sampling. J. Comput. Phys. 23, 187–199 (1977).

    Google Scholar 

  52. Souaille, M. & Roux, B. Extension to the weighted histogram analysis method: combining umbrella sampling with free energy calculations. Comput. Phys. Commun. 135, 40–57 (2001).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank staff at the Advanced Instrumentation Research Facility at Jawaharlal Nehru University, Delhi and DBT grant (no. BT/PR3130/INF/22/139/2011) for use of their confocal microscopy facility.

Author information

Authors and Affiliations

Authors

Contributions

N.G. conceived the idea, led the overall study, prepared and characterized the conjugates. G.B. and N.G. designed the linkers. A.A. synthesized and characterized the linkers and P.K.D. performed initial optimizations under supervision of G.B., A.Sarkar and S.K.M. G.V.D. performed all of the in silico research under the supervision of S.R. M.C. performed the ITC experiments under the supervision of V.R. and M.M. R.G. purified the linkers by HPLC. J.S., S.K. and S.C. performed all of the biological studies under supervision of A.Sengupta. M.S. analysed the PK data. M.R. and S.S. supervised all of the studies. N.G., M.R. and S.S. wrote the manuscript with input from all of the authors.

Corresponding authors

Correspondence to Nimish Gupta or Shiladitya Sengupta.

Ethics declarations

Competing interests

N.G., A.Sarkar, A.Sengupta and M.R. are employees of Akamara Therapeutics and own equity. S.S. is a cofounder and board member of Akamara Therapeutics and owns equity in Akamara Therapeutics. N.G., S.S. and M.R. are listed as inventors on a patent on this technology (US Patent App. 15/124,058; WO2015148126A1).

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 figures 1–27 and tables 1–7.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gupta, N., Ansari, A., Dhoke, G.V. et al. Computationally designed antibody–drug conjugates self-assembled via affinity ligands. Nat Biomed Eng 3, 917–929 (2019). https://doi.org/10.1038/s41551-019-0470-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-019-0470-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer