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:

In-solution enrichment identifies peptide inhibitors of protein–protein interactions

An Author Correction to this article was published on 13 May 2019

This article has been updated

Abstract

The use of competitive inhibitors to disrupt protein–protein interactions (PPIs) holds great promise for the treatment of disease. However, the discovery of high-affinity inhibitors can be a challenge. Here we report a platform for improving the affinity of peptide-based PPI inhibitors using non-canonical amino acids. The platform utilizes size exclusion-based enrichment from pools of synthetic peptides (1.5–4 kDa) and liquid chromatography-tandem mass spectrometry-based peptide sequencing to identify high-affinity binders to protein targets, without the need for ‘reporter’ or ‘encoding’ tags. Using this approach—which is inherently selective for high-affinity binders—we realized gains in affinity of up to ~100- or ~30-fold for binders to the oncogenic ubiquitin ligase MDM2 or HIV capsid protein C-terminal domain, which inhibit MDM2–p53 interaction or HIV capsid protein C-terminal domain dimerization, respectively. Subsequent macrocyclization of select MDM2 inhibitors rendered them cell permeable and cytotoxic toward cancer cells, demonstrating the utility of the identified compounds as functional PPI inhibitors.

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: Affinity selection platform for the maturation of known peptide binders.
Fig. 2: Affinity selection identifies hotspot residues for MDM2 binding.
Fig. 3: Affinity selection identifies potent variants containing non-canonical amino acids.
Fig. 4: Discovery of potent mirror image knottin derived peptide binders to MDM2.
Fig. 5: Macrocyclic variants were cell penetrating and active against oncogenic cells overexpressing MDM2.

Similar content being viewed by others

Data availability

The authors declare that all data supporting the findings of this study are available within the manuscript, its Supplementary Information, and Supplementary Notes.

Change history

  • 13 May 2019

    In the version of this article originally published, the peptide sequences of compounds 90, 92 and 93 in Fig. 5b and Supplementary Table 7 contained several errors. In Fig. 5b, position 6 of compound 90 should be Tyr instead of Phe. In both Fig. 5b and Supplementary Table 7, position 9 of compounds 92 and 93 should be Gln instead of Glu. Additionally, the surname of co-author Anupam Bandyopadhyay was incorrectly spelled as Bandyopdhyay. The errors have been corrected in the HTML and PDF versions of the paper and in the Supplementary Information PDF.

References

  1. Wells, J. A. & McClendon, C. L. Reaching for high-hanging fruit in drug discovery at protein–protein interfaces. Nature 450, 1001–1009 (2007).

    Article  CAS  Google Scholar 

  2. Petta, I., Lievens, S., Libert, C., Tavernier, J. & De Bosscher, K. Modulation of protein–protein interactions for the development of novel therapeutics. Mol. Ther. 24, 707–718 (2016).

    Article  CAS  Google Scholar 

  3. Modell, A. E., Blosser, S. L. & Arora, P. S. Systematic targeting of protein–protein interactions. Trends Pharmacol. Sci. 37, 702–713 (2016).

    Article  CAS  Google Scholar 

  4. Pelay-Gimeno, M., Glas, A., Koch, O. & Grossmann, T. N. Structure-based design of inhibitors of protein–protein interactions: mimicking peptide binding epitopes. Angew. Chem. Int. Ed. Engl. 54, 8896–8927 (2015).

  5. Valeur, E. et al. New modalities for challenging targets in drug discovery. Angew. Chem. Int. Ed. Engl. 56, 10294–10323 (2017).

    Article  CAS  Google Scholar 

  6. Grossmann, T. N. et al. Inhibition of oncogenic Wnt signaling through direct targeting of β-catenin. Proc. Natl Acad. Sci. USA 109, 17942–17947 (2012).

    Article  CAS  Google Scholar 

  7. Spokoyny, A. M. et al. A perfluoroaryl-cysteine SNAr chemistry approach to unprotected peptide stapling. J. Am. Chem. Soc. 135, 5946–5949 (2013).

    Article  CAS  Google Scholar 

  8. Lautrette, G., Touti, F., Lee, H. G., Dai, P. & Pentelute, B. L. Nitrogen arylation for macrocyclization of unprotected peptides. J. Am. Chem. Soc. 138, 8340–8343 (2016).

    Article  CAS  Google Scholar 

  9. Renfrew, P. D., Choi, E. J., Bonneau, R. & Kuhlman, B. Incorporation of noncanonical amino acids into Rosetta and use in computational protein–peptide interface design. PLoS One 7, 1–15 (2012).

    Article  Google Scholar 

  10. Drew, K. et al. Adding diverse noncanonical backbones to Rosetta: enabling peptidomimetic design. PLoS One 8, 1–17 (2013).

    Google Scholar 

  11. Rognan, D., Scapozza, L. & Folkers, G. & Daser A. Rational design of nonnatural peptides as high-affinity ligands for the HLA-B*2705 human leukocyte antigen. Proc. Natl Acad. Sci. USA 92, 753–757 (1995)..

  12. Zhan, C. et al. An ultrahigh affinity d-peptide antagonist Of MDM2. J. Med. Chem. 55, 6237–6241 (2012).

    Article  CAS  Google Scholar 

  13. Zhou, H.-B. et al. Structure-based design of high-affinity macrocyclic peptidomimetics to block the menin-MLL1 protein–protein interaction. J. Med. Chem. 1, 1113–1123 (2012).

    Google Scholar 

  14. Kritzer, J. A., Luedtke, N. W., Harker, E. A. & Schepartz, A. A rapid library screen for tailoring β-peptide structure and function. J. Am. Chem. Soc. 127, 14584–14585 (2005).

    Article  CAS  Google Scholar 

  15. Upadhyaya, P. et al. Inhibition of Ras signaling by blocking Ras-effector interactions with cyclic peptides. Angew. Chem. Int. Ed. Engl. 54, 7602–7606 (2015).

  16. Annis, D. A., Nazef, N., Chuang, C. C., Scott, M. P. & Nash, H. M. A general technique to rank protein–ligand binding affinities and determine allosteric versus direct binding site competition in compound mixtures. J. Am. Chem. Soc. 126, 15495–15503 (2004).

    Article  CAS  Google Scholar 

  17. Zuckermann, R. N., Kerr, J. M., Siani, M. A., Banville, S. C. & Santi, D. V. Identification of highest-affinity ligands by affinity selection from equimolar peptide mixtures generated by robotic synthesis. Proc. Natl Acad. Sci. USA 89, 4505–4509 (1992).

    Article  CAS  Google Scholar 

  18. Annis, D. A., Nickbarg, E., Yang, X., Ziebell, M. R. & Whitehurst, C. E. Affinity selection-mass spectrometry screening techniques for small molecule drug discovery. Curr. Opin. Chem. Biol. 11, 518–526 (2007).

    Article  CAS  Google Scholar 

  19. Comess, K. M. et al. An ultraefficient affinity-based high-throughout screening process: application to bacterial cell wall biosynthesis enzyme MurF. J. Biomol. Screen. 11, 743–754 (2006).

    Article  CAS  Google Scholar 

  20. Dunayevskiy, Y. M., Lai, J.-J., Quinn, C., Talley, F. & Vouros, P. Mass spectrometric identification of ligands selected from combinatorial libraries using gel filtration. Rapid Commun. Mass Spectrom. 11, 1178–1184 (1997).

    Article  CAS  Google Scholar 

  21. Huyer, G. et al. Affinity selection from peptide libraries to determine substrate specificity of protein tyrosine phosphatases. Anal. Biochem. 258, 19–30 (1998).

    Article  CAS  Google Scholar 

  22. Vinogradov, A. A. et al. Library design-facilitated high-throughput sequencing of synthetic peptide libraries. ACS Comb. Sci. 19, 694–701 (2017).

    Article  CAS  Google Scholar 

  23. Muckenschnabel, I., Falchetto, R., Mayr, L. M. & Filipuzzi, I. SpeedScreen: label-free liquid chromatography-mass spectrometry-based high-throughput screening for the discovery of orphan protein ligands. Anal. Biochem. 324, 241–249 (2004).

    Article  CAS  Google Scholar 

  24. O’Connell, T. N., Ramsay, J., Rieth, S. F., Shapiro, M. J. & Stroh, J. G. Solution-based indirect affinity selection mass spectrometry—a general tool for high-throughput screening of pharmaceutical compound libraries. Anal. Chem. 86, 7413–7420 (2014).

    Article  Google Scholar 

  25. Kussie, P. H. et al. Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain. Science 274, 948–953 (1996).

    Article  CAS  Google Scholar 

  26. Vassilev, L. T. et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844–848 (2004).

    Article  CAS  Google Scholar 

  27. Li, C. et al. Systematic mutational analysis of peptide inhibition of the p53–MDM2/MDMX interactions. J. Mol. Biol. 398, 200–213 (2010).

    Article  CAS  Google Scholar 

  28. Böttger, A. et al. Molecular characterization of the hdm2–p53 interaction. J. Mol. Biol. 269, 744–756 (1997).

    Article  Google Scholar 

  29. Phan, J. et al. Structure-based design of high affinity peptides inhibiting the interaction of p53 with MDM2 and MDMX. J. Biol. Chem. 285, 2174–2183 (2010).

    Article  CAS  Google Scholar 

  30. Pazgier, M. et al. Structural basis for high-affinity peptide inhibition of p53 interactions with MDM2 and MDMX. Proc. Natl Acad. Sci. USA 106, 4665–4670 (2009).

    Article  CAS  Google Scholar 

  31. Liu, R., Li, X. & Lam, K. S. Combinatorial chemistry in drug discovery. Curr. Opin. Chem. Biol. 38, 117–126 (2017).

    Article  CAS  Google Scholar 

  32. Ferrer, M. et al. Selection of gp41-mediated HIV-1 cell entry inhibitors from biased combinatorial libraries of non-natural binding elements. Nat. Struct. Biol. 6, 953–960 (1999).

    Article  CAS  Google Scholar 

  33. Wang, X., Peng, L., Liu, R., Xu, B. & Lam, K. S. Applications of topologically segregated bilayer beads in combinatorial libraries. J. Pept. Res. 65, 130–138 (2005).

    Article  CAS  Google Scholar 

  34. Lam, K. S. et al. A new type of synthetic peptide library for identifying ligand-binding activity. Nature 354, 82–84 (1991).

    Article  CAS  Google Scholar 

  35. Lam, K. S. & Lebl, M. Selectide technology: bead-binding screening. Methods 6, 372–380 (1994).

    Article  CAS  Google Scholar 

  36. Chen, C. L., Strop, P., Lebl, M. & Lam, K. S. Synthesis of libraries for bead-binding screening. Methods Enzymol. 267, 211–219 (1996).

    Article  CAS  Google Scholar 

  37. Alluri, P. G., Reddy, M. M., Bachhawat-Sikder, K., Olivos, H. J. & Kodadek, T. Isolation of protein ligands from large peptoid libraries. J. Am. Chem. Soc. 125, 13995–14004 (2003).

    Article  CAS  Google Scholar 

  38. Reddy, M. M., Bachhawat-Sikder, K. & Kodadek, T. Transformation of low-affinity lead compounds into high-affinity protein capture agents. Chem. Biol. 11, 1127–1137 (2004).

    Article  CAS  Google Scholar 

  39. Zondlo, S. C., Lee, A. E. & Zondlo, N. J. Determinants of specificity of MDM2 for the activation domains of p53 and p65: Proline27 disrupts the MDM2-binding motif of p53. Biochemistry 45, 11945–11957 (2006).

    Article  Google Scholar 

  40. Furman, J. L., Chiu, M. & Hunter, M. J. Early engineering approaches to improve peptide developability and manufacturability. AAPS J. 17, 111–120 (2015).

    Article  CAS  Google Scholar 

  41. Chang, Y. S. et al. Stapled α-helical peptide drug development: a potent dual inhibitor of MDM2 and MDMX for p53-dependent cancer therapy. Proc. Natl Acad. Sci. USA 110, E3445–E3454 (2013).

    Article  CAS  Google Scholar 

  42. Adamson, C. S. & Freed, E. O. Anti-HIV-1 therapeutics: from FDA-approved drugs to hypothetical future targets. Mol. Interv. 9, 70–74 (2009).

    Article  CAS  Google Scholar 

  43. Bartonova, V. et al. Residues in the HIV-1 capsid assembly inhibitor binding site are essential for maintaining the assembly-competent quaternary structure of the capsid protein. J. Biol. Chem. 283, 32024–32033 (2008).

    Article  CAS  Google Scholar 

  44. Ternois, F., Sticht, J., Duquerroy, S., Kräusslich, H. G. & Rey, F. A. The HIV-1 capsid protein C-terminal domain in complex with a virus assembly inhibitor. Nat. Struct. Mol. Biol. 12, 678–682 (2005).

    Article  CAS  Google Scholar 

  45. Heitz, a, Le-Nguyen, D. & Chiche, L. Min-21 and min-23, the smallest peptides that fold like a cystine-stabilized β-sheet motif: design, solution structure, and thermal stability. Biochemistry 38, 10615–10625 (1999).

    Article  CAS  Google Scholar 

  46. Celie, P. H. N. et al. UV-induced ligand exchange in MHC class I protein crystals. J. Am. Chem. Soc. 131, 12298–12304 (2009).

    Article  CAS  Google Scholar 

  47. Zhang, H. et al. A cell-penetrating helical peptide as a potential HIV-1 inhibitor. J. Mol. Biol. 378, 565–580 (2008).

    Article  CAS  Google Scholar 

  48. Wachter, F. et al. Mechanistic validation of a clinical lead stapled peptide that reactivates p53 by dual HDM2 and HDMX targeting. Oncogene 36, 2184–2190 (2017).

    Article  CAS  Google Scholar 

  49. Goodnow, R. A., Dumelin, C. E. & Keefe, A. D. DNA-encoded chemistry: enabling the deeper sampling of chemical space. Nat. Rev. Drug Discov. 16, 131–147 (2017).

    Article  CAS  Google Scholar 

  50. Rabideau, A. E., Liao, X. & Pentelute, B. L. Delivery of mirror image polypeptides into cells. Chem. Sci. 6, 648–653 (2015).

    Article  CAS  Google Scholar 

  51. Vinogradov, A. A., Choo, Z. N., Totaro, K. A. & Pentelute, B. L. Macrocyclization of unprotected peptide isocyanates. Org. Lett. 18, 1226–1229 (2016).

    Article  CAS  Google Scholar 

  52. Rodenko, B. et al. Class I major histocompatibility complexes loaded by a periodate trigger. J. Am. Chem. Soc. 131, 12305–12313 (2009).

    Article  CAS  Google Scholar 

  53. Mijalis, A. J. et al. A fully automated flow-based approach for accelerated peptide synthesis. Nat. Chem. Biol. 13, 464–466 (2017).

    Article  CAS  Google Scholar 

  54. Kim, Y.-W., Grossmann, T. N. & Verdine, G. L. Synthesis of all-hydrocarbon stapled α-helical peptides by ring-closing olefin metathesis. Nat. Protoc. 6, 761 (2011).

    Article  CAS  Google Scholar 

  55. Illien, F. et al. Quantitative fluorescence spectroscopy and flow cytometry analyses of cell-penetrating peptides internalization pathways: optimization, pitfalls, comparison with mass spectrometry quantification. Sci. Rep. 6, 1–13 (2016).

    Article  Google Scholar 

Download references

Acknowledgements

The Bettencourt Schueller Foundation is gratefully acknowledged for postdoctoral support to F.T. The Human Frontier Science Program Organization is thanked for a cross-disciplinary fellowship (LT000745/2014-C) to F.T. This work was supported in part by Servier, the Defense Advanced Research Projects Agency (award no. 023504-001 to B.L.P.), the NIH National Institute of General Medical Sciences (grant no. 5-R01-GM110535 to B.L.P.), a Bristol-Myers Squibb unrestricted grant in Synthetic Organic Chemistry (B.L.P.), and a Novartis Early Career Award (B.L.P). F. Lefoulon (Servier) is thankfully acknowledged for his suggestions and comments on the manuscript. FACS experiments were performed at the MIT Koch Institute Flow Cytometry Core. The authors thank the Biophysical Instrumentation Facility at MIT for providing access to the Octet Bio-Layer Interferometry System (NIH S10OD016326) and D. Pheasant for her technical assistance and B. Dass (Pall Fortebio) for his help with data analysis. We gratefully acknowledge A. Rabideau for peptide synthesis and characterization training, A. Quartararo for help with the Orbitrap LC–MS instrument, and A. Vinogradov and M. Simon for scientific discussions.

Author information

Authors and Affiliations

Authors

Contributions

F.T. and B.L.P. conceived the study with input from Z.P.G. F.T. and B.L.P. directed the project and designed experiments. F.T. performed most experiments. Z.P.G. and A.B. contributed to OBOC screen design and experiments, and G.L. contributed to library purification and binder validation. F.T., Z.P.G., and B.L.P. wrote the manuscript. All authors contributed to the analysis, interpretation, and validation of the data.

Corresponding authors

Correspondence to Fayçal Touti or Bradley L. Pentelute.

Ethics declarations

Competing interests

A patent application covering this work has been filed by MIT-TLO.

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 Tables 1–7, Supplementary Figures 1–39

Reporting Summary

Supplementary Note 1

Library 1 binder validation traces (Fig. 2 and Supplementary Fig. 13).

Supplementary Note 2

HPSEC and LC–MS traces for model binders; LC–MS characterization of numbered compounds.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Touti, F., Gates, Z.P., Bandyopadhyay, A. et al. In-solution enrichment identifies peptide inhibitors of protein–protein interactions. Nat Chem Biol 15, 410–418 (2019). https://doi.org/10.1038/s41589-019-0245-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0245-2

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