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:

A biomimetic approach for enhancing the in vivo half-life of peptides

Abstract

The tremendous therapeutic potential of peptides has not yet been realized, mainly owing to their short in vivo half-life. Although conjugation to macromolecules has been a mainstay approach for enhancing protein half-life, the steric hindrance of macromolecules often harms the binding of peptides to target receptors, compromising the in vivo efficacy. Here we report a new strategy for enhancing the in vivo half-life of peptides without compromising their potency. Our approach involves endowing peptides with a small molecule that binds reversibly to the serum protein transthyretin. Although there are a few molecules that bind albumin reversibly, we are unaware of designed small molecules that reversibly bind other serum proteins and are used for half-life extension in vivo. We show here that our strategy was effective in enhancing the half-life of an agonist for GnRH receptor while maintaining its binding affinity, which was translated into superior in vivo efficacy.

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: Crystal structure of hTTR bound to AG10, and effect of binding to TTR on the half-life of AG10.
Figure 2: TLHE1 and its peptide conjugates bind selectively to hTTR in buffer and human serum.
Figure 3: Binding to TTR increased the stability of TLHE1–peptides 5, 6 and 7 in vitro, and extended the t1/2 of 7 in rats.
Figure 4: Compound 8 preferentially binds to GnRH-R over hTTR and does not interfere with holo-RBP-TTR interaction.
Figure 5: Compound 8 displayed extended t1/2 and superior efficacy in rats.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Boohaker, R.J., Lee, M.W., Vishnubhotla, P., Perez, J.M. & Khaled, A.R. The use of therapeutic peptides to target and to kill cancer cells. Curr. Med. Chem. 19, 3794–3804 (2012).

    Article  CAS  Google Scholar 

  2. Kaspar, A.A. & Reichert, J.M. Future directions for peptide therapeutics development. Drug Discov. Today 18, 807–817 (2013).

    Article  CAS  Google Scholar 

  3. Tweedle, M.F. Peptide-targeted diagnostics and radiotherapeutics. Acc. Chem. Res. 42, 958–968 (2009).

    Article  CAS  Google Scholar 

  4. Morgat, C. et al. Targeting neuropeptide receptors for cancer imaging and therapy: perspectives with bombesin, neurotensin, and neuropeptide-Y receptors. J. Nucl. Med. 55, 1650–1657 (2014).

    Article  CAS  Google Scholar 

  5. Kontermann, R. in Therapeutic Proteins: Strategies to Modulate Their Plasma Half-lives (Wiley-VCH, Weinheim, 2012).

  6. Gaberc-Porekar, V., Zore, I., Podobnik, B. & Menart, V. Obstacles and pitfalls in the PEGylation of therapeutic proteins. Curr. Opin. Drug Discov. Devel. 11, 242–250 (2008).

    CAS  PubMed  Google Scholar 

  7. Bendele, A., Seely, J., Richey, C., Sennello, G. & Shopp, G. Short communication: renal tubular vacuolation in animals treated with polyethylene-glycol-conjugated proteins. Toxicol. Sci. 42, 152–157 (1998).

    Article  CAS  Google Scholar 

  8. Schellenberger, V. et al. A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat. Biotechnol. 27, 1186–1190 (2009).

    Article  CAS  Google Scholar 

  9. Mitragotri, S., Burke, P.A. & Langer, R. Overcoming the challenges in administering biopharmaceuticals: formulation and delivery strategies. Nat. Rev. Drug Discov. 13, 655–672 (2014).

    Article  CAS  Google Scholar 

  10. Alconcel, S.N., Baas, A.S. & Maynard, H.D. FDA-approved poly(ethylene glycol)–protein conjugate drugs. Polymer Chemistry 2, 1442–1448 (2011).

    Article  CAS  Google Scholar 

  11. Hopp, J. et al. The effects of affinity and valency of an albumin-binding domain (ABD) on the half-life of a single-chain diabody-ABD fusion protein. Protein Eng. Des. Sel. 23, 827–834 (2010).

    Article  CAS  Google Scholar 

  12. Levy, O.E. et al. Novel exenatide analogs with peptidic albumin binding domains: potent anti-diabetic agents with extended duration of action. PLoS ONE 9, e87704 (2014).

    Article  Google Scholar 

  13. Dennis, M.S. et al. Albumin binding as a general strategy for improving the pharmacokinetics of proteins. J. Biol. Chem. 277, 35035–35043 (2002).

    Article  CAS  Google Scholar 

  14. Zobel, K., Koehler, M.F., Beresini, M.H., Caris, L.D. & Combs, D. Phosphate ester serum albumin affinity tags greatly improve peptide half-life in vivo. Bioorg. Med. Chem. Lett. 13, 1513–1515 (2003).

    Article  CAS  Google Scholar 

  15. Trüssel, S. et al. New strategy for the extension of the serum half-life of antibody fragments. Bioconjug. Chem. 20, 2286–2292 (2009).

    Article  Google Scholar 

  16. Ahlskog, J.K., Dumelin, C.E., Trüssel, S., Mårlind, J. & Neri, D. In vivo targeting of tumor-associated carbonic anhydrases using acetazolamide derivatives. Bioorg. Med. Chem. Lett. 19, 4851–4856 (2009).

    Article  CAS  Google Scholar 

  17. Lubberink, M. et al. 110mIn-DTPA-D-Phe1-octreotide for imaging of neuroendocrine tumors with PET. J. Nucl. Med. 43, 1391–1397 (2002).

    CAS  PubMed  Google Scholar 

  18. Engstrøm, T., Barth, T., Melin, P. & Vilhardt, H. Oxytocin receptor binding and uterotonic activity of carbetocin and its metabolites following enzymatic degradation. Eur. J. Pharmacol. 355, 203–210 (1998).

    Article  Google Scholar 

  19. Ingenbleek, Y. & Young, V. Transthyretin (prealbumin) in health and disease: nutritional implications. Annu. Rev. Nutr. 14, 495–533 (1994).

    Article  CAS  Google Scholar 

  20. Johnson, S.M. et al. Native state kinetic stabilization as a strategy to ameliorate protein misfolding diseases: a focus on the transthyretin amyloidoses. Acc. Chem. Res. 38, 911–921 (2005).

    Article  CAS  Google Scholar 

  21. Alhamadsheh, M.M. et al. Potent kinetic stabilizers that prevent transthyretin-mediated cardiomyocyte proteotoxicity. Sci. Transl. Med. 3, 97ra81 (2011).

    Article  Google Scholar 

  22. Penchala, S.C. et al. AG10 inhibits amyloidogenesis and cellular toxicity of the familial amyloid cardiomyopathy-associated V122I transthyretin. Proc. Natl. Acad. Sci. USA 110, 9992–9997 (2013).

    Article  CAS  Google Scholar 

  23. Sundelin, J. et al. The primary structure of rabbit and rat prealbumin and a comparison with the tertiary structure of human prealbumin. J. Biol. Chem. 260, 6481–6487 (1985).

    CAS  PubMed  Google Scholar 

  24. Dickson, P.W., Howlett, G.J. & Schreiber, G. Metabolism of prealbumin in rats and changes induced by acute inflammation. Eur. J. Biochem. 129, 289–293 (1982).

    Article  CAS  Google Scholar 

  25. Mager, D.E. & Jusko, W.J. General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J. Pharmacokinet. Pharmacodyn. 28, 507–532 (2001).

    Article  CAS  Google Scholar 

  26. Choi, S. & Kelly, J.W. A competition assay to identify amyloidogenesis inhibitors by monitoring the fluorescence emitted by the covalent attachment of a stilbene derivative to transthyretin. Bioorg. Med. Chem. 19, 1505–1514 (2011).

    Article  CAS  Google Scholar 

  27. Wegener, D., Wirsching, F., Riester, D. & Schwienhorst, A. A fluorogenic histone deacetylase assay well suited for high-throughput activity screening. Chem. Biol. 10, 61–68 (2003).

    Article  CAS  Google Scholar 

  28. Seminara, S.B., Hayes, F.J. & Crowley, W.F. Gonadotropin-releasing hormone deficiency in the human: pathophysiological and genetic considerations. Endocr. Rev. 19, 521–539 (1998).

    CAS  PubMed  Google Scholar 

  29. Barelli, H. et al. Role of endopeptidase 3.4.24.16 in the catabolism of neurotensin, in vivo, in the vascularly perfused dog ileum. Br. J. Pharmacol. 112, 127–132 (1994).

    Article  CAS  Google Scholar 

  30. Hayden, C. GnRH analogues: applications in assisted reproductive techniques. Eur. J. Endocrinol. 159, S17–S25 (2008).

    Article  CAS  Google Scholar 

  31. Nagy, A. & Schally, A.V. Targeting of cytotoxic luteinizing hormone-releasing hormone analogs to breast, ovarian, endometrial, and prostate cancers. Biol. Reprod. 73, 851–859 (2005).

    Article  CAS  Google Scholar 

  32. Halmos, G., Schally, A.V., Pinski, J., Vadillo-Buenfil, M. & Groot, K. Down-regulation of pituitary receptors for luteinizing hormone-releasing hormone (LH-RH) in rats by LH-RH antagonist Cetrorelix. Proc. Natl. Acad. Sci. USA 93, 2398–2402 (1996).

    Article  CAS  Google Scholar 

  33. Monaco, H.L., Rizzi, M. & Coda, A. Structure of a complex of two plasma proteins: transthyretin and retinol-binding protein. Science 268, 1039–1041 (1995).

    Article  CAS  Google Scholar 

  34. Naylor, H.M. & Newcomer, M.E. The structure of human retinol-binding protein (RBP) with its carrier protein transthyretin reveals an interaction with the carboxy terminus of RBP. Biochemistry 38, 2647–2653 (1999).

    Article  CAS  Google Scholar 

  35. Anderes, K.L. et al. Biological characterization of a novel, orally active small molecule gonadotropin-releasing hormone (GnRH) antagonist using castrated and intact rats. J. Pharmacol. Exp. Ther. 305, 688–695 (2003).

    Article  CAS  Google Scholar 

  36. Mock, E.J., Norton, H.W. & Frankel, A.I. Daily rhythmicity of serum testosterone concentration in the male laboratory rat. Endocrinology 103, 1111–1121 (1978).

    Article  CAS  Google Scholar 

  37. Vickery, B.H. Comparison of the potential for therapeutic utilities with gonadotropin-releasing hormone agonists and antagonists. Endocr. Rev. 7, 115–124 (1986).

    Article  CAS  Google Scholar 

  38. Fenalti, G. et al. Molecular control of δ-opioid receptor signalling. Nature 506, 191–196 (2014).

    Article  CAS  Google Scholar 

  39. Sali, A. & Blundell, T.L. Comparative protein modelling by satisfaction of spatial restraints. J. Mol. Biol. 234, 779–815 (1993).

    Article  CAS  Google Scholar 

  40. Lang, P.T. et al. DOCK 6: combining techniques to model RNA-small molecule complexes. RNA 15, 1219–1230 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by a New Investigator Award from the American Association of Colleges of Pharmacy and the US National Institutes of Health grant 1R15GM110677-01 (M.M.A.). The support by a National Science Foundation Instrumentation grant (NSF-MRI-0722654) is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Contributions

M.M.A. conceived and supervised the project. M.M.A, S.C.P. and M.R.M. designed the experiments. S.C.P., M.R.M. and A.P. performed the chemical synthesis, PK studies and biological assays. H.J. and J.T. performed the modeling study. J.D. and P.B. helped with mass spectrometry analysis. N.R.M., V.S. and A.F. did the NMR spectrometry analysis. J.X. and W.K.C. did the western blot assay and provided advice. M.S.P. designed and analyzed the PK studies. T.C. and J.M. helped with chemical synthesis. M.M.A., S.C.P. and M.R.M. wrote the manuscript, and all authors refined the manuscript.

Corresponding author

Correspondence to Mamoun M Alhamadsheh.

Ethics declarations

Competing interests

The co-authors have filed a provisional patent application related to this work.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Table 1, Supplementary Figures 1–10 and Supplementary Notes 1 and 2 (PDF 3752 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Penchala, S., Miller, M., Pal, A. et al. A biomimetic approach for enhancing the in vivo half-life of peptides. Nat Chem Biol 11, 793–798 (2015). https://doi.org/10.1038/nchembio.1907

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1907

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