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.

A brush-polymer/exendin-4 conjugate reduces blood glucose levels for up to five days and eliminates poly(ethylene glycol) antigenicity

Abstract

The delivery of therapeutic peptides and proteins is often challenged by short half-lives and the consequent need for frequent injections that limit efficacy, reduce patient compliance and increase treatment cost. Here, we demonstrate that a single subcutaneous injection of site-specific (C-terminal) conjugates of exendin-4 (exendin)—a therapeutic peptide that is clinically used to treat type 2 diabetes mellitus—and poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) with precisely controlled molecular weights lowered blood glucose for up to 120 h in fed mice. Most notably, we show that an exendin-C-POEGMA conjugate with an average of nine side-chain ethylene glycol (EG) repeats exhibits significantly lower reactivity towards patient-derived anti-poly(ethylene glycol) (anti-PEG) antibodies than two US FDA-approved PEGylated drugs, and that reducing the side-chain length to three EG repeats completely eliminates PEG antigenicity without compro­mising in vivo efficacy. Our findings establish the site-specific conjugation of POEGMA as a next-generation PEGylation technology for improving the pharmacological performance of traditional PEGylated drugs, whose safety and efficacy are hindered by pre-existing anti-PEG antibodies in patients.

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: Synthesis of exendin-C-POEGMA.
Figure 2: Characterization of the exendin-C-Br macroinitiator and EG9 exendin-C-POEGMA conjugates.
Figure 3: Assessment of the Mw -dependent in vivo efficacy of EG9 exendin-C-POEGMA conjugates.
Figure 4: IPGTT for an EG9 exendin-C-POEGMA conjugate in mice.
Figure 5: Assessment of the reactivity of exendin-C-POEGMA conjugates towards anti-PEG antibodies in patient plasma samples.
Figure 6: Assessment of the in vivo efficacy and pharmacokinetics of exendin-C-POEGMA conjugates.

Similar content being viewed by others

References

  1. Leader, B., Baca, Q. J. & Golan, D. E. Protein therapeutics: a summary and pharmacological classification. Nat. Rev. Drug Discov. 7, 21–39 (2008).

    Article  Google Scholar 

  2. Craik, D. J., Fairlie, D. P., Liras, S. & Price, D. The future of peptide-based drugs. Chem. Biol. Drug Des. 81, 136–147 (2013).

    Article  Google Scholar 

  3. Peters, T. J. Serum albumin. Adv. Protein Chem. 37, 161–245 (1985).

    Article  Google Scholar 

  4. Awai, M. & Brown, E. B. Studies of the metabolism of I-131-labeled human transferrin. J. Lab. Clin. Med. 61, 363–396 (1963).

    Google Scholar 

  5. Banga, A. K. Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems (CRC, 2015).

    Book  Google Scholar 

  6. Caliceti, P. & Veronese, F. M. Pharmacokinetic and biodistribution properties of poly(ethylene glycol)-protein conjugates. Adv. Drug Deliv. Rev. 55, 1261–1277 (2003).

    Article  Google Scholar 

  7. Malik, D. K., Baboota, S., Ahuja, A., Hasan, S. & Ali, J. Recent advances in protein and peptide drug delivery systems. Curr. Drug Deliv. 2, 141–151 (2007).

    Article  Google Scholar 

  8. Werle, M. & Bernkop-Schnurch, A. Strategies to improve plasma half life time of peptide and protein drugs. Amino Acids 30, 351–367 (2006).

    Article  Google Scholar 

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

    Article  Google Scholar 

  10. Nucci, M. L., Shorr, R. G. & Abuchowski, A. The therapeutic value of poly(ethylene glycol)-modified proteins. Adv. Drug Deliv. Rev. 6, 133–151 (1991).

    Article  Google Scholar 

  11. Youn, Y. S., Na, D. H. & Lee, K. C. High-yield production of biologically active mono-PEGylated salmon calcitonin by site-specific PEGylation. J. Control. Release 117, 371–379 (2007).

    Article  Google Scholar 

  12. Gauthier, M. A. & Klok, H. Peptide/protein–polymer conjugates: synthetic strategies and design concepts. Chem. Commun. 2591–2611 (2008).

  13. Qi, Y. & Chilkoti, A. Growing polymers from peptides and proteins: a biomedical perspective. Polym. Chem. 5, 266–276 (2014).

    Article  Google Scholar 

  14. 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).

    Google Scholar 

  15. Veronese, F. M. Peptide and protein PEGylation: a review of problems and solutions. Biomaterials 22, 405–417 (2001).

    Article  Google Scholar 

  16. Ganson, N. J., Kelly, S. J., Scarlett, E., Sundy, J. S. & Hershfield, M. S. Control of hyperuricemia in subjects with refractory gout, and induction of antibody against poly(ethylene glycol) (PEG), in a phase I trial of subcutaneous PEGylated urate oxidase. Arthritis Res. Ther. 8, R12–R22 (2006).

    Article  Google Scholar 

  17. Hershfield, M. S. et al. Induced and pre-existing anti-polyethylene glycol antibody in a trial of every 3-week dosing of pegloticase for refractory gout, including in organ transplant recipients. Arthritis Res. Ther. 16, R63 (2014).

    Article  Google Scholar 

  18. Armstrong, J. K. et al. Antibody against poly(ethylene glycol) adversely affects PEG-asparaginase therapy in acute lymphoblastic leukemia patients. Cancer 110, 103–111 (2007).

    Article  Google Scholar 

  19. Richter, A. W. & Akerblom, E. Polyethylene glycol reactive antibodies in man: titer distribution in allergic patients treated with monomethoxy polyethylene glycol modified allergens or placebo, and in healthy blood donors. Int. Arch. Allergy Appl. Immunol. 74, 36–39 (1984).

    Article  Google Scholar 

  20. Garay, R. P., El-Gewely, R., Armstrong, J. K., Garratty, G. & Richette, P. Antibodies against polyethylene glycol in healthy subjects and in patients treated with PEG-conjugated agents. Expert Opin. Drug Deliv. 9, 1319–1323 (2012).

    Article  Google Scholar 

  21. Ganson, N. J. et al. Pre-existing anti-PEG antibody linked to first-exposure allergic reactions to Pegnivacogin, a PEGylated RNA aptamer. J. Allergy Clin. Immunol. 137, 1610–1613 (2016).

    Article  Google Scholar 

  22. Qi, Y., Amiram, M., Gao, W., McCafferty, D. G. & Chilkoti, A. Sortase-catalyzed initiator attachment enables high yield growth of a stealth polymer from the C terminus of a protein. Macromol. Rapid Commun. 34, 1256–1260 (2013).

    Article  Google Scholar 

  23. Matyjaszewski, K. & Tsarevsky, N. V. Macromolecular engineering by atom transfer radical polymerization. J. Am. Chem. Soc. 136, 6513–6533 (2014).

    Article  Google Scholar 

  24. Matyjaszewski, K. & Xia, J. Atom transfer radical polymerization. Chem. Rev. 101, 2921–2990 (2001).

    Article  Google Scholar 

  25. Bontempo, D. & Maynard, H. D. Streptavidin as a macroinitiator for polymerization: in situ protein-polymer conjugate formation. J. Am. Chem. Soc. 127, 6508–6509 (2005).

    Article  Google Scholar 

  26. Gao, W. et al. In situ growth of a stoichiometric PEG-like conjugate at a protein’s N-terminus with significantly improved pharmacokinetics. Proc. Natl Acad. Sci. USA 106, 15231–15236 (2009).

    Article  Google Scholar 

  27. Gao, W., Liu, W., Christensen, T., Zalutsky, M. R. & Chilkoti, A. In situ growth of a PEG-like polymer from the C terminus of an intein fusion protein improves pharmacokinetics and tumor accumulation. Proc. Natl Acad. Sci. USA 107, 16432–16437 (2010).

    Article  Google Scholar 

  28. Peeler, J. C. et al. Genetically encoded initiator for polymer growth from proteins. J. Am. Chem. Soc. 132, 13575–13577 (2010).

    Article  Google Scholar 

  29. Lele, B. S., Murata, H., Matyjaszewski, K. & Russell, A. J. Synthesis of uniform protein-polymer conjugates. Biomacromolecules 6, 3380–3387 (2005).

    Article  Google Scholar 

  30. Ryan, S. M. et al. PK/PD modelling of comb-shaped PEGylated salmon calcitonin conjugates of differing molecular weights. J. Control. Release 149, 126–132 (2011).

    Article  Google Scholar 

  31. Magnusson, J. P., Bersani, S., Salmaso, S., Alexander, C. & Caliceti, P. In situ growth of side-chain PEG polymers from functionalized human growth hormone—a new technique for preparation of enhanced protein-polymer conjugates. Bioconjugate Chem. 21, 671–678 (2010).

    Article  Google Scholar 

  32. Lovshin, J. A. & Drucker, D. J. Incretin-based therapies for type 2 diabetes mellitus. Nat. Rev. Endocrinol. 5, 262–269 (2009).

    Article  Google Scholar 

  33. Boekhorst, J., de Been, M. W., Kleerebezem, M. & Siezen, R. J. Genome-wide detection and analysis of cell wall-bound proteins with LPxTG-like sorting motifs. J. Bacteriol. 187, 4928–4934 (2005).

    Article  Google Scholar 

  34. Meyer, D. E. & Chilkoti, A. Purification of recombinant proteins by fusion with thermally-responsive polypeptide. Nat. Biotechnol. 14, 1112–1115 (1999).

    Article  Google Scholar 

  35. Mao, H., Hart, S. A., Schink, A. & Pollok, B. A. Sortase-mediated protein ligation: a new method for protein engineering. J. Am. Chem. Soc. 126, 2670–2671 (2004).

    Article  Google Scholar 

  36. Jakubowski, W. & Matyjaszewski, K. Activators regenerated by electron transfer for atom-transfer radical polymerization of (meth)acrylates and related block copolymers. Angew. Chem. Int. Ed. 45, 4482–4486 (2006).

    Article  Google Scholar 

  37. Goke, R. et al. Exendin-4 is a high potency agonist and truncated exendin-(9-39)-amide an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells. J. Biol. Chem . 268, 19650–19655 (1993).

    Google Scholar 

  38. Winzell, M. S. & Ahren, B. The high-fat diet-fed mouse: a model for studying mechanisms and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes 53, S215–S219 (2004).

    Article  Google Scholar 

  39. Surwit, R. S., Kuhn, C. M., Cochrane, C., McCubbin, J. A. & Feinglos, M. N. Diet-induced type II diabetes in C57BL/6J mice. Diabetes 37, 1163–1167 (1988).

    Article  Google Scholar 

  40. Mack, C. M. et al. Antiobesity action of peripheral exenatide (exendin-4) in rodents: effects on food intake, body weight, metabolic status and side-effect measures. Int. J. Obes. 30, 1332–1340 (2006).

    Article  Google Scholar 

  41. Kanoski, S. E., Rupperecht, L. W., Fortin, S. M., De Jonghe, B. C. & Hayes, M. R. The role of nausea in food intake and body weight suppression by peripheral GLP-1 receptor agonists, exendin-4 and liraglutide. Neuropharmacology 62, 1916–1927 (2012).

    Article  Google Scholar 

  42. Richter, A. W. & Akerblom, E. Antibodies against polyethylene glycol produced in animals by immunization with monomethoxy polyethylene glycol modified proteins. Int. Arch. Allergy Appl. Immunol. 70, 124–131 (1983).

    Article  Google Scholar 

  43. Tsarevsky, N. V., Pintauer, T. & Matyjaszewski, K. Deactivation efficiency and degree of control over polymerization in ATRP in protic solvents. Macromolecules 37, 9768–9778 (2004).

    Article  Google Scholar 

  44. Averick, S. et al. Protein-polymer hybrids: conducting ARGET ATRP from a genetically encoded cleavable ATRP initiator. Eur. Polym. J. 49, 2919–2924 (2013).

    Article  Google Scholar 

  45. Bellucci, J. J., Bhattacharyya, J. & Chilkoti, A. A noncanonical function of sortase enables site-specific conjugation of small molecules to lysine residues in proteins. Angew. Chem. Int. Ed. 54, 441–445 (2015).

    Google Scholar 

  46. Amiram, M., Luginbuhl, K. M., Li, X., Feinglos, M. N. & Chilkoti, A. Injectable protease-operated depots of glucagon-like peptide-1 provide extended and tunable glucose control. Proc. Natl Acad. Sci. USA 110, 2792–2797 (2013).

    Article  Google Scholar 

  47. Schellenberger, V. et al. A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat. Biotechnol. 27, 1186–1188 (2009).

    Article  Google Scholar 

  48. Liao, Y.-D., Jeng, J.-C., Wang, C.-F., Wang, S.-C. & Chang, S.-T. Removal of N-terminal methionine from recombinant proteins by engineered E. coli methionine aminopeptidase. Prot. Sci. 13, 1802–1810 (2004).

    Article  Google Scholar 

  49. McDaniel, J. R., Mackay, J. A., Quiroz, F. G. & Chilkoti, A. Recursive directional ligation by plasmid reconstruction allows rapid and seamless cloning of oligomeric genes. Biomacromolecules 11, 944–952 (2010).

    Article  Google Scholar 

  50. Ilangovan, U., Ton-That, H., Iwahara, J., Schneewind, I. & Clubb, R. T. Structure of sortase, the transpeptidase that anchors proteins to the cell wall of Staphylococcus aureus . Proc. Natl Acad. Sci. USA 98, 6056–6061 (2001).

    Article  Google Scholar 

  51. Drucker, D. J. & Nauck, M. A. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, 1696–1705 (2006).

    Article  Google Scholar 

  52. Baggio, L. L., Huang, Q. L., Brown, T. J. & Drucker, D. J. A recombinant human glucagon-like peptide (GLP)-1- albumin protein (Albugon) mimics peptidergic activation of GLP-1 receptor-dependent pathways coupled with satiety, gastrointestinal motility, and glucose homeostasis. Diabetes 53, 2492–2500 (2004).

    Article  Google Scholar 

  53. Goutelle, S. et al. The Hill equation: a review of its capabilities in pharmacological modelling. Fundam. Clin. Pharmacol. 22, 633–648 (2008).

    Article  Google Scholar 

  54. Qi, Y. et al. Dataset for A brush-polymer/exendin-4 conjugate reduces blood glucose levels for up to five days and eliminates poly(ethylene glycol) antigenicity. figshare http://dx.doi.org/10.6084/m9.figshare.3976761 (2016).

  55. Zong, Y., Bice, T. W., Ton-That, H., Schneewind, O. & Narayana, S. V. Crystal structures of Staphylococcus aureus sortase A and its substrate complex. J. Biol. Chem. 279, 31383–31389 (2004).

    Article  Google Scholar 

  56. Neidigh, J. W., Fesinmeyer, R. M., Prickett, K. S. & Andersen, N. H. Exendin-4 and glucagon-like-peptide-1: NMR structural comparisons in the solution and micelle-associated states. Biochemistry 40, 13188–13200 (2001).

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank D.M. Gooden at the Duke Small Molecule Synthesis Facility for the synthesis of AEBMP, E.J. Soderblom at the Duke Proteomics Facility for conducting LC/MS-MS, G. Dubay at the Duke Chemistry Mass Spectrometry Facility for LC/ESI-MS support and M.N. Feinglos for discussion of the in vivo results. BHK cells expressing GLP-1R were a gift from the Drucker group (University of Toronto, Canada). This work was supported by the National Institutes of Health (R01-DK092665 to A.C.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Q. and A.C. conceived and designed the research. Y.Q., A.S., N.J.G., X.L, I.O. and W.L. performed the experiments. A.S. and K.M. provided technical expertise in polymer chemistry. K.M.L. and W.L. contributed to the design of the in vivo studies. N.J.G. and M.S.H. provided materials and technical expertise for the antigenicity studies. Y.Q., N.J.G., K.M.L., M.S.H., K.M. and A.C. analysed and interpreted the results. Y.Q. and A.C. wrote the manuscript and A.S., N.J.G., K.M.L., M.S.H. and K.M. edited the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Ashutosh Chilkoti.

Ethics declarations

Competing interests

A.C. and Y.Q. have a pending patent on the sortase-catalysed C-terminal polymer conjugation technology (WO 2014194244 A1). M.S.H. is a co-inventor of Pegloticase (Krystexxa) and receives royalties from sales of Pegloticase, along with his employer, Duke University. The results reported in this paper form the basis of US provisional patent applications (62/270,401; 62/329,800; 62/310,534; 62/407,403) filled by A.C., Y.Q., M.S.H. and N.J.G. through the Duke University Office of Licensing & Ventures.

Supplementary information

Supplementary information

Supplementary methods, figures and tables (PDF 1731 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qi, Y., Simakova, A., Ganson, N. et al. A brush-polymer/exendin-4 conjugate reduces blood glucose levels for up to five days and eliminates poly(ethylene glycol) antigenicity. Nat Biomed Eng 1, 0002 (2017). https://doi.org/10.1038/s41551-016-0002

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-016-0002

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