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:

Bifunctional small molecules that mediate the degradation of extracellular proteins

Abstract

Targeted protein degradation (TPD) has emerged as a promising therapeutic strategy. Most TPD technologies use the ubiquitin–proteasome system, and are therefore limited to targeting intracellular proteins. To address this limitation, we developed a class of modular, bifunctional synthetic molecules called MoDE-As (molecular degraders of extracellular proteins through the asialoglycoprotein receptor (ASGPR)), which mediate the degradation of extracellular proteins. MoDE-A molecules mediate the formation of a ternary complex between a target protein and ASGPR on hepatocytes. The target protein is then endocytosed and degraded by lysosomal proteases. We demonstrated the modularity of the MoDE-A technology by synthesizing molecules that induce depletion of both antibody and proinflammatory cytokine proteins. These data show experimental evidence that nonproteinogenic, synthetic molecules can enable TPD of extracellular proteins in vitro and in vivo. We believe that TPD mediated by the MoDE-A technology will have widespread applications for disease treatment.

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: Bifunctional MoDE-A molecule design and chemical structures.
Fig. 2: Bifunctional MoDE-A molecules mediate endocytosis of target proteins.
Fig. 3: Target protein α-DNP antibody is trafficked to lysosomes and degraded.
Fig. 4: D-MoDE-A mediates the accelerated depletion of α-DNP antibodies from serum in vivo.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article (and its Supplementary information files). Source data are provided with this paper.

References

  1. Sun, X. et al. PROTACs: great opportunities for academia and industry. Sig. Transduct. Target Ther. 4, 64 (2019).

    Article  Google Scholar 

  2. Chen, B., Shi, X., Cui, Y., Hou, A. & Zhao, P. A review of PCSK9 inhibitors and their effects on cardiovascular diseases. Curr. Top. Med. Chem. 19, 1790–1817 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Kalliolias, G. D. & Ivashkiv, L. B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 12, 49–62 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Martínez-Fábregas, J. et al. Lysosomal protease deficiency or substrate overload induces an oxidative-stress mediated STAT3-dependent pathway of lysosomal homeostasis. Nat. Commun. 9, 5343 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Zoja, C., Benigni, A. & Remuzzi, G. Protein overload activates proximal tubular cells to release vasoactive and inflammatory mediators. Nephron Exp. Nephrol. 7, 420–428 (1999).

    Article  CAS  Google Scholar 

  6. Nakajima, H. et al. Activation of the signal transducer and activator of transcription signaling pathway in renal proximal tubular cells by albumin. J. Am. Soc. Nephrol. 15, 276–285 (2004).

    Article  CAS  PubMed  Google Scholar 

  7. Tanowitz, M. et al. Asialoglycoprotein receptor 1 mediates productive uptake of N-acetylgalactosamine-conjugated and unconjugated phosphorothioate antisense oligonucleotides into liver hepatocytes. Nucleic Acids Res. 45, 12388–12400 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mi, Y., Lin, A., Fiete, D., Steirer, L. & Baenziger, J. U. Modulation of mannose and asialoglycoprotein receptor expression determines glycoprotein hormone half-life at critical points in the reproductive cycle. J. Biol. Chem. 289, 12157–12167 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yang, W. H. et al. An intrinsic mechanism of secreted protein aging and turnover. Proc. Natl Acad. Sci. USA 112, 13657–13662 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Steirer, L. M., Park, E. I., Townsend, R. R. & Baenziger, J. U. The asialoglycoprotein receptor regulates levels of plasma glycoproteins terminating with sialic acid α2, 6-galactose. J. Biol. Chem. 284, 3777–3783 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Grozovsky, R. et al. The Ashwell–Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling. Nat. Med. 21, 47–54 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Huang, X., Leroux, J. C. & Castagner, B. Well-defined multivalent ligands for hepatocytes targeting via asialoglycoprotein receptor. Bioconjugate Chem. 28, 283–295 (2016).

    Article  CAS  Google Scholar 

  13. D’Souza, A. A. & Devarajan, P. V. Asialoglycoprotein receptor mediated hepatocyte targeting—strategies and applications. J. Control. Release 203, 126–139 (2015).

    Article  PubMed  CAS  Google Scholar 

  14. Matsuda, S. et al. siRNA conjugates carrying sequentially assembled trivalent N-acetylgalactosamine linked through nucleosides elicit robust gene silencing in vivo in hepatocytes. ACS Chem. Biol. 10, 1181–1187 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Biessen, E. A. et al. Novel hepatotrophic prodrugs of the antiviral nucleoside 9-(2-phosphonylmethoxyethyl) adenine with improved pharmacokinetics and antiviral activity. FASEB J. 14, 1784–1792 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Pujol, A. M. et al. Hepatocyte targeting and intracellular copper chelation by a thiol-containing glycocyclopeptide. J. Am. Chem. Soc. 133, 286–296 (2010).

    Article  PubMed  CAS  Google Scholar 

  17. Akinc, A. et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol. Ther. 18, 1357–1364 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bocci, V. The role of sialic acid in determining the life-span of circulating cells and glycoproteins. Experientia 32, 135–140 (1976).

    Article  CAS  PubMed  Google Scholar 

  19. Baenziger, J. U. & Fiete, D. Galactose and N-acetylgalactosamine-specific endocytosis of glycopeptides by isolated rat hepatocytes. Cell 22, 611–620 (1980).

    Article  CAS  PubMed  Google Scholar 

  20. Merwin, J. R. et al. Targeted delivery of DNA using YEE (GalNAcAH)3, a synthetic glycopeptide ligand for the asialoglycoprotein receptor. Bioconjugate Chem. 5, 612–620 (1994).

    Article  CAS  Google Scholar 

  21. Rensen, P. C. et al. Determination of the upper size limit for uptake and processing of ligands by the asialoglycoprotein receptor on hepatocytes in vitro and in vivo. J. Biol. Chem. 276, 37577–37584 (2001).

    Article  CAS  PubMed  Google Scholar 

  22. Mamidyala, S. K. et al. Glycomimetic ligands for the human asialoglycoprotein receptor. J. Am. Chem. Soc. 134, 1978–1981 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Onizuka, T. et al. NMR study of ligand release from asialoglycoprotein receptor under solution conditions in early endosomes. FEBS J. 279, 2645–2656 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. LaBadie, J. H., Chapman, K. P. & Aronson, N. N. Glycoprotein catabolism in rat liver: lysosomal digestion of iodinated asialo-fetuin. Biochem. J. 152, 271–279 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bilsborrow, J. B., Doherty, E., Tilstam, P. V. & Bucala, R. Macrophage migration inhibitory factor (MIF) as a therapeutic target for rheumatoid arthritis and systemic lupus erythematosus. Expert Opin. Ther. 23, 733–744 (2019).

    Article  CAS  Google Scholar 

  26. Banik, S. M. et al. Lysosome-targeting chimaeras for degradation of extracellular proteins. Nature 584, 291–297 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhou, Y., Teng, P., Montgomery, N. T., Li, X. & Tang, W. Development of triantennary N-acetylgalactosamine conjugates as degraders for extracellular proteins. ACS Cent. Sci. https://doi.org/10.1021/acscentsci.1c00146 (2021).

  28. Cotton, A. D., Nguyen, D. P., Gramespacher, J. A., Seiple, I. B. & Wells, J. A. Development of antibody-based PROTACs for the degradation of the cell-surface immune checkpoint protein PD-L1. J. Am. Chem. Soc. 143, 593–598 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Spiegel, D. A., Caianiello, D. & Zhang, M. Bifunctional small molecules to target the selective degradation of circulating proteins. PCT patent WO 2019/199634. US patent US62/655,055 US62/788,040 (2019).

  30. Cisneros, J. A., Robertson, M. J., Valhondo, M. & Jorgensen, W. L. A fluorescence polarization assay for binding to macrophage migration inhibitory factor and crystal structures for complexes of two potent inhibitors. J. Am. Chem. Soc. 138, 8630–8638 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Douglass, E. F. Jr., Miller, C. J., Sparer, G., Shapiro, H. & Spiegel, D. A. A comprehensive mathematical model for three-body binding equilibria. J. Am. Chem. Soc. 135, 6092–6099 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Brown, W., Saunders, N., Møsllgård, K. & Dziegielewska, K. Fetuin–an old friend revisited. BioEssays 14, 749–755 (1992).

    Article  CAS  PubMed  Google Scholar 

  33. Fernandes, C. L., Ligabue-Braun, R. & Verli, H. Structural glycobiology of human α1-acid glycoprotein and its implications for pharmacokinetics and inflammation. Glycobiology 25, 1125–1133 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Shi, B., Abrams, M. & Sepp-Lorenzino, L. Expression of asialoglycoprotein receptor 1 in human hepatocellular carcinoma. J. Histochem. Cytochem. 61, 901–909 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cooper, A. & Shaul, Y. Clathrin-mediated endocytosis and lysosomal cleavage of hepatitis B virus capsid-like core particles. J. Biol. Chem. 281, 16563–16569 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Guo, S. et al. Selectivity of commonly used inhibitors of clathrin-mediated and caveolae-dependent endocytosis of G protein–coupled receptors. BBA-Biomembranes 1848, 2101–2110 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Cervia, L. D., Chang, C. C., Wang, L. & Yuan, F. Distinct effects of endosomal escape and inhibition of endosomal trafficking on gene delivery via electrotransfection. PloS ONE 12, e42703 (2017).

    Article  CAS  Google Scholar 

  38. Ippoliti, R. et al. The effect of monensin and chloroquine on the endocytosis and toxicity of chimeric toxins. Cell. Mol. Life Sci. 54, 866–875 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Ba, Q. et al. Dihydroartemisinin exerts its anticancer activity through depleting cellular iron via transferrin receptor-1. PloS ONE 7, e42703 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zegers, M. M., Zaal, K. J., van IJzendoorn, S. C., Klappe, K. & Hoekstra, D. Actin filaments and microtubules are involved in different membrane traffic pathways that transport sphingolipids to the apical surface of polarized HepG2 cells. Mol. Biol. Cell 9, 1939–1949 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Maidorn, R., Cragoe, E. & Tannock, I. Therapeutic potential of analogues of amiloride: inhibition of the regulation of intracellular pH as a possible mechanism of tumour selective therapy. Br. J. Cancer 67, 297–303 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Oka, J., Christensen, M. & Weigel, P. Hyperosmolarity inhibits galactosyl receptor-mediated but not fluid phase endocytosis in isolated rat hepatocytes. J. Biol. Chem. 264, 12016–12024 (1989).

    Article  CAS  PubMed  Google Scholar 

  43. Schwartz, A. L., Bolognesi, A. & Fridovich, S. E. Recycling of the asialoglycoprotein receptor and the effect of lysosomotropic amines in hepatoma cells. J. Cell Biol. 98, 732–738 (1984).

    Article  CAS  PubMed  Google Scholar 

  44. Dziedzic, P. et al. Design, synthesis, and protein crystallography of biaryltriazoles as potent tautomerase inhibitors of macrophage migration inhibitory factor. J. Am. Chem. Soc. 137, 2996–3003 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bridges, K., Harford, J., Ashwell, G. & Klausner, R. D. Fate of receptor and ligand during endocytosis of asialoglycoproteins by isolated hepatocytes. Proc. Natl Acad. Sci. USA 79, 350–354 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ye, S., Reardon, C. & Getz, G. S. Inhibition of apolipoprotein E degradation in a post-Golgi compartment by a cysteine protease inhibitor. J. Biol. Chem. 268, 8497–8502 (1993).

    Article  CAS  PubMed  Google Scholar 

  47. Jevsevar, S. & Kunstelj, M. in Therapeutic Proteins: Strategies to Modulate Their Plasma Half-lives (ed. Kontermann, R.) Ch. 3 (Wiley-Blackwell, 2012).

  48. Wall, D. A., Wilson, G. & Hubbard, A. L. The galactose-specific recognition system of mammalian liver: the route of ligand internalization in rat hepatocytes. Cell 21, 79–93 (1980).

    Article  CAS  PubMed  Google Scholar 

  49. Guy, C. S., Rankin, S. L. & Michalak, T. I. Hepatocyte cytotoxicity is facilitated by asialoglycoprotein receptor. Hepatol 54, 1043–1050 (2011).

    Article  CAS  Google Scholar 

  50. Benseler, V. et al. Hepatocyte entry leads to degradation of autoreactive CD8 T cells. Proc. Natl Acad. Sci. USA 108, 16735–16740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Weigel, P. & Oka, J. Temperature dependence of endocytosis mediated by the asialoglycoprotein receptor in isolated rat hepatocytes. Evidence for two potentially rate-limiting steps. J. Biol. Chem. 256, 2615–2617 (1981).

    Article  CAS  PubMed  Google Scholar 

  52. Eshhar, Z., Ofarim, M. & Waks, T. Generation of hybridomas secreting murine reaginic antibodies of anti-DNP specificity. J. Immunol. Res. 124, 775–780 (1980).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Paulsen and S. Miller for use of equipment and the Eshhar laboratory for providing hybridomas for antibody production. We are thankful for financial support from the National Institute of Health Yale Chemical Biology Training grants to J.D.R. (no. 2T32GM06754 3-17), E.M.J.B. (5T32GM06754 3-12), R.A.H. (T32 GM067543) and D.F.C. (T32GM067543), the Department of Defense (BC120554) and Yale University (Michele Dufault Summer Research Fellowship to A.Z.G.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

D.F.C., M.Z., V.M. and D.A.S. conceived the project and designed experiments. D.F.C., M.Z., E.C., A.Z.G. and J.C.S synthesized compounds. D.F.C., J.D.R., J.C.S., E.M.J.B. and R.A.H. performed in vitro and cellular experiments. V.R.S. performed in vivo experiments. D.F.C. and D.M. performed statistical analysis. D.F.C., J.D.R., D.M.M. and D.A.S. wrote the manuscript with input from all authors. D.A.S. provided supervision.

Corresponding author

Correspondence to David A. Spiegel.

Ethics declarations

Competing interests

This work is the subject of two published PCT patent applications: PCT/US2019/026239 and PCT/US2019/026260. D.A.S. holds equity in Biohaven Pharmaceuticals, and also receives honoraria for consulting for Biohaven. Biohaven has licensed intellectual property from Yale University pertaining to the subject matter of this study.

Additional information

Peer review information Nature Chemical Biology thanks Padma Devarajan, Joshiawa Paulk and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1

a) Chemical structure of α-DNP antibody binding control molecule DNP-OH3. b) D-MoDE-A-mediated ternary complex formation is inhibited by competitive binders of either ASGPR (GalNAc, ‘GN’) or α-DNP antibody (‘DNP-OH3’). Each data point represents the mean ± SEM of a distinct biological replicate (n = 3). The line represents the mean. c) Endocytosis of α-DNP antibody in hepatocytes is dependent on both α-DNP antibody and D-MoDE-A concentration (12 hour time point). Each value represents a single biological replicate. d) Endocytosis of fluorescently labeled MIF is dependent on M-MoDE-A concentration. Each data point represents a distinct biological replicate (n = 4). The line represents the mean. e) Representative gating strategy for flow cytometry experiments.

Source data

Extended Data Fig. 2

a) Intracellular AF488 signal in colocalization studies is dependent on the presence of α-DNP antibody and D-MoDE-A. In cells treated with an isotopye control primary antibody, no EEA1 or LAMP2 signal is observed. The scale bar is 10 μm. These images are representative of three replicates. b) The majority of AF488 signal in cell culture supernatants is associated with full-length antibody. Experiments were performed in HepG2 cells. This blot is representative of three biological replicates. Uncropped blots are shown in Source data.

Extended Data Fig. 3

a) Plasma concentration of D-MoDE-A over time following a 1 mg/kg IP dose in male nude mice. b) Mouse body weight following treatment with D-MoDE-A or DNP-OH3 (data collected from the mice in the study described by Fig. 4b). Statistical differences were analyzed by ANOVA with Kruskall-Wallis test for post-hoc comparison between each treatment group and the PBS group. No significant differences were found (P > 0.9 in all cases). Each data point represents the mean ± SEM of a distinct mouse (n = 5). c) Levels of aspartate transaminase (AST) in treated mice (data from pooled serum from mice in the study described by Fig. 4b). Dashed lines represent the normal range. Each data point represents a technical replicate (n = 2). d) Levels of alanine transaminase (ALT) in treated mice (data from pooled serum from mice in the study described by Fig. 4b). Dashed lines represent the normal range. Each data point represents a technical replicate (n = 2). e) Decrease in serum levels of α-DNP antibody following a single variable dose of D-MoDE-A. Each experimental group contained at least eight mice. Statistical differences were assessed by repeated measures two-way ANOVA with Dunnett’s test for post-hoc comparison of simple effects between each of the treatment groups and PBS. Each data point represents the mean ± SEM of a distinct biological replicate (n = 10 for 0.1 mpk D-MoDE-A; n = 8 for all others). f) α-DNP antibody was co-injected with MIF. Mice treated with M-MoDE-A, D-MoDE-A, and 3w do not demonstrate rapid clearance of α-DNP antibody from circulation at early time points. Each data point represents the mean ± SEM of the concentration of α-DNP antibody in serum collected from five mice. Statistical differences were assessed by repeated measures two-way ANOVA with Tukey’s tests for post-hoc comparison of simple effects between each of the treatment groups and PBS.

Source data

Supplementary information

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

a) Uncropped α-AF488 blot corresponding to Fig. 3c. b) Uncropped α-actin blot corresponding to Fig. 3c. c) Uncropped α-AF488 blot corresponding to Fig. 3d. d) Uncropped α-actin blot corresponding to Fig. 3d.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

a) Uncropped α-AF488 blot corresponding to Extended Data Fig. 2b. b) Uncropped α-actin blot corresponding to Extended Data Fig. 2b.

Source Data Extended Data Fig. 3

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caianiello, D.F., Zhang, M., Ray, J.D. et al. Bifunctional small molecules that mediate the degradation of extracellular proteins. Nat Chem Biol 17, 947–953 (2021). https://doi.org/10.1038/s41589-021-00851-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00851-1

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