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:

Chemically engineered antibodies for autophagy-based receptor degradation

Abstract

Cell surface receptor-targeted protein degraders hold promise for drug discovery. However, their application is restricted because of the complexity of creating bifunctional degraders and the reliance on specific lysosome-shuttling receptors or E3 ubiquitin ligases. To address these limitations, we developed an autophagy-based plasma membrane protein degradation platform, which we term AUTABs (autophagy-inducing antibodies). Through covalent conjugation with polyethylenimine (PEI), the engineered antibodies acquire the capacity to degrade target receptors through autophagy. The degradation activities of AUTABs are self-sufficient, without necessitating the participation of lysosome-shuttling receptors or E3 ubiquitin ligases. The broad applicability of this platform was then illustrated by targeting various clinically important receptors. Notably, combining specific primary antibodies with a PEI-tagged secondary nanobody also demonstrated effective degradation of target receptors. Thus, our study outlines a strategy for directing plasma membrane proteins for autophagic degradation, which possesses desirable attributes such as ease of generation, independence from cell type and broad applicability.

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: PEI induces LC3C-dependent autophagy.
Fig. 2: Development of Atz-AUTAB to target PDL1.
Fig. 3: Atz-AUTAB drives PDL1 degradation through autophagic–lysosomal pathway.
Fig. 4: Degradation of EGFR and CD73 by AUTABs.
Fig. 5: AUTAB’s function depends on the cationic property of PEI.
Fig. 6: Development of secondary nanobody-based AUTABs.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the paper and its Supplementary Information and in the ScienceDB repository (https://doi.org/10.57760/sciencedb.11745). The RNA-seq data generated in this study are available from the Gene Expression Omnibus database under accession code GSE279343. The proteomic raw data were deposited to the ProteomeXchange Consortium through the iProX repository with the dataset identifier PXD056766. The tandem mass spectra obtained from quantitative proteomic analysis were searched against the full UniProt human database. Source data are provided with this paper.

References

  1. Hu, Z. et al. The Cancer Surfaceome Atlas integrates genomic, functional and drug response data to identify actionable targets. Nat. Cancer 2, 1406–1422 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tozzoli, R. Receptor autoimmunity: diagnostic and therapeutic implications. Autoimmun. Highlights 11, 1 (2020).

    Article  Google Scholar 

  3. Yin, H. & Flynn, A. D. Drugging membrane protein interactions. Annu. Rev. Biomed. Eng. 18, 51–76 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Santos, R. et al. A comprehensive map of molecular drug targets. Nat. Rev. Drug Discov. 16, 19–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Ahn, G., Banik, S. M. & Bertozzi, C. R. Degradation from the outside in: targeting extracellular and membrane proteins for degradation through the endolysosomal pathway. Cell Chem. Biol. 28, 1072–1080 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Alabi, S. B. & Crews, C. M. Major advances in targeted protein degradation: PROTACs, LYTACs, and MADTACs. J. Biol. Chem. 296, 100647 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Ahn, G. et al. LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation. Nat. Chem. Biol. 17, 937–946 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Caianiello, D. F. et al. Bifunctional small molecules that mediate the degradation of extracellular proteins. Nat. Chem. Biol. 17, 947–953 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Marei, H. et al. Antibody targeting of E3 ubiquitin ligases for receptor degradation. Nature 610, 182–189 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Pance, K. et al. Modular cytokine receptor-targeting chimeras for targeted degradation of cell surface and extracellular proteins. Nat. Biotechnol. 41, 273–281 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Mizushima, N. & Komatsu, M. Autophagy: renovation of cells and tissues. Cell 147, 728–741 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Takahashi, D. et al. AUTACs: cargo-specific degraders using selective autophagy. Mol. Cell 76, 797–810 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Li, Z. et al. Allele-selective lowering of mutant HTT protein by HTT–LC3 linker compounds. Nature 575, 203–209 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Ji, C. H. et al. The AUTOTAC chemical biology platform for targeted protein degradation via the autophagy–lysosome system. Nat. Commun. 13, 904 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gao, X. et al. The association of autophagy with polyethylenimine-induced cytotoxicity in nephritic and hepatic cell lines. Biomaterials 32, 8613–8625 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Lin, C. W., Jan, M. S. & Kuo, J. H. Autophagy-related gene expression analysis of wild-type and atg5 gene knockout mouse embryonic fibroblast cells treated with polyethylenimine. Mol. Pharm. 11, 3002–3008 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Coelho, P. P. et al. Endosomal LC3C-pathway selectively targets plasma membrane cargo for autophagic degradation. Nat. Commun. 13, 3812 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Vargas, J. N. S., Hamasaki, M., Kawabata, T., Youle, R. J. & Yoshimori, T. The mechanisms and roles of selective autophagy in mammals. Nat. Rev. Mol. Cell Biol. 24, 167–185 (2023).

    Article  CAS  PubMed  Google Scholar 

  21. Millarte, V., Schlienger, S., Kalin, S. & Spiess, M. Rabaptin5 targets autophagy to damaged endosomes and Salmonella vacuoles via FIP200 and ATG16L1. EMBO Rep. 23, e53429 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Pied, N. et al. TBK1 is part of a galectin 8 dependent membrane damage recognition complex and drives autophagy upon adenovirus endosomal escape. PLoS Pathog. 18, e1010736 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Casper, J. et al. Polyethylenimine (PEI) in gene therapy: current status and clinical applications. J. Control. Release 362, 667–691 (2023).

    Article  CAS  PubMed  Google Scholar 

  24. Guardiola, S., Varese, M., Sanchez-Navarro, M. & Giralt, E. A third shot at EGFR: new opportunities in cancer therapy. Trends Pharmacol. Sci. 40, 941–955 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Bendell, J. et al. First-in-human study of oleclumab, a potent, selective anti-CD73 monoclonal antibody, alone or in combination with durvalumab in patients with advanced solid tumors. Cancer Immunol. Immunother. 72, 2443–2458 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Burslem, G. M. et al. The advantages of targeted protein degradation over inhibition: an RTK case study. Cell Chem. Biol. 25, 67–77 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Miao, Y. et al. Bispecific aptamer chimeras enable targeted protein degradation on cell membranes. Angew. Chem. 60, 11267–11271 (2021).

    Article  CAS  Google Scholar 

  28. Zhang, H. et al. Covalently engineered nanobody chimeras for targeted membrane protein degradation. J. Am. Chem. Soc. 143, 16377–16382 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Zhou, Y. X., Teng, P., Montgomery, N. T., Li, X. L. & Tang, W. P. Development of triantennary N-acetylgalactosamine conjugates as degraders for extracellular proteins. ACS Cent. Sci. 7, 499–506 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zheng, J. W. et al. Bifunctional compounds as molecular degraders for integrin-facilitated targeted protein degradation. J. Am. Chem. Soc. 144, 21831–21836 (2022).

    Article  CAS  PubMed  Google Scholar 

  31. Wu, Y. et al. Aptamer-LYTACs for targeted degradation of extracellular and membrane proteins. Angew. Chem. Int. Ed. Engl. 62, e202218106 (2023).

    Article  CAS  PubMed  Google Scholar 

  32. Zhu, C. H., Wang, W. S., Wang, Y., Zhang, Y. & Li, J. B. Dendronized DNA chimeras harness scavenger receptors to degrade cell membrane proteins. Angew. Chem. Int. Ed. Engl. 62, e202300694 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Wang, K. et al. Nano-LYTACs for degradation of membrane proteins and inhibition of CD24/Siglec-10 signaling pathway. Adv. Sci. 10, e2300288 (2023).

    Article  Google Scholar 

  34. Gatica, D., Lahiri, V. & Klionsky, D. J. Cargo recognition and degradation by selective autophagy. Nat. Cell Biol. 20, 233–242 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fan, X., Jin, W. Y., Lu, J., Wang, J. & Wang, Y. T. Rapid and reversible knockdown of endogenous proteins by peptide-directed lysosomal degradation. Nat. Neurosci. 17, 471–480 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Shao, X. et al. Mammalian Numb protein antagonizes Notch by controlling postendocytic trafficking of the Notch ligand Delta-like 4. J. Biol. Chem. 292, 20628–20643 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Claude-Taupin, A. et al. ATG9A protects the plasma membrane from programmed and incidental permeabilization. Nat. Cell Biol. 23, 846–858 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Z. Li for her help in plasmid preparation. This work was supported by grant 2023YFA0915400 (to L.C. and H.L.) from the National Key R&D Program of China, grants 32371450 (to X.S.), 82304546 (to M.L.) and 21977111 (to L.F.) from the National Natural Science Foundation of China, grants 202381515040008 (to L.M. and H.L.), 2023A1515011765 (to L.F.) and 2021A1515012114 (to X.S.) from the Natural Science Foundation of Guangdong Province, grant 2020B1111540001 (to L.C.) from the Guangdong Provincial Key Area R&D Program, grants JCYJ20200109114608075 (to X.S.), JCYJ20210324120200001 (to H.L.) and JCYJ20210324101805014 (to K.L.) from the Shenzhen Science and Technology Program, grant D2301003 (to L.F.) from Shenzhen Medical Research Funds, grants JCYJ20220818100412028 (to L.F.) and JCYJ20220818101404010 (to L.F.) from the Shenzhen Fundamental Research Program and a grant from the SIAT Innovation Program for Excellent Young Researchers (to H.L.).

Author information

Authors and Affiliations

Authors

Contributions

H.L., L.F., X.S. and L.C. conceptualized the project and supervised the study. B.C., M.L. J.L. and X.S. performed most of the cell biological, biochemical and animal experiments. J.Z., H.T. and L.F. synthesized and characterized the AUTAB conjugates. Y.L., Z.Z., L.D., W.S., W.Z. and K.L. participated in cell culture, immunofluorescence staining and flow cytometric analysis. R.L., J.R., H.H. and L.M. provided special technical support and discussed the data. B.C., M.L., X.S., L.F. and H.L. wrote the manuscript.

Corresponding authors

Correspondence to Lintao Cai, Ximing Shao, Lijing Fang or Hongchang Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Autophagy activation upon PEI treatment.

a, The corresponding quantifications of confocal images in Fig. 1c–h and Fig. 1j (Fig. 1c, n = 10 cells; Fig. 1d, n = 12 cells; Fig. 1e, n = 15 cells; Fig. 1f, n = 15 cells; Fig. 1g, n = 12 cells; Fig. 1h, n = 10 cells; Fig. 1j, n = 15 cells, means ± s.d.; Quantification is unnecessary for the untreated group due to the absence of PEI signal in cell.). b,c, Representative images and relative quantifications of mCherry-LC3B (b) or mCherry-LC3C (c) stably expressing HeLa cells upon treatment with 1 μM of L25K PEI for indicated period of times. The quantifications are shown on the right (n = 15 cells per group, means ± s.d.). DNA in b and c was counterstained with DAPI. Scale bars, 10 μm.

Extended Data Fig. 2 AUTAB induces LC3C-mediated autophagy in a target receptor-dependent manner.

a, The corresponding quantifications of confocal images in Fig. 2d–f (Fig. 2d, n = 10 cells per group; Fig. 2e, n = 15 cells per group; Fig. 2f, Atz group, n = 14 cells, Atz-AUTAB group, n = 15 cells, means ± s.d.). b, HA-PD-L1 and mCherry-LC3C expressing HeLa cells transfected with scramble or PD-L1 siRNA were incubated with 100 nM Atz-AUTAB for 1 h. Cells were then fixed, and stained with antibodies against Atz. Enlarged images of the white box are presented on the right, with arrowheads indicating the colocalization of mCherry-LC3C and Atz-AUTAB. The quantification is shown on the right of the panel (n = 15 cells per group, means ± s.d.). DNA in b was counterstained with DAPI. Scale bars, 10 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (a, b).

Extended Data Fig. 3 Atz-AUTAB accelerates PD-L1 endocytosis.

a, Schematic representation of Atz-AUTAB uptake assay (see Methods section for details). b, Confirmation by western blotting of PD-L1 knockdown in stable expressing HA-PD-L1 HeLa cells. c, HA-PD-L1 expressing HeLa cells with scramble or PD-L1 siRNA were incubated with 1 nM Atz or Atz-AUTAB for 1 h. After fixation, the surface-remaining and internalized Atz/Atz-AUTAB were sequentially stained before or after cell permeabilization. The fluorescence intensities of internalized Atz-AUTAB were quantified, and the data are expressed as each normalized value relative to the scramble group (scramble group, n = 16 cells; siPD-L1 group, n = 14, means ± s.d.). d, HA-PD-L1 expressing HeLa cells were incubated with 1 nM Atz or Atz-AUTAB for 1 h. After fixation, the surface-remaining and internalized Atz/Atz-AUTAB were sequentially stained before or after cell permeabilization. The quantification of the internalized Atz or Atz-AUTAB is shown on the right (Atz group, n = 28 cells; Atz-AUTAB group, n = 22; means ± s.d.). e, HeLa cells stably expressing HA-PD-L1 were subjected to antibody feeding-based internalization assay. Representative examples of internalized PD-L1 and surface-remaining PD-L1 are shown. The quantification of the internalized PD-L1 is shown below as indicated (untreated group, n = 15 cells for 1 h internalization, n = 16 for 2 h; L25K PEI group, n = 14 for 1 h, n = 17 for 2 h; Atz group, n = 27 for 1 h, n = 26 for 2 h; Atz-AUTAB group, n = 18 for 1 h, n = 16 for 2 h; means ± s.d.). DNA in c and d was counterstained with DAPI. Scale bars, 10 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (c-e).

Source data

Extended Data Fig. 4 Atz-AUTAB treatment damages endosome membrane and triggers LC3C-mediated autophagy.

a,b, HeLa cells stably expressing HA-PD-L1 were treated with 100 nM Atz or Atz-AUTAB for 1 h. Cells were then fixed, co-stained using antibodies against galectin-3, Atz, and Rab5 (a) or Rab7 (b), and imaged by confocal microscopy. Enlarged views of the white boxed regions are shown below. The arrowheads indicate the triple colocalization of galectin-3, Atz-AUTAB, and Rab5 or Rab7. The relative quantification is shown on the right (n = 15 cells per group, means ± s.d.). c,d, HeLa cells concurrently expressing HA-PD-L1 and mCherry-LC3C were treated with 100 nM Atz or Atz-AUTAB for 20 min. After fixation, cells were co-stained with antibodies against Atz and Rab5 (c) or Rab7 (d), followed by confocal microscopy imaging. The white boxed regions are magnified below. The arrowheads indicate the colocalization of mCherry-LC3C and Atz-AUTAB on either early (Rab5-positive) or late (Rab7-positive) endosomes. The quantification is shown on the right (n = 15 cells per group, means ± s.d.). Scale bars, 10 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (a-d).

Extended Data Fig. 5 AUTAB induces membrane damage at cell surface.

a, Over-expressing HA-PD-L1 and mCherry-LC3C HeLa cells were treated with 100 nM Atz or Atz-AUTAB for 1 h, then fixed and co-stained with antibodies against ATG9A and Atz. Images were taken by confocal microscopy. The white boxed regions in the images are enlarged below and the arrowheads point to the colocalization between ATG9A, mCherry-LC3C and Atz-AUTAB. The corresponding colocalization quantization is on the right (n = 15 cells per group, means ± s.d.). b, Plasma membrane permeabilization was evaluated with PI uptake assay in stably expressing HA-PD-L1 HeLa cells treated with 250 nM L25K PEI, 100 nM Atz, 25 nM and 100 nM Atz-AUTAB for 1 h. Percentage of cells with PI-positive was quantified (n = 3 biological replicates, means ± s.d.). DNA in b was counterstained with Hoechst 33258. Scale bars, 10 μm (a) or 100 μm (b). Statistical significance was calculated via unpaired two-tailed Student’s t-test (a, b).

Extended Data Fig. 6 Mechanical characterization of Atz-AUTAB-mediated PD-L1 degradation.

a-c, Degradation of exogenously expressed PD-L1 assessed by western blotting in HA-PD-L1 stably expressing HeLa cells following treatment as in Fig. 3a–c. d, Western blot of PD-L1 in MDA-MB-231 cells treated with 6.25 nM Atz-AUTAB for 24 h in the presence or absence of 200 μM chloroquine. e, Western blot analysis of PD-L1 and LC3B in MDA-MB-231 cells treated with indicated concentrations of Atz, L25K PEI, and Atz-AUTAB for 12 h and followed by release for 12 h. f, MDA-MB-231 cells with or without siRNA mediated knockdown of LC3B were subjected to treatment with 6.25 nM Atz-AUTAB for 12 h. The PD-L1 levels were detected by immunoblotting. g, Western blot analysis of GABARAP, GABARAPL1, GABARAPL2 in LC3C knockout MDA-MB-231 cells treated with or without 6.25 nM Atz-AUTAB for 12 h. h, Western blot of PD-L1 in MDA-MB-231 cells treated with 6.25 nM Atz-AUTAB for 12 h under siRNA-mediated knockdown of GABARAP, GABARAPL1, and GABARAPL2 respectively. i, HA-PD-L1 stable expressing HeLa cells transfected with scramble or LC3C, GABARAP, GABARAPL1, and GABARAPL2 siRNA were incubated with 100 nM Atz-AUTAB for 1 h. Cells were then fixed and co-stained with antibodies against galectin-3, Lamp1 and Atz. The enlarged images of the white box are presented below, with arrowheads indicating the colocalization of galectin-3, Lamp1 and Atz-AUTAB. The quantification is shown on the right (n = 15 cells per group, means ± s.d.). j, Western blot of PD-L1 in MDA-MB-231 cells treated with 6.25 nM Atz-AUTAB for the indicated time (0, 12, 16, 20 h) under siRNA-mediated knockdown of ATG4B. Scale bars, 10 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (i).

Source data

Extended Data Fig. 7 EGFR degradation driven by Ctx-AUTAB.

a, SDS-PAGE analysis of the Ctx-AUTAB with Coomassie blue staining. b, Representative surface EGFR immunostaining images of HeLa cells treated with 10 nM L25K PEI, 5 nM Ctx or 5 nM Ctx-AUTAB for 4 h. c, Flow cytometry analysis of surface EGFR levels in live HeLa cells upon treatment with 10 nM L25K PEI, 5 nM Ctx or 5 nM Ctx-AUTAB for 4 h. Mean fluorescence intensity of surface EGFR relative to the untreated group was quantified (n = 3 biological replicates, means ± s.d.). d, Quantification of triple colocalization between mCherry-LC3C, EGFR and Ctx-AUTAB or Ctx in Fig. 4a (n = 15 cells per group, means ± s.d.). e,f, Representative confocal images (e) and quantification (f) showing the colocalization of EGFR and RFP-Lamp1 in HeLa cells following 1 h of treatment with 10 nM L25K PEI, 5 nM Ctx or 5 nM Ctx-AUTAB (n = 10 cells per group, means ± s.d.). g, Western blot analysis of EGFR in HeLa cells treated with increasing concentrations of Ctx-AUTAB for 24 h. h, Western blot of EGFR in HeLa cells following treatment with 25 nM of Ctx-AUTAB for the indicated time periods. DNA in b and e was counterstained with DAPI. Scale bars, 10 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (c, d, f).

Source data

Extended Data Fig. 8 Ole-AUTAB accelerates CD73 degradation.

a, SDS-PAGE analysis of the Ole-AUTAB with Coomassie blue staining. b, Quantification of colocalization between mCherry-CD73 and LysoTracker in Fig. 4h (n = 15 cells per group, means ± s.d.). c, Live-cell flow cytometry analysis of surface CD73 levels in mCherry-CD73 stably expressing HeLa cells upon treatment with 200 nM L25K PEI, 100 nM Ole or 100 nM Ole-AUTAB for 12 h. Mean fluorescence intensity of surface CD73 relative to the untreated group was quantified (n = 3 biological replicates, means ± s.d.). d,e, Western blot analysis of CD73 in U87-MG cells upon treatment with increasing concentrations of Ole-AUTAB for 24 h (d), or upon treatment with 100 nM of Ole-AUTAB for different times (e). f, Western blotting to detect the CD73 degradation in Ole-AUTAB treated U87-MG cells in the presence or absence of bafilomycin A1 or chloroquine for 24 h. Statistical significance was calculated via unpaired two-tailed Student’s t-test (b, c).

Source data

Extended Data Fig. 9 Gel electrophoresis validation and functional examination of various types of established AUTABs.

a,b, SDS-PAGE analysis of distinct Atz-AUTABs covalently tagged with various types of PEIs, including L2.5 K PEI, B800 PEI, B2K PEI and B25 K PEI (a), and Ctx-AUTAB and Ole-AUTAB that were both conjugated with L2.5K PEI (b). c, Western blot analysis of EGFR in HeLa cells or CD73 in U87-MG cells that were respectively treated with Ctx-AUTABs or Ole-AUTABs for 24 h at the indicated concentrations. d, SDS-PAGE analysis of Atz-AUTABs respectively prepared using acetylated L2.5 K PEI. e, Quantification of colocalization between Atz-AUTAB (L2.5 K), mCherry-LC3C and HA-PD-L1 in Fig. 5f (n = 15 cells per group, means ± s.d.). f, SDS-PAGE analysis of Atz-AUTABs respectively prepared using poly-L-Lys with varying sizes (4–15 K, and 15–30 K). g, Quantification of colocalization between PolyLys-Atz-AUTABs, mCherry-LC3C and HA-PD-L1 in Fig. 5h (n = 15 cells per group, means ± s.d.). Statistical significance was calculated via unpaired two-tailed Student’s t-test (e, g).

Source data

Extended Data Fig. 10 Therapeutic potential of AUTAB in vivo.

a, Schematic illustration of the general treatment procedure for anti-tumor study. C57BL/6 J mice with MC38 subcutaneous xenografts were administered doses of 5 mg/kg mouse PD-L1 antibody (mPD-L1-Ab) or mPD-L1-Ab-AUTAB by intravenous injection every 2 ~ 3 days for 5 times. b, SDS-PAGE analysis of the mPD-L1-Ab-AUTAB with Coomassie blue staining. c,g, Body weights (c) and MC38 tumor volumes (d) of mice were monitored during administration with mPD-L1-Ab or mPD-L1-Ab-AUTAB (n = 8 mice per group, means ± s.d.). e,f, Photographs of excised tumors are shown (e) and the weight of tumors was quantified (f) at the end of the treatments (n = 8 mice per group, means ± s.d.). g, Western blot of PD-L1 in MC38 xenografts in e (n = 4 mice per group). h, Tumor tissues in e were sectioned, stained for CD8, and imaged. The mean number of CD8-positive cells per field was quantified and is shown on the right (n = 4 mice, means ± s.d.). DNA in h was counterstained with DAPI. Scale bars, 50 μm. Statistical significance was calculated via unpaired two-tailed Student’s t-test (d, f, h).

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–4, Figs. 1–16, unprocessed blots for supplementary figures and Supplementary Note.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Unprocessed gels.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots and gels.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Unprocessed western blots and gels.

Source Data Extended Data Fig. 8

Unprocessed western blots and gels.

Source Data Extended Data Fig. 9

Unprocessed western blots and gels.

Source Data Extended Data Fig. 10

Unprocessed western blots and gels.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cheng, B., Li, M., Zheng, J. et al. Chemically engineered antibodies for autophagy-based receptor degradation. Nat Chem Biol 21, 855–866 (2025). https://doi.org/10.1038/s41589-024-01803-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41589-024-01803-1

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