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:

Snapshots and ensembles of BTK and cIAP1 protein degrader ternary complexes

Abstract

Heterobifunctional chimeric degraders are a class of ligands that recruit target proteins to E3 ubiquitin ligases to drive compound-dependent protein degradation. Advancing from initial chemical tools, protein degraders represent a mechanism of growing interest in drug discovery. Critical to the mechanism of action is the formation of a ternary complex between the target, degrader and E3 ligase to promote ubiquitination and subsequent degradation. However, limited insights into ternary complex structures exist, including a near absence of studies on one of the most widely co-opted E3s, cellular inhibitor of apoptosis 1 (cIAP1). In this work, we use a combination of biochemical, biophysical and structural studies to characterize degrader-mediated ternary complexes of Bruton’s tyrosine kinase and cIAP1. Our results reveal new insights from unique ternary complex structures and show that increased ternary complex stability or rigidity need not always correlate with increased degradation efficiency.

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

Fig. 1: IAP degrades BTK via BC5P.
Fig. 2: The IAP–BTK–BC5P ternary complex is non-cooperative.
Fig. 3: The IAP–BTK–BC5P ternary complex is flexible.
Fig. 4: Structure-based linker design yields a degrader with distinct biochemical properties.
Fig. 5: Distinct IAP–BTK crystal structure.
Fig. 6: IAP–BTK can form higher-order assemblies.

Similar content being viewed by others

Data availability

Structural coordinates from X-ray crystallography experiments have been deposited at RCSB with the following accession codes: 6W74 (cIAP1–BC5P binary structure), 6W7O (BTK–BCPyr–cIAP1 ternary structure) and 6W8I (BTK–BC5P–cIAP1 ternary structure). NMR data have been deposited to the Biological Magnetic Resonance Data Bank. Source data are provided with this paper.

References

  1. Sakamoto, K. M. et al. Protacs: chimeric molecules that target proteins to the Skp1–cullin–F box complex for ubiquitination and degradation. Proc. Natl Acad. Sci. USA 98, 8554–8559 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ohoka, N. et al. In vivo knockdown of pathogenic proteins via specific and nongenetic inhibitor of apoptosis protein (IAP)-dependent protein erasers (SNIPERs). J. Biol. Chem. 292, 4556–4570 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bondeson, D. P. et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sun, X. et al. A chemical approach for global protein knockdown from mice to non-human primates. Cell Discov. 5, 10 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Schneekloth, A. R., Pucheault, M., Tae, H. S. & Crews, C. M. Targeted intracellular protein degradation induced by a small molecule: en route to chemical proteomics. Bioorg. Med. Chem. Lett. 18, 5904–5908 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Mullard, A. First targeted protein degrader hits the clinic. Nat. Rev. Drug Discov. 18, 237–239 (2019).

    Google Scholar 

  7. Schapira, M., Calabrese, M. F., Bullock, A. N. & Crews, C. M. Targeted protein degradation: expanding the toolbox. Nat. Rev. Drug Discov. 18, 949–963 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Lu, J. et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755–763 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nowak, R. P. et al. Plasticity in binding confers selectivity in ligand-induced protein degradation. Nat. Chem. Biol. 14, 706–714 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zorba, A. et al. Delineating the role of cooperativity in the design of potent PROTACs for BTK. Proc. Natl Acad. Sci. USA 115, E7285–E7292 (2018).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  11. Winter, G. E. et al. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 348, 1376–1381 (2015).

    Article  PubMed Central  CAS  Google Scholar 

  12. Farnaby, W. et al. BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nat. Chem. Biol. 15, 672–680 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gadd, M. S. et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol. 13, 514–521 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Min, J.-H. et al. Structure of an HIF-1α–pVHL complex: hydroxyproline recognition in signaling. Science 296, 1886–1889 (2002).

    Article  Google Scholar 

  15. Zoppi, V. et al. Iterative design and optimization of initially inactive proteolysis targeting chimeras (PROTACs) identify VZ185 as a potent, fast, and selective von Hippel–Lindau (VHL) based dual degrader probe of BRD9 and BRD7. J. Med. Chem. 62, 699–726 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Peng, Y. et al. Bivalent Smac mimetics with a diazabicyclic core as highly potent antagonists of XIAP and cIAP1/2 and novel anticancer agents. J. Med. Chem. 55, 106–114 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Sun, H., Lu, J., Liu, L., Yang, C.-Y. & Wang, S. Potent and selective small-molecule inhibitors of cIAP1/2 proteins reveal that the binding of Smac mimetics to XIAP BIR3 is not required for their effective induction of cell death in tumor cells. ACS Chem. Biol. 9, 994–1002 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Varfolomeev, E. et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-κB activation, and TNFα-dependent apoptosis. Cell 131, 669–681 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Chessari, G. et al. Fragment-based drug discovery targeting inhibitor of apoptosis proteins: discovery of a non-alanine lead series with dual activity against cIAP1 and XIAP. J. Med. Chem. 58, 6574–6588 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Tamanini, E. et al. Discovery of a potent nonpeptidomimetic, small-molecule antagonist of cellular inhibitor of apoptosis protein 1 (cIAP1) and X-linked inhibitor of apoptosis protein (XIAP). J. Med. Chem. 60, 4611–4625 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Sun, H. et al. Nonpeptidic and potent small-molecule inhibitors of cIAP-1/2 and XIAP proteins. J. Med. Chem. 53, 6361–6367 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ohoka, N. et al. Derivatization of inhibitor of apoptosis protein (IAP) ligands yields improved inducers of estrogen receptor α degradation. J. Biol. Chem. 293, 6776–6790 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Okuhira, K. et al. Specific degradation of CRABP-II via cIAP1-mediated ubiquitylation induced by hybrid molecules that crosslink cIAP1 and the target protein. FEBS Lett. 585, 1147–1152 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Itoh, Y., Ishikawa, M., Naito, M. & Hashimoto, Y. Protein knockdown using methyl bestatin–ligand hybrid molecules: design and synthesis of inducers of ubiquitination-mediated degradation of cellular retinoic acid-binding proteins. J. Am. Chem. Soc. 132, 5820–5826 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Itoh, Y. et al. Development of target protein-selective degradation inducer for protein knockdown. Bioorg. Med. Chem. 19, 3229–3241 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Itoh, Y., Kitaguchi, R., Ishikawa, M., Naito, M. & Hashimoto, Y. Design, synthesis and biological evaluation of nuclear receptor-degradation inducers. Bioorg. Med. Chem. 19, 6768–6778 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Demizu, Y. et al. Design and synthesis of estrogen receptor degradation inducer based on a protein knockdown strategy. Bioorg. Med. Chem. Lett. 22, 1793–1796 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Tinworth, C. P. et al. PROTAC-mediated degradation of Bruton’s tyrosine kinase is inhibited by covalent binding. ACS Chem. Biol. 14, 342–347 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Casillas, L. N., Harling, J. D. M., Afjal H., Smith, I. E. D. & Rackham, M. D. Preparation of PROTAC compounds conjugates comprising RIPK2 inhibitors, especially IAP binder linked RIPK2 inhibitors, for treating inflammations. International patent WO2017182418 A1 (2017).

  30. Shibata, N. et al. Development of protein degradation inducers of oncogenic BCR–ABL protein by conjugation of ABL kinase inhibitors and IAP ligands. Cancer Sci. 108, 1657–1666 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Huang, H.-T. et al. A chemoproteomic approach to query the degradable kinome using a multi-kinase degrader. Cell Chem. Biol. 25, 88–99 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Byrd, J. C. et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 369, 32–42 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wang, M. L. et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N. Engl. J. Med. 369, 507–516 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pan, Z. et al. Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem 2, 58–61 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Honigberg, L. A. et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc. Natl Acad. Sci. USA 107, 13075–13080 (2010).

    Article  PubMed Central  Google Scholar 

  36. Buhimschi, A. D. et al. Targeting the C481S ibrutinib-resistance mutation in Bruton’s tyrosine kinase using PROTAC-mediated degradation. Biochemistry 57, 3564–3575 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Woyach, J. A. et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N. Engl. J. Med. 370, 2286–2294 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Sun, Y. et al. PROTAC-induced BTK degradation as a novel therapy for mutated BTK C481S induced ibrutinib-resistant B-cell malignancies. Cell Res. 28, 779–781 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Borzilleri, R. M. P. et al. Preparation of peptide IAP antagonists for cancer treatment. International patent WO2014047024A1 (2013).

  40. Feltham, R. et al. Smac mimetics activate the E3 ligase activity of cIAP1 protein by promoting RING domain dimerization. J. Biol. Chem. 286, 17015–17028 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Horst, R., Horwich, A. L. & Wüthrich, K. Translational diffusion of macromolecular assemblies measured using transverse-relaxation-optimized pulsed field gradient NMR. J. Am. Chem. Soc. 133, 16354–16357 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sekine, K. et al. Small molecules destabilize cIAP1 by activating auto-ubiquitylation. J. Biol. Chem. 283, 8961–8968 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Dueber, E. C. et al. Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination. Science 334, 376–380 (2011).

    Article  CAS  Google Scholar 

  44. Lopez, J. et al. CARD-mediated autoinhibition of cIAP1’s E3 ligase activity suppresses cell proliferation and migration. Mol. Cell 42, 569–583 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Ciulli, A., Testa, A., Hughes, S. J., Lucas, X. & Wright, J. E. Structure-based design of a macrocyclic PROTAC. Angew. Chem. Int. Ed. Engl. https://doi.org/10.1002/anie.201914396 (2019).

  46. Duda, D. M. et al. Structural insights into NEDD8 activation of cullin-RING ligases: conformational control of conjugation. Cell 134, 995–1006 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Baek, K. et al. NEDD8 nucleates a multivalent cullin–RING–UBE2D ubiquitin ligation assembly. Nature 578, 461–466 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Mares, A. et al. Extended pharmacodynamic responses observed upon PROTAC-mediated degradation of RIPK2. Commun. Biol. 3, 140 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhang, X. et al. Discovery of IAP-recruiting BCL-XL PROTACs as potent degraders across multiple cancer cell lines. Eur. J. Med. Chem. 199, 112397 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Condon, S. M. et al. Birinapant, a Smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies. J. Med. Chem. 57, 3666–3677 (2014).

    Article  CAS  PubMed  Google Scholar 

  52. Bender, A. T. et al. Ability of Bruton’s tyrosine kinase inhibitors to sequester Y551 and prevent phosphorylation determines potency for inhibition of Fc receptor but not B-cell receptor signaling. Mol. Pharmacol. 91, 208 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Blanc, E. et al. Refinement of severely incomplete structures with maximum likelihood in BUSTER-TNT. Acta Crystallogr. D Biol. Crystallogr. 60, 2210–2221 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  CAS  Google Scholar 

  55. Drummond, M. L. & Williams, C. I. In silico modeling of PROTAC-mediated ternary complexes: validation and application. J. Chem. Inf. Model. 59, 1634–1644 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Harder, E. et al. OPLS3: a force field providing broad coverage of drug-like small molecules and proteins. J. Chem. Theory Comput. 12, 281–296 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Watts, K. S. et al. ConfGen: a conformational search method for efficient generation of bioactive conformers. J. Chem. Inf. Model. 50, 534–546 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Bax, A. & Grzesiek, S. Methodological advances in protein NMR. Acc. Chem. Res. 26, 131–138 (1993).

    Article  CAS  Google Scholar 

  59. Bartels, C., Xia, T.-H, Billeter, M., Güntert, P. & Wüthrich, K. The program XEASY for computer-supported NMR spectral analysis of biological macromolecules. J. Biomol. NMR 6, 1–10 (1995).

    Article  CAS  PubMed  Google Scholar 

  60. Keller, R. Optimizing the Process of Nuclear Magnetic Resonance Spectrum Analysis and Computer Aided Resonance Assignment PhD thesis, ETH Zurich (2005).

Download references

Acknowledgements

We thank K. Fennell and P. Montanaro for expression of [15N13C]cIAP1Bir3 used in the pilot NMR experiments, J. Chang for the protein purification schemes of cIAP1Bir3 and biotinylated BTKKD and L. Byrnes and A. Varghese for SEC MALS troubleshooting and analysis. We also thank J. Knafels for crystal preparation before synchrotron shipment and J. Smith for the THP-1 cell culture and degradation protocols. BC5P synthesis was assisted by A. Davies. Compounds BC2P and BCPyr were synthesized by Wuxi PharmaTech. We also thank K. Farley, A. Ratnayake and T. Ryder for assistance with small-molecule NMR analysis. All individuals named are current or former members of Pfizer Worldwide Research and Development, Groton CT, USA.

Author information

Authors and Affiliations

Authors

Contributions

R.H. performed all protein NMR experiments and analyses. Y.M. performed computational modeling and analysis with guidance from Y.C. J.I.M. designed BCPyr and BCPip. X.F. performed native ESI MS and subsequent analysis. S.B. and D.P.U. performed compound synthesis. C.L., Y.X., M.F.B., M.M.H., A.M.G. and M.C.N. contributed to the interpretation of experimental data. J.S. and M.F.C. conceptualized the experimental work and contributed to data interpretation, manuscript drafting and revision. J.S. and M.F.C. performed crystal structure refinement. J.S. performed BLI experiments and analysis with guidance from K.B. J.S. performed cellular degradation assays, in vitro ubiquitination, protein expression and purification, crystallization, SEC MALS and cross-linking studies.

Corresponding author

Correspondence to Matthew F. Calabrese.

Ethics declarations

Competing interests

All authors are current or former employees of Pfizer, Inc.

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 The BC2P Linker is Non-Permissive.

a, Two representative replicate experiments (top and bottom) of dose dependent BTK degradation in THP1 cells and associated DC50 curve-fit. b, Schematic representation of 2-PEG ‘BC2P’ (2), with regions corresponding to BTK ligand (magenta), linker (black) and cIAP1 ligand (blue) indicated. c, Western blot analysis of dose dependent BTK degradation in THP1 cells by BC5P (left), BC2P (right) and associated DC50 curve-fit. The experiment was repeated for a total of 3 independent experiments with reproducible findings (d) Rescue of BC5P dependent degradation using a BTK ligand, and corresponding vinculin loading control with densiometric quantification shown on the left. The experiment was repeated for a total of 5 independent experiments with reproducible findings. Filled circles and hollow triangles represent untreated or BTK ligand treated samples, respectively. Error bars represent ± 1 standard deviation and these data are a result of n=5 independent experiments. Statistical analysis was performed using a two-sided student’s unpaired T Test where significance (*) a p value of 0.022 and 0.033 for 2.5 μM and 0.083 μM treatments, respectively.

Extended Data Fig. 2 Anisotropic vs Isotropic Resolution of BC5P.

Electron density difference map examining ternary complex Pose 2, with BC5P. Anisotropic data (left), and isotropic data (right) with 2Fo-Fc σ = 1.0.

Extended Data Fig. 3 NMR Analysis and Backbone Assignments.

a, Overlay of [15N,1H]-COSY spectra with15N13C cIAP1Bir3 as either apo (red) or an IAP-ligand binary complex (blue). b, [15N,1H]-HSQC spectrum of uniformly 15N,13C-enriched cIAP1Bir3 in complex with the IAP-ligand. Backbone resonance assignments are indicated by the one-letter amino acid code and the sequence number. The backbone cross-peaks of G46 and C62 are outside of the region shown. c, TRO-STE spectra of cIAP1Bir3/BC5P/BTKKD. The relative signal intensity ln(S/So) is plotted versus the square of the field gradient strength, \(G_D^2\). Green triangles and black squares represent data for the apo form of cIAP1Bir3 and for the cIAP1Bir3/BC5P/BTKKD ternary complex, respectively. Translational diffusion coefficients, Dt, as calculated from the diffusion data are indicated.

Extended Data Fig. 4 BC5P Binary Crystal Structure.

a, Electron density difference map examining the binary crystal structure of cIAP1Bir3 and BC5P. Density substantially weakens over the benzyl portion of the tetrahydroisoquinoline, thus preventing further modeling of the linker. Protein is omitted on the right panel for clarity. b, Protein-Ligand interactions between BC5P and cIAP1Bir3 in the binary crystal structure. Dashed lines indicate a set of polar interactions ≤ 3.5 Å in length.

Extended Data Fig. 5 Ternary Complex Kinetics between cIAP1Bir3 and BTKKD.

a, Western blot image of in vitro ubiquitination of BTKKD by cIAP1BUCR1 in the presence of BC5P, or BCPyr. Quantification of mUb and pUb signal was done using ImageJ analysis (v1.4.3.67). b, Ternary complex assay sensorgram of a binary [cIAP1Bir3-BC5P] complex binding to BTKKD, and (c) [cIAP1Bir3-BCPyr] complex binding to BTKKD. For panel B, data was fit globally (left) or independently for Kon and Koff (right) yielding consistent affinity values.

Extended Data Fig. 6 Asymmetric Unit Analysis.

Electron density for the BTKKD/BCPyr/cIAP1Bir3 ternary complex structure covering a portion of the protein-protein interface (proximal to BTKKD R487). Left and middle panels show map (2Fo-Fc contoured at σ = 1.0) for the two copies in the asymmetric unit. Panel on the right shows an overlay of the two copies with density omitted. R487 sidechain is modeled in different conformations in the two copies, projecting toward either one or both aspartic acids presented by cIAP1.

Extended Data Fig. 7 BCPip Design and Characteristics.

a, Top panel: Crystallographic pose of BTKKD (gray), cIAP1Bir3 (orange), and BCPyr (light orange). Bottom panel: Linker substitution modeled to generate Piperidine ‘BCPip’ (4) (purple). b, Schematic representation of BCPip, with regions corresponding to BTK ligand (Magenta), linker (Black) and IAP ligand (Blue) indicated. c, Ternary complex assay sensorgram of a binary [cIAP1BUCR1-BCPip] complex binding to BTKKD. d, Western blot analysis of dose dependent BTK degradation in THP1 cells by BCPip and associated DC50 curve-fit.

Extended Data Fig. 8 Native ESI and SEC MALS Analysis.

a, Native ESI Mass spectrometry showing the spectrum for monomeric cIAP1BUCR1 (Top Panel) or a spectrum for cIAP1BUCR1/ BC5P (Bottom Panel). A Distinct M/Z shift between cIAP1BUCR1 monomer (+) and cIAP1BUCR1 monomer after BC5P (‡) treatment is indicative of a single molecule of BC5P binding to the monomer. The dimer fraction (#) is comprised of 2 molecules of BC5P bound to 2 molecules of cIAP1BUCR1. These data demonstrate a 1:1 or 2:2 stoichiometry of BC5P for cIAP1BUCR1. b, Size Exclusion Chromatography Multi-Angle Light Scattering (SEC MALS) chromatograph of a cIAP1BUCR1/BC5P dimer (red), a lower concentration intermediate (blue), and a DMSO treated cIAP1BUCR1 monomer (black). Calculated molecular weights are shown as squares, change in refractive index (dRI) is shown as dotted lines, light scattering (LS) is shown as solid lines, and expected molecular weights are shown as dashed light blue lines. c, SEC MALS chromatograph of a cIAP1BUCR1/BCPyr dimer (red), and a cIAP1BUCR1/BCPyr/BTKKD dimer of ternary complexes (purple). Calculated molecular weights shown as squares, change in refractive index (dRI) is shown as dotted lines, light scattering (LS) is shown as solid lines, and expected molecular weights are shown as dashed light blue lines. This experiment was performed 3 times with reproducible results.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2, Fig. 1 and Note

Reporting Summary

Source data

Source Data Fig. 1.

Full scan images.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schiemer, J., Horst, R., Meng, Y. et al. Snapshots and ensembles of BTK and cIAP1 protein degrader ternary complexes. Nat Chem Biol 17, 152–160 (2021). https://doi.org/10.1038/s41589-020-00686-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-00686-2

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