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:

Discovery and resistance mechanism of a selective CDK12 degrader

Abstract

Cyclin-dependent kinase 12 (CDK12) is an emerging therapeutic target due to its role in regulating transcription of DNA-damage response (DDR) genes. However, development of selective small molecules targeting CDK12 has been challenging due to the high degree of homology between kinase domains of CDK12 and other transcriptional CDKs, most notably CDK13. In the present study, we report the rational design and characterization of a CDK12-specific degrader, BSJ-4-116. BSJ-4-116 selectively degraded CDK12 as assessed through quantitative proteomics. Selective degradation of CDK12 resulted in premature cleavage and poly(adenylation) of DDR genes. Moreover, BSJ-4-116 exhibited potent antiproliferative effects, alone and in combination with the poly(ADP-ribose) polymerase inhibitor olaparib, as well as when used as a single agent against cell lines resistant to covalent CDK12 inhibitors. Two point mutations in CDK12 were identified that confer resistance to BSJ-4-116, demonstrating a potential mechanism that tumor cells can use to evade bivalent degrader molecules.

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: Development and characterization of CDK12 degrader BSJ-4-116.
Fig. 2: CDK12 degradation preferentially leads to premature cleavage and poly(adenylation) of long genes enriched with DDR genes.
Fig. 3: BSJ-4-116 inhibits the growth of T-ALL cells and sensitizes them to PARP inhibition.
Fig. 4: Chronic exposure leads to acquired resistance to BSJ-4-116 mediated by G-loop mutations.
Fig. 5: Chronic exposure leads to acquired resistance to BSJ-4-116 mediated by G-loop mutations.

Similar content being viewed by others

Data availability

Dataset of exonic nonsynonymous variants excluding loss-of-function mutations in Jurkat-resistant cells is available in Supplementary Dataset 1. KINOMEscan data are provided in Supplementary Dataset 2. A complete GSEA result is provided in Supplementary Dataset 3. Whole-exome sequencing data of parental and resistant cell lines to BSJ-4-116 have been deposited at the National Center for Biotechnology Information (NCBI) Sequence Read Archive with BioProject accession no. PRJNA634900. Poly(A) 3′-sequencing data have been deposited at the NCBI’s Gene Expression Omnibus (accession no. GSE161650). Crystal structure of human CRBN in complex with DDB1 and lenalidomide has a PDB accession no. 4TZ4. Crystal structures of the human CDK12–cyclin K complex have PDB accession nos. 5ACB, 6CKX and 6B3E. Crystal structure of the human CDK13–cyclin K complex has PDB accession no. 5EFQ. Source data are provided with this paper.

References

  1. Hydbring, P., Malumbres, M. & Sicinski, P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat. Rev. Mol. Cell Biol. 17, 280–292 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Shiekhattar, R. et al. Cdk-activating kinase complex is a component of human transcription factor TFIIH. Nature 374, 283–287 (1995).

    Article  CAS  PubMed  Google Scholar 

  3. Fisher, R. P. Secrets of a double agent: CDK7 in cell-cycle control and transcription. J. Cell Sci. 118, 5171–5180 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Galbraith, M. D., Bender, H. & Espinosa, J. M. Therapeutic targeting of transcriptional cyclin-dependent kinases. Transcription 10, 118–136 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Gajduskova, P. et al. CDK11 is required for transcription of replication-dependent histone genes. Nat. Struct. Mol. Biol. https://doi.org/10.1038/s41594-020-0406-8 (2020).

  6. Ramanathan, Y. et al. Three RNA polymerase II carboxyl-terminal domain kinases display distinct substrate preferences. J. Biol. Chem. 276, 10913–10920 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Davidson, L., Muniz, L. & West, S. 3′ end formation of pre-mRNA and phosphorylation of Ser2 on the RNA polymerase II CTD are reciprocally coupled in human cells. Genes Dev. 28, 342–356 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bartkowiak, B. et al. CDK12 is a transcription elongation-associated CTD kinase, the metazoan ortholog of yeast Ctk1. Genes Dev. 24, 2303–2316 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Blazek, D. et al. The cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev. 25, 2158–2172 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, X., Chatterjee, N., Spirohn, K., Boutros, M. & Bohmann, D. Cdk12 is a gene-selective RNA polymerase II kinase that regulates a subset of the transcriptome, including Nrf2 target genes. Sci. Rep. 6, 21455 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Iniguez, A. B. et al. EWS/FLI confers tumor cell synthetic lethality to CDK12 inhibition in Ewing sarcoma. Cancer Cell 33, 202–216.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Dubbury, S. J., Boutz, P. L. & Sharp, P. A. CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature 564, 141–145 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Krajewska, M. et al. CDK12 loss in cancer cells affects DNA damage response genes through premature cleavage and polyadenylation. Nat. Commun. 10, 1757 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Negrini, S., Gorgoulis, V. G. & Halazonetis, T. D. Genomic instability—an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220–228 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Lui, G. Y. L., Grandori, C. & Kemp, C. J. CDK12: an emerging therapeutic target for cancer. J. Clin. Pathol. 71, 957–962 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bayles, I. et al. Ex vivo screen identifies CDK12 as a metastatic vulnerability in osteosarcoma. J. Clin. Invest. 129, 4377–4392 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Bradner, J. E., Hnisz, D. & Young, R. A. Transcriptional addiction in cancer. Cell 168, 629–643 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Paculova, H. et al. BRCA1 or CDK12 loss sensitizes cells to CHK1 inhibitors. Tumour Biol. 39, 1010428317727479 (2017).

  19. Zhang, T. et al. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat. Chem. Biol. 12, 876–884 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gao, Y. et al. Overcoming resistance to the THZ series of covalent transcriptional CDK inhibitors. Cell Chem. Biol. 25, 135–142 e135 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Johannes, J. W. et al. Structure-based design of selective noncovalent CDK12 inhibitors. ChemMedChem 13, 231–235 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Ito, M. et al. Discovery of 3-benzyl-1-(trans-4-((5-cyanopyridin-2-yl)amino)cyclohexyl)-1-arylurea derivatives as novel and selective cyclin-dependent kinase 12 (CDK12) inhibitors. J. Med. Chem. 61, 7710–7728 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Quereda, V. et al. Therapeutic targeting of CDK12/CDK13 in triple-negative breast cancer. Cancer Cell 36, 545–558 e547 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Fan, Z. et al. CDK13 cooperates with CDK12 to control global RNA polymerase II processivity. Sci. Adv. 6, https://doi.org/10.1126/sciadv.aaz5041 (2020).

  25. Liang, K. et al. Characterization of human cyclin-dependent kinase 12 (CDK12) and CDK13 complexes in C-terminal domain phosphorylation, gene transcription, and RNA processing. Mol. Cell Biol. 35, 928–938 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Greifenberg, A. K. et al. Structural and functional analysis of the Cdk13/cyclin K complex. Cell Rep. 14, 320–331 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Olson, C. M. et al. Pharmacological perturbation of CDK9 using selective CDK9 inhibition or degradation. Nat. Chem. Biol. 14, 163–170 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Jiang, B. et al. Development of dual and selective degraders of cyclin-dependent kinases 4 and 6. Angew. Chem. Int. Ed. Engl. 58, 6321–6326 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Brand, M. et al. Homolog-selective degradation as a strategy to probe the function of CDK6 in AML. Cell Chem. Biol. 26, 300–306 e309 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Salami, J. et al. Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance. Commun. Biol. 1, 100 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sun, Y. et al. Degradation of Bruton’s tyrosine kinase mutants by PROTACs for potential treatment of ibrutinib-resistant non-Hodgkin lymphomas. Leukemia 33, 2105–2110 (2019).

    Article  PubMed  Google Scholar 

  32. Dobrovolsky, D. et al. Bruton tyrosine kinase degradation as a therapeutic strategy for cancer. Blood 133, 952–961 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cooper, J. M. et al. Overcoming BET inhibitor resistance in malignant peripheral nerve sheath tumors. Clin. Cancer Res. 25, 3404–3416 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cheng, M. et al. Discovery of potent and selective epidermal growth factor receptor (EGFR) bifunctional small-molecule degraders. J. Med. Chem. 63, 1216–1232 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Burslem, G. M. & Crews, C. M. Small-molecule modulation of protein homeostasis. Chem. Rev. 117, 11269–11301 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Watt, G. F., Scott-Stevens, P. & Gaohua, L. Targeted protein degradation in vivo with proteolysis targeting chimeras: current status and future considerations. Drug Discov. Today Technol. 31, 69–80 (2019).

    Article  PubMed  Google Scholar 

  37. Burslem, G. M. & Crews, C. M. Proteolysis-targeting chimeras as therapeutics and tools for biological discovery. Cell 181, 102–114 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang, H. T. et al. A chemoproteomic approach to query the degradable kinome using a multi-kinase degrader. Cell Chem. Biol. 25, 88–99.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Choi, S. H. et al. CDK12 phosphorylates 4E-BP1 to enable mTORC1-dependent translation and mitotic genome stability. Genes Dev. 33, 418–435 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hafner, M., Niepel, M., Chung, M. & Sorger, P. K. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat. Methods 13, 521–527 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bosken, C. A. et al. The structure and substrate specificity of human Cdk12/cyclin K. Nat. Commun. 5, 3505 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Hopkins, A. L., Groom, C. R. & Alex, A. Ligand efficiency: a useful metric for lead selection. Drug Discov. Today 9, 430–431 (2004).

    Article  PubMed  Google Scholar 

  43. Chirackal Manavalan, A. P. et al. CDK12 controls G1/S progression by regulating RNAPII processivity at core DNA replication genes. EMBO Rep. 20, e47592 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zhang, L., Riley-Gillis, B., Vijay, P. & Shen, Y. Acquired resistance to BET-PROTACs (proteolysis-targeting chimeras) caused by genomic alterations in core components of E3 ligase complexes. Mol. Cancer Ther. 18, 1302–1311 (2019).

    Article  PubMed  Google Scholar 

  45. Song, Y. et al. A dynamic view of the proteomic landscape during differentiation of ReNcell VM cells, an immortalized human neural progenitor line. Sci. Data 6, 190016 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Wessel, D. & Flügge, U. I. A method for the quantitative recovery of protein in dilute solution in the presence of detergents and lipids. Anal. Biochem. 138, 141–143 (1984).

    Article  CAS  PubMed  Google Scholar 

  47. Ting, L., Rad, R., Gygi, S. P. & Haas, W. MS3 eliminates ratio distortion in isobaric multiplexed quantitative proteomics. Nat. Methods 8, 937–940 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. McAlister, G. C. et al. MultiNotch MS3 enables accurate, sensitive, and multiplexed detection of differential expression across cancer cell line proteomes. Anal. Chem. 86, 7150–7158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Paulo, J. A. et al. Quantitative mass spectrometry-based multiplexing compares the abundance of 5000 S. cerevisiae proteins across 10 carbon sources. J. Proteomics 148, 85–93 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Eng, J. K., McCormack, A. L. & Yates, J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass. Spectrom. 5, 976–989 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Bieniossek, C., Imasaki, T., Takagi, Y. & Berger, I. MultiBac: expanding the research toolbox for multiprotein complexes. Trends Biochem. Sci. 37, 49–57 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Kostic for her editing of this manuscript. This work was supported by a National Institutes of Health grant (nos. PO1 CA154303 to N.S.G. and U24-DK116204 to P.K.S.) and Deutsche Forschungsgemeinschaft GE 976/9-2 (to M.K.).

Author information

Authors and Affiliations

Authors

Contributions

N.S.G. and T.Z. conceived the project. B.J. performed the compound synthesis and structure determination with help from I.Y. Y.G. and W. L. executed cellular biological experimental research with help from J.J. K.R and D.D. performed the NanoBRET ternary complex assays. J.C. executed computational modeling, whole-exome sequencing analysis and mutational experiments design. R.D. and Y.G. performed genomic data analysis. I.K and M.G. executed CDK12 in vitro kinase assay. M.B. M.K and P.K.S. performed proteomic analysis. T.Z. Y.G. J.C. B.J. and N.S.G. co-wrote the paper. All authors edited the manuscript.

Corresponding authors

Correspondence to Tinghu Zhang or Nathanael S. Gray.

Ethics declarations

Competing interests

N.S.G. is a founder, science advisory board member and equity holder in Gatekeeper, Syros, Petra, C4, B2S, Aduro and Soltego. The Gray lab receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Janssen, Kinogen, Voronoi, Her2llc, Deerfield, Ephiphanes and Sanofi. J.C. is a consultant to Soltego, Jengu and Allorion, and equity holder for Soltego, Allorion and M3 bioinformatics & technology Inc. P.K.S. is a member of the science advisory board or board of directors of Merrimack Pharmaceutical, Glencoe Software, Applied Biomath and RareCyte Inc., and has equity in these companies. B.J., J.C., Y.G., N.K., T.Z. and N.S.G. are inventors on CDK12 degrader patents. All remaining authors declare no competing interests.

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 Development of CDK12 degraders BSJ-4-23 and BSJ-4-116.

a, Chemical structures of THZ531 and its 3 fragments with ligand efficiency values. b, In vitro CDK12 kinase assay. Assays were performed at an ATP concentration of 30 μM (apparent Km). Data are presented as mean ± s.d. of n = 3 biologically independent samples. c, Preliminary screening immunoblots for CDK12, CDK13 and β-Actin in Jurkat cells after 6 h treatment with DMSO or different CDK12 degraders at the indicated concentrations. d, Binding groove for BSJ-4-23 in modeled ternary complex of CDK12/BSJ-4-23/CRBN (CDK12 in blue, PDB ID: 5ACB, CRBN in orange, PDB ID: 4TZ4, BSJ-4-23 carbons in light grey). e, Time-dependent effect of BSJ-4-116 (50 nM) on CDK12, CDK13 and cyclin K protein levels after 2 h, 4 h, 8 h, 16 h and 24 h treatment in Jurkat cells. f, Left: Immunoblots for CDK12, CRBN and β-Actin in WT and CRBN null Jurkat cells after 6 h treatment with DMSO, BSJ-4-23 (250 nM) and BSJ-4-116 (50 nM); Right: Immunoblots for CDK12 and α-tubulin in Jurkat cells following 2 h pre-treatment with DMSO, Carfizomib (400 nM), MLN4924 (1000 nM), Thalidomide (1000 nM) and THZ531 (250 nM) followed by 6 h co-treatment with DMSO or BSJ-4-116 (50 nM). g, KinomeScan kinase selectivity profile for BSJ-4-116. BSJ-4-116 was profiled at a concentration of 1 μM against a panel of 468 human kinases. The results for the binding interactions are reported as a percent of the DMSO control (% control), where larger red circles indicate stronger binding hits. The selectivity score was defined as the ratio of the number of kinases inhibited to a specified percentage versus the total number of kinases. For this experiment, specified percent inhibition was set at 10%, resulting in S(10) value of 0.017 for BSJ-4-116. h, Degradation effect of BSJ-4-116 and BSJ-4-116NC at indicated doses prechecked by western blots for the proteomics experiment in Jurkat cells. Data in (c), (e), (f) and (h) represent n=2 independent experiments. i, NanoBRET live cell ternary complex assays performed in MOLT-4 cells co-expressing HaloTag-CRBN and one of the following C-terminal NanoLuc fusions: CDK12, CDK12 (K745R), CDK12 (L752M), CDK12 (K745R/L752M) or CDK13, CDK13 (R723K), CDK13 (M730L), CDK13 (R723K/M730L). The fold increase in NanoBRET signal relative to BSJ-4-116NC was plotted after 3 h treatment with the indicated compounds with n=6 biologically independent samples.

Source data

Extended Data Fig. 2 CDK12 degradation preferentially leads to premature cleavage and polyadenylation (PCPA) of long genes enriched with DDR genes.

a, Genome-wide correlation analysis for replicates from each condition showing significant correlation between BSJ-4-116 vs THZ531, and DMSO vs BSJ-4-116NC. b, Immunoblots for CDK12 and α-tubulin in Jurkat cells treated with DMSO or BSJ-4-116 (50 nM) for indicated hours. Data are representative of n=2 independent experiments. c, Fisher exact test showing significant overlap in genes downregulated by BSJ-4-116 vs THZ531 (p=0). There was also significant overlap in the small numbers of gene upregulated (p=1.42e-136). d, GSEA of downregulated genes in Jurkat cells treated with BSJ-4-116 and THZ531. e, Additional enriched GSEA signatures enriched by BSJ-4-116 treatment. f, Left: qRT-PCR analysis of the indicated DDR gene expression in Jurkat and MOLT4 cells treated with BSJ-4-116 (50 nM) or BSJ-4-116NC (100 nM) for 10 h. Data were normalized to GAPDH and compared to DMSO-treated controls (n=3). Right: Immunoblots for indicated DDR and cell death markers in Jurkat and MOLT4 cells treated with DMSO or BSJ-4-116 (50 nM) for indicated hours. Data are representative of n=2 independent experiments. g, Bar plot showing the frequency of retrieved polyadenylation site (PAS) motifs 100bp upstream of the poly(A) 3’-seq peaks. h, Average metagene profiles of normalized poly(A) 3’-seq reads over gene bodies and extending –2 to +2 kb of all detected genes in Jurkat cells treated with BSJ-4-116 (50 nM) or THZ531 (250 nM) vs DMSO for 8 h. Sense and antisense reads are depicted by solid and dashed lines, respectively. i, Boxplots showing the differential usage (log2 fold-change) of polyadenylation sites at three different genomic locations. The comparison BSJ-4-116 vs. BSJ-4-116NC is shown in red and THZ531 vs. DMSO is shown in green. j, Schematic illustration of PCPA caused by CDK12 inhibition or degradation.

Source data

Extended Data Fig. 3 BSJ-4-116 inhibits the growth of T-ALL cells and sensitizes them to PARP inhibition.

a, Cell-cycle analysis of Jurkat and MOLT4 cells treated with BSJ-4-116 (50 nM) and BSJ-4-116NC (100 nM) for 24 h. DNA was stained with propidium iodide (PI) before flow cytometry analysis. G/M% values are presented as mean ± s.d. of n=3 biologically independent samples and are representative of n=2 independent experiments. b, Excess over Bliss synergy plots for serial dilutions of BSJ-4-116 in combination with Olaparib in CRBN null Jurkat (top) and MOLT4 (bottom) cells. n=3 replicates.

Extended Data Fig. 4 Chronic exposure leads to acquired resistance to BSJ-4-116 mediated by G-loop mutations.

a, Dose response curves for parental and resistant Jurkat and MOLT4 cells treated with BSJ-4-23 at indicated dose range for 72 h. Percent cell growth relative to DMSO-treated was analyzed using growth rate inhibition assay method. Data are presented as mean ± s.d. of n=3 biologically independent samples. b, Detection of heterozygous G739S mutation in Jurkat resistant cells. DNA chromatograms of sanger sequencing shows region of mutation from PCR-amplified CDK12 cDNA. c, Immunoblots for CDK12, CDK9 and GAPDH in parental and resistant Jurkat and MOLT4 cells treated with DMSO, BSJ-4-23 (250 nM), BSJ-4-116 (50 nM) or THAL-SNS-032 (250 nM) for 8 h. Data represent n=2 independent experiments. d, CDK12 kinase domain structure (PDB code: 5ACB) showing the locations of G-loop mutations I733 and G739.

Source data

Supplementary information

Supplementary Information

Supplementary Note Synthetic procedures.

Reporting Summary

Supplementary Data 1

Exonic nonsynonymous variants excluding loss of function mutations in Jurkat-resistant cells.

Supplementary Data 2

Full KINOMEscan dataset.

Supplementary Data 3

Complete GSEA result.

Supplementary Data 4

Compound structures in Fig. 1a.

Source data

Source Data Fig. 1

Uncropped western blots for Fig. 1.

Source Data Fig. 3

Uncropped western blots for Fig. 3.

Source Data Fig. 4

Uncropped western blots for Fig. 4.

Source Data Extended Data Fig. 1

Uncropped western blots for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Uncropped western blots for Extended Data Fig. 2.

Source Data Extended Data Fig. 4

Uncropped western blots for Extended Data Fig. 4.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiang, B., Gao, Y., Che, J. et al. Discovery and resistance mechanism of a selective CDK12 degrader. Nat Chem Biol 17, 675–683 (2021). https://doi.org/10.1038/s41589-021-00765-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00765-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer