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 of a first-in-class CDK2 selective degrader for AML differentiation therapy

Abstract

The discovery of effective therapeutic treatments for cancer via cell differentiation instead of antiproliferation remains a great challenge. Cyclin-dependent kinase 2 (CDK2) inactivation, which overcomes the differentiation arrest of acute myeloid leukemia (AML) cells, may be a promising method for AML treatment. However, there is no available selective CDK2 inhibitor. More importantly, the inhibition of only the enzymatic function of CDK2 would be insufficient to promote notable AML differentiation. To further validate the role and druggability of CDK2 involved in AML differentiation, a suitable chemical tool is needed. Therefore, we developed first-in-class CDK2-targeted proteolysis-targeting chimeras (PROTACs), which promoted rapid and potent CDK2 degradation in different cell lines without comparable degradation of other targets, and induced remarkable differentiation of AML cell lines and primary patient cells. These data clearly demonstrated the practicality and importance of PROTACs as alternative tools for verifying CDK2 protein functions.

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: The design of PROTAC compounds and their potencies in vitro.
Fig. 2: Potent and selective CDK2 degradation induced by CPS2.
Fig. 3: CDK2 degradation and AML cell differentiation triggered by CPS2.
Fig. 4: The antileukemic effects of CPS2 are mediated by CDK2 degradation.
Fig. 5: Transcriptome changes in AML cells following CPS2 and shCDK2 treatment.
Fig. 6: Effects of CPS2 on primary AML cell and HSC differentiation.

Similar content being viewed by others

Data availability

All raw gels are included in source data files. The source structure of CDK2 and CRBN can be accessed via PDB IDs-4CI3 and 3PY0 (http://www.rcsb.org/). The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org) via the iProX partner repository46 with the dataset identifier PXD021592. The raw sequence data reported in this paper have been deposited in the Genome Sequence Archive47 in National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences48, under accession number CRA003240, which is publicly accessible at https://bigd.big.ac.cn/gsa. Source data are provided with this paper.

References

  1. Dohner, H., Weisdorf, D. J. & Bloomfield, C. D. Acute myeloid leukemia. N. Engl. J. Med. 373, 1136–1152 (2015).

    Article  PubMed  CAS  Google Scholar 

  2. Shih, A. H. et al. AG-221, a small molecule mutant IDH2 inhibitor, remodels the epigenetic state of IDH2-mutant cells and induces alterations in self-renewal/differentiation in IDH2-mutant AML model in vivo. Blood 124, 437–437 (2014).

    Article  Google Scholar 

  3. Zarrinkar, P. P. et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood 114, 2984–2992 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zimmerman, E. I. et al. Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia. Blood 122, 3607–3615 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zeidan, A. M. et al. Clinical benefit of glasdegib in combination with azacitidine or low-dose cytarabine in patients with acute myeloid leukemia. Blood 134, 3916–3916 (2019).

    Article  Google Scholar 

  6. Wei, A. et al. Safety and efficacy of venetoclax plus low-dose cytarabine in treatment-naive patients aged ≥65 years with acute myeloid leukemia. Blood 128, 102–102 (2016).

    Article  Google Scholar 

  7. Scappini, B. et al. Cytarabine and clofarabine after high-dose cytarabine in relapsed or refractory AML patients. Am. J. Hematol. 87, 1047–1051 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Ryningen, A. et al. In vivo biological effects of ATRA in the treatment of AML. Expert Opin. Investig. Drugs 17, 1623–1633 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Kelly, L. M. et al. PML/RARα and FLT3-ITD induce an APL-like disease in a mouse model. Proc. Natl Acad. Sci. USA 99, 8283–8288 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ohnishi, K. PML-RARα inhibitors (ATRA, tamibaroten, arsenic trioxide) for acute promyelocytic leukemia. Int. J. Clin. Oncol. 12, 313–317 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. de Botton, S. et al. Early onset of chemotherapy can reduce the incidence of ATRA syndrome in newly diagnosed acute promyelocytic leukemia (APL) with low white blood cell counts: results from APL 93 trial. Leukemia 17, 339–342 (2003).

    Article  PubMed  CAS  Google Scholar 

  12. Ying, M. et al. Ubiquitin-dependent degradation of CDK2 drives the therapeutic differentiation of AML by targeting PRDX2. Blood 131, 2698–2711 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Berthet, C., Aleem, E., Coppola, V., Tessarollo, L. & Kaldis, P. Cdk2 knockout mice are viable. Curr. Biol. 13, 1775–1785 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Takada, M. et al. FBW7 loss promotes chromosomal instability and tumorigenesis via cyclin E1/CDK2-mediated phosphorylation of CENP-A. Cancer Res. 77, 4881–4893 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tadesse, S., Caldon, E. C., Tilley, W. & Wang, S. Cyclin-dependent kinase 2 inhibitors in cancer therapy: an update. J. Med. Chem. 62, 4233–4251 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Fujimoto, T., Anderson, K., Jacobsen, S. E., Nishikawa, S. I. & Nerlov, C. Cdk6 blocks myeloid differentiation by interfering with Runx1 DNA binding and Runx1-C/EBPα interaction. EMBO J. 26, 2361–2370 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Neklesa, T. K., Winkler, J. D. & Crews, C. M. Targeted protein degradation by PROTACs. Pharmacol. Ther. 174, 138–144 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bai, L. et al. A potent and selective small-molecule degrader of STAT3 achieves complete tumor regression in vivo. Cancer Cell 36, 498–511.e17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Luo, M. Current chemical biology approaches to interrogate protein methyltransferases. ACS Chem. Biol. 7, 443–463 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Su, S. et al. Potent and preferential degradation of CDK6 via proteolysis targeting chimera degraders. J. Med. Chem. 62, 7575–7582 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rana, S. et al. Selective degradation of CDK6 by a palbociclib based PROTAC. Bioorg. Med. Chem. Lett. 29, 1375–1379 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Brand, M. et al. Homolog-selective degradation as a strategy to probe the function of CDK6 in AML. Cell Chem. Biol. 26, 300–306.e9 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. 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 

  26. Zhou, F. et al. Development of selective mono or dual PROTAC degrader probe of CDK isoforms. Eur. J. Med. Chem. 187, 111952 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Lin, R. et al. 1-Acyl-1H-[1,2,4]triazole-3,5-diamine analogues as novel and potent anticancer cyclin-dependent kinase inhibitors: synthesis and evaluation of biological activities. J. Med. Chem. 48, 4208–4211 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Emanuel, S. et al. The in vitro and in vivo effects of JNJ-7706621: a dual inhibitor of cyclin-dependent kinases and aurora kinases. Cancer Res. 65, 9038–9046 (2005).

    Article  CAS  PubMed  Google Scholar 

  29. Huang, S., Connolly, P. J., Lin, R., Emanuel, S. & Middleton, S. A. Synthesis and evaluation of N-acyl sulfonamides as potential prodrugs of cyclin-dependent kinase inhibitor JNJ-7706621. Bioorg. Med. Chem. Lett. 16, 3639–3641 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. de The, H. Differentiation therapy revisited. Nat. Rev. Cancer 18, 117–127 (2018).

    Article  PubMed  CAS  Google Scholar 

  31. Pettersson, M. & Crews, C. M. PROteolysis TArgeting Chimeras (PROTACs)—past, present and future. Drug Discov. Today Technol. 31, 15–27 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. 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 

  34. Bondeson, D. P. et al. Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem. Biol. 25, 78–87.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Grishina, I. & Lattes, B. A novel Cdk2 interactor is phosphorylated by Cdc7 and associates with components of the replication complexes. Cell Cycle 4, 4120–4126 (2005).

    Article  Google Scholar 

  36. Chunder, N., Wang, L., Chen, C., Hancock, W. W. & Wells, A. D. Cyclin-dependent kinase 2 controls peripheral immune tolerance. J. Immunol. 189, 5659–5666 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Saurus, P. et al. Cyclin-dependent kinase 2 protects podocytes from apoptosis. Sci. Rep. 6, 21664 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Granes, F., Roig, M. B., Brady, H. J. & Gil-Gomez, G. Cdk2 activation acts upstream of the mitochondrion during glucocorticoid induced thymocyte apoptosis. Eur. J. Immunol. 34, 2781–2790 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Teitz, T. et al. CDK2 inhibitors as candidate therapeutics for cisplatin- and noise-induced hearing loss. J. Exp. Med. 215, 1187–1203 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hsu, A. Y. et al. Phenotypical microRNA screen reveals a noncanonical role of CDK2 in regulating neutrophil migration. Proc. Natl Acad. Sci. USA 116, 18561–18570 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Drayson, M. T., Michell, R. H., Durham, J. & Brown, G. Cell proliferation and CD11b expression are controlled independently during HL60 cell differentiation initiated by 1,25α-dihydroxyvitamin D(3) or all-trans-retinoic acid. Exp. Cell Res. 266, 126–134 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Chou, T. C. Drug combination studies and their synergy quantification using the Chou–Talalay method. Cancer Res. 70, 440–446 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Shao, X. et al. CDK2 suppression synergizes with all-trans-retinoic acid to overcome the myeloid differentiation blockade of AML cells. Pharmacol. Res. https://doi.org/10.1016/j.phrs.2019.104545 (2020).

  44. Jiang, X. et al. Proteomic analysis of eIF5B silencing-modulated proteostasis. PLoS ONE 11, e0168387 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Ma, J. et al. iProX: an integrated proteome resource. Nucleic Acids Res. 47, D1211–D1217 (2019).

    Article  PubMed  Google Scholar 

  47. Wang, Y. et al. GSA: genome sequence archive. Genomics Proteomics Bioinformatics 15, 14–18 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. National Genomics Data Center, M., Partners Database resources of the National Genomics Data Center in 2020. Nucleic Acids Res. 48, D24–D33 (2020).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge and thank the patients who provided the primary samples for this research. The laboratory of Q. Wang, Beijing Institute Genomics, Chinese Academy of Science kindly donated MV-4-11, OCI-AML2, OCI-AML3 and Kasumi-1 cell lines. MOLT-4 cells were provided by Stem Cell Bank, Chinese Academy of Sciences. NB4 cells were provided by L. Wu, University of Southern California. We acknowledge them for their support for this work. We acknowledge Q. Tao, Z. Yang, H. Yang, W. Lv, S. Zhu, C. Yang, X. Yang, Y. Su, J. Wang, Y. Zhang and C. Qian for their help. This work was supported by the National Natural Science Foundation of China (nos. 81573277, 81622042 and 81773567), National Major Scientific and Technological Special Project for ‘Significant New Drugs Development’ (nos. SQ2017ZX095003 and 2018ZX09711001) and ‘National Key R&D Program of China’ (no. 2020YFE0202200).

Author information

Authors and Affiliations

Authors

Contributions

L.W., X. Shao, T.Z., Y.W. and A.X. contributed equally to this work. Y.R. and M.Y. designed the project. L.W., X.Shao, Y.R. and M.Y. wrote the paper. X. Sun helped organize the paper. L.W., Y.L. and Y.W. designed, simulated and synthesized the molecules. Y.W. and T. Lan helped synthesize the molecules. L.W., T.Z. and X.T. performed western blot procedures, cell proliferation experiments and acute toxicity experiments in vivo. X. Shao and A.X. examined the western blot, cell proliferation, cell differentiation effect and cell apoptosis in AML. Y.T. and H.G. helped prepare the samples for western blot. X. Shao, A.X., W.D., W.W. and Y.C. performed and analyzed RNA-sequencing data, collected patient primary cell samples and performed HSC toxicity experiments. X. Shao, A.X., W.D. and W.W. performed the target validation experiment. T. Li, X.M. and H.D. helped to prepare samples for proteomics and results analysis. B.Y. and Q.H. provided support for some experiments.

Corresponding authors

Correspondence to Meidan Ying or Yu Rao.

Ethics declarations

Competing interests

The 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 Ternary complexes model of a POI, PROTAC and E3 ligase.

Ternary complexes model of a POI, PROTAC and E3 ligase. Ternary complex formation is mediated by PROTAC technology. Apo CDK2 protein (PDB ID: 3PY0) and CRBN (PDB ID: 4CI3) are correspondingly shown as the ribbon and surface in the model. The ternary complex model was based on docking and molecular dynamics simulations with Schrodinger and Desmond software.

Extended Data Fig. 2 Acute toxicity and cell proliferation inhibition of CPS2.

Acute toxicity and cell proliferation inhibition of CPS2. a, Acute toxicity experiment. Vehicle, 400 mg/kg CPS2 and 400 mg/kg J2 were independently intraperitoneally administered into mice at a single dose. The survival time and body weight were recorded (n = 6 for each group). For body weight, ***: p<0.001; J2 vs. Control. #: p<0.05, ##: p<0.01; CPS2 vs. Control. b, Cell proliferation assay of NB4 cells treated with several concentrations of CPS2 and J2 for the indicated times. **: p<0.01, ***: p<0.001; CPS2 or J2 vs. Control. c, Cells apoptosis experiment. PI/Annexin V apoptosis assay of the NB4 cells treated with several concentrations of CPS2 and J2 for the indicated times. *: p<0.05, **: p<0.01, ***: p<0.001; CPS2 or J2 vs. Control. b-c, All experiments were conducted with three replicas. a-c, The error bars represent the means±s.d. The significance analysis was conducted by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 3 Synergetic effect of CPS2 with conventional inhibitors.

Synergetic effect of CPS2 with conventional inhibitors. a-c, In MV-4-11 cell line, CPS2 could enhance the effect of Copanlisib and Palbociclib. a, Cell viability assessment under CPS2 and Copanlisib treatment. For the combo curve, CPS2 was used as indicated concentration with 100 nM Copanlisib. CPS2 and Copanlisib independently were used as control. 5000 MV-4-11 cells were seeded into 96-wells plates. The cells were treated for 3 days. b, Cell viability assessment under CPS2 and Palbociclib treatment. For the combo curve, CPS2 was used as indicated concentration with 120 nM Palbociclib. CPS2 and Palbociclib independently were used as control. 5000 MV-4-11 cells were seeded into 96-wells plates. The cells were treated for 3 days. c, FA-CI curve of CPS2 combo treatment. FA-CI is calculated by CompuSyn (Ver. 1.0) based on the data in (a) and (b). The curve is draw by GraphPad prism 7, nonlinear regression (curve fit). d, Cell viability assessment under CPS2 and Palbociclib treatment in Kasumi-1 cells. For the combo curve, CPS2 was used as indicated concentration with 100 nM Palbociclib. CPS2 and Palbociclib independently were used as control. 5000 Kasumi-1 cells were seeded into 96-wells plates. The cells were treated for 3 days. a-d, The data are individual replicate values and shown as means±s.d. (n= 3 for biologically independent samples per group).

Extended Data Fig. 4 The effect of CPS2 on CDK2 degradation and AML cell differentiation.

The effect of CPS2 on CDK2 degradation and AML cell differentiation. a, Protein expression levels of different CDKs after CPS2 treatment for the indicated times in U937 and HL60 cells. b, Effects of CPS2 on the protein level of CDK2 in AML cells. NB4 and U937 cells were treated with the indicated concentrations of CPS2 for 3 h and 6 h. **: p<0.01, ***: p<0.001; CPS2 vs. the Control. c-d, Cell differentiation analysis of HL60 cells treated with different concentrations of CPS2 for the indicated times. c, CD11b expression. **: p<0.01, ***: p<0.001; CPS2 vs. the Control. d, Cell morphological analysis (RG) and NBT-reducing activity assay (NBT). Left: Representative images of RG and NBT assays. Right: The statistical data from the NBT assay. ***: p<0.001; CPS2 vs. the Control. a-d, The western-blot and images are representative of minimally three independent experiments. The data are individual replicate values and shown as means±s.d. (n= 3 for biologically independent samples per group).

Source data

Extended Data Fig. 5 Effects of AURKA and IKZF1 inhibition on AML cells differentiation.

Effects of AURKA and IKZF1 inhibition on AML cells differentiation. NB4 cells were infected with lentivirus-shAURKA (#1 or #2) or shIKZF1 (#1 or #2) for indicated times. shCDK2 #2 as positive group. a, The silencing efficiency of shRNA against AURKA for 3 days. **: p<0.01, ***: p<0.001; shRNA vs. shCtrl. b, The silencing efficiency of shRNA against IKZF1 for 3 days. ***: p<0.001; shRNA vs. shCtrl. c, CD11b expression. **: p<0.01, ***: p<0.001; shRNA vs. shCtrl. d, NBT-reducing activity assay (NBT). Left: the representative images of NBT assays. Right: The statistical data of NBT assay. *, p<0.05; ***: p<0.001; shRNA vs. shCtrl. e, Cell morphological analysis. f, The overexpression of IKZF1 in NB4 cells. ***, p<0.001; pCLS vs. IKZF1. All data was shown as mean±s.d., n = 3. Other data are representative of at least three individual experiments and one representative image is shown. The significance analysis was conducted by two-tailed unpaired Student’s t test.

Source data

Extended Data Fig. 6 The protein level of CDK2 in AML cells following CPS2 and shCDK2 treatment.

The protein level of CDK2 in AML cells following CPS2 and shCDK2 treatment. a, Protein expression levels of CDK2 after CPS2 treatment for the indicated times in NB4 cells. b, Western-blot analysis of CDK2 expression in NB4 cells transduced with different shRNAs (#1 and #2) against CDK2 for the indicated times. a-b, The gels are representative of minimally three biologically independent experiments. The error bars represent the mean±s.d. of n=3 in biologically duplicates. The significance analysis was conducted by two-tailed unpaired Student’s t-test. (a) ***: p<0.001. Each time point group vs. 0 h group. (b) ***: p<0.001. Each shCDK2 group as indicated vs. shCtrl.

Source data

Extended Data Fig. 7 Schematic representation of comparing gene expression profiles of between CDK2 decline induced by CPS2, shCDK2 and CDK2 inhibition driven by J2.

Schematic representation of comparing gene expression profiles of between CDK2 decline induced by CPS2, shCDK2 and CDK2 inhibition driven by J2. Red circles reflect the differential genes between CPS2 and DMSO, blue circles reflect the overlay differential genes between shCDK2 (#1 & #2) and shCtrl, green circles reflect the differential genes between J2 and DMSO. The overlay shows the shared differential genes between (a) CPS2 (12 h) & shCDK2 (D2.5) & J2 (12 h) and (b) CPS2 (24 h) & shCDK2 (D2.5) & J2 (24 h). The yellow box showed the genes responding to CPS2 and shCDK2, but not J2; The green box showed the genes responding to CPS2, shCDK2 and J2. The differentiation-related genes are highlighted with red.

Extended Data Fig. 8 CPS2 induces primary AML cell differentiation.

CPS2 induces primary AML cell differentiation. a-c, Primary AML samples collected at diagnosis from the peripheral blood of patients were treated with CPS2 for 7 days. a, CD11b expression. b, The protein level of CDK2. c, Cell morphological analysis.

Source data

Supplementary information

Supplementary Information

Supplementary Notes 1 and 2, Figs. 1–8 and Tables 1–5.

Reporting Summary

Supplementary Data 1

The affinity of CPS2 with representative targets.

Supplementary Data 2

Protein level change after CPS2 treatment.

Supplementary Data 3

List of overlapped genes in NB4 cells in response to CPS2

Supplementary Data 4

Three clusters from Metascape pathway enrichment analysis of the changed genes in response to CPS2 and lentivirus-shRNA-CDK2.

Supplementary Data 5

P value of each supplementary figure.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Shao, X., Zhong, T. et al. Discovery of a first-in-class CDK2 selective degrader for AML differentiation therapy. Nat Chem Biol 17, 567–575 (2021). https://doi.org/10.1038/s41589-021-00742-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00742-5

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