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:

Cellular and Molecular Biology

The novel CDK9 inhibitor, XPW1, alone and in combination with BRD4 inhibitor JQ1, for the treatment of clear cell renal cell carcinoma

Abstract

Background

Clear cell renal cell carcinoma (ccRCC) is a highly lethal malignancy with few therapeutic options. Cyclin‑dependent kinase 9 (CDK9), a potential therapeutic target of many cancers, has been recently observed to be upregulated in ccRCC patients. Therefore, we aimed to investigate the therapeutic potential of CDK9 in ccRCC and develop a novel CDK9 inhibitor with low toxicity for ccRCC treatment.

Methods

The expression of CDK9 in ccRCC was checked using the online database and tissue microarray analysis. shRNA-mediated CDK9 knockdown and CDK inhibitor were applied to evaluate the effect of CDK9 on ccRCC. Medicinal chemistry methods were used to develop a new CDK9 inhibitor with drugability. RNA-seq and ChIP-seq experiments were conducted to explore the mechanism of action. MTS, western blotting, and colony formation assays were performed to evaluate the anti-ccRCC effects of CDK9 knockdown and inhibition in vitro. The in vivo anti-tumour efficacy was evaluated in a xenograft model.

Results

CDK9 is overexpressed and associated with poor survival in ccRCC. Knockdown or inhibition of CDK9 significantly suppressed ccRCC cells. XPW1 was identified as a new potent and selective CDK9 inhibitor with excellent anti-ccRCC activity and low toxicity. In mechanism, XPW1 transcriptionally inhibited DNA repair programmes in ccRCC cells, resulting in an excellent anti-tumour effect. CDK9 and BRD4 were two highly correlated transcriptional regulators in ccRCC patients, and the BRD4 inhibitor JQ1 enhanced XPW1’s anti-ccRCC effects in vitro and in vivo.

Conclusions

This work provides valuable insights into the therapeutic potential of CDK9 in ccRCC. The CDK9 inhibitor XPW1 would be a novel therapeutic agent for targeting ccRCC, alone or in rational combinations.

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: CDK9 is upregulated and associated with poor survival in ccRCC.
Fig. 2: The shRNA-mediated CDK9 knockdown and CDK9 inhibitor suppressed cell proliferation, colony formation, and tumour growth in ccRCC.
Fig. 3: Development of novel potential CDK9 inhibitors.
Fig. 4: Anti-ccRCC effect of XPW1 and iCDK9 in vivo.
Fig. 5: XPW1 preferentially suppresses gene expression in terms of transcriptional manner.
Fig. 6: Identification of BRD4 inhibitor JQ1 as a potential enhancer for combination therapy with XPW1 in ccRCC cells.
Fig. 7: Efficacy of combined XPW1 and JQ1 treatment in vivo.

Similar content being viewed by others

Data availability

The data used to support the findings of this study are available from the corresponding author upon request.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.

    PubMed  Google Scholar 

  2. Atkins MB, Tannir NM. Current and emerging therapies for first-line treatment of metastatic clear cell renal cell carcinoma. Cancer Treat Rev. 2018;70:127–37.

    CAS  PubMed  Google Scholar 

  3. Gershman B, Moreira DM, Thompson RH, Boorjian SA, Lohse CM, Costello BA, et al. Renal cell carcinoma with isolated lymph node involvement: long-term natural history and predictors of oncologic outcomes following surgical resection. Eur Urol. 2017;72:300–6.

    PubMed  Google Scholar 

  4. Kim MC, Jin Z, Kolb R, Borcherding N, Chatzkel JA, Falzarano SM, et al. Updates on immunotherapy and immune landscape in renal clear cell carcinoma. Cancers. 2021;13:5856.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mollica V, Di Nunno V, Gatto L, Santoni M, Scarpelli M, Cimadamore A, et al. Resistance to systemic agents in renal cell carcinoma predict and overcome genomic strategies adopted by tumor. Cancers. 2019;11:830.

    PubMed  PubMed Central  Google Scholar 

  6. Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022;15:143.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Motzer RJ, Escudier B, George S, Hammers HJ, Srinivas S, Tykodi SS, et al. Nivolumab versus everolimus in patients with advanced renal cell carcinoma: Updated results with long-term follow-up of the randomized, open-label, phase 3 CheckMate 025 trial. Cancer. 2020;126:4156–67.

    CAS  PubMed  Google Scholar 

  8. Roskoski R Jr. Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs. Pharm Res. 2019;139:471–88.

    CAS  Google Scholar 

  9. García-Reyes B, Kretz AL, Ruff JP, von Karstedt S, Hillenbrand A, Knippschild U, et al. The emerging role of cyclin-dependent kinases (CDKs) in pancreatic ductal adenocarcinoma. Int J Mol Sci. 2018;19:3219.

    PubMed  PubMed Central  Google Scholar 

  10. Bacon CW, D’Orso I. CDK9: a signaling hub for transcriptional control. Transcription. 2019;10:57–75.

    CAS  PubMed  Google Scholar 

  11. Nepomuceno TC, Fernandes VC, Gomes TT, Carvalho RS, Suarez-Kurtz G, Monteiro AN, et al. BRCA1 recruitment to damaged DNA sites is dependent on CDK9. Cell Cycle. 2017;16:665–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ghia P, Scarfo L, Pathiraja K, Derosier M, Small K, Patton N. A phase 3 study to evaluate the efficacy and safety of dinaciclib compared to ofatumumab in patients with refractory chronic lymphocytic leukemia. Blood. 2015;126:4171.

    Google Scholar 

  13. Anshabo AT, Milne R, Wang S, Albrecht H. CDK9: a comprehensive review of its biology, and its role as a potential target for anti-cancer agents. Front Oncol. 2021;11:678559.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Mandal R, Becker S, Strebhardt K. Targeting CDK9 for anti-cancer therapeutics. Cancers. 2021;13:2181.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Alsfouk A. Small molecule inhibitors of cyclin-dependent kinase 9 for cancer therapy. J Enzym Inhib Med Chem. 2021;36:693–706.

    CAS  Google Scholar 

  16. Taniguchi Y. The bromodomain and extra-terminal domain (BET) family: functional anatomy of BET paralogous proteins. Int J Mol Sci. 2016;17:1849.

    PubMed  PubMed Central  Google Scholar 

  17. Wu X, Liu D, Gao X, Xie F, Tao D, Xiao X, et al. Inhibition of BRD4 suppresses cell proliferation and induces apoptosis in renal cell carcinoma. Cell Physiol Biochem: Int J Exp Cell Physiol Biochem Pharmacol. 2017;41:1947–56.

    CAS  Google Scholar 

  18. Rodriguez RM, Huidobro C, Urdinguio RG, Mangas C, Soldevilla B, Dominguez G, et al. Aberrant epigenetic regulation of bromodomain BRD4 in human colon cancer. J Mol Med. 2012;90:587–95.

    CAS  PubMed  Google Scholar 

  19. Donati B, Lorenzini E, Ciarrocchi ABRD4. and Cancer: going beyond transcriptional regulation. Mol Cancer. 2018;17:164.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Anaya J. OncoLnc: linking TCGA survival data to mRNAs, miRNAs, and lncRNAs. PeerJ Prepr. 2016;4:e1780v1.

    Google Scholar 

  21. Chen F, Chandrashekar DS, Varambally S, Creighton CJ. Pan-cancer molecular subtypes revealed by mass-spectrometry-based proteomic characterization of more than 500 human cancers. Nat Commun. 2019;10:5679.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pontén F, Jirström K, Uhlen M. The Human Protein Atlas—a tool for pathology. J Pathol. 2008;216:387–93.

    PubMed  Google Scholar 

  23. Dziekan JM, Wirjanata G, Dai L, Go KD, Yu H, Lim YT, et al. Cellular thermal shift assay for the identification of drug-target interactions in the Plasmodium falciparum proteome. Nat Protoc. 2020;15:1881–921.

    CAS  PubMed  Google Scholar 

  24. Lu J, Zhu L, Zheng LP, Cui Q, Zhu HH, Zhao H, et al. Overexpression of ULK1 represents a potential diagnostic marker for clear cell renal carcinoma and the antitumor effects of SBI-0206965. EBioMedicine. 2018;34:85–93.

    PubMed  PubMed Central  Google Scholar 

  25. Shen HF, Zhang WJ, Huang Y, He YH, Hu GS, Wang L, et al. The dual function of KDM5C in both gene transcriptional activation and repression promotes breast cancer cell growth and tumorigenesis. Adv Sci. 2021;8:2004635.

    CAS  Google Scholar 

  26. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7:562–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Gao WW, Xiao RQ, Zhang WJ, Hu YR, Peng BL, Li WJ, et al. JMJD6 licenses ERα-dependent enhancer and coding gene activation by modulating the recruitment of the CARM1/MED12 co-activator complex. Mol Cell. 2018;70:340–57.e348.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang M, Kuang Z, He Y, Cao Y, Yu T, Cheng J, et al. SNAPIN regulates cell cycle progression to promote pancreatic beta cell growth. Front Endocrinol. 2021;12:624309.

    Google Scholar 

  29. Xiao H, Xiao W, Cao J, Li H, Guan W, Guo X, et al. miR-206 functions as a novel cell cycle regulator and tumor suppressor in clear-cell renal cell carcinoma. Cancer Lett. 2016;374:107–16.

    CAS  PubMed  Google Scholar 

  30. Shen S, Dean DC, Yu Z, Hornicek F, Kan Q, Duan Z. Aberrant CDK9 expression within chordoma tissues and the therapeutic potential of a selective CDK9 inhibitor LDC000067. J Cancer. 2020;11:132–41.

    CAS  PubMed Central  Google Scholar 

  31. Zhang M, Zhang L, Hei R, Li X, Cai H, Wu X, et al. CDK inhibitors in cancer therapy, an overview of recent development. Am J Cancer Res. 2021;11:1913–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Polier G, Ding J, Konkimalla BV, Eick D, Ribeiro N, Köhler R, et al. Wogonin and related natural flavones are inhibitors of CDK9 that induce apoptosis in cancer cells by transcriptional suppression of Mcl-1. Cell Death Dis. 2011;2:e182.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Ao M, Wu J, Cao Y, He Y, Zhang Y, Gao X, et al. The synthesis of PROTAC molecule and new target KAT6A identification of CDK9 inhibitor iCDK9. Chin Chem Lett. 2022;34:107741.

    Google Scholar 

  34. Jiang X, Yu J, Zhou Z, Kongsted J, Song Y, Pannecouque C, et al. Molecular design opportunities presented by solvent-exposed regions of target proteins. Med Res Rev. 2019;39:2194–238.

    PubMed  Google Scholar 

  35. Axelsson H, Almqvist H, Seashore-Ludlow B, Lundbäck T, Markossian S, Grossman A, et al. Screening for target engagement using the cellular thermal shift assay—CETSA. In: S. Markossian et al. editors. Assay Guidance Manual [Internet]. Bethesda (MD): Eli Lilly & Company and the National Center for Advancing Translational Sciences; 2004; PMID, 27478247: 873–97.

  36. Glover-Cutter K, Larochelle S, Erickson B, Zhang C, Shokat K, Fisher RP, et al. TFIIH-associated Cdk7 kinase functions in phosphorylation of C-terminal domain Ser7 residues, promoter-proximal pausing, and termination by RNA polymerase II. Mol Cell Biol. 2009;29:5455–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Chlamydas S, Holz H, Samata M, Chelmicki T, Georgiev P, Pelechano V, et al. Functional interplay between MSL1 and CDK7 controls RNA polymerase II Ser5 phosphorylation. Nat Struct Mol Biol. 2016;23:580–9.

    CAS  PubMed  Google Scholar 

  38. Moreno N, Holsten T, Mertins J, Zhogbi A, Johann P, Kool M, et al. Combined BRD4 and CDK9 inhibition as a new therapeutic approach in malignant rhabdoid tumors. Oncotarget. 2017;8:84986–95.

    PubMed  PubMed Central  Google Scholar 

  39. Sakaguchi T, Yoshino H, Sugita S, Miyamoto K, Yonemori M, Osako Y, et al. Bromodomain protein BRD4 inhibitor JQ1 regulates potential prognostic molecules in advanced renal cell carcinoma. Oncotarget. 2018;9:23003–17.

    PubMed  PubMed Central  Google Scholar 

  40. Ma H, Seebacher NA, Hornicek FJ, Duan Z. Cyclin-dependent kinase 9 (CDK9) is a novel prognostic marker and therapeutic target in osteosarcoma. EBioMedicine. 2019;39:182–93.

    PubMed  Google Scholar 

  41. Chou J, Quigley DA, Robinson TM, Feng FY, Ashworth A. Transcription-associated cyclin-dependent kinases as targets and biomarkers for cancer therapy. Cancer Discov. 2020;10:351–70.

    CAS  PubMed  Google Scholar 

  42. Shen YL, Wang YM, Zhang YX, Ma SJ, Yang LH, Zhao CG, et al. Targeting cyclin-dependent kinase 9 in cancer therapy. Acta Pharm Sin. 2022;43:1633–45.

    CAS  Google Scholar 

  43. Xue S, Shao Q, Zhu LB, Jiang YF, Wang C, Xue B, et al. LDC000067 suppresses RANKL-induced osteoclastogenesis in vitro and prevents LPS-induced osteolysis in vivo. Int Immunopharmacol. 2019;75:105826.

    CAS  PubMed  Google Scholar 

  44. Rahaman MH, Yu Y, Zhong L, Adams J, Lam F, Li P, et al. CDKI-73: an orally bioavailable and highly efficacious CDK9 inhibitor against acute myeloid leukemia. Invest N Drugs. 2019;37:625–35.

    CAS  Google Scholar 

  45. Zhou F, Zhao J, Cicic D, Lan J, Lu Q. AML-259 pharmacokinetics and bioequivalence of two formulations of GFH009 maleate injection in Sprague Dawley rats. Clin Lymphoma Myeloma Leuk. 2022;22:S230–1.

    CAS  Google Scholar 

  46. Cidado J, Boiko S, Proia T, Ferguson D, Criscione SW, San Martin M, et al. AZD4573 is a highly selective CDK9 inhibitor that suppresses MCL-1 and induces apoptosis in hematologic cancer cells. Clin Cancer Res. 2020;26:922–34.

    CAS  PubMed  Google Scholar 

  47. Diamond JR, Boni V, Lim E, Nowakowski G, Cordoba R, Morillo D, et al. First-in-human dose-escalation study of cyclin-dependent kinase 9 inhibitor VIP152 in patients with advanced malignancies shows early signs of clinical efficacy. Clin Cancer Res. 2022;28:1285–93.

    CAS  PubMed  Google Scholar 

  48. Dey J, Deckwerth TL, Kerwin WS, Casalini JR, Merrell AJ, Grenley MO, et al. Voruciclib, a clinical stage oral CDK9 inhibitor, represses MCL-1 and sensitizes high-risk Diffuse Large B-cell Lymphoma to BCL2 inhibition. Sci Rep. 2017;7:18007.

    PubMed  PubMed Central  Google Scholar 

  49. Bhurta D, Bharate SB. Analyzing the scaffold diversity of cyclin-dependent kinase inhibitors and revisiting the clinical and preclinical pipeline. Med Res Rev. 2022;42:654–709.

    CAS  PubMed  Google Scholar 

  50. Fujinaga K, Huang F, Peterlin BM. P-TEFb: The master regulator of transcription elongation. Mol Cell. 2023;83:393–403.

    CAS  PubMed  Google Scholar 

  51. Sausville EA. Complexities in the development of cyclin-dependent kinase inhibitor drugs. Trends Mol Med. 2022;8:S32–7.

    Google Scholar 

  52. Huang CH, Lujambio A, Zuber J, Tschaharganeh DF, Doran MG, Evans MJ, et al. CDK9-mediated transcription elongation is required for MYC addiction in hepatocellular carcinoma. Genes Dev. 2014;28:1800–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. David SY, David C. A role for CDK9-cyclin K in maintaining genome integrity. Cell Cycle. 2011;10:28–32.

    Google Scholar 

  54. Liu X, Zhao B, Shaw TI, Fridley BL, Duckett DR, Tan AC, et al. Summarizing internal dynamics boosts differential analysis and functional interpretation of super enhancers. Nucleic Acids Res. 2022;50:3115–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Minzel W, Venkatachalam A, Fink A, Hung E, Brachya G, Burstain I, et al. Small molecules co-targeting CKIα and the transcriptional kinases CDK7/9 control AML in preclinical models. Cell. 2018;175:171–85.e125.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Shi ZD, Pang K, Wu ZX, Dong Y, Hao L, Qin JX, et al. Tumor cell plasticity in targeted therapy-induced resistance: mechanisms and new strategies. Signal Transduct Target Ther. 2023;8:113.

    PubMed  PubMed Central  Google Scholar 

  57. McLaughlin RP, He J, van der Noord VE, Redel J, Foekens JA, Martens JWM, et al. A kinase inhibitor screen identifies a dual cdc7/CDK9 inhibitor to sensitise triple-negative breast cancer to EGFR-targeted therapy. Breast Cancer Res. 2019;21:77.

    PubMed  PubMed Central  Google Scholar 

  58. Chu YY, Chen MK, Wei Y, Lee HH, Xia W, Wang YN, et al. Targeting the ALK-CDK9-Tyr19 kinase cascade sensitizes ovarian and breast tumors to PARP inhibition via destabilization of the P-TEFb complex. Nat Cancer. 2022;3:1211–27.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Kim SY, Zhang X, Schiattarella GG, Altamirano F, Ramos TAR, French KM, et al. Epigenetic reader BRD4 (Bromodomain-Containing Protein 4) governs nucleus-encoded mitochondrial transcriptome to regulate cardiac function. Circulation. 2020;142:2356–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Fukui T, Yik JHN, Doyran B, Davis J, Haudenschild AK, Adamopoulos IE, et al. Bromodomain-containing-protein-4 and cyclin-dependent-kinase-9 inhibitors interact synergistically in vitro and combined treatment reduces post-traumatic osteoarthritis severity in mice. Osteoarthr Cartil. 2021;29:68–77.

    CAS  Google Scholar 

Download references

Acknowledgements

The authors thank all staffs who assisted with the study.

Funding

This work was supported by grants from the National Natural Science Foundation of China (82125028 and 82102746) and the Fundamental Research Funds for the Central Universities of China (No. 20720180051).

Author information

Authors and Affiliations

Authors

Contributions

Study design: ZK, KG, TW, WL and MF; data collection: ZK, KG, YC, MJ, CW, QW, TZ and SL; bioinformatics and statistics: GH and TW; methodology: ZK, KG, MA and JQ; data analysis: ZK, KG, YC, MJ and MH; technical support: CS and ML; study supervision: WL and MF; writing—original draft: ZK, KG, YC and MJ; writing—review and editing: TW, WL and MF.

Corresponding authors

Correspondence to Tong Wu, Wen Liu or Meijuan Fang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

The animal studies were approved by the Ethics Committee for Animal Experimentation and Animal Welfare at Xiamen University. The study involved no human subjects.

Consent for publication

Not applicable.

Additional information

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

Supplementary information

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

Kuang, Z., Guo, K., Cao, Y. et al. The novel CDK9 inhibitor, XPW1, alone and in combination with BRD4 inhibitor JQ1, for the treatment of clear cell renal cell carcinoma. Br J Cancer 129, 1915–1929 (2023). https://doi.org/10.1038/s41416-023-02464-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41416-023-02464-y

Search

Quick links