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.

  • Review Article
  • Published:

Targeting transcription cycles in cancer

Abstract

Accurate control of gene expression is essential for normal development and dysregulation of transcription underpins cancer onset and progression. Similar to cell cycle regulation, RNA polymerase II-driven transcription can be considered as a unidirectional multistep cycle, with thousands of unique transcription cycles occurring in concert within each cell. Each transcription cycle comprises recruitment, initiation, pausing, elongation, termination and recycling stages that are tightly controlled by the coordinated action of transcriptional cyclin-dependent kinases and their cognate cyclins as well as the opposing activity of transcriptional phosphatases. Oncogenic dysregulation of transcription can entail defective control of gene expression, either at select loci or more globally, impacting a large proportion of the genome. The resultant dependency on the core-transcriptional machinery is believed to render ‘transcriptionally addicted’ cancers sensitive to perturbation of transcription. Based on these findings, small molecules targeting transcriptional cyclin-dependent kinases and associated proteins hold promise for the treatment of cancer. Here, we utilize the transcription cycles concept to explain how dysregulation of these finely tuned gene expression processes may drive tumorigenesis and how therapeutically beneficial responses may arise from global or selective transcriptional perturbation. This conceptual framework helps to explain tumour-selective transcriptional dependencies and facilitates the rational design of combination therapies.

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: An updated view of Pol II transcription cycles.
Fig. 2: Multi-tiered regulation of transcription cycles.
Fig. 3: Distinct properties define the vulnerability of transcription cycles to dysregulation in cancer.
Fig. 4: Selective dysregulation of transcription cycles.
Fig. 5: Distinct features of oncogenic transcription are targetable by tCDK inhibitors.

Similar content being viewed by others

References

  1. Bradner, J. E., Hnisz, D. & Young, R. A. Transcriptional addiction in cancer. Cell 168, 629–643 (2017). Important review article detailing the conceptual framework for transcriptional addiction in cancer and how this information might be exploited to develop new therapeutic modalities.

    CAS  PubMed Central  PubMed  Google Scholar 

  2. Lee, T. I. & Young, R. A. Transcriptional regulation and its misregulation in disease. Cell 152, 1237–1251 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  3. Parua, P. K. & Fisher, R. P. Dissecting the Pol II transcription cycle and derailing cancer with CDK inhibitors. Nat. Chem. Biol. 16, 716–724 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  4. Martin, R. D., Hébert, T. E. & Tanny, J. C. Therapeutic targeting of the general RNA polymerase II transcription machinery. Int. J. Mol. Sci. 21, 3354 (2020).

    CAS  PubMed Central  Google Scholar 

  5. Schier, A. C. & Taatjes, D. J. Structure and mechanism of the RNA polymerase II transcription machinery. Genes Dev. 34, 465–488 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  6. Chen, X. et al. Structures of the human mediator and mediator-bound preinitiation complex. Science 372, 52–56 (2021).

    PubMed Central  PubMed  Google Scholar 

  7. Abdella, R. et al. Structure of the human mediator-bound transcription preinitiation complex. Science 372, 52–56 (2021).

    CAS  PubMed Central  PubMed  Google Scholar 

  8. Ebmeier, C. C. et al. Human TFIIH Kinase CDK7 regulates transcription-associated chromatin modifications. Cell Rep. 20, 1173–1186 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  9. Glover-Cutter, K. 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. 29, 5455–5464 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  10. Nilson, K. A. et al. THZ1 reveals roles for Cdk7 in co-transcriptional capping and pausing. Mol. Cell 59, 576–587 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  11. Noe Gonzalez, M. et al. CTD-dependent and -independent mechanisms govern co-transcriptional capping of Pol II transcripts. Nat. Commun. 9, 3392 (2018).

    PubMed Central  PubMed  Google Scholar 

  12. Fant, C. B. & Taatjes, D. J. Regulatory functions of the mediator kinases CDK8 and CDK19. Transcription 10, 76–90 (2019).

    PubMed  Google Scholar 

  13. Vos, S. M. et al. Structure of paused transcription complex Pol II-DSIF-NELF. Nature 560, 601–606 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Chen, F. X., Smith, E. R. & Shilatifard, A. Born to run: control of transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 19, 464–478 (2018).

    CAS  PubMed  Google Scholar 

  15. Booth, G. T. et al. Cdk9 regulates a promoter-proximal checkpoint to modulate RNA polymerase II elongation rate in fission yeast. Nat. Commun. 9, 543 (2018).

    PubMed Central  PubMed  Google Scholar 

  16. Jonkers, I., Kwak, H. & Lis, J. T. Genome-wide dynamics of Pol II elongation and its interplay with promoter proximal pausing, chromatin, and exons. eLife 3, e02407 (2014).

    PubMed Central  PubMed  Google Scholar 

  17. Vos, S. M. et al. Structure of activated transcription complex Pol II-DSIF-PAF-SPT6. Nature 560, 607–612 (2018). Together with Vos et al. (reference 13), this study presents the structures of the transcriptionally paused and elongating Pol II complexes and demonstrate how CDK9-mediated phosphorylation modulates complex assembly.

    CAS  PubMed  Google Scholar 

  18. Cortazar, M. A. et al. Control of RNA Pol II Speed by PNUTS-PP1 and Spt5 Dephosphorylation Facilitates Termination by a “Sitting Duck Torpedo” Mechanism. Mol. Cell 76, 896–908.e4 (2019). This study demonstrates a role for the PNUTS-PP1 phosphatase complex for the control of Pol II elongation rate and termination through the modulation of Spt5 phosphorylation.

    CAS  PubMed Central  PubMed  Google Scholar 

  19. Dubbury, S. J., Boutz, P. L. & Sharp, P. A. CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature 564, 141–145 (2018). This study demonstrates that CDK12 suppresses intronic polyadenylation and is required for the full-length expression of homologous recombination genes.

    CAS  PubMed Central  PubMed  Google Scholar 

  20. Fan, Z. et al. CDK13 cooperates with CDK12 to control global RNA polymerase II processivity. Sci. Adv. 6, eaaz5041 (2020). Using a chemical-genetic analogue-sensitive mutant kinase approach, this study demonstrates the molecular redundancy between CDK13 and CDK12 for the control of Pol II processivity and elongation.

    CAS  PubMed Central  PubMed  Google Scholar 

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

    PubMed Central  PubMed  Google Scholar 

  22. 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).

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Tellier, M. et al. CDK12 globally stimulates RNA polymerase II transcription elongation and carboxyl-terminal domain phosphorylation. Nucleic Acids Res. 48, 7712–7727 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  24. Fisher, R. P. Cdk7: a kinase at the core of transcription and in the crosshairs of cancer drug discovery. Transcription 10, 47–56 (2019).

    CAS  PubMed  Google Scholar 

  25. Olson, C. M. et al. Development of a selective CDK7 covalent inhibitor reveals predominant cell-cycle phenotype. Cell Chem. Biol. 26, 792–803.e10 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  26. Schachter, M. M. et al. A Cdk7-Cdk4 T-loop phosphorylation cascade promotes G1 progression. Mol. Cell 50, 250–260 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  27. Petretti, C. et al. The PITSLRE/CDK11p58 protein kinase promotes centrosome maturation and bipolar spindle formation. EMBO Rep. 7, 418–424 (2006).

    CAS  PubMed Central  PubMed  Google Scholar 

  28. Gajdušková, P. et al. CDK11 is required for transcription of replication-dependent histone genes. Nat. Struct. Mol. Biol. 27, 500–510 (2020).

    PubMed Central  PubMed  Google Scholar 

  29. Trembley, J. H. et al. Casein kinase 2 interacts with cyclin-dependent kinase 11 (CDK11) in vivo and phosphorylates both the RNA polymerase II carboxyl-terminal domain and CDK11 in vitro. J. Biol. Chem. 278, 2265–2270 (2003).

    CAS  PubMed  Google Scholar 

  30. Heim, A., Rymarczyk, B. & Mayer, T. U. Regulation of cell division. Adv. Exp. Med. Biol. 953, 83–116 (2017).

    CAS  PubMed  Google Scholar 

  31. Hnisz, D. et al. Convergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers. Mol. Cell 58, 362–370 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  32. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    CAS  PubMed  Google Scholar 

  33. Lin, C. Y. et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell 151, 56–67 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Nie, Z. et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell 151, 68–79 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  35. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 351, 1454–1458 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Lovén, J. et al. Revisiting global gene expression analysis. Cell 151, 476–482 (2012).

    PubMed Central  PubMed  Google Scholar 

  37. Chou, J. et al. Transcription-associated cyclin-dependent kinases as targets and biomarkers for cancer therapy. Cancer Discov. 10, 351–370 (2020).

    CAS  PubMed  Google Scholar 

  38. Shan, W. et al. Systematic characterization of recurrent genomic alterations in cyclin-dependent kinases reveals potential therapeutic strategies for cancer treatment. Cell Rep. 32, 107884 (2020). This study characterizes genetic alterations in genes encoding cyclin-dependent kinases and cyclins across >10,000 tumours and identifies correlations between CDK-associated mutations and sensitivity to DNA-damaging agents.

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Clark, V. E. et al. Recurrent somatic mutations in POLR2A define a distinct subset of meningiomas. Nat. Genet. 48, 1253–1259 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  40. Li, Y. et al. Heterozygous deletion of chromosome 17p renders prostate cancer vulnerable to inhibition of RNA polymerase II. Nature Commun. 9, 4394 (2018).

    Google Scholar 

  41. Bancerek, J. et al. CDK8 kinase phosphorylates transcription factor STAT1 to selectively regulate the interferon response. Immunity 38, 250–262 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  42. Nitulescu, I. I. et al. Mediator kinase phosphorylation of STAT1 S727 promotes growth of neoplasms with JAK-STAT activation. EBioMedicine 26, 112–125 (2017).

    PubMed Central  PubMed  Google Scholar 

  43. Tsai, K. L. et al. A conserved mediator-CDK8 kinase module association regulates mediator-RNA polymerase II interaction. Nat. Struct. Mol. Biol. 20, 611–619 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  44. Firestein, R. et al. CDK8 expression in 470 colorectal cancers in relation to beta-catenin activation, other molecular alterations and patient survival. Int. J. Cancer 126, 2863–2873 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  45. Martin, E. S. et al. Common and distinct genomic events in sporadic colorectal cancer and diverse cancer types. Cancer Res. 67, 10736–10743 (2007).

    CAS  PubMed  Google Scholar 

  46. Jaeger, M. G. et al. Selective mediator dependence of cell-type-specifying transcription. Nat. Genet. 52, 719–727 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  47. Pelish, H. E. et al. Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature 526, 273–276 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  48. Philip, S. et al. Cyclin-dependent kinase 8: a new hope in targeted cancer therapy? J. Med. Chem. 61, 5073–5092 (2018).

    CAS  PubMed  Google Scholar 

  49. Morris, E. J. et al. E2F1 represses beta-catenin transcription and is antagonized by both pRB and CDK8. Nature 455, 552–556 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  50. Zhao, J., Ramos, R. & Demma, M. CDK8 regulates E2F1 transcriptional activity through S375 phosphorylation. Oncogene 32, 3520–3530 (2013).

    CAS  PubMed  Google Scholar 

  51. McCleland, M. L. et al. Cdk8 deletion in the Apc(Min) murine tumour model represses EZH2 activity and accelerates tumourigenesis. J. Pathol. 237, 508–519 (2015).

    CAS  PubMed  Google Scholar 

  52. Kämpjärvi, K. et al. Somatic MED12 mutations in prostate cancer and uterine leiomyomas promote tumorigenesis through distinct mechanisms. Prostate 76, 22–31 (2016).

    PubMed  Google Scholar 

  53. Mäkinen, N. et al. MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas. Science 334, 252–255 (2011).

    PubMed  Google Scholar 

  54. Turunen, M. et al. Uterine leiomyoma-linked MED12 mutations disrupt mediator-associated CDK activity. Cell Rep. 7, 654–660 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Barbieri, C. E. et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat. Genet. 44, 685–689 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  56. Klatt, F. et al. A precisely positioned MED12 activation helix stimulates CDK8 kinase activity. Proc. Natl Acad. Sci. USA 117, 2894–2905 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  57. Knuesel, M. T. et al. The human CDK8 subcomplex is a molecular switch that controls Mediator coactivator function. Genes Dev. 23, 439–451 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  58. Baker, A. et al. The CDK9 inhibitor dinaciclib exerts potent apoptotic and antitumor effects in preclinical models of MLL-rearranged acute myeloid leukemia. Cancer Res. 76, 1158–1169 (2016).

    CAS  PubMed  Google Scholar 

  59. Gregory, G. P. et al. CDK9 inhibition by dinaciclib potently suppresses Mcl-1 to induce durable apoptotic responses in aggressive MYC-driven B-cell lymphoma in vivo. Leukemia 29, 1437–1441 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Wang, Y. et al. CDK7-dependent transcriptional addiction in triple-negative breast cancer. Cell 163, 174–186 (2015). This study linked therapeutic efficacy of covalent inhibitors of CDK7/12/13 in TNBC to highly selective inhibition of an ‘Achilles cluster’ of super-enhancer-associated genes.

    CAS  PubMed Central  PubMed  Google Scholar 

  61. Rahl, P. B. et al. c-Myc regulates transcriptional pause release. Cell 141, 432–445 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  62. Rahl, P. B. & Young, R. A. MYC and transcription elongation. Cold Spring Harb. Perspect. Med. 4, a020990 (2014).

    PubMed Central  PubMed  Google Scholar 

  63. Eberhardy, S. R. & Farnham, P. J. Myc recruits P-TEFb to mediate the final step in the transcriptional activation of the cad promoter. J. Biol. Chem. 277, 40156–40162 (2002).

    CAS  PubMed  Google Scholar 

  64. Gargano, B. et al. P-TEFb is a crucial co-factor for Myc transactivation. Cell Cycle 6, 2031–2037 (2007).

    CAS  PubMed  Google Scholar 

  65. Kanazawa, S. et al. c-Myc recruits P-TEFb for transcription, cellular proliferation and apoptosis. Oncogene 22, 5707–5711 (2003).

    CAS  PubMed  Google Scholar 

  66. Walz, S. et al. Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature 511, 483–487 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  67. Kalkat, M. et al. MYC deregulation in primary human cancers. Genes 8, 151 (2017).

    PubMed Central  Google Scholar 

  68. Dalla-Favera, R. et al. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc. Natl Acad. Sci. USA 79, 7824–7827 (1982).

    CAS  PubMed Central  PubMed  Google Scholar 

  69. Taub, R. et al. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc. Natl Acad. Sci. USA 79, 7837–7841 (1982).

    CAS  PubMed Central  PubMed  Google Scholar 

  70. Walker, B. A. et al. Translocations at 8q24 juxtapose MYC with genes that harbor superenhancers resulting in overexpression and poor prognosis in myeloma patients. Blood Cancer J. 4, e191 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  71. Affer, M. et al. Promiscuous MYC locus rearrangements hijack enhancers but mostly super-enhancers to dysregulate MYC expression in multiple myeloma. Leukemia 28, 1725–1735 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  72. Zhang, X. et al. Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat. Genet. 48, 176–182 (2016).

    CAS  PubMed  Google Scholar 

  73. Herranz, D. et al. A NOTCH1-driven MYC enhancer promotes T cell development, transformation and acute lymphoblastic leukemia. Nat. Med. 20, 1130–1137 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  74. Shi, J. et al. Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev. 27, 2648–2662 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  75. Liang, J. et al. Epstein-Barr virus super-enhancer eRNAs are essential for MYC oncogene expression and lymphoblast proliferation. Proc. Natl Acad. Sci. USA 113, 14121–14126 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Sabò, A. et al. Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature 511, 488–492 (2014).

    PubMed Central  PubMed  Google Scholar 

  77. Chandriani, S. et al. A core MYC gene expression signature is prominent in basal-like breast cancer but only partially overlaps the core serum response. PLoS ONE 4, e6693 (2009).

    PubMed Central  PubMed  Google Scholar 

  78. Baluapuri, A., Wolf, E. & Eilers, M. Target gene-independent functions of MYC oncoproteins. Nat. Rev. Mol. Cell Biol. 21, 255–267 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  79. Ji, H. et al. Cell-type independent MYC target genes reveal a primordial signature involved in biomass accumulation. PLoS ONE 6, e26057 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  80. Lin, C. et al. AFF4, a component of the ELL/P-TEFb elongation complex and a shared subunit of MLL chimeras, can link transcription elongation to leukemia. Mol. Cell 37, 429–437 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Basu, S., Nandy, A. & Biswas, D. Keeping RNA polymerase II on the run: Functions of MLL fusion partners in transcriptional regulation. Biochim. Biophys. Acta Gene Regul. Mech. 1863, 194563 (2020).

    CAS  PubMed  Google Scholar 

  82. Bolouri, H. et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat. Med. 24, 103–112 (2018).

    CAS  PubMed  Google Scholar 

  83. Meyer, C. et al. The MLL recombinome of acute leukemias in 2017. Leukemia 32, 273–284 (2018).

    CAS  PubMed  Google Scholar 

  84. Milne, T. A. et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol. Cell 10, 1107–1117 (2002).

    CAS  PubMed  Google Scholar 

  85. Milne, T. A. et al. MLL associates specifically with a subset of transcriptionally active target genes. Proc. Natl Acad. Sci. USA 102, 14765–14770 (2005).

    CAS  PubMed Central  PubMed  Google Scholar 

  86. Armstrong, S. A. et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat. Genet. 30, 41–47 (2002).

    CAS  PubMed  Google Scholar 

  87. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006).

    CAS  PubMed  Google Scholar 

  88. Somervaille, T. C. & Cleary, M. L. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell 10, 257–268 (2006).

    CAS  PubMed  Google Scholar 

  89. Bernt, K. M. et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 20, 66–78 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  90. Nguyen, A. T. et al. DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood 117, 6912–6922 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  91. Okuda, H. et al. Cooperative gene activation by AF4 and DOT1L drives MLL-rearranged leukemia. J. Clin. Invest. 127, 1918–1931 (2017).

    PubMed Central  PubMed  Google Scholar 

  92. Mueller, D. et al. A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood 110, 4445–4454 (2007).

    CAS  PubMed Central  PubMed  Google Scholar 

  93. Dawson, M. A. et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  94. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  95. Phillips, D. C. et al. A novel CDK9 inhibitor increases the efficacy of venetoclax (ABT-199) in multiple models of hematologic malignancies. Leukemia 34, 1646–1657 (2020).

    CAS  PubMed  Google Scholar 

  96. Luo, Z., Lin, C. & Shilatifard, A. The super elongation complex (SEC) family in transcriptional control. Nat. Rev. Mol. Cell Biol. 13, 543–547 (2012).

    CAS  PubMed  Google Scholar 

  97. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

    Google Scholar 

  98. Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell 163, 1011–1025 (2015).

    Google Scholar 

  99. Wu, Y. M. et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell 173, 1770–1782.e14 (2018). This study demonstrated increased neoantigen burden and T cell infiltration in metastatic castration-resistant prostate cancers characterized by inactivating CDK12 mutations and focal tandem duplications.

    CAS  PubMed  Google Scholar 

  100. Liang, S. et al. CDK12: a potent target and biomarker for human cancer therapy. Cells 9, 1483 (2020).

    CAS  PubMed Central  Google Scholar 

  101. Joshi, P. M. et al. Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J. Biol. Chem. 289, 9247–9253 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  102. Ciriello, G. et al. Comprehensive molecular portraits of invasive lobular breast cancer. Cell 163, 506–519 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  103. Zang, Z. J. et al. Genetic and structural variation in the gastric cancer kinome revealed through targeted deep sequencing. Cancer Res. 71, 29–39 (2011).

    CAS  PubMed  Google Scholar 

  104. Tien, J. F. et al. CDK12 regulates alternative last exon mRNA splicing and promotes breast cancer cell invasion. Nucleic Acids Res. 45, 6698–6716 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  105. 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).

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Menghi, F. et al. The tandem duplicator phenotype is a prevalent genome-wide cancer configuration driven by distinct gene mutations. Cancer Cell 34, 197–210.e5 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  107. Popova, T. et al. Ovarian cancers harboring inactivating mutations in CDK12 display a distinct genomic instability pattern characterized by large tandem duplications. Cancer Res. 76, 1882–1891 (2016). Together with Menghi et al. (2018), this study identifies genomic instability associated with large tandem duplications in cancers with inactivating CDK12 mutations in ovarian cancers.

    CAS  PubMed  Google Scholar 

  108. Lord, C. J. & Ashworth, A. BRCAness revisited. Nat. Rev. Cancer 16, 110–120 (2016).

    CAS  PubMed  Google Scholar 

  109. Tufegdžić Vidaković, A. et al. Regulation of the RNAPII Pool Is Integral to the DNA damage response. Cell 180, 1245–1261.e21 (2020).

    PubMed Central  PubMed  Google Scholar 

  110. Insco, M. L. et al. Mutations drive melanoma via accumulation of prematurely terminated transcripts. Preprint at bioRxiv https://doi.org/10.1101/824193 (2019).

    Article  Google Scholar 

  111. Dong, X. et al. CDK13 RNA over-editing mediated by ADAR1 associates with poor prognosis of hepatocellular carcinoma patients. Cell Physiol. Biochem. 47, 2602–2612 (2018).

    CAS  PubMed  Google Scholar 

  112. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    PubMed Central  PubMed  Google Scholar 

  113. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013). Together with Loven et al. (2013), this study shows that large enhancer elements, coined super-enhancer elements, drive the expression of key oncogenes, such as MYC and cell identity genes, and are more sensitive to perturbation with BET inhibitors.

    CAS  PubMed Central  PubMed  Google Scholar 

  114. Hyde, R. K., Liu, P. & Friedman, A. D. RUNX1 and CBFβ mutations and activities of their wild-type alleles in AML. Adv. Exp. Med. Biol. 962, 265–282 (2017).

    CAS  PubMed  Google Scholar 

  115. Breit, T. M. et al. Site-specific deletions involving the tal-1 and sil genes are restricted to cells of the T cell receptor alpha/beta lineage: T cell receptor delta gene deletion mechanism affects multiple genes. J. Exp. Med. 177, 965–977 (1993).

    CAS  PubMed  Google Scholar 

  116. Mansour, M. R. et al. Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346, 1373–1377 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  117. Choi, Y. et al. Erythroid inhibition by the leukemic fusion AML1-ETO is associated with impaired acetylation of the major erythroid transcription factor GATA-1. Cancer Res. 66, 2990–2996 (2006).

    CAS  PubMed  Google Scholar 

  118. Ptasinska, A. et al. Identification of a dynamic core transcriptional network in t(8;21) AML that regulates differentiation block and self-renewal. Cell Rep. 8, 1974–1988 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  119. Saeed, S. et al. Chromatin accessibility, p300, and histone acetylation define PML-RARα and AML1-ETO binding sites in acute myeloid leukemia. Blood 120, 3058–3068 (2012).

    CAS  PubMed  Google Scholar 

  120. Vangala, R. K. et al. The myeloid master regulator transcription factor PU.1 is inactivated by AML1-ETO in t(8;21) myeloid leukemia. Blood 101, 270–277 (2003).

    CAS  PubMed  Google Scholar 

  121. Saeed, S. et al. Genome-wide functions of PML-RARα in acute promyelocytic leukaemia. Br. J. Cancer 104, 554–558 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  122. Brooks, A. N. et al. A pan-cancer analysis of transcriptome changes associated with somatic mutations in U2AF1 reveals commonly altered splicing events. PLoS ONE 9, e87361 (2014).

    PubMed Central  PubMed  Google Scholar 

  123. Dalton, W. B. et al. Hotspot SF3B1 mutations induce metabolic reprogramming and vulnerability to serine deprivation. J. Clin. Invest. 129, 4708–4723 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  124. Darman, R. B. et al. Cancer-associated SF3B1 hotspot mutations induce cryptic 3′ splice site selection through use of a different branch point. Cell Rep. 13, 1033–1045 (2015).

    CAS  PubMed  Google Scholar 

  125. Graubert, T. A. et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat. Genet. 44, 53–57 (2011).

    PubMed Central  PubMed  Google Scholar 

  126. Kim, E. et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell 27, 617–630 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  127. Liang, Y. et al. SRSF2 mutations drive oncogenesis by activating a global program of aberrant alternative splicing in hematopoietic cells. Leukemia 32, 2659–2671 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  128. Przychodzen, B. et al. Patterns of missplicing due to somatic U2AF1 mutations in myeloid neoplasms. Blood 122, 999–1006 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  129. Rahman, M. A. et al. Recurrent SRSF2 mutations in MDS affect both splicing and NMD. Genes Dev. 34, 413–427 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  130. Zhang, J. et al. Disease-causing mutations in SF3B1 alter splicing by disrupting interaction with SUGP1. Mol. Cell 76, 82–95.e7 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  131. Bartkowiak, B., Yan, C. & Greenleaf, A. L. Engineering an analog-sensitive CDK12 cell line using CRISPR/Cas. Biochim. Biophys. Acta 1849, 1179–1187 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  132. Rimel, J. K. et al. Selective inhibition of CDK7 reveals high-confidence targets and new models for TFIIH function in transcription. Genes Dev. 34, 1452–1473 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  133. Thapar, R. Structural basis for regulation of RNA-binding proteins by phosphorylation. ACS Chem. Biol. 10, 652–666 (2015).

    CAS  PubMed  Google Scholar 

  134. Bensaude, O. Inhibiting eukaryotic transcription: Which compound to choose? How to evaluate its activity? Transcription 2, 103–108 (2011).

    PubMed Central  PubMed  Google Scholar 

  135. Falini, B., Brunetti, L. & Martelli, M. P. Dactinomycin in NPM1-mutated acute myeloid leukemia. N. Engl. J. Med. 373, 1180–1182 (2015).

    PubMed  Google Scholar 

  136. George, B. et al. A phase I, first-in-human, open-label, dose-escalation, safety, pharmacokinetic, and pharmacodynamic study of oral TP-1287 administered daily to patients with advanced solid tumors. J. Clin. Oncol. 38 (Suppl. 15), 3611–3611 (2020).

    Google Scholar 

  137. Bishop, A. C. et al. A chemical switch for inhibitor-sensitive alleles of any protein kinase. Nature 407, 395–401 (2000). This study presents a chemical-genetic strategy for highly selective protein kinase inhibition through the generation of ATP analogue-sensitive mutant alleles.

    CAS  PubMed  Google Scholar 

  138. Decker, T. M. et al. Analog-sensitive cell line identifies cellular substrates of CDK9. Oncotarget 10, 6934–6943 (2019).

    PubMed Central  PubMed  Google Scholar 

  139. Galbraith, M. D. et al. CDK8 kinase activity promotes glycolysis. Cell Rep. 21, 1495–1506 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  140. Larochelle, S. et al. Cyclin-dependent kinase control of the initiation-to-elongation switch of RNA polymerase II. Nat. Struct. Mol. Biol. 19, 1108–1115 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  141. Clarke, P. A. et al. Assessing the mechanism and therapeutic potential of modulators of the human mediator complex-associated protein kinases. eLife 5, e20722. (2016).

    PubMed Central  PubMed  Google Scholar 

  142. Mallinger, A. et al. Discovery of potent, orally bioavailable, small-molecule inhibitors of WNT signaling from a cell-based pathway screen. J. Med. Chem. 58, 1717–1735 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  143. Czodrowski, P. et al. Structure-based optimization of potent, selective, and orally bioavailable CDK8 inhibitors discovered by high-throughput screening. J. Med. Chem. 59, 9337–9349 (2016).

    CAS  PubMed  Google Scholar 

  144. Dale, T. et al. A selective chemical probe for exploring the role of CDK8 and CDK19 in human disease. Nat. Chem. Biol. 11, 973–980 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  145. Rzymski, T. et al. SEL120-34A is a novel CDK8 inhibitor active in AML cells with high levels of serine phosphorylation of STAT1 and STAT5 transactivation domains. Oncotarget 8, 33779–33795 (2017).

    PubMed Central  PubMed  Google Scholar 

  146. McDermott, M. S. et al. Inhibition of CDK8 mediator kinase suppresses estrogen dependent transcription and the growth of estrogen receptor positive breast cancer. Oncotarget 8, 12558–12575 (2017).

    PubMed Central  PubMed  Google Scholar 

  147. Zheng, C., Liu, M. & Fan, H. Targeting complexes of super-enhancers is a promising strategy for cancer therapy. Oncol. Lett. 20, 2557–2566 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  148. Kwiatkowski, N. et al. Targeting transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 511, 616–620 (2014). This study presents the first covalent inhibitor of CDK7 and demonstrates the potent disruption of the core transcriptional regulatory circuitry following targeted inhibition of CDK7 in T-ALL.

    CAS  PubMed Central  PubMed  Google Scholar 

  149. Hu, S. et al. Discovery and characterization of SY-1365, a selective, covalent inhibitor of CDK7. Cancer Res. 79, 3479–3491 (2019).

    PubMed  Google Scholar 

  150. Slobodin, B. et al. Transcription dynamics regulate poly(A) tails and expression of the RNA degradation machinery to balance mRNA Levels. Mol. Cell 78, 434–444.e5 (2020). This study demonstrates the coordination between Pol II-driven mRNA production and mRNA degradation for the control of balanced gene expression.

    CAS  PubMed  Google Scholar 

  151. Robinson, D. R. et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat. Genet. 45, 1446–1451 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  152. Toy, W. et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat. Genet. 45, 1439–1445 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  153. Chen, D. et al. Activation of estrogen receptor alpha by S118 phosphorylation involves a ligand-dependent interaction with TFIIH and participation of CDK7. Mol. Cell 6, 127–137 (2000).

    CAS  PubMed  Google Scholar 

  154. Harrod, A. et al. Genomic modelling of the ESR1 Y537S mutation for evaluating function and new therapeutic approaches for metastatic breast cancer. Oncogene 36, 2286–2296 (2017).

    CAS  PubMed  Google Scholar 

  155. Jeselsohn, R. et al. Allele-specific chromatin recruitment and therapeutic vulnerabilities of ESR1 activating mutations. Cancer Cell 33, 173–186.e5 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  156. Patel, H. et al. ICEC0942, an orally bioavailable selective inhibitor of CDK7 for cancer treatment. Mol. Cancer Ther. 17, 1156–1166 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  157. Liang, H. et al. Recent progress in development of cyclin-dependent kinase 7 inhibitors for cancer therapy. Expert Opin. Investig. Drugs 30, 61–76 (2021).

    CAS  PubMed  Google Scholar 

  158. Satyam, L. K. et al. Potent anti-tumor activity of AUR102, a selective covalent inhibitor of CDK7. Eur. J. Cancer 138, S27 (2020).

    Google Scholar 

  159. Kent, L. N. & Leone, G. The broken cycle: E2F dysfunction in cancer. Nat. Rev. Cancer 19, 326–338 (2019).

    CAS  PubMed  Google Scholar 

  160. Zhang, H. et al. CDK7 inhibition potentiates genome instability triggering anti-tumor immunity in small cell lung cancer. Cancer Cell 37, 37–54.e9 (2020).

    CAS  PubMed  Google Scholar 

  161. Zhang, Z. et al. Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol. Cancer Ther. 16, 1739–1750 (2017).

    CAS  PubMed  Google Scholar 

  162. Christensen, C. L. et al. Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909–922 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  163. Hashiguchi, T. et al. Cyclin-dependent kinase-9 is a therapeutic target in MYC-expressing diffuse large B-cell lymphoma. Mol. Cancer Ther. 18, 1520–1532 (2019).

    CAS  PubMed  Google Scholar 

  164. Rahaman, M. H. et al. Targeting CDK9 for treatment of colorectal cancer. Mol. Oncol. 13, 2178–2193 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  165. Blake, D. R. et al. Application of a MYC degradation screen identifies sensitivity to CDK9 inhibitors in KRAS-mutant pancreatic cancer. Sci. Signal. 12, eaav7259 (2019).

    PubMed Central  PubMed  Google Scholar 

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

  167. Day, M. A. et al. Abstract 1141: CDK9 inhibition is selective for transcriptionally addicted tumors harboring MYC genomic amplifications. Cancer Res. 81 (Suppl. 13), 1141 (2021).

    Google Scholar 

  168. Scheepstra, M., Hekking, K. F. W., van Hijfte, L. & Folmer, R. H. A. Bivalent ligands for protein degradation in drug discovery. Comput. Struct. Biotechnol. J. 17, 160–176 (2019).

  169. Frame, S. et al. Fadraciclib (CYC065), a novel CDK inhibitor, targets key pro-survival and oncogenic pathways in cancer. PLoS ONE 15, e0234103 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  170. Garcia-Cuellar, M. P. et al. Efficacy of cyclin-dependent-kinase 9 inhibitors in a murine model of mixed-lineage leukemia. Leukemia 28, 1427–1435 (2014).

    CAS  PubMed  Google Scholar 

  171. Rahaman, M. H. et al. CDKI-73: an orally bioavailable and highly efficacious CDK9 inhibitor against acute myeloid leukemia. Invest. New Drugs 37, 625–635 (2019).

    CAS  PubMed  Google Scholar 

  172. Barlaam, B. et al. Discovery of AZD4573, a potent and selective inhibitor of CDK9 that enables short duration of target engagement for the treatment of hematological malignancies. J. Med. Chem. 63, 15564–15590 (2020).

    CAS  PubMed  Google Scholar 

  173. Vervoort, S. J. et al. The PP2A-Integrator-CDK9 axis fine-tunes transcription and can be targeted therapeutically in cancer. Cell 184, 3143–3162.e32 (2021). This study demonstrates the interplay between CDK9 and PP2A for the control of Pol II-driven transcription and the efficacy of combined CDK9 inhibition and PP2A activation in pre-clinical cancer models.

    CAS  PubMed  Google Scholar 

  174. Brägelmann, J. et al. Systematic kinase inhibitor profiling identifies CDK9 as a synthetic lethal target in NUT midline carcinoma. Cell Rep. 20, 2833–2845 (2017).

    PubMed Central  PubMed  Google Scholar 

  175. Richters, A. et al. Modulating androgen receptor-driven transcription in prostate cancer with selective CDK9 inhibitors. Cell Chem. Biol. 28, 134–147.e14 (2021).

    CAS  PubMed  Google Scholar 

  176. Chen, S. et al. Androgen receptor serine 81 phosphorylation mediates chromatin binding and transcriptional activation. J. Biol. Chem. 287, 8571–8583 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  177. Gordon, V. et al. CDK9 regulates AR promoter selectivity and cell growth through serine 81 phosphorylation. Mol. Endocrinol. 24, 2267–2280 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  178. Zhang, T. et al. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat. Chem. Biol. 12, 876–884 (2016). This study presents a selective covalent inhibitor of CDK12 and CDK13 and demonstrated its efficacy in reducing the expression of DNA damage response and super-enhancer-associated transcription factor genes in cancer cell lines.

    CAS  PubMed Central  PubMed  Google Scholar 

  179. Geng, M. et al. Targeting CDK12-mediated transcription regulation in anaplastic thyroid carcinoma. Biochem. Biophys. Res. Commun. 520, 544–550 (2019).

    CAS  PubMed  Google Scholar 

  180. Liu, H., Liu, K. & Dong, Z. Targeting CDK12 for cancer therapy: function, mechanism, and drug discovery. Cancer Res. 81, 18–26 (2021).

    CAS  PubMed  Google Scholar 

  181. Quereda, V. et al. Therapeutic targeting of CDK12/CDK13 in triple-negative breast cancer. Cancer Cell 36, 545–558.e7 (2019).

    CAS  PubMed  Google Scholar 

  182. Jiang, B. et al. Discovery and resistance mechanism of a selective CDK12 degrader. Nat. Chem. Biol. 17, 675–683 (2021).

    CAS  PubMed Central  PubMed  Google Scholar 

  183. Johnson, S. F. et al. CDK12 inhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer. Cell Rep. 17, 2367–2381 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  184. 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).

    CAS  PubMed Central  PubMed  Google Scholar 

  185. Liu, Y. et al. Discovery of MFH290: a potent and highly selective covalent inhibitor for cyclin-dependent kinase 12/13. J. Med. Chem. 63, 6708–6726 (2020).

    CAS  PubMed  Google Scholar 

  186. Li, Y. et al. CDK12/13 inhibition induces immunogenic cell death and enhances anti-PD-1 anticancer activity in breast cancer. Cancer Lett. 495, 12–21 (2020). This study highlights the effective combination of the selective CDK12 and CDK13 inhibitor SR-4835 with anti-PD1 immune-checkpoint inhibitors in pre-clinical breast cancer models.

    CAS  PubMed  Google Scholar 

  187. Eick, D. & Geyer, M. The RNA polymerase II carboxy-terminal domain (CTD) code. Chem. Rev. 113, 8456–8490 (2013).

    CAS  PubMed  Google Scholar 

  188. Mayfield, J. E. et al. Tyr1 phosphorylation promotes phosphorylation of Ser2 on the C-terminal domain of eukaryotic RNA polymerase II by P-TEFb. eLife 8, e48725 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  189. Hintermair, C. et al. Threonine-4 of mammalian RNA polymerase II CTD is targeted by Polo-like kinase 3 and required for transcriptional elongation. EMBO J. 31, 2784–2797 (2012).

    CAS  PubMed Central  PubMed  Google Scholar 

  190. Nieto Moreno, N. et al. GSK-3 is an RNA polymerase II phospho-CTD kinase. Nucleic Acids Res. 48, 6068–6080 (2020).

    PubMed Central  PubMed  Google Scholar 

  191. Bonnet, F. et al. Transcription-independent phosphorylation of the RNA polymerase II C-terminal domain (CTD) involves ERK kinases (MEK1/2). Nucleic Acids Res. 27, 4399–4404 (1999).

    CAS  PubMed Central  PubMed  Google Scholar 

  192. Asghar, U. et al. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 14, 130–146 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  193. Hoshii, T. et al. A non-catalytic function of SETD1A regulates cyclin K and the DNA damage response. Cell 172, 1007–1021.e17 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  194. Marx, A. et al. Upregulation of phosphatase 1 nuclear-targeting subunit (PNUTS) is an independent predictor of poor prognosis in prostate cancer. Dis. Markers 2020, 7050146 (2020).

    PubMed Central  PubMed  Google Scholar 

  195. Byrne, M. et al. Phase I study of the PTEFb inhibitor BAY 1251152 in patients with acute myelogenous leukemia. Blood 132 (Suppl. 1), 4055 (2018).

    Google Scholar 

  196. Song, H. et al. Targeting cyclin-dependent kinase 9 sensitizes medulloblastoma cells to chemotherapy. Biochem. Biophys. Res. Commun. 520, 250–256 (2019).

    CAS  PubMed  Google Scholar 

  197. Tee, A. E. et al. Combination therapy with the CDK7 inhibitor and the tyrosine kinase inhibitor exerts synergistic anticancer effects against MYCN-amplified neuroblastoma. Int. J. Cancer 147, 1928–1938 (2020).

    CAS  PubMed  Google Scholar 

  198. McDermott, M. S. J. et al. CDK7 inhibition is effective in all the subtypes of breast cancer: determinants of response and synergy with EGFR inhibition. Cells 9, 638 (2020).

    CAS  PubMed Central  Google Scholar 

  199. Wang, J. et al. CDK7 inhibitor THZ1 enhances antiPD-1 therapy efficacy via the p38α/MYC/PD-L1 signaling in non-small cell lung cancer. J. Hematol. Oncol. 13, 99 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  200. Chen, R. et al. The multi-kinase inhibitor TG02 induces apoptosis and blocks B-cell receptor signaling in chronic lymphocytic leukemia through dual mechanisms of action. Blood Cancer J. 11, 57 (2021).

    PubMed Central  PubMed  Google Scholar 

  201. Liang, K. et al. Targeting processive transcription elongation via SEC disruption for MYC-induced cancer therapy. Cell 175, 766–779.e17 (2018). This study presents KL-1/KL-2 compounds, which impaired Pol II pause release through the disruption of superelongation complex-mediated CDK9-recruitment to delay tumour progression in xenograft models of MYC-driven cancer.

    CAS  PubMed Central  PubMed  Google Scholar 

  202. Krivtsov, A. V. et al. A menin-MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL-rearranged leukemia. Cancer Cell 36, 660–673.e11 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  203. Brzezinka, K. et al. Characterization of the Menin-MLL interaction as therapeutic cancer target. Cancers 12, 201 (2020).

    CAS  PubMed Central  Google Scholar 

  204. Senisterra, G. et al. Small-molecule inhibition of MLL activity by disruption of its interaction with WDR5. Biochem. J. 449, 151–159 (2013).

    CAS  PubMed  Google Scholar 

  205. Karatas, H. et al. High-affinity, small-molecule peptidomimetic inhibitors of MLL1/WDR5 protein-protein interaction. J. Am. Chem. Soc. 135, 669–682 (2013).

    CAS  PubMed  Google Scholar 

  206. Ye, X. et al. The development of inhibitors targeting the mixed lineage leukemia 1 (MLL1)-WD repeat domain 5 protein (WDR5) protein-protein interaction. Curr. Med. Chem. 27, 5530–5542 (2020).

    CAS  PubMed  Google Scholar 

  207. Dölle, A. et al. Design, synthesis, and evaluation of WD-repeat-containing protein 5 (WDR5) degraders. J. Med. Chem. 64, 10682–10710 (2021).

    PubMed  Google Scholar 

  208. Thomas, L. R. et al. Targeting MYC through WDR5. Mol. Cell Oncol. 7, 1709388 (2020).

    PubMed Central  PubMed  Google Scholar 

  209. Erb, M. A. et al. Transcription control by the ENL YEATS domain in acute leukaemia. Nature 543, 270–274 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  210. Garnar-Wortzel, L. et al. Chemical inhibition of ENL/AF9 YEATS domains in acute leukemia. ACS Central Sci. 7, 815–830 (2021).

    CAS  Google Scholar 

  211. Li, X. et al. Structure-guided development of YEATS domain inhibitors by targeting π-π-π stacking. Nat. Chem. Biol. 14, 1140–1149 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  212. Doroshow, D. B., Eder, J. P. & LoRusso, P. M. BET inhibitors: a novel epigenetic approach. Ann. Oncol. 28, 1776–1787 (2017).

    CAS  PubMed  Google Scholar 

  213. Hogg, S. J. et al. Targeting histone acetylation dynamics and oncogenic transcription by catalytic P300/CBP inhibition. Mol. Cell 81, 2183–2200.e13 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Lasko, L. M. et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 550, 128–132 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  215. Hnisz, D. et al. A phase separation model for transcriptional control. Cell 169, 13–23 (2017).

    CAS  PubMed Central  PubMed  Google Scholar 

  216. Klein, I. A. et al. Partitioning of cancer therapeutics in nuclear condensates. Science 368, 1386–1392 (2020). This study demonstrates that physiochemically dependent partitioning of small-molecule cancer therapeutics in phase-separated condensates influences their on-target activity and suggests a mechanism for the selective sensitivity of super-enhancer-associated genes to global transcriptional inhibitors.

    CAS  PubMed Central  PubMed  Google Scholar 

  217. Hao, S. & Baltimore, D. The stability of mRNA influences the temporal order of the induction of genes encoding inflammatory molecules. Nat. Immunol. 10, 281–288 (2009).

    CAS  PubMed Central  PubMed  Google Scholar 

  218. Jia, Q. et al. Oncogenic super-enhancer formation in tumorigenesis and its molecular mechanisms. Exp. Mol. Med. 52, 713–723 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  219. Meisner, N. C. et al. Identification and mechanistic characterization of low-molecular-weight inhibitors for HuR. Nat. Chem. Biol. 3, 508–515 (2007).

    CAS  PubMed  Google Scholar 

  220. Caizzi, L. et al. Efficient RNA polymerase II pause release requires U2 snRNP function. Mol. Cell 81, 1920–1934.e9 (2021). This study demonstrates that U2 small nuclear ribonucleoprotein-dependent splicing is required for efficient CDK9 recruitment and Pol II elongation, highlighting feedback mechanisms between different phases of the transcription cycle.

    CAS  PubMed  Google Scholar 

  221. Li, N. et al. Cyclin C is a haploinsufficient tumour suppressor. Nat. Cell Biol. 16, 1080–1091 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  222. van Delft, F. W. et al. Clonal origins of relapse in ETV6-RUNX1 acute lymphoblastic leukemia. Blood 117, 6247–6254 (2011).

    PubMed  Google Scholar 

  223. Ohata, N. et al. Highly frequent allelic loss of chromosome 6q16-23 in osteosarcoma: involvement of cyclin C in osteosarcoma. Int. J. Mol. Med. 18, 1153–1158 (2006).

    CAS  PubMed  Google Scholar 

  224. Yang, S. et al. Identification of genes with correlated patterns of variations in DNA copy number and gene expression level in gastric cancer. Genomics 89, 451–459 (2007).

    CAS  PubMed  Google Scholar 

  225. Ding, L. et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 455, 1069–1075 (2008).

    CAS  PubMed Central  PubMed  Google Scholar 

  226. Ekumi, K. M. et al. Ovarian carcinoma CDK12 mutations misregulate expression of DNA repair genes via deficient formation and function of the Cdk12/CycK complex. Nucleic Acids Res. 43, 2575–2589 (2015).

    CAS  PubMed Central  PubMed  Google Scholar 

  227. Naidoo, K. et al. Evaluation of CDK12 protein expression as a potential novel biomarker for DNA damage response-targeted therapies in breast cancer. Mol. Cancer Ther. 17, 306–315 (2018).

    CAS  PubMed  Google Scholar 

  228. Quigley, D. A. et al. Genomic hallmarks and structural variation in metastatic prostate. Cancer Cell 174, 758–769.e9 (2018).

    CAS  Google Scholar 

  229. Reimers, M. A. et al. Clinical outcomes in cyclin-dependent kinase 12 mutant advanced prostate cancer. Eur. Urol. 77, 333–341 (2020).

    CAS  PubMed  Google Scholar 

  230. Haesen, D. et al. Recurrent PPP2R1A mutations in uterine cancer act through a dominant-negative mechanism to promote malignant cell growth. Cancer Res. 76, 5719–5731 (2016).

    CAS  PubMed  Google Scholar 

  231. Shih, l-M. et al. Somatic mutations of PPP2R1A in ovarian and uterine carcinomas. Am. J. Pathol. 178, 144C2–1447 (2011).

    Google Scholar 

  232. Kauko, O. et al. PP2A inhibition is a druggable MEK inhibitor resistance mechanism in KRAS-mutant lung cancer cells. Sci. Transl Med. 10, eaaq1093 (2018).

    PubMed Central  PubMed  Google Scholar 

  233. Pippa, R. et al. MYC-dependent recruitment of RUNX1 and GATA2 on the SET oncogene promoter enhances PP2A inactivation in acute myeloid leukemia. Oncotarget 8, 53989–54003 (2017).

    PubMed  Google Scholar 

  234. De Meyer, T. et al. E2Fs mediate a fundamental cell-cycle deregulation in high-grade serous ovarian carcinomas. J. Pathol. 217, 14–20 (2009).

    PubMed  Google Scholar 

  235. Di Fiore, R. et al. RB1 in cancer: different mechanisms of RB1 inactivation and alterations of pRb pathway in tumorigenesis. J. Cell Physiol. 228, 1676–1687 (2013).

    PubMed  Google Scholar 

  236. Fry, E. A., Mallakin, A. & Inoue, K. Translocations involving ETS family proteins in human cancer. Integr. Cancer Sci. Ther. 5, 10.15761/ICST.1000281 (2018).

    PubMed Central  Google Scholar 

  237. Sizemore, G. M. et al. The ETS family of oncogenic transcription factors in solid tumours. Nat. Rev. Cancer 17, 337–351 (2017).

    CAS  PubMed  Google Scholar 

  238. Igelmann, S., Neubauer, H. A. & Ferbeyre, G. STAT3 and STAT5 activation in solid cancers. Cancers 11, 1428 (2019).

    CAS  PubMed Central  Google Scholar 

  239. Shahmarvand, N. et al. Mutations in the signal transducer and activator of transcription family of genes in cancer. Cancer Sci. 109, 926–933 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  240. DiMartino, J. F. et al. The AF10 leucine zipper is required for leukemic transformation of myeloid progenitors by MLL-AF10. Blood 99, 3780–3785 (2002).

    CAS  PubMed  Google Scholar 

  241. Russler-Germain, D. A. et al. The R882H DNMT3A mutation associated with AML dominantly inhibits wild-type DNMT3A by blocking its ability to form active tetramers. Cancer Cell 25, 442–454 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  242. Yan, X. J. et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat. Genet. 43, 309–315 (2011).

    CAS  PubMed  Google Scholar 

  243. Lio, C. J., Yuita, H. & Rao, A. Dysregulation of the TET family of epigenetic regulators in lymphoid and myeloid malignancies. Blood 134, 1487–1497 (2019).

    PubMed Central  PubMed  Google Scholar 

  244. Waitkus, M. S., Diplas, B. H. & Yan, H. Biological role and therapeutic potential of IDH mutations in cancer. Cancer Cell 34, 186–195 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  245. Borrow, J. et al. The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nat. Genet. 14, 33–41 (1996).

    CAS  PubMed  Google Scholar 

  246. Panagopoulos, I. et al. Fusion of the MORF and CBP genes in acute myeloid leukemia with the t(10;16)(q22;p13). Hum. Mol. Genet. 10, 395–404 (2001).

    CAS  PubMed  Google Scholar 

  247. Chaffanet, M. et al. MOZ is fused to p300 in an acute monocytic leukemia with t(8;22). Genes Chromosomes Cancer 28, 138–144 (2000).

    CAS  PubMed  Google Scholar 

  248. Gayther, S. A. et al. Mutations truncating the EP300 acetylase in human cancers. Nat. Genet. 24, 300–303 (2000).

    CAS  PubMed  Google Scholar 

  249. Mullighan, C. G. et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 471, 235–239 (2011).

    CAS  PubMed Central  PubMed  Google Scholar 

  250. Kishimoto, M. et al. Mutations and deletions of the CBP gene in human lung cancer. Clin. Cancer Res. 11, 512–519 (2005).

    CAS  PubMed  Google Scholar 

  251. Levine, A. J. The many faces of p53: something for everyone. J. Mol. Cell Biol. 11, 524–530 (2019).

    CAS  PubMed Central  PubMed  Google Scholar 

  252. Morel, D. et al. Combining epigenetic drugs with other therapies for solid tumours — past lessons and future promise. Nat. Rev. Clin. Oncol. 283, 91–107 (2020).

    Google Scholar 

  253. Gibcus, J. H. & Dekker, J. The hierarchy of the 3D genome. Mol. Cell 49, 773–782 (2013).

    CAS  PubMed Central  PubMed  Google Scholar 

  254. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    CAS  PubMed Central  PubMed  Google Scholar 

  255. Bonev, B. & Cavalli, G. Organization and function of the 3D genome. Nat. Rev. Genet. 17, 661–678 (2016).

    CAS  PubMed  Google Scholar 

  256. Kim, S. & Shendure, J. Mechanisms of interplay between transcription factors and the 3D genome. Mol. Cell 76, 306–319 (2019).

    CAS  PubMed  Google Scholar 

  257. Noble, C. G. et al. Key features of the interaction between Pcf11 CID and RNA polymerase II CTD. Nat. Struct. Mol. Biol. 12, 144–151 (2005).

    CAS  PubMed  Google Scholar 

  258. Becker, R., Loll, B. & Meinhart, A. Snapshots of the RNA processing factor SCAF8 bound to different phosphorylated forms of the carboxyl-terminal domain of RNA polymerase II. J. Biol. Chem. 283, 22659–22669 (2008).

    CAS  PubMed  Google Scholar 

  259. Ni, Z. et al. Control of the RNA polymerase II phosphorylation state in promoter regions by CTD interaction domain-containing proteins RPRD1A and RPRD1B. Transcription 2, 237–242 (2011).

    PubMed Central  PubMed  Google Scholar 

  260. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    PubMed Central  PubMed  Google Scholar 

  261. Boehning, M. et al. RNA polymerase II clustering through carboxy-terminal domain phase separation. Nat. Struct. Mol. Biol. 25, 833–840 (2018).

    CAS  PubMed  Google Scholar 

  262. Boija, A. et al. Transcription factors activate genes through the phase-separation capacity of their activation domains. Cell 175, 1842–1855.e16 (2018).

    CAS  PubMed  Google Scholar 

  263. Lu, H. et al. Phase-separation mechanism for C-terminal hyperphosphorylation of RNA polymerase II. Nature 558, 318–323 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  264. Harlen, K. M. & Churchman, L. S. The code and beyond: transcription regulation by the RNA polymerase II carboxy-terminal domain. Nat. Rev. Mol. Cell Biol. 18, 263–273 (2017).

    CAS  PubMed  Google Scholar 

  265. Archambault, J. et al. An essential component of a C-terminal domain phosphatase that interacts with transcription factor IIF in Saccharomyces cerevisiae. Proc. Natl Acad. Sci. USA 94, 14300–14305 (1997).

    CAS  PubMed Central  PubMed  Google Scholar 

  266. Kobor, M. S. et al. An unusual eukaryotic protein phosphatase required for transcription by RNA polymerase II and CTD dephosphorylation in S. cerevisiae. Mol. Cell 4, 55–62 (1999).

    CAS  PubMed  Google Scholar 

  267. Krishnamurthy, S. et al. Ssu72 Is an RNA polymerase II CTD phosphatase. Mol. Cell 14, 387–394 (2004).

    CAS  PubMed  Google Scholar 

  268. Schreieck, A. et al. RNA polymerase II termination involves C-terminal-domain tyrosine dephosphorylation by CPF subunit Glc7. Nat. Struct. Mol. Biol. 21, 175–179 (2014).

    CAS  PubMed Central  PubMed  Google Scholar 

  269. Parua, P. K. et al. A Cdk9-PP1 switch regulates the elongation-termination transition of RNA polymerase II. Nature 558, 460–464 (2018).

    CAS  PubMed Central  PubMed  Google Scholar 

  270. Parua, P. K. et al. Distinct Cdk9-phosphatase switches act at the beginning and end of elongation by RNA polymerase II. Nat. Commun. 11, 4338 (2020). Together with Parua et al. (2018), this study demonstrates distinct regulatory roles for protein phosphatases, PP1 and PP4, highlighting the balance between tCDK kinase activity and phosphatase activity at multiple phases of Pol II transcription cycles.

    CAS  PubMed Central  PubMed  Google Scholar 

  271. Sen, I. et al. DAF-16/FOXO requires Protein Phosphatase 4 to initiate transcription of stress resistance and longevity promoting genes. Nat. Commun. 11, 138 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  272. Huang, K. L. et al. Integrator recruits protein phosphatase 2A to prevent pause release and facilitate transcription termination. Mol. Cell 80, 345–358.e9 (2020).

    CAS  PubMed Central  PubMed  Google Scholar 

  273. Zheng, H. et al. Identification of Integrator-PP2A complex (INTAC), an RNA polymerase II phosphatase. Science 370, eabb5872 (2020). Together with Huang and al. (2020), This study presents the identification of PP2A-dependent control of Pol II transcription cycles and structural characterization of the human Integator–PP2A complex.

    CAS  PubMed  Google Scholar 

  274. Kavela, S. et al. PNUTS functions as a proto-oncogene by sequestering PTEN. Cancer Res. 73, 205–214 (2013).

    CAS  PubMed  Google Scholar 

  275. Dingar, D. et al. MYC dephosphorylation by the PP1/PNUTS phosphatase complex regulates chromatin binding and protein stability. Nat. Commun. 9, 3502–350 (2018).

    PubMed Central  PubMed  Google Scholar 

  276. Farrington, C. C. et al. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers. J. Biol. Chem. 295, 757–770 (2020).

    PubMed  Google Scholar 

  277. Leonard, D. et al. Selective PP2A enhancement through biased heterotrimer stabilization. Cell 181, 688–701.e16 (2020). Together with Farrington et al. (2020), this study demonstrates the therapeutic properties and biochemical activities of small-molecule activators of protein phosphatase 2A, which exhibit efficacy in preclinical models of MYC and KRAS mutant-driven cancer.

    CAS  PubMed Central  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

All authors made substantial contributions to researching data for the article, the discussion of content, writing and editing of the manuscript before final submission.

Corresponding authors

Correspondence to Nathanael S. Gray or Ricky W. Johnstone.

Ethics declarations

Competing Interests

The Johnstone laboratory receives research support from Roche, BMS, AstraZeneca and MecRx. R.W.J. is a scientific consultant and shareholder in MecRx. N.S.G. is a founder, science advisor and equity holder in C4, Syros, Soltego, B2S, Jengu, Allorion, EoCys and Larkspur Pharmaceuticals. The Gray laboratory receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Janssen, Kinogen, Voronoi, Arbella, Deerfield and Sanofi. N.S.G. and N.K. are inventors on a patent application covering THZ1, which is licensed to a company co-founded by N.S.G.. N.K. is an employee of Kymera Therapeutics. S.J.V., J.R.D. and M.T. declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks Ivan D’Orso, Shudong Wang, who co-reviewed with Muhammed Rahaman, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vervoort, S.J., Devlin, J.R., Kwiatkowski, N. et al. Targeting transcription cycles in cancer. Nat Rev Cancer 22, 5–24 (2022). https://doi.org/10.1038/s41568-021-00411-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00411-8

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