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:

Bromodomains: a new target class for drug development

Abstract

Less than a decade ago, it was shown that bromodomains, acetyl lysine ‘reader’ modules found in proteins with varied functions, were highly tractable small-molecule targets. This is an unusual property for protein–protein or protein–peptide interaction domains, and it prompted a wave of chemical probe discovery to understand the biological potential of new agents that targeted bromodomains. The original examples, inhibitors of the bromodomain and extra-terminal (BET) class of bromodomains, showed enticing anti-inflammatory and anticancer activities, and several compounds have since advanced to human clinical trials. Here, we review the current state of BET inhibitor biology in relation to clinical development, and we discuss the next wave of bromodomain inhibitors with clinical potential in oncology and non-oncology indications. The lessons learned from BET inhibitor programmes should affect efforts to develop drugs that target non-BET bromodomains and other epigenetic readers.

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: Bromodomain structure and function.
Fig. 2: Bromodomain proteins and inhibition mechanisms.
Fig. 3: Ubiquitin-mediated proteolysis re-engineered to eliminate a therapeutic target of interest.
Fig. 4: BETi and resistance mechanisms.

Similar content being viewed by others

References

  1. Filippakopoulos, P. et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell 149, 214–231 (2012). This now-classic review of the BD protein family is an essential introduction for those new to the field.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Vollmuth, F. & Geyer, M. Interaction of propionylated and butyrylated histone H3 lysine marks with Brd4 bromodomains. Angew. Chem. Int. Ed. 49, 6768–6772 (2010).

    CAS  Google Scholar 

  3. Hewings, D. S. et al. Progress in the development and application of small molecule inhibitors of bromodomain-acetyl-lysine interactions. J. Med. Chem. 55, 9393–9413 (2012).

    CAS  PubMed  Google Scholar 

  4. Huang, D., Rossini, E., Steiner, S. & Caflisch, A. Structured water molecules in the binding site of bromodomains can be displaced by cosolvent. ChemMedChem 9, 573–579 (2014).

    CAS  PubMed  Google Scholar 

  5. Flynn, E. M. et al. A subset of human bromodomains recognizes butyryllysine and crotonyllysine histone peptide modifications. Structure 23, 1801–1814 (2015).

    CAS  PubMed  Google Scholar 

  6. Fedorov, O. et al. Selective targeting of the BRG/PB1 bromodomains impairs embryonic and trophoblast stem cell maintenance. Sci. Adv. 1, e1500723 (2015).

    PubMed  PubMed Central  Google Scholar 

  7. Crawford, T. D. et al. Diving into the water: inducible binding conformations for BRD4, TAF1(2), BRD9, and CECR2 bromodomains. J. Med. Chem. 59, 5391–5402 (2016).

    CAS  PubMed  Google Scholar 

  8. Liu, J. et al. The polar warhead of a TRIM24 bromodomain inhibitor rearranges a water-mediated interaction network. FEBS J. 284, 1082–1095 (2017).

    CAS  PubMed  Google Scholar 

  9. Aldeghi, M. et al. Large-scale analysis of water stability in bromodomain binding pockets with grand canonical Monte Carlo. Commun. Chem. 1, 19 (2018).

    PubMed  PubMed Central  Google Scholar 

  10. Shadrick, W. R. et al. Exploiting a water network to achieve enthalpy-driven, bromodomain-selective BET inhibitors. Bioorg. Med. Chem. 26, 25–36 (2018).

    CAS  PubMed  Google Scholar 

  11. Harner, M. J., Chauder, B. A., Phan, J. & Fesik, S. W. Fragment-based screening of the bromodomain of ATAD2. J. Med. Chem. 57, 9687–9692 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, X., Chen, K., Wu, Y. D. & Wiest, O. Protein dynamics and structural waters in bromodomains. PLOS ONE 12, e0186570 (2017).

    PubMed  PubMed Central  Google Scholar 

  13. Jennings, L. E., Measures, A. R., Wilson, B. G. & Conway, S. J. Phenotypic screening and fragment-based approaches to the discovery of small-molecule bromodomain ligands. Future Med. Chem. 6, 179–204 (2014).

    CAS  PubMed  Google Scholar 

  14. Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 11, 384–400 (2012).

    CAS  PubMed  Google Scholar 

  15. Filippakopoulos, P. & Knapp, S. Targeting bromodomains: epigenetic readers of lysine acetylation. Nat. Rev. Drug Discov. 13, 337–356 (2014).

    CAS  PubMed  Google Scholar 

  16. Sanchez, R., Meslamani, J. & Zhou, M. M. The bromodomain: from epigenome reader to druggable target. Biochim. Biophys. Acta 1839, 676–685 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Ferri, E., Petosa, C. & McKenna, C. E. Bromodomains: structure, function and pharmacology of inhibition. Biochem. Pharmacol. 106, 1–18 (2016).

    CAS  PubMed  Google Scholar 

  18. Shi, J. & Vakoc, C. R. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol. Cell 54, 728–736 (2014).

    CAS  PubMed  Google Scholar 

  19. Xu, Y. & Vakoc, C. R. Targeting cancer cells with BET bromodomain inhibitors. Cold Spring Harb. Perspect. Med. 7, a026674 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Brand, M. et al. Small molecule inhibitors of bromodomain-acetyl-lysine interactions. ACS Chem. Biol. 10, 22–39 (2015).

    CAS  PubMed  Google Scholar 

  21. Romero, F. A. et al. Disrupting acetyl-lysine recognition: progress in the development of bromodomain inhibitors. J. Med. Chem. 59, 1271–1298 (2016).

    CAS  PubMed  Google Scholar 

  22. Theodoulou, N. H., Tomkinson, N. C., Prinjha, R. K. & Humphreys, P. G. Clinical progress and pharmacology of small molecule bromodomain inhibitors. Curr. Opin. Chem. Biol. 33, 58–66 (2016).

    CAS  PubMed  Google Scholar 

  23. Clegg, M. A., Tomkinson, N. C. O., Prinjha, R. K. & Humphreys, P. G. Advancements in the development of non-BET bromodomain chemical probes. ChemMedChem 14, 362–385 (2019). This review is a comprehensive update of the 2016 ChemMedChem review from the same group.

    CAS  PubMed  Google Scholar 

  24. Liu, Z. et al. Drug discovery targeting bromodomain-containing protein 4. J. Med. Chem. 60, 4533–4558 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Perez-Salvia, M. & Esteller, M. Bromodomain inhibitors and cancer therapy: From structures to applications. Epigenetics 12, 323–339 (2017).

    PubMed  Google Scholar 

  26. Marchand, J. R. & Caflisch, A. Binding mode of acetylated histones to bromodomains: variations on a common motif. ChemMedChem 10, 1327–1333 (2015).

    CAS  PubMed  Google Scholar 

  27. Lloyd, J. T. & Glass, K. C. Biological function and histone recognition of family IV bromodomain-containing proteins. J. Cell. Physiol. 233, 1877–1886 (2018).

    CAS  PubMed  Google Scholar 

  28. Wu, Q. et al. A chemical toolbox for the study of bromodomains and epigenetic signaling. Nat. Commun. 10, 1915 (2019).

    PubMed  PubMed Central  Google Scholar 

  29. Fujisawa, T. & Filippakopoulos, P. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat. Rev. Mol. Cell Biol. 18, 246–262 (2017). This review surveys known biochemical functions of BD-containing proteins, including incorporation into multi-protein complexes and the presence of additional functional domains. In addition, it provides information curated from cancer sequencing databases (for example, The Cancer Genome Atlas) on genomic alterations found in BD proteins (translocations, mutations and expression changes), and they map onto structures those point mutations that occur in the BDs themselves, providing insight into whether the mutations are likely to alter BD function.

    CAS  PubMed  Google Scholar 

  30. Tough, D. F., Tak, P. P., Tarakhovsky, A. & Prinjha, R. K. Epigenetic drug discovery: breaking through the immune barrier. Nat. Rev. Drug Discov. 15, 835–853 (2016).

    CAS  PubMed  Google Scholar 

  31. Jain, A. K. & Barton, M. C. Bromodomain histone readers and cancer. J. Mol. Biol. 429, 2003–2010 (2017).

    CAS  PubMed  Google Scholar 

  32. Pfister, S. X. & Ashworth, A. Marked for death: targeting epigenetic changes in cancer. Nat. Rev. Drug Discov. 16, 241–263 (2017).

    CAS  PubMed  Google Scholar 

  33. Shortt, J., Ott, C. J., Johnstone, R. W. & Bradner, J. E. A chemical probe toolbox for dissecting the cancer epigenome. Nat. Rev. Cancer 17, 160–183 (2017).

    CAS  PubMed  Google Scholar 

  34. Gacias, M. et al. Selective chemical modulation of gene transcription favors oligodendrocyte lineage progression. Chem. Biol. 21, 841–854 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Raux, B. et al. Exploring selective inhibition of the first bromodomain of the human bromodomain and extra-terminal domain (BET) proteins. J. Med. Chem. 59, 1634–1641 (2016).

    CAS  PubMed  Google Scholar 

  36. Cheung, K. et al. BET N-terminal bromodomain inhibition selectively blocks Th17 cell differentiation and ameliorates colitis in mice. Proc. Natl Acad. Sci. USA 114, 2952–2957 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Jahagirdar, R. et al. RVX-297, a BET bromodomain inhibitor, has therapeutic effects in preclinical models of acute inflammation and autoimmune disease. Mol. Pharmacol. 92, 694–706 (2017).

    CAS  PubMed  Google Scholar 

  38. Odore, E. et al. Phase I population pharmacokinetic assessment of the oral bromodomain inhibitor OTX015 in patients with haematologic malignancies. Clin. Pharmacokinet. 55, 397–405 (2016).

    CAS  PubMed  Google Scholar 

  39. Amorim, S. et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 3, e196–e204 (2016).

    PubMed  Google Scholar 

  40. Berthon, C. et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 3, e186–e195 (2016).

    PubMed  Google Scholar 

  41. Massard, C. et al. A phase Ib trial with MK-8628/OTX015, a small molecule inhibitor of bromodomain (BRD) and extra-terminal (BET). proteins, in patients with selected advanced solid tumors. Eur. J. Cancer 69 (Suppl. 1), S2–S3 (2016).

    Google Scholar 

  42. Abramson, J. S. et al. BET inhibitor CPI-0610 is well tolerated and induces responses in diffuse large B-cell lymphoma and follicular lymphoma: preliminary analysis of an ongoing phase 1 study. Blood 126, 1491 (2015).

    Google Scholar 

  43. O’Dwyer, P. J. et al. GSK525762, a selective bromodomain (BRD) and extra terminal protein (BET) inhibitor: results from part 1 of a phase I/II open-label single-agent study in patients with NUT midline carcinoma (NMC) and other cancers [abstract]. Cancer Res. 76 (Suppl. 14), CT014 (2016).

    Google Scholar 

  44. Mottamal, M., Zheng, S., Huang, T. L. & Wang, G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 20, 3898–3941 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kuter, D. J. Managing thrombocytopenia associated with cancer chemotherapy. Oncology 29, 282–294 (2015).

    PubMed  Google Scholar 

  46. Postel-Vinay, S. et al. First-in-human phase I study of the bromodomain and extraterminal motif inhibitor BAY 1238097: emerging pharmacokinetic/pharmacodynamic relationship and early termination due to unexpected toxicity. Eur. J. Cancer 109, 103–110 (2019).

    CAS  PubMed  Google Scholar 

  47. Kati, W. ABBV-744: a first-in-class highly BDII-selective BET bromodomain inhibitor [abstract]. Cancer Res. 78 (Suppl. 13), DDT01-05 (2018).

    Google Scholar 

  48. Tanaka, M. et al. Design and characterization of bivalent BET inhibitors. Nat. Chem. Biol. 12, 1089–1096 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Waring, M. J. et al. Potent and selective bivalent inhibitors of BET bromodomains. Nat. Chem. Biol. 12, 1097–1104 (2016).

    CAS  PubMed  Google Scholar 

  50. Ren, C. et al. Spatially constrained tandem bromodomain inhibition bolsters sustained repression of BRD4 transcriptional activity for TNBC cell growth. Proc. Natl Acad. Sci. USA 115, 7949–7954 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Rhyasen, G. W. et al. AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies. Mol. Cancer Ther. 15, 2563–2574 (2016).

    CAS  PubMed  Google Scholar 

  52. French, C. NUT midline carcinoma. Nat. Rev. Cancer 14, 149–150 (2014).

    CAS  PubMed  Google Scholar 

  53. Stathis, A. et al. Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to the targeted bromodomain inhibitor OTX015/MK-8628. Cancer Discov. 6, 492–500 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Duan, Q. et al. BET bromodomain inhibition suppresses innate inflammatory and profibrotic transcriptional networks in heart failure. Sci. Transl Med. 9, eaah5084 (2017).

    PubMed  PubMed Central  Google Scholar 

  55. Nicholls, S. J. et al. Selective BET protein inhibition with apabetalone and cardiovascular events: a pooled analysis of trials in patients with coronary artery disease. Am. J. Cardiovasc. Drugs 18, 109–115 (2018).

    CAS  PubMed  Google Scholar 

  56. Gilham, D. et al. RVX-208, a BET-inhibitor for treating atherosclerotic cardiovascular disease, raises ApoA-I/HDL and represses pathways that contribute to cardiovascular disease. Atherosclerosis 247, 48–57 (2016).

    CAS  PubMed  Google Scholar 

  57. Siebel, A. L. et al. Effects of the BET-inhibitor, RVX-208 on the HDL lipidome and glucose metabolism in individuals with prediabetes: a randomized controlled trial. Metabolism 65, 904–914 (2016).

    CAS  PubMed  Google Scholar 

  58. Ghosh, G. C., Bhadra, R., Ghosh, R. K., Banerjee, K. & Gupta, A. RVX 208: a novel BET protein inhibitor, role as an inducer of apo A-I/HDL and beyond. Cardiovasc. Ther. 35, e12265 (2017).

    Google Scholar 

  59. Wasiak, S. et al. Downregulation of the complement cascade in vitro, in mice and in patients with cardiovascular disease by the BET protein inhibitor apabetalone (RVX-208). J. Cardiovasc. Transl Res. 10, 337–347 (2017).

    PubMed  PubMed Central  Google Scholar 

  60. Mirguet, O. et al. From ApoA1 upregulation to BET family bromodomain inhibition: discovery of I-BET151. Bioorg. Med. Chem. Lett. 22, 2963–2967 (2012).

    CAS  PubMed  Google Scholar 

  61. Kempen, H. J. et al. Stimulation of hepatic apolipoprotein A-I production by novel thieno-triazolodiazepines: roles of the classical benzodiazepine receptor, PAF receptor, and bromodomain binding. Lipid Insights 6, 47–54 (2013).

    PubMed  PubMed Central  Google Scholar 

  62. Gosmini, R. et al. The discovery of I-BET726 (GSK1324726A), a potent tetrahydroquinoline ApoA1 up-regulator and selective BET bromodomain inhibitor. J. Med. Chem. 57, 8111–8131 (2014).

    CAS  PubMed  Google Scholar 

  63. Picaud, S. et al. RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain. Proc. Natl Acad. Sci. USA 110, 19754–19759 (2013). This article is the first to provide a clear explanation of why RVX-208 showed a different pharmacology from other BETis. The authors show the selectivity of the inhibitor for BD2 and demonstrate the mild transcriptional effects resulting from BD2 inhibition as compared with pan-BET, BD1 and BD2 inhibition.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Wasiak, S. et al. Data on gene and protein expression changes induced by apabetalone (RVX-208) in ex vivo treated human whole blood and primary hepatocytes. Data Brief 8, 1280–1288 (2016).

    PubMed  PubMed Central  Google Scholar 

  65. Boehm, D. et al. BET bromodomain-targeting compounds reactivate HIV from latency via a Tat-independent mechanism. Cell Cycle 12, 452–462 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Li, Z., Guo, J., Wu, Y. & Zhou, Q. The BET bromodomain inhibitor JQ1 activates HIV latency through antagonizing Brd4 inhibition of Tat-transactivation. Nucleic Acids Res. 41, 277–287 (2013). This article and the one preceding it are the first articles to show that BETi may have a therapeutic impact on HIV infection, even if the relationship of BRD4 with HIV Tat and P-TEFb had been known for several years.

    CAS  PubMed  Google Scholar 

  67. Lu, P. et al. The BET inhibitor OTX015 reactivates latent HIV-1 through P-TEFb. Sci. Rep. 6, 24100 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Keck, K. M. et al. BET inhibitors block the Epstein-Barr virus lytic cycle at two distinct steps. J. Biol. Chem. 292, 13284–13295 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Mietton, F. et al. Selective BET bromodomain inhibition as an antifungal therapeutic strategy. Nat. Commun. 8, 15482 (2017). This report is the first of the generation of in vitro active, fungal-selective BDis for the elimination of a human pathogen. The authors describe a chemical starting point for future exploration of a potent anti-fungal agent with limited human toxicity in infection models.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Schulz, D. et al. Bromodomain proteins contribute to maintenance of bloodstream form stage identity in the African trypanosome. PLOS Biol. 13, e1002316 (2015).

    PubMed  PubMed Central  Google Scholar 

  71. Drouin, L. et al. Structure enabled design of BAZ2-ICR, a chemical probe targeting the bromodomains of BAZ2A and BAZ2B. J. Med. Chem. 58, 2553–2559 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Chen, P. et al. Discovery and characterization of GSK2801, a selective chemical probe for the bromodomains BAZ2A and BAZ2B. J. Med. Chem. 59, 1410–1424 (2016).

    CAS  PubMed  Google Scholar 

  73. Humphreys, P. G. et al. Discovery of a potent, cell penetrant, and selective p300/CBP-associated factor (PCAF)/general control nonderepressible 5 (GCN5) bromodomain chemical probe. J. Med. Chem. 60, 695–709 (2017).

    CAS  PubMed  Google Scholar 

  74. Moustakim, M. et al. Discovery of a PCAF bromodomain chemical probe. Angew. Chem. Int. Ed. 56, 827–831 (2017).

    CAS  Google Scholar 

  75. Crawford, T. D. et al. GNE-886: a potent and selective inhibitor of the cat eye syndrome chromosome region candidate 2 bromodomain (CECR2). ACS Med. Chem. Lett. 8, 737–741 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Attar, N. & Kurdistani, S. K. Exploitation of EP3000 and CREBBP lysine acetyltransferases by cancer. Cold Spring Harb. Perspect. Med. 7, a026534 (2017).

    PubMed  PubMed Central  Google Scholar 

  77. Sachchidanand et al. Target structure-based discovery of small molecules that block human p53 and CREB binding protein association. Chem. Biol. 13, 81–90 (2006).

    CAS  PubMed  Google Scholar 

  78. Borah, J. C. et al. A small molecule binding to the coactivator CREB-binding protein blocks apoptosis in cardiomyocytes. Chem. Biol. 18, 531–541 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Hay, D. A. et al. Discovery and optimization of small-molecule ligands for the CBP/p300 bromodomains. J. Am. Chem. Soc. 136, 9308–9319 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chekler, E. L. et al. Transcriptional profiling of a selective CREB binding protein bromodomain inhibitor highlights therapeutic opportunities. Chem. Biol. 22, 1588–1596 (2015).

    CAS  PubMed  Google Scholar 

  81. Picaud, S. et al. Generation of a selective small molecule inhibitor of the CBP/p300 bromodomain for leukemia therapy. Cancer Res. 75, 5106–5119 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Abruzzese, M. P. et al. Inhibition of bromodomain and extra-terminal (BET) proteins increases NKG2D ligand MICA expression and sensitivity to NK cell-mediated cytotoxicity in multiple myeloma cells: role of cMYC-IRF4-miR-125b interplay. J. Hematol. Oncol. 9, 134 (2016).

    PubMed  PubMed Central  Google Scholar 

  83. Conery, A. R. et al. Bromodomain inhibition of the transcriptional coactivators CBP/EP300 as a therapeutic strategy to target the IRF4 network in multiple myeloma. eLife 5, e10483 (2016). This article describes a CREBBP-dependent and EP300-dependent transcriptional programme, identified by chemical biology approaches, that converges on MYC but is distinct from a BET-driven transcriptional programme. This article not only highlights IRF4 as a potential biomarker for future development of CREBBP and EP300 inhibitors in myeloma but also points to the possibility of combined inhibition of BET, CREBBP and EP300 BDs as a therapeutic strategy.

    PubMed  PubMed Central  Google Scholar 

  84. Hammitzsch, A. et al. CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses. Proc. Natl Acad. Sci. USA 112, 10768–10773 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Mele, D. A. et al. BET bromodomain inhibition suppresses TH17-mediated pathology. J. Exp. Med. 210, 2181–2190 (2013). This article and that of Hammitzsch et al. (2015) suggest that, similar to the MYC programme, the T H 17 transcriptional programme is regulated by both CREBBP–EP300 and BET BDs. Whether inhibition of a single node or both nodes will have better efficacy or therapeutic index in autoimmune disorders remains to be tested.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Ebrahimi, A. et al. Bromodomain inhibition of the coactivators CBP/EP300 facilitate cellular reprogramming. Nat. Chem. Biol. 15, 519–528 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Olzscha, H., Fedorov, O., Kessler, B. M., Knapp, S. & La Thangue, N. B. CBP/p300 bromodomains regulate amyloid-like protein aggregation upon aberrant lysine acetylation. Cell Chem. Biol. 24, 9–23 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Taylor, A. M. et al. Fragment-based discovery of a selective and cell-active benzodiazepinone CBP/EP300 bromodomain inhibitor (CPI-637). ACS Med. Chem. Lett. 7, 531–536 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Ghosh, S. et al. Regulatory T cell modulation by CBP/EP300 bromodomain inhibition. J. Biol. Chem. 291, 13014–13027 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Crawford, T. D. et al. Discovery of a potent and selective in vivo probe (GNE-272) for the bromodomains of CBP/EP300. J. Med. Chem. 59, 10549–10563 (2016).

    CAS  PubMed  Google Scholar 

  91. Jin, L. et al. Therapeutic targeting of the CBP/p300 bromodomain blocks the growth of castration-resistant prostate cancer. Cancer Res. 77, 5564–5575 (2017). This article is the first to demonstrate the feasibility of generating a molecule that is highly selective for CREBBP and EP300 BDs relative to BET BDs through structure-based optimization starting with the molecule described in Crawford et al. (2016). The inhibitor served as a starting point for the highly optimized molecule described in Romero et al. (2017).

    CAS  PubMed  Google Scholar 

  92. Raisner, R. et al. Enhancer activity requires CBP/P300 bromodomain-dependent histone H3K27 acetylation. Cell Rep. 24, 1722–1729 (2018).

    CAS  PubMed  Google Scholar 

  93. Romero, F. A. et al. GNE-781, a highly advanced potent and selective bromodomain inhibitor of cyclic adenosine monophosphate response element binding protein, binding protein (CBP). J. Med. Chem. 60, 9162–9183 (2017).

    CAS  PubMed  Google Scholar 

  94. Pegg, N. A., Taddei, D. M. A., Brown, R. & Keen, C. M. Isoxazolyl substituted imidazopyridines. WO2016170323 (2016).

  95. Pegg, N. et al. Characterisation of CCS1477: a novel small molecule inhibitor of p300/CBP for the treatment of castration resistant prostate cancer [abstract]. J. Clin. Oncol. 35 (Suppl. 15), 11590 (2017).

    Google Scholar 

  96. Kadoch, C. & Crabtree, G. R. Mammalian SWI/SNF chromatin remodeling complexes and cancer: mechanistic insights gained from human genomics. Sci. Adv. 1, e1500447 (2015).

    PubMed  PubMed Central  Google Scholar 

  97. Kadoch, C. et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet. 45, 592–601 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Wang, X., Haswell, J. R. & Roberts, C. W. Molecular pathways: SWI/SNF (BAF) complexes are frequently mutated in cancer—mechanisms and potential therapeutic insights. Clin. Cancer Res. 20, 21–27 (2014).

    PubMed  Google Scholar 

  99. Oike, T. et al. A synthetic lethality-based strategy to treat cancers harboring a genetic deficiency in the chromatin remodeling factor BRG1. Cancer Res. 73, 5508–5518 (2013).

    CAS  PubMed  Google Scholar 

  100. Hoffman, G. R. et al. Functional epigenetics approach identifies BRM/SMARCA2 as a critical synthetic lethal target in BRG1-deficient cancers. Proc. Natl Acad. Sci. USA 111, 3128–3133 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Kaelin, W. G. Jr The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5, 689–698 (2005).

    CAS  PubMed  Google Scholar 

  102. Wang, X. et al. Oncogenesis caused by loss of the SNF5 tumor suppressor is dependent on activity of BRG1, the ATPase of the SWI/SNF chromatin remodeling complex. Cancer Res. 69, 8094–8101 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Helming, K. C. et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nat. Med. 20, 251–254 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Lissanu Deribe, Y. et al. Mutations in the SWI/SNF complex induce a targetable dependence on oxidative phosphorylation in lung cancer. Nat. Med. 24, 1047–1057 (2018).

    CAS  PubMed  Google Scholar 

  105. Molina, J. R. et al. An inhibitor of oxidative phosphorylation exploits cancer vulnerability. Nat. Med. 24, 1036–1046 (2018).

    CAS  PubMed  Google Scholar 

  106. Vangamudi, B. et al. The SMARCA2/4 ATPase domain surpasses the bromodomain as a drug target in SWI/SNF-mutant cancers: insights from cDNA rescue and PFI-3 inhibitor studies. Cancer Res. 75, 3865–3878 (2015). This important study invalidates the BRM BD as a therapeutic target in BRG1-mutant cancers.

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Papillon, J. P. N. et al. Discovery of orally active inhibitors of Brahma homolog (BRM)/SMARCA2 ATPase activity for the treatment of Brahma related gene 1 (BRG1)/SMARCA4-mutant cancers. J. Med. Chem. 61, 10155–10172 (2018).

    CAS  PubMed  Google Scholar 

  108. Farnaby, W. et al. BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design. Nat. Chem. Biol. https://doi.org/10.1038/s41589-019-0294-6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Gerstenberger, B. S. et al. Identification of a chemical probe for family VIII bromodomains through optimization of a fragment hit. J. Med. Chem. 59, 4800–4811 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Sutherell, C. L. et al. Identification and development of 2,3-dihydropyrrolo[1,2-a]quinazolin-5(1H)-one inhibitors targeting bromodomains within the switch/sucrose nonfermenting complex. J. Med. Chem. 59, 5095–5101 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Albrecht, B. K. et al. Therapeutic pyridazine compounds and uses thereof. WO2016138114A1 (2016).

  112. Morrison, E. A. et al. DNA binding drives the association of BRG1/hBRM bromodomains with nucleosomes. Nat. Commun. 8, 16080 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Miller, T. C. et al. A bromodomain-DNA interaction facilitates acetylation-dependent bivalent nucleosome recognition by the BET protein BRDT. Nat. Commun. 7, 13855 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Porter, E. G. & Dykhuizen, E. C. Individual bromodomains of polybromo-1 contribute to chromatin association and tumor suppression in clear cell renal carcinoma. J. Biol. Chem. 292, 2601–2610 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Oppikofer, M. et al. Non-canonical reader modules of BAZ1A promote recovery from DNA damage. Nat. Commun. 8, 862 (2017).

    PubMed  PubMed Central  Google Scholar 

  116. Mashtalir, N. et al. Modular organization and assembly of SWI/SNF family chromatin remodeling complexes. Cell 175, 1272–1288 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Picaud, S. et al. 9H-purine scaffold reveals induced-fit pocket plasticity of the BRD9 bromodomain. J. Med. Chem. 58, 2718–2736 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Clark, P. G. et al. LP99: discovery and synthesis of the first selective BRD7/9 bromodomain inhibitor. Angew. Chem. Int. Ed. 54, 6217–6221 (2015).

    CAS  Google Scholar 

  119. Theodoulou, N. H. et al. Discovery of I-BRD9, a selective cell active chemical probe for bromodomain containing protein 9 inhibition. J. Med. Chem. 59, 1425–1439 (2016).

    CAS  PubMed  Google Scholar 

  120. Hay, D. A. et al. Design and synthesis of potent and selective inhibitors of BRD7 and BRD9 bromodomains. Med. Chem. Commun. 6, 1381–1386 (2015).

    CAS  Google Scholar 

  121. Martin, L. J. et al. Structure-based design of an in vivo active selective BRD9 inhibitor. J. Med. Chem. 59, 4462–4475 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Crawford, T. D. et al. Inhibition of bromodomain-containing protein 9 for the prevention of epigenetically-defined drug resistance. Bioorg. Med. Chem. Lett. 27, 3534–3541 (2017).

    CAS  PubMed  Google Scholar 

  123. Karim, R. M. & Schonbrunn, E. An advanced tool to interrogate BRD9. J. Med. Chem. 59, 4459–4461 (2016).

    CAS  PubMed  Google Scholar 

  124. Hohmann, A. F. et al. Sensitivity and engineered resistance of myeloid leukemia cells to BRD9 inhibition. Nat. Chem. Biol. 12, 672–679 (2016). This exceptionally thorough study investigates the biological role of BRD9. The authors make use of BD chimeric proteins and chemical probes to differentiate the function of the BRD9 BD from the closely related BRD7 BD and from BET-class BD proteins.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Sharma, S. V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Remillard, D. et al. Degradation of the BAF complex factor BRD9 by heterobifunctional ligands. Angew. Chem. Int. Ed. 56, 5738–5743 (2017).

    CAS  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  128. Michel, B. C. et al. A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nat. Cell Biol. 20, 1410–1420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Albright, S. R. & Tjian, R. TAFs revisited: more data reveal new twists and confirm old ideas. Gene 242, 1–13 (2000).

    CAS  PubMed  Google Scholar 

  130. Nogales, E., Louder, R. K. & He, Y. Structural insights into the eukaryotic transcription initiation machinery. Annu. Rev. Biophys. 46, 59–83 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Jacobson, R. H., Ladurner, A. G., King, D. S. & Tjian, R. Structure and function of a human TAFII250 double bromodomain module. Science 288, 1422–1425 (2000).

    CAS  PubMed  Google Scholar 

  132. Bouche, L. et al. Benzoisoquinolinediones as potent and selective inhibitors of BRPF2 and TAF1/TAF1L bromodomains. J. Med. Chem. 60, 4002–4022 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Wang, S. et al. GNE-371, a potent and selective chemical probe for the second bromodomains of human transcription-initiation-factor TFIID subunit 1 and transcription-initiation-factor TFIID subunit 1-like. J. Med. Chem. 61, 9301–9315 (2018).

    CAS  PubMed  Google Scholar 

  134. Sdelci, S. et al. Mapping the chemical chromatin reactivation landscape identifies BRD4-TAF1 cross-talk. Nat. Chem. Biol. 12, 504–510 (2016).

    CAS  PubMed  Google Scholar 

  135. Suh, J. L. et al. Quantitative characterization of bivalent probes for a dual bromodomain protein, transcription initiation factor TFIID subunit 1. Biochemistry 57, 2140–2149 (2018).

    CAS  PubMed  Google Scholar 

  136. Klein, B. J., Lalonde, M. E., Cote, J., Yang, X. J. & Kutateladze, T. G. Crosstalk between epigenetic readers regulates the MOZ/MORF HAT complexes. Epigenetics 9, 186–193 (2014).

    CAS  PubMed  Google Scholar 

  137. Yang, X. J. MOZ and MORF acetyltransferases: molecular interaction, animal development and human disease. Biochim. Biophys. Acta 1853, 1818–1826 (2015).

    CAS  PubMed  Google Scholar 

  138. Demont, E. H. et al. 1,3-dimethyl benzimidazolones are potent, selective inhibitors of the BRPF1 bromodomain. ACS Med. Chem. Lett. 5, 1190–1195 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Bamborough, P. et al. GSK6853, a chemical probe for inhibition of the BRPF1 bromodomain. ACS Med. Chem. Lett. 7, 552–557 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Igoe, N. et al. Design of a biased potent small molecule inhibitor of the bromodomain and PHD finger-containing (BRPF) proteins suitable for cellular and in vivo studies. J. Med. Chem. 60, 668–680 (2017).

    CAS  PubMed  Google Scholar 

  141. Igoe, N. et al. Design of a chemical probe for the bromodomain and plant homeodomain finger-containing (BRPF) family of proteins. J. Med. Chem. 60, 6998–7011 (2017).

    CAS  PubMed  Google Scholar 

  142. Meier, J. C. et al. Selective targeting of bromodomains of the bromodomain-PHD fingers family impairs osteoclast differentiation. ACS Chem. Biol. 12, 2619–2630 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Palmer, W. S. et al. Structure-guided design of IACS-9571, a selective high-affinity dual TRIM24-BRPF1 bromodomain inhibitor. J. Med. Chem. 59, 1440–1454 (2016).

    CAS  PubMed  Google Scholar 

  144. Bennett, J. et al. Discovery of a chemical tool inhibitor targeting the bromodomains of TRIM24 and BRPF. J. Med. Chem. 59, 1642–1647 (2016).

    CAS  PubMed  Google Scholar 

  145. Hsia, E. Y., Zou, J. X. & Chen, H. W. The roles and action mechanisms of p160/SRC coactivators and the ANCCA coregulator in cancer. Prog. Mol. Biol. Transl Sci. 87, 261–298 (2009).

    CAS  PubMed  Google Scholar 

  146. Boussouar, F., Jamshidikia, M., Morozumi, Y., Rousseaux, S. & Khochbin, S. Malignant genome reprogramming by ATAD2. Biochim. Biophys. Acta 1829, 1010–1014 (2013).

    CAS  PubMed  Google Scholar 

  147. Cattaneo, M. et al. Lessons from yeast on emerging roles of the ATAD2 protein family in gene regulation and genome organization. Mol. Cells 37, 851–856 (2014).

    PubMed  PubMed Central  Google Scholar 

  148. Vidler, L. R., Brown, N., Knapp, S. & Hoelder, S. Druggability analysis and structural classification of bromodomain acetyl-lysine binding sites. J. Med. Chem. 55, 7346–7359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Demont, E. H. et al. Fragment-based discovery of low-micromolar ATAD2 bromodomain inhibitors. J. Med. Chem. 58, 5649–5673 (2015).

    CAS  PubMed  Google Scholar 

  150. Bamborough, P. et al. Structure-based optimization of naphthyridones into potent ATAD2 bromodomain inhibitors. J. Med. Chem. 58, 6151–6178 (2015).

    CAS  PubMed  Google Scholar 

  151. Bamborough, P. et al. A chemical probe for the ATAD2 bromodomain. Angew. Chem. Int. Ed. 55, 11382–11386 (2016).

    CAS  Google Scholar 

  152. Fernandez-Montalvan, A. E. et al. Isoform-selective ATAD2 chemical probe with novel chemical structure and unusual mode of action. ACS Chem. Biol. 12, 2730–2736 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Martin, M. P., Olesen, S. H., Georg, G. I. & Schonbrunn, E. Cyclin-dependent kinase inhibitor dinaciclib interacts with the acetyl-lysine recognition site of bromodomains. ACS Chem. Biol. 8, 2360–2365 (2013).

    CAS  PubMed  Google Scholar 

  154. Dittmann, A. et al. The commonly used PI3-kinase probe LY294002 is an inhibitor of BET bromodomains. ACS Chem. Biol. 9, 495–502 (2014).

    CAS  PubMed  Google Scholar 

  155. Arrowsmith, C. H. et al. The promise and peril of chemical probes. Nat. Chem. Biol. 11, 536–541 (2015). This must-read review for researchers uses chemical probes to explore new biology.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Mahadevan, D. et al. Phase I pharmacokinetic and pharmacodynamic study of the pan-PI3K/mTORC vascular targeted pro-drug SF1126 in patients with advanced solid tumours and B cell malignancies. Eur. J. Cancer 48, 3319–3327 (2012).

    CAS  PubMed  Google Scholar 

  157. Singh, A. R. et al. Single agent and synergistic activity of the “first-in-class” dual PI3K/BRD4 inhibitor SF1126 with sorafenib in hepatocellular carcinoma. Mol. Cancer Ther. 15, 2553–2562 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Ciceri, P. et al. Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat. Chem. Biol. 10, 305–312 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Ember, S. W. et al. Acetyl-lysine binding site of bromodomain-containing protein 4 (BRD4) interacts with diverse kinase inhibitors. ACS Chem. Biol. 9, 1160–1171 (2014). This study and that of Ciceri et al. (2014) reveal that many advanced preclinical (or even clinically used) kinase inhibitors have significant affinity for BDs, calling into question whether BD inhibition might contribute to the biological activities of these kinase inhibitors and suggesting the possibility of optimizing inhibitors for dual activity.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Allen, B. K. et al. Large-scale computational screening identifies first in class multitarget inhibitor of EGFR kinase and BRD4. Sci. Rep. 5, 16924 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Chen, L. et al. BRD4 structure-activity relationships of dual PLK1 kinase/BRD4 bromodomain inhibitor BI-2536. ACS Med. Chem. Lett. 6, 764–769 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Carlino, L. & Rastelli, G. Dual kinase-bromodomain inhibitors in anticancer drug discovery: a structural and pharmacological perspective. J. Med. Chem. 59, 9305–9320 (2016).

    CAS  PubMed  Google Scholar 

  163. Liu, S. et al. Structure-guided design and development of potent and selective dual bromodomain 4 (BRD4)/polo-like kinase 1 (PLK1) inhibitors. J. Med. Chem. 61, 7785–7795 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Peters, J. U. Polypharmacology - foe or friend? J. Med. Chem. 56, 8955–8971 (2013).

    CAS  PubMed  Google Scholar 

  165. Fiskus, W. et al. BET protein antagonist JQ1 is synergistically lethal with FLT3 tyrosine kinase inhibitor (TKI) and overcomes resistance to FLT3-TKI in AML cells expressing FLT-ITD. Mol. Cancer Ther. 13, 2315–2327 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Ember, S. W. et al. Potent dual BET bromodomain-kinase inhibitors as value-added multitargeted chemical probes and cancer therapeutics. Mol. Cancer Ther. 16, 1054–1067 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Bondeson, D. P. & Crews, C. M. Targeted protein degradation by small molecules. Annu. Rev. Pharmacol. Toxicol. 57, 107–123 (2017).

    CAS  PubMed  Google Scholar 

  168. Cromm, P. M. & Crews, C. M. Targeted protein degradation: from chemical biology to drug discovery. Cell Chem. Biol. 24, 1181–1190 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Lai, A. C. & Crews, C. M. Induced protein degradation: an emerging drug discovery paradigm. Nat. Rev. Drug Discov. 16, 101–114 (2017).

    CAS  PubMed  Google Scholar 

  170. Lucas, X. & Ciulli, A. Recognition of substrate degrons by E3 ubiquitin ligases and modulation by small-molecule mimicry strategies. Curr. Opin. Struct. Biol. 44, 101–110 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  172. Ottis, P. & Crews, C. M. Proteolysis-targeting chimeras: induced protein degradation as a therapeutic strategy. ACS Chem. Biol. 12, 892–898 (2017).

    CAS  PubMed  Google Scholar 

  173. Raina, K. & Crews, C. M. Targeted protein knockdown using small molecule degraders. Curr. Opin. Chem. Biol. 39, 46–53 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Zengerle, M., Chan, K. H. & Ciulli, A. Selective small molecule induced degradation of the BET bromodomain protein BRD4. ACS Chem. Biol. 10, 1770–1777 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Lu, J. et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem. Biol. 22, 755–763 (2015). This paper and those of Winter et al. (2015) and Zengerle et al. (2015) describe the first examples of PROTACs targeting BRD4.

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Robb, C. M. et al. Chemically induced degradation of CDK9 by a proteolysis targeting chimera (PROTAC). Chem. Commun. 53, 7577–7580 (2017).

    CAS  Google Scholar 

  178. Raina, K. et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc. Natl Acad. Sci. USA 113, 7124–7129 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Saenz, D. T. et al. Novel BET protein proteolysis-targeting chimera exerts superior lethal activity than bromodomain inhibitor (BETi) against post-myeloproliferative neoplasm secondary (s) AML cells. Leukemia 9, 1951–1961 (2017).

    Google Scholar 

  180. Sun, B. et al. BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells. Leukemia 32, 343–352 (2018).

    CAS  PubMed  Google Scholar 

  181. Ran, X. et al. Structure-based design of gamma-carboline analogues as potent and specific BET bromodomain inhibitors. J. Med. Chem. 58, 4927–4939 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Zhou, B. et al. Discovery of a small-molecule degrader of bromodomain and extra-terminal (BET) proteins with picomolar cellular potencies and capable of achieving tumor regression. J. Med. Chem. 61, 462–481 (2018).

    CAS  PubMed  Google Scholar 

  183. Bai, L. et al. Targeted degradation of BET proteins in triple-negative breast cancer. Cancer Res. 77, 2476–2487 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Qin, C. et al. Discovery of QCA570 as an exceptionally potent and efficacious proteolysis targeting chimera (PROTAC) degrader of the bromodomain and extra-terminal (BET) proteins capable of inducing complete and durable tumor regression. J. Med. Chem. 61, 6685–6704 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Wurz, R. P. et al. A “click chemistry platform” for the rapid synthesis of bispecific molecules for inducing protein degradation. J. Med. Chem. 61, 453–461 (2018).

    CAS  PubMed  Google Scholar 

  186. Lebraud, H., Wright, D. J., Johnson, C. N. & Heightman, T. D. Protein degradation by in-cell self-assembly of proteolysis targeting chimeras. ACS Cent. Sci. 2, 927–934 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Chan, K. H., Zengerle, M., Testa, A. & Ciulli, A. Impact of target warhead and linkage vector on inducing protein degradation: comparison of bromodomain and extra-terminal (BET) degraders derived from triazolodiazepine (JQ1) and tetrahydroquinoline (I-BET726) BET inhibitor scaffolds. J. Med. Chem. 61, 504–513 (2018).

    CAS  PubMed  Google Scholar 

  188. Gadd, M. S. et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat. Chem. Biol. 13, 514–521 (2017). This article describes the first published structure of a ternary complex of a PROTAC bound to its target protein and a recruited E3 ubiquitin ligase. This structure supports the idea that protein–protein interactions between E3 and target contribute to PROTAC degradation activity.

    CAS  PubMed  PubMed Central  Google Scholar 

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

  190. Roy, M. J. et al. SPR-measured dissociation kinetics of PROTAC ternary complexes influence target degradation rate. ACS Chem. Biol. 14, 361–368 (2019).

  191. Riching, K. M. et al. Quantitative live-cell kinetic degradation and mechanistic profiling of PROTAC mode of action. ACS Chem. Biol. 13, 2758–2770 (2018). This paper describes a panel of cell-based assays aimed at independent evaluation (and optimization) of different aspects of the PROTAC mechanism.

    CAS  PubMed  Google Scholar 

  192. 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  PubMed Central  Google Scholar 

  193. Mertz, J. A. et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc. Natl Acad. Sci. USA 108, 16669–16674 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Shin, H. Y. Targeting super-enhancers for disease treatment and diagnosis. Mol. Cells 41, 506–514 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Lin, X. et al. HEXIM1 as a robust pharmacodynamic marker for monitoring target engagement of BET family bromodomain inhibitors in tumors and surrogate tissues. Mol. Cancer Ther. 16, 388–396 (2017).

    CAS  PubMed  Google Scholar 

  196. Yeh, T. C. et al. Identification of CCR2 and CD180 as robust pharmacodynamic tumor and blood biomarkers for clinical use with BRD4/BET inhibitors. Clin. Cancer Res. 23, 1025–1035 (2017).

    CAS  PubMed  Google Scholar 

  197. Collins, T. A. et al. Translational modeling of drug-induced myelosuppression and effect of pretreatment myelosuppression for AZD5153, a selective BRD4 inhibitor. CPT Pharmacometrics Syst. Pharmacol. 6, 357–364 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Kurtz, S. E. et al. Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic malignancies. Proc. Natl Acad. Sci. USA 114, E7554–E7563 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Dawson, M. A. et al. Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia. Leukemia 28, 311–320 (2014). This report is one of the earliest of a BET-dependent transcriptional programme that can be targeted therapeutically. In this case, the transcriptional programme is driven by a recurrent mutation that could in principle be used as a selection biomarker in specific subtypes of AML.

    CAS  PubMed  Google Scholar 

  200. Bergsagel, P. L. & Kuehl, W. M. Chromosome translocations in multiple myeloma. Oncogene 20, 5611–5622 (2001).

    CAS  PubMed  Google Scholar 

  201. 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  PubMed Central  Google Scholar 

  202. Ceribelli, M. et al. Blockade of oncogenic IkappaB kinase activity in diffuse large B cell lymphoma by bromodomain and extraterminal domain protein inhibitors. Proc. Natl Acad. Sci. USA 111, 11365–11370 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Janouskova, H. et al. Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat. Med. 23, 1046–1054 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Zhang, P. et al. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT-mTORC1 activation. Nat. Med. 23, 1055–1062 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Dai, X. et al. Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat. Med. 23, 1063–1071 (2017). This article and that of Janouskova et al. (2017) and Zhang et al. (2017) provide evidence that supports the use of genetic features that lead to changes in BRD4 protein expression to identify a dependency that can be therapeutically targeted. Notably, SPOP is mutated in a significant number of prostate cancers, which suggests that it may be a relevant patient selection biomarker.

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Jin, X. et al. DUB3 promotes BET inhibitor resistance and cancer progression by deubiquitinating BRD4. Mol. Cell 71, 592–605 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Xiang, Q. et al. Discovery and optimization of 1-(1H-indol-1-yl)ethanone derivatives as CBP/EP300 bromodomain inhibitors for the treatment of castration-resistant prostate cancer. Eur. J. Med. Chem. 147, 238–252 (2018).

    CAS  PubMed  Google Scholar 

  208. Ogiwara, H. et al. Targeting p300 addiction in CBP-deficient cancers causes synthetic lethality by apoptotic cell death due to abrogation of MYC expression. Cancer Discov. 6, 430–445 (2016).

    CAS  PubMed  Google Scholar 

  209. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    CAS  PubMed  Google Scholar 

  210. Schmitt, M. W., Loeb, L. A. & Salk, J. J. The influence of subclonal resistance mutations on targeted cancer therapy. Nat. Rev. Clin. Oncol. 13, 335–347 (2016).

    CAS  PubMed  Google Scholar 

  211. Settleman, J. Cancer: bet on drug resistance. Nature 529, 289–290 (2016).

    CAS  PubMed  Google Scholar 

  212. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Rathert, P. et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature 525, 543–547 (2015). This article and that of Fong et al. (2015) highlight the powerful effects of transcriptional plasticity on promoting tumour resistance to natural and therapeutic antitumour mechanisms. Furthermore, the findings suggest that intervening at multiple nodes of a transcriptional network may be required for tumour elimination.

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Kumar, K. et al. GLI2-dependent c-MYC upregulation mediates resistance of pancreatic cancer cells to the BET bromodomain inhibitor JQ1. Sci. Rep. 5, 9489 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  216. Jang, J. E. et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin. Cancer Res. 23, 2781–2794 (2017).

    CAS  PubMed  Google Scholar 

  217. Lasorsa, E. et al. Mitochondrial protection impairs BET bromodomain inhibitor-mediated cell death and provides rationale for combination therapeutic strategies. Cell Death Dis. 6, e2014 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Conery, A. R. et al. Preclinical anticancer efficacy of BET bromodomain inhibitors is determined by the apoptotic response. Cancer Res. 76, 1313–1319 (2016). This article and that of Lasorsa et al. (2015) provide mechanistic support for ongoing trials testing the combination of BETis and BH3 mimetics.

    CAS  PubMed  Google Scholar 

  219. Zhao, Y. et al. High-resolution mapping of RNA polymerases identifies mechanisms of sensitivity and resistance to BET inhibitors in t(8;21) AML. Cell Rep. 16, 2003–2016 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Shu, S. et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 529, 413–417 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Herquel, B. et al. Transcription cofactors TRIM24, TRIM28, and TRIM33 associate to form regulatory complexes that suppress murine hepatocellular carcinoma. Proc. Natl Acad. Sci. USA 108, 8212–8217 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Shi, X. et al. Loss of TRIM33 causes resistance to BET bromodomain inhibitors through MYC- and TGF-beta-dependent mechanisms. Proc. Natl Acad. Sci. USA 113, E4558–E4566 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Xi, Q. et al. A poised chromatin platform for TGF-beta access to master regulators. Cell 147, 1511–1524 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Ma, Y. et al. The MAPK pathway regulates intrinsic resistance to BET inhibitors in colorectal cancer. Clin. Cancer Res. 23, 2027–2037 (2017).

    CAS  PubMed  Google Scholar 

  225. Kurimchak, A. M. et al. Resistance to BET bromodomain inhibitors is mediated by kinome reprogramming in ovarian cancer. Cell Rep. 16, 1273–1286 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Marcotte, R. et al. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell 164, 293–309 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Stuhlmiller, T. J. et al. Inhibition of lapatinib-induced kinome reprogramming in ERBB2-positive breast cancer by targeting BET family bromodomains. Cell Rep. 11, 390–404 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Stratikopoulos, E. E. et al. Kinase and BET inhibitors together clamp inhibition of PI3K signaling and overcome resistance to therapy. Cancer Cell 27, 837–851 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Asangani, I. A. et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510, 278–282 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  230. Asangani, I. A. et al. BET bromodomain inhibitors enhance efficacy and disrupt resistance to AR antagonists in the treatment of prostate cancer. Mol. Cancer Res. 14, 324–331 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Faivre, E. J. et al. Exploitation of castration-resistant prostate cancer transcription factor dependencies by the novel BET inhibitor ABBV-075. Mol. Cancer Res. 15, 35–44 (2017).

    CAS  PubMed  Google Scholar 

  232. Constellation Pharmaceuticals. Constellation Pharmaceuticals enhances and expands phase 2 MANIFEST study of CPI-0610 in myelofibrosis. Constellation Pharmaceuticals http://ir.constellationpharma.com/news-releases/news-release-details/constellation-pharmaceuticals-enhances-and-expands-phase-2 (2018).

  233. Schaffer, M. et al. Identification of potential ibrutinib combinations in hematological malignancies using a combination high-throughput screen. Leuk. Lymphoma 59, 931–940 (2017).

    PubMed  Google Scholar 

  234. Lam, L. T. et al. Vulnerability of small cell lung cancer to apoptosis induced by the combination of BET bromodomain proteins and BCL2 inhibitors. Mol. Cancer Ther. 16, 1511–1520 (2017).

    CAS  PubMed  Google Scholar 

  235. Peirs, S. et al. Targeting BET proteins improves the therapeutic efficacy of BCL-2 inhibition in T cell acute lymphoblastic leukemia. Leukemia 31, 2037–2047 (2017).

    CAS  PubMed  Google Scholar 

  236. Bui, M. H. et al. Preclinical characterization of BET family bromodomain inhibitor ABBV-075 suggests combination therapeutic strategies. Cancer Res. 77, 2976–2989 (2017).

    CAS  PubMed  Google Scholar 

  237. Wang, H. et al. JQ1 synergizes with the Bcl-2 inhibitor ABT-263 against MYCN-amplified small cell lung cancer. Oncotarget 8, 86312–86324 (2017).

    PubMed  PubMed Central  Google Scholar 

  238. Esteve-Arenys, A. et al. The BET bromodomain inhibitor CPI203 overcomes resistance to ABT-199 (venetoclax) by downregulation of BFL-1/A1 in in vitro and in vivo models of MYC+/BCL2+ double hit lymphoma. Oncogene 37, 1830–1844 (2018).

    CAS  PubMed  Google Scholar 

  239. Johnson-Farley, N., Veliz, J., Bhagavathi, S. & Bertino, J. R. ABT-199, a BH3 mimetic that specifically targets Bcl-2, enhances the antitumor activity of chemotherapy, bortezomib and JQ1 in “double hit” lymphoma cells. Leuk. Lymphoma 56, 2146–2152 (2015).

    CAS  PubMed  Google Scholar 

  240. Karakashev, S. et al. BET bromodomain inhibition synergizes with PARP inhibitor in epithelial ovarian cancer. Cell Rep. 21, 3398–3405 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Sun, C. et al. BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell 33, 401–416 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Wilson, A. J., Stubbs, M., Liu, P., Ruggeri, B. & Khabele, D. The BET inhibitor INCB054329 reduces homologous recombination efficiency and augments PARP inhibitor activity in ovarian cancer. Gynecol. Oncol. 149, 575–584 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Zenith Epigenetics. Zenith Epigenetics announces clinical trial collaboration with Pfizer. Zenith Epigenetics https://www.zenithepigenetics.com/newsroom/news-releases.html?article=28 (2018).

  244. Moros, A. et al. Synergistic antitumor activity of lenalidomide with the BET bromodomain inhibitor CPI203 in bortezomib-resistant mantle cell lymphoma. Leukemia 28, 2049–2059 (2014).

    CAS  PubMed  Google Scholar 

  245. Zhu, H. et al. BET bromodomain inhibition promotes anti-tumor immunity by suppressing PD-L1 expression. Cell Rep. 16, 2829–2837 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Hogg, S. J. et al. BET-bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1. Cell Rep. 18, 2162–2174 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Kagoya, Y. et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J. Clin. Invest. 126, 3479–3494 (2016). This article is the first to demonstrate the potential for ex vivo treatment of T cells with BETis to enhance antitumour immunity, which would mitigate any toxic effects or immune suppression that may arise from systemic delivery of BETis.

    PubMed  PubMed Central  Google Scholar 

  248. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Wai, D. C. C. et al. The BRD3 ET domain recognizes a short peptide motif through a mechanism that is conserved across chromatin remodelers and transcriptional regulators. J. Biol. Chem. 293, 7160–7175 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Lambert, J.-P. et al. Interactome rewiring following pharmacological targeting of BET bromodomains. Mol. Cell 73, 621–638 (2019).

  251. Tsai, W. W. et al. TRIM24 links a non-canonical histone signature to breast cancer. Nature 468, 927–932 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Thakur, J. K., Yadav, A. & Yadav, G. Molecular recognition by the KIX domain and its role in gene regulation. Nucleic Acids Res. 42, 2112–2125 (2014).

    CAS  PubMed  Google Scholar 

  253. Delvecchio, M., Gaucher, J., Aguilar-Gurrieri, C., Ortega, E. & Panne, D. Structure of the p300 catalytic core and implications for chromatin targeting and HAT regulation. Nat. Struct. Mol. Biol. 20, 1040–1046 (2013).

    CAS  PubMed  Google Scholar 

  254. Zhang, Y. et al. The ZZ domain of p300 mediates specificity of the adjacent HAT domain for histone H3. Nat. Struct. Mol. Biol. 25, 841–849 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Park, S. et al. Role of the CBP catalytic core in intramolecular SUMOylation and control of histone H3 acetylation. Proc. Natl Acad. Sci. USA 114, E5335–E5342 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  256. Huang, B., Yang, X. D., Zhou, M. M., Ozato, K. & Chen, L. F. Brd4 coactivates transcriptional activation of NF-kappaB via specific binding to acetylated RelA. Mol. Cell. Biol. 29, 1375–1387 (2009).

    CAS  PubMed  Google Scholar 

  257. Lamonica, J. M. et al. Bromodomain protein Brd3 associates with acetylated GATA1 to promote its chromatin occupancy at erythroid target genes. Proc. Natl Acad. Sci. USA 108, E159–E168 (2011).

    PubMed  PubMed Central  Google Scholar 

  258. Gamsjaeger, R. et al. Structural basis and specificity of acetylated transcription factor GATA1 recognition by BET family bromodomain protein Brd3. Mol. Cell. Biol. 31, 2632–2640 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Shi, J. et al. Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell 25, 210–225 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Liu, J. et al. Targeting the BRD4/FOXO3a/CDK6 axis sensitizes AKT inhibition in luminal breast cancer. Nat. Commun. 9, 5200 (2018). This article and those from Huang et al. (2009), Lamonica et al. (2011), Gamsjaeger et al. (2011) and Shi et al. (2014) demonstrate that BD function is not restricted to histone binding and that BDs can recruit transcription factors in functionally relevant ways.

    PubMed  PubMed Central  Google Scholar 

  261. Bassi, Z. I. et al. Modulating PCAF/GCN5 immune cell function through a PROTAC approach. ACS Chem. Biol. 13, 2862–2867 (2018).

    CAS  PubMed  Google Scholar 

  262. Gechijian, L. N. et al. Functional TRIM24 degrader via conjugation of ineffectual bromodomain and VHL ligands. Nat. Chem. Biol. 14, 405–412 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  263. Galdeano, C. et al. Structure-guided design and optimization of small molecules targeting the protein-protein interaction between the von Hippel-Lindau (VHL) E3 ubiquitin ligase and the hypoxia inducible factor (HIF) alpha subunit with in vitro nanomolar affinities. J. Med. Chem. 57, 8657–8663 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

A.R.C. thanks colleagues at Constellation Pharmaceuticals for input and discussions about the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed significantly to discussion of the content and reviewed and edited the article before submission. A.G.C. and A.R.C. researched data and wrote the article.

Corresponding authors

Correspondence to Andrea G. Cochran or Andrew R. Conery.

Ethics declarations

Competing interests

A.G.C. is an employee of Genentech, R.J.S. was, and A.R.C. is, an employee of Constellation Pharmaceuticals. Constellation Pharmaceuticals has progressed a bromodomain and extra-terminal (BET) inhibitor into clinical trials.

Additional information

Publisher’s note

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

Related links

RCSB Protein Data Bank: https://www.rcsb.org/

Glossary

Phenotypic screening

Chemical compounds can be screened for a desired biological effect or phenotype (usually in cell culture) rather than screening for binding to, or inhibition of, a purified protein.

Fragment-based ligand discovery

Fragments are low-molecular-weight chemical compounds that, although binding weakly to targets (with high micromolar to millimolar dissociation constants (Kd)), can be used to efficiently explore chemical space with a small compound collection.

BET bromodomains

Bromodomain and extra-terminal (BET) family proteins include BRD2, BRD3, BRD4 and bromodomain testis-specific protein (BRDT). Each BET-family protein has two bromodomains (BD1 and BD2). BD1 and BD2 are highly similar to one another (therefore many drugs bind to both BD1 and BD2), and each is nearly identical across the BET family. BET inhibitors therefore bind to all four BET-family proteins, with any selectivity limited to the differences between BD1 and BD2.

Chemotypes

Groups of small molecules related to one another by structure. Different chemotypes exhibiting the same expected mechanism of action (for example, bromodomain inhibition) are useful in establishing that cellular effects are likely caused by modulation of the intended target.

NUT midline carcinoma

A rare but aggressive squamous cell carcinoma that results from chromosomal rearrangements of the gene encoding nuclear protein in testis (NUT), frequently resulting in fusion with BET proteins (BRD4–NUT or BRD3–NUT).

Pleiotropic effects

A drug may modulate the intended target or may bind to an unrelated (and often unknown) target. Each of these can produce biological effects. One challenge in the use of chemical probes to learn about biology is separating on-target effects from any other effects.

Positive transcription elongation factor b

(P-TEFb). Also known as the CDK9–cyclinT heterodimer, P-TEFb phosphorylates and activates RNA polymerase II. The BRD4 carboxy-terminal domain binds P-TEFb at promoters and enhancer regions.

Proteolysis-targeting chimaera

(PROTAC). Bifunctional molecules that induce targeted protein degradation through co-opting the cellular ubiquitin machinery.

Nucleosome core particle

This basic unit of chromatin is a histone protein octamer (two copies each of histones H2A, H2B, H3 and H4) wrapped twice by ~147 bp of DNA.

NanoBRET assay

A cellular assay measuring the proximity of two proteins through bioluminescence resonance energy transfer (BRET), as developed by the Promega Corporation. For bromodomain (BD) research, the assay usually involves an expressed fusion of the luciferase variant nanoLuc to a histone protein. The BD protein is fused to the HaloTag protein. Cells are treated with a fluorescent ligand that covalently attaches to the expressed HaloTag protein. If the tagged BD binds to the histone, BRET occurs. Addition of a BD inhibitor reduces the BRET signal.

Fluorescence recovery after photobleaching (FRAP) assay

A cellular assay in which laser-induced photobleaching of a fluorescent protein localized in an area of a cell results in a dark spot that can be repopulated by dynamically relocalizing fluorescent proteins. The half-time (t1/2) for recovery of fluorescence is a measure of how strongly localized the bulk population is. For example, a bromodomain (BD) binding to chromatin is less dynamic than an inhibitor-bound BD that is not bound to chromatin. The inhibitor will result in a reduced recovery t1/2 in a FRAP assay.

Degron

Substrates degraded by the cellular ubiquitin system are often recognized through sequence motifs (degrons) that may or may not include post-translational modifications regulating degradation.

‘Click’ chemistry

‘Click’ reactions are crosslinking chemistries that are both selective and sufficiently mild to be useful in complex biological samples.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cochran, A.G., Conery, A.R. & Sims, R.J. Bromodomains: a new target class for drug development. Nat Rev Drug Discov 18, 609–628 (2019). https://doi.org/10.1038/s41573-019-0030-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-019-0030-7

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