Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Rewiring cancer drivers to activate apoptosis

An Author Correction to this article was published on 18 August 2023

This article has been updated

Abstract

Genes that drive the proliferation, survival, invasion and metastasis of malignant cells have been identified for many human cancers1,2,3,4. Independent studies have identified cell death pathways that eliminate cells for the good of the organism5,6. The coexistence of cell death pathways with driver mutations suggests that the cancer driver could be rewired to activate cell death using chemical inducers of proximity (CIPs). Here we describe a new class of molecules called transcriptional/epigenetic CIPs (TCIPs) that recruit the endogenous cancer driver, or a downstream transcription factor, to the promoters of cell death genes, thereby activating their expression. We focused on diffuse large B cell lymphoma, in which the transcription factor B cell lymphoma 6 (BCL6) is deregulated7. BCL6 binds to the promoters of cell death genes and epigenetically suppresses their expression8. We produced TCIPs by covalently linking small molecules that bind BCL6 to those that bind to transcriptional activators that contribute to the oncogenic program, such as BRD4. The most potent molecule, TCIP1, increases binding of BRD4 by 50% over genomic BCL6-binding sites to produce transcriptional elongation at pro-apoptotic target genes within 15 min, while reducing binding of BRD4 over enhancers by only 10%, reflecting a gain-of-function mechanism. TCIP1 kills diffuse large B cell lymphoma cell lines, including chemotherapy-resistant, TP53-mutant lines, at EC50 of 1–10 nM in 72 h and exhibits cell-specific and tissue-specific effects, capturing the combinatorial specificity inherent to transcription. The TCIP concept also has therapeutic applications in regulating the expression of genes for regenerative medicine and developmental disorders.

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: Production of TCIPs.
Fig. 2: TCIP1 functions by inducing ternary complex formation.
Fig. 3: TCIP1 represses MYC and its targets while activating pro-apoptotic genes.
Fig. 4: Rapid activation of BCL6 target genes by recruitment of BRD4.
Fig. 5: Toxicity of TCIP1 in mice and primary human cells and generalization to ER-positive cancers.

Similar content being viewed by others

Data availability

Uncropped blots of western blots and Coomassie gels of recombinant proteins are available in Supplementary Fig. 1a,b, respectively. The flow gating strategy is available in Supplementary Fig. 2. Select gene expression changes in tissue from mice treated with TCIP1 are annotated in Supplementary Table 1. Source data for mouse drug levels in plasma and tissue and for body weight changes are provided. Sequencing data have been deposited to GSE211282Source data are provided with this paper.

Change history

References

  1. Weinberg, R. A. The action of oncogenes in the cytoplasm and nucleus. Science 230, 770–776 (1985).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sanchez-Vega, F. et al. Oncogenic signaling pathways in The Cancer Genome Atlas. Cell 173, 321–337.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Denny, S. K. et al. Nfib promotes metastasis through a widespread increase in chromatin accessibility. Cell 166, 328–342 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hengartner, M. O. & Horvitz, H. R. C. elegans cell survival gene ced-9 encodes a functional homolog of the mammalian proto-oncogene bcl-2. Cell 76, 665–676 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Strasser, A., O’Connor, L. & Dixit, V. M. Apoptosis signaling. Annu. Rev. Biochem. 69, 217–245 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Schmitz, R. et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N. Engl. J. Med. 378, 1396–1407 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Phan, R. T. & Dalla-Favera, R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature 432, 635–639 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  9. Spencer, D. M., Wandless, T. J., Schreiber, S. L. & Crabtree, G. R. Controlling signal transduction with synthetic ligands. Science 262, 1019–1024 (1993).

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Spencer, D. M., Graef, I., Austin, D. J., Schreiber, S. L. & Crabtree, G. R. A general strategy for producing conditional alleles of Src-like tyrosine kinases. Proc. Natl Acad. Sci. USA 92, 9805–9809 (1995).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Graef, I. A., Holsinger, L. J., Diver, S., Schreiber, S. L. & Crabtree, G. R. Proximity and orientation underlie signaling by the non-receptor tyrosine kinase ZAP70. EMBO J. 16, 5618–5628 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ho, S. N., Biggar, S. R., Spencer, D. M., Schreiber, S. L. & Crabtree, G. R. Dimeric ligands define a role for transcriptional activation domains in reinitiation. Nature 382, 822–826 (1996).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Erwin, G. S. et al. Synthetic transcription elongation factors license transcription across repressive chromatin. Science 358, 1617–1622 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hathaway, N. A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stanton, B. Z., Chory, E. J. & Crabtree, G. R. Chemically induced proximity in biology and medicine. Science https://doi.org/10.1126/science.aao5902 (2018).

  16. Stanton, B. Z. et al. Smarca4 ATPase mutations disrupt direct eviction of PRC1 from chromatin. Nat. Genet. 49, 282–288 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gestwicki, J. E., Crabtree, G. R. & Graef, I. A. Harnessing chaperones to generate small-molecule inhibitors of amyloid β aggregation. Science 306, 865–869 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Freiberg, R. A. et al. Specific triggering of the Fas signal transduction pathway in normal human keratinocytes. J. Biol. Chem. 271, 31666–31669 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. MacCorkle, R. A., Freeman, K. W. & Spencer, D. M. Synthetic activation of caspases: artificial death switches. Proc. Natl Acad. Sci. USA 95, 3655–3660 (1998).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yang, X., Chang, H. Y. & Baltimore, D. Essential role of CED-4 oligomerization in CED-3 activation and apoptosis. Science 281, 1355–1357 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  22. Basso, K. et al. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood 115, 975–984 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kerres, N. et al. Chemically induced degradation of the oncogenic transcription factor BCL6. Cell Rep. 20, 2860–2875 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nagel, S. et al. Amplification at 11q23 targets protein kinase SIK2 in diffuse large B-cell lymphoma. Leuk. Lymphoma 51, 881–891 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Dyer, M. J., Fischer, P., Nacheva, E., Labastide, W. & Karpas, A. A new human B-cell non-Hodgkin’s lymphoma cell line (Karpas 422) exhibiting both t (14;18) and t(4;11) chromosomal translocations. Blood 75, 709–714 (1990).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yu, C. et al. High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines. Nat. Biotechnol. 34, 419–423 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  32. Slabicki, M. et al. Small-molecule-induced polymerization triggers degradation of BCL6. Nature 588, 164–168 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schultz, L. W. & Clardy, J. Chemical inducers of dimerization: the atomic structure of FKBP12-FK1012A-FKBP12. Bioorg. Med. Chem. Lett. 8, 1–6 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Schreiber, S. L. The rise of molecular glues. Cell 184, 3–9 (2021).

    Article  CAS  PubMed  Google Scholar 

  36. Barish, G. D. et al. Bcl-6 and NF-κB cistromes mediate opposing regulation of the innate immune response. Genes Dev. 24, 2760–2765 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Perez-Rosado, A. et al. BCL6 represses NFκB activity in diffuse large B-cell lymphomas. J. Pathol. 214, 498–507 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Brunet, A. et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96, 857–868 (1999).

    Article  CAS  PubMed  Google Scholar 

  39. Hatzi, K. et al. A hybrid mechanism of action for BCL6 in B cells defined by formation of functionally distinct complexes at enhancers and promoters. Cell Rep. 4, 578–588 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Renault, V. M. et al. The pro-longevity gene FoxO3 is a direct target of the p53 tumor suppressor. Oncogene 30, 3207–3221 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zou, Z., Ohta, T., Miura, F. & Oki, S. ChIP-Atlas 2021 update: a data-mining suite for exploring epigenomic landscapes by fully integrating ChIP-seq, ATAC-seq and Bisulfite-seq data. Nucleic Acids Res. https://doi.org/10.1093/nar/gkac199 (2022).

  44. Hurtz, C. et al. Rationale for targeting BCL6 in MLL-rearranged acute lymphoblastic leukemia. Genes Dev. 33, 1265–1279 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Winter, G. E. et al. BET bromodomain proteins function as master transcription elongation factors independent of CDK9 recruitment. Mol. Cell 67, 5–18.e19 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Adelman, K. & Lis, J. T. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nat. Rev. Genet. 13, 720–731 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat. Genet. 40, 897–903 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  51. Huang, C., Hatzi, K. & Melnick, A. Lineage-specific functions of Bcl-6 in immunity and inflammation are mediated by distinct biochemical mechanisms. Nat. Immunol. 14, 380–388 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang, X., Li, Z., Naganuma, A. & Ye, B. H. Negative autoregulation of BCL-6 is bypassed by genetic alterations in diffuse large B cell lymphomas. Proc. Natl Acad. Sci. USA 99, 15018–15023 (2002).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pasqualucci, L. et al. Mutations of the BCL6 proto-oncogene disrupt its negative autoregulation in diffuse large B-cell lymphoma. Blood 101, 2914–2923 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Gearhart, M. D., Corcoran, C. M., Wamstad, J. A. & Bardwell, V. J. Polycomb group and SCF ubiquitin ligases are found in a novel BCOR complex that is recruited to BCL6 targets. Mol. Cell. Biol. 26, 6880–6889 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Huang, C. et al. The BCL6 RD2 domain governs commitment of activated B cells to form germinal centers. Cell Rep. 8, 1497–1508 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Uhlen, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  57. Davis, O. A. et al. Optimizing shape complementarity enables the discovery of potent tricyclic BCL6 inhibitors. J. Med. Chem. 65, 8169–8190 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bellenie, B. R. et al. Achieving in vivo target depletion through the discovery and optimization of benzimidazolone BCL6 degraders. J. Med. Chem. 63, 4047–4068 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ho, S. N., Boyer, S. H., Schreiber, S. L., Danishefsky, S. J. & Crabtree, G. R. Specific inhibition of formation of transcription complexes by a calicheamicin oligosaccharide: a paradigm for the development of transcriptional antagonists. Proc. Natl Acad. Sci. USA 91, 9203–9207 (1994).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  60. Corsello, S. M. et al. Discovering the anti-cancer potential of non-oncology drugs by systematic viability profiling. Nat. Cancer 1, 235–248 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. McCoull, W. et al. Development of a novel B-cell lymphoma 6 (BCL6) PROTAC to provide insight into small molecule targeting of BCL6. ACS Chem. Biol. 13, 3131–3141 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Stead, M. A. et al. Structure of the wild-type human BCL6 POZ domain. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 64, 1101–1104 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10 (2011).

    Article  Google Scholar 

  64. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  65. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Zhu, A., Ibrahim, J. G. & Love, M. I. Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences. Bioinformatics 35, 2084–2092 (2019).

    Article  CAS  PubMed  Google Scholar 

  67. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE Blacklist: identification of problematic regions of the genome. Sci. Rep. https://doi.org/10.1038/s41598-019-45839-z (2019).

  73. Bal, E. et al. Super-enhancer hypermutation alters oncogene expression in B cell lymphoma. Nature 607, 808–815 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The studies described in this article were funded from a grant from the HHMI to G.R.C. and NIH grants CA276167, CA163915 and MH126720-01 to G.R.C. Funding was also provided by a grant from the Mary Kay Foundation. Funding was provided to S.G. from the NIH grant 5F31HD103339-03. G.R.C., S.G., A.K., S.H.K., C.-Y.C. and J.M.S. were mentored and financially supported by Stanford’s SPARK Translational Research Program. G.R.C. was supported by the David Korn Professorship. This research was financially supported by Stanford Bio-X. Funding was provided to N.S.G. from departmental funds from Chemical and Systems Biology and the Stanford Cancer Institute, and the Gray laboratory also receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Jansen, Kinogen, Arbella, Deerfield, Springworks, Interline and Sanofi. Funding for pharmacokinetic studies was provided by NIH grant number 1 S10OD030332-01. M.R.G. is supported by a Leukemia and Lymphoma Society Scholar award. S.G. thanks T. Reindl, E. Bruguera and S. Hinshaw for helpful advice for the biochemical studies. We thank I. A. Graef for thoughtful comments on the manuscript, and members of the Crabtree and Gray laboratories for constructive comments.

Author information

Authors and Affiliations

Authors

Contributions

G.R.C. conceived the TCIP strategy to regulate endogenous genes and the rewiring of cancer drivers to activate cell death pathways. S.G. contributed many inventive ideas, designed the first effective TCIP and performed the biochemical, cell biological and genomic studies. A.K. contributed many inventive ideas, defined the conditions for TCIP use, carried out the first successful TCIP experiments and conducted the cell biological and genomic studies. C.-Y.C. suggested the use of BCL6 as a means of producing cell death. S.H.K. screened the cell death genes to detect those that would directly kill cancer cells. N.S.G. and S.G. designed the first effective TCIPs. X.L. synthesized the first effective TCIP. Z.L. and T.Z. contributed to TCIP design and optimization. W.J. synthesized TCIP1, the most potent TCIP to date. W.W. contributed innovative ideas to the computational analysis for the selection of TCIP components and helped fund the studies by writing grants with G.R.C. J.M.S. carried out experiments designed by G.R.C., S.G. and A.K. H.V. conducted the histopathology studies. H.Y. and M.R.G. contributed to TCIP1 application in DLBCL. The manuscript was written by G.R.C., S.G., A.K. and N.S.G. with input from all authors.

Corresponding authors

Correspondence to Nathanael S. Gray or Gerald R. Crabtree.

Ethics declarations

Competing interests

G.R.C. is a founder and scientific advisor for Foghorn Therapeutics and Shenandoah Therapeutics. N.S.G. is a founder, science advisory board member (SAB) and equity holder in Syros, C4, Allorion, Lighthorse, Voronoi, Inception, Matchpoint, CobroVentures, GSK, Shenandoah (board member), Larkspur (board member) and Soltego (board member). The Gray laboratory receives or has received research funding from Novartis, Takeda, Astellas, Taiho, Jansen, Kinogen, Arbella, Deerfield, Springworks, Interline and Sanofi. T.Z. is a scientific founder, equity holder and consultant of Matchpoint, equity holder of Shenandoah, and consultant of Lighthorse. M.R.G. reports research funding from Sanofi, Kite/Gilead, Abbvie and Allogene; consulting for Abbvie, Allogene and Bristol Myers Squibb; honoraria from Tessa Therapeutics, Monte Rosa Therapeutics and Daiichi Sankyo; and stock ownership of KDAc Therapeutics. Shenandoah has a license from Stanford for the TCIP technology that was invented by G.R.C., S.G., A.K., C-Y.C, W.W., S.H.K., N.S.G., W.J., X.L. and Z.L. All other authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Potency of TCIP1 in cancer cell lines and correlation with BCL6 level.

a. Comparison of TCIP1 effect on cell viability to effect of negative controls Neg1 and Neg2, or single-sided molecules JQ1 and BI3812, or the additive effect of JQ1+BI3812. mean±s.d., 72 h drug treatment. b. TCIP1 EC50 of cell viability is anti-correlated with BCL6 content across 14 different cancer cell lines, p-values computed by Students’ t-test, two-sided, not adjusted for multiple comparisons. For a,b: n = 4 biological replicates with 3 technical replicates each, mean±s.d 72 h drug treatment. c. Measurement of BCL6, BRD4, p53 and BCL2 status of DLBCL cell lines ranked from left to right from high to low-BCL6 protein content. d. Unbiased screen of the effect of TCIP1 on the viability of 906 barcoded cancer cell lines (PRISM). Drug was dosed for 120 h in triplicate (Methods).

Extended Data Fig. 2 Rescue of TCIP1-induced cell death by competitive titration of BCL6 inhibitors.

a. Rescue of TCIP1-induced cell death across cancer cell lines that are highly sensitive to TCIP1, b. moderately sensitive, or c. not at all sensitive. d. Comparison of JQ1, TCIP1, and Neg2, which contains a functional BRD4 inhibitor but very low-affinity BCL6 binder (KD ~ 10 µM) in e. cell lines that have low or no BCL6. For a,b,c, e: n = 3 biological replicates, mean±s.d. Viability curves in a, b, c, and e are after 72 h drug treatment.

Extended Data Fig. 3 Biochemical studies of ternary complex binding affinities of TCIPs.

a. Ternary complex formation by TCIPs with related chemistries. TCIP1 plotted on every graph as a comparison. Each point represents an independent replicate which is the mean of 3 technical repeats, mean value line drawn. b. Isothermal calorimetry experiments to measure binary affinities of TCIP1 to BRD4BD1, BCL6BTB, and associated controls. Representative data from 1-2 independent experiments shown. c. Representative biolayer interferometry measurements (BLI) of ternary complex kinetics from 3 independent replicates shown with biotinylated BCL6BTB on the tip and excess BRD4BD1 in the well with titration of TCIP1. d. Off-rate and e. half-life of TCIP1 calculated from BLI dissociation curve measurements, 7-8 different doses for each of n = 3 independent replicates, mean±s.d. f. Area under curve of TR-FRET correlates with potency of TCIPs on cell death (KARPAS422 cells, viability at 72 h). Representative cellular EC50s labeled, mean of 4 biological replicates. Each area under the curve point represents an independent replicate which is the mean of 3 technical repeats of the TR-FRET experiment.

Extended Data Fig. 4 TCIP1 induces apoptosis at every stage of the cell cycle.

a. Dose-dependent induction of apoptosis at 24 h by TCIP1 as measured by AnnexinV-positive cells. b. Kinetics of TCIP1-induced apoptosis in KARPAS422 cells. For a, b: n = 2-6 biological replicates, mean(±s.d) shown as appropriate. c. Design of assay to measure cell cycle progression simultaneously with apoptosis using Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. d. TCIP1 induction of cell cycle arrest compared to controls, representative of 2 biological replicates, see Extended Data Fig. 5 for both replicates’ flow cytometry graphs. e. 100 nM TCIP1 induction of apoptosis as measured by DNA fragmentation at each stage of the cell cycle, n = 2 biological replicates, mean shown. f. Measurement of cell viability after cell cycle arrest in G0/G1 by serum starvation in SUDHL5 cells and TCIP1 addition, n = 3 biological replicates, mean±s.d.

Extended Data Fig. 5 Cell-cycle block and apoptosis induction by TCIP1.

a. 100 nM TCIP1 addition at 24 h and simultaneous measurement of cell cycle block and apoptosis in KARPAS422 cells, two separate experiments on different passages of cells shown. Gates were set based on no-stain controls detailed in the Supplementary information.

Extended Data Fig. 6 Robust and dose-dependent gene regulation by TCIP1.

a. Principal component analysis of RNA-seq data after addition of TCIP1 for 20 h in 2 biological replicates of KARPAS422 cells. b. Gene expression changes after addition of 100 nM TCIP1 for 20 h in KARPAS422 cells. Adjusted p-values computed by two-sided Wald test and adjusted for multiple comparisons by Benjamini-Hochberg. Significance cutoffs were padj ≤ 0.05 and |log2(Drug/DMSO)| ≥ 1), n = 2 biological replicates. c. Dose-dependent change in gene expression. d. Enrichment analysis of upregulated genes (MSigDB Hallmark Pathways). e. Analysis of TF binding at the top upregulated genes in over 4,500 public transcription factor ChIP-seq datasets from blood-lineage cells. For d, e: adjusted p-values computed by two-sided Fisher’s exact test and adjusted for multiple comparisons by Benjamini-Hochberg.

Extended Data Fig. 7 Specific activation of gene expression by TCIP1 but not related controls.

a. Gene expression changes after 1 h or 4 h addition of 10 nM TCIP1 in KARPAS422 cells. Changes at 2 h was shown in Fig. 4a. Adjusted p-values computed by two-sided Wald test and adjusted for multiple comparisons by Benjamini-Hochberg. Significance cutoffs were padj ≤ 0.05 and |log2(Drug/DMSO)| ≥ 0.5), n = 3 biological replicates. b. Specific effects of TCIP1 across transcriptome. For Neg1 and Neg2, n = 2 biological replicates. For TCIP1, n = 3 biological replicates. c. Enrichment analysis of upregulated and downregulated genes (MSigDB Hallmark Pathways). Adjusted p-values computed by two-sided Fisher’s exact test and adjusted for multiple comparisons by Benjamini-Hochberg.

Extended Data Fig. 8 ChIP-seq analyses of BRD4, H3K27ac, and RNA Pol II in response to TCIP1.

a. PCA plots of each ChIP-seq experiment at indicated timepoints of 10 nM TCIP1 addition: 0hr (DMSO), 15 min, 1 h, 2 h, and 4 h. b. Browser tracks of Pol II Ser2 phos, Pol II Ser5 phos, H3K27ac, and BRD4 at BCL6-target genes and TCIP1-upregulated genes FOXO3 and BCL2L11/BIM. c. Volcano plots of Pol II ser 2 phos, Pol II ser 5 phos, and H3K27ac after 2 h 10 nM TCIP1 addition. Adjusted p-values computed by two-sided Wald test and adjusted for multiple comparisons by Benjamini-Hochberg. Peaks were classified as differential after reads in peaks-based regulative log expression (RLE) normalization and cutoffs padj ≤ 0.05 and |log2(Drug/DMSO)|≥0.5. d. Enhancer and super-enhancer classification in KARPAS422 cells based on H3K27ac ChIP-seq and the ROSE algorithm (Methods). e. BRD4 and H3K27ac ChIP-seq track at the known OCA-B super-enhancer after TCIP1 addition for indicated timepoints. In b, e, Pol II Ser2 phos, Pol II Ser5 phos, and H3K27ac tracks in are spike-in- and input-normalized, BRD4 tracks are sequence-depth- and input-normalized.

Extended Data Fig. 9 Conversion of BCL6 auto-inhibitory pathway to feedforward loop.

a. Control Neg1 and Neg2 effect on BCL6 protein levels at 20 h treatment in KARPAS422 cells. b. Effect on BRD4 levels at 20 h treatment with TCIP1 in KARPAS422 cells. c. Kinetics of BCL6 upregulation in two separate DLBCL cell lines, KARPAS422 and SUDHL5, after addition of 10 nM TCIP1. Blots in a–c representative of 2 biological replicates (for SUDHL5) or 3 (for KARPAS422). d. Model for conversion of BCL6 auto-inhibitory circuit to a positive feedback loop.

Supplementary information

Supplementary Information

This file contains Supplementary Notes regarding the chemical synthesis and protein constructs and purification, Supplementary Figs 1–2, and a full description for Supplementary Table 1 (table supplied separately)

Reporting Summary

Supplementary Table 1

Mouse Tissue RNA-seq

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gourisankar, S., Krokhotin, A., Ji, W. et al. Rewiring cancer drivers to activate apoptosis. Nature 620, 417–425 (2023). https://doi.org/10.1038/s41586-023-06348-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06348-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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