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:

Therapeutic targeting of the p300/CBP bromodomain enhances the efficacy of immune checkpoint blockade therapy

Abstract

Blockade of immune checkpoints, such as programmed death-ligand 1 (PD-L1), has shown promise in cancer treatment; however, clinical response remains limited in many cancer types. Our previous research demonstrated that p300/CBP mediates the acetylation of the PD-L1 promoter, regulating PD-L1 expression. In this study, we further investigated the role of the p300/CBP bromodomain in regulating PD-L1 expression using CCS1477, a selective bromodomain inhibitor developed by our team. We found that the p300/CBP bromodomain is essential for H3K27 acetylation at PD-L1 enhancers. Inhibiting this modification significantly reduced enhancer activity and PD-L1 transcription, including exosomal PD-L1, which has been implicated as key contributors to resistance against PD-L1 blockade therapy in various cancers. Furthermore, CCS1477 treatment resulted in a marked reduction of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TME) by inhibiting key cytokines such as IL6, CSF1, and CSF2, which are crucial for MDSC differentiation and recruitment. By reducing PD-L1 expression and modulating the immunosuppressive TME, CCS1477 creates a more favorable environment for tumor-infiltrating lymphocytes, significantly enhancing the efficacy of immune checkpoint blockade (ICB) therapy. Notably, these effects were observed in both prostate cancer and melanoma models, underscoring the broad therapeutic potential of p300/CBP bromodomain inhibition in improving ICB outcomes.

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: Targeting p300/CBP bromodomain decreases PCa PD-L1 expression in vitro.
Fig. 2: CCS1477 decreases PCa PD-L1 expression in vivo.
Fig. 3: CCS1477 enhances PD-L1 blockade therapy in PCa.
Fig. 4: CCS1477 enhances the efficacy of ICB therapy in melanoma.
Fig. 5: CCS1477 treatment reduces MDSCs population in tumor bearing mice.
Fig. 6: CCS1477 down-regulated IL6/JAK pathway.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are either included in the manuscript or available from the corresponding authors upon reasonable request.

References

  1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48.

    Article  PubMed  Google Scholar 

  2. Swami U, McFarland TR, Nussenzveig R, Agarwal N. Advanced prostate cancer: treatment advances and future directions. Trends Cancer. 2020;6:702–15.

    Article  CAS  PubMed  Google Scholar 

  3. Chen Y, Zhou Q, Hankey W, Fang X, Yuan F. Second generation androgen receptor antagonists and challenges in prostate cancer treatment. Cell Death Dis. 2022;13:632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125:3384–91.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61.

    Article  CAS  PubMed  Google Scholar 

  6. Sharma P, Pachynski RK, Narayan V, Flechon A, Gravis G, Galsky MD, et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell. 2020;38:489–99.e483.

    Article  CAS  PubMed  Google Scholar 

  7. Liu J, He D, Cheng L, Huang C, Zhang Y, Rao X, et al. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene. 2020;39:3939–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lu X, Horner JW, Paul E, Shang X, Troncoso P, Deng P, et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature. 2017;543:728–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang J, Zhang HL, Sun X, et al. Exosomal PD-L1 and N-cadherin predict pulmonary metastasis progression for osteosarcoma patients. J Nanobiotechnol. 2020;18:151. https://doi.org/10.1186/s12951-020-00710-6.

    Article  CAS  Google Scholar 

  10. Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560:382–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177:414–27.e413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang G, Lu X, Dey P, Deng P, Wu CC, Jiang S, et al. Targeting YAP-dependent MDSC infiltration impairs tumor progression. Cancer Discov. 2016;6:80–95.

    Article  PubMed  Google Scholar 

  13. Zhao D, Cai L, Lu X, Liang X, Li J, Chen P, et al. Chromatin regulator CHD1 remodels the immunosuppressive tumor microenvironment in PTEN-deficient. Prostate Cancer Cancer Discov. 2020;10:1374–87.

    Article  CAS  PubMed  Google Scholar 

  14. Hossain DM, Pal SK, Moreira D, Duttagupta P, Zhang Q, Won H, et al. TLR9-Targeted STAT3 silencing abrogates immunosuppressive activity of myeloid-derived suppressor cells from prostate cancer patients. Clin Cancer Res. 2015;21:3771–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jin L, Garcia J, Chan E, de la Cruz C, Segal E, Merchant M, et al. Therapeutic targeting of the CBP/p300 bromodomain blocks the growth of castration-resistant prostate cancer. Cancer Res. 2017;77:5564–75.

    Article  CAS  PubMed  Google Scholar 

  16. Zhong J, Ding L, Bohrer LR, Pan Y, Liu P, Zhang J, et al. p300 acetyltransferase regulates androgen receptor degradation and PTEN-deficient prostate tumorigenesis. Cancer Res. 2014;74:1870–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lasko LM, Jakob CG, Edalji RP, Qiu W, Montgomery D, Digiammarino EL, et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature. 2017;550:128–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fu M, Wang C, Reutens AT, Wang J, Angeletti RH, Siconolfi-Baez L, et al. p300 and p300/cAMP-response element-binding protein-associated factor acetylate the androgen receptor at sites governing hormone-dependent transactivation. J Biol Chem. 2000;275:20853–60.

    Article  CAS  PubMed  Google Scholar 

  19. Welti J, Sharp A, Brooks N, Yuan W, McNair C, Chand SN, et al. Targeting the p300/CBP axis in lethal. Prostate Cancer Cancer Discov. 2021;11:1118–37.

    Article  CAS  PubMed  Google Scholar 

  20. Nicosia L, Spencer GJ, Brooks N, Amaral FMR, Basma NJ, Chadwick JA, et al. Therapeutic targeting of EP300/CBP by bromodomain inhibition in hematologic malignancies. Cancer Cell. 2023;41:2136–53.e2113.

    Article  CAS  PubMed  Google Scholar 

  21. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.

    Article  CAS  PubMed  Google Scholar 

  23. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu J, Zhao Y, He D, Jones KM, Tang S, Allison DB, et al. A kinome-wide CRISPR screen identifies CK1alpha as a target to overcome enzalutamide resistance of prostate cancer. Cell Rep Med. 2023;4:101015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J, Umansky V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol. 2024;21:147–64.

    Article  CAS  PubMed  Google Scholar 

  27. Weber R, Groth C, Lasser S, Arkhypov I, Petrova V, Altevogt P, et al. IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021;359:104254.

    Article  CAS  PubMed  Google Scholar 

  28. Neo SY, Tong L, Chong J, Liu Y, Jing X, Oliveira MMS, et al. Tumor-associated NK cells drive MDSC-mediated tumor immune tolerance through the IL-6/STAT3 axis. Sci Transl Med. 2024;16:eadi2952.

    Article  CAS  PubMed  Google Scholar 

  29. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kumar A, Taghi Khani A, Sanchez Ortiz A, Swaminathan S. GM-CSF: A Double-Edged Sword in Cancer Immunotherapy. Front Immunol. 2022;13:901277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cordonnier M, Nardin C, Chanteloup G, Derangere V, Algros MP, Arnould L, et al. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J Extracell Vesicles. 2020;9:1710899.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ebrahimi A, Sevinc K, Gurhan Sevinc G, Cribbs AP, Philpott M, Uyulur F, et al. Bromodomain inhibition of the coactivators CBP/EP300 facilitate cellular reprogramming. Nat Chem Biol. 2019;15:519–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Raisner R, Kharbanda S, Jin L, Jeng E, Chan E, Merchant M, et al. Enhancer activity requires CBP/P300 bromodomain-dependent histone H3K27 acetylation. Cell Rep. 2018;24:1722–9.

    Article  CAS  PubMed  Google Scholar 

  34. Lu J, Luo Y, Rao D, Wang T, Lei Z, Chen X, et al. Myeloid-derived suppressor cells in cancer: therapeutic targets to overcome tumor immune evasion. Exp Hematol Oncol. 2024;13:39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6:362.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185:2273–84.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants R01 CA256893 (XL), R01 CA264652 (XL), R01 CA157429 (XL), R01 CA272483 (XL). This research was supported by pilot funding (JL) provided by the Support from the University of Kentucky Markey Cancer Center’s Cancer Center Support Grant (P30 CA177558). The work was also supported by Biospecimen Procurement and Translational Pathology, Biostatistics and Bioinformatics, Flow Cytometry and Immune Monitoring Shared Resources of the University of Kentucky Markey Cancer Center (P30CA177558).

Author information

Authors and Affiliations

Authors

Contributions

XL designed and supervised the project. JL designed and performed the experiments, analyzed the data, and wrote the manuscript. XW conducted xenograft tumor harvesting and flow cytometry analysis. DH performed RNA-seq data analysis. HM and MN carried out the CRISPR knockout assays. MW and JP performed and evaluated immunofluorescence staining. XR, RW, SW, JW, and ZL contributed to manuscript editing. NB, NP, and KF developed and provided CCS1477.

Corresponding authors

Correspondence to Jinghui Liu or Xiaoqi Liu.

Ethics declarations

Competing interests

NB, NP, and KF are employees and shareholders of CellCentric Ltd. Additionally, Neil Pegg serves as a board director and is listed as the inventor on patents related to CCS1477.

Ethics approval and consent to participate

All procedures involving animals were carried out in compliance with institutional guidelines and approved by the Institutional Animal Care and Use Committee (IACUC) at the University of Kentucky (Protocol No.: 2020-3685). No human subjects were involved in this study.

Additional information

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

Supplementary information

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Wang, X., He, D. et al. Therapeutic targeting of the p300/CBP bromodomain enhances the efficacy of immune checkpoint blockade therapy. Oncogene 44, 2386–2395 (2025). https://doi.org/10.1038/s41388-025-03417-w

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41388-025-03417-w

This article is cited by

Search

Quick links