B7-H1 Antigen; Immune Checkpoint Inhibitors; Nuclear Proteins; Transcription Factors; Paclitaxel; Humans; Animals; Mice; B7-H1 Antigen/metabolism; Immune Checkpoint Inhibitors/therapeutic use; Paclitaxel/pharmacology; Paclitaxel/therapeutic use; Immunotherapy/methods; Triple Negative Breast Neoplasms/pathology; Immunotherapy; Triple Negative Breast Neoplasms; Oncology; Cancer Research
Abstract :
[en] Immune checkpoint inhibition combined with chemotherapy is currently approved as first-line treatment for patients with advanced PD-L1-positive triple-negative breast cancer (TNBC). However, a significant proportion of metastatic TNBC is PD-L1-negative and, in this population, chemotherapy alone largely remains the standard-of-care and novel therapeutic strategies are needed to improve clinical outcomes. Here, we describe a triple combination of anti-PD-L1 immune checkpoint blockade, epigenetic modulation thorough bromodomain and extra-terminal (BET) bromodomain inhibition (BBDI), and chemotherapy with paclitaxel that effectively inhibits both primary and metastatic tumor growth in two different syngeneic murine models of TNBC. Detailed cellular and molecular profiling of tumors from single and combination treatment arms revealed increased T- and B-cell infiltration and macrophage reprogramming from MHCIIlow to a MHCIIhigh phenotype in mice treated with triple combination. Triple combination also had a major impact on gene expression and chromatin profiles shifting cells to a more immunogenic and senescent state. Our results provide strong preclinical evidence to justify clinical testing of BBDI, paclitaxel, and immune checkpoint blockade combination.
Disciplines :
Oncology
Author, co-author :
Alečković, Maša ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Li, Zheqi ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Zhou, Ningxuan ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Qiu, Xintao ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Lulseged, Bethlehem ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
Foidart, Pierre ; Centre Hospitalier Universitaire de Liège - CHU > > Service d'oncologie médicale ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Huang, Xiao-Yun ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
Garza, Kodie ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
Shu, Shaokun ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Kesten, Nikolas ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Li, Rong ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Lim, Klothilda ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Garrido-Castro, Ana C ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Guerriero, Jennifer L ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Surgery, Division of Breast Surgery, Brigham and Women's Hospital, Boston, Massachusetts
Qi, Jun ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
Long, Henry W ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Harvard University, Cambridge, Massachusetts
Polyak, Kornelia ; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts ; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts ; Department of Medicine, Harvard Medical School, Boston, Massachusetts
NCI - National Cancer Institute Susan G. Komen Breast Cancer Foundation
Funding text :
We thank members of the Polyak laboratory for their critical reading of the article and useful advice. Financial support: Funded by the NCI R35 CA197623 (K. Polyak), P50 CA168504 (J.L. Guerriero, K. Polyak), P01CA250959 (K. Polyak, H.W. Long), the Ludwig Center at Harvard (K. Polyak), and the Susan G. Komen Foundation PDF14302777 (S. Shu).
scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.
Bibliography
Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 2020;396:1817–28.
Kagihara JA, Andress M, Diamond JR. Nab-paclitaxel and atezolizumab for the treatment of PD-L1-positive, metastatic triple-negative breast cancer: review and future directions. Expert Rev Precis Med Drug Dev 2020;5:59–65.
Miles D, Gligorov J, Andre F, Cameron D, Schneeweiss A, Barrios C, et al. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol 2021;32:994–1004.
Shu S, Lin CY, He HH, Witwicki RM, Tabassum DP, Roberts JM, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 2016;529:413–7.
Shu S, Wu HJ, Ge JY, Zeid R, Harris IS, Jovanovic B, et al. Synthetic lethal and resistance interactions with BET bromodomain inhibitors in triple-negative breast cancer. Mol Cell 2020;78:1096–113.
Zhu H, Bengsch F, Svoronos N, Rutkowski MR, Bitler BG, Allegrezza MJ, et al. BET bromodomain inhibition promotes anti-tumor immunity by suppressing PD-L1 expression. Cell Rep 2016;16:2829–37.
Hogg SJ, Vervoort SJ, Deswal S, Ott CJ, Li J, Cluse LA, et al. BET-Bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1. Cell Rep 2017;18:2162–74.
Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, et al. Selective inhibition of BET bromodomains. Nature 2010;468:1067–73.
Zhao Y, Yang CY, Wang S. The making of I-BET762, a BET bromodomain inhibitor now in clinical development. J Med Chem 2013;56:7498–500.
Huh SJ, Clement K, Jee D, Merlini A, Choudhury S, Maruyama R, et al. Age- and pregnancy-associated DNA methylation changes in mammary epithelial cells. Stem Cell Rep 2015;4:297–311.
Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: open source software for digital pathology image analysis. Sci Rep 2017;7:16878.
Gil Del Alcazar CR, Trinh A, Aleckovic M, Rojas Jimenez E, Harper NW, Oliphant MU, et al. Insights into immune escape during tumor evolution and response to immunotherapy using a rat model of breast cancer. Cancer Immunol Res 2022;10:680–97.
Ghoshdastider U, Rohatgi N, Mojtabavi Naeini M, Baruah P, Revkov E, Guo YA, et al. Pan-cancer analysis of ligand-receptor cross-talk in the tumor microenvironment. Cancer Res 2021;81:1802–12.
Corces MR, Trevino AE, Hamilton EG, Greenside PG, Sinnott-Armstrong NA, Vesuna S, et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat Methods 2017;14:959–62.
Taing L, Cousins C, Bai G, Paloma C, Qiu X, Brown M, et al. CHIPS: a snakemake pipeline for quality control and reproducible processing of chromatin profiling data. F1000Research 2021;10:517.
Qiu X, Feit AS, Feiglin A, Xie Y, Kesten N, Taing L, et al. CoBRA: containerized bioinformatics workflow for reproducible ChIP/ATAC-seq analysis. Genomics Proteomics Bioinformatics 2021;19:652–61.
Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009;25:1754–60.
Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol 2008;9:R137.
Neph S, Kuehn MS, Reynolds AP, Haugen E, Thurman RE, Johnson AK, et al. BEDOPS: high-performance genomic feature operations. Bioinformatics 2012; 28:1919–20.
Ge JY, Shu S, Kwon M, Jovanovic B, Murphy K, Gulvady A, et al. Acquired resistance to combined BET and CDK4/6 inhibition in triple-negative breast cancer. Nat Commun 2020;11:2350.
Tooley KA, Escobar G, Anderson AC. Spatial determinants of CD8(þ) T cell differentiation in cancer. Trends Cancer 2022;8:642–54.
Qin Q, Fan J, Zheng R, Wan C, Mei S, Wu Q, et al. Lisa: inferring transcriptional regulators through integrative modeling of public chromatin accessibility and ChIP-seq data. Genome Biol 2020;21:32.
Hu Q, Hong Y, Qi P, Lu G, Mai X, Xu S, et al. Atlas of breast cancer infiltrated B-lymphocytes revealed by paired single-cell RNA-sequencing and antigen receptor profiling. Nat Commun 2021;12:2186.
Dobenecker MW, Park JS, Marcello J, McCabe MT, Gregory R, Knight SD, et al. Signaling function of PRC2 is essential for TCR-driven T cell responses. J Exp Med 2018;215:1101–13.
Milazzo G, Perini G, Giorgi FM. Single-cell sequencing identifies master regulators affected by panobinostat in neuroblastoma cells. Genes 2022;13: 2240.
Darling TK, Lamb TJ. Emerging roles for Eph receptors and ephrin ligands in immunity. Front Immunol 2019;10:1473.
Song L, Cohen D, Ouyang Z, Cao Y, Hu X, Liu XS. TRUST4: immune repertoire reconstruction from bulk and single-cell RNA-seq data. Nat Methods 2021;18: 627–30.
Mungrue IN, Pagnon J, Kohannim O, Gargalovic PS, Lusis AJ. CHAC1/MGC4504 is a novel proapoptotic component of the unfolded protein response, downstream of the ATF4-ATF3-CHOP cascade. J Immunol 2009;182:466–76.
Budanov AV, Sablina AA, Feinstein E, Koonin EV, Chumakov PM. Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of bacterial AhpD. Science 2004;304:596–600.
Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, et al. Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat Protoc 2013;8: 2502–15.
Debien V, De Caluwe A, Wang X, Piccart-Gebhart M, Tuohy VK, Romano E, et al. Immunotherapy in breast cancer: an overview of current strategies and perspectives. NPJ Breast Cancer 2023;9:7.
Jacobs F, Agostinetto E, Miggiano C, De Sanctis R, Zambelli A, Santoro A. Hope and hype around immunotherapy in triple-negative breast cancer. Cancers 2023; 15:2933.
Halder TG, Soldi R, Sharma S. Bromodomain and extraterminal domain protein bromodomain inhibitor based cancer therapeutics. Curr Opin Oncol 2021;33: 526–31.
Mao W, Ghasemzadeh A, Freeman ZT, Obradovic A, Chaimowitz MG, Nirschl TR, et al. Immunogenicity of prostate cancer is augmented by BET bromodomain inhibition. J Immunother Cancer 2019;7:277.
Adeegbe DO, Liu S, Hattersley MM, Bowden M, Zhou CW, Li S, et al. BET bromodomain inhibition cooperates with PD-1 blockade to facilitate antitumor response in Kras-mutant non-small cell lung cancer. Cancer Immunol Res 2018; 6:1234–45.
Wang H, Liu G, Jin X, Song S, Chen S, Zhou P, et al. BET inhibitor JQ1 enhances anti-tumor immunity and synergizes with PD-1 blockade in CRC. J Cancer 2022; 13:2126–37.
Sauvage D, Bosseler M, Viry E, Kanli G, Oudin A, Berchem G, et al. The BET protein inhibitor JQ1 decreases hypoxia and improves the therapeutic benefit of anti-PD-1 in a high-risk neuroblastoma mouse model. Cells 2022; 11:2783.
Liu C, Miao X, Wang Y, Wen L, Cheng X, Kong D, et al. Bromo- and extraterminal domain protein inhibition improves immunotherapy efficacy in hepatocellular carcinoma. Cancer Sci 2020;111:3503–15.
Griss J, Bauer W, Wagner C, Simon M, Chen M, Grabmeier-Pfistershammer K, et al. B cells sustain inflammation and predict response to immune checkpoint blockade in human melanoma. Nat Commun 2019;10:4186.
Petitprez F, de Reynies A, Keung EZ, Chen TW, Sun CM, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020;577:556–60.
Kim SS, Sumner WA, Miyauchi S, Cohen EEW, Califano JA, Sharabi AB. Role of B cells in responses to checkpoint blockade immunotherapy and overall survival of cancer patients. Clin Cancer Res 2021;27:6075–82.
Chen HA, Ho YJ, Mezzadra R, Adrover JM, Smolkin R, Zhu C, et al. Senescence rewires microenvironment sensing to facilitate antitumor immunity. Cancer Discov 2023;13:432–53.
Marin I, Boix O, Garcia-Garijo A, Sirois I, Caballe A, Zarzuela E, et al. Cellular senescence is immunogenic and promotes antitumor immunity. Cancer Discov 2023;13:410–31.
Acosta JC, O’Loghlen A, Banito A, Raguz S, Gil J. Control of senescence by CXCR2 and its ligands. Cell Cycle 2008;7:2956–9.
Mabrouk N, Tran T, Sam I, Pourmir I, Gruel N, Granier C, et al. CXCR6 expressing T cells: functions and role in the control of tumors. Front Immunol 2022;13:1022136.
Cousin S, Blay JY, Garcia IB, de Bono JS, Le Tourneau C, Moreno V, et al. Safety, pharmacokinetic, pharmacodynamic and clinical activity of molibresib for the treatment of nuclear protein in testis carcinoma and other cancers: results of a phase I/II open-label, dose escalation study. Int J Cancer 2022; 150:993–1006.
Shapiro GI, LoRusso P, Dowlati A, TD K, Jacobson CA, Vaishampayan U, et al. A phase 1 study of RO6870810, a novel bromodomain and extra-terminal protein inhibitor, in patients with NUT carcinoma, other solid tumours, or diffuse large B-cell lymphoma. Br J Cancer 2021;124:744–53.
Piha-Paul SA, Hann CL, French CA, Cousin S, Brana I, Cassier PA, et al. Phase 1 study of molibresib (GSK525762), a bromodomain and extra-terminal domain protein inhibitor, in NUT carcinoma and other solid tumors. JNCI Cancer Spectr 2020;4:pkz093.
Cheng ML, Huang Y, Luong N, LoPiccolo J, Nishino M, Sholl LM, et al. Exceptional response to bromodomain and extraterminal domain inhibitor therapy with BMS-986158 in BRD4-NUTM1 NUT carcinoma harboring a BRD4 splice site mutation. JCO Precis Oncol 2023;7:e2200633.
Aggarwal RR, Schweizer MT, Nanus DM, Pantuck AJ, Heath EI, Campeau E, et al. A phase Ib/IIa study of the Pan-BET inhibitor ZEN-3694 in combination with enzalutamide in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res 2020;26:5338–47.
Aftimos PG, Oliveira M, Punie K, Boni V, Hamilton EP, Gucalp A, et al. A phase 1b/2 study of the BET inhibitor ZEN-3694 in combination with talazoparib for treatment of patients with TNBC without gBRCA1/2 mutations. J Clin Oncol 40: 16s, 2022 (suppl; abstr 1023).
Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer 2021;124:1478–90.
This website uses cookies to improve user experience. Read more
Save & Close
Accept all
Decline all
Show detailsHide details
Cookie declaration
About cookies
Strictly necessary
Performance
Strictly necessary cookies allow core website functionality such as user login and account management. The website cannot be used properly without strictly necessary cookies.
This cookie is used by Cookie-Script.com service to remember visitor cookie consent preferences. It is necessary for Cookie-Script.com cookie banner to work properly.
Performance cookies are used to see how visitors use the website, eg. analytics cookies. Those cookies cannot be used to directly identify a certain visitor.
Used to store the attribution information, the referrer initially used to visit the website
Cookies are small text files that are placed on your computer by websites that you visit. Websites use cookies to help users navigate efficiently and perform certain functions. Cookies that are required for the website to operate properly are allowed to be set without your permission. All other cookies need to be approved before they can be set in the browser.
You can change your consent to cookie usage at any time on our Privacy Policy page.