Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Breast Cancer | Research

Activity of BET-proteolysis targeting chimeric (PROTAC) compounds in triple negative breast cancer

Authors: María del Mar Noblejas-López, Cristina Nieto-Jimenez, Miguel Burgos, Mónica Gómez-Juárez, Juan Carlos Montero, Azucena Esparís-Ogando, Atanasio Pandiella, Eva M. Galán-Moya, Alberto Ocaña

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Triple negative breast cancer (TNBC) is an incurable disease where novel therapeutic strategies are needed. Proteolysis targeting chimeric (PROTAC) are novel compounds that promote protein degradation by binding to an ubiquitin ligase. In this work, we explored the antitumoral activity of two novel BET-PROTACs, MZ1 and ARV-825, in TNBC, ovarian cancer and in a BET inhibitor resistant model.

Methods

OVCAR3, SKOV3, BT549, MDA-MB-231 cell lines and the JQ1 resistant cell line MDA-MB-231R were evaluated. MTTs, colony-forming assay, three-dimensional cultures in matrigel, flow cytometry, and western blots were performed to explore the anti-proliferative effect and biochemical mechanism of action of MZ1 and ARV-825. In vivo studies included BALB/c nu/nu mice engrafted with MDA-MB-231R cells.

Results

The BET-PROTACs MZ1 and ARV-825 efficiently downregulated the protein expression levels of the BET protein BRD4, in MDA-MB-231 and MDA-MB-231R. MZ1 and ARV-825 also showed an antiproliferative effect on sensitive and resistant cells. This effect was corroborated in other triple negative (BT549) and ovarian cancer (SKOV3, OVCAR3) cell lines. MZ1 provoked G2/M arrest in MDA-MB-231. In addition, a profound effect on caspase-dependent apoptosis was observed in both sensitive and resistant cells. No synergistic activity was observed when it was combined with docetaxel, cisplatin or olaparib. Finally, in vivo administration of MZ1 rescued tumor growth in a JQ1-resistant xenograft model, reducing the expression levels of BRD4.

Conclusions

Using both in vitro and in vivo approaches, we describe the profound activity of BET-PROTACs in parental and BETi-resistant TNBC models. This data provides options for further clinical development of these agents in TNBC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ocana A, Pandiella A. Targeting oncogenic vulnerabilities in triple negative breast cancer: biological bases and ongoing clinical studies. Oncotarget. 2017;8(13):22218–34.CrossRef Ocana A, Pandiella A. Targeting oncogenic vulnerabilities in triple negative breast cancer: biological bases and ongoing clinical studies. Oncotarget. 2017;8(13):22218–34.CrossRef
2.
go back to reference Ocana A, Pandiella A. Identifying breast cancer druggable oncogenic alterations: lessons learned and future targeted options. Clin Cancer Res. 2008;14:961–70.CrossRef Ocana A, Pandiella A. Identifying breast cancer druggable oncogenic alterations: lessons learned and future targeted options. Clin Cancer Res. 2008;14:961–70.CrossRef
3.
go back to reference Filippakopoulos P, Qi J, Picaud S, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.CrossRef Filippakopoulos P, Qi J, Picaud S, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–73.CrossRef
4.
go back to reference Ocana A, Nieto-Jimenez C, Pandiella A. BET inhibitors as novel therapeutic agents in breast cancer. Oncotarget. 2017;8:71285–91.PubMedPubMedCentral Ocana A, Nieto-Jimenez C, Pandiella A. BET inhibitors as novel therapeutic agents in breast cancer. Oncotarget. 2017;8:71285–91.PubMedPubMedCentral
5.
go back to reference Perez-Pena J, Serrano-Heras G, Montero JC, et al. In silico analysis guides selection of BET inhibitors for triple-negative breast Cancer treatment. Mol Cancer Ther. 2016;15:1823–33.CrossRef Perez-Pena J, Serrano-Heras G, Montero JC, et al. In silico analysis guides selection of BET inhibitors for triple-negative breast Cancer treatment. Mol Cancer Ther. 2016;15:1823–33.CrossRef
6.
go back to reference Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54:728–36.CrossRef Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54:728–36.CrossRef
7.
go back to reference Shu S, Lin CY, He HH, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–7.CrossRef Shu S, Lin CY, He HH, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–7.CrossRef
8.
go back to reference Stratikopoulos EE, Dendy M, Szabolcs M, et al. Kinase and BET inhibitors together clamp inhibition of PI3K signaling and overcome resistance to therapy. Cancer Cell. 2015;27:837–51.CrossRef Stratikopoulos EE, Dendy M, Szabolcs M, et al. Kinase and BET inhibitors together clamp inhibition of PI3K signaling and overcome resistance to therapy. Cancer Cell. 2015;27:837–51.CrossRef
9.
go back to reference Nieto-Jimenez C, Alcaraz-Sanabria A, Perez-Pena J, et al. Targeting basal-like breast tumors with bromodomain and extraterminal domain (BET) and polo-like kinase inhibitors. Oncotarget. 2017;8(12):19478–90.CrossRef Nieto-Jimenez C, Alcaraz-Sanabria A, Perez-Pena J, et al. Targeting basal-like breast tumors with bromodomain and extraterminal domain (BET) and polo-like kinase inhibitors. Oncotarget. 2017;8(12):19478–90.CrossRef
10.
go back to reference Ma Y, Wang L, Neitzel LR, et al. The MAPK pathway regulates intrinsic resistance to BET inhibitors in colorectal Cancer. Clin Cancer Res. 2017;23:2027–37.CrossRef Ma Y, Wang L, Neitzel LR, et al. The MAPK pathway regulates intrinsic resistance to BET inhibitors in colorectal Cancer. Clin Cancer Res. 2017;23:2027–37.CrossRef
11.
go back to reference Jang JE, Eom JI, Jeung HK, et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin Cancer Res. 2017;23(11):2781–94.CrossRef Jang JE, Eom JI, Jeung HK, et al. AMPK-ULK1-mediated autophagy confers resistance to BET inhibitor JQ1 in acute myeloid leukemia stem cells. Clin Cancer Res. 2017;23(11):2781–94.CrossRef
12.
go back to reference Fong CY, Gilan O, Lam EY, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–42.CrossRef Fong CY, Gilan O, Lam EY, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–42.CrossRef
13.
go back to reference Wang P, Zhou J. Proteolysis targeting chimera (PROTAC): a paradigm-shifting approach in small molecule drug discovery. Curr Top Med Chem. 2018;18:1354–6.CrossRef Wang P, Zhou J. Proteolysis targeting chimera (PROTAC): a paradigm-shifting approach in small molecule drug discovery. Curr Top Med Chem. 2018;18:1354–6.CrossRef
14.
go back to reference Zengerle M, Chan KH, Ciulli A. Selective small molecule induced degradation of the BET Bromodomain protein BRD4. ACS Chem Biol. 2015;10:1770–7.CrossRef Zengerle M, Chan KH, Ciulli A. Selective small molecule induced degradation of the BET Bromodomain protein BRD4. ACS Chem Biol. 2015;10:1770–7.CrossRef
15.
go back to reference Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase Cereblon to efficiently target BRD4. Chem Biol. 2015;22:755–63.CrossRef Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase Cereblon to efficiently target BRD4. Chem Biol. 2015;22:755–63.CrossRef
16.
go back to reference Sun B, Fiskus W, Qian Y, et al. BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells. Leukemia. 2018;32:343–52.CrossRef Sun B, Fiskus W, Qian Y, et al. BET protein proteolysis targeting chimera (PROTAC) exerts potent lethal activity against mantle cell lymphoma cells. Leukemia. 2018;32:343–52.CrossRef
17.
go back to reference Ocana A, Garcia-Alonso S, Amir E, Pandiella A. Refining early Antitumoral drug development. Trends Pharmacol Sci. 2018;39:922–5.CrossRef Ocana A, Garcia-Alonso S, Amir E, Pandiella A. Refining early Antitumoral drug development. Trends Pharmacol Sci. 2018;39:922–5.CrossRef
18.
go back to reference Saenz DT, Fiskus W, Qian Y, 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. 2017;31:1951–61.CrossRef Saenz DT, Fiskus W, Qian Y, 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. 2017;31:1951–61.CrossRef
19.
go back to reference Zhang X, Lee HC, Shirazi F, et al. Protein targeting chimeric molecules specific for bromodomain and extra-terminal motif family proteins are active against pre-clinical models of multiple myeloma. Leukemia. 2018;32:2224–39.CrossRef Zhang X, Lee HC, Shirazi F, et al. Protein targeting chimeric molecules specific for bromodomain and extra-terminal motif family proteins are active against pre-clinical models of multiple myeloma. Leukemia. 2018;32:2224–39.CrossRef
Metadata
Title
Activity of BET-proteolysis targeting chimeric (PROTAC) compounds in triple negative breast cancer
Authors
María del Mar Noblejas-López
Cristina Nieto-Jimenez
Miguel Burgos
Mónica Gómez-Juárez
Juan Carlos Montero
Azucena Esparís-Ogando
Atanasio Pandiella
Eva M. Galán-Moya
Alberto Ocaña
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1387-5

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine