The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in triple-negative breast cancer models as single agent and in combination with everolimus

Ramiro Vázquez, María E Riveiro, Lucile Astorgues-Xerri, Elodie Odore, Keyvan Rezai, Eugenio Erba, Nicolò Panini, Andrea Rinaldi, Ivo Kwee, Luca Beltrame, Mohamed Bekradda, Esteban Cvitkovic, Francesco Bertoni, Roberta Frapolli, Maurizio D'Incalci, Ramiro Vázquez, María E Riveiro, Lucile Astorgues-Xerri, Elodie Odore, Keyvan Rezai, Eugenio Erba, Nicolò Panini, Andrea Rinaldi, Ivo Kwee, Luca Beltrame, Mohamed Bekradda, Esteban Cvitkovic, Francesco Bertoni, Roberta Frapolli, Maurizio D'Incalci

Abstract

Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous subgroup of breast tumors clinically defined by the lack of estrogen, progesterone and HER2 receptors, limiting the use of the targeted therapies employed in other breast malignancies. Recent evidence indicates that c-MYC is a key driver of TNBC. The BET-bromodomain inhibitor OTX015 (MK-8628) has potent antiproliferative activity accompanied by c-MYC down-regulation in several tumor types, and has demonstrated synergism with the mTOR inhibitor everolimus in different models. The aim of this study was to evaluate the anti-tumor activity of OTX015 as single agent and in combination with everolimus in TNBC models. OTX015 was assayed in three human TNBC-derived cell lines, HCC1937, MDA-MB-231 and MDA-MB-468, all showing antiproliferative activity after 72 h (GI50 = 75-650 nM). This was accompanied by cell cycle arrest and decreased expression of cancer stem cells markers. However, c-MYC protein and mRNA levels were only down-regulated in MDA-MB-468 cells. Gene set enrichment analysis showed up-regulation of genes involved in epigenetic control of transcription, chromatin and the cell cycle, and down-regulation of stemness-related genes. In vitro, combination with everolimus was additive in HCC1937 and MDA-MB-231 cells, but antagonistic in MDA-MB-468 cells. In MDA-MB-231 murine xenografts, tumor mass was significantly (p < 0.05) reduced by OTX015 with respect to vehicle-treated animals (best T/C = 40.7%). Although everolimus alone was not active, the combination was more effective than OTX015 alone (best T/C = 20.7%). This work supports current clinical trials with OTX015 in TNBC (NCT02259114).

Keywords: OTX015 (MK-8628); bromodomain inhibitor; everolimus; triple-negative breast cancer.

Conflict of interest statement

CONFLICTS OF INTEREST

Esteban Cvitkovic was founder and CSO of Oncoethix SA, and is CEO of Oncology Therapeutic Development. Lucile Astorgues-Xerri, Elodie Odore, Mohamed Bekradda and María E. Riveiro were or are employees of Oncology Therapeutic Development. Francesco Bertoni and Maurizio D’Incalci received institutional funds from Oncology Therapeutic Development. The other authors disclosed no potential conflicts of interest.

Figures

Figure 1. Characterization of OTX015 antiproliferative activity…
Figure 1. Characterization of OTX015 antiproliferative activity in three TNBC cell models
(A) Cells were seeded under normoxic and hypoxic conditions and treated with OTX015 for 72 h. The antiproliferative effect was determined by cell counting. The diagonal-line pattern () indicates OTX015 concentrations that decreased cell growth by 50% (GI50). For each treatment, the influence of hypoxia on antiproliferative activity was evaluated with the Student t-test. Significant differences in the OTX015 treatments were determined with a one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test; substantial differences (p < 0.05) are indicated as letters above the bars. (B) Cell lines were treated with OTX015 for 72 h (indicated by the green area) and then cultured in drug-free medium. The Y axis represents the number of cells per well evaluated every 24 h after cell seeding (time 0). Significant differences in the number of control and treated cells at each time point were determined by two-way ANOVA test (p < 0.001) followed by a Bonferroni a posteriori test (*p < 0.05, **p < 0.01, ***p < 0.001). Significant differences at each time point after washout versus 72 h-treated cells were determined with one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test (#p < 0.05). (C) Cells were treated with OTX015 for 24, 48 and 72 h and the latter cells were further cultured without OTX015 for 48 h (drug washout). Significant differences in the percentage of OTX015-treated cells in the G1, S and G2/M phases with respect to untreated controls were evaluated by a one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test. After washout, differences in the percentage of cells in a given cell cycle phase between control and pretreated cells were determined with the Student t-test. Significant differences between treated cells and controls were reported for the G1 phase (*p < 0.05, **p < 0.01), S phase (#p < 0.05, ##p < 0.01) and G2/M phase (×p < 0.05, ××p < 0.01). Each dot or bar and vertical line represents the mean ± SEM, respectively (n ≥ 3).
Figure 2. Baseline and post-OTX015 expression of…
Figure 2. Baseline and post-OTX015 expression of BRD2/3/4 and c-MYC
Basel expression of BRD2/3/4 and c-MYC in terms of protein (A) and mRNA (B) levels were evaluated in the three cell lines by Western blotting and qRT-PCR, respectively. Protein (C) and mRNA (D) levels were evaluated after 24-, 48- and 72-h OTX015 (650 nM in HCC1937 and MDA-MB-468 cells, and 75 nM in the MDA-MB-231 cell line). Significant differences in mRNA levels were determined by one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test (*p < 0.05, **p < 0.01). Western blots are representative of at least three independent experiments. Bars and vertical lines represent the mean ± SEM, respectively (n = 3).
Figure 3. Gene expression profiling in OTX015-treated…
Figure 3. Gene expression profiling in OTX015-treated TNBC cell models
(A) GEP heat maps in OTX015-treated MDA-MB-231 and MDA-MB-468 cell lines, compared to vehicle-treated control cells. Red and blue indicate up-regulated and down-regulated genes respectively. (B) Venn diagram of 81 and 1286 differentially expressed genes in MDA-MD-231 and MDA-MB-468 OTX015-treated cells, respectively. GSEA analyses showed common signatures enriched, using the complete MSigDB (C), up-regulated and (D), down-regulated) and CMAP databases (E) in OTX015-treated MDA-MB-231 and MDA-MB-468 cell lines. FDR, false discovery rate; NES, normalized enrichment score.
Figure 4. Effect of OTX015 on the…
Figure 4. Effect of OTX015 on the expression of cancer stem cell markers in TNBC cell models
Cells were exposed for 24, 48 and 72 h to OTX015 (650 nM for HCC1937 - ■ - and MDA-MB-468 - ■ -, and 75 nM for MDA-MB-231 cells - ■ -) and expression of stemness-related genes was determined by qRT-PCR. Differences in the mRNA levels of OTX015-treated cells vs. controls were determined by a one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test (*p < 0.05, **p < 0.01). Bars and vertical lines represent the mean ± SEM, respectively (n = 3).
Figure 5. Antitumor activity of OTX015 in…
Figure 5. Antitumor activity of OTX015 in combination with everolimus in vitro and in MDA-MB-231 xenografts
(A) Antiproliferative effects of concomitant OTX015 and everolimus exposure for 72 h was evaluated with the MTT assay in all three TNBC cell lines. Synergism is defined as a combination index < 0.9; values between 0.9 and 1.10 indicate an additive effect (shown as dotted lines); and values > 1.10, indicate antagonism (n = 3). (B) Antitumor effects of 50 mg/kg OTX015 twice daily (□) and 2 mg/kg everolimus thrice daily (RAD001; ) were evaluated as single agents and in combination () in MDA-MB-231 murine xenografts. Tumor weight was compared between drug regimens at each time point using a two-way ANOVA test (p < 0.001) followed by Bonferroni a posteriori test on log-transformed data. Color-filled symbols indicate significant (p < 0.05) differences between treated vs. vehicle mice (·). Significant differences in tumor mass between the combination versus single agent treatment after drug withdrawal are shown (*p < 0.05, **p < 0.01 and ***p < 0.001). Dotted lines define the treatment period. (C) Animal body weight during treatment (dotted lines) and subsequently. Results represent the mean ± SEM (during treatment, n = 9). Arrows in B and C indicate the change in the everolimus dosing schedule. (D) OTX015 levels in plasma and tissues from OTX015-treated mice, 4 h after the last dose. Variations in OTX015 concentrations detected between plasma, tumor and peritumoral tissue were evaluated by a one-way ANOVA test (p < 0.01) followed by an SNK a posteriori test. Letters above the bars indicate substantial differences (p < 0.05). (E) mRNA levels of c-MYC, BRD2/3/4, and CSC markers were determined by qRT-PCR in MDA-MB-231 tumors after 4-week treatment with OTX015 (■), everolimus (RAD001, ■) or the combination (■). Variations between the experimental groups were evaluated by a one-way ANOVA test (p < 0.001) followed by an SNK a posteriori test. Letters above the bars indicate significant differences (p < 0.05) in mRNA levels among the experimental groups. Bars and vertical lines represent the mean ± SEM, respectively (n = 4).

References

    1. Chacón RD, Costanzo MV. Triple-negative breast cancer. Breast Cancer Res. 2010;12:S3.
    1. Bottino MC, Cerliani JP, Rojas P, Giulianelli S, Soldati R, Mondillo C, Gorostiaga MA, Pignataro OP, Calvo JC, Gutkind JS, Amornphimoltham Panowat, Molinolo AA, Lüthy IA, et al. Classical membrane progesterone receptors in murine mammary carcinomas: agonistic effects of progestins and RU-486 mediating rapid non-genomic effects. Breast Cancer Res Treat. 2011;126:621–36.
    1. Montero JC, Esparís-Ogando A, Re-Louhau MF, Seoane S, Abad M, Calero R, Ocaña A, Pandiella A. Active kinase profiling, genetic and pharmacological data define mTOR as an important target in triple-negative breast cancer. Oncogene. 2014;33:148–156.
    1. Horiuchi D, Kusdra L, Huskey NE, Chandriani S, Lenburg ME, Gonzalez-Angulo AM, Creasman KJ, Bazarov AV, Smyth JW, Davis SE, Yaswen P, Mills GB, Esserman LJ, et al. MYC pathway activation in triple-negative breast cancer is synthetic lethal with CDK inhibition. J Exp Med. 2012;209:679–96.
    1. Saal LH, Holm K, Maurer M, Memeo L, Su T, Wang X, Js Yu, Malmström PO, Mansukhani M, Enoksson J, Hibshoosh H, Borg A, Parsons R. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast cancer. Cancer Res. 2005;65:2554–9.
    1. Downward J. PI 3-kinase, Akt and cell survival. Semin Cell Dev Biol. 2004;15:177–82.
    1. Martins FC, De S, Almendro V, Gönen M, Park SY, Blum JL, Herlihy W, Ethington G, Schnitt SJ, Tung N, Garber JE, Fetten K, Michor F, et al. Evolutionary pathways in BRCA1-associated breast tumors. Cancer Discov. 2012;2:503–11.
    1. Wu Y, Sarkissyan M, Elshimali Y, Vadgama JV. Triple negative breast tumors in African-American and Hispanic/Latina women are high in CD44+, low in CD24+, and have loss of PTEN. PLoS One. 2013;8:e78259.
    1. Marty B, Maire V, Gravier E, Rigaill G, Vincent-Salomon A, Kappler M, Lebigot I, Djelti F, Tourdès A, Gestraud P, Hupé P, Barillot E, Cruzalegui F, et al. Frequent PTEN genomic alterations and activated phosphatidylinositol 3-kinase pathway in basal-like breast cancer cells. Breast Cancer Res. 2008;10:R101.
    1. Gordon V, Banerji S. Molecular pathways: PI3K pathway targets in triple-negative breast cancers. Clin Cancer Res. 2013;19:3738–44.
    1. Berrada N, Delaloge S, André F. Treatment of triple-negative metastatic breast cancer: toward individualized targeted treatments or chemosensitization? Ann Oncol. 2010;21:vii30–5.
    1. Muller S, Filippakopoulos P, Knapp S. Bromodomains as therapeutic targets. Expert Rev Mol Med. 2011;13:e29.
    1. Noel JK, Iwata K, Ooike S, Sugahara K, Nakamura H, Daibata M. Development of the BET bromodomain inhibitor OTX015 (Abstract) Mol Cancer Ther. 2013;12:C244.
    1. Boi M, Gaudio E, Bonetti P, Kwee I, Bernasconi E, Tarantelli C, Rinaldi A, Testoni M, Cascione L, Ponzoni M, Mensah AA, Stathis A, Stussi G, et al. The BET bromodomain inhibitor OTX015 affects pathogenetic pathways in preclinical B-cell tumor models and synergizes with targeted drugs. Clin Cancer Res. 2015;2:1628–38.
    1. Coudé MM, Braun T, Berrou J, Dupont M, Bertrand S, Masse A, Raffoux E, Itzykson R, Delord M, Riveiro ME, Herait P, Baruchel A, Dombret H, et al. BET inhibitor OTX015 targets BRD2 and BRD4 and decreases c-MYC in acute leukemia cells. Oncotarget. 2015;6:17698–712. doi: 10.18632/oncotarget.4131.
    1. Henssen A, Althoff K, Odersky A, Beckers A, Koche R, Speleman F, Schäfers S, Bell E, Nortmeyer M, Westermann F, K De Preters, Florin A, Heukamp L, et al. Targeting MYCN-driven transcription by BET-bromodomain inhibition. Clin Cancer Res. 2015. [Epub ahead of print]
    1. Ansagami IA, Wilder-Roman K, Dommeti VL, Krishnamurthy PM, Apel IJ, Escara-Wilke J, Plymate SR, Navone NM, Wang S, Feng FY, Chinnaiyan AM. BET bromodomain inhibitors enhace efficacy and disrupt resistance to AR antagonists in the treatment of prostate cancer. Mol Cancer Res. 2016;14:324–31.
    1. Berenguer-Daizé C, Astorgues-Xerri L, Odore E, Cayol M, Cvitkovic E, Noel K, Bekradda M, MacKenzie S, Rezai K, Lokiec F, Riveiro ME, Ouafik L. OTX015 (MK-8628), a novel BET inhibitor, displays in vitro and in vivo antitumor effects alone and in combination with conventional therapies in glioblastoma . J Cancer. 2016;139:2047–55.
    1. Stathis A, Zucca E, Bekradda M, Gómez-Roca C, Delord JP, de La Motte Rouge T, Uro-Coste E, de Braud F, Pelosi G, French CA. Clinical response of carcinomas harboring the BRD4-NUT oncoprotein to targeted bromodomain inhibitor OTX015/MK-8628. Cancer Discov. 2016;6:492–500.
    1. Berthon C, Raffoux E, Thomas X, Vey N, Gómez-Roca C, Yee K, Taussing DC, Rezai K, Roumier C, Herait P, Kahatt C, Quesnel B, Michallet M, et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 2016;3:e186–95.
    1. Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, Morschhauser F, Karlin L, Broussais F, Rezai K, Herait P, Kahatt C, Lokiec F, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 2016;3:e196–204.
    1. Shu S, Lin CY, HE HH, Witwicki RM, Tabassum DP, Roberts JM, Janiszewska M, Huh SJ, Liang Y, Ryan J, Doherty E, Mohammed H, Guo H, et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature. 2016;529:413–7.
    1. Gaudio E, Tarantelli C, Ponzoni M, Odore E, Rezai K, Bernasconi E, Cascione L, Rinaldi A, Stathis A, Riveiro E, Cvitkovic E, Zucca E, Bertoni F. Bromodomain inhibitor OTX015 (MK-8628) combined with targeted agents shows strong in vivo antitumor activity in lymphoma. Oncotarget. 2016;7:58142–58147. doi: 10.18632/oncotarget.10983.
    1. Chávez KJ, Garimella SV, Lipñowitz S. Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer. Breast Dis. 2012;32:35–48.
    1. Vaupel P, Höckel M, Mayer A. Detection and characterization of tumor hypoxia using pO2 histography. Antioxid Redox Signal. 2007;9:1221–35.
    1. Brown JM. Tumor microenvironment and the response to anticancer therapy. Cancer Biol Ther. 2002;1:453–8.
    1. Jo M, Eastman BM, Webb DL, Stoletov K, Klemke R, Gonias SL. Cell signaling by urokinase-type plasminogen activator receptor induces stem cell-like properties in breast cancer cells. Cancer Res. 2010;70:8945–58.
    1. Laster WR, Jr, Schabel FM, Jr, Skipper HE, Wilcox WS, Jr Thompson. Experimental evaluation of potential anticancer agents. IV. Host weight loss as it relates to false positives in drug evaluation. Cancer Res. 1961;21:895–906.
    1. Moher ED, Reilly M, Grieco PA, Corbett TH, Valeriote FA. Synthetic studies on quassinoids: transformation of (-)-glaucarubolone into (-)-peninsularinone. In vivo antitumor evaluation of (-)-glaucarubolone, (-)-chaparrinone, and (-)-peninsularinone. J Org Chem. 1998;63:3508–10.
    1. Fowler T, Ghatak P, Price DH, Conaway R, Conaway J, Chiang CM, Bradner JE, Shilatifard A, Roy AL. Regulation of MYC expression and differential JQ1 sensitivity in cancer cells. PloS One. 2014;9:e87003.
    1. Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, Bergeron L, Sims RJ., 3rd Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci USA. 2011;108:16669–74.
    1. Yunokawa M, Koizumi F, Kitamura Y, Katanasaka Y, Okamato N, Kodaira M, Yonemori K, Shimizu C, Ando M, Masutomi K, Yoshida T, Fujiwara Y, Tamura K. Efficacy of everolimus, a novel mTOR inhibitor, against basal-like triple-negative breast cancer cells. Cancer Sci. 2012;103:1668–71.
    1. Hurvitz SA, Kalous O, Conklin D, Desai AJ, Dering J, Anderson L, O’Brien NA, Kolarova T, Finn RS, Linnartz R, Chen D, Slamon DJ. In vitro activity of the mTOR inhibitor everolimus, in a large panel of breast cancer cell lines and analysis for predictors of response. Breast Cancer Res Treat. 2015;149:669–680.
    1. Yardley DA, Noguchi S, Pritchard KI, Burris HA, 3rd, Baselga J, Gnant M, Hortobagyi GN, Campone M, Pistilli B, Piccart B, Piccart M, Melichar B, Petrakova K, et al. Everolimus plus exemestane in postmenopausal patients with HR(+) breast cancer: BOLERO-2 final progression-free survival analysis. Adv Ther. 2013;30:870–84.
    1. Piccart M, Hortobagyi GN, Campone M, Pritchard KI, Lebrun F, Ito Y, Noguchi S, Perez A, Rugo HS, Deleu I, Burris HA, 3rd, Provencher L, Neven P, et al. Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2. Ann Oncol. 2014;25:2357–62.
    1. Stuhmiller TJ, Miller SM, Zawistowski JS, Nakamura K, Beltran AS, Duncan JS, Angus SP, Collins KA, Granger DA, Reuthrt RA, Graves LM, Gomez SM, Kuan PF, et al. Inhibition of lapatinib-induced kinome reprogramming in ERBB2-positive breast cancer by targeting BET family bromodomains. Cell Rep. 2015;11:390–404.
    1. Bihani T, Ezell SA, Ladd B, Grosskurth SE, Mazzola AM, Pietras M, Reimer C, Zinda M, Fawell S, D’Cruz CM. Resistance to everolimus driven by epigenetic regulation of MYC in ER+ breast cancers. Oncotarget. 2015;6:2407–20. doi: 10.18632/oncotarget.2964.
    1. Liu CY, Hu MH, Hsu CJ, Huang CT, Wang DS, Tsai WC, Chen YT, Lee CH, Chu PY, Hsu CC, Chen MH, Shiau CW, Tseng LM, et al. Lapatinib inhibits CIP2A/PP2A/p-Akt signaling and induces apoptosis in triple negative breast cancer cells. Oncotarget. 2016;7:9135–49. doi: 10.18632/oncotarget.7035.
    1. Eaves CJ. Cancer stem cells: Here, there, everywhere? Nature. 2008;456:581–2.
    1. Abubaker K, Latifi A, Luwor R, Nazaretian S, Zhu H, Quinn MA, Thompson EW, Findlay JK, Ahmed N. Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to and increased tumor burden. Mol Cancer. 2013;12:24. doi: 10.1186/1476-4598-12-24.
    1. Bhola NE, Balko JM, Dugger TC, Kuba MG, Sánchez V, Sanders M, Satanford J, Cook RS, Arteaga CL. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013;123:1348–58.
    1. Horne GA, Stewart HJ, Dickson J, Knapp S, Ramsahoye B, Chevassut T. Nanog requires BRD4 to mantain murine embryonic stem cell pluripotency and is suppressed by bromodomain inhibitor JQ1 together with Lefty1. Stem Cells Dev. 2015. pp. 4879–91.
    1. Shi J, Wang Y, Zeng L, Wu Y, Deng J, Zhang Q, Lin Y, Li J, Kang T, Tao M, Rusinova E, Zhang G, Wang C, et al. Disrupting the interaction of BRD4 with diacetylated Twist suppresses tumorigenesis in basal-like breast cancer. Cancer Cell. 2014;25:210–25.
    1. Herrmann H, Blatt K, Shi J, Gleixner KV, Cerny-Reiterer S, Müllauer L, Vakoc CR, Sperr WR, Horny HP, Bradner JE, Zuber J, Valent P. Small-molecule inhibitor of BRD4 as a new potent approach to eliminate leukemic stem- and progenitor cell in acute myeloid leukemia (AML) Oncotarget. 2012;3:1588–99. doi: 10.18632/oncotarget.733.
    1. Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–81.
    1. Bertuzzi A, Gandolfi A, Germani A, Spano M, Starace G, Vitelli R. Analysis of DNA synthesis rate of cultured cells from flow cytometric data. Cytometry. 1984;5:619–28.
    1. Vandesompele J, K De Peter, Pattyn F, Poppe B, N Van Roy, A De Paepe, Speleman F. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol. 2002;3:RESEARCH0034.
    1. Odore E, Lokiec F, Weill S, Noel JK, Herait P, Bekradda M, Riveiro ME, Rezai K. Development and validation of an UPLC-MS/MS method for quantitative analysis of OTX015 in human plasma samples. Anal Methods. 2014;6:9108–15.

Source: PubMed

3
Subscribe