Skip to main content
Top
Published in: Breast Cancer Research 1/2021

01-12-2021 | Breast Cancer | Research article

First in class dual MDM2/MDMX inhibitor ALRN-6924 enhances antitumor efficacy of chemotherapy in TP53 wild-type hormone receptor-positive breast cancer models

Authors: Seyed Pairawan, Ming Zhao, Erkan Yuca, Allen Annis, Kurt Evans, David Sutton, Luis Carvajal, Jian-Guo Ren, Solimar Santiago, Vincent Guerlavais, Argun Akcakanat, Coya Tapia, Fei Yang, Priya Subash Chandra Bose, Xiaofeng Zheng, Ecaterina Ileana Dumbrava, Manuel Aivado, Funda Meric-Bernstam

Published in: Breast Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

MDM2/MDMX proteins are frequently elevated in hormone receptor-positive (ER+) breast cancer. We sought to determine the antitumor efficacy of the combination of ALRN-6924, a dual inhibitor of MDM2/MDMX, with chemotherapy in ER+ breast cancer models.

Methods

Three hundred two cell lines representing multiple tumor types were screened to confirm the role of TP53 status in ALRN-6924 efficacy. ER+ breast cancer cell lines (MCF-7 and ZR-75-1) were used to investigate the antitumor efficacy of ALRN-6924 combination. In vitro cell proliferation, cell cycle, and apoptosis assays were performed. Xenograft tumor volumes were measured, and reverse-phase protein array (RPPA), immunohistochemistry (IHC), and TUNEL assay of tumor tissues were performed to evaluate the in vivo pharmacodynamic effects of ALRN-6924 with paclitaxel.

Results

ALRN-6924 was active in wild-type TP53 (WT-TP53) cancer cell lines, but not mutant TP53. On ER+ breast cancer cell lines, it was synergistic in vitro and had enhanced in vivo antitumor activity with both paclitaxel and eribulin. Flow cytometry revealed signs of mitotic crisis in all treatment groups; however, S phase was only decreased in MCF-7 single agent and combinatorial ALRN-6924 arms. RPPA and IHC demonstrated an increase in p21 expression in both combinatorial and single agent ALRN-6924 in vivo treatment groups. Apoptotic assays revealed a significantly enhanced in vivo apoptotic rate in ALRN-6924 combined with paclitaxel treatment arm compared to either single agent.

Conclusion

The significant synergy observed with ALRN-6924 in combination with chemotherapeutic agents supports further evaluation in patients with hormone receptor-positive breast cancer.
Appendix
Available only for authorised users
Literature
2.
go back to reference Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502(7471):333–9.PubMedPubMedCentralCrossRef Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502(7471):333–9.PubMedPubMedCentralCrossRef
3.
go back to reference Ying M, Zhang L, Zhou Q, Shao X, Cao J, Zhang N, et al. The E3 ubiquitin protein ligase MDM2 dictates all-trans retinoic acid-induced osteoblastic differentiation of osteosarcoma cells by modulating the degradation of RARalpha. Oncogene. 2016;35(33):4358–67.PubMedCrossRef Ying M, Zhang L, Zhou Q, Shao X, Cao J, Zhang N, et al. The E3 ubiquitin protein ligase MDM2 dictates all-trans retinoic acid-induced osteoblastic differentiation of osteosarcoma cells by modulating the degradation of RARalpha. Oncogene. 2016;35(33):4358–67.PubMedCrossRef
6.
go back to reference Shadfan M, Lopez-Pajares V, Yuan ZM. MDM2 and MDMX: alone and together in regulation of p53. Transl Cancer Res. 2012;1(2):88–9.PubMed Shadfan M, Lopez-Pajares V, Yuan ZM. MDM2 and MDMX: alone and together in regulation of p53. Transl Cancer Res. 2012;1(2):88–9.PubMed
7.
go back to reference Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387(6630):296–9.PubMedCrossRef Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387(6630):296–9.PubMedCrossRef
8.
go back to reference Oliner JD, Saiki AY, Caenepeel S. The role of MDM2 amplification and overexpression in tumorigenesis. Cold Spring Harb Perspect Med. 2016;6(6):1–16.CrossRef Oliner JD, Saiki AY, Caenepeel S. The role of MDM2 amplification and overexpression in tumorigenesis. Cold Spring Harb Perspect Med. 2016;6(6):1–16.CrossRef
9.
go back to reference Howlader N, Altekruse SF, Li CI, Chen VW, Clarke CA, Ries LA, et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J Natl Cancer Inst. 2014;106(5):55–63.CrossRef Howlader N, Altekruse SF, Li CI, Chen VW, Clarke CA, Ries LA, et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. J Natl Cancer Inst. 2014;106(5):55–63.CrossRef
10.
go back to reference Bertheau P, Lehmann-Che J, Varna M, Dumay A, Poirot B, Porcher R, et al. p53 in breast cancer subtypes and new insights into response to chemotherapy. Breast. 2013;22:S27–S9.PubMedCrossRef Bertheau P, Lehmann-Che J, Varna M, Dumay A, Poirot B, Porcher R, et al. p53 in breast cancer subtypes and new insights into response to chemotherapy. Breast. 2013;22:S27–S9.PubMedCrossRef
11.
go back to reference Haupt S, Buckley D, Pang JM, Panimaya J, Paul PJ, Gamell C, et al. Targeting Mdmx to treat breast cancers with wild-type p53. Cell Death Dis. 2015;6:e1821.PubMedPubMedCentralCrossRef Haupt S, Buckley D, Pang JM, Panimaya J, Paul PJ, Gamell C, et al. Targeting Mdmx to treat breast cancers with wild-type p53. Cell Death Dis. 2015;6:e1821.PubMedPubMedCentralCrossRef
12.
go back to reference Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303(5659):844–8.PubMedCrossRef Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303(5659):844–8.PubMedCrossRef
13.
go back to reference Skalniak L, Surmiak E, Holak TA. A therapeutic patent overview of MDM2/X-targeted therapies (2014-2018). Expert Opin Ther Pat. 2019;29(3):151–70.PubMedCrossRef Skalniak L, Surmiak E, Holak TA. A therapeutic patent overview of MDM2/X-targeted therapies (2014-2018). Expert Opin Ther Pat. 2019;29(3):151–70.PubMedCrossRef
14.
go back to reference Lu J, McEachern D, Li S, Ellis MJ, Wang S. Reactivation of p53 by MDM2 inhibitor MI-77301 for the treatment of endocrine-resistant breast cancer. Mol Cancer Ther. 2016;15(12):2887–93.PubMedPubMedCentralCrossRef Lu J, McEachern D, Li S, Ellis MJ, Wang S. Reactivation of p53 by MDM2 inhibitor MI-77301 for the treatment of endocrine-resistant breast cancer. Mol Cancer Ther. 2016;15(12):2887–93.PubMedPubMedCentralCrossRef
15.
go back to reference Espadinha M, Barcherini V, Lopes EA, Santos MMM. An update on MDMX and dual MDM2/X inhibitors. Curr Top Med Chem. 2018;18(8):647–60.PubMedCrossRef Espadinha M, Barcherini V, Lopes EA, Santos MMM. An update on MDMX and dual MDM2/X inhibitors. Curr Top Med Chem. 2018;18(8):647–60.PubMedCrossRef
16.
go back to reference Carvajal LA, Ben Neriah D, Senecal A, Benard L, Thiruthuvanathan V, Yatsenko T, et al. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Sci Transl Med. 2018;10(436):3003–42.CrossRef Carvajal LA, Ben Neriah D, Senecal A, Benard L, Thiruthuvanathan V, Yatsenko T, et al. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Sci Transl Med. 2018;10(436):3003–42.CrossRef
17.
go back to reference Meric-Bernstam F SM, Infante JR, Goel S, Falchook GS, Shapiro G, et al. Phase I trial of a novel stapled peptide ALRN-6924 disrupting MDMX- and MDM2-mediated inhibition of WT p53 in patients with solid tumors and lymphomas. J Clin Oncol. 2017;35:2505-2505 (poster). Meric-Bernstam F SM, Infante JR, Goel S, Falchook GS, Shapiro G, et al. Phase I trial of a novel stapled peptide ALRN-6924 disrupting MDMX- and MDM2-mediated inhibition of WT p53 in patients with solid tumors and lymphomas. J Clin Oncol. 2017;35:2505-2505 (poster).
18.
go back to reference Ng SY, Yoshida N, Christie AL, Ghandi M, Dharia NV, Dempster J, et al. Targetable vulnerabilities in T- and NK-cell lymphomas identified through preclinical models. Nat Commun. 2018;9(1):2024.PubMedPubMedCentralCrossRef Ng SY, Yoshida N, Christie AL, Ghandi M, Dharia NV, Dempster J, et al. Targetable vulnerabilities in T- and NK-cell lymphomas identified through preclinical models. Nat Commun. 2018;9(1):2024.PubMedPubMedCentralCrossRef
19.
go back to reference Seidman AD, Tiersten A, Hudis C, Gollub M, Barrett S, Yao TJ, et al. Phase II trial of paclitaxel by 3-hour infusion as initial and salvage chemotherapy for metastatic breast cancer. J Clin Oncol. 1995;13(10):2575–81.PubMedCrossRef Seidman AD, Tiersten A, Hudis C, Gollub M, Barrett S, Yao TJ, et al. Phase II trial of paclitaxel by 3-hour infusion as initial and salvage chemotherapy for metastatic breast cancer. J Clin Oncol. 1995;13(10):2575–81.PubMedCrossRef
21.
go back to reference Garrone O, Miraglio E, Vandone AM, Vanella P, Lingua D, Merlano MC. Eribulin in advanced breast cancer: safety, efficacy and new perspectives. Future Oncol. 2017;13(30):2759–69.PubMedCrossRef Garrone O, Miraglio E, Vandone AM, Vanella P, Lingua D, Merlano MC. Eribulin in advanced breast cancer: safety, efficacy and new perspectives. Future Oncol. 2017;13(30):2759–69.PubMedCrossRef
23.
go back to reference Sun W, Tang L. MDM2 increases drug resistance in cancer cells by inducing EMT independent of p53. Curr Med Chem. 2016;23(40):4529–39.PubMedCrossRef Sun W, Tang L. MDM2 increases drug resistance in cancer cells by inducing EMT independent of p53. Curr Med Chem. 2016;23(40):4529–39.PubMedCrossRef
24.
go back to reference Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer. 2008;15(1):257–66.PubMedCrossRef Moreno A, Akcakanat A, Munsell MF, Soni A, Yao JC, Meric-Bernstam F. Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors. Endocr Relat Cancer. 2008;15(1):257–66.PubMedCrossRef
25.
go back to reference Gonzalez-Angulo AM, Hennessy BT, Meric-Bernstam F, Sahin A, Liu W, Ju Z, et al. Functional proteomics can define prognosis and predict pathologic complete response in patients with breast cancer. Clin Proteomics. 2011;8(1):11.PubMedPubMedCentralCrossRef Gonzalez-Angulo AM, Hennessy BT, Meric-Bernstam F, Sahin A, Liu W, Ju Z, et al. Functional proteomics can define prognosis and predict pathologic complete response in patients with breast cancer. Clin Proteomics. 2011;8(1):11.PubMedPubMedCentralCrossRef
26.
go back to reference Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol. 2012;13(1):33–42.PubMedCrossRef Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, et al. EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: analysis of data from the phase 3 FLEX study. Lancet Oncol. 2012;13(1):33–42.PubMedCrossRef
27.
go back to reference Rad FH, Le Buanec H, Paturance S, Larcier P, Genne P, Ryffel B, et al. VEGF kinoid vaccine, a therapeutic approach against tumor angiogenesis and metastases. Proc Natl Acad Sci U S A. 2007;104(8):2837–42.PubMedPubMedCentralCrossRef Rad FH, Le Buanec H, Paturance S, Larcier P, Genne P, Ryffel B, et al. VEGF kinoid vaccine, a therapeutic approach against tumor angiogenesis and metastases. Proc Natl Acad Sci U S A. 2007;104(8):2837–42.PubMedPubMedCentralCrossRef
28.
go back to reference Vu B, Wovkulich P, Pizzolato G, Lovey A, Ding Q, Jiang N, et al. Discovery of RG7112: a small-molecule MDM2 inhibitor in clinical development. ACS Med Chem Lett. 2013;4(5):466–9.PubMedPubMedCentralCrossRef Vu B, Wovkulich P, Pizzolato G, Lovey A, Ding Q, Jiang N, et al. Discovery of RG7112: a small-molecule MDM2 inhibitor in clinical development. ACS Med Chem Lett. 2013;4(5):466–9.PubMedPubMedCentralCrossRef
29.
go back to reference Ding Q, Zhang Z, Liu JJ, Jiang N, Zhang J, Ross TM, et al. Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development. J Med Chem. 2013;56(14):5979–83.PubMedCrossRef Ding Q, Zhang Z, Liu JJ, Jiang N, Zhang J, Ross TM, et al. Discovery of RG7388, a potent and selective p53-MDM2 inhibitor in clinical development. J Med Chem. 2013;56(14):5979–83.PubMedCrossRef
30.
go back to reference Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T, Brinkley BR, et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma. 1997;106(6):348–60.PubMedCrossRef Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T, Brinkley BR, et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation. Chromosoma. 1997;106(6):348–60.PubMedCrossRef
31.
go back to reference Antonucci LA, Egger JV, Krucher NA. Phosphorylation of the retinoblastoma protein (Rb) on serine-807 is required for association with Bax. Cell Cycle. 2014;13(22):3611–7.PubMedPubMedCentralCrossRef Antonucci LA, Egger JV, Krucher NA. Phosphorylation of the retinoblastoma protein (Rb) on serine-807 is required for association with Bax. Cell Cycle. 2014;13(22):3611–7.PubMedPubMedCentralCrossRef
32.
go back to reference Mori S, Nada S, Kimura H, Tajima S, Takahashi Y, Kitamura A, et al. The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase. Plos One. 2014;9(2):1–12.CrossRef Mori S, Nada S, Kimura H, Tajima S, Takahashi Y, Kitamura A, et al. The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase. Plos One. 2014;9(2):1–12.CrossRef
33.
go back to reference Morrish F, Neretti N, Sedivy JM, Hockenbery DM. The oncogene c-Myc coordinates regulation of metabolic networks to enable rapid cell cycle entry. Cell Cycle. 2008;7(8):1054–66.PubMedCrossRef Morrish F, Neretti N, Sedivy JM, Hockenbery DM. The oncogene c-Myc coordinates regulation of metabolic networks to enable rapid cell cycle entry. Cell Cycle. 2008;7(8):1054–66.PubMedCrossRef
35.
go back to reference Dufner A, Thomas G. Ribosomal S6 kinase signaling and the control of translation. Exp Cell Res. 1999;253(1):100–9.PubMedCrossRef Dufner A, Thomas G. Ribosomal S6 kinase signaling and the control of translation. Exp Cell Res. 1999;253(1):100–9.PubMedCrossRef
36.
go back to reference Meric-Bernstam F, Zheng X, Shariati M, Damodaran S, Wathoo C, Brusco L, et al. Survival outcomes by TP53 mutation status in metastatic breast cancer. JCO Precis Oncol. 2018;2018. Meric-Bernstam F, Zheng X, Shariati M, Damodaran S, Wathoo C, Brusco L, et al. Survival outcomes by TP53 mutation status in metastatic breast cancer. JCO Precis Oncol. 2018;2018.
37.
go back to reference Yu Q, Li Y, Mu K, Li ZS, Meng QY, Wu XJ, et al. Amplification of Mdmx and overexpression of MDM2 contribute to mammary carcinogenesis by substituting for p53 mutations. Diagn Pathol. 2014;9. Yu Q, Li Y, Mu K, Li ZS, Meng QY, Wu XJ, et al. Amplification of Mdmx and overexpression of MDM2 contribute to mammary carcinogenesis by substituting for p53 mutations. Diagn Pathol. 2014;9.
38.
go back to reference Kim JY, Jeong HS, Chung T, Kim M, Lee JH, Jung WH, et al. The value of phosphohistone H3 as a proliferation marker for evaluating invasive breast cancers: a comparative study with Ki67. Oncotarget. 2017;8(39):65064–76.PubMedPubMedCentralCrossRef Kim JY, Jeong HS, Chung T, Kim M, Lee JH, Jung WH, et al. The value of phosphohistone H3 as a proliferation marker for evaluating invasive breast cancers: a comparative study with Ki67. Oncotarget. 2017;8(39):65064–76.PubMedPubMedCentralCrossRef
39.
go back to reference Ozawa K. Reduction of phosphorylated histone H3 serine 10 and serine 28 cell cycle marker intensities after DNA damage. Cytometry A. 2008;73(6):517–27.PubMedCrossRef Ozawa K. Reduction of phosphorylated histone H3 serine 10 and serine 28 cell cycle marker intensities after DNA damage. Cytometry A. 2008;73(6):517–27.PubMedCrossRef
40.
go back to reference Wang D, Lippard SJ. Cisplatin-induced post-translational modification of histones H3 and H4. J Biol Chem. 2004;279(20):20622–5.PubMedCrossRef Wang D, Lippard SJ. Cisplatin-induced post-translational modification of histones H3 and H4. J Biol Chem. 2004;279(20):20622–5.PubMedCrossRef
41.
go back to reference Flores ML, Castilla C, Avila R, Ruiz-Borrego M, Saez C, Japon MA. Paclitaxel sensitivity of breast cancer cells requires efficient mitotic arrest and disruption of Bcl-xL/Bak interaction. Breast Cancer Res Treat. 2012;133(3):917–28.PubMedCrossRef Flores ML, Castilla C, Avila R, Ruiz-Borrego M, Saez C, Japon MA. Paclitaxel sensitivity of breast cancer cells requires efficient mitotic arrest and disruption of Bcl-xL/Bak interaction. Breast Cancer Res Treat. 2012;133(3):917–28.PubMedCrossRef
42.
go back to reference Funda Meric-Bernstam MNS, Jeffrey R. Infante, Sanjay Goel, Gerald Steven Falchook, Geoffrey Shapiro, Ki Y Chung, Robert Martin Conry, David S. Hong, Judy Sing-Zan Wang, Ulrich Steidl, Loren D. Walensky, Vincent Guerlavais, Marie Payton, D. Allen Annis, Manuel Aivado, Manish R. Patel. Phase I trial of a novel stapled peptide ALRN-6924 disrupting MDMX- and MDM2-mediated inhibition of WT p53 in patients with solid tumors and lymphomas. J Clin Oncol 2017;35(15):Suppl. Funda Meric-Bernstam MNS, Jeffrey R. Infante, Sanjay Goel, Gerald Steven Falchook, Geoffrey Shapiro, Ki Y Chung, Robert Martin Conry, David S. Hong, Judy Sing-Zan Wang, Ulrich Steidl, Loren D. Walensky, Vincent Guerlavais, Marie Payton, D. Allen Annis, Manuel Aivado, Manish R. Patel. Phase I trial of a novel stapled peptide ALRN-6924 disrupting MDMX- and MDM2-mediated inhibition of WT p53 in patients with solid tumors and lymphomas. J Clin Oncol 2017;35(15):Suppl.
43.
go back to reference Konopleva M, Martinelli G, Daver N, Papayannidis C, Wei A, Higgins B, et al. MDM2 inhibition: an important step forward in cancer therapy. Leukemia. 2020;34(11):2858–74.PubMedCrossRef Konopleva M, Martinelli G, Daver N, Papayannidis C, Wei A, Higgins B, et al. MDM2 inhibition: an important step forward in cancer therapy. Leukemia. 2020;34(11):2858–74.PubMedCrossRef
44.
go back to reference Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, et al. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature. 2012;485(7396):55–61.PubMedPubMedCentralCrossRef Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, et al. The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature. 2012;485(7396):55–61.PubMedPubMedCentralCrossRef
45.
go back to reference Kanaizumi H, Higashi C, Tanaka Y, Hamada M, Shinzaki W, Azumi T, et al. PI3K/Akt/mTOR signalling pathway activation in patients with ER-positive, metachronous, contralateral breast cancer treated with hormone therapy. Oncol Lett. 2019;17(2):1962–8.PubMed Kanaizumi H, Higashi C, Tanaka Y, Hamada M, Shinzaki W, Azumi T, et al. PI3K/Akt/mTOR signalling pathway activation in patients with ER-positive, metachronous, contralateral breast cancer treated with hormone therapy. Oncol Lett. 2019;17(2):1962–8.PubMed
46.
go back to reference Chumsri S, Sabnis G, Tkaczuk K, Brodie A. mTOR inhibitors: changing landscape of endocrine-resistant breast cancer. Future Oncol. 2014;10(3):443–56.PubMedCrossRef Chumsri S, Sabnis G, Tkaczuk K, Brodie A. mTOR inhibitors: changing landscape of endocrine-resistant breast cancer. Future Oncol. 2014;10(3):443–56.PubMedCrossRef
47.
go back to reference Mayer I. Role of mTOR inhibition in preventing resistance and restoring sensitivity to hormone-targeted and HER2-targeted therapies in breast cancer. Clin Adv Hematol Oncol. 2013;11(4):217–24.PubMedPubMedCentral Mayer I. Role of mTOR inhibition in preventing resistance and restoring sensitivity to hormone-targeted and HER2-targeted therapies in breast cancer. Clin Adv Hematol Oncol. 2013;11(4):217–24.PubMedPubMedCentral
48.
go back to reference Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, Sahmoud T, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366(6):520–9.PubMedCrossRef Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, Sahmoud T, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366(6):520–9.PubMedCrossRef
50.
go back to reference Walerych D, Pruszko M, Zyla L, Wezyk M, Gaweda-Walerych K, Zylicz A. Wild-type p53 oligomerizes more efficiently than p53 hot-spot mutants and overcomes mutant p53 gain-of-function via a “dominant-positive” mechanism. Oncotarget. 2018;9(62):32063–80.PubMedPubMedCentralCrossRef Walerych D, Pruszko M, Zyla L, Wezyk M, Gaweda-Walerych K, Zylicz A. Wild-type p53 oligomerizes more efficiently than p53 hot-spot mutants and overcomes mutant p53 gain-of-function via a “dominant-positive” mechanism. Oncotarget. 2018;9(62):32063–80.PubMedPubMedCentralCrossRef
51.
go back to reference Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. 2017;23(15):4242–50.PubMedPubMedCentralCrossRef Kato S, Goodman A, Walavalkar V, Barkauskas DA, Sharabi A, Kurzrock R. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. 2017;23(15):4242–50.PubMedPubMedCentralCrossRef
52.
go back to reference Munoz-Fontela C, Mandinova A, Aaronson SA, Lee SW. Emerging roles of p53 and other tumour-suppressor genes in immune regulation. Nat Rev Immunol. 2016;16(12):741–50.PubMedPubMedCentralCrossRef Munoz-Fontela C, Mandinova A, Aaronson SA, Lee SW. Emerging roles of p53 and other tumour-suppressor genes in immune regulation. Nat Rev Immunol. 2016;16(12):741–50.PubMedPubMedCentralCrossRef
54.
go back to reference Luis ACPJR, Solimar S, Manoj S, David S, Vincent G, et al. The stapled peptide ALRN-6924, a dual inhibitor of MDMX and MDM2, displays immunomodulatory activity and enhances immune checkpoint blockade in syngeneic mouse models. J Immunotherapy Cancer. 2018;115. Luis ACPJR, Solimar S, Manoj S, David S, Vincent G, et al. The stapled peptide ALRN-6924, a dual inhibitor of MDMX and MDM2, displays immunomodulatory activity and enhances immune checkpoint blockade in syngeneic mouse models. J Immunotherapy Cancer. 2018;115.
Metadata
Title
First in class dual MDM2/MDMX inhibitor ALRN-6924 enhances antitumor efficacy of chemotherapy in TP53 wild-type hormone receptor-positive breast cancer models
Authors
Seyed Pairawan
Ming Zhao
Erkan Yuca
Allen Annis
Kurt Evans
David Sutton
Luis Carvajal
Jian-Guo Ren
Solimar Santiago
Vincent Guerlavais
Argun Akcakanat
Coya Tapia
Fei Yang
Priya Subash Chandra Bose
Xiaofeng Zheng
Ecaterina Ileana Dumbrava
Manuel Aivado
Funda Meric-Bernstam
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2021
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-021-01406-x

Other articles of this Issue 1/2021

Breast Cancer Research 1/2021 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