40
Views
0
CrossRef citations to date
0
Altmetric
SHORT REPORT

Role of AURKB Inhibition in Reducing Proliferation and Enhancing Effects of Radiotherapy in Triple-Negative Breast Cancer

, , & ORCID Icon
Pages 341-346 | Received 17 Oct 2023, Accepted 04 Jun 2024, Published online: 09 Jul 2024

References

  • Arnold M, Morgan E, Rumgay H, et al. Current and future burden of breast cancer: global statistics for 2020 and 2040. Breast. 2022;66:15–23.
  • Mahmoud R, Ordóñez-Morán P, Allegrucci C. Challenges for triple negative breast cancer treatment: defeating heterogeneity and cancer stemness. Cancers. 2022;14(17):4280. doi:10.3390/cancers14174280
  • Burguin A, Diorio C, Durocher F. Breast cancer treatments: updates and new challenges. J Pers Med. 2021;11(8):808. doi:10.3390/jpm11080808
  • Moran MS. Radiation therapy in the locoregional treatment of triple-negative breast cancer. Lancet Oncol. 2015;16(3):e113–e122. doi:10.1016/S1470-2045(14)71104-0
  • Ahmed M, Jozsa F, Douek M. A systematic review of neo-adjuvant radiotherapy in the treatment of breast cancer. Ecancermedicalscience. 2021;15:1175. doi:10.3332/ecancer.2021.1175
  • Sousa C, Cruz M, Neto A, et al. Neoadjuvant radiotherapy in the approach of locally advanced breast cancer. ESMO Open. 2020;4(Suppl 2):e000640.
  • Bhat V, Pellizzari S, Allan AL, et al. Radiotherapy and radiosensitization in breast cancer: molecular targets and clinical applications. Crit Rev Oncol Hematol. 2022;169:103566. doi:10.1016/j.critrevonc.2021.103566
  • Ahmad SS, Crittenden MR, Tran PT, et al. Clinical development of novel drug-radiotherapy combinations. Clin Cancer Res. 2019;25(5):1455–1461. doi:10.1158/1078-0432.CCR-18-2466
  • Sharma RA, Plummer R, Stock JK, et al. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol. 2016;13(10):627–642.
  • Parsyan A, Cruickshank J, Hodgson K, et al. Anticancer effects of radiation therapy combined with Polo-Like Kinase 4 (PLK4) inhibitor CFI-400945 in triple negative breast cancer. Breast. 2021;58:6–9. doi:10.1016/j.breast.2021.03.011
  • Huang R-X, Zhou P-K. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther. 2020;5(1):60. doi:10.1038/s41392-020-0150-x
  • Alam MS, Sultana A, Wang G, Haque Mollah MN. Gene expression profile analysis to discover molecular signatures for early diagnosis and therapies of triple-negative breast cancer. Front Mol Biosci. 2022;9:1049741. doi:10.3389/fmolb.2022.1049741
  • Tchatchou S, Wirtenberger M, Hemminki K, et al. Aurora kinases A and B and familial breast cancer risk. Cancer Lett. 2007;247(2):266–272. doi:10.1016/j.canlet.2006.05.002
  • Yuan K, Wu M, Lyu S, Li Y. Identification of prognostic genes for early basal-like breast cancer with weighted gene co-expression network analysis. Medicine. 2022;101(42):e30581. doi:10.1097/MD.0000000000030581
  • Liu M, Li Y, Zhang C, Zhang Q. Role of Aurora kinase B in regulating resistance to paclitaxel in breast cancer cells. Hum Cell. 2022;35(2):678–693. doi:10.1007/s13577-022-00675-8
  • Ahmed A, Shamsi A, Mohammad T, Hasan GM, Islam A, Hassan MI. Aurora B kinase: a potential drug target for cancer therapy. J Cancer Res Clin Oncol. 2021;147(8):2187–2198. doi:10.1007/s00432-021-03669-5
  • Gully CP, Zhang F, Chen J, et al. Antineoplastic effects of an Aurora B kinase inhibitor in breast cancer. Mol Cancer. 2010;9(1):42. doi:10.1186/1476-4598-9-42
  • Falchook GS, Bastida CC, Kurzrock R. Aurora kinase inhibitors in oncology clinical trials: current state of the progress. Semin Oncol. 2015;42(6):832–848. doi:10.1053/j.seminoncol.2015.09.022
  • Zhang J, Lin X, Wu L, et al. Aurora B induces epithelial-mesenchymal transition by stabilizing Snail1 to promote basal-like breast cancer metastasis. Oncogene. 2020;39(12):2550–2567. doi:10.1038/s41388-020-1165-z
  • Niermann KJ, Moretti L, Giacalone NJ, et al. Enhanced radiosensitivity of androgen-resistant prostate cancer: AZD1152-mediated Aurora kinase B inhibition. Radiat Res. 2011;175(4):444–451. doi:10.1667/RR2317.1
  • Tao Y, Leteur C, Calderaro J, et al. The Aurora B kinase inhibitor AZD1152 sensitizes cancer cells to fractionated irradiation and induces mitotic catastrophe. Cell Cycle. 2009;8(19):3172–3181. doi:10.4161/cc.8.19.9729
  • Tao Y, Zhang P, Girdler F, et al. Enhancement of radiation response in p53-deficient cancer cells by the Aurora-B kinase inhibitor AZD1152. Oncogene. 2008;27(23):3244–3255. doi:10.1038/sj.onc.1210990
  • Jungles KM, Wang Z, Bishop C, et al. Targeting Aurora kinase B (AURKB) as a radiosensitizing strategy in syngeneic models of triple negative breast cancer (TNBC) (Abstract 708). Cancer Resear. 2024;84(6_Supplement):708. doi:10.1158/1538-7445.AM2024-708
  • Mason JM, Lin DC, Wei X, et al. Functional characterization of CFI-400945, a Polo-like kinase 4 inhibitor, as a potential anticancer agent. Cancer Cell. 2014;26(2):163–176. doi:10.1016/j.ccr.2014.05.006
  • Ianevski A, Giri AK, Aittokallio T. SynergyFinder 3.0: an interactive analysis and consensus interpretation of multi-drug synergies across multiple samples. Nucleic Acids Res. 2022;50(W1):W739–W743. doi:10.1093/nar/gkac382
  • Yang J, Ikezoe T, Nishioka C, et al. AZD1152, a novel and selective Aurora B kinase inhibitor, induces growth arrest, apoptosis, and sensitization for tubulin depolymerizing agent or topoisomerase II inhibitor in human acute leukemia cells in vitro and in vivo. Blood. 2007;110(6):2034–2040. doi:10.1182/blood-2007-02-073700
  • Borah NA, Reddy MM. Aurora kinase B inhibition: a potential therapeutic strategy for cancer. Molecules. 2021;26(7):1981. doi:10.3390/molecules26071981
  • Sak A, Stuschke M, Groneberg M, Kübler D, Pöttgen C, Eberhardt WE. Inhibiting the Aurora B kinase potently suppresses repopulation during fractionated irradiation of human lung cancer cell lines. Int J Radiat Oncol Biol Phys. 2012;84(2):492–499. doi:10.1016/j.ijrobp.2011.12.021
  • Marampon F, Gravina GL, Popov VM, et al. Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. Int J Oncol. 2014;44(1):285–294. doi:10.3892/ijo.2013.2167
  • Schwartz GK, Carvajal RD, Midgley R, et al. Phase I study of barasertib (AZD1152), a selective inhibitor of Aurora B kinase, in patients with advanced solid tumors. Invest New Drugs. 2013;31(2):370–380. doi:10.1007/s10637-012-9825-7
  • Cheung CH, Sarvagalla S, Lee JY, Huang YC, Coumar MS. Aurora kinase inhibitor patents and agents in clinical testing: an update (2011–2013). Expert Opin Ther Pat. 2014;24(9):1021–1038. doi:10.1517/13543776.2014.931374
  • Burris HA, Wang JS-Z, Johnson ML, et al. A Phase I, open-label, first-time-in-patient dose escalation and expansion study to assess the safety, tolerability, and pharmacokinetics of nanoparticle encapsulated Aurora B kinase inhibitor AZD2811 in patients with advanced solid tumours. J clin oncol. 2017;35(15_suppl):TPS2608. doi:10.1200/JCO.2017.35.15_suppl.TPS2608
  • Loap P, Loirat D, Berger F, et al. Combination of olaparib and radiation therapy for triple negative breast cancer: preliminary results of the RADIOPARP Phase 1 trial. Int J Radiat Oncol Biol Phys. 2021;109(2):436–440. doi:10.1016/j.ijrobp.2020.09.032
  • Loap P, Loirat D, Berger F, et al. Concurrent olaparib and radiotherapy in patients with triple-negative breast cancer: the Phase 1 olaparib and radiation therapy for triple-negative breast cancer trial. JAMA Oncol. 2022;8(12):1802–1808. doi:10.1001/jamaoncol.2022.5074
  • Ho AY, Barker CA, Arnold BB, et al. A Phase 2 clinical trial assessing the efficacy and safety of pembrolizumab and radiotherapy in patients with metastatic triple-negative breast cancer. Cancer. 2020;126(4):850–860. doi:10.1002/cncr.32599
  • Bavetsias V, Linardopoulos S. Aurora Kinase Inhibitors: current Status and Outlook. Front Oncol. 2015;5:278. doi:10.3389/fonc.2015.00278
  • Schöffski P, Jones SF, Dumez H, et al. Phase I, open-label, multicentre, dose-escalation, pharmacokinetic and pharmacodynamic trial of the oral Aurora kinase inhibitor PF-03814735 in advanced solid tumours. Eur J Cancer. 2011;47(15):2256–2264. doi:10.1016/j.ejca.2011.07.008
  • Schöffski P, Besse B, Gauler T, et al. Efficacy and safety of biweekly i.v. administrations of the Aurora kinase inhibitor danusertib hydrochloride in independent cohorts of patients with advanced or metastatic breast, ovarian, colorectal, pancreatic, small-cell and non-small-cell lung cancer: a multi-tumour, multi-institutional Phase II study. Ann Oncol. 2015;26(3):598–607. doi:10.1093/annonc/mdu566
  • Carducci M, Shaheen M, Markman B, et al. A phase 1, first-in-human study of AMG 900, an orally administered pan-Aurora kinase inhibitor, in adult patients with advanced solid tumors. Invest New Drugs. 2018;36(6):1060–1071. doi:10.1007/s10637-018-0625-6
  • Floc’h N, Ashton S, Taylor P, et al. Optimizing therapeutic effect of aurora B inhibition in acute myeloid leukemia with AZD2811 nanoparticles. Mol Cancer Ther. 2017;16(6):1031–1040. doi:10.1158/1535-7163.MCT-16-0580
  • Floc’h N, Ashton S, Ferguson D, et al. Modeling dose and schedule effects of AZD2811 nanoparticles targeting aurora B kinase for treatment of diffuse large B-cell lymphoma. Mol Cancer Ther. 2019;18(5):909–919. doi:10.1158/1535-7163.MCT-18-0577
  • Ashton S, Song YH, Nolan J, et al. Aurora kinase inhibitor nanoparticles target tumors with favorable therapeutic index in vivo. Sci Transl Med. 2016;8(325):325ra317. doi:10.1126/scitranslmed.aad2355