Skip to main content
Top
Published in: Cancer Cell International 1/2020

01-12-2020 | Neuroblastoma | Primary research

Silencing of AURKA augments the antitumor efficacy of the AURKA inhibitor MLN8237 on neuroblastoma cells

Authors: Yan Yang, Lili Ding, Qi Zhou, Li Fen, Yuhua Cao, Junjie Sun, Xuefeng Zhou, Aiguo Liu

Published in: Cancer Cell International | Issue 1/2020

Login to get access

Abstract

Background

Aurora kinase A (AURKA) has been implicated in the regulation of cell cycle progression, mitosis and a key number of oncogenic signaling pathways in various malignancies including neuroblastoma. Small molecule inhibitors of AURKA have shown potential, but still not as good as expected effects in clinical trials. Little is known about this underlying mechanism. Here, we evaluated the inhibitory effects of AURKA inhibitor MLN8237 on neuroblastoma cells to understand the potential mechanisms responsible for tumor therapy.

Methods

MLN8237 treatment on neuroblastoma cell line IMR32 was done and in vivo inhibitory effects were investigated using tumor xenograft model. Cellular senescence was evaluated by senescence-associated β-gal Staining assay. Flow cytometry was used to tested cell cycle arrest and cell apoptosis. Senescence-associated signal pathways were detected by western blot. CD133 microbeads and microsphere formation were used to separate and enrich CD133+ cells. AURKA small interfering RNA transfection was carried to downregulate AURKA level. Finally, the combination of MLN8237 treatment with AURKA small interfering RNA transfection were adopted to evaluate the inhibitory effect on neuroblastoma cells.

Results

We demonstrate that MLN8237, an inhibitor of AURKA, induces the neuroblastoma cell line IMR32 into cellular senescence and G2/M cell phase arrest. Inactivation of AURKA results in MYCN destabilization and inhibits cell growth in vitro and in a mouse model. Although MLN8237 inhibits AURKA kinase activity, it has almost no inhibitory effect on the AURKA protein level. By contrast, MLN8237 treatment leads to abnormal high expression of AURKA in vitro and in vivo. Knockdown of AURKA reduces cell survival. The combination of MLN8237 with AURKA small interfering RNA results in more profound inhibitory effects on neuroblastoma cell growth. Moreover, MLN8237 treatment followed by AURKA siRNA forces senescent cells into apoptosis via suppression of the Akt/Stat3 pathway.

Conclusions

The effect of AURKA-targeted inhibition of tumor growth plays roles in both the inactivation of AURKA activity and the decrease in the AURKA protein expression level.
Literature
1.
go back to reference Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010;362(23):2202–11.CrossRef Maris JM. Recent advances in neuroblastoma. N Engl J Med. 2010;362(23):2202–11.CrossRef
2.
go back to reference Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med. 2015;66:49–63.CrossRef Louis CU, Shohet JM. Neuroblastoma: molecular pathogenesis and therapy. Annu Rev Med. 2015;66:49–63.CrossRef
3.
go back to reference Dang CV, Reddy EP, Shokat KM, Soucek L. Drugging the ‘undruggable’ cancer targets. Nat Rev Cancer. 2017;17(8):502–8.CrossRef Dang CV, Reddy EP, Shokat KM, Soucek L. Drugging the ‘undruggable’ cancer targets. Nat Rev Cancer. 2017;17(8):502–8.CrossRef
4.
go back to reference Dutertre S, Descamps S, Prigent C. On the role of aurora-A in centrosome function. Oncogene. 2002;21(40):6175–83.CrossRef Dutertre S, Descamps S, Prigent C. On the role of aurora-A in centrosome function. Oncogene. 2002;21(40):6175–83.CrossRef
5.
go back to reference Marumoto T, Zhang D, Saya H. Aurora-A—a guardian of poles. Nat Rev Cancer. 2005;5(1):42–50.CrossRef Marumoto T, Zhang D, Saya H. Aurora-A—a guardian of poles. Nat Rev Cancer. 2005;5(1):42–50.CrossRef
6.
go back to reference Shang X, Burlingame SM, Okcu MF, Ge N, Russell HV, Egler RA, David RD, Vasudevan SA, Yang J, Nuchtern JG. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol Cancer Ther. 2009;8(8):2461–9.CrossRef Shang X, Burlingame SM, Okcu MF, Ge N, Russell HV, Egler RA, David RD, Vasudevan SA, Yang J, Nuchtern JG. Aurora A is a negative prognostic factor and a new therapeutic target in human neuroblastoma. Mol Cancer Ther. 2009;8(8):2461–9.CrossRef
7.
go back to reference Otto T, Horn S, Brockmann M, Eilers U, Schuttrumpf L, Popov N, Kenney AM, Schulte JH, Beijersbergen R, Christiansen H, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell. 2009;15(1):67–78.CrossRef Otto T, Horn S, Brockmann M, Eilers U, Schuttrumpf L, Popov N, Kenney AM, Schulte JH, Beijersbergen R, Christiansen H, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell. 2009;15(1):67–78.CrossRef
8.
go back to reference Sells TB, Chau R, Ecsedy JA, Gershman RE, Hoar K, Huck J, Janowick DA, Kadambi VJ, LeRoy PJ, Stirling M, et al. MLN8054 and Alisertib (MLN8237): discovery of selective oral aurora A inhibitors. ACS Med Chem Lett. 2015;6(6):630–4.CrossRef Sells TB, Chau R, Ecsedy JA, Gershman RE, Hoar K, Huck J, Janowick DA, Kadambi VJ, LeRoy PJ, Stirling M, et al. MLN8054 and Alisertib (MLN8237): discovery of selective oral aurora A inhibitors. ACS Med Chem Lett. 2015;6(6):630–4.CrossRef
9.
go back to reference Multicenter First. Randomized phase 3 study in patients (Pts) with relapsed/refractory (R/R) peripheral T-cell lymphoma (PTCL): alisertib (MLN8237) versus investigator’s choice (LUMIERE trial; NCT01482962). Clin Adv Hematol Oncol. 2016;14(2 Suppl 1):12–3. Multicenter First. Randomized phase 3 study in patients (Pts) with relapsed/refractory (R/R) peripheral T-cell lymphoma (PTCL): alisertib (MLN8237) versus investigator’s choice (LUMIERE trial; NCT01482962). Clin Adv Hematol Oncol. 2016;14(2 Suppl 1):12–3.
10.
go back to reference Lin J, Patel SA, Sama AR, Hoffman-Censits JH, Kennedy B, Kilpatrick D, Ye Z, Yang H, Mu Z, Leiby B, et al. A phase I/II study of the investigational drug alisertib in combination with abiraterone and prednisone for patients with metastatic castration-resistant prostate cancer progressing on abiraterone. Oncologist. 2016;21(11):1296–1297e.CrossRef Lin J, Patel SA, Sama AR, Hoffman-Censits JH, Kennedy B, Kilpatrick D, Ye Z, Yang H, Mu Z, Leiby B, et al. A phase I/II study of the investigational drug alisertib in combination with abiraterone and prednisone for patients with metastatic castration-resistant prostate cancer progressing on abiraterone. Oncologist. 2016;21(11):1296–1297e.CrossRef
11.
go back to reference Bringold F, Serrano M. Tumor suppressors and oncogenes in cellular senescence. Exp Gerontol. 2000;35(3):317–29.CrossRef Bringold F, Serrano M. Tumor suppressors and oncogenes in cellular senescence. Exp Gerontol. 2000;35(3):317–29.CrossRef
12.
go back to reference Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochem Biophys Acta. 1998;1378(2):F115–77.PubMed Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochem Biophys Acta. 1998;1378(2):F115–77.PubMed
13.
go back to reference Kaelin WG Jr. Functions of the retinoblastoma protein. BioEssays. 1999;21(11):950–8.CrossRef Kaelin WG Jr. Functions of the retinoblastoma protein. BioEssays. 1999;21(11):950–8.CrossRef
14.
go back to reference Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci USA. 1999;96(8):4240–5.CrossRef Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci USA. 1999;96(8):4240–5.CrossRef
15.
go back to reference Tomoda K, Kubota Y, Kato J. Degradation of the cyclin-dependent-kinase inhibitor p27Kip1 is instigated by Jab1. Nature. 1999;398(6723):160–5.CrossRef Tomoda K, Kubota Y, Kato J. Degradation of the cyclin-dependent-kinase inhibitor p27Kip1 is instigated by Jab1. Nature. 1999;398(6723):160–5.CrossRef
16.
go back to reference Noda A, Ning Y, Venable SF, Pereira-Smith OM, Smith JR. Cloning of senescent cell-derived inhibitors of DNA synthesis using an expression screen. Exp Cell Res. 1994;211(1):90–8.CrossRef Noda A, Ning Y, Venable SF, Pereira-Smith OM, Smith JR. Cloning of senescent cell-derived inhibitors of DNA synthesis using an expression screen. Exp Cell Res. 1994;211(1):90–8.CrossRef
17.
go back to reference Tahara H, Sato E, Noda A, Ide T. Increase in expression level of p21sdi1/cip1/waf1 with increasing division age in both normal and SV40-transformed human fibroblasts. Oncogene. 1995;10(5):835–40.PubMed Tahara H, Sato E, Noda A, Ide T. Increase in expression level of p21sdi1/cip1/waf1 with increasing division age in both normal and SV40-transformed human fibroblasts. Oncogene. 1995;10(5):835–40.PubMed
18.
go back to reference Ramljak D, Calvert RJ, Wiesenfeld PW, Diwan BA, Catipovic B, Marasas WF, Victor TC, Anderson LM, Gelderblom WC. A potential mechanism for fumonisin B(1)-mediated hepatocarcinogenesis: cyclin D1 stabilization associated with activation of Akt and inhibition of GSK-3beta activity. Carcinogenesis. 2000;21(8):1537–46.PubMed Ramljak D, Calvert RJ, Wiesenfeld PW, Diwan BA, Catipovic B, Marasas WF, Victor TC, Anderson LM, Gelderblom WC. A potential mechanism for fumonisin B(1)-mediated hepatocarcinogenesis: cyclin D1 stabilization associated with activation of Akt and inhibition of GSK-3beta activity. Carcinogenesis. 2000;21(8):1537–46.PubMed
19.
go back to reference Kim M, Baek M, Kim DJ. Protein tyrosine signaling and its potential therapeutic implications in carcinogenesis. Curr Pharm Des. 2017;23(29):4226–46.PubMedPubMedCentral Kim M, Baek M, Kim DJ. Protein tyrosine signaling and its potential therapeutic implications in carcinogenesis. Curr Pharm Des. 2017;23(29):4226–46.PubMedPubMedCentral
20.
go back to reference Huang R, Cheung NK, Vider J, Cheung IY, Gerald WL, Tickoo SK, Holland EC, Blasberg RG. MYCN and MYC regulate tumor proliferation and tumorigenesis directly through BMI1 in human neuroblastomas. FASEB J. 2011;25(12):4138–49.CrossRef Huang R, Cheung NK, Vider J, Cheung IY, Gerald WL, Tickoo SK, Holland EC, Blasberg RG. MYCN and MYC regulate tumor proliferation and tumorigenesis directly through BMI1 in human neuroblastomas. FASEB J. 2011;25(12):4138–49.CrossRef
21.
go back to reference Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell. 2013;24(1):75–89.CrossRef Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell. 2013;24(1):75–89.CrossRef
22.
go back to reference He S, Sharpless NE. Senescence in health and disease. Cell. 2017;169(6):1000–11.CrossRef He S, Sharpless NE. Senescence in health and disease. Cell. 2017;169(6):1000–11.CrossRef
23.
go back to reference Sui X, Fang Y, Lou H, Wang K, Zheng Y, Lou F, Jin W, Xu Y, Chen W, Pan H, et al. p53 suppresses stress-induced cellular senescence via regulation of autophagy under the deprivation of serum. Mol Med Rep. 2015;11(2):1214–20.CrossRef Sui X, Fang Y, Lou H, Wang K, Zheng Y, Lou F, Jin W, Xu Y, Chen W, Pan H, et al. p53 suppresses stress-induced cellular senescence via regulation of autophagy under the deprivation of serum. Mol Med Rep. 2015;11(2):1214–20.CrossRef
24.
go back to reference Burton DG, Krizhanovsky V. Physiological and pathological consequences of cellular senescence. CMLS. 2014;71(22):4373–86.CrossRef Burton DG, Krizhanovsky V. Physiological and pathological consequences of cellular senescence. CMLS. 2014;71(22):4373–86.CrossRef
25.
go back to reference Katsha A, Arras J, Soutto M, Belkhiri A, El-Rifai W. AURKA regulates JAK2-STAT3 activity in human gastric and esophageal cancers. Mol Oncol. 2014;8(8):1419–28.CrossRef Katsha A, Arras J, Soutto M, Belkhiri A, El-Rifai W. AURKA regulates JAK2-STAT3 activity in human gastric and esophageal cancers. Mol Oncol. 2014;8(8):1419–28.CrossRef
26.
go back to reference Kamran M, Long ZJ, Xu D, Lv SS, Liu B, Wang CL, Xu J, Lam EW, Liu Q. Aurora kinase A regulates Survivin stability through targeting FBXL7 in gastric cancer drug resistance and prognosis. Oncogenesis. 2017;6(2):e298.CrossRef Kamran M, Long ZJ, Xu D, Lv SS, Liu B, Wang CL, Xu J, Lam EW, Liu Q. Aurora kinase A regulates Survivin stability through targeting FBXL7 in gastric cancer drug resistance and prognosis. Oncogenesis. 2017;6(2):e298.CrossRef
27.
go back to reference Zheng F, Yue C, Li G, He B, Cheng W, Wang X, Yan M, Long Z, Qiu W, Yuan Z, et al. Nuclear AURKA acquires kinase-independent transactivating function to enhance breast cancer stem cell phenotype. Nat Commun. 2016;7:10180.CrossRef Zheng F, Yue C, Li G, He B, Cheng W, Wang X, Yan M, Long Z, Qiu W, Yuan Z, et al. Nuclear AURKA acquires kinase-independent transactivating function to enhance breast cancer stem cell phenotype. Nat Commun. 2016;7:10180.CrossRef
Metadata
Title
Silencing of AURKA augments the antitumor efficacy of the AURKA inhibitor MLN8237 on neuroblastoma cells
Authors
Yan Yang
Lili Ding
Qi Zhou
Li Fen
Yuhua Cao
Junjie Sun
Xuefeng Zhou
Aiguo Liu
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2020
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-1072-y

Other articles of this Issue 1/2020

Cancer Cell International 1/2020 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