390
Views
15
CrossRef citations to date
0
Altmetric
Review

Issues in interpreting the in vivo activity of Aurora-A

, &

Bibliography

  • Weinstein IB. Cancer. Addiction to oncogenes – the Achilles heal of cancer. Science 2002;297:63-4
  • Matallanas D, Birtwistle M, Romano D, et al. Raf family kinases: old dogs have learned new tricks. Genes Cancer 2011;2:232-60
  • Meraldi P, Honda R, Nigg EA. Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53-/- cells. EMBO J 2002;21:483-92
  • Anand S, Penrhyn-Lowe S, Venkitaraman AR. AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. Cancer Cell 2003;3:51-62
  • Vader G, Lens SM. The Aurora kinase family in cell division and cancer. Biochim Biophys Acta 2008;1786:60-72
  • Zhang D, Hirota T, Marumoto T, et al. Cre-loxP-controlled periodic Aurora-A overexpression induces mitotic abnormalities and hyperplasia in mammary glands of mouse models. Oncogene 2004;23:8720-30
  • Tatsuka M, Katayama H, Ota T, et al. Multinuclearity and increased ploidy caused by overexpression of the aurora- and Ipl1-like midbody-associated protein mitotic kinase in human cancer cells. Cancer Res 1998;58:4811-16
  • Nikonova AS, Astsaturov I, Serebriiskii IG, et al. Aurora A kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci 2013;70:661-87
  • Agnese V, Bazan V, Fiorentino FP, et al. The role of Aurora-A inhibitors in cancer therapy. Ann Oncol 2007;18(Suppl 6):47-52
  • Nadler Y, Camp RL, Schwartz C, et al. Expression of Aurora A (but not Aurora B) is predictive of survival in breast cancer. Clin Cancer Res 2008;14:4455-62
  • Ratushny V, Pathak HB, Beeharry N, et al. Dual inhibition of SRC and Aurora kinases induces postmitotic attachment defects and cell death. Oncogene 2012;31:1217-27
  • Otto T, Horn S, Brockmann M, et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 2009;15:67-78
  • Liu Q, Kaneko S, Yang L, et al. Aurora-A abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem 2004;279:52175-82
  • Wu CC, Yang TY, Yu CT, et al. p53 negatively regulates Aurora A via both transcriptional and posttranslational regulation. Cell Cycle 2012;11:3433-42
  • de Carcer G, Malumbres M. A centrosomal route for cancer genome instability. Nat Cell Biol 2014;16:504-6
  • Friedberg JW, Mahadevan D, Cebula E, et al. Phase II study of alisertib, a selective Aurora A kinase inhibitor, in relapsed and refractory aggressive B- and T-cell non-Hodgkin lymphomas. J Clin Oncol 2014;32:44-50
  • Matulonis UA, Sharma S, Ghamande S, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol 2012;127:63-9
  • Abe Y, Ohsugi M, Haraguchi K, et al. LATS2-Ajuba complex regulates gamma-tubulin recruitment to centrosomes and spindle organization during mitosis. FEBS Lett 2006;580:782-8
  • Mori D, Yano Y, Toyo-oka K, et al. NDEL1 phosphorylation by Aurora-A kinase is essential for centrosomal maturation, separation, and TACC3 recruitment. Mol Cell Biol 2007;27:352-67
  • Nurse P. Universal control mechanism regulating onset of M-phase. Nature 1990;344:503-8
  • Dutertre S, Cazales M, Quaranta M, et al. Phosphorylation of CDC25B by Aurora-A at the centrosome contributes to the G2-M transition. J Cell Sci 2004;117:2523-31
  • Nam HJ, van Deursen JM. Cyclin B2 and p53 control proper timing of centrosome separation. Nat Cell Biol 2014;16:538-49
  • Ouchi M, Fujiuchi N, Sasai K, et al. BRCA1 phosphorylation by Aurora-A in the regulation of G2 to M transition. J Biol Chem 2004;279:19643-8
  • Kashatus DF, Lim KH, Brady DC, et al. RALA and RALBP1 regulate mitochondrial fission at mitosis. Nat Cell Biol 2011;13:1108-15
  • Floyd S, Pines J, Lindon C. APC/C Cdh1 targets aurora kinase to control reorganization of the mitotic spindle at anaphase. Curr Biol 2008;18:1649-58
  • Perez de Castro I, Aguirre-Portoles C, Martin B, et al. A SUMOylation Motif in Aurora-A: implications for Spindle Dynamics and Oncogenesis. Front Oncol 2011;1:50
  • Littlepage LE, Wu H, Andresson T, et al. Identification of phosphorylated residues that affect the activity of the mitotic kinase Aurora-A. Proc Natl Acad Sci USA 2002;99:15440-5
  • Mori D, Yamada M, Mimori-Kiyosue Y, et al. An essential role of the aPKC-Aurora A-NDEL1 pathway in neurite elongation by modulation of microtubule dynamics. Nat Cell Biol 2009;11:1057-68
  • Diaz B, Barnard D, Filson A, et al. Phosphorylation of Raf-1 serine 338-serine 339 is an essential regulatory event for Ras-dependent activation and biological signaling. Mol Cell Biol 1997;17:4509-16
  • Rowan FC, Richards M, Bibby RA, et al. Insights into aurora-A kinase activation using unnatural amino acids incorporated by chemical modification. ACS Chem Biol 2013;8:2184-91
  • Dodson CA, Kosmopoulou M, Richards MW, et al. Crystal structure of an Aurora-A mutant that mimics Aurora-B bound to MLN8054: insights into selectivity and drug design. Biochem J 2010;427:19-28
  • Zorba A, Buosi V, Kutter S, et al. Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2. Elife 2014;3:e02667
  • Bayliss R, Sardon T, Vernos I, Conti E. Structural basis of Aurora-A activation by TPX2 at the mitotic spindle. Mol Cell 2003;12:851-62
  • Zhao B, Smallwood A, Yang J, et al. Modulation of kinase-inhibitor interactions by auxiliary protein binding: crystallography studies on Aurora A interactions with VX-680 and with TPX2. Protein Sci 2008;17:1791-7
  • Clark MA, Acharya RA, Arico-Muendel CC, et al. Design, synthesis and selection of DNA-encoded small-molecule libraries. Nat Chem Biol 2009;5:647-54
  • Bibby RA, Tang C, Faisal A, et al. A cancer-associated aurora A mutant is mislocalized and misregulated due to loss of interaction with TPX2. J Biol Chem 2009;284:33177-84
  • Eyers PA, Erikson E, Chen LG, Maller JL. A novel mechanism for activation of the protein kinase Aurora A. Curr Biol 2003;13:691-7
  • Tsai MY, Wiese C, Cao K, et al. A Ran signalling pathway mediated by the mitotic kinase Aurora A in spindle assembly. Nat Cell Biol 2003;5:242-8
  • Eyers PA, Maller JL. Regulation of xenopus aurora A activation by TPX2. J Biol Chem 2004;279:9008-15
  • Giubettini M, Asteriti IA, Scrofani J, et al. Control of Aurora-A stability through interaction with TPX2. J Cell Sci 2011;124:113-22
  • Kufer TA, Sillje HH, Korner R, et al. Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol 2002;158:617-23
  • Dodson CA, Bayliss R. Activation of Aurora-A kinase by protein partner binding and phosphorylation are independent and synergistic. J Biol Chem 2012;287:1150-7
  • Xu X, Wang X, Xiao Z, et al. Two TPX2-dependent switches control the activity of Aurora A. PLoS One 2011;6:e16757
  • Product data sheet purified anti-aurora A (aurora 2)-phosphorylated (Thr288) antibody. Available from: http://www.biolegend.com/purified-anti-aurora-a-aurora-2-phosphorylated-thr288-antibody-2176.html 2014. [Last accessed August 2014]
  • Plotnikova OV, Nikonova AS, Loskutov YV, et al. Calmodulin activation of Aurora-A kinase (AURKA) is required during ciliary disassembly and in mitosis. Mol Biol Cell 2012;23:2658-70
  • Plotnikova OV, Pugacheva EN, Dunbrack RL, Golemis EA. Rapid calcium-dependent activation of Aurora-A kinase. Nat Commun 2010;1:64
  • Reboutier D, Troadec MB, Cremet JY, et al. Nucleophosmin/B23 activates Aurora A at the centrosome through phosphorylation of serine 89. J Cell Biol 2012;197:19-26
  • Haydon CE, Eyers PA, Aveline-Wolf LD, et al. Identification of novel phosphorylation sites on Xenopus laevis Aurora A and analysis of phosphopeptide enrichment by immobilized metal-affinity chromatography. Mol Cell Proteomics 2003;2:1055-67
  • Pascreau G, Delcros JG, Morin N, et al. Aurora-A kinase Ser349 phosphorylation is required during Xenopus laevis oocyte maturation. Dev Biol 2008;317:523-30
  • Lawrence HR, Martin MP, Luo Y, et al. Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J Med Chem 2012;55:7392-416
  • Sarkissian M, Mendez R, Richter JD. Progesterone and insulin stimulation of CPEB-dependent polyadenylation is regulated by Aurora A and glycogen synthase kinase-3. Genes Dev 2004;18:48-61
  • Krystyniak A, Garcia-Echeverria C, Prigent C, Ferrari S. Inhibition of Aurora A in response to DNA damage. Oncogene 2006;25:338-48
  • Littlepage LE, Ruderman JV. Identification of a new APC/C recognition domain, the A box, which is required for the Cdh1-dependent destruction of the kinase Aurora-A during mitotic exit. Genes Dev 2002;16:2274-85
  • Pugacheva EN, Golemis EA. The focal adhesion scaffolding protein HEF1 regulates activation of the Aurora-A and Nek2 kinases at the centrosome. Nat Cell Biol 2005;7:937-46
  • Zhao ZS, Lim JP, Ng YW, et al. The GIT-associated kinase PAK targets to the centrosome and regulates Aurora-A. Mol Cell 2005;20:237-49
  • Hirota T, Kunitoku N, Sasayama T, et al. Aurora-A and an interacting activator, the LIM protein Ajuba, are required for mitotic commitment in human cells. Cell 2003;114:585-98
  • Satinover DL, Leach CA, Stukenberg PT, Brautigan DL. Activation of Aurora-A kinase by protein phosphatase inhibitor-2, a bifunctional signaling protein. Proc Natl Acad Sci USA 2004;101:8625-30
  • Molli PR, Li DQ, Bagheri-Yarmand R, et al. Arpc1b, a centrosomal protein, is both an activator and substrate of Aurora A. J Cell Biol 2010;190:101-14
  • Hutterer A, Berdnik D, Wirtz-Peitz F, et al. Mitotic activation of the kinase Aurora-A requires its binding partner Bora. Dev Cell 2006;11:147-57
  • Pascreau G, Delcros JG, Cremet JY, et al. Phosphorylation of maskin by Aurora-A participates in the control of sequential protein synthesis during Xenopus laevis oocyte maturation. J Biol Chem 2005;280:13415-23
  • Sloane DA, Trikic MZ, Chu ML, et al. Drug-resistant aurora A mutants for cellular target validation of the small molecule kinase inhibitors MLN8054 and MLN8237. ACS Chem Biol 2010;5:563-76
  • Joukov V, De Nicolo A, Rodriguez A, et al. Centrosomal protein of 192 kDa (Cep192) promotes centrosome-driven spindle assembly by engaging in organelle-specific Aurora A activation. Proc Natl Acad Sci USA 2010;107:21022-7
  • Nikonova AS, Gaponova AV, Kudinov AE, Golemis EA. CAS proteins in health and disease: An update. IUBMB Life 2014;66(6):387-95
  • Dadke D, Jarnik M, Pugacheva EN, et al. Deregulation of HEF1 impairs M-phase progression by disrupting the RhoA activation cycle. Mol Biol Cell 2006;17:1204-17
  • Ice RJ, McLaughlin SL, Livengood RH, et al. NEDD9 depletion destabilizes Aurora A kinase and heightens the efficacy of Aurora A inhibitors: implications for treatment of metastatic solid tumors. Cancer Res 2013;73:3168-80
  • Sabino D, Brown NH, Basto R. Drosophila Ajuba is not an Aurora-A activator but is required to maintain Aurora-A at the centrosome. J Cell Sci 2011;124:1156-66
  • Bai M, Ni J, Shen S, et al. Aurora-A kinase-inactive mutants disrupt the interaction with Ajuba and cause defects in mitotic spindle formation and G2/M phase arrest in HeLa cells. BMB Rep 2014; pii:2575. [Epub ahead of print]
  • Bai M, Ni J, Shen S, et al. Two newly identified sites in the N-terminal regulatory domain of Aurora-A are essential for auto-inhibition. Biotechnol Lett 2014;36:1595-604
  • Bai M, Ni J, Wu J, et al. A novel mechanism for activation of Aurora-A kinase by Ajuba. Gene 2014;543:133-9
  • Ritchey L, Ottman R, Roumanos M, Chakrabarti R. A functional cooperativity between Aurora A kinase and LIM kinase1: implication in the mitotic process. Cell Cycle 2012;11:296-309
  • Seki A, Coppinger JA, Jang CY, et al. Bora and the kinase Aurora a cooperatively activate the kinase Plk1 and control mitotic entry. Science 2008;320:1655-8
  • Radu M, Semenova G, Kosoff R, Chernoff J. PAK signalling during the development and progression of cancer. Nat Rev Cancer 2014;14:13-25
  • Katayama H, Zhou H, Li Q, et al. Interaction and feedback regulation between STK15/BTAK/Aurora-A kinase and protein phosphatase 1 through mitotic cell division cycle. J Biol Chem 2001;276:46219-24
  • Zeng K, Bastos RN, Barr FA, Gruneberg U. Protein phosphatase 6 regulates mitotic spindle formation by controlling the T-loop phosphorylation state of Aurora A bound to its activator TPX2. J Cell Biol 2010;191:1315-32
  • Chen SS, Chang PC, Cheng YW, et al. Suppression of the STK15 oncogenic activity requires a transactivation-independent p53 function. EMBO J 2002;21:4491-9
  • Shao S, Wang Y, Jin S, et al. Gadd45a interacts with aurora-A and inhibits its kinase activity. J Biol Chem 2006;281:28943-50
  • Castro A, Arlot-Bonnemains Y, Vigneron S, et al. APC/Fizzy-Related targets Aurora-A kinase for proteolysis. EMBO Rep 2002;3:457-62
  • Crane R, Kloepfer A, Ruderman JV. Requirements for the destruction of human Aurora-A. J Cell Sci 2004;117:5975-83
  • Yu X, Minter-Dykhouse K, Malureanu L, et al. Chfr is required for tumor suppression and Aurora A regulation. Nat Genet 2005;37:401-6
  • Shi Y, Solomon LR, Pereda-Lopez A, et al. Ubiquitin-specific cysteine protease 2a (USP2a) regulates the stability of Aurora-A. J Biol Chem 2011;286:38960-8
  • Huang YH, Wu CC, Chou CK, Huang CY. A translational regulator, PUM2, promotes both protein stability and kinase activity of Aurora-A. PLoS One 2011;6:e19718
  • Johnson EO, Chang KH, Ghosh S, et al. LIMK2 is a crucial regulator and effector of Aurora-A-kinase-mediated malignancy. J Cell Sci 2012;125:1204-16
  • Johnson EO, Chang KH, de Pablo Y, et al. PHLDA1 is a crucial negative regulator and effector of Aurora A kinase in breast cancer. J Cell Sci 2011;124:2711-22
  • Zheng M, McKeown-Longo PJ. Cell adhesion regulates Ser/Thr phosphorylation and proteasomal degradation of HEF1. J Cell Sci 2006;119:96-103
  • Chowdhury A, Chowdhury S, Tsai MY. A novel Aurora kinase A inhibitor MK-8745 predicts TPX2 as a therapeutic biomarker in non-Hodgkin lymphoma cell lines. Leuk Lymphoma 2012;53:462-71
  • Pugacheva EN, Jablonski SA, Hartman TR, et al. HEF1-dependent Aurora A activation induces disassembly of the primary cilium. Cell 2007;129:1351-63
  • Plotnikova OV, Pugacheva EN, Golemis EA. Aurora A kinase activity influences calcium signaling in kidney cells. J Cell Biol 2011;193:1021-32
  • Yuan K, Frolova N, Xie Y, et al. Primary cilia are decreased in breast cancer: analysis of a collection of human breast cancer cell lines and tissues. J Histochem Cytochem 2010;58:857-70
  • Han YG, Kim HJ, Dlugosz AA, et al. Dual and opposing roles of primary cilia in medulloblastoma development. Nat Med 2009;15:1062-5
  • Schneider L, Clement CA, Teilmann SC, et al. PDGFRalphaalpha signaling is regulated through the primary cilium in fibroblasts. Curr Biol 2005;15:1861-6
  • Seeger-Nukpezah T, Little JL, Serzhanova V, Golemis EA. Cilia and cilia-associated proteins in cancer. Drug Discov Today Dis Mech 2013;10:e135-e42
  • Plotnikova OV, Golemis EA, Pugacheva EN. Cell cycle-dependent ciliogenesis and cancer. Cancer Res 2008;68:2058-61
  • Jackman M, Lindon C, Nigg EA, Pines J. Active cyclin B1-Cdk1 first appears on centrosomes in prophase. Nat Cell Biol 2003;5:143-8
  • Yamada M, Hirotsune S, Wynshaw-Boris A. The essential role of LIS1, NDEL1 and Aurora-A in polarity formation and microtubule organization during neurogensis. Cell Adh Migr 2010;4:180-4
  • Lefkowitz GK, Gleeson JG. Aurora A moonlights in neurite extension. Nat Cell Biol 2009;11:1053-4
  • Manfredi MG, Ecsedy JA, Chakravarty A, et al. Characterization of Alisertib (MLN8237), an investigational small-molecule inhibitor of aurora A kinase using novel in vivo pharmacodynamic assays. Clin Cancer Res 2011;17:7614-24
  • Do TV, Xiao F, Bickel LE, et al. Aurora kinase A mediates epithelial ovarian cancer cell migration and adhesion. Oncogene 2014;33:539-49
  • Zhou N, Singh K, Mir MC, et al. The investigational Aurora kinase A inhibitor MLN8237 induces defects in cell viability and cell-cycle progression in malignant bladder cancer cells in vitro and in vivo. Clin Cancer Res 2013;19:1717-28
  • Sehdev V, Katsha A, Ecsedy J, et al. The combination of alisertib, an investigational Aurora kinase A inhibitor, and docetaxel promotes cell death and reduces tumor growth in preclinical cell models of upper gastrointestinal adenocarcinomas. Cancer 2013;119:904-14
  • Sehdev V, Peng D, Soutto M, et al. The aurora kinase A inhibitor MLN8237 enhances cisplatin-induced cell death in esophageal adenocarcinoma cells. Mol Cancer Ther 2012;11:763-74
  • Brewer Savannah KJ, Demicco EG, Lusby K, et al. Dual targeting of mTOR and aurora-A kinase for the treatment of uterine Leiomyosarcoma. Clin Cancer Res 2012;18:4633-45
  • Carol H, Boehm I, Reynolds CP, et al. Efficacy and pharmacokinetic/pharmacodynamic evaluation of the Aurora kinase A inhibitor MLN8237 against preclinical models of pediatric cancer. Cancer Chemother Pharmacol 2011;68:1291-304
  • Patel AV, Eaves D, Jessen WJ, et al. Ras-driven transcriptome analysis identifies aurora kinase A as a potential malignant peripheral nerve sheath tumor therapeutic target. Clin Cancer Res 2012;18:5020-30
  • Muscal JA, Scorsone KA, Zhang L, et al. Additive effects of vorinostat and MLN8237 in pediatric leukemia, medulloblastoma, and neuroblastoma cell lines. Invest New Drugs 2013;31:39-45
  • Gorgun G, Calabrese E, Hideshima T, et al. A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma. Blood 2010;115:5202-13
  • Mahadevan D, Stejskal A, Cooke LS, et al. Aurora A inhibitor (MLN8237) plus vincristine plus rituximab is synthetic lethal and a potential curative therapy in aggressive B-cell non-Hodgkin lymphoma. Clin Cancer Res 2012;18:2210-19
  • Qi W, Spier C, Liu X, et al. Alisertib (MLN8237) an investigational agent suppresses Aurora A and B activity, inhibits proliferation, promotes endo-reduplication and induces apoptosis in T-NHL cell lines supporting its importance in PTCL treatment. Leuk Res 2013;37:434-9
  • Qi W, Cooke LS, Liu X, et al. Aurora inhibitor MLN8237 in combination with docetaxel enhances apoptosis and anti-tumor activity in mantle cell lymphoma. Biochem Pharmacol 2011;81:881-90
  • Tomita M, Mori N. Aurora A selective inhibitor MLN8237 suppresses the growth and survival of HTLV-1-infected T-cells in vitro. Cancer Sci 2010;101:1204-11
  • Kelly KR, Ecsedy J, Medina E, et al. The novel Aurora A kinase inhibitor MLN8237 is active in resistant chronic myeloid leukaemia and significantly increases the efficacy of nilotinib. J Cell Mol Med 2011;15:2057-70
  • Infante J DE, Cohen RB, et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of MLN8237, a selective aurora A kinase inhibitor, in the United States [abstract 280]. Eur J Cancer Suppl 2008;6:90
  • Dees EC, Cohen RB, von Mehren M, et al. Phase I study of aurora A kinase inhibitor MLN8237 in advanced solid tumors: safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations. Clin Cancer Res 2012;18:4775-84
  • Cervantes A, Elez E, Roda D, et al. Phase I pharmacokinetic/pharmacodynamic study of MLN8237, an investigational, oral, selective aurora a kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2012;18:4764-74
  • Cervantes-Ruiperez A, Burris HA, Cohen RB, et al. Pharmacokinetic and pharmacodynamic results from two phase I studies of the investigational selective aurora A kinase (AAK) inhibitor MLN8237: Exposure-dependent AAK inhibition in human tumors. J Clin Oncol 2010;28(Suppl):abstract 3031
  • Kelly KR, Shea TC, Goy A, et al. Phase I study of MLN8237-investigational Aurora A kinase inhibitor-in relapsed/refractory multiple myeloma, Non-Hodgkin lymphoma and chronic lymphocytic leukemia. Invest New Drugs 2014;32(3):489-99
  • Brockmann M, Poon E, Berry T, et al. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell 2013;24:75-89
  • Zhang M HJ, Sells T, et al. In vivo characterization of the aurora A kinase inhibitor MLN8237 in subcutaneous and disseminated models of human cancer. Proc Am Assoc Cancer Res 2009;49:abstract 5646
  • Aliagas-Martin I, Burdick D, Corson L, et al. A class of 2,4-bisanilinopyrimidine Aurora A inhibitors with unusually high selectivity against Aurora B. J Med Chem 2009;52:3300-7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.