Emerging Role of Aurora A in Radioresistance: A Comprehensive Review

Radiotherapy is one of the most conventional modes of treatment in several cancers. Failure of radiotherapy followed by acquisition of radioresistance is one of the emerging challenges faced by clinical experts. Unusual expression and functional implications of several molecules are observed to facilitate radioresistance. Aurora A, a member of the Aurora kinase (serine/threonine kinase) family, is one such molecule that shows significantly altered expression as well as non-canonical functional crosstalk with other associated factors (cell cycle regulators, signaling molecules, stemness markers, etc.) to favour the adaptations for the acquirement of radioresistance. These mechanisms include progression of cell cycle, stimulatory activation of factors by phosphorylation for enhancing the chance of cellular survivability, and prevention of apoptosis. This review article summarises how Aurora A is responsible for radioresistance in cancer and why this kinase should be considered a negative biomarker of radiosensitivity. This review discloses a wider opportunity in the field of research to find the mechanistic key regulatory pathway of Aurora A, which can be a potential target for enhancing the efficiency of treatment. Further investigations are required to explore the potential of Aurora A inhibitors as reliable radiosensitisers.

[1]  Anna Cha,et al.  Cyclin-dependent Kinase 1 and Aurora Kinase choreograph mitotic storage and redistribution of a growth factor receptor , 2021, PLoS biology.

[2]  Arnoud Boot,et al.  Multiomic analysis and immunoprofiling reveal distinct subtypes of human angiosarcoma. , 2020, The Journal of clinical investigation.

[3]  P. Setny Conserved internal hydration motifs in protein kinases , 2020, Proteins.

[4]  James B. Mitchell,et al.  Radiation-induced Adaptive Response: New Potential for Cancer Treatment , 2020, Clinical Cancer Research.

[5]  M. Kavallaris,et al.  Interplay between MycN and c-Myc regulates radioresistance and cancer stem cell phenotype in neuroblastoma upon glutamine deprivation , 2020, Theranostics.

[6]  J. Chen,et al.  Long Noncoding RNA MALAT1 Contributes to Sorafenib Resistance by Targeting miR-140-5p/Aurora-A Signaling in Hepatocellular Carcinoma , 2020, Molecular Cancer Therapeutics.

[7]  Long-Bang Chen,et al.  Aurora-a confers radioresistance in human hepatocellular carcinoma by activating NF-κB signaling pathway , 2019, BMC Cancer.

[8]  R. Hoffman,et al.  Aurora-A/NF-ĸB Signaling Is Associated With Radio-resistance in Human Lung Adenocarcinoma , 2019, AntiCancer Research.

[9]  Ping Wang,et al.  Inhibition of Aurora A enhances radiosensitivity in selected lung cancer cell lines , 2019, Respiratory Research.

[10]  M. Dutreix,et al.  Altering DNA Repair to Improve Radiation Therapy: Specific and Multiple Pathway Targeting , 2019, Front. Oncol..

[11]  Y. Hosokawa,et al.  Understanding the mechanism underlying the acquisition of radioresistance in human prostate cancer cells. , 2019, Oncology letters.

[12]  L. Magnaghi-Jaulin,et al.  Aurora A Protein Kinase: To the Centrosome and Beyond , 2019, Biomolecules.

[13]  M. de Ridder,et al.  Hypoxic Radioresistance: Can ROS Be the Key to Overcome It? , 2019, Cancers.

[14]  Yan Chen,et al.  LncRNA TUG1 promotes cells proliferation and inhibits cells apoptosis through regulating AURKA in epithelial ovarian cancer cells , 2018, Medicine.

[15]  Hyuk Cho,et al.  Differential Selective Pressures Experienced by the Aurora Kinase Gene Family , 2017, International journal of molecular sciences.

[16]  R. Bronson,et al.  Cell cycle-targeting microRNAs promote differentiation by enforcing cell-cycle exit , 2017, Proceedings of the National Academy of Sciences.

[17]  T. Cech,et al.  How do lncRNAs regulate transcription? , 2017, Science Advances.

[18]  Yu-Chieh Su,et al.  Targeting the PI3K/AKT/mTOR signaling pathway as an effectively radiosensitizing strategy for treating human oral squamous cell carcinoma in vitro and in vivo , 2017, Oncotarget.

[19]  Guangyuan Song,et al.  AURKA promotes cancer metastasis by regulating epithelial-mesenchymal transition and cancer stem cell properties in hepatocellular carcinoma. , 2017, Biochemical and biophysical research communications.

[20]  Wenbo Li,et al.  Down-regulation of LncRNA TUG1 enhances radiosensitivity in bladder cancer via suppressing HMGB1 expression , 2017, Radiation oncology.

[21]  K. Shah,et al.  The Aurora-A–Twist1 axis promotes highly aggressive phenotypes in pancreatic carcinoma , 2017, Journal of Cell Science.

[22]  Ruozheng Wang,et al.  Aurora-A affects radiosenstivity in cervical squamous cell carcinoma and predicts poor prognosis , 2017, Oncotarget.

[23]  Masaru Ishii,et al.  Cell-cycle-controlled radiation therapy was effective for treating a murine malignant melanoma cell line in vitro and in vivo , 2016, Scientific Reports.

[24]  E. Sajo,et al.  The radiosensitizing effect of the aurora kinase inhibitors, ENMD-2076, on canine mast cell tumours in vitro. , 2016, Veterinary and comparative oncology.

[25]  B. Haffty,et al.  The relevance of molecular biomarkers in cervical cancer patients treated with radiotherapy. , 2015, Annals of translational medicine.

[26]  A. Musacchio,et al.  The Aurora B Kinase in Chromosome Bi-Orientation and Spindle Checkpoint Signaling , 2015, Front. Oncol..

[27]  K. Schindler,et al.  Functions of Aurora kinase C in meiosis and cancer , 2015, Front. Cell Dev. Biol..

[28]  J. Seong,et al.  Daurinol Enhances the Efficacy of Radiotherapy in Lung Cancer via Suppression of Aurora Kinase A/B Expression , 2015, Molecular Cancer Therapeutics.

[29]  B. Neumann,et al.  The Aurora-A inhibitor MLN8237 affects multiple mitotic processes and induces dose-dependent mitotic abnormalities and aneuploidy , 2014, Oncotarget.

[30]  Gong Yang,et al.  Corrigendum to “Aurora-A controls cancer cell radio- and chemoresistance via ATM/Chk2-mediated DNA repair networks” [Biochim. Biophys. Acta 1843 (2014) 934–944] , 2014 .

[31]  D. Chung,et al.  Targeting aurora kinase A inhibits hypoxia-mediated neuroblastoma cell tumorigenesis. , 2014, Anticancer research.

[32]  Gong Yang,et al.  Aurora-A controls cancer cell radio- and chemoresistance via ATM/Chk2-mediated DNA repair networks. , 2014, Biochimica et biophysica acta.

[33]  J. Crispino,et al.  The Aurora Kinases in Cell Cycle and Leukemia , 2014, Oncogene.

[34]  B. Teh,et al.  A novel panel of biomarkers predicts radioresistance in patients with squamous cell carcinoma of the head and neck. , 2014, European journal of cancer.

[35]  U. Mayor,et al.  Ubiquitination site preferences in anaphase promoting complex/cyclosome (APC/C) substrates , 2013, Open Biology.

[36]  W. De,et al.  The role of Aurora A in hypoxia-inducible factor 1α-promoting malignant phenotypes of hepatocelluar carcinoma , 2013, Cell cycle.

[37]  R. Bayliss,et al.  Insights into Aurora-A Kinase Activation Using Unnatural Amino Acids Incorporated by Chemical Modification , 2013, ACS chemical biology.

[38]  T. Kundu,et al.  Surface enhanced Raman spectroscopy of Aurora kinases: direct, ultrasensitive detection of autophosphorylation , 2013 .

[39]  L. Moretti,et al.  MLN8054, a small molecule inhibitor of aurora kinase a, sensitizes androgen-resistant prostate cancer to radiation. , 2011, International journal of radiation oncology, biology, physics.

[40]  K. Pollok,et al.  Preclinical activity of a novel multiple tyrosine kinase and aurora kinase inhibitor, ENMD‐2076, against multiple myeloma , 2010, British journal of haematology.

[41]  B. Lim,et al.  Aurora-A Contributes to Radioresistance by Increasing NF-&kgr;B DNA Binding , 2010, Radiation research.

[42]  D. Lane,et al.  Discovery of N-phenyl-4-(thiazol-5-yl)pyrimidin-2-amine aurora kinase inhibitors. , 2010, Journal of medicinal chemistry.

[43]  L. Pustilnik,et al.  PF-03814735, an Orally Bioavailable Small Molecule Aurora Kinase Inhibitor for Cancer Therapy , 2010, Molecular Cancer Therapeutics.

[44]  R. Fucini,et al.  SNS-314, a pan-Aurora kinase inhibitor, shows potent anti-tumor activity and dosing flexibility in vivo , 2010, Cancer Chemotherapy and Pharmacology.

[45]  Jeffrey Ecsedy,et al.  A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma. , 2009, Blood.

[46]  A. Wynshaw-Boris,et al.  An essential role of the aPKC–Aurora A–NDEL1 pathway in neurite elongation by modulation of microtubule dynamics , 2009, Nature Cell Biology.

[47]  Y. Toiyama,et al.  Correlation of CD133, OCT4, and SOX2 in Rectal Cancer and Their Association with Distant Recurrence After Chemoradiotherapy , 2009, Annals of Surgical Oncology.

[48]  Mladen Vinković,et al.  Fragment-based discovery of the pyrazol-4-yl urea (AT9283), a multitargeted kinase inhibitor with potent aurora kinase activity. , 2009, Journal of medicinal chemistry.

[49]  A. Dar,et al.  The Aurora kinase A Regulates GSK-3β in Gastric Cancer Cells , 2008, Oncogene.

[50]  S. Dimitrov,et al.  Localization of Aurora A and Aurora B kinases during interphase: Role of the N-Terminal domain , 2008, Cell cycle.

[51]  Paola Storici,et al.  PHA-739358, a potent inhibitor of Aurora kinases with a selective target inhibition profile relevant to cancer , 2007, Molecular Cancer Therapeutics.

[52]  J. Bourhis,et al.  Enhancement of radiation response by inhibition of Aurora-A kinase using siRNA or a selective Aurora kinase inhibitor PHA680632 in p53-deficient cancer cells , 2007, British Journal of Cancer.

[53]  Wenlin Huang,et al.  Aurora-A, a negative prognostic marker, increases migration and decreases radiosensitivity in cancer cells. , 2007, Cancer research.

[54]  F. Gergely,et al.  Aurora-A: the maker and breaker of spindle poles , 2007, Journal of Cell Science.

[55]  Wei Chen,et al.  Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase , 2007, Proceedings of the National Academy of Sciences.

[56]  Q. Zhan,et al.  Overexpression of Aurora-A kinase promotes tumor cell proliferation and inhibits apoptosis in esophageal squamous cell carcinoma cell line , 2006, Cell Research.

[57]  A. Puisieux,et al.  A twist for survival and cancer progression , 2005, British Journal of Cancer.

[58]  L. Lim,et al.  The GIT-associated kinase PAK targets to the centrosome and regulates Aurora-A. , 2005, Molecular cell.

[59]  Philippe Sanseau,et al.  Evolutionary relationships of Aurora kinases: Implications for model organism studies and the development of anti-cancer drugs , 2004, BMC Evolutionary Biology.

[60]  R. Knegtel,et al.  Crystal Structure of Aurora-2, an Oncogenic Serine/Threonine Kinase* , 2002, The Journal of Biological Chemistry.

[61]  Jun Yang,et al.  Cell-cycle-dependent Regulation of Human aurora A Transcription Is Mediated by Periodic Repression of E4TF1* , 2002, The Journal of Biological Chemistry.

[62]  W. Earnshaw,et al.  Chromosomal passengers and the (aurora) ABCs of mitosis. , 2001, Trends in cell biology.

[63]  E. Lees,et al.  The mitotic serine/threonine kinase Aurora2/AIK is regulated by phosphorylation and degradation , 2000, Oncogene.

[64]  R. Brown,et al.  Cell cycle arrests and radiosensitivity of human tumor cell lines: dependence on wild-type p53 for radiosensitivity. , 1994, Cancer research.

[65]  M. Mapelli,et al.  The Aurora-A/TPX2 Axis Directs Spindle Orientation by Regulating NuMa and Microtubule Dynamics , 2020, Social Science Research Network.

[66]  Ruozheng Wang,et al.  Mechanism of cervical squamous cell carcinoma radioresistance conferred by Aurora-A , 2017 .

[67]  Sun Hui-zhe Mechanistic study of pancreatic cancer cell radioresistance conferred by Aurora-A , 2013 .

[68]  V. Markovtsov,et al.  Preclinical characterization of Aurora kinase inhibitor R763/AS703569 identified through an image-based phenotypic screen , 2009, Journal of Cancer Research and Clinical Oncology.

[69]  Chi-Ying F. Huang,et al.  Gene expression profiles of the aurora family kinases. , 2006, Gene expression.

[70]  J. Weitzman Radiation resistance , 2001, Genome Biology.