Enhancement of radiation response in p53-deficient cancer cells by the Aurora-B kinase inhibitor AZD1152

Overexpression of the Aurora-B kinase correlates with oncogenic transformation and poor prognosis. We evaluated the effects of the bona fide Aurora-B kinase inhibitor AZD1152 on tumor responses to ionizing radiation (IR). When p53wt HCT116 and A549 cells were pretreated with AZD1152-HQPA prior to IR, additive effects were observed. Interestingly, more pronounced tumoricidal effects were observed in p53-deficient HCT116 and HT29 cells, as well as A549 cells treated with the p53 inhibitor cyclic pifithrin-α. In vivo studies on xenografted mice confirmed enhanced tumor growth delay after the combination of IR plus AZD1152-IR as compared to IR alone. Again, this effect was more pronounced with p53−/− HCT116 and p53-mutant xenografts. The AZD1152-mediated radiosensitization was mimicked by knockdown of Aurora-B with a short interference RNA or by inhibition of Aurora-B by transfection with an inducible kinase-dead Aurora-B. The radiosensitizing effect of AZD1152 was lost in CHK2−/− and 14-3-3−/− HCT116 cells. Altogether, these data indicate that AZD1152 can radiosensitize tumor cell lines in vitro and in vivo, the fact that these effects are exacerbated in p53-deficient cancer cells is of potential interest for further clinical development.

[1]  L. Milas,et al.  Combination of taxanes with radiation: preclinical studies. , 1999, Seminars in radiation oncology.

[2]  David Bebbington,et al.  VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth in vivo , 2004, Nature Medicine.

[3]  T. Tuschl,et al.  Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells , 2001, Nature.

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

[5]  P. D. Andrews,et al.  Aurora kinases: shining lights on the therapeutic horizon? , 2005, Oncogene.

[6]  W. Earnshaw,et al.  The cellular geography of Aurora kinases , 2003, Nature Reviews Molecular Cell Biology.

[7]  K. Kinzler,et al.  Requirement for p53 and p21 to sustain G2 arrest after DNA damage. , 1998, Science.

[8]  Erich A. Nigg,et al.  Cell division: Mitotic kinases as regulators of cell division and its checkpoints , 2001, Nature Reviews Molecular Cell Biology.

[9]  A. Venkitaraman,et al.  AURORA-A amplification overrides the mitotic spindle assembly checkpoint, inducing resistance to Taxol. , 2003, Cancer cell.

[10]  M V Chernov,et al.  A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. , 1999, Science.

[11]  Stephen S. Taylor,et al.  Validating Aurora B as an anti-cancer drug target , 2006, Journal of Cell Science.

[12]  Stephen S. Taylor,et al.  Aurora B couples chromosome alignment with anaphase by targeting BubR1, Mad2, and Cenp-E to kinetochores , 2003, The Journal of cell biology.

[13]  Brian Schryver,et al.  A homologue of Drosophila aurora kinase is oncogenic and amplified in human colorectal cancers , 1998, The EMBO journal.

[14]  J. Bourhis,et al.  Caspase independence of radio-induced cell death , 2006, Oncogene.

[15]  Guido Kroemer,et al.  Cell death by mitotic catastrophe: a molecular definition , 2004, Oncogene.

[16]  H. Saya,et al.  Aurora-A and an Interacting Activator, the LIM Protein Ajuba, Are Required for Mitotic Commitment in Human Cells , 2003, Cell.

[17]  K. Kinzler,et al.  14-3-3σ is required to prevent mitotic catastrophe after DNA damage , 1999, Nature.

[18]  Stephen S. Taylor,et al.  Aurora-kinase inhibitors as anticancer agents , 2004, Nature Reviews Cancer.

[19]  Kaori Sasai,et al.  Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53 , 2004, Nature Genetics.

[20]  Jiri Bartek,et al.  Cell-cycle checkpoints and cancer , 2004, Nature.

[21]  B. Vogelstein,et al.  The Chk2 Tumor Suppressor Is Not Required for p53 Responses in Human Cancer Cells* , 2003, Journal of Biological Chemistry.

[22]  Erich A Nigg,et al.  Aurora kinases link chromosome segregation and cell division to cancer susceptibility. , 2004, Current opinion in genetics & development.

[23]  Rajesh Odedra,et al.  AZD1152, a Selective Inhibitor of Aurora B Kinase, Inhibits Human Tumor Xenograft Growth by Inducing Apoptosis , 2007, Clinical Cancer Research.

[24]  J. Ruderman,et al.  Aurora kinase inhibitor ZM447439 blocks chromosome-induced spindle assembly, the completion of chromosome condensation, and the establishment of the spindle integrity checkpoint in Xenopus egg extracts. , 2005, Molecular biology of the cell.

[25]  Weikang Tao,et al.  Induction of apoptosis by an inhibitor of the mitotic kinesin KSP requires both activation of the spindle assembly checkpoint and mitotic slippage. , 2005, Cancer cell.

[26]  M. Loda,et al.  The Aurora kinase inhibitor VX-680 induces endoreduplication and apoptosis preferentially in cells with compromised p53-dependent postmitotic checkpoint function. , 2006, Cancer research.

[27]  J. Peters,et al.  The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore–microtubule attachment and in maintaining the spindle assembly checkpoint , 2003, The Journal of cell biology.

[28]  T. Pawlik,et al.  Role of cell cycle in mediating sensitivity to radiotherapy. , 2004, International journal of radiation oncology, biology, physics.

[29]  R. Klinke,et al.  A Chemical Inhibitor of p 53 That Protects Mice from the Side Effects of Cancer Therapy , 1999 .

[30]  C. Prigent,et al.  Aurora kinases, aneuploidy and cancer, a coincidence or a real link? , 2005, Trends in cell biology.

[31]  Jiri Bartek,et al.  Targeting the checkpoint kinases: chemosensitization versus chemoprotection , 2004, Nature Reviews Cancer.