Click here to close
Hello! We notice that you are using Internet Explorer, which is not supported by Xenbase and may cause the site to display incorrectly.
We suggest using a current version of Chrome,
FireFox, or Safari.
J Cancer
2020 Feb 03;118:2241-2251. doi: 10.7150/jca.31989.
Show Gene links
Show Anatomy links
Therapeutic Rationale to Target Highly Expressed Aurora kinase A Conferring Poor Prognosis in Cholangiocarcinoma.
Ding X
,
Huang T
,
Peng C
,
Ahn KS
,
Andersen JB
,
Lewinska M
,
Cao Y
,
Xu G
,
Chen G
,
Kong B
,
Friess H
,
Shen S
,
Roberts LR
,
Wang L
,
Zou X
.
???displayArticle.abstract???
Background: Cholangiocarcinoma is a highly lethal neoplasm for which the currently available chemotherapeutic agents are suboptimal. Numerous studies show that alterations in expression of genes related to mitotic spindle and mitotic checkpoint are involved in chromosomal instability and tumor progression in various malignancies. This study aimed to evaluate these genes in cholangiocarcinoma patients. Material and methods: Different public datasets were analyzed to examine the expression of 76 selected mitotic spindle checkpoint genes including Aurora Kinase A (AURKA) in cholangiocarcinoma. Afterwards, cell number counting, CCK-8 assay, and Caspase 3/7 assay were used to explore the antitumor effect of AURKA inhibitor Alisertib in vitro. In addition, xenograft model was used to evaluate the antitumor effect of Alisertib in vivo. Furthermore, siRNA mediated silencing of AURKA was used to verify the function of AURKA in cholangiocarcinoma. Results: Components of the mitotic spindle checkpoint, including AURKA, were broadly dysregulated in human cholangiocarcinoma. High AURKA mRNA expression was associated with poor survival in cholangiocarcinoma patients within different datasets. AURKA specific inhibitor Alisertib, inhibited cell growth, induced cell cycle arrest in G2/M phase, and promoted apoptosis in cholangiocarcinoma cell lines. Additionally, Alisertib also inhibited tumor growth in a cholangiocarcinoma xenograft mouse model. Furthermore, AURKA knockdown by siRNA recapitulated the antitumor effect of Alisertib. AURKA expression was also highly correlated with its interaction proteins Polo-like kinase 1(PLK1) and Targeting protein for xenopus kinesin-like protein2 (TPX2) in different cholangiocarcinoma datasets. Conclusions: Highly expressed AURKA confers poor outcomes in cholangiocarcinoma and may represent a rational therapeutic target.
???displayArticle.pubmedLink???
32127951
???displayArticle.pmcLink???PMC7052919 ???displayArticle.link???J Cancer
Figure 1. Mitotic spindle checkpoint genes were broadly overexpressed in human cholangiocarcinoma. A) A heat map was drawn to show the differentially expressed gene transcripts related to the mitotic spindle checkpoint in cholangiocarcinoma tissues and normal bile duct tissues in the TCGA RNA sequencing data. B-C) Heat maps were drawn to show the differentially expressed gene transcripts related to the mitotic spindle checkpoint in cholangiocarcinoma tissues, normal surrounding liver tissues and normal bile duct tissues in the GSE107943 and GSE26566 datasets. D) Venn diagram of dysregulated genes in TCGA, GSE107943, and GSE26566 datasets.
Figure 2. AURKA expression was upregulated in cholangiocarcinoma and correlated with cell proliferation. A) AURKA mRNA expression was markedly upregulated in cholangiocarcinoma in TCGA, GSE107943, and GSE26566 datasets. B) The expression level of AURKA showed a significant positive correlation with Ki-67 expression in the dataset of TCGA, GSE107943, and GSE26566. The r and P values were determined by Pearson correlation analysis.
Figure 3. High AURKA expression correlated with poor survival in cholangiocarcinoma tumors. A) Kaplan-Meier survival curves showing the relationship between AUKRA mRNA expression and disease free survival and overall survival in the Korea cohort GSE107943 (n = 30). B) Kaplan-Meier survival curves showing the relationship between AURKA mRNA expression and overall survival in the Copenhagen cohort GSE26566 (n = 102). C) Kaplan-Meier survival curves showing the relationship between AURKA mRNA expression and overall survival in the Japan cohort EGAD00001001693 (n=111).
Figure 4. Effect of Alisertib on tumor growth in vitro and in vivo. A) Cells were treated with Alisertib at different concentrations (10nM-10 μM) for 72 h and cell proliferation was determined by cell number counting. B) IC50 values of Alisertib in cholangiocarcinoma cells following 72 h of treatment. C) Time-response curves of HCCC9810 and HuCCT1 cell lines to Alisertib treatment (100 nM). D) HCCC9810 and HuCCT1 were treated with Alisertib at different concentrations for 72 h and cell viability was determined by CCK-8 assay. E) Plated HCCC9810 and HuCCT1 were treated with Alisertib for 7 days and colonies were stained with 0.5% crystal violet. F) HuCCT1 cells were injected subcutaneously into the right flanks of athymic nude mice. When tumors reached a size of approximately150- 200 mm3, mice were randomized to receive vehicle (n=6) or Alisertib (n=6) by oral gavage. Tumor volume was measured every 3-4 days. Values shown are the mean difference in tumor volume ± SEM. G) Tumor volume in each group on day 25. Data represented are mean ± min/max group values. H) Change of body weight in mice treated with vehicle control or Alisertib. All results are presented as mean ± SEM from 3 independent experiments unless otherwise pointed, * P < 0.05, ** P < 0.01, compared with control.
Figure 5. AURKA inhibition induced cell cycle arrest and apoptosis. A) Quantification of percentage of HCCC9810 and HuCCT1 cells in different cell cycle phases following treatment with Alisertib for 48 h. B) Apoptosis analysis via Annexin V/propidium iodide staining of cholangiocarcinoma cells at 100 nM Alisertib following 72 h of treatment. C) Apoptosis analysis via Caspase 3/7 activity of cholangiocarcinoma cells at various concentrations of Alisertib following 48 h of treatment. All data shown were Mean ± SEM from 3 independent experiments, * P < 0.05, ** P < 0.01, compared with control.
Figure 6. Knockdown of AURKA diminished cell proliferation, viability, and increased G2/M phase arrest and apoptosis. A) Western immunoblotting analysis of AURKA protein expression 48 h post-siRNA knockdown of AURKA in cholangiocarcinoma cells. B) Cell proliferation following 72 h AUKRA knockdown in cholangiocarcinoma cells. Cell proliferation was determined by cell number counting. Cells number was normalized to siControl (siCont). C) CCK-8 cell viability assay following 72 h AURKA knockdown in HCCC9810 and HuCCT1 cells (n= 3). Cell viability was normalized to siControl. D) Cell cycle analysis following 48 h AURKA knockdown in HCCC9810 cells. E) Annexin V-PI assay and Caspase 3/7 activity analysis of apoptosis following 72 h AURKA knockdown in HCCC9810 cells. All data shown were Mean ± SEM from 3 independent experiments, * P < 0.05, ** P < 0.01, compared with control.
Figure 7. AURKA correlated with TPX2 and PLK1. A) Genetic interaction network of AURKA generated with the help of the GeneMANIA online tool. The top 20 genes were shown. Blue and pink lines denote pathways and physical interactions, respectively. B) Top 20 correlated genes with AURKA in the TCGA dataset of cholangiocarcinoma based on the Person's analysis. C) The expression level of AURKA showed a significant positive correlation with TPX2 and PLK1 expression in the dataset of GSE107943 and GSE26566. The r and P values were determined by Pearson correlation analysis.
Ahn,
Prognostic subclass of intrahepatic cholangiocarcinoma by integrative molecular-clinical analysis and potential targeted approach.
2019, Pubmed
Ahn,
Prognostic subclass of intrahepatic cholangiocarcinoma by integrative molecular-clinical analysis and potential targeted approach.
2019,
Pubmed
Andersen,
Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors.
2012,
Pubmed
Bièche,
Expression analysis of mitotic spindle checkpoint genes in breast carcinoma: role of NDC80/HEC1 in early breast tumorigenicity, and a two-gene signature for aneuploidy.
2011,
Pubmed
Dar,
Frequent overexpression of Aurora Kinase A in upper gastrointestinal adenocarcinomas correlates with potent antiapoptotic functions.
2008,
Pubmed
Dees,
Phase I study of aurora A kinase inhibitor MLN8237 in advanced solid tumors: safety, pharmacokinetics, pharmacodynamics, and bioavailability of two oral formulations.
2012,
Pubmed
Dickson,
Phase II study of MLN8237 (Alisertib) in advanced/metastatic sarcoma.
2016,
Pubmed
Ding,
Triptolide induces apoptotic cell death of human cholangiocarcinoma cells through inhibition of myeloid cell leukemia-1.
2014,
Pubmed
Diogo,
Spindle Assembly Checkpoint as a Potential Target in Colorectal Cancer: Current Status and Future Perspectives.
2017,
Pubmed
DuBois,
Phase I Study of the Aurora A Kinase Inhibitor Alisertib in Combination With Irinotecan and Temozolomide for Patients With Relapsed or Refractory Neuroblastoma: A NANT (New Approaches to Neuroblastoma Therapy) Trial.
2016,
Pubmed
Durlacher,
An update on the pharmacokinetics and pharmacodynamics of alisertib, a selective Aurora kinase A inhibitor.
2016,
Pubmed
Fathi,
Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia.
2017,
Pubmed
Gabrielli,
Aurora A Is Critical for Survival in HPV-Transformed Cervical Cancer.
2015,
Pubmed
Goldenson,
The aurora kinases in cell cycle and leukemia.
2015,
Pubmed
Graff,
Open-label, multicenter, phase 1 study of alisertib (MLN8237), an aurora A kinase inhibitor, with docetaxel in patients with solid tumors.
2016,
Pubmed
Humme,
Aurora Kinase A Is Upregulated in Cutaneous T-Cell Lymphoma and Represents a Potential Therapeutic Target.
2015,
Pubmed
Jeng,
Overexpression and amplification of Aurora-A in hepatocellular carcinoma.
2004,
Pubmed
Katsha,
Aurora kinase A in gastrointestinal cancers: time to target.
2015,
Pubmed
Katsha,
AURKA regulates JAK2-STAT3 activity in human gastric and esophageal cancers.
2014,
Pubmed
Kelly,
Phase I Study of the Investigational Aurora A Kinase Inhibitor Alisertib plus Rituximab or Rituximab/Vincristine in Relapsed/Refractory Aggressive B-cell Lymphoma.
2018,
Pubmed
Lampis,
MIR21 Drives Resistance to Heat Shock Protein 90 Inhibition in Cholangiocarcinoma.
2018,
Pubmed
Liewer,
Alisertib: a review of pharmacokinetics, efficacy and toxicity in patients with hematologic malignancies and solid tumors.
2018,
Pubmed
Lin,
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.
2016,
Pubmed
Maia,
A growing role for Aurora A in chromosome instability.
2014,
Pubmed
Marques,
Targeting the spindle assembly checkpoint for breast cancer treatment.
2015,
Pubmed
Melichar,
Safety and activity of alisertib, an investigational aurora kinase A inhibitor, in patients with breast cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma: a five-arm phase 2 study.
2015,
Pubmed
Nakamura,
Genomic spectra of biliary tract cancer.
2015,
Pubmed
Necchi,
An open-label, single-arm, phase 2 study of the Aurora kinase A inhibitor alisertib in patients with advanced urothelial cancer.
2016,
Pubmed
Onesti,
Recent advances for the treatment of pancreatic and biliary tract cancer after first-line treatment failure.
2015,
Pubmed
Razumilava,
Cholangiocarcinoma.
2014,
Pubmed
Rosenthal,
A Phase Ib Study of the combination of the Aurora Kinase Inhibitor Alisertib (MLN8237) and Bortezomib in Relapsed Multiple Myeloma.
2016,
Pubmed
Sehdev,
HDM2 regulation by AURKA promotes cell survival in gastric cancer.
2014,
Pubmed
Van Brocklyn,
Aurora-A inhibition offers a novel therapy effective against intracranial glioblastoma.
2014,
Pubmed
Xu,
Aurora-A contributes to cisplatin resistance and lymphatic metastasis in non-small cell lung cancer and predicts poor prognosis.
2014,
Pubmed
Yan,
Aurora-A Kinase: A Potent Oncogene and Target for Cancer Therapy.
2016,
Pubmed
Yang,
Preclinical drug metabolism and pharmacokinetics, and prediction of human pharmacokinetics and efficacious dose of the investigational Aurora A kinase inhibitor alisertib (MLN8237).
2014,
Pubmed
Yang,
Aurora kinase A promotes ovarian tumorigenesis through dysregulation of the cell cycle and suppression of BRCA2.
2010,
Pubmed