Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 May 1;133(9):e161929.
doi: 10.1172/JCI161929.

Aurora A kinase inhibition compromises its antitumor efficacy by elevating PD-L1 expression

Affiliations

Aurora A kinase inhibition compromises its antitumor efficacy by elevating PD-L1 expression

Xiaobo Wang et al. J Clin Invest. .

Abstract

Aurora A plays a critical role in G2/M transition and mitosis, making it an attractive target for cancer treatment. Aurora A inhibitors showed remarkable antitumor effects in preclinical studies, but unsatisfactory outcomes in clinical trials have greatly limited their development. In this study, the Aurora A inhibitor alisertib upregulated programmed death ligand 1 (PD-L1) expression in a panel of tumor cells both in vitro and in vivo. Upregulation of the checkpoint protein PD-L1 reduced antitumor immunity in immune-competent mice, paradoxically inhibiting the antitumor effects of alisertib. Mechanistically, Aurora A directly bound to and phosphorylated cyclic GMP-AMP synthase (cGAS), suppressing PD-L1 expression in tumor cells. Aurora A inhibition by alisertib activated the cGAS/stimulator of IFN genes (STING)/NF-κB pathway and promoted PD-L1 expression. Combining alisertib with anti-PD-L1 antibody improved antitumor immunity and enhanced the antitumor effects of alisertib in immune-competent mice. Our results, which reveal the immunomodulatory functions of Aurora A inhibitors and provide a plausible explanation for the poor clinical outcomes with their use, offer a potential approach to improve the antitumor efficacy of these inhibitors.

Keywords: Cancer; Drug therapy; Oncology.

PubMed Disclaimer

Figures

Figure 1
Figure 1. Aurora A kinase inhibitor elevates PD-L1 expression in tumor cells.
(A) Flow cytometric analysis of cell-surface PD-L1 expression in BxPC3 cells following treatment for 72 hours with 1 μmol/L alisertib, 0.5 μmol/L AZD1152, or 0.5 μmol/L tozasertib. (B) Normalized MFI for the data shown in A (n = 3). (C) Western blot showing PD-L1 expression in BxPC3 cells after treatment for 72 hours with the indicated concentrations of alisertib. (D and E) qRT-PCR analysis of PDL1 expression in BxPC3 cells after treatment with alisertib at the indicated concentrations (D) and for the indicated durations (E) (n = 3). (F) Flow cytometric analysis of cell-surface PD-1 binding of BxPC3 cells after treatment for 72 hours with the indicated concentrations of alisertib. (G) Normalized MFI for the data in F (n = 3). hFc, human Fc. Data indicate the mean ± SD. **P < 0.01 and ****P < 0.0001, by 1-way ANOVA.
Figure 2
Figure 2. Knockdown of AURKA elevates PD-L1 expression in tumor cells.
(A) Western blot analysis of the expression of indicated proteins following treatment with an AURKA siRNA or a control siRNA. (B) Flow cytometric analysis of surface expression of PD-L1 on BxPC3 cells after treatment with the indicated siRNA. (C) Normalized MFI for the data shown in B (n = 3). (D) qRT-PCR analysis of PDL1 expression in BxPC3 cells after treatment with the indicated siRNA (n = 3). (E) Flow cytometric analysis of cell-surface PD-1 binding of BxPC3 cells after treatment with the indicated siRNAs. (F) Normalized MFI for the data in E (n = 3). Data indicate the mean ± SD. ***P < 0.001 and ****P < 0.0001, by 1-way ANOVA.
Figure 3
Figure 3. Aurora A inhibition upregulates PD-L1 expression in vivo, which compromises the inhibitor’s antitumor efficacy.
(AC) (A) Effect of vehicle or alisertib treatment on CT26 growth in BALB/c WT or BALB/c nude mice (n = 6). (B) Excised tumor tissues were digested to a single-cell suspension, and PD-L1+ cells were evaluated by flow cytometry. SSC-A, side scatter area. (C) Cumulative data for the percentage of PD-L1+ cells in B (n = 6). (D) Effect of vehicle or alisertib treatment on tumor growth in WT or Pdl1–/– MC38 mouse tumor models (n = 6). (E) Effect of vehicle or alisertib treatment on MC38 growth in WT or Pdl1–/– C57BL/6 mice (n = 6). Data indicate the mean ± SD. A 2-way ANOVA was applied to compare time-dependent tumor growth. *P < 0.05 and ****P < 0.0001, by unpaired t test.
Figure 4
Figure 4. PD-L1 upregulation caused by Aurora A inhibition depends on STING/NF-κB activation.
(A and B) RNA-Seq analysis was performed on BxPC3 cells after treatment for 72 hours with DMSO or 1 μmol/L alisertib. (A) KEGG pathway analysis of genes differentially expressed between the DMSO- and alisertib-treated groups. The most substantially enriched pathways are shown. p.adjust, adjusted P value. (B) Heatmap of gene expression levels of the indicated cytokines or chemokines in DMSO- or alisertib-treated BxPC3 cells. (C and D) BxPC3 cells were pretreated for 6 hours with 10 μmol/L TPCA-1 or with 5 μmol/L BAY11-7082, followed by treatment for 72 hours with 1 μmol/L alisertib, and PD-L1 expression was assessed by Western blotting (C) and qRT-PCR (D). (E and F) qRT-PCR analysis of IFNB expression in BxPC3 cells after the indicated concentrations (E) and durations (F) of alisertib treatment (n = 3). (G) qRT-PCR analysis of IFNB expression in BxPC3 cells after treatment with the indicated siRNA (n = 3). (H) qRT-PCR analysis of IFNB expression (n = 3). (IK) WT BxPC3 cells or STING–/– BxPC3 cells were treated with 1 μmol/L alisertib for 72 hours. (I) Western blot analysis of PD-L1 and STING protein levels. qRT-PCR analysis of PDL1 (J) and IFNB (K) mRNA levels (n = 3). Data indicate the mean ± SD. ***P < 0.001 and ****P < 0.0001, by 1-way ANOVA (DH) and 2-way ANOVA (J and K).
Figure 5
Figure 5. Aurora A inhibition–induced PD-L1 expression is mediated by cGAS dephosphorylation.
(AC) WT BxPC3 cells or CGAS–/– BxPC3 cells were treated with 1 μmol/L alisertib for 72 hours. (A) Western blot analysis of PD-L1 and cGAS protein levels. PD-L1 and cGAS were detected separately in 2 gels using the same biological samples, and GAPDH in each gel served as the loading control. qRT-PCR analysis of PDL1 (B) and IFNB (C) mRNA levels (n = 3). (D) Co-immunoprecipitation of Aurora A and cGAS. HEK293T cells were transfected with the indicated vectors encoding HA–Aurora A and Flag-cGAS. Whole-cell lysates were immunoprecipitated with anti-Flag beads, and the interactions were analyzed by Western blotting. (E) BxPC3 cells were synchronized with 10 μmol/L Ro-3306 for 16 hours and released into mitosis in the presence of alisertib at the indicated concentrations. Phosphorylation of cGAS was analyzed by Phos-tag electrophoresis. Data indicate the mean ± SD. ****P < 0.0001, by 2-way ANOVA.
Figure 6
Figure 6. PD-L1 blockade therapy improves the antitumor activity of alisertib.
(AE) BALB/c mice were inoculated with CT26 cells and administered alisertib, anti–PD-L1 antibody alone, or their combination. (A) Tumor growth curves of CT26 in BALB/c mice. Tumor volumes were measured at the indicated time points (n = 6). (B and D) Flow cytometric analysis of tumor-infiltrating CD8+ T cells (B) and granzyme B+CD8+ T cells (D). Representative plots are shown. (C and E) Cumulative data for B and D (n = 6). Data indicate the mean ± SD. A 2-way ANOVA was applied to compare time-dependent tumor growth. *P < 0.05, **P < 0.01, and ****P < 0.0001, by unpaired t test.
Figure 7
Figure 7. Active Aurora A levels are negatively associated with PD-L1 expression in human tumor tissues.
Representative images of IHC staining for p–Aurora A and PD-L1 in human colon cancer tissues. Scale bar: 500 μm (enlarged insets).

References

    1. Barr AR, Gergely F. Aurora-A: the maker and breaker of spindle poles. J Cell Sci. 2007;120(pt 17):2987–2996. doi: 10.1242/jcs.013136. - DOI - PubMed
    1. Liewer S, Huddleston A. Alisertib: a review of pharmacokinetics, efficacy and toxicity in patients with hematologic malignancies and solid tumors. Expert Opin Investig Drugs. 2018;27(1):105–112. doi: 10.1080/13543784.2018.1417382. - DOI - PubMed
    1. Durlacher CT, et al. An update on the pharmacokinetics and pharmacodynamics of alisertib, a selective Aurora kinase A inhibitor. Clin Exp Pharmacol Physiol. 2016;43(6):585–601. doi: 10.1111/1440-1681.12571. - DOI - PubMed
    1. Bavetsias V, Linardopoulos S. Aurora kinase inhibitors: current status and outlook. Front Oncol. 2015;5:278. - PMC - PubMed
    1. O’Connor OA, et al. Randomized phase III study of alisertib or investigator’s choice (selected single agent) in patients with relapsed or refractory peripheral T-cell lymphoma. J Clin Oncol. 2019;37(8):613–623. doi: 10.1200/JCO.18.00899. - DOI - PMC - PubMed

Publication types