Cancer Res Treat.  2020 Jul;52(3):945-956. 10.4143/crt.2020.080.

Inhibition of ATR Increases the Sensitivity to WEE1 Inhibitor in Biliary Tract Cancer

Affiliations
  • 1Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
  • 2Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea

Abstract

Purpose
Currently, the DNA damage response (DDR) pathway represents a key target for new cancer drug development. Advanced biliary tract cancer (BTC) has a poor prognosis because of the lack of efficacious treatment options. Although DNA repair pathway alterations have been reported in many patients with BTC, little is known regarding the effects of DDR-targeted agents against BTC.
Materials and Methods
In this study, nine BTC cell lines were exposed to the WEE1 inhibitor (AZD1775). In vitro, MTT assay, colony-forming assay, cell cycle analysis, phospho-histone H3 staining assay, Transwell migration assay, and western blot were performed. Then, to enhance the antitumor effect of AZD1775, the combination treatment of WEE1 inhibitor and ataxia telangiectasia mutated and Rad3 related (ATR) inhibitor (AZD6738) was conducted using MTT assay and comet assay. Finally, HuCCT-1 and SNU2670 xenograft models were established to confirm the anti-tumor effect of AZD1775 alone. Furthermore, the combination treatment was also evaluated in SNU2670 xenograft models.
Results
AZD1775 blocked the phosphorylation of CDC2 and CDC25C in all cell lines, but significantly increased apoptosis and S phase arrest in sensitive cells. However, increased p-ATR and phosphorylated ataxia telangiectasia mutated levels were observed in less sensitive cells. In addition, in vitro and in vivo data illustrated that AZD1775 combined with AZD6738 exerted more potent anti-tumor effects than either drug alone. Although WEE1 inhibition has promising anti-tumor effects in some BTC cells, the addition of ATR inhibitors could enhance its efficacy.
Conclusion
Taken together, this study supports further clinical development of DDR-targeted strategies as monotherapy or combination regimens for BTC.

Keyword

Biliary tract neoplasms; DNA damage response; WEE1; ATR

Figure

  • Fig. 1. Anti-proliferative effect of AZD1775 in biliary tract cancer (BTC) cells. (A) The anti-proliferative effects of AZD1775 in nine BTC cell lines were evaluated using the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide (MTT) assay after 72 hours. (B) The half-maximal inhibitory concentration (IC50) of AZD1775 in the MTT assay was calculated. (C) Western blot analyses of the effects of AZD1775 on signaling pathways in four BTC cell lines. Cells were treated with increasing concentrations of AZD1775 (0, 0.1, 0.5, and 1 μM) for 24 hours.

  • Fig. 2. The effect of AZD1775 alone in biliary tract cancer (BTC) cells. (A) Cell cycle analyses were performed by flow cytometry after treatment with increasing concentrations of AZD1775 (0, 0.1, 0.5, and 1 μM) for 24 hours. The data represent three independent experiments. *p < 0.05. (B) The phospho-histone H3 (p-HH3)–positive population in S phase was analyzed using flow cytometry after treatment with AZD1775 (0 and 1 μM) for 24 hours. The data represent three independent experiments. *p < 0.05. (C) Western blot analyses of the effects of AZD1775 on cell cycle pathways in four BTC cell lines. Cells were treated with increasing concentrations of AZD1775 (0, 0.1, 0.5, and 1 μM) for 24 hours. PARP, poly(ADP-ribose) polymerase. (D, E) Transwell migration assays were conducted with or without AZD1775 treatment (1 μM). The image was captured at 24 hours after treatment. The percentage of migratory cells was analyzed after images were captured. Experiments were repeated three times. *p < 0.05.

  • Fig. 3. Combination effects of WEE1 and ataxia telangiectasia mutated and Rad3 related (ATR) inhibitors in biliary tract cancer. (A) Western blotting was performed after treated with various concentrations of AZD1775 (0, 0.1, 0.5, and 1 μM) for 24 hours. ATM, ataxia telangiectasia mutated. (B) The combination indices (CIs) in the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide assay after combination treatment with AZD1775 (0, 0.001, 0.01, 0.1, 1, and 10 μM) and AZD0156 (0, 0.001, 0.01, 0.1, 1, and 10 μM)/AZD6738 (0, 0.001, 0.01, 0.1, 1, and 10 μM) for 72 hours. CI value was analyzed using CalcuSyn software. CI > 1, 1, and < 1 indicate antagonistic, additive, and synergistic effects, respectively. (C, D) Comet assays were conducted after treatment with AZD1775 (1 μM), AZD6738 (1 μM), or both for 24 hours. The tail intensity and moment were analyzed using the Comet Assay IV program. Experiments were repeated three times. *p < 0.05, **p < 0.01. (E) Cells were incubated with AZD1775 (1 μM), AZD6738 (1 μM), or both for 24 hours, and then western blotting was performed.

  • Fig. 4. Anti-tumor effects of AZD1775 alone and in combination with AZD6738 in xenograft mouse models. (A) HuCCT-1 and SNU2670 xenograft mice were administrated vehicle or AZD1775 (25 or 90 mg/kg) for 3 weeks (5 days on/2 days off). Each group contained five mice. *p < 0.05. (B) SNU2670 xenograft mice were treated with vehicle, AZD1775 (60 mg/kg), AZD6738 (25 mg/kg), or both AZD1775 and AZD6738 for 3 weeks (5 days on/2 days off). Each group contained five mice. TE, end of treatment. *p < 0.05. (C) H&E staining (×400) and immunohistochemical analysis (×400) of p-ataxia telangiectasia mutated and Rad3 related (ATR), Ki-67, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) expression were performed using isolated HuCCT-1 and SNU2670 tumors. (D) Western blot analysis of various proteins in isolated tumors.


Cited by  2 articles

Enrichment of Wee1/CDC2 and NF-κB Signaling Pathway Constituents Mutually Contributes to CDDP Resistance in Human Osteosarcoma
Zhengbo Hu, Lugen Li, Wenxing Lan, Xiao Wei, Xiangyuan Wen, Penghuan Wu, Xianliao Zhang, Xinhua Xi, Yufa Li, Liqi Wu, Wenhu Li, Xiaohong Liao
Cancer Res Treat. 2022;54(1):277-293.    doi: 10.4143/crt.2021.320.

Inhibition of WEE1 Potentiates Sensitivity to PARP Inhibitor in Biliary Tract Cancer
Hye-Rim Seo, Ah-Rong Nam, Ju-Hee Bang, Kyoung-Seok Oh, Jae-Min Kim, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh
Cancer Res Treat. 2022;54(2):541-553.    doi: 10.4143/crt.2021.473.


Reference

References

1. Pilie PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019; 16:81–104.
Article
2. Bogenberger JM, DeLeon TT, Arora M, Ahn DH, Borad MJ. Emerging role of precision medicine in biliary tract cancers. NPJ Precis Oncol. 2018; 2:21.
Article
3. Hyung J, Kim B, Yoo C, Kim KP, Jeong JH, Chang HM, et al. Clinical benefit of maintenance therapy for sdvanced biliary tract cancer patients showing no progression after first-line gemcitabine plus cisplatin. Cancer Res Treat. 2019; 51:901–9.
4. Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015; 47:1003–10.
Article
5. Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 2017; 17:93–115.
Article
6. Matheson CJ, Backos DS, Reigan P. Targeting WEE1 kinase in cancer. Trends Pharmacol Sci. 2016; 37:872–81.
Article
7. Geenen JJ, Schellens JH. Molecular pathways: targeting the protein kinase Wee1 in cancer. Clin Cancer Res. 2017; 23:4540–4.
Article
8. Krajewska M, Heijink AM, Bisselink YJ, Seinstra RI, Sillje HH, de Vries EG, et al. Forced activation of Cdk1 via wee1 inhibition impairs homologous recombination. Oncogene. 2013; 32:3001–8.
Article
9. Richer AL, Cala JM, O'Brien K, Carson VM, Inge LJ, Whitsett TG. WEE1 kinase inhibitor AZD1775 has preclinical efficacy in LKB1-deficient non-small cell lung cancer. Cancer Res. 2017; 77:4663–72.
Article
10. Nam AR, Kim JW, Cha Y, Ha H, Park JE, Bang JH, et al. Therapeutic implication of HER2 in advanced biliary tract cancer. Oncotarget. 2016; 7:58007–21.
Article
11. Nam AR, Jin MH, Park JE, Bang JH, Oh DY, Bang YJ. Therapeutic targeting of the DNA damage response using an ATR inhibitor in biliary tract cancer. Cancer Res Treat. 2019; 51:1167–79.
Article
12. Oku Y, Nishiya N, Tazawa T, Kobayashi T, Umezawa N, Sugawara Y, et al. Augmentation of the therapeutic efficacy of WEE1 kinase inhibitor AZD1775 by inhibiting the YAP-E2F1-DNA damage response pathway axis. FEBS Open Bio. 2018; 8:1001–12.
13. Wright G, Golubeva V, Remsing Rix LL, Berndt N, Luo Y, Ward GA, et al. Dual targeting of WEE1 and PLK1 by AZD1775 elicits single agent cellular anticancer activity. ACS Chem Biol. 2017; 12:1883–92.
Article
14. Jin MH, Oh DY. ATM in DNA repair in cancer. Pharmacol Ther. 2019; 203:107391.
Article
15. Lecona E, Fernandez-Capetillo O. Targeting ATR in cancer. Nat Rev Cancer. 2018; 18:586–95.
Article
16. Lin X, Chen D, Zhang C, Zhang X, Li Z, Dong B, et al. Augmented antitumor activity by olaparib plus AZD1775 in gastric cancer through disrupting DNA damage repair pathways and DNA damage checkpoint. J Exp Clin Cancer Res. 2018; 37:129.
Article
17. Jin J, Fang H, Yang F, Ji W, Guan N, Sun Z, et al. Combined inhibition of ATR and WEE1 as a novel therapeutic strategy in triple-negative breast cancer. Neoplasia. 2018; 20:478–88.
Article
18. Chen D, Lin X, Gao J, Shen L, Li Z, Dong B, et al. Wee1 inhibitor AZD1775 combined with cisplatin potentiates anticancer activity against gastric cancer by increasing DNA damage and cell apoptosis. Biomed Res Int. 2018; 2018:5813292.
Article
19. Chen WT, Ebelt ND, Stracker TH, Xhemalce B, Van Den Berg CL, Miller KM. ATM regulation of IL-8 links oxidative stress to cancer cell migration and invasion. Elife. 2015; 4:e07270.
Article
20. Zheng H, Shao F, Martin S, Xu X, Deng CX. WEE1 inhibition targets cell cycle checkpoints for triple negative breast cancers to overcome cisplatin resistance. Sci Rep. 2017; 7:43517.
Article
21. Ahn DH, Javle M, Ahn CW, Jain A, Mikhail S, Noonan AM, et al. Next-generation sequencing survey of biliary tract cancer reveals the association between tumor somatic variants and chemotherapy resistance. Cancer. 2016; 122:3657–66.
Article
22. Sun L, Moore E, Berman R, Clavijo PE, Saleh A, Chen Z, et al. WEE1 kinase inhibition reverses G2/M cell cycle checkpoint activation to sensitize cancer cells to immunotherapy. Oncoimmunology. 2018; 7:e1488359.
Article
23. Sun LL, Yang RY, Li CW, Chen MK, Shao B, Hsu JM, et al. Inhibition of ATR downregulates PD-L1 and sensitizes tumor cells to T cell-mediated killing. Am J Cancer Res. 2018; 8:1307–16.
24. Kim HJ, Min A, Im SA, Jang H, Lee KH, Lau A, et al. Antitumor activity of the ATR inhibitor AZD6738 in HER2 positive breast cancer cells. Int J Cancer. 2017; 140:109–19.
Article
25. Toulany M, Lee KJ, Fattah KR, Lin YF, Fehrenbacher B, Schaller M, et al. Akt promotes post-irradiation survival of human tumor cells through initiation, progression, and termination of DNA-PKcs-dependent DNA double-strand break repair. Mol Cancer Res. 2012; 10:945–57.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr