Cancer Res Treat.  2022 Apr;54(2):541-553. 10.4143/crt.2021.473.

Inhibition of WEE1 Potentiates Sensitivity to PARP Inhibitor in Biliary Tract Cancer

Affiliations
  • 1Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
  • 2Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, Korea
  • 3Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea

Abstract

Purpose
Up to 20% of patients with biliary tract cancer (BTC) have alterations in DNA damage response (DDR) genes, including homologous recombination (HR) genes. Therefore, the DDR pathway could be a promising target for new drug development in BTC. We aim to investigate the anti-tumor effects using poly(ADP-ribose) polymerase (PARP) and WEE1 inhibitors in BTC.
Materials and Methods
We used 10 BTC cell lines to evaluate an anti-tumor effect of olaparib (a PARP inhibitor) and AZD1775 (a WEE1 inhibitor) in in vitro. Additionally, we established SNU869 xenograft model for in vivo experiments.
Results
In this study, we observed a modest anti-proliferative effect of olaparib. DNA double-strand break (DSB) and apoptosis were increased by olaparib in BTC cells. However, olaparib-induced DNA DSB was repaired through the HR pathway, and G2 arrest was induced to secure the time for repair. As AZD1775 typically regulates the G2/M checkpoint, we combined olaparib with AZD1775 to abrogate G2 arrest. We observed that AZD1775 downregulated p-CDK1, a G2/M cell cycle checkpoint protein, and induced early mitotic entry. AZD1775 also decreased CtIP and RAD51 expression and disrupted HR repair. In xenograft model, olaparib plus AZD1775 treatment reduced tumor growth more potently than did monotherapy with either drug.
Conclusion
This is the first study to suggest that olaparib combined with AZD1775 can induce synergistic anti-tumor effects against BTC. Combination therapy that blocks dual PARP and WEE1 has the potential to be further clinically developed for BTC patients.

Keyword

Poly(ADP-ribose) polymerase; WEE1; Biliary tract neoplasms; DNA damage response; Cell cycle checkpoint; Homologous recombination

Figure

  • Fig. 1 Olaparib-induced DNA damage and simultaneously activated G2/M checkpoint. (A) MTT assay was performed in 10 biliary tract cancer cell lines for 5 days after treatment with olaparib (0, 0.01, 0.1, 1, and 10 μM). The half-maximal inhibitory concentration (IC50) of olaparib was calculated using SigmaPlot. Error bars represent mean±standar deviation (SD). (B) The proteins that indicated the DNA damage and apoptosis were analyzed by western blotting after treatment with olaparib (0, 1, and 10 μM) for 72 hours. (C) Cell cycle stages were determined by flow cytometry after 72-hour treatment with olaparib (0, 1, and 10 μM). Error bars represent mean±SD. *p < 0.05, **p < 0.01, ***p < 0.001. (D) The levels of cell cycle proteins and homologous recombination proteins were assessed by western blotting of cells treated with 0, 1, and 10 μM olaparib for 24 hours.

  • Fig. 2 Combination therapy with olaparib and AZD1775 induced more potent DNA damage and apoptosis. (A) Combination effect was investigated by MTT for 5 days before treatment with olaparib (0, 0.01, 0.1, 1, and 10 μM) and AZD1775 (0, 0.001, 0.01, 0.1, and 1 μM), dose ratio was 10:1. CI value was analyzed by CalcuSyn software. If the value is lower than 1 is synergistic, and the cell lines that higher than 1 exhibit an antagonistic effect. (B) Colony-forming analysis was performed with olaparib (0, 1, 5 μM), AZD1775 (0, 0.01, 0.05, 0.1 μM) or both for 10 days. Error bars represent mean±standard deviation (SD) (n=3). (C) Comet analysis was conducted after treatment with olaparib (5 μM), AZD1775 (0.2 μM) or both for 72 hours. The tail intensity and tail moment were analyzed using the Comet assay IV program. The experiment was repeated three times. Data are expressed as mean±standrad error of mean (n=100). (D) Apoptosis analysis was performed using annexin V/propidium iodide (PI) double staining after 24-hour and 72-hour treatment with olaparib (5 μM), AZD1775 (0.1 μM) or both. Error bars represent mean±SD (n=4). (E) Cell cycle analysis were performed with PI-stained BTC cells using flow cytometry after 72-hour treatment with olaparib (5 μM), AZD1775 (0.2 μM) or both. Error bars represent mean±SD (n=3). (F) Western blotting of two cell lines was performed after 72-hour incubation with olaparib, AZD1775, or both. *p < 0.05, **p < 0.01, ***p < 0.001.

  • Fig. 3 AZD1775 abrogated G2 phase arrest and induced early mitotic entry. (A) The phospho-histone H3 (p-HH3)-positive population was analyzed by flow cytometry after 6-hour and 24-hour treatment with olaparib (5 μM), AZD1775 (0.1 μM), or both. The experiment was repeated four times. #p < 0.05, ##p < 0.01, in G2/M phase. *p < 0.05, in all cell cycle phases. (B) Cell cycle assay was performed after 6-hour and 24-hour treatment with olaparib (5 μM), AZD1775 (0.1 μM), or both (n=3). Error bars represent mean±standard deviation. *p < 0.05. (C) Western blotting of cell cycle arrest-related signaling proteins was performed in biliary tract cancer cells treated with olaparib (5 μM), AZD1775 (0.2 μM), or both for 24 hours.

  • Fig. 4 AZD1775 disrupted homologous recombination (HR) repair and enhanced the effects of olaparib. (A) Immunofluorescence staining of γ-H2AX (red) and RAD51 (green) in SNU869 cells treated with olaparib (5 μM), AZD1775 (0.2 μM), or a combination of olaparib and AZD1775 for 24 hours. Cells with more than five foci were counted; 100 cells were analyzed, with experiments repeated 3 times. A confocal microscope at ×400 magnification was used. Error bars represent mean±standard deviation. *p < 0.05, **p < 0.01, ***p < 0.001. (B) Western blotting was performed after 24-hour treatment to show decrease in HR-related proteins. (C) Cells were transfected with WEE1-specific siRNAs (50 nM) for 6 hours and cells were used for western blotting. (D) After transfected with siWEE1, cells were treated with olaparib 24 hours, and western blotting was conducted. (E) After transfected with siWEE1, we treated olaparib for 72 hours, and then performed western blotting.

  • Fig. 5 Combination treatment of olaparib with AZD1775 showed Anti-tumor growth effects in a xenograft mouse model. (A) Vehicle, olaparib (50 mg/kg), AZD1775 (25 mg/kg), or both drugs were administered orally once a day for 4 weeks (5 days on/2 days off) to all groups of SNU869 xenograft mice (n=5 per group). Tumor volumes were measured three times weekly. Data are expressed as mean±standard error of mean (SEM). *p < 0.05. (B) Mouse weights (n=5 per group) were measured three times weekly. Data are expressed as mean±SEM. (C) Western blotting was performed for tumor-specific proteins in each group after tumor isolation. (D) SNU869-xenografted tumors were stained with H&E (×200), Ki67 (×200), terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL; ×200), and p-CDK1 (×400) for immunohistochemical analysis. Scale bars=200 μm (H&E, Ki67, and TUNEL), 60 μm (p-CDK1). Immunohistochemical staining was conducted with identical blocks per group.


Reference

References

1. Gottifredi V. Targeting DNA damage response kinases in cancer therapy. Mutat Res. 2020; 821:111725.
Article
2. Tariq NU, McNamara MG, Valle JW. Biliary tract cancers: current knowledge, clinical candidates and future challenges. Cancer Manag Res. 2019; 11:2623–42.
3. Ricci AD, Rizzo A, Bonucci C, Tober N, Palloni A, Mollica V, et al. PARP inhibitors in biliary tract cancer: a new kid on the block? Medicines (Basel). 2020; 7:54.
Article
4. Ahn DH, Bekaii-Saab T. Biliary tract cancer and genomic alterations in homologous recombinant deficiency: exploiting synthetic lethality with PARP inhibitors. Chin Clin Oncol. 2020; 9:6.
Article
5. Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017; 377:523–33.
6. Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019; 381:317–27.
Article
7. Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2018; 379:2495–505.
Article
8. de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020; 382:2091–102.
Article
9. Pommier Y, O’Connor MJ, de Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016; 8:362ps17.
Article
10. Shrivastav M, De Haro LP, Nickoloff JA. Regulation of DNA double-strand break repair pathway choice. Cell Res. 2008; 18:134–47.
Article
11. Sunada S, Nakanishi A, Miki Y. Crosstalk of DNA double-strand break repair pathways in poly(ADP-ribose) polymerase inhibitor treatment of breast cancer susceptibility gene 1/2-mutated cancer. Cancer Sci. 2018; 109:893–9.
Article
12. Lee EK, Matulonis UA. PARP inhibitor resistance mechanisms and implications for post-progression combination therapies. Cancers (Basel). 2020; 12:2054.
Article
13. Jelinic P, Levine DA. New insights into PARP inhibitors’ effect on cell cycle and homology-directed DNA damage repair. Mol Cancer Ther. 2014; 13:1645–54.
Article
14. Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene. 2001; 20:1803–15.
Article
15. Patil M, Pabla N, Dong Z. Checkpoint kinase 1 in DNA damage response and cell cycle regulation. Cell Mol Life Sci. 2013; 70:4009–21.
Article
16. Ghelli Luserna di Rora A, Cerchione C, Martinelli G, Simonetti G. A WEE1 family business: regulation of mitosis, cancer progression, and therapeutic target. J Hematol Oncol. 2020; 13:126.
Article
17. Schmidt M, Rohe A, Platzer C, Najjar A, Erdmann F, Sippl W. Regulation of G2/M transition by inhibition of WEE1 and PKMYT1 kinases. Molecules. 2017; 22:2045.
Article
18. Yang L, Shen C, Pettit CJ, Li T, Hu AJ, Miller ED, et al. Wee1 kinase inhibitor AZD1775 effectively sensitizes esophageal cancer to radiotherapy. Clin Cancer Res. 2020; 26:3740–50.
Article
19. Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004; 23:2825–37.
Article
20. 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
21. 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
22. 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
23. Geenen JJ, Schellens JH. Molecular pathways: targeting the protein kinase Wee1 in cancer. Clin Cancer Res. 2017; 23:4540–4.
Article
24. Liu K, Zheng M, Lu R, Du J, Zhao Q, Li Z, et al. The role of CDC25C in cell cycle regulation and clinical cancer therapy: a systematic review. Cancer Cell Int. 2020; 20:213.
Article
25. Wang J, Ding Q, Fujimori H, Motegi A, Miki Y, Masutani M. Loss of CtIP disturbs homologous recombination repair and sensitizes breast cancer cells to PARP inhibitors. Oncotarget. 2016; 7:7701–14.
Article
26. Zhao Q, Guan J, Zhang Z, Lv J, Wang Y, Liu L, et al. Inhibition of Rad51 sensitizes breast cancer cells with wild-type PTEN to olaparib. Biomed Pharmacother. 2017; 94:165–8.
Article
27. Lim G, Chang Y, Huh WK. Phosphoregulation of Rad51/ Rad52 by CDK1 functions as a molecular switch for cell cycle-specific activation of homologous recombination. Sci Adv. 2020; 6:eaay2669.
28. Buis J, Stoneham T, Spehalski E, Ferguson DO. Mre11 regulates CtIP-dependent double-strand break repair by interaction with CDK2. Nat Struct Mol Biol. 2012; 19:246–52.
Article
29. Wang H, Shi LZ, Wong CC, Han X, Hwang PY, Truong LN, et al. The interaction of CtIP and Nbs1 connects CDK and ATM to regulate HR-mediated double-strand break repair. PLoS Genet. 2013; 9:e1003277.
Article
30. Nam AR, Jin MH, Bang JH, Oh KS, Seo HR, Oh DY, et al. Inhibition of ATR increases the sensitivity to WEE1 inhibitor in biliary tract cancer. Cancer Res Treat. 2020; 52:945–56.
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