Korean J Physiol Pharmacol.  2022 Sep;26(5):377-387. 10.4196/kjpp.2022.26.5.377.

Anti-cancer effects of fenbendazole on 5-fluorouracil-resistant colorectal cancer cells

Affiliations
  • 1Department of Histology, College of Medicine, Jeju National University, Jeju 63243, Korea
  • 2Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju 61452, Korea
  • 3Department of Anatomy, College of Medicine, Jeju National University, Jeju 63243, Korea

Abstract

Benzimidazole anthelmintic agents have been recently repurposed to overcome cancers resistant to conventional therapies. To evaluate the anti-cancer effects of benzimidazole on resistant cells, various cell death pathways were investigated in 5-fluorouracil-resistant colorectal cancer cells. The viability of wild-type and 5-fluorouracil-resistant SNU-C5 colorectal cancer cells was assayed, followed by Western blotting. Flow cytometry assays for cell death and cell cycle was also performed to analyze the anti-cancer effects of benzimidazole. When compared with albendazole, fenbendazole showed higher susceptibility to 5-fluorouracil-resistant SNU-C5 cells and was used in subsequent experiments. Flow cytometry revealed that fenbendazole significantly induces apoptosis as well as cell cycle arrest at G2/ M phase on both cells. When compared with wild-type SNU-C5 cells, 5-fluorouracilresistant SNU-C5 cells showed reduced autophagy, increased ferroptosis and ferroptosis-augmented apoptosis, and less activation of caspase-8 and p53. These results suggest that fenbendazole may be a potential alternative treatment in 5-fluorouracilresistant cancer cells, and the anticancer activity of fenbendazole does not require p53 in 5-fluorouracil-resistant SNU-C5 cells.

Keyword

Apoptosis; Colorectal cancer; Drug resistance; Fenbendazole; Fenbendazole

Figure

  • Fig. 1 Anti-cancer effects of fenbendazole in colorectal cancer cells. (A) The SNU-C5 and 5-FU-resistant SNU-C5 (SNU-C5/5-FUR) cells were mock-treated with dimethyl sulfoxide (DMSO) or treated with indicated concentrations of fenbendazole or albendazole for 3 days. The extent of cell viability was determined by MTT assay as described in Methods. Data are presented as mean ± SD. **p < 0.01 and ***p < 0.001 vs. DMSO. #p < 0.05, ##p < 0.01, and ###p < 0.001 vs. SNU-C5. (B) The SNU-C5 and SNU-C5/5-FUR cells were mock-treated with DMSO or treated with indicated doses of fenbendazole for indicated days. Cell viability was determined as described in (A). (C) Representative images of SNU-C5 and SNU-C5/5-FUR cells treated with fenbendazole. Scale bar = 200 μm.

  • Fig. 2 Fenbendazole-induced cell cycle arrest at G2/M phase with increased p21 expression in colorectal cancer cells. (A) Cell cycle distribution in SNU-C5 and SNU-C5/5-FUR cells were assessed by flow cytometry after treatment with mock (dimethyl sulfoxide, DMSO) or indicated dose of fenbendazole for 3 days. The percentages of cells in each phase for SNU-C5 (left) and SNU-C5/5-FUR (right) cells are presented as the mean ± SD. *p < 0.05, **p < 0.01, and ***p < 0.001 vs. DMSO. (B) SNU-C5 (left) and SNU-C5/5-FUR (right) cells were mock-treated with DMSO or treated with indicated dose of fenbendazole. 3 days after treatment, cells were harvested and whole cell extracts were subjected to immunoblotting using the indicated antibodies. Signal intensities of p27, p21, Cyclin B1, and C-Myc were measured by AzureSpot analysis software. Data are presented as the mean ± SD. *p < 0.05, **p < 0.01, and ***p < 0.001 vs. DMSO.

  • Fig. 3 Fenbendazole-induced apoptosis via mitochondrial injury and caspase-3-poly (ADP-ribose) polymerase (PARP) pathways in colorectal cancer cells. (A) Cell death distribution was assessed by annexin V/PI staining and flow cytometry after treatment with mock (dimethyl sulfoxide, DMSO) or indicated dose of fenbendazole for 3 days. The percentages of normal, early apoptosis, late apoptosis, and necrosis in SNU-C5 (left) and SNU-C5/5-FUR (right) cells are presented as the mean ± SD. **p < 0.01 and ***p < 0.001 vs. DMSO. (B) SNU-C5 and SNU-C5/5-FUR cells were mock-treated with DMSO or treated with indicated dose of fenbendazole. 3 days after treatment, cells were harvested and whole cell extracts were subjected to immunoblotting using the anti-PARP, anti-caspase-3, and anti-cytochrome C antibodies. Signal intensity of each proteins was measured by AzureSpot analysis software. Data are presented as the mean ± SD. *p < 0.05, **p < 0.01, and ***p < 0.001 vs. DMSO.

  • Fig. 4 Fenbendazole-induced autophagy via beclin-1 in colorectal cancer cells. Effect of fenbendazole on the expression of autophagy-related proteins in SNU-C5 and SNU-C5/5-FUR cells was detected by immunoblotting. SNU-C5 and SNU-C5/5-FUR cells were mock-treated with dimethyl sulfoxide (DMSO) or treated with indicated dose of fenbendazole for 3 days. Total protein was collected, and immunoblotting analysis was performed for light chain 3 (LC3) (I/II), Beclin-1, Atg16L1, Atg3, Atg5, Atg12 (detected Atg12-5 complex also), and Atg7. GAPDH was used for a loading control. Band density was analyzed by AzureSpot analysis software, and results are expressed as the mean ± SD. *p < 0.05, **p < 0.01 vs. DMSO.

  • Fig. 5 Fenbendazole-induced ferroptosis via glutathione peroxidase 4 (GPX4) in colorectal cancer cells. (A) Effect of fenbendazole on the expression of ferroptosis-related proteins in SNU-C5 and SNU-C5/5-FUR cells was detected by immunoblotting. SNU-C5 and SNU-C5/5-FUR cells were mock-treated with dimethyl sulfoxide (DMSO) or treated with indicated dose of fenbendazole for 3 days. Total protein was collected, and immunoblotting analysis was performed for GPX4, high mobility group box 1 (HMGB1), SCL7A11, ferritin heavy chain (FTH1), ferroportin (FPN), and transferrin receptor (TfRC). GAPDH was used for a loading control. Band density was analyzed by AzureSpot analysis software, and results are expressed as the mean ± SD. *p < 0.05, **p < 0.01 vs. DMSO. (B) The SNU-C5 and SNU-C5/5-FUR cells were treated with fenbendazole alone or with fenbendazole in combination with indicated doses of ferrostatin-1 or deferoxamine mesylate (DFOM) for 3 days. The extent of cell viability was determined by MTT assay. Data are presented as mean ± SD.

  • Fig. 6 Fenbendazole-induced necroptosis in colorectal cancer cells. Effect of fenbendazole on the expression of necroptosis-related proteins in SNU-C5 and SNU-C5/5-FUR cells was detected by immunoblotting. SNU-C5 and SNU-C5/5-FUR cells were mock-treated with dimethyl sulfoxide (DMSO) or treated with indicated dose of fenbendazole for 3 days. Total protein was collected, and immunoblotting analysis was performed for phospho-receptor-interacting protein kinase (pRIP), RIP, pRIP3, RIP3, phosphor-mixed lineage kinase domain-like protein (pMLKL), MLKL, and caspase-8. GAPDH was used for a loading control. Band density was analyzed by AzureSpot analysis software, and results are expressed as the mean ± SD. *p < 0.05, **p < 0.01 vs. DMSO.

  • Fig. 7 Fenbendazole-induced p53 activation in colorectal cancer cells. SNU-C5 and SNU-C5/5-FUR cells were mock-treated with dimethyl sulfoxide (DMSO) or treated with indicated dose of fenbendazole. 3 days after treatment, cells were harvested and whole cell extracts were subjected to immunoblotting using the indicated antibodies. Signal intensity of each proteins was measured by AzureSpot analysis software. Data are presented as the mean ± SD. *p < 0.05 vs. DMSO.

  • Fig. 8 Schematic representation of cell death pathways in colorectal cancer (CRC) cells following fenbendazole treatment. Fenbendazole induces G2/M arrest and apoptosis in both (A) 5-FU-sensitive SNU-C5 and (B) 5-FU-resistant SNU-C5 (SNU-C5/5-FUR) CRC cells. In SNU-C5 cells, fenbendazole is presumed to activate p53-mediated apoptosis by increasing p53 expression. In SNU-C5/5-FUR cells, fenbendazole triggers apoptosis without affecting p53 expression, whereas fenbendazole enhances ferroptosis by inhibiting the expression of GPX4 and SLC7A11. Therefore, although fenbendazole has anti-cancer effects on both 5-FU-sensitive and resistant CRC cells, the mechanism of action appears to be different. That is, fenbendazole promotes cell death by activating p53-mediated apoptosis in SNU-C5 cells, whereas by both enhancing p53-independent apoptosis and ferroptosis-augmented apoptosis in SNU-C5/5-FUR cells. PARP, poly (ADP-ribose) polymerase; HMGB1, high mobility group box 1; GPX4, glutathione peroxidase 4; LC3, light chain 3.


Reference

1. Son DS, Lee ES, Adunyah SE. 2020; The antitumor potentials of benzimidazole anthelmintics as repurposing drugs. Immune Netw. 20:e29. DOI: 10.4110/in.2020.20.e29. PMID: 32895616. PMCID: PMC7458798.
Article
2. Mukhopadhyay T, Sasaki J, Ramesh R, Roth JA. 2002; Mebendazole elicits a potent antitumor effect on human cancer cell lines both in vitro and in vivo. Clin Cancer Res. 8:2963–2969. PMID: 12231542.
3. Sasaki J, Ramesh R, Chada S, Gomyo Y, Roth JA, Mukhopadhyay T. 2002; The anthelmintic drug mebendazole induces mitotic arrest and apoptosis by depolymerizing tubulin in non-small cell lung cancer cells. Mol Cancer Ther. 1:1201–1209. PMID: 12479701.
4. Martarelli D, Pompei P, Baldi C, Mazzoni G. 2008; Mebendazole inhibits growth of human adrenocortical carcinoma cell lines implanted in nude mice. Cancer Chemother Pharmacol. 61:809–817. DOI: 10.1007/s00280-007-0538-0. PMID: 17581752.
Article
5. Dogra N, Mukhopadhyay T. 2012; Impairment of the ubiquitin-proteasome pathway by methyl N-(6-phenylsulfanyl-1H-benzimidazol-2-yl)carbamate leads to a potent cytotoxic effect in tumor cells: a novel antiproliferative agent with a potential therapeutic implication. J Biol Chem. 287:30625–30640. DOI: 10.1074/jbc.M111.324228. PMID: 22745125. PMCID: PMC3436308.
Article
6. Lin S, Yang L, Yao Y, Xu L, Xiang Y, Zhao H, Wang L, Zuo Z, Huang X, Zhao C. 2019; Flubendazole demonstrates valid antitumor effects by inhibiting STAT3 and activating autophagy. J Exp Clin Cancer Res. 38:293. DOI: 10.1186/s13046-019-1303-z. PMID: 31287013. PMCID: PMC6615228. PMID: 3e2c66b97c604ee1b9fbb07e1c470dc9.
Article
7. Michaelis M, Agha B, Rothweiler F, Löschmann N, Voges Y, Mittelbronn M, Starzetz T, Harter PN, Abhari BA, Fulda S, Westermann F, Riecken K, Spek S, Langer K, Wiese M, Dirks WG, Zehner R, Cinatl J, Wass MN, Cinatl J Jr. 2015; Identification of flubendazole as potential anti-neuroblastoma compound in a large cell line screen. Sci Rep. 5:8202. DOI: 10.1038/srep08202. PMID: 25644037. PMCID: PMC4314641.
Article
8. Heo DS. 2020; Anthelmintics as potential anti-cancer drugs? J Korean Med Sci. 35:e75. DOI: 10.3346/jkms.2020.35.e75. PMID: 32056406. PMCID: PMC7025903.
Article
9. Yamaguchi T, Shimizu J, Oya Y, Horio Y, Hida T. 2021; Drug-induced liver injury in a patient with nonsmall cell lung cancer after the self-administration of fenbendazole based on social media information. Case Rep Oncol. 14:886–891. DOI: 10.1159/000516276. PMID: 34248555. PMCID: PMC8255718.
Article
10. Chiang RS, Syed AB, Wright JL, Montgomery B, inlavs S Sr. 2021; Fenbendazole enhancing anti-tumor effect: a case series. Clin Oncol Case Rep. 4:2. https://www.scitechnol.com/peer-review/fenbendazole-enhancing-antitumor-effect-a-case-series-P3SV.pdf.
11. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. 2018; Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. Erratum in: CA Cancer J Clin. 2020;70:313. DOI: 10.3322/caac.21492. PMID: 30207593.
Article
12. Azwar S, Seow HF, Abdullah M, Faisal Jabar M, Mohtarrudin N. 2021; Recent updates on mechanisms of resistance to 5-fluorouracil and reversal strategies in colon cancer treatment. Biology (Basel). 10:854. DOI: 10.3390/biology10090854. PMID: 34571731. PMCID: PMC8466833. PMID: 63c56935834c487a856258521353921b.
Article
13. Pan MH, Lai CS, Wu JC, Ho CT. 2011; Molecular mechanisms for chemoprevention of colorectal cancer by natural dietary compounds. Mol Nutr Food Res. 55:32–45. DOI: 10.1002/mnfr.201000412. PMID: 21207511.
Article
14. Fridman JS, Lowe SW. 2003; Control of apoptosis by p53. Oncogene. 22:9030–9040. DOI: 10.1038/sj.onc.1207116. PMID: 14663481.
Article
15. Li XL, Zhou J, Chen ZR, Chng WJ. 2015; P53 mutations in colorectal cancer- molecular pathogenesis and pharmacological reactivation. World J Gastroenterol. 21:84–93. DOI: 10.3748/wjg.v21.i1.84. PMID: 25574081. PMCID: PMC4284363.
Article
16. Thornton TM, Rincon M. 2009; Non-classical p38 map kinase functions: cell cycle checkpoints and survival. Int J Biol Sci. 5:44–51. DOI: 10.7150/ijbs.5.44. PMID: 19159010. PMCID: PMC2610339.
Article
17. Grossi V, Peserico A, Tezil T, Simone C. 2014; p38α MAPK pathway: a key factor in colorectal cancer therapy and chemoresistance. World J Gastroenterol. 20:9744–9758. DOI: 10.3748/wjg.v20.i29.9744. PMID: 25110412. PMCID: PMC4123363.
Article
18. Kim EJ, Kang JI, Kwak JW, et al. 2015; The anticancer effect of (1S,2S,3E,7E,11E)-3,7,11, 15-cembratetraen-17,2-olide(LS-1) through the activation of TGF-β signaling in SNU-C5/5-FU, fluorouracil-resistant human colon cancer cells. Mar Drugs. 13:1340–1359. DOI: 10.3390/md13031340. PMID: 25786063. PMCID: PMC4377987.
Article
19. Moon D, Kang HK, Kim J, Yoon SP. 2020; Yeast extract induces apoptosis and cell cycle arrest via activating p38 signal pathway in colorectal cancer cells. Ann Clin Lab Sci. 50:31–44. PMID: 32161010.
20. Arkun Y. 2016; Dynamic modeling and analysis of the cross-talk between insulin/AKT and MAPK/ERK signaling pathways. PLoS One. 11:e0149684. DOI: 10.1371/journal.pone.0149684. PMID: 26930065. PMCID: PMC4773096.
Article
21. Nygren P, Fryknäs M, Agerup B, Larsson R. 2013; Repositioning of the anthelmintic drug mebendazole for the treatment for colon cancer. J Cancer Res Clin Oncol. 139:2133–2140. DOI: 10.1007/s00432-013-1539-5. PMID: 24135855. PMCID: PMC3825534.
Article
22. Al-Douh MH, Sahib HB, Osman H, Abd Hamid S, Salhimi SM. 2012; Anti-proliferation effects of benzimidazole derivatives on HCT-116 colon cancer and MCF-7 breast cancer cell lines. Asian Pac J Cancer Prev. 13:4075–4079. DOI: 10.7314/APJCP.2012.13.8.4075. PMID: 23098519.
Article
23. Chen K, Chu BZ, Liu F, Li B, Gao CM, Li LL, Sun QS, Shen ZF, Jiang YY. 2015; New benzimidazole acridine derivative induces human colon cancer cell apoptosis in vitro via the ROS-JNK signaling pathway. Acta Pharmacol Sin. 36:1074–1084. DOI: 10.1038/aps.2015.44. PMID: 26235743. PMCID: PMC4561968.
Article
24. Králová V, Hanušová V, Rudolf E, Čáňová K, Skálová L. 2016; Flubendazole induces mitotic catastrophe and senescence in colon cancer cells in vitro. J Pharm Pharmacol. 68:208–218. DOI: 10.1111/jphp.12503. PMID: 26730435.
Article
25. Hanušová V, Skálová L, Králová V, Matoušková P. 2018; The effect of flubendazole on adhesion and migration in SW480 and SW620 colon cancer cells. Anticancer Agents Med Chem. 18:837–846. DOI: 10.2174/1871520618666171213141911. PMID: 29237383.
Article
26. Williamson T, Bai RY, Staedtke V, Huso D, Riggins GJ. 2016; Mebendazole and a non-steroidal anti-inflammatory combine to reduce tumor initiation in a colon cancer preclinical model. Oncotarget. 7:68571–68584. DOI: 10.18632/oncotarget.11851. PMID: 27612418. PMCID: PMC5356574.
Article
27. Duan Q, Liu Y, Rockwell S. 2013; Fenbendazole as a potential anticancer drug. Anticancer Res. 33:355–362. PMID: 23393324. PMCID: PMC3580766.
28. Doudican N, Rodriguez A, Osman I, Orlow SJ. 2008; Mebendazole induces apoptosis via Bcl-2 inactivation in chemoresistant melanoma cells. Mol Cancer Res. 6:1308–1315. DOI: 10.1158/1541-7786.MCR-07-2159. PMID: 18667591.
Article
29. Bai RY, Staedtke V, Aprhys CM, Gallia GL, Riggins GJ. 2011; Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro Oncol. 13:974–982. DOI: 10.1093/neuonc/nor077. PMID: 21764822. PMCID: PMC3158014.
Article
30. Mrkvová Z, Uldrijan S, Pombinho A, Bartůněk P, Slaninová I. 2019; Benzimidazoles downregulate Mdm2 and MdmX and activate p53 in MdmX overexpressing tumor cells. Molecules. 24:2152. DOI: 10.3390/molecules24112152. PMID: 31181622. PMCID: PMC6600429.
Article
31. Tang Y, Liang J, Wu A, Chen Y, Zhao P, Lin T, Zhang M, Xu Q, Wang J, Huang Y. 2017; Co-delivery of trichosanthin and albendazole by nano-self-assembly for overcoming tumor multidrug-resistance and metastasis. ACS Appl Mater Interfaces. 9:26648–26664. Erratum in: ACS Appl Mater Interfaces. 2020;12:3275. DOI: 10.1021/acsami.7b05292. PMID: 28741923.
Article
32. Kim JW, Kim SH, Mariappan R, Moon D, Kim J, Yoon SP. 2021; Anti-cancer effects of the aqueous extract of Orostachys japonica A. Berger on 5-fluorouracil-resistant colorectal cancer via MAPK signalling pathways in vitro and in vivo. J Ethnopharmacol. 280:114412. DOI: 10.1016/j.jep.2021.114412. PMID: 34265383.
Article
33. Feng R, Li S, Lu C, Andreas C, Stolz DB, Mapara MY, Lentzsch S. 2011; Targeting the microtubular network as a new antimyeloma strategy. Mol Cancer Ther. 10:1886–1896. DOI: 10.1158/1535-7163.MCT-11-0234. PMID: 21825007.
Article
34. Wales CT, Taylor FR, Higa AT, McAllister HA, Jacobs AT. 2015; ERK-dependent phosphorylation of HSF1 mediates chemotherapeutic resistance to benzimidazole carbamates in colorectal cancer cells. Anticancer Drugs. 26:657–666. DOI: 10.1097/CAD.0000000000000231. PMID: 25811962.
Article
35. Dogra N, Kumar A, Mukhopadhyay T. 2018; Fenbendazole acts as a moderate microtubule destabilizing agent and causes cancer cell death by modulating multiple cellular pathways. Sci Rep. 8:11926. DOI: 10.1038/s41598-018-30158-6. PMID: 30093705. PMCID: PMC6085345.
Article
36. Yang WS, iRamaratnam R Sr, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, Brown LM, Girotti AW, Cornish VW, Schreiber SL, Stockwell BR. 2014; Regulation of ferroptotic cancer cell death by GPX4. Cell. 156:317–331. DOI: 10.1016/j.cell.2013.12.010. PMID: 24439385. PMCID: PMC4076414.
Article
37. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, Basavarajappa D, Rådmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I, Wanke R, Förster H, Yefremova O, et al. 2014; Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 16:1180–1191. DOI: 10.1038/ncb3064. PMID: 25402683. PMCID: PMC4894846.
Article
38. Latunde-Dada GO. 2017; Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim Biophys Acta Gen Subj. 1861:1893–1900. DOI: 10.1016/j.bbagen.2017.05.019. PMID: 28552631.
Article
39. Lee YS, Lee DH, Jeong SY, Park SH, Oh SC, Park YS, Yu J, Choudry HA, Bartlett DL, Lee YJ. 2019; Ferroptosis-inducing agents enhance TRAIL-induced apoptosis through upregulation of death receptor 5. J Cell Biochem. 120:928–939. DOI: 10.1002/jcb.27456. PMID: 30160785. PMCID: PMC6249082.
40. Kaczmarek A, Vandenabeele P, Krysko DV. 2013; Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity. 38:209–223. DOI: 10.1016/j.immuni.2013.02.003. PMID: 23438821.
Article
41. Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, Ward Y, Wu LG, Liu ZG. 2014; Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 16:55–65. Erratum in: Nat Cell Biol. 2014;16:200. DOI: 10.1038/ncb2883. PMID: 24316671. PMCID: PMC8369836.
Article
42. Nirmala JG, Lopus M. 2020; Cell death mechanisms in eukaryotes. Cell Biol Toxicol. 36:145–164. DOI: 10.1007/s10565-019-09496-2. PMID: 31820165.
Article
43. Zhong B, Liu M, Bai C, Ruan Y, Wang Y, Qiu L, Hong Y, Wang X, Li L, Li B. 2020; Caspase-8 induces lysosome-associated cell death in cancer cells. Mol Ther. 28:1078–1091. DOI: 10.1016/j.ymthe.2020.01.022. PMID: 32053770. PMCID: PMC7132614.
Article
44. Younis NS, Ghanim AMH, Saber S. 2019; Mebendazole augments sensitivity to sorafenib by targeting MAPK and BCL-2 signalling in n-nitrosodiethylamine-induced murine hepatocellular carcinoma. Sci Rep. 9:19095. DOI: 10.1038/s41598-019-55666-x. PMID: 31836811. PMCID: PMC6911098.
Article
45. Zhang F, Li Y, Zhang H, Huang E, Gao L, Luo W, Wei Q, Fan J, Song D, Liao J, Zou Y, Liu F, Liu J, Huang J, Guo D, Ma C, Hu X, Li L, Qu X, Chen L, et al. 2017; Anthelmintic mebendazole enhances cisplatin's effect on suppressing cell proliferation and promotes differentiation of head and neck squamous cell carcinoma (HNSCC). Oncotarget. 8:12968–12982. DOI: 10.18632/oncotarget.14673. PMID: 28099902. PMCID: PMC5355070.
Article
46. Xu D, Tian W, Jiang C, Huang Z, Zheng S. 2019; The anthelmintic agent oxfendazole inhibits cell growth in non-small cell lung cancer by suppressing c-Src activation. Mol Med Rep. 19:2921–2926. DOI: 10.3892/mmr.2019.9897.
Article
47. Mirzayans R, Andrais B, Scott A, Murray D. 2012; New insights into p53 signaling and cancer cell response to DNA damage: implications for cancer therapy. J Biomed Biotechnol. 2012:170325. DOI: 10.1155/2012/170325. PMID: 22911014. PMCID: PMC3403320.
Article
48. Wang DB, Kinoshita C, Kinoshita Y, Morrison RS. 2014; p53 and mitochondrial function in neurons. Biochim Biophys Acta. 1842:1186–1197. DOI: 10.1016/j.bbadis.2013.12.015. PMID: 24412988. PMCID: PMC4074561.
Article
49. Moulder DE, Hatoum D, Tay E, Lin Y, McGowan EM. 2018; The roles of p53 in mitochondrial dynamics and cancer metabolism: the pendulum between survival and death in breast cancer? Cancers (Basel). 10:189. DOI: 10.3390/cancers10060189. PMID: 29890631. PMCID: PMC6024909.
Article
50. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. 2019; The molecular machinery of regulated cell death. Cell Res. 29:347–364. DOI: 10.1038/s41422-019-0164-5. PMID: 30948788. PMCID: PMC6796845.
Article
51. Kang R, Kroemer G, Tang D. 2019; The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 133:162–168. DOI: 10.1016/j.freeradbiomed.2018.05.074. PMID: 29800655. PMCID: PMC6251771.
Article
52. Lei G, Mao C, Yan Y, Zhuang L, Gan B. 2021; Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell. 12:836–857. DOI: 10.1007/s13238-021-00841-y. PMID: 33891303. PMCID: PMC8563889.
Article
53. Ren LW, Li W, Zheng XJ, Liu JY, Yang YH, Li S, Zhang S, Fu WQ, Xiao B, Wang JH, Du GH. 2022; Benzimidazoles induce concurrent apoptosis and pyroptosis of human glioblastoma cells via arresting cell cycle. Acta Pharmacol Sin. 43:194–208. DOI: 10.1038/s41401-021-00752-y. PMID: 34433903.
Article
54. Elayapillai S, Ramraj S, Benbrook DM, Bieniasz M, Wang L, Pathuri G, Isingizwe ZR, Kennedy AL, Zhao YD, Lightfoot S, Hunsucker LA, Gunderson CC. 2021; Potential and mechanism of mebendazole for treatment and maintenance of ovarian cancer. Gynecol Oncol. 160:302–311. DOI: 10.1016/j.ygyno.2020.10.010. PMID: 33131904. PMCID: PMC8820236.
Article
Full Text Links
  • KJPP
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