Korean J Physiol Pharmacol.  2024 May;28(3):197-207. 10.4196/kjpp.2024.28.3.197.

Tivozanib-induced activation of the mitochondrial apoptotic pathway and suppression of epithelial-to-mesenchymal transition in oral squamous cell carcinoma

Affiliations
  • 1Department of Oral Anatomy, School of Dentistry, Pusan National University, Yangsan 50612, Korea
  • 2Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea

Abstract

The potential of tivozanib as a treatment for oral squamous cell carcinoma (OSCC) was explored in this study. We investigated the effects of tivozanib on OSCC using the Ca9-22 and CAL27 cell lines. OSCC is a highly prevalent cancer type with a significant risk of lymphatic metastasis and recurrence, which necessitates the development of innovative treatment approaches. Tivozanib, a vascular endothelial growth factor receptor inhibitor, has shown efficacy in inhibiting neovascularization in various cancer types but has not been thoroughly studied in OSCC. Our comprehensive assessment revealed that tivozanib effectively inhibited OSCC cells. This was accompanied by the suppression of Bcl-2, a reduction in matrix metalloproteinase levels, and the induction of intrinsic pathway-mediated apoptosis. Furthermore, tivozanib contributed to epithelial-to-mesenchymal transition (EMT) inhibition by increasing E-cadherin levels while decreasing N-cadherin levels. These findings highlight the substantial anticancer potential of tivozanib in OSCC and thus its promise as a therapeutic option. Beyond reducing cell viability and inducing apoptosis, the capacity of tivozanib to inhibit EMT and modulate key proteins presents the possibility of a paradigm shift in OSCC treatment.

Keyword

Apoptosis; EMT suppression; Squamous cell carcinoma of head and neck; Tivozanib

Figure

  • Fig. 1 Tivozanib exhibits cytotoxic and antiproliferative effects and induces cell cycle arrest in OSCC cells. (A) Ca9-22 and CAL27 cells were treated with varying concentrations of tivozanib (0–200 μM) for 24 h. (B, C) Cells were treated with 1 or 10 μM tivozanib for 7 days. Cell proliferation was assessed by staining with 1% crystal violet. Cell viability and proliferation values are presented as percentages (compared to control cells with 0 μM tivozanib). (D) Cells were treated with 0–75 μM tivozanib for 24 h. After staining with PI, the cells were analyzed using FACS. The proportion of cells in the G2 phase after tivozanib treatment is presented as a histogram. (E) The protein levels of cell cycle-related factors were confirmed via Western blot. β-actin was used as a loading control. The results are presented as mean ± SD and were derived from three independent experiments (Ca9-22: *p < 0.05, **p < 0.01, ***p < 0.001; CAL27: #p < 0.05, ##p < 0.01, ###p < 0.001). OSCC, oral squamous cell carcinoma; PI, propidium iodide.

  • Fig. 2 Apoptotic responses in OSCC cells induced by tivozanib. After treating cells with 0–75 μM tivozanib for 24 h, (A) nuclear condensation and formation were observed. The number of condensed nuclei was quantified and is displayed in the graph. (B) Apoptosis in OSCC cells following tivozanib treatment was quantified using annexin V/PI staining and FACS. (C) The levels of apoptosis-related proteins were analyzed via Western blot. β-actin was used as a loading control. The results are presented as mean ± SD and were derived from three independent experiments (Ca9-22: *p < 0.05, **p < 0.01; CAL27: #p < 0.05, ##p < 0.01). OSCC, oral squamous cell carcinoma; PI, propidium iodide.

  • Fig. 3 Intrinsic pathway-mediated apoptosis in OSCC cells triggered by tivozanib. Cells were treated with 0.75 μM tivozanib for 24 h. After staining with JC-1, the cells were observed using fluorescence microscopy (A) and FACS (B). The green fluorescence signal in the cells treated with tivozanib was compared with that in the control cells. (C) An increase in the release of cytochrome c from the mitochondria suggested the activation of apoptotic pathways. (D) The levels of proteins involved in intrinsic pathway-mediated apoptosis were analyzed via Western blot. β-actin was used as a loading control. OSCC, oral squamous cell carcinoma; AIF, apoptosis-inducing factor.

  • Fig. 4 Tivozanib inhibited EMT in OSCC cells. (A, B) The wound area and number of invaded cells both significantly decreased with tivozanib treatment. (C) Tivozanib regulated the expression of EMT-related proteins in OSCC cells. (D) qPCR analysis of the mRNA levels of EMT-related markers. (E) E-cadherin expression increased with tivozanib treatment, as confirmed by confocal microscopy. The results are expressed as mean ± SD. Data were derived from three independent experiments (Ca9-22: *p < 0.05, **p < 0.01; CAL27: #p < 0.05). EMT, epithelial-to-mesenchymal transition; OSCC, oral squamous cell carcinoma; MMP9, matrix metalloproteinase 9; VEGFR2, vascular endothelial growth factor receptor 2.


Reference

1. Borse V, Konwar AN, Buragohain P. 2020; Oral cancer diagnosis and perspectives in India. Sens Int. 1:100046. DOI: 10.1016/j.sintl.2020.100046. PMID: 34766046. PMCID: PMC7515567.
Article
2. Shahoumi LA. 2021; Oral cancer stem cells: therapeutic implications and challenges. Front Oral Health. 2:685236. DOI: 10.3389/froh.2021.685236. PMID: 35048028. PMCID: PMC8757826.
Article
3. Pisani P, Airoldi M, Allais A, Aluffi Valletti P, Battista M, Benazzo M, Briatore R, Cacciola S, Cocuzza S, Colombo A, Conti B, Costanzo A, Della Vecchia L, Denaro N, Fantozzi C, Galizia D, Garzaro M, Genta I, Iasi GA, Krengli M, et al. 2020; Metastatic disease in head & neck oncology. Acta Otorhinolaryngol Ital. 40(SUPPL. 1):S1–S86. DOI: 10.14639/0392-100X-suppl.1-40-2020. PMID: 32469009. PMCID: PMC7263073.
Article
4. Kerawala C, Roques T, Jeannon JP, Bisase B. 2016; Oral cavity and lip cancer: United Kingdom national multidisciplinary guidelines. J Laryngol Otol. 130:S83–S89. DOI: 10.1017/S0022215116000499. PMID: 27841120. PMCID: PMC4873943.
Article
5. Voss JO, Freund L, Neumann F, Mrosk F, Rubarth K, Kreutzer K, Doll C, Heiland M, Koerdt S. 2022; Prognostic value of lymph node involvement in oral squamous cell carcinoma. Clin Oral Investig. 26:6711–6720. DOI: 10.1007/s00784-022-04630-7. PMID: 35895143. PMCID: PMC9643253.
Article
6. Opferman JT, Kothari A. 2018; Anti-apoptotic BCL-2 family members in development. Cell Death Differ. 25:37–45. DOI: 10.1038/cdd.2017.170. PMID: 29099482. PMCID: PMC5729530.
Article
7. Kashyap D, Garg VK, Goel N. 2021; Intrinsic and extrinsic pathways of apoptosis: role in cancer development and prognosis. Adv Protein Chem Struct Biol. 125:73–120. DOI: 10.1016/bs.apcsb.2021.01.003. PMID: 33931145.
Article
8. Jan R, Chaudhry GE. 2019; Understanding apoptosis and apoptotic pathways targeted cancer therapeutics. Adv Pharm Bull. 9:205–218. DOI: 10.15171/apb.2019.024. PMID: 31380246. PMCID: PMC6664112.
Article
9. O'Brien LE. 2022; Tissue homeostasis and non-homeostasis: from cell life cycles to organ states. Annu Rev Cell Dev Biol. 38:395–418. DOI: 10.1146/annurev-cellbio-120420-114855. PMID: 35850152. PMCID: PMC10182861.
10. Dai Y, Jin F, Wu W, Kumar SK. 2019; Cell cycle regulation and hematologic malignancies. Blood Sci. 1:34–43. DOI: 10.1097/BS9.0000000000000009. PMID: 35402801. PMCID: PMC8975093.
Article
11. Matthews HK, Bertoli C, de Bruin RAM. 2022; Cell cycle control in cancer. Nat Rev Mol Cell Biol. 23:74–88. DOI: 10.1038/s41580-021-00404-3. PMID: 34508254.
Article
12. Assani G, Zhou Y. 2019; Effect of modulation of epithelial-mesenchymal transition regulators Snail1 and Snail2 on cancer cell radiosensitivity by targeting of the cell cycle, cell apoptosis and cell migration/invasion. Oncol Lett. 17:23–30. DOI: 10.3892/ol.2018.9636. PMID: 30655734. PMCID: PMC6313178.
13. Das V, Bhattacharya S, Chikkaputtaiah C, Hazra S, Pal M. 2019; The basics of epithelial-mesenchymal transition (EMT): a study from a structure, dynamics, and functional perspective. J Cell Physiol. 234:14535–14555. DOI: 10.1002/jcp.28160. PMID: 30723913.
Article
14. Huang Y, Hong W, Wei X. 2022; The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:129. DOI: 10.1186/s13045-022-01347-8. PMID: 36076302. PMCID: PMC9461252.
Article
15. Gaianigo N, Melisi D, Carbone C. 2017; EMT and treatment resistance in pancreatic cancer. Cancers (Basel). 9:122. DOI: 10.3390/cancers9090122. PMID: 28895920. PMCID: PMC5615337.
Article
16. Van den Eynde C, De Clercq K, Van Bree R, Luyten K, Annibali D, Amant F, Han S, Van Nieuwenhuysen E, Baert T, Peeraer K, Voets T, Van Gorp T, Vriens J. 2021; TRP channel expression correlates with the epithelial-mesenchymal transition and high-risk endometrial carcinoma. Cell Mol Life Sci. 79:26. DOI: 10.1007/s00018-021-04023-1. PMID: 34936030. PMCID: PMC8732886.
Article
17. Momeny M, Moghaddaskho F, Gortany NK, Yousefi H, Sabourinejad Z, Zarrinrad G, Mirshahvaladi S, Eyvani H, Barghi F, Ahmadinia L, Ghazi-Khansari M, Dehpour AR, Amanpour S, Tavangar SM, Dardaei L, Emami AH, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. 2017; Blockade of vascular endothelial growth factor receptors by tivozanib has potential anti-tumour effects on human glioblastoma cells. Sci Rep. 7:44075. DOI: 10.1038/srep44075. PMID: 28287096. PMCID: PMC5347040.
Article
18. Passi I, Billowria K, Kumar B, Chawla PA. 2023; Tivozanib: a new hope for treating renal cell carcinoma. Anticancer Agents Med Chem. 23:562–570. DOI: 10.2174/1871520622666220617103126. PMID: 35718972.
Article
19. Schiavoni V, Campagna R, Pozzi V, Cecati M, Milanese G, Sartini D, Salvolini E, Galosi AB, Emanuelli M. 2023; Recent advances in the management of clear cell renal cell carcinoma: novel biomarkers and targeted therapies. Cancers (Basel). 15:3207. DOI: 10.3390/cancers15123207. PMID: 37370817. PMCID: PMC10296504.
Article
20. Moore JD, Yang J, Truant R, Kornbluth S. 1999; Nuclear import of Cdk/cyclin complexes: identification of distinct mechanisms for import of Cdk2/cyclin E and Cdc2/cyclin B1. J Cell Biol. 144:213–224. DOI: 10.1083/jcb.144.2.213. PMID: 9922449. PMCID: PMC2132890.
Article
21. Qian S, Wei Z, Yang W, Huang J, Yang Y, Wang J. 2022; The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front Oncol. 12:985363. DOI: 10.3389/fonc.2022.985363. PMID: 36313628. PMCID: PMC9597512.
Article
22. Anaya-Saavedra G, Ramírez-Amador V, Irigoyen-Camacho ME, García-Cuellar CM, Guido-Jiménez M, Méndez-Martínez R, García-Carrancá A. 2008; High association of human papillomavirus infection with oral cancer: a case-control study. Arch Med Res. 39:189–197. DOI: 10.1016/j.arcmed.2007.08.003. PMID: 18164962.
Article
23. Ram H, Sarkar J, Kumar H, Konwar R, Bhatt ML, Mohammad S. 2011; Oral cancer: risk factors and molecular pathogenesis. J Maxillofac Oral Surg. 10:132–137. DOI: 10.1007/s12663-011-0195-z. PMID: 22654364. PMCID: PMC3177522.
Article
24. Bugshan A, Farooq I. 2020; Oral squamous cell carcinoma: metastasis, potentially associated malignant disorders, etiology and recent advancements in diagnosis. F1000Res. 9:229. DOI: 10.12688/f1000research.22941.1. PMID: 32399208. PMCID: PMC7194458.
Article
25. Cao LM, Zhong NN, Li ZZ, Huo FY, Xiao Y, Liu B, Bu LL. 2023; Lymph node metastasis in oral squamous cell carcinoma: Where we are and where we are going. Clin Transl Discov. 3:e227. DOI: 10.1002/ctd2.227.
Article
26. Canino C, Perrone L, Bosco E, Saltalamacchia G, Mosca A, Rizzo M, Porta C. 2019; Targeting angiogenesis in metastatic renal cell carcinoma. Expert Rev Anticancer Ther. 19:245–257. DOI: 10.1080/14737140.2019.1574574. PMID: 30678509.
Article
27. Momeny M, Sabourinejad Z, Zarrinrad G, Moghaddaskho F, Eyvani H, Yousefi H, Mirshahvaladi S, Poursani EM, Barghi F, Poursheikhani A, Dardaei L, Bashash D, Ghazi-Khansari M, Tavangar SM, Dehpour AR, Yaghmaie M, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. 2017; Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci Rep. 7:45954. DOI: 10.1038/srep45954. PMID: 28383032. PMCID: PMC5382685.
Article
28. Pietenpol JA, Stewart ZA. 2002; Cell cycle checkpoint signaling: cell cycle arrest versus apoptosis. Toxicology. 181-182:475–481. DOI: 10.1016/S0300-483X(02)00460-2. PMID: 12505356.
29. Raleigh JM, O'Connell MJ. 2000; The G(2) DNA damage checkpoint targets both Wee1 and Cdc25. J Cell Sci. 113:1727–1736. DOI: 10.1242/jcs.113.10.1727. PMID: 10769204.
Article
30. Rogalińska M. 2002; Alterations in cell nuclei during apoptosis. Cell Mol Biol Lett. 7:995–1018.
31. Lucken-Ardjomande S, Martinou JC. 2005; Regulation of Bcl-2 proteins and of the permeability of the outer mitochondrial membrane. C R Biol. 328:616–631. DOI: 10.1016/j.crvi.2005.05.002. PMID: 15992745.
Article
32. Gottlieb E, Armour SM, Harris MH, Thompson CB. 2003; Mitochondrial membrane potential regulates matrix configuration and cytochrome c release during apoptosis. Cell Death Differ. 10:709–717. DOI: 10.1038/sj.cdd.4401231. PMID: 12761579.
Article
33. Sivandzade F, Bhalerao A, Cucullo L. 2019; Analysis of the mitochondrial membrane potential using the cationic JC-1 dye as a sensitive fluorescent probe. Bio Protoc. 9:e3128. DOI: 10.21769/BioProtoc.3128. PMID: 30687773. PMCID: PMC6343665.
Article
34. Santucci R, Sinibaldi F, Cozza P, Polticelli F, Fiorucci L. 2019; Cytochrome c: an extreme multifunctional protein with a key role in cell fate. Int J Biol Macromol. 136:1237–1246. DOI: 10.1016/j.ijbiomac.2019.06.180. PMID: 31252007.
Article
35. Sevrioukova IF. 2011; Apoptosis-inducing factor: structure, function, and redox regulation. Antioxid Redox Signal. 14:2545–2579. DOI: 10.1089/ars.2010.3445. PMID: 20868295. PMCID: PMC3096518.
Article
36. Noguti J, De Moura CF, De Jesus GP, Da Silva VH, Hossaka TA, Oshima CT, Ribeiro DA. 2012; Metastasis from oral cancer: an overview. Cancer Genomics Proteomics. 9:329–335.
37. Chen T, You Y, Jiang H, Wang ZZ. 2017; Epithelial-mesenchymal transition (EMT): a biological process in the development, stem cell differentiation, and tumorigenesis. J Cell Physiol. 232:3261–3272. DOI: 10.1002/jcp.25797. PMID: 28079253. PMCID: PMC5507753.
Article
38. Guarino M, Rubino B, Ballabio G. 2007; The role of epithelial-mesenchymal transition in cancer pathology. Pathology. 39:305–318. DOI: 10.1080/00313020701329914. PMID: 17558857.
Article
39. Hu QP, Kuang JY, Yang QK, Bian XW, Yu SC. 2016; Beyond a tumor suppressor: soluble E-cadherin promotes the progression of cancer. Int J Cancer. 138:2804–2812. DOI: 10.1002/ijc.29982. PMID: 26704932.
Article
40. Luo SL, Xie YG, Li Z, Ma JH, Xu X. 2014; E-cadherin expression and prognosis of oral cancer: a meta-analysis. Tumour Biol. 35:5533–5537. DOI: 10.1007/s13277-014-1728-0. PMID: 24573611.
Article
41. Marla V, Hegde V, Shrestha A. 2015; Relationship of angiogenesis and oral squamous cell carcinoma. Kathmandu Univ Med J (KUMJ). 13:178–185. DOI: 10.3126/kumj.v13i2.16796. PMID: 26643840.
Article
42. Foda HD, Zucker S. 2001; Matrix metalloproteinases in cancer invasion, metastasis and angiogenesis. Drug Discov Today. 6:478–482. DOI: 10.1016/S1359-6446(01)01752-4. PMID: 11344033.
Article
43. Takahashi S. 2011; Vascular endothelial growth factor (VEGF), VEGF receptors and their inhibitors for antiangiogenic tumor therapy. Biol Pharm Bull. 34:1785–1788. DOI: 10.1248/bpb.34.1785. PMID: 22130231.
Article
44. Park SJ, Lee N, Jeong CH. 2024; ACY-241, a histone deacetylase 6 inhibitor, suppresses the epithelial-mesenchymal transition in lung cancer cells by downregulating hypoxia-inducible factor-1 alpha. Korean J Physiol Pharmacol. 28:83–91. DOI: 10.4196/kjpp.2024.28.1.83. PMID: 38154967. PMCID: PMC10762487.
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