Generic placeholder image

Current Cancer Therapy Reviews

Editor-in-Chief

ISSN (Print): 1573-3947
ISSN (Online): 1875-6301

Review Article

Emerging Role of Wnt/Beta-Catenin Signalling Pathways in Cancer Progression and Role of Small Molecule Tankyrase Inhibitors in Combating Multistage Cancers

Author(s): Babli Khatun, Venkatesh Kamath, Muddukrishna Badamane Sathyanarayana, Aravinda Pai*, Ramji Gupta and Rishabha Malviya*

Volume 17, Issue 4, 2021

Published on: 28 June, 2021

Page: [304 - 311] Pages: 8

DOI: 10.2174/1573394717666210628122306

Price: $65

Abstract

In the present review, an attempt has been made to summarize the development of various Tankyrase inhibitors focussing on Wnt/beta-Catenin pathways along with other cancer targets. The last decade witnessed a plethora of research related to the role of various genetic and epigenetic events that are responsible for the progression of multistage cancers. As a result, the discovery of various signalling pathways responsible for the development of different types of cancers has resulted in the development of molecularly targeted anticancer agents. Out of the many signalling pathways, the Wnt/beta-Catenin pathways have attracted the attention of many research groups owing to their involvement in cell proliferation, role in apoptosis induction, cellular differentiation and also cell migration. The abnormal activation of this pathways has been documented in a variety of tumour cells. Another crucial factor that makes this pathway attractive to the researches is its direct involvement with poly ADP ribose polymerases. Tankyrases are poly ADP (Adenosine Diphosphate) ribose polymerases that have the capacity to inhibit Wnt/beta-Catenin pathways and become an attractive target for anticancer drugs.

Keywords: Wnt-beta catenin, cancer, colorectal, tankyrases, telomere, cell death.

Graphical Abstract
[1]
Otto H, Reche PA, Bazan F, Dittmar K, Haag F, Koch-Nolte F. In silico characterization of the family of PARP-like poly(ADP-ribosyl)transferases (pARTs). BMC Genomics 2005; 6: 139.
[http://dx.doi.org/10.1186/1471-2164-6-139] [PMID: 16202152]
[2]
Smith S, Schmitt A, Langei- T, De. Tan Kyrase. A Poly ( ADP-Ri Bose ) polymerase a t human telomeres. Science 1998; 282: 1-4.
[3]
Haikarainen T, Krauss S, Lehtio L. Tankyrases: Structure, function and therapeutic implications in cancer. Curr Pharm Des 2014; 20(41): 6472-88.
[http://dx.doi.org/10.2174/1381612820666140630101525] [PMID: 24975604]
[4]
Kim MK. Novel insight into the function of tankyrase. Oncol Lett 2018; 16(6): 6895-902.
[http://dx.doi.org/10.3892/ol.2018.9551] [PMID: 30546421]
[5]
Gao J, Zhang J, Long Y, Tian Y, Lu X. Expression of tankyrase 1 in gastric cancer and its correlation with telomerase activity. Pathol Oncol Res 2011; 17(3): 685-90.
[http://dx.doi.org/10.1007/s12253-011-9369-8] [PMID: 21455637]
[6]
Gelmini S, Quattrone S, Malentacchi F, et al. Tankyrase-1 mRNA expression in bladder cancer and paired urine sediment: Preliminary experience. Clin Chem Lab Med 2007; 45(7): 862-6.
[http://dx.doi.org/10.1515/CCLM.2007.133] [PMID: 17617028]
[7]
La Torre D, Conti A, Aguennouz MH, et al. Telomere length modulation in human astroglial brain tumors. PLoS One 2013; 8(5): e64296.
[http://dx.doi.org/10.1371/journal.pone.0064296] [PMID: 23691191]
[8]
Tang B, Wang J, Fang J, et al. No texpression of tnks1 is correlated with pathologic grade and wnt/-catenin pathway in human astrocytomasitle. 2012; 139-43.
[9]
Zhao F, Vermeer B, Lehmann U, et al. Identification of a novel murine pancreatic tumour antigen, which elicits antibody responses in patients with pancreatic carcinoma. Immunology 2009; 128(1): 134-40.
[http://dx.doi.org/10.1111/j.1365-2567.2009.03090.x] [PMID: 19689742]
[10]
Gelmini S, Poggesi M, Distante V, et al. Tankyrase, a positive regulator of telomere elongation, is over expressed in human breast cancer. Cancer Lett 2004; 216(1): 81-7.
[http://dx.doi.org/10.1016/j.canlet.2004.05.010] [PMID: 15500951]
[11]
Gelmini S, Poggesi M, Pinzani P, et al. Distribution of Tankyrase-1 mRNA expression in colon cancer and its prospective correlation with progression stage. Oncol Rep 2006; 16(6): 1261-6.
[http://dx.doi.org/10.3892/or.16.6.1261] [PMID: 17089047]
[12]
Shebzukhov YV, Lavrik IN, Karbach J, et al. Human tankyrases are aberrantly expressed in colon tumors and contain multiple epitopes that induce humoral and cellular immune responses in cancer patients. Cancer Immunol Immunother 2008; 57(6): 871-81.
[http://dx.doi.org/10.1007/s00262-007-0423-z] [PMID: 18026951]
[13]
Sharma HW, Maltese JY, Zhu X, Kaiser HE, Narayanan R. Telomeres, telomerase and cancer: Is the magic bullet real? Anticancer Res 1996; 16(1): 511-5.
[PMID: 8615664]
[14]
Orlando C, Gelmini S. Telomerase in endocrine and endocrine-dependent tumors. J Steroid Biochem Mol Biol 2001; 78(3): 201-14.
[http://dx.doi.org/10.1016/S0960-0760(01)00101-7] [PMID: 11595501]
[15]
Herbert BS, Wright WE, Shay JW. Telomerase and breast cancer. Breast Cancer Res 2001; 3(3): 146-9.
[http://dx.doi.org/10.1186/bcr288] [PMID: 11305948]
[16]
Hsiao SJ, Smith S. Tankyrase function at telomeres, spindle poles, and beyond. Biochimie 2008; 90(1): 83-92.
[http://dx.doi.org/10.1016/j.biochi.2007.07.012] [PMID: 17825467]
[17]
Ha GH, Kim HS, Go H, et al. Tankyrase-1 function at telomeres and during mitosis is regulated by Polo-like kinase-1-mediated phosphorylation. Cell Death Differ 2012; 19(2): 321-32.
[http://dx.doi.org/10.1038/cdd.2011.101] [PMID: 21818122]
[18]
Yan Y, Lackner MR. FOXO3a and β-catenin co-localization: Double trouble in colon cancer? Nat Med 2012; 18(6): 854-6.
[http://dx.doi.org/10.1038/nm.2799] [PMID: 22673992]
[19]
Pai SG, Carneiro BA, Mota JM, et al. Wnt/beta-catenin pathway: Modulating anticancer immune response. J Hematol Oncol 2017; 10(1): 101.
[http://dx.doi.org/10.1186/s13045-017-0471-6] [PMID: 28476164]
[20]
Mariotti L, Pollock K, Guettler S. Regulation of Wnt/β-catenin signalling by tankyrase-dependent poly(ADP-ribosyl)ation and scaffolding. Br J Pharmacol 2017; 174(24): 4611-36.
[http://dx.doi.org/10.1111/bph.14038] [PMID: 28910490]
[21]
von Kries JP, Winbeck G, Asbrand C, et al. Hot spots in β-catenin for interactions with LEF-1, conductin and APC. Nat Struct Biol 2000; 7(9): 800-7.
[http://dx.doi.org/10.1038/79039] [PMID: 10966653]
[22]
Mazieres J, He B, You L, Xu Z, Jablons DM. Wnt signaling in lung cancer. Cancer Lett 2005; 222(1): 1-10.
[http://dx.doi.org/10.1016/j.canlet.2004.08.040] [PMID: 15837535]
[23]
Uematsu K, He B, You L, Xu Z, McCormick F, Jablons DM. Activation of the Wnt pathway in non small cell lung cancer: Evidence of dishevelled overexpression. Oncogene 2003; 22(46): 7218-21.
[http://dx.doi.org/10.1038/sj.onc.1206817] [PMID: 14562050]
[24]
Peters , Xylia Q. Zoning in on Tankyrases: A brief review on the past, present and prospective studies. Anti-cancer agents in medicinal chemistry (formerly current medicinal chemistry-anti- cancer agents) 2019; 16: 1920-34.
[25]
Levaot N, Voytyuk O, Dimitriou I, et al. Loss of Tankyrase-mediated destruction of 3BP2 is the underlying pathogenic mechanism of cherubism. Cell 2011; 147(6): 1324-39.
[http://dx.doi.org/10.1016/j.cell.2011.10.045] [PMID: 22153076]
[26]
Voronkov A, Krauss S. Wnt/beta-catenin signaling and small molecule inhibitors. Curr Pharm Des 2013; 19(4): 634-64.
[http://dx.doi.org/10.2174/138161213804581837] [PMID: 23016862]
[27]
Guettler S, LaRose J, Petsalaki E, et al. Structural basis and sequence rules for substrate recognition by Tankyrase explain the basis for cherubism disease. Cell 2011; 147(6): 1340-54.
[http://dx.doi.org/10.1016/j.cell.2011.10.046] [PMID: 22153077]
[28]
Zhang Y, Liu S, Mickanin C, et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat Cell Biol 2011; 13(5): 623-9.
[http://dx.doi.org/10.1038/ncb2222] [PMID: 21478859]
[29]
SEIMIYA Hiroyuki. Tankyrase 1 as a target for telomere-directed molecular cancer therapeutics. Cancer cell 2014; 1: 25-37.
[30]
Kim M K, Smith S. Persistent telomere cohesion triggers a prolonged anaphase. Mol Biol Cell 2014; 25(1): 30-40.
[31]
Liu S, Chen X, Chen R, et al. Diagnostic role of Wnt pathway gene promoter methylation in non small cell lung cancer. Oncotarget 2017; 8(22): 36354-67.
[http://dx.doi.org/10.18632/oncotarget.16754] [PMID: 28422739]
[32]
Steinhagen H. Igniting small-molecule drug discovery. ChemMedChem 2016; 11(2): 148-9.
[http://dx.doi.org/10.1002/cmdc.201500580] [PMID: 26781775]
[33]
Short B, Preisinger C, Körner R, Kopajtich R, Byron O, Barr FAA. A GRASP55-rab2 effector complex linking Golgi structure to membrane traffic. J Cell Biol 2001; 155(6): 877-83.
[http://dx.doi.org/10.1083/jcb.200108079] [PMID: 11739401]
[34]
Sbodio JI, Chi NW. Identification of a tankyrase-binding motif shared by IRAP, TAB182, and human TRF1 but not mouse TRF1. NuMA contains this RXXPDG motif and is a novel tankyrase partner. J Biol Chem 2002; 277(35): 31887-92.
[http://dx.doi.org/10.1074/jbc.M203916200] [PMID: 12080061]
[35]
Zamudio-Martinez E, Herrera-Campos AB, Muñoz A, Rodríguez- Vargas JM, Oliver FJ. Tankyrases as modulators of pro-tumoral functions: Molecular insights and therapeutic opportunities. J Exp Clin Cancer Res 2021; 40(1): 144.
[http://dx.doi.org/10.1186/s13046-021-01950-6] [PMID: 33910596]
[36]
Chen G, Dimitriou ID, La Rose J, et al. The 3BP2 adapter protein is required for optimal B-cell activation and thymus-independent type 2 humoral response. Mol Cell Biol 2007; 27(8): 3109-22.
[http://dx.doi.org/10.1128/MCB.01014-06] [PMID: 17283041]
[37]
Eisemann T, McCauley M, Langelier M-F, et al. Tankyrase-1 ankyrin repeats form an adaptable binding platform for targets of adp-ribose modification. Structure 2016; 24(10): 1679-92.
[http://dx.doi.org/10.1016/j.str.2016.07.014] [PMID: 27594684]
[38]
Lehtiö L, Chi N-W, Krauss S. Tankyrases as drug targets. FEBS J 2013; 280(15): 3576-93.
[http://dx.doi.org/10.1111/febs.12320] [PMID: 23648170]
[39]
Guo H-L, Zhang C, Liu Q, et al. The Axin/TNKS complex interacts with KIF3A and is required for insulin-stimulated GLUT4 translocation. Cell Res 2012; 22(8): 1246-57.
[http://dx.doi.org/10.1038/cr.2012.52] [PMID: 22473005]
[40]
Chi NW, Lodish HF. Tankyrase is a golgi-associated mitogen-activated protein kinase substrate that interacts with IRAP in GLUT4 vesicles. J Biol Chem 2000; 275(49): 38437-44.
[http://dx.doi.org/10.1074/jbc.M007635200] [PMID: 10988299]
[41]
Chang W, Dynek JN, Smith S. NuMA is a major acceptor of poly(ADP-ribosyl)ation by tankyrase 1 in mitosis. Biochem J 2005; 391(Pt 2): 177-84.
[http://dx.doi.org/10.1042/BJ20050885] [PMID: 16076287]
[42]
Lau T, Chan E, Callow M, et al. A novel tankyrase small-molecule inhibitor suppresses APC mutation-driven colorectal tumor growth. Cancer Res 2013; 73(10): 3132-44.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-4562] [PMID: 23539443]
[43]
Arqués O, Chicote I, Puig I, et al. Tankyrase inhibition blocks wnt/β-catenin pathway and reverts resistance to pi3k and akt inhibitors in the treatment of colorectal cancer. Clin Cancer Res 2016; 22(3): 644-56.
[http://dx.doi.org/10.1158/1078-0432.CCR-14-3081] [PMID: 26224873]
[44]
Bao R, Christova T, Song S, Angers S, Yan X, Attisano L. Inhibition of tankyrases induces Axin stabilization and blocks Wnt signalling in breast cancer cells. PLoS One 2012; 7(11): e48670.
[http://dx.doi.org/10.1371/journal.pone.0048670] [PMID: 23144924]
[45]
Quackenbush KS, Bagby S, Tai WM, et al. The novel tankyrase inhibitor (AZ1366) enhances irinotecan activity in tumors that exhibit elevated tankyrase and irinotecan resistance. Oncotarget 2016; 7(19): 28273-85.
[http://dx.doi.org/10.18632/oncotarget.8626] [PMID: 27070088]
[46]
Li B, Liang J, Lu F, et al. Discovery of novel inhibitor for WNT/β-catenin pathway by tankyrase 1/2 structure- based virtual screening. Molecules 2020; 25(7): 1680.
[http://dx.doi.org/10.3390/molecules25071680] [PMID: 32268564]
[47]
Tian X, Hou W, Bai S, Fan J, Tong H, Xu H. XAV939 inhibits the stemness and migration of neuroblastoma cancer stem cells via repression of tankyrase 1. Int J Oncol 2014; 45(1): 121-8.
[http://dx.doi.org/10.3892/ijo.2014.2406] [PMID: 24789807]
[48]
Iwai Y, Hamanishi J, Chamoto K, Honjo T. Cancer immunotherapies targeting the PD-1 signaling pathway. J Biomed Sci 2017; 24(1): 26.
[http://dx.doi.org/10.1186/s12929-017-0329-9] [PMID: 28376884]
[49]
Tai D, Wells K, Arcaroli J, et al. Targeting the WNT signaling pathway in cancer therapeutics. Oncologist 2015; 20(10): 1189-98.
[http://dx.doi.org/10.1634/theoncologist.2015-0057] [PMID: 26306903]
[50]
Roelink H, Wagenaar E, Lopes da Silva S, Nusse R. Wnt-3, a gene activated by proviral insertion in mouse mammary tumors, is homologous to int-1/Wnt-1 and is normally expressed in mouse embryos and adult brain. Proc Natl Acad Sci USA 1990; 87(12): 4519-23.
[http://dx.doi.org/10.1073/pnas.87.12.4519] [PMID: 2162045]
[51]
Yang J-M, Huang H-M, Cheng J-J, et al. LGK974, a PORCUPINE inhibitor, mitigates cytotoxicity in an in vitro model of Parkinson’s disease by interfering with the WNT/β-CATENIN pathway. Toxicology 2018; 410: 65-72.
[http://dx.doi.org/10.1016/j.tox.2018.09.003] [PMID: 30205152]
[52]
Pavlovic Z, Adams JJ, Blazer LL, et al. A synthetic anti-Frizzled antibody engineered for broadened specificity exhibits enhanced anti-tumor properties. MAbs 2018; 10(8): 1157-67.
[http://dx.doi.org/10.1080/19420862.2018.1515565] [PMID: 30183492]
[53]
Huang S-MA, Mishina YM, Liu S, et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009; 461(7264): 614-20.
[http://dx.doi.org/10.1038/nature08356] [PMID: 19759537]
[54]
Stratford EW, Daffinrud J, Munthe E, et al. The tankyrase-specific inhibitor JW74 affects cell cycle progression and induces apoptosis and differentiation in osteosarcoma cell lines. Cancer Med 2014; 3(1): 36-46.
[http://dx.doi.org/10.1002/cam4.170] [PMID: 24403055]
[55]
Tian X-H, Hou WJ, Fang Y, et al. XAV939, a tankyrase 1 inhibitior, promotes cell apoptosis in neuroblastoma cell lines by inhibiting Wnt/β-catenin signaling pathway. J Exp Clin Cancer Res 2013; 32(1): 100.
[http://dx.doi.org/10.1186/1756-9966-32-100] [PMID: 24308762]
[56]
Scarborough HA, Helfrich BA, Casás-Selves M, et al. AZ1366: An inhibitor of tankyrase and the canonical Wnt pathway that limits the persistence of non–small cell lung cancer cells following EGFR inhibition. Clin Cancer Res 2017; 23(6): 1531-41.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-1179] [PMID: 27663586]
[57]
Busch AM, Johnson KC, Stan RV, et al. Evidence for tankyrases as antineoplastic targets in lung cancer. BMC Cancer 2013; 13(1): 211.
[http://dx.doi.org/10.1186/1471-2407-13-211] [PMID: 23621985]
[58]
Mizutani A, Yashiroda Y, Muramatsu Y, et al. RK-287107, a potent and specific tankyrase inhibitor, blocks colorectal cancer cell growth in a preclinical model. Cancer Sci 2018; 109(12): 4003-14.
[http://dx.doi.org/10.1111/cas.13805] [PMID: 30238564]
[59]
Waaler J, Machon O, Tumova L, et al. A novel tankyrase inhibitor decreases canonical Wnt signaling in colon carcinoma cells and reduces tumor growth in conditional APC mutant mice. Cancer Res 2012; 72(11): 2822-32.
[http://dx.doi.org/10.1158/0008-5472.CAN-11-3336] [PMID: 22440753]
[60]
Shultz MD, Cheung AK, Kirby CA, et al. Identification of NVP-TNKS656: The use of structure-efficiency relationships to generate a highly potent, selective, and orally active tankyrase inhibitor. J Med Chem 2013; 56(16): 6495-511.
[http://dx.doi.org/10.1021/jm400807n] [PMID: 23844574]
[61]
James RG, et al. WIKI4, a novel inhibitor of tankyrase and Wnt/ss-catenin signaling. PloS one 2012; 12: e50457.
[62]
Thomson DW, Wagner AJ, Bantscheff M, et al. Discovery of a highly selective tankyrase inhibitor displaying growth inhibition effects against a diverse range of tumor derived cell lines. J Med Chem 2017; 60(13): 5455-71.
[http://dx.doi.org/10.1021/acs.jmedchem.7b00137] [PMID: 28591512]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy