Targeting Telomerase and ATRX/DAXX Inducing Tumor Senescence and Apoptosis in the Malignant Glioma
Abstract
1. Introduction
2. Clinical Characteristics of GBM
3. Risk Factors
4. Etiology and Pathogenesis of GBM
4.1. Isocitrate Dehydrogenase (IDH)
4.2. Co-Deletion at Chromosome Regions 1p/19q
4.3. Mutations of H3F3A
4.4. Telomeres and Telomerase
4.5. Alternative Lengthening of Telomeres, α-Thalassemia X-Linkedintellectual Disability, and Death-Domain-Associated Protein
5. Novel and Alternative Therapeutic Options for Treating GBM
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- de Robles, P.; Fiest, K.M.; Frolkis, A.D.; Pringsheim, T.; Atta, C.; St Germaine-Smith, C.; Day, L.; Lam, D.; Jette, N. The worldwide incidence and prevalence of primary brain tumors: A systematic review and meta-analysis. Neuro Oncol. 2015, 17, 776–783. [Google Scholar] [CrossRef] [PubMed]
- Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.M.; Simjee Sh, U. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pac. J. Cancer Prev. 2017, 18, 3–9. [Google Scholar] [PubMed]
- Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K.; Burger, P.C.; Jouvet, A.; Scheithauer, B.W.; Kleihues, P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007, 114, 97–109. [Google Scholar] [CrossRef] [PubMed]
- van den Bent, M.J. Interobserver variation of the histopathological diagnosis in clinical trials on glioma: A clinician’s perspective. Acta Neuropathol. 2010, 120, 297–304. [Google Scholar] [CrossRef]
- Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed]
- CBTRUS. Central Brain Tumor Registry of the United States CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2004–2008; CBTRUS Central Brain Tumor Registry of the United States: Hinsdale, IL, USA, 2014. [Google Scholar]
- Ostrom, Q.T.; Bauchet, L.; Davis, F.G.; Deltour, I.; Fisher, J.L.; Langer, C.E.; Pekmezci, M.; Schwartzbaum, J.A.; Turner, M.C.; Walsh, K.M.; et al. The epidemiology of glioma in adults: A “state of the science” review. Neuro Oncol. 2014, 16, 896–913. [Google Scholar] [CrossRef] [PubMed]
- Chien, L.N.; Gittleman, H.; Ostrom, Q.T.; Hung, K.S.; Sloan, A.E.; Hsieh, Y.C.; Kruchko, C.; Rogers, L.R.; Wang, Y.F.; Chiou, H.Y.; et al. Comparative Brain and Central Nervous System Tumor Incidence and Survival between the United States and Taiwan Based on Population-Based Registry. Front. Public Health 2016, 4, 151. [Google Scholar] [CrossRef]
- Dolecek, T.A.; Propp, J.M.; Stroup, N.E.; Kruchko, C. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro Oncol. 2012, 14 (Suppl. 5), v1–v49. [Google Scholar] [CrossRef]
- Rock, K.; McArdle, O.; Forde, P.; Dunne, M.; Fitzpatrick, D.; O’Neill, B.; Faul, C. A clinical review of treatment outcomes in glioblastoma multiforme—The validation in a non-trial population of the results of a randomised Phase III clinical trial: Has a more radical approach improved survival? Br. J. Radiol. 2012, 85, e729–e733. [Google Scholar] [CrossRef]
- Tamimi, A.F.; Juweid, M. Epidemiology and Outcome of Glioblastoma. In Glioblastoma; De Vleeschouwer, S., Ed.; Codon Publications: Brisbane, Australia, 2017. [Google Scholar]
- Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed]
- Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
- Fan, H.C.; Chi, C.S.; Chang, Y.K.; Tung, M.C.; Lin, S.Z.; Harn, H.J. The Molecular Mechanisms of Plant-Derived Compounds Targeting Brain Cancer. Int. J. Mol. Sci. 2018, 19, 395. [Google Scholar] [CrossRef] [PubMed]
- Ohka, F.; Natsume, A.; Wakabayashi, T. Current trends in targeted therapies for glioblastoma multiforme. Neurol. Res. Int. 2012, 2012, 878425. [Google Scholar] [CrossRef] [PubMed]
- Malmstrom, A.; Gronberg, B.H.; Marosi, C.; Stupp, R.; Frappaz, D.; Schultz, H.; Abacioglu, U.; Tavelin, B.; Lhermitte, B.; Hegi, M.E.; et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: The Nordic randomised, phase 3 trial. Lancet Oncol. 2012, 13, 916–926. [Google Scholar] [CrossRef]
- Available online: https://brainfoundation.org.au/medical-information/c4-brain-tumour/diagnosis-of-brain-tumour (accessed on 12 December 2018).
- Nakada, M.; Kita, D.; Watanabe, T.; Hayashi, Y.; Teng, L.; Pyko, I.V.; Hamada, J. Aberrant signaling pathways in glioma. Cancers 2011, 3, 3242–3278. [Google Scholar] [CrossRef] [PubMed]
- Agnihotri, S.; Burrell, K.E.; Wolf, A.; Jalali, S.; Hawkins, C.; Rutka, J.T.; Zadeh, G. Glioblastoma, a brief review of history, molecular genetics, animal models and novel therapeutic strategies. Arch. Immunol. Ther. Exp. (Warsz) 2013, 61, 25–41. [Google Scholar] [CrossRef]
- Adamson, C.; Kanu, O.O.; Mehta, A.I.; Di, C.; Lin, N.; Mattox, A.K.; Bigner, D.D. Glioblastoma multiforme: A review of where we have been and where we are going. Expert Opin. Investig. Drugs 2009, 18, 1061–1083. [Google Scholar] [CrossRef]
- Fisher, J.L.; Schwartzbaum, J.A.; Wrensch, M.; Wiemels, J.L. Epidemiology of brain tumors. Neurol. Clin. 2007, 25, 867–890. [Google Scholar] [CrossRef]
- Inskip, P.D.; Tarone, R.E.; Hatch, E.E.; Wilcosky, T.C.; Shapiro, W.R.; Selker, R.G.; Fine, H.A.; Black, P.M.; Loeffler, J.S.; Linet, M.S. Cellular-telephone use and brain tumors. N. Engl. J. Med. 2001, 344, 79–86. [Google Scholar] [CrossRef]
- Ohgaki, H. Epidemiology of brain tumors. Methods Mol. Biol. 2009, 472, 323–342. [Google Scholar]
- Kabat, G.C.; Etgen, A.M.; Rohan, T.E. Do steroid hormones play a role in the etiology of glioma? Cancer Epidemiol. Biomarkers Prev. 2010, 19, 2421–2427. [Google Scholar] [CrossRef] [PubMed]
- Bondy, M.L.; Scheurer, M.E.; Malmer, B.; Barnholtz-Sloan, J.S.; Davis, F.G.; Il’yasova, D.; Kruchko, C.; McCarthy, B.J.; Rajaraman, P.; Schwartzbaum, J.A.; et al. Brain tumor epidemiology: Consensus from the Brain Tumor Epidemiology Consortium. Cancer 2008, 113 (Suppl. 7), 1953–1968. [Google Scholar] [CrossRef] [PubMed]
- Berg-Beckhoff, G.; Schuz, J.; Blettner, M.; Munster, E.; Schlaefer, K.; Wahrendorf, J.; Schlehofer, B. History of allergic disease and epilepsy and risk of glioma and meningioma (INTERPHONE study group, Germany). Eur. J. Epidemiol. 2009, 24, 433–440. [Google Scholar] [CrossRef] [PubMed]
- Il’yasova, D.; McCarthy, B.; Marcello, J.; Schildkraut, J.M.; Moorman, P.G.; Krishnamachari, B.; Ali-Osman, F.; Bigner, D.D.; Davis, F. Association between glioma and history of allergies, asthma, and eczema: A case-control study with three groups of controls. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 1232–1238. [Google Scholar] [CrossRef]
- McCarthy, B.J.; Rankin, K.; Il’yasova, D.; Erdal, S.; Vick, N.; Ali-Osman, F.; Bigner, D.D.; Davis, F. Assessment of type of allergy and antihistamine use in the development of glioma. Cancer Epidemiol. Biomarkers Prev. 2011, 20, 370–378. [Google Scholar] [CrossRef]
- Turner, M.C.; Krewski, D.; Armstrong, B.K.; Chetrit, A.; Giles, G.G.; Hours, M.; McBride, M.L.; Parent, M.E.; Sadetzki, S.; Siemiatycki, J.; et al. Allergy and brain tumors in the INTERPHONE study: Pooled results from Australia, Canada, France, Israel, and New Zealand. Cancer Causes Control 2013, 24, 949–960. [Google Scholar] [CrossRef]
- Linos, E.; Raine, T.; Alonso, A.; Michaud, D. Atopy and risk of brain tumors: A meta-analysis. J. Natl. Cancer Inst. 2007, 99, 1544–1550. [Google Scholar] [CrossRef]
- Mulvihill, J.J.; Parry, D.M.; Sherman, J.L.; Pikus, A.; Kaiser-Kupfer, M.I.; Eldridge, R. NIH conference. Neurofibromatosis 1 (Recklinghausen disease) and neurofibromatosis 2 (bilateral acoustic neurofibromatosis). An update. Ann. Intern. Med. 1990, 113, 39–52. [Google Scholar] [CrossRef]
- Cristofori, E.; Colvecchio, A.; Rescaldani, R.; Micoli, G. Bilateral renal angiomyoliposarcoma and glioblastoma multiforme in tuberous sclerosis. Folia Hered. Pathol. (Milano) 1968, 17, 79–83. [Google Scholar]
- Therkildsen, C.; Ladelund, S.; Rambech, E.; Persson, A.; Petersen, A.; Nilbert, M. Glioblastomas, astrocytomas and oligodendrogliomas linked to Lynch syndrome. Eur. J. Neurol. 2015, 22, 717–724. [Google Scholar] [CrossRef]
- Bahuau, M.; Vidaud, D.; Jenkins, R.B.; Bieche, I.; Kimmel, D.W.; Assouline, B.; Smith, J.S.; Alderete, B.; Cayuela, J.M.; Harpey, J.P.; et al. Germ-line deletion involving the INK4 locus in familial proneness to melanoma and nervous system tumors. Cancer Res. 1998, 58, 2298–2303. [Google Scholar] [PubMed]
- Mellon, C.D.; Carter, J.E.; Owen, D.B. Ollier’s disease and Maffucci’s syndrome: Distinct entities or a continuum. Case report: Enchondromatosis complicated by an intracranial glioma. J. Neurol. 1988, 235, 376–378. [Google Scholar] [CrossRef] [PubMed]
- Lynch, H.T.; McComb, R.D.; Osborn, N.K.; Wolpert, P.A.; Lynch, J.F.; Wszolek, Z.K.; Sidransky, D.; Steg, R.E. Predominance of brain tumors in an extended Li-Fraumeni (SBLA) kindred, including a case of Sturge-Weber syndrome. Cancer 2000, 88, 433–439. [Google Scholar] [CrossRef]
- Sanson, M.; Hosking, F.J.; Shete, S.; Zelenika, D.; Dobbins, S.E.; Ma, Y.; Enciso-Mora, V.; Idbaih, A.; Delattre, J.Y.; Hoang-Xuan, K.; et al. Chromosome 7p11.2 (EGFR) variation influences glioma risk. Hum. Mol. Genet. 2011, 20, 2897–2904. [Google Scholar] [CrossRef] [PubMed]
- Shete, S.; Hosking, F.J.; Robertson, L.B.; Dobbins, S.E.; Sanson, M.; Malmer, B.; Simon, M.; Marie, Y.; Boisselier, B.; Delattre, J.Y.; et al. Genome-wide association study identifies five susceptibility loci for glioma. Nat. Genet. 2009, 41, 899–904. [Google Scholar] [CrossRef] [PubMed]
- Stacey, S.N.; Sulem, P.; Jonasdottir, A.; Masson, G.; Gudmundsson, J.; Gudbjartsson, D.F.; Magnusson, O.T.; Gudjonsson, S.A.; Sigurgeirsson, B.; Thorisdottir, K.; et al. A germline variant in the TP53 polyadenylation signal confers cancer susceptibility. Nat. Genet. 2011, 43, 1098–1103. [Google Scholar] [CrossRef]
- Walsh, K.M.; Codd, V.; Smirnov, I.V.; Rice, T.; Decker, P.A.; Hansen, H.M.; Kollmeyer, T.; Kosel, M.L.; Molinaro, A.M.; McCoy, L.S.; et al. Variants near TERT and TERC influencing telomere length are associated with high-grade glioma risk. Nat. Genet. 2014, 46, 731–735. [Google Scholar] [CrossRef]
- Wrensch, M.; Jenkins, R.B.; Chang, J.S.; Yeh, R.F.; Xiao, Y.; Decker, P.A.; Ballman, K.V.; Berger, M.; Buckner, J.C.; Chang, S.; et al. Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility. Nat. Genet. 2009, 41, 905–908. [Google Scholar] [CrossRef]
- Eckel-Passow, J.E.; Lachance, D.H.; Molinaro, A.M.; Walsh, K.M.; Decker, P.A.; Sicotte, H.; Pekmezci, M.; Rice, T.; Kosel, M.L.; Smirnov, I.V.; et al. Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors. N. Engl. J. Med. 2015, 372, 2499–2508. [Google Scholar] [CrossRef]
- Kannan, K.; Inagaki, A.; Silber, J.; Gorovets, D.; Zhang, J.; Kastenhuber, E.R.; Heguy, A.; Petrini, J.H.; Chan, T.A.; Huse, J.T. Whole-exome sequencing identifies ATRX mutation as a key molecular determinant in lower-grade glioma. Oncotarget 2012, 3, 1194–1203. [Google Scholar] [CrossRef]
- Killela, P.J.; Reitman, Z.J.; Jiao, Y.; Bettegowda, C.; Agrawal, N.; Diaz, L.A.; Jr Friedman, A.H.; Friedman, H.; Gallia, G.L.; Giovanella, B.C.; et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc. Natl. Acad. Sci. USA 2013, 110, 6021–6026. [Google Scholar] [CrossRef] [PubMed]
- Koelsche, C.; Sahm, F.; Capper, D.; Reuss, D.; Sturm, D.; Jones, D.T.; Kool, M.; Northcott, P.A.; Wiestler, B.; Bohmer, K.; et al. Distribution of TERT promoter mutations in pediatric and adult tumors of the nervous system. Acta Neuropathol. 2013, 126, 907–915. [Google Scholar] [CrossRef] [PubMed]
- Parsons, D.W.; Jones, S.; Zhang, X.; Lin, J.C.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Siu, I.M.; Gallia, G.L.; et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008, 321, 1807–1812. [Google Scholar] [CrossRef] [PubMed]
- Yan, H.; Parsons, D.W.; Jin, G.; McLendon, R.; Rasheed, B.A.; Yuan, W.; Kos, I.; Batinic-Haberle, I.; Jones, S.; Riggins, G.J.; et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 2009, 360, 765–773. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Zhao, J.; Xu, Z.; Peng, B.; Huang, Q.; Arnold, E.; Ding, J. Structures of human cytosolic NADP-dependent isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J. Biol. Chem. 2004, 279, 33946–33957. [Google Scholar] [CrossRef] [PubMed]
- Joseph, J.W.; Jensen, M.V.; Ilkayeva, O.; Palmieri, F.; Alarcon, C.; Rhodes, C.J.; Newgard, C.B. The mitochondrial citrate/isocitrate carrier plays a regulatory role in glucose-stimulated insulin secretion. J. Biol. Chem. 2006, 281, 35624–35632. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.H.; Jo, S.H.; Lee, S.M.; Koh, H.J.; Song, H.; Park, J.W.; Lee, W.H.; Huh, T.L. Role of NADP+-dependent isocitrate dehydrogenase (NADP+-ICDH) on cellular defence against oxidative injury by gamma-rays. Int. J. Radiat. Biol. 2004, 80, 635–642. [Google Scholar] [CrossRef] [PubMed]
- Ward, P.S.; Patel, J.; Wise, D.R.; Abdel-Wahab, O.; Bennett, B.D.; Coller, H.A.; Cross, J.R.; Fantin, V.R.; Hedvat, C.V.; Perl, A.E.; et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010, 17, 225–234. [Google Scholar] [CrossRef] [PubMed]
- Figueroa, M.E.; Abdel-Wahab, O.; Lu, C.; Ward, P.S.; Patel, J.; Shih, A.; Li, Y.; Bhagwat, N.; Vasanthakumar, A.; Fernandez, H.F.; et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010, 18, 553–567. [Google Scholar] [CrossRef]
- Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar] [CrossRef]
- Sturm, D.; Witt, H.; Hovestadt, V.; Khuong-Quang, D.A.; Jones, D.T.; Konermann, C.; Pfaff, E.; Tonjes, M.; Sill, M.; Bender, S.; et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 2012, 22, 425–437. [Google Scholar] [CrossRef] [PubMed]
- Cairncross, J.G.; Ueki, K.; Zlatescu, M.C.; Lisle, D.K.; Finkelstein, D.M.; Hammond, R.R.; Silver, J.S.; Stark, P.C.; Macdonald, D.R.; Ino, Y.; et al. Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J. Natl. Cancer Inst. 1998, 90, 1473–1479. [Google Scholar] [CrossRef]
- Kraus, J.A.; Koopmann, J.; Kaskel, P.; Maintz, D.; Brandner, S.; Schramm, J.; Louis, D.N.; Wiestler, O.D.; von Deimling, A. Shared allelic losses on chromosomes 1p and 19q suggest a common origin of oligodendroglioma and oligoastrocytoma. J. Neuropathol. Exp. Neurol. 1995, 54, 91–95. [Google Scholar] [CrossRef] [PubMed]
- Polivka, J., Jr.; Polivka, J.; Repik, T.; Rohan, V.; Hes, O.; Topolcan, O. Co-deletion of 1p/19q as Prognostic and Predictive Biomarker for Patients in West Bohemia with Anaplastic Oligodendroglioma. Anticancer Res. 2016, 36, 471–476. [Google Scholar] [PubMed]
- Reifenberger, J.; Reifenberger, G.; Liu, L.; James, C.D.; Wechsler, W.; Collins, V.P. Molecular genetic analysis of oligodendroglial tumors shows preferential allelic deletions on 19q and 1p. Am. J. Pathol. 1994, 145, 1175–1190. [Google Scholar] [PubMed]
- Bettegowda, C.; Agrawal, N.; Jiao, Y.; Sausen, M.; Wood, L.D.; Hruban, R.H.; Rodriguez, F.J.; Cahill, D.P.; McLendon, R.; Riggins, G.; et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science 2011, 333, 1453–1455. [Google Scholar] [CrossRef] [PubMed]
- Jiao, Y.; Killela, P.J.; Reitman, Z.J.; Rasheed, A.B.; Heaphy, C.M.; de Wilde, R.F.; Rodriguez, F.J.; Rosemberg, S.; Oba-Shinjo, S.M.; Nagahashi Marie, S.K.; et al. Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget 2012, 3, 709–722. [Google Scholar] [CrossRef]
- Labussiere, M.; Idbaih, A.; Wang, X.W.; Marie, Y.; Boisselier, B.; Falet, C.; Paris, S.; Laffaire, J.; Carpentier, C.; Criniere, E.; et al. All the 1p19q codeleted gliomas are mutated on IDH1 or IDH2. Neurology 2010, 74, 1886–1890. [Google Scholar] [CrossRef]
- Duan, J.; Bao, X.; Ma, X.; Zhang, Y.; Ni, D.; Wang, H.; Zhang, F.; Du, Q.; Fan, Y.; Chen, J.; et al. Upregulation of Far Upstream Element-Binding Protein 1 (FUBP1) Promotes Tumor Proliferation and Tumorigenesis of Clear Cell Renal Cell Carcinoma. PLoS ONE 2017, 12, e0169852. [Google Scholar] [CrossRef]
- Han, F.; Zhang, J.; Ma, S.; Chen, X.; Liu, W.; He, X.; Fei, Z.; Wang, Y. Altered capicua transcriptional repressor gene expression exhibits distinct prognostic value for isocitrate dehydrogenase-mutant oligodendroglial tumors. Oncol. Lett. 2018, 15, 1459–1468. [Google Scholar] [CrossRef]
- Cairncross, G.; Wang, M.; Shaw, E.; Jenkins, R.; Brachman, D.; Buckner, J.; Fink, K.; Souhami, L.; Laperriere, N.; Curran, W.; et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: Long-term results of RTOG 9402. J. Clin. Oncol. 2013, 31, 337–343. [Google Scholar] [CrossRef] [PubMed]
- van den Bent, M.J.; Carpentier, A.F.; Brandes, A.A.; Sanson, M.; Taphoorn, M.J.; Bernsen, H.J.; Frenay, M.; Tijssen, C.C.; Grisold, W.; Sipos, L.; et al. Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: A randomized European Organisation for Research and Treatment of Cancer phase III trial. J. Clin. Oncol. 2006, 24, 2715–2722. [Google Scholar] [PubMed]
- Kaneshiro, D.; Kobayashi, T.; Chao, S.T.; Suh, J.; Prayson, R.A. Chromosome 1p and 19q deletions in glioblastoma multiforme. Appl. Immunohistochem. Mol. Morphol. 2009, 17, 512–516. [Google Scholar] [CrossRef] [PubMed]
- Nakamura, M.; Yang, F.; Fujisawa, H.; Yonekawa, Y.; Kleihues, P.; Ohgaki, H. Loss of heterozygosity on chromosome 19 in secondary glioblastomas. J. Neuropathol. Exp. Neurol. 2000, 59, 539–543. [Google Scholar] [CrossRef] [PubMed]
- Ohgaki, H.; Kleihues, P. Genetic pathways to primary and secondary glioblastoma. Am. J. Pathol. 2007, 170, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
- Filipescu, D.; Muller, S.; Almouzni, G. Histone H3 variants and their chaperones during development and disease: Contributing to epigenetic control. Annu. Rev. Cell Dev. Biol. 2014, 30, 615–646. [Google Scholar] [CrossRef] [PubMed]
- Saleem, M.; Abbas, K.; Manan, M.; Ijaz, H.; Ahmed, B.; Ali, M.; Hanif, M.; Farooqi, A.A.; Qadir, M.I. Review-Epigenetic therapy for cancer. Pak. J. Pharm. Sci. 2015, 28, 1023–1032. [Google Scholar] [PubMed]
- Szenker, E.; Ray-Gallet, D.; Almouzni, G. The double face of the histone variant H3.3. Cell Res. 2011, 21, 421–434. [Google Scholar] [CrossRef] [PubMed]
- Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
- Wu, G.; Broniscer, A.; McEachron, T.A.; Lu, C.; Paugh, B.S.; Becksfort, J.; Qu, C.; Ding, L.; Huether, R.; Parker, M.; et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 2012, 44, 251–253. [Google Scholar] [CrossRef]
- Bender, S.; Tang, Y.; Lindroth, A.M.; Hovestadt, V.; Jones, D.T.; Kool, M.; Zapatka, M.; Northcott, P.A.; Sturm, D.; Wang, W.; et al. Reduced H3K27me3 and DNA hypomethylation are major drivers of gene expression in K27M mutant pediatric high-grade gliomas. Cancer Cell 2013, 24, 660–672. [Google Scholar] [CrossRef] [PubMed]
- Buczkowicz, P.; Hoeman, C.; Rakopoulos, P.; Pajovic, S.; Letourneau, L.; Dzamba, M.; Morrison, A.; Lewis, P.; Bouffet, E.; Bartels, U.; et al. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 2014, 46, 451–456. [Google Scholar] [CrossRef]
- Fang, J.; Huang, Y.; Mao, G.; Yang, S.; Rennert, G.; Gu, L.; Li, H.; Li, G.M. Cancer-driving H3G34V/R/D mutations block H3K36 methylation and H3K36me3-MutSalpha interaction. Proc. Natl. Acad. Sci. USA 2018, 115, 9598–9603. [Google Scholar] [CrossRef] [PubMed]
- Bjerke, L.; Mackay, A.; Nandhabalan, M.; Burford, A.; Jury, A.; Popov, S.; Bax, D.A.; Carvalho, D.; Taylor, K.R.; Vinci, M.; et al. Histone H3.3. mutations drive pediatric glioblastoma through upregulation of MYCN. Cancer Discov. 2013, 3, 512–519. [Google Scholar] [CrossRef]
- Martin, C.L.L. DH Telomeres. In Handbook of Human Molecular Evolution; John Wiley & Sons: Hoboken, NJ, USA, 2008; pp. 721–724. [Google Scholar]
- Ruiz-Herrera, A.; Nergadze, S.G.; Santagostino, M.; Giulotto, E. Telomeric repeats far from the ends: Mechanisms of origin and role in evolution. Cytogenet. Genome Res. 2008, 122, 219–228. [Google Scholar] [CrossRef] [PubMed]
- Liu, D.; O’Connor, M.S.; Qin, J.; Songyang, Z. Telosome, a mammalian telomere-associated complex formed by multiple telomeric proteins. J. Biol. Chem. 2004, 279, 51338–51342. [Google Scholar] [CrossRef]
- Blasco, M.A. Telomere length, stem cells and aging. Nat. Chem. Biol. 2007, 3, 640–649. [Google Scholar] [CrossRef]
- McClintock, B. The Stability of Broken Ends of Chromosomes in Zea Mays. Genetics 1941, 26, 234–282. [Google Scholar]
- O’Sullivan, R.J.; Karlseder, J. Telomeres: Protecting chromosomes against genome instability. Nat. Rev. Mol. Cell Biol. 2010, 11, 171–181. [Google Scholar] [CrossRef]
- Gong, J.G.; Costanzo, A.; Yang, H.Q.; Melino, G.; Kaelin, W.G., Jr.; Levrero, M.; Wang, J.Y. The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature 1999, 399, 806–809. [Google Scholar] [CrossRef]
- Stiewe, T.; Putzer, B.M. p73 in apoptosis. Apoptosis 2001, 6, 447–452. [Google Scholar] [CrossRef] [PubMed]
- Vaziri, H.; Dragowska, W.; Allsopp, R.C.; Thomas, T.E.; Harley, C.B.; Lansdorp, P.M. Evidence for a mitotic clock in human hematopoietic stem cells: Loss of telomeric DNA with age. Proc. Natl. Acad. Sci. USA 1994, 91, 9857–9860. [Google Scholar] [CrossRef] [PubMed]
- Brouilette, S.; Singh, R.K.; Thompson, J.R.; Goodall, A.H.; Samani, N.J. White cell telomere length and risk of premature myocardial infarction. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 842–846. [Google Scholar] [CrossRef] [PubMed]
- Valdes, A.M.; Andrew, T.; Gardner, J.P.; Kimura, M.; Oelsner, E.; Cherkas, L.F.; Aviv, A.; Spector, T.D. Obesity, cigarette smoking, and telomere length in women. Lancet 2005, 366, 662–664. [Google Scholar] [CrossRef]
- Frenck, R.W., Jr.; Blackburn, E.H.; Shannon, K.M. The rate of telomere sequence loss in human leukocytes varies with age. Proc. Natl. Acad. Sci. USA 1998, 95, 5607–5610. [Google Scholar] [CrossRef] [PubMed]
- Dalgard, C.; Benetos, A.; Verhulst, S.; Labat, C.; Kark, J.D.; Christensen, K.; Kimura, M.; Kyvik, K.O.; Aviv, A. Leukocyte telomere length dynamics in women and men: Menopause vs age effects. Int. J. Epidemiol. 2015, 44, 1688–1695. [Google Scholar] [CrossRef] [PubMed]
- Benetti, R.; Garcia-Cao, M.; Blasco, M.A. Telomere length regulates the epigenetic status of mammalian telomeres and subtelomeres. Nat. Genet. 2007, 39, 243–250. [Google Scholar] [CrossRef] [PubMed]
- Celli, G.B.; de Lange, T. DNA processing is not required for ATM-mediated telomere damage response after TRF2 deletion. Nat. Cell Biol. 2005, 7, 712–718. [Google Scholar] [CrossRef]
- Steinert, S.; Shay, J.W.; Wright, W.E. Modification of subtelomeric DNA. Mol. Cell. Biol. 2004, 24, 4571–4580. [Google Scholar] [CrossRef]
- Adams, J.; Martin-Ruiz, C.; Pearce, M.S.; White, M.; Parker, L.; von Zglinicki, T. No association between socio-economic status and white blood cell telomere length. Aging Cell 2007, 6, 125–128. [Google Scholar] [CrossRef]
- Cherkas, L.F.; Hunkin, J.L.; Kato, B.S.; Richards, J.B.; Gardner, J.P.; Surdulescu, G.L.; Kimura, M.; Lu, X.; Spector, T.D.; Aviv, A. The association between physical activity in leisure time and leukocyte telomere length. Arch. Intern. Med. 2008, 168, 154–158. [Google Scholar] [CrossRef] [PubMed]
- Nordfjall, K.; Eliasson, M.; Stegmayr, B.; Melander, O.; Nilsson, P.; Roos, G. Telomere length is associated with obesity parameters but with a gender difference. Obesity 2008, 16, 2682–2689. [Google Scholar] [CrossRef] [PubMed]
- van der Harst, P.; van der Steege, G.; de Boer, R.A.; Voors, A.A.; Hall, A.S.; Mulder, M.J.; van Gilst, W.H.; van Veldhuisen, D.J.; MERIT-HF Study Group. Telomere length of circulating leukocytes is decreased in patients with chronic heart failure. J. Am. Coll. Cardiol. 2007, 49, 1459–1464. [Google Scholar] [CrossRef]
- Brouilette, S.W.; Moore, J.S.; McMahon, A.D.; Thompson, J.R.; Ford, I.; Shepherd, J.; Packard, C.J.; Samani, N.J.; West of Scotland Coronary Prevention Study Group. Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: A nested case-control study. Lancet 2007, 369, 107–114. [Google Scholar] [CrossRef]
- Fitzpatrick, A.L.; Kronmal, R.A.; Gardner, J.P.; Psaty, B.M.; Jenny, N.S.; Tracy, R.P.; Walston, J.; Kimura, M.; Aviv, A. Leukocyte telomere length and cardiovascular disease in the cardiovascular health study. Am. J. Epidemiol. 2007, 165, 14–21. [Google Scholar] [CrossRef] [PubMed]
- Zee, R.Y.; Michaud, S.E.; Germer, S.; Ridker, P.M. Association of shorter mean telomere length with risk of incident myocardial infarction: A prospective, nested case-control approach. Clin. Chim. Acta 2009, 403, 139–141. [Google Scholar] [CrossRef] [PubMed]
- Sampson, M.J.; Winterbone, M.S.; Hughes, J.C.; Dozio, N.; Hughes, D.A. Monocyte telomere shortening and oxidative DNA damage in type 2 diabetes. Diabetes Care 2006, 29, 283–289. [Google Scholar] [CrossRef] [PubMed]
- Valdes, A.M.; Richards, J.B.; Gardner, J.P.; Swaminathan, R.; Kimura, M.; Xiaobin, L.; Aviv, A.; Spector, T.D. Telomere length in leukocytes correlates with bone mineral density and is shorter in women with osteoporosis. Osteoporos. Int. 2007, 18, 1203–1210. [Google Scholar] [CrossRef] [PubMed]
- Cawthon, R.M.; Smith, K.R.; O’Brien, E.; Sivatchenko, A.; Kerber, R.A. Association between telomere length in blood and mortality in people aged 60 years or older. Lancet 2003, 361, 393–395. [Google Scholar] [CrossRef]
- Farzaneh-Far, R.; Cawthon, R.M.; Na, B.; Browner, W.S.; Schiller, N.B.; Whooley, M.A. Prognostic value of leukocyte telomere length in patients with stable coronary artery disease: Data from the Heart and Soul Study. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 1379–1384. [Google Scholar] [CrossRef]
- Yang, Z.; Huang, X.; Jiang, H.; Zhang, Y.; Liu, H.; Qin, C.; Eisner, G.M.; Jose, P.A.; Rudolph, L.; Ju, Z. Short telomeres and prognosis of hypertension in a chinese population. Hypertension 2009, 53, 639–645. [Google Scholar] [CrossRef] [PubMed]
- Chin, L.; Artandi, S.E.; Shen, Q.; Tam, A.; Lee, S.L.; Gottlieb, G.J.; Greider, C.W.; DePinho, R.A. p53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 1999, 97, 527–538. [Google Scholar] [CrossRef]
- De Lange, T. Telomere-related genome instability in cancer. Cold Spring Harb. Symp. Quant. Biol. 2005, 70, 197–204. [Google Scholar] [CrossRef] [PubMed]
- Meeker, A.K. Telomeres and telomerase in prostatic intraepithelial neoplasia and prostate cancer biology. Urol. Oncol. 2006, 24, 122–130. [Google Scholar] [CrossRef] [PubMed]
- Shay, J.W.; Wright, W.E. Role of telomeres and telomerase in cancer. Semin. Cancer Biol. 2011, 21, 349–353. [Google Scholar] [CrossRef]
- Wu, K.; Higashi, N.; Hansen, E.R.; Lund, M.; Bang, K.; Thestrup-Pedersen, K. Telomerase activity is increased and telomere length shortened in T cells from blood of patients with atopic dermatitis and psoriasis. J. Immunol. 2000, 165, 4742–4747. [Google Scholar] [CrossRef]
- Greider, C.W.; Blackburn, E.H. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell 1985, 43, 405–413. [Google Scholar] [CrossRef]
- Moyzis, R.K.; Buckingham, J.M.; Cram, L.S.; Dani, M.; Deaven, L.L.; Jones, M.D.; Meyne, J.; Ratliff, R.L.; Wu, J.R. A highly conserved repetitive DNA sequence, (TTAGGG)n, present at the telomeres of human chromosomes. Proc. Natl. Acad. Sci. USA 1988, 85, 6622–6626. [Google Scholar] [CrossRef]
- Shippen-Lentz, D.; Blackburn, E.H. Functional evidence for an RNA template in telomerase. Science 1990, 247, 546–552. [Google Scholar] [CrossRef]
- Hiyama, K.; Hirai, Y.; Kyoizumi, S.; Akiyama, M.; Hiyama, E.; Piatyszek, M.A.; Shay, J.W.; Ishioka, S.; Yamakido, M. Activation of telomerase in human lymphocytes and hematopoietic progenitor cells. J. Immunol. 1995, 155, 3711–3715. [Google Scholar]
- Kim, N.W.; Piatyszek, M.A.; Prowse, K.R.; Harley, C.B.; West, M.D.; Ho, P.L.; Coviello, G.M.; Wright, W.E.; Weinrich, S.L.; Shay, J.W. Specific association of human telomerase activity with immortal cells and cancer. Science 1994, 266, 2011–2015. [Google Scholar] [CrossRef] [PubMed]
- Meyne, J.; Ratliff, R.L.; Moyzis, R.K. Conservation of the human telomere sequence (TTAGGG)n among vertebrates. Proc. Natl. Acad. Sci. USA 1989, 86, 7049–7053. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Funk, W.D.; Wang, S.S.; Weinrich, S.L.; Avilion, A.A.; Chiu, C.P.; Adams, R.R.; Chang, E.; Allsopp, R.C.; Yu, J.; et al. The RNA component of human telomerase. Science 1995, 269, 1236–1241. [Google Scholar] [CrossRef] [PubMed]
- Mocellin, S.; Pooley, K.A.; Nitti, D. Telomerase and the search for the end of cancer. Trends Mol. Med. 2013, 19, 125–133. [Google Scholar] [CrossRef]
- Smekalova, E.M.; Shubernetskaya, O.S.; Zvereva, M.I.; Gromenko, E.V.; Rubtsova, M.P.; Dontsova, O.A. Telomerase RNA biosynthesis and processing. Biochemistry 2012, 77, 1120–1128. [Google Scholar] [CrossRef]
- Horn, S.; Figl, A.; Rachakonda, P.S.; Fischer, C.; Sucker, A.; Gast, A.; Kadel, S.; Moll, I.; Nagore, E.; Hemminki, K.; et al. TERT promoter mutations in familial and sporadic melanoma. Science 2013, 339, 959–961. [Google Scholar] [CrossRef]
- Huang, F.W.; Hodis, E.; Xu, M.J.; Kryukov, G.V.; Chin, L.; Garraway, L.A. Highly recurrent TERT promoter mutations in human melanoma. Science 2013, 339, 957–959. [Google Scholar] [CrossRef]
- Liu, X.; Wu, G.; Shan, Y.; Hartmann, C.; von Deimling, A.; Xing, M. Highly prevalent TERT promoter mutations in bladder cancer and glioblastoma. Cell Cycle 2013, 12, 1637–1638. [Google Scholar] [CrossRef]
- Arita, H.; Narita, Y.; Takami, H.; Fukushima, S.; Matsushita, Y.; Yoshida, A.; Miyakita, Y.; Ohno, M.; Shibui, S.; Ichimura, K. TERT promoter mutations rather than methylation are the main mechanism for TERT upregulation in adult gliomas. Acta Neuropathol. 2013, 126, 939–941. [Google Scholar] [CrossRef]
- Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
- Nonoguchi, N.; Ohta, T.; Oh, J.E.; Kim, Y.H.; Kleihues, P.; Ohgaki, H. TERT promoter mutations in primary and secondary glioblastomas. Acta Neuropathol. 2013, 126, 931–937. [Google Scholar] [CrossRef] [PubMed]
- Vinagre, J.; Almeida, A.; Populo, H.; Batista, R.; Lyra, J.; Pinto, V.; Coelho, R.; Celestino, R.; Prazeres, H.; Lima, L.; et al. Frequency of TERT promoter mutations in human cancers. Nat. Commun. 2013, 4, 2185. [Google Scholar] [CrossRef] [PubMed]
- Heaphy, C.M.; de Wilde, R.F.; Jiao, Y.; Klein, A.P.; Edil, B.H.; Shi, C.; Bettegowda, C.; Rodriguez, F.J.; Eberhart, C.G.; Hebbar, S.; et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science 2011, 333, 425. [Google Scholar] [CrossRef] [PubMed]
- Schwartzentruber, J.; Korshunov, A.; Liu, X.Y.; Jones, D.T.; Pfaff, E.; Jacob, K.; Sturm, D.; Fontebasso, A.M.; Quang, D.A.; Tonjes, M.; et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 2012, 482, 226–231. [Google Scholar] [CrossRef] [PubMed]
- Molano, M.; Martin, D.C.; Moreno-Acosta, P.; Hernandez, G.; Cornall, A.; Buitrago, O.; Gamboa, O.; Garland, S.; Tabrizi, S.; Munoz, N. Telomerase activity in cervical scrapes of women with high-grade cervical disease: A nested case-control study. Oncol. Lett. 2018, 15, 354–360. [Google Scholar] [CrossRef]
- Wang, N.; Xu, D.; Sofiadis, A.; Hoog, A.; Vukojevic, V.; Backdahl, M.; Zedenius, J.; Larsson, C. Telomerase-dependent and independent telomere maintenance and its clinical implications in medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2014, 99, E1571–E1579. [Google Scholar] [CrossRef] [PubMed]
- Bornstein-Quevedo, L.; Garcia-Hernandez, M.L.; Camacho-Arroyo, I.; Herrera, M.F.; Angeles, A.A.; Trevino, O.G.; Gamboa-Dominguez, A. Telomerase activity in well-differentiated papillary thyroid carcinoma correlates with advanced clinical stage of the disease. Endocr. Pathol. 2003, 14, 213–219. [Google Scholar] [CrossRef]
- Fernandez-Marcelo, T.; Gomez, A.; Pascua, I.; de Juan, C.; Head, J.; Hernando, F.; Jarabo, J.R.; Calatayud, J.; Torres-Garcia, A.J.; Iniesta, P. Telomere length and telomerase activity in non-small cell lung cancer prognosis: Clinical usefulness of a specific telomere status. J. Exp. Clin. Cancer Res. 2015, 34, 78. [Google Scholar] [CrossRef]
- Satyanarayana, A.; Manns, M.P.; Rudolph, K.L. Telomeres and telomerase: A dual role in hepatocarcinogenesis. Hepatology 2004, 40, 276–283. [Google Scholar] [CrossRef]
- Bryan, T.M.; Englezou, A.; Dalla-Pozza, L.; Dunham, M.A.; Reddel, R.R. Evidence for an alternative mechanism for maintaining telomere length in human tumors and tumor-derived cell lines. Nat. Med. 1997, 3, 1271–1274. [Google Scholar] [CrossRef]
- Slatter, T.L.; Tan, X.; Yuen, Y.C.; Gunningham, S.; Ma, S.S.; Daly, E.; Packer, S.; Devenish, C.; Royds, J.A.; Hung, N.A. The alternative lengthening of telomeres pathway may operate in non-neoplastic human cells. J. Pathol. 2012, 226, 509–518. [Google Scholar] [CrossRef] [PubMed]
- Heaphy, C.M.; Subhawong, A.P.; Hong, S.M.; Goggins, M.G.; Montgomery, E.A.; Gabrielson, E.; Netto, G.J.; Epstein, J.I.; Lotan, T.L.; Westra, W.H.; et al. Prevalence of the alternative lengthening of telomeres telomere maintenance mechanism in human cancer subtypes. Am. J. Pathol. 2011, 179, 1608–1615. [Google Scholar] [CrossRef] [PubMed]
- Wiestler, B.; Capper, D.; Holland-Letz, T.; Korshunov, A.; von Deimling, A.; Pfister, S.M.; Platten, M.; Weller, M.; Wick, W. ATRX loss refines the classification of anaplastic gliomas and identifies a subgroup of IDH mutant astrocytic tumors with better prognosis. Acta Neuropathol. 2013, 126, 443–451. [Google Scholar] [CrossRef] [PubMed]
- Flynn, R.L.; Cox, K.E.; Jeitany, M.; Wakimoto, H.; Bryll, A.R.; Ganem, N.J.; Bersani, F.; Pineda, J.R.; Suva, M.L.; Benes, C.H.; et al. Alternative lengthening of telomeres renders cancer cells hypersensitive to ATR inhibitors. Science 2015, 347, 273–277. [Google Scholar] [CrossRef] [PubMed]
- Shay, J.W.; Reddel, R.R.; Wright, W.E. Cancer. Cancer and telomeres—An ALTernative to telomerase. Science 2012, 336, 1388–1390. [Google Scholar] [CrossRef] [PubMed]
- Cesare, A.J.; Reddel, R.R. Alternative lengthening of telomeres: Models, mechanisms and implications. Nat. Rev. Genet. 2010, 11, 319–330. [Google Scholar] [CrossRef] [PubMed]
- Gibbons, R. Alpha thalassaemia-mental retardation, X linked. Orphanet J. Rare Dis. 2006, 1, 15. [Google Scholar] [CrossRef] [PubMed]
- Dyer, M.A.; Qadeer, Z.A.; Valle-Garcia, D.; Bernstein, E. ATRX and DAXX: Mechanisms and Mutations. Cold Spring Harb. Perspect. Med. 2017, 7, a026567. [Google Scholar] [CrossRef]
- Pekmezci, M.; Rice, T.; Molinaro, A.M.; Walsh, K.M.; Decker, P.A.; Hansen, H.; Sicotte, H.; Kollmeyer, T.M.; McCoy, L.S.; Sarkar, G.; et al. Adult infiltrating gliomas with WHO 2016 integrated diagnosis: Additional prognostic roles of ATRX and TERT. Acta Neuropathol. 2017, 133, 1001–1016. [Google Scholar] [CrossRef]
- Clynes, D.; Higgs, D.R.; Gibbons, R.J. The chromatin remodeller ATRX: A repeat offender in human disease. Trends Biochem. Sci. 2013, 38, 461–466. [Google Scholar] [CrossRef]
- Napier, C.E.; Huschtscha, L.I.; Harvey, A.; Bower, K.; Noble, J.R.; Hendrickson, E.A.; Reddel, R.R. ATRX represses alternative lengthening of telomeres. Oncotarget 2015, 6, 16543–16558. [Google Scholar] [CrossRef] [PubMed]
- Clynes, D.; Jelinska, C.; Xella, B.; Ayyub, H.; Taylor, S.; Mitson, M.; Bachrati, C.Z.; Higgs, D.R.; Gibbons, R.J. ATRX dysfunction induces replication defects in primary mouse cells. PLoS ONE 2014, 9, e92915. [Google Scholar] [CrossRef] [PubMed]
- Eid, R.; Demattei, M.V.; Episkopou, H.; Auge-Gouillou, C.; Decottignies, A.; Grandin, N.; Charbonneau, M. Genetic Inactivation of ATRX Leads to a Decrease in the Amount of Telomeric Cohesin and Level of Telomere Transcription in Human Glioma Cells. Mol. Cell. Biol. 2015, 35, 2818–2830. [Google Scholar] [CrossRef] [PubMed]
- Lovejoy, C.A.; Li, W.; Reisenweber, S.; Thongthip, S.; Bruno, J.; de Lange, T.; De, S.; Petrini, J.H.; Sung, P.A.; Jasin, M.; et al. Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway. PLoS Genet. 2012, 8, e1002772. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Khosravi-Far, R.; Chang, H.Y.; Baltimore, D. Daxx, a novel Fas-binding protein that activates JNK and apoptosis. Cell 1997, 89, 1067–1076. [Google Scholar] [CrossRef]
- Drane, P.; Ouararhni, K.; Depaux, A.; Shuaib, M.; Hamiche, A. The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev. 2010, 24, 1253–1265. [Google Scholar] [CrossRef] [PubMed]
- Goldberg, A.D.; Banaszynski, L.A.; Noh, K.M.; Lewis, P.W.; Elsaesser, S.J.; Stadler, S.; Dewell, S.; Law, M.; Guo, X.; Li, X.; et al. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 2010, 140, 678–691. [Google Scholar] [CrossRef]
- Lewis, P.W.; Elsaesser, S.J.; Noh, K.M.; Stadler, S.C.; Allis, C.D. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc. Natl. Acad. Sci. USA 2010, 107, 14075–14080. [Google Scholar] [CrossRef]
- Amorim, J.P.; Santos, G.; Vinagre, J.; Soares, P. The Role of ATRX in the Alternative Lengthening of Telomeres (ALT) Phenotype. Genes 2016, 7, 66. [Google Scholar] [CrossRef]
- Hu, Y.; Shi, G.; Zhang, L.; Li, F.; Jiang, Y.; Jiang, S.; Ma, W.; Zhao, Y.; Songyang, Z.; Huang, J. Switch telomerase to ALT mechanism by inducing telomeric DNA damages and dysfunction of ATRX and DAXX. Sci. Rep. 2016, 6, 32280. [Google Scholar] [CrossRef]
- Available online: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm614128.htm (accessed on 12 December 2018).
- Available online: https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm569482.htm (accessed on 12 December 2018).
- Available online: https://clinicaltrials.gov/ct2/show/NCT02977689 (accessed on 12 December 2018).
- Available online: https://clinicaltrials.gov/ct2/show/NCT02746081 (accessed on 12 December 2018).
- Lee, W.Y.; Chen, K.C.; Chen, H.Y.; Chen, C.Y. Potential mitochondrial isocitrate dehydrogenase R140Q mutant inhibitor from traditional Chinese medicine against cancers. Biomed. Res. Int. 2014, 2014, 364625. [Google Scholar] [CrossRef] [PubMed]
- Parsch, D.; Brassat, U.; Brummendorf, T.H.; Fellenberg, J. Consequences of telomerase inhibition by BIBR1532 on proliferation and chemosensitivity of chondrosarcoma cell lines. Cancer Investig. 2008, 26, 590–596. [Google Scholar] [CrossRef] [PubMed]
- Roth, A.; Harley, C.B.; Baerlocher, G.M. Imetelstat (GRN163L)—Telomerase-based cancer therapy. Recent Results Cancer Res. 2010, 184, 221–234. [Google Scholar] [PubMed]
- Nava-Parada, P.; Emens, L.A. GV-1001, an injectable telomerase peptide vaccine for the treatment of solid cancers. Curr. Opin. Mol. Ther. 2007, 9, 490–497. [Google Scholar] [PubMed]
- Ouellette, M.M.; Wright, W.E.; Shay, J.W. Targeting telomerase-expressing cancer cells. J. Cell. Mol. Med. 2011, 15, 1433–1442. [Google Scholar] [CrossRef]
- Kosmatopoulos KB, E.; Aggouraki, D.; Nikoloudi, E.; Kanellou, P.; Papadimitraki, E.; Kotsakis, A.; Mavroudis, D.; Georgoulias, V. Clinical efficacy and immunogenicity of the optimized cryptic peptide TERT572Y vaccine (Vx-001) in patients with advanced non-small cell lung cancer (NSCLC). In Proceedings of the ASCO Meeting, Atlanta, GA, USA, 2–6 June 2006. [Google Scholar]
- Taetz, S.; Baldes, C.; Murdter, T.E.; Kleideiter, E.; Piotrowska, K.; Bock, U.; Haltner-Ukomadu, E.; Mueller, J.; Huwer, H.; Schaefer, U.F.; et al. Biopharmaceutical characterization of the telomerase inhibitor BRACO19. Pharm. Res. 2006, 23, 1031–1037. [Google Scholar] [CrossRef]
- Grenert, J.P.; Sullivan, W.P.; Fadden, P.; Haystead, T.A.; Clark, J.; Mimnaugh, E.; Krutzsch, H.; Ochel, H.J.; Schulte, T.W.; Sausville, E.; et al. The amino-terminal domain of heat shock protein 90 (hsp90) that binds geldanamycin is an ATP/ADP switch domain that regulates hsp90 conformation. J. Biol. Chem. 1997, 272, 23843–23850. [Google Scholar] [CrossRef]
- Lee, J.H.; Chung, I.K. Curcumin inhibits nuclear localization of telomerase by dissociating the Hsp90 co-chaperone p23 from hTERT. Cancer Lett. 2010, 290, 76–86. [Google Scholar] [CrossRef]
- Tauchi, T.; Shin-ya, K.; Sashida, G.; Sumi, M.; Okabe, S.; Ohyashiki, J.H.; Ohyashiki, K. Telomerase inhibition with a novel G-quadruplex-interactive agent, telomestatin: In vitro and in vivo studies in acute leukemia. Oncogene 2006, 25, 5719–5725. [Google Scholar] [CrossRef]
- Li, G.D.; Kawashima, H.; Ogose, A.; Ariizumi, T.; Hotta, T.; Kuwano, R.; Urata, Y.; Fujiwara, T.; Endo, N. Telomelysin shows potent antitumor activity through apoptotic and non-apoptotic cell death in soft tissue sarcoma cells. Cancer Sci. 2013, 104, 1178–1188. [Google Scholar] [CrossRef]
- Burchett, K.M.; Yan, Y.; Ouellette, M.M. Telomerase inhibitor Imetelstat (GRN163L) limits the lifespan of human pancreatic cancer cells. PLoS ONE 2014, 9, e85155. [Google Scholar] [CrossRef] [PubMed]
- Salloum, R.; Hummel, T.R.; Kumar, S.S.; Dorris, K.; Li, S.; Lin, T.; Daryani, V.M.; Stewart, C.F.; Miles, L.; Poussaint, T.Y.; et al. A molecular biology and phase II study of imetelstat (GRN163L) in children with recurrent or refractory central nervous system malignancies: A pediatric brain tumor consortium study. J. Neurooncol. 2016, 129, 443–451. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, M.M.S.; Fukuoka, K.; Maida, Y.; Hayashi, M.; Hamada, A.; Nishikawa, R.; Nagane, M.; Maruyama, T.; Mukasa, A.; Arakawa, Y.; et al. EXTH-50. Development of investigator initiated clinical trial of TERT-targeting therapy using eribulin mesylate in patients with recurrent glioblastoma. Neuro Oncol. 2017, 19, vi83. [Google Scholar] [CrossRef]
- Gibbons, R.J.; McDowell, T.L.; Raman, S.; O’Rourke, D.M.; Garrick, D.; Ayyub, H.; Higgs, D.R. Mutations in ATRX, encoding a SWI/SNF-like protein, cause diverse changes in the pattern of DNA methylation. Nat. Genet. 2000, 24, 368–371. [Google Scholar] [CrossRef]
- Yang, Y.L.; Huang, P.H.; Chiu, H.C.; Kulp, S.K.; Chen, C.S.; Kuo, C.J.; Chen, H.D.; Chen, C.S. Histone deacetylase inhibitor AR42 regulates telomerase activity in human glioma cells via an Akt-dependent mechanism. Biochem. Biophys. Res. Commun. 2013, 435, 107–112. [Google Scholar] [CrossRef] [PubMed]
- Grasso, C.S.; Tang, Y.; Truffaux, N.; Berlow, N.E.; Liu, L.; Debily, M.A.; Quist, M.J.; Davis, L.E.; Huang, E.C.; Woo, P.J.; et al. Functionally defined therapeutic targets in diffuse intrinsic pontine glioma. Nat. Med. 2015, 21, 827. [Google Scholar] [CrossRef] [PubMed]
- Lee, P.; Murphy, B.; Miller, R.; Menon, V.; Banik, N.L.; Giglio, P.; Lindhorst, S.M.; Varma, A.K.; Vandergrift, W.A.; Patel, S.J.; et al. Mechanisms and clinical significance of histone deacetylase inhibitors: Epigenetic glioblastoma therapy. Anticancer Res. 2015, 35, 615–625. [Google Scholar]
- Hashizume, R.; Andor, N.; Ihara, Y.; Lerner, R.; Gan, H.; Chen, X.; Fang, D.; Huang, X.; Tom, M.W.; Ngo, V.; et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat. Med. 2014, 20, 1394–1396. [Google Scholar] [CrossRef]
- Ashour, M.L.; Wink, M. Genus Bupleurum: A review of its phytochemistry, pharmacology and modes of action. J. Pharm. Pharmacol. 2011, 63, 305–321. [Google Scholar] [CrossRef]
- Adorisio, S.; Fierabracci, A.; Gigliarelli, G.; Muscari, I.; Cannarile, L.; Liberati, A.M.; Marcotullio, M.C.; Riccardi, C.; Curini, M.; Robles Zepeda, R.E.; et al. The Hexane Fraction of Bursera microphylla A Gray Induces p21-Mediated Antiproliferative and Proapoptotic Effects in Human Cancer-Derived Cell Lines. Integr. Cancer Ther. 2017, 16, 426–435. [Google Scholar] [CrossRef]
- Chen, Y.L.; Lin, P.C.; Chen, S.P.; Lin, C.C.; Tsai, N.M.; Cheng, Y.L.; Chang, W.L.; Lin, S.Z.; Harn, H.J. Activation of nonsteroidal anti-inflammatory drug-activated gene-1 via extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase revealed a isochaihulactone-triggered apoptotic pathway in human lung cancer A549 cells. J. Pharmacol. Exp. Ther. 2007, 323, 746–756. [Google Scholar] [CrossRef] [PubMed]
- Cochrane, C.B.; Nair, P.K.; Melnick, S.J.; Resek, A.P.; Ramachandran, C. Anticancer effects of Annona glabra plant extracts in human leukemia cell lines. Anticancer Res. 2008, 28, 965–971. [Google Scholar] [PubMed]
- Tsai, N.M.; Lin, S.Z.; Lee, C.C.; Chen, S.P.; Su, H.C.; Chang, W.L.; Harn, H.J. The antitumor effects of Angelica sinensis on malignant brain tumors in vitro and in vivo. Clin. Cancer Res. 2005, 11, 3475–3484. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.L.; Su, K.J.; Chen, C.J.; Wei, C.W.; Lin, C.J.; Yiang, G.T.; Lin, S.Z.; Harn, H.J.; Chen, Y.L. Synergistic anti-tumor activity of isochaihulactone and paclitaxel on human lung cancer cells. J. Cell. Physiol. 2012, 227, 213–222. [Google Scholar] [CrossRef] [PubMed]
- Lin, P.C.; Lin, S.Z.; Chen, Y.L.; Chang, J.S.; Ho, L.I.; Liu, P.Y.; Chang, L.F.; Harn, Y.C.; Chen, S.P.; Sun, L.Y.; et al. Butylidenephthalide suppresses human telomerase reverse transcriptase (TERT) in human glioblastomas. Ann. Surg. Oncol. 2011, 18, 3514–3527. [Google Scholar] [CrossRef] [PubMed]
- Tsai, N.M.; Chen, Y.L.; Lee, C.C.; Lin, P.C.; Cheng, Y.L.; Chang, W.L.; Lin, S.Z.; Harn, H.J. The natural compound n-butylidenephthalide derived from Angelica sinensis inhibits malignant brain tumor growth in vitro and in vivo. J. Neurochem. 2006, 99, 1251–1262. [Google Scholar] [CrossRef]
- Yen, S.Y.; Chen, S.R.; Hsieh, J.; Li, Y.S.; Chuang, S.E.; Chuang, H.M.; Huang, M.H.; Lin, S.Z.; Harn, H.J.; Chiou, T.W. Biodegradable interstitial release polymer loading a novel small molecule targeting Axl receptor tyrosine kinase and reducing brain tumour migration and invasion. Oncogene 2016, 35, 2156–2165. [Google Scholar] [CrossRef]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Fan, H.-C.; Chen, C.-M.; Chi, C.-S.; Tsai, J.-D.; Chiang, K.-L.; Chang, Y.-K.; Lin, S.-Z.; Harn, H.-J. Targeting Telomerase and ATRX/DAXX Inducing Tumor Senescence and Apoptosis in the Malignant Glioma. Int. J. Mol. Sci. 2019, 20, 200. https://doi.org/10.3390/ijms20010200
Fan H-C, Chen C-M, Chi C-S, Tsai J-D, Chiang K-L, Chang Y-K, Lin S-Z, Harn H-J. Targeting Telomerase and ATRX/DAXX Inducing Tumor Senescence and Apoptosis in the Malignant Glioma. International Journal of Molecular Sciences. 2019; 20(1):200. https://doi.org/10.3390/ijms20010200
Chicago/Turabian StyleFan, Hueng-Chuen, Chuan-Mu Chen, Ching-Shiang Chi, Jeng-Dau Tsai, Kuo-Liang Chiang, Yu-Kang Chang, Shinn-Zong Lin, and Horng-Jyh Harn. 2019. "Targeting Telomerase and ATRX/DAXX Inducing Tumor Senescence and Apoptosis in the Malignant Glioma" International Journal of Molecular Sciences 20, no. 1: 200. https://doi.org/10.3390/ijms20010200
APA StyleFan, H.-C., Chen, C.-M., Chi, C.-S., Tsai, J.-D., Chiang, K.-L., Chang, Y.-K., Lin, S.-Z., & Harn, H.-J. (2019). Targeting Telomerase and ATRX/DAXX Inducing Tumor Senescence and Apoptosis in the Malignant Glioma. International Journal of Molecular Sciences, 20(1), 200. https://doi.org/10.3390/ijms20010200