Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2020

01-12-2020 | Neuroblastoma | Review

Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma

Authors: S. L. George, V. Parmar, F. Lorenzi, L. V. Marshall, Y. Jamin, E. Poon, P. Angelini, L. Chesler

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2020

Login to get access

Abstract

The majority of high-risk neuroblastomas can be divided into three distinct molecular subgroups defined by the presence of MYCN amplification, upstream TERT rearrangements or alternative lengthening of telomeres (ALT). The common defining feature of all three subgroups is altered telomere maintenance; MYCN amplification and upstream TERT rearrangements drive high levels of telomerase expression whereas ALT is a telomerase independent telomere maintenance mechanism. As all three telomere maintenance mechanisms are independently associated with poor outcomes, the development of strategies to selectively target either telomerase expressing or ALT cells holds great promise as a therapeutic approach that is applicable to the majority of children with aggressive disease.
Here we summarise the biology of telomere maintenance and the molecular drivers of aggressive neuroblastoma before describing the most promising therapeutic strategies to target both telomerase expressing and ALT cancers. For telomerase-expressing neuroblastoma the most promising targeted agent to date is 6-thio-2′-deoxyguanosine, however clinical development of this agent is required. In osteosarcoma cell lines with ALT, selective sensitivity to ATR inhibition has been reported. However, we present data showing that in fact ALT neuroblastoma cells are more resistant to the clinical ATR inhibitor AZD6738 compared to other neuroblastoma subtypes. More recently a number of additional candidate compounds have been shown to show selectivity for ALT cancers, such as Tetra-Pt (bpy), a compound targeting the telomeric G-quadruplex and pifithrin-α, a putative p53 inhibitor. Further pre-clinical evaluation of these compounds in neuroblastoma models is warranted.
In summary, telomere maintenance targeting strategies offer a significant opportunity to develop effective new therapies, applicable to a large proportion of children with high-risk neuroblastoma. In parallel to clinical development, more pre-clinical research specifically for neuroblastoma is urgently needed, if we are to improve survival for this common poor outcome tumour of childhood.
Literature
1.
2.
go back to reference Ladenstein R, et al. Busulfan and melphalan versus carboplatin, etoposide, and melphalan as high-dose chemotherapy for high-risk neuroblastoma (HR-NBL1/SIOPEN): an international, randomised, multi-arm, open-label, phase 3 trial. Lancet Oncol. 2017;18(4):500–14.PubMedCrossRef Ladenstein R, et al. Busulfan and melphalan versus carboplatin, etoposide, and melphalan as high-dose chemotherapy for high-risk neuroblastoma (HR-NBL1/SIOPEN): an international, randomised, multi-arm, open-label, phase 3 trial. Lancet Oncol. 2017;18(4):500–14.PubMedCrossRef
3.
go back to reference Amoroso L, et al. Topotecan-Vincristine-Doxorubicin in Stage 4 High-Risk Neuroblastoma Patients Failing to Achieve a Complete Metastatic Response to Rapid COJEC: A SIOPEN Study. Cancer Res Treat. 2018;50(1):148–55.PubMedCrossRef Amoroso L, et al. Topotecan-Vincristine-Doxorubicin in Stage 4 High-Risk Neuroblastoma Patients Failing to Achieve a Complete Metastatic Response to Rapid COJEC: A SIOPEN Study. Cancer Res Treat. 2018;50(1):148–55.PubMedCrossRef
4.
go back to reference Valentijn LJ, et al. TERT rearrangements are frequent in neuroblastoma and identify aggressive tumors. Nat Genet. 2015;47(12):1411–4.PubMedCrossRef Valentijn LJ, et al. TERT rearrangements are frequent in neuroblastoma and identify aggressive tumors. Nat Genet. 2015;47(12):1411–4.PubMedCrossRef
7.
go back to reference Shay JW, Wright WE. Hayflick, his limit, and cellular ageing. Nat Rev Mol Cell Biol. 2000;1(1):72–6.PubMedCrossRef Shay JW, Wright WE. Hayflick, his limit, and cellular ageing. Nat Rev Mol Cell Biol. 2000;1(1):72–6.PubMedCrossRef
8.
go back to reference Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20(5):299–309.PubMedCrossRef Shay JW, Wright WE. Telomeres and telomerase: three decades of progress. Nat Rev Genet. 2019;20(5):299–309.PubMedCrossRef
9.
10.
go back to reference Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. Eur J Cancer. 1997;33(5):787–91.PubMedCrossRef Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. Eur J Cancer. 1997;33(5):787–91.PubMedCrossRef
11.
go back to reference Kim NW, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266(5193):2011–5.PubMedCrossRef Kim NW, et al. Specific association of human telomerase activity with immortal cells and cancer. Science. 1994;266(5193):2011–5.PubMedCrossRef
12.
go back to reference Wright WE, et al. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18(2):173–9.PubMedCrossRef Wright WE, et al. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet. 1996;18(2):173–9.PubMedCrossRef
14.
go back to reference Henson JD, Reddel RR. Assaying and investigating Alternative Lengthening of Telomeres activity in human cells and cancers. FEBS Lett. 2010;584(17):3800–11.PubMedCrossRef Henson JD, Reddel RR. Assaying and investigating Alternative Lengthening of Telomeres activity in human cells and cancers. FEBS Lett. 2010;584(17):3800–11.PubMedCrossRef
15.
go back to reference Amorim JP, et al. The Role of ATRX in the Alternative Lengthening of Telomeres (ALT) Phenotype. Genes (Basel). 2016;7(9):66.CrossRef Amorim JP, et al. The Role of ATRX in the Alternative Lengthening of Telomeres (ALT) Phenotype. Genes (Basel). 2016;7(9):66.CrossRef
16.
17.
go back to reference Mafficini A, Scarpa A. Genomic landscape of pancreatic neuroendocrine tumours: the International Cancer Genome Consortium. J Endocrinol. 2018;236(3):R161–7.PubMedPubMedCentralCrossRef Mafficini A, Scarpa A. Genomic landscape of pancreatic neuroendocrine tumours: the International Cancer Genome Consortium. J Endocrinol. 2018;236(3):R161–7.PubMedPubMedCentralCrossRef
19.
go back to reference O'Sullivan RJ, et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nat Struct Mol Biol. 2014;21(2):167–74.PubMedPubMedCentralCrossRef O'Sullivan RJ, et al. Rapid induction of alternative lengthening of telomeres by depletion of the histone chaperone ASF1. Nat Struct Mol Biol. 2014;21(2):167–74.PubMedPubMedCentralCrossRef
21.
go back to reference Royle NJ, et al. The role of recombination in telomere length maintenance. Biochem Soc Trans. 2009;37(Pt 3):589–95.PubMedCrossRef Royle NJ, et al. The role of recombination in telomere length maintenance. Biochem Soc Trans. 2009;37(Pt 3):589–95.PubMedCrossRef
23.
go back to reference Chang FT, et al. PML bodies provide an important platform for the maintenance of telomeric chromatin integrity in embryonic stem cells. Nucleic Acids Res. 2013;41(8):4447–58.PubMedPubMedCentralCrossRef Chang FT, et al. PML bodies provide an important platform for the maintenance of telomeric chromatin integrity in embryonic stem cells. Nucleic Acids Res. 2013;41(8):4447–58.PubMedPubMedCentralCrossRef
25.
go back to reference Lewis PW, et al. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A. 2010;107(32):14075–80.PubMedPubMedCentralCrossRef Lewis PW, et al. Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A. 2010;107(32):14075–80.PubMedPubMedCentralCrossRef
26.
go back to reference Clynes D, et al. Suppression of the alternative lengthening of telomere pathway by the chromatin remodelling factor ATRX. Nat Commun. 2015;6:7538.PubMedCrossRef Clynes D, et al. Suppression of the alternative lengthening of telomere pathway by the chromatin remodelling factor ATRX. Nat Commun. 2015;6:7538.PubMedCrossRef
29.
go back to reference Graf M, et al. Telomere Length Determines TERRA and R-Loop Regulation through the Cell Cycle. Cell. 2017;170(1):72–85 e14.PubMedCrossRef Graf M, et al. Telomere Length Determines TERRA and R-Loop Regulation through the Cell Cycle. Cell. 2017;170(1):72–85 e14.PubMedCrossRef
31.
go back to reference Graham MK, et al. Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells. Mol Cancer Res. 2019;17(12):2480–91.PubMedCrossRefPubMedCentral Graham MK, et al. Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells. Mol Cancer Res. 2019;17(12):2480–91.PubMedCrossRefPubMedCentral
32.
go back to reference Brosnan-Cashman JA, et al. ATRX loss induces multiple hallmarks of the alternative lengthening of telomeres (ALT) phenotype in human glioma cell lines in a cell line-specific manner. PLoS One. 2018;13(9):e0204159.PubMedPubMedCentralCrossRef Brosnan-Cashman JA, et al. ATRX loss induces multiple hallmarks of the alternative lengthening of telomeres (ALT) phenotype in human glioma cell lines in a cell line-specific manner. PLoS One. 2018;13(9):e0204159.PubMedPubMedCentralCrossRef
33.
go back to reference Li F, et al. ATRX loss induces telomere dysfunction and necessitates induction of alternative lengthening of telomeres during human cell immortalization. EMBO J. 2019;38(19):e96659.PubMedCrossRefPubMedCentral Li F, et al. ATRX loss induces telomere dysfunction and necessitates induction of alternative lengthening of telomeres during human cell immortalization. EMBO J. 2019;38(19):e96659.PubMedCrossRefPubMedCentral
35.
go back to reference Salloum R, 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(3):443–51.PubMedPubMedCentralCrossRef Salloum R, 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(3):443–51.PubMedPubMedCentralCrossRef
36.
go back to reference Dagg RA, et al. Extensive Proliferation of Human Cancer Cells with Ever-Shorter Telomeres. Cell Rep. 2017;19(12):2544–56.PubMedCrossRef Dagg RA, et al. Extensive Proliferation of Human Cancer Cells with Ever-Shorter Telomeres. Cell Rep. 2017;19(12):2544–56.PubMedCrossRef
37.
go back to reference Onitake Y, et al. Telomere biology in neuroblastoma: telomere binding proteins and alternative strengthening of telomeres. J Pediatr Surg. 2009;44(12):2258–66.PubMedCrossRef Onitake Y, et al. Telomere biology in neuroblastoma: telomere binding proteins and alternative strengthening of telomeres. J Pediatr Surg. 2009;44(12):2258–66.PubMedCrossRef
38.
go back to reference Yeager TR, et al. Telomerase-negative immortalized human cells contain a novel type of promyelocytic leukemia (PML) body. Cancer Res. 1999;59(17):4175–9.PubMed Yeager TR, et al. Telomerase-negative immortalized human cells contain a novel type of promyelocytic leukemia (PML) body. Cancer Res. 1999;59(17):4175–9.PubMed
39.
go back to reference Henson JD, et al. DNA C-circles are specific and quantifiable markers of alternative-lengthening-of-telomeres activity. Nat Biotechnol. 2009;27(12):1181–5.PubMedCrossRef Henson JD, et al. DNA C-circles are specific and quantifiable markers of alternative-lengthening-of-telomeres activity. Nat Biotechnol. 2009;27(12):1181–5.PubMedCrossRef
40.
go back to reference Henson JD, et al. The C-Circle Assay for alternative-lengthening-of-telomeres activity. Methods. 2017;114:74–84.PubMedCrossRef Henson JD, et al. The C-Circle Assay for alternative-lengthening-of-telomeres activity. Methods. 2017;114:74–84.PubMedCrossRef
41.
go back to reference Lee JW, et al. Clinical significance of MYCN amplification in patients with high-risk neuroblastoma. Pediatr Blood Cancer. 2018;65(10):e27257.PubMedCrossRef Lee JW, et al. Clinical significance of MYCN amplification in patients with high-risk neuroblastoma. Pediatr Blood Cancer. 2018;65(10):e27257.PubMedCrossRef
42.
go back to reference Kuzyk A, Gartner J, Mai S. Identification of Neuroblastoma Subgroups Based on Three-Dimensional Telomere Organization. Transl Oncol. 2016;9(4):348–56.PubMedPubMedCentralCrossRef Kuzyk A, Gartner J, Mai S. Identification of Neuroblastoma Subgroups Based on Three-Dimensional Telomere Organization. Transl Oncol. 2016;9(4):348–56.PubMedPubMedCentralCrossRef
44.
go back to reference Kawashima M, et al. Telomere biology including TERT rearrangements in neuroblastoma: a useful indicator for surgical treatments. J Pediatr Surg. 2016;51(12):2080–5.PubMedCrossRef Kawashima M, et al. Telomere biology including TERT rearrangements in neuroblastoma: a useful indicator for surgical treatments. J Pediatr Surg. 2016;51(12):2080–5.PubMedCrossRef
45.
go back to reference Hertwig F, Peifer M, Fischer M. Telomere maintenance is pivotal for high-risk neuroblastoma. Cell Cycle. 2016;15(3):311–2.PubMedCrossRef Hertwig F, Peifer M, Fischer M. Telomere maintenance is pivotal for high-risk neuroblastoma. Cell Cycle. 2016;15(3):311–2.PubMedCrossRef
46.
go back to reference Ohali A, et al. Telomere length is a prognostic factor in neuroblastoma. Cancer. 2006;107(6):1391–9.PubMedCrossRef Ohali A, et al. Telomere length is a prognostic factor in neuroblastoma. Cancer. 2006;107(6):1391–9.PubMedCrossRef
47.
go back to reference Roderwieser A, Sand F, Walter E, Fischer J, Getcht J, Batenhagen C, Ackermann S, Otte F, Welte A, Kahlert Y, Lieberz D, Hertwig F, Reinhardt C, Simon T, Peifer M, Ortmann M, Buttner R, Her B, O'Sullivan RJ, Berthold F, Fischer M. Telomerase is a prognostic marker of poor outcome and a therapeutic target in neuroblastoma. JCO Precision Oncol. 2019;3:1–20. Roderwieser A, Sand F, Walter E, Fischer J, Getcht J, Batenhagen C, Ackermann S, Otte F, Welte A, Kahlert Y, Lieberz D, Hertwig F, Reinhardt C, Simon T, Peifer M, Ortmann M, Buttner R, Her B, O'Sullivan RJ, Berthold F, Fischer M. Telomerase is a prognostic marker of poor outcome and a therapeutic target in neuroblastoma. JCO Precision Oncol. 2019;3:1–20.
48.
go back to reference Schleiermacher G, et al. Stepwise occurrence of a complex unbalanced translocation in neuroblastoma leading to insertion of a telomere sequence and late chromosome 17q gain. Oncogene. 2005;24(20):3377–84.PubMedCrossRef Schleiermacher G, et al. Stepwise occurrence of a complex unbalanced translocation in neuroblastoma leading to insertion of a telomere sequence and late chromosome 17q gain. Oncogene. 2005;24(20):3377–84.PubMedCrossRef
49.
go back to reference Walsh KM, et al. Common genetic variants associated with telomere length confer risk for neuroblastoma and other childhood cancers. Carcinogenesis. 2016;37(6):576–82.PubMedPubMedCentralCrossRef Walsh KM, et al. Common genetic variants associated with telomere length confer risk for neuroblastoma and other childhood cancers. Carcinogenesis. 2016;37(6):576–82.PubMedPubMedCentralCrossRef
51.
go back to reference Roth A, Harley CB, Baerlocher GM. Imetelstat (GRN163L)--telomerase-based cancer therapy. Recent Results Cancer Res. 2010;184:221–34.PubMedCrossRef Roth A, Harley CB, Baerlocher GM. Imetelstat (GRN163L)--telomerase-based cancer therapy. Recent Results Cancer Res. 2010;184:221–34.PubMedCrossRef
52.
go back to reference Dikmen ZG, et al. In vivo inhibition of lung cancer by GRN163L: a novel human telomerase inhibitor. Cancer Res. 2005;65(17):7866–73.PubMedCrossRef Dikmen ZG, et al. In vivo inhibition of lung cancer by GRN163L: a novel human telomerase inhibitor. Cancer Res. 2005;65(17):7866–73.PubMedCrossRef
53.
go back to reference Gellert GC, et al. Effects of a novel telomerase inhibitor, GRN163L, in human breast cancer. Breast Cancer Res Treat. 2006;96(1):73–81.PubMedCrossRef Gellert GC, et al. Effects of a novel telomerase inhibitor, GRN163L, in human breast cancer. Breast Cancer Res Treat. 2006;96(1):73–81.PubMedCrossRef
54.
go back to reference Barszczyk M, et al. Telomerase inhibition abolishes the tumorigenicity of pediatric ependymoma tumor-initiating cells. Acta Neuropathol. 2014;128(6):863–77.PubMedPubMedCentralCrossRef Barszczyk M, et al. Telomerase inhibition abolishes the tumorigenicity of pediatric ependymoma tumor-initiating cells. Acta Neuropathol. 2014;128(6):863–77.PubMedPubMedCentralCrossRef
55.
go back to reference Thompson PA, et al. A phase I trial of imetelstat in children with refractory or recurrent solid tumors: a Children's Oncology Group Phase I Consortium Study (ADVL1112). Clin Cancer Res. 2013;19(23):6578–84.PubMedPubMedCentralCrossRef Thompson PA, et al. A phase I trial of imetelstat in children with refractory or recurrent solid tumors: a Children's Oncology Group Phase I Consortium Study (ADVL1112). Clin Cancer Res. 2013;19(23):6578–84.PubMedPubMedCentralCrossRef
56.
go back to reference Pascolo E, et al. Mechanism of human telomerase inhibition by BIBR1532, a synthetic, non-nucleosidic drug candidate. J Biol Chem. 2002;277(18):15566–72.PubMedCrossRef Pascolo E, et al. Mechanism of human telomerase inhibition by BIBR1532, a synthetic, non-nucleosidic drug candidate. J Biol Chem. 2002;277(18):15566–72.PubMedCrossRef
57.
go back to reference Hosoi T, et al. Inhibition of telomerase causes vulnerability to endoplasmic reticulum stress-induced neuronal cell death. Neurosci Lett. 2016;629:241–4.PubMedCrossRef Hosoi T, et al. Inhibition of telomerase causes vulnerability to endoplasmic reticulum stress-induced neuronal cell death. Neurosci Lett. 2016;629:241–4.PubMedCrossRef
58.
go back to reference Phatak P, et al. KML001 cytotoxic activity is associated with its binding to telomeric sequences and telomere erosion in prostate cancer cells. Clin Cancer Res. 2008;14(14):4593–602.PubMedCrossRef Phatak P, et al. KML001 cytotoxic activity is associated with its binding to telomeric sequences and telomere erosion in prostate cancer cells. Clin Cancer Res. 2008;14(14):4593–602.PubMedCrossRef
59.
go back to reference Repetto G, Sanz P, Repetto M. Comparative in vitro effects of sodium arsenite and sodium arsenate on neuroblastoma cells. Toxicology. 1994;92(1–3):143–53.PubMedCrossRef Repetto G, Sanz P, Repetto M. Comparative in vitro effects of sodium arsenite and sodium arsenate on neuroblastoma cells. Toxicology. 1994;92(1–3):143–53.PubMedCrossRef
60.
go back to reference Edelman MJ, et al. Phase I and pharmacokinetic evaluation of the anti-telomerase agent KML-001 with cisplatin in advanced solid tumors. Cancer Chemother Pharmacol. 2016;78(5):959–67.PubMedCrossRef Edelman MJ, et al. Phase I and pharmacokinetic evaluation of the anti-telomerase agent KML-001 with cisplatin in advanced solid tumors. Cancer Chemother Pharmacol. 2016;78(5):959–67.PubMedCrossRef
61.
go back to reference Kim MY, et al. Telomestatin, a potent telomerase inhibitor that interacts quite specifically with the human telomeric intramolecular g-quadruplex. J Am Chem Soc. 2002;124(10):2098–9.PubMedCrossRef Kim MY, et al. Telomestatin, a potent telomerase inhibitor that interacts quite specifically with the human telomeric intramolecular g-quadruplex. J Am Chem Soc. 2002;124(10):2098–9.PubMedCrossRef
62.
go back to reference Binz N, et al. Telomerase inhibition, telomere shortening, cell growth suppression and induction of apoptosis by telomestatin in childhood neuroblastoma cells. Eur J Cancer. 2005;41(18):2873–81.PubMedCrossRef Binz N, et al. Telomerase inhibition, telomere shortening, cell growth suppression and induction of apoptosis by telomestatin in childhood neuroblastoma cells. Eur J Cancer. 2005;41(18):2873–81.PubMedCrossRef
63.
go back to reference Mender I, et al. Induction of telomere dysfunction mediated by the telomerase substrate precursor 6-thio-2′-deoxyguanosine. Cancer Discov. 2015;5(1):82–95.PubMedCrossRef Mender I, et al. Induction of telomere dysfunction mediated by the telomerase substrate precursor 6-thio-2′-deoxyguanosine. Cancer Discov. 2015;5(1):82–95.PubMedCrossRef
65.
go back to reference Mender I, et al. Telomerase-Mediated Strategy for Overcoming Non-Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance. Neoplasia. 2018;20(8):826–37.PubMedPubMedCentralCrossRef Mender I, et al. Telomerase-Mediated Strategy for Overcoming Non-Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance. Neoplasia. 2018;20(8):826–37.PubMedPubMedCentralCrossRef
66.
go back to reference Sengupta S, et al. Induced Telomere Damage to Treat Telomerase Expressing Therapy-Resistant Pediatric Brain Tumors. Mol Cancer Ther. 2018;17(7):1504–14.PubMedCrossRef Sengupta S, et al. Induced Telomere Damage to Treat Telomerase Expressing Therapy-Resistant Pediatric Brain Tumors. Mol Cancer Ther. 2018;17(7):1504–14.PubMedCrossRef
68.
go back to reference Barone A, et al. FDA Approval Summary: Trabectedin for Unresectable or Metastatic Liposarcoma or Leiomyosarcoma Following an Anthracycline-Containing Regimen. Clin Cancer Res. 2017;23(24):7448–53.PubMedCrossRef Barone A, et al. FDA Approval Summary: Trabectedin for Unresectable or Metastatic Liposarcoma or Leiomyosarcoma Following an Anthracycline-Containing Regimen. Clin Cancer Res. 2017;23(24):7448–53.PubMedCrossRef
69.
go back to reference Baruchel S, et al. A phase 2 trial of trabectedin in children with recurrent rhabdomyosarcoma, Ewing sarcoma and non-rhabdomyosarcoma soft tissue sarcomas: a report from the Children’s Oncology Group. Eur J Cancer. 2012;48(4):579–85.PubMedCrossRef Baruchel S, et al. A phase 2 trial of trabectedin in children with recurrent rhabdomyosarcoma, Ewing sarcoma and non-rhabdomyosarcoma soft tissue sarcomas: a report from the Children’s Oncology Group. Eur J Cancer. 2012;48(4):579–85.PubMedCrossRef
70.
go back to reference Tian X, et al. XAV939 promotes apoptosis in a neuroblastoma cell line via telomere shortening. Oncol Rep. 2014;32(5):1999–2006.PubMedCrossRef Tian X, et al. XAV939 promotes apoptosis in a neuroblastoma cell line via telomere shortening. Oncol Rep. 2014;32(5):1999–2006.PubMedCrossRef
71.
go back to reference Koneru BLG, Nguyen L, Chen WH, Macha S, Farooqi A, Hindle A, Davidson H, Mccoy K, Yang S, Maris J, Reynolds P. Alternate Telomere Lengthening (ALT) neuroblastoma is a highly aggressive subgroup for which ATM kinase provides a novel therapeutic target. Advances in Neuroblastoma Conference, 2018. Abstract 272. Advances in Neuroblastoma Abstract Book. Adv Neuroblastoma Res. 2018. https://www.anrmeeting.org/dl/ANR2018/ANR_Abstract_Book_5-3-18.pdf. Koneru BLG, Nguyen L, Chen WH, Macha S, Farooqi A, Hindle A, Davidson H, Mccoy K, Yang S, Maris J, Reynolds P. Alternate Telomere Lengthening (ALT) neuroblastoma is a highly aggressive subgroup for which ATM kinase provides a novel therapeutic target. Advances in Neuroblastoma Conference, 2018. Abstract 272. Advances in Neuroblastoma Abstract Book. Adv Neuroblastoma Res. 2018. https://​www.​anrmeeting.​org/​dl/​ANR2018/​ANR_​Abstract_​Book_​5-3-18.​pdf.
72.
go back to reference Abida WBYJ, Azaro A, Krebs M, Im S, Chen T, Buil-Bruna N, Li Y, Eaton D, Stephens C, Ross G, Pass M, Rodon J, Dean E. Abstract A094: Phase 1 molecular study of AZD0156, a first-in-class oral selective inhibitor of ataxia telangiectasia mutated protein kinase (ATM), in combination with olaparib (AtoM Study, Module 1). Mol Cancer Ther. 2018;17(1):Abstract AO94. Abida WBYJ, Azaro A, Krebs M, Im S, Chen T, Buil-Bruna N, Li Y, Eaton D, Stephens C, Ross G, Pass M, Rodon J, Dean E. Abstract A094: Phase 1 molecular study of AZD0156, a first-in-class oral selective inhibitor of ataxia telangiectasia mutated protein kinase (ATM), in combination with olaparib (AtoM Study, Module 1). Mol Cancer Ther. 2018;17(1):Abstract AO94.
73.
74.
go back to reference Foote KM, et al. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J Med Chem. 2018;61(22):9889–907.PubMedCrossRef Foote KM, et al. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J Med Chem. 2018;61(22):9889–907.PubMedCrossRef
75.
go back to reference Farooqi AS, et al. Alternative lengthening of telomeres in neuroblastoma cell lines is associated with a lack of MYCN genomic amplification and with p53 pathway aberrations. J Neurooncol. 2014;119(1):17–26.PubMedCrossRef Farooqi AS, et al. Alternative lengthening of telomeres in neuroblastoma cell lines is associated with a lack of MYCN genomic amplification and with p53 pathway aberrations. J Neurooncol. 2014;119(1):17–26.PubMedCrossRef
77.
go back to reference Deeg KI, et al. Cancer Cells with Alternative Lengthening of Telomeres Do Not Display a General Hypersensitivity to ATR Inhibition. Front Oncol. 2016;6:186.PubMedPubMedCentralCrossRef Deeg KI, et al. Cancer Cells with Alternative Lengthening of Telomeres Do Not Display a General Hypersensitivity to ATR Inhibition. Front Oncol. 2016;6:186.PubMedPubMedCentralCrossRef
78.
go back to reference Murga M, et al. Exploiting oncogene-induced replicative stress for the selective killing of Myc-driven tumors. Nat Struct Mol Biol. 2011;18(12):1331–5.PubMedPubMedCentralCrossRef Murga M, et al. Exploiting oncogene-induced replicative stress for the selective killing of Myc-driven tumors. Nat Struct Mol Biol. 2011;18(12):1331–5.PubMedPubMedCentralCrossRef
79.
go back to reference Muralidharan SV, et al. BET bromodomain inhibitors synergize with ATR inhibitors to induce DNA damage, apoptosis, senescence-associated secretory pathway and ER stress in Myc-induced lymphoma cells. Oncogene. 2016;35(36):4689–97.PubMedCrossRef Muralidharan SV, et al. BET bromodomain inhibitors synergize with ATR inhibitors to induce DNA damage, apoptosis, senescence-associated secretory pathway and ER stress in Myc-induced lymphoma cells. Oncogene. 2016;35(36):4689–97.PubMedCrossRef
80.
go back to reference Zimmerman MW, et al. MYC Drives a Subset of High-Risk Pediatric Neuroblastomas and Is Activated through Mechanisms Including Enhancer Hijacking and Focal Enhancer Amplification. Cancer Discov. 2018;8(3):320–35.PubMedCrossRef Zimmerman MW, et al. MYC Drives a Subset of High-Risk Pediatric Neuroblastomas and Is Activated through Mechanisms Including Enhancer Hijacking and Focal Enhancer Amplification. Cancer Discov. 2018;8(3):320–35.PubMedCrossRef
81.
go back to reference Buchel G, et al. Association with Aurora-A Controls N-MYC-Dependent Promoter Escape and Pause Release of RNA Polymerase II during the Cell Cycle. Cell Rep. 2017;21(12):3483–97.PubMedPubMedCentralCrossRef Buchel G, et al. Association with Aurora-A Controls N-MYC-Dependent Promoter Escape and Pause Release of RNA Polymerase II during the Cell Cycle. Cell Rep. 2017;21(12):3483–97.PubMedPubMedCentralCrossRef
82.
go back to reference Zheng XH, et al. A Cisplatin Derivative Tetra-Pt (bpy) as an Oncotherapeutic Agent for Targeting ALT Cancer. J Natl Cancer Inst. 2017;109(10). Zheng XH, et al. A Cisplatin Derivative Tetra-Pt (bpy) as an Oncotherapeutic Agent for Targeting ALT Cancer. J Natl Cancer Inst. 2017;109(10).
83.
84.
85.
go back to reference Walton MI, et al. An evaluation of the ability of pifithrin-alpha and -beta to inhibit p53 function in two wild-type p53 human tumor cell lines. Mol Cancer Ther. 2005;4(9):1369–77.PubMedCrossRef Walton MI, et al. An evaluation of the ability of pifithrin-alpha and -beta to inhibit p53 function in two wild-type p53 human tumor cell lines. Mol Cancer Ther. 2005;4(9):1369–77.PubMedCrossRef
88.
go back to reference M'Kacher R, et al. The Transition between Telomerase and ALT Mechanisms in Hodgkin Lymphoma and Its Predictive Value in Clinical Outcomes. Cancers (Basel). 2018;10(6):169.PubMedCentralCrossRef M'Kacher R, et al. The Transition between Telomerase and ALT Mechanisms in Hodgkin Lymphoma and Its Predictive Value in Clinical Outcomes. Cancers (Basel). 2018;10(6):169.PubMedCentralCrossRef
89.
go back to reference Venturini L, et al. Telomere maintenance in Wilms tumors: first evidence for the presence of alternative lengthening of telomeres mechanism. Genes Chromosomes Cancer. 2011;50(10):823–9.PubMedCrossRef Venturini L, et al. Telomere maintenance in Wilms tumors: first evidence for the presence of alternative lengthening of telomeres mechanism. Genes Chromosomes Cancer. 2011;50(10):823–9.PubMedCrossRef
90.
go back to reference Pezzolo A, et al. Intratumoral diversity of telomere length in individual neuroblastoma tumors. Oncotarget. 2015;6(10):7493–503.PubMedCrossRef Pezzolo A, et al. Intratumoral diversity of telomere length in individual neuroblastoma tumors. Oncotarget. 2015;6(10):7493–503.PubMedCrossRef
92.
go back to reference Chen W, et al. Telomerase inhibition alters telomere maintenance mechanisms in laryngeal squamous carcinoma cells. J Laryngol Otol. 2010;124(7):778–83.PubMedCrossRef Chen W, et al. Telomerase inhibition alters telomere maintenance mechanisms in laryngeal squamous carcinoma cells. J Laryngol Otol. 2010;124(7):778–83.PubMedCrossRef
93.
go back to reference Bechter OE, et al. Telomeric recombination in mismatch repair deficient human colon cancer cells after telomerase inhibition. Cancer Res. 2004;64(10):3444–51.PubMedCrossRef Bechter OE, et al. Telomeric recombination in mismatch repair deficient human colon cancer cells after telomerase inhibition. Cancer Res. 2004;64(10):3444–51.PubMedCrossRef
95.
go back to reference Westermann F, et al. Distinct transcriptional MYCN/c-MYC activities are associated with spontaneous regression or malignant progression in neuroblastomas. Genome Biol. 2008;9(10):R150.PubMedPubMedCentralCrossRef Westermann F, et al. Distinct transcriptional MYCN/c-MYC activities are associated with spontaneous regression or malignant progression in neuroblastomas. Genome Biol. 2008;9(10):R150.PubMedPubMedCentralCrossRef
96.
go back to reference Qadeer ZA, et al. ATRX In-Frame Fusion Neuroblastoma Is Sensitive to EZH2 Inhibition via Modulation of Neuronal Gene Signatures. Cancer Cell. 2019;36(5):512–27 e9.PubMedCrossRefPubMedCentral Qadeer ZA, et al. ATRX In-Frame Fusion Neuroblastoma Is Sensitive to EZH2 Inhibition via Modulation of Neuronal Gene Signatures. Cancer Cell. 2019;36(5):512–27 e9.PubMedCrossRefPubMedCentral
98.
99.
go back to reference Grobner SN, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555(7696):321–7.PubMedCrossRef Grobner SN, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555(7696):321–7.PubMedCrossRef
Metadata
Title
Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma
Authors
S. L. George
V. Parmar
F. Lorenzi
L. V. Marshall
Y. Jamin
E. Poon
P. Angelini
L. Chesler
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2020
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01582-2

Other articles of this Issue 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine