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

Open Access 01.12.2020 | Review

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

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

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

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.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
TERT
Telomerase reverse transcriptase
ALT
Alternative lengthening of telomeres
TMM
Telomere maintenance mechanism
hTR
Human telomerase RNA
LoF
Loss of function
ATRX
Alpha Thalassemia mental Retardation-X linked
HR
Homologous recombination
MRN
Mre11-RAD50-Nbs1
PML
Pro-myelocytic Leukaemia
TERRA
RNA TElomeric Repeat-containing RNA
DAXX
Death domain associated protein
TP53
Tumour protein 53
RB1
Retinoblastoma 1
TRAP
Telomere repeat amplification protocol
FFPE
Formalin fixed paraffin embedded
ATM
Ataxia telangiectasia mutated
ATR
Ataxia Telangiectasia and Rad3 related
FISH
Fluorescence in-situ hybridisation

Background

Neuroblastoma is a common childhood malignancy arising from the sympathetic nervous system. It most commonly arises from the adrenal gland and presents with an abdominal mass, but has a heterogeneous clinical phenotype. Children aged less than 18 months often present with widespread metastases to the skin, liver and bone marrow (stage MS disease) [1], however this can spontaneously resolve without treatment. Conversely, distant metastatic spread in children aged greater than 18 months (stage M disease) is associated with an aggressive clinical phenotype and poor survival [1, 2].
Children aged > 18 months with stage M disease and/or amplification of the MYCN oncogene are classified as having clinical high-risk disease. High-risk neuroblastoma remains a major therapeutic challenge with survival rates of < 50% despite intensification of therapy [2, 3]. However, until recently, in the absence of MYCN amplification, the molecular drivers of aggressive disease were unknown.
In 2015 it was reported that aggressive neuroblastoma can be divided into 3 almost mutually exclusive subgroups with either MYCN amplification, rearrangements upstream to the telomerase reverse transcriptase (TERT) gene or alternative lengthening of telomeres (ALT) [4, 5]. Each subgroup is associated with the activation of a telomere maintenance mechanism (TMM) and poor outcomes. Conversely, it is thought that the absence of a TMM is associated with spontaneous regression and excellent survival [6].
Here we summarise TMMs in cancer, specifically focusing on the molecular alterations driving telomere maintenance in neuroblastoma, before describing potential novel therapeutic strategies to directly target TMMs for children with neuroblastoma.

Biology of telomere maintenance

Telomeres are regions of repetitive nucleotide sequences (TTAGGG) located at the ends of chromosomes that protect chromosomes from DNA damage, unnecessary DNA repair and fusion with other chromosomes. In normal dividing cells, with each cell replication telomeres gradually shorten, until a critical level is reached, the Hayflick limit, after which cells undergo senescence [7]. This gradual shortening of telomeres associated with cellular aging is believed to be a protective mechanism against uncontrolled growth, preventing cancer development in humans and other mammals [8]. In keeping with this, the activation of a TMM to prevent the shortening of telomeres is necessary for the continued sustained proliferation of cancer cells and hence a hallmark of cancer [9].
Telomerase, a functional ribonucleoprotein enzyme complex, maintains telomere length by adding telomeric DNA repeats at the 3′ ends of linear chromosomes. It is a reverse transcriptase that consists of a catalytic protein subunit TERT, and an essential RNA component known as human telomerase RNA (hTR), encoded by hTERC. hTR acts as a template for the synthesis of telomere DNA and is involved in the catalysis, localisation, and assembly of the telomerase holoenzyme [10, 11]. Telomerase is widely expressed in human embryos between 16 and 20 weeks gestation, however by the early neonatal period telomerase activity can no longer be detected in most somatic tissues [12]. In contrast, the majority of cancers overexpress telomerase: in a systematic analysis of 31 tumour types, over-expression of TERT was identified in 73% of all cancers [13]. This was most commonly associated with genetic alterations in either the TERT gene/promoter or TERT promoter methylation.
ALT is defined as maintenance of telomeres in the absence of telomerase activity [14]. It can be detected in 10–15% cancers overall but is particularly prevalent in tumours of mesenchymal origin [14, 15]. There is a strong association between ALT and loss of function (LoF) genetic alterations in ATRX (Alpha Thalassemia mental Retardation-X linked) in multiple malignancies, including neuroblastoma [13, 1618].
A number of different non-canonical homologous recombination (HR) based mechanisms have been proposed to play a role in ALT telomere maintenance [1922]. Furthermore, a number of studies have focused on the underlying basis for the relationship between ATRX LoF and the development of the non-canonical HR mechanisms implicated in ALT (summarised in Fig. 1). Firstly an established role of ATRX is the maintenance of genomic stability via the deposition of H3.3 into telomeric regions [24, 25]. In the absence of ATRX, G4 quadruplex structures may occur in guanine rich regions of DNA such as telomeres, resulting in stalling of replication forks, which provides a substrate for HR [26, 27]. Secondly, in the absence of ATRX, the MRN (Mre11-RAD50-Nbs1) complex is redistributed to ALT associated PML body sites where it is thought to also facilitate HR mechanisms [26]. Finally, it has been shown that the long non-coding RNA TElomeric Repeat-containing RNA (TERRA) is functionally antagonistic with ATRX [28], and therefore in the absence of ATRX, TERRA can form DNA-RNA hybrids known as R loops, that promote homology directed repair of telomeres [29]. Further confirming the role of ATRX as an ALT repressor, ATRX knockdown has been shown to induce ALT activity in cells of mesenchymal origin [30]. However, ATRX depletion does not promote ALT activity in all cell types [31, 32] suggesting that ATRX LoF alone is not sufficient to induce ALT and that additional, as yet unidentified mechanisms are also needed. Reinforcing the notion that ALT arises as a result a combination of ATRX loss and other factors, it has recently been shown that during the immortalisation process, ATRX loss results in a progressive chromatin de-compaction and a gradual induction of telomere replication dysfunction which triggers an adaptive response eventually resulting in ALT activation [33]. Furthermore the authors show that the telomere dysfunction induced by ATRX loss cannot be overcome by endogenous telomerase activity.
Genetic alterations in the histone chaperone and ATRX binding partner DAXX (death domain associated protein) have also been shown to result in ALT due to a failure to localise ATRX to PML bodies [34]. In addition genetic alterations in tumour protein 53 (TP53) and retinoblastoma 1 (RB1) have also been associated with ALT [13] however the mechanistic basis for these associations is currently unknown.

Biomarkers of TMM

In large-scale studies telomerase expression data is often used as a surrogate biomarker for telomerase activity [13], however quantification of telomerase activity by standardised assays based on the telomere repeat amplification (TRAP) protocol is also feasible in clinical trial settings both as a predictive biomarker in tumour tissue and as pharmacodynamic biomarker in peripheral blood mononuclear cells [35].
The identification of robust biomarkers of ALT has proved more challenging. The gold standard assay for ALT is to confirm the maintenance of telomeres, in the absence of telomerase activity through successive population doublings. However, this is only practical in cell lines, non quantitative, and requires long-term cell culture [14]. Although telomere length and telomere heterogeneity are often used as biomarkers of ALT the lack of specificity of these assays for ALT is becoming increasingly apparent [14, 36, 37].
PML bodies are ubiquitous throughout the genome and responsible for diverse functions including DNA repair. In ALT cancer cells PML bodies specifically co-localise to telomeric DNA, and are thought to facilitate HR [14, 26]. ALT associated PML bodies are now a well-established biomarker of ALT activity and have the advantage that they can be visualised in formalin fixed paraffin embedded (FFPE) material [14, 38].
ALT cells are also characterised by the presence of c-circles: single stranded, telomeric circular DNA strands which are thought to provide a template for ALT telomere synthesis [14]. The c-circle assay is a rolling circle PCR amplification assay, based on the self-priming nature of c-circles. The c-circle assay requires fresh-frozen tissue but is advantageous as c-circles are quantifiable, specific for ALT and c-circle levels can be used to evaluate response to ALT targeted agents [39, 40].

Telomeres and telomere lengthening in neuroblastoma

Amplification of the MYCN oncogene is found in almost 40% of clinical high-risk neuroblastomas [4, 41], and is associated with up-regulation of TERT expression and telomere dysfunction [4, 42]. In an additional 23–31% of high-risk neuroblastomas, TERT is activated through chromosomal rearrangements involving 5p15.33, proximal to the TERT gene, which induces transcriptional up-regulation of TERT by juxtaposing the TERT coding sequence with strong enhancer elements [4, 5]. The third distinct sub-group, accounting for 24% of high-risk neuroblastoma cases are those with ALT [36]. Approximately half of ALT neuroblastomas are associated with somatic alterations in ATRX [4, 5, 36]. In neuroblastoma, genetic alterations in ATRX are associated with a distinct clinical phenotype including older age at diagnosis and a chronic progressive disease course [43].
Neuroblastoma harbouring a TMM is associated with a poor prognosis regardless of clinical stage [4, 5, 4446]. More recently it has been shown that in the presence of a TMM a concurrent mutation in a TP53 or RAS pathway associated gene is associated with an even worse prognosis [6]. Conversely, neuroblastoma with TP53 or RAS pathway mutations (including canonical ALK mutations) in the absence of a concurrent TMM are not associated with worse survival and can spontaneously regress [6]. This molecular risk stratification of neuroblastoma, as described by Ackermann et al. [6] is summarised in Fig. 2.
Although the somatic alterations driving TMM’s in the majority of neuroblastoma are well defined, high telomerase expression can occur in the absence of either a TERT translocation or MYCN amplification [6]. Also in the absence of an ATRX alteration the underlying drivers of ALT neuroblastoma are currently unknown, and somatic alterations in other genes, known to be associated with ALT in other malignancies are rarely found in neuroblastoma [47]. Numerous additional mechansims for telomere maintenance in neuroblastoma have been suggested. A study has identified interstitial telomeric sequences at sites of unbalanced translocations in neuroblastoma cell lines and postulated that these may contribute to a defective telomere maintenance pathway [48]. Another possibility is that genetic predisposition may contribute to the development of TMMs. In keeping with this, six common single nucleotide polymorphisms known to be associated with telomere lengthening have been found to be associated with an increased risk of neuroblastoma [49]. In keeping with the hypothesis that additional underlying predisposing factors may contribute to TMMs, a substantial increase in telomeric DNA damage and active telomere trimming has been described consistently across all high-risk neuroblastomas, regardless of telomerase or ALT status [50].
In summary, data is accumulating to support the hypothesis that TMMs are the common defining molecular feature of aggressive disease in the majority of children with clinical high-risk neuroblastoma. Therefore, both telomerase and ALT represent attractive targets for the development of novel therapeutic strategies with the potential to benefit a significant proportion of high-risk neuroblastoma patients. Here, we review the current pre-clinical and clinical research focused on targeting telomere maintenance with a specific focus on the potential relevance to patients with neuroblastoma.

Drugs targeting telomerase activity

Imetelstat (GRN163L) is a competitive telomerase inhibitor with a complementary structure to the template region of the RNA component of telomerase that binds to and blocks the active site of the enzyme [51]. Confirmation of target inhibition and pre-clinical efficacy of imetelstat has been demonstrated in multiple cancer subtypes [5254].
A phase I trial of imetelstat in 20 children with refractory or recurrent solid tumours demonstrated telomerase inhibition, which was sustained through to day 8 of cycle 1 of therapy [55]. Two of 16 patients had a partial response, however no responses were seen in any of the 6 patients with neuroblastoma enrolled on the trial. The main dose limiting toxicities were neutropenia, thrombocytopenia, and lymphopenia. A Phase II study on imetelstat in children with recurrent or refractory central nervous system malignancies was also associated with significant haematological toxicity and was discontinued after two children died of intra-tumoral haemorrhage [35], thus paediatric development for this compound has subsequently been discontinued.
BIBR1532 is a potent synthetic, non-nucleoside telomerase inhibitor [56]. However like imetelstat there are significant concerns regarding toxicity [57], and this agent has not yet been evaluated in clinical trials.
Sodium metaarsenite (KML001) binds to telomeric sequences, displacing hTERT from the nucleus into the cytoplasm [58] and has been shown to be cytotoxic in neuroblastoma cells in vitro [59]. In a phase I trial in adults with advanced solid tumours, objective responses to KML001 and cisplatin were seen in four out of 18 patients, however this trial was also discontinued due to toxicity [60].
Telomestatin stabilises G-quadruplexes, which in turn, inhibits telomerase activity [61]. This compound has been shown to induce apoptosis in-vitro in telomerase expressing neuroblastoma cell lines [62] but is not yet in clinical development.
6-thio-2′-deoxyguanosine (6-thio-dG) is a nucleoside analogue, which in telomerase active cells is recognised by telomerase and incorporated into telomeres resulting in telomere dysfunction [63]. Pre-clinical efficacy has been demonstrated in melanoma, non-small cell lung cancer and paediatric brain tumour models [6466]. Although 6-thio-dG has not yet been evaluated in clinical trials, due to the novel mechanism of action of the compound it is thought to be less toxic than traditional telomerase inhibitors [67]. 6-thio-dG has been shown to be effective in-vivo in neuroblastoma models with TERT activation. However, the response in MYCN amplified xenografts was mixed, likely reflective of the additional oncogenic pathways activated by MYCN [47]. This is a priority compound for further development for neuroblastoma (Table 1).
Table 1
Compounds shown pre-clinically to target telomere maintenance mechanisms, to be prioritised for evaluation in neuroblastoma
Drug
Target
Pre-clinical Data
Clinical Trials
6-thio-dG
Telomerase
Pre-clinical efficacy in TERT activated neuroblastoma models
No
Tetra-Pt (bpy)
ALT
In-vitro and in-vivo activity in U2OS ALT osteosarcoma model
No
CX-5461
ALT
In-vitro activity in U2OS and SAOS2 ALT osteosarcoma cell lines
On-going adult phase I clinical trial (NCT0271997)
Pifithrin-α
ALT
In-vitro and in-vivo activity in U2OS ALT osteosarcoma model
No
Trabectedin
ALT
In-vitro activity in a panel of ALT cell lines (sarcoma, breast cancer and melanoma)
-FDA approval for certain soft tissue sarcomas [68]
-Paediatric sarcoma phase II data [69]
-On-going paediatric clinical trials (NCT04067115)
XAV939 is an inhibitor of tankyrase, a positive regulator or telomerase. It has been shown to induce apoptosis in the telomerase expressing neuroblastoma cell line SH-SY5Y [70].

Drugs targeting ALT

Ataxia telangiectasia mutated (ATM) inhibitor combination therapy

It has been reported by Koneru et al. that ALT neuroblastoma cell lines are more resistant to topoisomerase inhibitors and that activation of ATM at ALT telomeres is associated with chemo-resistance. The ATM inhibitor AZD0156 was found to be synergistic with temozolomide and irinotecan therapy in both in vitro and in-vivo models of ALT neuroblastoma [71]. AZD0156 is currently in phase I trials in adults [72].

Ataxia telangiectasia and Rad3 related (ATR) inhibitor

In 2015 Flynn et al. reported that the presence of ALT renders cells hypersensitive to ATR inhibition in osteosarcoma models [73]. We have evaluated the clinical ATR inhibitor AZD6738 [74], in a panel of neuroblastoma cell lines and found that in contrast, ALT cell lines are relatively resistant to ATR inhibition in comparison to other neuroblastoma cell lines (Fig. 3a-b). This is in keeping with a subsequent publication which directly refuted the findings of Flynn et al. and concluded that differences in ATR inhibitor sensitivity were not related to ALT [77]. Taken together this data does not currently support a role of ATR inhibitors as an ALT specific therapy.
Conversely, we found that MYCN amplified neuroblastoma cell lines show a significantly greater in vitro sensitivity to single agent AZD6738 than non-MYCN amplified neuroblastoma cell lines (Fig. 3a-c), and a preliminary dose finding study demonstrated sensitivity to single agent AZD6738 in the Th-MYCN transgenic mouse model of MYCN driven neuroblastoma [76] (Fig. 3d-e). This is in keeping with other cMYC driven tumours where oncogene driven replicative stress results in a reliance on ATR signalling [78, 79]. Also in keeping with this, the SKNAS neuroblastoma cell line is relatively sensitive to AZD6738 (Fig. 3a) and known to over-express cMYC [80]. In MYCN amplified neuroblastoma models, ATR has also been proposed to play a role in resolution of transcription/replication conflicts [81].

Pre-clinical research in other ALT cancers

Recent pre-clinical data focused on the therapeutic targeting of ALT in other cancer subtypes may give important insight into potential therapeutic vulnerabilities that can be exploited for ALT neuroblastoma (Table 1): Tetra-Pt (bpy) is a cisplatin derivative that inhibits telomeric homologous recombination by targeting the telomeric G-quadruplex and has been shown to inhibit growth of ALT-cell xenograft tumours in mice [82]. Thus far this agent has not entered clinical trials nor has been evaluated in neuroblastoma models. CX5461 is an RNA polymerase I inhibitor which has been shown to selectively kill ATRX mutant cells due to its effects on ribosomal RNA transcription. In-vitro sensitivity in ALT cancer cell lines has been demonstrated [83]. It has also recently been reported that some ALT cancer cells rely on p53 and AKT activity to suppress apoptosis. Furthermore the authors go on to show that the p53 inhibitor pifithrin-α suppresses tumour growth in an ALT xenograft model [84] although others have shown that pifithrin-α is not a specific inhibitor of p53 [85], calling into question the underlying mechanism of the demonstrated pifithrin-α response. This strategy may however represent a relevant novel therapeutic opportunity for ALT neuroblastoma, particularly in view of the fact that the majority of neuroblastoma is TP53 wild type [86]. Finally, preliminary data has shown that ALT cancer cell lines are in general more sensitive to trabectedin, although the mechanism for this is not clear [87].

Tumour heterogeneity, evolution and TMM targeted therapeutics

In some other paediatric malignancies, ALT (identified by the presence of ALT associated PML bodies) and telomerase activation have been shown to co-exist in the same tumour [88, 89]. In neuroblastoma, intra-tumoural diversity of telomere length in individual tumours has been identified using quantitative telomere fluorescence in-situ hybridisation (FISH) [90], however this is not a sufficiently sensitive or specific biomarker of ALT activity [14, 36]. In fact, one study has identified a subset of neuroblastomas with extremely long telomeres in the absence of either telomerase activity or ALT (detected by c-circle assay and ALT associated PML bodies) [36]. Further studies have shown that although TERT alterations and MYCN amplification do co-exist in a small proportion of cases, there is no overlap between the telomerase expressing and ALT positive neuroblastoma [47]. This is in keeping with a recent publication also showing that ATRX mutations and MYCN amplification are synthetically lethal in neuroblastoma [91]. Taken together, the evidence in neuroblastoma to date is that the presence of either ALT or telomerase activation is associated with differing distinct genetic drivers and occurs in mutually exclusive nature [4, 5].
Although data thus far indicates that subgroups of neuroblastoma are driven by either telomerase or ALT activation, it is highly likely that a selective pressure targeting one TMM will support the emergence of an alternative mechanism. In multiple other cancer subtypes it has been shown that long-term telomerase inhibition results in the emergence of features consistent with ALT [9294]. Conversely, with the development of ALT targeted therapeutics it is probable that an up-regulation of telomerase will be seen. However, encouragingly, following ATRX LoF, it appears that ALT is a necessary adaption for cancer cell survival, and that reactivation of telomerase activity cannot overcome endogenous telomere dysfunction [33].
Finally although it has been shown that activation of a TMM is the key determinant of poor outcome in neuroblastoma, the key drivers of TMM’s in neuroblastoma; MYCN amplification and genetic alterations in ATRX are also associated with distinct patterns of wider transcriptional activation which drive malignant transformation [91, 95, 96]. Furthermore it is known that the co-occurrence of RAS/TP53 pathway alterations with a TMM is associated with a particularly dismal outcome [6] and accordingly, alterations in the RAS and TP53 pathways are enriched at the time of neuroblastoma relapse [97, 98]. Therefore, TMM targeting strategies will only be beneficial when given in combination with agents targeting these key pathways and the evaluation of combination therapies is urgently needed.

Conclusion

Despite evidence that telomere maintenance is a key driver of aggressive biology in neuroblastoma, clinical translation of novel therapeutics specifically targeting telomere maintenance remains extremely challenging. The only compound to make it into paediatric clinical trials so far, imetelstat is excessively toxic and pre-clinical data on other compounds targeting telomerase activity is extremely limited. The most promising telomerase targeting candidate to date is 6-thio-dG, however this agent is yet to make it into clinical trials. It must also be noted that in an aggressive malignancy such as neuroblastoma, rapid development of resistance to single-agent targeting of telomerase activation is highly likely and that combination therapies will be needed to overcome this. Also, in the case of MYCN amplified tumours, transcriptional up-regulation of TERT is only one of many oncogenic programmes up-regulated in MYCN amplified neuroblastoma cells.
For ALT driven cancers, there is preliminary data supporting specific roles of a handful of targeted therapeutic approaches but a dearth of robust evidence of pre-clinical efficacy specifically for neuroblastoma. The combination of ATM inhibition with chemotherapy is currently the most promising option for ALT neuroblastoma, although regimens combining cytotoxic chemotherapy agents and inhibition of master upstream regulators of DNA damage repair such as ATM are likely to be prone to significant toxicities.
Despite these challenges, the development of effective new strategies for neuroblastoma by either selective targeting of telomerase or ALT offers great potential to treat the underlying drivers of aggressive disease biology, and is applicable to the greater proportion of neuroblastoma patients. Furthermore, the hypothesis that TMM targeting strategies may be particularly effective in neuroblastoma is supported by the fact that significantly fewer mutations are found in neuroblastoma in comparison to adult malignancies, which often arise due to an accumulation of oncogenic mutations over time [99].
In addition, as recent data identifies an ‘ultra-high risk’ group of neuroblastoma patients with both telomere maintenance and mutations in RAS and TP53 pathway genes [6], rational combinations of telomere targeting agents with other targeted therapeutics must be sought. As pre-clinical data develops, rationally designed paediatric clinical trials will be required to personalise therapy to simultaneously target multiple drivers of aggressive biology in an individual patient.
As our understanding of the molecular drivers of fatal neuroblastoma has significantly expanded in recent years, pre-clinical research efforts must now focus on translating this knowledge into effective, less toxic new therapies for children with neuroblastoma.

Acknowledgements

Not applicable.
This study did not involve human participants, data or tissue. For mouse experiments, protocols were approved and monitored by The Institute of Cancer Research Animal Welfare and Ethical Review Body (PPL 70/7945, later PPL P91E52C32), in compliance with the UK Home Office Animals (Scientific Procedures) Act 1986.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Monclair T, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG Task Force report. J Clin Oncol. 2009;27(2):298–303.PubMedPubMedCentralCrossRef Monclair T, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG Task Force report. J Clin Oncol. 2009;27(2):298–303.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.PubMedCrossRef
10.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
13.
14.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
24.
25.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
43.
44.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
64.
65.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
87.
88.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
94.
95.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat Carr-Wilkinson J, et al. High Frequency of p53/MDM2/p14ARF Pathway Abnormalities in Relapsed Neuroblastoma. Clin Cancer Res. 2010;16(4):1108–18.PubMedPubMedCentralCrossRef Carr-Wilkinson J, et al. High Frequency of p53/MDM2/p14ARF Pathway Abnormalities in Relapsed Neuroblastoma. Clin Cancer Res. 2010;16(4):1108–18.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat 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
Metadaten
Titel
Novel therapeutic strategies targeting telomere maintenance mechanisms in high-risk neuroblastoma
verfasst von
S. L. George
V. Parmar
F. Lorenzi
L. V. Marshall
Y. Jamin
E. Poon
P. Angelini
L. Chesler
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01582-2

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.