Purpose:

Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors. Whereas most PPGLs are benign, up to 20% may become metastatic with SDHB- and FH-mutated tumors showing the higher risk. We aimed at determining the contribution of immortalization mechanisms to metastatic progression.

Experimental Design: Immortalization mechanisms were investigated in 200 tumors. To identify telomerase (+) tumors, we analyzed genomic alterations leading to transcriptional activation of TERT comprising promoter mutations, hypermethylation and gain copy number. To identify tumors that activated the alternative lengthening of telomere (ALT) mechanism, we combined analyses of telomere length by slot blot, telomere heterogeneity by telomere FISH, and ATRX mutations by next-generation sequencing. Univariate/multivariate and metastasis-free survival (MFS) and overall survival (OS) analyses were carried out for assessment of risk factors and clinical outcomes.

Results:

Only 37 of 200 (18.5%) tumors achieved immortalization. Telomerase activation occurred in 12 metastatic tumors and was prevalent in SDHB-mutated paragangliomas (P = 2.42e−09). ALT features were present in 25 tumors, mostly pheochromocytomas, regardless of metastatic status or molecular group (P = 0.169), yet ATRX mutations were found preferentially in SDHB/FH-mutated metastatic tumors (P = 0.0014). Telomerase activation and ATRX mutations were independent factors of poor prognosis: MFS (hazard ratio, 48.2 and 33.1; P = 6.50E−07 and 1.90E−07, respectively); OS (hazard ratio, 97.4 and 44.1; P = 4.30E−03 and 2.00E−03, respectively) and were associated with worse MFS and OS (log-rank tests P < 0.0001).

Conclusions:

Assessment of telomerase activation and ATRX mutations could be used to identify metastatic PPGLs, particularly in tumors at high risk of progression.

Translational Relevance

Pheochromocytomas and paragangliomas (PPGLs) are tumors of neural crest origin with SDHB/FH-mutated carriers showing a high risk of metastatic progression. At present, it is not possible to distinguish benign from metastatic PPGLs based on histopathologic features. A better understanding of the underlying mechanisms of metastatic PPGLs is critical for the successful identification of risk factors able to predict PPGL behavior. Here, we identified genomic alterations leading to telomerase activation (TERT promoter mutation/hypermethylation and gain copy number) and ATRX mutations in most metastatic PPGLs, particularly SDHB/FH-mutated tumors. Remarkably, these immortalization-related mechanisms were independent factors of poor prognosis associated with shorter metastasis-free and overall survival. Therefore, assessment of telomerase activation mechanisms and screening of ATRX mutations could be used in the clinical routine to discriminate metastatic from nonmetastatic PPGLs at high risk of progression.

Pheochromocytomas and paragangliomas (PPGLs) are neuroendocrine tumors arising from adrenal medulla and paraganglia, respectively. Although the majority of PPGLs never progress, it has been estimated that up to 20% can develop overt metastases (1).

Metastatic PPGLs represent a major clinical challenge due to the limitations in accurate diagnosis and effective treatments. Indeed, reliable tumor biomarkers are still lacking to distinguish, at the time of diagnosis, tumors that will remain benign from those that will progress to metastasis. Consequently, the diagnosis of metastasis remains the only definitive criterion to define malignant PPGLs according to the most recent World Health Organization (WHO) classification (2) and a life-long follow-up is recommended for every patient presenting with PPGLs, despite the fact that most are usually cured after surgery.

PPGLs are characterized by a remarkable genetic determinism with up to 40% of cases explained by germline mutations in 14 susceptibility genes comprising SDHA, SDHB, SDHC, SDHD, SDHAF2 (collectively referred to as SDHx), FH, MDH2, SLC25A11 (3), EPAS1, VHL, NF1, RET, TMEM127, and MAX (4, 5). Importantly, SDHB/FH mutations are associated with high risk of metastatic progression and poor prognosis (6–8), with about half of the mutation carriers presenting synchronous or metachronous metastases. Clinical characteristics such as primary tumor size and extra-adrenal location have been suggested as risk factors as well (9).

Genomics studies revealed that PPGL tumorigenesis is mainly driven by germline or somatic mutations in susceptibility genes, but they failed to identify recurrent genetic alterations linked to metastatic progression (10). Transcriptomic unsupervised classification established 3 main clusters: Krebs cycle cluster C1A, comprising tumors at high risk of metastatic progression (SDHx, FH-, MDH2- and SLC25A11-mutated), the pseudohypoxic cluster C1B mostly specific of VHL-mutated tumors, and the kinase signaling cluster C2 comprising NF1-, RET-, HRAS-, TMEM127-, MAX-mutated tumors (11).

We hypothesized that immortalization in primary PPGLs could be an important risk factor for progression toward metastasis. Immortalization is achieved by reactivation of telomerase in about 85% of human carcinomas (12) or by a recombination-based alternative lengthening of telomeres (ALT) pathway mainly in sarcomas and tumors arising from endocrine and neural tissues (13). The specific contribution of these immortalization mechanisms to PPGL progression remains undetermined.

Telomerase activation can be assessed by evaluation of TERT expression (14). However, given the possible lymphocytic infiltration in tumors, expression of TERT alone should be taken with caution and additional efforts are required to analyze underlying mechanisms such as TERT promoter mutations (15), hypermethylation (16), amplifications (17), and genomic rearrangements (15). On the other hand, the genetic basis of ALT tumors remains poorly defined, although mutations in ATRX have been reported to be associated with this phenotype (18).

Early studies reported telomerase overexpression in malignant pheochromocytomas (19–23), whereas TERT promoter mutations (24, 25) and hypermethylation (26) were recently found in a few SDHx-deficient paragangliomas. The phenotypic assignment of ALT based on telomeric analysis of histological preparations remains limited to 2 discrepant studies reporting a prevalence of 4% (27) and 27% (28) of 75 and 22 PPGLs, respectively. Also, mutations in ATRX, were reported to occur in 12.6% of PPGLs, some associated with ALT and aggressive features (28). However, fewer mutations were detected in exome sequencing studies (29–33). So far, no systematic study has concomitantly analyzed both telomere maintenance mechanisms, which is necessary to determine the actual prevalence of immortalization and its relative contribution for metastatic progression.

Here, we performed a comprehensive analysis of immortalization in a well-characterized series of 200 PPGLs by combining previous multiomics data (29) with experimental validations (Supplementary Fig. S1). We aimed at identifying telomerase (+) and ALT (+) tumors while ascertaining their association with underlying mechanisms, genetic status, clinical features and outcomes of affected patients. We provide solid evidence that telomerase activation and ATRX mutations help to discriminate metastatic from non-metastatic tumors in high-risk PPGLs, thus suggesting a great clinical potential for diagnostic and eventually for therapeutic purposes.

PPGL cohort

Two hundred tumor samples from 190 patients with PPGLs, collected by the French “Cortico et Médullosurrénale: les Tumeurs Endocrines” (COMETE) network (34) were analyzed in this study. The study was conducted in accordance with the Declaration of Helsinki. Ethical approval for the study was obtained from the institutional review board [Comité de Protection des Personnes (CPP) Ile de France III, June 2012]. Written informed consent for the sample collection and subsequent analyses was obtained from all patients. Mutation status (germline or somatic) for the main PPGL susceptibility genes and integrative genomic characterization of the cohort were previously reported (29). For this study, we also identified 7/200 (3.5%) MAML3 (+) tumors (Supplementary Fig. S2). Primer sequences for amplification of UBTFMAML3 fusion transcripts are provided in Supplementary Table S1.

Availability of data

The datasets analyzed during the current study are available in the following repositories: Gene-expression profiling (ArrayExpress entry E-MTAB-733); copy-number alterations (ArrayExpress entry E-MTAB-2817); whole-genome DNA methylation [Gene Expression Omnibus (GEO) entry GSE43298]. Whole-exome sequencing data [European Genome-phenome Archive (entry EGAS00001000933)].

RT-qPCR

Total RNA was purified with the miRNeasy Mini Kit (Qiagen) and cDNAs prepared with the Superscript III Kit (Thermo fisher scientific). Quantitative PCR was performed with iTaq Universal SYBR Green Supermix (Bio-Rad). Primer sequences are provided in Supplementary Table S1. For absolute quantifications of TERT variants, cDNA amplicons were purified from agarose gels using NucleoSpin Gel and PCR Clean-up Kit (Macherey-Nagel) to be used as standards. Cycling conditions were as reported (35).

TERT promoter mutations and methylation

Amplicons of 163bp were amplified from tumor DNAs using a set of primers listed in Supplementary Table S1 to target the TERT promoter region (chr5:1295151-1295313). PCR was performed with KAPA HiFi HotStart ReadyMix kit (Kapa Biosystems) using 50-ng genomic DNA. Sequencing reactions in both directions were performed in an Applied Biosystems 3730xl DNA Analyzer. TERT mutations were confirmed to be present only at the somatic level. For analysis of promoter methylation, pyrosequencing of a region of 36 bp containing 5 CpG sites (chr5:1295586-1295621;GRCh37/hg19) was amplified from sodium bisulfite modified genomic DNA and analyzed as described previously (16). Primers are listed in Supplementary Table S1.

Whole-genome sequencing

To identify TERT rearrangements, PCR-free libraries were prepared with NEBNext Ultra II DNA Library Prep Kit following supplier recommendations. Briefly, 300-ng double-strand gDNA from tumor sample CIT_015 were fragmented using a sonication method to obtain 400-bp average sizes. After ligation with paired-end adaptor oligonucleotides (xGen TS-LT Adapter Duplexes from IDT), fragments were purified for direct sequencing. DNA PCR-free library sequencing was performed on an Illumina HiSeq 4000 instrument with a paired-end read length of 150 nucleotides. Base calling and image analysis were performed using Illumina Real Time Analysis (RTA) Pipeline version 1.12.4.2 with default parameters. The Manta software was used for identification of somatic structural variants nearby the TERT locus.

Slot blot

50 ng of tumor DNAs were spotted onto Biodyne B nylon membranes in alkaline conditions, hybridized to an oligonucleotide labeled with digoxigenin (DIG) and revealed for the DIG signal following Roche instructions. After stripping, membranes were rehybridized with DIG labeled total genomic DNA from HCA2 primary cells. Quantification of telomeric and genomic signals was performed using the MultiGauge software. Telomeric intensities were normalized with the genomic signal to correct for the differences in ploidy.

APB assays

Detection of ALT-associated PML bodies (APBs) was based on colocalization of a telomeric FISH signal with the PML protein. Tissue sections (4 μm) were steamed with citrate buffer and serially incubated with anti-PML antibody (Chemicon, AB1370) diluted 1/1,000, and goat anti-rabbit alexa 488 antibody diluted 1/100. Slides were post fixed with 3.7% formaldehyde and dehydrated. Hybridization with a Cy3-labeled peptide nucleic acid (PNA) telomere probe (Panagene, F1002) was performed overnight. Slides were imaged with a Nikon 80i epifluorescence microscope. The following ALT characteristics were evaluated: (i) dramatic cell-to-cell telomere length heterogeneity, (ii) the presence of large, ultra-bright nuclear foci of telomere FISH signals in tumor cells, and (iii) colocalization of PML protein with telomeric FISH (APBs).

Screening of ATRX mutations

All coding regions of ATRX were amplified using the MASTR Plus technology (Agilent technologies) to allow their analysis using Next-Generation Sequencing (NGS). Amplicon libraries from different samples were further processed by bridge amplification followed by sequencing on the MiSeq Instrument using the MiSeq reagent kit v2, 300 cycles. NGS data were analyzed using the SeqNext module v4.3.1 (JSI medical systems) and the PolyDiag pipeline developed by the Paris Descartes bioinformatic platform. Mutations with a ratio >10% were considered relevant. ATRX mutations were confirmed to be present only at the somatic level.

Statistical analyses

Associations between molecular and clinical characteristics were evaluated with 2-sided χ2 or Fisher-exact tests with significance set to P < 0.05. Differential analysis was determined using moderated T-tests or ANOVA models for multigroup comparison. In case of significant P values for ANOVA tests, we calculated pairwise comparisons with corrections for multiple testing. Local false discovery rate was used to control for multiple testing using the Benjamini and Hochberg method. The significance of a Pearson coefficient computed on 2 quantitative covariates was estimated by a correlation test based on a Student t distribution of the Pearson correlation coefficient. Univariate and multivariate cox models were built to find covariates related to survival. Survival curves were calculated with the Kaplan–Meier method and differences between curves were determined using the log-rank test.

Patient and tumor characteristics

The study cohort consisted of 200 tumors corresponding to 166 (83%) pheochromocytomas, 28 (14%) paragangliomas, and 6 (3%) metastases, collected from 190 patients (115 women and 75 men) with a mean age at diagnosis of 42.53 years (range, 7–82 years). Twenty-three patients presented a metastatic disease at diagnosis or within the follow-up period (median of 7.7 years from the initial diagnosis) with an equal distribution between synchronous and metachronous metastases. The proportion of patients with metastatic disease was consistent with their genetic status: 52% were metastatic in the group at high risk of progression (12/23 SDHx/FH/SLC25A11-mutated, cluster C1A) and 6.6% in other molecular groups (11/167 non-SDHx/FH/SLC25A11-mutated, clusters C1B, C2A/B/C). Twenty-seven samples from metastatic patients correspond to 21 primary tumors and 6 metastases. Using genomic data previously generated for this cohort (29), we first searched which tumors achieved immortalization.

Telomerase activation is prevalent in SDHx-metastatic PPGLs

To identify telomerase (+) tumors we analyzed expression levels of TERT using transcriptome data (11). General expression levels of TERT were very low in all PPGLs. Anyhow, we found a significant increase in TERT in transcriptomic cluster C1A enriched in tumors at high risk of metastatic progression (SDHx/FH-mutated), compared with other clusters (Supplementary Fig. S3A). Importantly, SDHx metastatic cases display the highest TERT expression even when compared with nonmetastatic tumors with the same genotype (Supplementary Fig. S3B). Given that full-length TERT can be co-expressed with a splice variant lacking the catalytic domain, the so-called β-deletion (35), we performed absolute quantifications of these variants using RT-qPCR. This validation revealed equivalent abundance of both full-length and β-deletion transcripts (≥1.5 × 10−4 copies) in 9 of 14 metastatic tumors from the high-risk group (Fig. 1A) and in 3 of 10 metastatic tumors from the other molecular groups (Supplementary Fig. S3C).

Figure 1.

Expression of TERT and underlying mechanisms. A, Expression levels of full-length TERT (black) and β-deletion splice variant (gray) in all tumors from the group at high risk of progression (SDHx/FH-mutated, cluster C1A). Absolute transcript copy numbers of TERT splice variants were normalized by transcript numbers of GAPDH. Mean ± SD from 2 independent RT reactions. B, Chromatograms corresponding to the identified TERT promoter mutations in SDHx-mutated patients. At the bottom, primary tumor and matched metastases from the same patient. Samples from patients with metastatic disease are in red print. C, Correlation analysis of TERT expression (Affymetrix data) and promoter methylation levels (Illumina 27 K, probe cg02545192) in the group of tumors at high risk of progression. The Pearson correlation coefficient and the correlation test P value are at the top left. D, Schematic representation of Chr 5 amplifications involving the TERT locus in 2 SDHB-mutated metastatic PPGLs.

Figure 1.

Expression of TERT and underlying mechanisms. A, Expression levels of full-length TERT (black) and β-deletion splice variant (gray) in all tumors from the group at high risk of progression (SDHx/FH-mutated, cluster C1A). Absolute transcript copy numbers of TERT splice variants were normalized by transcript numbers of GAPDH. Mean ± SD from 2 independent RT reactions. B, Chromatograms corresponding to the identified TERT promoter mutations in SDHx-mutated patients. At the bottom, primary tumor and matched metastases from the same patient. Samples from patients with metastatic disease are in red print. C, Correlation analysis of TERT expression (Affymetrix data) and promoter methylation levels (Illumina 27 K, probe cg02545192) in the group of tumors at high risk of progression. The Pearson correlation coefficient and the correlation test P value are at the top left. D, Schematic representation of Chr 5 amplifications involving the TERT locus in 2 SDHB-mutated metastatic PPGLs.

Close modal

There are 3 major genetic/epigenetic alterations that can lead to transcriptional activation of TERT in tumors: enhancing promoter mutations (15), hypermethylation of the promoter region that includes the so so-called THOR region (TERT hypermethylated Oncological Region; ref. 16), and amplification of the locus (17). To determine the mechanisms underlying telomerase activation, we sequenced the TERT promoter region in tumor DNAs of the 200 PPGLs. We found 1 SDHC-mutated benign tumor carrying a non-enhancing C228A mutation (15) and 6 SDHx-mutated metastatic tumors carrying an enhancing C228T mutation (Fig. 1B). Interestingly, analysis of 3 samples from 1 of the individuals revealed that the C228T mutation was not present in the primary tumor, whereas it appeared in 1 out of the 2 metachronous metastases. Further analysis of methylation levels at the TERT promoter region using methylome data (36) revealed hypermethylation in 5 SDHB-mutated metastatic cases, including the aforementioned metachronous metastases (Supplementary Fig. S4A). Hypermethylation was significantly correlated with expression of TERT (Fig. 1C) and covered CpG islands spanning 4 Kb, including the so-called THOR region (ref. 16; Supplementary Fig. S4B), a finding confirmed using pyrosequencing (Supplementary Fig. S4C). Next, by examining the SNP array data, we identified copy-number gains involving the TERT locus (5p15.33) in 2 SDHB-mutated metastatic tumors (Fig. 1D). Interestingly, these samples also exhibit hypermethylation at the THOR region and presented the highest expression levels of TERT. Finally, no chromosomal rearrangements within 100 kb nearby the TERT locus were identified by whole-genome sequencing in the metastatic sporadic tumor CIT_015.

Taking into account a high co-expression of TERT splice variants, which was coupled to an underlying mechanism in most cases, we assigned a telomerase (+) status to 12 PPGLs, all of which were metastatic. Interestingly, 9 of 12 were tumors classified in the group at high risk of progression (P = 2.42e−09).

The ALT mechanism is activated irrespective of tumor subtype or metastatic status

ALT (+) tumors are characterized by displaying long and heterogeneous telomeres. We first estimated the telomere length of each tumor by applying slot blot analysis, and we focused on the 48 tumors exhibiting long telomeres, defined as those that fall above the third quartile (Fig. 2A). Next, samples for which material was available (n = 22 with long telomeres and n = 7 with intermediate/short telomeres used as controls) were assayed for telomere heterogeneity using APB assays (Fig. 2B; ref. 13). Twelve out of the 22 tumors with long telomeres were classified as ALT (+), whereas samples with intermediate/short telomeres were negative for the presence of APBs (Supplementary Table S2). We found that tumor samples with long telomeres harbor the lowest ATRX mRNA expression (Fig. 2C). Therefore, we used this criterion to assign the ALT (+) status to 9 additional tumors in which we could not perform APB assays.

Figure 2.

Identification of ALT features in PPGLs. A, Box-plot shows normalized telomere intensities (measured by slot blot) discretized in 3 groups: Short, intermediate, and long, defined with the first, second, and third quartiles, respectively. B, Screening for the presence of large telomeric foci by telomeric FISH (red) and costaining with PML (green) for detection of ALT-APBs (arrow). Average projections of representative images with long and short telomeres are shown. T.I., telomere intensity. Each image is composed of 16 deconvolved stacks, 200 μm each; scale bar, 10 μm. C, Correlation analysis of ATRX mRNA expression level (Affymetrix data, n = 187) and telomere size for the entire PPGL cohort. The Pearson correlation coefficient and the correlation test P value are shown. The first and third quartiles are indicated as grey dotted lines. Red dots indicate tumor samples displaying both long telomeres and low ATRX mRNA expression. D,TERRA expression levels relative to GAPDH as determined by RT-qPCR are higher in ALT (+) than in telomerase (+) PPGLs (*, P < 0.05).

Figure 2.

Identification of ALT features in PPGLs. A, Box-plot shows normalized telomere intensities (measured by slot blot) discretized in 3 groups: Short, intermediate, and long, defined with the first, second, and third quartiles, respectively. B, Screening for the presence of large telomeric foci by telomeric FISH (red) and costaining with PML (green) for detection of ALT-APBs (arrow). Average projections of representative images with long and short telomeres are shown. T.I., telomere intensity. Each image is composed of 16 deconvolved stacks, 200 μm each; scale bar, 10 μm. C, Correlation analysis of ATRX mRNA expression level (Affymetrix data, n = 187) and telomere size for the entire PPGL cohort. The Pearson correlation coefficient and the correlation test P value are shown. The first and third quartiles are indicated as grey dotted lines. Red dots indicate tumor samples displaying both long telomeres and low ATRX mRNA expression. D,TERRA expression levels relative to GAPDH as determined by RT-qPCR are higher in ALT (+) than in telomerase (+) PPGLs (*, P < 0.05).

Close modal

Finally, we sought to determine the prevalence of ATRX mutations using a next-generation sequencing approach. In addition to the tumor carrying a frameshift mutation in ATRX that we have described previously (29), we identified 7 PPGLs, with damaging mutations (Table 1). Among them, expression of ATRX was absent at the protein level as confirmed by immunohistochemistry in available tissues (Supplementary Fig. S5). ALT characteristics were present in 6 of 8 (75%) tumors harboring ATRX mutations (Table 1), of which 2 had already been identified in the APBs screening, and 4 were thus considered as new ALT (+) cases.

Table 1.

ATRX mutations identified in the PPGL cohort

Sample IDDriver PPGL mutationATRX cDNA mutationExon/IntronProtein alterationConsequenceLoss-of-functionTelomere intensity ± SEMAPB assayALT status
CIT_119 MET 6869A>G Exon 32 Asn2290Ser Missense Yes 2.39 ± 0.74 Positive ALT 
CIT_191 SDHB 3967_3970del Exon 12 Glu1323Glnfs*22 Frameshift Yes 2.26 ± 0.10 N.A. ALT 
CIT_121 FH 5793del Exon 25 Lys1936Argfs*19 Frameshift Yes 2.13 ± 0.22 Positive ALT 
CIT_073  5114_5122del Exon 19 Asn1705_Leu 1708delinsMet In frame N.D. 2.08 ± 0.08 N.A. Probably ALT 
CIT_078 VHL 7283A>G Exon 35 Tyr2428Cys Missense Predicted 1.52 ± 0.14 N.A. Probably ALT 
CIT_149 SDHB 4778C>T Exon 17 Ala1593Val Missense Predicted 1.39 ± 0.07 N.A. Probably ALT 
CIT_146  242+2T>A Intron 4 p.? Essential splicing Yes 1.16 ± 0.29 N.A. Probably WT 
CIT_085 SDHB 1470_1548del Exon 9 Gly491Leufs*45 Frameshift Yes 1.11 ± 0.22 N.A. Probably WT 
Sample IDDriver PPGL mutationATRX cDNA mutationExon/IntronProtein alterationConsequenceLoss-of-functionTelomere intensity ± SEMAPB assayALT status
CIT_119 MET 6869A>G Exon 32 Asn2290Ser Missense Yes 2.39 ± 0.74 Positive ALT 
CIT_191 SDHB 3967_3970del Exon 12 Glu1323Glnfs*22 Frameshift Yes 2.26 ± 0.10 N.A. ALT 
CIT_121 FH 5793del Exon 25 Lys1936Argfs*19 Frameshift Yes 2.13 ± 0.22 Positive ALT 
CIT_073  5114_5122del Exon 19 Asn1705_Leu 1708delinsMet In frame N.D. 2.08 ± 0.08 N.A. Probably ALT 
CIT_078 VHL 7283A>G Exon 35 Tyr2428Cys Missense Predicted 1.52 ± 0.14 N.A. Probably ALT 
CIT_149 SDHB 4778C>T Exon 17 Ala1593Val Missense Predicted 1.39 ± 0.07 N.A. Probably ALT 
CIT_146  242+2T>A Intron 4 p.? Essential splicing Yes 1.16 ± 0.29 N.A. Probably WT 
CIT_085 SDHB 1470_1548del Exon 9 Gly491Leufs*45 Frameshift Yes 1.11 ± 0.22 N.A. Probably WT 

NOTE: Next-generation sequencing identified 8/200 samples with ATRX mutations. Metastatic cases are highlighted in bold print. Positions of mutations with respect to their genomic location in cDNA, exon/intron positions and introduced changes at the protein level, as well as their functional consequences are specified. ALT (+) status was assigned on the basis of the presence of APBs or applying the combined criteria loss-of-function ATRX (predicted or determined by immunohistochemistry when possible) and long telomeres (telomere intensity ≥1.34).

Abbreviations: NA, not available material for analysis; ND, not determined; WT, Wild-type.

Altogether, we assigned the ALT (+) status to 25 out of the 200 (12.5%) PPGLs of the cohort. In support of an ALT phenotype, we found that these tumors exhibit, as expected, higher levels of telomere repeat-containing RNA (TERRA) than telomerase (+) samples (Fig. 2D). Of note, although the ALT mechanism was activated irrespective of metastatic status or tumor subtype (P = 0.169), ATRX mutations were more frequent in tumors at high risk of metastatic progression (P = 0.0014).

Telomerase activation and ATRX mutations are independent prognostic factors

The exhaustive analysis of immortalization enabled us to determine that only 37 of 200 (18.5%) PPGLs activated a telomere maintenance mechanism (Fig. 3A), probably explaining why the great majority of these tumors never progress. Interestingly, telomerase activation appears to be more frequent in paraganglioma than in pheochromocytoma and the opposite was found for ALT (+) tumors (Fig. 3A). Furthermore, telomerase (+) but not ALT (+) tumors had a larger size than nonimmortalized tumors (P < 0.05; Fig. 3B).

Figure 3.

Prevalence of immortalization mechanisms in PPGLs and association with clinical outcome. A, Distribution of telomere maintenance mechanisms among PPGL tumors (pie) and among tumor types (chart below). B, Box-plot of tumor size according to the immortalization status: telomerase (+), ALT (+), and nonimmortalized (wild-type) PPGLs (*, P < 0.05). C, MFS and (D) OS analyses of PPGLs for telomerase (+; red), ATRX-mutated (yellow) and wild-type tumors (blue) in the entire PPGL cohort. The log-rank test P values are shown.

Figure 3.

Prevalence of immortalization mechanisms in PPGLs and association with clinical outcome. A, Distribution of telomere maintenance mechanisms among PPGL tumors (pie) and among tumor types (chart below). B, Box-plot of tumor size according to the immortalization status: telomerase (+), ALT (+), and nonimmortalized (wild-type) PPGLs (*, P < 0.05). C, MFS and (D) OS analyses of PPGLs for telomerase (+; red), ATRX-mutated (yellow) and wild-type tumors (blue) in the entire PPGL cohort. The log-rank test P values are shown.

Close modal

It has been previously shown that tumor size (>5 cm), extra-adrenal location and SDHB-mutated status are clinical factors associated with metastatic progression and decreased overall survival (OS; ref. 9). Given that these variables appeared highly correlated with a telomerase (+) status, and given the tight association of ATRX mutations and the ALT phenotype in some metastatic tumors, we sought to identify which of these variables impact the most on the clinical outcome of affected patients.

Univariate Cox regression models showed a strong impact of all of these covariates except tumor size and ALT phenotype, on both metastasis-free survival (MFS) and OS. Remarkably, multivariate analysis performed on the remaining covariates revealed that only telomerase activation and ATRX mutations were independent risk factors: MFS (hazard ratio, 48.2 and 33.1; P = 6.50E−07 and 1.90E−07, respectively); OS (hazard ratio, 33.1 and 12.9; P = 2.60E−03 and 2.20E−03, respectively; Table 2). In fact, 18 out of 27 tumor samples (67%) from the 23 patients with confirmed metastatic status harbor 1 of these alterations regardless the tumor subtype. Of note, these alterations were present not only in 7 primary tumors from patients with synchronous metastases, but also in 7 primary tumors from 6 patients with metachronous metastases (Supplementary Table S3). When combined, detection of telomerase activation and ATRX mutations reaches the best sensitivity (0.70) and specificity (0.99; Supplementary Table S4), thus performing more accurately (AUC 0.84) than the previously suggested risk factors on the ability to discriminate metastatic from nonmetastatic PPGLs.

Table 2.

Univariate and multivariate cox analyses for MFS and OS

Univariate analysis (MFS)Multivariate analysis (MFS)
nEvent (n)HR (95% CI)PnHR (95% CI)P
Tumor size 152 18 0.455 (0.18–1.2) 0.1    
Tumor location 183 20 9.76 (4–24) 4.20E−07 183 1.2 (0.34–4.2) 0.78 
SDHB mutation 186 23 13.1 (5.7–30) 1.20E−09 183 1.51 (0.51–4.5) 0.45 
Telomerase (+) 186 23 28.9 (12–68) 1.00E−14 183 48.2 (10–220) 6.50E07 
ALT (+) 182 20 3.38 (1.3–8.8) 0.013    
ATRX mutation 186 23 10 (3.7–27) 6.20E−06 183 33.1 (8.9–120) 1.90E07 
 Univariate analysis (OS) Multivariate analysis (OS) 
 n Event (n) HR (95% CI) P n HR (95% CI) P 
Tumor size 151 0.413 (0.09–1.7) 0.230    
Tumor location 182 11 11.9 (3.4–42) 1.00E−04 182 1.34 (0.09–19) 0.83 
SDHB mutation 185 11 11.3 (3.4–37) 7.00E−05 182 1.06 (0.26–3.4) 0.93 
Telomerase (+) 185 11 39.3 (10–150) 1.10E−07 182 97.4 (4.2–2,300) 4.30E03 
ALT (+) 181 0.63 (0.07–5.1) 0.670    
ATRX mutation 185 11 6.68 (1.7–26) 6.50E−03 182 44.1 (4–490) 2.00E03 
Univariate analysis (MFS)Multivariate analysis (MFS)
nEvent (n)HR (95% CI)PnHR (95% CI)P
Tumor size 152 18 0.455 (0.18–1.2) 0.1    
Tumor location 183 20 9.76 (4–24) 4.20E−07 183 1.2 (0.34–4.2) 0.78 
SDHB mutation 186 23 13.1 (5.7–30) 1.20E−09 183 1.51 (0.51–4.5) 0.45 
Telomerase (+) 186 23 28.9 (12–68) 1.00E−14 183 48.2 (10–220) 6.50E07 
ALT (+) 182 20 3.38 (1.3–8.8) 0.013    
ATRX mutation 186 23 10 (3.7–27) 6.20E−06 183 33.1 (8.9–120) 1.90E07 
 Univariate analysis (OS) Multivariate analysis (OS) 
 n Event (n) HR (95% CI) P n HR (95% CI) P 
Tumor size 151 0.413 (0.09–1.7) 0.230    
Tumor location 182 11 11.9 (3.4–42) 1.00E−04 182 1.34 (0.09–19) 0.83 
SDHB mutation 185 11 11.3 (3.4–37) 7.00E−05 182 1.06 (0.26–3.4) 0.93 
Telomerase (+) 185 11 39.3 (10–150) 1.10E−07 182 97.4 (4.2–2,300) 4.30E03 
ALT (+) 181 0.63 (0.07–5.1) 0.670    
ATRX mutation 185 11 6.68 (1.7–26) 6.50E−03 182 44.1 (4–490) 2.00E03 

NOTE: Independent risk factors are highlighted in bold.

Abbreviations: CI, confidence interval; HR, hazard ratio; n, number of analyzed individuals.

To examine further the relationship between these alterations and prognosis, we performed survival analyses of the entire cohort. We found that patients with ATRX-mutated or telomerase (+) tumors exhibited a significantly shorter MFS and OS than patients without such alterations (log-rank tests P < 0.001; Fig. 3C and D). Similar results were obtained after excluding 6 metastatic samples from the analysis, of which 4 harbored these alterations (Supplementary Fig. S6). No statistically significant differences in MFS and OS between telomerase (+) and ATRX-mutated PPGLs were identified.

Strikingly, although the presence of telomerase activation and ATRX mutations greatly improved the stratification of patients at high risk of metastatic progression (SDHx/FH-mutated, cluster C1A; Fig. 4A–C), these alterations were present only in 5 out of the 13 (38.4%) metastatic tumors from the other molecular groups (clusters C1B, C2A/B/C; Fig. 4B and C). Therefore, we concluded that assessment of telomerase activation mechanisms and screening of ATRX mutations could be used to identify the metastatic potential of PPGLs, especially in tumors at high risk of progression.

Figure 4.

Telomerase activation and ATRX mutations improve the stratification of high-risk SDHB/FH-mutated PPGLs. A, MFS and OS analyses of patients with PPGLs with telomerase (+) or ATRX-mutated tumors (green) compared with wild-type tumors (blue) from the high-risk group (cluster C1A) or from the other clusters (B). The log-rank test P values are shown. C, Summary of the genomic alterations linked to TERT overexpression, ATRX mutations, and ALT status found in the analyzed PPGL cohort. The metastatic status (black) and the PPGL driver mutation of each tumor are given at the top.

Figure 4.

Telomerase activation and ATRX mutations improve the stratification of high-risk SDHB/FH-mutated PPGLs. A, MFS and OS analyses of patients with PPGLs with telomerase (+) or ATRX-mutated tumors (green) compared with wild-type tumors (blue) from the high-risk group (cluster C1A) or from the other clusters (B). The log-rank test P values are shown. C, Summary of the genomic alterations linked to TERT overexpression, ATRX mutations, and ALT status found in the analyzed PPGL cohort. The metastatic status (black) and the PPGL driver mutation of each tumor are given at the top.

Close modal

We here assigned a telomere maintenance mechanism to most tumor samples of the well-characterized cohort of 200 PPGLs collected by the COMETE network. We identified 12 telomerase (+) PPGLs, all of which having a metastatic status, and showed that both isoforms, full-length TERT and β-deletion, were overexpressed in these tumors. Although full-length is required for telomere maintenance, the catalytically dead β-deletion has also oncogenic functions (35, 37). Therefore, expression of both isoforms may promote cellular growth and progression to metastasis in PPGL tumors.

Mechanistically, hotspot mutations in the TERT promoter, C250T and C228T activate telomerase expression in human cancers (15) by creating a de novo binding motif for the transcription factor GABPA (38) and by driving an epigenetic switch (39, 40). We found 6 C228T hotspot promoter mutations exclusively in SDHx/FH-mutated metastatic tumors. This is consistent with previous studies, suggesting that although this mutation is not frequent in PPGLs, it can be found in SDHx-deficient tumors (24, 25, 41). Our study is the first to show that this recurrent mutation is associated with overt metastatic disease.

Hypermethylation has been linked to TERT overexpression in numerous tumor types (16, 42), sometimes associated with worse prognosis (43, 44). A recent report also suggested this hypermethylation to be restricted to metastatic paraganglioma (26). Here, we reinforce this observation and highlight its relevance from the standpoint of diagnosis because even though SDHx/FH-mutated tumors display a hypermethylator phenotype (36), this is the first time that a specific alteration in methylation is associated directly with the metastatic status. In addition, we observed that hypermethylation is not mutually exclusive with TERT promoter mutations and copy-number gains. In fact, it has been shown that genomic rearrangements involving the TERT locus also coexist with hypermethylation of the THOR region in high-risk neuroblastomas (45). These observations suggest that distinct genomic alterations cooperate in driving the transcriptional activation of TERT in PPGLs and that analysis of TERT promoter mutations and methylation could be useful in the clinical routine to identify metastatic PPGLs in high-risk tumors.

Regarding the ALT immortalization mechanism, discrepant prevalences of 4% PPGLs (27) and 27% PPGLs (28) were reported based only histological data. To clarify this issue, we used a combined analysis of APB assays and low expression of ATRX in tumors with long telomeres, which enabled us to estimate that 25 of 200 (12.5%) tumors, the great majority benign pheochromocytomas, activated the ALT mechanism.

We also find ATRX mutations in 8 of 200 PPGLs (4%), which contrasts with the original estimated prevalence of 12.6% (28). Accordingly, when exome-sequencing studies for PPGLs were revisited (Supplementary Table S5), the prevalence for ATRX mutations is 29 of 593 (4.8%). Of these, half have been associated with clinically aggressive behavior mostly in SDHx-mutated cases. Given that ATRX mutations are more frequent in this tumor subtype, this might explain why cohorts enriched for SDHx patients present a higher prevalence of ATRX mutations (28). We further found that only 6 of 25 (24%) ALT (+) PPGLs were linked to ATRX mutations and that these mutations had a stronger impact on poor prognosis than the ALT (+) status alone, particularly in PPGLs at high risk of metastatic progression (SDHx/FH-mutated, cluster C1A).

Our findings highlight a prominent role of immortalization-related mechanisms for the progression of neural-crest derived tumors, as noticed recently in neuroblastomas in which telomerase activation, ATRX mutations and MYCN amplifications define 3 nonoverlapping high-risk subgroups (46), or in gliomas in which mutations in the isocitrate dehydrogenase (IDH) and TERT/ATRX are also concurrent events that guide molecular classification and diagnosis (47).

Importantly, we could not assign any immortalization mechanism to 8 metastatic tumors, 6 of which from molecular groups C1B, C2A/B/C (Fig. 4C). This observation supports recent reports indicating that a subset of metastatic tumors, including melanomas and neuroblastomas could have progressed towards metastasis without activating a telomere maintenance mechanism (48, 49). On the contrary, our most striking result is that telomerase activation and ATRX mutations do impact the prognosis, particularly in the group at high risk of progression (cluster C1A). In fact, these alterations were present not only in metastatic samples and primary tumors from patients with synchronous metastases, but also in primary tumors long before the first metastasis appeared.

Interestingly, we found 1 patient with an SDHB mutation who developed 2 telomerase (+) metastases, even though his primary tumor operated on 7 years earlier was telomerase-negative (CIT_087). In addition, 1 patient presented with 2 primary tumors (CIT_073 and CIT_074), harboring an ATRX mutation and telomerase overexpression, respectively, 4 years before the first metastasis appeared. Thus, our results point these somatic alterations as key drivers of metastatic PPGLs. Nevertheless, we acknowledge caution with regard to the extent of heterogeneity within primary tumors of metastatic cases without evidence of telomerase activation or ATRX mutations. Given that extensive analyses of the whole tumor is impractical in the clinical routine, detection of these alterations in liquid biopsies would be of utmost importance to capture this tumor heterogeneity.

A key question in PPGL research is the identification of biomarkers able to distinguish between potentially metastatic and nonmetastatic tumors, which is crucial for diagnosis, treatment and follow-up. Although the present study is limited to the retrospective analysis of a modest sample of metastatic cases per molecular groups, our findings suggest that assessment of telomerase activation mechanisms (TERT promoter mutation/hypermethylation/copy-number gain or chromosomal rearrangement) and screening of ATRX mutations can identify potentially metastatic PPGLs, particularly in tumors carrying SDHx/FH mutations that are currently considered at high risk of progression. In addition, discrimination of immortalization mechanisms may become relevant to identify patients that would benefit from therapies targeting either telomerase or ALT (50). Prospective large multicenter studies will be required to address these issues, and to ascertain whether patients with SDHx-/FH–mutated PPGLs but without TERT activation or ATRX mutations would be henceforth considered as “low-risk” hereditary PPGLs for their surveillance.

No potential conflicts of interest were disclosed.

Conception and design: J. Favier, L.J. Castro-Vega, A.-P. Gimenez-Roqueplo

Development of methodology: I. Draskovic, L.J. Castro-Vega, A.-P. Gimenez-Roqueplo

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): I. Draskovic, A. Buffet, J. Cros, C. Lépine, T. Meatchi, M. Sibony, L. Amar, J. Bertherat, A. Londoño-Vallejo, J. Favier, L.J. Castro-Vega, A.-P. Gimenez-Roqueplo

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Job, I. Draskovic, N. Burnichon, A. Buffet, J. Cros, A. Venisse, V. Verkarre, A. de Reyniès, J. Favier, L.J. Castro-Vega

Writing, review, and/or revision of the manuscript: S. Job, I. Draskovic, N. Burnichon, J. Cros, L. Amar, J. Bertherat, A. de Reyniès, A. Londoño-Vallejo, J. Favier, L.J. Castro-Vega, A.-P. Gimenez-Roqueplo

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C. Lépine, A. Venisse, E. Robidel, J. Favier, A.-P. Gimenez-Roqueplo

Study supervision: J. Favier, L.J. Castro-Vega, A.-P. Gimenez-Roqueplo

A.-P. Gimenez-Roqueplo was supported by European Commission grants FP7 Research and Innovation funding program for 2007-2013 (n° 259735) and Horizon 2020 (n° 633983), and from Institut National du Cancer and Direction Générale de l'Offre de Soins (DGOS), Programme de Recherche Translationnelle en cancérologie (PRT-K 2014, COMETE-TACTIC, INCa_DGOS_8663). J. Favier received funding from Agence Nationale de la Recherche (ANR-2011-JCJC-00701 MODEOMAPP) and the Alliance nationale pour les sciences de la vie et de la santé (AVIESAN), Plan Cancer: Appel à projets Epigénétique et Cancer 2013 (EPIG201303 METABEPIC). N. Burnichon is funded by the Cancer Research for Personalized Medicine—CARPEM project (Site de Recherche Intégré sur le Cancer—SIRIC). The group is supported by the Ligue Nationale contre le Cancer (Equipe Labellisée). This work is part of the "Cartes d'Identité des Tumeurs (CIT) program" funded and developed by the Ligue Nationale contre le Cancer. Work in the Telomere and Cancer lab is supported by grants from La Ligue Nationale contre le Cancer (Equipe Labellisée) and Fondation ARC pour la recherche sur le cancer. We thank Profs Pierre-François Plouin and Xavier Bertagna for making this work possible through the COMETE Network, and to all members of the Genetics Department, Biological Resources Center and Tumor Bank Platform, Hôpital européen Georges Pompidou (BB-0033-00063), and of the Nikon Imaging Centre @ Institut Curie-CNRS (ANR-10-INSB-04) for technical support. We also thank Dr. Pedro Castelo-Branco and Dr. Joana Apolónio for providing technical assistance with pyrosequencing assay of the THOR region.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Lenders
JW
,
Duh
QY
,
Eisenhofer
G
,
Gimenez-Roqueplo
AP
,
Grebe
SK
,
Murad
MH
, et al
Pheochromocytoma and paraganglioma: an endocrine society clinical practice guideline
.
J Clin Endocrinol Metab
2014
;
99
:
1915
42
.
2.
Lloyd
RV
,
Osamura
RY
,
Kloppel
G
,
Rosai
J
.
WHO classification of tumours: pathology and genetics of tumours of endocrine organs
.
Lyon
:
IARC
; 
2017
.
3.
Buffet
A
,
Morin
A
,
Castro-Vega
LJ
,
Habarou
F
,
Lussey-Lepoutre
C
,
Letouze
E
, et al
Germline mutations in the mitochondrial 2-oxoglutarate/malate carrier SLC25A11 gene confer a predisposition to metastatic paragangliomas
.
Cancer Res
2018
;
78
:
1914
22
.
4.
Favier
J
,
Amar
L
,
Gimenez-Roqueplo
A
. 
Paraganglioma and phaeochromocytoma: from genetics to personalized medicine
.
Nat Rev Endocrinol
2015
;
11
:
101
11
.
5.
Dahia
PL
. 
Pheochromocytoma and paraganglioma pathogenesis: learning from genetic heterogeneity
.
Nat Rev Cancer
2014
;
14
:
108
19
.
6.
Amar
L
,
Bertherat
J
,
Baudin
E
,
Ajzenberg
C
,
Bressac-de Paillerets
B
,
Chabre
O
, et al
Genetic testing in pheochromocytoma or functional paraganglioma
.
J Clin Oncol
2005
;
23
:
8812
8
.
7.
Castro-Vega
LJ
,
Buffet
A
,
De Cubas
AA
,
Cascon
A
,
Menara
M
,
Khalifa
E
, et al
Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas
.
Hum Mol Genet
2014
;
23
:
2440
6
.
8.
Amar
L
,
Baudin
E
,
Burnichon
N
,
Peyrard
S
,
Silvera
S
,
Bertherat
J
, et al
Succinate dehydrogenase B gene mutations predict survival in patients with malignant pheochromocytomas or paragangliomas
.
J Clin Endocrinol Metab
2007
;
92
:
3822
8
.
9.
Ayala-Ramirez
M
,
Feng
L
,
Johnson
MM
,
Ejaz
S
,
Habra
MA
,
Rich
T
, et al
Clinical risk factors for malignancy and overall survival in patients with pheochromocytomas and sympathetic paragangliomas: primary tumor size and primary tumor location as prognostic indicators
.
J Clin Endocrinol Metab
2011
;
96
:
717
25
.
10.
Castro-Vega
LJ
,
Lepoutre-Lussey
C
,
Gimenez-Roqueplo
AP
,
Favier
J
. 
Rethinking pheochromocytomas and paragangliomas from a genomic perspective
.
Oncogene
2016
;
35
:
1080
9
.
11.
Burnichon
N
,
Vescovo
L
,
Amar
L
,
Libe
R
,
de Reynies
A
,
Venisse
A
, et al
Integrative genomic analysis reveals somatic mutations in pheochromocytoma and paraganglioma
.
Hum Mol Genet
2011
;
20
:
3974
85
.
12.
Shay
JW
,
Bacchetti
S
. 
A survey of telomerase activity in human cancer
.
Eur J Cancer
1997
;
33
:
787
91
.
13.
Dilley
RL
,
Greenberg
RA
. 
ALTernative telomere maintenance and cancer
.
Trends Cancer
2015
;
1
:
145
56
.
14.
Borah
S
,
Xi
L
,
Zaug
AJ
,
Powell
NM
,
Dancik
GM
,
Cohen
SB
, et al
Cancer. TERT promoter mutations and telomerase reactivation in urothelial cancer
.
Science
2015
;
347
:
1006
10
.
15.
Bell
RJ
,
Rube
HT
,
Xavier-Magalhaes
A
,
Costa
BM
,
Mancini
A
,
Song
JS
, et al
Understanding TERT promoter mutations: a common path to immortality
.
Mol Cancer Res
2016
;
14
:
315
23
.
16.
Castelo-Branco
P
,
Leao
R
,
Lipman
T
,
Campbell
B
,
Lee
D
,
Price
A
, et al
A cancer specific hypermethylation signature of the TERT promoter predicts biochemical relapse in prostate cancer: a retrospective cohort study
.
Oncotarget
2016
;
7
:
57726
36
.
17.
Zhang
A
,
Zheng
C
,
Hou
M
,
Lindvall
C
,
Wallin
KL
,
Angstrom
T
, et al
Amplification of the telomerase reverse transcriptase (hTERT) gene in cervical carcinomas
.
Genes Chromosomes Cancer
2002
;
34
:
269
75
.
18.
Lovejoy
CA
,
Li
W
,
Reisenweber
S
,
Thongthip
S
,
Bruno
J
,
de Lange
T
, et al
Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway
.
PLoS Genet
2012
;
8
:
e1002772
.
19.
Kinoshita
H
,
Ogawa
O
,
Mishina
M
,
Oka
H
,
Okumura
K
,
Yamabe
H
, et al
Telomerase activity in adrenal cortical tumors and pheochromocytomas with reference to clinicopathologic features
.
Urol Res
1998
;
26
:
29
32
.
20.
Kubota
Y
,
Nakada
T
,
Sasagawa
I
,
Yanai
H
,
Itoh
K
. 
Elevated levels of telomerase activity in malignant pheochromocytoma
.
Cancer
1998
;
82
:
176
9
.
21.
Boltze
C
,
Mundschenk
J
,
Unger
N
,
Schneider-Stock
R
,
Peters
B
,
Mawrin
C
, et al
Expression profile of the telomeric complex discriminates between benign and malignant pheochromocytoma
.
J Clin Endocrinol Metab
2003
;
88
:
4280
6
.
22.
Isobe
K
,
Yashiro
T
,
Omura
S
,
Kaneko
M
,
Kaneko
S
,
Kamma
H
, et al
Expression of the human telomerase reverse transcriptase in pheochromocytoma and neuroblastoma tissues
.
Endocr J
2004
;
51
:
47
52
.
23.
Luo
Z
,
Li
J
,
Qin
Y
,
Ma
Y
,
Liang
X
,
Xian
J
, et al
Differential expression of human telomerase catalytic subunit mRNA by in situ hybridization in pheochromocytomas
.
Endocr Pathol
2006
;
17
:
387
98
.
24.
Liu
T
,
Brown
TC
,
Juhlin
CC
,
Andreasson
A
,
Wang
N
,
Backdahl
M
, et al
The activating TERT promoter mutation C228T is recurrent in subsets of adrenal tumors
.
Endocr Relat Cancer
2014
;
21
:
427
34
.
25.
Papathomas
TG
,
Oudijk
L
,
Zwarthoff
EC
,
Post
E
,
Duijkers
FA
,
van Noesel
MM
, et al
Telomerase reverse transcriptase promoter mutations in tumors originating from the adrenal gland and extra-adrenal paraganglia
.
Endocr Relat Cancer
2014
;
21
:
653
61
.
26.
Svahn
F
,
Juhlin
CC
,
Paulsson
JO
,
Fotouhi
O
,
Zedenius
J
,
Larsson
C
, et al
Telomerase reverse transcriptase promoter hypermethylation is associated with metastatic disease in abdominal paraganglioma
.
Clin Endocrinol (Oxf)
2018
;
88
:
343
45
.
27.
Heaphy
CM
,
Subhawong
AP
,
Hong
SM
,
Goggins
MG
,
Montgomery
EA
,
Gabrielson
E
, et al
Prevalence of the alternative lengthening of telomeres telomere maintenance mechanism in human cancer subtypes
.
Am J Pathol
2011
;
179
:
1608
15
.
28.
Fishbein
L
,
Khare
S
,
Wubbenhorst
B
,
DeSloover
D
,
D'Andrea
K
,
Merrill
S
, et al
Whole-exome sequencing identifies somatic ATRX mutations in pheochromocytomas and paragangliomas
.
Nat Commun
2015
;
6
:
6140
.
29.
Castro-Vega
LJ
,
Letouze
E
,
Burnichon
N
,
Buffet
A
,
Disderot
PH
,
Khalifa
E
, et al
Multi-omics analysis defines core genomic alterations in pheochromocytomas and paragangliomas
.
Nat Commun
2015
;
6
:
6044
.
30.
Flynn
A
,
Benn
D
,
Clifton-Bligh
R
,
Robinson
B
,
Trainer
AH
,
James
P
, et al
The genomic landscape of phaeochromocytoma
.
J Pathol
2015
;
236
:
78
89
.
31.
Juhlin
CC
,
Stenman
A
,
Haglund
F
,
Clark
VE
,
Brown
TC
,
Baranoski
J
, et al
Whole-exome sequencing defines the mutational landscape of pheochromocytoma and identifies KMT2D as a recurrently mutated gene
.
Genes Chromosomes Cancer
2015
;
54
:
542
54
.
32.
Toledo
RA
,
Qin
Y
,
Cheng
ZM
,
Gao
Q
,
Iwata
S
,
Silva
GM
, et al
Recurrent mutations of chromatin-remodeling genes and kinase receptors in pheochromocytomas and paragangliomas
.
Clin Cancer Res
2016
;
22
:
2301
10
.
33.
Fishbein
L
,
Leshchiner
I
,
Walter
V
,
Danilova
L
,
Robertson
AG
,
Johnson
AR
, et al
Comprehensive molecular characterization of pheochromocytoma and paraganglioma
.
Cancer Cell
2017
;
31
:
181
93
.
34.
Plouin
PF
,
Gimenez-Roqueplo
AP
,
Bertagna
X
. 
[COMETE, a network for the study and management of hypersecreting adrenal tumors]
.
Bull Acad Natl Med
2008
;
192
:
73
82
.
35.
Listerman
I
,
Sun
J
,
Gazzaniga
FS
,
Lukas
JL
,
Blackburn
EH
. 
The major reverse transcriptase-incompetent splice variant of the human telomerase protein inhibits telomerase activity but protects from apoptosis
.
Cancer Res
2013
;
73
:
2817
28
.
36.
Letouze
E
,
Martinelli
C
,
Loriot
C
,
Burnichon
N
,
Abermil
N
,
Ottolenghi
C
, et al
SDH mutations establish a hypermethylator phenotype in paraganglioma
.
Cancer cell
2013
;
23
:
739
52
.
37.
Hrdlickova
R
,
Nehyba
J
,
Bose
HR
 Jr.
Alternatively spliced telomerase reverse transcriptase variants lacking telomerase activity stimulate cell proliferation
.
Mol Cell Biol
2012
;
32
:
4283
96
.
38.
Bell
RJ
,
Rube
HT
,
Kreig
A
,
Mancini
A
,
Fouse
SD
,
Nagarajan
RP
, et al
Cancer. The transcription factor GABP selectively binds and activates the mutant TERT promoter in cancer
.
Science
2015
;
348
:
1036
9
.
39.
Stern
JL
,
Theodorescu
D
,
Vogelstein
B
,
Papadopoulos
N
,
Cech
TR
. 
Mutation of the TERT promoter, switch to active chromatin, and monoallelic TERT expression in multiple cancers
.
Genes Dev
2015
;
29
:
2219
24
.
40.
Akincilar
SC
,
Khattar
E
,
Boon
PL
,
Unal
B
,
Fullwood
MJ
,
Tergaonkar
V
. 
Long-Range chromatin interactions drive mutant TERT promoter activation
.
Cancer Discov
2016
;
6
:
1276
91
.
41.
Vinagre
J
,
Almeida
A
,
Populo
H
,
Batista
R
,
Lyra
J
,
Pinto
V
, et al
Frequency of TERT promoter mutations in human cancers
.
Nat Commun
2013
;
4
:
2185
.
42.
Barthel
FP
,
Wei
W
,
Tang
M
,
Martinez-Ledesma
E
,
Hu
X
,
Amin
SB
, et al
Systematic analysis of telomere length and somatic alterations in 31 cancer types
.
Nat Genet
2017
;
49
:
349
57
.
43.
Castelo-Branco
P
,
Choufani
S
,
Mack
S
,
Gallagher
D
,
Zhang
C
,
Lipman
T
, et al
Methylation of the TERT promoter and risk stratification of childhood brain tumours: an integrative genomic and molecular study
.
Lancet Oncol
2013
;
14
:
534
42
.
44.
Bougel
S
,
Lhermitte
B
,
Gallagher
G
,
de Flaugergues
JC
,
Janzer
RC
,
Benhattar
J
. 
Methylation of the hTERT promoter: a novel cancer biomarker for leptomeningeal metastasis detection in cerebrospinal fluids
.
Clin Cancer Res
2013
;
19
:
2216
23
.
45.
Peifer
M
,
Hertwig
F
,
Roels
F
,
Dreidax
D
,
Gartlgruber
M
,
Menon
R
, et al
Telomerase activation by genomic rearrangements in high-risk neuroblastoma
.
Nature
2015
;
526
:
700
4
.
46.
Valentijn
LJ
,
Koster
J
,
Zwijnenburg
DA
,
Hasselt
NE
,
van Sluis
P
,
Volckmann
R
, et al
TERT rearrangements are frequent in neuroblastoma and identify aggressive tumors
.
Nature Genet
2015
;
47
:
1411
4
.
47.
Cancer Genome Atlas Research N
,
Brat
DJ
,
Verhaak
RG
,
Aldape
KD
,
Yung
WK
,
Salama
SR
, et al
Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas
.
N Engl J Med
2015
;
372
:
2481
98
.
48.
Dagg
RA
,
Pickett
HA
,
Neumann
AA
,
Napier
CE
,
Henson
JD
,
Teber
ET
, et al
Extensive proliferation of human cancer cells with ever-shorter telomeres
.
Cell Rep
2017
;
19
:
2544
56
.
49.
Viceconte
N
,
Dheur
MS
,
Majerova
E
,
Pierreux
CE
,
Baurain
JF
,
van Baren
N
, et al
Highly aggressive metastatic melanoma cells unable to maintain telomere length
.
Cell Rep
2017
;
19
:
2529
43
.
50.
Reddel
RR
. 
Telomere maintenance mechanisms in cancer: clinical implications
.
Curr Pharm Des
2014
;
20
:
6361
74
.

Supplementary data