Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

TERT expression is associated with metastasis from thin primaries, exhausted CD4+ T cells in melanoma and with DNA repair across cancer entities

  • Christina Katharina Kuhn ,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Susanne.Horn@medizin.uni-leipzig.de (SH); Christina.Kuhn@medizin.uni-leipzig.de (CKK)

    Affiliation Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Medical Faculty, Leipzig, Germany

  • Jaroslawna Meister,

    Roles Writing – original draft, Writing – review & editing

    Affiliations Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Medical Faculty, Leipzig, Germany, Institute for Clinical Diabetology, German Diabetes Centre, Leibniz Centre for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany

  • Sophia Kreft,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium Partner Site Essen/Düsseldorf, Essen, Germany

  • Mathias Stiller,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Institute of Pathology, University of Leipzig Medical Center, Leipzig, Germany

  • Sven-Holger Puppel,

    Roles Data curation, Resources, Writing – review & editing

    Affiliation Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Medical Faculty, Leipzig, Germany

  • Anne Zaremba,

    Roles Writing – original draft, Writing – review & editing

    Affiliation Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium Partner Site Essen/Düsseldorf, Essen, Germany

  • Björn Scheffler,

    Roles Writing – original draft, Writing – review & editing

    Affiliation DKFZ-Division Translational Neurooncology at the West German Cancer Center, University Hospital Essen/University of Duisburg-Essen, Essen, Germany

  • Vivien Ullrich,

    Roles Writing – original draft, Writing – review & editing

    Affiliation DKFZ-Division Translational Neurooncology at the West German Cancer Center, University Hospital Essen/University of Duisburg-Essen, Essen, Germany

  • Torsten Schöneberg,

    Roles Funding acquisition, Project administration, Supervision

    Affiliations Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Medical Faculty, Leipzig, Germany, School of Medicine, University of Global Health Equity, Kigali, Rwanda

  • Dirk Schadendorf,

    Roles Funding acquisition, Project administration

    Affiliation Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium Partner Site Essen/Düsseldorf, Essen, Germany

  • Susanne Horn

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    Susanne.Horn@medizin.uni-leipzig.de (SH); Christina.Kuhn@medizin.uni-leipzig.de (CKK)

    Affiliations Rudolf Schönheimer Institute of Biochemistry, University of Leipzig, Medical Faculty, Leipzig, Germany, Department of Dermatology, University Hospital Essen, University Duisburg-Essen, and German Cancer Consortium Partner Site Essen/Düsseldorf, Essen, Germany

Abstract

Telomerase reverse transcriptase (TERT) promoter mutations occur frequently in cancer, have been associated with increased TERT expression and cell proliferation, and could potentially influence therapeutic regimens for melanoma. As the role of TERT expression in malignant melanoma and the non-canonical functions of TERT remain understudied, we aimed to extend the current knowledge on the impact of TERT promoter mutations and expression alterations in tumor progression by analyzing several highly annotated melanoma cohorts. Using multivariate models, we found no consistent association for TERT promoter mutations or TERT expression with the survival rate in melanoma cohorts under immune checkpoint inhibition. However, the presence of CD4+ T cells increased with TERT expression and correlated with the expression of exhaustion markers. While the frequency of promoter mutations did not change with Breslow thickness, TERT expression was increased in metastases arising from thinner primaries. As single-cell RNA-sequencing (RNA-seq) showed that TERT expression was associated with genes involved in cell migration and dynamics of the extracellular matrix, this suggests a role of TERT during invasion and metastasis. Co-regulated genes found in several bulk tumors and single-cell RNA-seq cohorts also indicated non-canonical functions of TERT related to mitochondrial DNA stability and nuclear DNA repair. This pattern was also evident in glioblastoma and across other entities. Hence, our study adds to the role of TERT expression in cancer metastasis and potentially also immune resistance.

Introduction

Telomere maintenance mechanisms play a critical role in cellular survival as they are known to enhance the proliferative capacity of cancer cells [1]. Frequent mutations in the promoter region of the telomerase reverse transcriptase (TERT) gene were shown to be associated with the upregulation of telomerase expression [24]. TERT promoter mutations have been proposed as independent prognostic markers in non-acral cutaneous melanoma, since they correlated in a multivariate analysis with decreased overall survival (OS) without analysis of therapy [5]. The co-occurrence of BRAF or NRAS mutations with TERT promoter mutations was also prognostic for poor disease-free survival in multivariate analyses in primary melanoma [6]. TERT promoter mutations may serve as predictive biomarkers as they were independently associated with improved survival in patients receiving BRAF/MEK inhibitor therapy in univariate and multivariate analyses [7, 8]. Also, patients undergoing anti-CTLA4 therapy showed better overall survival with TERT promoter mutations, implying its potential as a predictive marker for immune checkpoint inhibitor (ICI) therapy [9]. Also, TERT expression has been suggested to be an independent prognostic marker associated with poor survival in human cancer patients beyond melanoma [1012]. However, many variables, such as mutational load, tumor heterogeneity, and prior therapies, could be appended to existing multivariate models to test additional potentially stronger predictors.

The extent to which genetic alterations of TERT play a role in cancer progression and the molecular mechanisms affected by reactivation of TERT transcription vary widely among cancer entities, making it crucial to understand the underlying mechanisms [13]. One way cancer cells regain their proliferative ability is by forming a new ETS binding motif at the promoter mutation [2]. This binding motif attracts transcription factors, such as TCF1, Myc, Wnt/β-catenin, NF-kB, and GAPB, which are responsible for regulating numerous cellular processes including tumorigenesis [2, 14]. The canonical function of TERT is telomere maintenance, however, non-canonical functions of TERT have been discussed, e.g. related to reactive oxygen species (ROS), mitochondria and aging. TERT is imported into the mitochondria, where it binds mitochondrial DNA, protecting it from oxidative stress–induced damage, reducing apoptosis in human endothelial cells [15]. Furthermore, besides the full-length expressed TERT, alternatively spliced isoforms exist, which lack telomerase activity and may account for up to 10% of endogenous TERT. These are continuously expressed after telomerase silencing during human embryonic development [16] and are potentially associated with telomere-independent effects [17].

Here, we test if TERT promoter mutations and TERT expression are associated with OS and progression free survival (PFS) under ICI, as well as clinicopathological and demographic parameters in multiple highly annotated melanoma cohorts with exome- and transcriptome sequencing. To examine the non-canonical functions of TERT, we investigate associations with ROS-related genes, hypoxia, chromothripsis, and TERT isoform expression. Using molecular pathway analyses, we replicate known and identify new signaling pathways regulated with genetically modified TERT in bulk tumors, cell lines, and single-cells. Tumor immune signatures are estimated to elucidate the role of TERT during tumor rejection. Finally, our analyses are extended to additional cancer entities from the Pan-Cancer Analysis of Whole Genomes (PCAWG) cohort and glioblastoma (GBM) to determine the signaling pathways co-regulated with TERT across entities.

Materials and methods

Next generation sequencing data analysis

We analyzed several bulk RNA-seq cohorts with respective survival data (Table 1, Transcriptomics in S1 File). These included five melanoma cohorts with samples taken prior to or early-on ICI therapy (Gide, Hugo, Liu, Riaz, Van Allen cohorts), one melanoma cohort without ICI therapy (Skin Cutaneous Melanoma cohort, SKCM), two cross-entity datasets: cell lines from the Cancer Cell Line Encyclopedia (CCLE), and bulk tumors of the Pan-Cancer Analysis of Whole Genomes (PCAWG). We further included melanoma single-cell RNA-seq (scRNA-seq) and glioblastoma tumors in our workflow (S1 Figin S1 File). In brief, we extended the available gene expression levels with additional metadata, as described below and in the S1 File. We annotated published TERT promoter mutations in the RNA-seq datasets of two melanoma cohorts (Van Allen, SKCM) and two cross-entity datasets (CCLE and PCAWG, TERT promoter mutation calls in S1 Material). We added immune signatures for the six melanoma bulk RNA-seq cohorts using TIMER 2.0 [18] (Immune signatures in S1 Material). Isoform analysis was performed for the Van Allen, SKCM and PCAWG cohorts. To analyze mitochondria-related features in the PCAWG and SKCM cohorts, we further integrated measurements for hypoxia, chromothripsis [19] and mitochondrial mutations [19, 20]. Hypoxia scores for PCAWG were determined as in Buffa et al. 2010 [21], and hypoxia scores for the SKCM cohort were obtained from the cBioPortal (www.cbioportal.org/study/clinicalData?id=skcm_tcga_pan_can_atlas_2018). We reanalyzed the MSK IMPACT Clinical Sequencing Cohort (dataset “MSKCC, Nat Med 2017”) [9] as obtained from www.cbioportal.org annotating the corresponding therapy data from Samstein [22].

Enrichment analysis

Enrichment analysis of top genes associated with TERT promoter mutation and TERT expression using bulk RNA-seq cohorts was conducted using the gene set enrichment analysis tool https://maayanlab.cloud/Enrichr/ [23] with default settings. For enrichment analysis in the melanoma single-cell context, the top 100 genes associated with TERT expression in the scRNA-seq dataset were queried with Enrichr using default settings. TERT promoter mutation status was not available for single-cell data. The top ten results of each enrichment set from gene ontology pathways, ontologies and cell types were then used for discussion of the most frequently occurring terms.

TERT isoform analysis

Short names, coding information, biotype, and transcript lengths of TERT isoforms were collected from Ensembl (GRCh38.p13) using ENSG gene identifiers (TERT isoform information in S1 Material). The presence of the mitochondrial targeting protein sequence ‘MPRAPRCRAVRSLLRSHYRE’ [24] in the translated isoforms was analyzed using the UCSC Human Genome Browser. In detail, we verified whether transcripts included the targeting sequence by aligning the protein sequence with the genome (GRCh38.p13) and comparing the start/stop positions with the isoform exons. The isoform expressed in normal control tissues was assessed using the GTEx portal (https://gtexportal.org/home/gene/TERT#gene-transcript-browser-block, S2 Fig in S1 File). We then summed over the measured transcripts per million (TPM) of all isoforms and calculated the proportion of isoforms with mitochondrial targeting sequence. Data on isoforms lacking the targeting sequence were available only for the CCLE and Van Allen cohorts.

Statistics

Correlation of TERT mRNA expression, isoform expression and clinical variables was tested using Spearman correlations. For comparison between samples with or without TERT promoter mutation, two-sided Wilcoxon rank tests were applied. The correlates associated with TERT expression or TERT promoter mutations were ranked by p-value with false discovery rate control, and overlaps of the gene lists from several cohorts were performed. For survival analysis, the cohorts were divided into low vs. high TERT expression groups, with approximately one-third of the patients showing high TERT expression. If available, cohorts (SKCM and Van Allen cohort) were also divided into two groups by TERT promoter mutation absence/presence. Kaplan-Meier survival curves, log-rank tests, multivariate Cox Proportional-Hazards (Cox PH) analyses, and odds ratios were computed using R v.4.0.1 [25].

Data availability statement

The data used in this study are available within this manuscript and its supplementary files. Previously published, processed next generation sequencing data can be accessed at https://tools.hornlab.org/cru337phenotime/.

Results

In each of the following sections, we start at the genomic level by analyzing the role of TERT promoter mutations in malignant melanoma and extend the analysis to the transcriptomic level by examining the impact of TERT expression on clinical and molecular associations.

Association of TERT promoter mutation status and TERT expression with survival

TERT promoter mutations and TERT expression are proposed as prognostic biomarkers in melanoma and TERT promoter mutations might be predictive under anti-CTLA4 therapy [5, 6, 9, 26, 27]. Therefore we investigated the impact of both TERT alterations using univariate and multivariate survival analyses based on six bulk RNA-seq cohorts of metastatic melanoma treated with ICI (Gide, Riaz, Van Allen, Liu, Hugo cohorts) and without targeted therapy or ICI (SKCM). Our analysis showed that TERT promoter mutations and TERT expression were not associated with survival (S3-S5 Figs, S1 Table). PFS under anti-PD1 immunotherapy with pembrolizumab appeared decreased with higher TERT expression in only one analyzed cohort with small sample size (n = 8 vs. 24, Gide cohort, PFS log rank p = 0.038), however, it was not significantly associated in multivariate Cox PH including age, sex and BRAF mutation status (p = 0.071, S1A Table).

In contrast to our finding, an association of TERT promoter mutations with survival under anti-CTLA4 therapy was described previously [9], however, no multivariate model was attempted. Therefore, we reanalyzed the melanoma subset of the MSK Impact 2017 cohort [9] and the corresponding therapy data and included tumor mutational burden (TMB), sex, and BRAFV600 variants in a multivariate model. The previously described association of TERT promoter mutations with survival still holds, albeit with reduced significance (p = 0.015 compared to the previous univariate analysis p = 0.0007, S6A Fig in S1 File).

TERT expression and increased intratumoral CD4+ T helper type 1 cells

To investigate the role of TERT during tumor rejection, we correlated immune signatures with TERT expression. The CD4+ T helper type 1 cell signature repeatedly increased with TERT in five of the six cohorts analyzed here (Fig 1A, S2 Table). However, within a patient population with high CD4+ T helper type 1 cell estimates, survival did not differ with elevated TERT expression (S7 Fig in S1 File). To find a possible interaction of CD4+ T cells, we tested the presence of potential exhaustion markers [28]. Indeed, CTLA4, PDCD1IPD1, HAVCR2/TIM3, and TIGIT were expressed at higher levels (Fig 1B) in CD4+ T cells of tumors with high TERT expression of malignant cells.

thumbnail
Fig 1. TERT expression and immune signatures.

(A) Five out of six cohorts showed correlation of TERT expression and a CD4+ Th1 T cell immune signature (xCell assay). (B) Expression of T cell exhaustion markers in single CD4+ T cells of patients with low vs. high TERT expression in malignant tumor cells (</>0.05 TPM). Jerby-Arnon cohort, pre-therapy samples. Th1: T-helper type 1; TME: tumor microenvironment; TPM: transcripts per million; xCell: xCell cell types enrichment analysis. The figure was created with BioRender.com.

https://doi.org/10.1371/journal.pone.0281487.g001

Primary tumor thickness

Our examination of clinical variables in association with TERT promoter mutations and expression was based on the two largest melanoma cohorts, the SKCM and Liu cohorts (S3A–S3C Table). There was no obvious difference in the primary tumor thickness between metastatic samples with vs. without TERT promoter mutations (Wilcoxon p = 0.508, Fig 2A). Notably, TERT expression was higher in metastases of tumors arising from thinner primaries (Spearman p = 0.01, coeff = -0.16, Fig 2A; Wilcoxon p = 0.011, Breslow >2 mm: 8.95 TPM vs. Breslow <2 mm: 11.89 TPM, Fig 2B).

thumbnail
Fig 2. Primary tumor thickness and TERT in corresponding metastases.

(A) Breslow depth of primaries with TERT promoter mutations and TERT expression measured in corresponding metastatic tumors of the SKCM cohort (without one outlier; TERT expression >600, for data including outliers see S8 Fig in S1 File; also see S3D Table). (B) TERT expression in metastatic tumors arising from thin (Breslow depth <2 mm) and thick tumors (>2 mm). Information on TERT alterations in thin primaries are not available. TERTpm: TERT promoter mutations; TPM: transcripts per million. The figure was created with BioRender.com.

https://doi.org/10.1371/journal.pone.0281487.g002

TERT in melanoma subtypes

As the mitogen-activated protein kinase (BRAF-MAPK) signaling pathway is frequently activated in melanoma [29], we analyzed TERT in melanoma subtypes with hotspot mutations in BRAF, RAS (NRAS, KRAS, HRAS) and NF1 genes in the SKCM cohort. Melanomas with hotspot mutations carried TERT promoter mutations more often than triple wild-type melanomas (odds ratios >20, S4A Table). With respect to TERT expression, we found significantly lower TERT expression in BRAF hotspot mutants than in RAS hotspot mutants (Wilcoxon p = 0.0004, S4B Table). The group of patients with TERT promoter mutations showed down-regulated TERT expression in BRAF mutants and up-regulated TERT expression in RAS mutants (mutated RAS: Wilcoxon p = 0.024, mutated BRAF: Wilcoxon p = 0.004, S4C Table).

Mutational load

A common pattern of TERT promoter mutations and TERT expression was the association with more nuclear mutational load. In the entire SKCM cohort, samples with TERT promoter mutations showed higher overall mutation counts (Wilcoxon p = 0.014, Fig 3A, not significant for the metastatic subgroup) and higher TERT expression was also associated with mutation counts (Spearman p = 0.002, coeff = 0.16, S3B Fig in S1 File, metastatic subgroup: Spearman p = 0.017, coeff = 0.14, Fig 3A), however, not in the Liu cohort (Spearman: 0.194, coeff = 0.12, S3A and S3B Table).

thumbnail
Fig 3. Genetic associates of TERT alterations.

(A) TERT alterations and exome mutation load in the SKCM cohort. (B) TERT expression and DNA repair genes associated across all cohorts. (C) TERT alterations, mitochondrial mutation load and hypoxia (PCAWG cohort). TPM: transcripts per million; FPKM: fragments per kilobase of transcript per million fragments mapped; nsSNP: non-synonymous mitochondrial SNP; TERTpm: TERT promoter mutations. The figure was created with BioRender.com.

https://doi.org/10.1371/journal.pone.0281487.g003

Moreover, in the SKCM cohort we did not find TERT promoter mutations associated with Breslow depth or age at diagnosis, and we did not find TERT expression associated with sex (SKCM, Liu), age at diagnosis (SKCM), or various clinicopathological variables of the Liu cohort, such as heterogeneity or total neoantigens (S3 Table).

Gene expression associated with TERT alterations

At first, we replicated the previously described association of higher TERT expression levels in the presence of TERT promoter mutations in three independent cohorts (S5A Table). Since alternative splicing may result from mutation of the TERT promoter [17], we analyzed the expression of TERT isoforms. However, none of the isoforms were consistently regulated with TERT promoter mutation (S5A Table, S9 Fig in S1 File).

Next, we aimed to identify other cellular signaling pathways that may be driven by or co-regulated with either TERT promoter mutations (S6 Table) or TERT expression (S7 Table) followed by an enrichment analysis of associated gene expression. Associations with the presence of TERT promoter mutations revealed an overlap of 30 genes in the three bulk RNA-seq cohorts (in melanoma and across entities), of which six genes were consistently up- or down-regulated (up: ARL17A, FBF1, KAZALD1, PRAF2; down: PLEKHB2, RASGRP3; S6 Table). One gene (PRAF2) showed a consistent up-regulation also in the cell line data comprising other cancer entities (CCLE cohort). Enrichment analysis of the 30 genes associated with TERT promoter mutations showed overlaps with genes involved in ‘cilium organization’; ‘plasma membrane bound cell projection assembly’ and ‘actin cytoskeleton’ (S7A Table). To elucidate whether our findings are transferable to brain tumors, we extended our analyses to glioblastoma, in which TERT promoter mutations are highly recurrent [3] and diagnostic biomarkers [30]. Analysis of genes associated with TERT promoter mutations in 28 bulk tumor samples from the glioblastoma subcohort in PCAWG showed that of the six genes consistently associated with TERT promoter mutations in all bulk RNA-Seq cohorts, only ARL17A was also upregulated in glioblastoma (S6A Table).

In the context of TERT expression, 17 genes were consistently upregulated with TERT expression in all seven bulk RNA-seq cohorts, comprising melanoma and a cross-entity cancer cohort, as well as a cross-entity cell line cohort (Table 2, S7 Table). We confirmed that these TERT correlates were mostly tumor cell-specific genes within melanoma, as 15 genes were predominantly expressed in malignant tumor cells in our scRNA-seq data (Table 2, S7A and S7H Table). Enrichment analysis of the 17 genes upregulated with TERT expression revealed ‘DNA repair’, ‘double-strand break repair’, and ‘replication fork processing’ as relevant processes (Fig 3B, S8A Table). We replicated most of these associations in glioblastoma (Klughammer cohort) where 14 of 17 genes were also positively associated with TERT expression, including four genes related to DNA repair (XRCC2, MSH5, TRAIP, BRIP1, S6 Table).

thumbnail
Table 2. Genes repeatedly associated with TERT mRNA in 9 cohorts (S7 Table).

https://doi.org/10.1371/journal.pone.0281487.t002

To further elucidate tumor cell-specific signaling pathways, we used single-cell RNA-seq data and analyzed the top 100 genes co-expressed with TERT mRNA and performed an enrichment analysis with pathways, ontologies and cell type enrichment sets (S8C, S8D Table). Several cellular expression programs were repeatedly enriched, including the extracellular matrix, integrins, stromal cells, and smooth muscle cells (S8B Table).

Association of TERT and mitochondrial variables

To further elucidate the alternative role of TERT beyond telomere elongation in the molecular biology of mitochondria and reactive oxygen species (ROS) [15, 31], we first tested associations of several ROS-related candidate genes with the presence of genetic alterations in TERT. However, we did not observe consistent up- or downregulation off the examined genes NOX1-5, DUOX1/2, SOD1/2, OPA1, MFN1/2, PINK1, FUNDC1, and GPX4 in correlation with TERT promoter mutations or expression (S5B and S5C Table). Yet, within our discovery approach, we identified a consistent upregulation of DNA2 with higher TERT expression in several bulk tumor cohorts, potentially pointing to a role of TERT in DNA repair (Table).

We then analyzed the mutational load specifically in the mitochondria using the cross-entity PCAWG cohort (n = 1200 with data on mitochondrial mutations, S3C and S3E Table). TERT promoter mutation status was not associated with nonsynonymous mitochondrial or other mitochondrial mutations (S3C and S3E Table). However, TERT expression correlated with fewer, especially nonsynonymous mitochondrial mutations (Wilcoxon p = 0.007, Spearman p = 0.049, coeff = -0.06, S3D Table, Fig 3C) in stark contrast to the increased overall, and mainly nuclear, mutations assessed above (in SKCM metastases, n = 363 with data on overall mutations, Fig 3B). We further tested whether the expression of the mitochondrial targeting sequence, which is required for the transport of TERT into mitochondria [24], could be affected by TERT promoter mutations. Here, we did not detect a change in the proportion of expressed isoforms with mitochondrial targeting sequence (S5A Table) and conclude that TERT promoter mutations do not promote or restrain transcripts with mitochondrial targeting (Fig 3C).

TERT alterations in hypoxia and chromothripsis

We next tested whether TERT promoter mutations and TERT expression correlated with hypoxia and chromothripsis in the PCAWG and SKCM cohorts (S3B–S3E Table). Ragnum hypoxia scores in melanoma patients (SKCM, Wilcoxon p = 0.024, S3C Table) and Buffa scores across entities (PCAWG, Wilcoxon p = 4.678e-21, Fig 3C) were higher in samples carrying a TERT promoter mutation. We also observed upregulation of TERT expression with all hypoxia scores in the melanoma cohort (S3B Table) and with Buffa hypoxia scores across entities (Wilcoxon p = 7.43e-22). We observed significantly more chromothripsis in samples carrying TERT promoter mutations and a higher median TERT expression in samples with chromothripsis (PCAWG, S3D, S3E Table).

Discussion

TERT and ICI therapy

TERT promoter mutations have been discussed as potential prognostic factors in metastatic melanoma [5, 6, 8, 26, 27] and as potentially predictive most recently under BRAF/MEK [7] and anti-CTLA4 therapy [9]. TMB is discussed as a biomarker for ICI success [32, 33] and TERT promoter mutations showed association with higher TMB [9], however, multivariate approaches controlling for TMB were missing. Our analyses show that, when controlling for mutational load and prior MAPK inhibitor therapy, the association of TERT promoter mutations in the tested anti-CTLA4 and non-ICI cohorts was weak or absent. Hence, we believe that the extent to which TERT promoter mutations govern survival under anti-CTLA4 therapy is not statistically supported [7, 8]. In addition, since elevated TERT expression was not associated with survival, we conclude that melanoma differs from other cancer entities that showed an association between TERT levels and prognosis in lung cancer, oral squamous cell carcinoma, and colorectal carcinoma [1012]. To our best knowledge, TMB, however, was not assessed in these studies, and could possibly be improved by multivariate analyses. A striking correlation of TERT expression in our study was a higher estimate of tumor-infiltrating CD4+ T cells, which were previously associated with improved clinical outcome of ICI therapy [34]. There is evidence for a baseline TERT-specific CD4+ T cell immune response, occurring in over 50% of melanoma patients, rising to 80% in ICI responders [35, 36]. Consequently, a higher CD4+ T cell response, possibly explained by an anti-TERT T-helper type 1 cell response, could define a subgroup of patients with better ICI therapy response. However, our analyses of single-cell expression showed that, if malignant cells expressed higher levels of TERT, the corresponding CD4+ T cells from these tumors expressed exhaustion markers at higher levels (Fig 1B). Therefore, in TERT-high hypoxic melanoma, dysfunctional CD4+ T cells may accumulate employing these immuno-suppressive checkpoints [37]. As previously suspected, this could argue for a defect in anti-TERT CD4+ response during melanoma progression [36] and may explain why in our analysis, patients with high CD4+ T cell estimates and increased TERT expression did not show a survival benefit.

TERT expression in metastases from thin primaries

TERT promoter mutations were previously described to upregulate TERT expression [38, 39] involving the formation of ETS/TCF transcription factor binding sites [2, 38], and have been found in primary and metastatic melanoma. Here, metastatic melanomas showed higher TERT expression if they originated from thinner primaries, prompting the question of increased TERT expression in efficient early metastasis. This is especially relevant as TERT expression has been shown to be involved in most steps of the invasion-metastasis cascade [40]. The local invasion of primary melanoma cells might be regulated through increased TERT expression affecting cellular programs, such as NF-kB or metalloproteinases in addition to canonical telomerase activity [41, 42]. As the gene expression enrichment of specifically malignant melanoma cells revealed that TERT expression is co-regulated with integrins, TGF-beta, and interleukins, a TERT expression shift may be involved in remodeling the extracellular matrix, which is seen as a driving force for cancer stemness [43] and would aid invasion. It remains unclear whether TERT expression was elevated already in the thinner primary tumors corresponding to the metastases measured for RNA (Fig 2B). Higher Breslow depth was previously shown to be associated with the presence of TERT mutations in primary tumors [5, 26]. However, this association was not apparent in our analysis, probably because only thicker primaries were selected for the RNA-seq experiment in the SCKM cohort.

In conclusion, our data indicate that TERT expression may contribute to early metastasis from thin primaries, potentially through extracellular matrix remodeling, without a clear role for promoter mutations in this respect.

Hotspot mutations, mutational load and DNA repair

Initially, we confirmed the known co-occurrence of TERT promoter mutations and hotspot mutations in BRAF, RAS and NF1 as previously described for melanoma [5]. While, thus a co-mutation of the TERT promoter and members of the MAPK pathway is evident, TERT expression varied significantly between mutants, showing lower TERT expression in BRAF mutants compared to RAS mutants. Moreover, our finding that BRAF mutants exhibited lower TERT expression compared with non-BRAF mutants in TERT promoter-mutated samples somewhat contradicts the proposed model of TERT reactivation in BRAF-mutated melanoma samples through acquisition of TERT promoter mutations, subsequent binding of TCF, and thus increased TERT expression [44]. These contradicting findings are also of great interest for thyroid tumors, as the status of TERT promoter mutations together with BRAF mutations have been shown to be associated with clinicopathological aggressiveness [4547]. As our analysis of co-mutations were only available in one cohort, further investigations may improve our understanding of the MAPK pathway and TERT activation via TERT promoter mutations.

As a source of hotspot mutations, mutational load is also correlated with TERT expression in metastatic melanomas here and previously described [9]. The increased co-expression of DNA repair-related genes suggests that TERT expression may be involved in establishing genomic stability under replication stress since TERT expression regulates the chromatin state and DNA damage responses in normal human fibroblasts [48]. Beyond melanoma, co-expression of TERT and DNA repair genes was shared in various cancer entities including GBM, however, common gene expression patterns were not observed in association with TERT promoter mutations. To date, studies of TERT alterations in GBM have focused on the fairly frequent promoter mutations. Hence, our results may spark interest in assessing TERT expression levels in GBM to test sensitivity to DNA damage [49]. This could be relevant in the context of chemo- and radiotherapy independent of TERT promoter mutations, since TERT expression is also mediated via rearrangements and hypermethylation [50]. Moreover, our results support the view that this non-canonical function of TERT could be especially relevant under hypoxic conditions and after chromothripsis, where TERT expression was increased. In line with the proposed concept of a two-step activation of TERT [38], the up-regulation of TERT expression in later stages of melanoma development could aid the survival of melanoma cells in hypoxic tumors and metastasis to other sites.

TERT and mitochondrial stability

In cancer cells exposed to hypoxia, ROS levels produced by mitochondria are increased [51]. Moreover, TERT has previously been shown to be involved in the protection of cells from ROS-mediated oxidative stress, delaying aging processes supposedly via TERT activity in mitochondria [15]. As these effects were described predominantly in non-cancer tissues, we aimed to identify potential mitochondrial correlates of TERT expression in cancer and, specifically, in melanoma. While at first, we did not find associations of TERT with a set of fifteen ROS-related candidate genes, our less-biased discovery approach identified a consistently upregulated DNA2, known to be involved in DNA repair in the nucleus and mitochondria [52]. This is in line with TERT translocation into mitochondria to protect mitochondrial DNA from damage and implicates a dedicated repair gene in the described process [15, 24]. The correlation between TERT expression and fewer nonsynonymous mitochondrial mutations further supports this rationale. It contrasts though, with the fact that TERT expression and TERT promoter mutations were associated with more overall mutations in the nuclear genome. Therefore, DNA repair via TERT and possibly DNA2 may be mitochondria-specific. Interestingly, differing mutational processes have already been described in colorectal and gastric cancers, where microsatellite instability was observed in the nuclear, but not in the mitochondrial genome [53]. Thus, it would be highly interesting to explore the cellular localization of TERT and DNA2 in various cancers and in the context of hypoxia.

A potential limitation of our study is that different subtypes of TERT promoter mutations may differentially affect TERT expression and, therefore clinical factors. However, individual promoter mutations were only available for the SKCM and CCLE cohorts. Indeed, in both cohorts, the C288T mutation is the only mutation showing association with increased TERT expression (S5D Table). However, analyzing the different promoter mutations individually, whenever possible, did not reveal any differences in our results (S10 Fig). We strongly believe that, as the assessment of mutation types is currently part of routine testing, the different effects of the subtypes of TERT promoter mutations could be taken into account in future studies.

In conclusion, our analyses did not support a significant association between TERT genetic alterations and survival in melanoma patients. Hence, its presumed role as a biomarker, especially for anti-CTLA4 or MAPK inhibitor therapy, needs further validation. In addition, our data indicate dysfunctional CD4+ T cells in TERT high hypoxic melanoma. Since melanoma metastases with high TERT expression were shown to have originated from thinner primary tumors, we conclude that upregulation of TERT expression may contribute to metastasis. This is also supported by the involvement of TERT expression in the dynamics of the extracellular matrix. We add support to the non-canonical function of TERT expression related to mitochondrial and nuclear DNA stability even across cancer entities, especially under hypoxic conditions and after chromothripsis. In summary, our results demonstrate the possible role of TERT expression in metastasis formation and invasion in melanoma and highlight its role in DNA repair mechanisms across cancer entities.

Supporting information

S1 File. Supplementary material including the supplementary methods, description of supplementary tables and S1-10 Figs.

https://doi.org/10.1371/journal.pone.0281487.s001

(DOCX)

S1 Table. Multivariate survival analysis.

Multivariate analysis using Cox proportional hazard models for progression free survival (PFS) (S1A Table) and overall survival (OS) (S1B Table) subsetted for ICI therapy type in cohorts Liu, Gide, Hugo, Riaz, Van Allen and SKCM.

https://doi.org/10.1371/journal.pone.0281487.s002

(XLSX)

S2 Table. TERT expression and immune signatures.

Significant immune signatures in SKCM associated with TERT expression and comparison against other bulk RNA-seq cohorts (Gide, Hugo, Liu, Riaz, Van Allen).

https://doi.org/10.1371/journal.pone.0281487.s003

(XLSX)

S3 Table. TERT with respect to clinical variables.

Wilcoxon tests for group comparisons and Spearman correlations of clinical variables against TERT expression and TERT promoter mutation in Liu, SKCM and PCAWG cohorts (S3A–S3E Table).

https://doi.org/10.1371/journal.pone.0281487.s004

(XLSX)

S4 Table. TERT in melanoma subtypes.

Mutation subtypes against TERT expression (S4A Table) and TERT promoter mutation status (S4B Table) in the SKCM melanoma cohort. Effect of hotspot mutations on TERT expression in TERT promoter mutated samples (S4C Table).

https://doi.org/10.1371/journal.pone.0281487.s005

(XLSX)

S5 Table. TERT isoforms and mutation subtypes and influence of TERT alterations on ROS.

Influence of TERT promoter mutation on TERT isoform on mitochondrial targeting sequence (S5A Table), associations of ROS-related genes with TERT expression levels (S5B Table), and TERT promoter mutation status (S5C Table) in Gide, Riaz, Hugo, Liu, Van Allen, SKCM, PCAWG, CCLE cohorts, and single-cell. Difference on TERT promoter mutation subtypes on TERT expression on SKCM and CCLE cohorts (S5D Table).

https://doi.org/10.1371/journal.pone.0281487.s006

(XLSX)

S6 Table. Genetic correlates with TERT promoter mutations.

Genes associated with TERT promoter mutation status for the Van Allen, SKCM, PCAWG, and CCLE cohorts, and overlap of cohorts (S6A–S6E Table).

https://doi.org/10.1371/journal.pone.0281487.s007

(XLSX)

S7 Table. Genetic correlates with TERT expression.

Genes associated with TERT expression in each cohort (Gide, Riaz, Van Allen, Hugo, Liu, SKCM, PCAWG, CCLE, single-cell, and Klughammer) and overlap of cohorts (S7A–S7K Table).

https://doi.org/10.1371/journal.pone.0281487.s008

(XLSX)

S8 Table. Enrichment analysis.

Enrichment analysis of 17 genes associated with TERT expression and 30 genes associated with TERT promoter mutation in bulk tumors and cell lines (S8A Table), and for the top 100 genes associated with TERT expression in single-cells (S8B Table).

https://doi.org/10.1371/journal.pone.0281487.s009

(XLSX)

Acknowledgments

We thank Udo Stenzel for help with the data analysis.

References

  1. 1. Shay JW, Bacchetti S. A survey of telomerase activity in human cancer. European Journal of Cancer. 1997. pp. 787–791. pmid:9282118
  2. 2. Horn S, Figl A, Rachakonda PS, Fischer C, Sucker A, Gast A, et al. TERT promoter mutations in familial and sporadic melanoma. Science. 2013;339: 959–961. pmid:23348503
  3. 3. Killela PJ, Reitman ZJ, Jiao Y, Bettegowda C, Agrawal N, Diaz LA Jr, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci U S A. 2013;110: 6021–6026. pmid:23530248
  4. 4. Huang FW, Hodis E, Xu MJ, Kryukov GV, Chin L, Garraway LA. Highly recurrent TERT promoter mutations in human melanoma. Science. 2013;339: 957–959. pmid:23348506
  5. 5. Griewank KG, Murali R, Puig-Butille JA, Schilling B, Livingstone E, Potrony M, et al. TERT promoter mutation status as an independent prognostic factor in cutaneous melanoma. J Natl Cancer Inst. 2014;106. pmid:25217772
  6. 6. Nagore E, Heidenreich B, Rachakonda S, Garcia-Casado Z, Requena C, Soriano V, et al. TERT promoter mutations in melanoma survival. Int J Cancer. 2016;139: 75–84. pmid:26875008
  7. 7. Thielmann CM, Matull J, Zaremba A, Murali R, Chorti E, Lodde G, et al. TERT promoter mutations are associated with longer progression-free and overall survival in patients with BRAF-mutant melanoma receiving BRAF and MEK inhibitor therapy. Eur J Cancer. 2022;161: 99–107. pmid:34936949
  8. 8. Andrés-Lencina JJ, Rachakonda S, García-Casado Z, Srinivas N, Skorokhod A, Requena C, et al. TERT promoter mutation subtypes and survival in stage I and II melanoma patients. International Journal of Cancer. 2019. pp. 1027–1036. pmid:30070694
  9. 9. Li H, Li J, Zhang C, Zhang C, Wang H. TERT mutations correlate with higher TMB value and unique tumor microenvironment and may be a potential biomarker for anti-CTLA4 treatment. Cancer Med. 2020;9: 7151–7160. pmid:32810393
  10. 10. Chen H-H, Yu C-H, Wang J-T, Liu B-Y, Wang Y-P, Sun A, et al. Expression of human telomerase reverse transcriptase (hTERT) protein is significantly associated with the progression, recurrence and prognosis of oral squamous cell carcinoma in Taiwan. Oral Oncol. 2007;43: 122–129. pmid:16798059
  11. 11. Zhu C-Q, Cutz J-C, Liu N, Lau D, Shepherd FA, Squire JA, et al. Amplification of telomerase (hTERT) gene is a poor prognostic marker in non-small-cell lung cancer. Br J Cancer. 2006;94: 1452–1459. pmid:16641908
  12. 12. Bertorelle R, Briarava M, Rampazzo E, Biasini L, Agostini M, Maretto I, et al. Telomerase is an independent prognostic marker of overall survival in patients with colorectal cancer. Br J Cancer. 2013;108: 278–284. pmid:23322193
  13. 13. Guterres AN, Villanueva J. Targeting telomerase for cancer therapy. Oncogene. 2020;39: 5811–5824. pmid:32733068
  14. 14. Bell RJA, 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: 1036–1039. pmid:25977370
  15. 15. Haendeler J, Dröse S, Büchner N, Jakob S, Altschmied J, Goy C, et al. Mitochondrial Telomerase Reverse Transcriptase Binds to and Protects Mitochondrial DNA and Function From Damage. Arteriosclerosis, Thrombosis, and Vascular Biology. 2009. pp. 929–935. pmid:19265030
  16. 16. Ludlow AT, Slusher AL, Sayed ME. Insights into Telomerase/hTERT Alternative Splicing Regulation Using Bioinformatics and Network Analysis in Cancer. Cancers. 2019;11. pmid:31091669
  17. 17. Bollmann FM. Physiological and pathological significance of human telomerase reverse transcriptase splice variants. Biochimie. 2013;95: 1965–1970. pmid:23933091
  18. 18. Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48: W509–W514. pmid:32442275
  19. 19. Cortés-Ciriano I, Lee JJ-K, Xi R, Jain D, Jung YL, Yang L, et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat Genet. 2020;52: 331–341. pmid:32025003
  20. 20. Yuan Y, Ju YS, Kim Y, Li J, Wang Y, Yoon CJ, et al. Comprehensive molecular characterization of mitochondrial genomes in human cancers. Nat Genet. 2020;52: 342–352. pmid:32024997
  21. 21. Buffa FM, Harris AL, West CM, Miller CJ. Large meta-analysis of multiple cancers reveals a common, compact and highly prognostic hypoxia metagene. Br J Cancer. 2010;102: 428–435. pmid:20087356
  22. 22. Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51: 202–206. pmid:30643254
  23. 23. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Research. 2016. pp. W90–W97. pmid:27141961
  24. 24. Santos JH, Meyer JN, Skorvaga M, Annab LA, Van Houten B. Mitochondrial hTERT exacerbates free-radical-mediated mtDNA damage. Aging Cell. 2004;3: 399–411. pmid:15569357
  25. 25. Ihaka R, Gentleman R. R: A Language for Data Analysis and Graphics. J Comput Graph Stat. 1996;5: 299–314.
  26. 26. Roh MR, Park K-H, Chung KY, Shin SJ, Rha SY, Tsao H. Telomerase reverse transcriptase (TERT) promoter mutations in Korean melanoma patients. Am J Cancer Res. 2017;7: 134–138. pmid:28123854
  27. 27. Ofner R, Ritter C, Heidenreich B, Kumar R, Ugurel S, Schrama D, et al. Distribution of TERT promoter mutations in primary and metastatic melanomas in Austrian patients. J Cancer Res Clin Oncol. 2017;143: 613–617. pmid:27990595
  28. 28. Miggelbrink AM, Jackson JD, Lorrey SJ, Srinivasan ES, Waibl-Polania J, Wilkinson DS, et al. CD4 T-Cell Exhaustion: Does It Exist and What Are Its Roles in Cancer? Clin Cancer Res. 2021;27: 5742–5752. pmid:34127507
  29. 29. Sumimoto H, Imabayashi F, Iwata T, Kawakami Y. The BRAF–MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med. 2006;203: 1651–1656. pmid:16801397
  30. 30. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021;23: 1231–1251. pmid:34185076
  31. 31. Indran IR, Hande MP, Pervaiz S. hTERT overexpression alleviates intracellular ROS production, improves mitochondrial function, and inhibits ROS-mediated apoptosis in cancer cells. Cancer Res. 2011;71: 266–276. pmid:21071633
  32. 32. Choucair K, Morand S, Stanbery L, Edelman G, Dworkin L, Nemunaitis J. TMB: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials. Cancer Gene Ther. 2020;27: 841–853. pmid:32341410
  33. 33. Addeo A, Friedlaender A, Banna GL, Weiss GJ. TMB or not TMB as a biomarker: That is the question. Crit Rev Oncol Hematol. 2021;163: 103374. pmid:34087341
  34. 34. Fridman WH, Zitvogel L, Sautès–Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nature Reviews Clinical Oncology. 2017. pp. 717–734. pmid:28741618
  35. 35. Dosset M, Castro A, Carter H, Zanetti M. Telomerase and CD4 T Cell Immunity in Cancer. Cancers. 2020;12. pmid:32630460
  36. 36. Nardin C, Laheurte C, Puzenat E, Boullerot L, Ramseyer M, Marguier A, et al. Naturally Occurring Telomerase-Specific CD4 T-Cell Immunity in Melanoma. J Invest Dermatol. 2022;142: 435–444. pmid:34352265
  37. 37. Wherry EJ, John Wherry E, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nature Reviews Immunology. 2015. pp. 486–499. pmid:26205583
  38. 38. Chiba K, Lorbeer FK, Shain AH, McSwiggen DT, Schruf E, Oh A, et al. Mutations in the promoter of the telomerase gene contribute to tumorigenesis by a two-step mechanism. Science. 2017;357: 1416–1420.
  39. 39. Heidenreich B, Nagore E, Sivaramakrishna Rachakonda P, Garcia-Casado Z, Requena C, Traves V, et al. Telomerase reverse transcriptase promoter mutations in primary cutaneous melanoma. Nature Communications. 2014. pmid:24569790
  40. 40. Zou Y, Cong Y-S, Zhou J. Implications of telomerase reverse transcriptase in tumor metastasis. BMB Rep. 2020;53: 458–465. pmid:32731912
  41. 41. Ding D, Xi P, Zhou J, Wang M, Cong Y-S. Human telomerase reverse transcriptase regulates MMP expression independently of telomerase activity via NF-κB-dependent transcription. FASEB J. 2013;27: 4375–4383.
  42. 42. Ghosh A, Saginc G, Leow SC, Khattar E, Shin EM, Yan TD, et al. Telomerase directly regulates NF-κB-dependent transcription. Nat Cell Biol. 2012;14: 1270–1281.
  43. 43. Nallanthighal S, Heiserman JP, Cheon D-J. The Role of the Extracellular Matrix in Cancer Stemness. Frontiers in Cell and Developmental Biology. 2019. pmid:31334229
  44. 44. Li Y, Cheng HS, Chng WJ, Tergaonkar V. Activation of mutant TERT promoter by RAS-ERK signaling is a key step in malignant progression of BRAF-mutant human melanomas. Proc Natl Acad Sci U S A. 2016;113: 14402–14407. pmid:27911794
  45. 45. Liu X, Bishop J, Shan Y, Pai S, Liu D, Murugan AK, et al. Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr Relat Cancer. 2013;20: 603–610. pmid:23766237
  46. 46. Xing M, Liu R, Liu X, Murugan AK, Zhu G, Zeiger MA, et al. BRAF V600E and TERT promoter mutations cooperatively identify the most aggressive papillary thyroid cancer with highest recurrence. J Clin Oncol. 2014;32: 2718–2726. pmid:25024077
  47. 47. Liu X, Qu S, Liu R, Sheng C, Shi X, Zhu G, et al. TERT promoter mutations and their association with BRAF V600E mutation and aggressive clinicopathological characteristics of thyroid cancer. J Clin Endocrinol Metab. 2014;99: E1130–6. pmid:24617711
  48. 48. Masutomi K, Possemato R, Wong JMY, Currier JL, Tothova Z, Manola JB, et al. The telomerase reverse transcriptase regulates chromatin state and DNA damage responses. Proc Natl Acad Sci U S A. 2005;102: 8222–8227. pmid:15928077
  49. 49. Amen AM, Fellmann C, Soczek KM, Ren SM, Lew RJ, Knott GJ, et al. Cancer-specific loss of activation sensitizes glioblastoma to DNA damage. Proc Natl Acad Sci U S A. 2021;118. pmid:33758097
  50. 50. Patel B, Taiwo R, Kim AH, Dunn GP. TERT, a promoter of CNS malignancies. Neuro-Oncology Advances. 2020. pmid:32226942
  51. 51. Tafani M, Sansone L, Limana F, Arcangeli T, De Santis E, Polese M, et al. The Interplay of Reactive Oxygen Species, Hypoxia, Inflammation, and Sirtuins in Cancer Initiation and Progression. Oxid Med Cell Longev. 2016;2016: 3907147. pmid:26798421
  52. 52. Zheng L, Meng Y, Campbell JL, Shen B. Multiple roles of DNA2 nuclease/helicase in DNA metabolism, genome stability and human diseases. Nucleic Acids Res. 2020;48: 16–35. pmid:31754720
  53. 53. Gorelick AN, Kim M, Chatila WK, La K, Hakimi AA, Berger MF, et al. Respiratory complex and tissue lineage drive recurrent mutations in tumour mtDNA. Nat Metab. 2021;3: 558–570. pmid:33833465