2023 年 70 巻 11 号 p. 1035-1049
TERT promoter mutations (TERT-p mutations) have been found in many types of cancer and have emerged to play critical roles in tumor progression. The mutations upregulate TERT transcription, and TERT not only elongates telomeres and confers unlimited proliferative capacity on tumor cells, but is also involved in tumor progression and aggressiveness. In differentiated thyroid carcinoma, TERT-p mutations are associated with a number of high-risk clinicopathological aggressiveness and worse prognosis, making it the best molecular marker to predict tumor aggressiveness so far. This review summarizes recent relevant findings regarding TERT-p mutations and their functional/mechanistic aspects.
Telomerase Reverse Transcriptase (TERT) is a catalytic subunit of telomerase which maintains telomere structures at DNA ends and is not expressed in most human somatic cells, which is thought to be one of the mechanisms that prevents cancer in humans. In the vast majority of cancers, however, increased expression of the TERT gene is achieved by several mechanisms, including mutations in the core promoter region of the TERT gene (TERT-p mutations), thereby allowing cell immortalization which is a hallmark of cancer cells.
The first report of TERT-p mutations in cancer was published in 2013 [1, 2]. Whole genome sequencing analysis of malignant melanoma tissues revealed two highly recurrent somatic mutations in the TERT promoter: mutually exclusive heterozygous C > T mutations at –124 and –146 bp upstream from the initiation codon. They are also referred to as chr5:1,295,228 C > T (C228T) and chr5:1,295,250 C > T (C250T), respectively, based on the hg19 coordinate, but the latest human genome reference is GRCh38/hg38. TERT-p mutations have also been discovered in various other types of malignancies including bladder cancer, renal pelvic cancer, hepatocellular carcinoma, glioblastoma, and thyroid carcinoma. On the other hand, they are rarely detected in hematological malignancy, prostate cancer, gastrointestinal cancer, breast cancer, and lung cancer. Both mutations create binding sites for ETS family transcription factors and upregulate the transcription of the TERT gene.
Regarding the difference between the –124 and –146 mutations, several papers have reported that the transcriptional activity of –124 was higher than –146 in reporter assays [3-7], while others demonstrated no difference [1, 8]. As enhancers strongly regulate transcription of the human TERT gene, reporter assays using only the promoter region may not be able to reach a conclusion [9]. In studies comparing mRNA expression in clinical specimens, the results were also inconclusive [8, 10]. In melanomas, only the –124 mutation, but not –146, was associated with TERT mRNA upregulation [11]. There is also a report indicating that the –146 mutation, but not –124, requires not only ETS transcription factors but also NF-κB p52 for its transcriptional enhancement [12]. In general, the –124 mutation seems to be the more important of the two, but this too is still inconclusive and could differ depending on the type of cancer.
Interestingly, TERT-p mutations were scarcely detected in cancers arising from tissues that are always self-renewing (e.g., myeloid leukemia, breast cancer, and prostate cancer), while tumors arising from cells with low rates of self-renewal were found to have higher prevalence of TERT-p mutations (e.g., melanoma, hepatocellular carcinoma, urothelial carcinoma, glioblastoma) [13]. This may be because that telomerase is already epigenetically active in the progenitor cells of tissues that are constantly self-renewing, hence the mutation would not confer any growth advantage [14]. In addition, there is a strong correlation between the presence of the mutations and the age of patients in many types of cancers [4, 15-22].
A two-step mechanism has been proposed for the involvement of TERT-p mutations in tumorigenesis [23]. TERT-p mutations upregulate telomerase activity, but this is not enough to prevent bulk telomere shortening, only delaying senescence. The number of critically short telomeres then increases, causing genomic instability and selective pressure for further upregulation of TERT expression to sustain cell proliferation. Indeed, the telomere length in tumors with TERT-p mutations is shorter than that in normal tissue [24, 25].
In 2013, we and others demonstrated that TERT-p mutations were also detected in thyroid cancers, especially in aggressive histological types such as poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) [26, 27]. These mutations were found in all four major histological types derived from thyroid follicular cells, papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC), PDTC, and ATC, and were particularly prevalent in the aggressive latter two, but not found in medullary thyroid carcinoma (MTC), which originates from parafollicular C cells. Reported frequencies of TERT-p mutations in thyroid cancer are listed in Table 1. The –124 mutation is more prevalent than the –146 mutation. Shortly after the discovery of the mutations, a number of studies which analyzed the association between the presence of TERT-p mutations and clinicopathological parameters, especially in cases of differentiated thyroid carcinoma (DTC), which consists of PTC and FTC, were done.
Frequencies of the TERT promoter mutations in each histological subtype of thyroid cancer
No. | ATC | PDTC | micro PTC | PTC | HCC | FTC | FT-UMP | FTA/benign | MTC | Method | Country | Reference, PMID | |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
1 | 10/20 (50.0%) |
30/58 (52.0%) | 18/80 (22.5%) | 4/25 (16.0%) |
Sanger Sequencing | USA, JAPAN | Landa (2013) JCEM 23833040 |
[26] | |||||
2 | 23/54 (42.6%) |
3/8 (37.5%) | 30/257 (11.7%) | 9/79 (11.4%) | 0/85 (0%) |
0/16 (0%) |
Sanger Sequencing | USA? | Liu (2013) Endocr Relat Cancer 23766237 |
[27] | |||
3 | 2/16 (12.5%) |
3/14 (21.4%) | 13/169 (7.7%) | 9/64 (14.1%) | 0/81 (0%) |
0/28 (0%) |
Sanger Sequencing | Portugal | Vinagre (2013) Nat Commun 23887589 |
[15] | |||
4 | 10/20 (50.0%) |
13/51 (25.5%) | 8/36 (22.2%) | 0/37 (0%) |
Sanger Sequencing | Sweden | Liu (2014) Oncogene 24141777 |
[17] | |||||
5 | 46/408 (11.3%) | 8/22 (36.4%) | 0/44 (0%) |
Sanger Sequencing | China | Liu (2014) JCEM 24617711 |
[18] | ||||||
6 | 12/36 (33.3%) |
9/31 (29.0%) | 25/332 (7.5%) | 12/70 (17.1%) | Sanger Sequencing | Portugal | Melo (2014) JCEM 24476079 |
[16] | |||||
7 | 9/52 (17.3%) | 1/58 (1.7%) final diagnosis FTC 3/18 (16.7%) AFTA |
Sanger Sequencing | Sweden | Wang (2014) Cancer 24898513 |
[28] | |||||||
8 | 61/507 (12.0%) | Sanger Sequencing | USA | Xing (2014) J Clin Oncol 25024077 |
[19] | ||||||||
9 | 36/384 (9.4%) | NGS | USA | TCGA (2014) Cell 25417114 |
[132] | ||||||||
10 | 0/42 (0%) |
Sanger Sequencing | Sweden | Wang (2014) JCEM 24758186 |
[133] | ||||||||
11 | 8/61 (13.1%) |
Sanger Sequencing | USA | Chindris (2015) JCEM 25259908 |
[134] | ||||||||
12 | 22/182 (12.1%) | 8/58 (13.8%) | 0/6 (0%) |
0/14 (0%) |
Sanger Sequencing | Italy? | Muzza (2015) Mol Cell Endo 25448848 |
[23] | |||||
13 | 41/106 (38.7%) |
Sanger Sequencing | USA, China | Shi (2015) JCEM 25584719 |
[69] | ||||||||
14 | 19/404 (4.7%) |
NGS | Italy | de Biase (2015) Thyroid 26148423 |
[59] | ||||||||
15 | 21/121 (17.4%) | Sanger Sequencing | Italy | Gandolfi (2015) Eur J Endocrinol 25583906 |
[20] | ||||||||
16 | 6/14 (42.9%) | 26/243 (10.7%) | 1/3 (33.3%) |
1/5 (20%) | 0/17 (0%) |
Sanger Sequencing | Saudi Arabia | Qasem (2015) Endocr Relat Cancer 26354077 |
[135] | ||||
17 | 24/33 (72.7%) |
34/84 (40.5%) | NGS (MSK-IMPACT) | USA | Landa (2016) JCI 26878173 |
[70] | |||||||
18 | 18/432 (4.2%) | 7/119 (5.9%) | Sanger Sequencing | Korea | Song (2016) Cancer 26969876 |
[31] | |||||||
19 | 19/434 (4.4%) | Sanger Sequencing | China | Sun (2016) PLoS One 27064992 |
[32] | ||||||||
20 | 7/16 (43.8%) |
32/327 (9.8%) | 11/66 (16.7%) | Sanger Sequencing | Korea | Kim (2016) Endocr Relat Cancer 27528624 |
[136] | ||||||
21 | 64/1,051 (6.1%) | Sanger Sequencing | USA | Liu (2017) JAMA 27581851 |
[137] | ||||||||
22 | 36/357 (10.1%) | Sanger Sequencing | Japan | Matsuse (2017) Sci Rep 28150740 |
[46] | ||||||||
23 | 2/6 (33.3%) |
21/174 (12.1%) | 6/14 (42.9%) | Sanger Sequencing | Portugal, Spain | Melo (2017) JCEM 28323937 |
[34] | ||||||
24 | 98/144 (68.1%) |
Sanger Sequencing | France | Bonhomme (2017) Thyroid 28351340 |
[71] | ||||||||
25 | 86/118 (72.9%) |
Sanger Sequencing | Germany? | Tiedje (2017) Oncotarget 28489587 |
[72] | ||||||||
26 | 0/26 (0%) |
Sanger Sequencing | Japan | Yabuta (2017) Thyroid 28614984 |
[68] | ||||||||
27 | 19/35 (54.3%) fatal PDTC |
11/18 (61.1%) fatal PDTC |
2/4 (50%) fatal PDTC |
NGS | USA | Ibrahimpasic (2017) Clin Cancer Res 28634282 |
[138] | ||||||
28 | 7/355 (2.0%) | NGS | China | Liang (2018) J Pathol 29144541 |
[139] | ||||||||
29 | 24/94 (25.5%) | Sanger Sequencing | France | de la Fouchardiere (2018) Eur J Cancer 29413688 |
[140] | ||||||||
30 | 127/196? (65%) |
285/468? (61%) advanced PTC 0/15? (0%) pediatric PTC |
21/35? (59%) advanced HCC |
46/65? (71%) advanced FTC |
NGS | USA | Pozdeyev (2018) Clin Cancer Res 29615459 |
[73] | |||||
31 | 19/94 (20.2%) | 6/32 (18.8%) |
0/42 (0%) |
Sanger Sequencing | Sweden | Paulsson (2018) Endcr Relat Cancer 29692346 |
[35] | ||||||
32 | 8/21 (38.1%) |
9/21 (42.9%) | Sanger Sequencing | Italy | Romei (2018) Oncol Lett 29805648 |
[74] | |||||||
33 | 8/33 (24.2%) RAIR 0/34 (0%) age and sex matched disease-free PTC |
Sanger Sequencing | China | Meng (2019) IUBMB Life 31026111 |
[53] | ||||||||
34 | 20/159 (12.6%) by seq plus 3/159 (1.9%) by ddPCR |
Sanger Sequencing ddPCR | Japan | Tanaka (2019) Thyroid 31286848 |
[98] | ||||||||
35 | 77/568 (13.6%) | Sanger Sequencing | Poland | Trybek (2019) Endocrinology 31305897 |
[141] | ||||||||
36 | 12/14 (85.7%) |
14/198 (7.1%) | 10/40 (25%) |
6/34 (17.6%) | 0/15 (0%) |
Sanger Sequencing | USA | Panebianco (2019) Cancer Medicine 31408918 |
[6] | ||||
37 | 15/27 (55.6%) |
7/15 (46.7%) 11/28 (39.3%) focal ATC/PDTC |
15/31 (48.4%) metastatic PTC |
9/12 (75%) WI-FTC |
NGS | Korea | Yoo (2019) Nat Commun 31235699 |
[75] | |||||
38 | 4/40 (10.0%) pN1b 0/71 (0%) pN0 |
NGS | USA | Perera (2019) JCEM 31237614 |
[60] | ||||||||
39 | 75/101 (74.3%) |
NGS | USA, Australia | Xu (2020) Thyroid 32284020 |
[76] | ||||||||
40 | 5/266 (1.9%) |
61/712 (8.6%) | Sanger Sequencing | Korea | Song (2020) Cancers 32466217 |
[61] | |||||||
41 | 133/685 (19.4%) | SNaPshot Multiplex system | Japan | Ebina (2020) Cancers 32751594 |
[47] | ||||||||
42 | 47/164 (28.7%) | Sanger Sequencing | USA | Liu (2020) J Nucl Med 31375570 |
[51] | ||||||||
43 | 0/98 (0%) |
Sanger Sequencing | Italy | Sama (2021) Endocrine 32621051 |
[62] | ||||||||
44 | 5/133 (3.8%) < 40 mm w/o DM 2/34 (5.9%) > 40 mm w/o DM 10/35 (28.6%) w/ DM |
5/16 (31.3%) > 40 mm w/o DM 5/11 (45.5%) w/ DM |
Sanger Sequencing | Italy | Vianello (2021) Sci Rep 33790328 |
[142] | |||||||
45 | 16/504 (3.2%) | Sanger Sequencing | Korea | Lee (2021) Surgery 33952391 |
[63] | ||||||||
46 | 27/89 (30.3%) > 55 y.o. |
ddPCR | Japan | Nakao (2021) Clin Endocrinol 34322882 |
[79] | ||||||||
47 | 16/184 (8.7%) | Sanger Sequencing | Saudi Arabia | Parvathareddy (2022) Front Endocrinol 35360077 |
[64] | ||||||||
48 | 4/39 (10.3%) MI-FTC 5/24 (20.8%) EA-FTC 5/14 (35.7%) WI-FTC |
Sanger Sequencing | Korea | Park (2022) Mod Pathol 34497362 |
[37] | ||||||||
49 | 6/1,143 (0.5%) | 51/877 (5.8%) | Sanger Sequencing | Korea | Yang (2022) Endocrinol Metab 35864728 |
[36] | |||||||
50 | 20/126 (15.9%) all had RAI therapy |
Sanger Sequencing | China | Cao (2022) Eur J Nucl Med Mol Imaging 35501518 |
[54] | ||||||||
51 | 39/430 (9.1%) |
Sanger Sequencing | Poland | Kuchareczko (2022) Thyroid 35950639 |
[65] |
RAIR; radioiodine refractory, DM; distant metastasis, HCC; Hürthle cell carcinoma, WI-FTC; widely invasive FTC, MI-FTC; minimally invasive FTC, EA-FTC; encapsulated angioinvasive FTC, FT-UMP; follicular tumor of uncertain malignant potential, AFTA; autonomously functioning thyroid adenomas
The frequency of TERT-p mutations in DTC was in the range of 10–20% but varied more from report to report (Table 1). As discussed below, TERT-p mutations correlate with tumor aggressiveness and thus may be more frequent in institutions that treat advanced cases.
TERT-p mutations were strongly associated with high-risk clinicopathological characteristics such as age, tumor size, extrathyroidal extension, and distant metastasis, and they are now the best molecular marker so far to assess the risk for recurrence [15-20, 28-39]. Both the number of studies on FTC and the number of analyzed FTC cases are still small compared with those on PTC, hence the evidence for FTC may not be as conclusive as that for PTC. In PTC, the presence of TERT-p mutations is associated with that of the BRAFV600E mutation, and likewise with the RAS mutation in FTC [27, 38-40]. This means that the probability of having one of these mutations (either TERT-p/BRAFV600E or TERT-p/RAS) is high in TERT-p mutation-positive tumors. The coexistence of these mutations, especially with the BRAFV600E mutation, has stronger correlations with the above high-risk clinicopathological parameters [18, 19, 31, 40]. This seems reasonable because many of the ETS family transcription factors are upregulated through the mitogen-activated protein kinase (MAPK) pathway [41], which is strongly activated by the BRAFV600E mutation.
The BRAFV600E mutation is the most prevalent driver oncogene found in PTCs and constitutively activates the MAPK signaling pathway, but there is a controversy regarding the clinical significance of this mutation. Many papers especially from Western countries have demonstrated the association between the presence of the BRAFV600E mutation and tumor aggressiveness and worse prognosis [42, 43]. However, we and others failed to prove such a clinical significance in Japanese PTCs [44-47]. In addition, the prevalence of the BRAFV600E mutation in Japanese PTCs is higher (70–80%) than that in Western reports (50–60%). There is no definitive explanation for these discrepancies, but a regional difference probably exists [40]. Regarding TERT-p mutations, there seems to be no such difference. The clinicopathological significance of these mutations has been consistently demonstrated everywhere, including Japan [46, 47].
In addition, we have demonstrated that the combination of TERT-p mutations and Ki-67 labeling index, which is another prognostic marker in PTC, enables more precise prediction of prognosis than the TERT-p mutations alone [46]. This was thought to be because TERT-p mutations and Ki-67 represent different biological properties.
Radioactive iodine (RAI) treatment is the standard systemic treatment for advanced, persistent, recurrent, and metastatic DTCs, and the response to RAI treatment has a direct impact on prognosis. It has been reported that the presence of the BRAFV600E mutation is linked to RAI refractoriness (RAI-R) since the activation of the MAPK pathway causes dedifferentiation, leading to impairment of sodium iodide symporter [48-51]. Again, however, there seems to be a regional difference. In a subgroup analysis in the meta-analysis by Luo et al., Asian studies failed to show a significant association between the BRAFV600E mutation and RAI-R, whereas Western studies demonstrated significance and no heterogeneity [52].
TERT-p mutations have also been demonstrated to be associated with RAI-R by several studies [33, 51-55]. This significance was shown in both Asian and Western studies [52]. The combination of the BRAFV600E mutation and TERT-p mutations seems to have a stronger link to RAI-R [51, 54, 55]. Interestingly, although the number of studied cases was relatively small, the positive predictive value of TERT-p mutations for RAI-R was very high (most of the cases with TERT-p mutations showed RAI-R), and TERT-p mutations were strongly associated with reduced iodine uptake [33, 53]. On the other hand, however, its sensitivity was not high (there were many RAI-R cases without the TERT-p mutations), suggesting that there are other mechanisms to induce RAI-R. The prevalence of TERT-p mutations in RAI-R cases was 24.2–45.5% [33, 51, 53].
Recently, the category of low-risk papillary thyroid microcarcinoma (PTMC) has gained important clinical significance as active surveillance has been introduced as alternative management for this type of cancer [56-58]. Therefore, the analysis of TERT-p mutations, the best molecular marker to predict prognosis so far, in PTMCs is of great interest. The frequency of TERT-p mutations in PTMCs has been reported to be 0–8.7% [59-66]. These are generally lower than those of all PTCs including advanced cases. One should bear in mind that the detection sensitivity of the next-generation sequencing (NGS), especially deep sequencing used in recent studies, is theoretically higher than that of Sanger sequencing. However, there is no such tendency in the above studies.
At Kuma Hospital in Japan, where a program of active surveillance was started in 1993, and they reported that, over a 10-year observation period for 1,235 cases, 8% had increased in size ≥ 3 mm and 3.8% had novel appearance of lymph node metastasis [67]. We analyzed the specimens from three groups of patients who underwent surgery from the above cohort: 11 patients whose tumors did not grow for >5 years (but who requested surgery), 10 patients whose tumors increased by >4 mm in size, and 5 patients who had novel appearance of lymph node metastasis. TERT-p mutations were not detected by Sanger sequencing in any of the above cases [68]. Although the number of analyzed cases in this study was small, it is important to note that the background population was large and was actually under active surveillance.
The clinical significance of TERT-p mutations in PTMCs is still inconclusive. Four studies reported no association [59, 63, 65, 68]. One study showed an association with age, pT, and pN [36]. A report from Saudi Arabia demonstrated an association with distant metastasis and shorter metastasis-free survival even in multivariate analysis [64]. However, the prevalence of TERT-p mutations in this study was the highest (8.7%), and moreover, distant metastasis and recurrence were found in 6.0% and 15.8% of the cases, respectively, indicating that the PTMC cases in this study may have been unusually aggressive. Overall, the clinical significance of TERT-p mutations in PTMCs remains to be determined.
The frequencies of TERT-p mutations in PDTC and ATC have been reported to be 21.4–51.7% and 12.6–75.0%, respectively, which are much higher than that in DTC [15-17, 26, 27, 39, 69-76]. In some studies, TERT-p mutations were the most prevalent genetic alterations in ATC, suggesting the high importance of these mutations in the most aggressive type of thyroid cancers.
To the best of our knowledge, there are only two studies that analyze the prognostic significance of TERT-p mutations among ATC cases [70, 76]. Overall survival (OS) and OS time have been shown to be shorter in cases with TERT-p mutations, particularly those with concomitant BRAFV600E or RAS mutations, highlighting the important point that TERT-p mutations make the already highly malignant character of ATCs even more aggressive.
Regarding the anaplastic transformation from DTC, Oishi et al. analyzed the genetic status of 27 ATCs that were accompanied with PTC components [77]. TERT-p mutations were detected in 95% and 91% of the ATC and PTC components, respectively, indicating that its mutational status in both components was virtually identical. These results suggest that TERT-p mutations are associated with a higher risk for anaplastic transformation. Taken together, TERT-p mutations seem to play a key role in accelerating aggressiveness even at different stages.
It has been suggested that TERT-p mutations are a subclonal event in PTCs, whereas they are clonal in PDTCs and ATCs [70, 78]. Average allelic mutant frequency corrected for tumor purity using NGS was 23%, which means that 46% of the PTC cells harbored the mutations. These data prompted us to investigate the detection sensitivity by fine-needle aspiration (FNA), as preoperative detection of TERT-p mutations is likely to be clinically beneficial. We have developed a highly sensitive and specific detection method using droplet digital PCR (ddPCR) [79]. Using this method even for wash solution after FNA, highly concordant results were obtained between FNA and postoperative tissue samples, suggesting that preoperative detection of TERT-p mutations is a useful method for predicting tumor aggressiveness [79] and may enable more precise tailored management. In the same study, the clonality of TERT-p mutations was also assessed in tumors with the BRAFV600E mutation which was used as a reference of cancer cells. In the majority of the tumors, 80–120% of PTC cells carried the mutation, implying that TERT-p mutations are nearly clonal in most PTCs [79]. We do not yet have a good explanation for the discrepancy from the previous study. However, it should be noted that only relatively older cases (age ≥ 55) were used in our study. The issue of clonality still remains to be explored.
As described in the beginning of this paper, TERT-p mutations create a binding site for the ETS transcription factor family which includes many transcription factors such as ELFs, GABPA, ERG, ETSs, ETVs, and ELKs. First, Bell et al. showed that the most important transcription factor for enhancement of TERT transcription at the TERT-p mutation site was GABPA in glioblastomas [80]. Furthermore, Akincilar et al. found that a long-range interaction between the TERT promoter and 300 kb upstream region through GABPA proteins upregulates TERT transcription [81].
In thyroid cancer cells, Liu et al. demonstrated that BRAFV600E-MAPK activated FOS, which further upregulated GABPB transcription, and that the GABPA-GABPB complex bound to the mutated TERT promoter and increased TERT expression [82]. Recently, however, several papers have shown that GABPA is not important for TERT promoter-mutated thyroid cancer cells. Because GABPA expression was low in thyroid cancer tissues and also did not correlate well with the activation of the MAPK pathway, several groups performed experiments using thyroid cancer cell lines which suggested that ETV1, ETV4, ETV5 [83], ETV5 [84], ETV1, ELK1, ELF3, and ERF [85] play important roles at the mutated TERT promoter in thyroid cancer. These findings suggest that a definitive mechanism for the enhancement of TERT transcription by TERT-p mutations exists but has yet to be identified. Carefully designed experimental systems will be needed to identify the mechanism, especially in DTCs, where good cell lines are not available.
As described above, TERT-p mutations are one of the mechanisms by which TERT transcription is reactivated in cancer cells, but there are other modes of re-upregulation of TERT transcription such as genomic rearrangement, promoter methylation, and gene amplification [86–90]. According to the Cancer Genome Atlas (TCGA) and the Pan-Cancer Analysis of Whole Genomes (PCAWG) data, genomic rearrangement, and gene amplification were not detected in PTCs [91, 92]. However, another group identified MTMR12-TERT fusion in PTC (TCGA-BJ-A4O9-01) from the TCGA cohort (TCGA Fusion Gene Database). Subsequently, Yoo et al. identified two other structural rearrangements of TERT in widely invasive FTC [75]. Other studies reported that a small fraction of DTCs and MTCs had gene amplification, and its frequency may be higher in PDTCs and ATCs, but the number of analyzed cases was limited [6, 90, 93, 94]. In thyroid cancer, the most frequent of the other mechanisms of reactivation of TERT transcription is promoter methylation.
In general, DNA methylation at CpG sites in a core promoter region is associated with gene silencing. However, it has been reported that DNA methylation in the TERT promoter region distal to the core promoter causes increased TERT expression. It has been proposed that this is because transcriptional repressors such as CTCF cannot bind to the methylated region [95], leading to transcriptional upregulation.
A connection between TERT-p methylation/transcriptional upregulation and tumor aggressiveness has been demonstrated in childhood brain tumor, prostate cancer, melanoma, and also MTC [88, 89, 94, 96]. This relationship has also been demonstrated in DTC [35, 97]. However, further studies are still needed to determine their difference from TERT-p mutations and the significance of the combination of methylation and mutation.
We investigated the prognostic significance of high TERT expression but without TERT-p mutations in PTCs. Disease-free survival was significantly shorter in cases with high TERT expression compared with expression-free cases [98]. Interestingly, high TERT expression (without TERT-p mutations) was rather common in younger patients and was not associated with the BRAFV600E mutation or other high-risk clinicopathological parameters. This suggests that high TERT expression may be useful in predicting prognosis in young PTC patients. In addition, the results also suggest that although TERT is highly expressed in both situations (by TERT-p mutations or presumably by promoter methylation), their respective biological significance is different.
PTCs with TERT-p mutations have shorter telomeres than PTCs with the wild-type TERT promoter, even in cases older than 45 years old [17]. Another study demonstrated that TERT re-expression and promoter mutation/methylation/amplification co-occurred in clinically aggressive PTCs [93]. Moreover, significant telomere shortening was observed in these tumors, which was correlated with a higher Ki-67 index. The expression of subtelomeric genes, defined as genes located within a 5 Mb window adjacent to telomeres, was significantly increased in cases with shorter telomeres. Functional annotation revealed that there are many telomere-related genes in the subtelomeric region (e.g., TERT is 1.2 Mb from the 5p terminus). It was suggested that telomere shortening may lead to disruption of the loop structure, resulting in more open chromatin configuration, which may induce mutations in the TERT locus.
Several single nucleotide polymorphisms (SNPs) have been reported to affect TERT transcription, albeit controversial [99, 100]. Among them, rs2853669 located at the TERT promoter may have an interaction with TERT-p mutations. TERT-p mutations create a binding site for ETS transcription factors, whereas the rs2853669 disrupts another ETS site on the TERT promoter [101]. It has been reported that rs2853669 suppresses TERT transcriptional activity [3, 100], and that TERT mRNA expression is low in cancer cases carrying the minor allele [101]. Several studies suggest that patients with TERT-p mutations showed poor survival in the absence but not in the presence of rs2853669 in bladder cancer, glioblastoma, and oral squamous cell carcinoma [3, 102-105]. However, there are reports that rs2853669 has no effect on the prognostic impact of TERT-p mutations in glioma including glioblastoma [101, 106]. Conversely, it has been also reported that the combination of rs2853669 and the TERT-p mutation increased TERT expression and worsened prognosis in glioblastoma and liver cancer [107, 108]. There are inconsistencies even within the same type of cancer. In thyroid cancer, the coexistence of rs2853669 and the TERT-p mutation has been reported to be associated with an increase in tumor size [109, 110]. Overall, however, the biological and clinical significance of rs2853669, especially in relation to TERT-p mutations, is still inconclusive, and further studies are definitely needed.
It is easy to understand how telomerase activity would be important for overcoming the Hayflick limit and acquiring the infinite proliferative capacity of cancer cells. While it could well be associated with a faster growth rate, telomere elongation cannot adequately explain the other higher malignant properties such as invasion and metastasis. Indeed, novel TERT functions other than the above canonical telomerase activity have long been proposed [111].
The earliest evidence for a telomere-independent role for Tert comes from studies in mice. Since mice have very long telomeres (20–50 kb) compared to humans (5–10 kb), increased telomerase activity is not required in mouse tumors to prevent telomere shortening and replicative senescence. However, Tert was found to be upregulated in mouse breast cancer [112] and skin cancer [113], suggesting that Tert may also play a role other than telomere maintenance. Overexpression of Tert in mouse thymocytes caused T-cell lymphoma without affecting telomere length, also supporting a telomerase-independent role for Tert [114]. In addition, in the same study, it was shown that chromosomal instability was enhanced in cells with Tert overexpression after gamma irradiation, suggesting that Tert interferes with DNA damage response [114].
In normal cells, Sarin et al. showed that conditional expression of Tert in mouse skin epithelium induced proliferation of hair follicle stem cells, and that it was independent of telomerase activity [115]. The same group also demonstrated that a mutant Tert lacking RT function enhanced keratinocyte proliferation and activated hair follicle stem cells through Wnt and Myc [116]. In human cells, Smith et al. demonstrated that ectopic expression of TERT in human mammary epithelial cells enhanced cell proliferation through EGFR [117]. Hrdlickova et al. found that the overexpression of a naturally occurring alternative splicing variant (Δ4–13) of TERT accelerated the growth of several types of human cells without telomerase activity by stimulating the WNT signaling [118]. Telomerase-inactive mutant TERT has also been reported to be implicated in anti-apoptosis [119, 120].
TERT has also been reported to regulate angiogenesis. Zhou et al. showed that TERT activated VEGF transcription in WI-38 and HeLa cells, which was independent of telomerase activity [121]. Conversely, other papers demonstrated that VEGF upregulated TERT expression [122, 123], suggesting the existence of a possible positive feedback loop.
TERT has also been shown to interact with other molecules that are important in tumor progression. Park et al. reported that TERT interacted with BRG1, a chromatin-remodelling factor, and upregulated Wnt/β-catenin target genes [124]. Ghosh et al. demonstrated that TERT associated with RELA (P65) and induced NF-κB-dependent gene expression such as IL-6, IL-8, and TNF-α [125]. These cytokines play pivotal roles in inflammation, anti-apoptosis, and cancer progression. Interestingly, both Wnt/β-catenin and NF-κB have been shown to upregulate TERT transcription [126, 127], suggesting that this feed-forward loop may maintain the critical activation levels of these pathways.
An interesting novel enzymatic activity of TERT has also been reported. Phosphorylated TERT at threonine 249 by CDK1 acquired RNA dependent RNA polymerase (RdRP) activity. The complementary RNA of the mRNA of FOXO4, a tumor suppressor gene, was synthesized by TERT-RdRP, and this dsRNA complex was degraded, resulting in the downregulation of FOXO4 expression [128]. As FOXO4 has a tumor suppressor role, reduced FOXO4 expression is associated with tumor progression.
TERT has been shown to be involved in DNA damage response. Masutomi et al. reported that damage response to DNA double-strand breaks was impaired in cells lacking TERT. This is not recovered by a catalytically inactive TERT mutant, suggesting that telomerase activity may be required for this response [129]. Liu et al. used an irreversible TERT inhibitor, NU-1, and demonstrated that inhibiting TERT sensitized cancer cells to chemotherapy and ionizing radiation [130]. NU-1 delayed DNA repair of double-strand breaks, promoting cell senescence. This NU-1 effect was not observed in cells without TERT expression.
In thyroid cancer, the association between TERT and autophagy was demonstrated. Overexpression of TERT activated autophagy, which was suggested to be one of the mechanisms of tumor aggressiveness [131].
These findings indicate that TERT has non-canonical functions that play important roles in cancer progression. As TERT-p mutations induce TERT expression, these mechanisms are likely to be involved in enhancing tumor aggressiveness in thyroid cancer.
TERT-p mutations are strongly associated with tumor aggressiveness, response to treatment, and prognosis in thyroid cancer, at least in DTC. They are thus expected to be a good molecular marker for tailored management of the condition and could also be a novel therapeutic target for aggressive thyroid cancers.
The authors declare no conflict of interests.